WO2023183755A1 - Pyrimidones tricycliques - Google Patents

Pyrimidones tricycliques Download PDF

Info

Publication number
WO2023183755A1
WO2023183755A1 PCT/US2023/064622 US2023064622W WO2023183755A1 WO 2023183755 A1 WO2023183755 A1 WO 2023183755A1 US 2023064622 W US2023064622 W US 2023064622W WO 2023183755 A1 WO2023183755 A1 WO 2023183755A1
Authority
WO
WIPO (PCT)
Prior art keywords
trifluoromethyl
cancer
dihydro
quinazolin
title compound
Prior art date
Application number
PCT/US2023/064622
Other languages
English (en)
Inventor
Marcos GONZALEZ-LOPEZ
Jean-Michel Vernier
Ping Chen
Original Assignee
Erasca, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Erasca, Inc. filed Critical Erasca, Inc.
Publication of WO2023183755A1 publication Critical patent/WO2023183755A1/fr

Links

Classifications

    • 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
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains three hetero rings
    • C07D513/16Peri-condensed systems

Definitions

  • Embodiments herein relate to compounds and methods for the treatment of RAS- mediated disease.
  • embodiments herein relate to compounds and methods for treating diseases such as cancer via targeting oncogenic mutants of the K-RAS isoform.
  • Ras proteins are small guanine nucleotide-binding proteins that act as molecular switches by cycling between active GTP-bound and inactive GDP-bound conformations. Ras signaling is regulated through a balance between activation by guanine nucleotide exchange factors (GEFs), most commonly son of sevenless (SOS), and inactivation by GTPase -activating proteins (GAPs) such as neurofibromin or pl20GAP.
  • GEFs guanine nucleotide exchange factors
  • SOS most commonly son of sevenless
  • GAPs GTPase -activating proteins
  • Ras proteins play an important role in the regulation of cell proliferation, differentiation, and survival. Dysregulation of the Ras signaling pathway is almost invariably associated with disease . Hyper-activating somatic mutations in Ras are among the most common lesions found in human cancer. Most of these mutations have been shown to decrease the sensitivity of Ras to GAP stimulation and decrease its intrinsic GTPase activity, leading to an increase in the active GTP-bound population. Although mutation of any one of the three Ras isoforms (K- Ras, N-Ras, or H-Ras) has been shown to lead to oncogenic transformation, K-Ras mutations are by far the most common in human cancer.
  • K- Ras mutations are known to be often associated with pancreatic, colorectal and non-small-cell lung carcinomas.
  • H-Ras mutations are common in cancers such as papillary thyroid cancer, lung cancers and skin cancers.
  • N-Ras mutations occur frequently in hepatocellular carcinoma.
  • A is alkyl, alkenyl, alkynyl, alkoxy, cycloalkoxy, cyano, hydrogen, trifluoromethyl, trifluoromethoxy, halogen, -SF5, cycloalkyl, cycloalkenyl, bicycloalkyl, tricycloalkyl, tetracycloalkyl, or pentacycloalkyl, wherein when A comprises one or more C-H groups, one or more carbon atom is optionally substituted with 1 to 3 F, Cl, alkyl, or CF3; and any of which optionally replace 1 or 2 carbon atoms with a heteroatom selected from NR, O, or S; wherein R is H or Me;
  • X is O or S(O) P , wherein p is an integer from 0 to 2;
  • R 2 is selected from the group consisting of:
  • each R’ or R is selected from the group consisting of hydrogen, alkyl, alkoxy, alkoxyalkyl, hydroxy, cycloalkoxy, cyano, cyanoalkyl, amido, amidoalkyl, N-alkylamido, N-alkylamidoalkyl, N,N- dialkylamido, N,N-dialkylamidoalkyl, amino, aminoalkyl, N-alkyl amino, N-alkyl aminoalkyl, N,N-dialkylamino, and N,N -dialkylaminoalkyl, any of which are optionally substituted; or any two R’ and R” combine to form 3-7-membered ring, optionally comprising 1 or 2 heteroatoms selected from N, O, or S; or any two R 2 combine to form a spirocycle comprising 0 to 2 heteroatoms selected from
  • embodiments herein relate to methods of treating a subject with cancer associated with a G12C Kras mutation comprising administering to the subject a compound, as disclosed herein, in a pharmaceutically acceptable vehicle.
  • potent and selective tricyclic quinazoline-2-ones compounds which have been found to be useful as inhibitors of oncogenic mutants of RAS proteins.
  • the compounds disclosed herein are selective for oncogenic RAS mutants over wild-type RAS proteins.
  • compounds disclosed herein may exhibit selectivity for oncogenic mutants of K-RAS over other mutated K- RAS proteins, as well as mutants of the N-RAS and H-RAS isoforms.
  • the compounds disclosed herein may exhibit selectivity for K-RAS, N-RAS, and H-RAS mutants having a common G12C mutation.
  • compositions comprising these compounds, and their application in the treatment of disease, such as cancer.
  • diseases such as cancer.
  • Methods of inhibition of oncogenic mutant K-RAS, N-RAS, and H-RAS activity are also provided, as well as methods for the treatment of oncogenic mutant RAS-mediated diseases, especially those involving elevated levels of oncogenic mutated RAS, in particular cancer.
  • A is alkyl, alkenyl, alkynyl, alkoxy, cycloalkoxy, cyano, hydrogen, trifluoromethyl, trifluoromethoxy, halogen, -SF5, cycloalkyl, cycloalkenyl, bicycloalkyl, tricycloalkyl, tetracycloalkyl, or pentacycloalkyl, wherein when A comprises one or more C-H groups, one or more carbon atom is optionally substituted with 1 to 3 F, Cl, alkyl, or CF3; and any of which optionally replace 1 or 2 carbon atoms with a heteroatom selected from NR, O, or S; wherein R is H or Me;
  • X is O or S(O) P , wherein p is an integer from 0 to 2;
  • R 2 is selected from the group consisting of:
  • each R’ or R is selected from the group consisting of hydrogen, alkyl, alkoxy, alkoxyalkyl, hydroxy, cycloalkoxy, cyano, cyanoalkyl, amido, amidoalkyl, N-alkylamido, N-alkylamidoalkyl, N,N- dialkylamido, N,N-dialkylamidoalkyl, amino, aminoalkyl, N-alkyl amino, N-alkyl aminoalkyl, N,N-dialkylamino, and N,N -dialkylaminoalkyl, any of which are optionally substituted; or any two R’ and R” combine to form 3-7-membered ring, optionally comprising 1 or 2 heteroatoms selected from N, O, or S; or any two R 2 combine to form a spirocycle comprising 0 to 2 heteroatoms selected from
  • compositions comprising one or more compounds disclosed herein together with a pharmaceutically acceptable carrier, as well as methods of making and using the compounds and compositions.
  • Embodiments disclosed herein provide methods for selectively inhibiting the RAS that are oncogenic mutants having the G12C mutation.
  • methods for treating an oncogenic mutant K- RAS-mediated disorder in a subject comprising administering to the subject a therapeutically effective amount of a compound or pharmaceutical composition according to the various embodiments disclosed herein.
  • Related embodiments disclose the use of the compounds disclosed herein as therapeutic agents, for example, in treating cancer and other diseases involving elevated levels of oncogenic mutant K-RAS.
  • the various embodiments disclosed herein also contemplate the use of the compounds disclosed herein for use in the manufacture of a medicament for the treatment of a disease or condition ameliorated by the inhibition of oncogenic mutant K-RAS.
  • the disease or condition is cancer.
  • Each of the aforementioned methods apply equally to the similar mutation in N-RAS and H-RAS bearing the G12C mutation.
  • Compounds of the various embodiments disclosed herein may be selective amongst the RAS oncogenic mutant forms in various ways. For example, compounds described herein may be selective for G12C mutants of K-RAS, N-RAS, or H-RAS. In certain embodiments, compounds of the various embodiments disclosed herein may be selective for K-RAS G12C over other K-RAS mutants and Wild Type K-RAS. Likewise, compounds of various embodiments disclosed herein may be selective for N-RAS and H- RAS bearing the same G12C mutation.
  • the various embodiments disclosed herein also relate to methods of inhibiting at least one RAS function comprising the step of contacting an oncogenic mutant RAS with a compound of Formula I, as described herein.
  • the cell phenotype, cell proliferation, activity of the mutant RAS, change in biochemical output produced by active mutant RAS, expression of mutant RAS, or binding of mutant RAS with a natural binding partner may be affected.
  • Such methods may be embrace modes of treatment of disease, biological assays, cellular assays, biochemical assays, or the like.
  • ‘A,” “an,” or “the” as used herein not only include aspects with one member, but also include aspects with more than one member.
  • the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise.
  • a cell includes a plurality of such cells and reference to “the agent” includes reference to one or more agents known to those skilled in the art, and so forth.
  • An “alkylcarbonyl” or “alkanoyl” group refers to an alkyl group attached to the parent molecular moiety through a carbonyl group. Examples of such groups include, without limitation, methylcarbonyl and ethylcarbonyl.
  • an “arylcarbonyl” or “aroyl” group refers to an aryl group attached to the parent molecular moiety through a carbonyl group.
  • examples of such groups include, without limitation, benzoyl and naphthoyl.
  • generic examples of acyl groups include alkanoyl, aroyl, heteroaroyl, and so on.
  • Specific examples of acyl groups include, without limitation, formyl, acetyl, acryloyl, benzoyl, trifluoroacetyl and the like.
  • alkenyl refers to a straightchain or branched-chain hydrocarbon radical having one or more double bonds and containing from 2 to 20 carbon atoms.
  • the alkenyl may comprise from 2 to 6 carbon atoms, or from 2 to 4 carbons, either of which may be referred to as “lower alkenyl.”
  • Alkenyl can include any number of carbons, such as C2, C23, C24, C25, C26, C27, C28, C29, C210, C3, C34, C35, C36, C4, C45, C46, C5, C56, and Ce, and so on up to 20 carbon atoms.
  • Alkenyl groups can have any suitable number of double bonds, including, but not limited to, 1, 2, 3, 4, 5 or more.
  • alkenyl groups include, but are not limited to, vinyl (ethenyl), propenyl, isopropenyl, Ibutenyl, 2butenyl, isobutenyl, butadienyl, Ipentenyl, 2pentenyl, isopentenyl, l,3pentadienyl, l,4pentadienyl, lhexenyl, 2hexenyl, 3hexenyl, l,3hexadienyl, l,4hexadienyl, l,5hexadienyl, 2,4hexadienyl, or l,3,5hexatrienyl.
  • Alkenyl groups can be substituted or unsubstituted. Unless otherwise specified, the term “alkenyl” may include “alkenylene” groups.
  • alkoxy refers to an alkyl ether radical, wherein the term alkyl is as defined below.
  • Alkoxy groups may have the general formula: alkyl-O-.
  • alkyl group alkoxy groups can have any suitable number of carbon atoms, such as Ci6.
  • Alkoxy groups include, for example, methoxy, ethoxy, propoxy, isopropoxy, butoxy, 2butoxy, isobutoxy, secbutoxy, tertbutoxy, pentoxy, hexoxy, and the like.
  • the alkoxy groups can be further optionally substituted as defined herein.
  • alkyl refers to a straight-chain or branched-chain alkyl radical containing from 1 to 20 carbon atoms.
  • the alkyl may comprise from 1 to 10 carbon atoms.
  • the alkyl may comprise from 1 to 6 carbon atoms, or from 1 to 4 carbon atoms.
  • Alkyl can include any number of carbons, such as C12, C13, C14, C15, Ci6, C17, Cis, C19, C110, C23, C24, C25, C26, C34, C35, C36, C45, C46 and C56.
  • Ci6 alkyl includes, but is not limited to, methyl, ethyl, propyl, isopropyl, butyl, isobutyl, secbutyl, tertbutyl, pentyl, isopentyl, hexyl, etc.
  • Alkyl can also refer to alkyl groups having up to 20 carbons atoms, such as, but not limited to heptyl, octyl, nonyl, decyl, etc. Alkyl groups can be substituted or unsubstituted.
  • alkylene refers to a saturated aliphatic group derived from a straight or branched chain saturated hydrocarbon attached at two or more positions, such as methylene (— CH2-). Unless otherwise specified, the term “alkyl” may include “alkylene” groups. When the alkyl is methyl, it may be represented structurally as CH3, Me, or just a single bond terminating with no end group substitution.
  • alkylamino refers to an alkyl group attached to the parent molecular moiety through an amino group. Suitable alkylamino groups may be mono- or dialkylated, forming groups such as, for example, N- methylamino (— NHMe), N-ethylamino (— NHEt), N,N-dimethylamino (— NMe2), N,N- ethylmethylamino (— NMeEt) and the like.
  • aminoalkyl refers to reverse orientation in which the amino group appears distal to the parent molecular moiety and attachment to the parent molecular moiety is through the alkyl group.
  • NEb CFEjn describes an aminoalkyl group with a terminal amine at the end of an alkyl group attached to the parent molecular moiety.
  • alkylamino and aminoalkyl can be combined to describe an “alkylaminoalkyl” group in which an alkyl group resides on a nitrogen atom distal to the parent molecular moiety, such as MeNH(CH2)n— .
  • an aryl group as defined herein, may combine in a similar fashion providing an arylaminoalkyl group ArNH(CH2)n— .
  • A- in the name, such as A-arylaminoalkyl, which is understood to mean that the aryl group is a substituent on the nitrogen atom of the aminoalkyl group, the alkyl being attached the parent molecular moiety.
  • alkylidene refers to an alkenyl group in which one carbon atom of the carbon-carbon double bond belongs to the moiety to which the alkenyl group is attached.
  • alkylthio refers to an alkyl thioether (AlkS-) radical wherein the term alkyl is as defined above and wherein the sulfur may be singly or doubly oxidized.
  • alkyl thioether radicals include methylthio, ethylthio, n-propylthio, isopropylthio, n-butylthio, iso-butylthio, sec-butylthio, tert-butylthio, methanesulfonyl, ethanesulfinyl, and the like.
  • arylthio refers to arylthioether (ArS-) radical wherein the term aryl is as defined herein and wherein the sulfur may be singly or double oxidized.
  • alkynyl refers to a straightchain or branched chain hydrocarbon radical having one or more triple bonds and containing from 2 to 20 carbon atoms. In certain embodiments, said alkynyl comprises from 2 to 6 carbon atoms. In further embodiments, said alkynyl comprises from 2 to 4 carbon atoms.
  • alkynyl ene refers to a carbon-carbon triple bond attached at two positions such as ethynylene.
  • Alkynyl can include any number of carbons, such as C2, C23, C24, C25, C26, C27, C28, C29, C210, C3, C34, C35, C36, C4, C45, C46, C5, C56, and Ce.
  • alkynyl groups include, but are not limited to, acetylenyl, propynyl, Ibutynyl, 2butynyl, butadiynyl, Ipentynyl, 2pentynyl, isopentynyl, l,3pentadiynyl, l,4pentadiynyl, lhexynyl, 2hexynyl, 3hexynyl, l,3hexadiynyl, l,4hexadiynyl, l,5hexadiynyl, 2,4hexadiynyl, or l,3,5hexatriynyl.
  • Alkynyl groups can be substituted or unsubstituted. Unless otherwise specified, the term “alkynyl” may include “alkynylene” groups.
  • acylamino as used herein, alone or in combination, refers to an amino group as described below attached to the parent molecular moiety through a carbonyl group.
  • acylamino as used herein, alone or in combination, embraces an acyl group attached to the parent moiety through an amino group.
  • An example of an “acylamino” group is acetylamino (CH3C(O)NH— ).
  • amino refers to — N(R)(R') or — N + (R)(R')(R"), wherein R, R and R" are independently selected from the group consisting of hydrogen, alkyl, acyl, heteroalkyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl, any of which may themselves be optionally substituted.
  • amino acid means a substituent of the form — NRCH(R')C(O)OH, wherein R is typically hydrogen, but may be cyclized with N (for example, as in the case of the amino acid proline), and R is selected from the group consisting of hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, amino, amido, cycloalkylalkyl, heterocycloalkylalkyl, arylalkyl, heteroarylalkyl, aminoalkyl, amidoalkyl, hydroxyalkyl, thiol, thioalkyl, alkylthioalkyl, and alkylthio, any of which may be optionally substituted.
  • amino acid includes all naturally occurring amino acids as well as synthetic analogues.
  • aryl as used herein, alone or in combination, means a carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendent manner or may be fused.
  • aryl embraces aromatic radicals such as benzyl, phenyl, naphthyl, anthracenyl, phenanthryl, indanyl, indenyl, annulenyl, azulenyl, tetrahydronaphthyl, and biphenyl.
  • arylalkenyl or “aralkenyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkenyl group.
  • arylalkoxy or “aralkoxy,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkoxy group.
  • arylalkyl or “aralkyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkyl group.
  • arylalkynyl or “aralkynyl,” as used herein, alone or in combination, refers to an aryl group attached to the parent molecular moiety through an alkynyl group.
  • arylalkanoyl or “aralkanoyl” or “aroyl,” as used herein, alone or in combination, refers to an acyl radical derived from an aryl-substituted alkanecarboxylic acid such as benzoyl, naphthoyl, phenylacetyl, 3 -phenylpropionyl (hydrocinnamoyl), 4- phenylbutyryl, (2-naphthyl)acetyl, 4-chlorohydrocinnamoyl, and the like.
  • aryloxy refers to an aryl group attached to the parent molecular moiety through an oxy.
  • benzo and “benz,” as used herein, alone or in combination, refer to the divalent radical CeH-i- derived from benzene. Examples include benzothiophene and benzimidazole.
  • carbamate refers to an ester of carbamic acid (— NHCOO— ) which may be attached to the parent molecular moiety from either the nitrogen or acid (oxygen) end, and which may be optionally substituted as defined herein.
  • O-carbamyl as used herein, alone or in combination, refers to a — OC(O)NRR', group, with R and R' as defined herein.
  • N-carbamyl as used herein, alone or in combination, refers to a R0C(0)NR— group, with R and R' as defined herein.
  • cyano as used herein, alone or in combination, refers to — CN.
  • cycloalkyl refers to a saturated or partially saturated monocyclic, bicyclic or tricyclic alkyl radical wherein each cyclic moiety contains from 3 to 12 carbon atom ring members and which may optionally be a benzo fused ring system which is optionally substituted as defined herein.
  • a cycloalkyl may comprise from from 3 to 7 carbon atoms, or from 5 to 7 carbon atoms.
  • cycloalkyl radicals examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, octahydronaphthyl, 2,3- dihydro-lH-indenyl, adamantyl and the like.
  • “Bicyclic” and “tricyclic” as used herein are intended to include both fused ring systems, such as decahydronaphthalene, octahydronaphthalene as well as the multicyclic (multicentered) saturated or partially unsaturated type. The latter type of isomer is exemplified in general by, bicyclofl. l .l]pentane, camphor, adamantane, and bicyclo[3.2.1]octane.
  • electrophilic moiety is used in accordance with its plain ordinary chemical meaning and refers to a chemical group that is electrophilic.
  • electrophilic moieties include, without limitation, unsaturated carbonyl containing compounds such as acrylamides, acrylates, unsaturated (i.e., vinyl) sulfones or phosphates, epoxides, and vinyl epoxides.
  • ether typically refers to an oxy group bridging two moieties linked at carbon atoms. “Ether” may also include polyethers, such as, for example, — RO CEE O CEE O CEE OR', — RO(CH2)2O(CH 2 )2OR, -RO(CH 2 )2OR, and -RO(CH 2 )2OH.
  • halo or halogen, as used herein, alone or in combination, refers to fluorine, chlorine, bromine, or iodine.
  • haloalkoxy refers to a haloalkyl group attached to the parent molecular moiety through an oxygen atom.
  • haloalkyl refers to an alkyl radical having the meaning as defined above wherein one or more hydrogens are replaced with a halogen. Specifically embraced are monohaloalkyl, dihaloalkyl, trihaloalkyl and polyhaloalkyl radicals.
  • a monohaloalkyl radical for one example, may have an iodo, bromo, chloro or fluoro atom within the radical.
  • Dihalo and polyhaloalkyl radicals may have two or more of the same halo atoms or a combination of different halo radicals.
  • haloalkyl radicals include fluoromethyl, difluoromethyl, trifluoromethyl, chloromethyl, dichloromethyl, trichloromethyl, pentafluoroethyl, heptafluoropropyl, difluorochloromethyl, dichlorofluoromethyl, difluoroethyl, difluoropropyl, dichloroethyl and dichloropropyl.
  • “Haloalkylene” refers to a haloalkyl group attached at two or more positions. Examples include fluoromethylene (— CFH— ), difluoromethylene (— CF2— ), chloromethylene (--CHC1— ) and the like.
  • heteroalkyl refers to a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, fully saturated or containing from 1 to 3 degrees of unsaturation, consisting of the stated number of carbon atoms and from one to three heteroatoms selected from the group consisting of O, N, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quatemized (i.e. bond to 4 groups).
  • the heteroatom(s) O, N and S may be placed at any interior position of the heteroalkyl group. Up to two heteroatoms may be consecutive, such as, for example, — CH2NHOCH3.
  • the term heteroalkyl may include ethers.
  • heteroaryl refers to 3 to 7 membered unsaturated heteromonocyclic rings, or fused polycyclic rings, each of which is 3 to 7 membered, in which at least one of the fused rings is unsaturated, wherein at least one atom is selected from the group consisting of O, S, and N.
  • a heteroaryl may comprise from 5 to 7 carbon atoms.
  • the term also embraces fused polycyclic groups wherein heterocyclic radicals are fused with aryl radicals, wherein heteroaryl radicals are fused with other heteroaryl radicals, or wherein heteroaryl radicals are fused with cycloalkyl radicals.
  • heteroaryl groups include pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, pyranyl, furyl, thienyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, thiadiazolyl, isothiazolyl, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, quinoxalinyl, quinazolinyl, indazolyl, benzotriazolyl, benzodioxolyl, benzopyranyl, benzoxazolyl, benzoxadiazolyl, benzothiazolyl, benzothiadiazolyl, benzofuryl, benzothienyl, chro
  • Exemplary tricyclic heterocyclic groups include carbazolyl, benzidolyl, phenanthrolinyl, dibenzofuranyl, acridinyl, phenanthridinyl, xanthenyl and the like.
  • Heteroaryl groups can include any number of ring atoms, such as, 5 to 6, 3 to 8, 4 to 8, 5 to 8, 6 to 8, 3 to 9, 3 to 10, 3 to 11, or 3 to 12 ring members. Any suitable number of heteroatoms can be included in the heteroaryl groups, such as 1, 2, 3, 4, or 5, or 1 to 2, 1 to 3, 1 to 4, 1 to 5, 2 to 3, 2 to 4, 2 to 5, 3 to 4, or 3 to 5. Heteroaryl groups can have from 5 to 8 ring members and from 1 to 4 heteroatoms, or from 5 to 8 ring members and from 1 to 3 heteroatoms, or from 5 to 6 ring members and from 1 to 4 heteroatoms, or from 5 to 6 ring members and from 1 to 3 heteroatoms.
  • the heteroaryl group can include groups such as pyrrole, pyridine, imidazole, pyrazole, triazole, tetrazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,3, 5 -isomers), thiophene, furan, thiazole, isothiazole, oxazole, and isoxazole.
  • heteroaryl groups can also be fused to aromatic ring systems, such as a phenyl ring, to form members including, but not limited to, benzopyrroles such as indole and isoindole, benzopyridines such as quinoline and isoquinoline, benzopyrazine (quinoxaline), benzopyrimidine (quinazoline), benzopyridazines such as phthalazine and cinnoline, benzothiophene, and benzofuran.
  • Other heteroaryl groups include heteroaryl rings linked by a bond, such as bipyridine.
  • Heteroaryl groups can be substituted or unsubstituted.
  • the heteroaryl groups can be linked via any position on the ring.
  • pyrrole includes 1-, 2- and 3-pyrrole
  • pyridine includes 2-, 3- and 4-pyridine
  • imidazole includes 1-, 2-, 4- and 5-imidazole
  • pyrazole includes 1-, 3-, 4- and 5-pyrazole
  • triazole includes 1-, 4- and 5-triazole
  • tetrazole includes 1- and 5-tetrazole
  • pyrimidine includes 2-, 4-, 5- and 6- pyrimidine
  • pyridazine includes 3- and 4-pyridazine
  • 1,2,3-triazine includes 4- and 5-triazine
  • 1 ,2,4-triazine includes 3-, 5- and 6-triazine
  • 1,3, 5 -triazine includes 2- triazine
  • thiophene includes 2- and 3-thiophene
  • furan includes 2- and 3-furan
  • thiazole includes 2-, 4- and 5-thiazole
  • isothiazole includes 3-,
  • heteroaryl groups include those having from 5 to 10 ring members and from 1 to 3 ring atoms including N, O or S, such as pyrrole, pyridine, imidazole, pyrazole, triazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,3,5-isomers), thiophene, furan, thiazole, isothiazole, oxazole, isoxazole, indole, isoindole, quinoline, isoquinoline, quinoxaline, quinazoline, phthalazine, cinnoline, benzothiophene, and benzofuran.
  • N, O or S such as pyrrole, pyridine, imidazole, pyrazole, triazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,
  • heteroaryl groups include those having from 5 to 8 ring members and from 1 to 3 heteroatoms, such as pyrrole, pyridine, imidazole, pyrazole, triazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,3,5-isomers), thiophene, furan, thiazole, isothiazole, oxazole, and isoxazole.
  • heteroatoms such as pyrrole, pyridine, imidazole, pyrazole, triazole, pyrazine, pyrimidine, pyridazine, triazine (1,2,3-, 1,2,4- and 1,3,5-isomers), thiophene, furan, thiazole, isothiazole, oxazole, and isoxazole.
  • heteroaryl groups include those having from 9 to 12 ring members and from 1 to 3 heteroatoms, such as indole, isoindole, quinoline, isoquinoline, quinoxaline, quinazoline, phthalazine, cinnoline, benzothiophene, benzofuran and bipyridine.
  • heteroaryl groups include those having from 5 to 6 ring members and from 1 to 2 ring atoms including N, O or S, such as pyrrole, pyridine, imidazole, pyrazole, pyrazine, pyrimidine, pyridazine, thiophene, furan, thiazole, isothiazole, oxazole, and isoxazole.
  • heterocycloalkyl and, interchangeably, “heterocycle,” or “heterocyclyl” as used herein, alone or in combination, each refer to a saturated, partially unsaturated, or fully unsaturated monocyclic, bicyclic, or tricyclic heterocyclic radical containing at least one heteroatom as ring members, wherein each heteroatom may be independently selected from the group consisting of nitrogen, oxygen, and sulfur.
  • a heterocycloalkyl may comprise from 1 to 4 heteroatoms as ring members.
  • a heterocycloalkyl may comprise from 1 to 2 heteroatoms ring members.
  • a heterocycloalkyl may comprise from 3 to 8 ring members in each ring. In further embodiments, a heterocycloalkyl may comprise from 3 to 7 ring members in each ring. In yet further embodiments, a heterocycloalkyl may comprise from 5 to 6 ring members in each ring.
  • “Heterocycloalkyl” and “heterocycle” are intended to include sugars, sulfones, sulfoxides, N-oxides of tertiary nitrogen ring members, and carbocyclic fused and benzo fused ring systems; additionally, both terms also include systems where a heterocycle ring is fused to an aryl group, as defined herein, or an additional heterocycle group.
  • heterocycloalkyl groups include aziridinyl, azetidinyl, 1,3-benzodioxolyl, dihydroisoindolyl, dihydroisoquinolinyl, dihydrocinnolinyl, dihydrobenzodioxinyl, dihydro[l,3]oxazolo[4,5-b]pyridinyl, benzothiazolyl, dihydroindolyl, dihy-dropyridinyl, 1,3-dioxanyl, 1,4-dioxanyl, 1,3- dioxolanyl, epoxy, isoindolinyl, morpholinyl, piperazinyl, pyrrolidinyl, tetrahydropyridinyl, piperidinyl, thiomorpholinyl, and the like.
  • Heterocycloalkyl may refer to a saturated ring system having from 3 to 12 ring members and from 1 to 5 heteroatoms of N, O and S.
  • the heteroatoms can also be oxidized, such as, but not limited to, S(O) and S(O)2 .
  • Heterocycloalkyl groups can include any number of ring atoms, such as, 3 to 6, 4 to 6, 5 to 6, 3 to 8, 4 to 8, 5 to 8, 6 to 8, 3 to 9, 3 to 10, 3 to 11, or 3 to 12 ring members.
  • heteroatoms can be included in the heterocycloalkyl groups, such as 1, 2, 3, 4, or 5, or 1 to 2, 1 to 3, 1 to 4, 1 to 5, 2 to 3, 2 to 4, 2 to 5, 3 to 4 or 3 to 5.
  • the heterocycloalkyl group can include any number of carbons, such as C3-6, C4-6, C5-6, C3-8, C4-8, C5-8, Ce-8, C3-9, C3-10, C3-11, and C3-12.
  • the heterocycloalkyl group can include groups such as aziridine, azetidine, pyrrolidine, piperidine, azepane, diazepane, azocane, quinuclidine, pyrazolidine, imidazolidine, piperazine (1,2-, 1,3- and 1,4-isomers), oxirane, oxetane, tetrahydrofuran, oxane (tetrahydropyran), oxepane, thiirane, thietane, thiolane (tetrahydrothiophene), thiane (tetrahydrothiopyran), oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, dioxolane, dithiolane, morpholine, thiomorpholine, dioxane, or dithiane.
  • groups such as aziridine, azetidine, pyrrolidine,
  • heterocycloalkyl groups can also be fused to aromatic or non-aromatic ring systems to form members including, but not limited to, indoline, diazabicycloheptane, diazabicyclooctane, diazaspirooctane or diazaspirononane.
  • Heterocycloalkyl groups can be unsubstituted or substituted.
  • Heterocycloalkyl groups can also include a double bond or a triple bond, such as, but not limited to dihydropyridine or 1,2,3,6-tetrahydropyridine.
  • the heterocycloalkyl groups can be linked via any position on the ring.
  • aziridine can be 1- or 2-aziridine
  • azetidine can be 1- or 2- azetidine
  • pyrrolidine can be 1-, 2- or 3 -pyrrolidine
  • piperidine can be 1-, 2-, 3- or 4-piperidine
  • pyrazolidine can be 1-, 2-, 3-, or 4-pyrazolidine
  • imidazolidine can be 1-, 2-, 3- or 4-imidazolidine
  • piperazine can be 1-, 2-, 3- or 4-piperazine
  • tetrahydrofuran can be 1- or 2-tetrahydrofuran
  • oxazolidine can be 2-, 3-, 4- or 5 -oxazolidine
  • isoxazolidine can be 2-, 3-, 4- or 5- isoxazolidine
  • thiazolidine can be 2-, 3-, 4- or 5 -thiazolidine
  • isothiazolidine can be 2-, 3-, 4- or 5- iso
  • heterocycloalkyl includes 3 to 8 ring members and 1 to 3 heteroatoms
  • representative members include, but are not limited to, pyrrolidine, piperidine, tetrahydrofuran, oxane, tetrahydrothiophene, thiane, pyrazolidine, imidazolidine, piperazine, oxazolidine, isoxzoalidine, thiazolidine, isothiazolidine, morpholine, thiomorpholine, dioxane and dithiane.
  • Heterocycloalkyl can also form a ring having 5 to 6 ring members and 1 to 2 heteroatoms, with representative members including, but not limited to, pyrrolidine, piperidine, tetrahydrofuran, tetrahydrothiophene, pyrazolidine, imidazolidine, piperazine, oxazolidine, isoxazolidine, thiazolidine, isothiazolidine, and morpholine.
  • hydrazinyl refers to two amino groups joined by a single bond, i.e., — N— N— .
  • the hydrazinyl group has optional substitution on at least one NH hydrogen to confer stability.
  • hydroxamic acid or its ester as used herein, refers to — C(O)ON(R)O(R'), wherein R and R are as defined herein, or the corresponding “hydroxamate” anion, including any corresponding hydroxamic acid salt.
  • hydroxyalkyl refers to a hydroxy group attached to the parent molecular moiety through an alkyl group.
  • “Hydroxyalkyl” or “alkylhydroxy” refers to an alkyl group, as defined above, where at least one of the hydrogen atoms is replaced with a hydroxy group.
  • hydroxyalkyl or alkylhydroxy groups can have any suitable number of carbon atoms, such as Ci-6.
  • Exemplary Ci-4 hydroxyalkyl groups include, but are not limited to, hydroxymethyl, hydroxyethyl (where the hydroxy is in the 1 or 2position), hydroxypropyl (where the hydroxy is in the 1, 2 or 3position), hydroxybutyl (where the hydroxy is in the 1, 2, 3 or 4position), l,2dihydroxyethyl, and the like.
  • isocyanate refers to a — NCO group.
  • isothiocyanate refers to a — NCS group.
  • linear chain of atoms refers to the longest straight chain of atoms independently selected from carbon, nitrogen, oxygen and sulfur.
  • linking group refers to any nitrogen containing organic fragment that serves to connect the pyrimidine or pyridone core of the compounds disclosed herein to the electrophilic moiety E, as defined herein.
  • exemplary linking groups include piperazines, aminoalkyls, alkyl- or aryl-based diamines, aminocycloalkyls, amine- containing spirocyclics, any of which may be optionally substituted as defined herein.
  • linking groups may comprise the substructure L-Q-L’-E wherein Q is a monocyclic 4 to 7 membered ring or a bicyclic, bridged, or fused, or spiro 6-11 membered ring, any of which optionally include one or more nitrogen atoms, E is the electrophilic group, L is bond, Ci-6 alkylene, — O — Co-5 alkylene, — S — Co-5 alkylene, or — NH — Co-5 alkylene, and for C2-6 alkylene, — O — C2-5 alkylene, — S — C2-5 alkylene, and NH — C2-5 alkylene, one carbon atom of any of the alkylene groups can optionally be replaced with O, S, or NH; and L’ is bond when Q comprises a nitrogen to link to E, otherwise L’ is NR, where R is hydrogen or alkyl.
  • lower means containing from 1 to and including 6 carbon atoms, or from 1 to 4 carbon atoms.
  • mercaptyl as used herein, alone or in combination, refers to an RS— group, where R is as defined herein.
  • nitro refers to — NO2.
  • perhaloalkoxy refers to an alkoxy group where all of the hydrogen atoms are replaced by halogen atoms.
  • perhaloalkyl refers to an alkyl group where all of the hydrogen atoms are replaced by halogen atoms.
  • phosphonate refers to a group of the form ROP(OR')(OR)O— wherein R and R' are selected from the group consisting of hydrogen, alkyl, acyl, heteroalkyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl, any of which may themselves be optionally substituted.
  • Phosphonate includes “phosphate [(OH)2P(O)O— ] and related phosphoric acid anions which may form salts.
  • sulfonate refers to the -SO3H group and its anion as the sulfonic acid is used in salt formation or sulfonate ester where OH is replaced by OR, where R is not hydrogen, but otherwise is as defined herein, and typically being alkyl or aryl.
  • thia and thio refer to a — S— group or an ether wherein the oxygen is replaced with sulfur.
  • the oxidized derivatives of the thio group namely sulfinyl and sulfonyl, are included in the definition of thia and thio.
  • thiol refers to an — SH group.
  • thiocyanate refers to a — CNS group.
  • trimethoxy refers to a X3CO— group where X is a halogen.
  • trimethysilyl as used herein, alone or in combination, refers to a silicone group substituted at its three free valences with groups as listed herein under the definition of substituted amino. Examples include trimethysilyl, tert-butyldimethylsilyl, triphenylsilyl and the like.
  • any one of the positions that is understood to have a hydrogen may also exist or understood to be isotopically enriched.
  • any atom not specifically designated as a particular isotope is meant to represent any stable isotope of that atom. Obtaining 100% deuteration at any relevant site of a compound in an amount of milligram or greater can be difficult.
  • a benzene ring may be optionally exist as -CeDs, -CeDH4, -C6D2H3, -C6D3H2, and -C6D4H.
  • a cyclohexyl group may optionally exist as -CeDn.
  • any definition herein may be used in combination with any other definition to describe a composite structural group.
  • the trailing element of any such definition is that which attaches to the parent moiety.
  • the composite group alkylamido would represent an alkyl group attached to the parent molecule through an amido group
  • the term alkoxyalkyl would represent an alkoxy group attached to the parent molecule through an alkyl group.
  • null When a group is defined to be “null,” what is meant is that said group is absent.
  • a “null” group occurring between two other group may also be understood to be a collapsing of flanking groups. For example, if in — (CH2) X G 1 G 2 G 3 , the element G 2 were null, said group would become — (CH2) X G 1 G 3 .
  • optionally substituted means the anteceding group or groups may be substituted or unsubstituted. Groups constituting optional substitution may themselves be optionally substituted. For example, where an alkyl group is embraced by an optional substitution, that alkyl group itself may also be optionally substituted.
  • the substituents of an “optionally substituted” group may include, without limitation, one or more substituents independently selected from the following groups or a particular designated set of groups, alone or in combination: alkyl, alkenyl, alkynyl, alkanoyl, heteroalkyl, heterocycloalkyl, haloalkyl, haloalkenyl, haloalkynyl, lower perhaloalkyl, perhaloalkoxy, cycloalkyl, phenyl, aryl, aryloxy, alkoxy, haloalkoxy, oxo, acyloxy, carbonyl, carboxyl, alkylcarbonyl, carboxyester, carboxamido, cyano, hydrogen, halogen, hydroxy, amino, alkylamino, arylamino, amido, nitro, thiol, alkylthio, haloalkylthio, perhaloalkylthio, arylthi
  • optional substitution include, without limitation: (1) alkyl, halo, and alkoxy; (2) alkyl and halo; (3) alkyl and alkoxy; (4) alkyl, aryl, and heteroaryl; (5) halo and alkoxy; and (6) hydroxyl, alkyl, halo, alkoxy, and cyano.
  • an optional substitution comprises a heteroatom-hydrogen bond (-NH-, SH, OH)
  • further optional substitution of the heteroatom hydrogen is contemplated and includes, without limitation optional substitution with alkyl, acyl, alkoxymethyl, alkoxyethyl, arylsulfonyl, alkyl sulfonyl, any of which are further optionally substituted.
  • Optionally substituted may include any of the chemical functional groups defined hereinabove and throughout this disclosure. Two optional substituents may be joined together to form a fused five-, six-, or seven-membered carbocyclic or heterocyclic ring consisting of zero to three heteroatoms, for example forming methylenedioxy or ethylenedioxy.
  • An optionally substituted group may be unsubstituted (e.g., — CH2CH3), fully substituted (e.g., — CF2CF3), monosubstituted (e.g., — CH2CH2F) or substituted at a level anywhere in-between fully substituted and monosubstituted (e.g., — CH2CF3).
  • a carbon chain may be substituted with an alkyl group, a halo group, and an alkoxy group.
  • substituents are recited without qualification as to substitution, both substituted and unsubstituted forms are encompassed.
  • substituent is qualified as “substituted,” the substituted form is specifically intended.
  • different sets of optional substituents to a particular moiety may be defined as needed; in these cases, the optional substitution will be as defined, often immediately following the phrase, “optionally substituted with.”
  • R or the term R' appearing by itself and without a number designation, unless otherwise defined, refers to a moiety selected from the group consisting of hydrogen, hydroxyl, halogen, alkyl, cycloalkyl, heteroalkyl, aryl, heteroaryl and heterocycloalkyl, any of which may be optionally substituted.
  • Each such R and R groups should be understood to be optionally substituted as defined herein.
  • every substituent, and every term should be understood to be independent of every other in terms of selection from a group. Should any variable, substituent, or term (e.g. aryl, heterocycle, R, etc.) occur more than one time in a formula or generic structure, its definition at each occurrence is independent of the definition at every other occurrence.
  • substituent, or term e.g. aryl, heterocycle, R, etc.
  • its definition at each occurrence is independent of the definition at every other occurrence.
  • certain groups may be attached to a parent molecule or may occupy a position in a chain of elements from either end as written. Thus, by way of example only, an unsymmetrical group such as — C(O)N(R)— may be atached to the parent moiety at either the carbon or the nitrogen.
  • Asymmetric centers, axial asymmetry (non-interchanging rotamers), or the like may exist in the compounds of the various embodiments disclosed herein. Such chirality may be designated by the symbols “R” or “S,” depending on the configuration of substituents around the chiral carbon atom or the relevant axis. It should be understood that embodiments encompasses all stereochemical isomeric forms, including diastereomeric, enantiomeric, and epimeric forms, d-isomers and 1-isomers, and mixtures thereof.
  • Individual stereoisomers of compounds can be prepared synthetically from commercially available starting materials which contain chiral centers or by preparation of mixtures of enantiomeric products followed by separation such as conversion to a mixture of diastereomers followed by separation or recrystallization, chromatographic techniques, direct separation of enantiomers on chiral chromatographic columns, or any other appropriate method known in the art.
  • Starting compounds of particular stereochemistry are either commercially available or can be made and resolved by techniques known in the art.
  • the compounds of the various embodiments disclosed herein may exist as geometric isomers.
  • the various embodiments disclosed herein includes all cis, trans, syn, anti,
  • E
  • Z tautomers
  • compounds may exist as tautomers, including keto-enol tautomers; all tautomeric isomers are embraced by the embodiments disclosed herein.
  • the compounds of the various embodiments disclosed herein can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • the solvated forms are considered equivalent to the unsolvated forms for the purposes of the various embodiments disclosed herein.
  • bond refers to a covalent linkage between two atoms, or two moieties when the atoms joined by the bond are considered part of larger substructure.
  • a bond may be single, double, or triple unless otherwise specified.
  • a dashed line between two atoms in a drawing of a molecule indicates that an additional bond may be present or absent at that position.
  • the compounds disclosed herein can exist as pharmaceutically acceptable salts, including acid addition salts. Suitable salts include those formed with both organic and inorganic acids. Such acid addition salts will normally be pharmaceutically acceptable. However, salts of non-pharmaceutically acceptable salts may be of utility in the preparation and purification of the compound in question. Basic addition salts may also be formed and be pharmaceutically acceptable. For a more complete discussion of the preparation and selection of salts, refer to Pharmaceutical Salts: Properties, Selection, and Use (Stahl, P. Heinrich. Wiley-VCHA, Zurich, Switzerland, 2002). It is understood that each of the compounds disclosed herein, and each embodiment of the compounds set forth herein, include pharmaceutically acceptable salts of such compounds.
  • the salts can be prepared during the final isolation and purification of the compounds or separately by reacting the appropriate compound in the form of the free base with a suitable acid.
  • Representative acid addition salts include acetate, adipate, alginate, L-ascorbate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, butyrate, camphorate, camphorsulfonate, citrate, digluconate, formate, fumarate, gentisate, glutarate, glycerophosphate, glycolate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethansulfonate (isethionate), lactate, maleate, malonate, DL-mandelate, mesitylene sulfonate, methane sulfonate, naphthylenesulfonate, nicotinate, 2- naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-
  • basic groups in the compounds of the various embodiments disclosed herein can be quatemized with methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides; dimethyl, diethyl, dibutyl, and diamyl sulfates; decyl, lauryl, myristyl, and steryl chlorides, bromides, and iodides; and benzyl and phenethyl bromides.
  • acids which can be employed to form pharmaceutically acceptable addition salts include inorganic acids such as hydrochloric, hydrobromic, sulfuric, and phosphoric, and organic acids such as oxalic, maleic, succinic, and citric.
  • Salts can also be formed by coordination of the compounds with an alkali metal or alkaline earth ion.
  • the various embodiments disclosed herein contemplates sodium, potassium, magnesium, and calcium salts of the compounds disclosed herein, and the like.
  • Basic addition salts can be prepared during the final isolation and purification of the compounds by reacting a carboxyl group with a suitable base such as the hydroxide, carbonate, or bicarbonate of a metal cation or with ammonia or an organic primary, secondary, or tertiary amine.
  • the cations of pharmaceutically acceptable salts include lithium, sodium, potassium, calcium, magnesium, and aluminum, as well as nontoxic quaternary amine cations such as ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, diethylamine, ethylamine, tributylamine, pyridine, N,N-dimethylaniline, N- methylpiperidine, N-methylmorpholine, dicyclohexylamine, procaine, dibenzylamine, N,N-dibenzylphenethylamine, 1-ephenamine, and N,N'-dibenzylethylenediamine.
  • Other representative organic amines useful for the formation of base addition salts include ethylenediamine, ethanolamine, diethanolamine, piperidine, and piperazine.
  • disease as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder” and “condition” (as in medical condition), in that all reflect an abnormal condition of the body or of one of its parts that impairs normal functioning and is typically manifested by distinguishing signs and symptoms.
  • cancer refers to all types of cancer, neoplasm or malignant tumors found in mammals (e.g., but not limited to, humans), including leukemia, lymphomas, carcinomas and sarcomas.
  • Exemplary cancers that may be treated with a compound or method provided herein include cancer of the thyroid, endocrine system, brain, breast, cervix, colon, head & neck, liver, kidney, lung, non-small cell lung, melanoma, mesothelioma, ovary, sarcoma, stomach, uterus, Medulloblastoma, colorectal cancer, pancreatic cancer.
  • Additional examples include, Hodgkin's Disease, NonHodgkin's Lymphoma, multiple myeloma, neuroblastoma, glioma, glioblastoma multiforme, ovarian cancer, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, primary brain tumors, cancer, malignant pancreatic insulanoma, malignant carcinoid, urinary bladder cancer, premalignant skin lesions, testicular cancer, lymphomas, thyroid cancer, neuroblastoma, esophageal cancer, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenal cortical cancer, neoplasms of the endocrine or exocrine pancreas, medullary thyroid cancer, medullary thyroid carcinoma, melanoma, colorectal cancer, papillary thyroid cancer, hepatocellular carcinoma, or prostate cancer.
  • leukemia refers broadly to progressive, malignant diseases of the blood- forming organs and is generally characterized by a distorted proliferation and development of leukocytes and their precursors in the blood and bone marrow. Leukemia is generally clinically classified on the basis of (1) the duration and character of the disease-acute or chronic; (2) the type of cell involved; myeloid (myelogenous), lymphoid (lymphogenous), or monocytic; and (3) the increase or non-increase in the number abnormal cells in the blood -leukemic or aleukemic (subleukemic).
  • Exemplary leukemias that may be treated with a compound or method provided herein include, for example, acute nonlymphocytic leukemia, chronic lymphocytic leukemia, acute granulocytic leukemia, chronic granulocytic leukemia, acute promyelocytic leukemia, adult T-cell leukemia, aleukemic leukemia, a leukocythemic leukemia, basophylic leukemia, blast cell leukemia, bovine leukemia, chronic myelocytic leukemia, leukemia cutis, embryonal leukemia, eosinophilic leukemia, Gross' leukemia, hairy-cell leukemia, hemoblastic leukemia, hemocytoblastic leukemia, histiocytic leukemia, stem cell leukemia, acute monocytic leukemia, leukopenic leukemia, lymphatic leukemia, lymphoblastic leukemia, lymphocytic leukemia, lymphogenous leukemia,
  • lymphoma refers to a group of cancers affecting hematopoietic and lymphoid tissues. It begins in lymphocytes, the blood cells that are found primarily in lymph nodes, spleen, thymus, and bone marrow. Two main types of lymphoma are non-Hodgkin lymphoma and Hodgkin’s disease. Hodgkin’s disease represents approximately 15% of all diagnosed lymphomas. This is a cancer associated with Reed-Sternberg malignant B lymphocytes. Non-Hodgkin’s lymphomas (NHL) can be classified based on the rate at which cancer grows and the type of cells involved.
  • B-cell lymphomas that may be treated with a compound or method provided herein include, but are not limited to, small lymphocytic lymphoma, Mantle cell lymphoma, follicular lymphoma, marginal zone lymphoma, extranodal (MALT) lymphoma, nodal (monocytoid B- cell) lymphoma, splenic lymphoma, diffuse large cell B -lymphoma, Burkitt’s lymphoma, lymphoblastic lymphoma, immunoblastic large cell lymphoma, or precursor B -lymphoblastic lymphoma.
  • T-cell lymphomas that may be treated with a compound or method provided herein include, but are not limited to, cunateous T-cell lymphoma, peripheral T-cell lymphoma, anaplastic large cell lymphoma, mycosis fungoides, and precursor T- lymphoblastic lymphoma.
  • sarcoma generally refers to a tumor which is made up of a substance like the embryonic connective tissue and is generally composed of closely packed cells embedded in a fibrillar or homogeneous substance.
  • Sarcomas that may be treated with a compound or method provided herein include a chondrosarcoma, fibrosarcoma, lymphosarcoma, melanosarcoma, myxosarcoma, osteosarcoma, Abernethy's sarcoma, adipose sarcoma, liposarcoma, alveolar soft part sarcoma, ameloblastic sarcoma, botryoid sarcoma, chloroma sarcoma, chorio carcinoma, embryonal sarcoma, Wilms' tumor sarcoma, endometrial sarcoma, stromal sarcoma, Ewing's sarcoma, fascial sarcom
  • melanoma is taken to mean a tumor arising from the melanocytic system of the skin and other organs.
  • Melanomas that may be treated with a compound or method provided herein include, for example, acral -lentiginous melanoma, amelanotic melanoma, benign juvenile melanoma, Cloudman's melanoma, S91 melanoma, Harding- Passey melanomajuvenile melanoma, lentigo maligna melanoma, malignant melanoma, nodular melanoma, subungal melanoma, or superficial spreading melanoma.
  • carcinoma refers to a malignant new growth made up of epithelial cells tending to infiltrate the surrounding tissues and give rise to metastases.
  • exemplary carcinomas that may be treated with a compound or method provided herein include, for example, medullary thyroid carcinoma, familial medullary thyroid carcinoma, acinar carcinoma, acinous carcinoma, adenocystic carcinoma, adenoid cystic carcinoma, carcinoma adenomatosum, carcinoma of adrenal cortex, alveolar carcinoma, alveolar cell carcinoma, basal cell carcinoma, carcinoma basocellulare, basaloid carcinoma, basosquamous cell carcinoma, bronchioalveolar carcinoma, bronchiolar carcinoma, bronchogenic carcinoma, cerebriform carcinoma, cholangiocellular carcinoma, chorionic carcinoma, colloid carcinoma, comedo carcinoma, corpus carcinoma, cribriform carcinoma, carcinoma en cuirasse, carcinoma cutaneum, cylindrical carcinoma, cylindrical cell carcinoma, duct carcinoma, carcinoma durum, embryonal carcinoma, encephaloid
  • Ras associated cancer refers to a cancer caused by aberrant Ras activity or signaling.
  • A”cancer associated with aberrant K-Ras activity is a cancer caused by aberrant K-Ras activity or signaling (e.g. a mutant K-Ras).
  • K-Ras related cancers may include lung cancer, non- small cell lung cancer, breast cancer, leukemia, pancreatic cancer, colon cancer, colorectal cancer.
  • cancers that are associated with aberrant activity of one or more of Ras, K-Ras, H- Ras, N-Ras, mutant K-Ras (including K-Ras G12C, K-Ras G12V, K-Ras G13C, K-Ras G12D, K-Ras G13D mutants), mutant N-Ras, and mutant H-Ras are well known in the art, including G12C in both N-Ras and H-Ras, and determining such cancers are within the skill of a person of skill in the art.
  • administering means administering a compound that inhibits the activity or level (e.g. amount) or level of a signaling pathway of one or more Ras proteins (e.g. a Ras inhibitor, K-Ras inhibitor, N- Ras inhibitor, H-Ras inhibitor, mutant K-Ras inhibitor, K-Ras G12C inhibitor, K-Ras G12V inhibitor, K-Ras G13C inhibitor, K-Ras G12D inhibitor, K-Ras G13D inhibitor) to a subject.
  • a Ras inhibitor e.g. a Ras inhibitor, K-Ras inhibitor, N- Ras inhibitor, H-Ras inhibitor, mutant K-Ras inhibitor, K-Ras G12C inhibitor, K-Ras G12V inhibitor, K-Ras G13C inhibitor, K-Ras G12D inhibitor, K-Ras G13D inhibitor
  • Administration may include, without being limited by mechanism, allowing sufficient time for the Ras inhibitor to reduce the activity of one or more Ras proteins or for the Ras inhibitor to reduce one or more symptoms of a disease (e.g. cancer, wherein the Ras inhibitor may arrest the cell cycle, slow the cell cycle, reduce DNA replication, reduce cell replication, reduce cell growth, reduce metastasis, or cause cell death).
  • a K-Ras inhibitor means administering a compound that inhibits the activity or level (e.g.
  • the administering does not include administration of any active agent other than the recited active agent.
  • a disease e.g. Ras (e.g., human K-Ras or human H-Ras) activity, a protein associated disease, a cancer associated with aberrant Ras activity, K-Ras associated cancer, mutant K-Ras associated cancer, activated K-Ras associated cancer, K-RasG12C associated cancer, K-Ras G12V associated cancer, K-Ras G13C associated cancer, K-Ras G12D associated cancer, K-Ras G13D associated cancer) means that the disease (e.g.
  • a cancer associated with aberrant Ras activity or function may be a cancer that results (entirely or partially) from aberrant Ras activity or function (e.g. enzyme activity, protein-protein binding, signaling pathway) or a cancer wherein a particular symptom of the disease is caused (entirely or partially) by aberrant Ras activity or function.
  • aberrant Ras activity or function e.g. enzyme activity, protein-protein binding, signaling pathway
  • a cancer wherein a particular symptom of the disease is caused (entirely or partially) by aberrant Ras activity or function.
  • what is described as being associated with a disease if a causative agent, could be a target for treatment of the disease .
  • a cancer associated with aberrant Ras activity or function or a Ras associated cancer may be treated with a Ras modulator or Ras inhibitor, in the instance where increased Ras activity or function (e.g., signaling pathway activity) causes the cancer.
  • a cancer associated with K-Ras G12C may be a cancer that a subject with K-Ras G12C is at higher risk of developing as compared to a subject without K-Ras G12C.
  • a cancer associated with K-Ras G12V may be a cancer that a subject with K-Ras G 12V is at higher risk of developing as compared to a subject without K-Ras G12V.
  • Ras GTPase proteins e.g. K-Ras, H-Ras, N-Ras.
  • K-Ras refers to the nucleotide sequences or proteins of human K-Ras (e.g. human K-Ras4A (NP 203524.1), human K-Ras4B (NP 004976.2), or both K-Ras4A and K-Ras4B).
  • K-Ras includes both the wild-type form of the nucleotide sequences or proteins as well as any mutants thereof.
  • K-Ras is wild- type K-Ras. In some embodiments, “K-Ras” is one or more mutant forms.
  • K-Ras” XYZ refers to a nucleotide sequence or protein of a mutant K-Ras wherein the Y numbered amino acid of K-Ras that has an X amino acid in the wildtype instead has a Z amino acid in the mutant (e.g. K-Ras G12C has a G in wildtype protein but a C in the K-Ras G12C mutantprotein).
  • K-Ras refers to K-Ras4A and K-Ras4B.
  • K-Ras refers to K-Ras4A. In some embodiments, K-Ras refers to K-Ras4B (e.g., NM_004985.4 or NP_004976.2). In some embodiments, K-Ras refers to the protein including (e.g., consisting of) the amino acid sequence below or including the sequence below with one or more mutations (e.g., G12C, G12V, or G13C):
  • K-Ras refers to the protein including (e.g., consisting of) the amino acid sequence below or including (e.g., consisting of) the sequence below with one or more mutations (e.g., G12C, G12V, or G13C):
  • Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients or in multiple, separate capsules for each active ingredient.
  • administration also encompasses use of each type of therapeutic agent in a sequential manner. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.
  • K-RAS inhibitor is used herein to refer to a compound that exhibits an IC50 with respect to K-RAS activity of no more than about 100 mM and more typically not more than about 50 mM, as measured in the K-RAS assay described generally hereinbelow. “IC50” is that concentration of inhibitor that reduces the activity of an enzyme (e.g., K-RAS) to half-maximal level.
  • an enzyme e.g., K-RAS
  • compounds will exhibit an IC50 with respect to oncogenic mutant K-RAS of no more than about 10 mM; in further embodiments, compounds will exhibit an IC50 with respect to K-RAS of no more than about 5 mM; in yet further embodiments, compounds will exhibit an IC50 with respect to K-RAS of not more than about 1 mM, as measured in the K-RAS assay described herein. In yet further embodiments, compounds will exhibit an IC50 with respect to K-RAS of not more than about 200 nM.
  • the K-RAS inhibitor is an irreversible inhibitor by way of covalent bond formation to the cysteine at the G 12C mutation site.
  • the phrase “therapeutically effective” is intended to qualify the amount of active ingredients used in the treatment of a disease or disorder. This amount will achieve the goal of reducing or eliminating the the disease or disorder.
  • treatment of a subject is intended to include prophylaxis.
  • subject means all mammals, including humans. Examples of subjects include humans, cows, dogs, cats, goats, sheep, pigs, and rabbits. In some embodiments, the subject is a human.
  • prodrug refers to a compound that is made active in vivo through chemical reaction in vivo thereby releasing an active compound.
  • Compounds disclosed herein can be modified to exist as prodrugs, as described in Hydrolysis in Drug and Prodrug Metabolism: Chemistry, Biochemistry, and Enzymology (Testa, Bernard and Mayer, Joachim M. Wiley-VHCA, Zurich, Switzerland 2003).
  • Prodrugs of the compounds described herein are structurally modified forms of the compound that readily undergo chemical changes under physiological conditions to provide the active compound. Additionally, prodrugs can be converted to the active compounds by chemical or biochemical methods in an ex vivo environment.
  • prodrugs can be slowly converted to a compound when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • Prodrugs are often useful because, in some situations, they may be easier to administer than the active compound, or parent drug. They may, for instance, be bioavailable by oral administration whereas the parent drug is not.
  • the prodrug may also have improved solubility in pharmaceutical compositions over the parent drug.
  • a wide variety of prodrug derivatives are known in the art, such as those that rely on hydrolytic cleavage or oxidative activation of the prodrug.
  • prodrug is a compound which is administered as an ester (the “prodrug”), which is then metabolically hydrolyzed to the carboxylic acid, as the active entity. Additional examples include peptidyl derivatives of a compound.
  • therapeutically acceptable prodrug refers to those prodrugs or zwitterions which are suitable for use in contact with the tissues of subjects without undue toxicity, irritation, and allergic response, are commensurate with a reasonable benefit/risk ratio, and are effective for their intended use.
  • A is alkyl, alkenyl, alkynyl, alkoxy, cycloalkoxy, cyano, hydrogen, trifluoromethyl, trifluoromethoxy, halogen, -SF5, cycloalkyl, cycloalkenyl, bicycloalkyl, tricycloalkyl, tetracycloalkyl, or pentacycloalkyl, wherein when A comprises one or more C-H groups, one or more carbon atom is optionally substituted with 1 to 3 F, Cl, alkyl, or CF3; and any of which optionally replace 1 or 2 carbon atoms with a heteroatom selected from NR, O, or S; wherein R is H or Me;
  • X is O or S(O) P , wherein p is an integer from 0 to 2;
  • R 2 is selected from the group consisting of:
  • each R’ or R is selected from the group consisting of hydrogen, alkyl, alkoxy, alkoxyalkyl, hydroxy, cycloalkoxy, cyano, cyanoalkyl, amido, amidoalkyl, N-alkylamido, N-alkylamidoalkyl, N,N- dialkylamido, N,N-dialkylamidoalkyl, amino, aminoalkyl, N-alkyl amino, N-alkyl aminoalkyl, N,N-dialkylamino, and N,N -dialkylaminoalkyl, any of which are optionally substituted; or any two R’ and R” combine to form 3-7-membered ring, optionally comprising 1 or 2 heteroatoms selected from N, O, or S; or any two R 2 combine to form a spirocycle comprising 0 to 2 heteroatoms selected from
  • R 2 is aryl, heteroaryloxy, heteroaryl, or N-linked heteroaryl any of which are optionally substituted with 1 to 4 halogens.
  • A is alkyl, alkenyl, alkynyl, alkoxy, cycloalkoxy, cyano, hydrogen, trifluoromethyl, trifluoromethoxy, halogen, or - SFs, wherein when A comprises one or more C-H groups, one or more carbon atoms is optionally substituted with F, Cl, alkyl, or CF3.
  • A is halogen.
  • the halogen is F, Cl, Br, or I.
  • the halogen is Cl.
  • A is cycloalkyl, cycloalkenyl, bicycloalkyl, tricycloalkyl, tetracycloalkyl, or pentacycloalkyl, any of which comprise one or more carbon atoms optionally substituted with F, Cl, alkyl, or CF3; and any of which optionally replace 1 or 2 carbon atoms with a heteroatom selected from NR, O, or S; wherein R is H or CH3.
  • A is selected from: wherein Z is NH, NMe, O, or S; each R 4 is independently H, Me, F, Cl, or CF3; and p is 1 or 2.
  • R 2 is selected from:
  • R' is H, CH3, F, Cl, or CF3; and, wherein R is
  • compositions which comprise one or more of the compounds disclosed herein, or one or more pharmaceutically acceptable salts, esters, prodrugs, amides, or solvates thereof, together with one or more pharmaceutically acceptable carriers and optionally one or more other therapeutic ingredients.
  • the carrier(s) should be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Proper formulation is dependent upon the route of administration chosen.
  • compositions disclosed herein may be manufactured in any manner known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or compression processes.
  • the pharmaceutical compositions may include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous, intraarticular, and intramedullary), intraperitoneal, transmucosal, transdermal, rectal and topical (including dermal, buccal, sublingual and intraocular) administration although the most suitable route may depend upon for example the condition and disorder of the recipient.
  • the pharmaceutical composition may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Typically, these methods include the step of bringing into association a compound disclosed herein or a pharmaceutically acceptable salt, ester, amide, prodrug or solvate thereof (“active ingredient”) with the carrier which constitutes one or more accessory ingredients.
  • active ingredient a pharmaceutically acceptable salt, ester, amide, prodrug or solvate thereof
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation
  • compositions of the various embodiments disclosed herein suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • compositions that can be used orally include tablets.
  • Tablets may be made by compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration.
  • the compounds disclosed herein may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in powder form or in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or sterile pyrogen-free water, immediately prior to use.
  • sterile liquid carrier for example, saline or sterile pyrogen-free water
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • Formulations for parenteral administration include aqueous and non-aqueous (oily) sterile injection solutions of the active compounds which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
  • the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the compounds disclosed herein may be administered orally or via injection at a dose of from 0.1 to 500 mg/kg per day.
  • a common dose range for adult humans is generally from 5 mg to 2 g/day.
  • Tablets or other forms of presentation provided in discrete units may conveniently contain an amount of one or more compounds which is effective at such dosage or as a multiple of the same, for instance, units containing 5 mg to 500 mg, usually around 10 mg to 200 mg.
  • the amount of active ingredient that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated and the particular mode of administration.
  • the compounds disclosed herein can be administered in various modes, e.g. orally, topically, or by injection.
  • the precise amount of compound administered to a subject will be the responsibility of the attendant physician.
  • the specific dose level for any particular subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diets, time of administration, route of administration, rate of excretion, drug combination, the precise disorder being treated, and the severity of the indication or condition being treated.
  • the route of administration may vary depending on the condition and its severity.
  • the multiple therapeutic agents may be administered in any order or even simultaneously. If simultaneously, the multiple therapeutic agents may be provided in a single, unified form, or in multiple forms (by way of example only, either as a single pill or as two separate pills). One of the therapeutic agents may be given in multiple doses, or both may be given as multiple doses. If not simultaneous, the timing between the multiple doses may be any duration of time ranging from a few minutes to four weeks.
  • embodiments herein provide methods for treating K- RAS-mediated disorders in a human or animal subject in need of such treatment comprising administering to said subject an amount of a compound of the various embodiments disclosed herein effective to reduce or prevent said disorder in the subject in combination with at least one additional agent for the treatment of said disorder that is known in the art.
  • the various embodiments disclosed herein provides therapeutic compositions comprising at least one compound of the various embodiments disclosed herein in combination with one or more additional agents for the treatment of K- RAS-mediated disorders.
  • the K-RAS-mediated disease is cancer and the K-RAS presents in an oncogenic mutated form.
  • embodiments herein provide methods for treating a subject with cancer comprising administering to the subject a compound disclosed herein, or pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
  • the cancer comprises a K-Ras G12 mutation.
  • the G12 mutation is G12C.
  • the cancer is one or more of pancreatic, lung, and colorectal cancer.
  • the pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC).
  • the cancer is a CNS cancer.
  • the CNS cancer is a primary cancer.
  • the primary cancer comprises one or more of a glioma, meningioma, medulloblastoma, ganblioglioma, schwannoma, and craniopharyngioma.
  • the glioma comprises one or more of an astrocytoma, a glioblastoma, an oligodendroglioma, and a ependymoma.
  • the CNS cancer comprises a metastatic or secondary cancer.
  • the CNS cancer comprises a cancer metastasized from one or more of melanoma, breast cancer, colon cancer, kidney cancer, nasopharyngeal cancer, leukemia, lymphoma, myeloma, and other of unknown primary site.
  • the present embodiments provide for the use of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as disclosed herein, for the treatment of cancer, the cancer characterized by the presence of a KRAS G12C mutation.
  • the present embodiments provide for the use of a compound disclosed herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as disclosed herein, for the manufacture of a medicament for the treatment of cancer in a subject, the cancer characterized by the presence of a KRAS G12C mutation.
  • the present embodiments provide a method of preventing or reducing the spread of cancer via CNS pathways comprising administering to the subject a compound disclosed herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition as disclosed herein.
  • Compounds disclosed herein may be used in combination therapies.
  • the compounds disclosed herein may be used in combination with inhibitors of mammalian target of rapamycin (mTOR), insulin growth factor 1 receptor (IGF1R), and combinations thereof.
  • mTOR mammalian target of rapamycin
  • IGF1R insulin growth factor 1 receptor
  • Such combination therapies may be particularly suited to certain cancer types such as lung cancer. See Molinas-Arcas et al. Sci. Trans. Med. 18 Sep. 2019 11:510 eaaw7999 at stm.sciencemag.org/content/l l/510/eaaw7999.
  • Compounds disclosed herein may be combined with modulators the ULK family of proteins, which regulate autophagy.
  • Other compounds of interest in combination therapy include inhibitors of SHP2.
  • SHP2 inhibitors include those disclosed in W02016/203404, WO2018/136264, WO2018/057884, WO2019/067843, WO2019/183367, W02016/203405, W02019/051084, W02018/081091, WO2019/165073,
  • compounds disclosed herein may be combined with an EGFR inihibitor.
  • the EGFR inhibitor is selective for a mutant EGFR, including, without limitation, C797X, L718Q, G724S, S768I, G719X, L792X, G796X, T263P, A289D/V, G598V, and EGFRvIII high expression.
  • the combination therapy with EGFR agents tracked by mutation and indication are shown in Table CT-1 below.
  • EGFR inhibitors include those disclosed in US Pat. Nos. 5,747,498, 8,946,235, and 9,732,058, W02002030926, US 20040048880, US20050165035, and WO2019067543. All patents and applications, including compositions, methods of using, and methods of making compounds disclosed therein are incorporated herein by reference in their entirety.
  • the second agent of the pharmaceutical combination formulation or dosing regimen may have complementary activities to the compounds disclosed herein such that they do not adversely affect each other.
  • the compounds may be administered together in a unitary pharmaceutical composition or separately.
  • a compound or a pharmaceutically acceptable salt can be co-administered with a cytotoxic agent to treat proliferative diseases and cancer.
  • co-administering refers to either simultaneous administration, or any manner of separate sequential administration, of a compound disclosed herein or a salt thereof, and a further active pharmaceutical ingredient or ingredients, including cytotoxic agents and radiation treatment. If the administration is not simultaneous, the compounds are administered in a close time proximity to each other. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered topically and another compound may be administered orally.
  • Those additional agents may be administered separately from an inventive compound-containing composition, as part of a multiple dosage regimen.
  • those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
  • compositions of this invention are formulated such that a dosage of between 0.01 - 100 mg/kg body weight/day of an inventive can be administered.
  • any agent that has activity against a disease or condition being treated may be co-administered.
  • agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Hellman (editors), 6 th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers.
  • a person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the disease involved.
  • the treatment method includes the co-administration of a compound disclosed herein or a pharmaceutically acceptable salt thereof and at least one cytotoxic agent.
  • cytotoxic agent refers to a substance that inhibits or prevents a cellular function and/or causes cell death or destruction.
  • Cytotoxic agents include, but are not limited to, radioactive isotopes (e.g., At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu); chemotherapeutic agents; growth inhibitory agents; enzymes and fragments thereof such as nucleolytic enzymes; and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof.
  • radioactive isotopes e.g., At 211 , 1 131 , 1 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 and radioactive isotopes of Lu
  • chemotherapeutic agents e.g., At 211 , 1 131 , 1 125
  • Exemplary cytotoxic agents can be selected from anti-microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor tyrosine kinase angiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, inhibitors of LDH-A; inhibitors of fatty acid biosynthesis; cell cycle signalling inhibitors; HDAC inhibitors, proteasome inhibitors; and inhibitors of cancer metabolism.
  • “Chemotherapeutic agent” includes chemical compounds useful in the treatment of cancer.
  • chemotherapeutic agents include erlotinib (TARCEVA®, Genentech/OSI Pharm.), bortezomib (VELCADE®, Millennium Pharm.), disulfiram , epigallocatechin gallate , salinosporamide A, carfilzomib, 17-AAG(geldanamycin), radicicol, lactate dehydrogenase A (LDH-A), fulvestrant (FASLODEX®, AstraZeneca), sunitib (SUTENT®, Pfizer/Sugen), letrozole (FEMARA®, Novartis), imatinib mesylate (GLEEVEC®., Novartis), finasunate (VATALANIB®, Novartis), oxaliplatin (ELOXATIN®, Sanofi), 5-FU (5 -fluorouracil), leucovorin,
  • dynemicin including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antibiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxy doxorubicin), epirubicin,
  • GEMZAR® (gemcitabine); 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; etoposide (VP- 16); ifosfamide; mitoxantrone; vincristine; NAVELBINE® (vinorelbine); novantrone; teniposide; edatrexate; daunomycin; aminopterin; capecitabine (XELODA®); ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylomithine (DMFO); retinoids such as retinoic acid; and pharmaceutically acceptable salts, acids and derivatives of any of the above.
  • Chemotherapeutic agent also includes (i) anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti -estrogens and selective estrogen receptor modulators (SERMs), including, for example, tamoxifen (including NOLVADEX®; tamoxifen citrate), raloxifene, droloxifene, iodoxyfene , 4- hydroxytamoxifen, trioxifene, keoxifene, LY117018, onapristone, and FARESTON® (toremifine citrate); (ii) aromatase inhibitors that inhibit the enzyme aromatase, which regulates estrogen production in the adrenal glands, such as, for example, 4(5)-imidazoles, aminoglutethimide, MEGASE® (megestrol acetate), AROMASIN® (exemestane; Pfizer), formestanie, fadrozole, RIVISOR® (vorozole),
  • Chemotherapeutic agent also includes antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen pie), pertuzumab (OMNITARG®, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth).
  • antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RIT
  • Additional humanized monoclonal antibodies with therapeutic potential as agents in combination with the compounds of the invention include: apolizumab, aselizumab, atlizumab, bapineuzumab, bivatuzumab mertansine, cantuzumab mertansine, cedelizumab, certolizumab pegol, cidfusituzumab, cidtuzumab, daclizumab, eculizumab, efalizumab, epratuzumab, erlizumab, felvizumab, fontolizumab, gemtuzumab ozogamicin, inotuzumab ozogamicin, ipilimumab, labetuzumab, lintuzumab, matuzumab, mepolizumab, motavizumab, motovizumab, natalizumab, nimotuzumab, nolovizum
  • Chemotherapeutic agent also includes “EGFR inhibitors,” which refers to compounds that bind to or otherwise interact directly with EGFR and prevent or reduce its signaling activity, and is alternatively referred to as an “EGFR antagonist.”
  • EGFR inhibitors refers to compounds that bind to or otherwise interact directly with EGFR and prevent or reduce its signaling activity
  • Examples of such agents include antibodies and small molecules that bind to EGFR.
  • antibodies which bind to EGFR include MAb 579 (ATCC CRL HB 8506), MAb 455 (ATCC CRL HB8507), MAb 225 (ATCC CRL 8508), MAb 528 (ATCC CRL 8509) (see, US Patent No.
  • EMD 55900 Stragliotto et al. Eur. J. Cancer 32A:636-640 (1996)
  • EMD7200 (matuzumab) a humanized EGFR antibody directed against EGFR that competes with both EGF and TGF-alpha for EGFR binding (EMD/Merck); human EGFR antibody, HuMax-EGFR (GenMab); fully human antibodies known as ELI, E2.4, E2.5, E6.2, E6.4, E2.l l, E6. 3 and E7.6.
  • the anti-EGFR antibody may be conjugated with a cytotoxic agent, thus generating an immunoconjugate (see, e.g., EP659,439A2, Merck Patent GmbH).
  • EGFR antagonists include small molecules such as compounds described in US Patent Nos: 5,616,582, 5,457,105, 5,475,001, 5,654,307, 5,679,683, 6,084,095, 6,265,410, 6,455,534, 6,521,620, 6,596,726, 6,713,484, 5,770,599, 6,140,332, 5,866,572, 6,399,602, 6,344,459, 6,602,863, 6,391,874, 6,344,455, 5,760,041, 6,002,008, and 5,747,498, as well as the following PCT publications: WO98/14451, W098/50038, W099/09016, and WO99/24037.
  • EGFR antagonists include OSI-774 (CP-358774, erlotinib, TARCEVA® Genentech/OSI Pharmaceuticals); PD 183805 (CI 1033, 2- propenamide, N-[4-[(3-chloro-4-fluorophenyl)amino]-7-[3-(4-morpholinyl)propoxy]-6- quinazolinyl]-, dihydrochloride, Pfizer Inc.); ZD1839, gefitinib (IRESSA®) 4-(3’-Chloro- 4 ’ -fluoroanilino)-7 -methoxy-6-(3 -morpholinopropoxy)quinazoline, AstraZeneca); ZM 105180 ((6-amino-4-(3-methylphenyl-amino)-quinazoline, Zeneca); BIBX-1382 (N8-(3- chloro-4-fluoro-phenyl)-N2-(l-
  • Chemotherapeutic agents also include “tyrosine kinase inhibitors” including the EGFR-targeted drugs noted in the preceding paragraph; small molecule HER2 tyrosine kinase inhibitor such as TAK165 available from Takeda; CP-724,714, an oral selective inhibitor of the ErbB2 receptor tyrosine kinase (Pfizer and OSI); dual-HER inhibitors such as EKB-569 (available from Wyeth) which preferentially binds EGFR but inhibits both HER2 and EGFR-overexpressing cells; lapatinib (GSK572016; available from GlaxoSmithKline), an oral HER2 and EGFR tyrosine kinase inhibitor; PKI-166 (available from Novartis); pan-HER inhibitors such as canertinib (CI-1033; Pharmacia); Raf-1 inhibitors such as antisense agent ISIS-5132 available from ISIS Pharmaceuticals which inhibit Raf- 1 signaling; non-
  • Chemotherapeutic agents also include dexamethasone, interferons, colchicine, metoprine, cyclosporine, amphotericin, metronidazole, alemtuzumab, alitretinoin, allopurinol, amifostine, arsenic trioxide, asparaginase, BCG live, bevacuzimab, bexarotene, cladribine, clofarabine, darbepoetin alfa, denileukin, dexrazoxane, epoetin alfa, elotinib, filgrastim, histrelin acetate, ibritumomab, interferon alfa-2a, interferon alfa- 2b, lenalidomide, levamisole, mesna, methoxsalen, nandrolone, nelarabine, nofetumomab, opre
  • Chemotherapeutic agents also include hydrocortisone, hydrocortisone acetate, cortisone acetate, tixocortol pivalate, triamcinolone acetonide, triamcinolone alcohol, mometasone, amcinonide, budesonide, desonide, fluocinonide, fhiocinolone acetonide, betamethasone, betamethasone sodium phosphate, dexamethasone, dexamethasone sodium phosphate, fluocortolone, hydrocortisone- 17-butyrate, hydrocortisone- 17-valerate, aclometasone dipropionate, betamethasone valerate, betamethasone dipropionate, prednicarbate, clobetasone- 17-butyrate, clobetasol-17-propionate, fluocortolone caproate, fluocortolone pivalate and flupredn
  • celecoxib or etoricoxib proteosome inhibitor
  • CCI-779 tipifamib (R11577); orafenib, ABT510
  • Bcl-2 inhibitor such as oblimersen sodium (GENASENSE®)
  • pixantrone famesyltransferase inhibitors such as lonafamib (SCH 6636, SARASARTM)
  • pharmaceutically acceptable salts, acids or derivatives of any of the above as well as combinations of two or more of the above such as CHOP, an abbreviation for a combined therapy of cyclophosphamide, doxorubicin, vincristine, and prednisolone
  • FOLFOX an abbreviation for a treatment regimen with oxaliplatin (ELOXATIN TM) combined with 5-FU and leucovorin.
  • Chemotherapeutic agents also include non-steroidal anti-inflammatory drugs with analgesic, antipyretic and anti-inflammatory effects.
  • NSAIDs include non-selective inhibitors of the enzyme cyclooxygenase.
  • Specific examples of NSAIDs include aspirin, propionic acid derivatives such as ibuprofen, fenoprofen, ketoprofen, flurbiprofen, oxaprozin and naproxen, acetic acid derivatives such as indomethacin, sulindac, etodolac, diclofenac, enolic acid derivatives such as piroxicam, meloxicam, tenoxicam, droxicam, lomoxicam and isoxicam, fenamic acid derivatives such as mefenamic acid, meclofenamic acid, flufenamic acid, tolfenamic acid, and COX-2 inhibitors such as celecoxib, etoricoxib, lumiracoxi
  • NSAIDs can be indicated for the symptomatic relief of conditions such as rheumatoid arthritis, osteoarthritis, inflammatory arthropathies, ankylosing spondylitis, psoriatic arthritis, Reiter's syndrome, acute gout, dysmenorrhoea, metastatic bone pain, headache and migraine, postoperative pain, mild-to-moderate pain due to inflammation and tissue injury, pyrexia, ileus, and renal colic.
  • conditions such as rheumatoid arthritis, osteoarthritis, inflammatory arthropathies, ankylosing spondylitis, psoriatic arthritis, Reiter's syndrome, acute gout, dysmenorrhoea, metastatic bone pain, headache and migraine, postoperative pain, mild-to-moderate pain due to inflammation and tissue injury, pyrexia, ileus, and renal colic.
  • chemotherapeutic agents include, but are not limited to, doxorubicin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, interferons, platinum derivatives, taxanes (e.g., paclitaxel, docetaxel), vinca alkaloids (e.g., vinblastine), anthracyclines (e.g., doxorubicin), epipodophyllotoxins (e.g., etoposide), cisplatin, an mTOR inhibitor (e.g., a rapamycin), methotrexate, actinomycin D, dolastatin 10, colchicine, trimetrexate, metoprine, cyclosporine, daunorubicin, teniposide, amphotericin, alkylating agents (e.g., chlorambucil), 5-fluorouracil, campthothecin,
  • compounds disclosed herein, or a pharmaceutically acceptable composition thereof are administered in combination with an antiproliferative or chemotherapeutic agent selected from any one or more of abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, amifostine, anastrozole, arsenic trioxide, asparaginase, azacitidine, BCG live, bevacuzimab, fluorouracil, bexarotene, bleomycin, bortezomib, busulfan, calusterone, capecitabine, camptothecin, carboplatin, carmustine, cetuximab, chlorambucil, cladribine, clofarabine, cyclophosphamide, cytarabine, dactinomycin, darbepoetin alfa, daunorubicin, denileukin, dex
  • Chemotherapeutic agents also include treatments for Alzheimer's Disease such as donepezil hydrochloride and rivastigmine; treatments for Parkinson's Disease such as L- DOPA/carbidopa, entacapone, ropinrole, pramipexole, bromocriptine, pergolide, trihexephendyl, and amantadine; agents for treating multiple sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®), glatiramer acetate, and mitoxantrone; treatments for asthma such as albuterol and montelukast sodium; agents for treating schizophrenia such as zyprexa, risperdal, seroquel, and haloperidol; anti-inflammatory agents such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such
  • chemotherapeutic agents include pharmaceutically acceptable salts, acids or derivatives of any of chemotherapeutic agents, described herein, as well as combinations of two or more of them.
  • Example 1 (S)-7-((3S,57?)-4-Acryloyl-3,5-dimethylpiperazin-l-yl)-10-(4,4- difluorocyclohex-l-en-l-yl)-3-(methoxymethyl)-9-(trifluoromethyl)-2,3-dihydro-5H- [ 1 ,4]thiazino[2,3 ,4-ij] quinazolin-5 -one [0196] Step 1: tert-butyl (2S,6R)-4-((S)-10-(4,4-difhiorocyclohex-l-en-l-yl)-3- (methoxymethyl)-5-oxo-9-(trifluoromethyl)-2,3-dihydro-5H-[l,4]thiazino[2,3,4- ij]quinazolin-7-yl)-2,6-dimethylpiperazine-l-carboxylate
  • Step 2 (S)-10-(4,4-difluorocyclohex-l-en-l-yl)-7-((3S,5R)-3,5- dimethylpiperazin-l-yl)-3-(methoxymethyl)-9-(trifluoromethyl)-2,3-dihydro-5H- [ 1 ,4]thiazino[2,3 ,4-ij] quinazolin-5 -one
  • Step 3 (S)-7-((3S,5R)-4-Acryloyl-3,5-dimethylpiperazin-l-yl)-10-(4,4- difluorocyclohex-l-en-l-yl)-3-(methoxymethyl)-9-(trifluoromethyl)-2,3-dihydro-5H- [ 1 ,4]thiazino[2,3 ,4-ij] quinazolin-5 -one
  • Example 2 (S)-8-((3S,5R)-4-Acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difhiorocyclohex-l-en-l-yl)-3-(pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro- 2H,6H- [ 1 ,4]thiazepino [2, 3 ,4-ij ] quinazolin-6 -one
  • Step 1 tert-Butyl (2S,6R)-4-((S)-l l-(4,4-difluorocyclohex-l-en-l-yl)-6-oxo-3- (pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate
  • Step 2 (S)-l l-(4,4-difluorocyclohex-l-en-l-yl)-8-((3S,5R)-3,5- dimethylpiperazin-l-yl)-3-(pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Step 3 (S)-8-((3S,5R)-4-Acryloyl-3,5-dimethylpiperazin-l-yl)-ll-(4,4- difhiorocyclohex-l-en-l-yl)-3-(pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro- 2H,6H- [ 1 ,4]thiazepino [2, 3 ,4-ij ] quinazolin-6 -one
  • Example 3 (S)-7-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-10-(4,4- difluorocyclohexyl)-3-(methoxymethyl)-9-(trifluoromethyl)-2,3-dihydro-5H-
  • Step 1 (S)-10-(4,4-difluorocyclohex-l-en-l-yl)-3-(methoxymethyl)-9- (trifluoromethyl)-2,3-dihydro-5H-[l,4]thiazino[2,3,4-ij]quinazoline-5,7(6H)-dione
  • Step 2 (.S)-l0-(4.4-Difluorocyclohcxyl)-3-(mcthoxymcthyl)-9-(trifliioromcthyl)- 2,3-dihydro-5H-[l,4]thiazino[2,3,4-ij]quinazoline-5,7(6H)-dione
  • Step 3 (S)-10-(4,4-difluorocyclohexyl)-7-((3S,5R)-3,5-dimethylpiperazin-l-yl)- 3 -(methoxymethyl)-9-(trifluoromethyl)-2,3 -dihydro-5H-[ 1 ,4]thiazino [2,3 ,4-ij ]quinazolin- 5 -one
  • Step 4 (S)-7-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-10-(4,4- difluorocyclohexyl)-3-(methoxymethyl)-9-(trifluoromethyl)-2,3-dihydro-5H- [ 1 ,4]thiazino[2,3 ,4-ij] quinazolin-5 -one
  • Example 4a and Example 4b (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin- 1 -yl)- 11 -(4,4-difluorocyclohex-l -en- 1 -yl)-3 -(pyridin-2-yl)- 10-(trifluoromethyl)-3,4- dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one (Example 4a) and (S)-8- ((3 S,5R)-4-acryloyl-3,5-dimethylpiperazin- 1 -yl)- 11 -(4,4-difluorocyclohex- 1 -en- 1 -yl)-3 - (pyridin-2-yl)- 10-(trifhioromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thia
  • Step 1 2,4,7-Trichloro-8-iodo-6-(trifluoromethyl)quinazoline
  • Step 2 / /7- But l (2S,6R)-4-(2,7 -dichloro-8-iodo-6-(trifluoromethyl)quinazolin- 4-yl)-2,6-dimethylpiperazine- 1 -carboxylate
  • Step 3 tert-Butyl (2S,6R)-4-(7-chloro-8-iodo-2-oxo-6-(trifluoromethyl)-l,2- dihydroquinazolin-4-yl)-2,6-dimethylpiperazine- 1 -carboxylate
  • Step 6 3-((tert-butyldiphenylsilyl)oxy)-2-(pyridin-2-yl)propan-l-ol
  • Step 7 S-(3-((tert-butyldiphenylsilyl)oxy)-2-(pyridin-2-yl)propyl) ethanethioate
  • Step 8 3-((tert-butyldiphenylsilyl)oxy)-2-(pyridin-2-yl)propane-l-thiol
  • Step 9 tert-Butyl (2.S'.6/?)-4-(8-((3-((/ert-biityldiphcnylsilyl)oxy)-2-(pyridin-2- yl)propyl)thio)-7-chloro-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 10 tert-butyl (2S,6R)-4-(7-chloro-8-((3-hydroxy-2-(pyridin-2- yl)propyl)thio)-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 11 tert-butyl (2S,6R)-4-(l l-chloro-6-oxo-3-(pyridin-2-yl)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine- 1 -carboxylate [0242] To a 0 °C solution of tert-butyl (2.S'.6/?)-4-(7-chloro-8-((3-hydroxy-2-(pyridin-4- yl)propyl)thio)-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4-yl)-2,6- dimethylpiperazine-1 -carboxylate (2.11 mmol) in tetrahydrofuran (100 mb) was added triphenylphos
  • Step 12 tert-Butyl (2S,6R)-4-(l l-(4,4-difluorocyclohex-l-en-l-yl)-6-oxo-3- (pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate
  • Step 13 ll-(4,4-Difluorocyclohex-l-en-l-yl)-8-((3S,5R)-3,5-dimethylpiperazin- l-yl)-3-(pyridin-2-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Step 14 (J?)-8-((3S,5J?)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohex- 1 -en- 1 -yl)-3-(pyridin-2-yl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [l,4]thiazepino[2,3,4-ij]quinazolin-6-one (Example 4a) and (.S)-8-((3.S'.5/?)-4-acryloyl-3.5- dimethylpiperazin- 1 -yl)- 11 -(4,4-difluorocyclohex- 1 -en- 1 -yl)-3-(pyridin-2-yl)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-i
  • Example 4a and Example 4b were prepared analogously to Example 1, step 3, where (.S')- 10-(4,4-difluorocyclohex- 1 -en- 1 -yl)-7 -((3.S'.5 /?)-3.5 -di methyl pipe razi n- 1 -y 1 ) -3 - (methoxymethyl)-9-(trifluoromethyl)-2,3-dihydro-5H-[l,4]thiazino[2,3,4-ij]quinazolin-5- one was replaced with l l-(4,4-difluorocyclohex-l-en-l-yl)-8-((3S,5R)-3,5- dimethylpiperazin- 1 -yl)-3-(pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H- [l,4]thiazepino[2,3,4-ij]quina
  • Example 4a and Example 4b were isolated as single diastereomers after purification of the aforementioned mixture by SFC using a RegisPack 5/100 column (25cmxl0 mm, 5 micron) with 50:50 heptane :isopropanol as mobile phase, a flow rate of 3.0 mL/min and a fixed temperature of 35 °C.
  • Example 5 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohex-l-en-l-yl)-3-(pyrazin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one [0255] Step 1: (S)-2-((l-((tert-butyldiphenylsilyl)oxy)-3-(tritylthio)propan-2- yl)oxy)pyrazine
  • Step 2 (S)-3-((tert-butyldiphenylsilyl)oxy)-2-(pyrazin-2-yloxy)propane-l-thiol
  • Step 3 (S)-8-((3-((tert-butyldiphenylsilyl)oxy)-2-(pyrazin-2-yloxy)propyl)thio)- 7-chloro-6-(trifluoromethyl)quinazoline-2,4(lH,3H)-dione
  • Step 4 (S)-7-chloro-8-((3-hydroxy-2-(pyrazin-2-yloxy)propyl)thio)-6- (trifluoromethyl)quinazoline-2,4(lH,3H)-dione
  • Step 6 tert-butyl (2S,6R)-4-((S)-l l-chloro-6-oxo-3-(pyrazin-2-yloxy)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 7 tert-butyl (2S,6R)-4-((S)-l l-(4,4-difluorocyclohex-l-en-l-yl)-6-oxo-3- (pyrazin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate
  • Step 8 (S)-l l-(4,4-difluorocyclohex-l-en-l-yl)-8-((3S,5R)-3,5- dimethylpiperazin-l-yl)-3-(pyrazin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-
  • Step 9 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohex-l-en-l-yl)-3-(pyrazin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Example 6 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohex- 1 -en- 1 -yl)-3-(pyridin-2-yloxy)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Example 7 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohex- 1 -en- 1 -yl)-3-(pyridin-4-yl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one hloro-8-iodo-6-(trifluoromethyl)quinazoline
  • Step 2 / /7- But l (2S,6R)-4-(2,7 -dichloro-8-iodo-6-(trifluoromethyl)quinazolin- 4-yl)-2,6-dimethylpiperazine- 1 -carboxylate
  • Step 3 tert-Butyl (2S,6R)-4-(7-chloro-8-iodo-2-oxo-6-(trifluoromethyl)-l,2- dihydroquinazolin-4-yl)-2,6-dimethylpiperazine- 1 -carboxylate
  • Step 5 3 -Hydroxy-2 -(pyridin-4-yl)propyl 4-methylbenzenesulfonate
  • Step 6 S-(3-Hydroxy-2-(pyridin-4-yl)propyl) ethanethioate
  • Step 7 tert-Butyl (2.S'.6/?)-4-(7-chloro-8-((3-hydroxy-2-(pyridin-4- yl)propyl)thio)-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 8 tert-Butyl (2.S'.6/?)-4-((/?)- l l -chloro-6-oxo-3-(pyridin-4-yl)- I O- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-1 -carboxylate (Int-7a) and tert-butyl (2S,6R)-4-((S)-l l-chloro-6-oxo- 3-(pyridin-4-yl)-10-(trifhioromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate (Int-7b)
  • Pure diastereomers Int-7a and Int- 7b were obtained by purification of the aforementioned mixture by semi-preparative reverse phase-HPLC (column: DAICEL CHIRALCEL OD (250 mm x 30mm, 10 pm); mobile phase: [Neu-MeOH];B%: 50%-50%,6.4min) and characterized by SFC (Column: Chiracel OD-3, 50 x 4.6 mm ED., 3 pm stabilized at 35 °C with a back pressure of 100 bar.
  • Mobile phase and gradient 40% methanol containing 0.05% diethylamine in CO2, using a flow rate of 3 mL/min.
  • Detector Photodiode-array).
  • Step 9 tert-butyl (2.S',6/?)-4-((.S)- 1 l-(4,4-difluorocyclohex-l-en-l-yl)-6-oxo-3- (pyridin-4-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-I-carboxylate
  • Step 11 (S)-8-((3S,57?)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohex- 1 -en- 1 -yl)-3-(pyridin-4-yl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Example 8 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-ll-(4,4- difluorocyclohexyl)-3-(pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Step 1 Methyl 3-amino-4',4'-difluoro-2',3',4',5'-tetrahydro-[l,r-biphenyl]-4- carboxylate
  • Step 3 Methyl 2-amino-4-(4,4-difluorocyclohexyl)-5 -iodobenzoate
  • Step 4 Methyl 2-acetamido-4-(4,4-difluorocyclohexyl)-5-iodobenzoate
  • Step 7 Methyl 2-amino-4-(4,4-difluorocyclohexyl)-3-iodo-5- (trifluoromethyl)benzoate
  • Step 8 2-Amino-4-(4,4-difluorocyclohexyl)-3-iodo-5-(trifluoromethyl)benzoic acid
  • Step 9 7-(4,4-Difluorocyclohexyl)-8-iodo-6-(trifluoromethyl)quinazoline- 2,4(lH,3H)-dione
  • a mixture of 2-amino-4-(4,4-difluorocyclohexyl)-3-iodo-5- (trifluoromethyl)benzoic acid (24 mmol) and urea (78 mmol) was heated at 210 °C for one hour. After cooling down to room temperature, water was added and the solid filtered and washed with methanol multiple times. The filtrate was concentrated and the residue purified by HPLC to afford the title compound as a yellow solid.
  • Step 10 (S)-2-((l-((tert-butyldiphenylsilyl)oxy)-3-(tritylthio)propan-2- yl)oxy)pyrimidine
  • Step 11 (S)-3-((tert-butyldiphenylsilyl)oxy)-2-(pyrimidin-2-yloxy)propane-l- thiol
  • Step 12 (S)-8-((3-((tert-butyldiphenylsilyl)oxy)-2-(pyrimidin-2- yloxy)propyl)thio)-7-(4,4-difluorocyclohexyl)-6-(trifluoromethyl)quinazoline-2,4(lH,3H)-
  • Step 13 (S)-7-(4,4-difluorocyclohexyl)-8-((3-hydroxy-2-(pyrimidin-2- yloxy)propyl)thio)-6-(trifluoromethyl)quinazoline-2,4(lH,3H)-dione
  • Step 14 (S)-l l-(4,4-difluorocyclohexyl)-3-(pyrimidin-2-yloxy)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazoline-6,8(7H)-dione
  • Step 15 tert-butyl (2S,6R)-4-((S)-l l-(4,4-difluorocyclohexyl)-6-oxo-3- (pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate
  • Step 16 (S)-l l-(4,4-difluorocyclohexyl)-8-((3S,5R)-3,5-dimethylpiperazin-l- yl)-3-(pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Step 17 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohexyl)-3-(pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Example 9 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohexyl)-3-(pyridin-2-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Step 1 8-((3-((tert-butyldiphenylsilyl)oxy)-2-(pyridin-2-yl)propyl)thio)-7-(4,4- difluorocyclohexyl)-6-(trifluoromethyl)quinazoline-2,4(lH,3H)-dione
  • Step 2 7-(4,4-difluorocyclohexyl)-8-((3-hydroxy-2-(pyridin-2-yl)propyl)thio)-6- (trifluoromethyl)quinazoline-2,4(lH,3H)-dione
  • Step 3 l l-(4,4-difluorocyclohexyl)-3-(pyridin-2-yl)-10-(trifluoromethyl)-3,4- dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazoline-6,8(7H)-dione
  • Step 4 tert-butyl (2S,6R)-4-(l l-(4,4-difluorocyclohexyl)-6-oxo-3-(pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 5 l l-(4,4-difluorocyclohexyl)-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3- (pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Step 6 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohexyl)-3-(pyridin-2-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Example 10 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohexyl)-3-(pyridin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Example 11 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l 1-chloro- 3 -(pyrimidin-2-yloxy)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H-[ 1 ,4]thiazepino [2,3,4- ij ] quinazolin-6-one
  • Step 1 (S)-l l-chloro-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3-(pyrimidin-2- yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Example 12 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(3,6- dihydro-2H-thiopyran-4-yl)-3-(pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro- 2H,6H- [ 1 ,4]thiazepino [2, 3 ,4-ij ] quinazolin-6 -one
  • Step 1 tert-butyl-(2S,6R)-4-((S)-ll-(3,6-dihydro-2H-thiopyran-4-yl)-6-oxo-3-
  • Step 2 (S)-l l-(3,6-dihydro-2H-thiopyran-4-yl)-8-((3S,5R)-3,5- dimethylpiperazin-l-yl)-3-(pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Step 3 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(3,6-dihydro- 2H-thiopyran-4-yl)-3-(pyrimidin-2-yloxy)-10-(trifIuoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one [0369] The title compound was prepared analogously to Example 1, step 3, where (S)- 10-(4,4-difluorocyclohex- 1 -en- 1 -yl)-7 -((3 S,5 R)-3 ,5 -dimethylpiperazin- 1 -yl )- 3 - (methoxymethyl)-9-(trifluoromethyl)-2,3-dihydro-5H-[l,4]thiazino[2,3,
  • Example 13 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(3,6- dihydro-2H-pyran-4-yl)-3-(pyrimidin-2-yloxy)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Example 14 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-ll-chloro-3- cyclopropyl-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin- 6-one
  • Step 1 Ethyl 2-cyclopropylacetate
  • Step 2 l-(tert-Butyl) 3-ethyl 2-cyclopropylmalonate
  • Step 3 2-Cyclopropylpropane- 1,3 -diol
  • Step 4 3-((tert-butyldiphenylsilyl)oxy)-2-cyclopropylpropan-l-ol
  • Step 5 S-(3-((tert-butyldiphenylsilyl)oxy)-2-cyclopropylpropyl) ethanethioate
  • Step 6 3-((tert-butyldiphenylsilyl)oxy)-2-cyclopropylpropane-l-thiol
  • Step 7 8-((3-((tert-butyldiphenylsilyl)oxy)-2-cyclopropylpropyl)thio)-7-chloro- 6-(trifluoromethyl)quinazoline-2,4(lH,3H)-dione
  • Step 9 l l-chloro-3-cyclopropyl-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazoline-6,8(7H)-dione
  • Step 10 tert-Butyl (2.S'.6/?)-4-((/?)-l l -chloro-3-cyclopropyl-6-oxo-IO- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-1 -carboxylate and tert-butyl (2.S',6/?)-4-((.S)-l l -chloro-3-cyclopropyl- 6-oxo-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)- 2,6-dimethylpiperazine- 1 -carboxylate
  • Step 11 (R)-l l-chloro-3-cyclopropyl-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Step 12 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-ll-chloro-3- cyclopropyl-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin- 6-one
  • Example 15 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-chloro-3- cyclopropyl-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin- 6-one
  • Step 1 (S)-l l-chloro-3-cyclopropyl-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-10-
  • Step 2 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-chloro-3- cyclopropyl-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin- 6-one
  • Example 16 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-ll-(4,4- difluorocyclohex- 1 -en- 1 -yl)-3-(4-fluorophenyl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Step 1 2-(4-Fluorophenyl)prop-2-en-l-ol
  • Step 2 tert-butyl((2-(4-fluorophenyl)allyl)oxy)diphenylsilane
  • Step 3 3-((tert-butyldiphenylsilyl)oxy)-2-(4-fluorophenyl)propan-l-ol
  • reaction mixture was partitioned between water (50 mL) and ethyl acetate (100 mL) and the aqueous layer was extracted with ethyl acetate twice. The combined organic layers were concentrated under reduced pressure to afford a residue that was purified by silica gel chromatography (10%-25% ethyl acetate in hexanes). The title compound was isolated in 66% yield as a yellow oil.
  • Step 4 S-(3-((tert-butyldiphenylsilyl)oxy)-2-(4-fluorophenyl)propyl) ethanethioate
  • Ethanethioic .S'- acid (22 mmol) was added to a solution of 3-((tert- butyldiphenylsilyl)oxy)-2-(4-fluorophenyl)propan-l-ol (11 mmol), diisopropyl azodicarboxylate (22 mmol) and triphenylphosphine (22 mmol) in tetrahydrofuran (50 mL). After 16 hours, the volatiles were removed under reduced pressure and the resulting residue was purified by silica gel chromatography (5-33% ethyl acetate in hexanes). The title compound was isolated in 81% yield as a yellow oil.
  • Step 5 tert-butyl (2.S'.6/?)-4-(8-((3-((tcrt-biityldiphcnylsilyl)oxy)-2-(4- fluorophenyl)propyl)thio)-7-chloro-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4- yl)-2,6-dimethylpiperazine- 1 -carboxylate
  • Step 6 tert-butyl (2S,6R)-4-(7-chloro-8-((2-(4-fluorophenyl)-3- hydroxypropyl)thio)-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 7 tert-butyl (2.S'.6/?)-4-((/?)-l l-chloro-3-(4-fluorophcnyl)-6-oxo- IO- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-1 -carboxylate and tert-butyl (2.S'.6/?)-4-((.S)-l l -chloro-3-(4- fluorophenyl)-6-oxo-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate [0423] The title compounds were prepared analogously to Example 4,
  • tert-butyl (2S,6R)-4-((R)-l l-chloro-3-(4-fluorophenyl)-6-oxo-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-1 -carboxylate and tert-butyl (2S,6R)-4-((S)-ll-chloro-3-(4- fluorophenyl)-6-oxo-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate were obtained as single diastereomers by purification of the aforementioned mixture by SFC (column: REGI
  • tert-butyl (2.S'.6/?)-4-((/?)- l l-chloro-3-(4-fluorophcnyl)-6-oxo- IO- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-l-carboxylate: Rt 1.29 min; MS (ESI) m/z: 627.1 [M+l]+ [0426] tert-butyl (2.S'.6/?)-4-((.S')-l l -chloro-3-(4-fluorophcnyl)-6-oxo-IO- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2
  • Step 8 tert-butyl (2.S'.6/?)-4-((/?)-l l-(4.4-difliiorocyclohcx- l-cn-l-yl)-3-(4- fhrorophenyl)-6-oxo-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate
  • Step 9 (7?)-l l-(4,4-difluorocyclohex-l-en-l-yl)-8-((3S,57?)-3,5- dimethylpiperazin-l-yl)-3-(4-fluorophenyl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Step 10 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohex- 1 -en- 1 -yl)-3-(4-fluorophenyl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Example 17 (.S)-8-((3.S'.5/ )-4-acryloyl-3.5-dimcthylpipcrazin- l -yl)- l l-(4,4- difluorocyclohex- 1 -en- 1 -yl)-3-(4-fluorophenyl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Step 1 tert-butyl (2S,67?)-4-((S)-l l-(4,4-difluorocyclohex-l-en-l-yl)-3-(4- fluorophenyl)-6-oxo-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate
  • Step 2 (7?)-l l-(4,4-difluorocyclohex-l-en-l-yl)-8-((3S,57?)-3,5- dimethylpiperazin-l-yl)-3-(4-fluorophenyl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Step 3 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-(4,4- difluorocyclohex- 1 -en- 1 -yl)-3-(4-fluorophenyl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Example 18 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-ll-chloro-3- (pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Step 1 3-Hydroxy-2-(pyridin-2-yl)propyl 4-methylbenzenesulfonate HO ⁇ > ⁇ OTs
  • Step 2 S-(3-Hydroxy-2-(pyridin-2-yl)propyl) ethanethioate
  • Step 3 tert-Butyl (2.S'.6/?)-4-(7-chloro-8-((3-hydroxy-2-(pyridin-2- yl)propyl)thio)-6-(trifluoromethyl)- 1 ,2-dihydroquinazolin-4-yl)-2,6-dimethylpiperazine - 1 - carboxylate
  • Step 4 tert-Butyl (2.S'.6/?)-4-((.S)- l l -chloro-6-oxo-3-(pyridin-2-yl)- I O- (trifhioromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine- 1 -carboxylate and tert-Butyl (2.S'.6/?)-4-((/?)- l l -chloro-6-oxo-3- (pyridin-2-yl)- 10-(trifhioromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate
  • Step 5 (/?)- l l -chloro-8-((3.S'.5/?)-3.5-dimcthylpipcrazin- l -yl)-3-(pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Step 6 (/?)-8-((3.S'.5/ )-4-acryloyl-3.5-dimcthylpipcrazin- l-yl)- l l -chloro-3- (pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thiazepino[2,3,4-
  • Example 19 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l- cyclopropyl-3-(pyridin-2-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazolin-6-one
  • Step 1 tert-butyl (2S,6R)-4-((R)-l l-cyclopropyl-6-oxo-3-(pyridin-2-yl)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 2 (R)-l l-cyclopropyl-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3-(pyridin-2- yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Step 3 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-cyclopropyl-3- (pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • the title compound was prepared analogously to Example 1, step 3, where ( S)- 10-(4,4-difluorocyclohex- 1 -en- 1 -yl)-7 -((3 S,5 R)-3 ,5 -dimethylpiperazin- 1 -yl )- 3 - (methoxymethyl)-9-(trifluoromethyl)-2,3-dihydro-5
  • Example 21 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l 1-chloro- 3 -(5 -fluoropyridin-3-yl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3,4- ij ] quinazolin-6-one
  • Step 1 tert-butyl (2S,6R)-4-((R)-l l-chloro-3-(5-fluoropyridin-3-yl)-6-oxo-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-1 -carboxylate and tert-butyl (2S,6R)-4-((S)-l l-chloro-3-(5- fluoropyridin-3-yl)-6-oxo-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate [0468] The title compounds were prepared analogously to Example 16, where
  • Step 2 (S)-l l-chloro-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3-(5-fluoropyridin- 3-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Example 22 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-chloro-3- (pyrazin-2-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Step 1 tert-butyldimethyl((2-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)allyl)oxy)silane
  • Step 2 2-(3-((tert-butyldimethylsilyl)oxy)prop-l-en-2-yl)pyrazine
  • Step 3 S-(3-((tert-butyldimethylsilyl)oxy)-2-(pyrazin-2-yl)propyl) ethanethioate
  • Step 4 tert-butyl (2S,6R)-4-(8-((3-((tert-butyldimethylsilyl)oxy)-2-(pyrazin-2- yl)propyl)thio)-7-chloro-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 5 tert-butyl (2S,6R)-4-(7-chloro-8-((3-hydroxy-2-(pyrazin-2- yl)propyl)thio)-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 6 tert-butyl (2S,6R)-4-((S)-ll-chloro-6-oxo-3-(pyrazin-2-yl)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-1 -carboxylate and tert-butyl (2S,6R)-4-((R)-l l-chloro-6-oxo-3- (pyrazin-2-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2,6-dimethylpiperazine-l-carboxylate
  • the title compounds were obtained as a 1: 1 mixture of diastereomers in 59% yield.
  • Each individual diastereomer was obtained by purification of the aforementioned mixture by SFC (Chiracel Chiralpak AS 250x30mm, 10 pm using 50% methanol in CO2 as mobile phase).
  • Each individual diastereomer was characterized by analytical SFC (Chiralpak IC-3, 50x4.6mm, 3 pm using as mobile phase 5-40% methanol (containing 0.05% of diethylamine) in CO2 at 3 mL/min).
  • Step 7 (R)-l l-chloro-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3-(pyrazin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Step 8 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-chloro-3- (pyrazin-2-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Example 24 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-chloro- 3-(pyridin-4-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-
  • Step 1 (S)-l l-chloro-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3-(pyridin-4-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Example 25 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l 1-chloro- 3 -(3 -fluoropyridin-4-yl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3,4- ij ] quinazolin-6-one [0503] Step 1: tert-butyl (2S,6R)-4-((S)-ll-chloro-3-(3-fluoropyridin-4-yl)-6-oxo-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-1 -carboxylate and tert-butyl (2S,6R)-4-acryloy
  • Step 2 (S)-l l-chloro-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3-(3-fluoropyridin- 4-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Step 3 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-chloro-3-(3- fluoropyridin-4-yl)- 10-(trifluoromethyl)-3 ,4-dihydro-2H,6H-[ 1 ,4]thiazepino [2,3,4- ij ] quinazolin-6-one
  • Example 28 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-bromo- 3-(pyridin-2-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Step 4 7-Bromo-8-iodo-6-(trifluoromethyl)quinazoline-2,4(lH,3H)-dione
  • reaction mixture was diluted with water, extracted with ethyl acetate three times and the combined organic layers were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to afford a residue that was purified by silica gel chromatography (0-15% ethyl acetate in hexanes). The title compound was isolated in 91% yield as yellow solid.
  • Step 5 tert-butyl (2S,6R)-4-(2,7-dibromo-8-iodo-6-(trifluoromethyl)quinazolin- 4-yl)-2,6-dimethylpiperazine- 1 -carboxylate
  • Step 7 tert-Butyl (2S,6R)-4-(7-bromo-8-((3-((tert-butyldiphenylsilyl)oxy)-2- (pyridin-2-yl)propyl)thio)-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 8 tert-butyl (2S,6R)-4-(7-bromo-8-((3-hydroxy-2-(pyridin-2- yl)propyl)thio)-2-oxo-6-(trifluoromethyl)-l,2-dihydroquinazolin-4-yl)-2,6- dimethylpiperazine- 1 -carboxylate
  • Step 9 tert-butyl (2S,6R)-4-((R)-l l-bromo-6-oxo-3-(pyridin-2-yl)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-1 -carboxylate and tert-butyl (2S,6R)-4-((S)-l l-bromo-6-oxo-3- (pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thiazepino[2,3,4- ij]quinazolin-8-yl)-2
  • Step 10 (R)-l l-chloro-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3-(pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Step 10 (R)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-bromo-3- (pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[ 1 ,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Example 29 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l 1-chloro- 3-(4-fluoropyridin-3-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij ] quinazolin-6-one [0542] Step 1: tert-butyl (2S,6R)-4-((S)-ll-chloro-3-(4-fluoropyridin-3-yl)-6-oxo-10-
  • Step 2 (S)-l l-chloro-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3-(4-fluoropyridin-
  • Step 3 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-chloro-3-(4- fluoropyridin-3-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Example 32 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-chloro- 3-(pyridin-3-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Step 1 tert-butyl (2S,6R)-4-((S)-l l-chloro-6-oxo-3-(pyridin-3-yl)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)-2,6- dimethylpiperazine-1 -carboxylate and tert-butyl (2S,6R)-4-((R)-l l-chloro-6-oxo-3-
  • Step 2 (S)-l l-chloro-8-((3S,5R)-3,5-dimethylpiperazin-l-yl)-3-(pyridin-3-yl)- 10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Step 3 (S)-8-((3S,5R)-4-acryloyl-3,5-dimethylpiperazin-l-yl)-l l-chloro-3- (pyridin-3-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4- ij ] quinazolin-6-one
  • Example 33 (R)-8-(4-acryloylpiperazin-l-yl)-l l-chloro-3-(pyridin-2-yl)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • Step 1 8-((3-((tert-butyldiphenylsilyl)oxy)-2-(pyridin-2-yl)propyl)thio)-7- chloro-6-(trifluoromethyl)quinazoline-2,4(lH,3H)-dione
  • Step 3 l l-Chloro-3-(pyridin-2-yl)-10-(trifluoromethyl)-3,4-dihydro-2H,6H- [ 1 ,4]thiazepino [2,3 ,4-ij ] quinazoline-6,8(7H)-dione
  • Step 4 tert-butyl (S)-4-(l l-chloro-6-oxo-3-(pyri din-2 -yl)-10-(trifluoromethyl)- 3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)piperazine-l-carboxylate and tert-butyl (R)-4-( 11 -chloro-6-oxo-3-(pyridin-2-yl)- 10-(trifluoromethyl)-3,4-dihydro- 2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-8-yl)piperazine-l-carboxylate
  • Step 5 (R)-l l-chloro-8-(piperazin-l-yl)-3-(pyridin-2-yl)-10-(trifluoromethyl)- 3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • the title compound was prepared analogously to Example 1, step 2 where tertbutyl (2S,6R)-4-((S)-10-(4,4-difluorocyclohex-l-en-l-yl)-3-(methoxymethyl)-5-oxo-9- (trifluoromethyl)-2,3-dihydro-5H-[l,4]thiazino[2,3,4-ij]quinazolin-7-yl)-2,6- dimethylpiperazine-1 -carboxylate was replaced with tert-butyl (R)-4-(l l-chloro-6-oxo-3
  • Step 6 (R)-8-(4-acryloylpiperazin-l-yl)-l l-chloro-3-(pyridin-2-yl)-10- (trifluoromethyl)-3,4-dihydro-2H,6H-[l,4]thiazepino[2,3,4-ij]quinazolin-6-one
  • R' is H, CH3, F, Cl, or CF3; and, wherein R is
  • This Example provides a protocol for assessing covalent adduct formation (CAF) between the compounds shown in Tables 1 to 4 above and KRAS.
  • Recombinant Human KRAS 4B protein containing the G12C mutation was used in compound screening experiments. This protein contained 188 amino acids in total, including an N-terminal 6-Histidine tag, followed by a Tobacco Etch Virus (TEV) tag, followed by residues 1-169 of the native KRAS 4B sequence. The exact mass of the protein was 21,310 Da as determined by mass spectrometry. The full amino acid sequence is shown below:
  • the assay can be conducted using a KRAS 4b G12C protein having 170 amino acids, a mass of 19,336 Da, and the amino acid sequence SM TEYKLVVVGA CGVGKSALTI QLIQNHFVDE YDPTIEDSYR KQVVIDGETC LLDILDTAGQ EEYSAMRDQY MRTGEGFLCV FAINNTKSFE DIHHYREQIK RVKDSEDVPM VLVGNKCDLP SRTVDTKQAQ DLARSYGIPF IETSAKTRQG VDDAFYTLVR EIRKHKEK (SEQ ID NO.: 5).
  • the recombinant protein was expressed in E. coli BL21 cells and purified using affinity chromatography via a Ni-NTA column. Protein stocks were nucleotide-exchanged to >95 % GDP, concentrated to 4 mg/mL, and stored at -80 °C in storage buffer (50 mM HEPES pH 7.4, 50 mM NaCl, 5 mM MgC12, 1 mM DTT). Pure KRAS 4B G12C protein was diluted to a concentration of 5 pM in Tris Buffered Saline, pH 7.4. The compounds were dissolved in DMSO and added to the diluted protein to make a 10 pM concentration. The total DMSO concentration in the reaction was 4%.
  • the reaction was mixed by pipetting and incubated at 22 °C for one hour. Aliquots of the reaction were taken over time and diluted 2: 1 in 0.1% formic acid.
  • the intact mass of the protein samples was measured by LC-MS using a QExactive+ mass spectrometer (Thermo Scientific). An amount of 500 ng total protein was injected onto a C8 reverse phase column, eluted with a seven-minute gradient of 30%-90% acetonitrile/0.1% formic acid, and analyzed for intact mass by the mass spectrometer. Adducts identified were confirmed to be within 1 Dalton of the expected mass, and the relative ratios of free: adduct protein were used to quantify the percentage of protein bound by the compound. CAF reactions were run in duplicate, with a typical variability of ⁇ 5%.
  • KRAS G12C mutant cell lines, NCI H358 (ATCC, CRL-5807), and Ras Initiative (RI) KRAS G12C were cultured according to published protocols and maintained at 37 °C in 5% CO2.
  • the phospho-ERK HTRF assay was executed following provider’s protocol (CisBio #64AERPEH).
  • NCI-H358 or RI KRAS G12C cells were plated at a density of 50,000 cells per well in a 96-well plate (Coming #3903) in respective medias (for NCI-H358, RPMI + 10% FBS + 1% Pen/Strep, and for RI KRAS G12C, DMEM + 10% FBS + 1% Pen/Strep + 4 pg/mL Blasticidin) and maintained at 37 °C in 5% CO2.
  • Cells were allowed to adhere overnight and treated the following day with a Tecan D300e Digital Dispenser (Tecan Group Ltd., Switzerland) using an 11-point dose response starting at 2,500 nM of exemplified compounds followed by sequential 1:3 dilutions for either 4 hours or 16 hours. Following compound treatment, the cells were washed once with ice-cold PBS. Cells were lysed by adding 50 pL of lysis buffer (lx) supplemented with lx Pierce Halt Protease and Phosphatase inhibitor and incubated for 30 minutes at 4 °C with shaking.
  • lysis buffer lx

Landscapes

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

Abstract

L'invention concerne des composés ayant une activité inhibitrice de KRAS G12C et des procédés d'utilisation de ceux-ci pour traiter un cancer comprenant une mutation K-Ras G12C.
PCT/US2023/064622 2022-03-21 2023-03-17 Pyrimidones tricycliques WO2023183755A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263269676P 2022-03-21 2022-03-21
US63/269,676 2022-03-21

Publications (1)

Publication Number Publication Date
WO2023183755A1 true WO2023183755A1 (fr) 2023-09-28

Family

ID=88102144

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/064622 WO2023183755A1 (fr) 2022-03-21 2023-03-17 Pyrimidones tricycliques

Country Status (1)

Country Link
WO (1) WO2023183755A1 (fr)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104844573A (zh) * 2015-04-17 2015-08-19 中国药科大学 嘧啶类btk抑制剂、其制备方法及医药用途
WO2019110751A1 (fr) * 2017-12-08 2019-06-13 Astrazeneca Ab Composés tétracycliques en tant qu'inhibiteurs de la protéine ras mutante g12c, destinés à être utilisés en tant qu'agents anticancéreux
WO2020239123A1 (fr) * 2019-05-31 2020-12-03 上海翰森生物医药科技有限公司 Modulateur de dérivé hétérocyclique aromatique et son procédé de préparation et son utilisation
WO2021127404A1 (fr) * 2019-12-20 2021-06-24 Erasca, Inc. Pyridones et pyrimidones tricycliques

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104844573A (zh) * 2015-04-17 2015-08-19 中国药科大学 嘧啶类btk抑制剂、其制备方法及医药用途
WO2019110751A1 (fr) * 2017-12-08 2019-06-13 Astrazeneca Ab Composés tétracycliques en tant qu'inhibiteurs de la protéine ras mutante g12c, destinés à être utilisés en tant qu'agents anticancéreux
WO2020239123A1 (fr) * 2019-05-31 2020-12-03 上海翰森生物医药科技有限公司 Modulateur de dérivé hétérocyclique aromatique et son procédé de préparation et son utilisation
WO2021127404A1 (fr) * 2019-12-20 2021-06-24 Erasca, Inc. Pyridones et pyrimidones tricycliques

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
I. V. UKRAINETS ; E. V. MOSPANOVA ; L. V. SIDORENKO: "4-Hydroxy-2-quinolones. 122. 1-Hydroxy-3-oxo-5,6-dihydro-3H-pyrrolo[3,2,1-ij]-quinoline-2-carboxylic acid hetarylamides as potential antitubercular agents", CHEMISTRY OF HETEROCYCLIC COMPOUNDS, KLUWER ACADEMIC PUBLISHERS-PLENUM PUBLISHERS, NL, vol. 43, no. 7, 1 July 2007 (2007-07-01), NL , pages 863 - 870, XP019550110, ISSN: 1573-8353, DOI: 10.1007/s10593-007-0137-3 *

Similar Documents

Publication Publication Date Title
WO2021173923A1 (fr) Hétérocycles fusionnés à la pyrrolidine
WO2021127404A1 (fr) Pyridones et pyrimidones tricycliques
EP4217071A1 (fr) Pyridones et pyrimidones tricycliques
US11845761B2 (en) Tricyclic pyridones and pyrimidones
EP3218375B1 (fr) Pyrrolopyridines substituées comme inhibiteurs de bromodomaines
EP2970307B1 (fr) Composés pyrazolo et leurs utilisations
EP3204379A1 (fr) Composés de pyrrolidine à utiliser en tant qu'inhibiteurs de l'histone déméthylase
WO2022266206A1 (fr) Conjugués d'inhibiteurs de kras
US10358437B2 (en) Therapeutic compounds and uses thereof
WO2022271658A1 (fr) Inhibiteurs de kras tricycliques
WO2022266069A1 (fr) Inhibiteurs tricycliques de kras g12d
WO2022265974A1 (fr) Inhibiteurs de kras tricycliques substitués par un aminohétérocycle
KR20240026948A (ko) 황 함유 헤테로방향족 트리사이클릭 kras 억제제
WO2022266167A1 (fr) Inhibiteurs de kras tricycliques contenant un amide et de l'urée
WO2015049325A1 (fr) Inhibiteurs thérapeutiques de cdk8 et utilisations de ceux-ci
WO2016112251A1 (fr) Dérivés 4,5-dihydroimidazole et leur utilisation comme inhibiteurs d'histone déméthylase (kdm2b)
CN116615428A (zh) 三环吡啶酮和嘧啶酮
WO2023183755A1 (fr) Pyrimidones tricycliques
WO2023212548A1 (fr) Pyridones et pyrimidones tricycliques
WO2023212549A1 (fr) Pyridones et pyrimidones tricycliques
WO2024040080A1 (fr) Conjugués inhibiteurs de kras
CN118076612A (en) Sulfur-containing heteroaromatic tricyclic KRAS inhibitors
JP2024520457A (ja) 硫黄含有ヘテロ芳香族三環式kras阻害剤

Legal Events

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

Ref document number: 23775809

Country of ref document: EP

Kind code of ref document: A1