WO2023175184A1 - 2,4-dioxo-1,4-dihydroquinazoline derivatives as parg inhibitors for the treatment of cancer - Google Patents

2,4-dioxo-1,4-dihydroquinazoline derivatives as parg inhibitors for the treatment of cancer Download PDF

Info

Publication number
WO2023175184A1
WO2023175184A1 PCT/EP2023/056968 EP2023056968W WO2023175184A1 WO 2023175184 A1 WO2023175184 A1 WO 2023175184A1 EP 2023056968 W EP2023056968 W EP 2023056968W WO 2023175184 A1 WO2023175184 A1 WO 2023175184A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
alkylene
heterocycloalkyl
heteroaryl
aryl
Prior art date
Application number
PCT/EP2023/056968
Other languages
French (fr)
Inventor
Ulrich LÜCKING
Andreas Goutopoulos
Zaixu Xu
Sotirios Sotiriou
Luca IACOVINO
Alena FREUDENMANN
Olivier Querolle
Original Assignee
Forx Therapeutics Ag
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 Forx Therapeutics Ag filed Critical Forx Therapeutics Ag
Publication of WO2023175184A1 publication Critical patent/WO2023175184A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/95Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in positions 2 and 4
    • C07D239/96Two oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • 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
    • C07D405/06Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • the present invention relates to a compound of formula (I): or a pharmaceutically acceptable salt or a prodrug thereof.
  • the present invention further relates to the compound of formula (I) of the present invention for use in therapy.
  • Instant compounds are particularly useful as PARG inhibitors, preferably as covalent PARG inhibitors, and can be used in a method of treatment of a proliferative disorder, preferably of cancer.
  • Cancer is a leading cause of death worldwide. Although progression-free survival and overall survival of cancer patients has improved over the past two decades, millions of cancer patients still have few therapeutic options and poor survival outcomes (Jemal et al., J. Natl. Cancer Inst. 2017, 109, 1975).
  • DRS DNA replication stress
  • DRS refers to the deregulation of DNA replication and cell cycle progression. DRS can be induced from endogenous or exogenous causes such as oncogene activation and chemotherapeutics, respectively (Zeman and Cimprich, Nat. Cell Biol. 2013, 16, 2). At the level of the replication fork, DRS leads to replication fork stalling, disengagement of the replisome and eventually collapse.
  • Poly(ADP)ribosylation is a transient and reversible post-translational modification that occurs at DNA damaged sites and is catalyzed by the poly (ADP-ribose) polymerase (PARP) family of proteins (Cohen and Chang, Nat. Chem. Biol. 2018, 14, 236). PARylation of various DNA repair proteins leads to their activation. Degradation of the poly(ADP) ribose chains is mediated primarily by the poly(ADP-ribose) glycohydrolase (PARG) protein. DNA damage dependent PARylation/dePARylation is a rapid and dynamic process which needs to be well regulated since imbalances between the two processes can lead to DNA damage.
  • PARP poly (ADP-ribose) polymerase
  • Human PARG encodes a 111 kDa protein of 976 amino acids. It contains a N-terminal regulatory domain, a catalytic domain and an ADP-ribose binding macrodomain. Five human PARG transcripts have been identified. Full length PARG is mostly nuclear; the smaller isoforms localize primarily to the cytoplasm. PARG functions primarily as an exo-hydrolase and it releases mainly mono(ADP-ribose) by hydrolyzing the a-O-glycosidic ribose-ribose bond in PAR. PARG can also act as an endo-hydrolase. PARG preferentially degrades long and linear PAR chains whereas its activity with small and branched PAR chains is significantly reduced (O’Sullivan et al., Nat. Commun. 2019, 10, 1182).
  • PARG is the dominant cellular PAR degrading enzyme, it cannot act on the terminal protein-ribose bond.
  • Additional hydrolases such as terminal ADP-ribose protein glycohydrolase (TARG1) and ADP-ribosylhydrolase 3 (ARH3) are also known to catalyze PAR-degradation.
  • TARG1 and ARH3 complete the reversal of PARylation by removing protein-bound mono(ADP-ribose) moieties (a) Fontana et al., Elife 2017, doi: 10.7554/eLife.28533; b) Rack et al., Genes Dev. 2020, 34, 263).
  • TARG1 is located in the nucleus and cytoplasm.
  • ARH3 is found primarily in the cytoplasm but it can also be found in the mitochondria and in the nucleus (Rack et al., Genes Dev. 2020, 34, 263).
  • PARG participates in DNA replication and in various DNA repair mechanisms including singlestrand break (SSB) repair and replication fork restart.
  • SSB singlestrand break
  • PARG inhibitors have shown synthetic lethal phenotype in cells with high levels of DRS caused by low expression of genes involved in DNA replication and/or replication fork stability (Pillay et al., Cancer Cell. 2019, 35, 519).
  • PARG inactivation, depletion or inhibition sensitizes cells to irradiation and to DNA damaging agents such as alkylating agents (e.g. temozolomide and methyl methanesulfonate) (a) Fujihara et al., Curr. Cancer Drug Targets 2009, 9, 953; b) Gogola et al., Cancer Cell 2018, 33, 1078; c) Houl et al., Nat Commun. 2019, 10, 5654).
  • alkylating agents e.g. temozolomide and methyl methanesulfonate
  • Certain compounds that are useful as PARG inhibitors are further disclosed in documents WO 2016/092326, WO 2016/097749 and WO 2021/055744.
  • the present invention provides a compound of formula (I): or a pharmaceutically acceptable salt thereof.
  • compound of formula (I) preferably encompasses also an enantiomer, diastereoisomer, tautomer, pharmaceutically acceptable solvate, pharmaceutically acceptable crystal form, pharmaceutically acceptable salt or a prodrug thereof.
  • a further embodiment of the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the compound of formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the present invention relates to the compound of formula (I) of the present invention or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present invention, for use in therapy.
  • the compounds of formula (I) are useful for treating a disease or disorder in which PARG activity is implicated.
  • the compounds of formula (I) are useful for a method of treating a proliferative disorder.
  • the proliferative disorder is cancer, preferably a human cancer.
  • hydrogen is herein used to refer to protium, deuterium and/or tritium, preferably to protium. Accordingly, the term “non-hydrogen atom” refers to any atoms that is not hydrogen, i.e. that is not protium, deuterium or tritium.
  • hydrocarbon group refers to a group consisting of carbon atoms and hydrogen atoms.
  • alicyclic is used in connection with cyclic groups and denotes that the corresponding cyclic group is non-aromatic.
  • alkyl refers to a monovalent saturated acyclic (i.e., non-cyclic) hydrocarbon group which may be linear or branched. Accordingly, an “alkyl” group does not comprise any carbon-to-carbon double bond or any carbon-to-carbon triple bond.
  • a “C1-5 alkyl” denotes an alkyl group having 1 to 5 carbon atoms. Preferred exemplary alkyl groups are methyl, ethyl, propyl (e.g., n-propyl or isopropyl), or butyl (e.g., n-butyl, isobutyl, sec-butyl, or tert-butyl).
  • alkyl preferably refers to C1-4 alkyl, more preferably to methyl or ethyl, and even more preferably to methyl.
  • alkenyl refers to a monovalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon double bonds while it does not comprise any carbon-to-carbon triple bond.
  • C2-5 alkenyl denotes an alkenyl group having 2 to 5 carbon atoms.
  • Preferred exemplary alkenyl groups are ethenyl, propenyl (e.g., prop-1 -en-1-yl, prop-1 -en-2-yl, or prop-2-en-1 -yl), butenyl, butadienyl (e.g., buta-1 ,3-dien-1 -yl or buta-1 ,3- dien-2-yl), pentenyl, or pentadienyl (e.g., isoprenyl).
  • alkenyl preferably refers to C2-4 alkenyl.
  • alkynyl refers to a monovalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon triple bonds and optionally one or more (e.g., one or two) carbon-to-carbon double bonds.
  • C2-5 alkynyl denotes an alkynyl group having 2 to 5 carbon atoms.
  • Preferred exemplary alkynyl groups are ethynyl, propynyl (e.g., propargyl), or butynyl.
  • alkynyl preferably refers to C2-4 alkynyl.
  • alkylene refers to an alkanediyl group, i.e. a divalent saturated acyclic hydrocarbon group which may be linear or branched.
  • a “C1-5 alkylene” denotes an alkylene group having 1 to 5 carbon atoms, and the term “C0-3 alkylene” indicates that a covalent bond (corresponding to the option “Co alkylene”) or a C1-3 alkylene is present.
  • Preferred exemplary alkylene groups are methylene (- CH2-), ethylene (e.g., -CH2-CH2- or -CH(-CH 3 )-), propylene (e.g., -CH2-CH2-CH2-, -CH(-CH 2 -CH 3 )-, -CH2- CH(-CH3)-, or -CH(-CH3)-CH2-), or butylene (e.g., -CH2-CH2-CH2-CH2-).
  • alkylene preferably refers to C1-4 alkylene (including, in particular, linear C1-4 alkylene), more preferably to methylene or ethylene, and even more preferably to methylene.
  • alkenylene refers to an alkenediyl group, i.e. a divalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon double bonds while it does not comprise any carbon-to-carbon triple bond.
  • a “C2- 5 alkenylene” denotes an alkenylene group having 2 to 5 carbon atoms.
  • alkenylene preferably refers to C2-4 alkenylene (including, in particular, linear C2-4 alkenylene).
  • alkynylene refers to an alkynediyl group, i.e. a divalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon triple bonds and optionally one or more (e.g., one or two) carbon-to-carbon double bonds.
  • a “C2-5 alkynylene” denotes an alkynylene group having 2 to 5 carbon atoms.
  • alkynylene preferably refers to C2-4 alkynylene (including, in particular, linear C2-4 alkynylene).
  • carbocyclyl refers to a hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings), wherein said ring group may be saturated, partially unsaturated (i.e., unsaturated but not aromatic) or aromatic.
  • “carbocyclyl” preferably refers to aryl, cycloalkyl or cycloalkenyl.
  • heterocyclyl refers to a ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings), wherein said ring group comprises one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group), and further wherein said ring group may be saturated, partially unsaturated (i.e., unsaturated but not aromatic) or aromatic.
  • each heteroatom-containing ring comprised in said ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • heterocyclyl preferably refers to heteroaryl, heterocycloalkyl or heterocycloalkenyl.
  • aryl refers to an aromatic hydrocarbon ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic).
  • Aryl may, e.g., refer to phenyl, naphthyl, dialinyl (i.e., 1 ,2-dihydronaphthyl), tetralinyl (i.e., 1 ,2,3,4-tetrahydronaphthyl), indanyl, indenyl (e.g., 1 H-indenyl), anthracenyl, phenanthrenyl, 9H- fluorenyl, or azulenyl.
  • an “aryl” preferably has 6 to 14 ring atoms, more preferably 6 to 10 ring atoms, even more preferably refers to phenyl or naphthyl, and most preferably refers to phenyl.
  • arylene refers to an aryl group, as defined herein above, but having two points of attachment, i.e. a divalent aromatic hydrocarbon ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic).
  • “Arylene” may, e.g., refer to phenylene (e.g., phen-1 ,2-diyl, phen-1 , 3-diyl, or phen-1 ,4-diyl), naphthylene (e.g., naphthalen-1 ,2-diyl, naphthalen-1 ,3-diyl, naphthalen-1 ,4-diyl, naphthalen-1 ,5-diyl, naphthalen-1 ,6- diyl, naphthalen-1 , 7-diyl, naphthalen-2, 3-diyl, naphthalen-2, 5-diyl, naphthalen-2, 6-diyl, naphthalen-2, 7- diyl, or naphthalen-2, 8-diyl), 1 ,2-dihydronaphthylene, 1 ,2,3,4-tetrahydronaph
  • an “arylene” preferably has 6 to 14 ring atoms, more preferably 6 to 10 ring atoms, even more preferably refers to phenylene or naphthylene, and most preferably refers to phenylene (particularly phen- 1 ,4-diyl).
  • heteroaryl refers to an aromatic ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic), wherein said aromatic ring group comprises one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group).
  • aromatic ring group comprises one or more (such as, e.g., one, two, three
  • each heteroatom-containing ring comprised in said aromatic ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • Heteroaryl may, e.g., refer to thienyl (i.e., thiophenyl), benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl (i.e., furanyl), benzofuranyl, isobenzofuranyl, chromanyl, chromenyl (e.g., 2H-1- benzopyranyl or 4H-1 -benzopyranyl), isochromenyl (e.g., 1 H-2-benzopyranyl), chromonyl, xanthenyl, phenoxathiinyl, pyrrolyl (e.g., 1 H-pyrrolyl), imidazolyl, pyrazolyl, pyridyl (i.e., pyridinyl; e.g., 2-pyridyl, 3- pyridyl, or 4-pyridyl), pyr
  • heteroaryl preferably refers to a 5 to 14 membered (more preferably 5 to 10 membered) monocyclic ring or fused ring system comprising one or more (e.g., one, two, three or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; even more preferably, a “heteroaryl” refers to a 5 or 6 membered monocyclic ring comprising one or more (e.g., one, two or three) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized.
  • heteroarylene refers to a heteroaryl group, as defined herein above, but having two points of attachment, i.e. a divalent aromatic ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic), wherein said aromatic ring group comprises one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i).
  • each heteroatom-containing ring comprised in said aromatic ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three, or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • Heteroarylene may, e.g., refer to thienylene (i.e., thiophenylene; e.g., thien-2,3-diyl, thien-2,4-diyl, or thien-2,5-diyl), benzo[b]thienylene, naphtho[2,3-b]thienylene, thianthrenylene, furylene (i.e., furanylene; e.g., furan-2,3-diyl, furan-2,4-diyl, or furan-2,5-diyl), benzofuranylene, isobenzofuranylene, chromanylene, chromenylene, isochromenylene, chromonylene, xanthenylene, phenoxathiinylene, pyrrolylene, imidazolylene, pyrazolylene, pyridylene (i.e., pyridinylene),
  • heteroarylene preferably refers to a divalent 5 to 14 membered (more preferably 5 to 10 membered) monocyclic ring or fused ring system comprising one or more (e.g., one, two, three or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; even more preferably, a “heteroarylene” refers to a divalent 5 or 6 membered monocyclic ring comprising one or more (e.g., one, two or three) ring heteroatoms independently selected from 0, S, and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optional
  • heteroarylene including any of the specific heteroarylene groups described herein, may be attached through two carbon ring atoms, particularly through those two carbon ring atoms that have the greatest distance from one another (in terms of the number of ring atoms separating them by the shortest possible connection) within one single ring or within the entire ring system of the corresponding heteroarylene.
  • cycloalkyl refers to a saturated hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings).
  • Cycloalkyl may, e.g., refer to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, decalinyl (i.e., decahydronaphthyl), or adamantyl.
  • cycloalkyl preferably refers to a C3-11 cycloalkyl, and more preferably refers to a C3-7 cycloalkyl.
  • a particularly preferred “cycloalkyl” is a monocyclic saturated hydrocarbon ring having 3 to 7 ring members (e.g., cyclopropyl or cyclohexyl).
  • cycloalkylene refers to a cycloalkyl group, as defined herein above, but having two points of attachment, i.e. a divalent saturated hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings).
  • Cycloalkylene may, e.g., refer to cyclopropylene (e.g., cyclopropan-1 ,1 -diyl or cyclopropan-1 ,2-diyl), cyclobutylene (e.g., cyclobutan-1, 1 -diyl, cyclobutan-1 ,2-diyl, or cyclobutan-1 ,3-diyl), cyclopentylene (e.g., cyclopentan-1,1 -diyl, cyclopentan-1 , 2-diyl, or cyclopentan-1 , 3-diyl), cyclohexylene (e.g., cyclohexan-1 ,1-diyl, cyclohexan-1 , 2-diyl, cyclohexan-1 , 3-diyl, or cyclohexan-1 ,4-diyl), cycloheptylene
  • cycloalkylene preferably refers to a C3-11 cycloalkylene, and more preferably refers to a C3-7 cycloalkylene.
  • a particularly preferred “cycloalkylene” is a divalent monocyclic saturated hydrocarbon ring having 3 to 7 ring members (e.g., cyclopropylene or cyclohexylene).
  • heterocycloalkyl refers to a saturated ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said ring group contains one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group).
  • each heteroatom-containing ring comprised in said saturated ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • Heterocycloalkyl may, e.g., refer to aziridinyl, azetidinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, azepanyl, diazepanyl (e.g., 1 ,4-diazepanyl), oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, morpholinyl (e.g., morpholin-4-yl), thiomorpholinyl (e.g., thiomorpholin-4-yl), oxazepanyl, oxiranyl, oxetanyl, tetrahydrofuranyl, 1 ,3-dioxolanyl, tetrahydropyranyl, 1 ,4-dioxanyl, oxepany
  • heterocycloalkyl preferably refers to a 3 to 11 membered saturated ring group, which is a monocyclic ring or a fused ring system (e.g., a fused ring system composed of two fused rings), wherein said ring group contains one or more (e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; more preferably, “heterocycloalkyl” refers to a 5 to 7 membered saturated monocyclic ring group containing one or more (e.g., one, two, or three) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms
  • A/-heterocycloalkyl refers to the heterocycloalkyl groups as defined hereinabove wherein said heterocycloalkyl includes at least one nitrogen atom which serves as an attachment point of said heterocycloalkyl.
  • heterocycloalkylene refers to a heterocycloalkyl group, as defined herein above, but having two points of attachment, i.e. a divalent saturated ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said ring group contains one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo
  • each heteroatom-containing ring comprised in said saturated ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • Heterocycloalkylene may, e.g., refer to aziridinylene, azetidinylene, pyrrolidinylene, imidazolidinylene, pyrazolidinylene, piperidinylene, piperazinylene, azepanylene, diazepanylene (e.g., 1 ,4-diazepanylene), oxazolidinylene, isoxazolidinylene, thiazolidinylene, isothiazolidinylene, morpholinylene, thiomorpholinylene, oxazepanylene, oxiranylene, oxetanylene, tetrahydrofuranylene, 1 ,3-dioxolanylene, tetrahydropyranylene, 1 ,4-dioxanylene, oxepanylene, thiiranylene, thietanylene, tetrahydrothiophenylene (
  • heterocycloalkylene preferably refers to a divalent 3 to 11 membered saturated ring group, which is a monocyclic ring or a fused ring system (e.g., a fused ring system composed of two fused rings), wherein said ring group contains one or more (e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; more preferably, “heterocycloalkylene” refers to a divalent 5 to 7 membered saturated monocyclic ring group containing one or more (e.g., one, two, or three) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N
  • cycloalkenyl refers to an unsaturated alicyclic (non-aromatic) hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said hydrocarbon ring group comprises one or more (e.g., one or two) carbon-to-carbon double bonds and does not comprise any carbon-to-carbon triple bond.
  • Cycloalkenyl may, e.g., refer to cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, or cycloheptadienyl.
  • cycloalkenyl preferably refers to a C3-11 cycloalkenyl, and more preferably refers to a C3-7 cycloalkenyl.
  • a particularly preferred “cycloalkenyl” is a monocyclic unsaturated alicyclic hydrocarbon ring having 3 to 7 ring members and containing one or more (e.g., one or two; preferably one) carbon-to-carbon double bonds.
  • heterocycloalkenyl refers to an unsaturated alicyclic (non-aromatic) ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said ring group contains one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group), and further wherein said ring group comprises at least one double bond between adjacent
  • each heteroatom-containing ring comprised in said unsaturated alicyclic ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring.
  • Heterocycloalkenyl may, e.g., refer to imidazolinyl (e.g., 2-imidazolinyl (i.e., 4,5-dihydro-1 H-imidazolyl), 3-imidazolinyl, or 4-imidazolinyl), tetrahydropyridinyl (e.g., 1 ,2,3,6-tetrahydropyridinyl), dihydropyridinyl (e.g., 1,2- dihydropyridinyl or 2,3-dihydropyridinyl), pyranyl (e.g., 2H-pyranyl or 4H-pyranyl), thiopyranyl (e.g., 2H-thiopyranyl or 4H-thiopyranyl), dihydropyranyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrazinyl, dihydroisoindolyl, oc
  • heterocycloalkenyl preferably refers to a 3 to 11 membered unsaturated alicyclic ring group, which is a monocyclic ring or a fused ring system (e.g., a fused ring system composed of two fused rings), wherein said ring group contains one or more (e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, wherein one or more carbon ring atoms are optionally oxidized, and wherein said ring group comprises at least one double bond between adjacent ring atoms and does not comprise any triple bond between adjacent ring atoms; more preferably, “heterocycloalkenyl” refers to a 5 to 7 membered monocyclic unsaturated non-aromatic ring group containing one or more (e.g
  • halogen refers to fluoro (-F), chloro (-CI), bromo (-Br), or iodo (-I).
  • haloalky I refers to an alkyl group substituted with one or more (preferably 1 to 6, more preferably 1 to 3) halogen atoms which are selected independently from fluoro, chloro, bromo and iodo, and are preferably all fluoro atoms. It will be understood that the maximum number of halogen atoms is limited by the number of available attachment sites and, thus, depends on the number of carbon atoms comprised in the alkyl moiety of the haloalkyl group.
  • Haloalkyl may, e.g., refer to -CF3, -CHF 2 , -CH 2 F, -CF 2 -CH 3 , -CH 2 -CF 3 , -CH 2 -CHF 2 , -CH 2 -CF 2 -CH 3 , -CH 2 -CF 2 -CF 3 , or -CH(CF 3 ) 2 .
  • a particularly preferred “haloalkyl” group is -CF 3 .
  • the terms “optional”, “optionally” and “may” denote that the indicated feature may be present but can also be absent.
  • the present invention specifically relates to both possibilities, i.e., that the corresponding feature is present or, alternatively, that the corresponding feature is absent.
  • the expression “X is optionally substituted with Y” (or “X may be substituted with Y”) means that X is either substituted with Y or is unsubstituted.
  • a component of a composition is indicated to be “optional”, the invention specifically relates to both possibilities, i.e., that the corresponding component is present (contained in the composition) or that the corresponding component is absent from the composition.
  • substituents such as, e.g., one, two, three or four substituents. It will be understood that the maximum number of substituents is limited by the number of attachment sites available on the substituted moiety.
  • the “optionally substituted” groups referred to in this specification carry preferably not more than two substituents and may, in particular, carry only one substituent.
  • the optional substituents are absent, i.e. that the corresponding groups are unsubstituted.
  • substituent groups comprised in the compounds of the present invention may be attached to the remainder of the respective compound via a number of different positions of the corresponding specific substituent group. Unless defined otherwise, the preferred attachment positions for the various specific substituent groups are as illustrated in the examples.
  • compositions comprising “a” compound of formula (I) can be interpreted as referring to a composition comprising “one or more” compounds of formula (I).
  • the term “about” preferably refers to ⁇ 10% of the indicated numerical value, more preferably to ⁇ 5% of the indicated numerical value, and in particular to the exact numerical value indicated. If the term “about” is used in connection with the endpoints of a range, it preferably refers to the range from the lower endpoint -10% of its indicated numerical value to the upper endpoint +10% of its indicated numerical value, more preferably to the range from of the lower endpoint -5% to the upper endpoint +5%, and even more preferably to the range defined by the exact numerical values of the lower endpoint and the upper endpoint.
  • the term “comprising” (or “comprise”, “comprises”, “contain”, “contains”, or “containing”), unless explicitly indicated otherwise or contradicted by context, has the meaning of “containing, inter alia”, i.e., “containing, among further optional elements, ...”. In addition thereto, this term also includes the narrower meanings of “consisting essentially of’ and “consisting of’.
  • a comprising B and C has the meaning of “A containing, inter alia, B and C”, wherein A may contain further optional elements (e.g., “A containing B, C and D” would also be encompassed), but this term also includes the meaning of “A consisting essentially of B and C” and the meaning of “A consisting of B and C” (i.e., no other components than B and C are comprised in A).
  • the present invention relates to a compound of formula (I): or a pharmaceutically acceptable salt thereof.
  • R oov is selected from C2 alkenyl, C2 alkynyl, -CH2CI, -CH2CN, and wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(Ci-4 alkyl), -CO-(Ci-4 alkyl), -CONH-(CI-4 alkyl), -OOC-(Ci-4 alkyl), -NHCO-(CI-4 alkyl), -(C1-4 alkylene)N(Ci-4 alkyl)(Ci-4 alkyl), -(C1-4 alkylene)-(A/-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, said alkynyl is optionally substituted with an optional substituent selected from C1-4 alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroary
  • Suitable cycloalkyl group is for example a cyclopropyl group.
  • Suitable aryl group is for example a phenyl group.
  • R cov is selected from C2 alkenyl, C2 alkynyl, and -CH2Cl, wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -OOC-(C1-4 alkyl), -NHCO-(C1-4 alkyl), - (C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, preferably selected from C1-4 alkyl, -COO
  • R cov is selected from C2 alkenyl, and -CH2Cl, wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -OOC-(C1-4 alkyl), -NHCO-(C1-4 alkyl), - (C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, preferably selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -(C1-4 alkylene)N(C1-4 alkyl)
  • R cov is C2 alkenyl, wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -OOC-(C1-4 alkyl), -NHCO-(C1-4 alkyl), -(C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N- heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, preferably selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -(C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C
  • R cov is C 2 alkenyl.
  • -W cov - is selected from -CO-, -SO- and -SO2-.
  • -W cov - is selected from - CO-, and -SO2-.
  • -W cov - is -CO-.
  • R N is selected from hydrogen and C1-4 alkyl. Suitable C1-4 alkyl is for example methyl or ethyl.
  • R N is hydrogen.
  • R 1 is hydrogen, chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl) (preferably R 1 is chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl)); and R 2 and R 3 are independently each C1-2 alkyl or C1-2 haloalkyl, or R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F; or -CR 1 R 2 R 3 is bicyclo[1,1,1]pent-1-yl.
  • R 1 is hydrogen, chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl) (preferably R 1 is chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl)); and R 2 and R 3 are independently each C1-2 alkyl or C1-2 haloalkyl, or R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F.
  • R 1 is hydrogen, chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1- 2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl) (preferably R 1 is chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl)); and R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F.
  • R 1 is preferably -CN, methyl, fluoromethyl, difluoromethyl or trifluoromethyl, more preferably R 1 is methyl or fluoromethyl.
  • R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F.
  • W is selected from -NHS(O)y-, -S(O)yNH-, -NHS(O)(NH)-, -NHS(O)(N-C1-2 alkyl)-, -S(O)(NH)-NH- , -S(O)(N-C1-2 alkyl)-NH-, wherein y is 1 or 2.
  • y is 2.
  • W is selected from -NHS(O)2-, -S(O)2NH-, -NHS(O)(NH)-, and -S(O)(NH)-NH-. More preferably, W is selected from -NHS(O)2-, and -S(O)2NH-, even more preferably W is -NHS(O)2-.
  • W is selected from -NHS(O)2-, and -S(O)2NH-, even more preferably W is -NHS(O)2-.
  • the left side of W as defined herein is attached to the carbon atom that carries R1, R2 and R3, and the right side of W as defined herein is attached to the ring system shown in formula (I).
  • X1 and X3 are independently selected from the group consisting of N, CH, C(C1-2 alkyl), C-Cl and CF, preferably independently selected from the group consisting of N, CH and CF.
  • X 1 is CF or CH and X3 is CH, more preferably X1 and X3 are each CH.
  • X2 is N or C-YC2-RC2, wherein Y C2 is selected from a covalent bond, C1-8 alkylene, C2-8 alkenylene, C2-8 alkynylene, cycloalkylene and heterocycloalkylene wherein said alkylene, said alkenylene and said alkynylene are each optionally substituted with one or more groups independently selected from R S1 , and further wherein one or more -CH2- units comprised in said alkylene, said alkenylene or said alkynylene are each optionally replaced by a group independently selected from -O-, -NH-, -N(C1-5 alkyl)-, -CO-, -S-, -SO-, and -SO2-, and wherein said cycloalkylene and heterocycloalkylene are each optionally substituted with one or more groups independently selected R S2 ; and wherein R C2 is selected from hydrogen, halo, -OH, -NH2, -SH,
  • YC2 is selected from a covalent bond, -(C1-3 alkylene)-, -CO-(C1-3 alkylene)-, (C1-3 alkylene)-CO-, -CONH-(C1-3 alkylene)-, -(C1-3 alkylene)-CONH-, -NHCO-(C1-3 alkylene)-, -(C1-3 alkylene)- NHCO-, -NH-(C1-3 alkylene)-, -(C1-3 alkylene)-NH-, -N(C1-5 alkyl)-, -O-(C1-3 alkylene)-, -(C1-3 alkylene)-O- , -SO2-(C1-3 alkylene)-, -(C1-3 alkylene)-SO2-, -CONH-, -NHCO-, -NH-, -O-, -CO- and -SO2-, wherein said alkylene, said alkenylene and said alkynylene are each optional
  • C1-3 alkylene is herein preferably a -CH2- group.
  • RC2 is selected from hydrogen, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from R S2 .
  • RC2 is selected from cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in X 2 are each optionally substituted with one or more groups independently selected from R S2 .
  • RC2 is selected from heterocycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in X 2 are each optionally substituted with one or more groups independently selected from R S2 .
  • X2 is C-YC2-RC2
  • -YC2-RC2 is is selected from -O-C1-12 alkyl, -NH-C1-12 alkyl, - N(C1-5 alkyl)-C1-12 alkyl, -O-C2-12 alkenyl, -NH-C2-12 alkenyl, -N(C1-5 alkyl)-C2-12 alkenyl, -O-C2-12 alkynyl, - NH-C2-12 alkynyl, -N(C1-5 alkyl)-C2-12 alkynyl, -(C0-3 alkylene)-cycloalkyl, -CO-(C0-3 alkylene)-cycloalkyl, - (C 0-3 alkylene)-CO-cycloalkyl, -CONH-(C 0-3 alkylene)-cycloalkyl, (C 0-3 alkylene)-CONH-cycloalkyl, (
  • -YC2-RC2 is selected from -(C0-3 alkylene)-heterocycloalkyl, -CO-(C0-3 alkylene)heterocycloalkyl, -(C0-3 alkylene)-CO-heterocycloalkyl, -CONH-(C0-3 alkylene)heterocycloalkyl, - (C0-3 alkylene)-CONH-heterocycloalkyl, -NHCO-(C0-3 alkylene)heterocycloalkyl, -(C0-3 alkylene)-NHCO- heterocycloalkyl, -NH-(C0-3 alkylene)heterocycloalkyl, -(C0-3 alkylene)-NH-heterocycloalkyl, -O-(C0-3 alkylene) heterocycloalkyl, (C0-3 alkylene)-O-cycloalkyl, -SO2-(C0-3 alkylene)-he
  • -YC2-RC2 is selected from -(C0-3 alkylene)-heterocycloalkyl, -CONH- heterocycloalkyl, -NHCO-heterocycloalkyl, -NH-heterocycloalkyl, -O-heterocycloalkyl, -CO- heterocycloalkyl, -SO2-heterocycloalkyl, -(C0-3 alkylene)aryl, -CONH-aryl, -NHCO-aryl, -NH-aryl, -O-aryl, -CO-aryl, -SO2-aryl, -(C0-3 alkylene)heteroaryl, -CONH-heteroaryl, -NHCO-heteroaryl, -NH-heteroaryl, -O- heteroaryl, -CO-heteroaryl and -SO2-heteroaryl, wherein said alkylene is optionally substituted
  • -YC2-RC2 is selected from -(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)aryl, and -(C0-3 alkylene)heteroaryl, wherein said heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from R S2. .
  • -YC2-RC2 is selected from heterocycloalkyl, aryl, and heteroaryl wherein said heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from R S2 .
  • -YC2-RC2 is selected from heterocycloalkyl and heteroaryl, wherein said heterocycloalkyl, and heteroaryl are each optionally substituted with one or more groups independently selected from R S2 . Even more preferably, -YC2-RC2 is heterocycloalkyl wherein said heterocycloalkyl is optionally substituted with one or more groups independently selected from R S2 .
  • the moiety represented with a partial formula is a moiety selected from wherein: R 7 is hydrogen, -CN, -Hal, or a moiety of the formula: wherein: L 71 is a bond or C1-5 alkylene optionally substituted with halo or oxo; L 72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH-, -CON(C1- 6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, -NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, -SO2N(C1-6 alkyl)-, -NHSO2-,
  • R 7 is a moiety of the formula: wherein: L 71 is a bond or C1-5 alkylene optionally substituted with halo or oxo; L 72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH-, -CON(C1- 6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, -NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, -SO2N(C1-6 alkyl)-, -NHSO2-, or -N(C1-6 alkyl)SO2-, and Q 7 is hydrogen, C1-6 alkyl, C2-6 alkeny
  • L 71 is a bond.
  • L 72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH- , -CON(C1-6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, -NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, or -NHSO2-, wherein said alkyl in is optionally substituted with one or more optional substituents selected from R S1 .
  • L 72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -CO-, -COO-, -OCO-, -CONH-, - SO2NH- or -NHSO2-. Even more preferably, L 72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, or -CO-. Even more preferably, L 72 is a bond, -O-, or -S-. Even more preferably, L 72 is a bond. It is noted that in the case of two bivalent chemical groups attached to each other, e.g.
  • -L 71 -L 72 - if each of them is defined to be a chemical bond, it is to be understood that the whole moiety comprising two bivalent chemical groups attached to each other is a bond.
  • -L 71 - is a bond
  • -L 72 - is a bond
  • -L 71 -L 72 - is a bond
  • Q 7 is C2-6 alkynyl, -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)- heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkynyl and alkylene are each optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 .
  • Q 7 is -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)- heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkylene is optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 .
  • Q 7 is -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkylene is optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 . It is to be understood that if L 71 is a bond and L 72 is a bond, then R 7 is preferably Q 7 .
  • R 7 is preferably Q 7 is hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, -(C0-2 alkylene)- cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkyl, alkenyl, alkynyl, and alkylene in R 7 are each optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R 7 are each optionally substituted with one or more optional substituents selected from R S2 .
  • R 7 is C2-6 alkynyl, -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkynyl and alkylene are each optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 .
  • R 7 is -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)- heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkylene is optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 .
  • R 7 is -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkylene is optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 .
  • R 8 is a moiety of the formula: wherein: L 81 is a bond, C1-5 alkylene optionally substituted with halo or oxo; L 82 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH-, -CON(C1- 6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, -NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, -SO2N(C1-6 alkyl)-, -NHSO2-, or -N(C1-6 alkyl)SO2-; and Q 8 is hydrogen, -CN, C1-6 alkyl, C2
  • L 81 is -(C0-2 alkylene)-, wherein said alkylene is optionally substituted with one or more optional substituents selected from R S1 .
  • L 82 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -CO-, -COO-, -OCO-, -CONH-, -SO2NH-, or -NHSO2-. More preferably, L 82 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, or -CO-. Even more preferably, L 82 is a bond, -O-, or -S-.
  • L 82 is a bond.
  • Q 8 is C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, heterocycloalkyl or heteroaryl; wherein said alkyl, alkenyl, and alkynyl are each optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 .
  • Q 8 is C2-6 alkynyl, cycloalkyl, aryl, heterocycloalkyl or heteroaryl; wherein said alkynyl is optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 .
  • Q 8 is cycloalkyl, aryl, heterocycloalkyl or heteroaryl; wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 .
  • Q 8 is cycloalkyl, heterocycloalkyl or heteroaryl; wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with one or more optional substituents selected from R S2 .
  • R 8 is preferably selected from -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl, wherein said alkylene is optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R 8 are each optionally substituted with one or more optional substituents selected from R S2 .
  • R 8 is selected from -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl, wherein said alkylene is optionally substituted with one or more optional substituents selected from R S1 ; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R 8 are each optionally substituted with one or more optional substituents selected from R S2 .
  • Particularly preferred -(C0-2 alkylene)- in R 8 is methylene.
  • R 8 is selected from - CH 2 -cycloalkyl, -CH 2 -aryl, -CH 2 -heterocycloalkyl or -CH 2 -heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R 8 are each optionally substituted with one or more optional substituents selected from R S2 . More preferably R 8 is selected from -CH2-cycloalkyl, -CH2-heterocycloalkyl or -CH2-heteroaryl, wherein said cycloalkyl, heterocycloalkyl, and heteroaryl in R 8 are each optionally substituted with one or more optional substituents selected from R S2 .
  • L 81 is methylene
  • -L 82 is a covalent bond
  • Q 8 is cycloalkyl, aryl, heterocycloalkyl or heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R 8 are each optionally substituted with one or more optional substituents selected from R S2 .
  • R 8 is methylene, -L 82 is a covalent bond, and Q 8 is cycloalkyl, heterocycloalkyl or heteroaryl, wherein said cycloalkyl, heterocycloalkyl, and heteroaryl in R 8 are each optionally substituted with one or more optional substituents selected from R S2 .
  • Particularly preferred R 8 is Preferably, the moiety represented with a partial formula Therein, R 8 is defined as defined hereinabove.
  • R S1 is selected from halogen, -CN, -OH, -O(C1-5 alkyl), -O(C1-5 haloalkyl), C1-5 haloalkyl, -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1- 5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C 1-5 alkyl)(C 1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C 1-5 alkyl), -N(C 1-5 alkyl),
  • R S1 is selected from halogen, -CN, -OH, -O(C1-5 alkyl), -O(C1-5 haloalkyl), C1-5 haloalkyl, -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1- 5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C1-5 alkyl), -N(C1-5 alkyl)-CO-(N-heterocyclo
  • R S1 is selected from halogen, -CN, -OH, -O(C1-5 alkyl), -O(C1-5 haloalkyl), C1-5 haloalkyl, -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), and -(N-heterocycloalkyl).
  • R S1 is selected from halogen, -CN, -OH, -SH, and -NH2.
  • R S2 is selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1- 5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1- 5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C1-5 alkyl), -CON
  • R S2 is selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1- 5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C1-5 alkyl), -N(C1-5 alkyl)-N(C
  • R S2 is selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C 1-5 haloalkyl), -SH, -S(C 1-5 alkyl), -S(C 1-5 haloalkyl), -NH 2 , -NH(C 1-5 alkyl), -NH(C 1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -(C1-5 alkylene)- CN, -(C1-5 alkylene)-OH, -(C1-5 alkylene)-O(C1-5 alkyl), -(C1-5 alkylene)-O(C1-5 haloalkyl), -(C1-5 halo
  • R S2 is selected from halogen, -CN, -OH, -SH, -NH2, -(C1-5 alkylene)-CN, - (C1-5 alkylene)-OH, -(C1-5 alkylene)-SH, and -(C1-5 alkylene)-NH2. Even more preferably, R S2 is selected from halogen, -CN, -OH, -SH, and -NH2.
  • -CR 1 R 2 R 3 is bicyclo[1,1,1]pent-1-yl.
  • R N is C1-4 alkyl. Particularly suitable C1-4 alkyl are methyl and ethyl groups.
  • X 2 is CH.
  • X1 is CF and X3 is CH.
  • -YC2-RC2 is aryl, preferably -YC2-RC2 is phenyl, wherein said aryl (said phenyl) is optionally substituted with one or more groups independently selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -CO(C1-5 al
  • -YC2-RC2 is heteroaryl, preferably selected from imidazolyl, pyridazinyl, thiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and indazolyl, wherein said heteroaryl is optionally substituted with one or more groups independently selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -CO(C1-5 alkyl),
  • -YC2-RC2 is heterocycloalkyl, preferably selected from morpholinyl, 1,1-dioxothiomorpholinyl, azetinyl, pyrrolidinyl, piperidinyl, 6-oxo-1,6- dihydropyridinyl, or piperazinyl, wherein said heterocycloalkyl is optionally substituted with one or more groups independently selected from R S2 .
  • -YC2-RC2 is piperazinyl, optionally substituted with one or more groups independently selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C 1-5 alkyl), -N(C 1-5 haloalkyl)(C 1-5 alkyl), -CO(C 1-5 alkyl), -CONH 2 , -CONH(C 1-5 alkyl), and -CON(C 1- 5 alkyl)(C1-5 alkyl).
  • -YC2-RC2 is piperazinyl (preferably N-piperazinyl) optionally substituted (preferably N-substituted) with -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), and -CON(C1-5 alkyl)(C1-5 alkyl).
  • -YC2-RC2 is piperazinyl (preferably N-piperazinyl) substituted (preferably N-substituted, preferably at a different N-atom than that attached to the ring system as shown in formula (I)), with -CON(C1-5 alkyl)(C1-5 alkyl), preferably with -CON(CH3)2.
  • -YC2-RC2 is heterocycloalkyl, wherein said heterocycle comprises a spiro ring system, optionally selected from 2-oxaspiro[3.5]non-6-en-7-yl, 2-oxaspiro[3.5]non- 7-yl, 2-oxa-8-azaspiro[4.5]dec-8-yl, 9-oxa-3-azaspiro[5.5]undec-3-yl, 2-oxa-6-azaspiro[3.4]oct-6-yl, 1- oxa-7-azaspiro[3.5]non-7-yl, 1-oxa-8-azaspiro[4.5]dec-8-yl, 6-oxa-2-azaspiro[3.3]hept-2-yl, 2,8- diazaspiro[4.5]dec-8-yl, 7-oxa-3-azabicyclo[3.3.0]oct-3-yl, 8-oxa-3-azabicy
  • R 7 is hydrogen, -CN, or -Hal.
  • R 7 is hydrogen.
  • the moiety represented with a partial formula is .
  • R 8 is hydrogen, -CN, or -Hal.
  • R 8 is hydrogen.
  • the moiety represented with a partial formula is in an eleventh specific embodiment, R 8 is -CH2C ⁇ CH.
  • R 8 is -CH 2 -cycloalkyl or -CO-cycloalkyl. Particularly preferred cycloalkyl is cyclopropyl.
  • R cov is C2 alkenyl or .
  • R cov is .
  • W is -NHS(O)y- wherein y is 1 or 2.
  • y is 2. It is to be understood that the left side of W as defined herein is attached to the carbon atom that carries R1, R2 and R3.
  • Preferred compound of formula (I) are selected from the following compounds or their pharmaceutically acceptable salts:
  • Particularly preferred compounds of formula (I) are selected from the following compounds or their pharmaceutically acceptable salts:
  • the present invention also relates to each of the intermediates described further below in the examples section of this specification, including any one of these intermediates in non-salt form or in the form of a salt (e.g., a pharmaceutically acceptable salt) of the respective compound.
  • a salt e.g., a pharmaceutically acceptable salt
  • Such intermediates can be used, in particular, in the synthesis of the compounds of formula (I).
  • the scope of the invention embraces all pharmaceutically acceptable salt forms of the compounds of formula (I) which may be formed, e.g., by protonation of an atom carrying an electron lone pair which is susceptible to protonation, such as an amino group, with an inorganic or organic acid, or as a salt of an acid group (such as a carboxylic acid group) with a physiologically acceptable cation.
  • Exemplary base addition salts comprise, for example: alkali metal salts such as sodium or potassium salts; alkaline earth metal salts such as calcium or magnesium salts; zinc salts; ammonium salts; aliphatic amine salts such as trimethylamine, triethylamine, dicyclohexylamine, ethanolamine, diethanolamine, triethanolamine, procaine salts, meglumine salts, ethylenediamine salts, or choline salts; aralkyl amine salts such as N,N- dibenzylethylenediamine salts, benzathine salts, benethamine salts; heterocyclic aromatic amine salts such as pyridine salts, picoline salts, quinoline salts or isoquinoline salts; quaternary ammonium salts such as tetramethylammonium salts, tetraethylammonium salts, benzyltrimethylammonium salts, benzyltriethylammoni
  • Exemplary acid addition salts comprise, for example: mineral acid salts such as hydrochloride, hydrobromide, hydroiodide, sulfate salts (such as, e.g., sulfate or hydrogensulfate salts), nitrate salts, phosphate salts (such as, e.g., phosphate, hydrogenphosphate, or dihydrogenphosphate salts), carbonate salts, hydrogencarbonate salts, perchlorate salts, borate salts, or thiocyanate salts; organic acid salts such as acetate, propionate, butyrate, pentanoate, hexanoate, heptanoate, octanoate, cyclopentanepropionate, decanoate, undecanoate, oleate, stearate, lactate, maleate, oxalate, fumarate, tartrate, malate, citrate, succinate, adipate, gluconate, glycolate, nic
  • Preferred pharmaceutically acceptable salts of the compounds of formula (I) include a hydrochloride salt, a hydrobromide salt, a mesylate salt, a sulfate salt, a tartrate salt, a fumarate salt, an acetate salt, a citrate salt, and a phosphate salt.
  • a particularly preferred pharmaceutically acceptable salt of the compound of formula (I) is a hydrochloride salt.
  • the compound of formula (I), including any one of the specific compounds of formula (I) described herein, is in the form of a hydrochloride salt, a hydrobromide salt, a mesylate salt, a sulfate salt, a tartrate salt, a fumarate salt, an acetate salt, a citrate salt, or a phosphate salt, and it is particularly preferred that the compound of formula (I) is in the form of a hydrochloride salt.
  • the present invention also specifically relates to the compound of formula (I), including any one of the specific compounds of formula (I) described herein, in non-salt form.
  • the scope of the invention embraces the compounds of formula (I) in any solvated form, including, e.g., solvates with water (i.e., as a hydrate) or solvates with organic solvents such as, e.g., methanol, ethanol, isopropanol, acetic acid, ethyl acetate, ethanolamine, DMSO, or acetonitrile. All physical forms, including any amorphous or crystalline forms (i.e., polymorphs), of the compounds of formula (I) are also encompassed within the scope of the invention. It is to be understood that such solvates and physical forms of pharmaceutically acceptable salts of the compounds of the formula (I) are likewise embraced by the invention.
  • the compounds of formula (I) may exist in the form of different isomers, in particular stereoisomers (including, e.g., geometric isomers (or cis/trans isomers), enantiomers and diastereomers) or tautomers (including, in particular, prototropic tautomers, such as keto/enol tautomers or thione/thiol tautomers). All such isomers of the compounds of formula (I) are contemplated as being part of the present invention, either in admixture or in pure or substantially pure form.
  • stereoisomers the invention embraces the isolated optical isomers of the compounds according to the invention as well as any mixtures thereof (including, in particular, racemic mixtures/racemates).
  • the racemates can be resolved by physical methods, such as, e.g., fractional crystallization, separation or crystallization of diastereomeric derivatives, or separation by chiral column chromatography.
  • the individual optical isomers can also be obtained from the racemates via salt formation with an optically active acid followed by crystallization.
  • the present invention further encompasses any tautomers of the compounds of formula (I). It will be understood that some compounds may exhibit tautomerism. In such cases, the formulae provided herein expressly depict only one of the possible tautomeric forms.
  • the formulae and chemical names as provided herein are intended to encompass any tautomeric form of the corresponding compound and not to be limited merely to the specific tautomeric form depicted by the drawing or identified by the name of the compound.
  • the scope of the invention also embraces compounds of formula (I), in which one or more atoms are replaced by a specific isotope of the corresponding atom.
  • the invention encompasses compounds of formula (I), in which one or more hydrogen atoms (or, e.g., all hydrogen atoms) are replaced by deuterium atoms (i.e., 2 H; also referred to as “D”).
  • deuterium atoms i.e., 2 H; also referred to as “D”.
  • the invention also embraces compounds of formula (I) which are enriched in deuterium.
  • Naturally occurring hydrogen is an isotopic mixture comprising about 99.98 mol-% hydrogen-1 ( 1 H) and about 0.0156 mol-% deuterium ( 2 H or D).
  • the content of deuterium in one or more hydrogen positions in the compounds of formula (I) can be increased using deuteration techniques known in the art.
  • a compound of formula (I) or a reactant or precursor to be used in the synthesis of the compound of formula (I) can be subjected to an H/D exchange reaction using, e.g., heavy water (D2O).
  • D2O heavy water
  • deuteration techniques are described in: Atzrodt J et al., Bioorg Med Chem, 20(18), 5658-5667, 2012; William JS et al., Journal of Labelled Compounds and Radiopharmaceuticals, 53(11 -12), 635-644, 2010; Modvig A et al., J Org Chem, 79, 5861 -5868, 2014.
  • the content of deuterium can be determined, e.g., using mass spectrometry or NMR spectroscopy.
  • it is preferred that the compound of formula (I) is not enriched in deuterium. Accordingly, the presence of naturally occurring hydrogen atoms or 1 H hydrogen atoms in the compounds of formula (I) is preferred.
  • the present invention also embraces compounds of formula (I), in which one or more atoms are replaced by a positron-emitting isotope of the corresponding atom, such as, e.g., 18 F, 11 C, 13 N, 15 0, 76 Br, 77 Br, 120 l and/or 124 l.
  • a positron-emitting isotope of the corresponding atom such as, e.g., 18 F, 11 C, 13 N, 15 0, 76 Br, 77 Br, 120 l and/or 124 l.
  • Such compounds can be used as tracers, trackers or imaging probes in positron emission tomography (PET).
  • the invention thus includes (i) compounds of formula (I), in which one or more fluorine atoms (or, e.g., all fluorine atoms) are replaced by 18 F atoms, (ii) compounds of formula (I), in which one or more carbon atoms (or, e.g., all carbon atoms) are replaced by 11 C atoms, (iii) compounds of formula (I), in which one or more nitrogen atoms (or, e.g., all nitrogen atoms) are replaced by 13 N atoms, (iv) compounds of formula (I), in which one or more oxygen atoms (or, e.g., all oxygen atoms) are replaced by 15 O atoms, (v) compounds of formula (I), in which one or more bromine atoms (or, e.g., all bromine atoms) are replaced by 76 Br atoms, (vi) compounds of formula (I), in which one or more bromine atoms (or, e.g., all
  • the present invention further embraces the prodrugs of the compounds of formula (I).
  • the term “prodrug” of the compound of formula (I) refers to a derivative of the compounds of formula (I) that upon administration to a subject becomes metabolized to the said compound of formula (I).
  • Said prodrugs of the compound of formula (I) may include modifications of -OH, -NH2, or -COOH group if present in the compound of formula (I), which preferably can be hydrolyzed to - OH, -NH2, or -COOH groups, respectively, e.g. upon administration to the subject.
  • such prodrugs may preferably include for the compounds of formula (I) which comprise -OH moiety derivatives wherein said -OH moiety is turned into an -ORx moiety, wherein R x preferably comprises a moiety selected from -CO-, -CH2-O-CO, -CH2-O-CO-O-, and -CH(CH3)-O-COO-, more preferably wherein Rx is selected from -CO-R y , -CH2-O-CO-R y , -CH2-O-CO-O-R y , and -CH(CH3)-O- COO-R y , wherein R y is preferably carbocyclyl, heterocyclyl, C1-5 alkyl, -NH-(Ci-s alkyl) or -S-(Ci-s alkyl), wherein the said alkyl is optionally substituted with a group selected from halogen, -CN, -OH, C
  • such prodrugs may preferably include for the compounds of formula (I) which comprise -NH2 moiety derivatives wherein said -NH2 moiety is turned into -NHCOO-R y moiety, wherein R y is as defined hereinabove.
  • such prodrugs may preferably include for the compounds of formula (I) which comprise -COOH moiety derivatives wherein said -COOH group is turned into -COOR y moiety, wherein R y is as defined hereinabove.
  • groups that can be derivatized to yield prodrugs are known to the skilled person.
  • the compounds provided herein may be administered as compounds perse or may be formulated as medicaments.
  • the medicaments/pharmaceutical compositions may optionally comprise one or more pharmaceutically acceptable excipients, such as carriers, diluents, fillers, disintegrants, lubricating agents, binders, colorants, pigments, stabilizers, preservatives, antioxidants, and/or solubility enhancers.
  • the pharmaceutical compositions may comprise one or more solubility enhancers, such as, e.g., poly(ethylene glycol), including poly(ethylene glycol) having a molecular weight in the range of about 200 to about 5,000 Da (e.g., PEG 200, PEG 300, PEG 400, or PEG 600), ethylene glycol, propylene glycol, glycerol, a non-ionic surfactant, tyloxapol, polysorbate 80, macrogol-15-hydroxystearate (e.g., Kolliphor ® HS 15, CAS 70142-34-6), a phospholipid, lecithin, dimyristoyl phosphatidylcholine, dipalmitoyl phosphatidylcholine, distearoyl phosphatidylcholine, a cyclodextrin, ⁇ -cyclodextrin, ⁇ -cyclodextrin, hydroxyethyl- ⁇ -cyclodextrs, a solubility enhancer
  • the pharmaceutical compositions may also comprise one or more preservatives, particularly one or more antimicrobial preservatives, such as, e.g., benzyl alcohol, chlorobutanol, 2-ethoxyethanol, m-cresol, chlorocresol (e.g., 2-chloro-3-methyl-phenol or 4-chloro-3-methyl-phenol), benzalkonium chloride, benzethonium chloride, benzoic acid (or a pharmaceutically acceptable salt thereof), sorbic acid (or a pharmaceutically acceptable salt thereof), chlorhexidine, thimerosal, or any combination thereof.
  • preservatives particularly one or more antimicrobial preservatives, such as, e.g., benzyl alcohol, chlorobutanol, 2-ethoxyethanol, m-cresol, chlorocresol (e.g., 2-chloro-3-methyl-phenol or 4-chloro-3-methyl-phenol), benzalkonium chloride, benzethonium chloride, benzoic
  • compositions can be formulated by techniques known to the person skilled in the art, such as the techniques published in “Remington: The Science and Practice of Pharmacy”, Pharmaceutical Press, 22 nd edition.
  • the pharmaceutical compositions can be formulated as dosage forms for oral, parenteral, such as intramuscular, intravenous, subcutaneous, intradermal, intraarterial, intracardial, rectal, nasal, topical, aerosol or vaginal administration.
  • Dosage forms for oral administration include coated and uncoated tablets, soft gelatin capsules, hard gelatin capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixirs, powders and granules for reconstitution, dispersible powders and granules, medicated gums, chewing tablets and effervescent tablets.
  • Dosage forms for parenteral administration include solutions, emulsions, suspensions, dispersions and powders and granules for reconstitution. Emulsions are a preferred dosage form for parenteral administration.
  • Dosage forms for rectal and vaginal administration include suppositories and ovula.
  • Dosage forms for nasal administration can be administered via inhalation and insufflation, for example by a metered inhaler.
  • Dosage forms for topical administration include creams, gels, ointments, salves, patches and transdermal delivery systems.
  • the compounds of formula (I) or the above described pharmaceutical compositions comprising a compound of formula (I) may be administered to a subject by any convenient route of administration, whether systemically/peripherally or at the site of desired action, including but not limited to one or more of: oral (e.g., as a tablet, capsule, or as an ingestible solution), topical (e.g., transdermal, intranasal, ocular, buccal, and sublingual), parenteral (e.g., using injection techniques or infusion techniques, and including, for example, by injection, e.g., subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, or intrasternal by, e.g., implant of a depot, for example, subcutaneously or intramuscularly), pulmonary (e
  • examples of such administration include one or more of: intravenously, intraarterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracardially, intracranially, intramuscularly or subcutaneously administering the compounds or pharmaceutical compositions, and/or by using infusion techniques.
  • parenteral administration the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood.
  • the aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary.
  • the preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
  • Said compounds or pharmaceutical compositions can also be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.
  • the tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules.
  • excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine
  • disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glyco
  • Preferred excipients in this regard include lactose, starch, a cellulose, or high molecular weight polyethylene glycols.
  • the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
  • the compounds or pharmaceutical compositions are preferably administered by oral ingestion, particularly by swallowing.
  • the compounds or pharmaceutical compositions can thus be administered to pass through the mouth into the gastrointestinal tract, which can also be referred to as “oral-gastrointestinal” administration.
  • said compounds or pharmaceutical compositions can be administered in the form of a suppository or pessary, or may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder.
  • the compounds of the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch.
  • sustained-release compositions include semi-permeable polymer matrices in the form of shaped articles, e.g., films, or microcapsules.
  • Sustained-release matrices include, e.g., polylactides, copolymers of L-glutamic acid and gamma-ethyl-L-glutamate, poly(2-hydroxyethyl methacrylate), ethylene vinyl acetate, or poly-D-(— )-3-hydroxybutyric acid.
  • Sustained-release pharmaceutical compositions also include liposomally entrapped compounds. The present invention thus also relates to liposomes containing a compound of the invention.
  • Said compounds or pharmaceutical compositions may also be administered by the pulmonary route, rectal routes, or the ocular route.
  • they can be formulated as micronized suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzalkonium chloride.
  • they may be formulated in an ointment such as petrolatum.
  • dry powder formulations of the compounds of formula (I) for pulmonary administration may be prepared by spray drying under conditions which result in a substantially amorphous glassy or a substantially crystalline bioactive powder. Accordingly, dry powders of the compounds of the present invention can be made according to an emulsification/spray drying process.
  • said compounds or pharmaceutical compositions can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water.
  • they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, 2-octyldodecanol, benzyl alcohol and water.
  • the present invention thus relates to the compounds or the pharmaceutical compositions provided herein, wherein the corresponding compound or pharmaceutical composition is to be administered by any one of: an oral route; topical route, including by transdermal, intranasal, ocular, buccal, or sublingual route; parenteral route using injection techniques or infusion techniques, including by subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, intrasternal, intraventricular, intraurethral, or intracranial route; pulmonary route, including by inhalation or insufflation therapy; gastrointestinal route; intrauterine route; intraocular route; subcutaneous route; ophthalmic route, including by intravitreal, or intracameral route; rectal route; or vaginal route.
  • Preferred routes of administration are oral administration or parenteral administration.
  • a physician will determine the actual dosage which will be most suitable for an individual subject.
  • the specific dose level and frequency of dosage for any particular individual subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual subject undergoing therapy.
  • a proposed, yet non-limiting dose of the compounds according to the invention for oral administration to a human may be 0.05 to 2000 mg, preferably 0.1 mg to 1000 mg, of the active ingredient per unit dose.
  • the unit dose may be administered, e.g., 1 to 3 times per day.
  • the unit dose may also be administered 1 to 7 times per week, e.g., with not more than one administration per day. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient/subject as well as the severity of the condition to be treated. The precise dose and also the route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
  • the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as defined herein for use in therapy.
  • the present invention provides compounds that function as inhibitors of PARG.
  • the present invention provides a method of inhibiting PARG enzyme activity in vitro or in vivo, said method comprising contacting a cell with an effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein.
  • the present inventors have demonstrated that certain compounds of formula (I) as described herein are covalent inhibitors of PARG enzyme.
  • the compounds of the present invention are significantly more potent (i.e., exhibit lower ICso) against the wild-type PARG protein in comparison to its C872A mutant, which is not capable of covalently binding the compounds of the present invention.
  • the inhibition of PARG by the compounds of the present invention is time-dependent, leading to lower ICso values upon 2-hour incubation when compared to a shorter incubation of 15 minutes.
  • the present invention also provides a method of selectively inhibiting PARG enzyme activity over PARP1 or ARH3 enzyme activity in vitro or in vivo.
  • the said method comprises the steps of contacting a cell with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as defined herein.
  • the present invention relates to the compound of formula (I), as disclosed herein, for use in a method of treating a disease or disorder in which PARG activity is implicated in a subject or patient in need of such treatment.
  • Said method of treatment comprises administering to said subject/patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as defined herein.
  • the present invention relates to the compound of formula (I), as disclosed herein, for use in treating a disease or disorder in which PARG activity is implicated.
  • the present invention relates to a method of inhibiting cell proliferation, in vitro or in vivo, said method comprising contacting a cell with an effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein.
  • the present invention relates to the compound of formula (I) or a pharmaceutically acceptable salt thereof for use in of inhibiting cell proliferation, in vitro or in vivo.
  • the present invention relates to a method of treating a proliferative disorder in a subject or patient in need of such treatment.
  • the said method of treating a proliferative disorder in a subject or patient in need thereof comprises administering to said subject/patient a therapeutically effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as defined herein.
  • the proliferative disorder is cancer.
  • the present invention relates to a method of treating cancer in a subject or patient in need thereof.
  • the said method of treating cancer in a subject or patient in need thereof comprises administering to said subject/patient a therapeutically effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as defined herein.
  • the cancer is human cancer.
  • the present invention relates to the compound of formula (I) or a pharmaceutically acceptable salt thereof, for use in treating a proliferative disorder.
  • the proliferative disorder is cancer. Therefore, the present invention relates to the compound of formula (I) or a pharmaceutically acceptable salt thereof for use in treating cancer.
  • the cancer is human cancer.
  • the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, for use in the manufacture of a medicament for the treatment of a proliferative condition.
  • the proliferative condition is cancer, more preferably a human cancer.
  • the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, for use in the manufacture of a medicament for the treatment of cancer, preferably for the treatment of human cancer.
  • the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, for use in the manufacture of a medicament for the inhibition of PARG enzyme activity.
  • the inhibition of PARG enzyme activity is selective inhibition of PARG enzyme activity over PARP1 or ARH3 enzyme activity.
  • the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, for use in the manufacture of a medicament for the selective inhibition of PARG enzyme activity over PARP1 or ARH3 enzyme activity.
  • the present invention further provides the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein for use in the manufacture of a medicament for the treatment of a disease or disorder in which PARG activity is implicated, as defined herein.
  • proliferative disorder are used interchangeably herein and pertain to an unwanted or uncontrolled cellular proliferation of excessive or abnormal cells which is undesired, such as, neoplastic or hyperplastic growth, whether in vitro or in vivo.
  • proliferative conditions include, but are not limited to, pre-malignant and malignant cellular proliferation, including but not limited to, malignant neoplasms and tumours, cancers, leukemias, psoriasis, bone diseases, fibroproliferative disorders (e.g., of connective tissues), and atherosclerosis. Any type of cell may be treated, including but not limited to, lung, colon, breast, ovarian, prostate, liver, pancreas, brain, and skin.
  • the anti-proliferative effects of the compound of formula (I) of the present invention have particular application in the treatment of human cancers (by virtue of their inhibition of PARG enzyme activity).
  • the anti-cancer effect may arise through one or more mechanisms, including but not limited to, the regulation of cell proliferation, the inhibition of angiogenesis (the formation of new blood vessels), the inhibition of metastasis (the spread of a tumour from its origin), the inhibition of invasion (the spread of tumour cells into neighbouring normal structures), or the promotion of apoptosis (programmed cell death).
  • the antiproliferative treatment with the compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined hereinbefore, may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional surgery or radiotherapy or chemotherapy.
  • Such chemotherapy may include one or more of the following categories of anti-tumour agents:
  • antiproliferative/antineoplastic drugs and combinations thereof as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblast
  • cytostatic agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestagens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5oc-reductase such as finasteride;
  • antioestrogens for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene
  • antiandrogens for example
  • anti-invasion agents for example c-Src kinase family inhibitors like 4-(6-chloro-2,3- methylenedioxyanilino)-7-[2-(4-methylpiperazin-1 -yl)ethoxy]-5-tetrahydropyran-4- yloxyquinazoline (AZD0530; International Patent Application WO 01/94341 ), N-(2-chloro-6- methylphenyl)-2- ⁇ 6-[4-(2- hydroxyethyl)piperazin-1 -yl]-2-methylpyrimidin-4-ylamino ⁇ thiazole- 5-carboxamide (dasatinib, BMS- 354825; J. Med.
  • anti-invasion agents for example c-Src kinase family inhibitors like 4-(6-chloro-2,3- methylenedioxyanilino)-7-[2-(4-methylpiperazin-1 -yl)ethoxy]-5-tetra
  • inhibitors of growth factor function include growth factor antibodies and growth factor receptor antibodies (for example the anti-erbB2 antibody trastuzumab [HerceptinTM], the anti-EGFR antibody panitumumab, the anti-erbB 1 antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al. (Critical reviews in oncology/haematology, 2005, Vol.
  • inhibitors also include tyrosine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N-(3-chloro- 4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy)quinazolin-4-amine (gefitinib, ZD1839), N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774) and 6- acrylamido-N-(3-chloro-4-fluorophenyl)-7-(3-morpholinopropoxy)-quinazolin-4-amine (Cl 1033), erbB2 tyrosine kinase inhibitors such as lapatinib); inhibitors of the hepatocyte growth factor family; inhibitors of the epidermal growth factor family; inhibitors of
  • antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (AvastinTM) and for example, a VEGF receptor tyrosine kinase inhibitor such as vandetanib (ZD6474), vatalanib (PTK787), sunitinib (SU1 1248), axitinib (AG-013736), pazopanib (GW 786034) and 4-(4-fluoro-2-methylindol-5- yloxy)-6-methoxy-7-(3-pyrrolidin-1 - ylpropoxy)quinazoline (AZD2171 ; Example 240 within WO 00/47212), compounds such as those disclosed in International Patent Applications W097/22596, WO 97/30035, WO 97/32856 and WO 98/13354 and compounds that work by other mechanisms (for example li
  • vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529, WO 00/41669, WO 01 Z92224, WO 02/04434 and WO 02/08213;
  • an endothelin receptor antagonist for example zibotentan (ZD4054) or atrasentan;
  • antisense therapies for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
  • (ix) gene therapy approaches including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multidrug resistance gene therapy; and
  • GDEPT gene-directed enzyme pro-drug therapy
  • (x) immunotherapy approaches including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies.
  • cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor
  • the antiproliferative treatment defined hereinbefore may involve, in addition to the compound of formula (I) of the invention, conventional surgery or radiotherapy or chemotherapy.Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment.
  • Such combination products employ the compounds of this invention within the dosage range described hereinbefore and the other pharmaceutically-active agent within its approved dosage range.
  • the present invention further relates to the compound of formula (I) or a pharmaceutically acceptable salt as defined herein, for use in the treatment of a cancer (for example a cancer involving a solid tumour) in combination with another anti-tumour agent.
  • the anti-tumour agent is preferably selected from the anti-tumour agents as listed hereinabove.
  • the term “combination” refers to simultaneous, separate or sequential administration. In one aspect of the invention “combination” refers to simultaneous administration. In another aspect of the invention “combination” refers to separate administration. In a further aspect of the invention “combination” refers to sequential administration. Where the administration is sequential or separate, the delay in administering the second component should not be such as to lose the beneficial effect of the combination.
  • Scheme 1 illustrates a preferred synthetic approach to compounds of the general formula A.
  • the scheme can also be extended to the compounds of formula (I) wherein W is -S(O) y NH-, -NHS(O)(NH)-, -NHS(O)(NCH 3 )-, -S(O)(NH)-NH-, or -S(O)(NCH 3 )-NH- upon the corresponding functionalization of the bromide of compound 1.
  • a compound 1 in which X 1 , X 2 , X 3 and R 7 are are as defined for the compound of formula (I) reacts with benzyl mercaptan to give compound 2 in which X 1 , X 2 , X 3 and R 7 are are as defined for formula (I).
  • This coupling reaction can be carried out by a palladium-catalyzed Carbon-Sulfur (C-S) cross-coupling reaction (see for example: Jiang, Buchwald in ‘Metal-Catalyzed Cross-Coupling Reactions’, 2 nd edition.: de Meijere, Diederich, Eds.: Wiley-VCH: Weinheim, Germany, 2004).
  • the reactions are preferably run under an atmosphere of argon for 1 - 48 hours at 80 - 100°C in a microwave oven or in an oil bath.
  • compound 2 in which X 1 , X 2 , X 3 and R 7 are as defined for the compound of formula (I) reacts with an brominating reagent to give compound 3 in which X 1 , X 2 , X 3 and R 7 are are as defined for formula (I).
  • This bromination can be carried out by treatment with A/-bromosuccinimide (NBS), Br2 etc., in MeCN, THF, dioxane, DMF etc. (see for example: Bentley et al; WO2011/138266).
  • NBS A/-bromosuccinimide
  • Pd(0) catalysts like tetrakis(triphenylphosphine) palladium(O) [Pd(PPh3)4], tris(dibenzylideneacetone) di-palladium(O) [Pd2(dba)3], or by Pd(ll) catalysts like dichlorobis(triphenylphosphine)-palladium(ll) [Pd(PPh3)2Cb], palladium(ll) acetate and triphenylphosphine or by [l,l'-bis(diphenylphosphino)ferrocene]palladium dichloride.
  • Pd(0) catalysts like tetrakis(triphenylphosphine) palladium(O) [Pd(PPh3)4], tris(dibenzylideneacetone) di-palladium(O) [Pd2(dba)3]
  • Pd(ll) catalysts like dichlorobis(triphenylphosphine
  • the reaction is preferably carried out in a solvent like 1 ,2-dimethoxyethane, dioxane, DMF, DME, THF, or isopropanol with water and in the presence of a base like potassium carbonate, sodium carbonate, sodium bicarbonate or potassium phosphate, (see for example: Hall, Boronic Acids, 2005 Wiley VCH Verlag GmbH & Co. KGaA, Weinheim, ISBN 3-527- 30991-8 and references cited therein).
  • the reaction is performed at temperatures ranging from room temperature to the boiling point of the respective solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven.
  • the reaction is preferably completed after 1 to 36 hours.
  • compound 5 in which X 1 , X 2 , X 3 , R 7 and R 8 are as defined for formula (I) reacts with chlorination reagent to give a sulfonyl chloride 6 in which X 1 , X 2 , X 3 , R 7 and R 8 are as defined for formula (I).
  • This sulfonyl chloride formation can be carried out by treatment with NCS (N- chlorosuccinimide), sulfonyl chloride, DCDMH (1 ,3-dichloro-5,5-dimethylhydantoin), CI2 etc., in MeCN with equivalent acetic acid and water (see for example: Sutton et al, WO 2021/055744). Preferred is the herein described use of DCDMH in MeCN with equivalent acetic acid and water.
  • the reactions are preferably run under an atmosphere of argon for 0.5-5 hours at 0°C to room temperature.
  • compound 6 in which X 1 , X 2 , X 3 , R 7 and R 8 are as defined for formula (I) reacts with an amine 7 in which R1 , R 2 and R 3 are as defined for formula (I) to give compound 8 in which X 1 , X 2 , X 3 , R 7 and R 8 are as defined for formula (I).
  • This reaction can be carried out under basic conditions (see for example: Guo et al, WO2013/006394). Preferred is the herein described use of trimethylamine, pyridine etc., in DCM, THF or DMF.
  • the reactions are preferably run under an atmosphere of argon for 0.5 - 24 hours at 0 °C to room temperature.
  • compound 8 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 7 and R 8 are as defined for formula (I) is converted to a hydrazide compound 9 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 7 and R 8 are as defined for formula (I).
  • This hydrazide formation is preferably carried out by treating with an amination reagent.
  • the reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 60 °C to 100 °C. (see for example: Boyles et al. Org. Progress. Res. Dev., 2002, 6, 230 - 233).
  • compound 9 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 7 and R 8 are as defined for formula (I) reacts with compound 10 in which R N is as defined for formula (I) and LG is a leaving group such as CI-, Br-, I-, MsO- or an aldehyde to give compound 11 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 7 , R 8 and R N are as defined for formula (I).
  • This alkylation is preferably carried out in basic condition. Preferred is the herein described use NaH, K2CO3 or CS2CO3 etc.in DCM, DMF or THF.
  • the alkylation is preferably run under an atmosphere of argon for 3-24 hours at 0 °C to 80 °C.
  • this reaction is preferably carried out with reductive amination reaction.
  • the reducing reagent can be, but not limited to, sodium cyanoborohydride or sodium triacetoxyborohydride.
  • the reductive amination are preferably run under an atmosphere of argon for 12-24 hours at room temperature to 80 °C.(see for example: Ong et al, US2013203686).
  • compound 11 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 7 , R 8 and R N are as defined for formula (I) reacts with compound 12 in which R oov and W oov are as defined for formula (I) and LG is a leaving group such as HO-, Cl- or -O-W oov -R oov ; to give a compound A in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 8 , R 7 , R N , R oov and W oov are as defined for formula (I).
  • this acylhydrazine formation is preferably carried out by condensation.
  • EEDQ A/-Ethoxycarbonyl-2-ethoxy-1,2- dihydroquinoline
  • CMPI 2-Chloro-1 -methylpyridinium
  • the reaction is preferably run under an atmosphere of argon for 12-36 hours at room temperature to 80 °C (see for example: Dominic et Org. Process Res.Dev. 2005, 9, 499 - 507).
  • this reaction can be carried out under basic conditions.
  • Preferred is the herein described use of triethylamine, pyridine, di-/so-propylethylamine etc in DCM or THF under an atmosphere of argon for 2- 24 hours at 0 °C to 50 °C. (see for example: Lyer et al, Chem. Communications, 2018, 54, 11021 - 11024).
  • Scheme 2 illustrates a preferred synthetic approach to compounds of the general formula B.
  • the scheme can also be extended to the compounds of formula (I) wherein W is -S(O) y NH-, -NHS(O)(NH)-, -NHS(O)(NCH 3 )-, -S(O)(NH)-NH-, or -S(O)(NCH 3 )-NH- starting from appropriate starting materials.
  • compound 13 in which X 1 , X 2 and X 3 are are as defined for formula (I) reacts with chlorosulfonic acid to give compound 14 in which X 1 , X 2 and X 3 are are as defined for formula (I).
  • chlorosulfonic acid under an atmosphere of argon, (see for example: Adams et al, W02008070707).
  • the reactions are preferably run in an oil bath for 2-24 hours at 0-140°C.
  • compound 14 in which X 1 , X 2 and X 3 are as defined for formula (I) reacts with an amine 7 in which R1 , R 2 and R 3 are as defined for formula (I) to give compound 15 in which X 1 , X 2 , X 3 , R 1 , R 2 and R 3 as defined for formula (I).
  • This reaction can be carried out under basic conditions (see for example: Guo et al, WO2013/006394). Preferred is the herein described use of trimethylamine, pyridine etc., in DCM, THF or DMF.
  • the reactions are preferably run under an atmosphere of argon for 0.5-24 hours at 0 °C to room temperature.
  • compound 15 in which X 1 , X 2 , X 3 , R 1 , R 2 and R 3 are as defined for formula (I) reacts with compound 16 in which R 7 as defined for formula (I) to give compound 17 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 7 are as defined for formula (I).
  • R 7 as defined for formula (I)
  • the conditions for this reaction can be found in: McGonagle et al, WO2016/092326.
  • compound 17 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 7 are as defined for formula (I) is converted to a hydrazide compound 18 n which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 7 are as defined for formula (I).
  • This hydrazide formation is preferably carried out by treating with amination reagent.
  • the reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 60°C to 100°C. (see for example: Boyles et al. Org. Progress. Res. Dev., 2002, 6, 230 - 233).
  • compound 18 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 7 are as defined for formula (I) reacts with compound 10 in which R N is as defined for formula (I) and LG is a leaving group such as CI-, Br-, I-, MsO- or an aldehyde to give compound 19 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 7 and R N are as defined for formula (I).
  • This alkylation is preferably carried out in basic condition. Preferred is the herein described use NaH, K2CO3 or CS2CO3 etc.in DCM, DMF or THF.
  • the alkylation is preferably run under an atmosphere of argon for 3-24 hours at 0 °C to 80 °C.
  • this reaction is preferably carried out with reductive amination reaction.
  • the reducing reagent can be, but not limited to, sodium cyanoborohydride or sodium triacetoxyborohydride.
  • the reductive amination are preferably run under an atmosphere of argon for 12-24 hours at room temperature to 80 °C.(see for example: Ong et al, US2013203686).
  • compound 19 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 7 and R N are as defined for formula (I) reacts with compound 12 in which R oov and W oov are as defined for formula (I) and LG is leaving group such as HO-, Cl- or -O-W oov -R oov to give compound B in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 7 , R N , R oov and W oov are as defined for formula (I).
  • LG is HO-, this acylhydrazine formation is preferably carried out by condensation.
  • CMPI 2-chloro-1 -methylpyridinium
  • this reaction can be carried out under basic conditions.
  • Scheme 3 illustrates an alternative pathway to access to compounds of the general formula 17.
  • the scheme can also be extended to compounds of formula (I) wherein W is -S(O) y NH-, -NHS(O)(NH)-, -NHS(O)(NCH 3 )-, -S(O)(NH)-NH-, or -S(O)(NCH 3 )-NH-.
  • a compound of formula 15 in which X 1 , X 2 , X 3 , R 1 , R 2 and R 3 are as defined for the compound of formula (I) is converted to a compound of formula 18 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 are as defined for the compound of formula (I).
  • This amide formation can be carried out with ammonium hydroxide or ammonium salt and base such as triethylamine, pyridine, di-/so-propylethylamine etc.
  • HATU 1-[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5- b]pyridinium 3-Oxide Hexafluorophosphate
  • EDCI/HOBt N-(3-Dimethylaminopropyl)-N'- ethylcarbodiimide hydrochloride/ Hydroxybenzotriazole
  • T3P T3P
  • GDI di(1 H-imidazol-1-yl)methanone
  • DCM or DMF see for example: Weaver et al, W02020/257940.
  • This amide formation can also be conducted by acyl chloride strategy.
  • the reactions are preferably run under an atmosphere of argon for 2-24 hours at room temperature.
  • compound 20 in which X 1 , X 2 , X 3 , R 1 , R 2 and R 3 are as defined for formula (I) reacts with compound 19 in which R 7 is as defined for formula (I) to give compound 17 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 7 are as defined forformula (I).
  • R 7 is as defined for formula (I) to give compound 17 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 7 are as defined forformula (I).
  • the conditions ofthis reaction can be found for example in: Mcgonagle et al, WO2016/092326.
  • Scheme 4 illustrates a preferred synthetic approach to compounds of the general formula C.
  • the scheme can also be extended to the compounds of formula (I) wherein W is -S(O) y NH-, -NHS(O)(NH)-, -NHS(O)(NCH 3 )-, -S(O)(NH)-NH-, or -S(O)(NCH 3 )-NH- starting from appropriate starting materials.
  • compound 22 in which X 1 , X 2 and X 3 are as defined for formula (I) reacts with amine 7 in which R 1 ’ R 2 and R 3 are as defined for formula (I) to give compound 23 in which X 1 , X 2 , X 3 , R 1 , R 2 and R 3 are as defined for formula (I).
  • This reaction can be carried out under basic conditions, (see for example: Guo et al, WO2013/006394). Preferred is the herein described use of triethylamine, pyridine, di-/so- propylethylamine etc in DCM, THF or DMF.
  • the reactions are preferably run under an atmosphere of argon for 0.5 - 24 hours at 0 °C to room temperature.
  • compound 23 in which X 1 , X 2 , X 3 , R 1 , R 2 and R 3 are as defined for formula (I) reacts with amine 24 in which R 8 is as defined for formula (I) to give compound 25 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I).
  • R 8 is as defined for formula (I) to give compound 25 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I).
  • Preferred is the herein described use of triethylamine, di-/so- propylethylamine etc in DMF, acetonitrile or dioxane, (see for example: Liu et al, Eur. J. Med. Chem., 2021 , 222, 113565).
  • the reactions are preferably run under an atmosphere of argon for 2 - 24 hours
  • compound 25 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I) is converted to compound 26 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I).
  • This amide formation can be carried out with ammonium hydroxide or ammonium salt and base such as triethylamine, pyridine, di-7so-propylethylamine etc.
  • compound 26 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I) is converted to compound 27 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I).
  • This cyclization is preferably carried out with GDI in the presence of base such as triethylamine, di-/so- propylethylamine etc. in DMF, NMP or DMA.
  • the reactions are preferably run for 0.5 - 16 hours at 80 - 120°C in a microwave oven or in an oil bath (see for example: Velaparthi et al, WO2021/133751).
  • This cyclization also can be carried out with GDI in DMF, NMP or DMA etc. without base at 100 - 150°C, or with triphosgene and base in DCM, THF etc. at 0 °C to room temperature.
  • compound 27 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I) is converted to hydrazide compound 28 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I).
  • This hydrazide formation is preferably carried out by treating with amination reagent. Preferred is the herein described use of O-(4-nitrophenyl)hydroxylamine or O-(2,4-dinitrophenyl)hydroxylamine with potassium carbonate in DMF and dioxane.
  • the reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 60 °C to 100 °C (see for example: Boyles et al, Eur. J. Med. Chem.,Org. Pro. Res. Dev., 2002, 6, 230 - 233).
  • compound 28 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I) reacts with compound 10 in which R N is as defined for formula (I) and LG is a leaving group such as CI-, Br-, I-, MsO- or an aldehyde to give compound 29 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 8 and R N are as defined for formula (I).
  • This alkylation is preferably carried out in basic condition.
  • the alkylation is preferably run under an atmosphere of argon for 3-24 hours at 0 °C to 80 °C.
  • LG is aldehyde group
  • this reaction is preferably carried out with reductive amination reaction.
  • the reducing reagent can be, but not limited to, sodium cyanoborohydride or sodium triacetoxyborohydride.
  • the reductive amination are preferably run under an atmosphere of argon for 12-24 hours at room temperature to 80 °C.(see for example: Ong et al, US2013203686).
  • compound 29 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 8 and R N are as defined for formula (I) reacts with compound 12 in which R oov and W oov are as defined for formula (I) and LG is leaving group such as HO-, Cl- or -O-W oov -R oov to give compound C in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 8 , W, R N , R oov and W oov are as defined for formula (I).
  • LG is HO-, this acylhydrazine formation is preferably carried out by condensation.
  • EEDQ A/-Ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline
  • CMPI 2-Chloro-1 -methylpyridinium
  • the reaction is preferably run under an atmosphere of argon for 12-36 hours at room temperature to 80 °C (see for example: Dominic et Org. Process Res. Dev. 2005, 9, 499 - 507).
  • this reaction can be carried out under basic conditions.
  • Scheme 5 illustrates an alternative pathway to access to compounds of the general formula 26.
  • the scheme can also be extended to the compounds of formula (I) wherein W is -S(O) y NH-, -NHS(O)(NH)-, -NHS(O)(NCH 3 )-, -S(O)(NH)-NH-, or -S(O)(NCH 3 )-NH-.
  • compound 30 in which X 1 , X 2 and X 3 are as defined for formula (I) reacts with chlorosulfonic acid to give compound 31 in which X 1 , X 2 and X 3 are as defined for formula (I).
  • chlorosulfonic acid under an atmosphere of argon (see for example: Adams et al, W02008070707).
  • the reactions are preferably run in an oil bath for 2 - 24 hours at 0 - 140°C.
  • compound 31 in which X 1 , X 2 and X 3 are as defined for formula (I) reacts with compound 7 in which R 1 , R 2 and R 3 are as defined for formula (I) to give compound 32 in which X 1 , X 2 , X 3 , R 1 , R 2 and R 3 are as defined for formula (I).
  • This reaction can be carried out under basic conditions (see for example: Sari et al, Eur. J. Med. Chem., 2017, 138, 407 - 421). Preferred is the herein described use of triethylamine, pyridine, di-/so-propylethylamine etc. in DCM, THF or DMF.
  • the reactions are preferably run under an atmosphere of argon for 0.5 - 24 hours at 0 °C to room temperature.
  • compound 32 in which X 1 , X 2 , X 3 , R 1 , R 2 and R 3 are as defined for formula (I) reacts with amine 24 in which R 8 is as defined for formula (I) to give compound 26.
  • amine 24 in which R 8 is as defined for formula (I) to give compound 26.
  • the reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 80 - 110°C in a microwave oven or in an oil bath.
  • Scheme 6 illustrates a preferred synthetic approach to compounds of the general formula D.
  • the scheme can also be extended to the compounds of formula (I) wherein W is -S(O) y NH-, -NHS(O)(NH)-, -NHS(O)(NCH 3 )-, -S(O)(NH)-NH-, or -S(O)(NCH 3 )-NH- starting from appropriate starting materials.
  • compound 33 in which X 1 , X 2 and X 3 are as defined for formula (I) reacts with compound 34 in which R 8 is as defined for formula (I) to give compound 35 in which X 1 , X 2 , X 3 and R 8 are as defined for formula (I).
  • R 8 is as defined for formula (I)
  • the condition of this reaction can be found for example in Mcgonagle et al, WO2016/092326.
  • compound 35 in which X 1 , X 2 , X 3 and R 8 are as defined for formula (I) reacts with hydrazine hydrate to give compound 36 in which X 1 , X 2 , X 3 and R 8 are as defined for formula (I).
  • the condition of this reaction can be found for example in Mcgonagle et al, WO2016/092326.
  • compound 36 in which X 1 , X 2 , X 3 and R 8 are as defined for formula (I) reacts with benzyl mercaptan to give compound 37 in which X 1 , X 2 , X 3 and R 8 are as defined for formula (I).
  • This coupling reaction can be carried out by a palladium-catalyzed carbon-sulfur (C-S) cross-coupling reaction (see for example: Jiang, Buchwald in ‘Metal-Catalyzed Cross-Coupling Reactions’, 2 nd edition.: de Meijere, Diederich, Eds.: Wiley-VCH: Weinheim, Germany, 2004).
  • the reactions are preferably run under an atmosphere of argon for 1 - 48 hours at 80 - 100°C in a microwave oven or in an oil bath.
  • compound 37 in which X 1 , X 2 , X 3 and R 8 are as defined for formula (I) reacts with a chlorination reagent to give sulfonyl chloride compound 38 in which X 1 , X 2 , X 3 and R 8 are as defined for formula (I).
  • This sulfonyl chloride formation can be carried out by treatment with NCS, sulfonyl chloride, DCDMH, CI2 etc., in MeCN with equivalent acetic acid and water (see for example: Sutton et al, WO 2021/055744).
  • the reactions are preferably run under an atmosphere of argon for 0.5 - 5 hours at 0 °C to room temperature.
  • compound 38 in which X 1 , X 2 , X 3 , R 8 are as defined for formula (I) reacts with amine 7 in which R 1 ’ R 2 and R 3 are as defined for formula (I) to give compound 39 in which X 1 , X 2 , X 3 , R 1 ’ R 2 , R 3 and R 8 are as defined for formula (I).
  • This reaction can be carried out under basic conditions (see for example: Guo et al, WO2013/006394). Preferred is the herein described use of trimethylamine, pyridine etc., in DCM, THF or DMF.
  • the reactions are preferably run under an atmosphere of argon for 0.5 - 24 hours at 0 °C to room temperature.
  • compound 39 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I) is converted to a hydrazide compound 40 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I).
  • This hydrazide formation is preferably carried out by treating with amination reagent. Preferred is the herein described use of O-(4-nitrophenyl)hydroxylamine or O-(2,4-dinitrophenyl)hydroxylamine with potassium carbonate in DMF and dioxane.
  • the reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 60 °C to 100 °C (see for example: Boyles et al, Eur. J. Med. Chem.,Org. Pro. Res. Dev., 2002, 6, 230 - 233).
  • compound 40 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 and R 8 are as defined for formula (I) reacts with compound 10 in which R N is as defined for formula (I) and LG is a leaving group such as CI-, Br-, I-, MsO- or an aldehyde to give compound 41 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 8 and R N are as defined for formula (I).
  • This alkylation is preferably carried out in basic condition.
  • the alkylation is preferably run under an atmosphere of argon for 3-24 hours at 0 °C to 80 °C.
  • LG is aldehyde group
  • this reaction is preferably carried out with reductive amination reaction.
  • the reducing reagent can be, but not limited to, sodium cyanoborohydride or sodium triacetoxyborohydride.
  • the reductive amination are preferably run under an atmosphere of argon for 12-24 hours at room temperature to 80 °C.(see for example: Ong et al, US2013203686).
  • compound 41 in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 8 and R N are as defined for formula (I) reacts with compound 12 in which R oov and W oov are as defined for formula (I) and LG is leaving group such as HO-, Cl- or -O-W oov -R oov to give a compound D in which X 1 , X 2 , X 3 , R 1 , R 2 , R 3 , R 8 , R N , R oov and W oov are as defined for formula (I).
  • LG is HO-, this acylhydrazine formation is preferably carried out by condensation.
  • CMPI 2-Chloro-1 -methylpyridinium
  • this reaction can be carried out under basic conditions.
  • the compounds described in this section are defined by their chemical formulae and their corresponding chemical names.
  • the present invention relates to both the compound defined by the chemical formula and the compound defined by the chemical name, and particularly relates to the compound defined by the chemical formula.
  • Method 1 SHIMADZU LCMS-2020 Kinetex EVO C18 2.1X30 mm, 5 urn at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5 mL/min; eluted with the mobile phase over 0.80 min employing UV detection at 220 nm and 254 nm.
  • Method 2 SHIMADZU LCMS-2020 Kinetex EVO C18 2.1X30 mm, 5 urn at 40°C ;
  • Mobile Phase A: 0.025% NH3-H2O in water (v/v) , B: MeCN; flow rate held at 1 .5 mL/min; eluted with the mobile phase over 1.55 min employing UV detection at 220 nm and 254 nm.
  • Gradient information 0-0.80 min, ramped from 95% A-5% B to 5% A-95% B; 0.80-1 .20 min, held at 5% A-95% B; 1 .20-1 .21 min, returned to 95% A-5% B, 1.21-1 .55 min, held at 95% A-5% B.
  • Method 3 Agilent 1200 ⁇ G6110A Kinetex EVO C18 2.1X30 mm, 5 urn at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5.0 mL/min; eluted with the mobile phase over 0.80 min employing UV detection at 220 nm and 254 nm.
  • Method 4 SHIMADZU LCMS-2020 Kinetex® EVO C18 2.1X30 mm 5 urn at 50°C
  • A 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5 mL/min; eluted with the mobile phase over 1.00 min employing UV detection at 220 nm and 254 nm.
  • Gradient information 0.01-0.80 min, ramped from 95% A-5% B to 5% A-95% B; 0.80-0.95 min, held at 5% A-95% B; 0.95-0.96 min, returned to 95% A-5% B, 0.96-1 .00 min, held at 95% A-5% B.
  • Method 5 SHIMADZU LCMS-2020 Kinetex® EVO C18 2.1X20 mm 2.6 urn at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 2.0 mL/min; eluted with the mobile phase over 1.00 min employing UV detection at 220 nm and 254 nm. Gradient information: 0.01-0.60 min, ramped from 95% A-5% B to 5% A-95% B; 0.61-0.78 min, held at 5% A-95% B; 0.78-0.79 min, returned to 95% A-5% B, 0.79-0.80 min, held at 95% A-5% B.
  • Method 6 SHIMADZU LCMS-2020 Kinetex® EVO C18 2.1X30 mm 5 urn at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5 mL/min; eluted with the mobile phase over 1.00 min employing UV detection at 220 nm and 254 nm. Gradient information: 0.01-0.60 min, ramped from 95% A-5% B to 5% A-95% B; 0.60-0.78 min, held at 5% A-95% B; 0.78-0.79 min, returned to 95% A-5% B, 0.79-0.80 min, held at 95% A-5% B.
  • Method 7 SHIMADZU LCMS-2020 Kinetex EVO C18 2.1X30mm,5um at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 2.0 mL/min; eluted with the mobile phase over 0.80 min employing UV detection at 220 nm and 254 nm. Gradient information: 0- 0.80 min, ramped from 95% A-5% B to 5% A-95% B; 0.80-1 .20 min, held at 5% A-95% B; 1 .20-1 .21 min, returned to 95% A-5% B, 1.21-1 .55 min, held at 95% A-5% B.
  • Method 8 SHIMADZU LCMS-2020 Kinetex EVO C18 2.1X30mm,5pm at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5 mL/min; eluted with the mobile phase over 1 .55 min employing UV detection at 220 nm and 254 nm.
  • 1 H NMR spectra were acquired on a Bruker Avance IH spectrometer at 400 MHz using residual undeuterated solvent as reference. 1 H NMR signals are specified with their multiplicity / combined multiplicities as apparent from the spectrum; possible higher-order effects are not considered. Chemical shifts of the signals (5) are specified as ppm (parts per million).
  • the mixture was stirred at 100 °C for 8 h.
  • the reaction mixture was adjusted to pH ⁇ 6 with aqueous HCl solution (1N).
  • the mixture was extracted with EtOAc (100 mL, 2x).
  • the combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue, which was triturated with petroleum ether/ ethyl acetate 10:1 (30 mL) at 20 °C for 10 min.
  • Example 7 2-cyano-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acetamide
  • 2-cyanoacetic acid 8.99 mg, 105.70 ⁇ mol
  • THF 0.5 mL
  • ethyl 2- ethoxy-2H-quinoline-1-carboxylate 26.14 mg, 105.70 ⁇ mol
  • Example 8 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)-2-(trifluoromethyl)acrylamide
  • 2-(trifluoromethyl) acrylic acid 14.80 mg, 105.70 ⁇ mol
  • THF 0.5 mL
  • ethyl 2-ethoxy-2H-quinoline-1-carboxylate 26.14 mg, 105.70 ⁇ mol
  • Example 10 N-methyl-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acrylamide
  • 3-(methylamino)-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl) methyl)-2,4- dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (10 mg, 25.48 ⁇ mol) in THF (0.3 mL) was added NaHCO3 (21.40 mg, 254.80 ⁇ mol) in H2O (0.3 mL), followed by acryloyl chloride (23.06 mg, 254.80 ⁇ mol, 20.78 ⁇ L) at 0 °C.
  • reaction mixture was stirred at 0 °C for 1 h, then, diluted with H2O (10mL) and extracted with EtOAc (10 mL; 2x). The combined organic layer was washed with brine (10mL; 2x), dried over Na2SO4 and concentrated under reduced pressure.
  • Example 12 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)methacrylamide
  • TEA 40.11 mg, 396.36 ⁇ mol
  • methacrylic anhydride 28.52 mg, 184.97 ⁇ mol
  • Example 14 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)pent-2-ynamide
  • pent-2-ynoic acid 10.37 mg, 105.70 ⁇ mol
  • THF 2 mL
  • ethyl 2- ethoxyquinoline-1(2H)-carboxylate 26.14 mg, 105.70 ⁇ mol
  • Example 16 3-cyclopropyl-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo- 1,4-dihydroquinazolin-3(2H)-yl)propiolamide
  • 3-cyclopropylpropiolic acid 5.82 mg, 52.84 ⁇ mol
  • THF 0.5 mL
  • ethyl 2-ethoxyquinoline-1(2H)-carboxylate 13.07 mg, 52.84 ⁇ mol
  • the residue was purified by reversed-phase flash (ISCO®; 330 g Flash Coulmn Welch Ultimate XB_C1820-40 ⁇ m; 120 A, Eluent of 8 ⁇ 50% ACN/H2O (0.1% HCl condition) @ 100 mL/min).
  • the ACN of the resulting solution was concentrated under vacuum, and the aqueous layer was adjusted to pH>7 with sat. NaHCO3 solution.
  • the solution was extracted with EtOAc (100 mL; 2x).
  • the organic phase was washed with brine (100 mL; 2x).
  • Example 17 N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)but-2-ynamide
  • ethyl 2- ethoxyquinoline-1(2H)-carboxylate 95.00 mg, 384.17 ⁇ mol
  • Example 20 N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)acrylamide
  • NaHCO 3 36.88 mg, 439.06 ⁇ mol
  • H 2 O 0.5 mL
  • 3- amino-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide 40 mg, 109.76 ⁇ mol
  • THF 0.5 mL
  • reaction mixture was stirred at 20 °C for 1 h and pyridine (366.15 mg, 4.63 mmol, 373.62 ⁇ L) was added.
  • the combined organic layer was dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum to give the product 5-(N- (bicyclo[1.1.1]pentan-1-yl)sulfamoyl)-2-fluorobenzamide (550 mg, 1.93 mmol, 83.58% yield) as a yellow solid.
  • Step 1 To a solution of methyl 3-chlorocyclobutanecarboxylate (1 eq) in THF (100 mg/mL), which was degassed and purged with N2 (3x), was added drop-wise LiHMDS in THF (1 M, 1 .0 -1 .5 eq) at 0 °C. The solution was stirred at 0 °C for 0.5 to 3 h under a N2 atmosphere leading to a solution of methyl bicyclo [1 .1 .0] butane- 1 -carboxylate.
  • Step 2 Then, this newly prepared methyl bicyclo[1.1.0]butane-1 -carboxylate solution (1.0-1.5 eq) was added to a solution of the hydrazide compound (1eq) in THF (100 mg/L) previously cooled to 0°C and LiHMDS in THF (1 M, 2.0-3.5 eq) was added at 0 °C The resulting mixture was heated to 20-40 °C and stirred for additional 1 ⁇ 3 h. The reaction mixture was quenched with NH4Cl (aq., sat.) and extracted with EtOAc.
  • the resulting mixture was stirred at 20 °C for 2 h then heated at 40 °C and stirred for another 1 h.
  • the mixture was poured into NH4Cl (aq., sat., 10 mL).
  • the aqueous layer was extracted with EtOAc (3 mL, 3x).
  • the combined organic layer was washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum.
  • the mixture was stirred at 80°C for 14 h.
  • the reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue.
  • the crude product was purified by reversed-phase flash (ISCO ® ; 48 g Flash Column Welch Ultimate XB_C1820-40 ⁇ m; 120 A, Eluent of 5 ⁇ 95% ACN/H2O (0.1% FA condition) @ 80 mL/min).
  • the desired fraction was lyophilized to give the product 2-(((2,2-difluorocyclopropyl)methyl)amino)-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (200 mg, 556.50 ⁇ mol, 37.88% yield) as a white solid.
  • the crude product was purified by preparative-HPLC (column: Phenomenex C18150*25 mm*10 ⁇ m; mobile phase: A: 0.05% NH3 ⁇ H2O in water; B: MeCN; B%: 22%-52%, 8 min).
  • Example 27 N-(1-((2,4-dimethylthiazol-5-yl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
  • the reaction was conducted according to the general procedure 1 starting from 3-amino-1-((2,4- dimethylthiazol-5-yl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide.
  • Example 28 N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)-N-methylbicyclo[1.1.0]butane-1-carboxamide
  • the reaction was conducted according to the general procedure 1 starting from 3-(methylamino)- N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide.
  • Example 30 N-(1-methyl-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)bicyclo[1.1.0]butane-1-carboxamide
  • the reaction was conducted according to the general procedure 1 starting from 3-amino-1-methyl- N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide.
  • n-BuLi 2.5 M, 719.94 ⁇ L
  • n-BuLi 2.5 M, 719.94 ⁇ L
  • the mixture was stirred at - 78 °C for 15 min.
  • the mixture was warmed to 0 °C and TIPSCl (115.67 mg, 599.95 ⁇ mol, 128.38 ⁇ L) was added dropwise via syringe over 1 min and the mixture was stirred at 0 °C for 1.5 h.
  • the reaction mixture was quenched with NH4Cl (aq., sat., 10 mL) at 0 °C and extracted with EtOAc (15 mL, 3x).
  • reaction mixture was diluted with water (10 mL) at 20 °C and extracted with EtOAc (15 mL, 3x). The combined organic layer was washed with brine (20 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate concentrated under reduced pressure.
  • Example 33b RT 0.292 min (Method 7); m/z 391.3 (M+H) + (ESI + ); 1 H NMR (400 MHz, DMSO-
  • Example 34 N-(1-(cyclopentylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
  • the reaction was conducted according to the general procedure 1 starting from 3-amino-1- (cyclopentylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide.
  • Example 36 N-(1-(cyclohexylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
  • the reaction was conducted according to the general procedure 1 starting from 3-amino-1- (cyclohexylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide.
  • Example 37 N-(1-(cyclopropylmethyl)-7-methyl-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acrylamide
  • 3-amino-1-(cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (30.00 mg, 79.27 ⁇ mol) in THF (1 mL) and H2O (1 mL) was added NaHCO3 (59.93 mg, 713.45 ⁇ mol), followed by the addition of prop-2-enoyl chloride (14.35 mg, 158.54 ⁇ mol) at 0 °C.
  • the product solution was lyophilized to give the product N-(1-(cyclopropylmethyl)-7-methyl-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)acrylamide (7.87 mg, 17.81 ⁇ mol, 22.46% yield, 97.85% purity) as a yellow solid.
  • Exemplary compounds of formula (I) were tested in selected biological and/or physicochemical assays one or more times.
  • data are reported as either average values or as median values, wherein the average value, also referred to as the arithmetic mean value, represents the sum of the values obtained divided by the number of times tested, and the median value represents the middle number of the group of values when ranked in ascending or descending order. If the number of values in the data set is odd, the median value is the middle value. If the number of values in the data set is even, the median is the arithmetic mean of the two middle values.
  • the in vitro pharmacological, pharmacokinetic and physicochemical properties of the compounds can be determined according to the following assays and methods.
  • Protein was purified by IMAC and SEC: frozen cell pellets (typically 40 g wet weight) were resuspended by homogenization in 5 volumes buffer A (25 mM Tris/HCI pH 8.0, 200 mM NaCI, 2 mM DTT), supplemented with 1 mg of DNase I from bovine pancreas (Sigma-Aldrich) and protease inhibitors (Roche CompleteTM EDTA-free protease inhibitor tablet), and lysed by passage through a Constant Systems BasicZ homogenizer.
  • buffer A 25 mM Tris/HCI pH 8.0, 200 mM NaCI, 2 mM DTT
  • protease inhibitors Roche CompleteTM EDTA-free protease inhibitor tablet
  • the lysate was clarified by centrifugation for 60 minutes at 25,000 g, 4°C, and the lysate supernatant was loaded onto 5 ml StrepTrap HP (Cytiva) pre-equilibrated with buffer A.
  • the column was washed with buffer A ( ⁇ 10 CV), then buffer B containing 1 M KCI ( ⁇ 5 CV), and then the protein was eluted with buffer A containing 2.5 mM d-Desthiobiotin. Pooled fractions containing 6HisTwinStrep-TEV-hPARG were incubated with TEV protease overnight at 4°C.
  • hPARG was separated from uncleaved material and Thrombin protease through gel filtration with Superdex75 sizing column (GE Healthcare) pre-equilibrated with SEC buffer (15 mM Tris/HCI pH 8.5, 100 mM NaCI, 2 mM DTT). Pooled fractions containing pure hPARG were concentrated using a 10 k MWCO spin concentrator (VivaSpin) to 10 mg/mL, and then either used immediately for crystallisation or snap-frozen in liquid nitrogen for storage at -80°C.
  • SEC buffer 15 mM Tris/HCI pH 8.5, 100 mM NaCI, 2 mM DTT.
  • Protein was purified by IMAC and SEC: frozen cell pellets (typically 40 g wet weight) were resuspended by homogenization in 5 volumes buffer A (25 mM Tris/HCI pH 8.0, 200 mM NaCI, 2 mM DTT), supplemented with 1 mg of DNase I from bovine pancreas (Sigma-Aldrich) and protease inhibitors (Roche CompleteTM EDTA-free protease inhibitor tablet), and lysed by passage through a Constant Systems BasicZ homogenizer.
  • buffer A 25 mM Tris/HCI pH 8.0, 200 mM NaCI, 2 mM DTT
  • protease inhibitors Roche CompleteTM EDTA-free protease inhibitor tablet
  • the lysate was clarified by centrifugation for 60 minutes at 25,000 g, 4°C, and the lysate supernatant was loaded onto 5 ml StrepTrap HP (Cytiva) pre-equilibrated with buffer A.
  • hPARG C872A was separated from uncleaved material and Thrombin protease through gel filtration with Superdex75 sizing column (GE Healthcare) preequilibrated with SEC buffer (15 mM Tris/HCI pH 8.5, 100 mM NaCI, 2 mM DTT). Pooled fractions containing pure hPARG C872A were concentrated using a 10 k MWCO spin concentrator (VivaSpin) to 10 mg/mL, and then snap-frozen in liquid nitrogen for storage at -80°C.
  • PARG enzyme as incubated with compound or vehicle (DMSO) for 15 minutes or 2 hours in a 384 well plate. After adding the PARG substrate ADP-ribose-pNP, the plate was read for absorbance intensity at 405 nm.
  • the vehicle (DMSO) with high absorbance intensity represents no inhibition of enzymatic reaction while the low control (no enzyme) with low absorbance intensity represents full inhibition of enzymatic reaction.
  • Substrate ADP-pNP, 800 pM, Jena Bioscience catalog # NU-955
  • Assay buffer 50 mM Tris-HCI pH 8.0, 100 mM NaCI, 2 mM DTT
  • the ability of compounds to inhibit PARG in response to DNA damage was assessed with U2OS cells pretreated with the compounds for 1 hour, following a 1 -hour treatment with or without the DNA alkylating agent temozolomide (TMZ).
  • TTZ DNA alkylating agent temozolomide
  • the cells were harvested and fixed in 70% ethanol, rehydrated with glucose and EDTA in PBS and subsequently blocked for 1 hour with PBS 1 % BSA and 0.01% Tween-20 (PBT).
  • PBT PBS 1 % BSA and 0.01% Tween-20
  • the cells were incubated for 2 hours at room temperature with a mouse monoclonal antibody against poly (ADP) ribose (PAR) polymer.
  • the cells were washed and incubated with an anti-mouse Alexa-488 conjugated secondary antibody for 1 hour at room temperature.
  • NCIH-460 as a PARG-inhibition sensitive cell line and U2OS as PARG-inhibition insensitive cell line were plated at 1000 cells/well and 2000 cells/well, respectively, in 96-well white plates with clear flat bottom. After 24 hours, the compounds were added with the Tecan digital dispenser (D300e) in duplicates. The outer wells of the plate were excluded. After 96 hours of incubation, 150 pl of the growth medium were removed and 50 pl of Cell Titer-Gio (Promega) were added per well. Following an incubation of 10 minutes, luminescence was read using a plate reader (Tecan). Averaged values of the samples were normalized to DMSO treated control samples. Curves were fit as % of the control vs. log of the compound concentration using a 4 parameter log-logistic function:
  • Table 2 Inhibition of PARG by compounds according to the present invention and cellular activity of compounds according to the present invention.
  • the ICso (inhibitory concentration at 50% of maximal effect) values are indicated in pM, empty space means that the corresponding compounds have not been tested in the respective assay.
  • I C50 in pM determined in PARG enzymatic assay (PARG C872A protein and 2 hours incubation) described under PARG enzymatic IC50 assay.
  • the Kinetic solubility assay employs the shake flask method followed by HPLC-UV analysis.
  • the kinetic solubility was measured according to the following protocol:
  • Test compounds and controls (10 mM in DMSO, 10 ⁇ L/tube) were added into the buffer (490 ⁇ L/well) which placed in a Minni-Uniprep filter.
  • the buffer was prepared as the customer’s requirement.
  • Caco-2 cells purchased from ATCC were seeded onto polyethylene membranes (PET) in 96- well BD Insert plates at 1 x 105 cells/ cm2, and refreshed medium every 4 ⁇ 5 days until to the 21st to 28th day for confluent cell monolayer formation.
  • PET polyethylene membranes
  • the quality of the monolayer is verified by measuring the Unidirectional (A— >B) permeability of fenoterol/nadolol (low permeability marker), propranolol/metopronolol (high permeability marker) and Bi-directional permeability of Digoxin (a P-glycoprotein substrate marker) in duplicate wells.
  • bi-directional transport including A ⁇ B and B ⁇ A;
  • MMS Microsome metabolic stability
  • the stability of the exemplary compounds was measured in the microsome metabolic stability assay as follows:
  • Test compounds will be incubated at 37°C with liver microsomes (pooled from multiple donors) at 1 pM in the presence of a NADPH regenerating system at 0.5 mg/ml microsomal protein.
  • Positive controls include Testosterone (3A4 substrate), Propafenone (2D6) and Diclofenac (2C9). They will be incubated with microsomes in the presence of a NADPH regenerating system.
  • int(mic) 0.693/half life/mg microsome protein per mLwt: 40 g/kg, 30 g/kg, 32 g/kg, 20 g/kg and
  • test compound is assessed based on peak area ratios of analyte/IS (no standard curve).

Abstract

The present invention relates to a compound of formula (I) or an enantiomer, diastereoisomer, tautomer, pharmaceutically acceptable solvate, pharmaceutically acceptable crystal form, or pharmaceutically acceptable salt thereof. The present invention further relates to the compound of formula (I) of the present invention for use in therapy. Instant compounds are particularly useful as PARG inhibitors, preferably as covalent PARG inhibitors, and can be used in a method of treatment of a proliferative disorder, preferably of cancer.

Description

2.4-DIOXO-1 ,4-DIHYDROQUINAZOLINE DERIVATIVES AS PARG INHIBITORS FOR THE TREATMENT OF CANCER
Field of the invention
The present invention relates to a compound of formula (I):
Figure imgf000002_0001
or a pharmaceutically acceptable salt or a prodrug thereof. The present invention further relates to the compound of formula (I) of the present invention for use in therapy. Instant compounds are particularly useful as PARG inhibitors, preferably as covalent PARG inhibitors, and can be used in a method of treatment of a proliferative disorder, preferably of cancer.
Background of the invention
Cancer is a leading cause of death worldwide. Although progression-free survival and overall survival of cancer patients has improved over the past two decades, millions of cancer patients still have few therapeutic options and poor survival outcomes (Jemal et al., J. Natl. Cancer Inst. 2017, 109, 1975).
DNA replication stress (DRS) is a hallmark of cancer cells and a major source of genomic instability (a) Halazonetis et al., Science 2008, 319, 1352; b) Negrini et al., Nat. Rev. Mol. Cell Biol. 2010, 11 , 220). In broad terms, DRS refers to the deregulation of DNA replication and cell cycle progression. DRS can be induced from endogenous or exogenous causes such as oncogene activation and chemotherapeutics, respectively (Zeman and Cimprich, Nat. Cell Biol. 2013, 16, 2). At the level of the replication fork, DRS leads to replication fork stalling, disengagement of the replisome and eventually collapse. Several DNA repair proteins are involved in replication fork stability, protection, and restart under DRS conditions (a) Costantino et al., Science 2014, 343, 88; b) Scully et al., Curr. Opin. Genet. Dev. 2021 71 , 154).
Poly(ADP)ribosylation (PARylation) is a transient and reversible post-translational modification that occurs at DNA damaged sites and is catalyzed by the poly (ADP-ribose) polymerase (PARP) family of proteins (Cohen and Chang, Nat. Chem. Biol. 2018, 14, 236). PARylation of various DNA repair proteins leads to their activation. Degradation of the poly(ADP) ribose chains is mediated primarily by the poly(ADP-ribose) glycohydrolase (PARG) protein. DNA damage dependent PARylation/dePARylation is a rapid and dynamic process which needs to be well regulated since imbalances between the two processes can lead to DNA damage.
Human PARG encodes a 111 kDa protein of 976 amino acids. It contains a N-terminal regulatory domain, a catalytic domain and an ADP-ribose binding macrodomain. Five human PARG transcripts have been identified. Full length PARG is mostly nuclear; the smaller isoforms localize primarily to the cytoplasm. PARG functions primarily as an exo-hydrolase and it releases mainly mono(ADP-ribose) by hydrolyzing the a-O-glycosidic ribose-ribose bond in PAR. PARG can also act as an endo-hydrolase. PARG preferentially degrades long and linear PAR chains whereas its activity with small and branched PAR chains is significantly reduced (O’Sullivan et al., Nat. Commun. 2019, 10, 1182).
Although PARG is the dominant cellular PAR degrading enzyme, it cannot act on the terminal protein-ribose bond. Additional hydrolases such as terminal ADP-ribose protein glycohydrolase (TARG1) and ADP-ribosylhydrolase 3 (ARH3) are also known to catalyze PAR-degradation. TARG1 and ARH3 complete the reversal of PARylation by removing protein-bound mono(ADP-ribose) moieties (a) Fontana et al., Elife 2017, doi: 10.7554/eLife.28533; b) Rack et al., Genes Dev. 2020, 34, 263). TARG1 is located in the nucleus and cytoplasm. ARH3 is found primarily in the cytoplasm but it can also be found in the mitochondria and in the nucleus (Rack et al., Genes Dev. 2020, 34, 263).
Genomic aberrations targeting tumor suppressor genes or oncogenes, often make cancer cells dependent on specific DNA repair pathways. For instance, it is well known that PARP inhibitors are particularly effective against tumors carrying mutations in the BRCA1 and BRCA2 genes (a) Bryant et al., Nature 2005, 434, 913; b) Farmer et al., Nature 2005, 434, 917). Targeting synthetic lethal interactions like the one between PARP and BRCA is an attractive novel therapeutic approach for cancer treatment.
PARG participates in DNA replication and in various DNA repair mechanisms including singlestrand break (SSB) repair and replication fork restart. PARG inhibitors have shown synthetic lethal phenotype in cells with high levels of DRS caused by low expression of genes involved in DNA replication and/or replication fork stability (Pillay et al., Cancer Cell. 2019, 35, 519). Moreover, PARG inactivation, depletion or inhibition sensitizes cells to irradiation and to DNA damaging agents such as alkylating agents (e.g. temozolomide and methyl methanesulfonate) (a) Fujihara et al., Curr. Cancer Drug Targets 2009, 9, 953; b) Gogola et al., Cancer Cell 2018, 33, 1078; c) Houl et al., Nat Commun. 2019, 10, 5654).
Given the therapeutic potential of PARG inhibitors in cancer treatment, there is an increased need for the development of highly potent and selective PARG inhibitors beyond the ones that have already been described (a) James et al., ACS Chem. Biol. 2016, 11 , 3179; b) Waszkowycz et al., J. Med. Chem. 2018, 61 , 10767).
Certain compounds that are useful as PARG inhibitors are further disclosed in documents WO 2016/092326, WO 2016/097749 and WO 2021/055744.
Summary of the invention
It was an objective technical problem of the present invention to provide compounds that are cell- permeable inhibitors of PARG. The technical problem of the present invention is solved by the embodiments described herein and as characterized by the claims.
Accordingly, in a first embodiment, the present invention provides a compound of formula (I):
Figure imgf000004_0001
or a pharmaceutically acceptable salt thereof. It is to be understood that the term “compound of formula (I) preferably encompasses also an enantiomer, diastereoisomer, tautomer, pharmaceutically acceptable solvate, pharmaceutically acceptable crystal form, pharmaceutically acceptable salt or a prodrug thereof.
A further embodiment of the present invention relates to a pharmaceutical composition comprising the compound of formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
In a further embodiment, the present invention relates to the compound of formula (I) of the present invention or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of the present invention, for use in therapy.
The compounds of formula (I) are useful for treating a disease or disorder in which PARG activity is implicated.
The compounds of formula (I) are useful for a method of treating a proliferative disorder. In a preferred embodiment of the present invention, the proliferative disorder is cancer, preferably a human cancer.
Definitions
The following definitions apply throughout the present specification and the claims, unless specifically indicated otherwise.
The term “hydrogen” is herein used to refer to protium, deuterium and/or tritium, preferably to protium. Accordingly, the term “non-hydrogen atom” refers to any atoms that is not hydrogen, i.e. that is not protium, deuterium or tritium.
The term “hydrocarbon group” refers to a group consisting of carbon atoms and hydrogen atoms.
The term “alicyclic” is used in connection with cyclic groups and denotes that the corresponding cyclic group is non-aromatic.
As used herein, the term “alkyl” refers to a monovalent saturated acyclic (i.e., non-cyclic) hydrocarbon group which may be linear or branched. Accordingly, an “alkyl” group does not comprise any carbon-to-carbon double bond or any carbon-to-carbon triple bond. A “C1-5 alkyl” denotes an alkyl group having 1 to 5 carbon atoms. Preferred exemplary alkyl groups are methyl, ethyl, propyl (e.g., n-propyl or isopropyl), or butyl (e.g., n-butyl, isobutyl, sec-butyl, or tert-butyl). Unless defined otherwise, the term “alkyl” preferably refers to C1-4 alkyl, more preferably to methyl or ethyl, and even more preferably to methyl.
As used herein, the term “alkenyl” refers to a monovalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon double bonds while it does not comprise any carbon-to-carbon triple bond. The term “C2-5 alkenyl” denotes an alkenyl group having 2 to 5 carbon atoms. Preferred exemplary alkenyl groups are ethenyl, propenyl (e.g., prop-1 -en-1-yl, prop-1 -en-2-yl, or prop-2-en-1 -yl), butenyl, butadienyl (e.g., buta-1 ,3-dien-1 -yl or buta-1 ,3- dien-2-yl), pentenyl, or pentadienyl (e.g., isoprenyl). Unless defined otherwise, the term “alkenyl” preferably refers to C2-4 alkenyl.
As used herein, the term “alkynyl” refers to a monovalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon triple bonds and optionally one or more (e.g., one or two) carbon-to-carbon double bonds. The term “C2-5 alkynyl” denotes an alkynyl group having 2 to 5 carbon atoms. Preferred exemplary alkynyl groups are ethynyl, propynyl (e.g., propargyl), or butynyl. Unless defined otherwise, the term “alkynyl” preferably refers to C2-4 alkynyl.
As used herein, the term “alkylene” refers to an alkanediyl group, i.e. a divalent saturated acyclic hydrocarbon group which may be linear or branched. A “C1-5 alkylene” denotes an alkylene group having 1 to 5 carbon atoms, and the term “C0-3 alkylene” indicates that a covalent bond (corresponding to the option “Co alkylene”) or a C1-3 alkylene is present. Preferred exemplary alkylene groups are methylene (- CH2-), ethylene (e.g., -CH2-CH2- or -CH(-CH3)-), propylene (e.g., -CH2-CH2-CH2-, -CH(-CH2-CH3)-, -CH2- CH(-CH3)-, or -CH(-CH3)-CH2-), or butylene (e.g., -CH2-CH2-CH2-CH2-). Unless defined otherwise, the term “alkylene” preferably refers to C1-4 alkylene (including, in particular, linear C1-4 alkylene), more preferably to methylene or ethylene, and even more preferably to methylene. As used herein, the term “alkenylene” refers to an alkenediyl group, i.e. a divalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon double bonds while it does not comprise any carbon-to-carbon triple bond. A “C2- 5 alkenylene” denotes an alkenylene group having 2 to 5 carbon atoms. Unless defined otherwise, the term “alkenylene” preferably refers to C2-4 alkenylene (including, in particular, linear C2-4 alkenylene).
As used herein, the term “alkynylene” refers to an alkynediyl group, i.e. a divalent unsaturated acyclic hydrocarbon group which may be linear or branched and comprises one or more (e.g., one or two) carbon-to-carbon triple bonds and optionally one or more (e.g., one or two) carbon-to-carbon double bonds. A “C2-5 alkynylene” denotes an alkynylene group having 2 to 5 carbon atoms. Unless defined otherwise, the term “alkynylene” preferably refers to C2-4 alkynylene (including, in particular, linear C2-4 alkynylene).
As used herein, the term “carbocyclyl” refers to a hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings), wherein said ring group may be saturated, partially unsaturated (i.e., unsaturated but not aromatic) or aromatic. Unless defined otherwise, “carbocyclyl” preferably refers to aryl, cycloalkyl or cycloalkenyl.
As used herein, the term “heterocyclyl” refers to a ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings), wherein said ring group comprises one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group), and further wherein said ring group may be saturated, partially unsaturated (i.e., unsaturated but not aromatic) or aromatic. For example, each heteroatom-containing ring comprised in said ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring. Unless defined otherwise, “heterocyclyl” preferably refers to heteroaryl, heterocycloalkyl or heterocycloalkenyl.
As used herein, the term “aryl” refers to an aromatic hydrocarbon ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic). “Aryl” may, e.g., refer to phenyl, naphthyl, dialinyl (i.e., 1 ,2-dihydronaphthyl), tetralinyl (i.e., 1 ,2,3,4-tetrahydronaphthyl), indanyl, indenyl (e.g., 1 H-indenyl), anthracenyl, phenanthrenyl, 9H- fluorenyl, or azulenyl. Unless defined otherwise, an “aryl” preferably has 6 to 14 ring atoms, more preferably 6 to 10 ring atoms, even more preferably refers to phenyl or naphthyl, and most preferably refers to phenyl.
As used herein, the term “arylene” refers to an aryl group, as defined herein above, but having two points of attachment, i.e. a divalent aromatic hydrocarbon ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic). “Arylene” may, e.g., refer to phenylene (e.g., phen-1 ,2-diyl, phen-1 , 3-diyl, or phen-1 ,4-diyl), naphthylene (e.g., naphthalen-1 ,2-diyl, naphthalen-1 ,3-diyl, naphthalen-1 ,4-diyl, naphthalen-1 ,5-diyl, naphthalen-1 ,6- diyl, naphthalen-1 , 7-diyl, naphthalen-2, 3-diyl, naphthalen-2, 5-diyl, naphthalen-2, 6-diyl, naphthalen-2, 7- diyl, or naphthalen-2, 8-diyl), 1 ,2-dihydronaphthylene, 1 ,2,3,4-tetrahydronaphthylene, indanylene, indenylene, anthracenylene, phenanthrenylene, 9H-fluorenylene, or azulenylene. Unless defined otherwise, an “arylene” preferably has 6 to 14 ring atoms, more preferably 6 to 10 ring atoms, even more preferably refers to phenylene or naphthylene, and most preferably refers to phenylene (particularly phen- 1 ,4-diyl).
As used herein, the term “heteroaryl” refers to an aromatic ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic), wherein said aromatic ring group comprises one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group). For example, each heteroatom-containing ring comprised in said aromatic ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring. “Heteroaryl” may, e.g., refer to thienyl (i.e., thiophenyl), benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl (i.e., furanyl), benzofuranyl, isobenzofuranyl, chromanyl, chromenyl (e.g., 2H-1- benzopyranyl or 4H-1 -benzopyranyl), isochromenyl (e.g., 1 H-2-benzopyranyl), chromonyl, xanthenyl, phenoxathiinyl, pyrrolyl (e.g., 1 H-pyrrolyl), imidazolyl, pyrazolyl, pyridyl (i.e., pyridinyl; e.g., 2-pyridyl, 3- pyridyl, or 4-pyridyl), pyrazinyl, pyrimidinyl, pyridazinyl, indolyl (e.g., 3H-indolyl), isoindolyl, indazolyl, indolizinyl, purinyl, quinolyl, isoquinolyl, phthalazinyl, naphthyridinyl, quinoxalinyl, cinnolinyl, pteridinyl, carbazolyl, p-carbolinyl, phenanthridinyl, acridinyl, perimidinyl, phenanthrolinyl (e.g., [1 ,10]phenanthrolinyl, [1 ,7]phenanthrolinyl, or [4,7]phenanthrolinyl), phenazinyl, thiazolyl, isothiazolyl, phenothiazinyl, oxazolyl, isoxazolyl, oxadiazolyl (e.g., 1 ,2,4-oxadiazolyl, 1 ,2,5-oxadiazolyl (i.e., furazanyl), or 1 ,3,4-oxadiazolyl), thiadiazolyl (e.g., 1 ,2,4-thiadiazolyl, 1 ,2,5-thiadiazolyl, or 1 ,3,4-thiadiazolyl), phenoxazinyl, pyrazolo[1 ,5-a]pyrimidinyl (e.g., pyrazolo[1 ,5-a]pyrimidin-3-yl), 1 ,2-benzoisoxazol-3-yl, benzothiazolyl, benzothiadiazolyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzo[b]thiophenyl (i.e., benzothienyl), triazolyl (e.g., 1 H-1 ,2,3-triazolyl, 2H-1 ,2,3-triazolyl, 1 H-1 ,2,4-triazolyl, or 4H-1 ,2,4-triazolyl), benzotriazolyl, 1 H-tetrazolyl, 2H-tetrazolyl, triazinyl (e.g., 1,2,3-triazinyl, 1 ,2,4-triazinyl, or 1 ,3,5-triazinyl), furo[2,3-c]pyridinyl, dihydrofuropyridinyl (e.g., 2,3-dihydrofuro[2,3-c]pyridinyl or 1 ,3-dihydrofuro[3,4- c]pyridinyl), imidazopyridinyl (e.g., imidazo[1 ,2-a]pyridinyl or imidazo[3,2-a]pyridinyl), quinazolinyl, thienopyridinyl, tetrahydrothienopyridinyl (e.g., 4,5,6,7-tetrahydrothieno[3,2-c]pyridinyl), dibenzofuranyl, 1 ,3-benzodioxolyl, benzodioxanyl (e.g., 1 ,3-benzodioxanyl or 1 ,4-benzodioxanyl), or coumarinyl. Unless defined otherwise, the term “heteroaryl” preferably refers to a 5 to 14 membered (more preferably 5 to 10 membered) monocyclic ring or fused ring system comprising one or more (e.g., one, two, three or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; even more preferably, a “heteroaryl” refers to a 5 or 6 membered monocyclic ring comprising one or more (e.g., one, two or three) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized.
As used herein, the term “heteroarylene” refers to a heteroaryl group, as defined herein above, but having two points of attachment, i.e. a divalent aromatic ring group, including monocyclic aromatic rings as well as bridged ring and/or fused ring systems containing at least one aromatic ring (e.g., ring systems composed of two or three fused rings, wherein at least one of these fused rings is aromatic; or bridged ring systems composed of two or three rings, wherein at least one of these bridged rings is aromatic), wherein said aromatic ring group comprises one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group). For example, each heteroatom-containing ring comprised in said aromatic ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three, or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring. “Heteroarylene” may, e.g., refer to thienylene (i.e., thiophenylene; e.g., thien-2,3-diyl, thien-2,4-diyl, or thien-2,5-diyl), benzo[b]thienylene, naphtho[2,3-b]thienylene, thianthrenylene, furylene (i.e., furanylene; e.g., furan-2,3-diyl, furan-2,4-diyl, or furan-2,5-diyl), benzofuranylene, isobenzofuranylene, chromanylene, chromenylene, isochromenylene, chromonylene, xanthenylene, phenoxathiinylene, pyrrolylene, imidazolylene, pyrazolylene, pyridylene (i.e., pyridinylene), pyrazinylene, pyrimidinylene, pyridazinylene, indolylene, isoindolylene, indazolylene, indolizinylene, purinylene, quinolylene, isoquinolylene, phthalazinylene, naphthyridinylene, quinoxalinylene, cinnolinylene, pteridinylene, carbazolylene, p-carbolinylene, phenanthridinylene, acridinylene, perimidinylene, phenanthrolinylene, phenazinylene, thiazolylene (e.g., thiazol-2,4-diyl, thiazol-2,5-diyl, or thiazol-4,5-diyl), isothiazolylene (e.g., isothiazol-3,4-diyl, isothiazol-3,5-diyl, or isothiazol-4,5-diyl), phenothiazinylene, oxazolylene (e.g., oxazol-2,4-diyl, oxazol-2,5-diyl, or oxazol-4,5-diyl), isoxazolylene (e.g., isoxazol-3,4-diyl, isoxazol-3,5-diyl, or isoxazol-4,5-diyl), oxadiazolylene (e.g., 1 ,2,4-oxadiazol-3,5-diyl, 1 ,2,5-oxadiazol-3,4-diyl, or 1,3,4- oxadiazol-2,5-diyl), thiadiazolylene (e.g., 1 ,2,4-thiadiazol-3,5-diyl, 1 ,2,5-thiadiazol-3,4-diyl, or 1,3,4- thiadiazol-2,5-diyl), phenoxazinylene, pyrazolo[1 ,5-a]pyrimidinylene, 1 ,2-benzoisoxazolylene, benzothiazolylene, benzothiadiazolylene, benzoxazolylene, benzisoxazolylene, benzimidazolylene, benzo[b]thiophenylene (i.e., benzothienylene), triazolylene (e.g., 1 H-1 ,2,3-triazolylene, 2H-1,2,3- triazolylene, 1 H-1 ,2,4-triazolylene, or 4H-1 ,2,4-triazolylene), benzotriazolylene, 1 H-tetrazolylene, 2H-tetrazolylene, triazinylene (e.g., 1 ,2,3-triazinylene, 1,2,4-triazinylene, or 1 ,3,5-triazinylene), furo[2,3- c]pyridinylene, dihydrofuropyridinylene (e.g., 2,3-dihydrofuro[2,3-c]pyridinylene or 1 ,3-dihydrofuro[3,4-c]pyridinylene), imidazopyridinylene (e.g., imidazo[1 ,2-a]pyridinylene or imidazo[3,2-a]pyridinylene), quinazolinylene, thienopyridinylene, tetrahydrothienopyridinylene (e.g., 4, 5, 6, 7-tetrahyd rothieno[3, 2-c]pyridi nylene) , dibenzofuranylene, 1 ,3-benzodioxolylene, benzodioxanylene (e.g., 1 ,3-benzodioxanylene or 1 ,4-benzodioxanylene), or coumarinylene. Unless defined otherwise, the term “heteroarylene” preferably refers to a divalent 5 to 14 membered (more preferably 5 to 10 membered) monocyclic ring or fused ring system comprising one or more (e.g., one, two, three or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; even more preferably, a “heteroarylene” refers to a divalent 5 or 6 membered monocyclic ring comprising one or more (e.g., one, two or three) ring heteroatoms independently selected from 0, S, and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized. A “heteroarylene”, including any of the specific heteroarylene groups described herein, may be attached through two carbon ring atoms, particularly through those two carbon ring atoms that have the greatest distance from one another (in terms of the number of ring atoms separating them by the shortest possible connection) within one single ring or within the entire ring system of the corresponding heteroarylene.
As used herein, the term “cycloalkyl” refers to a saturated hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings). “Cycloalkyl” may, e.g., refer to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, decalinyl (i.e., decahydronaphthyl), or adamantyl. Unless defined otherwise, “cycloalkyl” preferably refers to a C3-11 cycloalkyl, and more preferably refers to a C3-7 cycloalkyl. A particularly preferred “cycloalkyl” is a monocyclic saturated hydrocarbon ring having 3 to 7 ring members (e.g., cyclopropyl or cyclohexyl).
As used herein, the term “cycloalkylene” refers to a cycloalkyl group, as defined herein above, but having two points of attachment, i.e. a divalent saturated hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings). “Cycloalkylene” may, e.g., refer to cyclopropylene (e.g., cyclopropan-1 ,1 -diyl or cyclopropan-1 ,2-diyl), cyclobutylene (e.g., cyclobutan-1, 1 -diyl, cyclobutan-1 ,2-diyl, or cyclobutan-1 ,3-diyl), cyclopentylene (e.g., cyclopentan-1,1 -diyl, cyclopentan-1 , 2-diyl, or cyclopentan-1 , 3-diyl), cyclohexylene (e.g., cyclohexan-1 ,1-diyl, cyclohexan-1 , 2-diyl, cyclohexan-1 , 3-diyl, or cyclohexan-1 ,4-diyl), cycloheptylene, decalinylene (i.e., decahydronaphthylene), or adamantylene. Unless defined otherwise, “cycloalkylene” preferably refers to a C3-11 cycloalkylene, and more preferably refers to a C3-7 cycloalkylene. A particularly preferred “cycloalkylene” is a divalent monocyclic saturated hydrocarbon ring having 3 to 7 ring members (e.g., cyclopropylene or cyclohexylene).
As used herein, the term “heterocycloalkyl” refers to a saturated ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said ring group contains one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group). For example, each heteroatom-containing ring comprised in said saturated ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring. “Heterocycloalkyl” may, e.g., refer to aziridinyl, azetidinyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, piperidinyl, piperazinyl, azepanyl, diazepanyl (e.g., 1 ,4-diazepanyl), oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, morpholinyl (e.g., morpholin-4-yl), thiomorpholinyl (e.g., thiomorpholin-4-yl), oxazepanyl, oxiranyl, oxetanyl, tetrahydrofuranyl, 1 ,3-dioxolanyl, tetrahydropyranyl, 1 ,4-dioxanyl, oxepanyl, thiiranyl, thietanyl, tetrahydrothiophenyl (i.e., thiolanyl), 1 ,3-dithiolanyl, thianyl, 1 ,1 -dioxothianyl, thiepanyl, decahydroquinolinyl, decahydroisoquinolinyl, or 2-oxa-5-aza-bicyclo[2.2.1]hept-5-yl. Unless defined otherwise, “heterocycloalkyl” preferably refers to a 3 to 11 membered saturated ring group, which is a monocyclic ring or a fused ring system (e.g., a fused ring system composed of two fused rings), wherein said ring group contains one or more (e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; more preferably, “heterocycloalkyl” refers to a 5 to 7 membered saturated monocyclic ring group containing one or more (e.g., one, two, or three) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized.
As used herein, the term “A/-heterocycloalkyl” refers to the heterocycloalkyl groups as defined hereinabove wherein said heterocycloalkyl includes at least one nitrogen atom which serves as an attachment point of said heterocycloalkyl.
As used herein, the term “heterocycloalkylene” refers to a heterocycloalkyl group, as defined herein above, but having two points of attachment, i.e. a divalent saturated ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said ring group contains one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, and further wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group). For example, each heteroatom-containing ring comprised in said saturated ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring. “Heterocycloalkylene” may, e.g., refer to aziridinylene, azetidinylene, pyrrolidinylene, imidazolidinylene, pyrazolidinylene, piperidinylene, piperazinylene, azepanylene, diazepanylene (e.g., 1 ,4-diazepanylene), oxazolidinylene, isoxazolidinylene, thiazolidinylene, isothiazolidinylene, morpholinylene, thiomorpholinylene, oxazepanylene, oxiranylene, oxetanylene, tetrahydrofuranylene, 1 ,3-dioxolanylene, tetrahydropyranylene, 1 ,4-dioxanylene, oxepanylene, thiiranylene, thietanylene, tetrahydrothiophenylene (i.e., thiolanylene), 1 ,3-dithiolanylene, thianylene, 1 ,1 -dioxothianylene, thiepanylene, decahydroquinolinylene, decahydroisoquinolinylene, or 2-oxa-5-aza-bicyclo[2.2.1]hept-5-ylene. Unless defined otherwise, “heterocycloalkylene” preferably refers to a divalent 3 to 11 membered saturated ring group, which is a monocyclic ring or a fused ring system (e.g., a fused ring system composed of two fused rings), wherein said ring group contains one or more (e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized; more preferably, “heterocycloalkylene” refers to a divalent 5 to 7 membered saturated monocyclic ring group containing one or more (e.g., one, two, or three) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, and wherein one or more carbon ring atoms are optionally oxidized.
As used herein, the term “cycloalkenyl” refers to an unsaturated alicyclic (non-aromatic) hydrocarbon ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said hydrocarbon ring group comprises one or more (e.g., one or two) carbon-to-carbon double bonds and does not comprise any carbon-to-carbon triple bond. “Cycloalkenyl” may, e.g., refer to cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, or cycloheptadienyl. Unless defined otherwise, “cycloalkenyl” preferably refers to a C3-11 cycloalkenyl, and more preferably refers to a C3-7 cycloalkenyl. A particularly preferred “cycloalkenyl” is a monocyclic unsaturated alicyclic hydrocarbon ring having 3 to 7 ring members and containing one or more (e.g., one or two; preferably one) carbon-to-carbon double bonds.
As used herein, the term “heterocycloalkenyl” refers to an unsaturated alicyclic (non-aromatic) ring group, including monocyclic rings as well as bridged ring, spiro ring and/or fused ring systems (which may be composed, e.g., of two or three rings; such as, e.g., a fused ring system composed of two or three fused rings), wherein said ring group contains one or more (such as, e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, and the remaining ring atoms are carbon atoms, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) may optionally be oxidized, wherein one or more carbon ring atoms may optionally be oxidized (i.e., to form an oxo group), and further wherein said ring group comprises at least one double bond between adjacent ring atoms and does not comprise any triple bond between adjacent ring atoms. For example, each heteroatom-containing ring comprised in said unsaturated alicyclic ring group may contain one or two 0 atoms and/or one or two S atoms (which may optionally be oxidized) and/or one, two, three or four N atoms (which may optionally be oxidized), provided that the total number of heteroatoms in the corresponding heteroatom-containing ring is 1 to 4 and that there is at least one carbon ring atom (which may optionally be oxidized) in the corresponding heteroatom-containing ring. “Heterocycloalkenyl” may, e.g., refer to imidazolinyl (e.g., 2-imidazolinyl (i.e., 4,5-dihydro-1 H-imidazolyl), 3-imidazolinyl, or 4-imidazolinyl), tetrahydropyridinyl (e.g., 1 ,2,3,6-tetrahydropyridinyl), dihydropyridinyl (e.g., 1,2- dihydropyridinyl or 2,3-dihydropyridinyl), pyranyl (e.g., 2H-pyranyl or 4H-pyranyl), thiopyranyl (e.g., 2H-thiopyranyl or 4H-thiopyranyl), dihydropyranyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrazinyl, dihydroisoindolyl, octahydroquinolinyl (e.g., 1 ,2,3,4,4a,5,6,7-octahydroquinolinyl), or octahydroisoquinolinyl (e.g., 1 ,2,3,4,5,6,7,8-octahydroisoquinolinyl). Unless defined otherwise, “heterocycloalkenyl” preferably refers to a 3 to 11 membered unsaturated alicyclic ring group, which is a monocyclic ring or a fused ring system (e.g., a fused ring system composed of two fused rings), wherein said ring group contains one or more (e.g., one, two, three, or four) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, wherein one or more carbon ring atoms are optionally oxidized, and wherein said ring group comprises at least one double bond between adjacent ring atoms and does not comprise any triple bond between adjacent ring atoms; more preferably, “heterocycloalkenyl” refers to a 5 to 7 membered monocyclic unsaturated non-aromatic ring group containing one or more (e.g., one, two, or three) ring heteroatoms independently selected from 0, S and N, wherein one or more S ring atoms (if present) and/or one or more N ring atoms (if present) are optionally oxidized, wherein one or more carbon ring atoms are optionally oxidized, and wherein said ring group comprises at least one double bond between adjacent ring atoms and does not comprise any triple bond between adjacent ring atoms.
As used herein, the term “halogen” refers to fluoro (-F), chloro (-CI), bromo (-Br), or iodo (-I).
As used herein, the term “haloalky I” refers to an alkyl group substituted with one or more (preferably 1 to 6, more preferably 1 to 3) halogen atoms which are selected independently from fluoro, chloro, bromo and iodo, and are preferably all fluoro atoms. It will be understood that the maximum number of halogen atoms is limited by the number of available attachment sites and, thus, depends on the number of carbon atoms comprised in the alkyl moiety of the haloalkyl group. “Haloalkyl” may, e.g., refer to -CF3, -CHF2, -CH2F, -CF2-CH3, -CH2-CF3, -CH2-CHF2, -CH2-CF2-CH3, -CH2-CF2-CF3, or -CH(CF3)2. A particularly preferred “haloalkyl” group is -CF3.
The terms “bond” and “covalent bond” are used herein synonymously, unless explicitly indicated otherwise or contradicted by context.
As used herein, the terms “optional”, “optionally” and “may” denote that the indicated feature may be present but can also be absent. Whenever the term “optional”, “optionally” or “may” is used, the present invention specifically relates to both possibilities, i.e., that the corresponding feature is present or, alternatively, that the corresponding feature is absent. For example, the expression “X is optionally substituted with Y” (or “X may be substituted with Y”) means that X is either substituted with Y or is unsubstituted. Likewise, if a component of a composition is indicated to be “optional”, the invention specifically relates to both possibilities, i.e., that the corresponding component is present (contained in the composition) or that the corresponding component is absent from the composition.
Various groups are referred to as being “optionally substituted” in this specification. Generally, these groups may carry one or more substituents, such as, e.g., one, two, three or four substituents. It will be understood that the maximum number of substituents is limited by the number of attachment sites available on the substituted moiety. Unless defined otherwise, the “optionally substituted” groups referred to in this specification carry preferably not more than two substituents and may, in particular, carry only one substituent. Moreover, unless defined otherwise, it is preferred that the optional substituents are absent, i.e. that the corresponding groups are unsubstituted.
A skilled person will appreciate that the substituent groups comprised in the compounds of the present invention may be attached to the remainder of the respective compound via a number of different positions of the corresponding specific substituent group. Unless defined otherwise, the preferred attachment positions for the various specific substituent groups are as illustrated in the examples.
As used herein, unless explicitly indicated otherwise or contradicted by context, the terms “a”, “an” and “the” are used interchangeably with “one or more” and “at least one”. Thus, for example, a composition comprising “a” compound of formula (I) can be interpreted as referring to a composition comprising “one or more” compounds of formula (I).
It is to be understood that wherever numerical ranges are provided/disclosed herein, all values and subranges encompassed by the respective numerical range are meant to be encompassed within the scope of the invention. Accordingly, the present invention specifically and individually relates to each value that falls within a numerical range disclosed herein, as well as each subrange encompassed by a numerical range disclosed herein.
As used herein, the term “about” preferably refers to ±10% of the indicated numerical value, more preferably to ±5% of the indicated numerical value, and in particular to the exact numerical value indicated. If the term “about” is used in connection with the endpoints of a range, it preferably refers to the range from the lower endpoint -10% of its indicated numerical value to the upper endpoint +10% of its indicated numerical value, more preferably to the range from of the lower endpoint -5% to the upper endpoint +5%, and even more preferably to the range defined by the exact numerical values of the lower endpoint and the upper endpoint.
As used herein, the term “comprising” (or “comprise”, “comprises”, “contain”, “contains”, or “containing”), unless explicitly indicated otherwise or contradicted by context, has the meaning of “containing, inter alia”, i.e., “containing, among further optional elements, ...”. In addition thereto, this term also includes the narrower meanings of “consisting essentially of’ and “consisting of’. For example, the term “A comprising B and C” has the meaning of “A containing, inter alia, B and C”, wherein A may contain further optional elements (e.g., “A containing B, C and D” would also be encompassed), but this term also includes the meaning of “A consisting essentially of B and C” and the meaning of “A consisting of B and C” (i.e., no other components than B and C are comprised in A).
Detailed description of the invention
The invention is described in detail in the following. It is to be understood that the present invention specifically relates to each and every combination of features and embodiments described herein, including any combination of general and/or preferred features/embodiments.
In a first embodiment, the present invention relates to a compound of formula (I):
Figure imgf000015_0001
or a pharmaceutically acceptable salt thereof.
In formula (I), Roov is selected from C2 alkenyl, C2 alkynyl, -CH2CI, -CH2CN, and
Figure imgf000015_0002
wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(Ci-4 alkyl), -CO-(Ci-4 alkyl), -CONH-(CI-4 alkyl), -OOC-(Ci-4 alkyl), -NHCO-(CI-4 alkyl), -(C1-4 alkylene)N(Ci-4 alkyl)(Ci-4 alkyl), -(C1-4 alkylene)-(A/-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, said alkynyl is optionally substituted with an optional substituent selected from C1-4 alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, and the -CH2- group in said -CH2Cl and the -CH2- group in said -CH2CN are each optionally substituted with one or more optional substituents selected from -Hal, C1-4 alkyl and -CF3. Suitable cycloalkyl group is for example a cyclopropyl group. Suitable aryl group is for example a phenyl group. Preferably, Rcov is selected from C2 alkenyl, C2 alkynyl, and -CH2Cl, wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -OOC-(C1-4 alkyl), -NHCO-(C1-4 alkyl), - (C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, preferably selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -(C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, more preferably selected from C1-4 alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, said alkynyl is optionally substituted with an optional substituent selected from C1-4 alkyl, cycloalkyl, heterocycloalkyl, aryl phenyl and heteroaryl, preferably selected from C1-4 alkyl, cycloalkyl, and aryl and the -CH2- group in said -CH2Cl is optionally substituted with one or more optional substituents selected from -Hal, C1-4 alkyl and -CF3. More preferably, Rcov is selected from C2 alkenyl, and -CH2Cl, wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -OOC-(C1-4 alkyl), -NHCO-(C1-4 alkyl), - (C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, preferably selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -(C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, more preferably selected from C1-4 alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, and the -CH2- group in said -CH2Cl is optionally substituted with one or more optional substituents selected from -Hal, C1-4 alkyl and -CF3. Even more preferably, Rcov is C2 alkenyl, wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -OOC-(C1-4 alkyl), -NHCO-(C1-4 alkyl), -(C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N- heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, preferably selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -(C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and - CF3, more preferably selected from C1-4 alkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3. Even more preferably, Rcov is C2 alkenyl. In formula (I), -Wcov- is selected from -CO-, -SO- and -SO2-. Preferably, -Wcov- is selected from - CO-, and -SO2-. More preferably, -Wcov- is -CO-. RN is selected from hydrogen and C1-4 alkyl. Suitable C1-4 alkyl is for example methyl or ethyl. Preferably, RN is hydrogen. In formula (I), R1 is hydrogen, chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl) (preferably R1 is chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl)); and R2 and R3 are independently each C1-2 alkyl or C1-2 haloalkyl, or R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F; or -CR1R2R3 is bicyclo[1,1,1]pent-1-yl. Preferably, R1 is hydrogen, chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl) (preferably R1 is chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl)); and R2 and R3 are independently each C1-2 alkyl or C1-2 haloalkyl, or R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F. More preferably, R1 is hydrogen, chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1- 2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl) (preferably R1 is chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl)); and R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F. R1 is preferably -CN, methyl, fluoromethyl, difluoromethyl or trifluoromethyl, more preferably R1 is methyl or fluoromethyl. Preferably, R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F. W is selected from -NHS(O)y-, -S(O)yNH-, -NHS(O)(NH)-, -NHS(O)(N-C1-2 alkyl)-, -S(O)(NH)-NH- , -S(O)(N-C1-2 alkyl)-NH-, wherein y is 1 or 2. Preferably, y is 2. Thus, in a preferred embodiment, W is selected from -NHS(O)2-, -S(O)2NH-, -NHS(O)(NH)-, and -S(O)(NH)-NH-. More preferably, W is selected from -NHS(O)2-, and -S(O)2NH-, even more preferably W is -NHS(O)2-. Preferably as understood herein, the left side of W as defined herein is attached to the carbon atom that carries R1, R2 and R3, and the right side of W as defined herein is attached to the ring system shown in formula (I). X1 and X3 are independently selected from the group consisting of N, CH, C(C1-2 alkyl), C-Cl and CF, preferably independently selected from the group consisting of N, CH and CF. Preferably, X1 is CF or CH and X3 is CH, more preferably X1 and X3 are each CH. X2 is N or C-YC2-RC2, wherein YC2 is selected from a covalent bond, C1-8 alkylene, C2-8 alkenylene, C2-8 alkynylene, cycloalkylene and heterocycloalkylene wherein said alkylene, said alkenylene and said alkynylene are each optionally substituted with one or more groups independently selected from RS1, and further wherein one or more -CH2- units comprised in said alkylene, said alkenylene or said alkynylene are each optionally replaced by a group independently selected from -O-, -NH-, -N(C1-5 alkyl)-, -CO-, -S-, -SO-, and -SO2-, and wherein said cycloalkylene and heterocycloalkylene are each optionally substituted with one or more groups independently selected RS2; and wherein RC2 is selected from hydrogen, halo, -OH, -NH2, -SH, -CN, C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; wherein said alkyl, alkenyl, and alkynyl in X2 are each optionally substituted with one or more groups independently selected from RS1, and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in X2 are each optionally substituted with one or more groups independently selected from RS2. Preferably, YC2 is selected from a covalent bond, -(C1-3 alkylene)-, -CO-(C1-3 alkylene)-, (C1-3 alkylene)-CO-, -CONH-(C1-3 alkylene)-, -(C1-3 alkylene)-CONH-, -NHCO-(C1-3 alkylene)-, -(C1-3 alkylene)- NHCO-, -NH-(C1-3 alkylene)-, -(C1-3 alkylene)-NH-, -N(C1-5 alkyl)-, -O-(C1-3 alkylene)-, -(C1-3 alkylene)-O- , -SO2-(C1-3 alkylene)-, -(C1-3 alkylene)-SO2-, -CONH-, -NHCO-, -NH-, -O-, -CO- and -SO2-, wherein said alkylene, said alkenylene and said alkynylene are each optionally substituted with one or more groups independently selected from RS1. C1-3 alkylene is herein preferably a -CH2- group. Preferably, RC2 is selected from hydrogen, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from RS2. More preferably, RC2 is selected from cycloalkyl, heterocycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in X2 are each optionally substituted with one or more groups independently selected from RS2. Even more preferably, RC2 is selected from heterocycloalkyl, aryl, and heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in X2 are each optionally substituted with one or more groups independently selected from RS2. Thus preferably, if X2 is C-YC2-RC2, -YC2-RC2 is is selected from -O-C1-12 alkyl, -NH-C1-12 alkyl, - N(C1-5 alkyl)-C1-12 alkyl, -O-C2-12 alkenyl, -NH-C2-12 alkenyl, -N(C1-5 alkyl)-C2-12 alkenyl, -O-C2-12 alkynyl, - NH-C2-12 alkynyl, -N(C1-5 alkyl)-C2-12 alkynyl, -(C0-3 alkylene)-cycloalkyl, -CO-(C0-3 alkylene)-cycloalkyl, - (C0-3 alkylene)-CO-cycloalkyl, -CONH-(C0-3 alkylene)-cycloalkyl, (C0-3 alkylene)-CONH-cycloalkyl, - NHCO-(C0-3 alkylene)-cycloalkyl, -(C0-3 alkylene)-NHCO-cycloalkyl, -NH-(C0-3 alkylene)-cycloalkyl, -(C0-3 alkylene)-NH-cycloalkyl, -O-(C0-3 alkylene)-cycloalkyl, -(C0-3 alkylene)-O-cycloalkyl, -SO2-(C0-3 alkylene)- cycloalkyl, -(C0-3 alkylene)-SO2-cycloalkyl, -CONH-cycloalkyl, -NHCO-cycloalkyl, -NH-cycloalkyl, -O- cycloalkyl, -CO-cycloalkyl, -SO2-cycloalkyl, -(C0-3 alkylene)-heterocycloalkyl, -CO-(C0-3 alkylene)- heterocycloalkyl, -(C0-3 alkylene)-CO-heterocycloalkyl, -CONH-(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)-CONH-heterocycloalkyl, -NHCO-(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)-NHCO- heterocycloalkyl, -NH-(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)-NH-heterocycloalkyl, -O-(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)-O-cycloalkyl, -SO2-(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)-SO2-heterocycloalkyl, -CONH-heterocycloalkyl, -NHCO-heterocycloalkyl, -NH-heterocycloalkyl, -O-heterocycloalkyl, -CO-heterocycloalkyl, -SO2-heterocycloalkyl, -(C0-3 alkylene)-aryl, -CO-(C0-3 alkylene)-aryl, -(C0-3 alkylene)-CO-aryl, -CONH-(C0-3 alkylene)-aryl, -(C0-3 alkylene)-CONH-aryl, -NHCO- (C0-3 alkylene)-aryl, -(C0-3 alkylene)-NHCO-aryl, -NH-(C0-3 alkylene)-aryl, -(C0-3 alkylene)-NH-aryl, -O-(C0- 3 alkylene)-aryl, -(C0-3 alkylene)-O-aryl, -SO2-(C0-3 alkylene)-aryl, -(C0-3 alkylene)-SO2-aryl, -CONH-aryl, - NHCO-aryl, -NH-aryl, -O-aryl, -CO-aryl, -SO2-aryl, -(C0-3 alkylene)-heteroaryl, -CO-(C0-3 alkylene)- heteroaryl, -(C0-3 alkylene)-CO-heteroaryl, -CONH-(C0-3 alkylene)-heteroaryl, -(C0-3 alkylene)-CONH- heteroaryl, -NHCO-(C0-3 alkylene)-heteroaryl, -(C0-3 alkylene)-NHCO-heteroaryl, -NH-(C0-3 alkylene)- heteroaryl, -(C0-3 alkylene)-NH-heteroaryl, -O-(C0-3 alkylene)-heteroaryl, -(C0-3 alkylene)-O- heteroaryl, -SO2-(C0-3 alkylene)-heteroaryl, -(C0-3 alkylene)-SO2-heteroaryl, -CONH-heteroaryl, -NHCO- heteroaryl, -NH-heteroaryl, -O-heteroaryl, -CO-heteroaryl and -SO2-heteroaryl, wherein said alkylene is optionally substituted with one or more groups independently selected from RS1, and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from RS2. More preferably, -YC2-RC2 is selected from -(C0-3 alkylene)-heterocycloalkyl, -CO-(C0-3 alkylene)heterocycloalkyl, -(C0-3 alkylene)-CO-heterocycloalkyl, -CONH-(C0-3 alkylene)heterocycloalkyl, - (C0-3 alkylene)-CONH-heterocycloalkyl, -NHCO-(C0-3 alkylene)heterocycloalkyl, -(C0-3 alkylene)-NHCO- heterocycloalkyl, -NH-(C0-3 alkylene)heterocycloalkyl, -(C0-3 alkylene)-NH-heterocycloalkyl, -O-(C0-3 alkylene) heterocycloalkyl, (C0-3 alkylene)-O-cycloalkyl, -SO2-(C0-3 alkylene)heterocycloalkyl, -(C0-3 alkylene)-SO2-heterocycloalkyl, -CONH-heterocycloalkyl, -NHCO-heterocycloalkyl, -NH-heterocycloalkyl, -O-heterocycloalkyl, -CO-heterocycloalkyl, -SO2-heterocycloalkyl, -(C0-3 alkylene)aryl, -CO-(C0-3 alkylene)aryl, -(C0-3 alkylene)-CO-aryl, -CONH-(C0-3 alkylene)aryl, -(C0-3 alkylene)-CONH-aryl, -NHCO- (C0-3 alkylene)aryl, -(C0-3 alkylene)-NHCO-aryl, -NH-(C0-3 alkylene)aryl, -(C0-3 alkylene)-NH-aryl, -O-(C0-3 alkylene)aryl, -(C0-3 alkylene)-O-aryl, -SO2-(C0-3 alkylene)aryl, -(C0-3 alkylene)-SO2-aryl, -CONH-aryl, - NHCO-aryl, -NH-aryl, -O-aryl, -CO-aryl, -SO2-aryl, -(C0-3 alkylene)heteroaryl, -CO-(C0-3 alkylene)heteroaryl, -(C0-3 alkylene)-CO-heteroaryl, -CONH-(C0-3 alkylene)heteroaryl, -(C0-3 alkylene)- CONH-heteroaryl, -NHCO-(C0-3 alkylene)heteroaryl, -(C0-3 alkylene)-NHCO-heteroaryl, -NH-(C0-3 alkylene)heteroaryl, -(C0-3 alkylene)-NH-heteroaryl, -O-(C0-3 alkylene)heteroaryl, -(C0-3 alkylene)-O- heteroaryl, -SO2-(C0-3 alkylene)heteroaryl, -(C0-3 alkylene)-SO2-heteroaryl, -CONH-heteroaryl, -NHCO- heteroaryl, -NH-heteroaryl, -O-heteroaryl, -CO-heteroaryl and -SO2-heteroaryl, wherein said alkylene is optionally substituted with one or more groups independently selected from RS1, and wherein said heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from RS2. Even more preferably, -YC2-RC2 is selected from -(C0-3 alkylene)-heterocycloalkyl, -CONH- heterocycloalkyl, -NHCO-heterocycloalkyl, -NH-heterocycloalkyl, -O-heterocycloalkyl, -CO- heterocycloalkyl, -SO2-heterocycloalkyl, -(C0-3 alkylene)aryl, -CONH-aryl, -NHCO-aryl, -NH-aryl, -O-aryl, -CO-aryl, -SO2-aryl, -(C0-3 alkylene)heteroaryl, -CONH-heteroaryl, -NHCO-heteroaryl, -NH-heteroaryl, -O- heteroaryl, -CO-heteroaryl and -SO2-heteroaryl, wherein said alkylene is optionally substituted with one or more groups independently selected from RS1, and wherein said heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from RS2. Even more preferably, -YC2-RC2 is selected from -(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)aryl, and -(C0-3 alkylene)heteroaryl, wherein said heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from RS2.. Even more preferably, -YC2-RC2 is selected from heterocycloalkyl, aryl, and heteroaryl wherein said heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from RS2. Even more preferably -YC2-RC2 is selected from heterocycloalkyl and heteroaryl, wherein said heterocycloalkyl, and heteroaryl are each optionally substituted with one or more groups independently selected from RS2. Even more preferably, -YC2-RC2 is heterocycloalkyl wherein said heterocycloalkyl is optionally substituted with one or more groups independently selected from RS2. In formula (I), the moiety represented with a partial formula is a moiety selected from
Figure imgf000021_0001
Figure imgf000021_0002
wherein: R7 is hydrogen, -CN, -Hal, or a moiety of the formula:
Figure imgf000021_0003
wherein: L71 is a bond or C1-5 alkylene optionally substituted with halo or oxo; L72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH-, -CON(C1- 6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, -NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, -SO2N(C1-6 alkyl)-, -NHSO2-, or -N(C1-6 alkyl)SO2-, and Q7 is hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)- aryl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkyl, alkenyl, alkynyl, and alkylene in R7 are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R7 are each optionally substituted with one or more optional substituents selected from RS2; R8 is hydrogen, -CN, -Hal, or a moiety of the formula:
Figure imgf000021_0004
wherein: L81 is a bond, C1-5 alkylene optionally substituted with halo or oxo; L82 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH-, -CON(C1- 6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, -NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, -SO2N(C1-6 alkyl)-, -NHSO2-, or -N(C1-6 alkyl)SO2-; and Q8 is hydrogen, -CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, heterocycloalkyl or heteroaryl; wherein said alkyl, alkenyl, alkynyl, and alkylene in R8 are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R8 are each optionally substituted with one or more optional substituents selected from RS2. Preferably, R7 is a moiety of the formula:
Figure imgf000022_0001
wherein: L71 is a bond or C1-5 alkylene optionally substituted with halo or oxo; L72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH-, -CON(C1- 6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, -NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, -SO2N(C1-6 alkyl)-, -NHSO2-, or -N(C1-6 alkyl)SO2-, and Q7 is hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)- aryl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkyl, alkenyl, alkynyl, and alkylene in R7 are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R7 are each optionally substituted with one or more optional substituents selected from RS2. Preferably, L71 is a bond. Preferably, L72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH- , -CON(C1-6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, -NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, or -NHSO2-, wherein said alkyl in is optionally substituted with one or more optional substituents selected from RS1. More preferably, L72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -CO-, -COO-, -OCO-, -CONH-, - SO2NH- or -NHSO2-. Even more preferably, L72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, or -CO-. Even more preferably, L72 is a bond, -O-, or -S-. Even more preferably, L72 is a bond. It is noted that in the case of two bivalent chemical groups attached to each other, e.g. -L71-L72-, if each of them is defined to be a chemical bond, it is to be understood that the whole moiety comprising two bivalent chemical groups attached to each other is a bond. In other words, for example, is -L71- is a bond and -L72- is a bond, then -L71-L72- is a bond. Preferably, Q7 is C2-6 alkynyl, -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)- heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkynyl and alkylene are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. More preferably, Q7 is -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)- heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkylene is optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. Even more preferably, Q7 is -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkylene is optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. It is to be understood that if L71 is a bond and L72 is a bond, then R7 is preferably Q7. Accordingly, R7 is preferably Q7 is hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, -(C0-2 alkylene)- cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkyl, alkenyl, alkynyl, and alkylene in R7 are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R7 are each optionally substituted with one or more optional substituents selected from RS2. More preferably, R7 is C2-6 alkynyl, -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkynyl and alkylene are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. Even more preferably, R7 is -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)- heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkylene is optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. Even more preferably, R7 is -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkylene is optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. Preferably R8 is a moiety of the formula:
Figure imgf000024_0001
wherein: L81 is a bond, C1-5 alkylene optionally substituted with halo or oxo; L82 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH-, -CON(C1- 6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, -NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, -SO2N(C1-6 alkyl)-, -NHSO2-, or -N(C1-6 alkyl)SO2-; and Q8 is hydrogen, -CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, heterocycloalkyl or heteroaryl; wherein said alkyl, alkenyl, alkynyl, and alkylene in R8 are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R8 are each optionally substituted with one or more optional substituents selected from RS2. Preferably, L81 is -(C0-2 alkylene)-, wherein said alkylene is optionally substituted with one or more optional substituents selected from RS1. Preferably, L82 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -CO-, -COO-, -OCO-, -CONH-, -SO2NH-, or -NHSO2-. More preferably, L82 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, or -CO-. Even more preferably, L82 is a bond, -O-, or -S-. Even more preferably, L82 is a bond. Preferably, Q8 is C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, heterocycloalkyl or heteroaryl; wherein said alkyl, alkenyl, and alkynyl are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. More preferably, Q8 is C2-6 alkynyl, cycloalkyl, aryl, heterocycloalkyl or heteroaryl; wherein said alkynyl is optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. Even more preferably, Q8 is cycloalkyl, aryl, heterocycloalkyl or heteroaryl; wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. Even more preferably, Q8 is cycloalkyl, heterocycloalkyl or heteroaryl; wherein said cycloalkyl, heterocycloalkyl, and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. Thus accordingly, R8 is preferably selected from -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl, wherein said alkylene is optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R8 are each optionally substituted with one or more optional substituents selected from RS2. More preferably, R8 is selected from -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl, wherein said alkylene is optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R8 are each optionally substituted with one or more optional substituents selected from RS2. Particularly preferred -(C0-2 alkylene)- in R8 is methylene. Thus, preferably R8 is selected from - CH2-cycloalkyl, -CH2-aryl, -CH2-heterocycloalkyl or -CH2-heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R8 are each optionally substituted with one or more optional substituents selected from RS2. More preferably R8 is selected from -CH2-cycloalkyl, -CH2-heterocycloalkyl or -CH2-heteroaryl, wherein said cycloalkyl, heterocycloalkyl, and heteroaryl in R8 are each optionally substituted with one or more optional substituents selected from RS2. In other words, preferably in R8 L81 is methylene, -L82 is a covalent bond, and Q8 is cycloalkyl, aryl, heterocycloalkyl or heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R8 are each optionally substituted with one or more optional substituents selected from RS2. More preferably, in R8 L81 is methylene, -L82 is a covalent bond, and Q8 is cycloalkyl, heterocycloalkyl or heteroaryl, wherein said cycloalkyl, heterocycloalkyl, and heteroaryl in R8 are each optionally substituted with one or more optional substituents selected from RS2. Particularly preferred R8 is
Figure imgf000026_0001
Preferably, the moiety represented with a partial formula
Figure imgf000026_0002
Therein, R8 is defined as defined hereinabove. In formula (I), RS1 is selected from halogen, -CN, -OH, -O(C1-5 alkyl), -O(C1-5 haloalkyl), C1-5 haloalkyl, -SH, -S(C1-5 alkyl), -SO2(C1-5 alkyl), -S(O)(NH)(C1-5 alkyl), -S(O)(N-C1-3 alkyl)(C1-5 alkyl), -N=S(O)(C1-5 alkyl)(C1-5 alkyl), -S(C1-5 haloalkyl), -P(O)(C1-5 alkyl)(C1-5 alkyl), -P(O)(O-C1-5 alkyl) (O-C1-5 alkyl), -P(O)(O-C1-5 alkyl)(C1-5 alkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C1-5 alkyl), -N(C1-5 alkyl)-CO-(C1-5 alkyl), -NHCONH2, -NHCONH-(C1-5 alkyl), -NHCON(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 alkyl)CONH2, -N(C1-5 alkyl)CONH-(C1-5 alkyl), and -N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl). Preferably, RS1 is selected from halogen, -CN, -OH, -O(C1-5 alkyl), -O(C1-5 haloalkyl), C1-5 haloalkyl, -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1- 5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C1-5 alkyl), -N(C1-5 alkyl)-CO-(C1-5 alkyl), -NHCONH2, -NHCONH-(C1-5 alkyl), -NHCON(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 alkyl)CONH2, -N(C1-5 alkyl)CONH-(C1-5 alkyl), and -N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl). More preferably, RS1 is selected from halogen, -CN, -OH, -O(C1-5 alkyl), -O(C1-5 haloalkyl), C1-5 haloalkyl, -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1- 5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C1-5 alkyl), -N(C1-5 alkyl)-CO-(C1-5 alkyl), -NHCONH2, -NHCONH-(C1-5 alkyl), -NHCON(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 alkyl)CONH2, -N(C1-5 alkyl)CONH-(C1-5 alkyl), and -N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl). Even more preferably, RS1 is selected from halogen, -CN, -OH, -O(C1-5 alkyl), -O(C1-5 haloalkyl), C1-5 haloalkyl, -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), and -(N-heterocycloalkyl). Even more preferably, RS1 is selected from halogen, -CN, -OH, -SH, and -NH2. In formula (I), RS2 is selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), - O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -SO2(C1-5 alkyl), -S(O)(NH)(C1-5 alkyl), -S(O)(N-C1-3 alkyl)(C1-5 alkyl), -N=S(O)(C1-5 alkyl)(C1-5 alkyl), -S(C1-5 haloalkyl), -P(O)(C1-5 alkyl)(C1-5 alkyl), -P(O)(O-C1-5 alkyl)(O- C1-5 alkyl), -P(O)(O-C1-5 alkyl)(C1-5 alkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C1-5 alkyl), -N(C1-5 alkyl)-CO-(C1-5 alkyl), -NHCONH2, -NHCONH-(C1-5 alkyl), -NHCON(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 alkyl)CONH2, -N(C1-5 alkyl)CONH-(C1-5 alkyl), and -N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-CN, -(C1-5 alkylene)-OH, -(C1-5 alkylene)-O(C1-5 alkyl), -(C1-5 alkylene)-O(C1-5 haloalkyl), -(C1-5 alkylene)-SH, -(C1-5 alkylene)-S(C1-5 alkyl), -(C1-5 alkylene)-SO2(C1-5 alkyl), -(C1-5 alkylene)-S(O)(NH)(C1-5 alkyl), -(C1-5 alkylene)-S(O)(N-C1-3 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-N=S(O)(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)- S(C1-5 haloalkyl), -(C1-5 alkylene)-P(O)(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-P(O)(O-C1-5 alkyl)(O-C1-5 alkyl), -(C1-5 alkylene)-P(O)(O-C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-NH2, -(C1-5 alkylene)-NH(C1-5 alkyl), - (C1-5 alkylene)-NH(C1-5 haloalkyl), -(C1-5 alkylene)-N(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)(C1- 5 haloalkyl), -(C1-5 alkylene)-(N-heterocycloalkyl), -(C1-5 alkylene)-N(C1-5 haloalkyl)(C1-5 alkyl), -(C1-5 alkylene)-CO(C1-5 alkyl), -(C1-5 alkylene)-CONH2, -(C1-5 alkylene)-CONH(C1-5 alkyl), -(C1-5 alkylene)-CON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-CO-(N-heterocycloalkyl), -(C1-5 alkylene)-NHCO-(C1- 5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)-CO-(C1-5 alkyl), -(C1-5 alkylene)-NHCONH2, -(C1-5 alkylene)-NHCONH-(C1-5 alkyl), -(C1-5 alkylene)-NHCON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)CONH2, -(C1-5 alkylene)-N(C1-5 alkyl)CONH-(C1-5 alkyl), and -(C1-5 alkylene)-N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl). Preferably, RS2 is selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1- 5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1- 5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C1-5 alkyl), -N(C1-5 alkyl)-CO-(C1-5 alkyl), -NHCONH2, -NHCONH-(C1-5 alkyl), -NHCON(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 alkyl)CONH2, -N(C1-5 alkyl)CONH-(C1-5 alkyl), -N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-CN, -(C1-5 alkylene)-OH, -(C1-5 alkylene)-O(C1-5 alkyl), -(C1-5 alkylene)-O(C1-5 haloalkyl), -(C1-5 alkylene)-SH, -(C1-5 alkylene)-S(C1-5 alkyl), -(C1-5 alkylene)-S(C1-5 haloalkyl), -(C1-5 alkylene)-NH2, -(C1-5 alkylene)-NH(C1-5 alkyl), -(C1-5 alkylene)-NH(C1-5 haloalkyl), -(C1-5 alkylene)-N(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)(C1-5 haloalkyl), -(C1-5 alkylene)-(N-heterocycloalkyl), -(C1-5 alkylene)-N(C1-5 haloalkyl)(C1-5 alkyl), -(C1-5 alkylene)-CO(C1-5 alkyl), -(C1-5 alkylene)-CONH2, -(C1-5 alkylene)-CONH(C1-5 alkyl), -(C1-5 alkylene)-CON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-CO-(N-heterocycloalkyl), -(C1-5 alkylene)-NHCO-(C1- 5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)-CO-(C1-5 alkyl), -(C1-5 alkylene)-NHCONH2, -(C1-5 alkylene)-NHCONH-(C1-5 alkyl), -(C1-5 alkylene)-NHCON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)CONH2, -(C1-5 alkylene)-N(C1-5 alkyl)CONH-(C1-5 alkyl), and -(C1-5 alkylene)-N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl). More preferably, RS2 is selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1- 5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO-(C1-5 alkyl), -N(C1-5 alkyl)-CO-(C1-5 alkyl), -NHCONH2, -NHCONH-(C1-5 alkyl), -NHCON(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 alkyl)CONH2, -N(C1-5 alkyl)CONH-(C1-5 alkyl), -N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-CN, -(C1-5 alkylene)-OH, -(C1-5 alkylene)-O(C1-5 alkyl), -(C1-5 alkylene)-O(C1-5 haloalkyl), -(C1-5 alkylene)-SH, -(C1-5 alkylene)-S(C1-5 alkyl), -(C1-5 alkylene)-S(C1-5 haloalkyl), -(C1-5 alkylene)-NH2, -(C1-5 alkylene)-NH(C1-5 alkyl), -(C1-5 alkylene)-NH(C1-5 haloalkyl), -(C1-5 alkylene)-N(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)(C1-5 haloalkyl), -(C1-5 alkylene)-(N-heterocycloalkyl), -(C1-5 alkylene)-N(C1-5 haloalkyl)(C1-5 alkyl), -(C1-5 alkylene)-CONH2, -(C1-5 alkylene)-CONH(C1-5 alkyl), -(C1-5 alkylene)-CON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-CO-(N-heterocycloalkyl), -(C1-5 alkylene)-NHCO-(C1-5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)-CO- (C1-5 alkyl), -(C1-5 alkylene)-NHCONH2, -(C1-5 alkylene)-NHCONH-(C1-5 alkyl), -(C1-5 alkylene)-NHCON(C1- 5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)CONH2, -(C1-5 alkylene)-N(C1-5 alkyl)CONH-(C1-5 alkyl), and -(C1-5 alkylene)-N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl). Even more preferably, RS2 is selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -(C1-5 alkylene)- CN, -(C1-5 alkylene)-OH, -(C1-5 alkylene)-O(C1-5 alkyl), -(C1-5 alkylene)-O(C1-5 haloalkyl), -(C1-5 alkylene)-SH, -(C1-5 alkylene)-S(C1-5 alkyl), -(C1-5 alkylene)-S(C1-5 haloalkyl), -(C1-5 alkylene)-NH2, -(C1-5 alkylene)-NH(C1-5 alkyl), -(C1-5 alkylene)-NH(C1-5 haloalkyl), -(C1-5 alkylene)-N(C1-5 alkyl)(C1-5 alkyl), -(C1- 5 alkylene)-N(C1-5 alkyl)(C1-5 haloalkyl), -(C1-5 alkylene)-(N-heterocycloalkyl), -(C1-5 alkylene)-N(C1-5 haloalkyl)(C1-5 alkyl), -(C1-5 alkylene)-CONH2, -(C1-5 alkylene)-CONH(C1-5 alkyl), -(C1-5 alkylene)-CON(C1- 5 alkyl)(C1-5 alkyl), and -(C1-5 alkylene)-CO-(N-heterocycloalkyl). Even more preferably, RS2 is selected from halogen, -CN, -OH, -SH, -NH2, -(C1-5 alkylene)-CN, - (C1-5 alkylene)-OH, -(C1-5 alkylene)-SH, and -(C1-5 alkylene)-NH2. Even more preferably, RS2 is selected from halogen, -CN, -OH, -SH, and -NH2. In a first specific embodiment, -CR1R2R3 is bicyclo[1,1,1]pent-1-yl. In a second specific embodiment, RN is C1-4 alkyl. Particularly suitable C1-4 alkyl are methyl and ethyl groups. In a third specific embodiment, X2 is CH. In a fourth specific embodiment, X1 is CF and X3 is CH. In a fifth specific embodiment, -YC2-RC2 is aryl, preferably -YC2-RC2 is phenyl, wherein said aryl (said phenyl) is optionally substituted with one or more groups independently selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), and -CON(C1-5 alkyl)(C1-5 alkyl). In a sixth specific embodiment, -YC2-RC2 is heteroaryl, preferably selected from imidazolyl, pyridazinyl, thiazolyl, pyridinyl, pyrimidinyl, pyrazinyl, and indazolyl, wherein said heteroaryl is optionally substituted with one or more groups independently selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), and -CON(C1-5 alkyl)(C1-5 alkyl). In a seventh specific embodiment, -YC2-RC2 is heterocycloalkyl, preferably selected from morpholinyl, 1,1-dioxothiomorpholinyl, azetinyl, pyrrolidinyl, piperidinyl, 6-oxo-1,6- dihydropyridinyl, or piperazinyl, wherein said heterocycloalkyl is optionally substituted with one or more groups independently selected from RS2. More preferably, -YC2-RC2 is piperazinyl, optionally substituted with one or more groups independently selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), and -CON(C1- 5 alkyl)(C1-5 alkyl). Even more preferably, -YC2-RC2 is piperazinyl (preferably N-piperazinyl) optionally substituted (preferably N-substituted) with -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), and -CON(C1-5 alkyl)(C1-5 alkyl). Most preferably, -YC2-RC2 is piperazinyl (preferably N-piperazinyl) substituted (preferably N-substituted, preferably at a different N-atom than that attached to the ring system as shown in formula (I)), with -CON(C1-5 alkyl)(C1-5 alkyl), preferably with -CON(CH3)2. In an eighth specific embodiment, -YC2-RC2 is heterocycloalkyl, wherein said heterocycle comprises a spiro ring system, optionally selected from 2-oxaspiro[3.5]non-6-en-7-yl, 2-oxaspiro[3.5]non- 7-yl, 2-oxa-8-azaspiro[4.5]dec-8-yl, 9-oxa-3-azaspiro[5.5]undec-3-yl, 2-oxa-6-azaspiro[3.4]oct-6-yl, 1- oxa-7-azaspiro[3.5]non-7-yl, 1-oxa-8-azaspiro[4.5]dec-8-yl, 6-oxa-2-azaspiro[3.3]hept-2-yl, 2,8- diazaspiro[4.5]dec-8-yl, 7-oxa-3-azabicyclo[3.3.0]oct-3-yl, 8-oxa-3-azabicyclo[4.3.0]non-3-yl, 2-oxa-6- azaspiro[3.5]non-6-yl, 7-oxo-3,6,8-triazabicyclo[4.3.0]non-3-yl, 3-pyrrolino[3,4-c]pyrazol-2-yl, 3,6- diazabicyclo[3.1.1]hept-3-yl, and 2,7-diazaspiro[3.5]non-7-yl. In a ninth specific embodiment, R7 is hydrogen, -CN, or -Hal. Preferably, R7 is hydrogen. In this ninth specific embodiment, the moiety represented with a partial formula
Figure imgf000030_0001
is
Figure imgf000030_0002
. In a tenth specific embodiment, R8 is hydrogen, -CN, or -Hal. Preferably, R8 is hydrogen. In this tenth specific embodiment, the moiety represented with a partial formula
Figure imgf000030_0003
is
Figure imgf000030_0004
In an eleventh specific embodiment, R8 is -CH2C≡CH. In this eleventh specific embodiment, the moiety represented with a partial formula
Figure imgf000031_0001
. In a twelfth specific embodiment, R8 is -CH2-cycloalkyl or -CO-cycloalkyl. Particularly preferred cycloalkyl is cyclopropyl. In this twelfth embodiment, the moiety represented with a partial formula
Figure imgf000031_0002
In a thirteenth specific embodiment, Rcov is C2 alkenyl or
Figure imgf000031_0003
. In a fourteenth specific embodiment, Rcov is
Figure imgf000031_0004
. This embodiment is particularly preferred. In a fifteenth specific embodiment, W is -NHS(O)y- wherein y is 1 or 2. Preferably, y is 2. It is to be understood that the left side of W as defined herein is attached to the carbon atom that carries R1, R2 and R3. Preferred compound of formula (I) are selected from the following compounds or their pharmaceutically acceptable salts:
Figure imgf000031_0005
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Further preferred compounds of formula (I) are selected from the following compounds, or their pharmaceutically acceptable salts:
Figure imgf000035_0001
Figure imgf000036_0001
Further preferred compounds of formula (I) are selected from the following compounds, or their pharmaceutically acceptable salts:
Figure imgf000036_0002
Particularly preferred compounds of formula (I) are selected from the following compounds or their pharmaceutically acceptable salts:
Figure imgf000036_0003
Figure imgf000037_0001
Figure imgf000038_0001
The present invention also relates to each of the intermediates described further below in the examples section of this specification, including any one of these intermediates in non-salt form or in the form of a salt (e.g., a pharmaceutically acceptable salt) of the respective compound. Such intermediates can be used, in particular, in the synthesis of the compounds of formula (I).
The scope of the invention embraces all pharmaceutically acceptable salt forms of the compounds of formula (I) which may be formed, e.g., by protonation of an atom carrying an electron lone pair which is susceptible to protonation, such as an amino group, with an inorganic or organic acid, or as a salt of an acid group (such as a carboxylic acid group) with a physiologically acceptable cation. Exemplary base addition salts comprise, for example: alkali metal salts such as sodium or potassium salts; alkaline earth metal salts such as calcium or magnesium salts; zinc salts; ammonium salts; aliphatic amine salts such as trimethylamine, triethylamine, dicyclohexylamine, ethanolamine, diethanolamine, triethanolamine, procaine salts, meglumine salts, ethylenediamine salts, or choline salts; aralkyl amine salts such as N,N- dibenzylethylenediamine salts, benzathine salts, benethamine salts; heterocyclic aromatic amine salts such as pyridine salts, picoline salts, quinoline salts or isoquinoline salts; quaternary ammonium salts such as tetramethylammonium salts, tetraethylammonium salts, benzyltrimethylammonium salts, benzyltriethylammonium salts, benzyltributylammonium salts, methyltrioctylammonium salts or tetrabutylammonium salts; and basic amino acid salts such as arginine salts, lysine salts, or histidine salts. Exemplary acid addition salts comprise, for example: mineral acid salts such as hydrochloride, hydrobromide, hydroiodide, sulfate salts (such as, e.g., sulfate or hydrogensulfate salts), nitrate salts, phosphate salts (such as, e.g., phosphate, hydrogenphosphate, or dihydrogenphosphate salts), carbonate salts, hydrogencarbonate salts, perchlorate salts, borate salts, or thiocyanate salts; organic acid salts such as acetate, propionate, butyrate, pentanoate, hexanoate, heptanoate, octanoate, cyclopentanepropionate, decanoate, undecanoate, oleate, stearate, lactate, maleate, oxalate, fumarate, tartrate, malate, citrate, succinate, adipate, gluconate, glycolate, nicotinate, benzoate, salicylate, ascorbate, pamoate (embonate), camphorate, glucoheptanoate, or pivalate salts; sulfonate salts such as methanesulfonate (mesylate), ethanesulfonate (esylate), 2-hydroxyethanesulfonate (isethionate), benzenesulfonate (besylate), p-toluenesulfonate (tosylate), 2-naphthalenesulfonate (napsylate), 3-phenylsulfonate, or camphorsulfonate salts; glycerophosphate salts; and acidic amino acid salts such as aspartate or glutamate salts. Preferred pharmaceutically acceptable salts of the compounds of formula (I) include a hydrochloride salt, a hydrobromide salt, a mesylate salt, a sulfate salt, a tartrate salt, a fumarate salt, an acetate salt, a citrate salt, and a phosphate salt. A particularly preferred pharmaceutically acceptable salt of the compound of formula (I) is a hydrochloride salt. Accordingly, it is preferred that the compound of formula (I), including any one of the specific compounds of formula (I) described herein, is in the form of a hydrochloride salt, a hydrobromide salt, a mesylate salt, a sulfate salt, a tartrate salt, a fumarate salt, an acetate salt, a citrate salt, or a phosphate salt, and it is particularly preferred that the compound of formula (I) is in the form of a hydrochloride salt.
The present invention also specifically relates to the compound of formula (I), including any one of the specific compounds of formula (I) described herein, in non-salt form.
Moreover, the scope of the invention embraces the compounds of formula (I) in any solvated form, including, e.g., solvates with water (i.e., as a hydrate) or solvates with organic solvents such as, e.g., methanol, ethanol, isopropanol, acetic acid, ethyl acetate, ethanolamine, DMSO, or acetonitrile. All physical forms, including any amorphous or crystalline forms (i.e., polymorphs), of the compounds of formula (I) are also encompassed within the scope of the invention. It is to be understood that such solvates and physical forms of pharmaceutically acceptable salts of the compounds of the formula (I) are likewise embraced by the invention.
Furthermore, the compounds of formula (I) may exist in the form of different isomers, in particular stereoisomers (including, e.g., geometric isomers (or cis/trans isomers), enantiomers and diastereomers) or tautomers (including, in particular, prototropic tautomers, such as keto/enol tautomers or thione/thiol tautomers). All such isomers of the compounds of formula (I) are contemplated as being part of the present invention, either in admixture or in pure or substantially pure form. As for stereoisomers, the invention embraces the isolated optical isomers of the compounds according to the invention as well as any mixtures thereof (including, in particular, racemic mixtures/racemates). The racemates can be resolved by physical methods, such as, e.g., fractional crystallization, separation or crystallization of diastereomeric derivatives, or separation by chiral column chromatography. The individual optical isomers can also be obtained from the racemates via salt formation with an optically active acid followed by crystallization. The present invention further encompasses any tautomers of the compounds of formula (I). It will be understood that some compounds may exhibit tautomerism. In such cases, the formulae provided herein expressly depict only one of the possible tautomeric forms. The formulae and chemical names as provided herein are intended to encompass any tautomeric form of the corresponding compound and not to be limited merely to the specific tautomeric form depicted by the drawing or identified by the name of the compound.
The scope of the invention also embraces compounds of formula (I), in which one or more atoms are replaced by a specific isotope of the corresponding atom. For example, the invention encompasses compounds of formula (I), in which one or more hydrogen atoms (or, e.g., all hydrogen atoms) are replaced by deuterium atoms (i.e., 2H; also referred to as “D”). Accordingly, the invention also embraces compounds of formula (I) which are enriched in deuterium. Naturally occurring hydrogen is an isotopic mixture comprising about 99.98 mol-% hydrogen-1 (1H) and about 0.0156 mol-% deuterium (2H or D). The content of deuterium in one or more hydrogen positions in the compounds of formula (I) can be increased using deuteration techniques known in the art. For example, a compound of formula (I) or a reactant or precursor to be used in the synthesis of the compound of formula (I) can be subjected to an H/D exchange reaction using, e.g., heavy water (D2O). Further suitable deuteration techniques are described in: Atzrodt J et al., Bioorg Med Chem, 20(18), 5658-5667, 2012; William JS et al., Journal of Labelled Compounds and Radiopharmaceuticals, 53(11 -12), 635-644, 2010; Modvig A et al., J Org Chem, 79, 5861 -5868, 2014. The content of deuterium can be determined, e.g., using mass spectrometry or NMR spectroscopy. Unless specifically indicated otherwise, it is preferred that the compound of formula (I) is not enriched in deuterium. Accordingly, the presence of naturally occurring hydrogen atoms or 1H hydrogen atoms in the compounds of formula (I) is preferred.
The present invention also embraces compounds of formula (I), in which one or more atoms are replaced by a positron-emitting isotope of the corresponding atom, such as, e.g., 18F, 11C, 13N, 150, 76Br, 77Br, 120l and/or 124l. Such compounds can be used as tracers, trackers or imaging probes in positron emission tomography (PET). The invention thus includes (i) compounds of formula (I), in which one or more fluorine atoms (or, e.g., all fluorine atoms) are replaced by 18F atoms, (ii) compounds of formula (I), in which one or more carbon atoms (or, e.g., all carbon atoms) are replaced by 11C atoms, (iii) compounds of formula (I), in which one or more nitrogen atoms (or, e.g., all nitrogen atoms) are replaced by 13N atoms, (iv) compounds of formula (I), in which one or more oxygen atoms (or, e.g., all oxygen atoms) are replaced by 15O atoms, (v) compounds of formula (I), in which one or more bromine atoms (or, e.g., all bromine atoms) are replaced by 76Br atoms, (vi) compounds of formula (I), in which one or more bromine atoms (or, e.g., all bromine atoms) are replaced by 77Br atoms, (vii) compounds of formula (I), in which one or more iodine atoms (or, e.g., all iodine atoms) are replaced by 120l atoms, and (viii) compounds of formula (I), in which one or more iodine atoms (or, e.g., all iodine atoms) are replaced by 124l atoms. In general, it is preferred that none of the atoms in the compounds of formula (I) are replaced by specific isotopes.
The present invention further embraces the prodrugs of the compounds of formula (I). As preferably understood herein, the term “prodrug” of the compound of formula (I) refers to a derivative of the compounds of formula (I) that upon administration to a subject becomes metabolized to the said compound of formula (I). Said prodrugs of the compound of formula (I) may include modifications of -OH, -NH2, or -COOH group if present in the compound of formula (I), which preferably can be hydrolyzed to - OH, -NH2, or -COOH groups, respectively, e.g. upon administration to the subject. For example, as known to the skilled person, such prodrugs may preferably include for the compounds of formula (I) which comprise -OH moiety derivatives wherein said -OH moiety is turned into an -ORx moiety, wherein Rx preferably comprises a moiety selected from -CO-, -CH2-O-CO, -CH2-O-CO-O-, and -CH(CH3)-O-COO-, more preferably wherein Rx is selected from -CO-Ry, -CH2-O-CO-Ry, -CH2-O-CO-O-Ry, and -CH(CH3)-O- COO-Ry, wherein Ry is preferably carbocyclyl, heterocyclyl, C1-5 alkyl, -NH-(Ci-s alkyl) or -S-(Ci-s alkyl), wherein the said alkyl is optionally substituted with a group selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(Ci-5 alkyl), -O(Ci-5 haloalkyl), -SH, -S(Ci-5 alkyl), -S(Ci-5 haloalkyl), -NH2, -NH(Ci-s alkyl), -NH(CI-5 haloalkyl), -N(Ci-s alkyl)(Ci-5 alkyl), -N(Ci-s haloalkyl)(Ci-5 alkyl), -CONH2, -CONH(CI-5 alkyl), and -CON(Ci-s alkyl)(Ci 5 alkyl), and wherein the said carbocyclyl and heterocyclyl are each optionally substituted with a group selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(Ci-s alkyl), -O(Ci-5 haloalkyl), -SH, -S(Ci-5 alkyl), -S(Ci-5 haloalkyl), -NH2, -NH(Ci-s alkyl), -NH(Ci-s haloalkyl), -N(Ci-s alkyl)(Ci-5 alkyl), -N(Ci-s haloalkyl)(Ci-5 alkyl), -CONH2, -CONH(CI-5 alkyl), and -CON(CI-5 alkyl) (Ci -5 alkyl). Furthermore, for example, as known to the skilled person, such prodrugs may preferably include for the compounds of formula (I) which comprise -NH2 moiety derivatives wherein said -NH2 moiety is turned into -NHCOO-Ry moiety, wherein Ry is as defined hereinabove. Furthermore, for examples, as known to the skilled person, such prodrugs may preferably include for the compounds of formula (I) which comprise -COOH moiety derivatives wherein said -COOH group is turned into -COORy moiety, wherein Ry is as defined hereinabove. Further examples of groups that can be derivatized to yield prodrugs are known to the skilled person.
Pharmaceutical compositions
The compounds provided herein may be administered as compounds perse or may be formulated as medicaments. The medicaments/pharmaceutical compositions may optionally comprise one or more pharmaceutically acceptable excipients, such as carriers, diluents, fillers, disintegrants, lubricating agents, binders, colorants, pigments, stabilizers, preservatives, antioxidants, and/or solubility enhancers. The pharmaceutical compositions may comprise one or more solubility enhancers, such as, e.g., poly(ethylene glycol), including poly(ethylene glycol) having a molecular weight in the range of about 200 to about 5,000 Da (e.g., PEG 200, PEG 300, PEG 400, or PEG 600), ethylene glycol, propylene glycol, glycerol, a non-ionic surfactant, tyloxapol, polysorbate 80, macrogol-15-hydroxystearate (e.g., Kolliphor® HS 15, CAS 70142-34-6), a phospholipid, lecithin, dimyristoyl phosphatidylcholine, dipalmitoyl phosphatidylcholine, distearoyl phosphatidylcholine, a cyclodextrin, α-cyclodextrin, β-cyclodextrin, γ- cyclodextrin, hydroxyethyl-β-cyclodextrin, hydroxypropyl-β-cyclodextrin, hydroxyethyl-γ-cyclodextrin, hydroxypropyl-γ-cyclodextrin, dihydroxypropyl-β-cyclodextrin, sulfobutylether-β-cyclodextrin, sulfobutylether-γ-cyclodextrin, glucosyl-α-cyclodextrin, glucosyl-β-cyclodextrin, diglucosyl-β-cyclodextrin, maltosyl-α-cyclodextrin, maltosyl-β-cyclodextrin, maltosyl-γ-cyclodextrin, maltotriosyl-β-cyclodextrin, maltotriosyl-γ-cyclodextrin, dimaltosyl-β-cyclodextrin, methyl-β-cyclodextrin, a carboxyalkyl thioether, hydroxypropyl methylcellulose, hydroxypropylcellulose, polyvinylpyrrolidone, a vinyl acetate copolymer, vinyl pyrrolidone, sodium lauryl sulfate, dioctyl sodium sulfosuccinate, or any combination thereof. The pharmaceutical compositions may also comprise one or more preservatives, particularly one or more antimicrobial preservatives, such as, e.g., benzyl alcohol, chlorobutanol, 2-ethoxyethanol, m-cresol, chlorocresol (e.g., 2-chloro-3-methyl-phenol or 4-chloro-3-methyl-phenol), benzalkonium chloride, benzethonium chloride, benzoic acid (or a pharmaceutically acceptable salt thereof), sorbic acid (or a pharmaceutically acceptable salt thereof), chlorhexidine, thimerosal, or any combination thereof. The pharmaceutical compositions can be formulated by techniques known to the person skilled in the art, such as the techniques published in “Remington: The Science and Practice of Pharmacy”, Pharmaceutical Press, 22nd edition. The pharmaceutical compositions can be formulated as dosage forms for oral, parenteral, such as intramuscular, intravenous, subcutaneous, intradermal, intraarterial, intracardial, rectal, nasal, topical, aerosol or vaginal administration. Dosage forms for oral administration include coated and uncoated tablets, soft gelatin capsules, hard gelatin capsules, lozenges, troches, solutions, emulsions, suspensions, syrups, elixirs, powders and granules for reconstitution, dispersible powders and granules, medicated gums, chewing tablets and effervescent tablets. Dosage forms for parenteral administration include solutions, emulsions, suspensions, dispersions and powders and granules for reconstitution. Emulsions are a preferred dosage form for parenteral administration. Dosage forms for rectal and vaginal administration include suppositories and ovula. Dosage forms for nasal administration can be administered via inhalation and insufflation, for example by a metered inhaler. Dosage forms for topical administration include creams, gels, ointments, salves, patches and transdermal delivery systems. The compounds of formula (I) or the above described pharmaceutical compositions comprising a compound of formula (I) may be administered to a subject by any convenient route of administration, whether systemically/peripherally or at the site of desired action, including but not limited to one or more of: oral (e.g., as a tablet, capsule, or as an ingestible solution), topical (e.g., transdermal, intranasal, ocular, buccal, and sublingual), parenteral (e.g., using injection techniques or infusion techniques, and including, for example, by injection, e.g., subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, or intrasternal by, e.g., implant of a depot, for example, subcutaneously or intramuscularly), pulmonary (e.g., by inhalation or insufflation therapy using, e.g., an aerosol, e.g., through mouth or nose), gastrointestinal, intrauterine, intraocular, subcutaneous, ophthalmic (including intravitreal or intracameral), rectal, or vaginal administration.
If said compounds or pharmaceutical compositions are administered parenterally, then examples of such administration include one or more of: intravenously, intraarterially, intraperitoneally, intrathecally, intraventricularly, intraurethrally, intrasternally, intracardially, intracranially, intramuscularly or subcutaneously administering the compounds or pharmaceutical compositions, and/or by using infusion techniques. For parenteral administration, the compounds are best used in the form of a sterile aqueous solution which may contain other substances, for example, enough salts or glucose to make the solution isotonic with blood. The aqueous solutions should be suitably buffered (preferably to a pH of from 3 to 9), if necessary. The preparation of suitable parenteral formulations under sterile conditions is readily accomplished by standard pharmaceutical techniques well known to those skilled in the art.
Said compounds or pharmaceutical compositions can also be administered orally in the form of tablets, capsules, ovules, elixirs, solutions or suspensions, which may contain flavoring or coloring agents, for immediate-, delayed-, modified-, sustained-, pulsed- or controlled-release applications.
The tablets may contain excipients such as microcrystalline cellulose, lactose, sodium citrate, calcium carbonate, dibasic calcium phosphate and glycine, disintegrants such as starch (preferably corn, potato or tapioca starch), sodium starch glycolate, croscarmellose sodium and certain complex silicates, and granulation binders such as polyvinylpyrrolidone, hydroxypropylmethylcellulose (HPMC), hydroxypropylcellulose (HPC), sucrose, gelatin and acacia. Additionally, lubricating agents such as magnesium stearate, stearic acid, glyceryl behenate and talc may be included. Solid compositions of a similar type may also be employed as fillers in gelatin capsules. Preferred excipients in this regard include lactose, starch, a cellulose, or high molecular weight polyethylene glycols. For aqueous suspensions and/or elixirs, the agent may be combined with various sweetening or flavoring agents, coloring matter or dyes, with emulsifying and/or suspending agents and with diluents such as water, ethanol, propylene glycol and glycerin, and combinations thereof.
For oral administration, the compounds or pharmaceutical compositions are preferably administered by oral ingestion, particularly by swallowing. The compounds or pharmaceutical compositions can thus be administered to pass through the mouth into the gastrointestinal tract, which can also be referred to as “oral-gastrointestinal” administration.
Alternatively, said compounds or pharmaceutical compositions can be administered in the form of a suppository or pessary, or may be applied topically in the form of a gel, hydrogel, lotion, solution, cream, ointment or dusting powder. The compounds of the present invention may also be dermally or transdermally administered, for example, by the use of a skin patch.
Said compounds or pharmaceutical compositions may also be administered by sustained release systems. Suitable examples of sustained-release compositions include semi-permeable polymer matrices in the form of shaped articles, e.g., films, or microcapsules. Sustained-release matrices include, e.g., polylactides, copolymers of L-glutamic acid and gamma-ethyl-L-glutamate, poly(2-hydroxyethyl methacrylate), ethylene vinyl acetate, or poly-D-(— )-3-hydroxybutyric acid. Sustained-release pharmaceutical compositions also include liposomally entrapped compounds. The present invention thus also relates to liposomes containing a compound of the invention.
Said compounds or pharmaceutical compositions may also be administered by the pulmonary route, rectal routes, or the ocular route. For ophthalmic use, they can be formulated as micronized suspensions in isotonic, pH adjusted, sterile saline, or, preferably, as solutions in isotonic, pH adjusted, sterile saline, optionally in combination with a preservative such as a benzalkonium chloride. Alternatively, they may be formulated in an ointment such as petrolatum.
It is also envisaged to prepare dry powder formulations of the compounds of formula (I) for pulmonary administration, particularly inhalation. Such dry powders may be prepared by spray drying under conditions which result in a substantially amorphous glassy or a substantially crystalline bioactive powder. Accordingly, dry powders of the compounds of the present invention can be made according to an emulsification/spray drying process.
For topical application to the skin, said compounds or pharmaceutical compositions can be formulated as a suitable ointment containing the active compound suspended or dissolved in, for example, a mixture with one or more of the following: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, emulsifying wax and water. Alternatively, they can be formulated as a suitable lotion or cream, suspended or dissolved in, for example, a mixture of one or more of the following: mineral oil, sorbitan monostearate, a polyethylene glycol, liquid paraffin, polysorbate 60, cetyl esters wax, 2-octyldodecanol, benzyl alcohol and water. The present invention thus relates to the compounds or the pharmaceutical compositions provided herein, wherein the corresponding compound or pharmaceutical composition is to be administered by any one of: an oral route; topical route, including by transdermal, intranasal, ocular, buccal, or sublingual route; parenteral route using injection techniques or infusion techniques, including by subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, intrasternal, intraventricular, intraurethral, or intracranial route; pulmonary route, including by inhalation or insufflation therapy; gastrointestinal route; intrauterine route; intraocular route; subcutaneous route; ophthalmic route, including by intravitreal, or intracameral route; rectal route; or vaginal route. Preferred routes of administration are oral administration or parenteral administration. For each of the compounds or pharmaceutical compositions provided herein, it is particularly preferred that the respective compound or pharmaceutical composition is to be administered orally (particularly by oral ingestion).
Typically, a physician will determine the actual dosage which will be most suitable for an individual subject. The specific dose level and frequency of dosage for any particular individual subject may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the individual subject undergoing therapy.
A proposed, yet non-limiting dose of the compounds according to the invention for oral administration to a human (of approximately 70 kg body weight) may be 0.05 to 2000 mg, preferably 0.1 mg to 1000 mg, of the active ingredient per unit dose. The unit dose may be administered, e.g., 1 to 3 times per day. The unit dose may also be administered 1 to 7 times per week, e.g., with not more than one administration per day. It will be appreciated that it may be necessary to make routine variations to the dosage depending on the age and weight of the patient/subject as well as the severity of the condition to be treated. The precise dose and also the route of administration will ultimately be at the discretion of the attendant physician or veterinarian.
Therapeutic use
In one embodiment, the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as defined herein for use in therapy.
The present invention provides compounds that function as inhibitors of PARG. Thus, the present invention provides a method of inhibiting PARG enzyme activity in vitro or in vivo, said method comprising contacting a cell with an effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein.
Without wishing to be bound by the theory, the present inventors have demonstrated that certain compounds of formula (I) as described herein are covalent inhibitors of PARG enzyme. As shown in Table 2, the compounds of the present invention are significantly more potent (i.e., exhibit lower ICso) against the wild-type PARG protein in comparison to its C872A mutant, which is not capable of covalently binding the compounds of the present invention. As further demonstrated in Table 2, the inhibition of PARG by the compounds of the present invention is time-dependent, leading to lower ICso values upon 2-hour incubation when compared to a shorter incubation of 15 minutes.
The present invention also provides a method of selectively inhibiting PARG enzyme activity over PARP1 or ARH3 enzyme activity in vitro or in vivo. The said method comprises the steps of contacting a cell with an effective amount of a compound, or a pharmaceutically acceptable salt thereof, as defined herein.
In a further embodiment, the present invention relates to the compound of formula (I), as disclosed herein, for use in a method of treating a disease or disorder in which PARG activity is implicated in a subject or patient in need of such treatment. Said method of treatment comprises administering to said subject/patient a therapeutically effective amount of a compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as defined herein. In other words, in one embodiment the present invention relates to the compound of formula (I), as disclosed herein, for use in treating a disease or disorder in which PARG activity is implicated.
In a further embodiment, the present invention relates to a method of inhibiting cell proliferation, in vitro or in vivo, said method comprising contacting a cell with an effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein. Thus, the present invention relates to the compound of formula (I) or a pharmaceutically acceptable salt thereof for use in of inhibiting cell proliferation, in vitro or in vivo.
Thus, in a further embodiment, the present invention relates to a method of treating a proliferative disorder in a subject or patient in need of such treatment. The said method of treating a proliferative disorder in a subject or patient in need thereof comprises administering to said subject/patient a therapeutically effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as defined herein. Preferably as disclosed herein, the proliferative disorder is cancer. Thus, the present invention relates to a method of treating cancer in a subject or patient in need thereof. The said method of treating cancer in a subject or patient in need thereof comprises administering to said subject/patient a therapeutically effective amount of the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as defined herein. In a particular embodiment, the cancer is human cancer.
In one embodiment, the present invention relates to the compound of formula (I) or a pharmaceutically acceptable salt thereof, for use in treating a proliferative disorder. Preferably as disclosed herein, the proliferative disorder is cancer. Therefore, the present invention relates to the compound of formula (I) or a pharmaceutically acceptable salt thereof for use in treating cancer. In a particular embodiment, the cancer is human cancer.
In a further embodiment, the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, for use in the manufacture of a medicament for the treatment of a proliferative condition. In a preferred embodiment, the proliferative condition is cancer, more preferably a human cancer. Thus, preferably the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, for use in the manufacture of a medicament for the treatment of cancer, preferably for the treatment of human cancer.
In a further embodiment, the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, for use in the manufacture of a medicament for the inhibition of PARG enzyme activity. Preferably, the inhibition of PARG enzyme activity is selective inhibition of PARG enzyme activity over PARP1 or ARH3 enzyme activity. Thus, the present invention relates to the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein, for use in the manufacture of a medicament for the selective inhibition of PARG enzyme activity over PARP1 or ARH3 enzyme activity.
The present invention further provides the compound of formula (I), or a pharmaceutically acceptable salt thereof, as defined herein for use in the manufacture of a medicament for the treatment of a disease or disorder in which PARG activity is implicated, as defined herein.
As understood herein, the term "proliferative disorder" are used interchangeably herein and pertain to an unwanted or uncontrolled cellular proliferation of excessive or abnormal cells which is undesired, such as, neoplastic or hyperplastic growth, whether in vitro or in vivo. Examples of proliferative conditions include, but are not limited to, pre-malignant and malignant cellular proliferation, including but not limited to, malignant neoplasms and tumours, cancers, leukemias, psoriasis, bone diseases, fibroproliferative disorders (e.g., of connective tissues), and atherosclerosis. Any type of cell may be treated, including but not limited to, lung, colon, breast, ovarian, prostate, liver, pancreas, brain, and skin.
The anti-proliferative effects of the compound of formula (I) of the present invention have particular application in the treatment of human cancers (by virtue of their inhibition of PARG enzyme activity). The anti-cancer effect may arise through one or more mechanisms, including but not limited to, the regulation of cell proliferation, the inhibition of angiogenesis (the formation of new blood vessels), the inhibition of metastasis (the spread of a tumour from its origin), the inhibition of invasion (the spread of tumour cells into neighbouring normal structures), or the promotion of apoptosis (programmed cell death).
The antiproliferative treatment with the compound of formula (I) or a pharmaceutically acceptable salt thereof, as defined hereinbefore, may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional surgery or radiotherapy or chemotherapy. Such chemotherapy may include one or more of the following categories of anti-tumour agents:
(i) other antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and taxotere and polokinase inhibitors); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and camptothecin);
(ii) cytostatic agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestagens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5oc-reductase such as finasteride;
(iii) anti-invasion agents [for example c-Src kinase family inhibitors like 4-(6-chloro-2,3- methylenedioxyanilino)-7-[2-(4-methylpiperazin-1 -yl)ethoxy]-5-tetrahydropyran-4- yloxyquinazoline (AZD0530; International Patent Application WO 01/94341 ), N-(2-chloro-6- methylphenyl)-2-{6-[4-(2- hydroxyethyl)piperazin-1 -yl]-2-methylpyrimidin-4-ylamino}thiazole- 5-carboxamide (dasatinib, BMS- 354825; J. Med. Chem., 2004, 47, 6658-6661 ) and bosutinib (SKI-606), and metalloproteinase inhibitors like marimastat, inhibitors of urokinase plasminogen activator receptor function or antibodies to Heparanase];
(iv) inhibitors of growth factor function: for example such inhibitors include growth factor antibodies and growth factor receptor antibodies (for example the anti-erbB2 antibody trastuzumab [Herceptin™], the anti-EGFR antibody panitumumab, the anti-erbB 1 antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al. (Critical reviews in oncology/haematology, 2005, Vol. 54, pp1 1 -29); such inhibitors also include tyrosine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as N-(3-chloro- 4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy)quinazolin-4-amine (gefitinib, ZD1839), N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774) and 6- acrylamido-N-(3-chloro-4-fluorophenyl)-7-(3-morpholinopropoxy)-quinazolin-4-amine (Cl 1033), erbB2 tyrosine kinase inhibitors such as lapatinib); inhibitors of the hepatocyte growth factor family; inhibitors of the insulin growth factor family; inhibitors of the platelet-derived growth factor family such as imatinib and/or nilotinib (AMN107); inhibitors of serine/threonine kinases (for example Ras/Raf signalling inhibitors such as farnesyl transferase inhibitors, for example sorafenib (BAY 43-9006), tipifarnib (R1 15777) and lonafarnib (SCH66336)), inhibitors of cell signalling through MEK and/or AKT kinases, c-kit inhibitors, abl kinase inhibitors, PI3 kinase inhibitors, Plt3 kinase inhibitors, CSF-1 R kinase inhibitors, IGF receptor (insulin-like growth factor) kinase inhibitors; aurora kinase inhibitors (for example AZD1 152, PH739358, VX-680, MLN8054, R763, MP235, MP529, VX-528 AND AX39459) and cyclin dependent kinase inhibitors such as CDK2 and/or CDK4 inhibitors;
(v) antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (Avastin™) and for example, a VEGF receptor tyrosine kinase inhibitor such as vandetanib (ZD6474), vatalanib (PTK787), sunitinib (SU1 1248), axitinib (AG-013736), pazopanib (GW 786034) and 4-(4-fluoro-2-methylindol-5- yloxy)-6-methoxy-7-(3-pyrrolidin-1 - ylpropoxy)quinazoline (AZD2171 ; Example 240 within WO 00/47212), compounds such as those disclosed in International Patent Applications W097/22596, WO 97/30035, WO 97/32856 and WO 98/13354 and compounds that work by other mechanisms (for example linomide, inhibitors of integrin av03 function and angiostatin)];
(vi) vascular damaging agents such as Combretastatin A4 and compounds disclosed in International Patent Applications WO 99/02166, WO 00/40529, WO 00/41669, WO 01 Z92224, WO 02/04434 and WO 02/08213; (vii) an endothelin receptor antagonist, for example zibotentan (ZD4054) or atrasentan;
(viii) antisense therapies, for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
(ix) gene therapy approaches, including for example approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multidrug resistance gene therapy; and
(x) immunotherapy approaches, including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies.
In a particular embodiment, the antiproliferative treatment defined hereinbefore may involve, in addition to the compound of formula (I) of the invention, conventional surgery or radiotherapy or chemotherapy.Such conjoint treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment. Such combination products employ the compounds of this invention within the dosage range described hereinbefore and the other pharmaceutically-active agent within its approved dosage range.
According to this aspect the present invention further relates to the compound of formula (I) or a pharmaceutically acceptable salt as defined herein, for use in the treatment of a cancer (for example a cancer involving a solid tumour) in combination with another anti-tumour agent. The anti-tumour agent is preferably selected from the anti-tumour agents as listed hereinabove.
As understood herein, the term "combination" refers to simultaneous, separate or sequential administration. In one aspect of the invention "combination" refers to simultaneous administration. In another aspect of the invention "combination" refers to separate administration. In a further aspect of the invention "combination" refers to sequential administration. Where the administration is sequential or separate, the delay in administering the second component should not be such as to lose the beneficial effect of the combination.
Examples
The following examples are merely illustrative of the present invention and should not be construed to limit the scope of the invention which is defined by the appended claims. The compounds described in this section are defined by their chemical formulae and their corresponding chemical names. In case of conflict between any chemical formula and the corresponding chemical name indicated herein, the present invention relates to both the compound defined by the chemical formula and the compound defined by the chemical name, and particularly relates to the compound defined by the chemical formula.
Synthesis of the compounds of formula (I)
The syntheses of embodiments A, B, C and D of the compounds of formula (I) according to the present invention are preferably carried out according to the general synthetic sequences as shown in Schemes 1-6.
In addition to said routes described below, also other routes may be used to synthesize the target compounds, in accordance with common general knowledge of a person skilled in the art of organic synthesis. The order of transformations exemplified in the following Schemes is therefore not intended to be limiting, and suitable synthesis steps from various schemes can be combined to form additional synthesis sequences. In addition, modification of any of the substituents can be achieved before and/or after the exemplified transformations. These modifications can be such as the introduction of protective groups, cleavage of protective groups, reduction or oxidation of functional groups, halogenation, metalation, metal-catalyzed coupling reactions, substitution or other reactions known to a person skilled in the art. These transformations include those which introduce a functionality allowing for further interconversion of substituents. Appropriate protective groups and their introduction and cleavage are well-known to a person skilled in the art (see for example: Greene's Protective Groups in Organic Synthesis; Editor: P.G.M. Wuts, 5th edition, Wiley 2014). Specific examples are described in the subsequent paragraphs. Further, it is possible that two or more successive steps may be performed without work-up being performed between said steps, e.g. a “one-pot” reaction, as it is well-known to a person skilled in the art.
Scheme 1
Figure imgf000051_0001
Scheme 1 illustrates a preferred synthetic approach to compounds of the general formula A. As it is to be understandable to the skilled person, the scheme can also be extended to the compounds of formula (I) wherein W is -S(O)yNH-, -NHS(O)(NH)-, -NHS(O)(NCH3)-, -S(O)(NH)-NH-, or -S(O)(NCH3)-NH- upon the corresponding functionalization of the bromide of compound 1.
Figure imgf000052_0001
In the first step, a compound 1 in which X1, X2, X3 and R7 are are as defined for the compound of formula (I) reacts with benzyl mercaptan to give compound 2 in which X1, X2, X3 and R7 are are as defined for formula (I). This coupling reaction can be carried out by a palladium-catalyzed Carbon-Sulfur (C-S) cross-coupling reaction (see for example: Jiang, Buchwald in ‘Metal-Catalyzed Cross-Coupling Reactions’, 2nd edition.: de Meijere, Diederich, Eds.: Wiley-VCH: Weinheim, Germany, 2004). Preferred is the herein described use of tris(dibenzylideneacetone) dipalladium(O), (9,9-dimethyl-9H-xanthene-4,5- diyl)bis(diphenylphosphane) and N-ethyl-N-isopropylpropan-2-amine in dioxane. The reactions are preferably run under an atmosphere of argon for 1 - 48 hours at 80 - 100°C in a microwave oven or in an oil bath.
Figure imgf000052_0002
In the second step, compound 2 in which X1, X2, X3 and R7 are as defined for the compound of formula (I) reacts with an brominating reagent to give compound 3 in which X1, X2, X3 and R7 are are as defined for formula (I). This bromination can be carried out by treatment with A/-bromosuccinimide (NBS), Br2 etc., in MeCN, THF, dioxane, DMF etc. (see for example: Bentley et al; WO2011/138266). Preferred is the herein described use of NBS in MeCN. The reactions are preferably run under an atmosphere of argon for 0.5 - 5 hours at 0 °C to room temperature.
Figure imgf000052_0003
In the third step, compound 3 in which X1, X2, X3 and R7 are as defined for formula (I) reacts with 4 in which R8 is as defined for formula (I) to give compound 5 in which X1, X2, X3, R7 and R8 are as defined for formula (I). The coupling reaction is catalyzed by palladium catalysts, e.g. by Pd(0) catalysts like tetrakis(triphenylphosphine) palladium(O) [Pd(PPh3)4], tris(dibenzylideneacetone) di-palladium(O) [Pd2(dba)3], or by Pd(ll) catalysts like dichlorobis(triphenylphosphine)-palladium(ll) [Pd(PPh3)2Cb], palladium(ll) acetate and triphenylphosphine or by [l,l'-bis(diphenylphosphino)ferrocene]palladium dichloride. The reaction is preferably carried out in a solvent like 1 ,2-dimethoxyethane, dioxane, DMF, DME, THF, or isopropanol with water and in the presence of a base like potassium carbonate, sodium carbonate, sodium bicarbonate or potassium phosphate, (see for example: Hall, Boronic Acids, 2005 Wiley VCH Verlag GmbH & Co. KGaA, Weinheim, ISBN 3-527- 30991-8 and references cited therein). The reaction is performed at temperatures ranging from room temperature to the boiling point of the respective solvent. Further on, the reaction can be performed at temperatures above the boiling point using pressure tubes and a microwave oven. The reaction is preferably completed after 1 to 36 hours.
Figure imgf000053_0001
In the fourth step, compound 5 in which X1, X2, X3, R7 and R8 are as defined for formula (I) reacts with chlorination reagent to give a sulfonyl chloride 6 in which X1, X2, X3, R7 and R8 are as defined for formula (I). This sulfonyl chloride formation can be carried out by treatment with NCS (N- chlorosuccinimide), sulfonyl chloride, DCDMH (1 ,3-dichloro-5,5-dimethylhydantoin), CI2 etc., in MeCN with equivalent acetic acid and water (see for example: Sutton et al, WO 2021/055744). Preferred is the herein described use of DCDMH in MeCN with equivalent acetic acid and water. The reactions are preferably run under an atmosphere of argon for 0.5-5 hours at 0°C to room temperature.
Figure imgf000053_0002
In the fifth step, compound 6 in which X1, X2, X3, R7 and R8 are as defined for formula (I) reacts with an amine 7 in which R1 , R2 and R3 are as defined for formula (I) to give compound 8 in which X1, X2, X3, R7 and R8 are as defined for formula (I). This reaction can be carried out under basic conditions (see for example: Guo et al, WO2013/006394). Preferred is the herein described use of trimethylamine, pyridine etc., in DCM, THF or DMF. The reactions are preferably run under an atmosphere of argon for 0.5 - 24 hours at 0 °C to room temperature.
Figure imgf000054_0001
In the sixth step, compound 8 in which X1, X2, X3, R1, R2, R3, R7 and R8 are as defined for formula (I) is converted to a hydrazide compound 9 in which X1, X2, X3, R1, R2, R3, R7 and R8 are as defined for formula (I). This hydrazide formation is preferably carried out by treating with an amination reagent. Preferred is the herein described use of O-(4-nitrophenyl)hydroxylamine or O-(2,4- dinitrophenyl)hydroxylamine with potassium carbonate in DMF and dioxane. The reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 60 °C to 100 °C. (see for example: Boyles et al. Org. Progress. Res. Dev., 2002, 6, 230 - 233).
Figure imgf000054_0002
In the seventh step, compound 9 in which X1, X2, X3, R1, R2, R3, R7 and R8are as defined for formula (I) reacts with compound 10 in which RN is as defined for formula (I) and LG is a leaving group such as CI-, Br-, I-, MsO- or an aldehyde to give compound 11 in which X1, X2, X3, R1, R2, R3, R7, R8 and RN are as defined for formula (I). This alkylation is preferably carried out in basic condition. Preferred is the herein described use NaH, K2CO3 or CS2CO3 etc.in DCM, DMF or THF. (see for example: Sankaranarayananv et al, US2004106802). The alkylation is preferably run under an atmosphere of argon for 3-24 hours at 0 °C to 80 °C. When LG is aldehyde group, this reaction is preferably carried out with reductive amination reaction. Preferred is the herein described use aldehyde, acetic acid (cat.), molecular sieve in present of reducing agent in MeOH, dichloroethane (DCE), /-PrOH et. The reducing reagent can be, but not limited to, sodium cyanoborohydride or sodium triacetoxyborohydride. The reductive amination are preferably run under an atmosphere of argon for 12-24 hours at room temperature to 80 °C.(see for example: Ong et al, US2013203686).
Figure imgf000055_0001
In the final step, compound 11 in which X1, X2, X3, R1, R2, R3, R7, R8 and RN are as defined for formula (I) reacts with compound 12 in which Roov and Woov are as defined for formula (I) and LG is a leaving group such as HO-, Cl- or -O-Woov-Roov; to give a compound A in which X1, X2, X3, R1, R2, R3, R8, R7, RN, Roov and Woov are as defined for formula (I). When LG is HO-, this acylhydrazine formation is preferably carried out by condensation. Preferred is the herein described use A/-Ethoxycarbonyl-2-ethoxy-1,2- dihydroquinoline (EEDQ) in DCM or THF, or use 2-Chloro-1 -methylpyridinium (CMPI) in present of a base such as triethylamine, pyridine, di-7so-propylethylamine etc in DMSO, DMF, DCM or THF. The reaction is preferably run under an atmosphere of argon for 12-36 hours at room temperature to 80 °C (see for example: Dominic et Org. Process Res.Dev. 2005, 9, 499 - 507). When LG is Cl- or -O-Woov-Roov, this reaction can be carried out under basic conditions. Preferred is the herein described use of triethylamine, pyridine, di-/so-propylethylamine etc in DCM or THF under an atmosphere of argon for 2- 24 hours at 0 °C to 50 °C. (see for example: Lyer et al, Chem. Communications, 2018, 54, 11021 - 11024).
Scheme 2
Figure imgf000055_0002
Scheme 2 illustrates a preferred synthetic approach to compounds of the general formula B. As it is to be understandable to the skilled person, the scheme can also be extended to the compounds of formula (I) wherein W is -S(O)yNH-, -NHS(O)(NH)-, -NHS(O)(NCH3)-, -S(O)(NH)-NH-, or -S(O)(NCH3)-NH- starting from appropriate starting materials.
Figure imgf000056_0001
In the first step, compound 13 in which X1, X2 and X3 are are as defined for formula (I) reacts with chlorosulfonic acid to give compound 14 in which X1, X2 and X3 are are as defined for formula (I). Preferred is the herein described use of chlorosulfonic acid under an atmosphere of argon, (see for example: Adams et al, W02008070707). The reactions are preferably run in an oil bath for 2-24 hours at 0-140°C.
Figure imgf000056_0002
In the second step, compound 14 in which X1, X2 and X3 are as defined for formula (I) reacts with an amine 7 in which R1 , R2 and R3 are as defined for formula (I) to give compound 15 in which X1, X2, X3, R1, R2 and R3 as defined for formula (I). This reaction can be carried out under basic conditions (see for example: Guo et al, WO2013/006394). Preferred is the herein described use of trimethylamine, pyridine etc., in DCM, THF or DMF. The reactions are preferably run under an atmosphere of argon for 0.5-24 hours at 0 °C to room temperature.
Figure imgf000056_0003
In the third step, compound 15 in which X1, X2, X3, R1, R2 and R3 are as defined for formula (I) reacts with compound 16 in which R7 as defined for formula (I) to give compound 17 in which X1, X2, X3, R1, R2, R3 and R7 are as defined for formula (I). The conditions for this reaction can be found in: McGonagle et al, WO2016/092326.
Figure imgf000057_0001
In the fourth step, compound 17 in which X1, X2, X3, R1, R2, R3 and R7 are as defined for formula (I) is converted to a hydrazide compound 18 n which X1, X2, X3, R1, R2, R3 and R7 are as defined for formula (I). This hydrazide formation is preferably carried out by treating with amination reagent. Preferred is the herein described use of O-(4-nitrophenyl)hydroxylamine or O-(2,4-dinitrophenyl)hydroxylamine with potassium carbonate in DMF and dioxane. The reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 60°C to 100°C. (see for example: Boyles et al. Org. Progress. Res. Dev., 2002, 6, 230 - 233).
Figure imgf000057_0002
In the fifth step, compound 18 in which X1, X2, X3, R1, R2, R3 and R7 are as defined for formula (I) reacts with compound 10 in which RN is as defined for formula (I) and LG is a leaving group such as CI-, Br-, I-, MsO- or an aldehyde to give compound 19 in which X1, X2, X3, R1, R2, R3, R7 and RN are as defined for formula (I). This alkylation is preferably carried out in basic condition. Preferred is the herein described use NaH, K2CO3 or CS2CO3 etc.in DCM, DMF or THF. (see for example: Sankaranarayananv et al, US2004106802). The alkylation is preferably run under an atmosphere of argon for 3-24 hours at 0 °C to 80 °C. When LG is aldehyde group, this reaction is preferably carried out with reductive amination reaction. Preferred is the herein described use aldehyde, acetic acid (cat.), molecular sieve in present of reducing agent in MeOH, dichloroethane (DCE), /-PrOH et. The reducing reagent can be, but not limited to, sodium cyanoborohydride or sodium triacetoxyborohydride. The reductive amination are preferably run under an atmosphere of argon for 12-24 hours at room temperature to 80 °C.(see for example: Ong et al, US2013203686).
Figure imgf000058_0003
In the final step, compound 19 in which X1, X2, X3, R1, R2, R3, R7 and RN are as defined for formula (I) reacts with compound 12 in which Roov and Woov are as defined for formula (I) and LG is leaving group such as HO-, Cl- or -O-Woov-Roov to give compound B in which X1, X2, X3, R1, R2, R3, R7, RN, Roov and Woov are as defined for formula (I). When LG is HO-, this acylhydrazine formation is preferably carried out by condensation. Preferred is the herein described use A/-Ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ) in DCM or THF, or use 2-chloro-1 -methylpyridinium (CMPI) in present of a base such as triethylamine, pyridine, di-/so-propylethylamine etc in DMSO, DMF, DCM or THF. The reaction is preferably run under an atmosphere of argon for 12-36 hours at room temperature to 80 °C (see for example: Dominic et Org. Process Res. Dev. 2005, 9, 499 - 507). When LG is Cl- or -O-Woov-Roov, this reaction can be carried out under basic conditions. Preferred is the herein described use of triethylamine, pyridine, di-/so-propylethylamine etc in DCM or THF under an atmosphere of argon for 2-24 hours at 0 °C to 50 °C. (see for example: Lyer et al, Chem. Communications, 2018, 54, 11021 - 11024).
Scheme 3
Figure imgf000058_0001
Scheme 3 illustrates an alternative pathway to access to compounds of the general formula 17. As it is to be understandable to the skilled person, the scheme can also be extended to compounds of formula (I) wherein W is -S(O)yNH-, -NHS(O)(NH)-, -NHS(O)(NCH3)-, -S(O)(NH)-NH-, or -S(O)(NCH3)-NH-.
Figure imgf000058_0002
In the first step, a compound of formula 15 in which X1, X2, X3, R1, R2 and R3 are as defined for the compound of formula (I) is converted to a compound of formula 18 in which X1, X2, X3, R1, R2, R3 are as defined for the compound of formula (I). This amide formation can be carried out with ammonium hydroxide or ammonium salt and base such as triethylamine, pyridine, di-/so-propylethylamine etc. in the presence of condensating agents such as HATU (1 -[Bis(dimethylamino)methylene]-1 H-1 ,2,3-triazolo[4,5- b]pyridinium 3-Oxide Hexafluorophosphate), EDCI/HOBt (N-(3-Dimethylaminopropyl)-N'- ethylcarbodiimide hydrochloride/ Hydroxybenzotriazole), T3P, GDI (di(1 H-imidazol-1-yl)methanone)etc.in DCM or DMF (see for example: Weaver et al, W02020/257940). This amide formation can also be conducted by acyl chloride strategy. The reactions are preferably run under an atmosphere of argon for 2-24 hours at room temperature.
Figure imgf000059_0001
In the final step, compound 20 in which X1, X2, X3, R1, R2 and R3 are as defined for formula (I) reacts with compound 19 in which R7 is as defined for formula (I) to give compound 17 in which X1, X2, X3, R1, R2, R3and R7 are as defined forformula (I). The conditions ofthis reaction can be found for example in: Mcgonagle et al, WO2016/092326.
Scheme 4
Figure imgf000060_0001
Scheme 4 illustrates a preferred synthetic approach to compounds of the general formula C. As it is to be understandable to the skilled person, the scheme can also be extended to the compounds of formula (I) wherein W is -S(O)yNH-, -NHS(O)(NH)-, -NHS(O)(NCH3)-, -S(O)(NH)-NH-, or -S(O)(NCH3)-NH- starting from appropriate starting materials.
Figure imgf000060_0002
22 23
In the first step, compound 22 in which X1, X2 and X3 are as defined for formula (I) reacts with amine 7 in which R1’ R2 and R3 are as defined for formula (I) to give compound 23 in which X1, X2, X3, R1, R2 and R3 are as defined for formula (I). This reaction can be carried out under basic conditions, (see for example: Guo et al, WO2013/006394). Preferred is the herein described use of triethylamine, pyridine, di-/so- propylethylamine etc in DCM, THF or DMF. The reactions are preferably run under an atmosphere of argon for 0.5 - 24 hours at 0 °C to room temperature.
Figure imgf000061_0001
In the second step, compound 23 in which X1, X2, X3, R1, R2 and R3 are as defined for formula (I) reacts with amine 24 in which R8 is as defined for formula (I) to give compound 25 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I). Preferred is the herein described use of triethylamine, di-/so- propylethylamine etc in DMF, acetonitrile or dioxane, (see for example: Liu et al, Eur. J. Med. Chem., 2021 , 222, 113565). The reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 80 - 110°C in a microwave oven or in an oil bath.
Figure imgf000061_0003
In the third step, compound 25 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I) is converted to compound 26 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I). This amide formation can be carried out with ammonium hydroxide or ammonium salt and base such as triethylamine, pyridine, di-7so-propylethylamine etc. in the presence of coulping agents such as HATU, EDCI/HOBt, T3P, GDI etc.in DCM or DMF, (see for example: Weaver et al, W02020/257940). or in the presence of an acyl chloride. The reactions are preferably run under an atmosphere of argon for 2 - 24 hours at room temperature.
Figure imgf000061_0002
In the fourth step, compound 26 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I) is converted to compound 27 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I). This cyclization is preferably carried out with GDI in the presence of base such as triethylamine, di-/so- propylethylamine etc. in DMF, NMP or DMA. The reactions are preferably run for 0.5 - 16 hours at 80 - 120°C in a microwave oven or in an oil bath (see for example: Velaparthi et al, WO2021/133751). This cyclization also can be carried out with GDI in DMF, NMP or DMA etc. without base at 100 - 150°C, or with triphosgene and base in DCM, THF etc. at 0 °C to room temperature.
Figure imgf000062_0001
In the fifth step, compound 27 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I) is converted to hydrazide compound 28 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I). This hydrazide formation is preferably carried out by treating with amination reagent. Preferred is the herein described use of O-(4-nitrophenyl)hydroxylamine or O-(2,4-dinitrophenyl)hydroxylamine with potassium carbonate in DMF and dioxane. The reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 60 °C to 100 °C (see for example: Boyles et al, Eur. J. Med. Chem.,Org. Pro. Res. Dev., 2002, 6, 230 - 233).
Figure imgf000062_0002
In the sixth step, compound 28 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I) reacts with compound 10 in which RN is as defined for formula (I) and LG is a leaving group such as CI-, Br-, I-, MsO- or an aldehyde to give compound 29 in which X1, X2, X3, R1, R2, R3, R8 and RN are as defined for formula (I). This alkylation is preferably carried out in basic condition. Preferred is the herein described use NaH, K2CO3 or CS2CO3 etc.in DCM, DMF or THF (see for example: Sankaranarayananv et al, US2004106802). The alkylation is preferably run under an atmosphere of argon for 3-24 hours at 0 °C to 80 °C. When LG is aldehyde group, this reaction is preferably carried out with reductive amination reaction. Preferred is the herein described use aldehyde, acetic acid (cat.), molecular sieve in present of reducing agent in MeOH, dichloroethane (DCE), /-PrOH et. The reducing reagent can be, but not limited to, sodium cyanoborohydride or sodium triacetoxyborohydride. The reductive amination are preferably run under an atmosphere of argon for 12-24 hours at room temperature to 80 °C.(see for example: Ong et al, US2013203686).
Figure imgf000063_0001
In the final step, compound 29 in which X1, X2, X3, R1, R2, R3, R8 and RN are as defined for formula (I) reacts with compound 12 in which Roov and Woov are as defined for formula (I) and LG is leaving group such as HO-, Cl- or -O-Woov-Roov to give compound C in which X1, X2, X3, R1, R2, R3, R8, W, RN, Roov and Woov are as defined for formula (I). When LG is HO-, this acylhydrazine formation is preferably carried out by condensation. Preferred is the herein described use A/-Ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ) in DCM or THF, or use 2-Chloro-1 -methylpyridinium (CMPI) in presence of a base such as triethylamine, pyridine, di-/so-propylethylamine etc in DMSO, DMF, DCM or THF. The reaction is preferably run under an atmosphere of argon for 12-36 hours at room temperature to 80 °C (see for example: Dominic et Org. Process Res. Dev. 2005, 9, 499 - 507). When LG is Cl- or -O-Woov-Roov, this reaction can be carried out under basic conditions. Preferred is the herein described use of triethylamine, pyridine, di-/so-propylethylamine etc in DCM or THF under an atmosphere of argon for 2-24 hours at 0 °C to 50 °C. (see for example: Lyer et al, Chem. Communications, 2018, 54, 11021 - 11024).
Scheme 5
Figure imgf000063_0002
Scheme 5 illustrates an alternative pathway to access to compounds of the general formula 26. As it is to be understandable to the skilled person, the scheme can also be extended to the compounds of formula (I) wherein W is -S(O)yNH-, -NHS(O)(NH)-, -NHS(O)(NCH3)-, -S(O)(NH)-NH-, or -S(O)(NCH3)-NH-.
Figure imgf000064_0001
In the first step, compound 30 in which X1, X2 and X3 are as defined for formula (I) reacts with chlorosulfonic acid to give compound 31 in which X1, X2 and X3 are as defined for formula (I). Preferred is the herein described use of chlorosulfonic acid under an atmosphere of argon (see for example: Adams et al, W02008070707). The reactions are preferably run in an oil bath for 2 - 24 hours at 0 - 140°C.
Figure imgf000064_0002
In the second step, compound 31 in which X1, X2 and X3 are as defined for formula (I) reacts with compound 7 in which R1, R2 and R3 are as defined for formula (I) to give compound 32 in which X1, X2, X3, R1, R2 and R3 are as defined for formula (I). This reaction can be carried out under basic conditions (see for example: Sari et al, Eur. J. Med. Chem., 2017, 138, 407 - 421). Preferred is the herein described use of triethylamine, pyridine, di-/so-propylethylamine etc. in DCM, THF or DMF. The reactions are preferably run under an atmosphere of argon for 0.5 - 24 hours at 0 °C to room temperature.
Figure imgf000064_0003
In the final step, compound 32 in which X1, X2, X3, R1, R2 and R3 are as defined for formula (I) reacts with amine 24 in which R8 is as defined for formula (I) to give compound 26. Preferred is the herein described use of triethylamine, di-/so-propylethylamine etc in DMF, acetonitrile or dioxane (see for example: Putman et al, WO2016144792). The reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 80 - 110°C in a microwave oven or in an oil bath. Scheme 6
Figure imgf000065_0001
Scheme 6 illustrates a preferred synthetic approach to compounds of the general formula D. As it is to be understandable to the skilled person, the scheme can also be extended to the compounds of formula (I) wherein W is -S(O)yNH-, -NHS(O)(NH)-, -NHS(O)(NCH3)-, -S(O)(NH)-NH-, or -S(O)(NCH3)-NH- starting from appropriate starting materials.
Figure imgf000065_0002
In the first step, compound 33 in which X1, X2 and X3 are as defined for formula (I) reacts with compound 34 in which R8 is as defined for formula (I) to give compound 35 in which X1, X2, X3 and R8 are as defined for formula (I). The condition of this reaction can be found for example in Mcgonagle et al, WO2016/092326.
Figure imgf000066_0001
In the second step, compound 35 in which X1, X2, X3 and R8 are as defined for formula (I) reacts with hydrazine hydrate to give compound 36 in which X1, X2, X3 and R8 are as defined for formula (I). The condition of this reaction can be found for example in Mcgonagle et al, WO2016/092326.
Figure imgf000066_0002
In the third step, compound 36 in which X1, X2, X3 and R8 are as defined for formula (I) reacts with benzyl mercaptan to give compound 37 in which X1, X2, X3 and R8 are as defined for formula (I). This coupling reaction can be carried out by a palladium-catalyzed carbon-sulfur (C-S) cross-coupling reaction (see for example: Jiang, Buchwald in ‘Metal-Catalyzed Cross-Coupling Reactions’, 2nd edition.: de Meijere, Diederich, Eds.: Wiley-VCH: Weinheim, Germany, 2004). Preferred is the herein described use of tris(dibenzylideneacetone) dipalladium(O), (9,9-dimethyl-9H-xanthene-4,5-diyl)bis(diphenylphosphane) and A/-ethyl-N-isopropylpropan-2-amine in dioxane. The reactions are preferably run under an atmosphere of argon for 1 - 48 hours at 80 - 100°C in a microwave oven or in an oil bath.
Figure imgf000066_0003
In the fourth step, compound 37 in which X1, X2, X3 and R8 are as defined for formula (I) reacts with a chlorination reagent to give sulfonyl chloride compound 38 in which X1, X2, X3 and R8 are as defined for formula (I). This sulfonyl chloride formation can be carried out by treatment with NCS, sulfonyl chloride, DCDMH, CI2 etc., in MeCN with equivalent acetic acid and water (see for example: Sutton et al, WO 2021/055744). Preferred is the herein described use of DCDMH in MeCN with equivalent acetic acid and water. The reactions are preferably run under an atmosphere of argon for 0.5 - 5 hours at 0 °C to room temperature.
Figure imgf000067_0001
In the fifth step, compound 38 in which X1, X2, X3, R8 are as defined for formula (I) reacts with amine 7 in which R1’ R2 and R3 are as defined for formula (I) to give compound 39 in which X1, X2, X3, R1’ R2, R3 and R8 are as defined for formula (I). This reaction can be carried out under basic conditions (see for example: Guo et al, WO2013/006394). Preferred is the herein described use of trimethylamine, pyridine etc., in DCM, THF or DMF. The reactions are preferably run under an atmosphere of argon for 0.5 - 24 hours at 0 °C to room temperature.
Figure imgf000067_0002
39 40
In the sixth step, compound 39 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I) is converted to a hydrazide compound 40 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I). This hydrazide formation is preferably carried out by treating with amination reagent. Preferred is the herein described use of O-(4-nitrophenyl)hydroxylamine or O-(2,4-dinitrophenyl)hydroxylamine with potassium carbonate in DMF and dioxane. The reactions are preferably run under an atmosphere of argon for 2 - 24 hours at 60 °C to 100 °C (see for example: Boyles et al, Eur. J. Med. Chem.,Org. Pro. Res. Dev., 2002, 6, 230 - 233).
Figure imgf000067_0003
In the seventh step, compound 40 in which X1, X2, X3, R1, R2, R3 and R8 are as defined for formula (I) reacts with compound 10 in which RN is as defined for formula (I) and LG is a leaving group such as CI-, Br-, I-, MsO- or an aldehyde to give compound 41 in which X1, X2, X3, R1, R2, R3, R8 and RN are as defined for formula (I). This alkylation is preferably carried out in basic condition. Preferred is the herein described use NaH, K2CO3 or CS2CO3 etc.in DCM, DMF or THF (see for example: Sankaranarayananv et al, US2004106802). The alkylation is preferably run under an atmosphere of argon for 3-24 hours at 0 °C to 80 °C. When LG is aldehyde group, this reaction is preferably carried out with reductive amination reaction. Preferred is the herein described use aldehyde, acetic acid (cat.), molecular sieve in present of reducing agent in MeOH, dichloroethane (DCE), /-PrOH et. The reducing reagent can be, but not limited to, sodium cyanoborohydride or sodium triacetoxyborohydride. The reductive amination are preferably run under an atmosphere of argon for 12-24 hours at room temperature to 80 °C.(see for example: Ong et al, US2013203686).
Figure imgf000068_0001
In the final step, compound 41 in which X1, X2, X3, R1, R2, R3, R8 and RN are as defined for formula (I) reacts with compound 12 in which Roov and Woov are as defined for formula (I) and LG is leaving group such as HO-, Cl- or -O-Woov-Roov to give a compound D in which X1, X2, X3, R1, R2, R3, R8, RN, Roov and Woov are as defined for formula (I). When LG is HO-, this acylhydrazine formation is preferably carried out by condensation. Preferred is the herein described use A/-Ethoxycarbonyl-2-ethoxy-1 ,2-dihydroquinoline (EEDQ) in DCM or THF, or use 2-Chloro-1 -methylpyridinium (CMPI) in present of a base such as triethylamine, pyridine, di-7so-propylethylamine etc in DMSO, DMF, DCM or THF. The reaction is preferably run under an atmosphere of argon for 12-36 hours at room temperature to 80 °C (see for example: Dominic et Org. Process Res.Dev. 2005, 9, 499 - 507). When LG is Cl- or -O- Woov-Roov, this reaction can be carried out under basic conditions. Preferred is the herein described use of triethylamine, pyridine, di-7so-propylethylamine etc in DCM or THF under an atmosphere of argon for 2-24 hours at 0 °C to 50 °C. (see for example: Lyer et al, Chem. Communications, 2018, 54, 11021 - 11024).
Preparative examples
The compounds described in this section are defined by their chemical formulae and their corresponding chemical names. In case of conflict between any chemical formula and the corresponding chemical name indicated herein, the present invention relates to both the compound defined by the chemical formula and the compound defined by the chemical name, and particularly relates to the compound defined by the chemical formula.
General considerations
Abbreviations used in the descriptions that follows are: aq. (aqueous); br. (broad, 1H NMR signal); BuLi (Butyl lithium); CDCh (deuterated chloroform); GDI (di(1 H-imidazol-1-yl)methanone); cHex (cyclohexane); CS2CO3 (cesium carbonate); DCE (dichloroethane); d (doublet, 1H NMR signal); DCC (N,N'-Dicyclohexylcarbodiimide) DCM (dichloromethane); DIEA (di-/so-propylethylamine); DIPEA (di-/so- propylethylamine); DMAP (4- A/-A/-dimethylaminopyridine), DME (1 ,2-dimethoxyethane), DMF (N-N- dimethylformamide); DMF (dimethylformamide); DMSO (dimethyl sulfoxide); EEDQ (2-ethoxyquinoline- 1 (2H)-carboxylate); ES (electrospray); EtOAc (ethyl acetate); EtOH (ethanol); h (hour(s)); HATU (1- [Bis(dimethyl ami no)methylene]- 1 H-1 , 2, 3-triazolo[4, 5-b] pyridi ni um 3-oxide hexafluorophosphate,
Hexafluorophosphate Azabenzotriazole Tetramethyl Uranium); 1H NMR (proton nuclear magnetic resonance spectroscopy); H2O (water); HCI (hydrochloric acid); HF (hydrogen fluoride); HPLC (High Performance Liquid Chromatography); HSO3CI (chlorosulfuric acid); iPrOH (/so-propanol); K2CO3 (potassium carbonate); KOCN ((potassium cyanate); LCMS (liquid chromatograpy mass spectrometry); □HMDS (Lithium bis(trimethylsilyl)amide); m (multiplet, 1H NMR signal); mCPBA (meta- chloroperoxybenzoic acid), MeCN (acetonitrile), MeLi (methyl lithium); MeOH (methanol); min (minute(s)); Mn02 (manganese dioxide); MS (mass spectrometry); MTBE (methyl ferf-butyl ether); Na2SO4 (sodium sulfate); NaHCOs (sodium hydrogenacarbonate); NBS (N-bromosuccinimide); NH4HCO3 (ammonium bicarbonate); NH3.H2O (aqueous ammonia); N2 (nitrogen); NMP (N-methyl-2pyrrolidine); NMR (nuclear magnetic resonance); Pd2dba3 (tris(dibenzylideneacetone)dipalladium(O)); prep-HPLC (preparative High Performance Liquid Chromatography); q (quartet, 1 H NMR signal); quin (quintet, 1 H NMR signal); rac (racemic); RT (retention time); s (singlet, 1H NMR signal); sat (saturated); t (triplet, 1H NMR signal); TBAF (tetrabutylammonium fluoride); TEA (triethylamine); TFA (trifluoroacetic acid); TFAA (trifluoroacetic anhydride), THF (tetrahydrofuran); TIPSCI (triisopropylsilyl chloride); TLC (Thin Layer Chromatography); UPLC (Ultra-High Performance Liquid Chromatography), UV (ultraviolet), wt-% (percent by weight); Xantphos (4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene); Zn (Zinc).
General Procedure: All starting materials and solvents were obtained either from commercial sources or prepared according to literature references. Commercially available reagents and anhydrous solvents were used as supplied, without further purification. Unless otherwise stated all reactions were stirred. All air- and moisture-sensitive reactions were carried out in oven-dried (at 120°C) glassware under an inert atmosphere of nitrogen or argon. Compound names were generated using ChemDraw Professional (Perkin Elmer). In some cases, generally accepted names of commercially available reagents were used in place of ChemDraw generated names.
Reversed Phase HPLC conditions for LCMS Analysis of final compounds:
Method 1 : SHIMADZU LCMS-2020 Kinetex EVO C18 2.1X30 mm, 5 urn at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5 mL/min; eluted with the mobile phase over 0.80 min employing UV detection at 220 nm and 254 nm. Gradient information: 0- 0.80 min, ramped from 95% A-5% B to 5% A-95% B; 0.80-1 .20 min, held at 5% A-95% B; 1 .20-1 .21 min, returned to 95% A-5% B, 1.21-1 .55 min, held at 95% A-5% B.
Method 2 : SHIMADZU LCMS-2020 Kinetex EVO C18 2.1X30 mm, 5 urn at 40°C ; Mobile Phase : A: 0.025% NH3-H2O in water (v/v) , B: MeCN; flow rate held at 1 .5 mL/min; eluted with the mobile phase over 1.55 min employing UV detection at 220 nm and 254 nm. Gradient information: 0-0.80 min, ramped from 95% A-5% B to 5% A-95% B; 0.80-1 .20 min, held at 5% A-95% B; 1 .20-1 .21 min, returned to 95% A-5% B, 1.21-1 .55 min, held at 95% A-5% B.
Method 3: Agilent 1200\G6110A Kinetex EVO C18 2.1X30 mm, 5 urn at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5.0 mL/min; eluted with the mobile phase over 0.80 min employing UV detection at 220 nm and 254 nm. Gradient information: 0.01-0.80 min, ramped from 95% A-5% B to 5% A-95% B; 0.80-1 .20 min, held at 5% A-95% B; 1 .20-1 .21 min, returned to 95% A-5% B, 1.21-1.50 min, held at 95% A-5% B.
Method 4: SHIMADZU LCMS-2020 Kinetex® EVO C18 2.1X30 mm 5 urn at 50°C Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5 mL/min; eluted with the mobile phase over 1.00 min employing UV detection at 220 nm and 254 nm. Gradient information: 0.01-0.80 min, ramped from 95% A-5% B to 5% A-95% B; 0.80-0.95 min, held at 5% A-95% B; 0.95-0.96 min, returned to 95% A-5% B, 0.96-1 .00 min, held at 95% A-5% B.
Method 5: SHIMADZU LCMS-2020 Kinetex® EVO C18 2.1X20 mm 2.6 urn at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 2.0 mL/min; eluted with the mobile phase over 1.00 min employing UV detection at 220 nm and 254 nm. Gradient information: 0.01-0.60 min, ramped from 95% A-5% B to 5% A-95% B; 0.61-0.78 min, held at 5% A-95% B; 0.78-0.79 min, returned to 95% A-5% B, 0.79-0.80 min, held at 95% A-5% B.
Method 6: SHIMADZU LCMS-2020 Kinetex® EVO C18 2.1X30 mm 5 urn at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5 mL/min; eluted with the mobile phase over 1.00 min employing UV detection at 220 nm and 254 nm. Gradient information: 0.01-0.60 min, ramped from 95% A-5% B to 5% A-95% B; 0.60-0.78 min, held at 5% A-95% B; 0.78-0.79 min, returned to 95% A-5% B, 0.79-0.80 min, held at 95% A-5% B.
Method 7: SHIMADZU LCMS-2020 Kinetex EVO C18 2.1X30mm,5um at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 2.0 mL/min; eluted with the mobile phase over 0.80 min employing UV detection at 220 nm and 254 nm. Gradient information: 0- 0.80 min, ramped from 95% A-5% B to 5% A-95% B; 0.80-1 .20 min, held at 5% A-95% B; 1 .20-1 .21 min, returned to 95% A-5% B, 1.21-1 .55 min, held at 95% A-5% B.
Method 8: SHIMADZU LCMS-2020 Kinetex EVO C18 2.1X30mm,5pm at 50°C; Mobile Phase: A: 0.0375% TFA in water (v/v); B: 0.01875% TFA in MeCN (v/v); flow rate held at 1 .5 mL/min; eluted with the mobile phase over 1 .55 min employing UV detection at 220 nm and 254 nm. Gradient information: 0- 0.80 min, ramped from 95% A-5% B to 5% A-95% B; 0.80-1 .20 min, held at 5% A-95% B; 1 .20-1 .21 min, returned to 95% A-5% B, 1.21-1 .55 min, held at 95% A-5% B.
1H N MR Spectroscopy:
1H NMR spectra were acquired on a Bruker Avance IH spectrometer at 400 MHz using residual undeuterated solvent as reference. 1H NMR signals are specified with their multiplicity / combined multiplicities as apparent from the spectrum; possible higher-order effects are not considered. Chemical shifts of the signals (5) are specified as ppm (parts per million).
Salt stoichiometry:
In the present text, in particular in the experimental section, for the synthesis of intermediates and of examples of the present invention, when a compound is mentioned as a salt form with the corresponding base or acid, the exact stoichiometric composition of said salt form, as obtained by the respective preparation and/or purification process, is, in most cases, unknown. Unless specified otherwise, suffixes to chemical names or structural formulae such as "hydrochloride", "trifluoroacetate", "sodium salt", or "x HO", "x CF3COOH", "x Na+", for example, are to be understood as not a stoichiometric specification, but solely as a salt form. This applies analogously to cases in which synthesis intermediates or example compounds or salts thereof have been obtained, by the preparation and/or purification processes described, as solvates, such as hydrates with (if defined) unknown stoichiometric composition.
Preparation of Intermediate 1.1
3-carbamoyl-4-fluorobenzenesulfonyl chloride
Figure imgf000071_0001
A solution of 2-fluorobenzamide (95 g, 682.83 mmol) in HSO3Cl (477.40 g, 4.10 mol) was stirred at 140 °C for 2 h. LCMS showed starting material remained. The mixture was stirred at 140 °C for another 1 h. The mixture was poured into ice (2 L) carefully and filtered. The cake was dried in vacuum to give the product 3-carbamoyl-4-fluorobenzenesulfonyl chloride (150 g, 631.22 mmol, 92.44% yield) as a white solid. RT 0.376 min (Method 4); m/z 237.9 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 7.87 (dd, J = 6.8, 2.0 Hz, 1H), 7.78 (br s, 1H), 7.72-7.67 (m, 1H), 7.61 (br s, 1H), 7.21 (dd, J = 10.4, 8.4 Hz, 1H). Preparation of Intermediate 1.2 2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000072_0001
To a solution of 1-methylcyclopropanamine (0.8 g, 11.25 mmol, HCl salt) and TEA (2.13 g, 21.09 mmol) in NMP (6 mL) at 0 °C was added dropwise a solution of 3-carbamoyl-4-fluorobenzenesulfonyl chloride (3.34 g, 14.06 mmol) in DCM (25 mL). The mixture was stirred at 20 °C for 16 h. The resulting mixture was distilled off under vacuum to give the NMP solution. The solution was diluted with water (20 mL), then adjusted with aq. HCl(1N) to pH 5-6. The aqueous phase was extracted with EtOAc (100 mL, 2x). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 2-fluoro-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (1.3 g, 4.23 mmol, 30.07% yield, 88.55% purity) as a white solid. RT 0.352 min (Method 4); m/z 273.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 8.17 (s, 1H), 8.06 (dd, J = 6.8, 2.4 Hz, 1H), 7.94-7.89 (m, 2H), 7.87-7.83 (m, 1H), 7.52 (t, J = 10.0 Hz, 1H), 1.07 (s, 3H), 0.63-0.57 (m, 2H), 0.43-0.37 (m, 2H) Preparation of Intermediate 1.3 1-methylcyclopropane-1-carboxamide
Figure imgf000072_0002
To a solution of 1-methylcyclopropane-1-carboxylic acid (22.0 g, 219.75 mmol) in DCM (220 mL) was added DMF (1.61 g, 21.97 mmol, 1.69 mL). Then oxalyl dichloride (33.47 g, 263.70 mmol, 23.08 mL) was added dropwise to the mixture at 0°C under N2. The mixture was stirred at 20 °C for 2 h under N2. The mixture was concentrated in vacuum at 20 °C to give methylcyclopropanecarbonyl chloride (26.05 g, crude) which was used for next step without further purification. To a solution of methylcyclopropanecarbonyl chloride (26.05 g, crude) in THF (330 mL) was added NH3.H2O (82.50 g, 659.15 mmol, 90.66 mL, 28% purity) dropwise at 0 °C under N2. The mixture was stirred at 20 °C for 2 h under N2 before it was concentrated and filtered to give a solid which was dried under reduced pressure to give the product 1-methylcyclopropane-1-carboxamide (14.8 g, 149.30 mmol, 67.95% yield) as a white solid. 1 HNMR (CDCl3, 400 MHz): δ = 5.70 (s, 2H), 1.34 (s, 3H), 1.23-1.20 (m, 2H), 0.64-0.60 (m, 2H). Preparation of Intermediate 1.4 (1-methylcyclopropyl)methanamine
Figure imgf000073_0001
To a solution of 1-methylcyclopropane-1-carboxamide (14.8 g, 149.30 mmol) in THF (200 mL) at 0 °C was added dropwise BH3.THF (1 M, 328.46 mL). The mixture was stirred at 20 °C for 16 h. To the mixture was added MeOH (50 mL) and aqueous HCl solution (1N, 50 mL) drop-wise at 20°C. Then the mixture was heated to 50°C and stirred for 0.5 h. The mixture was concentrated under vacuum to give the crude product. The crude product was triturated with EtOAc (200 mL) at 20 °C for 10 min and filtered. The filter cake was dried under vacuum to give the product (1-methylcyclopropyl)methanamine (23.0 g, crude, HCl salt) as a white solid. 1 HNMR (DMSO-d6, 400 MHz): δ = 6.54 (s, 2H), 2.67 (s, 2H), 1.10 (s, 3H), 0.51-0.57 (m, 2H), 0.32- 0.38 (m, 2H). Preparation of Intermediate 1.5 2-(((1-methylcyclopropyl)methyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl) benzamide
Figure imgf000073_0002
To a solution of 2-fluoro-5-[(1-methylcyclopropyl)sulfamoyl]benzamide (1.4 g, 5.14 mmol) in MeCN (15 mL) was added DIPEA (3.99 g, 30.85 mmol) and (1-methylcyclopropyl)methanamine (2.03 g, 16.71 mmol, HCl salt). The mixture was stirred at 85 °C for 16 h. To the mixture was added (1- methylcyclopropyl)methanamine (1.63 g, 13.37 mmol, HCl salt) and DIPEA (1.99 g, 15.42 mmol). The mixture was stirred at 100 °C for 48 h. To the mixture was added (1-methylcyclopropyl)methanamine (1.88 g, 15.42 mmol) and DIPEA (3.32 g, 25.71 mmol). The mixture was stirred at 100 °C for 16 h. To the mixture was added potassium carbonate (7.11 g, 51.41 mmol). The mixture was stirred at 100 °C for 16 h. To the mixture was added K2CO3 (2 g, 14.47 mmol). The mixture was stirred at 100 °C for 8 h. The reaction mixture was adjusted to pH<6 with aqueous HCl solution (1N). The mixture was extracted with EtOAc (100 mL, 2x). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue, which was triturated with petroleum ether/ ethyl acetate 10:1 (30 mL) at 20 °C for 10 min. The mixture was filtered and the filter cake was dried under reduced pressure to give the product 2-(((1- methylcyclopropyl)methyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl) benzamide (1.28 g, 3.56 mmol, 69.16% yield, 93.96% purity) as a white solid. RT 0.524 min (method 4); m/z 338.1 (M+H)+ (ESI+); 1 HNMR (DMSO-d6, 400 MHz): 8.73 (s, 1H), 8.19-8.03 (m, 1H), 7.87 (d, J = 2.0 Hz, 1H), 7.57 (dd, J = 8.8, 1.6 Hz, 1H), 7.52 (s, 1H), 7.30 (s, 1H), 6.77 (d, J = 9.2 Hz, 1H), 3.05 (d, J = 3.6 Hz, 2H), 1.11 (s, 3H), 1.05 (s, 3H), 0.62- 0.57 (m, 2H), 0.47-0.42 (m, 2H), 0.37-0.31 (m, 4H). Preparation of Intermediate 1.6 N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline- 6-sulfonamide
Figure imgf000074_0001
To a solution of 2-(((1-methylcyclopropyl)methyl)amino)-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (1.8 g, 5.33 mmol) in 1-methylpyrrolidin-2-one (20 mL) was added 1,1’-carbonyldiimidazole (6.92 g, 42.67 mmol). The mixture was stirred at 130 °C for 3 h before it was poured into aq. HCl solution (400 mL, 1N) and extracted with EtOAc (200 mL, 2x). The combined organic layers were washed with brine (200 mL, 2x), dried over with anhydrous Na2SO4, filtered and the filtrate was dried under reduced pressure to give the product N-(1-methylcyclopropyl)-1-((1- methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (1.7 g, 4.36 mmol, 81.65% yield, 93.12% purity) as a yellow solid. RT 0.372 min (method 5); m/z 364.1 (M+H)+ (ESI+); 1 HNMR (DMSO-d6, 400 MHz): δ = 11.85 (s, 1H), 8.35 (d, J = 2.4 Hz, 1H), 8.16 (s, 1H), 8.03 (dd, J = 8.8, 2.4 Hz, 1H) 7.72 (d, J = 9.2 Hz, 1H), 4.12 (s, 2H), 1.07 (s, 3H), 1.02 (s, 3H), 0.62-0.57 (m, 2H), 0.56-0.52 (m, 2H), 0.41-0.36 (m, 2H), 0.32-0.27 (m, 2H). Preparation of Intermediate 1.7 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000075_0001
To a solution of N-(1-methylcyclopropyl)-1-[(1-methylcyclopropyl)methyl]-2,4-dioxo-quinazoline-6- sulfonamide (300 mg, 825.5 µmol) in dioxane (3 mL) and DMF (3 mL) was added potassium carbonate (171.13 mg, 1.24 mmol). The mixture was stirred at 70 °C for 0.5 h., Then, O-(4- nitrophenyl)hydroxylamine (152.67 mg, 990.56 µmol) was added and the mixture was stirred at 70 °C for 20 h. The reaction mixture was poured into water (50 mL) and extracted with EtOAc (50 mL; 2x). The combined organic layer was washed with brine (20 mL; 2x), dried over Na2SO4, filtered and the filtrate was concentrated under reduce pressure. The resulting residue was triturated with MTBE (30 mL) for 15 min to give the product 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (230 mg, 607.75 µmol, 73.62% yield) as a yellow solid. RT 0.555 min (method 1); m/z 379.0 (M+H)+ (ESI+); Preparation of Example 1 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acrylamide
Figure imgf000075_0002
To a solution of 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (50 mg, 132.12 µmol) in DCM (1 mL) were added TEA (26.74 mg, 264.24 µmol) and acryloyl chloride (23.33 mg, 184.97 µmol) at 0 °C. The mixture was stirred at 0 °C for 2 h and, then, concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: Waters Xbridge 150*25 mm* 5 µm; mobile phase: A: 1 mM aqueous solution of NH4HCO3, B: MeCN; B%: 31%-61%, 8 min) to give the product N-(1-((1-methylcyclopropyl)methyl)-6- (N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)acrylamide (4.32 mg, 9.59 µmol, 7.26% yield, 96% purity) as a white solid. RT 0.784 min (method 2); m/z 433.1 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 8.42 (d, J = 2.4 Hz, 1H), 8.22-8.30 (m, 1H), 8.12 (dd, J = 8.8, 2.4 Hz, 1H), 7.85 (d, J = 9.2 Hz, 1H), 6.42-6.50 (m, 1H), 6.24-6.32 (m, 1H), 5.88 (dd, J = 9.2 Hz, 1H), 4.16-4.31 (m, 2H), 1.09 (s, 3H), 1.04 (s, 3H), 0.53-0.62 (m, 4H), 0.40-0.43 (m, 2H), 0.32-0.30 (m, 2H). Preparation of Example 2 2-chloro-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acetamide
Figure imgf000076_0001
To a solution of 3-amino-N-(1-methylcyclopropyl)-1-[(1-methylcyclopropyl)methyl]-2,4-dioxo- quinazoline-6-sulfonamide (70 mg, 184.97 µmol) and TEA (56.15 mg, 554.91 µmol, 77.24 µL) in DCM (2 mL) was added 2-chloroacetyl chloride (20.89 mg, 184.97 µmol, 14.71 µL) at 0 °C. The mixture was warmed to 20 °C, stirred at 20 °C for 2 h and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: Waters Xbridge 150*25 mm* 5 µm; mobile phase: A: 1 mM aqueous solution of NH4HCO3, B: MeCN; B%: 31%-61%,9 min) and lyophilized directly to give the product 2-chloro-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acetamide (13 mg, 27.43 µmol, 14.83% yield, 96% purity) as a white solid. RT 0.846 min (method 1); m/z 455.1 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 8.42 (d, J = 2.4 Hz, 1H), 8.20-8.35 (m, 1H), 8.12 (m, 1H), 7.85 (d, J = 9.2 Hz, 1H), 4.38 (s, 2H), 4.15-4.34 (m, 2H), 1.09 (s, 3H), 1.04 (s, 3H), 0.49-0.63 (m, 4H), 0.38-0.44 (m, 2H), 0.32-0.30 (m, 2H). Preparation of Example 3 2-fluoro-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acrylamide
Figure imgf000076_0002
To a solution of 2-fluoroacrylic acid (4.76 mg, 52.85 µmol) in THF (0.5 mL) was added ethyl 2- ethoxy-2H-quinoline-1-carboxylate (13.07 mg, 52.85 µmol). The mixture was stirred at 20 °C for 1 h. Then, to the mixture was added 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4- dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (20 mg, 52.85 µmol). The mixture was stirred at 20 °C for 16 h. The mixture was concentrated under reduced pressure. The resulting residue was purified by preparative-HPLC (column: Phenomenex Synergi C18150*25 mm*10 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 31%-61%, 10 min) to give the product 2-fluoro-N-(1-((1- methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)acrylamide (1.47 mg, 2.60 µmol, 4.92% yield, 96% purity, 2 FA salt) as white solid. RT 0.858 min (method 1); m/z 522.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz) 8.37-8.46 (m, 3H), 8.25 (dd, J = 7.2, 4.0 Hz, 1H), 8.12 (dd, J = 8.8, 2.4 Hz, 1H), 7.85 (d, J = 8.8 Hz, 1H), 5.63 (d, J = 2.4 Hz, 1H), 5.49 (d, J = 2.4 Hz, 1H), 4.17-4.31 (m, 2H), 1.07 (s, 3H), 1.03 (s, 3H), 0.51-0.63 (m, 4H), 0.38- 0.43 (m, 2H), 0.31 (s, 2H). Preparation of Intermediate 4.1 But-2-ynoic anhydride
Figure imgf000077_0001
To a solution of but-2-ynoic acid (500 mg, 5.95 mmol) in DCM (10 mL) was added DCC (736.24 mg, 3.57 mmol) at 0 °C. Then, the mixture was stirred at 0 °C for 1 h, warmed to 20 °C and stirred for 12 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure. The residue was purified by column chromatography (SiO2, petroleum ether/EtOAc =1/0 to 100/1) to give the product but-2-ynoic anhydride (230 mg, 1.53 mmol, 25.76% yield) as a white solid. 1H NMR (DMSO-d6, 400 MHz): δ = 1.93 (s, 6H). Preparation of Example 4 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)but-2-ynamide
Figure imgf000077_0002
To a solution of 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (120 mg, 317.09 µmol) and TEA (128.34 mg, 1.27 mmol) in DCM (2 mL) was added but-2-ynoic anhydride (95.21 mg, 634.17 µmol) at 0 °C. The reaction mixture was stirred at 0 °C for 2 h under N2 and was concentrated under reduced pressure to give a residue, which was purified by preparative HPLC (column: Phenomenex Synergi C18150*25 mm* 10 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 25%-56%, 10min) to give the product N-(1-((1- methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)but-2-ynamide (3 mg, 6.68 µmol, 2.11% yield, 99% purity) as a white solid. RT 0.807 min (method 3); m/z 445.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 8.48-8.38 (m, 2H), 8.34-8.16 (br, 1H), 8.10 (dd, J = 8.8, 2.0 Hz, 1H), 7.83 (d, J = 9.2 Hz, 1H), 4.28-4.14 (m, 2H), 2.08 (s, 3H), 1.07 (s, 3H), 1.04-1.00 (m, 3H), 0.62-0.50 (m, 4H), 0.42-0.37 (m, 2H), 0.33-0.28 (m, 2H). Preparation of Intermediate 5.1 (Z)-but-2-enoic anhydride
Figure imgf000078_0001
To a solution of (Z)-but-2-enoic acid (500 mg, 5.81 mmol) in DCM (10 mL) was added DCC (719.00 mg, 3.48 mmol) at 0 °C. The mixture was stirred at 0 °C for 1 h, then, was warmed to 20 °C and stirred for 1 h. The reaction mixture was filtered and the filtrate was concentrated. The residue was purified by column chromatography (SiO2, Petroleum ether/EtOAc =100/1 to 20/1) to give the product (Z)-but-2-enoic anhydride (250 mg, 1.62 mmol, 27.92% yield) as yellow oil. Preparation of Example 5 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)but-2-enamide
Figure imgf000078_0002
To a solution of 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (80 mg, 211.39 µmol) and TEA (106.95 mg, 1.06 mmol, 147.12 µL) in DCM (3 mL) was added (Z)-but-2-enoic anhydride (39.11 mg, 253.67 µmol) at 0 °C. The reaction mixture was stirred at 20 °C for 1 h under N2 and concentrated under reduced pressure. The resulting residue was purified by preparative HPLC (column: Phenomenex luna C18150*40 mm* 15 µm; mobile phase: A: 0.1% TFA in water, B: MeCN; B%: 1%-20%,10 min) and lyophilized directly to give the product N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)but-2-enamide (2.5 mg, 5.18 µmol, 2.5% yield, 92.5% purity, cis/trans: 2/3 evaluated by 1H NMR) as a white solid. RT 0.850 min (method 1); m/z 446.9 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 10.79 (s, 1H), 8.41 (t, J = 2.0 Hz, 1H), 8.27-8.20 (m, 1H), 8.11 (d, J = 8.8 Hz, 1H), 7.83 (d, J = 9.2 Hz, 1H), 6.89- 5.84 (m, 1H), 5.30-5.15 (m, 1H), 4.30-4.14 (m, 2H), 3.20-1.83 (m, 3H), 1.08 (s, 3H), 1.03 (s, 3H), 0.62- 0.50 (m, 4H), 0.44-0.39 (m, 2H), 0.33 (s, 2H). Preparation of Example 6 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)but-2-enamide
Figure imgf000079_0001
To a solution of 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (80 mg, 211.39 µmol) in DCM (1 mL) was added TEA (64.17 mg, 634.17 µmol) and (E)-but-2-enoic anhydride (45.62 mg, 295.95 µmol) at 0 °C. The mixture was stirred at 20 °C for 12 h and, then concentrated under reduced pressure to give a residue, which was purified by preparative HPLC (column: Unisil 3-100 C18 Ultra 150*50 mm*3 µm; mobile phase: A: 0.225% formic acid in water, B%: 28%-58%,10 min) to give the product N-(1-((1-methylcyclopropyl)methyl)-6-(N- (1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)but-2-enamide (20 mg, 44.34 µmol, 20.98% yield, 99% purity, cis/trans: 1/1 evaluated by 1H NMR) as a white solid. RT 0.789 min (method 3); m/z 447.3 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 10.79 (br, 1H), 8.41 (t, J = 2.0 Hz, 1H), 8.27-8.20 (m, 1H), 8.11 (d, J = 8.8 Hz, 1H), 7.83 (d, J = 9.2 Hz, 1H), 6.89- 5.84 (m, 1H), 5.30-5.15 (m, 1H), 4.30-4.14 (m, 2H), 3.20-1.83 (m, 3H), 1.08 (s, 3H), 1.03 (s, 3H), 0.62- 0.50 (m, 4H), 0.44-0.39 (m, 2H), 0.33 (s, 2H). Preparation of Example 7 2-cyano-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acetamide
Figure imgf000079_0002
To a solution of 2-cyanoacetic acid (8.99 mg, 105.70 µmol) in THF (0.5 mL) was added ethyl 2- ethoxy-2H-quinoline-1-carboxylate (26.14 mg, 105.70 µmol), The mixture was stirred at 20 °C for 30 min. Then, to the mixture was added 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4- dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (20 mg, 52.85 µmol). The mixture was stirred at 20 °C for 16 h and then, concentrated under reduced pressure to give a residue, which was purified by preparative -HPLC (column: Phenomenex Synergi C18150* 25 mm* 10 µm; mobile phase: A: 1 mM aqueous solution of NH4HCO3, B: MeCN; B%: 3%-33%, 9 min) to give the product 2-cyano-N-(1-((1- methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)acetamide (7.52 mg, 16.66 µmol, 31.52% yield, 98.67% purity) as an off-white solid. RT 0.820 min (method 1); m/z 446.1 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 8.38-8.43 (m, 1H), 8.25 (br, 1H), 8.10 (dd, J = 8.8, 2.0 Hz, 1H), 7.83 (d, J = 8.8 Hz, 1H), 4.13-4.32 (m, 2H), 3.98 (s, 2H), 1.07 (s, 3H), 1.03 (s, 3H), 0.49-0.65 (m, 4H), 0.40 (m, 2H), 0.28-0.34 (m, 2H). Preparation of Example 8 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)-2-(trifluoromethyl)acrylamide
Figure imgf000080_0001
To a solution of 2-(trifluoromethyl) acrylic acid (14.80 mg, 105.70 µmol) in THF (0.5 mL) was added ethyl 2-ethoxy-2H-quinoline-1-carboxylate (26.14 mg, 105.70 µmol). The mixture was stirred at 20 °C for 30 min followed by addition of3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4- dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (20 mg, 52.85 µmol). The mixture was stirred at 20 °C for 16 h and, then concentrated under reduced pressure to give a residue, which was purified by preparative -HPLC (column: Phenomenex Synergi C18150*25 mm*10 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 37%-67%, 10min) to give the product N-(1-((1- methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)-2-(trifluoromethyl)acrylamide (7.03 mg, 13.68 µmol, 25.89% yield, 97.4% purity) as an off-white solid. RT 0.883 min (method 1); m/z 501.1 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz) 8.42 (d, J = 2.4 Hz, 1H), 8.27 (s, 1H), 8.13 (dd, J = 8.8, 2.4 Hz, 1H), 7.87 (d, J = 9.2 Hz, 1H), 6.67 (s, 2H), 4.17-4.31 (m, 2H), 1.08 (s, 3H), 1.03 (s, 3H), 0.51-0.62 (m, 4H), 0.38-0.44 (m, 2H), 0.32 (s, 2H). Preparation of Intermediate 9.1 3-(methylamino)-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000080_0002
3 batches as in the following protocol were run in parallel and mixed for the purification: To a solution of formaldehyde (19.84 mg, 198.18 µmol, 18.20 µL, 30% purity in water), 3-amino-N-(1- methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide (50 mg, 132.12 µmol) and 4 Å molecular sieve -325 mesh particle size (20 mg) in MeOH (1 mL) was added acetic acid (793.38 ug, 13.21 µmol). The mixture was stirred at 20 °C for 12 h and, then sodium cyanoborohydride (66.42 mg, 1.06 mmol) was added. The mixture was stirred at 20 °C for 2 h and, then, cyanoborohydride (50 mg) was added. This operation was repeated one additional time followed by stirring at 20°C during 12h. Then, the mixture was filtered and the filtrate. was concentrated under reduced pressure. The resulting residue was purified by preparative-HPLC (column: Waters Xbridge 150*50 mm*10 µm; mobile phase: A: 1 mM aqueous solution of NH4HCO3, B: MeCN; B%: 35%- 61%, 11 min) to give the product 3-(methylamino)-N-(1-methylcyclopropyl)-1-((1- methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (40 mg, 101.92 µmol, 25.71% yield) as a white solid. 1H NMR (DMSO-d6, 400 MHz): δ = 8.41 (d, J = 2.4 Hz, 1H), 8.19 (s, 1H), 8.04 (dd, J = 9.2, 2.4 Hz, 1H), 7.76 (d, J = 9.2 Hz, 1H), 6.02 (q, J = 5.6 Hz, 1H), 4.21 (s, 2H), 2.60 (d, J = 5.6 Hz, 3H), 1.07 (s, 3H), 1.03 (s, 3H), 0.54-0.62 (m, 4H), 0.36-0.43 (m, 2H), 0.29-0.33 (m, 2H). Preparation of Example 9 2-chloro-N-methyl-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4- dioxo-1,4-dihydroquinazolin-3(2H)-yl)acetamide
Figure imgf000081_0001
To a solution of 3-(methylamino)-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4- dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (10 mg, 25.48 µmol), TEA (5.16 mg, 50.96 µmol, 7.09 µL) in DCM (0.2 mL) was added 2-chloroacetyl chloride (5.76 mg, 50.96 µmol, 4.05 µL) in DCM (0.2 mL) at 0 °C. The mixture was stirred at 20 °C for 1 h, then, diluted with H2O (10mL) and extracted with EtOAc (10mL; 2x). The combined organic layer was washed with brine (10mL; 2x), dried over Na2SO4 and concentrated under reduced pressure. The resulting residue was purified by preparative HPLC (column: Waters Xbridge 150*50 mm*10 µm; mobile phase: A: 1 mM aqueous solution of NH4HCO3, B: MeCN; B%: 30%-60%, 9 min) to give the product 2-chloro-N-methyl-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)acetamide (2.78 mg, 5.93 µmol, 23.27% yield, 100% purity) as a yellow solid. RT 0.891 min (method 1); m/z 467.1 (M+H)+ (ESI+); 1H NMR (CDCl3, 400 MHz) 8.64 (dd, J = 14.4, 2.4 Hz, 1H), 8.07-8.16 (m, 1H), 7.34-7.46 (m, 1H), 4.90 (d, J=16.8 Hz, 1H), 3.83-4.32 (m, 4H), 3.20-3.49 (m, 3H), 1.22 (d, J = 12.8 Hz, 3H), 1.05 (s, 3H), 0.68-0.75 (m, 2H), 0.45-0.57 (m, 4H), 0.34-0.43 (m, 2H). Preparation of Example 10 N-methyl-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acrylamide
Figure imgf000082_0001
To a solution of 3-(methylamino)-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl) methyl)-2,4- dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (10 mg, 25.48 µmol) in THF (0.3 mL) was added NaHCO3 (21.40 mg, 254.80 µmol) in H2O (0.3 mL), followed by acryloyl chloride (23.06 mg, 254.80 µmol, 20.78 µL) at 0 °C. The reaction mixture was stirred at 0 °C for 1 h, then, diluted with H2O (10mL) and extracted with EtOAc (10 mL; 2x). The combined organic layer was washed with brine (10mL; 2x), dried over Na2SO4 and concentrated under reduced pressure. The resulting residue was purified by preparative HPLC (column: Waters Xbridge 150*50 mm*10 µm; mobile phase: A: 1 mM aqueous solution of NH4HCO3, B: MeCN; B%: 37%-67%, 8 min) to give the product N-methyl-N-(1-((1- methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)acrylamide (1.46 mg, 3.20 µmol, 12.54% yield, 97.74% purity) as a yellow solid. RT 0.844 min (method 1); m/z 447.2 (M+H)+ (ESI+); 1H NMR (CDCl3, 400 MHz): δ = 8.70-8.75 (m, 1 H), 8.13-8.23 (m, 1 H), 7.42-7.51 (m, 1 H), 6.64-6.73 (m, 1 H), 6.41-6.54 (m, 1 H), 6.09 (m, 1 H), 5.61- 5.96 (m, 1 H), 4.92-5.03 (m, 1 H), 4.11-4.40 (m, 2 H), 3.29-3.56 (m, 3 H), 1.26-1.33 (m, 3 H), 1.09-1.16 (m, 3 H), 0.76-0.83 (m, 2 H), 0.52-0.66 (m, 4 H), 0.40-0.49 (m, 2 H). Preparation of Intermediate 11.1 2-chloro-2-fluoroacetyl chloride
Figure imgf000082_0002
To a solution of ethyl 2-chloro-2-fluoroacetate (4 g, 28.46 mmol) in chlorosulfonic acid (5 mL) was added benzene-1,2-dicarbonyl chloride (11.56 g, 56.92 mmol). The mixture was stirred at 120 °C for 4 h in a still connected to an dry ice-cooled receiver, and the product was collected in the dry ice-cooled receiver to give the product 2-chloro-2-fluoroacetyl chloride (1.5 g, 11.46 mmol, 40.25% yield) as a fuming colorless liquid which was used in the next step. 1H NMR (CDCl3, 400 MHz): δ = 6.33-6.52 (d, J = 50.8 Hz, 1H). Preparation of Example 11 2-chloro-2-fluoro-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4- dioxo-1,4-dihydroquinazolin-3(2H)-yl)acetamide
Figure imgf000083_0001
To a solution of 3-amino-N-(1-methylcyclopropyl)-1-[(1-methylcyclopropyl)methyl]-2,4-dioxo- quinazoline-6-sulfonamide (30 mg, 79.27 µmol) and TEA (32.09 mg, 317.09 µmol, 44.13 µL) in THF (1 mL) was added 2-chloro-2-fluoro-acetyl chloride (12.46 mg, 95.13 µmol) in DCM (0.5 mL) at 0 °C under N2. The mixture was warmed to 20 °C stirred at 20 °C for 0.5 h and concentrated under reduced pressure. The resulting residue was purified by preparative HPLC (column: Phenomenex Synergi Polar-RP 100*25 mm*4 µm; mobile phase: A: 0.1% TFA in water, B: MeCN; B%: 45%-65%, 7 min) and lyophilized directly to give the product 2-chloro-2-fluoro-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)acetamide (6 mg, 12.43 µmol, 15.68% yield, 98% purity) as a white solid. RT 0.767 min (method 1); m/z 473.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 11.81 (d, J = 2.8 Hz, 1H), 8.43 (t, J = 2.4 Hz, 1H), 8.27 (s, 1H), 8.14 (m, 1H), 7.88 (d, J = 9.2 Hz, 1H), 7.07-7.26 (m, 1H), 4.12-4.36 (m, 2H), 1.09 (s, 3H), 1.04 (s, 3H), 0.49-0.67 (m, 4H), 0.39-0.45 (m, 2H), 0.33 (s, 2H). Preparation of Example 12 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)methacrylamide
Figure imgf000083_0002
To a solution of 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (50 mg, 132.12 µmol) in DCM (0.5 mL) was added TEA (40.11 mg, 396.36 µmol) and methacrylic anhydride (28.52 mg, 184.97 µmol) at 0 °C. The mixture was stirred at 20 °C for 12 h. Methacrylic anhydride (30.55 mg, 198.18 µmol) was added and the mixture was stirred at 20 °C for 12 h. The reaction mixture was concentrated under reduced pressure to give a residue which was purified by preparative HPLC (column: Phenomenex Synergi Polar-RP 100*25 mm*4 µm; mobile phase: A: 0.1% TFA in water, B: MeCN; B%: 42%-62%, 7 min) to give the product N-(1-((1- methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)methacrylamide (4.1 mg, 9.09 µmol, 6.88% yield, 99% purity) as an off-white solid. RT 0.768 min (method 1); m/z 447.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 10.87 (s, 1H), 8.43 (d, J = 2.4 Hz, 1H), 8.27 (s, 1H), 8.13 (dd, J = 8.8, 2.4 Hz, 1H), 7.87 (d, J = 9.2 Hz, 1H), 5.95 (s, 1H), 5.66 (s, 1H), 4.33-4.17 (m, 2H), 1.98-1.91 (m, 3H), 1.06 (s, 3H), 1.04(s, 3H), 0.64-0.51 (m, 4H), 0.44- 0.38 (m, 2H), 0.4-0.30 (m, 2H). Preparation of Intermediate 13.1 (E)-4-(dimethylamino)but-2-enoyl chloride
Figure imgf000084_0001
To a solution of (E)-4-(dimethylamino)but-2-enoic acid (100 mg, 603.80 µmol, HCl salt) in THF (3 mL) was added DMF (4.41 mg, 60.38 µmol, 4.65 µL) and oxalyl chloride (72.81 mg, 573.61 µmol, 50.21 µL,) at 0 °C. The mixture was stirred at 20 °C for 1.5 hr. The mixture was concentrated under vacuum to give (E)-4-(dimethylamino)but-2-enoyl chloride (80 mg, 542.00 µmol, 89.76% yield) as a yellow oil which was directly used for next step without purification. Preparation of Example 13 (E)-4-(dimethylamino)-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)- 2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)but-2-enamide
Figure imgf000084_0002
To a solution of 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (10 mg, 26.42 µmol), TEA (5.35 mg, 52.84 µmol, 7.35 µL) in DCM (0.5 mL) were added (E)-4-(dimethylamino)but-2-enoyl chloride (7.80 mg, 52.84 µmol) at 0 °C and the mixture was stirred at 20 °C for 1 h. Then, additional TEA (25 mg) and (E)-4-(dimethylamino)but- 2-enoyl chloride (30 mg, 203.25 µmol) were added The mixture was stirred at 20 °C for 2 h and concentrated under reduced pressure. The resulting residue was purified by reparative HPLC (column: Phenomenex C1875*30 mm*3 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 6%- 36%, 10 min) to give the product (E)-4-(dimethylamino)-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)but-2-enamide (2.25 mg, 4.60 µmol, 17.39% yield, 100% purity) as a yellow solid. RT 0.718 min (method 1); m/z 490.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 11.13 (br, 1H), 8.41 (d, J = 2.4 Hz, 1H), 8.26 (s, 1H), 8.08-8.17 (m, 1H), 7.85 (d, J = 9.2 Hz, 1H), 6.75-6.84 (m, 1H), 6.43 (d, J = 16.0 Hz, 1H), 4.11-4.34 (m, 2 H), 3.62-3.83 (m, 2 H), 2.55-2.65 (m, 6 H), 1.08 (s, 3 H), 1.03 (s, 3 H), 0.50-0.64 (m, 4 H), 0.37-0.44 (m, 2 H), 0.31 (s, 2 H). Preparation of Example 14 N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)pent-2-ynamide
Figure imgf000085_0001
To a solution of pent-2-ynoic acid (10.37 mg, 105.70 µmol) in THF (2 mL) was added ethyl 2- ethoxyquinoline-1(2H)-carboxylate (26.14 mg, 105.70 µmol). The mixture was stirred at 20 °C for 5 min. Then, 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (20 mg, 52.85 µmol) was added. The mixture was stirred at 20 °C for 16 h and concentrated under reduced pressure. The residue was purified by preparative HPLC (column: Phenomenex C1875*30 mm*3 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 34%-64%, 10 min) to give the product N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)pent-2-ynamide (3.02 mg, 11.33% yield, 100% purity, FA salt) as an off-white solid. RT 0.877 min (method 1); m/z 459.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 8.45 (s, 1H), 8.40 (d, J = 2.4 Hz, 1H), 8.18-8.29 (m, 1H), 8.10 (dd, J = 9.2, 2.0 Hz, 1H), 7.81 (d, J = 9.2 Hz, 1H), 4.14-4.27 (m, 2H), 2.43-2.44 (m, 2H), 1.17 (t, J = 7.2 Hz, 3H), 1.07 (s, 3H), 1.01 (s, 3H), 0.49-0.62 (m, 4H), 0.37-0.44 (m, 2H), 0.26-0.33 (m, 2H). Preparation of Intermediate 15.1 4-methylpent-2-ynoic acid
Figure imgf000085_0002
To a solution of 3-methylbut-1-yne (1 g, 14.68 mmol, 1.50 mL) in THF (10 mL) was added n-BuLi (2.5 M, 5.87 mL, 14.68 µmol) slowly at -78° C. The mixture was stirred at -78 °C for 1 h. Then, dry ice (1 g) was added and the reaction temperature was gently raised to 20 °C. The mixture was stirred at 20 °C for 30 min, then poured into an aqueous ammonium chloride solution (sat., 50 mL) and extracted with EtOAc (50 ml; ×3). The combined organic layer was dried over Na2SO4 and concentrated under vacuum to give residue, which was purified by column chromatography (SiO2, DCM: MeOH = 1: 0 to 10 : 1) to give the product 4-methylpent-2-ynoic acid (300 mg, 2.68 mmol, 18.23% yield) as colorless oil. 1H NMR (DMSO-d6, 400 MHz): δ = 13.34 (br, 1H), 2.71-2.78 (m, 1H), 1.16 (s, 3H), 1.15 (s, 3H) Preparation of Example 15 4-methyl-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)pent-2-ynamide
Figure imgf000086_0001
Two batches as in the following protocol were run in parallel and mixed for the purification: To a solution of 4-methylpent-2-ynoic acid (5.92 mg, 52.84 µmol) in THF (0.5 mL) was added ethyl 2- ethoxyquinoline-1(2H)-carboxylate (13.07 mg, 52.84 µmol). The mixture was stirred at 20 °C for 2 h. Then, 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (10 mg, 26.42 µmol) was added. The mixture was stirred at 20 °C for 16 h and concentrated under reduced pressure to give residue, which was purified by preparative HPLC (column: Phenomenex C1875*30 mm*3 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 37%-67%, 2 min) to give the product 4-methyl-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)pent-2-ynamide (2.09 mg, 4.38 µmol, 8.29% yield, 99% purity) as an off-white solid. RT 0.923 min (method 1); m/z 473.3 (M+H)+ (ESI+); 1H NMR (CDCl3, 400 MHz): δ = 8.71 (d, J = 2.4 Hz, 1H), 8.17 (dd, J = 8.8, 2.4 Hz, 1H), 7.76 (s, 1H), 7.45 (d, J = 9.2 Hz, 1H), 5.02 (s, 1H), 4.22 (d, J = 8.0 Hz, 2H), 2.81-2.71 (m, 1H), 1.29 (s, 3H), 1.27 (s, 6H), 1.12 (s, 3H), 0.80 (d, J = 6.4 Hz, 2H), 0.52- 0.63 (m, 4H), 0.44 (s, 2H). Preparation of Intermediate 16.1 3-cyclopropylpropiolic acid
Figure imgf000087_0001
To a solution of ethynylcyclopropane (500 mg, 7.56 mmol, 627.35 µL) in THF (5 mL) was added n- BuLi (2.5 M, 3.03 mL) slowly at -78 °C and the mixture was stirred at -78 °C for 1 h. Then, dry ice (1 g) was added. and the reaction temperature was gently raised to 20 °C. The mixture was stirred at 20 °C for 30 min, poured into an aqueous ammonium chloride solution (sat., 50 mL) and extracted with EtOAc (50 ml; ×3). The combined organic layer was dried over Na2SO4,filtered and the filtrate was concentrated under vacuum to give a residue, which was purified by column chromatography (SiO2, DCM: MeOH = 1 : 0 to 10 : 1) to give the product 3-cyclopropylpropiolic acid (400 mg, 3.63 mmol, 48.03% yield) as a yellow oil. 1H NMR (DMSO-d6, 400 MHz): δ = 13.20 (s, 1H), 1.46-1.56 (m, 1H), 0.91-0.97 (m, 2H), 0.76-0.82 (m, 2H). Preparation of Example 16 3-cyclopropyl-N-(1-((1-methylcyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo- 1,4-dihydroquinazolin-3(2H)-yl)propiolamide
Figure imgf000087_0002
To a solution of 3-cyclopropylpropiolic acid (5.82 mg, 52.84 µmol) in THF (0.5 mL) was added ethyl 2-ethoxyquinoline-1(2H)-carboxylate (13.07 mg, 52.84 µmol). The mixture was stirred at 20 °C for 30 min. Then, 3-amino-N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl) methyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (10 mg, 26.42 µmol) was added. The mixture was stirred at 20 °C for 16 h, then concentrated under reduced pressure to give a residue, which was purified by preparative HPLC (column: Phenomenex C1875*30 mm*3 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 35%-65%, 10 min) to give the product 3-cyclopropyl-N-(1-((1-methylcyclopropyl)methyl)-6- (N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)propiolamide (1.47 mg, 3.12 µmol, 11.82% yield, 100% purity) as an off-white solid. RT 0.843 min (method 1); m/z 471.1 (M+H)+ (ESI+); 1H NMR (CDCl3, 400 MHz) 8.71 (s, 1H), 8.17 (d, J = 9.2 Hz, 1H), 7.73 (s, 1H), 7.44 (d, J = 8.8 Hz, 1H), 5.05 (s, 1H), 4.15-4.28 (m, 2H), 1.41-1.46 (m, 1H), 1.27 (s, 3H), 1.12 (s, 3H), 0.93-1.03 (m, 4H), 0.80 (d, J = 5.2 Hz, 2 H), 0.51-0.63 (m, 4H), 0.39-0.48 (m, 2H). Preparation of Intermediate 17.1 2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzoic acid
Figure imgf000088_0001
To a solution of 1-methylcyclopropanamine (6.76 g, 62.86 mmol, HCl salt) in DCM (150 mL) at -5 °C was added TEA (15.90 g, 157.15 mmol, 21.87 mL). Then 5-chlorosulfonyl-2-fluoro-benzoic acid (15 g, 62.86 mmol) was added in batches and stirred at -5 °C for 2 h. The mixture was concentrated under reduced pressure to give a residue. The residue was dissolved with saturated aq. NaHCO3 (400 mL) and the solution was washed with EtOAc (200 mL; 2x). The aqueous phase was collected and it was adjusted to pH=6 with aq. HCl (1N). The resulting solution turned to a suspension before it was extracted by EtOAc (200 mL; 2x). The organic phase was collected and washed with brine (200 mL; 2x). The organic phase was collected and dried over anhydrous Na2SO4. The organic phase was filtered and the filtrate was concentrated under reduced pressure to give the product 2-fluoro-5-(N-(1- methylcyclopropyl)sulfamoyl)benzoic acid (11.0 g, 40.25 mmol, 64.03% yield) as a white solid. Preparation of Intermediate 17.2 2-((cyclopropylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzoic acid
Figure imgf000088_0002
To a solution of 2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzoic acid (5.0 g, 18.30 mmol) in MeCN (50 mL) was added DIPEA (3.55 g, 27.44 mmol) and cyclopropylmethanamine (1.43 g, 20.13 mmol). The mixture was stirred at 85 °C for 16 h. The mixture was concentrated under reduced pressure. The residue was diluted with aq. NaHCO3 (400 mL) and extracted with EtOAc (200 mL; 2x). The aqueous phase was adjust to pH<6 with HCl solution (aq., 1 N). The aqueous phase was extracted with EtOAc (200 mL; 2x). The combined organic phases were washed with brine (200 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 2- ((cyclopropylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzoic acid (4.86 g, 14.98 mmol, 81.87% yield) as a white solid. RT 0.407 min (Method 6); m/z 325.0 (M+H)+ (ESI+); 1 HNMR (DMSO-d6, 400 MHz): δ = 13.16 (s, 1H), 8.29 (dd, J = 6.8, 2.4 Hz, 1H), 8.23 (s, 1H), 8.03 (m, 1H), 7.71 (s, 1H), 7.56 (dd, J = 10.4, 9.2 Hz, 1H), 3.13 (d, J = 7.2 Hz, 2H), 1.11-1.08 (m, 1H),1.07 (s, 3H), 0.62-0.58 (m, 4H), 0.43-0.39 (m, 2H), 0.37- 0.32 (m, 2H) Preparation of Intermediate 17.3 2-((cyclopropylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000089_0001
To a solution of 2-((cyclopropylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzoic acid (4.3 g, 13.26 mmol) and DIPEA (8.57 g, 66.28 mmol, 11.54 mL) in DMF (43 mL) was added HATU (6.05 g, 15.91 mmol). The mixture was stirred at 20 °C for 0.5 h. After 0.5 h, NH4Cl (2.13 g, 39.77 mmol) was added and the mixture was stirred at 20 °C for 2 h. The mixture was diluted with water (400 mL) and extracted with EtOAc (200 mL; 2x). The organic phase was washed with brine (200 mL; 2x). The organic phase was dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase flash (ISCO®; 330 g Flash Coulmn Welch Ultimate XB_C1820-40μm; 120 A, Eluent of 8~50% ACN/H2O (0.1% HCl condition) @ 100 mL/min). The ACN of the resulting solution was concentrated under vacuum, and the aqueous layer was adjusted to pH>7 with sat. NaHCO3 solution. The solution was extracted with EtOAc (100 mL; 2x). The organic phase was washed with brine (100 mL; 2x). The organic phase was dried over with anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 2- (cyclopropylmethylamino)-5-[(1-methylcyclopropyl)sulfamoyl]benzamide (1.9 g, 5.87 mmol, 44.28% yield) as a yellow solid. RT 0.490 min (method 4); m/z 324.0 (M+H)+ (ESI+); 1 HNMR (DMSO-d6, 400 MHz): δ = 8.60 (t, J = 4.8 Hz, 1H), 8.05 (s, 1H), 7.98 (d, J = 2.4 Hz, 1H), 7.58 (dd, J = 9.2, 2.4 Hz, 1H), 7.52 (s, 1H) 7.30 (s, 1H), 6.78 (d, J = 8.8 Hz, 1H), 3.05 (dd, J = 6.4, 5.2 Hz, 2H), 1.13-1.07 (m, 1H), 1.05 (s, 3H), 0.63-0.56 (m, 2H), 0.54-0.47 (m, 2H), 0.35-0.30 (m, 2H), 0.28-0.21 (m, 2H). Preparation of Intermediate 17.4 1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000089_0002
To a solution of 2-(cyclopropylmethylamino)-5-[(1-methylcyclopropyl)sulfamoyl]benzamide (1.3 g, 4.02 mmol) in 1-methylpyrrolidin-2-one (15 mL) was added di(1H-imidazol-1-yl)methanone (5.21 g, 32.16 mmol).The mixture was stirred at 130 °C for 3 h. The mixture was partitioned between aq. HCl (1 N, 400 mL) and EtOAc (200 mL; 2x). The aqueous phase was extracted with EtOAc (200 mL; 2x). The organic phase was washed with brine (200 mL; 2x). The organic phase was dried over with anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 1- (cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (1.4 g, 4 mmol, 100% yield) as a yellow solid. RT 0.347 min (Method 6); m/z 350.0 (M+H)+ (ESI+); 1 HNMR (DMSO-d6, 400 MHz): δ = 11.85 (s, 1H), 8.37 (d, J = 2.4 Hz, 1H), 8.17 (s, 1H), 8.05 (dd, J = 8.8, 2.0 Hz, 1H), 7.77 (d, J = 8.8 Hz, 1H), 4.01 (d, J = 6.8 Hz, 2H), 1.27-1.16 (m, 1H), 1.08 (s, 3H), 0.65-0.56 (m, 2H), 0.50-0.42 (m, 4H), 0.42-0.38 (m, 2H). Preparation of Intermediate 17.5 3-amino-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000090_0001
To a solution of 1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (500 mg, 1.43 mmol) in dioxane (5 mL) and DMF (5 mL) was added potassium carbonate (296.66 mg, 2.15 mmol). The mixture was stirred at 70 °C for 0.5 h, then O-(2,4- dinitrophenyl)hydroxylamine (341.93 mg, 1.72 mmol) was added. The mixture was stirred 70 °C for 12 h then partitioned between water (200 mL) and EtOAc (100 mL). The layers were separated and the aqueous phase was extracted with EtOAc (100 mL; 2x). The combined organic phase was washed with brine (100 mL; 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (column: Welch Ultimate XB-CN 250*50*10 µm; mobile phase: A: Hexane, B: EtOH; 10%-50%, 12 min) to give, after solvent concentration, the product 3-amino-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (363 mg, 896.50 µmol, 62.65% yield, 90% purity) as a yellow solid. RT 0.332 min (Method 4); m/z 365.1 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 8.44 (d, J = 2.0 Hz, 1H), 8.21 (s, 1H), 8.06 (dd, J = 8.8, 2.4 Hz, 1H), 7.83 (d, J = 8.8 Hz, 1H), 5.67 (s, 2H), 4.11 (d, J = 7.2 Hz, 2H), 1.29-1.18 (m, 1H), 1.07 (s, 3H), 0.60 (m, 2H), 0.52-0.45 (m, 4H), 0.42-0.37 (m, 2H). Preparation of Example 17 N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)but-2-ynamide
Figure imgf000091_0001
To a solution of but-2-ynoic acid (32.30 mg, 384.17 µmol) in THF (2 mL) was added ethyl 2- ethoxyquinoline-1(2H)-carboxylate (95.00 mg, 384.17 µmol) and the mixture was stirred at 20 °C for 0.5 h. Then, 3-amino-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (70 mg, 192.09 µmol) was added. The reaction mixture was stirred at 20 °C for 16 h and concentrated under reduced pressure. The crude product was purified by preparative HPLC (column: Phenomenex luna C18150*25 mm*10 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 25%-55%, 10 min) and lyophilized directly to give to give the product N-(1- (cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)but- 2-ynamide (15 mg, 34.68 µmol, 18.05% yield, 99.52% purity) as a white solid. RT 0.411 min(Method 6); m/z 430.8 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 11.6 (s, 1H), 8.49-8.41 (d, J = 2.4 Hz, 1H), 8.25 (s, 1H), 8.12 (dd, J = 8.8, 2.0 Hz, 1H), 7.86 (d, J = 8.8 Hz, 1H), 4.09 (d, J = 6.8 Hz, 2H), 2.08 (s, 3H), 1.27-1.20 (m, 1H), 1.08 (s, 3H), 0.63-0.58 (m, 2H), 0.53-0.48 (m, 2H), 0.45-0.44 (m, 2H), 0.43-0.39 (m, 2H). Preparation of Example 18 (E)-N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)but-2-enamide
Figure imgf000091_0002
To a solution of 3-amino-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (50 mg, 137.20 µmol), NaHCO3 (115.26 mg, 1.37 mmol) in THF (1.25 mL) and H2O (1.25 mL) at 0°C was added (E)-but-2-enoyl chloride (43.03 mg, 411.61 µmol). The mixture was stirred at 20 °C for 2 h, quenched with water (50 mL) and extracted with EtOAc (20 mL; 3x). The combined organic phase was washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum. The residue was purified by preparative-TLC (EtOAc: Petroleum ether = 1:1) to give the product (E)-N-(1-(cyclopropylmethyl)-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)but-2-enamide (3.64 mg, 6.01 µmol, 4.4% yield, 96.8% purity) as a white solid. RT 0.480 min (Method 4); m/z 433.1 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6): 10.76 (br, 1H), 8.43 (d, J = 2.0 Hz, 1H), 8.25 (s, 1H), 8.13 (dd, J = 8.8, 2.4 Hz, 1H), 7.87 (d, J = 9.2 Hz, 1H), 6.78- 6.90 (m, 1H), 6.13 (dd, J = 15.2, 1.6 Hz, 1 H), 4.10 (d, J = 6.8 Hz, 2H), 1.89 (dd, J = 6.8, 1.2 Hz, 3H), 1.21-1.27 (m, 1H), 1.09 (s, 3H), 0.57-0.65 (m, 2H), 0.48-0.54 (m, 2H), 0.46 - 0.44 (d, J = 3.2 Hz, 2H), 0.40-0.43 (m, 2H). Preparation of Intermediate 19.1 1-(cyclopropylmethyl)-3-(methylamino)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000092_0001
To a solution of 3-amino-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (30 mg, 82.32 µmol) and paraformaldehyde (14.83 mg, 493.92 µmol,) in i-PrOH (1.5 mL) was added acetic acid (494.35 ug, 8.23 µmol). The mixture was stirred at 20 °C for 2 h, then sodium cyanoborohydride (25.87 mg, 411.60 µmol) was added. The mixture was stirred at 20 °C for 32 h. The reaction mixture was quenched with water (10 mL), then extracted with EtOAc (150 mL; 3x). The combined organic phase was washed with brine (50 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum. The residue was purified by preparative-TLC (SiO2, EtOAc: petroleum ether = 2:1) to give the product 1-(cyclopropylmethyl)-3-(methylamino)-N-(1- methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (18 mg, 4.7 µmol, 5.94% yield, 91% purity) as a white solid. RT 0.467 min (Method 4); m/z 379.1 (M+H)+ (ESI+) Preparation of Example 19 N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)-N-methylacrylamide
Figure imgf000092_0002
To a solution of 1-(cyclopropylmethyl)-3-(methylamino)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (20 mg, 52.85 µmol) in THF (2 mL) and water (2 mL) at 0 °C was added NaHCO3 (44.40 mg, 528.48 µmol) and acryloyl chloride (14.35 mg, 158.54 µmol). Then, the mixture was stirred at 20 °C for 2 h, quenched with water (50 mL) and extracted with EtOAc (20 mL; 3x). The combined organic layer was washed with brine (50 mL), dried with anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum. The residue was purified by preparative TLC (EtOAc: petroleum ether = 1:1) to give the product N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)- 2,4-dioxo-1,2-dihydroquinazolin-3(4H)-yl)-N-methylacrylamide (5.44 mg, 9.99 µmol, 18.9% yield, 96.9% purity) as a white solid. RT 0.501 min (method 4); m/z 433.0 (M+H)+ (ESI+).1H NMR (400 MHz, DMSO-d6): δ = 8.43 (d, J = 2.4 Hz, 1H), 8.28 (s, 1H), 8.09-8.19 (m, 1H), 7.81-7.94 (m, 1H), 6.40-7.01 (m, 1H), 6.18-6.36 (m, 1H), 5.96 - 5.65 (m, 1H), 4.10 (dd, J = 6.8, 2.8 Hz, 2H), 3.17 (s, 3H), 1.26 - 1.22 (m, 1H), 1.09 (s, 3H), 0.57- 0.69 (m, 2H), 0.34-0.56 (m, 6H). Preparation of Example 20 N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)acrylamide
Figure imgf000093_0001
To a solution of NaHCO3 (36.88 mg, 439.06 µmol) in H2O (0.5 mL) was added a solution of 3- amino-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide (40 mg, 109.76 µmol) in THF (0.5 mL). The mixture was stirred at 20 °C for 5 min, then, cooled to -10°C and prop-2-enoyl chloride (29.80 mg, 329.29 µmol) was added dropwise. The mixture was stirred at -10 °C for 0.5 h and quenched with water (20 mL). The aqueous phase was extracted with EtOAc (10 mL; 2x). The combined organic phase was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum. The residue was purified by preparative- HPLC (column: Phenomenex Luna C18150*25 mm*10 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 25%-55%, 10 min) to give an impure fraction which was further purified by preparative- TLC to give the product N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl) sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acrylamide (4 mg, 23.42 µmol, 21.34% yield, 98% purity) as a white solid. RT 0.342 min (Method 4); m/z 419.1 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 10.99 ( s, 1H), 8.43 (d, J = 2.4 Hz, 1H), 8.26 (s, 1H), 8.14 (dd, J = 9.2, 2.4 Hz, 1H), 7.88 (d, J = 8.8 Hz, 1H), 6.50- 6.41 (m, 1H), 6.34-6.25 (m, 1H), 5.91-5.85 (m, 1H), 4.10 (d, J = 6.8 Hz, 2H), 1.27-1.22 (m, 1 H), 1.09 (s, 3H), 0.65-0.59 (m, 2H), 0.52-0.40 (m, 6H). Preparation of Intermediate 21.1 5-(N-(bicyclo[1.1.1]pentan-1-yl)sulfamoyl)-2-fluorobenzamide
Figure imgf000094_0001
To a solution of bicyclo[1.1.1]pentan-1-amine (192.40 mg, 1.61 mmol, HCl) in DCM (10 mL) cooled to 0 °C was added TEA (234.20 mg, 2.31 mmol, 322.14 µL). The solution was stirred for 0.5 h and a 3- carbamoyl-4-fluorobenzenesulfonyl chloride (550 mg, 2.31 mmol) in DCM (10 mL) was added dropwise very slowly keeping the temperature at 0 °C. Then, the reaction mixture was stirred at 20 °C for 1 h and pyridine (366.15 mg, 4.63 mmol, 373.62 µL) was added. The reaction mixture was adjusted to pH=6 with HCl (1 N), then extracted with DCM (50 mL; 2x). The combined organic layer was dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum to give the product 5-(N- (bicyclo[1.1.1]pentan-1-yl)sulfamoyl)-2-fluorobenzamide (550 mg, 1.93 mmol, 83.58% yield) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz): δ = 8.72 (s, 1H), 8.08 (dd, J = 6.8, 2.4 Hz, 1H), 7.82-7.97 (m, 3H), 7.52 (dd, J = 10.0, 8.8 Hz, 1H), 2.29 (s, 1H), 1.72 (s, 6H). Preparation of Intermediate 21.2 5-(N-(bicyclo[1.1.1]pentan-1-yl)sulfamoyl)-2-((cyclopropylmethyl)amino)benzamide
Figure imgf000094_0002
To a solution of 5-(N-(bicyclo[1.1.1]pentan-1-yl)sulfamoyl)-2-fluorobenzamide (520 mg, 1.83 mmol) in MeCN (5 mL) was added DIPEA (709.16 mg, 5.49 mmol, 955.74 µL) and cyclopropylmethanamine (260.16 mg, 3.66 mmol). The mixture was stirred at 85 °C for 12 hand concentrated under reduced pressure. The resulting residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0~40% EtOAc /Petroleum ether gradient @ 60 mL/min) to give the product 5-(N-(bicyclo[1.1.1]pentan-1-yl)sulfamoyl)-2-((cyclopropylmethyl)amino)benzamide (340 mg, 1.01 mmol, 55.42% yield) as a white solid. 1H NMR (DMSO-d6, 400 MHz): δ = 8.64 (s, 1H), 8.01-8.20 (m, 2H), 7.98 (m, 1H), 7.60 (d, J = 8.0 Hz, 1H), 7.13-7.48 (m, 1H), 6.78 (d, J = 8.8 Hz, 1H), 3.05 (t, J = 5.6 Hz, 2H), 2.24 (s, 1H), 1.68 (s, 6H), 1.09 (s, 1H), 0.51 (d, J = 7.2 Hz, 2H), 0.25 (d, J = 4.4 Hz, 2H). Preparation of Intermediate 21.3 N-(bicyclo[1.1.1]pentan-1-yl)-1-(cyclopropylmethyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000095_0001
To a solution of 5-(N-(bicyclo[1.1.1]pentan-1-yl)sulfamoyl)-2-((cyclopropylmethyl)amino)benzamide (340 mg, 1.01 mmol) in 1-methylpyrrolidin-2-one (4.5 mL) was added di(1H-imidazol-1-yl)methanone (1.31 g, 8.11 mmol). The mixture was stirred at 130 °C for 2 hr, then, cooled to room temperature, diluted with H2O (30 mL) and extracted with EtOAc (20 mL; 2x). The combined organic phase was washed with brine (30 mL; 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum. The resulting residue, was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0~50% EtOAc /Petroleum ether gradient @ 50 mL/min) to give the product N- (bicyclo[1.1.1]pentan-1-yl)-1-(cyclopropylmethyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (300 mg, 830.07 µmol, 81.89% yield) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz): δ = 11.86 (s, 1H), 8.73 (s, 1H), 8.38 (d, J = 2.4 Hz, 1H), 8.07 (dd, J = 8.8, 2.4 Hz, 1H), 7.76 (d, J = 8.8 Hz, 1H), 3.97-4.06 (m, 2H), 2.29 (s, 1H), 1.72 (s, 6H), 1.20-1.24 (m, 1H), 0.41-0.51 (m, 4H). Preparation of Intermediate 21.4 N-(bicyclo[1.1.1]pentan-1-yl)-1-(cyclopropylmethyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000095_0002
To a solution of N-(bicyclo[1.1.1]pentan-1-yl)-1-(cyclopropylmethyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (300 mg, 830.07 µmol) in dioxane (2 mL) and DMF (2 mL) was added potassium carbonate (229.45 mg, 1.66 mmol). The mixture was stirred at 70 °C for 0.5 h followed by addition of O-(2, 4-dinitrophenyl)hydroxylamine (214.87 mg, 1.08 mmol). The mixture was stirred at 70 °C for 12 h, cooled to room temperature, diluted with H2O (50 mL) and extracted with EtOAc (50 mL; 2x). The combined organic phase was washed with brine (50 mL; 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated. The crude was purified by preparative TLC (Petroleum ether : EtOAc = 1 : 1) to give the product 3-amino-N-(bicyclo[1.1.1]pentan-1-yl)-1-(cyclopropylmethyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (80 mg, 212.52 µmol, 25.60% yield) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz): δ = 8.76 (s, 1H), 8.45 (d, J = 2.4 Hz, 1H), 8.08 (dd, J = 8.8, 2.4 Hz, 1H), 7.83 (d, J = 9.2 Hz, 1H), 5.67 (br, 2H), 4.12 (d, J = 6.8 Hz, 2H), 2.4 (s, 1H), 1.72 (s, 6H), 1.21-1.28 (m, 1H), 0.44-0.55 (m, 4H). Preparation of Example 21 N-(6-(N-(bicyclo[1.1.1]pentan-1-yl)sulfamoyl)-1-(cyclopropylmethyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acrylamide
Figure imgf000096_0001
To a solution of 3-amino-N-(bicyclo[1.1.1]pentan-1-yl)-1-(cyclopropylmethyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (64 mg, 170.02 µmol) in THF (1 mL) was added TEA (51.61 mg, 510.06 µmol, 70.99 µL). The mixture was stirred at 0 °C for 10 min. Then, acrylic anhydride (107.21 mg, 850.09 µmol) in THF (1 mL) was added dropwise at 0 °C. The mixture was stirred at 0 °C for 2 hr, then concentrated under reduced pressure to give a residue, which was purified by preparative-HPLC (column: Waters Xbridge 150*25 mm* 5 µm; mobile phase: A: 1 mM aqueous solution of NH4HCO3, B: MeCN; B%: 25%-55%, 9 min) to give the product N-(6-(N-(bicyclo[1.1.1]pentan-1-yl)sulfamoyl)-1-(cyclopropylmethyl)- 2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)acrylamide (1.83 mg, 4.17 µmol, 2.45% yield, 98% purity) as a light yellow solid. RT 0.817 min (method 1); m/z 431.0 (M+H)+ (ESI+); 1H NMR (CDCl3, 400 MHz): δ = 8.75 (d, J = 2.4 Hz, 1H), 8.20 (dd, J = 8.8, 2.4 Hz, 1H), 7.89 (s, 1H), 7.48 (d, J = 8.8 Hz, 1H), 6.51-6.61 (m, 1H), 6.32- 6.45 (m, 1H), 5.94 (d, J = 10.0 Hz, 1H), 5.57 (s, 1 H), 4.04-4.18 (m, 2H), 2.39 (s, 1H) 1.85-1.99 (m, 6H), 0.85-0.94 (m, 1H), 0.49-0.69 (m, 4 H). Preparation of Intermediate 22.1 2-fluoro-5-(N-(1-(fluoromethyl)cyclopropyl)sulfamoyl)benzamide
Figure imgf000097_0001
To a solution of 1-(fluoromethyl)cyclopropan-1-amine (200 mg, 1.59 mmol, HCl) in DCM (4 mL), cooled to 0 °C, was added pyridine (343.59 mg, 4.34 mmol, 350.60 µL) and the solution was stirred for 0.5 h. Then, 3-carbamoyl-4-fluorobenzenesulfonyl chloride (344.08 mg, 1.45 mmol)was added and the solution was stirred at 20 °C for 1h. The reaction mixture was adjusted to pH = 6 with HCl (1 M), then extracted with DCM (50 mL; 2x). The combined organic phase was washed with brine (30 mL; 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum to give the product 2- fluoro-5-(N-(1-(fluoromethyl)cyclopropyl)sulfamoyl)benzamide (240 mg, 826.77 µmol, 57.10% yield) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz): δ = 8.57 (s, 1H), 8.05 (dd, J = 6.4, 2.4 Hz, 1H), 7.88-7.94 (m, 2H), 7.85 (s, 1H), 7.52 (dd, J = 10.0, 8.8 Hz, 1H), 4.20 (d, J = 48 Hz, 2H), 0.70-0.77 (m, 2H), 0.66-0.71 (m, 2H). Preparation of Intermediate 22.2 2-((cyclopropylmethyl)amino)-5-(N-(1-(fluoromethyl)cyclopropyl)sulfamoyl)benzamide
Figure imgf000097_0002
To a solution of 2-fluoro-5-(N-(1-(fluoromethyl)cyclopropyl)sulfamoyl)benzamide (240 mg, 826.77 µmol) in MeCN (2.5 mL) was added DIPEA (320.56 mg, 2.48 mmol, 432.02 µL) and cyclopropylmethanamine (117.60 mg, 1.65 mmol). The mixture was stirred at 85 °C for 12 h, then cooled to room temperature, diluted with H2O (30 mL) and extracted with EtOAc (20 mL; 2x). The combined organic phase was washed with brine (30 mL; 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum to give the product 2-((cyclopropylmethyl)amino)-5-(N-(1- (fluoromethyl)cyclopropyl)sulfamoyl)benzamide (220 mg, 644.41 µmol, 77.94% yield) as a yellow solid 1H NMR (DMSO-d6, 400 MHz): δ = 8.63 (t, J = 5.2 Hz, 1H), 8.00-8.15 (m, 1H), 7.97 (d, J = 2.0 Hz, 1H), 7.92 (s, 1H), 7.57 (dd, J = 8.8, 2.0 Hz, 1H), 6.78 (d, J = 9.2 Hz, 1H), 4.09-4.33 (m, 2H), 3.05 (dd, J = 6.8, 5.2 Hz, 2H), 1.00-1.11 (m, 1H), 0.65 (s, 4H), 0.43-0.54 (m,2 H), 0.20-0.30 (m, 2H). Preparation of Intermediate 22.3 N-(bicyclo[1.1.1]pentan-1-yl)-1-(cyclopropylmethyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000098_0001
To a solution of 2-(cyclopropylmethylamino)-5-[[1 -(fluoromethyl)cyclopropyl]sulfamoyl]benzamide (220 mg, 644.41 pmol) and DIPEA (832.83 mg, 6.44 mmol, 1.12 mL) in DMF (3 mL) was added di(1 H- imidazol-1-yl)methanone (1.04 g, 6.44 mmol). The mixture was stirred at 140 °C for 2 h and cooled to room temperature. The reaction mixture was justed to pH = 6 with HCI (1 N) and extracted with EtOAc (30 mL; 2x). The combined organic layer was washed with brine (30 mL; 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated in vacuum to give the product 1 -(cyclopropylmethyl)-N- [1-(fluoromethyl)cyclopropyl]-2,4-dioxo-quinazoline-6-sulfonamide (200 mg, 544.37 pmol, 84.48% yield) as a yellow solid.
1H NMR (DMSO-cfe, 400 MHz): 5 = 11 .85 (s, 1 H), 8.56 (s, 1 H), 8.36 (d, J = 2.4 Hz, 1 H), 8.04 (dd, J = 8.8, 2.4 Hz, 1 H), 7.76 (d, J = 8.8 Hz, 1 H), 4.10-4.33 (m, 2H), 4.01 (d, J = 6.8 Hz, 2H), 1.20-1.25 (m, 1 H), 0.65-0.78 (m, 4H), 0.38-0.56 (m, 4H).
Preparation of Intermediate 22.4
3-amino-1-(cyclopropylmethyl)-N-(1-(fluoromethyl)cyclopropyl)-2, 4-dioxo-1 , 2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000098_0002
To a solution of 1-(cyclopropylmethyl)-N-(1-(fluoromethyl)cyclopropyl)-2,4-dioxo-1, 2,3,4- tetrahydroquinazoline-6-sulfonamide (200 mg, 544.37 pmol) in dioxane (1.5 mL) and DMF (1.5 mL) was added potassium carbonate (150.48 mg, 1 .09 mmol). The mixture was stirred at 70 °C for 0.5 h followed by addition of O-(4-nitrophenyl)hydroxylamine (109.07 mg, 707.69 pmol). The mixture was stirred at 70 °C for 12 h, cooled to room temperature, diluted with H2O (20 mL) and extracted with EtOAc (20 mL; 2x). The combined organic layer was washed with brine (20 mL; 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated. The resulting residue was purified by flash silica gel chromatography (ISCO®; 4 g SepaFlash® Silica Flash Column, Eluent of 0-60% EtOAc /Petroleum ether gradient @ 40 mL/min) to give the product 3-amino-1-(cyclopropylmethyl)-N-(1-(fluoromethyl)cyclopropyl)-2,4-dioxo- 1 ,2,3,4-tetrahydroquinazoline-6-sulfonamide (100 mg, 261.50 pmol, 48.04% yield) as a yellow solid. 1H NMR (DMSO-cfe, 400 MHz): 5 = 8.60 (s, 1 H), 8.43 (d, J = 2.4 Hz, 1 H), 8.05 (dd, J = 8.8, 2.4 Hz, 1 H), 7.82 (d, J=9.2 Hz, 1 H), 5.68 (s, 2H), 4.13-4.27 (m, 2H), 4.11 (d, J = 6.8 Hz, 2H), 1.21-1.29 (m, 1 H), 0.65-0.76 (m, 4H), 0.44-0.53 (m, 4H).
Preparation of Example 22
N-(1-(cyclopropylmethyl)-6-(N-(1-(fluoromethyl)cyclopropyl)sulfamoyl)-2,4-dioxo-1 ,4- dihydroquinazolin-3(2H)-yl)acrylamide
Figure imgf000099_0001
Two batches as in the following protocol were run in parallel and mixed for the purification: To a solution of 3-amino-1-(cyclopropylmethyl)-N-(1 -(fluoromethyl)cyclopropyl)-2,4-dioxo-1 , 2,3,4- tetrahydroquinazoline-6-sulfonamide (20 mg, 52.30 pmol) in THF (0.3 mL) were added dropwise at 0 °C TEA (15.88 mg, 156.90 pmol, 21.84 μL) and acrylic anhydride (32.98 mg, 261.50 pmol) in THF (0.3 mL) . The mixture was stirred at 20 °C for 1 h, then with H2O (20 mL), extracted with EtOAc (20mL; 2x). The combined organic layer was washed with brine (20 mL; 2x), dried over Na2SO4 and concentrated. The resulting residue was purified by preparative TLC (Petroleum ether: EtOAc = 1 : 1) to give an impure product which was further purified by preparative HPLC (column: Waters Xbridge 150*50 mm*10 pm; mobile phase: A: 1 mM aqueous solution of NH4HCO3, B: MeCN; B%: 25%-55%, 8 min) to give the product N-(1-(cyclopropylmethyl)-6-(N-(1 -(fluoromethyl)cyclopropyl)sulfamoyl)-2,4-dioxo-1 ,4-dihydroquinazolin- 3(2H)-yl)acrylamide (2.94 mg, 6.74 pmol, 6.44% yield, 100% purity) as an off-white solid.
RT 0.801 min (method 1); m/z 437.1 (M+H)+ (ESI*); 1H NMR (CDCI3, 400 MHz): 5 = 8.75 (d, J = 2.4 Hz, 1 H), 8.19 (dd, J = 8.8, 2.4 Hz, 1 H), 7.79 (s, 1 H), 7.49 (d, J = 8.8 Hz, 1 H), 6.51-6.59 (m, 1 H), 6.31- 6.44 (m, 1 H), 5.94 (d, J = 10.4 Hz, 1 H), 5.49 (s, 1 H), 4.06-4.33 (m, 4H), 1.16-1 .26 (m, 1 H), 0.94-1 .06 (m, 2H), 0.74-0.86 (m, 2H), 0.50-0.71 (m, 4H).
General procedure 1 :
Step 1 : To a solution of methyl 3-chlorocyclobutanecarboxylate (1 eq) in THF (100 mg/mL), which was degassed and purged with N2 (3x), was added drop-wise LiHMDS in THF (1 M, 1 .0 -1 .5 eq) at 0 °C. The solution was stirred at 0 °C for 0.5 to 3 h under a N2 atmosphere leading to a solution of methyl bicyclo [1 .1 .0] butane- 1 -carboxylate.
Step 2: Then, this newly prepared methyl bicyclo[1.1.0]butane-1 -carboxylate solution (1.0-1.5 eq) was added to a solution of the hydrazide compound (1eq) in THF (100 mg/L) previously cooled to 0°C and LiHMDS in THF (1 M, 2.0-3.5 eq) was added at 0 °C The resulting mixture was heated to 20-40 °C and stirred for additional 1~3 h. The reaction mixture was quenched with NH4Cl (aq., sat.) and extracted with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue which used for further purification. Preparation of Example 23 using General procedure 1 N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000100_0001
To a solution of methyl 3-chlorocyclobutanecarboxylate (200 mg, 1.35 mmol) in THF (2 mL) was added LiHMDS in THF (1 M, 1.62 mmol, 1.62 mL) drop-wise at 0 °C under N2 and the solution was stirred at 0 °C for 3 h. Then, this solution of methyl bicyclo[1.1.0]butane-1-carboxylate in THF (123 µL , 164.65 µmol , 1.34 mol/L) was added to a solution of 3-amino-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)- 2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (50 mg, 137.20 µmol) in THF (0.5 mL) at 0 °C and LiHMDS in THF (1 M, 480.22 µL) was added drop-wise at 0 °C. The resulting mixture was stirred at 20 °C for 2 h then heated at 40 °C and stirred for another 1 h. The mixture was poured into NH4Cl (aq., sat., 10 mL). The aqueous layer was extracted with EtOAc (3 mL, 3x). The combined organic layer was washed with brine (10 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum. The resulting residue was purified by preparative HPLC (column: Phenomenex luna C18150*25 mm*10 µm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 25%-55%, 10 min) and lyophilized directly to give the product N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)- 2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide (4.99 mg, 10.58 µmol, 7.71% yield, 94.27% purity) as a yellow solid. RT 0.443 min (method 7); m/z 445.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz,): 10.56 (s, 1H), 8.43 (d, J = 2.0 Hz, 1H), 8.24 (s, 1H), 8.13 (dd, J = 2.0, 8.8 Hz, 1H), 7.86 (d, J = 8.8 Hz, 1H), 4.17 - 4.02 (m, 2H), 2.40 (d, J = 3.2 Hz, 2H), 2.27 - 2.23 (m, 1H), 1.28 - 1.20 (m, 1H), 1.14 (d, J = 2.0 Hz, 2H), 1.09 (s, 3H), 0.64 - 0.59 (m, 2H), 0.54 - 0.39 (m, 6H) Preparation of Intermediate 24.1 2-(((2,2-difluorocyclopropyl)methyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000101_0001
To a solution of 2-fluoro-5-((1-methylcyclopropyl)sulfamoyl)benzamide (400 mg, 1.47 mmol) in DMF (4 mL) was added K2CO3 (406.06 mg, 2.94 mmol) and (2,2-difluorocyclopropyl)methanamine (231.98 mg, 1.62 mmol, HCl salt). The mixture was stirred at 80°C for 14 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The crude product was purified by reversed-phase flash (ISCO®; 48 g Flash Column Welch Ultimate XB_C1820-40μm; 120 A, Eluent of 5~95% ACN/H2O (0.1% FA condition) @ 80 mL/min). The desired fraction was lyophilized to give the product 2-(((2,2-difluorocyclopropyl)methyl)amino)-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (200 mg, 556.50 µmol, 37.88% yield) as a white solid. RT 0.476 min (Method 7); m/z 359.8 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6) 8.65 (t, J = 5.2 Hz, 1H), 8.15 - 8.04 (m, 1H), 8.00 (d, J = 2.4 Hz, 1H), 7.61 (dd, J = 8.8, 2.0 Hz, 1H), 7.58 (s, 1H), 7.43 - 7.31 (m, 1H), 6.88 (d, J = 9.2 Hz, 1H), 3.41 - 3.37 (m, 2H), 2.14 - 2.00 (m, 1H), 1.65 - 1.61 (m, 1H), 1.43 - 1.32 (m, 1H), 1.05 (s, 3H), 0.63 - 0.56 (m, 2H), 0.37 - 0.30 (m, 2H) Preparation of Intermediate 24.2 1-((2,2-difluorocyclopropyl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000101_0002
To a solution of 2-(((2,2-difluorocyclopropyl)methyl)amino)-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (190 mg, 528.67 µmol) in NMP (3 mL) was added CDI (685.79 mg, 4.23 mmol) and DIEA (546.62 mg, 4.23 mmol, 736.68 µL) .The mixture was stirred at 140 °C for 2 h. The mixture was poured into water (30 mL). The aqueous layer was extracted with EtOAc (15 mL, 3x). The combined organic layer was washed with HCl aqueous solution (20 mL, 0.5 N) and brine (20 mL, 2x), dried with anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum to give the product 1-((2,2-difluorocyclopropyl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide (200 mg, 518.96 µmol, 98.16% yield) as a brown solid. RT 0.37 min (Method 7); m/z 386.2 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6) 11.92 (s, 1H), 8.37 (d, J = 2.4 Hz, 1H), 8.19 (s, 1H), 8.05 (dd, J = 9.2, 2.4 Hz, 1H), 7.75 (d, J = 8.8 Hz, 1H), 4.26 (d, J = 6.8 Hz, 2H), 2.26 - 2.14 (m, 1H), 1.71 - 1.57 (m, 1H), 1.55 - 1.43 (m, 1H), 1.08 (s, 3H), 0.63 - 0.57 (m, 2H), 0.43 - 0.37 (m, 2H) Preparation of Intermediate 24.3 3-amino-1-((2,2-difluorocyclopropyl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000102_0001
To a solution of 1-((2,2-difluorocyclopropyl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (190 mg, 493.01 µmol) in DMF (2 mL) and dioxane (2 mL) was added K2CO3 (136.28 mg, 986.03 µmol). The mixture was stirred at 70 °C for 0.5 h. O-(2,4- dinitrophenyl)hydroxylamine (147.25 mg, 739.52 µmol) was added at 70 °C. The resulting mixture was stirred at 70 °C for 13.5 h. The mixture was poured into water (30 mL). The aqueous layer was extracted with EtOAc (15 mL, 3x). The combined organic layer was washed with brine (20 mL, 3x), dried with anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum to give the product 3-amino- 1-((2,2-difluorocyclopropyl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide (197 mg, 492.01 µmol, 99.80% yield) as a yellow oil. RT 0.36 min (Method 7); m/z 401.2 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6) 8.44 (d, J = 2.4 Hz, 1H), 8.22 (s, 1H), 8.06 (dd, J = 9.2, 2.4 Hz, 1H), 7.81 (d, J = 8.8 Hz, 1 H), 5.67 (s, 2H), 4.43 - 4.29 (m, 2H), 2.26 - 2.13 (m, 1H), 1.71 - 1.61 (m, 1H), 1.56 - 1.46 (m, 1H), 1.08 (s, 3H), 0.63 - 0.57 (m, 2H), 0.43 - 0.37 (m, 2H) Preparation of Example 24 N-(1-((2,2-difluorocyclopropyl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000103_0001
The reaction was conducted according to the general procedure 1 starting from 3-amino-1-((2,2- difluorocyclopropyl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide. The residue was purified by preparative-HPLC (column: Waters Xbridge 150*25 mm*5 µm; mobile phase: A: 10 mM aqueous solution of NH4HCO3 in water, B: MeCN; B%: 25%-55%, 8 min) and lyophilized directly to give the product N-(1-((2,2-difluorocyclopropyl)methyl)-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1- carboxamide (7.85 mg, 15.83 µmol, 12.67% yield, 96.87% purity) as a white solid. RT 0.384 min (Method 7); m/z 481.3 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6) 10.63 (d, J = 11.6 Hz, 1H), 8.43 (s, 1H), 8.28 (s, 1H), 8.13 (d, J = 8.8 Hz, 1H), 7.87 (d, J = 9.2 Hz, 1H), 4.55 - 4.10 (m, 2H), 2.40 (d, J = 1.6 Hz, 2H), 2.30 - 2.16 (m, 2H), 1.73 - 1.62 (m, 1H), 1.54 - 1.40 (m, 1H), 1.25 - 1.12 (m, 2H), 1.09 (s, 3H), 0.67 - 0.55 (m, 2H), 0.48 - 0.36 (m, 2H) Preparation of Example 25 N-(1-(cyclopropylmethyl)-6-(N-(1-(fluoromethyl)cyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000103_0002
The reaction was conducted according to the general procedure 1 starting from 3-amino-1- (cyclopropylmethyl)-N-(1-(fluoromethyl)cyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide. The residue was purified by preparative-HPLC (column: Waters Xbridge 150*25 mm*5 µm; mobile phase: A: 10 mM aqueous solution of NH4HCO3 in water, B: MeCN; B%: 15%-45%, 8 min) and lyophilized directly to give the product N-(1-(cyclopropylmethyl)-6-(N-(1- (fluoromethyl)cyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1- carboxamide (4.43 mg, 9.35 µmol, 8.94% yield, 97.59% purity) as a white solid. RT 0.364 min (Method 7); m/z 463.4 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6) 10.59 (br, 1 H), 8.65 (s, 1 H), 8.42 (d, J = 2.4 Hz, 1 H), 8.12 (dd, J = 8.8, 2.4 Hz, 1H), 7.86 (d, J = 9.2 Hz, 1H), 4.26 (d, J = 48.4 Hz, 2H), 4.13 - 4.02 (m, 2H), 2.40 (d, J = 3.6 Hz, 2H), 2.23 - 2.29 (m, 1H), 1.28 - 1.23 (m, 1H), 1.13 (d, J = 2.0 Hz, 2H), 0.83 - 0.58 (m, 4H), 0.57 - 0.48 (m, 2H), 0.39 - 0.48 (m, 2H). Preparation of Intermediate 26.1 2-fluoro-4-methyl-benzamide
Figure imgf000104_0001
To a solution of 2-fluoro-4-methyl-benzoic acid (3 g, 19.46 mmol) in THF (30 mL) was added CDI (3.79 g, 23.36 mmol) and DIEA (3.77 g, 29.19 mmol, 5.09 mL,). The mixture was stirred at 20 °C for 2 h. After 1 h, NH3.H2O (10.92 g, 87.25 mmol, 12.00 mL, 28% purity) was added at 0 °C and the mixture was stirred at 20 °C for 0.5 h. The reaction mixture was diluted with H2O (250 mL) and extracted with EtOAc (100 mL, 3x). The organic phase was washed with brine (100 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give 2-fluoro-4-methyl-benzamide (1.9 g, 12.41 mmol, 63.74% yield) as a white solid. RT 0.295 min (Method 1); m/z 153.9 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 7.68-7.44 (m, 3H), 7.20-7.00 (m, 2H), 2.34 (s, 3H) Preparation of Intermediate 26.2 5-carbamoyl-4-fluoro-2-methyl-benzenesulfonyl chloride
Figure imgf000104_0002
2-fluoro-4-methyl-benzamide (1.9 g, 12.41 mmol) was added to HSO3Cl (8.67 g, 74.44 mmol, 4.96 mL). and the mixture was stirred at 140 °C for 2 h. The mixture was poured into ice-water (500 mL), and extracted with EtOAc (200 mL, 3x). The organic layer was washed with brine (200 mL, 2x), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure. The resulting residue was purified by column chromatography (SiO2: ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0~50% EtOAc/Petroleum ether gradient @ 70 mL/min 30 min) to give the product 5-carbamoyl-4-fluoro-2-methyl- benzenesulfonyl chloride (2 g, 7.95 mmol, 64.06% yield) as a brown solid. RT 0.400 min (Method 7); m/z 251.8 (M+H)+ (ESI+);1H NMR (DMSO-d6, 400 MHz): 8.04 (d, J = 8.0 Hz, 1H), 7.65-7.42 (m, 2H), 7.06 (d, J = 12 Hz, 1H), 2.53 (s, 3H) Preparation of Intermediate 26.3 2-fluoro-4-methyl-5-[(1-methylcyclopropyl)sulfamoyl]benzamide
Figure imgf000105_0001
To a solution of 5-carbamoyl-4-fluoro-2-methylbenzenesulfonyl chloride (2 g, 7.95 mmol) in THF (20 mL) and H2O (15 mL) at 0°C was added NaHCO3 (6.01 g, 71.52 mmol, 2.78 mL) and 1- methylcyclopropanamine (1.71 g, 15.89 mmol, HCl salt). The mixture was stirred at 0 °C for 3 h. The reaction mixture was diluted with H2O (300 mL) and extracted with EtOAc (150 mL, 3x). Then, the combined organic layer was washed with brine (150 mL, 2x), dried over anhydrous Na2SO4 filtered and the filtrate was concentrated under reduced pressure to give the product 2-fluoro-4-methyl-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (1.77 g, 6.18 mmol, 77.79% yield) as a white solid. RT 0.373 min (Method 7); m/z 287.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.23 (s, 1H), 8.19 (d, J = 7.2 Hz, 1H), 7.76 (br d, J = 7.2 Hz, 2H), 7.38 (d, J = 11.6 Hz, 1H), 2.56 (s, 3H), 1.09 (s, 3H), 0.59-0.53 (m, 2H), 0.41-0.35 (m, 2H) Preparation of Intermediate 26.4 2-(cyclopropylmethylamino)-4-methyl-5-[(1-methylcyclopropyl)sulfamoyl]benzamide
Figure imgf000105_0002
To a solution of 2-fluoro-4-methyl-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (1.77 g, 6.18 mmol) in MeCN (25 mL) was added K2CO3 (1.71 g, 12.36 mmol) and cyclopropylmethanamine (879.31 mg, 12.36 mmol). The mixture was stirred at 100°C for 8 h. The reaction mixture was diluted with H2O (100 mL) and extracted with EtOAc (30 mL, 2x). The combined organic layer was washed with brine (30 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 2-((cyclopropylmethyl)amino)-4-methyl-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (1.8 g, 5.33 mmol, 86.29% yield) as a white solid. RT 0.421 min (Method 7); m/z 338.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.57 (s, 1H), 8.04 (s, 1H), 7.64 (s, 1H), 7.20 (s, 1H), 6.58 (s, 1H), 3.10-2.98 (m, 2H), 2.44 (s, 3H), 1.12-1.08 (m, 1H), 1.07 (s, 3H), 0.59-0.54 (m, 2H), 0.53- 0.47 (m, 2H), 0.33-0.29 (m, 2H), 0.26-0.21 (m, 2H) Preparation of Intermediate 26.5 1-(cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-quinazoline-6-sulfonamide
Figure imgf000106_0001
To a solution of 2-((cyclopropylmethyl)amino)-4-methyl-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (700 mg, 2.07 mmol) in DMF (15 mL) was added CDI (2.69 g, 16.60 mmol) and DIEA (2.14 g, 16.60 mmol, 2.89 mL). The mixture was stirred at 100 °C for 2 h. The resulting mixture was diluted with H2O (200 mL) and extracted with EtOAc (50 mL, 3x). The organic layers were washed with HCl solution (0.5 N, 50 mL, 2x), combined, washed with brine (50 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate concentrated under reduced pressure to give the product 1- (cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide (750 mg, 2.06 mmol, 99.48% yield) as a pink solid. RT 0.471 min (Method 7); m/z 364.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 11.76 (s, 1H), 8.43 (s, 1H), 8.23 (s, 1H), 7.57 (s, 1H), 4.00 (d, J = 7.2 Hz, 2H), 2.66 (s, 3H), 1.25-1.21 (m, 1H), 1.11 (s, 3H), 0.60-0.54 (m, 2H), 0.51-0.42 (m, 4H), 0.42-0.35 (m, 2H) Preparation of Intermediate 26.6 3-amino-1-(cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-quinazoline-6- sulfonamide
Figure imgf000106_0002
To a solution of 1-(cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (350 mg, 963.05 µmol) in DMF (3 mL) and dioxane (3 mL) was added K2CO3 (266.20 mg, 1.93 mmol). The mixture was stirred at 70°C for 0.5 h. O-(2,4- dinitrophenyl)hydroxylamine (383.52 mg, 1.93 mmol) was added and the reaction mixture stirred at 70 °C for 16 h. The reaction mixture was diluted with H2O (100 mL) and extracted with EtOAc (30 mL, 3x). The combined organic layer was washed with brine (30 mL, 5x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to the product 3-amino-1-(cyclopropylmethyl)- 7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (310 mg, 819.14 µmol, 85.06% yield) as a brown solid. RT 0.374 min (Method 7); m/z 379.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.50 (s, 1H), 8.27 (s, 1H), 7.63 (s, 1H), 5.64 (s, 2H), 4.11 (d, J = 6.8 Hz, 2H), 2.68 (s, 3H), 1.29-1.25(m, 1H), 1.11 (s, 3H), 0.59-0.54 (m, 2H), 0.50-0.46 (m, 4H), 0.41-0.37 (m, 2H) Preparation of Example 26 N-(1-(cyclopropylmethyl)-7-methyl-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000107_0001
The reaction was conducted according to the general procedure 1 starting from 3-amino-1- (cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-quinazoline-6-sulfonamide. The crude product was purified by preparative-HPLC (column: Phenomenex C18150*25 mm*10 µm; mobile phase: A: 0.05% NH3·H2O in water; B: MeCN; B%: 22%-52%, 8 min). The product solution was lyophilized to give the product N-(1-(cyclopropylmethyl)-7-methyl-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide (13.82 mg, 29.84 µmol, 3.76% yield, 99.012% purity) as a white solid. RT 0.383 min (Method 7); m/z 459.4 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz):10.53 (br, 1H), 8.48 (s, 1H), 8.32 (s, 1H), 7.68 (s, 1H), 4.16-4.06 (m, 2H), 2.69 (s, 3H), 2.39 (d, J = 2.8 Hz, 2H), 2.33 (s, 1H), 1.31-1.23 (m, 1H), 1.14-1.12 (m, 2H), 1.12 (s, 3H), 0.67-0.66 (m, 1H) ,0.66-0.44 (m, 5H), 0.42-0.38 (m, 2H) Preparation of Intermediate 27.1 2,4-dimethylthiazole-5-carbaldehyde
Figure imgf000107_0002
To a solution of (2,4-dimethylthiazol-5-yl)methanol (4.5 g, 31.42 mmol) in DCE (50 mL) was added MnO2 (27.32 g, 314.23 mmol). The mixture was stirred at 80 °C for 1.5 h. The mixture was filtered and the filtrate was concentrated under reduced pressure to give the product 2,4-dimethylthiazole-5- carbaldehyde (3.57 g, 25.28 mmol, 80.47% yield) as yellow oil. RT 0.234 min (Method 7); m/z 142.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 10.03 (s, 1H), 2.69 (s, 3H), 2.64 (s, 3H). Preparation of Intermediate 27.2 trans&cis-2,4-dimethylthiazole-5-carbaldehyde oxime
Figure imgf000108_0002
To a mixture of 2,4-dimethylthiazole-5-carbaldehyde (3 g, 21.25 mmol) in EtOH (25 mL) was added AcONa (3.49 g, 42.50 mmol) and hydroxylamine hydrochloride (2.21 g, 31.87 mmol). The mixture was stirred at 20 °C for 16 h. The reaction mixture was filtered and diluted with H2O (10 mL). The solution was extracted with EtOAc (10 mL, 3x). The combined organic leyers were washed with brine (20 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 2,4-dimethylthiazole-5-carbaldehyde oxime (3.3 g, 21.13 mmol, 99.43% yield) as a yellow solid and a mixture of cis/trans (1/1). RT 0.170 min (Method 7); m/z 157.1 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): δ = 11.90 (s, 0.5 H), 11.24 (s, 0.5H), 8.32 (s, 0.5H), 7.85 (s, 0.5H), 2.58 (s, 1.5H), 2.52(s, 1.5 H) 2.46 (s, 1.5H), 2.35 (s, 1.5H) Preparation of Intermediate 27.3 (2,4-dimethylthiazol-5-yl)methanamine
Figure imgf000108_0001
To a solution of trans and cis-2,4-dimethylthiazole-5-carbaldehyde oxime (3.3 g, 21.13 mmol) in EtOH (20 mL) and AcOH (20 mL) at 0 °C was added Zn (7.26 g, 111.03 mmol). The mixture was stirred at 20 °C for 16 h. The reaction mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase flash (ISCO®; 330 g Flash Coulmn Welch Ultimate XB_C 1820-40 μm; 120 A, Eluent of 5~50% ACN/H2O (0.1% formic acid) @150 mL/min) and lyophilized directly to give the product (2,4-dimethylthiazol-5-yl)methanamine (6 g, crude, FA) as a colorless oil. 1H NMR (DMSO-d6, 400 MHz): 8.25 (s, 1H), 3.90 (s, 2H), 2.55 (s, 3H), 2.24 (s, 3H). Preparation of Intermediate 27.4 2-(((2,4-dimethylthiazol-5-yl)methyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000109_0001
To a mixture of (2, 4-dimethylthiazol-5-yl)methanamine (5.7 g, 18.17 mmol, 60% purity) in MeCN (50 mL) were added K2CO3 (9.39 g, 67.94 mmol) and 2-fluoro-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (3.7 g, 13.59 mmol). The mixture was stirred at 100 °C for 16 h. The mixture was filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase flash (ISCO®; 330 g Flash Column Welch Ultimate XB_C 1820- 40 μm; 120 A, Eluent of 5~50% ACN/H2O (0.1% formic acid) @120 mL/min). The solvent was concentrated under reduced pressure to give the product 2-(((2,4-dimethylthiazol-5-yl)methyl)amino)-5- (N-(1-methylcyclopropyl)sulfamoyl)benzamide (2.46 g, 6.24 mmol, 45.89% yield) as a white solid. RT 0.324 min (Method 7); m/z 395.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.83 (t, J = 5.6 Hz,1H), 8.14-8.10 (m, 1H), 8.00 (d, J = 2.0 Hz, 1H), 7.63-7.56 (m, 2H), 7.38 (d, J=5.2 Hz, 1H), 6.83 (d, J = 9.2 Hz, 1H), 4.55 (d, J = 5.6 Hz, 2H), 2.52 (s, 3H), 2.31 (s, 3 H), 1.05 (s, 3H), 0.61 - 0.56 (m, 2H), 0.36 - 0.31 (m, 2H). Preparation of Intermediate 27.5 1-((2,4-dimethylthiazol-5-yl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000109_0002
To a mixture of 2-(((2,4-dimethylthiazol-5-yl)methyl)amino)-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (1.23 g, 3.12 mmol) in DMF (10 mL) was added CDI (4.04 g, 24.94 mmol) and DIEA (3.22 g, 24.94 mmol, 4.34 mL). The mixture was stirred at 100 °C for 3 h. The mixture filtered and the filtrate was purified by reversed-phase flash (ISCO®; 330 g Flash Column Welch Ultimate XB_C 1820-40 μm; 120 A, Eluent of 5~50% ACN/H2O (0.1% NH3•H2O) @120 mL/min) and lyophilized directly to give the product 1-((2,4-dimethylthiazol-5-yl)methyl)-N-(1-methylcyclopropyl)-2,4- dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (1.93 g, 4.59 mmol, 73.61% yield) as a yellow solid RT 0.320 min (Method 1); m/z 421.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.37 (d, J = 2.4 Hz, 1H), 8.19 (s, 1H), 8.08 (dd, J = 8.8, 2.4 Hz, 1H), 7.59 (d, J=8.8 Hz, 1H), 5.39 (s, 2H), 2.48 (s, 3H), 2.45 (s, 3H), 1.07 (s, 3H), 0.61-0.56 (m, 2H), 0.42-0.37 (m, 2H). Preparation of Intermediate 27.6 3-amino-1-((2,4-dimethylthiazol-5-yl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000110_0001
To a mixture of 1-((2,4-dimethylthiazol-5-yl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (200 mg, 475.62 µmol) in DMF (1 mL) and dioxane (1 mL) was added K2CO3 (98.60 mg, 713.43 µmol). The mixture was stirred at 70 °C for 0.5 h. Then, O-(2,4- dinitrophenyl)hydroxylamine (94.71 mg, 475.62 µmol) was added and the reaction mixture was stirred at 70 °C for 16 h. The mixture was filtered and the filtrate was purified by reversed-phase flash (ISCO®; 48 g Flash Column Welch Ultimate XB_C 1820-40 μm; 120 A, Eluent of 5~50% ACN/H2O (0.1% NH3•H2O) @120 mL/min) and lyophilized directly to give the product 3-amino-1-((2,4-dimethylthiazol-5-yl)methyl)- N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (90 mg, 206.65 µmol, 43.45% yield) as a gray solid. RT 0.337 min (Method 7); m/z 436.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.44 (d, J = 2.0 Hz, 1H), 8.22 (s, 1H), 8.10 (dd, J = 8.8, 2.4 Hz, 1H), 7.70 (d, J = 9.2 Hz, 1H), 5.68 (s, 2H), 5.49 (s, 2H), 2.48 (s, 3H), 2.47 (s, 3H), 1.07 (s, 3H), 0.61-0.57 (m, 2H), 0.42 - 0.37 (m, 2H). Preparation of Example 27 N-(1-((2,4-dimethylthiazol-5-yl)methyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000110_0002
The reaction was conducted according to the general procedure 1 starting from 3-amino-1-((2,4- dimethylthiazol-5-yl)methyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide. The residue was purified by preparative-HPLC (column: Waters Xbridge 150*25 mm*5 µm; mobile phase: A: 10 mM aqueous solution of NH4HCO3 in water, B: MeCN; B%: 18%-48%, 9 min) and lyophilized directly to give the product N-(1-((2,4-dimethylthiazol-5-yl)methyl)-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1- carboxamide (2.35 mg, 4.44 µmol, 4.83% yield, 97.33% purity) as a white solid. RT 0.486 min (Method 7); m/z 516.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 10.67 (s, 1H), 8.43 (d, J = 2.4 Hz, 1H), 8.28 (s, 1H), 8.17 (dd, J = 8.8, 2.2 Hz, 1H), 7.72 (d, J = 8.8 Hz, 1H), 5.53-5.47 (m, 2H), 2.52 (s, 3H), 2.45 (s, 3H), 2.40 (d, J=3.2 Hz, 2H), 2.29-2.26 (m, 1H), 1.14 (d, J =2.0 Hz, 2H), 1.08 (s, 3H), 0.62-0.58 (m, 2H), 0.43-0.39 (m, 2H). Preparation of Example 28 N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)-N-methylbicyclo[1.1.0]butane-1-carboxamide
Figure imgf000111_0001
The reaction was conducted according to the general procedure 1 starting from 3-(methylamino)- N-(1-methylcyclopropyl)-1-((1-methylcyclopropyl)methyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide. The residue was purified by preparative HPLC (column: Phenomenex luna C18150*25 mm* 10 µm; mobile phase: A: 10 mM aqueous solution of NH4HCO3 in water, B: MeCN; 28%-58%, 9 min) and lyophilized directly to give the product N-(1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)- 2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)-N-methylbicyclo[1.1.0]butane-1-carboxamide (1.63 mg, 3.54 µmol, 13.39% yield, 99.53% purity) was obtained as a white solid. RT 0.480 min (Method 7); m/z m/z 459.4 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz,): 8.54-8.36 (m, 1H), 8.28 (s, 1H), 8.19-8.09 (m, 1H), 7.91 (d, J = 8.8 Hz, 1H), 4.39-3.94 (m, 2H), 3.51 (s, 1H), 3.15 (s, 2H), 2.55 (br s, 1H), 2.41-2.15 (m, 2H), 1.98-1.87 (m, 1H), 1.47-1.22 (m, 2H), 1.20-1.00 (m, 3H), 0.85- 0.58 (m, 3H), 0.57-0.36 (m, 5H) Preparation of Intermediate 29.1 5-(N-(1-ethynylcyclopropyl)sulfamoyl)-2-fluorobenzamide
Figure imgf000112_0001
To a solution of 3-carbamoyl-4-fluoro-benzene-1-sulfonyl chloride (1.85 g, 7.79 mmol,) in THF (15 mL) and H2O (10 mL) was added NaHCO3 (5.89 g, 70.07 mmol, 2.72 mL). Then, 1- ethynylcyclopropanamine (1.01 g, 8.56 mmol, HCl) was added at 0°C and the mixture was stirred at 0 °C for 1 h. The reaction mixture was diluted with H2O (300 mL) and extracted with EtOAc (150 mL, 2x). Then, the organic layers were combined, washed with brine (150 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 5-(N-(1- ethynylcyclopropyl)sulfamoyl)-2-fluorobenzamide (1.5 g, 5.31 mmol, 68.26% yield) as a white solid. RT 0.306 min (Method 7); m/z 282.8 (M+H)+ (ESI+);1H NMR (DMSO-d6, 400 MHz): 8.73 (s, 1H), 8.12 (dd, J = 6.4, 2.4 Hz, 1H), 7.97-7.92 (m, 1H), 7.82 (s, 2H), 7.55-7.45(m, 1H), 2.71 (s, 1H), 1.17-1.13 (m, 2H), 1.04-0.99 (m, 2H) Preparation of Intermediate 29.2 5-(N-(1-ethylcyclopropyl)sulfamoyl)-2-fluorobenzamide
Figure imgf000112_0002
To a solution of 5-(N-(1-ethynylcyclopropyl)sulfamoyl)-2-fluorobenzamide (500 mg, 1.77 mmol) in THF (6.5 mL) was added quinoline (2.29 g, 17.71 mmol, 2.10 mL). Then, to the mixture was added Lindlar catalyst (99.86 mg, 483.54 µmol, 99.86 µL) under N2 atmosphere. The suspension was degassed and purged with H2 (3x). The mixture was stirred under H2 (15 PSI) at 20 °C for 24 h. The reaction mixture was filtered and the filtrate was concentrated under vacuum. The resulting residue was diluted with H2O (50 mL) and extracted with EtOAc (20 mL, 3x). The organic layers were combined, washed with HCl solution (0.5 N, 30 mL, 3x) and brine (10 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue. The residue was purified by reversed-phase flash (48 g Flash column C1820-40 µm, 120 A, Eluent of 0~24% ACN/H2O (0.1% FA condition) @ 70 mL/min) to give the product 5-(N-(1-ethylcyclopropyl)sulfamoyl)-2-fluorobenzamide (310 mg, 1.08 mmol, 61.13% yield) as a white solid. RT 0.395 min (Method 7); m/z 287.0 (M+H)+ (ESI+);1H NMR (DMSO-d6, 400 MHz): 8.19 (s, 1H), 8.05 (dd, J = 6.4, 2.4 Hz, 1H), 7.90 (m, 2H), 7.85 (s, 1H), 7.51 (dd, J = 10.0, 8.8 Hz, 1H), 1.29 (q, J = 7.2 Hz, 2H, 0.77 (t, J = 7.2 Hz, 3H), 0.53-0.48 (m, 2H), 0.43-0.37 (m, 2H) Preparation of Intermediate 29.3 2-((cyclopropylmethyl)amino)-5-(N-(1-ethylcyclopropyl)sulfamoyl)benzamide
Figure imgf000113_0001
To a solution of 5-(N-(1-ethylcyclopropyl)sulfamoyl)-2-fluorobenzamide (310 mg, 1.08 mmol) in MeCN (10 mL) was added K2CO3 (299.27 mg, 2.17 mmol) and cyclopropylmethanamine (231.00 mg, 3.25 mmol). The mixture was stirred at 100°C for 16 h. The mixture was diluted with H2O (60 mL) and extracted with EtOAc (30 mL, 2x). Then, the combined layer was washed with brine (30 mL, 2x), dried over anhydrous Na2SO4 filtered and the filtrate was concentrated under reduced pressure to give the product 2-((cyclopropylmethyl)amino)-5-(N-(1-ethylcyclopropyl)sulfamoyl)benzamide (340 mg, 1.01 mmol, 93.06% yield) as a yellow solid. RT 0.518 min (Method 7); m/z 338.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.60 (t, J = 4.2 Hz, 1H), 8.04 (s, 1H), 7.97 (d, J = 2.0 Hz, 1 H), 7.57 (dd, J = 9.2, 2.4 Hz, 1H), 7.54 (s, 1H), 7.31 (s, 1H), 6.77 (d, J = 9.2 Hz, 1H), 3.05 (dd, J = 7.2, 4.2 Hz, 2H), 1.29 (m, 2H), 1.14-1.04 (m, 1H), 0.77 (t, J = 7.2 Hz, 3H), 0.54-0.47 (m, 4H), 0.36-0.31 (m, 2H), 0.28-0.22 (m, 2H) Preparation of Intermediate 29.4 1-(cyclopropylmethyl)-N-(1-ethylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000113_0002
To a solution of 2-((cyclopropylmethyl)amino)-5-(N-(1-ethylcyclopropyl)sulfamoyl)benzamide (340 mg, 1.01 mmol) in DMF (5 mL) was added CDI (1.31 g, 8.06 mmol) and DIEA (1.04 g, 8.06 mmol, 1.40 mL). The mixture was stirred at 100 °C for 2 h. The reaction was diluted with H2O (100 mL) and extracted with EtOAc (50 mL, 3x). The organic layers were combined; washed with HCl solution (0.5 N, 50 mL, 2x) and brine (50 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 1-(cyclopropylmethyl)-N-(1-ethylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (300 mg, 825.47 µmol, 81.92% yield) as a white solid. RT 0.398 min (Method 7); m/z 364.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 11.85 (s, 1H), 8.36 (d, J = 2.4 Hz, 1H), 8.18 (s, 1H), 8.04 (dd, J = 9.2, 2.4 Hz, 1H), 7.76 (d, J = 8.8 Hz, 1H), 4.01 (d, J = 6.8 Hz, 2H), 1.31 (m, 2H), 1.24-1.15 (m, 1H), 0.78 (t, J = 7.2 Hz, 3H), 0.55-0.37 (m, 8H) Preparation of Intermediate 29.5 3-amino-1-(cyclopropylmethyl)-N-(1-ethylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000114_0001
To a solution of 1-(cyclopropylmethyl)-N-(1-ethylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (100 mg, 275.16 µmol) in DMF (1 mL) and dioxane (1 mL) was added K2CO3 (76.06 mg, 550.31 µmol). The mixture was stirred at 80°C for 0.5 h. O-(2,4- dinitrophenyl)hydroxylamine (109.58 mg, 550.31 µmol) was added and the reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was diluted with H2O (50 mL) and extracted with EtOAc (20 mL, 3x). Then, the combined organic layer was washed with brine (20 mL, 5x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 3-amino-1- (cyclopropylmethyl)-N-(1-ethylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (70 mg, 184.97 µmol, 67.22% yield) as a yellow solid. RT 0.381 min (Method 7); m/z 379.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.43 (d, J = 2.4 Hz, 1H), 8.21 (s, 1H), 8.05 (dd, J = 8.8, 2.4 Hz, 1H), 7.82 (d, J = 8.8 Hz, 1H), 5.67 (s, 2H), 4.11 (d, J = 7.2 Hz, 2H), 1.35-1.27 (m, 2H), 1.27-1.20 (m, 1H), 0.77 (t, J =7.2 Hz, 3H), 0.54-0.38 (m, 8H) Preparation of Example 29 N-(1-(cyclopropylmethyl)-6-(N-(1-ethylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000114_0002
The reaction was conducted according to the general procedure 1 starting from 3-amino-1- (cyclopropylmethyl)-N-(1-ethylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide. The residue was purified by preparative HPLC (column: Waters Xbridge 150*25 mm 10 µm; mobile phase: A: 0.05% NH3·H2O in water; B: MeCN; B%: 24%-54%, 11 min). The product solution was lyophilized to give the product N-(1-(cyclopropylmethyl)-6-(N-(1-ethylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide (8.52 mg, 18.58 µmol, 23.44% yield, 99.99% purity) as a white solid. RT 0.403 min (Method 7); m/z 459.3 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 10.58 (s, 1H), 8.41 (d, J = 2.4 Hz, 1H), 8.27 (s, 1H), 8.12 (dd, J = 8.8, 2.0 Hz, 1 H), 7.86 (d, J = 9.2 Hz, 1H), 4.18-4.02 (m, 2H), 2.40 (d, J = 3.2 Hz, 2H), 2.30-2.22 ,(m 1H), 1.32 (m, 2H), 1.27-1.19 (m, 1H), 1.13 (d, J = 2 Hz, 2H), 0.78 (t, J = 7.2 Hz, 3H), 0.55-0.47 (m, 4H), 0.47-0.39 (m, 4H) Preparation of Intermediate 30.1 1-methylquinazoline-2,4(1H,3H)-dione
Figure imgf000115_0001
To a stirred solution of methyl 2-(methylamino)benzoate (25 g, 151.34 mmol) in AcOH (120 mL) was added KOCN (15.46 g, 181.61 mmol) in H2O (30 mL). The solution was stirred at 20 °C for 3 h. The temperature was raised to 80°C and stirring was pursued for 5 h. The reaction mixture was diluted with water (200 mL). The precipitate was collected by filtration, triturated in EtOH (150 mL) at 20 °C for 0.5 h, filtered and dried under vacuum to afford the product 1-methylquinazoline-2,4(1H,3H)-dione (20 g, 113.53 mmol, 75.01% yield) as a white solid. RT 0.394 min (Method 7); m/z 177.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 11.54 (br, 1H), 8.04-7.98 (m, 1H), 7.81-7.74 (m, 1H), 7.43 (d, J = 8.4 Hz, 1H), 7.29 (t, J = 7.6 Hz, 1H), 3.45 (s, 3H). Preparation of Intermediate 30.2 1-methyl-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonyl chloride
Figure imgf000115_0002
1-methylquinazoline-2,4(1H,3H)-dione (4 g, 22.71 mmol) was added to chlorosulfonic acid (4.76 g, 40.87 mmol, 2.72 mL) and the mixture was stirred at 50°C for 16 h. The resulting mixture was poured to ice/water (200 mL). The precipitate was collected by filtration, washed with water (20 mL, 2x) and dried under vacuum to afford the product 1-methyl-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonyl chloride (2.5 g, 9.10 mmol, 40.09% yield) as a white solid. RT 0.673 min (Method 8); m/z 275.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 11.56 (br, 1H), 8.19 (d, J = 2.0 Hz, 1H), 7.91 (dd, J = 2.1, 8.6 Hz, 1H), 7.37 (d, J = 8.8 Hz, 1H), 3.44 (s, 3H). Preparation of Intermediate 30.3 1-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide
Figure imgf000116_0001
To a solution of 1-methylcyclopropanamine (1.17 g, 10.92 mmol, HCl salt) in saturated aqueous NaHCO3 (15.00 mL) was added 1-methyl-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonyl chloride (1.5 g, 5.46 mmol) in THF (15 mL) at 0°C. The mixture was stirred at 20°C for 16 h. The mixture was filtered, and a yellow solid was collected and dried under vacuum to afford the product 3-amino-1-methyl-N-(1- methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (1 g, 3.04 mmol, 55.65% yield, 94% purity) as a yellow solid. RT 0.757 min (Method 8); m/z 310.1 (M+H)+ (ES+); 1H NMR (DMSO-d6, 400 MHz): 8.34 (d, J = 2.4 Hz, 1H), 8.04 (dd, J = 2.4, 8.8 Hz, 1H), 7.58 (d, J = 8.8 Hz, 1H), 3.46 (s, 3H), 1.06 (s, 3H), 0.62-0.55 (m, 2H), 0.41-0.36 (m, 2H) Preparation of Intermediate 30.4 3-amino-1-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide
Figure imgf000116_0002
To a solution of 1-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide (500 mg, 1.62 mmol) in DMF (2.5 mL) and dioxane (2.5 mL) was added K2CO3 (446.78 mg, 3.23 mmol). The mixture was stirred at 80°C for 0.5 h. O-(2,4-dinitrophenyl)hydroxylamine (643.69 mg, 3.23 mmol) was added to the reaction and the mixture was stirred at 80°C for 16 h. The mixture was quenched by cold water (10 mL) and extracted with EtOAc (10 mL, 2x). The combined organic layers were washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The resulting residue was purified by silica gel chromatography flash (ISCO®; 20 g SepaFlash® Silica Flash Column, Eluent of 0~10% DCM /MeOH @ 60 mL/min) to afford the product 3-amino-1-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (200 mg, 542.62 µmol, 33.57% yield, 88% purity) as a yellow solid. RT 0.453 min (Method 7); m/z 325.0 (M+H)+ 1H NMR (DMSO-d6, 400 MHz): 8.42 (d, J = 2.0 Hz, 1H), 8.19 (s, 1H), 8.07 (dd, J = 2.48.8 Hz, 1H), 7.67 (d, J = 8.8 Hz, 1H), 5.8-5.4 (br, 2H), 3.59 (s, 3H), 1.07 (s, 3H), 0.63-0.55 (m, 2H), 0.44-0.33 (m, 2H). Preparation of Example 30 N-(1-methyl-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000117_0001
The reaction was conducted according to the general procedure 1 starting from 3-amino-1-methyl- N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide. The residue was purified by prep-HPLC (column: Waters Xbridge C18150*50 mm* 10 µm;mobile phase: A: 10 mM aqueous solution of NH4HCO3, B: MeCN; B%: 16%-46%,10 min) to afford N-(1-methyl-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1- carboxamide (15.44 mg, 38.18 µmol, 24.77% yield, 100% purity) as a white solid. RT 0.782 min (Method 8); m/z 405.1 (M+H)+ 1H NMR (DMSO-d6, 400 MHz): 10.59 (br, 1H), 8.40 (d, J = 2.0 Hz, 1H), 8.27-8.22 (m, 1H), 8.14 (dd, J = 2.4, 8.8 Hz, 1H), 7.71 (d, J = 8.8 Hz, 1H), 3.58 (s, 3H), 2.41 (d, J = 3.2 Hz, 2H), , 2.24 (t, J = 3.2 Hz, 1H), 1.18-1.12 (m, 2H), 1.07 (s, 3H), 0.63-0.58 (m, 2H), 0.44-0.34 (m, 2H). Preparation of Intermediate 31.1 7-(benzylthio)isoquinolin-1(2H)-one
Figure imgf000117_0002
To a mixture of 7-bromoisoquinolin-1(2H)-one (10 g, 44.63 mmol), Pd2(dba)3 (2.04 g, 2.23 mmol), Xantphos (2.58 g, 4.46 mmol) and DIPEA (6.35 g, 49.10 mmol, 8.55 mL) in dioxane (100 mL) was added phenylmethanethiol (6.10 g, 49.10 mmol, 5.75 mL). Then, the mixture was degassed and purged with N2 (3x) before it was stirred at 80°C for 5 h under N2 atmosphere. The reaction mixture was poured into DCM (50 mL) and stirred at 20°C for 15 min. The precipitate was collected by filtration, triturated with EtOAc (20 mL) at 25°C for 15 min and dried under reduce pressure to give the product 7-(benzylthio)isoquinolin- 1(2H)-one (10 g, 37.40 mmol, 83.81% yield) as a yellow solid RT 0.904 min (Method 7); m/z 268.1 (M+H)+ (ESI+);1H NMR (DMSO-d6, 400 MHz): 11.27 (s, 1H), 8.05 (s, 1H), 7.62-7.67 (m, 1H), 7.56-7.61 (m, 1H), 7.33-7.40 (m, 2H), 7.19-7.33 (m, 3H), 7.13 (t, J = 6.4 Hz, 1H), 6.51 (d, -J = 6.8 Hz, 1H), 4.32 (s, 2H) Preparation of Intermediate 31.2 7-(benzylthio)-4-bromoisoquinolin-1(2H)-one
Figure imgf000118_0001
To a solution of 7-(benzylthio)isoquinolin-1(2H)-one (8.5 g, 31.79 mmol) in MeCN (200 mL) was added NBS (6.22 g, 34.97 mmol). The suspension was stirred at 20°C for 12 h. The solid was collected by filtration, triturated with EtOAc (20 mL) at 25°C for 15 min and dried under reduce pressure to give the product 7-(benzylthio)-4-bromoisoquinolin-1(2H)-one (5 g, 14.44 mmol, 45.42% yield) as a brown solid. RT 0.924 min (Method 8); m/z 346.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz) 11.62 (d, J = 4.4 Hz, 1H), 8.08 (d, J = 2.0 Hz, 1H), 7.80 (dd, J = 8.4, 2.0 Hz, 1H), 7.67 (d, J = 8.8 Hz, 1H), 7.49-7.52 (m, 1H), 7.37-7.42 (m, 2H), 7.28-7.33 (m, 2H), 7.20-7.26 (m, 1H), 4.37 (s, 2H) Preparation of Intermediate 31.3 7-(benzylthio)-4-(cyclopropanecarbonyl)isoquinolin-1(2H)-one
Figure imgf000118_0002
MeLi (1.6 M, 9.34 mL) was added to a suspension of 7-(benzylthio)-4-bromoisoquinolin-1(2H)-one (4.5 g, 13.00 mmol) in THF (15 mL) at 20°C. After 10 min, the reaction mixture was cooled to -78°C and n-BuLi (2.5 M, 5.98 mL) was added dropwise. After 10 min, cyclopropanecarbonyl chloride (1.56 g, 14.95 mmol, 1.36 mL) was added and stirred for 10 min. Then, the mixture was warmed to 20°C, stirred at 20 °C for 1h under N2, poured into ice-water (100 mL) and EtOAc (50 mL) and stirred for 15 min. The precipitate was collected by filtration, triturated with EtOAc (20 mL) at 25°C for 15 min and dried under reduce pressure to give the product 7-(benzylthio)-4-(cyclopropanecarbonyl)isoquinolin-1(2H)-one (2 g, 5.96 mmol, 45.88% yield) as a yellow solid RT 0.635 min (Method 7); m/z 336.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz) 12.00 (s, 1H), 8.65 (d, J = 8.8 Hz, 1H), 8.28 (s, 1H), 8.10 (d, J = 2.0 Hz, 1H), 7.73 (dd, J = 8.8, 2.4 Hz, 1H), 7.37-7.42 (m, 2 H), 7.27-7.33 (m, 2H), 7.21-7.26 (m, 1H), 4.36 (s, 2H), 2.67-2.71 (m, 1H), 0.93-1.03 (m, 4H) Preparation of Intermediate 31.4 4-(cyclopropanecarbonyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonyl chloride
Figure imgf000119_0001
At 0 °C, to a solution of 7-(benzylthio)-4-(cyclopropanecarbonyl)isoquinolin-1(2H)-one (400 mg, 1.19 mmol) in MeCN (4 mL), AcOH (0.06mL) and H2O (0.06mL) was added 1,3-dichloro-5,5-dimethyl- imidazolidine-2,4-dione (305 mg, 1.55 mmol) portionwise. After stirring at 0°C for 5 min, the mixture was filtered to give the product 4-(cyclopropanecarbonyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonyl chloride (350 mg,1.12 mmol, 94.15 % yield) as a yellow solid. Preparation of Intermediate 31.5 4-(cyclopropanecarbonyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonamide
Figure imgf000119_0002
To a solution of 1-methylcyclopropanamine (241.57 mg, 2.25 mmol, HCl salt) in saturated, aqueous NaHCO3 solution (1.12 mmol, 4 mL) was dropwise added 4-(cyclopropanecarbonyl)-1-oxo--1,2-dihydro 2H-isoquinoline-7-sulfonyl chloride (350 mg, 1.12 mmol) in THF (4 mL) in 0°C. The mixture was stirred at 20°C for 2 h. The reaction mixture was diluted with H2O (20 mL) and extracted with EtOAc (20mL, 2x). The organic layer was washed with brine (20mL, 2x), dried over Na2SO4 and concentrated under reduced pressure to give a residue. The residue was triturated with petroleum ether and EtOAc (v/v= 1/1, 5 mL) for 15 min. The solid was dried under reduce pressure to give the product 4-(cyclopropanecarbonyl)-N- (1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonamide (100 mg, 288.68 µmol, 25.71% yield) as a yellow solid. RT 0.557 min (Method 7); m/z 347.1 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz) 12.20 (br, 1H), 8.89 (d, J = 8.8 Hz, 1H), 8.64 (d, J = 2.0 Hz, 1H), 8.46 (s, 1 H), 8.23 (s, 1H), 8.09 (dd, J = 8.8, 2.0 Hz, 1H), 2.70-2.78 (m, 1H), 0.97-1.07 (m, 7H), 0.55-0.62 (m, 2H), 0.35-0.42 (m, 2H) Preparation of Intermediate 31.6 2-amino-4-(cyclopropanecarbonyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7- sulfonamide
Figure imgf000120_0001
To a solution of 4-(cyclopropanecarbonyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline- 7-sulfonamide (220 mg, 0.635 mmol) in DMF (3mL) was added Cs2CO3 (248 mg, 0.762 mmol). The reaction mixture was stirred at 20 °C for 15 min. (Aminooxy)diphenylphosphine oxide (178 mg, 0.762 mmol) was aded to the mixture and the reaction mixture was stirred at 20°C for 1 h and subsequently at 50°C for 12 h. The reaction mixture was cooled to RT, diluted with H2O (30 mL) and extracted with EtOAc (20 mL, 2x). The organic layers were combined; washed with brine (30 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum. The resulting residue was triturated with petroleum ether : EtOAc (v/v=1/1, 5 mL) for 15 min. The solid was filtered and dried under reduce pressure to give the product 2-amino-4-(cyclopropanecarbonyl)-N-(1-methylcyclopropyl)-1-oxo-1,2- dihydroisoquinoline-7-sulfonamide (110 mg, 0.304 mmol, 47.92 % yield) as a yellow solid. RT 0.551 min (Method 8); m/z 362.3 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.92 (d, J = 8.8 Hz, 1H), 8.78 (s, 1 H), 8.71 (d, J = 2.0 Hz, 1H), 8.28 (s, 1 H), 8.06-8.12 (m, 1H), 6.29 (s, 2H), 2.74- 2.80 (m, 1H), 0.97-1.09 (m, 7H), 0.53-0.63 (m, 2 H), 0.34-0.43 (m, 2H) Preparation of Example 31 N-(4-(cyclopropanecarbonyl)-7-(N-(1-methylcyclopropyl)sulfamoyl)-1-oxoisoquinolin-2(1H)- yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000120_0002
The reaction was conducted according to the general procedure 1 starting from 2-amino-4- (cyclopropanecarbonyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonamide. The residue was purified by preparative HPLC (column: Phenomenex Synergi C18150*25 mm* 5 µm; mobile phase: A: 10 mM aqueous solution of NH4HCO3, B: MeCN; B%: 31%-61%, 9 min) to give the product N- (4-(cyclopropanecarbonyl)-7-(N-(1-methylcyclopropyl)sulfamoyl)-1-oxoisoquinolin-2(1H)- yl)bicyclo[1.1.0]butane-1-carboxamide (1.6 mg, 0.00342 mmol, 8.24 %yield) as an off-white solid RT 0.825 min (Method 8); m/z 442.2 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 11.25 (s, 1H), 8.88 (d, J = 8.8 Hz, 1H), 8.70 (s, 1 H), 8.68 (d, J = 2.0 Hz, 1H), 8.32 (s, 1 H), 8.16 (dd, J = 8.8, 2.0 Hz, 1H), 2.71-2.77 (m, 1H), 2.58-2.64 (m, 2H), 2.37-2.41 (m, 1H), 1.21 (d, J = 2.4 Hz, 2H), 0.99-1.09 (m, 7H), 0.56-0.64 (m, 2H), 0.36-0.42 (m, 2H). Preparation of Intermediate 32.1 5-(N-(1-methylcyclopropyl)sulfamoyl)-2-(prop-2-yn-1-ylamino)benzamide
Figure imgf000121_0001
To a solution of 2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (2.0 g, 7.34 mmol) and prop-2-yn-1-amine (404.55 mg, 7.34 mmol, 470.40 µL) in MeCN (20 mL) was added K2CO3 (1.02 g, 7.34 mmol). The mixture was stirred at 70°C for 16h and prop-2-yn-1-amine (809.10 mg, 14.69 mmol, 940.81 µL) was added. Then, the mixture was stirred at 70°C for another 16 h and cooled to 20 °C. After filtration, the filtrate was concentrated under vacuum to give a residue which was purified by flash silica gel chromatography (ISCO®; 40 g SepaFlash® Silica Flash Column, Eluent of 0~47% Petroleum ether/Ethyl acetate @ 50 mL/min) to give the product 5-(N-(1-methylcyclopropyl)sulfamoyl)-2-(prop-2-yn-1- ylamino)benzamide (2.02 g, 5.59 mmol, 76.05% yield, 85% purity) as a brown solid. RT 0.348 min (Method 7); m/z 308.0 (M+H)+ (ESI+), 1H NMR (DMSO-d6, 400 MHz): 8.61 (t, J = 5.6 Hz, 1H), 8.16-8.04 (m, 1H), 8.01 (d, J = 2.0 Hz, 1H), 7.66 (dd, J = 8.8 Hz, 2.0 Hz, 1H), 7.59 (s, 1H), 7.40 (br s, 1H), 6.87 (d, J = 9.2 Hz, 1H), 4.15-4.08 (m, 2H), 3.18-3.16 (m, 1H), 1.06 (s, 3H), 0.65-0.55 (m, 2H), 0.42-0.30 (m, 2H). Preparation of Intermediate 32.2 N-(1-methylcyclopropyl)-2,4-dioxo-1-(prop-2-yn-1-yl)-1,2,3,4-tetrahydroquinazoline-6-sulfonamide
Figure imgf000122_0001
To a solution of 5-(N-(1-methylcyclopropyl)sulfamoyl)-2-(prop-2-yn-1-ylamino)benzamide (1.82 g, 5.92 mmol) and CDI (2.88 g, 17.76 mmol) in DMF (46 mL) was added DIEA (2.29 g, 17.76 mmol, 3.09 mL) and the mixture was stirred at 130°C for 1 h. Then, the reaction mixture was cooled to 20°C. After filtration, the filtrate was diluted with water. The aqueous layer was extracted with EtOAc (80 mL; 2x) and the combined organic layer washed with brine (80 mL; 2x). The combined organic layer was dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum to give the product N-(1- methylcyclopropyl)-2,4-dioxo-1-(prop-2-yn-1-yl)-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (2.02 g, 5.15 mmol, 86.99% yield, 85% purity) as a black brown oil. RT 0.324 min (Method 7); m/z 334.0 (M+H)+ (ESI+), 1H NMR (DMSO-d6, 400 MHz): 11.99 (br-, 1H), 8.38 (d, J = 2.4 Hz, 1H), 8.20 (s, 1H), 8.12 (dd, J = 8.8 Hz, 2.4 Hz, 1H), 7.70-7.62 (m, 1H), 3.38 (t, J = 2.4 Hz, 1H), 3.29 (s, 2H), 1.08 (s, 3H), 0.67-0.55 (m, 2H), 0.44-0.35 (m, 2H). Preparation of Intermediate 32.3 N-(1-methylcyclopropyl)-2,4-dioxo-1-(3-(triisopropylsilyl)prop-2-yn-1-yl)-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000122_0002
A solution of N-(1-methylcyclopropyl)-2,4-dioxo-1-(prop-2-yn-1-yl)-1,2,3,4-tetrahydroquinazoline-6- sulfonamide (200 mg, 599.95 µmol) in THF (2 mL) was degassed and purged with N2 (3x). Then, n-BuLi (2.5 M, 719.94 µL) was added dropwise via syringe over 5 min at -78 °C and the mixture was stirred at - 78 °C for 15 min. Then, the mixture was warmed to 0 °C and TIPSCl (115.67 mg, 599.95 µmol, 128.38 µL) was added dropwise via syringe over 1 min and the mixture was stirred at 0 °C for 1.5 h. The reaction mixture was quenched with NH4Cl (aq., sat., 10 mL) at 0 °C and extracted with EtOAc (15 mL, 3x). The combined organic layer was washed with brine (20 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The residue was purified by preparative TLC (Petroleum ether: Ethyl acetate= 2:1) to give the product N-(1-methylcyclopropyl)-2,4-dioxo-1-(3- (triisopropylsilyl)prop-2-yn-1-yl)-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (120 mg, 245.05 µmol, 40.84% yield) as a yellow solid. RT 0.584 min (Method 7); m/z 490.4 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 12.02 (br-, 1H), 8.37 (d, J = 2.0 Hz, 1H), 8.19 (s, 1H), 8.07 (dd, J = 2.4, 8.8 Hz, 1H), 7.72 (d, J = 8.8 Hz, 1H), 5.01 (s, 2H), 1.04 (s, 3H), 0.93 (s, 21H), 0.60-0.54 (m, 2H), 0.41-0.35 (m, 2H). Preparation of Intermediate 32.4 3-amino-N-(1-methylcyclopropyl)-2,4-dioxo-1-(3-(triisopropylsilyl)prop-2-yn-1-yl)-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000123_0001
To a solution of N-(1-methylcyclopropyl)-2,4-dioxo-1-(3-(triisopropylsilyl)prop-2-yn-1-yl)-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (120 mg, 245.05 µmol) in DMF (1 mL) and dioxane (1 mL) was added K2CO3 (50.80 mg, 367.57 µmol) and the mixture was stirred at 70 °C for 0.5 h. Then, O-(2,4- dinitrophenyl)hydroxylamine (48.79 mg, 245.05 µmol) was added and the mixture was stirred at 70 °C for 16 h. Then, the reaction mixture was diluted with water (10 mL) at 0 °C and extracted with EtOAc (15 mL, 3x). The combined organic layer was washed with brine (20 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The resulting residue was purified by reversed-phase flash (column: 48 g Flash Column Welch Ultimate XB_C1820-40 μm; mobile phase: A: 0.1% NH3•H2O in water, B: MeCN; B%: 5%-50%, 50 min). The solvent was concentrated under reduced pressure to give the product 3-amino-N-(1-methylcyclopropyl)-2,4-dioxo-1-(3-(triisopropylsilyl)prop-2-yn- 1-yl)-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (105 mg, 208.04 µmol, 84.90% yield) as a light yellow solid. RT 0.582 min (Method 7); m/z 505.6 (M+H)+ (ESI+), 1H NMR (DMSO-d6, 400 MHz): 8.44 (d, J = 2.4 Hz, 1H), 8.23 (s, 1H), 8.08 (dd, J = 8.8, 2.4 Hz, 1H), 7.77 (d, J =8.8 Hz, 1 H), 5.68 (s, 2H), 5.12 (s, 2H), 1.04 (s, 3H), 0.96-0.89 (m, 21H), 0.60-0.55 (m, 2H), 0.40-0.34 (m, 2H). Preparation of Intermediate 32.5 N-(6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1-(3-(triisopropylsilyl)prop-2-yn-1-yl)-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000124_0001
The reaction was conducted according to the general procedure 1 starting from 3-amino-N-(1- methylcyclopropyl)-2,4-dioxo-1-(3-(triisopropylsilyl)prop-2-yn-1-yl)-1,2,3,4-tetrahydroquinazoline-6- sulfonamide. The residue was purified by preparative TLC (Petroleum ether: Ethyl acetate= 1:1) to give the product N-(6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1-(3-(triisopropylsilyl)prop-2-yn-1-yl)-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide (10 mg, 16.8 µmol, 24.17% yield, 98% purity) as a yellow solid. RT 0.578 min (Method 7); m/z 585.6 (M+H)+ (ESI+); 1H NMR (CDCl3, 400 MHz) δ 8.64 (d, J = 2.1 Hz, 1H), 8.12 (dd, J = 2.2, 8.9 Hz, 1H), 7.61 (s, 1H), 7.55 (d, J = 8.8 Hz, 1H), 5.25-4.66 (m, 3H), 2.44 (d, J = 2.6 Hz, 2H), 2.31-2.18 (m, 1H), 1.23-1.21 (m, 2H), 1.16 (s, 3H), 0.98-0.90 (m, 21H), 0.70- 0.69 (m, 2H), 0.47 - 0.38 (m, 2H) Preparation of Example 32 N-(6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1-(prop-2-yn-1-yl)-1,4-dihydroquinazolin- 3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000124_0002
To a solution of N-(6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1-(3-(triisopropylsilyl)prop-2- yn-1-yl)-1,4-dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide (10 mg, 0.0171 mmol) in THF (0.3 mL) was added AcOH (2.9 mg, 0.0479 mmol) and TBAF (1 M, 0.089 mL) at 0°C and the mixture was stirred at 20°C for 1 h. The reaction mixture was diluted with water (10 mL) at 20 °C and extracted with EtOAc (15 mL, 3x). The combined organic layer was washed with brine (20 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate concentrated under reduced pressure. The resulting residue was purified by preparative TLC (Petroleum ether: Ethyl acetate=2:1) twice followed by a purification via preparative HPLC (column: Waters Xbridge 150*25 mm 10 µm; mobile phase: A: 10 mM NH4HCO3 in water, B: MeCN; B%: 15%-45%,9 min) and, then lyophilized directly to give the product N-(6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1-(prop-2-yn-1-yl)-1,4-dihydroquinazolin-3(2H)- yl)bicyclo[1.1.0]butane-1-carboxamide (1.2 mg, 2.6 µmol, 15.37% yield, 93.82% purity) as a white solid. RT 0.344 min (Method 7); m/z 429.3 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 10.64 (s, 1H), 8.43 (s, 1H), 8.28 (s, 1H), 8.23-8.18 (m, 1H), 7.77 (br d, J = 9.2 Hz, 1H), 5.01 (br d, J = 5.6 Hz, 2H), 3.45- 3.44 (m, 1H), 2.41 (br d, J = 2.0 Hz, 2H), 2.26 (s, 1H), 1.15 (br s, 2H), 1.08 (s, 3H), 0.61 (br s, 2H), 0.41 (br s, 2H). Preparation of Intermediate 33.1 5-(N-(1-methylcyclopropyl)sulfamoyl)-2-((3,3,3-trifluoropropyl)amino)benzamide
Figure imgf000125_0001
To a solution of 2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (650 mg, 2.39 mmol) in DMF (6 mL) was added Cs2CO3 (3.11 g, 9.55 mmol) and 3,3,3-trifluoropropan-1-amine (713.95 mg, 4.77 mmol) and the mixture was stirred at 80 °C for 16 h. The mixture was poured into water (30 mL) and extracted with EtOAc (15 mL, 3x). The combined organic layer was washed with brine (20 mL, 3x), dried over anhydrous Na2SO4, filtered and the filtrate concentrated under vacuum. The residue was purified by flash silica gel chromatography (ISCO®; 12 g SepaFlash® Silica Flash Column, Eluent of 0~50% Ethyl acetate/Petroleum ether @ 35 mL/min) and concentrated under vacuum to give the product 5-(N-(1- methylcyclopropyl)sulfamoyl)-2-((3,3,3-trifluoropropyl)amino)benzamide (306.5 mg, 838.87 µmol, 35.14% yield) as a yellow solid. RT 0.392 min (Method 7); m/z 366.2 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6); 8.67 (t, J = 5.6 Hz, 1H), 8.00 (d, J = 2.0 Hz, 1H), 7.94-7.88 (m, 1H), 7.63 (dd, J = 8.8, 2.0 Hz, 1H), 7.59 (s, 1H), 7.45-7.32 (m, 1H), 6.86 (d, J = 8.8 Hz, 1H), 3.51-3.44 (m, 2H), 2.67-2.56 (m, 2H), 1.06 (s, 3H), 0.61-0.58 (m, 2H), 0.37-0.31 (m, 2H) Preparation of Intermediate 33.2 N-(1-methylcyclopropyl)-2,4-dioxo-1-(3,3,3-trifluoropropyl)-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000126_0001
To a solution of 5-(N-(1-methylcyclopropyl)sulfamoyl)-2-((3,3,3-trifluoropropyl)amino)benzamide (310 mg, 848.45 µmol) in DMF (5 mL) was added CDI (1.10 g, 6.79 mmol) and DIEA (877.23 mg, 6.79 mmol, 1.18 mL) and the mixture was stirred at 90°C for 2 h. The mixture was poured into water (40 mL) and extracted with EtOAc (20 mL; 2x). The combined organic layer was washed with HCl (aq., 0.5 N, 30 mL) and brine (20 mL; 3x), dried over anhydrous Na2SO4, filtered and the filtrate concentrated under vacuum to give the product N-(1-methylcyclopropyl)-2,4-dioxo-1-(3,3,3-trifluoropropyl)-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (260 mg, 664.34 µmol, 78.30% yield) as a yellow solid. RT 0.395 min (Method 7); m/z 392.1 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6) 11.95 (s, 1H), 8.37 (d, J = 2.4 Hz, 1H), 8.20 (s, 1H), 8.06 (dd, J = 8.4, 2.0 Hz, 1H), 7.64 (d, J = 9.2 Hz, 1H), 4.34 (t, J = 6.8 Hz, 2H), 2.76-2.65 (m, 2H), 1.08 (s, 3H), 0.61-0.57 (m, 2H), 0.42-0.37 (m, 2H) Preparation of Intermediate 33.3 3-amino-N-(1-methylcyclopropyl)-2,4-dioxo-1-(3,3,3-trifluoropropyl)-1,2,3,4-tetrahydroquinazoline- 6-sulfonamide
Figure imgf000126_0002
To a solution of N-(1-methylcyclopropyl)-2,4-dioxo-1-(3,3,3-trifluoropropyl)-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (100 mg, 255.52 µmol) in dioxane (1 mL) and DMF (1 mL) was added K2CO3 (70.63 mg, 511.03 µmol) and the mixture was stirred at 70°C for 0.5 h. Then, O-(2,4- dinitrophenyl)hydroxylamine (76.32 mg, 383.27 µmol) was added. The resulting mixture was stirred at 70°C for 13.5 h. Then, the mixture was poured into water (30 mL) and extracted with EtOAc (15 mL; 3x). The combined organic layer was washed with brine (20 mL; 5x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under vacuum to give the product 3-amino-N-(1-methylcyclopropyl)-2,4- dioxo-1-(3,3,3-trifluoropropyl)quinazoline-6-sulfonamide (85 mg, 209.16 µmol, 81.86% yield) as a brown solid. RT 0.359 min (Method 7); m/z 407.2 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6) 8.44 (d, J = 2.4 Hz, 1H), 8.23 (s, 1H), 8.07 (dd, J = 8.8, 2.0 Hz, 1H), 7.71 (d, J = 8.8 Hz, 1H), 5.68 (s, 2H), 4.43 (t, J = 6.8 Hz, 2H), 2.81-2.70 (m, 2H), 1.08 (s, 3H), 0.64-0.55 (m, 2H), 0.46-0.35 (m, 2H) Preparation of Example 33a N-(6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1-(3,3,3-trifluoropropyl)-1,4-dihydroquinazolin- 3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide and example 33b N-(6-(N-(1-methylcyclopropyl)sulfamoyl)- 2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000127_0001
The reaction was conducted according to the general procedure 1 starting from 3-amino-N-(1- methylcyclopropyl)-2,4-dioxo-1-(3,3,3-trifluoropropyl)quinazoline-6-sulfonamide. The residue was purified by preparative-HPLC (column: Waters Xbridge 150*25 mm*10 µm; mobile phase: A: 10 mM NH4HCO3 in water, B: MeCN; B%: 34%-64%, 9 min) and lyophilized directly to give the product N-(6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1-(3,3,3-trifluoropropyl)-1,4-dihydroquinazolin-3(2H)- yl)bicyclo[1.1.0]butane-1-carboxamide (4.48 mg, 9.20 µmol, 4.67% yield, 99.78% purity) as a white solid, and the product N-(6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)bicyclo[1.1.0]butane-1-carboxamide (1.42 mg, 3.60 µmol, 1.81% yield, 97.99% purity) as a white solid. Example 33a RT 0.382 min (Method 7); m/z 487.3 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6): 10.64 (s, 1H), 8.43 (d, J = 2.0 Hz, 1H), 8.28 (s, 1H), 8.14 (dd, J = 8.8, 2.4 Hz, 1H), 7.74 (d, J = 8.8 Hz, 1H), 4.51-4.35 (m, 2H), 2.81-2.70 (m, 2H), 2.41 (d, J = 3.2 Hz, 2H), 2.27-2.25 (m, 1H), 1.14 (d, J=2.0 Hz, 2H), 1.09 (s, 3H), 0.63-0.59 (m, 2H), 0.43-0.39 (m, 2H) Example 33b RT 0.292 min (Method 7); m/z 391.3 (M+H)+ (ESI+); 1H NMR (400 MHz, DMSO-d6): 12.24 – 11.93 (m, 1H), 10.47 (s, 1H), 8.31 (d, J = 2.0 Hz, 1H), 8.18 (s, 1H), 8.05 (dd, J = 8.4, 2.0 Hz, 1H), 7.38 (d, J = 8.8 Hz, 1H), 2.40 (d, J = 2.8 Hz, 2H), 2.27-2.22 (m, 1H), 1.13 (d, J = 1.6 Hz, 2H), 1.06 (s, 3H), 0.63-0.54 (m, 2H), 0.43-0.36 (m, 2H) Preparation of Intermediate 34.1 2-((cyclopentylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000128_0001
To a mixture of 2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (1.50 g, 5.51 mmol) in MeCN (20 mL) was added 1-cyclopentylmethanamine (1.09 g, 11.0 mmol) and K2CO3 (5.38 g, 16.5 mmol) at 20°C, then the mixture was heated to 100°C and stirred for 16 h. The resulting mixture was diluted with MeCN (20 mL), filtered and the filtrate was concentrated under reduced pressure to afford the product 2- ((cyclopentylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (2.00 g, 5.24 mmol, 95.07 % yield) as a yellow solid. RT 0.344 min (method 5); m/z 429.3 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.70 (t, J = 4.8 Hz, 1H), 8.07 (br, 1H), 7.99 (d, J = 2.0 Hz, 1H), 7.60 (dd, J = 8.8, 2.0 Hz, 1H), 7.53 (s, 1H), 7.32 (br, 1H), 6.81 (d, J = 8.8 Hz, 1H), 3.10 (t, J = 6.8 Hz, 2H), 2.20-2.09 (m, 1H), 1.80-1.71 (m, 2H), 1.66-1.49 (m, 4H), 1.29-1.21 (m, 2H), 1.06 (s, 3H), 0.63-0.57 (m, 2H), 0.38-0.30 (m, 2H). Preparation of Intermediate 34.2 1-(cyclopentylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000128_0002
To a mixture of 2-((cyclopentylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (2.00 g, 5.69 mmol) in DMF (25 mL) was added CDI (14.83 g, 45.5 mmol) and DIEA (7.5 mL, 45.5 mmol) at 20°C, then the mixture was heated to 100°C and stirred for 1h. The resulting mixture was poured into H2O (40 mL), extracted with EtOAc (20 mL, 3x), and the combined organic phase was washed with brine (15 mL, 3x), dried over anhydrous Na2SO4, filtered and the filtrated was concentrated under reduced pressure to give the product 1-(cyclopentylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-quinazoline-6- sulfonamide (1.90 g, 4.88 mmol, 85.75 % yield) as a yellow solid. RT 0.433 min (method 5); m/z 378.1 (M+H)+ (ESI+); 1H NMR (CDCl3, 400 MHz): 8.69 (d, J = 2.0 Hz, 1H), 8.63 (br, 1 H), 8.16 (dd, J = 8.8, 2.0 Hz, 1 H), 7.35 (d, J = 8.8 Hz, 1 H), 5.15 (br, 1H), 4.15-4.11 (m, 2 H), 1.77-1.72 (m, 4 H), 1.61-1.58 (m, 5 H), 1.25 (s, 3H), 0.81-0.76 (m, 2 H), 0.55-0.51 (m, 2 H). Preparation of Intermediate 34.3 3-amino-1-(cyclopentylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline- 6-sulfonamide
Figure imgf000129_0001
To a mixture of 1-(cyclopentylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (500 mg, 1.32 mmol) in dioxane (4 mL) and DMF (4 mL) was added K2CO3 ( 275 mg, 1.99 mmol) at 20 °C, and then the mixture was stirred at 20 °C for 15 min, followed by the addition of O-(2,4-dinitrophenyl)hydroxylamine (264 mg, 1.32 mmol). The mixture was heated to 70 °C and stirred for 16 h. The resulting mixture was poured into H2O (20 mL), extracted with EtOAc (20 mL, 3x), and the combined organic phase was washed with brine (15 mL, 3x), dried over anhydrous Na2SO4, filtered and concentrated under reduced pressure to give a residue, which was purified by reversed-phase flash (ISCO®; 48 g Flash Coulmn Welch Ultimate XB_C1820-40 μm; 120 A, Eluent of 5~95% ACN/H2O (addition of 0.1% formic acid) @ 25 mL/min) to give the product 3-amino-1-(cyclopentylmethyl)-N-(1- methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (310 mg,0.763 mmol, 59.71 % yield) as a yellow solid. RT 0.421 min (method 5); m/z 393.3 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.43 (d, J = 2.4 Hz, 1H), 8.20 (br, 1H), 8.04 (dd, J = 8.8, 2.4 Hz, 1H), 7.75 (d, J = 8.8 Hz, 1H), 5.67 (br, 2H), 4.15 (d, J = 7.6 Hz, 2H), 2.38-2.27 (m, 1H), 1.69-1.61 (m, 4H), 1.53-1.44 (m, 2H), 1.38-1.29 (m, 2H), 1.07 (s, 3H), 0.63-0.55 (m, 2H), 0.42-0.35 (m, 2H). Preparation of Intermediate 34.4 methyl bicyclo[1.1.0]butane-1-carboxylate
Figure imgf000129_0002
To a mixture of methyl 3-chlorocyclobutane-1-carboxylate (70 mg, 0.471 mmol) in THF (1 mL) was added a solution of LiHMDS in THF (1 M, 0.50 mL) at 0°C under N2 atmosphere. The mixture was stirred at 0°C for 1h. The resulting yellow solution of methyl bicyclo[1.1.0]butane-1-carboxylate (theoretical quantity: 52 mg, 1.5 mL) was used in the next step without further purification. Preparation of Example 34 N-(1-(cyclopentylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000130_0001
The reaction was conducted according to the general procedure 1 starting from 3-amino-1- (cyclopentylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide. The residue was purified by preparative HPLC (column: Waters Xbridge 150*25 mm*10 μm; mobile phase: A: 10 mM aqueous solution of NH3.H2O in water, B: MeCN; B%: 36%-66%, 15 min) and lyophilized directly to give the product N-(1-(cyclopentylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide (30 mg, 0.0610 mmol, 96.38 % yield) as a white solid. RT 0.454 min (method 5), m/z, 474.2(M+H)+ (ESl+). 1H NMR (CDCl3, 400 MHz): 8.70 (d, J = 2.4 Hz, 1H), 8.16 (dd, J = 8.8, 2.4 Hz, 1H), 7.94 (br, 1H), 7.35 (d, J = 8.8 Hz, 1H), 5.33 (br, 1H), 3.94-4.34 (m, 2H), 2.53 (s, 2H), 2.28-2.42 (m, 2H), 1.66-1.83 (m, 4H), 1.56 (s, 2H), 1.33-1.46 (m, 2H), 1.27 (s, 5H), 0.69-0.85 (m, 2H), 0.44-0.61 (m, 2H). Preparation of Intermediate 35.1 2-amino-5-bromo-4-fluoro-N-(4-methoxybenzyl)benzamide
Figure imgf000130_0002
To a solution of 2-amino-5-bromo-4-fluorobenzoic acid (16.00 g, 68.37 mmol) andHATU (33.80 g, 88.88 mmol) in DMF (180 mL) was added DIPEA (17.67 g, 136.74 mmol, 23.82 mL), and then the mixture was stirred at 20 °C for 0.5 h. To the mixture was added (4-methoxyphenyl)methanamine (9.38 g, 68.37 mmol, 8.85 mL), and the mixture was stirred at 20 °C for 5 h. The resulting mixture was cooled to 20 °C and poured into ice-water (w/w = 1/1) (300 mL). The mixture was stirred for 10 min, filtered to give a solid, which was dried under reduced pressure to give the product 2-amino-5-bromo-4-fluoro-N-(4- methoxybenzyl)benzamide (22 g, 62.29 mmol, 91.11% yield) as a yellow solid. RT 0.919 min (method 1); m/z 353.0 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz) 8.83 (t, J = 5.6 Hz, 1 H), 7.85 (d, J = 8.0 Hz, 1 H), 7.23 (d, J = 8.4 Hz, 2 H), 6.84-6.95 (m, 4 H), 6.63 (d, J = 11.6 Hz, 1 H), 4.33 (d, J = 5.6 Hz, 2 H), 3.72 (s, 3 H). Preparation of Intermediate 35.2 6-bromo-7-fluoro-3-(4-methoxybenzyl)quinazoline-2,4(1H,3H)-dione
Figure imgf000131_0001
To a mixture of 2-amino-5-bromo-4-fluoro-N-(4-methoxybenzyl)benzamide (22.00 g, 62.29 mmol) and TEA (13.87 g, 137.04 mmol, 19.07 mL) in THF (220 mL) was added bis(trichloromethyl) carbonate (12.38 g, 41.73 mmol) at 0 °C, then the mixture was stirred at 20 °C for 5 h. The resulting mixture was poured into water (400 mL) and extracted with EtOAc (300 mL, 2x). The combined organic layers were washed with brine (200 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure. The residue was triturated with Petroleum ether/Ethyl acetate = 1/1 at 25 °C for 1 h .The mixture was filtered to give a solid, which was dried under vacuum to give the product 6-bromo- 7-fluoro-3-(4-methoxybenzyl)quinazoline-2,4(1H,3H)-dione (15.00 g, 39.56 mmol, 63.51% yield) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz): 11.74 (br, 1 H), 8.15 (d, J = 7.6 Hz, 1 H), 7.27 (d, J = 8.8 Hz, 2 H), 7.07 (d, J = 9.2 Hz, 1 H), 6.86 (d, J = 8.8 Hz, 2 H), 4.98 (s, 2 H), 3.71 (s, 3 H). Preparation of Intermediate 35.3 6-bromo-1-(cyclopropylmethyl)-7-fluoro-3-(4-methoxybenzyl)quinazoline-2,4(1H,3H)-dione
Figure imgf000131_0002
To a solution of 6-bromo-7-fluoro-3-(4-methoxybenzyl)quinazoline-2,4(1H,3H)-dione (8.00 g, 21.10 mmol) in DMF (80 mL) was added Na2CO3 (6.71 g, 63.29 mmol) and (bromomethyl)cyclopropane (5.70 g, 42.20 mmol, 4.04 mL), and then the mixture was stirred at 80 °C for 12 h. The resulting mixture was cooled to room temperature and diluted with H2O (200 mL) before it was extracted with EtOAc (200 mL, 2x). The combined organic phase was washed with brine (200 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated in vacuum to give a residue, which was purified by flash silica gel chromatography (ISCO®; 80 g SepaFlash® Silica Flash Column, Eluent of 0~15% Ethyl acetate/Petroleum ether gradient @ 100 mL/min) to give the product 6-bromo-1-(cyclopropylmethyl)-7- fluoro-3-(4-methoxybenzyl)quinazoline-2,4(1H,3H)-dione (6.00 g, 13.85 mmol, 65.64% yield) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz): 8.26 (d, J = 7.6 Hz, 1 H), 7.74 (d, J = 11.2 Hz, 1 H), 7.28 (d, J = 8.8 Hz, 2 H), 6.86 (d, J = 8.8 Hz, 2 H), 5.05 (s, 2 H), 3.99-4.07 (m, 2 H), 3.70 (s, 3 H), 1.19-1.26 (m, 1 H), 0.37-0.49 (m, 4 H). Preparation of Intermediate 35.4 6-bromo-1-(cyclopropylmethyl)-7-fluoroquinazoline-2,4(1H,3H)-dione
Figure imgf000132_0001
To a solution of 6-bromo-1-(cyclopropylmethyl)-7-fluoro-3-(4-methoxybenzyl)quinazoline- 2,4(1H,3H)-dione (6.00 g, 13.85 mmol) in MeCN (60 mL) and H2O (20 mL) was added CAN (22.78 g, 41.54 mmol, 20.71 mL), and then the mixture was stirred at 20 °C for 12 h. The resulting mixture was diluted with H2O (20 mL) and extracted with EtOAc (20 mL, 2x). The combined organic layer was washed with brine (20 mL, 2x), dried over Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue, which was triturated with EtOAc (5 mL) at 25 °C for 1 h. The mixture was filtered. The cake was collected and dried under vacuum to give the product 6-bromo-1- (cyclopropylmethyl)-7-fluoroquinazoline-2,4(1H,3H)-dione (3.50 g, 11.18 mmol, 80.72% yield) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz): 11.77 (br, 1 H), 8.18 (d, J = 7.6 Hz, 1 H), 7.69 (d, J = 11.2 Hz, 1 H), 3.96 (d, J = 6.8 Hz, 2 H), 1.12-1.28 (m, 1 H), 0.38-0.52 (m, 4 H). Preparation of Intermediate 35.5 6-(benzylthio)-1-(cyclopropylmethyl)-7-fluoroquinazoline-2,4(1H,3H)-dione
Figure imgf000132_0002
To a mixture of 6-bromo-1-(cyclopropylmethyl)-7-fluoroquinazoline-2,4(1H,3H)-dione (3.50 g, 11.18 mmol), phenylmethanethiol (1.67 g, 13.41 mmol, 1.57 mL), Xantphos (1.29 g, 2.24 mmol) and DIEA (2.89 g, 22.36 mmol, 3.89 mL) in dioxane (35 mL) was added Pd2(dba)3 (1.02 g, 1.12 mmol) under nitrogen atmosphere, and then the mixture was stirred at 90 °C for 2 h. The mixture was diluted with H2O (300 mL) and extracted with EtOAc (300 mL, 2x). The combined organic layers were washed with brine (150 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue, which was triturated with petroleum ether/EtOAc = 1/1 at 20 °C for 1 h. The mixture was filtered, and the solid was collected and dried under vacuum to give the product 6-(benzylthio)-1- (cyclopropylmethyl)-7-fluoroquinazoline-2,4(1H,3H)-dione (2.4 g, 6.73 mmol, 60.24% yield) as a yellow solid. RT 0.956 min (method 1); m/z 357.0 (M+H)+ (ES+); 1H NMR (DMSO-d6, 400 MHz): 11.66 (br, 1 H), 7.95 (d, J = 8.0 Hz, 1 H), 7.53 (d, J = 11.6 Hz, 1 H), 7.21-7.35 (m, 5 H), 4.23 (s, 2 H), 3.94 (d, J = 6.8 Hz, 2 H), 1.12-1.22 (m, 1 H), 0.33-0.52 (m, 4 H). Preparation of Intermediate 35.6 1-(cyclopropylmethyl)-7-fluoro-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonyl chloride
Figure imgf000133_0001
To a solution of 6-(benzylthio)-1-(cyclopropylmethyl)-7-fluoroquinazoline-2,4(1H,3H)-dione (200 mg, 561.15 μmol) in ACN (1 mL), acetic acid (0.04 mL) and H2O (0.04 mL) was added 1,3-dichloro-5,5- dimethylimidazolidine-2,4-dione (221.11 mg, 1.12 mmol) at 0 °C, then the mixture was stirred at 0 °C for 1 h. The mixture was filtered, the solid was collected and dried under reduced pressure to give the product 1-(cyclopropylmethyl)-7-fluoro-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonyl chloride (400 mg, 1.20 mmol, 71.41% yield) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz) 11.62 (s, 1 H), 8.29 (d, J = 8.0 Hz, 1 H), 7.39 (d, J = 11.6 Hz, 1 H), 3.96 (d, J = 6.8 Hz, 2 H), 1.14-1.22 (m, 1 H), 0.34-0.54 (m, 4 H). Preparation of Intermediate 35.7 1-(cyclopropylmethyl)-7-fluoro-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000133_0002
To a solution of 1 -methylcyclopropan-1 -amine (170.72 mg, 1.59 mmol, HCI salt) and TEA (401.43 mg, 3.97 mmol, 552.17 μL) in DCM (5 mL) was added 1-(cyclopropylmethyl)-7-fluoro-2,4-dioxo-1 , 2,3,4- tetrahydroquinazoline-6-sulfonyl chloride (440 mg, 1.32 mmol) at O °C. The mixture was stirred at 20 °C for 2 h. The pH of the resulting mixture was adjusted with HCI (1 N) to pH = 3 and the mixture was extracted with EtOAc (20 mL, 2x). The combined organic layer was washed with brine (20 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 1-(cyclopropylmethyl)-7-fluoro-N-(1 -methylcyclopropyl)-2,4-dioxo-1 ,2,3,4-tetrahydroquinazoline-6- sulfonamide (400 mg, 1.09 mmol, 82.33% yield) as a yellow solid.
RT 0.831 min (method 1); m/z 368.1 (M+H)+ (ES+); 1H NMR (DMSO-ofe, 400 MHz) 11 .88 (br, 1 H), 8.49 (s, 1 H), 8.34 (d, J = 8.0 Hz, 1 H), 7.69 (d, J = 12.4 Hz, 1 H), 3.98 (d, J = 6.8 Hz, 2 H), 1.18-1.22 (m, 1 H), 1.14 (s, 3 H), 0.59-0.67 (m, 2 H), 0.40-0.51 (m, 6 H).
Preparation of Intermediate 35.8
3-amino-1-(cyclopropylmethyl)-7-fluoro-N-(1-methylcyclopropyl)-2, 4-dioxo-1, 2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000134_0001
To a solution of 1-(cyclopropylmethyl)-7-fluoro-N-(1-methylcyclopropyl)-2,4-dioxo-1 , 2,3,4- tetrahydroquinazoline-6-sulfonamide (100.00 mg, 272.19 pmol) in dioxane (1 mL) and DMF (1 mL) was added K2CO3 (56.43 mg, 408.28 pmol), and then the mixture was stirred at 70 °C for 0.5 h. O-(4- nitrophenyl)hydroxylamine (50.34 mg, 326.62 pmol) was added, and the mixture was stirred at 70 °C for 12 h. The resulting mixture was cooled to room temperature, diluted with H2O (20 mL) and extracted with EtOAc (20 mL, 2x). The combined organic phase was washed with brine (20 mL, 2x), dried with anhydrous Na2SO4, filtered and the filtrate was concentrated in vacuum. The residue was purified by flash silica gel chromatography (ISCO®; 4 g SepaFlash® Silica Flash Column, Eluent of 0-50% Ethyl acetate/Petroleum ethergradient @ 30 mL/min) to give the product 3-amino-1-(cyclopropylmethyl)-7-fluoro-N-(1- methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (45 mg, 117.68 pmol, 43.23% yield) as a yellow solid.
RT 0.786 min (method 1); m/z 383.1 (M+H)+ (ESI*)
Preparation of Example 35 N-(1-(cyclopropylmethyl)-7-fluoro-6-(N-(1 -methylcyclopropyl)sulfamoyl)-2,4-dioxo-1 ,4- dihydroquinazolin-3(2H)-yl)acrylamide
Figure imgf000135_0001
To a solution of 3-amino-1 -(cyclop ropy Imethyl) -7-fl uoro-N- (1 -methy Icyclop ropy l)-2, 4-dioxo- 1 ,2,3,4- tetrahydroquinazoline-6-sulfonamide (10 mg, 26.15 pmol) in THF (0.3 mL) was added NaHCOs (8.79 mg, 104.60 pmol) in H2O (0.3 mL), followed by the addition of acryloyl chloride (4.73 mg, 52.30 pmol, 4.26 μL) to the mixture at 0 °C. The mixture was stirred at 0 °C for 1 h. The resulting mixture was diluted with H2O (20 mL) and extracted with EtOAc (20 mL, 2x). The combined organic layer was washed with brine (20 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue, which was purified by preparative HPLC (column: Phenomenex luna C18 150*25 mm* 10 pm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 25%-55%, 10 min) and lyophilized to give the product N-(1 -(cyclopropyl methy l)-7-fl uoro-6- (N- (1 -methy Icyclop ropy l)s u lfamoyl)-2, 4-dioxo- 1 ,4- dihydroquinazolin-3(2H)-yl)acrylamide (2.29 mg, 5.25 pmol, 20.06% yield, 100% purity) as a white solid
RT 0.805 min (method 1); m/z 437.1 (M+H)+ (ESI*); 1H NMR (CDCI3, 400 MHz): 8.83 (d, J = 8.0 Hz, 1 H), 7.33-7.39 (br, 1 H), 7.16 (d, J = 11.2 Hz, 1 H), 6.50-6.60 (m, 1 H), 6.30-6.42 (m, 1 H), 5.98-5.91 (m, 1 H), 5.19 (br, 1 H), 4.05 (d, J = 7.2 Hz, 2H), 1.26 (s, 3H), 1.16-1.21 (m, 1 H), 0.80-0.96 (m, 2H), 0.54-0.66 (m, 6H).
Preparation of Intermediate 36.1
2-((cyclohexylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000135_0002
To a mixture of 2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (1.50 g, 5.51 mmol) in MeCN (20 mL) were added K2CO3 (5.38 g, 16.5 mmol) and cyclohexylmethanamine (0.75 g, 6.61 mmol) at 20°C. The mixture was stirred at 100 °C for 16 h. After cooling, the resulting mixture was acidified with HCI (aq., 1 N) to pH = 3 and extracted with EtOAc (40 mL 3x). The combined organic phase was washed with brine (35 mL 3x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 2-((cyclohexylmethyl)amino)-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (2.00 g, 5.24 mmol, 95.07 % yield) as a yellow solid. Rt 0.484 min (method 5); m/z 366.1 (M+H)+ (ESI+); 1H NMR (DMSO-d6400 MHz): 8.70 (t, J = 5.6 Hz, 1H), 8.06 (br, 1H), 7.98 (d, J = 1.2 Hz, 1H), 7.58 (dd, J = 8.8, 1.2 Hz, 1H), 7.52 (s, 1H), 7.30 (br, 1H), 6.79 (d, J = 8.8 Hz, 1H), 3.03 (t, J = 5.6 Hz, 2H), 1.78-1.68 (m, 4H), 1.59-1.51 (m, 1H), 1.26-1.14 (m, 4H), 1.06 (s, 3H), 1.03-1.96 (m, 2H), 0.62-0.57 (m, 2H), 0.35-0.30 (m, 2H). Preparation of Intermediate 36.2 1-(cyclohexylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000136_0001
To a mixture of 2-((cyclohexylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (2.00 g, 5.47 mmol) in DMF (25 mL) were added CDI (17.06 g, 52.4 mmol) and DIEA (8.7 mL, 52.4 mmol) at 20°C, then the mixture was stirred at 100°C for 1 h. The resulting solution was poured into H2O (30 mL), extracted with EtOAc (30 mL 3x) and the combined organic phase was washed with brine (25 mL 3x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 1-(cyclohexylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide (1.50 g, 3.67 mmol, 95.75% yield) as a yellow solid. Rt 0.453 min (method 5); m/z 392.3 (M+H)+ (ESI+); 1H NMR (CDCl3, 400 MHz): 8.69 (d, J = 2.4 Hz, 1H), 8.16 (dd, J = 8.8, 2.4 Hz, 1H), 8.03 (br, 1H), 7.30 (d, J = 8.8 Hz, 1H), 5.60 (br, 1H), 4.01 (d, J = 7.2 Hz, 2H), 1.81-1.83 (m, 1H), 1.73 (m, 6H), 1.27 (s, 3H), 1.24-1.19 (m, 4H), 0.84-0.80 (m, 2H), 0.54-0.52 (m, 2H). Preparation of Intermediate 36.3 3-amino-1-(cyclohexylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide
Figure imgf000137_0001
To a mixture of 1-(cyclohexylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (500 mg, 1.28 mmol) in dioxane (4 mL) and DMF (4 mL) was added K2CO3 (265 mg, 1.92 mmol) at 20°C. The mixture was stirred for 15 min, and then O-(2,4- dinitrophenyl)hydroxylamine (254 mg, 1.28 mmol) was added. The mixture was heated to 70°C and stirred for 16 h. After cooling, the resulting mixture was added to H2O (20 mL), extracted with EtOAc (20 mL 3x). The combined organic layer was washed with brine (15 mL 3x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue, which was purified by reversed-phase flash (ISCO®; 48 g Flash Coulmn Welch Ultimate XB_C1820-40 μm; 120 A, Eluent of 5~95% ACN/H2O (addition of 0.1% formic acid) @ 25 mL/min) to give the product 3-amino-1- (cyclohexylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (310 mg,0.763 mmol, 59.71 % yield) as a yellow solid. 1H NMR (DMSO-d6, 400 MHz): 8.43 (d, J = 2.4 Hz, 1H), 8.20 (br, 1H), 8.04 (dd, J = 8.8, 2.4 Hz, 1H), 7.73 (d, J = 8.8 Hz, 1H), 5.66 (s, 2H), 4.05 (d, J = 7.2 Hz, 2H), 1.84-1.75 (m, 1H), 1.73-1.59 (m, 6H), 1.16-1.12 (m, 4H), 1.09 (s, 3H), 0.63-0.58 (m, 2H), 0.42-0.38 (m, 2H). Preparation of Example 36 N-(1-(cyclohexylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin- 3(2H)-yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000137_0002
The reaction was conducted according to the general procedure 1 starting from 3-amino-1- (cyclohexylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide. The residue was purified by prep-HPLC (column: Waters Xbridge 150*25 mm 5 μm; mobile phase: A: 10 mM aqueous solution of NH3.H2O in water, B: MeCN; B%: 36%-66%, 15 min) to give the product N-(1- (cyclohexylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)- yl)bicyclo[1.1.0]butane-1-carboxamide (14.53 mg, 0.0286 mmol, 16.64 % yield, 95.94% purity) as a white solid. Rt 0.454 min (method 5); m/z 487.3 (M+H)+ (ESI+), 1H NMR (DMSO-d6, 400 MHz): 10.57 (br, 1H), 8.41 (d, J = 2.4 Hz, 1H), 8.26 (br, 1H), 8.10 (dd, J = 8.8, 2.4 Hz, 1H), 7.75 (d, J = 8.8 Hz, 1H), 4.03 (d, J = 6.8 Hz, 2H), 2.40 (d, J = 3.2 Hz, 2H), 2.25 (t, J = 3.2 Hz, 1H), 1.82-1.74 (m, 1H), 1.71-1.56 (m, 6H), 1.19-1.11 (m, 6H), 1.09 (s, 3H), 0.63-0.59 (m,2H), 0.43-0.39 (m, 2H). Preparation of Intermediate 37.1 2-fluoro-4-methylbenzamide
Figure imgf000138_0001
To a solution of 2-fluoro-4-methylbenzoic acid (3.00 g, 19.46 mmol) in THF (30 mL) was added CDI (3.79 g, 23.36 mmol) and DIEA (3.77 g, 29.19 mmol) and the mixture was stirred at 20 °C for 2 h. To the mixture was added NH3·H2O (10.92 g, 87.25 mmol, 28% purity) at 0 °C. The mixture was stirred at 20 °C for 0.5 h. The reaction mixture was diluted with H2O (250 mL) and extracted with EtOAc (100 mL, 3x). The combined organic phase was washed with brine (100 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 2-fluoro-4-methyl-benzamide (1.90 g, 12.41 mmol, 63.74% yield) as a white solid. RT 0.293 min (method 4); m/z 153.9 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 7.68-7.44 (m, 3H), 7.20-7.00 (m, 2H), 2.34 (s, 3H). Preparation of Intermediate 37.2 5-carbamoyl-4-fluoro-2-methylbenzenesulfonyl chloride
Figure imgf000138_0002
A solution of 2-fluoro-4-methylbenzamide (1.90 g, 12.41 mmol) in HSO3Cl (8.67 g, 74.44 mmol) was stirred at 140 °C for 2 h. The reaction mixture was poured to 500 g ice slowly andextracted with EtOAc (200 mL, 3x). The combined organic phase was washed with brine (200 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 5-carbamoyl-4-fluoro-2-methylbenzenesulfonyl chloride (2.00 g, 7.95 mmol, 64.06% yield) as a brown solid. RT 0.398 min (method 4); m/z 251.7 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.04 (d, J = 8.0 Hz, 1H), 7.65-7.42 (m, 2H), 7.06 (d, J = 11.2 Hz, 1H), 2.53 (s, 3H). Preparation of Intermediate 37.3 2-fluoro-4-methyl-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000139_0001
To a solution of 5-carbamoyl-4-fluoro-2-methylbenzenesulfonyl chloride (430 mg, 1.71 mmol) in THF (6 mL) and H2O (5 mL) was added NaHCO3 (1.29 g, 15.38 mmol), followed by the addition of 1- methylcyclopropan-1-amine hydrochloride (367.64 mg, 3.42 mmol, HCl salt) at 0 °C. The mixture was stirred at 0 °C for 1 h. The reaction mixture was diluted with water (60 mL) and extracted with EtOAc (30 mL, 3x). The combined organic phase was washed with brine (30 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 2-fluoro-4-methyl-5- (N-(1-methylcyclopropyl)sulfamoyl)benzamide (350 mg, 1.22 mmol, 71.54% yield) as a brown solid. RT 0.350 min (method 4); m/z 286.9 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.22 (s, 1H), 8.19 (d, J = 7.6 Hz, 1H), 7.76 (br, 2H), 7.38 (d, J = 11.2 Hz, 1H), 2.56 (s, 3H), 1.09 (s, 3H), 0.63-0.52 (m, 2H), 0.43-0.34 (m, 2H). Preparation of Intermediate 37.4 2-((cyclopropylmethyl)amino)-4-methyl-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000139_0002
To a solution of 2-fluoro-4-methyl-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (350.00 mg, 1.22 mmol) in MeCN (6 mL) was added K2CO3 (337.90 mg, 2.44 mmol) and cyclopropylmethanamine (173.88 mg, 2.44 mmol), then the mixture was stirred at 100 °C for 16 h. The reaction mixture was diluted with H2O (60 mL) and extracted with EtOAc (30 mL, 3x). The combined organic phase was washed with brine (30 mL, 3x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a residue, which was purified by reversed-phase flash (ISCO®; 48 g Flash Coulmn Welch Ultimate XB_C1820-40 μm; 120 A, Eluent of 28~35% ACN/H2O (addition of 0.1% FA) @70 mL/min). The product solution was concentrated under reduced pressure to remove MeCN, then extracted with ethyl acetate (30 mL, 3x). The combined organic phase was washed with brine (20 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 2-((cyclopropylmethyl)amino)-4-methyl-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (200 mg, 592.70 μmol, 48.49% yield) as a brown solid. RT 0.432 min (method 4); m/z 337.8 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.57 (t, J = 4.8 Hz, 1H), 8.04 (s, 1H), 7.92 (br, 1H), 7.64 (s, 1H), 7.19 (br, 1H), 6.58 (s, 1H), 3.04 (t, J = 6.4 Hz, 2H), 2.44 (s, 3H), 1.12-1.08 (m, 1H), 1.07 (s, 3H), 0.59-0.55 (m, 2H), 0.54-0.48 (m, 2H), 0.33-0.29 (m, 2H), 0.27- 0.22 (m, 2H). Preparation of Intermediate 37.5 1-(cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline- 6-sulfonamide
Figure imgf000140_0001
To a solution of 2-((cyclopropylmethyl)amino)-4-methyl-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (120.00 mg, 355.62 μmol) in DMF (3 mL) were added CDI (461.31 mg, 2.84 mmol) and DIEA (367.69 mg, 2.84 mmol), and the mixture was stirred at 100 °C for 3 h. The reaction was diluted with water (30 mL) and extracted with EtOAc (15 mL, 3x). The combined organic phase was washed with HCl (aq., 0.5 N, 10 mL, 2x), followed by washing with brine (10 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 1-(cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (120 mg, 330.19 μmol, 92.85% yield) as a brown solid RT 0.442 min (method 4); m/z 363.9 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 11.75 (br, 1H), 8.43 (s, 1H), 8.23 (s, 1H), 7.57 (s, 1H), 4.00 (d, J = 7.2 Hz, 2H), 2.66 (s, 3H), 1.25- 1.21 (m, 1H), 1.11 (s, 3H), 0.60-0.55 (m, 2H), 0.50-0.42 (m, 4H), 0.41-0.35(m, 2H). Preparation of Intermediate 37.6 3-amino-1-(cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000140_0002
To a solution of 1-(cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (50.00 mg, 137.58 μmol) in DMF (1 mL) and dioxane (1 mL) was added K2CO3 (38.03 mg, 275.16 μmol). The mixture was stirred at 70 °C for 0.5 h, then to the mixture was added O-(2,4-dinitrophenyl)hydroxylamine (54.79 mg, 275.16 μmol). The mixture was stirred at 70 °C for 16 h. The residue was diluted with H2O (50 mL) and extracted with EtOAc (25 mL, 3x). The combined organic phase was washed with brine (20 mL, 3x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give the product 3-amino-1-(cyclopropylmethyl)- 7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6-sulfonamide (35 mg, 92.48 μmol, 67.22% yield) as a yellow solid. RT 0.433 min (method 4); m/z 378.9 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 8.50 (s, 1H), 8.26 (br, 1H), 7.63 (s, 1H), 5.64 (br, 2H), 4.11 (d, J = 6.8 Hz, 2H), 2.67 (s, 3H), 1.28-1.23 (m, 1H), 1.11 (s, 3H), 0.60-0.54 (m, 2H), 0.51-0.45 (m, 4H), 0.42-0.35 (m, 2H). Preparation of Example 37 N-(1-(cyclopropylmethyl)-7-methyl-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acrylamide
Figure imgf000141_0001
To a solution of 3-amino-1-(cyclopropylmethyl)-7-methyl-N-(1-methylcyclopropyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (30.00 mg, 79.27 μmol) in THF (1 mL) and H2O (1 mL) was added NaHCO3 (59.93 mg, 713.45 μmol), followed by the addition of prop-2-enoyl chloride (14.35 mg, 158.54 μmol) at 0 °C. tThen the mixture was stirred at 0 °C for 10 min. The resulting mixture was diluted with H2O (10 mL) and extracted with EtOAc (5 mL, 3x). The combined organic phase was washed with brine (5 mL, 2x), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated under reduced pressure to give a crude product, which was purified by prep-HPLC (column: Phenomenex luna C18 150*25 mm* 10 μm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 22%-52%, 8 min). The product solution was lyophilized to give the product N-(1-(cyclopropylmethyl)-7-methyl-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)acrylamide (7.87 mg, 17.81 μmol, 22.46% yield, 97.85% purity) as a yellow solid. RT 0.478 min (method 4); m/z 432.9 (M+H)+ (ESI+); 1H NMR (DMSO-d6, 400 MHz): 10.96 (br, 1H), 8.49 (s, 1H), 8.33 (br, 1H), 7.69 (s, 1H), 6.56-6.39 (dd, J = 16.8, 10.4 Hz,, 1H), 6.28 (dd, J = 16.8, 1.6 Hz, 1H), 5.87 (dd, J = 10.4, 1.6 Hz, 1H), 4.10 (d, J = 6.8 Hz, 2H), 2.70 (s, 3H), 1.31-1.23 (m, 1H), 1.12 (s, 3H), 0.65-0.53 (m, 2H), 0.53-0.44(m, 4H), 0.44-0.38 (m, 2H). Preparation of Intermediate 38.1 5-bromo-4-chloro-2-fluorobenzamide
Figure imgf000142_0001
To a solution of 5-bromo-4-chloro-2-fluorobenzoic acid (10.00 g, 39.46 mmol) in DMF (80 mL) was added NH4Cl (2.32 g, 43.40 mmol), DIEA (15.30 g, 118.37 mmol, 20.62 mL) and HATU (22.50 g, 59.18 mmol), then the reaction was stirred at 20 °C for 16 h. The resutling mixture was poured into water (500 mL) andextracted with EtOAc (300 mL; 2x). The combined organic layer was washed with brine (500 mL), dried over Na2SO4, filtered and the filtrate was concentrated to give a residue, which was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0~40% Ethyl acetate/Petroleum @ 100 mL/min) to give the product 5-bromo-4-chloro-2-fluorobenzamide (8.5 g, 33.67 mmol, 85.33% yield) as a brown solid. RT 0.593 min (method 1); m/z 253.7 (M+H)+ Preparation of Intermediate 38.2 5-(benzylthio)-4-chloro-2-fluorobenzamide
Figure imgf000142_0002
To a solution of 5-bromo-4-chloro-2-fluorobenzamide (8.50 g, 33.67 mmol) in dioxane (100 mL) was added DIEA (8.70 g, 67.34 mmol, 11.73 mL), Xantphos (974.03 mg, 1.68 mmol), Pd2(dba)3 (1.54 g, 1.68 mmol) and BnSH (4.60 g, 37.03 mmol, 4.34 mL) under N2, then the mixture was stirred at 90 °C for 4 h under N2. The reaction mixture was poured into water (500 mL) and extracted with EtOAc (300 mL, 2x). The combined organic layer was washed with brine (500 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated to give a residue. The residue was purified by flash silica gel chromatography (ISCO®; 120 g SepaFlash® Silica Flash Column, Eluent of 0~60% Ethyl acetate/Petroleum @ 100 mL/min) to give the product 5-(benzylthio)-4-chloro-2-fluorobenzamide (8.00 g, 27.05 mmol, 80.34% yield) as a yellow solid. RT 0.823 min (method 1); m/z 296.1 (M+H)+, 1H NMR (DMSO-d6, 400 MHz): 7.81 (br, 1H), 7.70 (br, 1H), 7.66 (d, J = 6.0 Hz, 1H), 7.60 (d, J = 10.0 Hz, 1H), 7.38-7.25 (m, 5H), 4.28 (s, 2H). Preparation of Intermediate 38.3 5-carbamoyl-2-chloro-4-fluorobenzenesulfonyl chloride
Figure imgf000143_0001
To a solution of 5-(benzylthio)-4-chloro-2-fluorobenzamide (6.00 g, 20.28 mmol) in H2O (48 mL) and AcOH (12 mL) was added NCS (8.12 g, 60.84 mmol) at 0 °C, and then the reaction mixture was stirred at 0 °C for 0.1 h. The reaction mixture was poured into NaHCO3 (aq., sat., 120 mL) and extracted with EtOAc (120 mL, 3x). The combined organic layer was washed with brine (120 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated to give the product 5-carbamoyl-2-chloro-4- fluorobenzenesulfonyl chloride (3.00 g, 11.03 mmol, 54.35% yield) as a white solid. RT 0.514 min (method 1); m/z 271.9 (M+H)+ Preparation of Intermediate 38.4 4-chloro-2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000143_0002
To a solution of 1-methylcyclopropan-1-amine hydrochloride (941.02 mg, 13.23 mmol, HCl salt) in THF (30 mL) was added and aqeuous, saturated solution of NaHCO3 (10 mL) and 5-carbamoyl-2-chloro- 4-fluorobenzenesulfonyl chloride (3.00 g, 11.03 mmol) at 0 °C and the mixture was stirred at 0 °C for 1 h. The reaction mixture was diluted with H2O (50 mL) and extracted with EtOAc (50mL, 2x). The combined organic layer was washed with brine (20 mL, 2x), dried over anhydrous Na2SO4 and concentrated under reduced pressure to give a residue, which was purified by by flash silica gel chromatography (ISCO®; 60 g SepaFlash® Silica Flash Column, Eluent of 0~60% Ethyl acetate/Petroleum @ 80 mL/min) to give the product 4-chloro-2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (1.3 g, 4.24 mmol, 38.44% yield) as a white solid. RT 0.457 min (method 1); m/z 307.0 (M+H)+, 1H NMR (DMSO-d6, 400 MHz): 8.54 (s, 1H), 8.27(d, J = 6.0 Hz, 1H), 7.93 (m, 2H), 7.85-7.80 (d, J = 10.0 Hz, 1H), 1.13 (s, 3H), 0.66-0.57 (m, 2H), 0.46-0.41 (m, 2H). Preparation of Intermediate 38.5. 4-chloro-2-((cyclopropylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide
Figure imgf000144_0001
To a solution of 4-chloro-2-fluoro-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (650.00 mg, 2.12 mmol) and cyclopropylmethanamine (150.71 mg, 2.12 mmol) in MeCN (7 mL) was added K2CO3 (585.73 mg, 4.24 mmol), and the mixture was stirred at 100 °C for 4 h. The resulting mixture was poured into water (100 mL) and extracted with EtOAc (60 mL, 2x). The combined organic layer was washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated to give a residue, which was purified by preparative TLC (Petroleum ether : Ethyl acetate=1:1) to give the product 4-chloro- 2-((cyclopropylmethyl)amino)-5-(N-(1-methylcyclopropyl)sulfamoyl)benzamide (500 mg, 1.40 mmol, 65.94% yield) as a yellow solid RT 0.585 min (method 1); m/z 358.0 (M+H)+, 1H NMR (DMSO-d6, 400 MHz): 8.67 (br, 1H), 8.22 (br, 1H), 8.19 (s, 1 H), 7.37 (br, 1H), 6.78 (s, 1H), 3.05 (d, J = 6.8 Hz, 2H), 1.10 (s, 3H), 1.09 -1.07 (m, 1H), 0.65-0.57 (m, 2H), 0.54-0.45 (m, 2H), 0.40-0.31 (m, 2H), 0.30-0.14 (m, 2H). Preparation of Intermediate 38.6 7-chloro-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline- 6-sulfonamide
Figure imgf000144_0002
To a solution of 4-chloro-2-((cyclopropylmethyl)amino)-5-(N-(1- methylcyclopropyl)sulfamoyl)benzamide (250.00 mg, 698.61 μmol) in DMF (3 mL) was added CDI (339.84 mg, 2.10 mmol) and DIEA (270.87 mg, 2.10 mmol, 365.05 μL), and the mixture was stirred at 100 °C for 4 h. The resulting mixture was poured into water (10 mL) and extracted with EtOAc (20 mL, 2x). The combined organic layer was washed with brine (20 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated to give a residue, which was purified by preparative TLC (Petroleum ether : Ethyl acetate=1:1) to give the product 7-chloro-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (260 mg, 677.35 μmol, 96.96% yield) as a white solid. RT 0.550 min (method 1); m/z 384.0 (M+H)+, 1H NMR (DMSO-d6, 400 MHz): 11.95 (br, 1H), 8.50 (s, 1H), 8.45 (s, 1H), 7.81 (s, 1H), 4.02 (d, J = 6.8 Hz, 2H), 1.18-1.16 (m, 1H), 1.14 (s, 3H), 0.64-0.60 (m, 2H), 0.50-0.46 (m, 2H), 0.44-0.41 (m, 4H). Preparation of Intermediate 38.7 3-amino-7-chloro-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide
Figure imgf000145_0001
To a solution of 7-chloro-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4- tetrahydroquinazoline-6-sulfonamide (50.00 mg, 130.26 μmol) and O-(2,4-dinitrophenyl)hydroxylamine (31.12 mg, 156.31 μmol) in DMF (0.3 mL) and dioxane (0.3 mL) was added K2CO3 (27.00 mg, 195.39 μmol), and the mixture was stirred at 70 °C for 3 h. The reaction mixture was poured into water (100 mL) and extracted with DCM (60 mL, 2x). The combined organic layers were washed with brine (100 mL), dried over anhydrous Na2SO4, filtered and the filtrate was concentrated to give a residue. The residue was purified by preparative TLC (Petroleum ether : Ethyl acetate=1:1) to give the product 3-amino-7- chloro-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo-1,2,3,4-tetrahydroquinazoline-6- sulfonamide (45.00 mg, 112.82 μmol, 86.61% yield) as a white solid. RT 0.544 min (method 1); m/z 399.1 (M+H)+. Preparation of Example 38 N-(7-chloro-1-(cyclopropylmethyl)-6-(N-(1-methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4- dihydroquinazolin-3(2H)-yl)acrylamide
Figure imgf000145_0002
To a solution of 3-amino-7-chloro-1-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-2,4-dioxo- 1,2,3,4-tetrahydroquinazoline-6-sulfonamide (45.00 mg, 112.82 μmol) in THF (0.25 mL) and H2O (0.25 mL) was added NaHCO3 (18.96 mg, 225.64 μmol) andprop-2-enoyl chloride (10.21 mg, 112.82 μmol, 9.20 μL) at 0 °C, and the mixture was stirred at 0 °C for 15 min. The reaction mixture was concentrated under reduced pressure to remove the solvent. The residue was purified by preparative HPLC (column: Phenomenex C1875*30 mm*3 μm; mobile phase: A: 0.225% formic acid in water, B: MeCN; B%: 30%- 60%, 7min) and lyophilized directly to give the product N-(7-chloro-1-(cyclopropylmethyl)-6-(N-(1- methylcyclopropyl)sulfamoyl)-2,4-dioxo-1,4-dihydroquinazolin-3(2H)-yl)acrylamide (7.58 mg, 16.72 μmol, 14.82% yield, 99.9% purity) as a white solid. RT 0.562 min (method 1); m/z 453.2 (M+H)+, 1H NMR (DMSO-d6, 400 MHz): 11.01 (br, 1H), 8.56 (s, 1H), 8.55 (br, 1H), 7.94 (s, 1H), 6.45 (dd, J = 17.2, 10.4 Hz, 1H), 6.29 (d, J = 17.2, 2.0 Hz, 1H), 5.87 (d, J = 10.4, 2.0 Hz, 1H), 4.12 (d, J = 6.8 Hz, 2H), 1.26-1.21 (m, 1H), 1.15 (s, 3H), 0.70-0.57 (m, 2H), 0.52-0.42 (m, 6H). Compounds listed in the table below were prepared accordingly to closely related compounds and starting from the corresponding intermediates
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Preparation of Intermediate 77.1 4-(cyclopropanecarbonyl)-1 -oxo-1 , 2-dihydroisoquinoline-7-sulfonyl chloride
Figure imgf000163_0001
To a solution of 7-(benzylthio)-4-(cyclopropanecarbonyl)isoquinolin-1 (2H)-one ( 2.60 g, 7.75 mmol) in MeCN (3.5 mL) (preparation reported in WO2016092326) was added acetic acid (1.8 mL, 31.0 mmol) and H2O (0.56 mL, 31.0 mmol) at 0°C followed by 1,3-dichloro-5,5-dimethylimidazolidine-2, 4-dione ( 2.29 g, 11.6 mmol) in batches. Then, the reaction mixture was stirred at 0 °C for 15 min and the precipitate was filtered. The filter cake was washed with MeCN (1 mL, 2x) and dried under vacuum to give the product 4-(cyclopropanecarbonyl)-1 -oxo-1 , 2-dihydroisoquinoline-7-sulfonyl chloride (2.40 g, 7.70 mmol, 99.32 % yield) as a white solid, which was used for the next step directly without purification.
RT 0.510 min (Method 1 ); m/z 311 .9 (M+H)+ (ESI +) .
Preparation of Intermediate 77.2
4-(cyclopropanecarbonyl)-N-(1 -methylcyclopropyl)-1 -oxo-1 ,2-dihydroisoquinoline-7-sulfonamide
Figure imgf000163_0002
To a solution of 1-methylcyclopropan-1 -amine hydrochloride (1.66 g, 15.4 mmol, HCI salt) in NaHC03 (aq, sat., 16 mL) and THF (10 mL) was added dropwise 4-(cyclopropanecarbonyl)-1-oxo-1,2- dihydroisoquinoline-7-sulfonyl chloride (2.40 g, 7.70 mmol) in THF (14 mL) at 0 °C. The resulting mixture was stirred at 25 °C for 1 h, then poured into water (40 mL) and extracted with EtOAc (40 mL; 2x). The combined organic layer was washed with brine (40 mL; 2x), dried over anhydrous Na2SO4, filtered and concentrated. The crude product was triturated with MeOH (5 mL) at room temperature for 10 min and filtered. The filter cake was collected, dried under reduced pressure to give the product 4- (cyclopropanecarbonyl)-N-(1-methylcyclopropyl)-1 -oxo-1 , 2-dihydroisoquinoline-7-sulfonamide (710 mg, 2.05 mmol, 26.62 % yield) as a white solid. RT 0.474 min (Method 1); m/z 347.1 (M+H)+ (ESI+); 1 HNMR (DMSO-d6, 400 MHz): 12.32 (s, 1H), 8.89 (d, J = 8.8 Hz, 1H), 8.64 (d, J = 2.0 Hz, 1H), 8.46 (s, 1H), 8.24 (s, 1H), 8.09 (dd, J = 8.8, 2.0 Hz, 1H), 2.80-2.67 (m, 1H), 1.04 (s, 3H), 1.03-1.01 (m, 2H), 1.00-0.96 (m, 2H), 0.61-0.55 (m, 2H), 0.41-0.36 (m, 2H) Preparation of Intermediate 77.3 4-(cyclopropyl(hydroxy)methyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7- sulfonamide
Figure imgf000164_0001
To a solution of 4-(cyclopropanecarbonyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7- sulfonamide (710 mg, 2.05 mmol)) in EtOH (7 mL) was added sodium borohydride (155 mg, 4.10 mmol) in batches at 0 °C under N2. The mixture was stirred at 25°C for 2 h, then quenched with H2O (15 mL) and filtered. The filter cake was collected, triturated in MeOH (2 mL) at room temperature for 10 min filtered and dried under vacuum to give the product 4-(cyclopropyl(hydroxy)methyl)-N-(1- methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonamide (330 mg,0.890 mmol, 43.44 % yield) as a light yellow solid. RT 0.419 min (Method 1); m/z 349.2 (M+H)+ (ESI+); 1 HNMR (DMSO-d6, 400 MHz): 8.64 (d, J = 2.0 Hz, 1H), 8.22 (d, J = 8.8 Hz, 1H), 8.03 (dd, J = 2.0, 8.8 Hz, 1H), 7.33 (s, 1H), 5.23 (s, 1H), 4.26 (d, J = 7.2 Hz, 1H), 1.36-1.24 (m, 1H), 1.05 (s, 3H), 0.61-0.24 (m, 8H). Preparation of Intermediate 77.4 4-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonamide
Figure imgf000164_0002
To a solution of 4-(cyclopropyl(hydroxy)methyl)-N-(1-methylcyclopropyl)-1-oxo-1,2- dihydroisoquinoline-7-sulfonamide (280 mg, 0.804 mmol) in DCM (2.8 mL) was added triethylsilane (0.51 mL, 3.21 mmol) followed by addition of TFA (0.24 mL, 3.21 mmol) dropwise over 5 min. The reaction mixture was stirred at 20°C for 1 h, then quenched with water (15 mL) and extracted with EtOAc (15 mL; 2x). The combined organic layer was washed with brine (15 mL; 2x), dried over anhydrous Na2SO4, filtered and concentrated. The crude product was triturated in MeOH (3 mL) at 20°C for 15 min and filtered. The filter cake was collected and dried under reduced pressure to give the product 4-(cyclopropylmethyl)- N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonamide (250 mg, 0.752 mmol, 93.58 % yield) as a white solid. RT 0.485 min (Method 1); m/z 333.1 (M+H)+ (ESI+); 1 HNMR (DMSO-d6, 400 MHz): 11.51 (s, 1H), 8.64 (d, J = 2.0 Hz, 1H), 8.22 (s, 1H), 8.05 (dd, J = 2.0, 8.0 Hz, 1H), 7.96 (d, J = 8.0 Hz, 1H), 7.23 (s, 1H), 2.59 (d, J = 7.0 Hz, 2H), 1.05 (s, 3H), 1.04-1.02 (m, 1H), 0.60-0.56 (m, 2H), 0.53-0.48 (m, 2H), 0.40-0.36 (m, 2H), 0.23-0.18 (m, 2H). Preparation of Intermediate 77.5 2-amino-4-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7- sulfonamide
Figure imgf000165_0001
To a solution of 4-(cyclopropylmethyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7- sulfonamide (30 mg, 0.0902 mmol) in DMF (0.5 mL) was added (aminooxy)diphenylphosphine oxide (42 mg, 0.180 mmol) and t-BuOK (20 mg, 0.180 mmol). The mixture was stirred at 25 °C for 1 h, then poured into water (10 mL) and extracted with EtOAc (10 mL; 2x). The combined organic layer was washed with brine (10 mL; 2x), dried over anhydrous Na2SO4, filtered and concentrated. The residue was purified by preparative TLC (PE:EtOAc=1:2) to give the product 2-amino-4-(cyclopropylmethyl)-N-(1- methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonamide (10 mg, 0.0268 mmol, 29.66 % yield) as a white solid. RT 0.475 min (Method 1); m/z 348.2 (M+H)+ (ESI+). Preparation of Intermediate 77 N-(4-(cyclopropylmethyl)-7-(N-(1-methylcyclopropyl)sulfamoyl)-1-oxoisoquinolin-2(1H)- yl)bicyclo[1.1.0]butane-1-carboxamide
Figure imgf000166_0001
The reaction was conducted according to the general procedure 1 starting from 2-amino-4- (cyclopropylmethyl)-N-(1-methylcyclopropyl)-1-oxo-1,2-dihydroisoquinoline-7-sulfonamide.The residue was purified by preparative HPLC (column:Waters Xbridge 150*25 mm* 5 μm; mobile phase: A: 10 mM aqueous solution of NH4HCO3, B: MeCN; B%: 32%-62%, 10 min) and lyophilized to give the product N- (4-(cyclopropylmethyl)-7-(N-(1-methylcyclopropyl)sulfamoyl)-1-oxoisoquinolin-2(1H)- yl)bicyclo[1.1.0]butane-1-carboxamide (5.8 mg, 0.0128 mmol, 44.30 % yield) as a white solid. RT 0.508 min (Method 1); m/z 428.2 (M+H)+ (ESI+); 1 HNMR (CDCl3, 400 MHz): 9.13 (s, 1H), 9.01 (d, J = 2.0 Hz, 1H), 8.15 (dd, J = 2.0, 8.8 Hz, 1H), 7.78 (d, J = 8.8 Hz, 1H), 6.10 (s, 1H), 2.62 (d, J = 6.4 Hz, 2H), 2.59 (d, J = 3.2 Hz, 2H), 2.36-2.34 (m, 1H), 1.32 (d, J = 3.2 Hz, 2H), 1.26 (s, 3H), 1.09-0.98 (m, 1H), 0.88-0.82 (m, 2H), 0.68-0.57 (m, 2H), 0.55-0.49 (m, 2H), 0.28-0.21 (m, 2H) The following Table 1 provides an overview on the compounds described in the example section:
Figure imgf000166_0002
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Yin
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Biological evaluation of the exemplary compounds
Exemplary compounds of formula (I) were tested in selected biological and/or physicochemical assays one or more times. When tested more than once, data are reported as either average values or as median values, wherein the average value, also referred to as the arithmetic mean value, represents the sum of the values obtained divided by the number of times tested, and the median value represents the middle number of the group of values when ranked in ascending or descending order. If the number of values in the data set is odd, the median value is the middle value. If the number of values in the data set is even, the median is the arithmetic mean of the two middle values. The in vitro pharmacological, pharmacokinetic and physicochemical properties of the compounds can be determined according to the following assays and methods.
PARG protein expression and purification
A codon optimized gene encoding human PARG (448-976 [H446G, L447S, L473S, N479S, S802A, R81 1 K, M841 I, S858P, 1916T, T924D, D927K, C963S, A967T]) was synthesized by Genscript, and cloned into pET15b (Ncol/BamHI) with an N-terminal, Thrombin protease cleavable 6His-TwinStrep tag. Expression of the protein in E. coli BL21 (DE3) was induced by addition of 0.2 mM IPTG to a shake flask culture grown to GD600=0.8 at 37°C. Growth was allowed to continue at 30°C for a further 20 hours before harvesting by centrifugation and storage of the cell pellet at -80°C.
Protein was purified by IMAC and SEC: frozen cell pellets (typically 40 g wet weight) were resuspended by homogenization in 5 volumes buffer A (25 mM Tris/HCI pH 8.0, 200 mM NaCI, 2 mM DTT), supplemented with 1 mg of DNase I from bovine pancreas (Sigma-Aldrich) and protease inhibitors (Roche Complete™ EDTA-free protease inhibitor tablet), and lysed by passage through a Constant Systems BasicZ homogenizer. The lysate was clarified by centrifugation for 60 minutes at 25,000 g, 4°C, and the lysate supernatant was loaded onto 5 ml StrepTrap HP (Cytiva) pre-equilibrated with buffer A. The column was washed with buffer A (~ 10 CV), then buffer B containing 1 M KCI (~5 CV), and then the protein was eluted with buffer A containing 2.5 mM d-Desthiobiotin. Pooled fractions containing 6HisTwinStrep-TEV-hPARG were incubated with TEV protease overnight at 4°C. hPARG was separated from uncleaved material and Thrombin protease through gel filtration with Superdex75 sizing column (GE Healthcare) pre-equilibrated with SEC buffer (15 mM Tris/HCI pH 8.5, 100 mM NaCI, 2 mM DTT). Pooled fractions containing pure hPARG were concentrated using a 10 k MWCO spin concentrator (VivaSpin) to 10 mg/mL, and then either used immediately for crystallisation or snap-frozen in liquid nitrogen for storage at -80°C.
PARG C872A protein expression and purification
A codon optimized gene encoding human PARG C872A (448-976 [L473S, N479S, S802A, R81 1 K, M8411, S858P, C872A, 1916T, T924D, D927K, C963S, A967T]) was synthesized by Genscript, and cloned into pET15b (Ncol/BamHI) with an N-terminal, Thrombin protease cleavable 6His-TwinStrep tag. Expression of the protein in E. coli BL21 (DE3) was induced by addition of 0.2 mM IPTG to a shake flask culture grown to GD600 = 0.8 at 37°C. Growth was allowed to continue at 30°C for a further 20 hours before harvesting by centrifugation and storage of the cell pellet at -80°C.
Protein was purified by IMAC and SEC: frozen cell pellets (typically 40 g wet weight) were resuspended by homogenization in 5 volumes buffer A (25 mM Tris/HCI pH 8.0, 200 mM NaCI, 2 mM DTT), supplemented with 1 mg of DNase I from bovine pancreas (Sigma-Aldrich) and protease inhibitors (Roche Complete™ EDTA-free protease inhibitor tablet), and lysed by passage through a Constant Systems BasicZ homogenizer. The lysate was clarified by centrifugation for 60 minutes at 25,000 g, 4°C, and the lysate supernatant was loaded onto 5 ml StrepTrap HP (Cytiva) pre-equilibrated with buffer A.
The column was washed with buffer A (~10 CV), then buffer B containing 1 M KCI (~5 CV), and then the protein was eluted with buffer A containing 2.5 mM d-Desthiobiotin. Pooled fractions were incubated with TEV protease overnight at 4°C. hPARG C872A was separated from uncleaved material and Thrombin protease through gel filtration with Superdex75 sizing column (GE Healthcare) preequilibrated with SEC buffer (15 mM Tris/HCI pH 8.5, 100 mM NaCI, 2 mM DTT). Pooled fractions containing pure hPARG C872A were concentrated using a 10 k MWCO spin concentrator (VivaSpin) to 10 mg/mL, and then snap-frozen in liquid nitrogen for storage at -80°C.
PARG enzymatic I Cso assay
PARG enzyme as incubated with compound or vehicle (DMSO) for 15 minutes or 2 hours in a 384 well plate. After adding the PARG substrate ADP-ribose-pNP, the plate was read for absorbance intensity at 405 nm. The vehicle (DMSO) with high absorbance intensity represents no inhibition of enzymatic reaction while the low control (no enzyme) with low absorbance intensity represents full inhibition of enzymatic reaction.
Materials: hPARG: Peak Protein, 30 nM
Substrate: ADP-pNP, 800 pM, Jena Bioscience catalog # NU-955
Reaction time: 60 minutes
Assay buffer: 50 mM Tris-HCI pH 8.0, 100 mM NaCI, 2 mM DTT
Temperature: 30 °C
Total volume: 30 μL
Controls:
• 0% inhibition control: DMSO
• 100% inhibition control: No enzyme
The protocol that was used for enzyme reaction and detection is as follows:
1. Transfer 100 nL of the final concentration of test compounds or vehicle (DMSO) to the appropriate wells of a microtiter plate.
2. Centrifuge the plate at 1000 rpm for 1 minute.
3. Transfer 14.6 μL of 2x final concentration of enzyme in assay buffer or assay buffer alone to the appropriate wells.
4. Centrifuge the plate at 1000 rpm for 1 minute.
5. Incubate the plate at room temperature for 15 minutes.
6. Transfer 15.4 μL of 2x substrate in assay buffer to all the test wells.
7. Centrifuge the plate at 1000 rpm for 1 minute.
8. Read the plate on a plate reader (e.g., Spark Tecan).
The Absorbance ICso value of compounds of Formula (I) in Examples 1 to 85 are provided in Table 2 below.
Cellular PAR chain assay
The ability of compounds to inhibit PARG in response to DNA damage, was assessed with U2OS cells pretreated with the compounds for 1 hour, following a 1 -hour treatment with or without the DNA alkylating agent temozolomide (TMZ). The cells were harvested and fixed in 70% ethanol, rehydrated with glucose and EDTA in PBS and subsequently blocked for 1 hour with PBS 1 % BSA and 0.01% Tween-20 (PBT). The cells were incubated for 2 hours at room temperature with a mouse monoclonal antibody against poly (ADP) ribose (PAR) polymer. The cells were washed and incubated with an anti-mouse Alexa-488 conjugated secondary antibody for 1 hour at room temperature. Propidium iodide staining was used to determine DNA content in the cells (staining at 4°C overnight). The fluorescence intensity of the cells was assessed by flow cytometry (Cytoflex from Beckmann) and the percentage of PAR chain positive cells (gated in relation to TMZ+DMSO treated control) was determined. PAR chain positive cells % were fit against the concentration of the compound using a 4 parameter log-logistic function, generating PAR chain ECso values:
Figure imgf000186_0001
The PAR chain ECso value for compounds of Formula (I) in Examples 1 to 85 are provided in Table 2 below.
Cellular Viability Assay
NCIH-460 as a PARG-inhibition sensitive cell line and U2OS as PARG-inhibition insensitive cell line were plated at 1000 cells/well and 2000 cells/well, respectively, in 96-well white plates with clear flat bottom. After 24 hours, the compounds were added with the Tecan digital dispenser (D300e) in duplicates. The outer wells of the plate were excluded. After 96 hours of incubation, 150 pl of the growth medium were removed and 50 pl of Cell Titer-Gio (Promega) were added per well. Following an incubation of 10 minutes, luminescence was read using a plate reader (Tecan). Averaged values of the samples were normalized to DMSO treated control samples. Curves were fit as % of the control vs. log of the compound concentration using a 4 parameter log-logistic function:
Figure imgf000186_0002
The PARGi (NCIH-460 and U2OS) cellular viability ECso values for compounds of Formula (I) in Examples 1 to 85 are provided in Table 2 below.
Table 2: Inhibition of PARG by compounds according to the present invention and cellular activity of compounds according to the present invention. The ICso (inhibitory concentration at 50% of maximal effect) values are indicated in pM, empty space means that the corresponding compounds have not been tested in the respective assay.
(T) Example number
@ Structure @ IC50 in pM determined in PARG enzymatic assay (PARG protein and 15 mn incubation) described under PARG enzymatic IC50 assay
(4) IC50 in pM determined in PARG enzymatic assay (PARG protein and 2 hours incubation) described under PARG enzymatic IC50 assay.
(5) I C50 in pM determined in PARG enzymatic assay (PARG C872A protein and 2 hours incubation) described under PARG enzymatic IC50 assay.
@ EC50 in pM determined in cellular assay as described under Cellular PAR chain assay (conditions with treatment of TMZ).
(7) EC50 in pM determined in cellular assay as described under Cellular PAR chain assay (conditions without treatment of TMZ).
(8) EC50 in pM determined in NCIH-460 cells as described under Cellular viability assay.
(9) EC50 in pM determined in U2OS cells as described under Cellular viability assay.
Figure imgf000187_0001
Figure imgf000188_0001
Figure imgf000189_0001
Figure imgf000190_0001
Further assays
Kinetic solubility assay
The Kinetic solubility assay employs the shake flask method followed by HPLC-UV analysis. For exemplary compounds, the kinetic solubility was measured according to the following protocol:
1 ) Samples were weighed and dissolved in 100% DMSO to make a stock solution of 10 mM. About 100 μL of stock solution is needed to cover this assay.
2) Test compounds and controls (10 mM in DMSO, 10 μL/tube) were added into the buffer (490 μL/well) which placed in a Minni-Uniprep filter. The buffer was prepared as the customer’s requirement.
3) Vortex the kinetic solubility samples for 2 minutes.
4) Incubate and shake the solubility solutions on an orbital shaker for 24 hr at room temperature
5) Transfer 200 μL each of solubility solution into 96-deep well for analysis when the samples were directly filtered by the syringeless filter device
6) Determine the test compound concentration of the filtrate using HPLC-UV.
7) Injected three UV standard solutions into HPLC from low to high concentration, followed by testing of the K.S. supernatant. Testing samples are injected in duplicate.
Bidirectional permeability in Caco2
The bidirectional permeability in Caco-2 cells assay was performed for the exemplary compounds of formula (I) according to the following protocol:
1. Caco-2 cells purchased from ATCC were seeded onto polyethylene membranes (PET) in 96- well BD Insert plates at 1 x 105 cells/ cm2, and refreshed medium every 4~5 days until to the 21st to 28th day for confluent cell monolayer formation.
2. The integrity of the monolayer is verified by performing Lucifer yellow rejection assay.
3. The quality of the monolayer is verified by measuring the Unidirectional (A— >B) permeability of fenoterol/nadolol (low permeability marker), propranolol/metopronolol (high permeability marker) and Bi-directional permeability of Digoxin (a P-glycoprotein substrate marker) in duplicate wells.
4. Standard assay conditions for test compounds:
-Test concentration: 2 pM (DMS0<1 %);
-Replicates: n=2;
-Directions: bi-directional transport including A^B and B^A;
-Incubation time: single time point, 2hours;
-Transport buffer: HBSS containing 10 mM HEPES, pH7.40±0.05;
-Incubation condition: 37±1 °C, 5% CO2, relatively saturated humidity.
5. Spike dosing solution and mix with transport buffer and Stop Solution (containing an appropriate internal standard (IS)) as TO sample.
6. At the end of incubation, sample solutions from both donor and receiver wells and mix with Stop Solution immediately.
7. All samples including TO samples, donor samples and receiver samples are analyzed using LC/MS/MS. Concentrations of test compound are expressed as peak area ratio of analytes versus IS without a standard curve.
Microsome metabolic stability (MMS) assay
The stability of the exemplary compounds was measured in the microsome metabolic stability assay as follows:
1) Test compounds will be incubated at 37°C with liver microsomes (pooled from multiple donors) at 1 pM in the presence of a NADPH regenerating system at 0.5 mg/ml microsomal protein.
2) Positive controls include Testosterone (3A4 substrate), Propafenone (2D6) and Diclofenac (2C9). They will be incubated with microsomes in the presence of a NADPH regenerating system.
3) Time samples (0, 5, 15, 30, 45 and 60 minutes) will be removed, immediately mixed with cold acetonitrile containing internal standard (IS). Test compound incubated with microsomes without NADPH regenerating system for 60min will be also included.
4) Single point for each test condition (n=1).
5) Samples will be analyzed by LC/MS/MS; disappearance of test compound will be assessed base on peak area ratios of analyte/l S(no standard curve).
6) An excel data summary, calculated intrinsic clearance and t1/2 values will be provided.
7) Using the following equation to calculate the microsome clearance:
, int(mic) = 0.693/half life/mg microsome protein per mLwt: 40 g/kg, 30 g/kg, 32 g/kg, 20 g/kg and
88 g/kg for rat, monkey, dog, human and mouse.CLint(mic) to calculate the whole the liver clearance: microsomal protein / g liver weight: 45 mg/g for 5 speciesint(liver) = CLint(mic) * mg microsomal protein/g liver weight * g liver weight/kg body weight .
In vitro metabolic stability of test compounds in CD-1 mouse, SD rat, beagle dog, cynomolqus monkey and human cryopreserved hepatocytes
1 . Test compound (at 1 pM) is incubated with cryopreserved hepatocytes (0.5 x 106 cells per mL) in duplicates (n=2) at 37°C using 96-well plate format.
2) Time points are 0, 15, 30, 60 and 90 minutes in separate plates and medium control samples without cells at 0 and 90 minutes are also incubated. At each time point the reaction will be stopped by adding organic solution containing internal standard (IS).
3. Positive controls 7-ethoxycoumarin and 7-hydroxycoumarin are included in parallel.
4. Samples are analyzed by LC-MS/MS. Disappearance of test compound is assessed based on peak area ratios of analyte/IS (no standard curve).

Claims

New PCT-Patent Application based on US 63/320,873 and US 63/391,166 FoRx Therapeutics AG Vossius Ref.: AE3522 PCT BS Claims 1. A compound of formula:
Figure imgf000193_0001
wherein: Rcov is selected from C2 alkenyl, C2 alkynyl, -CH2Cl, -CH2CN, and
Figure imgf000193_0002
wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(C1-4 alkyl), -CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -OOC-(C1-4 alkyl), -NHCO-(C1-4 alkyl), -(C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3, wherein said alkynyl is optionally substituted with an optional substituent selected from C1-4 alkyl, cycloalkyl, heterocycloalkyl, aryl and heteroaryl, and wherein the -CH2- group in said -CH2Cl and the -CH2- group in said -CH2CN are each optionally substituted with one or more optional substituents selected from -Hal, C1-4 alkyl and -CF3; -Wcov- is selected from -CO-, -SO- and -SO2-; RN is selected from hydrogen and C1-4 alkyl; R1 is hydrogen, chloro, fluoro, -CN, formyl, C1-2 alkyl, C2 alkenyl, C2 alkynyl, C1-2 haloalkyl, -(C1-2 alkylene)-OH or -(C1-2 alkylene)-O-(C1-2 alkyl); R2 and R3 are independently each C1-2 alkyl or C1-2 haloalkyl, or R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F; or -CR1R2R3 is bicyclo[1,1,1]pent-1-yl; W is selected from -NHS(O)y-, -S(O)yNH-, -NHS(O)(NH)-, -NHS(O)(N-C1-2 alkyl)-, -S(O)(NH)-NH-, -S(O)(N-C1-2 alkyl)-NH-, wherein y is 1 or 2; X1 and X3 are independently selected from the group consisting of N, CH, and CF; X2 is N or C-YC2-RC2, wherein YC2 is selected from a covalent bond, C1-8 alkylene, C2-8 alkenylene, C2-8 alkynylene, cycloalkylene and heterocycloalkylene wherein said alkylene, said alkenylene and said alkynylene are each optionally substituted with one or more groups independently selected from RS1, and further wherein one or more -CH2- units comprised in said alkylene, said alkenylene or said alkynylene are each optionally replaced by a group independently selected from -O-, -NH-, -N(C1-5 alkyl)-, -CO-, -S-, -SO-, and -SO2-, and wherein said cycloalkylene and heterocycloalkylene are each optionally substituted with one or more groups independently selected RS2; and wherein RC2 is selected from hydrogen, halo, -OH, -NH2, -SH, -CN, C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl; wherein said alkyl, alkenyl, and alkynyl in RC2 are each optionally substituted with one or more groups independently selected from RS1, and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in RC2 are each optionally substituted with one or more groups independently selected from RS2; and the moiety represented with a partial formula is a moiety selected from
Figure imgf000195_0001
Figure imgf000195_0002
wherein: R7 is hydrogen, -CN, -Hal, or a moiety of the formula
Figure imgf000195_0003
wherein: L71 is a bond or C1-5 alkylene optionally substituted with halo or oxo; L72 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH- , -CON(C1-6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, - NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, -SO2N(C1-6 alkyl)-, - NHSO2-, or -N(C1-6 alkyl)SO2-, and Q7 is hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, -(C0-2 alkylene)-cycloalkyl, -(C0-2 alkylene)-aryl, -(C0-2 alkylene)-heterocycloalkyl or -(C0-2 alkylene)-heteroaryl; wherein said alkyl, alkenyl, alkynyl, and alkylene in R7 are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R7 are each optionally substituted with one or more optional substituents selected from RS2; R8 is hydrogen, -CN, -Hal, or a moiety of the formula:
Figure imgf000196_0001
wherein: L81 is a bond, C1-5 alkylene optionally substituted with halo or oxo; L82 is a bond, -O-, -S-, -SO-, -SO2-, -NH-, -N(C1-6 alkyl)-, -CO-, -COO-, -OCO-, -CONH- , -CON(C1-6 alkyl)-, -NHCO-, N(C1-6 alkyl)CO-, -NHCONH-, -N(C1-6 alkyl)CONH-, - NHCON(C1-6 alkyl)-, N(C1-6 alkyl)CON(C1-6 alkyl)-, -SO2NH-, -SO2N(C1-6 alkyl)-, - NHSO2-, or -N(C1-6 alkyl)SO2-; and Q8 is hydrogen, -CN, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, cycloalkyl, aryl, heterocycloalkyl or heteroaryl; wherein said alkyl, alkenyl, alkynyl, and alkylene in R8 are each optionally substituted with one or more optional substituents selected from RS1; and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl in R8 are each optionally substituted with one or more optional substituents selected from RS2; wherein RS1 is selected from halogen, -CN, -OH, -O(C1-5 alkyl), -O(C1-5 haloalkyl), C1-5 haloalkyl, -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO- (C1-5 alkyl), -N(C1-5 alkyl)-CO-(C1-5 alkyl), -NHCONH2, -NHCONH-(C1-5 alkyl), -NHCON(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 alkyl)CONH2, -N(C1-5 alkyl)CONH-(C1-5 alkyl), and -N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl), and wherein RS2 is selected from halogen, -CN, -OH, C1-5 alkyl, C1-5 haloalkyl, -O(C1-5 alkyl), -O(C1-5 haloalkyl), -SH, -S(C1-5 alkyl), -S(C1-5 haloalkyl), -NH2, -NH(C1-5 alkyl), -NH(C1-5 haloalkyl), -N(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 haloalkyl)(C1-5 alkyl), -(N-heterocycloalkyl), -CO(C1-5 alkyl), -CONH2, -CONH(C1-5 alkyl), -CON(C1-5 alkyl)(C1-5 alkyl), -CO-(N-heterocycloalkyl), -NHCO- (C1-5 alkyl), -N(C1-5 alkyl)-CO-(C1-5 alkyl), -NHCONH2, -NHCONH-(C1-5 alkyl), -NHCON(C1-5 alkyl)(C1-5 alkyl), -N(C1-5 alkyl)CONH2, -N(C1-5 alkyl)CONH-(C1-5 alkyl), -N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-CN, -(C1-5 alkylene)-OH, -(C1-5 alkylene)-O(C1-5 alkyl), -(C1-5 alkylene)-O(C1-5 haloalkyl), -(C1-5 alkylene)-SH, -(C1-5 alkylene)-S(C1-5 alkyl), -(C1-5 alkylene)-S(C1- 5 haloalkyl), -(C1-5 alkylene)-NH2, -(C1-5 alkylene)-NH(C1-5 alkyl), -(C1-5 alkylene)-NH(C1-5 haloalkyl), -(C1-5 alkylene)-N(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)(C1-5 haloalkyl), -(C1-5 alkylene)-(N-heterocycloalkyl), -(C1-5 alkylene)-N(C1-5 haloalkyl)(C1-5 alkyl), -(C1-5 alkylene)-CO(C1- 5 alkyl), -(C1-5 alkylene)-CONH2, -(C1-5 alkylene)-CONH(C1-5 alkyl), -(C1-5 alkylene)-CON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-CO-(N-heterocycloalkyl), -(C1-5 alkylene)-NHCO-(C1-5 alkyl), -(C1- 5 alkylene)-N(C1-5 alkyl)-CO-(C1-5 alkyl), -(C1-5 alkylene)-NHCONH2, -(C1-5 alkylene)-NHCONH-(C1- 5 alkyl), -(C1-5 alkylene)-NHCON(C1-5 alkyl)(C1-5 alkyl), -(C1-5 alkylene)-N(C1-5 alkyl)CONH2, -(C1-5 alkylene)-N(C1-5 alkyl)CONH-(C1-5 alkyl), and -(C1-5 alkylene)-N(C1-5 alkyl)CON(C1-5 alkyl)(C1-5 alkyl); or a pharmaceutically acceptable salt thereof.
2. The compounds of claim 1, wherein Rcov is
Figure imgf000197_0001
3. The compound of claim 1, wherein Rcov is C2 alkenyl, wherein said alkenyl is optionally substituted with one or more optional substituents selected from C1-4 alkyl, -COO-(C1-4 alkyl), - CO-(C1-4 alkyl), -CONH-(C1-4 alkyl), -OOC-(C1-4 alkyl), -NHCO-(C1-4 alkyl), -(C1-4 alkylene)N(C1-4 alkyl)(C1-4 alkyl), -(C1-4 alkylene)-(N-heterocycloalkyl), cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -Hal, -CN and -CF3,.
4. The compound of any one of claims 1 to 3, wherein R1 is methyl or fluoromethyl.
5. The compound of any one of claims 1 to 4, wherein R2 and R3 together with the carbon atom to which they are attached form cyclopropyl optionally substituted with one or more -F.
6. The compound of any one of claims 1 to 5, wherein W is -NHS(O)2-, preferably wherein the left side of W as defined herein is attached to the carbon atom that carries R1, R2 and R3.
7. The compound of any one of claims 1 to 6, wherein X1 is CF or CH and X3 is CH, preferably wherein X1 and X3 are each CH.
8. The compound of any one of claims 1 to 7, wherein X2 is C-YC2-RC2, wherein -YC2-RC2 is selected from -O-C1-12 alkyl, -NH-C1-12 alkyl, -N(C1-5 alkyl)-C1-12 alkyl, -O-C2-12 alkenyl, -NH-C2-12 alkenyl, - N(C1-5 alkyl)-C2-12 alkenyl, -O-C2-12 alkynyl, -NH-C2-12 alkynyl, -N(C1-5 alkyl)-C2-12 alkynyl, -(C0-3 alkylene)-cycloalkyl, -CO-(C0-3 alkylene)-cycloalkyl, -(C0-3 alkylene)-CO-cycloalkyl, -CONH-(C0-3 alkylene)-cycloalkyl, (C0-3 alkylene)-CONH-cycloalkyl, -NHCO-(C0-3 alkylene)-cycloalkyl, -(C0-3 alkylene)-NHCO-cycloalkyl, -NH-(C0-3 alkylene)-cycloalkyl, -(C0-3 alkylene)-NH-cycloalkyl, -O-(C0-3 alkylene)-cycloalkyl, -(C0-3 alkylene)-O-cycloalkyl, -SO2-(C0-3 alkylene)-cycloalkyl, -(C0-3 alkylene)-SO2-cycloalkyl, -CONH-cycloalkyl, -NHCO-cycloalkyl, -NH-cycloalkyl, -O-cycloalkyl, - CO-cycloalkyl, -SO2-cycloalkyl, -(C0-3 alkylene)-heterocycloalkyl, -CO-(C0-3 alkylene)- heterocycloalkyl, -(C0-3 alkylene)-CO-heterocycloalkyl, -CONH-(C0-3 alkylene)-heterocycloalkyl, - (C0-3 alkylene)-CONH-heterocycloalkyl, -NHCO-(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)- NHCO-heterocycloalkyl, -NH-(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)-NH- heterocycloalkyl, -O-(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)-O-cycloalkyl, -SO2-(C0-3 alkylene)-heterocycloalkyl, -(C0-3 alkylene)-SO2-heterocycloalkyl, -CONH-heterocycloalkyl, - NHCO-heterocycloalkyl, -NH-heterocycloalkyl, -O-heterocycloalkyl, -CO-heterocycloalkyl, -SO2- heterocycloalkyl, -(C0-3 alkylene)-aryl, -CO-(C0-3 alkylene)-aryl, -(C0-3 alkylene)-CO-aryl, -CONH- (C0-3 alkylene)-aryl, -(C0-3 alkylene)-CONH-aryl, -NHCO-(C0-3 alkylene)-aryl, -(C0-3 alkylene)- NHCO-aryl, -NH-(C0-3 alkylene)-aryl, -(C0-3 alkylene)-NH-aryl, -O-(C0-3 alkylene)-aryl, -(C0-3 alkylene)-O-aryl, -SO2-(C0-3 alkylene)-aryl, -(C0-3 alkylene)-SO2-aryl, -CONH-aryl, -NHCO-aryl, - NH-aryl, -O-aryl, -CO-aryl, -SO2-aryl, -(C0-3 alkylene)-heteroaryl, -CO-(C0-3 alkylene)-heteroaryl, - (C0-3 alkylene)-CO-heteroaryl, -CONH-(C0-3 alkylene)-heteroaryl, -(C0-3 alkylene)-CONH- heteroaryl, -NHCO-(C0-3 alkylene)-heteroaryl, -(C0-3 alkylene)-NHCO-heteroaryl, -NH-(C0-3 alkylene)-heteroaryl, -(C0-3 alkylene)-NH-heteroaryl, -O-(C0-3 alkylene)-heteroaryl, -(C0-3 alkylene)- O-heteroaryl, -SO2-(C0-3 alkylene)-heteroaryl, -(C0-3 alkylene)-SO2-heteroaryl, -CONH-heteroaryl, - NHCO-heteroaryl, -NH-heteroaryl, -O-heteroaryl, -CO-heteroaryl and -SO2-heteroaryl, wherein said alkylene is optionally substituted with one or more groups independently selected from RS1, and wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more groups independently selected from RS2. The compound of any one of claims 1 to 7, wherein X2 is CH. The compound of any one of claims 1 to 9, wherein the moiety represented with a partial formula
Figure imgf000199_0001
The compound of claim 10, wherein R8 is a moiety of the formula -L81-L82-Q8. The compound of claim 11 , wherein L81 is methylene, -L82 is a covalent bond, and wherein Q8 is cycloalkyl, aryl, heterocycloalkyl or heteroaryl, wherein said cycloalkyl, heterocycloalkyl, aryl and heteroaryl are each optionally substituted with one or more optional substituents selected from RS2. The compound of any one of claims 1 to 12, wherein RN is H and/or Woov is -CO-. The compound of claim 1 , selected from:
Figure imgf000199_0002
Figure imgf000200_0001
Figure imgf000201_0001
pharmaceutically acceptable salt thereof. A pharmaceutical composition comprising the compound of any one of claims 1 to 14 or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. The compound of any one of claims 1 to 14 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 15, for use in therapy. The compound for use or the pharmaceutical composition for use of claim 16, for use in a method of treating a disease or disorder in which PARG activity is implicated. The compound for use or the pharmaceutical composition for use of claim 16, for use in a method of treating a proliferative disorder, preferably wherein the proliferative disorder is cancer, preferably a human cancer.
PCT/EP2023/056968 2022-03-17 2023-03-17 2,4-dioxo-1,4-dihydroquinazoline derivatives as parg inhibitors for the treatment of cancer WO2023175184A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US202263320873P 2022-03-17 2022-03-17
US63/320,873 2022-03-17
US202263391166P 2022-07-21 2022-07-21
US63/391,166 2022-07-21

Publications (1)

Publication Number Publication Date
WO2023175184A1 true WO2023175184A1 (en) 2023-09-21

Family

ID=85795351

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/056968 WO2023175184A1 (en) 2022-03-17 2023-03-17 2,4-dioxo-1,4-dihydroquinazoline derivatives as parg inhibitors for the treatment of cancer

Country Status (1)

Country Link
WO (1) WO2023175184A1 (en)

Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
WO1999002166A1 (en) 1997-07-08 1999-01-21 Angiogene Pharmaceuticals Ltd. Use of colchinol derivatives as vascular damaging agents
WO2000040529A1 (en) 1999-01-07 2000-07-13 Angiogene Pharmaceuticals Ltd. Colchinol derivatives as vascular damaging agents
WO2000041669A2 (en) 1999-01-15 2000-07-20 Angiogene Pharmaceuticals Ltd. Benzimidazole vascular damaging agents
WO2000047212A1 (en) 1999-02-10 2000-08-17 Astrazeneca Ab Quinazoline derivatives as angiogenesis inhibitors
WO2001092224A1 (en) 2000-05-31 2001-12-06 Astrazeneca Ab Indole derivatives with vascular damaging activity
WO2001094341A1 (en) 2000-06-06 2001-12-13 Astrazeneca Ab Quinazoline derivatives for the treatment of tumours
WO2002004434A1 (en) 2000-07-07 2002-01-17 Angiogene Pharmaceuticals Limited Colchinol derivatives as vascular damaging agents
WO2002008213A1 (en) 2000-07-07 2002-01-31 Angiogene Pharmaceuticals Limited Colchinol derivatives as angiogenesis inhibitors
US20040106802A1 (en) 2002-04-08 2004-06-03 Torrent Pharmaceuticals Ltd. Novel compounds and therapeutic uses thereof
WO2008070707A1 (en) 2006-12-05 2008-06-12 Smithkline Beecham Corporation Il-8 receptor antagonists
WO2011138266A1 (en) 2010-05-03 2011-11-10 Evotec Ag Indolizine and imidazopyridine derivatives as orexin receptor antagonists
WO2013006394A1 (en) 2011-07-01 2013-01-10 Institute For Hepatitis And Virus Research Sulfamoylbenzamide derivatives as antiviral agents against hbv infection
US20130203686A1 (en) 2007-06-18 2013-08-08 Huy Ong Azapeptides as cd36 binding compounds
WO2016092326A1 (en) 2014-12-12 2016-06-16 Cancer Research Technology Limited 2,4-dioxo-quinazoline-6-sulfonamide derivatives as inhibitors of parg
WO2016097749A1 (en) 2014-12-19 2016-06-23 Cancer Research Technology Limited Parg inhibitory compounds
WO2016144792A1 (en) 2015-03-06 2016-09-15 Alpharmagen, Llc Alpha 7 nicotinic acetylcholine receptor allosteric modulators, their derivatives and uses thereof
WO2020257940A1 (en) 2019-06-26 2020-12-30 University Health Network Furosemide analogues and compositions and uses thereof for treatment of alzheimer's disease
WO2021055744A1 (en) 2019-09-20 2021-03-25 Ideaya Biosciences, Inc. 4-substituted indole and indazole sulfonamido derivatives as parg inhibitors
WO2021133751A1 (en) 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted quinazolinyl compounds useful as t cell activators

Patent Citations (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
WO1999002166A1 (en) 1997-07-08 1999-01-21 Angiogene Pharmaceuticals Ltd. Use of colchinol derivatives as vascular damaging agents
WO2000040529A1 (en) 1999-01-07 2000-07-13 Angiogene Pharmaceuticals Ltd. Colchinol derivatives as vascular damaging agents
WO2000041669A2 (en) 1999-01-15 2000-07-20 Angiogene Pharmaceuticals Ltd. Benzimidazole vascular damaging agents
WO2000047212A1 (en) 1999-02-10 2000-08-17 Astrazeneca Ab Quinazoline derivatives as angiogenesis inhibitors
WO2001092224A1 (en) 2000-05-31 2001-12-06 Astrazeneca Ab Indole derivatives with vascular damaging activity
WO2001094341A1 (en) 2000-06-06 2001-12-13 Astrazeneca Ab Quinazoline derivatives for the treatment of tumours
WO2002004434A1 (en) 2000-07-07 2002-01-17 Angiogene Pharmaceuticals Limited Colchinol derivatives as vascular damaging agents
WO2002008213A1 (en) 2000-07-07 2002-01-31 Angiogene Pharmaceuticals Limited Colchinol derivatives as angiogenesis inhibitors
US20040106802A1 (en) 2002-04-08 2004-06-03 Torrent Pharmaceuticals Ltd. Novel compounds and therapeutic uses thereof
WO2008070707A1 (en) 2006-12-05 2008-06-12 Smithkline Beecham Corporation Il-8 receptor antagonists
US20130203686A1 (en) 2007-06-18 2013-08-08 Huy Ong Azapeptides as cd36 binding compounds
WO2011138266A1 (en) 2010-05-03 2011-11-10 Evotec Ag Indolizine and imidazopyridine derivatives as orexin receptor antagonists
WO2013006394A1 (en) 2011-07-01 2013-01-10 Institute For Hepatitis And Virus Research Sulfamoylbenzamide derivatives as antiviral agents against hbv infection
WO2016092326A1 (en) 2014-12-12 2016-06-16 Cancer Research Technology Limited 2,4-dioxo-quinazoline-6-sulfonamide derivatives as inhibitors of parg
WO2016097749A1 (en) 2014-12-19 2016-06-23 Cancer Research Technology Limited Parg inhibitory compounds
WO2016144792A1 (en) 2015-03-06 2016-09-15 Alpharmagen, Llc Alpha 7 nicotinic acetylcholine receptor allosteric modulators, their derivatives and uses thereof
WO2020257940A1 (en) 2019-06-26 2020-12-30 University Health Network Furosemide analogues and compositions and uses thereof for treatment of alzheimer's disease
WO2021055744A1 (en) 2019-09-20 2021-03-25 Ideaya Biosciences, Inc. 4-substituted indole and indazole sulfonamido derivatives as parg inhibitors
WO2021133751A1 (en) 2019-12-23 2021-07-01 Bristol-Myers Squibb Company Substituted quinazolinyl compounds useful as t cell activators

Non-Patent Citations (32)

* Cited by examiner, † Cited by third party
Title
"Greene's Protective Groups in Organic Synthesis", 2014, WILEY
ATZRODT J ET AL., BIOORG MED CHEM, vol. 20, no. 18, 2012, pages 5658 - 5667
BOYLES ET AL., EUR. J. MED. CHEM.,ORG. PRO. RES. DEV., vol. 6, 2002, pages 230 - 233
BOYLES ET AL., ORG. PROGRESS. RES. DEV., vol. 6, 2002, pages 230 - 233
CAS, no. 70142-34-6
COHENCHANG, NAT. CHEM. BIOL., vol. 14, 2018, pages 236
COSTANTINO ET AL., SCIENCE, vol. 343, 2014, pages 88
DOMINIC, ORG. PROCESS RES.DEV., vol. 9, 2005, pages 499 - 507
FARMER ET AL., NATURE, vol. 434, 2005, pages 917
FONTANA ET AL., ELIFE, 2017
FUJIHARA ET AL., CURR. CANCER DRUG TARGETS, vol. 9, 2009, pages 953
GOGOLA ET AL., CANCER CELL, vol. 33, 2018, pages 1078
HALAZONETIS ET AL., SCIENCE, vol. 319, 2008, pages 1352
HOUL ET AL., NAT COMMUN, vol. 10, 2019, pages 5654
J. MED. CHEM., vol. 47, 2004, pages 6658 - 6661
JAMES ET AL., ACS CHEM. BIOL., vol. 11, 2016, pages 3179
JEMA ET AL., J. NATL. CANCER INST., vol. 109, 2017, pages 1975
JIANGBUCHWALD: "Metal-Catalyzed Cross-Coupling Reactions", 2004, WILEY-VCH
LIU ET AL., EUR. J. MED. CHEM., vol. 222, 2021, pages 113565
LYER ET AL., CHEM. COMMUNICATIONS, vol. 54, 2018, pages 11021 - 11024
MODVIG A ET AL., J ORG CHEM, vol. 79, 2014, pages 5861 - 5868
NEGRINI ET AL., NAT. REV. MOL. CELL BIOL., vol. 11, 2010, pages 220
O'SULLIVAN ET AL., NAT. COMMUN., vol. 10, 2019, pages 1182
PILLAY ET AL., CANCER CELL, vol. 35, 2019, pages 519
RACK ET AL., GENES DEV, vol. 34, 2020, pages 263
SARI ET AL., EUR. J. MED. CHEM., vol. 138, 2017, pages 407 - 421
SCULLY ET AL., CURR. OPIN. GENET. DEV., vol. 71, 2021, pages 154
STERN ET AL., CRITICAL REVIEWS IN ONCOLOGY/HAEMATOLOGY, vol. 54, 2005, pages 1 1 - 29
WASZKOWYCZ BOHDAN ET AL: "Cell-Active Small Molecule Inhibitors of the DNA-Damage Repair Enzyme Poly(ADP-ribose) Glycohydrolase (PARG): Discovery and Optimization of Orally Bioavailable Quinazolinedione Sulfonamides", JOURNAL OF MEDICINAL CHEMISTRY, vol. 61, no. 23, 7 November 2018 (2018-11-07), US, pages 10767 - 10792, XP055945870, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.8b01407 *
WASZKOWYCZ ET AL., J. MED. CHEM., vol. 61, 2018, pages 10767
WILLIAM JS ET AL., JOURNAL OF LABELLED COMPOUNDS AND RADIOPHARMACEUTICALS,, vol. 53, no. 11-12, 2010, pages 635 - 644
ZEMANCIMPRICH, NAT. CELL BIOL., vol. 16, 2013, pages 2

Similar Documents

Publication Publication Date Title
US10995073B2 (en) PARG inhibitory compounds
CN110506043B (en) Compounds useful as RET inhibitors
JP5188988B2 (en) Benzazole derivatives, compositions, and methods of use as Aurora kinase inhibitors
CN102171214B (en) Dihydropyridophthalazinone inhibitors of poly(ADP-ribose)polymerase (PARP)
US10239843B2 (en) 2,4-dioxo-quinazoline-6-sulfonamide derivatives as inhibitors of PARG
KR101738866B1 (en) Cyclic N,N&#39;-diarylthioureas and N,N&#39;-diarylureas as androgen receptor antagonists, anti-cancer agent, method for producing and using same
JP5508257B2 (en) Arylamidopyrimidone compounds
KR20100137564A (en) Pyridazinone derivatives
JP5509070B2 (en) Arylamidopyrimidone derivatives for the treatment of neurodegenerative diseases
WO2019089835A1 (en) Diazanaphthalen-3-yl carboxamides and preparation and use thereof
WO2012059932A1 (en) 2, 4 -diaminopyrimidine derivatives as protein kinase inhibitors
KR20130116070A (en) Heterocyclic compounds, their preparation and their therapeutic application
WO2020150545A1 (en) Pyrazole derivatives as modulators of the wnt/b-catenin signaling pathway
CA2841897A1 (en) Novel compound having parp inhibitory activity
WO2023057389A1 (en) Parg inhibitory compounds
WO2023175184A1 (en) 2,4-dioxo-1,4-dihydroquinazoline derivatives as parg inhibitors for the treatment of cancer
WO2023175185A1 (en) 2,4-dioxo-1,4-dihydroquinazoline derivatives as parg inhibitors for the treatment of cancer
WO2023057394A1 (en) N,n-dimethyl-4-(7-(n-(1-methylcyclopropyl)sulfamoyl)-imidazo[1,5-a]pyridin-5-yl)piperazine-1-carboxamide derivatives and the corresponding pyrazolo[1,5-a]pyridine derivatives as parg inhibitors for the treatment of cancer
CN111868060A (en) Dihydropyridophthalazinone derivative, preparation method and application thereof
WO2024074497A1 (en) Parg inhibitory compound
KR20210048701A (en) Novel pyridinyltriazine derivatives as protein kinase inhibitors and compositions for preventing, improving or treating cancer containing the same
MX2008009811A (en) Benzazole derivatives, compositions, and methods of use as aurora kinase inhibitors

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: 23714476

Country of ref document: EP

Kind code of ref document: A1