WO2018184590A1 - 作为A 2A受体抑制剂的[1,2,4]***并[1,5-c]嘧啶衍生物 - Google Patents

作为A 2A受体抑制剂的[1,2,4]***并[1,5-c]嘧啶衍生物 Download PDF

Info

Publication number
WO2018184590A1
WO2018184590A1 PCT/CN2018/082119 CN2018082119W WO2018184590A1 WO 2018184590 A1 WO2018184590 A1 WO 2018184590A1 CN 2018082119 W CN2018082119 W CN 2018082119W WO 2018184590 A1 WO2018184590 A1 WO 2018184590A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
group
isomer
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/CN2018/082119
Other languages
English (en)
French (fr)
Inventor
陈新海
于衍新
陈鑫德
张丽
陈兆国
谢程
王校飞
吴灵慧
胡国平
黎健
陈曙辉
Original Assignee
南京明德新药研发股份有限公司
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
Application filed by 南京明德新药研发股份有限公司 filed Critical 南京明德新药研发股份有限公司
Priority to US16/500,901 priority Critical patent/US11117899B2/en
Priority to CN201880020259.9A priority patent/CN110446712B/zh
Priority to DK18781739.0T priority patent/DK3611174T3/da
Priority to JP2019554827A priority patent/JP7065113B2/ja
Priority to ES18781739T priority patent/ES2919474T3/es
Priority to EP18781739.0A priority patent/EP3611174B1/en
Priority to PL18781739.0T priority patent/PL3611174T3/pl
Publication of WO2018184590A1 publication Critical patent/WO2018184590A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to a compound of the formula (I), an isomer thereof or a pharmaceutically acceptable salt thereof, and to its use in the preparation of a medicament for treating a disease associated with an A 2A receptor.
  • the adenosine A 2A receptor is widely distributed in human tissues. This receptor is highly expressed in tissues such as spleen, thymus, white blood cells, platelets, GABA-type neurons and olfactory bulbs. It is also expressed in the heart, lungs, blood vessels, and other parts of the brain. Adenosine A 2A receptors are commonly co-existing with other GPCRs and become heterodimers. For example, A 2A receptors can form heterodimers with dopamine D 2 , cannabinoid CB 1 , glutamic acid mGluR5 , etc. .
  • Adenosine A 2A receptors play an important role in regulating vasodilation, supporting the formation of new blood vessels, protecting body tissues from damage caused by inflammation, and adenosine A 2A receptors also affect the indirect pathways of the basal ganglia. The degree of activity.
  • the decomposition of cellular tissues and the environment of hypoxia cause a large amount of decomposition of ATP, which leads to extracellular adenosine enrichment, and the concentration is abnormally high, which is 10-20 times of the normal value.
  • Binding of high concentrations of adenosine to the A2A receptor activates the adenosine signaling pathway.
  • This signaling pathway is a mechanism that protects the body's tissues through immunosuppression in the event of damage to the body's tissues.
  • adenosine signaling pathway leads to long-term inhibition of the innate immune response, which produces immune tolerance, which in turn leads to the loss of controlled growth of adenosine and A2A receptors in white blood cells (such as lymphocytes).
  • white blood cells such as lymphocytes.
  • the combination of T lymphocytes, natural killer cells, dendritic cells, etc. inhibits the effector functions that these white blood cells should have in the immune system.
  • Binding of adenosine to the A2A receptor increases the expression of CD39, CD73 and CTLA4 (T cell checkpoints), resulting in more Treg cells with greater immunosuppression.
  • Blocking the A 2A receptor adenosine signaling pathway can reduce the inhibition of the immune system and enhance the immune function of T cells, and is therefore considered to be a promising negative feedback mechanism to inhibit tumor growth.
  • the present invention provides a compound of the formula (I) or a pharmaceutically acceptable salt thereof,
  • R 2 is each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R: C 3-6 cycloalkyl , C 1-6 alkyl or C 1-6 heteroalkyl;
  • n is selected from 0, 1, 2 or 3;
  • n 0, 1, 2 or 3;
  • Ring A is selected from the group consisting of: 6 to 10 membered aryl, 5 to 10 membered heteroaryl, 5 to 10 membered heterocycloalkyl or 5 to 10 membered heterocycloalkenyl;
  • Ring B is selected from the group consisting of: phenyl or 5- to 6-membered heteroaryl;
  • R is selected from F, Cl, Br, I, OH, NH 2 , CN, or selected from C 1 1-3 alkyl, C 3-6 cycloalkyl optionally substituted by 1, 2 or 3 R', C 3-6 cycloalkyl-NH-, or phenyl;
  • R' is selected from the group consisting of: F, Cl, Br, I, OH, NH 2 ,
  • the number of the above heteroatoms or heteroatoms is independently selected from 1, 2, 3 or 4.
  • R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, or selected from the group consisting of 1, 2 or 3 R' substitutions: Me, Et,
  • the above R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, Me, Et,
  • R 1 is selected from H or is selected from the group consisting of: 1, 2 or 3 R: Me, Et,
  • R 1 is selected from the group consisting of: H, Me, Et, CF 3 ,
  • R 2 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R. : C 3-6 cycloalkyl, C 1-3 alkyl or C 1-3 alkoxy.
  • R 2 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 ,
  • said R 3 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R : C 1-3 alkyl or C 1-3 alkoxy.
  • R 3 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 or
  • the ring A is selected from the group consisting of phenyl, pyridyl, tetrahydropyranyl, 3,6-dihydro-2H-pyranyl, piperidinyl, 1,2,3,6- Tetrahydropyridyl, 1H-indenyl, 1H-carbazolyl, 1H-benzo[d]imidazolyl, benzo[d][1,3]dioxolyl, indoline- 2-keto, 1H-benzo[d][1,2,3]triazolyl, quinolyl or 1,2,3,4-tetrahydroquinolinyl.
  • the structural unit From:
  • the structural unit From:
  • the structural unit From:
  • the ring B is selected from the group consisting of phenyl, pyridyl, imidazolyl, pyrazolyl, furyl, thienyl, thiazolyl.
  • the structural unit From:
  • the structural unit From:
  • the structural unit From:
  • R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, or selected from the group consisting of 1, 2 or 3 R' substitutions: Me, Et, Other variables are as defined above.
  • the above R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, Me, Et, Other variables are as defined above.
  • R 1 is selected from H or is selected from the group consisting of: 1, 2 or 3 R: Me, Et, Other variables are as defined above.
  • R 1 is selected from the group consisting of: H, Me, Et, CF 3 , Other variables are as defined above.
  • R 2 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R. : C 3-6 cycloalkyl, C 1-3 alkyl or C 1-3 alkoxy, the other variables are as defined above.
  • R 2 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 , Other variables are as defined above.
  • said R 3 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R : C 1-3 alkyl or C 1-3 alkoxy, other variables are as defined above.
  • R 3 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 or Other variables are as defined above.
  • the ring A is selected from the group consisting of phenyl, pyridyl, tetrahydropyranyl, 3,6-dihydro-2H-pyranyl, piperidinyl, 1,2,3,6- Tetrahydropyridyl, 1H-indenyl, 1H-carbazolyl, 1H-benzo[d]imidazolyl, benzo[d][1,3]dioxolyl, indoline- 2-keto, 1H-benzo[d][1,2,3]triazolyl, quinolyl or 1,2,3,4-tetrahydroquinolinyl, other variables are as defined above.
  • the ring B is selected from the group consisting of phenyl, pyridyl, imidazolyl, pyrazolyl, furyl, thienyl, thiazolyl, and other variables are as defined above.
  • the structural unit From: or Other variables are as defined above.
  • the present invention provides a compound of the formula (I) or a pharmaceutically acceptable salt thereof,
  • R 2 is each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R: C 3-6 cycloalkyl , C 1-6 alkyl or C 1-6 heteroalkyl;
  • R 3 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from: C 1-6 alkyl optionally substituted by 1, 2 or 3 R C 1-6 heteroalkyl;
  • n is selected from 0, 1, 2 or 3;
  • n 0, 1, 2 or 3;
  • Ring A is selected from the group consisting of: 6 to 10 membered aryl, 5 to 10 membered heteroaryl, 5 to 10 membered heterocycloalkyl or 5 to 10 membered heterocycloalkenyl;
  • Ring B is selected from the group consisting of: phenyl or 5- to 6-membered heteroaryl;
  • R is selected from F, Cl, Br, I, OH, NH 2 , CN, or selected from C 1 1-3 alkyl, C 3-6 cycloalkyl optionally substituted by 1, 2 or 3 R', C 3-6 cycloalkyl-NH-, or phenyl;
  • R' is selected from the group consisting of: F, Cl, Br, I, OH, NH 2 ,
  • the number of the above heteroatoms or heteroatoms is independently selected from 1, 2, 3 or 4.
  • R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, or selected from the group consisting of 1, 2 or 3 R' substitutions: Me, Et,
  • the above R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, Me, Et,
  • R 1 is selected from H or is selected from the group consisting of: 1, 2 or 3 R: Me, Et,
  • R 1 is selected from the group consisting of: H, Me, Et, CF 3 ,
  • R 2 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R. : C 3-6 cycloalkyl, C 1-3 alkyl or C 1-3 alkoxy.
  • R 2 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 ,
  • said R 3 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R : C 1-3 alkyl or C 1-3 alkoxy.
  • R 3 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 or
  • the ring A is selected from the group consisting of phenyl, pyridyl, tetrahydropyranyl, 3,6-dihydro-2H-pyranyl, piperidinyl, 1,2,3,6- Tetrahydropyridyl, 1H-indenyl, 1H-carbazolyl, 1H-benzo[d]imidazolyl, benzo[d][1,3]dioxolyl, indoline- 2-keto, 1H-benzo[d][1,2,3]triazolyl, quinolyl or 1,2,3,4-tetrahydroquinolinyl.
  • the structural unit From:
  • the structural unit From:
  • the structural unit From:
  • the ring B is selected from the group consisting of phenyl, pyridyl, imidazolyl, pyrazolyl, furyl, thienyl, thiazolyl.
  • the structural unit From:
  • the structural unit From:
  • the structural unit From:
  • R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, or selected from the group consisting of 1, 2 or 3 R' substitutions: Me, Et, Other variables are as defined above.
  • the above R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, Me, Et, Other variables are as defined above.
  • R 1 is selected from H or is selected from the group consisting of: 1, 2 or 3 R: Me, Et, Other variables are as defined above.
  • R 1 is selected from the group consisting of: H, Me, Et, CF 3 , Other variables are as defined above.
  • R 2 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R. : C 3-6 cycloalkyl, C 1-3 alkyl or C 1-3 alkoxy, the other variables are as defined above.
  • R 2 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 , Other variables are as defined above.
  • said R 3 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R : C 1-3 alkyl or C 1-3 alkoxy, other variables are as defined above.
  • R 3 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 or Other variables are as defined above.
  • the ring A is selected from the group consisting of phenyl, pyridyl, tetrahydropyranyl, 3,6-dihydro-2H-pyranyl, piperidinyl, 1,2,3,6- Tetrahydropyridyl, 1H-indenyl, 1H-carbazolyl, 1H-benzo[d]imidazolyl, benzo[d][1,3]dioxolyl, indoline- 2-keto, 1H-benzo[d][1,2,3]triazolyl, quinolyl or 1,2,3,4-tetrahydroquinolinyl, other variables are as defined above.
  • the ring B is selected from the group consisting of phenyl, pyridyl, imidazolyl, pyrazolyl, furyl, thienyl, thiazolyl, and other variables are as defined above.
  • the present invention provides a compound of the formula (I) or a pharmaceutically acceptable salt thereof,
  • R 2 is each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R: C 3-6 cycloalkyl , C 1-6 alkyl or C 1-6 heteroalkyl;
  • R 3 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from: C 1-6 alkyl optionally substituted by 1, 2 or 3 R C 1-6 heteroalkyl;
  • n is selected from 0, 1, 2 or 3;
  • n 0, 1, 2 or 3;
  • Ring A is selected from the group consisting of: 6 to 10 membered aryl, 5 to 10 membered heteroaryl, 5 to 10 membered heterocycloalkyl or 5 to 10 membered heterocycloalkenyl;
  • Ring B is selected from the group consisting of: phenyl or 5- to 6-membered heteroaryl;
  • R is selected from F, Cl, Br, I, OH, NH 2 , CN, or selected from, optionally substituted by 1, 2 or 3 R': C 1-3 alkyl, C 1-3 alkylamino, C 1-3 alkoxy group, C 3-6 cycloalkyl group, C 3-6 cycloalkyl-NH-, 3- to 6-membered heterocycloalkyl group, 3- to 6-membered heterocycloalkyl-O-, or phenyl group ;
  • R' is selected from the group consisting of: F, Cl, Br, I, OH, NH 2 ,
  • the number of the above heteroatoms or heteroatoms is independently selected from 1, 2, 3 or 4.
  • R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, or selected from the group consisting of 1, 2 or 3 R' substitutions: Me, Et, Other variables are as defined by the present invention.
  • the above R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, Me, Et, Other variables are as defined by the present invention.
  • R 1 is selected from the group consisting of H, CN, Or selected from those optionally substituted by 1, 2 or 3 R: Me, Et, Other variables are as defined by the present invention.
  • R 1 is selected from the group consisting of: H, CN, Me, Et, CF 3 , Other variables are as defined by the present invention.
  • R 2 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R. : C 3-6 cycloalkyl, C 1-3 alkyl or C 1-3 alkoxy, other variables are as defined in the invention.
  • R 2 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 , Other variables are as defined by the present invention.
  • said R 3 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R : C 1-3 alkyl or C 1-3 alkoxy, other variables are as defined in the present invention.
  • R 3 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 or Other variables are as defined by the present invention.
  • the ring A is selected from the group consisting of phenyl, pyridyl, tetrahydropyranyl, 3,6-dihydro-2H-pyranyl, piperidinyl, 1,2,3,6- Tetrahydropyridyl, 1H-indenyl, 1H-carbazolyl, 1H-benzo[d]imidazolyl, benzo[d][1,3]dioxolyl, indoline- 2-keto, 1H-benzo[d][1,2,3]triazolyl, quinolyl or 1,2,3,4-tetrahydroquinolinyl, other variables are as defined herein.
  • Ring B is selected from the group consisting of phenyl, pyridyl, imidazolyl, pyrazolyl, furyl, thienyl, thiazolyl, and other variables are as defined herein.
  • the above compound, or a pharmaceutically acceptable salt thereof is selected from the group consisting of
  • R 1 , R 2 , R 3 are as defined in the present invention.
  • the present invention provides a compound of the formula (I), an isomer thereof or a pharmaceutically acceptable salt thereof,
  • R 2 is each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R: C 3-6 cycloalkyl , C 1-6 alkyl or C 1-6 heteroalkyl;
  • R 3 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from: C 1-6 alkyl optionally substituted by 1, 2 or 3 R C 1-6 heteroalkyl;
  • n is selected from 0, 1, 2 or 3;
  • n 0, 1, 2 or 3;
  • Ring A is selected from the group consisting of: 6 to 10 membered aryl, 5 to 10 membered heteroaryl, 5 to 10 membered heterocycloalkyl or 5 to 10 membered heterocycloalkenyl;
  • Ring B is selected from the group consisting of: phenyl or 5- to 6-membered heteroaryl;
  • R is selected from F, Cl, Br, I, OH, NH 2 , CN, or selected from, optionally substituted by 1, 2 or 3 R': C 1-3 alkyl, C 1-3 alkylamino, C 1-3 alkoxy group, C 3-6 cycloalkyl group, C 3-6 cycloalkyl-NH-, 3- to 6-membered heterocycloalkyl group, 3- to 6-membered heterocycloalkyl-O-, or phenyl group ;
  • R' is selected from the group consisting of: F, Cl, Br, I, OH, NH 2 ,
  • the number of the above heteroatoms or heteroatoms is independently selected from 1, 2, 3 or 4.
  • R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, or selected from the group consisting of 1, 2 or 3 R' substitutions: Me, Et, Other variables are as defined by the present invention.
  • the above R is selected from the group consisting of F, Cl, Br, I, OH, NH 2 , CN, Me, Et, Other variables are as defined by the present invention.
  • R 1 is selected from the group consisting of H, CN, COOH, Or selected from those optionally substituted by 1, 2 or 3 R: Me, Et, Other variables are as defined by the present invention.
  • R 1 is selected from the group consisting of: H, CN, COOH, Me, Et, CF 3 , Other variables are as defined by the present invention.
  • R 2 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R. : C 3-6 cycloalkyl, C 1-3 alkyl or C 1-3 alkoxy, other variables are as defined in the invention.
  • R 2 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 , Other variables are as defined by the present invention.
  • said R 3 are each independently selected from H, F, Cl, Br, I, OH, NH 2 , CN, or independently selected from, optionally substituted by 1, 2 or 3 R : C 1-3 alkyl or C 1-3 alkoxy, other variables are as defined in the present invention.
  • R 3 are independently selected from the group consisting of: H, F, Cl, Br, I, OH, NH 2 , CN, Me, Et, CF 3 or Other variables are as defined by the present invention.
  • the ring A is selected from the group consisting of phenyl, pyridyl, tetrahydropyranyl, 3,6-dihydro-2H-pyranyl, piperidinyl, 1,2,3,6- Tetrahydropyridyl, 1H-indenyl, 1H-carbazolyl, 1H-benzo[d]imidazolyl, benzo[d][1,3]dioxolyl, indoline- 2-keto, 1H-benzo[d][1,2,3]triazolyl, quinolyl or 1,2,3,4-tetrahydroquinolinyl, other variables are as defined herein.
  • Ring B is selected from the group consisting of phenyl, pyridyl, imidazolyl, pyrazolyl, furyl, thienyl, thiazolyl, and other variables are as defined herein.
  • the above compound, an isomer thereof, or a pharmaceutically acceptable salt thereof is selected from the group consisting of:
  • R 1 , R 2 , R 3 are as defined in the present invention.
  • the present invention provides a compound represented by the following formula, an isomer thereof or a pharmaceutically acceptable salt thereof, which is selected from the group consisting of:
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the above compound, an isomer thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the present invention also provides the use of the above compound, an isomer thereof or a pharmaceutically acceptable salt thereof for the preparation of a medicament for treating a disease associated with an A 2A receptor.
  • the present invention also provides the use of the above pharmaceutical composition for the preparation of a medicament for treating a disease associated with an A 2A receptor.
  • the present invention synthesizes a compound of formula (I) to obtain a novel class of adenosine A2a antagonists, which are used alone or in combination with antibodies for tumor immunotherapy.
  • the present invention increases the solubility of the compound while significantly improving the pharmacokinetic profile.
  • pharmaceutically acceptable salt refers to a salt of a compound of the invention prepared from a compound having a particular substituent found in the present invention and a relatively non-toxic acid or base.
  • a base addition salt can be obtained by contacting a neutral amount of such a compound with a sufficient amount of a base in a neat solution or a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salts or similar salts.
  • an acid addition salt can be obtained by contacting a neutral form of such a compound with a sufficient amount of an acid in a neat solution or a suitable inert solvent.
  • pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, hydrogencarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, Hydrogen sulfate, hydroiodic acid, phosphorous acid, etc.; and an organic acid salt, such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, Similar acids such as fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, and me
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing an acid group or a base by conventional chemical methods.
  • such salts are prepared by reacting these compounds in water or an organic solvent or a mixture of the two via a free acid or base form with a stoichiometric amount of the appropriate base or acid.
  • the compounds provided herein also exist in the form of prodrugs.
  • Prodrugs of the compounds described herein are readily chemically altered under physiological conditions to convert to the compounds of the invention.
  • prodrugs can be converted to the compounds of the invention by chemical or biochemical methods in an in vivo setting.
  • Certain compounds of the invention may exist in unsolvated or solvated forms, including hydrated forms.
  • the solvated forms are equivalent to the unsolvated forms and are included within the scope of the invention.
  • the compounds of the invention may exist in specific geometric or stereoisomeric forms.
  • the present invention contemplates all such compounds, including the cis and trans isomers, the (-)- and (+)-p-enantiomers, the (R)- and (S)-enantiomers, and the diastereomeric a conformation, a (D)-isomer, a (L)-isomer, and a racemic mixture thereof, and other mixtures, such as enantiomerically or diastereomeric enriched mixtures, all of which belong to It is within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in the substituents such as alkyl groups. All such isomers, as well as mixtures thereof, are included within the scope of the invention.
  • enantiomer or “optical isomer” refer to stereoisomers that are mirror images of one another.
  • cis-trans isomer or “geometric isomer” is caused by the inability to freely rotate a single bond due to a double bond or a ring-forming carbon atom.
  • diastereomer refers to a stereoisomer in which the molecule has two or more chiral centers and the molecules are in a non-mirrored relationship.
  • wedge-shaped dashed keys Represents the absolute configuration of a solid center with straight solid keys
  • straight dashed keys Indicates the relative configuration of the stereocenter, using wavy lines Indicates a wedge solid key Or wedge-shaped dotted key Or with wavy lines Represents a straight solid key And straight dashed keys
  • tautomer or “tautomeric form” mean that the different functional isomers are in dynamic equilibrium at room temperature and can be rapidly converted into each other. If tautomers are possible (as in solution), the chemical equilibrium of the tautomers can be achieved.
  • proton tautomers also known as prototropic tautomers
  • prototropic tautomers include interconversions by proton transfer, such as keto-enol isomerization and imine-enes. Amine isomerization.
  • the valence tautomer includes the mutual transformation of some of the bonding electrons.
  • keto-enol tautomerization is the interconversion between two tautomers of pentane-2,4-dione and 4-hydroxypent-3-en-2-one.
  • the terms "enriched in one isomer”, “isomer enriched”, “enriched in one enantiomer” or “enantiomeric enriched” refer to one of the isomers or pairs
  • the content of the oligo is less than 100%, and the content of the isomer or enantiomer is 60% or more, or 70% or more, or 80% or more, or 90% or more, or 95% or more, or 96% or more, or 97% or more, 98% or more, 99% or more, 99.5% or more, 99.6% or more, 99.7% or more, 99.8% or more, or greater than or equal to 99.9%.
  • the term “isomer excess” or “enantiomeric excess” refers to the difference between the two isomers or the relative percentages of the two enantiomers. For example, if one of the isomers or enantiomers is present in an amount of 90% and the other isomer or enantiomer is present in an amount of 10%, the isomer or enantiomeric excess (ee value) is 80%. .
  • optically active (R)- and (S)-isomers as well as the D and L isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If an enantiomer of a compound of the invention is desired, it can be prepared by asymmetric synthesis or by derivatization with a chiral auxiliary wherein the resulting mixture of diastereomers is separated and the auxiliary group cleaved to provide pure The desired enantiomer.
  • a diastereomeric salt is formed with a suitable optically active acid or base, followed by conventional methods well known in the art.
  • the diastereomers are resolved and the pure enantiomer is recovered.
  • the separation of enantiomers and diastereomers is generally accomplished by the use of chromatography using a chiral stationary phase, optionally in combination with chemical derivatization (eg, formation of an amino group from an amine). Formate).
  • the compounds of the present invention may contain unnatural proportions of atomic isotopes on one or more of the atoms that make up the compound.
  • radiolabeled compounds can be used, such as tritium (3 H), iodine -125 (125 I) or C-14 (14 C).
  • hydrogen can be replaced by heavy hydrogen to form a deuterated drug.
  • the bond composed of barium and carbon is stronger than the bond composed of common hydrogen and carbon.
  • deuterated drugs have reduced side effects and increased drug stability. Enhance the efficacy and prolong the biological half-life of the drug. Alterations of all isotopic compositions of the compounds of the invention, whether radioactive or not, are included within the scope of the invention.
  • pharmaceutically acceptable carrier refers to any formulation or carrier medium that is capable of delivering an effective amount of an active substance of the present invention, does not interfere with the biological activity of the active substance, and has no toxic side effects to the host or patient, including water, oil, Vegetables and minerals, cream bases, lotion bases, ointment bases, etc. These bases include suspending agents, tackifiers, transdermal enhancers and the like. Their formulations are well known to those skilled in the cosmetic or topical pharmaceutical arts.
  • excipient generally refers to the carrier, diluent and/or vehicle required to formulate an effective pharmaceutical composition.
  • an "effective amount” or “therapeutically effective amount” with respect to a pharmaceutical or pharmacologically active agent refers to a sufficient amount of a drug or agent that is non-toxic but that achieves the desired effect.
  • an "effective amount” of an active substance in a composition refers to the amount required to achieve the desired effect when used in combination with another active substance in the composition. The determination of the effective amount will vary from person to person, depending on the age and general condition of the recipient, and also on the particular active substance, and a suitable effective amount in a case can be determined by one skilled in the art based on routine experimentation.
  • active ingredient refers to a chemical entity that is effective in treating a target disorder, disease or condition.
  • substituted means that any one or more hydrogen atoms on a particular atom are replaced by a substituent, and may include variants of heavy hydrogen and hydrogen, as long as the valence of the particular atom is normal and the substituted compound is stable. of.
  • oxygen i.e., 0
  • substituent two hydrogen atoms are substituted.
  • Oxygen substitution does not occur on the aromatic group.
  • optionally substituted means that it may or may not be substituted, and unless otherwise specified, the kind and number of substituents may be arbitrary on the basis of chemically achievable.
  • any variable eg, R
  • its definition in each case is independent.
  • the group may optionally be substituted with at most two R, and each case has an independent option.
  • combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
  • linking group When the number of one linking group is 0, such as -(CRR) 0 -, it indicates that the linking group is a single bond.
  • one of the variables When one of the variables is selected from a single bond, it means that the two groups to which it is attached are directly linked. For example, when L represents a single bond in A-L-Z, the structure is actually A-Z.
  • a substituent When a substituent is vacant, it means that the substituent is absent. For example, when X is vacant in AX, the structure is actually A.
  • the substituent can be attached to more than one atom on a ring, the substituent can be bonded to any atom on the ring, for example, a structural unit. or It is indicated that the substituent R can be substituted at any position on the cyclohexyl group or cyclohexadiene.
  • substituents When the listed substituents are not indicated by which atom is attached to the substituted group, such a substituent may be bonded through any atom thereof, for example, a pyridyl group as a substituent may be passed through any one of the pyridine rings.
  • a carbon atom is attached to the substituted group.
  • the listed linking group does not indicate its direction of attachment, its connection direction is arbitrary, for example,
  • the medium linking group L is -MW-, and at this time, -MW- can be connected in the same direction as the reading order from left to right to form ring A and ring B. It is also possible to connect the ring A and the ring B in a direction opposite to the reading order from left to right.
  • Combinations of the linking groups, substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
  • hetero denotes a hetero atom or a hetero atomic group (ie, a radical containing a hetero atom), including atoms other than carbon (C) and hydrogen (H), and radicals containing such heteroatoms, including, for example, oxygen (O).
  • ring means substituted or unsubstituted cycloalkyl, heterocycloalkyl, cycloalkenyl, heterocycloalkenyl, cycloalkynyl, heterocycloalkynyl, aryl or heteroaryl. So-called rings include single rings, interlocking rings, spiral rings, parallel rings or bridge rings. The number of atoms on the ring is usually defined as the number of elements of the ring. For example, "5 to 7-membered ring” means 5 to 7 atoms arranged in a circle. Unless otherwise specified, the ring optionally contains from 1 to 3 heteroatoms.
  • the "5-7 membered ring” includes, for example, phenyl, pyridine and piperidinyl; on the other hand, the term “5-7 membered heterocycloalkyl ring” includes pyridyl and piperidinyl, but does not include phenyl.
  • the term “ring” also includes ring systems containing at least one ring, each of which "ring” independently conforms to the above definition.
  • heterocycle or “heterocyclyl” means a stable monocyclic, bicyclic or tricyclic ring containing a hetero atom or a heteroatom group which may be saturated, partially unsaturated or unsaturated ( Aromatic) which comprise a carbon atom and 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S, wherein any of the above heterocycles may be fused to a phenyl ring to form a bicyclic ring.
  • the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O)p, p is 1 or 2).
  • the nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents as already defined herein).
  • the heterocyclic ring can be attached to the side groups of any hetero atom or carbon atom to form a stable structure. If the resulting compound is stable, the heterocycles described herein can undergo substitutions at the carbon or nitrogen sites.
  • the nitrogen atom in the heterocycle is optionally quaternized.
  • a preferred embodiment is that when the total number of S and O atoms in the heterocycle exceeds 1, these heteroatoms are not adjacent to each other. Another preferred embodiment is that the total number of S and O atoms in the heterocycle does not exceed one.
  • aromatic heterocyclic group or "heteroaryl” as used herein means a stable 5, 6, or 7 membered monocyclic or bicyclic or aromatic ring of a 7, 8, 9 or 10 membered bicyclic heterocyclic group, It contains carbon atoms and 1, 2, 3 or 4 ring heteroatoms independently selected from N, O and S.
  • the nitrogen atom can be substituted or unsubstituted (i.e., N or NR, wherein R is H or other substituents as already defined herein).
  • the nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., NO and S(O)p, p is 1 or 2).
  • bridged rings are also included in the definition of heterocycles.
  • a bridged ring is formed when one or more atoms (ie, C, O, N, or S) join two non-adjacent carbon or nitrogen atoms.
  • Preferred bridged rings include, but are not limited to, one carbon atom, two carbon atoms, one nitrogen atom, two nitrogen atoms, and one carbon-nitrogen group. It is worth noting that a bridge always converts a single ring into a three ring. In the bridged ring, a substituent on the ring can also be present on the bridge.
  • heterocyclic compounds include, but are not limited to, acridinyl, octanoyl, benzimidazolyl, benzofuranyl, benzofuranylfuranyl, benzindenylphenyl, benzoxazolyl, benzimidin Oxazolinyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, oxazolyl, 4aH-carbazolyl, Porphyrin, chroman, chromene, porphyrin-decahydroquinolinyl, 2H, 6H-1,5,2-dithiazinyl, dihydrofuro[2,3-b] Tetrahydrofuranyl, furyl, furfuryl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-carbazolyl, nonenyl,
  • hydrocarbyl or its subordinate concept (such as alkyl, alkenyl, alkynyl, aryl, etc.), by itself or as part of another substituent, is meant to be straight-chain, branched or cyclic.
  • the hydrocarbon atom group or a combination thereof may be fully saturated (such as an alkyl group), a unit or a polyunsaturated (such as an alkenyl group, an alkynyl group, an aryl group), may be monosubstituted or polysubstituted, and may be monovalent (such as Methyl), divalent (such as methylene) or polyvalent (such as methine), may include divalent or polyvalent radicals with a specified number of carbon atoms (eg, C 1 -C 12 represents 1 to 12 carbons) , C 1-12 is selected from C 1 , C 2 , C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , C 10 , C 11 and C 12 ; C 3-12 is selected from C 3 , C 4 , C 5 , C 6 , C 7 , C 8 , C 9 , C 10 , C 11 and C 12 .).
  • C 1-12 is selected from C 1
  • Hydrocarbyl includes, but is not limited to, aliphatic hydrocarbyl groups including chain and cyclic, including but not limited to alkyl, alkenyl, alkynyl groups including, but not limited to, 6-12 members.
  • An aromatic hydrocarbon group such as benzene, naphthalene or the like.
  • hydrocarbyl means a straight or branched chain radical or a combination thereof, which may be fully saturated, unitary or polyunsaturated, and may include divalent and multivalent radicals.
  • saturated hydrocarbon radicals include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, isobutyl, cyclohexyl, (cyclohexyl).
  • a homolog or isomer of a methyl group, a cyclopropylmethyl group, and an atomic group such as n-pentyl, n-hexyl, n-heptyl, n-octyl.
  • the unsaturated hydrocarbon group has one or more double or triple bonds, and examples thereof include, but are not limited to, a vinyl group, a 2-propenyl group, a butenyl group, a crotyl group, a 2-isopentenyl group, and a 2-(butadienyl group). , 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and higher homologs and isomers body.
  • heterohydrocarbyl or its subordinate concept (such as heteroalkyl, heteroalkenyl, heteroalkynyl, heteroaryl, etc.), by itself or in combination with another term, means a stable straight chain, branched chain. Or a cyclic hydrocarbon radical or a combination thereof having a number of carbon atoms and at least one heteroatom.
  • heteroalkyl by itself or in conjunction with another term refers to a stable straight chain, branched hydrocarbon radical or combination thereof, having a number of carbon atoms and at least one heteroatom.
  • the heteroatoms are selected from the group consisting of B, O, N, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen heteroatoms are optionally quaternized.
  • the hetero atom or heteroatom group may be located at any internal position of the heterohydrocarbyl group, including where the hydrocarbyl group is attached to the rest of the molecule, but the terms "alkoxy”, “alkylamino” and “alkylthio” (or thioalkoxy). By customary expression, those alkyl groups which are attached to the remainder of the molecule through an oxygen atom, an amino group or a sulfur atom, respectively.
  • Up to two heteroatoms may be consecutive, for example, -CH 2 -NH-OCH 3.
  • cycloalkyl refers to any heterocyclic alkynyl group, etc., by itself or in combination with other terms, denotes a cyclized “hydrocarbyl group” or “heterohydrocarbyl group”, respectively.
  • a hetero atom may occupy a position at which the hetero ring is attached to the rest of the molecule.
  • cycloalkyl groups include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
  • heterocyclic groups include 1-(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothiophen-2-yl, tetrahydrothiophen-3-yl, 1-piperazinyl and 2-piperazinyl.
  • alkyl is used to denote a straight or branched saturated hydrocarbon group, which may be monosubstituted (eg, -CH 2 F) or polysubstituted (eg, -CF 3 ), and may be monovalent (eg, Methyl), divalent (such as methylene) or polyvalent (such as methine).
  • alkyl group include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, s-butyl). , t-butyl), pentyl (eg, n-pentyl, isopentyl, neopentyl) and the like.
  • alkenyl refers to an alkyl group having one or more carbon-carbon double bonds at any position of the chain, which may be mono- or poly-substituted, and may be monovalent, divalent or multivalent.
  • alkenyl group include a vinyl group, a propenyl group, a butenyl group, a pentenyl group, a hexenyl group, a butadienyl group, a pentadienyl group, a hexadienyl group and the like.
  • alkynyl refers to an alkyl group having one or more carbon-carbon triple bonds at any position of the chain, which may be mono- or poly-substituted, and may be monovalent, divalent or multivalent.
  • alkynyl groups include ethynyl, propynyl, butynyl, pentynyl and the like.
  • a cycloalkyl group includes any stable cyclic or polycyclic hydrocarbon group, any carbon atom which is saturated, may be monosubstituted or polysubstituted, and may be monovalent, divalent or multivalent.
  • Examples of such cycloalkyl groups include, but are not limited to, cyclopropyl, norbornyl, [2.2.2]bicyclooctane, [4.4.0]bicyclononane, and the like.
  • a cycloalkenyl group includes any stable cyclic or polycyclic hydrocarbon group which contains one or more unsaturated carbon-carbon double bonds at any position of the ring, and may be monosubstituted or polysubstituted, It can be one price, two price or multiple price.
  • Examples of such cycloalkenyl groups include, but are not limited to, cyclopentenyl, cyclohexenyl, and the like.
  • a cycloalkynyl group includes any stable cyclic or polycyclic hydrocarbon group which contains one or more carbon-carbon triple bonds at any position of the ring, which may be monosubstituted or polysubstituted, and may be one Price, price or price.
  • halo or “halogen”, by itself or as part of another substituent, denotes a fluorine, chlorine, bromine or iodine atom.
  • haloalkyl is intended to include both monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl is intended to include, but is not limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like. Wait.
  • examples of haloalkyl include, but are not limited to, trifluoromethyl, trichloromethyl, pentafluoroethyl, and pentachloroethyl.
  • alkoxy represents attached through an oxygen bridge
  • C 1-6 alkoxy groups include C 1, C 2, C 3 , C 4, C 5 , and C 6 alkoxy groups.
  • alkoxy groups include, but are not limited to, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentyloxy and S- Pentyloxy.
  • aryl denotes a polyunsaturated, aromatic hydrocarbon substituent which may be monosubstituted or polysubstituted, which may be monovalent, divalent or polyvalent, which may be monocyclic or polycyclic ( For example, 1 to 3 rings; at least one of which is aromatic), they are fused together or covalently linked.
  • heteroaryl refers to an aryl (or ring) containing one to four heteroatoms. In an illustrative example, the heteroatoms are selected from the group consisting of B, N, O, and S, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom is optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • aryl or heteroaryl groups include phenyl, naphthyl, biphenyl, pyrrolyl, pyrazolyl, imidazolyl, pyrazinyl, oxazolyl, phenyl-oxazolyl, isomerism Azyl, thiazolyl, furyl, thienyl, pyridyl, pyrimidinyl, benzothiazolyl, indolyl, benzimidazolyl, indolyl, isoquinolyl, quinoxalinyl, quinolinyl, 1 -naphthyl, 2-naphthyl, 4-biphenylyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl
  • aryl groups when used in conjunction with other terms (e.g., aryloxy, arylthio, aralkyl), include aryl and heteroaryl rings as defined above.
  • aralkyl is intended to include those radicals to which an aryl group is attached to an alkyl group (eg, benzyl, phenethyl, pyridylmethyl, and the like), including wherein the carbon atom (eg, methylene) has been, for example, oxygen.
  • alkyl groups substituted by an atom such as phenoxymethyl, 2-pyridyloxymethyl 3-(1-naphthyloxy)propyl and the like.
  • leaving group refers to a functional group or atom which may be substituted by another functional group or atom by a substitution reaction (for example, an affinity substitution reaction).
  • substituent groups include triflate; chlorine, bromine, iodine; sulfonate groups such as mesylate, tosylate, p-bromobenzenesulfonate, p-toluenesulfonic acid Esters and the like; acyloxy groups such as acetoxy, trifluoroacetoxy and the like.
  • protecting group includes, but is not limited to, "amino protecting group", “hydroxy protecting group” or “thiol protecting group”.
  • amino protecting group refers to a protecting group suitable for preventing side reactions at the amino nitrogen position.
  • Representative amino protecting groups include, but are not limited to, formyl; acyl, such as alkanoyl (e.g., acetyl, trichloroacetyl or trifluoroacetyl); alkoxycarbonyl, e.g., tert-butoxycarbonyl (Boc) Arylmethoxycarbonyl, such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc); arylmethyl, such as benzyl (Bn), trityl (Tr), 1, 1-di -(4'-methoxyphenyl)methyl; silyl groups such as trimethylsilyl (TMS) and tert-
  • hydroxy protecting group refers to a protecting group suitable for use in preventing hydroxy side reactions.
  • Representative hydroxy protecting groups include, but are not limited to, alkyl groups such as methyl, ethyl and t-butyl groups; acyl groups such as alkanoyl groups (e.g., acetyl); arylmethyl groups such as benzyl (Bn), Oxybenzyl (PMB), 9-fluorenylmethyl (Fm) and diphenylmethyl (diphenylmethyl, DPM); silyl groups such as trimethylsilyl (TMS) and tert-butyl Dimethylsilyl (TBS) and the like.
  • alkyl groups such as methyl, ethyl and t-butyl groups
  • acyl groups such as alkanoyl groups (e.g., acetyl)
  • arylmethyl groups such as benzyl (Bn), Oxybenzyl (PMB), 9-fluoreny
  • the compounds of the present invention can be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments set forth below, combinations thereof with other chemical synthetic methods, and those well known to those skilled in the art. Equivalent alternatives, preferred embodiments include, but are not limited to, embodiments of the invention.
  • the solvent used in the present invention is commercially available.
  • the present invention employs the following abbreviations: aq for water; HATU for O-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate ; EDC stands for N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride; m-CPBA stands for 3-chloroperoxybenzoic acid; eq stands for equivalent, equivalent; CDI stands for Carbonyldiimidazole; DCM stands for dichloromethane; PE stands for petroleum ether; DIAD stands for diisopropyl azodicarboxylate; DMF stands for N,N-dimethylformamide; DMSO stands for dimethyl sulfoxide; EtOAc stands for acetic acid Esters; EtOH for ethanol; MeOH for methanol; CBz for benzyl
  • Synthetic compound A-1 is prepared from (A-1-1), and is synthesized in detail as shown in the following figure:
  • Correlation characterization data is as follows: LCMS m / z: 206.1 [M + H] (shows hydrolysis of borate ester to boric acid).
  • reaction mixture was concentrated by rotary evaporation, the residue was diluted with 1,4-dioxane (5.00 mL) with, To this mixture was added Pd (dppf) Cl 2 .CH 2 Cl 2 (294.97mg, 361.20 ⁇ mol, 0.21eq) and K 2 CO 3 (2M, 1.81 mL, 2.10 eq), the reaction atmosphere was replaced with nitrogen, and the reaction mixture was stirred for 2 hours in a microwave oven at 120 °C. LCMS monitored a large amount of material remaining and the product was detected.
  • the reaction liquid was directly concentrated and dried, and the residue was purified by a column chromatography to obtain a crude product. The crude product was purified by preparative HPLC.
  • the preparation of the compound of the example of Table 1 can be carried out by referring to the similar step method of the above-mentioned Preparation Example A-1, except that the starting material is used in the following step to replace the starting material 1a with the boric acid in the following table.
  • Synthetic compound A-1 was synthesized from (A-1-1a), and the synthesis route 2 was as follows, as shown in the following figure:
  • Example A-8 The preparation of the compound of this example can be carried out by referring to the procedure similar to the route of the above Preparation Example A-7, except that the starting material A-8-1 is used instead of the starting material A in the step 2. 7-1, the crude product was separated and purified by a plate and sent to a neutral machine (mobile phase: water/acetonitrile) to obtain A-8.
  • a neutral machine mobile phase: water/acetonitrile
  • the preparation of the compound of the example in Table 2 can be carried out by referring to the procedure similar to the route of the above Preparation Example A-6, except that the starting material is used in the step 3, and the boronic acid in the following table is substituted for the raw material 1a.
  • EXAMPLE B-7 The preparation of the compound of this example can be carried out by referring to the above-mentioned method for preparing the route of Example A-1 except that in the step 7, the starting material 2 g of boric acid is used instead of the starting material 2a.
  • EXAMPLE B-10 The preparation of the compound of this example can be carried out by referring to the method of Scheme 2 of the above-mentioned Preparation Example A-1, except that the starting material boric acid 2j is used instead of the starting material 2a in the step 8. The crude product was purified to give the desired product B-10.
  • the preparation of the compound of the example in Table 5 can be carried out by referring to the above-mentioned method for preparing the route of the example A-1, except that the starting material is used in the step 8 in the following table to obtain the corresponding compound instead of the raw material 2a.
  • the preparation of B-13-1 can be carried out by referring to the above-mentioned method for preparing the route of Example A-1 except that the starting material 2m of the starting material is used in place of the raw material 2a.
  • the crude product was purified by preparative TLC EtOAc (EtOAc (EtOAc)
  • the reaction solution was diluted with acetic acid (6 mL), and isopropyl nitrite (1.01 g, 8.66 mmol, 1.17 mL, 1.10 eq) was added dropwise to the reaction mixture, and the reaction mixture was stirred at 25 ° C for 3 hours. After LC-MS showed that the reaction mixture was completed, the reaction mixture was diluted with water (10 mL), and extracted with ethyl acetate and saturated sodium bicarbonate, and the organic phase was washed twice with brine. The organic layer was concentrated under reduced pressure, and the crude material was purified from m.
  • EXAMPLE B-14 The preparation of the compound of this example can be carried out by referring to the method of the above-mentioned Preparation Example A-1, Route 2, except that the starting material, boric acid 2n, is used in the step 8 instead of the starting material 2a. Compound B-14 was obtained.
  • EXAMPLE B-15 The preparation of the compound of this example can be carried out by referring to the method of the above-mentioned Preparation Example A-1, Route 2, except that the starting material boric acid 2o is used in the step 8 instead of the starting material 2a. Compound B-15 was obtained.
  • Example B-16 The preparation of the compound of this example can be carried out by referring to the method of the above-mentioned Preparation Example A-1, Route 2, except that the starting material 2b of the starting material is used instead of the starting material 2a in the step 8. The crude product was isolated and purified by pre-HPLC to give Compound B-16.
  • Example B-17 The preparation of the compound of this example can be carried out by referring to the method of the above-mentioned Preparation Example A-1, Route 2, except that the starting material boric acid 2q is used in the step 8 instead of the starting material 2a.
  • the crude product was purified by pre-HPLC to give compound B-17.
  • the preparation of the compound B-18-1 can be carried out by referring to the above-mentioned method for preparing the route of the example A-1 except that the starting material boric acid 2r is used instead of the starting material 2a in the step 8.
  • Example B-22-1 The preparation of the compound of this example can be carried out by referring to the above-mentioned method for preparing the route of Example A-1 except that the starting material 2t of the starting material is used instead of the starting material 2a in the step 8.
  • reaction solution was replaced with nitrogen three times, replaced with hydrogen three times, and stirred at 40 ° C under a hydrogen pressure of 50 psi for 12 hours.
  • LC-MS showed that the starting material was consumed completely, filtered with celite, and the filtrate was decompressed at 50 ° C. Concentrated and the crude was purified by prep HPLC to afford B-22.
  • the preparation of the present compound can be carried out by referring to the method of the above-mentioned Preparation Example A-1, Route 1, except that the starting material, boric acid 2u-2, is used in the first step, and the crude product, which is concentrated in the reaction liquid, is directly used in the next step.
  • EXAMPLE B-23 The preparation of the compound of this example can be carried out by referring to the method of the eighth step of the above-mentioned Preparation Example A-1, Route 2, except that in the step 8, the starting material 2u boric acid is used instead of the starting material 2a. Compound B-23 was obtained.
  • Methyl tert-butyl ether (10 mL) was added to the compound (1,5-cyclooctadiene)(methoxy)indole (I) dimer (54.69 mg, 82.50 ⁇ mol, 0.03 eq) under nitrogen.
  • EXAMPLE B-28 The preparation of the compound of this example can be carried out by referring to the method of the eighth step of the above-mentioned Preparation Example A-1, Route 2, except that the starting material boric acid 2y is used instead of the starting material 2a in the step 8. Compound B-28 was obtained.
  • the preparation of the compound of Example 6 can be carried out by referring to the above-mentioned preparation method of Example B-28, except that the starting material is used in step 1 instead of the starting material 2y-1 to obtain the corresponding compound.
  • Diterpenes (I) (57.47 mg, 86.70 ⁇ mol, 0.03 eq) and 4,4'-di-tert-butyl-2,2'-bipyridine (23.27 mg, 86.70 ⁇ mol, 0.03 eq), this mixture at 70-80
  • the compound 2ab-2 507.00 mg, 2.89 mmol, 1.00 eq was added to the reaction system.
  • EXAMPLE B-32 The preparation of the compound of this example can be carried out by referring to the method of the eighth step of the above Preparation Example A-1, Route 2, except that the starting material boric acid 2ab is used in the step 8 instead of the starting material 2a. Compound B-32 was obtained.
  • EXAMPLE B-34 The preparation of the compound of this example can be carried out by referring to the above-mentioned Preparation Example B-32 route except that the starting material isopropylmagnesium bromide is used in the step 1 instead of the starting material 2u-1b. Compound B-34 was obtained.
  • Example B-23 The preparation of the compound of the present example can be carried out by referring to the method of the eighth step of the above Preparation Example A-1, Route 2, except that the starting materials boric acid 2ac and 2ac-a are used in the step 8 instead of the raw material. 2a. Compounds B-33 and B-33-1a were obtained.
  • the synthesized compound C-1 was started from (C-1-1), and the detailed synthesis route was as follows:
  • Trifluoroacetic anhydride 3a (4.74 g, 22.56 mmol, 3.14 mL, 1.2 eq.) was added dropwise to a solution of compound C-1-2 (3.00 g, 18.80 mmol, 1.00 eq) in DMF (20.00 mL). Stir at 0 ° C for 4 hours. The solvent was evaporated to dryness under reduced pressure and filtered to afford crude compound C-1-3.
  • reaction solution was stirred at 15 ° C for 60 hours, poured into ice water (140 mL), stirred for 15 min, and extracted with ethyl acetate (75 mL) three times. The extract was washed twice with water (40 mL) and brine (50 mL) Dry, filter and concentrate to give crude C-12-1.
  • the preparation of the compound of the example in Table 9 can be carried out by referring to the above-mentioned preparation example C-19 route method.
  • the crude product obtained by concentration of the mixture was prepared by reversed phase (column: Phenomenex Gemini 150*25 mm*10 ⁇ m; mobile phase: [A: purified water (10 mM NH 4 HCO 3 ), B: ACN]; B%: 18% - 48%, After 3 min), C-56 was obtained after purification.
  • the C-60 correlation characterization data is as follows: LCMS m/z: 471.2 [M+H].
  • Example C-73 The preparation of the compound of this example can be carried out by the method of the above-mentioned preparation of the compound C-62 to give the compound C-73.
  • Example C-74 The preparation of the compound of this example can be carried out by the method of the above-mentioned preparation of the compound C-62 to give the compound C-74.
  • Ethylene glycol monomethyl ether (89.38 mg, 1.17 mmol, 92.62 ⁇ L, 2.5 eq) was dissolved in tetrahydrofuran (3 mL), then sodium hydrogen (46.98 mg, 1.17 mmol, 60% purity, 2.5 eq) was added at 20 ° C Stir for 2 hours.
  • Compound C-65-2 (0.13 g, 469.83 ⁇ mol, 1 eq) was then added and this mixture was stirred at 70-80 ° C for 5 hours.
  • the preparation of the compound of the example in Table 10 can be carried out by referring to the above-mentioned preparation example C-68 route method.
  • Example C-77 The preparation of the compound of this example can be carried out by the method of the above-mentioned preparation of the compound C-75 to give the compound C-77.
  • Example C-78 The preparation of the compound of this example can be carried out by the method of the above-mentioned preparation of Example C-75 to give Compound C-78.
  • Example C-79 The preparation of the compound of this example can be carried out by the method of the above-mentioned preparation of the compound C-77 to give the compound C-79.
  • the mixture was extracted three times with ethyl acetate (60 mL each), and the organic phase was combined and washed twice with saturated brine (30 mL), dried over anhydrous sodium sulfate, filtered, and evaporated.
  • the column was purified by preparative separation: Phenomenex Gemini 150*25 mm*10 ⁇ m; mobile phase: [H 2 O (10 mM NH 4 HCO 3 )-ACN]; B%: 10%-40%, 10 min.
  • the target compound C-80 was collected.
  • the pharmacological activity of the compounds of the invention is determined by the following in vitro assay for detecting A2a receptor activity.
  • the A 2a stable cell line was constructed by Shanghai WuXi PharmaTech, host cell CHO.
  • Fluo-4Direct kit (Invitrogen, Cat. No. F10471). After the fluorescent detection reagent (specifically binding to calcium ions and causing an increase in fluorescence signal) in the kit is incubated with the cells for a suitable period of time, the addition of the compound stimulates the cells to cause changes in intracellular calcium flux, thereby causing changes in the fluorescent signal, which can be reflected The strength of the agonistic or inhibitory activity of the compound.
  • test compound was dissolved in DMSO to prepare a 10 mM mother liquor.
  • the test compound was diluted to 0.2 mM with DMSO, and the reference compound CGS-15943 was diluted with DMSO to 0.015 mM.
  • 10 points of serial dilutions were performed with ECHO, 900 nl was transferred to the compound plate (Greiner-781280), and 30 ul of compound dilution buffer was added.
  • the final starting concentration of the test compound was 1 uM and CGS-15943 was 0.075 uM.
  • a 2A cells were resuspended in culture medium to 1 ⁇ 10 6 cells/ml after resuscitation, and 20 ⁇ l/well was seeded into 384-well polylysine-coated cell plates (Greiner-781946), 5% CO 2 , 37 ° C. Incubate overnight in the incubator.
  • the prepared cell plates from the previous day were taken out from the incubator, and 20 ⁇ l of 2X Fluo-4DirectTM buffer, 5% CO 2 was added to each well, and incubated in a 37 ° C incubator for 50 minutes at room temperature for 10 minutes.
  • Agonist NECA dilution NECA with a starting concentration of 0.15 mM was subjected to 10-point 3-fold serial dilution with Echo, followed by transfer of 900 nL to the corresponding compound plate; then 30 ⁇ l of compound dilution buffer was added to the corresponding compound plate. The final starting concentration was 750 nM. Run the FLIPR instrument software and add 10 ul of compound dilution buffer to the cell plate according to the setup procedure to read the fluorescence signal. An additional 10 ul of the agonist reference compound at the indicated concentration was added to the cell plate and the fluorescent signal was read.
  • the data was exported by the "Max-Min”, "Read 90to Maximum allowed” method in the software, and the EC80 of the A2A cell line was calculated to prepare an agonist at a concentration of 6X EC80.
  • a reference compound agonist of the corresponding cell 6X EC80 concentration was prepared with a buffered saline solution, and 30 ul/well was added to the corresponding compound plate for use.
  • test compound was dissolved in DMSO to prepare a 10 mmol/L stock solution.
  • stock solution was diluted with 50% ACN/50 mM phosphate buffer solution (pH 7.4) to prepare a standard solution (1 ⁇ M, 20 ⁇ M, 200 ⁇ M).
  • the rodent pharmacological characteristics of the compound after intravenous injection and oral administration were tested by a standard protocol.
  • the candidate compound was formulated into a clear solution, and the mice were administered a single intravenous injection and a uniform suspension to the mice for single oral administration.
  • the vehicle was intravenously injected with 5% DMSO/95% 10% Cremophor EL, and the oral vehicle was 1% tween 80, 9% PEG 400, 90% water.
  • the compounds of the present invention can significantly increase the pharmacokinetic index of mice.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

式(Ⅰ)所示化合物、其异构体或其药学上可接受的盐,以及其在制备治疗与A 2A受体相关疾病药物中的应用,其中R 1、R 2、R 3、环A、环B、n和m为如本文中定义的。

Description

作为A 2A受体抑制剂的[1,2,4]***并[1,5-c]嘧啶衍生物
相关申请的引用
本申请要求于2017年04月07日向中华人民共和国国家知识产权局提交的第201710224553.9号、2017年08月24日向中华人民共和国国家知识产权局提交的第201710737871.5号和2018年02月08日向中华人民共和国国家知识产权局提交的第201810129208.1号中国发明专利申请的权益,在此将其全部内容以援引的方式整体并入本文中。
技术领域
本发明涉及式(Ⅰ)所示化合物、其异构体或其药学上可接受的盐,并涉及其在制备治疗与A 2A受体相关疾病药物中的应用。
背景技术
腺苷A 2A受体在人体组织中有广泛的分布,这个受体在脾脏,胸腺,白血球,血小板,GABA型神经元和嗅球等组织器官中有高表达。同时在心脏,肺部,血管,和脑等其他部位中也有表达。腺苷A 2A受体一般和其他GPCR共同存在并结合在一起成为异质二聚体,比如A 2A受体可以和多巴胺D 2,***素CB 1,谷氨酸mGluR5等形成异质二聚体。腺苷A 2A受体在调节血管舒张,支持新血管的形成,保护身体组织免受由炎症引起的伤害等生命活动中有着重要的作用;腺苷A 2A受体也影响基底神经节间接通路的活性程度。
在实体瘤中,细胞组织的分解和缺氧的环境造成了ATP大量分解,因此导致细胞外腺苷富集,浓度异常地高,为正常值的10-20倍。高浓度的腺苷和A 2A受体的结合会激活腺苷信号通路。这个信号通路是一种在机体组织损伤时通过免疫抑制来保护了机体组织的机制。腺苷信号通路的激活导致了对先天性免疫应答的长期抑制,这种长期抑制会产生免疫耐受性,进而导致恶性肿瘤失去控制的生长腺苷和A 2A受体在白血球里(譬如淋巴细胞,T淋巴细胞,自然杀手细胞,树突状细胞等)的结合抑制了这些白血球在免疫***中应有的效应子功能。腺苷与A 2A受体的结合使CD39,CD73和CTLA4(T细胞检查点)的表达增加,从而产生更多的具有更强免疫抑制性的T reg细胞。阻断A 2A受体的腺苷信号通路可以减少对免疫***的抑制作用,增强T细胞的免疫功能,因而被认为是很有希望的能抑制肿瘤生长的负面反馈机制。
文献J.Med.Chem.2012,55,1898-1903报道了化合物A,作为腺苷A 2A受体拮抗剂,用于治疗帕金森疾病的治疗。
发明内容
本发明提供了式(Ⅰ)所示化合物或其药学上可接受的盐,
Figure PCTCN2018082119-appb-000002
其中,
R 1选自H,或选自任选被1、2或3个R取代的:C 1-3烷基、C 1-3烷基-O-C 1-3烷基-或C 3-6环烷基、C 1-3烷基-C(=O)NH-;
R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-6烷基或C 1-6杂烷基;
R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-6烷基或C 1-6杂烷基;
n选自0、1、2或3;
m选自0、1、2或3;
环A选自:6~10元芳基、5~10元杂芳基、5~10元杂环烷基或5~10元杂环烯基;
环B选自:苯基或5~6元杂芳基;
R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:C 1-3烷基、C 3-6环烷基、C 3-6环烷基-NH-、或苯基;
R’选自:F、Cl、Br、I、OH、NH 2
Figure PCTCN2018082119-appb-000003
所述C 1-6杂烷基、5~10元杂芳基、5~10元杂环烷基或5~10元杂环烯基之“杂”分别独立地选自:N、O、S、NH、-C(=O)-、-C(=O)O-或-C(=O)NH-;
上述杂原子或杂原子团的数目分别独立地选自1、2、3或4。
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’ 取代的:Me、Et、
Figure PCTCN2018082119-appb-000004
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN、Me、Et、
Figure PCTCN2018082119-appb-000005
Figure PCTCN2018082119-appb-000006
本发明的一些方案中,上述R 1选自H,或选自任选被1、2或3个R取代的:Me、Et、
Figure PCTCN2018082119-appb-000007
Figure PCTCN2018082119-appb-000008
本发明的一些方案中,上述R 1选自:H、Me、Et、CF 3
Figure PCTCN2018082119-appb-000009
Figure PCTCN2018082119-appb-000010
本发明的一些方案中,上述R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-3烷基或C 1-3烷氧基。
本发明的一些方案中,上述R 2分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000011
本发明的一些方案中,上述R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-3烷基或C 1-3烷氧基。
本发明的一些方案中,上述R 3分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000012
本发明的一些方案中,上述环A选自:苯基、吡啶基、四氢吡喃基、3,6-二氢-2H-吡喃基、哌啶基、1,2,3,6-四氢吡啶基、1H-吲哚基、1H-吲唑基、1H-苯并[d]咪唑基、苯并[d][1,3]二氧杂环戊烯基、二氢吲哚-2-酮基、1H-苯并[d][1,2,3]***基、喹啉基或1,2,3,4-四氢喹啉基。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000013
选自:
Figure PCTCN2018082119-appb-000014
Figure PCTCN2018082119-appb-000015
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000016
选自:
Figure PCTCN2018082119-appb-000017
Figure PCTCN2018082119-appb-000018
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000019
选自:
Figure PCTCN2018082119-appb-000020
Figure PCTCN2018082119-appb-000021
本发明的一些方案中,上述环B选自:苯基、吡啶基、咪唑基、吡唑基、呋喃基、噻吩基、噻唑基。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000022
选自:
Figure PCTCN2018082119-appb-000023
Figure PCTCN2018082119-appb-000024
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000025
选自:
Figure PCTCN2018082119-appb-000026
Figure PCTCN2018082119-appb-000027
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000028
选自:
Figure PCTCN2018082119-appb-000029
Figure PCTCN2018082119-appb-000030
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:Me、Et、
Figure PCTCN2018082119-appb-000031
其他变量如上述所定义。
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN、Me、Et、
Figure PCTCN2018082119-appb-000032
Figure PCTCN2018082119-appb-000033
其他变量如上述所定义。
本发明的一些方案中,上述R 1选自H,或选自任选被1、2或3个R取代的:Me、Et、
Figure PCTCN2018082119-appb-000034
Figure PCTCN2018082119-appb-000035
其他变量如上述所定义。
本发明的一些方案中,上述R 1选自:H、Me、Et、CF 3
Figure PCTCN2018082119-appb-000036
Figure PCTCN2018082119-appb-000037
其他变量如上述所定义。
本发明的一些方案中,上述R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-3烷基或C 1-3烷氧基,其他变量如上述所定义。
本发明的一些方案中,上述R 2分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000038
其他变量如上述所定义。
本发明的一些方案中,上述R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-3烷基或C 1-3烷氧基,其他变量如上述所定义。
本发明的一些方案中,上述R 3分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000039
其他变量如上述所定义。
本发明的一些方案中,上述环A选自:苯基、吡啶基、四氢吡喃基、3,6-二氢-2H-吡喃基、哌啶基、1,2,3,6-四氢吡啶基、1H-吲哚基、1H-吲唑基、1H-苯并[d]咪唑基、苯并[d][1,3]二氧杂环戊烯基、二氢吲哚-2-酮基、1H-苯并[d][1,2,3]***基、喹啉基或1,2,3,4-四氢喹啉基,其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000040
选自:
Figure PCTCN2018082119-appb-000041
Figure PCTCN2018082119-appb-000042
Figure PCTCN2018082119-appb-000043
其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000044
选自:
Figure PCTCN2018082119-appb-000045
Figure PCTCN2018082119-appb-000046
Figure PCTCN2018082119-appb-000047
其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000048
选自:
Figure PCTCN2018082119-appb-000049
Figure PCTCN2018082119-appb-000050
Figure PCTCN2018082119-appb-000051
其他变量如上述所定义。
本发明的一些方案中,上述环B选自:苯基、吡啶基、咪唑基、吡唑基、呋喃基、噻吩基、噻唑基,其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000052
选自:
Figure PCTCN2018082119-appb-000053
Figure PCTCN2018082119-appb-000054
其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000055
选自:
Figure PCTCN2018082119-appb-000056
Figure PCTCN2018082119-appb-000057
Figure PCTCN2018082119-appb-000058
其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000059
选自:
Figure PCTCN2018082119-appb-000060
Figure PCTCN2018082119-appb-000061
Figure PCTCN2018082119-appb-000062
其他变量如上述所定义。
本发明提供了式(Ⅰ)所示化合物或其药学上可接受的盐,
Figure PCTCN2018082119-appb-000063
其中,
R 1选自H,或选自任选被1、2或3个R取代的:C 1-3烷基、C 1-3烷基-O-C 1-3烷基-或C 3-6环烷基、C 1-3 烷基-C(=O)NH-;
R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-6烷基或C 1-6杂烷基;
R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-6烷基或C 1-6杂烷基;
n选自0、1、2或3;
m选自0、1、2或3;
环A选自:6~10元芳基、5~10元杂芳基、5~10元杂环烷基或5~10元杂环烯基;
环B选自:苯基或5~6元杂芳基;
R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:C 1-3烷基、C 3-6环烷基、C 3-6环烷基-NH-、或苯基;
R’选自:F、Cl、Br、I、OH、NH 2
Figure PCTCN2018082119-appb-000064
所述C 1-6杂烷基、5~10元杂芳基、5~10元杂环烷基或5~10元杂环烯基之“杂”分别独立地选自:N、O、S、NH、-C(=O)-、-C(=O)O-或-C(=O)NH-;
上述杂原子或杂原子团的数目分别独立地选自1、2、3或4。
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:Me、Et、
Figure PCTCN2018082119-appb-000065
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN、Me、Et、
Figure PCTCN2018082119-appb-000066
Figure PCTCN2018082119-appb-000067
本发明的一些方案中,上述R 1选自H,或选自任选被1、2或3个R取代的:Me、Et、
Figure PCTCN2018082119-appb-000068
Figure PCTCN2018082119-appb-000069
本发明的一些方案中,上述R 1选自:H、Me、Et、CF 3
Figure PCTCN2018082119-appb-000070
Figure PCTCN2018082119-appb-000071
本发明的一些方案中,上述R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-3烷基或C 1-3烷氧基。
本发明的一些方案中,上述R 2分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000072
本发明的一些方案中,上述R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-3烷基或C 1-3烷氧基。
本发明的一些方案中,上述R 3分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000073
本发明的一些方案中,上述环A选自:苯基、吡啶基、四氢吡喃基、3,6-二氢-2H-吡喃基、哌啶基、1,2,3,6-四氢吡啶基、1H-吲哚基、1H-吲唑基、1H-苯并[d]咪唑基、苯并[d][1,3]二氧杂环戊烯基、二氢吲哚-2-酮基、1H-苯并[d][1,2,3]***基、喹啉基或1,2,3,4-四氢喹啉基。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000074
选自:
Figure PCTCN2018082119-appb-000075
Figure PCTCN2018082119-appb-000076
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000077
选自:
Figure PCTCN2018082119-appb-000078
Figure PCTCN2018082119-appb-000079
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000080
选自:
Figure PCTCN2018082119-appb-000081
Figure PCTCN2018082119-appb-000082
本发明的一些方案中,上述环B选自:苯基、吡啶基、咪唑基、吡唑基、呋喃基、噻吩基、噻唑基。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000083
选自:
Figure PCTCN2018082119-appb-000084
Figure PCTCN2018082119-appb-000085
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000086
选自:
Figure PCTCN2018082119-appb-000087
Figure PCTCN2018082119-appb-000088
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000089
选自:
Figure PCTCN2018082119-appb-000090
Figure PCTCN2018082119-appb-000091
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:Me、Et、
Figure PCTCN2018082119-appb-000092
其他变量如上述所定义。
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN、Me、Et、
Figure PCTCN2018082119-appb-000093
Figure PCTCN2018082119-appb-000094
其他变量如上述所定义。
本发明的一些方案中,上述R 1选自H,或选自任选被1、2或3个R取代的:Me、Et、
Figure PCTCN2018082119-appb-000095
Figure PCTCN2018082119-appb-000096
其他变量如上述所定义。
本发明的一些方案中,上述R 1选自:H、Me、Et、CF 3
Figure PCTCN2018082119-appb-000097
Figure PCTCN2018082119-appb-000098
其他变量如上述所定义。
本发明的一些方案中,上述R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-3烷基或C 1-3烷氧基,其他变量如上述所定义。
本发明的一些方案中,上述R 2分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000099
其他变量如上述所定义。
本发明的一些方案中,上述R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-3烷基或C 1-3烷氧基,其他变量如上述所定义。
本发明的一些方案中,上述R 3分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000100
其他变量如上述所定义。
本发明的一些方案中,上述环A选自:苯基、吡啶基、四氢吡喃基、3,6-二氢-2H-吡喃基、哌啶基、1,2,3,6-四氢吡啶基、1H-吲哚基、1H-吲唑基、1H-苯并[d]咪唑基、苯并[d][1,3]二氧杂环戊烯基、二氢吲哚-2-酮基、1H-苯并[d][1,2,3]***基、喹啉基或1,2,3,4-四氢喹啉基,其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000101
选自:
Figure PCTCN2018082119-appb-000102
Figure PCTCN2018082119-appb-000103
Figure PCTCN2018082119-appb-000104
其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000105
选自:
Figure PCTCN2018082119-appb-000106
Figure PCTCN2018082119-appb-000107
Figure PCTCN2018082119-appb-000108
其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000109
选自:
Figure PCTCN2018082119-appb-000110
Figure PCTCN2018082119-appb-000111
Figure PCTCN2018082119-appb-000112
其他变量如上述所定义。
本发明的一些方案中,上述环B选自:苯基、吡啶基、咪唑基、吡唑基、呋喃基、噻吩基、噻唑基,其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000113
选自:
Figure PCTCN2018082119-appb-000114
Figure PCTCN2018082119-appb-000115
其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000116
选自:
Figure PCTCN2018082119-appb-000117
Figure PCTCN2018082119-appb-000118
Figure PCTCN2018082119-appb-000119
其他变量如上述所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000120
选自:
Figure PCTCN2018082119-appb-000121
Figure PCTCN2018082119-appb-000122
Figure PCTCN2018082119-appb-000123
其他变量如上述所定义。
本发明提供了式(Ⅰ)所示化合物或其药学上可接受的盐,
Figure PCTCN2018082119-appb-000124
其中,
R 1选自H、CN、
Figure PCTCN2018082119-appb-000125
或选自任选被1、2或3个R取代的:C 1-3烷基、C 1-3烷基-O-C 1-3烷基-或C 3-6环烷基、C 1-3烷基-C(=O)NH-;
R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-6烷基或C 1-6杂烷基;
R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-6烷基或C 1-6杂烷基;
n选自0、1、2或3;
m选自0、1、2或3;
环A选自:6~10元芳基、5~10元杂芳基、5~10元杂环烷基或5~10元杂环烯基;
环B选自:苯基或5~6元杂芳基;
R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:C 1-3烷基、C 1-3烷氨基、C 1-3烷氧基、C 3-6环烷基、C 3-6环烷基-NH-、3~6元杂环烷基、3~6元杂环烷基-O-、或苯基;
R’选自:F、Cl、Br、I、OH、NH 2
Figure PCTCN2018082119-appb-000126
所述C 1-6杂烷基、5~10元杂芳基、5~10元杂环烷基、3~6元杂环烷基或5~10元杂环烯基之“杂”分别独立地选自:N、O、S、NH、-C(=O)-、-C(=O)O-或-C(=O)NH-;
上述杂原子或杂原子团的数目分别独立地选自1、2、3或4。
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:Me、Et、
Figure PCTCN2018082119-appb-000127
其他变量如本发明所定义。
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN、Me、Et、
Figure PCTCN2018082119-appb-000128
Figure PCTCN2018082119-appb-000129
其他变量如本发明所定义。
本发明的一些方案中,上述R 1选自H、CN、
Figure PCTCN2018082119-appb-000130
或选自任选被1、2或3个R取代的:Me、Et、
Figure PCTCN2018082119-appb-000131
其他变量如本发明所定义。
本发明的一些方案中,上述R 1选自:H、CN、
Figure PCTCN2018082119-appb-000132
Me、Et、CF 3
Figure PCTCN2018082119-appb-000133
Figure PCTCN2018082119-appb-000134
其他变量如本发明所定义。
本发明的一些方案中,上述R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-3烷基或C 1-3烷氧基,其他变量如本发明所定义。
本发明的一些方案中,上述R 2分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000135
其他变量如本发明所定义。
本发明的一些方案中,上述R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-3烷基或C 1-3烷氧基,其他变量如本发明所定义。
本发明的一些方案中,上述R 3分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000136
其他变量如本发明所定义。
本发明的一些方案中,上述环A选自:苯基、吡啶基、四氢吡喃基、3,6-二氢-2H-吡喃基、哌啶基、1,2,3,6-四氢吡啶基、1H-吲哚基、1H-吲唑基、1H-苯并[d]咪唑基、苯并[d][1,3]二氧杂环戊烯基、二氢吲哚-2-酮基、1H-苯并[d][1,2,3]***基、喹啉基或1,2,3,4-四氢喹啉基,其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000137
选自:
Figure PCTCN2018082119-appb-000138
Figure PCTCN2018082119-appb-000139
Figure PCTCN2018082119-appb-000140
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000141
选自:
Figure PCTCN2018082119-appb-000142
Figure PCTCN2018082119-appb-000143
Figure PCTCN2018082119-appb-000144
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000145
选自:
Figure PCTCN2018082119-appb-000146
Figure PCTCN2018082119-appb-000147
Figure PCTCN2018082119-appb-000148
其他变量如本发明所定义。
本发明的一些方案中,上述环B选自:苯基、吡啶基、咪唑基、吡唑基、呋喃基、噻吩基、噻唑基,其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000149
选自:
Figure PCTCN2018082119-appb-000150
Figure PCTCN2018082119-appb-000151
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000152
选自:
Figure PCTCN2018082119-appb-000153
Figure PCTCN2018082119-appb-000154
Figure PCTCN2018082119-appb-000155
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000156
选自:
Figure PCTCN2018082119-appb-000157
Figure PCTCN2018082119-appb-000158
Figure PCTCN2018082119-appb-000159
其他变量如本发明所定义。
本发明的一些方案中,上述化合物或其药学上可接受的盐,其选自:
Figure PCTCN2018082119-appb-000160
其中,
R 1、R 2、R 3如本发明所定义。
本发明提供了式(Ⅰ)所示化合物、其异构体或其药学上可接受的盐,
Figure PCTCN2018082119-appb-000161
其中,
R 1选自H、CN、COOH、
Figure PCTCN2018082119-appb-000162
或选自任选被1、2或3个R取代的:C 1-3烷基、C 1-3烷基-O-C 1- 3烷基-或C 3-6环烷基、C 1-3烷基-C(=O)NH-;
R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-6烷基或C 1-6杂烷基;
R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-6烷基或C 1-6杂烷基;
n选自0、1、2或3;
m选自0、1、2或3;
环A选自:6~10元芳基、5~10元杂芳基、5~10元杂环烷基或5~10元杂环烯基;
环B选自:苯基或5~6元杂芳基;
R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:C 1-3烷基、C 1-3烷氨基、C 1-3烷氧基、C 3-6环烷基、C 3-6环烷基-NH-、3~6元杂环烷基、3~6元杂环烷基-O-、或苯基;
R’选自:F、Cl、Br、I、OH、NH 2
Figure PCTCN2018082119-appb-000163
所述C 1-6杂烷基、5~10元杂芳基、5~10元杂环烷基、3~6元杂环烷基或5~10元杂环烯基之“杂”分别独立地选自:N、O、S、NH、-C(=O)-、-C(=O)O-或-C(=O)NH-;
上述杂原子或杂原子团的数目分别独立地选自1、2、3或4。
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:Me、Et、
Figure PCTCN2018082119-appb-000164
其他变量如本发明所定义。
本发明的一些方案中,上述R选自F、Cl、Br、I、OH、NH 2、CN、Me、Et、
Figure PCTCN2018082119-appb-000165
Figure PCTCN2018082119-appb-000166
其他变量如本发明所定义。
本发明的一些方案中,上述R 1选自H、CN、COOH、
Figure PCTCN2018082119-appb-000167
或选自任选被1、2或3个R取代的:Me、Et、
Figure PCTCN2018082119-appb-000168
其他变量如本发明所定义。
本发明的一些方案中,上述R 1选自:H、CN、COOH、
Figure PCTCN2018082119-appb-000169
Me、Et、CF 3
Figure PCTCN2018082119-appb-000170
Figure PCTCN2018082119-appb-000171
其他变量如本发明所定义。
本发明的一些方案中,上述R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-3烷基或C 1-3烷氧基,其他变量如本发明所定义。
本发明的一些方案中,上述R 2分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000172
其他变量如本发明所定义。
本发明的一些方案中,上述R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-3烷基或C 1-3烷氧基,其他变量如本发明所定义。
本发明的一些方案中,上述R 3分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
Figure PCTCN2018082119-appb-000173
其他变量如本发明所定义。
本发明的一些方案中,上述环A选自:苯基、吡啶基、四氢吡喃基、3,6-二氢-2H-吡喃基、哌啶基、1,2,3,6-四氢吡啶基、1H-吲哚基、1H-吲唑基、1H-苯并[d]咪唑基、苯并[d][1,3]二氧杂环戊烯基、二氢吲哚-2-酮基、1H-苯并[d][1,2,3]***基、喹啉基或1,2,3,4-四氢喹啉基,其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000174
选自:
Figure PCTCN2018082119-appb-000175
Figure PCTCN2018082119-appb-000176
Figure PCTCN2018082119-appb-000177
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000178
选自:
Figure PCTCN2018082119-appb-000179
Figure PCTCN2018082119-appb-000180
Figure PCTCN2018082119-appb-000181
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000182
选自:
Figure PCTCN2018082119-appb-000183
Figure PCTCN2018082119-appb-000184
Figure PCTCN2018082119-appb-000185
其他变量如本发明所定义。
本发明的一些方案中,上述环B选自:苯基、吡啶基、咪唑基、吡唑基、呋喃基、噻吩基、噻唑基,其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000186
选自:
Figure PCTCN2018082119-appb-000187
Figure PCTCN2018082119-appb-000188
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000189
选自:
Figure PCTCN2018082119-appb-000190
Figure PCTCN2018082119-appb-000191
Figure PCTCN2018082119-appb-000192
其他变量如本发明所定义。
本发明的一些方案中,上述结构单元
Figure PCTCN2018082119-appb-000193
选自:
Figure PCTCN2018082119-appb-000194
Figure PCTCN2018082119-appb-000195
Figure PCTCN2018082119-appb-000196
其他变量如本发明所定义。
本发明的一些方案中,上述化合物、其异构体或其药学上可接受的盐,其选自:
Figure PCTCN2018082119-appb-000197
其中,
R 1、R 2、R 3如本发明所定义。
本发明还有一些方案是由上述各变量任意组合而来。
本发明提供了下式所示化合物、其异构体或其药学上可接受的盐,其选自:
Figure PCTCN2018082119-appb-000198
Figure PCTCN2018082119-appb-000199
Figure PCTCN2018082119-appb-000200
Figure PCTCN2018082119-appb-000201
Figure PCTCN2018082119-appb-000202
Figure PCTCN2018082119-appb-000203
本发明还提供了一种药物组合物,包括治疗有效量的上述化合物、其异构体或其药学上可接受的盐,以及药学上可接受的载体。
本发明还提供了上述化合物、其异构体或其药学上可接受的盐在制备治疗与A 2A受体相关疾病的药物中的应用。
本发明还提供了上述药物组合物在制备治疗与A 2A受体相关疾病的药物中的应用。
技术效果:
本发明合成了式(I)化合物,获得一类新的腺苷A2a拮抗剂,单药或与抗体联用用于肿瘤免疫治疗。本发明提高化合物的溶解度,同时显著改善了药代动力学方面的特征。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。当本文中出现商品名时,意在指代其对应的商品或其活性成分。这里所采用的术语“药学上可接受的”,是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物的中性形式接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机氨或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物的中性形式接触的方式获得酸加成盐。药学上可接受的酸加成盐的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸,碳酸氢根,磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等;以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡糖醛酸等有机酸的盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与 化学计量的适当的碱或酸反应来制备。
除了盐的形式,本发明所提供的化合物还存在前药形式。本文所描述的化合物的前药容易地在生理条件下发生化学变化从而转化成本发明的化合物。此外,前体药物可以在体内环境中通过化学或生化方法被转换到本发明的化合物。
本发明的某些化合物可以以非溶剂化形式或者溶剂化形式存在,包括水合物形式。一般而言,溶剂化形式与非溶剂化的形式相当,都包含在本发明的范围之内。
本发明的化合物可以存在特定的几何或立体异构体形式。本发明设想所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,所有这些混合物都属于本发明的范围之内。烷基等取代基中可存在另外的不对称碳原子。所有这些异构体以及它们的混合物,均包括在本发明的范围之内。
除非另有说明,术语“对映异构体”或者“旋光异构体”是指互为镜像关系的立体异构体。
除非另有说明,术语“顺反异构体”或者“几何异构体”系由因双键或者成环碳原子单键不能自由旋转而引起。
除非另有说明,术语“非对映异构体”是指分子具有两个或多个手性中心,并且分子间为非镜像的关系的立体异构体。
除非另有说明,“(D)”或者“(+)”表示右旋,“(L)”或者“(-)”表示左旋,“(DL)”或者“(±)”表示外消旋。
除非另有说明,用楔形实线键
Figure PCTCN2018082119-appb-000204
和楔形虚线键
Figure PCTCN2018082119-appb-000205
表示一个立体中心的绝对构型,用直形实线键
Figure PCTCN2018082119-appb-000206
和直形虚线键
Figure PCTCN2018082119-appb-000207
表示立体中心的相对构型,用波浪线
Figure PCTCN2018082119-appb-000208
表示楔形实线键
Figure PCTCN2018082119-appb-000209
或楔形虚线键
Figure PCTCN2018082119-appb-000210
或用波浪线
Figure PCTCN2018082119-appb-000211
表示直形实线键
Figure PCTCN2018082119-appb-000212
和直形虚线键
Figure PCTCN2018082119-appb-000213
本发明的化合物可以存在特定的。除非另有说明,术语“互变异构体”或“互变异构体形式”是指在室温下,不同官能团异构体处于动态平衡,并能很快的相互转化。若互变异构体是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异构体(proton tautomer)(也称质子转移互变异构体(prototropic tautomer))包括通过质子迁移来进行的互相转化,如酮-烯醇异构化和亚胺-烯胺异构化。价键异构体(valence tautomer)包括一些成键电子的重组来进行的相互转化。其中酮-烯醇互变异构化的具体实例是戊烷-2,4-二酮与4-羟基戊-3-烯-2-酮两个互变异构体之间的互变。
除非另有说明,术语“富含一种异构体”、“异构体富集”、“富含一种对映体”或者“对映体富集”指其中一种异构体或对映体的含量小于100%,并且,该异构体或对映体的含量大于等于60%,或者大于等于70%,或者大于等于80%,或者大于等于90%,或者大于等于95%,或者大于等于96%,或者大 于等于97%,或者大于等于98%,或者大于等于99%,或者大于等于99.5%,或者大于等于99.6%,或者大于等于99.7%,或者大于等于99.8%,或者大于等于99.9%。
除非另有说明,术语“异构体过量”或“对映体过量”指两种异构体或两种对映体相对百分数之间的差值。例如,其中一种异构体或对映体的含量为90%,另一种异构体或对映体的含量为10%,则异构体或对映体过量(ee值)为80%。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明某化合物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。本发明的化合物可以在一个或多个构成该化合物的原子上包含非天然比例的原子同位素。例如,可用放射性同位素标记化合物,比如氚( 3H),碘-125( 125I)或C-14( 14C)。又例如,可用重氢取代氢形成氘代药物,氘与碳构成的键比普通氢与碳构成的键更坚固,相比于未氘化药物,氘代药物有降低毒副作用、增加药物稳定性、增强疗效、延长药物生物半衰期等优势。本发明的化合物的所有同位素组成的变换,无论放射性与否,都包括在本发明的范围之内。术语“药学上可接受的载体”是指能够递送本发明有效量活性物质、不干扰活性物质的生物活性并且对宿主或者患者无毒副作用的任何制剂或载体介质代表性的载体包括水、油、蔬菜和矿物质、膏基、洗剂基质、软膏基质等。这些基质包括悬浮剂、增粘剂、透皮促进剂等。它们的制剂为化妆品领域或局部药物领域的技术人员所周知。
术语“赋形剂”通常是指配制有效的药物组合物所需要载体、稀释剂和/或介质。
针对药物或药理学活性剂而言,术语“有效量”或“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。对于本发明中的口服剂型,组合物中一种活性物质的“有效量”是指与该组合物中另一种活性物质联用时为了达到预期效果所需要的用量。有效量的确定因人而异,取决于受体的年龄和一般情况,也取决于具体的活性物质,个案中合适的有效量可以由本领域技术人员根据常规试验确定。
术语“活性成分”、“治疗剂”,“活性物质”或“活性剂”是指一种化学实体,它可以有效地治疗目标紊乱、疾病或病症。
“任选”或“任选地”指的是随后描述的事件或状况可能但不是必需出现的,并且该描述包括其中所述事件或状况发生的情况以及所述事件或状况不发生的情况。
术语“被取代的”是指特定原子上的任意一个或多个氢原子被取代基取代,可以包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧(即=O)时,意味着两个氢 原子被取代。氧取代不会发生在芳香基上。术语“任选被取代的”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当一个连接基团的数量为0时,比如-(CRR) 0-,表示该连接基团为单键。
当其中一个变量选自单键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表单键时表示该结构实际上是A-Z。
当一个取代基为空缺时,表示该取代基是不存在的,比如A-X中X为空缺时表示该结构实际上是A。当一个取代基可以连接到一个环上的一个以上原子时,这种取代基可以与这个环上的任意原子相键合,例如,结构单元
Figure PCTCN2018082119-appb-000214
Figure PCTCN2018082119-appb-000215
表示取代基R可在环己基或者环己二烯上的任意一个位置发生取代。当所列举的取代基中没有指明其通过哪一个原子连接到被取代的基团上时,这种取代基可以通过其任何原子相键合,例如,吡啶基作为取代基可以通过吡啶环上任意一个碳原子连接到被取代的基团上。当所列举的连接基团没有指明其连接方向,其连接方向是任意的,例如,
Figure PCTCN2018082119-appb-000216
中连接基团L为-M-W-,此时-M-W-既可以按与从左往右的读取顺序相同的方向连接环A和环B构成
Figure PCTCN2018082119-appb-000217
也可以按照与从左往右的读取顺序相反的方向连接环A和环B构成
Figure PCTCN2018082119-appb-000218
所述连接基团、取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
除非另有规定,术语“杂”表示杂原子或杂原子团(即含有杂原子的原子团),包括碳(C)和氢(H)以外的原子以及含有这些杂原子的原子团,例如包括氧(O)、氮(N)、硫(S)、硅(Si)、锗(Ge)、铝(Al)、硼(B)、-O-、-S-、=O、=S、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)、-S(=O) 2-,以及任选被取代的-C(=O)N(H)-、-N(H)-、-C(=NH)-、-S(=O) 2N(H)-或-S(=O)N(H)-。
除非另有规定,“环”表示被取代或未被取代的环烷基、杂环烷基、环烯基、杂环烯基、环炔基、杂环炔基、芳基或杂芳基。所谓的环包括单环、联环、螺环、并环或桥环。环上原子的数目通常被定义为环的元数,例如,“5~7元环”是指环绕排列5~7个原子。除非另有规定,该环任选地包含1~3个杂原子。因此,“5~7元环”包括例如苯基、吡啶和哌啶基;另一方面,术语“5~7元杂环烷基环”包括吡 啶基和哌啶基,但不包括苯基。术语“环”还包括含有至少一个环的环系,其中的每一个“环”均独立地符合上述定义。
除非另有规定,术语“杂环”或“杂环基”意指稳定的含杂原子或杂原子团的单环、双环或三环,它们可以是饱和的、部分不饱和的或不饱和的(芳族的),它们包含碳原子和1、2、3或4个独立地选自N、O和S的环杂原子,其中上述任意杂环可以稠合到一个苯环上形成双环。氮和硫杂原子可任选被氧化(即NO和S(O)p,p是1或2)。氮原子可以是被取代的或未取代的(即N或NR,其中R是H或本文已经定义过的其他取代基)。该杂环可以附着到任何杂原子或碳原子的侧基上从而形成稳定的结构。如果产生的化合物是稳定的,本文所述的杂环可以发生碳位或氮位上的取代。杂环中的氮原子任选地被季铵化。一个优选方案是,当杂环中S及O原子的总数超过1时,这些杂原子彼此不相邻。另一个优选方案是,杂环中S及O原子的总数不超过1。如本文所用,术语“芳族杂环基团”或“杂芳基”意指稳定的5、6、7元单环或双环或7、8、9或10元双环杂环基的芳香环,它包含碳原子和1、2、3或4个独立地选自N、O和S的环杂原子。氮原子可以是被取代的或未取代的(即N或NR,其中R是H或本文已经定义过的其他取代基)。氮和硫杂原子可任选被氧化(即NO和S(O)p,p是1或2)。值得注意的是,芳香杂环上S和O原子的总数不超过1。桥环也包含在杂环的定义中。当一个或多个原子(即C、O、N或S)连接两个不相邻的碳原子或氮原子时形成桥环。优选的桥环包括但不限于:一个碳原子、两个碳原子、一个氮原子、两个氮原子和一个碳-氮基。值得注意的是,一个桥总是将单环转换成三环。桥环中,环上的取代基也可以出现在桥上。
杂环化合物的实例包括但不限于:吖啶基、吖辛因基、苯并咪唑基、苯并呋喃基、苯并巯基呋喃基、苯并巯基苯基、苯并恶唑基、苯并恶唑啉基、苯并噻唑基、苯并***基、苯并四唑基、苯并异恶唑基、苯并异噻唑基、苯并咪唑啉基、咔唑基、4aH-咔唑基、咔啉基、苯并二氢吡喃基、色烯、噌啉基十氢喹啉基、2H,6H-1,5,2-二噻嗪基、二氢呋喃并[2,3-b]四氢呋喃基、呋喃基、呋咱基、咪唑烷基、咪唑啉基、咪唑基、1H-吲唑基、吲哚烯基、二氢吲哚基、中氮茚基、吲哚基、3H-吲哚基、异苯并呋喃基、异吲哚基、异二氢吲哚基、异喹啉基、异噻唑基、异恶唑基、亚甲二氧基苯基、吗啉基、萘啶基,八氢异喹啉基、恶二唑基、1,2,3-恶二唑基、1,2,4-恶二唑基、1,2,5-恶二唑基、1,3,4-恶二唑基、恶唑烷基、恶唑基、羟吲哚基、嘧啶基、菲啶基、菲咯啉基、吩嗪、吩噻嗪、苯并黄嘌呤基、酚恶嗪基、酞嗪基、哌嗪基、哌啶基、哌啶酮基、4-哌啶酮基、胡椒基、蝶啶基、嘌呤基、吡喃基、吡嗪基、吡唑烷基、吡唑啉基、吡唑基、哒嗪基、吡啶并恶唑、吡啶并咪唑、吡啶并噻唑、吡啶基、吡咯烷基、吡咯啉基、2H-吡咯基、吡咯基、喹唑啉基、喹啉基、4H-喹嗪基、喹喔啉基、奎宁环基、四氢呋喃基、四氢异喹啉基、四氢喹啉基、四唑基,6H-1,2,5-噻二嗪基、1,2,3-噻二唑基、1,2,4-噻二唑基、1,2,5-噻二唑基、1,3,4-噻二唑基、噻蒽基、噻唑基、异噻唑基噻吩基、噻吩并恶唑基、噻吩并噻唑基、噻吩并咪唑基、噻吩基、三嗪基、1H-1,2,3-***基、2H-1,2,3-***基、1H-1,2,4-***基、4H-1,2,4-***基和呫吨基。还包括稠环和螺环化 合物。
除非另有规定,术语“烃基”或者其下位概念(比如烷基、烯基、炔基、芳基等等)本身或者作为另一取代基的一部分表示直链的、支链的或环状的烃原子团或其组合,可以是完全饱和的(如烷基)、单元或多元不饱和的(如烯基、炔基、芳基),可以是单取代或多取代的,可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基),可以包括二价或多价原子团,具有指定数量的碳原子(如C 1-C 12表示1至12个碳,C 1-12选自C 1、C 2、C 3、C 4、C 5、C 6、C 7、C 8、C 9、C 10、C 11和C 12;C 3-12选自C 3、C 4、C 5、C 6、C 7、C 8、C 9、C 10、C 11和C 12。)。“烃基”包括但不限于脂肪烃基和芳香烃基,所述脂肪烃基包括链状和环状,具体包括但不限于烷基、烯基、炔基,所述芳香烃基包括但不限于6-12元的芳香烃基,例如苯、萘等。在一些实施例中,术语“烃基”表示直链的或支链的原子团或它们的组合,可以是完全饱和的、单元或多元不饱和的,可以包括二价和多价原子团。饱和烃原子团的实例包括但不限于甲基、乙基、正丙基、异丙基、正丁基、叔丁基、异丁基、仲丁基、异丁基、环己基、(环己基)甲基、环丙基甲基,以及正戊基、正己基、正庚基、正辛基等原子团的同系物或异构体。不饱和烃基具有一个或多个双键或三键,其实例包括但不限于乙烯基、2-丙烯基、丁烯基、巴豆基、2-异戊烯基、2-(丁二烯基)、2,4-戊二烯基、3-(1,4-戊二烯基)、乙炔基、1-和3-丙炔基,3-丁炔基,以及更高级的同系物和异构体。
除非另有规定,术语“杂烃基”或者其下位概念(比如杂烷基、杂烯基、杂炔基、杂芳基等等)本身或者与另一术语联合表示稳定的直链的、支链的或环状的烃原子团或其组合,有一定数目的碳原子和至少一个杂原子组成。在一些实施例中,术语“杂烷基”本身或者与另一术语联合表示稳定的直链的、支链的烃原子团或其组合物,有一定数目的碳原子和至少一个杂原子组成。在一个典型实施例中,杂原子选自B、O、N和S,其中氮和硫原子任选地被氧化,氮杂原子任选地被季铵化。杂原子或杂原子团可以位于杂烃基的任何内部位置,包括该烃基附着于分子其余部分的位置,但术语“烷氧基”、“烷氨基”和“烷硫基”(或硫代烷氧基)属于惯用表达,是指分别通过一个氧原子、氨基或硫原子连接到分子的其余部分的那些烷基基团。实例包括但不限于-CH 2-CH 2-O-CH 3、-CH 2-CH 2-NH-CH 3、-CH 2-CH 2-N(CH 3)-CH 3、-CH 2-S-CH 2-CH 3、-CH 2-CH 2、-S(O)-CH 3、-CH 2-CH 2-S(O) 2-CH 3、-CH=CH-O-CH 3、-CH 2-CH=N-OCH 3和–CH=CH-N(CH 3)-CH 3。至多两个杂原子可以是连续的,例如-CH 2-NH-OCH 3
除非另有规定,术语“环烃基”、“杂环烃基”或者其下位概念(比如芳基、杂芳基、环烷基、杂环烷基、环烯基、杂环烯基、环炔基、杂环炔基等等)本身或与其他术语联合分别表示环化的“烃基”、“杂烃基”。此外,就杂烃基或杂环烃基(比如杂烷基、杂环烷基)而言,杂原子可以占据该杂环附着于分子其余部分的位置。环烃基的实例包括但不限于环戊基、环己基、1-环己烯基、3-环己烯基、环庚基等。杂环基的非限制性实例包括1-(1,2,5,6-四氢吡啶基)、1-哌啶基、2-哌啶基,3-哌啶基、4-吗啉基、3-吗啉基、四氢呋喃-2-基、四氢呋喃吲哚-3-基、四氢噻吩-2-基、四氢噻吩-3-基,1-哌嗪基和2-哌嗪基。
除非另有规定,术语“烷基”用于表示直链或支链的饱和烃基,可以是单取代(如-CH 2F)或多取代的(如-CF 3),可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。烷基的例子包括甲基(Me),乙基(Et),丙基(如,n-丙基和异丙基),丁基(如,n-丁基,异丁基,s-丁基,t-丁基),戊基(如,n-戊基,异戊基,新戊基)等。
除非另有规定,“烯基”指在链的任何位点上具有一个或多个碳碳双键的烷基,可以是单取代或多取代的,可以是一价、二价或者多价。烯基的例子包括乙烯基,丙烯基,丁烯基,戊烯基,己烯基,丁间二烯基,戊间二烯基,己间二烯基等。
除非另有规定,“炔基”指在链的任何位点上具有一个或多个碳碳三键的烷基,可以是单取代或多取代的,可以是一价、二价或者多价。炔基的例子包括乙炔基,丙炔基,丁炔基,戊炔基等。
除非另有规定,环烷基包括任何稳定的环状或多环烃基,任何碳原子都是饱和的,可以是单取代或多取代的,可以是一价、二价或者多价。这些环烷基的实例包括,但不限于,环丙基、降冰片烷基、[2.2.2]二环辛烷、[4.4.0]二环癸烷等。
除非另有规定,环烯基包括任何稳定的环状或多环烃基,该烃基在环的任何位点含有一个或多个不饱和的碳-碳双键,可以是单取代或多取代的,可以是一价、二价或者多价。这些环烯基的实例包括,但不限于,环戊烯基、环己烯基等。
除非另有规定,环炔基包括任何稳定的环状或多环烃基,该烃基在环的任何位点含有一个或多个碳-碳三键,可以是单取代或多取代的,可以是一价、二价或者多价。
除非另有规定,术语“卤代素”或“卤素”本身或作为另一取代基的一部分表示氟、氯、溴或碘原子。此外,术语“卤代烷基”意在包括单卤代烷基和多卤代烷基。例如,术语“卤代(C 1-C 4)烷基”意在包括但不仅限于三氟甲基、2,2,2-三氟乙基、4-氯丁基和3-溴丙基等等。除非另有规定,卤代烷基的实例包括但不仅限于:三氟甲基、三氯甲基、五氟乙基,和五氯乙基。
“烷氧基”代表通过氧桥连接的具有特定数目碳原子的上述烷基,除非另有规定,C 1-6烷氧基包括C 1、C 2、C 3、C 4、C 5和C 6的烷氧基。烷氧基的例子包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、仲丁氧基、叔丁氧基、正戊氧基和S-戊氧基。
除非另有规定,术语“芳基”表示多不饱和的芳族烃取代基,可以是单取代或多取代的,可以是一价、二价或者多价,它可以是单环或多环(比如1至3个环;其中至少一个环是芳族的),它们稠合在一起或共价连接。术语“杂芳基”是指含有一至四个杂原子的芳基(或环)。在一个示范性实例中,杂原子选自B、N、O和S,其中氮和硫原子任选地被氧化,氮原子任选地被季铵化。杂芳基可通过杂原子连接到分子的其余部分。芳基或杂芳基的非限制性实施例包括苯基、萘基、联苯基、吡咯基、吡唑基、咪唑基、吡嗪基、恶唑基、苯基-恶唑基、异恶唑基、噻唑基、呋喃基、噻吩基、吡啶基、嘧啶基、苯并噻唑基、嘌呤基、苯并咪唑基、吲哚基、异喹啉基、喹喔啉基、喹啉基、1-萘基、2-萘基、4-联苯基、1-吡咯 基、2-吡咯基、3-吡咯基、3-吡唑基、2-咪唑基、4-咪唑基、吡嗪基、2-恶唑基、4-恶唑基、2-苯基-4-恶唑基、5-恶唑基、3-异恶唑基、4-异恶唑基、5-异恶唑基、2-噻唑基、4-噻唑基、5-噻唑基、2-呋喃基、3-呋喃基、2-噻吩基、3-噻吩基、2-吡啶基、3-吡啶基、4-吡啶基、2-嘧啶基、4-嘧啶基、5-苯并噻唑基、嘌呤基、2-苯并咪唑基、5-吲哚基、1-异喹啉基、5-异喹啉基、2-喹喔啉基、5-喹喔啉基、3-喹啉基和6-喹啉基。上述任意一个芳基和杂芳基环系的取代基选自下文所述的可接受的取代基。
除非另有规定,芳基在与其他术语联合使用时(例如芳氧基、芳硫基、芳烷基)包括如上定义的芳基和杂芳基环。因此,术语“芳烷基”意在包括芳基附着于烷基的那些原子团(例如苄基、苯乙基、吡啶基甲基等),包括其中碳原子(如亚甲基)已经被例如氧原子代替的那些烷基,例如苯氧基甲基、2-吡啶氧甲基3-(1-萘氧基)丙基等。
术语“离去基团”是指可以被另一种官能团或原子通过取代反应(例如亲和取代反应)所取代的官能团或原子。例如,代表性的离去基团包括三氟甲磺酸酯;氯、溴、碘;磺酸酯基,如甲磺酸酯、甲苯磺酸酯、对溴苯磺酸酯、对甲苯磺酸酯等;酰氧基,如乙酰氧基、三氟乙酰氧基等等。
术语“保护基”包括但不限于“氨基保护基”、“羟基保护基”或“巯基保护基”。术语“氨基保护基”是指适合用于阻止氨基氮位上副反应的保护基团。代表性的氨基保护基包括但不限于:甲酰基;酰基,例如链烷酰基(如乙酰基、三氯乙酰基或三氟乙酰基);烷氧基羰基,如叔丁氧基羰基(Boc);芳基甲氧羰基,如苄氧羰基(Cbz)和9-芴甲氧羰基(Fmoc);芳基甲基,如苄基(Bn)、三苯甲基(Tr)、1,1-二-(4'-甲氧基苯基)甲基;甲硅烷基,如三甲基甲硅烷基(TMS)和叔丁基二甲基甲硅烷基(TBS)等等。术语“羟基保护基”是指适合用于阻止羟基副反应的保护基。代表性羟基保护基包括但不限于:烷基,如甲基、乙基和叔丁基;酰基,例如链烷酰基(如乙酰基);芳基甲基,如苄基(Bn),对甲氧基苄基(PMB)、9-芴基甲基(Fm)和二苯基甲基(二苯甲基,DPM);甲硅烷基,如三甲基甲硅烷基(TMS)和叔丁基二甲基甲硅烷基(TBS)等等。
本发明的化合物可以通过本领域技术人员所熟知的多种合成方法来制备,包括下面列举的具体实施方式、其与其他化学合成方法的结合所形成的实施方式以及本领域技术上人员所熟知的等同替换方式,优选的实施方式包括但不限于本发明的实施例。
本发明所使用的溶剂可经市售获得。本发明采用下述缩略词:aq代表水;HATU代表O-(7-氮杂苯并***-1-基)-N,N,N',N'-四甲基脲六氟磷酸盐;EDC代表N-(3-二甲基氨基丙基)-N'-乙基碳二亚胺盐酸盐;m-CPBA代表3-氯过氧苯甲酸;eq代表当量、等量;CDI代表羰基二咪唑;DCM代表二氯甲烷;PE代表石油醚;DIAD代表偶氮二羧酸二异丙酯;DMF代表N,N-二甲基甲酰胺;DMSO代表二甲亚砜;EtOAc代表乙酸乙酯;EtOH代表乙醇;MeOH代表甲醇;CBz代表苄氧羰基,是一种胺保护基团;BOC代表叔丁基羰基是一种胺保护基团;HOAc代表乙酸;NaCNBH 3代表氰基硼氢化钠;r.t.代表室温;O/N代表过夜;THF代表四氢呋喃;Boc 2O代表二-叔丁基二碳酸酯;TFA代表三氟乙酸; DIPEA代表二异丙基乙基胺;SOCl 2代表氯化亚砜;CS 2代表二硫化碳;TsOH代表对甲苯磺酸;NFSI代表N-氟-N-(苯磺酰基)苯磺酰胺;NCS代表1-氯吡咯烷-2,5-二酮;n-Bu 4NF代表氟化四丁基铵;iPrOH代表2-丙醇;mp代表熔点;LDA代表二异丙基胺基锂;EDCI代表碳化二亚胺;HOBt代表1-羟基苯并***;Pd(dppf)Cl 2代表[1,1'-双(二苯基膦)二茂铁]二氯化钯。
化合物经手工或者
Figure PCTCN2018082119-appb-000219
软件命名,市售化合物采用供应商目录名称。
具体实施方式
下面通过实施例对本发明进行详细描述,但并不意味着对本发明任何不利限制。本文已经详细地描述了本发明,其中也公开了其具体实施例方式,对本领域的技术人员而言,在不脱离本发明精神和范围的情况下针对本发明具体实施方式进行各种变化和改进将是显而易见的。
实施例A-1:
Figure PCTCN2018082119-appb-000220
合成化合物A-1以(A-1-1)为起始原料,详细合成路线1,如下图:
Figure PCTCN2018082119-appb-000221
第一步(化合物2a的合成)
将化合物联二嚬哪醇硼酸酯(25.61g,100.85mmol,0.65eq),(1,5)-环辛二烯甲氧基铱二聚体(308.56mg,465.48μmol,0.003eq)和4,4-二叔丁基-2,2联吡啶(249.88mg,930.96μmol,0.006eq),溶于正己烷(250mL)中,反应液在氮气保护下50℃下搅拌至反应液成深红色。向上述溶液中加入化合物2a-1,然后反应液在氮气保护下50℃下搅拌3小时。LC-MS显示全部转化为化合物2a硼酸酯水解的产物。将反应液减压浓缩,得到粗品化合物2a,无需纯化,直接用于下一步。
相关表征数据如下:LCMS m/z:206.1[M+H](显示硼酸酯水解为硼酸).
1H NMR(400MHz,CDCl 3):δ7.83(s,1H),7.70(s,1H),1.37(s,12H).
第二步(化合物A-1-2的合成)
将化合物A-1-1(5g,30.49mmol,1.00eq)溶于无水二氧六环(80mL)和水(15mL)混合溶剂中,加入苯硼酸1a(2.97g,24.39mmol,0.80eq)和碳酸氢钠(5.12g,60.98mmol,2.37mL,2.00eq),四三苯基磷钯(1.76g,1.52mmol,0.05eq)。反应液在100℃下搅拌12小时在氮气保护下。当LC-MS显示反应完全,将反应液减压浓缩后加水100mL,用乙酸乙酯(50mL x 3)萃取,有机层用饱和食盐水溶液洗涤,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,层析柱(石油醚:乙酸乙酯:二氯甲烷=20:1:1-5:1:1),得到化合物A-1-2。
相关表征数据如下:LCMS m/z:206.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.12-8.14(m,2H),7.53-7.57(m,3H),7.29(s,1H),7.23(brs,2H).
第三步(化合物A-1-3的合成)
将化合物A-1-2(3.00g,14.59mmol,1.00eq)溶于乙腈(80.00mL),再加入NBS(5.19g,29.18mmol,2.00eq)反应液在80℃下搅拌2小时。当LC-MS显示反应完全,将反应液减压浓缩后加水100mL,用乙酸乙酯(50mL x 3)萃取,有机层用饱和食盐水溶液洗涤,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,层析柱(石油醚:乙酸乙酯=30:1-15:1),得到化合物A-1-3。
相关表征数据如下:LCMS m/z:283.9[M+H].
1H NMR(400MHz,DMSO-d 6):δ7.62-7.67(m,2H),7.52-7.54(m,3H),7.43(brs,2H)
第四步(化合物A-1-4的合成)
将化合物A-1-3(2.18g,7.66mmol,1.00eq)溶于水(40.00mL),再加入水合肼(1.92g,38.31mmol,1.86mL,5.00eq)反应液在100℃下搅拌12小时。当LC-MS显示反应完全,将反应液减压浓缩,得到粗品化合物A-1-4,直接用于下一步。
相关表征数据如下:LCMS m/z:280.0[M+H].
第五步(化合物A-1-5的合成)
将化合物A-1-4(4.00g,14.28mmol,1.00eq)溶于甲酸(50.00mL),反应液在100℃下搅拌12小时。当LC-MS显示反应完全,将反应液减压浓缩,得到粗品化合物A-1-5,直接用于下一步。
相关表征数据如下:LCMS m/z:309.9[M+H].
第六步(化合物A-1-6的合成)
将化合物A-1-5(4.00g,12.98mmol,1.00eq)溶于BSTA(30.00mL),反应液在100℃下搅拌12小时。当LC-MS显示反应完全,将反应液减压浓缩后加水50mL,用乙酸乙酯(50mL x 3)萃取,有机层用饱和食盐水溶液洗涤,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,层析柱(石油醚:乙酸乙酯=20:1-2:1),得到化合物A-1-6。
相关表征数据如下:LCMS m/z:289.9[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.61(s,1H),7.76-7.74(m,2H),7.51-7.43(m,5H)
第七步(化合物A-1的合成)
将化合物A-1-6(500.00mg,1.72mmol,1.00eq),化合物2a(602.43mg,2.10mmol,1.22eq)溶于四氢呋喃(3mL)中,将K 3PO 4(2M,1.77mL,2.05eq)和XPHOS-PD-G2(297.73mg,378.40μmol,0.22eq)加入到此溶液中,反应氛围置换为氮气,反应液在70℃下搅拌12小时。LCMS监测大量原料剩余,微量产物被检测到。反应液浓缩旋干,剩余物用1,4-二氧六环(5.00mL)稀释,向此混合物中加入Pd(dppf)Cl 2.CH 2Cl 2(294.97mg,361.20μmol,0.21eq)和K 2CO 3(2M,1.81mL,2.10eq),反应氛围置换为氮气,反应液在120℃微波环境下搅拌2小时。LCMS监测大量原料剩余,产物被检测到。反应液直接浓缩旋干,剩余物使用过柱机纯化得到粗产品,粗产品使用制备级高效液相色谱分离提纯,得到目标产物A-1。
相关表征数据如下:LCMS m/z:371.3[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.56(s,1H),8.40-8.30(brs,2H),7.54(s,1H),7.43(s,1H),7.37-7.33(m,5H),2.46(s,3H).
表1实施例化合物的制备可以参照前述制备实施例A-1的路线1类似的步骤方法进行,不同之处在于在步骤2中使用起始原料以下表中的硼酸代替原料1a得相应化合物
表1
Figure PCTCN2018082119-appb-000222
Figure PCTCN2018082119-appb-000223
合成化合物A-1以(A-1-1a)为起始原料,详细合成路线2,如下图:
Figure PCTCN2018082119-appb-000224
第一步(化合物A-1-2a的合成)
将化合物A-1-1a(40g,205.05mmol,1.00eq)溶于无水四氢呋喃(800mL),在0℃下缓慢滴加水合肼(24.15g,410.10mmol,23.45mL,2.00eq),0℃搅拌一小时1小时。然后升温到20℃反应14小时。LC-MS显示原料反应完全后,将反应液减压浓缩。粗品中加入500mL水,搅拌30分钟后过滤,用水洗涤得到化合物A-1-2a。
相关表征数据如下:LCMS m/z:190.4[M+H].
第二步(化合物A-1-3a的合成)
在25℃下将化合物A-1-2a(41.00g,195.70mmol,1.00eq),原甲酸三乙酯溶于冰醋酸(800mL),将混合物搅拌10min后升温到120℃搅拌16小时。LC-MS显示原料反应完全后,冷却到室温,将反应液在60℃下减压浓缩。得到的粗品,加入800mL水搅拌10分钟。用碳酸氢钠将pH调到7-8,用乙酸乙酯萃取用无水硫酸钠对有机相进行干燥,并对有有机相减压浓缩得到化合物A-1-3a。
相关表征数据如下:LCMS m/z:200.4[M+H].
第三步(化合物A-1-4a的合成)
将化合物A-1-3a(38.00g,180.86mmol,1.00eq),氢氧化钠(0.25M,217.03mL,0.30eq)溶于无水四氢呋喃(450mL)中,在25℃下搅拌16小时。LC-MS显示原料反应完全后,将反应液过滤并用四氢呋喃(50mL*4)洗涤,将滤渣干燥得到化合物A-1-4a。
相关表征数据如下:LCMS m/z:170.3[M+H].
第四步(化合物A-1-5a的合成)
在25℃下将化合物A-1-4a(5.50g,31.60mmol,1.00eq)溶于三氯氧磷(165.00g,1.08mol,100.00mL,34.05eq)中,向反应液中慢慢加入N,N-二甲基苯胺(382.95mg,3.16mmol,398.91μL,0.10eq)。反应液升温至110℃并搅拌16小时。LC-MS显示原料反应完全后,将反应液在50℃下减压浓缩。向得到的粗品中慢慢加入水(100mL)并搅拌20分钟。水相通过乙酸乙酯(50mL*4)萃取,合并有机相,依次用饱和碳酸氢钠水溶液(50mL*3)和饱和氯化钠水溶液(50mL*2)洗涤,再通过无水硫酸钠干燥过滤,减压浓缩得到化合物A-1-5a。
相关表征数据如下:LCMS m/z:188.3[M+H].
第五步(化合物A-1-6a的合成)
将化合物A-1-5a(3.05g,16.14mmol,1.00eq)溶解到二氧六环(50mL)中,在0℃下向反应液中通入氨气20分钟。将反应液倒入100mL的闷罐中,升温至90℃搅拌12个小时。LC-MS显示化合物1-5完全消耗,66%为目标产物。将反应液减压浓缩得到目标产物A-1-6a。
相关表征数据如下:LCMS m/z:169.4[M].
第六步(化合物A-1-7a的合成)
将化合物A-1-6a(3.58g,15.20mmol,1.00eq),苯硼酸1a(2.22g,18.24mmol,1.20eq),碳酸钾(4.20g,30.40mmol,2.00eq)溶于二氧六环(80mL)和水(16mL)中,向反应液中加入Pd(dppf)Cl 2.CH 2Cl 2(1.24g,1.52mmol,0.10eq)。反应液用氮气置换5次并在90℃氮气保护下搅拌12小时。LC-MS显示原料消耗完全,将反应液在50℃下减压浓缩。粗品通过正相硅胶柱纯化(PE:EA=10:1-0:1)得化合物A-1-7a。
相关表征数据如下:LCMS m/z:211.5[M+H].
第七步(化合物A-1-8a的合成)
在25℃下将化合物A-1-7a(2.80g,11.27mmol,1.00eq)溶于乙腈(50.00mL)中,向反应液中慢慢加入N-碘代丁二酰亚胺(5.07g,22.54mmol,2.00eq)。反应液升温至90℃并搅拌12小时。LC-MS显示原料反应完全后,将反应液在50℃下减压浓缩。向得到的粗品中加入硫代硫酸钠(100mL)并搅拌5分钟。水相通过乙酸乙酯(100mL*8)萃取,合并有机相,并用饱和氯化钠水溶液(50mL*2)洗涤,再通过无水硫酸钠干燥过滤,减压浓缩得到化合物A-1-8a。
相关表征数据如下:LCMS m/z:337.6[M+H].
第八步(实施例A-1的合成)
将化合物A-1-8a(2.00g,5.52mmol,1.00eq),2a(2.64g,8.28mmol,1.50eq),碳酸钾(1.53g,11.04mmol,2.00eq)溶于二氧六环(60mL)和水(12mL)中,向反应液中加入Pd(dppf)Cl 2.CH 2Cl 2(450.79mg,552.00μmol,0.10eq)。反应液用氮气置换5次并在90℃氮气保护下搅拌16小时。LC-MS显示原料消耗完全,将反应液在50℃下减压浓缩。粗品通过正相硅胶柱(PE:EA=10:1-0:1)及pre-HPLC纯化得到目标化合物A-1。
相关表征数据如下:LCMS m/z:370.7[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.55(s,1H),8.36-8.29(m,2H),7.53(s,1H),7.43(s,1H),7.38-7.31(m,5H),2.45(s,3H).
实施例A-6:
Figure PCTCN2018082119-appb-000225
合成化合物实施例A-6以(A-1-6a)为起始原料,详细合成路线,如下图:
Figure PCTCN2018082119-appb-000226
第一步(化合物A-6-1的合成)
在25℃下将化合物A-1-6a(4.00g,22.30mmol,1.00eq),NIS(10.04g,44.60mmol,2.00eq)溶于MeCN(40mL),将反应液升温到90℃,搅拌14小时。LC-MS显示原料反应完全后,冷却到室温,将反应液在50℃下减压浓缩得到化合物A-6-1。
相关表征数据如下:LCMS m/z:295.8[M+H].
1H NMR(400MHz,DMSO-d 6):δ11.05(brs,2H),8.48(s,1H)
第二步(化合物A-6-2的合成)
在25℃下将化合物A-6-1(2.00g,6.23mmol,1.00eq),2a(2.38g,7.48mmol,1.20eq),磷酸钾(2.64g,12.46mmol,2.00eq),DTBPF(PdCl 2)(406.04mg,623.00μmol,0.10eq)溶于二氧六环/水(30mL/6mL),将混合物用氮气换气三次后升温到110℃,搅拌14小时。LC-MS显示原料反应完全后,冷却到室温,将反应液在50℃下减压浓缩,粗品通过柱层析方法分离(100-200目硅胶,石油醚/乙酸乙酯=5:1,1:1)纯化后,中性p-HPLC机分(流动相:水/乙腈)得到化合物A-6-2。
A-6-2相关表征数据如下:LCMS m/z:328.9[M+H].
第三步(化合物A-6的合成)
在25℃下将化合物A-6-2(40.00mg,121.70μmol,1.00eq),1f(27.74mg,182.55μmol,1.50eq),碳酸钾(33.64mg,243.40μmol,2.00eq),Pd(dppf)Cl 2.CH 2Cl 2(9.94mg,12.17μmol,0.10eq)溶于二氧六环/水(3mL/0.6mL),将混合物用氮气换气三次后升温到100℃,搅拌14小时。LC-MS显示原料反应完全后,冷却到室温,将反应液在50℃下减压浓缩,粗品通过柱层析方法分离(100-200目硅胶,石油醚/乙酸乙酯=5:1,1:1)纯化后送中性机分(流动相:水/乙腈)得到化合物A-6。
相关表征数据如下:LCMS m/z:401.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.52(s,1H),7.54(s,1H),7.47(s,1H),7.29(d,J=8.8Hz,2H),6.89(d,J=8.8Hz,2H),6.05(br s,2H),3.75(s,3H),2.48(s,3H).
实施例A-7:
Figure PCTCN2018082119-appb-000227
在25℃下将化合物A-6-2(40.00mg,121.70μmol,1.00eq),A-7-1(145.46mg,304.25μmol,2.50eq),LiCl(15.48mg,365.10μmol,7.48uL,3.00eq),Pd(dppf)Cl 2.CH 2Cl 2(19.88mg,24.34μmol,0.20eq)溶于二氧六环(4.00mL),将混合物用氮气换气三次后升温到125℃,搅拌48小时。LC-MS显示原料反应完全后,冷却到室温,将反应液在50℃下减压浓缩,粗品通过柱层析方法分离(100-200目硅胶,石油醚/乙酸乙酯=5:1,1:1)纯化后送中性机分(流动相:水/乙腈)得到化合物A-7。
相关表征数据如下:LCMS m/z:372.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.59(s,1H),8.41(br s,2H),8.35(d,J=4.0Hz,1H),7.92(dt,J=1.6,8.0Hz,1H),7.80(d,J=8.0Hz,1H),7.49(s,1H),7.39(ddd,J=1.2,4.8,7.6Hz,1H),7.33(s,1H),2.47(s,3H).
实施例A-8:
Figure PCTCN2018082119-appb-000228
实施例A-8:本实施例化合物的制备可以参照前述制备实施例A-7的路线类似的步骤方法进行,不同之处在于在步骤2中使用起始原料A-8-1代替原料A-7-1,粗品通过板分离纯化后送中性机分(流动相:水/乙腈)得到A-8。
相关表征数据如下:LCMS m/z:378.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.53(s,1H),7.87(d,J=3.2Hz,1H),7.73(d,J=3.2Hz,1H),7.67(s,1H),7.62(s,1H),2.58(s,3H).
表2中实施例化合物的制备可以参照前述制备实施例A-6的路线类似的步骤方法进行,不同之处在于在步骤3中使用起始原料以下表中硼酸代替原料1a
表2
Figure PCTCN2018082119-appb-000229
Figure PCTCN2018082119-appb-000230
Figure PCTCN2018082119-appb-000231
表3中实施例化合物的制备可以参照前述制备实施例路线1类似的步骤方法进行,不同之处在于在步骤6和8中使用起始原料以下表中的硼酸代替原料1a和2a得相应化合物
表3
Figure PCTCN2018082119-appb-000232
Figure PCTCN2018082119-appb-000233
Figure PCTCN2018082119-appb-000234
表4中实施例化合物的制备可以参照前述制备实施例A-1路线1的类似方法进行,不同之处在于在步骤7中使用起始原料以下表中的硼酸代替原料2a得相应化合物
表4
Figure PCTCN2018082119-appb-000235
Figure PCTCN2018082119-appb-000236
实施例B-4:
Figure PCTCN2018082119-appb-000237
将化合物B-3(117.00mg,212.85μmol,1.00eq)溶于无水二氯甲烷(5mL)中,在0℃氮气保护下慢慢加入三溴化硼(533.24mg,2.13mmol,205.09μL,10.00eq)。反应液在0℃下搅拌2小时然后升到20℃搅拌10小时。当LC-MS显示反应完全,向反应液中加入甲醇10mL并搅拌10分钟,用三乙胺将反应液pH值调到8。将反应液在40℃下减压浓缩,用制备级高效液相色谱分离提纯得到产物B-4。
相关表征数据如下:LCMS m/z:338.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.48(s,1H),8.02(s,2H),7.35-7.38(m,2H),7.28-7.31(m,4H),6.91-6.94(d,J=8.4Hz,1H),6.84-6.86(d,J=8.4Hz,1H).
实施例B-7:
Figure PCTCN2018082119-appb-000238
第一步(化合物2g的合成)
将化合物2g-1(500.00mg,2.54mmol,1.00eq)和联二嚬哪醇硼酸酯(1.10g,4.32mmol,1.70eq)溶于无水二氧六环(10mL)中,加入醋酸钾和Pd(dppf)Cl 2(55.76mg,76.20μmol,0.03eq)。反应液在氮气保护下90℃下搅拌16小时。LC-MS显示15%是化合物2g-1,51%是化合物2g。向反应液中加入化合物联二嚬哪醇硼酸酯(645.01mg,2.54mmol,1.00eq)。反应液在氮气保护下100℃下搅拌2小时。LC-MS显示9%是化合物2g-1,62%是化合物2g。将反应液减压浓缩。得到的粗品通过正相硅胶柱(PE:EA=1:0-5:1)纯化得到化合物2g。
相关表征数据如下:LCMS m/z:245.2[M+H].
第二步(化合物B-7的合成)
实施例B-7:本实施例化合物的制备可以参照前述制备施例A-1路线1的方法进行,不同之处在于在步骤7中使用起始原料硼酸2g代替原料2a。产物经过正相硅胶柱(PE:EA=10:1-0:1)以及制备级高效液相色谱分离提纯得到目标产物B-7。
相关表征数据如下:LCMS m/z:328.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ13.06(brs,1H),8.47(s,1H),8.02-7.99(m,3H),7.73(s,1H),7.43-7.15(m,7H).
实施例B-10:
Figure PCTCN2018082119-appb-000239
第一步(化合物2j-2的合成)
二异丙基胺(635.57mg,6.28mmol,882.74uL,1.10eq)溶于THF(20.00mL),于-78℃下滴加入n-BuLi(2.5M,2.51mL,1.10eq),反应液于-78℃下反应1小时,然后滴加入化合物2j-1(1.00g,5.71mmol,628.93μL,1.00eq)的THF(10.00mL)溶液,于-78℃下反应1小时后加入三氟乙酸乙酯(893.07mg,6.28mmol,867.06uL,1.10eq)的THF(10.00mL)溶液,继续于0℃下反应2小时。TLC检测化合物 2j-1消耗完全,生成一个极性较大的点。0℃下加入20mL氯化铵水溶液淬灭反应,乙酸乙酯萃取(20mL*2),合并有机层,饱和食盐水(40mL)洗涤,无水硫酸钠干燥,过滤,蒸干溶剂。粗品用正相硅胶柱(PE:EA=1:0-3:1)纯化得到化合物2j-2,产率:42.0%。
相关表征数据如下: 1H NMR(400MHz,CHLOROFORM-d):δ7.91(dd,J=2.4,6.0Hz,1H),7.71(ddd,J=2.4,4.4,8.8Hz,1H),7.07(dd,J=8.8,10.2Hz,1H)
第二步(化合物2j-3的合成)
化合物2j-2(5.00g,18.45mmol,1.00eq)溶于正丁醇(70.00mL),加入水合肼(18.47g,369.00mmol,17.93mL,20.00eq),反应液于120℃反应6小时。反应完成后,反应液冷却至室温,加入100mL水,乙酸乙酯萃取(100mL*3),合并有机层,饱和食盐水洗涤(100mL),无水硫酸钠干燥,过滤,蒸干溶剂。粗品用正相硅胶柱(PE:EA=1:0-2:3)纯化得到化合物2j-3。
相关表征数据如下: 1H NMR(400MHz,CHLOROFORM-d):δ10.28-10.89(m,1H),8.04(s,1H),7.60(dd,J=1.6,8.8Hz,1H),7.47(d,J=8.8Hz,1H)
第三步(化合物2j的合成)
化合物2j-3(630.00mg,2.38mmol,1.00eq),联硼酸频那醇酯(1.51g,5.95mmol,2.50eq),醋酸钾(700.72mg,7.14mmol,3.00eq),Pd(dppf)Cl 2.CH 2Cl 2(97.18mg,119.00μmol,0.05eq)混于二氧六环(30.00mL),氮气置换3次,反应液于90℃反应16小时。TLC显示反应完全后,加入30mL水淬灭反应,乙酸乙酯萃取(30mL*3),合并有机层,饱和水盐水(50mL)洗涤,无水硫酸钠干燥,过滤,蒸干溶剂。粗品用正相硅胶柱(PE:EA=1:0-4:1)纯化得到化合物2j。
相关表征数据如下: 1H NMR(400MHz,CHLOROFORM-d):δ10.97(br s,1H),8.31(s,1H),7.83(d,J=8.8Hz,1H),7.45-7.51(m,1H),7.19(s,1H),1.31(s,12H).
第四步(化合物B-10的合成)
实施例B-10:本实施例化合物的制备可以参照前述制施例A-1路线2的方法进行,不同之处在于在步骤8中使用起始原料硼酸2j代替原料2a。粗品经过纯化得到目标产物B-10。
相关表征数据如下:LCMS m/z:396.2[M+H].
1H NMR(400MHz,DMSO-d 6):δ14.00(br s,1H),8.51(s,1H),8.08(br s,2H),7.76(s,1H),7.58(d,J=8.78Hz,1H),7.34(br d,J=8.78Hz,3H),7.20-7.27(m,3H).
表5中实施例化合物的制备可以参照前述制备施例A-1路线2的方法进行,不同之处在于在步骤8中使用起始原料以下表中的硼酸代替原料2a得相应化合物
表5
Figure PCTCN2018082119-appb-000240
Figure PCTCN2018082119-appb-000241
实施例B-13:
Figure PCTCN2018082119-appb-000242
第一步(化合物2m-2的合成)
本化合物的制备可以参照前述制备施例B-10方法进行,不同之处在于在步骤3中使用起始原料硼酸2m-1代替原料,粗品通过硅胶色谱柱纯化后(填充料:100-200目硅胶粉,流动相为石油醚/乙酸乙酯=50/1,10/1)得到化合物2m。
相关表征数据如下:LCMS m/z:289.0[M+H].
第二步(化合物2m的合成)
在25℃下将化合物2m-2(600.00mg,1.12mmol,1.00eq),苄基溴(287.33mg,1.68mmol,199.53uL,1.50 eq),碳酸钾(309.59mg,2.24mmol,2.00eq),溶于乙腈(8mL),升温到90℃搅拌10小时。LC-MS显示原料反应完全后,冷却到室温,将反应液在50℃下减压浓缩,粗品通过硅胶色谱柱纯化后(填充料:100-200目硅胶粉,流动相为石油醚/乙酸乙酯=20/1,10/1)得到2m。
相关表征数据如下:LCMS m/z:379.0[M+H].
第三步(化合物B-13-1的合成)
B-13-1的制备可以参照前述制备施例A-1路线2的方法进行,不同之处在于在步骤8中使用起始原料硼酸2m代替原料2a。粗品通过制备TLC大板(石油醚/乙酸乙酯=1:1)纯化得到化合物B-13-1。
相关表征数据如下:LCMS m/z:461.4[M+H].
第四步(化合物B-13的合成)
在25℃下将化合物B-13(50.00mg,78.02μmol,1.00eq),Pd/C(5.00mg,7.80μmol,10%纯度),溶于四氢呋喃/甲醇(6mL/8mL),将混合物用氢气换气三次后,在氢气保护下25℃下搅拌20小时。LC-MS显示原料反应完全后,将反应液在50℃下减压浓缩,粗品通过硅胶色谱柱纯化后(填充料:100-200目硅胶粉,流动相为石油醚/乙酸乙酯=5/1,1/1)得到粗品,将粗品送中性Pre-HPLC机分(流动相:水/乙腈)得到B-13。
相关表征数据如下:LCMS m/z:372.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ10.52(br s,1H),8.49(s,1H),8.03(br s,2H),7.43(d,J=2.0Hz,1H),7.36-7.24(m,6H),6.89(d,J=8.4Hz,1H)
实施例B-14:
Figure PCTCN2018082119-appb-000243
第一步(化合物2n-2的合成)
在25℃下将化合物2n-1(2.00g,11.42mmol,1.00eq)溶于乙腈(55.00mL)中,向反应液中慢慢加入N-溴代丁二酰亚胺(2.03g,11.42mmol,1.00eq)。反应液升温至25℃并搅拌1小时。LC-MS显示原料反应 完全后,将反应液在50℃下减压浓缩。向得到的粗品中加入饱和的食盐水。水相通过乙酸乙酯(50mL*3)萃取,合并有机相,减压浓缩,粗品通过正相硅胶柱(PE:EA=1:0)纯化得到目标化合物2n-2。
相关表征数据如下:LCMS m/z:253.9[M+H].
1H NMR(400MHz,CDCl 3):δ7.44(s,1H),7.32(s,1H),4.25(brs,1H),2.18(s,3H).
第二步(化合物2n-3的合成)
在25℃下将化合物2n-2(2.00g,7.87mmol,1.00eq)和乙酸(7.09g,118.05mmol,6.75mL,15.00eq)溶于甲苯(40.00mL)中,向反应液中加入乙酸钾(6.18g,62.96mmol,8.00eq)。反应液25℃搅拌10分钟有大量沉淀生成。反应液用乙酸(6mL)稀释,在将亚硝酸异戊脂(1.01g,8.66mmol,1.17mL,1.10eq)逐滴加入反应液中,反应液在25℃下搅拌3小时。LC-MS显示原料反应完全后,将反应液在加入10mL水中稀释,在用乙酸乙酯和饱和的碳酸氢钠萃取,有机相在用食盐水洗2次,合并有机相用无水硫酸钠干燥,减压浓缩,粗品通过正相硅胶柱(PE:EA=20:1到10:1)纯化得到目标化合物2n-3。
相关表征数据如下:LCMS m/z:264.9[M+H].
第三步(化合物2n的合成)
本化合物的制备可以参照前述制备施例B-10方法进行,不同之处在于在步骤3中使用起始原料硼酸2n-3代替原料,粗品通过硅胶色谱柱纯化后(填充料:100-200目硅胶粉,流动相为石油醚/乙酸乙酯=1/0,10/1)得到化合物2n。
相关表征数据如下:LCMS m/z:313.1[M+H].
1H NMR(400MHz,CDCl 3):δ10.4(brs,1H),8.46(s,1H),8.18(s,1H),8.08(s,1H),1.38(s,12H).
第四步(化合物B-14的合成)
实施例B-14:本实施例化合物的制备可以参照前述制备实施例A-1路线2的方法进行,不同之处在于在步骤8中使用起始原料硼酸2n代替原料2a。得到化合物B-14。
相关表征数据如下:LCMS m/z:396.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ13.65(s,1H),8.51(s,1H),8.25(m,1H),8.11(br s,2H),8.07(s,1H),7.53(s,1H),7.38-7.31(m,2H),7.29-7.21(m,3H).
实施例B-15:
Figure PCTCN2018082119-appb-000244
第一步(化合物2o的合成)
本化合物的制备可以参照前述制备施例B-10方法进行,不同之处在于在步骤3中使用起始原料硼酸2o-1代替原料,粗品通过硅胶色谱柱纯化后(填充料:100-200目硅胶粉,流动相为石油醚/乙酸乙酯=1/1)得到化合物2o。
相关表征数据如下: 1H NMR(400MHz,DMSO-d 6):δ7.50(s,2H),1.27(s,12H).
第二步(化合物B-15的合成)
实施例B-15:本实施例化合物的制备可以参照前述制备实施例A-1路线2的方法进行,不同之处在于在步骤8中使用起始原料硼酸2o代替原料2a。得到化合物B-15。
相关表征数据如下:LCMS m/z:372.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.48(s,1H),8.01(br s,2H),7.41-7.36(m,2H),7.34-7.27(m,3H),7.13(s,2H).
实施例B-16:
Figure PCTCN2018082119-appb-000245
第一步(化合物2p的合成)
本化合物的制备可以参照前述制备施例B-10方法进行,不同之处在于在步骤3中使用起始原料硼酸2p-1代替原料,粗品用正相硅胶柱纯化(PE:EA=1:0-10:1)纯化得到化合物2p。
相关表征数据如下: 1H NMR(400MHz,CHLOROFORM-d):δ8.95(dd,J=1.6,4.0Hz,1H),8.35(s,1H),8.20(dd,J=1.6,8.0Hz,1H),8.09(m,2H),7.42(dd,J=4.0,8.0Hz,1H),1.40(s,12H).
第二步(化合物B-16的合成)
实施例B-16:本实施例化合物的制备可以参照前述制备实施例A-1路线2的方法进行,不同之处在于在步骤8中使用起始原料硼酸2p代替原料2a。粗品通过pre-HPLC分离纯化得到化合物B-16。
相关表征数据如下:LCMS m/z:339.2[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.88(dd,J=1.6,4.0Hz,1H),8.52(s,1H),8.27(d,J=8.0Hz,1H),8.05-8.21(m,2H),8.01(d,J=1.6Hz,1H),7.87(d,J=8.4Hz,1H),7.48-7.55(m,2H),7.35-7.41(m,2H),7.20-7.28(m,3H).
实施例B-17:
Figure PCTCN2018082119-appb-000246
第一步(化合物2q-2的合成)
化合物2q-1(3.00g,22.52mmol,1.00eq)溶于乙腈(50.00mL),0℃下分批加入NBS(3.81g,21.39mmol,0.95eq),反应液于0℃反应3小时。0℃加入50mL水淬灭反应,乙酸乙酯萃取(50mL*3),合并有机层,饱和水盐水(50mL)洗涤,无水硫酸钠干燥,过滤,蒸干溶剂,粗品用正相硅胶柱分离(PE:EA=1:0-10:1)纯化得到化合物2q-2。
相关表征数据如下: 1H NMR(400MHz,CHLOROFORM-d):δ6.98-7.07(m,2H),6.34(d,J=8.0Hz,1H),3.25-3.31(m,2H),2.72(t,J=6.40Hz,2H),1.85-1.95(m,2H)
第二步(化合物2q的合成)
本化合物的制备可以参照前述制备施例B-10方法进行,不同之处在于在步骤3中使用起始原料硼酸2q-2代替原料,粗品用正相硅胶柱(PE:EA=1:0-10:1)纯化得到化合物2q。
相关表征数据如下: 1H NMR(400MHz,CHLOROFORM-d):δ7.40-7.46(m,2H),6.45(d,J=8.28Hz,1H),3.30-3.38(m,2H),2.78(t,J=6.27Hz,2H),1.89-1.98(m,2H),1.33(s,12H).
第三步(化合物B-17的合成)
实施例B-17:本实施例化合物的制备可以参照前述制备实施例A-1路线2的方法进行,不同之处在于在步骤8中使用起始原料硼酸2q代替原料2a。粗品通过pre-HPLC分离得纯化得到化合物B-17。
相关表征数据如下:LCMS m/z:343.2[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.44(s,1H),7.83(br s,2H),7.35-7.41(m,2H),7.24-7.30(m,3H),6.86(s,1H),6.63(d,J=8.4Hz,1H),6.27(d,J=8.4Hz,1H),5.69(s,1H),3.17(br d,J=4.8Hz,2H),2.54-2.59(m,2H),1.77(br d,J=4.8Hz,2H).
实施例B-19:
Figure PCTCN2018082119-appb-000247
第一步(化合物B-19-1的合成)
化合物B-18-1的制备可以参照前述制备施例A-1路线2的方法进行,不同之处在于在步骤8中使用起始原料硼酸2r代替原料2a。粗品通过柱层析方法分离(100-200目硅胶,石油醚/乙酸乙酯=5:1,1:1)纯化得到化合物B-19-1。
相关表征数据如下:LCMS m/z:377.1[M+H].
1H NMR(300MHz,DMSO-d 6):δ8.46(s,1H),7.94(br s,2H),7.58(br d,J=3.2Hz,2H),7.39(br dd,J=1.6,5.0Hz,3H),5.60(br s,1H),3.80(br s,2H),3.45(br s,2H),2.24(br s,2H),1.40(s,9H).
第二步(化合物B-19-2的合成)
在20℃下将化合物B-19-1(100.00mg,249.71μmol,1.00eq),Pd/C(20.00mg,10%纯度)溶于MeOH(15.00mL),然后将反应液在20℃,氢气压力为30Psi下搅拌36小时。LC-MS显示原料反应完全后,将反应液过滤后在50℃下减压浓缩得到化合物B-19-2。
相关表征数据如下:LCMS m/z:339.1[M+H].
第三步(化合物B-19的合成)
在20℃下将化合物B-19-2(74.00mg,166.58μmol,1.00eq),溶于EtOAc(2.00mL),然后将HCl/EtOAc(2.00mL,1.00eq)滴加到反应液当中,在20℃下搅拌1小时。LC-MS显示原料反应完全后,将反应液在50℃下减压浓缩后加水溶解,然后滴加2M的NaOH溶液(2mL),过滤得到化合物B-19。
相关表征数据如下:LCMS m/z:295.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.51(s,1H),7.75(br s,2H),7.54-7.38(m,5H),2.95(br d,J=10.8Hz,2H),2.73-2.67(m,2H),2.45-2.42(m,1H),2.37-2.24(m,2H),1.46(br d,J=11.6Hz,2H).
实施例B-22:
Figure PCTCN2018082119-appb-000248
第一步(化合物B-22-1的合成)
实施例B-22-1:本实施例化合物的制备可以参照前述制备施例A-1路线2的方法进行,不同之处在于在步骤8中使用起始原料硼酸2t代替原料2a。
第二步(化合物B-22的合成)
将化合物B-22-1(30.00mg,101.25μmol,1.00eq),溶于甲醇(10mL)中,向反应液中加入Pd/C(30.00mg,10%纯度)。反应液用氮气置换3次,在用氢气置换3次,并在30℃氢气压力30psi下搅拌12小时,LC-MS显示原料还有87%没有反应,用硅藻土过滤,将滤液液在50℃下减压浓缩,加入甲醇(10mL),向 反应液中加入Pd/C(30.00mg,10%纯度)。反应液用氮气置换3次,在用氢气置换3次,并在40℃氢气压力50psi下搅拌12小时,LC-MS显示原料消耗完全,用硅藻土过滤,将滤液液在50℃下减压浓缩,粗品通过prep HPLC纯化得到B-22。
相关表征数据如下:LCMS m/z:296.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.52(s,1H),7.79(br s,2H),7.53-7.42(m,5H),3.91-3.86(m,2H),3.20-3.17(m,2H),2.90-2.81(m,1H),2.71-2.58(m,2H),1.46(m,2H).
实施例B-25:
Figure PCTCN2018082119-appb-000249
在25℃下将化合物B-24(38.00mg,118.24μmol,1.00eq)和湿钯炭(20.00mg)溶于甲醇(10mL),将反应液用氮气换气三次后置于50psi的氢气环境下,并升温到40℃搅拌32小时。反应完成后反应液用硅藻土过滤,滤液旋干后粗品送中性机分(流动相:水/乙腈)得到B-25。
相关表征数据如下:LCMS m/z:324.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.52(s,1H),7.79(br s,2H),7.44-7.48(m,5H),3.30–3.25(m,2H),2.94–2.96(m,1H),2.17-2.23(m,2H),1.49-1.52(m,2H),1.08(d,J=6.0Hz,6H).
实施例B-23:
Figure PCTCN2018082119-appb-000250
第一步(化合物2u-2的合成)
在25℃下将化合物2u-1(1.00g,5.51mmol,1.00eq),环丙基硼酸2u-1a(709.96mg,8.27mmol,1.50eq),碳酸钾(1.52g,11.02mmol,2.00eq)溶于二氧六环(20mL)和水(4mL)中,在加入Pd(dppf)Cl 2.CH 2Cl 2(449.97mg,551.00μmol,0.10eq)。反应液升温至100℃并搅拌6小时。LC-MS显示原料反应完全后,将反应液用硅藻土过滤,在50℃下减压浓缩。得到目标化合物2u-2。
相关表征数据如下:LCMS m/z:188.0[M+H].
第二步(化合物2u的合成)
本化合物的制备可以参照前述制备实施例A-1路线1方法进行,不同之处在于在步骤1中使用起始原料硼酸2u-2代替原料,反应液浓缩的粗品2u,直接用于下一步。
第三步(化合物B-23的合成)
实施例B-23:本实施例化合物的制备可以参照前述制备实施例A-1路线2第8步的方法进行,不同之处在于在步骤8中使用起始原料硼酸2u代替原料2a。得到化合物B-23。
相关表征数据如下:LCMS m/z:397.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.56(s,1H),8.34(br s,2H),7.41(d,J=5.2Hz,2H),7.39-7.32(m,5H),2.10-2.04(m,1H),1.01-0.93(m,2H),0.79-0.72(m,2H).
实施例B-28:
Figure PCTCN2018082119-appb-000251
第一步(化合物2y的合成)
在氮气保护下将甲基叔丁基醚(10mL)加入化合物(1,5-环辛二烯)(甲氧基)铱(I)二聚体(54.69mg,82.50μmol,0.03eq),4,4'-二叔丁基-2,2'-联吡啶(44.29mg,165.00μmol,0.06eq)和频哪醇硼酸酯(698.33mg,2.75mmol,1.00eq)中,加完后的混合物在50℃下搅拌30分钟直到溶液变为暗红色,然后将化合物2y-1(500.00mg,2.75mmol,1.00eq)加入该混合物中,反应液在50℃继续搅拌12小时。反应完全后,反应液用硅藻土过滤,滤液浓缩得到粗品,粗品通过硅胶色谱柱纯化后(填充料:200-300目硅胶粉,流动相为石油醚/乙酸乙酯=100/0~10/1)得到化合物2y。
相关表征数据如下: 1H NMR(400MHz,CDCl 3-d)δppm 7.85(s,1H),7.79(s,1H),1.29(s,12H).
第二步(化合物B-28的合成)
实施例B-28:本实施例化合物的制备可以参照前述制备实施例A-1路线2第8步的方法进行,不同之处在于在步骤8中使用起始原料硼酸2y代替原料2a。得到化合物B-28。
相关表征数据如下:LCMS m/z:391.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.61(s,1H),7.79(s,1H),7.66(d,J=0.8Hz,1H),7.37-7.44(m,5H).表6中实施例化合物的制备可以参照前述制备实施例B-28路线方法进行,不同之处在于在步骤1中使用起始原料代替原料2y-1得相应化合物
表6
Figure PCTCN2018082119-appb-000252
Figure PCTCN2018082119-appb-000253
实施例B-32:
Figure PCTCN2018082119-appb-000254
第一步(化合物2ab-2的合成)
冰浴(0℃)下,化合物2u-1(1.00g,5.51mmol,1.00eq)和乙酰丙酮化铁(389.08mg,1.10mmol,0.20eq)加入到无水四氢呋喃(20.00mL)和N-甲基吡咯烷酮(2.00mL),然后将乙基溴化镁(3M,3.67mL,2.00eq)加入其中,这个混合物在0℃搅拌0.5个小时。LCMS跟踪显示反应完成,碳酸氢钠的水溶液(60mL)淬灭反应,然后用乙酸乙酯(60mL*3)萃取,有机相用饱和食盐水(40mL*2)洗,用无水硫酸钠干燥,过滤在减压下浓缩,残渣用柱层析纯化(硅胶,淋洗液为石油醚/乙酸乙酯=100:1到20:1)收集到目标化合物2ab-2。
相关表征数据如下:LCMS m/z:176.0[M+H].
第二步(化合物2ab的合成)
将联硼酸频哪醇酯(763.24mg,3.01mmol,1.04eq)溶于甲基叔丁基醚(10.00mL),然后加入二(1,5-环辛二烯)二-μ-甲氧基二铱(I)(57.47mg,86.70μmol,0.03eq)和4,4'-二叔丁基-2,2'-联吡啶(23.27mg,86.70μmol,0.03eq),这个混合物在70-80℃下搅拌15分钟,然后化合物2ab-2(507.00mg,2.89mmol,1.00eq)加入到这个反应体系中。这个混合物在70-80℃下搅拌了9个小时。LCMS跟踪显示反应完成,这个混合物在减压下浓缩。液体浓缩后用柱层析纯化(硅胶,淋洗液为石油醚/乙酸乙酯=10:1到1:1))收集到目标化合物2ab。
相关表征数据如下:LCMS m/z:220.1[M+H].
第三步(化合物B-32的合成)
实施例B-32:本实施例化合物的制备可以参照前述制备实施例A-1路线2第8步的方法进行,不同之处在于在步骤8中使用起始原料硼酸2ab代替原料2a。得到化合物B-32。
相关表征数据如下:LCMS m/z:385.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.58(s,1H),8.38(br s,2H),7.64(s,1H),7.47-7.18(m,6H),2.68(q,J=7.6Hz,2H),1.05(t,J=7.6Hz,3H)
实施例B-34:
Figure PCTCN2018082119-appb-000255
实施例B-34:本实施例化合物的制备可以参照前述制备实施例B-32路线方法进行,不同之处在于在步骤1中使用起始原料异丙基溴化镁代替原料2u-1b。得到化合物B-34。
实施例B-33:
Figure PCTCN2018082119-appb-000256
第一步(化合物2ac-2和2ac-2-a的合成)
将混合物2ac-1(250.00mg,1.26mmol,1.00eq),3,4-二氢-2H-吡喃(108.33mg,1.29mmol,117.75μL,1.02eq)和DDQ(28.66mg,126.25μmol,0.10eq)的乙腈(10.00mL)溶液在20℃下搅拌16小时。反应完全后,反应液浓缩得到粗品,粗品通过硅胶色谱层析板纯化后(层析液为石油醚/乙酸乙酯=3/1)得到混合物2ac-2和2ac-2-a。
第二步(化合物2ac和2ac-a的合成)
在25℃下将混合物2ac-2和2ac-2-1(160.00mg,567.09μmol,1.00eq),频哪醇硼酸酯(158.41mg,623.80μmol,1.10eq),Pd(dppf)Cl 2(82.99mg,113.42μmol,0.20eq),醋酸钾(166.96mg,1.70mmol,3.00eq)溶于1,4-二氧六环(5mL),将反应液用氮气换气三次后升温到90℃,并在氮气保护下继续搅拌16小时。反应完全后,冷却到25℃,将反应液用硅藻土过滤,滤液浓缩后粗品通过硅胶色谱层析板纯化后(层析液为石油醚/乙酸乙酯=1/1)得到混合物2ac和2ac-a。
第三步(化合物B-33-1和B-33-1a的合成)
实施例B-23:本实施例化合物的制备可以参照前述制备实施例A-1路线2第8步的方法进行,不同之处在于在步骤8中使用起始原料硼酸2ac和2ac-a代替原料2a。得到化合物B-33和B-33-1a。
相关表征数据如下:LCMS m/z:413.1[M+H].
第四步(化合物B-33的合成)
在25℃下向化合物B-33和B-33-1a(50.00mg,121.23μmol,1.00eq)的甲醇(4mL)溶液中一次加入氯化氢甲醇溶液(4M,2.00mL,65.99eq),并升温到70℃搅拌3小时。反应完成后反应液旋干后,粗品溶于甲醇(5mL)并用碳酸氢钠固体调节pH=8~9后送中性机分(流动相:水/乙腈)得到B-33。
相关表征数据如下:LCMS m/z:329.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.50(s,1H),8.12(brs,2H),7.76-7.84(m,2H),7.35(m,2H),7.24(m, 4H).
实施例C-1:
Figure PCTCN2018082119-appb-000257
合成化合物C-1以(C-1-1)为起始原料,详细合成路线如下:
Figure PCTCN2018082119-appb-000258
第一步(化合物C-1-2的合成)
将化合物C-1-1(20.00g,121.96mmol,1.00eq.)溶解于甲醇(1.40L),并向溶液中滴加水合肼(14.37g,243.92mmol,13.95mL,85%纯度,2.00eq),室温下搅拌20小时。反应结束后旋蒸除去大部分溶剂,过滤得到化合物C-1-2粗品。
相关表征数据如下:LCMS m/z:159.8[M+H]
第二步(化合物C-1-3的合成)
0℃下,将三氟乙酸酐3a(4.74g,22.56mmol,3.14mL,1.2eq.)滴入化合物C-1-2(3.00g,18.80mmol,1.00eq)的DMF(20.00mL)的溶液,在0℃搅拌4小时。溶剂减压旋蒸至固体析出,过滤得到化合物C-1-3粗品。
相关表征数据如下:LCMS m/z:255.9[M+H]
第三步(化合物C-1-4的合成)
将化合物C-1-3(1.09g,7.43mmol,1.00eq),溶于N,O-双三甲硅基乙酰胺(20mL),80℃搅拌15小时。反应结束后将反应液缓慢滴加到甲醇(50mL),再将将甲醇溶液浓缩得到固体,柱分离得到化合物4。
相关表征数据如下:LCMS m/z:237.9[M+H]
1H NMR(400MHz,DMSO-d 6):δ7.23(s,1H),8.72(br s,2H)
第四步(化合物C-1-5的合成)
将化合物C-1-4(100mg,421μmol,1.00eq),苯硼酸1a(62mg,505μmol,1.20eq),Pd(dppf)Cl 2.CH 2Cl 2(34mg,42μmol,0.10eq)和碳酸钾(116mg,842μmol,2.00eq)溶于1,4-二氧六环(5mL)和水(1mL)的混合溶剂中,脱气氮气置换三次,然后在氮气保护下,在90℃下搅拌2小时。反应完成后,冷却至室温,溶剂减压旋干,剩余固体加水(20mL)稀释,乙酸乙酯萃取(300mL*3),合并有机相,无水硫酸钠干燥,过滤,旋干,柱分离,得化合物C-1-5。
相关表征数据如下:LCMS m/z:279.9[M+H]
1H NMR(400MHz,DMSO-d 6):δ8.33(brs,2H),8.17-8.15(m,2H),7.71(s,1H),7.54-7.47(m,3H).
第五步(化合物C-1-6的合成)
将化合物C-1-5(90mg,322.3μmol,1.00eq),NIS(145.0mg,664.6μmol,2eq)溶于乙腈(4mL),80℃下搅拌2小时。冷却至室温,减压除去溶剂,剩余固体加硫代硫酸钠水溶液(20mL)稀释,乙酸乙酯萃取(30mL*3),合并有机相,无水硫酸钠干燥,过滤,旋干,得到粗品C-1-6。
相关表征数据如下:LCMS m/z:405.9[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.42(brs,2H),7.61-7.59(m,2H),7.51-7.41(m,3H)
第六步(化合物C-1的合成)
将化合物C-1-6(126.0mg,311μmol,1.00eq),化合物2a(267.9mg,933μmol,3eq),Pd(dppf)Cl 2.CH 2Cl 2(45mg,55μmol,0.18eq)和碳酸钠(107mg,777μmol,2.50eq)溶于1,4-二氧六环(4mL)和水(1mL)的混合溶剂中,脱气氮气置换三次,然后在氮气保护下,90℃下搅拌16小时。冷却至室温,减压除去溶剂,剩余固体用甲醇(20mL)稀释,抽滤,滤液旋干得到固体。制备液相色谱分离得化合物C-1。
相关表征数据如下:LCMS m/z:439.3[M+H]
1H NMR(400MHz,DMSO-d 6):δ7.53(s,1H),7.38-7.32(m,6H),2.48(s,3H).
表7中实施例化合物的制备可以参照前述制备实施例C-1路线类似方法进行,不同之处在于在步骤2,4和6使用起始原料以下表中的原料代替得相应化合物
表7
Figure PCTCN2018082119-appb-000259
Figure PCTCN2018082119-appb-000260
Figure PCTCN2018082119-appb-000261
Figure PCTCN2018082119-appb-000262
Figure PCTCN2018082119-appb-000263
Figure PCTCN2018082119-appb-000264
Figure PCTCN2018082119-appb-000265
Figure PCTCN2018082119-appb-000266
Figure PCTCN2018082119-appb-000267
Figure PCTCN2018082119-appb-000268
实施例C-12:
Figure PCTCN2018082119-appb-000269
第一步(化合物C-12-1的合成)
向化合物3j(5.0g,30.09mmol,4.31mL,1eq)和N,N-二异丙基乙胺(4.67g,36.11mmol,6.29mL,1.2eq)的DMF(80mL)分批加入CDI(5.85g,36.11mmol,1.2eq),混合物在15℃下搅拌1小时后向其中加入4-氯-6-肼基-嘧啶-2-胺C-1-2(4.80g,30.09mmol,1eq),反应液在15℃搅拌60小时,倒入冰水中(140mL),搅拌15分钟,用乙酸乙酯(75mL)萃取三次,萃取液用水(40mL)洗两次,盐水(50mL)洗一次,硫酸钠干燥,过滤,浓缩得粗品C-12-1。
相关表征数据如下:LCMS m/z:308.2[M+H]
第二步(化合物C-12-2的合成)
将化合物C-12-1(10.00g,25.67mmol,1eq)和N,O-二(三甲基硅)乙酰胺(60.00mL)的悬浊液加热到120℃搅拌2小时。冷却,剧烈搅拌下将反应液缓慢倒入冷的甲醇(200mL)中,控制温度在5-10℃,将该澄清的混合液浓缩,向残余物加入乙酸乙酯(30mL)打浆2次,滤液浓缩,经硅胶柱(石油醚:乙酸乙酯3:1-2:1)纯化得C-12-2。
第三步(化合物C-12-3的合成)
化合物C-12-2(1.35g,4.66mmol,1.0eq)和苯硼酸(852.29mg,6.99mmol,1.5eq)溶解在40毫升二氧六环和8毫升水中,氮气置换三遍后依次加入碳酸钾(643.99mg,4.66mmol,1.0eq),催化剂Pd(dppf)Cl 2(380.53mg,520.05ummol,0.112eq),然后氮气再置换三遍后升温到80-90℃搅拌3小时。LCMS显示反应完成且检测到目标化合物。反应降到室温后浓缩干,然后加入乙酸乙酯大约200毫升稀释,然后用水洗涤4次,每次50毫升。有机物浓缩后经硅胶柱(200-300目硅胶,石油醚/乙酸乙酯=2:1)纯化后,得到产物C-12-3。
相关表征数据如下:LCMS m/z:332.0[M+H]
1HNMR(400MHz,DMSO-d 6)δ8.12(t,J=5.6Hz,1H),8.01(m,2H),7.52(s,1H),7.45-7.48(m,3H),7.30-7.37(m,6H),4.71(s,2H),4.64(s,2H).
第四步(化合物C-12-4的合成)
化合物C-12-3(800mg,2.41mmol,1.0eq)溶解在20毫升乙腈中,然后加入碘代丁二酰亚胺(814.72mg,3.62mmol,1.5eq),反应混合物加热到100℃搅拌4小时。薄层色谱分析(石油醚/乙酸乙酯=1:1,Rf=0.36)显示反应原料消失,新点生成。溶剂乙腈减压浓缩干后,用100毫升乙酸乙酯稀释,然后好用20毫升饱和亚硫酸钠水溶液洗涤两次,20毫升水洗涤两次,20毫升饱和食盐水洗涤两次,无水硫酸钠干燥后过滤浓缩得到粗产品C-12-4,直接用于下一步反应。
第五步(C-12的合成)
将化合物C-12-4(1.10g,2.41mmol,1.0eq),2,6-二甲基苯硼酸(363.84mg,2.41mmol,1.0eq)和碳酸钾(999.23mg,7.23mmol,3eq)溶解在20毫升二氧六环和4毫升水中,氮气置换3次加入催化剂Pd(dppf)Cl 2(176.34mg,241.00ummol,0.10eq),然后加热升温到80-90℃并搅拌16小时,薄层色谱(石油醚/乙酸乙酯=1:1)跟踪显示反应完成,有机溶剂减压浓缩干后加入150毫升乙酸乙酯,然后好用20毫升水,20毫升包和实验室分别依次洗涤两次,然后浓缩干。残余物用硅胶柱(200-300目,石油醚/乙酸乙酯=1:1)纯化得到粗品,进一步用制备色谱纯化得到纯品C-12。
相关表征数据如下:LCMS m/z:437.1[M+H]
1HNMR(400MHz,CD 3OD):δ7.42-7.30(m,10H),7.03(s,2H),4.79(s,2H),4.70(s,2H),2.41(s,6H).
表8中实施例化合物的制备可以参照前述制备实施例C-12路线类似方法进行,不同之处在于在步骤1,3和5使用起始原料以下表中的原料代替得相应化合物
表8
Figure PCTCN2018082119-appb-000270
Figure PCTCN2018082119-appb-000271
Figure PCTCN2018082119-appb-000272
实施例C-18:
Figure PCTCN2018082119-appb-000273
第一步(化合物C-18-1的合成)
将化合物C-1-2(2.00g,12.53mmol,1.00eq)和二异丙基乙基胺(2.43g,18.79mmol,3.28mL,1.50eq)的二氯甲烷(40.00mL)悬浊液冷却至-40℃,向其中缓慢滴加草酰氯单乙酯(1.63g,11.90mmol,1.33mL,0.95eq)的二氯甲烷(5.00mL)溶液,控制反应液温度在-20℃至-40℃,1小时滴加完毕。反应液缓慢升温至15℃(室温)并搅拌15小时。过滤,固体用二氯甲烷(3mL)洗涤两次,干燥得C-18-1直 接用于下一步反应。
相关表征数据如下:LCMS m/z:260.0[M+H].
第二步(化合物C-18-2的合成)
将化合物C-18-1(8.00g,30.81mmol,1.00eq)和N,O-二(三甲基硅)乙酰胺(80.00mL)的悬浊液加热到120℃搅拌16小时。冷却,剧烈搅拌下将反应液缓慢倒入0℃的甲醇(100mL)中,搅拌15分钟,将该澄清的混合液浓缩,向残余物加入乙酸乙酯(100mL)搅拌30分钟,过滤,滤液浓缩,经硅胶柱(石油醚:乙酸乙酯3:1-1:1)纯化得C-18-2。
第三步(化合物C-18-3的合成)
将化合物C-18-2(2.00g,8.28mmol,1.00eq),苯硼酸(1.11g,9.11mmol,1.10eq),Pd(dppf)Cl 2DCM络合物(338.09mg,414.00μmol,0.05eq)和碳酸钾(2.29g,16.56mmol,2.00eq)加入1,4-二氧六环(20mL)和水(4mL)中,氮气置换数次后加热到100℃搅拌14小时。冷却到室温后过滤,固体用乙酸乙酯(10mL)洗涤后用水(40mL)打浆,过滤,真空干燥得到灰色固体C-18-3。
相关表征数据如下:LCMS m/z:256.2[M+H].
HNMR(400MHz,DMSO-d 6):δ8.12(s,2H),7.87(s,2H),7.49-7.42(s,4H).
第四步(化合物C-18-4的合成)
室温(15-25℃)下,往化合物C-18-3(200mg,0.783mmol,1.0eq)和二异丙基乙基胺(0.205mL,1.18mmol)的DMF(5毫升)悬浮物中分批加入HATU(327.75mg,0.862mmol),反应混合物在10-25度下搅拌10分钟,然后再加入2,4-二氟苄胺(123.37mg,0.862mmol),反应混合物在15-25度下搅拌16小时,LCMS显示得到了目标产品,反应混合物倒入水中,析出固体,固体收集后悬浮在100毫升乙酸乙酯中然后再过滤,滤液浓缩之后用制备剥层色谱纯化(乙酸乙酯/石油醚=1:1)得到化合物C-18-4。
相关表征数据如下:LCMS m/z:381.0[M+H].
第五步(化合物C-18-5的合成)
化合物C-18-4(36mg,94.65μmol)溶解在3毫升乙腈中,氮气保护后加入NIS(25.55mg,113.58ummol),在15-25℃下搅拌10分钟后升温到100℃搅拌16小时。LCMS暗示反应完成并监测到目标化合物的MS,反应溶液浓缩掉溶剂后得到粗品C-18-5。
相关表征数据如下:LCMS m/z:506.9[M+H].
第六步(化合物C-18的合成)
化合物C-18-5(47.92mg,94.65μmol,1.00eq),2,6-二甲基苯硼酸(14.29mg,94.65μmol,1.00eq)溶解在2.5毫升二氧六环和0.5毫升水中,然后一次性加入碳酸钾(39.24mg,283.95μmol,3.00eq)和催化剂二苯基二茂铁氯化钯二氯甲烷络合物(15.46mg,18.93μmol,0.2eq),然后氮气置换三次,缓慢 加热到100℃并搅拌16小时,LCMS显示反应完全,目标化合物的MS被检测到,反应混合物直接减压浓缩,残余物首先用制备剥层色谱纯化(二氯甲烷/甲醇=10:1)纯化,然后再用制备剥层色谱(乙酸乙酯/石油醚=2:1)得到一个粗品,制备高效液相色谱纯化得到目标化合物C-18。
相关表征数据如下:LCMS m/z:486.0[M+H].
1HNMR(400MHz,DMSO-d 6):δ9.16(t,J=6.0Hz,1H),8.32(brs,1H),7.06-7.40(m,8H),6.95(s,1H),4.51(d,J=6.0Hz,2H),2.33(s,3H),2.08(s,3H)。
实施例C-19:
Figure PCTCN2018082119-appb-000274
C12(1.7g,3.89mmol)溶解在20毫升三氟乙酸中,然后升温到70℃,反应1小时,LCMS显示脱苄干净。多余的三氟乙酸通过减压旋蒸除去,剩余物加甲醇(4mL)稀释,NaHCO3调pH值到7.0,过滤滤液浓缩得粗产品,反相制备得到纯品C-19。
相关表征数据如下:LCMS m/z:347.2[M+H].
1HNMR(400MHz,DMSO-d 6):δ8.12(brs,2H),8.36-7.29(m,5H),6.92(s,2H),5.54-5.51(t,J=6.4Hz,1H),4.64-4.62(d,J=6.4Hz,,2H),2.32(s,6H).
表9中实施例化合物的制备可以参照前述制备实施例C-19路线方法进行
表9
Figure PCTCN2018082119-appb-000275
Figure PCTCN2018082119-appb-000276
Figure PCTCN2018082119-appb-000277
Figure PCTCN2018082119-appb-000278
实施例C-20:
Figure PCTCN2018082119-appb-000279
第一步(化合物C-20-1的合成)
在20℃下,向化合物C-19(270mg,779.48μmol,1eq)的DCM(5mL)溶液加入DMP(95.91mg,1.17mmol,361.98μL,1.5eq),反应液在20℃搅拌12小时。加水(10mL),过滤水洗,滤饼减压浓缩得C-20-1粗品。
相关表征数据如下:LCMS m/z:345.0[M+H]
第二步(化合物C-20的合成)
向化合物C-20-1(100mg,290.39μmol,1eq)的MeOH(3.00mL)溶液加环丙基胺(24.87mg,435.58μmol,30.18μL,1.5eq),在20℃搅拌半小时。然后加入NaBH 3CN(36.50mg,580.77μmol,2eq),反应液在20℃搅拌反应1.5个小时。向反应液中加水(20mL),乙酸乙酯萃取(20mL*2),有机相饱和食盐水洗涤,无水硫酸钠干燥,浓缩,浓缩,经制备硅胶(DCM/MeOH=10/1)和制备色谱柱纯化得C-20.
表征数据如下:LCMS m/z:386.1[M+H]
1H NMR(400MHz,CDCl 3):δ7.37-7.27(m,5H),6.94(s,2H),6.03(brs,2H),4.15(s,2H),2.46(s,6H),2.32-2.28(m,1H),0.52-0.48(m,4H)
实施例C-56:
Figure PCTCN2018082119-appb-000280
第一步(化合物C-56-1的合成)
室温下,向化合物C-55(150mg,433.04μmol,1.0eq)的二氯甲烷溶液中加入戴斯-马丁过碘烷(275.51mg,649.56μmol,201.10μL,1.5eq),在氮气保护下,反应三个小时。混合物过滤得到滤液,滤饼用甲醇(10mL*3)洗涤,合并滤液并浓缩得到化合物C-56-1。
相关表征数据如下:LCMS m/z:345.0[M+H].
第二步(化合物C-56的合成)
在室温下,将化合物C-56-1(50mg,145.19μmol,1eq)溶解于水(3mL)、乙醇(3mL)和四氢呋喃(3mL)中,向混合物中加入羟胺盐酸盐(15.13mg,217.79μmol,1.5eq)和醋酸钠(23.82mg,290.39μmol,2.0eq),混合物加热至80℃,在80℃下搅拌两个小时。混合物浓缩得到的粗品用反相制备(柱子:Phenomenex Gemini 150*25mm*10μm;流动相:[A:纯净水(10mM NH 4HCO 3),B:ACN];B%:18%-48%,3min)纯化后得C-56。
相关表征数据如下:LCMS m/z:360.1[M+H].
1H NMR(400MHz,METHANOL-d 4):δ8.24(s,1H),7.40-7.27(m,5H),7.02(m,2H),2.40(s,6H)。
实施例C-60和C-61:
Figure PCTCN2018082119-appb-000281
第一步(化合物C-60-1的合成)
将化合物C-33(200mg,499.55μmol,1eq)溶于无水二氯甲烷(5mL)后冷至0℃,向该溶液中一次加入氯化亚砜(297.16mg,2.50mmol,181.19μL,5eq),该混合物在20℃下搅拌2小时。反应完成后反应液浓缩,剩余物用二氯甲烷(10mL)打浆,形成的悬浊液过滤,滤饼用二氯甲烷(10mL)洗涤后干燥,得到化合物C-60-1。
相关表征数据如下:LCMS(Ms+1):419.0.
第二步(化合物C-60的合成)
将化合物C-60-1(42.92mg,487.10μmol,1.2eq)溶于DMF(5mL)后冷至0℃,向该溶液中一次加入NaH(24.36mg,608.88μmol,60%纯度,1.5eq),该混合物在20℃下搅拌0.5小时后向其中加入化合物3-S-羟基-四氢呋喃(170mg,405.92μmol,1eq),此反应液在20℃下搅拌16小时。反应完成后反应液用饱和NH 4Cl水溶液(50mL)淬灭,然后用乙酸乙酯(20mL)稀释并用乙酸乙酯(20mL)萃取两次,合并的有机相用饱和食盐水(50mL)洗,再用无水Na 2SO 4干燥,过滤后旋干,得到的粗品送中性积分(流动相:水/乙腈)得到C-60和C-61。
C-60相关表征数据如下:LCMS m/z:471.2[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.34(br s,2H),7.54(s,1H),7.41(s,1H),7.33-7.39(m,5H),4.69(s,2H),4.36(br d,J=3.2Hz,1H),3.72-3.77(m,2H),3.66-3.69(m,2H),2.47(s,3H),1.95-2.00(m,2H).C-61相关表征数据如下:LCMS m/z:428.2[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.30(br s,2H),7.54(s,1H),7.41(s,1H),7.32-7.39(m,5H),3.67(s,2H),2.47(s,3H),2.25(s,6H).
实施例C-62:
Figure PCTCN2018082119-appb-000282
在室温下,向三氯氧磷(5mL)中加入化合物C-56,体系加热至70℃,在70℃下搅拌2小时。混合物浓缩,加入甲醇(3mL),用固体碳酸氢钠调pH至7,混合物过滤得到滤液,滤液用反相制备(柱子:Phenomenex Gemini 150*25mm*10μm;流动相:[A:纯净水(10mM NH 4HCO 3),B:ACN];B%:20%-50%,3min)纯化分离得到化合物C-62。
相关表征数据如下:LCMS m/z:342.1[M+H].
1H NMR(400MHz,METHANOL-d 4):δ7.42~7.40(m,2H),7.32~7.28(m,2H),7.02(s,2H),2.40(s,6H)。
实施例C-73:
Figure PCTCN2018082119-appb-000283
实施例C-73:本实施例化合物的制备可以参照前述制备施例C-62的方法进行,得到化合物C-73。
相关表征数据如下:LCMS m/z:380.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.73(br s,2H),7.40-7.43(m,2H),7.23(t,J=8.8Hz,2H),7.15(d,J=5.2Hz,2H),2.40(s,3H).
实施例C-74:
Figure PCTCN2018082119-appb-000284
实施例C-74:本实施例化合物的制备可以参照前述制备施例C-62的方法进行,得到化合物C-74。
相关表征数据如下:LCMS m/z:400.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.58(br s,2H),7.65(d,J=2.0Hz,2H),6.77(d,J=3.2Hz,1H),6.25(d,J=2.8Hz,1H),2.63(s,3H),2.05(s,3H).
实施例C-65
Figure PCTCN2018082119-appb-000285
第一步(化合物C65-2的合成)
向化合物C-65-1(3.8g,14.66mmol,1eq)的二氯甲烷((60mL)溶液加入氯化亚砜(10.46g,87.95mmol,6.38mL,6eq),反应液在氮气50℃搅拌5小时。反应液过滤,二氯甲烷洗涤,滤饼减压浓缩干得C-65-2。
相关表征数据如下:LCMS m/z:278.0[M+H]
第二步(化合物C-65-3的合成)
向化合物C-65-2(700mg,2.52mmol,1eq)的二氧六环((10mL)溶液加入二甲胺(3.44g,25.21mmol,33%,10eq),反应液在30mL闷罐中60℃搅拌12小时。反应液减压浓缩干,加MeOH(20mL),H 2O(20mL)打浆得C-65-3。
相关表征数据如下:LCMS m/z:287.1[M+H]
第三步(化合物C-65-4的合成)
向化合物C-65-3(600mg,2.10mmol,1eq)的ACN(8mL)溶液中加入NIS(942.97mg,4.19mmol,2eq),反应液在,80℃搅拌12小时。反应液加饱和Na 2SO 3(20mL),H2O(10mL),用EA(20mL*2)萃取,有机相饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得化合物C-65-4。
相关表征数据如下:LCMS m/z:413.0[M+H]
第四步(化合物C-65的合成)
向化合物C-65-4(250mg,606.50μmol,1eq)加1,4-二氧六环(5mL)和H 2O(1mL)溶液加入Pd(dppf)Cl 2(44.38mg,60.65μmol,0.1eq),K 2CO 3(167.65mg,1.21mmol,2eq),C-65-4a(184.52mg,727.79μmol,1.2eq),氮气置换三次,在90℃搅拌2小时。浓缩,经硅胶柱(PE/EA=5/1to PE/EA/EtOH=2/3/1)和制备硅胶板(DCM/MeOH=10/1)纯化,再制备色谱柱纯化得C-65。
相关表征数据如下:LCMS m/z:412.1[M+H]
1H NMR(400MHz,DMSO-d 6):δ8.26(br s,2H),7.40-7.37(m,2H),7.20-7.12(m,4H),3.64(s,2H),2.36(s,3H),2.24(s,6H).
实施例C-68
Figure PCTCN2018082119-appb-000286
第一步(化合物C-68-1的合成)
乙二醇单甲醚(89.38mg,1.17mmol,92.62μL,2.5eq)溶于四氢呋喃(3mL),然后加入钠氢(46.98mg,1.17mmol,60%纯度,2.5eq),这个混合物在20℃下搅拌2个小时。然后加入化合物C-65-2(0.13g,469.83μmol,1eq),这个混合物在70-80℃下搅拌5个小时。LCMS跟踪显示有目标化合物的MS生成,反应液在15℃下用10mL水淬灭,混合物在减压水泵下除去溶剂四氢呋喃,然后每次用10mL乙酸乙酯萃取3次,合并有机相然后用饱和食盐水(10mL)洗涤一次,无水硫酸钠干燥,过滤,在减压下浓缩有机相得到残渣。收集到目标化合物C-68-1。
相关表征数据如下:LCMS m/z:318.0[M+H].
第二步(化合物C-68-2的合成)
化合物C-68-1(0.12g,378.17μmol,1eq)溶于乙腈(5mL)然后加入N-碘代丁二酰亚胺(170.16mg,756.34μmol,2eq),反应液在70-80℃下搅拌5个小时。LCMS跟踪显示有目标化合物的MS生成,反应液在15℃下用20mL亚硫酸钠水溶液淬灭,然后每次用50mL乙酸乙酯萃取两次,合并有机相然后用饱和食盐水(10mL)洗涤一次,无水硫酸钠干燥,过滤,在减压下浓缩有机相得到残渣。收集到目标化合物C-68-2。
相关表征数据如下:LCMS m/z:443.9[M+H].
第三步(化合物C-68的合成)
往化合物C-68-2(0.3g,676.87μmol,1eq)中加入1,4-二氧六环(20mL)和水(5mL),然后加入2a(194.32mg,676.87μmol,1eq),磷酸钾(287.35mg,1.35mmol,2eq),1,1-双(二-叔-丁基膦)二茂铁二氯化钯(88.23mg,135.37μmol,0.2eq),这个混合物用氮气置换三次,并在氮气保护下在100℃下搅拌八个小时。LCMS显示有目标化合物的MS生成。溶剂减压浓缩干,残渣残留物用硅胶柱纯化(硅胶,淋洗液为石油醚/乙酸乙酯=3:1到1:1),然后用柱子:Phenomenex Gemini 150*25mm*10um;流动相:([A:纯净水(10mM NH 4HCO 3),B:ACN];B%:40%-70%,3min收集到目标化合物C-68。
相关表征数据如下:LCMS m/z:477.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.35(br s,2H),7.54(s,1H),7.44(s,1H),7.39(dd,J=5.7,8.7Hz,2H),7.18(t,J=8.8Hz,2H),4.70(s,2H),3.72-3.66(m,2H),3.50(dd,J=3.8,5.6Hz,2H),3.25(s,3H),2.48(s,3H).
表10中实施例化合物的制备可以参照前述制备实施例C-68路线方法进行
表10
Figure PCTCN2018082119-appb-000287
Figure PCTCN2018082119-appb-000288
实施例C-75:
Figure PCTCN2018082119-appb-000289
第一步(化合物C-75-1的合成)
将化合物C-18-2(500mg,2.07mmol,1eq)溶于无水二氯甲烷(20mL)中,然后在氮气保护下,向其中一次加入甲基溴化镁(3M,2.07mL,3eq),反应液在25℃下搅拌0.5小时。反应完成后,反应液用饱和氯化铵水溶液(50mL)淬灭,水相用二氯甲烷(100mL)萃取,有机相通过无水硫酸钠干燥,过滤,旋干,最终经硅胶板分离纯化(层析液为石油醚/乙酸乙酯=1/1),得C-75-1。
相关表征数据如下:LCMS m/z:227.9[M+H]
第二步(化合物C-75-2的合成)
将化合物C-75-1(110mg,483.20μmol,1eq),化合物1m(81.13mg,579.84μmol,1.2eq),1,1'-双(二-叔丁基膦基)二茂铁二氯化钯(62.98mg,96.64μmol,0.20eq)和磷酸钾(205.14mg,966.39μmol,2.00eq)溶于1,4-二氧六环(10mL)和水(1mL)的混合溶剂中,脱气氮气置换三次,然后在氮气保护下,在100℃下搅拌12小时。反应完成后,冷却至室温,溶液用硅藻土过滤,旋干,所得粗品经硅胶板分离纯化(层析液为石油醚/乙酸乙酯=1/1)得C-75-2。
相关表征数据如下:LCMS m/z:288.0[M+H].
第三步(化合物C-75-3的合成)
将化合物C-75-2(110mg,382.89μmol,1.00eq)和N-碘代丁二酰亚胺(172.28mg,765.77μmol,2eq) 溶于乙腈(5mL),80℃下搅拌3小时。冷却至室温,反应液用饱和亚硫酸钠水溶液(20mL)淬灭,乙酸乙酯萃取(20mL*2),合并有机相,饱和食盐水(30mL)洗,无水硫酸钠干燥,过滤,旋干得C-75-3。
相关表征数据如下:LCMS m/z:413.8[M+H].
第四步(化合物C-75的合成)
将化合物C-75-3(150mg,363.03μmol,1eq),化合物2a(125.06mg,435.64μmol,1.2eq),1,1'-双(二-叔丁基膦基)二茂铁二氯化钯(47.32mg,72.61μmol,0.20eq)和磷酸钾(154.12mg,726.06μmol,2eq)溶于1,4-二氧六环(5mL)和水(1mL)的混合溶剂中,脱气氮气置换三次,然后在氮气保护下,100℃下搅拌12小时。冷却至室温,反应液用硅藻土过滤,滤液旋干所得粗品用制备液相色谱分离得C-75。
相关表征数据如下:LCMS m/z:447.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.22(br s,2H),7.55(s,1H),7.46(s,1H),7.38(dd,J=1.2,8.8Hz,2H),7.18(t,J=8.8Hz,2H),5.32(s,1H),2.49(s,3H),1.59(s,6H).
实施例C-77:
Figure PCTCN2018082119-appb-000290
实施例C-77:本实施例化合物的制备可以参照前述制备施例C-75的方法进行,得到化合物C-77。
相关表征数据如下:LCMS m/z:467.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.37(br s,2H),7.83(s,1H),7.66(s,1H),7.43(dd,J=6.0,8.4Hz,2H),7.23(t,J=8.8Hz,2H),5.38(s,1H),1.60(s,6H).
实施例C-78:
Figure PCTCN2018082119-appb-000291
实施例C-78:本实施例化合物的制备可以参照前述制备施例C-75的方法进行,得到化合物C-78。
相关表征数据如下:LCMS m/z:413.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.16(br s,2H),7.41-7.34(m,2H),7.21-7.15(m,4H),5.35(s,1H),2.36(s,3H),1.58(s,6H).
实施例C-76:
Figure PCTCN2018082119-appb-000292
第一步(化合物C-76-1的合成)
在25℃下向化合物C-65-1(1.3g,5.01mmol,1eq)的二氯甲烷(50mL)悬浊液中加入戴斯-马丁过碘烷(3.19g,7.52mmol,2.33mL,1.5eq),该反应液在25℃下搅拌12小时。反应完成后,反应液用饱和碳酸氢钠水溶液(40mL)和饱和硫代硫酸钠水溶液(40mL)淬灭,并在室温下搅拌30分钟,该水溶液用乙酸乙酯(100mL)萃取三次,合并有机相用饱和氯化钠水溶液(200mL)洗涤,再通过无水硫酸钠干燥过滤,减压浓缩得到化合物C-76-1。
相关表征数据如下:LCMS(Ms+1):257.9.
1H NMR(400MHz,DMSO-d 6):δ10.13(s,1H),8.32(br s,2H),8.21-8.24(m,2H),7.68(s,1H),7.32-7.37(m,2H).
第二步(化合物C-76-2的合成)
向化合物C-76-1(1g,3.89mmol,1eq)的水(6mL)、乙醇(6mL)和四氢呋喃(6mL)的混合溶液中依次加入盐酸羟胺(405.24mg,5.83mmol,1.5eq)和醋酸钠(637.81mg,7.78mmol,2eq),反应液升温至80℃并搅拌2小时。原料反应完全后,将反应液减压浓缩,所得固体用水(20mL)洗涤三次,并在真空条件下干燥得到化合物C-76-2。
相关表征数据如下:LCMS(Ms+1):273.0.
第三步(化合物C-76-3的合成)
在25℃下将化合物C-76-2(0.65g,2.39mmol,1eq)溶于***(10mL)中,该反应液在70℃下搅拌2小时。将反应液浓缩,向剩余物中加入饱和碳酸氢钠水溶液调节反应液pH至7~8,所形成溶液用乙酸乙酯(50mL)萃取三次,合并有机相用饱和氯化钠水溶液(100mL)洗涤,再通过无水硫酸钠干燥过滤,减压浓缩得到化合物C-76-3。
相关表征数据如下:LCMS m/z:254.9[M+H].
第四步(化合物C-76-4的合成)
将化合物C-76-3(100mg,393.36μmol,1.00eq)和N-碘代丁二酰亚胺(176.99mg,786.71μmol,2eq)溶于乙腈(5mL)中,反应液升温至80℃并搅拌2小时。原料反应完全后,将反应液降至室温,并用饱和亚硫酸钠水溶液(20mL)淬灭,所得混合物通过乙酸乙酯(20mL*2)萃取,合并有机相,并用饱和氯化钠水溶液(30mL)洗涤,再通过无水硫酸钠干燥过滤,减压浓缩得到化合物C-76-4。
相关表征数据如下:LCMS m/z:380.6[M+H]
第五步(化合物C-76的合成)
在25℃下将化合物C-76-4(180mg,473.54μmol,1eq),6(163.13mg,568.24μmol,1.2eq),1,1'-双(二-叔丁基膦基)二茂铁二氯化钯(61.73mg,94.71μmol,0.20eq)和磷酸钾(201.04mg,947.07μmol,2.00eq)溶于二氧六环/水(10mL/2mL),将反应液用氮气换气三次后升温到100℃,并在氮气保护下继续搅拌12小时。反应完成后,反应液用硅藻土过滤,将滤液旋干后所得粗品送中性积分(流动相:水/乙腈)得化合物C-76。
相关表征数据如下:LCMS m/z:414.1[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.77(br s,2H),7.54(s,1H),7.38-7.41(m,3H),7.21(t,J=8.8Hz,2H),2.51(br s,3H).
实施例C-79:
Figure PCTCN2018082119-appb-000293
实施例C-79:本实施例化合物的制备可以参照前述制备施例C-77的方法进行,得到化合物C-79。
相关表征数据如下:LCMS m/z:434.0[M+H].
1H NMR(400MHz,DMSO-d 6):δ8.93(br s,2H),7.78(s,1H),7.62(s,1H),7.44(dd,J=5.6,8.8Hz,2H),7.25(t,J=8.8Hz,2H).
实施例C-80:
Figure PCTCN2018082119-appb-000294
第一步(化合物C-80的合成)
将化合物C-33(0.5g,1.25mmol,1eq)加入水(25mL),然后加入高锰酸钾(394.73mg,2.50mmol, 2eq)和氢氧化钠(99.90mg,2.50mmol,2eq)。此混合物在40℃下搅拌8个小时。LCMS和HPLC跟踪显示有目标化合物的MS生成,往反应液中加入1克亚硫酸钠固体淬灭反应,然后用2M盐酸将pH调到5-6。混合物用乙酸乙酯萃取3次(每次60mL),合并有机相然后用饱和食盐水(30mL)洗涤两次,无水硫酸钠干燥,过滤,在减压下浓缩有机相得到残渣,残渣残留物用制备分离纯化柱子:Phenomenex Gemini 150*25mm*10μm;流动相:[H 2O(10mM NH 4HCO 3)-ACN];B%:10%-40%,10min。收集到目标化合物C-80。
相关表征数据如下:LCMS m/z:415.2[M+H].
1H NMR(400MHz,METHANOL-d 4):δ7.72(s,1H),7.45-7.27(m,6H),2.56(s,3H).
本发明的化合物的药理活性通过下述检测A 2a受体活性的体外测定法来确定。
实验例1:体外活性测试实验
人类腺苷A 2a受体钙流检测实验
细胞来源:
A 2a稳定细胞株由上海药明康德构建,宿主细胞CHO。
检测试剂盒:
Fluo-4Direct试剂盒,(Invitrogen,货号F10471)。在试剂盒中的荧光检测试剂(与钙离子特异性结合并引起荧光信号的增加)与细胞孵育适当时间后,加入化合物刺激细胞引起胞内钙流发生变化,从而引起荧光信号的变化,可以反映出化合物的激动或抑制活性的强弱。
细胞培养基:
F12+10%胎牛血清+遗传霉素300ug/ml+杀稻瘟菌素2ug/ml
化合物稀释缓冲液:
Hanks平衡盐缓冲液(Invitrogen)+20mM HEPES,每次使用前配置
激动剂:
NECA(Sigma-E2387)
参考化合物(拮抗剂):
CGS-15943(Sigma-C199)
化合物稀释:
待测化合物用DMSO溶解配制成10mM母液。待测化合物用DMSO稀释成0.2mM,参考化合物CGS-15943用DMSO稀释成0.015mM。接着用ECHO进行10个点3倍连续稀释,转移900nl到化合物板(Greiner-781280)中,加入30ul化合物稀释缓冲液。待测化合物最终起始浓度为1uM,CGS-15943为0.075uM。
测定方法:
细胞准备:
取冻存的A 2A细胞,复苏之后用培养基重悬至1x10 6个/ml,20μl/孔种入384孔多聚赖氨酸包被细胞板(Greiner-781946),5%CO2,37℃培养箱孵育过夜。
将前一天准备好的细胞板从培养箱中取出,每孔加入20ul 2X Fluo-4DirectTM缓冲液,5%CO2,37℃培养箱孵育50分钟,室温放置10分钟。
激动剂NECA的EC80测定:
激动剂NECA稀释:将起始浓度为0.15mM的NECA用Echo进行10个点3倍连续稀释,接着转移900nL至相应化合物板;然后加30μl化合物稀释缓冲液至相应的化合物板。最终起始浓度为750nM。运行FLIPR仪器软件,按照设定程序,添加10ul化合物稀释缓冲液到细胞板中,读取荧光信号。再添加10ul既定浓度的激动剂参考化合物到细胞板中,读取荧光信号。读数后,通过软件中“Max-Min”,“Read 90to Maximum allowed”方法导出数据,计算A 2A细胞系的EC80,准备6X EC80浓度的激动剂。用缓冲盐溶液配制相应细胞6X EC80浓度的参考化合物激动剂,30ul/孔添加至相应的化合物板,备用。
待测化合物的IC50测定:
运行FLIPR仪器软件,按照设定程序,添加10μl既定浓度的检测化合物及参考化合物到细胞板中,读取荧光信号。再添加10ul 6X EC80浓度的参考化合物激动剂到细胞板中,读取荧光信号。对于化合物的激动剂检测,通过软件中“Max-Min”,“Read 1to 90”方法导出数据。对于化合物的拮抗剂检测,通过软件中“Max-Min”,“Read 90to Maximum allowed”方法导出数据。数据用GraphPad Prism 5.0进行数据分析,计算待测化合物的IC50值。
表11 本发明专利化合物体外筛选实验结果
Figure PCTCN2018082119-appb-000295
Figure PCTCN2018082119-appb-000296
结论:如表11中所示,本发明化合物表现出优异的腺苷A2a受体拮抗活性。
实验例2:动力学溶解度的测定
将受试化合物溶解在DMSO中,以制备10mmol/L的储备溶液。用50%ACN/50mM磷酸盐缓冲溶液(pH 7.4)稀释储备液,以配制标准溶液(1μM,20μM,200μM)。
用移液器(Eppendorf Research公司)将490μL溶解介质(50mM磷酸盐缓冲溶液,pH7.4)加入到单孔容积为2mL的96孔板中。分别将10μL各受试化合物的原液以及QC样品(10mmol/L DMSO储备溶液)添加到溶解介质中,用封板膜密封样品板。受试化合物和DMSO在样品溶液中的终浓度分别是为200μM和2%。室温下以每分钟600转的速度旋转摇动该混合物24小时。用移液器将200μL样品溶液转移到96孔滤膜板中,抽滤。用高效液相色谱法(HPLC-DAD)对滤液和标准溶液进行检测,用外标法计算滤液中受试化合物的浓度,即为该化合物的动力学溶解度。
表12
化合物 溶解度(μM)pH 7.4
化合物A 29.2
实施例A-1 185.0
实施例A-3 92.5
实施例A-4 160.0
实施例B-5 197.0
实施例B-28 54.5
实施例B-31 56.0
实施例C-19 194.0
实施例C-33 >200
实施例C-43 168.0
实施例C-46 172.0
实施例C-58 78.9
结论:如表12中所示,与化合物A相比,本发明化合物表现出优异的水溶解度(在pH=7.4)。
实验例3:化合物药代动力学评价
实验目的:测试化合物在雌性Balb/c小鼠体内药代动力学
实验材料:
Balb/c小鼠(雌性,15-30g,7~9周龄,上海灵畅)
实验操作:
以标准方案测试化合物静脉注射及口服给药后的啮齿类动物药代特征,实验中候选化合物配成澄清溶液给予小鼠单次静脉注射及均一混悬液给予小鼠单次口服给药。静注溶媒5%DMSO/95%10%Cremophor EL,口服溶媒为1%tween80,9%PEG400,90%water。收集24小时内的全血样品,在4度下3000g离心15分钟,分离上清得血浆样品,加入20倍体积含内标的乙腈溶液沉淀蛋白,离心取上清液加入等倍体积的水再离心取上清进样,以LC-MS/MS分析方法定量分析血药浓度,并计算药代参数,如达峰浓度,达峰时间,清除率,半衰期,药时曲线下面积,生物利用度等。
实验结果:
表13 药代动力学测试结果
Figure PCTCN2018082119-appb-000297
结论:本发明化合物可以显著提高小鼠药代动力学指标。

Claims (21)

  1. 式(Ⅰ)所示化合物、其异构体或其药学上可接受的盐,
    Figure PCTCN2018082119-appb-100001
    其中,
    R 1选自H、CN、COOH、
    Figure PCTCN2018082119-appb-100002
    或选自任选被1、2或3个R取代的:C 1-3烷基、C 1-3烷基-O-C 1-3烷基-、C 1-3烷基-C(=O)NH-、或C 3-6环烷基;
    R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1-6烷基或C 1-6杂烷基;
    R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-6烷基或C 1-6杂烷基;
    n选自:0、1、2或3;
    m选自:0、1、2或3;
    环A选自:6~10元芳基、5~10元杂芳基、5~10元杂环烷基或5~10元杂环烯基;
    环B选自:苯基或5~6元杂芳基;
    R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:C 1-3烷基、C 1-3烷氨基、C 1-3烷氧基、C 3-6环烷基、C 3-6环烷基-NH-、3~6元杂环烷基、3~6元杂环烷基-O-、或苯基;
    R’选自:F、Cl、Br、I、OH、NH 2、Me、
    Figure PCTCN2018082119-appb-100003
    所述C 1-6杂烷基、5~10元杂芳基、5~10元杂环烷基、3~6元杂环烷基或5~10元杂环烯基之“杂”分别独立地选自:N、O、S、NH、-C(=O)-、-C(=O)O-或-C(=O)NH-;
    上述杂原子或杂原子团的数目分别独立地选自1、2、3或4。
  2. 根据权利要求1所述化合物、其异构体或其药学上可接受的盐,其中,R选自F、Cl、Br、I、OH、NH 2、CN,或选自任选被1、2或3个R’取代的:Me、Et、
    Figure PCTCN2018082119-appb-100004
    Figure PCTCN2018082119-appb-100005
  3. 根据权利要求2所述化合物、其异构体或其药学上可接受的盐,其中,R选自F、Cl、Br、I、OH、 NH 2、CN、Me、Et、
    Figure PCTCN2018082119-appb-100006
    Figure PCTCN2018082119-appb-100007
  4. 根据权利要求1~3任意一项所述化合物、其异构体或其药学上可接受的盐,其中,R 1选自H、CN、COOH、
    Figure PCTCN2018082119-appb-100008
    或选自任选被1、2或3个R取代的:Me、Et、
    Figure PCTCN2018082119-appb-100009
  5. 根据权利要求4所述化合物、其异构体或其药学上可接受的盐,其中,R 1选自:H、CN、COOH、
    Figure PCTCN2018082119-appb-100010
  6. 根据权利要求1~3任意一项所述化合物、其异构体或其药学上可接受的盐,其中,R 2分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 3-6环烷基、C 1- 3烷基或C 1-3烷氧基。
  7. 根据权利要求6所述化合物、其异构体或其药学上可接受的盐,其中,R 2分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
    Figure PCTCN2018082119-appb-100011
  8. 根据权利要求1~3任意一项所述化合物、其异构体或其药学上可接受的盐,其中,R 3分别独立地选自H、F、Cl、Br、I、OH、NH 2、CN,或分别独立地选自任选被1、2或3个R取代的:C 1-3烷基或C 1-3烷氧基。
  9. 根据权利要求8所述化合物、其异构体或其药学上可接受的盐,其中,R 3分别独立地选自:H、F、Cl、Br、I、OH、NH 2、CN、Me、Et、CF 3
    Figure PCTCN2018082119-appb-100012
  10. 根据权利要求1~3任意一项所述化合物、其异构体或其药学上可接受的盐,其中,环A选自:苯基、吡啶基、四氢吡喃基、3,6-二氢-2H-吡喃基、哌啶基、1,2,3,6-四氢吡啶基、1H-吲哚基、1H-吲唑基、1H-苯并[d]咪唑基、苯并[d][1,3]二氧杂环戊烯基、二氢吲哚-2-酮基、1H-苯并[d][1,2,3]***基、喹啉基或1,2,3,4-四氢喹啉基。
  11. 根据权利要求10所述化合物、其异构体或其药学上可接受的盐,结构单元
    Figure PCTCN2018082119-appb-100013
    选自:
    Figure PCTCN2018082119-appb-100014
  12. 根据权利要求11所述化合物、其异构体或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2018082119-appb-100015
    选自:
    Figure PCTCN2018082119-appb-100016
  13. 根据权利要求7或12所述化合物、其异构体或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2018082119-appb-100017
    选自:
    Figure PCTCN2018082119-appb-100018
    Figure PCTCN2018082119-appb-100019
    Figure PCTCN2018082119-appb-100020
  14. 根据权利要求1~3任意一项所述化合物、其异构体或其药学上可接受的盐,其中,环B选自:苯基、吡啶基、咪唑基、吡唑基、呋喃基、噻吩基、噻唑基。
  15. 根据权利要求14所述化合物、其异构体或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2018082119-appb-100021
    选自:
    Figure PCTCN2018082119-appb-100022
    Figure PCTCN2018082119-appb-100023
  16. 根据权利要求15所述化合物、其异构体或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2018082119-appb-100024
    选自:
    Figure PCTCN2018082119-appb-100025
    Figure PCTCN2018082119-appb-100026
  17. 根据权利要求9或16所述化合物、其异构体或其药学上可接受的盐,其中,结构单元
    Figure PCTCN2018082119-appb-100027
    选自:
    Figure PCTCN2018082119-appb-100028
    Figure PCTCN2018082119-appb-100029
  18. 根据权利要求1~9任意一项所述化合物、其异构体或其药学上可接受的盐,其选自:
    Figure PCTCN2018082119-appb-100030
    其中,
    R 1、R 2、R 3如权利要求1~9所定义。
  19. 下式所示化合物、其异构体或其药学上可接受的盐,其选自:
    Figure PCTCN2018082119-appb-100031
    Figure PCTCN2018082119-appb-100032
    Figure PCTCN2018082119-appb-100033
    Figure PCTCN2018082119-appb-100034
    Figure PCTCN2018082119-appb-100035
    Figure PCTCN2018082119-appb-100036
  20. 一种药物组合物,包括治疗有效量的根据权利要求1~19任意一项所述的化合物、其异构体或其药学上可接受的盐,以及药学上可接受的载体。
  21. 根据权利要求1~19任意一项所述的化合物、其异构体或其药学上可接受的盐或根据权利要求20所述组合物在制备治疗与A 2A受体相关疾病的药物中的应用。
PCT/CN2018/082119 2017-04-07 2018-04-08 作为A 2A受体抑制剂的[1,2,4]***并[1,5-c]嘧啶衍生物 WO2018184590A1 (zh)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US16/500,901 US11117899B2 (en) 2017-04-07 2018-04-08 [1,2,4]triazolo[1,5-c]pyrimidine derivative as A2A receptor inhibitor
CN201880020259.9A CN110446712B (zh) 2017-04-07 2018-04-08 作为A2A受体抑制剂的[1,2,4]***并[1,5-c]嘧啶衍生物
DK18781739.0T DK3611174T3 (da) 2017-04-07 2018-04-08 [1,2,4]triazolo[1,5-c]pyrimidinderivat som a2a-receptorinhibitor
JP2019554827A JP7065113B2 (ja) 2017-04-07 2018-04-08 A2A受容体阻害剤としての[1,2,4]トリアゾロ[1,5-c]ピリミジン誘導体
ES18781739T ES2919474T3 (es) 2017-04-07 2018-04-08 Derivado de [1,2,4]triazolo[1,5-c]pirimidina como inhibidor del receptor A2A
EP18781739.0A EP3611174B1 (en) 2017-04-07 2018-04-08 [1,2,4]triazolo[1,5-c]pyrimidine derivative as a2a receptor inhibitor
PL18781739.0T PL3611174T3 (pl) 2017-04-07 2018-04-08 Pochodne [1,2,4]triazolo[1,5-c]pirymidyny jako inhibitor receptora a2a

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN201710224553 2017-04-07
CN201710224553.9 2017-04-07
CN201710737871 2017-08-24
CN201710737871.5 2017-08-24
CN201810129208.1 2018-02-08
CN201810129208 2018-02-08

Publications (1)

Publication Number Publication Date
WO2018184590A1 true WO2018184590A1 (zh) 2018-10-11

Family

ID=63712048

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/082119 WO2018184590A1 (zh) 2017-04-07 2018-04-08 作为A 2A受体抑制剂的[1,2,4]***并[1,5-c]嘧啶衍生物

Country Status (9)

Country Link
US (1) US11117899B2 (zh)
EP (1) EP3611174B1 (zh)
JP (1) JP7065113B2 (zh)
CN (1) CN110446712B (zh)
DK (1) DK3611174T3 (zh)
ES (1) ES2919474T3 (zh)
PL (1) PL3611174T3 (zh)
PT (1) PT3611174T (zh)
WO (1) WO2018184590A1 (zh)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019168847A1 (en) * 2018-02-27 2019-09-06 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as a2a / a2b inhibitors
WO2019206336A1 (zh) * 2018-04-28 2019-10-31 南京明德新药研发有限公司 一种***并嘧啶类化合物的晶型、盐型及其制备方法
WO2019222677A1 (en) * 2018-05-18 2019-11-21 Incyte Corporation Fused pyrimidine derivatives as a2a / a2b inhibitors
WO2020010197A1 (en) * 2018-07-05 2020-01-09 Incyte Corporation Fused pyrazine derivatives as a2a / a2b inhibitors
WO2020052631A1 (en) * 2018-09-12 2020-03-19 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Triazolo-pyrimidine compounds and uses thereof
WO2020106560A1 (en) 2018-11-20 2020-05-28 Merck Sharp & Dohme Corp. Substituted amino triazolopyrimidine and amino triazolopyrazine adenosine receptor antagonists, pharmaceutical compositions and their use
WO2020106558A1 (en) 2018-11-20 2020-05-28 Merck Sharp & Dohme Corp. Substituted amino triazolopyrimidine and amino triazolopyrazine adenosine receptor antagonists, pharmaceutical compositions and their use
WO2020108590A1 (zh) * 2018-11-30 2020-06-04 上海拓界生物医药科技有限公司 嘧啶并五元氮杂环类衍生物、其制备方法及其在医药上的应用
CN111848625A (zh) * 2019-04-24 2020-10-30 江苏恒瑞医药股份有限公司 一种杂芳基并[4,3-c]嘧啶-5-胺类化合物的制备方法及中间体
WO2021041360A1 (en) * 2019-08-26 2021-03-04 Incyte Corporation Triazolopyrimidines as a2a / a2b inhibitors
WO2021156439A1 (en) 2020-02-06 2021-08-12 Astrazeneca Ab Triazole compounds as adenosine receptor antagonists
RU2775229C1 (ru) * 2018-11-30 2022-06-28 Тоцзе Биотек (Шанхай) Ко., Лтд. Производное пиримидина и пятичленного азотсодержащего гетероцикла, способ его получения и его медицинские применения
US11390624B2 (en) 2019-01-29 2022-07-19 Incyte Corporation Pyrazolopyridines and triazolopyridines as A2A / A2B inhibitors
US11697666B2 (en) 2021-04-16 2023-07-11 Gilead Sciences, Inc. Methods of preparing carbanucleosides using amides
US11767337B2 (en) 2020-02-18 2023-09-26 Gilead Sciences, Inc. Antiviral compounds

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109535161B (zh) * 2017-09-22 2021-09-03 江苏恒瑞医药股份有限公司 ***并嘧啶类衍生物、其制备方法及其在医药上的应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0976753A1 (en) * 1997-03-24 2000-02-02 Kyowa Hakko Kogyo Co., Ltd. [1,2,4]TRIAZOLO[1,5-c]PYRIMIDINE DERIVATIVES
WO2004092173A2 (en) * 2003-04-09 2004-10-28 Biogen Idec Ma Inc. A2a adenosine receptor antagonists

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI330183B (zh) 2001-10-22 2010-09-11 Eisai R&D Man Co Ltd
AR038366A1 (es) 2001-11-30 2005-01-12 Schering Corp Compuestos de 1,2,4-triazolo [1,5-c] pirimidinas sustituidas, antagonistas del receptor de adenosina a2a, composiciones farmaceuticas, el uso de dichos compuestos para la manufactura de un medicamento para el tratamiento de enfermedades del sistema nervioso central y un kit que comprende combinacion
AU2003272886A1 (en) * 2002-09-24 2004-04-19 Kyowa Hakko Kogyo Co., Ltd. (1,2,4)-TRIAZOLO(1,5-c)PYRIMIDINE DERIVATIVE
CN109535161B (zh) * 2017-09-22 2021-09-03 江苏恒瑞医药股份有限公司 ***并嘧啶类衍生物、其制备方法及其在医药上的应用
CN117903140A (zh) * 2018-02-27 2024-04-19 因赛特公司 作为a2a/a2b抑制剂的咪唑并嘧啶和***并嘧啶
CN109535162B (zh) 2018-12-13 2020-03-24 浙江圣达生物药业股份有限公司 一种n-对氨基苯甲酰谷氨酰叶酸的制备方法

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0976753A1 (en) * 1997-03-24 2000-02-02 Kyowa Hakko Kogyo Co., Ltd. [1,2,4]TRIAZOLO[1,5-c]PYRIMIDINE DERIVATIVES
WO2004092173A2 (en) * 2003-04-09 2004-10-28 Biogen Idec Ma Inc. A2a adenosine receptor antagonists

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
J. MED. CHEM., vol. 55, 2012, pages 1898 - 1903
See also references of EP3611174A4

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112384515A (zh) * 2018-02-27 2021-02-19 因赛特公司 作为a2a/a2b抑制剂的咪唑并嘧啶和***并嘧啶
US20190292188A1 (en) * 2018-02-27 2019-09-26 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as a2a / a2b inhibitors
JP7474709B2 (ja) 2018-02-27 2024-04-25 インサイト・コーポレイション A2a/a2b阻害剤としてのイミダゾピリミジン及びトリアゾロピリミジン
JP2021515036A (ja) * 2018-02-27 2021-06-17 インサイト・コーポレイションIncyte Corporation A2a/a2b阻害剤としてのイミダゾピリミジン及びトリアゾロピリミジン
AU2019227607C1 (en) * 2018-02-27 2023-07-20 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as A2A / A2B inhibitors
US11673894B2 (en) 2018-02-27 2023-06-13 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as A2A / A2B inhibitors
AU2019227607B2 (en) * 2018-02-27 2023-04-27 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as A2A / A2B inhibitors
WO2019168847A1 (en) * 2018-02-27 2019-09-06 Incyte Corporation Imidazopyrimidines and triazolopyrimidines as a2a / a2b inhibitors
WO2019206336A1 (zh) * 2018-04-28 2019-10-31 南京明德新药研发有限公司 一种***并嘧啶类化合物的晶型、盐型及其制备方法
EP3766884A4 (en) * 2018-04-28 2021-03-31 Medshine Discovery Inc. CRYSTAL SHAPE AND SALT TYPE OF A TRIAZOLOPYRIMIDINE COMPOUND AND METHOD FOR MANUFACTURING IT
WO2019222677A1 (en) * 2018-05-18 2019-11-21 Incyte Corporation Fused pyrimidine derivatives as a2a / a2b inhibitors
US11873304B2 (en) 2018-05-18 2024-01-16 Incyte Corporation Fused pyrimidine derivatives as A2A/A2B inhibitors
US11168089B2 (en) 2018-05-18 2021-11-09 Incyte Corporation Fused pyrimidine derivatives as A2A / A2B inhibitors
JP7391046B2 (ja) 2018-05-18 2023-12-04 インサイト・コーポレイション A2a/a2b阻害剤としての縮合ピリミジン誘導体
JP2021524457A (ja) * 2018-05-18 2021-09-13 インサイト・コーポレイションIncyte Corporation A2a/a2b阻害剤としての縮合ピリミジン誘導体
WO2020010197A1 (en) * 2018-07-05 2020-01-09 Incyte Corporation Fused pyrazine derivatives as a2a / a2b inhibitors
CN113166153A (zh) * 2018-07-05 2021-07-23 因赛特公司 作为a2a/a2b抑制剂的稠合吡嗪衍生物
US11999740B2 (en) 2018-07-05 2024-06-04 Incyte Corporation Fused pyrazine derivatives as A2A / A2B inhibitors
US11161850B2 (en) 2018-07-05 2021-11-02 Incyte Corporation Fused pyrazine derivatives as A2A / A2B inhibitors
WO2020052631A1 (en) * 2018-09-12 2020-03-19 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Triazolo-pyrimidine compounds and uses thereof
US11629147B2 (en) 2018-09-12 2023-04-18 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Triazolo-pyrimidine compounds and uses thereof
JP2022500402A (ja) * 2018-09-12 2022-01-04 ディザル(ジァンスー)ファーマシューティカル・カンパニー・リミテッド トリアゾロ−ピリミジン化合物およびそれらの使用
US10858365B2 (en) 2018-09-12 2020-12-08 Dizal (Jiangsu) Pharmaceutical Co., Ltd. Triazolo-pyrimidine compounds and uses thereof
EP3883576A4 (en) * 2018-11-20 2022-06-22 Merck Sharp & Dohme Corp. SUBSTITUTED AMINOTRIAZOLOPYRIMIDINES AND AMINO-TRIAZOLOPYRAZINE ADENOSINE RECEPTOR ANTAGONISTS, PHARMACEUTICAL COMPOSITIONS AND THEIR USE
EP3883575A4 (en) * 2018-11-20 2022-06-15 Merck Sharp & Dohme Corp. SUBSTITUTE AMINO-TRIAZOLOPYRIMIDINE AND AMINO-TRIAZOLOPYRAZINE ADENOSINE RECEPTOR ANTAGONIST, PHARMACEUTICAL COMPOSITIONS AND THEIR USE
WO2020106558A1 (en) 2018-11-20 2020-05-28 Merck Sharp & Dohme Corp. Substituted amino triazolopyrimidine and amino triazolopyrazine adenosine receptor antagonists, pharmaceutical compositions and their use
WO2020106560A1 (en) 2018-11-20 2020-05-28 Merck Sharp & Dohme Corp. Substituted amino triazolopyrimidine and amino triazolopyrazine adenosine receptor antagonists, pharmaceutical compositions and their use
RU2775229C1 (ru) * 2018-11-30 2022-06-28 Тоцзе Биотек (Шанхай) Ко., Лтд. Производное пиримидина и пятичленного азотсодержащего гетероцикла, способ его получения и его медицинские применения
CN113272303A (zh) * 2018-11-30 2021-08-17 上海拓界生物医药科技有限公司 嘧啶并五元氮杂环类衍生物、其制备方法及其在医药上的应用
WO2020108590A1 (zh) * 2018-11-30 2020-06-04 上海拓界生物医药科技有限公司 嘧啶并五元氮杂环类衍生物、其制备方法及其在医药上的应用
US11390624B2 (en) 2019-01-29 2022-07-19 Incyte Corporation Pyrazolopyridines and triazolopyridines as A2A / A2B inhibitors
US11884665B2 (en) 2019-01-29 2024-01-30 Incyte Corporation Pyrazolopyridines and triazolopyridines as A2A / A2B inhibitors
CN111848625A (zh) * 2019-04-24 2020-10-30 江苏恒瑞医药股份有限公司 一种杂芳基并[4,3-c]嘧啶-5-胺类化合物的制备方法及中间体
CN111848625B (zh) * 2019-04-24 2022-07-26 江苏恒瑞医药股份有限公司 一种杂芳基并[4,3-c]嘧啶-5-胺类化合物的制备方法及中间体
CN114585625A (zh) * 2019-08-26 2022-06-03 因赛特公司 作为a2a/a2b抑制剂的***并嘧啶
WO2021041360A1 (en) * 2019-08-26 2021-03-04 Incyte Corporation Triazolopyrimidines as a2a / a2b inhibitors
WO2021156439A1 (en) 2020-02-06 2021-08-12 Astrazeneca Ab Triazole compounds as adenosine receptor antagonists
US11767337B2 (en) 2020-02-18 2023-09-26 Gilead Sciences, Inc. Antiviral compounds
US11697666B2 (en) 2021-04-16 2023-07-11 Gilead Sciences, Inc. Methods of preparing carbanucleosides using amides

Also Published As

Publication number Publication date
EP3611174A4 (en) 2020-09-09
PT3611174T (pt) 2022-07-07
JP2020516606A (ja) 2020-06-11
CN110446712B (zh) 2021-09-14
ES2919474T3 (es) 2022-07-26
JP7065113B2 (ja) 2022-05-11
CN110446712A (zh) 2019-11-12
EP3611174B1 (en) 2022-06-08
US11117899B2 (en) 2021-09-14
US20200131184A1 (en) 2020-04-30
PL3611174T3 (pl) 2022-08-08
DK3611174T3 (da) 2022-07-04
EP3611174A1 (en) 2020-02-19

Similar Documents

Publication Publication Date Title
WO2018184590A1 (zh) 作为A 2A受体抑制剂的[1,2,4]***并[1,5-c]嘧啶衍生物
JP7377798B2 (ja) c-MET/AXL阻害剤としてのウラシル系化合物
WO2019015655A1 (zh) 作为egfr激酶抑制剂的芳基磷氧化合物
EP3470409B1 (en) Benzotriazole-derived alpha and beta-unsaturated amide compound used as tgf-beta ri inhibitor
JP6836693B2 (ja) A2a受容体アンタゴニストとしての縮合環誘導体
TW201706277A (zh) Janus激酶抑制劑
CN109661395B (zh) Pde4抑制剂
WO2015192760A1 (zh) 作为pi3k抑制剂的吡啶并[1,2-a]嘧啶酮类似物
WO2017177974A1 (zh) 用于治疗纤维化和炎性疾病的含杂原子环丁烷取代基的吡啶酮衍生物
WO2019154364A1 (zh) 作为fgfr抑制剂的吡嗪-2(1h)-酮类化合物
TWI801394B (zh) 作為mek抑制劑的類香豆素環類化合物及其應用
WO2018036470A1 (zh) 作为pde4抑制剂的并环类化合物
WO2018103757A1 (zh) 作为ccr2/ccr5受体拮抗剂的联苯化合物
WO2019072235A1 (zh) 2,6-二氧杂螺[4,5]癸烷类衍生物、其制备方法及其在医药上的应用
WO2019134662A1 (zh) 作为csf-1r抑制剂的杂环化合物及其应用
WO2019134661A1 (zh) 异吲哚啉酮及其衍生物作为csf-1r抑制剂
WO2017215588A1 (zh) 作为Akt抑制剂的二氢吡唑氮杂卓类化合物
WO2018028557A1 (zh) 三环类化合物及其应用
WO2016169514A1 (zh) 咪唑类化合物
WO2018228435A1 (zh) 作为gls1抑制剂的化合物
TWI402264B (zh) 芳基甲基苯并喹唑啉酮m1受體之正向異位性調節劑
WO2017202376A1 (zh) 磺酰胺衍生物

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: 18781739

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019554827

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: 2018781739

Country of ref document: EP

Effective date: 20191107