WO2015057938A1 - Pyrimidine fgfr4 inhibitors - Google Patents

Pyrimidine fgfr4 inhibitors Download PDF

Info

Publication number
WO2015057938A1
WO2015057938A1 PCT/US2014/060857 US2014060857W WO2015057938A1 WO 2015057938 A1 WO2015057938 A1 WO 2015057938A1 US 2014060857 W US2014060857 W US 2014060857W WO 2015057938 A1 WO2015057938 A1 WO 2015057938A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
compound
methyl
yield
added
Prior art date
Application number
PCT/US2014/060857
Other languages
French (fr)
Inventor
Dominic Reynolds
Ming-Hong Hao
John Wang
Sudeep PRAJAPATI
Takashi Satoh
Anand SELVARAJ
Original Assignee
Eisai R&D Management Co., Ltd.
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
Priority to RS20180834A priority Critical patent/RS57444B1/en
Priority to ES14790956.8T priority patent/ES2679521T3/en
Priority to CA2924206A priority patent/CA2924206C/en
Priority to JP2016523266A priority patent/JP6139788B2/en
Priority to NZ717559A priority patent/NZ717559A/en
Priority to EP14790956.8A priority patent/EP3057943B1/en
Application filed by Eisai R&D Management Co., Ltd. filed Critical Eisai R&D Management Co., Ltd.
Priority to MYPI2016000489A priority patent/MY184733A/en
Priority to UAA201605322A priority patent/UA116920C2/en
Priority to SG11201602069WA priority patent/SG11201602069WA/en
Priority to LTEP14790956.8T priority patent/LT3057943T/en
Priority to AU2014337291A priority patent/AU2014337291B9/en
Priority to SI201430792T priority patent/SI3057943T1/en
Priority to EP18167989.5A priority patent/EP3421457B1/en
Priority to KR1020167009625A priority patent/KR101826015B1/en
Priority to MX2016004933A priority patent/MX2016004933A/en
Priority to CN201480056358.4A priority patent/CN105899490B/en
Priority to PL14790956T priority patent/PL3057943T3/en
Priority to RU2016118981A priority patent/RU2715708C2/en
Priority to BR112016008110-2A priority patent/BR112016008110B1/en
Priority to DK14790956.8T priority patent/DK3057943T3/en
Publication of WO2015057938A1 publication Critical patent/WO2015057938A1/en
Priority to US15/000,659 priority patent/US9434697B2/en
Priority to IL244373A priority patent/IL244373B/en
Priority to PH12016500676A priority patent/PH12016500676A1/en
Priority to US15/221,018 priority patent/US9730931B2/en
Priority to HK16111183.6A priority patent/HK1223091A1/en
Priority to US15/627,851 priority patent/US10537571B2/en
Priority to HRP20181129TT priority patent/HRP20181129T1/en
Priority to CY181100754T priority patent/CY1121129T1/en
Priority to AU2018271284A priority patent/AU2018271284B2/en
Priority to US16/717,080 priority patent/US10912774B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0021Intradermal administration, e.g. through microneedle arrays, needleless injectors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C53/00Saturated compounds having only one carboxyl group bound to an acyclic carbon atom or hydrogen
    • C07C53/15Saturated compounds having only one carboxyl group bound to an acyclic carbon atom or hydrogen containing halogen
    • C07C53/16Halogenated acetic acids
    • C07C53/18Halogenated acetic acids containing fluorine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D403/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Dermatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Urology & Nephrology (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Oncology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Hospice & Palliative Care (AREA)
  • Food Science & Technology (AREA)
  • Biotechnology (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Provided herein are compounds of Formula I useful as FGFR4 inhibitors, as well as methods of use of the same.

Description

PYRIMIDINE FGFR4 INHIBITORS
BACKGROUND
Fibroblast growth factors (FGF) are a family of more than 20 structurally related proteins with a variety of biological activities. Their main receptors, the fibroblast growth factor receptors (FGFR1, FGFR2, FGFR3 and FGFR4), are a family of receptor tyrosine kinases that bind FGF and are involved in processes of cell proliferation and differentiation. Deregulation of FGFR signaling networks is implicated in a number of pathophysiological conditions, including many types of human cancers.
"Fibroblast Growth Factor Receptor 4" or "FGFR4" is known to regulate proliferation and antiapoptosis and is expressed or highly expressed in many cancers. See, e.g., Dieci et al. 2013, Cancer Discovery, 0F1-0F16, Studies have shown that expression of FGFR4 is predictive of a more aggressive phenotype of the cancer, and knockdown or reduction of FGFR4 expression serves to reduce proliferation and promote apoptosis. See, e.g., Wesche et al. 2011, Biochem J 437:199-213.
For example, FGFR4 expression or overexpression is associated with cancer
aggressiveness in gastric cancer (Ye et al. 2011, Cancer, 5304-5313), prostate cancer (Xu et al.
2011, BMC Cancer, 1 1;84), sarcoma such as rhabdomyosarcoma (Taylor VI et al. 2009, J Clin Invest, 119(11):3395-3407), skin cancer such as melanoma (Streit et al. 2006, British J Cancer, 94:1879-1886), liver cancer such as cholangiocarcinoma (Sia et al. 2013, Gastroenterology 144:829-840) and hepatocellular carcinoma (French et al 2012, PLoS ONE 7(5): e367313; Miura et al. 2012, BMC Cancer 12:56; Chiang et al. 2008, Cancer Res 68(16):6779-6788; Sawey et al. 2011, Cancer Cell 19:347-358), pancreatic cancer such as pancreatic intraepithelial neoplasia and pancreatic ductal adenocarcinoma (Motoda et al. 2011, Int'l J Oncol 38:133-143), lung cancer such as non-small-cell lung cancer (Fawdar et al. 2013, PNAS 110(30): 12426- 12431), colorectal cancer (Pelaez-Garcia et al. 2013, PLoS ONE 8(5): e63695; Barderas et al.
2012, J Proteomics 75:4647-4655), and ovarian cancer (Zaid et al. 2013, Clin Cancer Res 19:809-820).
Clinical development of several FGFR inhibitors have confirmed their utility as antitumor agents. Dieci et al. 2013, Cancer Discovery, OF 1 -OF 16. However, new agents are needed that are useful to target FGFR, and FGFR4, in particular. SUMMARY
A purpose of the present invention is to provide a compound of Formula I:
Figure imgf000003_0001
ΐ
wherein:
R3 is selected from the group consisting of: C1-6alkyl, Ci-6alkox Ci-6alk l, N ^R11^. 6alkyl, R10heterocyclylCi-6alkyl, R10arylC[-6alkyl, and R¾eteroarylCi.6alkyl, wherein R10 and R11 are each independently selected from the group consisting of: hydrogen and Ci-6alkyl;
E is selected from the group consisting of:
-NRI3C(0)CRI4-CHRl5 f and
~NR13C(0)C≡CR14,
wherein R13 is selected from the group consisting of: hydrogen and methyl, and R14 and R15 are each independently selected from the group consisting of; hydrogen, methyl, fluoro and chloro;
R12 is selected from the group consisting of: hydroge -6aIkoxy, hydroxyCi-ealkyl, hydroxyC1-6alkoxy}Ci-6alkoxyCi-6alkoxy,
Figure imgf000003_0002
R5R6heterocyciyl, -C(0)heterocyclylR5R6 , R5R6heterocyclylC^alkyl, NR5R6, NR5R6Ci-6alkyl, -C(0)NR5R6 5 and NRsR6Ci-6alkyoxyt wherein R5 and R6 are each independently selected from the group consisting of hydrogen, C^alkyl, hydroxyCi^aikyl, aminoCi-6alkyl,
-C(0)C1.6alkyl and
Figure imgf000003_0003
and
R1 is phenyl, wherein said phenyl is substituted 2, 3, or 4 times with independently selected halo or C1-6alkoxy,
or a pharmaceutically acceptable salt thereof. In some embodiments, R is Ci^alkyl.
In some embodiments, R3 is selected from the group consisting of: methyl, methoxyethyl, 4-pyridylmethyl, 3-pyridylmethyi, 2-pyridylmethyl, benzyl, N,N-dimethylaminopropyl, 3- methylisoxazol-5-yl-methyl, and 4-methylpiperazin-l-yl-propyl.
In some embodiments, E is -NR13C(0)CH-CHR15 or -NR,3C(0)CF=CH2, wherein R13 and R15 are as defined above. In some embodiments, E is -NHCCC CH^CHs. In some embodiments, R is selected from the group consisting of: hydrogen, fiuoro, chloro, methyl, methoxy, Ν,Ν-dimethylaminoethyl, piperazin-l-yl, 4-ethylpiperazm-l-yl, 4- ethylpiperazin-l-yl-methyl, l-methylpiperidine-4-yl, l-ethylpiperidine-4-yl, N,N- dimethylaminomethyl, N,N-dimethylammopropyl, piperidine-4-yl, morpholino, 3,5- dimethylpiperazin-l-yl, 4-(methylsuIfonyl)piperazin-l-yl, N,N-dimethylaminoethoxy, 4-(2- hydroxyethyl)piperazm-l-yl, hydroxyethoxy, methoxyethoxy, hydroxymethyl, methoxymethyl, 2-methoxypropyI, 2-hydroxypropyl, 2-aminopropyl,4-methylpiperazin-l-yl-carbonyl, 4- ethylpiperazin-l-yl-carbonyl, 4-[2-propyl]piperazin-l -yl, 4-acetylpiperazin-l-yl, N-methyl-N- hydroxyethyl-amino, Ν,Ν-dimethylamido, and 4-(2-ammoethyl)piperazin-l-yl.
In some embodiments, R12 is selected from the group consisting of: hydrogen, Ci.6alkyl, hydroxyd-ealkyl, RsR6heterocyclyl, RsR6heterocyclylCi-6alkyl, -C(0)NR5R6, NR5R6C1-6alkyl, NR5R6C1-galkyoxyJ Q^alkoxy, and Ci-galko yCi-ealkyl, whererin R5 and R6 are each independently selected from the group consisting of: hydrogen, Ci-ealkyl, hydroxyCi-6alkyl , ^C(0)Ci-6alkyI and C1-6alkylsulfonyl.
In some embodiments, R12 is R5R6heterocyclyl, wherein R5 and R6 are as defined above.
In some embodiments, R5R6heterocyclyl is R5R6piperazinyl, wherein R5 and R6 are as defined above.
In some embodiments, R is 4-ethylpiperazin-l-yl.
In some embodiments, R is not hydrogen.
In some embodiments, R1 is 2,6-dichloro-3,5-dimethoxyphenyl.
In some embodiments, the compound is a compound of Formula 1(a):
Figure imgf000004_0001
1(a) wherein R3, E, R12 and R1 are as defined above,
or a pharmaceutically acceptable salt thereof.
A further purpose is a pharmaceutical composition comprising a compound or salt as described herein and a pharmaceutically acceptable carrier. In some embodiments, the composition is formulated for oral or parenteral administration.
A further purpose is a method of treating hepatocellular carcinoma in a subject in need thereof comprising administering to said subject a treatment effective amount of a compound or salt or composition as described herein. In some embodiments, hepatocellular carcinoma has altered FGFR4 and/or FGF19 status (e.g., increased expression of FGFR4 and/or FGF19).
A further purpose is a method of treating hepatocellular carcinoma in a subject in need thereof, comprising: detecting an altered FGFR4 and/or FGF19 status (e.g., increased expression of FGFR4 and/or FGF19) in a biological sample containing cells of said hepatocellular carcinoma, and if said hepatocellular carcinoma has said altered FGFR4 and/or FGF19 status, administering a compound or composition described herein to said subject in a treatment- effective amount.
A further purpose is the use of a compound or salt or a composition as described herein in a method of treatment of hepatocellular carcinoma.
A further purpose is the use of a compound or salt described herein in the preparation of a medicament for the treatment of hepatocellular carcinoma.
BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 presents the results of in vivo efficacy testing in hepatocellular carcinoma model using HUH7 cells. Compound 108 (25 mg kg or 37.5 mg/kg) or Vehicle control was
administered via intraperitoneal injection, and tumor volume was measured twice weekly over the course of 15 days.
FIG. 2 presents the results of in vivo efficacy testing in hepatocellular carcinoma model using HEP3B cells. Compound 108 (12.5 mg/kg, 25 mg/kg or 37.5 mg/kg) or Vehicle control was administered via intraperitoneal injection, and tumor volume was measured twice weekly over the course of 15 days.
FIG. 3 presents the results of in vivo efficacy testing in hepatocellular carcinoma model using JHH7 cells. Compound 108 (12.5 mg/kg, 25 mg/kg or 37.5 mg/kg) or Vehicle control was administered via intraperitoneal injection, and tumor volume was measured twice weekly over the course of 15 days.
FIG. 4 presents the results of comparative in vivo efficacy testing in hepatocellular carcinoma model using HEP3B cells. Compound 108 (25 mg/kg, 37.5 mg/kg or 50 mg/kg) was administered twice daily via intraperitoneal injection, or BGJ398 (30 mg/kg or 60 mg kg) was administered orally twice daily.
DETAILED DESCRIPTION OF EMBODIMENTS
Provided herein are compounds useful as FGFR4 inhibitors. In some embodiments, the compounds are selective FGFR4 inhibitors in that they have a greater binding affinity and/or inhibitory effect of FGFR4 as compared to that of FGFRl and/or FGFR2 and/or FGFR3 (e.g., by 10-fold, 100-fold, or 1000-fold greater or more).
A. Definitions
Compounds useful as active agents in accordance with the present disclosure include those described generally above and below, and are further illustrated by the embodiments, sub- embodiments, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated.
As described herein, compounds of the invention may optionally be substituted with one or more substituents, such as those illustrated generally herein, or as exemplified by particular classes, subclasses, and species of the invention. In general, the term "substituted" refers to the replacement of hydrogen in a given structure with a specified substituent. Unless otherwise indicated, a substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable compounds. "Stable" as used herein refers to chemically feasible compound is one that is not substantially altered when kept at a temperature of 40 °C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
As would be understood by those of skill in the art, as used herein "H" is hydrogen, "C" is carbon, "N" is nitrogen, "S" is sulfur, and "O" is oxygen.
"Alkyl" or "alkyl group," as used herein, means a straight-chain (i.e., unbranched), or branched hydrocarbon chain that is completely saturated. In some embodiments, the alkyl has 1, 2, 3, 4, 5 or 6 carbon atoms. In certain embodiments, alkyl groups contain 1-6 carbon atoms (Ci_ 6alkyl). In certain embodiments, alkyl groups contain 1-4 carbon atoms (C1-4alkyl). In certain embodiments, alkyl groups contain 1-3 carbon atoms (Ci-jalkyi). In still other embodiments, alkyl groups contain 2-3 carbon atoms (Chalky!), and in yet other embodiments alkyl groups contain 1-2 carbon atoms (Ci-2alkyl).
"Alkenyl" or "alkenyl group," as used herein, refers to a straight-chain (i.e., unbranched), or branched hydrocarbon chain that has one or more double bonds. In some embodiments, the alkenyl has 2, 3, 4, 5 or 6 carbon atoms. In certain embodiments, alkenyl groups contain 2-8 carbon atoms (C2-8alkyl). In certain embodiments, alkenyl groups contain 2-6 carbon atoms (C2- 6alkyl). In still other embodiments, alkenyl groups contain 3-4 carbon atoms (Cs^ lkyl), and in yet other embodiments alkenyl groups contain 2-3 carbon atoms (C2.3 alkyl). According to another aspect, the term alkenyl refers to a straight chain hydrocarbon having two double bonds, also referred to as "diene." Non-limiting examples of exemplary alkenyl groups include
-CH=CH2, ~CH2CH=CH2j -CH-CHCH3, -CH2CH2CH=CH2,
~CH2CH=CHC¾, -CH-CHCH2CH3} and -CH=CHCH=C¾.
"Alkynyl" or "alkynyl group" as used herein refers to a straight-chain (i.e., unbranched), or branched hydrocarbon chain that has one or more triple bonds. In some embodiments, the alkynyl has 2, 3, 4, 5 or 6 carbon atoms. In certain embodiments, alkynyl groups contain 2-8 carbon atoms (C2.g lkynyl). In certain embodiments, alkynyl groups contain 2-6 carbon atoms (C2-6alkynyl). In still other embodiments, alkynyl groups contain 3-4 carbon atoms (Ca^alkynyl), and in yet other embodiments alkynyl groups contain 2-3 carbon atoms (C2-3alkynyl).
"Ar" or "aryl" refer to an aromatic carbocyclic moiety having one or more closed rings. Examples include, without limitation, phenyl, naphthyl, anthracenyl, phenanthracenyl, biphenyl, and pyrenyl.
"Halo" refers to chloro (CI), fluoro (F), bromo (Br) or iodo (I).
"Haloalkyl" refers to one or more halo groups appended to the parent molecular moiety through an alkyl group. Examples include, but are not limited to, chloromethyl, fluoromethyl, trifiuoromethyl, etc.
"Heteroaryl" refers to a cyclic moiety having one or more closed rings, with one or more heteroatoms (oxygen, nitrogen or sulfur) in at least one of the rings, wherein at least one of the rings is aromatic, and wherein the ring or rings may independently be fused, and/or bridged. Examples include, without limitation, quinolinyl, isoquinolinyl, indolyl, furyl, thienyl, pyrazoiyl, quinoxalinyl, pyrrolyl, indazolyl, thieno [2, 3 -c] pyrazoiyl, benzofuryl, pyrazolo[l,5-a]pyridyl, thiophenylpyrazolyl, benzothienyl, benzothiazolyl, thiazolyl, 2-phenylthiazolyl, and isoxazolyl.
"-OR" or "oxy" refers to an R group appended to the parent molecular moiety through an oxygen atom, wherein R is H, alkyl, alkenyl, alkynyl, and the like.
" Alkoxy" refers to an alkyl group, as herein defined, attached to the principal carbon chain through an oxygen ("alkoxy") atom. Representative examples of "alkoxy" include, but are not limited to, methoxy, ethoxy, propoxy, phenoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, hexyloxy and the like.
"Hydroxy" refers to an -OH group.
"Carbonyl" is a group having a carbon atom double-bonded to an oxygen atom (C-O), often depicted in chemical formulae as C(0).
An "acetyl" is a group -C(0)C¾.
An "amine" or "aimno" refers to a group ~~NH2, wherein none, one or two of the hydrogens may replaced by a suitable substituent as described herein, such as alkyl, aikenyl, alkynyl, and the like.
An "amide" or "amido" refers to a group having a carbonyl bonded to a nitrogen atom, such as -~C(0)NH2, wherein none, one or two of the hydrogens may replaced by a suitable substituent as described herein, such as alkyl, aikenyl, alkynyl, and the like.
"-SR" refers to an R group appended to the parent molecular moiety through a sulfur atom, wherein R is alkyl, aikenyl, alkynyl, aryl, cycloalkyl, heterocyclo, or heteroaryl.
Representative examples of "-SR" include, but are not limited to, ethanethiol, 3 -methyl- 1- butanethiol, phenylthiol and the like.
"Cycloalkyl" as used herein, refers to a saturated cyclic hydrocarbon group containing from 3 to 8 carbons or more. Representative examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
"Cycloalkenyl" as used herein, refers to an unsaturated cyclic hydrocarbon group containing from 3 to 8 carbons or more and having one or more double bonds.
"Cycloalkynyl" as used herein, refers to an unsaturated cyclic hydrocarbon group containing from 3 to 8 carbons or more and having one or more triple bonds.
"Electrophile" as used herein refers to a group having reduced electron density, typically comprising a carbon atom that is directly bonded to a more electronegative atom, such as an oxygen, nitrogen or halo. Exemplary electrophiles include, but are not limited to, diazomethane, trimethylsilyldiazomethane, alkyl halides, such as for example methyl iodide, benzyl bromide and the like, alkyl triflates, such as for example methyl triflate and the like, alkyl sulfonates, such as for example ethyl toluenesulfonate, butyl methanesulfonate and the like, acyl halides, such as for example acetyl chloride, benzoyl bromide and the like, acid anhydrides, such as for example acetic anhydride, succinic anhydride, maleic anhydride and the like, isocyanates, such as for example methyl isocyanate, phenyl isocyanate and the like, isothiocyanates, such as for example methyl isothiocyanate, phenyl isothiocyanate and the like, chloroformates, such as for example methyl chloroformate, ethyl chloroformate, benzyl chloroformate and the like, sulfonyl halides, such as for example methanesulfonyi chloride, methanesulfonyl fluoride, p-toluene sulfonyl chloride and the like, silyl halides, such as for example trimethylsilyl chloride, tert- butyldimethylsilyl chloride and the like, phosphoryl halides such as for example dimethyl chlorophosphate and the like, epoxides such as for example 2-methyloxirane, aziridines such as for example 2-methylaziridine, alpha-ha oketone such as for example l-chloro-2-propanone, alpha-beta-unsaturated carbonyl compounds such as for example acrolein, methyl vinyl ketone, cinnamaldehyde, Ν,Ν-dimethylacr lamide and the like, and gamma-halo-alpha-beta-unsaturated carbonyl compounds such as for example (E)-6-chlorohex-4-en-3-one, In some embodiments, electrophiles are alpha-haloketones, isothiocyanates, epoxides, aziridines, sulfonyl halides, or alpha-beta-unsaturated carbonyls.
In some embodiments, the electrophile is a Michael acceptor. As known in the art, a
"Michael acceptor" is an alkene or alkyne of the form 2 ; wherein Z comprises an electron withdrawing group, including, but not limited to, CHO, COR, COOR, CONRR', CONROR', CN, N02, SOR, S02R. R may be H, alkyl, or aryl; wherein R is alkyl, alkenyl, alkoxy or aryl. In another embodiment, azodicarboxamides and quinones are Michael acceptors. See, Santos, M.M.M. and Moreira, R., Mini-Reviews in Medicinal Chemistry, 7:1040-1050, 2007.
An example of the Mich l cti n is de i in he hem low:
Figure imgf000009_0001
Michae onor Michael acceptor wherein electron withdrawing groups Z, Z' and Z" are as described above. In some embodiments, the Michael acceptors are alpha-beta-unsaturated carbonyl compounds including, but not limited to, alpha-beta-unstaturated amides, alpha-beta-unstaturated ketones, alpha-beta-unstaturated esters, conjugated alkynyl carbonyls and alpha-beta-unsaturated nitriles.
"Alpha-beta-unsaturated amide" or "unsaturated amide" as used herein refers to an amide comprising an alkene or alkyne bonded directly to the amide carbonyl group and is represented
by the structure
Figure imgf000009_0002
wherein R is hydrogen or alkyl.
"Heteroatom" refers to O, S or N.
"Heterocycle" or "heterocyclyl" as used herein, means a monocyclic heterocycle, a bicyclic heterocycle, or a tricyclic heterocycle containing at least one heteroatom in the ring.
The monocyclic heterocycle is a 3-, 4-, 5-, 6-, 7, or 8-membered ring containing at least one heteroatom independently selected from the group consisting of O, N, and S. In some embodiments, the heterocycle is a 3- or 4-membered ring containing one heteroatom selected from the group consisting of O, N and S. In some embodiments, the heterocycle is a 5-membered ring containing zero or one double bond and one, two or three heteroatoms selected from the group consisting of O, N and S. In some embodiments, the heterocycle is a 6-, 7-, or 8-membered ring containing zero, one or two double bonds and one, two or three heteroatoms selected from the group consisting of O, N and S. Representative examples of monocyclic heterocycle include, but are not limited to, azetidinyl, azepanyl, aziridinyl, diazepanyl, 1 ,3-dioxanyl, 1,3-dioxolanyl, dihydropyranyl (including 3,4-dihydro-2H-pyran-6-yl), 1,3-dithiolanyl, 1,3-dithianyl, imidazolinyl, imidazolidinyl, isotliiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolinyl, oxadiazolidinyl, oxazolinyl, oxazolidinyl, piperazinyl, piperidinyl, pyranyl, pyrazolinyl, pyrazolidinyl, pyrrolinyi, pyrrolidinyl, tetrahydrofuranyl, tetrahydropyranyl (including tetrahydro-2H-pyran-4-yl), tetrahydrothienyl, thiadiazolinyl, thiadiazolidinyl, thiazolinyl, thiazolidinyl, thiomorpholinyl, 1,1-dioxidothiomorpholinyl (thiomorpholine sulfone), thiopyranyl, and trithianyl.
The bicyclic heterocycles of the present invention may be exemplified by a monocyclic heterocycie fused to an aryl group, or a monocyclic heterocycle fused to a monocyclic cycloalkyl, or a monocyclic heterocycle fused to a monocyclic cycloalkenyl, or a monocyclic heterocycle fused to a monocyclic heterocycle. Representative examples of bicyclic heterocycles include, but are not limited to, 3,4-dihydro-2H-pyranyl, 1,3-benzodioxolyL 1,3-benzodithiolyl, 2,3-dihydro-l,4-benzodioxinyl, 2,3-dihydro-l-benzofuranyl, 2,3-dihydro-l-benzothienyl5 2,3- dihydro-lH-indolyl, 3,4-dihydroquinolin-2(lH)-one and 1,2,3,4- tetrahydroquinolinyl.
The tricyclic heterocycle is a bicyclic heterocycle fused to an aryl group, or a bicyclic heterocycle fused to a monocyclic cycloalkyl, or a bicyclic heterocycle fused to a monocyclic cycloalkenyl, or a bicyclic heterocycle fused to a monocyclic heterocycle. Representative examples of tricyclic heterocycles include, but are not limited to, 2,3,4,4a,9,9a-hexahydro~lH- carbazolyl, 5a;6,7,8,9;9a-hexahydrodibenzo[b,d]furanyl, and Sa^ ^^^a-he ahydrodibenzoib, d]thienyl.
In the above heteroaryl and heterocycles the nitrogen or sulfur atoms can be optionally oxidized to various oxidation states. In a specific example, the group S(0)o-2 refers to -S- (sulfide), -S(0)- (sulfoxide), and -S02- (sulfone) respectively. For convenience, nitrogens, particularly but not exclusively, those defined as annular aromatic nitrogens, are meant to include those corresponding N-oxide forms.
"Pharmaceutically acceptable salt" as used herein refer to acid addition salts or base addition salts of the compounds in the present disclosure. A pharmaceutically acceptable salt is any salt which retains the activity of the parent compound and does not impart any unduly deleterious or undesirable effect on a subject to whom it is administered and in the context in which it is administered. Pharmaceutically acceptable salts include, but are not limited to, metal complexes and salts of both inorganic and carboxylic acids. Pharmaceutically acceptable salts also include metal salts such as aluminum, calcium, iron, magnesium, manganese and complex salts. In addition, pharmaceutically acceptable salts include, but are not limited to, acid salts such as acetic, aspartic, alkylsulfonic, arylsulfonic, axetil, benzenesulfonic, benzoic, bicarbonic, bisulfuric, bitartaric, butyric, calcium edetate, camsylic, carbonic, chlorobenzoic, citric, edetic, edisylic, estolic, esyl, esylic, formic, fumaric, gluceptic, gluconic, glutamic, glycolic, glycolylarsanilic, hexamic, hexylresorcinoic, hydrabamic, hydrobromic, hydrochloric, hydroiodic, hydroxynaphthoic, isethionic, lactic, Iactobionic, maleic, malic, malonic, mandelic, methanesulfonic, methylmtric, methyl sulfuric, mucic, muconic, napsylic, nitric, oxalic, p-mtromethanesulfonic, pamoic, pantothenic, phosphoric, monohydrogen phosphoric, dihydrogen phosphoric, phthalic, polygalactouronic, propionic, salicylic, stearic, succinic, sulfamic, sulfanlic, sulfonic, sulfuric, tannic, tartaric, teoclic, toluenesulfonic, and the like.
Pharmaceutically acceptable salts may be derived from amino acids including, but not limited to, cysteine. Methods for producing compounds as salts are known to those of skill in the art {see, e.g., Stahl et al., Handbook of Pharmaceutical Salts: Properties, Selection, and Use, Wiley-VCH; Verlag Helvetica Chimica Acta, Zurich, 2002; Berge et al., J. Pharm. Sci. 66: 1, 1977).
Unless indicated otherwise, nomenclature used to describe chemical groups or moieties as used herein follow the convention where, reading the name from left to right, the point of attachment to the rest of the molecule is at the right-hand side of the name. For example, the group "arylCi-6alkyl" is attached to the rest of the molecule at the alkyl end.
Unless indicated otherwise, where a chemical group is described by its chemical formula, including a terminal bond moiety indicated by it will be understood that the attachment is read from left to right. For example, -C(0)Ci-6alkyl is attached to the rest of the molecule at the carbonyl end.
Unless otherwise stated, structures depicted herein are also meant to include all enantiomeric, diastereomeric, and geometric (or conformational) forms of the structure; for example, the and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. In addition, unless otherwise stated, all rotamer forms of the compounds of the invention are within the scope of the invention. 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 having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C -enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools or probes in biological assays.
"Isomers" refer to compounds having the same number and kind of atoms and hence the same molecular weight, but differing with respect to the arrangement or configuration of the atoms. It will be understood, however, that some isomers or racemates or others mixtures of isomers may exhibit more activity than others. "Stereoisomers" refer to isomers that differ only in the arrangement of the atoms in space. "Diastereoisomers" refer to stereoisomers that are not mirror images of each other. "Enantiomers" refers to stereoisomers that are non-superimposable mirror images of one another.
In some embodiments, enantiomeric compounds taught herein may be "enantiomerically pure" isomers that comprise substantially a single enantiomer, for example, greater than or equal to 90%, 92%, 95%, 98%, or 99%, or equal to 100% of a single enantiomer.
In some embodiments, enantiomeric compounds taught herein may be stereomerically pure. "Stereomerically pure" as used herein means a compound or composition thereof that comprises one stereoisomer of a compound and is substantially free of other stereoisomers of that compound. For example, a stereomerically pure composition of a compound having one chiral center will be substantially free of the opposite enantiomer of the compound. A
stereomerically pure composition of a compound having two chiral centers will be substantially free of diastereomers, and substantially free of the opposite enantiomer, of the compound. A typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of the other stereoisomers of the compound, more preferably greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, even more preferably greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, and most preferably greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound. See, e.g., US Patent No. 7, 189,715.
"R" and "S" as terms describing isomers are descriptors of the stereochemical configuration at an asymmetrically substituted carbon atom. The designation of an
asymmetrically substituted carbon atom as "R" or "S" is done by application of the Cahn-Ingold- Prelog priority rules, as are well known to those skilled in the art, and described in the
International Union of Pure and Applied Chemistry (IUPAC) Rules for the Nomenclature of Organic Chemistry. Section E, Stereochemistry.
"Enantiomeric excess" (ee) of an enantiomer is [(the mole fraction of the major enantiomer) minus (the mole fraction of the minor enantiomer)] x 100. B. Compounds
Provided herein as active agents according to some embodiments is a compound of Formula I:
Figure imgf000013_0001
wherein:
is selected from the group consisting of: Chalky!,
Figure imgf000013_0002
NR L1100RRnn'Ci.
6alkyl, R'uheterocyclylC1-6alkyl, Rluaryl 1-6alkyl, and RluheteroarylCwalk l, wherein R10 and R11 are each independently selected from the group consisting of: hydrogen and Cj-6alkyl;
E is selected from the group consisting of:
-NRt3C(0)CR14=CHR15, and
-NRi3C(0)C≡CR14,
wherein R13 is selected from the group consisting of: hydrogen and methyl, and R14 and R15 are each independently selected from the group consisting of: hydrogen, methyl, fluoro and chloro;
R12 is selected from the group consisting of: hydrogen, halo, Ci-6alkyl, C^alkoxy, hydroxyC(-6alkyl, hydroxyCj-ealkox A^alkoxyCi-galkoxy, Ci^alkoxy ^alkyL
R5R6heterocyclyl, -C(0)heterocyclylR5R6 , RsR6heterocyclylC1-6alkyl, NR5R6, NR5R6C1.6alkyl, -C(0)NR5R6 s and NR5R6Ci-6aikyoxy, wherein R5 and R6 are each independently selected from the group consisting of hydrogen, Ci^alkyl, hydroxyCi^aU yl, aminoCj-6alkyl5
-C(0)Ci_6alkyl and Ct-ealkylsulfonyl; and
R1 is phenyl, wherein said phenyl is substituted 2, 3, or 4 times with independently selected halo or Q.galkoxy, or a pharmaceutically acceptable salt thereof. In some embodiments, R3 is ^alkyl.
In some embodiments, R3 is selected from the group consisting of: methyl, methoxyethyl,
4-pyridylmethyl, 3-pyridylmethyl, 2-pyridylmethyl} benzyl, N,N-dimethylaminopropyl, 3- methylisoxazol-5-yl-methyl, and 4-methylpiperazin-l-yl-propyl.
In some embodiments, E is -NR13C(0)CH=CHR15 or -NR33C(0)CF-CH2, wherein R° and R15 are as defined above. In some embodiments, E is -NHC(0)CH=C¾.
In some embodiments, R12 is selected from the group consisting of: hydrogen, fluoro, chloro, methyl, methoxy, Ν,Ν-dimethylaminoethyl, piperazin-l-yl, 4-ethylpiperazin-l-yl, 4- ethylpiperazin-l-yl-methyl, l-methylpiperidine-4-yi5 l-ethylpiperidine-4-yl, N,N- dimethylaminomethyl, Ν,Ν-dimethylaminopropyl, piperidine-4-yl, morpholino, 3,5- dimethylpiperazin-l-yl, 4-(methylsulfonyl)piperazm-l-yl, Ν,Ν-dimethylaminoethoxy, 4-(2- hydroxyethyl)piperazin-l-yl, hydroxyethoxy, methoxyethoxy, hydroxymethyl, methoxymethyl, 2-methoxypropyl, 2-hydroxypropyl, 2-aminopropyl,4-methylpiperaz; n-l-yl-carbonyl, 4- ethylpiperazin-l-yl-carbonyl, 4-[2-propyl]piperazin-l-yl, 4-acetylpiperazin-l-yl, N-methyl-N- hydroxyethyl-amino, N,N-dimethylamido, and 4-(2-aminoethyl)piperazin-l-yl.
In some embodiments, R12 is selected from the group consisting of: hydrogen, Ci-6alkyl, hydroxyC3-6alkyl, R5R6heterocyclyl, R5R6heterocyclylC1-6alkyl, -C(0)NR5R6, NRsR6C1-6alkyl, NR5R6C1-6alkyoxy, Ci-6alkoxy, and Ci-ealko yC^alk l, whererin R5 and R6 are each independently selected from the group consisting of: hydrogen, Ci-6alkyl, hydroxyCj-ealkyl , -C(0)C1-6alkyl and CwaIkylsulfonyl.
In some embodiments, R12 is R5R6heterocyclyi5 wherein R5 and R6 are as defined above.
In some embodiments, R5R6heterocycIyl is R5R6piperazinyl, wherein R5 and R6 are as defined above.
In some embodiments, R12 is 4-ethylpiperazin-l-yl.
In some embodiments, R12 is not hydrogen.
In some embodiments, R1 is 256-dichIoro-3,5-dimethoxyphenyl
In some embodiments, the compound is a compound of Formula 1(a):
Figure imgf000014_0001
1(a) wherein R3, E, R!2 and R1 are as defined above,
or a pharmaceutically acceptable salt thereof.
C. Pharmaceutical formulations
Active agents of the present invention can be combined with a pharmaceutically acceptable carrier to provide pharmaceutical formulations thereof. The particular choice of carrier and formulation will depend upon the particular route of administration for which the composition is intended.
"Pharmaceutically acceptable carrier" as used herein refers to a nontoxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention include, but are not limited to, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrroHdone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene glycol and wool fat.
The compositions of the present invention may be suitable for parenteral, oral, inhalation spray, topical, rectal, nasal, buccal, vaginal or implanted reservoir administration, etc. In some embodiments, the formulation comprise ingredients that are from natural or non-natural sources. In some embodiments, the formulation or carrier may be provided in a sterile form. Non-limiting examples of a sterile carrier include endotoxin-free water or pyrogen-free water.
The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. In particular embodiments, the compounds are administered intravenously, orally, subcutaneously, or via intramuscular administration. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
For this purpose, any bland fixed oil may be employed including synthetic mono- or di- glycerides. Fatty acids and their glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically acceptable oils, such as olive oil or castor oil, especially in their pol oxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents that are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
For oral administration, a compound or salt may be provided in an acceptable oral dosage form, including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, may also be added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient may be combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added. In addition preservatives may also be added. Suitable examples of pharmaceutically acceptable preservatives include, but are not limited to, various antibacterial and antifungal agents such as solvents, for example ethanol, propylene glycol, benzyl alcohol, chlorobutanol, quaternary ammonium salts, and parabens (such as methyl paraben, ethyl paraben, propyl paraben, etc.).
D. Subjects and methods of use
Active agents of the present invention may be used to treat hepatocellular carcinoma.
"Treatment," "treat," and "treating" refer to reversing, alleviating, delaying the onset of, inhibiting the progress of, or otherwise ameliorating a disease or disorder as described herein. In some embodiments, treatment may be administered after one or more symptoms have developed, In other embodiments, treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors).
Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence.
"Patient" or "subject", as used herein, means an animal subject, preferably a mammalian subject, and particularly human subjects (including both male and female subjects, and including neonatal, infant, juvenile, adolescent, adult and geriatric subjects). Subjects may also include other mammalian subjects (e.g., dog, cat, horse, cow, sheep, goat, monkey, bird, etc.), for laboratory or veterinary purposes.
In some embodiments, treatment is provided to a subject having hepatocellular carcinoma with altered FGFR4 and /or FGF19 (fibroblast growth factor 19) status.
In some embodiments, treatment may include or be performed in conjunction with analyzing FGFR4 and/or FGF19 status in a biological sample containing cells of said
hepatocellular carcinoma, and if said hepatocellular carcinoma exhibits an FGFR4 and/or FGF19 alteration, treating a subject with a treatment effective amount of an active agent as described herein.
"Altered status" as used herein with reference to FGFR4 and/or FGF19 includes an increased expression thereof (e.g., increased levels of the mRNA or increased levels of the protein), increased copy number in the genome, and/or increased activity of the encoded protein as a result of mutation, etc., as compared to a corresponding non- cancerous tissue. In some embodiments, altered status of FGFR4 and/or FGF19 includes gene and/or encoded protein mutations that result in an increase in activity or are otherwise associated with a more aggressive form of hepatocellular carcinoma.
"Expression" of FGFR4 and/or FGF19 means that a gene encoding the same is transcribed, and preferably, translated. Typically, expression of a coding region will result in production of the encoded polypeptide.
The FGFR4 and FGF19 proteins are known, and their altered status and/or expression may be measured using techniques standard in the art, e.g., genomic analysis of mutations or copy number aberrations such as by nucleic acid amplification, sequencing analysis, and/or hybridization-based techniques, RNA expression analysis such as northern blot or qRT-PCR, western blot or other immunoblot or immunoassay, fluorescent activated cell sorting (FACS), etc.
In order that the invention described herein may be more fully understood, the following examples are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting,
EXAMPLES
General:
Microwave heating was done using a Biotage Emrys Liberator or Initiator microwave. Column chromatography was carried out using an Isco Rf200d. Solvent removal was carried out using either a Buchi rotary evaporator or a Genevac centrifugal evaporator. Preparative LC/MS was conducted using a Waters autopurifier and 19 x 100mm XTe ra 5 micron MS CI 8 column under acidic mobile phase conditions. NMR spectra were recorded using a Varian 400MHz spectrometer.
When the term "inerted" is used to describe a reactor (e.g., a reaction vessel, flask, glass reactor, and the like) it is meant that the air in the reactor has been replaced with an essentially moisture-free or dry, inert gas (such as nitrogen, argon, and the like), General methods and experimentals for preparing compounds of the present invention are set forth below. In certain cases, a particular compound is described by way of example.
However, it will be appreciated that in each case a series of compounds of the present invention were prepared in accordance with the schemes and experimentals described below.
Preparative HPLC Conditions for the Purification of Target Compounds
Chromatography Conditions:
Instrument: Waters 2767-SQD Mass trigger Prep System
Column : Waters Xbridge C 18 150mm* 19mm* 5 μπι
Detector: VWD SQD
Flow Rate : 15 rnL/min
Gradient Time:
Time(min) B%
0 5
7.5 70
8 95
1 1 95
Representative Mobile Phase:
1)
Mobile Phase: A: 0.1%TFA in water
Mobile Phase: B: ACN
2)
Mobile Phase: A: 0.1%NH4HC03 in water
Mobile Phase: B: ACN
3)
Mobile Phase: A: 0.1 %NH OAc in water
Mobile Phase: B: ACN
4)
Mobile Phase: A: 0.1%NH4OH in water
Mobile Phase: B: ACN
Definitions: The following abbreviations have the indicated meanings:
ACN: Acetonitrile
Boc20: Di-tert-butyl dicarbonate
Brettphos: 2-(Dicyclohexylphosphino)-356-dimethoxy-2'J4',6'-triisopropyl-lj -biphenyl tBuONa: Sodium tert-butoxide
CH3I: Iodomethane
CS2CO3: Cesium carbonate
D CC : N, ' -dicy clohexy lcarbodiimide
DCM: Dichloromethane DIEA; Ν,Ν-diisopropylemylamine
DIPEA: Ν,Ν-diisopropyiethylamine
DMAP: 4-(Din ethylamino)pyridine
DME: Dimethyl ether
DMF: Dimethylformamide
DMSO: Dimethyl sulfoxide
EGTA: Ethylene glycol tetraacetic acid
ES1-MS: Electrospray ionization - mass spectrometry
EtOH: Ethanol
HATU: 1 - [Bis(dimethylamino)methylene]~ 1 H- 1 ,2,3 -trizolo[4,5 -b]pyridinium 3-oxid hexafmorophosphate
¾S04: Sulfuric acid
iPrOH: Isopropanol
K2CO3 : Potassium carbonate
HMDS: Potassium bis(trimethylsilyl)amide
OH: Potassium hydroxide
LCMS: Liquid chromatography - mass spectrometry
MeOH: Methanol
MsCl: Methansulfonyl chloride
NaBH3CN: Sodium cyanoborohydride
NaBH(OAc)3; Sodium triacetoxyborohydride
NH4C1: Ammonium chloride
NH4HCO3: Ammonium bicarbonate
Nal: Sodium iodide
NaN03: Sodium nitrate
NaOAc: Sodium acetate
nBuOH: n-Butanol
prep-HPLC: Preparative high-performance liquid chromatography
prep-TLC: Preparative thin layer chromatography
TBAF: Tetrabutylammonium fluoride
TBDMS-CL: tert-Butyldimethylsilyl chloride
TBSC1: tert-Butyldimethylsilyl chloride
TBSOTf: tert-Butyldimethylsilyl trifluoromethanesulfonate
TEA: Triethylamine TESC1: Chiorotriethylsilane
TFA: Trifluoroacetic acid
THF: Tetrahydrofuran
ΤΪ(0!ΡΓ)4: Titanium isopropoxide
TLC: Thin-layer chromatography
PPTS: Pyridinium p-toluenesulfonate
PE: Petroleum ether
PEG: Poly(ethylene glycol)
Pt02: platinum dioxide
EtOAc: Ethyl acetate
Pd/C: Palladium (0) on carbon
Pd2(dba)3.- Tris(dibenzylideneacetone) dipalladium(O)
Pd(dppf)2Cl2: [l,l'-Bis(diphenylphosphino)ferrocene]dichloropalladium(II)
Ruphos : 2-Dicyclohexylphosphino-2'J6'-diisopropoxybiphenyl
Xantphos: 4,5-Bis(diphenylphosphino)-9J9-dimethyIxanthene
Materials: The following compounds are commercially available and/or can be prepared in a number of ways well known to one skilled in the art of organic synthesis. More specifically, disclosed compounds can be prepared using the reactions and techniques described herein. In the description of the synthetic methods described below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of the experiment, and workup procedures, can be chosen to be the conditions standard for that reaction, unless otherwise indicated. It is understood by one skilled in the art of organic synthesis that the functionality present on various portions of the molecule should be compatible with the reagents and reactions proposed. Substituents not compatible with the reaction conditions will be apparent to one skilled in the art, and alternate methods are therefore indicated. The starting materials for the examples are either commercially available or are readily prepared by standard methods from known materials. SYNTHESIS AND TESTING OF EXAMPLE COMPOUNDS
Compounds of Table 1 were prepared by the Procedures of 2A- 2L.
TABLE 1
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
Figure imgf000026_0001
o
136 < 0.001 0.112
0
137 < 0.001 >10.0
0
139 0.006 >20.0
OMe
0
ιΓΤΥτι 140 < 0.001 >20.0
'" 0
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Procedure 2A: Example
Figure imgf000035_0002
yield'. 95% ^Q-^^Q^ yield: 34% yield: 76% o
Figure imgf000035_0003
N-iZ-ie-p-^-dichloro-S^-dimethoxy-phenylH^^
phenyl)-acry lamid e
Figure imgf000036_0001
a. N-(3,5→Dimethoxy-phenyl)-acetamide
To a solution of 3,5-dimethoxy-phenylamine (20 g, 0.131 mol) in toluene (110 mL) was added acetic anhydride (14 g, 0.137 mmol) at room temperature. The resulting mixture was stirred for 18 hours at room temperature. PE (55 mL) was added, the precipitate was filtered and washed with PE (100 mL) to obtain the title compound (24.2 g, yield: 95%). 1H-NMR (400 MHz, CDCI3) 6 2.16 (s, 3H), 3.77 (s, 6H), 6.23 (s, 1H), 6.75 (s, 2H), 7.20 (s, 1H).
O O HN^ HN^
fS -^- CIYVCI b. N-(2,6-Dichloro-3,S-dimethoxy-phenyl)-acetamide
To a solution of N-(3,5-dimethoxy-phenyl)-acetamide (5 g, 25.6 mmol) in ACN (75 mL) was added suifuryl chloride (6,9 g, 51.2 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 30 minutes at this temperature and quenched with saturated aqueous NaHC03 (40 mL). The precipitate was filtered, washed with water and dried to obtain the title compound (2.3 g, yield: 34%). 1H-NMR (400 MHz, CDC13) 6 2.25 (s, 3H), 3,86 (s, 6H)5 6.54 (s, 1H), 6.90 (s, 1H).
Figure imgf000036_0002
c. 2, 6-Dich loro-3, 5-dimeth oxy-phenyl min e
A solution of N-(2?6-dichloro-3J5-dimethoxy-phenyl)-acetamide (3.6 g, 13.7 mmol) in EtOH (130 mL) and KOH (2M, 75 mL) was heated to reflux for 24 hours. The reaction was cooled to 0 °C and stirred for 1 hour at this temperature. The precipitate was filtered and dried to obtain the title compound (2.3 g, yield: 76%). 1H-NMR (400 MHz, CDC13) δ 3.90 (s, 6H), 4.57 (bs, 2H), 6.05 (s, 1H).
Figure imgf000037_0001
d. 2, 4-Dichloro-3-isocyanafo-l, 5-dimethoxy-benzene
A mixture of 2,6-dichioro- 3,5-dimethoxy-phenylamine (500 mg, 2.25 mmol), triphosgene (335 mg, 1.12 mmol) and TEA (342 mg, 3.38 mmol) in dioxane (15 mL) was heated to 130 °C for 2 hours under microwave. The reaction was concentrated and the residue was purified by flash chromatography on silica eiuting with DCM to obtain the title compound (450 mg, yield: 80%). 1H-NMR (400 MHz, CDC13) δ 3.92 (s, 6H), 6.42 (s, 1H).
N^ tPrOH N
C ^^-^^CI methyiamine in THF Cl N'
H
e. ( 6-Ch loro-pyrimidin~4~yl)~methyl-amine
To a solution of 4,6-dic oro-pyrimidine (7.45 g, 50 mmol) in iPrOH (50 mL) was added a solution of methyl amine in THF (2M, 30 mL, 60 mmol) at room temperature. The resulting mixture was stirred for 18 hours. The mixture was concentrated and the residue was purified by flash chromatography on silica eiuting with DCM:EtOAc = 6:1-1 :1 to obtain the title compound (4.4 g, yield: 62%) as a white solid. lU NMR (400 MHz, CDC13) δ 2.96 (d, 3H), 5.22-5.36 (bs, 1H), 6.35 (s, 1H), 8.35 (s, 1H); MS (ESI): 144 [M+H]+.
Figure imgf000037_0002
N-Methyl~N'~(2-nitro-phenyl)-pyrimidine~4,6-diamine
A mixture of (6-chloro-pyrimidin-4-yl)-methyl-amine (1 g, 7 mmol), 2-nitro-phenylamine (965 mg, 7 mmol), Brettphos (279 mg, 0.35 mmol) and tBuONa (2 g, 21 mmol) in DME (50 mL) was heated to 90 °C for 1 hour under nitrogen atmosphere. The reaction was concentrated, and the residue was purified by flash chromatography on silica eiuting with DCM:EtOAc = 10:1—1 : 1 to obtain the title compound (600 mg, yield: 35%) as a yellow solid. 1H NMR (400 MHz, CDC13) δ 2.94 (d, 3H), 4.99 (bs, 1H), 5.82 (s, ΪΗ), 7.04 (t, 1H), 7.60 (t, 1H), 8.21 (d, 1H), 8.33 (s, 1H), 8.75 (d, 1H), 9.91 (s, 1H); MS (ESI): 246 [M+H]+.
Figure imgf000038_0001
g. 3-(2,6^ichloro-3i5-dimethoxy-phenyl) -methyl^l-[6-(2-nitro^
urea
To a solution of N-methyl-N'-tl-nitro-pheny^-pyrimidine^^-diamine (150 mg, 0.61 mmol) in THF (15 mL) was added NaH (60%, 60 mg, 1.5 mmol) at 0 °C, and the mixture was stirred for 30 minutes at room temperature. A solution of 2,4-dichloro-3-isocyanato-l,5-dimethoxy- benzene (180 mg, 0.73 mmol) was added dropwise at room temperature. The resulting mixture was stirred for 2 hours. Water (2 mL) was added to quench the reaction. The mixture was concentrated, and the residue was purified by flash chromatography on silica eluting with DCM:EtOAc == 6:1-1:1 to obtain the title compound (54 mg, yield: 18%) as a yellow solid. lH NMR (400 MHz, DMSO-d6) δ 3.38 (s, 3H), 3.93 (s, 6H), 6.75 (s, 1H), 6.91 (s, 1H), 7.34 (t} 1H), 7.72 (t, 1H), 7.79 (d, 1H), 8.01 (d, 1H), 8.38 (s, 1H)5 9.99 (s, 1H), 11.78 (s, 1H); MS (ESI): 493 [M+H]+.
Figure imgf000038_0002
h. l-[6-(2-Amino-phenylamino)-pyrimidin~4~yl]-3-(2,6-dwhlo^
methyl-urea
To a solution of 3-(2,6-dichloro-3,5-dimethoxy-phenyl)-l-methyl-l-[6-(2-nitro-phenylamino)- pyrimidin-4-yl]-urea (50 mg, 0.1 mmol) in THF (10 mL) and MeOH (10 mL) was added Raney- Ni (suspension in water) at room temperature, the resulting mixture was stirred for 2 hours under hydrogen atmosphere. The reaction was filtered and concentrated to obtain the title compound (38 mg, yield: 82%), which was used directly in the next step. 1H NMR (400 MHz, CDClj) δ 3.28 (s, 3H), 3.85 (s, 2H), 3.94 (s, 6H), 5,86 (s, 1H), 6.52 (s, 1H), 6.78-6.87 (m, 3H), 7.16-7.20 (m, 2H), 8.39 (s, 1H), 12.62 (s, 1H); MS (ESI): 463 [M+H]+.
Figure imgf000039_0001
i. N-(2-{6-[3-(2,6^ichloro-3,5-dimethoxy-phenyl)-l-methyl~ureido]-^^
phenyl)- crylamide
To a solution of l-[6-(2-amino- henylamino)- yrimidin-4-yl]-3-(2,6-dic loro-3,5-dimethoxy- henyl)-l-methyi-uΓea (25 mg, 0.05 mmol) in THF (10 mL) was added a solution of acryloyl chloride in THF (20 mg/mL, 0.5 mL, 0.1 mmol) at -10 °C, and the resulting mixture was stirred for 1 hour at this temperature. MeOH (1 mL) was added to quench the reaction. The mixture was concentrated and the residue was purified by prep-TLC to obtain the title compound (12 mg, yield: 43%). 1H NMR (400 MHz, DMSO-d6) δ 3.26 (s, 3H), 3.94 (s, 6H), 5.74 (d, 1H), 6.24 (d, 1H), 6.37 (s, 1H), 6.47-6.54 (m, 1H), 6.90 (s, 2H), 7.20 (d, 2H), 7.56-7.58 (m, 1H), 7.66-7.68 (m, 1H), 8.38 (s, 1H), 9.99 (s, 1H), 9.70 (s, 1H), 11.99 (s, 1H); MS (ESI): 517 [M+H .
Compounds 102, 103 and 105 were synthesized in a similar manner as compound 100.
Procedure 2B: Example - 107
Figure imgf000040_0001
N-(2-{6-[3-(2,6-dichloro-3,5-dimethoxy-phenyl)-l-pyridin-3-ylmethyi-ureido]-pyrimidin-4- ylamino}-phenyl)-acrylamide
Figure imgf000040_0002
rt, overnight
a. (6-Chloro-pyrimidin-4-yl)^yridin-3~ylmethyl-amine
To a solution of 4,6-dichloro-pyrimidine (1 g, 6.71 mmol) in dioxane (20 mL) was added a solution of pyridin-3-yl-methylamine (745 mg, 6.9 mmol) at room temperature. The resultmg mixture was stirred at room temperature overnight. The mixture was concentrated and the residue was purified by flash chromatography on silica to obtain the title compound (680 mg, yield: 46%). MS (ESI): 221 [M+H]+.
Figure imgf000041_0001
b. N~(2~Nitro-phenyl)-N'^yridin-3-ylmethy yrimidine-4,6~diam
A degassed mixture of (6-chloro-pyrimidin-4-yl)-pyridin-3-ylmethyl-amine (300 mg, 1 ,36 mmol), 2-nitro-phenylamine (188 mg, 1.36 mmol), Pd2(dba)3 (128 mg, 0.14 mmol), Xantphos
(161 mg, 0.28 mmol) and Cs2C03 (913 rag, 2,8 mmol) in toluene (10 mL) was heated at 100 °C for 4 hours. The reaction was concentrated, and the residue was purified by flash
chromatography on silica to obtain the title compound (150 mg, yield: 34%). MS (ESI): 323
[M+Hf.
Figure imgf000041_0002
c. 3'(2,6^ichtoro-3,5-dlmethoxy^henyl)-l-[6-(2-nitro~phenylamino)-pyrM
pyridin-3-ylmethyl-urea
To a solution of N-(2-nitro-phenyl)-N'-pyridin-3-ylmethyl-pyrimidine-4;6-diamine (150 mg, 0.467 mmol) in THF (15 mL) was added NaH (60%, 48 mg, 1.2 mmol) at 0 °C, and the mixture was stirred for 30 minutes at room temperature. A solution of 2,4-dichloro-3-isocyanato-l,5- dimethoxy-benzene (procedure 2A, steps a-d; 180 mg, 0.73 mmol) was added dropwise at room temperature. The resulting mixture was stirred for 2 hours. Water (2 mL) was added to quench the reaction. The mixture was concentrated, and the residue was purified by flash
chromatography on silica to obtain the title compound (85 mg, yield: 32%). MS (ESI): 570
Figure imgf000042_0001
d. l-{6-(2-Amino-phenylamino)-pyrimidin-4-yl]^^
pyridin-3-ylmethyl-urea
A mixture of 3-(2,6-dichloro-3,5-dimethoxy-phenyl)-l-[6-(2-nitro-phenylamino)- pyrimidin-4- yl]-l-pyridin-3-ylmethyl-urea (85 mg, 0.149 mmol) and Fe (84 mg, 1.5 mmol) in AcOH (5 mL) was heated at 50°C for 2 hours. The reaction mixture was filtered and the filtrate was concentrated in vacuo to give crude product, which was purified by silica gel column
chromatography to afford the title compound (53 mg, yield: 66%). MS (ESI): 540 [M+H]+.
Figure imgf000042_0002
e. N-(2-{6-[3-(2t6~Dichloro-3t5-dimethoxy^henyl) -pyridin-3-ylm thy^
ylaminoj-ph enyl)-acrylamide
To a solution of l-[6-(2-amino-phenylamino)-pyrimidin-4-yl]-3-(2,6-dichloro-355-dimethoxy- phenyl)-l-pyridin-3-ylmethyl-urea (53 mg, 0.1 mmol) in THF (10 mL) was added a solution of acryloyl chloride in THF (20 mg mL, 0.5 mL, 0.1 mmol) at -10 °C, and the mixture was stirred for 1 hour at this temperature. MeOH (1 mL) was added to quench the reaction. The mixture was concentrated and the residue was purified by prep-TLC to obtain the title compound (9 mg, yield: 15%). 1H NMR (400 MHz, CDC13) δ 3.84 (s, 6H), 5.01 (s, 2H), 5.69 (d, 1H), 5.75 (s, IH), 6.10 (dd, 1H), 6.34 (d, 1H), 6.47 (s, 1H), 7.00 (d, IH), 7.09-7.24 (m, 2H), 7.28 (t, IH), 7.32 (s, IH), 7.47 (d, IH), 7.69-7.71 (m, 2H), 8.31-8.34 (m, 2H), 8.40-8.42 (m, IH), 12.60 (s, H); MS (ESI): 594 [M+H]+. Procedure 2C: Example - 108
Figure imgf000043_0001
Figure imgf000043_0002
yield: 29%
Figure imgf000043_0003
N-[2-{6-[3-(2,6-dichloro-3,5-dimethoxy-phenyI)-l~methyl-ureido]-pyrimidin-4-ylam (4-ethyl-piperazin-l-y])-phenyl]-acryIamidemethaBe
Figure imgf000043_0004
«. tert-Butyl 4-bromo-2-nitrophenylc rbamate
A mixture of 4-bromo-2-nitroaniline (4 g, 18.4 mmol), (Boc)20 (4.4 g, 20.24 mmol) in THF (50 mL) was heated under reflux overnight. The mixture was concentrated and the residue was purified by flash chromatography on silica eluting with PE:EtOAc = 20:1 to obtain the title compound (5.4 g, yield: 93%). MS (ESI): 317, 319 [M+Hf .
Figure imgf000044_0001
100 °C, 4h
b. tert-Butyl 4-(4-ethylpiperazin-l-yl)-2~nitrophenylcarbamate
A degassed mixture of tert-butyl 4-brorno-2-nitrophenylcarbarnate (5.4 g, 17 mmol), 1- ethylpiperazine (2.91 g, 25.5 mmol), Pd2(dba)3 (2.1 g, 3.4 mmol), xantphos (3.92 g, 6.8 mmol) and CS2CO3 (11.1 g, 34 mmol) in toluene (85 mL) was heated at 100 °C for 4 hours. The reaction was concentrated, and the residue was purified by flash chromatography on silica eluting with MeOH: DCM = 1 :50-1:20 to obtain the title compound (3.3 g, yield: 55%). MS (ESI): 351 [ +HJ+.
Figure imgf000044_0002
c. 4-(4~Ethylpiperazin-l-yl)-2-nitroaniline
To a solution of tert-butyl 4-(4-ethylpiperazin-l-yl)-2-nitrophenylcarbamate (3.3 g, 9.43 mmol) in DCM (50 mL) was added TFA (20 mL) at 0 °C, the resulting mixture was stirred for 3 hours at r After removal of all volatiles in vacuo, the residue was re-dissolved in DCM, neutralized with saturated aqueous K2C03 and extracted with DCM. The combined extracts were concentrated to obtain the title compound (2.1 g, yield: 90%), which was used directly in the next step. JH NMR (400 MHz, DMSO-d6) δ 1.02 (t, 3H), 2.36 (q, 2H), 2.47-2.49 (m, 4H) 2.97- 3.00 (m, 4H), 6.97 (d, 1H), 7.20 (s, 2H), 7.25 (s, 1H), 7.34 (dd, 1H); MS (ESI): 251 [M+H]+.
Figure imgf000044_0003
(L rf- - -ethylpiperazin-l-yfy- -nitroph enyl)-T^-methylpyrimidine-4, 6-diamine
A degassed mixture of 4-(4-ethylpiperazin-l-yl)-2-nitroaniline (2.1 g, 8.4 mmol), 6-chloro-jV- methylpyrimidin-4-amine (Procedure 2A, step e; 1.2 g, 8.4 mmol), Pd2(dba)3 (1.54 g, 1.68 mmol), xantphos (1.94 g, 3.36 mmol) and Cs2C0 (5.48 g, 16.8 mmol) in toluene (45 mL) was heated at 100 °C for 1 hour. The reaction was concentrated, and the residue was purified by flash chromatography on silica eluting with MeOH: DCM = 1 :40~1 :20 to obtain the title compound (870 mg, yield: 29%). MS (ESI): 358 [M+H]+.
Figure imgf000045_0001
e. 3-(2f6-Dichloro~3,5-dimethoxyphenyl)-l-(6~(4-(4-ethylpiperazin-l~yl^
nitrophenylamino)pyrimidin-4-yl)~l-methyturea
To a solution of N4-(4-(4-ethylpiperazin-l-yl)-2-nitrophenyl)-N6-methylpyrirnidme-4,6- diamine (870 mg, 2.44 mmol) in THF (15 mL) was added NaH (60%, 200 mg, 5 mmol) at 0 °C, and the mixture was stirred for 30 minutes at room temperature. A solution of 2,4-dichloro-3- isocyanato- 1,5-dimethoxy-benzene (Procedure 2A, steps a-d; 908 mg, 3.66 mmol) in THF was added dropwise at 0 °C. The resulting mixture was stirred at room temperature for 2 hours. Saturated aqueous NH4C1 solution (2 mL) was added to quench the reaction. The mixture was concentrated and extracted with DCM. The combined extracts were washed with brine, dried over anhydrous Na2S04, and concentrated to give the crude product, which was purified by flash chromatography on silica to obtain the title compound (330 mg, yield: 21%) as a red oil. 1H NMR (400 MHz, CDC13) 5 1.44 (t, 3H), 3.01 (t, 2H), 3.21 (q, 2H), 3.41-3.49 (m, 5H), 3.73-3.80 (ms 4H), 3.92 (s, 6H), 6.27 (s, IH), 6.55 (s, IH), 7.25 (d, IH), 7.69 (s, IH), 8.32 (d, IH), 8.52 (s, IH), 10.28 (br s, IH), 12.05 (br s, IH); MS (ESI): 605 [M+H]+.
Figure imgf000045_0002
l-(6~(2-Amino-4-(4-ethylpiperazin-l-yl)phenylamino)pyrimid^
dimethoxyphenyl)-l-methylurea
To a solution of 3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-(6-(4-(4-ethylpiperazin-l-yl)-2- nitrophenylaraino)pyrimidin-4-yl)-l-methylurea (330 mg, 0.546 mmol) in THF (20 mL) and MeOH (20 mL) was added Raney-Ni (suspension in water) at room temperature, the resulting mixture was stirred for 3 hours under hydrogen atmosphere (1 atm). The reaction was filtered and concentrated. The residue was washed twice with MeOH to obtain the title compound (280 mg, purity: 90%), which was used directly in the next step. MS (ESI): 575 [M+H]+.
Figure imgf000046_0001
g, N~(2-(6~(3~(2,6^ichloro-3,5-dimethoxyphenyl)~l~methylureido)pyrM
ethylpiperazin-l-yl)phenyl)acrylamide
To a solution of l-(6-(2-amino-4-(4-ethylpiperazin-l-yl)phenylamino)pyrimidin-4-yl)-3-(2,6- dichloro-3,5-dimethoxyphenyl)-l-methylurea (280 mg, purity: 90%, 0.44 mmol) in THF (30 mL) was added a solution of acryloyl chloride in THF (20 mg/mL, 2 mL, 0.44 mmol) at -10 °C, and the resulting mixture was stirred for 1 hour at this temperature. MeOH (1 mL) was added to quench the reaction. The mixture was concentrated and the residue was purified by prep-HPLC and prep-TLC to obtain the title compound (20 mg, yield: 7%). 1H NMR (400 MHz, CDC13) δ 1.31 (t, 3H), 2.65 (q, 2H), 2.62-2.68 (m, 4H), 3.27 (s, 3H), 3.36-3.38 (m, 4H), 3.91 (s, 6H), 5.76 (d, 1H), 5.90 (s5 1H), 6.24 (dd, 1H), 6.41 (d, 1H), 6.52 (s, 1H), 6.74 (dd, 1H), 7.07 (br s, 1H), 7.23 (d, 1H), 7.72 (br s, 1H), 7.98 (br s, 1H), 8.37 (s, 1H), 12.52 (s, 1H); MS (ESI): 629 [M+H]+.
Example - 110
Figure imgf000047_0001
N-(2-(6-(3 -(296-dich lo ro-3,5-dimeth oxy pheny -methy lureido)py rimid in-4-y lamin o)- phenyI)-2-fluoroacrylamide
The compound was synthesized following the approach outlined in Procedure 2 A (Example 100), modifying step (i) to the following procedure: To a solution of I-[6-(2-amino- phenylamino)-pyrimidin- 4-yl]-3-(2,6-dichloro-3,5-dimethoxy-phenyl)-l-methyl-urea (130 mg, mixed with tetrachloro aniline) and DCC (118 mg, 0.56 mmol) in chloroform (100 mL) was added a solution of 2-fluoroacrylic acid (50 mg, 0.56 mmol) in chloroform (50 mL) at 0 °C, and the resulting mixture was stirred at room temperature overnight. Water (1 mL) was added to quench the reaction. The mixture was concentrated and the residue was purified by reverse phase column and prep-TLC to obtain the title compound (4 mg, yield: 5%). !H NMR (400 MHz,
CDC13) δ 12.17 (s, 1H), 8.45 (s, 1H), 8.35 (s, 1H), 7.84 (d, 1H), 7.37 (d, 1H), 7.29 (t, 1H), 7.26 (t, 1H), 6.47 (s, 1H), 5.94 (s, 1H), 5.78 (dd, 1H), 5,21 (dd, 1H), 3.85 (s, 6H), 3.25 (s, 3H); MS (ESI): 535 [M+H]+.
Procedure 2E: Example
Figure imgf000048_0001
yield: 10%
N 2-(6-(3-(2,6-dichIoro-3,5-dimethoxyphenyI)-l-methylureido)pyrimidin-4-ylamino)-5-(l- ethylpiperidin-4-yl)phenyl)acryIamide
Figure imgf000049_0001
dioxane/H20, reflux, 3h
a. tert-butyl 4~(4-amino-3-nitrophenyl)-5, 6-dihydropyridine-l (2H)-carboxylate
To a degassed mixture of 4-bromo-2-nitroaniline (1 g, 4.6 mmol), tert-butyl 4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)-5,6-dihydropyridine-l(2H)-carboxylate (1.42 g, 4.6 mmol), tripotassium phosphate trihydrate (3.9 g, 14.64 mmol) in dioxane and water (30 mL, 8:1) was added Pd(dppf)2Cl2 (337 mg, 0.46 mmol). The mixture was refluxed at 110 °C for 3 hours.
Filtration and concentration gave crude product, which was purified by silica gel column chromatography to afford the title compound (1,1 g, yield: 75%). MS (ESI): 320 [M+H]+.
Figure imgf000049_0002
b. l-(6-chloropyrimidin^-yl)-3-(2,6-dfchloro-3,5-dimethoxyphen^
(trimeth Is ilyfyethoxy) methyl) urea
To a solution of 6-chloro-N-methylpyrimidin-4-amine (Procedure 2A, step e; 460 mg, 3.21 mmol) in DMF (15 mL) was added NaH (60%, 193 mg, 4.81 mmol) at 0 °C, and the mixture was stirred for 30 minutes at room temperature. A solution of 2,4-dichloro-3-isocyanato- 1,5- dimethoxy-benzene (Procedure H, steps a-d; 1.03 g, 4.17 mmol) in DMF (5 mL) was added dropwise at room temperature. The resulting mixture was stirred for 0.5 hour. SEMC1 (804 mg, 4.81 mmol) in DMF (2 mL) was added. The reaction mixture was stirred at room temperature for 1 hour. Saturated aqueous NH4CI was added to quench the reaction. The mixture was diluted with water and extracted with EtOAc. The combined extracts were washed with water and brine, dried over anhydrous Na S04 and filtered. The filtrate was evaporated under vacuum to give crude product, which was purified by flash chromatography on silica to obtain the title compound (470 mg, yield: 28%). MS (ESI): 521 [M+Hf.
Figure imgf000050_0001
c. tert-b utyl-(4~(6-(3-(2, 6-dichloro-3, 5-dimeth oxyph enyl)-l-methyl-3-( (2-
(trimethylsilyl)ethoxy)methyl)ureido)pyrimid^
dihydropyridine-l(2H)~carboxylate
A degassed mixture of l-(6-chloropyrimidin-4-yl)-3-(2;,6-dichloro-3,5-dimethoxyphenyl)-l- methyl-3-((2-(trimethylsilyl)ethoxy)methyl)urea (470 mg, 0.9 mmol), tert-butyl 4-(4-amino-3- nitrophenyl)-5,6-dihydropyridine-l(2H)-carboxylate (320 mg, 1 mmol), Pd2(dba)3 (92 mg, 0.1 mmol), xantphos (115 mg, 0.2 mmol) and Cs2C(¾ (652 mg, 2 mmol) in toluene (10 mL) was heated at 100 °C for 5 hours. The reaction was concentrated, and the residue was purified by flash chromatography on silica to obtain the title compound (400 mg, yield: 57%). MS (ESI): 804 [M+H]+.
Figure imgf000050_0002
d, tert-butyl-4^-amino^-(6-(3~(2,6-dichloro-3,5-dimethoxyphen^ -((2~
(trimethylsilyfyethoxy) methyl) ureido)pyrimidin-4~ylamino)phenyl)piperidine~l-carboxylate
To a solution of tert-butyl-4-(4-(6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl-3-((2- (trimethyl silyl)ethoxy)methyl)ureido)pyrimidin-4-ylamino)-3 -nitrophenyl) -5 , 6-dihy dropyridine- l(2H)-carboxylate (380 mg, 0.473 mmol) in MeOH (10 mL) was added Pt02 (38 mg, 10% wt) and one drop of chlorobenzene at room temperature, the resulting mixture was stirred under hydrogen atmosphere (1 atm) overnight. The reaction was filtered and concentrated. The residue was purified by flash chromatography on silica to obtain the title compound (130 mg, yield: 37%). MS (ESI): 776 [M+H ,
Figure imgf000051_0001
e. N-(2-( 6-(3-(2, 6-dichloro-3, S-dimeth oxyphenyl)-l~methylureido)pyrimidin-4-ylamin o)-5- (piperidin-4-yl)phenyl)acrylamide TFA salt
To a solution of tert-butyl-4-(3-amino-4-(6-(3-(2,6-dichloro-3,5-dirnethoxyphenyl)-l-methyl-3- ((2- (trimemylsilyI)ethoxy)methyl)u^ 1 -carboxylate
(130 mg, 0.168 mmol) in THF (15 mL) was added a solution of acryloyl chloride (10 mg/niL, 1.7 mL, 0.19 mmol) dropwise at -10 °C, and the resulting mixture was stirred at 0 °C for 1 hour. LC-MS showed that the reaction was complete. MeOH (5 mL) was added to quench the reaction, and the reaction was concentrated. The residue in DCM (2 mL) was added dropwise to a mixture of DCM/TFA (2/1, v/v, 3 mL). The mixture was stirred at room temperature for 1 hour and then concentrated under vacuum. The residue (50 mg, quant.) was used directly for the next step without further purification. MS (ESI): 600 [M+H .
Figure imgf000051_0002
N-(2-(6-(3-(2y6-dichloro~3,5'dimethoxyphenyl)-l-methylureido)pyrimM (1- ethylpiperidin-4~yl)phenyl)acryl mide
To a solution of N-(2-(6-(3-(2,6-dichloro-3J5-dimethoxyphenyl)-l-methylureido)pyrimidin-4- ylamino)- 5-(piperidin-4-yl)phenyl)acrylamide TFA salt (35 mg, 0.049 mmol) in EtOH (1 mL), was added NaOAc (4 mg, 0.05 mmol) and aqueous acetaldehyde (1 mL, 0.9 mmol, 40%). After the mixture was stirred at room temperature for 1 hour, NaB¾CN (12 mg, 0.18 mmol) was added, and the solution was stined at room temperature for another 3 hours. After removal of all volatiles in vacuo, the residue was partitioned between DCM and water. The aqueous layer was extracted with chloroform twice. The combined extracts were washed with brine, dried over anhydrous Na2S0 and filtered. The filtrate was evaporated under vacuum to give crude product, which was purified by prep-HPLC to afford the title compound (3 mg, yield: 10%). !H NM (400 MHz, MeOH-d4) δ 8.38 (s, IH), 7.68 (s, IH), 7.54 (d, IH), 7.24 (dd, IH), 6.83 (s, IH), 6.46-6.35 (m, 3H), 5.81 (d, IH), 3.97 (s, 6H), 3.74-3.70 (m, 2H), 3.37 (s, 3H), 3.26 (q, 2H), 3.17- 3.1 1 (m, 2H), 2.99-2.96 (m, IH), 2.26-2.22 (m, 2H), 2.06-1.99 (m, 2H), 1.43 (t, 3H); MS (ESI): 628 [M+H]+.
Example - 112
Figure imgf000052_0001
N-(2-(6~(3-(2,6-dichIoro-3,5-dimethoxyphenyI)-l-methylureido)pyrimidin-4-ylamino)-5-(l- raethylpiperidin-4-yl)phenyl)acrylamide
The compound was synthesized following the approach outlined in Procedure 2E (Example 11 1), substituting formaldehyde in step (f) to afford the title compound (1.5 mg, yield: 11.6%). !H NMR (400 MHz, MeOH-d4) δ 8.26 (s, IH), 7.55 (s, IH), 7.41 (d, IH), 7.11 (d, IH), 6.71 (s, IH), 6.34-6.22 (m, 3H), 5.68 (d, IH), 3.84 (s, 6H), 3.55-3.52 (m, 2H), 3.25 (s, 3H), 3.21-3.08 (m, 2H), 2.90-2.83 (m, 4H), 2.1 1-2.08 (m, 2H), 1.89-1.85 (m, 2H); MS (ESI): 614 [M+H]\
Procedure 2F: Example -
Figure imgf000053_0001
yield: 53% yield: 53%
Figure imgf000053_0002
yield: 30%
Figure imgf000053_0003
N-(2-{6-[3-(2,6-dichioro-3,5-dimethoxy-phe^
dimethyJaminomethyl-phenyI)-acrylaniide
Figure imgf000053_0004
a. 4-Fluoro-3-nitro-benzaldehyde
To a stirred solution of (4-fluoro-3-nitro-phenyl)-methanol (750 mg, 4.4 mmol) in DCM (40 mL) at 0 °C was added Dess-Martin Reagent (3.0 g, 7 mmol). The solution was stirred at room temperature for 4 hours. TLC showed disappearance of starting material. The reaction was quenched with 10% NaHC03 and 10% Na2S203 aqueous solution and DCM layer separated and washed with water (100 mL) and brine (50 mL). The reaction was concentrated, and the residue was purified by flash chromatography on silica to obtain the title compound (570 mg, yield: 75%). 1H-NMR (400 MHz, DMSO-d6) δ 10.09 (d, 1H), 8.36 (t, 1H), 8.06 (dd, 1H), 7.97 (m, 1H).
Figure imgf000054_0001
b. 4-Atnino-3-nitrobenzaldehyde
To a solution of 4-fluoro-3-nitro-benzaldehyde (570 mg, 3.3 mmol) in THF (20 mL) was added NH4OH (5 mL). The reaction mixture was stirred at room temperature for 1 hour. The resulting yellow solid was collected and washed with water, dried under vacuum to give the title compound (300 mg, yield: 53%). 1H-NMR (300 MHz, DMSO-d6) δ 9.76 (s, 1H), 8.57 (d, 1H), 8.18 (br s, 2H), 7.80 (dd, 1H), 7.10 (d, lH).
Figure imgf000054_0002
c. 4-Dimethyl minomethyl-2-nitro-phenyl mine
To a stirred solution of dimethylamine (4.0 mL, 2 M, 8.0 mmol) in MeOH ( 4 mL) was added Ti(0'Pr)4 (1.15 g, 4 mmol) and the solution was stirred at room temperature for 15 minutes Then 4-amino-3-nitro-benzaldehyde (160 mg, 1.0 mmol) in MeOH (2 mL) was added and the solution was stirred at room temperature overnight. Then NaBH4 (78 mg, 2 mmol) was added and the solution was stirred at room temperature for 1 hour. LCMS showed major product peak. The solution was diluted with EtOAc (60 mL) and washed with water (2 xlOO mL) and brine (50 mL), dried over anhydrous Na2S04, The solution was evaporated to dryness and 130 mg of crude product was collected, which was used for the next step without further purification, Ή NMR (400 MHz, DMSO-d6) δ 7.82 (s, 1H), 7.35 (br s, 2H), 7.31 (dd, 1H), 6.97 (d, 1H), 3.26 (s, 2H), 2.12 (s, 6H).
Figure imgf000054_0003
d. l-(2,6-Dichloro-3,5-dimethoxy^henyl)-3-[6-(4^imethylaminome phenylamino)~ pyrimidin-4-ylj-3-methyl~l-(2-MmethyMlany ethoxymethyl)-urea
To a stirred solution of l-(6-chloro-pyrimidin-4-yl)-3-(2,6-dichloro-3,5-dimethoxy- phenyl)- 1- methyl-3- (2-trimethyisilanyl-ethoxymethyl)-urea (procedure L, step b; 260 mg, 0.5 mmol) in toluene (5 mL) was added 4-dimethylammomethyl-2-nitro-phenylamine (100 mg, 0.5 mmol), Cs2C03 (400 mg, 1.25 mmol), Pd2(dba)3 (46 mg, 0.05 mmol), xantphos (90 mg, 0.15 mmol). The solution was stirred at 100 °C overnight. LCMS showed major product peak. The solution was evaporated with silica gel and purified by flash chromatography on silica eluting with EtOAc (w(/ 0.5% TEA):MeOH (w/0.5% TEA) = 10-10:0.5 to afford the desired product (100 mg, yield: 30%).
Figure imgf000055_0001
Figure imgf000055_0002
e. l6-(2~Amino-4-dimethylamiwmethyl~phenylamino ^
dimethoxy^henyl)~l~methyl-3-(2-trimethylsil nyl^ethoxymethyl)~ure
To a stirred solution of l-(2,6-dichloro-3,5-dimethoxy-phenyl)-3-[6-(4-dimethyIaminomethyl-2- nitro- phenylamino)-pyrimidin-4-yl]-3-methyl-l-(2-trimethylsilanyl-ethoxymethyl)-urea (100 mg, 0.15 mmol) in MeOH (10 mL) was added 4 drops of chlorobenzene and then Pt02 (30 mg, 30% wt). The solution was stirred under hydrogen atmosphere at room temperature overnight. The reaction was filtered and concentrated. The residue was taken to the next step without further purification. MS (ESI): 650 [M+H]+.
Figure imgf000055_0003
l-[6-(2-A mino-4-dimethylaminomethyl-ph enylamino)-pyrimidin~4~yl]-3-(2, 6- dich loro-3, 5~ dimeth oxy~phenyl)-l-methyl-urea
To a stirred solution of l-[6-(2-amino-4-dimethylaminomethyl-phenylamino)-pyrimidin-4-yl]-3- (2,6-dichloro-3 ,5 -dimethoxy-phenyl)- 1 -methyl-3 -(2 rimethylsilanyl-ethoxymethyl)-urea in anhydrous DCM (10 mL) was added TFA (10 mL) at room temperature, The solution was stirred at room temperature for 3 hours. LCMS showed major product peak. The solution was evaporated to dryness, diluted with DCM (40 mL) and washed with 10% saturated Na2C03 (10 mL). The DCM layer was dried over anhydrous Na2S04. Concentration under vacuum gave crude product, which was purified by silica gel column chromatography (10% MeOH/DCM with 0.5% Et3N) to afford the title compound (45 mg, yield: 58% in two steps). MS (ESI): 520 [M+H]+.
Figure imgf000056_0001
g. N-(2-{6~[3~(2,6^ichloro-3,5-dimethoxy-phenyl)-I~methyl~ureido
dim thylaminomethyl~phenyl)~acrylamide
To a stirred solution of l-[6-(2-amirio-4-dimethylaminomethyl-phenylammo)-pyrimidin-4-yl}-3- (2,6-dichloro-3,5-dimethoxy-phenyl)-l -methyl-urea (45 mg, 0.11 mmol) in THF (40 mL) at -10 °C was added acryloyl chloride (30 mg, 0,33 mmol) in THF (3 mL). The solution was stirred at -10 °C for 5 hours. LCMS showed major product peak. The reaction was quenched with MeOH (3 mL) and evaporated. The residue was purified by prep-HPLC (water/ ACN in NH4HC03 condition) to afford the title compound (6 mg, yield: 15%). 1H-NMR (400 MHz, MeOH-d^ δ 8.25 (s, 1H), 7.47 (d, 1H), 7.44 (s, 1H), 7.16 (dd, 1H), 6.70 (s, 1H), 6.33-6.20 (ra, 3H), 5.67 (dd, 1H), 3.84 (s, 6H), 3.43 (s, 2H) 3.22 (s, 3H), 2.20 (s, 6H); MS (ESI): 574 [M+Hf .
Example - 114
Figure imgf000056_0002
N-(2-(6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-raethylureido)pyrimidiii-4- ylam o)phenyl)propiolamide
The compound was synthesized following the approach outlined in Procedure 2C (Example 108) modifying step (g) to the following procedure: To a solution of l-[6-(2-amino-phenylamino)- pyrimidin- 4-yl]-3-(2,6-dichloro-3J5-dimethoxy-phenyl)- 1 -methyl-urea (50 mg, 0.108 mmol) and DCC (46 mg, 0.22 mmol) in chloroform (50 mL) was added a solution of propiolic acid (16 mg, 0.22 mmol) in chloroform (50 mL) at 0 °C, and the resulting mixture was stirred at room temperature overnight. Water (1 mL) was added to quench the reaction, The mixture was concentrated and the residue was purified by reverse phase column and prep-TLC to obtain the title compound (5 mg, yield: 9.1%). 1H NMR (400 MHz, DMSO-d6) 6 12.04 (s, IH), 10.36 (s, IH), 9.03 (s, IH), 8.45 (s, IH), 7.67 (d, IH), 7.59 (d, IH), 7.30-7.25 (m, 2H), 6.96 (s, IH), 6.50 (s, IH), 4.42 (s, IH), 4.00 (s, 6H), 3.35 (s, 3H); MS (ESI): 515 [M+H]+
Example - 116
Figure imgf000057_0001
N-(2-{6-[3-(2,6-Dichloro-3,5~dimethoxy-phenyl)-l-(2-met^^
ylaminoj-ph enyl)-acrylamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 1 -(6-chloro-pyrimidin-4-yl)-3-(2,6-dichloro-3,5-dimethoxy-phenyl)- 1 -(2- methoxy-ethyl)-3-(2-trimethylsilanyl-ethoxymethyl)-urea (preparation shown below) in step (d) to afford the title compound (40 mg, yield: 16% over five steps). lH NMR (400 MHz, DMSO- d6) δ 11.28 (s, IH), 9.71 (s, IH), 8.85 (s, IH), 8.38 (s, IH), 7.70-7.68 (m, IH), 7.55-7.53 (m, IH), 7.20-7.18 (m, 2H), 6.89 (s, IH), 6.69 (s, IH), 6.50 (dd, IH), 6.26 (d, IH), 5.75 (d, IH), 4.03 (t, 2H)5 3.94 (s, 6H), 3.56 (t, 2H), 3.24 (s, 3H); MS (ESI): 437 [M+H]+.
Preparation of l-(6-Chloro-pyriraidin-4-yl)-3-(2,6-dichloro-3,5-dimethoxy-phenyl)-l-(2- methoxy-ethyl)-3-(2-trimethylsilanyI-ethoxymethyl)-urea:
Figure imgf000058_0001
a. ( 6-Chloro-pyrimidin-4-yl)~(2~meth oxy-ethyl)-amine
To a solution of 4,6-dichloro-pyrimidine (2 g, 14 mmol) in iPrOH (70 mL) and DIPEA (1.94 g, 15 mmol) was added a solution of 2-methoxy-ethylamine (1.13 g, 15 mmol) at room
temperature. The resulting mixture was stirred at room temperature for 1 hour. Water was added and the mixture was extracted with DCM. The combined extracts were washed with brine, dried over anhydrous Na2S04, and concentrated to give the crude product, which was purified by flash chromatography on si!ica to obtain the title compound (1.95 g, yield: 82%). MS (ESI): 188 [M+H]+.
Figure imgf000058_0002
b. l-(6-Chloro-pyrimidin-4-yt)-3-(2f6~dichloro-3,5-dimethoxy^hen^
(2-trimethylsilanyl-ethoxymethyl)-urea
To a solution of (6-chloro-pyrimidin-4-yl)-(2-methoxy-ethyl)-amme (300 mg, 1.6 mmol) in DMF (10 mL) was added NaH (60%, 96 mg, 2.4 mmol) at 0 °C, and the mixture was stirred for 10 minutes at room temperature. A solution of l-isocyanato-3,5-dimethoxy-benzene (590 mg, 2.4 mmol) in DMF (5 mL) was added dropwise at 0 °C. The resulting mixture was stirred for 30 minutes. SEMC1 (400 mg, 2.4 mmol) in DMF (2 mL) was added and the reaction mixture was stirred at room temperature for 1 hour. Saturated aqueous NH4C1 was added to quench the reaction. The mixture was diluted with water and extracted with EtOAc. The combined extracts were washed with water and brine, dried over anhydrous Na2SC>4, and concentrated to afford the crude product, which was purified by flash chromatography on silica to obtain the title compound (720 mg, yield: 78%). MS (ESI): 565 [M+Hf. Example - 120
Figure imgf000059_0001
N-(2-{6-[3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-l-(3-dimethylamino-propyl)-ureidoJ- pyrimidin-4-ylamino}~phenyl)-aciylamide trifl oroacetic acid
The compound was synthesized following the approach outlined in Procedure 21 (Example 142), substituting 2-nitroaniline and N-(3-Dimethylamino-propyl)-N'-(2-nitro-phenyl)-pyrimidine-4,6- diamine (prepared by the method outlined below) in step (c) to afford the title compound (11 mg, yield: 7.5% over six steps). 1H NMR (300 MHz, DMSO-d6) δ 11.30 (s, IH), 9.80 (s, IH), 9.28 (m, IH), 8.92 (s, IH), 8.42 (s, IH), 7.64 (d, IH), 7.55 (d, IH), 7.24-7.16 (m, 2H), 6.90 (s, IH), 6.53 (s, IH), 6.49 (dd, IH), 6.25 (d, IH), 5.76 (d, IH), 3.95 (s, 6H), 3.91-3.88 (m, 2H), 3.11-3.05 (m, 2H), 2.74 (d, 6H), 1.97-1.92 (m, 2H); MS (ESI): 588 [M+H]+.
Preparation of N-iS-Dimethylaraino-propy -N'-ii-nitro-phe -pyrimidme^^-diamine
Figure imgf000059_0002
a. N'^-C loro-pyrinddin-^yl^N^-dimethyl^ropane-l^-diamine
To a solution of 4,6-dichloro-pyrimidine (1 g, 6.7 mmol) and DIPEA (1.03 g; 8 mmol) in iPrOH (20 mL) was added N,N-dimethyl-propane-l53-diarnine (714 mg, 7 mmol) at room temperature. The resulting mixture was stirred for 2 hours. Water was added and the mixture was extracted with DCM. The combined extracts were washed with brine, dried over anhydrous Na2S04j and concentrated to give the crude product, which was purified by flash chromatography on silica to obtain the title compound (1.15 g, yield: 80%). MS (ESI): 215 [M+H]+.
Figure imgf000060_0001
toluene, 100°C, 4h
b. N-(3^imethylamino-propyl)-N'-(2-nitro-phenyl)-pyrim^
A degassed mixture of N,-(6-chloro-pyrimidin-4-yl)-N,N-dimethyl-propane-l,3-diamine (800 mg, 3.74 mmol), nitro aniline (525 mg, 3.8 mmol), Pd2(dba)3 (348 mg, 038 mmol), Xantphos (438 mg, 0.76 mmol) and Cs2C03 (3.05 g, 9.35 mmol) in toluene (15 mL) was heated at 100 °C for 4 hours. The reaction was concentrated, and the residue was purified by reverse phase chromatography to obtain the title compound (530 mg, yield: 45%). MS (ESI): 317 [M+H]+.
Example - 121
Figure imgf000060_0002
N-(2-(6-{3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-l-[3-(4-methyI-piperazin-l-yI)-propyl]- ureido}-pyrimidin-4-y]amino)-phenyl]-acryIamide
The compound was synthesized following the approach outlined in Procedure 21 (Example 142), substituting 2-nitroaniline and N-[3-(4-Methyl-piperazin-l-yl)-propyl]-N'-(2-nitro-phenyl)- pyrimidine-436-diamine (preparation shown below) in step (e) to afford the title compound (8 mg, yield: 1.2% over six steps). 1H NMR (300 MHz, Methanol-d4) δ 8.17 (s, 1H), 7.46 (d, 1H), 7.32 (d, 1H), 7.12-7.08 (m, 2H), 6.62 (s, 1H), 6.29-6.14 (m, 3H), 5.58 (d, 1H), 3.82 (t, 2H)5 3.75 (s, 6H), 2.27-2.19 (m, 8H), 2.06 (s, 3H), 1.67 (t, 2H); MS (ESI): 643 [M+H]+. Preparation of N-[3-(4-Methyl-piperazm-l-yl)-propyl]-N'-(2-nitro-phenyI)-pyrimidme-4,6- diamine
Figure imgf000061_0001
a. (6-Chloro-pyrimidin-4-yl)~[3-(4-methyl^iper zin-l-yl)-propy
To a solution of 4,6-dichIoro- pyrimidine (1.5 g, 10 mmol) and DIPEA (1.55 g, 12 mmol) in iPrOH (50 mL) was added a solution of 3-(4-methyI-piperazin-l-yl)-propylamine (1.73 g, 11 mmol) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. Water was added and the mixture was extracted with DCM. The combined extracts were washed with brine, dried over anhydrous Na2S04, and concentrated to afford the title compound (1.4 g, 51%), which was used directly in the next step without further purification. MS (ESI): 270
[M+Hf.
Figure imgf000061_0002
b. N-[3-(4-Methyl~piperazin-l-yl) ropyl]-N'-(2-nitro^henyl)-pyn
A degassed mixture of (6-chloro-pyrimidin-4-yl)-[3-(4-methyl-piperazin-l-yl)-propyl]-amine (600 mg, 2.22 mmol), nitroanline (317 mg, 2.3 mmol), Pd2(dba)3 (210 mg} 0.23 mmol), Xantphos (265 mg, 0.46 mmol) and Cs2C0 (1.81 g, 5.55 mmol) in toluene (15 mL) was heated at 100 °C for 4 hours. The reaction was concentrated, and the residue was purified by reverse phase chromatography to obtain the title compound (400 mg, yield: 48%). MS (ESI): 372
[M+H]+.
Example - 122
Figure imgf000062_0001
N-(2-{6-[3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-l-m
piperidm-4-yl-phenyl)-acrylamidc trifluoroacetic acid
The compound was synthesized following the approach outlined in Procedure 2E (Example 111), replacing steps (e) and (f) with the following procedure: To a solution of fert-butyl 4-(3-amino-4- (6-(3-(2,6-dichloro-3,S-dimethoxyphenyl)-l-methyl-3-((2-(1rimethylsilyl)ethoxy)
me1hyl)iffeido)pyrimidin-4-ylarnino)phenyl)piperidine-l-carboxylate (Procedure 2E, Example 111, 65 mg, 0.084 mmol) in THF (15 mL) was added a solution of acryloyl chloride (10 mg/mL, 0.9 mL, 0.1 mmol) dropwise at -10 °C, and the resulting mixture was stirred at 0 °C for 1 hour. MeOH (5 mL) was added to quench the reaction, and the reaction was concentrated. The residue was dissolved in DCM (2 mL) and added dropwise to a mixture of DCM/TFA (2:1 , 3 mL). The mixture was stirred at room temperature for 1 hour and then concentrated. The residue was purified by prep-HPLC to afford the title compound (23 mg, yield: 47%). Ή NMR (300 MHz, DMSO-d6) 6 1 1.92 (s, 1H), 9.64 (s, 1H), 8.98 (s, 1H), 8.60 (m, 1H), 8.36 (s, 1H), 8.25 (m, 1H), 7.66 (s, 1H), 7.51 (d, 1H), 7.05 (d, 1H), 6.90 (s, 1H), 6.52 (dd, 1H), 6.41 (s, 1H), 6.24 (d, 1H), 5.74 (d, 1H), 3.96 (s, 6H), 3.49 (d, 2H), 3.28 (s, 3H), 3.02 (q, 2H), 2.85 (t, 1H), 1.96 (d, 2H), 1.78 (q, 2H); MS (ESI): 600 [M+Hj+.
- 123
Figure imgf000063_0001
toluene, 100°C, 1 h
Figure imgf000063_0002
toluene, 100 °C, 2.5 h
Figure imgf000063_0003
N-(2- {6- [3-(2,6-Dichloro-3 ,5-dim ethoxy-pheny I)-l -methy 1-u reido] -py rimidin-4-y la mino}-5- morphoIin-4-yl-phenyl)-acryIamide
Figure imgf000063_0004
a. di-tert-Butyl 4-bromo-2-nitrophenylcarbamate
A mixture of 4-bromo-2-nitroaniline (10 g, 46 mmol), (Boc)20 (20.7 g, 95 mmol) in THF (250 mL) was heated under reflux overnight. The mixture was concentrated to afford the title compound (19.2 g, yield: quant.) which was used directly in next step without further purification.
Figure imgf000064_0001
toluene, 100 °C, 1 h
b. (4~Morpholin-4-yl-2-nitro~phenyl)-carbamic acid di-tert-butyl ester
A degassed mixture of tert-butyl 4-bromo-2-nitrophenyIcarbamate (1 g, 2.4 mmol), morpholine (314 mg, 3.6 mmol), Pd2(dba)3 (220 mg, 0.24 mmol), Xantphos (278 mg, 0.48 mmol) and
CS2CO3 (1.56 g, 4.8 mmol) in toluene (30 mL) was heated at 100°C for 1 hour. The reaction was concentrated, and the residue was purified by flash chromatography on silica to obtain a crude mixture of the title compound and mono-Boc product (744 mg). The mixture was used directly in the next step without further purification. MS (ESI): 324 [M-Boc+HJ+.
Figure imgf000064_0002
c. 4-Morpholitt~4-yl-2-nitro-ph enylamine
To a solution of (4-morpholin-4-yl-2-nitro-phenyl)-carbamic acid di-tert-butyl ester and mono- Boc product (744 mg) in DCM (20 mL) was added TFA (10 mL) at 0 °C, the resulting mixture was stirred for 3 hours at room temperature. After removal of all volatiles in vacuo, the residue was re-dissolved in DCM, neutralized by saturated aqueous NaHC03> and extracted with DCM. The combined extracts were concentrated and the residue was purified by flash chromatography on silica to obtain the title compound (290 mg, yield: 54% over two steps). MS (ESI): 251
[M+H]+.
Figure imgf000065_0001
rf. l-(2,6-Dichloro~3,5-dimethoxy-phenyl)-3-methyl~3-[6-(4-morp^
phenylamino)~pyrimidin-4~yl]-l-(2~trimethylsilanyl-etto
A degassed mixture of 4-morpholin-4-yl-2-nitro-phenylamine (290 mg, 1.3 mmol), l-(6-chloro- pyrimidin-4-yl)-3-(2,6-dichioro-3,5~dimemoxy-ph
ethoxymethyl)-urea (Procedure 2E , step b; 624 mg, 1.2 mmol), Pd2(dba)3 (110 mg, 0.12 mmol), Xantphos (139 mg, 0.24 mmol) and Cs2C(¾ (782 mg, 2.4 mmol) in toluene ( 5 mL) was heated at 100°C for 2.5 hours. The reaction was concentrated, and the residue was purified by chromatography flash on silica to obtain the title compound (440 mg, yield: 49%) as a red solid. MS (ESI): 708 [M+H]+.
Figure imgf000065_0002
e. {6^-(2,6-Dichloro'3,5-dimethoxy-phenyl)-l-methyl-3-(2-MmethylsU
ureido]-pyrimidin^-yl}-(4~morpholin-4-yl-2-nitro-phenyl)-carba acid tert-butyl ester
A mixture of l-(2,6-dichloro-3J5-dimethoxy-phenyl)-3~methyl-3-[6-(4-morpholm-4-yl-2-nitro- phenylamino)-pyrimidin-4-yl]-l-(2-trimethylsilanyl-ethoxymethyl)-urea (200 mg, 0.28 mmol), (Boc)20 (93 mg, 0.42 mmol) and catalytic amount of DMAP in THF (10 mL) was heated under reflux for 1 hour. The mixture was concentrated and the residue was used for the next step without further purification. MS (ESI); 808 [M+H .
Figure imgf000066_0001
(2-Amino-4-morpholin-4-yl-phenyl)-{6-[3-(2,6-dichloro-3,5-d^
(2-trimethylsilanyl-ethoxymethyl)-ureidoJ-pyrimidin-4-yl}-carb m acid tert-butyl ester
To a solution of {6-[3-(2,6-dichloro-3,5-dimethoxy-pheny
ethoxymemylj-ureidoj-pyrimidin-^ acid tert- butyl ester (crude, prepared above) in MeOH (20 mL) was added Raney-Ni (suspension in water) at room temperature, the resulting mixture was stirred for 2 hours under hydrogen atmosphere (1 atm). The reaction was filtered and concentrated. The residue was washed twice with MeOH to obtain title product (160 mg, yield: 70%). MS (ESI):778 [M+H]+.
Figure imgf000066_0002
g. (2-Acryloylamino-4-morphoUn~4-yl-phenyl)-{6-[3~(2,6-dichloro-3,5-dim
methyl-3-(2-trimethyteilanyl-ethoxymethyl)-ureid^ acid tert-butyl ester
To a solution of (2-amino-4-morpholin-4-yl-phenyl)-{6-[3-(2,6-dichloro-3,5-dimethoxy-phenyl)- l-methyl-3-(2-trimethylsilanyl-ethoxymethyl)-ureido]-pyrimidin-4-yl}-carbamic acid tert-butyl ester (80 mg, 0.103 mmol) in DCM (5 mL) was added a solution of TEA (10 mg/mL, 1.2 mL,
0.12 mmol) and a solution of acryloyl chloride (10 mg/mL, 1 mL, 0.11 mmol) dropwise at 0 °C, and the resulting mixture was stirred at room temperature for 1 hour. LC-MS showed that the reaction was complete. Water (5 mL) was added to quench the reaction, and the reaction mixture was extracted with DCM. The combined extracts were washed with brine, dried over anhydrous sodium sulfate and filtered. The filtrate was evaporated under vacuum to give crude product, which was used for the next step without further purification.
Figure imgf000067_0001
h. N-(2-{6-[3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-l-methyl~ureifo
morpholin~4-yl~phenyl)-acrylamide
To a solution of (2-acryloylamino-4-morphol n-4-yl-ph^^
phenyl)-l-methyl-3-(2-trimethylsilanyl-ethoxymethyl)-ureido]-pyrimidi acid tert-butyl ester (crude, prepared above) in CH2CI2 (10 mL) was added TFA (3 n L) at 0 °C, the resulting mixture was stirred for 1 hour at room temperature. The reaction mixture was concentrated and neutralized wifhNH3.H20 to give the crude compound, which was purified by Prep-HPLC to obtain title compound (10 mg, yield: 16% in two steps). lH NMR (300 MHz,
DMSO-d6) δ 12.05 (s, IH), 9.60 (s, IH), 8.74 (s, IH), 8.32 (s; IH), 7.33-7.30 (ms 2H), 6.89 (s, IH), 6.83 (d, IH), 6.48 (dd, IH), 6.25-6.21 (m, 2H), 5.72 (d, IH), 3.96 (s, 6H), 3.73 (br, 4H), 3.44 (s, 3H), 3.10 (br, 4H); MS (ESI): 602 [M+H]+
Example - 124
Figure imgf000067_0002
N-{2-{6-[3-(2,6-DichIoro-3,5-dimethoxy-phenyI)-l^
(3 ,5-dim ethyl-piperazin-1 -y l)-p henyl] -acrylamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 4~(3,5-dimethyl-piperazin-l-yl)-2-nitro-phenylamine (preparation shown below) in step (d) to afford the title compound (26 mg, yield: 30% over three steps). 1H NMR
(300 MHz, Methanol^) δ 8.32 (s, IH), 7.36-7.33 (m, 2H), 6.95 (d, IH), 6.80 (s, IH), 6.43-6.37 (m, 2H), 6.15 (s, IH), 5.77 (d, IH), 3.95 (s, 6H), 3.70 (d, 2H), 3.29 (s, 3H), 3.18-3.13 (m, 2H), 2.45 (t, 2H), 1.24 (d, 6H); MS (ESI): 629 [M+H .
Preparation of 4-(3, -Dimethyl-piperazin-l-yl)-2-nitro-phenyIamine
Figure imgf000068_0001
. di-tert-Butyl 4-brotno-2-nitrophenylcatbamate
A mixture of 4~bromo-2-nitroanilme (10 g, 46 mmol), (Boc)20 (20.7 g, 95 mmol) in THF (250 mL) was heated under reflux overnight. The mixture was concentrated to afford the title compound (19.2 g, quant) which was used for the next step without further purification.
Figure imgf000068_0002
b. f4-(3,5^imet yl-piperazin~l~yl)-2-nitro-phenylJ-carbamic acid di~tert-butyl ester
A degassed mixture of di-tert-Butyl 4-bromo-2-nitrophenylcarbamate (1 g, 2.4 mmol), 2,6- dimethyl-piperazine (410 mg, 3.6 mmol), Pd2(dba)3 (220 mg, 0.24 mmol), Xantphos (278 mg, 0.48 mmol) and Cs2C03 (1.56 g, 4.8 mmol) in toluene (30 mL) was heated at 100 °C for 1 hour. The reaction was concentrated, and the residue was purified by flash chromatography on silica to obtain a mixture of the title compound and mono-Boc product (600 mg). The mixture was used directly in the next step without further purification. MS (ESI): 350 [M-Boc+HV\
Figure imgf000068_0003
c. 4~(3,5~Dimethyl-piperazin-l-yl)~2-nitro-phenyl mine
To a solution of [4-(3,5-dimethyl-piperazin-l-yl)-2-nitro-phenyl]-carbamic acid di-tert-buty ester and mono-Boc product (600 mg) in DCM (20 mL) was added TFA (10 mL) at 0 °C, the resulting mixture was stin-ed for 3 hours at room temperature. After removal of all voiatiles in vacuo, the residue was re-dissolved in DCM, neutralized by saturated aqueous NaHC03, and extracted with DCM. The combined extracts were concentrated and the residue was purified by flash chromatography on silica to obtain the title compound (233 mg, yield: 39% over two steps), MS (ESI): 251 [M+H]+.
Example - 125
Figure imgf000069_0001
N-[2-{6-[3-(2i6-Dichloro-3,5-dimethoxy-phenyI)-l-methyl-ureidoJ-pyrimidin-4-ylamiao}-5- (4-methanesulfonyI-piperazm-l-yl)-phenyI]-acrylamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 4-(4-methanesulfonyl-piperazin-l-yl)-2-nitro-phenylamine (preparatin shown below) in step (d) to afford the title compound (15 mg, yield; 9.6% in five steps). 1H NMR (300 MHz, DMSO-d6) δ 12.03 (s, 1H), 9.62 (s, 1H); 8.76 (s, 1H), 8.33 (s, IK), 7.37-7.32 (m, 2H), 6.89-6.83 (m, 2H), 6.46 (dd, 1H), 6.26-6.21 (m, 2H), 5.72 (d, 1H), 3.93 (s, 6H), 3.34-3.16 (m, 11H), 2.93 (s, 3H); MS (ESI): 679 [M+H]+.
Preparation of 4-(4-Methanesulfonyl-piperazra-l-yI)-2-nitro-phenylamine
Figure imgf000069_0002
a. di-tert~Butyl 4-bromo-2-nitrophenylcarbamate
A mixture of 4-bromo-2-nitroaniline (10 g, 46 mmol), (Boc)20 (20.7 g, 95 mmol) in THF (250 mL) was heated under reflux overnight. The mixture was concentrated to afford the title compound (19.2 g, quant) which was used directly in next step without further purification.
Figure imgf000070_0001
toluene, 100°C, 1 h
b. [4-(4-Methanesulfonyl^iperazin-l-yl)~2-nitro-phenyl]-carbamic acid di-tert-butyl ester A degassed mixture of di-tert-butyl 4-bromo-2-nitrophenylcarbamate (1 g, 2.4 mmol), 1- methanesulfonyl-piperazine (590 mg, 3.6 mmol), Pd2(dba)3 (220 mg, 0.24 mmol), Xantphos (278 mg, 0.48 mmol) and Cs2C03 (1.56 g, 4.8 mmol) in toluene (30 mL) was heated at 100 °C for 1 hour. The reaction was concentrated, and the residue was purified by flash chromatography on silica to obtain a mixture of the title compound and mono-Boc product (755 mg). The mixture was used directly in next step without further purification. MS (ESI): 400 [M-Boc+H]+.
Figure imgf000070_0002
c. 4-(4-Methanesulfonyl~piperazin-l-yl)-2-nitro-phenylamine
To a solution of [4-(4-methanesulfonyl-piperazin-l-yl)-2-nitro-phenyl]-carbamic acid di-tert- butyl ester and mono-Boc product (755 mg) in DCM (20 mL) was added TFA (10 mL) at 0 °C, the resulting mixture was stirred for 3 hours at RT. After removal of all volatiles in vacuo, the residue was re-dissolved in DCM, neutralized by saturated aqueous NaHC03, and extracted with DCM. The combined extracts were concentrated and the residue was purified by flash chromatography on silica to obtain the title compound (290 mg, yield: 40% in two steps). MS (ESI): 301 [M+H]+.
- 126
Figure imgf000071_0001
N-[2-{6-[3-(2,6-DICHLORO-3,5-DIMETHOXY-PHENYL)-l-METHYL-URErDO]-
PYRIMIDIN-4-YLAMINO}-5-(3-DIMETHYLAMINO-PROPYL)-PHENYL]-
ACRYLAMIDE
The compound was synthesized following the approach outlined in Procedure 2G (Example 123)j substituting 4-(3-Dimethylamino-prop-l-ynyl)-2-nitro-phenylamine (preparatin shown below) in step (d) and platinum oxide in step (f) to afford the title compound (4 mg, yield: 1.4% in five steps). 1H-NM (300 MHz, DMSO-d6) δ 11.98 (s, IH), 9.66 (d, IH), 8.88 (s, IH), 8.36 (s, IH), 7.52 (d, IE), 7.44 (d, IH), 7.04 (dd, IH), 6.90 (s, IH), 6.48 (dd, IH), 6.34 (s, IH), 6.24 (dd, IH), 5.72 (dd, IH), 3.94 (s, 6H), 3.25 (s, 3H), 2.59 (t, 2H), 2.24 (t, 2H), 2.11 (s. 6H), 1.71 (m, 2H); MS (ESI) 602 [M+H]+.
Preparation of 4-(3-DimethyIamino-prop-l-ynyl)-2-nitro-phenylamine
Figure imgf000071_0002
a. 4-(3-Dimethylcffliino-prop-l-ynyl)-2~nitro-phenylamine
To a stirred solution of 4-bromo-2-nitro-phenylaraine (1.08 g, 5 mmol) and dimethyl-prop-2- ynyl-amine (1.0 g, 12 mmol) in TEA (20 mL) was added Pd(PPh3)2Cl2 (0.7 g, 1 mmol) and Cul (360 mg, 2 mmol). The solution was stirred at 60 °C under nitrogen for 3 hours. The solution was evaporated with silica gel and purified by flash chromatography on silica to give the title compound (1.1 g, 70% purity). The title compound was taken directly to the next step without further purification. MS (ESI) 221 [M+H] +. Example - 127
Figure imgf000072_0001
N-[2-{6-[3-(2,6~DichIoro-3,5-dimethoxy
(2-dimethyIamino-ethoxy)-phenyl]-acrylamide
The compound was synthesized following the approach outlined in Procedure 2G for compound 123, substituting 4-(2-Dimethylamino-ethoxy)-2-nitro-phenylarnine (preparatin shown below) in step (d) and platinum oxide in step (f) to afford the title compound (4.3 mg, yield: 8% over five steps). 1H-NM (300 MHz, DMSO-d6) δ 12.04 (s, IH), 9.58 (s, IH), 8.81 (s, IH), 8.33 (s, IH), 7.43 (d, IH), 7.33( d, IH), 6.89 (s, IH), 6.79 (dd, IH), 6.51 (dd, IH), 6.26-6.20 (m, 2H), 5.73 (dd, IH), 4.04 (t, 2H), 3.96 (s, 6H), 3.27 (s, 3H), 2.67 (t, 2H), 2.24 (s, 6H); MS (ESI) 604 [M+H]+.
Preparation of 4-(2-Dimethylammo-ethoxy)-2-nitro-phenylamine
Figure imgf000072_0002
a. 4-(2-Dimethylamino~eth oxy)-2-n itro-phenylamine
To a stirred solution of 4-amino-3-nitro-phenol (1.54 g, 10 mmol) and (2-chIoro-ethyl)-dimethyl- amine hydrogen chloride (1.43 g, 10 mmol) in butanone (40 mL) was added Cs2C03 (10 g, 30 mmol) and Nal (150 mgt 1 mmol). The solution was slowly heated to 80 °C over one hour. Then the solution was stirred at 80 °C for 2 hours. The solution was filtered through Celite® and washed with acetone. The solution was evaporated with silica gel and purified by flash chromatography on silica to afford the title compound (1.0 g, yield: 45%) as a brown solid. 1H-NMR (300 MHz, DMSO~d6) δ 7.38 (d, IH), 7.24 (s, 2H), 7.16 (dd, 2H), 6.99 (d, IH), 3.98 (t, 2H), 2.58 (t, 2H), 2.20 (s, 6H); MS (ESI) 226 [M+H]+. Example - 128
Figure imgf000073_0001
But-2-ynoic acid (2-{6-[3-(2,6-dichloro-3i5-dimethoxy-phenyl)-l-methyI-ureido]-pyrimidin- 4-yIamino}~phenyI)-amide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 2-nitro-phenylamine in step (d), omitting step (e), and replacing step (g) with the following procedure below to afford the title compound (7 mg, yield: 4.8% over five steps). 1H NM (300 MHz, DMSO-d6) 6 11.97 (s, 1H), 10.07 (s, 1H), 8.89 (s, 1H), 8.39 (s, 1H), 7.60 (d, 1H), 7.53 (d, 1H), 7.24-7.15 (m, 2H), 6.90 (s, 1H), 6.44 (s, 1H), 3.93 (s, 6H), 3.29 (s, 3H), 2.22 (s, 3H); MS (ESI): 529 [M+H]+.
Figure imgf000073_0002
To a solution of (2-amino-phenyl)-{6-[3-(2,6-dichloro-3,5-dimethoxy-phenyl)-l-methyl-3-(2- trimethylsilanyl-ethoxymethyl)-ureido]-pyrimidin-4-yl}-carbamic acid tert-butyl ester (50 mg, 0.075 mmol) and DCC (42 mg, 0.2 mmol) in DCM (50 mL) was added a solution of but-2~ynoic acid (13 mg, 0.15 mmol) in DCM (10 mL) at 0 °C, and the resulting mixture was stirred at room temperature overnight. Water (1 mL) was added to quench the reaction. The mixture was concentrated and the residue was purified by reverse phase chromatography to obtain the title compound (20 mg, yield: 34%). MS (ESI): 659 [M+H]+.
Compound 129 was synthesized in a similar manner as compound 100. - 130
Figure imgf000074_0001
N-(2-{6-[l-Ben2yl-3-(2,6-dichloro~3^
ph eny 1)- acryJamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 2-nitro-phenylamine and 1 -benzyl- l-(6-chloro-pyrimidin-4-yl)-3 -(2,6- dichloro-3 }5-dimethoxy-phenyl)-3-(2-trimethylsilanyl-ethoxymethyl)-urea (preparation shown below) in step (d) to afford the title compound (26 mg, yield: 20% over five steps).1H NMR (300 MHz, DMSO-d6) δ 12.27 (s, IH), 9.68 (s, IH), 8.90 (s, IH), 8.39 (s, IH), 7.69 (d, IH), 7.33-7.19 (m, 6H), 6.92 (s, IH), 6.45 (dd, 1H)} 6.24 (d, IH), 6.18 (s, IH), 5.74 (d, IH), 5.07 (s, 2H), 3.95 (s, 6H); MS (ESI): 593 [M+H]+.
Pre aration of 1 -B enzyl-1 -(6-ch loro-py rimidin-4- l)-3-(2,6-d ieh Ioro~3 ,5-dimethoxy- ph eny l)-3-(2-trim ethylsilany 1-eth oxym eth yl)-urea
Figure imgf000074_0002
a. Benzyl-(6-chloro-pyrimidin~4-yl)-amine
To a solution of 4,6-dichloro-pyrimidine (1.5 g, 10 mmol) in iPrOH (40 mL) and DIPEA (1.55 g, 12 mmol) was added a solution of benzylamine (1.28 g, 12 mmol) at room temperature. The resulting mixture was stirred at room temperature for 2.5 hours. Water was added and the mixture was extracted with DCM. The combined extracts were washed with brine, dried over anhydrous N 2S04, and concentrated to give the crude product, which was purified by flash chromatography on silica to obtain the title compound (1.5 g, yield: 68%) as a white solid. 1H
NMR (300 MHz, CDC13) δ 8.35 (br s, IH), 7.20-7.38 (m, 5 H), 6.35 (s, IH), 4.52 (s, 2H); MS (ESI): 220 [M+H]+.
Figure imgf000075_0001
b. l-Benzyl-l-(6-chloro-pyrimidin-4-yl)-3-(2,6-dichhrO'3,5'dim
trimethylsilanyl~ethoxymethyl)-urea
To a solution of benzyl"(6-chloro-pyrimidin-4-yl)-amine (800 mg, 3,64 mmol) in DMF (15 mL) was added NaH (60%, 218 mg, 5.45 mmol) at 0 °C, and the mixture was stirred for 10 minutes at room temperature. A solution of l-isocyanato-3,5-dimethoxy-benzene (Procedure 2A , steps a-d; 1.35 g, 5.45 mmol) in DMF (2 mL) was added dropwise at 0 °C. The resulting mixture was stirred at room temperature for 2 hours. Saturated aqueous NH4CI (2 mL) was added to quench the reaction. The mixture was concentrated and extracted with DCM. The combined extracts were washed with brine, dried over anhydrous Na2S04, and concentrated to give the crude product, which was purified by flash chromatography on silica to obtain the title compound (1.7 g, yield: 77%). MS (ESI): 599 [M+H]+.
Example - 131
Figure imgf000075_0002
N-(2-{6-[3~(2,6-Dic oro-3,5-dimethoxy- he^
ylamino}-phenyl)-acrylamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 2-nitro-phenylamine and l-(6-chloro-pyrimidin-4-yl)-3-(2,6-dichloro~3,5- dimemoxy-phenyl)-l-pyridin-2-ylmethyl-3-(2-trimethylsilanyl-emoxymethyl)-urea (prepar shown below) in step (d) to afford the title compound (35 mg, yield: 13% over five steps).1H NM (300 MHz, DMSO-d6) δ 12,15 (s, 1H), 9.65 (s, 1H), 8.83 (s, 1H), 8.50 (s, 1H), 8.39 (s, 1H), 7.80 (t, 1H), 7.66 (d, 1H), 7.31-7.25 (m, 3H), 7.17 (t, 1H), 7.09 (t, 1H), 6.91 (s, 1H), 6.45 (dd, 1H), 6.39 (s, 1H), 6.23 (d, 1H), 5.74 (d, 1H), 5.17 (s, 2H), 3.94 (s, 6H); MS (ESI): 594
[M+HJ+.
Preparation of l-(6-ChIoro-pynmidin-4-yl)-3-(2,6-dichloro-3,5-dimethoxy-phenyl)-l- pyridin-2-ylmethyI-3-(2-trimethylsilanyl-ethoxyraethyl)~urea
Figure imgf000076_0001
a. ( loro-pyrimidin~4~yl)-pyridin-2-ylmethyl-amine
To a solution of 4,6-dichloro-pyrimidine (1 g, 7 mmol) in iPrOH (40 mL) and DIPEA (1.16 g, 9 mmol) was added a solution of 2-pyridinylmethanamine (970 mg, 9 mmol) at room temperature. The resulting mixture was stirred at room temperature for 2.5 hours. Water was added and the mixture was extracted with DCM. The combined extracts were washed with brine, dried over anhydrous sodium sulfate and concentrated to give a crude product, which was purified by flash chromatography on silica to obtain the title compound (1.2 g, yield: 78%). MS (ESI): 221
[M+H]+.
Figure imgf000076_0002
b. l-(6-Chloro-pyrimidin-4-yl)-3-(2,6-d hloro-3,5-dimethoxy-^^
(2-trimethylsilanyl-ethoxymethyl)-urea
To a solution of (6-chloro-pyrimidin-4-yl)-pyridin-2-ylmethyl-amine (200 mg, 0.91 mmol) in
DMF (5 mL) was added NaH (60%, 55 mg, 1.37 mmol) at 0 °C, and the mixture was stirred for
10 minutes at room temperature. A solution of l-isocyanato-3,5-dimethoxy-benzene (Procedure
2A , a-d; 337 mg, L37 mmol) in DMF (2 mL) was added dropwise at 0 °C. The resulting mixture was stirred for 30 minutes. SEMCI (230 mg, 1.37 mmol) in DMF (2 mL) was added and the reaction mixture was stirred at room temperature for 1 hour. Saturated aqueous NH4C1 was added to quench the reaction. The mixture was diluted with water and extracted with EtOAc. The combined extracts were washed with water and brine, dried over anhydrous Na2S04 and filtered. The filtrate was concentrated to give a crude product, which was purified by flash
chromatography on silica to obtain the title product (420 mg, yield: 78%). MS (ESI): 598
Example - 132
Figure imgf000077_0001
N-(2-{6-[3-(2,6-DICHLORO-3,5-DIMETHOXY-PHENYL)-l-PYRIDIN-4-YLMETHYL- UREIDO]-PYRIMIDIN-4-YLAMINO}-PHENYL)~ACRYLAMIDE
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-l-[6-(2-nitro-phenylamino)- pyrimidin-4-yl]-l-pyridin-4-ylmethyl-urea in step (e) to afford the title compound (14 mg, yield: 8.1%). 1H NMR (300 MHz, DMSO-d6) δ 12.03 (s, 1H), 9.68 (s, 1H), 8.92 (s, 1H), 8.49 (d, 2H), 8.39 (s, 1H), 7.67 (d, 1H)5 7.27-7.17 (m, 4H), 7.10 (t, 1H), 6.92 (s, 1H), 6.48 (dd, 1H), 6.24 (d, 1H), 6.15 (s, 1H), 5.74 (d, 1H), 5.09 (s, 2H), 3.94 (s, 6H); MS (ESI): 594 [M+Hf.
Preparation of 3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-l-[6-(2-nitro-phenyIamino)- pyrimidin-4-ylj-l-pyridin-4-ylmethyl-urea
Figure imgf000077_0002
a. ( 6-Chloro-pyrimidin-4-yi)-pyridin-4-ylmethyl-amine
To a solution of 4,6-dichloro-pyrimidine (1.5 g, 10.5 mmol) and DIPEA (1.62 g, 12.6 mmol) in iPrOH (40 mL) was added 4-pyridinylmethanamine (1.2 g, 11 mmol) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. Water was added and the mixture was extracted with DCM. The combined extracts were washed with brine, dried over anhydrous Na2S04, and concentrated to give the crude product, which was purified by flash chromatography on silica to obtain the title compound (1.8 g, yield: 80%). MS (ESI): 221
[M+H]+.
Figure imgf000078_0001
b. N-(2-Nitro^henyl)-N'^yndin^-ylmethyl^yrimidine-4,6-diamine
A degassed mixture of (6-chloro-pyrimidin-4-yl)-pyridin-4-ylmethyl-amine (500 mg, 2.27 mmol), 2-nitroaniline (317mg, 2.3 mmol), Pd2(dba)3 (200 mg, 0.22 mmol), Xantphos (253 mg, 0.44 mmol) and Cs2C03 (1.48 g, 9.35 mmol) in toluene (10 mL) was heated at 100 °C for 3 hours. The reaction was concentrated, and the residue was purified by reverse phase
chromatography followed by flash chromatography on silica to obtain the title compound (330 mg, yield: 45 +.
Figure imgf000078_0002
c 3-(2,6~Dichloro~3t5-dimethoxy-phenyl)-l-[6-(2-nitro-phenylam
pyridin-4-ylmethyl-ure
To a solution of N-(2-mtro-phenyl)-N'-pyridin-4-ylmethyl-pyrimidine-4,6-diamine (330 mg, 1.02 mmol) in DMF (10 mL) was added NaH (60%, 56 mg, 1.4 mmol) at 0 °C, and the mixture was stirred for 30 minutes at room temperature. A solution of l-isocyanato-3,5-dimethoxy- benzene (Procedure 2A , steps a-d; 345 mg, 1.4 mmol) in DMF (2 mL) was added dropwise at 0 °C. The resulting mixture was stirred at room temperature for 2 hours. Saturated aqueous NH4CI (2 mL) was added to quench the reaction. The mixture was concentrated and extracted with DCM. The combined extracts were washed with brine, dried over anhydrous Na2S04, and concentrated to give the crude product, which was purified by prep-TLC to obtain the title compound (190 mg, yield: 33%). MS (ESI): 570 [M+H]+. Example - 133
Figure imgf000079_0001
Preparation of 5-(4-ethyl-piperazin-l-yl)-2-nitro-phenylamine
Figure imgf000079_0002
1. 5-(4-Ethyl-piperazin-l-yl)-2-nitro-phenyl mine
A mixture of 1 -ethyl-piperazine (1.2 mL, 9.6 mmol). 5-fluoro-2-nitro-phenylamine (1 g, 6.4 mmol), DIPEA (1.24 g, 9.6 mmol) in DMF (15 mL) was heated at 80 °C overnight. The reaction mixture was poured into ice water and extracted with EtOAc. The combined extract was washed with brine, dried over anhydrous Na2S04, and concentrated to obtain a crude product, which was purified by flash chromatography on silica to afford the title compound (1 g, yield: 63%). ESI- MS: 251 [M+Hf.
N- [2- {6- [3-(2 ,6-Dichloro-3,5-dimethoxy~ph en ^
(4-ethyl-piperazin-l-yl)-phenyl]-acrylamide trifluoroacetic acid salt
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 5-(4-ethyl-piperazin-l-yl)-2-nitro-phenylamine in step (a) to afford the title compound (20 mg, yield: 39%). 1H NMR (300 MHz, Methanol-d4) δ 8.37 (s, 1H), 7.49 (d, 1H), 7.25 (s, 1H), 6.98 (d, 1H), 6.82 (s, 1H), 6.43-6.38 (m, 3H), 5.78 (d, 1H), 3.96-3.88 (m, 8H), 3.68- 3.64 (m, 2H), 3.37 (s, 3H), 3.33-3.08 (m, 6H), 1.40 (t, 3H); MS (ESI): 629 [M+H]+. Example - 135
Figure imgf000080_0001
N- [2- {6-[3-(2 ,6-Dichloro-3 ,5-d hnethoxy-ph enyl)-l-methyl-ureido -pyrimidra-4-ylamrao}-5- (4-ethyl-piperazin~l-ylniethyl)-phenyl]-acrylamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 4-(4-Ethyl-piperazin-l-ylmethyl)-2-nitro-phenylamine (preparation shown below) in step (d) to afford the title compound (30 mg, yield: 31% over five steps). 1H NMR (300 MHz, DMSO-d6) δ 11.97 (s, IH), 9.71 (s, IH), 8.95 (s, IH), 8.37 (s, IH), 7.60 (s, IH), 7.50 (d, IH), 7.12 (d, IH), 6.90 (s, IH), 6.50 (dd, IH), 6.39 (s, IH), 6.25 (d, IH), 5.73 (d, IH), 3.93 (s, 6H), 3.48 (s, 2H), 3.32 (s, 3H), 2,50-2.25 (m, 10H), 0.98 (t, 3H); MS (ESI): 643 [M+H]+.
Preparation of 4-( -Ethyl-pipera¾in-l-ylmethyl)-2-nitro-phenylaraine
Figure imgf000080_0002
a, 4-(4-Ethyl-piperazin-l-ylmethyl)-2-nitro-phenylamine.
To a stirred solution of 1 -ethyl-piperazine (1.37 g, 12 mmol) in MeOH ( 30 mL) was added Ti(0'Pr)4 (1.73 g, 6 mmol). Then the solution was stirred at room temperature for 15 min. Then 4-amino-3-nitro-benzaldehyde (Procedure 2F, steps a-b, 1.5 g, 9 mmol) in MeOH (10 mL) was added and the solution was stirred at room temperature overnight, ThenNaBH4 (380 mg, 10 mmol) was added and the solution was stirred at room temperature for 1 hour. The solution was diluted with EtOAc and filtered. The filtrate was washed with water and brine, dried over anhydrous Na2S04. Concentration gave product, which was purified by flash chromatography on silica to obtain the title compound (800 mg, yield: 34%). MS (ESI): 265 [M+H]+
Figure imgf000081_0001
N-(2-{6-[3-(2,6^ichloro-3 -dimethoxy-phenyl)-l-(3-methyl-isoxazoI-5-ylmethyI)-ureido]- pyrimidin-4-ylamino}-phenyl)~acryIamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting l-(6-Chloro-pyrimidin-4-yl)-3-(2,6-dichloro-3,5-dimethoxy-phenyl)-l-(3- memyl-isoxazol-5-ylmemyl)-3-(2-trimemylsilanyl-ethoxymethyl)-urea (preparation shown below) in step (d) and iron acetic acid at 60 °C in step (f) to afford the title compound (26 rag, yield: 12% in five steps).1H NM (300 MHz, DMSO-d6) δ 11.78 (s, 1H), 9.72 (s, 1H), 8.97 (s, 1H), 8.40 (s, 1H), 7.66 (d, 1H), 7.45 (d, 1H), 7.21-7.15 (m, 2H)S 6.90 (s, 1H), 6.52-6.43 (m, 2H), 6.26 (d, 1H), 6.09 (s, 1H), 5.74 (d, 1H), 5.16 (s, 2H), 3.93 (s, 6H), 2.18 (s, 3H); MS (ESI): 598 [M+H]+
Prep aration of 1 -(6-Chlor o-py r imidin~4-yl)-3-(2,6~dkh loro-3 ,5-dimeth oxy-ph enyl)- 1-(3- methyI-isoxazol-5-ylmethyl)-3-(2-trimethyIsiIanyI-ethoxymethyI)-urea
Figure imgf000081_0002
a. (6-Chloro-pyrimidin-4-yl)-(3-methyl-isoxazol-5-ylmethyl) -amin e.
To a solution of 4,6-dichloro-pyrimidine (660 mg, 4.46 mmol) in iPrOH (40 mL) and DIEA (690 mg, 5.35 mmol) was added a solution of C-pyridin-2-yl-methylamine (560 mg, 5 mmol) at room temperature. The resulting mixture was stirred at room temperature for 2 hours. Water was added and the mixture was extracted with DCM. The combined extracts were washed with brine, dried over anhydrous sodium sulfate, and concentration under vacuum to give the crude product. The crude product was purified by flash chromatography on silica to obtain the title compound (650 mg, yield: 65%). MS (ESI): 225 [M+H]+.
Figure imgf000082_0001
b. l-(6-Chloro^pyrimidin-4-yl)-3-(2,6-dichloro-3,5-dimethoxy-phen^
ylmethyl)-3-(2-trimethylsilanyl-ethoxymethyl)~urea.
To a solution of (6-chloro-pyrimidin-4-yl)-(3-memyl-isoxazol-5-ylmethyl)-araine (300 mg, 1.34 mmol) in DMF (5 mL) was added NaH (60%, 80 mg, 2 mmol) at 0 °C, and the mixture was stirred for 10 minutes at room temperature. A solution of l-isocyanato-3,5-dimethoxy-benzene (Procedure 2A, steps a-d, 337 mg, 1.37 mmol) in DMF (2 mL) was added dropwise at 0 °C. The resulting mixture was stirred for 0.5 hour. SEMC1 (230 mg, 1.37 mmol) in DMF (2 mL) was added. The reaction mixture was stirred at room temperature for 1 hour. Sat. aqueous NH4C1 was added to quench the reaction. The mixture was diluted with water and extracted with EtOAc. The combined extracts were washed with water and brine, dried over anhydrous sodium sulfate and filtered. The filtrate was evaporated under vacuum to give the crude product, which was purified by flash chromatography on silica to obtain the title product (440 mg, yield: 55%). MS (ESI): 604 [M+H
Example - 137
Figure imgf000082_0002
(2-dimethyIamino-ethyI)-phenyI]"acrylamide TFA salt
The compound was synthesized following the approach outlined in Procedure 2G substituting 5- (2-dimethylamino-ethyl)-2-nitro-phenylamine (prepared by the method outlined below) in step (d) to afford the title compound (39 mg, yield: 29%) as a TFA salt. 1H NMR (300 MHz, DMSO- d6) 5 11.86 (s, IH), 9.76-9.70 (m, 2H), 9.02 (s, IH), 8.40 (s, IH), 7.86 (d, IH), 7.51 (s, IH), 7.12 (d, IH), 6.90 (s, IH), 6.57-6.48 (m, 2H), 6.24 (d, IH), 5.74 (d, 1H)} 3.94 (s, 6H), 3.35-3.28 (m, 5H), 2.98-2.92 (m, 2H), 2.79 (s, 6H); MS (ESI): 588 [M+H .
Preparation of 5-(2-Dimethy!amino-ethyi)-2- tro-phenyIamine
Figure imgf000083_0001
a. [2-(3-Fluoro-4-nitro-phenyl)-vinyl]-dimethyl-amine.
A mixture of 2-fluoro-4-methyl-l-nitro-benzene (3 g, 19.3 mmol), N,N-dimethylformamide dimethylacetal (10 mL) and 3 mL of DMF (30 mL) was heated at 125°C for 1 hour. The mixture was cooled and concentrated under reduced pressure to give a purple solid. Trituration with hexanes gave the pure title pr 211 [M+H]+.
Figure imgf000083_0002
b. [2-(3-Fluoro-4-nitro-phenyl)-ethyl]~dimethyl-amine.
To a solution of [2-(3-fluoro-4-nitro-phenyl)-vinyl]-dimethyl-amine (1.7 g, 8 mmol) in MeOH, was added NaBH3CN (770 mg, 12 mmol) and one drop of AcOH. The reaction mixture was stirred at room temperature for 2 hours and quenched with water. After removal of all volatiles in vacuo, the residue was extracted with 10% methanol in DCM twice. The combined extracts were washed with brine, dried over anhydrous sodium sulfate. Concentration under vacuum gave a crude product, which was purified by reverse phase column to afford the title compound (1.08 g, yield: 63%). MS (ESI): 21 +H]+
Figure imgf000083_0003
c. S-(2-Dimethylamino~ethyl)-2-nitro-phenylamine
To a solution of [2-(3-fiuoro-4-nitro-phenyl)-ethyl]-dimethyl-amine (800 mg, 3.76 mmol) in MeOH (20 mL) was added ammonia hydroxide (5 mL). The reaction mixture was heated at 100°C under microwave heating for 1.5 hours. The resulting yellow solid was collected and washed with water, dried under vacuum and combined with a previous batch to give the pure title compound (560 mg, yield: 55%). MS (ESI): 210 [M+H]+.
Procedure 2H: Example - 139
Figure imgf000084_0001
Toluene, 100 °C overnight
Figure imgf000084_0002
N-(2-((6-(3-(3,S-dimethoxyphenyl)-l-methylureido)pyrimidin-4~yl)am
l-yl)phenyl)acrylamide
Figure imgf000085_0001
, I-ft-chloropyrimidin^-yty-S-fttS-dimethoxyphenylJ-l-methylurea
To a stirred solution of 6-chloro-N-methylpyrimidin-4-amine (lg, 6.965 mmol) in dioxane (10 mL) was added DIPEA (3.6 mL, 20.895 mmol) and triphosgene (0.8 lg, 2.786 mmol) under argon atmosphere at 0 °C. The resulting mixture was stirred for lh at 70 °C, and then allowed to cool to room temperature. The resulting mixture was added via cannula to a solution of 3,5- dimethoxyaniline (1.2g, 8.358 mmol) and DIPEA (1.2 mL, 6.965 mmol) in dioxane (4mL) under argon atmosphere at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred for 12h. After completion of the reaction by TLC (Hexanes:EtOAc, 7:3), reaction mixture was diluted with ethyl acetate and a saturated aqueous solution of NaHC03. The aqueous layer was separated and extracted with ethyl acetate (2x5 OmL). The combined organic phase washed with brine, dried on a2S04, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography (Hexanes:EtOAc, 70:30) to afford l-(6-chloropyrimidin-4- yl)-3-(3,5-dimethoxyphenyi)-l-methylurea (1.1 g, yield: 50%) as a white solid. !HNMR (CDCl3j 300MHz): δ 12.38 (s, 1H), 8.71 (s, 1H), 6.99 (s, 1H), 6.79 (d, 2H), 6.26 (t, 1H), 3.81 (s, 6H), 3,45 (s, 3H); 10 [M+H .
Figure imgf000085_0002
b. l-(6-chloropyrimidin-4-yl)-3-(3,5-dimethoxyphenyl)-l-metfy carbonate urea
NaH (0.124g, 3.098 mmol) was added to a stirred solution of l-(6-chloropyrimidin-4-yl)-3-(3,5- dimethoxyphenyl)-l-methylurea (0.5g, 1.549 mmol) in anhydrous DMF (4mL) under an argon atmosphere at 0 °C. The resulting mixture was stirred for 15 min. and added the Di~ rf-butyl dicarbonate (0.50 mL, 2.323 mmol) at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 16h. The reaction mixture was diluted with ethyl acetate and an excess of cold water. The aqueous layer was separated and extracted with ethyl acetate (3x25mL). The combined organic phase was washed with brine, dried over Na2S04, filtered, and concentrated under vacuum. The residue was purified by silica gel column chromatography (Hexanes-.EtOAc, 80:20) to afford the title compound (0.4Sg, 69% of yield) as a white solid. 'HNMR (CDC13, 300MHz): 6 8.72 (d, 1H), 8.78 (d, IH), 6.43-6.37 (m, 3H), 3.77 (s, 6H), 3.49 (s, 3H)} 1.40 (s, 9H); MS (ESI): 424.10 [M+2]+.
Figure imgf000086_0001
overnight
c. l-(3,S-dimethoxyphenyl)-3-(6-((4-(4~ethylpiperazin-l-yl)~2-nitw
yl)-3~methyl~l~tert-butyl carbonate urea
Pd2(dba)3 (0.095g, 0.104 mmol) and Xantphos (0.1202 g, 0.208 mmol) was taken in lOmL of dry toluene in a seal tube under Argon atmosphere at room temperature. The Argon gas purging was continued for additional 5-10 min. Then l-(6-chloropyrimidin-4-yl)-3-(3,5-dimethoxyphenyl)-l- dimethyl-3-te -butyl carbonate urea (0.43g, 1.042 mmol) and 4-(4-ethylpiperazin-l-yl)-2- nitroaniline (Procedure 2C, Steps a - c; 0.317g, 1.25 mmol) was added and the resulting reaction mixture was purged with argon gas for 5 min., and then Cs2C03 (0.676g, 2.08 mmol) was added. The argon gas purging was continued for additional 5 min. before sealing the reaction vial. Then the reaction mixture was heated at 100 °C for 12h. After completion of the reaction by TLC (DCM:MeOH, 98:2), reaction mass was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc (2x25mL) and the combined organic layer was washed with water, brine, dried over Na2S04 and evaporated under vacuum. The crude residue was purified by column chromatography on silica gel (DCM:MeOH/97:3) to afford the title compound (0.250g, 37% of yield). 1HNMR (CDC13, 300MHz): δ 9.30 (s, 1H), 8.58 (s, 1H), 8.17 (d, IH), 7.59 (d, IH), 7.14 (s, IH), 7.08 (dd, IH), 6.42 (d, 2H), 6.36 (t, IH), 3.75 (s, 6H), 3.49 (s, 3H), 3.23 (t, 4H), 2.62 (t, 4H), 2.49 (q, 2H), 1.39 (s, 9H), 1.14 (t, 3H); MS (ESI): 637.4 [M+H]+.
Figure imgf000086_0002
d. tert-butyl (6-(3-(3,5-dimethoxyphenyl)-l-methyl~3~tert-butyl carbonate ureido)pyrimidin-4- yl)(4-(4-ethylpiperazin-l-yl)-2-n rophenyl)carbamate
NaH (0.0314g, 0.786 mmol) was added to a stirred solution of l-(3,5-dimethoxyphenyl)-3-(6- ((4-(4-ethylpiperazin- 1 -yl)-2-nitrophenyl)amino)pyrimidin-4-yl)-3-methyl- 1 -teri-butyl carbonate urea (0.25g, 0.393 mmol) in anhydrous DMF (4mL) under an argon atmosphere at 0 °C. The resulting mixture was stirred for 15 min. Then Di-¾rt-butyl dicarbonate (0.12 mL, 0.589 mmol) was added at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 18h. The reaction mixture was diluted with ethyl acetate and cold water. The aqueous layer was separated and extracted with ethyl acetate (3x20mL). The combined organic phase was washed with brine, dried over Na2S0 , filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography (DCM:MeOH, 97:3) to afford the title compound (0.280g, 96% of yield) as a brown solid. 1HNMR (CDCI3, 300MHz): δ 8.44 (s, IH), 8.30 (s, 1H)5 7.60 (d, IH), 7.14-7.03 (m, 2H), 6.48 (d, IH), 6.44-6.21 (m, 2H), 3.75 (s, 6H), 3.49 (s, 3H), 3.49 (t, 4H), 2.62 (t, 4H), 2.49 (q, 2H), 1 ,38 (d, 18H), 1.14 (t, 3H); MS (ESI):737.5 [M+H]+.
Figure imgf000087_0001
e. tert-butyl (2-amino-4~(4~ethylpiperazin-l-yl)phenyl)(6~(3-(3,5-dimethoxypfa
tert-butyl carbonate ureido)pyrimidin-4~yl)carbamate
A mixture of tert-butyl (6-(3-(3,5-dimethoxyphenyl)-l-methyl-3-/ert-butyl carbonate
ureido)pyrimidin-4-yl)(4-(4-ethylpiperazin-l-yl)-2-nitrophenyl)carbamate (0.280g, 0.380 mmol) and Raney nickel (0.05g) in mixture of MeOH and THF (1 : 1) (lOmL) was stirred for 36h at room temperature under a hydrogen atmosphere (balloon). The reaction mixture was filtered through Celite pad. The filtrate was concentrated to afford the title compound (0.14g, 52% of yield). MS (ESI): 707.7 [M+H]+.
Figure imgf000087_0002
/. tert-butyl (2-acrylamido-4-(4-ethylpiperazin-l-yl)phenyl)(6-(3-(3tS-dimethoxyp methyl-3-tert-butyl carbonate ureido)pyrimidin-4-yl)carbamate
To a stirred solution of fert-butyl (2-amino-4-(4-ethylpiperazin-l-yl)phenyl)(6-(3-(3,5- dimethoxyphenyl)-l-methyl-3-ier/-butyl carbonate ureido)pyrimidin-4-yl)carbamate (Q.14g, 0.183 ramol) in anhydrous DCM (5 mL) was added TEA (0,08mL, 0.594 mmol) under argon atmosphere at 0 °C. The resulting mixture was stirred for 15 m n. and slowly added the acryloyl chloride (0.03 mL, 0.396 mmol) at 0 °C. The resulting reaction mixture was allowed to warm to room temperature stirred for 3h. The reaction mixture was diluted with DCM and water. The aqueous layer was separated and extracted with DCM (3 20mL). The organic phase was washed with brine, dried over Na2SC>4, filtered, and concentrated. The residue was purified by silica gel column chromatography (DCM:MeOH, 97:3) to afford the title compound (0.070g, 46% of yield) as a brown solid. MS (ESI): 761.4 [M+H]+.
Figure imgf000088_0001
g. N-(2-((6^(3-(3,5-dimethoxyphenyl)-l-methylureido)pyrimidin-4-yl)a
ethylpiperazin~l-yl)phenyl)acrylamide
TFA (0.35 mL, 5 vol) was slowly added to a stirred solution of tert~bxxty\ (2-acrylamido-4-(4- ethylpiperazin- 1 -yl)phenyl)(6-(3-(3 ,5-dimethoxyphenyl)- 1 -methyi-3 -tert-butyl carbonate ureido)pyrimidin-4-yl)carbamate (0.070g, 0.124 mmol) in dry DCM (2 mL) under argon atmosphere at 0 °C. The resulting reaction mixture was allowed, to warm to room temperature and stirred for 24h. Reaction progress was monitored by LCMS, after completion of the reaction, excess solvents were removed under reduced pressure. The resulting residue was diluted with
DCM and a saturated aqueous solution of NaHC03. The aqueous layer was separated and extracted with DCM (3x1 OmL). The organic phase was washed with brine, dried on Na2S04, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography (DCM/MeOH, 97:3) to afford 45mg of desired product with HPLC purity 80%, which was purified by preparative HPLC (Conditions: Column: XBRIDGE-C18 (19.0x150mm,
5micron); (Mobile Phase: A; 0.1% TFA in Water, B; ACN) to afford the title compound (19 mg,
37% of yield) as a off-white solid. JHNMR (CDC13, 300MHz): δ 12.85 (s, 1H), 8.39 (s, 1H), 7.72
(s, 1H), 7.55 (s, 1H), 7.19 (d, 1H), 6.79 (s, 2H), 6.75 (d, 1H), 6.64 (s, 1H), 6.39 (d, 1H), 6.22-
6.13 (m, 2H), 5.80-5.73 (m, 2H), 3.78 (s, 6H), 3.28 (t, 4H), 3.21 (s, 3H), 2.60 (t, 4H), 2.47 (q, 2H), 1.12 (t, 3H) MS (ESI): 561.60 [M+H]+; HPLC: 96.04%, rt: 6.40 min.
Example - 14Θ
Figure imgf000089_0001
N-(2-((6-(3-(2,6-dichlorophenyl)-l-methylureido)pyriniidiri-4-yl)amiao)-5-(4- ethylpiperazin-l-yl)phenyl)acrylamide
The compound was synthesized following the approach outlined in Procedure 2H (Example 139), substituting 2,6-dichloroaniline in step (a) and (2-(chloromethoxy)ethyl)trimethylsilane in step (b) to afford the title compound (18 mg, 3.2% of yield) as a off-white solid. 1HNMR (CDCI3, 300MHz): δ 12.58 (s, IH), 8.39 (s, IH), 7.75 (s, IH), 7.61 (s, IH), 7.37 (d, 2H), 7.22 (d, IH), 7.14 (t, IH), 6.77 (dd, IH), 6.67 (s, IH), 6.42 (d, IH), 6.25-6.16 (m, IH), 5.85 (s, IH), 5.78 (d, IH), 3.35-3.24 (m, 7H), 2.62 (t, 4H), 2.49 (q, 2H), 1.14 (t, 3H); MS (ESI): 569.10 [M]+; HPLC: 96.98%, rt: 3.49 min.
Example - 141
Figure imgf000089_0002
N-(2-((6-(3-(2-chloro-3,5-dimethoxyphenyl)-l-methylureido)pyrim^
ethylpiperazin~l~yl)phenyl)acrylamide
The compound was synthesized following the approach outlined in Procedure 2H (Example 139), substituting 2-chloro-3,5-dimethoxyaniline (procedure shown below) in step (a) to afford the title compound (20 mg, 6.8 % of yield) as a off-white solid. IH-NMR (CDC13, 400MHz): δ 13.46 (s, IH), 8.43 (s, IH), 7.79-7.71 (m, 2H), 7.56 (s, IH), 7.18 (d, IH), 6.74 (dd, IH), 6.61 (s, IH), 6.38 (d, IH), 6.26 (d5 IH), 6.21-6.15 (m, IH), 5.79-5.74 (m, 2H), 3.86 (s, 3H), 3.81 (s, 3H), 3.28 (t, 4H), 3.23 (s, 3H), 2.59 (t, 4H), 2.47 (q, 2H), 1.12 (t, 3H); MS (ESI): 595.15 [M]+;
HPLC: 98.14%, rt: 3.49 min.
Preparation of 2-chloro~3,5-dimethoxyaniline
Figure imgf000090_0001
15h
a. N-(3,5-dimethoxyphenyl)acetamide
Acetic anhydride (6.5 mL)was added slowly to a stirred solution of 3,5-dimethoxy aniline (lOg, 65.359 mmol) in toluene (50 mL) under argon atmosphere at room temperature and resulting reaction mixture was stirred for 15h. After completion of the reaction, reaction was diluted with Hexane and resulted precipitate was collected by filtration and dried under vacuum to afford the title compound (12.5g, 98% of yield) as a off-white solid. !HNMR (CDC13, 300MHz): δ 7.38 (s, IH), 6.75 (d, 2H), 6.23 s, 1H), 3.76 (s, 6H)} 2.15 (s, 3H); MS (ESI):196.1 [M+H]+.
Figure imgf000090_0002
b, N-(2~chloro-3,5-dimethoxyphenyl) cetamide
To a stirred solution of N-(3,5-dimethoxy-phenyl)-acetamide (5g, 25.64 mmol) in acetic acid (17 mL) was added 32% aqueous hydrochloric acid solution (14 mL), followed by a solution of sodium chlorate (1.16g, 11 mmol) in water (1.5 mL) at 0 °C. The resulting reaction mixture was stirred for 30 min. at 0 °C. Thereafter reaction mixture was poured into ice water and made it basic with 2C03 powder. The precipitate was filtered off and washed with water. The residue was purified by silica gel column chromatography (Hexane/EtOAc, 88:12) to afford the title compound (1.8g, 31% of yield) as a white solid. 'HNMR (DMSO-£¾, 300MHz): δ 9.36 (s, 1H), 7.03 (d, 1H), 6.53 (d, 1H), 3.84 (s, 3H), 3.74 (s, 3H), 2.08 (s, 3H); MS (ESI): 230.2 [M+H]+.
Figure imgf000090_0003
c. 2-chloro~3f5-dimethoxy aniline
Potassium hydroxide (2.19g} 39.18 mmol) was added to a solution of N-(2-chloro-3}5- dimethoxyphenyl)acetamide (1.8g, 7.837 mmol) in EtOH (lOOmL) and water (lOmL) and the reaction mixture heated to reflux for 12h. Excess EtOH was removed under reduced pressure to obtain a residue. The residue was then partitioned between water and diethyl ether. The organic layer was separated, dried over sodium sulfate, filtered and concentrated under vacuum to afford the title compound (1.2g, 82% of yield) as a white solid. 1HNMR (CDC13, 300MHz): δ 5.97 (s, 2H)S 4.08 (brs, 2H), 3,84 (s, 3H), 3.75 (s, 3H); MS (ESI): 188.1 [M+H .
Procedure 21: Example
Figure imgf000091_0001
tphos (20%)
50°C, Q Cs2C03 (2.5 eqv.), Toluene
100 °C
yield: 70%
Figure imgf000091_0002
N-(2-((6-(3-(2)6-dichloro-3,5-dimethoxyphenyl)-l-methylureido)pyrimidin~4-yl)araino)-5- (2-methoxyethoxy)phenyl)acrylamide
Figure imgf000092_0001
a. 4-(2-methoxyethoxy)-2-nitroaniline
Powdered and dried Potassium carbonate (3.58g, 0.025mol) was added to a solution of 4-amino- 3-nitropheiioI (2g, 0.012moi) in DMF (20mL) at 0°C under nitrogen atmosphere. To this, 1- bromo-2-methoxyethane (1.34mL, Q.014mol) was added dropwise and the resultant reaction mixture was refiuxed for overnight. Then, the reaction mixture was filtered through celite. The fiiterate was concentrated and diluted with ethyl acetate and water. The aqueous layer was separated and extracted with ethyl acetate (3 30mL). The organic phase was washed with brine, dried on Na2S04, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography to obtain the title compound (0.6g, yield: 24%) as a solid. MS (ESI):
213.15 [M+H .
Figure imgf000092_0002
b. ]rf-(4-(2-methoxyethoxy)-2-nitrophenyl)-]^-methylpyrM
4-(2-methoxyethoxy)-2-nitroaniline (0.6g, 2.830mmol) and 6-chloro-N-methylpyrimidin-4- amine (0.404 g, 2.830 mmol) were taken in lOmL of dry toluene in a seal tube under Argon atmosphere at room temperature. The Argon gas purging was continued for additional 5-10 min. Then Cs2C03 (2.3 g, 7.075 mmol, 2.5eq) and Xantphos (0.490g, 0.849 mmol) were added and the resulting reaction mixture was purged with argon gas for 5 min, followed by Pd2(dba)3 (0.518 g, 0.566 mmol) was added. The argon gas purging was continued for additional 5 min before sealing the reaction vial. Then the reaction mixture was heated at 100 °C for 7h. After completion of the reaction by TLC (DCM:MeOH, 98:2), reaction mass was filtered through celite and the filtrate was evaporated under vacuum to get a crude residue. The crude residue was purified by column chromatography on silica gel to afford the title compound (0.93g, yield:
70%); MS (ESI): 320.3 [M+H]+.
Figure imgf000093_0001
c. 3-(2,6-dichloro~3,5-dimethoxyphenyl)-l~(6-((4-(2-methoxyethoxy
pyrimidin-4-yl)-l-methylurea
To a stirred solution of 2,6-dichloro-3,5-dimetrioxyaniIine (500mg, 2.252 mmol) in dioxane (10 mL) was added 20% phosgene in toluene (4.4mL, 9.0mmol) under argon atmosphere at 0 °C. The resulting mixture was stirred for 6h at 90 °C, and then allowed to cool to room temperature. The solvents were removed and the residue was dissolved in toluene (lOmL). To this, was added N4-(4-(2-methoxyethoxy)-2-nitrophenyl)-N6-methylpyrimidine-4,6-diamine (0,718g, 8.358 mmol, l.Oeq). The resultant reaction mixture was then refluxed for 6h. After completion of the reaction by TLC (Hexanes:EtOAc, 7:3), reaction mixture was cooled to room temperature, concentrated under vacuum to obtain a crude reaction mixture. The solid precipitated on addition of ethyl acetate to the crude reaction mixture was filtered, washed with ether and pentane to afford the title compound (0.285, yield: 32%) as a white solid. MS (ESI): 567.0 [M+H]+.
Figure imgf000093_0002
d. tert-butyl (6-(3~(2,6-dichlow~3,5-dimethoxyphenyl)-l-methyl-3-tert-butyl carbonate ureido)pyrimidin-4-yl)(4-(2-methoxyethoxy)-2-nitrophenyl)carbamate
DMAP(0.025g, 0.2 mmol), and Di-terf-butyl dicarbonate (0.438g, 2.009 mmol) was added to a stirred solution of 3 -(2 ,6-dicbioro-3 , 5 -dimethoxyphenyi)- 1 -(6 -((4-(2-methoxyethoxy)-2 - nitrophenyl)amino) pyrimidin-4-yl)-l-methylurea (0.285g, 0.502 mmol) in anhydrous THF (lOmL) under an argon atmosphere at 0 °C. The resulting mixture was refluxed for 3-4h. After completion of the reaction by TLC (Hexanes:EtOAc, 1:1), reaction mixture was cooled to room temperature, concentrated under vacuum to obtain a crude residue. The residue was purified by silica gel column chromatography to afford the title compound (0.35g, yield: 88%) as an off- white solid. MS (ESI): 767.1 [M+H]+.
Figure imgf000094_0001
e. tert-butyl (2-amino-4-(2-methoxyethoxy)phenyl)(6-(3~(2,6 dichloro-3,5-dim
methyl-3-tert-butyl carbonate ureido)pyri idin-4-yl)carbamate
Raney nickel (0.05g) was added to a solution of tert-butyl (6-(3-(2,6-dichloro-3,5- dimethoxyphenyl)-l -methyl-3-iert-butyl carbonate ureido)pyrimidin-4-yl)(4-(2-methoxyethoxy)- 2-nitrophenyl)carbamate (0.350g, 0.456mmol) in mixture of THF and MeOH (lOmL) and the resultant reaction mixture was stirred for 14h at room temperature under a hydrogen atmosphere (balloon). The reaction mixture was filtered through Celite pad. The filtrate was concentrated to afford a crude residue. The residue was purified by silica gel column chromatography
(MeOH:DCM, 5:95) to afford the title eomopund (0.26g, yield: 77% ) as solid. MS (ESI): 737.2 [M+Hf.
Figure imgf000094_0002
tert-butyl (2~acrylamido»4"(2-methoxyethoxy)phenyl)(6-(3-(2,6-dichloro~3,5- dimethoxyphenyl)-l-methyl-3-tert~butyl carbonate ureido)pyrimidin-4-yl)carbamate
To a stirred solution of tert-butyl (2-amino-4-(2-methoxyethoxy)phenyl)(6-(3-(2s6-dichloro-3J5- dimethoxyphenyl)-l-rnethyI-3 -tert-butyl carbonate ureido)pyrimidin-4-yl)carbamate (0.26g, 0.352 mmol) in anhydrous DCM (6 mL) was added TEA (0.09mL, 0.704 mmol) under argon atmosphere at 0 °C. The resulting mixture was stirred for 15 min. and slowly added the acryloyl chloride (0.04 mL, 0.528 mmol) at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 2h. The reaction mixture was diluted with DCM and water. The aqueous layer was separated and extracted with DCM (3x30mL). The organic phase was washed with brine, dried over Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography to afford the title compound (0.200g, yield: 72%) as a solid. MS (ESI):
Figure imgf000094_0003
Figure imgf000095_0001
g. N-(2-((6-(3-(2,6'dichloro-3,5-dimethoxyphenyl)-l-methylureido)py
methoxyethoxy)phenyl)acrylamide
TFA (0.38 mL, 5.05mmol) was slowly added to a stirred solution of fert-butyl (2-acrylamido-4- (2~memoxyethoxy)phenyl)(6-(3-(256-dichloro-3,5-d^
carbonate ureido)pyrimidin-4-yl)carbamate (0.2g, 0.252 mmol) in dry DCM (2 mL) under argon atmosphere at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 12h. Reaction progress was monitored by LCMS. After completion of the reaction, excess solvents were removed under reduced pressure. The crude solid was washed with ether to afford the title compound (86 mg, yield: 58%) as a white solid. lHNMR (DMSO- d6, 400MHz): 312.01 (s, 1H), 9.60 (s, 1H), 8.85 (s, 1H), 8.33 (s, 1H); 7.42 (s, 1H), 7.34 (d, 1H), 6.89 (s,lH), 6.79 (d, 1H), 6.51 (dd, 1H), 6.23 (d, 2H), 5.72(d, ΪΗ), 4.08 (m,2H), 3.93 (s, 6H), 3.66 (m, 2H), 3.31(3,3Η), 3.23(s,3H). MS (ESI): 591.3 [M+H]+; HPLC: 96.04%, rt: 3.72 min.
Example: 144
Figure imgf000095_0002
N-(2-((6-(3-(2-chloro-6-fluoro-3,5-dimethoxyphenyl)-l-methyIureido) pyrimidin-4- yl)amino)-5-(4-ethylpiperazitt-l-yl)phenyl)acryIamide
The title compound was synthesized following the approach outlined in Procedure 21 (Example 142), substituting N4-(4-(4-ethylpiperazin- 1 -yl)-2-nitrophenyl)-N6-methylpyrimidine-4,6- diamine (Procedure 2C, Example 108, Steps a- d) and 2-chloro-6-fiuoro-3,5-dimethoxyaniline (Procedure shown below) in step (c) to afford the pen-ultimate title compound (0.46 mg, yield: 2.6 % over five steps) as an off-white solid. MS (ESI): 813.1 [M+H]+. The free amine (46rng, 0.075mmol) was dissolved in ethyl acetate: DCM: MeOH mixture and treated with phosphoric acid (7mg, 0.075mmol). After stirring for lh, the solid precipitated was filtered and washed with ether and pentane to afford the title compound (34.7 mg, yield: 65%) as an off-white solid.
'HNMR (DMSO-d6, 300MHz): δ 12.15 (s, IH), 9.60 (s, IH), 8.76 (s, IH), 8.32 (s, IH), 7.29 (d, 2H), 6.9 (d, 2H), 6.81 (dd, IH), 6.51-6.45 (m, IH), 6.25-6.20 (m, 2H), 5.72 (d, IH), 3.91 (s, 3H), 3.89 (s, 3H), 3.23 (t, 4H), 3.14 (t, 4H), 2.54-2.50 (m, 5H), 1.05 (t, 3H); MS (ESI): 613.2 (M- ¾P04]+; HPLC: 98.6%, rt: 6.13 min.
Preparation of2-ch loro-6-fluoro-3, 5-dimethoxyaniline
Figure imgf000096_0001
a. methyl 2-fluoro-3f5~dimethoxybenzoate
A suspension of Selectfluor (48.9g, 0,15mol) in acetonitrile (1.1L) was added to a solution of methyi-3,5-dimethoxy benzoate (20g, O.lOmol) in acetonitrile at 0°C under nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature and stirred overnight. The reaction mixture was concentrated under vaccum, diluted with saturated sodium carbonate solution and ethyl acetate. The aqueous layer was separated and extracted with ethyl acetate (3x200mL). The organic phase was washed with brine, dried over Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography (gradient hexane/ether 30:1 to 4:1) to afford the title compound (4g, yield: 16.9%).
Figure imgf000096_0002
b. methyl 2-chloro-6-fluoro-3,5-dimethoxybenzoate
S02C12 (2.20g, 0.016mol) was added dropwise to a solution of methyI-2-fluoro-3,5-dimethoxy benzoate (3.5g, 0.016 mol) in acetonitrile (40mL) at 0°C under nitrogen atmosphere. The resulting reaction mixture was warmed to room temperature slowly and stirred for lh. The reaction mixture was quenched with saturated sodium bicarbonate solution, and extracted with ethyl acetate (3x30mL). The organic phase was washed with brine, dried over Na2S0 , filtered, and concentrated. The residue was purified by silica gel column chromatography using eluent (gradient hexane/ether (20:1) to hexane/ether (5:1) to afford the title compound (2.7g, yield: 67%) as a solid.
Figure imgf000097_0001
c. 2-chloro-6-fluoro-3,5-dimethoxybenzoic acid
A suspension of methyl 2-chloro-6-fiuoro-3,5-dimethoxybenzoate (2.7g, O.OlOmol) and sodium hydroxide (1.088g, 0.0272) in anhydrous ethanol (30mL) was refluxed for 24h. The resulting reaction mixture was cooled to room temperature and concentrated under vacuum to get a crude residue. The crude residue was dissolved in water and extracted with ether (3X30mL). The aqueous layer was acidified with conc.HCl and the precipitated solid was filtered, washed with cold water and dried in vacuo to afford the title compound (1.8g, yield: 71%) as a solid.
Figure imgf000097_0002
(L 2-chloro-6~fluoro-3,5-dimethoxyaniline
A suspension of 2-chloro-6-fluoro-3,5-dimethoxybenzoic acid (1.8g, 0.0077 mol) and triethyl amine (0.934g, 0.0092mol) in tert-BuOH(50mL) was stirred for 5 rnin, To the resulting reaction mixture, Diphenyl phosphoryl azide (2.53g, 0.0092mol) was added and heated up to 82 °C and kept at this temperature for overnight . The reaction mixture was then concentrated in vacuo to obtain a crude residue. The crude residue was dissolved in dichloromethane (20mL) and cooled to 0°C. TFA (4mL) was added to the reaction mixture and the resultant reaction mixture was then stirred at room temperature for 2h. The solvents were removed under vaccum and the crude residue was diluted with ethyl acetate and saturated sodium carbonate solution. The aqueous layer was separated and extracted with ethyl acetate (3x30mL). The organic phase was washed with brine, dried over Na2S04, filtered, and concentrated. The residue was purified by silica gel column chromatography using the eluent (gradient hexane to hexane-ether (65:35)) to afford the title compound (0.95g, yield: 60%) as solid. MS (ESI): 205.7 [M+H]+. Procedure 2J - Example 145
Figure imgf000098_0001
N-(2-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methylureido) pyrimidin-4-yl)amino)-5~ (hydroxymethyl)phen l)acryl mide
Figure imgf000098_0002
a. l-(6-chloropyrimidin-4-yl)-3-(2f6-dichloro-3,5~dimethoxyphenyl)-l^
car on ate urea
DMAP (0.080g, 0.655 mmol) and Di-tirt-butyl dicarbonate (2.9mL, 12.6 mmol) was added to a stirred solution of l-(6-chloropyrimidin-4-yl)-3-(256-dichloro-3,5-dimethoxyphenyl)-l- methylurea (Procedure 2E, step b; 2.6g; 6.632mmoi) in anhydrous THF (20mL) under an argon atmosphere at 0 °C. The resulting mixture was then refluxed for 2h. After completion of the reaction by TLC (EtOAc:Hexane 3:7), reaction mixture was cooled to room temperature, concentrated under vacuum to obtain a crude residue. The residue was purified by silica gel column chromatography to afford the title compound (2.5g, yield: 69.4%) as a solid. 1H-NMR (CDCI3, 300MHz): 5 8.73 (d, 1H), 7.95 (d, 1H)} 6.61 (s, 1H), 3.95 (s, 6H), 3.63 (s, 3H), 1.35 (s, 9H).
Figure imgf000099_0001
b. methyl 4-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl~3-tert-butyl carbonate ureido)pyrimidin~4~yl)amino)-3-nitrobenzoate
Methyl 4-amino-3-nitrobenzoate (0.956g, 0.004mol) and l-(6-chloropyrimidin-4-yl)-3-(2,6- dichloro-3,5-dimethoxyphenyl)-l-methyl-3-/ert-butyl caronate urea (2g, 0.004mol) were taken in iOmL of dry toluene in a seal tube at room temperature and Argon gas was purged for 5-10 min. Then Cs2C03 (3.25g, O.Olmol) and Xantphos (0.46g, 0,0008 mol) were added and the resulting reaction mixture was purged with argon gas for 5 min, followed by Pd2(dba)3 (0.36g, 0.0004mol) was added. The argon gas purging was continued for additional 5 min before sealing the reaction vial. Then the reaction mixture was heated at 100 °C for 12h. After completion of the reaction by TLC, reaction mass was filtered through celite and the fllterate was evaporated under vacuum to get a crude residue. The crude residue was purified by column chromatography on silica gel to afford the title compound (2.10g, yield: 79%); 1H-NMR (CDC13, 300MHz): δ 10.3 (s, 1H), 9.00- 8.95 (m, 2H), 8.75 (s, 1H), 8.22 (dd, 1H), 7.63 (s, 1H), 6.61 (s, 1H), 3.95 (s, 6H), 3.94 (s, 3H), 3.67 (s, 3H), 1.39 (s, 9H).
Figure imgf000099_0002
c. methyl 4-((tert^utoxycarbonyl)(6-(3-(2,6-dw' hloro-3,5-dimetho
butyl ureido)pyrimidin-4-yl)amino)~3~nitrobenzoate
DMAP (0.039g, 0.322 mmol) and Di-¾rt-butyl dicarbonate (I.48mL, 6.45mmol) was added to a stirred solution of methyl 4-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl-3-tert-butyl carbonate ureido)pyrimidin-4-yl)amino)-3-mtrobenzoate (2.1g, 3.22 mmol) in anhydrous THF
(5mL) under an argon atmosphere at 0 °C. The resulting mixture was refluxed for 12h. After completion of the reaction by TLC (EtOAc:Hexane 40:60), reaction mixture was cooled to room temperature, concentrated under vacuum to obtain a crude residue. The residue was purified by silica gel column chromatography to afford the title compound (2.1g, yield: 87%) as a solid. MS (ESI): 751.0 [M .
Figure imgf000100_0001
d. tert-butyl (6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)~l-met yl-3-tert-butyl carbonate ureido)pyrimidin-4-yl)(4-(hydroxymethyl)-2'nitrophenyl)carbamate
Lithium borohydride (0.049g, 2.26mmol) was added to a solution of methyl A-{{tert- butoxycarbonyl)(6-(3 -(2,6-dichloro-3 ,5 -dimethoxyphenyl)- 1 -methyl-3~tert-butyl
ureido)pyrimidin-4-yl)amino)-3-nitrobenzoate (1.7g, 2.26 mmol) in anhydrous THF (18mL) under argon atmosphere at 0 °C, The resulting reaction mixture was then allowed to warm to room temperature by 2.5h. The reaction mixture was then quenched with ice- water and diluted with ethylacetate. The aqueous layer was separated and extracted with ethyl acetate (3x20mL). The organic phase was washed with brine, dried over Na2S(¾, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography to afford the title compound
Figure imgf000100_0002
e. tert-butyl (4-(((tert-butyldimethyltilyl)oxy)methyl)-2-nitro
dimethoxyphenyl)-l-methyl-3-tert~butyl carbonate ureido)pyrimidin~4-yl) carbamate
Imidazole (0.07 l g, 1.051 mmol) and TBDMS-C1 (0.118g, 0.787 mmol) were added to a stirred solution of ier/-butyl (6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)"l-methyl-3-/ert-butyl carbonate ureido)pyrimidin-4-yl)(4-(hydroxymethyl)-2-nitrophenyl)carbamate (0.380g, 0.525 mmol) in anhydrous dichloromethane (5mL) under an argon atmosphere at 0 °C. The resultant reaction mixture was stirred for 4h at room temperature. The reaction mixture was then diluted with DCM and water. The aqueous layer was separated and extracted with DCM (3 l0mL). The organic phase was washed with brine, dried over N 2S04, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography to afford the title compound (0.330g, yield: 75%) as a solid. MS (ESI): 837.4 [M]+.
Figure imgf000101_0001
tert-butyl (2~amino-4-(((tert-butyldimethykilyl)oxy)methyl)phen^
dimethoxyphenyl)-l~methyl-3-tert-butyl carbonate ureido)pyrimidin-4~y I) carbamate
Raney nickel (0.06g) was added to a solution of tert-butyl (4-(((/ert-butyldimethylsilyl) oxy)methyI)-2-nitrophenyl)(6-(3-(2,6-dichloro-3,5-dimemoxyphenyl)-l-methyl-3- carbonate ureido)pyriraidin-4-yl)carbaraate (0.3g, 0.358 mmol) in MeOH (5mL) and the resultant reaction mixture was stirred for 12h at room temperature under a hydrogen atmosphere (balloon). The reaction mixture was filtered through Celite pad. The filtrate was concentrated to afford a crude residue which was then purified by column chromatography to afford the title compound (0.180g, yield: 64%) as a solid. MS (ESI): 807.2 [M]+.
Figure imgf000101_0002
g. tert-butyl (2-acrylamido~4-(((tert-butyldimeikylsilyl}oxy)methyl)phenyl)(6-(3
3, 5-dimethoxyphenyl)~l-methyl-3-tert-butyl carbonate ureido)pyrimidin-4-yl)carbamate
To a stirred solution of ierf-butyl (2-amino-4-(((tert-butyldimethylsilyl)oxy)methyl)phenyl)(6-(3- (2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl-3-ierf-butyl carbonate ureido)pyrimidin-4- yl)carbamate (0.180g, 0.223 mmol) in anhydrous DCM (5 mL) was added TEA (0.08mL, 0.557 mmol) under argon atmosphere at 0 °C. The resulting mixture was stirred for 15 min. and slowly added the acryloyl chloride (0.03 mL, 0.334 mmol) at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 4h. The reaction mixture was then concentrated and the residue was purified by silica gel column chromatography to afford desired the title compound (0.130& yield: 68%) as a solid. MS (ESI): 861.3 [M .
Figure imgf000102_0001
h. N-(2-((6-(3-(2,6-dichloro-3,5-dimeihoxyphenyl)-l-methylureido)pyrimffl
(hydroxymethyl)phenyl)acrylamide
TFA (1.0 mL) was slowly added to a stirred solution of ierf -butyl (2-acrylamido-4-(((tert- butyldimeth.ylsilyl)oxy)metb.yl)plienyl)(6-(3 -(2,6-dichloro-3 ,5-dimethoxyphenyl)- 1 -methyl-3 - terr-butyl carbonate ureido)pyrimidin-4-yl)carbamate (0.130g, 0.150mmol) in dry DCM (3 mL) under argon atmosphere at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 13h. Reaction progress was monitored by LCMS, after completion of the reaction, excess solvents were removed under reduced pressure. The resulting residue was diluted with DCM and a saturated aqueous solution of NaHC03. The aqueous layer was separated and extracted with DCM (3 1 OmL). The organic phase was washed with brine, dried over Na2S04, filtered and concentrated under vacuum. The residue was purified by silica gel column chromatography to afford 95mg of desired product with HPLC purity 84%, which was then purified by preparative HPLC (Conditions: Column: X bridge CI 8 ( 19mm x 150mm, 5μπι); (Mobile Phase: A; 0.01% TFA in Water, B; ACN) to afford the title compound (16mg, yield: 16%) as a white solid. 1H-NMR (DMSO-d6, 400MHz): δ 12.08 (s, IH), 9.70 (s, IH), 8,95 (s, IH), 8.37 (s, IH), 7.64 (s, IH), 7.49 (d, IH), 7.15 (s, IH), 6.90 (s, IH), 6.53-7.46 (q, IH), 6.32 (s, IH), 6.25 (d, IH), 5.74 (d, 1H)S 5.42 (s, IH), 5.35 (s, IH), 4.49 (s, 2H), 3.94 (s, 6H), 3.25 (s, 3H); MS (ESI): 547.0 [M+H]+; HPLC: 97.4%, rt: 3.83 min.
Example 147
Figure imgf000102_0002
N-(2~((6~(3-(2,6~dichloro-3,S-dimethoxyphenyl)-l~methylureido) pyrimidin-4-yl)amino)-5- (methoxymethyfyphenyl) acrylamide
The title compound was synthesized following the approach outlined in Procedure 21 (Example 142), substituting N4-(4-(methoxymethyl)-2-nitrophenyl)-N6-methylpyrimidine-4,6-diamine (procedure shown below) in step (c) to afford the title compound (5.0 mg, yield: 1.8%) as an off- white solid. IH-NMR (CDC13, 400MHz): δ 12.30 (s, IH), 8.41 (s3 IH), 7.86 (s, IH), 7.81 (s, IH), 7.52 (d, IH), 7.49 (s, IH), 730 (d, IH), 6.54 (s, IH), 6.46 (d, IH), 6.26 (dd, IH), 6.07 (s, IH), 5.83 (d, IH), 5.18 (s, 2H), 3.93 (s3 6H), 3.82 (s, 3H), 3.34 (s, 3H); MS (ESI): 561.1 [M+H ; HPLC: 95.76%, rt: 4.36 min.
Preparaton ofJ^-(4-(methoxymethyl)-2-nitrophenyl)~lS^-methylpyrM
Figure imgf000103_0001
a. methyl 4-amino-3-nitrobenzoate
Thionyl chloride (19.4g, 164.85 mmol) was added to a solution of 4-amino-3-nitrobenzoic acid (20g, 109.89 mmol) in methanol (200mL) at 0 °C. The resulting mixture was then refiuxed for 12h. The reaction mixture was allowed to cool to room temperature. The yellow solid
precipitated was filtered and dried to afford the title compound (22g, yield: 100%) as a solid. IH- NMR (CDC13, 300MHz): δ 8.85 (d, IH), 7.99 (dd, IH), 6.83 (d, IH), 6.40 (s, 2H), 3.90 (s, 3H).
Figure imgf000103_0002
110°C, 16h
b. methyl 4-((6-(methyl mino)pyrimidin-4-yl)amino)-3-nitrobenzoate
6-chloro-N-methylpyrimidin-4-amine, 3 (3g, 20.97mrnol) and methyl 4-amino-3-nitrobenzoate (3.9g, 20.97mmol) were taken in toluene (5mL) in a seal tube under Argon atmosphere at room temperature. The Argon gas was purged for 5-10 min. Then Cs2C03 (17.0g, 52.4mmol) and Xantphos (3.6g, 6.29mmol) were added and the resulting reaction mixture was purged with argon gas for 5 min, followed by Pd2(dba)3 (3.8g, 4.19mmol) was added. The argon gas purging was continued for additional 5 min before sealing the reaction vial. Then the reaction mixture was heated at 110 °C for 16h. After completion of the reaction by TLC, the reaction mixture was cooled to rt, filtered through celite bed and the filtrate was evaporated under vacuum to get a crude residue. The crude residue was purified by column chromatography on silica gel to afford the title compound (3.2g, yield: 51%) as a solid; MS (ESI): 304.2 [M+H]+.
Figure imgf000104_0001
c. methyl 4~((tert-butoxycarbonyl)(6-((tert-butoxycarbonyl)(methyl) mino)pyrimidin^ yl) amin o)-3-nitrobenzoate
DMAP (0.497 g, 4.078 mmol) and Di-tert-butyl dicarbonate (8.89g, 40.78 mmol) were added to a stirred solution of intermediate-4 (3.1g, 10.197mmol) in anhydrous THF (35mL) under an argon atmosphere at room temperature. The resulting mixture was refluxed for 2-3h. After completion of the reaction by TLC, reaction mixture was cooled to room temperature, concentrated under vacuum to obtain a crude residue. The residue was purified by silica gel column chromatography to afford the title compound (1.9g, yield: 37%) as a solid. MS (ESI): 504.0 [M+H]+.
Figure imgf000104_0002
d. tert-butyl (6-((tert-butoxycarbonyl)(4-(hydroxymethyl)-2-nitrophenyl)am
yl) (methyl) carbamate
Lithium borohydride (0.157g, 7.14 mmol) was added to a solution of methyl 4-((teri- butoxycarbonyl)(6-((ie^butoxycarbonyl)(methyl)amino)pyrimidin-4-yl)amino)-3-nitrobenzoate (1.8g, 3.57 rrirnol) in THF (20mL) under an argon atmosphere at 0 °C. The resulting mixture was then allowed to warm to rt and stirred for 12h. The reaction mixture was then quenched with ice- water and diluted with ethyl acetate. The aqueous layer was separated and extracted with ethyl acetate (3x40mL). The organic phase was washed with brine, dried over Na2S04, filtered, and concentrated to get a crude residue. The residue was purified by silica gel column
chromatography to afford desired intermediate-6 (1.2g, 75%) as a solid. MS (ESI): 476.1
[M+H]÷.
Figure imgf000104_0003
e, tert-butyl (6-((tert-butoxycarbonyl)(4-(methoxymethyl)-2-nitrophenyl)amino)pyrm yl) ( methyfycarbamate
To a solution of tert-butyl (6-((te^butoxycarbonyl)(4-0iydroxymethyl)-2-nitrophenyl)arnino) pyrimidin-4-yl)(methyl)carbamate (1.5g, 3.488ramol) in acetone (20mL) under an argon atmosphere at 0 °C, was added potassium carbonate (0.48g, 6.97mmol) followed by dimethyl sulfate (0.87g, 6.97mmol). The resulting reaction mixture was then refmxed for 24h and then allowed to cool to rt. The reaction mixture was concentrated under vacuum to obtain a crude residue. The residue was purified by silica gel column chromatography to afford the title compound (0.3g, yield: 19%) as a solid. MS (ESI): 490.55 [M+H]+.
Figure imgf000105_0001
lt^-(4-(methoxymethyl)-2-nitrophenyl)-lSf-methylpyH
TFA (5mL) was slowly added to a stirred solution of tert-butyl (6-((tert-butoxycarbonyl)(4- (methoxymethyl)-2-nitrophenyl)amino)pyrimidin-4-yl)(methyl)carbamate (0.85g) in dry DCM (10 mL) under argon atmosphere at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for lh. After completion of the reaction, excess solvents were removed under reduced pressure and washed with ether to afford the title compound (0.7g crude) as a solid. MS (ESI); 290.2 [M+H]+.
Procedure 2K - Example 148
Figure imgf000106_0001
N-(2-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l^
(4-methyIpiperaziae-i-carbonyl)phenyI)acrylamide
Figure imgf000106_0002
a. 4-((tert-butoxycarbonyl)(6-(3-(teH-butoxycarbonyl)-3-(2,6'diM
l-methylureido)pyrimidin-4-yl)amino)-3-nitrobenzoic acid
Lithium hydroxide (0.08g, 2.99mmol) was added to a solution of methyl 4-((tert- butoxycarbonyl)(6-(3 -(ferf-butoxycarbonyl)-3 -{2 , 6-dichloro-3 , 5 -dimethoxyphenyl) - 1 - methylureido)pyrimidin-4-yl)amino)-3-nitrobenzoate (1.5g, 1.997mmol) (Procedure 2J, Step c) in a mixture of THF (lOmL) and water (2mL) at 0°C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 5h. After completion of the reaction by TLC
(EtOAc;Hexane 3:7), the reaction mixture was concentrated under vacuum to get a crude residue. The crude residue was dissolved in water and extracted with ether (3X30mL). The aqueous layer was acidified with 10% citric acid solution and the precipitated solid was filtered, washed with cold water and dried in vacuo to afford the title compound (1.3g, yield: 93%) as a light brown solid. MS (ESI): 737.2 [M+Hf .
Figure imgf000107_0001
b. tert-butyl (2-nitro~4~(4~methylpiperazine-l-carbonyl)phenyl)(6-(3-(2,6-M
dimethoxyphenyl)-3-tert-butoxycarbonyl~l~methylureido)pyrM
DIPEA (0.3mL, 1.62mmol), HATU (0.515g, 1.355mmol) and N-methyl piperazine, 6 (0.09mL, 0.813mmol) were added to a solution of 4-((tert-butoxycarbonyl)(6-(3-(¾rt-butoxycarbonyl)-3- (2,6-dichloro-3,5-dimethoxyphenyl)-l-methylur acid (0.4g, 0.542mmol) in DMF (5mL) at 0°C under argon atmosphere. The resulting reaction mixture was then allowed to warm to rt and stirred for 18h. After completion of the reaction by TLC (MeOH:DCM 1 :19), water was added to the reaction mixture. The precipitated crude solid was filtered, dried and purified by silica gel column chromatography to obtain the title compound (0.27g, yield: 61%) as a solid. MS (ESI): 819.1 [M+H]+.
Figure imgf000107_0002
c. tert-butyl (2-amino-4-(4-methylpiperazine-l~c rbonyl)phenyl)(6-(3-(2, 6-dichloro-3,5- dimet oxyphenyl)-3-tert-butoxycarbonyl-l- ihylureido)pyrlmMin^
Raney nickel (0.06g) was added to a solution of tert-butyl (2-nitro-4-(4-methylpiperazine-l- carbonyl)phenyl)(6-(3-(2}6-dichloro-3,5-dimethoxyphenyl)-3-¾rt-butoxycai-bonyl-l- methylureido)pyrimidin-4-yl)carbamate (0.27g, 0,329mmol) in a mixture of MeOH (4mL) and THF (4mL) and the resultant reaction mixture was stirred for 20h at room temperature under a hydrogen atmosphere. The reaction mixture was filtered through Celite bed. The filtrate was concentrated, to afford crude title compound (0.22g, yield: 84%) as a solid. MS (ESI): 789.4 [M+Hf.
Figure imgf000108_0001
d. tert-butyl (2-acrylamido-4-(4-methylpiperazine-l-carbonyl)ph enyl) (6-(3-(2, 6~dichloro-3, 5- dimethoxyphenyl)-3-tert-butoxycarbonyl-l-methylureido)pyrimi^^
To a stirred solution of tert-butyl (2-amino-4-(4-rnethylpiperazine-l-carbonyl)phenyl)(6-(3-(2,6- dichloro-3,5-dimethoxyphenyl)-3-tert-butoxycarbonyl-l-me
(0,22g, 0.278mmoi) in anhydrous DCM (5 mL) was added TEA (0.08mL, 0.557 mmol) under argon atmosphere at 0 °C. The resulting mixture was stirred for 15 min. and slowly added the acryloyl chloride (0.037g, 0.417 mmol) at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 2h. The reaction mixture was quenched with a saturated sodium bicarbonate solution and diluted with DCM. The aqueous layer was separated and extracted with DCM (3x20mL). The organic phase was washed with brine, dried over Na2S04, filtered, and concentrated to get a crude residue. The residue was purified by silica gel column chromatography to afford the title compound (0.060g, yield: 25%) as a solid. MS (ESI): 843.3 [M+H]+.
Figure imgf000108_0002
e. N-(2-((6-(3-(2,6-dichtoro-3,5-dimethoxyphenyl)-l-methylureido^
methylpiperazine~l-carbonyl)ph enyl) acrylamide
TFA (0.2mL) was slowly added to a stirred solution of tert-butyl (2-acrylamido-4-(4- methylpiperazine-l-carbonyl)phenyl)(6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-3-feri- butylcarbonyl-l-methylureido)pyrimidin-4-yl)carbamate (0.060g, 0.071 lmmol) in dry DCM (2 mL) under argon atmosphere at 0 °C, The resulting reaction mixture was allowed to warm to room temperature and stirred for 18h. Reaction progress was monitored by LCMS, after completion of the reaction, excess solvents were removed under reduced pressure. The resulting residue was diluted with DCM and quenched with a saturated aqueous solution of NaHC03. The aqueous layer was separated and extracted with DCM (3x1 OmL). The organic phase was washed with brine, dried over Na2S04, filtered and concentrated under vacuum to get a crude residue. The residue was purified by silica gel column chromatography to afford 70mg of desired product with HPLC purity 35% which was then purified by preparative HPLC (Conditions: Column: Gemini NX CIS ( 21.2mm x 150mm particle size 5μπι); (Mobile Phase: A; 0.1% Ammonium bicarbonate in Water, B; ACN) to afford the desired compound. The compound was then diluted with dichloromethane and water. The aqueous layer was separated and extracted with DCM (3xl0mL). The organic phase was washed with brine, dried over Na2S04, filtered and
concentrated under vacuum to afford the title compound (0.005g, yield; 11%) as a white solid.
1H-NMR (CD3OD, 400MHz): δ 8.41 (s, IH), 7.79-7.76 (m, 2H), 7.36 (dd, IH), 6.82 (s, IH), 6.47-6.42 (m, 3H), 5.82 (d, IH), 3.96 (s, 6H), 3.81-3.55 (m, 8H), 2.54 (s, 3H), 2.37 (s, 3H), ; MS (ESI): 643.1 [M+Hf ; HPLC: 97.26%, rt: 6.19 min.
Example 149
Figure imgf000109_0001
3-aciylamido-4-((6-(3-(2,6-dichloro-3,5-dim^
yl)amino)-N,N-dimet ylbenzamide
The title compound was synthesized following the approach outlined in Procedure 2K (Example 148), substituting dimethylamine in step (b) to afford the title compound (12.0 mg, yield: 6.1%) as an off-white solid. 1H-NMR (CD3OD, 400MHz): δ 8.38 (s, IH), 7.78- 7.72 (m, 2H), 7.34 (dd, IH), 6.80 (s, IH), 6.46-6.38 (m, 3H), 5.80 (dd, IH), 3.94 (s, 6H), 3.36 (s, 3H), 3.10 (s, 6H); MS (ESI): 587.9 [M+H]+; HPLC: 99.04%, rt: 3.98 min.
Example 150
Figure imgf000109_0002
N-(2-((6-(3-(2,6-dichIoro~3,5-dimethoxyphe^^
(4~ethylpiperazine-l-carbonyl)phenyI)acrylamide
The title compound was synthesized following the approach outlined in Procedure 2 (Example 148), substituting 1-ethylpiperazine in step (b) to afford the title compound (10.0 mg, yield: 9.7%) as an off-white solid. 1H-NM (CD3OD, 400MHz): δ 8.41 (s, 1H), 7.76-7.79 (m, 2H), 7.36 (d, 1H), 6.82 (s, 1H), 6.38-6.47 (m, 3H), 5.82 (d5 1H), 3.96 (s, 6H), 3.72-3.82 (m, 2H), 3.53- 3.65 (m, 3H), 3.39 (s, 4H), 2.49-2.54 (m, 6H), 1.15 (t, 3H) ; MS (ESI): 657.0 |M+H]+; HPLC: 95.98%, rt: 6.25 mm.
Example 151
Figure imgf000110_0001
N-(2-((6-(3-(2,6-dichIoro-3,5-diraethoxyphenyl)-l-methyIureido)pyrimidia-4~yl)amino)-5- (4-isopropylpiperazin-l-yi)phenyl)acrylamide phosphoric acid
The title compound was synthesized following the approach outlined in Procedure 2J (Example 145), substituting 4-(4-isopropylpiperazin-l-yl)-2-nitroaniline (procedure shown below) in step (b) and omitting steps (d) and (e) to afford the free base of the title compound (0.1 g, overall yield: 9.7%) as an off-white solid. MS (ESI): 643.1 [M+H]+. 85% H3P04 was slowly added to a solution ofN-(2-((6-(3-(256-dichloro-3,5-dimethoxyphenyl)-l-methylureido)pyrimidin-4- yl)amino)-5-(4-isopropyIpiperazin-l-yl)phenyl)acrylamide (O.lg) in 95% THF-MeOH (5mL). The resulting reaction mixture was then allowed to stir at room temperature for 30min., evaporated the solvent and triturated with diethyl ether, dried under vacuum to afford the title compound (0.16g) as a off white solid. 'HNMRpMSO-ifc, 400MHz): δ 12.08 (s, 1H), 9.62 (s, 1H), 8.76 (s, 1H), 8.32 (s, 1H), 7.30 (d, 2H), 6.89 (d, 1H), 6.81 (d, 1H), 6.46-6.49 (m, 1H), 6.21- 6.25 (m, 2H), 5.72 (d, 1H), 3.93 (s, 6H), 3.35-3.41 (m, 1H), 3.23 (s, 3H), 3.12-3.17 (m, 4H), 2.67-2.70 (m, 4H), 1.05-1.11 (m, 6H); MS (ESI): 643.3 (M +1); HPLC: 95.41%, rt: 6.48min.
Preparation of 4~(4-isopropylpiperazin-l-yl)-2-nitroaniline
Figure imgf000111_0001
a. tert-butyl 4-isopropylpiperazine-l-carboxylate
Boc piperizine, 1 (10g, 53.76ramol) and acetone (4mL) was taken in a mixture of dry DCM (lOOmL) and acetic acid (3.2mL). Stirred at room temperature for 20 min. Added Na(OAc)3BH (17g? 80.2mmol) and stirring continued at room temperature for 12hr. The reaction mixture was diluted with water, extracted with ethyl acetate (3x25mL). The organic layer was washed with brine, dried over Na2SC>4, filtered and concentrated under vacuum to get crude title compound (13g, crude). MS (ESI): 229.2 [M+H .
b. 1-isopropylpiperazine
TFA (15mL) was slowly added to a stirred solution of fe t-butyl 4-isopropylpiperazine-l- carboxylate (13g crude) in dry DCM (20mL) under argon atmosphere at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 18hr. The reaction mass was concentrated under vacuum and triturated the residue with rc-hexane and diethyl ether, Dried under vacuum to afford the title compound (7g, yield: 97%). MS (ESI): 129.1 [M+H]+.
Figure imgf000111_0003
Dioxane, 100 °C
c. tert-butyl (4-(44sopropylpiperazin l-yl)-2-nitrophenyl)carbamate
1-isopropylpiperazine (1.29g, 5.66mmol) and feri-butyl (4-bromo-2-nitrophenyl)carbamate
(1.5g, 4.71mmol) were taken in a mixture of dry toluene (15mL) and dioxane (2mL) in a seal tube under argon atmosphere at room temperature. The Argon gas was purged for 5-10rain. Then
CS2CO3 (3.06g, 9.43mmol) and Xantphos (0.54g, 0.94mmol) were added and the resulting reaction mixture was purged with argon gas for 5min.? followed by Pd2(dba)3 (0.43g, 0.47mmol).
The argon gas purging was continued for additional 5min. before sealing the reaction vial. Then the reaction mixture was heated at 100 °C for 12hr. After completion of the reaction by TLC, the reaction mixture was cooled to room temperature, filtered through celite bed and concentrated. The residue was purified by silica gel column chromatography (DCM;MeOH/97:3) to afford the title compound (1.2g, yield: 70.5%). MS (ESI): 365.5 [M+H]+.
Figure imgf000112_0001
d. 4-(4-isopropylpiperazin-l-yl)-2-nUroanitine
TFA (3mL) was slowly added to a stirred solution of tert-buty\ (4-(4-isopropylpiperazin-l-yl)-2- mtrophenyl)carbamate (1.2g, 3.29mmol) in dry DCM (5mL) under argon atmosphere at 0 °C. The resulting reaction mixture was allowed to warm to room temperature and stirred for 12hr. Reaction progress was monitored by LCMS, after completion of the reaction, excess solvents were removed under reduced pressure to afford a crude residue. The crude residue was repeatedly washed with ether to get the title compound (lg, yield: 80.6%) as a red solid. MS (ESI): 265.1 [M+H]+. - 154
Figure imgf000112_0002
Preparation of N-(5-(4-acetylpiperazin-l-yl)-2-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l- methyIureido)pyrimidm-4-yI)amino)phenyl)acrylamide
N- (2-((6-(3 - (2 ?6 -dichloro-3 , 5-dimethoxyphenyl)- 1 -methylureido)pyrimidin-4-yl)amino)- 5 -
(piperazin-l-yl)phenyl)acryl amide (Procedure 2L, Example 157) (8.6 mg, 0.014 rnrnoi) and
DIEA (7,5 μΐ, 0.043 mrnol) were stirred in DCM (1.0 ml) at room temperature under nitrogen atmosphere. A solution of acetyl chloride (1.0 μΐ, 0.016 mrnol) in DCM (1 lul) was added and the reaction mixture was stirred at room temperature for 2 hours. The solvent was evaporated and the resulting material was dissolved in 400ul of DMSO. The DMSO solution was diluted with
1.0 ml of MeOH and purified by prep-HPLC (water/ACN in formic acid condition) to afford the title compound (1.4 mg, yield: 15%). 1H-NMR (400 MHz, MeOH-dt) δ 2.16 (s, 3 H) 3.65 - 3.80 (m, 4 H) 3.94 (s, 6 H) 5.76 (dd, 1 H) 6.15 (s, 1 H) 6.28 - 6.49 (m, 2 H) 6.78 - 6.82 (m, 1 H) 6.95 (dd, 2.89 Hz, 1 H) 7.28 - 7.38 (m, 2 H) 8.31 (d, 1 H) 8.58 (br. s., 1 H); ESI-MS: 643 [M+H]+. - 155 and 156
Figure imgf000113_0001
Preparation of N-(2-((6-(3-(2,6-dichtoro-3>5-dimethoxyphenyI)-l-methylureido)pyrimidin- 4-yI)amino)-5~(l-methoxyethyl)pbenyl)acrylamide and N-(2-((6-(3-(2,6-dichloro-3,5- dimethoxyphenyl)-l-methylureido)pyrimidin-4-yl)aniino)-5-(l-hydroxyethyI)phenyl)
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 4-(l-methoxyethyI)-2-nitroaniline (procedure shown below) in step (d) to afford the title compounds after purification using flash chromatography on silica eluting with 90% to 100% EtOAc/Hexane. Non-polar fraction was N-(2-((6-(3-(2,6-dichloro-3,5- dimethoxyphenyl)- 1 -methylureido)pyrimidin-4-yI)amino)-5-( 1 -methoxyethyl)phenyl)acrylamide (16 mg, yield: 6% in five steps) 1H-NMR (400 MHz, MeOH-d4) δ 1.46 (d, 3 H) 3.29 (s, 3 H) 3.96 (s, 6 H) 4.12 (d, 1 H) 4.40 (q, 1 H) 5.78 - 5.82 (m, 1 H) 6.33 - 6.50 (m, 3 H) 6.82 (s, 1 H) 7.27 (dd, 1 H) 7.55 - 7.64 (m, 2 H) 8.37 (d, 1 H); ESI-MS: 575 [M+H]+. Polar fraction was N-(2- ((6-(3 -(2)6-dichloro-3,5-dimethoxyphenyl)- 1 -methylureido)pyrimidin~4-yl)amino)-5-(l - hydroxyethyl)phenyl) acrylamide (22 mg, yield: 8% in five steps). 1H-NMR (400 MHz, MeOH- d4) δ 1.53 (d, 3 H) 4.00 (s, 6 H) 5.83 (dd, 1 H) 6.29 - 6.56 (m, 3 H) 6.86 (s, 1 H) 7.38 (dd, 1 H) 7.57 (d, 1 H) 7.69 (s, 1 H) 8.40 (d, 1 H); ESI-MS: 661 [M+H]+.
Preparatin of 4-(l-methoxyetbyI)-2-nitroaniline
Figure imgf000113_0002
yield: 87%
a. l-(4~fluoro-3-nitrophenyl)ethanol
l-(4-fluoro-3-nitrophenyl)ethanone (1.0 g, 5.46 mmol) was dissolved in MeOH (15.0 ml) and stirred over ice bath. NaBH4 (0.62 g, 16.0 mmol) was added portionwise. Upon completion of the addition, the reaction mixture was stirred at rt for 10 minutes. The reaction mixture was poured into EtOAc and brine. The organic layer was separated, dried over MgS04 and evaporated. The resulting material was purified by flash chromatography on silica eluting with 20% to 70% EtOAc/Hexane to afford the title compound (881 mg, yield: 87%). %). 1H-NMR (400 MHz, CDClj) δ 1.53 (d,4 H) 4.99 (dd, 1 H) 7.22 - 7.33 (m, 1 H) 7.66 (ddd, 1 H) 8.09 (dd, 1 H)
Figure imgf000114_0001
microwave, 150°C, 5min
yield: 71 %
b. l-fluoro-4-(l-m thoxyethyl)-2-nitrobenzene
l-(4-fluoro-3-nitrophenyl)ethanol (870 mg, 4.7 mmol) was dissolved in MeOH (10 ml) and concentrated sulfuric acid (2.5 ml, 47 mmol) was added carefully. The reaction mixture was heated at 150 °C using microwave (Biotage Initiator) for 5 minutes. After cooling to room temperature, the reation mixture was poured into EtO Ac/water. The organic layer was washed with brine, dried over MgS04 and evaporated. The resulting material was purified by flash chromatography on silica eluting with 0% to 50% EtOAc/Hexane to afford the title compound
(664 mg, yield: 71%). 1H-NMR (400 MHz, CDC13) δ 1.45 (d, 3 H) 3.27 (s, 3 H) 4.36 (q, 1 H) 7.28 - 7.32 (m, 1 H) 7.56 - 7.63 (m, 1 H) 7.98 - 8.04 (m, 1 H)
Figure imgf000114_0002
microwave, 140°C 50mirt
yield: 44%
c. 4-(l-methoxyethyl)-2-nitroaniline
l-fluoro-4-(l-methoxyethyl) -2 -nitrobenzene (664 mg, 3.33 mmol) was stirred in THF (10 ml). NH4OH (0.39 ml, 10.0 mmol) was added and the reaction mixture was stirred at room temperature for 2 hours. Further 780ul of NH4OH was added and the reaction mixture was heated at 120 °C for 10 minutes using microwave (Biotage Initiator) and then at 140 °C for 50 minutes. After cooling to room temperature, the reation mixture was poured into EtOAc/water. The organic layer was washed with brine, dried over MgS04 and evaporated. The resulting material was purified by flash chromatography on silica eluting with 0% to 50% EtOAc Hexane to afford the title compound (290 mg, yield: 44%). 1H-NMR (400 MHz, CDCI3) 8 1.42 (d, 3 H) .22 (s, 3 H) 4.24 (q, 1 H) 6.05 (br. s., 2 H) 6.83 (d, 1 H) 7.38 (dd, 2.01 Hz, 1 H) 8.04 (d, 1 H)
Procedure 2L: Example
Figure imgf000115_0001
Figure imgf000115_0002
Preparation of N-(2-((6-(3-(2,6-dichl0ro-355-dimethoxyphenyl)-l-methylureido)pyrimidin- 4-y amino)-5-(piperazm-l»yl)phenyl)acrylamide
Figure imgf000115_0003
Microwave, 200 °C, 1min
yield: 92%
a, 2-(4-bromo-2~nitrophenyl)isoindoline~l,3-dione 4-bromo-l-fiuoro- -nitrobenzene (2.0 g, 9.1 mmol) and potassium l,3-dioxoisoindolin-2-ide (2.0 g, 10,9 mmol) were placed in microwave vial (0.5-2ml) and NMP (12.0 ml) was added. The reaction mixture was heated at 200 °C using microvave (Biotage, initiator) for lmin. After cooling to rt, the reaction mixture was added dropwise to stirring water which resulted in formation of precipitate. The solid was collected, washed with water and dried under stream of nitrogen to afford the title compound (2.89 g, yield: 92%). 1H-NMR (400 MHz, CDC13) δ 7.26 (s, 2 H) 7.43 (d, 2H) 7.81 - 7.87 (m, 3H) 7.91 (dd, ,2H) 7.95 - 8.05 (m, 3H) 8.33 (d, 1H)
Figure imgf000116_0001
Toluene, 100°C. 5
yield: 30%
b. tert-butyl4-(4-(l,3-dioxoisoindolin~2-yl)-3-nitropheny
2-(4-bromo-2-nitrophenyl)isoindoline-l,3-dione (500 mg, 1.4 mmol), tert-butyl piperazine-1- carboxylate (402 mg, 2.2 mmol), Pd2(dba)3 (66.0 mg, .07 mmol), Xantphos (83 mg, .144 mmol) and CS2CO3 (939 mg, 2.9 mmol) were placed in reaction vial (microwave reaction vial 10-20ml) and purged with nitrogen. Toluene (5.0 ml) was added and nitrogen was bubbled for lOmin. The reaction mixture was heated at 100 °C for 5 hours. Heating was stopped and cooled to rt. The reaction mixture was filtered through a pad of celite and filtrate was evaporated. The remaining material was purified by flash chromatography on silica eluting with 20% to 70% EtOAc/Hexane to afford the title compound (275 mg, yield: 42%). 1H-NMR (400 MHz, CDC13) δ 1.51 (s, 6H) 3.28 - 3.39 (m, 2H) 3.57 - 3.70 (m, 2H) 7.20 - 7.24 (m, 1H) 7.35 (d, 1H) 7.66 (d, 1H) 7.75 - 7.87 (m, lH) 7.96 (dd, 1H)
Figure imgf000116_0002
c. tert-butyl 4-(4-amino-3-nitrophenyl)piperazine-l-carboxylate
To a suspension of tert-butyl 4-(4-(l,3-dioxoisoindolin-2-yl)-3-nitrophenyl)piperazine-l- carboxylate (275.4 mg, 0.6 mmol) in THF (5.0 ml) was added 1.0M Hydrazine in THF (1.8 ml,
1.8 mmol) at room temperature. The reaction mixture was stirred at room temperature for 2 hours. The reaction mixture was poured into EtO Ac/water and organic layer was washed with brine, dried over MgSC<4 and evaporated. The resulting oil was purified by flash chromatography on silica eluting with 20% to 70% EtOAc/Hexane to afford the title compound (182 mg, 93 % yield). 1H-NMR (400 MHz, DMSO-d6) 6 1.42 (s, 9 H) 2.89 - 2.99 (m, 4 H) 3.44 (d, 4 H) 6.98 (d, 1 H) 7.22 (s, 2H) 7.26 - 7.38 (m, 2 H)
Figure imgf000117_0001
yield: 30%
d. tert~butyl 4-(4-((6-(3-(2,6-dichloro~3,5-dimethoxyphenyl)-l-methyl^
(trimethylsilyl) eth oxy) methyl) ureido)pyrimidin~4~yl)amino)~3-n itrophenytypiperazin e-l~ carboxylate
l-(6-chloropyrimidin-4-yl)-3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl~3-((2- (trimethylsilyl)ethoxy)methyI)urea (175 mg, 0.34 mmol), tert-butyl 4-(4-amino-3- ratrophenyl)piperazine-l -carboxylate (90 mg, 0.28 mmol), Pd2(dba)3 (13 mg, 0.014 mmol), Brettphos (13 mg, 0.028 mmol) and sodium tert-butoxide (54 mg, 0.56 mmol) were placed in a reaction vial (2 to 5ml) and purged with nitrogen. Toluene (1.0 ml) was added and nitrogen was bubbled for 5min then the reaction mixture was heated at 100 °C overnight. Heating was stopped and cooled to room temperature. The reaction mixture was filtered through pad of Celite®. The filtrate was evaporated and resulting material was purified by flash chromatography on silica eluting with 20% to 70% EtOAc/Hexane to afford the title compound (102.4 mg, yield: 45%). 1H-NMR (400 MHz, CDC13) δ 0.01 (s, 9 H) 0.85 - 0.98 (m, 2 H) 1.50 (s, 9 H) 3.03 (s, 3 H) 3.17 (br. s, 4 H) 3.56- 3.66 (m, 4 H) 3.75 - 3.97 (m, 8 H) 5.22 (s, 2 H) 6.50 (s, 1 H) 6.95 (s, 1 H) 7.65 (s, 1 H) 8.46 (s, 1 H)
Figure imgf000117_0002
e. tert-b tyl 4-(4~((ten-butoxycarbonyl)(6-(3-(2,6~dichloro-^
(trimethylsilyl)ethoxy) methyl) ureido)pyrimidin-4-yl) mino)-3-nitrophenyl)piperazine-l- carboxylate
tert-butyl 4-(4-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)- 1 -methyl-3-((2- (trimethylsilyl)ethoxy)methyl)ureido)pyrimidin-4-yl)amino)-3 -nitrophenyl)piperazine- 1 - carboxylate (162 mg,0 0.2 mmol), di-tert-butyl dicarbonate (53 mg, 0.24 mmol) and DMAP (4.9 mg, 0.04 mmol) were stirred in THF (2.0 ml) at room temperature under nitrogen atmosphere for 1 hour. The reaction mixture was poured into EtOAc and water. The organic layer was washed with brine, dried over MgS04 and evaporated. The resulting maerial was purified by flash chromatography on silica eluting with 10% to 70% EtOAc/Hexane to afford the title compound (148 mg, yield: 82%). 1H-NMR (400 MHz, CDC13) δ 0.01 (s, 9 H) 0.80 - 1.04 (m, 2 H) 1.4 (s, 9 H) 1.49 (s, 9 H) 3.12 (s, 3 H) 3.19 - 3.36 (m, 4 H) 3.50 - 3.70 (m, 4 H) 3.83 - 3.92 (m, 9 H) 3.96 (d, 1H) 5.14 (d, 1 H) 5.42 (d, 1 H) 6.46 (s, 1 H) 7.05 - 7.19 (m, 2 H) 7.58 (d, 1 H) 8.03 (s, 1 H) 8.29 (s,l H)
Figure imgf000118_0001
te -butyl 4-(3~ mino-4-((tert-butoxycarbonyl)(6-(3-(2,6-dichloro-3tS-dim
methyl-3-((2-(trimethyl$ilyl)ethoxy)methyl)ureido)pyrim
caroxylate
tert-butyl 4-(4~((tert-butoxycarbonyl)(6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl-3-((2- (trimethylsilyl)ethoxy)methyl)ureido)pyrimidin-4-yl)amino)-3-nitrophenyl)piperazine-l- carboxylate (149 mg, 0.164 mmol) was stirred in THF (1.5 ml) and eOH (1.5 ml). Five drops of Raney Nickel suspension in water was added. The solution was stirred under hydrogen atmosphere at room temperature overnight. The reaction was filtered through a pad of Celite®. and filtrate was concentrated to give crude title compound which was taken to the next step without further purification. ESI-MS: 877 [M+H]+.
Figure imgf000119_0001
g. tert-butyl 4-(3-acrylamido-4-((tert-butoxycarbonyl)(6-(3~(2,6-dichloro^
l-methyl-3-((2-(trimethylsilyl)ethoxy) methyl) ureido)pyrimidin-4-yl)amino)phenyl)piperazine-l- carboxylate
Crude tert-butyl 4-(3-arnino-4-((tert-butoxycarbonyl)(6-(3-(2}6-dichloro-3,5-dimethoxyphenyl)- 1 -methyl-3 -((2-(trimethyl silyl)ethoxy)methy l)ureido)pyrimidin-4-y l)amino)phenyl)piperazine- 1 - carboxylate (95 mg, 0.11 mmol) was dissolved in THF (1.5 ml) and stirred over ice bath under nitrogen atmosphere. DIEA (57 μΐ, 0.30 mmol) followed by acryloyl chloride (13 μΐ, 0.16 mmol) were added and the reaction mixture was stirred at room temperature for 1 hour. The reaction mixture was poured into EtOAc and brine. The organic, layer was separated, dried over MgS04 and evaporated. The resulting maerial was purified by flash chromatography on silica eluting with 30% to 100% EtOAc/Hexane to afford the title compound (47 mg, yield: 47%). 1H-NMR
(400 MHz, CDC13) δ 0.02 (s, 9 H) 0.79 - 1.04 (m, 2 H) 1.36 (s,9 H) 1.49 (s, 9 H) 3.08 (s, 4 H) 3.05 (s, 3 H) 3.25 (br. s., 4 H) 3.64 (br. s., 4 H) 3.86 (s, 6 H) 5.27 (s, 1 H) 5.67 - 5,79 (m, 1 H) 6,15 - 6.56 (m, 2 H) 6.48 (s, 1 H) 7.03 - 7.14 (m, 1 H) 7.87 ~ 8.09 (m, 2 H) 8.19 - 8.29 (br. s., 1 H) 8.44 (s, 1 H).
h. N-(2-((6-(3-(2,6-dichloro~3,5-dimethoxyphenyl)-l-methylureido
(piperazin-l-yl)phenyl)acrylamide
/ert-butyl 4-(3-acrylamido-4-((tert-butoxycarbonyl)(6-(3-(2,6-dicM
memyl-3-((2-(1iimethylsilyl)ethoxy)methy
carboxylate (59 mg, 0.06 mmol) was stirred in DCM (2.0 ml). TFA (97 μΐ, 1,3 mmol) was added and the mixture was stirred at room temperature for 6 hours. The reaction mixture was evaporated. The remaining residue was dissolved in DCM and washed with saturated NaHCC , dried over MgS04 and evaporated. The resulting material was dissolved in THF (2.0 ml) and N¾OH (74 μϊ, 1.9 mmol) was added. The reaction mixture was stirred at room temperature for 5 minutes. The reaction mixture was evaporated to obtain the title compound (34 mg, yield:
90%) which was taken to the next step without further purification. !H-NMR (400 MHz, DMSO- d6) δ 2.62 (br. s., 4 H) 3.08 - 3.27 (m, 7 H) 3.77 - 4.03 (m, 6 H) 5.72 (d, 1 H) 6.11 - 6.34 (m, 2 H) 6.40 - 6.62 (m, 1 H) 6.70 - 6.98 (m, 2 H) 7.22 - 7.37 (m, 2 H) 8.32 (s, 1 H) 8.70 (s, 1 H) 9.58 (br. s., 1 H) 12.06 (s, 1 H); ESI-MS: 601 [M+H]+.
Example - 158
Figure imgf000120_0001
N-(2-((6-(3-(2,6-dichloro~3,5-dimethoxyphenyl)-l^
hydroxy ethyl)(methy.)amino)phenyl)acryIamide
The compound was synthesized following the approach outlined in Procedure 2L (Example 157), substituting 2-((tert-butyldimethylsilyl)oxy)-N-methylethanamine, Pd(dba)2 and Ruphos in step (b), and Pd(dba)2, Brettphos and sodium tert-butoxide in step (d) to afford the title compound (64 mg, yield: 16% in seven steps) 1H-NMR (400 MHz, MeOH-d4) δ 3.04 (s, 3 H) 3.26 (ss 3 H) 3.51 (s, 2 H) 3.69 - 3.81 (m, 2 H) 3.94 (s, 6 H) 5.71 - 5.78 (m, 1 H) 6.03 - 6.10 (m, 1 H) 6.26 - 6.45 (m, 2 H) 6.72 (d, 1 H) 6.79 (s, 1 H) 7.04 - 7.10 (m, 1 H) 7.23 (d, 1 H) 8.28 (d, 1 H); ESI- MS: 590 [M+H].
Preparation of 2-((te -butyIdimethylsilyl)oxy)-N-methyIethanamine
Figure imgf000120_0002
DCM, rt, overnight
yield: 56%
a. 2-((tert-butyldimethylsilyl)oxy)~N-methytethanamine
2-(methylamino)ethanol (1.0 g, 13.3 mmol) was stirred in DCM (25.0 ml) under atmosphere of nitrogen. DIEA (3.23 ml, 18.6 mmol) followed by tert-butylchlorodimethylsilane (2.0 g, 13.3 mmol) was added and the reaction mixture was stirred at room temperature over night. The reaction mixture was poured into ether/water. The aqueous layer was extracted with ether three times. The combined organic layer was dried over MgS0 and evaporated and remaining material was dried under high vacuum to afford title compound (1.4 g, yield: 56%), 1H-NMR (400 MHz, CDCla) δ 0.08 (s, 6 H) 0.91 (s, 9 H) 1.93 (br. s., 1 H) 2.48 (s, 3 H) 2.71 (t, 2 H) 3.75 (t, 2 H).
Example - 160
Figure imgf000121_0001
N-(5-(l-amiBoethyl)-2-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methylureido)pyrimidin- 4-yl)amiBo)phenyl)acrylamide TFA salt
The compound was synthesized following the approach outlined in Procedure 2G (Example 123) substituting teri-butyl (l-(4-amino-3-nitrophenyl)ethyl)carbamate, Pd(dba)2, Brettphos and sodium tert-butoxide in step (d) to afford the free base which was converted to TFA salt. The free base was dissolved in DCM and 1 equivalent of TFA was added. The mixture was evaporated and trituration with ether afforded the title compound (65 mg, yield: 23% in five steps). 1H-NMR (400 MHz, MeOH-dj) δ 1.67 (d, 3 H) 3.33 - 3.37 (s, 3 H) 3.92 - 3.97 (s, 6 H) 4.49 (m, 1 H) 5.75 - 5.83 (m, 1 H) 6.30 - 6.50 (m, 3 H) 6.81 (s, 1 H) 7.36 (dd„ 1 H) 7.69 (d, 1 H) 7.82 (d, 1 H) 8.36 - 8.43 (m, 1 H); ESI-MS: 560 [M+H].
Preparation of tert-butyl (l-(4-amioo-3-nitrophenyl)etbyl)carbamate
Figure imgf000121_0002
yield: 96%
a. l-(4-fluoro-3-nitrophenyl)ethanamine
Fuming HN03 (0.48 ml, 10.8 mmol) was added carefully to ice bath cooled concentrated H2S04 (3.6 ml, 68.2 mmol). To the mixture was added l-(4-fluorophenyl)ethanamine (1.0 g, 7.2 mmol) dropwise. The reaction mxiture was stirred with cooling for 50 minutes. The reaction mixture was poured into ice and basified with 3M NaOH solution (24 ml, 72.00 mmol) to about pH8.0. The alkaline solution was extracted with DCM tiwce. The combined organic layer was dried over Na2S04 and evaporated to afford title compound (1,3 g, yield: 96%). 1H-NMR (400 MHz, CDC13) δ 1.41 (d, 6 H) 4.25 (q, 1 H) 7.21 - 7,26 (m, 1 H) 7.67 (ddd, 1 H) 8.10 (dd, 1 H)
Figure imgf000122_0001
b. tert-butyl (l-(4-fluoro-3-nitrophenyl)ethyl)carbamate
To the suspension of l-(4-fluoro-3-nitrophenyl)ethanamine (1.3 g, 6.9 mmol) in THF (10.0 ml) was added DIEA (2.4 ml, 13.8 mmol) followed by di-tert-butyl dicarbonate (1.9 ml, 8.3 mmol). The reaction mixture was stirred at room temperature under nitrogen atmosphere for 2 hours. The reaction mixture was poured into EtO Ac/water and organic layer was washed with brine, dried over MgS04 and evaporated. The resulting maerial was purified by flash chromatography on silica eluting with 10% to 70% EtOAc Hexane to afford the title compound (1.5 g, yield:
75%). 1H-NMR (400 MHz, CDC13) δ 1.37 - 1.50 (m, 12 H) 4.81 (br. s., 2H) 7,23 - 7.26 (;
7.59 (ddd, 1 H) 8.01 (dd, 1 H).
Figure imgf000122_0002
c. tert-butyl (l~(4-amino-3-nitrophenyl)ethyl)carbamate
Tert-butyl (l-(4-fiuoro-3-nitrophenyi)ethyl)carbamate (500 mg, 1.75 mmol) was stirred in THF (2 ml). Ν¾ΟΗ (0.978 ml, 7.0 mmol) was added and the reaction mixture was heated at 1 0 °C for 30 minutes using microwave (Biotage Initiator). N¾OH (0.6 00 ml) was added and then heated at 180 °C. After cooling to room temperature, the reation mixture was poured into EtO Ac/water. The organic layer was washed with brine, dried over MgS04 and evaporated. The resulting material was purified by flash chromatography on silica eluting with 0% to 50% EtOAc/Hexane to afford the title compound (376 mg, yield: 76%) lH-NMR (400 MHz, CDC13) δ 1.43 - 1.48 (m, 12 H) 4.71 (br. s., 2 H) 6.79 (d, 1 H) 7.34 (dd, 1 H) 8.05 (d, 1 H). - 161
Figure imgf000123_0001
Preparation of N-(5-(4-(2-aminoethyl)piperazin-l-yl)-2-((6-(3-(2,6-dichloro-3,5- dimethoxyphenyl)-l-raethylureido)pyrimidin-4-yI)amino)phenyl)acrylamide 2 TFA salt
Figure imgf000123_0002
. tert-butyl (2-(4-(3-acrylamido-4-( (6-(3-(2, 6-dichloro~3, 5-dimethoxyphenyl)-l- methytureido)pyrimMin^-yl)amino)phenyl)piperazin-l-yl)ethy
To the solution of N-(2-((6-(3-(256-dichloro-3,5-dimethoxyphenyl)-l-methylureido)pyrimidin-4- yl)amino)-5-(piperazin-l-yl)phenyl)acrylamide 2,2,2-trifluoroacetate (Procedure 2L, Example · 157) (30 mg, 0.042 mmol) in THF (1.0 ml) and MeOH (1.0 ml) was added tert-butyl (2- oxoethyl)carbamate (13 mg, 0.08 mmol). The reaction mixture was stirred at room temperature for 10 minutes. Sodium cyanoborohydride (7.0 mg, 0.12 mmol) was added and the reaction mixture was stirred at room temperture over night. Solvent was evaporated and saturated NaHC03 solution was added. The mixture was extracted three times with DCM. Combined organic layer was dried over a2S0 and evaporated. The remaining material was purified by flash chromatography on silica eluting with 0% to 15% MeOH in DCM to afford the title compound (17 mg, yield: 54%) ESI-MS: 744 [M+H]+.
Figure imgf000124_0001
N-(5-(4-(2-aminoethyl)piperazin-l-yl)-2-((6-(3-(2,6-dichloro~3,5-dimethoxyphenyl)-l- methylureido)pyrimidiu-4-yl)amino)phenyl)acrylamide 2 TFA salt
feri-butyi (2-acryiamido-4-(4-(2-((tert-butoxycarbonyl)ammo)ethyl)piperazin- 1 -yl)phenyl)(6-(3- (2,6-dichioro-3,5-dimethoxyphenyl)-l-metbyI-3-((2-(trimethylsilyl)ethoxy)m
pyrimidin-4-yl)carbamate (17 mg, 0.017 mmol) was stirred in DCM (1.0 ml). TFA (200 μΐ, 2.6 rnmol) was added and the mixture was stirred at room temperature for 2 hours. Solvent was evaporated and remaining material was triturated with ether. The resulting solid was collected washed with ether and dried under stream of nitrogen to afford the title compound (15 mg, yield:
96%). 1H-NM (400 MHz, MeOH-d4) δ 2.90 (br. s„ 6 H) 3.15 - 3.22 (m, 2 H) 3.33 - 3.41 (m, 4 H) 3.94 (s, 6 H) 5.74 - 5.79 (m, 1 H) 6.17 (s, 1 H) 6.32 - 6.44 (m, 2 H) 6,81 (s, 1 H) 6.92 - 6.98 (m, 1 H) 7.35 (d, 2 H) 8.32 (d, 1 H); ESI-MS: 644 [M+H]+. - 162
Figure imgf000124_0002
N-(2-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methylureido)pyrimidin-4-yl)ainino)-3- fluGFophenyl)acrylamide 2,2,2-trifluoroacetate
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 2-fluoro-6-nitroaniline, Pd(dba)2; Brettphos and sodium tert-butoxide in step (d) to afford the free base, which was converted to TFA salt. The free base was dissolved in DCM and 1 equivalent of TFA was added. The mixture was evaporated and trituration with ether afforded the title compound (47 mg, yield: 22% in four steps). 1H-NMR (400 MHz, MeOH-d4) δ 3.30 (s, 3 H) 3.93 (s, 6 H) 5.72 - 5.78 (m, 2 H) 6.21 - 6.29 (m, 1 H) 6.23 (d, 1 H) 6.27 (d, 1 H) 6.32 - 6.40 (m, 1 H) 6.56 (dd, 1 H) 6.90 (s, 1 H) 7.10 (t, 1 H) 7.25 - 7.37 (m, 1 H) 7.76 (d, 1 H) 8.32 (s, 1 H) 8.89 (s, 1 H) 9.66 (s, 1 H) 11.96 (s, 1 H) ; ESI-MS: 535 [M+H] + - 163
Figure imgf000125_0001
N-(2-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l^
(2-(dimetbylam o)ethyI)phenyl)acrylamide 2,2,2-trifluoroacetate
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting iert-butyl 4-amino-3-nitrophenethylcarbamate (procedure shown below), Pd(dba)2 and Brettphos in step (d) to afford crude TFA salt of the free terminal amine, which was used as-is in the following additional step: To the solution of N-(5-(2-am noethyl)-2-((6-(3- (2,6-dichloro-3}5-dimethoxyphenyl)-3-(hydroxymethyl)-l-methylureido)pyrimidin-4- yl)amino)phenyl)acrylamide 2,2,2-trifluoroacetate (9.2 mg, 0.014 mmol) in THF (1.0 ml) and MeOH (1.0 ml) was added formaldehyde (5.1 μΐ, 0.068 mmol). The reaction mixture was stirred at room temperature for 10 minutes. Sodium cyanoborohydride (3.4 mg, 0.055 mmol) was added and the reaction mixture was stirred at room temperture over night. Solvent was evaporated and saturated NaHC03 solution was added. The mixture was extracted three times with DCM.
Combined organic layer was dried over Na2S04 and evaporated. The remaining material was purified by flash chromatography on silica eluting with 10% to 70% MeOH in DCM to afford free base. To a solution of free base in DCM, TFA(11 μΐ, 0.014 mmol) was added. The solvent was evaporated and dried under high vacuum to afford the title compound (6.3 mg, yield: 66%). 1H-NMR (400 MHz, MeOH-d δ 2.97 (s, 6 H) 3.10 (dd, 2 H) 3.34 (s, 3 H) 3.45 (dd, 2 H) 3.94 (s, 6 H) 4.63 - 4.63 (m5 1 H) 5.76 - 5.81 (m, 1 H) 6.31 - 6.48 (m, 3 H) 6.81 (s, 1 H) 7.25 (dd, 1 H) 7.55 (d, 1 H) 7.62 - 7.72 (m, 1 H) 8.35 (s, 1 H) ; ESI-MS: 588 [M+H].
Preparation of tert-butyl 4-amino-3-nitrophenethylcarbamate
Figure imgf000126_0001
0°C, 45min yield: 96%
ii. (Boc)20, DIEA, THF, o/n
yield: 28% two steps
a, tert-butyl 4-fluoro-3-nitrophenethylcarbamate
2-(4-fluorophenyl)ethanamine (1.0 g, 7.2 mmol) was dissolved in concentrated ¾S04 (4.0 ml, 75 mmol) and cooled over ice bath. Fuming H 03 (0.48 ml, 10.8 mmol) was added carefully dropwise to the mixture. The reaction mxiture was stirred with cooling for 45 minutes and poured into ice. The mixture was basified with 3MNaOH solution (60 ml, 180 mmol) and the alkaline solution was extracted with DCM three times. The combined organic layer was dried over Na2S04 and evaporated to afford crude amine. The crude material was dissolved in THF (15.0ml). Di-tert-butyl dicarbonate (1.7g, 7.9mmol) and DIEA (2.5ml, 14.4mmol) were added and the reaction mixture was stirred at room temperature for 12 hours. The reaction mixture was poured into water and extracted with EtOAc. The organic layer was washed with brine, dried over MgS04 and evaporated. The resulting material was purified by flash chromatography on silica eluting with 0% to 20% EtOAc/Hexane to afford title compound (569 mg, yield: 28%). 1H- NMR (400 MHz, CDC13) 6 1.44 (s, 9 H) 2.88 (t, 2 H) 3.40 (d, 2 H) 7.24 (t, 1 H) 7.42 - 7.54 (ms 1 H) 7.89 (dd, 1 H)
Figure imgf000126_0002
yield: 46%
b. tert-butyl 4-amino-3-nitrophenethylcarbamate
Tert-butyl 4-fluoro-3-nitrophenethyicarbamate (569 mg, 2.00 mmol) was stirred in THF (2.64 ml). NH4OH (2.50 ml, 17.97 mmol) was added and the reaction mixture was heated at 150 °C for 30 minutes using microwave (Biotage Initiator). NH40H (0.600 ml) was added and then heated at 180 °C. After cooling to room temperature, the reation mixture was poured into EtOAc/water. The organic layer was washed with brine, dried over MgS04 and evaporated. The resulting material was purified by flash chromatography on silica eluting with 0% to 30% EtOAc/Hexane to afford the title compound (257 mg, yield: 46%) 1H-NMR (400 MHz, CDC13) δ 1.44 (s, 9 H) 2.73 (t, J=7.03 Hz, 2 H) 3.34 (m, 2 H) 4.46 - 4.62 (m, 1 H) 6.78 (d,J=8.53 Hz, 1 H) 7.24 (dd, J-8.53, 1.76 Hz, 1 H) 7.94 (d, J-t.76 Hz, 1 H).
Exam le - 164
Figure imgf000127_0001
N-(2-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)~l~m
(2-(dimethylamino)ethyl)phenyl)acrylamide 2,2,2-trifluoroacetate
The compound was synthesized following the approach outlined in Procedure 2L (Example 157), substituting 2-nitro-4-(2-((tetrahydro-2H-pyran-2-yl)oxy)ethoxy)aniline (preparation shown below) and sodium ter/-butoxide in step (d) to afford the title compound (7mg, yield: 2.7% over four steps). 1H-NMR (400 MHz, MeOH-d*) δ 3.28 (s, 3 H) 3.88 - 3.97 (m, 8 H) 4.06 - 4.14 (m, 2 H) 5.76 (dd, 1 H) 6.12 (s, 1 H) 6.32 - 6.46 (m, 2 H) 6.79 (s, 1 H) 6.91 (dd, 1 H) 7.34 - 7.41 (m, 2 H) 8.31 (d, 1 H); ESI-MS: 577 [M+H]+.
Preparation o -nitro-4-(2-((tetrahydro-2H-pyran-2-yl)oxy)ethoxy)aniline
Figure imgf000127_0002
yield: 38%
a. 2-nitro-4-(2-((ietrahydro~2H-pyr n-2-yl)oxy)ethoxy)aniline
4-amino-3-nitrophenol (2.0 g, 12.977 mmol) was dissolved in DMF (20 ml) and Potassium carbonate (3.59 g, 25.953 mmol) was added. To the mixture was added 2-(2- bromoethoxy)tetrahydro-2H-pyran (2.55 ml, 16.87 mmol) and the reaction mixture was stirred at 50 °C for 6 hours and then for 3 days at room temperature. The reation mixture was poured into EtOAc/brine. The aqueous layer was extracted twice with EtOAc. The combined organic layer was washed with brine three times, dried over MgS04 and evaporated. The resulting material was purified by flash chromatography on silica eluting with 5% to 40% EtOAc/Hexane to afford title compound (1.4 g, yield: 38%). 1H-NMR (400 MHz, CDC13) δ 1.51 - 1.68 (m, 4 H) 1.71 - 1.89 (m, 2 H) 3.46 - 3.61 (m, 1 H) 3.72 - 3.99 (m, 2 H) 4.03 - 4.24 (m, 3 H) 4.64 - 4.79 (m, 1 H) 6.78 (d, 1 H) 7.09 - 7.17 (m, 1 H) 7.60 (d, 1 H). - 165
Figure imgf000128_0001
N-(2-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methylureido)pyrimidin-4-yl)amino)-3- methylphenyl)acrylamide
The compound was synthesized following the approach outlined in Procedure 2G (Example
123), substituting 2-methyl-6-nitroaniline, Pd(dba)2, Brettphos and sodium tert-butoxide in step (d) to afford the title compound (3.0 mg, yield: 6% in five steps). 1H-NMR (400 MHz, MeOH- d4) δ 3.52 (m, 3 H) 3.95 (m, 6 H) 5.69 (dd, 1 H) 6.21 (dd, 1 H) 6.46 (dd, 1 H) 6.64 (d, 1 H) 6.73 (d, 1 H) 6.82 (s, 1 H) 7.11 (t, 1 H) 8.06 (d, 1 H) 8.55 (d, 1 H) ; ESI- S: 531 [M+H] + - 166
Figure imgf000128_0002
N-(2-((6-(3-(2,6-dichIoro-3,5-dimethoxyphenyl)-l-^
methoxyphenyI)acrylamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 2-methoxy-6-nitroaniline, Pd(dba)2, Brettphos and sodium ter/-butoxide in step (d) to afford the title compound (33.0 mg, yield: 6.1% in five steps). 1H-NMR (400 MHz, MeOH-d4) δ 3.76 (s, 3 H) 3.93 (s, 6 H) 5.70 (dd, 1 H) 6.23 (d, 1 H) 6.52 (d, 1 H) 6.88 - 6.95 (m, 2 H) 7.27 (t, 1 H) 7.44 - 7.56 (m, 1 H) 8.28 (s, 1 H) 8.57 (br. s., 1 H) 9.40 - 9.57 (m, 1 H) 12.15 (s, 1 H) ; ESI-MS: 547 [M+H] + - 167
Figure imgf000129_0001
N-(3-chloro-2-((6-(3-(2,6-dichloro-3,5-dimethoxyphenyI)-l-methylureido)pyrimidin-4- yl)am.n o)pheny I) acrylamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 2-chloro-6-nitroaniline, Pd(dba)2, Brettphos and sodium teri-butoxide in step (d) to afford the title compound (11.0 mg, yield: 5% in five steps). 1H-NMR (400 MHz, MeOH- d4) δ 3.29 (s, 3 H) 3.94 (s, 6 H) 5.71 - 5.75 (m, 1 H) 6.23 (dd, 1 H) 6.56 (dd, 1 H) 6.90 (s, 1 H) 7.31 - 7.39 (m, 2 H) 7.94 (d, 1 H) 8.30 (s, 1 H) 9.01 (s, 1 H) 9.51 - 9.70 (m, 1 H) 12.04 (s, 1 H) ;
Figure imgf000129_0002
Example - 168
Figure imgf000130_0001
Preparation of N-(2-((6-(3-(2,6-dichIoro-3^-dimethoxyphenyI)-l-methylureido)pyrimidin- 4-y!)amino)-5-(4-(2-hydroxyethyI)piperaziii-l-yl)phenyl)acry]amide TFA salt
The compound was synthesized following the approach outlined in Example 161, substituting 2- ((tert-butyldimethylsilyl)oxy)acetaldehyde in step (a) to afford the title compound (13 mg, yield: 42% two steps) 1H-NMR (400 MHz, MeOH-d4) δ 3.14 - 3.20 (m 2 H) 3.35 - 3.37 (m, 2H) 3.73 - 3.76 (m, 2 H) 3.84 - 3.96 (m, 10 H) 5.75 - 5.79 (m, 1 H) 6.24 (s, 1 H) 6.37 - 6.47 (m, 2 H) 6.81 (s, 1 H) 6.99 (dd, 1 H) 7.37 - 7.46 (m, 2 H) 8.34 - 8.36 (m, 1 H) ; ESI-MS: 645 [M+H] +.
Example - 170
Figure imgf000130_0002
(£)-3-chIoro-N-(2-((6-(3-(2,6-dichIoro-3,5-dimethoxyphenyi)-l-methylureido)pyrimidin-4- y 1) ami no) ph eny l)acrylamid e
The title compound was synthesized following the approach outlined in Procedure 2A (Example 100), modifying step (i) to the following procedure: To a solution of l-(6-((2- aminophenyl)amino) pyrimidin-4-yl)-3 -(2,6-dichloro-3 , 5 -dimethoxyphenyl)- 1 -methylurea ( 10 mg, 0.022 mmol), triethylamine (10.9 mg, 0.11 mmol), and (£)-3-chioroacrylic acid (2.76 mg5 0.026 mmol) in DCM (0.4 ml, 6.22 mmol), cooled to 0 °C, was added 2,4,6-tripropyM, 3,5,2,4,6- trioxatriphosphinane 2,4,6-trioxide (0.023 ml, 0.039 mmol, 50% solution in EtOAc). The resulting mixture was stirred at room temperature for 4h and concentrated. The remaining residue was purified by silica gel flash chromatography to obtain the title compound (7.2 mg, yield: 61%). 1H NMR (400 MHz, CDC13) δ 3.30 (s, 3 H) 3.90 (s, 6 H) 6.02 (s, 1 H) 6.36 (d, J=12.92 Hz, 1 H) 6.49 (s, 1 H) 7.21 - 7.33 (m, 2 H) 7.36 - 7.50 (m, 2 H) 7.75 (d, J=6.53 Hz, 1 H) 7.93 (br. s., 1 H) 8.41 (s, 1 H) 12.50 (s, 1 H); MS (ESI); 551.0 [M+H]+
Example - 171
Figure imgf000131_0001
(2)-3-chloro-N-(2-((6-(3-(2,6-dichIoro-3,5-dimethoxyphenyl)-l-methy!ureido)pyrimidin-4- yl)amino)phenyl)acrylamide
The title compound was synthesized following the approach outlined in Procedure 2A (Example 100), modifying step (i) to the following procedure: To a solution of l-(6-((2- aminophenyl)amino) pyrimidin-4-yl)-3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methylurea (20 mg, 0.043 mmol), triethylamine (21.8 mg, 0.22 mmol), and (Z)-3-chioroacr lic acid (5.5 mg, 0.052 mmol) in DCM (0.86 ml, 13.3 mmol), cooled to 0 °C was added 2,4,6-tripropyl- 1 ,3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (49 mg, 0.78 mmrnol, 50% solution of EtOAc). The resulting mixture was stirred at room temperature for 2 h and concentrated. The residue was purified by silica gel column chromatography to obtain the title compound (15 mg, yield: 63%). 1H NMR (400 MHz, CDC13) δ X; MS (ESI): 551.0 [M+H]+. - 172
Figure imgf000132_0001
(Z)-N-(2-((6-(3-(2 ,6-dich !oro-3,5-d imeth oxyphenyl)- 1-m ethylu reido)py rimidin-4- y l)amino)ph enyl)b ut-2- enamid e
The title compound was synthesized following the approach outlined in Procedure 2A (Example 100), modifying step (i) to the following procedure: To a solution of l-(6-((2- aminophenyl)amino) pyrimidin-4-yl)-3 -(2,6-dichloro-3,5-dimethoxyphenyl)- 1 -methylurea (20 mg, 0.043 mmol), triethylamine (21.8 mg, 0.22 mmol), and (Z)-but-2-enoic acid (4.5 mg, 0.052 mmol) in DCM (0.86 ml, 13.3 mmol) was added a solution of 2,4,6-tripropyl- 1,3,5,2,4,6- trioxatriphosphinane 2,4,6-trioxide (49.4 mg, 0.78 mmol, 50% solution EtOAc) at 0 °C, and the resulting mixture was stirred at room temperature for 2h. The mixture was concentrated and the residue was purified by silica gel column to obtain the title compound (8.5 mg, yield: 37%). 1H NMR (400 MHz, CDC13) δ 2.20 (dd, J=7.28, 1.76 Hz, 3 H) 3.30 (s, 3 H) 3.92 (s, 6 H) 5.85 (dd, J=1 L42, 1.76 Hz, 1 H) 5.99 (s, 1 H) 6.29 (dd, J=l L36, 7.34 Hz, 1 H) 6.52 (s, 1 H) 7.23 - 7.34 (m, 2 H) 7.46 (d, J=7.40 Hz, 1 H) 7.55 (br. s., 1 H) 7.78 (d, J=7.53 Hz, 1 H) 8.40 (d, J=0.88 Hz, ί H) 12.50 (s, 1 H); MS (ESI): 531.3 [M+H]+.
Example - 175
Figure imgf000132_0002
(SiZ)~N-(2-((6-(3-(2J6-dichloro-3,5-dimethoxyphenyl)-l-methylureido)pyrimidiii-4- yl)amino)phenyl)-4-hydr0xypent-2-enamide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 2-nitroamlme in step (d) and raethacryloyl chloride in step (g) to afford the title compound (13 mg, yield: 72%). 1H NMR (400 MHz, CDC13) 2.03 (br. s., 3 H) 3.31 (s, 3 H) 3.92 (s, 6 H) 5.49 (s5 1 H) 5.81 (s, 1 H) 6.00 (s, 1 H) 6.53 (s, 1 H) 7.29 (m, 3 H) 7.44 (d, J-7.91 Hz, 1 H) 7.83 (d, J-7.53 Hz, 1 H) 7.92 (s, 1 H) 8.41 (s, 1 H) 12.34 (s} 1 H). MS (ESI): 531.1 [M+H .
Example - 181
Figure imgf000133_0001
(Z)-3-chloro-N-(2-((6-(3-(2,6-dichIoro-3,5-dimethoxyphenyl)-l-methyIureido)pyrimidin-4- yl)amino)-5-(4-ethylpiperazin-l-yl)phenyI)acrylamide
The title compound was synthesized following the approach outlined in Procedure 2C (Example 108), modifying step (g) to the following procedure: To a solution of l-(6-((2-amino-4-(4- ethylpiperazin- 1 -yl)phenyl)amino)pyrimidin-4-yl)-3-(2,6-dichloro-3 ,5-dimethoxyphenyl)- 1 - methylurea (1 1 mg, 0.019 mmol), triethylamine (9.67 mg, 0.096 mmol), and (Z)-3-chloroacrylic acid (2.43 mg, 0.023 mmol) in DCM (0.4 ml, 6.21 mmol) was added a solution of 2,4,6- tripropyl-l?3,5,2,4,6-trioxatriphosphinane 2,4,6-trioxide (21.9 mg, 0.034 mmol) at 0 °C, and the resulting mixture was stirred at room temperature for 30 min. The mixture was concentrated and the residue was purified by silica gel column to obtain the title compound (7.5 mg, yield: 59%). 1H NMR (400 MHz, CDC13) δ 1.29 (t, J=8.0 Hz, 3 H) 2.90 (br. s., 4 H) 3.27 (s, 3 H) 3.45 (t, J=4.64 Hz, 4 H) 3.91 (s, 6 H) 5.90 (s, 1 H) 6.32 (d, J=8.41 Hz, 1 H) 6.52 (s, 1 H) 6.59 (d, J=8.41 Hz, ί H) 6.73 (dd, J-8.78, 2.76 Hz, 1 H) 7.09 (br. s., 1H) 7.24 (s, 2 H) 7.83 (br. s.5 1 H) 8.36 (s, 1 H) 8.69 (br. s., 1 H) 12.52 (s, 1 H); MS (ESI): 663.1 [M+H]+.
- 185
Figure imgf000134_0001
N-(2-((6-(3-(2,6-dich loro-3 ,5-dim eth oxyphenyl) -methyIureido)pyrimidin-4-yl)amino)-6- fluorophenyl)acry!amide
The compound was synthesized following the approach outlined in Procedure 2G (Example 123), substituting 3-fluoro-2-nitroaniline in step (d), omitting step (e), and modifying step (g) to the following procedure: To a solution of l-(6-((2-amino-3-methylphenyl)amino)pyrimidin-4- yl)-3-(2,6-dichloro-3,5-dimethoxyphenyl)-l-methyl~3-((2-(trimethyisilyl)ethoxy)methyl)urea (31 mg, 0.051 mmol), diisopropyl ethylamine (13.1 mg, 0.103 mmol), and acrylic acid (39.9 mg, 0.055 mmol) in DCM (1 ml, 15.54 mmol) was added a solution of 2,4,6-tripropyl-l ,3,5,2,4,6- trioxatriphosphinane 2,4,6-trioxide (48.1 mg, 0.076 mmol, 50% solution EtOAc) at 0 °C, and the resulting mixture was stirred at room temperature for 2h. The mixture was concentrated and the residue was purified by silica gel column to obtain the SEM protected title compound (24 mg, yield: 71% in three steps). Following the final step (g), the title compound was isolated (12 mg, yield: 62%). 1H-NMR (400 MHz, CDC13) δ 3.40 (s, 3 H) 3.93 (m, 6 H) 5.91 (d, 1 H) 6.20 (s, 1 H) 6.30 - 6.45 (m, 1 H) 6.50 - 6.58 (m, 2 H) 7.01 (t, 1 H) 7.28 - 7.35 (m, 1 H) 7.42 - 7.63 (m, 2 H) 8.41 (s, 1 H) 12.43 (br. s., 1 H) ; ESI-MS: 535 [M+H]\
Assays of Biological Activity
Assay of Binding to FGFR4. Purified, recombinant FGFR4 was pre-incubated with 10 μΜ compound overnight at 4 °C, or for 1 hour at room temperature. Following pre-incubation, protein samples were separated using SDS-PAGE and gels were stained with SimplyBlue™ SafeStain (Life Technologies, Grand Island, New York). FGFR bands were cut out and digested using an In-Gel Tryptic Digestion Kit (Thermo Scientific, Waltham, Massachusetts). Digested samples were run on a Thermo Scientific Q Exactive™ LCMS using reverse phase separation and tandem mass spectrometry to identify modified peptides.
Alternatively, following pre-incubation FGFR4 was concentrated and buffer exchanged on an OPTI-TRAP protein concentrating and desalting C4 column (Optimize Technologies). Protein was eluted in acetonitrile containing 0.1% formic acid and run by direct injection on a Thermo Scientific Q Exactive™ LCMS to identify modified, intact FGFR4.
Results provided below in Table 2 confirm covalent adduct formation of the tested compounds with the peptides by correspondence of the expected mass of the peptide-ligand adduct with the mass observed.
TABLE 2
Figure imgf000135_0001
ICs Profiling of Kinase Activity Inhibition. Compounds were profiled for FGFR inhibition activity at Reaction Biology Corporation (Malvern, Pennsylvania) with their Kinase HotSpotSM assay. See, Anastassiadis et al, 2011, Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat Biotechnol 29, 1039-1045.
Recombinant FGFR1 (2.5 nM), FGFR2 (1 nM), FGFR3 (5 nM), or FGFR4 (12 nM)
(Invitrogen™) was prepared as a mixture with substrate KKKSPGEYVNIEFG (SEQ ID NO:l)
(20 μΜ, FGFR1 substrate); and Poly [E,Y] 4:1 (0.2 mg/ml, FGFR2,3,4 substrate)] in kinase reaction buffer (20 mM HEPES-HC1, pH 7.5, 10 mM MgCl2, 2 mM MnCl2, 1 mM EGTA,
0.02% Brij35, 0.1 mM Na3V04, 0.02 mg/ml BSA, 2 mM DTT, and 1% DMSO). Compound was added to the enzyme/substrate mixture using acoustic technology (Labcyte® Echo 550,
Sunnyvale, California) (see, Olechno et al, 2006, Improving IC50 results with acoustic droplet ejection. JALA ί 1, 240-246) and pre-incubated for 0, 15, or 60 minutes at room temperature. After compound pre-incubation, a mixture of ATP (Sigma-Aldrich®) and 33Ρ-γ-ΑΤΡ
(PerkinElmer) was added to a final concentration of 10 μΜ to initiate kinase reactions. Reactions were incubated for 120 minutes at room temperature and then spotted onto Whatman™ P81 ion exchange filter paper. Unbound phosphate was removed by extensively washing filters in 0.75% phosphoric acid. See, Anastassiadis et al, 2011, Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat Biotechnol 29, 1039-1045.
Results for FGFR4 and FGFRl are shown next to individual compounds listed in Table 1 above. The compounds showed selective inhibition of FGFR4, with a higher IC50 for FGFRl.
Without wishing to be bound by theory, the IC50 activity with respect to FGFRl is generally representative of the activity with respect to FGFRl, FGFR2, and FGFR3. See also, Dieci et al., 2013, Fibroblast Growth Factor Receptor inhibitors as a Cancer Treatment: From a Biologic Rationale to Medical Perspectives, Cancer Discovery, F1-F16.
To confirm, some of the compounds were also tested for FGFR2 and FGFR3 inhibition. These results shown below in Table 3 are consistent with the IC50 activity of FGFRl being generally representative of the activity of FGFRl, FGFR2, and FGFR3, and further demonstrates the selectivity of these FGFR4 inhibitors.
TABLE 3
Figure imgf000136_0001
In vivo efficacy in tumor models. Compound 108 was evaluated for its ability to inhibit tumor growth in nude mice bearing tumor xenografts from three different human hepatocellular carcinoma tumor cell lines. These cell lines are representative of cancers having an altered FGFR4 and/or FGF19 status. See Sawey et al., Cancer Cell 19(3): 347-358 (2011).
Animals: Nude mice, ages 6-8 weeks, and weighing approximately 19-25 g, were purchased from Taconic (Taconic, Hudson, New York). All animal experiments were done in accordance with protocols approved by the Institutional Animal Care and Use Committee.
Tumor xenografts and treatment: 7.5 x 106 HUH7 cells (HSRRB cat. no. JCRB0403), 5 x 106 Hep3B (ATCC cat. no. HB8064), or 2.5 x 106 JHH7 cells (HS RB cat. no. JCRB1031), each in a total volume of 100 μΐ, 1:1 Matrigel (Corning Inc, Corning, NY), were injected subcutanousiy (s.c) into the right lateral flank. When tumors reached 150-200 mm3, the mice were randomized into treatment groups of 5-10 animals. Dosing was peformed twice daily by
intraperitoneal injection at the indicated dosages for 15 days using Compound 108, formulated in a vehicle of 5% DMSO (Alfa Aesar, Ward Hill, MA), 10% PEG300 (Sigma, St. Louis, MO), 8% TWEEN® 80 (Sigma, St. Louis, MO), 77% USP Saline at the desired concentration. Tumor volumes were collected twice weekly using the formula Volume = (length* width2)/2. Body weights were collected twice weekly, as well. All animals were observed and cared for in accordance with The Guide for Care and Use of Laboratory Animals, 8th edition (National Academies Press, Washington D.C.).
Statistical methods: Statistical comparisons were made at the end of the experiment using Repeated Measures Anova with Bonferroni post-test for comparisons of treatment groups, using GraphPad Prism 5. The following criteria were used to determine Progressive Disease, Stable Disease, Partial Regression, and Complete Regression. Progressive Disease is defined as three consecutive measurements increasing from best response or >120% initial tumor volume. Stable Disease is three consecutive measurements <120% and >50% of initial tumor volume, whereas three consecutive measurements <50% initial tumor volume qualifies as a Partial Regression. A Complete Regression is three consecutive measurements <30 mm3. Chi-squared test was used to compare responses between treatment groups (Microsoft Excel).
Results from animals bearing tumors from HUH7, HEP3B, and JHH7 cancer cells are shown in FIGS. 1-3, respectively, and are also reflected in Table 4.
TABLE 4 - Inhibition of Tumor Growth in FGF19 amplified HCC xenografts
HUH7 (n=10 per group)
Figure imgf000137_0001
Figure imgf000138_0001
JHH7 (n=10 per group)
Figure imgf000138_0002
These data demonstrate that compound 108 is efficacious in all models. Among the three models, HEP3B is the most sensitive, JHH7 the least sensitive and HUH7 showing intermediate sensitivity to compound 108. Although a dose response can be seen in FIG. 3 for JHH7, there was Progressive Disease in all dose levels tested.
Comparative studies of Compound 108 with BGJ398. Comparative studies were done with Compound 108 and the known FGFR inhibitor BJG398.
Biochemical Kinase assay protocol to obtain ICso: Recombinant FGFR1 (2.5 nM), or FGFR4 (12 nM) was prepared as a mixture with substrate KK SPGEYVNIEFG (SEQ ID NO:l) (20 μΜ, FGFR1 substrate); Poly [E,Y] 4: 1 (0.2 mg/ml, FGFR2,3,4 substrate)] in kinase reaction buffer (20 mM HEPES-HCl, pH 7.5, 10 mM MgCl2? 2 mM MnCl2, 1 mM EGTA, 0.02% Brij35, 0.1 mM Na3V04, 0.02 mg/ml BSA, 2 mM DTT, and 1% DMSO). Compound was added to the enzyme/substrate mixture using acoustic technology and pre-incubated for 0, 15, or 60 minutes at room temperature. After compound pre-incubation, 33Ρ-γ-ΑΤΡ was added at a final concentration of 10 μΜ to initiate kinase reactions. Reactions were incubated for 120 minutes at room temperature. Substrate phosphorylation was monitored by filter assay, as above. Results are shown in Table 5. The results reported show that compound 108 is a more potent FGFR4 inhibitor, whereas BGJ398 is a more potent FGFR1 inhibitor.
TABLE 5 - Comparative Testing of Compound 108 and BGJ398 with Biochemical Kinase assay
Figure imgf000138_0003
FGFR1 513 1.0
Cellular Viability assay protocol to obtain GI5o: Cells lines were cultured at 37°C, 5% C02 and 95% humidity. Culture media were purchased from GIBCO®, USA. For viability assay, 2000 cells/well were seeded in 96 well plates, incubated for 24h before compound treatment. Following compound addition, plates were incubate for 72h at 37°C with 5% C02) and then measured by means of CTG assay (CellTiter-Glo® Luminescent Cell Viability Assay, Cat. No.: G7572, Promega). Results are shown in Table 6. The table shows compound 108 is more potent than BGJ398 in Hep3B cells, an FGF19 amplified line. The potency in HUH7 and JHH7, the other two FGF19 amplified lines, are comparable between compound 108 and BGJ398. HepG2 (ATCC cat. no. HB-8065), SNU398 (ATCC cat. no. CRL-2233) and SNU449 (ATCC cat. no. CRL-2234) are FGF 19 non-amplified cell lines that were used as controls,
GI5o is the concentration of test drug where 100 (T - T0)/(C - TO) = 50, See, e.g., Monks et al., Feasibility of a High-Flux Anticancer Drug Screen Using a Diverse Panel of Cultured Human Tumor Cell Lines, J Natl Cancer Inst (1991) 83(11):757-766; Boyd et al., Data Display and Analysis Strategies for the NCI Disease-oriented In Vitro Antitumor Drug Screen, in CYTOTOXIC ANTICANCER DRUGS: MODELS AND CONCEPTS FOR DRUG DISCOVERY AND
DEVELOPMENT, Valeriote et al,, eds. (1990), pp. 11-34. The luminescence of the test well after a 72h period of exposure to test drug is T, the luminescence at time zero is TO, and the control luminescence is C. The GI5o measures the growth inhibitory power of the test agent.
TABLE 6 - Comparative Testing of Compound 108 and BGJ398 in Cellular Viability assays
Figure imgf000139_0001
In vivo efficacy comparison: Nude mice were used for these experiments as above. 5.0 x 106 Hep3B cells in a total volume of 100 μΐ, 1 :1 Matrigel (Corning Inc, Corning, New York), were injected s.c, into the right lateral flank, When tumors reached 150-200 mm3 the mice were randomized into treatment groups of 5-10 animals. Treatment was then started using Compound 108, formulated in a vehicle of 5% DMSO (Alfa Aesar, Ward Hill, MA), 10% PEG300 (Sigma, St. Louis, MO), 8% TWEEN® 80 (Sigma, St. Louis, MO), 77% USP Saline at the desired concentration. BGJ398, formulated as a suspension in 0.5% Methylcelmlose (Sigma)/0.2% TWEEN® 80, was suspended at the desired concentration. Both drugs were dosed for 18 dyas, except for one treatment group (see below). Tumor volumes were collected twice weekly using the formula Volume = (length*width2)/2. Body weights were collected twice weekly as well. All animals were observed and cared for in accordance with The Guide for Care and Use of Laboratory Animals, 8th edition (National Academies Press, Washington D.C,)- The results of this comparative in vivo study are shown in FIG. 4.
The data show that compound 108 is more efficacious than BGJ398 at tolerable dosage levels. Although BGJ398 at 60mg/kg showed efficacy comparable to compound 108, the dosing of this BGJ398 60mg/kg group had to be terminated on Day 11 due to poor health of animals. This difference in toxicity is not due to routes of administration because the group of animals dosed orally with BGJ398 at 30mg/kg did not exhibit poor health.
The foregoing is illustrative of the present invention, and is not to be construed as limiting thereof. The invention is defined by the following claims, with equivalents of the claims to be included therein.

Claims

We claim:
1. A compound of Formula I:
Figure imgf000141_0001
wherein:
R3 is selected from the group consisting of: Chalk !,
Figure imgf000141_0002
6alkyl, R^heterocyciyld-ealkyl, R10arylCi-6alkyl, and R10heteroarylCi-6alkyl, wherein R10 and R11 are each independently selected from the group consisting of; hydrogen and Ci-6alkyl;
E is selected from the group consisting of:
-NR13C(0)CR14=CHR15, and
-NR13C(0)C≡CR14,
wherein R° is selected from the group consisting of: hydrogen and methyl, and R14 and R15 are each independently selected from the group consisting of: hydrogen, methyl, fluoro and chloro;
R12 is selected from the group consisting of: hydrogen, halo, Ci-ealkyl,
Figure imgf000141_0003
hydroxyC 1-6alkyl, hy droxy C i -6alkoxy ,C i ,6alkoxyC 1-6alkoxy 5 C \ -6alkoxyC i -6alky 1 ,
R5R6heterocyclyl, ~C(0)heterocyclylR5R , R5R6heterocyclylC3-6alkyl, NR5R6, NR5R6C1-6alkyi, -C(0)NR5R6, and NR5R6C1.6alkyoxyJ wherein R5 and R6 are each independently selected from the group consisting of hydrogen, Ci-ealkyl, hydrox Ci-6alkyl, ammoCi-eaikyl,
-C(0)C1-6alkyl and
Figure imgf000141_0004
and
R1 is phenyl, wherein said phenyl is substituted 2, 3, or 4 times with independently selected halo or Cj-galkoxy, or a pharmaceutically acceptable salt thereof,
2. The compound of claim 1 , wherein R3 is
Figure imgf000141_0005
or a pharmaceutically acceptable salt thereof.
3. The compound of claim 1, wherein R3 is selected from the group consisting of: methyl, methoxyethyl, 4-pyridylmethyl, 3-pyridylmethyl, 2-pyridylmethyl, benzyl, N,N- dimethylaminopropyl, 3-methylisoxazol-5-yl-methyl, and 4-methylpiperazin-l-yl-propyl, or a pharmaceutically acceptable salt thereof.
4. The compound of any one of claims 1-3, wherein E is -NR C(0)CH-CHR15 or
-NR13C(0)CF=CH2, wherein R13 and R15 are as defined above, or a pharmaceutically acceptable salt thereof,
5. The compound of any one of claims 1 -3, wherein E is
Figure imgf000142_0001
or a pharmaceutically acceptable salt thereof.
6. The compound of any one of claims 1-5, wherein R is selected from the group consisting of: hydrogen, fluoro, chloro, methyl, methoxy, Ν,Ν-dimethylaminoethyl, piperazin-1- yl5 4-ethylpiperazm-l-yl, 4-ethylpiperazin-l-yl-methyl, l-methylpiperidine-4-yl, 1- ethylpiperidine-4-yl, N,N-dimethylaminomethyl, N,N-dimethylaminopropyl, piperidine-4-yl, morpholino, 3,5-dimethylpiperazin-l-yl, 4-(methylsulfonyl)piperazin-l-yl, N,N- dimethylaminoethoxy, 4-(2-hydroxyethyl)piperazin-l-yl, hydroxyefhoxy, methoxyethoxy, hydroxymethyl, methoxymethyl, 2-methoxypropyl, 2-hydroxypropyl, 2-aminopropyl,4- methylpiperazin-l-yl-carbonyl, 4-ethylpiperazin-l-yl-carbonyl, 4-[2-propyl]piperazin-l-yl, 4- acetylpiperazin-l-yl, N-methyl-N-hydroxyethyl-amino, Ν,Ν-dimethylamido, and 4-(2- aminoethyI)piperazin-l-yl, or a pharmaceutically acceptable salt thereof.
7. The compound of any one of claims 1-5, wherein R12 is selected from the group consisting of: hydrogen, C1-6alkyl, hydroxyC1-6alkyl, R5R6heterocyclyl, RsR6heterocyclyiC1- 6alkyl, -C(0)NR5R6, NR5R6Ct-6aikyl, NR5R Ci-6alkyoxy, C1-6alkoxy, and Ci.6alkoxyCwaIkyl, whererin R5 and R6 are each independently selected from the group consisting of: hydrogen, Q. 6alkyl, hydroxyCi.6alkyl , -C(0)Ct-6alkyl and C]-6alkylsulfonyl, or a pharmaceutically acceptable salt thereof.
8. The compound of any one of claims 1-5, wherein R12 is RsR6heterocyclyl, wherein R5 and 6 are as defined above, or a pharmaceutically acceptable salt thereof.
9. The compound of any one of claims 1-8, wherein R5R6heterocyclyl is R5R6piperazinyl, wherein R5 and R6 are as defined above, or a pharmaceutically acceptable salt thereof.
10. The compound of any one of claims 1-5, wherein R12 is 4-ethylpiperazin-l-yl, or a pharmaceutically acceptable salt thereof.
11. The compound of any one of claims 1-7, wherein R is not hydrogen, or a
pharmaceutically acceptable salt thereof.
12. The compound of any one of claims 1-11, wherein R1 is 2,6-dichloro-3,5- dimethoxyphenyl, or a pharmaceutically acceptable salt thereof.
13. The compound of any one of claims 1-12, wherein said compound is a compound of Formula 1(a):
Figure imgf000143_0001
1(a) wherein R3, E, Ri2 and R1 are as defined above,
or a pharmaceutically acceptable salt thereof.
The compound of claim 1, wherein said compound is selected from the group consisting
Figure imgf000143_0002
Figure imgf000144_0001
Figure imgf000145_0001
ı44
Figure imgf000146_0001
Figure imgf000147_0001

Figure imgf000148_0001
or a pharmaceutically acceptable salt thereof. The compound of claim 1, wherein said compound is selected from the group consisting
Figure imgf000149_0001
Figure imgf000150_0001

Figure imgf000151_0001
or a pharmaceutically acceptable salt thereof.
The compound of claim 1, wherein said compound is selected from the group consisting
Figure imgf000151_0002
or a pharmaceutically acceptable salt thereof.
17. The compound of claim 1 which is:
Figure imgf000152_0001
or a pharmaceutically acceptable salt thereof.
18. A pharmaceutical composition comprising a compound or salt of any one of claims 1-17 and a pharmaceutically acceptable carrier.
19. The pharmaceutical composition of claim 18, wherein said composition is formulated for oral, intravenous or subcutaneous administration.
20. A method of treating hepatocellular carcinoma in a subject in need thereof comprising administering to said subject a treatment effective amount of a compound or salt of any one of claims 1-17 or the pharmaceutical composition of any one of claims 18 or 19.
21. The method of claim 20, wherein said hepatocellular carcinoma has altered FGFR4 and/or FGF 19 status.
22. The method of claim 21, wherein said altered FGFR4 and/or FGF19 status comprises increased expression of FGFR4 and/or FGF 19.
23. A method of treating hepatocellular carcinoma in a subject in need thereof, comprising: detecting an altered FGFR4 and/or FGF19 status in a biological sample containing cells of said hepatocellular carcinoma, and if said hepatocellular carcinoma has said altered FGFR4 and/or FGF 19 status,
administering the compound or salt of any one of claims 1-17 or the pharmaceutical composition of any one of claims 18 or 19 to said subject in a treatment-effective amount.
24. The method of claim 23, wherein said altered FGFR4 and/or FGF19 status comprises increased expression of FGFR4 and/or FGF19.
Use of a compound or salt of any one of claims 1-1 or the pharmaceutical composition of any one of claims 18 or 19 in a method of treatment for hepatocellular carcinoma.
26. The use of claim 25, wherein said hepatocellular carcinoma has altered FGFR4 and or FGF19 status.
27. The use of claim 26, wherein said altered FGFR4 and/or FGF19 status comprises increased expression of FGFR4 and/or FGF1 .
28. Use of a compound or salt of any one of claims 1-17 in the preparation of a medicament.
29. The use of claim 28, wherein said medicament is for the treatment of hepatocellular carcinoma.
30. The use of claim 29, wherein said hepatocellular carcinoma has altered FGFR4 and/or FGF19 status.
31. The use of claim 30, wherein said altered FGFR4 and/or FGF 19 status comprises increased expression of FGFR4 and/or FGF 19.
PCT/US2014/060857 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors WO2015057938A1 (en)

Priority Applications (30)

Application Number Priority Date Filing Date Title
KR1020167009625A KR101826015B1 (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
CA2924206A CA2924206C (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
JP2016523266A JP6139788B2 (en) 2013-10-18 2014-10-16 Pyrimidine FGFR4 inhibitor
NZ717559A NZ717559A (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
EP14790956.8A EP3057943B1 (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
CN201480056358.4A CN105899490B (en) 2013-10-18 2014-10-16 Pyrimidine FGFR4 inhibitor
MYPI2016000489A MY184733A (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
UAA201605322A UA116920C2 (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
SG11201602069WA SG11201602069WA (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
LTEP14790956.8T LT3057943T (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
ES14790956.8T ES2679521T3 (en) 2013-10-18 2014-10-16 FGFR4 pyrimidine inhibitors
SI201430792T SI3057943T1 (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
EP18167989.5A EP3421457B1 (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
MX2016004933A MX2016004933A (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors.
AU2014337291A AU2014337291B9 (en) 2013-10-18 2014-10-16 Pyrimidine FGFR4 inhibitors
RS20180834A RS57444B1 (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
PL14790956T PL3057943T3 (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
RU2016118981A RU2715708C2 (en) 2013-10-18 2014-10-16 Fgfr4 inhibitors
BR112016008110-2A BR112016008110B1 (en) 2013-10-18 2014-10-16 FGFR4 PYRIMIDINE INHIBITOR COMPOSITION, PHARMACEUTICAL COMPOSITION UNDERSTANDING COMPOUND AND THERAPEUTIC USES OF THESE
DK14790956.8T DK3057943T3 (en) 2013-10-18 2014-10-16 PYRIMIDINE-FGFR4 INHIBITORS
US15/000,659 US9434697B2 (en) 2013-10-18 2016-01-19 Pyrimidine FGFR4 inhibitors
IL244373A IL244373B (en) 2013-10-18 2016-03-01 Pyrimidine fgfr4 inhibitors
PH12016500676A PH12016500676A1 (en) 2013-10-18 2016-04-12 Pyrimidine fgfr4 inhibitors
US15/221,018 US9730931B2 (en) 2013-10-18 2016-07-27 Pyrimidine FGFR4 inhibitors
HK16111183.6A HK1223091A1 (en) 2013-10-18 2016-09-23 Pyrimidine fgfr4 inhibitors fgfr4
US15/627,851 US10537571B2 (en) 2013-10-18 2017-06-20 Pyrimidine FGFR4 inhibitors
HRP20181129TT HRP20181129T1 (en) 2013-10-18 2018-07-17 Pyrimidine fgfr4 inhibitors
CY181100754T CY1121129T1 (en) 2013-10-18 2018-07-18 TOY FGFR4 INHIBITORS
AU2018271284A AU2018271284B2 (en) 2013-10-18 2018-11-27 Pyrimidine FGFR4 inhibitors
US16/717,080 US10912774B2 (en) 2013-10-18 2019-12-17 Pyrimidine FGFR4 inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361892881P 2013-10-18 2013-10-18
US61/892,881 2013-10-18

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/000,659 Continuation US9434697B2 (en) 2013-10-18 2016-01-19 Pyrimidine FGFR4 inhibitors

Publications (1)

Publication Number Publication Date
WO2015057938A1 true WO2015057938A1 (en) 2015-04-23

Family

ID=51842899

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2014/060857 WO2015057938A1 (en) 2013-10-18 2014-10-16 Pyrimidine fgfr4 inhibitors
PCT/US2014/060902 WO2015057963A1 (en) 2013-10-18 2014-10-16 Fgfr4 inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2014/060902 WO2015057963A1 (en) 2013-10-18 2014-10-16 Fgfr4 inhibitors

Country Status (33)

Country Link
US (4) US9434697B2 (en)
EP (2) EP3421457B1 (en)
JP (2) JP6139788B2 (en)
KR (1) KR101826015B1 (en)
CN (2) CN105899490B (en)
AR (1) AR098048A1 (en)
AU (2) AU2014337291B9 (en)
BR (1) BR112016008110B1 (en)
CA (1) CA2924206C (en)
CL (1) CL2016000918A1 (en)
CY (1) CY1121129T1 (en)
DK (1) DK3057943T3 (en)
ES (1) ES2679521T3 (en)
HK (1) HK1223091A1 (en)
HR (1) HRP20181129T1 (en)
HU (1) HUE039268T2 (en)
IL (1) IL244373B (en)
JO (1) JO3515B1 (en)
LT (1) LT3057943T (en)
MX (1) MX2016004933A (en)
MY (1) MY184733A (en)
NZ (1) NZ717559A (en)
PE (1) PE20160679A1 (en)
PH (1) PH12016500676A1 (en)
PL (1) PL3057943T3 (en)
PT (1) PT3057943T (en)
RS (1) RS57444B1 (en)
RU (1) RU2715708C2 (en)
SG (1) SG11201602069WA (en)
SI (1) SI3057943T1 (en)
TW (1) TWI597268B (en)
UA (1) UA116920C2 (en)
WO (2) WO2015057938A1 (en)

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9321786B2 (en) 2013-03-15 2016-04-26 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9434697B2 (en) 2013-10-18 2016-09-06 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
WO2016164703A1 (en) * 2015-04-09 2016-10-13 Eisai R & D Management Co., Ltd. Fgfr4 inhibitors
WO2016168331A1 (en) * 2015-04-14 2016-10-20 Eisai R&D Management Co., Ltd. Crystalline fgfr4 inhibitor compound and uses thereof
WO2017015363A1 (en) * 2015-07-20 2017-01-26 Dana-Farber Cancer Institute, Inc. Novel pyrimidines as egfr inhibitors and methods of treating disorders
US9663524B2 (en) 2013-03-15 2017-05-30 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as protein kinase inhibitors
WO2017118438A1 (en) * 2016-01-08 2017-07-13 杭州英创医药科技有限公司 Heterocyclic compound used as fgfr inhibitor
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
WO2017196854A1 (en) * 2016-05-10 2017-11-16 Eisai R&D Management Co., Ltd. Drug combinations for reducing cell viability and/or cell proliferation
WO2017198221A1 (en) 2016-05-20 2017-11-23 浙江海正药业股份有限公司 Pyrimidine derivative, method for preparing same and use thereof in medicine
CN107400092A (en) * 2016-05-20 2017-11-28 浙江海正药业股份有限公司 Pyridine derivatives and preparation method thereof and its purposes in medicine
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
WO2018039324A1 (en) 2016-08-23 2018-03-01 Eisai R&D Management Co., Ltd. Combination therapies for the treatment of hepatocellular carcinoma
WO2018049233A1 (en) 2016-09-08 2018-03-15 Nicolas Stransky Inhibitors of the fibroblast growth factor receptor in combination with cyclin-dependent kinase inhibitors
WO2018055503A1 (en) 2016-09-20 2018-03-29 Novartis Ag Combination comprising a pd-1 antagonist and an fgfr4 inhibitor
WO2018083603A1 (en) 2016-11-02 2018-05-11 Novartis Ag Combinations of fgfr4 inhibitors and bile acid sequestrants
US10000490B2 (en) 2014-01-15 2018-06-19 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10065966B2 (en) 2013-03-15 2018-09-04 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
CN108503593A (en) * 2017-02-28 2018-09-07 暨南大学 2- amino-metadiazine compounds and its application
JP2018527321A (en) * 2015-07-20 2018-09-20 タイペイ・メディカル・ユニバーシティTaipei Medical University Azaaryl compounds substituted with chlorobenzene
US10131667B2 (en) 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
CN105884760B (en) * 2016-06-13 2019-01-04 厦门市蔚嘉化学科技有限公司 A kind of preparation method than his intermediate of department
US10196436B2 (en) 2012-07-11 2019-02-05 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10221154B2 (en) 2013-10-25 2019-03-05 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
CN109422760A (en) * 2017-09-01 2019-03-05 南京圣和药业股份有限公司 FGFR4 inhibitor and its application
WO2019047826A1 (en) 2017-09-05 2019-03-14 博奥阿迪斯生物科技公司 Aromatic derivative, preparation method for same, and medical applications thereof
WO2019139977A1 (en) 2018-01-10 2019-07-18 Eisai R&D Management Co., Ltd. Combination therapies for the treatment of hepatocellular carcinoma
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11008292B2 (en) 2016-11-17 2021-05-18 Guangdong Zhongsheng Pharmaceutical Co., Ltd FGFR4 inhibitor and preparation method and use thereof
WO2021129561A1 (en) 2019-12-23 2021-07-01 北京赛特明强医药科技有限公司 Cyano-substituted pyridine and cyano-substituted pyrimidine compound and preparation method therefor and application thereof
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
WO2022075482A1 (en) 2020-10-05 2022-04-14 株式会社カイオム・バイオサイエンス Medicine for treating cancer
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
WO2022262577A1 (en) * 2021-06-17 2022-12-22 浙江海正药业股份有限公司 Salt of selective fgfr4 inhibitor, and preparation method therefor and application thereof
WO2022268063A1 (en) 2021-06-23 2022-12-29 浙江海正药业股份有限公司 Crystal form of pyrimidine derivative and preparation method therefor
RU2787993C2 (en) * 2018-01-10 2023-01-16 ЭЙСАЙ Р энд Д МЭНЭДЖМЕНТ КО., ЛТД. (EISAI R&D MANAGEMENT CO., LTD.) Combination therapies for treatment of hepatocellular carcinoma
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11976058B2 (en) 2019-03-05 2024-05-07 Bioardis Llc Aromatic derivatives, preparation methods, and medical uses thereof

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201602183QA (en) 2013-10-25 2016-05-30 Novartis Ag Ring-fused bicyclic pyridyl derivatives as fgfr4 inhibitors
EP3200786B1 (en) 2014-10-03 2019-08-28 Novartis AG Use of ring-fused bicyclic pyridyl derivatives as fgfr4 inhibitors
US9802917B2 (en) 2015-03-25 2017-10-31 Novartis Ag Particles of N-(5-cyano-4-((2-methoxyethyl)amino)pyridin-2-yl)-7-formyl-6-((4-methyl-2-oxopiperazin-1-yl)methyl)-3,4-dihydro-1,8-naphthyridine-1(2H)-carboxamide
CN105418441A (en) * 2015-12-22 2016-03-23 中国工程物理研究院化工材料研究所 Preparation method for 2,3-dichloro-4-hydroxyaniline
KR101812266B1 (en) 2016-11-25 2017-12-27 한국과학기술연구원 4-((2-Acrylamidophenyl)amino)thieno[3,2-d]pyrimidin-7-carboxamide derivatives as protein kinase inhibitors
CN108239069B (en) * 2016-12-26 2021-01-05 南京药捷安康生物科技有限公司 Inhibitor for fibroblast growth factor receptor and application thereof
EP3663285B1 (en) * 2017-08-04 2024-01-24 Abbisko Therapeutics Co., Ltd. Formylpyridine derivative having fgfr4 inhibitory activity, preparation method therefor and use thereof
CN112313207B (en) * 2018-06-22 2023-02-14 北京赛特明强医药科技有限公司 Cyano-substituted pyridine and cyano-substituted pyrimidine compounds, and preparation methods and applications thereof
LT3966206T (en) 2019-05-10 2023-11-10 Deciphera Pharmaceuticals, Llc Heteroarylaminopyrimidine amide autophagy inhibitors and methods of use thereof
EP3966207B1 (en) 2019-05-10 2023-11-01 Deciphera Pharmaceuticals, LLC Phenylaminopyrimidine amide autophagy inhibitors and methods of use thereof
EP3983081A1 (en) 2019-06-17 2022-04-20 Deciphera Pharmaceuticals, LLC Aminopyrimidine amide autophagy inhibitors and methods of use thereof
CN112341344B (en) * 2019-08-09 2021-10-26 上海喀露蓝科技有限公司 Preparation method of kinase inhibitor intermediate
JP2023533903A (en) 2020-05-15 2023-08-07 エーザイ・アール・アンド・ディー・マネジメント株式会社 Method for treating cancer with oral formulation of FGFR4 inhibitor
US20230172867A1 (en) 2020-05-15 2023-06-08 Eisai R&D Management Co., Ltd. A method for treating cancer with an oral dosage form of an fgfr4 inhibitor
CN114276266A (en) * 2021-12-30 2022-04-05 兰州康鹏威耳化工有限公司 Preparation method of 4-amino-2-fluorobenzamide
CN114560815A (en) * 2022-03-04 2022-05-31 北京工业大学 (2- ((5-chloro-substituted phenylaminopyrimidin-2-yl) amino) phenyl) carbamic acid tert-butyl ester derivative
CN114835640B (en) * 2022-05-24 2024-02-13 中国药科大学 Fibroblast growth factor receptor inhibitor, preparation method and application

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7189715B2 (en) 2002-10-24 2007-03-13 Sepracor Inc. Compositions comprising zopiclone derivatives and methods of making and using the same
WO2011071821A1 (en) * 2009-12-07 2011-06-16 Novartis Ag Crystalline forms of 3-(2,6-dichloro-3,5-dimethoxy-phenyl)-1-{6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimidin-4-yl}-1-methyl-urea and salts thereof.
WO2014149164A1 (en) * 2013-03-15 2014-09-25 Celgene Avilomics Research, Inc. Mk2 inhibitors and uses thereof

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3881320D1 (en) * 1987-09-28 1993-07-01 Ciba Geigy Ag PEST CONTROL.
ID27589A (en) 1998-05-26 2001-04-12 Warner Lambert Comapny BICYCLIC PYYRIMINE AND DIHYDROPIRYMIDINE BICYCLIC 3,4 AS A RESULTS OF MOBILE PROLIFERATION
GB0103926D0 (en) 2001-02-17 2001-04-04 Astrazeneca Ab Chemical compounds
MXPA06001098A (en) 2003-07-29 2006-04-24 Irm Llc Compounds and compositions as protein kinase inhibitors.
GB0512324D0 (en) * 2005-06-16 2005-07-27 Novartis Ag Organic compounds
WO2006038112A1 (en) 2004-10-01 2006-04-13 Warner-Lambert Company Llc Use of kinase inhibitors to promote neochondrogenesis
KR101406956B1 (en) 2005-12-21 2014-06-13 노파르티스 아게 Pyrimidinyl aryl urea derivatives being fgf inhibitors
EP1918376A1 (en) 2006-11-03 2008-05-07 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. FGFR4 promotes cancer cell resistance in response to chemotherapeutic drugs
CA2702647C (en) * 2007-01-31 2016-03-22 Ym Biosciences Australia Pty Ltd Thiopyrimidine-based compounds and uses thereof
US20090062320A1 (en) 2007-08-28 2009-03-05 Vito Guagnano Method of Treating Disorders Mediated by the Fibroblast Growth Factor Receptor
ES2711249T3 (en) 2008-06-27 2019-04-30 Celgene Car Llc Heteroaryl compounds and uses thereof
EP2519517B1 (en) 2009-12-29 2015-03-25 Dana-Farber Cancer Institute, Inc. Type ii raf kinase inhibitors
AU2011205297A1 (en) 2010-01-14 2012-06-28 Yale University Inhibitors of receptor tyrosine kinases (RTK) and methods of use thereof
WO2011153553A2 (en) 2010-06-04 2011-12-08 The Regents Of The University Of California Methods and compositions for kinase inhibition
WO2012136732A1 (en) 2011-04-08 2012-10-11 Ab Science Treatment of multiple myeloma with masitinib
CN102816162B (en) 2011-06-10 2016-04-27 中国科学院广州生物医药与健康研究院 Pyrimido-pyrimidine ketone compounds and medicinal compositions thereof and application
RU2014133547A (en) 2012-01-18 2016-03-10 Дженентек, Инк. WAYS OF APPLICATION OF FGF19 MODULATORS
CN104144915B (en) 2012-02-28 2016-08-24 安斯泰来制药有限公司 Nitrogenous aromatic heterocyclic compounds
DK2872491T3 (en) 2012-07-11 2021-08-09 Blueprint Medicines Corp Inhibitors of the fibroblast growth factor receptor
CN111793068A (en) * 2013-03-15 2020-10-20 西建卡尔有限责任公司 Heteroaryl compounds and uses thereof
US9783504B2 (en) 2013-07-09 2017-10-10 Dana-Farber Cancer Institute, Inc. Kinase inhibitors for the treatment of disease
TW201605452A (en) 2013-08-28 2016-02-16 安斯泰來製藥股份有限公司 Pharmaceutical composition containing pyrimidine compound as an active ingredient
TWI597268B (en) * 2013-10-18 2017-09-01 衛材R&D企管股份有限公司 Fgfr4 inhibitors
JP6458023B2 (en) 2013-10-25 2019-01-23 ブループリント メディシンズ コーポレイション Inhibitors of fibroblast growth factor receptor
US9695165B2 (en) 2014-01-15 2017-07-04 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
CN108884097B (en) 2016-05-20 2021-05-28 浙江海正药业股份有限公司 Pyrimidine derivatives, preparation method thereof and application thereof in medicine
JP7103952B2 (en) 2016-05-20 2022-07-20 江▲蘇▼豪森▲薬▼▲業▼集▲団▼有限公司 FGFR4 inhibitor, its manufacturing method and application

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7189715B2 (en) 2002-10-24 2007-03-13 Sepracor Inc. Compositions comprising zopiclone derivatives and methods of making and using the same
WO2011071821A1 (en) * 2009-12-07 2011-06-16 Novartis Ag Crystalline forms of 3-(2,6-dichloro-3,5-dimethoxy-phenyl)-1-{6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimidin-4-yl}-1-methyl-urea and salts thereof.
WO2014149164A1 (en) * 2013-03-15 2014-09-25 Celgene Avilomics Research, Inc. Mk2 inhibitors and uses thereof

Non-Patent Citations (24)

* Cited by examiner, † Cited by third party
Title
ANASTASSIADIS ET AL.: "Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity", NAT BIOTECHNOL, vol. 29, 2011, pages 1039 - 1045, XP055035782, DOI: doi:10.1038/nbt.2017
BARDERAS ET AL., J PROTEOMICS, vol. 75, 2012, pages 4647 - 4655
BERGE ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1
BOYD ET AL.: "Data Display and Analysis Strategies far the NCI Disease-oriented In Vitro Antitumor Drug Screen", CYTOTOXIC ANTICANCER DRUGS: MODELS AND CONCEPTS FOR DRUG DISCOVERY AND DEVELOPMENT, 1990, pages 11 - 34
CHIANG ET AL., CANCER RES, vol. 68, no. 16, 2008, pages 6779 - 6788
DIECI ET AL., CANCER DISCOVERY, 2013, pages OF1 - OF16
FAWDAR ET AL., PNAS, vol. 110, no. 30, 2013, pages 12426 - 12431
FRENCH ET AL., PLOS ONE, vol. 7, no. 5, 2012, pages E367313
GUAGNANO V ET AL: "Discovery of 3-(2,6-Dichloro-3,5-dimethoxy-phenyl)-1-{6-[4-(4-ethyl-piperazin-1-yl)-phenylamino]-pyrimidin-4-yl}-1-methyl-urea (NVP-BGJ398), A Potent and Selective Inhibitor of the Fibroblast Growth Factor Receptor Family of Receptor Tyrosine Kinase", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 54, no. 20, 27 October 2011 (2011-10-27), pages 7066 - 7083, XP002689272, ISSN: 0022-2623, [retrieved on 20110921], DOI: 10.1021/JM2006222 *
MIURA ET AL., BMC CANCER, vol. 12, 2012, pages 56
MONKS ET AL.: "Feasibility of a High-Flux Anticancer Drug Screen Using a Diverse Panel of Cultured Human Tumor Cell Lines", J NATL CANCER INST, vol. 83, no. 11, 1991, pages 757 - 766, XP002951637, DOI: doi:10.1093/jnci/83.11.757
MOTODA ET AL., INT'L J ONCOL, vol. 38, 2011, pages 133 - 143
OLECHNO ET AL.: "Improving IC results with acoustic droplet ejection", JALA, vol. 11, 2006, pages 240 - 246, XP024969892, DOI: doi:10.1016/j.jala.2006.06.007
PELAEZ-GARCIA ET AL., PLOS ONE, vol. 8, no. 5, 2013, pages E63695
SAWEY ET AL., CANCER CELL, vol. 19, 2011, pages 347 - 358
SAWEY ET AL., CANCER CELL, vol. 19, no. 3, 2011, pages 347 - 358
SIA ET, GASTROENTEROLOGY, vol. 144, 2013, pages 829 - 840
STAHL ET AL.: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH; VERLAG HELVETICA CHIMICA ACTA
STREIT ET AL., BRITISH J CANCER, vol. 94, 2006, pages 1879 - 1886
TAYLOR VI ET AL., J CLIN INVEST, vol. 119, no. 11, 2009, pages 3395 - 3407
WESCHE ET AL., BIOCHEM J, vol. 437, 2011, pages 199 - 213
XU ET AL., BMC CANCER, vol. 11, 2011, pages 84
YE ET AL., CANCER, 2011, pages 5304 - 5313
ZAID ET AL., CLIN CANCER RES, vol. 19, 2013, pages 809 - 820

Cited By (107)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10813930B2 (en) 2010-12-22 2020-10-27 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US11053246B2 (en) 2012-06-13 2021-07-06 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US10131667B2 (en) 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US11840534B2 (en) 2012-06-13 2023-12-12 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US10196436B2 (en) 2012-07-11 2019-02-05 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9321786B2 (en) 2013-03-15 2016-04-26 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US10774052B2 (en) 2013-03-15 2020-09-15 Celgene Car Llc Heteroaryl compounds and uses thereof
US10618902B2 (en) 2013-03-15 2020-04-14 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US9695132B2 (en) 2013-03-15 2017-07-04 Celgene Car Llc Heteroaryl compounds and uses thereof
US9663524B2 (en) 2013-03-15 2017-05-30 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as protein kinase inhibitors
US10189794B2 (en) 2013-03-15 2019-01-29 Celgene Car Llc Heteroaryl compounds and uses thereof
US10065966B2 (en) 2013-03-15 2018-09-04 Celgene Car Llc Substituted pyrido[2,3-d]pyrimidines as inhibitors of protein kinases
US10450313B2 (en) 2013-04-19 2019-10-22 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US11530214B2 (en) 2013-04-19 2022-12-20 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10947230B2 (en) 2013-04-19 2021-03-16 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US9730931B2 (en) 2013-10-18 2017-08-15 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US10537571B2 (en) 2013-10-18 2020-01-21 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US10912774B2 (en) 2013-10-18 2021-02-09 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US9434697B2 (en) 2013-10-18 2016-09-06 Eisai R&D Management Co., Ltd. Pyrimidine FGFR4 inhibitors
US10221154B2 (en) 2013-10-25 2019-03-05 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10875837B2 (en) 2013-10-25 2020-12-29 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10000490B2 (en) 2014-01-15 2018-06-19 Blueprint Medicines Corporation Inhibitors of the fibroblast growth factor receptor
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11667635B2 (en) 2015-02-20 2023-06-06 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11014923B2 (en) 2015-02-20 2021-05-25 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10632126B2 (en) 2015-02-20 2020-04-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11173162B2 (en) 2015-02-20 2021-11-16 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10016438B2 (en) 2015-02-20 2018-07-10 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10251892B2 (en) 2015-02-20 2019-04-09 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10738048B2 (en) 2015-02-20 2020-08-11 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214528B2 (en) 2015-02-20 2019-02-26 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
WO2016164703A1 (en) * 2015-04-09 2016-10-13 Eisai R & D Management Co., Ltd. Fgfr4 inhibitors
JP2018188465A (en) * 2015-04-14 2018-11-29 エーザイ・アール・アンド・ディー・マネジメント株式会社 Crystalline fgfr4 inhibitor compound and uses thereof
US11498916B2 (en) 2015-04-14 2022-11-15 Eisai R&D Management Co., Ltd. Crystalline FGFR4 inhibitor compound and uses thereof
KR20180003559A (en) * 2015-04-14 2018-01-09 에자이 알앤드디 매니지먼트 가부시키가이샤 Crystalline FGFR4 inhibitor compounds and uses thereof
CN107660200A (en) * 2015-04-14 2018-02-02 卫材R&D管理有限公司 Crystallize FGFR4 inhibitor compounds and its purposes
CN107660200B (en) * 2015-04-14 2022-01-11 卫材R&D管理有限公司 Crystalline FGFR4 inhibitor compounds and uses thereof
AU2016248056B2 (en) * 2015-04-14 2020-07-23 Eisai R&D Management Co., Ltd. Crystalline FGFR4 inhibitor compound and uses thereof
US20180093972A1 (en) * 2015-04-14 2018-04-05 Eisai R&D Management Co., Ltd. Crystalline fgfr4 inhibitor compound and uses thereof
KR102548229B1 (en) 2015-04-14 2023-06-27 에자이 알앤드디 매니지먼트 가부시키가이샤 Crystalline FGFR4 inhibitor compounds and uses thereof
US10562888B2 (en) 2015-04-14 2020-02-18 Eisai R&D Management Co., Ltd. Crystalline FGFR4 inhibitor compound and uses thereof
WO2016168331A1 (en) * 2015-04-14 2016-10-20 Eisai R&D Management Co., Ltd. Crystalline fgfr4 inhibitor compound and uses thereof
JP2018511635A (en) * 2015-04-14 2018-04-26 エーザイ・アール・アンド・ディー・マネジメント株式会社 Crystalline FGFR4 inhibitor compounds and uses thereof
US10550112B2 (en) 2015-07-20 2020-02-04 Dana-Farber Cancer Institute, Inc. Pyrimidines as EGFR inhibitors and methods of treating disorders
WO2017015363A1 (en) * 2015-07-20 2017-01-26 Dana-Farber Cancer Institute, Inc. Novel pyrimidines as egfr inhibitors and methods of treating disorders
JP2018527321A (en) * 2015-07-20 2018-09-20 タイペイ・メディカル・ユニバーシティTaipei Medical University Azaaryl compounds substituted with chlorobenzene
EP3324970A4 (en) * 2015-07-20 2019-03-06 Taipei Medical University Chlorobenzene substituted azaaryl compounds
US10624897B2 (en) 2015-07-20 2020-04-21 Taipei Medical University Chlorobenzene substituted azaaryl compounds
US10934311B2 (en) 2016-01-08 2021-03-02 Hangzhou Innogate Pharma Co., Ltd. Heterocyclic compound used as FGFR inhibitor
WO2017118438A1 (en) * 2016-01-08 2017-07-13 杭州英创医药科技有限公司 Heterocyclic compound used as fgfr inhibitor
US20190117640A1 (en) * 2016-05-10 2019-04-25 Eisai R&D Management Co., Ltd. Drug combinations for reducing cell viability and/or cell proliferation
US11357769B2 (en) 2016-05-10 2022-06-14 Eisai R&D Management Co., Ltd. Drug combinations for reducing cell viability and/or cell proliferation
JP2019515010A (en) * 2016-05-10 2019-06-06 エーザイ・アール・アンド・ディー・マネジメント株式会社 Drug combinations to reduce cell viability and / or cell proliferation
WO2017196854A1 (en) * 2016-05-10 2017-11-16 Eisai R&D Management Co., Ltd. Drug combinations for reducing cell viability and/or cell proliferation
US11827625B2 (en) 2016-05-20 2023-11-28 Zhejiang Hisun Pharmaceutical Co., Ltd. Pyrimidine derivative, method for preparing same and use thereof in medicine
US10654836B2 (en) 2016-05-20 2020-05-19 Zhejiang Hisun Pharmaceutical Co., Ltd. Pyrimidine derivative, method for preparing same and use thereof in medicine
CN108884097B (en) * 2016-05-20 2021-05-28 浙江海正药业股份有限公司 Pyrimidine derivatives, preparation method thereof and application thereof in medicine
WO2017198221A1 (en) 2016-05-20 2017-11-23 浙江海正药业股份有限公司 Pyrimidine derivative, method for preparing same and use thereof in medicine
CN108884097A (en) * 2016-05-20 2018-11-23 浙江海正药业股份有限公司 Pyridine derivatives, preparation method and its purposes in medicine
JP2019519512A (en) * 2016-05-20 2019-07-11 浙江海正薬業股▲ふん▼有限公司Zhejiang Hisun Pharmaceutical CO.,LTD. Pyrimidine derivatives, process for their preparation and use in medicine
TWI669300B (en) * 2016-05-20 2019-08-21 浙江海正藥業股份有限公司 Pyrimidine derivatives, its preparation method, its pharmaceutical composition and its use in medicine
CN107400092A (en) * 2016-05-20 2017-11-28 浙江海正药业股份有限公司 Pyridine derivatives and preparation method thereof and its purposes in medicine
US11001572B2 (en) 2016-05-20 2021-05-11 Zhejiang Hisun Pharmaceutical Co., Ltd. Pyrimidine derivative, method for preparing same and use thereof in medicine
EP3459952A4 (en) * 2016-05-20 2020-01-15 Zhejiang Hisun Pharmaceutical Co., Ltd. Pyrimidine derivative, method for preparing same and use thereof in medicine
CN105884760B (en) * 2016-06-13 2019-01-04 厦门市蔚嘉化学科技有限公司 A kind of preparation method than his intermediate of department
RU2769251C2 (en) * 2016-08-23 2022-03-29 Эйсай Ар Энд Ди Менеджмент Ко., Лтд. Combination therapy for hepatocellular carcinoma
JP7190425B2 (en) 2016-08-23 2022-12-15 エーザイ・アール・アンド・ディー・マネジメント株式会社 Combination therapy for the treatment of hepatocellular carcinoma
CN109803684B (en) * 2016-08-23 2022-08-23 卫材 R&D 管理有限公司 Combination therapy for the treatment of hepatocellular carcinoma
AU2017315357B2 (en) * 2016-08-23 2022-12-01 Eisai R&D Management Co., Ltd. Combination therapies for the treatment of hepatocellular carcinoma
JP2019528302A (en) * 2016-08-23 2019-10-10 エーザイ・アール・アンド・ディー・マネジメント株式会社 Combination therapy for the treatment of hepatocellular carcinoma
CN109803684A (en) * 2016-08-23 2019-05-24 卫材 R&D 管理有限公司 For treating the combination treatment of hepatocellular carcinoma
WO2018039324A1 (en) 2016-08-23 2018-03-01 Eisai R&D Management Co., Ltd. Combination therapies for the treatment of hepatocellular carcinoma
IL264950B1 (en) * 2016-08-23 2023-09-01 Eisai R&D Man Co Ltd Combination therapies for the treatment of hepatocellular carcinoma
WO2018049233A1 (en) 2016-09-08 2018-03-15 Nicolas Stransky Inhibitors of the fibroblast growth factor receptor in combination with cyclin-dependent kinase inhibitors
WO2018055503A1 (en) 2016-09-20 2018-03-29 Novartis Ag Combination comprising a pd-1 antagonist and an fgfr4 inhibitor
WO2018083603A1 (en) 2016-11-02 2018-05-11 Novartis Ag Combinations of fgfr4 inhibitors and bile acid sequestrants
US11008292B2 (en) 2016-11-17 2021-05-18 Guangdong Zhongsheng Pharmaceutical Co., Ltd FGFR4 inhibitor and preparation method and use thereof
CN108503593A (en) * 2017-02-28 2018-09-07 暨南大学 2- amino-metadiazine compounds and its application
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11472801B2 (en) 2017-05-26 2022-10-18 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
CN109422760A (en) * 2017-09-01 2019-03-05 南京圣和药业股份有限公司 FGFR4 inhibitor and its application
CN109422760B (en) * 2017-09-01 2022-05-27 南京圣和药物研发有限公司 FGFR4 inhibitor and application thereof
WO2019047826A1 (en) 2017-09-05 2019-03-14 博奥阿迪斯生物科技公司 Aromatic derivative, preparation method for same, and medical applications thereof
US11279697B2 (en) 2017-09-05 2022-03-22 Bioardis Llc Aromatic derivative, preparation method for same, and medical applications thereof
US11833119B2 (en) 2018-01-10 2023-12-05 Eisai R&D Management Co., Ltd. Combination therapies for the treatment of hepatocellular carcinoma
WO2019139977A1 (en) 2018-01-10 2019-07-18 Eisai R&D Management Co., Ltd. Combination therapies for the treatment of hepatocellular carcinoma
RU2787993C2 (en) * 2018-01-10 2023-01-16 ЭЙСАЙ Р энд Д МЭНЭДЖМЕНТ КО., ЛТД. (EISAI R&D MANAGEMENT CO., LTD.) Combination therapies for treatment of hepatocellular carcinoma
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11976058B2 (en) 2019-03-05 2024-05-07 Bioardis Llc Aromatic derivatives, preparation methods, and medical uses thereof
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
WO2021129561A1 (en) 2019-12-23 2021-07-01 北京赛特明强医药科技有限公司 Cyano-substituted pyridine and cyano-substituted pyrimidine compound and preparation method therefor and application thereof
WO2022075482A1 (en) 2020-10-05 2022-04-14 株式会社カイオム・バイオサイエンス Medicine for treating cancer
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
WO2022262577A1 (en) * 2021-06-17 2022-12-22 浙江海正药业股份有限公司 Salt of selective fgfr4 inhibitor, and preparation method therefor and application thereof
WO2022268063A1 (en) 2021-06-23 2022-12-29 浙江海正药业股份有限公司 Crystal form of pyrimidine derivative and preparation method therefor

Also Published As

Publication number Publication date
AU2014337291B9 (en) 2020-05-07
AR098048A1 (en) 2016-04-27
NZ717559A (en) 2021-07-30
IL244373A0 (en) 2016-04-21
RS57444B1 (en) 2018-09-28
CN105899490A (en) 2016-08-24
RU2715708C2 (en) 2020-03-03
CY1121129T1 (en) 2019-12-11
CA2924206C (en) 2022-10-25
IL244373B (en) 2018-10-31
TWI597268B (en) 2017-09-01
US9730931B2 (en) 2017-08-15
JO3515B1 (en) 2020-07-05
HRP20181129T1 (en) 2018-09-21
AU2014337291A1 (en) 2016-03-17
EP3421457B1 (en) 2021-10-06
KR101826015B1 (en) 2018-02-06
US9434697B2 (en) 2016-09-06
KR20160072113A (en) 2016-06-22
DK3057943T3 (en) 2018-07-30
MY184733A (en) 2021-04-20
ES2679521T3 (en) 2018-08-28
JP2017160233A (en) 2017-09-14
US10912774B2 (en) 2021-02-09
BR112016008110B1 (en) 2019-07-02
PL3057943T3 (en) 2018-09-28
JP6139788B2 (en) 2017-05-31
AU2018271284A1 (en) 2018-12-20
EP3057943A1 (en) 2016-08-24
RU2016118981A3 (en) 2018-06-25
PT3057943T (en) 2018-07-24
PH12016500676A1 (en) 2016-05-30
CN105899490B (en) 2019-07-23
US10537571B2 (en) 2020-01-21
PE20160679A1 (en) 2016-08-04
HUE039268T2 (en) 2018-12-28
CA2924206A1 (en) 2015-04-23
UA116920C2 (en) 2018-05-25
EP3057943B1 (en) 2018-04-25
CL2016000918A1 (en) 2016-11-11
HK1223091A1 (en) 2017-07-21
TW201605802A (en) 2016-02-16
SI3057943T1 (en) 2018-08-31
SG11201602069WA (en) 2016-04-28
AU2014337291B2 (en) 2018-12-13
US20170007601A1 (en) 2017-01-12
WO2015057963A1 (en) 2015-04-23
US20200206222A1 (en) 2020-07-02
RU2016118981A (en) 2017-11-23
LT3057943T (en) 2018-11-12
CN110354128A (en) 2019-10-22
MX2016004933A (en) 2016-12-20
AU2018271284B2 (en) 2020-05-28
JP2016535000A (en) 2016-11-10
US20160130237A1 (en) 2016-05-12
US20170360785A1 (en) 2017-12-21
EP3421457A1 (en) 2019-01-02

Similar Documents

Publication Publication Date Title
AU2018271284B2 (en) Pyrimidine FGFR4 inhibitors
ES2879474T3 (en) Compounds and compositions to modulate the activities of mutant EGFR kinase
EP3173412B1 (en) 2,4-disubstituted 7h-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medicinal use thereof
US11498916B2 (en) Crystalline FGFR4 inhibitor compound and uses thereof
WO2016164703A1 (en) Fgfr4 inhibitors
JP7036939B2 (en) A novel pyrimidine derivative showing a growth inhibitory effect on cancer cells and a pharmaceutical composition containing the same.
WO2016164754A1 (en) Fgfr4 inhibitors

Legal Events

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

Ref document number: 14790956

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 244373

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2924206

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2014337291

Country of ref document: AU

Date of ref document: 20141016

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 000460-2016

Country of ref document: PE

ENP Entry into the national phase

Ref document number: 20167009625

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12016500676

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2016523266

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: IDP00201602520

Country of ref document: ID

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/004933

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 16097757

Country of ref document: CO

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016008110

Country of ref document: BR

REEP Request for entry into the european phase

Ref document number: 2014790956

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014790956

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: A201605322

Country of ref document: UA

ENP Entry into the national phase

Ref document number: 2016118981

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016008110

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20160412