WO2012118492A1 - Heterocyclic sulfonamides as raf inhibitors - Google Patents

Heterocyclic sulfonamides as raf inhibitors Download PDF

Info

Publication number
WO2012118492A1
WO2012118492A1 PCT/US2011/026700 US2011026700W WO2012118492A1 WO 2012118492 A1 WO2012118492 A1 WO 2012118492A1 US 2011026700 W US2011026700 W US 2011026700W WO 2012118492 A1 WO2012118492 A1 WO 2012118492A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
mmol
phenyl
hydrogen
sulfonamide
Prior art date
Application number
PCT/US2011/026700
Other languages
French (fr)
Inventor
Jonas Grina
Joshua D. Hansen
Ellen Laird
Simon Mathieu
David Moreno
Li Ren
Joachim Rudolph
Steven Mark Wenglowsky
Original Assignee
Array Biopharma Inc.
Genentech, 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 Array Biopharma Inc., Genentech, Inc. filed Critical Array Biopharma Inc.
Priority to PCT/US2011/026700 priority Critical patent/WO2012118492A1/en
Publication of WO2012118492A1 publication Critical patent/WO2012118492A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/88Oxygen atoms
    • 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
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/74Quinazolines; Hydrogenated quinazolines with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, attached to ring carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/36Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems
    • C07D241/38Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems with only hydrogen or carbon atoms directly attached to the ring nitrogen atoms
    • C07D241/40Benzopyrazines
    • C07D241/44Benzopyrazines with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • Raf/MEK/ERK pathway is critical for cell survival, growth, proliferation and tumorigenesis.
  • Li Nanxin, et al. "B-Raf kinase inhibitors for cancer treatment.” Current Opinion in Investigational Drugs. Vol. 8, No. 6 (2007): 452-456.
  • Raf kinases exist as three isoforms, A-Raf, B-Raf and C-Raf. Among the three isoforms, studies have shown that B- Raf functions as the primary MEK activator.
  • B-Raf is one of the most frequently mutated genes in human cancers.
  • B-Raf kinase represents an excellent target for anticancer therapy based on preclinical target validation, epidemiology and drugability.
  • Another aspect of the present invention provides methods of preventing or treating a disease or disorder modulated by B-Raf, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof.
  • diseases and disorders include, but are not limited to, hyperproliferative disorders (such as cancer, including melanoma and other cancers of the skin), neurodegeneration, cardiac hypertrophy, pain, migraine and neurotraumatic disease.
  • Another aspect of the present invention provides methods of preventing or treating cancer, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention, or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds having anti-cancer properties.
  • Another aspect of the present invention provides methods of preventing or treating cancer, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention, or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds having anti-cancer properties.
  • Another aspect of the present invention provides a method of treating a hyperproliferative disease in a mammal comprising administering a therapeutically effective amount of a compound of this invention to the mammal.
  • Another aspect of the present invention provides methods of preventing or treating kidney disease, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention, or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds.
  • Another aspect of the present invention provides methods of preventing or treating polycystic kidney disease, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention, or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds.
  • Another aspect of the present invention provides the compounds of the present invention for use in therapy.
  • Another aspect of the present invention provides the compounds of the present invention for use in the treatment of a hyperproliferative disease.
  • the hyperproliferative disease may be cancer (or still further, a specific cancer as defined herein).
  • kidney disease may be polycystic kidney disease.
  • Another aspect of the present invention provides the use of a compound of this invention in the manufacture of a medicament for the treatment of a hyperproliferative disease.
  • the hyperproliferative disease may be cancer (or still further, a specific cancer as defined herein).
  • kidney disease may be polycystic kidney disease.
  • Another aspect of the present invention provides the use of a compound of the present invention in the manufacture of a medicament, for use as a B-Raf inhibitor in the treatment of a patient undergoing cancer therapy.
  • Another aspect of the present invention provides the use of a compound of the present invention in the manufacture of a medicament, for use as a B-Raf inhibitor in the treatment of a patient undergoing polycystic kidney disease therapy.
  • Another aspect of the present invention provides a pharmaceutical composition comprising a compound of the present invention for use in the treatment of a hyperproliferative disease.
  • Another aspect of the present invention provides a pharmaceutical composition comprising a compound of the present invention for use in the treatment of cancer.
  • Another aspect of the present invention provides a pharmaceutical composition comprising a compound of the present invention for use in the treatment of polycystic kidney disease.
  • Another aspect of the present invention provides a pharmaceutical composition comprising a compound of this invention, a stereoisomer, tautomer, solvate, prodrug or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
  • Another aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of this invention or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
  • Another aspect of the present invention provides intermediates for preparing compounds of Formulas I, II, III, IV, V, VI, and VII. Certain compounds of Formulas I, II, III, IV, V, VI, and VII may be used as intermediates for other compounds of Formulas I, II, III, IV, V, VI, and VII.
  • Another aspect of the present invention includes methods of preparing, methods of separation, and methods of purification of the compounds of this invention.
  • alkyl includes linear or branched-chain radicals of carbon atoms.
  • alkyl moieties have been abbreviated, for example, methyl (“Me”), ethyl (“Et”), propyl (“Pr”) and butyl (“Bu”), and further abbreviations are used to designate specific isomers of compounds, for example, 1 -propyl or n-propyl (“n-Pr”), 2-propyl or isopropyl (“i-Pr”), 1- butyl or n-butyl (“n-Bu”), 2-methyl-l -propyl or isobutyl (“i-Bu”), 1-methylpropyl or s-butyl (“s-Bu”), 1,1-dimethylethyl or t-butyl (“t-Bu”) and the like.
  • n-Pr 1-propyl or n-propyl
  • i-Pr 2-propyl or isopropyl
  • i-Bu 2-methyl-l -propyl or isobutyl
  • s-Bu 1,1-d
  • DMSO dimethylsulfoxide
  • DMF dimethylformamide
  • DCM dichloromethane
  • EtOAc ethyl acetate
  • THF tetrahydrofuran
  • alkenyl refers to linear or branched-chain monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having "cis” and “trans” orientations, or alternatively, "E” and "Z” orientations.
  • the alkenyl radical is two to six carbon atoms (C 2 - C 6 ).
  • the alkenyl radical is C 2 -C 5 , C 2 -C 4 or C 2 -C 3 .
  • alkynyl refers to a linear or branched monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein.
  • the alkynyl radical is two to six carbon atoms (C 2 -C 6 ).
  • the alkynyl radical is C 2 -C 5 , C 2 -C 4 or C 2 -C 3 .
  • cycloalkyl refers to a non-aromatic, saturated or partially unsaturated hydrocarbon ring group, wherein the cycloalkyl group may be optionally substituted independently with one or more substituents described herein.
  • the cycloalkyl group is 3 to 6 carbon atoms (C 3 -C 6 ).
  • cycloalkyl is C 3 -C 4 or C 3 -C 5 .
  • the cycloalkyl group, as a monocycle is C 3 -C 6 or C 5 -C 6 .
  • the cycloalkyl group, as a bicycle is C 7 -C 12 .
  • halogen refers to F, CI, Br or I.
  • heterocyclic or “heterocycle” or “heterocyclyl” refers to a saturated or a partially unsaturated (i.e., having one or more double and/or triple bonds within the ring) cyclic group in which at least one ring atom is a heteroatom independently selected from nitrogen, oxygen, and sulfur, the remaining ring atoms being carbon.
  • heterocyclyl includes saturated or partially unsaturated 3-7 membered heterocyclyl groups, another embodiment includes 4-6 membered heterocyclyl groups, and another embodiment includes 5-6 membered heterocyclyl groups.
  • the heterocyclyl group may be optionally substituted with one or more substituents described herein.
  • Heterocycles include 3 to 7 membered rings containing one or two heteroatoms selected from oxygen, nitrogen and sulfur.
  • heteroaryl refers to an aromatic cyclic group in which at least one ring atom is a heteroatom independently selected from nitrogen, oxygen and sulfur, the remaining ring atoms being carbon. Heteroaryl groups may be optionally substituted with one or more substituents described herein. In one example, heteroaryl includes 5-6 membered heteroaryl groups. Heteroaryls includes 5 to 6 membered aromatic rings containing one, two, three or four heteroatoms selected from oxygen, nitrogen and sulfur.
  • a “solvate” refers to an association or complex of one or more solvent molecules and a compound of the invention.
  • solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
  • hydrate refers to the complex where the solvent molecule is water.
  • TLC thin layer chromatography
  • treat refers to therapeutic, prophylactic, palliative or preventative measures.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable.
  • Treatment can also mean prolonging survival as compared to expected survival if not receiving treatment.
  • Those in need of treatment include those already with the condition or disorder, as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
  • phrases "therapeutically effective amount” or “effective amount” mean an amount of a compound described herein that, when administered to a mammal in need of such treatment, sufficient to (i) treat or prevent the particular disease, condition, or disorder, (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, condition, or disorder, or (iii) prevent or delay the onset of one or more symptoms of the particular disease, condition, or disorder described herein.
  • the amount of a compound that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the mammal in need of treatment, but can nevertheless be routinely determined by one skilled in the art.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by abnormal or unregulated cell growth.
  • a “tumor” comprises one or more cancerous cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • squamous cell cancer e.g., epithelial squamous cell cancer
  • lung cancer including small- cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, skin cancer, including melanoma, as well as head and neck cancer.
  • NSCLC non-small cell lung cancer
  • adenocarcinoma of the lung and squamous carcinoma of the lung cancer of the peritoneum, hepatocellular cancer
  • phrases "pharmaceutically acceptable” indicates that the substance or composition is compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • phrases "pharmaceutically acceptable salt,” as used herein, refers to pharmaceutically acceptable organic or inorganic salts of a compound described herein.
  • the compounds described herein also include other salts of such compounds that are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds described herein and/or for separating enantiomers of compounds described herein.
  • mammal means a warm-blooded animal that has or is at risk of developing a disease described herein and includes, but is not limited to, guinea pigs, dogs, cats, rats, mice, hamsters, and primates, including humans.
  • compound of this invention and “compounds of the present invention”, unless otherwise indicated, include compounds of Formulas I, II, III, IV, V, VI, and VII, stereoisomers, tautomers, solvates, prodrugs and salts (e.g., pharmaceutically acceptable salts) thereof.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds of Formulas I, II, III, IV, V, VI, and VII wherein one or more hydrogen atoms are replaced deuterium or tritium, or one or more carbon atoms are replaced by a 13 C- or 14 C-enriched carbon are within the scope of this invention.
  • Y is selected from N, NR 10 , CR 11 and CHR 11 ;
  • Z is selected from N, NR 12 , CR 13 and CHR 13 ;
  • L is selected from O, NR 14 , S, C(-O) and CR 15 R 16 ;
  • R and R" are independently selected from hydrogen, halogen, CN, Ci-C 3 alkyl, C1-C3 alkoxy and C 2 -C 3 alkynyl;
  • R is selected from hydrogen, halogen and C1-C3 alkyl
  • R 4 is selected from C 3 -C 5 cycloalkyl, C C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, phenyl, a 5-6 membered heteroaryl, or NR a R b , wherein the cycloalkyl, alkyl, alkenyl, alkynyl, phenyl and heteroaryl are optionally substituted with OR 0 , halogen, phenyl, C 3 -C 4 cycloalkyl, or C C4 alkyl optionally substituted with halogen;
  • R 5 is selected from hydrogen, C1-C3 alkyl and Ci-C 3 alkoxy, wherein the alkyl and alkoxy are optionally substituted with R d ;
  • R 6 is selected from hydrogen, Ci-C6 alkyl, Ci-Ce alkenyl, Q-C6 alkynyl, C 3 -C 6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), OR e , SR e , NR f R g , phenyl, Ci-C 4 alkyl, C1-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with R p ; R 7 is selected from hydrogen, C C 6 alkyl, Ci-C alkenyl, Ci-C ⁇ alkynyl, Q-
  • R is selected from hydrogen, Q-Q alkyl, C C 6 alkenyl, C C 6 alkynyl, C 3 -C 6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), OR", SR J , NR k R*, phenyl, Q-C4 alkyl, Q-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with R p ;
  • R 9 is selected from hydrogen, C!-C 6 alkyl, Ci-C 6 alkenyl, C!-C 6 alkynyl, Ci-Ce alkoxy, C 3 -C 6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, a 5 to 6 membered heteroaryl, and NR m R n , wherein the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), OR J , SR J , NR k R*, C C 4 alkyl, d-C 4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with R p ;
  • R 10 is selected from hydrogen and halogen
  • R 11 is selected from hydrogen and halogen, or
  • R 11 and R 14 together with the atoms to which they are attached form a 5-6 membered heterocyclyl or heteroaryl optionally substituted with oxo, or
  • R 11 and R 15 together with the atoms to which they are attached form a 5-6 membered carbocyclyl
  • R 12 is hydrogen
  • R 13 is selected from hydrogen and halogen
  • R 14 is selected from hydrogen and C C 4 alkyl
  • R 15 and R 16 are independently selected from hydrogen and Q-C3 alkyl
  • R a and R b are independently selected from hydrogen or C Cs alkyl optionally substituted by halogen, or
  • R a and R b are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C!-C 3 alkyl;
  • R c is selected from hydrogen and C!-C 6 alkyl optionally substituted by halogen;
  • each R d is selected from halogen and OH;
  • R e is selected from hydrogen and Q-C4 alkyl;
  • R f and R 8 are independently selected from hydrogen and Q-C4 alkyl optionally substituted with halogen;
  • R h and R 1 are independently selected from hydrogen and C 1 -C4 alkyl optionally substituted with halogen;
  • R J is selected from hydrogen and C 1 -C 4 alkyl
  • R k and R 1 are independently selected from hydrogen and Q-C 4 alkyl optionally substituted with halogen;
  • R m and R" are independently selected from hydrogen and C!-C4 alkyl optionally substituted with halogen;
  • each R p is independently selected from halogen, oxo, Q-C 4 alkyl and Q-C4 alkoxy, wherein the alkyl and alkoxy are optionally substituted with halogen.
  • W is selected from N, .
  • R 6 is selected from hydrogen, C!-C 6 alkyl, C C 6 alkenyl, C C 6 alkynyl, C 3 -C 6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), OR e , SR e , NR f R 8 , phenyl, Q-Q alkyl, C C 4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with R p .
  • R 6 is selected from C ⁇ - e alkyl, phenyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, phenyl and heteroaryl are optionally substituted with phenyl or C1-C 4 alkyl optionally substituted with R p .
  • R 6 is methyl, benzyl, phenyl or 1 -methyl- 1H- pyrazol-4-yl.
  • R is selected from hydrogen, C ⁇ -C(, alkyl, Q-Ce alkenyl, Q-Q alkynyl, Ci-C 6 alkoxy, C 3 -C 6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, a 5 to 6 membered heteroaryl, and NR h R', wherein the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), OR e , SR e , NR f R g , Q-C4 alkyl, C1-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with R p .
  • R 7 is selected from hydrogen, C ⁇ -C(, alkoxy and NR h R'.
  • R h and R 1 are independently selected from hydrogen and C 1 -C 4 alkyl optionally substituted with halogen.
  • R 7 is hydrogen, OCH 3 or NHCH3.
  • R is selected from hydrogen, Q-Q alkyl, Q-C6 alkenyl, C ! -C 6 alkynyl, C 3 -C 6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), OR J , SR J , NR k R*, phenyl, Q-C4 alkyl, Q-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with R p .
  • R 8 is C - C 6 alkyl.
  • R is methyl, ethyl, methyl, ethyl, methyl
  • R 9 is selected from hydrogen, C ⁇ -C(, alkyl, C C 6 alkenyl, C C 6 alkynyl, C C 6 alkoxy, C 3 -C 6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, a 5 to 6 membered heteroaryl, and NR m R n , wherein the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), OR J , SR J , NR k R', Q-C4 alkyl, Q-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with R p .
  • R 9 is selected from hydrogen, Cy-Ce alkoxy, a 3 to 6 membered heterocyclyl, and NR m R n , wherein the alkoxy and heterocyclyl are optionally substituted with C C 4 alkyl optionally substituted R p .
  • R m and R n are independently selected from hydrogen and C1-C4 alkyl optionally substituted with halogen.
  • each R p is independently selected from halogen, oxo, Ct-Q alkyl and C 1 -C 4 alkoxy, wherein the alkyl and alkoxy are optionally substituted with halogen.
  • R 9 is hydrogen, OCH 3 , OCH 2 CH 3 , NHCH 3 , N(CH 3 ) 2 , piperazin-l-yl, 4- methylpirperazin-l-yl or tert-butyl 4-piperazine-l-carboxylate.
  • Y is selected from N, NR 10 , CR 11 and CHR 11 . In certain embodiments, Y is CR 11 . In certain embodiments, R 11 is selected from hydrogen and halogen. In certain embodiments, Y is CH or CC1.
  • Z is selected from N, NR 12 , CR 13 and CHR 13 .
  • Z is CR .
  • R is hydrogen.
  • R is selected from hydrogen and halogen.
  • R is hydrogen.
  • Z is CH.
  • the residue: wherein the wavy line represents the point of attachment of the residue to L, is selected from:
  • wav line represents the point of attachment of the residue to L, is selected from:
  • L is selected from O and NR 14 .
  • R 14 is selected from hydrogen and Ci-C 4 alkyl.
  • L is O, NH, N(CH 3 ) or N(CH 2 CH 3 ).
  • L is O.
  • L is NR 14 .
  • R 14 is selected from hydrogen and C C 4 alkyl.
  • R 14 is hydrogen.
  • R 14 is Ci-C 4 alkyl.
  • L is NH, N(CH 3 ) or N(CH 2 CH 3 ).
  • L is N(CH 3 ) or N(CH 2 CH 3 ).
  • L is NH.
  • R and R are independently selected from hydrogen, halogen, CN, Ci-C 3 alkyl, Ci-C 3 alkoxy or C 1 -C 3 alkynyl.
  • R 1 and R 2 are independently selected from hydrogen, halogen, CN, C C 3 alkyl or C C 3 alkoxy.
  • R , R and R are independently selected from hydrogen, halogen or 1 2 3
  • R , R and R are independently selected from hydrogen, F, CI or methyl.
  • R 1 is hydrogen, halogen, CN, C C 3 alkyl, C1-C3 alkoxy or C 2 -C 3 alkynyl. In certain embodiments, R 1 is hydrogen, halogen, CN, Q-C3 alkyl or Ci-C 3 alkoxy. In certain embodiments, R 1 is hydrogen or halogen. In certain embodiments, R 1 is hydrogen, CI or F.
  • R 1 is hydrogen
  • R 1 is halogen. In certain embodiments, R 1 is F or CI.
  • R 1 is C!-C 3 alkyl. In certain embodiments, R 1 is methyl.
  • R is hydrogen, halogen, CN, C C 3 alkyl, Q-C3 alkoxy or C 2 -C 3 alkynyl. In certain embodiments, R is hydrogen, halogen, CN, Ci-C 3 alkyl
  • R is halogen or CN. In certain embodiments, R is CI, F or CN. In certain embodiments, R 2 is CI or CN.
  • R is hydrogen
  • R is halogen. In certain embodiments, R is F or CI.
  • R is CI
  • R is C1-C3 alkyl. In certain embodiments, R is methyl.
  • R is hydrogen, halogen or C1-C3 alkyl. In certain embodiments, R is hydrogen.
  • R is halogen. In certain embodiments, R is F or CI.
  • R and R are F and R is hydrogen.
  • R is hydrogen and R and R are F.
  • R is hydrogen, R is F and R is CI.
  • R is F and R is CI and R is hydrogen.
  • R 1 is CI and R 2 is F and R 3 is hydrogen.
  • R is F and R and R are hydrogen.
  • R and R are hydrogen and R is F.
  • R and R are hydrogen and R is CI.
  • R is CI and R and R are hydrogen.
  • R 1 , R 2 and R 3 are F.
  • R is F and R is methyl and R is hydrogen.
  • R is methyl and R is F and R is hydrogen.
  • R 2 is F and R 1 and R 3 are hydrogen.
  • R is CI, R is ethynyl and R is hydrogen.
  • R is F
  • R is ethynyl
  • R is hydrogen
  • R 1 and R 3 are hydrogen and R 2 is CN.
  • R 1 is F
  • R 2 is CN
  • R 3 is hydrogen.
  • R 1 is CI
  • R 2 is CN
  • R 3 is hydrogen
  • R is methyl and R and R are hydrogen.
  • wav line represents the point of attachment of the residue to L, is selected from:
  • R 4 is selected from C 3 -C 5 cycloalkyl, C ! -C 6 alkyl, C 2 - C 6 alkenyl, C 2 -C 6 alkynyl, phenyl, a 5-6 membered heteroaryl, or NR a R b , wherein the cycloalkyl, alkyl, alkenyl, alkynyl, phenyl and heteroaryl are optionally substituted with OR c , halogen, phenyl, C 3 -C cycloalkyl, or Ci-C 4 alkyl optionally substituted with halogen.
  • R 4 is selected from Ci-Ce alkyl, a 5-6 membered heteroaryl, or NR a R b , wherein the alkyl and heteroaryl are optionally substituted with halogen or C 3 -C 4 cycloalkyl.
  • R a and R b are independently selected from hydrogen or C Cs alkyl optionally substituted by halogen.
  • R a and R b are Q-Cs alkyl.
  • R a and R b are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or Ci- C 3 alkyl.
  • R a and R b are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl. In certain embodiments, R a and R b are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl, wherein the heterocyclyl contains one nitrogen heteroatom. In certain embodiments, R a and R b are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl, wherein the heterocyclyl is pyrrolidine. In certain embodiments, R 4 is propyl, isobutyl, -CH 2 CH 2 CH 2 F, cyclopropylmethyl, -N(CH 3 ) 2 , pyrrolidine, or furan-2-yl.
  • R 4 is C 3 -C 4 cycloalk l, Q-Q alkyl optionally substituted with halogen or C 3 -C 4 cycloalkyl, or NR a R b .
  • R a and R b are independently selected from hydrogen and Q-Cs alkyl.
  • R 4 is C 3 -C 5 cycloalkyl, C C6 alkyl, C 2 -C 6 alkenyl or C 2 -C 6 alkynyl, wherein the cycloalkyl, alkyl, alkenyl and alkynyl are optionally substituted with OR c , halogen or C3-C4 cycloalkyl.
  • R 4 is cyclopropyl, ethyl, propyl, butyl, isobutyl, -CH 2 C1, -CH 2 CF 3 , -CH 2 CH 2 CH 2 F, ⁇ CH 2 CH 2 CF 3 , phenylmethyl, cyclopropylmethyl, phenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2,5-difluorophenyl, 4-chloro-3- trifluoromethylphenyl, 1 -methyl- lH-imidazol-4-yl, furan-2-yl, pyridin-2-yl, pyridin-3-yl, thiophen-2-yl, -NHCH 2 CH 3 , -NHCH 2 CH 2 CH 3 , -N(CH 3 )CH 2 CH 3 , -N(CH 3 ) 2 , or pyrrolidine.
  • R 4 is cyclopropyl, propyl, butyl, isobutyl, -CH 2 C1, -CH 2 CF 3 , -CH 2 CH 2 CH 2 F, -CH 2 CH 2 CF 3 , cyclopropylmethyl, -NHCH 2 CH 2 CH 3 , -N(CH 3 )CH 2 CH 3 , -N(CH 3 ) 2 , or pyrrolidine.
  • R 4 is cyclopropyl, propyl, butyl, isobutyl, -CH 2 C1, - €H 2 CF 3 , -CH 2 CH 2 CH 2 F, -CH 2 CH 2 CF 3 , cyclopropylmethyl or -NHCH 2 CH 2 CH 3 .
  • R 4 is propyl, butyl, isobutyl, -CH 2 CH 2 CH 2 F, -CH 2 CH 2 CF 3 or cyclopropylmethyl.
  • R 4 is C 3 -C 5 cycloalkyl or C C 6 alkyl optionally substituted with OH, halogen or C 3 -C 4 cycloalkyl.
  • R 4 is C 3 -C 5 cycloalkyl. In certain embodiments, R 4 is C 3 -C 4 cycloalkyl. In certain embodiments, R 4 is cyclopropyl or cyclobutyl. In certain embodiments, R 4 is cyclopropyl.
  • R 4 is Ci-C 6 alkyl. In certain embodiments, R 4 is ethyl, propyl, butyl or isobutyl. In certain embodiments, R 4 is propyl or isobutyl.
  • R 4 is C C 6 alkyl optionally substituted with halogen.
  • R 4 is -CF 3 , -CH 2 C1, -CH 2 CF 3 , -CH 2 CH 2 CH 2 F, -CH 2 CH 2 CF 3 , -CF 2 CF 3 or -CF 2 CF 2 CF 3 .
  • R 4 is -CH 2 CH 2 CH 2 F or -CH 2 CH 2 CF 3 .
  • R 4 is -CH 2 CH 2 CH 2 F.
  • R 4 is Ci-Ce alkyl optionally substituted with OH, halogen or C 3 -C 4 cycloalkyl.
  • R 4 is cyclopropylmethyl (-CH 2 - cyclopropyl) or cyclobutylmethyl (-CH 2 -cyclobutyl).
  • R 4 is cyclopropylmethyl .
  • R 4 is Q-C 6 alkyl optionally substituted with phenyl. In certain embodiments, R 4 is phenylmethyl.
  • R 4 is phenyl optionally substituted with OR c , halogen, C 3 -C 4 cycloalkyl, or C 1 -C 4 alkyl optionally substituted with halogen. In certain embodiments, R 4 is phenyl optionally substituted with halogen. In certain embodiments, R 4 is phenyl optionally substituted with C C 4 alkyl optionally substituted with halogen. In certain embodiments, R 4 is phenyl optionally substituted with halogen and Q-C 4 alkyl optionally substituted with halogen. In certain embodiments, R 4 is phenyl.
  • R 4 is phenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2,5- difluorophenyl or 4-chloro-3-trifluoromethylphenyl.
  • R 4 is a 5-6 membered heteroaryl optionally substituted with OR c , halogen, C 3 -C 4 cycloalkyl or C t -C4 alkyl optionally substituted with halogen.
  • R 4 is a 5-6 membered heteroaryl optionally substituted with C!-C 4 alkyl.
  • R 4 is a 5-6 membered heteroaryl, wherein the heteroaryl contains one or two heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur.
  • R 4 is a 5-6 membered heteroaryl, wherein the heteroaryl is imidazolyl, furanyl, pyridinyl or thiophenyl.
  • R 4 is 1- methyl-lH-imidazol-4-yl, furan-2-yl, pyridin-2-yl, pyridin-3-yl or thiophen-2-yl.
  • R 4 is furan-2-yl.
  • R 4 is NR a R b .
  • R a and R b are independently selected from hydrogen and Q-Cs alkyl.
  • R a is hydrogen.
  • R a is Ci-C 6 alkyl.
  • R a is ethyl or propyl.
  • R 4 is selected from the group consisting of -NHCH 2 CH 3 , -NHCH 2 CH 2 CH 3 , -N(CH 3 )CH 2 CH 3 and -N(CH 3 ) 2 .
  • R 4 is -N(CH 3 ) 2 .
  • R a and R b together with the nitrogen to which they are attached form a 3 to 6 membered heterocyclyl ring. In certain embodiments, R a and R b together with the nitrogen to which they are attached form a 3 to 6 membered heterocyclic ring, wherein the heterocyclic ring contains one nitrogen heteroatom.
  • R 4 is pyrrolidine. In certain embodiments, R 4 is pyrrolidin-l-yl.
  • R 4 is selected from propyl, cyclopropylmethyl, -CH 2 CH 2 CH 2 F and phenyl. In a further embodiment, R 4 is selected from propyl, cyclopropylmethyl and -CH 2 CH 2 CH 2 F.
  • R 5 is selected from hydrogen, Q-C 3 alkyl and C ! -C 3 alkoxy, wherein the alkyl and alkoxy are optionally substituted with R d . In certain embodiments, R 5 is hydrogen.
  • R 11 and R 14 together with the atoms to which they are attached form a 5-6 membered heterocyclyl or heteroaryl optionally substituted with oxo.
  • This embodiment includes the following formula:
  • V is O, N or CH; x is 1 or 2; the dashed lines represent optional double bonds, and W, X, Z, R 1 , R 2 , R 3 , R 4 and R 5 are as defined herein.
  • compounds of the present invention have the following formulas:
  • R 11 and R 15 together with the atoms to which they are attached form a 5-6 membered carbocyclyl.
  • This embodiment includes the following formula:
  • V is O, N or CH; x is 1 or 2; the dashed lines represent optional double bonds, and W, X, Z, R 1 , R 2 , R 3 , R 4 and R 5 are as defined herein.
  • compounds of the present invention have the following formulas:
  • Y is CR 11 ;
  • R 5 is hydrogen
  • Y is CR 11 ;
  • L is selected from O and NR 14 ;
  • R 5 is hydrogen
  • R 6 is selected from C C 6 alkyl, phenyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, phenyl and heteroaryl are optionally substituted with phenyl or C1-C4 alkyl optionally substituted with R p ;
  • R 7 is selected from hydrogen, CrC 6 alkoxy and NR h R';
  • R 8 is Ci-Ce alkyl
  • R 9 is selected from hydrogen, Ci-C , alkoxy, a 3 to 6 membered heterocyclyl, and NR m R", wherein the alkoxy and heterocyclyl are optionally substituted with C C 4 alkyl optionally substituted R ;
  • R 11 is selected from hydrogen and halogen
  • R is hydrogen
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butyloxycarbonyl ("Boc”), benzyloxycarbonyl ("CBz”) and 9-fluorenylmethyleneoxycarbonyl ("Fmoc”).
  • Boc trifluoroacetyl
  • CBz benzyloxycarbonyl
  • Fmoc 9-fluorenylmethyleneoxycarbonyl
  • Schemes 1-18 show general methods for preparing the compounds described herein, as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
  • Scheme 1 shows a general method for preparing a compound 1.6, wherein R , R 2 , R 3 and R 4 are as defined herein.
  • a benzoic acid 1.1 is esterified to an alkyl benzoate 1.2 (wherein R is alkyl), e.g., by treatment with tnmethylsilyl diazomethane in MeOH or via Fischer esterification conditions, such as treatment with trimethylsilyl chloride ("TMSC1") in MeOH.
  • TMSC1 trimethylsilyl chloride
  • Reduction of nitro intermediate 1.2 to its amino analog 1.3 is performed using a standard condition, such as treatment with Pd/C and H 2 .
  • Bis-sulfonamide 1.4 is obtained by treatment of the aniline 1.3 with a sulfonyl chloride, R 4 S0 2 C1, in the presence of a base, such as NEt 3 , in an organic solvent, such as dichloromethane ("DCM"). Hydrolysis of compound 1.4 is accomplished under basic conditions, such as aqueous NaOH, in the appropriate solvent system, such as THF and/or MeOH, to provide a carboxylic acid 1.5.
  • This compound in a suitable solvent, such as THF is treated with diphenylphosphonic azide (“DPP A”) and a base such as triethylamine, and subsequently hydrolyzed to form an amine 1.6.
  • DPP A diphenylphosphonic azide
  • Scheme 2 describes the synthesis of aniline intermediates 2.7, wherein R 1 , R 2 ', R 3 , R 4 and R" are as defined herein.
  • a benzoic acid ester 2.1 is treated with an alkoxide,
  • Benzylation with an optionally substituted benzyl halide for example p- methoxybenzyl chloride, (wherein LG is a leaving group such as chloro, bromo, iodo, triflate, tosylate; and R" is hydrogen, C C 3 alkyl or Ci-C alkoxy; and in one example, R" is hydrogen, in another example, R" is OMe) in the presence of a base, such as sodium hydride, yields the protected sulfonamide ester 2.5, which is hydrolyzed with aqueous base, such as NaOH, to form the acid 2.6.
  • a base such as sodium hydride
  • Scheme 3 shows a procedure for generating the aniline intermediate 3.1, wherein R" and LG are defined in Scheme 2 and R 1 , R 2 , R 3 and R 4 are as defined herein, through protection of the sulfonamide moiety of aniline 1.6.
  • This transformation can be accomplished by treatment with an optionally substituted benzyl halide (e.g., p- methox benzyl chloride) and a base, such as sodium hydride.
  • Scheme 4 describes the synthesis of an aniline ester of Formula 1.3, wherein R , R and R J are defined herein and R is alkyl, such as methyl or ethyl, or benzyl.
  • R is alkyl, such as methyl or ethyl, or benzyl.
  • the amino group of an aniline 4.1 is protected by reacting with hexane-2,5-dione in the presence of a catalytic amount of an acid, such as p-toluenesulfonic acid, in a solvent, such as toluene, to form the 2,5-dimethylpyrrole derivative 4.2.
  • R 1', R2", and R 3 J are defined herein and R is small alkyl, such as methyl or ethyl, or benzyl.
  • R is small alkyl, such as methyl or ethyl, or benzyl.
  • the amino group of an aniline 4.1 is protected by reacting with 1,2- bis(chlorodimethylsilyl)ethane in the presence of a strong base, such as n-butyllithium, in a suitable solvent, such as THF, at low temperatures, e.g., -78°C, to form the l-aza-2,5- disilacyclopentane intermediate 5.1.
  • Scheme 6 describes another way of synthesizing an intermediate of Formula 1.6, wherein R 1 , R 2 , R 3 and R 4 are as defined herein.
  • Bis-sulfonamide 6.2 is obtained by treatment of the aniline 6.1 with a sulfonyl chloride, R 4 S0 2 C1, in the presence of a base, such as NEt 3 , in an organic solvent, such as dichloromethane.
  • Hydrolysis of compound 6.2 is accomplished under basic conditions, such as aqueous NaOH, in the appropriate solvent system, such as THF and/or MeOH, to provide the mono-sulfonamide 6.3.
  • This compound in a suitable solvent, such as ethanol is treated with a reducing agent, such as iron, and additional reagents, such as ammonium chloride, to form an amine 1.6.
  • Scheme 7 shows another way of preparing an intermediate of Formula 1.6. This transformation is accomplished by mono-sulfonylation of a diamino derivative 7.1 with a sulfonyl chloride, R 4 S0 2 C1, in the presence of a base, such as pyridine, in an organic solvent, such as dichloromethane.
  • a base such as pyridine
  • Scheme 8 describes the synthesis of an intermediate of Formula 8.2, a subset of Formula 1.6 compounds, wherein R 1 , R 3 and R 4 are as defined herein and R 2 is hydrogen. This transformation is accomplished by reduction of the chloro atom of compound 8.1 using reducing conditions, such as hydrogen in the presence of a palladium catalyst in a suitable solvent, such as ethanol.
  • Scheme 9 describes the synthesis of an intermediate of Formula 9.2, a subset of Formula 1.6 compounds, wherein R 2 , R 3 and R 4 are as defined herein and R 1 is hydrogen. This transformation is accomplished by reduction of the chloro atom of compound 9.1 using reducing conditions, such as hydrogen in the presence of a palladium catalyst in a suitable solvent, such as ethanol.
  • Scheme 10 shows a method for preparing nitrile-substituted aniline intermediates 10.2. Reaction of fluoronitrile 10.1 with the sodium salt of H 2 NS0 2 R 4 (generated by a strong base, such as sodium hydride) in a suitable solvent, such as dimethylsulfoxide or N-methylpyrrolidone, at elevated temperature, results in the formation of intermediate 10.2.
  • a strong base such as sodium hydride
  • Scheme 11 shows a general method for preparing compounds 11.2 and 11.6, wherein W, X, Y, Z, R 1 , R 2 , R 3 , R 4 and R 8 are as defined herein.
  • Amino heterocycle 11.1 can be coupled with aniline 1.6 under standard Buchwald coupling conditions utilizing a Pd catalyst, such as Pd(dba) 2 , and a ligand, such as BINAP, to give biarylamine 11.2.
  • aniline 1.6 can be protected with a suitable protecting group, for example PMB, with a suitable base, such as NaH, in a solvent, such as DMF, to give 11.3, and then coupled to give 11.4 and deprotected, for example with neat TFA at room temperature or DCM/TFA at elevated temperatures to provide 11.2.
  • Biaryl amine 11.4 may also be alkylated on nitrogen utilizing a suitable base, preferably NaH, and an alkylating agent, such as Mel, Etl, allyl bromide or cyclopropylmethyl methanesulfonate. Subsequent deprotection provides compound 11.6.
  • Scheme 12 shows a general method for preparing compound 12.2, wherein R 1 , R 2 , R 3 , R 4 , R 6 and R 14 are as defined herein.
  • Biaryl amine 12.1 can be chlorinated in the presence of a chlorinating reagent, such as n-chlorosuccinimide, in a variety of solvents, including THF, DMF or CHC1 3 , to provide compound 12.2.
  • a chlorinating reagent such as n-chlorosuccinimide
  • PG is a protecting group
  • Scheme 13 shows a general method for preparing compounds 13.2 and 13.3, wherein R and R is as defined herein.
  • Quinoxalinone 13.1 can be alkylated with an alkylating agent, such as Mel, EtI, allyl bromide or cyclopropylmethyl methanesulfonate, utilizing a base, such as NaH, in a solvent, such as DMF, giving a mixture of heterocycles 13.2 and 13.3.
  • Heterocycles 13.2 and 13.3 can be separated and coupled separately in the Buchwald reaction according to Scheme 11.
  • Scheme 14 describes the preparation of nitriles 14.3, wherein W, X, R 1 , R 3 , R 4 and R 11 are as defined herein.
  • An alkali metal salt of a hydroxyl-containing heterocycle 14.1 is generated using a suitable base, such as sodium hydride, in an appropriate solvent, such as THF, DMF or NMP.
  • a suitable base such as sodium hydride
  • an appropriate solvent such as THF, DMF or NMP.
  • the resulting phenoxide is treated with an optionally substituted difluorobenzonitrile at ambient or elevated temperature to afford ether intermediate 14.2.
  • Nitrile 14.3 is obtained by reacting, at elevated temperature, intermediate 14.2 with an anion of an optionally protected sulfonamide or sulfamide, formed by the action of a strong base, such as sodium hydride, in a solvent, such as NMP, DMF, or DMSO.
  • a strong base such as sodium hydride
  • Scheme 15 describes the preparation of nitriles 15.3, wherein W, X, R 1 , R 3 , R 4 and R 11 are as defined herein.
  • An aniline is coupled to a heterocycle 15.1 under standard Buchwald coupling conditions utilizing a Pd catalyst, such as Pd(dba) 2 , and a ligand, such as ⁇ , to give biarylamine 15.2.
  • Nitrile 15.3 is obtained by reacting, at elevated temperature, intermediate 15.2 with an anion of an optionally protected sulfonamide or sulfamide, formed by the action of a strong base, such as sodium hydride, in a solvent, such as NMP DMF, or DMSO.
  • Scheme 16 shows a general method for preparing compounds 16.2 and 16.3, wherein R 6 , R m and R n are as defined herein.
  • a quinazolinone such as 16.1 can be treated with a coupling agent, such as BOP, in a suitable solvent, such as DMF or acetonitrile, and subsequently treated with an amine to provide quinazoline amines 16.3.
  • a coupling agent such as BOP
  • a suitable solvent such as DMF or acetonitrile
  • 16.1 can be treated with the coupling agent FLATU in a suitable solvent, such as DMF or acetonitrile, and subsequently treated with an amine to provide quinazolines 16.2.
  • Scheme 17 shows a general method for preparing sulfamides 17.2, wherein W, X, R 1 , R 2 , R 3 , R 4 , R 11 , R a and R b are as defined herein.
  • a sulfonamide 17.1 is treated with a sulfamoyl chloride in a solvent, such as DMF, and subsequently hydrolyzed to a sulfamide 17.2 by addition of a base and water, such as sodium h droxide.
  • Scheme 18 shows a general method for preparing compounds 18.2, wherein R z is Ci-C 6 alkyl optionally substituted with halogen, oxo, OR J , SR j , NR k R', phenyl, d-C 4 alkyl, C ! -C 4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with R p .
  • 4-Chloroquinazolinone 18.1 can be treated with a nucleophile, such as NaOR z , in a suitable solvent, such as R z OH, to provide 4-alkoxyquinazole 18.2.
  • reaction products from one another and/or from starting materials.
  • the desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art.
  • separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography.
  • Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography.
  • SMB simulated moving bed
  • Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers.
  • an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
  • Enantiomers can also be separated by use of a chiral HPLC column.
  • a single stereoisomer e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S. Stereochemistry of Organic Compounds. New York: John Wiley & Sons, Inc., 1994; Lochmuller, C. H., et al. "Chromatographic resolution of enantiomers: Selective review.” J. Chromatogr., 113(3) (1975): pp. 283-302).
  • Racemic mixtures of chiral compounds described herein may be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. See: Wainer, Irving W., Ed. Drug Stereochemistry: Analytical Methods and Pharmacology. New York: Marcel Dekker, Inc., 1993.
  • diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, a-methyl- ⁇ -phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid.
  • the diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography.
  • the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and Wilen, S. Stereochemistry of Organic Compounds. New York: John Wiley & Sons, Inc., 1994, p. 322).
  • Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer.
  • a method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, a-methoxy-a- (trifluoromethyl)phenyl acetate (Jacob III, Peyton. "Resolution of ( ⁇ )-5-Bromonornicotine. Synthesis of (R)- and (S)-Nornicotine of High Enantiomeric Purity.” J. Org. Chem. Vol. 47, No. 21 (1982): pp.
  • chiral esters such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, a-methoxy-a- (trifluoromethyl)phenyl acetate (Jacob III, Peyton. "Resolution of ( ⁇ )-5-Bromonornicotine. Synthesis of (R)- and
  • Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (WO 96/15111).
  • a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase (Lough, W.J., Ed. Chiral Liquid Chromatography. New York: Chapman and Hall, 1989; Okamoto, Yoshio, et al. "Optical resolution of dihydropyridine enantiomers by high-performance liquid chromatography using phenylcarbamates of polysaccharides as a chiral stationary phase.” J. of Chromatogr. Vol. 513 (1990): pp. 375-378).
  • Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
  • B-Raf mutant protein 447-717 (V600E) was co-expressed with the chaperone protein Cdc37, complexed with Hsp90 (Roe, S. Mark, et al. "The Mechanism of Hsp90 Regulation by the Protein Kinase-Specific Cochaperone p50cdc37.” Cell. Vol. 116 (2004): pp. 87-98; Stancato, LF, et al. "Raf exists in a native heterocomplex with Hsp90 and p50 that can be reconstituted in a cell free system.” J. Biol. Chem. 268(29) (1993): pp. 21711-21716).
  • the compounds described herein may be administered by any convenient route appropriate to the condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal.
  • the compounds may be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches, etc.
  • Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents. If parenteral administration is desired, the compositions will be sterile and in a solution or suspension form suitable for injection or infusion.
  • a typical formulation is prepared by mixing a compound described herein and a carrier or excipient.
  • Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C, et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005.
  • the formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound described herein or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
  • buffers stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound described herein or pharmaceutical composition thereof) or aid in the manufacturing of the
  • One embodiment includes a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer or pharmaceutically acceptable salt thereof.
  • a further embodiment provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer or pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier or excipient.
  • Another embodiment of the present invention provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of a hyperproliferative disease.
  • Another embodiment of the present invention provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of cancer.
  • Another embodiment of the present invention provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of kidney disease.
  • a further embodiment of the present invention provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of polycystic kidney disease.
  • the invention includes methods of treating or preventing disease or condition by administering one or more compounds of this invention, or a stereoisomer or pharmaceutically acceptable salt thereof.
  • a human patient is treated with a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle in an amount to detectably inhibit B-Raf activity.
  • a human patient is treated with a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle in an amount to detectably inhibit B-Raf activity.
  • a method of treating a hyperproliferative disease in a mammal comprising administering a therapeutically effective amount of the compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, to the mammal is provided.
  • kidney disease in another embodiment, a method of treating kidney disease in a mammal comprising administering a therapeutically effective amount of the compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, to the mammal is provided.
  • the kidney disease is polycystic kidney disease.
  • a method of treating or preventing cancer in a mammal in need of such treatment comprises administering to said mammal a therapeutically effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof.
  • the cancer is selected from breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, NSCLC, small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system, Hodgkin's and leukemia.
  • a method of treating or preventing cancer in a mammal in need of such treatment comprises administering to said mammal a therapeutically effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof.
  • Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
  • Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of kidney disease.
  • the kidney disease is polycystic kidney disease.
  • a method of preventing or treating cancer comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds having anti-cancer properties.
  • a method of preventing or treating cancer comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds having anti-cancer properties.
  • the cancer is a sarcoma.
  • the cancer is a carcinoma.
  • the carcinoma is squamous cell carcinoma.
  • the carcinoma is an adenoma or adenocarcinoma.
  • a method of treating or preventing a disease or disorder modulated by B-Raf comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof.
  • diseases and disorders include, but are not limited to, cancer.
  • the cancer is selected from breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, NSCLC, small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system, Hodgkin's and leukemia.
  • a method of treating or preventing a disease or disorder modulated by B-Raf comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof.
  • a method of preventing or treating kidney disease comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds.
  • a method of preventing or treating polycystic kidney disease comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds.
  • Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
  • the cancer is selected from breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, NSCLC, small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system, Hodgkin's and leukemia.
  • oral lip,
  • Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
  • Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of polycystic kidney disease.
  • the kidney disease is polycystic kidney disease.
  • Another embodiment of the present invention provides the compounds of
  • Another embodiment of the present invention provides the compounds of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of a hyperproliferative disease.
  • the hyperproliferative disease is cancer (as further defined and may be individually selected from those above).
  • kidney disease is polycystic kidney disease.
  • the compounds described herein and stereoisomers and pharmaceutically acceptable salts thereof may be employed alone or in combination with other therapeutic agents for treatment.
  • the compounds described herein may be used in combination with one or more additional drugs, for example an anti-hyperproliferative (or anti-cancer) agent that works through action on a different target protein.
  • the second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound described herein, such that they do not adversely affect each other.
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • the compounds may be administered together in a unitary pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order. Such sequential administration may be close in time or remote in time.
  • a “chemotherapeutic agent” is a chemical compound useful in the treatment of cancer, regardless of mechanism of action.
  • Chemotherapeutic agents include compounds used in "targeted therapy” and conventional chemotherapy.
  • a number of suitable chemotherapeutic agents to be used as combination therapeutics are contemplated for use in the methods of the present invention.
  • the present invention contemplates, but is not limited to, administration of numerous anticancer agents, such as: agents that induce apoptosis; polynucleotides (e.g., ribozymes); polypeptides (e.g., enzymes); drugs; biological mimetics; alkaloids; alkylating agents ⁇ antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal antibodies conjugated with anticancer drugs, toxins, and/or radionuclides; biological response modifiers (e.g., interferons [e.g., IFN-a, etc.] and interleukins [e.g., IL-2, etc.], etc.); adoptive immunotherapy agents; hematopoietic growth factors; agents that induce tumor cell differentiation (e.g., all-trans-retinoic acid, etc.); gene therapy reagents; antisense therapy reagents and nucleotides; tumor vaccines; inhibitors of angiogenesis, and the like.
  • chemotherapeutic agents include Erlotinib (TARCEVA®, Genentech/OSI Pharm.), Bortezomib (VELCADE®, Millennium Pharm.), Fulvestrant (FASLODEX®, AstraZeneca), Sunitinib (SUTENT®, Pfizer), Letrozole (FEMARA®, Novartis), Imatinib mesylate (GLEEVEC®, Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin (Eloxatin®, Sanofi), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNE®, Wyeth), Lapatinib (TYKERB®, GSK572016, Glaxo Smith Kline), Lonafarnib (SCH 66336), Sorafenib (NEXAVAR®, Bayer), Irinotecan (CAMPTOSAR®, Pfizer) and
  • 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, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6- diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), morpholino-doxorubicin, cyanomo holino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin
  • chemotherapeutic agent also included in the definition of "chemotherapeutic agent” are: (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, 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, RTVISOR® (vorozole), FEMARA®
  • chemotherapeutic agent therapeutic 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).
  • therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RIT
  • Humanized monoclonal antibodies with therapeutic potential as chemotherapeutic agents in combination with the Raf inhibitors of the invention include: alemtuzumab, apolizumab, aselizumab, atlizumab, bapineuzumab, bevacizumab, 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, moto
  • Activity of human recombinant B-Raf protein may be assessed in vitro by assay of the incorporation of radio labeled phosphate to recombinant MAP kinase (MEK), a known physiologic substrate of B-Raf, according to US 2004/0127496 and WO 03/022840.
  • Catalytically active human recombinant B-Raf protein is obtained by purification from sf9 insect cells infected with a human B-Raf recombinant baculovirus expression vector.
  • V600E full-length B-Raf The activity/inhibition of V600E full-length B-Raf was estimated by measuring the incorporation of radio labeled phosphate from [ ⁇ - 33 ⁇ ] ATP into FSBA-modified wild-type MEK.
  • the 30- ⁇ _ ⁇ assay mixtures contained 25mM Na Pipes, pH 7.2, lOOmM KC1, lOmM MgCl 2 , 5mM ⁇ -glycerophosphate, ⁇ Na Vanadate, 4 ⁇ ATP, 500 nCi [ ⁇ - 33 P]ATP, ⁇ ⁇ FSBA-MEK and 20nM V600E full-length B-Raf. Incubations were carried out at 22°C in a Costar 3365 plate (Corning).
  • the B-Raf and FSBA-MEK were preincubated together in assay buffer at 1.5x (20 ⁇ ⁇ of 30nM and 1.5 ⁇ , respectively) for 15 minutes, and the assay was initiated by the addition of 10 ⁇ , of 10 ⁇ ATP.
  • the assay mixtures were quenched by the addition of 100 ⁇ iL of 25% TCA, the plate was mixed on a rotary shaker for 1 minute, and the product was captured on a Perkin-Elmer GF/B filter plate using a Tomtec Mach III Harvester. After sealing the bottom of the plate, 35 ⁇ ⁇ of Bio-Safe II (Research Products International) scintillation cocktail were added to each well and the plate was top-sealed and counted in a Topcount NXT (Packard).
  • Step A A 1 L flask was charged with 2,6-difluoro-3-nitrobenzoic acid (17.0 g, 83.7 mmol) and MeOH (170 mL, 0.5M). The flask was placed in a cold water bath, and an addition funnel charged with a 2M solution of trimethylsilyl (“TMS") diazomethane in hexanes (209 mL, 419 mmol) was attached to the flask. The TMS diazomethane solution was added slowly to the reaction flask over the course of 2 hours. A large excess of reagent was required in order for the reaction to reach completion as determined by the ceased evolution of N 2 upon further addition of reagent. The volatiles were removed in vacuo to afford crude methyl 2,6-difluoro-3-nitrobenzoate as a solid (18.2 g). The material was taken directly to Step B.
  • TMS trimethylsilyl
  • Step B 10% (wt.) Pd on activated carbon (4.46 g, 4.19 mmol) was added to a 1 L flask charged with methyl 2,6-difluoro-3-nitrobenzoate (18.2 g, 83.8 mmol) under a nitrogen atmosphere. To the flask was added EtOH (350 mL, 0.25 M), and H 2 gas was passed through the mixture for 15 minutes. The reaction mixture was stirred under two H 2 balloons overnight. The balloons were recharged with H 2 gas and the mixture was stirred an additional 4 hours. Upon consumption of the starting material and intermediate hydroxylamine as determined by TLC, N 2 gas was flushed through the reaction mixture.
  • Step C Propane- 1-sulfonyl chloride (23.46 mL, 209.3 mmol) was slowly added to a solution of methyl 3-amino-2,6-difluorobenzoate (15.66 g, 83.7 mmol) and triethylamine (35.00 mL, 251.1 mmol) in CH 2 C1 2 (175 mL, 0.5M) maintained in a cool water bath. The reaction mixture was stirred for 1 hour at room temperature. Water (300 mL) was added and the organic layer was separated, washed with water (2 X 300 mL) and brine (200 mL), then dried (Na 2 S04), filtered and concentrated to an oil.
  • a IN aqueous NaOH solution 150 mL, 150 mmol was added to a solution of methyl 2,6-difluoro-3-(N-(propylsulfonyl)propylsulfonamido)benzoate (20.0 g, 50.1 mmol) in 4:1 THF/MeOH (250 mL, 0.2M).
  • the reaction mixture was stirred at room temperature overnight. The majority of the organic solvents were removed in vacuo (water bath temperature 35°C).
  • IN HC1 150 mL was slowly added to the mixture, and the resulting solid was filtered and rinsed with water (4 X 50 mL).
  • the aqueous layer was washed with ethyl acetate (50 mL) and then acidified with concentrated HC1 to a pH of about 1.
  • the acidified aqueous layer was extracted with ethyl acetate (2 x 50 mL), and the combined ethyl acetate extracts were dried (over Na 2 S0 4 ), filtered and concentrated.
  • the resulting residue was triturated with hexanes to afford 2,6-difluoro-3-(N-(propylsulfonyl)propyl- sulfonamido)benzoic acid as a solid (0.948 g, 74% yield).
  • Step A Into a 20-L 4-neck round flask was placed a solution of 2-chloro-4- fluorobenzenamine (1300 g, 8.82 mol, 1.00 equiv, 99%) in toluene (10 L), 4- methylbenzenesulfonic acid (3.1 g, 17.84 mmol, 99%), and hexane-2,5-dione (1222.5 g, 10.62 mol, 1.20 equiv, 99%). The resulting solution was heated to reflux for 1 hour in an oil bath and cooled. The pH value of the solution was adjusted to 8 with sodium carbonate (1 mol/L). The resulting mixture was washed with water (5000 mL) and concentrated under vacuum.
  • Step B Into a 5000-mL 4-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed a solution of l-(2-chloro-4-fluorophenyl)- 2,5-dimethyl-lH-pyrrole (390 g, 1.65 mol, 1.00 equiv, 95%) in tetrahydrofuran (2000 mL). The reaction vessel was cooled to -78°C.
  • n-BuLi 800 mL, 1.10 equiv, 2.5%) was added dropwise with stirring to the reaction vessel over 80 minutes, and methyl carbonochloridate (215.5 g, 2.27 mol, 1.20 equiv, 99%) was added dropwise with stirring over 90 minutes.
  • the reaction solution was further stirred for 60 minutes at -78°C and quenched by the addition of NH 4 Cl/water (1000 mL). The resulting solution was extracted with ethyl acetate (1500 mL).
  • Step C A solution of methyl 2-chloro-3-(2,5-dimethyl-lH-pyrrol-l-yl)-6- fluorobenzoate (1500 g, 5.05 mol, 1.00 equiv, 95%) in ethanol/H 2 0 (7500/2500 mL), NH 2 OH-HCl (5520 g, 79.20 mol, 15.00 equiv, 99%), and triethylamine (2140 g, 20.98 mol, 4.00 equiv, 99%) was placed into five 5000-mL 4-neck round-bottom flasks.
  • Step D A solution of methyl 3-amino-2-chloro-6-fluorobenzoate (980 g, 4.76 mol, 1.00 equiv, 99%) in dichloromethane (8000 mL) was placed into four 5000-mL 4-neck round-bottom flasks. Triethylamine (1454 g, 14.25 mol, 3.00 equiv, 99%) was added dropwise to this reaction vessel with stirring at 0°C over 80 minutes followed by the addition of propane- 1-sulfonyl chloride (1725 g, 11.94 mol, 2.50 equiv, 99%). The resulting solution was stirred at room temperature for 2 hours, diluted with water (1000 mL).
  • Step E A solution of methyl 2-chloro-6-fluoro-3-
  • Step A A flame dried flask equipped with a stir bar and rubber septum was charged with 4-chloro-2-fluoroaniline (5.00 g, 34.35 mmol) and anhydrous THF (170 mL). This solution was chilled to -78°C, and n-BuLi (14.7 mL, 1.07 eq. of 2.5M solution in hexanes) was then added over a 15 minute period. This mixture was stirred at -78°C for 20 minutes, and then a THF solution (25 mL) of l,2-bis(chlorodimethylsilyl)ethane (7.76 g, 1.05 eq.) was added slowly (over a 10 minute period) to the reaction mixture.
  • Step B Benzyl 3-amino-6-chloro-2-fluorobenzoate (4.3 g, 15.37 mmol) was dissolved in dry dichloromethane (270 mL). Triethylamine (5.36 mL, 2.5 eq.) was added, and the mixture was chilled to 0°C. Propane- 1-sulfonyl chloride (3.63 mL, 32.3 mmol, 2.1 eq.) was then added via syringe, and a precipitate resulted. Once the addition was complete, the mixture was allowed to warm to room temperature, and the starting material was consumed as determined by TLC (3:1 hexanerethyl acetate).
  • Step C Benzyl 6-chloro-2-fluoro-3-(N-(propylsulfonyl)propylsulfonamido) benzoate (5.4 g, 10.98 mmol) was dissolved in THF (100 mL) and 1M aqueous KOH (100 mL). This mixture was refluxed for 16 hours and then allowed to cool to room temperature. The mixture was then acidified to a pH of 2 with 2M aqueous HCl and extracted with EtOAc (2 X).
  • Triethylamine (4.68 mL, 33.59 mmol) and diphenylphosphonic azide (3.73 mL, 16.79 mmol) were added to a solution of 2,6-difluoro-3-(propylsulfonamido)benzoic acid (4.078 g, 14.6 mmol) in THF (60 mL).
  • the reaction mixture was stirred at room temperature for 3 hours and then warmed to 80°C for 2 hours. Water (10 mL) was added, and the mixture was stirred at 80°C for 15 hours.
  • the reaction mixture was diluted with EtOAc (300 mL), and the organic layer was washed with saturated aqueous NaHC0 3 solution and brine.
  • Triethylamine (1.84 mL, 13.2 mmol) and diphenylphosphonic azide (1.43 mL, 6.61 mmol) were added to a solution of 6-chloro-2-fluoro-3-(propylsulfonamido)benzoic acid (1.70 g, 5.75 mmol) in THF (23 mL).
  • the reaction mixture was stirred at room temperature for 1 hour, warmed to 70°C and stirred for 1 hour. Water (6 mL) was added, after which the reaction mixture was stirred again at 70°C for 3 hours.
  • the mixture was cooled to room temperature, ethyl acetate was added, and the layers were separated.
  • Triethylamine (8.02 mL, 57.61 mmol) and diphenylphosphonic azide (6.21 mL, 28.81 mmol) were added to a solution of 2,6-difluoro-3-(propylsulfonamido)benzoic acid (6.643 g, 25.1 mmol) in THF (50 mL).
  • the reaction mixture was stirred at room temperature for 3 hours and then warmed to 80°C for 2 hours. Water (15 mL, 830 mmol) was added, and the mixture stirred at 80°C for 15 hours.
  • reaction mixture was diluted with EtOAc (500 mL), and the organic layer was washed with saturated aqueous NaHC0 3 solution and brine. The solvent was removed under reduced pressure, and the residual purified via silica gel column chromatography eluting with EtOAc/hexane (30/70) to obtain 3.03 g (50%) of the title compound.
  • Triethylamine (9.10 mL, 65.3 mmol) and diphenylphosphonic azide (7.04 mL, 32.7 mmol) was added to a solution of 6-chloro-3-(ethylsulfonamido)-2-fluorobenzoic acid (8.00 g, 28.4 mmol) in THF (115 mL).
  • the reaction mixture was stirred at room temperature for 4 hours, warmed to 70°C and stirred for 2 hours. Water (27 mL) was then added, after which the reaction mixture was stirred again at 70°C for 16 hours.
  • the mixture was cooled to room temperature, ethyl acetate and a saturated solution of NaHC0 3 were added, and the layers were separated.
  • Step A Cyclopropylmethanesulfonyl chloride (4.74 g, 30.6 mmol) was added dropwise to a solution of methyl 3-amino-2-chloro-6-fluorobenzoate (2.97 g, 14.6 mmol) in THF (26 mL) and triethylamine (6.10 mL, 43.8 mmol) at 0°C. The reaction mixture was stirred at 0°C for 90 minutes, after which 8N NaOH (18.2 mL, 140 mmol) was added. The reaction mixture was then warmed up at 40°C and stirred for 16 hours.
  • Step B Triethylamine (2.05 mL, 14.7 mmol) was added to a solution of 2- chloro-3-(cyclopropylmethylsulfonamido)-6-fluorobenzoic acid (4.11 g, 13.4 mmol) in 1,4- dioxane (30 mL), followed by diphenylphosphonic azide (3.12 mL, 14.0 mmol) at room temperature. The reaction was stirred at room temperature for 4 hours, and the resulting mixture added dropwise, via an addition funnel, over 15 minutes in a round-bottom flask containing 1,4-dioxane (16 mL) and water (1.20 mL, 66.8 mmol) at 95°C.
  • Step A 2-Methylpropane-l-sulfonyl chloride (4.80 g, 30.6 mmol) was added dropwise to a solution of methyl 3-amino-2-chloro-6-fluorobenzoate (2.97 g, 14.6 mmol) in THF (20 mL) and triethylamine (6.10 mL, 43.8 mmol) at 0°C. The reaction mixture was stirred at 0°C for 90 minutes, after which 8N aqueous NaOH (18.2 mL, 140 mmol) was added. The reaction mixture was warmed up at 40°C and stirred for 16 hours.
  • Step A 2,6-Difluoro-3-(2-methylpropylsulfonamido)benzoic acid was prepared according to the general procedure for Example N, Step A, substituting methyl 3- amino-2,6-difluorobenzoate for methyl 3-amino-2-chloro-6-fluorobenzoate.
  • Step A 3-(Cyclopropylmethylsulfonamido)-2,6-difluorobenzoic acid was prepared according to the general procedure for Example M, Step A, substituting methyl 3- amino-2,6-difluorobenzoate for methyl 3-amino-2-chloro-6-fluorobenzoate.
  • Step A Fuming nitric acid (4.98 mL, 119 mmol) was added to a solution of methyl 5-chloro-2-fluorobenzoate (16.0 g, 84.8 mmol) in sulfuric acid (100 mL) at 0°C. The reaction mixture was stirred at room temperature for 3 hours, poured into ice/water, and the resulting precipitate was filtered. The obtained solid was purified by flash chromatography to afford methyl 5-chloro-2-fluoro-3-nitrobenzoate (6.78 g, 30%), contaminated with 10% of an undesired regioisomer.
  • Step B A round-bottom flask was charged with 5-chloro-2-fluoro-3- nitrobenzoate (6.78 g, 29.0 mmol), iron (16.2 g, 290 mmol), ammonium chloride (5.43 g, 102 mmol), ethanol (100 mL) and water (30 mL). The reaction mixture was stirred at 85°C for 2 hours and then cooled to room temperature. The mixture was diluted with ethyl acetate and a saturated solution of NaHC0 3 , and the layers were separated. The aqueous layer was extracted twice with ethyl acetate. The organic layers were combined, dried with sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by flash chromatography to afford methyl 3-amino-5-chloro-2-fluorobenzoate (3.7 g, 63%), contaminated with 10% of an undesired regioisomer.
  • Step C Propane- 1-sulfonyl chloride (3.12 mL, 27.8 mmol) was added dropwise to a solution of methyl 3-amino-5-chloro-2-fluorobenzoate (2.71 g, 13.3 mmol) in THF (25 mL) and triethylamine (5.54 mL, 39.8 mmol) at 0°C. The reaction mixture was stirred at 0°C for 90 minutes, after which 8N aqueous NaOH (16.6 mL, 130 mmol) was added. The reaction mixture was then warmed up at 40°C and stirred for 16 hours.
  • Step A Benzyl 6-chloro-2-fluoro-3-(N-(isobutylsulfonyl)-2-methylpropyl- sulfonamido)benzoate was prepared according to the general procedure for Example F, Step B, substituting 2-methylpropane-l-sulfonyl chloride for propane- 1-sulfonyl chloride.
  • Step B 6-Chloro-2-fluoro-3-(2-methylpropylsulfonamido)benzoic acid was prepared according to the general procedure for Example F, Step C, substituting benzyl 6- cUoro-2-fluoro-3-(N-(isobutylsulfonyl)-2-methylpropylsulfonamido)benzoate for benzyl 6- chloro-2-fluoro-3 -(N-(propylsulfonyl)propylsulfonamido) benzoate.
  • the aqueous layer was extracted twice with ethyl acetate. The organic phases were combined, dried with sodium sulfate, filtered and concentrated in vacuo.
  • the crude mixture was dissolved in tetrahydrofuran (15 mL) and methanol (5 mL), and then 1.0M of sodium hydroxide in water (6.3 mL) was added. The reaction mixture was stirred at room temperature for 30 minutes. An aqueous saturated solution of NaHC0 3 and ethyl acetate were added, and the layers were separated. The aqueous layer was extracted twice with ethyl acetate. The organic layers were combined, dried with sodium sulfate, filtered and concentrated in vacuo.
  • Step A 2,6-Dichloro-3-nitrobenzoic acid (2.13 g, 9.03 mmol) was dissolved in 2:1 THF:saturated aqueous N3 ⁇ 4C1 and cooled to 0°C. The mixture was treated with zinc (11.8 g, 181 mmol) and then allowed to warm to ambient temperature and stir for 24 hours. The reaction mixture was filtered through GF/F paper while rinsing with THF. The mixture was acidified to a pH of 1 using 1.0 M HC1 and extracted with 15% 2-propanol/DCM (3 X).
  • Step B 3-Amino-2,6-dichlorobenzoic acid (1.40 g, 6.82 mmol) was dissolved in dry dichloromethane (66.7 mL). Triethylamine (4.09 mL, 29.4 mmol) was added, and the mixture was chilled to 0°C. Propane- 1-sulfonyl chloride (2.48 mL, 22 mmol) was then added using a syringe. When the addition was complete, the mixture was allowed to warm to ambient temperature and stir for 1 hour. The mixture was concentrated in vacuo and diluted with diethyl ether.
  • Step C Triethylamine (2.863 mL, 20.5 mmol) and diphenylphosphonic azide (2.282 mL, 10.2 mmol) was added to a solution of 2,6-dichloro-3- (propylsulfonamido)benzoic acid (2.788 g, 8.93 mmol in THF (40 mL). The reaction mixture was stirred for 6 hours at room temperature. Then water (8 mL, 400 mmol) was added, and the reaction mixture was heated under reflux overnight. Ethyl acetate (300 mL) was added, followed by washing with saturated aqueous NaHC0 3 solution and brine.
  • Step A Methyl 2,6-difluoro-3-(N-(3-fluoropropylsulfonyl)-3-fluoropropyl- sulfonamido)benzoate was prepared according to the general procedure described in Example B, Step C, substituting 3-fluoropropyl sulfonyl chloride for propane- 1-sulfonyl chloride.
  • 1H NMR 400 MHz, (CD 3 ) 2 SO) ⁇ 8.05-7.99 (m, 1H), 7.44 (t, 1H), 4.62 (t, 2H), 4.50 (t, 2H), 3.93 (s, 3H), 3.89-3.74 (m, 4H), 2.26-2.11 (m, 4H).
  • Step B 2,6-Difluoro-3-(3-fluoropropylsulfonamido)benzoic acid was prepared according to the general procedure in Example C, substituting methyl 2,6-difluoro- 3-(N-(3-fluoropropylsulfonyl)-3-fluoropropylsulfonamido)benzoate for methyl 2,6-difluoro- 3-(N-(propylsulfonyl)-propylsulfonamido)benzoate.
  • Step A Benzyl 6-chloro-2-fluoro-3-(3-fiuoro-N-(3-fluoropropylsulfonyl)- propylsulfonamido)benzoate (92%) was prepared according to the general procedure described in Example F, Step B, substituting 3-fluoropropane-l-sulfonyl chloride for propane- 1-sulfonyl chloride.
  • Step B 6-Chloro-2-fluoro-3-(3-fluoropropylsulfonamido)benzoic acid (71%) was prepared according to the general procedure for Example F, Step C, substituting benzyl 6-chloro-2-fluoro-3 -(3 -fluoro-N-(3 -fluoropropylsulfonyl)propylsulfonamido)benzoate for benzyl 6-chloro-2-fluoro-3-(N-(propylsulfonyl)propylsulfonamido)benzoate.
  • Step A Methyl 2-chloro-6-fluoro-3-(3-fluoro-N-(3-fluoropropylsulfonyl)- propylsulfonamido)benzoate was prepared according to the general procedure in Example B, Step C, substituting 3-fluoropropyl sulfonyl chloride for propane- 1-sulfonyl chloride.
  • Step B 2-Chloro-6-fiuoro-3-(3-fluoropropylsulfonamido)benzoic acid (93% over 2 steps) was prepared according to the general procedure for Example F, Step C, substituting methyl 2-chloro-6-fluoro-3-(3-fiuoro-N-(3- fluoropropylsulfonyl)propylsulfonamido)benzoate for benzyl 6-chloro-2-fluoro-3-(N- (propylsulfonyl)propylsulfonamido)benzoate.
  • Triethylamine (0.682 mL, 4.89 mmol) and diphenylphosphoryl azide (0.547 mL, 2.45 mmol) were added to a solution of 2,6-difluoro-3-(3- fluoropropylsulfonamido)benzoic acid (0.485 g, 1.63 mmol) in DMF (10 mL).
  • the reaction mixture was stirred at room temperature for 1 hour. Water (1.18 mL, 65.2 mmol) was added, and the mixture was stirred at 80°C for 15 hours.
  • the reaction mixture was diluted with ethyl acetate (100 mL), and the organic layer was washed with saturated aqueous NaHC0 3 , dried over sodium sulfate and concentrated.
  • the crude product was purified via silica gel chromatography, eluting with hexanes/ethyl acetate (4:1) to give the title compound (0.34 g, 77%).
  • N-(3-Amino-4-chloro-2-fluorophenyl)-3 -fluoropropane- 1 -sulfonamide (8.8 g, 52%) was prepared according to the general procedure in Example Y, substituting 6-chloro- 2-fluoro-3-(3-fluoropropylsulfonamido)benzoic acid for 2,6-difluoro-3-(3-fluoropropyl- sulfonamido)benzoic acid.
  • Step A TEA (4.58 mL, 32.9 mmol) was added to 4-methyl-3-nitroaniline (1.0 g, 6.57 mmol) in DCM (30 mL) at 0°C, followed by propane- 1-sulfonyl chloride (1.84 mL, 16.4 mmol). The solution was allowed to warm to ambient temperature and stirred for 2 hours. The solution was diluted with aqueous bicarbonate (100 mL) and extracted with EtOAc (3 X 40 mL).
  • Step B 2M NaOH (16 mL, 33 mmol) was added to N-(4-methyl-3- nitrophenyl)-N-(propylsulfonyl)propane-l -sulfonamide (2.4 g, 6.6 mmol) in 4:1 THF:MeOH (75 mL). The solution was warmed to 50°C for 3 hours. The cooled solution was concentrated under reduced pressure, and the residue was diluted with aqueous ammonium chloride (100 mL) and extracted with EtOAc (3 X 40 mL).
  • Step C 10% Pd/C (1.32 g, 1.24 mmol) was added to N-(4-methyl-3- nitrophenyl)propane-l -sulfonamide (1.6 g, 6.19 mmol) in EtOH (30 mL). The suspension was stirred under a balloon of hydrogen at ambient temperature for 16 hours. The suspension was filtered and concentrated to afford (3 -amino-4-methylphenyl)propane-l -sulfonamide (1.38 g, 97.6%).
  • Step A Triethylamine (4.19 mL, 29.0 mmol) and propane- 1-sulfonyl chloride (1.63 mL, 14.5 mmol) was added to 4-chloro-3-nitroaniline (29.0 mL, 5.79 mmol) in DCM (30 mL) at 0°C. The solution was allowed to warm to ambient temperature and stirred for 1 hour before dilution with aqueous bicarbonate (50 mL) and extraction with EtOAc (3 X 40 mL).
  • Step B 2M NaOH (16 mL, 33 mmol) was added to N-(N-(4-chloro-3- nitrophenyl)-N-(propylsulfonyl)propane-l -sulfonamide (2.4 g, 6.6 mmol) in 4:1 THF:MeOH (75 mL). The solution was warmed to 50°C for 3 hours.
  • Step C 2M HC1 (2 mL) and Fe (0) (0.301 g, 5.38 mmol) were added to N-(4- chloro-3-nitrophenyl)propane-l -sulfonamide (0.50 g, 1.79 mmol) in MeOH (10 mL). The suspension was warmed to reflux for 4 hours, then cooled and filtered through GF/F paper. The filtrate was concentrated under reduced pressure to afford N-(3-amino-4- chlorophenyl)propane-l -sulfonamide (0.40 g, 89%).
  • Step A 2-Chloro-3-nitroaniline (Sienkowska, et. al, Tetrahedron 56 (2000) 165) (0.36 g, 2.086 mmol) was dissolved in DCM (20 mL) and cooled to 0°C. Triethylamine (0.8723 mL, 6.258 mmol) was added followed by propane- 1-sulfonyl chloride (0.5847 mL, 5.215 mmol) and the reaction was stirred at room temperature overnight. The reaction was quenched with 0.1N HC1 (10 mL), and the layers were separated.
  • Step B N-(2-Chloro-3-nitrophenyl)-N-(propylsulfonyl)propane-l- sulfonamide (0.8028 g, 2.086 mmol) was dissolved in 3:1 THF/MeOH (4.0 mL). NaOH (2.0 M, 2.086 mL, 4.172 mmol) was added, and the reaction was stirred for five minutes at room temperature. The reaction was quenched with 0.1N HC1 (5 mL), and the volatiles were removed by rotary evaporation.
  • Step C N-(2-Chloro-3-nitrophenyl)propane-l -sulfonamide (0.580 g, 2.08 mmol) was dissolved in 4:1 EtOH/water (10 mL). Fe (0) (1.16 g, 20.8 mmol) was added, followed by a catalytic amount of NH4CI (5 mg), and the reaction was heated to 80°C for 3 hours.
  • N-(3-Amino-2-chloro-4-fluorophenyl) propane- 1 -sulfonamide (668 mg, 2.5 mmol) was dissolved in methanol (100 mL), and the solution was run through an H-Cube hydrogenator at 50°C and 10 bar H 2 pressure at a 1 mL/min flow rate. After removal of the solvent, 481 mg (83%) of N-(3-amino-4-fluorophenyl) propane- 1 -sulfonamide were obtained.
  • N-(3-Amino-4-chloro-2-fluorophenyl) propane- 1 -sulfonamide (477 mg, 1.8 mmol) was dissolved in methanol (100 mL), and the solution was run through an H-Cube hydrogenator at room temperature and ambient pressure at a 1 mL/min flow rate. After removal of the solvent, N-(3-amino-2-fluorophenyl) propane- 1 -sulfonamide was obtained (251 mg, 60%).
  • Step A Concentrated ammonium hydroxide (5.16 mL, 76.4 mmol) was added to 2,3,6-trifluorobenzonitrile (2.0 g, 12.7 mmol) in isopropanol (5 mL). The colorless solution was heated at 80°C in a sealed vial overnight. The reaction mixture was concentrated, and the residue partitioned between EtOAc and water. The EtOAc was washed with brine, dried over MgS0 4 , filtered, and evaporated to yield 2-amino-3,6- difluorobenzonitrile (1.93 g, 12.5 mmol, 98.4% yield) as a solid.
  • Step B 60% sodium hydride (0.558 g, 14.0 mmol) was added slowly in portions to propane- 1 -sulfonamide (1.68 g, 13.6 mmol) in DMSO (10 mL) with water bath cooling. After gas evolution ceased, the mixture was diluted with DMSO (5 mL) to aid in dissolution, and was stirred an additional 30 minutes at ambient temperature. A solution of 2-amino-3,6-difluorobenzonitrile (1.00 g, 6.49 mmol) in DMSO (20 mL) was added to the reaction, and the resulting mixture was heated at 100°C for 20 hours, then 120°C for 16 hours.
  • Step A 14.8M ammonium hydroxide (4.67 mL, 69.1 mmol) was added to 3- chloro-2,6-difluorobenzonitrile (2.00 g, 11.5 mmol) in isopropanol (5 mL). The colorless solution was heated at 80°C in a sealed vial. After 2 hours, the reaction mixture was concentrated, and the residue partitioned between EtOAc and water. The EtOAc was washed with brine, dried over MgS0 4 , filtered, and evaporated to yield 2-amino-3-chlorO ' 6- fluorobenzonitrile (1.63 g, 9.56 mmol, 82.9% yield) as a solid.
  • Step B 60% sodium hydride (0.252 g, 6.30 mmol) was added slowly in portions to propane- 1 -sulfonamide (0.740 g, 6.01 mmol) in NMP (10 mL) with water bath cooling. Gas evolution continued as the mixture was stirred an additional 30 minutes at ambient temperature and then heated 1 hour at 40°C. The mixture was cooled to room temperature, and 2-amino-3-chloro-6-fluorobenzonitrile (0.50 g, 2.93 mmol) was added. The resulting mixture was heated in a sealed vial at 120°C overnight. The reaction mixture was diluted with 0.5M NaOH and washed twice with EtOAc.
  • Step A A 250 mL round bottom flask was charged with methyl 2,6-difluoro- 3-nitrobenzoate (10.03 g, 46.18 mmol) and methanol (60 mL, 1000 mmol) and was then cooled over a brine / ice bath at -4°C for 20 minutes. A 5M solution of sodium methoxide in methanol (11.98 mL, 59.88 mmol) was added dropwise to this solution over 20 minutes, while maintaining the reaction temperature at -4°C over the course of the addition. The reaction mixture was allowed to stir overnight, gradually rising to room temperature.
  • Step B A 250 mL round bottom flask was charged with methyl 6-fluoro-2- methoxy-3-nitrobenzoate (3.37 g, 14.71 mmol) dissolved in methanol (125 mL, 3080 mmol). Nitrogen was passed through the reaction mixture, and 10% palladium on activated carbon (1.3 g, 1.2 mmol) was added. The flask was capped and evacuated and then allowed to stir for 60 hours under an atmosphere of hydrogen at ambient temperature and pressure. The mixture was then filtered through Celite® to remove the solid catalyst and washed with methanol (500 mL).
  • Step C A 250 mL round bottom flask was charged with a solution of methyl 3-amino-6-fluoro-2-methoxybenzoate (3.656 g, 18.36 mmol) in methylene chloride (100 mL). A solution of 4-dimethylaminopyridine (113 mg, 0.925 mmol), pyridine (7.45 mL, 92.1 mmol) and propane- 1-sulfonyl chloride (8.25 mL, 73.6 mmol) in methylene chloride (10 mL) was added to the reaction mixture over the course of five minutes. The reaction mixture was stirred at room temperature for 14 hours.
  • Step D A 100 mL round bottom flask was charged with methyl 6-fluoro-2- methoxy-3-(propylsulfonamido)benzoate (4.91 g, 16.1 mmol) dissolved in N,N- dimethylformamide (16 mL, 210 mmol) and was cooled over an ice/ brine bath. Sodium hydride (0.676 g, 16.9 mmol) was added in four portions. After the vigorous bubbling subsided, the reaction mixture was stirred for 1 hour at room temperature. The reaction mixture was cooled over an ice/brine bath, and p-methoxybenzyl chloride (2.646 g, 16.90 mmol) was added.
  • the reaction was allowed to warm to room temperature over the next three hours and then quenched by adding a semi-saturated aqueous ammonium chloride solution (200 mL) at 0°C. After stirring at room temperature overnight, the aqueous layer was discarded, and the remaining oil washed with heptanes to remove the mineral oil. The residual oil was dissolved in ethyl acetate, dried over magnesium sulfate, filtered and concentrated to remove the ethyl acetate.
  • the crude product was purified by flash chromatography (120 g column), using a gradient of 0-100% ethyl acetate: heptanes to give methyl 6-fluoro-2-methoxy-3-(N-(4-methoxybenzyl)propylsulfonamido)benzoate (3.71 g, 57%) as an oil.
  • Step E A 250 mL round bottom flask was charged with methyl 6-fluoro-2- methoxy-3-(N-(4-methoxybenzyl)propylsulfonamido)benzoate (4.42 g, 10.4 mmol) dissolved in tetrahydrofuran (70 mL, 900 mmol). 1M of sodium hydroxide in water (67.8 mL, 67.8 mmol) was added, and the mixture was stirred at 60°C for 48 hours. After cooling, the THF was removed under reduced pressure. The basic aqueous solution was diluted with water to a volume of 100 mL and then extracted once with ethyl acetate (200 mL).
  • aqueous layer was acidified with concentrated hydrochloric acid (5 mL) to a pH of 2 and extracted three times with ethyl acetate (100 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to afford 6-fluoro-2-methoxy-3-(N-(4- methoxybenzyl)propylsulfonamido)benzoic acid as a solid (4.253 g, 99%). !
  • Step F Under a nitrogen atmosphere, a dry 100 mL round bottom, stir bar and reflux condenser were charged with 6-fluoro-2-methoxy-3-(N-(4-methoxybenzyl)- propylsulfonamido)benzoic acid (379 mg, 0.921 mmol) dissolved in 1,4-dioxane (10 mL, 83 mmol). Triethylamine (295.3 ⁇ , 2.12 mmol) and then diphenylphosphonic azide (228.3 ⁇ , 1.06 mmol) were added. The reaction mixture was stirred at room temperature for 3 hours and then heated to reflux for 1 hour.
  • N-(3-Amino-2-chloro-4-fluorophenyl)propane-l -sulfonamide (75 g, 280 mmol) was dissolved in N,N-dimethylformamide (200 mL, 2000 mmol).
  • a 60% sodium hydride suspension in mineral oil (6:4, sodium hydride:mineral oil, 11.85 g, 296 mmol) was added in multiple portions over a period of fifteen minutes. The reaction mixture was stirred at room temperature for 90 minutes and was then warmed to 40°C for two hours.
  • Step A 2-Chloro-l,3-dinitrobenzene (0.500 g, 2.47 mmol), Cul (0.0940 g, 0.494 mmol), P(t-Bu) 3 (1.51 mL, 0.494 mmol) and ethynyltriisopropylsilane (0.658 mL, 2.96 mmol) were dissolved in 5:1 acetonitrile/TEA (10 mL). Nitrogen gas was bubbled through for 5 minutes, and PdCl 2 (MeCN) 2 (0.0640 g, 0.247 mmol) was added. Nitrogen gas was bubbled though for another 10 minutes.
  • Step B ((2,6-Dinitrophenyl)ethynyl)triisopropylsilane (0.310 g, 0.890 mmol) was dissolved in 1:1 DCM/DMF (30 mL). SnCl 2 dihydrate (10.0 g, 44.5 mmol) was added and stirred for one hour at room temperature. The reaction was poured into saturated aqueous NaHC0 3 (200 mL) giving a precipitate, which was stirred at room temperature for 30 minutes and filtered through Celite®. The layers were separated, the aqueous was extracted with DCM, and the combined organics washed with water (2 X) and brine, dried over Na 2 S0 4 and concentrated.
  • Step C 2-((Triisopropylsilyl)ethynyl)benzene-l,3-diamine (0.072 g, 0.249 mmol) was dissolved in THF (5 mL), and n-chlorosuccinimide (0.036 g, 0.286 mmol) was added and stirred at room temperature for one hour. The crude reaction was diluted with EtOAc, washed with water (3 X) and brine, and then dried over Na 2 S0 4 and concentrated.
  • Step D 4-Chloro-2-((triisopropylsilyl)ethynyl)benzene- 1,3 -diamine (0.0554 g, 0.172 mmol) was dissolved in 10:1 dichloroethane/pyridine (1 mL) and cooled to 0°C. Propane- 1-sulfonyl chloride (0.0193 mL, 0.172 mmol) was added, and the reaction was stirred at 50°C overnight. The reaction was concentrated, dissolved in EtOAc and washed with 0.1N HC1, water, and brine, dried over Na 2 S0 4 and concentrated.
  • reaction mixture was cooled to room temperature and diluted with ethyl acetate (100 mL) and water (30 mL). The pH of the aqueous layer was adjusted to about 6 with AcOH. The reaction mixture was filtered, and the filter cake was washed with ethyl acetate (50 mL). The filtrate was transferred to a separatory funnel, and the layers were separated. The aqueous layer was extracted with ethyl acetate (1 X 50 mL). The combined organics were dried, filtered and concentrated.
  • Step A 6-Bromo-3-methylquinazolin-4(3H)-one (0.200 g, 0.837 mmol), N- (3-amino-2,4-difluorophenyl)-N-(4-methoxybenzyl)propane-l-sulfonamide (0.310 g, 0.837 mmol), NaOtBu (0.241 g, 2.51 mmol), and Binap-rac (0.0521 g, 0.0837 mmol) were taken up in toluene as a slurry in a sealed vial.
  • Nitrogen gas was bubbled through for 5 minutes, and Pd 2 dba 3 CHCI3 (0.0433 g, 0.0418 mmol) was added. Nitrogen gas was bubbled through for another 10 minutes, and the reaction was heated to 100°C overnight. The reaction was concentrated and partitioned between EtOAc and 0.1N HC1. The organic layer was washed with water and brine, dried over Na 2 S0 , filtered and concentrated.
  • Step B N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.010 g, 0.019 mmol) was dissolved in neat TFA with one drop of water and stirred at room temperature for 30 minutes. The reaction was concentrated, and the residual solid was taken up in EtOAc and washed with saturated NaHC0 3 and brine, dried over Na 2 S0 4 , and concentrated.
  • Step A N-(2,4-Difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.015 g, 0.0284 mmol) was dissolved in DMF (1 mL) and cooled to 0°C. NaH (0.00170 g, 0.0426 mmol) was added and stirred for 10 minutes. Mel (0.00195 mL, 0.0312 mmol; d 2.275) was added and stirred for 10 minutes at room temperature. The reaction was partitioned between EtOAc and water.
  • Step B N-(2,4-Difluoro-3-(methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6- yl)amino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.0154 g, 0.0284 mmol) was dissolved in neat TFA with a few drops of water and stirred at room temperature for 30 minutes. The reaction was concentrated, and the residual solid was taken up in EtOAc and washed with saturated NaHC0 3 and brine, and dried over Na 2 S0 4 and concentrated.
  • N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.050 g, 28% yield) was prepared according to the general procedure of Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1-sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide.
  • N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-l-cyclopropylmethanesulfonamide (0.082 g, 48% yield) was prepared according to the general procedure of Example 1, substituting N-(2-chloro-4-fluoro-3-(3- methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)- 1 -cyclopropylmethanesulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide.
  • Step A N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.45 g, 49% yield) was prepared according to the general procedure of Example 1, substituting N-(3-amino-2-chloro- 4-fluorophenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide for N-(3 -amino-4-chloro-2- fluorophenyl)propane-l -sulfonamide.
  • LC/MS m/z 545.2, 547.2 [M+l].
  • Step B N-(2-Chloro-4-fluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide was prepared according to the general procedure of Example 3, Step A, substituting N-(2-chloro- 4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide.
  • LC/MS m/z 559.
  • Step C N-(2-Chloro-4-fluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)propane-l -sulfonamide (0.025 g, 74% yield over 2 steps) was prepared according to the general procedure of Example 3, Step B substituting N- (2-chloro-4-fluoro-3 -(methyl(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6-yl)amino)phenyl)-N- (4-methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3 -(methyl(3 -methyl-4-oxo- 3,4-dihydroquinazolin-6-yl)amino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide.
  • N-(2-Chloro-4-fiuoro-3 -(quinoxalin-6-ylamino)phenyl)propane- 1 -sulfonamide (53%) was prepared according to Example 1, using 6-bromoquinoxaline and N-(3-amino-2- chloro-4-fluorophenyl)propane-l -sulfonamide.
  • Step A 6-Bromo-4-chloroquinazoline (1.00 g, 4.11 mmol) was treated with 0.5M NaOMe (82.1 mL, 41.1 mmol) at room temperature for 2 hours. The reaction mixture was concentrated, and the crude solids were washed with water and dried in vacuo to give 6- bromo-4-methoxyquinazoline (0.83 g, 84% yield) as a solid.
  • LC/MS m/z 239.0, 241.0 [M+l].
  • Step B N-(2-Chloro-4-fluoro-3-(4-methoxyquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.100 g, 55% yield) was prepared according to the general procedure of Example 1, substituting 6-bromo-4-methoxyquinazoline for 6-bromo-3- methylquinazolin-4(3H)-one.
  • Step A 6-Bromo-4-ethoxyquinazoline (0.61 g, 59% yield) was prepared according to the general procedure in Example 8, Step A, substituting NaOEt for NaOMe.
  • LC/MS m/z 253.1, 255.1 [M+l].
  • Step B N-(2-Chloro-3-(4-ethoxyquinazolin-6-ylamino)-4- fluorophenyl)propane-l -sulfonamide (0.100 g, 55% yield) was prepared according to the general procedure in Example 1, substituting 6-bromo-4-ethoxyquinazoline for 6-bromo-3- methylquinazolin-4(3H)-one.
  • Step A HATU (0.329 g, 0.867 mmol), DBU (0.152 g, 1.000 mmol) and 1- methyl-lH-pyrazol-4-amine hydrochloride (0.0745 mL, 0.800 mmol) were added to 6- bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water.
  • Step B N-(2-Chloro-4-fluoro-3-(3-(l-methyl-lH-pyrazol-4-yl)-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)propane-l -sulfonamide (0.005 g, 17% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro- 4-fluorophenyl)propane- 1 -sulfonamide for N-(3 -amino-4-chloro-2-fluorophenyl)propane- 1 - sulfonamide and 6-bromo-3-methylquinazolin-4(3H)-one for 6 -bromo-3-(l -methyl- 1H- pyrazol-4-yl)quinazolin-4(3H)-one.
  • N-(2,4-Difluoro-3-(3-(l-methyl-lH-pyrazol-4-yl)-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)propane-l -sulfonamide (0.006 g, 21% yield) was prepared according to the Example 10, Step B, substituting N-(3-amino-4-chloro-2- fluorophenyl)propane-l -sulfonamide for N-(3-amino-2,4-difIuorophenyl)propane-l - sulfonamide.
  • Step A HATU (0.329 g, 0.867 mmol), DBU (0.152 g, 1.000 mmol) and aniline (0.0745 mL, 0.800 mmol) were added to 6-bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water.
  • Step B N-(2-Chloro-4-fluoro-3-(4-oxo-3-phenyl-3,4-dihydroquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.005 g, 7.4% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo- 3-methylquinazolin-4(3H)-one for 6-bromo-3-phenylquinazolin-4(3H)-one.
  • Step A HATU (0.329 g, 0.867 mmol), DBU (0.152 g, 1.000 mmol) and phenylmethanamine (0.0902 mL, 0.800 mmol) were added to 6-bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water.
  • Step B N-(3-(3-Benzyl-4-oxo-3,4-dihydroquinazolin-6-ylamino)-2-chloro-4- fluorophenyl)propane-l -sulfonamide (0.020 g, 36% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo- 3-methylquinazolin-4(3H)-one for 3-benzyl-6-bromoquinazolin-4(3H)-one.
  • Step A PyBOP (0.347 g, 0.667 mmol), DBU (0.152 g, 1.000 mmol), and methylamine (0.400 mL, 0.800 mmol) were added to 6-bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water. The concentrated organics were purified via trituration with DCM to provide 6-bromo-N-methylquinazolin-4-amine (0.055 g, 0.231 mmol, 35.7% yield).
  • Step B N-(2-Chloro-4-fluoro-3-(4-(methylamino)quinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.003 g, 3.1% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo- 3-methylquinazolin-4(3H)-one for 6-bromo-N-methylquinazolin-4-amine.
  • Step A PyBOP (0.347 g, 0.667 mmol), DBU (0.152 g, 1.000 mmol), and dimethylamine (0.400 mL, 0.800 mmol) were added to 6-bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water. The concentrated organics were purified via silica gel chromatography eluting with EtOAc to provide 6-bromo-N,N-dimethylquinazolin-4-amine (0.042 g, 0.167 mmol, 25% yield).
  • Step B N-(2-Chloro-3-(4-(dimethylamino)quinazolin-6-ylamino)-4- fluorophenyl)propane-l -sulfonamide (0.005 g, 6.9% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo- 3-methylquinazolin-4(3H)-one for 6-bromo-N,N-dimethylquinazolin-4-amine.
  • N-(2-Chloro-4-fluoro-3 -(4-(4-methylpiperazin- 1 -yl)quinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.002 g, 2.5% yield) was prepared according to the general procedure in Example 14, substituting methylamine for methyl piperazine and substituting 6-bromo-N-methylquinazolin-4-amine for 6-bromo-4-(4-methylpiperazin-l- yl)quinazoline.
  • tert-Butyl 4-(6-(2-cMoro-6-fluoro-3- ( ropylsulfonarnido)phenylamino)quinazolin-4-yl)piperazine- 1 -carboxylate sulfonamide (0.020 g, 27% yield) was prepared according to the general procedure in Example 14, substituting methylamine for tert-butyl piperazine-l-carboxylate and substituting 6-bromo-N- methylquinazolin-4-amine for tert-butyl 4-(6-bromoquinazolin-4-yl)piperazine-l -carboxylate.
  • N-(2-Chloro-4-fluoro-3-(quinazolin-6-ylamino)phenyl)propane-l -sulfonamide (0.050 g, 26.5% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane-l -sulfonamide for N-(3-amino-4- chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline.
  • Step A N-(2,4-Difluoro-3-(quinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (0.050 g, 28% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3 -amino-2,4-difluorophenyl)-N-(4-methoxybenzyl)propane- 1 - sulfonamide and 6-bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline.
  • LC/MS m/z 499.2 [M+l].
  • Step B N-(2,4-Difluoro-3-(quinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (0.010 g, 0.020 mmol) was taken up in TFA (1 mL) and warmed to 50°C for 30 minutes. Concentration under reduced pressure and trituration of the residue with DCM provided N-(2,4-difluoro-3-(quinazolin-6-ylamino)phenyl)propane-l- sulfonamide (0.004 g, 0.011 mmol, 53% yield).
  • Step A N-(2,4-Difluoro-3-(quinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (0.050 g, 28% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3 -amino-2,4-difluorophenyl)-N-(4-methoxybenzyl)propane- 1 - sulfonamide and 6-bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline.
  • LC MS m/z 499.2 [M+l].
  • Step B NaH (0.00882 g, 0.221 mmol) was added to N-(2,4-difluoro-3- (quinazolin-6-ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.044 g, 0.0883 mmol) in DMF (1 mL) at ambient temperature. After 10 minutes, Mel (0.0275 mL, 0.441 mmol; d 2.275) was added and stirred for 1 hour. The suspension was diluted with EtOAc and washed with brine-water.
  • Step C N-(2,4-Difluoro-3-(methyl(quinazolin-6-yl)amino)phenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (0.010 g, 0.020 mmol) was taken up in TFA (1 mL) and warmed to 50°C for 30 minutes. Concentration under reduced pressure and trituration of the residue with DCM provided N-(2,4-difluoro-3-(methyl(quinazolin-6- yl)amino)phenyl)propane-l -sulfonamide (0.004 g, 0.010 mmol, 52% yield).
  • N-(2-Chloro-4-fluoro-3 -(methyl(quinazolin-6-yl)amino)phenyl)propane- 1 - sulfonamide (0.004 g, 52% yield) was prepared according to the procedure in Example 21, substituting N-(3-amino-2,4-difluorophenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide for N-(3-amino-2-chloro-4-fluorophenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide.
  • Step A 2-Fluoro-6-(quinazolin-6-ylamino)benzonitrile (0.100 g, 26.4% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2- chloro-4-fluorophenyl)propane-l -sulfonamide for 2-amino-6-fluorobenzonitrile and 6- bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline.
  • LC/MS m/z 265.1 [M+l].
  • Step B NaH (0.020 g, 0.50 mmol) was added to propane- 1 -sulfonamide (0.062 g, 0.50 mmol) in DMA (0.7 mL). After 20 minutes, a solution of 2-fluoro-6- (quinazolin-6-ylamino)benzonitrile (0.066 g, 0.25 mmol) in DMA (1 mL) was slowly added. The mixture was sealed and placed in a microwave for 6 hours at 210°C. The resulting solution was diluted with EtOAc and washed with NRjCl-brine-water. The concentrated residue was then partitioned between EtAOc and aqueous sodium bicarbonate.
  • Step A 2-Fluoro-6-(quinazolin-6-ylamino)benzonitrile (0.100 g, 26.4% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2- chloro-4-fluorophenyl)propane-l -sulfonamide for 2-amino-6-fluorobenzonitrile and 6- bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline.
  • LC/MS m/z 265.1 [M+l].
  • Step B NaH (0.00568 g, 0.142 mmol) was added to 2-fIuoro-6-(quinazolin-6- ylamino)benzonitrile (0.592 mL, 0.118 mmol) in DMF at 0°C. After 10 minutes at ambient temperature, Mel (0.0148 mL, 0.237 mmol) was added and stirred for 20 minutes. The suspension was diluted with EtOAc and washed with brine-water. The organics were concentrated under reduced pressure to afford 2-fluoro-6-(methyl(quinazolin-6- yl)amino)benzonitrile (0.035 g, 0.126 mmol, 106%).
  • Step C NaH (0.010 g, 0.25 mmol) was added to propane- 1 -sulfonamide (0.031 g, 0.25 mmol) in DMA (0.5 mL). After 20 minutes, a solution of 2-fluoro-6- (methyl(quinazolin-6-yl)amino)benzonitrile (0.035 g, 0.13 mmol) in DMA (1 mL) was slowly added. The mixture was sealed and placed in a microwave for 6 hours at 210°C. The resulting solution was diluted with EtOAc and washed with NFL t Cl-brine-water. The concentrated residue was then partitioned between EtAOc and aqueous sodium bicarbonate.
  • Step A 60% Sodium hydride (0.0522 g, 1.31 mmol) was slowly added to 6- hydroxy-3-methylquinazolin-4(3H)-one (0.2 g, 1.14 mmol; Maillard, Jacques, et al. "(3H)- Quinazolin-4-one derivatives with antiinflammatory activity. II. Derivatives substituted in the aromatic nucleus, and related compounds.” Chim. Ther. 2(4) (1967): 231-9) in DMF (4 mL) cooled in ice.
  • Step B 60% Sodium hydride (0.0287 g, 0.718 mmol) was slowly added to propane- 1 -sulfonamide (0.0826 g, 0.670 mmol) in NMP (5 mL) cooled in ice. After addition, the mixture was stirred at 50°C for 30 minutes. 3,6-Difluoro-2-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yloxy)benzonitrile (0.10 g, 0.319 mmol; Step A) was added to the mixture. The reaction was stirred at 120°C overnight. The cooled reaction mixture was partitioned between 0.1M NaOH and EtOAc.
  • Step A 60% Sodium hydride (0.0522 g, 1.31 mmol) was slowly added to 6- hydroxy-3-methylquinazolin-4(3H)-one (0.200 g, 1.14 mmol) in DMF (3 mL) cooled in ice. When gas evolution ceased, the mixture was stirred 15 minutes at room temperature, then 2,6-difluorobenzonitrile (0.158 g, 1.14 mmol) was added, and the mixture was stirred at 80°C. After 1 hour, the reaction mixture was allowed to cool and was diluted with water. The resulting solid was collected by vacuum filtration, washed with water and some hexane.
  • Step B 60% Sodium hydride (0.0291 g, 0.728 mmol) was slowly added to propane- 1 -sulfonamide (0.0876 g, 0.711 mmol) in NMP (5 mL) cooled in ice. After addition, the mixture was stirred at 50°C for 30 minutes. 2-Fluoro-6-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yloxy)benzonitrile (0.10 g, 0.339 mmol) was added to the mixture. The reaction mixture was stirred at 140°C overnight, then at 180°C for 8 hours. The cooled reaction mixture was partitioned between 0.5M NaOH and EtOAc.
  • Step A 60% Sodium hydride (0.0522 g, 1.31 mmol) was slowly added to 6- hydroxy-3-methylquinazolin-4(3H)-one (0.200 g, 1.14 mmol) in DMF (4 mL). When gas evolution ceased, a solution of 3-chloro-2,6-difluorobenzonitrile (0.197 g, 1.14 mmol) in DMF (1 mL) was added, and the mixture was stirred at ambient temperature.
  • Step B 60% Sodium hydride (0.0255 g, 0.637 mmol) was added to propane-
  • N-(4-Chloro-2-cyano-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.024 g, 22% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-4-chloro-2-cyanophenyl)propane- 1-sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide.
  • N-(2-Cyano-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.0295 g, 18.7% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-cyanophenyl)propane-l- sulfonamide for N-(3 -amino-4-chloro-2-fluorophenyl)propane- 1 -sulfonamide .
  • Step B N-(2,4-Difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-3 -fluoropropane- 1 -sulfonamide (74%) was prepared according to the general procedure of Example 2, Step B, using N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)-3 -fluoro-N-(4-methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6-ylamino)phenyl)-3-fluoro-N- (4-methoxybenzyl)propane-l-sulfonamide.
  • Step A N-(2,4-Difluoro-3-(methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6- yl)amino)phenyl)-3 -fluoro-N-(4-methoxybenzyl)propane- 1 -sulfonamide ( 100%) was prepared according to the general procedure of Example 3, Step A, substituting N-(2,4- difluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)-3 -fluoro-N-(4- methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3 -(3-methyl-4-oxo-3 ,4- dihydroquinazolin-6-ylamino)phenyl)-N-(4-methoxybenzyl)propane-l-s
  • Step B N-(2,4-Difluoro-3-(methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6- yl)amino)phenyl)-3 -fluoropropane- 1 -sulfonamide (33%) was prepared according to the general procedure of Example 3, Step B, substituting N-(2,4-difluoro-3-(methyl(3-methyl-4- oxo-3,4-dihydroquinazolin-6-yl)amino)phenyl)-3-fluoro-N-(4-methoxybenzyl)propane-l- sulfonamide for N-(2,4-difluoro-3 -(methyl(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- yl)amino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide for N-(
  • Step A N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-3 -fluoropropane- 1 -sulfonamide (44%) was prepared according to the general procedure of Example 4, Step A, substituting N-(3-amino-2-chloro-4-fluorophenyl)- 3 -fluoro-N-(4-methoxybenzyl)propane- 1 -sulfonamide for N-(3 -amino-2,4-difluorophenyl)- N-(4-methoxybenzyl)propane- 1 -sulfonamide.
  • Step B N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-3 -fluoropropane- 1 -sulfonamide (37%) was prepared according to the general procedure of Example 4, Step B, substituting N-(2-chloro-4-fluoro-3-(3-methyl-4- oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)-3 -fluoro-N-(4-methoxybenzyl)propane- 1 - sulfonamide for N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-3-fluoro-N-(4-methoxybenzyl)propane-l -sulfonamide.
  • Step A N-(4-Chloro-2-fluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide ( 100%) was prepared according to the general procedure of Example 3, Step A, substituting N-(4- chloro-2-fluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3-(3 -methyl-4-oxo-3 ,4- dihydroquinazolin-6-ylammo)phenyl)-3-fluoro-N-(4-methoxybenzyl)propane-l-sulfonamide.
  • Step B N-(4-Chloro-2-fluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)propane-l -sulfonamide (71%) was prepared according to the general procedure of Example 3, Step B, substituting N-(4-chloro-2-fluoro-3- (methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)phenyl)-N-(4- methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide.
  • Step A N-(4-Chloro-2-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6 ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.0235 g, 0.0431 mmol) was dissolved in THF. N-Chlorosuccinimide (0.00576 g, 0.0431 mmol) was added and stirred at room temperature for 3 days. The reaction was concentrated and partitioned between EtOAc and water. The organic layer was washed with water and brine, dried over Na 2 S0 4 , filtered and concentrated.
  • Step B N-Benzyl-N-(4-chloro-3-(5-chloro-3-methyl-4-oxo-3,4- dihydroqumazolin-6-ylamino)-2-fluorophenyl)propane-l -sulfonamide was dissolved in neat TFA with one drop of water and stirred at room temperature for 30 minutes. The reaction was concentrated, and the residual solid was taken up in EtOAc and washed with saturated NaHC0 3 and brine, dried over Na 2 S0 4 , and concentrated.
  • Biotage chromatography (hexanes/EtO Ac) provided N-(4-chloro-3 -(5 -chloro-3 -methyl-4-oxo-3 ,4-dihydroquinazolin- 6-ylamino)-2-fiuorophenyl)propane-l -sulfonamide (0.0056g, 71%) as a solid.
  • N-(2-Chloro-4-fluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- ylamino)phenyl)-3-fluoropropane-l -sulfonamide (0.0207 g, 0.0467 mmol) was dissolved in THF.
  • N-Chlorosuccinimide (0.00624 g, 0.0467 mmol) was added, and the reaction was stirred at room temperature overnight. The reaction was concentrated and partitioned between EtOAc and water. The organic layer was washed with water and brine, dried over Na 2 S0 4 , filtered and concentrated.
  • Step A 7-Bromoquinoxalin-2(lH)-one (145.0 mg, 0.644 mmol) was dissolved in 0.1M MeCN (6.4 mL) and treated sequentially with 2.0M methylamine in tetrahydrofuran (354.4 ⁇ , 0.709 mmol), l,8-diazabicyclo[5.4.0]undec-7-ene (115.6 ⁇ , 0.773 mmol), and benzotriazol-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate (342.0 mg, 0.773 mmol). The reaction mixture was stirred at ambient temperature for 16 hours and then concentrated in vacuo.
  • Step B N-(2-Chloro-4-fluoro-3-(3-(methylamino)quinoxalin-6- ylamino)phenyl)propane-l -sulfonamide (10.0 mg, 0.024 mmol, 17.9% yield) was prepared according to the general procedure in Example 1 , substituting 7-bromo-N-methylquinoxalin- 2-amine for 6-bromo-3-methylquinazolin-4(3H)-one.
  • Step A 7-Bromoquinoxalin-2(lH)-one (313.1 mg, 1.391 mmol) was dissolved in 0.15M DMF (9.2 mL) and treated with potassium carbonate (288.4 mg, 2.087 mmol) and iodomethane (95.5 iL, 1.530 mmol). The reaction mixture was stirred at ambient temperature for 30 minutes. The reaction mixture was then diluted with water and extracted with EtOAc (2 X). The organics were washed with water (3 X) and brine (1 X), dried over Na 2 S0 4 , filtered and concentrated.
  • Step B N-(2-Chloro-4-fluoro-3-(3-methoxyquinoxalin-6- ylamino)phenyl)propane-l -sulfonamide (12.1 mg, 0.029 mmol, 27.0% yield) was prepared according to the general procedure in Example 1, substituting 7-bromo-2- methoxyquinoxaline for 6-bromo-3-methylquinazolin-4(3H)-one.
  • Step A 7-Bromoquinoxalin-2(lH)-one (313.1 mg, 1.391 mmol) was dissolved in 0.15M DMF (9.2 mL) and treated with potassium carbonate (288.4 mg, 2.087 mmol) and iodomethane (95.5 ⁇ ,, 1.530 mmol). The reaction mixture was stirred at ambient temperature for 30 minutes. The reaction mixture was then diluted with water and extracted with EtOAc (2 X). The organics were washed with water (3 X) and brine (1 X), dried over Na 2 S04, filtered and concentrated.
  • Step B N-(2-Chloro-4-fluoro-3-(4-methyl-3-oxo-3,4-dihydroquinoxalin-6- ylamino)phenyl)propane-l -sulfonamide (1.8 mg, 0.004 mmol, 1.2% yield) was prepared according to the general procedure in Example 1, substituting 7-bromo-l-methylquinoxalin- 2(lH)-one for 6-bromo-3-methylquinazolin-4(3H)-one.
  • Step A Methyl 3-amino-2,6-difluorobenzoate (652.8 mg, 3.488 mmol) was dissolved in 0.2M DCM (17.4 mL), chilled to 0°C, and then treated sequentially with triethylamine (1.42 mL, 10.46 mmol) and furan-2-sulfonyl chloride (1.16 mg, 6.98 mmol). The reaction mixture was warmed to ambient temperature and stirred for 16 hours. The reaction mixture was diluted with EtOAc, washed with water (2 X) and brine (1 X), dried over Na 2 S0 4 , filtered and concentrated.
  • Step B 2,6-Difluoro-3-(furan-2-sulfonamido)benzoic acid (475.0 mg, 1.57 mmol) was dissolved in 0.1M DMF (15.7 mL) and treated with triethylamine (637.4 ⁇ , 4.70 mmol) and then diphenylphosphoryl azide (523.7 ⁇ , 2.350 mmol) at ambient temperature for 1 hour. The reaction mixture was then treated with deionized water (5.0 mL) and heated to 80°C for 16 hours. The reaction mixture was cooled to ambient temperature, diluted with EtOAc and washed with water (3 X) and brine (1 X), dried over Na 2 S0 4 , filtered and concentrated.
  • Step C N-(2,4-Difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)furan-2-sulfonamide (4.5 mg, 0.010 mmol, 9.6% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2,4- difluorophenyl)furan-2-sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l - sulfonamide.
  • Step A 7-Bromo-l-ethylquinoxalin-2(lH)-one: 7-bromoquinoxalin-2(lH)- one (313.6 mg, 1.39 mmol) was dissolved in 0.2M DMF (7.0 mL) and treated with potassium carbonate (288.9 mg, 2.09 mmol) and iodoethane (123.7 ⁇ L, 1.53 mmol) at ambient temperature for 1 hour. The reaction mixture was then diluted with water and extracted with EtOAc (2 X). The organics were washed with water (3 X) and brine (1 X), dried over Na 2 S0 4 , filtered and concentrated.
  • Step B N-(2-Chloro-3-(4-ethyl-3-oxo-3,4-dihydroquinoxalin-6-ylamino)-4- fluorophenyl)propane-l -sulfonamide (5.4 mg, 0.012 mmol, 1.9% yield) was prepared according to the general procedure in Example 1, substituting 7-bromo-l-ethylquinoxalin- 2(lH)-one for 6-bromo-3-methylquinazolin-4(3H)-one.
  • Step A 7-Bromoquinoxalin-2(lH)-one (322.6 mg, 1.43 mmol) was dissolved in 0.1 M DMF (14.4 mL) and treated with potassium carbonate (297.2 mg, 2.15 mmol) and 2- iodopropane (157.7 ih, 1.58 mmol) at 50°C for 2 hours.
  • the reaction mixture was cooled to ambient temperature, diluted with water and extracted with EtOAc (2 X). The organics were washed with water (3 X) and brine (1 X), dried over Na 2 S0 4 , filtered and concentrated.
  • Step B N-(2-Chloro-4-fluoro-3-(4-isopropyl-3-oxo-3,4-dihydroquinoxalin-6- ylamino)phenyl)propane-l -sulfonamide (5.4 mg, 0.012 mmol, 27.7% yield) was prepared according to the general procedure in Example 1, substituting 7-bromo-l- isopropylquinoxalin-2(lH)-one for 6-bromo-3-methylquinazolin-4(3H)-one.
  • Step A DMAP (0.096 g, 0.79 mmol), pyridine (1.67 mL, 20.7 mmol) and propane- 1-sulfonyl chloride (2.32 mL, 20.7 mmol) were added to a solution of 4-mefhyl-3- nitroaniline (3 g, 19.7 mmol) in DCM (25 mL). The reaction was stirred at 45°C for 18 hours. The reaction mixture was then diluted with ethyl acetate and washed with water.
  • Step B A sealed microwave vial was charged with 6-bromo-3- methylquinazolin-4(3H)-one (0.075 g, 0.31 mmol), N-(3-amino-4-methylphenyl)propane-l- sulfonamide (0.079 g, 0.35 mmol), Pd 2 (dba) 3 (0.014 g, 0.016 mmol), (R)-BINAP (0.020 g, 0.031 mmol), sodium tert-butoxide (0.106 g, 1.10 mmol) and toluene (2.3 mL). The mixture was heated in a microwave reactor at 120°C for 15 minutes.
  • dihydroquinazolin-6- 5 9.68 (s, 1H), ylamino)phenyl)-2- 8.46 (s, 1H), methylpropane- 1 - 8.16 (s, 1H),

Abstract

Compounds of Formula (I) are useful for inhibition of Raf kinases. Methods of using compounds of Formula (I), stereoisomers, tautomers and pharmaceutically acceptable salts thereof, for in vitro, in situ, and in vivo diagnosis, prevention or treatment of such disorders in mammalian cells, or associated pathological conditions are disclosed.

Description

HETEROCYCLIC SULFONAMIDES AS RAF INHIBITORS
BACKGROUND OF THE INVENTION
[0001] FIELD OF THE INVENTION
[0002] Compounds, pharmaceutical compositions comprising the compounds, a process for making the compounds and the use of the compounds in therapy are provided herein. More particularly, certain quinazolinone compounds useful for inhibiting Raf kinase and for treating disorders mediated thereby are provided herein.
[0003] DESCRIPTION OF THE STATE OF THE ART
[0004] The Raf/MEK/ERK pathway is critical for cell survival, growth, proliferation and tumorigenesis. Li, Nanxin, et al. "B-Raf kinase inhibitors for cancer treatment." Current Opinion in Investigational Drugs. Vol. 8, No. 6 (2007): 452-456. Raf kinases exist as three isoforms, A-Raf, B-Raf and C-Raf. Among the three isoforms, studies have shown that B- Raf functions as the primary MEK activator. B-Raf is one of the most frequently mutated genes in human cancers. B-Raf kinase represents an excellent target for anticancer therapy based on preclinical target validation, epidemiology and drugability.
[0005] Small molecule inhibitors of B-Raf are being developed for anticancer therapy. Nexavar® (sorafenib tosylate) is a multikinase inhibitor, which includes inhibition of B-Raf, and is approved for the treatment of patients with advanced renal cell carcinoma and unresectable hepatocellular carcinoma. Other Raf inhibitors have also been disclosed or have entered clinical trials, for example SB-590885, RAF-265, PLX-4032, XL-281 and GS - 2118436.
SUMMARY OF THE INVENTION
[0006] Compounds that are inhibitors of Raf kinase are described herein. Certain hyperproliferative disorders are characterized by the over activation of Raf kinase function, for example by mutations or over expression of the protein. Accordingly, the compounds are useful in the treatment of hyperproliferative disorders, such as cancer.
[0007] More specifically, one aspect provides compounds of Formula I:
Figure imgf000003_0001
I
and stereoisomers and pharmaceutically acceptable salts thereof, wherein W, X, Y, Z, L, R1, R2, R3, R4 and R5 are as defined herein and the dashed lines represent optional double bonds.
[0008] Another aspect of the present invention provides methods of preventing or treating a disease or disorder modulated by B-Raf, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof. Examples of such diseases and disorders include, but are not limited to, hyperproliferative disorders (such as cancer, including melanoma and other cancers of the skin), neurodegeneration, cardiac hypertrophy, pain, migraine and neurotraumatic disease.
[0009] Another aspect of the present invention provides methods of preventing or treating cancer, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention, or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds having anti-cancer properties.
[0010] Another aspect of the present invention provides methods of preventing or treating cancer, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention, or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds having anti-cancer properties.
[0011] Another aspect of the present invention provides a method of treating a hyperproliferative disease in a mammal comprising administering a therapeutically effective amount of a compound of this invention to the mammal.
[0012] Another aspect of the present invention provides methods of preventing or treating kidney disease, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention, or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds. Another aspect of the present invention provides methods of preventing or treating polycystic kidney disease, comprising administering to a mammal in need of such treatment an effective amount of a compound of this invention, or a stereoisomer, tautomer, solvate, prodrug or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds.
[0013] Another aspect of the present invention provides the compounds of the present invention for use in therapy.
[0014] Another aspect of the present invention provides the compounds of the present invention for use in the treatment of a hyperproliferative disease. In a further embodiment, the hyperproliferative disease may be cancer (or still further, a specific cancer as defined herein).
[0015] Another aspect of the present invention provides the compounds of the present invention for use in the treatment of a kidney disease. In a further embodiment, the kidney disease may be polycystic kidney disease.
[0016] Another aspect of the present invention provides the use of a compound of this invention in the manufacture of a medicament for the treatment of a hyperproliferative disease. In a further embodiment, the hyperproliferative disease may be cancer (or still further, a specific cancer as defined herein).
[0017] Another aspect of the present invention provides the use of a compound of this invention in the manufacture of a medicament for the treatment of a kidney disease. In a further embodiment, the kidney disease may be polycystic kidney disease.
[0018] Another aspect of the present invention provides the use of a compound of the present invention in the manufacture of a medicament, for use as a B-Raf inhibitor in the treatment of a patient undergoing cancer therapy.
[0019] Another aspect of the present invention provides the use of a compound of the present invention in the manufacture of a medicament, for use as a B-Raf inhibitor in the treatment of a patient undergoing polycystic kidney disease therapy.
[0020] Another aspect of the present invention provides a pharmaceutical composition comprising a compound of the present invention for use in the treatment of a hyperproliferative disease.
[0021] Another aspect of the present invention provides a pharmaceutical composition comprising a compound of the present invention for use in the treatment of cancer.
[0022] Another aspect of the present invention provides a pharmaceutical composition comprising a compound of the present invention for use in the treatment of polycystic kidney disease. [0023] Another aspect of the present invention provides a pharmaceutical composition comprising a compound of this invention, a stereoisomer, tautomer, solvate, prodrug or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
[0024] Another aspect of the present invention provides a pharmaceutical composition comprising a compound of this invention or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier or excipient.
[0025] Another aspect of the present invention provides intermediates for preparing compounds of Formulas I, II, III, IV, V, VI, and VII. Certain compounds of Formulas I, II, III, IV, V, VI, and VII may be used as intermediates for other compounds of Formulas I, II, III, IV, V, VI, and VII.
[0026] Another aspect of the present invention includes methods of preparing, methods of separation, and methods of purification of the compounds of this invention.
DETAILED DESCRIPTION OF THE INVENTION
[0027] Reference will now be made in detail to certain embodiments, examples of which are illustrated in the accompanying structures and formulas. While enumerated embodiments will be described, it will be understood that they are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents, which may be included within the scope of the present invention as defined by the claims. One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described. In the event that one or more of the incorporated literature and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls.
[0028] DEFINITIONS
[0029] The term "alkyl" includes linear or branched-chain radicals of carbon atoms.
Some alkyl moieties have been abbreviated, for example, methyl ("Me"), ethyl ("Et"), propyl ("Pr") and butyl ("Bu"), and further abbreviations are used to designate specific isomers of compounds, for example, 1 -propyl or n-propyl ("n-Pr"), 2-propyl or isopropyl ("i-Pr"), 1- butyl or n-butyl ("n-Bu"), 2-methyl-l -propyl or isobutyl ("i-Bu"), 1-methylpropyl or s-butyl ("s-Bu"), 1,1-dimethylethyl or t-butyl ("t-Bu") and the like. The abbreviations are sometimes used in conjunction with elemental abbreviations and chemical structures, for example, methanol ("MeOH") or ethanol ("EtOH"). [0030] Additional abbreviations used throughout the application may include, for example, benzyl ("Bn"), phenyl ("Ph") and acetate ("Ac").
[0031] The following terms are abbreviated: dimethylsulfoxide ("DMSO"), dimethylformamide ("DMF"), dichloromethane ("DCM"), ethyl acetate ("EtOAc") and tetrahydrofuran ("THF").
[0032] The term "alkenyl" refers to linear or branched-chain monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having "cis" and "trans" orientations, or alternatively, "E" and "Z" orientations. In one example, the alkenyl radical is two to six carbon atoms (C2- C6). In other examples, the alkenyl radical is C2-C5, C2-C4 or C2-C3.
[0033] The term "alkynyl" refers to a linear or branched monovalent hydrocarbon radical with at least one site of unsaturation, i.e., a carbon-carbon triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein. In one example, the alkynyl radical is two to six carbon atoms (C2-C6). In other examples, the alkynyl radical is C2-C5, C2-C4 or C2-C3.
[0034] The term "cycloalkyl" refers to a non-aromatic, saturated or partially unsaturated hydrocarbon ring group, wherein the cycloalkyl group may be optionally substituted independently with one or more substituents described herein. In one example, the cycloalkyl group is 3 to 6 carbon atoms (C3-C6). In other examples, cycloalkyl is C3-C4 or C3-C5. In other examples, the cycloalkyl group, as a monocycle, is C3-C6 or C5-C6. In another example, the cycloalkyl group, as a bicycle, is C7-C12.
[0035] The term "halogen" refers to F, CI, Br or I.
[0036] The terms "heterocyclic" or "heterocycle" or "heterocyclyl" refers to a saturated or a partially unsaturated (i.e., having one or more double and/or triple bonds within the ring) cyclic group in which at least one ring atom is a heteroatom independently selected from nitrogen, oxygen, and sulfur, the remaining ring atoms being carbon. In one embodiment, heterocyclyl includes saturated or partially unsaturated 3-7 membered heterocyclyl groups, another embodiment includes 4-6 membered heterocyclyl groups, and another embodiment includes 5-6 membered heterocyclyl groups. The heterocyclyl group may be optionally substituted with one or more substituents described herein. Heterocycles include 3 to 7 membered rings containing one or two heteroatoms selected from oxygen, nitrogen and sulfur.
[0037] The term "heteroaryl" refers to an aromatic cyclic group in which at least one ring atom is a heteroatom independently selected from nitrogen, oxygen and sulfur, the remaining ring atoms being carbon. Heteroaryl groups may be optionally substituted with one or more substituents described herein. In one example, heteroaryl includes 5-6 membered heteroaryl groups. Heteroaryls includes 5 to 6 membered aromatic rings containing one, two, three or four heteroatoms selected from oxygen, nitrogen and sulfur.
[0038] A "solvate" refers to an association or complex of one or more solvent molecules and a compound of the invention. Examples of solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. The term "hydrate" refers to the complex where the solvent molecule is water.
[0039] The abbreviation "TLC" stands for thin layer chromatography.
[0040] The terms "treat" or "treatment" refer to therapeutic, prophylactic, palliative or preventative measures. Beneficial or desired clinical results include, but are not limited to, alleviation of symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder, as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.
[0041] The phrases "therapeutically effective amount" or "effective amount" mean an amount of a compound described herein that, when administered to a mammal in need of such treatment, sufficient to (i) treat or prevent the particular disease, condition, or disorder, (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, condition, or disorder, or (iii) prevent or delay the onset of one or more symptoms of the particular disease, condition, or disorder described herein. The amount of a compound that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the mammal in need of treatment, but can nevertheless be routinely determined by one skilled in the art.
[0042] The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by abnormal or unregulated cell growth. A "tumor" comprises one or more cancerous cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g., epithelial squamous cell cancer), lung cancer including small- cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, skin cancer, including melanoma, as well as head and neck cancer.
[0043] The phrase "pharmaceutically acceptable" indicates that the substance or composition is compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
[0044] The phrase "pharmaceutically acceptable salt," as used herein, refers to pharmaceutically acceptable organic or inorganic salts of a compound described herein.
[0045] The compounds described herein also include other salts of such compounds that are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds described herein and/or for separating enantiomers of compounds described herein.
[0046] The term "mammal" means a warm-blooded animal that has or is at risk of developing a disease described herein and includes, but is not limited to, guinea pigs, dogs, cats, rats, mice, hamsters, and primates, including humans.
[0047] The terms "compound of this invention," and "compounds of the present invention", unless otherwise indicated, include compounds of Formulas I, II, III, IV, V, VI, and VII, stereoisomers, tautomers, solvates, prodrugs and salts (e.g., pharmaceutically acceptable salts) thereof. Unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds of Formulas I, II, III, IV, V, VI, and VII, wherein one or more hydrogen atoms are replaced deuterium or tritium, or one or more carbon atoms are replaced by a 13C- or 14C-enriched carbon are within the scope of this invention.
[0048] RAF INHIBITORS
[0049] Provided herein are compounds, and pharmaceutical formulations thereof, that are potentially useful in the treatment of diseases, conditions and/or disorders modulated by B-Raf.
[0050] One embodiment provides compounds of Formula I:
Figure imgf000009_0001
I
and stereoisomers, tautomers, solvates, prodrugs and pharmaceutically acceptable salts thereof, wherein:
the dashed lines represent optional double bonds;
W is selected from N, NR6, C(=0), CR7 and CHR7;
X is selected from N, NR8, C(=0), CR9 and CHR9, provided when:
(i) W is N, then X is CR9,
(ii) W is NR6, then X is C(=0) or CHR9,
(iii) W is C(=0), then X is NR8 or CHR9,
(iv) W is CR7, then X is N or CR9, and
(v) W is CHR7, then X is NR8, C(=0) or CHR9;
Y is selected from N, NR10, CR11 and CHR11;
Z is selected from N, NR12, CR13 and CHR13;
L is selected from O, NR14, S, C(-O) and CR15R16;
R and R" are independently selected from hydrogen, halogen, CN, Ci-C3 alkyl, C1-C3 alkoxy and C2-C3 alkynyl;
R is selected from hydrogen, halogen and C1-C3 alkyl;
R4 is selected from C3-C5 cycloalkyl, C C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, phenyl, a 5-6 membered heteroaryl, or NRaRb, wherein the cycloalkyl, alkyl, alkenyl, alkynyl, phenyl and heteroaryl are optionally substituted with OR0, halogen, phenyl, C3-C4 cycloalkyl, or C C4 alkyl optionally substituted with halogen;
R5 is selected from hydrogen, C1-C3 alkyl and Ci-C3 alkoxy, wherein the alkyl and alkoxy are optionally substituted with Rd;
R6 is selected from hydrogen, Ci-C6 alkyl, Ci-Ce alkenyl, Q-C6 alkynyl, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORe, SRe, NRfRg, phenyl, Ci-C4 alkyl, C1-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp; R7 is selected from hydrogen, C C6 alkyl, Ci-C alkenyl, Ci-C^ alkynyl, Q-Q alkoxy, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, a 5 to 6 membered heteroaryl, and NRhR', wherein the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORe, SRe, NRfRg, Q- i alkyl, C C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with R ;
Q
R is selected from hydrogen, Q-Q alkyl, C C6 alkenyl, C C6 alkynyl, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), OR", SRJ, NRkR*, phenyl, Q-C4 alkyl, Q-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp;
R9 is selected from hydrogen, C!-C6 alkyl, Ci-C6 alkenyl, C!-C6 alkynyl, Ci-Ce alkoxy, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, a 5 to 6 membered heteroaryl, and NRmRn, wherein the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORJ, SRJ, NRkR*, C C4 alkyl, d-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp;
R10 is selected from hydrogen and halogen;
R11 is selected from hydrogen and halogen, or
R11 and R14 together with the atoms to which they are attached form a 5-6 membered heterocyclyl or heteroaryl optionally substituted with oxo, or
R11 and R15 together with the atoms to which they are attached form a 5-6 membered carbocyclyl;
R12 is hydrogen;
R13 is selected from hydrogen and halogen;
R14 is selected from hydrogen and C C4 alkyl;
R15 and R16 are independently selected from hydrogen and Q-C3 alkyl;
Ra and Rb are independently selected from hydrogen or C Cs alkyl optionally substituted by halogen, or
Ra and Rb are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or C!-C3 alkyl;
Rc is selected from hydrogen and C!-C6 alkyl optionally substituted by halogen;
each Rd is selected from halogen and OH; Re is selected from hydrogen and Q-C4 alkyl;
Rf and R8 are independently selected from hydrogen and Q-C4 alkyl optionally substituted with halogen;
Rh and R1 are independently selected from hydrogen and C1-C4 alkyl optionally substituted with halogen;
RJ is selected from hydrogen and C1-C4 alkyl;
Rk and R1 are independently selected from hydrogen and Q-C4 alkyl optionally substituted with halogen;
Rm and R" are independently selected from hydrogen and C!-C4 alkyl optionally substituted with halogen; and
each Rp is independently selected from halogen, oxo, Q-C4 alkyl and Q-C4 alkoxy, wherein the alkyl and alkoxy are optionally substituted with halogen.
[0051] One embodiment provides compounds of Formula II:
Figure imgf000011_0001
1 2 3 and stereoisomers and pharmaceutically acceptable salts thereof, wherein W, X, L, R , R , R , R4, R5 and Rn are as defined herein and the dashed lines represent optional double bonds.
[0052] One embodiment provides compounds of Formula III:
Figure imgf000011_0002
1 2 3 and stereoisomers and pharmaceutically acceptable salts thereof, wherein W, X, L, R , R , R , R4, R5 and R11 are as defined herein and the dashed line between W and X represents an optional double bond.
[0053] One embodiment provides compounds of Formula IV:
Figure imgf000012_0001
IV
and stereoisomers and pharmaceutically acceptable salts thereof, wherein L, R1, R2, R3, R4, R5, R6 and Rn are as defined herein.
[0054] One embodiment provides compounds of Formula V:
Figure imgf000012_0002
V
and stereoisomers and pharmaceutically acceptable salts thereof, wherein L, R1, R2, R3, R4, R5, R7 and R11 are as defined herein.
[0055] One embodiment provides compounds of Formula VI:
Figure imgf000012_0003
VI
and stereoisomers and pharmaceutically acceptable salts thereof, wherein L, R1, R2, R3, R4, R5, R9 and Rn are as defined herein.
[0056] One embodiment provides compounds of Formula VII:
Figure imgf000012_0004
VII
and stereoisomers and pharmaceutically acceptable salts thereof, wherein L, R1, R2, R3, R4, R5, R7, R8 and R11 are as defined herein. [0057] In certain embodiments, W is selected from N, NR6, C(=0), CR7 and CHR7.
In certain embodiments, W is selected from N,
Figure imgf000013_0001
.
[0058] In certain embodiments, R6 is selected from hydrogen, C!-C6 alkyl, C C6 alkenyl, C C6 alkynyl, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORe, SRe, NRfR8, phenyl, Q-Q alkyl, C C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp. In certain embodiments, R6 is selected from C\- e alkyl, phenyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, phenyl and heteroaryl are optionally substituted with phenyl or C1-C4 alkyl optionally substituted with Rp. In certain embodiments, R6 is methyl, benzyl, phenyl or 1 -methyl- 1H- pyrazol-4-yl.
[0059] In certain embodiments, R is selected from hydrogen, C\-C(, alkyl, Q-Ce alkenyl, Q-Q alkynyl, Ci-C6 alkoxy, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, a 5 to 6 membered heteroaryl, and NRhR', wherein the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORe, SRe, NRfRg, Q-C4 alkyl, C1-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp. In certain embodiments, R7 is selected from hydrogen, C\-C(, alkoxy and NRhR'. In certain embodiments, Rh and R1 are independently selected from hydrogen and C1-C4 alkyl optionally substituted with halogen. In certain embodiments, R7 is hydrogen, OCH3 or NHCH3.
[0060] In certain embodiments, X is selected from N, NR8, C(=0), CR9 and CHR9.
Q
In certain embodiments, X is selected from N, NR , C(=0) and CR .
o
[0061] In certain embodiments, R is selected from hydrogen, Q-Q alkyl, Q-C6 alkenyl, C!-C6 alkynyl, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORJ, SRJ, NRkR*, phenyl, Q-C4 alkyl, Q-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp. In certain embodiments, R8 is C - C6 alkyl. In certain embodiments, R is methyl, ethyl or isopropyl.
[0062] In certain embodiments, R9 is selected from hydrogen, C\-C(, alkyl, C C6 alkenyl, C C6 alkynyl, C C6 alkoxy, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, a 5 to 6 membered heteroaryl, and NRmRn, wherein the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORJ, SRJ, NRkR', Q-C4 alkyl, Q-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp. In certain embodiments, R9 is selected from hydrogen, Cy-Ce alkoxy, a 3 to 6 membered heterocyclyl, and NRmRn, wherein the alkoxy and heterocyclyl are optionally substituted with C C4 alkyl optionally substituted Rp. In certain embodiments, Rm and Rn are independently selected from hydrogen and C1-C4 alkyl optionally substituted with halogen. In certain embodiments, each Rp is independently selected from halogen, oxo, Ct-Q alkyl and C1-C4 alkoxy, wherein the alkyl and alkoxy are optionally substituted with halogen. In certain embodiments, R9 is hydrogen, OCH3, OCH2CH3, NHCH3, N(CH3)2, piperazin-l-yl, 4- methylpirperazin-l-yl or tert-butyl 4-piperazine-l-carboxylate.
[0063] In the present invention, when:
(i) W is N, then X is CR9,
(ii) W is NR6, then X is C(=0) or CHR9,
(iii) W is C(=0), then X is NR8 or CHR9,
(iv) W is CR7, then X is N or CR9, and
(v) W is CHR7, then X is NR8, C(=0) or CHR9.
[0064] In certain embodiments, when:
(i) W is N, then X is CR9,
(ii) W is NR6, then X is C(=0),
(iii) W is C(=0), then X is NR8, and
(iv) W is CR7, then X is N or CR9.
[0065] In certain embodiments, Y is selected from N, NR10, CR11 and CHR11. In certain embodiments, Y is CR11. In certain embodiments, R11 is selected from hydrogen and halogen. In certain embodiments, Y is CH or CC1.
[0066] In certain embodiments, Z is selected from N, NR12, CR13 and CHR13. In
13 12
certain embodiments, Z is CR . In certain embodiments, R is hydrogen. In certain embodiments, R is selected from hydrogen and halogen. In certain embodiments, R is hydrogen. In certain embodiments, Z is CH.
[0067] In certain embodiments the residue:
Figure imgf000014_0001
wherein the wavy line represents the point of attachment of the residue to L, is selected from:
Figure imgf000015_0001

Figure imgf000016_0001
Figure imgf000016_0002
wherein the wav line represents the point of attachment of the residue to L, is selected from:
Figure imgf000016_0003
[0069] In certain embodiments, L is selected from O, NR. , S, C(=0) and CR R .
In certain embodiments, L is selected from O and NR14. In certain embodiments, R14 is selected from hydrogen and Ci-C4 alkyl. In certain embodiments, L is O, NH, N(CH3) or N(CH2CH3).
[0070] In certain embodiments, L is O.
[0071] In certain embodiments, L is NR14. In certain embodiments, R14 is selected from hydrogen and C C4 alkyl. In certain embodiments, R14 is hydrogen. In certain embodiments, R14 is Ci-C4 alkyl. In certain embodiments, L is NH, N(CH3) or N(CH2CH3). In certain embodiments, L is N(CH3) or N(CH2CH3). In certain embodiments, L is NH.
1
[0072] In certain embodiments, R and R are independently selected from hydrogen, halogen, CN, Ci-C3 alkyl, Ci-C3 alkoxy or C1-C3 alkynyl. In certain embodiments, R1 and R2 are independently selected from hydrogen, halogen, CN, C C3 alkyl or C C3 alkoxy.
1 9
[0073] In certain embodiments, R , R and R are independently selected from hydrogen, halogen or 1 2 3
C1-C3 alkyl. In certain embodiments, R , R and R are independently selected from hydrogen, F, CI or methyl.
[0074] In certain embodiments, R1 is hydrogen, halogen, CN, C C3 alkyl, C1-C3 alkoxy or C2-C3 alkynyl. In certain embodiments, R1 is hydrogen, halogen, CN, Q-C3 alkyl or Ci-C3 alkoxy. In certain embodiments, R1 is hydrogen or halogen. In certain embodiments, R1 is hydrogen, CI or F.
[0075] In certain embodiments, R1 is hydrogen.
[0076] In certain embodiments, R1 is halogen. In certain embodiments, R1 is F or CI.
[0077] In certain embodiments, R1 is C!-C3 alkyl. In certain embodiments, R1 is methyl.
[0078] In certain embodiments, R is hydrogen, halogen, CN, C C3 alkyl, Q-C3 alkoxy or C2-C3 alkynyl. In certain embodiments, R is hydrogen, halogen, CN, Ci-C3 alkyl
2 2 or C C3 alkoxy. In certain embodiments, R is halogen or CN. In certain embodiments, R is CI, F or CN. In certain embodiments, R2 is CI or CN.
[0079] In certain embodiments, R is hydrogen.
9 9
[0080] In certain embodiments, R is halogen. In certain embodiments, R is F or CI.
In certain embodiments, R is CI.
[0081] In certain embodiments, R is C1-C3 alkyl. In certain embodiments, R is methyl.
[0082] In certain embodiments, R is hydrogen, halogen or C1-C3 alkyl. In certain embodiments, R is hydrogen.
[0083] In certain embodiments, R is halogen. In certain embodiments, R is F or CI.
1 9 '
[0084] In certain embodiments, R and R are F and R is hydrogen.
1 9 ^
[0085] In certain embodiments, R is hydrogen and R and R are F.
[0086] In certain embodiments, R is hydrogen, R is F and R is CI.
[0087] In certain embodiments, R is F and R is CI and R is hydrogen.
[0088] In certain embodiments, R1 is CI and R2 is F and R3 is hydrogen.
1 9
[0089] In certain embodiments, R is F and R and R are hydrogen.
[0090] In certain embodiments, R and R are hydrogen and R is F.
[0091] In certain embodiments, R and R are hydrogen and R is CI.
1 9
[0092] In certain embodiments, R is CI and R and R are hydrogen.
[0093] In certain embodiments, R1, R2 and R3 are F.
1 9 "X
[0094] In certain embodiments, R is F and R is methyl and R is hydrogen.
1 9 "X
[0095] In certain embodiments, R is methyl and R is F and R is hydrogen.
[0096] In certain embodiments, R2 is F and R1 and R3 are hydrogen.
1 9 "X
[0097] In certain embodiments, R is CI, R is ethynyl and R is hydrogen.
1 9 "
[0098] In certain embodiments, R is F, R is ethynyl and R is hydrogen.
[0099] In certain embodiments, R1 and R3 are hydrogen and R2 is CN. [00100] In certain embodiments, R1 is F, R2 is CN and R3 is hydrogen.
[00101] In certain embodiments, R1 is CI, R2 is CN and R3 is hydrogen.
[00102] In certain embodiments, R is methyl and R and R are hydrogen.
[00103] In certain embodiments, the residue:
Figure imgf000018_0001
wherein the wav line represents the point of attachment of the residue to L, is selected from:
Figure imgf000018_0002
Figure imgf000019_0001
[00104] In certain embodiments, R4 is selected from C3-C5 cycloalkyl, C!-C6 alkyl, C2- C6 alkenyl, C2-C6 alkynyl, phenyl, a 5-6 membered heteroaryl, or NRaRb, wherein the cycloalkyl, alkyl, alkenyl, alkynyl, phenyl and heteroaryl are optionally substituted with ORc, halogen, phenyl, C3-C cycloalkyl, or Ci-C4 alkyl optionally substituted with halogen. In certain embodiments, R4 is selected from Ci-Ce alkyl, a 5-6 membered heteroaryl, or NRaRb, wherein the alkyl and heteroaryl are optionally substituted with halogen or C3-C4 cycloalkyl. In certain embodiments, Ra and Rb are independently selected from hydrogen or C Cs alkyl optionally substituted by halogen. In certain embodiments, Ra and Rb are Q-Cs alkyl. In certain embodiments, Ra and Rb are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or Ci- C3 alkyl. In certain embodiments, Ra and Rb are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl. In certain embodiments, Ra and Rb are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl, wherein the heterocyclyl contains one nitrogen heteroatom. In certain embodiments, Ra and Rb are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl, wherein the heterocyclyl is pyrrolidine. In certain embodiments, R4 is propyl, isobutyl, -CH2CH2CH2F, cyclopropylmethyl, -N(CH3)2, pyrrolidine, or furan-2-yl.
[00105] In certain embodiments, R4 is C3-C4 cycloalk l, Q-Q alkyl optionally substituted with halogen or C3-C4 cycloalkyl, or NRaRb. In certain embodiments, Ra and Rb are independently selected from hydrogen and Q-Cs alkyl.
[00106] In certain embodiments, R4 is C3-C5 cycloalkyl, C C6 alkyl, C2-C6 alkenyl or C2-C6 alkynyl, wherein the cycloalkyl, alkyl, alkenyl and alkynyl are optionally substituted with ORc, halogen or C3-C4 cycloalkyl.
[00107] In certain embodiments, R4 is cyclopropyl, ethyl, propyl, butyl, isobutyl, -CH2C1, -CH2CF3, -CH2CH2CH2F, ^CH2CH2CF3, phenylmethyl, cyclopropylmethyl, phenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2,5-difluorophenyl, 4-chloro-3- trifluoromethylphenyl, 1 -methyl- lH-imidazol-4-yl, furan-2-yl, pyridin-2-yl, pyridin-3-yl, thiophen-2-yl, -NHCH2CH3, -NHCH2CH2CH3, -N(CH3)CH2CH3, -N(CH3)2, or pyrrolidine.
[00108] In certain embodiments, R4 is cyclopropyl, propyl, butyl, isobutyl, -CH2C1, -CH2CF3, -CH2CH2CH2F, -CH2CH2CF3, cyclopropylmethyl, -NHCH2CH2CH3, -N(CH3)CH2CH3, -N(CH3)2, or pyrrolidine.
[00109] In certain embodiments, R4 is cyclopropyl, propyl, butyl, isobutyl, -CH2C1, -€H2CF3, -CH2CH2CH2F, -CH2CH2CF3, cyclopropylmethyl or -NHCH2CH2CH3.
[00110] In certain embodiments, R4 is propyl, butyl, isobutyl, -CH2CH2CH2F, -CH2CH2CF3 or cyclopropylmethyl.
[00111] In certain embodiments, R4 is C3-C5 cycloalkyl or C C6 alkyl optionally substituted with OH, halogen or C3-C4 cycloalkyl.
[00112] In certain embodiments, R4 is C3-C5 cycloalkyl. In certain embodiments, R4 is C3-C4 cycloalkyl. In certain embodiments, R4 is cyclopropyl or cyclobutyl. In certain embodiments, R4 is cyclopropyl.
[00113] In certain embodiments, R4 is Ci-C6 alkyl. In certain embodiments, R4 is ethyl, propyl, butyl or isobutyl. In certain embodiments, R4 is propyl or isobutyl.
[00114] In certain embodiments, R4 is C C6 alkyl optionally substituted with halogen. In certain embodiments, R4 is -CF3, -CH2C1, -CH2CF3, -CH2CH2CH2F, -CH2CH2CF3, -CF2CF3 or -CF2CF2CF3. In certain embodiments, R4 is -CH2CH2CH2F or -CH2CH2CF3. In certain embodiments, R4 is -CH2CH2CH2F.
[00115] In certain embodiments, R4 is Ci-Ce alkyl optionally substituted with OH, halogen or C3-C4 cycloalkyl. In certain embodiments, R4 is cyclopropylmethyl (-CH2- cyclopropyl) or cyclobutylmethyl (-CH2-cyclobutyl). In certain embodiments, R4 is cyclopropylmethyl .
[00116] In certain embodiments, R4 is Q-C6 alkyl optionally substituted with phenyl. In certain embodiments, R4 is phenylmethyl.
[00117] In certain embodiments, R4 is phenyl optionally substituted with ORc, halogen, C3-C4 cycloalkyl, or C1-C4 alkyl optionally substituted with halogen. In certain embodiments, R4 is phenyl optionally substituted with halogen. In certain embodiments, R4 is phenyl optionally substituted with C C4 alkyl optionally substituted with halogen. In certain embodiments, R4 is phenyl optionally substituted with halogen and Q-C4 alkyl optionally substituted with halogen. In certain embodiments, R4 is phenyl. In certain embodiments, R4 is phenyl, 2-fluorophenyl, 3 -fluorophenyl, 4-fluorophenyl, 2,5- difluorophenyl or 4-chloro-3-trifluoromethylphenyl.
[00118] In certain embodiments, R4 is a 5-6 membered heteroaryl optionally substituted with ORc, halogen, C3-C4 cycloalkyl or Ct-C4 alkyl optionally substituted with halogen. In certain embodiments, R4 is a 5-6 membered heteroaryl optionally substituted with C!-C4 alkyl. In certain embodiments, R4 is a 5-6 membered heteroaryl, wherein the heteroaryl contains one or two heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur. In certain embodiments, R4 is a 5-6 membered heteroaryl, wherein the heteroaryl is imidazolyl, furanyl, pyridinyl or thiophenyl. In certain embodiments, R4 is 1- methyl-lH-imidazol-4-yl, furan-2-yl, pyridin-2-yl, pyridin-3-yl or thiophen-2-yl. In certain embodiments, R4 is furan-2-yl.
[00119] In certain embodiments, R4 is NRaRb. In certain embodiments, Ra and Rb are independently selected from hydrogen and Q-Cs alkyl. In certain embodiments, Ra is hydrogen. In certain embodiments, Ra is Ci-C6 alkyl. In certain embodiments, Ra is ethyl or propyl. In certain embodiments, R4 is selected from the group consisting of -NHCH2CH3, -NHCH2CH2CH3, -N(CH3)CH2CH3 and -N(CH3)2. In certain embodiments, R4 is -N(CH3)2.
[00120] In certain embodiments, Ra and Rb together with the nitrogen to which they are attached form a 3 to 6 membered heterocyclyl ring. In certain embodiments, Ra and Rb together with the nitrogen to which they are attached form a 3 to 6 membered heterocyclic ring, wherein the heterocyclic ring contains one nitrogen heteroatom. In certain embodiments, R4 is pyrrolidine. In certain embodiments, R4 is pyrrolidin-l-yl.
[00121] In certain embodiments, R4 is selected from propyl, cyclopropylmethyl, -CH2CH2CH2F and phenyl. In a further embodiment, R4 is selected from propyl, cyclopropylmethyl and -CH2CH2CH2F.
[00122] In certain embodiments, R5 is selected from hydrogen, Q-C3 alkyl and C!-C3 alkoxy, wherein the alkyl and alkoxy are optionally substituted with Rd. In certain embodiments, R5 is hydrogen.
[00123] In certain embodiments, R11 and R14 together with the atoms to which they are attached form a 5-6 membered heterocyclyl or heteroaryl optionally substituted with oxo. This embodiment includes the following formula:
Figure imgf000022_0001
wherein V is O, N or CH; x is 1 or 2; the dashed lines represent optional double bonds, and W, X, Z, R1, R2, R3, R4 and R5 are as defined herein. In certain embodiments, compounds of the present invention have the following formulas:
Figure imgf000022_0002
[00124] In certain embodiments, R11 and R15 together with the atoms to which they are attached form a 5-6 membered carbocyclyl. This embodiment includes the following formula:
Figure imgf000023_0001
wherein V is O, N or CH; x is 1 or 2; the dashed lines represent optional double bonds, and W, X, Z, R1, R2, R3, R4 and R5 are as defined herein. In certain embodiments, compounds of the present invention have the following formulas:
Figure imgf000023_0002
[00125] In certain embodiments,
W is selected from N, NR6, C(=0) and CR7;
X is selected from N, NR8, C(=0) and CR9;
Y is CR11;
Z is CH; and
R5 is hydrogen.
[00126] In certain embodiments,
W is selected from N, NR6, C(=0) and CR7;
X is selected from N, NR8, C(=0) and CR9, provided when:
(i) W is N, then X is CR9, (ii) W is NR.6, then X is C(=0),
(iii) W is C(=0), then X is NR8, and
(iv) W is CR7, then X is N or CR9;
Y is CR11;
Z is CH;
L is selected from O and NR14;
R5 is hydrogen;
R6 is selected from C C6 alkyl, phenyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, phenyl and heteroaryl are optionally substituted with phenyl or C1-C4 alkyl optionally substituted with Rp;
R7 is selected from hydrogen, CrC6 alkoxy and NRhR';
R8 is Ci-Ce alkyl;
R9 is selected from hydrogen, Ci-C , alkoxy, a 3 to 6 membered heterocyclyl, and NRmR", wherein the alkoxy and heterocyclyl are optionally substituted with C C4 alkyl optionally substituted R ;
R11 is selected from hydrogen and halogen; and
R is hydrogen.
[00127] It will be appreciated that certain compounds described herein may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of the compounds described herein, including but not limited to, diastereomers, enantiomers and atropisomers, as well as mixtures thereof such as racemic mixtures, form part of the present compounds.
[00128] In the structures shown herein, where the stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds described herein. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined.
[00129] It will also be appreciated that certain compounds of Formulas I, II, III, IV, V, VI, and VII may be used as intermediates for further compounds of Formulas I, II, III, IV, V, VI, and VII.
[00130] It will be further appreciated that the compounds described herein may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the compounds embrace both solvated and unsolvated forms.
[00131] SYNTHESIS OF COMPOUNDS [00132] Compounds described herein may be synthesized by synthetic routes that include processes analogous to those well-known in the chemical arts, particularly in light of the description contained herein. The starting materials are generally available from commercial sources such as Sigma-Aldrich (St. Louis, MO), Alfa Aesar (Ward Hill, MA), or TCI (Portland, OR), or are readily prepared using methods well known to those skilled in the art (e.g., prepared by methods generally described in Louis F. Fieser and Mary Fieser, Reagents for Organic Synthesis, v. 1-23, New York: Wiley 1967-2006 ed. (also available via the Wiley InterScience® website), or Beilsteins Handbuch der organischen Chemie, 4, Aufl. ed. Springer- Verlag, Berlin, including supplements (also available via the Beilstein online database)).
[00133] In preparing compounds of Formula I, II, III, IV, V, VI, and VII, protection of remote functionalities (e.g., primary or secondary amines, etc.) of intermediates may be necessary. The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods. Suitable amino-protecting groups (NH-Pg) include acetyl, trifluoroacetyl, t-butyloxycarbonyl ("Boc"), benzyloxycarbonyl ("CBz") and 9-fluorenylmethyleneoxycarbonyl ("Fmoc"). The need for such protection is readily determined by one skilled in the art. For a general description of protecting groups and their use, see T. W. Greene, et al. Greene's Protective Groups in Organic Synthesis. New York: Wiley Interscience, 2006.
[00134] For illustrative purposes, Schemes 1-18 show general methods for preparing the compounds described herein, as well as key intermediates. For a more detailed description of the individual reaction steps, see the Examples section below. Those skilled in the art will appreciate that other synthetic routes may be used to synthesize the compounds. Although specific starting materials and reagents are depicted in the Schemes and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions. In addition, many of the compounds prepared by the methods described below can be further modified in light of this disclosure using conventional chemistry well known to those skilled in the art.
Figure imgf000026_0001
1.6
Scheme 1
[00135] Scheme 1 shows a general method for preparing a compound 1.6, wherein R , R2, R3 and R4 are as defined herein. A benzoic acid 1.1 is esterified to an alkyl benzoate 1.2 (wherein R is alkyl), e.g., by treatment with tnmethylsilyl diazomethane in MeOH or via Fischer esterification conditions, such as treatment with trimethylsilyl chloride ("TMSC1") in MeOH. Reduction of nitro intermediate 1.2 to its amino analog 1.3 is performed using a standard condition, such as treatment with Pd/C and H2. Bis-sulfonamide 1.4 is obtained by treatment of the aniline 1.3 with a sulfonyl chloride, R4S02C1, in the presence of a base, such as NEt3, in an organic solvent, such as dichloromethane ("DCM"). Hydrolysis of compound 1.4 is accomplished under basic conditions, such as aqueous NaOH, in the appropriate solvent system, such as THF and/or MeOH, to provide a carboxylic acid 1.5. This compound in a suitable solvent, such as THF, is treated with diphenylphosphonic azide ("DPP A") and a base such as triethylamine, and subsequently hydrolyzed to form an amine 1.6.
Figure imgf000027_0001
Scheme 2
[00136] Scheme 2 describes the synthesis of aniline intermediates 2.7, wherein R1, R2', R3, R4 and R" are as defined herein. A benzoic acid ester 2.1 is treated with an alkoxide,
2' 2'
NaOR (wherein R is C1-C3 alkyl), in an appropriate solvent, such as methanol, to form the ether intermediate 2.2. Reduction of the nitro group affords aniline 2.3, which is reacted with a sulfonyl chloride, R4S02C1, in the presence of base, such as pyridine, to give a sulfonamide intermediate 2.4. Benzylation with an optionally substituted benzyl halide, for example p- methoxybenzyl chloride, (wherein LG is a leaving group such as chloro, bromo, iodo, triflate, tosylate; and R" is hydrogen, C C3 alkyl or Ci-C alkoxy; and in one example, R" is hydrogen, in another example, R" is OMe) in the presence of a base, such as sodium hydride, yields the protected sulfonamide ester 2.5, which is hydrolyzed with aqueous base, such as NaOH, to form the acid 2.6. In the last step, application of Curtius rearrangement conditions and subse uent hydrolysis gives the amino intermediate 2.7.
Figure imgf000027_0002
Scheme 3
[00137] Scheme 3 shows a procedure for generating the aniline intermediate 3.1, wherein R" and LG are defined in Scheme 2 and R1, R2, R3 and R4 are as defined herein, through protection of the sulfonamide moiety of aniline 1.6. This transformation can be accomplished by treatment with an optionally substituted benzyl halide (e.g., p- methox benzyl chloride) and a base, such as sodium hydride.
Figure imgf000028_0001
4.1 4.2 4.3 1.3
Scheme 4
[00138] Scheme 4 describes the synthesis of an aniline ester of Formula 1.3, wherein R , R and RJ are defined herein and R is alkyl, such as methyl or ethyl, or benzyl. The amino group of an aniline 4.1 is protected by reacting with hexane-2,5-dione in the presence of a catalytic amount of an acid, such as p-toluenesulfonic acid, in a solvent, such as toluene, to form the 2,5-dimethylpyrrole derivative 4.2. Reaction with a carbamoyl chloride, RO(C=0)Cl, in the presence of n-butyllithium or a comparable agent in a suitable solvent, such as THF, leads to formation of the ester analog 4.3. The amino function of compound 4.3 is deprotected by reaction with hydroxylamine in a suitable solvent, such as ethanol, leading to formation of intermediate 1.3.
Figure imgf000028_0002
4 1 5.1 5.2
Scheme 5
[00139] Scheme 5 describes the synthesis of an aniline ester of Formula 1.3, wherein
R 1', R2", and R 3J are defined herein and R is small alkyl, such as methyl or ethyl, or benzyl. The amino group of an aniline 4.1 is protected by reacting with 1,2- bis(chlorodimethylsilyl)ethane in the presence of a strong base, such as n-butyllithium, in a suitable solvent, such as THF, at low temperatures, e.g., -78°C, to form the l-aza-2,5- disilacyclopentane intermediate 5.1. Compound 5.1 is immediately reacted with a carbamoyl chloride, RO(C=0)Cl, in the presence of n-butyllithium or a comparable agent in a suitable solvent, such as THF, leading to formation of the ester analog 5.2. The amino function of compound 5.2 is deprotected by reaction with an acid, such as HC1, in a suitable solvent, leadin to formation of intermediate 1.3.
Figure imgf000029_0001
Scheme 6
[00140] Scheme 6 describes another way of synthesizing an intermediate of Formula 1.6, wherein R1, R2, R3 and R4 are as defined herein. Bis-sulfonamide 6.2 is obtained by treatment of the aniline 6.1 with a sulfonyl chloride, R4S02C1, in the presence of a base, such as NEt3, in an organic solvent, such as dichloromethane. Hydrolysis of compound 6.2 is accomplished under basic conditions, such as aqueous NaOH, in the appropriate solvent system, such as THF and/or MeOH, to provide the mono-sulfonamide 6.3. This compound in a suitable solvent, such as ethanol, is treated with a reducing agent, such as iron, and additional reagents, such as ammonium chloride, to form an amine 1.6.
Figure imgf000029_0002
Scheme 7
[00141] Scheme 7 shows another way of preparing an intermediate of Formula 1.6. This transformation is accomplished by mono-sulfonylation of a diamino derivative 7.1 with a sulfonyl chloride, R4S02C1, in the presence of a base, such as pyridine, in an organic solvent, such as dichloromethane.
Figure imgf000030_0001
8.2
Scheme 8
[00142] Scheme 8 describes the synthesis of an intermediate of Formula 8.2, a subset of Formula 1.6 compounds, wherein R1, R3 and R4 are as defined herein and R2 is hydrogen. This transformation is accomplished by reduction of the chloro atom of compound 8.1 using reducing conditions, such as hydrogen in the presence of a palladium catalyst in a suitable solvent, such as ethanol.
Figure imgf000030_0002
Scheme 9
[00143] Scheme 9 describes the synthesis of an intermediate of Formula 9.2, a subset of Formula 1.6 compounds, wherein R2, R3 and R4 are as defined herein and R1 is hydrogen. This transformation is accomplished by reduction of the chloro atom of compound 9.1 using reducing conditions, such as hydrogen in the presence of a palladium catalyst in a suitable solvent, such as ethanol.
Figure imgf000030_0003
Scheme 10
[00144] Scheme 10 shows a method for preparing nitrile-substituted aniline intermediates 10.2. Reaction of fluoronitrile 10.1 with the sodium salt of H2NS02R4 (generated by a strong base, such as sodium hydride) in a suitable solvent, such as dimethylsulfoxide or N-methylpyrrolidone, at elevated temperature, results in the formation of intermediate 10.2.
Figure imgf000031_0001
Figure imgf000031_0002
Figure imgf000031_0003
11.6 11 .5
Scheme 11
[00145] Scheme 11 shows a general method for preparing compounds 11.2 and 11.6, wherein W, X, Y, Z, R1, R2, R3, R4 and R8 are as defined herein. Amino heterocycle 11.1 can be coupled with aniline 1.6 under standard Buchwald coupling conditions utilizing a Pd catalyst, such as Pd(dba)2, and a ligand, such as BINAP, to give biarylamine 11.2. Alternatively, aniline 1.6 can be protected with a suitable protecting group, for example PMB, with a suitable base, such as NaH, in a solvent, such as DMF, to give 11.3, and then coupled to give 11.4 and deprotected, for example with neat TFA at room temperature or DCM/TFA at elevated temperatures to provide 11.2. Biaryl amine 11.4 may also be alkylated on nitrogen utilizing a suitable base, preferably NaH, and an alkylating agent, such as Mel, Etl, allyl bromide or cyclopropylmethyl methanesulfonate. Subsequent deprotection provides compound 11.6.
Figure imgf000032_0001
Scheme 12
[00146] Scheme 12 shows a general method for preparing compound 12.2, wherein R1, R2, R3, R4, R6 and R14 are as defined herein. Biaryl amine 12.1 can be chlorinated in the presence of a chlorinating reagent, such as n-chlorosuccinimide, in a variety of solvents, including THF, DMF or CHC13, to provide compound 12.2. Alternatively, protected sulfonamide 12.3, where PG is a protecting group, can be chlorinated and then deprotected with TFA to provide compound 12.2.
Figure imgf000032_0002
Scheme 13
[00147] Scheme 13 shows a general method for preparing compounds 13.2 and 13.3, wherein R and R is as defined herein. Quinoxalinone 13.1 can be alkylated with an alkylating agent, such as Mel, EtI, allyl bromide or cyclopropylmethyl methanesulfonate, utilizing a base, such as NaH, in a solvent, such as DMF, giving a mixture of heterocycles 13.2 and 13.3. Heterocycles 13.2 and 13.3 can be separated and coupled separately in the Buchwald reaction according to Scheme 11.
Figure imgf000033_0001
14.1 14.2 14.3
Scheme 14
[00148] Scheme 14 describes the preparation of nitriles 14.3, wherein W, X, R1, R3, R4 and R11 are as defined herein. An alkali metal salt of a hydroxyl-containing heterocycle 14.1 is generated using a suitable base, such as sodium hydride, in an appropriate solvent, such as THF, DMF or NMP. The resulting phenoxide is treated with an optionally substituted difluorobenzonitrile at ambient or elevated temperature to afford ether intermediate 14.2. Nitrile 14.3 is obtained by reacting, at elevated temperature, intermediate 14.2 with an anion of an optionally protected sulfonamide or sulfamide, formed by the action of a strong base, such as sodium hydride, in a solvent, such as NMP, DMF, or DMSO.
Figure imgf000033_0002
Scheme 15
[00149] Scheme 15 describes the preparation of nitriles 15.3, wherein W, X, R1, R3, R4 and R11 are as defined herein. An aniline is coupled to a heterocycle 15.1 under standard Buchwald coupling conditions utilizing a Pd catalyst, such as Pd(dba)2, and a ligand, such as ΒΓΝΑΡ, to give biarylamine 15.2. Nitrile 15.3 is obtained by reacting, at elevated temperature, intermediate 15.2 with an anion of an optionally protected sulfonamide or sulfamide, formed by the action of a strong base, such as sodium hydride, in a solvent, such as NMP DMF, or DMSO.
Figure imgf000033_0003
Scheme 16
[00150] Scheme 16 shows a general method for preparing compounds 16.2 and 16.3, wherein R6, Rm and Rn are as defined herein. A quinazolinone such as 16.1 can be treated with a coupling agent, such as BOP, in a suitable solvent, such as DMF or acetonitrile, and subsequently treated with an amine to provide quinazoline amines 16.3. Alternatively, 16.1 can be treated with the coupling agent FLATU in a suitable solvent, such as DMF or acetonitrile, and subsequently treated with an amine to provide quinazolines 16.2.
Figure imgf000034_0001
Scheme 17
[00151] Scheme 17 shows a general method for preparing sulfamides 17.2, wherein W, X, R1, R2, R3, R4, R11, Ra and Rb are as defined herein. A sulfonamide 17.1 is treated with a sulfamoyl chloride in a solvent, such as DMF, and subsequently hydrolyzed to a sulfamide 17.2 by addition of a base and water, such as sodium h droxide.
Figure imgf000034_0002
Scheme 18
[00152] Scheme 18 shows a general method for preparing compounds 18.2, wherein Rz is Ci-C6 alkyl optionally substituted with halogen, oxo, ORJ, SRj, NRkR', phenyl, d-C4 alkyl, C!-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp. 4-Chloroquinazolinone 18.1 can be treated with a nucleophile, such as NaORz, in a suitable solvent, such as RzOH, to provide 4-alkoxyquinazole 18.2.
[00153] METHODS OF SEPARATION
[00154] It may be advantageous to separate reaction products from one another and/or from starting materials. The desired products of each step or series of steps is separated and/or purified (hereinafter separated) to the desired degree of homogeneity by the techniques common in the art. Typically such separations involve multiphase extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography can involve any number of methods including, for example: reverse-phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small scale analytical; simulated moving bed (SMB) and preparative thin or thick layer chromatography, as well as techniques of small scale thin layer and flash chromatography. One skilled in the art will apply techniques most likely to achieve the desired separation.
[00155] Diastereomeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diastereomeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereoisomers to the corresponding pure enantiomers. Enantiomers can also be separated by use of a chiral HPLC column.
[00156] A single stereoisomer, e.g., an enantiomer, substantially free of its stereoisomer may be obtained by resolution of the racemic mixture using a method such as formation of diastereomers using optically active resolving agents (Eliel, E. and Wilen, S. Stereochemistry of Organic Compounds. New York: John Wiley & Sons, Inc., 1994; Lochmuller, C. H., et al. "Chromatographic resolution of enantiomers: Selective review." J. Chromatogr., 113(3) (1975): pp. 283-302). Racemic mixtures of chiral compounds described herein may be separated and isolated by any suitable method, including: (1) formation of ionic, diastereomeric salts with chiral compounds and separation by fractional crystallization or other methods, (2) formation of diastereomeric compounds with chiral derivatizing reagents, separation of the diastereomers, and conversion to the pure stereoisomers, and (3) separation of the substantially pure or enriched stereoisomers directly under chiral conditions. See: Wainer, Irving W., Ed. Drug Stereochemistry: Analytical Methods and Pharmacology. New York: Marcel Dekker, Inc., 1993.
[00157] Under method (1), diastereomeric salts can be formed by reaction of enantiomerically pure chiral bases such as brucine, quinine, ephedrine, strychnine, a-methyl- β-phenylethylamine (amphetamine), and the like with asymmetric compounds bearing acidic functionality, such as carboxylic acid and sulfonic acid. The diastereomeric salts may be induced to separate by fractional crystallization or ionic chromatography. For separation of the optical isomers of amino compounds, addition of chiral carboxylic or sulfonic acids, such as camphorsulfonic acid, tartaric acid, mandelic acid, or lactic acid, can result in formation of the diastereomeric salts.
[00158] Alternatively, by method (2), the substrate to be resolved is reacted with one enantiomer of a chiral compound to form a diastereomeric pair (Eliel, E. and Wilen, S. Stereochemistry of Organic Compounds. New York: John Wiley & Sons, Inc., 1994, p. 322). Diastereomeric compounds can be formed by reacting asymmetric compounds with enantiomerically pure chiral derivatizing reagents, such as menthyl derivatives, followed by separation of the diastereomers and hydrolysis to yield the pure or enriched enantiomer. A method of determining optical purity involves making chiral esters, such as a menthyl ester, e.g., (-) menthyl chloroformate in the presence of base, or Mosher ester, a-methoxy-a- (trifluoromethyl)phenyl acetate (Jacob III, Peyton. "Resolution of (±)-5-Bromonornicotine. Synthesis of (R)- and (S)-Nornicotine of High Enantiomeric Purity." J. Org. Chem. Vol. 47, No. 21 (1982): pp. 4165-4167), of the racemic mixture, and analyzing the 1H NMR spectrum for the presence of the two atropisomeric enantiomers or diastereomers. Stable diastereomers of atropisomeric compounds can be separated and isolated by normal- and reverse-phase chromatography following methods for separation of atropisomeric naphthyl-isoquinolines (WO 96/15111).
[00159] By method (3), a racemic mixture of two enantiomers can be separated by chromatography using a chiral stationary phase (Lough, W.J., Ed. Chiral Liquid Chromatography. New York: Chapman and Hall, 1989; Okamoto, Yoshio, et al. "Optical resolution of dihydropyridine enantiomers by high-performance liquid chromatography using phenylcarbamates of polysaccharides as a chiral stationary phase." J. of Chromatogr. Vol. 513 (1990): pp. 375-378). Enriched or purified enantiomers can be distinguished by methods used to distinguish other chiral molecules with asymmetric carbon atoms, such as optical rotation and circular dichroism.
[00160] BIOLOGICAL EVALUATION
[00161] B-Raf mutant protein 447-717 (V600E) was co-expressed with the chaperone protein Cdc37, complexed with Hsp90 (Roe, S. Mark, et al. "The Mechanism of Hsp90 Regulation by the Protein Kinase-Specific Cochaperone p50cdc37." Cell. Vol. 116 (2004): pp. 87-98; Stancato, LF, et al. "Raf exists in a native heterocomplex with Hsp90 and p50 that can be reconstituted in a cell free system." J. Biol. Chem. 268(29) (1993): pp. 21711-21716).
[00162] Determining the activity of Raf in the sample is possible by a number of direct and indirect detection methods (US 2004/0082014). Activity of human recombinant B-Raf protein may be assessed in vitro by assay of the incorporation of radio labeled phosphate to recombinant MAP kinase (MEK), a known physiologic substrate of B-Raf, according to US 2004/0127496 and WO 03/022840. The activity/inhibition of V600E full-length B-Raf was estimated by measuring the incorporation of radio labeled phosphate from [γ-33Ρ]ΑΤΡ into FSBA-modified wild-type MEK (see Example A).
[00163] ADMINISTRATION AND PHARMACEUTICAL FORMULATIONS
[00164] The compounds described herein may be administered by any convenient route appropriate to the condition to be treated. Suitable routes include oral, parenteral (including subcutaneous, intramuscular, intravenous, intraarterial, intradermal, intrathecal and epidural), transdermal, rectal, nasal, topical (including buccal and sublingual), vaginal, intraperitoneal, intrapulmonary and intranasal.
[00165] The compounds may be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches, etc. Such compositions may contain components conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents. If parenteral administration is desired, the compositions will be sterile and in a solution or suspension form suitable for injection or infusion.
[00166] A typical formulation is prepared by mixing a compound described herein and a carrier or excipient. Suitable carriers and excipients are well known to those skilled in the art and are described in detail in, e.g., Ansel, Howard C, et al., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems. Philadelphia: Lippincott, Williams & Wilkins, 2004; Gennaro, Alfonso R., et al. Remington: The Science and Practice of Pharmacy. Philadelphia: Lippincott, Williams & Wilkins, 2000; and Rowe, Raymond C. Handbook of Pharmaceutical Excipients. Chicago, Pharmaceutical Press, 2005. The formulations may also include one or more buffers, stabilizing agents, surfactants, wetting agents, lubricating agents, emulsifiers, suspending agents, preservatives, antioxidants, opaquing agents, glidants, processing aids, colorants, sweeteners, perfuming agents, flavoring agents, diluents and other known additives to provide an elegant presentation of the drug (i.e., a compound described herein or pharmaceutical composition thereof) or aid in the manufacturing of the pharmaceutical product (i.e., medicament).
[00167] One embodiment includes a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer or pharmaceutically acceptable salt thereof. A further embodiment provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer or pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier or excipient. [00168] Another embodiment of the present invention provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of a hyperproliferative disease.
[00169] Another embodiment of the present invention provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of cancer.
[00170] Another embodiment of the present invention provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of kidney disease. A further embodiment of the present invention provides a pharmaceutical composition comprising a compound of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of polycystic kidney disease.
[00171] METHODS OF TREATMENT WITH COMPOUNDS OF THE INVENTION
[00172] The invention includes methods of treating or preventing disease or condition by administering one or more compounds of this invention, or a stereoisomer or pharmaceutically acceptable salt thereof. In one embodiment, a human patient is treated with a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle in an amount to detectably inhibit B-Raf activity.
[00173] In another embodiment, a human patient is treated with a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle in an amount to detectably inhibit B-Raf activity.
[00174] In another embodiment of the present invention, a method of treating a hyperproliferative disease in a mammal comprising administering a therapeutically effective amount of the compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, to the mammal is provided.
[00175] In another embodiment of the present invention, a method of treating kidney disease in a mammal comprising administering a therapeutically effective amount of the compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, to the mammal is provided. In a further embodiment, the kidney disease is polycystic kidney disease.
[00176] In another embodiment, a method of treating or preventing cancer in a mammal in need of such treatment, wherein the method comprises administering to said mammal a therapeutically effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof. The cancer is selected from breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, NSCLC, small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system, Hodgkin's and leukemia. Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
[00177] In another embodiment, a method of treating or preventing cancer in a mammal in need of such treatment, wherein the method comprises administering to said mammal a therapeutically effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof.
[00178] Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
[00179] Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of kidney disease. In a further embodiment, the kidney disease is polycystic kidney disease.
[00180] In another embodiment, a method of preventing or treating cancer, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds having anti-cancer properties.
[00181] In another embodiment, a method of preventing or treating cancer, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds having anti-cancer properties.
[00182] In one further embodiment, the cancer is a sarcoma. [00183] In another further embodiment, the cancer is a carcinoma. In one further embodiment, the carcinoma is squamous cell carcinoma. In another further embodiment, the carcinoma is an adenoma or adenocarcinoma.
[00184] In another embodiment, a method of treating or preventing a disease or disorder modulated by B-Raf, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof. Examples of such diseases and disorders include, but are not limited to, cancer. The cancer is selected from breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, NSCLC, small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system, Hodgkin's and leukemia.
[00185] In another embodiment, a method of treating or preventing a disease or disorder modulated by B-Raf, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof.
[00186] In another embodiment of the present invention, a method of preventing or treating kidney disease, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds. In another embodiment of the present invention, a method of preventing or treating polycystic kidney disease, comprising administering to a mammal in need of such treatment an effective amount of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, alone or in combination with one or more additional compounds.
[00187] Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer. The cancer is selected from breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, NSCLC, small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system, Hodgkin's and leukemia. In a further embodiment, the use of a compound of Formulas I, II, III, IV, V, VI, or VII in the manufacture of a medicament, for use as a B-Raf inhibitor in the treatment of a patient undergoing cancer therapy.
[00188] Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
[00189] Another embodiment of the present invention provides the use of a compound of Formulas I, II, III, IV, V, VI, or VII, or a stereoisomer, tautomer, or pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of polycystic kidney disease. In a further embodiment, the kidney disease is polycystic kidney disease.
[00190] Another embodiment of the present invention provides the compounds of
Formulas I, II, III, IV, V, VI, or VII for use in therapy.
[00191] Another embodiment of the present invention provides the compounds of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of a hyperproliferative disease. In a further embodiment, the hyperproliferative disease is cancer (as further defined and may be individually selected from those above).
[00192] Another embodiment of the present invention provides the compounds of Formulas I, II, III, IV, V, VI, or VII for use in the treatment of kidney disease. In a further embodiment, the kidney disease is polycystic kidney disease.
[00193] COMBINATION THERAPY
[00194] The compounds described herein and stereoisomers and pharmaceutically acceptable salts thereof may be employed alone or in combination with other therapeutic agents for treatment. The compounds described herein may be used in combination with one or more additional drugs, for example an anti-hyperproliferative (or anti-cancer) agent that works through action on a different target protein. The second compound of the pharmaceutical combination formulation or dosing regimen preferably has complementary activities to the compound described herein, such that they do not adversely affect each other. Such molecules are suitably present in combination in amounts that are effective for the purpose intended. The compounds may be administered together in a unitary pharmaceutical composition or separately and, when administered separately this may occur simultaneously or sequentially in any order. Such sequential administration may be close in time or remote in time.
[00195] A "chemotherapeutic agent" is a chemical compound useful in the treatment of cancer, regardless of mechanism of action. Chemotherapeutic agents include compounds used in "targeted therapy" and conventional chemotherapy. A number of suitable chemotherapeutic agents to be used as combination therapeutics are contemplated for use in the methods of the present invention. The present invention contemplates, but is not limited to, administration of numerous anticancer agents, such as: agents that induce apoptosis; polynucleotides (e.g., ribozymes); polypeptides (e.g., enzymes); drugs; biological mimetics; alkaloids; alkylating agents^antitumor antibiotics; antimetabolites; hormones; platinum compounds; monoclonal antibodies conjugated with anticancer drugs, toxins, and/or radionuclides; biological response modifiers (e.g., interferons [e.g., IFN-a, etc.] and interleukins [e.g., IL-2, etc.], etc.); adoptive immunotherapy agents; hematopoietic growth factors; agents that induce tumor cell differentiation (e.g., all-trans-retinoic acid, etc.); gene therapy reagents; antisense therapy reagents and nucleotides; tumor vaccines; inhibitors of angiogenesis, and the like.
[00196] Examples of chemotherapeutic agents include Erlotinib (TARCEVA®, Genentech/OSI Pharm.), Bortezomib (VELCADE®, Millennium Pharm.), Fulvestrant (FASLODEX®, AstraZeneca), Sunitinib (SUTENT®, Pfizer), Letrozole (FEMARA®, Novartis), Imatinib mesylate (GLEEVEC®, Novartis), PTK787/ZK 222584 (Novartis), Oxaliplatin (Eloxatin®, Sanofi), 5-FU (5-fluorouracil), Leucovorin, Rapamycin (Sirolimus, RAPAMUNE®, Wyeth), Lapatinib (TYKERB®, GSK572016, Glaxo Smith Kline), Lonafarnib (SCH 66336), Sorafenib (NEXAVAR®, Bayer), Irinotecan (CAMPTOSAR®, Pfizer) and Gefitinib (IRESSA®, AstraZeneca), AG1478, AG1571 (SU 5271; Sugen), alkylating agents such as thiotepa and CYTOXAN® cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide and trimethylomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analog topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogs); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogs, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, chlorophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammall and calicheamicin omegall (Angew Chem. Intl. Ed. Engl. (1994) 33:183-186); 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, carminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6- diazo-5-oxo-L-norleucine, ADRIAMYCIN® (doxorubicin), morpholino-doxorubicin, cyanomo holino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, porfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6- mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfornithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK® polysaccharide complex (JHS Natural Products, Eugene, OR); razoxane; rhizoxin; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2',2"-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside ("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g., TAXOL® (paclitaxel; Bristol-Myers Squibb Oncology, Princeton, N.J.), ABRAXANE™ (Cremophor-free), albumin-engineered nanoparticle formulations of paclitaxel (American Pharmaceutical Partners, Schaumberg, Illinois), and TAXOTERE® (doxetaxel; Rhone-Poulenc Rorer, Antony, France); chloranmbucil; 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; difluoromethylornithine (DMFO); retinoids such as retinoic acid; and pharmaceutically acceptable salts, acids and derivatives of any of the above.
[00197] Also included in the definition of "chemotherapeutic agent" are: (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, 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, RTVISOR® (vorozole), FEMARA® (letrozole; Novartis), and ARIMIDEX® (anastrozole; AstraZeneca); (iii) anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; as well as troxacitabine (a 1,3-dioxolane nucleoside cytosine analog); (iv) protein kinase inhibitors; (v) lipid kinase inhibitors; (vi) antisense oligonucleotides, particularly those which inhibit expression of genes in signaling pathways implicated in aberrant cell proliferation, such as, for example, PKC-alpha, Raf and H-Ras; (vii) ribozymes such as VEGF expression inhibitors (e.g., ANGIOZYME®) and HER2 expression inhibitors; (viii) vaccines such as gene therapy vaccines, for example, ALLOVECTIN®, LEUVECTIN®, and VAXID®; PROLEUKIN® rIL-2; a topoisomerase 1 inhibitor such as LURTOTECAN®; ABARELIX® rmRH; (ix) anti-angiogenic agents such as bevacizumab (AVASTIN®, Genentech); (x) PI3k/AKT/mTOR pathway inhibitors, including GDC-0941 (2-( 1 H-Indazol-4-yl)-6-(4-methanesulfonyl-piperazin- 1 -ylmethyl)-4- morpholin-4-yl-thieno[3,2-d]pyrimidine), XL-147, GSK690693 and temsirolimus; (xi) Ras/Raf MEK/ERK pathway inhibitors; and (xii) pharmaceutically acceptable salts, acids and derivatives of any of the above.
[00198] Also included in the definition of "chemotherapeutic agent" are therapeutic antibodies such as alemtuzumab (Campath), bevacizumab (AVASTIN®, Genentech); cetuximab (ERBITUX®, Imclone); panitumumab (VECTIBIX®, Amgen), rituximab (RITUXAN®, Genentech/Biogen Idee), pertuzumab (OMNITARG®, 2C4, Genentech), trastuzumab (HERCEPTIN®, Genentech), tositumomab (Bexxar, Corixia), and the antibody drug conjugate, gemtuzumab ozogamicin (MYLOTARG®, Wyeth).
[00199] Humanized monoclonal antibodies with therapeutic potential as chemotherapeutic agents in combination with the Raf inhibitors of the invention include: alemtuzumab, apolizumab, aselizumab, atlizumab, bapineuzumab, bevacizumab, 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, nolovizumab, numavizumab, ocrelizumab, omalizumab, palivizumab, pascolizumab, pecfusituzumab, pectuzumab, pertuzumab, pexelizumab, ralivizumab, ranibizumab, reslivizumab, reslizumab, resyvizumab, rovelizumab, ruplizumab, sibrotuzumab, siplizumab, sontuzumab, tacatuzumab tetraxetan, tadocizumab, talizumab, tefibazumab, tocilizumab, toralizumab, trastuzumab, tucotuzumab celmoleukin, tucusituzumab, umavizumab, urtoxazumab, and visilizumab.
EXAMPLES
[00200] For illustrative purposes, the following Examples are included. However, it is to be understood that these Examples do not limit the invention and are only meant to suggest a method of practicing the invention. Persons skilled in the art will recognize that the chemical reactions described may be readily adapted to prepare a number of other compounds described herein, and alternative methods for preparing the compounds are deemed to be within the scope of this invention. For example, the synthesis of non- exemplified compounds may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds described herein.
[00201] In the Examples described below, unless otherwise indicated all temperatures are set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Sigma-Aldrich, Alfa Aesar, or TCI, and were used without further purification unless otherwise indicated. [00202] The reactions set forth below were done generally under a positive pressure of nitrogen or argon or with a drying tube (unless otherwise stated) in anhydrous solvents, and the reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried.
[00203] Column chromatography was done on a Biotage system (Manufacturer: Dyax Corporation) having a silica gel column or on a silica SepPak cartridge (Waters) (unless otherwise stated). 1H NMR spectra were recorded on a Varian instrument operating at 400 MHz. 1H-NMR spectra were obtained as CDC13, CD3OD, D20, (CD3)2SO, (CD3)2CO, C6D6, CD3CN solutions (reported in ppm), using tetramethylsilane (0.00 ppm) or residual solvent (CDC13: 7.26 ppm; CD3OD: 3.31 ppm; D20: 4.79 ppm; (CD3)2SO: 2.50 ppm; (CD3)2CO: 2.05 ppm; C6D6: 7.16 ppm; CD3CN: 1.94 ppm) as the reference standard. When peak multiplicities are reported, the following abbreviations are used: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broadened), dd (doublet of doublets), dt (doublet of triplets). Coupling constants, when given, are reported in Hertz (Hz).
Example A
B-Raf ICsn Assay Protocol
[00204] Activity of human recombinant B-Raf protein may be assessed in vitro by assay of the incorporation of radio labeled phosphate to recombinant MAP kinase (MEK), a known physiologic substrate of B-Raf, according to US 2004/0127496 and WO 03/022840. Catalytically active human recombinant B-Raf protein is obtained by purification from sf9 insect cells infected with a human B-Raf recombinant baculovirus expression vector.
[00205] The activity/inhibition of V600E full-length B-Raf was estimated by measuring the incorporation of radio labeled phosphate from [γ-33Ρ] ATP into FSBA-modified wild-type MEK. The 30-μΙ_< assay mixtures contained 25mM Na Pipes, pH 7.2, lOOmM KC1, lOmM MgCl2, 5mM β-glycerophosphate, ΙΟΟμΜ Na Vanadate, 4μΜ ATP, 500 nCi [γ- 33P]ATP, Ι μΜ FSBA-MEK and 20nM V600E full-length B-Raf. Incubations were carried out at 22°C in a Costar 3365 plate (Corning). Prior to the assay, the B-Raf and FSBA-MEK were preincubated together in assay buffer at 1.5x (20 μΐ^ of 30nM and 1.5μΜ, respectively) for 15 minutes, and the assay was initiated by the addition of 10 μΐ, of 10μΜ ATP. Following the 60-minute incubation, the assay mixtures were quenched by the addition of 100 \iL of 25% TCA, the plate was mixed on a rotary shaker for 1 minute, and the product was captured on a Perkin-Elmer GF/B filter plate using a Tomtec Mach III Harvester. After sealing the bottom of the plate, 35 μί^ of Bio-Safe II (Research Products International) scintillation cocktail were added to each well and the plate was top-sealed and counted in a Topcount NXT (Packard).
[00206] The compounds of Examples 1-49 were tested in the above assay and found to have an IC50 of less than 900 nM. The compounds of Examples 1-14, 17-29, 31-43 and 45- 49 were tested in the above assay and found to have an IC50 of less than 100 nM.
Example B
Figure imgf000047_0001
Methyl 2,6-difluoro-3-(N-(propylsulfonyl)propylsulfonamido')benzoate
[00207] Step A: A 1 L flask was charged with 2,6-difluoro-3-nitrobenzoic acid (17.0 g, 83.7 mmol) and MeOH (170 mL, 0.5M). The flask was placed in a cold water bath, and an addition funnel charged with a 2M solution of trimethylsilyl ("TMS") diazomethane in hexanes (209 mL, 419 mmol) was attached to the flask. The TMS diazomethane solution was added slowly to the reaction flask over the course of 2 hours. A large excess of reagent was required in order for the reaction to reach completion as determined by the ceased evolution of N2 upon further addition of reagent. The volatiles were removed in vacuo to afford crude methyl 2,6-difluoro-3-nitrobenzoate as a solid (18.2 g). The material was taken directly to Step B.
[00208] Step B: 10% (wt.) Pd on activated carbon (4.46 g, 4.19 mmol) was added to a 1 L flask charged with methyl 2,6-difluoro-3-nitrobenzoate (18.2 g, 83.8 mmol) under a nitrogen atmosphere. To the flask was added EtOH (350 mL, 0.25 M), and H2 gas was passed through the mixture for 15 minutes. The reaction mixture was stirred under two H2 balloons overnight. The balloons were recharged with H2 gas and the mixture was stirred an additional 4 hours. Upon consumption of the starting material and intermediate hydroxylamine as determined by TLC, N2 gas was flushed through the reaction mixture. The mixture was then filtered through glass microfibre filter ("GF/F") paper twice. The volatiles were removed to afford crude methyl 3-amino-2,6-difluorobenzoate as an oil (15.66 g). The material was taken directly onto the next step.
[00209] Step C: Propane- 1-sulfonyl chloride (23.46 mL, 209.3 mmol) was slowly added to a solution of methyl 3-amino-2,6-difluorobenzoate (15.66 g, 83.7 mmol) and triethylamine (35.00 mL, 251.1 mmol) in CH2C12 (175 mL, 0.5M) maintained in a cool water bath. The reaction mixture was stirred for 1 hour at room temperature. Water (300 mL) was added and the organic layer was separated, washed with water (2 X 300 mL) and brine (200 mL), then dried (Na2S04), filtered and concentrated to an oil. The crude product was purified by column chromatography, eluting with 15% ethyl acetate ("EtOAc")/hexane. The isolated fractions were triturated with hexanes to afford methyl 2,6-difluoro-3-(N- (propylsulfonyl)propylsulfonamido)benzoate as a solid (24.4 g, 73% yield for 3 steps). 1H NMR (400 MHz, CDC13) δ 7.52-7.45 (m, 1H), 7.08-7.02 (m, 1H), 3.97 (s, 3H), 3.68-3.59 (m, 2H), 3.53-3.45 (m, 2H), 2.02-1.89 (m, 4H), 1.10 (t, J = 7.4 Hz, 6H). m/z (APCI-neg) M- (S02Pr) = 292.2.
Example C
Figure imgf000048_0001
2,6-Difluoro-3-(propylsulfonamido)benzoic acid
[00210] A IN aqueous NaOH solution (150 mL, 150 mmol) was added to a solution of methyl 2,6-difluoro-3-(N-(propylsulfonyl)propylsulfonamido)benzoate (20.0 g, 50.1 mmol) in 4:1 THF/MeOH (250 mL, 0.2M). The reaction mixture was stirred at room temperature overnight. The majority of the organic solvents were removed in vacuo (water bath temperature 35°C). IN HC1 (150 mL) was slowly added to the mixture, and the resulting solid was filtered and rinsed with water (4 X 50 mL). The material was washed with Et20 (4 X 15 mL) to give 2,6-difluoro-3-(propylsulfonamido)benzoic acid as a solid (10.7 g, 77% yield). 1H NMR (400 MHz, (CD3)2SO) δ 9.74 (s, 1H), 7.57-7.50 (m, 1H), 7.23-7.17 (m, 1H), 3.11-3.06 (m, 2H), 1.79-1.69 (m, 2H), 0.98 (t, J - 7.4 Hz, 3H). m/z (APCI-neg) M-l = 278.0.
Example D
Figure imgf000048_0002
2,6-Difluoro-3-(N-(propylsulfonyl)propylsulfonamido)benzoic acid
[00211] Propane- 1-sulfonyl chloride (1.225 mL, 10.92 mmol) was added to a mixture of 3-amino-2,6-difluorobenzoic acid (0.573 g, 3.310 mmol), triethylamine (2.030 mL, 14.56 mmol) and CH2C12 (17 mL, 0.2M) cooled to 0°C. The reaction mixture was allowed to warm to room temperature and stirred for 1 hour. The mixture was then partitioned between saturated NaHC03 (100 mL) and ethyl acetate (75 mL). The aqueous layer was washed with ethyl acetate (50 mL) and then acidified with concentrated HC1 to a pH of about 1. The acidified aqueous layer was extracted with ethyl acetate (2 x 50 mL), and the combined ethyl acetate extracts were dried (over Na2S04), filtered and concentrated. The resulting residue was triturated with hexanes to afford 2,6-difluoro-3-(N-(propylsulfonyl)propyl- sulfonamido)benzoic acid as a solid (0.948 g, 74% yield). 1H NMR (400 MHz, (CD3)2SO) δ 7.90-7.84 (m, 1H), 7.39-7.34 (m, 1H), 3.73-3.58 (m, 4H), 1.88-1.74 (m, 4H), 1.01 (t, J = 7.5 Hz, 6H). m/z (APCI-neg) M-(S02Pr) = 278.1.
Example E
Figure imgf000049_0001
2-Chloro-6-fluoro-3 -(propylsulfonamido)benzoic acid
[00212] Step A: Into a 20-L 4-neck round flask was placed a solution of 2-chloro-4- fluorobenzenamine (1300 g, 8.82 mol, 1.00 equiv, 99%) in toluene (10 L), 4- methylbenzenesulfonic acid (3.1 g, 17.84 mmol, 99%), and hexane-2,5-dione (1222.5 g, 10.62 mol, 1.20 equiv, 99%). The resulting solution was heated to reflux for 1 hour in an oil bath and cooled. The pH value of the solution was adjusted to 8 with sodium carbonate (1 mol/L). The resulting mixture was washed with water (5000 mL) and concentrated under vacuum. The crude product was purified by distillation and the fraction was collected at 140°C to afford l-(2-chloro-4-fluorophenyl)-2,5-dimethyl-lH-pyrrole as a solid (1700 g, yield: 85%).
[00213] Step B: Into a 5000-mL 4-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen was placed a solution of l-(2-chloro-4-fluorophenyl)- 2,5-dimethyl-lH-pyrrole (390 g, 1.65 mol, 1.00 equiv, 95%) in tetrahydrofuran (2000 mL). The reaction vessel was cooled to -78°C. n-BuLi (800 mL, 1.10 equiv, 2.5%) was added dropwise with stirring to the reaction vessel over 80 minutes, and methyl carbonochloridate (215.5 g, 2.27 mol, 1.20 equiv, 99%) was added dropwise with stirring over 90 minutes. The reaction solution was further stirred for 60 minutes at -78°C and quenched by the addition of NH4Cl/water (1000 mL). The resulting solution was extracted with ethyl acetate (1500 mL). The organic layers were combined, washed with water (1500 mL) and aqueous sodium chloride (1500 mL), dried over anhydrous magnesium sulfate, and concentrated under vacuum to afford methyl 2-chloro-3-(2,5-dimethyl-lH-pyrrol-l-yl)-6-fluorobenzoate as an oil (crude, 566.7 g).
[00214] Step C: A solution of methyl 2-chloro-3-(2,5-dimethyl-lH-pyrrol-l-yl)-6- fluorobenzoate (1500 g, 5.05 mol, 1.00 equiv, 95%) in ethanol/H20 (7500/2500 mL), NH2OH-HCl (5520 g, 79.20 mol, 15.00 equiv, 99%), and triethylamine (2140 g, 20.98 mol, 4.00 equiv, 99%) was placed into five 5000-mL 4-neck round-bottom flasks. The resulting solution was refluxed for 18 hours in an oil bath, cooled to room temperature, concentrated, and extracted with ethyl acetate (3 X 3000 mL). The organic layers were combined, dried over anhydrous sodium sulfate, and concentrated under vacuum. The residue was purified using a silica gel column eluting with PE:EA (20:1-10:1) to afford methyl 3-amino-2-chloro- 6-fluorobenzoate as an oil (980 g, yield: 95%).
[00215] Step D: A solution of methyl 3-amino-2-chloro-6-fluorobenzoate (980 g, 4.76 mol, 1.00 equiv, 99%) in dichloromethane (8000 mL) was placed into four 5000-mL 4-neck round-bottom flasks. Triethylamine (1454 g, 14.25 mol, 3.00 equiv, 99%) was added dropwise to this reaction vessel with stirring at 0°C over 80 minutes followed by the addition of propane- 1-sulfonyl chloride (1725 g, 11.94 mol, 2.50 equiv, 99%). The resulting solution was stirred at room temperature for 2 hours, diluted with water (1000 mL). The organic layer was washed with hydrogen chloride (1000 mL) and water (1000 mL), dried over sodium sulfate, and concentrated to afford methyl 2-chloro-6-fluoro-3-(propylsulfonamido)benzoate as a solid (1500 g, 97%).
[00216] Step E: A solution of methyl 2-chloro-6-fluoro-3-
(propylsulfonamido)benzoate (1500 g, 4.61 mol, 1.00 equiv, 95%) in tetrahydrofuran/H20 (3000/3000 mL) and potassium hydroxide (1000 g, 17.68 mol, 4.50 equiv, 99%) was placed into a 10000-mL 4-necked round-bottom flask. The resulting solution was refluxed for 2 hours, cooled to room temperature and extracted with ethyl acetate (3 X 2000 mL). The aqueous layers were combined, and the pH was adjusted to 2 with hydrogen chloride (2 mol/L). The resulting solution was extracted with dichloromethane (2 X 3000 mL). The organic layers were combined, dried over anhydrous sodium sulfate and concentrated to afford 2-chloro-6-fluoro-3-(propylsulfonamido)benzoic acid as a solid (517.5 g, yield: 37%). (ES, m/z): [M+H]+ 296. 1H NMR (400 MHz, CDC13): δ 1.058-1.096 (3H, m, J = 15.2Hz), 1.856-1.933 (2H, m, J = 30.8 Hz), 3.073-3.112 (2H, m, J = 15.6 Hz); 6.811 (1H, s), 7.156- 7.199 (1H, d, J= 17.2 Hz), 7.827-7.863 (1H, d, J= 14.4Hz).
Example F
Figure imgf000051_0001
6-Chloro-2-fluoro-3 -('propylsulfonarnido)benzoic acid
[00217] Step A: A flame dried flask equipped with a stir bar and rubber septum was charged with 4-chloro-2-fluoroaniline (5.00 g, 34.35 mmol) and anhydrous THF (170 mL). This solution was chilled to -78°C, and n-BuLi (14.7 mL, 1.07 eq. of 2.5M solution in hexanes) was then added over a 15 minute period. This mixture was stirred at -78°C for 20 minutes, and then a THF solution (25 mL) of l,2-bis(chlorodimethylsilyl)ethane (7.76 g, 1.05 eq.) was added slowly (over a 10 minute period) to the reaction mixture. This was stirred for 1 hour, and then 2.5M n-BuLi in hexanes (15.11 mL, 1.1 eq.) was added slowly. After allowing the mixture to warm to room temperature for one hour, the mixture was chilled back to -78°C. A third allotment of n-BuLi (15.66 mL, 1.14 eq.) was added slowly, and the mixture was stirred at -78°C for 75 minutes. Benzyl chloroformate (7.40 g, 1.2 eq.) was then added slowly, and the mixture was stirred at -78°C for one hour. The cooling bath was then removed. The mixture was allowed to warm for 30 minutes and then quenched with water (70 mL) and concentrated HC1 (25 mL). The mixture was allowed to continue to warm to room temperature. The mixture was then extracted with EtOAc. The extracts were washed twice with a saturated Na2HC03 solution, once with water, dried over sodium sulfate and concentrated. The resulting residue was flashed on a 65 Biotage (30% ethyl acetate/hexane) to produce benzyl 3-amino-6-chloro-2-fluorobenzoate (4.3 g, 45%) as an oil.
1H NMR ((CD3)2SO, 400 MHz) δ 7.37-7.48 (m, 5H), 7.07 (dd, 1H, J = 8, 2), 6.87 (t, 1H, J = 8), 5.61 (br s, 2H), 5.40 (s, 2H).
[00218] Step B: Benzyl 3-amino-6-chloro-2-fluorobenzoate (4.3 g, 15.37 mmol) was dissolved in dry dichloromethane (270 mL). Triethylamine (5.36 mL, 2.5 eq.) was added, and the mixture was chilled to 0°C. Propane- 1-sulfonyl chloride (3.63 mL, 32.3 mmol, 2.1 eq.) was then added via syringe, and a precipitate resulted. Once the addition was complete, the mixture was allowed to warm to room temperature, and the starting material was consumed as determined by TLC (3:1 hexanerethyl acetate). The mixture was then diluted with dichloromethane (200 mL), washed with 2M aqueous HC1 (2 X 100 mL), saturated Na2HC03 solution, dried over sodium sulfate and concentrated. The resulting residue was purified on a 65 Biotage chromatography system (40% ethyl acetate/hexane) to produce benzyl 6-cliloro-2-fluoro-3-(N-(propylsulfonyl)propylsulfonamido)benzoate (5.5 g, 72%») as an oil that slowly solidified upon standing. NMR (CDC13, 400 MHz) δ 7.28-7.45 (m, 7H), 5.42 (s, 2H), 3.58-3.66 (m, 2H), 3.43-3.52 (m, 2H), 1.08 (t, 6H, J=8).
[00219] Step C: Benzyl 6-chloro-2-fluoro-3-(N-(propylsulfonyl)propylsulfonamido) benzoate (5.4 g, 10.98 mmol) was dissolved in THF (100 mL) and 1M aqueous KOH (100 mL). This mixture was refluxed for 16 hours and then allowed to cool to room temperature. The mixture was then acidified to a pH of 2 with 2M aqueous HCl and extracted with EtOAc (2 X). The extracts were washed with water, dried over sodium sulfate and concentrated to a solid that was triturated with hexanes/ether to give 6-chloro-2-fluoro-3- (propylsulfonamido)benzoic acid (2.2 g, 68%) as a solid. 1H NMR ((CD3)2SO, 400 MHz) δ 9.93 (s, 1H), 7.49 (t, 1H, J=8), 7.38 (dd, 1H, J = 8, 2), 3.11-3.16 (m, 2H), 1.68-1.78 (m, 2H), 0.97 (t, 3H, J = 8).
Exam le G
Figure imgf000052_0001
N-(3 - Amino-2,4-difluorophenyl)propane- 1 -sulfonamide
[00220] Triethylamine (4.68 mL, 33.59 mmol) and diphenylphosphonic azide (3.73 mL, 16.79 mmol) were added to a solution of 2,6-difluoro-3-(propylsulfonamido)benzoic acid (4.078 g, 14.6 mmol) in THF (60 mL). The reaction mixture was stirred at room temperature for 3 hours and then warmed to 80°C for 2 hours. Water (10 mL) was added, and the mixture was stirred at 80°C for 15 hours. The reaction mixture was diluted with EtOAc (300 mL), and the organic layer was washed with saturated aqueous NaHC03 solution and brine. The solvent was removed under reduced pressure, and the residual purified via silica gel column chromatography eluting with 30/70 EtOAc/hexane to obtain 2.03 g (55%) of the title compound. 1H NMR (400 MHz, DMSO-d6) δ 9.32 (s, 1H), 6.90-^.80 (m, 1H), 6.51 (td, J=8.7, 5.5, 1H), 5.28 (s, 2H), 3.05-2.96 (m, 2H), 1.82-1.64 (m, 2H), 1.01-0.90 (m, 3H). LC/MS: w/z 251.1 [M+l].
Exam le H
Figure imgf000052_0002
N-(3 - Amino-4-chloro-2-fluorophenyl)propane- 1 -sulfonamide
[00221] Triethylamine (1.84 mL, 13.2 mmol) and diphenylphosphonic azide (1.43 mL, 6.61 mmol) were added to a solution of 6-chloro-2-fluoro-3-(propylsulfonamido)benzoic acid (1.70 g, 5.75 mmol) in THF (23 mL). The reaction mixture was stirred at room temperature for 1 hour, warmed to 70°C and stirred for 1 hour. Water (6 mL) was added, after which the reaction mixture was stirred again at 70°C for 3 hours. The mixture was cooled to room temperature, ethyl acetate was added, and the layers were separated. The organic phase was dried with sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by flash silica gel chromatography using 0-50% EtO Ac/heptane gradient to afford N-(3- amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide (1.01 g, 66%) as a solid. 1H NMR (500 MHz, (CD3)2SO) δ 9.54 (s, 1H), 7.02 (d, 1H), 6.58 (t, 1H), 5.50 (s, 2H), 3.09-2.95 (t, 2H), 1.81-1.64 (m, 2H), 0.96 (t, 3H). LC/MS: m/z 267.1 [M+l].
Example I
Figure imgf000053_0001
N-(3-Amino-2-chloro-4-fluorophenvnpropane-l-sulfonamide
[00222] The compound was prepared using the procedure described in Example G using 2-chloro-6-fluoro-3-(propylsulfonamido)benzoic acid instead 2,6-difluoro-3 -(propyl - sulfonamido)benzoic acid as starting material. 1H NMR (400 MHz, (CD3)2SO) δ 9.20 (s, 1H), 7.28-6.99 (m, 1H), 6.63 (td, J=8.7, 5.5, 1H), 5.45 (s, 2H), 3.07-2.99 (m, 2H), 1.88-1.69 (m, 2H), 1.03-0.95 (m, 3H). LC/MS: m/z 267.1 [M+l].
Exam le J
Figure imgf000053_0002
N-(3-Amino-2,4-difluorophenyl)ethanesulfonamide
[00223] Triethylamine (8.02 mL, 57.61 mmol) and diphenylphosphonic azide (6.21 mL, 28.81 mmol) were added to a solution of 2,6-difluoro-3-(propylsulfonamido)benzoic acid (6.643 g, 25.1 mmol) in THF (50 mL). The reaction mixture was stirred at room temperature for 3 hours and then warmed to 80°C for 2 hours. Water (15 mL, 830 mmol) was added, and the mixture stirred at 80°C for 15 hours. The reaction mixture was diluted with EtOAc (500 mL), and the organic layer was washed with saturated aqueous NaHC03 solution and brine. The solvent was removed under reduced pressure, and the residual purified via silica gel column chromatography eluting with EtOAc/hexane (30/70) to obtain 3.03 g (50%) of the title compound. 1H NMR (400 MHz, (CD3)2SO) δ 9.37 (d, J=29.6, 1H), 6.86 (dd, J=10.7, 9.1, 1.9, 1H), 6.52 (td, J=8.7, 5.5, 1H), 5.28 (s, 2H), 3.03 (q, J=7.3, 2H), 1.25 (td, J=7.3, 2.5, 3H). LC/MS: m/z 237.1 [M+l].
Exam le K
Figure imgf000054_0001
N-(3-Amino-4-chloro-2-fluorophenyl ethanesulfonamide
[00224] Triethylamine (9.10 mL, 65.3 mmol) and diphenylphosphonic azide (7.04 mL, 32.7 mmol) was added to a solution of 6-chloro-3-(ethylsulfonamido)-2-fluorobenzoic acid (8.00 g, 28.4 mmol) in THF (115 mL). The reaction mixture was stirred at room temperature for 4 hours, warmed to 70°C and stirred for 2 hours. Water (27 mL) was then added, after which the reaction mixture was stirred again at 70°C for 16 hours. The mixture was cooled to room temperature, ethyl acetate and a saturated solution of NaHC03 were added, and the layers were separated. The organic phase was dried with sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by flash silica gel chromatography using 30-50% EtOAc/heptane gradient to afford N-(3-amino-4-chloro-2- fluorophenyl)ethanesulfonamide (4.24 g, 59%) as a solid. 1H NMR (500 MHz, (CD3)2SO) δ 9.48 (s, 1H), 7.02 (dd, J=8.8, 1.7 Hz, 1H), 6.59 (t, J=8.3 Hz, 1H), 5.44 (s, 2H), 3.07 (q, J=7.3 Hz, 2H), 1.24 (t, J=7.3 Hz, 3H). LC/MS: m/z 253.2 [M+l].
Exam le L
Figure imgf000054_0002
N-(3-Amino-2-chloro-4-fluorophenyl)ethanesulfonamide
[00225] 2-Chloro-6-fluoro-3-(ethylsulfonamido)benzoic acid (3.3 g, 12.0 mmol) was treated with thionyl chloride (21.0 mL, 0.29 mmol) and heated at reflux for 15 hours. The reaction mixture was concentrated and then azeotrophed with toluene (2 X 20 mL). The residue was treated with a solution of sodium azide (3.1 g, 48.0 mmol) dissolved in water (20 mL) and acetone (20 mL). After stirring at room temperature for 1 hour, the intermediate acyl azide was extracted into ethyl acetate (2 X 25 mL), dried with magnesium sulfate and concentrated. The residue was dissolved in dioxane (40 mL) and water (5 mL) and heated to reflux for 3 hours. After cooling to room temperature, the product was extracted into methylene chloride (2 X 25 mL), dried with magnesium sulfate and concentrated. The residue was purified by flash silica gel chromatography (2-30% isopropanol in methylene chloride) to afford N-(3-amino-2-chloro-4-fluorophenyl)ethanesulfonamide (2.0 g, 66%). 1H NMR (400 MHz, (CD3)2SO) 6 9.15 (s, 1H), 7.02 (dd, J= 10.7, 8.8 Hz, 1H), 6.64 (dd, J= 8.8, 5.1 Hz, 1H), 5.45 (s, 2H), 3.06 (q, J = 7.3 Hz, 2H), 0.96 (t, J = 7.3 Hz, 3H). LC/MS: m/z 253.0 [M+l].
Example M
Figure imgf000055_0001
N-(3 - Amino-2-chloro-4-fluorophenyl)- 1 -cyclopropylmethanesulfonamide
[00226] Step A: Cyclopropylmethanesulfonyl chloride (4.74 g, 30.6 mmol) was added dropwise to a solution of methyl 3-amino-2-chloro-6-fluorobenzoate (2.97 g, 14.6 mmol) in THF (26 mL) and triethylamine (6.10 mL, 43.8 mmol) at 0°C. The reaction mixture was stirred at 0°C for 90 minutes, after which 8N NaOH (18.2 mL, 140 mmol) was added. The reaction mixture was then warmed up at 40°C and stirred for 16 hours. The volatiles were removed in vacuo, and the mixture acidified with concentrated HC1 at 0°C to pH 1. The acidified mixture was extracted with ethyl acetate twice. The organic phases were combined, dried with sodium sulfate, filtered and concentrated in vacuo to obtain crude 2-chloro-3- (cyclopropylmethylsulfonamido)-6-fluorobenzoic acid, which was used directly in the next step without further purification.
[00227] Step B: Triethylamine (2.05 mL, 14.7 mmol) was added to a solution of 2- chloro-3-(cyclopropylmethylsulfonamido)-6-fluorobenzoic acid (4.11 g, 13.4 mmol) in 1,4- dioxane (30 mL), followed by diphenylphosphonic azide (3.12 mL, 14.0 mmol) at room temperature. The reaction was stirred at room temperature for 4 hours, and the resulting mixture added dropwise, via an addition funnel, over 15 minutes in a round-bottom flask containing 1,4-dioxane (16 mL) and water (1.20 mL, 66.8 mmol) at 95°C. The reaction mixture was stirred at this temperature for 16 hours. Half of the reaction mixture was concentrated in vacuo, then the rest of the solution was diluted with ethyl acetate and a saturated solution of NaHC03. The layers were separated, and the aqueous layer was extracted twice with ethyl acetate. The organic phases were combined, dried with sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by flash chromatography to afford N-(3-amino-2-chloro-4-fluorophenyl)-l- cyclopropylmethanesulfonamide (2.05 g, 55%). m/z (ES-MS) M+l = 279.2. 1H NMR (500 MHz, (CD3)2SO) δ 9.07 (s, 1H), 7.01 (dd, J = 10.7, 8.9 Hz, 1H), 6.66 (dd, J = 8.8, 5.1 Hz, 1H), 5.43 (s, 2H), 3.04 (d, J = 7.1 Hz, 2H), 1.12 - 0.99 (m, 1H), 0.59 - 0.52 (m, 2H), 0.36 - 0.30 (m, 2H).
Example N
Figure imgf000056_0001
N-(3 - Amino-2-chloro-4-fluorophenyl)-2-methylpropane- 1 -sulfonamide
[00228] Step A: 2-Methylpropane-l-sulfonyl chloride (4.80 g, 30.6 mmol) was added dropwise to a solution of methyl 3-amino-2-chloro-6-fluorobenzoate (2.97 g, 14.6 mmol) in THF (20 mL) and triethylamine (6.10 mL, 43.8 mmol) at 0°C. The reaction mixture was stirred at 0°C for 90 minutes, after which 8N aqueous NaOH (18.2 mL, 140 mmol) was added. The reaction mixture was warmed up at 40°C and stirred for 16 hours. The volatiles were then removed in vacuo, and the mixture was acidified with concentrated HCl at 0°C to pH 1. The acidified mixture was extracted with ethyl acetate twice. The organic phases were combined, dried with sodium sulfate, filtered and concentrated in vacuo to obtain crude 2- chloro-6-fluoro-3-(2-methylpropylsulfonamido)benzoic acid, which was used directly in the next step without further purification.
[00229] Step B: N-(3-Amino-2-chloro-4-fluorophenyl)-2-methylpropane-l- sulfonamide was prepared according to the general procedure for Example A, Step B, substituting 2-chloro-6-fluoro-3-(2-methylpropylsulfonamido)benzoic acid for 2-chloro-3- (cyclopropylmethylsulfonamido)-6-fluorobenzoic acid, m/z (ES-MS) M+l = 281.2. 1H NMR (500 MHz, (CD3)2SO) δ 9.14 (s, 1H), 7.02 (dd, 7 = 10.7, 8.9 Hz, 1H), 6.64 (dd, J = 8.8, 5.1 Hz, 1H), 5.44 (s, 2H), 2.96 (d, J= 6.4 Hz, 2H), 2.20-2.10 (m, 1H), 1.01 (d, J= 6.7 Hz, 6H).
Example O
Figure imgf000056_0002
N-(3-Amino-2,5-difluorophenyl)propane-l-sulfonamide
[00230] Propane- 1-sulfonyl chloride (1.867 mL, 16.65 mmol) at 0°C was added to a solution of 2,5-difluorobenzene-l,3-diamine (2.00 g, 13.9 mmol) (described in Moilliet, J. S. et al., EP415595, 1989, 4) in THF (40 mL) and pyridine (1.571 mL, 19.43 mmol). The reaction mixture was stirred at 50°C for 90 minutes, and DCM and a saturated solution of NaHC03 were then added. The layers were separated, and the aqueous layer was extracted twice with DCM. The organic layers were combined, dried with sodium sulfate, filtered and concentrated in vacuo. The crude mixture was re-submitted to exact same reaction conditions, and the reaction was stirred at 55°C for 16 hours. Ethyl acetate and a saturated solution of NaHC03 were then added. The layers were separated, and the aqueous layer extracted twice with ethyl acetate. The organic layers were combined, dried with sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by flash chromatography to afford N-(3-amino-2,5-difluorophenyl)propane-l -sulfonamide (485 mg, 14%).
Example P
Figure imgf000057_0001
N-(3-Amino-2,4-difluorophenyl)-2-methylpropane- 1 -sulfonamide
[00231] Step A: 2,6-Difluoro-3-(2-methylpropylsulfonamido)benzoic acid was prepared according to the general procedure for Example N, Step A, substituting methyl 3- amino-2,6-difluorobenzoate for methyl 3-amino-2-chloro-6-fluorobenzoate.
[00232] Step B: N-(3-Amino-2,4-difluorophenyl)-2-methylpropane-l -sulfonamide was prepared according to the general procedure for Example M, Step B, substituting 2,6- difluoro-3-(2-methylpropylsulfonamido)benzoic acid for 2-chloro-3-(cyclopropylmethyl- sulfonamido)-6-fluorobenzoic acid, m/z (ES-MS) M+l = 265.2. 1H NMR (400 MHz, (CD3)2SO) δ 9.36 (s, 1H), 6.86 (t, J= 9.8 Hz, 1H), 6.55-6.47 (m, 1H), 5.32 (s, 2H), 2.93 (d, J = 6.4 Hz, 2H), 2.21-2.10 m, 1H), 1.01 (d, J= 6.7 Hz, 6H).
Example Q
Figure imgf000057_0002
N-(3 - Amino-2,4-difluorophenylV 1 -cvclopropylmethanesulfonamide
[00233] Step A: 3-(Cyclopropylmethylsulfonamido)-2,6-difluorobenzoic acid was prepared according to the general procedure for Example M, Step A, substituting methyl 3- amino-2,6-difluorobenzoate for methyl 3-amino-2-chloro-6-fluorobenzoate.
[00234] Step B: N-(3-Amino-2,4-difluorophenyl)-l -cyclopropylmethanesulfonamide was prepared according to the general procedure for Example M, Step B, substituting 3- (cyclopropylmethylsulfonamido)-2,6-difluorobenzoic acid for 2-chloro-3-(cyclopropyl- methylsulfonamido)-6-fluorobenzoic acid, m/z (ES-MS) M+l = 263.2. 1H NMR (400 MHz, (CD3)2SO) δ 9.37 (s, 1H), 6.84 (t, J= 9.8 Hz, 1H), 6.57-6.50 (m, 1H), 5.30 (s, 2H), 3.01 (d, J = 7.1 Hz, 2H), 1.11 - 0.98 (m, 1H), 0.59 - 0.51 (m, 2H), 0.35 - 0.27 (m, 2H).
Example R
Figure imgf000058_0001
N-(3-Amino-5-chloro-2-fluorophenyl)propane-l-sulfonamide
[00235] Step A: Fuming nitric acid (4.98 mL, 119 mmol) was added to a solution of methyl 5-chloro-2-fluorobenzoate (16.0 g, 84.8 mmol) in sulfuric acid (100 mL) at 0°C. The reaction mixture was stirred at room temperature for 3 hours, poured into ice/water, and the resulting precipitate was filtered. The obtained solid was purified by flash chromatography to afford methyl 5-chloro-2-fluoro-3-nitrobenzoate (6.78 g, 30%), contaminated with 10% of an undesired regioisomer.
[00236] Step B: A round-bottom flask was charged with 5-chloro-2-fluoro-3- nitrobenzoate (6.78 g, 29.0 mmol), iron (16.2 g, 290 mmol), ammonium chloride (5.43 g, 102 mmol), ethanol (100 mL) and water (30 mL). The reaction mixture was stirred at 85°C for 2 hours and then cooled to room temperature. The mixture was diluted with ethyl acetate and a saturated solution of NaHC03, and the layers were separated. The aqueous layer was extracted twice with ethyl acetate. The organic layers were combined, dried with sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by flash chromatography to afford methyl 3-amino-5-chloro-2-fluorobenzoate (3.7 g, 63%), contaminated with 10% of an undesired regioisomer.
[00237] Step C: Propane- 1-sulfonyl chloride (3.12 mL, 27.8 mmol) was added dropwise to a solution of methyl 3-amino-5-chloro-2-fluorobenzoate (2.71 g, 13.3 mmol) in THF (25 mL) and triethylamine (5.54 mL, 39.8 mmol) at 0°C. The reaction mixture was stirred at 0°C for 90 minutes, after which 8N aqueous NaOH (16.6 mL, 130 mmol) was added. The reaction mixture was then warmed up at 40°C and stirred for 16 hours. The volatiles were removed in vacuo, and the mixture was then acidified with concentrated HCl at 0°C to pH 1. The acidified mixture was extracted with ethyl acetate twice. The organic layers were combined, dried with sodium sulfate, filtered and concentrated in vacuo to obtain crude 5-chloro-2-fluoro-3-(propylsulfonamido)benzoic acid, which was used in the next step without further purification. [00238] Step D: N-(3-Amino-5-chloro-2-fluorophenyl)propane-l -sulfonamide was prepared according to the general procedure for Example M, Step B, substituting 5-chloro-2- fluoro-3-(propylsulfonamido)benzoic acid for 2-chloro-3-(cyclopropylmethylsulfonamido)-6- fluorobenzoic acid, m/z (ES-MS) M+l = 267.0. 1H NMR (400 MHz, (CD3)2SO) δ 9.58 (s, 1H), 6.59 (dd, J = 7.1, 2.6 Hz, 1H), 6.53 (dd, J= 5.9, 2.6 Hz, 1H), 5.56 (s, 2H), 3.11 - 3.03 (m, 2H), 1.78 - 1.65 (m, 2H), 0.97 (t, J= 7.4 Hz, 3H).
Example S
Figure imgf000059_0001
N-(3-Arnmo-4-c oro-2-fluorophenyl)-2-methylpropane-l-sulfonamide
[00239] Step A: Benzyl 6-chloro-2-fluoro-3-(N-(isobutylsulfonyl)-2-methylpropyl- sulfonamido)benzoate was prepared according to the general procedure for Example F, Step B, substituting 2-methylpropane-l-sulfonyl chloride for propane- 1-sulfonyl chloride.
[00240] Step B: 6-Chloro-2-fluoro-3-(2-methylpropylsulfonamido)benzoic acid was prepared according to the general procedure for Example F, Step C, substituting benzyl 6- cUoro-2-fluoro-3-(N-(isobutylsulfonyl)-2-methylpropylsulfonamido)benzoate for benzyl 6- chloro-2-fluoro-3 -(N-(propylsulfonyl)propylsulfonamido) benzoate.
[00241] Step C: N-(3-Amino-4-chloro-2-fluorophenyl)-2-methylpropane-l- sulfonamide was prepared according to the general procedure for Example M, Step B, substituting 6-chloro-2-fluoro-3-(2-methylpropylsulfonamido)benzoic acid for 2-chloro-3- (cyclopropylmethylsulfonamido)-6-fluorobenzoic acid, m/z (ES-MS) M+l = 281.2. 1H NMR (400 MHz, (CD3)2SO) δ 9.50 (s, 1H), 7.02 (dd, J = 8.8, 1.8 Hz, 1H), 6.62 - 6.54 (m, 1H), 5.45 (s, 2H), 2.97 (d, J- 6.4 Hz, 2H), 2.21-2.10 (m, 1H), 1.01 (d, J= 6.7 Hz, 6H).
Example T
Figure imgf000059_0002
N-(3-Amino-2-chloro-5-fluorophenvDpropane- 1 -sulfonamide
[00242] Propane- 1-sulfonyl chloride (1.41 mL, 12.6 mmol) at 0°C was added to a solution of 2-chloro-5-fluorobenzene-l,3-diamine (1.01 g, 6.29 mmol; 70% purity) (described in Frutos, R. P. et al., US 2006/0258888, 2006, 3) in DCM (30 mL) and triethylamine (1.93 mL, 13.8 mmol). The reaction mixture was stirred at room temperature for 1 hour. An aqueous saturated solution of NaHC03 and ethyl acetate were added, and the layers were separated. The aqueous layer was extracted twice with ethyl acetate. The organic phases were combined, dried with sodium sulfate, filtered and concentrated in vacuo. The crude mixture was dissolved in tetrahydrofuran (15 mL) and methanol (5 mL), and then 1.0M of sodium hydroxide in water (6.3 mL) was added. The reaction mixture was stirred at room temperature for 30 minutes. An aqueous saturated solution of NaHC03 and ethyl acetate were added, and the layers were separated. The aqueous layer was extracted twice with ethyl acetate. The organic layers were combined, dried with sodium sulfate, filtered and concentrated in vacuo. The crude product was purified by flash chromatography to afford N- (3-amino-2-chloro-5-fluorophenyl)propane-l-sulfonamide (0.17 g, 7%). m/z (ES-MS) M+l = 281.2.
Exam le U
Figure imgf000060_0001
N-(3-Amino-2,4-dichlorophenyl)propane- 1 -sulfonamide
[00243] Step A: 2,6-Dichloro-3-nitrobenzoic acid (2.13 g, 9.03 mmol) was dissolved in 2:1 THF:saturated aqueous N¾C1 and cooled to 0°C. The mixture was treated with zinc (11.8 g, 181 mmol) and then allowed to warm to ambient temperature and stir for 24 hours. The reaction mixture was filtered through GF/F paper while rinsing with THF. The mixture was acidified to a pH of 1 using 1.0 M HC1 and extracted with 15% 2-propanol/DCM (3 X). The extracts were washed with water and brine, dried over sodium sulfate and concentrated to afford 3-amino-2,6-dichlorobenzoic acid (1.40 g, 6.82 mmol, 75.5% yield). MS (APCI- neg) w/z =203.6 (M-H).
[00244] Step B: 3-Amino-2,6-dichlorobenzoic acid (1.40 g, 6.82 mmol) was dissolved in dry dichloromethane (66.7 mL). Triethylamine (4.09 mL, 29.4 mmol) was added, and the mixture was chilled to 0°C. Propane- 1-sulfonyl chloride (2.48 mL, 22 mmol) was then added using a syringe. When the addition was complete, the mixture was allowed to warm to ambient temperature and stir for 1 hour. The mixture was concentrated in vacuo and diluted with diethyl ether. The mixture was washed with 0.25 M NaOH (80 mL), and the aqueous layer acidified to a pH of 1 using 1.0M HC1. The aqueous layer was extracted with 15% 2- propano DCM (2 X 300 mL). The organic layer was collected, dried over sodium sulfate, and concentrated to afford 2,6-dichloro-3-(propylsulfonamido)benzoic acid (1.55 g, 4.96 mmol, 74.4% yield). 1H NMR (400 MHz, (CD3)2SO) δ 9.77-9.75 (s, IH), 7.84-7.80 (d, IH), 7.71-7.68 (d, IH), 3.82-3.72 (m, 2H), 1.89-1.70 (m, 2H), 1.05-1.03 (m, 3H).
[00245] Step C: Triethylamine (2.863 mL, 20.5 mmol) and diphenylphosphonic azide (2.282 mL, 10.2 mmol) was added to a solution of 2,6-dichloro-3- (propylsulfonamido)benzoic acid (2.788 g, 8.93 mmol in THF (40 mL). The reaction mixture was stirred for 6 hours at room temperature. Then water (8 mL, 400 mmol) was added, and the reaction mixture was heated under reflux overnight. Ethyl acetate (300 mL) was added, followed by washing with saturated aqueous NaHC03 solution and brine. The solvent was removed under reduced pressure, and the crude product purified via silica gel flash chromatography using ethyl acetate/hexane (1 :1) as eluent to yield N-(3-amino-2,4- dichlorophenyl)propane-l -sulfonamide (834 mg, 33%). 1H NMR (500 MHz, (CD3)2SO) δ 9.24 (s, 1H), 7.20 (d, J - 8.7 Hz, 1H), 6.71 (d, J= 8.7 Hz, 1H), 5.55 (s, 2H), 3.13 - 2.92 (m, 2H), 1.73 (dd, J= 15.2, 7.6 Hz, 2H), 0.96 (t, J= 7.4 Hz, 3H). LC-MS [M+l] mJz 284.1.
Example V
Figure imgf000061_0001
2,6-Difluoro-3-(3-fluoropropylsulfonamido)benzoic acid
[00246] Step A: Methyl 2,6-difluoro-3-(N-(3-fluoropropylsulfonyl)-3-fluoropropyl- sulfonamido)benzoate was prepared according to the general procedure described in Example B, Step C, substituting 3-fluoropropyl sulfonyl chloride for propane- 1-sulfonyl chloride. 1H NMR (400 MHz, (CD3)2SO) δ 8.05-7.99 (m, 1H), 7.44 (t, 1H), 4.62 (t, 2H), 4.50 (t, 2H), 3.93 (s, 3H), 3.89-3.74 (m, 4H), 2.26-2.11 (m, 4H).
[00247] Step B: 2,6-Difluoro-3-(3-fluoropropylsulfonamido)benzoic acid was prepared according to the general procedure in Example C, substituting methyl 2,6-difluoro- 3-(N-(3-fluoropropylsulfonyl)-3-fluoropropylsulfonamido)benzoate for methyl 2,6-difluoro- 3-(N-(propylsulfonyl)-propylsulfonamido)benzoate. 1H NMR (400 MHz, CD3OD) δ 7.64 (m, 1H), 7.07 (m, 1H), 4.58 (m, 1H), 4.47 (m, 1H), 3.22 (m, 2H), 2.26-2.12 (m, 2H). MS m/z 296.1 [M-l].
Example W
Figure imgf000061_0002
6-Chloro-2-fluoro-3 -(3 -fluoropropylsulfonamido)benzoic acid [00248] Step A: Benzyl 6-chloro-2-fluoro-3-(3-fiuoro-N-(3-fluoropropylsulfonyl)- propylsulfonamido)benzoate (92%) was prepared according to the general procedure described in Example F, Step B, substituting 3-fluoropropane-l-sulfonyl chloride for propane- 1-sulfonyl chloride.
[00249] Step B: 6-Chloro-2-fluoro-3-(3-fluoropropylsulfonamido)benzoic acid (71%) was prepared according to the general procedure for Example F, Step C, substituting benzyl 6-chloro-2-fluoro-3 -(3 -fluoro-N-(3 -fluoropropylsulfonyl)propylsulfonamido)benzoate for benzyl 6-chloro-2-fluoro-3-(N-(propylsulfonyl)propylsulfonamido)benzoate. 1H NMR (400 MHz, (CD3)2SO) δ 9.65 (br s, 1H), 7.03 (m, 1H), 6.58 (m, 1H), 4.59 (m, 1H), 4.47 (m, 1H), 3.18 (m, 2H), 2.22-2.02 (m, 2H). MS m/z 312.1, 314.1 [M-l].
Example X
Figure imgf000062_0001
2-Chloro-6-fluoro-3 -(3 -fluoropropylsulfonamido)benzoic acid
[00250] Step A: Methyl 2-chloro-6-fluoro-3-(3-fluoro-N-(3-fluoropropylsulfonyl)- propylsulfonamido)benzoate was prepared according to the general procedure in Example B, Step C, substituting 3-fluoropropyl sulfonyl chloride for propane- 1-sulfonyl chloride.
[00251] Step B: 2-Chloro-6-fiuoro-3-(3-fluoropropylsulfonamido)benzoic acid (93% over 2 steps) was prepared according to the general procedure for Example F, Step C, substituting methyl 2-chloro-6-fluoro-3-(3-fiuoro-N-(3- fluoropropylsulfonyl)propylsulfonamido)benzoate for benzyl 6-chloro-2-fluoro-3-(N- (propylsulfonyl)propylsulfonamido)benzoate. 1H NMR (400 MHz, CD3OD) δ 7.63 (m, 1H), 7.19 (m, 1H), 4.56 (m, 1H), 4.44 (m, 1H), 3.21 (m, 2H), 2.25-2.12 (m, 2H). MS m/z 312.1, 314.1 [M-l].
Example Y
Figure imgf000062_0002
N-(3 - Amino-2,4-difluorophenvD-3 -fluoropropane- 1 -sulfonamide
[00252] Triethylamine (0.682 mL, 4.89 mmol) and diphenylphosphoryl azide (0.547 mL, 2.45 mmol) were added to a solution of 2,6-difluoro-3-(3- fluoropropylsulfonamido)benzoic acid (0.485 g, 1.63 mmol) in DMF (10 mL). The reaction mixture was stirred at room temperature for 1 hour. Water (1.18 mL, 65.2 mmol) was added, and the mixture was stirred at 80°C for 15 hours. The reaction mixture was diluted with ethyl acetate (100 mL), and the organic layer was washed with saturated aqueous NaHC03, dried over sodium sulfate and concentrated. The crude product was purified via silica gel chromatography, eluting with hexanes/ethyl acetate (4:1) to give the title compound (0.34 g, 77%). 1H NMR (400 MHz, (CD3)2SO) δ 9.49 (br s, 1H), 6.87 (m, 1H), 6.52 (m, 1H), 5.33 (br s, 2H), 4.60 (m, 1H), 4.48 (m, 1H), 3.14 (m, 2H), 2.16-2.03 (m, 2H). LC/MS: m/z 267.1 [M- 1]·
Example Z
Figure imgf000063_0001
N-(3 - Amino-4-chloro-2-fluorophenyl)-3 -fluoropropane- 1 -sulfonamide
[00253] N-(3-Amino-4-chloro-2-fluorophenyl)-3 -fluoropropane- 1 -sulfonamide (8.8 g, 52%) was prepared according to the general procedure in Example Y, substituting 6-chloro- 2-fluoro-3-(3-fluoropropylsulfonamido)benzoic acid for 2,6-difluoro-3-(3-fluoropropyl- sulfonamido)benzoic acid. 1H NMR (400 MHz, (CD3)2SO) δ 9.65 (br s, 1H), 7.03 (m, 1H), 6.58 (m, 1H), 5.50 (br s, 2H), 4.59 (m, 1H), 4.48 (m, 1H), 3.17 (m, 2H), 2.16-2.03 (m, 2H). LC/MS: m/z 283.1, 285.1 [M-1J.
Example AA
Figure imgf000063_0002
N-(3 -Amino-2-chloro-4-fluorophenyl -3 -fluoropropane- 1 -sulfonamide
[00254] N-(3-Amino-2-chloro-4-fluorophenyl)-3-fluoropropane-l-sulfonamide
(0.88 g, 42%) was prepared according to the general procedure in Example Y, substituting 2- chloro-6-fluoro-3-(3-fluoropropylsulfonamido)benzoic acid for 2,6-difluoro-3-(3-fluoro- propylsulfonamido)benzoic acid. 1H NMR (400 MHz, (CD3)2SO) δ 9.35 (br s, 1H), 7.03 (m, 1H), 6.63 (m, 1H), 5.50 (br s, 2H), 4.59 (m, 1H), 4.48 (m, 1H), 3.17 (m, 2H), 2.16-2.03 (m, 2H). LC/MS: m/z 283.1, 285.1 [M-l].
Exam le AB
Figure imgf000063_0003
(3-Amino-4-methylphenyl)propane- 1 -sulfonamide [00255] Step A: TEA (4.58 mL, 32.9 mmol) was added to 4-methyl-3-nitroaniline (1.0 g, 6.57 mmol) in DCM (30 mL) at 0°C, followed by propane- 1-sulfonyl chloride (1.84 mL, 16.4 mmol). The solution was allowed to warm to ambient temperature and stirred for 2 hours. The solution was diluted with aqueous bicarbonate (100 mL) and extracted with EtOAc (3 X 40 mL). The organics were dried over sodium sulfate, filtered and concentrated under reduced pressure to afford N-(4-methyl-3-nitrophenyl)-N-(propylsulfonyl)propane-l- sulfonamide (2.4 g, 100%).
[00256] Step B: 2M NaOH (16 mL, 33 mmol) was added to N-(4-methyl-3- nitrophenyl)-N-(propylsulfonyl)propane-l -sulfonamide (2.4 g, 6.6 mmol) in 4:1 THF:MeOH (75 mL). The solution was warmed to 50°C for 3 hours. The cooled solution was concentrated under reduced pressure, and the residue was diluted with aqueous ammonium chloride (100 mL) and extracted with EtOAc (3 X 40 mL). The combined organics were washed with aqueous bicarbonate (2 X 50 mL), then dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford N-(4-methyl-3-nitrophenyl)propane-l- sulfonamide (1.6 g, 94%).
[00257] Step C: 10% Pd/C (1.32 g, 1.24 mmol) was added to N-(4-methyl-3- nitrophenyl)propane-l -sulfonamide (1.6 g, 6.19 mmol) in EtOH (30 mL). The suspension was stirred under a balloon of hydrogen at ambient temperature for 16 hours. The suspension was filtered and concentrated to afford (3 -amino-4-methylphenyl)propane-l -sulfonamide (1.38 g, 97.6%). Ή NMR (400 MHz, CD3OD) δ 6.96-6.99 (m, 1H), 6.63-6.65 (m, 1H), 6.45- 6.49 (m, 1H), 6.38 (br s, 1H), 3.02-3.07 (m, 2H), 2.13 (s, 3H), 1.75-1.90 (m, 2H), 0.98-1.04 (m, 3H). LCMS: m/z 229.1 [M+l].
xample AC
Figure imgf000064_0001
N-(3 - Amino-4-chlorophenyl)propane- 1 -sulfonamide
[00258] Step A: Triethylamine (4.19 mL, 29.0 mmol) and propane- 1-sulfonyl chloride (1.63 mL, 14.5 mmol) was added to 4-chloro-3-nitroaniline (29.0 mL, 5.79 mmol) in DCM (30 mL) at 0°C. The solution was allowed to warm to ambient temperature and stirred for 1 hour before dilution with aqueous bicarbonate (50 mL) and extraction with EtOAc (3 X 40 mL). The organics were dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford N-(4-chloro-3-nitrophenyl)-N-(propylsulfonyl)propane-l -sulfonamide (2.4 g, 107%), which was used without further purification. [00259] Step B: 2M NaOH (16 mL, 33 mmol) was added to N-(N-(4-chloro-3- nitrophenyl)-N-(propylsulfonyl)propane-l -sulfonamide (2.4 g, 6.6 mmol) in 4:1 THF:MeOH (75 mL). The solution was warmed to 50°C for 3 hours. The cooled solution was concentrated under reduced pressure, and the residue was diluted with aqueous ammonium chloride (100 mL) and extracted with EtOAc (3 X 40 mL). The combined organic extracts were washed with aqueous bicarbonate (2 X 50 mL), then dried over sodium sulfate, filtered, and concentrated under reduced pressure to afford N-(4-chloro-3-nitrophenyl)propane-l- sulfonamide (1.5 g, 88%).
[00260] Step C: 2M HC1 (2 mL) and Fe (0) (0.301 g, 5.38 mmol) were added to N-(4- chloro-3-nitrophenyl)propane-l -sulfonamide (0.50 g, 1.79 mmol) in MeOH (10 mL). The suspension was warmed to reflux for 4 hours, then cooled and filtered through GF/F paper. The filtrate was concentrated under reduced pressure to afford N-(3-amino-4- chlorophenyl)propane-l -sulfonamide (0.40 g, 89%). 1H NMR (400 MHz, CD3OD) δ 7.16- 7.21 (m, 1H), 6.90 (s, 1H), 6.58-6.63 (m, 1H), 3.02-3.08 (m, 2H), 2.13 (s, 3H), 1.73-1.84 (m, 2H), 0.97-1.05 (m, 3H). LC/MS: m/z 247.1 [M-l].
Example AD
Figure imgf000065_0001
N-(3 - Amino-2-chlorophenyl)propane- 1 -sulfonamide
[00261] Step A: 2-Chloro-3-nitroaniline (Sienkowska, et. al, Tetrahedron 56 (2000) 165) (0.36 g, 2.086 mmol) was dissolved in DCM (20 mL) and cooled to 0°C. Triethylamine (0.8723 mL, 6.258 mmol) was added followed by propane- 1-sulfonyl chloride (0.5847 mL, 5.215 mmol) and the reaction was stirred at room temperature overnight. The reaction was quenched with 0.1N HC1 (10 mL), and the layers were separated. The organic layer was dried over Na2S04, and concentrated to give N-(2-chloro-3-nitrophenyl)-N- (propylsulfonyl)propane-l -sulfonamide as an oil, which was used directly in the next step.
[00262] Step B: N-(2-Chloro-3-nitrophenyl)-N-(propylsulfonyl)propane-l- sulfonamide (0.8028 g, 2.086 mmol) was dissolved in 3:1 THF/MeOH (4.0 mL). NaOH (2.0 M, 2.086 mL, 4.172 mmol) was added, and the reaction was stirred for five minutes at room temperature. The reaction was quenched with 0.1N HC1 (5 mL), and the volatiles were removed by rotary evaporation. EtOAc (10 mL) was added, and the organic layer was washed with water and brine, dried with Na2S04 and concentrated to give N-(2-chloro-3- nitrophenyl)propane-l -sulfonamide as an oil, which was used directly in the next step. [00263] Step C: N-(2-Chloro-3-nitrophenyl)propane-l -sulfonamide (0.580 g, 2.08 mmol) was dissolved in 4:1 EtOH/water (10 mL). Fe (0) (1.16 g, 20.8 mmol) was added, followed by a catalytic amount of NH4CI (5 mg), and the reaction was heated to 80°C for 3 hours. The reaction was cooled to room temperature, filtered through Celite®, concentrated, dissolved in EtOAc, washed with water, dried over Na2S04 and concentrated. Purification by silica gel chromatography (10% to 90% EtOAc/hexanes) gave N-(3-amino-2- chlorophenyl)propane-l -sulfonamide (259 mg, 1.04 mmol, 51%) as an oil. 1H NMR (400 MHz, (CD3)2SO) δ 9.06 (br s, 1H), 6.96-6.99 (d, 1H), 6.63-6.66 (m, 2H), 5.43 (br s, 1H), 3.03-3.07 (t, 1H), 1.71-1.77 (m, 2H), 0.94-0.98 (t, 3H); m/z (APCI-neg) M-l = 247.1, 249.0.
Example AE
Figure imgf000066_0001
N-(3 - Amino-4-fluorophenyl) propane- 1 -sulfonamide
[00264] N-(3-Amino-2-chloro-4-fluorophenyl) propane- 1 -sulfonamide (668 mg, 2.5 mmol) was dissolved in methanol (100 mL), and the solution was run through an H-Cube hydrogenator at 50°C and 10 bar H2 pressure at a 1 mL/min flow rate. After removal of the solvent, 481 mg (83%) of N-(3-amino-4-fluorophenyl) propane- 1 -sulfonamide were obtained. 1H NMR (500 MHz, (CD3)2SO) δ 9.37 (s, 1H), 6.89 (dd, J= 11.2, 8.7, 1H), 6.67 (dd, J= 8.1, 2.6, 1H), 6.49 - 6.24 (m, 1H), 5.19 (s, 2H), 3.09 - 2.86 (m, 2H), 1.67 (dq, J= 15.0, 7.5, 2H), 0.93 (t, J= 7.4, 3H). LC-MS [M+l] m/z 233.1.
Example AF
Figure imgf000066_0002
N-(3-Amino-2 -fluorophenyl) propane- 1 -sulfonamide
[00265] N-(3-Amino-4-chloro-2-fluorophenyl) propane- 1 -sulfonamide (477 mg, 1.8 mmol) was dissolved in methanol (100 mL), and the solution was run through an H-Cube hydrogenator at room temperature and ambient pressure at a 1 mL/min flow rate. After removal of the solvent, N-(3-amino-2-fluorophenyl) propane- 1 -sulfonamide was obtained (251 mg, 60%). 1H NMR (500 MHz, (CD3)2SO) δ 9.29 (s, 1H), 6.79 (t, J= 8.0, 1H), 6.58 (td, J = 8.1, 1.4, 1H), 6.55 - 6.49 (m, 1H), 5.17 (s, 2H), 3.02 (dd, J = 8.7, 6.7, 2H), 1.85 - 1.60 (m, 2H), 0.96 (t, J= 7.4, 3H). LC-MS [M+l] m/z 233.1.
Example AG
Figure imgf000067_0001
N-(3 - Amino-2 ,4,5 -trifluorophenyPpropane- 1 -sulfonamide
[00266] 2,4,5-Trifluorobenzene-l,3-diamine (1116 mg, 6.88 mmol) was dissolved in methylene chloride (27 mL, 420 mmol), and pyridine (557 μί, 6.88 mmol) was added. After cooling the mixture to 0°C, propane- 1-sulfonyl chloride (772 μί, 6.88 mmol) was added dropwise through a syringe. The ice bath was removed, and the mixture was stirred at room temperature overnight. The solvent was removed under reduced pressure, and the crude product purified via chromatography eluting with 1:1 ethyl acetate/hexane to afford N-(3- amino-2,4,5-trifluorophenyl)propane-l -sulfonamide (1847 mg, 83.6%). 1H NMR (400 MHz, (CD3)2SO) δ 9.58 (s, 1H), 6.53 (dt, J= 11.8, 7.5 Hz, 1H), 5.75 (s, 2H), 3.10 - 2.91 (m, 2H), 1.72 (dd, J= 15.1, 7.5 Hz, 2H), 0.96 (t, J= 7.4 Hz, 3H). LC-MS [M+l] m/z 269.0.
Exam le AH
Figure imgf000067_0002
N-(3-Amino-2-cyano-4-fluorophenyl)propane- 1 -sulfonamide
[00267] Step A: Concentrated ammonium hydroxide (5.16 mL, 76.4 mmol) was added to 2,3,6-trifluorobenzonitrile (2.0 g, 12.7 mmol) in isopropanol (5 mL). The colorless solution was heated at 80°C in a sealed vial overnight. The reaction mixture was concentrated, and the residue partitioned between EtOAc and water. The EtOAc was washed with brine, dried over MgS04, filtered, and evaporated to yield 2-amino-3,6- difluorobenzonitrile (1.93 g, 12.5 mmol, 98.4% yield) as a solid.
[00268] Step B: 60% sodium hydride (0.558 g, 14.0 mmol) was added slowly in portions to propane- 1 -sulfonamide (1.68 g, 13.6 mmol) in DMSO (10 mL) with water bath cooling. After gas evolution ceased, the mixture was diluted with DMSO (5 mL) to aid in dissolution, and was stirred an additional 30 minutes at ambient temperature. A solution of 2-amino-3,6-difluorobenzonitrile (1.00 g, 6.49 mmol) in DMSO (20 mL) was added to the reaction, and the resulting mixture was heated at 100°C for 20 hours, then 120°C for 16 hours. The reaction mixture was diluted with 0.5M NaOH and washed with 2 portions EtOAc. The aqueous layer was acidified with 12M HC1 to pH 4 and extracted twice with EtOAc. The organic layer was washed with brine (3 X), dried over MgS04, filtered, and evaporated to yield a thick oil (0.41 g). This was chromatographed on a 50 g Biotage SNAP column with 1 :1 hexane:EtOAc to afford N-(3-amino-2-cyano-4-fluorophenyl)propane-l- sulfonamide (0.33 g, 1.28 mmol, 19.8% yield). 1H NMR (400 MHz, CDC13) δ 7.12-7.18 (m, 1H), 6.90-6.94 (m, 1H), 6.50 (br s, 1H), 4.58 (br s, 2H), 3.11-3.15 (m, 2H), 1.85-1.95 (m, 2H), 1.06 (t, 3H). m/z 256.1 (LC/MS negative ionization) [M-l].
Exam le AI
Figure imgf000068_0001
N-(3 -Amino-4-chloro-2-cvanophenyl)propane- 1 -sulfonamide
[00269] Step A: 14.8M ammonium hydroxide (4.67 mL, 69.1 mmol) was added to 3- chloro-2,6-difluorobenzonitrile (2.00 g, 11.5 mmol) in isopropanol (5 mL). The colorless solution was heated at 80°C in a sealed vial. After 2 hours, the reaction mixture was concentrated, and the residue partitioned between EtOAc and water. The EtOAc was washed with brine, dried over MgS04, filtered, and evaporated to yield 2-amino-3-chlorO'6- fluorobenzonitrile (1.63 g, 9.56 mmol, 82.9% yield) as a solid.
[00270] Step B: 60% sodium hydride (0.252 g, 6.30 mmol) was added slowly in portions to propane- 1 -sulfonamide (0.740 g, 6.01 mmol) in NMP (10 mL) with water bath cooling. Gas evolution continued as the mixture was stirred an additional 30 minutes at ambient temperature and then heated 1 hour at 40°C. The mixture was cooled to room temperature, and 2-amino-3-chloro-6-fluorobenzonitrile (0.50 g, 2.93 mmol) was added. The resulting mixture was heated in a sealed vial at 120°C overnight. The reaction mixture was diluted with 0.5M NaOH and washed twice with EtOAc. The aqueous layer was acidified with 12M HC1 to pH 5 and extracted with 2 portions of EtOAc. The combined EtOAc extracts were washed twice with brine, dried over MgS04, filtered, and evaporated to afford a thick oil (0.65 g). This was chromatographed on a 50g Biotage SNAP column with DCM to yield N-(3-amino-4-chloro-2-cyanophenyl)propane-l -sulfonamide (0.29 g, 1.06 mmol, 36.1% yield) as a solid. 1H NMR (400 MHz, CDC13) δ 7.40 (d, 1H), 6.97 (d, 1H), 6.62 (br s, 1H), 4.90 (br s, 2H), 3.13-3.17 (m, 2H), 1.85-1.94 (m, 2H), 1.06 (t, 3H). m/z 272.1 (LC/MS negative ionization) [M-l].
Example AJ
Figure imgf000068_0002
N-(3 -Amino-2-cvanophenyl)propane- 1 -sulfonamide
[00271] 60% sodium hydride (0.194 g, 8.08 mmol) was added to propane-1- sulfonamide (0.950 g, 7.71 mmol) in N-methylpyrrolidone (7 mL) in a vial. After gas evolution ceased, the mixture was stirred 30 minutes at 40°C. 2-Amino-6-fluorobenzonitrile (0.500 g, 3.67 mmol) was added, and the sealed vial was heated at 120°C overnight, then 150°C overnight, then for 3 days at 150°C. The reaction mixture was partitioned between 0.5M NaOH and EtOAc. The aqueous layer was acidified to pH 5 with concentrated HCl and extracted with EtOAc. The EtOAc extract was washed with brine (2 X), dried over MgS04, filtered, and evaporated to yield an oil (0.73 g). This material was dissolved in ether and washed with 3 portions water to remove NMP, dried over MgS04, filtered, and evaporated to yield N-(3-amino-2-cyanophenyl)propane-l -sulfonamide (0.34 g, 1.42 mmol, 38.7% yield) as a film. 1H NMR (400 MHz, CDC13) δ 7.29 (t, 1H), 6.98 (dd, 1H), 6.70 (br s, 1H), 6.51 (dd, 1H), 4.52 (br s, 2H), 3.18-3.14 (m, 2H), 1.95-1.85 (m, 2H), 1.06 (t, 3H). m/z 238.1 (LC/MS negative ionization) [M-l].
Exam le AK
Figure imgf000069_0001
N-(3 -Amino-2,4-difluorophenyl pyrrolidine- 1 -sulfonamide
[00272] Potassium carbonate (0.414 g, 3.00 mmol) and pyrrolidine- 1-sulfonyl chloride (0.196 mL, 1.50 mmol) were added to N-(3-amino-2,4-difluorophenyl)propane-l- sulfonamide (0.250 g, 0.999 mmol) in DMF (4.5 mL). The suspension was stirred at ambient temperature for 18 hours. 2M NaOH (1 mL) was added to the suspension, which stirred at ambient temperature for 1 hour. The resulting solution was diluted with water (20 mL) and brought to pH 9 with HCl, followed by extraction with EtOAc (3 X 15 mL). The concentrated organics were purified via silica gel chromatography eluting with 1 : 1 hexane- EtOAc to provide N-(3-amino-2,4-difluorophenyl)pyrrolidine-l -sulfonamide (184 mg, 66%). 1H NMR (400 MHz, CD3OD) δ 6.86-6093 (m, 1H), 6.75-9.82 (m, 1H), 6.34 (br s, 1H), 3.78 (br s, 2 H), 3.28-3.33 (m, 4H), 1.82-1.87 (m, 4H). LC/MS: m/z 276.1 [M-l].
Example AL
Figure imgf000070_0001
N-(3 -Amino-4-fluoro-2-methoxyphenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide
[00273] Step A: A 250 mL round bottom flask was charged with methyl 2,6-difluoro- 3-nitrobenzoate (10.03 g, 46.18 mmol) and methanol (60 mL, 1000 mmol) and was then cooled over a brine / ice bath at -4°C for 20 minutes. A 5M solution of sodium methoxide in methanol (11.98 mL, 59.88 mmol) was added dropwise to this solution over 20 minutes, while maintaining the reaction temperature at -4°C over the course of the addition. The reaction mixture was allowed to stir overnight, gradually rising to room temperature. The methanol was removed under reduced pressure, and the residual oil quenched with a saturated aqueous solution of potassium bicarbonate (250 mL). The organic layer was saved, and the aqueous layer was extracted twice with ethyl acetate (250 mL). The combined organic layers were washed once with brine, dried over magnesium sulfate, filtered, and concentrated. The crude product was purified via flash chromatography (330 g ISCO column) using a gradient of 0 - 50% ethyl acetate: heptane to yield methyl 6-fluoro-2-methoxy-3-nitrobenzoate (3.37 g. 32%) as an oil. 1H NMR (400 MHz, (CD3)2SO) δ = 8.23 (dd, J=9.3, 5.9, 1H), 7.39 (t, J=8.9, 1H), 3.94 (s, 3H), 3.89 (s, 3H).
[00274] Step B: A 250 mL round bottom flask was charged with methyl 6-fluoro-2- methoxy-3-nitrobenzoate (3.37 g, 14.71 mmol) dissolved in methanol (125 mL, 3080 mmol). Nitrogen was passed through the reaction mixture, and 10% palladium on activated carbon (1.3 g, 1.2 mmol) was added. The flask was capped and evacuated and then allowed to stir for 60 hours under an atmosphere of hydrogen at ambient temperature and pressure. The mixture was then filtered through Celite® to remove the solid catalyst and washed with methanol (500 mL). The filtrate was concentrated to give methyl 3-amino-6-fluoro-2- methoxybenzoate as an oil (2.95 g, 100%). 1H NMR (400 MHz, (CD3)2SO) δ = 6.74-6.84 (m, 2H), 4.98 (s, 2H), 3.85 (s, 3H), 3.68 (s, 3H).
[00275] Step C: A 250 mL round bottom flask was charged with a solution of methyl 3-amino-6-fluoro-2-methoxybenzoate (3.656 g, 18.36 mmol) in methylene chloride (100 mL). A solution of 4-dimethylaminopyridine (113 mg, 0.925 mmol), pyridine (7.45 mL, 92.1 mmol) and propane- 1-sulfonyl chloride (8.25 mL, 73.6 mmol) in methylene chloride (10 mL) was added to the reaction mixture over the course of five minutes. The reaction mixture was stirred at room temperature for 14 hours. After removing the organic solvent under reduced pressure, saturated aqueous sodium bicarbonate (100 mL) was added followed by stirring for 10 minutes. The aqueous mixture was extracted twice with ethyl acetate (200 mL). The combined organic layers were washed with brine, dried over magnesium sulfate, filtered and concentrated. The crude product was purified using flash chromatography, eluting with 0- 30% ethyl acetate/heptanes to give methyl 6-fluoro-2-methoxy-3-(propylsulfonamido)- benzoate as an oil (4.914 g, 85%). 1H NMR (400 MHz, (CD3)2SO) δ = 9.27 (s, 1H), 7.48 (dd, J=9.1, 6.1, 1H), 7.09 (t, J=9.0, 1H), 3.89 (s, 3H), 3.85 - 3.74 (m, 3H), 3.29 (s, 15H).
[00276] Step D: A 100 mL round bottom flask was charged with methyl 6-fluoro-2- methoxy-3-(propylsulfonamido)benzoate (4.91 g, 16.1 mmol) dissolved in N,N- dimethylformamide (16 mL, 210 mmol) and was cooled over an ice/ brine bath. Sodium hydride (0.676 g, 16.9 mmol) was added in four portions. After the vigorous bubbling subsided, the reaction mixture was stirred for 1 hour at room temperature. The reaction mixture was cooled over an ice/brine bath, and p-methoxybenzyl chloride (2.646 g, 16.90 mmol) was added. The reaction was allowed to warm to room temperature over the next three hours and then quenched by adding a semi-saturated aqueous ammonium chloride solution (200 mL) at 0°C. After stirring at room temperature overnight, the aqueous layer was discarded, and the remaining oil washed with heptanes to remove the mineral oil. The residual oil was dissolved in ethyl acetate, dried over magnesium sulfate, filtered and concentrated to remove the ethyl acetate. The crude product was purified by flash chromatography (120 g column), using a gradient of 0-100% ethyl acetate: heptanes to give methyl 6-fluoro-2-methoxy-3-(N-(4-methoxybenzyl)propylsulfonamido)benzoate (3.71 g, 57%) as an oil. 1H NMR (400 MHz, (CD3)2SO) δ = 7.28 (dd, J=9.0, 6.3, 1H), 7.12 (d, J=8.7, 2H), 6.98 (t, J=8.9, 1H), 6.84 (dd, J=6.8, 4.8, 2H), 4.65 (s, 2H), 3.90 (d, J=7.6, 3H), 3.73 (s, 3H), 3.70 (s, 3H), 3.28 - 3.21 (m, 2H), 1.84 - 1.70 (m, 2H), 1.00 (q, J=7.2, 3H).
[00277] Step E: A 250 mL round bottom flask was charged with methyl 6-fluoro-2- methoxy-3-(N-(4-methoxybenzyl)propylsulfonamido)benzoate (4.42 g, 10.4 mmol) dissolved in tetrahydrofuran (70 mL, 900 mmol). 1M of sodium hydroxide in water (67.8 mL, 67.8 mmol) was added, and the mixture was stirred at 60°C for 48 hours. After cooling, the THF was removed under reduced pressure. The basic aqueous solution was diluted with water to a volume of 100 mL and then extracted once with ethyl acetate (200 mL). The aqueous layer was acidified with concentrated hydrochloric acid (5 mL) to a pH of 2 and extracted three times with ethyl acetate (100 mL). The combined organic layers were dried over magnesium sulfate, filtered and concentrated to afford 6-fluoro-2-methoxy-3-(N-(4- methoxybenzyl)propylsulfonamido)benzoic acid as a solid (4.253 g, 99%). !H NMR (400 MHz, (CD3)2SO) 6 = 13.86 (s, 1H), 7.19 (dd, J=8.9, 6.3, 1H), 7.12 (d, J=8.6, 2H), 6.93 (t, J=8.8, 1H), 6.83 (d, J=8.7, 2H), 4.65 (s, 2H), 3.80 (s, 3H), 3.70 (s, 3H), 3.27 - 3.19 (m, 2H), 1.78 (dd, J-15.3, 7.5, 2H), 1.01 (t, J=7.4, 3H).
[00278] Step F: Under a nitrogen atmosphere, a dry 100 mL round bottom, stir bar and reflux condenser were charged with 6-fluoro-2-methoxy-3-(N-(4-methoxybenzyl)- propylsulfonamido)benzoic acid (379 mg, 0.921 mmol) dissolved in 1,4-dioxane (10 mL, 83 mmol). Triethylamine (295.3 μί, 2.12 mmol) and then diphenylphosphonic azide (228.3 μί, 1.06 mmol) were added. The reaction mixture was stirred at room temperature for 3 hours and then heated to reflux for 1 hour. Water (10 mL, 36 mmol) was added to reaction mixture and heating to reflux continued for 2 hours. The reaction mixture was concentrated to remove the 1,4-dioxane. Residual material was stirred with a saturated aqueous solution of sodium bicarbonate for thirty minutes, and the aqueous layer was decanted and discarded. The residual oil was purified by flash chromatography (40 g column) using a gradient of 0- 100% ethyl acetate:heptanes to give N-(3-amino-4-fluoro-2-methoxyphenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (108 mg, 31%) as an oil. 1H NMR (400 MHz, (CD3)2SO) δ = 7.11 (d, J=8.7, 2H), 6.82 (d, J-8.7, 2H), 6.73 (dd, J=10.4, 8.9, 1H), 6.33 (dd, J=8.9, 5.8, 1H), 4.95 (s, 2H), 4.63 (s, 2H), 3.69 (s, 3H), 3.62 (s, 3H), 3.25 - 3.17 (m, 2H), 1.77 (dq, J=15.0, 7.4, 2H), 1.00 (t, J-7.4, 3H). MS m/z 383.2 [M+l].
Example AM
Figure imgf000072_0001
N-(3 -Amino-2-chloro-4-fluorophenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide
[00279] N-(3-Amino-2-chloro-4-fluorophenyl)propane-l -sulfonamide (75 g, 280 mmol) was dissolved in N,N-dimethylformamide (200 mL, 2000 mmol). A 60% sodium hydride suspension in mineral oil (6:4, sodium hydride:mineral oil, 11.85 g, 296 mmol) was added in multiple portions over a period of fifteen minutes. The reaction mixture was stirred at room temperature for 90 minutes and was then warmed to 40°C for two hours. This homogeneous mixture was cooled to 0°C, and p-methoxybenzyl chloride (40.03 mL, 295.25 mmol) was added over 5 minutes. The reaction was left to stir and warm to room temperature. After 14 hours, the reaction mixture was poured into a dilute ammonium chloride solution (1750 mL), and the water layer was decanted to leave an oil. This oil was triturated three times with water (2 L). The remaining product was transferred into a 1 L beaker, diluted with water (800 mL), sonicated for 30 minutes and then stirred at room temperature for 1 hour. The resulting solid was collected via filtration and dried by lyophilization to give N-(3-amino-2-chloro-4-fluorophenyl)-N-(4-methoxybenzyl)propane-l- sulfonamide (111.9 g, 99 %). 1H NMR (500 MHz, (CD3)2SO) 5 = 7.11 (d, J=8.6, 2H), 6.96 (dd, J=10.6, 8.8, 1H), 6.81 (t, J=5.7, 2H), 6.51 (dd, J=8.7, 5.1, 1H), 5.42 (s, 2H), 4.71 (d, J=14.4, 1H), 4.57 (d, J=14.4, 1H), 3.70 (s, 3H), 3.21 (td, J=6.7, 1.4, 2H), 1.77 (dd, J=15.3, 7.5, 2H), 1.00 (t, J=7.4, 3H). MS m/z 387.2 [M+l].
Example AN
Figure imgf000073_0001
7-Bromoquinoxalin-2( 1 HVone
[00280] Bromine (3.56 mL, 68.63 mmol) was slowly added to a solution of quinoxalin-2(lH)-one (10.03 g, 68.63 mmol) in 0.1M acetic acid (686.3 mL). The reaction mixture was stirred at ambient temperature for 1.5 hours. The resulting solids were collected by filtration and washed with hexanes to afford 7-bromoquinoxalin-2(lH)-one (4.02 g, 17.86 mmol, 26.0% yield). 1H NMR (400 MHz, (CD3)2SO) δ = 12.466 (s, 1H), 8.191 (s, 1H), 7.724-7.700 (d, 1H), 7.470-7.444 (m, 2H).
Example AO
Figure imgf000073_0002
N-(3-Amino-4-chloro-2-((triisopropylsilyl)ethvnyl)phenyl)propane-l -sulfonamide
[00281] Step A: 2-Chloro-l,3-dinitrobenzene (0.500 g, 2.47 mmol), Cul (0.0940 g, 0.494 mmol), P(t-Bu)3 (1.51 mL, 0.494 mmol) and ethynyltriisopropylsilane (0.658 mL, 2.96 mmol) were dissolved in 5:1 acetonitrile/TEA (10 mL). Nitrogen gas was bubbled through for 5 minutes, and PdCl2(MeCN)2 (0.0640 g, 0.247 mmol) was added. Nitrogen gas was bubbled though for another 10 minutes. The reaction was stirred at room temperature for 2 hours, diluted with EtOAc and filtered through Celite®. The filtrate was concentrated, dissolved in EtOAc, and washed with 0.1N HC1, water and brine, and dried over Na2S04 and concentrated. The product was purified by Biotage chromatography eluting with hexanes/EtOAc to give ((2,6-dinitrophenyl)ethynyl)triisopropylsilane (310 mg, 36%) as an oil. m/z (APCI-neg) M-l = 348.1.
[00282] Step B: ((2,6-Dinitrophenyl)ethynyl)triisopropylsilane (0.310 g, 0.890 mmol) was dissolved in 1:1 DCM/DMF (30 mL). SnCl2 dihydrate (10.0 g, 44.5 mmol) was added and stirred for one hour at room temperature. The reaction was poured into saturated aqueous NaHC03 (200 mL) giving a precipitate, which was stirred at room temperature for 30 minutes and filtered through Celite®. The layers were separated, the aqueous was extracted with DCM, and the combined organics washed with water (2 X) and brine, dried over Na2S04 and concentrated. The product was purified by Biotage chromatography eluting with hexanes/EtOAc to give 2-((triisopropylsilyl)ethynyl)benzene- 1,3 -diamine as an oil. 1H NMR (400 MHz, CDC13) δ 6.89 (t, 1H), 6.09 (d, 2H), 4.17 (br s, 4H), 1.14 (s, 21H). m/z (APCI- pos) M+l = 389.2.
[00283] Step C: 2-((Triisopropylsilyl)ethynyl)benzene-l,3-diamine (0.072 g, 0.249 mmol) was dissolved in THF (5 mL), and n-chlorosuccinimide (0.036 g, 0.286 mmol) was added and stirred at room temperature for one hour. The crude reaction was diluted with EtOAc, washed with water (3 X) and brine, and then dried over Na2S04 and concentrated. The product was purified by Biotage chromatography eluting with hexanes/DCM to give 4- chloro-2-((triisopropylsilyl)ethynyl)benzene- 1,3 -diamine (55 mg, 64% for two steps) as an oil. m/z (APCI-pos) M+l = 323.1, 325.2.
[00284] Step D: 4-Chloro-2-((triisopropylsilyl)ethynyl)benzene- 1,3 -diamine (0.0554 g, 0.172 mmol) was dissolved in 10:1 dichloroethane/pyridine (1 mL) and cooled to 0°C. Propane- 1-sulfonyl chloride (0.0193 mL, 0.172 mmol) was added, and the reaction was stirred at 50°C overnight. The reaction was concentrated, dissolved in EtOAc and washed with 0.1N HC1, water, and brine, dried over Na2S04 and concentrated. The product was purified by Biotage chromatography eluting with hexanes/DCM to give N-(3-amino-4- chloro-2-((triisopropylsilyl)ethynyl)phenyl)propane-l -sulfonamide (30 mg, 41%) as an oil. 1H NMR (400 MHz, CDC13) δ 7.17-7.19 (d, 1H), 6.93-6.95 (d, 1H), 6.89 (br s, 1H), 4.69 (br s, 2H), 3.04-3.08 (m, 2H), 1.77-1.87 (m, 2H), 1.14-1.16 (m, 21 H), 0.98-1.02 (t, 3H). m/z (APCI-neg) M-l = 427.2, 429.2.
Exam le 1
Figure imgf000074_0001
N-(4-Chloro-2-fluoro-3 -(3 -methyl-4-oxo-3 ,4-dihvdroquinazolin-6-ylamino)phenyl)propane-
1 -sulfonamide
[00285] 6-Bromo-3-methylquinazolin-4(3H)-one (0.100 g, 0.418 mmol), N-(3-amino- 4-chloro-2-fluorophenyl)propane-l -sulfonamide (0.112 g, 0.418 mmol), NaOtBu (0.121 g, 1.25 mmol), Binap-rac (0.0260 g, 0.0418 mmol) and Pd2dba3.CHCl3 (0.0216 g, 0.0209 mmol) were suspended in toluene (8 mL). Ar was bubbled through for 10 minutes, and the reaction was heated to 100°C for 16 hours under an Ar balloon. The next morning the reaction mixture was cooled to room temperature and diluted with ethyl acetate (100 mL) and water (30 mL). The pH of the aqueous layer was adjusted to about 6 with AcOH. The reaction mixture was filtered, and the filter cake was washed with ethyl acetate (50 mL). The filtrate was transferred to a separatory funnel, and the layers were separated. The aqueous layer was extracted with ethyl acetate (1 X 50 mL). The combined organics were dried, filtered and concentrated. The crude product was purified by column chromatography, eluting with DCM/ ethyl acetate (2:1), DCM/ ethyl acetate (1:1) to give N-(4-chloro-2-fluoro- 3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)propane- 1 -sulfonamide (0.020 g, 11%) as a solid. 1H NMR (400 MHz, (CD3)2SO) δ 9.83 (br s, 1H), 8.42 (br s, 1H), 8.16 (s, 1H), 7.53 (d, J=8.4 Hz, 1H), 7.42 (d, J=8.6 Hz, 1H), 7.32-7.24 (m, 2H), 7.15 (s, 1H), 3.44 (s, 3H), 3.12 (m, 2H), 1.73 (m, 2H), 0.96 (t, J=7.4 Hz, 3H). LC/MS: m/z 425.1, 427.1 [M+l].
Exam le 2
Figure imgf000075_0001
N-(2 ,4-Difluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)propane- 1 - sulfonamide
[00286] Step A: 6-Bromo-3-methylquinazolin-4(3H)-one (0.200 g, 0.837 mmol), N- (3-amino-2,4-difluorophenyl)-N-(4-methoxybenzyl)propane-l-sulfonamide (0.310 g, 0.837 mmol), NaOtBu (0.241 g, 2.51 mmol), and Binap-rac (0.0521 g, 0.0837 mmol) were taken up in toluene as a slurry in a sealed vial. Nitrogen gas was bubbled through for 5 minutes, and Pd2dba3 CHCI3 (0.0433 g, 0.0418 mmol) was added. Nitrogen gas was bubbled through for another 10 minutes, and the reaction was heated to 100°C overnight. The reaction was concentrated and partitioned between EtOAc and 0.1N HC1. The organic layer was washed with water and brine, dried over Na2S0 , filtered and concentrated. The crude product was purified by Biotage chromatography with 1% MeOH/EtOAc to give N-(2,4-difluoro-3-(3- methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)-N-(4-methoxybenzyl)propane- 1 - sulfonamide (28 mg, 6.3%) as a solid, m/z (APCI-pos) M+l = 429.2.
[00287] Step B: N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.010 g, 0.019 mmol) was dissolved in neat TFA with one drop of water and stirred at room temperature for 30 minutes. The reaction was concentrated, and the residual solid was taken up in EtOAc and washed with saturated NaHC03 and brine, dried over Na2S04, and concentrated. Biotage chromatography (hexanes/EtOAc) provided N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)propane-l -sulfonamide (5.6 mg, 72%) as a solid. lH NMR (400 MHz, (CD3)2SO) 6 9.68 (s, 1H), 8.46 (s, 1H), 8.16 (s, 1H), 7.53-7.56 (d, 1H), 7.27-7.30 (m, 1H), 7.20-7.23 (m, 2H), 3.45 (s, 3H), 3.06-3.10 (m, 2H), 1.72-1.78 (m, 2H), 0.95-0.99 (t, 3H); m/z (APCI-pos) M+l = 409.1.
Exam le 3
Figure imgf000076_0001
N-(2,4-Difluoro-3-(methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6- yl)arnino)phenyl)propane- 1 -sulfonamide
[00288] Step A: N-(2,4-Difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.015 g, 0.0284 mmol) was dissolved in DMF (1 mL) and cooled to 0°C. NaH (0.00170 g, 0.0426 mmol) was added and stirred for 10 minutes. Mel (0.00195 mL, 0.0312 mmol; d 2.275) was added and stirred for 10 minutes at room temperature. The reaction was partitioned between EtOAc and water. The organic layer was washed with water (2 X) and brine, dried over Na2S04 and concentrated to provide N-(2,4-difluoro-3-(methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6- yl)amino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide, which was used in the next step without further purification, m/z (APCI-pos) M+l = 543.2.
[00289] Step B: N-(2,4-Difluoro-3-(methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6- yl)amino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.0154 g, 0.0284 mmol) was dissolved in neat TFA with a few drops of water and stirred at room temperature for 30 minutes. The reaction was concentrated, and the residual solid was taken up in EtOAc and washed with saturated NaHC03 and brine, and dried over Na2S04 and concentrated. Biotage chromatography (hexanes/EtOAc) provided N-(2,4-difiuoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)propane-l -sulfonamide as an oil (7.7 mg, 64%). 1H NMR (400 MHz, (CD3)2SO) δ 9.73 (s, 1H), 8.18 (s, 1H), 7.54-7.56 (d, 1H), 7.41-7.47 (m, 1H), 7.27-7.32 (m, 1H), 7.21-7.22 (d, 1H), 7.13-7.16 (m, 1H), 3.46 (s, 3H), 3.33 (s, 3H), 3.07-3.11 (m, 2H), 1.70-1.76 (m, 2H), 0.93-0.96 (t, 3H); m/z (APCI-pos) M+l = 423.3.
xample 4
Figure imgf000077_0001
N-(2-CMoro-4-fluoro-3-(3-methyl-4-oxo-3^-dih^
1 -sulfonamide
[00290] N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.050 g, 28% yield) was prepared according to the general procedure of Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1-sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide. 1H NMR (400 MHz, (CD3)2SO) δ 9.54 (br s, 1H), 8.40 (br s, 1H), 8.15 (s, 1H), 7.53 (d, J=8.8 Hz, 1H), 7.37 (m, 2H), 7.25 (d, J=8.6 Hz, 1H), 7.14 (s, 1H), 3.44 (s, 3H), 3.11 (m, 2H), 1.77 (m, 2H), 0.98 (t, J=7.4 Hz, 3H). LC/MS: m/z 425.1, 427.1 [M+l].
Example 5
Figure imgf000077_0002
N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihvdroquinazolin-6-ylamino)phenyl)-l- cyclopropylmethanesulfonamide
[00291] N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-l-cyclopropylmethanesulfonamide (0.082 g, 48% yield) was prepared according to the general procedure of Example 1, substituting N-(2-chloro-4-fluoro-3-(3- methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)- 1 -cyclopropylmethanesulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide. 1H NMR (400 MHz, (CD3)2SO) δ 9.43 (br s, 1H), 8.34 (br s, 1H), 8.1 1 (s, 1H), 7.49 (d, J=8.6 Hz, 1H), 7.34 (m, 2H), 7.22 (m, 1H), 7.09 (s, 1H), 3.40 (s, 3H), 3.09 (d, J=7.1 Hz, 2H), 1.07 (m, 1H), 0.53 (m, 2H), 0.32 (m, 2H). LC/MS: m/z 437.1, 439.1 [M+l].
Example 6
Figure imgf000077_0003
N-(2-Chloro-4-fluoro-3 -(methyl(3 -methyl-4-oxo-3 ,4-dihvdroquinazolin-6- yl amino)phenyl)propane- 1 -sulfonamide
[00292] Step A: N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.45 g, 49% yield) was prepared according to the general procedure of Example 1, substituting N-(3-amino-2-chloro- 4-fluorophenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide for N-(3 -amino-4-chloro-2- fluorophenyl)propane-l -sulfonamide. LC/MS: m/z 545.2, 547.2 [M+l].
[00293] Step B: N-(2-Chloro-4-fluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide was prepared according to the general procedure of Example 3, Step A, substituting N-(2-chloro- 4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide. LC/MS : m/z 559.2, 561.2 [M+l].
[00294] Step C: N-(2-Chloro-4-fluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)propane-l -sulfonamide (0.025 g, 74% yield over 2 steps) was prepared according to the general procedure of Example 3, Step B substituting N- (2-chloro-4-fluoro-3 -(methyl(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6-yl)amino)phenyl)-N- (4-methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3 -(methyl(3 -methyl-4-oxo- 3,4-dihydroquinazolin-6-yl)amino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide. 1H NMR (400 MHz, (CD3)2SO) δ 9.56 (br s, 1H), 8.15 (s, 1H), 7.55-7.50 (m, 2H), 7.43 (m, 1H), 7.08 (d, J=3.0 Hz, 1H), 7.08 (m, 1H), 3.44 (s, 3H), 3.27 (s, 3H), 3.11 (m, 2H), 1.73 (m, 2H), 0.94 (t, J=7.4 Hz, 3H). LC/MS: m/z 439.1, 441.1 [M+l].
Exam le 7
Figure imgf000078_0001
N-(2-Chloro-4-fluoro-3 -(quinoxalin-6-ylamino)phenvDpropane- 1 -sulfonamide
[00295] N-(2-Chloro-4-fiuoro-3 -(quinoxalin-6-ylamino)phenyl)propane- 1 -sulfonamide (53%) was prepared according to Example 1, using 6-bromoquinoxaline and N-(3-amino-2- chloro-4-fluorophenyl)propane-l -sulfonamide. 1H NMR (400 MHz, (CD3)2SO) 5 9.57 (s, 1H), 8.72 (s, 1H), 8.70 (s, 1H), 8.61 (s, 1H), 7.91-7.93 (d, 1H), 7.42-7.48 (m, 3H), 6.73-6.76 (m, 1H), 3.12-3.16 (m, 2H), 1.75-1.80 (m, 2H), 0.96-1.00 (t, 3H); m/z (APCI-neg) M-l = 393.0, 395.0.
Example 8
Figure imgf000079_0001
N-(2-Chloro-4-fluoro-3 -(4-memoxyquinazolin-6-ylamino)phenyl)propane- 1 -sulfonamide
[00296] Step A: 6-Bromo-4-chloroquinazoline (1.00 g, 4.11 mmol) was treated with 0.5M NaOMe (82.1 mL, 41.1 mmol) at room temperature for 2 hours. The reaction mixture was concentrated, and the crude solids were washed with water and dried in vacuo to give 6- bromo-4-methoxyquinazoline (0.83 g, 84% yield) as a solid. LC/MS: m/z 239.0, 241.0 [M+l].
[00297] Step B: N-(2-Chloro-4-fluoro-3-(4-methoxyquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.100 g, 55% yield) was prepared according to the general procedure of Example 1, substituting 6-bromo-4-methoxyquinazoline for 6-bromo-3- methylquinazolin-4(3H)-one. 1H NMR (400 MHz, (CD3)2SO) δ 9.57 (br s, IH), 8.57 ( s, IH), 8.54 (s, IH), 7.78 (d, J=9.2 Hz, IH), 7.50 (m, IH), 7.40 (m, 2H), 6.91 (s, IH), 4.03 (s, 3H), 3.13 (m, 2H), 1.77 (m, 2H), 0.97 (t, J=7.0 Hz, 3H). LC/MS: m/z 425.1, 427.1 [M+l].
Exam le 9
Figure imgf000079_0002
N-(2-Chloro-3 -(4-ethoxyquinazolin-6-ylamino)-4-fluorophenyl)propane- 1 -sulfonamide
[00298] Step A: 6-Bromo-4-ethoxyquinazoline (0.61 g, 59% yield) was prepared according to the general procedure in Example 8, Step A, substituting NaOEt for NaOMe. LC/MS: m/z 253.1, 255.1 [M+l].
[00299] Step B: N-(2-Chloro-3-(4-ethoxyquinazolin-6-ylamino)-4- fluorophenyl)propane-l -sulfonamide (0.100 g, 55% yield) was prepared according to the general procedure in Example 1, substituting 6-bromo-4-ethoxyquinazoline for 6-bromo-3- methylquinazolin-4(3H)-one. 1H NMR (400 MHz, (CD3)2SO) δ 9.56 (br s, IH), 8.55 ( s, 2H), 7.76 (d, J=8.6 Hz, IH), 7.42-7.38 (m, 3H), 7.02 (s, IH), 4.53 (q, J=7.0 Hz, 2H), 3.13 (m, 2H), 1.77 (m, 2H), 1.37 (t, J=7.0 Hz, 3H), 0.97 (t, J=7.0 Hz, 3H). LC/MS: m/z 439.1, 441.1 [M+l].
Example 10
Figure imgf000080_0001
N-(2-Chloro-4-fluoro-3 -(3 -Π -methyl- 1 H-pyrazol-4- vD-4-oxo-3 ,4-dihydroquinazolin-6- ylamino)phenyQpropane- 1 -sulfonamide
[00300] Step A: HATU (0.329 g, 0.867 mmol), DBU (0.152 g, 1.000 mmol) and 1- methyl-lH-pyrazol-4-amine hydrochloride (0.0745 mL, 0.800 mmol) were added to 6- bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water. The concentrated organics were purified via silica gel chromatography eluting with 2% MeOH-EtOAc to yield 6-bromo-3-(l-methyl-lH-pyrazol-4-yl)quinazolin-4(3H)-one (0.018 g, 0.059 mmol, 18% yield).
[00301] Step B: N-(2-Chloro-4-fluoro-3-(3-(l-methyl-lH-pyrazol-4-yl)-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)propane-l -sulfonamide (0.005 g, 17% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro- 4-fluorophenyl)propane- 1 -sulfonamide for N-(3 -amino-4-chloro-2-fluorophenyl)propane- 1 - sulfonamide and 6-bromo-3-methylquinazolin-4(3H)-one for 6 -bromo-3-(l -methyl- 1H- pyrazol-4-yl)quinazolin-4(3H)-one. !H NMR (400 MHz, CD3OD) δ 9.22 (s, 1H), 8.12 (s, 1H), 7.81 (s, 1H), 7.67-7.71 (m, 1H), 7.51-7.56 (m, 1H), 7.43-7.47 (m, 1H), 7.34-7.36 (m, 1H), 7.24-7.30 (m, 1H), 3.98 (s, 3H), 3.09-3.15 (m, 2H), 1.81-1.91 (m, 2H), 1.01-1.06 (m, 3H). LC/MS: m/z 491.2 [M+l].
Example 11
Figure imgf000080_0002
N-(2,4-Difluoro-3 -(3 -( 1 -methyl- 1 H-pyrazol-4-yl)-4-oxo-3 ,4-dihydroquinazolin-6- ylamino)phenyl)propane- 1 -sulfonamide
[00302] N-(2,4-Difluoro-3-(3-(l-methyl-lH-pyrazol-4-yl)-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)propane-l -sulfonamide (0.006 g, 21% yield) was prepared according to the Example 10, Step B, substituting N-(3-amino-4-chloro-2- fluorophenyl)propane-l -sulfonamide for N-(3-amino-2,4-difIuorophenyl)propane-l - sulfonamide. 1H NMR (400 MHz, CD3OD) δ 8.20 (s, 1H), 8.07 (s, 1H), 7.79 (s, 1H), 7.59- 7.63 (m, 1H), 7.29-7.41 (m, 3H), 7.05-7.12 (m, 1H), 3.96 (s, 3H), 3.07-3.12 (m, 2H), 1.80- 1.91 (m, 2H), 1.00-1.05 (m, 3H). LC/MS: m/z 475.2 [M+l].
Example 12
Figure imgf000081_0001
N-(2-CMoro-4-fluoro-3-(4-oxo-3-phenyl-3,4-dihvo^oquinazolin-6-ylamino)phenyl)propane-
1 -sulfonamide
[00303] Step A: HATU (0.329 g, 0.867 mmol), DBU (0.152 g, 1.000 mmol) and aniline (0.0745 mL, 0.800 mmol) were added to 6-bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water. The concentrated organics were purified via silica gel chromatography eluting with 2% MeOH-EtOAc to yield 6-bromo- 3-phenylquinazolin-4(3H)-one (0.042 g, 0.139 mmol, 20.9% yield).
[00304] Step B: N-(2-Chloro-4-fluoro-3-(4-oxo-3-phenyl-3,4-dihydroquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.005 g, 7.4% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo- 3-methylquinazolin-4(3H)-one for 6-bromo-3-phenylquinazolin-4(3H)-one. 1H NMR (400 MHz, CD3OD) δ 8.09 (s, 1H), 7.61-7.64 (m, 1H), 7.43-7.58 (m, 6H), 7.34-7.38 (m, 1H), 7.30-7.32 (m, 1H), 7.18-7.24 (m, 1H), 3.07-3.13 (m, 2H), 1.80-1.90 (m, 2H), 0.99-1.04 (m, 3H). LC/MS: m/z 487.1 [M+l].
Example 13
Figure imgf000081_0002
N-(3-(3-Benzyl-4-oxo-3,4-dihydroquinazolin-6-ylamino)-2-chloro-4-fluorophenyl)propane-
1 -sulfonamide
[00305] Step A: HATU (0.329 g, 0.867 mmol), DBU (0.152 g, 1.000 mmol) and phenylmethanamine (0.0902 mL, 0.800 mmol) were added to 6-bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water. The concentrated organics were purified via silica gel chromatography eluting with 2% MeOH- EtOAc to yield 3-benzyl-6-bromoquinazolin-4(3H)-one (0.020 g, 0.0635 mmol, 9.5% yield).
[00306] Step B: N-(3-(3-Benzyl-4-oxo-3,4-dihydroquinazolin-6-ylamino)-2-chloro-4- fluorophenyl)propane-l -sulfonamide (0.020 g, 36% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo- 3-methylquinazolin-4(3H)-one for 3-benzyl-6-bromoquinazolin-4(3H)-one. 1H NMR (400 MHz, CD3OD) δ 8.43 (s, IH), 8.34 (s, IH), 7.54-7.58 (m, IH), 7.25-7.39 (m, 7H), 7.09-7.11 (m, IH), 5.14 (s, IH), 3.07-3.13 (m, 2H), 1.70-1.79 (m, 2H), 0.94-0.99 (m, 3H). LC/MS: m/z 501.2 [M+l].
Example 14
Figure imgf000082_0001
N-(2-Chloro-4-fluoro-3 -(4-(methylamino)quinazolin-6- ylamino)phenyl propane- 1 - sulfonamide
[00307] Step A: PyBOP (0.347 g, 0.667 mmol), DBU (0.152 g, 1.000 mmol), and methylamine (0.400 mL, 0.800 mmol) were added to 6-bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water. The concentrated organics were purified via trituration with DCM to provide 6-bromo-N-methylquinazolin-4-amine (0.055 g, 0.231 mmol, 35.7% yield).
[00308] Step B: N-(2-Chloro-4-fluoro-3-(4-(methylamino)quinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.003 g, 3.1% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo- 3-methylquinazolin-4(3H)-one for 6-bromo-N-methylquinazolin-4-amine. 1H NMR (400 MHz, CD3OD) δ 8.43 (s, IH), 8.34 (s, IH), 8.20-8.25 (br s, 2H), 7.60-7.64 (m, IH), 7.46-7.52 (m, 2H), 7.22-7.28 (m, IH), 7.02-7.05 (m, IH), 3.11 (s, 3H), 3.11-3.16 (m, 2H), 1.84-1.92 (m, 2H), 1.02-1.07 (m, 3H). LC/MS: m/z 424.1 [M+l].
Example 15
Figure imgf000082_0002
N-(2-Chloro-3 -(4-(dimethylamino)quinazolin-6-ylamino)-4-fluorophenyl)propane- 1 - sulfonamide
[00309] Step A: PyBOP (0.347 g, 0.667 mmol), DBU (0.152 g, 1.000 mmol), and dimethylamine (0.400 mL, 0.800 mmol) were added to 6-bromoquinazolin-4(3H)-one (0.150 g, 0.667 mmol) in MeCN (3.5 mL). The solution was aged for 25 hours at ambient temperature before it was diluted with EtOAc and washed with brine-water. The concentrated organics were purified via silica gel chromatography eluting with EtOAc to provide 6-bromo-N,N-dimethylquinazolin-4-amine (0.042 g, 0.167 mmol, 25% yield).
[00310] Step B: N-(2-Chloro-3-(4-(dimethylamino)quinazolin-6-ylamino)-4- fluorophenyl)propane-l -sulfonamide (0.005 g, 6.9% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo- 3-methylquinazolin-4(3H)-one for 6-bromo-N,N-dimethylquinazolin-4-amine. 1H NMR (400 MHz, CD3OD) 5 8.32 (s, 1H), 7.62-7.66 (m, 1H), 7.42-7.47 (m, 1H), 7.36-7.40 (m, 1H), 7.20-7.26 (m, 1H), 7.09-7.11 (m, 1H), 3.23 (s, 6H), 3.10-3.16 (m, 2H), 1.82-1.92 (m, 2H), 1.01-1.07 (m, 3H). LC/MS: m/z 438.2 [M+l].
Example 16
Figure imgf000083_0001
N-(2-Chloro-4-fluoro-3 -(4-(4-methylpiperazin- 1 -yl)quinazolin-6-ylamino)phenyl)propane- 1 - sulfonamide
[00311 ] N-(2-Chloro-4-fluoro-3 -(4-(4-methylpiperazin- 1 -yl)quinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.002 g, 2.5% yield) was prepared according to the general procedure in Example 14, substituting methylamine for methyl piperazine and substituting 6-bromo-N-methylquinazolin-4-amine for 6-bromo-4-(4-methylpiperazin-l- yl)quinazoline. 1H NMR (400 MHz, CD3OD) δ 8.71 (s, 1H), 7.80-7.83 (m, 1H), 7.62-7.66 (m, 1H), 7.46-7.52 (m, 1H), 7.29-7.34 (m, 1H), 6.97-6.99 (m, 1H), 3.29-3.32 (m, 8H), 3.18- 3.23 (m, 2H), 2.96 (s, 3H), 1.87-1.97 (m, 2H), 1.06-1.11 (m, 3H). LC/MS: m/z 493.2 [M+l].
Example 17
Figure imgf000084_0001
tert-Butyl 4-(6-(2-chloro-6-fluoro-3-(propylsulfonamido)phenylamino)quinazolin-4- vOpiperazine- 1 -carboxylate
[00312] tert-Butyl 4-(6-(2-cMoro-6-fluoro-3- ( ropylsulfonarnido)phenylamino)quinazolin-4-yl)piperazine- 1 -carboxylate sulfonamide (0.020 g, 27% yield) was prepared according to the general procedure in Example 14, substituting methylamine for tert-butyl piperazine-l-carboxylate and substituting 6-bromo-N- methylquinazolin-4-amine for tert-butyl 4-(6-bromoquinazolin-4-yl)piperazine-l -carboxylate. 1H NMR (400 MHz, CDC13) δ 8.68 (s, 1H), 7.84-7.87 (m, 1H), 7.47-7.53 (m, 1H), 7.34-7.38 (m, 1H), 7.15-7.20 (m, 1H), 6.97-7.0 (m, 1H), 6.67-6.71 (br s, 1H), 5.97-5.99 (br s, 1H), 3.54-3.61 (m, 8H), 3.10-3.15 (m, 2H), 1.85-1.95 (m, 2H), 1.50 (s, 9H), 1.04-1.09 (m, 3H). LC/MS: m/z 579.3 [M+l].
Example 18
Figure imgf000084_0002
N-(2-Chloro-4-fluoro-3 -(4-(piperazin- 1 -yl)quinazolin-6-ylamino phenyl)propane- 1 - sulfonamide
[00313] tert-Butyl 4-(6-(2-chloro-6-fluoro-3- (propylsulfonamido)phenylamino)quinazolin-4-yl)piperazine-l -carboxylate (0.020 g, 0.035 mmol) was taken up in DCM (1 mL) and cooled to 0°C. TFA (1 mL) was added, and after 1 hour at 0°C, the solution was concentrated under reduced pressure. The residue was then taken up in 1M HCl-dioxane (2 mL) and re-concentrated to provide N-(2-chloro-4-fluoro-3- (4-(piperazin- 1 -yl)quinazolin-6-ylamino)phenyl)propane- 1 -sulfonamide hydrochloride (0.015 g, 0.030 mmol, 90% yield). 1H NMR (400 MHz, CD3OD) δ 8.70 (s, 1H), 7.78-7.82 (m, 1H), 7.60-7.64 (m, 1H), 7.48-7.53 (m, 1H), 7.29-7.35 (m, 1H), 7.06-7.09 (m, 1H), 4.35-4.40 (m, 4H), 3.44-3.49 (m, 4H), 3.16-3.22 (m, 2H), 1.85-1.95 (m, 2H), 1.04-1.09 (m, 3H). LC/MS: m/z 47902 [M+l].
Example 19
Figure imgf000085_0001
N-(2-Chloro-4-fluoro-3 -(quinazolin-6-ylamino)phenyl)propane- 1 -sulfonamide
[00314] N-(2-Chloro-4-fluoro-3-(quinazolin-6-ylamino)phenyl)propane-l -sulfonamide (0.050 g, 26.5% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane-l -sulfonamide for N-(3-amino-4- chloro-2-fluorophenyl)propane-l -sulfonamide and 6-bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline. 1H NMR (400 MHz, CDC13) δ 9.18 (s, 1H), 9.21 (s, 1H), 7.96-7.99 (m, 1H), 7.54-7.59 (m, 1H), 7.49-7.54 (m, 1H), 7.17-7.23 (m, 1H), 6.93-6.96 (m, 1H), 6.74- 6.77 (br s, 1H), 5.99-6.02 (br s, 1H), 3.10-3.15 (m, 2H), 1.85-1.96 (m, 2H), 1.04-1.09 (m, 3H). LC/MS: m/z 395.1 [M+l].
Example 20
Figure imgf000085_0002
N-(2,4-Difluoro-3-(quinazolin-6-ylarnino)phenyl)propane-l-sulfonamide
[00315] Step A: N-(2,4-Difluoro-3-(quinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (0.050 g, 28% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3 -amino-2,4-difluorophenyl)-N-(4-methoxybenzyl)propane- 1 - sulfonamide and 6-bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline. LC/MS: m/z 499.2 [M+l].
[00316] Step B: N-(2,4-Difluoro-3-(quinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (0.010 g, 0.020 mmol) was taken up in TFA (1 mL) and warmed to 50°C for 30 minutes. Concentration under reduced pressure and trituration of the residue with DCM provided N-(2,4-difluoro-3-(quinazolin-6-ylamino)phenyl)propane-l- sulfonamide (0.004 g, 0.011 mmol, 53% yield). 1H NMR (400 MHz, CDC13) δ 9.21 (s, 1H), 9.18 (s, 1H), 7.96-8.00 (m, 1H), 7.50-7.55 (m, 1H), 7.37-7.43 (m, 1H), 7.04-7.10 (m, 1H), 6.99-7.01 (m, 1H), 6.43-6.47 (br s, 1H), 5.82-5.86 (br s, 1H), 3.10-3.15 (m, 2H), 1.86-1.96 (m, 2H), 1.05-1.09 (m, 3H). LC/MS: m/z 377.7 [M+l].
Example 21
Figure imgf000086_0001
N-(2^-Difluoro-3-(memyl(quinazolin-6-yl amm^
[00317] Step A: N-(2,4-Difluoro-3-(quinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (0.050 g, 28% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-chloro-4-fluorophenyl)propane- 1 -sulfonamide for N-(3 -amino-2,4-difluorophenyl)-N-(4-methoxybenzyl)propane- 1 - sulfonamide and 6-bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline. LC MS: m/z 499.2 [M+l].
[00318] Step B: NaH (0.00882 g, 0.221 mmol) was added to N-(2,4-difluoro-3- (quinazolin-6-ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.044 g, 0.0883 mmol) in DMF (1 mL) at ambient temperature. After 10 minutes, Mel (0.0275 mL, 0.441 mmol; d 2.275) was added and stirred for 1 hour. The suspension was diluted with EtOAc and washed with brine-water. The organics were concentrated under reduced pressure to afford N-(2,4-difluoro-3-(methyl(quinazolin-6-yl)amino)phenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (0.040 g, 0.0780 mmol, 88.4% yield).
[00319] Step C: N-(2,4-Difluoro-3-(methyl(quinazolin-6-yl)amino)phenyl)-N-(4- methoxybenzyl)propane-l -sulfonamide (0.010 g, 0.020 mmol) was taken up in TFA (1 mL) and warmed to 50°C for 30 minutes. Concentration under reduced pressure and trituration of the residue with DCM provided N-(2,4-difluoro-3-(methyl(quinazolin-6- yl)amino)phenyl)propane-l -sulfonamide (0.004 g, 0.010 mmol, 52% yield). 1H NMR (400 MHz, CDC13) δ 9.25 (s, 1H), 9.14 (s, 1H), 7.84-7.88 (m, 1H), 7.53-7.60 (m, 1H), 7.23-7.25 (m, 1H), 7.05-7.11 (m, 1H), 7.01-7.03 (m, 1H), 6.61-6.65 (br s, 1H), 3.44 (s, 3H), 3.09-3.15 (m, 2H), 1.88-1.95 (m, 2H), 1.03-1.09 (m, 3H). LC/MS: m/z 393.2 [M+l].
Example 22
Figure imgf000086_0002
N-(2-Chloro-4-fluoro-3 -(methyl(quinazolin-6- yl)amino)phenyl)propane- 1 -sulfonamide
[00320] N-(2-Chloro-4-fluoro-3 -(methyl(quinazolin-6-yl)amino)phenyl)propane- 1 - sulfonamide (0.004 g, 52% yield) was prepared according to the procedure in Example 21, substituting N-(3-amino-2,4-difluorophenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide for N-(3-amino-2-chloro-4-fluorophenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide. 1H NMR (400 MHz, CDC13) δ 9.26 (s, 1H), 9.13 (s, 1H), 7.85-7.88 (m, 1H), 7.70-7.75 (m, 1H), 7.20-7.25 (m, 1H), 7.12-7.16 (m, 1H), 6.94-6.97 (m, 1H), 6.77-6.80 (br s, 1H), 3.41 (s, 3H), 3.11-3.16 (m, 2H), 1.85-1.95 (m, 2H), 1.05-1.10 (m, 3H). LC/MS: m/z 409.1 [M+l].
Example 23
Figure imgf000087_0001
N-(2-C yano-3 -(quinazolin-6- ylamino)phenyl propane- 1 -sulfonamide
[00321] Step A: 2-Fluoro-6-(quinazolin-6-ylamino)benzonitrile (0.100 g, 26.4% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2- chloro-4-fluorophenyl)propane-l -sulfonamide for 2-amino-6-fluorobenzonitrile and 6- bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline. LC/MS: m/z 265.1 [M+l].
[00322] Step B: NaH (0.020 g, 0.50 mmol) was added to propane- 1 -sulfonamide (0.062 g, 0.50 mmol) in DMA (0.7 mL). After 20 minutes, a solution of 2-fluoro-6- (quinazolin-6-ylamino)benzonitrile (0.066 g, 0.25 mmol) in DMA (1 mL) was slowly added. The mixture was sealed and placed in a microwave for 6 hours at 210°C. The resulting solution was diluted with EtOAc and washed with NRjCl-brine-water. The concentrated residue was then partitioned between EtAOc and aqueous sodium bicarbonate. The aqueous was washed with EtOAc, then acidified to pH 6 and extracted with EtOAc. The organics were dried over sodium sulfate and concentrated under reduce pressure. The residue was purified via silica gel chromatography eluting with 1% MeOH-EtOAc to provide N-(2-cyano- 3-(quinazolin-6-ylamino)phenyl)propane-l -sulfonamide (0.010 g, 0.027 mmol, 11% yield). 1H NMR (400 MHz, CD3OD) δ 9.33 (s, 1H), 9.07 (s, 1H), 7.94-7.98 (m, 1H), 7.84-7.88 (m, 1H), 7.60-7.61 (m, 1H), 7.53-7.58 (m, 1H), 7.31-7.34 (m, 1H), 7.21-7.24 (m, 1H), 3.19-3.24 (m, 2H), 1.87-1.97 (m, 2H), 1.05-1.10 (m, 3H). LC/MS: m/z 368.1 [M+l].
Example 24
Figure imgf000087_0002
N-(2-Cyano-3 -(methyl(quinazolin-6-yl)amino)phenyl)propane- 1 -sulfonamide
[00323] Step A: 2-Fluoro-6-(quinazolin-6-ylamino)benzonitrile (0.100 g, 26.4% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2- chloro-4-fluorophenyl)propane-l -sulfonamide for 2-amino-6-fluorobenzonitrile and 6- bromo-3-methylquinazolin-4(3H)-one for 6-bromoquinazoline. LC/MS: m/z 265.1 [M+l].
[00324] Step B: NaH (0.00568 g, 0.142 mmol) was added to 2-fIuoro-6-(quinazolin-6- ylamino)benzonitrile (0.592 mL, 0.118 mmol) in DMF at 0°C. After 10 minutes at ambient temperature, Mel (0.0148 mL, 0.237 mmol) was added and stirred for 20 minutes. The suspension was diluted with EtOAc and washed with brine-water. The organics were concentrated under reduced pressure to afford 2-fluoro-6-(methyl(quinazolin-6- yl)amino)benzonitrile (0.035 g, 0.126 mmol, 106%).
[00325] Step C: NaH (0.010 g, 0.25 mmol) was added to propane- 1 -sulfonamide (0.031 g, 0.25 mmol) in DMA (0.5 mL). After 20 minutes, a solution of 2-fluoro-6- (methyl(quinazolin-6-yl)amino)benzonitrile (0.035 g, 0.13 mmol) in DMA (1 mL) was slowly added. The mixture was sealed and placed in a microwave for 6 hours at 210°C. The resulting solution was diluted with EtOAc and washed with NFLtCl-brine-water. The concentrated residue was then partitioned between EtAOc and aqueous sodium bicarbonate. The aqueous was washed with EtOAc, then acidified to pH 6 and extracted with EtOAc. The organics were dried over sodium sulfate and concentrated under reduced pressure. The residue was purified via silica gel chromatography eluting with 1% MeOH-EtOAc to provide N-(2-cyano-3-(methyl(quinazolin-6-yl)amino)phenyl)propane-l -sulfonamide (0.005 g, 0.013 mmol, 10% yield). 1H NMR (400 MHz, CD3OD) δ 9.35 (s, 1H), 9.02 (s, 1H), 7.83-7.86 (m, 1H), 7.69-7.74 (m, 1H), 7.49-7.54 (m, 2H), 7.34-7.36 (m, 1H), 7.27-7.31 (m, 2H), 3.53 (s, 3H), 3.20-3.25 (m, 2H), 1.85-1.95 (m, 2H), 1.04-1.09 (m, 3H). LC/MS: m/z 382.3 [M+l].
Example 25
O CN
N-(2-Cvano-4-fluoro-3-(3-methyl-4-oxo-3,4-dihvdroquinazolin-6-yloxy phenyl)propane-l- sulfonamide
[00326] Step A: 60% Sodium hydride (0.0522 g, 1.31 mmol) was slowly added to 6- hydroxy-3-methylquinazolin-4(3H)-one (0.2 g, 1.14 mmol; Maillard, Jacques, et al. "(3H)- Quinazolin-4-one derivatives with antiinflammatory activity. II. Derivatives substituted in the aromatic nucleus, and related compounds." Chim. Ther. 2(4) (1967): 231-9) in DMF (4 mL) cooled in ice. When gas evolution ceased, the mixture was stirred at room temperature for 15 minutes, then cooled in ice, and a solution of 2,3,6-trifluorobenzonitrile (0.178 g, 1.14 mmol) in DMF (1 mL) was added. The mixture was allowed to stir at ambient temperature. After 1 hour, the reaction mixture was diluted with water, and the resulting solid was collected by vacuum filtration, washed with water and a small amount of hexane. Ή NMR and LC/MS were consistent with the desired 3,6-difluoro-2-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yloxy)benzonitrile (0.294 g, 0.939 mmol, 82.7% yield). Ή NMR (400 MHz, CDC13) δ 8.01 (s, 1H), 7.77 (d, 1H), 7.55-7.61 (m, 2H), 7.41-7.48 (m, 1H), 7.09-7.14 (m, 1H), 3.58 (s, 3H). LC/MS: m/z 314.1 [M+l].
[00327] Step B: 60% Sodium hydride (0.0287 g, 0.718 mmol) was slowly added to propane- 1 -sulfonamide (0.0826 g, 0.670 mmol) in NMP (5 mL) cooled in ice. After addition, the mixture was stirred at 50°C for 30 minutes. 3,6-Difluoro-2-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yloxy)benzonitrile (0.10 g, 0.319 mmol; Step A) was added to the mixture. The reaction was stirred at 120°C overnight. The cooled reaction mixture was partitioned between 0.1M NaOH and EtOAc. The aqueous layer was washed with EtOAc, acidified to pH 4 with 6M HC1, and extracted with 2 portions of EtOAc. The combined EtOAc extracts were washed with 3 portions water, brine, dried over MgS04, filtered, and evaporated to yield a solid. This was absorbed on silica gel, dry-loaded on a 10 g Biotage SNAP column, and chromatographed with EtOAc. Fractions 16-24 contained solid. !H NMR and LC/MS were consistent with the desired N-(2-cyano-4-fluoro-3-(3-methyl-4-oxo- 3, 4-dihydroquinazolin-6-yloxy)phenyl)propane-l -sulfonamide (0.0136 g, 0.0327 mmol, 10.2% yield). 1H NMR (400 MHz, CD3OD) δ 8.27 (s, 1H), 7.77 (d, 1H), 7.62-7.67 (m, 2H), 7.54-7.57 (m, 2H), 3.58 (s, 3H), 3.19-3.23 (m, 2H), 1.86-1.96 (m, 2H), 1.07 (t, 3H). LC/MS: m/z AM 2 [M+l].
Exam le 26
Figure imgf000089_0001
N-(2-Cvano-3-(3-methyl-4-oxo-3,4-dihvdroquinazolin-6-yloxy)phenyl)propane-l- sulfonamide
[00328] Step A: 60% Sodium hydride (0.0522 g, 1.31 mmol) was slowly added to 6- hydroxy-3-methylquinazolin-4(3H)-one (0.200 g, 1.14 mmol) in DMF (3 mL) cooled in ice. When gas evolution ceased, the mixture was stirred 15 minutes at room temperature, then 2,6-difluorobenzonitrile (0.158 g, 1.14 mmol) was added, and the mixture was stirred at 80°C. After 1 hour, the reaction mixture was allowed to cool and was diluted with water. The resulting solid was collected by vacuum filtration, washed with water and some hexane. 1H NMR and LC/MS were consistent with the desired 2-fluoro-6-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yloxy)benzonitrile (0.297 g, 1.01 mmol, 88.6% yield). Ή NMR (400 MHz, CDC13) δ 8.05 (s, 1H), 7.92 (d, 1H), 7.80 (d, 1H), 7.48-7.56 (m, 2H), 6.98 (t, 1H), 6.72 (d, 1H), 3.61 (s, 3H). LC/MS: m/z 296.1 [M+l].
[00329] Step B: 60% Sodium hydride (0.0291 g, 0.728 mmol) was slowly added to propane- 1 -sulfonamide (0.0876 g, 0.711 mmol) in NMP (5 mL) cooled in ice. After addition, the mixture was stirred at 50°C for 30 minutes. 2-Fluoro-6-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yloxy)benzonitrile (0.10 g, 0.339 mmol) was added to the mixture. The reaction mixture was stirred at 140°C overnight, then at 180°C for 8 hours. The cooled reaction mixture was partitioned between 0.5M NaOH and EtOAc. The aqueous layer was washed with EtOAc, acidified to pH 5 with 6M HC1, and extracted with 2 portions of EtOAc. The combined EtOAc extracts were washed with twice with brine, dried over MgS04, filtered, and evaporated to yield solid (0.09 g). This was absorbed on silica gel, dry-loaded on a 10 g Biotage SNAP column, and chromatographed with 10:1 DCM:MeOH to afford N- (2-cyano-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6-yloxy)phenyl)propane-l-sulfonamide (0.0166 g, 0.0417 mmol, 12.3% yield) as a solid. 1H NMR (400 MHz, CD3OD) δ 8.29 (s, 1H), 7.80 (d, 1H), 7.79 (s, 1H), 7.58-7.65 (m, 2H), 7.40-7.47 (m, 2H), 3.59 (s, 3H), 3.21-3.27 (m, 2H), 1.82-1.86 (m, 2H), 1.08 (t, 3H). LC MS: m/z 399.2 [M+l].
Exam le 27
Figure imgf000090_0001
N-(4-Chloro-2-cyano-3 -(3 -methyl-4-oxo-3 ,4-dihvdroquinazolin-6-yloxy)phenyl)propane- 1 - sulfonamide
[00330] Step A: 60% Sodium hydride (0.0522 g, 1.31 mmol) was slowly added to 6- hydroxy-3-methylquinazolin-4(3H)-one (0.200 g, 1.14 mmol) in DMF (4 mL). When gas evolution ceased, a solution of 3-chloro-2,6-difluorobenzonitrile (0.197 g, 1.14 mmol) in DMF (1 mL) was added, and the mixture was stirred at ambient temperature. After 30 minutes, the reaction mixture was diluted with water, stirred for 20 minutes, and the resulting solid was collected by vacuum filtration, washed with water then a small amount of hexane, and dried under high vacuum to afford 3-chloro-6-fluoro-2-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yloxy)benzonitrile (0.279 g, 0.846 mmol, 74.5% yield). 1H NMR (400 MHz, CDC13) δ 8.01 (s, 1H), 7.78 (d, 1H), 7.72 (q, 1H), 7.56 (dd, 1H), 7.45 (d, 1H), 7.16 (dd, 1H), 3.58 (s, 3H). LC/MS: m/z 330.1 [M+l].
[00331] Step B: 60% Sodium hydride (0.0255 g, 0.637 mmol) was added to propane-
1 - sulfonamide (0.0785 g, 0.637 mmol) in NMP (4 mL) in a vial cooled in ice. After gas evolution ceased, the mixture was stirred at 40°C for 15 minutes, and then 3-chloro-6-fluoro-
2- (3-methyl-4-oxo-3,4-dihydroquinazolin-6-yloxy)benzonitrile (0.100 g, 0.303 mmol) was added. The mixture was stirred and heated at 120°C for 4 hours. The cooled reaction mixture was partitioned between 0.5M NaOH and EtOAc. The aqueous layer was washed with EtOAc, acidified to pH 5 with 6 M HCl, and extracted with 2 portions of EtOAc. The combined EtOAc extracts were washed twice with brine, dried over MgS04, filtered, and evaporated to yield glass (0.21 g). This was chromatographed on a 10 g Biotage SNAP column with 20:1 EtOAc:MeOH to afford impure desired product (0.0098 g). This was triturated with Et20 to provide N-(4-chloro-2-cyano-3-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yloxy)phenyl)propane-l -sulfonamide (0.0063 g, 0.0146 mmol, 4.80% yield). 1H NMR (400 MHz, CD3OD) δ 8.26 (s, 1H), 7.83 (d, 1H), 7.77 (d, 1H), 7.59 (dd, 1H), 7.42 (d, 1H), 3.57 (s, 3H), 3.20-3.24 (m, 2H), 1.85-1.94 (m, 2H), 1.06 (t, 3H). LC/MS: m/z 433.1 [M+l].
Ex mple 28
Figure imgf000091_0001
N-(4-Chloro-2-cyano-3 -(3 -methyl-4-oxo-3 ,4-dihvdroquinazolin-6-ylamino)phenyl)propane-
1 -sulfonamide
[00332] N-(4-Chloro-2-cyano-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.024 g, 22% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-4-chloro-2-cyanophenyl)propane- 1-sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l -sulfonamide. 1H NMR
(400 MHz, CD3OD) δ 8.16 (s, 1H), 7.76 (d, 1H), 7.61 (d, 1H), 7.39-7.44 (m, 3H), 3.57 (s, 3H), 3.18-3.22 (m, 2H), 1.84-1.94 (m, 2H), 1.05 (t, 3H). LC/MS: m/z 432.1 [M+l].
Example 29
Figure imgf000092_0001
N-(2-Cvano-3-(3-methyl-4-oxo-3^-dihydroquinazolin-6-ylarnino phenyl)propane-l- sulfonamide
[00333] N-(2-Cyano-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (0.0295 g, 18.7% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2-cyanophenyl)propane-l- sulfonamide for N-(3 -amino-4-chloro-2-fluorophenyl)propane- 1 -sulfonamide . 1 H NMR (400 MHz, CD3OD) δ 8.20 (s, 1H), 7.89 (d, 1H), 7.60-7.67 (m, 2H), 7.49 (dd, 1H), 7.21 (d, 1H), 7.15 (d, 1H), 3.59 (s, 3H), 3.18-3.22 (m, 2H), 1.87-1.96 (m, 2H), 1.07 (t, 3H). LC/MS: m/z 398.2 [M+l].
Exam le 30
Figure imgf000092_0002
N-(4-Chloro-2-fluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- ylammo)phenyl)dimethylsulfamide
[00334] Potassium carbonate (0.035 g, 0.25 mmol) and dimethylsulfarnoyl chloride (0.014 mL, 0.13 mmol) were added to N-(4-chloro-2-fiuoro-3-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)propane-l -sulfonamide (0.036 g, 0.085 mmol) in DMF (0.75 mL). The suspension was aged at ambient temperature for 48 hours before addition of 2M NaOH (1 mL). After an additional 2 hours, the solution was diluted with EtOAc and washed with brine-water. The organics were dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was triturated with DCM then EtOAc to provide N-(4-chloro-2-fluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- ylamino)phenyl)dimethylsulfamide (0.010 g, 0.023 mmol, 28% yield). 1H NMR (400 MHz, CD3OD) δ 8.12 (s, 1H), 7.55-7.58 (m, 1H), 7.40-7.45 (m, 1H), 7.29-7.36 (m, 2H), 7.24-7.27 (m, 1H), 3.55 (s, 3H), 2.81 (s, 6H). LC/MS: m/z 426.1, [M+l].
Exam le 31
Figure imgf000092_0003
N-(4-Chloro-2-fluoro-3-(3-methyl-4-oxo-3,4-dihvdroquinazolin-6- ylamino phenyl)pyrrolidine- 1 -sulfonamide
[00335] Potassium carbonate (0.037 g, 0.27 mmol) and pyrrolidine- 1-sulfonyl chloride (0.018 mL, 0.13 mmol) were added to N-(4-chloro-2-fluoro-3-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)propane-l -sulfonamide (0.038 g, 0.089 mmol) in DMF (0.75 mL). The suspension was aged at ambient temperature for 48 hours before addition of 2M NaOH (1 mL). After an additional 2 hours, the solution was diluted with EtOAc and washed with brine-water. The organics were dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was triturated with DCM then EtOAc to provide N-(4-chloro-2-fluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- ylamino)phenyl)pyrrolidine-l -sulfonamide (0.006 g, 0.013 mmol, 15% yield). 1H NMR (400 MHz, CD3OD) δ 8.11 (s, 1H), 7.54-7.57 (m, 1H), 7.42-7.47 (m, 1H), 7.29-7.35 (m, 2H), 7.24-7.27 (m, 1H), 3.55 (s, 3H), 2.27-2.30 (m, 4H), 1.83-1.87 (m, 4H). LC/MS: m/z 452.1, [M+l].
Exam le 32
Figure imgf000093_0001
N-(2,4-Difluoro-3 -(3 -methyl-4-oxo-3 ,4-dihvdroquinazolin-6-ylamino)phenyl)-3 - fluoropropane- 1 -sulfonamide
[00336] Step A: N-(2,4-Difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-3-fluoro-N-(4-methoxybenzyl)propane-l -sulfonamide (32%) was prepared according to the general procedure of Example 2, Step A, substituting N-(3-amino-2,4- difluorophenyl)-3 -fluoro-N-(4-methoxybenzyl)propane- 1 -sulfonamide for N-(3 -amino-2,4- difluorophenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide, m/z (APCI-pos) M+l = 547.1.
[00337] Step B: N-(2,4-Difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-3 -fluoropropane- 1 -sulfonamide (74%) was prepared according to the general procedure of Example 2, Step B, using N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4- dihydroquinazolin-6-ylamino)phenyl)-3 -fluoro-N-(4-methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6-ylamino)phenyl)-3-fluoro-N- (4-methoxybenzyl)propane-l-sulfonamide. 1H NMR (400 MHz, (CD3)2SO) δ 9.82 (s, 1H), 8.48 (s, 1H), 8.17 (s, 1H), 7.53-7.56 (d, 1H), 7.22-7.30 (m, 4H), 4.59-4.62 (t, 1H), 4.47-4.50 (t, 1H), 3.45 (s, 3H), 3.20-3.24 (m, 2H), 2.05-2.18 (m, 2H); m/z (APCI-neg) M-l = 425.1.
Example 33
Figure imgf000094_0001
N-(2,4-Difluoro-3-(methylQ-meth^
fluoropropane- 1 -sulfonamide
[00338] Step A: N-(2,4-Difluoro-3-(methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6- yl)amino)phenyl)-3 -fluoro-N-(4-methoxybenzyl)propane- 1 -sulfonamide ( 100%) was prepared according to the general procedure of Example 3, Step A, substituting N-(2,4- difluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)-3 -fluoro-N-(4- methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3 -(3-methyl-4-oxo-3 ,4- dihydroquinazolin-6-ylamino)phenyl)-N-(4-methoxybenzyl)propane-l-sulfonamide.
[00339] Step B: N-(2,4-Difluoro-3-(methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6- yl)amino)phenyl)-3 -fluoropropane- 1 -sulfonamide (33%) was prepared according to the general procedure of Example 3, Step B, substituting N-(2,4-difluoro-3-(methyl(3-methyl-4- oxo-3,4-dihydroquinazolin-6-yl)amino)phenyl)-3-fluoro-N-(4-methoxybenzyl)propane-l- sulfonamide for N-(2,4-difluoro-3 -(methyl(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- yl)amino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide. 1H NMR (400 MHz, (CD3)2SO) δ 9.88 (s, 1H), 8.18 (s, 1H), 7.42-7.48 (m, 1H), 7.27-7.33 (m, 1H), 7.22-7.23 (d, 1H), 7.13-7.16 (m, 1H), 4.57-4.59 (t, 1H), 4.45-4.48 (t, 1H), 3.46 (s, 3H), 3.33 (s, 3H), 3.20- 3.24 (m, 2H), 2.03-2.16 (m, 2H); m/z (APCI-neg) M-l = 439.1.
mple 34
Figure imgf000094_0002
N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihvdroquinazolin-6-ylamino)phenyl)-3- fluoropropane- 1 -sulfonamide
[00340] Step A: N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-3 -fluoropropane- 1 -sulfonamide (44%) was prepared according to the general procedure of Example 4, Step A, substituting N-(3-amino-2-chloro-4-fluorophenyl)- 3 -fluoro-N-(4-methoxybenzyl)propane- 1 -sulfonamide for N-(3 -amino-2,4-difluorophenyl)- N-(4-methoxybenzyl)propane- 1 -sulfonamide.
[00341] Step B: N-(2-Chloro-4-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-3 -fluoropropane- 1 -sulfonamide (37%) was prepared according to the general procedure of Example 4, Step B, substituting N-(2-chloro-4-fluoro-3-(3-methyl-4- oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)-3 -fluoro-N-(4-methoxybenzyl)propane- 1 - sulfonamide for N-(2,4-difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)-3-fluoro-N-(4-methoxybenzyl)propane-l -sulfonamide. !H NMR (400 MHz, (CD3)2SO) δ 9.71 (s, 1H), 8.42 (s, 1H), 8.15 (s, 1H), 7.52-7.55 (d, 1H), 7.37-7.39 (m, 2H), 7.24-7.27 (m, 1H), 7.14 (s, 1H) 4.60-4.63 (t, 1H), 4.48-4.51 (t, 1H), 3.45 (s, 3H), 3.23- 3.27 (m, 2H), 2.08-2.21 (m, 2H); m/z (APCI-pos) M+l = 441.0, 443.0.
Example 35
Figure imgf000095_0001
N-(4-Chloro-2-fluoro-3 -(methyl(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- yl amino)phenyl)propane- 1 -sulfonamide
[00342] Step A: N-(4-Chloro-2-fluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide ( 100%) was prepared according to the general procedure of Example 3, Step A, substituting N-(4- chloro-2-fluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)-N-(4- methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3-(3 -methyl-4-oxo-3 ,4- dihydroquinazolin-6-ylammo)phenyl)-3-fluoro-N-(4-methoxybenzyl)propane-l-sulfonamide.
[00343] Step B: N-(4-Chloro-2-fluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)propane-l -sulfonamide (71%) was prepared according to the general procedure of Example 3, Step B, substituting N-(4-chloro-2-fluoro-3- (methyl(3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)phenyl)-N-(4- methoxybenzyl)propane- 1 -sulfonamide for N-(2,4-difluoro-3-(methyl(3-methyl-4-oxo-3,4- dihydroquinazolin-6-yl)amino)phenyl)-N-(4-methoxybenzyl)propane- 1 -sulfonamide. !H NMR (400 MHz, (CD3)2SO) δ 9.90 (s, 1H), 8.17 (s, 1H), 7.53-7.55 (d, 1H), 7.46-7.50 (m, 2H), 7.12-7.13 (d, 1H), 7.03-7.06 (m, 1H), 3.46 (s, 3H), 3.30 (s, 3H), 3.11-3.15 (m, 2H), 1.66-1.76 (m, 2H), 0.92-0.95 (t, 3H); m/z (APCI-pos) M+l = 439.9.
Exam le 36
Figure imgf000095_0002
N-(4-C oro-3-(5-chloro-3-methyl-4-oxo-3,4-dihydroquinazolin-6-ylamino)-2- fluorophenvDpropane- 1 -sulfonamide
[00344] Step A: N-(4-Chloro-2-fluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6 ylamino)phenyl)-N-(4-methoxybenzyl)propane-l -sulfonamide (0.0235 g, 0.0431 mmol) was dissolved in THF. N-Chlorosuccinimide (0.00576 g, 0.0431 mmol) was added and stirred at room temperature for 3 days. The reaction was concentrated and partitioned between EtOAc and water. The organic layer was washed with water and brine, dried over Na2S04, filtered and concentrated. The crude product was purified by Biotage chromatography eluting with DCM/acetone to give N-benzyl-N-(4-chloro-3-(5-chloro-3-methyl-4-oxo-3,4- dihydroquinazolin-6-ylamino)-2-fluorophenyl)propane-l -sulfonamide (0.010 g, 40%) as an oil.
[00345] Step B: N-Benzyl-N-(4-chloro-3-(5-chloro-3-methyl-4-oxo-3,4- dihydroqumazolin-6-ylamino)-2-fluorophenyl)propane-l -sulfonamide was dissolved in neat TFA with one drop of water and stirred at room temperature for 30 minutes. The reaction was concentrated, and the residual solid was taken up in EtOAc and washed with saturated NaHC03 and brine, dried over Na2S04, and concentrated. Biotage chromatography (hexanes/EtO Ac) provided N-(4-chloro-3 -(5 -chloro-3 -methyl-4-oxo-3 ,4-dihydroquinazolin- 6-ylamino)-2-fiuorophenyl)propane-l -sulfonamide (0.0056g, 71%) as a solid. 1H NMR (400 MHz, CDC13) δ 7.95 (s, 1H), 7.51-7.49 (d, 1H), 7.41-7.45 (t, 1H), 7.29-7.32 (m, 1H), 6.96- 6.99 (m, 1H), 6.76 (br s, 1H), 6.47 (br s, 1H), 3.58 (s, 3H), 3.10-3.14 (m, 2H), 1.84-1.93 (m, 2H), 1.04-1.08 (t, 3H); m/z (APCI-neg) M-l = 459.3, 461.3.
Example 37
Figure imgf000096_0001
N-(2-CMoro-3-(5-cMoro-3-methyl-4-oxo-3^-dihvdroquinazolin-6-ylamino -4-fluorophenyl)-
3-fluoropropane- 1 -sulfonamide
[00346] N-(2-Chloro-4-fluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6- ylamino)phenyl)-3-fluoropropane-l -sulfonamide (0.0207 g, 0.0467 mmol) was dissolved in THF. N-Chlorosuccinimide (0.00624 g, 0.0467 mmol) was added, and the reaction was stirred at room temperature overnight. The reaction was concentrated and partitioned between EtOAc and water. The organic layer was washed with water and brine, dried over Na2S04, filtered and concentrated. The crude product was purified by Biotage chromatography eluting with DCM/acetone to give N-(2-chloro-3-(5-chloro-3-methyl-4-oxo- 3 ,4-dihydroquinazolin-6-ylamino)-4-fluorophenyl)-3-fluoropropane-l -sulfonamide (0.0131 g, 59%) as a solid. 1H NMR (400 MHz, CDC13) δ 7.94 (s, 1H), 7.54-7.58 (dd, 1H), 7.49-7.51 (d, 1H), 7.16-7.21 (t, 1H), 6.96-6.98 (dd, 1H), 6.86 (br s, 1H), 6.48 (br s, 1H), 4.60-4.63 (t, 2H), 4.49-4.51 (t, 2H), 3.57 (s, 3H), 3.26-3.30 (m, 2H), 2.19-2.32 (m, 2H); m/z (APCI-neg) M-l = 475.0, 477.0.
Example 38
Figure imgf000097_0001
N-(2-CMoro-4-fluoro-3-(3-(methylamino quinoxalin-6-ylamino)phenyl)propane-l- sulfonamide
[00347] Step A: 7-Bromoquinoxalin-2(lH)-one (145.0 mg, 0.644 mmol) was dissolved in 0.1M MeCN (6.4 mL) and treated sequentially with 2.0M methylamine in tetrahydrofuran (354.4 μί, 0.709 mmol), l,8-diazabicyclo[5.4.0]undec-7-ene (115.6 μί, 0.773 mmol), and benzotriazol-l-yloxytris(dimethylamino)phosphonium hexafluorophosphate (342.0 mg, 0.773 mmol). The reaction mixture was stirred at ambient temperature for 16 hours and then concentrated in vacuo. Biotage chromatography (hexanes/EtOAc) provided 7-bromo-N-methylquinoxalin-2-amine (31.4 mg, 0.132 mmol, 20.5% yield). LC/MS: m/z 240.0 [M+l].
[00348] Step B: N-(2-Chloro-4-fluoro-3-(3-(methylamino)quinoxalin-6- ylamino)phenyl)propane-l -sulfonamide (10.0 mg, 0.024 mmol, 17.9% yield) was prepared according to the general procedure in Example 1 , substituting 7-bromo-N-methylquinoxalin- 2-amine for 6-bromo-3-methylquinazolin-4(3H)-one. 1H NMR (400 MHz, (CD3)2SO) δ = 9.548 (s, 1H), 8.258 (s, 1H), 7.970 (s, 1H), 7.563-7.541 (d, 1H), 7.395-7.344 (m, 3H), 6.907- 6.880 (d, 1H), 6.352 (s, 1H), 3.145-3.107 (t, 2H), 2.825-2.813 (d, 3H), 1.799-1.742 (m, 2H), 1.000-0.963 (t, 3H). LC/MS: m/z 424.1, 426.1 [M+l].
Exam le 39
Figure imgf000097_0002
N-(2-Chloro-4-fluoro-3-(3-methoxyquinoxalin-6-ylamino phenvnpropane-l-sulfonamide
[00349] Step A: 7-Bromoquinoxalin-2(lH)-one (313.1 mg, 1.391 mmol) was dissolved in 0.15M DMF (9.2 mL) and treated with potassium carbonate (288.4 mg, 2.087 mmol) and iodomethane (95.5 iL, 1.530 mmol). The reaction mixture was stirred at ambient temperature for 30 minutes. The reaction mixture was then diluted with water and extracted with EtOAc (2 X). The organics were washed with water (3 X) and brine (1 X), dried over Na2S04, filtered and concentrated. Biotage chromatography (hexanes/EtOAc) provided 7- bromo-2-methoxyquinoxaline (25.2 mg, 0.105 mmol, 7.6% yield). 1H NMR (400 MHz, (CD3)2SO) δ = 8.642 (s, 1H), 8.070-8.064 (d, 1H), 7.971-7.950 (d, 1H), 7.801-7.774 (dd, 1H), 4.045 (s, 3H).
[00350] Step B: N-(2-Chloro-4-fluoro-3-(3-methoxyquinoxalin-6- ylamino)phenyl)propane-l -sulfonamide (12.1 mg, 0.029 mmol, 27.0% yield) was prepared according to the general procedure in Example 1, substituting 7-bromo-2- methoxyquinoxaline for 6-bromo-3-methylquinazolin-4(3H)-one. ]H NMR (400 MHz, (CD3)2SO) δ = 9.583 (s, 1H), 8.590 (s, 1H), 8.250 (s, 1H), 7.822-7.799 (d, 1H), 7.459-7.389 (m, 2H), 7.231-7.203 (dd, 1H), 6.541 (s, 1H), 3.935 (s, 3H), 3.157-3.119 (t, 2H), 1.824-1.729 (m, 2H), 1.004-0.967 (t, 3H). LC MS: m/z 425.1, 427.1 [M+l].
Example 40
Figure imgf000098_0001
N-f 2-Chloro-4-fluoro-3 -(4-methyl-3 -oxo-3 ,4-dihvdroquinoxalin-6-ylamino)phenyl)propane-
1 -sulfonamide
[00351] Step A: 7-Bromoquinoxalin-2(lH)-one (313.1 mg, 1.391 mmol) was dissolved in 0.15M DMF (9.2 mL) and treated with potassium carbonate (288.4 mg, 2.087 mmol) and iodomethane (95.5 μΐ,, 1.530 mmol). The reaction mixture was stirred at ambient temperature for 30 minutes. The reaction mixture was then diluted with water and extracted with EtOAc (2 X). The organics were washed with water (3 X) and brine (1 X), dried over Na2S04, filtered and concentrated. Biotage chromatography (hexanes/EtOAc) provided 7- bromo-l-methylquinoxalin-2(lH)-one (87.6 mg, 0.366 mmol, 26.3% yield). 1H NMR (400 MHz, (CD3)2SO) δ = 8.257 (s, 1H), 7.824 (s, 1H), 7.768-7.746 (d, 1H), 7.569-7.543 (dd, 1H), 3.591 (s, 3H).
[00352] Step B: N-(2-Chloro-4-fluoro-3-(4-methyl-3-oxo-3,4-dihydroquinoxalin-6- ylamino)phenyl)propane-l -sulfonamide (1.8 mg, 0.004 mmol, 1.2% yield) was prepared according to the general procedure in Example 1, substituting 7-bromo-l-methylquinoxalin- 2(lH)-one for 6-bromo-3-methylquinazolin-4(3H)-one. 1H NMR (400 MHz, (CD3)2SO) δ = 9.560 (s, 1H), 8.734 (s, 1H), 7.909 (s, 1H), 7.612-7.591 (d, 1H), 7.430-7.361 (m, 2H), 6.656- 6.635 (d, 1H), 6.578 (s, 1H), 3.457 (s, 3H), 3.147-3.109 (t, 2H), 1.817-1.724 (m, 2H), 0.999- 0.962 (t, 3H). LC/MS: m/z 425.2, 427.1 [M+l].
Example 41
Figure imgf000099_0001
N-(2,4-Difluoro-3 -(3 -methyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)f\iran-2- sulfonamide
[00353] Step A: Methyl 3-amino-2,6-difluorobenzoate (652.8 mg, 3.488 mmol) was dissolved in 0.2M DCM (17.4 mL), chilled to 0°C, and then treated sequentially with triethylamine (1.42 mL, 10.46 mmol) and furan-2-sulfonyl chloride (1.16 mg, 6.98 mmol). The reaction mixture was warmed to ambient temperature and stirred for 16 hours. The reaction mixture was diluted with EtOAc, washed with water (2 X) and brine (1 X), dried over Na2S04, filtered and concentrated. The crude reaction mixture was then dissolved in 4:1 THF:MeOH (17.5 mL) and treated with 2.0M KOH (8.70 mL, 17.45 mmol) at ambient temperature for 2 hours. The organic solvent was then concentrated off in vacuo, and the reaction mixture was diluted with water and extracted with EtOAc (2 X). The aqueous layer was then acidified to pH 3 using 6.0M HC1 and extracted with EtOAc (2 X). The organics were washed with water (2 X) and brine (1 X), dried over Na2S04, filtered and concentrated to afford 2,6-difluoro-3-(furan-2-sulfonamido)benzoic acid (475.0 mg, 1.57 mmol, 44.9% yield). LC MS: m/z 302.0 [M-l].
[00354] Step B: 2,6-Difluoro-3-(furan-2-sulfonamido)benzoic acid (475.0 mg, 1.57 mmol) was dissolved in 0.1M DMF (15.7 mL) and treated with triethylamine (637.4 μΕ, 4.70 mmol) and then diphenylphosphoryl azide (523.7 μί, 2.350 mmol) at ambient temperature for 1 hour. The reaction mixture was then treated with deionized water (5.0 mL) and heated to 80°C for 16 hours. The reaction mixture was cooled to ambient temperature, diluted with EtOAc and washed with water (3 X) and brine (1 X), dried over Na2S04, filtered and concentrated. Biotage chromatography (hexanes/EtOAc) provided N-(3-amino-2,4- difiuorophenyl)furan-2-sulfonamide (152.6 mg, 0.56 mmol, 35.5% yield). LC/MS: m/z 273.1 [M-l].
[00355] Step C: N-(2,4-Difluoro-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)furan-2-sulfonamide (4.5 mg, 0.010 mmol, 9.6% yield) was prepared according to the general procedure in Example 1, substituting N-(3-amino-2,4- difluorophenyl)furan-2-sulfonamide for N-(3-amino-4-chloro-2-fluorophenyl)propane-l - sulfonamide. 1H NM (400 MHz, (CD3)2SO) δ =10.534 (s, 1H), 8.394 (s, 1H), 8.167 (s, 1H), 7.997 (s, 1H), 7.546-7.524 (d, 1H), 7.221-7.134 (m, 2H), 7.089-7.031 (m, 2H), 6.652 (s, 1H), 3.459 (s, 3H). LC/MS: m/z 431.1 [M-l].
Figure imgf000100_0001
N-(2-Chloro-3 -(4-ethyl-3 -oxo-3 -άίΙινάΓο ηίηοχ-ΐΙίη-ό-νΐ ηιΐηο^^-ίΙ^ΓορΙιεηνΠρΓο εηε- 1 - sulfonamide
[00356] Step A: 7-Bromo-l-ethylquinoxalin-2(lH)-one: 7-bromoquinoxalin-2(lH)- one (313.6 mg, 1.39 mmol) was dissolved in 0.2M DMF (7.0 mL) and treated with potassium carbonate (288.9 mg, 2.09 mmol) and iodoethane (123.7 \L, 1.53 mmol) at ambient temperature for 1 hour. The reaction mixture was then diluted with water and extracted with EtOAc (2 X). The organics were washed with water (3 X) and brine (1 X), dried over Na2S04, filtered and concentrated. Biotage chromatography (hexanes/EtOAc) provided 7- bromo-l-ethylquinoxalin-2(lH)-one (169.2 mg, 0.67 mmol, 48.0% yield). 1H NMR (400 MHz, (CD3)2SO) δ = 8.611 (s, 1H), 8.038 (s, 1H), 7.954-7.937 (d, 1H), 7.787-7.764 (d, 1H), 4.517-4.474 (q, 2H), 1.431-1.402 (t, 3H).
[00357] Step B: N-(2-Chloro-3-(4-ethyl-3-oxo-3,4-dihydroquinoxalin-6-ylamino)-4- fluorophenyl)propane-l -sulfonamide (5.4 mg, 0.012 mmol, 1.9% yield) was prepared according to the general procedure in Example 1, substituting 7-bromo-l-ethylquinoxalin- 2(lH)-one for 6-bromo-3-methylquinazolin-4(3H)-one. 1H NMR (400 MHz, (CD3)2SO) δ = 9.568 (s, 1H), 8.700 (s, 1H), 7.901 (s, 1H), 7.624-7.602 (d, 1H), 7.406-7.386 (d, 2H), 6.677- 6.627 (t, 2H), 4.103-4.049 (q, 2H), 3.150-3.1 12 (t, 2H), 1.821-1.727 (m, 2H), 1.207-1.171 (t, 3H), 1.001-0.964 (t, 3H). LC/MS: m/z 437.1, 439.1 [M-l].
Example 43
Figure imgf000100_0002
N-(2-Chloro-4-fluoro-3 -(4-isopropyl-3 -oxo-3 ,4-dihydroquinoxalin-6- ylamino)phenvDpropane- 1 -sulfonamide
[00358] Step A: 7-Bromoquinoxalin-2(lH)-one (322.6 mg, 1.43 mmol) was dissolved in 0.1 M DMF (14.4 mL) and treated with potassium carbonate (297.2 mg, 2.15 mmol) and 2- iodopropane (157.7 ih, 1.58 mmol) at 50°C for 2 hours. The reaction mixture was cooled to ambient temperature, diluted with water and extracted with EtOAc (2 X). The organics were washed with water (3 X) and brine (1 X), dried over Na2S04, filtered and concentrated. Biotage chromatography (hexanes/EtOAc) provided 7-bromo-l-isopropylquinoxalin-2(lH)- one (1 1.5 mg, 0.043 mmol, 3.0% yield). 1H NMR (400 MHz, (CD3)2SO) 5 = 8.144 (s, 1H), 8.015 (s, 1H), 7.757-7.736 (d, 1H), 7.552-7.526 (dd, 2H), 5.133 (m, 1H), 1.531-1.515 (d, 6H).
[00359] Step B: N-(2-Chloro-4-fluoro-3-(4-isopropyl-3-oxo-3,4-dihydroquinoxalin-6- ylamino)phenyl)propane-l -sulfonamide (5.4 mg, 0.012 mmol, 27.7% yield) was prepared according to the general procedure in Example 1, substituting 7-bromo-l- isopropylquinoxalin-2(lH)-one for 6-bromo-3-methylquinazolin-4(3H)-one. 1H NMR (400 MHz, (CD3)2SO) δ = 9.576 (s, 1H), 8.648 (s, 1H), 7.831 (s, 1H), 7.601-7.578 (d, 1H), 7.412- 7.394 (d, 2H), 6.849 (s, 1H), 6.638-6.615 (d, 1H), 5.190 (m, 1H), 3.145-3.107 (t, 2H), 1.815- 1.722 (m, 2H), 1.472-1.454 (d, 6H), 0.997-0.959 (t, 3H). LC/MS: m/z 453.2, 455.1 [M+l].
Exam le 44
Figure imgf000101_0001
N-(4-Mefhyl-3 -(3 -mefhyl-4-oxo-3 ,4-dihydroquinazolin-6-ylamino)phenyl)propane- 1 - sulfonamide
[00360] Step A: DMAP (0.096 g, 0.79 mmol), pyridine (1.67 mL, 20.7 mmol) and propane- 1-sulfonyl chloride (2.32 mL, 20.7 mmol) were added to a solution of 4-mefhyl-3- nitroaniline (3 g, 19.7 mmol) in DCM (25 mL). The reaction was stirred at 45°C for 18 hours. The reaction mixture was then diluted with ethyl acetate and washed with water. The organics were dried with sodium sulfate, filtered and concentrated in vacuo to obtain a crude mixture of N-(4-methyl-3-nitrophenyl)propane-l -sulfonamide (5 g). Half of this crude mixture (2.5 g) was dissolved in ethanol (35 mL) and water (15 mL). Iron (5.4 g, 96.8 mmol) and ammonium chloride (2.1 g, 38.7 mmol) were subsequently added. The reaction was heated at reflux for 2 hours, after which salts were filtered off. The filtrate was diluted with ethyl acetate and water, and the layers were partitioned and washed with a saturated solution of NaCl. The aqueous layer was extracted twice with ethyl acetate. The organics were dried with sodium sulfate, filtered and concentrated in vacuo. The crude mixture was purified by flash chromatography to obtain N-(3-amino-4-methylphenyl)propane-l -sulfonamide (2.46 g, quantitative yield). 1H NMR (400 MHz, CDC13) δ 6.98 (d, J = 8.0 Hz, 1H), 6.63 (d, J = 2.2 Hz, 1H), 6.45 (dd, J = 8.0, 2.2 Hz, 1H), 6.29 (s, 1H), 3.08 - 3.01 (m, 2H), 2.13 (br s, 2H) 1.90 - 1.78 (m, 2H), 1.01 (t, J = 7.5 Hz, 3H); m/z (ES-MS) 229.3 (100%) [M+l].
[00361] Step B: A sealed microwave vial was charged with 6-bromo-3- methylquinazolin-4(3H)-one (0.075 g, 0.31 mmol), N-(3-amino-4-methylphenyl)propane-l- sulfonamide (0.079 g, 0.35 mmol), Pd2(dba)3 (0.014 g, 0.016 mmol), (R)-BINAP (0.020 g, 0.031 mmol), sodium tert-butoxide (0.106 g, 1.10 mmol) and toluene (2.3 mL). The mixture was heated in a microwave reactor at 120°C for 15 minutes. The reaction mixture was filtered, and the filtrate was directly concentrated in vacuo. The crude product was purified by reverse phase HPLC to give N-(4-methyl-3-(3-methyl-4-oxo-3,4-dihydroquinazolin-6- ylamino)phenyl)propane-l -sulfonamide (23 mg, 19%). 1H NMR (500 MHz, (CD3)2SO) δ 9.58 (s, 1H), 8.14 (s, 1H), 7.94 (s, 1H), 7.53 (d, J = 8.8 Hz, 1H), 7.45 (d, J = 2.6 Hz, 1H), 7.39 (dd, J = 8.8, 2.7 Hz, 1H), 7.18 (d, J = 8.2 Hz, 1H), 7.14 (d, J = 2.1 Hz, 1H), 6.85 (dd, J = 8.1, 2.2 Hz, 1H), 3.46 (s, 3H), 3.02 (dd, J = 8.6, 6.7 Hz, 2H), 2.14 (s, 3H), 1.74 - 1.60 (m, 2H), 0.94 (t, J = 7.5 Hz, 3H); m/z (ES-MS) 387.2 (96.1%) [M+1], LCMS retention time: 3.78 minutes.
[00362] The following compounds in Table 1 were prepared according to the general procedure of the Example Number given in the Method column using appropriate intermediates.
Table 1
Ex. Structure Name MS / NMR Method
#
45
Figure imgf000102_0001
Figure imgf000102_0002
[M+l]
46 N-(3-(ethyl(3-methyl- ¾ NMR (400 6
4-oxo-3,4- MHz, CDC13) δ
Figure imgf000103_0001
dihydroquinazolin-6- 7.89 (s, 1H),
yl)amino)-2,4- 7.56-7.51 (m, difluorophenyl)propan 3H), 7.09-6.97
e-1 -sulfonamide (m, 2H), 6.43 (s,
1H), 3.80 (m, 2H), 3.57 (s,
3H), 3.09 (m, 2H), 1.88 (m, 2H), 1.27 (m, 3H), 1.05 (t,
J=7.4 Hz, 3H);
LC/MS: m/z
437.2, 439.2
[M+l]
47 N-(3-(ethyl(3-methyl- Ή NMR (400 1
4-oxo-3,4- MHz, (CD3)2SO)
0 CI dihydroquinazolin-6- δ 9.40 (br s, 1H),
yl)amino)-2,4- 8.26 (br s, 1H), difluorophenyl)propan 8.21 (s, 1H),
e-1 -sulfonamide 7.62 (s, 1H),
7.58 (d, J=8.4
Hz, 1H), 7.51
(m, 1H), 7.29- 7.20 (m, 2H), 7.14 (m, 1H), 3.47 (s, 3H),
3.13 (m, 2H), 1.79 (m, 2H), 0.99 (t, J=7.4
Hz, 3H); LC/MS: m/z
425.1, 427.1
[M+l]
1 -cyclopropyl-N-(2,4- 'H NMR (400 1 difluoro-3 -(3 -methyl- MHz, (CD3)2SO)
H F H
4-oxo-3,4- δ 9.69 (s, 1H), dihydroquinazolin-6- 8.45 (s, 1H), ylamino)phenyl)metha 8.16 (s, 1H),
nesulfonamide 7.53-7.55 (d,
1H), 7.17-7.29 (m, 4H), 3.45 (s, 3H), 3.09-3.11 (d, 2H), 1.02- 1.13 (m, 1H), 0.58-0.53 (m, 2H), 0.33-0.36 (m, 2H); m/z
(APCI-pos) M+l
- 421.1
N-(2,4-difluoro-3-(3- 'H NMR (400 1 methyl-4-oxo-3,4- MHz, (CD3)2SO)
H F H
dihydroquinazolin-6- 5 9.68 (s, 1H), ylamino)phenyl)-2- 8.46 (s, 1H), methylpropane- 1 - 8.16 (s, 1H),
sulfonamide 7.53-7.55 (d,
1H), 7.27-7.29
(m, 1H), 7.21- 7.23 (m, 3H), 3.45 (s, 3H),
3.00-3.02 (m,
2H), 2.15-2.21 (m, 1H), 1.01- 1.03 (d, 6H); m/z
Figure imgf000105_0001
[00363] It will be understood that the enumerated embodiments are not intended to limit the invention to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications and equivalents, which may be included within the scope of the present invention as defined by the claims. Thus, the foregoing description is considered as illustrative only of the principles of the invention.
[00364] The words "comprise," "comprising," "include," "including," and "includes" when used in this specification and in the following claims are intended to specify the presence of stated features, integers, components, or steps, but they do not preclude the presence or addition of one or more other features, integers, components, steps, or groups thereof.

Claims

What is claimed is:
1. A compound selected from Formula I:
Figure imgf000106_0001
and stereoisomers, tautomers, and pharmaceutically acceptable salts thereof, wherein:
the dashed lines represent optional double bonds;
W is selected from N, NR6, C(=0), CR7 and CHR7;
X is selected from N, NR8, C(=0), CR9 and CHR9, provided when:
(i) W is N, then X is CR9,
(ii) W is NR6, then X is C(=0) or CHR9,
(iii) W is C(=0), then X is NR8 or CHR9,
(iv) W is CR7, then X is N or CR9, and
(v) W is CHR7, then X is NR8, C(=0) or CHR9;
Y is selected from N, NR10, CR11 and CHR11;
Z is selected from N, NR12, CR13 and CHR13;
L is selected from O, NR14, S, C(=0) and CR15R16;
1 2
R' and R' are independently selected from hydrogen, halogen, CN, Q-C3 alkyl, Ci-Ca alkoxy and C2-C3 alkynyl;
R is selected from hydrogen, halogen and C C3 alkyl;
R4 is selected from C3-C5 cycloalkyl, Ci-C^ alkyl, C2-C6 alkenyl, C2-C alkynyl, phenyl, a 5-6 membered heteroaryl, or NRaRb, wherein the cycloalkyl, alkyl, alkenyl, alkynyl, phenyl and heteroaryl are optionally substituted with ORc, halogen, phenyl, C3-C4 cycloalkyl, or C C4 alkyl optionally substituted with halogen;
R5 is selected from hydrogen, Cj-C3 alkyl and C!-C3 alkoxy, wherein the alkyl and alkoxy are optionally substituted with Rd;
R6 is selected from hydrogen, Q-C6 alkyl, Ci-C6 alkenyl, Ci-C^ alkynyl, C3-C cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORe, SRe, NRfRg, phenyl, Q-C4 alkyl, C1-C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp;
R is selected from hydrogen, Ci-Ce alkyl, C]-C6 alkenyl, C C6 alkynyl, C!-C6 alkoxy, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, a 5 to 6 membered heteroaryl, and NRhR', wherein the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORe, SRe, NRfR8, C!-C4 alkyl, C C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp;
R is selected from hydrogen, Q-C6 alkyl, Ci-C alkenyl, C\-Ce alkynyl, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, and a 5 to 6 membered heteroaryl, wherein the alkyl, alkenyl, alkynyl, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), OR", SR*, NRkR!, phenyl, Q-Gj alkyl, Q-Q alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp;
R9 is selected from hydrogen, Q-Q alkyl, Ci-Ce alkenyl, C!-C6 alkynyl, C^Ce alkoxy, C3-C6 cycloalkyl, phenyl, a 3 to 6 membered heterocyclyl, a 5 to 6 membered heteroaryl, and NRmRn, wherein the alkyl, alkenyl, alkynyl, alkoxy, cycloalkyl, phenyl, heterocyclyl and heteroaryl are optionally substituted with halogen, oxo (not on phenyl or heteroaryl), ORJ, SRj, NR^1, CrC4 alkyl, C C4 alkoxy, and cyclopropyl, wherein the alkyl, alkoxy and cyclopropyl are optionally substituted with Rp;
R10 is selected from hydrogen and halogen;
R11 is selected from hydrogen and halogen, or
R11 and R14 together with the atoms to which they are attached form a 5-6 membered heterocyclyl or heteroaryl optionally substituted with oxo, or
R11 and R15 together with the atoms to which they are attached form a 5-6 membered carbocyclyl;
R is hydrogen;
R is selected from hydrogen and halogen;
R14 is selected from hydrogen and C C4 alkyl;
R15 and R16 are independently selected from hydrogen and Q-C3 alkyl;
Ra and Rb are independently selected from hydrogen or Q-C5 alkyl optionally substituted by halogen, or
Ra and Rb are taken together with the nitrogen atom to which they are attached to form a 3-6 membered heterocyclyl optionally substituted by halogen, oxo or Ci-C3 alkyl; Rc is selected from hydrogen and C C6 alkyl optionally substituted by halogen;
each Rd is selected from halogen and OH;
Re is selected from hydrogen and C!-C4 alkyl;
Rf and Rg are independently selected from hydrogen and C C4 alkyl optionally substituted with halogen;
Rh and R1 are independently selected from hydrogen and Q-C4 alkyl optionally substituted with halogen;
RJ is selected from hydrogen and Q-C4 alkyl;
Rk and R1 are independently selected from hydrogen and C1-C4 alkyl optionally substituted with halogen;
Rm and Rn are independently selected from hydrogen and C1-C4 alkyl optionally substituted with halogen; and
each Rp is independently selected from halogen, oxo, C]-C4 alkyl and C!-C4 alkoxy, wherein the alkyl and alkoxy are optionally substituted with halogen.
2. The compound of Claim 1 wherein the residue:
Figure imgf000108_0001
wherein the wav line represents the point of attachment of the residue to L, is selected from:
Figure imgf000108_0002
Figure imgf000109_0001
Figure imgf000109_0002
Figure imgf000110_0001
4. The compound of any one of Claims 1 to 3, having the Formula II:
Figure imgf000110_0002
II
The compound of any one of Claims 1 to 4, having the Formula III:
Figure imgf000110_0003
III
The compound of any one of Claims 1 to 5, having the Formula IV:
Figure imgf000110_0004
IV
7. The compound of Claim 6, wherein R6 is methyl, benzyl, phenyl or 1 -methyl lH-pyrazol-4-yl.
8. The compound of any one of Claims 1 to 5, having the Formula V:
Figure imgf000110_0005
9. The compound of Claim 8, wherein R7 is hydrogen, OCH3 or NHCH3. The compound of any one of Claims 1 to 5, havin the Formula VI:
Figure imgf000111_0001
VI
11. The compound of Claim 10, wherein R9 is hydrogen, OCH3, OCH2CH3, NHCH3, N(CH3)2, piperazin-l-yl, 4-methylpirperazin-l-yl or tert-butyl 4-piperazine-l carboxylate.
12. The compound of any one of Claims 1 to 5, having the Formula VII:
Figure imgf000111_0002
VII
The compound of Claim 12, wherein R is methyl, ethyl or isopropyl.
The compound of any one of Claims 1 to 13, wherein L is selected from O and
NR 14
15. The compound of any one of Claims 1 to 14, wherein L is O, NH, N(CH3) or N(CH2CH3).
16. The compound of any one of Claims 1 to 15, wherein the residue:
Figure imgf000111_0003
wherein the wavy line represents the point of attachment of the residue to L, is selected from:
Figure imgf000111_0004
Figure imgf000112_0001
17. The compound of any one of Claims 1 to 15, wherein R1 is hydrogen, CI or F; R2 is CI, F or CN; and R3 is hydrogen.
18. The compound of any one of Claims 1 to 17, wherein R4 is propyl, isobutyl, -CH2CH2CH2F, cyclopropylmethyl, -N(CH3)2, pyrrolidine, or furan-2-yl.
19. The compound of any one of Claims 1 to 18, wherein R5 is hydrogen.
20. A compound of Claim 1 and named in any one of Examples 1-49.
21. A pharmaceutical composition, comprising a compound of any one of Claims 1-20 and a pharmaceutically acceptable carrier or excipient.
22. A method of preventing or treating a disease or disorder modulated by b-Raf, comprising administering to a mammal in need of such treatment an effective amount of a compound of any one of Claims 1-20.
23. A compound of any one of Claims 1-20 for use in therapy.
24. Use of a compound of any one of Claims 1-20 in the manufacture of a medicament for the treatment of cancer.
25. A pharmaceutical composition comprising a compound of any one of Claims 1-20 for use in the treatment of cancer.
PCT/US2011/026700 2011-03-01 2011-03-01 Heterocyclic sulfonamides as raf inhibitors WO2012118492A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/US2011/026700 WO2012118492A1 (en) 2011-03-01 2011-03-01 Heterocyclic sulfonamides as raf inhibitors

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US2011/026700 WO2012118492A1 (en) 2011-03-01 2011-03-01 Heterocyclic sulfonamides as raf inhibitors

Publications (1)

Publication Number Publication Date
WO2012118492A1 true WO2012118492A1 (en) 2012-09-07

Family

ID=44259778

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/026700 WO2012118492A1 (en) 2011-03-01 2011-03-01 Heterocyclic sulfonamides as raf inhibitors

Country Status (1)

Country Link
WO (1) WO2012118492A1 (en)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130053384A1 (en) * 2011-08-26 2013-02-28 Neupharma, Inc. Certain Chemical Entities, Compositions, and Methods
CN103242245A (en) * 2013-05-22 2013-08-14 苏州明锐医药科技有限公司 Preparation method of tandutinib
WO2014047648A1 (en) 2012-09-24 2014-03-27 Neupharma, Inc. Certain chemical entities, compositions, and methods
WO2014169167A1 (en) * 2013-04-12 2014-10-16 Asana Biosciences, Llc Quinazolines and azaquinazolines as dual inhibitors of ras/raf/mek/erk and pi3k/akt/pten/mtor pathways
US8895601B2 (en) 2010-04-30 2014-11-25 Astex Therapeutics Ltd Pyrazolyl quinoxaline kinase inhibitors
WO2014206344A1 (en) 2013-06-28 2014-12-31 Beigene, Ltd. Fused tricyclic amide compounds as multiple kinase inhibitors
EP2757885A4 (en) * 2011-09-21 2015-03-04 Neupharma Inc Certain chemical entites, compositions, and methods
WO2016010662A1 (en) * 2014-07-16 2016-01-21 Development Center For Biotechnology Quinoxaline compounds, method for preparing the same and use thereof
US9249111B2 (en) 2011-09-30 2016-02-02 Neupharma, Inc. Substituted quinoxalines as B-RAF kinase inhibitors
US9290478B2 (en) 2010-11-29 2016-03-22 Astex Therapeutics Ltd Substituted quinoxalines as FGFR kinase inhibitors
US9303030B2 (en) 2012-05-30 2016-04-05 Astex Therapeutics Limited Compounds
US9303029B2 (en) 2011-10-28 2016-04-05 Astex Therapeutics Ltd Substituted quinoxalines as FGFR kinase inhibitors
US9309242B2 (en) 2011-10-28 2016-04-12 Astex Therapeutics Ltd Substituted pyrido[2,3-b]pyrazines as FGFR kinase inhibitors
US9309241B2 (en) 2011-10-28 2016-04-12 Astex Therapeutics Ltd Naphthyridine derivative compounds
JP2016521276A (en) * 2013-04-26 2016-07-21 アステックス、セラピューティックス、リミテッドAstex Therapeutics Limited Quinazolinone derivatives useful as FGFR kinase modulators
US9439896B2 (en) 2011-10-28 2016-09-13 Astex Therapeutics Ltd Quinolines as FGFR kinase modulators
US9447098B2 (en) 2012-05-30 2016-09-20 Astex Therapeutics Ltd Pteridines as FGFR inhibitors
US9518029B2 (en) 2011-09-14 2016-12-13 Neupharma, Inc. Certain chemical entities, compositions, and methods
US9670180B2 (en) 2012-01-25 2017-06-06 Neupharma, Inc. Certain chemical entities, compositions, and methods
US9725421B2 (en) 2012-11-12 2017-08-08 Neupharma, Inc. Substituted quinoxalines as B-raf kinase inhibitors
US9902714B2 (en) 2014-03-26 2018-02-27 Astex Therapeutics Ltd Quinoxaline derivatives useful as FGFR kinase modulators
US10085982B2 (en) 2014-03-26 2018-10-02 Astex Therapeutics Ltd Combinations
WO2019014668A1 (en) * 2017-07-14 2019-01-17 Asana Biosciences, Llc Formulations, methods, kit, and dosage forms for improved stability of an active pharmaceutical ingredient
WO2019060611A1 (en) * 2017-09-20 2019-03-28 Abm Therapeutics, Inc. Cyclic iminopyrimidine derivatives as kinase inhibitors
US10478494B2 (en) 2015-04-03 2019-11-19 Astex Therapeutics Ltd FGFR/PD-1 combination therapy for the treatment of cancer
US10736900B2 (en) 2014-03-26 2020-08-11 Astex Therapeutics Ltd Combinations of an FGFR inhibitor and an IGF1R inhibitor
WO2020261156A1 (en) 2019-06-28 2020-12-30 Array Biopharma Inc. Quinazolin-4-one derivatives useful for the treatment of braf-associated diseases and disorders
US10898482B2 (en) 2015-02-10 2021-01-26 Astex Therapeutics Ltd Pharmaceutical compositions comprising N-(3,5-dimethoxyphenyl)-N'-1 methylethyl)-N-[3-(1-methyl-1H-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine
WO2021116050A1 (en) 2019-12-10 2021-06-17 F. Hoffmann-La Roche Ag New braf inhibitors as paradox breakers
WO2021116055A1 (en) 2019-12-10 2021-06-17 F. Hoffmann-La Roche Ag New methylquinazolinone derivatives
US11155555B2 (en) 2015-09-23 2021-10-26 Janssen Pharmaceutica Nv Compounds
WO2021250521A1 (en) * 2020-06-09 2021-12-16 Array Biopharma Inc. 4-oxo-3,4-dihydroquinazolinon compounds for the treatment of braf-associated diseases and disorders
CN113968803A (en) * 2020-07-24 2022-01-25 苏州匠化生物科技有限公司 Novel method for synthesizing 2, 6-difluoro-3-propyl sulfonamide benzoic acid
WO2022129260A1 (en) * 2020-12-18 2022-06-23 F. Hoffmann-La Roche Ag New aryl-pyrido-pyrimidin-one derivatives
WO2022129259A1 (en) * 2020-12-18 2022-06-23 F. Hoffmann-La Roche Ag New quinazolinone derivatives
WO2022261250A1 (en) * 2021-06-08 2022-12-15 C4 Therapeutics, Inc. Therapeutics for the degradation of mutant braf
WO2022258600A1 (en) 2021-06-09 2022-12-15 F. Hoffmann-La Roche Ag Combination of a particular braf inhibitor (paradox breaker) and a pd-1 axis binding antagonist for use in the treatment of cancer
WO2022258584A1 (en) 2021-06-09 2022-12-15 F. Hoffmann-La Roche Ag New solid forms of (3r)-n-[2-cyano-4-fluoro-3-(3-methyl-4-oxo-quinazolin-6-yl)oxy-phenyl]-3-fluoro-pyrrolidine-1-sulfonamide
US11542247B2 (en) 2015-09-23 2023-01-03 Janssen Pharmaceutica Nv Bi-heteroaryl substitute 1,4-benzodiazepines and uses thereof for the treatment of cancer
RU2797606C1 (en) * 2019-06-28 2023-06-07 Аррэй Байофарма Инк. Quinazolin-4-one derivatives useful in the treatment of braf-associated diseases and disorders
JP7289978B1 (en) 2021-06-09 2023-06-12 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Combination therapy for cancer treatment
WO2023105371A1 (en) * 2021-12-08 2023-06-15 Array Biopharma Inc. Crystalline form of n-(2-chloro-3-((5-chloro-3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4-fluorophenyl)-3-fluoroazetidine-1-sulfonamide
WO2024017294A1 (en) * 2022-07-19 2024-01-25 西藏海思科制药有限公司 Preparation and use of quinazolinone derivative as kinase inhibitor
WO2024054591A1 (en) * 2022-09-07 2024-03-14 Arvinas Operations, Inc. Rapidly accelerated fibrosarcoma (raf) degrading compounds and associated methods of use

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0415595A1 (en) 1989-08-31 1991-03-06 Imperial Chemical Industries Plc Fluorobenzene derivatives
WO1996015111A1 (en) 1994-11-15 1996-05-23 Regents Of The University Of Minnesota Method and intermediates for the synthesis of korupensamines
WO2003022840A1 (en) 2001-09-05 2003-03-20 Smithkline Beecham Plc Pyridine derivatives as raf kinase inhibitors
US20040082014A1 (en) 2001-01-22 2004-04-29 Michael Sendtner Method and test system for identifying substances which protect nerve cells
US20040127496A1 (en) 2001-05-19 2004-07-01 Dean David Kenneth Imidazole-2-carboxamide derivatives as raf kinase inhibitors
US20060258888A1 (en) 2005-05-12 2006-11-16 Boehringer Ingelheim International, Gmbh Bis-Amination of Aryl Halides
WO2008079903A1 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2, 3-b] pyridine derivatives as kinase modulators
WO2009012283A1 (en) * 2007-07-17 2009-01-22 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
WO2009111280A1 (en) * 2008-02-29 2009-09-11 Array Biopharma Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
WO2009111277A1 (en) * 2008-02-29 2009-09-11 Array Biopharma Inc. Imdizo [4. 5-b] pyridine derivatives used as raf inhibitors

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0415595A1 (en) 1989-08-31 1991-03-06 Imperial Chemical Industries Plc Fluorobenzene derivatives
WO1996015111A1 (en) 1994-11-15 1996-05-23 Regents Of The University Of Minnesota Method and intermediates for the synthesis of korupensamines
US20040082014A1 (en) 2001-01-22 2004-04-29 Michael Sendtner Method and test system for identifying substances which protect nerve cells
US20040127496A1 (en) 2001-05-19 2004-07-01 Dean David Kenneth Imidazole-2-carboxamide derivatives as raf kinase inhibitors
WO2003022840A1 (en) 2001-09-05 2003-03-20 Smithkline Beecham Plc Pyridine derivatives as raf kinase inhibitors
US20060258888A1 (en) 2005-05-12 2006-11-16 Boehringer Ingelheim International, Gmbh Bis-Amination of Aryl Halides
WO2008079903A1 (en) * 2006-12-21 2008-07-03 Plexxikon, Inc. Pyrrolo [2, 3-b] pyridine derivatives as kinase modulators
WO2009012283A1 (en) * 2007-07-17 2009-01-22 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor
WO2009111280A1 (en) * 2008-02-29 2009-09-11 Array Biopharma Inc. N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
WO2009111277A1 (en) * 2008-02-29 2009-09-11 Array Biopharma Inc. Imdizo [4. 5-b] pyridine derivatives used as raf inhibitors

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
"Beilsteins Handbuch der organischen Chemie", SPRINGER-VERLAG
"Chiral Liquid Chromatography", 1989, CHAPMAN AND HALL
"Drug Stereochemistry: Analytical Methods and Pharmacology", 1993, MARCEL DEKKER, INC.
ANGEW CHEM. INTL. ED. ENGL., vol. 33, 1994, pages 183 - 186
ANSEL, HOWARD C. ET AL.: "Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems", 2004, LIPPINCOTT, WILLIAMS & WILKINS
ELIEL, E.; WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC.
ELIEL, E.; WILEN, S.: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC., pages: 322
GENNARO, ALFONSO R. ET AL.: "Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT, WILLIAMS & WILKINS
JACOB III, PEYTON: "Resolution of (+)-5-Bromonornicotine. Synthesis of (R)- and (S)-Nornicotine of High Enantiomeric Purity", J. ORG. CHEM., vol. 47, no. 21, 1982, pages 4165 - 4167
LI, NANXIN ET AL.: "B-Raf kinase inhibitors for cancer treatment", CURRENT OPINION IN INVESTIGATIONAL DRUGS, vol. 8, no. 6, 2007, pages 452 - 456
LOCHMULLER, C. H. ET AL.: "Chromatographic resolution of enantiomers: Selective review", J. CHROMATOER., vol. 113, no. 3, 1975, pages 283 - 302
LOUIS F. FIESER; MARY FIESER: "Reagents for Organic Synthesis", vol. 1-23, 1967, WILEY
MAILLARD, JACQUES ET AL.: "(3H)-Quinazolin-4-one derivatives with antiinflammatory activity. II. Derivatives substituted in the aromatic nucleus, and related compounds", CHIM. THER., vol. 2, no. 4, 1967, pages 231 - 9
OKAMOTO, YOSHIO ET AL.: "Optical resolution of dihydropyridine enantiomers by high-performance liquid chromatography using phenylcarbamates of polysaccharides as a chiral stationary phase", J. OF CHROMATOGR., vol. 513, 1990, pages 375 - 378
ROE, S. MARK ET AL.: "The Mechanism of Hsp90 Regulation by the Protein Kinase-Specific Cochaperone p50cdc37", CELL, vol. 116, 2004, pages 87 - 98
ROWE, RAYMOND C: "Handbook of Pharmaceutical Excipients", 2005, PHARMACEUTICAL PRESS
SIENKOWSKA, TETRAHEDRON, vol. 56, 2000, pages 165
STANCATO, LF ET AL.: "Raf exists in a native heterocomplex with Hsp90 and p50 that can be reconstituted in a cell free system", J. BIOL. CHEM., vol. 268, no. 29, 1993, pages 21711 - 21716
T. W. GREENE ET AL.: "Greene's Protective Groups in Organic Synthesis", 2006, WILEY INTERSCIENCE

Cited By (123)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10519137B2 (en) 2010-04-30 2019-12-31 Astex Therapeutics Ltd Pyrazolyl quinoxaline kinase inhibitors
US9464071B2 (en) 2010-04-30 2016-10-11 Astex Therapeutics Ltd Pyrazolyl quinoxaline kinase inhibitors
US9850228B2 (en) 2010-04-30 2017-12-26 Astex Therapeutics Ltd Pyrazolyl quinoxaline kinase inhibitors
US8895601B2 (en) 2010-04-30 2014-11-25 Astex Therapeutics Ltd Pyrazolyl quinoxaline kinase inhibitors
US9856236B2 (en) 2010-11-29 2018-01-02 Astex Therapeutics Ltd Substituted quinoxalines as FGFR kinase inhibitors
US9290478B2 (en) 2010-11-29 2016-03-22 Astex Therapeutics Ltd Substituted quinoxalines as FGFR kinase inhibitors
US10137125B2 (en) * 2011-08-26 2018-11-27 Neupharma, Inc. Benzenesulfonamide derivatives of quinoxaline, pharmaceutical compositions thereof, and their use in methods for treating cancer
US10561652B2 (en) 2011-08-26 2020-02-18 Neupharma, Inc. Benzenesulfonamide derivatives of quinoxaline, pharmaceutical compositions thereof, and their use in methods for treating cancer
US20130053384A1 (en) * 2011-08-26 2013-02-28 Neupharma, Inc. Certain Chemical Entities, Compositions, and Methods
US9572808B2 (en) 2011-08-26 2017-02-21 Neupharma, Inc. Benzenesulfonamide derivatives of quinoxaline, pharmaceutical compositions thereof, and their use in methods for treating cancer
US9295671B2 (en) * 2011-08-26 2016-03-29 Neupharma, Inc. Benzenesulfonamide derivatives of quinoxaline, pharmaceutical compositions thereof, and their use in methods for treating cancer
US20160303118A1 (en) * 2011-08-26 2016-10-20 Neupharma, Inc. Benzenesulfonamide derivatives of quinoxaline, pharmaceutical compositions thereof, and their use in methods for treating cancer
US9822081B2 (en) 2011-09-14 2017-11-21 Neupharma, Inc. Certain chemical entities, compositions, and methods
US9518029B2 (en) 2011-09-14 2016-12-13 Neupharma, Inc. Certain chemical entities, compositions, and methods
US10759766B2 (en) 2011-09-14 2020-09-01 Neupharma, Inc. Certain chemical entities, compositions, and methods
US10065932B2 (en) 2011-09-14 2018-09-04 Neupharma, Inc. Certain chemical entities, compositions, and methods
US9249110B2 (en) 2011-09-21 2016-02-02 Neupharma, Inc. Substituted quinoxalines as B-raf kinase inhibitors
EP2757885A4 (en) * 2011-09-21 2015-03-04 Neupharma Inc Certain chemical entites, compositions, and methods
US9249111B2 (en) 2011-09-30 2016-02-02 Neupharma, Inc. Substituted quinoxalines as B-RAF kinase inhibitors
US9757364B2 (en) 2011-10-28 2017-09-12 Astex Therapeutics Ltd Naphthyridine derivative compounds
US9527844B2 (en) 2011-10-28 2016-12-27 Astex Therapeutics Limited Naphthyridine derivative compounds
US9309242B2 (en) 2011-10-28 2016-04-12 Astex Therapeutics Ltd Substituted pyrido[2,3-b]pyrazines as FGFR kinase inhibitors
US9439896B2 (en) 2011-10-28 2016-09-13 Astex Therapeutics Ltd Quinolines as FGFR kinase modulators
US10052320B2 (en) 2011-10-28 2018-08-21 Astex Therapeutics Ltd Substituted quinoxalines as FGFR kinase inhibitors
US9303029B2 (en) 2011-10-28 2016-04-05 Astex Therapeutics Ltd Substituted quinoxalines as FGFR kinase inhibitors
US10039759B2 (en) 2011-10-28 2018-08-07 Astex Therapeutics Ltd Quinolines as FGFR kinase modulators
US9309241B2 (en) 2011-10-28 2016-04-12 Astex Therapeutics Ltd Naphthyridine derivative compounds
US10045982B2 (en) 2011-10-28 2018-08-14 Astex Therapeutics Ltd Substituted pyrido[2,3-b]pyrazines as FGFR kinase inhibitors
US9908866B2 (en) 2012-01-25 2018-03-06 Neupharma, Inc. Certain chemical entities, compositions, and methods
US9670180B2 (en) 2012-01-25 2017-06-06 Neupharma, Inc. Certain chemical entities, compositions, and methods
US10590106B2 (en) 2012-01-25 2020-03-17 Neupharma, Inc. Certain chemical entities, compositions, and methods
US9447098B2 (en) 2012-05-30 2016-09-20 Astex Therapeutics Ltd Pteridines as FGFR inhibitors
US9303030B2 (en) 2012-05-30 2016-04-05 Astex Therapeutics Limited Compounds
US10272087B2 (en) 2012-05-30 2019-04-30 Astex Therapeutics Ltd Pteridines as FGFR inhibitors
US9737544B2 (en) 2012-05-30 2017-08-22 Astex Therapeutics Limited Compounds
EP2897618A4 (en) * 2012-09-24 2016-03-02 Neupharma Inc Certain chemical entities, compositions, and methods
WO2014047648A1 (en) 2012-09-24 2014-03-27 Neupharma, Inc. Certain chemical entities, compositions, and methods
US20150246885A1 (en) * 2012-09-24 2015-09-03 Xiangping Qian Certain chemical entites, compositions, and methods
US9688635B2 (en) * 2012-09-24 2017-06-27 Neupharma, Inc. Certain chemical entities, compositions, and methods
US10457641B2 (en) 2012-09-24 2019-10-29 Neupharma, Inc. Certain chemical entities, compositions, and methods
CN104812389A (en) * 2012-09-24 2015-07-29 润新生物公司 Certain chemical entities, compositions, and methods
US9725421B2 (en) 2012-11-12 2017-08-08 Neupharma, Inc. Substituted quinoxalines as B-raf kinase inhibitors
US10047059B2 (en) 2012-11-12 2018-08-14 Neupharma, Inc. Substituted quinoxalines for inhibiting kinase activity
US10226468B2 (en) 2013-04-12 2019-03-12 Asana Biosciences, Llc Quinazolines and azaquinazolines as dual inhibitors of RAS/RAF/MEK/ERK and PI3K/AKT/PTEN/mTOR pathways
WO2014169167A1 (en) * 2013-04-12 2014-10-16 Asana Biosciences, Llc Quinazolines and azaquinazolines as dual inhibitors of ras/raf/mek/erk and pi3k/akt/pten/mtor pathways
US10912779B2 (en) 2013-04-12 2021-02-09 Asana Biosciences, Llc Quinazolines and azaquinazolines as dual inhibitors of RAS/RAF/MEK/ERK and PI3K/AKT/PTEN/mTOR pathways
CN105283454A (en) * 2013-04-12 2016-01-27 阿萨纳生物科技有限责任公司 Quinazolines and azaquinazolines as dual inhibitors of RAS/RAF/MEK/ERK and PI3K/AKT/PTEN/MTOR pathways
US9499495B2 (en) 2013-04-12 2016-11-22 Asana Biosciences, Llc Quinazolines and azaquinazolines as dual inhibitors of RAS/RAF/MEK/ERK and PI3K/AKT/PTEN/mTOR pathways
JP2016519685A (en) * 2013-04-12 2016-07-07 アサナ・バイオサイエンシズ,リミテッド・ライアビリティ・カンパニー Quinazolines and azaquinazolines as dual inhibitors of the RAS / RAF / MEK / ERK pathway and the PI3K / AKT / PTEN / MTOR pathway
US9757382B2 (en) 2013-04-12 2017-09-12 Asana Biosciences, Llc Quinazolines and azaquinazolines as dual inhibitors of RAS/RAF/MEK/ERK and PI3K/AKT/PTEN/mTOR pathways
RU2701517C2 (en) * 2013-04-26 2019-09-27 Астекс Терапьютикс Лимитед Quinazolinone derivatives applicable as modulators of fgfr kinase
US9493426B2 (en) 2013-04-26 2016-11-15 Astex Therapeutics Limited Quinazolinone derivatives useful as FGFR kinase modulators
JP2016521276A (en) * 2013-04-26 2016-07-21 アステックス、セラピューティックス、リミテッドAstex Therapeutics Limited Quinazolinone derivatives useful as FGFR kinase modulators
CN103242245A (en) * 2013-05-22 2013-08-14 苏州明锐医药科技有限公司 Preparation method of tandutinib
WO2014206344A1 (en) 2013-06-28 2014-12-31 Beigene, Ltd. Fused tricyclic amide compounds as multiple kinase inhibitors
US9902714B2 (en) 2014-03-26 2018-02-27 Astex Therapeutics Ltd Quinoxaline derivatives useful as FGFR kinase modulators
US10085982B2 (en) 2014-03-26 2018-10-02 Astex Therapeutics Ltd Combinations
US11918576B2 (en) 2014-03-26 2024-03-05 Astex Therapeutics Ltd Combination of an FGFR inhibitor and a CMET inhibitor
US10736900B2 (en) 2014-03-26 2020-08-11 Astex Therapeutics Ltd Combinations of an FGFR inhibitor and an IGF1R inhibitor
US10716787B2 (en) 2014-03-26 2020-07-21 Astex Therapeutics Ltd Combinations
US10421747B2 (en) 2014-03-26 2019-09-24 Astex Therapeutics Ltd Quinoxaline derivatives useful as FGFR kinase modulators
US10745360B2 (en) * 2014-07-16 2020-08-18 Development Center For Biotechnology Quinoxaline compounds, method for preparing the same and use thereof
AU2015290176B2 (en) * 2014-07-16 2019-10-31 Dcb-Usa Llc Quinoxaline compounds, method for preparing the same and use thereof
US20180208561A1 (en) * 2014-07-16 2018-07-26 Development Center For Biotechnology Quinoxaline compounds, method for preparing the same and use thereof
TWI565698B (en) * 2014-07-16 2017-01-11 財團法人生物技術開發中心 Quinoxaline compounds, method for preparing the same and use thereof
KR20170030550A (en) * 2014-07-16 2017-03-17 재단법인 생물기술개발중심 Quinoxaline compounds, method for preparing the same and use thereof
WO2016010662A1 (en) * 2014-07-16 2016-01-21 Development Center For Biotechnology Quinoxaline compounds, method for preparing the same and use thereof
KR102433758B1 (en) * 2014-07-16 2022-08-19 재단법인 생물기술개발중심 Quinoxaline compounds, method for preparing the same and use thereof
EP3169670A4 (en) * 2014-07-16 2017-05-24 Development Center for Biotechnology Quinoxaline compounds, method for preparing the same and use thereof
CN106536488A (en) * 2014-07-16 2017-03-22 财团法人生物技术开发中心 Quinoxaline compounds, method for preparing the same and use thereof
JP2017521449A (en) * 2014-07-16 2017-08-03 ディベロップメント センター フォー バイオテクノロジーDevelopment Center For Biotechnology Quinoxaline compounds, methods of preparing them and uses thereof
US11684620B2 (en) 2015-02-10 2023-06-27 Astex Therapeutics Ltd Pharmaceutical compositions comprising N-(3,5-dimethoxyphenyl)-N′-(1-methylethyl)-N-[3-(1-methyl-1H-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine
US10898482B2 (en) 2015-02-10 2021-01-26 Astex Therapeutics Ltd Pharmaceutical compositions comprising N-(3,5-dimethoxyphenyl)-N'-1 methylethyl)-N-[3-(1-methyl-1H-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine
US10478494B2 (en) 2015-04-03 2019-11-19 Astex Therapeutics Ltd FGFR/PD-1 combination therapy for the treatment of cancer
US11542247B2 (en) 2015-09-23 2023-01-03 Janssen Pharmaceutica Nv Bi-heteroaryl substitute 1,4-benzodiazepines and uses thereof for the treatment of cancer
US11155555B2 (en) 2015-09-23 2021-10-26 Janssen Pharmaceutica Nv Compounds
US10695296B2 (en) 2017-07-14 2020-06-30 Asana Biosciences, Llc Formulations, methods, kit, and dosage forms for improved stability of an active pharmaceutical ingredient
WO2019014668A1 (en) * 2017-07-14 2019-01-17 Asana Biosciences, Llc Formulations, methods, kit, and dosage forms for improved stability of an active pharmaceutical ingredient
WO2019060611A1 (en) * 2017-09-20 2019-03-28 Abm Therapeutics, Inc. Cyclic iminopyrimidine derivatives as kinase inhibitors
US11932647B2 (en) 2017-09-20 2024-03-19 Abm Therapeutics Corporation Cyclic iminopyrimidine derivatives as kinase inhibitors
JP7394768B2 (en) 2017-09-20 2023-12-08 エイビーエム セラピューティックス コーポレイション Cyclic iminopyrimidine derivatives as kinase inhibitors
CN111247152A (en) * 2017-09-20 2020-06-05 璧辰医药技术股份有限公司 Cyclic iminopyrimidine derivatives as kinase inhibitors
JP2020534373A (en) * 2017-09-20 2020-11-26 エイビーエム セラピューティックス コーポレイション Cyclic iminopyrimidine derivative as a kinase inhibitor
US11254680B2 (en) 2017-09-20 2022-02-22 Abm Therapeutics Corporation Cyclic iminopyrimidine derivatives as kinase inhibitors
RU2801302C2 (en) * 2017-09-20 2023-08-07 Абм Терапьютикс Корпорейшн Cyclic iminopyrimidine derivatives as kinase inhibitors
WO2020261156A1 (en) 2019-06-28 2020-12-30 Array Biopharma Inc. Quinazolin-4-one derivatives useful for the treatment of braf-associated diseases and disorders
RU2797606C1 (en) * 2019-06-28 2023-06-07 Аррэй Байофарма Инк. Quinazolin-4-one derivatives useful in the treatment of braf-associated diseases and disorders
CN114026073A (en) * 2019-06-28 2022-02-08 Array生物制药公司 Quinazolin-4-one derivatives for the treatment of BRAF related diseases and disorders
US11634409B2 (en) 2019-06-28 2023-04-25 Array Biopharma Inc. Compounds for the treatment of BRAF-associated diseases and disorders
AU2020302352B2 (en) * 2019-06-28 2023-03-23 Array Biopharma Inc. Quinazolin-4-one derivatives useful for the treatment of BRAF-associated diseases and disorders
US11414404B2 (en) 2019-06-28 2022-08-16 Array Biopharma Inc. Compounds for the treatment of BRAF-associated diseases and disorders
TWI817018B (en) * 2019-06-28 2023-10-01 美商艾瑞生藥股份有限公司 Compounds for the treatment of braf-associated diseases and disorders
JP2022539056A (en) * 2019-06-28 2022-09-07 アレイ バイオファーマ インコーポレイテッド Quinazolin-4-one Derivatives Useful for Treating BRAF-Related Diseases and Disorders
JP7169469B2 (en) 2019-06-28 2022-11-10 アレイ バイオファーマ インコーポレイテッド Quinazolin-4-one Derivatives Useful for Treating BRAF-Related Diseases and Disorders
CN114746405A (en) * 2019-12-10 2022-07-12 豪夫迈·罗氏有限公司 Novel BRAF inhibitors as paradoxical breakers
RU2802968C1 (en) * 2019-12-10 2023-09-05 Ф. Хоффманн-Ля Рош Аг New methylquinazolinone derivatives
CN114746405B (en) * 2019-12-10 2024-03-26 豪夫迈·罗氏有限公司 Novel BRAF inhibitors as anomalous breakers
WO2021116050A1 (en) 2019-12-10 2021-06-17 F. Hoffmann-La Roche Ag New braf inhibitors as paradox breakers
AU2020403443B2 (en) * 2019-12-10 2023-02-23 F. Hoffmann-La Roche Ag New methylquinazolinone derivatives
JP7108146B2 (en) 2019-12-10 2022-07-27 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Novel methylquinazolinone derivative
CN114787156A (en) * 2019-12-10 2022-07-22 豪夫迈·罗氏有限公司 Novel methyl quinazolinone derivatives
WO2021116055A1 (en) 2019-12-10 2021-06-17 F. Hoffmann-La Roche Ag New methylquinazolinone derivatives
JP2022531609A (en) * 2019-12-10 2022-07-07 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト New methylquinazolinone derivative
RU2814662C1 (en) * 2020-06-09 2024-03-04 Аррэй Байофарма Инк. 4-oxo-3,4-dihydroquinazolinone compounds for treatment of braf-associated diseases and disorders
WO2021250521A1 (en) * 2020-06-09 2021-12-16 Array Biopharma Inc. 4-oxo-3,4-dihydroquinazolinon compounds for the treatment of braf-associated diseases and disorders
JP2023524910A (en) * 2020-06-09 2023-06-13 アレイ バイオファーマ インコーポレイテッド 4-Oxo-3,4-dihydroquinazolinone compounds for treating BRAF-related diseases and disorders
JP7312335B2 (en) 2020-06-09 2023-07-20 アレイ バイオファーマ インコーポレイテッド 4-Oxo-3,4-dihydroquinazolinone compounds for treating BRAF-related diseases and disorders
CN113968803A (en) * 2020-07-24 2022-01-25 苏州匠化生物科技有限公司 Novel method for synthesizing 2, 6-difluoro-3-propyl sulfonamide benzoic acid
CN113968803B (en) * 2020-07-24 2023-05-09 苏州匠化生物科技有限公司 Method for synthesizing 2, 6-difluoro-3-propyl sulfonamide benzoic acid
WO2022129259A1 (en) * 2020-12-18 2022-06-23 F. Hoffmann-La Roche Ag New quinazolinone derivatives
WO2022129260A1 (en) * 2020-12-18 2022-06-23 F. Hoffmann-La Roche Ag New aryl-pyrido-pyrimidin-one derivatives
WO2022261250A1 (en) * 2021-06-08 2022-12-15 C4 Therapeutics, Inc. Therapeutics for the degradation of mutant braf
JP7289961B1 (en) 2021-06-09 2023-06-12 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Combination therapy for cancer treatment
JP2023177177A (en) * 2021-06-09 2023-12-13 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Combination therapy for cancer treatment
JP2023177209A (en) * 2021-06-09 2023-12-13 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Combination therapy for cancer treatment
WO2022258600A1 (en) 2021-06-09 2022-12-15 F. Hoffmann-La Roche Ag Combination of a particular braf inhibitor (paradox breaker) and a pd-1 axis binding antagonist for use in the treatment of cancer
JP7289978B1 (en) 2021-06-09 2023-06-12 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Combination therapy for cancer treatment
WO2022258584A1 (en) 2021-06-09 2022-12-15 F. Hoffmann-La Roche Ag New solid forms of (3r)-n-[2-cyano-4-fluoro-3-(3-methyl-4-oxo-quinazolin-6-yl)oxy-phenyl]-3-fluoro-pyrrolidine-1-sulfonamide
WO2023105371A1 (en) * 2021-12-08 2023-06-15 Array Biopharma Inc. Crystalline form of n-(2-chloro-3-((5-chloro-3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4-fluorophenyl)-3-fluoroazetidine-1-sulfonamide
TWI836777B (en) 2021-12-08 2024-03-21 美商亞雷生物製藥股份有限公司 Crystalline form of n-(2-chloro-3-((5-chloro-3-methyl-4-oxo-3,4-dihydroquinazolin-6-yl)amino)-4-fluorophenyl)-3-fluoroazetidine-1-sulfonamide
WO2024017294A1 (en) * 2022-07-19 2024-01-25 西藏海思科制药有限公司 Preparation and use of quinazolinone derivative as kinase inhibitor
WO2024054591A1 (en) * 2022-09-07 2024-03-14 Arvinas Operations, Inc. Rapidly accelerated fibrosarcoma (raf) degrading compounds and associated methods of use
US11957759B1 (en) 2022-09-07 2024-04-16 Arvinas Operations, Inc. Rapidly accelerated fibrosarcoma (RAF) degrading compounds and associated methods of use

Similar Documents

Publication Publication Date Title
WO2012118492A1 (en) Heterocyclic sulfonamides as raf inhibitors
EP2265610B1 (en) Pyrazole [3, 4-b]pyridine raf inhibitors
EP2265609B1 (en) Imdizo [4. 5-b] pyridine derivatives used as raf inhibitors
US20130018033A1 (en) Raf inhibitor compounds and methods of use thereof
US20110003859A1 (en) N- (6-aminopyridin-3-yl) -3- (sulfonamido) benzamide derivatives as b-raf inhibitors for the treatment of cancer
EP2470539A1 (en) Raf inhibitor compounds and methods of use thereof
EP2470532A1 (en) 1h-pyrazolo [ 3, 4-b]pyridine compounds for inhibiting raf kinase
US20120157439A1 (en) Raf inhibitor compounds and methods of use thereof
CA2716951A1 (en) Raf inhibitor compounds and methods of use thereof
WO2011025965A1 (en) Raf inhibitor compounds and methods of use thereof
EP2470541A1 (en) Raf inhibitor compounds and methods of use thereof

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: 11707327

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11707327

Country of ref document: EP

Kind code of ref document: A1