US20220298142A1 - Heterocyclic compounds as adenosine antagonists - Google Patents

Heterocyclic compounds as adenosine antagonists Download PDF

Info

Publication number
US20220298142A1
US20220298142A1 US17/684,626 US202217684626A US2022298142A1 US 20220298142 A1 US20220298142 A1 US 20220298142A1 US 202217684626 A US202217684626 A US 202217684626A US 2022298142 A1 US2022298142 A1 US 2022298142A1
Authority
US
United States
Prior art keywords
alkylene
optionally substituted
alkyl
halogen
compound
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
US17/684,626
Inventor
Son Minh Pham
Jayakanth Kankanala
Pradeep S. JADHAVAR
Baban Mohan MULIK
Farha KHAN
Sreekanth A. Ramachandran
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nuvation Bio Inc
Nuvation Bio Operating Co LLC
Original Assignee
Nuvation Bio Inc
Nuvation Bio Operating Co LLC
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 Nuvation Bio Inc, Nuvation Bio Operating Co LLC filed Critical Nuvation Bio Inc
Priority to US17/684,626 priority Critical patent/US20220298142A1/en
Assigned to NUVATION BIO OPERATING COMPANY LLC reassignment NUVATION BIO OPERATING COMPANY LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: NUVATION BIO OPERATING COMPANY INC.
Assigned to INTEGRAL BIOSCIENCES PVT. LTD. reassignment INTEGRAL BIOSCIENCES PVT. LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: Jadhavar, Pradeep S., KHAN, Farha, MULIK, Baban Mohan, RAMACHANDRAN, Sreekanth A.
Assigned to NUVATION BIO INC. reassignment NUVATION BIO INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NUVATION BIO OPERATING COMPANY LLC (FORMALLY NUVATION BIO OPERATING COMPANY INC., WHICH WAS FORMALLY KNOWN AS NUVATION BIO INC.)
Assigned to NUVATION BIO OPERATING COMPANY INC. (FORMERLY KNOWN AS NUVATION BIO INC.) reassignment NUVATION BIO OPERATING COMPANY INC. (FORMERLY KNOWN AS NUVATION BIO INC.) ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GiraFpharma LLC
Assigned to GiraFpharma LLC reassignment GiraFpharma LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SPARCBIO LLC
Assigned to SPARCBIO LLC reassignment SPARCBIO LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: INTEGRAL BIOSCIENCES PVT. LTD.
Assigned to SPARCBIO LLC reassignment SPARCBIO LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KANKANALA, JAYAKANTH, PHAM, SON MINH
Publication of US20220298142A1 publication Critical patent/US20220298142A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • This disclosure relates generally to therapeutics for treatment mediated through a G-protein-coupled receptor (GPCR) signaling pathway and, more particularly, to compounds that inhibit an adenosine receptor (such as an A 2A antagonist).
  • GPCR G-protein-coupled receptor
  • the disclosure also provides pharmaceutically acceptable compositions comprising such compounds and methods of using the compounds or compositions in the treatment of a disease associated with a GPCR signaling pathway
  • Adenosine receptors are distributed throughout the body and are responsible for numerous biological functions.
  • the seven trans-membrane G-protein-coupled receptors (GPCRs) have been divided into four different subtypes: A 1 , A 2A , A 2B , and A 3 .
  • the A 2A and A 2B ARs stimulate activity of the adenylyl cyclase, inducing an increase of cAMP levels.
  • a 2A ARs have a distinct tissue localization, different biochemical pathways, and specific pharmacological profiles.
  • Adenosine is one of the human body's most important neuromodulators in both the central and the peripheral nervous systems. Adenosine is released from tumor cells and its concentration in the extracellular fluid of tumors can reach immunosuppressive levels (Blay et al. (1997), Cancer Res., 57(13), pp. 2602-5). The extracellular fluid of solid carcinomas contains immunosuppressive concentrations of adenosine. Id. This increase in adenosine concentration is a result of increases in CD73 (ecto-5′-nucleotidase) and CD39 (nucleoside triphosphate dephosphorylase) enzymes, which are responsible for directly catabolizing ATP into adenosine.
  • CD73 ecto-5′-nucleotidase
  • CD39 nucleoside triphosphate dephosphorylase
  • adenosine as an endogenous immunoregulator in cancer pathogenesis: where to go?
  • Adenosine whose concentration increases within hypoxic regions of solid tumors, has been recognized as being able to interfere with the recognition of tumor cells by cytolytic effector cells of the immune system. (Tuite and Riss (2013). Recent developments in the pharmacological treatment of Parkinson's disease. Expert Opin. Investig. Drugs, 12(8) pp 1335-52, Popoli et al. (2002).
  • Blockade of striatal adenosine A 2A receptor reduces, through a presynaptic mechanism, quinolinic acid-induced excitotoxicity: possible relevance to neuroprotective interventions in neurodegenerative diseases of the striatum, J. Neurosci, 22(5) pp. 1967-75, Gessi et al. (2011). Adenosine receptors and cancer. Biochim Biophys Acta, 1808(5), pp. 1400-12).
  • a 2A and A 3 subtypes appear promising targets for therapeutic development.
  • activation of A 2A receptors leads to immunosuppressive effects, which decreases anti-tumoral immunity and thereby encourages tumor growth.
  • the A 2B receptor is another potential target for therapeutic development. Autocrine/paracrine stimulation of A 2B expressed on tumor cells is believed to enhance their metastatic potential and A 2B blockade may reduce tumor metastasis in an immune-independent manner (Beavis et al. (2013). Blockade of A 2A receptors potently suppresses the metabolism of CD73 + Tumors. Proc. Natl. Acad. Sci., 110(36) pp. 14711-6). A 2B expression also correlates with relapse-free survival (RFS) in triple negative breast cancer suggesting that this pathway may be clinically relevant.
  • RFS relapse-free survival
  • a 2B blockade also has the potential to modulate the immunosuppressive properties of tumor-associated immune cells including dendritic cells and myeloid-derived suppressor cells (MDSCs) (Cekic et al. (2011). Adenosine A 2B receptor blockade slows growth of bladder and breast tumors. J. Immunol. 188(1), pp. 198-205; Sorrentino et al. (2015). Myeloid-derived suppressor cells contribute to A 2B adenosine receptor-induced VEGF production and angiogenesis in a mouse melanoma model. Oncotarget 6(29), pp. 27478-89; Iannone et al. (2013). Blockade of A 2B adenosine receptor reduces tumor growth and immune suppression mediated by myeloid-derived suppressor cells in a mouse model of melanoma. Neoplasia, 15(12), pp. 1400-9.
  • MDSCs myeloid-derived suppressor cells
  • the compound of the formula (I), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing is of the formula (II) or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, as detailed herein.
  • a disease such as a proliferative disease
  • the compound of formulae (I), (II) or (III), or a salt thereof is administered to the individual in combination with another therapeutic agent.
  • the compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing is administered to the individual in combination with another therapeutic agent.
  • the compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing is a compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • compositions comprising (A) a compound detailed herein, such as a compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, or a compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, and (B) a pharmaceutically acceptable carrier or excipient.
  • a pharmaceutical compositions comprising (A) a compound detailed herein, such as a compound of formulae (I), (II) or (III), or a salt thereof, and (B) a pharmaceutically acceptable carrier or excipient.
  • Kits comprising a compound detailed herein or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing and instructions for use are also provided. Kits comprising a compound detailed herein or a salt thereof and instructions for use are also provided. A compound detailed herein or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing is also provided for the manufacture of a medicament for the treatment of cancer. Compounds as detailed herein or a pharmaceutically acceptable salt thereof are also provided for the manufacture of a medicament for the treatment of cancer.
  • Alkenyl refers to an unsaturated linear or branched univalent hydrocarbon chain or combination thereof, having at least one site of olefinic unsaturation (i.e., having at least one moiety of the formula C ⁇ C) and having the number of carbon atoms designated (i.e., C 2 -C 10 means two to ten carbon atoms).
  • the alkenyl group may be in “cis” or “trans” configurations, or alternatively in “E” or “Z” configurations.
  • Particular alkenyl groups are those having 2 to 20 carbon atoms (a “C 2 -C 20 alkenyl”), having 2 to 8 carbon atoms (a “C 2 -C 8 alkenyl”), having 2 to 6 carbon atoms (a “C 2 -C 6 alkenyl”), or having 2 to 4 carbon atoms (a “C 2 -C 4 alkenyl”).
  • alkenyl examples include, but are not limited to, groups such as ethenyl (or vinyl), prop-1-enyl, prop-2-enyl (or allyl), 2-methylprop-1-enyl, but-1-enyl, but-2-enyl, but-3-enyl, buta-1,3-dienyl, 2-methylbuta-1,3-dienyl, homologs and isomers thereof, and the like.
  • alkyl refers to and includes saturated linear and branched univalent hydrocarbon structures and combination thereof, having the number of carbon atoms designated (i.e., C 1 -C 10 means one to ten carbons). Particular alkyl groups are those having 1 to 20 carbon atoms (a “C 1 -C 20 alkyl”).
  • alkyl groups are those having 1 to 8 carbon atoms (a “C 1 -C 8 alkyl”), 3 to 8 carbon atoms (a “C 3 -C 8 alkyl”), 1 to 6 carbon atoms (a “C 1 -C 6 alkyl”), 1 to 5 carbon atoms (a “C 1 -C 5 alkyl”), or 1 to 4 carbon atoms (a “C 1 -C 4 alkyl”).
  • alkyl examples include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • Alkylene refers to the same residues as alkyl, but having bivalency. Particular alkylene groups are those having 1 to 6 carbon atoms (a “C 1 -C 6 alkylene”), 1 to 5 carbon atoms (a “C 1 -C 5 alkylene”), 1 to 4 carbon atoms (a “C 1 -C 4 alkylene”) or 1 to 3 carbon atoms (a “C 1 -C 3 alkylene”).
  • alkylene examples include, but are not limited to, groups such as methylene (—CH 2 —), ethylene (—CH 2 CH 2 —), propylene (—CH 2 CH 2 CH 2 —), butylene (—CH 2 CH 2 CH 2 CH 2 —), isopropylene (—CH 2 C(H)(CH 3 )CH 2 —), and the like.
  • Alkynyl refers to an unsaturated linear or branched univalent hydrocarbon chain or combination thereof, having at least one site of acetylenic unsaturation (i.e., having at least one moiety of the formula C ⁇ C) and having the number of carbon atoms designated (i.e., C 2 -C 10 means two to ten carbon atoms).
  • Particular alkynyl groups are those having 2 to 20 carbon atoms (a “C 2 -C 20 alkynyl”), having 2 to 8 carbon atoms (a “C 2 -C 8 alkynyl”), having 2 to 6 carbon atoms (a “C 2 -C 6 alkynyl”), or having 2 to 4 carbon atoms (a “C 2 -C 4 alkynyl”).
  • alkynyl examples include, but are not limited to, groups such as ethynyl (or acetylenyl), prop-1-ynyl, prop-2-ynyl (or propargyl), but-1-ynyl, but-2-ynyl, but-3-ynyl, homologs and isomers thereof, and the like.
  • aryl refers to and includes polyunsaturated aromatic hydrocarbon groups.
  • Aryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings.
  • the aryl group contains from 6 to 14 annular carbon atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, biphenyl, and the like.
  • cycloalkyl refers to and includes cyclic univalent hydrocarbon structures, which may be fully saturated, mono- or polyunsaturated, but which are non-aromatic, having the number of carbon atoms designated (e.g., C 1 -C 10 means one to ten carbons). Cycloalkyl can consist of one ring, such as cyclohexyl, or multiple rings, such as adamantyl, but excludes aryl groups. A cycloalkyl comprising more than one ring may be fused, spiro or bridged, or combinations thereof. A preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 13 annular carbon atoms.
  • a more preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a “C 3 -C 8 cycloalkyl”).
  • cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, norbornyl, and the like.
  • Halo or “halogen” refers to elements of the Group 17 series having atomic number 9 to 85. Preferred halo groups include fluoro, chloro, bromo and iodo. Where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached, e.g., dihaloaryl, dihaloalkyl, trihaloaryl etc. refer to aryl and alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be but are not necessarily the same halo; thus 4-chloro-3-fluorophenyl is within the scope of dihaloaryl.
  • perhaloalkyl An alkyl group in which each hydrogen is replaced with a halo group is referred to as a “perhaloalkyl.”
  • a preferred perhaloalkyl group is trifluoroalkyl (—CF 3 ).
  • perhaloalkoxy refers to an alkoxy group in which a halogen takes the place of each H in the hydrocarbon making up the alkyl moiety of the alkoxy group.
  • An example of a perhaloalkoxy group is trifluoromethoxy (—OCF 3 ).
  • heteroaryl refers to and includes unsaturated aromatic cyclic groups having from 1 to 10 annular carbon atoms and at least one annular heteroatom, including but not limited to heteroatoms such as nitrogen, oxygen and sulfur, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule at an annular carbon or at an annular heteroatom.
  • Heteroaryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings.
  • heteroaryl groups include, but are not limited to, pyridyl, pyrimidyl, thiophenyl, furanyl, thiazolyl, and the like.
  • heteroaryl groups also include, but are not limited to, pyridyl, pyrimidyl, thiophenyl, furanyl, thiazolyl, oxazolyl, isoxazolyl, thiophenyl, pyrrolyl, pyrazolyl, 1,3,4-oxadiazolyl, imidazolyl, isothiazolyl, triazolyl, 1,3,4-thiadiazolyl, tetrazolyl, benzofuranyl, benzothiophenyl, pyrazolopyridinyl, indazolyl, benzothiazolyl, benzooxazolyl or benzoimidazolyl and the like.
  • a heteroaryl containing at least one additional fused ring that is nonaromatic is attached to the parent structure at an annular atom of the additional ring.
  • a heteroaryl containing at least one additional ring that is nonaromatic is attached to the parent structure at an annular atom of the aromatic ring.
  • heterocycle refers to a saturated or an unsaturated non-aromatic group having from 1 to 10 annular carbon atoms and from 1 to 4 annular heteroatoms, such as nitrogen, sulfur or oxygen, and the like, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • a heterocyclyl group may have a single ring or multiple condensed rings, but excludes heteroaryl groups.
  • a heterocycle comprising more than one ring may be fused, spiro or bridged, or any combination thereof. In fused ring systems, one or more of the fused rings can be aryl, cycloalyl or heterocyclyl.
  • heterocyclyl groups include, but are not limited to, tetrahydropyranyl, dihydropyranyl, piperidinyl, piperazinyl, pyrrolidinyl, thiazolinyl, thiazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, 2,3-dihydrobenzo[b]thiophen-2-yl, 4-amino-2-oxopyrimidin-1(2H)-yl, and the like.
  • a heterocyclyl containing at least one additional ring (such as a fused additional ring) that does not contain a heteroatom is attached to the parent structure at an annular atom of the additional ring.
  • a heterocyclyl containing at least one additional ring (such as a fused additional ring) that does not contain a heteroatom is attached to the parent structure at an annular atom of the ring containing a heteroatom.
  • Oxo refers to the moiety ⁇ O.
  • Optionally substituted unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 1, 2, 3, 4 or 5) of the substituents listed for that group in which the substituents may be the same of different.
  • an optionally substituted group has one substituent.
  • an optionally substituted group has two substituents.
  • an optionally substituted group has three substituents.
  • an optionally substituted group has four substituents.
  • an optionally substituted group has 1 to 2, 2 to 5, 3 to 5, 2 to 3, 2 to 4, 3 to 4, 1 to 3, 1 to 4 or 1 to 5 substituents.
  • a “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject.
  • a pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • treatment is an approach for obtaining beneficial or desired results including clinical results.
  • beneficial or desired results include, but are not limited to, one or more of the following: decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of individuals.
  • beneficial or desired results include shrinking a tumor (reducing tumor size); decreasing the growth rate of the tumor (such as to suppress tumor growth); reducing the number of cancer cells; inhibiting, retarding or slowing to some extent and preferably stopping cancer cell infiltration into peripheral organs; inhibiting (slowing to some extent and preferably stopping) tumor metastasis; inhibiting tumor growth; preventing or delaying occurrence and/or recurrence of tumor; and/or relieving to some extent one or more of the symptoms associated with the cancer.
  • beneficial or desired results include preventing or delaying occurrence and/or recurrence, such as of unwanted cell proliferation.
  • “delaying development of a disease” means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease (such as cancer). This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. For example, a late stage cancer, such as development of metastasis, may be delayed.
  • an “effective dosage” or “effective amount” of compound or salt thereof or pharmaceutical composition is an amount sufficient to effect beneficial or desired results.
  • beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity of, or delaying the onset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease.
  • beneficial or desired results include ameliorating, palliating, lessening, delaying or decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival.
  • an effective amount comprises an amount sufficient to cause a tumor to shrink and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation. In some embodiments, an effective amount is an amount sufficient to delay development. In some embodiments, an effective amount is an amount sufficient to prevent or delay occurrence and/or recurrence.
  • an effective amount can be administered in one or more administrations, in the case of cancer, the effective amount of the drug or composition may: (i) reduce the number of cancer cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and preferably stop cancer cell infiltration into peripheral organs; (iv) inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer.
  • An effective dosage can be administered in one or more administrations.
  • an effective dosage of compound or a salt thereof, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. It is intended and understood that an effective dosage of a compound or salt thereof, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • the term “individual” is a mammal, including humans.
  • An individual includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate.
  • the individual is human.
  • the individual (such as a human) may have advanced disease or lesser extent of disease, such as low tumor burden.
  • the individual is at an early stage of a proliferative disease (such as cancer).
  • the individual is at an advanced stage of a proliferative disease (such as an advanced cancer).
  • references to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”.
  • A is 4-hydroxyphenyl optionally further substituted by R 3 , 4-hydroxy-2-pyridyl optionally further substituted by R 4 , a naphthyl substituted by R 4 , a 9- or 10-membered bicyclic heterocyclyl optionally substituted by R 4 , or a 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 ;
  • B is a phenyl optionally substituted by R 3 , C 3 -C 6 cycloalkyl optionally substituted by R 4 , 3- to 6-membered heterocyclyl optionally substituted by R 4 or a 5- to 10-membered heteroaryl optionally substituted by R 4 ;
  • R 1 is a hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 6-membered heterocyclyl, 5- to 10-membered heteroaryl, —(C 1 -C 3 alkylene)(C 3 -C 6 cycloalkyl), —(C 1 -C 3 alkylene)(3-6-membered heterocyclyl), —(C 1 -C 3 alkylene)(5-6-membered heteroaryl), —(C 1 -C 3 alkylene)(C 6 aryl), —C(O)R 1a , —C(O)OR 1a , —C(O)NR 1b R 1c , —NR 1b R 1c , —S(O) 2 R 1a , —(C 1 -C 3 alkylene)C(O)NR 1b R 1c , —
  • each R 1a is independently hydrogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, 3-6-membered heterocyclyl, C 6 aryl, 5-6-membered heteroaryl, —(C 1 -C 3 alkylene)(C 3 -C 6 cycloalkyl), —(C 1 -C 3 alkylene)(3-6-membered heterocyclyl), —(C 1 -C 3 alkylene)(C 6 aryl) or —(C 1 -C 3 alkylene)(5-6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)OC 2 H 5 , —NH 2 or —NHCH 3 ;
  • each R 1b and R 1c is independently hydrogen, C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, 3-6-membered heterocyclyl, C 6 aryl, 5-6-membered heteroaryl, —(C 1 -C 3 alkylene)(C 3 -C 6 cycloalkyl), —(C 1 -C 3 alkylene)(3-6-membered heterocyclyl), —(C 1 -C 3 alkylene)(C 6 aryl) or —(C 1 -C 3 alkylene)(5-6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)OC 2 H 5 , —NH 2 or —NHCH 3 ;
  • R 2 is —OR 2a , —NHR 2b , —C(O)NHR 2b , or C 1 -C 6 alkyl, wherein the C 1 -C 6 alkyl of R 2 is substituted by —OR 2c , —NHR 2c , or —C(O)NHR 2c ;
  • each R 2a and R 2b is independently cyclohexane, 6-membered heterocyclyl, —(C 1 -C 3 alkylene)N(C 2 H 5 ) 2 , —(C 1 -C 3 alkylene)(C 3 -C 6 cycloalkyl), —(C 1 -C 3 alkylene)(3-6-membered heterocyclyl), or —(C 1 -C 3 alkylene)(5-6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)OC 2 H 5 , —NH 2 or —NHCH 3 ;
  • R 2 is 5- or 6-membered heteroaryl, wherein the 5- or 6-membered heteroaryl is further substituted by C 1 -C 6 alkyl optionally substituted by halogen, —OH or oxo;
  • each R 3 is independently C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —OC(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)OR 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , —NR 5 S(O)R 6 , —C(O)NR 5 S(O)R 6 , —NR 5 S(O) 2 R 6 , —C(O)NR 5 S(O) 2 R 6 , —S(O)NR 6 R 7
  • each R 4 is independently oxo or R 3 ;
  • R 5 is independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR 8 , —NR 8 R 9 , —C(O)R 8 , —C(O)OR 8 , —C(O)NR 8 R 9 , —NR 8 C(O)R 9 , —S(O)R 8 , —S(O) 2 R 8 , —S(O) 2 NR 8 R 9 , —NR 8 S(O) 2 R 9 , or C 1 -C 6 alkyl optionally substituted by oxo, —OH or halogen;
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR 8 , —NR 8 R 9 , —C(O)R 8 , —C(O)OR 8 , —C(O)NR 8 R 9 , —NR 8 C(O)R 9 , —S(O)R 8 , —S(O) 2 R 8 , —S(O) 2 NR 8 R 9 , —NR 8 S(O) 2 R 9 , or C 1 -C 6 alkyl optionally substituted by oxo, —OH or halogen;
  • R 6 and R 7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo, —CN, —OR 8 , —NR 8 R 9 , —C(O)R 8 , —C(O)ORB, —C(O)NR 8 R 9 , —NR 8 C(O)R 9 , —S(O)R 8 , —S(O) 2 R 8 , —S(O) 2 NR 8 R 9 , —NR 8 S(O) 2 R 9 or C 1 -C 6 alkyl optionally substituted by oxo, —OH or halogen;
  • R 8 and R 9 are each independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, OH, oxo or NH 2 ;
  • R 8 and R 9 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C 1 -C 6 alkyl optionally substituted by halogen, OH, oxo or NH 2 .
  • A is 4-hydroxyphenyl optionally further substituted by R 3 , 4-hydroxy-2-pyridyl optionally further substituted by R 4 , or a 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 ;
  • B is a phenyl optionally substituted by R 3 , or a 5- to 6-membered heteroaryl optionally substituted by R 4 ;
  • R 1 is a hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 6-membered heterocyclyl, —C(O)R 1a , —C(O)OR 1a , —C(O)NR 1b R 1c , or —NR 1b R 1c , wherein the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl and 3- to 6-membered heterocyclyl of R 1 are independently optionally substituted by R 4 ;
  • each R 1a is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • each R 1b and R 1c is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • R 2 is —OR 2a , —NHR 2b , —C(O)NHR 2b , or C 1 -C 6 alkyl, wherein the C 1 -C 6 alkyl of R 2 is substituted by —OR 2c , —NHR 2c , or —C(O)NHR 2c ;
  • each R 2a and R 2b is independently cyclohexane, 6-membered heterocyclyl, —(C 1 -C 3 alkylene)N(C 2 H 5 ) 2 , —(C 1 -C 3 alkylene)(C 3 -C 6 cycloalkyl), —(C 1 -C 3 alkylene)(3- to 6-membered heterocyclyl), or —(C 1 -C 3 alkylene)(5- or 6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)OC 2 H 5 , —NH 2 or —NHCH 3 ;
  • R 2 is 5- or 6-membered heteroaryl, wherein the 5- or 6-membered heteroaryl is further substituted by C 1 -C 6 alkyl optionally substituted by halogen, —OH or oxo;
  • each R 3 is independently C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —OC(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)OR 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , —NR 5 S(O)R 6 , —C(O)NR 5 S(O)R 6 , —NR 5 S(O) 2 R 6 , —C(O)NR 5 S(O) 2 R 6 , —S(O)NR 6 R 7
  • each R 4 is independently oxo or R 3 ;
  • R 5 is independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR 8 , —NR 8 R 9 , —C(O)R 8 , —C(O)OR 8 , —C(O)NR 8 R 9 , —NR 8 C(O)R 9 , —S(O)R 8 , —S(O) 2 R 8 , —S(O) 2 NR 8 R 9 , —NR 8 S(O) 2 R 9 , or C 1 -C 6 alkyl optionally substituted by oxo, —OH or halogen;
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR 8 , —NR 8 R 9 , —C(O)R 8 , —C(O)OR 8 , —C(O)NR 8 R 9 , —NR 8 C(O)R 9 , —S(O)R 8 , —S(O) 2 R 8 , —S(O) 2 NR 8 R 9 , —NR 8 S(O) 2 R 9 , or C 1 -C 6 alkyl optionally substituted by oxo, —OH or halogen;
  • R 6 and R 7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo, —CN, —OR 8 , —NR 8 R 9 , —C(O)R 8 , —C(O)OR 8 , —C(O)NR 8 R 9 , —NR 8 C(O)R 9 , —S(O)R 8 , —S(O) 2 R 8 , —S(O) 2 NR 8 R 9 , —NR 8 S(O) 2 R 9 or C 1 -C 6 alkyl optionally substituted by oxo, —OH or halogen; R 8 and R 9 are each independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally
  • R 1 is a hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 6-membered heterocyclyl, 5- to 10-membered heteroaryl, —C(O)R 1a , —C(O)OR 1a , —C(O)NR 1b R 1c , or —S(O) 2 R 1a , wherein the C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl, 3- to 6-membered heterocyclyl, and 5- to 10-membered heteroaryl are optionally substituted with R 4 .
  • R 1 is hydrogen, C 1 -C 6 alkyl or —C(O)R 1a In certain embodiments, R 1 is hydrogen. In certain embodiments, R 1 is —C(O)R 1a where R 1a is C 1 -C 6 alkyl (e.g., methyl) or C 3 -C 6 cycloalkyl.
  • each R 1 may be combined with each R 2 , A and/or B the same as if each and every combination of R 1 with R 2 , A and/or B were specifically and individually listed.
  • R 1 is hydrogen in formula (I), wherein R 2 , A and B are as defined herein.
  • R 2 is —OR 2a . In some embodiments, R 2 is —NHR 2b . In some embodiments, R 2 is —C(O)NHR 2b . In some embodiments, R 2 is —OR 2a , —NHR 2b , or —C(O)NHR 2b , wherein each R 2a and R 2b is independently cyclohexane, 6-membered heterocyclyl, —(C 1 -C 3 alkylene)N(C 2 H 5 ) 2 , —(C 1 -C 3 alkylene)(C 3 -C 6 cycloalkyl), —(C 1 -C 3 alkylene)(3-6-membered heterocyclyl), or —(C 1 -C 3 alkylene)(5-6-membered heteroaryl), and wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF 3 , —OH
  • R 2 and R 2b is independently cyclohexane or 6-membered heterocyclyl, wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)OC 2 H 5 , —NH 2 or —NHCH 3 .
  • R 2 and R 2b is independently —(C 1 -C 3 alkylene)N(C 2 H 5 ) 2 , —(C 1 -C 3 alkylene)(C 3 -C 6 cycloalkyl), —(C 1 -C 3 alkylene)(3- to 6-membered heterocyclyl), or —(C 1 -C 3 alkylene)(5- or 6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)OC 2 H 5 , —NH 2 or —NHCH 3 .
  • R 2 is substituted C 1 -C 6 alkyl. In some embodiments, R 2 is substituted C 1 -C 3 alkyl. In some embodiments, R 2 is C 1 -C 6 alkyl substituted by —OR 2 , —NHR 2c , —SR 2c , —S(O) 2 R 2c , —S(O) 2 NHR 2c , —NHS(O) 2 R 2c —, —C(O) R 2c , —NHC(O)R 2c , —NHC(O)NR 2c , —C(O)OR 2c , —C(O)ONHR 2c —, or —C(O)NHR 2c , wherein R 2 is 5- or 6-membered heteroaryl, and wherein the 5- or 6-membered heteroaryl is further substituted by C 1 -C 6 alkyl optionally substituted by halogen, —OH or oxo.
  • R 2 is C 1 -C 6 alkyl substituted by —NHR 2c , wherein R 2c is 5- or 6-membered heteroaryl, and wherein the 5- or 6-membered heteroaryl is further substituted by C 1 -C 6 alkyl optionally substituted by halogen, —OH or oxo.
  • R 2c is pyridyl further substituted by C 1 -C 6 alkyl optionally substituted by halogen, —OH or oxo.
  • R 2 is pyridyl further substituted by —C(CH 3 ) 2 OH.
  • R 2 is selected from the group consisting of:
  • R 2 is selected from the group consisting of:
  • R 2 is selected from the group consisting of:
  • R 2 is selected from the group consisting of:
  • each R 2 may be combined with each R 1 , A and/or B the same as if each and every combination of R 2 with R 1 , A and/or B were specifically and individually listed.
  • R 1 is hydrogen
  • each R 2 may be combined with each A and/or B the same as if each and every combination of R 2 with A and/or B were specifically and individually listed.
  • A is 9- or 10-membered bicyclic heteroaryl or a 9- or 10-membered bicyclic heterocyclyl, each of A is optionally substituted by R 4 ;
  • B is a phenyl optionally substituted by R 3 , or a 5- or 6-membered heteroaryl optionally substituted by R 4 ;
  • one of Q 1 and Q 2 is —O—, —NH—, or —C(O)NH— and the other is a bond;
  • L is a bond or C 1 -C 4 alkylene
  • D is —N(C 2 H 5 ) 2 , C 3 -C 6 cycloalkyl, 3- to 6-membered heterocyclyl, or 5- or 6-membered heteroaryl, wherein each of which is optionally substituted by halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)OC 2 H 5 , —NH 2 , —NHCH 3 or C 1 -C 6 alkyl optionally substituted by halogen, —OH or oxo, when L is not a bond, or D is cyclohexane, or 6-membered heterocyclyl, wherein the cyclohexane and 6-membered heterocyclyl is optionally substituted by halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)
  • each R 3 is independently C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —OC(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)OR 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , —NR 5 S(O)R 6 , —C(O)NR 5 S(O)R 6 , —NR 5 S(O) 2 R 6 , —C(O)NR 5 S(O) 2 R 6 , —S(O)NR 6 R 7
  • each R 4 is independently oxo or R 3 ;
  • R 5 is independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR 8 , —NR 8 R 9 , —C(O)R 8 , —C(O)ORB, —C(O)NR 8 R 9 , —NR 8 C(O)R 9 , —S(O)R 8 , —S(O) 2 R 8 , —S(O) 2 NR 8 R 9 , —NR 8 S(O) 2 R 9 , or C 1 -C 6 alkyl optionally substituted by oxo, —OH or halogen;
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR 8 , —NR 8 R 9 , —C(O)R 8 , —C(O)OR 8 , —C(O)NR 8 R 9 , —NR 8 C(O)R 9 , —S(O)R 8 , —S(O) 2 R 8 , —S(O) 2 NR 8 R 9 , —NR 8 S(O) 2 R 9 , or C 1 -C 6 alkyl optionally substituted by oxo, —OH or halogen;
  • R 6 and R 7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo, —CN, —OR 8 , —NR 8 R 9 , —C(O)R 8 , —C(O)OR 8 , —C(O)NR 8 R 9 , —NR 8 C(O)R 9 , —S(O)R 8 , —S(O) 2 R 8 , —S(O) 2 NR 8 R 9 , —NR 8 S(O) 2 R 9 or C 1 -C 6 alkyl optionally substituted by oxo, —OH or halogen;
  • R 8 and R 9 are each independently hydrogen, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 3 -C 6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, OH, oxo or NH 2 ;
  • Q 1 is —O—. In some embodiments Q 1 is —NH—. In some embodiments Q 1 is —C(O)NH—. In some embodiments Q 1 is a bond.
  • Q 2 is —O—. In some embodiments Q 2 is —NH—. In some embodiments Q 2 is —C(O)NH—. In some embodiments Q 2 is a bond.
  • Q 1 is —O—, —NH—, or —C(O)NH— and Q 2 is a bond.
  • Q 2 is —O—, —NH—, or —C(O)NH— and Q 1 is a bond.
  • L is a bond. In some embodiments, L is C 1 -C 4 alkylene, for example, —CH 2 —, —CH 2 CH 2 —, and —CH 2 CH 2 CH 2 —.
  • C 1 -C 3 or C 1 -C 4 alkylene as disclosed herein is a linear alkylene.
  • C 1 -C 3 or C 1 -C 4 alkylene is a branched alkylene, such as —CH(CH 3 )— and —C(CH 3 ) 2 —.
  • —(C 1 -C 3 alkylene)(5-6-membered heteroaryl) is —CH(CH 3 )-pyridyl.
  • D is —N(C 2 H 5 ) 2 , C 3 -C 6 cycloalkyl, 3- to 6-membered heterocyclyl, or 5- or 6-membered heteroaryl, wherein each of which is optionally substituted by halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)OC 2 H 5 , —NH 2 , —NHCH 3 or C 1 -C 6 alkyl optionally substituted by halogen, —OH or oxo.
  • D when L is not a bond, D is optionally substituted —N(C 2 H 5 ) 2 . In some embodiments, when L is not a bond, D is optionally substituted C 3 -C 6 cycloalkyl. In some embodiments, when L is not a bond, D is optionally substituted 3- to 6-membered heterocyclyl. In some embodiments, when L is not a bond, D is optionally substituted 5- or 6-membered heteroaryl. In some embodiments, D is pyridyl further substituted by C 1 -C 6 alkyl optionally substituted by halogen, —OH or oxo. For example, in certain embodiments, D is pyridyl further substituted by —C(CH 3 ) 2 OH.
  • D when L is a bond, D is cyclohexane, or 6-membered heterocyclyl, wherein the cyclohexane and 6-membered heterocyclyl is optionally substituted by halogen, oxo, —CF 3 , —OH, —OCH 3 , —CN, —C(O)OCH 3 , —C(O)OC 2 H 5 , —NH 2 , —NHCH 3 or C 1 -C 6 alkyl optionally substituted by halogen, —OH or oxo.
  • D when L is a bond, D is optionally substituted cyclohexane.
  • D when L is a bond, D is optionally substituted 6-membered heterocyclyl. In some embodiments, when L is a bond, Q 1 is —C(O)NH—, Q 2 is a bond, D is optionally substituted cyclohexane, or optionally substituted 6-membered heterocyclyl.
  • Q 1 , Q 2 , L and D together are
  • Q 1 , Q 2 , L and D together are
  • Q 1 , Q 2 , L and D together are
  • Q 1 , Q 2 , L and D together are
  • Q 1 , Q 2 , L and D together are
  • Q 1 , Q 2 , L and D may be combined with each A and/or B the same as if each and every combination of Q 1 , Q 2 , L or/and D of
  • A is 4-hydroxyphenyl optionally further substituted by R 3 , 4-hydroxy-2-pyridyl optionally further substituted by R 4 , or a 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 .
  • A is 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 . In some embodiments, A is a 9- or 10-membered bicyclic heterocyclyl optionally substituted by R 4 .
  • A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 . In some embodiments, A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 , wherein one ring is saturated. In some embodiments, A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 , wherein both rings are unsaturated.
  • A is selected from the group consisting of benzimidazolyl, benzoxazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, indazolyl, quinoxalinyl, quinazolinyl, cinnolinyl, naphthyridinyl and naphthyl.
  • A is selected from the group consisting of benzimidazolyl, benzoxazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, indazolyl, quinoxalinyl, quinazolinyl, cinnolinyl, naphthyridinyl and naphthyl, each of which is optionally substituted by R 4 .
  • A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 , comprising a first and second ring, wherein the first ring has a greater number of ring atoms than the second ring.
  • the point of attachment of A to the parent molecule is on the first ring having a greater number of ring atoms.
  • the point of attachment of A to the parent molecule is on the second ring having a smaller number of ring atoms.
  • A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 , wherein the two rings are selected from the group consisting of: a 5-membered ring and a 6-membered ring or two 6-membered rings.
  • A when A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R 4 , A is an unsubstituted 9- or 10-membered bicyclic heteroaryl containing at least one annular nitrogen atom, a 9- or 10-membered bicyclic heteroaryl containing at least two annular nitrogen atoms and optionally substituted by R 4 which R 4 groups are connected to the parent structure via a carbon atom, or a 10-membered bicyclic heteroaryl optionally substituted by R 4 .
  • A is a 9- or 10-membered bicyclic heteroaryl substituted with 0 to 3 R 4 groups which may be the same or different, and which may be present on either one ring or both rings.
  • A is a 9- or 10-membered bicyclic heteroaryl substituted with 0 to 3 R 3 groups which may be the same or different, and which may be present on either one ring or both rings.
  • A is a 9- or 10-membered bicyclic heteroaryl substituted with 1 R 3 group.
  • A is a 9- or 10-membered bicyclic heteroaryl substituted with 2 R 3 groups, which may be the same or different.
  • A is a 9- or 10-membered bicyclic heteroaryl substituted with 3 R 3 groups, which may be the same or different.
  • A is selected from the group consisting of:
  • R 3 if present, is attached at any available position on the bicyclic ring system. In one aspect, at least one R 3 is present and is attached at a position on the ring bearing the wavy line (on the ring that is the attachment point of the bicyclic ring to the parent molecule). In one aspect, at least one R 3 is present and is attached at a position on the ring that does not bear the wavy line (on the ring that is fused to the ring which is the attachment point of the bicyclic ring to the parent molecule).
  • A is a 9- or 10-membered bicyclic heteroaryl substituted with 0 to 3 R 3 groups which may be the same or different, and which may be present on either one ring or both rings. In some embodiments, A is selected from the group consisting of:
  • R 3 if present, is attached at any available position on the bicyclic ring system.
  • at least one R 3 is present and is attached at a position on the ring bearing the wavy line (on the ring that is the attachment point of the bicyclic ring to the parent molecule).
  • at least one R 3 is present and is attached at a position on the ring that does not bear the wavy line (on the ring that is fused to the ring which is the attachment point of the bicyclic ring to the parent molecule).
  • A is selected from the group consisting of:
  • A is selected from the group consisting of:
  • A is selected from the group consisting of:
  • A is selected from the group consisting of:
  • A is selected from the group consisting of:
  • A is
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • B is an unsubstituted phenyl. In some embodiments, B is a phenyl optionally substituted by R 3 . In some embodiments, B is a phenyl substituted by 1 to 3 R 3 which R 3 groups may be the same or different. In other embodiments, B is a 5- to 6-membered heteroaryl optionally substituted by R 4 . In other embodiments, B is a 5- to 6-membered heteroaryl substituted by 1 to 3 R 4 which R 4 may be the same or different.
  • the 5- to 6-membered heteroaryl of B is a 5-membered heteroaryl selected from the group consisting of furanyl, oxazolyl, thiophenyl, pyrazolyl, isoxazolyl, 1,3,4-oxadiazolyl, imidazolyl, thiazolyl, isothiazolyl, triazolyl, 1,3,4-thiadiazolyl and tetrazolyl, which 5-membered heteroaryl is optionally substituted by 1 to 3 R 4 which R 4 groups may be the same or different.
  • the 5- to 6-membered heteroaryl of B is a 6-membered heteroaryl selected from the group consisting of pyridyl, pyridazinyl and pyrimidinyl which 6-membered heteroaryl is optionally substituted to 1 to 3 R 4 which R 4 groups may be the same or different
  • B is an unsubstituted phenyl. In some embodiments, B is a phenyl optionally substituted by R 3 . In some embodiments, B is a phenyl substituted by 1 to 3 R 3 which R 3 groups may be the same or different. In other embodiments, B is a 5- to 6-membered heteroaryl optionally substituted by R 4 . In other embodiments, B is a 5- to 6-membered heteroaryl substituted by 1 to 3 R 4 which R 4 may be the same or different.
  • the 5- to 6-membered heteroaryl of B is a 5-membered heteroaryl selected from the group consisting of furanyl, oxazolyl, thiophenyl, pyrazolyl, isoxazolyl, 1,3,4-oxadiazolyl, imidazolyl, thiazolyl, isothiazolyl, triazolyl, 1,3,4-thiadiazolyl and tetrazolyl, which 5-membered heteroaryl is optionally substituted by 1 to 3 R 4 which R 4 groups may be the same or different.
  • the 5- to 6-membered heteroaryl of B is a 6-membered heteroaryl selected from the group consisting of pyridyl and pyrimidinyl which 6-membered heteroaryl is optionally substituted to 1 to 3 R 4 which R 4 groups may be the same or different.
  • each R 3 of B in one aspect is independently selected from the group consisting of halogen, —CN, —OR 5 , —NR 6 R 7 , —C(O)R 5 , C 3 -C 6 cycloalkyl and C 1 -C 6 alkyl optionally substituted by halogen.
  • each R 3 of B is independently selected from the group consisting of halogen and C 1 -C 6 alkyl optionally substituted by halogen (e.g., CF 3 ).
  • B is a phenyl substituted with 1 to 3 halo groups which may be the same or different.
  • B is phenyl, fluoro-phenyl, di-fluoro-phenyl, chloro-phenyl, di-chloro-phenyl or (fluoroxchloro)-phenyl.
  • B is selected from the group consisting of:
  • B is a phenyl substituted with 1 to 3 halo groups which may be the same or different.
  • B is phenyl, fluoro-phenyl, di-fluoro-phenyl, chloro-phenyl, di-chloro-phenyl or (fluoroxchloro)-phenyl.
  • B is selected from the group consisting of:
  • B is a 5-membered heteroaryl substituted with 0 to 3 R 4 groups which may be the same or different. In some embodiments, B is a 5-membered heteroaryl substituted with 0 to 3 R 3 groups which may be the same or different. In one such aspect, B is a 5-membered heteroaryl substituted with 1 R 3 group. In another such aspect, B is a 5-membered heteroaryl substituted with 2 R 3 groups, which may be the same or different. In another such aspect, B is a 5-membered heteroaryl substituted with 3 R 3 groups, which may be the same or different. In some embodiments, B is a 5-membered heteroaryl selected from the group consisting of:
  • the B ring can be substituted with 0, 1, 2, or 3 R 3 groups, as valence permits (e.g., when the maximum number of allowed substituents is 2, the B ring can be substituted with 0, 1, or 2 R 3 groups).
  • B is a 5-membered heteroaryl substituted with 0 to 3 R 3 groups which may be the same or different. In some embodiments, B is a 5-membered heteroaryl selected from the group consisting of:
  • the B ring can be substituted with 0, 1, 2, or 3 R 3 groups, as valence permits (e.g., when the maximum number of allowed substituents is 2, the B ring can be substituted with 0, 1, or 2 R 3 groups).
  • B is a 5-membered heteroaryl selected from the group consisting of:
  • B is a 5-membered heteroaryl selected from the group consisting of:
  • B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 R 4 , which R 4 may be the same or different. In some embodiments, B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 halo groups which may be the same or different. In some embodiments, B is a 6-membered heteroaryl selected from the group consisting of:
  • B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 R 4 , which R 4 may be the same or different. In some embodiments, B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 R 3 , which R 3 may be the same or different. In some embodiments, B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 halo groups which may be the same or different. In some embodiments, B is a 6-membered heteroaryl selected from the group consisting of:
  • B is selected from the group consisting of:
  • B is selected from the group consisting of:
  • B is
  • B is
  • B is
  • B is
  • each description of B may be combined with each description of R 1 and/or R 2 the same as if each and every combination were specifically and individually listed. It is similarly understood that each description of B may be combined with each description of A (and further with each description of R 1 and R 2 ) the same as if each and every combination were specifically and individually listed.
  • B is as defined in any variation herein
  • R 1 and R 2 are as defined in any variation herein and A is 4-hydroxyphenyl optionally further substituted by R 3 or 4-hydroxy-2-pyridyl optionally further substituted by R 4 .
  • B is as defined in any variation herein
  • R 1 and R 2 are as defined in any variation herein and
  • A is 9- or 10-membered bicyclic heteroaryl (eg., quinolinyl or indazolyl) optionally substituted by R 4 .
  • the compound of formula (I) is a compound of formula (IIIa):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 1 is independently O, S, NH, NR 4a , CH 2 , CHR 4b , CR 4b R 4b , N, CH or CR 4b ;
  • each X 2 is independently NH, NR 4a , CHR 4b , CR 4b R 4b , CH, CR 4b or N;
  • each is a single or double bond, provided that when is a double bond, is a single bond and when is a double bond, is a singe bond;
  • R 4a is C 1 -C 6 alkyl
  • each R 4b is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • the compound is other than a compound selected from Table 1X or a salt thereof.
  • the compound of formula (I) is a compound of formula (IIIb):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 1 is independently O, S, NH, NR 4a , CH 2 , CHR 4b , CR 4b R 4b , N, CH or CR 4b ;
  • each X 2 is independently NH, NR 4a , CH 2 , CHR 4b , CR 4b R 4b , CH, CR 4b or N;
  • each is a single or double bond
  • R 4a is C 1 -C 6 alkyl
  • each R 4 is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • the compound is other than a compound selected from Table 1X or a salt thereof.
  • the compound of formula (I) is a compound of formula (IIIc):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 1 is independently O, S, NH, NR 4a , CH 2 , CHR 4b , CR 4b R 4b , N, CH or CR 4b ;
  • each X 2 is independently CH, CR 4b or N;
  • R 4a is C 1 -C 6 alkyl
  • each R 4b is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • the compound is other than a compound selected from Table 1X or a salt thereof.
  • the compound of formula (I) is a compound of formula (IIIc-1):
  • R 1 and R 2 are as defined for formula (I); each X 1 and X 2 are as defined for formula (IIIc);
  • X 4 is C or N
  • the compound of formula (I) is a compound of formula (IIIc-2):
  • R 1 , R 2 and R 3 are as defined for formula (I);
  • each X 1 and X 2 are as defined for formula (IIc);
  • the compound of formula (I) is a compound of formula (IIId):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 1 is independently O, S, NH, CH 2 , CHR 4b , CR 4b R 4b , N, CH or CR 4b ;
  • each X 2 is independently CH, CR 4b or N;
  • each R 4 is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • the compound of formula (I) is a compound of formula (IIIe):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 1 is independently O, S, NH, NR 4a , CH 2 , CHR 4b , CR 4b R 4b , N, CH or CR 4b ;
  • each X 2 is independently O, CH 2 , CHR 4b , CR 4b R 4b , CH, CR 4b or N;
  • each is a single or double bond
  • R 4a is C 1 -C 6 alkyl
  • each R 4 is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • the compound is other than a compound selected from Table 1X or a salt thereof.
  • the compound of formula (I) is a compound of formula (IIIf):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 1 is independently O, S, NH, NR 4a , CH 2 , CHR 4b , CR 4b R 4b , N, CH or CR 4b ;
  • each X 2 is independently C, CH, CR 4b or N;
  • each is a single or double bond, provided that when is a double bond, is a single bond and when is a double bond, is a single bond;
  • R 4a is C 1 -C 6 alkyl
  • each R 4 is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • the compound is other than a compound selected from Table 1X or a salt thereof.
  • a compound of formula (IIIf), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing is provided.
  • the compound of formula (I) is a compound of formula (IIIg):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 1 is independently O, S, NH, NR 4a , N, CH or CR 4b ;
  • each X 2 is independently C, CH, CR 4b or N;
  • R 4a is C 1 -C 6 alkyl
  • each R 4 is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • the compound is other than a compound selected from Table 1X or a salt thereof.
  • R 4b is selected from the group consisting of halogen, —OR 5 and C 1 -C 6 alkyl optionally substituted by halogen.
  • one of X 1 is N, and the other one of X 1 is NR 4a , and each X 2 is CH or CR 4b .
  • one of X 1 is N, and the other one of X 1 is O or S, and each X 2 is CH or CR 4b .
  • the compound of formula (I) is a compound of formula (IVa):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 3 is independently NH, NR 4 , CH 2 , CHR 4 , CR 4 R 4 , CR 4 , CH, C ⁇ O, O or N;
  • each is a single or double bond
  • each R 4 is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, —(C 1 -C 3 alkylene)(6-membered aryl) optionally substituted by halogen or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • the compound of formula (I) is a compound of formula (IVa):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 3 is independently NH, NR 4 , CH 2 , CHR 4 , CR 4 R 4 , CR 4 , CH or N;
  • each is a single or double bond
  • each R 4 is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • R 6 and R 7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl
  • the compound is other than a compound selected from Table 1X or a salt thereof.
  • a compound of formula (Va), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing is provided.
  • the compound of formula (I) is a compound of formula (IVb):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 3 is independently NH, NR 4 , CH 2 , CHR 4 , CR 4 R 4 , CR 4 , CH or N;
  • each is a single or double bond
  • each R 4 is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • R 6 and R 7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl, provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • the compound of formula (I) is a compound of formula (IVc):
  • R 1 , R 2 and B are as defined for formula (I);
  • each X 3 is independently CR 4 , CH or N;
  • each R 4 is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • R 6 and R 7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl
  • the compound is other than a compound selected from Table 1X or a salt thereof.
  • R 4 is selected from the group consisting of halogen, —OR 5 and C 1 -C 6 alkyl optionally substituted by halogen.
  • one X 3 is N, and the remaining X 3 are each CR 4 . In some embodiments, two of the X 3 are N, and the remaining X 3 are each CR 4 .
  • the compound of formula (I) is a compound of formula (IVc-1):
  • R 1 and R 2 are as defined for formula (I);
  • each X 1 is independently O, S, NH, NR 4 , N, CH or CR 4 ;
  • X 4 is C or N
  • each X 3 is as defined for formula (IVc)
  • R 4a is C 1 -C 6 alkyl
  • each R b is independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen;
  • each R 5 is independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl
  • R 6 and R 7 are each independently hydrogen, C 1 -C 6 alkyl, or C 3 -C 6 cycloalkyl;
  • R 6 and R 7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl, provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • the compound of formula (I) is a compound of formula (IVc-2):
  • R 1 , R 2 and R 3 are as defined for formula (I); each X 3 is as defined for formula (IVc); provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently R 4 .
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen; and B is phenyl, optionally substituted with R 3 .
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen; and B is 5- to 6-membered heteroaryl, optionally substituted with R 4 .
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen; and B is 5-membered heteroaryl such as furanyl, oxazolyl, thiophenyl, pyrazolyl, is
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen; and B is selected from the group consisting of:
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently R 4 .
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR, —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen; and B is phenyl, optionally substituted with R 3 .
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen; and B is 5- to 6-membered heteroaryl, optionally substituted with R 4 .
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen; and B is 5-membered heteroaryl such as furanyl, oxazolyl, thiophenyl, pyrazolyl, is
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • R 401 , R 402 , R 403 , R 404 , R 405 , and R 406 are each independently halogen, —CN, —OR 5 , —SR 5 , —NR 6 R 7 , —NO 2 , —C(O)R 5 , —C(O)OR 5 , —C(O)NR 6 R 7 , —C(O)NR 5 S(O) 2 R 6 , —OC(O)R 5 , —OC(O)NR 6 R 7 , —NR 5 C(O)R 6 , —NR 5 C(O)NR 6 R 7 , —S(O)R 5 , —S(O) 2 R 5 , C 3 -C 6 cycloalkyl, or C 1 -C 6 alkyl optionally substituted by halogen; and B is selected from the group consisting of:
  • salts of compounds referred to herein such as pharmaceutically acceptable salts.
  • the invention also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms, and any tautomers or other forms of the compounds described.
  • compositions comprising a compound as detailed herein are provided, such as compositions of substantially pure compounds.
  • a composition containing a compound as detailed herein or a salt thereof is in substantially pure form.
  • substantially pure intends a composition that contains no more than 35% impurity, wherein the impurity denotes a compound other than the compound comprising the majority of the composition or a salt thereof.
  • a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 25%, 20%, 15%, 10%, or 5% impurity.
  • a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 3%, 2%, 1% or 0.5% impurity.
  • the compound of formula (I) is of formula (I-A):
  • R 2 and B are as defined for formula (I) or any embodiment or aspect or other variation thereof, wherein
  • R 2 is not
  • R 2 is not
  • the compound of formula (I) is of formula (I-B):
  • R 2 and B are as defined for formula (I) or any embodiment or aspect or other variation thereof, wherein
  • R 2 is not
  • R 2 is not
  • R 2 is not
  • R 2 is not
  • R 2 is not
  • R 2 is not
  • the compound is other than the compounds in Table 1X.
  • the compound of formula (II) is of formula (II-A):
  • the compound of formula (II) is of formula (II-B):
  • the compound is other than the compounds in Table 1X.
  • the compound is other than the compounds in Table 1X, a tautomer or isomer thereof, and a salt of any of the foregoing.
  • A is 9- or 10-membered bicyclic heteroaryl or a 9- or 10-membered bicyclic heterocyclyl, each of A is optionally substituted by R a ;
  • R a group is —CN
  • A is selected from the group consisting of
  • B is selected from the group consisting of
  • each A may be combined with each B the same as if each and every combination of A and/or B were specifically and individually listed.
  • B is
  • the compounds depicted herein may be present as salts even if salts are not depicted and it is understood that the present disclosure embraces all salts and solvates of the compounds depicted here, as well as the non-salt and non-solvate form of the compound, as is well understood by the skilled artisan.
  • the salts of the compounds provided herein are pharmaceutically acceptable salts. Where one or more tertiary amine moiety is present in the compound, the N-oxides are also provided and described.
  • tautomeric forms may be present for any of the compounds described herein, each and every tautomeric form is intended even though only one or some of the tautomeric forms may be explicitly depicted.
  • the tautomeric forms specifically depicted may or may not be the predominant forms in solution or when used according to the methods described herein.
  • the present disclosure also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms of the compounds described.
  • the structure or name is intended to embrace all possible stereoisomers of a compound depicted, and each unique stereoisomer has a compound number bearing a suffix “a”, “b”, etc. All forms of the compounds are also embraced by the invention, such as crystalline or non-crystalline forms of the compounds.
  • Compositions comprising a compound of the invention are also intended, such as a composition of substantially pure compound, including a specific stereochemical form thereof, or a composition comprising mixtures of compounds of the invention in any ratio, including two or more stereochemical forms, such as in a racemic or non-racemic mixture.
  • the invention also intends isotopically-labeled and/or isotopically-enriched forms of compounds described herein.
  • the compounds herein may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
  • the compound is isotopically-labeled, such as an isotopically-labeled compound of formulae (I), (II) or (III), or variations thereof described herein, where a fraction of one or more atoms are replaced by an isotope of the same element.
  • Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C 13 N, 15 O, 17 O, 32 P, 35 S, 18 F, 36 Cl.
  • Certain isotope labeled compounds e.g. 3 H and 14 C
  • Incorporation of heavier isotopes such as deuterium ( 2 H) can afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life, or reduced dosage requirements and, hence may be preferred in some instances.
  • Isotopically-labeled compounds of the present invention can generally be prepared by standard methods and techniques known to those skilled in the art or by procedures similar to those described in the accompanying Examples substituting appropriate isotopically-labeled reagents in place of the corresponding non-labeled reagent.
  • the invention also includes any or all metabolites of any of the compounds described.
  • the metabolites may include any chemical species generated by a biotransformation of any of the compounds described, such as intermediates and products of metabolism of the compound, such as would be generated in vivo following administration to a human.
  • Articles of manufacture comprising a compound described herein, or a salt or solvate thereof, in a suitable container are provided.
  • the container may be a vial, jar, ampoule, preloaded syringe, i.v. bag, and the like.
  • the compounds detailed herein are orally bioavailable.
  • the compounds may also be formulated for parenteral (e.g., intravenous) administration.
  • One or several compounds described herein can be used in the preparation of a medicament by combining the compound or compounds as an active ingredient with a pharmacologically acceptable carrier, which are known in the art.
  • a pharmacologically acceptable carrier which are known in the art.
  • the carrier may be in various forms.
  • the manufacture of a medicament is for use in any of the methods disclosed herein, e.g., for the treatment of cancer.
  • the compounds of the invention may be prepared by a number of processes as generally described below and more specifically in the Examples hereinafter (such as the schemes provided in the Examples below).
  • the symbols when used in the formulae depicted are to be understood to represent those groups described above in relation to the formulae herein.
  • enantiomer of a compound may be accomplished from a corresponding mixture of enantiomers using any suitable conventional procedure for separating or resolving enantiomers.
  • diastereomeric derivatives may be produced by reaction of a mixture of enantiomers, e.g., a racemate, and an appropriate chiral compound. The diastereomers may then be separated by any convenient means, for example by crystallization and the desired enantiomer recovered. In another resolution process, a racemate may be separated using chiral High Performance Liquid Chromatography. Alternatively, if desired a particular enantiomer may be obtained by using an appropriate chiral intermediate in one of the processes described.
  • Chromatography, recrystallization and other conventional separation procedures may also be used with intermediates or final products where it is desired to obtain a particular isomer of a compound or to otherwise purify a product of a reaction.
  • Solvates and/or polymorphs of a compound provided herein, or a pharmaceutically acceptable salt thereof are also contemplated.
  • Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent and are often formed during the process of crystallization. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol.
  • Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and/or solubility. Various factors such as the recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate.
  • compounds of the formula (I) may be synthesized according to Scheme 1. In some embodiments, compounds of the formula (I) may be synthesized according to Scheme 1, 2 or 3.
  • A, B and R 2 are as defined for formula (I), or any variation thereof detailed herein; and X is a leaving group (e.g., alkoxy or halogen).
  • A, B and R 2b are as defined for formula (I), or any variation thereof detailed herein; and X is a leaving group (e.g., alkoxy or halogen).
  • A, B and R 2 are as defined for formula (I), or any variation thereof detailed herein; and X is a leaving group (e.g., alkoxy or halogen).
  • compositions of any of the compounds detailed herein are embraced by this disclosure.
  • the present disclosure includes pharmaceutical compositions comprising a compound as detailed herein or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier or excipient.
  • the pharmaceutically acceptable salt is an acid addition salt, such as a salt formed with an inorganic or organic acid.
  • Pharmaceutical compositions may take a form suitable for oral, buccal, parenteral, nasal, topical or rectal administration or a form suitable for administration by inhalation.
  • compositions comprising a compound in purified forms are detailed herein.
  • Compositions comprising a compound as detailed herein or a salt thereof are provided, such as compositions of substantially pure compounds.
  • a composition containing a compound as detailed herein or a salt thereof is in substantially pure form.
  • the compounds herein are synthetic compounds prepared for administration to an individual.
  • compositions are provided containing a compound in substantially pure form.
  • the present disclosure embraces pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier.
  • methods of administering a compound are provided. The purified forms, pharmaceutical compositions and methods of administering the compounds are suitable for any compound or form thereof detailed herein.
  • a compound detailed herein or salt thereof may be formulated for any available delivery route, including an oral, mucosal (e.g., nasal, sublingual, vaginal, buccal or rectal), parenteral (e.g., intramuscular, subcutaneous or intravenous), topical or transdermal delivery form.
  • oral, mucosal e.g., nasal, sublingual, vaginal, buccal or rectal
  • parenteral e.g., intramuscular, subcutaneous or intravenous
  • topical or transdermal delivery form e.g., topical or transdermal delivery form.
  • a compound or salt thereof may be formulated with suitable carriers to provide delivery forms that include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultices), pastes, powders, dressings, creams, solutions, patches, aerosols (e.g., nasal spray or inhalers), gels, suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions or water-in-oil liquid emulsions), solutions and elixirs.
  • suitable carriers include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultic
  • One or several compounds described herein or a salt thereof can be used in the preparation of a formulation, such as a pharmaceutical formulation, by combining the compound or compounds, or a salt thereof, as an active ingredient with a pharmaceutically acceptable carrier, such as those mentioned above.
  • a pharmaceutically acceptable carrier such as those mentioned above.
  • the carrier may be in various forms.
  • pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants.
  • Formulations comprising the compound may also contain other substances which have valuable therapeutic properties.
  • Pharmaceutical formulations may be prepared by known pharmaceutical methods. Suitable formulations can be found, e.g., in Remington's Pharmaceutical Sciences , Mack Publishing Company, Philadelphia, Pa., 20 th ed. (2000), which is incorporated herein by reference.
  • Compounds as described herein may be administered to individuals in a form of generally accepted oral compositions, such as tablets, coated tablets, and gel capsules in a hard or in soft shell, emulsions or suspensions.
  • carriers which may be used for the preparation of such compositions, are lactose, corn starch or its derivatives, talc, stearate or its salts, etc.
  • Acceptable carriers for gel capsules with soft shell are, for instance, plant oils, wax, fats, semisolid and liquid poly-ols, and so on.
  • pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants.
  • any of the compounds described herein can be formulated in a tablet in any dosage form described, for example, a compound as described herein or a pharmaceutically acceptable salt thereof can be formulated as a 10 mg tablet.
  • compositions comprising a compound provided herein are also described.
  • the composition comprises a compound or salt thereof and a pharmaceutically acceptable carrier or excipient.
  • a composition of substantially pure compound is provided.
  • Compounds and compositions detailed herein such as a pharmaceutical composition containing a compound of any formula provided herein or a salt thereof and a pharmaceutically acceptable carrier or excipient, may be used in methods of administration and treatment as provided herein.
  • the compounds and compositions may also be used in in vitro methods, such as in vitro methods of administering a compound or composition to cells for screening purposes and/or for conducting quality control assays.
  • a method of treating a disease in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or any embodiment, variation or aspect thereof (collectively, a compound of formulae (I), (II) or (III), or the present compounds or the compounds detailed or described herein) or a pharmaceutically acceptable salt thereof, to the individual.
  • a method of treating a disease mediated by a G protein coupled receptor signaling pathway in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual.
  • the disease is mediated by a class A G protein coupled receptor.
  • the disease is mediated by a class B G protein coupled receptor. In some embodiments, the disease is mediated by a class C G protein coupled receptor. In some embodiments, the G protein coupled receptor is a purinergic G protein receptor. In some embodiments, the G protein coupled receptor is an adenosine receptor, such as any of the A 1 , A 2A , A 2B , and A 3 receptors.
  • the present compositions may be used to treat a proliferative disease, such as cancer.
  • the cancer is a solid tumor.
  • the cancer is any of adult and pediatric oncology, myxoid and round cell carcinoma, locally advanced tumors, metastatic cancer, human soft tissue sarcomas, including Ewing's sarcoma, cancer metastases, including lymphatic metastases, squamous cell carcinoma, particularly of the head and neck, esophageal squamous cell carcinoma, oral carcinoma, blood cell malignancies, including multiple myeloma, leukemias, including acute lymphocytic leukemia, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, and hairy cell leukemia, effusion lymphomas (body cavity based lymphomas), thymic lymphoma
  • the present compounds or salts thereof are used in treatment of tumors which produce high levels of ATP and/or adenosine.
  • the extracellular concentration of adenosine is 10-20 times higher in the tumor compared to adjacent tissue.
  • the present compounds or salts thereof are used in treatment of tumors that express high levels of an ectonucleotidase.
  • the ectonucleotidase is CD39.
  • the ectonucleotidase is CD73.
  • Also provided herein is a method of enhancing an immune response in an individual in need thereof comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual.
  • Adenosine receptors are known to play an immunosuppressive role in cancer biology. High levels of adenosine present in the tumor microenvironment bind to adenosine receptors on immune cells to provide an immunosuppressive microenvironment. Specifically, binding of adenosine to the A 2A receptor provides an immunosuppressive signal that inhibits T cell proliferation, cytokine production and cytotoxicity.
  • the immune response is enhanced by a compound of formulae (I), (II) or (III) or a salt thereof enhancing activity of natural killer (NK) cells.
  • the present compounds or salts thereof increase NK cell-meditated cytotoxicity.
  • the immune response is enhanced by enhancing the activity of CD8 + T cells.
  • the present compounds or salts thereof cause an inflammatory response in the tumor microenvironment.
  • the present disclosure further provides a method of increasing the activity of a natural killer cell in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual.
  • the present compounds or salts thereof increase NK cell-meditated cytotoxicity.
  • a compound of formulae (I), (II) or (III) or a salt thereof increases the number of NK cells.
  • a compound of formulae (I), (II) or (III) or a salt thereof may be useful for modulating the activity of G protein receptor coupled signaling pathway proteins.
  • a compound of formulae (I), (II) or (III) or a salt thereof activates a G protein receptor coupled signaling pathway protein (i.e. is an agonist of a G protein receptor).
  • a compound of formulae (I), (II) or (III) or a salt thereof inhibits a G protein receptor coupled signaling pathway protein (i.e., is a G protein receptor antagonist).
  • a compound of formulae (I), (II) or (III) or a salt thereof is an adenosine receptor antagonist.
  • a compound of formulae (I), (II) or (III) or a salt thereof is an antagonist of any of the A 1 , A 2A , A 2B , and A 3 receptors.
  • a method of modulating the activity of an A 2A receptor in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof to an individual.
  • a compound of formulae (I), (II) or (III) or a salt thereof is an A 2A receptor antagonist.
  • a compound of formulae (I), (II) or (III) or a salt thereof reduces A 2A receptor signaling by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • a compound of formulae (I), (II) or (III) or a salt thereof reduces A 2A receptor signaling by 40-99%, 50-99%, 60-99%, 70-99%, 80-99%, 90-99%, or 95-99%.
  • a compound of formulae (I), (II) or (III) or a salt thereof binds to the A 2A receptor with an IC 50 of less than 1 ⁇ M, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 10 nM, less than 1 nM or less than 100 pM.
  • [compound x] binds to the A 2A receptor with an IC 50 of 500 nM to 100 pM, 400 nM to 100 pM, 300 nM to 100 pM, 200 nM to 100 pM, or 100 nM to 100 pM.
  • Also provided herein is a method of modulating the activity of an A 2B receptor in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof to an individual.
  • a compound of formulae (I), (II) or (III) or a salt thereof is an A 2B receptor antagonist.
  • a compound of formulae (I), (II) or (III) or a salt thereof reduces A 2B receptor signaling by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • a compound of formulae (I), (II) or (III) or a salt thereof reduces A 2B receptor signaling by 40-99%, 50-99%, 60-99%, 70-99%, 80-99%, 90-99%, or 95-99%.
  • a compound of formulae (I), (II) or (III) or a salt thereof binds to the A 2B receptor with an IC 50 of less than 1 ⁇ M, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 10 nM, less than 1 nM or less than 100 pM.
  • a compound of formulae (I), (II) or (III) or a salt thereof binds to the A 2B receptor with an IC 50 of 500 nM to 100 pM, 400 nM to 100 pM, 300 nM to 100 pM, 200 nM to 100 pM, or 100 nM to 100 pM.
  • Also provided herein is a method of modulating the activity of an A 3 receptor in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof to an individual.
  • a compound of formulae (I), (II) or (III) or a salt thereof is an A 3 receptor antagonist.
  • a compound of formulae (I), (II) or (III) or a salt thereof reduces A 3 receptor signaling by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • a compound of formulae (I), (II) or (III) or a salt thereof reduces A 3 receptor signaling by 40-99%, 50-99%, 60-99%, 70-99%, 80-99%, 90-99%, or 95-99%.
  • a compound of formulae (I), (II) or (III) or a salt thereof binds to the A 3 receptor with an IC 50 of less than 1 ⁇ M, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 10 nM, less than 1 nM or less than 100 pM.
  • a compound of formulae (I), (II) or (III) or a salt thereof binds to the A 3 receptor with an IC 50 of 500 nM to 100 pM, 400 nM to 100 pM, 300 nM to 100 pM, 200 nM to 100 pM, or 100 nM to 100 pM.
  • the present invention comprises a method of inhibiting tumor metastasis in an individual in need thereof comprising administering a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual.
  • the metastasis is to the lung, liver, lymph node, bone, adrenal gland, brain, peritoneum, muscle, or vagina.
  • a compound of formulae (I), (II) or (III) or a salt thereof inhibits metastasis of melanoma cells.
  • the present disclosure includes a method of delaying tumor metastasis comprising administering a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual.
  • the time to metastasis is delayed by 1 month, 2 months 3 months, 4 months, 5 months, 6 months, 12 months, or more, upon treatment with the compounds of the present invention.
  • a compound of formulae (I), (II) or (III) or a salt thereof is used to treat an individual having a proliferative disease, such as cancer as described herein.
  • the individual is at risk of developing a proliferative disease, such as cancer.
  • the individual is determined to be at risk of developing cancer based upon one or more risk factors.
  • the risk factor is a family history and/or gene associated with cancer.
  • the individual has a cancer that expresses a high level of a nucleotide metabolizing enzyme.
  • the nucleotide metabolizing enzyme is a nucleotidase, such as CD73 (ecto-5′-nucleotidase, Ecto5′NTase).
  • the individual has a cancer that expresses a high level of a nucleotidase, such as CD73.
  • the nucleotide metabolizing enzyme is an ecto-nucleotidase.
  • the ecto-nucleotidase degrades adenosine monophosphate.
  • the nucleotide metabolizing enzyme is CD39 (ecto-nucleoside triphosphate diphosphohydrolase 1, E-NTPDase1).
  • the individual has a cancer that expresses a high level of CD39.
  • the individual has a cancer that expresses a high level of an adenosine receptor, such as the A 2A receptor.
  • the presently disclosed compounds or a salt thereof may activate the immune system by modulating the activity of a G protein coupled receptor signaling pathway, for example acting as an A 2A receptor antagonist, which results in significant anti-tumor effects. Accordingly, the present compounds or a salt thereof may be used in combination with other anti-cancer agents to enhance tumor immunotherapy.
  • a method of treating a disease mediated by a G protein coupled receptor signaling pathway in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, and an additional therapeutic agent to the individual.
  • the disease mediated by a G protein coupled receptor signaling pathway is a proliferative disease such as cancer.
  • the additional therapeutic agent is a cancer immunotherapy. In some embodiments, the additional therapeutic agent is an immunostimulatory agent. In some embodiments, the additional therapeutic agent targets a checkpoint protein. In some embodiments, the additional therapeutic agent is effective to stimulate, enhance or improve an immune response against a tumor.
  • a combination therapy in which a compound of formulae (I), (II) or (III) is coadministered (which may be separately or simultaneously) with one or more additional agents that are effective in stimulating immune responses to thereby further enhance, stimulate or upregulate immune responses in a subject.
  • a method for stimulating an immune response in a subject comprising administering to the subject a compound of formulae (I), (II) or (III) or a salt thereof and one or more immunostimulatory antibodies, such as an anti-PD-1 antibody, an anti-PD-L1 antibody and/or an anti-CTLA-4 antibody, such that an immune response is stimulated in the subject, for example to inhibit tumor growth.
  • the subject is administered a compound of formulae (I), (II) or (III) or a salt thereof and an anti-PD-1 antibody.
  • a method for stimulating an immune response in a subject comprising administering to the subject a compound of formulae (I), (II) or (III) or a salt thereof and one or more immunostimulatory antibodies or immunotherapy like Chimeric antigen receptor (CAR) T-cell therapy; immunostimulatory antibodies such as an anti-PD-1 antibody, an anti-PD-L1 antibody and/or an anti-CTLA-4 antibody, such that an immune response is stimulated in the subject, for example to inhibit tumor growth.
  • CAR Chimeric antigen receptor
  • the subject is administered a compound of formulae (I), (II) or (III) or a salt thereof and an anti-PD-L1 antibody.
  • the subject is administered a compound of formulae (I), (II) or (III) or a salt thereof and an anti-CTLA-4 antibody.
  • the immunostimulatory antibody e.g., anti-PD-1, anti-PD-L1 and/or anti-CTLA-4 antibody
  • the immunostimulatory antibody is a human antibody.
  • the immunostimulatory antibody can be, for example, a chimeric or humanized antibody (e.g., prepared from a mouse anti-PD-1, anti-PD-L1 and/or anti-CTLA-4 antibody).
  • the subject is administered a compound of formulae (I), (II) or (III) or a salt thereof and CAR T-cells (genetically modified T cells).
  • the present disclosure provides a method for treating a proliferative disease (e.g., cancer), comprising administering a compound of formulae (I), (II) or (III) or a salt thereof and an anti-PD-1 antibody to a subject.
  • a compound of formulae (I), (II) or (III) or a salt thereof is administered at a subtherapeutic dose
  • the anti-PD-1 antibody is administered at a subtherapeutic dose
  • both are administered at a subtherapeutic dose.
  • the present disclosure provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound of formulae (I), (II) or (III) or a salt thereof and a subtherapeutic dose of anti-PD-1 antibody to a subject.
  • the subject is human.
  • the anti-PD-1 antibody is a human sequence monoclonal antibody
  • the present invention provides a method for treating a hyperproliferative disease (e.g., cancer), comprising administering a compound of formulae (I), (II) or (III) or a salt thereof and an anti-PD-L1 antibody to a subject.
  • a compound of formulae (I), (II) or (III) or a salt thereof is administered at a subtherapeutic dose
  • the anti-PD-L1 antibody is administered at a subtherapeutic dose
  • both are administered at a subtherapeutic dose.
  • the present invention provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound of formulae (I), (II) or (III) or a salt thereof and a subtherapeutic dose of anti-PD-L1 antibody to a subject.
  • the subject is human.
  • the anti-PD-L1 antibody is a human sequence monoclonal antibody.
  • the combination of therapeutic agents discussed herein can be administered concurrently as a single composition in a pharmaceutically acceptable carrier, or concurrently as separate compositions each in a pharmaceutically acceptable carrier.
  • the combination of therapeutic agents can be administered sequentially.
  • an anti-CTLA-4 antibody and a compound of formulae (I), (II) or (III) or a salt thereof can be administered sequentially, such as anti-CTLA-4 antibody being administered first and a compound of formulae (I), (II) or (III) or a salt thereof second, or a compound of formulae (I), (II) or (III) or a salt thereof being administered first and anti-CTLA-4 antibody second.
  • an anti-PD-1 antibody and a compound of formulae (I), (II) or (III) or a salt thereof can be administered sequentially, such as anti-PD-1 antibody being administered first and a compound of formulae (I), (II) or (III) or a salt thereof second, or a compound of formulae (I), (II) or (III) or a salt thereof being administered first and anti-PD-1 antibody second.
  • an anti-PD-L1 antibody and a compound of formulae (I), (II) or (III) or a salt thereof can be administered sequentially, such as anti-PD-L1 antibody being administered first and a compound of formulae (I), (II) or (III) or a salt thereof second, or a compound of formulae (I), (II) or (III) or a salt thereof being administered first and anti-PD-L1 antibody second.
  • sequential administration can be reversed or kept in the same order at each time point of administration, sequential administrations can be combined with concurrent administrations, or any combination thereof.
  • a compound of formulae (I), (II) or (III) or a salt thereof can be further combined with an immunogenic agent, such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines.
  • an immunogenic agent such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines.
  • a compound of formulae (I), (II) or (III) or a salt thereof can also be further combined with standard cancer treatments.
  • a compound of formulae (I), (II) or (III) or a salt thereof can be effectively combined with chemotherapeutic regimes. In these instances, it is possible to reduce the dose of other chemotherapeutic reagent administered with the combination of the instant disclosure (Mokyr et al. (1998) Cancer Research 58: 5301-5304).
  • Other combination therapies with a compound of formulae (I), (II) or (III) or a salt thereof include radiation, surgery, or hormone deprivation.
  • Angiogenesis inhibitors can also be combined with a compound of formulae (I), (II) or (III) or a salt thereof. Inhibition of angiogenesis leads to tumor cell death, which can be a source of tumor antigen fed into host antigen presentation pathways.
  • a compound of formulae (I), (II) or (III) or a salt thereof can be used in conjunction with anti-neoplastic antibodies.
  • treatment with an anti-cancer antibody or an anti-cancer antibody conjugated to a toxin can lead to cancer cell death (e.g., tumor cells) which would potentiate an immune response mediated by CTLA-4, PD-1, PD-L1 or a compound of formulae (I), (II) or (III) or a salt thereof.
  • a treatment of a hyperproliferative disease can include an anti-cancer antibody in combination with a compound of formulae (I), (II) or (III) or a salt thereof and anti-CTLA-4 and/or anti-PD-1 and/or anti-PD-L1 antibodies, concurrently or sequentially or any combination thereof, which can potentiate anti-tumor immune responses by the host.
  • Other antibodies that can be used to activate host immune responsiveness can be further used in combination with a compound of formulae (I), (II) or (III) or a salt thereof.
  • a compound of formulae (I), (II) or (III) or a salt thereof can be combined with an anti-CD73 therapy, such as an anti-CD73 antibody.
  • a compound of formulae (I), (II) or (III) or a salt thereof can be combined with an anti-CD39 therapy, such as an anti-CD39 antibody.
  • a compound of formulae (I), (II) or (III) or a salt thereof is administered in combination another G protein receptor antagonist, such as an adenosine A 1 and/or A 3 antagonist.
  • the dose of a compound administered to an individual may vary with the particular compound or salt thereof, the method of administration, and the particular disease, such as type and stage of cancer, being treated.
  • the amount of the compound or salt thereof is a therapeutically effective amount.
  • the effective amount of the compound may in one aspect be a dose of between about 0.01 and about 100 mg/kg.
  • Effective amounts or doses of the compounds of the invention may be ascertained by routine methods, such as modeling, dose escalation, or clinical trials, taking into account routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the disease to be treated, the subject's health status, condition, and weight.
  • An exemplary dose is in the range of about from about 0.7 mg to 7 g daily, or about 7 mg to 350 mg daily, or about 350 mg to 1.75 g daily, or about 1.75 to 7 g daily.
  • Any of the methods provided herein may in one aspect comprise administering to an individual a pharmaceutical composition that contains an effective amount of a compound provided herein or a salt thereof and a pharmaceutically acceptable excipient.
  • a compound or composition of the invention may be administered to an individual in accordance with an effective dosing regimen for a desired period of time or duration, such as at least about one month, at least about 2 months, at least about 3 months, at least about 6 months, or at least about 12 months or longer, which in some variations may be for the duration of the individual's life.
  • the compound is administered on a daily or intermittent schedule.
  • the compound can be administered to an individual continuously (for example, at least once daily) over a period of time.
  • the dosing frequency can also be less than once daily, e.g., about a once weekly dosing.
  • the dosing frequency can be more than once daily, e.g., twice or three times daily.
  • the dosing frequency can also be intermittent, including a ‘drug holiday’ (e.g., once daily dosing for 7 days followed by no doses for 7 days, repeated for any 14 day time period, such as about 2 months, about 4 months, about 6 months or more). Any of the dosing frequencies can employ any of the compounds described herein together with any of the dosages described herein.
  • a drug holiday e.g., once daily dosing for 7 days followed by no doses for 7 days, repeated for any 14 day time period, such as about 2 months, about 4 months, about 6 months or more.
  • the compounds provided herein or a salt thereof may be administered to an individual via various routes, including, e.g., intravenous, intramuscular, subcutaneous, oral and transdermal.
  • a compound provided herein can be administered frequently at low doses, known as ‘metronomic therapy,’ or as part of a maintenance therapy using compound alone or in combination with one or more additional drugs.
  • Metronomic therapy or maintenance therapy can comprise administration of a compound provided herein in cycles.
  • Metronomic therapy or maintenance therapy can comprise intra-tumoral administration of a compound provided herein.
  • the invention provides a method of treating cancer in an individual by parenterally administering to the individual (e.g., a human) an effective amount of a compound or salt thereof.
  • the route of administration is intravenous, intra-arterial, intramuscular, or subcutaneous.
  • the route of administration is oral.
  • the route of administration is transdermal.
  • compositions including pharmaceutical compositions as described herein for the use in treating, preventing, and/or delaying the onset and/or development of cancer and other methods described herein.
  • the composition comprises a pharmaceutical formulation which is present in a unit dosage form.
  • articles of manufacture comprising a compound of the disclosure or a salt thereof, composition, and unit dosages described herein in suitable packaging for use in the methods described herein.
  • suitable packaging is known in the art and includes, for example, vials, vessels, ampules, bottles, jars, flexible packaging and the like.
  • An article of manufacture may further be sterilized and/or sealed.
  • kits for carrying out the methods of the invention which comprises one or more compounds described herein or a composition comprising a compound described herein.
  • the kits may employ any of the compounds disclosed herein.
  • the kit employs a compound described herein or a pharmaceutically acceptable salt thereof.
  • the kits may be used for any one or more of the uses described herein, and, accordingly, may contain instructions for the treatment of cancer.
  • Kits generally comprise suitable packaging.
  • the kits may comprise one or more containers comprising any compound described herein.
  • Each component if there is more than one component
  • kits may be in unit dosage forms, bulk packages (e.g., multi-dose packages) or sub-unit doses.
  • kits may be provided that contain sufficient dosages of a compound as disclosed herein and/or a second pharmaceutically active compound useful for a disease detailed herein (e.g., hypertension) to provide effective treatment of an individual for an extended period, such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 7 months, 8 months, 9 months, or more.
  • Kits may also include multiple unit doses of the compounds and instructions for use and be packaged in quantities sufficient for storage and use in pharmacies (e.g., hospital pharmacies and compounding pharmacies).
  • kits may optionally include a set of instructions, generally written instructions, although electronic storage media (e.g., magnetic diskette or optical disk) containing instructions are also acceptable, relating to the use of component(s) of the methods of the present invention.
  • electronic storage media e.g., magnetic diskette or optical disk
  • the instructions included with the kit generally include information as to the components and their administration to an individual.
  • Step-1 Synthesis of 6-phenylpyrazin-2-amine: To a stirred solution of 6-chloropyrazin-2-amine (50 g, 0.3861 mol) in dioxane:water (400 mL:100 mL) was added benzeneboronic acid (56.4 g, 0.46 mol). The reaction mixture was purged with nitrogen for 20 min then charged Na 2 CO 3 (70.6 g, 0.57 mol) and Pd(PPh 3 )Cl 2 (13.5 g, 0.01930 mol). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS.
  • Step-2 Synthesis of 5-bromo-6-phenylpyrazin-2-amine: To a stirred solution of 6-phenylpyrazin-2-amine (48 g, 0.2803 mol) in DMF was added NBS (49.9 g, 0.28 mol) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at RT for 16 h. The reaction was monitored by TLC and LCMS. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 100 mL).
  • Step-3 synthesis of 6-phenyl-5-(quinolin-6-yl)pyrazin-2-amine: To a stirred solution of 5-bromo-6-phenylpyrazin-2-amine (38 g, 0.1519 mol) in dioxane:water (320 mL:80 mL) was added quinolin-6-ylboronic acid (46.4 g, 0.18 mol). The reaction mixture was purged with nitrogen for 20 min then charged with Na 2 CO 3 (32.2 g, 0.3038 mol) and Pd(dppf)Cl 2 (6.19 g, 0.007 mol). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h.
  • Step-4 synthesis of 3-bromo-6-phenyl-5-(quinolin-6-yl)pyrazin-2-amine: To a stirred solution of 6-phenyl-5-(quinolin-6-yl) pyrazin-2-amine (21 g, 0.07 mol) in DMF was added NBS (12.5 g, 0.07 mol) at 0° C. under nitrogen atmosphere. The reaction mixture was stir at RT for 16 h. The reaction was monitored by TLC and LCMS. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 30 mL).
  • Step-5 Synthesis of N2-(2-(diethylamino)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2,3-diamine: To a stirred solution of N1,N1-diethylethane-1,2-diamine (0.155 g, 1.32 mmol, 5.0 eq) in DMF (5 mL) was added Cs 2 CO 3 (0.104 g, 0.31 mmol, 1.2 eq) and the mixture was stirred at RT for 15 min.
  • Example S-2 Synthesis of 3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 73)
  • Step-1 Synthesis of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carbonitrile: To a stirred solution of NaCN (1.56 g, 0.03 mol) and CuCN (5.7 g, 0.06 mol) in dry DMF (150 mL) was added 3-bromo-6-phenyl-5-(quinolin-6-yl)pyrazin-2-amine (12.0 g, 0.03 mol) at 120° C. The reaction mixture was stirred at 145° C. for 12 h. The reaction was monitored by TLC and LCMS. The reaction was distilled. The crude product was poured in ice-water the solid precipitate out.
  • Step-2 Synthesis of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid: To a stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carbonitrile (1 g, 3.08 mmol, 1 eq) in 1,4-dioxane (50 mL) and aqueous NaOH (10%, 50 mL) was heated at 100° C. for 48 h. Progress of reaction was monitored by LCMS.
  • Step-3 Synthesis of 3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To a stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.100 g, 0.30 mmol, 1 eq) and 1-(pyridin-2-yl)ethan-1-amine (0.07 g, 0.58 mmol, 2.0 eq) in DMF (2 mL), was added HOBT (0.06 g, 0.43 mmol, 1.5 eq), EDC.HCl (0.08 g, 0.43 mmol, 1.5 eq) and DIPEA (0.113 g, 0.87 mmol, 3.0 eq).
  • reaction mixture was allowed to stir at RT for 24 h. Progress of reaction was monitored by TLC and LCMS. On completion of the reaction, the reaction mixture was diluted with water (15 mL) and extracted with ethyl acetate (50 mL ⁇ 2). Combined organic layer was washed with water (50 mL ⁇ 2), dried over anhydrous Na 2 SO 4 and concentrated under vacuum to get the solid residue which was purified by normal phase column chromatography to get the desired product (5 mg, 2%).
  • Example-S-3 Synthesis of (S)-3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide. (Compound No. 75)
  • Step-1 Synthesis of (S, E)-2-methyl-N-(pyridin-2-ylmethylene)propane-2-sulfinamide: To a stirred solution of pyridine-2-carboxaldehyde (1 g, 9.34 mmol, 1.0 eq) and copper(II) sulfate (2.98 g, 18.69 mmol, 2.0 eq) in dichloromethane (15 mL) was added (S)-2-methylpropane-2-sulfinamide (1.13 g, 9.34 mmol, 1.0 eq) at RT. The resulting mixture was heated at 50° C. for 16 h.
  • reaction mixture was allowed to cool to room temperature, filtered through celite pad, the celite pad washed with dichloromethane (30 mL). The combined filtrate dried over anhydrous Na 2 SO 4 and concentrated under vacuum to get the solid residue which was purified by flash column chromatography to get the desired product as white solid (1.2 g, 61%).
  • Step-2 Synthesis of (S)-2-methyl-N—((S)-1-(pyridin-2-yl)ethyl)propane-2-sulfinamide: To a stirred solution of (S,E)-2-methyl-N-(pyridin-2-ylmethylene)propane-2-sulfinamide (1.0 g, 4.76 mmol, 1.0 eq) in tetrahydrofuran (15 mL) was added drop wise 3 M methylmagnesium bromide (2.38 mL, 7.14 mmol, 1.5 eq) at ⁇ 78° C. The resulting mixture was stirred for 4 h at same temperature. The reaction was then quenched by careful addition of saturated NH 4 Cl (10 mL).
  • Step-3 Synthesis of (S)-1-(pyridin-2-yl)ethanamine: To a stirred solution of (S)-2-methyl-N—((S)-1-(pyridin-2-yl)ethyl)propane-2-sulfinamide (0.7 g, 3.09 mmol, 1.0 eq) in methanol (5 mL) was added 4N HCl in dioxane (1.6 ml) at RT. The resulting mixture was stirred for 30 min. Following this the reaction mixture was evaporated under reduced pressure to get solid residue. The obtained solid was washed with diethyl ether, died under vacuum to get desired product as off white solid (0.35 g, 94%).
  • Step-4 Synthesis of (S)-3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.1 g, 0.29 mmol, 1.0 eq) in DMF (5.0 ml) was added (S)-1-(pyridin-2-yl)ethanamine (0.042 g, 0.35 mmol, 1.2 eq), DIPEA (0.15 mL, 0.87 mmol, 3 eq) and HATU (0.22 g, 0.58 mmol, 2 eq) at RT under inert condition.
  • Example-S-4 Synthesis of (R)-3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide. (Compound No. 76)
  • Step-1 Synthesis of 2-methyl-N-[(E)-pyridin-2-ylmethylidene]propane-2-sulfinamide: To a stirred solution of pyridine-2-carboxaldehyde (1 g, 9.34 mmol, 1.0 eq) and copper(II) sulfate (2.98 g, 18.69 mmol, 2.0 eq) in dichloromethane (15 mL) was added (R)-2-methylpropane-2-sulfinamide (1.13 g, 9.34 mmol, 1.0 eq) at RT. The resulting mixture was heated at 50° C. for 16 h.
  • Step-2 Synthesis of 2-methyl-N-[(1R)-1-(pyridin-2-yl)ethyl]propane-2-sulfinamide: To a stirred solution of 2-methyl-N-[(E)-pyridin-2-ylmethylidene]propane-2-sulfinamide (800 mg, 3.80 mmol, 1.0 eq) in tetrahydrofuran (10 mL) was added drop wise 3 M methylmagnesium bromide (2.5 mL, 7.61 mmol, 2.0 eq) at ⁇ 78° C. The resulting mixture was stirred for 4 h at same temperature. The reaction was then quenched by addition of saturated NH 4 Cl (10 mL).
  • Step-3 Synthesis of (1R)-1-(pyridin-2-yl)ethanamine: To a stirred solution of 2-methyl-N-[(1R)-1-(pyridin-2-yl)ethyl]propane-2-sulfinamide (600 mg, 2.65 mmol, 1.0 eq) in methanol (10 mL) was added 4N HCl in dioxane (2.5 ml) at RT. The resulting mixture was stirred for 30 min. Following this the reaction mixture was evaporated under reduced pressure to get solid residue. The obtained solid was washed with diethyl ether, died under vacuum to get desired product as solid (0.35 g, 94%).
  • Step-4 Synthesis of (R)-3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.1 g, 0.29 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine e (68 mg, 0.43 mmol, 1.5 eq), DIPEA (0.2 mL, 0.87 mmol, 3 eq) and HATU (220 mg, 0.58 mmol, 2 eq) at RT under inert condition.
  • Example S-5 Synthesis of 3-amino-S-phenyl-6-(quinolin-6-yl)-N-(2-(tetrahydro-2H-pyran-4-yl)ethyl)pyrazine-2-carboxamide. (Compound No. 89)
  • reaction was monitored by TLC and LCMS. On completion of the reaction, the reaction mixture was diluted with water (15 mL) and extracted with ethyl acetate (50 mL ⁇ 2). Combined organic layer were washed (brine), dried (anhydrous Na 2 SO 4 ) and concentrated under vacuum to get the solid residue which was purified by reverse phase column chromatography to get the desired product (3 mg, 2%).
  • Example-S-6 Synthesis of 3-(6-amino-3-(8-chloroquinolin-6-yl)pyrazin-2-yl)-2-methylbenzonitrile. (Compound No. 2-2)
  • Step-1 Synthesis of 6-chloro-5-(8-chloroquinolin-6-yl)pyrazin-2-amine: To a stirred solution of 5-bromo-6-chloropyrazin-2-amine (4.0 g, 19.23 mmol, 1 eq) in dioxane:water (180 mL:20 mL) was added 8-chloroquinolin-6-ylboronic acid (5.0 g, 17.30 mmol, 0.9 eq). The reaction mixture was purged with nitrogen for 20 min then charged with Na 2 CO 3 (4.1 g, 39.0 mmol, 2.0 eq) and Pd(dppf)Cl 2 .DCM (787 mg, 5 mol %).
  • reaction mixture was again purged with nitrogen.
  • the reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h.
  • the reaction was monitored by TLC and LCMS.
  • the reaction mixture was filtered through celite and distilled.
  • the reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL). The combined organic layers were washed (brine), dried (anhydrous Na 2 SO 4 ) and concentrated under vacuum to get the solid which was purified by reverse phase column chromatography to get the desired product (2.2 g, 78%).
  • Step-2 Synthesis of 3-(6-amino-3-(8-chloroquinolin-6-yl)pyrazin-2-yl)-2-methylbenzonitrile: To a stirred solution of 6-chloro-5-(8-chloroquinolin-6-yl)pyrazin-2-amine (0.100 g, 0.35 mmol, 1 eq) in dioxane:water (4 mL: 10 mL) was added 2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzonitrile (0.9 g, 0.31 mmol, 1.2 eq).
  • reaction mixture was purged with nitrogen for 20 min then charged with Na 2 CO 3 (0.073 g, 0.69 mmol, 2.0 eq) and Pd(dppf)Cl 2 .DCM (14 mg, 5 mol %). The reaction mixture was again purged with nitrogen. The reaction mixture was stiffed at RT for 10 min followed by heating at 100° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • Example-S-7 Synthesis of 3-(6-amino-3-(8-chloroquinolin-6-yl)pyrazin-2-yl)-2-fluorobenzonitrile (Compound No. 2-10)
  • the reaction mixture was stirred at RT for 10 min followed by heating at 100° C. for 16 h.
  • the reaction was monitored by TLC and LCMS.
  • the reaction mixture was filtered through celite and distilled.
  • the reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • the combined organic layers were washed (brine), dried (anhydrous Na 2 SO 4 ) and concentrated under vacuum to get the solid which was purified by reverse phase column chromatography to get the desired product (0.008 g, 12%).
  • LCMS 376 [M+1] + .
  • Example-S-8 Synthesis of 3-amino-N-((6-cyanopyridin-2-yl)methyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 98)
  • Step 1 Synthesis of 2-(pyridin-2-yl)propan-2-amine: To a stirred solution of anhydrous cerium (III) chloride (7.1 g, 0.0288 mmol, 3 eq) in THF (15 mL) was added methyl lithium (1.6 M, 18 mL, 0.0288 mmol, 3 eq) at ⁇ 78° C. The reaction was allowed to stir at same temperature for 30 min. Picolinonitrile (1 g, 0.0096 mmol, 3 eq) in THF was added at ⁇ 78° C. and reaction was allowed to stir at RT for 1 h. The reaction was cooled to ⁇ 40° C. then charged with 10 mL ammonium hydroxide.
  • Step 2 Synthesis of 3-amino-5-phenyl-N-(2-(pyridin-2-yl)propan-2-yl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To a stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (100 mg, 0.2924 mmol, 1.0 eq) and 2-(pyridin-2-yl)propan-2-amine (222 mg, 0.3508 mmol, 2 eq) in DMF (5 mL) was added DIPEA (0.150 mL, 0.8772 mmol, 3.0 eq) at 0° C.
  • Example S-10 Synthesis of 3-amino-N-(2-(diethylamino)ethyl)-S-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 107)
  • Step 1 Synthesis of 6-(4-fluorophenyl)pyrazin-2-amine: To a stirred solution of 6-chloropyrazin-2-amine (2 g, 15.50 mmol, 1 eq) in dioxane:water (50 mL:10 mL) was added 4-fluorobenzeneboronic acid (2.8 g, 20.15 mmol, 1.3 eq). The reaction mixture was purged with nitrogen for 20 min then charged K 2 CO 3 (4.2 g, 31.0 mmol, 2.0 eq) and Pd(dppf)Cl 2 .DCM complex (632 mg, 0.77 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen.
  • the reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h.
  • the reaction was monitored by TLC and LCMS.
  • the reaction mixture was filtered through celite and distilled.
  • the reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • the combined organic layers were washed (brine), dried (anhydrous Na 2 SO 4 ) and concentrated under vacuum to get the solid which was purified by column chromatography over silica gel (100-200 mesh) [Ethyl acetate:Hexane (5:5) as eluent] to get the title compound (2 g, 68%).
  • LCMS 190 [M+1] + .
  • Step 2 Synthesis of 5-bromo-6-(4-fluorophenyl)pyrazin-2-amine: To a stirred solution of 6-(4-fluorophenyl)pyrazin-2-amine (2 g, 10.50 mmol, 1.0 eq) in DMF (20 mL) was added NBS (1.9 g, 10.50 mmol, 1.0 eq) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at RT for 30 min. The reaction was monitored by TLC and LCMS. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 100 mL).
  • Step 3 5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)pyrazin-2-amine: To a stirred solution of 5-bromo-6-(4-fluorophenyl)pyrazin-2-amine (4.5 g, 16.79 mmol, 1 eq) in dioxane:water (50 mL:10 mL) was added 8-chloroquinolin-6-ylboronic acid (5.8 g, 20.14 mmol 1.2 eq).
  • reaction mixture was purged with nitrogen for 20 min then charged with K 2 CO 3 (4.6 g, 33.58 mmol, 2.0 eq) and Pd(dppf)Cl 2 .DCM complex (685 mg, 0.83 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • Step 4 Synthesis of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)pyrazin-2-amine.
  • NBS 498 mg, 2.85 mmol, 1 e.q
  • Step-5 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)pyrazine-2-carbonitrile: To a stirred solution of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)pyrazin-2-amine (500 mg, 1.16 mmol, 1.0 eq) in DMF (5 mL) was added cuprous cyanide (0.104 g, 3.50 mmol, 3.0 eq). The reaction mixture was allowed to heat at 150° C. for 1 h using microwave irradiation.
  • Step-6 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid: To a stirred solution of 3-bromo-6-phenyl-5-(quinolin-6-yl)pyrazin-2-amine (0.2 g, 0.53 mmol, 1.0 eq) in ethanol (5 mL) was added 6M NaOH solution (5 mL). The resulting reaction mixture was heated at 100° C. for 16 h. The reaction mixture was allowed to cool to RT. The solvent was evaporated under vacuum and acidified using 1N HCl to get the solid which was filtered and dried to get the title compound (0.20 g, 95%). LCMS: 394 [M+1] + .
  • Step-7 (R)-3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid (0.1 g, 0.25 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine (47 mg, 0.43 mmol, 1.5 eq), DIPEA (0.2 mL, 0.75 mmol, 3 eq) and HATU (190 mg, 0.50 mmol, 2 eq) at RT under inert condition.
  • the reaction mixture was diluted with water (50 mL) and extracted using ethyl acetate (3 ⁇ 50 mL). The separated organic layer was dried over sodium sulfate and concentrated under reduced pressure. The crude product was purified by normal phase silica gel column chromatography to afford (0.015 g, 11%) the title compound.
  • Example S-13 Synthesis of Synthesis of S-(6-amino-3-(8-chloroquinolin-6-yl)pyrazin-2-yl)-2-fluorobenzonitrile (Compound No. 2-13)
  • reaction mixture was stiffed at RT for 10 min followed by heating at 100° C. for 16 h.
  • the reaction was monitored by TLC and LCMS.
  • the reaction mixture was filtered through celite and distilled.
  • the reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL). The combined organic layers were washed (brine), dried (anhydrous Na 2 SO 4 ) and concentrated under vacuum to get the solid which was purified by reverse phase column chromatography to get the title compound (0.04 g, 15%).
  • Example S-15 Synthesis of (R)-3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 104)
  • Step-1 Synthesis of 5-(7-chloro-1H-indazol-5-yl)-6-(4-fluorophenyl)pyrazin-2-amine.
  • 5-bromo-6-(4-fluorophenyl)pyrazin-2-amine 1.3 g, 4.84 mmol, 1.0 eq
  • dioxane:water 50 mL:10 mL
  • 7-chloro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole 1.6 g, 5.80 mmol, 1.2 eq.
  • reaction mixture was purged with nitrogen for 20 min then charged with K 2 CO 3 (1.3 g 9.63 mmol, 2.0 eq) and Pd(dppf)Cl 2 .DCM complex (197 mg, 0.02 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • Step-2 Synthesis of 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(4-fluorophenyl)pyrazin-2-amine: To a stirred solution of 5-(7-chloro-1H-indazol-5-yl)-6-(4-fluorophenyl)pyrazin-2-amine (533 mg, 1.45 mmol, 1 eq) in DMF (10 ml) was added NBS (259 mg, 1.45 mmol, 1.0 eq) at 0° C. Reaction mixture was stirred at 0° C. for 30 min. The reaction was monitored by TLC and LCMS and found to be complete after 30 min.
  • Step-3 Synthesis 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)pyrazine-2-carbonitrile: To a stirred solution of 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(4-fluorophenyl)pyrazin-2-amine (483 mg, 1.15 mmol, 1.0 eq) in DMF (10 mL) was added cuprous cyanide (206 mg, 2.30 mmol, 2.0 eq). The reaction mixture was allowed to stir at 150° C. for 1 h under microwave irradiation.
  • Step-4 Synthesis of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid: To a stirred solution of -amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)pyrazine-2-carbonitrile (250 mg, 0.68 mmol, 1.0 eq) in ethanol (5 mL) was added 6M NaOH solution (5 mL). The resulting reaction mixture was heated at 100° C. for 16 h. The reaction mixture was allowed to cool to RT. The solvent was evaporated under vacuum and acidified using 1N HCl to get the solid which was filtered and dried to get the title compound (100 mg, 38%) LCMS: 384 (M+1) + .
  • Step-5 (R)-3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To stirred solution of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid (100 mg, 0.26 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine (63 mg, 0.39 mmol, 1.5 eq), DIPEA (0.2 mL, 0.75 mmol, 3 eq) and HATU (190 mg, 0.50 mmol, 2 eq) at RT under inert condition.
  • Example S-16 Synthesis of 5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)-3-(1-(pyridin-2-yl)ethoxy)pyrazin-2-amine (Compound No. 103)
  • Step-1 Synthesis of 5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine: To a stirred solution of 6-chloro-5-(quinolin-6-yl)pyrazin-2-amine (1.0 g, 3.44 mmol, 1.0 eq) in dioxane:water (16 mL: 4 mL) was added 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (0.860 g, 4.12 mmol, 1.2 eq).
  • reaction mixture was purged with nitrogen for 5 min then charged with Na 2 CO 3 (0.73 g, 6.88 mmol, 2.0 eq) and Pd(dppf)Cl 2 .DCM complex (0.080 g, 10 mol %). The reaction mixture was again purged with nitrogen. The reaction mixture was allowed to heat at 100° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • Step-2 Synthesis of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine.
  • 5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine (0.400 g, 1.18 mmol, 1 eq) in DMF (5 ml) was added NBS (210 mg, 1.18 mmol, 1.0 eq) at 0° C. Reaction mixture was stirred at 0° C. for 10 min. The reaction was monitored by TLC and LCMS and found to be complete after 10 min.
  • Step-3 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carbonitrile: To a stirred solution of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine (500 mg, 1.20 mmol, 1.0 eq) in DMF (10 mL) was added cuprous cyanide (120 mg, 1.35 mmol, 1.1 eq). The reaction mixture was allowed to stir at 120° C. for 45 min under microwave irradiation.
  • Step-4 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxylic acid: To a stirred solution of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine (170 mg, 0.48 mmol, 1.0 eq) in ethanol (5 mL) was added 6M NaOH solution (5 mL). The resulting reaction mixture was heated at 100° C. for 16 h. The reaction mixture was allowed to cool to RT. The solvent was evaporated under vacuum and acidified using 1N HCl to get the solid which was filtered and dried to get the product as yellow solid (110 mg, 60%) LCMS: 381 [M+1] + .
  • Step-3 Synthesis of (R)-3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To a stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxylic acid (50 mg, 0.13 mmol, 1 eq) in DMF (4 mL) was added (1R)-1-(pyridin-2-yl)ethanamine (23 mg, 0.19 mmol, 1.2 eq), DIPEA (0.1 mL, 0.39 mmol, 3 eq) and HATU (95 mg, 0.26 mmol, 2 eq) at RT under inert condition.
  • the reaction mixture was heated at 90° C. for 16 h under carbon monoxide. The reaction was monitored by TLC and LCMS. The reaction mixture was filter through celite. The reaction mixture was diluted with water (50 mL) and extracted using ethyl acetate (3 ⁇ 50 mL). The separated organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by normal phase silica-gel column chromatography followed by HPLC purification to get the title compound (4 mg, 2%).
  • Step-1 Synthesis of 6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine.
  • 6-chloro-5-(quinolin-6-yl)pyrazin-2-amine 500 mg, 1.95 mmol, 1 eq
  • DMF:water 10 mL:5 mL
  • 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (487 mg, 2.34 mmol, 1.2 eq).
  • reaction mixture was purged with nitrogen for 5 min then charged with K 3 PO 4 (1.2 g, 5.85 mmol, 3.0 eq) and Pd(PPh 3 ) 4 (90 mg, 0.07 mmol, 0.04 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 120° C. for 1 h using microwave irradiation. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • Step-2 Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine: To a stirred solution 6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine (226 mg, 0.74 mmol, 1 eq) in DMF (10 ml) was added NBS (133 mg, 0.61 mmol, 1.0 eq) at 0° C. Reaction mixture was stiffed at 0° C. for 10 min. The reaction was monitored by TLC and LCMS and found to be complete after 10 min.
  • Step-3 Synthesis of (R)-3-amino-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To a stirred solution of 3-bromo-6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine (189 mg, 0.49 mmol, 1 eq) in DMF (10 mL) was added (R)-1-(pyridin-2-yl)ethanamine (121 mg, 0.99 mmol, 1.2 eq).
  • reaction mixture was purged with nitrogen for 5 min then charged with Mo(CO) 6 (48 mg, 0.18 mmol, 0.37 eq) and PdCl 2 dppf (18 mg, 0.02 mmol, 0.05 eq).
  • the reaction mixture was again purged with nitrogen for 5 min and then stirred at RT for 1 h followed by the addition of DBU (0.2 mL, 1.07 mmol, 2.2 eq).
  • the reaction mixture was stirred at RT for 5 min and then heated at 120° C. for 3 h.
  • the reaction was monitored by TLC and LCMS.
  • the reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • Example S-24 Synthesis of 3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 111), (R)-3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No.
  • the title compound was purified by chiral HPLC to get the two enantiomers (R)-3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide and (S)-3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide.
  • Step-1 Synthesis of 5-(7-chloro-1H-indazol-5-yl)-6-4-fluorophenyl)pyrazin-2-amine.
  • 6-chloro-5-(7-chloro-1H-indazol-5-yl)pyrazin-2-amine 500 mg, 1.79 mmol, 1 eq
  • DMF:water 10 mL: 5 mL
  • 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (447 mg, 2.15 mmol, 1.2 eq).
  • reaction mixture was purged with nitrogen for 5 min then charged with K 3 PO 4 (1.2 g, 5.37 mmol, 3.0 eq) and Pd(PPh 3 ) 4 (82 mg, 0.07 mmol, 0.04 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 120° C. for 1 h using microwave irradiation. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and concentrated under vacuum. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • Step-2 Synthesis of 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine.
  • NBS 109 mg, 0.61 mmol, 1.0 eq
  • Step-3 Synthesis of (R)-3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To a stirred solution of 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine (138 mg, 0.34 mmol, 1 eq) in DMF (10 mL) was added (1R)-1-(pyridin-2-yl)ethanamine (83 mg, 2.15 mmol, 1.2 eq).
  • reaction mixture was purged with nitrogen for 5 min then charged with Mo(CO) 6 (33 mg, 0.12 mmol, 0.37 eq) and PdCl 2 dppf (12 mg, 0.01 mmol, 0.05 eq).
  • the reaction mixture was again purged with nitrogen for 5 min and then stirred at RT for 1 h followed by the addition of DBU (0.1 mL, 0.74 mmol, 2.2 eq).
  • the reaction mixture was stirred at RT for 5 min and then heated at 120° C. for 3 h.
  • the reaction was monitored by TLC and LCMS.
  • the reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL).
  • Step-1 Synthesis of 3-bromo-5-(8-chloroquinolin-6-yl)-6-phenylpyrazin-2-amine: To a solution of 5-(8-chloroquinolin-6-yl)-6-phenylpyrazin-2-amine (1.6 g, 4.81 mmol, 1 eq) in DMF (20 mL) was added N-bromosuccinimide (0.85 g, 4.81 mmol, 1 eq) at 0° C. The reaction mixture was stirred at same temperature for 2 h. The reaction was monitored by TLC. The reaction was added with water and the solid precipitates out. The solid was filtered and dried to use for next step without further purification (1.1 g, 55%). LCMS: 412[M+1] +
  • Step-2 Synthesis of (R)-3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide: To a solution of 3-bromo-5-(8-chloroquinolin-6-yl)-6-phenylpyrazin-2-amine (200 mg, 0.48 mmol, 1 eq) in DMF (10 mL) was added (R)-6-(1-aminoethyl)picolinonitrile hydrochloride (92 mg, 0.58 mmol, 1.2 eq) The reaction mixture was deoxygenated using N 2 atmosphere then charged MO(CO) 6 (51.3 mg, 0.19 mmol, 0.4 eq) and PdCl 2 (dppf).DCM complex (20 mg, 0.024 mmol, 0.05 eq), deoxygenating was continued for further 20 min.
  • MO(CO) 6 5
  • Example S-27 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide (Compound No. 114), (R)-3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide (Compound No.
  • Step-1 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2-carbonitrile: To a stirred solution of CuI (554 mg, 2.92 mmol, 1.5 eq) and CuCN (384 mg, 4.28 mol, 2.2 eq) in dry DMF (150 mL) was added 3-bromo-5-(8-chloroquinolin-6-yl)-6-phenylpyrazin-2-amine (800 mg, 1.94 mmol, 1 eq) at 100° C. The reaction mixture was stirred at 120° C. for 12 h. The reaction was monitored by TLC and LCMS. The crude product was poured in ice-water the solid precipitate out.
  • Step-2 Synthesis 3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2-carboxylic acid: To a solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2-carbonitrile (700 mg, 1.96 mmol, 1.0 eq) in ethanol (10 mL) was added aqeuous NaOH (6M, 10 mL) at 0° C. The reaction mixture was stirred at 120° C. The reaction was monitored by TLC. The reaction was distilled then charged ice water then acidifies with dilute HCl solid precipitates out. The solid was filtered and dried to use it for next step without further purification (610 g, 82%). LCMS: 377 [M+1] + .
  • Step-3 Synthesis of (R)-3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide: To a stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2-carboxylic acid (200 mg, 0.53 mmol, 1.0 eq) in DMF (10 mL) was added 6-(1-aminoethyl)picolinonitrile (94 mg, 0.63 mmol, 1.2 eq) and the mixture was stirred at RT for 5 min.
  • Example S-28 Synthesis of (R)-3-amino-5-(3-cyanophenyl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 117) and (S)-3-amino-5-(3-cyanophenyl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 118)
  • Step-1 Synthesis of methyl 3-amino-5-(3-cyanophenyl)-6-(quinolin-6-yl)pyrazine-2-carboxylate: To a stirred solution of 3-[6-amino-5-bromo-3-(quinolin-6-yl)pyrazin-2-yl]benzonitrile (700 mg, 1.74 mmol, 1.0 eq) in DMF (5 mL) and MeOH (10 mL). The reaction mixture was purged with nitrogen for 5 min then charged with Mo(CO) 6 (170 mg, 0.64 mmol, 0.37 eq) and PdCl 2 dppf (63 mg, 0.08 mmol, 0.05 eq).
  • Step-2 Synthesis of 3-amino-5-(3-cyanophenyl)-6-(quinolin-6-yl)pyrazine-2-carboxylic acid.
  • methyl 3-amino-5-(3-cyanophenyl)-6-(quinolin-6-yl)pyrazine-2-carboxylate 200 mg, 0.52 mmol, 1.0 eq
  • ethanol 5 mL
  • THF 5 mL
  • LiOH.H 2 O 88 mg, 2.09 mmol, 4.0 eq
  • Step-3 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To stirred solution of of 3-amino-5-(3-cyanophenyl)-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (100 mg, 0.27 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine (80 mg, 0.54 mmol, 2.0 eq), DIPEA (0.2 mL, 0.81 mmol, 3 eq) and HATU (205 mg, 0.54 mmol, 2 eq) at RT under inert condition.
  • Example S-29 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 119), (R)-3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No.
  • Step-1 Synthesis of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carbonitrile).
  • 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine 500 mg, 1.24 mmol, 1.0 eq
  • cuprous cyanide 122 mg, 1.36 mmol, 1.1 eq
  • copper iodide 354 mg, 1.86 mmol, 1.5 eq.
  • the reaction mixture was allowed to stir at 120° C.
  • Step-2 Synthesis of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxylic acid FK-GRF-633-84)
  • 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carbonitrile (170 mg, 0.48 mmol, 1.0 eq) in ethanol (5 mL) was added 6M NaOH solution (5 mL).
  • the resulting reaction mixture was heated at 100° C. for 16 h.
  • the reaction mixture was allowed to cool to RT.
  • the solvent was evaporated under vacuum and acidified using 1N HCl to get the solid which was filtered and dried to get the product as yellow solid (120 mg, 67%).
  • Step-3 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide; To stirred solution of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxylic acid (120 mg, 0.33 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine (96 mg, 0.65 mmol, 2.0 eq), DIPEA (0.2 mL, 0.97 mmol, 3 eq) and HATU (247 mg, 0.65 mmol, 2 eq) at RT under inert condition.
  • Example S-30 Synthesis of (3-amino-5-phenyl-N-(1-(pyrimidin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 120), (R)-3-amino-5-phenyl-N-(1-(pyrimidin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 125) and (S)-3-amino-5-phenyl-N-(1-(pyrimidin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 126)
  • Example S-31 Synthesis of 3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-(1-methyl-1H-pyrazol-3-yl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 127)
  • Example S-32 Synthesis of (R)-3-amino-5-phenyl-N-(1-(pyrazin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 128) and (S)-3-amino-5-phenyl-N-(1-(pyrazin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 129)
  • Example S-33 Synthesis of 3-amino-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyrimidin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 460)
  • reaction mixture was again purged with nitrogen for 5 min and then stirred at RT for 1 h followed by the addition of DBU (0.2 mL, 1.04 mmol, 2.2 eq).
  • the reaction mixture was stirred at RT for 5 min and then heated at 120° C. for 3 h.
  • the reaction was monitored by TLC and LCMS.
  • the reaction mixture was filtered through celite and distilled.
  • the reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 200 mL). The combined organic layers were washed (brine), dried (anhydrous Na 2 SO 4 ) and concentrated under vacuum to get the crude which was purified by reverse phase column chromatography to get the title compound (10 mg, 4.0%).
  • reaction mixture was again purged with nitrogen for 5 min and then stirred at RT for 1 h followed by the addition of DBU (0.2 mL, 1.15 mmol, 2.2 eq).
  • the reaction mixture was stirred at RT for 5 min and then heated at 120° C. for 4 h.
  • the reaction was monitored by TLC and LCMS.
  • the reaction mixture was filtered through celite and distilled.
  • the reaction was diluted with water and extracted with ethyl acetate (3 ⁇ 100 mL). The combined organic layers were washed (brine), dried (anhydrous Na 2 SO 4 ) and concentrated under vacuum to get the crude which was purified by reverse phase column chromatography to get the title compound (7 mg, 3%).
  • Binding of selected compounds to the adenosine A 2A , A 1 , A 2B , and A 3 receptors is tested using a binding competition assay.
  • the general protocol for the radioligand binding competition assay is as follows. Competition binding is performed in duplicate in the wells of a 96 well plate (Master Block, Greiner, 786201) containing binding buffer (optimized for each receptor), membrane extracts (amount of protein/well optimized for each receptor), radiotracer (final concentration optimized for each receptor), and test compound. Nonspecific binding is determined by co-incubation with 200-fold excess of cold competitor. The samples are incubated in a final volume of 0.1 mL at 25° C. for 60 minutes and then filtered over filter plates. Filters are washed six times with 0.5 mL of ice-cold washing buffer (optimized for each receptor) and 50 ⁇ L of Microscint 20 (Packard) are added on each filter. The filter plates are sealed, incubated 15 min on an orbital shaker and scintillation counted with a TopCount for 30 sec/filter.
  • a 2A adenosine receptor radioligand binding assay For the A 2A adenosine receptor radioligand binding assay, the following modifications are made to the general protocol. GF/C filters (Perkin Elmer, 6005174), presoaked in 0.01% Brij for 2 h at room temperature are used. Filters are washed six times with 0.5 mL of ice-cold washing buffer (50 mM Tris pH 7.4) and 50 ⁇ L of Microscint 20 (Packard) are added in each well. The plates are then incubated for 15 min on an orbital shaker and then counted with a TopCountTM for 1 min/well. Another radioligand binding assay used to evaluate the binding affinity for the adenosine A 2A receptor assay is performed in duplicate in the wells of a 384 plate.
  • Assay buffer contains DPBS 500 mM, MgCl 2 0.1 mM, and 1% DMSO.
  • Membrane-bead suspension is prepared by mixing 25.98 ⁇ L of human adenosine A 2A membrane preparation (Perkin Elmer, RBHA2AM400UA) at 33.4 ⁇ g/mL, 28 ⁇ L of ADA at 20 ⁇ g/mL, and 932 ⁇ L of SPA beads at 3.33 mg/mL) and the mixture is incubated for 20 min at room temperature.
  • 20 ⁇ L of radiotracer ( 3 H—SCH 58261) at 15 nM is mixed into each well containing test articles at various concentrations and the plate is centrifuged at 1000 rpm for 1 minute.
  • adenosine A 1 radioligand binding competition assay For the adenosine A 1 radioligand binding competition assay, a similar procedure is used except that the following reagents are used: CHO-K1-A1 cell membranes; binding buffer comprising HEPES 25 mM pH 7.4, MgCl 2 5 mM, CaCl 2 ) 1 mM, NaCl 100 mM, saponin 10 ⁇ g/mL; wash buffer comprising HEPES 25 mM pH 7.4, MgCl 2 5 mM, CaCl 2 ) 1 mM, NaCl 100 mM; a Unifilter GF/B—treated for 2 h with 0.5% PEI; and 1.6 nM of 3 H-DPCPX as the tracer.
  • binding buffer comprising HEPES 25 mM pH 7.4, MgCl 2 5 mM, CaCl 2 ) 1 mM, NaCl 100 mM, saponin 10 ⁇ g/mL
  • HEK-293-A 2B cell membranes 20 ⁇ g/well, preincubated 30 min at RT with 25 ⁇ g/mL Adenosine Deaminase; a binding buffer comprising HEPES 10 mM pH 7.4, EDTA 1 mM, 0.5% BSA; a wash buffer comprising HEPES 10 mM pH 7.4, EDTA 1 mM; a Unifilter GF/C—treated for 2 h with 0.5% PEI; and 10 nM 3 H-DPCPX as the tracer.
  • CHO-K1-A3 cell membranes 1.5 ⁇ g/well; a binding buffer comprising HEPES 25 mM pH 7.4, MgC 2 5 mM, CaCl 2 ) 1 mM, 0.5% BSA; a wash buffer comprising HEPES 25 mM pH 7.4, MgCl 2 5 mM, CaCl 2 1 mM; a Unifilter GF/C—treated for 2 h with 0.5% BS; and 0.4 nM of 125 I-AB-MECA as the tracer.
  • the results of the binding assay are given as percent residual binding at a given concentration. Percent of residual binding means binding of a compound in the presence of competitor normalized to the amount of binding in the absence of competitor.
  • a second A 2A adenosine receptor radioligand binding assay protocol was used.
  • the protocol used adenosine A2a (human) membrane (PerkinElmer RBHA2AM400UA) at a concentration of 5 ⁇ g/well/100 ⁇ l and the radioligand [3H] CGS-21680 (Cat No. PerkinElmer-NET1021250UC) at a final concentration of 6 nM.
  • Testing compounds were diluted with DMSO to make 8-point 4-fold serial dilution, starting at 0.2 mM.
  • CGS-15943 was the reference compound.
  • 1 ⁇ l of compounds/high control/low control was transferred to the assay plate according to a plate map, followed by 100 ⁇ l of membrane stocks and 100 ⁇ l of radioligand, in assay buffer (50 mM Tris-HCl, 10 mM MgCl 2 , 1 mM EDTA, pH 7.4). The plate was sealed and incubated at RT for 2 hours. Unifilter-96 GF/C filter plates (Perkin Elmer Cat #6005174) were soaked with 50 ⁇ l of 0.3% PEI per well for at least 0.5 hour at room temperature.
  • the reaction mixtures were filtered through GF/C plates using Perkin Elmer Filtermate Harvester, and each plate washed 4 times with cold wash buffer (50 mM Tris-HCl, 154 mM NaCl, pH 7.4).
  • the filter plates were dried for 1 hour at 50 degrees. After drying, the bottom of the filter plate wells was sealed, 50 ⁇ l of Perkin Elmer Microscint 20 cocktail was added, and the top of the filter plate was sealed. 3 H trapped on the filter was counted using Perkin Elmer MicroBeta2 Reader. The data were analyzed with GraphPad Prism 5 to obtain binding IC 50 values.
  • a second A 1 adenosine receptor radioligand binding assay protocol was used.
  • the protocol used adenosine A1 (human) membrane (PerkinElmer ES-010-M400UA) at a concentration of 2.5 ⁇ g/well/100 ⁇ l and the radioligand [3H] DPCPX (Cat No. PerkinElmer-NET974250UC) at a final concentration of 1 nM.
  • Testing compounds were tested at a final concentration of 200 nM.
  • CGS-15943, the reference compound was tested in an 8-point 4-fold serial dilution, starting at a top concentration of 1 ⁇ M.
  • 1 ⁇ l of compounds/high control/low control was transferred to the assay plate according to a plate map, followed by 100 ⁇ l of membrane stocks and 100 ⁇ l of radioligand, in assay buffer (25 mM HEPES, 5 mM MgCl 2 , 1 mM CaCl 2 ), 100 mM NaCl, pH 7.4).
  • assay buffer 25 mM HEPES, 5 mM MgCl 2 , 1 mM CaCl 2
  • 100 mM NaCl, pH 7.4 100 mM NaCl, pH 7.4
  • Assay 2 The functional activity of compounds was tested using Assay 2, below, to detect the presence of cAMP.
  • Assay 1 is an alternative assay for this purpose.
  • Activation of G-protein coupled receptors results in activation of adenylyl cyclase which converts ATP into cAMP which is used as a downstream signaling molecule. Therefore, molecules which act as GPCR (or specifically A 2A receptor) antagonists cause a decrease in intracellular cAMP concentration.
  • Assay 1 This assay uses HEK-293 cells expressing human recombinant adenosine A 2A receptor that are grown prior to the test in media without antibiotic. The cells are detached by gentle flushing with PBS-EDTA (5 mM EDTA), recovered by centrifugation and suspended in assay buffer (KRH: 5 mM KCl, 1.25 mM MgSO 4 , 124 mM NaCl, 25 mM HEPES, 13.3 mM Glucose, 1.25 mM KH 2 PO 4 , 1.45 mM CaCl 2 ), 0.5 g/L BSA, supplemented with Rolipram).
  • PBS-EDTA 5 mM EDTA
  • assay buffer KRH: 5 mM KCl, 1.25 mM MgSO 4 , 124 mM NaCl, 25 mM HEPES, 13.3 mM Glucose, 1.25 mM KH 2 PO 4 , 1.45
  • Assay 2 (Table B2): This assay used HEK-293 cells expressing human recombinant adenosine A 2A receptor (or, alternatively, A 1 receptor) that were grown prior to the test in media without antibiotic. 100 nL of test articles at 100 ⁇ of final concentration were transferred to assay plate by Echo. Cells were washed twice with 5 mL of PBS and 10 ⁇ L of cells were mixed with 5 mL PBS. After aspirating the PBS and adding 1.5 mL versine, cells were incubated at 37° C. for 2-5 min. After centrifugation, 4 mL of medium was added and adjusted cell density to 5,000 cells/well with Stimulation Buffer.
  • a scintillation proximity assay (SPA) is used to determine the kinetic profile of the binding of test compound to the A 2A receptor.
  • membrane extracts are prepared from HEK-293 cells expressing recombinant human A 2A receptor, are mixed with GDP (volume:volume) and are incubated in assay buffer comprising 20 mM HEPES pH 7.4; 100 mM NaCl, 10 ⁇ g/mL saponin, 5 mM MgCl 2 for at least 15 min on ice.
  • assay buffer comprising 20 mM HEPES pH 7.4; 100 mM NaCl, 10 ⁇ g/mL saponin, 5 mM MgCl 2 for at least 15 min on ice.
  • GTP ⁇ [ 35 S] is mixed with the beads (volume:volume) just before starting the reaction.
  • PBMCs peripheral blood mononuclear cells
  • Fresh human blood is diluted with the same volume of PBS and the buffy coat containing peripheral blood mononuclear cells (PBMCs) is prepared and resuspended in culture medium at a density of 2 ⁇ 10 6 /mL.
  • 2 ⁇ 10 5 PBMCs (in 100 ⁇ L) are plated to each well of a 96-well flat bottom plate.
  • 25 ⁇ L of 8 ⁇ final concentration of 10-fold serial diluted or single concentration compounds are added to indicated wells and incubated for 30 mins in 37° C./5% CO 2 .
  • 25 ⁇ L of 8 ⁇ final concentration of NECA (1 ⁇ M) is added to indicated wells and incubated for 30 min in 37° C./5% CO 2 .
  • T cell activation/expansion kit (Miltenyi biotec Cat #130-091-441) at a bead-to-cell ratio of 1:6 in 50 ⁇ L is added to all wells with the final concentration of DMSO at 0.1% and final volume at 200 ⁇ L. 60 ⁇ L of supernatant post 24 hr and 48 hr incubation is collected for TNF- ⁇ and IFN- ⁇ concentration evaluation using TNF- ⁇ ELISA ready-set-go kit (eBioscience, Cat #88-7346-77) and IFN- ⁇ ELISA ready-set-go kit (eBioscience, Cat #88-7316-77), respectively.
  • TNF- ⁇ ELISA ready-set-go kit eBioscience, Cat #88-7346-77
  • IFN- ⁇ ELISA ready-set-go kit eBioscience, Cat #88-7316-77
  • CD8 + T-cells (1 ⁇ 105) are cultured alone, with 3 ⁇ M of NECA, or in the presence of 1 ⁇ M of the compound of interest with or without 3 ⁇ M of NECA.
  • the cells are incubated for 30 min at 37° C. and 5% CO 2 , and the reaction is stopped by addition of 200 ⁇ L, 0.1 M hydrochloric acid.
  • cAMP levels are determined by an ELISA kit.
  • mice are evaluated in selective mouse models (e.g., syngeneic model, xenograft model, or PDX) as a monotherapy or combination therapies.
  • MC-38 syngeneic model as an example: female C 57 BL/6 mice are inoculated subcutaneously at right flank with MC-38 cells for tumor development. Five days after tumor inoculation, mice with tumor size ranging from 40-85 mm 3 are selected and assigned into sub-groups using stratified randomization with 10 mice per group based upon their tumor volumes. Mice receive pre-defined treatments include vehicle, test article at various doses alone, test article at various doses plus other anti-cancer therapy, and other anti-cancer therapy control.
  • the tumor sizes are used for the calculations of both tumor growth inhibition (TGI) and T/C values.
  • TGI tumor growth inhibition
  • the termination endpoint e.g., with TV >1000 mm 3
  • the time from inoculation to the termination are deemed as its survival time. Survival curves are plotted by the Kaplan-Meier method.
  • plasma and tumor samples are collected to explore biomarkers.
  • IC 50 values of compounds for reversal of NECA suppression of mIFN ⁇ release is determined in mouse splenocytes isolated from Balb/c mice.
  • the mIFN ⁇ release is CD3e/CD28-induced release.
  • Mouse splenocytes (2 ⁇ 10 5 cells/well) are activated with Anti-mouse CD3e (2.5 ⁇ g/ml, coated overnight at 4° C.; Cat #14-0032-82, eBioscience) and then incubated with serial dilutions of compounds (3 fold, 8 point dose response starting at 1 ⁇ M) in the presence of NECA (at a concentration such as 0.1, 3.0, or 6.0 ⁇ M; Cat #E2387, Sigma) for 30 min at 37° C., 5% CO 2 in an incubator (cell culture conditions) prior to treating them with Anti-mouse CD28 (0.1 ⁇ g/ml soluble; Cat #16-0289-81, eBiosciences).
  • Splenocytes are further incubated under cell culture conditions for 72 hr; the supernatant is then harvested and diluted to 1:100, and ELISA is performed as per the manufacturer's protocol (mIFN- ⁇ kit; Cat #555138 and 550534, BD Biosciences). Plates are read in a plate reader by measuring absorbance at 450 nm. Values for the reversal of NECA suppressed mIFN- ⁇ release by compounds are calculated by the following formula:
  • [mIFN- ⁇ ] test is the test reading
  • [mIFN- ⁇ ] blank is the average reading obtained from blank wells
  • [mIFN- ⁇ ] NECA is the average reading obtained from NECA treated, activated cells.
  • the IC 50 values are calculated by fitting the curve to the “four-parameter variable slope logistic model” using Graph Pad Prism.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

5,6-disubstituted 2-aminopyrazine compounds as modulators of an adenosine receptor are provided. The compounds may find use as therapeutic agents for the treatment of diseases mediated through a G-protein-coupled receptor signaling pathway and may find particular use in oncology.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 16/46,763, filed Jan. 17, 2020, which claims priority to U.S. Application Ser. No. 62/794,537, filed Jan. 18, 2019, and U.S. Application Ser. No. 62/796,046, filed Jan. 23, 2019, each of which is hereby incorporated herein by reference in its entirety.
  • FIELD OF THE INVENTION
  • This disclosure relates generally to therapeutics for treatment mediated through a G-protein-coupled receptor (GPCR) signaling pathway and, more particularly, to compounds that inhibit an adenosine receptor (such as an A2A antagonist). The disclosure also provides pharmaceutically acceptable compositions comprising such compounds and methods of using the compounds or compositions in the treatment of a disease associated with a GPCR signaling pathway
  • BACKGROUND OF THE INVENTION
  • Adenosine receptors (ARs) are distributed throughout the body and are responsible for numerous biological functions. The seven trans-membrane G-protein-coupled receptors (GPCRs) have been divided into four different subtypes: A1, A2A, A2B, and A3. The A2A and A2B ARs stimulate activity of the adenylyl cyclase, inducing an increase of cAMP levels. A2A ARs have a distinct tissue localization, different biochemical pathways, and specific pharmacological profiles.
  • Adenosine is one of the human body's most important neuromodulators in both the central and the peripheral nervous systems. Adenosine is released from tumor cells and its concentration in the extracellular fluid of tumors can reach immunosuppressive levels (Blay et al. (1997), Cancer Res., 57(13), pp. 2602-5). The extracellular fluid of solid carcinomas contains immunosuppressive concentrations of adenosine. Id. This increase in adenosine concentration is a result of increases in CD73 (ecto-5′-nucleotidase) and CD39 (nucleoside triphosphate dephosphorylase) enzymes, which are responsible for directly catabolizing ATP into adenosine. These upregulations are triggered by hypoxia and the generation of HIF-1α. High levels of adenosine around tumor cells act to regulate multiple immune cells (e.g., CD4+ T-cells and cytotoxic CD8+ T-cells) via activation of multiple adenosine receptor subtypes, but particularly A2A receptors, resulting in the suppressing of pro-inflammatory activities and upregulation of anti-inflammatory molecules and immunoregulatory cells (Kumar et al. (2013), Adenosine as an endogenous immunoregulator in cancer pathogenesis: where to go? Purinergic Signal., 9(2), pp 145-65 and Sitkowsky et al., Hostile, hypoxia-A2-adenosinergic tumor biology as the next barrier to overcome for tumor immunologists. Cancer Immunol. Res. 2(7), pp 598-605; Ohta (2016), A Metabolic Immune Checkpoint: Adenosine in Tumor Microenvironment. Frontiers in Immunology., 7 article #109, pp 1-11). It was demonstrated that chimeric antigen receptor (CAR) T cells upregulate A2ARs upon antigen-specific stimulation in vitro and in vivo (Beavls (2017), Targeting the Adenosine 2A Receptor Enhances Chimeric Antigen Receptor T Cell Efficacy. J of Clin Invest. 127 (3): pp 929-941).
  • Survival of cancer cells is dependent on their ability to avoid attack by the immune system. In addition, tumor cells can overtake the immune system to facilitate tumor survival and metastasis. Adenosine, whose concentration increases within hypoxic regions of solid tumors, has been recognized as being able to interfere with the recognition of tumor cells by cytolytic effector cells of the immune system. (Tuite and Riss (2013). Recent developments in the pharmacological treatment of Parkinson's disease. Expert Opin. Investig. Drugs, 12(8) pp 1335-52, Popoli et al. (2002). Blockade of striatal adenosine A2A receptor reduces, through a presynaptic mechanism, quinolinic acid-induced excitotoxicity: possible relevance to neuroprotective interventions in neurodegenerative diseases of the striatum, J. Neurosci, 22(5) pp. 1967-75, Gessi et al. (2011). Adenosine receptors and cancer. Biochim Biophys Acta, 1808(5), pp. 1400-12).
  • Although all adenosine receptors now have an increasing number of recognized biological roles in tumors, the A2A and A3 subtypes appear promising targets for therapeutic development. In particular, activation of A2A receptors leads to immunosuppressive effects, which decreases anti-tumoral immunity and thereby encourages tumor growth.
  • The A2B receptor is another potential target for therapeutic development. Autocrine/paracrine stimulation of A2B expressed on tumor cells is believed to enhance their metastatic potential and A2B blockade may reduce tumor metastasis in an immune-independent manner (Beavis et al. (2013). Blockade of A2A receptors potently suppresses the metabolism of CD73+ Tumors. Proc. Natl. Acad. Sci., 110(36) pp. 14711-6). A2B expression also correlates with relapse-free survival (RFS) in triple negative breast cancer suggesting that this pathway may be clinically relevant. A2B blockade also has the potential to modulate the immunosuppressive properties of tumor-associated immune cells including dendritic cells and myeloid-derived suppressor cells (MDSCs) (Cekic et al. (2011). Adenosine A2B receptor blockade slows growth of bladder and breast tumors. J. Immunol. 188(1), pp. 198-205; Sorrentino et al. (2015). Myeloid-derived suppressor cells contribute to A2B adenosine receptor-induced VEGF production and angiogenesis in a mouse melanoma model. Oncotarget 6(29), pp. 27478-89; Iannone et al. (2013). Blockade of A2B adenosine receptor reduces tumor growth and immune suppression mediated by myeloid-derived suppressor cells in a mouse model of melanoma. Neoplasia, 15(12), pp. 1400-9.
  • There remains a continuing need for new therapies for the treatment of diseases and disorders related to the adenosine signaling pathway.
  • BRIEF SUMMARY OF THE INVENTION
  • In one aspect, provided is a compound of the formula (I):
  • Figure US20220298142A1-20220922-C00001
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein A, B, R1 and R2 are as detailed herein. In some embodiments, provided is a compound of formula (I), or a salt thereof.
  • In one aspect, provided is a compound of the formula (II):
  • Figure US20220298142A1-20220922-C00002
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein L, Q1, Q2, A, B and D are as detailed herein. In some embodiments, provided is a compound of formula (II), or a salt thereof.
  • In some embodiments, the compound of the formula (I), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, is of the formula (II) or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, as detailed herein.
  • In another aspect, provided is a compound of the formula (III):
  • Figure US20220298142A1-20220922-C00003
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein A and B are as detailed herein.
  • In another aspect, provided is a method for any one or more of: (a) treating a disease, such as a proliferative disease, in an individual in need thereof; (b) enhancing an immune response in an individual in need thereof; (c) inhibiting tumor metastasis in an individual in need thereof; (d) modulating the activity of a G protein coupled receptor signaling pathway in an individual in need thereof; (e) modulating the activity of an adenosine receptor, such as an A2A receptor, in an individual in need thereof; and (f) increasing the activity of a natural killer cell in an individual in need thereof, wherein the method comprises administering to the individual an effective amount of a compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing. In some embodiments, provided is a method for any one or more of: (a) treating a disease, such as a proliferative disease, in an individual in need thereof; (b) enhancing an immune response in an individual in need thereof; (c) inhibiting tumor metastasis in an individual in need thereof; (d) modulating the activity of a G protein coupled receptor signaling pathway in an individual in need thereof; (e) modulating the activity of an adenosine receptor, such as an AA receptor, in an individual in need thereof; and (f) increasing the activity of a natural killer cell in an individual in need thereof, wherein the method comprises administering to the individual an effective amount of a compound of formulae (I), (II) or (III), or a salt thereof. In one aspect, the compound of formulae (I), (II) or (III), or a salt thereof is administered to the individual in combination with another therapeutic agent. In some embodiments, the compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing is administered to the individual in combination with another therapeutic agent. In some embodiments, the compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing is a compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • Also provided are pharmaceutical compositions comprising (A) a compound detailed herein, such as a compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, or a compound of formulae (I), (II) or (III), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, and (B) a pharmaceutically acceptable carrier or excipient. In some embodiments, provided are pharmaceutical compositions comprising (A) a compound detailed herein, such as a compound of formulae (I), (II) or (III), or a salt thereof, and (B) a pharmaceutically acceptable carrier or excipient. Kits comprising a compound detailed herein or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing and instructions for use are also provided. Kits comprising a compound detailed herein or a salt thereof and instructions for use are also provided. A compound detailed herein or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing is also provided for the manufacture of a medicament for the treatment of cancer. Compounds as detailed herein or a pharmaceutically acceptable salt thereof are also provided for the manufacture of a medicament for the treatment of cancer.
  • DETAILED DESCRIPTION OF THE INVENTION Definitions
  • For use herein, unless clearly indicated otherwise, use of the terms “a”, “an” and the like refers to one or more.
  • “Alkenyl” as used herein refers to an unsaturated linear or branched univalent hydrocarbon chain or combination thereof, having at least one site of olefinic unsaturation (i.e., having at least one moiety of the formula C═C) and having the number of carbon atoms designated (i.e., C2-C10 means two to ten carbon atoms). The alkenyl group may be in “cis” or “trans” configurations, or alternatively in “E” or “Z” configurations. Particular alkenyl groups are those having 2 to 20 carbon atoms (a “C2-C20 alkenyl”), having 2 to 8 carbon atoms (a “C2-C8 alkenyl”), having 2 to 6 carbon atoms (a “C2-C6 alkenyl”), or having 2 to 4 carbon atoms (a “C2-C4 alkenyl”). Examples of alkenyl include, but are not limited to, groups such as ethenyl (or vinyl), prop-1-enyl, prop-2-enyl (or allyl), 2-methylprop-1-enyl, but-1-enyl, but-2-enyl, but-3-enyl, buta-1,3-dienyl, 2-methylbuta-1,3-dienyl, homologs and isomers thereof, and the like.
  • The term “alkyl” refers to and includes saturated linear and branched univalent hydrocarbon structures and combination thereof, having the number of carbon atoms designated (i.e., C1-C10 means one to ten carbons). Particular alkyl groups are those having 1 to 20 carbon atoms (a “C1-C20 alkyl”). More particular alkyl groups are those having 1 to 8 carbon atoms (a “C1-C8 alkyl”), 3 to 8 carbon atoms (a “C3-C8 alkyl”), 1 to 6 carbon atoms (a “C1-C6 alkyl”), 1 to 5 carbon atoms (a “C1-C5 alkyl”), or 1 to 4 carbon atoms (a “C1-C4 alkyl”). Examples of alkyl include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • “Alkylene” as used herein refers to the same residues as alkyl, but having bivalency. Particular alkylene groups are those having 1 to 6 carbon atoms (a “C1-C6 alkylene”), 1 to 5 carbon atoms (a “C1-C5 alkylene”), 1 to 4 carbon atoms (a “C1-C4 alkylene”) or 1 to 3 carbon atoms (a “C1-C3 alkylene”). Examples of alkylene include, but are not limited to, groups such as methylene (—CH2—), ethylene (—CH2CH2—), propylene (—CH2CH2CH2—), butylene (—CH2CH2CH2CH2—), isopropylene (—CH2C(H)(CH3)CH2—), and the like.
  • “Alkynyl” as used herein refers to an unsaturated linear or branched univalent hydrocarbon chain or combination thereof, having at least one site of acetylenic unsaturation (i.e., having at least one moiety of the formula C≡C) and having the number of carbon atoms designated (i.e., C2-C10 means two to ten carbon atoms). Particular alkynyl groups are those having 2 to 20 carbon atoms (a “C2-C20 alkynyl”), having 2 to 8 carbon atoms (a “C2-C8 alkynyl”), having 2 to 6 carbon atoms (a “C2-C6 alkynyl”), or having 2 to 4 carbon atoms (a “C2-C4 alkynyl”). Examples of alkynyl include, but are not limited to, groups such as ethynyl (or acetylenyl), prop-1-ynyl, prop-2-ynyl (or propargyl), but-1-ynyl, but-2-ynyl, but-3-ynyl, homologs and isomers thereof, and the like.
  • The term “aryl” refers to and includes polyunsaturated aromatic hydrocarbon groups. Aryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings. In one variation, the aryl group contains from 6 to 14 annular carbon atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, biphenyl, and the like.
  • The term “cycloalkyl” refers to and includes cyclic univalent hydrocarbon structures, which may be fully saturated, mono- or polyunsaturated, but which are non-aromatic, having the number of carbon atoms designated (e.g., C1-C10 means one to ten carbons). Cycloalkyl can consist of one ring, such as cyclohexyl, or multiple rings, such as adamantyl, but excludes aryl groups. A cycloalkyl comprising more than one ring may be fused, spiro or bridged, or combinations thereof. A preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 13 annular carbon atoms. A more preferred cycloalkyl is a cyclic hydrocarbon having from 3 to 8 annular carbon atoms (a “C3-C8 cycloalkyl”). Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, norbornyl, and the like.
  • “Halo” or “halogen” refers to elements of the Group 17 series having atomic number 9 to 85. Preferred halo groups include fluoro, chloro, bromo and iodo. Where a residue is substituted with more than one halogen, it may be referred to by using a prefix corresponding to the number of halogen moieties attached, e.g., dihaloaryl, dihaloalkyl, trihaloaryl etc. refer to aryl and alkyl substituted with two (“di”) or three (“tri”) halo groups, which may be but are not necessarily the same halo; thus 4-chloro-3-fluorophenyl is within the scope of dihaloaryl. An alkyl group in which each hydrogen is replaced with a halo group is referred to as a “perhaloalkyl.” A preferred perhaloalkyl group is trifluoroalkyl (—CF3). Similarly, “perhaloalkoxy” refers to an alkoxy group in which a halogen takes the place of each H in the hydrocarbon making up the alkyl moiety of the alkoxy group. An example of a perhaloalkoxy group is trifluoromethoxy (—OCF3).
  • The term “heteroaryl” refers to and includes unsaturated aromatic cyclic groups having from 1 to 10 annular carbon atoms and at least one annular heteroatom, including but not limited to heteroatoms such as nitrogen, oxygen and sulfur, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized. A heteroaryl group can be attached to the remainder of the molecule at an annular carbon or at an annular heteroatom. Heteroaryl may contain additional fused rings (e.g., from 1 to 3 rings), including additionally fused aryl, heteroaryl, cycloalkyl, and/or heterocyclyl rings. Examples of heteroaryl groups include, but are not limited to, pyridyl, pyrimidyl, thiophenyl, furanyl, thiazolyl, and the like. Examples of heteroaryl groups also include, but are not limited to, pyridyl, pyrimidyl, thiophenyl, furanyl, thiazolyl, oxazolyl, isoxazolyl, thiophenyl, pyrrolyl, pyrazolyl, 1,3,4-oxadiazolyl, imidazolyl, isothiazolyl, triazolyl, 1,3,4-thiadiazolyl, tetrazolyl, benzofuranyl, benzothiophenyl, pyrazolopyridinyl, indazolyl, benzothiazolyl, benzooxazolyl or benzoimidazolyl and the like.
  • In one variation, a heteroaryl containing at least one additional fused ring that is nonaromatic (e.g., cycloakyl or heterocyclyl) is attached to the parent structure at an annular atom of the additional ring. In another variation, a heteroaryl containing at least one additional ring that is nonaromatic (e.g., cycloakyl or heterocyclyl) is attached to the parent structure at an annular atom of the aromatic ring.
  • The term “heterocycle” or “heterocyclyl” refers to a saturated or an unsaturated non-aromatic group having from 1 to 10 annular carbon atoms and from 1 to 4 annular heteroatoms, such as nitrogen, sulfur or oxygen, and the like, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized. A heterocyclyl group may have a single ring or multiple condensed rings, but excludes heteroaryl groups. A heterocycle comprising more than one ring may be fused, spiro or bridged, or any combination thereof. In fused ring systems, one or more of the fused rings can be aryl, cycloalyl or heterocyclyl. Examples of heterocyclyl groups include, but are not limited to, tetrahydropyranyl, dihydropyranyl, piperidinyl, piperazinyl, pyrrolidinyl, thiazolinyl, thiazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, 2,3-dihydrobenzo[b]thiophen-2-yl, 4-amino-2-oxopyrimidin-1(2H)-yl, and the like.
  • In one variation, a heterocyclyl containing at least one additional ring (such as a fused additional ring) that does not contain a heteroatom is attached to the parent structure at an annular atom of the additional ring. In another variation, a heterocyclyl containing at least one additional ring (such as a fused additional ring) that does not contain a heteroatom is attached to the parent structure at an annular atom of the ring containing a heteroatom.
  • “Oxo” refers to the moiety ═O.
  • “Optionally substituted” unless otherwise specified means that a group may be unsubstituted or substituted by one or more (e.g., 1, 2, 3, 4 or 5) of the substituents listed for that group in which the substituents may be the same of different. In one embodiment, an optionally substituted group has one substituent. In another embodiment, an optionally substituted group has two substituents. In another embodiment, an optionally substituted group has three substituents. In another embodiment, an optionally substituted group has four substituents. In some embodiments, an optionally substituted group has 1 to 2, 2 to 5, 3 to 5, 2 to 3, 2 to 4, 3 to 4, 1 to 3, 1 to 4 or 1 to 5 substituents.
  • A “pharmaceutically acceptable carrier” refers to an ingredient in a pharmaceutical formulation, other than an active ingredient, which is nontoxic to a subject. A pharmaceutically acceptable carrier includes, but is not limited to, a buffer, excipient, stabilizer, or preservative.
  • As used herein, “treatment” or “treating” is an approach for obtaining beneficial or desired results including clinical results. For example, beneficial or desired results include, but are not limited to, one or more of the following: decreasing symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, delaying the progression of the disease, and/or prolonging survival of individuals. In reference to cancers or other unwanted cell proliferation, beneficial or desired results include shrinking a tumor (reducing tumor size); decreasing the growth rate of the tumor (such as to suppress tumor growth); reducing the number of cancer cells; inhibiting, retarding or slowing to some extent and preferably stopping cancer cell infiltration into peripheral organs; inhibiting (slowing to some extent and preferably stopping) tumor metastasis; inhibiting tumor growth; preventing or delaying occurrence and/or recurrence of tumor; and/or relieving to some extent one or more of the symptoms associated with the cancer. In some embodiments, beneficial or desired results include preventing or delaying occurrence and/or recurrence, such as of unwanted cell proliferation.
  • As used herein, “delaying development of a disease” means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease (such as cancer). This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. For example, a late stage cancer, such as development of metastasis, may be delayed.
  • As used herein, an “effective dosage” or “effective amount” of compound or salt thereof or pharmaceutical composition is an amount sufficient to effect beneficial or desired results. For prophylactic use, beneficial or desired results include results such as eliminating or reducing the risk, lessening the severity of, or delaying the onset of the disease, including biochemical, histological and/or behavioral symptoms of the disease, its complications and intermediate pathological phenotypes presenting during development of the disease. For therapeutic use, beneficial or desired results include ameliorating, palliating, lessening, delaying or decreasing one or more symptoms resulting from the disease, increasing the quality of life of those suffering from the disease, decreasing the dose of other medications required to treat the disease, enhancing effect of another medication such as via targeting, delaying the progression of the disease, and/or prolonging survival. In reference to cancers or other unwanted cell proliferation, an effective amount comprises an amount sufficient to cause a tumor to shrink and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation. In some embodiments, an effective amount is an amount sufficient to delay development. In some embodiments, an effective amount is an amount sufficient to prevent or delay occurrence and/or recurrence. An effective amount can be administered in one or more administrations, in the case of cancer, the effective amount of the drug or composition may: (i) reduce the number of cancer cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and preferably stop cancer cell infiltration into peripheral organs; (iv) inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer. An effective dosage can be administered in one or more administrations. For purposes of this disclosure, an effective dosage of compound or a salt thereof, or pharmaceutical composition is an amount sufficient to accomplish prophylactic or therapeutic treatment either directly or indirectly. It is intended and understood that an effective dosage of a compound or salt thereof, or pharmaceutical composition may or may not be achieved in conjunction with another drug, compound, or pharmaceutical composition. Thus, an “effective dosage” may be considered in the context of administering one or more therapeutic agents, and a single agent may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable result may be or is achieved.
  • As used herein, the term “individual” is a mammal, including humans. An individual includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate. In some embodiments, the individual is human. The individual (such as a human) may have advanced disease or lesser extent of disease, such as low tumor burden. In some embodiments, the individual is at an early stage of a proliferative disease (such as cancer). In some embodiments, the individual is at an advanced stage of a proliferative disease (such as an advanced cancer).
  • Reference to “about” a value or parameter herein includes (and describes) embodiments that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”.
  • It is understood that aspects and variations described herein also include “consisting” and/or “consisting essentially of” aspects and variations.
  • Compounds
  • In one aspect, provided is a compound of the formula (I):
  • Figure US20220298142A1-20220922-C00004
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein:
  • A is 4-hydroxyphenyl optionally further substituted by R3, 4-hydroxy-2-pyridyl optionally further substituted by R4, a naphthyl substituted by R4, a 9- or 10-membered bicyclic heterocyclyl optionally substituted by R4, or a 9- or 10-membered bicyclic heteroaryl optionally substituted by R4;
  • B is a phenyl optionally substituted by R3, C3-C6 cycloalkyl optionally substituted by R4, 3- to 6-membered heterocyclyl optionally substituted by R4 or a 5- to 10-membered heteroaryl optionally substituted by R4;
  • R1 is a hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, 5- to 10-membered heteroaryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), —(C1-C3 alkylene)(5-6-membered heteroaryl), —(C1-C3 alkylene)(C6 aryl), —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —NR1bR1c, —S(O)2R1a, —(C1-C3alkylene)C(O)NR1bR1c, —(C1-C3 alkylene)C(O)R1a or —(C1-C3 alkylene)NR1bR1c, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, 5- to 10-membered heteroaryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), —(C1-C3 alkylene)(5-6-membered heteroaryl), and —(C1-C3 alkylene)(C6 aryl) of R1 are independently optionally substituted by R4;
  • each R1a is independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, 3-6-membered heterocyclyl, C6 aryl, 5-6-membered heteroaryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3alkylene)(3-6-membered heterocyclyl), —(C1-C3 alkylene)(C6 aryl) or —(C1-C3 alkylene)(5-6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3;
  • each R1b and R1c is independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, 3-6-membered heterocyclyl, C6 aryl, 5-6-membered heteroaryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), —(C1-C3 alkylene)(C6 aryl) or —(C1-C3alkylene)(5-6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3;
      • or R1b and R1c are taken together with the nitrogen atom to which they are attached to form a 3- to 6-membered heterocyclyl;
  • R2 is —OR2a, —NHR2b, —C(O)NHR2b, or C1-C6 alkyl, wherein the C1-C6 alkyl of R2 is substituted by —OR2c, —NHR2c, or —C(O)NHR2c;
  • each R2a and R2b is independently cyclohexane, 6-membered heterocyclyl, —(C1-C3alkylene)N(C2H5)2, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), or —(C1-C3 alkylene)(5-6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3;
  • R2 is 5- or 6-membered heteroaryl, wherein the 5- or 6-membered heteroaryl is further substituted by C1-C6 alkyl optionally substituted by halogen, —OH or oxo;
  • each R3 is independently C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —OC(O)R5, —C(O)OR5, —C(O)NR6R7, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)OR6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, —NR5S(O)R6, —C(O)NR5S(O)R6, —NR5S(O)2R6, —C(O)NR5S(O)2R6, —S(O)NR6R7, —S(O)2NR6R7, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR5, —(C1-C3 alkylene)SR5, —(C1-C3 alkylene)NR6R7, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)NO2, —C═NH(OR5), —(C1-C3 alkylene)C(O)R5, —(C1-C3 alkylene)OC(O)R5, —(C1-C3 alkylene)C(O)OR5, —(C1-C3 alkylene)C(O)NR6R7, —(C1-C3 alkylene)OC(O)NR6R7, —(C1-C3 alkylene)NR5C(O)R6, —(C1-C3 alkylene)NR5C(O)OR6, —(C1-C3 alkylene)NR5C(O)NR6R7, —(C1-C3 alkylene)S(O)R5, —(C1-C3 alkylene)S(O)2R5, —(C1-C3 alkylene)NR5S(O)R6, —C(O)(C1-C3 alkylene)NR5S(O)R6, —(C1-C3 alkylene)NR5S(O)2R6, —(C1-C3 alkylene)C(O)NR5S(O)2R6, —(C1-C3 alkylene)S(O)NR6R7, —(C1-C3 alkylene)S(O)2NR6R7, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), wherein each R3 is independently optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • each R4 is independently oxo or R3;
  • R5 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)ORB, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9 or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • R8 and R9 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, OH, oxo or NH2;
  • or R8 and R9 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by halogen, OH, oxo or NH2.
  • In some embodiments, provided is a compound of the formula (I):
  • Figure US20220298142A1-20220922-C00005
  • or a salt thereof, wherein:
  • A is 4-hydroxyphenyl optionally further substituted by R3, 4-hydroxy-2-pyridyl optionally further substituted by R4, or a 9- or 10-membered bicyclic heteroaryl optionally substituted by R4;
  • B is a phenyl optionally substituted by R3, or a 5- to 6-membered heteroaryl optionally substituted by R4;
  • R1 is a hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, or —NR1bR1c, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl and 3- to 6-membered heterocyclyl of R1 are independently optionally substituted by R4;
  • each R1a is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
  • each R1b and R1c is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
      • or R1b and R1c are taken together with the nitrogen atom to which they are attached to form a 3- to 6-membered heterocyclyl;
  • R2 is —OR2a, —NHR2b, —C(O)NHR2b, or C1-C6 alkyl, wherein the C1-C6 alkyl of R2 is substituted by —OR2c, —NHR2c, or —C(O)NHR2c;
  • each R2a and R2b is independently cyclohexane, 6-membered heterocyclyl, —(C1-C3 alkylene)N(C2H5)2, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), or —(C1-C3 alkylene)(5- or 6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3;
  • R2 is 5- or 6-membered heteroaryl, wherein the 5- or 6-membered heteroaryl is further substituted by C1-C6 alkyl optionally substituted by halogen, —OH or oxo;
  • each R3 is independently C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —OC(O)R5, —C(O)OR5, —C(O)NR6R7, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)OR6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, —NR5S(O)R6, —C(O)NR5S(O)R6, —NR5S(O)2R6, —C(O)NR5S(O)2R6, —S(O)NR6R7, —S(O)2NR6R7, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR5, —(C1-C3 alkylene)SR5, —(C1-C3 alkylene)NR6R7, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)NO2, —C═NH(OR5), —(C1-C3 alkylene)C(O)R5, —(C1-C3 alkylene)OC(O)R5, —(C1-C3 alkylene)C(O)OR5, —(C1-C3 alkylene)C(O)NR6R7, —(C1-C3 alkylene)OC(O)NR6R7, —(C1-C3 alkylene)NR5C(O)R6, —(C1-C3 alkylene)NR5C(O)OR6, —(C1-C3 alkylene)NR5C(O)NR6R7, —(C1-C3 alkylene)S(O)R5, —(C1-C3 alkylene)S(O)2R5, —(C1-C3 alkylene)NR5S(O)R6, —C(O)(C1-C3 alkylene)NR5S(O)R6, —(C1-C3 alkylene)NR5S(O)2R6, —(C1-C3 alkylene)C(O)NR5S(O)2R6, —(C1-C3 alkylene)S(O)NR6R7, —(C1-C3 alkylene)S(O)2NR6R7, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), wherein each R3 is independently optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • each R4 is independently oxo or R3;
  • R5 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9 or C1-C6 alkyl optionally substituted by oxo, —OH or halogen; R8 and R9 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, OH, oxo or NH2;
      • or R8 and R9 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by halogen, OH, oxo or NH2.
  • In some embodiments, R1 is a hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, 5- to 10-membered heteroaryl, —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, or —S(O)2R1a, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, and 5- to 10-membered heteroaryl are optionally substituted with R4. In some embodiments, R1 is hydrogen, C1-C6 alkyl or —C(O)R1aIn certain embodiments, R1 is hydrogen. In certain embodiments, R1 is —C(O)R1a where R1a is C1-C6 alkyl (e.g., methyl) or C3-C6 cycloalkyl.
  • It is understood that each R1 may be combined with each R2, A and/or B the same as if each and every combination of R1 with R2, A and/or B were specifically and individually listed. For example, in some embodiments, R1 is hydrogen in formula (I), wherein R2, A and B are as defined herein.
  • In some embodiments, R2 is —OR2a. In some embodiments, R2 is —NHR2b. In some embodiments, R2 is —C(O)NHR2b. In some embodiments, R2 is —OR2a, —NHR2b, or —C(O)NHR2b, wherein each R2a and R2b is independently cyclohexane, 6-membered heterocyclyl, —(C1-C3 alkylene)N(C2H5)2, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), or —(C1-C3 alkylene)(5-6-membered heteroaryl), and wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3.
  • In some embodiments, R2 and R2b is independently cyclohexane or 6-membered heterocyclyl, wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3.
  • In some embodiments, R2 and R2b is independently —(C1-C3 alkylene)N(C2H5)2, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3- to 6-membered heterocyclyl), or —(C1-C3alkylene)(5- or 6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3.
  • In some embodiments, R2 is substituted C1-C6 alkyl. In some embodiments, R2 is substituted C1-C3 alkyl. In some embodiments, R2 is C1-C6 alkyl substituted by —OR2, —NHR2c, —SR2c, —S(O)2R2c, —S(O)2NHR2c, —NHS(O)2R2c—, —C(O) R2c, —NHC(O)R2c, —NHC(O)NR2c, —C(O)OR2c, —C(O)ONHR2c—, or —C(O)NHR2c, wherein R2 is 5- or 6-membered heteroaryl, and wherein the 5- or 6-membered heteroaryl is further substituted by C1-C6 alkyl optionally substituted by halogen, —OH or oxo. In some embodiments, R2 is C1-C6 alkyl substituted by —NHR2c, wherein R2c is 5- or 6-membered heteroaryl, and wherein the 5- or 6-membered heteroaryl is further substituted by C1-C6 alkyl optionally substituted by halogen, —OH or oxo. In some embodiments, R2c is pyridyl further substituted by C1-C6 alkyl optionally substituted by halogen, —OH or oxo. For example, in certain embodiments, R2 is pyridyl further substituted by —C(CH3)2OH.
  • In some embodiments R2 is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00006
    Figure US20220298142A1-20220922-C00007
    Figure US20220298142A1-20220922-C00008
    Figure US20220298142A1-20220922-C00009
    Figure US20220298142A1-20220922-C00010
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments R2 is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00011
    Figure US20220298142A1-20220922-C00012
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments R2 is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00013
    Figure US20220298142A1-20220922-C00014
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments R2 is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00015
    Figure US20220298142A1-20220922-C00016
  • wherein the wavy lines denote attachment points to the parent molecule.
  • It is understood that each R2 may be combined with each R1, A and/or B the same as if each and every combination of R2 with R1, A and/or B were specifically and individually listed. In some embodiments, R1 is hydrogen, and each R2 may be combined with each A and/or B the same as if each and every combination of R2 with A and/or B were specifically and individually listed.
  • In one aspect, provided is a compound of the formula (II):
  • Figure US20220298142A1-20220922-C00017
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein:
  • A is 9- or 10-membered bicyclic heteroaryl or a 9- or 10-membered bicyclic heterocyclyl, each of A is optionally substituted by R4;
  • B is a phenyl optionally substituted by R3, or a 5- or 6-membered heteroaryl optionally substituted by R4;
  • one of Q1 and Q2 is —O—, —NH—, or —C(O)NH— and the other is a bond;
  • L is a bond or C1-C4 alkylene;
  • D is —N(C2H5)2, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, or 5- or 6-membered heteroaryl, wherein each of which is optionally substituted by halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2, —NHCH3 or C1-C6 alkyl optionally substituted by halogen, —OH or oxo, when L is not a bond, or D is cyclohexane, or 6-membered heterocyclyl, wherein the cyclohexane and 6-membered heterocyclyl is optionally substituted by halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2, —NHCH3 or C1-C6 alkyl optionally substituted by halogen, —OH or oxo, when L is a bond;
  • each R3 is independently C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —OC(O)R5, —C(O)OR5, —C(O)NR6R7, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)OR6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, —NR5S(O)R6, —C(O)NR5S(O)R6, —NR5S(O)2R6, —C(O)NR5S(O)2R6, —S(O)NR6R7, —S(O)2NR6R7, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR5, —(C1-C3 alkylene)SR5, —(C1-C3 alkylene)NR6R7, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)NO2, —C═NH(OR5), —(C1-C3 alkylene)C(O)R5, —(C1-C3 alkylene)OC(O)R5, —(C1-C3 alkylene)C(O)OR5, —(C1-C3 alkylene)C(O)NR6R7, —(C1-C3 alkylene)OC(O)NR6R7, —(C1-C3 alkylene)NR5C(O)R6, —(C1-C3 alkylene)NR5C(O)OR6, —(C1-C3 alkylene)NR5C(O)NR6R7, —(C1-C3 alkylene)S(O)R5, —(C1-C3 alkylene)S(O)2R5, —(C1-C3 alkylene)NR5S(O)R6, —C(O)(C1-C3 alkylene)NR5S(O)R6, —(C1-C3 alkylene)NR5S(O)2R6, —(C1-C3 alkylene)C(O)NR5S(O)2R6, —(C1-C3 alkylene)S(O)NR6R7, —(C1-C3 alkylene)S(O)2NR6R7, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), wherein each R3 is independently optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • each R4 is independently oxo or R3;
  • R5 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)ORB, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9 or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
  • R8 and R9 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, OH, oxo or NH2;
      • or R8 and R9 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by halogen, OH, oxo or NH2.
  • In some embodiments Q1 is —O—. In some embodiments Q1 is —NH—. In some embodiments Q1 is —C(O)NH—. In some embodiments Q1 is a bond.
  • In some embodiments Q2 is —O—. In some embodiments Q2 is —NH—. In some embodiments Q2 is —C(O)NH—. In some embodiments Q2 is a bond.
  • In some embodiments, Q1 is —O—, —NH—, or —C(O)NH— and Q2 is a bond. In other embodiments, Q2 is —O—, —NH—, or —C(O)NH— and Q1 is a bond.
  • In some embodiments, L is a bond. In some embodiments, L is C1-C4 alkylene, for example, —CH2—, —CH2CH2—, and —CH2CH2CH2—.
  • In one variation, C1-C3 or C1-C4 alkylene as disclosed herein (for example, both in formula (I) and formula (II)) is a linear alkylene. In other variation, C1-C3 or C1-C4 alkylene is a branched alkylene, such as —CH(CH3)— and —C(CH3)2—. For example, in certain embodiments, —(C1-C3 alkylene)(5-6-membered heteroaryl) is —CH(CH3)-pyridyl.
  • In some embodiments, when L is not a bond, D is —N(C2H5)2, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, or 5- or 6-membered heteroaryl, wherein each of which is optionally substituted by halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2, —NHCH3 or C1-C6 alkyl optionally substituted by halogen, —OH or oxo. In some embodiments, when L is not a bond, D is optionally substituted —N(C2H5)2. In some embodiments, when L is not a bond, D is optionally substituted C3-C6 cycloalkyl. In some embodiments, when L is not a bond, D is optionally substituted 3- to 6-membered heterocyclyl. In some embodiments, when L is not a bond, D is optionally substituted 5- or 6-membered heteroaryl. In some embodiments, D is pyridyl further substituted by C1-C6 alkyl optionally substituted by halogen, —OH or oxo. For example, in certain embodiments, D is pyridyl further substituted by —C(CH3)2OH.
  • In some embodiments, when L is a bond, D is cyclohexane, or 6-membered heterocyclyl, wherein the cyclohexane and 6-membered heterocyclyl is optionally substituted by halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2, —NHCH3 or C1-C6 alkyl optionally substituted by halogen, —OH or oxo. In some embodiments, when L is a bond, D is optionally substituted cyclohexane. In some embodiments, when L is a bond, D is optionally substituted 6-membered heterocyclyl. In some embodiments, when L is a bond, Q1 is —C(O)NH—, Q2 is a bond, D is optionally substituted cyclohexane, or optionally substituted 6-membered heterocyclyl.
  • In some embodiments, Q1, Q2, L and D together are
  • Figure US20220298142A1-20220922-C00018
  • group, which is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00019
    Figure US20220298142A1-20220922-C00020
    Figure US20220298142A1-20220922-C00021
    Figure US20220298142A1-20220922-C00022
    Figure US20220298142A1-20220922-C00023
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, Q1, Q2, L and D together are
  • Figure US20220298142A1-20220922-C00024
  • group, which is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00025
    Figure US20220298142A1-20220922-C00026
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, Q1, Q2, L and D together are
  • Figure US20220298142A1-20220922-C00027
  • group, which is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00028
    Figure US20220298142A1-20220922-C00029
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, Q1, Q2, L and D together are
  • Figure US20220298142A1-20220922-C00030
  • group, which is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00031
    Figure US20220298142A1-20220922-C00032
  • herein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, Q1, Q2, L and D together are
  • Figure US20220298142A1-20220922-C00033
  • group, which is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00034
  • wherein the wavy lines denote attachment points to the parent molecule.
  • It is understood that each description of every variable of
  • Figure US20220298142A1-20220922-C00035
  • (Q1, Q2, L and D) may be combined with each A and/or B the same as if each and every combination of Q1, Q2, L or/and D of
  • Figure US20220298142A1-20220922-C00036
  • with A and/or B were specifically and individually listed.
  • In some embodiments, A is 4-hydroxyphenyl optionally further substituted by R3, 4-hydroxy-2-pyridyl optionally further substituted by R4, or a 9- or 10-membered bicyclic heteroaryl optionally substituted by R4.
  • In some embodiments, A is 9- or 10-membered bicyclic heteroaryl optionally substituted by R4. In some embodiments, A is a 9- or 10-membered bicyclic heterocyclyl optionally substituted by R4.
  • In some embodiments, A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R4. In some embodiments, A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R4, wherein one ring is saturated. In some embodiments, A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R4, wherein both rings are unsaturated. In some embodiments, A is selected from the group consisting of benzimidazolyl, benzoxazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, indazolyl, quinoxalinyl, quinazolinyl, cinnolinyl, naphthyridinyl and naphthyl. In some embodiments, A is selected from the group consisting of benzimidazolyl, benzoxazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, indazolyl, quinoxalinyl, quinazolinyl, cinnolinyl, naphthyridinyl and naphthyl, each of which is optionally substituted by R4. In yet further embodiments, A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R4, comprising a first and second ring, wherein the first ring has a greater number of ring atoms than the second ring. In certain embodiments, the point of attachment of A to the parent molecule is on the first ring having a greater number of ring atoms.
  • In other embodiments, the point of attachment of A to the parent molecule is on the second ring having a smaller number of ring atoms. In some embodiments, A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R4, wherein the two rings are selected from the group consisting of: a 5-membered ring and a 6-membered ring or two 6-membered rings.
  • In one aspect, when A is a 9- or 10-membered bicyclic heteroaryl optionally substituted by R4, A is an unsubstituted 9- or 10-membered bicyclic heteroaryl containing at least one annular nitrogen atom, a 9- or 10-membered bicyclic heteroaryl containing at least two annular nitrogen atoms and optionally substituted by R4 which R4 groups are connected to the parent structure via a carbon atom, or a 10-membered bicyclic heteroaryl optionally substituted by R4.
  • In some embodiments, A is a 9- or 10-membered bicyclic heteroaryl substituted with 0 to 3 R4 groups which may be the same or different, and which may be present on either one ring or both rings. In one such aspect, A is a 9- or 10-membered bicyclic heteroaryl substituted with 0 to 3 R3 groups which may be the same or different, and which may be present on either one ring or both rings. In one such aspect, A is a 9- or 10-membered bicyclic heteroaryl substituted with 1 R3 group. In another such aspect, A is a 9- or 10-membered bicyclic heteroaryl substituted with 2 R3 groups, which may be the same or different. In another such aspect, A is a 9- or 10-membered bicyclic heteroaryl substituted with 3 R3 groups, which may be the same or different.
  • In some embodiments, A is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00037
    Figure US20220298142A1-20220922-C00038
  • where R3, if present, is attached at any available position on the bicyclic ring system. In one aspect, at least one R3 is present and is attached at a position on the ring bearing the wavy line (on the ring that is the attachment point of the bicyclic ring to the parent molecule). In one aspect, at least one R3 is present and is attached at a position on the ring that does not bear the wavy line (on the ring that is fused to the ring which is the attachment point of the bicyclic ring to the parent molecule).
  • In some embodiments, A is a 9- or 10-membered bicyclic heteroaryl substituted with 0 to 3 R3 groups which may be the same or different, and which may be present on either one ring or both rings. In some embodiments, A is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00039
    Figure US20220298142A1-20220922-C00040
  • and where R3, if present, is attached at any available position on the bicyclic ring system. In one aspect, at least one R3 is present and is attached at a position on the ring bearing the wavy line (on the ring that is the attachment point of the bicyclic ring to the parent molecule). In one aspect, at least one R3 is present and is attached at a position on the ring that does not bear the wavy line (on the ring that is fused to the ring which is the attachment point of the bicyclic ring to the parent molecule).
  • In some embodiments, A is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00041
    Figure US20220298142A1-20220922-C00042
    Figure US20220298142A1-20220922-C00043
    Figure US20220298142A1-20220922-C00044
    Figure US20220298142A1-20220922-C00045
  • In some embodiments, A is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00046
    Figure US20220298142A1-20220922-C00047
  • In certain embodiments, A is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00048
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In certain embodiments, A is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00049
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In certain embodiments, A is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00050
  • wherein the wavy lines denote attachment points to the parent molecule. In some embodiments, A is
  • Figure US20220298142A1-20220922-C00051
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00052
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00053
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00054
  • It is understood that each description of A may be combined with each description of B, R1 and/or R2 the same as if each and every combination were specifically and individually listed.
  • In some embodiments, B is an unsubstituted phenyl. In some embodiments, B is a phenyl optionally substituted by R3. In some embodiments, B is a phenyl substituted by 1 to 3 R3 which R3 groups may be the same or different. In other embodiments, B is a 5- to 6-membered heteroaryl optionally substituted by R4. In other embodiments, B is a 5- to 6-membered heteroaryl substituted by 1 to 3 R4 which R4 may be the same or different. In some embodiments, the 5- to 6-membered heteroaryl of B is a 5-membered heteroaryl selected from the group consisting of furanyl, oxazolyl, thiophenyl, pyrazolyl, isoxazolyl, 1,3,4-oxadiazolyl, imidazolyl, thiazolyl, isothiazolyl, triazolyl, 1,3,4-thiadiazolyl and tetrazolyl, which 5-membered heteroaryl is optionally substituted by 1 to 3 R4 which R4 groups may be the same or different. In other embodiments, the 5- to 6-membered heteroaryl of B is a 6-membered heteroaryl selected from the group consisting of pyridyl, pyridazinyl and pyrimidinyl which 6-membered heteroaryl is optionally substituted to 1 to 3 R4 which R4 groups may be the same or different
  • In some embodiments, B is an unsubstituted phenyl. In some embodiments, B is a phenyl optionally substituted by R3. In some embodiments, B is a phenyl substituted by 1 to 3 R3 which R3 groups may be the same or different. In other embodiments, B is a 5- to 6-membered heteroaryl optionally substituted by R4. In other embodiments, B is a 5- to 6-membered heteroaryl substituted by 1 to 3 R4 which R4 may be the same or different. In some embodiments, the 5- to 6-membered heteroaryl of B is a 5-membered heteroaryl selected from the group consisting of furanyl, oxazolyl, thiophenyl, pyrazolyl, isoxazolyl, 1,3,4-oxadiazolyl, imidazolyl, thiazolyl, isothiazolyl, triazolyl, 1,3,4-thiadiazolyl and tetrazolyl, which 5-membered heteroaryl is optionally substituted by 1 to 3 R4 which R4 groups may be the same or different. In other embodiments, the 5- to 6-membered heteroaryl of B is a 6-membered heteroaryl selected from the group consisting of pyridyl and pyrimidinyl which 6-membered heteroaryl is optionally substituted to 1 to 3 R4 which R4 groups may be the same or different.
  • In some embodiments of B in which B is a phenyl substituted by R3, such as when B is a phenyl substituted by 1 to 3 R3 which may be the same or different, each R3 of B in one aspect is independently selected from the group consisting of halogen, —CN, —OR5, —NR6R7, —C(O)R5, C3-C6 cycloalkyl and C1-C6 alkyl optionally substituted by halogen. In other embodiments, each R3 of B is independently selected from the group consisting of halogen and C1-C6 alkyl optionally substituted by halogen (e.g., CF3).
  • In some embodiments, B is a phenyl substituted with 1 to 3 halo groups which may be the same or different. In some embodiments, B is phenyl, fluoro-phenyl, di-fluoro-phenyl, chloro-phenyl, di-chloro-phenyl or (fluoroxchloro)-phenyl. In some embodiments, B is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00055
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, B is a phenyl substituted with 1 to 3 halo groups which may be the same or different. In some embodiments, B is phenyl, fluoro-phenyl, di-fluoro-phenyl, chloro-phenyl, di-chloro-phenyl or (fluoroxchloro)-phenyl. In some embodiments, B is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00056
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, B is a 5-membered heteroaryl substituted with 0 to 3 R4 groups which may be the same or different. In some embodiments, B is a 5-membered heteroaryl substituted with 0 to 3 R3 groups which may be the same or different. In one such aspect, B is a 5-membered heteroaryl substituted with 1 R3 group. In another such aspect, B is a 5-membered heteroaryl substituted with 2 R3 groups, which may be the same or different. In another such aspect, B is a 5-membered heteroaryl substituted with 3 R3 groups, which may be the same or different. In some embodiments, B is a 5-membered heteroaryl selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00057
  • wherein the wavy lines denote attachment points to the parent molecule. It is understood that
  • Figure US20220298142A1-20220922-C00058
  • means that the B ring can be substituted with 0, 1, 2, or 3 R3 groups, as valence permits (e.g., when the maximum number of allowed substituents is 2, the B ring can be substituted with 0, 1, or 2 R3 groups).
  • In some embodiments, B is a 5-membered heteroaryl substituted with 0 to 3 R3 groups which may be the same or different. In some embodiments, B is a 5-membered heteroaryl selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00059
  • wherein the wavy lines denote attachment points to the parent molecule. It is understood that
  • Figure US20220298142A1-20220922-C00060
  • means that the B ring can be substituted with 0, 1, 2, or 3 R3 groups, as valence permits (e.g., when the maximum number of allowed substituents is 2, the B ring can be substituted with 0, 1, or 2 R3 groups).
  • In some embodiments, B is a 5-membered heteroaryl selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00061
    Figure US20220298142A1-20220922-C00062
    Figure US20220298142A1-20220922-C00063
    Figure US20220298142A1-20220922-C00064
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, B is a 5-membered heteroaryl selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00065
    Figure US20220298142A1-20220922-C00066
    Figure US20220298142A1-20220922-C00067
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 R4, which R4 may be the same or different. In some embodiments, B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 halo groups which may be the same or different. In some embodiments, B is a 6-membered heteroaryl selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00068
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 R4, which R4 may be the same or different. In some embodiments, B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 R3, which R3 may be the same or different. In some embodiments, B is a pyridyl or pyrimidyl optionally substituted by 1 to 3 halo groups which may be the same or different. In some embodiments, B is a 6-membered heteroaryl selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00069
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, B is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00070
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, B is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00071
  • wherein the wavy lines denote attachment points to the parent molecule.
  • In some embodiments, B is
  • Figure US20220298142A1-20220922-C00072
  • In some embodiments, B is
  • Figure US20220298142A1-20220922-C00073
  • In some embodiments, B is
  • Figure US20220298142A1-20220922-C00074
  • In some embodiments, B is
  • Figure US20220298142A1-20220922-C00075
  • It is understood that each description of B may be combined with each description of R1 and/or R2 the same as if each and every combination were specifically and individually listed. It is similarly understood that each description of B may be combined with each description of A (and further with each description of R1 and R2) the same as if each and every combination were specifically and individually listed. In one variation, B is as defined in any variation herein, R1 and R2 are as defined in any variation herein and A is 4-hydroxyphenyl optionally further substituted by R3 or 4-hydroxy-2-pyridyl optionally further substituted by R4. In another variation, B is as defined in any variation herein, R1 and R2 are as defined in any variation herein and A is 9- or 10-membered bicyclic heteroaryl (eg., quinolinyl or indazolyl) optionally substituted by R4.
  • In some embodiments, the compound of formula (I) is a compound of formula (IIIa):
  • Figure US20220298142A1-20220922-C00076
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X1 is independently O, S, NH, NR4a, CH2, CHR4b, CR4bR4b, N, CH or CR4b;
  • each X2 is independently NH, NR4a, CHR4b, CR4bR4b, CH, CR4b or N;
  • each
    Figure US20220298142A1-20220922-P00001
    is a single or double bond, provided that when
    Figure US20220298142A1-20220922-P00002
    is a double bond,
    Figure US20220298142A1-20220922-P00003
    is a single bond and when
    Figure US20220298142A1-20220922-P00003
    is a double bond,
    Figure US20220298142A1-20220922-P00002
    is a singe bond;
  • R4a is C1-C6 alkyl;
  • each R4b is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
      • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
  • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IIIa), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IIIb):
  • Figure US20220298142A1-20220922-C00077
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X1 is independently O, S, NH, NR4a, CH2, CHR4b, CR4bR4b, N, CH or CR4b;
  • each X2 is independently NH, NR4a, CH2, CHR4b, CR4bR4b, CH, CR4b or N;
  • each
    Figure US20220298142A1-20220922-P00001
    is a single or double bond;
  • R4a is C1-C6 alkyl;
  • each R4 is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
      • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
  • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IIIb), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IIIc):
  • Figure US20220298142A1-20220922-C00078
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X1 is independently O, S, NH, NR4a, CH2, CHR4b, CR4bR4b, N, CH or CR4b;
  • each X2 is independently CH, CR4b or N;
  • R4a is C1-C6 alkyl;
  • each R4b is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
      • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
  • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IIIc), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IIIc-1):
  • Figure US20220298142A1-20220922-C00079
  • or a salt thereof, wherein R1 and R2 are as defined for formula (I);
    each X1 and X2 are as defined for formula (IIIc);
  • X4 is C or N;
      • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IIIc-1), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IIIc-2):
  • Figure US20220298142A1-20220922-C00080
  • or a salt thereof, wherein R1, R2 and R3 are as defined for formula (I);
  • each X1 and X2 are as defined for formula (IIc);
      • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IIIc-2), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IIId):
  • Figure US20220298142A1-20220922-C00081
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X1 is independently O, S, NH, CH2, CHR4b, CR4bR4b, N, CH or CR4b;
  • each X2 is independently CH, CR4b or N;
  • each R4 is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
      • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl.
  • In some embodiments, provided is a compound of formula (IIId), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IIIe):
  • Figure US20220298142A1-20220922-C00082
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X1 is independently O, S, NH, NR4a, CH2, CHR4b, CR4bR4b, N, CH or CR4b;
  • each X2 is independently O, CH2, CHR4b, CR4bR4b, CH, CR4b or N;
  • each
    Figure US20220298142A1-20220922-P00001
    is a single or double bond;
  • R4a is C1-C6 alkyl;
  • each R4 is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
      • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
  • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IIIe), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IIIf):
  • Figure US20220298142A1-20220922-C00083
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X1 is independently O, S, NH, NR4a, CH2, CHR4b, CR4bR4b, N, CH or CR4b;
  • each X2 is independently C, CH, CR4b or N;
  • each
    Figure US20220298142A1-20220922-P00001
    is a single or double bond, provided that when
    Figure US20220298142A1-20220922-P00004
    is a double bond,
    Figure US20220298142A1-20220922-P00005
    is a single bond and when
    Figure US20220298142A1-20220922-P00005
    is a double bond,
    Figure US20220298142A1-20220922-P00004
    is a single bond;
  • R4a is C1-C6 alkyl;
  • each R4 is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
      • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
  • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IIIf), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IIIg):
  • Figure US20220298142A1-20220922-C00084
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X1 is independently O, S, NH, NR4a, N, CH or CR4b;
  • each X2 is independently C, CH, CR4b or N;
  • R4a is C1-C6 alkyl;
  • each R4 is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
      • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
  • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IIIg), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments of the compound of formula (III), R4b is selected from the group consisting of halogen, —OR5 and C1-C6 alkyl optionally substituted by halogen.
  • In some embodiments of the compound of formula (III), one of X1 is N, and the other one of X1 is NR4a, and each X2 is CH or CR4b. In other embodiments of the compound of formula (III), one of X1 is N, and the other one of X1 is O or S, and each X2 is CH or CR4b.
  • In some embodiments, the compound of formula (I) is a compound of formula (IVa):
  • Figure US20220298142A1-20220922-C00085
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X3 is independently NH, NR4, CH2, CHR4, CR4R4, CR4, CH, C═O, O or N;
  • each
    Figure US20220298142A1-20220922-P00001
    is a single or double bond;
  • each R4 is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, —(C1-C3 alkylene)(6-membered aryl) optionally substituted by halogen or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
      • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
        provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, the compound of formula (I) is a compound of formula (IVa):
  • Figure US20220298142A1-20220922-C00086
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X3 is independently NH, NR4, CH2, CHR4, CR4R4, CR4, CH or N;
  • each
    Figure US20220298142A1-20220922-P00001
    is a single or double bond;
  • each R4 is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
  • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
  • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (Va), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IVb):
  • Figure US20220298142A1-20220922-C00087
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X3 is independently NH, NR4, CH2, CHR4, CR4R4, CR4, CH or N;
  • each
    Figure US20220298142A1-20220922-P00001
    is a single or double bond;
  • each R4 is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
  • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
    provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IVb), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IVc):
  • Figure US20220298142A1-20220922-C00088
  • or a salt thereof, wherein R1, R2 and B are as defined for formula (I);
  • each X3 is independently CR4, CH or N;
  • each R4 is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
  • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
  • provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IVc), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments of formula (IV), R4 is selected from the group consisting of halogen, —OR5 and C1-C6 alkyl optionally substituted by halogen.
  • In some embodiments, one X3 is N, and the remaining X3 are each CR4. In some embodiments, two of the X3 are N, and the remaining X3 are each CR4.
  • In some embodiments, the compound of formula (I) is a compound of formula (IVc-1):
  • Figure US20220298142A1-20220922-C00089
  • or a salt thereof, wherein R1 and R2 are as defined for formula (I);
  • each X1 is independently O, S, NH, NR4, N, CH or CR4;
  • X4 is C or N;
  • each X3 is as defined for formula (IVc)
  • R4a is C1-C6 alkyl;
  • each Rb is independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen;
  • where each R5 is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl; and
  • R6 and R7 are each independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
  • or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl,
    provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IVc-1), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments, the compound of formula (I) is a compound of formula (IVc-2):
  • Figure US20220298142A1-20220922-C00090
  • or a salt thereof, wherein R1, R2 and R3 are as defined for formula (I);
    each X3 is as defined for formula (IVc);
    provided the compound is other than a compound selected from Table 1X or a salt thereof.
  • In some embodiments, provided is a compound of formula (IVc-2), or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
  • In some embodiments of a compound of any of the foregoing formula, A is
  • Figure US20220298142A1-20220922-C00091
  • wherein R401, R402, R403, R404, R405, and R406 are each independently R4. In some embodiments, R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen.
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00092
  • wherein R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen; and B is phenyl, optionally substituted with R3.
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00093
  • wherein R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen; and B is 5- to 6-membered heteroaryl, optionally substituted with R4.
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00094
  • wherein R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen; and B is 5-membered heteroaryl such as furanyl, oxazolyl, thiophenyl, pyrazolyl, isoxazolyl, 1,3,4-oxadiazolyl, imidazolyl, thiazolyl, isothiazolyl, triazolyl, 1,3,4-thiadiazolyl and tetrazolyl, each of which optionally substituted with R4.
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00095
  • wherein R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen; and B is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00096
    Figure US20220298142A1-20220922-C00097
    Figure US20220298142A1-20220922-C00098
    Figure US20220298142A1-20220922-C00099
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00100
  • wherein R401, R402, R403, R404, R405, and R406 are each independently R4. In some embodiments, R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen.
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00101
  • wherein R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen; and B is phenyl, optionally substituted with R3.
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00102
  • wherein R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen; and B is 5- to 6-membered heteroaryl, optionally substituted with R4.
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00103
  • wherein R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen; and B is 5-membered heteroaryl such as furanyl, oxazolyl, thiophenyl, pyrazolyl, isoxazolyl, 1,3,4-oxadiazolyl, imidazolyl, thiazolyl, isothiazolyl, triazolyl, 1,3,4-thiadiazolyl and tetrazolyl, each of which optionally substituted with R4.
  • In some embodiments, A is
  • Figure US20220298142A1-20220922-C00104
  • wherein R401, R402, R403, R404, R405, and R406 are each independently halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl, or C1-C6 alkyl optionally substituted by halogen; and B is selected from the group consisting of:
  • Figure US20220298142A1-20220922-C00105
    Figure US20220298142A1-20220922-C00106
    Figure US20220298142A1-20220922-C00107
    Figure US20220298142A1-20220922-C00108
  • Also provided are salts of compounds referred to herein, such as pharmaceutically acceptable salts. The invention also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms, and any tautomers or other forms of the compounds described.
  • A compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein. Compositions comprising a compound as detailed herein or a salt thereof are provided, such as compositions of substantially pure compounds. In some embodiments, a composition containing a compound as detailed herein or a salt thereof is in substantially pure form. Unless otherwise stated, “substantially pure” intends a composition that contains no more than 35% impurity, wherein the impurity denotes a compound other than the compound comprising the majority of the composition or a salt thereof. In some embodiments, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains no more than 25%, 20%, 15%, 10%, or 5% impurity. In some embodiments, a composition of substantially pure compound or a salt thereof is provided wherein the composition contains or no more than 3%, 2%, 1% or 0.5% impurity.
  • Representative compounds of formula (I) or (II) are listed in Table 1. In some embodiments, provided herein are compounds described in Table 1, including pharmaceutically acceptable salts thereof, and uses thereof. It is understood that individual enantiomers and diastereomers if not depicted and their corresponding structures can be readily determined therefrom. For example, compounds 75 and 76 are representative stereoisomers of compound 73.
  • TABLE 1
    Compd
    No. Structure
     1
    Figure US20220298142A1-20220922-C00109
     2
    Figure US20220298142A1-20220922-C00110
     3
    Figure US20220298142A1-20220922-C00111
     4
    Figure US20220298142A1-20220922-C00112
     5
    Figure US20220298142A1-20220922-C00113
     6
    Figure US20220298142A1-20220922-C00114
     7
    Figure US20220298142A1-20220922-C00115
     8
    Figure US20220298142A1-20220922-C00116
     9
    Figure US20220298142A1-20220922-C00117
     10
    Figure US20220298142A1-20220922-C00118
     11
    Figure US20220298142A1-20220922-C00119
     12
    Figure US20220298142A1-20220922-C00120
     13
    Figure US20220298142A1-20220922-C00121
     14
    Figure US20220298142A1-20220922-C00122
     15
    Figure US20220298142A1-20220922-C00123
     16
    Figure US20220298142A1-20220922-C00124
     17
    Figure US20220298142A1-20220922-C00125
     18
    Figure US20220298142A1-20220922-C00126
     19
    Figure US20220298142A1-20220922-C00127
     20
    Figure US20220298142A1-20220922-C00128
     21
    Figure US20220298142A1-20220922-C00129
     22
    Figure US20220298142A1-20220922-C00130
     23
    Figure US20220298142A1-20220922-C00131
     24
    Figure US20220298142A1-20220922-C00132
     25
    Figure US20220298142A1-20220922-C00133
     26
    Figure US20220298142A1-20220922-C00134
     27
    Figure US20220298142A1-20220922-C00135
     28
    Figure US20220298142A1-20220922-C00136
     29
    Figure US20220298142A1-20220922-C00137
     30
    Figure US20220298142A1-20220922-C00138
     31
    Figure US20220298142A1-20220922-C00139
     32
    Figure US20220298142A1-20220922-C00140
     33
    Figure US20220298142A1-20220922-C00141
     34
    Figure US20220298142A1-20220922-C00142
     35
    Figure US20220298142A1-20220922-C00143
     36
    Figure US20220298142A1-20220922-C00144
     37
    Figure US20220298142A1-20220922-C00145
     38
    Figure US20220298142A1-20220922-C00146
     39
    Figure US20220298142A1-20220922-C00147
     40
    Figure US20220298142A1-20220922-C00148
     41
    Figure US20220298142A1-20220922-C00149
     42
    Figure US20220298142A1-20220922-C00150
     43
    Figure US20220298142A1-20220922-C00151
     44
    Figure US20220298142A1-20220922-C00152
     45
    Figure US20220298142A1-20220922-C00153
     46
    Figure US20220298142A1-20220922-C00154
     47
    Figure US20220298142A1-20220922-C00155
     48
    Figure US20220298142A1-20220922-C00156
     49
    Figure US20220298142A1-20220922-C00157
     50
    Figure US20220298142A1-20220922-C00158
     51
    Figure US20220298142A1-20220922-C00159
     52
    Figure US20220298142A1-20220922-C00160
     53
    Figure US20220298142A1-20220922-C00161
     54
    Figure US20220298142A1-20220922-C00162
     55
    Figure US20220298142A1-20220922-C00163
     56
    Figure US20220298142A1-20220922-C00164
     57
    Figure US20220298142A1-20220922-C00165
     58
    Figure US20220298142A1-20220922-C00166
     59
    Figure US20220298142A1-20220922-C00167
     60
    Figure US20220298142A1-20220922-C00168
     61
    Figure US20220298142A1-20220922-C00169
     62
    Figure US20220298142A1-20220922-C00170
     63
    Figure US20220298142A1-20220922-C00171
     64
    Figure US20220298142A1-20220922-C00172
     65
    Figure US20220298142A1-20220922-C00173
     66
    Figure US20220298142A1-20220922-C00174
     67
    Figure US20220298142A1-20220922-C00175
     68
    Figure US20220298142A1-20220922-C00176
     69
    Figure US20220298142A1-20220922-C00177
     70
    Figure US20220298142A1-20220922-C00178
     71
    Figure US20220298142A1-20220922-C00179
     72
    Figure US20220298142A1-20220922-C00180
     73
    Figure US20220298142A1-20220922-C00181
     74
    Figure US20220298142A1-20220922-C00182
     75
    Figure US20220298142A1-20220922-C00183
     76
    Figure US20220298142A1-20220922-C00184
     77
    Figure US20220298142A1-20220922-C00185
     78
    Figure US20220298142A1-20220922-C00186
     79
    Figure US20220298142A1-20220922-C00187
     80
    Figure US20220298142A1-20220922-C00188
     81
    Figure US20220298142A1-20220922-C00189
     82
    Figure US20220298142A1-20220922-C00190
     83
    Figure US20220298142A1-20220922-C00191
     84
    Figure US20220298142A1-20220922-C00192
     85
    Figure US20220298142A1-20220922-C00193
     86
    Figure US20220298142A1-20220922-C00194
     87
    Figure US20220298142A1-20220922-C00195
     88
    Figure US20220298142A1-20220922-C00196
     89
    Figure US20220298142A1-20220922-C00197
     90
    Figure US20220298142A1-20220922-C00198
     91
    Figure US20220298142A1-20220922-C00199
     92
    Figure US20220298142A1-20220922-C00200
     93
    Figure US20220298142A1-20220922-C00201
     94
    Figure US20220298142A1-20220922-C00202
     95
    Figure US20220298142A1-20220922-C00203
     96
    Figure US20220298142A1-20220922-C00204
     97
    Figure US20220298142A1-20220922-C00205
     98
    Figure US20220298142A1-20220922-C00206
     99
    Figure US20220298142A1-20220922-C00207
    100
    Figure US20220298142A1-20220922-C00208
    101
    Figure US20220298142A1-20220922-C00209
    102
    Figure US20220298142A1-20220922-C00210
    103
    Figure US20220298142A1-20220922-C00211
    104
    Figure US20220298142A1-20220922-C00212
    105
    Figure US20220298142A1-20220922-C00213
    106
    Figure US20220298142A1-20220922-C00214
    107
    Figure US20220298142A1-20220922-C00215
    108
    Figure US20220298142A1-20220922-C00216
    109
    Figure US20220298142A1-20220922-C00217
    110
    Figure US20220298142A1-20220922-C00218
    111
    Figure US20220298142A1-20220922-C00219
    112
    Figure US20220298142A1-20220922-C00220
    113
    Figure US20220298142A1-20220922-C00221
    114
    Figure US20220298142A1-20220922-C00222
    115
    Figure US20220298142A1-20220922-C00223
    116
    Figure US20220298142A1-20220922-C00224
    117
    Figure US20220298142A1-20220922-C00225
    118
    Figure US20220298142A1-20220922-C00226
    119
    Figure US20220298142A1-20220922-C00227
    120
    Figure US20220298142A1-20220922-C00228
    121
    Figure US20220298142A1-20220922-C00229
    122
    Figure US20220298142A1-20220922-C00230
    123
    Figure US20220298142A1-20220922-C00231
    124
    Figure US20220298142A1-20220922-C00232
    125
    Figure US20220298142A1-20220922-C00233
    126
    Figure US20220298142A1-20220922-C00234
    127
    Figure US20220298142A1-20220922-C00235
    128
    Figure US20220298142A1-20220922-C00236
    129
    Figure US20220298142A1-20220922-C00237
    130
    Figure US20220298142A1-20220922-C00238
    131
    Figure US20220298142A1-20220922-C00239
    132
    Figure US20220298142A1-20220922-C00240
    133
    Figure US20220298142A1-20220922-C00241
    134
    Figure US20220298142A1-20220922-C00242
    135
    Figure US20220298142A1-20220922-C00243
    136
    Figure US20220298142A1-20220922-C00244
    137
    Figure US20220298142A1-20220922-C00245
    138
    Figure US20220298142A1-20220922-C00246
    139
    Figure US20220298142A1-20220922-C00247
    140
    Figure US20220298142A1-20220922-C00248
    141
    Figure US20220298142A1-20220922-C00249
    142
    Figure US20220298142A1-20220922-C00250
    143
    Figure US20220298142A1-20220922-C00251
    144
    Figure US20220298142A1-20220922-C00252
    145
    Figure US20220298142A1-20220922-C00253
    146
    Figure US20220298142A1-20220922-C00254
    147
    Figure US20220298142A1-20220922-C00255
    148
    Figure US20220298142A1-20220922-C00256
    149
    Figure US20220298142A1-20220922-C00257
    150
    Figure US20220298142A1-20220922-C00258
    151
    Figure US20220298142A1-20220922-C00259
    152
    Figure US20220298142A1-20220922-C00260
    153
    Figure US20220298142A1-20220922-C00261
    154
    Figure US20220298142A1-20220922-C00262
    155
    Figure US20220298142A1-20220922-C00263
    156
    Figure US20220298142A1-20220922-C00264
    157
    Figure US20220298142A1-20220922-C00265
    158
    Figure US20220298142A1-20220922-C00266
    159
    Figure US20220298142A1-20220922-C00267
    160
    Figure US20220298142A1-20220922-C00268
    161
    Figure US20220298142A1-20220922-C00269
    162
    Figure US20220298142A1-20220922-C00270
    163
    Figure US20220298142A1-20220922-C00271
    164
    Figure US20220298142A1-20220922-C00272
    165
    Figure US20220298142A1-20220922-C00273
    166
    Figure US20220298142A1-20220922-C00274
    167
    Figure US20220298142A1-20220922-C00275
    168
    Figure US20220298142A1-20220922-C00276
    169
    Figure US20220298142A1-20220922-C00277
    170
    Figure US20220298142A1-20220922-C00278
    171
    Figure US20220298142A1-20220922-C00279
    172
    Figure US20220298142A1-20220922-C00280
    173
    Figure US20220298142A1-20220922-C00281
    174
    Figure US20220298142A1-20220922-C00282
    175
    Figure US20220298142A1-20220922-C00283
    176
    Figure US20220298142A1-20220922-C00284
    177
    Figure US20220298142A1-20220922-C00285
    178
    Figure US20220298142A1-20220922-C00286
    179
    Figure US20220298142A1-20220922-C00287
    180
    Figure US20220298142A1-20220922-C00288
    181
    Figure US20220298142A1-20220922-C00289
    182
    Figure US20220298142A1-20220922-C00290
    183
    Figure US20220298142A1-20220922-C00291
    184
    Figure US20220298142A1-20220922-C00292
    185
    Figure US20220298142A1-20220922-C00293
    186
    Figure US20220298142A1-20220922-C00294
    187
    Figure US20220298142A1-20220922-C00295
    188
    Figure US20220298142A1-20220922-C00296
    189
    Figure US20220298142A1-20220922-C00297
    190
    Figure US20220298142A1-20220922-C00298
    191
    Figure US20220298142A1-20220922-C00299
    192
    Figure US20220298142A1-20220922-C00300
    193
    Figure US20220298142A1-20220922-C00301
    194
    Figure US20220298142A1-20220922-C00302
    195
    Figure US20220298142A1-20220922-C00303
    196
    Figure US20220298142A1-20220922-C00304
    197
    Figure US20220298142A1-20220922-C00305
    198
    Figure US20220298142A1-20220922-C00306
    199
    Figure US20220298142A1-20220922-C00307
    200
    Figure US20220298142A1-20220922-C00308
    201
    Figure US20220298142A1-20220922-C00309
    202
    Figure US20220298142A1-20220922-C00310
    203
    Figure US20220298142A1-20220922-C00311
    204
    Figure US20220298142A1-20220922-C00312
    205
    Figure US20220298142A1-20220922-C00313
    206
    Figure US20220298142A1-20220922-C00314
    207
    Figure US20220298142A1-20220922-C00315
    208
    Figure US20220298142A1-20220922-C00316
    209
    Figure US20220298142A1-20220922-C00317
    210
    Figure US20220298142A1-20220922-C00318
    211
    Figure US20220298142A1-20220922-C00319
    212
    Figure US20220298142A1-20220922-C00320
    213
    Figure US20220298142A1-20220922-C00321
    214
    Figure US20220298142A1-20220922-C00322
    215
    Figure US20220298142A1-20220922-C00323
    216
    Figure US20220298142A1-20220922-C00324
    217
    Figure US20220298142A1-20220922-C00325
    218
    Figure US20220298142A1-20220922-C00326
    219
    Figure US20220298142A1-20220922-C00327
    220
    Figure US20220298142A1-20220922-C00328
    221
    Figure US20220298142A1-20220922-C00329
    222
    Figure US20220298142A1-20220922-C00330
    223
    Figure US20220298142A1-20220922-C00331
    224
    Figure US20220298142A1-20220922-C00332
    225
    Figure US20220298142A1-20220922-C00333
    226
    Figure US20220298142A1-20220922-C00334
    227
    Figure US20220298142A1-20220922-C00335
    228
    Figure US20220298142A1-20220922-C00336
    229
    Figure US20220298142A1-20220922-C00337
    230
    Figure US20220298142A1-20220922-C00338
    231
    Figure US20220298142A1-20220922-C00339
    232
    Figure US20220298142A1-20220922-C00340
    233
    Figure US20220298142A1-20220922-C00341
    234
    Figure US20220298142A1-20220922-C00342
    235
    Figure US20220298142A1-20220922-C00343
    236
    Figure US20220298142A1-20220922-C00344
    237
    Figure US20220298142A1-20220922-C00345
    238
    Figure US20220298142A1-20220922-C00346
    239
    Figure US20220298142A1-20220922-C00347
    240
    Figure US20220298142A1-20220922-C00348
    241
    Figure US20220298142A1-20220922-C00349
    242
    Figure US20220298142A1-20220922-C00350
    243
    Figure US20220298142A1-20220922-C00351
    244
    Figure US20220298142A1-20220922-C00352
    245
    Figure US20220298142A1-20220922-C00353
    246
    Figure US20220298142A1-20220922-C00354
    247
    Figure US20220298142A1-20220922-C00355
    248
    Figure US20220298142A1-20220922-C00356
    249
    Figure US20220298142A1-20220922-C00357
    250
    Figure US20220298142A1-20220922-C00358
    251
    Figure US20220298142A1-20220922-C00359
    252
    Figure US20220298142A1-20220922-C00360
    253
    Figure US20220298142A1-20220922-C00361
    254
    Figure US20220298142A1-20220922-C00362
    255
    Figure US20220298142A1-20220922-C00363
    256
    Figure US20220298142A1-20220922-C00364
    257
    Figure US20220298142A1-20220922-C00365
    258
    Figure US20220298142A1-20220922-C00366
    259
    Figure US20220298142A1-20220922-C00367
    260
    Figure US20220298142A1-20220922-C00368
    261
    Figure US20220298142A1-20220922-C00369
    262
    Figure US20220298142A1-20220922-C00370
    263
    Figure US20220298142A1-20220922-C00371
    264
    Figure US20220298142A1-20220922-C00372
    265
    Figure US20220298142A1-20220922-C00373
    266
    Figure US20220298142A1-20220922-C00374
    267
    Figure US20220298142A1-20220922-C00375
    268
    Figure US20220298142A1-20220922-C00376
    269
    Figure US20220298142A1-20220922-C00377
    270
    Figure US20220298142A1-20220922-C00378
    271
    Figure US20220298142A1-20220922-C00379
    272
    Figure US20220298142A1-20220922-C00380
    273
    Figure US20220298142A1-20220922-C00381
    274
    Figure US20220298142A1-20220922-C00382
    275
    Figure US20220298142A1-20220922-C00383
    276
    Figure US20220298142A1-20220922-C00384
    277
    Figure US20220298142A1-20220922-C00385
    278
    Figure US20220298142A1-20220922-C00386
    279
    Figure US20220298142A1-20220922-C00387
    280
    Figure US20220298142A1-20220922-C00388
    281
    Figure US20220298142A1-20220922-C00389
    282
    Figure US20220298142A1-20220922-C00390
    283
    Figure US20220298142A1-20220922-C00391
    284
    Figure US20220298142A1-20220922-C00392
    285
    Figure US20220298142A1-20220922-C00393
    286
    Figure US20220298142A1-20220922-C00394
    287
    Figure US20220298142A1-20220922-C00395
    288
    Figure US20220298142A1-20220922-C00396
    289
    Figure US20220298142A1-20220922-C00397
    290
    Figure US20220298142A1-20220922-C00398
    291
    Figure US20220298142A1-20220922-C00399
    292
    Figure US20220298142A1-20220922-C00400
    293
    Figure US20220298142A1-20220922-C00401
    294
    Figure US20220298142A1-20220922-C00402
    295
    Figure US20220298142A1-20220922-C00403
    296
    Figure US20220298142A1-20220922-C00404
    297
    Figure US20220298142A1-20220922-C00405
    298
    Figure US20220298142A1-20220922-C00406
    299
    Figure US20220298142A1-20220922-C00407
    300
    Figure US20220298142A1-20220922-C00408
    301
    Figure US20220298142A1-20220922-C00409
    302
    Figure US20220298142A1-20220922-C00410
    303
    Figure US20220298142A1-20220922-C00411
    304
    Figure US20220298142A1-20220922-C00412
    305
    Figure US20220298142A1-20220922-C00413
    306
    Figure US20220298142A1-20220922-C00414
    307
    Figure US20220298142A1-20220922-C00415
    308
    Figure US20220298142A1-20220922-C00416
    309
    Figure US20220298142A1-20220922-C00417
    310
    Figure US20220298142A1-20220922-C00418
    311
    Figure US20220298142A1-20220922-C00419
    312
    Figure US20220298142A1-20220922-C00420
    313
    Figure US20220298142A1-20220922-C00421
    314
    Figure US20220298142A1-20220922-C00422
    315
    Figure US20220298142A1-20220922-C00423
    316
    Figure US20220298142A1-20220922-C00424
    317
    Figure US20220298142A1-20220922-C00425
    318
    Figure US20220298142A1-20220922-C00426
    319
    Figure US20220298142A1-20220922-C00427
    320
    Figure US20220298142A1-20220922-C00428
    321
    Figure US20220298142A1-20220922-C00429
    322
    Figure US20220298142A1-20220922-C00430
    323
    Figure US20220298142A1-20220922-C00431
    324
    Figure US20220298142A1-20220922-C00432
    325
    Figure US20220298142A1-20220922-C00433
    326
    Figure US20220298142A1-20220922-C00434
    327
    Figure US20220298142A1-20220922-C00435
    328
    Figure US20220298142A1-20220922-C00436
    329
    Figure US20220298142A1-20220922-C00437
    330
    Figure US20220298142A1-20220922-C00438
    331
    Figure US20220298142A1-20220922-C00439
    332
    Figure US20220298142A1-20220922-C00440
    333
    Figure US20220298142A1-20220922-C00441
    334
    Figure US20220298142A1-20220922-C00442
    335
    Figure US20220298142A1-20220922-C00443
    336
    Figure US20220298142A1-20220922-C00444
    337
    Figure US20220298142A1-20220922-C00445
    338
    Figure US20220298142A1-20220922-C00446
    339
    Figure US20220298142A1-20220922-C00447
    340
    Figure US20220298142A1-20220922-C00448
    341
    Figure US20220298142A1-20220922-C00449
    342
    Figure US20220298142A1-20220922-C00450
    343
    Figure US20220298142A1-20220922-C00451
    344
    Figure US20220298142A1-20220922-C00452
    345
    Figure US20220298142A1-20220922-C00453
    346
    Figure US20220298142A1-20220922-C00454
    347
    Figure US20220298142A1-20220922-C00455
    348
    Figure US20220298142A1-20220922-C00456
    349
    Figure US20220298142A1-20220922-C00457
    350
    Figure US20220298142A1-20220922-C00458
    351
    Figure US20220298142A1-20220922-C00459
    352
    Figure US20220298142A1-20220922-C00460
    353
    Figure US20220298142A1-20220922-C00461
    354
    Figure US20220298142A1-20220922-C00462
    355
    Figure US20220298142A1-20220922-C00463
    356
    Figure US20220298142A1-20220922-C00464
    357
    Figure US20220298142A1-20220922-C00465
    358
    Figure US20220298142A1-20220922-C00466
    359
    Figure US20220298142A1-20220922-C00467
    360
    Figure US20220298142A1-20220922-C00468
    361
    Figure US20220298142A1-20220922-C00469
    362
    Figure US20220298142A1-20220922-C00470
    363
    Figure US20220298142A1-20220922-C00471
    364
    Figure US20220298142A1-20220922-C00472
    365
    Figure US20220298142A1-20220922-C00473
    366
    Figure US20220298142A1-20220922-C00474
    367
    Figure US20220298142A1-20220922-C00475
    368
    Figure US20220298142A1-20220922-C00476
    369
    Figure US20220298142A1-20220922-C00477
    370
    Figure US20220298142A1-20220922-C00478
    371
    Figure US20220298142A1-20220922-C00479
    372
    Figure US20220298142A1-20220922-C00480
    373
    Figure US20220298142A1-20220922-C00481
    374
    Figure US20220298142A1-20220922-C00482
    375
    Figure US20220298142A1-20220922-C00483
    376
    Figure US20220298142A1-20220922-C00484
    377
    Figure US20220298142A1-20220922-C00485
    378
    Figure US20220298142A1-20220922-C00486
    379
    Figure US20220298142A1-20220922-C00487
    380
    Figure US20220298142A1-20220922-C00488
    381
    Figure US20220298142A1-20220922-C00489
    382
    Figure US20220298142A1-20220922-C00490
    383
    Figure US20220298142A1-20220922-C00491
    384
    Figure US20220298142A1-20220922-C00492
    385
    Figure US20220298142A1-20220922-C00493
    386
    Figure US20220298142A1-20220922-C00494
    387
    Figure US20220298142A1-20220922-C00495
    388
    Figure US20220298142A1-20220922-C00496
    389
    Figure US20220298142A1-20220922-C00497
    390
    Figure US20220298142A1-20220922-C00498
    391
    Figure US20220298142A1-20220922-C00499
    392
    Figure US20220298142A1-20220922-C00500
    393
    Figure US20220298142A1-20220922-C00501
    394
    Figure US20220298142A1-20220922-C00502
    395
    Figure US20220298142A1-20220922-C00503
    396
    Figure US20220298142A1-20220922-C00504
    397
    Figure US20220298142A1-20220922-C00505
    398
    Figure US20220298142A1-20220922-C00506
    399
    Figure US20220298142A1-20220922-C00507
    400
    Figure US20220298142A1-20220922-C00508
    401
    Figure US20220298142A1-20220922-C00509
    402
    Figure US20220298142A1-20220922-C00510
    403
    Figure US20220298142A1-20220922-C00511
    404
    Figure US20220298142A1-20220922-C00512
    405
    Figure US20220298142A1-20220922-C00513
    406
    Figure US20220298142A1-20220922-C00514
    407
    Figure US20220298142A1-20220922-C00515
    408
    Figure US20220298142A1-20220922-C00516
    409
    Figure US20220298142A1-20220922-C00517
    410
    Figure US20220298142A1-20220922-C00518
    411
    Figure US20220298142A1-20220922-C00519
    412
    Figure US20220298142A1-20220922-C00520
    413
    Figure US20220298142A1-20220922-C00521
    414
    Figure US20220298142A1-20220922-C00522
    415
    Figure US20220298142A1-20220922-C00523
    416
    Figure US20220298142A1-20220922-C00524
    417
    Figure US20220298142A1-20220922-C00525
    418
    Figure US20220298142A1-20220922-C00526
    419
    Figure US20220298142A1-20220922-C00527
    420
    Figure US20220298142A1-20220922-C00528
    421
    Figure US20220298142A1-20220922-C00529
    422
    Figure US20220298142A1-20220922-C00530
    423
    Figure US20220298142A1-20220922-C00531
    424
    Figure US20220298142A1-20220922-C00532
    425
    Figure US20220298142A1-20220922-C00533
    426
    Figure US20220298142A1-20220922-C00534
    427
    Figure US20220298142A1-20220922-C00535
    428
    Figure US20220298142A1-20220922-C00536
    429
    Figure US20220298142A1-20220922-C00537
    430
    Figure US20220298142A1-20220922-C00538
    431
    Figure US20220298142A1-20220922-C00539
    432
    Figure US20220298142A1-20220922-C00540
    433
    Figure US20220298142A1-20220922-C00541
    434
    Figure US20220298142A1-20220922-C00542
    435
    Figure US20220298142A1-20220922-C00543
    436
    Figure US20220298142A1-20220922-C00544
    437
    Figure US20220298142A1-20220922-C00545
    438
    Figure US20220298142A1-20220922-C00546
    439
    Figure US20220298142A1-20220922-C00547
    440
    Figure US20220298142A1-20220922-C00548
    441
    Figure US20220298142A1-20220922-C00549
    442
    Figure US20220298142A1-20220922-C00550
    443
    Figure US20220298142A1-20220922-C00551
    444
    Figure US20220298142A1-20220922-C00552
    445
    Figure US20220298142A1-20220922-C00553
    446
    Figure US20220298142A1-20220922-C00554
    447
    Figure US20220298142A1-20220922-C00555
    448
    Figure US20220298142A1-20220922-C00556
    449
    Figure US20220298142A1-20220922-C00557
    450
    Figure US20220298142A1-20220922-C00558
    451
    Figure US20220298142A1-20220922-C00559
    452
    Figure US20220298142A1-20220922-C00560
    453
    Figure US20220298142A1-20220922-C00561
    454
    Figure US20220298142A1-20220922-C00562
    455
    Figure US20220298142A1-20220922-C00563
    456
    Figure US20220298142A1-20220922-C00564
    457
    Figure US20220298142A1-20220922-C00565
    458
    Figure US20220298142A1-20220922-C00566
    459
    Figure US20220298142A1-20220922-C00567
    460
    Figure US20220298142A1-20220922-C00568
    461
    Figure US20220298142A1-20220922-C00569
    462
    Figure US20220298142A1-20220922-C00570
  • In some embodiments, the compound of formula (I) is of formula (I-A):
  • Figure US20220298142A1-20220922-C00571
  • or a salt thereof, wherein R2 and B are as defined for formula (I) or any embodiment or aspect or other variation thereof, wherein
      • (i) when B is
  • Figure US20220298142A1-20220922-C00572
  • R2 is not
  • Figure US20220298142A1-20220922-C00573
      • (ii) when B is
  • Figure US20220298142A1-20220922-C00574
  • R2 is not
  • Figure US20220298142A1-20220922-C00575
  • In some embodiments, the compound of formula (I) is of formula (I-B):
  • Figure US20220298142A1-20220922-C00576
  • or a salt thereof, wherein R2 and B are as defined for formula (I) or any embodiment or aspect or other variation thereof, wherein
  • (i) when B is
  • Figure US20220298142A1-20220922-C00577
  • R2 is not
  • Figure US20220298142A1-20220922-C00578
  • In some embodiments of the compound of formula (I),
  • (i) when the compound is of formula (I-A) and B is
  • Figure US20220298142A1-20220922-C00579
  • R2 is no
  • Figure US20220298142A1-20220922-C00580
  • (ii) when the compound is of formula (I-A), and B is
  • Figure US20220298142A1-20220922-C00581
  • R2 is not
  • Figure US20220298142A1-20220922-C00582
  • (iii) when the compound is of formula (I-B), and B is
  • Figure US20220298142A1-20220922-C00583
  • R2 is not
  • Figure US20220298142A1-20220922-C00584
  • In some embodiments of the compound of any formulae disclosed herein (e.g., formulae (IIIa), (IIIb), (IIIc), (IIIc-1), (IIIc-2), (IIId), (IIIe), (IIIf), (IIIg), (IVa), (IVb), (IVc), (IVc-1), (IVc-2)),
  • (i) when the compound is of formula (I-A) and B is
  • Figure US20220298142A1-20220922-C00585
  • R2 is not
  • Figure US20220298142A1-20220922-C00586
  • (ii) when the compound is of formula (I-A), and B is
  • Figure US20220298142A1-20220922-C00587
  • R2 is not
  • Figure US20220298142A1-20220922-C00588
  • (iii) when the compound is of formula (I-B), and B is
  • Figure US20220298142A1-20220922-C00589
  • R2 is not
  • Figure US20220298142A1-20220922-C00590
  • In some embodiments of formula (I) as disclosed herein, the compound is other than the compounds in Table 1X.
  • In some embodiments, the compound of formula (II) is of formula (II-A):
  • Figure US20220298142A1-20220922-C00591
  • or a salt thereof, wherein Q1, Q2, L, D and B are as defined for formula (I) or any embodiment or aspect or other variation thereof, wherein
  • (i) when B is
  • Figure US20220298142A1-20220922-C00592
  • is not
  • Figure US20220298142A1-20220922-C00593
  • (ii) when B is
  • Figure US20220298142A1-20220922-C00594
  • is not
  • Figure US20220298142A1-20220922-C00595
  • In some embodiments, the compound of formula (II) is of formula (II-B):
  • Figure US20220298142A1-20220922-C00596
  • or a salt thereof, wherein Q1, Q2, L, D and B are as defined for formula (I) or any embodiment or aspect or other variation thereof, wherein
  • (i) when B is
  • Figure US20220298142A1-20220922-C00597
  • is not
  • Figure US20220298142A1-20220922-C00598
  • In some embodiments of the compound of formula (II),
  • (i) when the compound is of formula (II-A) and B is
  • Figure US20220298142A1-20220922-C00599
  • is not
  • Figure US20220298142A1-20220922-C00600
  • (ii) when the compound is of formula (II-A), and B is
  • Figure US20220298142A1-20220922-C00601
  • is not
  • Figure US20220298142A1-20220922-C00602
  • and
  • (iii) when the compound is of formula (II-B), and B is
  • Figure US20220298142A1-20220922-C00603
  • is not
  • Figure US20220298142A1-20220922-C00604
  • In some embodiments of formula (II) as disclosed herein, the compound is other than the compounds in Table 1X.
  • In some embodiments of a compound of formula (I) or (II), the compound is other than the compounds in Table 1X, a tautomer or isomer thereof, and a salt of any of the foregoing.
  • TABLE 1X
    Compound Name
    3-amino-5-(5-methylfuran-2-yl)-N-((6-methylpyridin-2-yl)methyl)-6-(quinolin-6-yl)pyrazine-
    2-carboxamide
    3-amino-N-((6-methylpyridin-2-yl)methyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide
    5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)-3-(2-(4-methylpiperazin-1-
    yl)ethoxy)pyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-3-(2-(diethylamino)ethoxy)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-
    amine
    6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N2-(morpholinomethyl)pyrazine-
    2,3-diamine
    6-(5-amino-3-(1-methyl-1H-pyrazol-3-yl)-6-(((1-methyl-1H-pyrrol-2-
    yl)methyl)amino)pyrazin-2-yl)isoquinolin-1(2H)-one
    3-amino-6-(8-chloroquinolin-6-yl)-5-(3-methyl-1H-pyrazol-1-yl)-N-((5-methylpyridin-2-
    yl)methyl)pyrazine-2-carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(3-methyl-1H-pyrazol-1-yl)-N-
    (morpholinomethyl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-((2-oxopiperidin-4-
    yl)methyl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(2-(3-hydroxypyrrolidin-1-yl)ethyl)-5-(2-oxopyridin-
    1(2H)-yl)pyrazine-2-carboxamide
    6-(7-chloro-1H-benzo[d]imidazol-5-yl)-5-(5-methylisothiazol-3-yl)-N2-((tetrahydro-2H-
    pyran-4-yl)methyl)pyrazine-2,3-diamine
    6-(5-amino-3-(1-methyl-1H-pyrazol-3-yl)-6-(((1-methyl-1H-pyrrol-2-
    yl)methyl)amino)pyrazin-2-yl)-8-methylisoquinolin-1(2H)-one
    3-amino-6-(8-chloroquinolin-6-yl)-5-(3-methyl-1H-pyrrol-1-yl)-N-((5-methylpyridin-2-
    yl)methyl)pyrazine-2-carboxamide
    3-amino-6-(7-chloro-1H-benzo[d]imidazol-5-yl)-5-(3-methyl-1H-pyrazol-1-yl)-N-
    (morpholinomethyl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrrol-3-yl)-N-((2-oxopiperazin-1-
    yl)methyl)pyrazine-2-carboxamide
    3-amino-6-(8-chloro-1,2,3,4-tetrahydroquinolin-6-yl)-N-((3-hydroxypyrrolidin-1-yl)methyl)-
    5-(2-oxopyridin-1(2H)-yl)pyrazine-2-carboxamide
    6-(8-chloroquinolin-6-yl)-N2-(2-(diethylamino)ethyl)-5-(3-methyl-1H-pyrazol-1-yl)pyrazine-
    2,3-diamine
    6-(8-chloroquinolin-6-yl)-N2-(2-(diethylamino)ethyl)-5-(2-methylthiazol-5-yl)pyrazine-2,3-
    diamine
    6-(7-chloro-1H-indazol-5-yl)-N2-(2-(diethylamino)ethyl)-5-(1-methyl-1H-pyrazol-3-
    yl)pyrazine-2,3-diamine
    6-(4-chlorobenzo[d]thiazol-6-yl)-N2-(2-(diethylamino)ethyl)-5-(1-methyl-1H-pyrazol-3-
    yl)pyrazine-2,3-diamine
    6-(4-chlorobenzo[d]oxazol-6-yl)-N2-(2-(diethylamino)ethyl)-5-(1-methyl-1H-pyrazol-3-
    yl)pyrazine-2,3-diamine
    5-(8-chloroquinolin-6-yl)-3-(2-(diethylamino)ethoxy)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-
    amine
    5-(8-chloroquinolin-6-yl)-3-(cyclopropylmethoxy)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-
    amine
    5-(8-chloroquinolin-6-yl)-3-(cyclobutylmethoxy)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-
    amine
    5-(8-chloroquinolin-6-yl)-3-(2-cyclopentylethoxy)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-
    amine
    5-(8-chloroquinolin-6-yl)-3-(cyclohexylmethoxy)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-
    amine
    5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)-3-((1-methylpyrrolidin-3-
    yl)methoxy)pyrazin-2-amine
    3-(2-(azetidin-1-yl)ethoxy)-5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-
    amine
    5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)-3-(2-(pyrrolidin-1-yl)ethoxy)pyrazin-
    2-amine
    5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)-3-((1-methylpiperidin-4-
    yl)methoxy)pyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)-3-(2-(3-(trifluoromethyl)-1H-pyrrol-
    1-yl)ethoxy)pyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)-3-(pyridin-3-ylmethoxy)pyrazin-2-
    amine
    5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)-3-((6-(methylamino)pyridin-3-
    yl)methoxy)pyrazin-2-amine
    6-(8-chloroquinolin-6-yl)-N2-(2-(diethylamino)ethyl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-
    2,3-diamine
    6-(8-chloroquinolin-6-yl)-N2-(cyclopropylmethyl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2,3-
    diamine
    6-(8-chloroquinolin-6-yl)-N2-(cyclobutylmethyl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2,3-
    diamine
    6-(8-chloroquinolin-6-yl)-N2-(2-cyclopentylethyl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2,3-
    diamine
    6-(8-chloroquinolin-6-yl)-N2-(cyclohexylmethyl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2,3-
    diamine
    1-(2-((3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-
    yl)amino)ethyl)piperidin-4-ol
    N2-(2-(azetidin-1-yl)ethyl)-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-
    2,3-diamine
    6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N2-(2-(pyrrolidin-1-
    yl)ethyl)pyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N2-((4-methylpiperazin-1-
    yl)methyl)pyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N2-(2-(3-(trifluoromethyl)-1H-
    pyrrol-1-yl)ethyl)pyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N2-(pyridin-3-ylmethyl)pyrazine-2,3-
    diamine
    6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N2-((6-(methylamino)pyridin-3-
    yl)methyl)pyrazine-2,3-diamine
    3-amino-6-(8-chloroquinolin-6-yl)-N-(2-(diethylamino)ethyl)-5-(1-methyl-1H-pyrazol-3-
    yl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(cyclopropylmethyl)-5-(1-methyl-1H-pyrazol-3-
    yl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-((3-hydroxycyclobutyl)methyl)-5-(1-methyl-1H-pyrazol-
    3-yl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(cyclopentylmethyl)-5-(1-methyl-1H-pyrazol-3-
    yl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(cyclohexylmethyl)-5-(1-methyl-1H-pyrazol-3-
    yl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-((5-oxopyrrolidin-3-
    yl)methyl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-((3-hydroxyazetidin-1-yl)methyl)-5-(1-methyl-1H-
    pyrazol-3-yl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-(2-(2-oxopyrrolidin-1-
    yl)ethyl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-((1-methylpiperidin-4-
    yl)methyl)pyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-((1-methyl-1H-pyrrol-3-
    yl)methyl)pyrazine-2-carboxamide
    ethyl 6-((3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-
    carboxamido)methyl)nicotinate
    5-(8-chloroquinolin-6-yl)-3-((diethylamino)methoxy)-6-phenylpyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-3-(cyclopropylmethoxy)-6-phenylpyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-3-(cyclobutylmethoxy)-6-phenylpyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-3-(2-cyclopentylethoxy)-6-phenylpyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-3-(cyclohexylmethoxy)-6-phenylpyrazin-2-amine
    4-(((3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazin-2-yl)oxy)methyl)-1-methylpyrrolidin-
    2-one
    5-(8-chloroquinolin-6-yl)-3-((1-methylazetidin-3-yl)methoxy)-6-phenylpyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-6-phenyl-3-(2-(pyrrolidin-1-yl)ethoxy)pyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-3-((1-methylpiperidin-4-yl)methoxy)-6-phenylpyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-6-phenyl-3-(2-(3-(trifluoromethyl)-1H-pyrrol-1-yl)ethoxy)pyrazin-
    2-amine
    5-(8-chloroquinolin-6-yl)-6-phenyl-3-(pyridin-3-ylmethoxy)pyrazin-2-amine
    5-(8-chloroquinolin-6-yl)-3-((6-(methylamino)pyridin-3-yl)methoxy)-6-phenylpyrazin-2-
    amine
    6-(8-chloroquinolin-6-yl)-N2-(2-(diethylamino)ethyl)-5-phenylpyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-N2-(cyclopropylmethyl)-5-phenylpyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-N2-(cyclobutylmethyl)-5-phenylpyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-N2-(2-cyclopentylethyl)-5-phenylpyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-N2-(cyclohexylmethyl)-5-phenylpyrazine-2,3-diamine
    4-(((3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazin-2-yl)amino)methyl)pyrrolidin-2-one
    N2-(2-(azetidin-1-yl)ethyl)-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-5-phenyl-N2-(2-(pyrrolidin-1-yl)ethyl)pyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-N2-((1-methylpiperidin-4-yl)methyl)-5-phenylpyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-5-phenyl-N2-(2-(3-(trifluoromethyl)-1H-pyrrol-1-yl)ethyl)pyrazine-
    2,3-diamine
    6-(8-chloroquinolin-6-yl)-5-phenyl-N2-(pyridin-3-ylmethyl)pyrazine-2,3-diamine
    6-(8-chloroquinolin-6-yl)-N2-((6-(methylamino)pyridin-3-yl)methyl)-5-phenylpyrazine-2,3-
    diamine
    3-amino-6-(8-chloroquinolin-6-yl)-N-(2-(diethylamino)ethyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(cyclopropylmethyl)-5-phenylpyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-((3-hydroxycyclobutyl)methyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(cyclopentylmethyl)-5-phenylpyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(cyclohexylmethyl)-5-phenylpyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(morpholinomethyl)-5-phenylpyrazine-2-carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(2-(3-hydroxyazetidin-1-yl)ethyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-(2-(2-oxopyrrolidin-1-yl)ethyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-((1-methylpiperidin-4-yl)methyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(8-chloroquinolin-6-yl)-N-((1-methyl-1H-pyrrol-3-yl)methyl)-5-phenylpyrazine-2-
    carboxamide
    ethyl 6-((3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2-
    carboxamido)methyl)nicotinate
    5-(7-chloro-1H-indazol-5-yl)-3-(2-(diethylamino)ethoxy)-6-phenylpyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-3-(cyclopropylmethoxy)-6-phenylpyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-3-(cyclobutylmethoxy)-6-phenylpyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-3-(2-cyclopentylethoxy)-6-phenylpyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-3-(cyclohexylmethoxy)-6-phenylpyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-3-((3-methylimidazolidin-1-yl)methoxy)-6-phenylpyrazin-2-
    amine
    5-(7-chloro-1H-indazol-5-yl)-3-((1-methylazetidin-3-yl)methoxy)-6-phenylpyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-6-phenyl-3-(2-(pyrrolidin-1-yl)ethoxy)pyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-3-((1-methylpiperidin-4-yl)methoxy)-6-phenylpyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-6-phenyl-3-(2-(3-(trifluoromethyl)-1H-pyrrol-1-
    yl)ethoxy)pyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-6-phenyl-3-(pyridin-3-ylmethoxy)pyrazin-2-amine
    5-(7-chloro-1H-indazol-5-yl)-3-((6-(methylamino)pyridin-3-yl)methoxy)-6-phenylpyrazin-2-
    amine
    6-(7-chloro-1H-indazol-5-yl)-N2-(2-(diethylamino)ethyl)-5-phenylpyrazine-2,3-diamine
    6-(7-chloro-1H-indazol-5-yl)-N2-(cyclopropylmethyl)-5-phenylpyrazine-2,3-diamine
    6-(7-chloro-1H-indazol-5-yl)-N2-(cyclobutylmethyl)-5-phenylpyrazine-2,3-diamine
    6-(7-chloro-1H-indazol-5-yl)-N2-(2-cyclopentylethyl)-5-phenylpyrazine-2,3-diamine
    6-(7-chloro-1H-indazol-5-yl)-N2-(cyclohexylmethyl)-5-phenylpyrazine-2,3-diamine
    6-(7-chloro-1H-indazol-5-yl)-N2-((3-methylimidazolidin-1-yl)methyl)-5-phenylpyrazine-2,3-
    diamine
    N2-(2-(azetidin-1-yl)ethyl)-6-(7-chloro-1H-indazol-5-yl)-5-phenylpyrazine-2,3-diamine
    6-(7-chloro-1H-indazol-5-yl)-5-phenyl-N2-(2-(pyrrolidin-1-yl)ethyl)pyrazine-2,3-diamine
    6-(7-chloro-1H-indazol-5-yl)-N2-((1-methylpiperidin-4-yl)methyl)-5-phenylpyrazine-2,3-
    diamine
    6-(7-chloro-1H-indazol-5-yl)-5-phenyl-N2-(2-(3-(trifluoromethyl)-1H-pyrrol-1-
    yl)ethyl)pyrazine-2,3-diamine
    6-(7-chloro-1H-indazol-5-yl)-5-phenyl-N2-(pyridin-3-ylmethyl)pyrazine-2,3-diamine
    6-(7-chloro-1H-indazol-5-yl)-N2-((6-(methylamino)pyridin-3-yl)methyl)-5-phenylpyrazine-
    2,3-diamine
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-(1-methyl-2-oxopiperidin-4-yl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-(2-(diethylamino)ethyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-(cyclopropylmethyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-((3-hydroxycyclobutyl)methyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-(cyclopentylmethyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-(cyclohexylmethyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-5-phenyl-N-((tetrahydro-2H-pyran-4-
    yl)methyl)pyrazine-2-carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-(2-(3-hydroxyazetidin-1-yl)ethyl)-5-phenylpyrazine-
    2-carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-((2-oxopyrrolidin-3-yl)methyl)-5-phenylpyrazine-2-
    carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-((1-methylpiperidin-4-yl)methyl)-5-phenylpyrazine-
    2-carboxamide
    3-amino-6-(7-chloro-1H-indazol-5-yl)-N-((1-methyl-1H-pyrazol-3-yl)methyl)-5-
    phenylpyrazine-2-carboxamide
    ethyl 6-((3-amino-6-(7-chloro-1H-indazol-5-yl)-5-phenylpyrazine-2-
    carboxamido)methyl)nicotinate
  • In another aspect, also provided herein is a compound of the formula (III):
  • Figure US20220298142A1-20220922-C00605
  • or a tautomer or isomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein:
  • A is 9- or 10-membered bicyclic heteroaryl or a 9- or 10-membered bicyclic heterocyclyl, each of A is optionally substituted by Ra;
  • B is a phenyl substituted with 1 to 3 Ra groups which may be the same or different, wherein at least one Ra group is —CN, and
      • Ra is halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2, —NHCH3 or C1-C6 alkyl optionally substituted by halogen, —OH or oxo.
  • In some embodiments of the compounds of formula (III), A is selected from the group consisting of
  • Figure US20220298142A1-20220922-C00606
  • In some embodiments of the compounds of formula (III), B is selected from the group consisting of
  • Figure US20220298142A1-20220922-C00607
  • It is understood that each A may be combined with each B the same as if each and every combination of A and/or B were specifically and individually listed. For example, in some embodiments, B is
  • Figure US20220298142A1-20220922-C00608
  • in formula (III), wherein A is as defined herein.
  • Representative compounds of formula (III) are listed in Table 2. In some embodiments, provided herein are compounds described in Table 2, including pharmaceutically acceptable salts thereof, and uses thereof. It is understood that individual enantiomers and diastereomers if not depicted and their corresponding structures can be readily determined therefrom.
  • TABLE 2
    Compd
    No. Structure
    2-1 
    Figure US20220298142A1-20220922-C00609
    2-2 
    Figure US20220298142A1-20220922-C00610
    2-3 
    Figure US20220298142A1-20220922-C00611
    2-4 
    Figure US20220298142A1-20220922-C00612
    2-5 
    Figure US20220298142A1-20220922-C00613
    2-6 
    Figure US20220298142A1-20220922-C00614
    2-7 
    Figure US20220298142A1-20220922-C00615
    2-8 
    Figure US20220298142A1-20220922-C00616
    2-9 
    Figure US20220298142A1-20220922-C00617
    2-10
    Figure US20220298142A1-20220922-C00618
    2-11
    Figure US20220298142A1-20220922-C00619
    2-12
    Figure US20220298142A1-20220922-C00620
    2-13
    Figure US20220298142A1-20220922-C00621
  • The embodiments and variations described herein are suitable for compounds of any formulae detailed herein, where applicable.
  • Representative examples of compounds detailed herein, including intermediates and final compounds according to the present disclosure are depicted herein. It is understood that in one aspect, any of the compounds may be used in the methods detailed herein, including, where applicable, intermediate compounds that may be isolated and administered to an individual.
  • The compounds depicted herein may be present as salts even if salts are not depicted and it is understood that the present disclosure embraces all salts and solvates of the compounds depicted here, as well as the non-salt and non-solvate form of the compound, as is well understood by the skilled artisan. In some embodiments, the salts of the compounds provided herein are pharmaceutically acceptable salts. Where one or more tertiary amine moiety is present in the compound, the N-oxides are also provided and described.
  • Where tautomeric forms may be present for any of the compounds described herein, each and every tautomeric form is intended even though only one or some of the tautomeric forms may be explicitly depicted. The tautomeric forms specifically depicted may or may not be the predominant forms in solution or when used according to the methods described herein.
  • The present disclosure also includes any or all of the stereochemical forms, including any enantiomeric or diastereomeric forms of the compounds described. The structure or name is intended to embrace all possible stereoisomers of a compound depicted, and each unique stereoisomer has a compound number bearing a suffix “a”, “b”, etc. All forms of the compounds are also embraced by the invention, such as crystalline or non-crystalline forms of the compounds. Compositions comprising a compound of the invention are also intended, such as a composition of substantially pure compound, including a specific stereochemical form thereof, or a composition comprising mixtures of compounds of the invention in any ratio, including two or more stereochemical forms, such as in a racemic or non-racemic mixture.
  • The invention also intends isotopically-labeled and/or isotopically-enriched forms of compounds described herein. The compounds herein may contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. In some embodiments, the compound is isotopically-labeled, such as an isotopically-labeled compound of formulae (I), (II) or (III), or variations thereof described herein, where a fraction of one or more atoms are replaced by an isotope of the same element. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, chlorine, such as 2H, 3H, 11C, 13C, 14C 13N, 15O, 17O, 32P, 35S, 18F, 36Cl. Certain isotope labeled compounds (e.g. 3H and 14C) are useful in compound or substrate tissue distribution study. Incorporation of heavier isotopes such as deuterium (2H) can afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life, or reduced dosage requirements and, hence may be preferred in some instances.
  • Isotopically-labeled compounds of the present invention can generally be prepared by standard methods and techniques known to those skilled in the art or by procedures similar to those described in the accompanying Examples substituting appropriate isotopically-labeled reagents in place of the corresponding non-labeled reagent.
  • The invention also includes any or all metabolites of any of the compounds described. The metabolites may include any chemical species generated by a biotransformation of any of the compounds described, such as intermediates and products of metabolism of the compound, such as would be generated in vivo following administration to a human.
  • Articles of manufacture comprising a compound described herein, or a salt or solvate thereof, in a suitable container are provided. The container may be a vial, jar, ampoule, preloaded syringe, i.v. bag, and the like.
  • Preferably, the compounds detailed herein are orally bioavailable. However, the compounds may also be formulated for parenteral (e.g., intravenous) administration.
  • One or several compounds described herein can be used in the preparation of a medicament by combining the compound or compounds as an active ingredient with a pharmacologically acceptable carrier, which are known in the art. Depending on the therapeutic form of the medication, the carrier may be in various forms. In one variation, the manufacture of a medicament is for use in any of the methods disclosed herein, e.g., for the treatment of cancer.
  • General Synthetic Methods
  • The compounds of the invention may be prepared by a number of processes as generally described below and more specifically in the Examples hereinafter (such as the schemes provided in the Examples below). In the following process descriptions, the symbols when used in the formulae depicted are to be understood to represent those groups described above in relation to the formulae herein.
  • Where it is desired to obtain a particular enantiomer of a compound, this may be accomplished from a corresponding mixture of enantiomers using any suitable conventional procedure for separating or resolving enantiomers. Thus, for example, diastereomeric derivatives may be produced by reaction of a mixture of enantiomers, e.g., a racemate, and an appropriate chiral compound. The diastereomers may then be separated by any convenient means, for example by crystallization and the desired enantiomer recovered. In another resolution process, a racemate may be separated using chiral High Performance Liquid Chromatography. Alternatively, if desired a particular enantiomer may be obtained by using an appropriate chiral intermediate in one of the processes described.
  • Chromatography, recrystallization and other conventional separation procedures may also be used with intermediates or final products where it is desired to obtain a particular isomer of a compound or to otherwise purify a product of a reaction.
  • Solvates and/or polymorphs of a compound provided herein, or a pharmaceutically acceptable salt thereof are also contemplated. Solvates contain either stoichiometric or non-stoichiometric amounts of a solvent and are often formed during the process of crystallization. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Polymorphs include the different crystal packing arrangements of the same elemental composition of a compound. Polymorphs usually have different X-ray diffraction patterns, infrared spectra, melting points, density, hardness, crystal shape, optical and electrical properties, stability, and/or solubility. Various factors such as the recrystallization solvent, rate of crystallization, and storage temperature may cause a single crystal form to dominate.
  • In some embodiments, compounds of the formula (I) may be synthesized according to Scheme 1. In some embodiments, compounds of the formula (I) may be synthesized according to Scheme 1, 2 or 3.
  • Figure US20220298142A1-20220922-C00622
  • wherein A, B and R2 are as defined for formula (I), or any variation thereof detailed herein; and X is a leaving group (e.g., alkoxy or halogen).
  • Figure US20220298142A1-20220922-C00623
  • wherein A, B and R2b are as defined for formula (I), or any variation thereof detailed herein; and X is a leaving group (e.g., alkoxy or halogen).
  • Figure US20220298142A1-20220922-C00624
  • wherein A, B and R2 are as defined for formula (I), or any variation thereof detailed herein; and X is a leaving group (e.g., alkoxy or halogen).
  • Pharmaceutical Compositions and Formulations
  • Pharmaceutical compositions of any of the compounds detailed herein are embraced by this disclosure. Thus, the present disclosure includes pharmaceutical compositions comprising a compound as detailed herein or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier or excipient. In one aspect, the pharmaceutically acceptable salt is an acid addition salt, such as a salt formed with an inorganic or organic acid. Pharmaceutical compositions may take a form suitable for oral, buccal, parenteral, nasal, topical or rectal administration or a form suitable for administration by inhalation.
  • A compound as detailed herein may in one aspect be in a purified form and compositions comprising a compound in purified forms are detailed herein. Compositions comprising a compound as detailed herein or a salt thereof are provided, such as compositions of substantially pure compounds. In some embodiments, a composition containing a compound as detailed herein or a salt thereof is in substantially pure form.
  • In one variation, the compounds herein are synthetic compounds prepared for administration to an individual. In another variation, compositions are provided containing a compound in substantially pure form. In another variation, the present disclosure embraces pharmaceutical compositions comprising a compound detailed herein and a pharmaceutically acceptable carrier. In another variation, methods of administering a compound are provided. The purified forms, pharmaceutical compositions and methods of administering the compounds are suitable for any compound or form thereof detailed herein.
  • A compound detailed herein or salt thereof may be formulated for any available delivery route, including an oral, mucosal (e.g., nasal, sublingual, vaginal, buccal or rectal), parenteral (e.g., intramuscular, subcutaneous or intravenous), topical or transdermal delivery form. A compound or salt thereof may be formulated with suitable carriers to provide delivery forms that include, but are not limited to, tablets, caplets, capsules (such as hard gelatin capsules or soft elastic gelatin capsules), cachets, troches, lozenges, gums, dispersions, suppositories, ointments, cataplasms (poultices), pastes, powders, dressings, creams, solutions, patches, aerosols (e.g., nasal spray or inhalers), gels, suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in-water emulsions or water-in-oil liquid emulsions), solutions and elixirs.
  • One or several compounds described herein or a salt thereof can be used in the preparation of a formulation, such as a pharmaceutical formulation, by combining the compound or compounds, or a salt thereof, as an active ingredient with a pharmaceutically acceptable carrier, such as those mentioned above. Depending on the therapeutic form of the system (e.g., transdermal patch vs. oral tablet), the carrier may be in various forms. In addition, pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants. Formulations comprising the compound may also contain other substances which have valuable therapeutic properties. Pharmaceutical formulations may be prepared by known pharmaceutical methods. Suitable formulations can be found, e.g., in Remington's Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, Pa., 20th ed. (2000), which is incorporated herein by reference.
  • Compounds as described herein may be administered to individuals in a form of generally accepted oral compositions, such as tablets, coated tablets, and gel capsules in a hard or in soft shell, emulsions or suspensions. Examples of carriers, which may be used for the preparation of such compositions, are lactose, corn starch or its derivatives, talc, stearate or its salts, etc. Acceptable carriers for gel capsules with soft shell are, for instance, plant oils, wax, fats, semisolid and liquid poly-ols, and so on. In addition, pharmaceutical formulations may contain preservatives, solubilizers, stabilizers, re-wetting agents, emulgators, sweeteners, dyes, adjusters, and salts for the adjustment of osmotic pressure, buffers, coating agents or antioxidants.
  • Any of the compounds described herein can be formulated in a tablet in any dosage form described, for example, a compound as described herein or a pharmaceutically acceptable salt thereof can be formulated as a 10 mg tablet.
  • Compositions comprising a compound provided herein are also described. In one variation, the composition comprises a compound or salt thereof and a pharmaceutically acceptable carrier or excipient. In another variation, a composition of substantially pure compound is provided.
  • Methods of Use
  • Compounds and compositions detailed herein, such as a pharmaceutical composition containing a compound of any formula provided herein or a salt thereof and a pharmaceutically acceptable carrier or excipient, may be used in methods of administration and treatment as provided herein. The compounds and compositions may also be used in in vitro methods, such as in vitro methods of administering a compound or composition to cells for screening purposes and/or for conducting quality control assays.
  • Provided herein is a method of treating a disease in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or any embodiment, variation or aspect thereof (collectively, a compound of formulae (I), (II) or (III), or the present compounds or the compounds detailed or described herein) or a pharmaceutically acceptable salt thereof, to the individual. In some embodiments, provided herein is a method of treating a disease mediated by a G protein coupled receptor signaling pathway in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual. In some embodiments, the disease is mediated by a class A G protein coupled receptor. In some embodiments, the disease is mediated by a class B G protein coupled receptor. In some embodiments, the disease is mediated by a class C G protein coupled receptor. In some embodiments, the G protein coupled receptor is a purinergic G protein receptor. In some embodiments, the G protein coupled receptor is an adenosine receptor, such as any of the A1, A2A, A2B, and A3 receptors.
  • The present compounds or salts thereof are believed to be effective for treating a variety of diseases and disorders. For example, in some embodiments, the present compositions may be used to treat a proliferative disease, such as cancer. In some embodiments the cancer is a solid tumor. In some embodiments the cancer is any of adult and pediatric oncology, myxoid and round cell carcinoma, locally advanced tumors, metastatic cancer, human soft tissue sarcomas, including Ewing's sarcoma, cancer metastases, including lymphatic metastases, squamous cell carcinoma, particularly of the head and neck, esophageal squamous cell carcinoma, oral carcinoma, blood cell malignancies, including multiple myeloma, leukemias, including acute lymphocytic leukemia, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, chronic myelocytic leukemia, and hairy cell leukemia, effusion lymphomas (body cavity based lymphomas), thymic lymphoma lung cancer, including small cell carcinoma, cutaneous T cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cancer of the adrenal cortex, ACTH-producing tumors, nonsmall cell cancers, breast cancer, including small cell carcinoma and ductal carcinoma, gastrointestinal cancers, including stomach cancer, colon cancer, colorectal cancer, polyps associated with colorectal neoplasia, pancreatic cancer, liver cancer, urological cancers, including bladder cancer, including primary superficial bladder tumors, invasive transitional cell carcinoma of the bladder, and muscle-invasive bladder cancer, prostate cancer, malignancies of the female genital tract, including ovarian carcinoma, primary peritoneal epithelial neoplasms, cervical carcinoma, uterine endometrial cancers, vaginal cancer, cancer of the vulva, uterine cancer and solid tumors in the ovarian follicle, malignancies of the male genital tract, including testicular cancer and penile cancer, kidney cancer, including renal cell carcinoma, brain cancer, including intrinsic brain tumors, neuroblastoma, astrocytic brain tumors, gliomas, metastatic tumor cell invasion in the central nervous system, bone cancers, including osteomas and osteosarcomas, skin cancers, including melanoma, tumor progression of human skin keratinocytes, squamous cell cancer, thyroid cancer, retinoblastoma, neuroblastoma, peritoneal effusion, malignant pleural effusion, mesothelioma, Wilms's tumors, gall bladder cancer, trophoblastic neoplasms, hemangiopericytoma, and Kaposi's sarcoma.
  • In some embodiments, the present compounds or salts thereof are used in treatment of tumors which produce high levels of ATP and/or adenosine. For example, in some embodiments the extracellular concentration of adenosine is 10-20 times higher in the tumor compared to adjacent tissue. In some embodiments, the present compounds or salts thereof are used in treatment of tumors that express high levels of an ectonucleotidase. In some embodiments, the ectonucleotidase is CD39. In some embodiments, the ectonucleotidase is CD73.
  • Also provided herein is a method of enhancing an immune response in an individual in need thereof comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual. Adenosine receptors are known to play an immunosuppressive role in cancer biology. High levels of adenosine present in the tumor microenvironment bind to adenosine receptors on immune cells to provide an immunosuppressive microenvironment. Specifically, binding of adenosine to the A2A receptor provides an immunosuppressive signal that inhibits T cell proliferation, cytokine production and cytotoxicity. The A2A receptor signaling has been implicated in adenosine-mediated inhibition of NK cell cytotoxicity, NKT cell cytokine production and CD40L upregulation. Therefore, use of an A2A receptor antagonist, such as those provided herein, may reverse the immunosuppressive effect of adenosine on immune cells. In some embodiments, the immune response is enhanced by a compound of formulae (I), (II) or (III) or a salt thereof enhancing activity of natural killer (NK) cells. In some embodiments, the present compounds or salts thereof increase NK cell-meditated cytotoxicity. In some embodiments, the immune response is enhanced by enhancing the activity of CD8+T cells. In some embodiments, the present compounds or salts thereof cause an inflammatory response in the tumor microenvironment.
  • The present disclosure further provides a method of increasing the activity of a natural killer cell in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual. In some of these embodiments, the present compounds or salts thereof increase NK cell-meditated cytotoxicity. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof increases the number of NK cells.
  • A compound of formulae (I), (II) or (III) or a salt thereof may be useful for modulating the activity of G protein receptor coupled signaling pathway proteins. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof activates a G protein receptor coupled signaling pathway protein (i.e. is an agonist of a G protein receptor). In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof inhibits a G protein receptor coupled signaling pathway protein (i.e., is a G protein receptor antagonist). In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof is an adenosine receptor antagonist. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof is an antagonist of any of the A1, A2A, A2B, and A3 receptors.
  • Accordingly, also provided herein is a method of modulating the activity of an A2A receptor in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof to an individual. In some embodiments a compound of formulae (I), (II) or (III) or a salt thereof is an A2A receptor antagonist. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof reduces A2A receptor signaling by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof reduces A2A receptor signaling by 40-99%, 50-99%, 60-99%, 70-99%, 80-99%, 90-99%, or 95-99%. In some of these embodiments, a compound of formulae (I), (II) or (III) or a salt thereof binds to the A2A receptor with an IC50 of less than 1 μM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 10 nM, less than 1 nM or less than 100 pM. In some embodiments, [compound x] binds to the A2A receptor with an IC50 of 500 nM to 100 pM, 400 nM to 100 pM, 300 nM to 100 pM, 200 nM to 100 pM, or 100 nM to 100 pM.
  • Also provided herein is a method of modulating the activity of an A2B receptor in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof to an individual. In some embodiments a compound of formulae (I), (II) or (III) or a salt thereof is an A2B receptor antagonist. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof reduces A2B receptor signaling by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof reduces A2B receptor signaling by 40-99%, 50-99%, 60-99%, 70-99%, 80-99%, 90-99%, or 95-99%. In some of these embodiments, a compound of formulae (I), (II) or (III) or a salt thereof binds to the A2B receptor with an IC50 of less than 1 μM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 10 nM, less than 1 nM or less than 100 pM.
  • In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof binds to the A2B receptor with an IC50 of 500 nM to 100 pM, 400 nM to 100 pM, 300 nM to 100 pM, 200 nM to 100 pM, or 100 nM to 100 pM.
  • Also provided herein is a method of modulating the activity of an A3 receptor in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof to an individual. In some embodiments a compound of formulae (I), (II) or (III) or a salt thereof is an A3 receptor antagonist. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof reduces A3 receptor signaling by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof reduces A3 receptor signaling by 40-99%, 50-99%, 60-99%, 70-99%, 80-99%, 90-99%, or 95-99%. In some of these embodiments, a compound of formulae (I), (II) or (III) or a salt thereof binds to the A3 receptor with an IC50 of less than 1 μM, less than 900 nM, less than 800 nM, less than 700 nM, less than 600 nM, less than 500 nM, less than 400 nM, less than 300 nM, less than 200 nM, less than 100 nM, less than 10 nM, less than 1 nM or less than 100 pM. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof binds to the A3 receptor with an IC50 of 500 nM to 100 pM, 400 nM to 100 pM, 300 nM to 100 pM, 200 nM to 100 pM, or 100 nM to 100 pM.
  • In some embodiments, the present invention comprises a method of inhibiting tumor metastasis in an individual in need thereof comprising administering a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual. In some embodiments, the metastasis is to the lung, liver, lymph node, bone, adrenal gland, brain, peritoneum, muscle, or vagina. In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof inhibits metastasis of melanoma cells. In some embodiments, the present disclosure includes a method of delaying tumor metastasis comprising administering a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, to the individual. In some of these embodiments, the time to metastasis is delayed by 1 month, 2 months 3 months, 4 months, 5 months, 6 months, 12 months, or more, upon treatment with the compounds of the present invention.
  • In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof is used to treat an individual having a proliferative disease, such as cancer as described herein. In some embodiments, the individual is at risk of developing a proliferative disease, such as cancer. In some of these embodiments, the individual is determined to be at risk of developing cancer based upon one or more risk factors. In some of these embodiments, the risk factor is a family history and/or gene associated with cancer. In some embodiments, the individual has a cancer that expresses a high level of a nucleotide metabolizing enzyme. In some embodiments, the nucleotide metabolizing enzyme is a nucleotidase, such as CD73 (ecto-5′-nucleotidase, Ecto5′NTase). In some of these embodiments, the individual has a cancer that expresses a high level of a nucleotidase, such as CD73. In any of these embodiments, the nucleotide metabolizing enzyme is an ecto-nucleotidase. In some embodiments, the ecto-nucleotidase degrades adenosine monophosphate. In some embodiments, the nucleotide metabolizing enzyme is CD39 (ecto-nucleoside triphosphate diphosphohydrolase 1, E-NTPDase1). In some of these embodiments, the individual has a cancer that expresses a high level of CD39. In some embodiments, the individual has a cancer that expresses a high level of an adenosine receptor, such as the A2A receptor.
  • Combination Therapy
  • As provided herein, the presently disclosed compounds or a salt thereof may activate the immune system by modulating the activity of a G protein coupled receptor signaling pathway, for example acting as an A2A receptor antagonist, which results in significant anti-tumor effects. Accordingly, the present compounds or a salt thereof may be used in combination with other anti-cancer agents to enhance tumor immunotherapy. In some embodiments, provided herein is a method of treating a disease mediated by a G protein coupled receptor signaling pathway in an individual comprising administering an effective amount of a compound of formulae (I), (II) or (III), or a pharmaceutically acceptable salt thereof, and an additional therapeutic agent to the individual. In some embodiments, the disease mediated by a G protein coupled receptor signaling pathway is a proliferative disease such as cancer.
  • In some embodiments, the additional therapeutic agent is a cancer immunotherapy. In some embodiments, the additional therapeutic agent is an immunostimulatory agent. In some embodiments, the additional therapeutic agent targets a checkpoint protein. In some embodiments, the additional therapeutic agent is effective to stimulate, enhance or improve an immune response against a tumor.
  • In another aspect, provided herein is a combination therapy in which a compound of formulae (I), (II) or (III) is coadministered (which may be separately or simultaneously) with one or more additional agents that are effective in stimulating immune responses to thereby further enhance, stimulate or upregulate immune responses in a subject. For example, provided is a method for stimulating an immune response in a subject comprising administering to the subject a compound of formulae (I), (II) or (III) or a salt thereof and one or more immunostimulatory antibodies, such as an anti-PD-1 antibody, an anti-PD-L1 antibody and/or an anti-CTLA-4 antibody, such that an immune response is stimulated in the subject, for example to inhibit tumor growth. In one embodiment, the subject is administered a compound of formulae (I), (II) or (III) or a salt thereof and an anti-PD-1 antibody. In another embodiment, provided is a method for stimulating an immune response in a subject comprising administering to the subject a compound of formulae (I), (II) or (III) or a salt thereof and one or more immunostimulatory antibodies or immunotherapy like Chimeric antigen receptor (CAR) T-cell therapy; immunostimulatory antibodies such as an anti-PD-1 antibody, an anti-PD-L1 antibody and/or an anti-CTLA-4 antibody, such that an immune response is stimulated in the subject, for example to inhibit tumor growth. In another embodiment, the subject is administered a compound of formulae (I), (II) or (III) or a salt thereof and an anti-PD-L1 antibody. In yet another embodiment, the subject is administered a compound of formulae (I), (II) or (III) or a salt thereof and an anti-CTLA-4 antibody. In another embodiment, the immunostimulatory antibody (e.g., anti-PD-1, anti-PD-L1 and/or anti-CTLA-4 antibody) is a human antibody. Alternatively, the immunostimulatory antibody can be, for example, a chimeric or humanized antibody (e.g., prepared from a mouse anti-PD-1, anti-PD-L1 and/or anti-CTLA-4 antibody). In another embodiment, the subject is administered a compound of formulae (I), (II) or (III) or a salt thereof and CAR T-cells (genetically modified T cells).
  • In one embodiment, the present disclosure provides a method for treating a proliferative disease (e.g., cancer), comprising administering a compound of formulae (I), (II) or (III) or a salt thereof and an anti-PD-1 antibody to a subject. In further embodiments, a compound of formulae (I), (II) or (III) or a salt thereof is administered at a subtherapeutic dose, the anti-PD-1 antibody is administered at a subtherapeutic dose, or both are administered at a subtherapeutic dose. In another embodiment, the present disclosure provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound of formulae (I), (II) or (III) or a salt thereof and a subtherapeutic dose of anti-PD-1 antibody to a subject. In certain embodiments, the subject is human. In certain embodiments, the anti-PD-1 antibody is a human sequence monoclonal antibody
  • In one embodiment, the present invention provides a method for treating a hyperproliferative disease (e.g., cancer), comprising administering a compound of formulae (I), (II) or (III) or a salt thereof and an anti-PD-L1 antibody to a subject. In further embodiments, a compound of formulae (I), (II) or (III) or a salt thereof is administered at a subtherapeutic dose, the anti-PD-L1 antibody is administered at a subtherapeutic dose, or both are administered at a subtherapeutic dose. In another embodiment, the present invention provides a method for altering an adverse event associated with treatment of a hyperproliferative disease with an immunostimulatory agent, comprising administering a compound of formulae (I), (II) or (III) or a salt thereof and a subtherapeutic dose of anti-PD-L1 antibody to a subject. In certain embodiments, the subject is human. In certain embodiments, the anti-PD-L1 antibody is a human sequence monoclonal antibody.
  • In certain embodiments, the combination of therapeutic agents discussed herein can be administered concurrently as a single composition in a pharmaceutically acceptable carrier, or concurrently as separate compositions each in a pharmaceutically acceptable carrier. In another embodiment, the combination of therapeutic agents can be administered sequentially. For example, an anti-CTLA-4 antibody and a compound of formulae (I), (II) or (III) or a salt thereof can be administered sequentially, such as anti-CTLA-4 antibody being administered first and a compound of formulae (I), (II) or (III) or a salt thereof second, or a compound of formulae (I), (II) or (III) or a salt thereof being administered first and anti-CTLA-4 antibody second. Additionally, or alternatively, an anti-PD-1 antibody and a compound of formulae (I), (II) or (III) or a salt thereof can be administered sequentially, such as anti-PD-1 antibody being administered first and a compound of formulae (I), (II) or (III) or a salt thereof second, or a compound of formulae (I), (II) or (III) or a salt thereof being administered first and anti-PD-1 antibody second. Additionally, or alternatively, an anti-PD-L1 antibody and a compound of formulae (I), (II) or (III) or a salt thereof can be administered sequentially, such as anti-PD-L1 antibody being administered first and a compound of formulae (I), (II) or (III) or a salt thereof second, or a compound of formulae (I), (II) or (III) or a salt thereof being administered first and anti-PD-L1 antibody second.
  • Furthermore, if more than one dose of the combination therapy is administered sequentially, the order of the sequential administration can be reversed or kept in the same order at each time point of administration, sequential administrations can be combined with concurrent administrations, or any combination thereof.
  • Optionally, the combination of a compound of formulae (I), (II) or (III) or a salt thereof can be further combined with an immunogenic agent, such as cancerous cells, purified tumor antigens (including recombinant proteins, peptides, and carbohydrate molecules), cells, and cells transfected with genes encoding immune stimulating cytokines.
  • A compound of formulae (I), (II) or (III) or a salt thereof can also be further combined with standard cancer treatments. For example, a compound of formulae (I), (II) or (III) or a salt thereof can be effectively combined with chemotherapeutic regimes. In these instances, it is possible to reduce the dose of other chemotherapeutic reagent administered with the combination of the instant disclosure (Mokyr et al. (1998) Cancer Research 58: 5301-5304). Other combination therapies with a compound of formulae (I), (II) or (III) or a salt thereof include radiation, surgery, or hormone deprivation. Angiogenesis inhibitors can also be combined with a compound of formulae (I), (II) or (III) or a salt thereof. Inhibition of angiogenesis leads to tumor cell death, which can be a source of tumor antigen fed into host antigen presentation pathways.
  • In another example, a compound of formulae (I), (II) or (III) or a salt thereof can be used in conjunction with anti-neoplastic antibodies. By way of example and not wishing to be bound by theory, treatment with an anti-cancer antibody or an anti-cancer antibody conjugated to a toxin can lead to cancer cell death (e.g., tumor cells) which would potentiate an immune response mediated by CTLA-4, PD-1, PD-L1 or a compound of formulae (I), (II) or (III) or a salt thereof. In an exemplary embodiment, a treatment of a hyperproliferative disease (e.g., a cancer tumor) can include an anti-cancer antibody in combination with a compound of formulae (I), (II) or (III) or a salt thereof and anti-CTLA-4 and/or anti-PD-1 and/or anti-PD-L1 antibodies, concurrently or sequentially or any combination thereof, which can potentiate anti-tumor immune responses by the host. Other antibodies that can be used to activate host immune responsiveness can be further used in combination with a compound of formulae (I), (II) or (III) or a salt thereof.
  • In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof can be combined with an anti-CD73 therapy, such as an anti-CD73 antibody.
  • In some embodiments, a compound of formulae (I), (II) or (III) or a salt thereof can be combined with an anti-CD39 therapy, such as an anti-CD39 antibody.
  • In yet further embodiments, a compound of formulae (I), (II) or (III) or a salt thereof is administered in combination another G protein receptor antagonist, such as an adenosine A1 and/or A3 antagonist.
  • Dosing and Method of Administration
  • The dose of a compound administered to an individual (such as a human) may vary with the particular compound or salt thereof, the method of administration, and the particular disease, such as type and stage of cancer, being treated. In some embodiments, the amount of the compound or salt thereof is a therapeutically effective amount.
  • The effective amount of the compound may in one aspect be a dose of between about 0.01 and about 100 mg/kg. Effective amounts or doses of the compounds of the invention may be ascertained by routine methods, such as modeling, dose escalation, or clinical trials, taking into account routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the disease to be treated, the subject's health status, condition, and weight. An exemplary dose is in the range of about from about 0.7 mg to 7 g daily, or about 7 mg to 350 mg daily, or about 350 mg to 1.75 g daily, or about 1.75 to 7 g daily.
  • Any of the methods provided herein may in one aspect comprise administering to an individual a pharmaceutical composition that contains an effective amount of a compound provided herein or a salt thereof and a pharmaceutically acceptable excipient.
  • A compound or composition of the invention may be administered to an individual in accordance with an effective dosing regimen for a desired period of time or duration, such as at least about one month, at least about 2 months, at least about 3 months, at least about 6 months, or at least about 12 months or longer, which in some variations may be for the duration of the individual's life. In one variation, the compound is administered on a daily or intermittent schedule. The compound can be administered to an individual continuously (for example, at least once daily) over a period of time. The dosing frequency can also be less than once daily, e.g., about a once weekly dosing. The dosing frequency can be more than once daily, e.g., twice or three times daily. The dosing frequency can also be intermittent, including a ‘drug holiday’ (e.g., once daily dosing for 7 days followed by no doses for 7 days, repeated for any 14 day time period, such as about 2 months, about 4 months, about 6 months or more). Any of the dosing frequencies can employ any of the compounds described herein together with any of the dosages described herein.
  • The compounds provided herein or a salt thereof may be administered to an individual via various routes, including, e.g., intravenous, intramuscular, subcutaneous, oral and transdermal. A compound provided herein can be administered frequently at low doses, known as ‘metronomic therapy,’ or as part of a maintenance therapy using compound alone or in combination with one or more additional drugs. Metronomic therapy or maintenance therapy can comprise administration of a compound provided herein in cycles. Metronomic therapy or maintenance therapy can comprise intra-tumoral administration of a compound provided herein.
  • In one aspect, the invention provides a method of treating cancer in an individual by parenterally administering to the individual (e.g., a human) an effective amount of a compound or salt thereof. In some embodiments, the route of administration is intravenous, intra-arterial, intramuscular, or subcutaneous. In some embodiments, the route of administration is oral. In still other embodiments, the route of administration is transdermal.
  • The invention also provides compositions (including pharmaceutical compositions) as described herein for the use in treating, preventing, and/or delaying the onset and/or development of cancer and other methods described herein. In certain embodiments, the composition comprises a pharmaceutical formulation which is present in a unit dosage form.
  • Also provided are articles of manufacture comprising a compound of the disclosure or a salt thereof, composition, and unit dosages described herein in suitable packaging for use in the methods described herein. Suitable packaging is known in the art and includes, for example, vials, vessels, ampules, bottles, jars, flexible packaging and the like. An article of manufacture may further be sterilized and/or sealed.
  • Kits
  • The present disclosure further provides kits for carrying out the methods of the invention, which comprises one or more compounds described herein or a composition comprising a compound described herein. The kits may employ any of the compounds disclosed herein. In one variation, the kit employs a compound described herein or a pharmaceutically acceptable salt thereof. The kits may be used for any one or more of the uses described herein, and, accordingly, may contain instructions for the treatment of cancer.
  • Kits generally comprise suitable packaging. The kits may comprise one or more containers comprising any compound described herein. Each component (if there is more than one component) can be packaged in separate containers or some components can be combined in one container where cross-reactivity and shelf life permit.
  • The kits may be in unit dosage forms, bulk packages (e.g., multi-dose packages) or sub-unit doses. For example, kits may be provided that contain sufficient dosages of a compound as disclosed herein and/or a second pharmaceutically active compound useful for a disease detailed herein (e.g., hypertension) to provide effective treatment of an individual for an extended period, such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 7 months, 8 months, 9 months, or more. Kits may also include multiple unit doses of the compounds and instructions for use and be packaged in quantities sufficient for storage and use in pharmacies (e.g., hospital pharmacies and compounding pharmacies).
  • The kits may optionally include a set of instructions, generally written instructions, although electronic storage media (e.g., magnetic diskette or optical disk) containing instructions are also acceptable, relating to the use of component(s) of the methods of the present invention.
  • The instructions included with the kit generally include information as to the components and their administration to an individual.
  • The invention can be further understood by reference to the following examples, which are provided by way of illustration and are not meant to be limiting.
  • EXAMPLES Synthetic Examples Example S-1: Synthesis of N2-(2-(diethylamino)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2,3-diamine (Compound No. 1)
  • Figure US20220298142A1-20220922-C00625
  • Step-1: Synthesis of 6-phenylpyrazin-2-amine: To a stirred solution of 6-chloropyrazin-2-amine (50 g, 0.3861 mol) in dioxane:water (400 mL:100 mL) was added benzeneboronic acid (56.4 g, 0.46 mol). The reaction mixture was purged with nitrogen for 20 min then charged Na2CO3 (70.6 g, 0.57 mol) and Pd(PPh3)Cl2 (13.5 g, 0.01930 mol). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filter through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography over silica gel (100-200 mesh) [Ethyl acetate:Hexane (3:7) as eluent] to get the desired product (55 g, 83%). LCMS: 172 [M+1]+
  • Step-2: Synthesis of 5-bromo-6-phenylpyrazin-2-amine: To a stirred solution of 6-phenylpyrazin-2-amine (48 g, 0.2803 mol) in DMF was added NBS (49.9 g, 0.28 mol) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at RT for 16 h. The reaction was monitored by TLC and LCMS. The reaction was diluted with water and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography silica gel (100-200 mesh) [Ethyl acetate:Hexane (1:4) as eluent] to get the desired product (38 g, 55%). LCMS: 252 [M+2]+
  • Step-3: synthesis of 6-phenyl-5-(quinolin-6-yl)pyrazin-2-amine: To a stirred solution of 5-bromo-6-phenylpyrazin-2-amine (38 g, 0.1519 mol) in dioxane:water (320 mL:80 mL) was added quinolin-6-ylboronic acid (46.4 g, 0.18 mol). The reaction mixture was purged with nitrogen for 20 min then charged with Na2CO3 (32.2 g, 0.3038 mol) and Pd(dppf)Cl2 (6.19 g, 0.007 mol). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography over basic alumina [Ethyl acetate:Hexane (3:7) as eluent] to get the desired product (31 g, 68%). LCMS: 299 [M+1]+
  • Step-4: synthesis of 3-bromo-6-phenyl-5-(quinolin-6-yl)pyrazin-2-amine: To a stirred solution of 6-phenyl-5-(quinolin-6-yl) pyrazin-2-amine (21 g, 0.07 mol) in DMF was added NBS (12.5 g, 0.07 mol) at 0° C. under nitrogen atmosphere. The reaction mixture was stir at RT for 16 h. The reaction was monitored by TLC and LCMS. The reaction was diluted with water and extracted with ethyl acetate (3×30 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography over basic alumina [Ethyl acetate:Hexane (3:7) as eluent] to get the desired product (18 g, 69%). LCMS: 377 [M+1]+
  • Step-5: Synthesis of N2-(2-(diethylamino)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2,3-diamine: To a stirred solution of N1,N1-diethylethane-1,2-diamine (0.155 g, 1.32 mmol, 5.0 eq) in DMF (5 mL) was added Cs2CO3 (0.104 g, 0.31 mmol, 1.2 eq) and the mixture was stirred at RT for 15 min. To this mixture 3-bromo-6-phenyl-5-(quinolin-6-yl)pyrazin-2-amine (0.100 g, 0.26 mmol, 1.0 eq) was added and the resultant mixture was allowed to heat at 120° C. for 16 h. The progress of reaction was monitored by TLC. Upon completion, the mixture was diluted with water (40 mL), extracted with EtOAc (2×100 mL). The combined organic layers were washed with water (40 mL), brine (40 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to afford a crude residue which was purified by reverse phase column chromatography to afford the desired product as an off-white solid (3 mg, 4%). LCMS: 413 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ 8.82 (d, J=2.63 Hz, 2H), 8.10-8.21 (m, 2H), 7.93 (s, 1H), 7.87 (d, J=7.45 Hz, 1H), 7.79 (d, J=8.77 Hz, 1H), 7.58 (d, J=7.02 Hz, 1H), 7.40-7.51 (m, 2H), 7.13-7.31 (m, 5H), 6.82 (br s, 1H), 6.36 (br s, 2H), 3.71 (br s, 2H), 3.12 (br s, 2H), 2.98 (br s, 4H), 1.09 (t, J=7.02 Hz, 6H).
  • Example S-2: Synthesis of 3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 73)
  • Figure US20220298142A1-20220922-C00626
  • Step-1: Synthesis of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carbonitrile: To a stirred solution of NaCN (1.56 g, 0.03 mol) and CuCN (5.7 g, 0.06 mol) in dry DMF (150 mL) was added 3-bromo-6-phenyl-5-(quinolin-6-yl)pyrazin-2-amine (12.0 g, 0.03 mol) at 120° C. The reaction mixture was stirred at 145° C. for 12 h. The reaction was monitored by TLC and LCMS. The reaction was distilled. The crude product was poured in ice-water the solid precipitate out. The reaction mixture pH was adjusted with aqueous ammonia and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography using basic alumina [Ethyl acetate:Hexane (1:1) as eluent] to get the desired product (3.8 g, 34%). LCMS: 324 [M+1]+.
  • Step-2: Synthesis of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid: To a stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carbonitrile (1 g, 3.08 mmol, 1 eq) in 1,4-dioxane (50 mL) and aqueous NaOH (10%, 50 mL) was heated at 100° C. for 48 h. Progress of reaction was monitored by LCMS. On completion of the reaction, the reaction mixture was concentrated under vacuum to get the solid residue which was diluted with water (15 mL) and acidified with 3N HCl solution (10 mL), and extracted with ethyl acetate (50 mL×2). Organic layer was washed with water (100 mL×2), dried over anhydrous Na2SO4 and concentrated under vacuum to get the solid residue which was used as such for next step without further purification (950 mg, 91%). LCMS: 343 [M+1]+.
  • Step-3: Synthesis of 3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To a stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.100 g, 0.30 mmol, 1 eq) and 1-(pyridin-2-yl)ethan-1-amine (0.07 g, 0.58 mmol, 2.0 eq) in DMF (2 mL), was added HOBT (0.06 g, 0.43 mmol, 1.5 eq), EDC.HCl (0.08 g, 0.43 mmol, 1.5 eq) and DIPEA (0.113 g, 0.87 mmol, 3.0 eq). The reaction mixture was allowed to stir at RT for 24 h. Progress of reaction was monitored by TLC and LCMS. On completion of the reaction, the reaction mixture was diluted with water (15 mL) and extracted with ethyl acetate (50 mL×2). Combined organic layer was washed with water (50 mL×2), dried over anhydrous Na2SO4 and concentrated under vacuum to get the solid residue which was purified by normal phase column chromatography to get the desired product (5 mg, 2%). LCMS: 447 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ 9.18 (d, J=7.45 Hz, 1H), 9.02 (br s, 1H), 8.62 (br s, 2H), 8.52 (s, 1H), 8.14 (br s, 1H), 7.98 (d, J=8.77 Hz, 2H), 7.84 (d, J=8.77 Hz, 1H), 7.68 (br s, 3H), 7.26-7.44 (m, 5H), 5.29 (d, J=6.58 Hz, 1H), 1.59 (d, J=7.02 Hz, 3H).
  • Example-S-3: Synthesis of (S)-3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide. (Compound No. 75)
  • Figure US20220298142A1-20220922-C00627
  • Step-1: Synthesis of (S, E)-2-methyl-N-(pyridin-2-ylmethylene)propane-2-sulfinamide: To a stirred solution of pyridine-2-carboxaldehyde (1 g, 9.34 mmol, 1.0 eq) and copper(II) sulfate (2.98 g, 18.69 mmol, 2.0 eq) in dichloromethane (15 mL) was added (S)-2-methylpropane-2-sulfinamide (1.13 g, 9.34 mmol, 1.0 eq) at RT. The resulting mixture was heated at 50° C. for 16 h. Following this, reaction mixture was allowed to cool to room temperature, filtered through celite pad, the celite pad washed with dichloromethane (30 mL). The combined filtrate dried over anhydrous Na2SO4 and concentrated under vacuum to get the solid residue which was purified by flash column chromatography to get the desired product as white solid (1.2 g, 61%). LCMS: 211[M+1]+
  • Step-2: Synthesis of (S)-2-methyl-N—((S)-1-(pyridin-2-yl)ethyl)propane-2-sulfinamide: To a stirred solution of (S,E)-2-methyl-N-(pyridin-2-ylmethylene)propane-2-sulfinamide (1.0 g, 4.76 mmol, 1.0 eq) in tetrahydrofuran (15 mL) was added drop wise 3 M methylmagnesium bromide (2.38 mL, 7.14 mmol, 1.5 eq) at −78° C. The resulting mixture was stirred for 4 h at same temperature. The reaction was then quenched by careful addition of saturated NH4Cl (10 mL). The aqueous layer was separated and extracted with ethyl acetate (3×50 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated to give crude solid residue which was purified by flash column chromatography to get the desired product as semi solid (0.72 g, 71%). LCMS: 227.0 [M+1]+;
  • Step-3: Synthesis of (S)-1-(pyridin-2-yl)ethanamine: To a stirred solution of (S)-2-methyl-N—((S)-1-(pyridin-2-yl)ethyl)propane-2-sulfinamide (0.7 g, 3.09 mmol, 1.0 eq) in methanol (5 mL) was added 4N HCl in dioxane (1.6 ml) at RT. The resulting mixture was stirred for 30 min. Following this the reaction mixture was evaporated under reduced pressure to get solid residue. The obtained solid was washed with diethyl ether, died under vacuum to get desired product as off white solid (0.35 g, 94%). LCMS: 123 [M+1]+
  • Step-4: Synthesis of (S)-3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.1 g, 0.29 mmol, 1.0 eq) in DMF (5.0 ml) was added (S)-1-(pyridin-2-yl)ethanamine (0.042 g, 0.35 mmol, 1.2 eq), DIPEA (0.15 mL, 0.87 mmol, 3 eq) and HATU (0.22 g, 0.58 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. Following this, ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by column chromatography using basic alumina (18 mg, 14%). LCMS: 447 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 9.20 (d, J=7.45 Hz, 1H), 9.10 (d, J=3.51 Hz, 1H), 8.68 (d, J=3.51 Hz, 2H), 8.25 (br s, 1H), 8.13 (br s, 1H), 8.03 (d, J=8.77 Hz, 1H), 7.91 (d, J=8.77 Hz, 1H), 7.75-7.84 (m, 3H), 7.58 (br s, 1H), 7.35-7.43 (m, 3H), 7.28-7.35 (m, 2H), 5.28-5.38 (m, 1H), 2.87-2.97 (m, 1H), 1.62 (d, J=7.02 Hz, 3H), 1.16 (t, J=7.24 Hz, 1H).
  • Example-S-4: Synthesis of (R)-3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide. (Compound No. 76)
  • Figure US20220298142A1-20220922-C00628
  • Step-1: Synthesis of 2-methyl-N-[(E)-pyridin-2-ylmethylidene]propane-2-sulfinamide: To a stirred solution of pyridine-2-carboxaldehyde (1 g, 9.34 mmol, 1.0 eq) and copper(II) sulfate (2.98 g, 18.69 mmol, 2.0 eq) in dichloromethane (15 mL) was added (R)-2-methylpropane-2-sulfinamide (1.13 g, 9.34 mmol, 1.0 eq) at RT. The resulting mixture was heated at 50° C. for 16 h. Following this, reaction was allowed to cool to room temperature, filtered through celite pad, the celite pad washed with dichloromethane (30 mL). The combined filtrate dried over anhydrous Na2SO4 and concentrated under vacuum to get the solid residue which was purified by flash column chromatography to get the desired product as white solid (800 mg, 40%) LCMS: 210 [M+1]+
  • Step-2: Synthesis of 2-methyl-N-[(1R)-1-(pyridin-2-yl)ethyl]propane-2-sulfinamide: To a stirred solution of 2-methyl-N-[(E)-pyridin-2-ylmethylidene]propane-2-sulfinamide (800 mg, 3.80 mmol, 1.0 eq) in tetrahydrofuran (10 mL) was added drop wise 3 M methylmagnesium bromide (2.5 mL, 7.61 mmol, 2.0 eq) at −78° C. The resulting mixture was stirred for 4 h at same temperature. The reaction was then quenched by addition of saturated NH4Cl (10 mL). The aqueous layer was separated and extracted with ethyl acetate (3×50 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated to give crude solid residue which was purified by flash column chromatography to get the desired product as semi solid (600 mg, 75%). LCMS: 227.0 [M+1]+
  • Step-3: Synthesis of (1R)-1-(pyridin-2-yl)ethanamine: To a stirred solution of 2-methyl-N-[(1R)-1-(pyridin-2-yl)ethyl]propane-2-sulfinamide (600 mg, 2.65 mmol, 1.0 eq) in methanol (10 mL) was added 4N HCl in dioxane (2.5 ml) at RT. The resulting mixture was stirred for 30 min. Following this the reaction mixture was evaporated under reduced pressure to get solid residue. The obtained solid was washed with diethyl ether, died under vacuum to get desired product as solid (0.35 g, 94%). LCMS: 123 [M+1]+
  • Step-4: Synthesis of (R)-3-amino-5-phenyl-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.1 g, 0.29 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine e (68 mg, 0.43 mmol, 1.5 eq), DIPEA (0.2 mL, 0.87 mmol, 3 eq) and HATU (220 mg, 0.58 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. Following this, ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the desired product (10 mg, 8%) LCMS: 447 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 9.18 (d, J=8.33 Hz, 2H), 8.91 (d, J=2.63 Hz, 1H), 8.57 (d, J=5.26 Hz, 1H), 8.29 (d, J=7.45 Hz, 1H), 8.03 (br s, 1H), 7.92 (d, J=8.77 Hz, 1H), 7.82 (d, J=7.02 Hz, 2H), 7.73 (d, J=7.02 Hz, 1H), 7.50-7.57 (m, 2H), 7.38-7.42 (m, 2H), 7.28-7.38 (m, 4H), 5.22-5.27 (m, 2H), 2.87-2.97 (m, 4H), 1.55 (d, J=7.02 Hz, 3H), 1.16 (t, J=7.45 Hz, 4H).
  • Example S-5: Synthesis of 3-amino-S-phenyl-6-(quinolin-6-yl)-N-(2-(tetrahydro-2H-pyran-4-yl)ethyl)pyrazine-2-carboxamide. (Compound No. 89)
  • Figure US20220298142A1-20220922-C00629
  • To a stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.100 g, 0.30 mmol, 1 eq) and 2-(tetrahydro-2H-pyran-4-yl)ethan-1-amine (0.07 g, 0.58 mmol, 2.0 eq) in DMF (5 mL), was added HOBT (0.06 g, 0.43 mmol, 1.5 eq), EDC.HCl (0.08 g, 0.43 mmol, 1.5 eq) and DIPEA (0.113 g, 0.87 mmol, 3.0 eq). The reaction mixture was allowed to stir at RT for 24 h. Progress of reaction was monitored by TLC and LCMS. On completion of the reaction, the reaction mixture was diluted with water (15 mL) and extracted with ethyl acetate (50 mL×2). Combined organic layer were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid residue which was purified by reverse phase column chromatography to get the desired product (3 mg, 2%). LCMS: 454 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ 8.84-8.90 (m, 1H), 8.74 (s, 1H), 8.43 (s, 1H), 8.21 (d, J=7.45 Hz, 1H), 8.03 (br s, 1H), 7.85 (d, J=8.33 Hz, 1H), 7.66-7.73 (m, 1H), 7.51 (dd, J=3.95, 8.33 Hz, 1H), 7.26-7.43 (m, 5H), 6.62 (s, 1H), 4.66 (s, 1H), 4.09 (s, 2H), 3.83 (d, J=7.89 Hz, 2H), 3.17 (d, J=4.39 Hz, 2H), 1.52 (d, J=7.45 Hz, 2H), 1.23 (br s, 4H).
  • Example-S-6: Synthesis of 3-(6-amino-3-(8-chloroquinolin-6-yl)pyrazin-2-yl)-2-methylbenzonitrile. (Compound No. 2-2)
  • Figure US20220298142A1-20220922-C00630
  • Step-1: Synthesis of 6-chloro-5-(8-chloroquinolin-6-yl)pyrazin-2-amine: To a stirred solution of 5-bromo-6-chloropyrazin-2-amine (4.0 g, 19.23 mmol, 1 eq) in dioxane:water (180 mL:20 mL) was added 8-chloroquinolin-6-ylboronic acid (5.0 g, 17.30 mmol, 0.9 eq). The reaction mixture was purged with nitrogen for 20 min then charged with Na2CO3 (4.1 g, 39.0 mmol, 2.0 eq) and Pd(dppf)Cl2.DCM (787 mg, 5 mol %). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by reverse phase column chromatography to get the desired product (2.2 g, 78%). LCMS: 292 [M+1]+
  • Step-2: Synthesis of 3-(6-amino-3-(8-chloroquinolin-6-yl)pyrazin-2-yl)-2-methylbenzonitrile: To a stirred solution of 6-chloro-5-(8-chloroquinolin-6-yl)pyrazin-2-amine (0.100 g, 0.35 mmol, 1 eq) in dioxane:water (4 mL: 10 mL) was added 2-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzonitrile (0.9 g, 0.31 mmol, 1.2 eq). The reaction mixture was purged with nitrogen for 20 min then charged with Na2CO3 (0.073 g, 0.69 mmol, 2.0 eq) and Pd(dppf)Cl2.DCM (14 mg, 5 mol %). The reaction mixture was again purged with nitrogen. The reaction mixture was stiffed at RT for 10 min followed by heating at 100° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by reverse phase column chromatography to get the desired product (0.05 g, 38%). LCMS: 372 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 8.94 (dd, J=1.53, 4.17 Hz, 1H), 8.24 (dd, J=1.75, 8.33 Hz, 1H), 8.11 (s, 1H), 7.82 (d, J=6.58 Hz, 1H), 7.73 (d, J=1.75 Hz, 1H), 7.66 (d, J=1.75 Hz, 1H), 7.51-7.60 (m, 2H), 7.35-7.42 (m, 1H), 6.94 (s, 2H), 2.20 (s, 3H).
  • Example-S-7: Synthesis of 3-(6-amino-3-(8-chloroquinolin-6-yl)pyrazin-2-yl)-2-fluorobenzonitrile (Compound No. 2-10)
  • Figure US20220298142A1-20220922-C00631
  • To a stirred solution of 6-chloro-5-(8-chloroquinolin-6-yl)pyrazin-2-amine (0.05 g, 0.18 mmol, 1 eq) in dioxane:water (4 mL:10 mL) was added 2-fluoro-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)benzonitrile (0.9 g, 0.15 mmol 1.2 eq). The reaction mixture was purged with nitrogen for 20 min then charged with Na2CO3 (0.073 g, 0.35 mmol, 2.0 eq) and Pd(dppf)Cl2.DCM (7 mg, 5 mol %). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 100° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by reverse phase column chromatography to get the desired product (0.008 g, 12%). LCMS: 376 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 8.93-8.99 (m, 1H), 8.30 (dd, J=1.53, 8.55 Hz, 1H), 8.12 (s, 1H), 7.98 (t, J=5.92 Hz, 1H), 7.88 (t, J=6.80 Hz, 1H), 7.83 (d, J=1.75 Hz, 1H), 7.75 (d, J=1.75 Hz, 1H), 7.59 (td, J=3.84, 8.11 Hz, 1H), 7.47 (t, J=7.67 Hz, 1H), 7.02 (s, 2H).
  • Example-S-8: Synthesis of 3-amino-N-((6-cyanopyridin-2-yl)methyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 98)
  • Figure US20220298142A1-20220922-C00632
  • To a stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (100 mg, 0.29 mmol, 1.0 eq) in DMF (10 mL) was added 6-(aminomethyl)pyridine-2-carbonitrile (58 mg, 0.43 mmol, 1.5 eq) and the mixture was stirred at RT for 5 min. To this mixture HATU (220 mg, 0.58 mmol, 2.0 eq) and DIPEA (108 mg, 0.87 mmol, 3.0 eq) was added and the resultant mixture was allowed to stir for 16 h. The progress of reaction was monitored by TLC. Upon completion, the mixture was diluted with water (40 mL), extracted with EtOAc (2×100 mL). The combined organic layers were washed with water (40 mL), brine (40 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to afford a crude residue which was purified by reverse phase column chromatography to afford the desired product as an off-white solid (14 mg, 11%). LCMS: 458[M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 9.50 (t, J=6.14 Hz, 1H), 8.87 (d, J=3.07 Hz, 1H), 8.21 (d, J=7.89 Hz, 1H), 8.00-8.08 (m, 2H), 7.95 (d, J=7.45 Hz, 1H), 7.86 (d, J=8.77 Hz, 1H), 7.68-7.77 (m, 3H), 7.50 (dd, J=4.39, 8.33 Hz, 1H), 7.27-7.45 (m, 5H), 4.69 (d, J=6.14 Hz, 2H).
  • Example S-9: Synthesis of 3-amino-S-phenyl-N-(2-(pyridin-2-yl)propan-2-yl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: (Compound No. 108)
  • Figure US20220298142A1-20220922-C00633
  • Step 1: Synthesis of 2-(pyridin-2-yl)propan-2-amine: To a stirred solution of anhydrous cerium (III) chloride (7.1 g, 0.0288 mmol, 3 eq) in THF (15 mL) was added methyl lithium (1.6 M, 18 mL, 0.0288 mmol, 3 eq) at −78° C. The reaction was allowed to stir at same temperature for 30 min. Picolinonitrile (1 g, 0.0096 mmol, 3 eq) in THF was added at −78° C. and reaction was allowed to stir at RT for 1 h. The reaction was cooled to −40° C. then charged with 10 mL ammonium hydroxide. The reaction was allowed to stir at RT for 16 h. The solid was filtered and wash with THF. The organic layer was concentrated under vacuum to get the title compound which was used directly for next step without further purification (0.460 g, Crude); LCMS: 137 [M+1]+
  • Step 2: Synthesis of 3-amino-5-phenyl-N-(2-(pyridin-2-yl)propan-2-yl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To a stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (100 mg, 0.2924 mmol, 1.0 eq) and 2-(pyridin-2-yl)propan-2-amine (222 mg, 0.3508 mmol, 2 eq) in DMF (5 mL) was added DIPEA (0.150 mL, 0.8772 mmol, 3.0 eq) at 0° C. The reaction was allowed to stir at same temperature for 10 min. Then charged HATU (222 mg, 0.5848 mmol, 2.0 eq) and the reaction was allowed to stir at RT for 16 h. The reaction mixture was quenched with cold water (5 mL) then extracted using ethyl acetate (2×50 mL). The combined organic layers were washed with brine, dried (anhydrous sodium sulphate) and concentrated under vacuum to get the solid which was purified by SFC to get the title compound (8 mg, 6%). LCMS: 461 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ 10.08 (s, 1H), 8.89 (d, J=2.63 Hz, 1H), 8.57 (d, J=4.82 Hz, 1H), 8.26 (d, J=7.89 Hz, 1H), 8.05 (s, 1H), 7.94 (d, J=8.77 Hz, 1H), 7.84-7.92 (m, 1H), 7.75 (d, J=8.33 Hz, 2H), 7.67 (d, J=8.33 Hz, 2H), 7.53 (dd, J=3.95, 8.33 Hz, 2H), 7.41-7.49 (m, 2H), 7.30-7.41 (m, 4H), 1.80 (s, 6H).
  • Example S-10: Synthesis of 3-amino-N-(2-(diethylamino)ethyl)-S-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 107)
  • Figure US20220298142A1-20220922-C00634
  • To a stirred solution of 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.1 g, 0.29 mmol, 1.0 eq) in DMF (10 mL) was added N,N-diethylethane-1,2-diamine (51 mg, 0.43 mmol, 1.5 eq), DIPEA (0.2 mL, 0.87 mmol, 3 eq) and HATU (220 mg, 0.58 mmol, 2 eq) at RT under inert condition. The resulting mixture was stirred for 16 h at same temperature. Following this, ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the desired product (8 mg, 7%). LCMS: 441 [M+1]+; 1H NMR (DMSO-d6, 400 MHz) δ 8.87 (dd, J=4.2, 1.5 Hz, 1H), 8.70 (t, J=5.7 Hz, 1H), 8.19 (d, J=7.5 Hz, 1H), 7.99 (d, J=2.2 Hz, 1H), 7.85 (d, J=8.8 Hz, 1H), 7.65 (dd, J=8.8, 2.2 Hz, 1H), 7.51 (dd, J=8.3, 4.4 Hz, 1H), 7.26-7.43 (m, 5H), 3.36-3.43 (m, 3H), 2.60 (t, J=6.8 Hz, 3H), 2.52-2.56 (m, 7H), 0.97 (t, J=7.0 Hz, 6H).
  • Example S-11: Synthesis of (R)-3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 106)
  • Figure US20220298142A1-20220922-C00635
    Figure US20220298142A1-20220922-C00636
  • Step 1: Synthesis of 6-(4-fluorophenyl)pyrazin-2-amine: To a stirred solution of 6-chloropyrazin-2-amine (2 g, 15.50 mmol, 1 eq) in dioxane:water (50 mL:10 mL) was added 4-fluorobenzeneboronic acid (2.8 g, 20.15 mmol, 1.3 eq). The reaction mixture was purged with nitrogen for 20 min then charged K2CO3 (4.2 g, 31.0 mmol, 2.0 eq) and Pd(dppf)Cl2.DCM complex (632 mg, 0.77 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography over silica gel (100-200 mesh) [Ethyl acetate:Hexane (5:5) as eluent] to get the title compound (2 g, 68%). LCMS: 190 [M+1]+.
  • Step 2: Synthesis of 5-bromo-6-(4-fluorophenyl)pyrazin-2-amine: To a stirred solution of 6-(4-fluorophenyl)pyrazin-2-amine (2 g, 10.50 mmol, 1.0 eq) in DMF (20 mL) was added NBS (1.9 g, 10.50 mmol, 1.0 eq) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at RT for 30 min. The reaction was monitored by TLC and LCMS. The reaction was diluted with water and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography silica gel (100-200 mesh) [Ethyl acetate:Hexane (2:8) as eluent] to get the title compound (2.5 g, 89%). LCMS: 269 [M+1]+.
  • Step 3: 5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)pyrazin-2-amine: To a stirred solution of 5-bromo-6-(4-fluorophenyl)pyrazin-2-amine (4.5 g, 16.79 mmol, 1 eq) in dioxane:water (50 mL:10 mL) was added 8-chloroquinolin-6-ylboronic acid (5.8 g, 20.14 mmol 1.2 eq). The reaction mixture was purged with nitrogen for 20 min then charged with K2CO3 (4.6 g, 33.58 mmol, 2.0 eq) and Pd(dppf)Cl2.DCM complex (685 mg, 0.83 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography over basic alumina [Ethyl acetate:Hexane (3:7) as eluent] to get the title compound (5 g, 86%). LCMS: 351 [M+1]+.
  • Step 4: Synthesis of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)pyrazin-2-amine. To a stirred solution of 5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)pyrazin-2-amine (1 g, 2.85 mmol, 1 eq) in DMF (20 mL) was added NBS (498 mg, 2.85 mmol, 1 e.q) at 0° C. under nitrogen atmosphere. The reaction mixture was stirred at RT for 30 min. The reaction was monitored by TLC and LCMS. The reaction was diluted with water and extracted with ethyl acetate (3×30 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography [Ethyl acetate:Hexane (7:3) as eluent] to get the title compound (600 mg, 73%). LCMS: 429 [M+1]+.
  • Step-5 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)pyrazine-2-carbonitrile: To a stirred solution of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)pyrazin-2-amine (500 mg, 1.16 mmol, 1.0 eq) in DMF (5 mL) was added cuprous cyanide (0.104 g, 3.50 mmol, 3.0 eq). The reaction mixture was allowed to heat at 150° C. for 1 h using microwave irradiation. The reaction mixture was allowed to cool to RT, diluted with aqueous ammonia (5 mL) and extracted using ethyl acetate (3×25 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by reverse phase column chromatography to get the title compound (0.20 g, 46%). LCMS: 376[M+1]+.
  • Step-6 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid: To a stirred solution of 3-bromo-6-phenyl-5-(quinolin-6-yl)pyrazin-2-amine (0.2 g, 0.53 mmol, 1.0 eq) in ethanol (5 mL) was added 6M NaOH solution (5 mL). The resulting reaction mixture was heated at 100° C. for 16 h. The reaction mixture was allowed to cool to RT. The solvent was evaporated under vacuum and acidified using 1N HCl to get the solid which was filtered and dried to get the title compound (0.20 g, 95%). LCMS: 394 [M+1]+.
  • Step-7: (R)-3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid (0.1 g, 0.25 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine (47 mg, 0.43 mmol, 1.5 eq), DIPEA (0.2 mL, 0.75 mmol, 3 eq) and HATU (190 mg, 0.50 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. Following this, ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the title compound (10 mg, 8%). LCMS: 499 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 9.28 (d, J=7.9 Hz, 1H), 9.00 (d, J=3.1 Hz, 1H), 8.58 (d, J=3.9 Hz, 1H), 8.32 (d, J=7.9 Hz, 1H), 8.02 (s, 1H), 7.90 (s, 1H), 7.81 (t, J=6.8 Hz, 2H), 7.63 (dd, J=8.3, 3.9 Hz, 1H), 7.44-7.54 (m, 3H), 7.29-7.36 (m, 1H), 7.19 (t, J=8.8 Hz, 2H), 5.18-5.30 (m, 1H), 1.55 (d, J=7.0 Hz, 3H).
  • Example S-12: Synthesis of 3-(6-amino-3-(8-chloroquinolin-6-yl)pyrazin-2-yl)benzonitrile (Compound No. 2-6)
  • Figure US20220298142A1-20220922-C00637
  • To a stirred solution of 6-chloro-5-(8-chloroquinolin-6-yl)pyrazin-2-amine (0.110 g 0.38 mmol, 1.0 eq) in dioxane (5 mL):water (1 mL) was added 3-cyanophenylboronic acid (0.103 g, 0.45 mmol, 1.2 eq), Na2CO3 (0.80 g, 0.76 mmol, 2.0 eq) and PdCl2(dppf).DCM complex (0.015 g, 5 mol %). The reaction mixture was deoxygenated using N2 atmosphere and the reaction mixture was heated at 100° C. for 18 h. The reaction was monitored by TLC and LCMS. The reaction mixture was diluted with water (50 mL) and extracted using ethyl acetate (3×50 mL). The separated organic layer was dried over sodium sulfate and concentrated under reduced pressure. The crude product was purified by normal phase silica gel column chromatography to afford (0.015 g, 11%) the title compound. LCMS: 358 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ 8.97 (dd, J=1.53, 4.17 Hz, 1H), 8.31 (dd, J=1.75, 8.33 Hz, 1H), 8.06 (s, 1H), 7.91 (s, 1H), 7.76-7.85 (m, 3H), 7.54-7.63 (m, 2H), 7.42-7.51 (m, 1H), 6.94 (s, 2H).
  • Example S-13: Synthesis of Synthesis of S-(6-amino-3-(8-chloroquinolin-6-yl)pyrazin-2-yl)-2-fluorobenzonitrile (Compound No. 2-13)
  • Figure US20220298142A1-20220922-C00638
  • To a stirred solution of 6-chloro-5-(8-chloroquinolin-6-yl)pyrazin-2-amine (0.200 g, 0.68 mmol, 1 eq) in dioxane:water (8 mL:2 mL) was added 3-cyano-4-fluorophenylboronic acid (0.136 g, 0.82 mmol 1.2 eq) The reaction mixture was purged with nitrogen for 20 min then charged with Na2CO3 (0.146 g, 1.38 mmol, 2.0 eq) and Pd(dppf)Cl2.DCM complex (28 mg, 5 mol %). The reaction mixture was again purged with nitrogen. The reaction mixture was stiffed at RT for 10 min followed by heating at 100° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by reverse phase column chromatography to get the title compound (0.04 g, 15%). LCMS: 376 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ8.92-9.02 (m, 1H), 8.33 (d, J=7.02 Hz, 1H), 8.06 (s, 1H), 8.01 (dd, J=2.19, 6.14 Hz, 1H), 7.81 (d, J=1.75 Hz, 1H), 7.85 (d, J=1.75 Hz, 1H), 7.57-7.70 (m, 2H), 7.43 (t, J=8.99 Hz, 1H), 6.94 (s, 2H).
  • Example S-14: Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-N-((6-cyanopyridin-2-yl)methyl)-S-(4-fluorophenyl)pyrazine-2-carboxamide (Compound No. 105)
  • Figure US20220298142A1-20220922-C00639
  • To stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid (0.1 g, 0.25 mmol, 1.0 eq) in DMF (10 ml) was added 6-(aminomethyl)picolinonitrile (51 mg, 0.38 mmol, 1.5 eq), DIPEA (0.2 mL, 0.75 mmol, 3 eq) and HATU (190 mg, 0.50 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. Following this, ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the desired product (10 mg, 7%) LCMS: 510 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 9.54 (t, 1H), 8.98 (d, 1H), 8.28 (d, 1H), 8.00-8.05 (m, 2H), 7.91-7.96 (m, 2H), 7.71 (d, 1H), 7.61 (dd, 1H), 7.48 (dd, 5.5, 2H), 7.19 (t, 2H), 4.69 ppm (d, 2H).
  • Example S-15: Synthesis of (R)-3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 104)
  • Figure US20220298142A1-20220922-C00640
    Figure US20220298142A1-20220922-C00641
  • Step-1 Synthesis of 5-(7-chloro-1H-indazol-5-yl)-6-(4-fluorophenyl)pyrazin-2-amine. To a stirred solution of 5-bromo-6-(4-fluorophenyl)pyrazin-2-amine (1.3 g, 4.84 mmol, 1.0 eq) in dioxane:water (50 mL:10 mL) was added 7-chloro-5-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-indazole (1.6 g, 5.80 mmol, 1.2 eq). The reaction mixture was purged with nitrogen for 20 min then charged with K2CO3 (1.3 g 9.63 mmol, 2.0 eq) and Pd(dppf)Cl2.DCM complex (197 mg, 0.02 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography over silica gel (100-200 mesh) [Ethyl acetate:Hexane (5:5) as eluent] to get the title compound (300 mg, 18%). LCMS: 340 [M+1]+.
  • Step-2 Synthesis of 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(4-fluorophenyl)pyrazin-2-amine: To a stirred solution of 5-(7-chloro-1H-indazol-5-yl)-6-(4-fluorophenyl)pyrazin-2-amine (533 mg, 1.45 mmol, 1 eq) in DMF (10 ml) was added NBS (259 mg, 1.45 mmol, 1.0 eq) at 0° C. Reaction mixture was stirred at 0° C. for 30 min. The reaction was monitored by TLC and LCMS and found to be complete after 30 min. The reaction mixture was quenched with cold water 10 mL and was extracted with EtOAc (2×20 ml). The resulting solution was concentrated under reduced pressure. The crude product was purified by normal phase silica gel column chromatography to get the title compound (483 mg, 73%) LCMS: 376[M+1]+.
  • Step-3 Synthesis 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)pyrazine-2-carbonitrile: To a stirred solution of 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(4-fluorophenyl)pyrazin-2-amine (483 mg, 1.15 mmol, 1.0 eq) in DMF (10 mL) was added cuprous cyanide (206 mg, 2.30 mmol, 2.0 eq). The reaction mixture was allowed to stir at 150° C. for 1 h under microwave irradiation. The reaction mixture was allowed to cool to RT, diluted with aqueous ammonia (5 mL) and extracted using ethyl acetate (3×25 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography to get the title compound (250 mg, 48%). LCMS: 336 [M+1]+.
  • Step-4 Synthesis of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid: To a stirred solution of -amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)pyrazine-2-carbonitrile (250 mg, 0.68 mmol, 1.0 eq) in ethanol (5 mL) was added 6M NaOH solution (5 mL). The resulting reaction mixture was heated at 100° C. for 16 h. The reaction mixture was allowed to cool to RT. The solvent was evaporated under vacuum and acidified using 1N HCl to get the solid which was filtered and dried to get the title compound (100 mg, 38%) LCMS: 384 (M+1)+.
  • Step-5: (R)-3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To stirred solution of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid (100 mg, 0.26 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine (63 mg, 0.39 mmol, 1.5 eq), DIPEA (0.2 mL, 0.75 mmol, 3 eq) and HATU (190 mg, 0.50 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. Following this, ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the title compound (10 mg, 8%) LCMS: 488[M+1]+; 1H NMR (400 MHz, DMSO-d6) δ 13.66 (br s, 1H), 9.20 (d, 1H), 8.57 (d, 1H), 8.18 (s, 1H), 7.78-7.83 (m, 1H), 7.69 (s, 1H), 7.47-7.52 (m, 2H), 7.43 (dd, 2H), 7.31 (dd, 2H), 7.17 (t, 2H), 5.19-5.25 (m, 1H), 1.53 (d, 3H)
  • Example S-16: Synthesis of 5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)-3-(1-(pyridin-2-yl)ethoxy)pyrazin-2-amine (Compound No. 103)
  • Figure US20220298142A1-20220922-C00642
  • To a stirred solution of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(4-fluorophenyl)pyrazin-2-amine (0.1 g, 0.23 mmol, 1.0 eq) in DMF (5 ml) was added Cs2CO3 (224 mg, 0.69 mmol, 3 eq) and it was stirred at RT for 5 min followed by the addition of 1-(pyridin-2-yl)ethanol (72 mg, 0.69 mmol, 2.5 eq) at RT under inert condition. The resulting mixture was stirred for 16 h at 120° C. Following this, ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the title compound (20 mg, 18%). LCMS: 472 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ8.93 (d, 1H), 8.58 (d, 1H), 8.19 (d, 1H), 7.84 (t, 1H), 7.64 (d, 1H), 7.52-7.61 (m, 3H), 7.26-7.37 (m, 3H), 7.11 (t, 2H), 6.83 (br s, 2H), 6.20-6.25 (m, 1H), 1.70 (d, J=6.6 Hz, 3H).
  • Example S-17: Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-(4-fluorophenyl)pyrazine-2-carboxamide (Compound No. 102)
  • Figure US20220298142A1-20220922-C00643
  • To the stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid (0.1 g, 0.25 mmol, 1.0 eq) in DMF (10 ml) was added 6-(1-aminoethyl)pyridine-2-carbonitrile (60 mg, 0.38 mmol, 1.5 eq), DIPEA (0.2 mL, 0.87 mmol, 3 eq) and HATU (190 mg, 0.50 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. The reaction was monitored by TLC and LCMS. ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the title compound (20 mg, 15%) LCMS:524 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ9.16 (d, 1H), 8.99 (dd, 1H), 8.34 (dd, 1H), 8.04-8.09 (m, 1H), 7.93-8.00 (m, 3H), 7.85 (d, 1H), 7.62 (dd, 1H), 7.46 (dd, 2H), 7.18 (t, 2H), 5.26-5.33 (m, 1H), 1.59 (d, 3H).
  • Example S-18: Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)-N-(2-(tetrahydro-2H-pyran-4-yl)ethyl)pyrazine-2-carboxamide (Compound No. 101)
  • Figure US20220298142A1-20220922-C00644
  • To the stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-(4-fluorophenyl)pyrazine-2-carboxylic acid (0.1 g, 0.25 mmol, 1.0 eq) in DMF (10 ml) was added 2-(tetrahydro-2H-pyran-4-yl)ethanamine (49 mg, 0.38 mmol, 1.5 eq), DIPEA (0.2 mL, 0.87 mmol, 3 eq) and HATU (220 mg, 0.58 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. The reaction was monitored by TLC and LCMS. Ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the title compound (80 mg, 62%), LCMS:506 [M+1]+. 1H NMR (400 MHz, DMSO-d6) δ 8.99 (dd, 1H), 8.81 (t, 1H), 8.25-8.32 (m, 1H), 7.99 (d, 1H), 7.91 (d, 1H), 7.62 (dd, 1H), 7.46 (dd, 2H), 7.18 (t, 2H), 3.83 (d, 2H), 3.35-3.40 (m, 2H), 3.27 (t, 3H), 1.64 (d, 2H), 1.47-1.55 (m, 3H), 1.11-1.25 (m, 3H).
  • Example S-19: Synthesis of (R)-3-amino-6-(8-chloroquinolin-6-yl)-S-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 100)
  • Figure US20220298142A1-20220922-C00645
    Figure US20220298142A1-20220922-C00646
  • Step-1 Synthesis of 5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine: To a stirred solution of 6-chloro-5-(quinolin-6-yl)pyrazin-2-amine (1.0 g, 3.44 mmol, 1.0 eq) in dioxane:water (16 mL: 4 mL) was added 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (0.860 g, 4.12 mmol, 1.2 eq). The reaction mixture was purged with nitrogen for 5 min then charged with Na2CO3 (0.73 g, 6.88 mmol, 2.0 eq) and Pd(dppf)Cl2.DCM complex (0.080 g, 10 mol %). The reaction mixture was again purged with nitrogen. The reaction mixture was allowed to heat at 100° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum, to get the crude which was purified by normal phase silica-gel column chromatography to get the title compound (0.400 g, 34%). LCMS: 337[M+1]+.
  • Step-2 Synthesis of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine. To a stiffed solution 5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine (0.400 g, 1.18 mmol, 1 eq) in DMF (5 ml) was added NBS (210 mg, 1.18 mmol, 1.0 eq) at 0° C. Reaction mixture was stirred at 0° C. for 10 min. The reaction was monitored by TLC and LCMS and found to be complete after 10 min. The reaction mixture was quenched with cold water 10 mL and was extracted with EtOAc (3×20 mL). The resulting solution was concentrated under reduced pressure. The crude product was purified by normal phase column chromatography to get the title compound (0.300 g, 61%). LCMS: 415 [M+1]+.
  • Step-3 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carbonitrile: To a stirred solution of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine (500 mg, 1.20 mmol, 1.0 eq) in DMF (10 mL) was added cuprous cyanide (120 mg, 1.35 mmol, 1.1 eq). The reaction mixture was allowed to stir at 120° C. for 45 min under microwave irradiation. The reaction mixture was allowed to cool to RT, diluted with aqueous ammonia (5 mL) and extracted using ethyl acetate (3×25 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography to get the title compound (170 mg, 46%). LCMS: 362[M+1]+.
  • Step-4 Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxylic acid: To a stirred solution of 3-bromo-5-(8-chloroquinolin-6-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine (170 mg, 0.48 mmol, 1.0 eq) in ethanol (5 mL) was added 6M NaOH solution (5 mL). The resulting reaction mixture was heated at 100° C. for 16 h. The reaction mixture was allowed to cool to RT. The solvent was evaporated under vacuum and acidified using 1N HCl to get the solid which was filtered and dried to get the product as yellow solid (110 mg, 60%) LCMS: 381 [M+1]+.
  • Step-3 Synthesis of (R)-3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To a stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxylic acid (50 mg, 0.13 mmol, 1 eq) in DMF (4 mL) was added (1R)-1-(pyridin-2-yl)ethanamine (23 mg, 0.19 mmol, 1.2 eq), DIPEA (0.1 mL, 0.39 mmol, 3 eq) and HATU (95 mg, 0.26 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. The reaction was monitored by TLC and LCMS. Ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the desired product (0.005 g, 8%). LCMS: 485 [M+1]+; 1H NMR (400 MHz, D2O) S 9.17 (d, J=7.45 Hz, 1H), 9.02 (dd, J=1.32, 3.95 Hz, 1H), 8.60 (d, J=4.82 Hz, 1H), 8.38-8.49 (m, 1H), 8.07 (d, J=1.75 Hz, 1H), 8.00 (d, J=1.32 Hz, 1H), 7.93 (br s, 1H), 7.63-7.72 (m, 2H), 7.60 (d, J=7.89 Hz, 1H), 7.42 (br s, 1H), 6.35 (d, J=2.19 Hz, 1H), 5.17-5.32 (m, 1H), 3.64-3.76 (m, 3H), 1.56 (d, J=7.02 Hz, 3H).
  • Example S-20: Synthesis of 3-(6-amino-3-(7-chloro-1H-indazol-5-yl)pyrazin-2-yl)benzonitrile (Compound No. 2-7)
  • Figure US20220298142A1-20220922-C00647
  • To a stirred solution of 6-chloro-5-(7-chloro-1H-indazol-5-yl)pyrazin-2-amine (160 mg, 0.57 mmol, 1 eq) in dioxane:water (10 mL: 5 mL) was added 3-cyanobenzeneboronic acid (197 mg, 0.86 mmol, 1.5 eq). The reaction mixture was purged with nitrogen for 20 min then charged K2CO3 (157 mg 1.14 mmol, 2.0 eq) and Pd(dppf)Cl2.DCM complex (23 mg, 0.02 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 90° C. for 16 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the crude which was purified by reverse phase column chromatography to get the title compound (50 mg, 25%). LCMS: 347[M+1]+; 1H NMR (DMSO-d6 400 MHz): δ 13.60 (br s, 1H), 8.10 (s, 1H), 8.01 (s, 1H), 7.84 (s, 1H), 7.78 (d, 1H), 7.52-7.56 (m, 2H), 7.41-7.47 (m, 2H), 7.35 (s, 1H), 6.75 (s, 2H).
  • Example S-21: Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxamide (Compound No. 99)
  • Figure US20220298142A1-20220922-C00648
  • To a stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxylic acid (50 mg, 0.13 mmol, 1 eq) in DMF (4 mL) was added 6-(1-aminoethyl)picolinonitrile (27 mg, 0.19 mmol, 1.2 eq), DIPEA (0.1 mL, 0.39 mmol, 3 eq) and HATU (95 mg, 0.26 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. The reaction was monitored by TLC and LCMS. Ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer was washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled under vacuum to get the crude which was purified by reverse phase column chromatography to get the title compound (0.003 g, 5%). LCMS: 510 [M+1]+; 1H NMR (400 MHz, METHANOL-d4) 68.91-8.97 (m, 1H), 8.48 (d, J=8.33 Hz, 1H), 8.13 (s, 1H), 7.98 (t, J=7.89 Hz, 1H), 7.90 (d, J=1.75 Hz, 1H), 7.77 (t, J=7.67 Hz, 2H), 7.63 (dd, J=4.39, 8.33 Hz, 1H), 7.56 (d, J=2.19 Hz, 1H), 6.31 (d, J=2.19 Hz, 1H), 5.34 (dd, J=6.80, 13.81 Hz, 1H), 3.82 (s, 3H), 1.62 (d, J=7.02 Hz, 3H)
  • Example S-22: Synthesis of (R)-5-(3-cyanophenyl)-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 109)
  • Figure US20220298142A1-20220922-C00649
  • To a solution of 3-(6-amino-5-bromo-3-(quinolin-6-yl)pyrazin-2-yl)benzonitrile (200 mg 0.49 mmol, 1 eq) in toluene (10 mL) was added (R)-1-(pyridin-2-yl)ethanamine (72 mg, 0.59 mmol, 1.2 eq), TEA (0.210 mL, 1.49 mol, 3.0 eq) in autoclave. The reaction mixture was deoxygenated using N2 atmosphere for 10 min, then charged Palladium Acetate (6 mg, 0.025 mmol, 0.05 eq) and Xanthphos (28 mg, 0.049 mmol, 0.1 eq). The reaction mixture was heated at 90° C. for 16 h under carbon monoxide. The reaction was monitored by TLC and LCMS. The reaction mixture was filter through celite. The reaction mixture was diluted with water (50 mL) and extracted using ethyl acetate (3×50 mL). The separated organic layer was dried over sodium sulphate and concentrated under reduced pressure. The crude product was purified by normal phase silica-gel column chromatography followed by HPLC purification to get the title compound (4 mg, 2%). LCMS: 481 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ 10.06 (br s, 1H) 9.21 (d, J=7.89 Hz, 1H) 8.99 (br s, 1H) 8.61 (d, J=4.82 Hz, 1H) 8.45 (d, J=8.33 Hz, 1H) 8.09 (d, J=1.75 Hz, 1H) 8.01 (d, J=8.77 Hz, 1H) 7.91-7.94 (m, 1H) 7.81-7.87 (m, 2H) 7.57-7.69 (m, 3H) 7.37-7.51 (m, 2H) 5.26-5.36 (m, 1H) 1.58 (d, J=7.02 Hz, 3H).
  • Example S-23: Synthesis of (R)-3-amino-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 110)
  • Figure US20220298142A1-20220922-C00650
  • Step-1: Synthesis of 6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine. To a stirred solution of 6-chloro-5-(quinolin-6-yl)pyrazin-2-amine (500 mg, 1.95 mmol, 1 eq) in DMF:water (10 mL:5 mL) was added 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (487 mg, 2.34 mmol, 1.2 eq). The reaction mixture was purged with nitrogen for 5 min then charged with K3PO4 (1.2 g, 5.85 mmol, 3.0 eq) and Pd(PPh3)4 (90 mg, 0.07 mmol, 0.04 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 120° C. for 1 h using microwave irradiation. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the crude which was purified by column chromatography to get the title compound (226 mg, 34%). LCMS: 303 [M+1]+.
  • Step-2: Synthesis of 3-bromo-6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine: To a stirred solution 6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine (226 mg, 0.74 mmol, 1 eq) in DMF (10 ml) was added NBS (133 mg, 0.61 mmol, 1.0 eq) at 0° C. Reaction mixture was stiffed at 0° C. for 10 min. The reaction was monitored by TLC and LCMS and found to be complete after 10 min. The reaction mixture was quenched with cold water 10 mL and was extracted with EtOAc (3×20 mL). The resulting solution was concentrated under reduced pressure. The crude product was purified by normal phase silica gel column chromatography to get the title compound. (189 mg, 66%). LCMS: 381[M+1]+.
  • Step-3 Synthesis of (R)-3-amino-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide: To a stirred solution of 3-bromo-6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine (189 mg, 0.49 mmol, 1 eq) in DMF (10 mL) was added (R)-1-(pyridin-2-yl)ethanamine (121 mg, 0.99 mmol, 1.2 eq). The reaction mixture was purged with nitrogen for 5 min then charged with Mo(CO)6 (48 mg, 0.18 mmol, 0.37 eq) and PdCl2dppf (18 mg, 0.02 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen for 5 min and then stirred at RT for 1 h followed by the addition of DBU (0.2 mL, 1.07 mmol, 2.2 eq). The reaction mixture was stirred at RT for 5 min and then heated at 120° C. for 3 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the crude which was purified by reverse phase column chromatography to get the title compound (30 mg, 9.0%). LCMS: 451[M+1]+; 1H NMR (DMSO-d6, 400 MHz) δ 9.08 (d, 1H), 8.90 (d, 1H), 8.53 (d, 1H), 8.35 (d, 1H), 8.07-8.13 (m, 1H), 7.95 (d, 1H), 7.70-7.83 (m, 3H), 7.64 (d, 2H), 7.54 (dd, 1H), 7.47 (d, 1H), 7.29 (dd, 1H), 6.19 (d, 1H), 5.22 (d, 1H), 3.71 (s, 3H) 1.52 (d, 3H).
  • Example S-24: Synthesis of 3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 111), (R)-3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 115) and (S)-3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-S-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 116)
  • Figure US20220298142A1-20220922-C00651
  • To the stirred solution 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.1 g, 0.29 mmol, 1.0 eq) in DMF (10 ml) was added 6-(1-aminoethyl)pyridine-2-carbonitrile (86 mg, 0.58 mmol, 2.0 eq), DIPEA (0.2 mL, 0.87 mmol, 3 eq) and HATU (220 mg, 0.58 mmol, 2 eq) at RT under inert condition. The resulting mixture stiffed for 16 h at same temperature. The reaction was monitored by TLC and LCMS. Ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the title compound (4 mg, 3.0%). The title compound was purified by chiral HPLC to get the two enantiomers (R)-3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide and (S)-3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxamide. LCMS:472 [M+1]+; 1H NMR (DMSO-d6, 400 MHz) δ 9.20 (d, 1H), 8.87 (s., 1H), 8.23 (d, 1H), 8.04-8.12 (m, 1H), 7.94-8.00 (m, 1H), 7.87 (d, 2H), 7.91 (d, 2H), 7.77 (d, 1H), 7.50 (dd, 2H), 7.41 (d, 1H), 7.32 (d, 1H), 7.36 (d, 1H), 5.26-5.32 (m, 1H), 1.57 (d, 3H).
  • Example S-25: Synthesis of (R)-3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 112)
  • Figure US20220298142A1-20220922-C00652
  • Step-1: Synthesis of 5-(7-chloro-1H-indazol-5-yl)-6-4-fluorophenyl)pyrazin-2-amine. To a stirred solution of 6-chloro-5-(7-chloro-1H-indazol-5-yl)pyrazin-2-amine (500 mg, 1.79 mmol, 1 eq) in DMF:water (10 mL: 5 mL) was added 1-methyl-3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-1H-pyrazole (447 mg, 2.15 mmol, 1.2 eq). The reaction mixture was purged with nitrogen for 5 min then charged with K3PO4 (1.2 g, 5.37 mmol, 3.0 eq) and Pd(PPh3)4 (82 mg, 0.07 mmol, 0.04 eq). The reaction mixture was again purged with nitrogen. The reaction mixture was stirred at RT for 10 min followed by heating at 120° C. for 1 h using microwave irradiation. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and concentrated under vacuum. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the crude which was purified by column chromatography to get the title compound (200 mg, 34%). LCMS: 326 [M+1]+.
  • Step-2: Synthesis of 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine. To a stirred solution of 5-(7-chloro-1H-indazol-5-yl)-6-(4-fluorophenyl)pyrazin-2-amine (200 mg, 0.61 mmol, 1 eq) in DMF (10 ml) was added NBS (109 mg, 0.61 mmol, 1.0 eq) at 0° C. Reaction mixture was stirred at 0° C. for 10 min. The reaction was monitored by TLC and LCMS and found to be complete after 10 min. The reaction mixture was quenched with cold water (10 mL) and was extracted with EtOAc (3×20 mL). The resulting solution was concentrated under reduced pressure. The crude product was purified by normal phase silica-gel column chromatography to get the title compound. (138 mg, 55%). LCMS: 404 [M+1]+.
  • Step-3: Synthesis of (R)-3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To a stirred solution of 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine (138 mg, 0.34 mmol, 1 eq) in DMF (10 mL) was added (1R)-1-(pyridin-2-yl)ethanamine (83 mg, 2.15 mmol, 1.2 eq). The reaction mixture was purged with nitrogen for 5 min then charged with Mo(CO)6 (33 mg, 0.12 mmol, 0.37 eq) and PdCl2dppf (12 mg, 0.01 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen for 5 min and then stirred at RT for 1 h followed by the addition of DBU (0.1 mL, 0.74 mmol, 2.2 eq). The reaction mixture was stirred at RT for 5 min and then heated at 120° C. for 3 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the crude which was purified by reverse phase column chromatography to get the title compound (20 mg, 12%). LCMS: 474 [M+1]+; 1H NMR (DMSO-d6, 400 MHz): δ13.63 (br s, 1H), 9.10 (d, 1H), 8.55 (d, 1H), 8.21 (s, 1H), 7.75-7.83 (m, 2H), 7.64 (d, 2H), 7.46-7.54 (m, 2H), 7.24-7.35 (m, 1H), 6.14 (d, 1H), 5.16-5.24 (m, 1H), 3.74 (s, 3H), 1.51 (d, 3H).
  • Example S-26: Synthesis of (R)-3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 113)
  • Figure US20220298142A1-20220922-C00653
  • Step-1: Synthesis of 3-bromo-5-(8-chloroquinolin-6-yl)-6-phenylpyrazin-2-amine: To a solution of 5-(8-chloroquinolin-6-yl)-6-phenylpyrazin-2-amine (1.6 g, 4.81 mmol, 1 eq) in DMF (20 mL) was added N-bromosuccinimide (0.85 g, 4.81 mmol, 1 eq) at 0° C. The reaction mixture was stirred at same temperature for 2 h. The reaction was monitored by TLC. The reaction was added with water and the solid precipitates out. The solid was filtered and dried to use for next step without further purification (1.1 g, 55%). LCMS: 412[M+1]+
  • Step-2: Synthesis of (R)-3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide: To a solution of 3-bromo-5-(8-chloroquinolin-6-yl)-6-phenylpyrazin-2-amine (200 mg, 0.48 mmol, 1 eq) in DMF (10 mL) was added (R)-6-(1-aminoethyl)picolinonitrile hydrochloride (92 mg, 0.58 mmol, 1.2 eq) The reaction mixture was deoxygenated using N2 atmosphere then charged MO(CO)6 (51.3 mg, 0.19 mmol, 0.4 eq) and PdCl2(dppf).DCM complex (20 mg, 0.024 mmol, 0.05 eq), deoxygenating was continued for further 20 min. and DBU (0.165 mL, 1.07 mmol, 2.2 eq) was charged and the reaction mixture was heated at 120° C. for 4 h. The reaction was monitored by TLC and LCMS. The reaction mixture was diluted with water (50 mL) and extracted using ethyl acetate (3×100 mL). The separated organic layer was dried over sodium sulfate and concentrated under reduced pressure. The crude product was purified by normal phase silica-gel column chromatography followed by HPLC purification to afford (40 mg, 17%) the title compound. LCMS: 481 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ 9.28 (d, J=7.9 Hz, 1H), 8.99 (d, J=3.9 Hz, 1H), 8.58 (d, J=3.5 Hz, 1H), 8.28 (d, J=7.0 Hz, 1H), 7.98 (d, J=1.8 Hz, 1H), 7.90 (d, J=1.8 Hz, 1H), 7.81 (dd, J=5.7, 7.5 Hz, 2H), 7.62 (dd, J=4.4, 8.3 Hz, 1H), 7.51 (d, J=7.9 Hz, 1H), 7.46-7.31 (m, 7H), 5.26-5.21 (m, 1H), 1.55 (d, J=6.6 Hz, 3H).
  • Example S-27: Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide (Compound No. 114), (R)-3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide (Compound No. 121) and (S)-3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide (Compound No. 122)
  • Figure US20220298142A1-20220922-C00654
  • Step-1: Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2-carbonitrile: To a stirred solution of CuI (554 mg, 2.92 mmol, 1.5 eq) and CuCN (384 mg, 4.28 mol, 2.2 eq) in dry DMF (150 mL) was added 3-bromo-5-(8-chloroquinolin-6-yl)-6-phenylpyrazin-2-amine (800 mg, 1.94 mmol, 1 eq) at 100° C. The reaction mixture was stirred at 120° C. for 12 h. The reaction was monitored by TLC and LCMS. The crude product was poured in ice-water the solid precipitate out. The reaction mixture PH was adjusted with aqueous ammonia until pH=9 then extracted with ethyl acetate (3×50 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography using basic Alumina [Ethyl acetate:Hexane (5:5)] to get the title compound (400 mg, 57%). LCMS: 358 [M+1]+
  • Step-2: Synthesis 3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2-carboxylic acid: To a solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2-carbonitrile (700 mg, 1.96 mmol, 1.0 eq) in ethanol (10 mL) was added aqeuous NaOH (6M, 10 mL) at 0° C. The reaction mixture was stirred at 120° C. The reaction was monitored by TLC. The reaction was distilled then charged ice water then acidifies with dilute HCl solid precipitates out. The solid was filtered and dried to use it for next step without further purification (610 g, 82%). LCMS: 377 [M+1]+.
  • Step-3: Synthesis of (R)-3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide: To a stirred solution of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenylpyrazine-2-carboxylic acid (200 mg, 0.53 mmol, 1.0 eq) in DMF (10 mL) was added 6-(1-aminoethyl)picolinonitrile (94 mg, 0.63 mmol, 1.2 eq) and the mixture was stirred at RT for 5 min. To this mixture HATU (404 mg, 1.06 mmol, 2.0 eq) and DIPEA (0.260 mL, 1.60 mmol, 3.0 eq) was added and the resultant mixture was allowed to stir for 16 h. The progress of reaction was monitored by TLC. Upon completion, the mixture was diluted with water (40 mL), extracted with EtOAc (2×100 mL). The combined organic layers were washed with water (40 mL), brine (40 mL), dried over Na2SO4, filtered and concentrated under reduced pressure to afford a crude residue which was purified by reverse phase column chromatography to afford the title compound (100 mg, 37%). The title compound was purified by chiral HPLC to get the two enantiomers (R)-3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide (4 mg) and (S)-3-amino-6-(8-chloroquinolin-6-yl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-phenylpyrazine-2-carboxamide (7 mg). LCMS: 506 [M+1]+; 1H NMR (400 MHz, DMSO-d6) δ 9.17 (d, J=7.89 Hz, 1H), 8.96-9.02 (m, 1H), 8.31 (d, J=8.33 Hz, 1H), 8.05 (d, J=7.89 Hz, 1H), 7.93-8.02 (m, 2H), 7.90 (d, J=1.75 Hz, 1H), 7.85 (d, J=8.33 Hz, 1H), 7.61 (dd, J=3.95, 8.33 Hz, 1H), 7.28-7.46 (m, 5H), 5.29 (s, 1H), 1.59 (d, J=7.02 Hz, 3H)
  • Example S-28: Synthesis of (R)-3-amino-5-(3-cyanophenyl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 117) and (S)-3-amino-5-(3-cyanophenyl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 118)
  • Figure US20220298142A1-20220922-C00655
  • Step-1: Synthesis of methyl 3-amino-5-(3-cyanophenyl)-6-(quinolin-6-yl)pyrazine-2-carboxylate: To a stirred solution of 3-[6-amino-5-bromo-3-(quinolin-6-yl)pyrazin-2-yl]benzonitrile (700 mg, 1.74 mmol, 1.0 eq) in DMF (5 mL) and MeOH (10 mL). The reaction mixture was purged with nitrogen for 5 min then charged with Mo(CO)6 (170 mg, 0.64 mmol, 0.37 eq) and PdCl2dppf (63 mg, 0.08 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen for 5 min and then stirred at RT for 1 h followed by the addition of DBU (0.6 mL, 3.82 mmol, 2.2 eq). The reaction mixture was stirred at RT for 5 min and then heated at 120° C. for 3 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled to get the crude which was purified by column chromatography to get the desired product (200 mg, 22%). LCMS: 382 [M+1].+
  • Step-2: Synthesis of 3-amino-5-(3-cyanophenyl)-6-(quinolin-6-yl)pyrazine-2-carboxylic acid. To a stirred solution of methyl 3-amino-5-(3-cyanophenyl)-6-(quinolin-6-yl)pyrazine-2-carboxylate (200 mg, 0.52 mmol, 1.0 eq) in ethanol (5 mL) and THF (5 mL) was added LiOH.H2O (88 mg, 2.09 mmol, 4.0 eq) dissolved in H2O (2 mL). The resulting reaction mixture was stirred at RT for 16 h. The reaction mixture was allowed to cool to RT. The solvent was evaporated under vacuum and acidified using 1N HCl to get the solid which was filtered and dried to get the product (100 mg, 52%) LCMS: 368 [M+1]+.
  • Step-3: Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide: To stirred solution of of 3-amino-5-(3-cyanophenyl)-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (100 mg, 0.27 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine (80 mg, 0.54 mmol, 2.0 eq), DIPEA (0.2 mL, 0.81 mmol, 3 eq) and HATU (205 mg, 0.54 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. Following this, ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the title compound (50 mg) which was further purified by chiral purification to get the two enantiomers (R)-3-amino-5-(3-cyanophenyl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (10 mg, 7.0%) and (S)-3-amino-5-(3-cyanophenyl)-N-(1-(6-cyanopyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (16 mg, 11.0%). LCMS: 497 [M+1]+; 1H NMR (DMSO-d6, 400 MHz): δ 9.25 (d, 1H), 8.89 (d, 1H), 8.27 (d, 1H), 8.05-8.10 (m, 1H), 7.96-8.02 (m, 4H), 7.88-7.94 (m, 5H), 7.62 (d, 1H), 7.46-7.54 (m, 2H), 5.27-5.33 (m, 1H), 1.58 (d, 3H).
  • Example S-29: Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 119), (R)-3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 123) and (S)-3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide (Compound No. 124)
  • Figure US20220298142A1-20220922-C00656
  • Step-1: Synthesis of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carbonitrile). To a stirred solution of 3-bromo-5-(7-chloro-1H-indazol-5-yl)-6-(1-methyl-1H-pyrazol-3-yl)pyrazin-2-amine (500 mg, 1.24 mmol, 1.0 eq) in DMF (10 mL) was added cuprous cyanide (122 mg, 1.36 mmol, 1.1 eq) and copper iodide (354 mg, 1.86 mmol, 1.5 eq). The reaction mixture was allowed to stir at 120° C. for 45 mins under microwave irradiation. The reaction mixture was allowed to cool to RT, diluted with aqueous ammonia (5 mL) and extracted using ethyl acetate (3×25 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the solid which was purified by column chromatography to get the desired product. (170 mg, 39%). LCMS: 351 [M+1]+.
  • Step-2: Synthesis of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxylic acid FK-GRF-633-84) To a stirred solution of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carbonitrile (170 mg, 0.48 mmol, 1.0 eq) in ethanol (5 mL) was added 6M NaOH solution (5 mL). The resulting reaction mixture was heated at 100° C. for 16 h. The reaction mixture was allowed to cool to RT. The solvent was evaporated under vacuum and acidified using 1N HCl to get the solid which was filtered and dried to get the product as yellow solid (120 mg, 67%). LCMS: 370 [M+1]+.
  • Step-3: Synthesis of 3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide; To stirred solution of 3-amino-6-(7-chloro-1H-indazol-5-yl)-5-(1-methyl-1H-pyrazol-3-yl)pyrazine-2-carboxylic acid (120 mg, 0.33 mmol, 1.0 eq) in DMF (10 ml) was added (1R)-1-(pyridin-2-yl)ethanamine (96 mg, 0.65 mmol, 2.0 eq), DIPEA (0.2 mL, 0.97 mmol, 3 eq) and HATU (247 mg, 0.65 mmol, 2 eq) at RT under inert condition. The resulting mixture stiffed for 16 h at the same temperature. Following this, ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the title compound (26 mg, 21%). The title compound was purified by chiral HPLC to get the two enantiomers (R)-3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide and (S)-3-amino-6-(8-chloroquinolin-6-yl)-5-phenyl-N-(1-(pyridin-2-yl)ethyl)pyrazine-2-carboxamide LCMS: 49 [M+1]+; 1H NMR (DMSO-d6, 400 MHz): δ 9.04 (d, 1H), 8.21 (s, 1H), 8.02-8.08 (m, 2H), 7.95 (d, 1H), 7.81-7.87 (m, 2H), 7.63 (d, 2H), 7.47 (s, 2H), 6.13 (d, 1H), 5.23-5.28 (m, 1H), 3.74 (s, 3H), 1.55 (d, 3H).
  • Example S-30: Synthesis of (3-amino-5-phenyl-N-(1-(pyrimidin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 120), (R)-3-amino-5-phenyl-N-(1-(pyrimidin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 125) and (S)-3-amino-5-phenyl-N-(1-(pyrimidin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 126)
  • Figure US20220298142A1-20220922-C00657
  • To the stirred solution 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.2 g, 0.58 mmol, 1.0 eq) in DMF (10 ml) was added 1-(pyrimidin-2-yl)ethanamine (143 mg, 1.16 mmol, 2.0 eq), DIPEA (0.3 mL, 1.74 mmol, 3 eq) and HATU (440 mg, 1.16 mmol, 2 eq) at RT under inert condition. The resulting mixture stiffed for 16 h at same temperature. The reaction was monitored by TLC and LCMS. Ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the desired product (26 mg, 10%) LCMS: 448 [M+1]+; 1H NMR (DMSO-d6, 400 MHz) δ 9.16 (d, 1H), 8.87-8.90 (m, 1H), 8.83 (d, 2H), 8.26 (d, 1H), 8.02 (d, 1H), 7.91-7.52 (m, 4H), 7.39-7.46 (m, 3H), 7.28-7.37 (m, 3H), 5.24 (dt, 1H), 1.60 (d, 3H).
  • Example S-31: Synthesis of 3-amino-N-(1-(6-cyanopyridin-2-yl)ethyl)-5-(1-methyl-1H-pyrazol-3-yl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 127)
  • Figure US20220298142A1-20220922-C00658
  • To the stirred solution 3-amino-5-(1-methyl-1H-pyrazol-3-yl)-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.2 g, 0.57 mmol, 1.0 eq) in DMF (10 ml) was added 6-(1-aminoethyl)pyridine-2-carbonitrile (170 mg, 1.15 mmol, 2.0 eq), DIPEA (0.3 mL, 1.71 mmol, 3 eq) and HATU (433 mg, 1.15 mmol, 2 eq) at RT under inert condition. The resulting mixture stirred for 16 h at same temperature. The reaction was monitored by TLC and LCMS. Ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the title compound (7 mg, 3%). LCMS: 476 [M+1]+. 1H NMR (DMSO-d6, 400 MHz): δ 9.10 (d, 1H), 8.97 (br s, 1H), 8.50 (br s, 1H), 8.18 (br s, 1H), 8.04-8.10 (m, 1H), 8.00 (d, 2H), 7.96 (d, 2H), 7.65-7.93 (m, 4H), 6.26 (br s, 1H), 5.20-5.29 (m, 1H), 3.71 (s, 3H), 1.55 (d, 0.3H).
  • Example S-32: Synthesis of (R)-3-amino-5-phenyl-N-(1-(pyrazin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 128) and (S)-3-amino-5-phenyl-N-(1-(pyrazin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 129)
  • Figure US20220298142A1-20220922-C00659
  • To the stirred solution 3-amino-5-phenyl-6-(quinolin-6-yl)pyrazine-2-carboxylic acid (0.2 g, 0.58 mmol, 1.0 eq) in DMF (10 mL) was added 1-(pyrazin-2-yl)ethanamine (143 mg, 1.16 mmol, 2.0 eq), DIPEA (0.3 mL, 1.74 mmol, 3 eq) and HATU (440 mg, 1.16 mmol, 2 eq) at RT under inert condition. The resulting mixture stiffed for 16 h at same temperature. The reaction was monitored by TLC and LCMS. Ice cold water (20 mL) was added and extracted with ethyl acetate (3×20 mL), the combined organic layer washed with brine solution (1×50 mL), dried over Na2SO4, filtered and distilled purified by reverse phase column chromatography to get the desired product (70 mg, 26%) the desired product obtained was further purified by chiral column to obtain the desired enantiomers (R)-3-amino-5-phenyl-N-(1-(pyrazin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide and (S)-3-amino-5-phenyl-N-(1-(pyrazin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide; LCMS:448 [M+1]+; 1H NMR (DMSO-d6, 400 MHz): δ 9.07 (s, 1H), 8.80 (d, 1H), 8.61-8.63 (m, 1H), 8.57 (d, 1H), 8.05 (s, 1H), 7.92 (d, 1H), 7.73-7.78 (m, 1H), 7.41 (s, 1H), 7.26-7.40 (m, 4H), 4.94-5.62 (m, 1H), 1.60 (d, 3H).
  • Example S-33: Synthesis of 3-amino-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyrimidin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 460)
  • Figure US20220298142A1-20220922-C00660
  • To a stirred solution of 3-bromo-6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine (200 mg, 0.52 mmol, 1.0 eq) in DMF (10 mL) was 1-(pyrimidin-2-yl)ethanamine (77 mg, 0.62 mmol, 1.2 eq). The reaction mixture was purged with nitrogen for 5 min then charged with Mo(CO)6 (51 mg, 0.19 mmol, 0.37 eq) and PdCl2dppf (19 mg, 0.02 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen for 5 min and then stirred at RT for 1 h followed by the addition of DBU (0.2 mL, 1.04 mmol, 2.2 eq). The reaction mixture was stirred at RT for 5 min and then heated at 120° C. for 3 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×200 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the crude which was purified by reverse phase column chromatography to get the title compound (10 mg, 4.0%). LCMS: 45 2[M+1]+; 1H NMR (DMSO-d6, 400 MHz) δ 8.99-9.09 (m, 2H), 8.81 (d, J=4.82 Hz, 2H), 8.62 (br s, 2H), 8.24 (br s, 1H), 8.03 (d, J=8.77 Hz, 1H), 7.85 (d, J=8.33 Hz, 1H), 7.61-7.76 (m, 3H), 7.43 (t, J=5.04 Hz, 1H), 6.27 (d, J=2.19 Hz, 1H), 5.17-5.26 (m, 1H), 3.70 (s, 3H), 1.58 (d, J=6.58 Hz, 3H).
  • Example S-34: Synthesis of 3-amino-5-(1-methyl-1H-pyrazol-3-yl)-N-(1-(pyridin-2-yl)ethyl)-6-(quinolin-6-yl)pyrazine-2-carboxamide (Compound No. 459)
  • Figure US20220298142A1-20220922-C00661
  • To a stirred solution of 3-bromo-6-(1-methyl-1H-pyrazol-3-yl)-5-(quinolin-6-yl)pyrazin-2-amine (200 mg, 0.52 mmol, 1 eq) in DMF (8 mL) was added 1-(pyridin-2-yl)ethanamine (129.4 mg, 1.052 mmol, 2.0 eq). The reaction mixture was purged with nitrogen for 5 min then charged with Mo(CO)6 (55 mg, 0.21 mmol, 0.4 eq) and PdCl2dppf (20 mg, 0.02 mmol, 0.05 eq). The reaction mixture was again purged with nitrogen for 5 min and then stirred at RT for 1 h followed by the addition of DBU (0.2 mL, 1.15 mmol, 2.2 eq). The reaction mixture was stirred at RT for 5 min and then heated at 120° C. for 4 h. The reaction was monitored by TLC and LCMS. The reaction mixture was filtered through celite and distilled. The reaction was diluted with water and extracted with ethyl acetate (3×100 mL). The combined organic layers were washed (brine), dried (anhydrous Na2SO4) and concentrated under vacuum to get the crude which was purified by reverse phase column chromatography to get the title compound (7 mg, 3%). LCMS: 451[M+1]+; 1H NMR (DMSO-d6, 400 MHz) δ 9.08 (d, 1H), 8.90 (d, 1H), 8.53 (d, 1H), 8.35 (d, 1H), 8.07-8.13 (m, 1H), 7.95 (d, 1H), 7.70-7.83 (m, 3H), 7.64 (d, 2H), 7.54 (dd, 1H), 7.47 (d, 1H), 7.29 (dd, 1H), 6.19 (d, 1H), 5.22 (d, 1H), 3.71 (s, 3H), 1.52 (d, 3H).
  • It is understood that compounds from the Tables (2-72, 74, 77-88, 90-97, 130-458, 461, 462, 2-1, 2-3, 2-4, 2-5, 2-8, 2-9, 2-11, 2-12) are synthesized using the General Synthetic Schemes 1 to 3 or using the experimental procedures as described above and the steps involved in the synthetic routes are clearly familiar to those skilled in the art, wherein the substituents described in compounds of Formula (I), (II) and (III) herein can be varied with a choice of appropriate starting materials and reagents utilized in the steps presented.
  • BIOLOGICAL EXAMPLES Example B1. Radioligand Binding Competition Assay Example B1(a)
  • Binding of selected compounds to the adenosine A2A, A1, A2B, and A3 receptors is tested using a binding competition assay.
  • The general protocol for the radioligand binding competition assay is as follows. Competition binding is performed in duplicate in the wells of a 96 well plate (Master Block, Greiner, 786201) containing binding buffer (optimized for each receptor), membrane extracts (amount of protein/well optimized for each receptor), radiotracer (final concentration optimized for each receptor), and test compound. Nonspecific binding is determined by co-incubation with 200-fold excess of cold competitor. The samples are incubated in a final volume of 0.1 mL at 25° C. for 60 minutes and then filtered over filter plates. Filters are washed six times with 0.5 mL of ice-cold washing buffer (optimized for each receptor) and 50 μL of Microscint 20 (Packard) are added on each filter. The filter plates are sealed, incubated 15 min on an orbital shaker and scintillation counted with a TopCount for 30 sec/filter.
  • For the A2A adenosine receptor radioligand binding assay, the following modifications are made to the general protocol. GF/C filters (Perkin Elmer, 6005174), presoaked in 0.01% Brij for 2 h at room temperature are used. Filters are washed six times with 0.5 mL of ice-cold washing buffer (50 mM Tris pH 7.4) and 50 μL of Microscint 20 (Packard) are added in each well. The plates are then incubated for 15 min on an orbital shaker and then counted with a TopCount™ for 1 min/well. Another radioligand binding assay used to evaluate the binding affinity for the adenosine A2A receptor assay is performed in duplicate in the wells of a 384 plate. Assay buffer contains DPBS 500 mM, MgCl2 0.1 mM, and 1% DMSO. Membrane-bead suspension is prepared by mixing 25.98 μL of human adenosine A2A membrane preparation (Perkin Elmer, RBHA2AM400UA) at 33.4 μg/mL, 28 μL of ADA at 20 μg/mL, and 932 μL of SPA beads at 3.33 mg/mL) and the mixture is incubated for 20 min at room temperature. 20 μL of radiotracer (3H—SCH 58261) at 15 nM is mixed into each well containing test articles at various concentrations and the plate is centrifuged at 1000 rpm for 1 minute. 30 μL of the membrane-bead suspension is added to each well. The plates are sealed and incubated for 1 hr at room temperature with vigorous mixing on a plate mixer. Plates are read on Microbeta2 (Perkin Elmer, 2450-0010).
  • For the adenosine A1 radioligand binding competition assay, a similar procedure is used except that the following reagents are used: CHO-K1-A1 cell membranes; binding buffer comprising HEPES 25 mM pH 7.4, MgCl2 5 mM, CaCl2) 1 mM, NaCl 100 mM, saponin 10 μg/mL; wash buffer comprising HEPES 25 mM pH 7.4, MgCl2 5 mM, CaCl2) 1 mM, NaCl 100 mM; a Unifilter GF/B—treated for 2 h with 0.5% PEI; and 1.6 nM of 3H-DPCPX as the tracer.
  • Similarly, the following reagents are used for the adenosine A2B radioligand binding competition assay: HEK-293-A2B cell membranes, 20 μg/well, preincubated 30 min at RT with 25 μg/mL Adenosine Deaminase; a binding buffer comprising HEPES 10 mM pH 7.4, EDTA 1 mM, 0.5% BSA; a wash buffer comprising HEPES 10 mM pH 7.4, EDTA 1 mM; a Unifilter GF/C—treated for 2 h with 0.5% PEI; and 10 nM 3H-DPCPX as the tracer.
  • For the adenosine A3 radioligand binding competition assay, the following reagents are used: CHO-K1-A3 cell membranes, 1.5 μg/well; a binding buffer comprising HEPES 25 mM pH 7.4, MgC2 5 mM, CaCl2) 1 mM, 0.5% BSA; a wash buffer comprising HEPES 25 mM pH 7.4, MgCl2 5 mM, CaCl2 1 mM; a Unifilter GF/C—treated for 2 h with 0.5% BS; and 0.4 nM of 125I-AB-MECA as the tracer.
  • The results of the binding assay are given as percent residual binding at a given concentration. Percent of residual binding means binding of a compound in the presence of competitor normalized to the amount of binding in the absence of competitor.
  • Example B1(b)
  • A second A2A adenosine receptor radioligand binding assay protocol was used. The protocol used adenosine A2a (human) membrane (PerkinElmer RBHA2AM400UA) at a concentration of 5 μg/well/100 μl and the radioligand [3H] CGS-21680 (Cat No. PerkinElmer-NET1021250UC) at a final concentration of 6 nM. Testing compounds were diluted with DMSO to make 8-point 4-fold serial dilution, starting at 0.2 mM. CGS-15943 was the reference compound. 1 μl of compounds/high control/low control was transferred to the assay plate according to a plate map, followed by 100 μl of membrane stocks and 100 μl of radioligand, in assay buffer (50 mM Tris-HCl, 10 mM MgCl2, 1 mM EDTA, pH 7.4). The plate was sealed and incubated at RT for 2 hours. Unifilter-96 GF/C filter plates (Perkin Elmer Cat #6005174) were soaked with 50 μl of 0.3% PEI per well for at least 0.5 hour at room temperature. When the binding assays were completed, the reaction mixtures were filtered through GF/C plates using Perkin Elmer Filtermate Harvester, and each plate washed 4 times with cold wash buffer (50 mM Tris-HCl, 154 mM NaCl, pH 7.4). The filter plates were dried for 1 hour at 50 degrees. After drying, the bottom of the filter plate wells was sealed, 50 μl of Perkin Elmer Microscint 20 cocktail was added, and the top of the filter plate was sealed. 3H trapped on the filter was counted using Perkin Elmer MicroBeta2 Reader. The data were analyzed with GraphPad Prism 5 to obtain binding IC50 values. The “Inhibition [% Control]” was calculated using the equation: % Inh=(1−Background subtracted Assay value/Background subtracted HC value)*100, where HC is high control. A2a binding IC50 values are shown in Table B1.
  • A second A1 adenosine receptor radioligand binding assay protocol was used. The protocol used adenosine A1 (human) membrane (PerkinElmer ES-010-M400UA) at a concentration of 2.5 μg/well/100 μl and the radioligand [3H] DPCPX (Cat No. PerkinElmer-NET974250UC) at a final concentration of 1 nM. Testing compounds were tested at a final concentration of 200 nM. CGS-15943, the reference compound, was tested in an 8-point 4-fold serial dilution, starting at a top concentration of 1 μM. 1 μl of compounds/high control/low control was transferred to the assay plate according to a plate map, followed by 100 μl of membrane stocks and 100 μl of radioligand, in assay buffer (25 mM HEPES, 5 mM MgCl2, 1 mM CaCl2), 100 mM NaCl, pH 7.4). The plate was sealed and incubated at RT for 1 hour. Unifilter-96 GF/C filter plates (Perkin Elmer Cat #6005174) were soaked with 50 μl of 0.3% PEI per well for at least 0.5 hour at room temperature. When the binding assays were completed, the reaction mixtures were filtered through GF/C plates using Perkin Elmer Filtermate Harvester, and each plate washed 4 times with cold wash buffer (25 mM HEPES, 5 mM MgCl2, 1 mM CaCl2), 100 mM NaCl, pH 7.4). The filter plates were dried for 1 hour at 50 degrees. After drying, the bottom of the filter plate wells was sealed, 50 μl of Perkin Elmer Microscint 20 cocktail was added, and the top of the filter plate was sealed. 3H trapped on the filter was counted using Perkin Elmer MicroBeta2 Reader. The data were analyzed with GraphPad Prism 5 to obtain binding IC50 values for the reference compound. The “Inhibition [% Control]” was calculated using the equation: % Inh=(1−Background subtracted Assay value/Background subtracted HC value)*100, where HC is high control. A1 binding inhibition values are shown in Table B1.
  • TABLE B1
    A2a binding A1 binding
    Compound No. IC50 (nM) inhibition at 200 nM (%)
    73 3.1 61
    75 2.5 63
    76 3.4 45
    89 16 ND
    98 0.8 92
    2-2 ND 30
    2-6 3.7 90
    2-7 ND 81
    2-10 3.1 79
    2-13 ND 54
    100 1.6 70
    101 ND 66
    102 1.9 93
    103 ND 19
    104 0.9 68
    105 1.2 94
    106 ND 34
    107 ND 49
    108 1.3 47
    109 3.8 49
    110 2.9 38
    111 3.9 92
    112 1.6 77
    113 4.7 58
    114 3.5 95
    115 64 74
    116 5.4 98
    117 269 56
    118 16 92
    119 12 92
    120 6.8 45
    121 1.1 80
    122 <0.6 96
    123 1.1 89
    124 3.7 95
    125 1.2 ND
    126 1.8 41
    127 3.9 84
    128 2 92
    129 3.9 61
    459 4.7 ND
    460 7.3 ND
    ND: Not Determined
  • Example B2. cAMP Assay
  • The functional activity of compounds was tested using Assay 2, below, to detect the presence of cAMP. Assay 1 is an alternative assay for this purpose. Activation of G-protein coupled receptors (such as A2A) results in activation of adenylyl cyclase which converts ATP into cAMP which is used as a downstream signaling molecule. Therefore, molecules which act as GPCR (or specifically A2A receptor) antagonists cause a decrease in intracellular cAMP concentration.
  • Assay 1: This assay uses HEK-293 cells expressing human recombinant adenosine A2A receptor that are grown prior to the test in media without antibiotic. The cells are detached by gentle flushing with PBS-EDTA (5 mM EDTA), recovered by centrifugation and suspended in assay buffer (KRH: 5 mM KCl, 1.25 mM MgSO4, 124 mM NaCl, 25 mM HEPES, 13.3 mM Glucose, 1.25 mM KH2PO4, 1.45 mM CaCl2), 0.5 g/L BSA, supplemented with Rolipram).
  • 12 μL of cells are mixed with 6 μL of the test compound at increasing concentrations and then incubated for 10 min. Thereafter 6 μL of the reference agonist is added at a final concentration corresponding to the historical EC50. The plates are then incubated for 30 min at room temperature. After addition of the lysis buffer and 1 hour incubation, cAMP concentrations are estimated, according to the manufacturer specification, with the HTRF® kit.
  • Assay 2 (Table B2): This assay used HEK-293 cells expressing human recombinant adenosine A2A receptor (or, alternatively, A1 receptor) that were grown prior to the test in media without antibiotic. 100 nL of test articles at 100× of final concentration were transferred to assay plate by Echo. Cells were washed twice with 5 mL of PBS and 10 μL of cells were mixed with 5 mL PBS. After aspirating the PBS and adding 1.5 mL versine, cells were incubated at 37° C. for 2-5 min. After centrifugation, 4 mL of medium was added and adjusted cell density to 5,000 cells/well with Stimulation Buffer. 10 μL of cells were aliquoted to the assay plate, centrifuged at 1000 rpm for 1 minute, and incubated for 60 minutes at room temperature. 5 μL 4×Eu-cAMP tracer solution and 5 μL 4×Ulight™-anti-cAMP solution were added to assay plate, followed by centrifugation and 60-minute incubation at room temperature. Plates were read on EnVision.
  • As shown in Table B2, certain of the compounds disclosed herein strongly reduced intracellular levels of cAMP. For example, compound 89 had an IC50 for reducing cAMP levels of 58 nM in the A2A assay.
  • TABLE B2
    A1 cAMP A2a cAMP
    Compound No. IC50 (nM) IC50 (nM)
    1 ND 262
    73 403 53
    75 219 25
    76 804 24
    89 ND 58
    98 11 18
    2-2 ND 327
    2-6 5 44
    2-7 ND 130
     2-10 27 39
     2-13 ND 527
    99 ND 72
    100 22 28
    101 ND 238
    102 24 143
    103 ND 1421
    104 117 76
    105 10 48
    106 ND 165
    107 ND 191
    108 326 55
    110 ND 75
    122 ND 33
    125 ND 27
    126 ND 110
    ND: Not Determined
  • Example B3 GTPγ35S Scintillation Proximity Assay for A2A Receptor
  • A scintillation proximity assay (SPA) is used to determine the kinetic profile of the binding of test compound to the A2A receptor.
  • For antagonist testing, membrane extracts are prepared from HEK-293 cells expressing recombinant human A2A receptor, are mixed with GDP (volume:volume) and are incubated in assay buffer comprising 20 mM HEPES pH 7.4; 100 mM NaCl, 10 μg/mL saponin, 5 mM MgCl2 for at least 15 min on ice. In parallel, GTPγ[35S] is mixed with the beads (volume:volume) just before starting the reaction. The following reagents are successively added in the wells of an Optiplate (Perkin Elmer): 25 μL of test compound or reference ligand, 25 μL of the membranes: GDP mix, 25 μL of reference agonist at historical EC50 and 25 μL of GTPγ[35S] (PerkinElmer NEG030X), diluted in assay buffer to give 0.1 nM. The plate is incubated at room temperature for 1 hour. Then, 20 μL of IGEPAL is added for 30 minutes at room temperature. Following this incubation, 20 μL of beads (PVT-anti rabbit (PerkinElmer, RPNQ0016)), diluted in assay buffer at 50 mg/mL (0.5 mg/10 μL) and 20 μL of an Anti-GαS/olf antibody are added for a final incubation of 3 hours at room temperature. Then, the plates are centrifuged for 10 min at 2000 rpm, incubated at room temperature for 1 hour and counted for 1 min/well with a PerkinElmer TopCount reader
  • Example B4 Functional T Cell Assay
  • Human T Cell Activation Assay: Fresh human blood is diluted with the same volume of PBS and the buffy coat containing peripheral blood mononuclear cells (PBMCs) is prepared and resuspended in culture medium at a density of 2×106/mL. 2×105 PBMCs (in 100 μL) are plated to each well of a 96-well flat bottom plate. 25 μL of 8× final concentration of 10-fold serial diluted or single concentration compounds are added to indicated wells and incubated for 30 mins in 37° C./5% CO2. 25 μL of 8× final concentration of NECA (1 μM) is added to indicated wells and incubated for 30 min in 37° C./5% CO2. Beads included in T cell activation/expansion kit (Miltenyi biotec Cat #130-091-441) at a bead-to-cell ratio of 1:6 in 50 μL is added to all wells with the final concentration of DMSO at 0.1% and final volume at 200 μL. 60 μL of supernatant post 24 hr and 48 hr incubation is collected for TNF-α and IFN-γ concentration evaluation using TNF-α ELISA ready-set-go kit (eBioscience, Cat #88-7346-77) and IFN-γ ELISA ready-set-go kit (eBioscience, Cat #88-7316-77), respectively.
  • Example B5 cAMP Assay
  • In a 96-well plate coated with anti-CD3 antibody, CD8+ T-cells (1×105) are cultured alone, with 3 μM of NECA, or in the presence of 1 μM of the compound of interest with or without 3 μM of NECA. The cells are incubated for 30 min at 37° C. and 5% CO2, and the reaction is stopped by addition of 200 μL, 0.1 M hydrochloric acid. cAMP levels are determined by an ELISA kit.
  • Example B6 Anti-Tumor Activities in Immuno-Oncology Mouse Models
  • The anti-tumor activities of test articles are evaluated in selective mouse models (e.g., syngeneic model, xenograft model, or PDX) as a monotherapy or combination therapies. Using MC-38 syngeneic model as an example: female C57BL/6 mice are inoculated subcutaneously at right flank with MC-38 cells for tumor development. Five days after tumor inoculation, mice with tumor size ranging from 40-85 mm3 are selected and assigned into sub-groups using stratified randomization with 10 mice per group based upon their tumor volumes. Mice receive pre-defined treatments include vehicle, test article at various doses alone, test article at various doses plus other anti-cancer therapy, and other anti-cancer therapy control. Body weight and tumor sizes are measured three times per week during the treatment. Tumor volume is expressed in mm3 using the formula: V=0.5 a×b2 where a and b are the long and short diameters of the tumor, respectively. The tumor sizes are used for the calculations of both tumor growth inhibition (TGI) and T/C values. When an individual animal reaches to the termination endpoint (e.g., with TV >1000 mm3), the mouse is euthanized. The time from inoculation to the termination are deemed as its survival time. Survival curves are plotted by the Kaplan-Meier method. At the end of study, plasma and tumor samples are collected to explore biomarkers.
  • Example B7 Mouse Splenocyte Assay
  • IC50 values of compounds for reversal of NECA suppression of mIFNγ release is determined in mouse splenocytes isolated from Balb/c mice. The mIFNγ release is CD3e/CD28-induced release. Mouse splenocytes (2×105 cells/well) are activated with Anti-mouse CD3e (2.5 μg/ml, coated overnight at 4° C.; Cat #14-0032-82, eBioscience) and then incubated with serial dilutions of compounds (3 fold, 8 point dose response starting at 1 μM) in the presence of NECA (at a concentration such as 0.1, 3.0, or 6.0 μM; Cat #E2387, Sigma) for 30 min at 37° C., 5% CO2 in an incubator (cell culture conditions) prior to treating them with Anti-mouse CD28 (0.1 μg/ml soluble; Cat #16-0289-81, eBiosciences). Splenocytes are further incubated under cell culture conditions for 72 hr; the supernatant is then harvested and diluted to 1:100, and ELISA is performed as per the manufacturer's protocol (mIFN-γ kit; Cat #555138 and 550534, BD Biosciences). Plates are read in a plate reader by measuring absorbance at 450 nm. Values for the reversal of NECA suppressed mIFN-γ release by compounds are calculated by the following formula:
  • Normalized mIFN - γ release = ( [ mIFN - γ ] test - [ mIFN - γ ] blank ) ( [ mIFN - γ ] NECA - [ mIFN - γ ] blank )
  • where [mIFN-γ]test is the test reading, [mIFN-γ]blank is the average reading obtained from blank wells, and [mIFN-γ]NECA is the average reading obtained from NECA treated, activated cells. The IC50 values are calculated by fitting the curve to the “four-parameter variable slope logistic model” using Graph Pad Prism.
  • Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is apparent to those skilled in the art that certain minor changes and modifications will be practiced in light of the above teaching. Therefore, the description and examples should not be construed as limiting the scope of the invention.
  • All references throughout, such as publications, patents, and published patent applications, are incorporated herein by reference in their entireties.

Claims (26)

1. A compound of the formula (I):
Figure US20220298142A1-20220922-C00662
or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein:
A is selected from the group consisting of:
Figure US20220298142A1-20220922-C00663
B is a phenyl optionally substituted by R3, C3-C6 cycloalkyl optionally substituted by R4, 3- to 6-membered heterocyclyl optionally substituted by R4 or a 5- to 10-membered heteroaryl optionally substituted by R4;
R1 is a hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, 5- to 10-membered heteroaryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), —(C1-C3 alkylene)(5-6-membered heteroaryl), —(C1-C3 alkylene)(C6 aryl), —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, —NR1bR1c, —S(O)2R1a, —(C1-C3 alkylene)C(O)NR1bR1c, —(C1-C3 alkylene)C(O)R1a or —(C1-C3 alkylene)NR1bR1c, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, 5- to 10-membered heteroaryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), —(C1-C3 alkylene)(5-6-membered heteroaryl), and —(C1-C3 alkylene)(C6 aryl) of R1 are independently optionally substituted by R4;
each R1a is independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, 3-6-membered heterocyclyl, C6 aryl, 5-6-membered heteroaryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3alkylene)(3-6-membered heterocyclyl), —(C1-C3 alkylene)(C6 aryl) or —(C1-C3 alkylene)(5-6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3;
each R1b and R1c is independently hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl, 3-6-membered heterocyclyl, C6 aryl, 5-6-membered heteroaryl, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), —(C1-C3 alkylene)(C6 aryl) or —(C1-C3alkylene)(5-6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3;
or R1b and R1c are taken together with the nitrogen atom to which they are attached to form a 3- to 6-membered heterocyclyl;
R2 is —OR2a, —NHR2b, —C(O)NHR2b, or C1-C6 alkyl, wherein the C1-C6 alkyl of R2 is substituted by —OR2c, —NHR2c, or —C(O)NHR2c;
each R2a and R2b is independently cyclohexane, 6-membered heterocyclyl, —(C1-C3alkylene)N(C2H5)2, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), or —(C1-C3 alkylene)(5-6-membered heteroaryl), wherein each of which is optionally substituted by methyl, ethyl, halogen, oxo, —CF3, —OH, —OCH3, —CN, —C(O)OCH3, —C(O)OC2H5, —NH2 or —NHCH3;
R2c is 5- or 6-membered heteroaryl, wherein the 5- or 6-membered heteroaryl is further substituted by C1-C6 alkyl optionally substituted by halogen, —OH or oxo;
each R3 is independently C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —OC(O)R5, —C(O)OR5, —C(O)NR6R7, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)OR6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, —NR5S(O)R6, —C(O)NR5S(O)R6, —NR5S(O)2R6, —C(O)NR5S(O)2R6, —S(O)NR6R7, —S(O)2NR6R7, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —(C1-C3 alkylene)CN, —(C1-C3 alkylene)OR5, —(C1-C3 alkylene)SR5, —(C1-C3 alkylene)NR6R7, —(C1-C3 alkylene)CF3, —(C1-C3 alkylene)NO2, —C═NH(OR5), —(C1-C3 alkylene)C(O)R5, —(C1-C3 alkylene)OC(O)R5, —(C1-C3 alkylene)C(O)OR5, —(C1-C3 alkylene)C(O)NR6R7, —(C1-C3 alkylene)OC(O)NR6R7, —(C1-C3 alkylene)NR5C(O)R6, —(C1-C3 alkylene)NR5C(O)OR6, —(C1-C3 alkylene)NR5C(O)NR6R7, —(C1-C3 alkylene)S(O)R5, —(C1-C3 alkylene)S(O)2R5, —(C1-C3 alkylene)NR5S(O)R6, —C(O)(C1-C3 alkylene)NR5S(O)R6, —(C1-C3 alkylene)NR5S(O)2R6, —(C1-C3 alkylene)C(O)NR5S(O)2R6, —(C1-C3 alkylene)S(O)NR6R7, —(C1-C3 alkylene)S(O)2NR6R7, —(C1-C3 alkylene)(C3-C6 cycloalkyl), —(C1-C3 alkylene)(3-6-membered heterocyclyl), wherein each R3 is independently optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
each R4 is independently oxo or R3;
R5 is independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
R6 and R7 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9, or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
or R6 and R7 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo, —CN, —OR8, —NR8R9, —C(O)R8, —C(O)OR8, —C(O)NR8R9, —NR8C(O)R9, —S(O)R8, —S(O)2R8, —S(O)2NR8R9, —NR8S(O)2R9 or C1-C6 alkyl optionally substituted by oxo, —OH or halogen;
R8 and R9 are each independently hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3- to 6-membered heterocyclyl, wherein each of which is optionally substituted by halogen, OH, oxo or NH2;
or R8 and R9 are taken together with the atom to which they attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C1-C6 alkyl optionally substituted by halogen, OH, oxo or NH2.
2. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein:
B is a phenyl optionally substituted by R3, or a 5- or 6-membered heteroaryl optionally substituted by R4;
R1 is a hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- to 6-membered heterocyclyl, —C(O)R1a, —C(O)OR1a, —C(O)NR1bR1c, or —NR1bR1c, wherein the C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl and 3- to 6-membered heterocyclyl of R1 are independently optionally substituted by R4;
each R1a is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
each R1b and R1c is independently hydrogen, C1-C6 alkyl, or C3-C6 cycloalkyl;
or R1b and R1c are taken together with the nitrogen atom to which they are attached to form a 3- to 6-membered heterocyclyl.
3. (canceled)
4. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein R1 is hydrogen.
5. (canceled)
6. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein R2 is —OR2a.
7. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein R2 is —NHR2b.
8. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein R2 is —C(O)NHR2b.
9. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein R2 is C1-C6 alkyl substituted by —OR2c, —NHR2c, or —C(O)NHR2c.
10-12. (canceled)
13. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein R4 is R3 and each R3 is independently selected from the group consisting of halogen, —CN, —OR5, —SR5, —NR6R7, —NO2, —C(O)R5, —C(O)OR5, —C(O)NR6R7, —C(O)NR5S(O)2R6, —OC(O)R5, —OC(O)NR6R7, —NR5C(O)R6, —NR5C(O)NR6R7, —S(O)R5, —S(O)2R5, C3-C6 cycloalkyl and C1-C6 alkyl optionally substituted by halogen.
14. The compound of claim 13, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein each R3 is independently selected from the group consisting of halogen, —OR5 and C1-C6 alkyl optionally substituted by halogen.
15. (canceled)
16. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein B is a phenyl optionally substituted by R3.
17. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein B is a 5- to 6-membered heteroaryl optionally substituted by R4.
18. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein B is a 6-membered heteroaryl selected from the group consisting of pyridyl and pyrimidinyl, which is optionally substituted by R4.
19-20. (canceled)
21. The compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, wherein B is selected from the group consisting of:
Figure US20220298142A1-20220922-C00664
22-29. (canceled)
30. A compound of Table 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
31. A pharmaceutical composition comprising a compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, and a pharmaceutically acceptable carrier.
32. A method of treating a disease mediated by an adenosine signaling pathway in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
33. A method of treating cancer in an individual in need thereof comprising administering to the individual a therapeutically effective amount of a compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing.
34. A method of inhibiting an adenosine receptor of subtype A2A, A2B or A3 in a cell, comprising administering a compound of claim 1, or a tautomer or stereoisomer thereof, or a pharmaceutically acceptable salt of any of the foregoing, to the cell.
35. The method of claim 34, wherein the adenosine receptor is of subtype A2A.
36-37. (canceled)
US17/684,626 2019-01-18 2022-03-02 Heterocyclic compounds as adenosine antagonists Pending US20220298142A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/684,626 US20220298142A1 (en) 2019-01-18 2022-03-02 Heterocyclic compounds as adenosine antagonists

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201962794537P 2019-01-18 2019-01-18
US201962796046P 2019-01-23 2019-01-23
US16/746,763 US11306071B2 (en) 2019-01-18 2020-01-17 Heterocyclic compounds as adenosine antagonists
US17/684,626 US20220298142A1 (en) 2019-01-18 2022-03-02 Heterocyclic compounds as adenosine antagonists

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/746,763 Continuation US11306071B2 (en) 2019-01-18 2020-01-17 Heterocyclic compounds as adenosine antagonists

Publications (1)

Publication Number Publication Date
US20220298142A1 true US20220298142A1 (en) 2022-09-22

Family

ID=71609717

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/746,763 Active 2040-02-10 US11306071B2 (en) 2019-01-18 2020-01-17 Heterocyclic compounds as adenosine antagonists
US17/684,626 Pending US20220298142A1 (en) 2019-01-18 2022-03-02 Heterocyclic compounds as adenosine antagonists

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/746,763 Active 2040-02-10 US11306071B2 (en) 2019-01-18 2020-01-17 Heterocyclic compounds as adenosine antagonists

Country Status (13)

Country Link
US (2) US11306071B2 (en)
EP (1) EP3911323A4 (en)
JP (1) JP2022517419A (en)
KR (1) KR20210116550A (en)
CN (1) CN113924095A (en)
AU (1) AU2020208644A1 (en)
BR (1) BR112021013936A2 (en)
CA (1) CA3126702A1 (en)
CL (1) CL2021001884A1 (en)
IL (1) IL284767A (en)
MX (1) MX2021008650A (en)
SG (1) SG11202107810WA (en)
WO (1) WO2020150674A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2018302179A1 (en) 2017-07-18 2020-02-13 Nuvation Bio Inc. Heterocyclic compounds as adenosine antagonists
EP3911324A4 (en) 2019-01-18 2022-08-17 Nuvation Bio Inc. 1,8-naphthyridinone compounds and uses thereof
US20220259195A1 (en) * 2021-02-03 2022-08-18 Nuvation Bio Inc. Crystalline forms of a cyclin-dependent kinase inhibitor
WO2023201267A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers

Family Cites Families (72)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE47351E1 (en) 1999-06-22 2019-04-16 Gilead Sciences, Inc. 2-(N-pyrazolo)adenosines with application as adenosine A2A receptor agonists
WO2003030909A1 (en) 2001-09-25 2003-04-17 Bayer Pharmaceuticals Corporation 2- and 4-aminopyrimidines n-substtituded by a bicyclic ring for use as kinase inhibitors in the treatment of cancer
US6992087B2 (en) 2001-11-21 2006-01-31 Pfizer Inc Substituted aryl 1,4-pyrazine derivatives
AP2114A (en) 2003-02-26 2010-03-04 Sugen Inc Aminoheteroaryl compounds as protein kinase inhibitors
WO2004084824A2 (en) 2003-03-24 2004-10-07 Merck & Co., Inc. Biaryl substituted 6-membered heterocyles as sodium channel blockers
EP1678160A1 (en) 2003-10-27 2006-07-12 Astellas Pharma Inc. Pyrazine derivatives and pharmaceutical use thereof
CA2544191A1 (en) 2003-11-04 2005-05-26 Merck & Co., Inc. Substituted naphthyridinone derivatives
US10028058B2 (en) 2003-11-27 2018-07-17 Yul Anderson VSR surround sound tube headphone
CA2562126A1 (en) 2004-04-01 2005-10-13 Astellas Pharma Inc. Pyrazine derivatives and pharmaceutical use thereof
ES2270715B1 (en) * 2005-07-29 2008-04-01 Laboratorios Almirall S.A. NEW DERIVATIVES OF PIRAZINA.
GB0800741D0 (en) 2008-01-16 2008-02-20 Univ Greenwich Cyclic triazo and diazo sodium channel blockers
US8436171B2 (en) 2008-02-01 2013-05-07 Akinion Pharmaceuticals Ab Amino substituted pyrazines as inhibitors or protein kinases
KR101626996B1 (en) 2008-03-31 2016-06-02 제넨테크, 인크. Benzopyran and benzoxepin pi3k inhibitor compounds and methods of use
CA2750051A1 (en) 2009-02-02 2010-08-05 Merck Sharp & Dohme Corp. Inhibitors of akt activity
TW201102065A (en) 2009-05-29 2011-01-16 Astrazeneca Ab Heterocyclic urea derivatives and methods of use thereof
PT2531492T (en) 2010-02-05 2016-07-07 Heptares Therapeutics Ltd 1,2,4-triazine-4-amine derivatives
EA022046B1 (en) 2010-07-14 2015-10-30 Новартис Аг Ip receptor agonist heterocyclic compounds
US9142781B2 (en) 2011-06-09 2015-09-22 Novaled Ag Compound for organic electronic device
TW201335160A (en) 2012-01-13 2013-09-01 Novartis Ag IP receptor agonist heterocyclic compounds
US9266891B2 (en) 2012-11-16 2016-02-23 Boehringer Ingelheim International Gmbh Substituted [1,2,4]triazolo[4,3-A]pyrazines that are BRD4 inhibitors
US9675697B2 (en) 2013-03-11 2017-06-13 The Regents Of The University Of Michigan BET bromodomain inhibitors and therapeutic methods using the same
EP3521284B1 (en) 2013-03-15 2020-12-02 The Trustees of Columbia University in the City of New York Pyrazine compounds as map kinase modulators and uses thereof
BR112015031073B1 (en) 2013-06-21 2022-11-29 Zenith Epigenetics Ltd BICYCLIC BROMODIMANIUM INHIBITORS AND PHARMACEUTICAL COMPOSITION CONTAINING SAID COMPOUNDS
CN104341386A (en) * 2013-07-23 2015-02-11 中国科学院上海药物研究所 Aryl heterocycle micromolecule compounds, derivatives thereof, and preparing methods and uses of the compounds and the derivatives
EP3027616B1 (en) 2013-07-30 2018-01-10 Boehringer Ingelheim International GmbH Azaindole compounds as modulators of rorc
EP3071205B1 (en) 2013-11-18 2020-02-05 Forma Therapeutics, Inc. Benzopiperazine compositions as bet bromodomain inhibitors
RU2727169C2 (en) 2013-11-18 2020-07-21 Форма Терапеутикс Инк. Composition of tetrahydroquinolines as bromodomain bet modifying agents
JP2017511801A (en) 2014-02-28 2017-04-27 ザ リージェンツ オブ ザ ユニヴァシティ オブ ミシガン 9H-pyrimido [4,5-b] indoles and related analogs as BET bromodomain inhibitors
EA039678B1 (en) 2014-04-23 2022-02-24 Инсайт Корпорейшн 1H-PYRROLO[2,3-c]PYRIDIN-7(6H)-ONES AND PYRAZOLO[3,4-c]PYRIDIN-7(6H)-ONES AS INHIBITORS OF BET PROTEINS
AU2015317322B2 (en) 2014-09-19 2020-09-17 Forma Therapeutics, Inc. Phenyl quinolinone derivatives as mutant-isocitrate dehydrogenase inhibitors
US10472347B2 (en) 2014-11-18 2019-11-12 Merck Sharp & Dohme Corp. Aminopyrazine compounds with A2A antagonist properties
WO2016092375A1 (en) 2014-12-11 2016-06-16 Zenith Epigenetics Corp. Substituted heterocycles as bromodomain inhibitors
US10307407B2 (en) 2015-02-27 2019-06-04 The Regents Of The University Of Michigan 9H-pyrimido [4,5-B] indoles as BET bromodomain inhibitors
JP2018527293A (en) 2015-06-16 2018-09-20 オリオン コーポレーション Spiro [cyclobutane-1,3'-indoline] -2'-one derivatives as bromodomain inhibitors
AU2016306090B2 (en) 2015-08-11 2019-05-02 Novartis Ag 5-bromo-2,6-di-(1H-pyrazol-l-yl)pyrimidin-4-amine for use in the treatment of cancer
US9771363B2 (en) 2015-09-21 2017-09-26 Plexxikon Inc. Heterocyclic compounds and uses thereof
WO2017050714A1 (en) 2015-09-22 2017-03-30 Glaxosmithkline Intellectual Property (No.2) Limited Pyridinone dicarboxamide for use as bromodomain inhibitors
EA201891509A1 (en) 2015-12-24 2019-01-31 Корвус Фармасьютикалз, Инк. METHODS OF TREATING CANCER
MX2019000200A (en) 2016-07-01 2019-09-26 G1 Therapeutics Inc Pyrimidine-based antiproliferative agents.
WO2018005533A1 (en) 2016-07-01 2018-01-04 G1 Therapeutics, Inc. Antiproliferative pyrimidine-based compounds
BR112018077155A2 (en) 2016-07-01 2019-04-02 G1 Therapeutics, Inc. process for preparing a compound and compound
GB201612092D0 (en) 2016-07-12 2016-08-24 Almac Discovery Ltd Pharmaceutical compounds
WO2018081863A1 (en) 2016-11-04 2018-05-11 University Of Wollongong 6-SUBSTITUTED DERIVATIVES OF HEXAMETHYLENE AMILORIDE AS INHIBITORS OF uPA AND USES THEREOF
EP3570844B1 (en) 2017-01-20 2023-09-06 Arcus Biosciences, Inc. Azolopyrimidine for the treatment of cancer-related disorders
TWI801372B (en) 2017-03-30 2023-05-11 比利時商艾特歐斯比利時有限公司 Thiocarbamate derivatives as a2a inhibitors and methods for use in the treatment of cancers
HRP20221039T1 (en) 2017-03-30 2022-11-11 iTeos Belgium SA 2-oxo-thiazole derivatives as a2a inhibitors and compounds for use in the treatment of cancers
CN107221611B (en) 2017-06-15 2019-02-05 江西冠能光电材料有限公司 A kind of stable easy processing organic semiconducting materials and its organic luminescent device application
BR112019027446A2 (en) 2017-06-30 2020-07-07 Ryvu Therapeutics S.A. adenosine a2a receptor modulators
CN111093666A (en) 2017-07-18 2020-05-01 诺维逊生物股份有限公司 1, 8-naphthyridinone compounds and uses thereof
AU2018302179A1 (en) 2017-07-18 2020-02-13 Nuvation Bio Inc. Heterocyclic compounds as adenosine antagonists
CN111565722A (en) 2017-11-06 2020-08-21 科尔沃斯制药股份有限公司 Adenosine pathway inhibitors for cancer treatment
CN109810111B (en) 2017-11-20 2023-10-27 上海医药集团股份有限公司 Pyrazolopyrimidine compound, and preparation method and application thereof
WO2019118313A1 (en) 2017-12-13 2019-06-20 Merck Sharp & Dohme Corp. Imidazo [1,2-c] quinazolin-5-amine compounds with a2a antagonist properties
AU2018391833A1 (en) 2017-12-20 2020-07-30 Betta Pharmaceuticals Co., Ltd Compound functioning as bromodomain protein inhibitor, and composition
WO2019134539A1 (en) 2018-01-05 2019-07-11 四川科伦博泰生物医药股份有限公司 Dihydropyrazolone and pyrimidine compound, preparation method and use therefor
CN110041333B (en) 2018-01-16 2022-03-01 中国科学院上海药物研究所 Bromodomain inhibitor compounds and uses thereof
TW201940166A (en) 2018-01-29 2019-10-16 美商貝達醫藥公司 2H-indazole derivatives as CDK4 and CDK6 inhibitors and therapeutic uses thereof
CA3089159A1 (en) 2018-02-06 2019-08-15 Jiangsu Hengrui Medicine Co., Ltd. Pyrazolo[1,5-a][1,3,5]triazine-2-amine derivative, preparation method therefor and medical use thereof
WO2019160829A1 (en) 2018-02-13 2019-08-22 Iovance Biotherapeutics, Inc. Expansion of tumor infiltrating lymphocytes (tils) with adenosine a2a receptor antagonists and therapeutic combinations of tils and adenosine a2a receptor antagonists
CN111989313A (en) 2018-02-15 2020-11-24 杭州阿诺生物医药科技有限公司 A2A and/or A2B receptor antagonists
WO2019165204A1 (en) 2018-02-23 2019-08-29 Newave Pharmaceutical Inc. 1,2-dihydro-3h-pyrazolo[3,4-d]pyrimidine-3-one compounds as inhibitors of wee-1 kinase
CN112384515A (en) 2018-02-27 2021-02-19 因赛特公司 Imidazopyrimidines and triazolopyrimidines as A2A/A2B inhibitors
WO2019169065A2 (en) 2018-02-28 2019-09-06 The Regents Of The University Of Colorado, A Body Corporate Wee1 kinase inhibitors and methods of treating cancer using the same
JP7300460B2 (en) 2018-03-09 2023-06-29 リキュリウム アイピー ホールディングス リミテッド ライアビリティー カンパニー Substituted 1,2-dihydro-3H-pyrazolo[3,4-d]pyrimidin-3-ones
US11220510B2 (en) 2018-04-09 2022-01-11 Incyte Corporation Pyrrole tricyclic compounds as A2A / A2B inhibitors
CN112055709A (en) 2018-08-17 2020-12-08 迪哲(江苏)医药股份有限公司 Pyrazine compounds and uses thereof
TWI820209B (en) 2018-09-12 2023-11-01 大陸商迪哲(江蘇)醫藥股份有限公司 Triazolo-pyrimidine compounds and uses thereof
EP3911324A4 (en) 2019-01-18 2022-08-17 Nuvation Bio Inc. 1,8-naphthyridinone compounds and uses thereof
CN113613655A (en) 2019-01-18 2021-11-05 诺维逊生物股份有限公司 Compound and use thereof
WO2020150677A1 (en) 2019-01-18 2020-07-23 Nuvation Bio Inc. Heterocyclic compounds as adenosine antagonists
WO2021146629A1 (en) 2020-01-17 2021-07-22 Nuvation Bio Inc. Heterocyclic compounds as adenosine antagonists
WO2021146631A1 (en) 2020-01-17 2021-07-22 Nuvation Bio Inc. Heterocyclic compounds as adenosine antagonists

Also Published As

Publication number Publication date
US20200231570A1 (en) 2020-07-23
MX2021008650A (en) 2021-11-03
KR20210116550A (en) 2021-09-27
CL2021001884A1 (en) 2022-03-04
AU2020208644A1 (en) 2021-08-26
CN113924095A (en) 2022-01-11
CA3126702A1 (en) 2020-07-23
US11306071B2 (en) 2022-04-19
EP3911323A1 (en) 2021-11-24
IL284767A (en) 2021-08-31
BR112021013936A2 (en) 2021-09-21
SG11202107810WA (en) 2021-08-30
WO2020150674A1 (en) 2020-07-23
JP2022517419A (en) 2022-03-08
EP3911323A4 (en) 2022-11-16

Similar Documents

Publication Publication Date Title
US20210292285A1 (en) Heterocyclic compounds as adenosine antagonists
US11306071B2 (en) Heterocyclic compounds as adenosine antagonists
US10793561B2 (en) 1,8-naphthyridinone compounds and uses thereof
US20220169648A1 (en) 1,8-naphthyridinone compounds and uses thereof
US20220119367A1 (en) Heterocyclic compounds as adenosine antagonists
US20230066315A1 (en) Compounds and uses thereof
US20220169646A1 (en) Heterocyclic compounds and uses thereof
WO2021146631A1 (en) Heterocyclic compounds as adenosine antagonists
WO2021146629A1 (en) Heterocyclic compounds as adenosine antagonists

Legal Events

Date Code Title Description
AS Assignment

Owner name: NUVATION BIO OPERATING COMPANY INC. (FORMERLY KNOWN AS NUVATION BIO INC.), NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GIRAFPHARMA LLC;REEL/FRAME:059163/0818

Effective date: 20211203

Owner name: GIRAFPHARMA LLC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:SPARCBIO LLC;REEL/FRAME:059163/0809

Effective date: 20211026

Owner name: SPARCBIO LLC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:INTEGRAL BIOSCIENCES PVT. LTD.;REEL/FRAME:059163/0786

Effective date: 20210727

Owner name: SPARCBIO LLC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PHAM, SON MINH;KANKANALA, JAYAKANTH;REEL/FRAME:059163/0766

Effective date: 20210913

Owner name: NUVATION BIO OPERATING COMPANY LLC, NEW YORK

Free format text: CHANGE OF NAME;ASSIGNOR:NUVATION BIO OPERATING COMPANY INC.;REEL/FRAME:059317/0852

Effective date: 20211229

Owner name: NUVATION BIO INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NUVATION BIO OPERATING COMPANY LLC (FORMALLY NUVATION BIO OPERATING COMPANY INC., WHICH WAS FORMALLY KNOWN AS NUVATION BIO INC.);REEL/FRAME:059163/0993

Effective date: 20220128

Owner name: INTEGRAL BIOSCIENCES PVT. LTD., INDIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JADHAVAR, PRADEEP S.;MULIK, BABAN MOHAN;KHAN, FARHA;AND OTHERS;REEL/FRAME:059317/0835

Effective date: 20210719

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION