WO2018049781A1 - 炔代杂环化合物、其制备方法及其在医药学上的应用 - Google Patents

炔代杂环化合物、其制备方法及其在医药学上的应用 Download PDF

Info

Publication number
WO2018049781A1
WO2018049781A1 PCT/CN2017/072570 CN2017072570W WO2018049781A1 WO 2018049781 A1 WO2018049781 A1 WO 2018049781A1 CN 2017072570 W CN2017072570 W CN 2017072570W WO 2018049781 A1 WO2018049781 A1 WO 2018049781A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
group
cycloalkyl
cyano
ethynyl
Prior art date
Application number
PCT/CN2017/072570
Other languages
English (en)
French (fr)
Inventor
陈向阳
高英祥
孔祥龙
Original Assignee
北京天诚医药科技有限公司
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 MX2019003143A priority Critical patent/MX2019003143A/es
Application filed by 北京天诚医药科技有限公司 filed Critical 北京天诚医药科技有限公司
Priority to CA3036594A priority patent/CA3036594C/en
Priority to EP17849992.7A priority patent/EP3517535B1/en
Priority to CN201780057036.5A priority patent/CN109843873B/zh
Priority to AU2017327954A priority patent/AU2017327954B2/en
Priority to ES17849992T priority patent/ES2925212T3/es
Priority to RU2019112046A priority patent/RU2729069C1/ru
Priority to JP2019515464A priority patent/JP6906811B2/ja
Priority to KR1020197011454A priority patent/KR20190052113A/ko
Publication of WO2018049781A1 publication Critical patent/WO2018049781A1/zh
Priority to US16/353,806 priority patent/US10710981B2/en
Priority to US16/925,738 priority patent/US11059805B2/en
Priority to US17/369,850 priority patent/US11572353B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41551,2-Diazoles non 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three 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, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three 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, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/44Oxygen and nitrogen or sulfur and 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
    • C07D401/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • 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
    • C07D403/06Heterocyclic 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 carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings

Definitions

  • the present invention relates to a novel acetylene heterocyclic compound as a FGFR inhibitor or a pharmaceutically acceptable salt thereof; a pharmaceutical composition comprising the acetylene heterocyclic compound or a pharmaceutically acceptable salt thereof; a method for producing a cyclic compound or a pharmaceutically acceptable salt thereof; the acetylene heterocyclic compound or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the acetylene heterocyclic compound or a pharmaceutically acceptable salt thereof, in the preparation Use in a medicament for the treatment and/or prevention of a FGFR-related disorder, in particular a tumor; and a method of using the compound or composition to treat and/or prevent a FGFR-related disorder, in particular a tumor.
  • Fibroblast Growth Factor Receptor is a class of receptor tyrosine kinases (RTKs) structurally composed of extramembranous ligand binding domains, a single transmembrane domain, and intramembrane tyrosine kinases.
  • the domain consists of four subtypes of FGFR1, FGFR2, FGFR3 and FGFR4. It and its ligand, Fibroblast Growth Factor (FGF) play an important regulatory role in cell signaling.
  • FGF As an extracellular stimulatory signal, FGF binds to the extracellular domain of FGFR, causing phosphorylation of its intramembrane tyrosine kinase, thereby activating a series of downstream signaling pathways that regulate cell proliferation, differentiation and metastasis.
  • a variety of tumors are closely related to FGF/FGFR expression and activation, such as non-small cell lung cancer, breast cancer, gastric cancer, liver cancer, bladder cancer, endometrial cancer, prostate cancer, cervical cancer, colon cancer, esophageal cancer, myeloma and melanin. Tumors, etc. (Clin. Cancer Res. 2012, 18, 1855). Studies have shown that FGFR1 amplification accounts for 20% of non-small cell lung cancer, FGFR2 amplification accounts for ⁇ 5% of gastric cancer, FGFR3 mutation accounts for ⁇ 70% of non-invasive bladder cancer, and FGFR4 is expanded in liver cancer (PloS One 2012). , 7, e36713). Therefore, the development of inhibitors targeting FGFR has become a hot topic in anti-tumor drug research (Drug Disc. Today 2014, 19, 51).
  • FGFR-specific drugs such as sunitinib from Pfizer, lenvatini from Eisai, and nintedanib from Boehringer Ingelheim, but no FGFR-specific inhibitors.
  • Specific FGFR inhibitors that enter the clinic include HMPL-453, BGJ-398, LY-2874455, AZ-4547, JNJ-42756493, TAS-120, ARQ-087, and BLU-554.
  • the present invention has designed a compound having a structure represented by the general formula (I), and it has been found that a compound having such a structure exhibits an excellent effect and effect.
  • the present invention provides a compound of the formula (I), a prodrug thereof, a stable isotope derivative, a pharmaceutically acceptable salt, an isomer, and a mixture thereof as an FGFR inhibitor:
  • A is N or CR 2 ;
  • Ring B is a benzene ring or a 5-6 membered heteroaryl ring, wherein the benzene ring and the heteroaryl ring are optionally substituted by one or more G 1 ;
  • R 1 is independently selected from H, halogen, cyano, C 1-6 alkyl or -NHR 3 ;
  • R 2 is independently selected from H, halo, cyano or C 1-6 alkyl, wherein said alkyl is optionally substituted by halogen, cyano, hydroxy or -OC 1-6 alkyl;
  • R 3 is independently selected from H, C 1-6 alkyl, C 3-6 cycloalkyl or 3-6 membered heterocyclyl, wherein the alkyl, cycloalkyl and heterocyclyl are optionally halogen, cyanide Substituted with -OR 4 , -NR 5 R 6 , C 1-6 alkyl, C 3-6 cycloalkyl or 3-6 membered heterocyclyl;
  • X is absent or is a C 1-6 alkylene group
  • Y is absent or selected from a C 3-8 cycloalkylene group, a 3-8 membered heterocyclylene group, an arylene group or a heteroarylene group, wherein the cycloalkylene group, the heterocyclylene group, the arylene group and The heteroarylene is optionally substituted with one or more G 2 ;
  • Z is independently selected from cyano, -NR 7 CN,
  • the key a is a double bond or a triple bond
  • R a , R b and R c are each independently selected from H, cyano, halogen, C 1-6 alkyl, C 3-6 cycloalkyl or 3-6 membered heterocyclic group. Wherein said alkyl, cycloalkyl and heterocyclic group are optionally substituted by one or more G 3 ;
  • R a and R b or R b and R c are optionally taken together with the carbon atom to which they are attached to form a 3-6 membered ring optionally containing a hetero atom;
  • R a and R c are absent, and R b is independently selected from H, cyano, halogen, C 1-6 alkyl, C 3-6 cycloalkyl or 3-6 membered heterocyclic ring. a group wherein the alkyl group, cycloalkyl group and heterocyclic group are optionally substituted by one or more G 4 ;
  • R 4 is independently selected from H, C 1-6 alkyl, C 3-6 cycloalkyl or 3-6 membered heterocyclyl, wherein the alkyl, cycloalkyl and heterocyclyl are optionally selected by one or Replaced by multiple G 5 ;
  • G 1 , G 2 , G 3 , G 4 and G 5 are each independently selected from the group consisting of halogen, cyano, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 ring Alkyl, 3-8 membered heterocyclic, C 6-10 aryl, 5-10 membered heteroaryl, -OR 8 , -OC(O)NR 8 R 9 , -C(O)OR 8 , -C (O)NR 8 R 9 , -C(O)R 8 , -NR 8 R 9 , -NR 8 C(O)R 9 , -NR 8 C(O)NR 9 R 10 , -S(O) m R 8 or -NR 8 S(O) m R 9 , wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, aryl and heteroaryl groups are optionally selected from one or more selected from halogen
  • R 4 , R 5 , R 6 , R 8 , R 9 , R 10 , R 11 , R 12 and R 13 are each independently selected from H, C 1-6 alkyl, C 3-8 cycloalkyl, 3- 8-membered monocyclic heterocyclic group, monocyclic heteroaryl or phenyl;
  • n 1 or 2.
  • One embodiment of the present invention relates to a compound represented by the above formula (I) or a prodrug thereof, a stable isotope derivative, a pharmaceutically acceptable salt, an isomer thereof and a mixture thereof, wherein A is N or CH, preferably N.
  • Another embodiment of the present invention relates to the compound of the above formula (I) or a prodrug thereof, a stable isotope derivative, a pharmaceutically acceptable salt, an isomer thereof and a mixture thereof, wherein Ring B is a benzene ring.
  • the invention provides a compound of formula (II) below, or a prodrug thereof, a stable isotope derivative, a pharmaceutically acceptable salt, an isomer, and mixtures thereof:
  • G a , G b , G c and G d are each independently selected from H, halogen, cyano, C 1-6 alkyl, C 3-8 cycloalkyl, 3-8 membered heterocyclic, -OR 8 , -NR 8 R 9 or -C(O)NR 8 R 9 , wherein the alkyl group, cycloalkyl group and heterocyclic group are optionally one or more selected from the group consisting of halogen, cyano, C 1-6 alkyl, Substituted by a C 3-8 cycloalkyl, 3-8 membered heterocyclyl, -OR 11 or -NR 11 R 12 substituent; wherein A, R 1 , R 8 , R 9 , R 11 , R 12 , X
  • Y, Z are as described above.
  • Another embodiment of the present invention relates to a compound represented by the above formula (I) or a prodrug thereof, a stable isotope derivative, a pharmaceutically acceptable salt, an isomer thereof, and a mixture thereof, which is a compound of the formula (III) a compound or a prodrug thereof, a stable isotope derivative, a pharmaceutically acceptable salt, an isomer, and a mixture thereof:
  • G a and G b are each independently selected from H, halogen, cyano, C 1-6 alkyl, C 3-8 cycloalkyl, 3-8 membered heterocyclyl, -OR 8 , -NR 8 R 9 or -C(O)NR 8 R 9 wherein the alkyl, cycloalkyl and heterocyclic groups are optionally selected from one or more selected from the group consisting of halogen, cyano, C 1-6 alkyl, C 3-8 naphthenic Substituted by a substituent of a 3-8 membered heterocyclic group, -OR 11 or -NR 11 R 12 ; wherein A, R 1 , R 8 , R 9 , R 11 , R 12 , X, Y, Z are defined As mentioned earlier.
  • Another embodiment of the present invention relates to the compound of the above formula (I) or a prodrug thereof, a stable isotopic derivative, a pharmaceutically acceptable salt, an isomer thereof and a mixture thereof, wherein R 1 is independently selected from H , -NH 2 or -NHC 1-6 alkyl.
  • R 1 may be H or -NH 2 .
  • G a , G b , G c and G d may each independently be —OC 1-2 alkyl or halogen.
  • R 1 is independently selected from H, -NH 2 or -NHR 3 ;
  • R 3 is independently selected from C 1-6 alkyl, C 3-6 cycloalkyl or 3-6 membered heterocyclic ring a group wherein the alkyl group, cycloalkyl group and heterocyclic group are optionally halogen, cyano, -OR 4 , -NR 5 R 6 , C 1-6 alkyl, C 3-6 cycloalkyl or 3- Substituted by a 6-membered heterocyclic group.
  • Another embodiment of the present invention relates to a compound represented by the above formula (I), (II) and (III) or a prodrug thereof, a stable isotope derivative, a pharmaceutically acceptable salt, an isomer and a mixture thereof, among them:
  • X is absent or is a C 1-6 alkylene group
  • Y is absent or selected from a C 3-8 cycloalkylene group or a 3-8 membered heterocyclylene group;
  • Z is independently selected from cyano, -NR 7 CN,
  • the key a is a double bond or a triple bond
  • R a , R b and R c are each independently selected from H, cyano, halogen, C 1-6 alkyl, C 3-6 cycloalkyl or 3-6 membered heterocyclic group.
  • the alkyl, cycloalkyl and heterocyclic groups are optionally one or more independently selected from the group consisting of halogen, cyano, C 1-6 alkyl, C 3-6 cycloalkyl, 3-6-membered Substituted by a ring group, a substituent of -OR 8 or -NR 8 R 9 ;
  • R a and R c are absent, and R b is independently selected from H, cyano, halogen, C 1-6 alkyl, C 3-6 cycloalkyl or 3-6 membered heterocyclic ring.
  • R b is independently selected from H, cyano, halogen, C 1-6 alkyl, C 3-6 cycloalkyl or 3-6 membered heterocyclic ring.
  • a group wherein the alkyl group, cycloalkyl group and heterocyclic group are optionally one or more independently selected from the group consisting of halogen, cyano, C 1-6 alkyl, C 3-6 cycloalkyl, 3-6 Substituted by a heterocyclic group, a substituent of -OR 8 or -NR 8 R 9 ;
  • R 4 , R 8 and R 9 are each independently selected from H or C 1-6 alkyl.
  • One embodiment of the invention relates to the compound of the above formula (I), wherein the compound is selected from the group consisting of:
  • a prodrug thereof a stable isotope derivative, a pharmaceutically acceptable salt, an isomer, and mixtures thereof.
  • the compounds of the invention have a significant inhibitory effect on the activity of FGFR.
  • the compounds of this invention effective to inhibit FGFR1, activity of FGFR2, FGFR3, or FGFR4, and preferably which inhibits FGFR1, FGFR2, FGFR3 or FGFR4 an IC 50 of 100 to 1000 nM, more preferably IC 50 of less than 10OnM, most preferably an IC 50 of less than 10nM.
  • the compounds of the present invention have a significant inhibitory effect on cell proliferation of tumor cells (e.g., Hep3B, RT4 and SNU-16 tumor cells), preferably having an IC 50 of 100 to 1000 nM, more preferably an IC 50 of less than 100 nM, most preferably The IC 50 is less than 10 nM.
  • tumor cells e.g., Hep3B, RT4 and SNU-16 tumor cells
  • the compounds of the invention are therefore useful in the treatment or prevention of FGFR-associated diseases including, but not limited to, tumors and inflammatory diseases such as osteoarthritis.
  • the compounds of the present invention are useful for treating or preventing FGFR-associated tumors, such as non-small cell lung cancer, esophageal cancer, melanoma, rhabdomyosarcoma, renal cell carcinoma, multiple myeloma, breast cancer, ovarian cancer, endometrial cancer, cervical cancer.
  • gastric cancer, colon cancer, bladder cancer, pancreatic cancer, lung cancer, breast cancer, prostate cancer and liver cancer (such as hepatocellular carcinoma), more specifically liver cancer, gastric cancer, non-small cell lung cancer and bladder cancer.
  • the present invention provides a method of treating or preventing a FGFR-mediated disease, such as the tumor, comprising administering to a patient in need thereof a therapeutically effective amount of a compound of the present invention or a prodrug thereof, and stabilizing Isotope derivatives, pharmaceutically acceptable salts, isomers, and mixtures thereof, or pharmaceutical compositions comprising the compounds.
  • Another aspect of the invention relates to a compound of the formula (I) or a prodrug thereof, a stable isotopic derivative, a pharmaceutically acceptable salt, an isomer thereof and a mixture thereof for use as a medicament or for medical use, for treatment or Prevention of FGFR-mediated diseases such as tumors or inflammatory diseases including, but not limited to, non-small cell lung cancer, esophageal cancer, melanoma, rhabdomyosarcoma, renal cell carcinoma, multiple myeloma, breast cancer, ovarian cancer, endometrium Cancer, cervical cancer, stomach cancer, colon cancer, bladder cancer, pancreatic cancer, lung cancer, breast cancer, prostate cancer and liver cancer.
  • diseases such as tumors or inflammatory diseases including, but not limited to, non-small cell lung cancer, esophageal cancer, melanoma, rhabdomyosarcoma, renal cell carcinoma, multiple myeloma, breast cancer, ovarian cancer, endometrium Cancer, cervical cancer, stomach
  • the invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention or a prodrug thereof, a stable isotope derivative, a pharmaceutically acceptable salt, an isomer, and mixtures thereof, and a pharmaceutically acceptable carrier , thinner, excipients.
  • Another aspect of the invention relates to a compound of the formula (I) or a prodrug thereof, a stable isotopic derivative, a pharmaceutically acceptable salt, an isomer thereof, and a mixture thereof, or a pharmaceutical composition for the preparation of a medicament Use, wherein the medicament is for treating or preventing a FGFR mediated disease, such as a tumor and an inflammatory disease.
  • a FGFR mediated disease such as a tumor and an inflammatory disease.
  • the medicament may be in any pharmaceutical dosage form including, but not limited to, tablets, capsules, solutions, lyophilized preparations, injections.
  • the pharmaceutical preparation of the present invention can be administered in the form of a dosage unit containing a predetermined amount of the active ingredient per dosage unit.
  • a dosage unit may comprise, for example, from 0.5 mg to 1 g, preferably from 1 mg to 700 mg, particularly preferably from 5 mg to 300 mg, of a compound of the invention, or a drug, depending on the condition being treated, the method of administration, and the age, weight and condition of the patient.
  • the formulations may be administered in the form of dosage units containing a predetermined amount of active ingredient per dosage unit.
  • Preferred dosage unit formulations are those containing the daily or divided doses indicated above or their corresponding fractions of the active ingredient.
  • pharmaceutical preparations of this type can be prepared using methods well known in the pharmaceutical art.
  • the pharmaceutical preparations of the invention may be adapted for administration by any suitable method desired, for example by oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral. (including subcutaneous, intramuscular, intravenous or intradermal) methods of administration.
  • Such formulations can be prepared by, for example, combining the active ingredient with one or more excipients or one or more adjuvants, using all methods known in the art of pharmacy.
  • C xy denotes a range of the number of carbon atoms, wherein x and y are both integers, for example, C 3-8 cycloalkyl represents a cycloalkyl group having 3 to 8 carbon atoms, that is, having 3 a cycloalkyl group of 4, 5, 6, 7 or 8 carbon atoms. It should also be understood that “C3-8” also encompasses any sub-ranges therein, such as C3-7, C3-6, C4-7, C4-6, C5-6, and the like.
  • Alkyl means a saturated group containing from 1 to 20 carbon atoms, for example from 1 to 18 carbon atoms, from 1 to 12 carbon atoms, from 1 to 8 carbon atoms, from 1 to 6 carbon atoms or from 1 to 4 carbon atoms. a linear or branched hydrocarbyl group.
  • Non-limiting examples of alkyl groups include methyl, ethyl, and positive Propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2 - dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-tri Methylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl and 2-ethylbutyl base.
  • the alkyl group can be substituted or unsubstituted.
  • alkenyl means a straight or branched chain hydrocarbon radical containing at least one carbon to carbon double bond and usually 2 to 20 carbon atoms, for example 2 to 8 carbon atoms, 2 to 6 carbon atoms or 2 to 4 carbon atoms. Group.
  • alkenyl groups include ethenyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2-methyl-2-propenyl, 1 , 4-pentadienyl and 1,4-butadienyl.
  • the alkenyl group can be substituted or unsubstituted.
  • Alkynyl means a straight or branched chain hydrocarbon radical containing at least one carbon to carbon triple bond and typically 2 to 20 carbon atoms, for example 2 to 8 carbon atoms, 2 to 6 carbon atoms or 2 to 4 carbon atoms. Group. Non-limiting examples of alkynyl groups include ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl and 3-butynyl. The alkynyl group can be substituted or unsubstituted.
  • Cycloalkyl means a saturated cyclic hydrocarbyl substituent containing from 3 to 14 carbon ring atoms.
  • the cycloalkyl group can be a single carbon ring and usually contains from 3 to 7 carbon ring atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • the cycloalkyl group may alternatively be a double or tricyclic ring fused together, such as decahydronaphthyl.
  • the cycloalkyl group can be substituted or unsubstituted.
  • Heterocyclic or heterocyclic group refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic group comprising from 3 to 20 ring atoms, for example from 3 to 16, from 3 to 14, from 3 to 12 , 3 to 10, 3 to 8, 3 to 6, or 5 to 6 ring atoms, wherein one or more ring atoms are selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2 ), but does not include the ring portion of -OO-, -OS- or -SS-, and the remaining ring atoms are carbon.
  • ring atoms Preferably, it comprises from 3 to 12 ring atoms, more preferably from 3 to 10 ring atoms, most preferably 5 or 6 ring atoms, wherein from 1 to 4 are heteroatoms, more preferably from 1 to 3 are heteroatoms, most preferably from 1 to Two are heteroatoms.
  • monocyclic heterocyclic groups include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, oxetanyl and azetidine base.
  • Polycyclic heterocyclic groups include fused, bridged or spiro polycyclic heterocyclic groups. The heterocyclic or heterocyclic group may be substituted or unsubstituted.
  • Aryl means an aromatic monocyclic or fused polycyclic group containing from 6 to 14 carbon atoms, preferably from 6 to 10 members, such as phenyl and naphthyl, most preferably phenyl.
  • the aryl ring may be fused to a heteroaryl, heterocyclic or cycloalkyl ring, wherein the ring to which the parent structure is attached is Non-limiting examples of aryl rings include:
  • the aryl group may be substituted or unsubstituted.
  • Heteroaryl or heteroaryl ring refers to a heteroaromatic system containing from 5 to 14 ring atoms, wherein from 1 to 4 ring atoms are selected from heteroatoms including oxygen, sulfur and nitrogen.
  • the heteroaryl group is preferably from 5 to 10 members. More preferably, the heteroaryl group is 5- or 6-membered, such as furyl, thienyl, pyridyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyrazolyl, imidazolyl, tetrazolyl, Oxazolyl, isoxazolyl, thiazolyl, isothiazolyl and the like.
  • the heteroaryl ring may be fused to an aryl, heterocyclic or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heteroaryl ring, non-limiting examples include:
  • the heteroaryl group can be substituted or unsubstituted.
  • Halogen means fluoro, chloro, bromo or iodo.
  • heterocyclic group optionally substituted by an alkyl group means that an alkyl group may be, but not necessarily, present, and the description includes the case where the heterocyclic group is substituted with an alkyl group and the case where the heterocyclic group is not substituted with an alkyl group.
  • Substituted refers to one or more hydrogen atoms in the group, preferably 5, more preferably 1 to 3 hydrogen atoms, independently of each other, substituted by a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and those skilled in the art will be able to determine (by experiment or theory) substitutions that may or may not be possible without undue effort. For example, an amino group or a hydroxyl group having a free hydrogen may be unstable when combined with a carbon atom having an unsaturated (e.g., olefinic) bond.
  • the substituents include, but are not limited to, hydroxyl, amino, halogen, cyano, C 1-6 alkyl, C 1-6 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl, C 3-8 Cycloalkyl and the like.
  • “Pharmaceutical composition” refers to a composition comprising one or more compounds described herein, or a pharmaceutically acceptable salt or prodrug thereof, and other components such as pharmaceutically acceptable carriers and excipients.
  • Drug group The purpose of the composition is to promote the administration of the organism, and to facilitate the absorption of the active ingredient to exert biological activity.
  • isomer refers to a compound having the same molecular formula but differing in the nature or sequence of its atomic bonding or in the spatial arrangement of its atoms, and is referred to as an "isomer.” Isomers whose atomic space is arranged differently are referred to as “stereoisomers”. Stereoisomers include optical isomers, geometric isomers, and conformational isomers.
  • optical isomer form The compounds of the invention may exist in optical isomer form. These optical isomers are in the "R” or “S” configuration depending on the configuration of the substituents around the chiral carbon atom. Optical isomers include enantiomers and diastereomers. Methods of preparing and isolating optical isomers are known in the art.
  • Geometric isomers may also be present in the compounds of the invention.
  • the present invention contemplates various geometric isomers and mixtures thereof resulting from the distribution of substituents around carbon-carbon double bonds, carbon-nitrogen double bonds, cycloalkyl groups or heterocyclic groups.
  • the substituents around the carbon-carbon double bond or carbon-nitrogen bond are designated as the Z or E configuration, and the substituents around the cycloalkyl or heterocycle are designated in the cis or trans configuration.
  • the compounds of the invention may also exhibit tautomerism, such as keto-enol tautomerization.
  • the invention includes any tautomeric or stereoisomeric forms and mixtures thereof, and is not limited to any one of the tautomeric or stereoisomeric forms used in the nomenclature or chemical structural formula of the compound.
  • isotopes are all isotopes of atoms occurring in the compounds of the invention. Isotopes include those atoms having the same atomic number but different mass numbers. Examples of isotopes suitable for incorporation into the compounds of the invention are hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, for example but not limited to 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • Isotopically labeled compounds of the invention can generally be prepared by conventional techniques known to those skilled in the art or by methods analogous to those described in the accompanying examples, using the appropriate isotopically labeled reagents in place of the non-isotopically labeled reagents.
  • Such compounds have a variety of potential uses, for example as a standard and reagent in the determination of biological activity. In the case of stable isotopes, such compounds have the potential to advantageously alter biological, pharmacological or pharmacokinetic properties.
  • Prodrug means that the compound of the invention can be administered in the form of a prodrug.
  • Prodrugs are derivatives which are converted to the biologically active compounds of the invention under physiological conditions in vivo, for example by oxidation, reduction, hydrolysis, etc., each of which is carried out using an enzyme or without the participation of an enzyme.
  • prodrugs are compounds wherein the amino group in the compound of the invention is acylated, alkyl Or phosphorylation, such as eicosylamino, alanylamino, pivaloyloxymethylamino, or wherein the hydroxy group is acylated, alkylated, phosphorylated or converted to a borate, such as an acetoxy group , palmitoyloxy, pivaloyloxy, succinyloxy, fumaryloxy, alanyloxy, or wherein the carboxyl group is esterified or amidated, or wherein the thiol is selectively attached to the target and/or
  • a carrier molecule such as a peptide, that delivers a drug to the cytosol of the cell forms a disulfide bridge.
  • “Pharmaceutically acceptable salt” or “pharmaceutically acceptable salt” refers to a salt made from a pharmaceutically acceptable base or acid, including inorganic bases or acids and organic bases or acids. Where the compounds of the invention contain one or more acidic or basic groups, the invention also includes their corresponding pharmaceutically acceptable salts. Thus, the compounds of the invention containing an acidic group may be present in the form of a salt and may be used according to the invention, for example as an alkali metal salt, an alkaline earth metal salt or as an ammonium salt. More specific examples of such salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as ethylamine, ethanolamine, triethanolamine or amino acids.
  • the compounds of the invention containing a basic group may be present in the form of a salt and may be used in accordance with the invention in the form of their addition salts with inorganic or organic acids.
  • suitable acids include hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, and C.
  • the compound of the invention contains both acidic and basic groups in the molecule, the invention includes, in addition to the salt forms mentioned, internal or internal ammonium salts.
  • Each salt can be obtained by conventional methods known to those skilled in the art, for example by contacting these with an organic or inorganic acid or base in a solvent or dispersant or by anion exchange or cation exchange with other salts.
  • tumor includes benign tumors and malignant tumors (eg, cancer).
  • cancer includes various malignancies in which FGFR is involved, including but not limited to non-small cell lung cancer, esophageal cancer, melanoma, rhabdomyosarcoma, renal cell carcinoma, multiple myeloma, breast cancer, ovary Cancer, endometrial cancer, cervical cancer, stomach cancer, colon cancer, bladder cancer, pancreatic cancer, lung cancer, breast cancer, prostate cancer and liver cancer (eg liver Cell carcinoma), more specifically liver cancer, gastric cancer, non-small cell lung cancer and bladder cancer.
  • non-small cell lung cancer esophageal cancer, melanoma, rhabdomyosarcoma, renal cell carcinoma, multiple myeloma, breast cancer, ovary Cancer, endometrial cancer, cervical cancer, stomach cancer, colon cancer, bladder cancer, pancreatic cancer, lung cancer, breast cancer, prostate cancer and liver cancer (eg liver Cell carcinoma), more specifically liver cancer, gastric cancer, non-small cell lung cancer and bladder cancer.
  • liver cancer eg liver Cell carcinoma
  • inflammatory disease refers to any inflammatory disease in which FGFR is involved in the onset of inflammation, such as osteoarthritis.
  • terapéuticaally effective amount refers to an amount that includes a compound of the invention that is effective to inhibit the function of FGFR and/or to treat or prevent the disease.
  • the invention also provides methods of making the compounds.
  • the preparation of the compounds of the general formula (I) of the present invention can be carried out by the following exemplary methods and examples, but the methods and examples should not be construed as limiting the scope of the invention in any way.
  • the compounds of the invention may also be synthesized by synthetic techniques known to those skilled in the art, or a combination of methods known in the art and methods of the invention may be employed.
  • the product obtained in each step of the reaction is obtained by separation techniques known in the art including, but not limited to, extraction, filtration, distillation, crystallization, chromatographic separation and the like.
  • the starting materials and chemical reagents required for the synthesis can be conventionally synthesized or purchased according to the literature (available from SciFinder).
  • the pyrazole compound of the formula (I) of the present invention can be synthesized according to the route described in Process A: 1) the starting material A1 is obtained by the Sandmeyer reaction to obtain A2, and can also be brominated to obtain A3, wherein R 1 may be -CN or an ester (-COOR, wherein R is an alkyl group); 2) A2 or A3 and a precursor XL to NP (wherein X is a leaving group and L to NP is a functional group containing a protected amino group) , P is a protecting group for an amino group) a substitution reaction occurs under base catalysis to form A4, and a precursor having a hydroxyl group (HO-L to NP) can also be subjected to a Mitsunobu reaction to obtain A4; 3) when R of A4 1 is -CN, hydrolyzed to amide A5 under NaOH/H 2 O 2 conditions; when R 1 of A4 is an ester (-COOR, wherein R is an alkyl group),
  • A5 A5 and alkyne are coupled by Sonogashira to obtain A6; 5) A6 in A6 deprotected to give A7; 6) Amino group in A7 is contained in the kinase ligand binding domain cysteine residue
  • the chemical reagent e.g., BrCN, acryloyl chloride, etc.
  • the reactive functional group is derivatized to give the target compound A8.
  • the pyrazole NH protecting group Q is introduced, and in the fifth step, the common intermediate B7 is obtained, and the pyrazole NH of B7 is different from that of the protected amino group.
  • the bulk reaction is substituted, deprotected and derivatized to give the target product A8.
  • the pyrazole compound of the formula (I) of the present invention can also be synthesized according to the route described in Process C: 1) A4 is first coupled with an alkyne via Sonogashira to obtain C1; 2) -NH 2 in C1 is substituted under base catalysis. Reaction or reductive amination to form C2; 3) C2 CN is hydrolyzed to amide C3 under NaOH/H 2 O 2 conditions, in some cases first protected with Boc -NH-, then hydrolyzed; finally deprotected and Derivatization to obtain the target product C5; C5 can also be obtained by direct substitution of A8.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or mass spectrometry (MS). NMR was measured using a Bruker AVANCE-400 or Varian Oxford-300 nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD). The internal standard is tetramethylsilane (TMS) and the chemical shift is given in units of 10 -6 (ppm).
  • NMR nuclear magnetic resonance
  • MS mass spectrometry
  • the MS was measured using an Agilent SQD (ESI) mass spectrometer (manufacturer: Agilent, model: 6120).
  • ESI Agilent SQD
  • the HPLC was measured using an Agilent 1200 DAD high pressure liquid chromatograph (Sunfirc C18, 150 x 4.6 mm, 5 ⁇ m column) and a Waters 2695-2996 high pressure liquid chromatograph (Gimini C18 150 x 4.6 mm, 5 ⁇ m column).
  • the thin-layer chromatography silica gel plate uses Qingdao Ocean GF254 silica gel plate, and the silica gel plate used for thin-layer chromatography (TLC) has a specification of 0.15 mm to 0.2 mm.
  • the specification for separation and purification of thin layer chromatography is 0.4 mm to 0.5 mm. silicone board.
  • the known starting materials of the present invention may be synthesized by or according to methods known in the art, or may be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc., Beijing. Coupling chemicals and other companies.
  • An argon atmosphere or a nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 L.
  • the hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon of about 1 L volume.
  • the hydrogenation reaction is usually evacuated, charged with hydrogen, and operated three times.
  • the microwave reaction used a CEM Discover-SP type microwave reactor.
  • the temperature of the reaction is room temperature, and the temperature range is from 20 ° C to 30 ° C.
  • reaction progress in the examples was monitored by thin layer chromatography (TLC), and the system used for the reaction was A: dichloromethane and methanol system; B: petroleum ether and ethyl acetate system, the volume ratio of the solvent was based on The polarity of the compound is adjusted to adjust.
  • TLC thin layer chromatography
  • the system for purifying the compound using the column chromatography eluent and the system for developing the thin layer chromatography include A: dichloromethane and methanol systems; B: petroleum ether and ethyl acetate system, the volume ratio of the solvent according to the compound The polarity is adjusted to adjust, and a small amount of triethylamine and an acidic or alkaline reagent may be added for adjustment.
  • the compound 5-amino-1H-pyrazole-4-carbonitrile 7a (20 g, 185 mmol) was dissolved in N,N-dimethylformamide (200 mL), cooled to 0 ° C, and N-bromobutane was added in portions.
  • the imide 34 g, 190 mmol was stirred at room temperature for 2 h.
  • the reaction solution was poured into a sodium sulfite solution, and extracted with ethyl acetate (200 mL ⁇ 3).
  • reaction solution was poured into water and extracted with dichloromethane (20 mL ⁇ 3).
  • the organic phase was combined and dried over anhydrous sodium sulfate, filtered, evaporated, evaporated, evaporated, evaporated, evaporated -dimethoxyphenyl)ethynyl)-1-(1-(4-(dimethylamino)but-2-enoyl)pyrrolidin-3-yl)-1H-pyrazole-4-carboxamide 8 (2.4 mg, white solid, formate), yield: 4%.
  • Example 13-16 was synthesized with reference to the operational steps of Example 7:
  • Example 17 was synthesized by referring to the procedure of the first to sixth steps of Example 7, but in the third step, 1-ethynyl-2-fluoro-3,5-dimethoxybenzene was substituted for 1-ethynyl-3. , 5-dimethoxybenzene.
  • Example 18 was synthesized by referring to the procedure of the first to sixth steps of Example 7, but in the third step, 1-chloro-2-ethynyl-1-fluorobenzene was substituted for 1-ethynyl-3,5-dimethyl Oxybenzene.
  • Methyl 4-chloro-3-((trimethylsilyl)ethynyl)benzoate 19c (1 g, 3.76 mmol) was dissolved in MeOH (20 mL) then EtOAc (EtOAc) After stirring at room temperature for 1 hour, it was desolvated under reduced pressure. The residue was washed with water and filtered tolululululululululululululu
  • 3-Bromo-5-methoxybenzoic acid 20a (500 mg, 2.17 mmol) was dissolved in N,N-dimethylformamide (15 mL), then methylamine hydrochloride (291 mg, 4. Diisopropylethylamine (1.12 g, 8.68 mmol) and 2-(7-oxobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (1.24 g, 3.26 mmol). The reaction mixture was stirred at room temperature for 2 hr then EtOAc (EtOAc)EtOAc. Methylbenzamide 20b (500 mg, white solid), yield: 95%.
  • Example 22 was synthesized by following the procedure of Example 21, but in the second step, 1-ethynyl-2-fluoro-3,5-dimethoxybenzene was substituted for 1-ethynyl-3,5-dimethoxy Base benzene.
  • Example 23 was synthesized by following the procedure of Example 21, but in the second step, 1-chloro-2-ethynyl-1-fluorobenzene was substituted for 1-ethynyl-3,5-dimethoxybenzene.
  • Example 25 was synthesized with reference to the operation steps of the second to fourth steps in Example 24.
  • Example 27 was synthesized by referring to the operation steps of the second to fourth steps in Example 24.
  • Example 28 was synthesized by following the procedure of Example 25, but in the first step, 2-bromopropane was replaced with 1-bromo-2-methoxyethane.
  • Example 30 was synthesized by reference to the procedure of Example 25 except that 2-bromopropane was replaced with 3-morpholinopropyl 4-methylbenzenesulfonate in the first step.
  • Example 31 was synthesized by reference to the procedure of Example 25, but in the first step, 2-bromopropane was replaced with 4-(2-chloroethyl)morpholine.
  • Example 32 was synthesized with reference to the operation steps of the second to fourth steps in Example 24.
  • Example 33 was synthesized by following the procedure of Example 24, but in the first step, ethyl iodoethane was replaced with 4-iodotetrahydro-2H-pyran.
  • the reaction buffer contained the following components: 5-fold diluted enzymatic buffer/kinase 5X (Cisbio, Cat. No. 62EZBFDD) (main component 50 mM HEPES, pH 7.0), 5 mM MgCl 2 and 1 mM DTT; human recombinant FGFR1 catalytic domain protein (Amino acid 308-731) was purified by the company itself and diluted to 0.6 ng/uL of kinase solution with reaction buffer; substrate reaction solution including biotin-labeled tyrosine kinase substrate diluted to 400 nM with reaction buffer (Cisbio , the product number is 62TK0PEC) and 40uM ATP; the test solution consists of a test buffer (Cisbio, Cat.
  • the phosphorylated product was simultaneously recognized by the Eu 3+ labeled cage antibody (donor) and the streptavidin-labeled XL665 antibody (receptor), after laser excitation, close to the donor and acceptor Energy resonance transfer occurred, and the energy transferred from the donor (620 nm) to the acceptor (665 nm) was detected by a microplate reader EnVision (Perkin Elmer).
  • the ratio of 665/620 is positively correlated with the degree of phosphorylation of the substrate, thus detecting the activity of FGFR1 kinase.
  • Percent inhibition 100-100* (ratio compound - ratio 100% inhibition ) / (ratio 0% inhibition - ratio 100% inhibition )
  • the IC 50 value of the compound was calculated from the 10 concentration points using the XLfit software in Excel by the following formula:
  • Y is the percent inhibition
  • Bottom is the bottom platform value of the S-curve
  • Top is the top platform value of the S-curve
  • X is the logarithm of the concentration of the test compound
  • slope factor is the slope coefficient of the curve.
  • the reaction buffer contained the following components: 5-fold diluted enzymatic buffer/kinase 5X (Cisbio, Cat. No. 62EZBFDD) (main component 50 mM HEPES, pH 7.0), 5 mM MgCl 2 and 1 mM DTT; human recombinant FGFR2 catalytic domain protein (Amino acid 400-821) purchased from Yishen Shenzhou Biotechnology Co., Ltd., diluted to 0.45 ng/uL of kinase solution with reaction buffer; substrate reaction solution including biotin-labeled tyrosine diluted to 800 nM with reaction buffer Kinase substrate (Cisbio, Cat. No.
  • the assay solution consisted of a test buffer (Cisbio, Cat. No. 62SDBRDF) diluted to 0.125 ng/uL Eu 3+ labeled cage antibody (Cisbio, Cat. No. 61T66KLB), 50 nM streptavidin-labeled XL665 (Cisbio, Cat. No. 610 SAXLB).
  • Percent inhibition 100-100* (ratio compound - ratio 100% inhibition ) / (ratio 0% inhibition - ratio 100% inhibition )
  • the IC 50 value of the compound was calculated from the 10 concentration points using the XLfit software in Excel by the following formula:
  • Y is the percent inhibition
  • Bottom is the bottom platform value of the S-curve
  • Top is the top platform value of the S-curve
  • X is the logarithm of the concentration of the test compound
  • slope factor is the slope coefficient of the curve.
  • the reaction buffer contained the following components: 5-fold diluted enzymatic buffer/kinase 5X (Cisbio, Cat. No. 62EZBFDD) (main component 50 mM HEPES, pH 7.0), 5 mM MgCl 2 and 1 mM DTT; human recombinant FGFR3 catalytic domain protein (Amino acid 399-806) was purchased from Yishen Shenzhou Biotechnology Co., Ltd. and diluted to 0.3 ng/uL of kinase solution with reaction buffer; substrate reaction solution including diluted with reaction buffer to 1000 nM biotin-labeled tyrosine The kinase substrate (Cisbio, Cat. No.
  • the assay solution consisted of a test buffer (Cisbio, Cat. No. 62SDBRDF) diluted to 0.125 ng/uL Eu 3+ labeled cage antibody (Cisbio, Cat. No. 61T66KLB), 62.5 nM streptavidin-labeled XL665 (Cisbio, Cat. No. 610 SAXLB).
  • Percent inhibition 100-100* (ratio compound - ratio 100% inhibition ) / (ratio 0% inhibition - ratio 100% inhibition )
  • the IC 50 value of the compound was calculated from the 10 concentration points using the XLfit software in Excel by the following formula:
  • Y is the percent inhibition
  • Bottom is the bottom platform value of the S-curve
  • Top is the top platform value of the S-curve
  • X is the logarithm of the concentration of the test compound
  • slope factor is the slope coefficient of the curve.
  • the reaction buffer contained the following components: 5-fold diluted enzymatic buffer/kinase 5X (Cisbio, Cat. No. 62EZBFDD) (main component 50 mM HEPES, pH 7.0), 5 mM MgCl 2 and 1 mM DTT; human recombinant FGFR4 catalytic domain protein (Amino acids 460-802) was purchased from Tsinghua University Protein Research Technology Center and diluted to 0.5 ng/uL of kinase solution with reaction buffer; substrate reaction solution including biotinylated tyrosine kinase diluted to 500 nM with reaction buffer Substrate (Cisbio, Cat. No.
  • assay solution consisting of diluted to 0.125 ng/uL Eu 3+ labeled cage antibody (Cisbio, Cat. No. 61T66KLB), 31.25, using assay buffer (Cisbio, Cat. No. 62SDBRDF) nM streptavidin-labeled XL665 (Cisbio, Cat. No. 610 SAXLB).
  • Percent inhibition 100-100* (ratio compound - ratio 100% inhibition ) / (ratio 0% inhibition - ratio 100% inhibition )
  • the IC 50 value of the compound was calculated from the 10 concentration points using the XLfit software in Excel by the following formula:
  • Y is the percent inhibition
  • Bottom is the bottom platform value of the S-curve
  • Top is the top platform value of the S-curve
  • X is the logarithm of the concentration of the test compound
  • slope factor is the slope coefficient of the curve.
  • Example compounds of the present invention have significant activity of FGFR inhibitory effect, preferably an IC 50 of 100 to 1000 nM, more preferably less than 10OnM IC 50, IC 50 and most preferably less than 10nM.
  • CellTilter-Glo reagent (Promega, product number G7572) consists of CTG lyophilized powder and CTG buffer. When used, the lyophilized powder can be dissolved in buffer.
  • the compound was diluted with DMSO (Sigma, Cat. No. D5879) to 5 mM, then serially diluted 4 times with DMSO to a minimum concentration of 0.31 uM, and each concentration point was again in DMEM medium without FBS (ThermoFisher, item number is 11995073) diluted 50 times. If the compound IC50 value is very low, the initial concentration of the compound can be lowered.
  • Hep3B cells (from the Cell Resource Center of Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences) were fully cultured in DMEM containing 10% FBS (GBICO, Cat. No. 10099-141) and 100 U/mL Streptomycin Mixture (ThermoFisher, Cat. No. 15140122). The medium is cultured. When the cells cover 80-90% in the culture vessel, they are digested with 0.25% trypsin (containing EDTA) (ThermoFisher, Cat. No. 25200056) and then planted in a white 384-well plate (ThermoFisher, item No. 164610), 1000 cells per well (27 uL DMEM complete medium), then 384-well plates were incubated overnight (18-20 hours) in a 37 ° C, 5% CO 2 incubator.
  • trypsin containing EDTA
  • the cold light signal of BLU9931 (Cancer Discovery 2015, 5, 424) group with 10uM Blueprint was used as signal 100% inhibition
  • the cold light signal of 0.2% DMSO group was used as signal 0% inhibition .
  • the percentage inhibition of Hep3B cell proliferation by the compound can be calculated by the following formula:
  • Percent inhibition 100-100* (signal compound- signal 100% inhibition ) / (signal 0% inhibition - signal 100% inhibition )
  • Y is the percent inhibition
  • Bottom is the bottom platform value of the S-curve
  • Top is the top platform value of the S-curve
  • X is the logarithm of the concentration of the test compound
  • slope factor is the slope coefficient of the curve.
  • Example compounds of the invention are cells Hep3B, RT4 and SNU-16 has significant proliferation inhibitory effect, preferably an IC 50 of 100 to 1000 nM, more preferably IC 50 less than 100nM.

Abstract

本发明涉及作为FGFR抑制剂的炔代杂环化合物、其制备方法及其在医药学上的应用。具体而言,本发明涉及一种通式(I)所示的化合物及其可药用的盐、含有所述化合物或其可药用的盐的药物组合物、应用所述化合物或其可药用的盐治疗和/或预防FGFR相关性病症、特别是肿瘤的方法以及所述化合物或其可药用的盐的制备方法。本发明还涉及所述化合物或其可药用的盐或含有所述化合物或其可药用的盐的药物组合物在制备用于治疗和/或预防FGFR相关性病症、特别是肿瘤的药物中的用途。其中通式(I)的各取代基与说明书中的定义相同。

Description

炔代杂环化合物、其制备方法及其在医药学上的应用 技术领域
本发明涉及一种作为FGFR抑制剂的新的炔代杂环化合物或其可药用的盐;含有所述炔代杂环化合物或其可药用的盐的药物组合物;所述炔代杂环化合物或其可药用的盐的制备方法;所述炔代杂环化合物或其可药用的盐、或含有所述炔代杂环化合物或其可药用的盐的药物组合物在制备用于治疗和/或预防FGFR相关性病症、特别是肿瘤的药物中的用途;以及应用所述化合物或组合物治疗和/或预防FGFR相关性病症、特别是肿瘤的方法。
背景技术
成纤维细胞生长因子受体(Fibroblast Growth Factor Receptor,FGFR)是一类受体酪氨酸激酶(RTK),结构上由膜外配体结合域、单一的跨膜域和膜内酪氨酸激酶域所组成,主要包括FGFR1、FGFR2、FGFR3和FGFR4四种亚型。它与其配体,成纤维细胞生长因子(Fibroblast Growth Factor,FGF)在细胞信号传递中起重要的调节作用。FGF作为细胞外刺激信号,与FGFR膜外区结合,引起其膜内酪氨酸激酶磷酸化,从而激活下游的一系列信号通路,对细胞的增殖、分化和转移等进行调控。
多种肿瘤与FGF/FGFR表达及激活密切相关,比如非小细胞肺癌、乳腺癌、胃癌、肝癌、膀胱癌、子宫内膜癌、***癌、***、结肠癌、食管癌、骨髓瘤和黑色素瘤等(Clin.Cancer Res.2012,18,1855)。研究显示,FGFRl扩增占非小细胞肺癌的20%、FGFR2扩增占胃癌的~5%、FGFR3突变占非侵袭性膀胱癌的~70%和FGFR4在肝癌中的扩增等(PloS One 2012,7,e36713)。因此,靶向FGFR的抑制剂的研发已成为抗肿瘤药物研究的前沿热点(Drug Disc.Today 2014,19,51)。
目前市场上已有一些非FGFR特异性药物,比如Pfizer的sunitinib、Eisai的lenvatini和Boehringer Ingelheim的nintedanib,但还没有FGFR特异性抑制剂。进入临床的FGFR特异性抑制剂有HMPL-453、BGJ-398、LY-2874455、AZ-4547、JNJ-42756493、TAS-120、ARQ-087和BLU-554等。
尽管FGFR抑制剂的开发吸引了众多生物制药公司的关注,由于其在治疗多种恶性肿瘤所展示的前景,仍需要开发新的化合物。经过不断努力,本发明设计具有通式(I)所示的结构的化合物,并发现具有此类结构的化合物表现出优异的效果和作用。
发明内容
本发明提供作为FGFR抑制剂的一种通式(I)所示的化合物、其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式:
Figure PCTCN2017072570-appb-000001
其中:
A为N或CR2
环B为苯环或5-6元杂芳环,其中所述苯环和杂芳环任选被一个或多个G1所取代;
R1独立地选自H、卤素、氰基、C1-6烷基或-NHR3
R2独立地选自H、卤素、氰基或C1-6烷基,其中所述烷基任选被卤素、氰基、羟基或-OC1-6烷基所取代;
R3独立地选自H、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被卤素、氰基、-OR4、-NR5R6、C1-6烷基、C3-6环烷基或3-6元杂环基所取代;
X不存在或为C1-6亚烷基;
Y不存在或选自C3-8亚环烷基、3-8元亚杂环基、亚芳基或亚杂芳基,其中所述亚环烷基、亚杂环基、亚芳基和亚杂芳基任选被一个或多个G2所取代;
Z独立地选自氰基、-NR7CN、
Figure PCTCN2017072570-appb-000002
键a为双键或三键;
当键a为双键时,Ra、Rb和Rc各自独立地选自H、氰基、卤素、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被一个或多个G3所取代;
Ra和Rb或Rb和Rc任选与它们连接的碳原子共同形成一任选含有杂原子的3-6元环;
当键a为三键时,Ra和Rc不存在,Rb独立地选自H、氰基、卤素、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被一个或多个G4所取代;
R4独立地选自H、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选可被一个或多个G5所取代;
G1、G2、G3、G4和G5各自独立地选自卤素、氰基、C1-6烷基、C2-6烯基、C2-6炔基、C3-8环烷基、3-8元杂环基、C6-10芳基、5-10元杂芳基、-OR8、-OC(O)NR8R9、-C(O)OR8、-C(O)NR8R9、-C(O)R8、-NR8R9、-NR8C(O)R9、-NR8C(O)NR9R10、-S(O)mR8或-NR8S(O)mR9,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选被一个或多个选自卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR11、-OC(O)NR11R12、-C(O)OR11、-C(O)NR11R12、-C(O)R11、-NR11R12、-NR11C(O)R12、-NR11C(O)NR12R13、-S(O)mR11或-NR11S(O)mR12的取代基所取代;
R4、R5、R6、R8、R9、R10、R11、R12和R13各自独立地选自H、C1-6烷基、C3-8环烷基、3-8元单环杂环基、单环杂芳基或苯基;且
m为1或2。
本发明的一个实施方案涉及上述通式(I)所示的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式,其中A为N或CH,优选为N。
本发明的另一个实施方案涉及上述通式(I)所示的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式,其中环B为苯环。
在一方面,本发明提供以下通式(II)的化合物、或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式:
Figure PCTCN2017072570-appb-000003
其中:
Ga、Gb、Gc和Gd各自独立地选自H、卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR8、-NR8R9或-C(O)NR8R9,其中所述烷基、 环烷基和杂环基任选被一个或多个选自卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR11或-NR11R12的取代基所取代;其中A、R1、R8、R9、R11、R12、X、Y、Z的定义如前所述。
本发明的另一个实施方案涉及上述通式(I)所示的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式,其为通式(III)所述的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式:
Figure PCTCN2017072570-appb-000004
其中:
Ga和Gb各自独立地选自H、卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR8、-NR8R9或-C(O)NR8R9,其中所述烷基、环烷基和杂环基任选被一个或多个选自卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR11或-NR11R12的取代基所取代;其中A、R1、R8、R9、R11、R12、X、Y、Z的定义如前所述。
本发明的另一个实施方案涉及上述通式(I)所示的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式,其中R1独立地选自H、-NH2或-NHC1-6烷基。
在本发明实施方案中,R1可以为H或-NH2
在本发明实施方案中,Ga、Gb、Gc和Gd可以各自独立地为-OC1-2烷基或卤素。
在本发明实施方案中,R1独立地选自H、-NH2或-NHR3;R3独立地选自C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被卤素、氰基、-OR4、-NR5R6、C1-6烷基、C3-6环烷基或3-6元杂环基所取代。
本发明的另一个实施方案涉及上述通式(I)、(II)和(III)所示的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式,其中:
X不存在或为C1-6亚烷基;
Y不存在或选自C3-8亚环烷基或3-8元亚杂环基;
Z独立地选自氰基、-NR7CN、
Figure PCTCN2017072570-appb-000005
键a为双键或三键;
当键a为双键时,Ra、Rb和Rc各自独立地选自H、氰基、卤素、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被一个或多个独立地选自卤素、氰基、C1-6烷基、C3-6环烷基、3-6元杂环基、-OR8或-NR8R9的取代基所取代;
当键a为三键时,Ra和Rc不存在,Rb独立地选自H、氰基、卤素、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被一个或多个独立地选自卤素、氰基、C1-6烷基、C3-6环烷基、3-6元杂环基、-OR8或-NR8R9的取代基所取代;
R4、R8、R9各自独立地选自H或C1-6烷基。
本发明的一个实施方案涉及上述通式(I)所示的化合物,其中所述化合物选自:
Figure PCTCN2017072570-appb-000006
Figure PCTCN2017072570-appb-000007
Figure PCTCN2017072570-appb-000008
Figure PCTCN2017072570-appb-000009
Figure PCTCN2017072570-appb-000010
Figure PCTCN2017072570-appb-000011
Figure PCTCN2017072570-appb-000012
或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式。
本发明化合物对FGFR的活性具有显著抑制效应。本发明化合物能够有效抑制FGFR1、FGFR2、FGFR3或FGFR4的活性,优选其抑制FGFR1、FGFR2、FGFR3或FGFR4的IC50为100至1000nM,更优选IC50小于100nM,最优选其IC50小于10nM。特别的是,本发明化合物对肿瘤细胞(例如Hep3B、RT4和SNU-16肿瘤细胞)的细胞增殖具有显著抑制效应,优选其IC50为100至1000nM,更优选其IC50小于100nM,最优选其IC50小于10nM。
因此本发明化合物可用于治疗或者预防FGFR相关性疾病,包括但不限于肿瘤和炎症性疾病,例如骨关节炎。本发明化合物可用于治疗或者预防FGFR相关性肿瘤,例如非小细胞肺癌、食管癌、黑色素瘤、横纹肌肉瘤、肾细胞癌、多发性骨髓瘤、乳腺癌、卵巢癌、子宫内膜癌、***、胃癌、结肠癌、膀胱癌、胰腺癌、肺癌、乳腺癌、***癌和肝癌(例如肝细胞癌),更具体为肝癌、胃癌、非小细胞肺癌和膀胱癌。因此,再一方面,本发明提供一种治疗或者预防FGFR介导的疾病(例如所述肿瘤)的方法,其包括给予有需要的患者治疗有效量的本发明所述化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物、或包含所述化合物的药物组合物。
本发明的另一方面涉及作为药物或者医药用途的通式(I)所示的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物,其用于治疗或者预防FGFR介导的疾病,例如肿瘤或炎症性疾病,包括但不限于非小细胞肺癌、食管癌、黑色素瘤、横纹肌肉瘤、肾细胞癌、多发性骨髓瘤、乳腺癌、卵巢癌、子宫内膜癌、***、胃癌、结肠癌、膀胱癌、胰腺癌、肺癌、乳腺癌、***癌和肝癌。
本发明进一步涉及一种药物组合物,所述药物组合物包含本发明所述化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物及药学上可接受的载体、稀释剂、赋形剂。
本发明的另一方面涉及通式(I)所示的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物、或所述药物组合物在制备药物中的用途,其中所述药物用于治疗或者预防FGFR介导的疾病,例如肿瘤和炎症性疾病。
根据本发明,所述药物可以是任何药物剂型,包括但不限于片剂、胶囊剂、溶液剂、冻干制剂、注射剂。
本发明的药物制剂可以以每剂量单位包含预定量的活性成分的剂量单位形式给药。这种单位可根据治疗的病症、给药方法和患者的年龄、体重和状况包含例如0.5毫克至1克,优选1毫克至700毫克,特别优选5毫克至300毫克的本发明的化合物,或药物制剂可以以每剂量单位包含预定量的活性成分的剂量单位形式给药。优选剂量单位制剂是包含如上指示的日剂量或分剂量或其相应分数的活性成分的那些。此外,可以使用制药领域中公知的方法制备这种类型的药物制剂。
本发明药物制剂可适于通过任何所需的合适方法给药,例如通过经口(包括口腔或舌下)、直肠、经鼻、局部(包括口腔、舌下或经皮)、***或肠道外(包括皮下、肌内、静脉内或皮内)方法给药。可以使用制药领域中已知的所有方法通过例如将活性成分与一种或多种赋形剂或一种或多种辅助剂合并来制备这样的制剂。
具体实施方式
除非有相反陈述,否则下列用在说明书和权利要求书中的术语具有下述含义。
在本文中使用的表示方式“Cx-y”表示碳原子数的范围,其中x和y均为整数,例如C3-8环烷基表示具有3-8个碳原子的环烷基,即具有3、4、5、6、7或8个碳原子的环烷基。还应理解,“C3-8”还包含其中的任意亚范围,例如C3-7、C3-6、C4-7、C4-6、C5-6等。
“烷基”指含有1至20个碳原子,例如1至18个碳原子、1至12个碳原子、1至8个碳原子、1至6个碳原子或1至4个碳原子的饱和的直链或支链的烃基基团。烷基的非限制性实例包括甲基、乙基、正 丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基和2-乙基丁基。所述烷基可以是取代的或未取代的。
“烯基”指含有至少一个碳碳双键和通常2至20个碳原子,例如2至8个碳原子、2至6个碳原子或2至4个碳原子的直链或支链的烃基基团。烯基的非限制性实例包括乙烯基、1-丙烯基、2-丙烯基、1-丁烯基、2-丁烯基、3-丁烯基、2-甲基-2-丙烯基、1,4-戊二烯基和1,4-丁二烯基。所述烯基可以是取代的或未取代的。
“炔基”指含有至少一个碳碳三键和通常2至20个碳原子,例如2至8个碳原子、2至6个碳原子或2至4个碳原子的直链或支链的烃基基团。炔基的非限制性实例包括乙炔基、1-丙炔基、2-丙炔基、1-丁炔基、2-丁炔基和3-丁炔基。所述炔基可以是取代的或未取代的。
“环烷基”指含有3至14个碳环原子的饱和环形烃基取代基。环烷基可以是单碳环,通常含有3至7个碳环原子。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环己基和环庚基。环烷基可选择地可以是稠合到一起的双或三环,如十氢萘基。所述环烷基可以是取代的或未取代的。
“杂环或杂环基”指饱和或部分不饱和的单环或多环环状基团,其包括3至20个环原子,例如可以是3至16个、3至14个、3至12个、3至10个、3至8个、3至6个或5至6个环原子,其中一个或多个环原子选自氮、氧或S(O)m(其中m是整数0至2),但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包括3至12个环原子,更优选3至10个环原子,最优选5或6个环原子,其中1~4个是杂原子,更优选1~3个是杂原子,最优选1~2个是杂原子。单环杂环基的非限制性实例包含吡咯烷基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基、氧杂环己烷基和氮杂环丁烷基。多环杂环基包括稠合、桥接或螺多环杂环基。所述杂环或杂环基可以是取代的或未取代的。
“芳基”指含有6至14个碳原子的芳香族单环或稠合多环基团,优选为6至10元,例如苯基和萘基,最优选苯基。所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为 芳基环,非限制性实例包括:
Figure PCTCN2017072570-appb-000013
所述芳基可以是取代的或未取代的。
“杂芳基或杂芳环”指包含5至14个环原子的杂芳族体系,其中1至4个环原子选自包括氧、硫和氮的杂原子。杂芳基优选为5至10元。更优选杂芳基是5元或6元,例如呋喃基、噻吩基、吡啶基、吡咯基、N-烷基吡咯基、嘧啶基、吡嗪基、吡唑基、咪唑基、四唑基、噁唑基、异噁唑基、噻唑基、异噻唑基等。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,非限制性实例包括:
Figure PCTCN2017072570-appb-000014
所述杂芳基可以是取代的或未取代的。
“卤素”指氟、氯、溴或碘。
“氰基”指-CN。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生地场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。所述取代基包括但不限于羟基、氨基、卤素、氰基、C1-6烷基、C1-6烷氧基、C2-6烯基、C2-6炔基、C3-8环烷基等。
“药物组合物”指含有一种或多种本文所述的化合物或其可药用的盐或前药以及其他组分例如可药用的载体和赋形剂的组合物。药物组 合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“异构体”指具有相同分子式但其原子结合的性质或顺序或其原子的空间排列不同的化合物称为“异构体”。其原子空间排列不同的异构体称为“立体异构体”。立体异构体包括光学异构体、几何异构体和构象异构体。
本发明的化合物可以以光学异构体形式存在。根据手性碳原子周围取代基的构型,这些光学异构体是“R”或“S”构型。光学异构体包括对映异构体和非对映异构体。制备和分离光学异构体的方法是本领域中已知的。
本发明的化合物也可以存在几何异构体。本发明考虑由碳-碳双键、碳-氮双键、环烷基或杂环基团周围的取代基的分布所产生的各种几何异构体和其混合物。碳-碳双键或碳-氮键周围的取代基指定为Z或E构型,环烷基或杂环周围的取代基指定为顺式或反式构型。
本发明的化合物还可能显示互变异构现象,例如酮-烯醇互变异构。
应该理解,本发明包括任何互变异构或立体异构形式和其混合物,并且不仅仅限于化合物的命名或化学结构式中所使用的任何一个互变异构或立体异构形式。
“同位素”是在本发明化合物中出现的原子的所有同位素。同位素包括具有相同原子序数但不同质量数的那些原子。适合并入本发明化合物中的同位素的实例是氢、碳、氮、氧、磷、氟和氯,分别例如但不限于2H、3H、13C、14C、15N、18O、17O、31P、32P、35S、18F和36Cl。本发明的同位素标记化合物通常可通过本领域技术人员已知的传统技术或通过与所附实施例中描述的那些类似的方法使用适当的同位素标记的试剂代替非同位素标记的试剂来制备。这样的化合物具有各种潜在用途,例如作为测定生物活性中的标样和试剂。在稳定同位素的情况下,这样的化合物具有有利地改变生物、药理学或药代动力学性质的潜力。
“前药”是指本发明的化合物可以以前药的形式给予。前药是指在活体内的生理条件下例如通过氧化、还原、水解等(它们各自利用酶或在没有酶参与下进行)转化成本发明的生物活性化合物的衍生物。前药的实例是下述化合物:其中本发明的化合物中的氨基被酰化、烷基 化或磷酸化,例如二十烷酰基氨基、丙氨酰氨基、新戊酰氧基甲基氨基,或其中羟基被酰化、烷基化、磷酸化或转化成硼酸盐,例如乙酰氧基、棕榈酰氧基、新戊酰氧基、琥珀酰氧基、富马酰氧基、丙氨酰氧基,或其中羧基被酯化或酰胺化,或其中巯基与选择性地向靶和/或向细胞的胞质溶胶递送药物的载体分子,例如肽形成二硫桥键。这些化合物可以由本发明的化合物根据公知方法制备。
“可药用的盐”或者“药学上可接受的盐”是指由可药用的碱或酸,包括无机碱或酸和有机碱或酸制成的盐。在本发明的化合物含有一个或多个酸性或碱性基团的情况下,本发明还包含它们相应的可药用盐。因此,含有酸性基团的本发明的化合物可以以盐形式存在并可根据本发明使用,例如作为碱金属盐、碱土金属盐或作为铵盐。这样的盐的更确切实例包括钠盐、钾盐、钙盐、镁盐或与氨或有机胺,例如乙胺、乙醇胺、三乙醇胺或氨基酸的盐。含有碱性基团的本发明的化合物可以以盐形式存在并可根据本发明以它们与无机或有机酸的加成盐的形式使用。合适的酸的实例包括盐酸、氢溴酸、磷酸、硫酸、硝酸、甲磺酸、对甲苯磺酸、萘二磺酸、草酸、乙酸、酒石酸、乳酸、水杨酸、苯甲酸、甲酸、丙酸、特戊酸、丙二酸、琥珀酸、庚二酸、富马酸、马来酸、苹果酸、氨基磺酸、苯基丙酸、葡糖酸、抗坏血酸、异烟酸、柠檬酸、己二酸和本领域技术人员已知的其它酸。如果本发明的化合物在分子中同时含有酸性和碱性基团,本发明除所提到的盐形式外还包括内盐或内铵盐。各盐可通过本领域技术人员已知的常规方法获得,例如通过在溶剂或分散剂中使这些与有机或无机酸或碱接触或通过与其它盐阴离子交换或阳离子交换。
因此,在本申请中当提及“化合物”、“本发明化合物”或“本发明所述化合物”时,包括所有所述化合物形式,例如其前药、稳定同位素衍生物、可药用的盐、异构体、内消旋体、外消旋体、对映异构体、非对映异构体及其混合物。
在本文中,术语“肿瘤”包括良性肿瘤和恶性肿瘤(例如癌症)。
在本文中,术语“癌症”包括FGFR参与其发生的各种恶性肿瘤,包括但不限于非小细胞肺癌、食管癌、黑色素瘤、横纹肌肉瘤、肾细胞癌、多发性骨髓瘤、乳腺癌、卵巢癌、子宫内膜癌、***、胃癌、结肠癌、膀胱癌、胰腺癌、肺癌、乳腺癌、***癌和肝癌(例如肝 细胞癌),更具体为肝癌、胃癌、非小细胞肺癌和膀胱癌。
在本文中,术语“炎症性疾病”是指FGFR参与其炎症发生的任何炎性疾病,例如骨关节炎。
在本文中,术语“治疗有效量”是指包括可有效抑制FGFR的功能和/或治疗或防止所述疾病的本发明化合物的量。
合成方法
本发明还提供制备所述化合物的方法。本发明通式(I)所述化合物的制备,可通过以下示例性方法和实施例完成,但这些方法和实施例不应以任何方式被认为是对本发明范围的限制。也可通过本领域技术人员所知的合成技术合成本发明所述的化合物,或者综合使用本领域已知方法和本发明所述方法。每步反应所得的产物用本领域已知的分离技术得到,包括但不限于萃取、过滤、蒸馏、结晶、色谱分离等。合成所需的起始原料和化学试剂可以根据文献(可从SciFinder上查询)常规合成或购买。
本发明通式(I)所述吡唑类化合物可按照方法A所述路线合成:1)起始物A1通过桑德迈尔(Sandmeyer)反应得到A2,也可被溴化得到A3,其中的R1可以是-CN或酯(-COOR,其中R为烷基);2)A2或A3与前体X-L~N-P(其中X为离去基团、L~N-P为含有带保护氨基的功能团、P为氨基的保护基)在碱催化下发生取代反应生成A4,也可与一带有羟基的前体(HO-L~N-P)通过光延反应(Mitsunobu反应)得到A4;3)当A4的R1是-CN,在NaOH/H2O2条件下水解成酰胺A5;当A4的R1是酯(-COOR,其中R为烷基),先在碱性条件(比如LiOH)下水解成羧酸,然后酰胺化得到A5;4)A5与炔通过Sonogashira偶联得到A6;5)A6中氨基去保护得到A7;6)A7中的氨基被含有和激酶配体结合域内半胱氨酸残基起反应的功能团的化学试剂(例如BrCN、烯丙酰氯等)衍生化得到目标化合物A8。
方法A:
Figure PCTCN2017072570-appb-000015
另外也可以按照方法B所述路线合成,在第二步引入吡唑NH保护基Q,在第五步脱保护得到共同的中间体B7,B7的吡唑NH再与含有带保护氨基的不同前体反应取代反应,经过脱保护和衍生化,从而得到目标产物A8。
方法B:
Figure PCTCN2017072570-appb-000016
本发明通式(I)所述吡唑类化合物还可以按照方法C所述路线合成:1)A4先与炔通过Sonogashira偶联得到C1;2)C1中的-NH2在碱催化下发生取代反应或通过还原氨化生成C2;3)C2的CN在NaOH/H2O2条件下水解成酰胺C3,在有些情况下需先用Boc保护-NH-,然后再水解;最后经过脱保护和衍生化,从而得到目标产物C5;C5也可通过对A8进行直接取代而得到。
方法C:
Figure PCTCN2017072570-appb-000017
实施例
化合物的结构是通过核磁共振(NMR)或质谱(MS)来确定的。NMR的测定是用Bruker AVANCE-400或Varian Oxford-300核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d6)、氘代氯仿(CDCl3)、氘代甲醇(CD3OD),内标为四甲基甲硅烷(TMS),化学位移是以10-6(ppm)作为单位给出。
MS的测定用Agilent SQD(ESI)质谱仪(生产商:Agilent,型号:6120)。
HPLC的测定使用安捷伦1200DAD高压液相色谱仪(Sunfirc C18,150×4.6mm,5μm色谱柱)和Waters 2695-2996高压液相色谱仪(Gimini C18 150×4.6mm,5μm色谱柱)。
薄层层析硅胶板使用青岛海洋GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm硅胶板。
柱层析一般使用青岛海洋200~300目硅胶为载体。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自ABCR GmbH&Co.KG、Acros Organics、Aldrich Chemical Company、韶远化学科技(Accela ChemBio Inc.)、北京耦合化学品等公司。
实施例中如无特殊说明,反应均在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
加压氢化反应使用北京佳维科创科技有限公司GCD-500G高纯氢 气发生器和BLT-2000中压氢化仪。
氢化反应通常抽真空,充入氢气,反复操作3次。
微波反应使用CEM Discover-SP型微波反应器。
实施例中如无特殊说明,反应的温度为室温,温度范围是20℃-30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂的体系有A:二氯甲烷和甲醇体系;B:石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节。
纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂的体系包括A:二氯甲烷和甲醇体系;B:石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和酸性或碱性试剂等进行调节。
实施例1
(S)-1-(1-丙烯酰基吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000018
Figure PCTCN2017072570-appb-000019
第一步
(R)-3-(甲苯磺酰氧代)吡咯烷-1-甲酸叔丁基酯
将化合物(R)-3-羟基吡咯烷-1-甲酸叔丁基酯1a(3.5g,18.7mmol)、三乙胺(5.25mL,37.9mmol)、4-二甲氨基吡啶(0.35g,2.87mmol)溶于二氯甲烷(50mL),加入对甲苯磺酰氯(5.4g,28.1mmol),将反应混合物在室温下搅拌12小时。加水(50mL)稀释,用乙酸乙酯(100mL×3)萃取。有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,在减压下除去溶剂,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=2/1),得到目标产物(R)-3-(甲苯磺酰氧代)吡咯烷-1-甲酸叔丁基酯1b(6.0g,黄色油状物),产率:94%。
MS m/z(ESI):364[M+23]
第二步
((3,5-二甲氧基苯基)乙炔基)三甲基甲硅烷
将混合物1-溴-3,5-二甲氧基苯1c(6.51g,30mmol)、三甲基甲硅基乙炔(8.8g,90mmol)、二(三苯基膦)氯化钯(1.05g,1.5mmol)、碘化亚铜(0.56g,3.0mmol)、三乙胺(80mL)和N,N-二甲基甲酰胺(150mL)加热至80℃,并在氮气保护下搅拌12小时。将反应混合物冷却至室温,减压浓缩,残余物用硅胶柱层析纯化(石油醚),得到目标产物((3,5-二甲氧基苯基)乙炔基)三甲基甲硅烷1d(6.2g,棕色固体),产率:88%。
MS m/z(ESI):235[M+1]
第三步
1-乙炔基-3,5-二甲氧基苯
将((3,5-二甲氧基苯基)乙炔基)三甲基甲硅烷1d(3.0g,12.8mmol)溶于甲醇(100mL),加入碳酸钾(3.5g,25.6mmol),并在室温下搅拌2小时。过滤,减压浓缩滤液,残余物用硅胶柱层析纯化(石油醚),得到目标产物1-乙炔基-3,5-二甲氧基苯1e(2g,黄色固体),产率:96%。
第四步
3-碘-1H-吡唑-4-甲酸乙酯
将3-氨基-1H-吡唑-4-甲酸乙酯1f(4.7g,30.3mmol)溶于浓盐酸(12M,40mL),并冷却至0℃,加入亚硝酸钠(4.25g,60mmol)溶液(7.5mL),搅拌5分钟,然后缓慢加入碘化钾(12.5g,75mmol)溶液(17.5mL),继续搅拌30分钟。将上述反应混合物倒入饱和硫代硫酸钠溶液(200mL),用乙酸乙酯(400mL×3)萃取。有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,在减压下除去溶剂,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=2/1),得到目标产物3-碘-1H-吡唑-4-甲酸乙酯1g(6.4g,浅黄色固体),产率:80%。
MS m/z(ESI):267[M+1]
第五步
(S)-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-3-碘-1H-吡唑-4-甲酸乙酯
将3-碘-1H-吡唑-4-甲酸乙酯1g(4.5g,17mmol)、(R)-3-(甲苯磺酰氧代)吡咯烷-1-甲酸叔丁基酯1b(6.1g,17.8mmol)、碳酸铯(7.5g,20.4mmol)和N,N-二甲基甲酰胺(50mL)的混合物加热到80℃,搅拌3小时。将反应混合物冷却至室温,倒入饱和碳酸氢钠溶液(200mL)中,用乙酸乙酯(300mL×3)萃取。有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,在减压下除去溶剂,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=5/1到2/1),得到目标产物(S)-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-3-碘-1H-吡唑-4-甲酸乙酯1h(3.1g,淡黄色固体),产率:42%。
MS m/z(ESI):458[M+23]
第六步
(S)-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酸乙酯
将(S)-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-3-碘-1H-吡唑-4-甲酸乙酯1h(1g,2.25mmol)、1-乙炔基-3,5-二甲氧基苯1e(0.75g,4.5mmol)、二(三苯基膦)氯化钯(175mg,0.25mmol)、碘化亚铜(95mg,0.5mmol)、三乙胺(12.5ml)和N,N-二甲基甲酰胺(12.5mL)的混合物加热到80℃,搅拌12小时。将反应混合物冷却至室温,在减压下除去溶剂,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=2/1),得到目标产物(S)-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酸乙酯1i(0.95g,黄色油状物),产率:90%。
MS m/z(ESI):414[M+1-56]
第七步
(S)-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酸
将(S)-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酸乙酯1i(0.30g,0.64mmol)溶于四氢呋喃(3mL),加入氢氧化钠溶液(4M,2mL),室温搅拌1小时。将反应混合物减压浓缩,残余物用盐酸(6M,1mL)酸化,用乙酸乙酯(10mL×3)萃取。有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,在减压下除去溶剂,得到目标产物(S)-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酸1j(200mg,淡黄色油状物),产率:71%。
MS m/z(ESI):386[M+1-56]
第八步
(S)-3-(4-氨甲酰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯
将(S)-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酸1j(220mg,0.5mmol)、氯化铵(270mg,5mmol)、O-(7-氮杂苯并***-1-基)-N,N,N′,N′-四甲基脲鎓六氟磷酸盐(HATU)(228mg,0.6mmol)、N,N-二异丙基乙胺(129mg,1mmol)和N,N-二甲基甲酰胺(5mL)的反应混合物在室温下搅拌过夜。加水稀释,用乙酸乙酯萃取。有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,在减压下除去溶剂,残余物用薄层硅胶制备色谱纯化(二氯甲烷/甲醇=20/1),得到目标产物(S)-3-(4-氨甲酰基-3-((3,5-二甲氧基苯基)乙炔 基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯1k(140mg,白色固体),产率:64%。
MS m/z(ESI):385[M+1-56]
第九步
(S)-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺
将(S)-3-(4-氨甲酰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯1k(50mg,0.11mmol)、盐酸(6M,5mL)和二氧六环(5mL)的反应混合物在室温下搅拌1小时。在减压下除去溶剂,得到目标产物(S)-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺1l(42mg,盐酸盐,粗品),产率:100%。
MS m/z(ESI):341[M+1]
第十步
(S)-1-(1-丙烯酰基吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
向(S)-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐1l(30mg,0.08mmol)、N,N-二异丙基乙胺(31mg,0.24mmol)和四氢呋喃(15mL)的混合物中滴加丙烯酰氯(11mg,0.12mmol)的四氢呋喃(5mL)溶液,将反应混合物在室温下搅拌30分钟。加水(30mL)淬灭,用乙酸乙酯萃取。有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,在减压下除去溶剂,残余物用薄层硅胶制备色谱纯化(二氯甲烷/甲醇=20/1),得到目标产物(S)-1-(1-丙烯酰基吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺1(15mg,白色固体),产率:50%。
MS m/z(ESI):395[M+1]
1H NMR(400MHz,CDCl3)δ8.10(d,J=9.8Hz,1H),6.96(brs,1H),6.71(d,J=2.3Hz,2H),6.54-6.52(m,1H),6.46-6.39(m,2H),5.80(brs,1H),5.76-5.72(m,1H),5.01-4.92(m,1H),4.13-4.00(m,2H),3.90-3.75(m,8H),2.62-2.44(m,2H)。
实施例2
1-(1-丙烯酰基哌啶-4-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑 -4-甲酰胺
Figure PCTCN2017072570-appb-000020
第一步
3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酸乙酯
将化合物3-碘-1H-吡唑-4-甲酸乙酯1g(2.01g,7.5mmol)溶于四氢呋喃(80mL)并冷却至0℃,加入氢化钠(60%矿物油分散体,0.42g,10.5mmol),室温下搅拌1小时。向反应混合物中加入2-(三甲基甲硅烷基)乙氧基甲基氯(1.76g,10.5mmol),继续搅拌15小时。向反应混合物中加入饱和食盐水(100mL),用乙酸乙酯萃取(150mL×2)。有机相合并后用饱和食盐水洗涤(100mL),在减压下除去溶剂,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=5/1到1/2),得到目标产物3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酸乙酯2a(2.6g,无色油状物),产率:87%。
MS m/z(ESI):397[M+1]
第二步
3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酸
将化合物3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酸乙酯2a(2.6g,6.5mmol)溶于四氢呋喃(40mL),加入氢氧化锂水溶液(1M,13mL)并在室温下搅拌15小时。加水(20mL)稀释,用 盐酸(1M)酸化至pH=4-5,用乙酸乙酯萃取(50mL×3)。有机相合并后用饱和食盐水(100mL)洗涤,在减压下除去溶剂,干燥后得到目标产物3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酸2b(2.03g,白色固体),产率:85%。
MS m/z(ESI):391[M+23]
第三步
3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酰胺
将化合物3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酸2b(2.03g,5.5mmol)、二异丙基乙基胺(2.13g,16.5mmol)和N,N-二甲基甲酰胺(20mL)混合,依次加入O-(7-氮杂苯并***-1-基)-N,N,N′,N′-四甲基脲鎓六氟磷酸盐(HATU)(2.5g,6.6mmol)和1-羟基苯并***(890mg,6.6mmol),室温下搅拌1小时后加入固体氯化铵(1.47g,27.5mmol),继续搅拌15小时。向反应混合物物中加入饱和食盐水(30mL),用乙酸乙酯萃取(50mL×3)。有机相合并后用饱和食盐水洗涤(100mL),在减压下除去溶剂后,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=20/1),得到目标产物3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酰胺2c(2.3g,黄色油状物),产率:100%。
MS m/z(ESI):368[M+1]
第四步
3-((3,5-二甲氧基苯基)乙炔基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酰胺
将化合物3-碘-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酰胺2c(2.7g,7.3mmol)、1-乙炔基-3,5-二甲氧基苯(1.78g,11mmol)、三乙胺(2.2g,21.9mmol)、二(三苯基磷)氯化钯(512mg,0.73mmol)和无水四氢呋喃(70mL)混合,除氧,在氩气气氛下室温搅拌15小时。在减压下除去溶剂,残余物用硅胶柱层析纯化(乙酸乙酯/石油醚=10/1至2/1)得到目标产物3-((3,5-二甲氧基苯基)乙炔基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酰胺2d(1.5g,黄色固体),产率:51%。
MS m/z(ESI):402[M+1]
第五步
3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
将3-((3,5-二甲氧基苯基)乙炔基)-1-((2-(三甲基甲硅烷基)乙氧基)甲基)-1H-吡唑-4-甲酰胺2d(1.4g,3.5mmol)、乙二胺(525mg,8.75mmol)和四氢呋喃(30mL)混合,加入四丁基氟化铵的四氢呋喃溶液(1M,17.5mL,17.5mmol)。加热回流15小时后,冷却至室温,加入饱和食盐水(20mL),用乙酸乙酯(100mL×3)萃取。有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,在减压下除去溶剂,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=20/1),得到目标产物3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺2e(600mg,白色固体),产率:63%。
MS m/z(ESI):272[M+1]
第六步
4-(4-氨甲酰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)哌啶-1-甲酸叔丁基酯
将化合物3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺2e(180mg,0.66mmol)、4-溴哌啶-1-甲酸叔丁基酯(264mg,0.99mmol)、碳酸钾(182mg,1.32mmol)和N,N-二甲基甲酰胺(10mL)混合后,加热到75℃,搅拌15小时。加入水(30mL),用乙酸乙酯(50mL×3)萃取。有机相合并后用饱和食盐水洗涤,并用无水硫酸钠干燥。过滤除去干燥剂,在减压下除去溶剂,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=20/1),得到目标产物4-(4-氨甲酰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)哌啶-1-甲酸叔丁基酯2f(120mg,黄色固体,含区域异构体),产率:40%。
MS m/z(ESI):477[M+23]
第七步
3-((3,5-二甲氧基苯基)乙炔基)-1-(哌啶-4-基)-1H-吡唑-4-甲酰胺
将化合物4-(4-氨甲酰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)哌啶-1-甲酸叔丁基酯2f(120mg,0.26mmol,混合物)溶于乙醇(20mL),加入氯化氢的乙醇溶液(4M,1mL,4mmol),室温下搅拌15小时。在减压下除去溶剂,残余物溶于甲醇(20mL)后,用饱和碳酸氢钠溶液调节至pH=8-9。再次在减压下除去溶剂后,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=10/1),得到目标产物3-((3,5-二甲氧基苯基)乙炔基)-1-(哌啶-4-基)-1H-吡唑-4-甲酰胺2g(25mg,白色固体), 产率:27%。
MS m/z(ESI):355[M+1]
第八步
1-(1-丙烯酰基哌啶-4-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
将化合物3-((3,5-二甲氧基苯基)乙炔基)-1-(哌啶-4-基)-1H-吡唑-4-甲酰胺2g(25mg,0.07mmol)、烯丙酰氯(10mg,0.11mmol)、固体碳酸氢钠(18mg,0.21mmol)、水(2mL)和四氢呋喃(10mL)在0℃混合并在此温度下搅拌10小时。用乙酸乙酯(20mL×3)萃取,有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,在减压下除去溶剂,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=10/1),得到目标产物1-(1-丙烯酰基哌啶-4-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺2(17mg,白色固体),产率:60%。
MS m/z(ESI):409[M+1]
1H NMR(400MHz,CDCl3)δ8.10(s,1H),7.01(brs,1H),6.72(d,J=2.2Hz,2H),6.62(dd,J=16.8,10.6Hz,1H),6.55(t,J=2.2Hz,1H),6.33(dd,J=16.8,1.5Hz,1H),5.80(brs,1H),5.76(dd,J=10.6,1.6Hz,1H),4.81(brs,1H),4.40(t,J=11.4Hz,1H),4.18(brs,1H),3.82(s,6H),3.26(brs,1H),2.89(brs,1H),2.42-2.25(m,2H),2.08-2.00(m,2H)。
参照实施例2的操作步骤合成实施例3-6:
实施例3
1-(1-丙烯酰基氮杂环丁烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000021
MS m/z(ESI):381[M+1]
1H NMR(400MHz,DMSO-d6)δ8.43(s,1H),7.30(s,2H),6.73(d,J=2.2Hz,2H),6.60(t,J=2.2Hz,1H),6.38(dd,J=17.0,10.3Hz,1H),6.16(dd,J=17.0,2.1Hz,1H),5.73(dd,J=10.3,2.1Hz,1H),5.41-5.28 (m,1H),4.71(t,J=8.6Hz,1H),4.50(dd,J=9.2,4.9Hz,1H),4.46-4.36(m,1H),4.20(dd,J=10.7,4.8Hz,1H),3.78(s,6H)。
实施例4
1-((1-丙烯酰基哌啶-4-基)甲基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000022
MS m/z(ESI):423[M+1]
1H NMR(400MHz,CDCl3)δ8.00(s,1H),6.98(brs,1H),6.72(s,2H),6.61-6.54(m,2H),6.28(d,J=16.8Hz,1H),5.87(brs,1H),5.70(d,J=10.5Hz,1H),4.72(brs,1H),4.04(brs,3H),3.82(s,6H),3.05(brs,1H),2.64(brs,1H),2.27(brs,1H),1.69(brs,2H),1.24(brs,2H)。
实施例5
1-(4-丙烯酰基氨基环己基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000023
MS m/z(ESI):423[M+1]
1H NMR(400MHz,CD3OD)δ8.25(s,1H),6.77(d,J=2.3Hz,2H),6.58(t,J=2.3Hz,1H),6.38(dd,J=17.1,10.0Hz,1H),6.26(dd,J=17.1,2.0Hz,1H),5.68(dd,J=10.1,2.0Hz,1H),4.38-4.33(m,1H),4.13-4.11(m,1H),3.82(s,6H),2.28-2.18(m,2H),2.07-2.02(m,2H),1.96-1.80(m,4H)。
实施例6
3-((3,5-二甲氧基苯基)乙炔基)-1-(2-(N-甲基丙烯酰基氨基)乙 基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000024
MS m/z(ESI):383[M+1]
1H NMR(300MHz,DMSO-d6)δ8.24(s,1H),7.10-6.90(m,2H),6.76(s,2H),6.69-6.54(m,2H),6.07(d,J=16.5Hz,1H),5.64(d,J=9.8Hz,1H),4.37(t,J=5.7Hz,2H),3.89-3.80(m,8H),2.94(s,3H)。
实施例7
(S)-1-(1-丙烯酰基吡咯烷-3-基)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000025
第一步
5-氨基-3-溴-1H-吡唑-4-甲腈
将化合物5-氨基-1H-吡唑-4-甲腈7a(20g,185mmol)溶于N,N-二甲基甲酰胺(200mL),冷却至0℃,分批加入N-溴代丁二酰亚胺(34g,190mmol),升至室温搅拌2小时。将反应液倒入亚硫酸钠溶液中,用乙酸乙酯(200mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤 除去干燥剂,减压浓缩,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=20/1),得到目标产物5-氨基-3-溴-1H-吡唑-4-甲腈7b(32g,黄色固体),产率:93%。
MS m/z(ESI):187/189[M+1]
第二步
(S)-3-(5-氨基-3-溴-4-氰基-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯
将5-氨基-3-溴-1H-吡唑-4-甲腈7b(10g,53.8mmol)、3-(甲苯磺酰氧代)吡咯烷-1-甲酸叔丁基酯(22g,64.5mmol)、碳酸铯(58g,107.6mmol)和乙腈(250mL)的混合物加热到90℃反应4小时。冷却至室温,过滤,滤饼用二氯甲烷洗涤,滤液合并后减压浓缩,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=5/1),得到目标产物(S)-3-(5-氨基-3-溴-4-氰基-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7c(5g,黄色油状物),产率:26%。
MS m/z(ESI):300/302[M+1-56]
第三步
(S)-3-(5-氨基-4-氰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯
将(S)-3-(5-氨基-3-溴-4-氰基-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7c(5g,14.1mmol)、碘化亚铜(0.6g,2.8mmol)、三乙胺(9mL)、[1,1′-双(二苯基膦基)二茂铁]二氯化钯(2g,2.8mmol)和N,N-二甲基甲酰胺(150mL)的混合物在氩气保护下加热至80℃,分批加入1-乙炔基-3,5-二甲氧基苯(14g,84.5mmol),搅拌2小时。冷却至室温,将反应液倒入水中,用乙酸乙酯(200mL×3)萃取。有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=5/1),得到目标产物(S)-3-(5-氨基-4-氰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7d(5g,棕色油状物),产率:81%。
MS m/z(ESI):382[M+1-56]
第四步
(S)-3-(5-氨基-4-氨甲酰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯
将(S)-3-(5-氨基-4-氰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1- 基)吡咯烷-1-甲酸叔丁基酯7d(5g,11.4mmol)、氢氧化钠(1.5g,37.5mmol,溶于2mL水)、乙醇(50mL)和二甲基亚砜(10mL)的混合物冷却至0℃,加入双氧水(20mL),室温搅拌2小时。将反应液倒入亚硫酸钠溶液中,用乙酸乙酯(100mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=1/1),得到目标产物(S)-3-(5-氨基-4-氨甲酰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7e(5g,棕色油状物),产率:96%。
MS m/z(ESI):400[M+1-56]
第五步
(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺
将化合物(S)-3-(5-氨基-4-氨甲酰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7e(5g,11mmol)溶于二氯甲烷(100mL)中,加入三氟乙酸(15mL),室温搅拌2小时。减压浓缩,得到目标产物(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺7f(7.1g,棕色油状物,三氟乙酸盐,粗品),产率:>100%,产物不经纯化直接用于下一步反应。
MS m/z(ESI):356[M+1]
第六步
(S)-1-(1-丙烯酰基吡咯烷-3-基)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
将化合物(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺7f(7.1g,11mmol,三氟乙酸盐,粗品)溶于四氢呋喃(50mL)中,冷却至0℃,先后加入碳酸氢钠饱和溶液(20mL)和丙烯酰氯(900mg,10mmol),搅拌30分钟。将反应液倒入水(100mL)中,并用二氯甲烷(100mL×3)萃取。有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=1/2),得到目标产物(S)-1-(1-丙烯酰基吡咯烷-3-基)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺7(1.9g,白色固体),产率:42%。
MS m/z(ESI):410[M+1]
1H NMR(400MHz,DMSO-d6)δ7.18(brs,1H),6.75(d,J=2.3Hz,2H),6.69-6.55(m,3H),6.20-6.14(m,1H),5.72-5.67(m,1H),5.03-4.91(m,1H),4.01-3.96(m,1H),3.84-3.70(m,7H),3.66-3.60(m,1H),3.55-3.48(m,1H),2.36-2.21(m,2H)。
实施例8
(S,E)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(1-(4-(二甲氨基)丁-2-烯酰基)吡咯烷-3-基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000026
第一步
(S,E)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(1-(4-(二甲氨基)丁-2-烯酰基)吡咯烷-3-基)-1H-吡唑-4-甲酰胺
将(E)-4-(二甲氨基)丁-2-烯酸(23mg,0.14mmol)、O-(7-氮杂苯并***-1-基)-N,N,N′,N′-四甲基脲鎓六氟磷酸盐(HATU)(64mg,0.17mmol)、(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺7f(50mg,0.14mmol)、N,N-二异丙基乙胺(2mL)和二氯甲烷(3mL)的反应混合物在室温下搅拌1小时。将反应液倒入水中,用二氯甲烷(20mL×3)萃取。有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用高效液相制备色谱纯化,得到目标产物(S,E)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(1-(4-(二甲氨基)丁-2-烯酰基)吡咯烷-3-基)-1H-吡唑-4-甲酰胺8(2.4mg,白色固体,甲酸盐),产率:4%。
MS m/z(ESI):467[M+1]
1H NMR(400MHz,DMSO-d6)δ8.27(brs,1H),7.20(brs,1H),6.75(d,J=2.3Hz,2H),6.70-6.61(m,3H),6.44-6.35(m,1H),5.01-4.93(m,1H),4.01-3.93(m,1H),3.77(s,6H),3.74-3.64(m,3H),3.06-3.03(m, 2H),2.38-2.24(m,2H),2.17-2.15(m,6H)。
实施例9
(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(1-(2-氟丙烯酰基)吡咯烷-3-基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000027
第一步
(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(1-(2-氟丙烯酰基)吡咯烷-3-基)-1H-吡唑-4-甲酰胺
将化合物(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺7f(50mg,0.14mmol)和2-氟丙烯酸(15mg,0.17mmol)溶于二氯甲烷,加入N,N-二异丙基乙胺(54mg,0.42mmol)和O-(7-氮杂苯并***-1-基)-N,N,N′,N′-四甲基脲鎓六氟磷酸盐(HATU)(69mg,0.18mmol),室温搅拌2小时。加水(10mL)稀释反应混合物,用二氯甲烷(10mL×3)萃取,有机相合并后减压浓缩。残余物用薄层硅胶制备色谱纯化(二氯甲烷/甲醇=20/1),得到目标产物(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(1-(2-氟丙烯酰基)吡咯烷-3-基)-1H-吡唑-4-甲酰胺9(3.6mg,白色固体),产率:6%。
MS m/z(ESI):428[M+1]
1H NMR(400MHz,CD3OD)δ6.62(t,J=2.5Hz,2H),6.47(t,J=2.3Hz,1H),5.39(dd,J=47.2,3.5Hz,1H),5.16(ddd,J=16.6,5.7,3.5Hz,1H),4.86-4.81(m,1H),4.02-3.91(m,2H),3.87-3.72(m,2H),3.71(s,6H),2.34-2.23(m,2H)。
实施例10
(S)-5-氨基-1-(1-(丁-2-炔酰基)吡咯烷-3-基)-3-((3,5-二甲氧基苯基) 乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000028
第一步
(S)-5-氨基-1-(1-(丁-2-炔酰基)吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
将(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺7f(50mg,0.14mmol)和2-丁炔酸(14mg,0.17mmol)溶于二氯甲烷中,加入N,N-二异丙基乙胺(54mg,0.42mmol)和O-(7-氮杂苯并***-1-基)-N,N,N′,N′-四甲基脲鎓六氟磷酸盐(HATU)(69mg,0.18mmol),室温搅拌2小时。反应混合物用水(10mL)稀释,用二氯甲烷(10mL×3)萃取,有机相合并后减压浓缩。残余物用薄层硅胶制备色谱纯化(二氯甲烷/甲醇=20/1),得到目标产物(S)-5-氨基-1-(1-(丁-2-炔酰基)吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺10(5.1mg,淡黄色固体),产率:9%。
MS m/z(ESI):422[M+1]
1H NMR(400MHz,CD3OD)δ6.62(t,J=2.1Hz,2H),6.47(t,J=2.2Hz,1H),4.85-4.81(m,1H),4.01-3.86(m,2H),3.77-3.62(m,7.5H),3.54-3.46(m,0.5H),2.32-2.27(m,2H),1.95-1.93(m,3H)。
实施例11
(S,E)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(1-(4-甲氧基丁-2-烯酰基)吡咯烷-3-基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000029
Figure PCTCN2017072570-appb-000030
第一步
(E)-4-溴丁-2-烯酸
将(E)-4-溴丁-2-烯酸甲酯11a(3g,16.8mmol)、氢氧化锂一水合物(1.1g,25.3mmol)、四氢呋喃(50mL)和水(50mL)在0℃混合并继续搅拌2小时。反应结束后,用石油醚洗去四氢呋喃,水相用2M盐酸调节至pH=1,然后用乙酸乙酯(100mL×2)萃取。有机相合并后在减压下除去溶剂,得到目标产物(E)-4-溴丁-2-烯酸11b(2.3g,黄色油状物),产率:83%。
MS m/z(ESI):163[M-1]
第二步
(E)-4-甲氧基丁-2-烯酸
将化合物(E)-4-溴丁-2-烯酸11b(100mg,0.61mmmol)溶于甲醇(5mL)后,加入甲醇钠的甲醇溶液(30%,0.55mL,3.05mmol)并搅拌15小时。反应混合物在减压下除去溶剂后溶于水,并用稀盐酸调节至pH=1,然后用二氯甲烷萃取(10mL×3)。有机相合并后在减压下除去溶剂,得到目标产物(E)-4-甲氧基丁-2-烯酸11c(50mg,黄色油状物),产率:71%。
1H NMR(400MHz,CDCl3)δ7.13-7.03(m,1H),6.15-6.07(m,1H),4.18-4.11(m,2H),3.48-3.38(s,3H)。
第三步
(S,E)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(1-(4-甲氧基丁-2-烯酰基)吡咯烷-3-基)-1H-吡唑-4-甲酰胺
将化合物(E)-4-甲氧基丁-2-烯酸11c(22mg,0.19mmol)、二异丙基乙基胺(67mg,0.52mmol)、(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺7f(50mg,0.13mmol)、2-(7-氧化苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(72mg,0.19mmol)和N,N-二甲基甲酰胺(10mL)混合并搅拌2小时。在减压下除去溶剂,残余物溶于乙酸乙酯(30mL)后依次用水和饱和食盐水洗涤。在减压下除去溶剂,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=20/1)得到 目标产物(S,E)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(1-(4-甲氧基丁-2-烯酰基)吡咯烷-3-基)-1H-吡唑-4-甲酰胺11(30mg,白色固体),产率:51%。
MS m/z(ESI):454[M+1]
1H NMR(400MHz,CDCl3)δ6.98(d,J=15.3Hz,1H),6.86(brs,1H),6.72(d,J=2.1Hz,2H),6.54(s,1H),6.39(dd,J=27.7,16.0Hz,1H),5.54(brs,1H),4.73-4.70(m,1H),4.14-4.12(m,2H),4.05-4.00(m,2H),3.95-3.93(m,1H),3.82(s,6H),3.77-3.68(m,1H),3.43(d,J=10.1Hz,3H),2.72(brs,0.5H),2.54(brs,0.5H),2.43-2.35(m,1H)。
实施例12
(S)-5-氨基-1-(1-氰基吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000031
第一步
(S)-5-氨基-1-(1-氰基吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
将化合物(S)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺7f(50mg,0.14mmol)溶于四氢呋喃(2mL),加入三乙胺(1mL),冷却至0℃,加入溴化氰(17mg,0.15mmol),0℃搅拌2小时,升至室温,继续搅拌2小时。反应混合物减压浓缩,残余物用薄层硅胶制备色谱纯化(二氯甲烷/甲醇=15/1),得到目标产物(S)-5-氨基-1-(1-氰基吡咯烷-3-基)-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺12(18mg,白色固体),产率:34%。
MS m/z(ESI):381[M+1]
1H NMR(400MHz,CDCl3)δ6.77(brs,1H),6.68(d,J=1.9Hz,2H), 6.50(s,1H),5.75(s,2H),5.67(brs,1H),4.79-4.73(m,1H),3.84-3.73(m,9H),3.61-3.53(m,1H),2.53-2.43(m,1H),2.37-2.26(m,1H)。
参照实施例7的操作步骤合成了实施例13-16:
实施例13
(R)-1-(1-丙烯酰基吡咯烷-3-基)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000032
MS m/z(ESI):410[M+1]
1H NMR(400MHz,CD3OD)δ6.73(d,J=1.9Hz,2H),6.71-6.60(m,1H),6.58(brs,1H),6.32(dd,J=16.8,1.7Hz,1H),5.84-5.74(m,1H),5.04-4.91(m,1H),4.09(m,0.5H),3.98(td,J=11.1,4.0Hz,1H),3.91(dd,J=7.8,5.6Hz,1H),3.86(dd,J=9.9,4.4Hz,1H),3.81(s,6H),3.73-3.63(m,0.5H),2.47(dd,J=13.2,6.7Hz,1H),2.38(dd,J=13.6,7.0Hz,1H)。
实施例14
1-(1-丙烯酰基氮杂环丁烷-3-基)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000033
MS m/z(ESI):396[M+1]
1H NMR(400MHz,CD3OD)δ6.75(d,J=2.3Hz,2H),6.60(t,J=2.2Hz,1H),6.45-6.28(m,2H),5.80(dd,J=10.1,2.1Hz,1H),5.29-5.21(m,1H),4.79-4.64(m,2H),4.54-4.47(m,1H),4.46-4.39(m,1H),3.82(s,6H)。
实施例15
1-(1-丙烯酰基哌啶-4-基)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000034
MS m/z(ESI):424[M+1]
1H NMR(400MHz,CD3OD)δ6.88-6.78(m,1H),6.73(d,J=2.2Hz,2H),6.58(t,J=2.2Hz,1H),6.24(dd,J=16.8,1.7Hz,1H),5.78(dd,J=10.7,1.7Hz,1H),4.73(d,J=13.2Hz,1H),4.47-4.36(m,1H),4.30(d,J=13.3Hz,1H),3.81(s,6H),3.32-3.24(m,1H),2.91(t,J=9.9Hz,1H),2.02(d,J=4.5Hz,4H)。
实施例16
1-((1-丙烯酰基吡咯烷-3-基)甲基)-5-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000035
MS m/z(ESI):424[M+1]
1H NMR(400MHz,CD3OD)δ6.73(s,2H),6.66-6.55(m,2H),6.28(d,J=16.7Hz,1H),5.75(d,J=10.4Hz,1H),4.13-3.99(m,2H),3.82(s,6H),3.78-3.61(m,2H),3.48(dd,J=14.8,7.4Hz,1H),3.39-3.34(m,1H),2.94-2.75(m,1H),2.20-2.02(m,1H),1.94-1.71(m,1H)。
实施例17
(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((2-氟-3,5-二甲氧苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000036
Figure PCTCN2017072570-appb-000037
第一步
1-乙炔基-2-氟-3,5-二甲氧基苯
将混合物1-乙炔基-3,5-二甲氧基苯1e(2g,12.3mmol)溶于乙腈(15mL)中,降温至0℃,并分批加入1-氯甲基-4-氟-1,4-重氮化二环2.2.2辛烷双(四氟硼酸)盐(6.6g,18.5mmol),然后室温搅拌过夜。将反应液倒入水(50mL)中,并用二氯甲烷(30mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=30/1),得到目标产物1-乙炔基-2-氟-3,5-二甲氧基苯17a(800mg,黄色固体),产率:36%。
1H NMR(400MHz,CDCl3)δ6.46(dd,J=6.9,2.9Hz,1H),6.41(dd,J=4.5,3.0Hz,1H),3.78(s,3H),3.69(s,3H),3.22(s,1H)。
参照实施例7第一至第六步的操作步骤合成实施例17,但在第三步中用1-乙炔基-2-氟-3,5-二甲氧基苯取代1-乙炔基-3,5-二甲氧基苯。
MS m/z(ESI):428[M+1]
1H NMR(400MHz,CDCl3)δ7.00(brs,1H),6.59-6.57(m,2H),6.49-6.39(m,2H),5.74-5.70(m,1H),5.52(d,J=8.5Hz,2H),5.35(brs,1H),4.73-4.64(m,1H),4.07-3.90(m,3H),3.88(s,3H),3.78(d,J=5.3Hz,3H),3.75-3.67(m,1H),2.72-2.67(m,0.5H),2.54-2.31(m,1.5H)。
实施例18
(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((5-氯-2-氟苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000038
第一步
((2-氟-5-氯苯基)乙炔基)三甲基硅烷
将2-氟-5-氯溴苯18a(11.0g,52.8mmol)、乙炔基三甲基硅烷(7.7g,79mmol)和三乙胺(60mL)混合,然后加入碘化亚铜(100mg,0.53mmol)和二三苯基膦氯化钯(1.86g,2.65mmol)。反应混合物在氮气保护气氛下加热到80℃并继续搅拌4小时。反应结束后,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=100/1)得到目标产物((2-氟-5-氯苯基)乙炔基)三甲基硅烷18b(11.0g,黄色油状物),产率:90%。
1H NMR(400MHz,CDCl3)δ7.45(dd,J=6.0,2.7Hz,1H),7.28-7.22(m,1H),7.02(t,J=8.8Hz,1H),0.29(s,9H)。
第二步
4-氯-2-乙炔基-1-氟苯
将((2-氟-5-氯苯基)乙炔基)三甲基硅烷18b(11.0g,48mmol)、碳酸钾(8.1g,58mmol)、二氯甲烷(80mL)和甲醇(40mL)混合后,室温下搅拌18小时。反应结束后,减压脱溶,残余物用硅胶柱层析(石油醚/乙酸乙酯=100/1)纯化得到目标产物4-氯-2-乙炔基-1-氟苯18c(5.5g,黄色固体),产率:74%。
1H NMR(400MHz,CDCl3)δ7.45(dd,J=6.0,2.7Hz,1H),7.31-7.27(m,1H),7.04(t,J=8.0,1H),3.35(s,1H)。
参照实施例7第一至第六步的操作步骤合成实施例18,但在第三步中用4-氯-2-乙炔基-1-氟苯取代1-乙炔基-3,5-二甲氧基苯。
MS m/z(ESI):402[M+1]
1H NMR(400MHz,CD3OD)δ7.62-7.61(m,1H),7.47-7.45(m,1H),7.24(t,J=9.0Hz,1H),6.69-6.56(m,1H),6.30(d,J=16.8Hz,1H),5.77(t,J=9.2Hz,1H),5.02-1.91(m,1H),4.09-3.95(m,2H),3.84-3.78(m,2H),2.46(dd,J=13.1,6.6Hz,1H),2.37(dd,J=13.6,6.9Hz,1H)。
实施例19
(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((2-氯-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000039
第一步
4-氯-3-溴苯甲酸甲酯
将4-氯-3-溴苯甲酸19a(2g,8.5mmol)溶于甲醇(400mL)并冷却至0℃,然后逐滴加入乙酰氯(2.3g,30mmol)并继续搅拌18小时。反应结束后减压脱溶,残余物用硅胶柱层析(石油醚/乙酸乙酯=10/1)纯化得到目标产物4-氯-3-溴苯甲酸甲酯19b(1.2g,黄色固体),产率:57%。
1H NMR(400MHz,CDCl3)δ8.31(d,J=1.9Hz,1H),7.93(dd,J=8.3,1.9Hz,1H),7.55(d,J=8.3Hz,1H),3.95(s,3H)。
第二步
4-氯-3-((三甲基甲硅烷基)乙炔基)苯甲酸甲酯
将化合物4-氯-3-溴苯甲酸甲酯19b(1.2g,4.8mmol)、三甲基硅基乙炔(0.95g,9.7mmol)、醋酸钯(108mg,0.48mmol)、三苯基膦(254mg,0.97mmol)、碘化亚铜(185mg,0.97mmol)和三乙胺(25mL)在封管中混合并在100℃下加热并搅拌15小时。反应完成后减压脱溶, 残余物用柱层析硅胶色谱(石油醚/乙酸乙酯=10/1)纯化得到目标产物4-氯-3-((三甲基甲硅烷基)乙炔基)苯甲酸甲酯19c(1g,黄色固体),产率:78%。
1H NMR(400MHz,CDCl3)δ8.19(d,J=2.0Hz,1H),7.91(dd,J=8.4,2.1Hz,1H),7.48(d,J=8.4Hz,1H),3.94(s,3H),0.30(s,9H)。
第三步
4-氯-3-乙炔基苯甲酸甲酯
将4-氯-3-((三甲基甲硅烷基)乙炔基)苯甲酸甲酯19c(1g,3.76mmol)溶于甲醇(20mL),然后加入碳酸钾(1.04g,7.52mmol)。室温搅拌1小时后,减压脱溶。残余物用水洗涤并过滤得到目标产品4-氯-3-乙炔基苯甲酸甲酯19d(380mg,黄色固体),产率:52%。
1H NMR(400MHz,CDCl3)δ8.23(d,J=2.1Hz,1H),7.96(dd,J=8.4,2.0Hz,1H),7.51(d,J=8.4Hz,1H),3.95(s,3H),3.44(s,1H)。
第四步
(S)-3-(5-氨基-3-溴-4-氨基甲酰-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将化合物(S)-3-(5-氨基-3-溴-4-氰基-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯7c(2.20g,6.2mmol)、氢氧化钠水溶液(0.5M,12.4mL,6.2mmol)、过氧化氢水溶液(30%,15mL)和二甲基亚砜(30mL)混合。室温搅拌2小时后,反应物用饱和食盐水(50mL)稀释,并用乙酸乙酯(50mL×3)萃取。有机相合并后减压脱溶,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=100/1至20/1)得到目标产物(S)-3-(5-氨基-3-溴-4-氨基甲酰-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯19e(1.92g,淡黄色固体),产率:83%。
MS m/z(ESI):374[M+1]
第五步
(S)-3-(5-氨基-4-氨基甲酰-3-((2-氯-5-(甲酯基<甲氧羰基>)苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将(S)-3-(5-氨基-3-溴-4-氨基甲酰-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯19e(770mg,2.1mmol)、三乙胺(6mL)、1,1′-双二苯基膦二茂铁二氯化钯(307mg,0.42mmol)、碘化亚铜(80mg,0.42mmol)和N,N-二甲基甲酰胺(20mL)混合,除氧,在氩气气氛下加热至90℃,然后逐滴加入4-氯-3-乙炔基苯甲酸甲酯19d(3.20g,16.5mmol)的N,N- 二甲基甲酰胺(2mL)溶液,并继续搅拌12小时。减压脱溶,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=20/1)得到目标产物(S)-3-(5-氨基-4-氨基甲酰-3-((2-氯-5-(甲酯基<甲氧羰基>)苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯19f(420mg,黄色固体),产率:41%。
MS m/z(ESI):488[M+1]
第六步
(S)-3-((5-氨基-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-4-氨基甲酰-1H-吡唑-3-基)乙炔基)-4-氯苯甲酸
将(S)-3-(5-氨基-4-氨基甲酰-3-((2-氯-5-(甲酯基<甲氧羰基>)苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯19f(100mg,0.2mmol)溶于甲醇(4mL)和水(4mL)的混合溶剂中,然后加入氢氧化钠(25mg,0.61mmol)并继续搅拌2小时。反应完成后,在减压条件下除去有机溶剂。残余物用盐酸(1M)调节至pH=4至5,然后用乙酸乙酯萃取(30mL×2)。有机相合并后用饱和食盐水洗涤,无水硫酸钠干燥后过滤。滤液减压脱溶得到目标产物(S)-3-((5-氨基-1-(1-(叔丁氧基羰基)吡咯烷-3-基)-4-氨基甲酰-1H-吡唑-3-基)乙炔基)-4-氯苯甲酸19g(80mg,棕色固体),产率:84%。
MS m/z(ESI):418[M+H-56]
第七步
(S)-3-(5-氨基-4-氨基甲酰-3-((2-氯-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将(S)-3-((5-氨基-1-(1-(叔-丁氧基羰基)吡咯烷-3-基)-4-氨基甲酰-1H-吡唑-3-基)乙炔基)-4-氯苯甲酸19g(80mg,0.17mmol)溶于N,N-二甲基甲酰胺(2.5mL)中,然后依次加入甲胺盐酸盐(34mg,0.50mmol)、二异丙基乙基胺(129mg,1mmol)和2-(7-氧化苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(64mg,0.17mmol)。反应物室温搅拌2小时后用水淬灭,然后用乙酸乙酯(20mL×3)萃取。有机相合并后减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=100/1至0/1)得到目标产物(S)-3-(5-氨基-4-氨基甲酰-3-((2-氯-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯19h(41mg,棕色固体),产率:50%。
MS m/z(ESI):387[M+H-Boc]
第八步
(S)-5-氨基-3-((2-氯-5-(甲基氨基甲酰)苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐
将(S)-3-(5-氨基-4-氨基甲酰-3-((2-氯-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯19h(40mg,0.08mmol)溶于乙酸乙酯(5mL)中,然后加入氯化氢的乙醇溶液(33%,3mL)并在室温下搅拌1小时。反应完成后,减压脱溶,得到目标产物(S)-5-氨基-3-((2-氯-5-(甲基氨基甲酰)苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐19i(40mg,粗品,棕色固体),该产品未经进一步纯化,直接用于下一步反应。
MS m/z(ESI):387[M+H]
第九步
(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((2-氯-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-4-甲酰胺
将化合物(S)-5-氨基-3-((2-氯-5-(甲基氨基甲酰)苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐19i(40mg,0.08mmol,粗品)、烯丙酰氯(7.5mg,0.08mmol)、碳酸钾水溶液(0.4M,1.0mL,0.4mmol)和四氢呋喃(5mL)在0℃混合并在此温度下搅拌0.5小时。用乙酸乙酯(20mL×2)萃取,有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=100/1至10/1),得到目标产物(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((2-氯-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-4-甲酰胺19(18mg,白色固体),产率:两步51%。
MS m/z(ESI):441[M+H]
1H NMR(400MHz,DMSO-d6)δ8.65(s,1H),8.18(s,1H),7.90(d,J=8.0Hz,1H),7.72(d,J=8.3Hz,1H),7.43(s,1H),6.70-6.62(m,4H),6.19-6.15(m,1H),5.70(t,J=10.2Hz,1H),5.03-4.94(m,1H),3.80-3.54(m,4H),2.78(d,J=3.8Hz,3H),2.36-2.25(m,2H)。
实施例20
(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((3-甲氧基-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000040
第一步
3-溴-5-甲氧基-N-甲基苯酰胺
将3-溴-5-甲氧基苯甲酸20a(500mg,2.17mmol)溶于N,N-二甲基甲酰胺(15mL)中,然后依次加入甲胺盐酸盐(291mg,4.35mmol)、二异丙基乙基胺(1.12g,8.68mmol)和2-(7-氧化苯并三氮唑)-N,N,N′,N′-四甲基脲六氟磷酸酯(1.24g,3.26mmol)。反应物室温搅拌2小时后用水淬灭,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=1/1)得到目标产物3-溴-5-甲氧基-N-甲基苯酰胺20b(500mg,白色固体),产率:95%。
MS m/z(ESI):244[M+H]
第二步
3-甲氧基-N-甲基-5-((三甲基甲硅烷基)乙炔基)苯酰胺
将化合物3-溴-5-甲氧基-N-甲基苯酰胺20b(500mg,2.1mmol)、三甲基硅基乙炔(302mg,3.1mmol)、醋酸钯(47mg,0.21mmol)、三苯基膦(110mg,0.42mmol)、碘化亚铜(80mg,0.42mmol)和三乙胺(20mL)在封管中混合,加热至100℃,搅拌15小时。反应完成后,减压脱溶,残余物用硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化,得到目标产物3-甲氧基-N-甲基-5-((三甲基甲硅烷基)乙炔基)苯酰胺 20c(220mg,黄色固体),产率:41%。
MS m/z(ESI):262[M+H]
第三步
3-乙炔基-5-甲氧基-N-甲基苯酰胺
将3-甲氧基-N-甲基-5-((三甲基甲硅烷基)乙炔基)苯酰胺20c(220mg,0.84mmol)溶于甲醇(8mL),然后加入碳酸钾(233mg,1.68mmol)。室温搅拌1小时后,减压脱溶。残余物用硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化,得到目标产物3-乙炔基-5-甲氧基-N-甲基苯酰胺20d(140mg,淡黄色固体),产率:88%。
MS m/z(ESI):190[M+H]
第四步
(S)-3-(5-氨基-4-氨基甲酰-3-((3-甲氧基-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将(S)-3-(5-氨基-3-溴-4-氨基甲酰-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯19e(329mg,0.88mmol)、三乙胺(2mL)、1,1′-双二苯基膦二茂铁二氯化钯(129mg,0.2mmol)、碘化亚铜(34mg,0.18mmol)和N,N-二甲基甲酰胺(8mL)混合,除氧,在氩气气氛下加热至90℃。然后逐滴加入3-乙炔基-5-甲氧基-N-甲基苯酰胺20d(1.00g,5.3mmol)的N,N-二甲基甲酰胺(2mL)溶液并继续搅拌12小时。减压脱溶,残余物用硅胶柱层析(二氯甲烷/甲醇=20/1)纯化,得到目标产物(S)-3-(5-氨基-4-氨基甲酰-3-((3-甲氧基-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯20e(400mg,粗品,棕色固体)。
MS m/z(ESI):383[M+H-100]
第五步
(S)-5-氨基-3-((3-甲氧基-5-(甲基氨基甲酰)苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐
将(S)-3-(5-氨基-4-氨基甲酰-3-((3-甲氧基-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯20e(400mg,粗品)溶于二氯甲烷(5mL)中,然后加入氯化氢的乙醇溶液(30%,3mL),并在室温下搅拌1小时。反应完成后,减压脱溶,得到目标产物(S)-5-氨基-3-((3-甲氧基-5-(甲基氨基甲酰)苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐20f(300mg,粗品,棕色固体)。产品不经纯化, 直接用于下一步反应。
MS m/z(ESI):383[M+H]
第六步
(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((3-甲氧基-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-4-甲酰胺
将化合物(S)-5-氨基-3-((3-甲氧基-5-(甲基氨基甲酰)苯基)乙炔基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐20f(150mg,0.39mmol,粗品)、烯丙酰氯(42mg,0.47mmol)、碳酸氢钠(131mg,1.56mmol)、水(4mL)和四氢呋喃(8mL)在0℃混合并在此温度下搅拌0.5小时。用乙酸乙酯(20mL×2)萃取,有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶,残余物用硅胶柱层析(二氯甲烷/甲醇=20/1)纯化,得到目标产物(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((3-甲氧基-5-(甲基氨基甲酰)苯基)乙炔基)-1H-吡唑-4-甲酰胺20(60mg,白色固体),产率:两步35%。
MS m/z(ESI):437[M+H]
1H NMR(400MHz,CD3OD)δ7.59(s,1H),7.45(s,1H),7.28(s,1H),6.73-6.58(m,1H),6.36-6.28(m,1H),5.83-5.75(m,1H),5.04-4.93(m,1H),4.12-3.91(m,2H),3.89(s,3H),3.86-3.66(m,2H),2.93(s,3H),2.51-2.44(m,1H),2.42-2.34(m,1H)。
实施例21
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000041
Figure PCTCN2017072570-appb-000042
第一步
(S)-3-(3-溴-4-氰基-5-(甲基氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将化合物(S)-3-(5-氨基-3-溴-4-氰基-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯7c(178mg,0.5mmol)和对甲基苯磺酸一水和物(12mg,0.07mmol)溶于原甲酸三乙酯(4mL)中,加热回流2小时。反应结束后减压脱溶,残余物分散在水中,然后用乙酸乙酯(30mL×2)萃取。有机相合并后,用无水硫酸钠干燥。过滤后滤液减压脱溶,残余物溶于乙醇(10mL)。冷却至0℃后,加入硼氢化钠(89mg,2.35mmol)并在室温下搅拌2小时。反应完成后,用饱和食盐水淬灭,然后用乙酸乙酯(30mL×2)萃取。有机相合并后,减压脱溶,残余物用硅胶柱层析(石油醚/乙酸乙酯=100/1至1/1)纯化得到目标产物(S)-3-(3-溴-4-氰基-5-(甲基氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯21a(178mg,白色固体),产率:100%。
MS m/z(ESI):314[M+H-56]
第二步
(S)-3-(4-氰基-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将(S)-3-(3-溴-4-氰基-5-(甲基氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯21a(1.85g,5.0mmol)、三乙胺(20mL)、1,1′-双二苯基膦二茂铁二氯化钯(816mg,1mmol)、碘化亚铜(190mg,1mmol)和N,N-二甲基甲酰胺(20mL)混合,除氧,在氩气气氛下加热至90℃,然后 逐滴加入1-乙炔基-3,5-二甲氧基苯(4.86g,30mmol)的N,N-二甲基甲酰胺(10mL)溶液并继续搅拌12小时。减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=50/1至0/1)得到目标产物(S)-3-(4-氰基-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯21b(2.1g,棕色固体),产率:80%。
MS m/z(ESI):496[M+H-56]
第三步
(S)-3-(5-((叔丁氧基羰基)(甲基)氨基)-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将(S)-3-(4-氰基-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯21b(225mg,0.5mmol)溶于二氯甲烷(10mL),然后依次加入三乙胺(150mg,1.5mmol)、Boc酸酐(218mg,1mmol)和4-二甲氨基吡啶(6mg,0.05mmol)。室温下搅拌2小时后,加入饱和食盐水(10mL)并用乙酸乙酯(20mL×2)萃取。有机相合并后,减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=50/1至1/1)得到目标产物(S)-3-(5-((叔丁氧基羰基)(甲基)氨基)-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯21c(200mg,淡黄色固体),产率:72%。
MS m/z(ESI):440[M+H-112]
第四步
(S)-3-(5-((叔丁氧基羰基)(甲基)氨基)-4-氨基甲酰-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将(S)-3-(5-((叔丁氧基羰基)(甲基)氨基)-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯21c(55mg,0.1mmol)、氢氧化钠水溶液(0.5M,0.1mL,0.05mmol)、过氧化氢水溶液(30%,0.5mL)和二甲基亚砜(1mL)混合,室温搅拌2小时。反应物用饱和食盐水(10mL)稀释并用乙酸乙酯(20mL×2)萃取。有机相合并后减压脱溶,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=100/1至1/100)得到(S)-3-(5-((叔丁氧基羰基)(甲基)氨基)-4-氨基甲酰-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯21d(30mg,棕色固体),产率:50%。
MS m/z(ESI):414[M+H-156]
第五步
(S)-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐
将(S)-3-(5-((叔丁氧基羰基)(甲基)氨基)-4-氨基甲酰-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯21d(570mg,1mmol)溶于乙酸乙酯(10mL)中,然后加入氯化氢的乙醇溶液(33%,5mL),并在室温下搅拌1小时。反应完成后,减压脱溶,得到目标产物(S)-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐21e(400mg,粗品,棕色固体),该产品未经进一步纯化,直接用于下一步反应。
MS m/z(ESI):370[M+H]
第六步
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1H-吡唑-4-甲酰胺
将化合物(S)-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺盐酸盐21e(870mg,2.35mmol,粗品)、烯丙酰氯(254mg,2.82mmol)、碳酸钾水溶液(2.5M,4.7mL,11.78mmol)和四氢呋喃(10mL)在0℃混合并在此温度下搅拌0.5小时。用乙酸乙酯(50mL×2)萃取,有机相合并后用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶。残余物用硅胶柱层析纯化(二氯甲烷/甲醇=100/1至10/1)得到目标产物(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1H-吡唑-4-甲酰胺21(720mg,白色固体),产率:76%。
MS m/z(ESI):424[M+H]
1HNMR(400MHz,CDCl3)δ6.88(s,1H),6.69(d,J=2.3Hz,2H),6.51(t,J=2.2Hz,1H),6.46-6.40(m,2H),5.74-5.72(m,1H),5.52-5.48(m,1H),5.06-5.01(m,1H),4.09-3.94(m,3H),3.80(s,6H),3.72-3.70(m,1H),3.00(s,3H),2.71-2.56(m,1H),2.45-2.35(m,1H)。
实施例22
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((2-氟-3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000043
参照实施例21的操作步骤合成实施例22,但在第二步中用1-乙炔基-2-氟-3,5-二甲氧基苯取代1-乙炔基-3,5-二甲氧基苯。
MS m/z(ESI):442[M+H]
1H NMR(400MHz,CDCl3)δ7.08(s,1H),6.68(d,J=7.2Hz,1H),6.60-6.57(m,2H),6.51-6.40(m,2H),5.74-5.69(m,1H),5.35(s,1H),5.08-4.99(m,1H),4.11-4.08(m,1H),4.05-3.94(m,2H),3.88(s,3H),3.79(s,3H),3.75-3.65(m,1H),3.00(t,J=5.2Hz,3H),2.72-2.58(m,1H),2.44-2.33(m,1H)。
实施例23
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((5-氯-2-氟苯基)乙炔基)-5-(甲基氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000044
参照实施例21的操作步骤合成实施例23,但在第二步中用4-氯-2-乙炔基-1-氟苯取代1-乙炔基-3,5-二甲氧基苯。
MS m/z(ESI):416[M+H]
1H NMR(400MHz,CDCl3)δ7.56-7.52(m,1H),7.35-7.33(m,1H),7.08(t,J=8.8Hz,1H),7.02-6.92(m,1H),6.51-6.39(m,2H),5.74(d,J=9.3Hz,1H),5.55-5.44(m,1H),5.09-4.98(m,1H),4.14-3.90(m,3H),3.80-3.65(m,1H),3.01(s,3H),2.74-2.55(m,1H),2.49-2.34(m,1H)。
实施例24
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-(乙基氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000045
第一步
(S)-3-(5-乙基氨基-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将混合物(S)-3-(5-氨基-4-氰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7d(500mg,1.14mmol)和氢化钠(91mg,2.28mmol,60%)加入到N,N-二乙基乙酰胺(5mL)中,搅拌10分钟,加入碘乙烷(106mg,0.68mmol),搅拌0.5小时。将反应液倒入水中,减压浓缩,残余物用反相高效液相制备色谱纯化[乙腈/水(含0.1%甲酸):50%-90%],得到目标产物(S)-3-(5-乙基氨基-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯24a(70mg,白色固体),产率:22%。
MS m/z(ESI):410[M+1-56]
第二步
(S)-3-(5-乙基氨基-4-氨基甲酰-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将(S)-3-(5-乙基氨基-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯24a(55mg,0.12mmol)溶于二甲基亚砜(3mL),加入双氧水(2mL)和氢氧化钠(300mg,7.5mmol),常温搅拌10分钟后,升温至40℃。待反应完成,冷却后用水(20mL)稀释,乙酸乙酯(30mL)萃取,并用水(20mL×3)洗涤,有机相减压浓缩,残余物用反相高效液相制备色谱纯化[乙腈/水(含0.1%甲酸): 50%-90%],得到目标产物(S)-3-(5-乙基氨基-4-氨基甲酰-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯24b(15mg),产率:26%。
MS m/z(ESI):484[M+1]
第三步
(S)-3-((3,5-二甲氧苯基)乙炔基)-5-(乙基氨基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺
将(S)-3-(5-乙基氨基-4-氨基甲酰-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯24b(15mg,0.031mmol)溶于二氯甲烷(2mL),加入三氟乙酸(0.5mL),搅拌半小时。待反应完成,减压浓缩,得到目标产物(S)-3-((3,5-二甲氧苯基)乙炔基)-5-(乙基氨基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺24c(20mg,粗品,棕色油状物),产率:>100%。产物不经纯化直接用于下一步反应。
MS m/z(ESI):384[M+1]
第四步
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-(乙基氨基)-1H-吡唑-4-甲酰胺
将化合物(S)-3-((3,5-二甲氧苯基)乙炔基)-5-(乙基氨基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺24c(20mg,0.031mmol,粗品)溶于四氢呋喃(5mL),加入饱和碳酸氢钠溶液(2mL),再加入丙烯酰氯(2.7mg,0.03mmol)的四氢呋喃溶液,搅拌0.5小时。反应液减压浓缩,残余物溶于乙酸乙酯(30mL),并用水(20mL×3)洗涤。有机相减压浓缩,残余物用反相高效液相制备色谱纯化[乙腈/水(含0.1%甲酸):20%-70%],得到目标产物(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-(乙基氨基)-1H-吡唑-4-甲酰胺24(4.7mg,白色固体),产率:24%。
MS m/z(ESI):438[M+1]
1H NMR(400MHz,CDCl3)δ8.87(brs,1H),6.74(s,2H),6.54(s,1H),6.52(s,1H),6.48-6.40(m,2H),5.74-5.69(m,1H),5.06-4.97(m,2H),4.13-3.93(m,3H),3.84(s,6H),3.80-3.67(m,1H),3.42(brs,2H),2.75-2.35(m,2H),1.31-1.25(m,3H)。
实施例25
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-(异丙基氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000046
第一步
(S)-3-(4-氰基-3-((3,5-二甲氧苯基)乙炔基)-5-(异丙基氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将混合物(S)-3-(5-氨基-4-氰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7d(600mg,1.37mmol)、碳酸铯(893mg,2.74mmol)和乙腈(25mL)搅拌10分钟,迅速加入2-溴丙烷(186mg,1.51mmol),加热至72℃,搅拌6小时。冷却至室温,减压浓缩,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=2/1),得到目标产物(S)-3-(4-氰基-3-((3,5-二甲氧苯基)乙炔基)-5-(异丙基氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯25a(600mg,浅黄色固体),产率:91%。
MS m/z(ESI):424[M+1-56]
参照实施例24中第二步至第四步的操作步骤合成实施例25。
MS m/z(ESI):452[M+1]
1H NMR(400MHz,CDCl3)δ6.88(brs,1H),6.70(s,2H),6.54(s,1H),6.51-6.39(m,2H),6.03(t,J=10.3Hz,1H),5.74-5.69(m,1H),5.49(brs,1H),4.96-4.87(m,1H),4.09-3.86(m,3H),3.80-3.66(m,7H),3.45-3.43(m,1H),2.69-2.32(m,2H),1.27-1.15(m,6H)。
实施例26
(S)-1-(1-丙烯酰吡咯烷-3-基)-5-((环丙基甲基)氨基)-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000047
第一步
(S)-1-(1-丙烯酰吡咯烷-3-基)-5-((环丙基甲基)氨基)-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-4-甲酰胺
将化合物(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-4-甲酰胺7(50mg,0.12mmol)溶于乙腈(2mL)中,加入碳酸铯(80mg,0.24mmol)和(溴甲基)环丙烷(19mg,0.13mmol),加热至70℃,搅拌4小时。将反应液倒入水(30mL)中,并用乙酸乙酯(30mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用薄层硅胶制备色谱(二氯甲烷/甲醇=12/1)纯化,得到目标产物(S)-1-(1-丙烯酰吡咯烷-3-基)-5-((环丙基甲基)氨基)-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-4-甲酰胺26(14mg,白色固体),产率:28%。
MS m/z(ESI):464[M+1]
1H NMR(400MHz,CDCl3)δ6.89(brs,1H),6.71(s,2H),6.54(s,1H),6.51-6.37(m,2H),5.76-5.71(m,1H),5.40(brs,1H),5.03-4.95(m,1H),4.06-3.89(m,3H),3.82(s,6H),3.78-3.67(m,1H),3.06-3.02(m,2H),2.69-2.52(m,1H),2.46-2.35(m,1H),1.15-0.98(m,1H),0.63-0.60(m,2H),0.29-0.27(m,2H)。
实施例27
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((2,2,2-三氟乙基)氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000048
第一步
(S)-3-(4-氰基-3-((3,5-二甲氧苯基)乙炔基)-5-((2,2,2-三氟乙基)氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将(S)-3-(5-氨基-4-氰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7d(430mg,0.98mmol)、三氟乙醛水溶液(75%)(304mg,1.96mmol)和钛酸四乙酯(448mg,1.96mmol)加入到二氯甲烷(15mL)中,并搅拌2小时。待反应完全,向反应液中加入硼氢化钠(75mg,1.96mmol),常温下继续搅拌1小时。将反应液倒入水中并用乙酸乙酯(20mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物经快速柱纯化,得到目标产物(S)-3-(4-氰基-3-((3,5-二甲氧苯基)乙炔基)-5-((2,2,2-三氟乙基)氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯27a(120mg,黄色油状物),产率:26%。
MS m/z(ESI):464[M+1-56]
参照实施例24中第二步至第四步的操作步骤合成实施例27。
MS m/z(ESI):492[M+1]
1H NMR(400MHz,CDCl3)δ6.92(brs,1H),6.70(s,2H),6.52(s,1H),6.47-6.39(m,2H),6.31-6.25(m,1H),5.75-5.65(m,1H),5.65(brs,1H),5.05-4.98(m,1H),4.10-3.88(m,3H),3.80(s,6H),3.75-3.61 (m,3H),2.63-2.34(m,2H)。
实施例28
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((2-甲氧基乙基)氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000049
参照实施例25的操作步骤合成实施例28,但在第一步中用1-溴-2-甲氧基乙烷取代2-溴丙烷。
MS m/z(ESI):468[M+1]
1H NMR(400MHz,DMSO-d6)δ7.32(brs,1H),6.74(d,J=2.2Hz,2H),6.64(dd,J=16.8,10.4Hz,1H),6.60(t,J=2.2Hz,1H),6.50(t,J=6.0Hz,1H),6.16(dd,J=16.8,5.0Hz,1H),5.68(t,J=10.8Hz,1H),5.15-5.05(m,1H),4.05-4.01(m,0.5H),3.86-3.81(m,1.5H),3.77(s,6H),3.70-3.61(m,1H),3.59-3.50(m,1H),3.46(t,J=5.1Hz,2H),3.39-3.34(m,2H),3.26(s,3H),2.42-2.23(m,2H)。
实施例29
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((2-羟基乙基)氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000050
Figure PCTCN2017072570-appb-000051
第一步
(S)-3-(5-((2-乙酰氧基乙基)氨基)-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将混合物(S)-3-(5-氨基-4-氰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7d(300mg,0.685mmol)、2-溴乙基乙酸酯(126mg,0.753mmol)、碳酸铯(447mg,1.37mmol)和乙腈(4mL)加热至90℃,并搅拌2小时。将反应液冷却至室温,倒入水(50mL)中,并用乙酸乙酯(30mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=15/1),得到目标产物(S)-3-(5-((2-乙酰氧基乙基)氨基)-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯29a(148mg,黄色固体),产率:41%。
MS m/z(ESI):468[M+1-56]
第二步
(S)-3-(4-氨基甲酰-3-((3,5-二甲氧苯基)乙炔基)-5-((2-羟基乙基)氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将混合物(S)-3-(5-((2-乙酰氧基乙基)氨基)-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯29a(68mg,0.146mmol)、乙醇(5mL)和二甲基亚砜(1mL),加入饱和氢氧化钠溶液(3mL)和双氧水(4mL),30℃下搅拌1小时。待反应完成,将反应液倒入饱和亚硫酸钠溶液(30mL)中,并用乙酸乙酯(30mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,得到目标产物(S)-3-(4-氨基甲酰-3-((3,5-二甲氧苯基)乙炔基)-5-((2-羟基乙基)氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯29b(110mg,粗品, 黄色油状物),产率:>100%,产物不经纯化直接用于下一步反应。
MS m/z(ESI):444[M+1-56]
第三步
(S)-3-((3,5-二甲氧苯基)乙炔基)-5-((2-羟基乙基)氨基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺
将化合物(S)-3-(4-氨基甲酰-3-((3,5-二甲氧苯基)乙炔基)-5-((2-羟基乙基)氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯29b(110mg,0.146mmol,粗品)溶于盐酸的甲醇溶液(5mL),加热至40℃搅拌1小时。待反应完成后,减压浓缩,得到目标产物(S)-3-((3,5-二甲氧苯基)乙炔基)-5-((2-羟基乙基)氨基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺29c(160mg,粗品,白色固体),产率:>100%,产物不经纯化直接用于下一步反应。
MS m/z(ESI):400[M+1]
第四步
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((2-羟基乙基)氨基)-1H-吡唑-4-甲酰胺
将化合物(S)-3-((3,5-二甲氧苯基)乙炔基)-5-((2-羟基乙基)氨基)-1-(吡咯烷-3-基)-1H-吡唑-4-甲酰胺29c(160mg,0.146mmol,粗品)溶于四氢呋喃(5mL)中,加入饱和碳酸氢钠溶液(10mL),再加入丙烯酰氯(12mg,0.13mmol),室温搅拌10分钟。待反应完成后,将反应液倒入水(50mL)中,并用乙酸乙酯(30mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物通过反相高效液相制备色谱[乙腈/水(含0.2%甲酸):20%-60%]纯化,得到目标产物(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((2-羟基乙基)氨基)-1H-吡唑-4-甲酰胺29(6mg,白色固体),产率:9%。
MS m/z(ESI):454[M+1]
1H NMR(400MHz,DMSO-d6)δ7.36(brs,1H),6.76(brs,1H),6.74(s,2H),6.70-6.60(m,2H),6.55-6.52(m,1H),6.17(d,J=16.9Hz,1H),5.69(t,J=10.9Hz,1H),5.16-5.10(m,1H),4.87(s,1H),4.06-4.0(m,0.5H),3.83-3.81(m,1.5H),3.77(s,6H),3.68-3.63(m,2H),3.55-3.53(m,2H),3.28-3.26(m,2H),2.38-2.27(m,2H)。
实施例30
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((3-吗啉代丙基)氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000052
第一步
3-吗啉代丙基4-甲基苯磺酸酯
将化合物3-吗啉代丙烷-1-醇30a(500mg,3.45mmol)溶于二氯甲烷(100ml),加入4-二甲氨基吡啶(42mg,0.34mmol)、三乙胺(1.04g,10.3mmol)和对甲苯磺酰氯(988mg,5.17mmol),室温搅拌过夜。待反应完成,将反应液倒入水(50mL)中,并用二氯甲烷(50mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=2/1,得到目标产物3-吗啉代丙基4-甲基苯磺酸酯30b(660mg,黄色油状物),产率:64%。
MS m/z(ESI):300[M+1]
参照实施例25的操作步骤合成实施例30,但在第一步中用3-吗啉代丙基4-甲基苯磺酸酯取代2-溴丙烷。
MS m/z(ESI):537[M+1]
1H NMR(400MHz,CDCl3)δ8.23(brs,1H),7.12(brs,1H),6.94(brs,1H),6.69(s,2H),6.52(s,1H),6.49-6.40(m,2H),5.92(brs,1H),5.74-5.70(m,1H),5.03-4.96(m,1H),4.09-3.90(m,3H),3.80-3.68(m,11H),3.28(brs,2H),2.89(brs,6H),2.69-2.33(m,2H),1.93(brs,2H)。
实施例31
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((2-吗啉代乙基)氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000053
参照实施例25的操作步骤合成实施例31,但在第一步中用4-(2-氯乙基)吗啉取代2-溴丙烷。
MS m/z(ESI):523[M+1]
1H NMR(400MHz,CDCl3)δ8.14(s,1H),6.95(brs,1H),6.69(s,2H),6.63(brs,1H),6.52(s,1H),6.49-6.39(m,2H),6.14(brs,1H),5.75-5.70(m,1H),5.06-4.98(m,1H),4.11-3.85(m,3H),3.80-3.72(m,11H),3.37-3.33(m,2H),2.80-2.73(m,2H),2.65(brs,4H),2.45-2.32(m,2H)。
实施例32
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((2-(吡咯烷-1-基)乙基)氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000054
第一步
2-溴乙基4-甲基苯磺酸酯
将化合物2-溴乙醇32a(500mg,4.0mmol)、4-二甲氨基吡啶(246mg,2.02mmol)和三乙胺(1.22g,12.1mmol)溶于二氯甲烷(50mL),降 温至0℃,再分批加对甲苯磺酰氯(1.15g,6.05mmol),加入完成后,升至室温搅拌过夜。待反应完成,将反应液倒入水(50mL)中并用二氯甲烷(50mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用硅胶柱层析纯化(石油醚/乙酸乙酯=10/1),得到目标产物2-溴乙基4-甲基苯磺酸酯32b(600mg,黄色油状物),产率:53%。
MS m/z(ESI):277[M+1]
第二步
(S)-3-(5-((2-溴乙基)氨基)-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将混合物(S)-3-(5-氨基-4-氰基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁基酯7d(400mg,0.92mmol)、2-溴乙基4-甲基苯磺酸酯(380mg,1.37mmol)、碳酸铯(600mg,1.84mmol)和乙腈(10mL)加热至70℃,并搅拌2小时。将反应液倒入水(50mL)中,并用乙酸乙酯(50mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物通过快速柱分离,得到目标产物(S)-3-(5-((2-溴乙基)氨基)-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯32c(240mg,棕色油状物),产率:48%。
MS m/z(ESI):408[M+1-56-80]
第三步
(S)-3-(4-氰基-3-((3,5-二甲氧苯基)乙炔基)-5-((2-(吡咯烷-1-基)乙基)氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯
将混合(S)-3-(5-((2-溴乙基)氨基)-4-氰基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯32c(240mg,0.44mmol)、吡咯烷(47mg,0.66mmol)、碳酸铯(288mg,0.88mmol)和乙腈(5mL)加热至70℃,并搅拌1.5小时。将反应液倒入水(30mL)中,并用乙酸乙酯(30mL×3)萃取,有机相合并后经无水硫酸钠干燥,过滤除去干燥剂,减压浓缩,残余物用硅胶柱层析纯化(二氯甲烷/甲醇=10/1),得到目标产物(S)-3-(4-氰基-3-((3,5-二甲氧苯基)乙炔基)-5-((2-(吡咯烷-1-基)乙基)氨基)-1H-吡唑-1-基)吡咯烷-1-甲酸叔丁酯32d(200mg,黄色油状物),产率:85%。
MS m/z(ESI):479[M+1-56]
参照实施例24中第二步至第四步的操作步骤合成实施例32。
MS m/z(ESI):507[M+1]
1H NMR(400MHz,CDCl3)δ8.38(s,1H),6.99(brs,1H),6.69(s,2H),6.51(s,1H),6.47-6.36(m,2H),5.72-5.67m,2H),5.16-5.08(m,1H),4.12-3.86(m,3H),3.80-3.62(m,9H),3.33-3.29(m,6H),2.62-2.34(m,2H),2.07(brs,4H)。
实施例33
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((四氢-2H-吡喃-4-基)氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000055
第一步
4-碘四氢-2H-吡喃
将4-羟基四氢-2H-吡喃33a(2.04g,20mmol)、三苯基膦(6.81g,26)和咪唑(2.04g,30mmol)溶于二氯甲烷(100mL)中,冷却至0℃,然后加入碘(6.09g,24mmol),并在45℃下搅拌14小时。反应用水淬灭并用乙酸乙酯(50mL×2)萃取,有机相合并后用无水硫酸钠干燥,过滤后滤液减压脱溶。残余物用硅胶柱层析(石油醚/乙酸乙酯=1/1)纯化得到目标产物4-碘四氢-2H-吡喃33b(2.12g,白色固体),产率:50%。
1H NMR(400MHz,DMSO-d6)δ4.62(dt,J=13.9,4.5Hz,1H),3.68-3.64(m,2H),3.47-3.42(m,2H),2.13-1.97(m,4H)。
参照实施例24的操作步骤合成实施例33,但在第一步中用4-碘四氢-2H-吡喃取代碘乙烷。
MS m/z(ESI):494[M+H]
1H NMR(400MHz,CD3OD)δ6.74(t,J=2.2Hz,2H),6.71-6.62 (m,1H),6.60-6.58(m,1H),6.36-6.30(m,1H),5.82-5.77(m,1H),5.18-5.12(m,1H),4.04-3.94(m,6H),3.81(s,6H),3.52-3.46(m,3H),2.55-2.39(m,2H),1.94-1.92(m,2H),1.60-1.55(m,2H)。
实施例34
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((1-甲基哌啶-4-基)氨基)-1H-吡唑-4-甲酰胺
Figure PCTCN2017072570-appb-000056
第一步
4-碘-1-甲基哌啶
将4-羟基-1-甲基哌啶34a(2.3g,20mmol)、三苯基膦(6.81g,26mmol)、咪唑(2.04g,30mmol)和二氯甲烷(100mL)混合并冷却至0℃,然后加入碘(6.09g,24mmol)并继续搅拌18小时。反应结束后用水淬灭,然后用二氯甲烷(50mL×2)萃取。有机相合并后用无水硫酸钠干燥并过滤,然后将滤液减压脱溶。残余物用硅胶柱层析(二氯甲烷/甲醇=10/1)纯化得到目标产物4-碘-1-甲基哌啶34b(2.25g,白色固体),产率:50%。
MS m/z(ESI):226[M+H]
第二步
(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((1-甲基哌啶-4-基)氨基)-1H-吡唑-4-甲酰胺
将化合物(S)-1-(1-丙烯酰吡咯烷-3-基)-5-氨基-3-((3,5-二甲氧苯基)乙炔基)-1H-吡唑-4-甲酰胺7(210mg,0.5mmmol)、4-碘-1-甲基哌啶34b(450mg,2mmol)、碳酸钾(207mg,1.5mmol)和乙腈(10mL)混合并在90℃下加热并搅拌13小时。反应混合物减压脱溶后溶于水,然后用乙酸乙酯萃取(50mL×2)。有机相合并后减压脱溶,残余物用 反向制备液相色谱纯化得到目标产物(S)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-((1-甲基哌啶-4-基)氨基)-1H-吡唑-4-甲酰胺34(8.1mg,白色固体),产率:3.2%。
MS m/z(ESI):507[M+H]
1H NMR(400MHz,CD3OD)δ6.77(s,2H),6.68-6.65(m,1H),6.59(s,1H),6.34-6.30(m,1H),5.81-5.78(m,1H),5.69-5.67(m,1H),5.03-5.00(m,1H),4.98-5.95(m,1H),4.92-4.90(m,1H),4.36(s,2H),4.12-4.07(m,1H),4.00-3.98(m,1H),3.93-3.90(m,1H),3.86-3.84(m,1H),3.81(s,6H),3.72-3.68(m,1H),2.53-2.47(m,3H),2.40-2.38(m,1H),2.32(s,3H),2.27-2.21(m,1H)。
生物学实验
FGFR的活性抑制测试
使用HTRF激酶检测试剂盒,通过检测激酶反应中底物的磷酸化水平来评估本发明的化合物对FGFR体外活性的影响(表1)。
FGFR1的活性抑制测试
实验方法概述如下:
反应缓冲液包含以下组分:5倍稀释的enzymatic buffer/kinase 5X(Cisbio,货号为62EZBFDD)(主要成分为50mM HEPES,pH7.0)、5mM MgCl2和1mM DTT;人重组FGFR1催化结构域蛋白(氨基酸308-731)由公司自己纯化,用反应缓冲液稀释成0.6ng/uL的激酶溶液;底物反应溶液包括用反应缓冲液稀释成400nM的生物素标记的酪氨酸激酶底物(Cisbio,货号为62TK0PEC)和40uM ATP;检测液包括用检测缓冲液(Cisbio,货号为62SDBRDF)稀释成0.125ng/uL Eu3+标记的笼状抗体(Cisbio,货号为61T66KLB)、25nM链霉亲和素标记的XL665(Cisbio,货号为610SAXLB)。
将化合物用DMSO溶解稀释至1mM,然后用DMSO进行4倍的系列稀释至最低浓度为0.061uM,每个浓度点再使用反应缓冲液稀释40倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。
向384孔检测板(Thermo,货号为264706)中添加4uL化合物溶液和2uL的FGFR1激酶溶液,混合均匀后室温孵育15分钟;随后加入4uL底物反应溶液,将反应混合物在室温孵育60分钟;随后加入与 反应等体积的10uL检测液终止反应,混合均匀后室温放置。60分钟后,磷酸化的产物同时被Eu3+标记的笼状抗体(供体)和链霉亲和素标记的XL665抗体(受体)识别,在激光激发后,靠近的供体和受体发生能量共振转移,其从供体(620nm)转移至受体(665nm)的能量用酶标仪EnVision(Perkin Elmer)检测。665/620的比值与底物的磷酸化程度呈正相关,因此从而检测FGFR1激酶的活性。
该实验中未加酶组作为100%抑制组,加酶但是未加化合物组作为0%抑制组。化合物对FGFR1活性的抑制百分比用以下公式计算:
抑制百分比=100-100*(比值化合物-比值100%抑制)/(比值0%抑制-比值100% 抑制)
化合物的IC50值由10个浓度点用Excel中XLfit软件通过以下公式来计算:
Y=Bottom+(Top-Bottom)/(1+10^((10gIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为S型曲线的底部平台值,Top为S型曲线的顶部平台值,X为待测化合物浓度的对数值,slope factor为曲线斜率系数。
FGFR2的活性抑制测试
实验方法概述如下:
反应缓冲液包含以下组分:5倍稀释的enzymatic buffer/kinase 5X(Cisbio,货号为62EZBFDD)(主要成分为50mM HEPES,pH7.0)、5mM MgCl2和1mM DTT;人重组FGFR2催化结构域蛋白(氨基酸400-821)购自义翘神州生物技术有限公司,用反应缓冲液稀释成0.45ng/uL的激酶溶液;底物反应溶液包括用反应缓冲液稀释成800nM的生物素标记的酪氨酸激酶底物(Cisbio,货号为62TK0PEC)和50uM ATP;检测液包括用检测缓冲液(Cisbio,货号为62SDBRDF)稀释成0.125ng/uL Eu3+标记的笼状抗体(Cisbio,货号为61T66KLB)、50nM链霉亲和素标记的XL665(Cisbio,货号为610SAXLB)。
将化合物用DMSO溶解稀释至1mM,然后用DMSO进行4倍的系列稀释至最低浓度为0.061uM,每个浓度点再使用反应缓冲液稀释40倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。
向384孔检测板(Thermo,货号为264706)中添加4uL化合物溶 液和2uL的FGFR2激酶溶液,混合均匀后室温孵育15分钟;随后加入4uL底物反应溶液,将反应混合物在室温孵育60分钟;随后加入与反应等体积的10uL检测液终止反应,混合均匀后室温放置。60分钟后,磷酸化的产物同时被Eu3+标记的笼状抗体(供体)和链霉亲和素标记的XL665抗体(受体)识别。在激光激发后,靠近的供体和受体发生能量共振转移,其从供体(620nm)转移至受体(665nm)的能量用酶标仪EnVision(Perkin Elmer)检测。665/620的比值与底物的磷酸化程度呈正相关,因此从而检测FGFR2激酶的活性。
该实验中未加酶组作为100%抑制组,加酶但是未加化合物组作为0%抑制组。化合物对FGFR2活性抑制百分比用以下公式计算:
抑制百分比=100-100*(比值化合物-比值100%抑制)/(比值0%抑制-比值100% 抑制)
化合物的IC50值由10个浓度点用Excel中XLfit软件通过以下公式来计算:
Y=Bottom+(Top-Bottom)/(1+10^((logIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为S型曲线的底部平台值,Top为S型曲线的顶部平台值,X为待测化合物浓度的对数值,slope factor为曲线斜率系数。
FGFR3的活性抑制测试
实验方法概述如下:
反应缓冲液包含以下组分:5倍稀释的enzymatic buffer/kinase 5X(Cisbio,货号为62EZBFDD)(主要成分为50mM HEPES,pH7.0)、5mM MgCl2和1mM DTT;人重组FGFR3催化结构域蛋白(氨基酸399-806)购自义翘神州生物技术有限公司,用反应缓冲液稀释成0.3ng/uL的激酶溶液;底物反应溶液包括用反应缓冲液稀释成1000nM的生物素标记的酪氨酸激酶底物(Cisbio,货号为62TK0PEC)和90uM ATP;检测液包括用检测缓冲液(Cisbio,货号为62SDBRDF)稀释成0.125ng/uL Eu3+标记的笼状抗体(Cisbio,货号为61T66KLB)、62.5nM链霉亲和素标记的XL665(Cisbio,货号为610SAXLB)。
将化合物用DMSO溶解稀释至1mM,然后用DMSO进行4倍的系列稀释至最低浓度为0.061uM,每个浓度点再使用反应缓冲液稀释 40倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。
向384孔检测板(Thermo,货号为264706)中添加4uL化合物溶液和2uL的FGFR3激酶溶液,混合均匀后室温孵育15分钟;随后加入4uL底物反应溶液,将反应混合物在室温孵育60分钟;随后加入与反应等体积的10uL检测液终止反应,混合均匀后室温放置。60分钟后,磷酸化的产物同时被Eu3+标记的笼状抗体(供体)和链霉亲和素标记的XL665抗体(受体)识别。在激光激发后,靠近的供体和受体发生能量共振转移,其从供体(620nm)转移至受体(665nm)的能量用酶标仪EnVision(Perkin Elmer)检测。665/620的比值与底物的磷酸化程度呈正相关,因此从而检测FGFR3激酶的活性。
该实验中未加酶组作为100%抑制组,加酶但是未加化合物组作为0%抑制组。化合物对FGFR3活性抑制百分比用以下公式计算:
抑制百分比=100-100*(比值化合物-比值100%抑制)/(比值0%抑制-比值100% 抑制)
化合物的IC50值由10个浓度点用Excel中XLfit软件通过以下公式来计算:
Y=Bottom+(Top-Bottom)/(1+10^((logIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为S型曲线的底部平台值,Top为S型曲线的顶部平台值,X为待测化合物浓度的对数值,slope factor为曲线斜率系数。
FGFR4的活性抑制测试
实验方法概述如下:
反应缓冲液包含以下组分:5倍稀释的enzymatic buffer/kinase 5X(Cisbio,货号为62EZBFDD)(主要成分为50mM HEPES,pH7.0)、5mM MgCl2和1mM DTT;人重组FGFR4催化结构域蛋白(氨基酸460-802)购自清华大学蛋白质研究技术中心,用反应缓冲液稀释成0.5ng/uL的激酶溶液;底物反应溶液包括用反应缓冲液稀释成500nM的生物素标记的酪氨酸激酶底物(Cisbio,货号为62TK0PEC)和90uM ATP;检测液包括用检测缓冲液(Cisbio,货号为62SDBRDF)稀释成0.125ng/uL Eu3+标记的笼状抗体(Cisbio,货号为61T66KLB)、31.25nM链霉亲和素标记的XL665(Cisbio,货号为610SAXLB)。
将化合物用DMSO溶解稀释至1mM,然后用DMSO进行4倍的系列稀释至最低浓度为0.061uM,每个浓度点再使用反应缓冲液稀释40倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。
向384孔检测板(Thermo,货号为264706)中添加4uL化合物溶液和2uL的FGFR4激酶溶液,混合均匀后室温孵育15分钟;随后加入4uL底物反应溶液,将反应混合物在室温孵育60分钟;随后加入与反应等体积的10uL检测液终止反应,混合均匀后室温放置。60分钟后,磷酸化的产物同时被Eu3+标记的笼状抗体(供体)和链霉亲和素标记的XL665抗体(受体)识别。在激光激发后,靠近的供体和受体发生能量共振转移,其从供体(620nm)转移至受体(665nm)的能量用酶标仪EnVision(Perkin Elmer)检测。665/620的比值与底物的磷酸化程度呈正相关,因此从而检测FGFR4激酶的活性。
该实验中未加酶组作为100%抑制组,加酶但是未加化合物组作为0%抑制组。化合物对FGFR4活性抑制百分比用以下公式计算:
抑制百分比=100-100*(比值化合物-比值100%抑制)/(比值0%抑制-比值100% 抑制)
化合物的IC50值由10个浓度点用Excel中XLfit软件通过以下公式来计算:
Y=Bottom+(Top-Bottom)/(1+10^((logIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为S型曲线的底部平台值,Top为S型曲线的顶部平台值,X为待测化合物浓度的对数值,slope factor为曲线斜率系数。
表1
Figure PCTCN2017072570-appb-000057
A<10nM;10nM≤B<100nM;100nM≤C<1000nM
本发明的实施例化合物对FGFR的活性具有显著抑制效应,优选IC50为100至1000nM,更优选IC50小于100nM,最优选IC50小于10nM。
Hep3B细胞增殖抑制的测定
使用发光细胞活力测试实验评估本发明的化合物对Hep3B肝癌细胞系细胞增殖的影响(表2)。
实验方法概述如下:
CellTilter-Glo试剂(Promega,货号为G7572)由CTG冻干粉和CTG缓冲液组成,使用时将冻干粉溶解到缓冲液中即可。
将化合物用DMSO(Sigma,货号为D5879)溶解稀释至5mM,然后用DMSO进行4倍的系列稀释至最低浓度为0.31uM,每个浓度点再用不含FBS的DMEM培养基(ThermoFisher,货号为11995073)稀释50倍。如果化合物IC50值非常低,可以降低化合物的起始浓度。
Hep3B细胞(来自中国科学院上海生命科学研究院细胞资源中心)在含有10%FBS(GBICO,货号为10099-141)和100U/mL青链霉素混合液(ThermoFisher,货号为15140122)的DMEM完全培养基中培养,当细胞在培养容器中覆盖率达80-90%时,用0.25%胰酶(含EDTA)(ThermoFisher,货号为25200056)消化吹散后种植于白色384孔板(ThermoFisher,货号为164610),每孔1000细胞(27uL DMEM完全培养基),然后把384孔板置于37℃、5%CO2的培养箱中培养过夜(18-20小时)。
过夜后每孔加入3uL DMEM稀释后的化合物,轻轻离心混匀,然后把384孔板置于37℃、5%CO2的培养箱中继续培养,72小时后取出于室温放置。30分钟,每孔加15uL平衡至室温的CTG试剂,置于振荡器上轻轻震荡3分钟以确保细胞裂解充分,放置10分钟使冷光信号稳定,然后用EnVision(Perkin Elmer)读取冷光信号。
其中,加10uM Blueprint的BLU9931(Cancer Discovery 2015,5,424)组的冷光信号作为signal100%抑制,加0.2%DMSO组的冷光信号作为signal0%抑制
化合物对Hep3B细胞增殖抑制的百分比可以用以下公式计算:
抑制百分比=100-100*(signal化合物-signal100%抑制)/(signal0%抑制-signal100%抑制)
化合物IC50值由8个浓度点用XLfit(ID Business Solutions Ltd.,UK)软件通过以下公式计算:
Y=Bottom+(Top-Bottom)/(1+10^((l0gIC50-X)*slope factor))
其中Y为抑制百分比,Bottom为S型曲线的底部平台值,Top为S型曲线的顶部平台值,X为待测化合物浓度的对数值,slope factor为曲线斜率系数。
RT4细胞增殖抑制的测定
使用发光细胞活力测试实验评估本发明的化合物对RT4膀胱癌细胞系细胞增殖的影响(表2)。
实验方法概述参照Hep3B细胞增殖抑制的测定方法,其中RT4细胞(来自中国科学院上海生命科学研究院细胞资源中心)、阳性对照物为Taiho专利申请WO2015008844A1中的实施例1((S)-1-(3-(4-氨基-3-((3,5-二甲氧基苯基)乙炔基)-1H-吡唑并[3,4-d]嘧啶-1-基)吡咯烷-1-基)丙-2-烯-1-酮)。
SNU-16细胞增殖抑制的测定
使用发光细胞活力测试实验评估本发明的化合物对SNU-16胃癌细胞系细胞增殖的影响(表2)。
实验方法概述参照Hep3B细胞增殖抑制的测定方法,其中SNU-16细胞(ATCC,HB-8064)、阳性对照物为Novartis的BJG398。
表2
Figure PCTCN2017072570-appb-000058
说明:A<10nM;10nM≤B<100nM;100nM≤C<1000Nm
N.D.:未进行检测
本发明的实施例化合物分别对Hep3B、RT4和SNU-16的细胞增殖具有显著抑制效应,优选IC50为100至1000nM,更优选IC50小于100nM。

Claims (13)

  1. 一种通式(I)所示的化合物:
    Figure PCTCN2017072570-appb-100001
    其中:
    A为N或CR2
    环B为苯环或5-6元杂芳环,其中所述苯环和杂芳环任选被一个或多个G1所取代;
    R1独立地选自H、卤素、氰基、C1-6烷基或-NHR3
    R2独立地选自H、卤素、氰基或C1-6烷基,其中所述烷基任选被卤素、氰基、羟基或-OC1-6烷基所取代;
    R3独立地选自H、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被卤素、氰基、-OR4、-NR5R6、C1-6烷基、C3-6环烷基或3-6元杂环基所取代;
    X不存在或为C1-6亚烷基;
    Y不存在或选自C3-8亚环烷基、3-8元亚杂环基、亚芳基或亚杂芳基,其中所述亚环烷基、亚杂环基、亚芳基和亚杂芳基任选被一个或多个G2所取代;
    Z独立地选自氰基、-NR7CN、
    Figure PCTCN2017072570-appb-100002
    键a为双键或三键;
    当键a为双键时,Ra、Rb和Rc各自独立地选自H、氰基、卤素、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被一个或多个G3所取代;
    Ra和Rb或Rb和Rc任选与它们连接的碳原子共同形成一任选含有杂原子的3-6元环;
    当键a为三键时,Ra和Rc不存在,Rb独立地选自H、氰基、卤素、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被一个或多个G4所取代;
    R7独立地选自H、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所 述烷基、环烷基和杂环基任选可被一个或多个G5所取代;
    G1、G2、G3、G4和G5各自独立地选自卤素、氰基、C1-6烷基、C2-6烯基、C2-6炔基、C3-8环烷基、3-8元杂环基、C6-10芳基、5-10元杂芳基、-OR8、-OC(O)NR8R9、-C(O)OR8、-C(O)NR8R9、-C(O)R8、-NR8R9、-NR8C(O)R9、-NR8C(O)NR9R10、-S(O)mR8或-NR8S(O)mR9,其中所述烷基、烯基、炔基、环烷基、杂环基、芳基和杂芳基任选被一个或多个选自卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR11、-OC(O)NR11R12、-C(O)OR11、-C(O)NR11R12、-C(O)R11、-NR11R12、-NR11C(O)R12、-NR11C(O)NR12R13、-S(O)mR11或-NR11S(O)mR12的取代基所取代;
    R4、R5、R6、R8、R9、R10、R11、R12和R13各自独立地选自H、C1-6烷基、C3-8环烷基、3-8元单环杂环基、单环杂芳基或苯基;且
    m为1或2;
    或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式。
  2. 根据权利要求1所述的化合物,其中A为N或CH,优选为N。
  3. 根据权利要求1或2所述的化合物,其中环B为苯环。
  4. 根据权利要求1所述的化合物,其为以下通式(II)的化合物:
    Figure PCTCN2017072570-appb-100003
    其中:
    Ga、Gb、Gc和Gd各自独立地选自H、卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR8、-NR8R9或-C(O)NR8R9,其中所述烷基、环烷基和杂环基任选被一个或多个选自卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR11或-NR11R12的取代基所取代,并且Ga、Gb、Gc和Gd各自独立地优选为-OC1-2烷基或卤素;
    A、R1、R8、R9、R11、R12、X、Y、Z的定义如权利要求1中所述。
  5. 根据权利要求1所述的化合物,其为以下通式(III)的化合物:
    Figure PCTCN2017072570-appb-100004
    其中:
    Ga和Gb各自独立地选自H、卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR8、-NR8R9或-C(O)NR8R9,其中所述烷基、环烷基和杂环基任选被一个或多个选自卤素、氰基、C1-6烷基、C3-8环烷基、3-8元杂环基、-OR11或-NR11R12的取代基所取代,并且Ga和Gb各自独立地优选为-OC1-2烷基;
    A、R1、R8、R9、R11、R12、X、Y、Z的定义如权利要求1中所述。
  6. 根据前述权利要求任一项所述的化合物,其中R1独立地选自H、-NH2或-NHR3
    R3独立地选自C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被卤素、氰基、-OR4、-NR5R6、C1-6烷基、C3-6环烷基或3-6元杂环基所取代。
  7. 根据前述权利要求任一项所述的化合物,其中R1独立地选自H、-NH2或-NHC1-6烷基。
  8. 根据前述权利要求任一项所述的化合物,其中:
    X不存在或为C1-6亚烷基;
    Y不存在或选自C3-8亚环烷基或3-8元亚杂环基;
    Z独立地选自氰基、-NR7CN、
    Figure PCTCN2017072570-appb-100005
    键a为双键或三键;
    当键a为双键时,Ra、Rb和Rc各自独立地选自H、氰基、卤素、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被一个或多个独立地选自卤素、氰基、C1-6烷基、C3-6环烷基、3-6元杂环基、-OR8或-NR8R9的取代基所取代;
    当键a为三键时,Ra和Rc不存在,Rb独立地选自H、氰基、卤素、C1-6烷基、C3-6环烷基或3-6元杂环基,其中所述烷基、环烷基和杂环基任选被一个或多个独立地选自卤素、氰基、C1-6烷基、C3-6环烷基、3-6元杂环基、-OR8或-NR8R9的取代基所取代;
    R4、R8、R9各自独立地选自H或C1-6烷基。
  9. 根据权利要求1所述的化合物,所述化合物选自:
    Figure PCTCN2017072570-appb-100006
    Figure PCTCN2017072570-appb-100007
    或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式。
  10. 一种药物组合物,所述药物组合物包含根据权利要求1-9任一项所述的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式和药学上可接受的载体和赋形剂。
  11. 根据权利要求1-9任一项所述的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式或根据权利要求10所述的药物组合物在制备用于治疗和/或预防FGFR相关性疾病、优选为肿瘤(例如非小细胞肺癌、食管癌、黑色素瘤、横纹肌肉瘤、肾细胞癌、多发性骨髓瘤、乳腺癌、卵巢癌、子宫内膜癌、***、胃癌、结肠癌、膀胱癌、胰腺癌、肺癌、乳腺癌、***癌和肝癌,更优选为肝癌、胃癌、非小细胞肺癌和膀胱癌)的药物中的用途。
  12. 权利要求1-9任一项的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式或者权利要求10所述的药物组合物,其用作药物。
  13. 一种治疗FGFR相关性疾病的方法,其包括给药有其需要的患者治疗有效量的根据权利要求1-9任一项所述的化合物或其前药、稳定同位素衍生物、可药用的盐、异构体及其混合物形式。
PCT/CN2017/072570 2016-09-19 2017-01-25 炔代杂环化合物、其制备方法及其在医药学上的应用 WO2018049781A1 (zh)

Priority Applications (12)

Application Number Priority Date Filing Date Title
ES17849992T ES2925212T3 (es) 2016-09-19 2017-01-25 Compuesto heterocíclico sustituido con alquinilo, método de preparación y uso médico del mismo
CA3036594A CA3036594C (en) 2016-09-19 2017-01-25 Alkynyl-substituted heterocyclic compound, preparation method thereof and medical use thereof
EP17849992.7A EP3517535B1 (en) 2016-09-19 2017-01-25 Alkynyl-substituted heterocyclic compound, preparation method therefor and medical use thereof
CN201780057036.5A CN109843873B (zh) 2016-09-19 2017-01-25 炔代杂环化合物、其制备方法及其在医药学上的应用
AU2017327954A AU2017327954B2 (en) 2016-09-19 2017-01-25 Alkynyl-substituted heterocyclic compound, preparation method therefor and medical use thereof
MX2019003143A MX2019003143A (es) 2016-09-19 2017-01-25 Compuesto heterociclico sustituido con alquinilo, su metodo de preparacion y su uso medico.
RU2019112046A RU2729069C1 (ru) 2016-09-19 2017-01-25 Алкинил-замещенное гетероциклическое соединение, способ его получения и его применение в медицине
JP2019515464A JP6906811B2 (ja) 2016-09-19 2017-01-25 アルキル置換複素環化合物、そのための調製方法およびその医学的用途
KR1020197011454A KR20190052113A (ko) 2016-09-19 2017-01-25 알키닐-치환된 헤테로시클릭 화합물, 이의 제조 방법 및 이의 의약 용도
US16/353,806 US10710981B2 (en) 2016-09-19 2019-03-14 Alkynyl-substituted heterocyclic compound, preparation method therefor and medical use thereof
US16/925,738 US11059805B2 (en) 2016-09-19 2020-07-10 Alkynyl-substituted heterocyclic compound, preparation method therefor and medical use thereof
US17/369,850 US11572353B2 (en) 2016-09-19 2021-07-07 Alkynyl-substituted heterocyclic compound, preparation method therefor and medical use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610833890.3 2016-09-19
CN201610833890.3A CN107840842A (zh) 2016-09-19 2016-09-19 炔代杂环化合物、其制备方法及其在医药学上的应用

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/353,806 Continuation-In-Part US10710981B2 (en) 2016-09-19 2019-03-14 Alkynyl-substituted heterocyclic compound, preparation method therefor and medical use thereof

Publications (1)

Publication Number Publication Date
WO2018049781A1 true WO2018049781A1 (zh) 2018-03-22

Family

ID=61618562

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/072570 WO2018049781A1 (zh) 2016-09-19 2017-01-25 炔代杂环化合物、其制备方法及其在医药学上的应用

Country Status (12)

Country Link
US (3) US10710981B2 (zh)
EP (1) EP3517535B1 (zh)
JP (1) JP6906811B2 (zh)
KR (1) KR20190052113A (zh)
CN (2) CN107840842A (zh)
AU (1) AU2017327954B2 (zh)
CA (1) CA3036594C (zh)
ES (1) ES2925212T3 (zh)
MX (1) MX2019003143A (zh)
RU (1) RU2729069C1 (zh)
SG (1) SG10202102822RA (zh)
WO (1) WO2018049781A1 (zh)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020131674A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. 7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021129565A1 (en) * 2019-12-26 2021-07-01 Beijing Innocare Pharma Tech Co., Ltd. Crystalline forms of (s) -1- (1-acryloylpyrrolidin-3-yl) -3- ( (3, 5-dimethoxyphenyl) ethynyl) -5- (methylamino) -1h-pyrazole-4-carboxamide
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
JP2022511948A (ja) * 2018-12-13 2022-02-01 インターベット インターナショナル ベー. フェー. 1-[(3r,4s)-4-シアノテトラヒドロピラン-3-イル]-3-[(2-フルオロ-6-メトキシ-4-ピリジル)アミノ]ピラゾール-4-カルボキサミドの調製方法
US11345681B1 (en) 2020-06-05 2022-05-31 Kinnate Biopharma Inc. Inhibitors of fibroblast growth factor receptor kinases
WO2022152090A1 (en) * 2021-01-12 2022-07-21 Beijing Innocare Pharma Tech Co., Ltd. Process for preparing (s)-1-(1-acryloylpyrrolidin-3-yl)-3-((3,5-dimethoxyphenyl) ethynyl)-5-(methylamino)-1h-pyrazole-4-carboxamide
WO2023078179A1 (en) * 2021-11-03 2023-05-11 Beijing Innocare Pharma Tech Co., Ltd. Amorphous solid dispersion comprising (s) -1- (1-acryloylpyrrolidin-3-yl) -3- ( (3, 5-dimethoxyphenyl) ethynyl) -5- (methylamino) -1h-pyrazole-4-carboxamide
WO2023088105A1 (en) * 2021-11-16 2023-05-25 Beijing Innocare Pharma Tech Co., Ltd. Method for treating head and neck cancer

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3181209A1 (en) * 2020-06-05 2021-12-09 Stephen W. Kaldor Inhibitors of fibroblast growth factor receptor kinases
CN114853739B (zh) * 2021-02-03 2023-09-22 药雅科技(上海)有限公司 一种炔代吡嗪类fgfr抑制剂及其制备方法和用途
CN115433190A (zh) * 2021-06-02 2022-12-06 药雅科技(上海)有限公司 不可逆杂环化合物fgfr抑制剂的制备方法和用途
CN115043832B (zh) * 2021-03-08 2023-08-22 药雅科技(上海)有限公司 一种fgfr抑制剂炔代杂环类化合物及其制备方法和用途
WO2023107979A1 (en) * 2021-12-08 2023-06-15 Kinnate Biopharma Inc. Treatment of cancer with an fgfr kinase inhibitor
TW202332432A (zh) * 2021-12-08 2023-08-16 美商奇奈特生物製藥公司 Egfr抑制劑的固態形式
WO2023107870A1 (en) * 2021-12-08 2023-06-15 Kinnate Biopharma Inc. Inhibitors of fibroblast growth factor receptor kinases

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013014039A1 (en) * 2011-07-28 2013-01-31 Nerviano Medical Sciences S.R.L. Alkynyl substituted pyrimidinyl-pyrroles active as kinases inhibitors
WO2015008844A1 (ja) 2013-07-18 2015-01-22 大鵬薬品工業株式会社 Fgfr阻害剤耐性癌の治療薬

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2833948B1 (fr) * 2001-12-21 2004-02-06 Sod Conseils Rech Applic Nouveaux derives de benzimidazole et leur utilisation en tant que medicament
RU2340605C2 (ru) 2002-06-27 2008-12-10 Ново Нордиск А/С Арилкарбонильные производные в качестве терапевтических средств
CN1508130A (zh) 2002-12-18 2004-06-30 中国科学院大连化学物理研究所 N-苯基-n’-嘧啶基取代脲类衍生物的合成方法
US7150894B2 (en) * 2003-05-01 2006-12-19 Kraft Foods Holdings, Inc. Acid whey texture system
DK1761520T3 (da) 2004-06-23 2008-10-27 Lilly Co Eli Kinaseinhibitorer
FR2889526B1 (fr) 2005-08-04 2012-02-17 Aventis Pharma Sa 7-aza-indazoles substitues, compositions les contenant, procede de fabrication et utilisation
WO2007059341A2 (en) 2005-11-16 2007-05-24 Sgx Pharmaceuticals, Inc. Pyrazolothiazole protein kinase modulators
FR2908409B1 (fr) 2006-11-10 2009-01-09 Sanofi Aventis Sa Pyrazoles substituees,compositions les contenant,procede de fabrication et utilisation
US7737149B2 (en) * 2006-12-21 2010-06-15 Astrazeneca Ab N-[5-[2-(3,5-dimethoxyphenyl)ethyl]-2H-pyrazol-3-yl]-4-(3,5-dimethylpiperazin-1-yl)benzamide and salts thereof
UA96969C2 (en) * 2006-12-21 2011-12-26 Астразенека Аб Acylaminopyrazoles as fgfr inhibitors
ES2927660T3 (es) 2008-06-16 2022-11-10 Univ Tennessee Res Found Compuestos para el tratamiento del cáncer
FR2947546B1 (fr) 2009-07-03 2011-07-01 Sanofi Aventis Derives de pyrazoles, leur preparation et leur application en therapeutique
GB2474748B (en) * 2009-10-01 2011-10-12 Amira Pharmaceuticals Inc Polycyclic compounds as lysophosphatidic acid receptor antagonists
KR20120130777A (ko) 2010-03-01 2012-12-03 유니버시티 오브 테네시 리서치 파운데이션 암 치료용 화합물
WO2013024427A1 (en) 2011-08-16 2013-02-21 Glenmark Pharmaceuticals S.A. Novel urea derivatives as tec kinase inhibitors and uses thereof
CN103848810A (zh) * 2012-11-30 2014-06-11 北京赛林泰医药技术有限公司 鲁顿酪氨酸激酶抑制剂
PL3023100T3 (pl) * 2013-07-18 2019-07-31 Taiho Pharmaceutical Co., Ltd. Lek przeciwnowotworowy do okresowego podawania inhibitora FGFR
SG11201602183QA (en) 2013-10-25 2016-05-30 Novartis Ag Ring-fused bicyclic pyridyl derivatives as fgfr4 inhibitors
CA2976766A1 (en) 2015-03-25 2016-09-29 Novartis Ag Formylated n-heterocyclic derivatives as fgfr4 inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013014039A1 (en) * 2011-07-28 2013-01-31 Nerviano Medical Sciences S.R.L. Alkynyl substituted pyrimidinyl-pyrroles active as kinases inhibitors
WO2015008844A1 (ja) 2013-07-18 2015-01-22 大鵬薬品工業株式会社 Fgfr阻害剤耐性癌の治療薬

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CLIN. CANCER RES., vol. 18, 2012
DRUG DISC. TODAY, vol. 19, 2014, pages 51
PLOS ONE, vol. 7, 2012, pages e36713
See also references of EP3517535A4

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2022511948A (ja) * 2018-12-13 2022-02-01 インターベット インターナショナル ベー. フェー. 1-[(3r,4s)-4-シアノテトラヒドロピラン-3-イル]-3-[(2-フルオロ-6-メトキシ-4-ピリジル)アミノ]ピラゾール-4-カルボキサミドの調製方法
WO2020131674A1 (en) 2018-12-19 2020-06-25 Array Biopharma Inc. 7-((3,5-dimethoxyphenyl)amino)quinoxaline derivatives as fgfr inhibitors for treating cancer
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021129565A1 (en) * 2019-12-26 2021-07-01 Beijing Innocare Pharma Tech Co., Ltd. Crystalline forms of (s) -1- (1-acryloylpyrrolidin-3-yl) -3- ( (3, 5-dimethoxyphenyl) ethynyl) -5- (methylamino) -1h-pyrazole-4-carboxamide
CN114787149A (zh) * 2019-12-26 2022-07-22 北京诺诚健华医药科技有限公司 (s)-1-(1-丙烯酰吡咯烷-3-基)-3-((3,5-二甲氧苯基)乙炔基)-5-(甲基氨基)-1h-吡唑-4-甲酰胺的晶型
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
US11345681B1 (en) 2020-06-05 2022-05-31 Kinnate Biopharma Inc. Inhibitors of fibroblast growth factor receptor kinases
WO2022152090A1 (en) * 2021-01-12 2022-07-21 Beijing Innocare Pharma Tech Co., Ltd. Process for preparing (s)-1-(1-acryloylpyrrolidin-3-yl)-3-((3,5-dimethoxyphenyl) ethynyl)-5-(methylamino)-1h-pyrazole-4-carboxamide
WO2023078179A1 (en) * 2021-11-03 2023-05-11 Beijing Innocare Pharma Tech Co., Ltd. Amorphous solid dispersion comprising (s) -1- (1-acryloylpyrrolidin-3-yl) -3- ( (3, 5-dimethoxyphenyl) ethynyl) -5- (methylamino) -1h-pyrazole-4-carboxamide
WO2023088105A1 (en) * 2021-11-16 2023-05-25 Beijing Innocare Pharma Tech Co., Ltd. Method for treating head and neck cancer

Also Published As

Publication number Publication date
EP3517535A1 (en) 2019-07-31
MX2019003143A (es) 2019-06-17
US10710981B2 (en) 2020-07-14
KR20190052113A (ko) 2019-05-15
JP6906811B2 (ja) 2021-07-21
EP3517535A4 (en) 2020-02-26
US20190210997A1 (en) 2019-07-11
US20200339541A1 (en) 2020-10-29
RU2729069C1 (ru) 2020-08-04
AU2017327954A1 (en) 2019-05-02
EP3517535B1 (en) 2022-06-29
ES2925212T3 (es) 2022-10-14
CN109843873A (zh) 2019-06-04
CA3036594C (en) 2024-02-13
CA3036594A1 (en) 2018-03-22
CN107840842A (zh) 2018-03-27
SG10202102822RA (en) 2021-05-28
JP2019529444A (ja) 2019-10-17
US20210332027A1 (en) 2021-10-28
AU2017327954B2 (en) 2022-02-03
US11572353B2 (en) 2023-02-07
US11059805B2 (en) 2021-07-13
CN109843873B (zh) 2022-06-17

Similar Documents

Publication Publication Date Title
CN109843873B (zh) 炔代杂环化合物、其制备方法及其在医药学上的应用
CN111212834A (zh) 作为变构shp2抑制剂的吡啶、吡嗪和三嗪化合物
JP7426398B2 (ja) Ras阻害剤としての新規イソインドリノン置換インドールおよび誘導体
ES2426482T3 (es) Inhibidor de IGF-1R
KR101637337B1 (ko) 5-ht2b 수용체 길항활성을 가지는 신규 피라졸-3-카복사미드 유도체
TWI669300B (zh) 嘧啶類衍生物、其製備方法、其藥物組合物以及其在醫藥上的用途
WO2021143701A1 (zh) 嘧啶-4(3h)-酮类杂环化合物、其制备方法及其在医药学上的应用
EP2736514A1 (en) Alkynyl substituted pyrimidinyl-pyrroles active as kinases inhibitors
WO2018010514A1 (zh) 作为fgfr抑制剂的杂环化合物
WO2020156283A1 (zh) 炔基嘧啶或炔基吡啶类化合物、及其组合物与应用
WO2020207260A1 (zh) 一种cdk激酶抑制剂及其应用
JP7338072B2 (ja) Retキナーゼ阻害剤としてのピラゾール誘導体
TWI727152B (zh) 炔代雜環化合物、其製備方法及其在醫藥學上的應用
WO2022007659A1 (zh) 杂环类免疫调节剂
WO2022127869A1 (zh) 杂环类jak抑制剂
TW202300485A (zh) Plk4抑制劑及其用途
WO2023109929A1 (zh) 一种具有抗肿瘤活性的杂环化合物及其用途
WO2023207556A1 (zh) Prmt5-mta抑制剂
CN115650974A (zh) N-[5-(嘧啶-2-氨基)-2,4-二取代苯基]-顺式戊二烯酰胺衍生物及应用
WO2019218928A1 (zh) 吲哚啉-1-甲酰胺类化合物、其制备方法及其在医药学上的应用

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: 17849992

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3036594

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2019515464

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20197011454

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017849992

Country of ref document: EP

Effective date: 20190423

ENP Entry into the national phase

Ref document number: 2017327954

Country of ref document: AU

Date of ref document: 20170125

Kind code of ref document: A