WO2022247796A1 - 周期蛋白依赖性激酶9抑制剂的用途 - Google Patents

周期蛋白依赖性激酶9抑制剂的用途 Download PDF

Info

Publication number
WO2022247796A1
WO2022247796A1 PCT/CN2022/094496 CN2022094496W WO2022247796A1 WO 2022247796 A1 WO2022247796 A1 WO 2022247796A1 CN 2022094496 W CN2022094496 W CN 2022094496W WO 2022247796 A1 WO2022247796 A1 WO 2022247796A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
independently selected
dichloromethane
mmol
Prior art date
Application number
PCT/CN2022/094496
Other languages
English (en)
French (fr)
Inventor
魏冰
范丽雪
赵传武
王佳
柴晓玲
高娜
郝红茹
Original Assignee
石药集团中奇制药技术(石家庄)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 石药集团中奇制药技术(石家庄)有限公司 filed Critical 石药集团中奇制药技术(石家庄)有限公司
Publication of WO2022247796A1 publication Critical patent/WO2022247796A1/zh

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4418Non condensed pyridines; Hydrogenated derivatives thereof having a carbocyclic group directly attached to the heterocyclic ring, e.g. cyproheptadine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/443Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4433Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4436Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a heterocyclic ring having sulfur as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present application relates to the field of medicine, in particular, to a cyclin-dependent kinase 9 (CDK9) inhibitor or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotope derivative or a prodrug thereof, and a pharmaceutical composition comprising the same , or the use of a medicament containing it, particularly the inhibitor or its pharmaceutically acceptable salt, its stereoisomer, isotopic derivative or prodrug, a pharmaceutical composition containing it, or a medicament containing it in the preparation of therapeutic blood Use in medicine for neoplastic diseases, especially hematological malignancies.
  • CDK9 cyclin-dependent kinase 9
  • CDKs Cyclin-dependent kinases
  • CDKs are a class of serine/threonine protein kinases that play key roles in regulating the cell cycle and transcription.
  • CDKs can be activated by cell cycle proteins and play different biological functions.
  • CDKs are classified according to The functions can be divided into two types, one controls the cell cycle and the other regulates cell transcription.
  • CDK1, 2, 3, 4, and 6 directly intervene in the cell cycle; CDK5 does not regulate the cell cycle, but plays a key role in the complex migration of neurons after mitosis; CDK7 acts indirectly as an activator of these CDKs; CDK9 acts only in the cell transcription play a role in the regulation of the cell cycle.
  • CDK9 is an important member of the transcriptional CDKs subfamily, a group of kinases whose function is to control the main steps in the synthesis and processing of mRNA by eukaryotic RNA polymerase II (Pol II).
  • CDK9 exists in all mammalian cells, and the activation of CDK9 in vivo depends on its combination with the corresponding cell cycle protein (Cyclin T/K), forming a heterodimer, that is, positive transcription elongation factor b (P-TEFb ).
  • NELF negative transcription elongation factor
  • NELFs negative transcription elongation factors
  • P-TEFb Phosphorylation of the CTD domain
  • CDK9 cyclin-dependent kinase inhibitors
  • CDK9 inhibitors for cancer treatment, for example, AZD4573 of AstraZeneca and BAY-1251152 of Bayer, both of which are in clinical I
  • AZD4573 of AstraZeneca and BAY-1251152 of Bayer both of which are in clinical I
  • BAY-1251152 of Bayer both of which are in clinical I
  • no CDK9 inhibitor has been approved for marketing.
  • CDK9 inhibitors Although some small molecules of CDK9 inhibitors have been disclosed (for example, WO2009047359, WO2014076091, etc.), it is still necessary to develop new compounds with good efficacy and safety, so as to benefit more patients clinically.
  • the present application provides a compound as shown in formula (I) or its pharmaceutically acceptable salt, its stereoisomer, isotopic derivative or prodrug used in the preparation of a drug for the treatment of hematological tumors, especially malignant hematological tumors Uses in medicine.
  • the present application provides a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof for use in the preparation and treatment of leukemia, lymphoma or myeloma, In particular, the use in the medicine of CDK9-related leukemia, lymphoma or myeloma.
  • the present application provides a CDK9 inhibitor compound 45 or a pharmaceutically acceptable salt thereof, its stereoisomer, isotopic derivative or prodrug in the preparation of a drug for treating hematological tumors, especially malignant hematological tumors the use of.
  • the present application provides a CDK9 inhibitor compound 45 or its pharmaceutically acceptable salt, its stereoisomer, isotopic derivative or prodrug for the preparation and treatment of leukemia, lymphoma or myeloma, especially CDK9 Related medicines for leukemia, lymphoma or myeloma.
  • the present application provides a pharmaceutical composition for treating hematological tumors, which comprises a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, its stereoisomer, isotopic derivative or pro medicine.
  • the application provides a pharmaceutical composition for treating leukemia, lymphoma and myeloma, which comprises a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, Isotopic derivatives or prodrugs.
  • the present application provides a pharmaceutical composition for treating hematological tumors, which comprises a CDK9 inhibitor compound 45 or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotope derivative or a prodrug thereof .
  • the present application provides a pharmaceutical composition for treating leukemia, lymphoma and myeloma, which comprises a CDK9 inhibitor compound 45 or a pharmaceutically acceptable salt thereof, stereoisomers, isotopic derivatives thereof or prodrugs of the drug.
  • X is selected from Cl, F, wherein F is preferred;
  • R 1 is selected from substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl; the "substituted" in R 1 means that it is independently replaced by 1, 2, 3, 4 or 5 selected from -F, -Cl, -Br, -NH 2 , -OH, -SH, -CN, -NO 2 , -N 3 , -C ⁇ CH, -COOH, -R 3 , -(CH 2 ) w O(CH 2 ) n R 3 , -(CH 2 ) w NH(CH 2 ) n R 3 , -(CH 2 ) w NR 3 (CH 2 ) n R 4 , -(CH 2 ) w S(CH 2 ) n R 3 , -(CH 2 ) w C(O)(CH 2 ) n R 3 , -(CH 2 ) w C(O)O(CH 2 ) n R 3
  • the A ring is selected from substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl; the "substituted" in the A ring refers to being replaced by 1, 2, 3, 4 or 5 independently selected from -F, -Cl, -Br, OH, NH 2 , SH, CN, NO 2 , -N 3 , -C ⁇ CH, COOH, R 5 , OR 5 , -NHR 5 , -NR 5 R 6 , -SR 5 , -NHCOR 5 , -CONHR 5 , -NHS(O) 2 R 5 , -S(O) 2 NHR 5 , -NR 5 S(O) 2 R 6 , -S(O) 2 NR 5 R 6 is replaced by a group, or one, two or more -CH 2 - groups in the A ring structure can be optionally replaced by a -C(O)- group; wherein, R 5
  • R 2 is selected from H, R 7 , -(CH 2 ) x R 7 , -(CH 2 ) x NH(CH 2 ) y R 7 , -(CH 2 ) x O(CH 2 ) y R 7 , -( CH 2 ) x NR 7 (CH 2 ) y R 8 , -(CH 2 ) x C(O)(CH 2 ) y H, -(CH 2 ) x C(O)(CH 2 ) y R 7 , - (CH 2 ) x S(O) 2 (CH 2 ) y R 7 , -(CH 2 ) x C(O)C(O)(CH 2 ) y R 7 , -(CH 2 ) x S(O) 2 NH 2 , -(CH 2 ) x NHS(O) 2 H, -(CH 2 ) x S(O) 2 NH(CH 2 ) y R 7 , -(
  • R 7 and R 8 are independently selected from substituted or unsubstituted R 9 , OR 9 , -R 10 -OR 9 , -R 10 -NH-R 9 , -R 10 -C(O)-R 9 , -R 10 -NHC(O)-R 9 , -R 10 -C(O)NH-R 9 , -R 10 -SR 9 , -R 10 -S(O)-R 9 , -R 10 -SC(O) -R 9 , cycloalkyl, heterocycloalkyl, aryl, heteroaryl, -R 10 -aryl, -R 10 -heteroaryl, -OR 10 -aryl, -OR 10 -heteroaryl, -R 10 -O-aryl, -R 10 -O-heteroaryl, -cycloalkyl-aryl, -cycloalkyl-aryl-heteroaryl,
  • -C ⁇ CH -COOH, C 1-6 alkyl, C 1-6 alkoxy, C 1-6 haloalkyl, C 1-6 haloalkoxy , -NHCN, -NHCONH 2 , NHC(O ) CH 3 , N(CH 3 ) 2 , N(C 2 H 5 ) 2 , -SC(O)CH 3 , -OC(O)-C 1-6 alkyl, etc.;
  • said isotopic derivatives are deuterated forms of derivatives.
  • aryl preferably contains 6-10 carbon atoms
  • cycloalkyl preferably contains 3-6 carbon atoms
  • heteroaryl is preferably 5-10 membered heteroaryl
  • heterocycloalkyl is preferably 3 ⁇ 8-membered heterocyclyl
  • heteroaryl or heterocycloalkyl preferably contains 1, 2 or 3 heteroatoms each independently selected from N, O or S, the remainder being carbon atoms.
  • the compound represented by formula (I) or its pharmaceutically acceptable salt, its stereoisomer, isotopic derivative or prodrug wherein, the A ring is selected from substituted or unsubstituted 4-6 membered Cycloalkyl, substituted or unsubstituted 4-6 membered heterocycloalkyl, or substituted or unsubstituted 5-6 cycloalkyl, or substituted or unsubstituted 5-6 membered heterocycloalkyl; more preferably, A The ring is selected from substituted or unsubstituted 5-6 cycloalkyl, or substituted or unsubstituted 5-6 membered heterocycloalkyl; more preferably, ring A is selected from substituted or unsubstituted 5-6 cycloalkyl.
  • ring A is selected from cyclohexane, tetrahydropyrrolyl, piperidinyl, piperazinyl, cyclopentyl or morpholinyl, further preferably, ring A is selected from cyclohexane, Cyclopentyl or tetrahydropyrrolyl.
  • the compound represented by the formula (I) or its pharmaceutically acceptable salt, its stereoisomer, isotopic derivative or prodrug wherein the "substitution" in the A ring refers to being replaced by 1 , 2, 3, 4 or 5 groups independently selected from -F, -Cl, -Br, OH, NH 2 , SH, CN, R 5 , OR 5 are substituted, wherein, R 5 is C 1-6 alkyl or C 1-6 alkoxy, and R 5 can be further selected from C 1-4 alkyl or C 1-4 alkoxy.
  • the compound represented by the formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof has a structure represented by the formula (II):
  • R 1 , R 2 and X are as shown in formula (I).
  • the compound represented by the formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof has a structure represented by the formula (IIa):
  • R 1 , R 2 and X are as shown in formula (I).
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof has a structure represented by formula (III):
  • R 1 , R 2 and X are as shown in formula (I).
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof has a structure represented by formula (IIIa):
  • R 1 , R 2 and X are as shown in formula (I).
  • the compound represented by formula (I) or its pharmaceutically acceptable salt, its stereoisomer, isotopic derivative or prodrug wherein, R is selected from substituted or unsubstituted 6-10 membered Aryl, or a substituted or unsubstituted 5-10 membered heteroaryl; the heteroaryl contains 1 or 2 heteroatoms independently selected from N or O; the number of the substituents is selected from 1, 2 or 3.
  • the compound represented by the formula (I) or its pharmaceutically acceptable salt, its stereoisomer, isotopic derivative or prodrug wherein, R is selected from substituted or unsubstituted benzene ring, pyridine ring, indole ring, indazole ring, benzofuran ring, pyrrolopyridine ring; more preferably substituted or unsubstituted benzene ring, pyridine ring, indole ring, benzofuran ring, pyrrolopyridine ring; more preferably substituted The benzene ring, pyridine ring and unsubstituted indole ring, benzofuran ring, pyrrolopyridine ring; more preferably substituted benzene ring.
  • the compound represented by the formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof wherein the substituents on the R1 are independently selected from -F, -Cl, -OH, -NH 2 , -R 3 , -(CH 2 ) w O(CH 2 ) n R 3 or -(CH 2 ) w OC(O)(CH 2 ) n R 3 ;
  • Each occurrence of w and n is independently selected from 0, 1 or 2, wherein R 3 is as defined in formula (I).
  • the substituents on R 1 are independently selected from -F-, -Cl, -OH, -R 3 , -(CH 2 ) w O(CH 2 ) n R 3 ; further preferably, the The substituents on R 1 are independently selected from -F-, -OH, -R 3 , -(CH 2 ) w O(CH 2 ) n R 3 .
  • R is a substituted benzene ring
  • the substituents are selected from -F, -OH or alkoxy, preferably 1 or 2 fluorine atoms and 1 -OH or alkoxy, preferably 1 or 2 Fluorine atom substitution and 1 alkoxyl substitution.
  • the compound represented by the formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof has a structure represented by the formula (II):
  • R is a substituted benzene ring, and the substituent is selected from -F, -OH or alkoxy; preferably 1 or 2 fluorine atoms are substituted and 1 -OH or alkoxy is substituted, preferably 1 or 2 fluorine atoms substituted and 1 alkoxy substituted;
  • R 2 and X are shown in formula (I).
  • the compound represented by the formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof has a structure represented by the formula (IIa):
  • R is a substituted benzene ring, and the substituent is selected from -F, -OH or alkoxy; preferably 1 or 2 fluorine atoms are substituted and 1 -OH or alkoxy is substituted, preferably 1 or 2 fluorine atoms substituted and 1 alkoxy substituted;
  • R 2 and X are shown in formula (I).
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof wherein R and R are independently selected from substituted or unsubstituted Substituted 6-membered aryl, substituted or unsubstituted 5-6-membered heteroaryl, substituted or unsubstituted C 1-3 alkyl, substituted or unsubstituted C 1-3 alkoxy, substituted or unsubstituted C 3-6 cycloalkyl, substituted or unsubstituted C 3-6 heterocycloalkyl, or when R 3 and R 4 are jointly linked to the same nitrogen atom, R 3 , R 4 and the nitrogen atom to which they are jointly linked 3-7 membered substituted or unsubstituted heterocycloalkyl; said heterocycloalkyl contains 1 or 2 heteroatoms independently selected from N, O or S; the "substitution" described in R3 and R4 refers to 1,
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof wherein R and R are independently selected from substituted or unsubstituted Substituted benzene ring, pyridine ring, methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, isopropoxy, cyclopropyl, cyclobutyl, cyclopentyl or Cyclohexyl, "substituted" in R 3 and R 4 refers to being 1, 2 or 3 independently selected from -F, -Cl, -Br, -OH, -CH 3 , -C 2 Substituents of H 5 , -OCH 3 , -OC 2 H 5 .
  • R and R are independently selected from substituted or unsubstituted methyl, ethyl, isopropyl, methoxy, ethoxy, isopropoxy, cyclopropyl, pyridine rings; more preferably, R 3 and R 4 are independently selected from substituted or unsubstituted methyl, ethyl, isopropyl, methoxy, ethoxy, isopropoxy; more preferably, R 3 and R 4 are independently selected from substituted or unsubstituted methyl and methoxy groups.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof wherein R 2 is selected from R 7 , -(CH 2 ) x R 7 , -(CH 2 ) x NH(CH 2 ) y R 7 , -(CH 2 ) x C(O)(CH 2 ) y R 7 , -(CH 2 ) x S(O) 2 (CH 2 ) y R 7 , -(CH 2 ) x C(O)C(O)(CH 2 ) y R 7 , -(CH 2 ) x C(O)O(CH 2 ) y R 7 , -(CH 2 ) x C(O)NH(CH 2 ) y R 7 , -(CH 2 ) x C(O)NR 7 (CH 2 ) y R 8 or -(CH 2 ) x NR 7 C(O)
  • R 2 is selected from R 7 , -(CH 2 ) x R 7 , -(CH 2 ) x C(O)(CH 2 ) y R 7 , -(CH 2 ) x S(O) 2 ( CH 2 ) y R 7 , -(CH 2 ) x C(O)C(O)(CH 2 ) y R 7 , -(CH 2 ) x C(O)O(CH 2 ) y R 7 , -( CH 2 ) x C(O)NH(CH 2 ) y R 7 ; more preferably, R 2 is selected from R 7 , -(CH 2 ) x R 7 , -(CH 2 ) x C(O)(CH 2 ) y R 7 ; more preferably, R 2 is selected from -(CH 2 ) x C(O)(CH 2 ) y R 7 .
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof wherein R 7 and R 8 are each independently selected from substituted or unsubstituted Substituted R 9 , OR 9 , -R 10 -OR 9 , -R 10 -NH-R 9 , -R 10 -C(O)-R 9 , -R 10 -NHC(O)-R 9 , -R 10 -C(O)NH-R 9 , -R 10 -SR 9 , -R 10 -SC(O)-R 9 , C 3-6 cycloalkyl, 3-6 membered heterocycloalkyl, C 6- 10 aryl, 5-10 membered heteroaryl, -R 10 -C 6-10 aryl, -R 10 -5-10 membered heteroaryl, -OR 10 -C 6-10 aryl, -OR 10 - 5-10
  • R 7 and R 8 form a substituted or unsubstituted 3-6 membered heterocycloalkyl group with the jointly connected nitrogen atom; further preferably, R 7 is independently selected from the group consisting of substituted Or unsubstituted R 9 , OR 9 , -R 10 -OR 9 , C 3-6 cycloalkyl, 3-6 membered heterocycloalkyl, C 6-10 aryl, 5-10 membered heteroaryl; further Preferably, R 7 is independently selected from substituted or unsubstituted R 9 , OR 9 , -R 10 -OR 9 , C 3-6 cycloalkyl, 3-6 membered heterocycloalkyl; more preferably, R 7 independently
  • the compound represented by the formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof wherein each of the R 7 and R 8 is independently selected from the substituent or unsubstituted methyl, ethyl, propyl, isopropyl, butyl, pentyl, methoxy, ethoxy, propoxy, isopropoxy, -CH 2 OCH 3 , -CH 2 OCH 2 CH 3 , -CH 2 CH 2 OCH 3 , -CH 2 CH 2 OCH 2 CH 3 , cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, aziridyl, azetidinyl, nitrogen Heterocyclopentyl, azacyclohexyl, oxirane, oxetanyl, oxolyl, oxanyl, phenyl, pyridyl,
  • R is independently selected from substituted or unsubstituted methyl, ethyl, methoxy, ethoxy, cyclopropyl, cyclobutyl, azetidinyl, azacyclohexyl, oxetane Butyl, oxanyl, phenyl, pyridyl, pyrazolyl, isoxazolyl, thienyl, thiazolyl, benzyl, vinyl, propenyl or ethynyl; further preferably, R independently selected from substituted or unsubstituted methyl, ethyl, methoxy, cyclopropyl, cyclobutyl, azacyclohexyl, oxetanyl, oxetyl, pyridyl, pyrazolyl, isoxane Azolyl, vinyl, propenyl or ethynyl; Further preferably, R is
  • R 7 and R 8 refers to being 1, 2 or 3 independently selected from -F, -Cl, -Br, -OH, -NH 2 , -SH, -CN, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkyl, C 1-3 haloalkoxy, -NHCN, -NHCONH 2 , NHC(O)CH 3 , N(CH 3 ) 2 , N(C 2 H 5 ) 2 , -SC(O)CH 3 , -OC(O)-C 1-6 alkyl, etc.; preferably , the ""Substituted” refers to being 1, 2 or 3 independently selected from -F, -OH, -SH, -CN, C 1-3 alkyl, C 1-3 alkoxy, C 1-3 haloalkane Oxygen, NHC(O)CH 3 , N(CH 3 ) 2 , N(C 2 H 5 ) 2
  • substitution in R7 and R8 refers to being substituted by 1, 2 or 3 groups independently selected from -F, -OH, -CN, etc.; Further preferably, the "substitution" in R 7 and R 8 refers to being substituted by 1, 2 or 3 groups independently selected from -OH, -CN, etc.; further preferably, R 7 and the "substituted” in R 8 means to be substituted by one group independently selected from -OH, -CN, etc.; further preferably, the "substituted" in R 7 and R 8 means Substituted by 1 group each independently selected from -OH.
  • the compound represented by the formula (I) or its pharmaceutically acceptable salt, its stereoisomer, isotopic derivative or prodrug wherein, R 9 is C 1-4 alkyl, R 10 is C 1-4 alkylene.
  • the hematological tumor is a malignant hematological tumor, preferably a relapsed or refractory hematological tumor.
  • the hematological tumor is selected from leukemia, lymphoma and myeloma, preferably relapsed or refractory leukemia, lymphoma and myeloma.
  • the leukemia, lymphoma or myeloma refers to CDK9-related leukemia, lymphoma or myeloma.
  • the leukemia is acute leukemia, preferably acute myeloid leukemia, more preferably relapsed or refractory acute myeloid leukemia, further preferably CDK9-related relapsed or refractory acute myeloid leukemia.
  • the blood tumor is selected from relapsed or refractory acute myeloid leukemia, lymphoma and myeloma; preferably lymphoma and myeloma; more preferably relapsed or refractory lymphoma and myeloma.
  • the lymphoma is non-Hodgkin's lymphoma, preferably diffuse large B-cell lymphoma or mantle cell lymphoma, more preferably CDK9-related diffuse large B-cell lymphoma or mantle cell lymphoma.
  • the myeloma is multiple myeloma, preferably CDK9-related multiple myeloma.
  • said CDK9 association refers to CDK9 overexpression.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt, stereoisomer, isotopic derivative or prodrug thereof is the only active ingredient in the drug.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof is used in combination with one or more other targeted or chemotherapeutic drugs.
  • the medicament is formulated into a clinically acceptable formulation.
  • the formulation is an oral formulation or an injectable formulation.
  • the compound represented by formula (I) or its pharmaceutically acceptable salt, its stereoisomer, isotopic derivative or prodrug is administered at a daily dose of from about 0.001 mg/kg to about 1000 mg/kg
  • the dosage range is administered; preferably 0.01 mg/kg to about 100 mg/kg, more preferably 0.02 mg/kg to about 10 mg/kg.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof is administered in a daily dosage range from about 0.001 mg to about 1000 mg ; Preferably 0.01mg to about 100mg, more preferably 0.1mg to about 80mg, further preferably 1mg to about 70mg, further preferably 1.5mg to about 60mg, further preferably 2mg to about 50mg; more preferably 2mg, 3mg, 4mg, 5mg, 6mg , 8mg, 10mg, 12mg, 16mg, 18mg, 20mg, 24mg, 25mg, 30mg, 32mg, 36mg, 40mg, 42mg, 45mg, or 50mg.
  • the dosing frequency is single dose or multiple doses.
  • the dosing frequency is once a day, twice a day, three times a day, once every two days, once every three days, once every four days, once every five days 1. Dosing once every six days or once every seven days.
  • the medicament contains a therapeutically effective amount of a compound represented by formula (I) or a pharmaceutically acceptable salt, stereoisomer, isotope derivative or prodrug thereof.
  • the therapeutically effective amount is 0.001-1000 mg.
  • the therapeutically effective amount is 0.01-100 mg.
  • the therapeutically effective amount is 0.1-50 mg; more preferably 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 8 mg, 10 mg, 12 mg, 16 mg, 18 mg, 20 mg, 24 mg, 25 mg, 30 mg, 32 mg , 36mg, 40mg, 42mg, 45mg, or 50mg.
  • the single dosage form of the drug contains 0.01-100mg of the compound represented by formula (I) or its pharmaceutically acceptable salt, stereoisomer, isotope derivative or prodrug; preferably 0.1-80mg; more preferably 1-60mg; further preferably 1-50mg; further preferably 2-20mg; further preferably 2-15mg; further preferably 2-10mg; -6mg; more preferably 2-5mg; further preferably 2-4mg; further preferably 2-3mg.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt, stereoisomer, isotopic derivative or prodrug thereof is administered in a single dose or in divided doses.
  • the medicament is administered orally or by injection. In a preferred embodiment, the medicament is administered orally.
  • a method for treating a hematological tumor in an individual comprising administering to the individual a therapeutically effective amount of a A compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof, or a compound represented by formula (I) as described in the first aspect or a pharmaceutically acceptable salts, stereoisomers, isotopic derivatives or prodrugs thereof.
  • a pharmaceutical composition which comprises the compound represented by formula (I) or its pharmaceutically acceptable salt, stereoisomer, isotopic derivative or A prodrug, and optionally a pharmaceutically acceptable carrier.
  • the pharmaceutical composition is used to treat hematological tumors.
  • the compound represented by formula (I) or a pharmaceutically acceptable salt, stereoisomer, isotopic derivative or prodrug thereof is the only active ingredient in the pharmaceutical composition.
  • the pharmaceutical composition further comprises one or more other targeted drugs or chemotherapeutic drugs as active ingredients.
  • the pharmaceutical composition is formulated into a clinically acceptable formulation.
  • the formulation is an oral formulation or an injectable formulation.
  • the pharmaceutical composition contains a therapeutically effective amount of a compound represented by formula (I) or a pharmaceutically acceptable salt, stereoisomer, isotopic derivative or prodrug thereof.
  • the therapeutically effective amount is 0.001-1000 mg.
  • the therapeutically effective amount is 0.01-100 mg.
  • the therapeutically effective amount is 0.1-50 mg.
  • the single dosage form of the pharmaceutical composition contains 0.01-100 mg of the compound represented by formula (I) or its pharmaceutically acceptable salt, stereoisomer, isotope derivative or prodrug; preferably 0.1-80mg; more preferably 1-60mg; further preferably 1-50mg; further preferably 2-20mg; further preferably 2-15mg; further preferably 2-10mg; -6mg; more preferably 2-5mg; further preferably 2-4mg; further preferably 2-3mg.
  • the pharmaceutical composition is administered orally or by injection. In a preferred embodiment, the pharmaceutical composition is administered orally.
  • the hematological tumor is a malignant hematological tumor, preferably a relapsed or refractory hematological tumor.
  • the hematological tumor is selected from leukemia, lymphoma and myeloma, preferably relapsed or refractory leukemia, lymphoma and myeloma.
  • the leukemia, lymphoma or myeloma refers to CDK9-related leukemia, lymphoma or myeloma.
  • the leukemia is acute leukemia, preferably acute myeloid leukemia, more preferably relapsed or refractory acute myeloid leukemia, further preferably CDK9-related relapsed or refractory acute myeloid leukemia.
  • the blood tumor is selected from relapsed or refractory acute myeloid leukemia, lymphoma and myeloma; preferably lymphoma and myeloma; more preferably relapsed or refractory lymphoma and myeloma.
  • the lymphoma is non-Hodgkin's lymphoma; preferably diffuse large B-cell lymphoma and mantle cell lymphoma; more preferably CDK9-related diffuse large B-cell lymphoma and mantle cell lymphoma.
  • the myeloma is multiple myeloma, preferably CDK9-related multiple myeloma.
  • said CDK9 association refers to CDK9 overexpression.
  • the compound 45 or a pharmaceutically acceptable salt, stereoisomer, isotopic derivative or prodrug thereof, is the sole active ingredient in the medicament.
  • the compound 45 or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotopic derivative or a prodrug thereof is used in combination with one or more other targeted drugs or chemotherapeutic drugs for the preparation of the drug.
  • the medicament is formulated into a clinically acceptable formulation.
  • the formulation is an oral formulation or an injectable formulation.
  • said Compound 45 or a pharmaceutically acceptable salt thereof, stereoisomer, isotopic derivative or prodrug thereof, is administered at a daily administered dosage ranging from about 0.001 mg/kg to about 1000 mg/kg; Preferably 0.01 mg/kg to about 100 mg/kg, more preferably 0.02 mg/kg to about 10 mg/kg.
  • the compound 45, or a pharmaceutically acceptable salt thereof, stereoisomer, isotopic derivative or prodrug thereof is administered at a daily dose ranging from about 0.001 mg to about 1000 mg; preferably 0.01 mg to About 100 mg, more preferably 0.1 mg to about 80 mg, further preferably 1 mg to about 70 mg, further preferably 1.5 mg to about 60 mg, further preferably 2 mg to about 50 mg; more preferably 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 8 mg, 10 mg, 12mg, 16mg, 18mg, 20mg, 24mg, 25mg, 30mg, 32mg, 36mg, 40mg, 42mg, 45mg, or 50mg.
  • the dosing frequency is single dose or multiple doses.
  • the dosing frequency is once a day, twice a day, three times a day, once every two days, once every three days, once every four days, once every five days 1. Dosing once every six days or once every seven days.
  • the medicament contains a therapeutically effective amount of Compound 45, or a pharmaceutically acceptable salt, stereoisomer, isotopic derivative, or prodrug thereof.
  • the therapeutically effective amount is 0.001-1000 mg.
  • the therapeutically effective amount is 0.01-100 mg.
  • the therapeutically effective amount is 0.1-50 mg; more preferably 2 mg, 3 mg, 4 mg, 5 mg, 6 mg, 8 mg, 10 mg, 12 mg, 16 mg, 18 mg, 20 mg, 24 mg, 25 mg, 30 mg, 32 mg , 36mg, 40mg, 42mg, 45mg, or 50mg.
  • the single dosage form of the drug contains 0.01-100 mg of compound 45 or its pharmaceutically acceptable salt, stereoisomer, isotope derivative or prodrug; preferably 0.1-80 mg; more preferably 1-60mg; more preferably 1-50mg; further preferably 2-20mg; further preferably 2-15mg; further preferably 2-10mg; further preferably 2-8mg; further preferably 2-6mg; -5 mg; more preferably 2-4 mg; more preferably 2-3 mg.
  • the compound 45 or a pharmaceutically acceptable salt, stereoisomer, isotopic derivative or prodrug thereof, is administered in a single dose or in divided doses.
  • the medicament is administered orally or by injection. In a preferred embodiment, the medicament is administered orally.
  • a pharmaceutical composition which comprises compound 45 or its pharmaceutically acceptable salt, stereoisomer, isotope derivative or prodrug as described in the fourth aspect, and any Optionally, a pharmaceutically acceptable carrier is included.
  • the pharmaceutical composition is used to treat hematological tumors.
  • the compound 45 or a pharmaceutically acceptable salt, stereoisomer, isotopic derivative or prodrug thereof, is the sole active ingredient in the pharmaceutical composition.
  • the pharmaceutical composition further comprises one or more other targeted drugs or chemotherapeutic drugs as active ingredients.
  • the pharmaceutical composition is formulated into a clinically acceptable formulation.
  • the formulation is an oral formulation or an injectable formulation.
  • the pharmaceutical composition contains a therapeutically effective amount of Compound 45, or a pharmaceutically acceptable salt, stereoisomer, isotopic derivative, or prodrug thereof.
  • the therapeutically effective amount is 0.001-1000 mg.
  • the therapeutically effective amount is 0.01-100 mg.
  • the therapeutically effective amount is 0.1-50 mg.
  • the single dosage form of the pharmaceutical composition contains 0.01-100 mg of the compound represented by formula (I) or its pharmaceutically acceptable salt, stereoisomer, isotope derivative or prodrug; preferably 0.1-80mg; more preferably 1-60mg; further preferably 1-50mg; further preferably 2-20mg; further preferably 2-15mg; further preferably 2-10mg; -6mg; more preferably 2-5mg; further preferably 2-4mg; further preferably 2-3mg.
  • the pharmaceutical composition is administered orally or by injection. In a preferred embodiment, the pharmaceutical composition is administered orally.
  • a method of treating a hematological tumor in an individual comprising administering to the individual a therapeutically effective amount of compound 45 as described in the fourth aspect or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotope derivative or a prodrug thereof, or compound 45 as described in the fourth aspect or a pharmaceutically acceptable salt thereof, a stereoisomer, an isotope derivative or a prodrug thereof pharmaceutical composition.
  • the compound of the present application has optical activity, and the compound of the present application can be a racemate, or an optical isomer or a mixture thereof, and the synthesis of the optical isomer in the compound of the present application can be achieved by the formation of the optical isomer It can also be prepared from the starting material, and can also be prepared by separation of the racemate.
  • ethyl is “optionally” substituted with halogen , meaning that the ethyl group can be unsubstituted ( CH2CH3 ), monosubstituted (eg CH2CH2F ), polysubstituted (eg CHFCH2F , CH 2 CHF 2 etc.) or fully substituted (CF 2 CF 3 ). It will be appreciated by those skilled in the art that for any group containing one or more substituents, no sterically impossible and/or synthetically impossible substitution or substitution pattern is introduced.
  • C mn herein, is that the moiety has an integer number of carbon atoms in the given range.
  • C 1-6 means that the group can have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms.
  • variable e.g, R
  • R any variable
  • its definition is independent at each occurrence. So, for example, if a group is substituted by 2 R's, each R has independent options.
  • alkyl refers to a monovalent saturated aliphatic hydrocarbon group, a linear or branched group containing 1-20 carbon atoms, preferably containing 1-10 carbon atoms (i.e. C 1-10 alkyl), more preferably Contains 1-8 carbon atoms (C 1-8 alkyl), more preferably contains 1-6 carbon atoms (ie C 1-6 alkyl), for example "C 1-6 alkyl” refers to the group is an alkyl group, and the number of carbon atoms in the carbon chain is between 1 and 6 (specifically 1, 2, 3, 4, 5 or 6).
  • Examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, neopentyl, 1,1-dimethyl Propyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, n-heptyl , N-octyl, etc.
  • cycloalkyl refers to a monocyclic saturated aliphatic hydrocarbon group having a specific number of carbon atoms, preferably containing 3-12 carbon atoms (i.e. C 3-12 cycloalkyl), more preferably containing 3-10 carbon atoms (C 3-10 cycloalkyl), more preferably 3-6 carbon atoms (C 3-6 cycloalkyl), 4-6 carbon atoms (C 4-6 cycloalkyl), 5-6 carbon atoms (C 5-6 cycloalkyl).
  • Examples include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methylcyclopropyl, 2-ethyl-cyclopentyl, dimethylcyclobutyl, and the like.
  • alkoxy refers to an -O-alkyl group as defined above, that is, containing 1-20 carbon atoms, preferably containing 1-10 carbon atoms, preferably 1-8 carbon atoms , more preferably 1 to 6 carbon atoms (specifically 1, 2, 3, 4, 5 or 6).
  • Representative examples include, but are not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy, 1-methylpropoxy, 2-methylpropoxy, tert-butoxy, pentyloxy, Oxygen, 1-methylbutoxy, 2-methylbutoxy, 3-methylbutoxy, 1,1-dimethylpropoxy, 1,2-dimethylpropoxy, 2 , 2-dimethylpropoxy, 1-ethylpropoxy, etc.
  • halogen refers to F, Cl, Br, I.
  • haloalkyl refers to an alkyl group as defined above in which one, two or more hydrogen atoms or all hydrogen atoms are replaced by halogen.
  • Representative examples of haloalkyl include CCl3 , CF3 , CHCl2, CH2Cl , CH2Br , CH2I , CH2CF3 , CF2CF3 , and the like .
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic, bicyclic or polycyclic cyclic hydrocarbon substituent, which is a non-aromatic structure, containing 3-20 ring atoms, of which 1, 2, 3 or more One ring atom is selected from N, O or S, and the remaining ring atoms are C.
  • It preferably contains 3-12 ring atoms (C 3-12 heterocyclyl), further preferably contains 3-10 ring atoms (C 3-10 heterocyclyl), or 3-8 ring atoms (C 3-8 heterocyclyl) ring group), or 3 to 6 ring atoms (C 3-6 heterocyclyl), or 4 to 6 ring atoms (C 4-6 heterocyclyl), or 5 to 6 ring atoms (C 5-6 heterocyclyl).
  • the number of heteroatoms is preferably 1-4, more preferably 1-3 (ie 1, 2 or 3).
  • Examples of monocyclic heterocyclyl groups include pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, dihydropyrrolyl, piperidinyl, piperazinyl, pyranyl, and the like.
  • Polycyclic heterocyclyls include spiro, fused and bridged heterocyclyls.
  • heterocycloalkyl means a saturated “heterocyclyl” as defined above, containing 3 to 20 ring atoms, of which 1, 2, 3 or more ring atoms are selected from N, O or S , and the remaining ring atoms are C.
  • It preferably contains 3-12 ring atoms (C 3-12 heterocycloalkyl), further preferably contains 3-10 ring atoms (C 3-10 heterocycloalkyl), or 3-8 ring atoms (C 3- 8 heterocycloalkyl), or 3-7 ring atoms (C 3-7 heterocycloalkyl), or 3-6 ring atoms (C 3-6 heterocycloalkyl), or 4-6 ring atoms (C 4-6 heterocycloalkyl), or 5-6 ring atoms (C 5-6 heterocycloalkyl).
  • the number of heteroatoms is preferably 1-4, more preferably 1-3 (ie 1, 2 or 3).
  • Examples include aziridinyl, oxiranyl, thietidinyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, tetrahydrofuryl, oxa Cyclohexane, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, dioxanyl, dithianyl, oxazolidinyl, thiazolidinyl, pyrazolidinyl, imidazolidinyl Wait.
  • aryl denotes monocyclic, bicyclic and tricyclic aromatic carbocyclic ring systems containing 6-16 carbon atoms, or 6-14 carbon atoms, or 6-12 carbon atoms, or 6-10 carbon atoms , preferably 6-10 carbon atoms, the term “aryl” may be used interchangeably with the term “aromatic ring”.
  • aryl groups may include, but are not limited to, phenyl, naphthyl, anthracenyl, phenanthrenyl, or pyrenyl, and the like.
  • heteroaryl means an aromatic monocyclic or polycyclic ring system containing a 5-12-membered structure, or preferably a 5-10-membered structure, a 5-8-membered structure, and more preferably a 5-6-membered structure, wherein one, 2, 3 or more ring atoms are heteroatoms and the remaining atoms are carbon, the heteroatoms are independently selected from O, N or S, and the number of heteroatoms is preferably 1, 2 or 3.
  • heteroaryl examples include, but are not limited to, furyl, thienyl, oxazolyl, thiazolyl, isoxazolyl, oxadiazolyl, thiadiazolyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl , tetrazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, thiodiazolyl, triazinyl, phthalazinyl, quinolinyl, isoquinolinyl, pteridyl, purinyl, indyl Indolyl, Isoindolyl, Indazolyl, Benzofuryl, Benzothienyl, Benzopyridyl, Benzopyrimidinyl, Benzopyrazinyl, Benzimidazolyl, Benzophthalazinyl, Pyrrole A[2,3
  • pharmaceutically acceptable salt or “pharmaceutically acceptable salt” means, within the scope of sound medical judgment, suitable for use in contact with the tissues of mammals, especially humans, without undue toxicity, irritation, allergic reaction, etc. and with reasonable benefits. /risk ratio with a grain of salt.
  • salt includes salts prepared from inorganic acids. If the compound of the present application is acidic, the pharmaceutically acceptable non-toxic bases include salts prepared with inorganic bases and organic bases.
  • stereoisomer refers to isomers produced by different arrangements of atoms in space, including configuration isomers and conformational isomers, wherein configuration isomers include geometric isomers isomers (or cis-trans isomers) and optical isomers (including enantiomers and diastereomers).
  • Geometric isomers may exist in the compounds of the present application.
  • the compounds of the present application may contain a carbon-carbon double bond or a carbon-nitrogen double bond of E or Z configuration, wherein the term “E” represents a higher order substituent on the opposite side of the carbon-carbon or carbon-nitrogen double bond, and the term “Z” represents a higher order substituent on the same side of a carbon-carbon or carbon-nitrogen double bond (determined using the Cahn-Ingold Prelog priority rule).
  • the compounds of the present application may also exist as a mixture of "E” and "Z” isomers. Substituents around a cycloalkyl or heterocycloalkyl group are referred to as cis or trans configurations.
  • Optical isomers refer to substances with exactly the same molecular structure, similar physical and chemical properties, but different optical activity.
  • the compounds of the present application may contain asymmetrically substituted carbon atoms in the R or S configuration, wherein the terms "R” and "S” are as in IUPAC 1974 Recommendations for Section E, Fundamental Stereochemistry, Pure Appl.Chem. (1976) 45, 13-10 as defined.
  • Compounds with asymmetrically substituted carbon atoms are racemic at those carbon atoms. Atoms having an excess of one configuration (relative to the other) such that that configuration is present in a higher amount, preferably an excess of about 85% to 90%, more preferably an excess of about 95% to 99%, and still more preferably an excess of greater than about 99% .
  • the present application includes racemic mixtures, relative and absolute stereoisomers and mixtures of relative and absolute stereoisomers.
  • isotopic derivatives may also be referred to as “isotopic forms", which means that the compounds of the present application may exist in isotope-labeled or enriched forms, containing one or more atoms whose atomic weight or mass number is different from that in nature The atomic mass or mass number of the most abundant atom found in .
  • Isotopes can be radioactive or non-radioactive isotopes.
  • Isotopes of atoms such as hydrogen, carbon, phosphorus, sulfur, fluorine, chlorine, and iodine include, but are not limited to: 2 H, 3 H, 13 C, 14 C, 15 N, 18 O, 32 P, 35 S, 18 F, 36 Cl and 125 I.
  • Compounds containing other isotopes of these and/or other atoms are within the scope of this application; deuterated forms are preferred.
  • the isotopically labeled compound contains a deuterium ( 2 H), tritium ( 3 H) or 14 C isotope.
  • the isotope-labeled compounds of the present application can be prepared using general methods well known to those of ordinary skill in the art.
  • relevant literature includes: Lizondo, J et al, Drugs Fut, 21(11), 1116(1996); Brickner, S J et al, J Med Chem, 39(3), 673(1996); Mallesham, B et al, Org Lett, 5(7), 963 (2003).
  • drugs containing non-radioactive isotopes such as deuterated drugs known as "heavy drugs" can be used to treat related diseases and conditions.
  • Increasing the amount of an isotope present in the above compounds above its natural abundance is called enrichment.
  • Examples of enriched amounts include about 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 16, 21, 25, 29, 33, 37, 42, 46, 50, 54, 58, 63, 67, 71, 75, 79, 84, 88, 92, 96 to about 100 mol%.
  • any possible position in the molecular structure can be replaced by an isotope to obtain an isotope derivative.
  • deuterium ( 2 H) can be substituted at any possible site in the molecule to give a deuterated form of the derivative.
  • Drugs labeled with stable isotopes can alter the physicochemical properties of the drug, such as pKa and lipid solubility. If isotopic substitutions affect regions involved in ligand-receptor interactions, these effects and changes can affect the pharmacodynamic response of drug molecules. Although some physical properties of stable isotope-labeled molecules differ from those of unlabeled molecules, the chemical and biological properties are the same, with one important difference being that due to the increased mass of the heavy isotope, Any bond to another atom is stronger than the same bond between the light isotope and that atom.
  • prodrug or “prodrug” is a designed derivative of an active drug that improves some identified, undesirable physical or biological property. Physical properties are usually related to solubility (too high or insufficient lipid or water solubility) or stability, while problematic biological properties include too fast metabolism or poor bioavailability, which may themselves be related to physicochemical properties.
  • Prodrugs are usually prepared by a) forming esters, half-esters, carbonates, nitrates, amides, hydroxamic acids, carbamates, imines, Mannich bases, phosphates, phosphate esters and Enamines, b) functionalization of drugs with azo, glycoside, peptide and ether functional groups, c) use of aminals, hemiaminals, polymers, salts, complexes, phosphoramides, acetals, hemiaminals of drugs Acetal and ketal forms.
  • Esters can be prepared from substrates containing hydroxyl or carboxyl groups using general methods known to those skilled in the art. A typical reaction in these compounds is the substitution of one heteroatom with another.
  • Amides can be prepared in a similar manner from substrates containing amino or carboxyl groups. Esters can also react with amines or ammonia to form amides. Another way to prepare amides is to heat carboxylic acids and amines together.
  • relapse refers to a condition in which cancer cells reappear in a subject or mammal that has achieved cancer remission following treatment.
  • refractory refers to a condition in which, despite intensive treatment, residual cancer cells remain in the subject or mammal.
  • lymphoma refers to a heterogeneous population of tumors arising in the reticuloendothelium and lymphatic system. It includes non-Hodgkin's lymphoma (NHL).
  • NHL non-Hodgkin's lymphoma
  • non-Hodgkin's lymphoma refers to a monoclonal proliferation of lymphoid cells in sites of the immune system, including lymph nodes, bone marrow, spleen, liver and gastrointestinal tract. Examples of NHL include, but are not limited to, diffuse large B-cell lymphoma, mantle cell lymphoma, and the like.
  • myeloma refers to a tumor made up of the type of cells normally found in the bone marrow. Examples include, but are not limited to, multiple myeloma.
  • treating means administering a compound or formulation described herein to ameliorate or eliminate a disease or one or more symptoms associated with the disease, and includes: (i) inhibiting a disease or disease state, i.e. curbing its development; (ii) remission of a disease or disease state, even if the disease or disease state regresses.
  • terapéuticaally effective amount means that amount of a compound of the present application that (i) treats a particular disease, condition or disorder, or (ii) alleviates, ameliorates or eliminates one or more symptoms of a particular disease, condition or disorder.
  • the amount of a compound of the present application that constitutes a “therapeutically effective amount” varies depending on the compound, the disease state and its severity, the mode of administration, and the individual characteristics (e.g., sensitivity, body weight, age, etc.) of the individual to be treated, However, it can be routinely determined by those skilled in the art based on their own knowledge and the present disclosure.
  • pharmaceutical composition refers to a mixture of one or more compounds of the present application or pharmaceutically acceptable salts, stereoisomers or prodrugs thereof and pharmaceutically acceptable auxiliary materials.
  • the purpose of the pharmaceutical composition is to facilitate administration of a compound of the present application, or a pharmaceutically acceptable salt, stereoisomer or prodrug thereof, to an organism.
  • the pharmaceutical composition of the present invention can be prepared by conventional methods in the art.
  • the term "pharmaceutically acceptable carrier” or “excipient” or “pharmaceutically acceptable adjuvant” or “pharmaceutically acceptable adjuvant” means no significant irritation to the organism, and Those excipients that do not impair the biological activity and performance of the active compound.
  • pharmaceutically acceptable excipients includes: solvents, propellants, solubilizers, co-solvents, emulsifiers, colorants, binders, disintegrants, fillers, lubricants, wetting agents, osmotic pressure regulators, Stabilizers, glidants, flavoring agents, preservatives, suspending agents, coating materials, fragrances, anti-adherents, antioxidants, chelating agents, penetration enhancers, pH regulators, buffers, plasticizers Agents, surfactants, foaming agents, defoamers, thickeners, inclusion agents, humectants, absorbents, diluents, flocculants and deflocculants, filter aids, release retardants, etc.
  • the compounds of the present application can be prepared by other synthetic methods, including the specific embodiments listed below, the embodiments formed by combining them with other chemical synthesis methods, and those known to those skilled in the art
  • preferred implementations include but are not limited to the examples of the present application.
  • the term "individual” or “subject” refers to a cell or a mammal, such as a human being, but can also be other mammals, such as livestock or laboratory animals and the like.
  • the application has evaluated the compound shown in the formula (I) for different subtypes of CDK In vitro kinase inhibitory activity and proliferation inhibitory activity of various myeloma, lymphoma and leukemia cell lines, and further evaluated the inhibitory effect of the compound represented by formula (I) on tumor growth for leukemia tumor xenograft models.
  • IC 50 is below 300nM, preferably below 200nM, more preferably below 100nM, more preferably below tens of nM, compound 45 has a certain activity on RPMI08826 cells, MM.1S cells, SU-DHL-4 cells and Jeko-1 cells Significant inhibitory activity, IC 50 below 100nM.
  • results of the in vivo test show that, compared with the reference compound, the compound of the present application has better anti-tumor effect in vivo, is better tolerated, and has a higher possibility of becoming a drug, which provides a better choice for inhibiting CDK9 target drugs.
  • Embodiment 6 is a diagrammatic representation of Embodiment 6
  • 6b (250mg, 0.52mmol) was dissolved in dichloromethane (10mL), then trifluoroacetic acid (1mL) was added, and reacted at room temperature for 1.5 hours. TLC followed and monitored that no raw material remained. Add saturated sodium bicarbonate (30mL) to the reaction solution, extract with dichloromethane (20mL ⁇ 3), combine the organic phases, wash with saturated sodium chloride (20mL ⁇ 2), dry over anhydrous sodium sulfate, and remove the solvent under reduced pressure to obtain 6c (130 mg, 65% yield).
  • Embodiment 7 is a diagrammatic representation of Embodiment 7:
  • Embodiment 8 is a diagrammatic representation of Embodiment 8
  • Embodiment 9 is a diagrammatic representation of Embodiment 9:
  • 2-Methoxyphenylboronic acid (0.42 g, 2.77 mmol) was dissolved in diethylene glycol dimethyl ether (30 mL) and water (6 mL), followed by the addition of the compound 5-fluoro-4-iodopyridin-2-amine ( 0.60 g, 2.52 mmol), [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (95 mg, 0.13 mmol) and potassium carbonate (1.04 g, 7.56 mmol). The system was protected with nitrogen, stirred at 100° C., and reacted for 4 hours. TLC monitored that there was no residue in the raw material reaction.
  • 2,6-Difluoropyridine-3-boronic acid 550 mg, 3.14 mmol
  • compound 5-fluoro-4-iodopyridin-2-amine 898 mg, 3.77 mmol
  • 1,4-dioxane 15 mL
  • sequentially added [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride (219mg, 0.30mmol)
  • potassium carbonate (1.30g, 9.42mmol
  • 23a (806mg, 1.74mmol) was dissolved in dichloromethane (10mL), then 5mL of trifluoroacetic acid was added in an ice-water bath, and the whole system was stirred at room temperature for 2 hours, and no raw material was detected by TLC.
  • Dissolve 24a 400mg, 0.86mmol
  • dichloromethane 25mL
  • trifluoroacetic acid 3mL
  • TLC monitors that there is no remaining raw material
  • the aqueous phase was extracted with dichloromethane (30 mL ⁇ 3), and the organic phases were combined and dried over anhydrous sodium sulfate.
  • 26a (0.30 g, 1.18 mmol) was dissolved in N,N-dimethylformamide (30 mL), followed by the addition of (1S,3R)-3-[(tert-butoxycarbonyl)amino]cyclohexanecarboxylic acid (0.35 g, 1.42mmol), 2-(7-benzotriazole oxide)-N,N,N',N'-tetramethyluronium hexafluorophosphate (0.54g, 1.42mmol) and N,N-di Methylethylamine (0.30 g, 2.36 mmol), the whole system was stirred overnight at room temperature, and no raw material was found by TLC.
  • 26b (0.20g, 0.42mmol) was dissolved in dichloromethane (30mL), then trifluoroacetic acid (2mL) was added under an ice-water bath, the whole system was stirred overnight at room temperature, and TLC was detected until no raw material remained.
  • 26c (0.11 g, 0.29 mmol) was dissolved in dichloromethane (35 mL), followed by the addition of methanesulfonyl chloride (0.40 g, 0.35 mmol) and triethylamine (44 mg, 0.46 mmol). The system was stirred at room temperature, followed by TLC monitoring until no raw material remained.
  • 2,2'-Dibromodiethyl ether (12.85 g, 55.60 mmol) was dissolved in N,N-dimethylformamide (10 mL), followed by the addition of methyl cyanoacetate (5.00 g, 50.45 mmol) and 1,8 - Diazabicyclo[5.4.0]undec-7-ene (11.50 g, 75.68 mmol), replaced with nitrogen three times, and kept the whole system under nitrogen atmosphere. The system was stirred at 85° C. and reacted for 4 hours. TLC monitored that there was no remaining raw material.
  • 35a (2.37g, 14.00mmol) was dissolved in ethanol (36mL) and water (5mL), then sodium hydroxide (2.24g, 56.00mmol) was added, and the whole system was stirred at room temperature for 4 hours, and no raw material was detected by TLC.
  • 35b (123 mg, 0.79 mmol) was dissolved in N,N-dimethylformamide (5 mL), followed by the addition of (1S,3R)-3-amino-N-(5-fluoro-4-(4-fluoro- 2-methoxyphenyl)pyridin-2-yl)cyclohexylamide (260mg, 0.72mmol), 2-(7-azobenzotriazole)-N,N,N',N'-tetramethyl Urea hexafluorophosphate (548mg, 1.44mmol) and N,N-diisopropylethylamine (357 ⁇ L, 2.16mmol), the whole system was stirred at room temperature, and reacted for 10 hours.
  • Triphosgene (33 mg, 0.11 mmol) was dissolved in dichloromethane (2 mL), followed by dropwise addition of a solution of 5b (61 mg, 0.17 mmol) and triethylamine (22 mg, 0.22 mmol) dissolved in dichloromethane (2 mL), The whole system was stirred at room temperature and reacted for 2 hours, then methanol solution (2 mL) was added, and the reaction was continued for 3 hours, monitored by TLC until no raw material remained.
  • 60a (623mg, 1.09mmol) was dissolved in dichloromethane (20mL), then trifluoroacetic acid (5mL) was added, and the whole system was stirred at room temperature for 2 hours, and no raw material was detected by TLC.
  • Dissolve 5b (0.07g, 0.19mmol) in N,N-dimethylformamide (10mL), add cyclopropylacetic acid (0.02g, 0.21mmol), 2-(7-azobenzotriazole )-N,N,N',N'-tetramethyluronium hexafluorophosphate (0.09g, 0.23mmol) and N,N-diisopropylethylamine (0.05g, 0.38mmol), react at room temperature for 8 hours , TLC monitors that there is no remaining raw material.
  • 26c (99 mg, 0.26 mmol) was dissolved in N,N-dimethylformamide (5 mL), followed by the addition of 1-cyano-1-cyclopropanecarboxylic acid (44 mg, 0.40 mmol), 2-(7- Nitrobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (152 mg, 0.40 mmol) and N,N-diisopropylethylamine (131 ⁇ L, 0.79 mmol), The whole system was stirred overnight at room temperature, and no raw material was found by TLC.
  • 1-cyano-1-cyclopropanecarboxylic acid 44 mg, 0.40 mmol
  • 2-(7- Nitrobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate 152 mg, 0.40 mmol
  • N,N-diisopropylethylamine 131 ⁇ L,
  • 26c (99 mg, 0.26 mmol) was dissolved in N,N-dimethylformamide (5 mL), followed by the addition of glycolic acid (20 mg, 0.26 mmol), 2-(7-azobenzotriazole)-N , N,N',N'-tetramethyluronium hexafluorophosphate (152mg, 0.40mmol) and N,N-diisopropylethylamine (131 ⁇ L, 0.79mmol), the whole system was stirred overnight at room temperature, TLC Check that there is no remaining raw material.
  • Dissolve 69a (0.03g, 0.07mmol) in absolute ethanol (10mL), add 2.0M ammonia ethanol solution (0.004g, 0.28mmol), heat to 90°C, seal the tube for 4 hours, TLC monitors that there is no remaining raw material .
  • the solvent was evaporated under reduced pressure, the residue was dissolved in absolute ethanol (10 mL), and 40% dichloroacetaldehyde aqueous solution (0.005 g, 0.07 mmol) was added, heated to 90° C. for 8 hours, and there was no remaining raw material as monitored by TLC.
  • 70b (1.17g, 4.83mmol) was dissolved in dichloromethane (30mL), then 10mL of trifluoroacetic acid was added under an ice-water bath, and the whole system was stirred at room temperature for 3 hours. The solvent was removed under reduced pressure to afford 70c (1.50 g) which was used directly in the next step.
  • 71a (0.45g, 0.94mmol) was dissolved in dichloromethane (30mL), then 2mL of trifluoroacetic acid was added in an ice-water bath, and the whole system was stirred at room temperature overnight, and no raw materials were detected by TLC.
  • 71b (0.31g, 0.82mmol) was dissolved in dichloromethane (35mL), then acetic anhydride (0.25g, 2.47mmol) and triethylamine (0.25g, 2.47mmol) were added, and the system was stirred at room temperature, monitored by TLC There are no raw materials left.
  • Add water (50mL) to the reaction solution, extract with dichloromethane (50mL ⁇ 3), combine the organic phases, wash with saturated sodium chloride (50mL ⁇ 2), dry over anhydrous sodium sulfate, remove the solvent under reduced pressure, and perform column chromatography Separation and purification (dichloromethane:methanol 50:1-10:1) gave the final product 71 (0.18g, yield 52%).
  • 6-Bromo-1-methyl-1H-indazole (211mg, 1.00mmol), bis-pinacol diborane (508mg, 2.00mmol), potassium acetate (294mg, 3.00mmol) and [1,1' - Bis(diphenylphosphino)ferrocene]palladium dichloride (73 mg, 0.10 mmol) was dissolved in 1,4-dioxane (20 mL). The reaction system was protected with nitrogen and reacted at 100° C. for 4 hours. TLC detected that there was no remaining raw material, and the heating was stopped.
  • 74b (0.12g, 0.25mmol) was dissolved in dichloromethane (20ml), 4mL of trifluoroacetic acid was added, and the reaction was carried out at room temperature for 4 hours, and there was no remaining raw material as monitored by TLC. Wash the reaction solution with water (30mL ⁇ 3), combine the aqueous phase, adjust the pH of the aqueous phase to 8-9 with sodium carbonate, extract with dichloromethane (30mL ⁇ 3), combine the organic phase, wash with saturated sodium chloride (50mL ⁇ 2 ), dried over anhydrous sodium sulfate. The solvent was removed by concentration under reduced pressure to give 74c (0.08 g, 84% yield).
  • Test Example 1 The inhibitory effect test of the compound of the present application on CDK9, CDK1, CDK2, CDK4, CDK5, CDK6 and CDK7
  • the compound was diluted with DMSO to 11 concentrations, 3-fold dilution, and the highest concentration of the compound to be tested was 10 ⁇ M.
  • Test Example 2 Inhibitory Effect of CDK9 Inhibitor on MV 4-11 Cell Proliferation in Vitro
  • MTT is thiazolium blue, which is a tetrazolium salt of a dye that can accept hydrogen atoms.
  • Amber dehydrogenase in the mitochondria of living cells can reduce exogenous MTT to insoluble blue-purple crystals and deposit them in cells, while dead cells have no such function.
  • Dimethyl sulfoxide can dissolve the blue-purple complex in the cells, and its light absorption value is measured at a wavelength of 490-550nm with an enzyme-linked immunosorbent detector, which can indirectly reflect the number of cells. Within a certain cell number range, the amount of MTT crystal formation is proportional to the cell number.
  • the OD value can reflect the number of living cells, and its IC 50 value is calculated with SPSS19.0.
  • MTT working solution Weigh 0.5g of MTT and dissolve in 100mL PBS, filter and sterilize with a 0.22 ⁇ m microporous membrane, and store in a 4°C refrigerator (use within two weeks) or -20°C for long-term storage.
  • Suspension cells Count after resuspension by centrifugation. After making a certain density of cell suspension with complete medium, pipette and blow evenly to inoculate in a 96-well plate, 100 ⁇ L per well, and then culture in a CO 2 incubator.
  • MTT method add 20 ⁇ L of MTT to each well, incubate in the incubator for about 4 hours, discard the liquid in the well, add 150 ⁇ L DMSO to each well, place in a shaker for 5-10 minutes, and detect at a wavelength of 550 nm with a microplate reader.
  • the compounds of the present application have a strong inhibitory effect on MV 4-11 cells, and the in vitro cell inhibitory activity is basically below 300nM, and the optimal amount can reach several nM.
  • Test Example 3 In vitro inhibitory effect of compounds on the proliferation of myeloma and lymphoma cells
  • the cell culture environment is 37°C, 5% CO 2 .
  • Inhibition rate (%) (well with normal OD value- well with drug-treated OD value)/(well with normal OD value- blank well with OD value) ⁇ 100%
  • the half inhibitory concentration IC 50 of the drug was calculated by SPSS19.0.
  • RPMI-8826 human multiple myeloma cells
  • MM.1S human multiple myeloma cells
  • SU-DHL-4 human diffuse large B lymphoma cells
  • Jeko-1 human mantle cell lymphoma cells.
  • Test Example 4 In vivo investigation of anti-tumor activity of drugs - in vivo pharmacodynamics of the compound of the present application in human acute myeloid leukemia MV 4-11 cell subcutaneous xenograft tumor model
  • IMDM medium containing 10% fetal bovine serum (FBS), 37°C, 5% CO 2 .
  • NOD-SCID mice female, 6-8 weeks old, weighing about 18-22 grams, were subcutaneously inoculated with 0.1 mL (1 ⁇ 10 8 ) of MV 4-11 cells on the right back of each mouse.
  • administration began, and the dosage and method of administration were shown in the table below.
  • the tumor volume was measured twice a week, and the volume was measured in cubic millimeters.
  • the administration was terminated to compare the difference in average tumor volume between the test compound group and the solvent group.
  • the antitumor efficacy of compounds was evaluated by TGI (%).
  • TGI (%) reflects tumor growth inhibition rate.
  • the solvent composition of the solvent group and the administration group DMSO: HP- ⁇ -CD (0.5g/mL): water ratio is 2%: 20%: 78% (v/v/v)
  • TGI (%) [1-(average tumor volume at the end of certain treatment group administration-average tumor volume at the beginning of this treatment group)/(average tumor volume at the end of solvent control group administration-solvent The average tumor volume at the beginning of the control group)] ⁇ 100%.
  • the compound of the application exhibits good drug efficacy in vivo in the subcutaneous xenograft tumor model of human acute myeloid leukemia MV 4-11 cells, and has significant tumor inhibitory effect.
  • Test Example 5 In vivo investigation of the antitumor activity of the drug - the in vivo drug effect of the compound of the present application in the subcutaneous xenograft tumor model of human promyelocytic acute leukemia HL-60 cells.
  • IMDM medium containing 20% fetal bovine serum (FBS), 37°C, 5% CO 2 ;
  • NU/NU mice female, 6-8 weeks old, weighing about 18-22 grams, each mouse was subcutaneously inoculated with 0.1 mL of HL-60 cell suspension (containing about 1 ⁇ 10 7 cells) in the armpit of the right forelimb.
  • the drug was started, and the dosage and method of administration were shown in the table below.
  • the tumor volume was measured 2-3 times a week, and the volume was measured in cubic millimeters.
  • the administration was terminated to compare the difference in average tumor volume between the test compound group and the solvent group .
  • the antitumor efficacy of compounds was evaluated by TGI (%).
  • TGI (%) reflects tumor growth inhibition rate.
  • the solvent composition of the solvent group and the administration group DMSO: HP- ⁇ -CD (0.5g/mL): water ratio is 2%: 20%: 78% (v/v/v)
  • TGI (%) [1-(average tumor volume at the end of certain treatment group administration-average tumor volume at the beginning of this treatment group)/(average tumor volume at the end of solvent control group administration-solvent The average tumor volume at the beginning of the control group)] ⁇ 100%.
  • the compound of the present application exhibits good drug efficacy in vivo in the subcutaneous xenograft tumor model of human promyelocytic acute leukemia HL-60 cells.
  • the compound of the present application has a significant antitumor effect.
  • Test Example 6 Investigation of hERG Inhibitory Activity in Vitro
  • This test includes the following aspects:
  • hERG currents were recorded using the whole-cell patch clamp technique. Take the cell suspension and add it to the cell tank, and place it on the stage of the upright microscope. After the cells adhered to the wall, they were perfused with extracellular fluid at a flow rate of 1–2 mL/min. The glass microelectrode is drawn in two steps by a microelectrode pulling instrument, and its water resistance value is 2-5M ⁇ . After establishing whole-cell recordings, the clamping potential was maintained at -80mV. Depolarization to +60mV and repolarization to -50mV elicited hERG tail currents when voltage stimulation was given. All recordings were performed after the current was stabilized.
  • the administration of extracellular perfusion starts at a low concentration, and each concentration lasts for 5-10 minutes until the current is stable, and then the next concentration is given.
  • the half maximal inhibitory concentration (IC50 ) of the test compound was obtained by the best fit of the Logistic equation.
  • Amitripyline is one of the most widely used drugs to block hERG current, so it was used as a positive control drug in this study.
  • the IC 50 result of the active control drug Amitriptyline on the inhibition of hERG current is consistent with the historical results of the test party, and also consistent with the results reported in the literature, indicating that the results of this test are credible.
  • the above test results show that the tested compound cannot achieve half inhibition of hERG current at the highest test concentration, so the IC50 cannot be measured, indicating that the compound of the present application has no obvious inhibitory effect on the hERG channel within the detection concentration range of this test. To a certain extent, it can reflect that the compound of the application has low or no cardiotoxicity, which has positive significance for drug safety evaluation.
  • mice ICR mice (5 weeks), male and female
  • the solvent composition of the solvent group and the administration group DMSO: HP- ⁇ -CD (0.5g/mL): water ratio is 2%: 20%: 78% (v/v/v)
  • Test method ICR mice were divided into 7 groups according to weight balance, with 5 males and 5 mice in each group.
  • the method of administration is intragastric administration, once a day, for 7 consecutive days, and the dosage and results are shown in the table below.

Abstract

式(I)所示的化合物或其可药用盐、其立体异构体、同位素衍生物或前药作为周期蛋白依赖性激酶9(CDK9)抑制剂在治疗血液肿瘤,特别是白血病、骨髓瘤和淋巴瘤中的各种应用,包括相关的制药用途、药物组合物或治疗方法等。

Description

周期蛋白依赖性激酶9抑制剂的用途
相关申请
本申请要求于2021年5月24日提交的中国专利申请202110582506.8号的优先权,通过引用的方式将该申请的全部内容整体并入本文,用于所有目的。
技术领域
本申请涉及医药领域,具体而言,涉及一种周期蛋白依赖性激酶9(CDK9)抑制剂或其可药用盐、其立体异构体、同位素衍生物或前药,包含其的药物组合物,或含有其的药物的用途,特别是该抑制剂或其可药用盐、其立体异构体、同位素衍生物或前药,包含其的药物组合物,或含有其的药物在制备治疗血液肿瘤疾病,特别是恶性血液肿瘤的药物中的用途。
背景技术
细胞周期蛋白依赖性激酶(CDKs)是一类丝氨酸/苏氨酸蛋白激酶,在调节细胞周期和转录中起关键作用。截至目前,已知约有20余种人类CDK亚型以及约30种细胞周期伴侣蛋白(Cao et al.2014),这些CDKs可以被细胞周期蛋白所激活,发挥着不同的生物学功能,CDK按照功能可分为两种,一种控制细胞周期,一种调节细胞转录。例如,CDK1、2、3、4和6直接干预细胞周期;CDK5不调节细胞周期,但在有丝分裂后神经元复杂迁移中起到关键作用;CDK7间接充当这些CDK的激活剂;CDK9仅在细胞转录中起作用,而不参与细胞周期的调节。
CDK9是转录CDKs亚家族的重要成员,该家族是一组激酶,其功能是控制真核RNA聚合酶II(Pol II)合成和加工mRNA的主要步骤。CDK9存在于所有的哺乳动物细胞内,体内CDK9的激活取决于其与对应的细胞周期蛋白(Cyclin T/K)的结合,形成异源二聚体,即正性转录延长因子b(P-TEFb)。当负性转录延长因子(NELF、NELFs)参与细胞转录的负性调节时,转录被抑制,P-TEFb被招募到负性转录延长因子抑制转录延长的体系中,催化RNA聚合酶II的碳末端结构域(CTD)磷酸化,同时催化NELFs的SPT5亚基和NELF的RD亚基磷酸化,致使负性转录延长因子从转录复合物上脱离,从而使转录得以继续。
肿瘤通常是由于细胞周期蛋白依赖性激酶抑制物(CDKI)表达缺失或者细胞周期蛋白的过量表达使细胞不受调控而过度增殖所导致的。鉴于上述调控机制,使用CDK9抑制剂,将能够阻止P-TEFb对RNA聚合酶II的碳末端结构域磷酸化,进一步阻碍NEFL的离去,加强负抑制作用,引起转录停止,使得细胞内mRNA及半衰期短的蛋白的水平快速下降,从而可以引起肿瘤细胞的凋亡。CDK9已成为开发有效癌症治疗的潜在蛋白靶标,近来已有制药公司对CDK9抑制剂用于癌症的治疗开展了研究,例如,阿斯利康的AZD4573和拜尔公司的BAY-1251152,均处于临床I期试验阶段,尚未有CDK9抑制剂获批上市。
尽管目前已公开了一些CDK9抑制剂小分子(例如,WO2009047359、WO2014076091等),仍有必要开发兼具良好药效、良好安全性的新化合物,使临床上更多患者受益。
发明内容
在一方面,本申请提供一种如式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药在用于制备治疗血液肿瘤,特别是恶性血液肿瘤的药物中的用途。
在另一方面,本申请提供一种如式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药在用于制备治疗白血病、淋巴瘤或骨髓瘤,特别是CDK9相关的白血病、淋巴瘤或骨髓瘤的药物中的用途。
在另一方面,本申请提供一种CDK9抑制剂化合物45或其可药用盐、其立体异构体、同位素衍生物或前药在用于制备治疗血液肿瘤,特别是恶性血液肿瘤的药物中的用途。
在另一方面,本申请提供一种CDK9抑制剂化合物45或其可药用盐、其立体异构体、同位素衍生物或前药在用于制备治疗白血病、淋巴瘤或骨髓瘤,特别是CDK9相关的白血病、淋巴瘤或骨髓瘤的药物中的用途。
在另一方面,本申请提供一种用于治疗血液肿瘤的药物组合物,其包含一种如式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药。
在另一方面,本申请提供一种用于治疗白血病、淋巴瘤和骨髓瘤的药物组合物,其包含一种如式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药。
在另一方面,本申请提供一种用于治疗血液肿瘤的药物组合物,其包含一种CDK9抑制剂化合物45或其可药用盐、其立体异构体、同位素衍生物或前药的药物。
在另一方面,本申请提供一种用于治疗白血病、淋巴瘤和骨髓瘤的药物组合物,其包含一种CDK9抑 制剂化合物45或其可药用盐、其立体异构体、同位素衍生物或前药的药物。
本申请的发明人先前的工作开发了具有周期蛋白依赖性激酶9(CDK9)抑制剂活性的新颖化合物(参见公布号WO2021/115335A1或申请号PCT/CN2020/134966,通过引用的方式将该申请的全部内容整体并入本文,用于所有目的)。在此基础上,本申请的发明人对该化合物的医药用途进行了更深入的研究,并得到了本申请的各项发明。
具体而言,在本申请的第一方面中,提供了一种如式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药在用于制备治疗血液肿瘤的药物中的用途,
Figure PCTCN2022094496-appb-000001
其中,
X选自Cl、F,其中优选F;
R 1选自取代或未取代的芳基,或取代或未取代的杂芳基;R 1中的所述“取代”是指被1个、2个、3个、4个或5个各自独立地选自-F、-Cl、-Br、-NH 2、-OH、-SH、-CN、-NO 2、-N 3、-C≡CH、-COOH、-R 3、-(CH 2) wO(CH 2) nR 3、-(CH 2) wNH(CH 2) nR 3、-(CH 2) wNR 3(CH 2) nR 4、-(CH 2) wS(CH 2) nR 3、-(CH 2) wC(O)(CH 2) nR 3、-(CH 2) wC(O)O(CH 2) nR 3、-(CH 2) wOC(O)(CH 2) nR 3、-(CH 2) wC(O)NH(CH 2) nR 3、-(CH 2) wNHC(O)(CH 2) nR 3、-(CH 2) wC(O)NR 3(CH 2) nR 4、-(CH 2) wNR 3C(O)(CH 2) nR 4、-(CH 2) wOS(O) 2(CH 2) nR 3或-(CH 2) wS(O) 2O(CH 2) nR 3的基团所取代;其中,w、n每次出现时各自独立地选自0、1、2、3或4;R 3和R 4分别独立地选自取代或未取代的芳基,取代或未取代的杂芳基,取代或未取代的C 1-6烷基,取代或未取代的C 1-6卤代烷基,取代或未取代的C 2-6烯基,取代或未取代的C 2-6炔基,取代或未取代的C 1-6烷氧基,取代或未取代的C 1-6卤代烷氧基,取代或未取代的环烷基,取代或未取代的杂环烷基,或者当R 3、R 4共同连接至同一个氮原子时,R 3、R 4和共同连接的氮原子组成取代或未取代的杂环烷基;R 3和R 4中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-NH 2、-OH、-SH、-CN、-NO 2、-N 3、-C≡CH、-COOH、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基等的基团所取代;
A环选自取代或未取代的环烷基、取代或未取代的杂环烷基;A环中的所述“取代”是指被1个、2个、3个、4个或5个各自独立地选自-F、-Cl、-Br、OH、NH 2、SH、CN、NO 2、-N 3、-C≡CH、COOH、R 5、OR 5、-NHR 5、-NR 5R 6、-SR 5、-NHCOR 5、-CONHR 5、-NHS(O) 2R 5、-S(O) 2NHR 5、-NR 5S(O) 2R 6、-S(O) 2NR 5R 6的基团所取代,或A环结构中1个、2个或多个-CH 2-基团可任选地被-C(O)-基团替代;其中,R 5和R 6独立地为C 1-6烷基、C 1-6卤代烷基。
R 2选自H、R 7、-(CH 2) xR 7、-(CH 2) xNH(CH 2) yR 7、-(CH 2) xO(CH 2) yR 7、-(CH 2) xNR 7(CH 2) yR 8、-(CH 2) xC(O)(CH 2) yH、-(CH 2) xC(O)(CH 2) yR 7、-(CH 2) xS(O) 2(CH 2) yR 7、-(CH 2) xC(O)C(O)(CH 2) yR 7、-(CH 2) xS(O) 2NH 2、-(CH 2) xNHS(O) 2H、-(CH 2) xS(O) 2NH(CH 2) yR 7、-(CH 2) xNHS(O) 2(CH 2) yR 7、-(CH 2) xS(O) 2NR 7(CH 2) yR 8、-(CH 2) xNR 7S(O) 2(CH 2) yR 8、-(CH 2) xC(O)O(CH 2) yR 7、-(CH 2) xOC(O)(CH 2) yR 7、-(CH 2) xC(O)NH 2、-(CH 2) xNHC(O)H、-(CH 2) xC(O)NH(CH 2) yR 7、-(CH 2) xNHC(O)(CH 2) yR 7、-(CH 2) xC(O)NR 7(CH 2) yR 8或-(CH 2) xNR 7C(O)(CH 2) yR 8;其中,1个、2个或多个-CH 2-基团可任选地被-C(O)-基团替代;x、y每次出现时各自独立地选自0、1、2、3或4;
R 7和R 8独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、-R 10-NH-R 9、-R 10-C(O)-R 9、-R 10-NHC(O)-R 9、-R 10-C(O)NH-R 9、-R 10-S-R 9、-R 10-S(O)-R 9、-R 10-S-C(O)-R 9、环烷基、杂环烷基、芳基、杂芳基、-R 10-芳基、-R 10-杂芳基、-O-R 10-芳基、-O-R 10-杂芳基、-R 10-O-芳基、-R 10-O-杂芳基、-环烷基-芳基、-环烷基-杂芳基、-杂环烷基-芳基、-杂环烷基-杂芳基、C 2-6烯烃以及C 2-6炔烃,或者当R 7和R 8共同连接至同一个氮原子时,R 7和R 8与共同连接的氮原子组成取代或未取代的杂环烷基;其中R 9为C 1-6烷基,R 10为C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-NH 2、-SH、-CN、-NO 2、-N 3、-C≡CH、-COOH、C 1-6烷基、C 1-6烷氧基、C 1-6卤代烷基、C 1-6卤代烷氧基、-NHCN、-NHCONH 2、NHC(O)CH 3、N(CH 3) 2、N(C 2H 5) 2、-SC(O)CH 3、-OC(O)-C 1-6烷基等的基团所取代;
优选地,所述同位素衍生物为氘代形式的衍生物。
本申请中关于“芳基”、“杂芳基”、“环烷基”、“杂环烷基”的定义如下文“定义”部分。
在一个实施方案中,芳基优选包含6~10个碳原子,环烷基优选包含3~6个碳原子,杂芳基优选为5~10元杂芳基,且杂环烷基优选为3~8元杂环基;杂芳基或杂环烷基优选含有1个、2个或3个各自独立地选自N、O或S的杂原子,其余为碳原子。
关于本申请中的“w”、“n”、“x”、“y”,如前所述可各自独立地选自0、1、2、3或4,当“w”和“n”、“x”和“y”同时出现在一个基团中时,具体来说,“w”和“n”、“x”和“y”的数值组合可选自(0,0)、(0,1)、(0,2)、 (0,3)、(0,4)、(1,0)、(1,1)、(1,2)、(1,3)、(1,4)、(2,0)、(2,1)、(2,2)、(2,3)、(2,4)、(3,0)、(3,1)、(3,2)、(3,3)、(3,4)、(4,0)、(4,1)、(4,2)、(4,3)、(4,4),该数值组合适用于R 1、R 2定义中的每一个相关基团,举例而言,R 1定义中的-(CH 2) wO(CH 2) nR 3相当于公开了-OR 3、-OCH 2R 3、-O(CH 2) 2R 3、-O(CH 2) 3R 3、-O(CH 2) 4R 3、-CH 2OR 3、-CH 2OCH 2R 3、-CH 2O(CH 2) 2R 3、-CH 2O(CH 2) 3R 3、-CH 2O(CH 2) 4R 3、-(CH 2) 2OR 3、-(CH 2) 2OCH 2R 3、-(CH 2) 2O(CH 2) 2R 3、-(CH 2) 2(CH 2) 3R 3、-(CH 2) 2O(CH 2) 4R 3、-(CH 2) 3OR 3、-(CH 2) 3OCH 2R 3、-(CH 2) 3O(CH 2) 3R 3、-(CH 2) 3O(CH 2) 3R 3、-(CH 2) 3O(CH 2) 4R 3、-(CH 2) 4OR 3、-(CH 2) 4OCH 2R 3、-(CH 2) 4O(CH 2) 4R 3、-(CH 2) 4O(CH 2) 3R 3、-(CH 2) 4O(CH 2) 4R 3等基团,与之相同,R 1、R 2中的其他相关基团也公开了这样的选择,不再一一赘述。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中,A环选自取代或未取代的4-6元环烷基,取代或未取代的4-6元杂环烷基,或取代或未取代的5-6环烷基,或取代未取代的5-6元杂环烷基;进一步优选地,A环选自取代或未取代的5-6环烷基,或取代未取代的5-6元杂环烷基;更进一步优选地,A环选自取代或未取代的5-6环烷基。
在一个实施方案中,A环选自环己烷基、四氢吡咯基、哌啶基、哌嗪基、环戊烷基或吗啉基,进一步优选地,A环选自环己烷基、环戊烷基或四氢吡咯基。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中A环中的所述“取代”是指被1个、2个、3个、4个或5个各自独立地选自-F、-Cl、-Br、OH、NH 2、SH、CN、R 5、OR 5的基团所取代,其中,R 5为C 1-6烷基或C 1-6烷氧基,R 5可进一步选自C 1-4烷基或C 1-4烷氧基。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其具有式(II)所示结构:
Figure PCTCN2022094496-appb-000002
其中,R 1、R 2和X如式(I)所示。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其具有式(IIa)所示结构:
Figure PCTCN2022094496-appb-000003
其中,R 1、R 2和X如式(I)所示。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其具有式(III)所示结构:
Figure PCTCN2022094496-appb-000004
其中,R 1、R 2和X如式(I)所示。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其具有式(IIIa)所示结构:
Figure PCTCN2022094496-appb-000005
Figure PCTCN2022094496-appb-000006
其中,R 1、R 2和X如式(I)所示。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中,R 1选自取代或未取代的6-10元芳基,或取代或未取代的5-10元杂芳基;所述杂芳基含有1或2个各自独立地选自N或O的杂原子;所述取代基的数量选自1、2或3个。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中,R 1选自取代或未取代的苯环、吡啶环、吲哚环、吲唑环、苯并呋喃环、吡咯并吡啶环;进一步优选取代或未取代的苯环、吡啶环、吲哚环、苯并呋喃环、吡咯并吡啶环;进一步优选取代的苯环、吡啶环和未取代的吲哚环、苯并呋喃环、吡咯并吡啶环;更进一步优选取代的苯环。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中,所述R 1上的取代基分别独立地选自-F、-Cl、-OH、-NH 2、-R 3、-(CH 2) wO(CH 2) nR 3或-(CH 2) wOC(O)(CH 2) nR 3;w和n每次出现时各自独立的选自0、1或2,其中,R 3定义如式(I)所述。优选地,所述R 1上的取代基分别独立地选自-F-、-Cl、-OH、-R 3、-(CH 2) wO(CH 2) nR 3;进一步优选地,所述R 1上的取代基分别独立地选自-F-、-OH、-R 3、-(CH 2) wO(CH 2) nR 3。例如,当R 1为取代的苯环时,取代基选自-F、-OH或烷氧基,优选1或2个氟原子取代和1个-OH或烷氧基取代,优选1或2个氟原子取代和1个烷氧基取代。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其具有式(II)所示结构:
Figure PCTCN2022094496-appb-000007
其中,R 1为取代的苯环,取代基选自-F、-OH或烷氧基;优选1或2个氟原子取代和1个-OH或烷氧基取代,优选1或2个氟原子取代和1个烷氧基取代;
R 2和X如式(I)所示。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其具有式(IIa)所示结构:
Figure PCTCN2022094496-appb-000008
其中,R 1为取代的苯环,取代基选自-F、-OH或烷氧基;优选1或2个氟原子取代和1个-OH或烷氧基取代,优选1或2个氟原子取代和1个烷氧基取代;
R 2和X如式(I)所示。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中,R 3和R 4分别独立地选自取代或未取代的6元芳基,取代或未取代的5-6元杂芳基,取代或未取代的C 1-3烷基,取代或未取代的C 1-3烷氧基,取代或未取代的C 3-6环烷基,取代或未取代的C 3-6杂环烷基,或者当R 3、R 4共同连接至同一个氮原子时,R 3、R 4和共同连接的氮原子组成3-7元取代或未取代的杂环烷基;所述杂环烷基含有1个或2独立地选自N、O或S的杂原子;R 3和R 4中所述的“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-CH 3、-C 2H 5、-OCH 3、-OC 2H 5的取代基所取代;优选地,R 3和R 4分别独立地选自取代或未取代的C 1-3烷基、取代或未取代的C 1-3烷氧基。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中,R 3和R 4分别独立地选自取代或未取代的苯环、吡啶环、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、丙氧基、异丙氧基、环丙基、环丁基、环戊基或环己基,R 3和R 4中所述的“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-CH 3、-C 2H 5、-OCH 3、-OC 2H 5的取代基所取代。优选地,R 3和R 4分别独立地选自取代或未取代的甲基、乙基、异丙基、甲氧基、乙氧基、异丙氧基、环丙基、吡啶环;进一步优选地,R 3和R 4分别独立地选自取代或未取代的甲基、乙基、异丙基、甲氧基、乙氧基、异丙氧基;更进一步优选地,R 3和R 4分别独立地选自取代或未取代的甲基、甲氧基。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其 中,R 2选自R 7、-(CH 2) xR 7、-(CH 2) xNH(CH 2) yR 7、-(CH 2) xC(O)(CH 2) yR 7、-(CH 2) xS(O) 2(CH 2) yR 7、-(CH 2) xC(O)C(O)(CH 2) yR 7、-(CH 2) xC(O)O(CH 2) yR 7、-(CH 2) xC(O)NH(CH 2) yR 7、-(CH 2) xC(O)NR 7(CH 2) yR 8或-(CH 2) xNR 7C(O)(CH 2) yR 8,其中,所述R 7和R 8定义同式(I)。进一步优选地,R 2选自R 7、-(CH 2) xR 7、-(CH 2) xC(O)(CH 2) yR 7、-(CH 2) xS(O) 2(CH 2) yR 7、-(CH 2) xC(O)C(O)(CH 2) yR 7、-(CH 2) xC(O)O(CH 2) yR 7、-(CH 2) xC(O)NH(CH 2) yR 7;更进一步优选地,R 2选自R 7、-(CH 2) xR 7、-(CH 2) xC(O)(CH 2) yR 7;更进一步优选地,R 2选自-(CH 2) xC(O)(CH 2) yR 7
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中,R 7和R 8各自独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、-R 10-NH-R 9、-R 10-C(O)-R 9、-R 10-NHC(O)-R 9、-R 10-C(O)NH-R 9、-R 10-S-R 9、-R 10-S-C(O)-R 9、C 3-6环烷基、3-6元杂环烷基、C 6-10芳基、5-10元杂芳基、-R 10-C 6-10芳基、-R 10-5-10元杂芳基、-O-R 10-C 6-10芳基、-O-R 10-5-10元杂芳基、-R 10-O-C 6-10芳基、-R 10-O-5-10元杂芳基、C 2-6烯烃以及C 2-6炔烃,或者当R 7和R 8共同连接至同一个氮原子时,R 7和R 8与共同连接的氮原子组成取代或未取代的3-6元杂环烷基;其中R 9为C 1-6烷基,R 10为C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-NH 2、-SH、-CN、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基、C 1-3卤代烷氧基、-NHCN、-NHCONH 2、NHC(O)CH 3、N(CH 3) 2、N(C 2H 5) 2、-SC(O)CH 3、-OC(O)-C 1-6烷基等的基团所取代;优选地,R 7独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、-R 10-NHC(O)-R 9、C 3-6环烷基、3-6元杂环烷基、C 6-10芳基、5-10元杂芳基、C 2-6烯烃以及C 2-6炔烃,或者当R 7和R 8共同连接至同一个氮原子时,R 7和R 8与共同连接的氮原子组成取代或未取代的3-6元杂环烷基;进一步优选地,R 7独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、C 3-6环烷基、3-6元杂环烷基、C 6-10芳基、5-10元杂芳基;进一步优选地,R 7独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、C 3-6环烷基、3-6元杂环烷基;进一步优选地,R 7独立地选自取代或未取代的R 9;R 7中所述的“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-NH 2、-SH、-CN、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基、C 1-3卤代烷氧基、-NHCN、-NHCONH 2、NHC(O)CH 3、N(CH 3) 2、N(C 2H 5) 2、-SC(O)CH 3、-OC(O)-C 1-6烷基等的基团所取代;优选地,R 7中所述的“取代”是指1个、2个或3个各自独立地选自-F、-OH、-NH 2、-SH、-CN、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基、NHC(O)CH 3、N(CH 3) 2、-OC(O)-C 1-6烷基等的基团所取代;进一步优选地,R 7中所述的“取代”是指1个、2个或3个各自独立地选自-F、-OH、-NH 2、-CN、C 1-3烷基、C 1-3烷氧基等的基团所取代;进一步优选地,R 7中所述的“取代”是指1个、2个或3个各自独立地选自-OH、-CN等的基团所取代;进一步优选地,R 7中所述的“取代”是指1个各自独立地选自-OH、-CN等的基团所取代;进一步优选地,R 7中所述的“取代”是指1个各自独立地选自-OH基团所取代。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中,所述R 7和R 8各自独立地选自取代或未取代的甲基、乙基、丙基、异丙基、丁基、戊基、甲氧基、乙氧基、丙氧基、异丙氧基、-CH 2OCH 3、-CH 2OCH 2CH 3、-CH 2CH 2OCH 3、-CH 2CH 2OCH 2CH 3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氮杂环戊基、氮杂环己基、氧杂环丙基、氧杂环丁基、氧杂环戊基、氧杂环己基、苯基、吡啶基、吡唑基、噁唑基、异噁唑基、噻吩基、噻唑基、苯甲基、苯乙基、乙烯基、丙烯基、乙炔基或丙炔基。优选地,R 7独立地选自取代或未取代的甲基、乙基、甲氧基、乙氧基、环丙基、环丁基、氮杂环丁基、氮杂环己基、氧杂环丁基、氧杂环己基、苯基、吡啶基、吡唑基、异噁唑基、噻吩基、噻唑基、苯甲基、乙烯基、丙烯基或乙炔基;进一步优选地,R 7独立地选自取代或未取代的甲基、乙基、甲氧基、环丙基、环丁基、氮杂环己基、氧杂环丁基、氧杂环己基、吡啶基、吡唑基、异噁唑基、乙烯基、丙烯基或乙炔基;进一步优选地,R 7独立地选自取代或未取代的甲基、乙基、环丙基、氮杂环己基、氧杂环丁基、吡唑基、乙烯基、丙烯基或乙炔基;更进一步优选地,R 7独立地选自取代或未取代的甲基、乙基、环丙基;更进一步优选地,R 7独立地选自取代或未取代的甲基。R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-NH 2、-SH、-CN、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基、C 1-3卤代烷氧基、-NHCN、-NHCONH 2、NHC(O)CH 3、N(CH 3) 2、N(C 2H 5) 2、-SC(O)CH 3、-OC(O)-C 1-6烷基等的基团所取代;优选地,R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-OH、-SH、-CN、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷氧基、NHC(O)CH 3、N(CH 3) 2、N(C 2H 5) 2、-OC(O)-C 1-6烷基等的基团所取代;优选地,R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-OH、-NH 2、-CN、C 1-3卤代烷氧基等的基团所取代;进一步优选地,R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-OH、-CN等的基团所取代;进一步优选地,R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-OH、-CN等的基团所取代;进一步优选地,R 7和R 8中的所述“取代”是指被1个各自独立地选自-OH、-CN等的基团所取代;进一步优选地,R 7和R 8中的所述“取代”是指被1个各自独立地选自-OH基团所取代。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其中,R 9为C 1-4烷基,R 10为C 1-4亚烷基。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,其 中,所述化合物具有如下结构:
Figure PCTCN2022094496-appb-000009
Figure PCTCN2022094496-appb-000010
Figure PCTCN2022094496-appb-000011
Figure PCTCN2022094496-appb-000012
在一个实施方案中,所述血液肿瘤为恶性血液肿瘤,优选为复发或难治性血液肿瘤。
在一个实施方案中,所述血液肿瘤选自白血病、淋巴瘤和骨髓瘤,优选为复发或难治性白血病、淋巴瘤和骨髓瘤。
在一个实施方案中,所述白血病、淋巴瘤或骨髓瘤是指CDK9相关的白血病、淋巴瘤或骨髓瘤。
在一个实施方案中,所述白血病为急性白血病,优选为急性髓性白血病,进一步优选为复发或难治性急性髓性白血病,进一步优选为CDK9相关的复发或难治性急性髓性白血病。
在一个实施方案中,所述血液肿瘤选自复发或难治性急性髓性白血病、淋巴瘤和骨髓瘤;优选为淋巴瘤和骨髓瘤;进一步优选为复发或难治性淋巴瘤和骨髓瘤。
在一个实施方案中,所述淋巴瘤为非霍奇金淋巴瘤,优选为弥漫大B细胞淋巴瘤或套细胞淋巴瘤,进一步优选CDK9相关的弥漫大B细胞淋巴瘤或套细胞淋巴瘤。
在一个实施方案中,所述骨髓瘤为多发性骨髓瘤,优选为CDK9相关的多发性骨髓瘤。
在一个实施方案中,所述CDK9相关是指CDK9过量表达。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药是所述药物中的唯一的活性成分。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药与一种或多种其它靶向药或化疗药联合使用。
在一个实施方案中,所述药物被制成临床接受的制剂。在一个优选的实施方案中,所述制剂是口服制剂或注射制剂。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药以从约0.001mg/kg至约1000mg/kg的每日给药剂量范围给予;优选0.01mg/kg至约100mg/kg,进一步优选0.02mg/kg至约10mg/kg。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药以从约0.001mg至约1000mg的每日给药剂量范围给予;优选0.01mg至约100mg,进一步优选0.1mg至约80mg,进一步优选1mg至约70mg,进一步优选1.5mg至约60mg,进一步优选2mg至约50mg;进一步优选为2mg、3mg、4mg、5mg、6mg、8mg、10mg、12mg、16mg、18mg、20mg、24mg、25mg、30mg、32mg、36mg、40mg、42mg、45mg、或50mg。给药频率为单次给药或多次给药。优选地,给药频率为每天给药一次、每天给药两次、每天给药三次、每两天给药一次、每三天给药一次、每四天给药一次、每五天给药一次、每六天给药一次或每七天给药一次。
在一个实施方案中,所述药物含有治疗有效量的式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药。在一个优选的实施方案中,所述治疗有效量是0.001-1000mg。在一个优选的实施方案中,所述治疗有效量是0.01-100mg。在一个优选的实施方案中,所述治疗有效量是0.1-50mg;进一步优选为2mg、3mg、4mg、5mg、6mg、8mg、10mg、12mg、16mg、18mg、20mg、24mg、25mg、30mg、32mg、36mg、40mg、42mg、45mg、或50mg。
在一个实施方案中,所述药物的单一剂量形式中含有0.01-100mg的式(I)所示化合物或其药学上可接受的盐、其立体异构体、同位素衍生物或前药;优选为0.1-80mg;进一步优选为1-60mg;进一步优选为1-50mg;进一步优选为2-20mg;进一步优选为2-15mg;进一步优选为2-10mg;进一步优选为2-8mg;进一步优选为2-6mg;进一步优选为2-5mg;进一步优选为2-4mg;进一步优选为2-3mg。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药以单剂量给予或分剂量给予。
在一个实施方案中,所述药物通过口服给予或注射给予。在一个优选的实施方案中,所述药物通过口服给予。
在本申请的第二方面中,提供了用于治疗个体(例如受试者或患者)的血液肿瘤的方法,所述方法包括给予所述个体治疗有效量的一种如第一方面所述的式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,或者包含一种如第一方面所述的式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药的药物。
在本申请的第三方面中,提供了一种药物组合物,其包含如第一方面所述的式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,以及任选地包含药学上可接受的载体。在具体实施方案中,所述药物组合物用于治疗血液肿瘤。
在一个实施方案中,所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药是所述药物组合物中的唯一的活性成分。
在一个实施方案中,所述药物组合物中还包含一种或多种其它靶向药或化疗药作为活性成分。
在一个实施方案中,所述药物组合物被制成临床接受的制剂。在一个优选的实施方案中,所述制剂是口服制剂或注射制剂。
在一个实施方案中,所述药物组合物含有治疗有效量的式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药。在一个优选的实施方案中,所述治疗有效量是0.001-1000mg。在一个优选的实施方案中,所述治疗有效量是0.01-100mg。在一个优选的实施方案中,所述治疗有效量是0.1-50mg。
在一个实施方案中,所述药物组合物的单一剂量形式中含有0.01-100mg的式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药;优选为0.1-80mg;进一步优选为1-60mg;进一步优选为1-50mg;进一步优选为2-20mg;进一步优选为2-15mg;进一步优选为2-10mg;进一步优选为2-8mg;进一步优选为2-6mg;进一步优选为2-5mg;进一步优选为2-4mg;进一步优选为2-3mg。
在一个实施方案中,所述药物组合物通过口服给予或注射给予。在一个优选的实施方案中,所述药物 组合物通过口服给予。
在不存在矛盾和冲突的情况下,第一方面所述的技术方案或技术特征均适用于第二方面和第三方面。
在本申请的第四方面中,提供了本申请的化合物45或其可药用盐、其立体异构体、同位素衍生物或前药在用于制备治疗血液肿瘤的药物中的用途,
Figure PCTCN2022094496-appb-000013
在一个实施方案中,所述血液肿瘤为恶性血液肿瘤,优选为复发或难治性血液肿瘤。
在一个实施方案中,所述血液肿瘤选自白血病、淋巴瘤和骨髓瘤,优选为复发或难治性白血病、淋巴瘤和骨髓瘤。
在一个实施方案中,所述白血病、淋巴瘤或骨髓瘤是指CDK9相关的白血病、淋巴瘤或骨髓瘤。
在一个实施方案中,所述白血病为急性白血病,优选为急性髓性白血病,进一步优选为复发或难治性急性髓性白血病,进一步优选为CDK9相关的复发或难治性急性髓性白血病。
在一个实施方案中,所述血液肿瘤选自复发或难治性急性髓性白血病、淋巴瘤和骨髓瘤;优选为淋巴瘤和骨髓瘤;进一步优选为复发或难治性淋巴瘤和骨髓瘤。
在一个实施方案中,所述淋巴瘤为非霍奇金淋巴瘤;优选为弥漫大B细胞淋巴瘤和套细胞淋巴瘤;进一步优选为CDK9相关的弥漫大B细胞淋巴瘤和套细胞淋巴瘤。
在一个实施方案中,所述骨髓瘤为多发性骨髓瘤,优选为CDK9相关的多发性骨髓瘤。
在一个实施方案中,所述CDK9相关是指CDK9过量表达。
在一个实施方案中,所述化合物45或其可药用盐、其立体异构体、同位素衍生物或前药是所述药物中的唯一的活性成分。
在一个实施方案中,所述化合物45或其可药用盐、其立体异构体、同位素衍生物或前药与一种或多种其它靶向药或化疗药联合用于制备所述药物。
在一个实施方案中,所述药物被制成临床接受的制剂。在一个优选的实施方案中,所述制剂是口服制剂或注射制剂。
在一个实施方案中,所述化合物45或其可药用盐、其立体异构体、同位素衍生物或前药以从约0.001mg/kg至约1000mg/kg的每日给药剂量范围给予;优选0.01mg/kg至约100mg/kg,进一步优选0.02mg/kg至约10mg/kg。
在一个实施方案中,所述化合物45或其可药用盐、其立体异构体、同位素衍生物或前药以从约0.001mg至约1000mg的每日给药剂量范围给予;优选0.01mg至约100mg,进一步优选0.1mg至约80mg,进一步优选1mg至约70mg,进一步优选1.5mg至约60mg,进一步优选2mg至约50mg;进一步优选为2mg、3mg、4mg、5mg、6mg、8mg、10mg、12mg、16mg、18mg、20mg、24mg、25mg、30mg、32mg、36mg、40mg、42mg、45mg、或50mg。给药频率为单次给药或多次给药。优选地,给药频率为每天给药一次、每天给药两次、每天给药三次、每两天给药一次、每三天给药一次、每四天给药一次、每五天给药一次、每六天给药一次或每七天给药一次。
在一个实施方案中,所述药物含有治疗有效量的化合物45或其可药用盐、其立体异构体、同位素衍生物或前药。在一个优选的实施方案中,所述治疗有效量是0.001-1000mg。在一个优选的实施方案中,所述治疗有效量是0.01-100mg。在一个优选的实施方案中,所述治疗有效量是0.1-50mg;进一步优选为2mg、3mg、4mg、5mg、6mg、8mg、10mg、12mg、16mg、18mg、20mg、24mg、25mg、30mg、32mg、36mg、40mg、42mg、45mg、或50mg。
在一个实施方案中,所述药物的单一剂量形式中含有0.01-100mg的化合物45或其可药用盐、其立体异构体、同位素衍生物或前药;优选为0.1-80mg;进一步优选为1-60mg;进一步优选为1-50mg;进一步优选为2-20mg;进一步优选为2-15mg;进一步优选为2-10mg;进一步优选为2-8mg;进一步优选为2-6mg;进一步优选为2-5mg;进一步优选为2-4mg;进一步优选为2-3mg。
在一个实施方案中,所述化合物45或其可药用盐、其立体异构体、同位素衍生物或前药以单剂量给予或分剂量给予。
在一个实施方案中,所述药物通过口服给予或注射给予。在一个优选的实施方案中,所述药物通过口服给予。
在本申请的第五方面中,提供了一种药物组合物,其包含如第四方面所述的化合物45或其可药用盐、 其立体异构体、同位素衍生物或前药,以及任选地包含药学上可接受的载体。在具体实施方案中,所述药物组合物用于治疗血液肿瘤。
在一个实施方案中,所述化合物45或其可药用盐、其立体异构体、同位素衍生物或前药是所述药物组合物中的唯一的活性成分。
在一个实施方案中,所述药物组合物中还包含一种或多种其它靶向药或化疗药作为活性成分。
在一个实施方案中,所述药物组合物被制成临床接受的制剂。在一个优选的实施方案中,所述制剂是口服制剂或注射制剂。
在一个实施方案中,所述药物组合物含有治疗有效量的化合物45或其可药用盐、其立体异构体、同位素衍生物或前药。在一个优选的实施方案中,所述治疗有效量是0.001-1000mg。在一个优选的实施方案中,所述治疗有效量是0.01-100mg。在一个优选的实施方案中,所述治疗有效量是0.1-50mg。
在一个实施方案中,所述药物组合物的单一剂量形式中含有0.01-100mg的式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药;优选为0.1-80mg;进一步优选为1-60mg;进一步优选为1-50mg;进一步优选为2-20mg;进一步优选为2-15mg;进一步优选为2-10mg;进一步优选为2-8mg;进一步优选为2-6mg;进一步优选为2-5mg;进一步优选为2-4mg;进一步优选为2-3mg。
在一个实施方案中,所述药物组合物通过口服给予或注射给予。在一个优选的实施方案中,所述药物组合物通过口服给予。
在本申请的第六方面中,提供了一种治疗个体(例如受试者或患者)的血液肿瘤的方法,所述方法包括给予所述个体治疗有效量的如第四方面所述的化合物45或其可药用盐、其立体异构体、同位素衍生物或前药,或者包含如第四方面所述的化合物45或其可药用盐、其立体异构体、同位素衍生物或前药的药物组合物。
在不存在矛盾和冲突的情况下,第四方面所述的技术方案或技术特征适用于第五方面和第六方面。
本申请化合物具有光学活性,本申请化合物既可以是外消旋体,也可以是光学异构体或其混合物,本申请化合物中的光学异构体的合成,既可以通过光学异构体的起始原料制备,也可以通过消旋体分离制备。
定义
除非另有说明,本申请中所用的下列术语具有下列含义。一个特定的术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,而应该按照本领域普通的含义去理解。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。
术语“任选”或“任选地”是指随后描述的事件或情况可以发生或不发生,该描述包括发生所述事件或情况和不发生所述事件或情况。例如,乙基“任选”被卤素取代,指乙基可以是未被取代的(CH 2CH 3)、单取代的(如CH 2CH 2F)、多取代的(如CHFCH 2F、CH 2CHF 2等)或完全被取代的(CF 2CF 3)。本领域技术人员可理解,对于包含一个或多个取代基的任何基团,不会引入任何在空间上不可能存在和/或不能合成的取代或取代模式。
本文中的C m-n,是该部分具有给定范围中的整数个碳原子。例如“C 1-6”是指该基团可具有1个碳原子、2个碳原子、3个碳原子、4个碳原子、5个碳原子或6个碳原子。
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2个R所取代,则每个R都有独立的选项。
术语“烷基”指一价饱和脂肪族烃基团,包含1-20个碳原子的直链或支链基团,优选包含1-10个碳原子(即C 1-10烷基),进一步优选包含1-8个碳原子(C 1-8烷基),更优选包含1-6个碳原子(即C 1-6烷基),例如“C 1-6烷基”指的是该基团为烷基,且碳链上的碳原子数量在1-6之间(具体地为1个、2个、3个、4个、5个或6个)。实例包括但不限于甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、新戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、正庚基、正辛基等。
术语“环烷基”指的是具有特定碳原子数的单环饱和脂烃基,优选地包含3-12个碳原子(即C 3-12环烷基),更优选包含3-10个碳原子(C 3-10环烷基),进一步优选3-6个碳原子(C 3-6环烷基)、4-6个碳原子(C 4-6环烷基)、5-6个碳原子(C 5-6环烷基)。实例包括但不限于环丙基、环丁基、环戊基、环己基、甲基环丙基、2-乙基-环戊基、二甲基环丁基等。
术语“烷氧基”指-O-烷基,所述烷基的定义同上,即包含1-20个碳原子,优选地,包含1-10个碳原子,较佳地1-8个碳原子,更佳地1~6个碳原子(具体地为1个、2个、3个、4个、5个或6个)。代表的例子包括但不限于甲氧基、乙氧基、丙氧基、异丙氧基、丁氧基、1-甲基丙氧基、2-甲基丙氧基、叔丁氧基、戊氧基、1-甲基丁氧基、2-甲基丁氧基、3-甲基丁氧基、1,1-二甲基丙氧基、1,2-二甲基丙氧基、2,2-二甲基丙氧基、1-乙基丙氧基等。
术语“卤素”或“卤代”是指F、Cl、Br、I。术语“卤代烷基”是指如上所定义的烷基中一个、两个或多个 氢原子或全部氢原子被卤素取代。卤代烷基的代表性例子包括CCl 3、CF 3、CHCl 2、CH 2Cl、CH 2Br、CH 2I、CH 2CF 3、CF 2CF 3等。
术语“杂环基”指饱和或部分不饱和单环、双环或多环环状烃取代基,为非芳香结构,包含3-20个环原子,其中1个、2个、3个或更多个环原子选自N、O或S,其余环原子为C。优选包含3-12个环原子(C 3-12杂环基),进一步优选包含3-10个环原子(C 3-10杂环基),或3~8个环原子(C 3-8杂环基),或3~6个环原子(C 3-6杂环基),或4~6个环原子(C 4-6杂环基),或5~6个环原子(C 5-6杂环基)。杂原子优选1-4个,更优选1~3个(即1个、2个或3个)。单环杂环基的实例包括吡咯烷基、咪唑烷基、四氢呋喃基、二氢吡咯基、哌啶基、哌嗪基、吡喃基等。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“杂环烷基”是指饱和的如上定义的“杂环基”,包含3-20个环原子,其中1个、2个、3个或更多个环原子选自N、O或S,其余环原子为C。优选包含3-12个环原子(C 3-12杂环烷基),进一步优选包含3-10个环原子(C 3-10杂环烷基),或3-8个环原子(C 3-8杂环烷基),或3-7个环原子(C 3-7杂环烷基),或3-6个环原子(C 3-6杂环烷基),或4-6个环原子(C 4-6杂环烷基),或5-6个环原子(C 5-6杂环烷基)。杂原子优选1-4个,更优选1~3个(即1个、2个或3个)。实例包括氮杂环丙烷基、氧杂环丙烷基、硫杂环丙烷基、氮杂环丁烷基、氧杂环丁烷基、硫杂环丁烷基、吡咯烷基、四氢呋喃基、氧杂环己烷、哌啶基、哌嗪基、吗啉基、硫代吗啉基、二噁烷基、二硫杂环己基、噁唑烷基、噻唑烷基、吡唑烷基、咪唑啉啶等。
术语“芳基”表示含有6-16个碳原子,或6-14个碳原子,或6-12个碳原子,或6-10个碳原子的单环、双环和三环的芳香碳环体系,优选6-10个碳原子,术语“芳基”可以和术语“芳香环”交换使用。芳基基团的实例可以包括但不限于苯基、萘基、蒽基、菲基或芘基等。
术语“杂芳基”表示含有5-12元结构,或优选5-10元结构,5-8元结构,更优选5-6元结构的芳香单环或者多环环状系统,其中1个、2个、3个或更多个环原子为杂原子且其余原子为碳,杂原子独立地选自O、N或S,杂原子数量优选为1个、2个或3个。杂芳基的实例包括但不限于呋喃基、噻吩基、噁唑基、噻唑基、异噁唑基、噁二唑基、噻二唑基、吡咯基、吡唑基、咪唑基、三唑基、四唑基、吡啶基、嘧啶基、吡嗪基、哒嗪基、硫代二唑基、三嗪基、酞嗪基、喹啉基、异喹啉基、喋啶基、嘌呤基、吲哚基、异吲哚基、吲唑基、苯并呋喃基、苯并噻吩基、苯并吡啶基、苯并嘧啶基、苯并吡嗪基、苯并咪唑基、苯并酞嗪基、吡咯并[2,3-b]吡啶基、咪唑并[1,2-a]吡啶基、吡唑并[1,5-a]吡啶基、吡唑并[1,5-a]嘧啶基、咪唑并[1,2-b]哒嗪基、[1,2,4]三唑并[4,3-b]哒嗪基、[1,2,4]三唑并[1,5-a]嘧啶基、[1,2,4]三唑并[1,5-a]吡啶基等。
术语“可药用盐”或“药学上可接受的盐”是指在合理医学判断范围内适用于与哺乳动物特别是人的组织接触而无过度毒性、刺激、过敏反应等并与合理的效益/风险比相称的盐。
术语“盐”包含得自无机酸制备的盐。如果本申请的化合物为酸性的,则药学上可接受的无毒碱包括无机碱及有机碱制备的盐。
术语“立体异构体”是指由分子中原子在空间上排列方式不同所产生的异构体,包括构型异构体和构象异构体,其中,构型异构体又包括几何异构体(或顺反异构体)和光学异构体(包含对映异构体和非对映异构体)。
几何异构体可以存在于本申请的化合物中。本申请的化合物可以含有E或Z构型的碳-碳双键或碳-氮双键,其中术语“E”代表碳-碳或碳-氮双键的对侧的更高顺序取代基,术语“Z”代表碳-碳或碳-氮双键的同侧上的更高顺序取代基(利用Cahn-Ingold Prelog优先规则确定)。本申请的化合物还可以以“E”和“Z”异构体的混合物形态存在。将环烷基或杂环烷基周围的取代基称为顺式或反式构型。
光学异构体指的是分子结构完全相同,物理化学性质相近,但旋光性不同的物质。
本申请的化合物在R或S构型中可以含有不对称取代的碳原子,其中术语“R”和“S”如IUPAC 1974 Recommendations for Section E,Fundamental Stereochemistry,Pure Appl.Chem.(1976)45,13-10所定义。具有不对称取代的碳原子的化合物(具有相等数量的R和S构型)在那些碳原子处是外消旋的。具有过量的一种构型(相对于另一个)的原子使该构型存在更高数量,优选过量大约85%-90%,更优选过量大约95%-99%,更加优选过量大于大约99%。相应地,本申请包括外消旋混合物、相对和绝对立体异构体和相对与绝对立体异构体的混合物。
术语“同位素衍生物”也可称为“同位素形式”,是指本申请的化合物可以以同位素示踪的或富集形式存在,含有一个或多个原子,这些原子的原子量或质量数不同于自然界中发现的最大量的原子的原子量或质量数。同位素可以是放射性或非放射性的同位素。原子例如氢、碳、磷、硫、氟、氯和碘的同位素包括但不局限于: 2H, 3H, 13C, 14C, 15N, 18O, 32P, 35S, 18F, 36Cl和 125I。含有这些和/或其它原子的其它同位素的化合物在本申请范围之内;优选为氘代形式。
在另一个实施方案中,同位素标记的化合物含有氘( 2H)、氚( 3H)或 14C同位素。本申请的同位素标记的化合物可以利用本领域普通技术人员熟知的一般方法来制备。在这方面,相关的文献包括:Lizondo,J et al,Drugs Fut,21(11),1116(1996);Brickner,S J et al,J Med Chem,39(3),673(1996);Mallesham,B et al,Org Lett,5(7),963(2003)。
含有同位素的化合物已经用于药物研究,通过评价非同位素示踪的母体化合物的作用机理和代谢途径,研究化合物的体内代谢结果(Blake et al,J.Pharm.Sci.64,3,367-391(1975))。在安全、有效的治疗药物设计方面,这种代谢研究是重要的,这是因为给予患者的体内活性化合物或因为母体化合物产生的代谢物被证明是毒性的或致癌的(Kushner et al,Can.J.Physiol.Pharmacol.,77,79-88(1999);Foster et al,Advances in Drug Research Vol.14,pp.2-36,Academic press,London,1985;Kato et al,J.Labelled Comp.Radiopharmaceut.,36(10):927-932(1995))。
另外,含有非放射性活性同位素的药物,例如称为“重药物”的氘代药物,可以用于治疗相关的疾病和病症。存在于上面化合物中的同位素的量提高到其天然丰度以上被称作富集。富集的量的例子包括大约0.5,1,2,3,4,5,6,7,8,9,10,12,16,21,25,29,33,37,42,46,50,54,58,63,67,71,75,79,84,88,92,96至大约100mol%。
分子结构中任意可能的位点均可被同位素取代,得到同位素衍生物。例如,在分子中任意可能的位点可被氘( 2H)取代,得到氘代形式的衍生物。
用稳定同位素示踪的药物,可以改变药物的理化性质,例如pKa和脂质溶解度。如果同位素取代影响参与配体-受体相互作用的区域,那么这些效果和改变可以影响药物分子的药效响应。尽管稳定同位素示踪的分子的一些物理性能不同于未示踪的分子的物理性能,但化学和生物学特性是相同的,一种重要的不同是:由于重同位素的质量增加,涉及重同位素和另一个原子的任何键比轻同位素和该原子之间的相同的键更强。相应地,在代谢或酶催转化的位点结合同位素可以使所述反应潜在地减缓,相对于非同位素化合物,可以改变药物动力学特性或效果。术语“前药”或“前体药物”是设计的活性药物的衍生物,其可以改善一些确定的、不合需要的物理或生物学性质。物理性能通常是相关的溶解度(过高或不足的脂质或水溶性)或稳定性,而有问题的生物学特性包括代谢太快或生物利用率差,这本身可能与物理化学性质相关。
前体药物通常如下制备:a)形成活性药物的酯、半酯、碳酸脂、硝酸酯、酰胺、异羟肟酸、氨基甲酸酯、亚胺、曼尼希碱、磷酸盐、磷酸酯和烯胺,b)用偶氮、糖苷、肽和醚官能团使药物功能化,c)使用药物的缩醛胺、半缩醛胺、聚合物、盐、复合物、磷酰胺、乙缩醛、半缩醛和缩酮形式。例如,参见Andrejus Korolkovas’s,“Essentials of Medicinal Chemistry”,John Wiley-Interscience Pulications,John Wiley and Sons,New York(1988),pp.97-118,本文结合其所有内容作为参考。酯可以利用本领域技术人员已知的一般方法,由含有羟基或羧基的底物来制备。这些化合物的典型反应是用另一个原子替代一个杂原子的取代。酰胺可以用类似的方式、由含有氨基或羧基的底物来制备。酯还可以与胺或氨反应,形成酰胺。制备酰胺的另一种方式是将羧酸和胺一起加热。
术语“复发”指这样一种情况,其中治疗后已获得癌症缓解的对象或哺乳动物的癌细胞重现。
术语“难治”指这样一种情况,其中即使经过强化治疗,对象或哺乳动物体内仍有残余的癌细胞。
术语“淋巴瘤”是指网状内皮和淋巴系统中出现的异质肿瘤群。其包括非霍奇金淋巴瘤(NHL)。术语“非霍奇金淋巴瘤”是指在免疫系统部位淋巴样细胞的单克隆增殖,免疫系统部位包括淋巴结、骨髓、脾、肝和胃肠道。NHL的实例包括但不限于弥漫大B细胞淋巴瘤、套细胞淋巴瘤等。
术语“骨髓瘤”是指由在骨髓中正常发现的类型的细胞组成的肿瘤。其实例包括但不限于多发性骨髓瘤。
术语“治疗”意为将本申请所述化合物或制剂进行给药以改善或消除疾病或与所述疾病相关的一个或多个症状,且包括:(i)抑制疾病或疾病状态,即遏制其发展;(ii)缓解疾病或疾病状态,即使该疾病或疾病状态消退。
术语“治疗有效量”意指(i)治疗特定疾病、病况或障碍,或(ii)减轻、改善或消除特定疾病、病况或障碍的一种或多种症状的本申请化合物的用量。构成“治疗有效量”的本申请化合物的量取决于该化合物、疾病状态及其严重性、给药方式以及待被治疗的个体的自身特点(例如,敏感性、体重、年龄等)而改变,但可例行性地由本领域技术人员根据其自身的知识及本公开内容而确定。
术语“药物组合物”是指一种或多种本申请的化合物或其可药用盐、其立体异构体或前药与药学上可接受的辅料组成的混合物。药物组合物的目的是有利于对有机体给予本申请的化合物或其可药用盐、其立体异构体或前药。本发明的药物组合物可以采用本领域的常规方法制备得到。
在本申请的上下文中,术语“药学上可接受的载体”或“赋形剂”或“药学上可接受的辅料”或“药用可接受的辅料”是指对有机体无明显刺激作用,而且不会损害该活性化合物的生物活性及性能的那些辅料。术语“药用可接受的辅料”包括:溶剂、抛射剂、增溶剂、助溶剂、乳化剂、着色剂、黏合剂、崩解剂、填充剂、润滑剂、润湿剂、渗透压调节剂、稳定剂、助流剂、矫味剂、防腐剂、助悬剂、包衣材料、芳香剂、抗黏着剂、抗氧剂、螯合剂、渗透促进剂、pH值调节剂、缓冲剂、增塑剂、表面活性剂、发泡剂、消泡剂、增稠剂、包合剂、保湿剂、吸收剂、稀释剂、絮凝剂与反絮凝剂、助滤剂、释放阻滞剂等。本领域技术人员可根据实际需要选择具体的药用可接受的辅料。有关辅料的知识是本领域技术人员众所周知的,例如可以参考《药剂学》(崔福德主编,第5版,人民卫生出版社,2003)。
词语“包括”、“包含”或“含有”及其相关变体应理解为开放的、非排他性的意义,即“包括但不限于”。
在本申请的范围中,任一特征的各种选项可以与其它特征的各种选项相互组合,从而构成许多不同的实施方案。本申请意欲包括由所有技术特征的各种选项所组成的所有可能的实施方案。
除了下面列举的具体实施方式,本申请的化合物可以通过其他合成方法来制备,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知晓的等同替换方式,优选的实施方式包括但不限于本申请的实施例。
本文所使用的术语“个体”或“对象”是指细胞或哺乳动物,如人类,但也可以是其它哺乳动物,如家畜或实验动物等。
以上实施方案代表了本申请的示例性实施方案,但是本申请并不限于以上实施方案。另外,本申请的以上实施方案中的各个技术特征可以相互组合,从而构成一个或多个新的技术方案,这些新的技术方案也落在本申请的范围内,只要这样的新的技术方案是在技术上可行的即可。
本申请具有以下一种或多种有益效果:
为证明本申请的式(I)所示化合物是对血液瘤(例如,骨髓瘤、淋巴瘤、白血病)有效的CDK9抑制剂,本申请评价了式(I)所示化合物对不同亚型CDK的体外激酶抑制活性及多种骨髓瘤、淋巴瘤和白血病细胞株的增殖抑制活性,并进一步针对白血病肿瘤异种移植模型评价了式(I)所示化合物对肿瘤生长的抑制效果。
体外激酶活性试验和细胞试验结果显示,本申请化合物对CDK9具有良好的体外激酶抑制活性,对其他CDK亚型具有良好的选择性;对白血病细胞系(MV 4-11细胞)、骨髓瘤细胞系(RPMI08826细胞和MM.1S细胞)及淋巴瘤细胞系(SU-DHL-4细胞和Jeko-1细胞)均具有较强的抑制作用,部分化合物在体外对MV 4-11细胞具有明显的抑制活性,IC 50在300nM以下,优选在200nM以下,进一步优选在100nM以下,更进一步优选在几十nM以下,化合物45对RPMI08826细胞、MM.1S细胞、SU-DHL-4细胞和Jeko-1细胞具有明显的抑制活性,IC 50在100nM以下。
体内试验结果表明,与对照化合物相比,本申请化合物具有更好的体内抗肿瘤效果,且耐受性好,成药可能性更高,为抑制CDK9靶点药物提供了更好的选择。
体内外安全性试验结果表明,与对照化合物相比,本申请式(I)所示化合物在检测浓度范围内,对体外hERG通道无明显抑制活性,说明本申请化合物具有较低的心脏毒性风险,且小鼠服药连续7天,随着剂量递增,未见动物死亡,可见动物耐受性良好。
本申请化合物的开发,扩展了治疗癌症药物的选择。
实施例
下面结合具体实施例,进一步阐述本申请。应理解,这些实施例仅用于说明本申请而不用于限制本申请的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件或者按照制造厂商所建议的条件。除非另行定义,文中所使用的所有专业与科学用语与本领域专业人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本申请方法之中。文中所示的较佳实施方法与材料仅做示范之用。
实施例1:
Figure PCTCN2022094496-appb-000014
中间体1a的合成:
将4-溴-5-氯吡啶-2-胺(3.00g,14.50mmol)溶解于乙二醇二甲醚(50mL)和水(10mL)中,接着加入4-氟-2-甲氧基苯硼酸(2.50g,14.70mmol),[1,1'-双(二苯基膦基)二茂铁]二氯化钯(1.06g,1.45mmol)和碳酸钾(6.00g,44.10mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。减压浓缩,经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到1a(3.11g,产率85%)。
中间体1b的合成:
将1a(0.89g,3.50mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环戊烷甲酸(0.85g,3.50mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(1.60g,4.20mmol)和N,N-二异丙基乙胺(0.91g,7.00mmol),整个体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到1b(0.96g,产率59%)。
中间体1c的合成:
将1b(0.96g,2.07mmol)溶解于二氯甲烷(30mL)中,接着冰浴下加入三氟乙酸(2mL),整个体系在室温下搅拌过夜,TLC监测直至原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,用二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1),得到1c(0.66g,产率为88%)。
终产物1的合成:
将1c(0.66g,1.80mmol)溶解于二氯甲烷中(35mL),接着加入乙酸酐(0.92g,9.00mmol)和三乙胺(0.91g,9.00mmol)。体系在室温下搅拌,TLC监测直至原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到到终产物1(0.48g,产率64%)。MS m/z(ESI):406.1[M+H] +
1H NMR(600MHz,CDCl 3)δ9.16(s,1H),8.28-8.25(m,2H),7.55(s,1H),7.18(d,J=7.2Hz,1H),6.79-6.71(m,2H),4.44(s,1H),3.80(s,3H),2.98(t,J=4.8Hz,1H),2.20-2.17(m,3H),1.97(s,3H),1.88-1.82(m,3H).
实施例2:
Figure PCTCN2022094496-appb-000015
中间体2a的合成:
将5-氟-4-碘吡啶-2-胺(1.00g,4.20mmol)溶解于乙二醇二甲醚(20mL)和水(4mL)中,接着加入4-氟-2-甲氧基苯硼酸(0.71g,4.20mmol),[1,1'-双(二苯基膦基)二茂铁]二氯化钯(0.31g,0.42mmol)和碳酸钾(1.70g,12.60mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。减压浓缩,经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到2a(0.80g,产率81%)。
中间体2b的合成:
将2a(0.80g,3.40mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环戊烷甲酸(0.83g,3.40mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(1.56g,4.10mmol)和N,N-二异丙基乙胺(0.88g,6.80mmol),整个体系在室温下搅拌过夜,TLC检测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到2b(0.82g,产率54%)。
中间体2c的合成:
将2b(0.82g,1.83mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入三氟乙酸(2mL),整个体系在室温下搅拌过夜,TLC检测直至原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1)后,得到2c(0.59g,产率93%)。
终产物2的合成:
将2c(0.59g,1.70mmol)溶解于二氯甲烷(35mL)中,接着加入乙酸酐(0.87g,8.50mmol)和三乙胺(0.86g,8.50mmol).体系在室温下搅拌,TLC跟踪监测直至原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经 柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物2(0.42g,产率63%)。MS m/z(ESI):390.2[M+H] +
1H NMR(600MHz,CDCl 3)δ9.20(s,1H),8.32(s,1H),8.12(s,1H),7.30(d,J=6.6Hz,1H),6.82-6.75(m,3H),4.44(s,1H),3.84(s,3H),2.99(q,J=3.6Hz,1H),2.22-2.15(m,3H),1.99(s,3H),1.89-1.85(m,3H).
实施例3:
Figure PCTCN2022094496-appb-000016
中间体3a的合成:
将2a(0.80g,3.40mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入顺-3-[(叔丁氧羰基)氨基]环己基烷甲酸(0.83g,3.40mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(1.56g,4.10mmol)和N,N-二异丙基乙胺(0.88g,6.80mmol),整个体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到3a(0.90g,产率57%)。
中间体3b的合成:
将3a(0.90g,1.95mmol)溶解于二氯甲烷(30mL)中,接着冰浴下加入三氟乙酸(2mL),整个体系在室温下搅拌过夜,TLC监测原料无剩余,向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,用二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1),得到3b(0.61g,产率87%)。
终产物3的合成:
将3b(0.61g,1.69mmol)溶解于二氯甲烷(35mL)中,接着加入乙酸酐(0.86g,8.40mmol)和三乙胺(0.85g,8.40mmol)。体系在室温下搅拌,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物3(0.38g,产率56%)。MS m/z(ESI):404.2[M+H] +
1H NMR(600MHz,CDCl 3) 1H NMR(600MHz,CDCl 3)δ8.88(s,1H),8.29(s,1H),8.10(s,1H),7.28(s,1H),6.77-6.72(m,3H),3.82(s,3H),2.52-2.49(m,1H),2.24-2.22(m,1H),2.00-1.95(m,4H),1.98(s,3H),1.48-1.38(m,3H),1.18-1.13(m,1H).
实施例4:
Figure PCTCN2022094496-appb-000017
中间体4a的合成:
将1a(0.40g,1.58mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1R,3S)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.38g,1.58mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(0.72g,1.90mmol)和N,N-二异丙基乙胺(0.41g,3.16mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到4a(0.45g,产率60%)。
中间体4b的合成:
将4a(0.45g,0.94mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入2mL的三氟乙酸,整个体系在室温下搅拌过夜,TLC检测直至原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1)后,得到4b(0.30g,产率85%)。
终产物4的合成:
将4b(0.30g,0.80mmol)溶解于二氯甲烷(35mL)中,接着加入乙酸酐(0.24g,2.39mmol)和三乙胺(0.24g,2.39mmol),体系在室温下搅拌,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到终产物4(0.17g,产率51%)。MS m/z(ESI):420.14[M+H] +
1H NMR(600MHz,CDCl 3) 1H NMR(600MHz,DMSO-d6)δ10.69(s,1H),8.41(s,1H),8.05(s,1H),7.78-7.73(m,1H),7.27-7.22(m,1H),7.10-7.08(m,1H),6.91-6.88(m,1H),3.76(s,3H),3.57-3.54(m,1H),2.62-2.59(m,1H),1.86(d,J=12.6Hz,1H),1.76(s,6H),1.31-1.23(m,3H),1.07-1.05(m,1H).
实施例5:
Figure PCTCN2022094496-appb-000018
中间体5a的合成:
将2a(0.80g,3.40mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.83g,3.40mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(1.56g,4.10mmol)和N,N-二异丙基乙胺(0.88g,6.80mmol),整个体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到5a(0.90g,产率58%)。
中间体5b的合成:
将5a(0.90g,1.95mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入三氟乙酸(2mL),整个体系在室温下搅拌过夜,TLC检测直至原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,用二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1)后,得到5b(0.61g,产率87%)。
终产物5的合成:
将5b(0.61g,1.69mmol)溶解于二氯甲烷(35mL)中,接着加入乙酸酐(0.86g,8.40mmol)和三乙胺(0.85g,8.40mmol),体系在室温下搅拌,TLC跟踪监测直至原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物5(0.38g,产率56%)。MS m/z(ESI):404.2[M+H] +
1H NMR(600MHz,CD 3OD)δ8.18(s,1H),8.09(s,1H),7.33-7.29(m,1H),6.94(d,J=10.8Hz,1H),6.84-6.81(m,1H),3.83(s,3H),3.76-3.72(m,1H),2.60-2.57(m,1H),2.06(d,J=12.0Hz,1H),1.96-1.90(m,3H),1.93(s,3H),1.51-1.39(m,3H),1.24-1.21(m,1H).
实施例6:
Figure PCTCN2022094496-appb-000019
中间体6a的合成:
将3,4-二氟-2-甲氧基苯硼酸(0.57g,3.03mmol)溶解于二氧六环(50mL)中,接着加入5-氟-4-碘吡啶-2-胺(0.60g,2.52mmol)、四三苯基膦钯(150mg,0.13mmol)和三水合磷酸钾(1.00g,3.78mmol),体系在氮气保护下升温至100℃,反应4小时,TLC跟踪监测原料无剩余。反应液降至室温,粗品经柱层析分离纯化(石油醚:乙酸乙酯=10:1-1:1),得到6a(0.40g,产率52%)。
中间体6b的合成:
将(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(348mg,1.43mmol)溶解于二氯甲烷(50mL)中,接着加入吡啶(572mg,7.24mmol)和二氯亚砜(300mg,2.52mmol)室温反应4小时,然后将6a(400mg,1.57mmol)直接加入到上述反应液中。室温继续反应过夜,TLC跟踪监测原料无剩余。向反应液中加入水(30mL)乙酸乙酯萃取(20mL×3),合并有机相,饱和氯化钠洗涤(20mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到6b(250mg,产率33%)。
中间体6c的合成:
将6b(250mg,0.52mmol)溶解于二氯甲烷(10mL)中,接着加入三氟乙酸(1mL),室温反应1.5小时,TLC跟踪监测原料无剩余。反应液中加入饱和碳酸氢钠(30mL),二氯甲烷萃取(20mL×3),合并有机相,饱和氯化钠洗涤(20mL×2),无水硫酸钠干燥,减压除去溶剂,得到6c(130mg,产率65%)。
终产物6的合成:
将6c(130mg,0.34mmol)溶解于二氯甲烷(10mL)中,接着加入乙酸酐(45mg,0.44mmol),和三乙胺(44mg,0.44mmol),室温反应1.5小时,TLC跟踪监测原料无剩余。反应液中加入饱和碳酸氢钠(30mL),二氯甲烷萃取(20mL×3),合并有机相,饱和氯化钠洗涤(20mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析纯化(二氯甲烷:甲醇=50:1-30:1),得到终产物6(120mg,产率84%)。MS m/z(ESI):422.2[M+H] +
1H NMR(600MHz,DMSO-d6)δ10.68(s,1H),8.43(s,1H),8.20(s,1H),7.78(d,J=7.8Hz,1H),7.33(t,J=7.8Hz,1H),7.21(t,J=7.8Hz,1H),3.95(s,3H),3.59-3.54(m,1H),2.63-2.59(m,1H),1.88-1.84(m,1H),1.80-1.72(m,3H),1.78(s,3H),1.31-1.25(m,3H),1.10-1.04(m,1H).
实施例7:
Figure PCTCN2022094496-appb-000020
中间体7a的合成:
将2-氨基-5-氟-4-碘吡啶(0.50g,2.10mmol)和5-氟-2-乙氧基苯硼酸(0.46g,2.50mmol)溶于乙二醇二甲醚(10mL)和水(2mL)中,加入[1,1’-双(二苯基膦基)二茂铁]二氯化钯(71mg,0.10mmol),碳酸钾(0.87g,6.30mmol)。用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下反应2小时。TLC监控显示无原料剩余。降温后,浓缩除去溶剂。粗品用柱层析进行分离纯化(石油醚:乙酸乙酯=10:1- 2:1),得到7a(0.50g,产率95%)。
中间体7b的合成:
将化合物(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.29g,1.20mmol)溶于二氯甲烷(10mL)内,冰浴加入吡啶(395mg,5.00mmol)和二氯亚砜(202mg,1.70mmol),体系处于室温反应2小时后浓缩,除去溶剂及多余的二氯亚砜。然后加入二氯甲烷(10mL)及化合物7a(250mg,1.00mmol)。体系处于室温反应过夜。TLC监控显示无原料剩余。将体系浓缩,粗品用柱层析进行分离纯化(石油醚:乙酸乙酯=10:1-1:1),得到7b(100mg,产率21%)。
中间体7c的合成:
将7b(47mg,0.10mmol)溶于二氯甲烷(5mL)内,加入三氟乙酸(2mL)。体系处于室温反应1小时。TLC监控显示原料无剩余。将体系浓缩,得到7c(50mg,粗品)。所得到产物未进一步纯化,直接用于下一步反应。
终产物7的合成:
将7c(37mg,0.10mmol)溶于二氯甲烷(2mL),加入三乙胺(20mg,0.20mmol)以及醋酸酐(20mg,0.20mmol)。体系处于室温反应1小时。TLC监控显示无原料剩余。将体系浓缩,所得到粗品用薄板层析分离纯化(二氯甲烷:甲醇=10:1),得到终产物7(30mg,产率72%)。MS:(m/z,ESI):417.2[M+H] +1H NMR(600MHz,DMSO-d6)δ10.57(s,1H),8.33(s,1H),8.11(d,J=5.4Hz,1H),7.79(d,J=7.8Hz,1H),7.35(d,J=7.8Hz,1H),7.09(d,J=11.4Hz,1H),6.90(d,J=7.8Hz,1H),4.10-4.07(m,2H),3.57-3.55(m,1H),2.59-2.57(m,1H),1.87-1.84(m,1H),1.77-1.75(m,5H),1.70-1.50(m,1H),1.31-1.26(m,3H),1.24-1.21(m,3H),1.07-1.05(m,1H).
实施例8:
Figure PCTCN2022094496-appb-000021
中间体8a的合成:
将5-氟-4-碘吡啶-2-胺(1.00g,4.20mmol)溶解于乙二醇二甲醚(20mL)和水(4mL)中,接着加入4-氟-2-异丙氧基苯硼酸(0.83g,4.20mmol),[1,1'-双(二苯基膦基)二茂铁]二氯化钯(0.31g,0.42mmol)和碳酸钾(1.74g,12.60mmol),用氮气置换三次,使整个体系处于氮气的氛围下.体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。减压浓缩,经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到8a(0.85g,产率77%)。
中间体8b的合成:
将8a(0.85g,3.20mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.78g,3.20mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(1.44g,3.80mmol)和N,N-二异丙基乙胺(0.83g,6.40mmol),整个体系在室温下搅拌过夜,TLC检测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到8b(0.90g,产率57%)。
中间体8c的合成:
将8b(0.90g,1.84mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入三氟乙酸(2mL),整个体系在室温下搅拌过夜,TLC检测直至原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,用二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1),得到8c(0.64g,产率89%)。
终产物8的合成:
将8c(0.64g,1.64mmol)溶解于二氯甲烷(35mL)中,接着加入乙酸酐(0.84g,8.20mmol)和三乙胺(0.83g,8.20mmol)。体系在室温下搅拌,TLC跟踪监测直至原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物8(0.42g,产率59%)。MS:(m/z,ESI):431.2[M+ H] +
1H NMR(600MHz,DMSO-d 6)δ10.57(s,1H),8.32(s,1H),8.12(s,1H),7.78(d,J=7.8Hz,1H),7.34(d,J=7.2Hz,1H),7.10(s,1H),6.88(d,J=8.4Hz,1H),4.72-4.69(m,1H),3.56(s,1H),2.62-2.58(m,1H),1.78(s,6H),1.31-1.23(m,4H),1.20(s,6H),1.09-1.04(m,1H).
实施例9:
Figure PCTCN2022094496-appb-000022
中间体9a的合成:
将1-溴-2-二氟甲氧基-4-氟苯(1.00g,4.15mmol),双联频哪醇硼酸酯(1.26g,4.98mmol),乙酸钾(1.22g,12.45mmol),[1,1'-双(二苯基膦基)二茂铁]二氯化钯(0.24g,0.33mmol),乙二醇二甲醚(30mL)加入到反应瓶中,在氮气保护下加热至100℃反应8小时,TLC监测无原料剩余。停止加热,降低至室温。减压浓缩,柱层析分离(正己烷:乙酸乙酯=10:1),得到9a(0.50g,产率42%)。
中间体9b的合成:
将9a(0.50g,1.74mmol),5-氟-4-碘-吡啶-2-胺(0.33g,1.39mmol),四三苯基膦钯(0.12g,0.10mmol),三水合磷酸三钾(0.60g,2.26mmol),二氧六环(30mL)加入到反应瓶中,在氮气保护下加热至100℃反应8小时,TLC监测原料无剩余。停止加热,降至室温。减压浓缩,柱层析分离(正己烷:乙酸乙酯=1:1),得到9b(0.39g,产率83%)。
中间体9c的合成:
将9b(0.39g,1.43mmol,)(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.35g,1.43mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(0.65g,1.72mmol),N,N-二异丙基乙胺(0.37g,2.86mmol)和N,N-二甲基甲酰胺(20mL)加入到反应瓶中,室温反应15小时,TLC监测原料无剩余。在反应液中加入水(30mL),乙酸乙酯萃取(30mL×3),合并有机相,饱和氯化钠洗涤(30mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(正己烷:乙酸乙酯=1:1),得到9c(0.16g,产率23%)。
中间体9d的合成:
将9c(0.16g,0.33mmol)溶于二氯甲烷(20mL)中,加入三氟乙酸(4mL),室温反应4小时,TLC监测原料无剩余。将反应液用水洗涤(20mL×3),合并水相,水相用碳酸钠调节pH为8-9,二氯甲烷萃取(30mL×3),合并有机相,无水硫酸钠干燥,减压除去溶剂,得到9d(0.10g,产率76%)。
终产物9的合成:
将9d(0.10g,0.26mmol)溶解于二氯甲烷(20mL)中,加入乙酸酐(0.05g,0.52mmol),三乙胺(0.05g,0.52mmol)室温反应2小时,TLC监测原料无剩余。反应液直接进行柱层析分离(二氯甲烷:甲醇=25:1),得到终产物9(0.05g,产率44%)。MS m/z(ESI):440.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.67(s,1H),8.41(s,1H),8.12(d,J=5.4Hz,1H),7.78(d,J=7.2Hz,1H),7.58-7.56(m,1H),7.42(s,1H),7.34-7.28(m,1H),3.57-3.56(m,1H),2.61-2.59(m,1H),1.89-1.78(m,4H),1.77(s,3H),1.31-1.24(m,3H),1.08-1.06(m,1H).
实施例10:
Figure PCTCN2022094496-appb-000023
中间体10a的合成:
将5-氟-4-碘吡啶-2-胺(500mg,2.10mmol)溶解于DME(20mL)和水(4mL)的混合溶剂中,接着加入2-苄氧基-4-氟苯硼酸(620mg,2.52mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(90mg,0.11mmol)和碳酸钾(870mg,6.30mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下回流搅拌,反应4小时,TLC监测原料完全反应。反应直接经柱层析分离纯化(石油醚:乙酸乙酯=5:1-2:1),得到10a(0.50g,产率76%)。
中间体10b的合成:
将10a(0.30g,0.96mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.25g,1.01mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(0.43g,1.15mmol)和N,N-二异丙基乙胺(0.25g,1.92mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,粗品经柱层析纯化(石油醚:乙酸乙酯=10:1-2:1),得到10b(0.30g,产率58%)。
中间体10c的合成:
将10b(0.30g,0.56mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入三氟乙酸(2mL),体系在室温下搅拌2小时,TLC监测原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,用二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1),得到10c(0.20g,产率82%)。
终产物10的合成:
将10c(0.20g,0.46mmol)用二氯甲烷(10mL)溶解,然后加入乙酸酐(94mg,0.92mmol),和三乙胺(93mg,0.92mmol),室温反应1.5小时,TLC(乙酸乙酯)监测原料完全反应。反应液中加入饱和碳酸氢钠(30mL),二氯甲烷萃取(20mL×3),合并有机相,饱和氯化钠洗涤(20mL×2),无水硫酸钠干燥,减压除去溶剂,粗品经柱层析纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物10(150mg,产率68%)。MS m/z(ESI):480.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.32(s,1H),8.14(d,J=5.4Hz,1H),7.76(d,J=7.8Hz,1H),7.37(dd,J=7.8Hz,J=7.2Hz,1H),7.32-7.27(m,5H),7.16(d,J=11.4Hz,1H),6.92(dd,J=8.4Hz,J=8.4Hz,1H),5.15(s,2H),3.55-3.54(m,1H),2.58-2.56(m,1H),1.86-1.84(m,1H),1.75-1.70(m,6H),1.33-1.15(m,4H).
实施例11:
Figure PCTCN2022094496-appb-000024
中间体11a的合成:
将5(0.10g,0.25mmol)溶于二氯甲烷(20mL)中,加入三溴化硼(0.12g,0.50mmol),室温反应4小时,TLC监测原料无剩余。反应液用碳酸氢钠水溶液调节pH为6左右,分离有机相,无水硫酸钠干燥。减压蒸除溶剂,得到11a(0.08g,产率82%)。
终产物11的合成:
将11a(0.08g,0.21mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入2-溴乙基甲基醚(0.03g,0.25mmol),碳酸钾(0.06g,0.42mmol),室温反应8小时,TLC监测原料无剩余。在反应液中加入水(20mL),乙酸乙酯萃取(20mL×3),合并有机相,无水硫酸钠干燥。有机相柱层析分离(二氯甲烷:甲醇=25:1),得到终产物11(0.05g,产率53%)。MS m/z(ESI):448.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.31(s,1H),8.11-8.10(m,1H),7.78-7.58(m,1H),7.36-7.33(m,1H),7.12-7.01(m,1H),6.92-6.89(m,1H),4.15-4.13(m,2H),3.55-3.54(m,2H),3.53-3.52(m,1H),3.15(s,3H),2.59-2.58(m,1H),1.85(s,3H),1.83-1.47(m,4H),1.29-1.04(m,4H).
实施例12:
Figure PCTCN2022094496-appb-000025
中间体12a的合成:
将2-氨基-5-氟-4-碘吡啶(0.50g,2.10mmol)和5-氯-2-甲氧基苯硼酸(0.47g,2.50mmol)溶解于乙二醇二甲醚(10mL)中,加入[1,1-双(二苯基膦基)二茂铁]二氯化钯(73mg,0.10mmol),碳酸钾(0.87g,6.3mmol)和水(2mL)。用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下反应2小时。TLC监控显示无原料剩余。降温后,浓缩除去溶剂。粗品用柱层析进行分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到12a(0.50g,产率95%)。
中间体12b的合成:
将(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.29g,1.20mmol)溶于二氯甲烷(10mL)内,冰浴加入吡啶(0.40g,5.00mmol)和二氯亚砜(0.20g,1.70mmol),体系处于室温反应2小时后浓缩,除去溶剂及多余的二氯亚砜。然后加入二氯甲烷(10mL)及化合物12a(0.25g,1.00mmol)。体系处于室温反应过夜。TLC监控显示无原料剩余。将体系浓缩,粗品用柱层析进行分离纯化(石油醚:乙酸乙酯=10:1-1:1),得到12b(0.10g,产率21%)。
中间体12c的合成:
将12b(48mg,0.10mmol)溶于二氯甲烷(5mL)内,加入三氟乙酸(2mL)。体系处于室温反应1小时。TLC监控显示原料无剩余。将体系浓缩,得到12c(50mg,粗品)。所得到产物未进一步纯化,直接用于下一步反应。
终产物12的合成:
将12c(50mg,0.10mmol)溶于二氯甲烷(2mL),加入三乙胺(20mg,0.20mmol)以及醋酸酐(20mg,0.20mmol)。体系处于室温反应1小时。TLC监控显示无原料剩余。将体系浓缩,所得到粗品用薄板层析分离纯化(二氯甲烷:甲醇=10:1),得到终产物12(30mg,产率72%)。MS:(m/z,ESI):420.1[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.60(s,1H),8.34(s,1H),8.08(d,J=5.4Hz,1H),7.78(d,J=7.8Hz,1H),7.33(d,J=8.4Hz,1H),7.27(s,1H),7.20-7.15(m,1H),3.80(s,3H),3.65-3.55(m,1H),2.68-2.62(m,1H),1.90-1.85(m,1H),1.76-1.55(m,6H),1.28-1.23(m,3H),1.15-1.10(m,1H).
实施例13:
Figure PCTCN2022094496-appb-000026
中间体13a的合成:
将3-甲氧基-4-溴苯酚(3.00g,14.80mmol)溶解于丙酮(50mL)中,接着加入溴甲基环丙烷(2.20g,16.30mmol),碘化钠(1.11g,7.40mmol),碳酸铯(9.64g,29.60mmol),在回流条件下搅拌,反应8小时,TLC监测原料无剩余。停止加热,降至室温。减压蒸除丙酮,残留物中加入水(50mL),乙酸乙酯萃取(30mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压浓缩除去溶剂,得到13a(3.70g,产率97%)。
中间体13b的合成:
将13a(3.20g,12.40mmol)溶解于乙二醇二甲醚(50mL)中,接着加入双联频哪醇硼酸酯(3.79g,14.90mmol),乙酸钾(3.65g,37.2mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(0.88g,1.2mmol),在氮气保护下加热至100℃反应8小时,TLC监测无原料剩余。停止加热,降至室温。减压除去溶剂,柱层析分离纯化(正己烷:乙酸乙酯=5:1),得到13b(2.50g,产率66%)。
中间体13c的合成:
将13b(2.19g,7.20mmol)溶解于二氧六环(50mL)中,接着加入5-氟-4-碘-吡啶-2-胺(1.38g,5.80mmol),四三苯基膦钯(0.46g,0.40mmol)和三水合磷酸钾(2.50g,9.40mmol),在氮气保护下加热至100℃反应8小时,TLC监测原料无剩余。停止加热,降至室温。减压浓缩除去溶剂,柱层析分离纯化(正己烷:乙酸乙酯=1:1),得到13c(1.90g,产率92%)。
中间体13d的合成:
将13c(0.60g,2.10mmol)溶解于N,N-二甲基甲酰胺(20mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.51g,2.10mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(0.95g,2.50mmol)和N,N-二异丙基乙胺(0.54g,4.20mmol),室温反应15小时,TLC监测原料无剩余。在反应液中加入水(50mL),乙酸乙酯萃取(30mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(正己烷:乙酸乙酯=1:1),得到13d(0.50g,产率46%)。
中间体13e的合成:
将13d(0.50g,0.97mmol)溶解于二氯甲烷(20mL)中,加入三氟乙酸(4mL),室温反应4小时,TLC监测原料无剩余。将反应液用水洗涤(30mL×3),合并水相,水相用碳酸钠调节pH为8-9,二氯甲烷萃取(30mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥。减压浓缩除去溶剂,得到13e(0.35g,产率88%)。
终产物13的合成:
将13e(0.06g,0.14mmol)溶解于二氯甲烷(20mL)中,接着加入乙酸酐(0.03g,0.28mmol),三乙胺(0.03g,0.28mmol),室温反应2h,TLC监测原料无剩余。反应液直接进行柱层析分离(DCM:MeOH=25:1),得到终产物13(0.02g,产率31%)。MS m/z(ESI):456.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.52(s,1H),8.28(s,1H),8.06(d,J=4.8Hz,1H),7.78(d,J=7.8Hz,1H),7.18(d,J=8.4Hz,1H),6.71(s,1H),6.63(d,J=8.4Hz,1H),3.89(d,J=7.2Hz,2H),3.76(s,3H),3.57-3.55(m,1H),2.61-2.60(m,1H),1.88-1.86(m,1H),1.77(s,3H),1.76-1.75(m,3H),1.31-1.23(m,4H),1.09-1.04(m,1H),0.60-0.59(m,2H),0.35-0.34(m,2H).
实施例14:
Figure PCTCN2022094496-appb-000027
中间体14a的合成:
将4-溴-3-甲氧基苯酚(0.40g,2.00mmol)溶解于二氧六环(50mL)中,接着加入双联频哪醇二硼烷(0.60g,2.40mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯(0.12g,0.16mmol)和乙酸钾(0.59g,6.00mmol),将体系用氮气保护,在100℃下搅拌,反应5小时,TLC监测原料反应完毕。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚-石油醚:乙酸乙酯=4:1),得到14a(0.36g,产率72%)。
中间体14b的合成:
将14a(0.36g,1.44mmol)溶解于二乙二醇二甲醚(50mL)和水(10mL)中,接着加入化合物5-氟-4-碘-吡啶-2-胺(0.29g,1.20mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯(0.44g,0.60mmol)和碳酸钾(0.50g,3.60mmol)。整个体系在80℃下搅拌5小时,TLC检测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(100mL×3),合并有机相,饱和氯化钠洗涤(100mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=4:1-2:3),得到14b(0.25g,产率74%)
中间体14c的合成:
将14b(0.15g,0.64mmol)溶解于N,N-二甲基甲酰胺(50mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.18g,0.72mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(0.58g,0.72mmol)和二异丙基乙胺(0.20mL,1.20mmol)整个体系在25℃下搅拌反应15小时,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(100mL×3),合并有机相,饱和氯化钠洗涤(100mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=4:1-2:1),得到14c(0.15g,产率51%)。
终产物14的合成:
将14c(0.15g,0.32mmol)溶解于二氯甲烷(50mL)之中,接着加入三氟乙酸(2mL)室温下搅拌4小时,TLC检测原料无剩余。向体系内加入饱和碳酸钠水溶液,将反应体系的pH值调节至9-10,二氯甲烷萃取(50mL×3),合并有机相浓缩至体积约为50mL,加入三乙胺(2mL)和乙酸酐(2mL)室温下反应30分钟后,加入碳酸钠水溶液洗涤有机相。分液,再用二氯甲烷萃取(50mL×3),减压浓缩,经柱层析分离(乙酸乙酯),得到终产物14(90mg,产率63%)。MS m/z(ESI):444.2[M+H] +
1H NMR(600MHz,CD 3OD)δ8.16(s,1H),8.10(s,1H),7.29(d,J=7.8Hz,1H),6.87(s,1H),6.78(t,J=7.8Hz,1H),3.79(s,3H),3.78-3.73(m,1H),2.72(t,J=12.0Hz,1H),2.31-2.29(m,1H),2.20-2.10(m,1H),2.00-1.80(m,6H),1.51-1.38(m,3H),1.27-1.21(m,2H).
实施例15:
Figure PCTCN2022094496-appb-000028
中间体15a的合成:
将3-甲氧基-4-溴苯酚(540mg,2.66mmol)溶解于N,N-二甲基甲酰胺(50mL)中,接着加入碳酸钾(735mg,5.32mmol)和溴化苄(910mg,5.32mmol)反应在室温下搅拌15小时,TLC检测原料无剩余,向体系中加入水(50mL),乙酸乙酯萃取(50mL×3)合并有机相,饱和氯化钠水溶液洗涤(50mL×2),无水硫酸钠干燥,减压浓缩,经柱层析分离纯化(石油醚-石油醚:乙酸乙酯=4:1),得到15a(662mg,收率85%)。
中间体15b的合成:
将化合物15a(662mg,2.27mmol),双联频哪醇二硼烷(1.15g,4.54mmol),乙酸钾(667mg,6.81mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(146mg,0.20mmol)溶解到无水1,4-二氧六环(100mL)中。将反应体系用氮气保护并在100℃反应4小时,TLC检测原料无剩余。向反应体系中加入水(100mL),乙酸乙酯萃取(100mL×3),合并有机相,无水硫酸钠干燥,减压浓缩除去溶剂,经柱层析分离纯化(石油醚-石油醚:乙酸乙酯=4:1),得到15b(510mg,产率65%)。
中间体15c的合成:
将15b(510mg,1.50mmol)溶解于二乙二醇二甲醚(150mL)中,室温下加入5-氟-4碘吡啶-2-胺(536mg,2.25mmol),[1,1'-双(二苯基膦基)二茂铁]二氯化钯(110mg,0.15mmol)和碳酸钾(621mg,4.50mmol),将反应体系在80℃下搅拌4小时,TLC检测原料无剩余。向反应体系中加入水(100mL),乙酸乙酯萃取(100mL×3),合并有机相,经无水硫酸钠干燥,减压除去溶剂,经柱层析纯化(石油醚-石油醚:乙酸乙酯=1:1),得到15c(263mg,收率54%)。
中间体15d的合成:
将15c(263mg,0.81mmol)和(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(290mg,1.23mmol)溶解于N,N-二甲基甲酰胺(20mL)中,依次加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(470mg,1.23mmol)和二异丙基乙胺(480mg,3.69mmol),将反应体系在室温反应4小时,TLC检测原料无剩余。向反应体系中加入水(100mL),乙酸乙酯萃取(100mL×3),合并有机相,经无水硫酸钠干燥,减压除去溶剂,经过柱层析纯化(石油醚:乙酸乙酯=4:1-2:1),得到15d(250mg,收率56%)。
中间体15e的合成:
将15d(250mg,0.45mmol)溶解到二氯甲烷(50mL)中,接着加入三氟乙酸(5mL)室温下搅拌4小时,TLC检测原料无剩余。向体系中加入饱和碳酸钠水溶液,将反应体系的pH值调节至9-10。分液后再用二氯甲烷萃取(100mL×3),合并有机相,浓缩至体积约为50mL,加入三乙胺(2mL)和乙酸酐(2mL)室温下反应30分钟后,加入碳酸钠水溶液洗涤有机相,二氯甲烷萃取(20mL×3),合并有机相,经无水硫酸钠干燥,减压除去溶剂,经柱层析分离(乙酸乙酯),得到15e(176mg,产率79%)
终产物15的合成:
将15e(175mg,0.36mmol)溶解于甲醇(20mL)中,加入钯碳(10mg),将反应体系用氢气保护,室温反应10小时,LC-MS检测原料无剩余,将钯碳滤掉之后,减压浓缩反应溶液,得到终产物15(117mg,收率81%)。MS m/z(ESI):402.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.49(s,1H),10.09(s,1H),8.25(s,1H),8.05(s,J=5.4Hz,1H),7.78(d,J=7.8Hz,1H),7.07(d,J=8.4Hz,1H),6.55(d,J=2.4Hz,1H),6.48(s,1H),3.71(s,3H),3.55-3.53(m,1H),2.61-2.57(m,1H),1.87(d,J=11.4Hz,1H),1.77(s,3H),1.77-1.75(m,3H),1.29-1.27(m,4H).
实施例16:
Figure PCTCN2022094496-appb-000029
中间体16a的合成:
将2-(3,4-二甲氧基苯基)-4,4,5,5-四甲基-1,3,2-二氧杂硼烷(150mg,0.57mmol)和5-氟-4-碘吡啶-2-胺(202mg,0.85mmol)溶解到二乙二醇二甲醚(50mL)中,依次加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(37mg,0.05mmol),碳酸钾(117mg,0.85mmol)和水(10mL),将反应体系用氮气保护在80℃反应4小时,TLC检测原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(100mL×3),合并有机相,无水硫酸钠干燥,减压除去溶剂,经过柱层析纯化(石油醚:乙酸乙酯=4:1-1:1),得到16a(124mg,收率88%)。
中间体16b的合成:
将化合物16a(36mg,0.15mmol)溶解于N,N-二甲基甲酰胺(10mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(71mg,0.29mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(110mg,0.29mmol)和二异丙基乙胺(57mg,0.44mmol),整个体系在25℃下搅拌反应15小时,TLC监测原料无剩余。向反应液中加入水(15mL),乙酸乙酯萃取(20mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经过柱层析分离纯化(石油醚:乙酸乙酯=4:1-2:1),得到16b(35mg,产率49%)。
终产物16的合成:
将16b(35mg,0.074mmol)溶解到二氯甲烷(20mL)中,接着加入三氟乙酸(2mL)室温下搅拌4小时,TLC检测原料无剩余。向体系中加入饱和碳酸钠水溶液,将反应体系的pH值调节至9-10,二氯甲烷萃取(50mL×3),合并有机相,浓缩至体积约为20mL,加入三乙胺(2mL)和乙酸酐(2mL)。室温下反应30分钟,加入碳酸钠水溶液洗涤有机相。二氯甲烷萃取水相(20mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经硅胶柱层析分离(乙酸乙酯),得到终产物16(15mg,产率49%)。MS m/z(ESI):416.2[M+H] +
1H NMR(600MHz,CD 3OD)δ8.12(s,1H),8.08(d,J=5.4Hz,1H),7.19(d,J=8.4Hz,1H),6.64(s,1H),6.60(s,1H),3.88(s,3H),3.79(s,3H),3.71(t,J=4.8Hz,2H),2.71-2.57(m,1H),2.24-2.22(m,1H),1.91(s,3H),1.89(s,2H),1.49-1.19(m,6H).
实施例17:
Figure PCTCN2022094496-appb-000030
中间体17a的合成:
将2-甲氧基苯硼酸(0.42g,2.77mmol)溶解于二乙二醇二甲醚(30mL)和水(6mL)中,接着加入化合物5-氟-4-碘吡啶-2-胺(0.60g,2.52mmol),[1,1'-双(二苯基膦基)二茂铁]二氯化钯(95mg,0.13mmol)和碳酸钾(1.04g,7.56mmol)。将体系用氮气保护,在100℃下搅拌,反应4小时,TLC监测原料反应无剩余。向反应体系中加入的水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=4:1-2:3),得到17a(0.60g,产率99%)。
中间体17b的合成:
将17a(0.60g,2.77mmol)溶解于N,N-二甲基甲酰胺(50mL)中,接着加入化合物(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.79g,3.20mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(1.22g,3.20mmol)和二异丙基乙胺(1.2mL,7.00mmol),整个体系在室温下搅拌15小时,TLC检测原料无剩余。向反应体系中加入的水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=4:1-2:3),得到17b(0.60g,产率49%)。
中间体17c的合成:
将17b(0.60g,1.35mmol)溶解于二氯甲烷(50mL)中,接着加入三氟乙酸(3mL)整个体系在25℃下搅拌反应,TLC监测跟踪直至原料无剩余。向反应液中加入碳酸钠水溶液(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,粗品17c可直接进行下一步反应。
终产物17的合成
将17c溶解到二氯甲烷(50mL)中,接着加入三乙胺(0.4mL,2.70mmol)和乙酸酐(0.3mL,2.70mmol)室温下搅拌30分钟,TLC检测原料反应完毕。向体系内加入饱和碳酸钠水溶液,二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=4:1-2:1),得到终产物17(0.35g,产率68%)。MS m/z(ESI):386.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.55(s,1H),8.30(s,1H),8.07(d,J=5.4Hz,1H),7.76(d,J=7.8Hz,1H),7.46(t,J=7.8Hz,1H),7.27(d,J=7.2Hz,1H),7.15(d,J=8.4Hz,1H),7.06(t,J=7.8Hz,1H),3.75(s,3H),3.55-3.54(m,1H),2.56-2.48(m,1H),1.86-1.84(m,1H),1.77-1.74(m,6H),1.31-1.02(m,4H).
实施例18:
Figure PCTCN2022094496-appb-000031
中间体18a的合成:
将4-(溴甲基)吡啶盐酸盐(1.72g,6.81mmol)溶解于20mL N,N-二甲基甲酰胺中,接着加入2-碘苯酚(1.50g,6.81mmol),碳酸钾(2.83g,20.45mmol),和碘化钠(1.02g,6.81mmol),体系在室温下搅拌,反应4小时,TLC监测原料无剩余。反应直接经柱层析分离纯化(石油谜:乙酸乙酯=2:1-石油谜:乙酸乙酯=1:1),得到18a(1.90g,产率90%)。
中间体18b的合成:
将18a(1.00g,3.21mmol)溶解于15mL乙二醇二甲醚中,接着加入双联频哪醇硼酸酯(980mg,3.85mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯(234mg,0.32mmol)和乙酸钾(0.95g,9.64mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。反应直接经柱层析分离纯化(石油谜:乙酸乙酯=2:1-石油谜:乙酸乙酯=1:1),得到化合物18b(0.80g,产率80%)。
中间体18c的合成:
将5-氟-4-碘吡啶-2-胺(802mg,3.37mmol)溶解于20mL乙二醇二甲醚中,接着加入18b(700mg,2.25mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯(168mg,0.23mmol)和碳酸钾(932mg,6.75mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。反应直接经柱层析分离纯化(二氯甲烷:甲醇=50:1-二氯甲烷:甲醇=10:1),得到18c(220mg,产率33%)。
中间体18d的合成:
将18c(220mg,0.75mmol)溶解于10mL N,N-二甲基甲酰胺中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(217mg,0.89mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(340mg,0.89mmol)和N,N-二甲基乙基胺(192mg,1.49mmol),整个体系在室温下搅拌过夜,TLC检测原料无剩余。向反应液中加入100mL水,乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-二氯甲烷:甲醇=20:1),得到18d(300mg,产率77%)。
中间体18e的合成:
将18d(0.30g,0.58mmol)溶解于30mL二氯甲烷中,接着冰水浴下加入2mL的三氟乙酸,整个体系在室温下搅拌过夜,TLC检测直至原料无剩余。向反应液中加入100mL水,再用饱和碳酸氢钠水溶液调节PH=9-10,用二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-二氯甲烷:甲醇=8:1)后,得到18e(0.16g,产率66%)。
终产物18的合成:
将18e(160mg,0.38mmol)溶解于二氯甲烷(5mL)中,接着加入乙酸酐(116mg,1.14mmol)和三乙胺(115mg,1.14mmol)。体系在室温下搅拌,TLC跟踪监测直至原料无剩余。向反应液中加入50mL水,二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(石油醚:乙酸乙酯=10:1-石油醚:乙酸乙酯=2:1)得到终产物18(60mg,产率34%)。MS m/z(ESI):463.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.61(s,1H),8.50(d,J=5.4Hz,2H),8.36(s,1H),8.20(d,J=5.4Hz, 1H),7.80(d,J=7.8Hz,1H),7.47-7.44(m,1H),7.35(d,J=7.2Hz,1H),7.30(d,J=5.4Hz,2H),7.18(d,J=9.0Hz,1H),7.15(t,J=7.8Hz,1H),5.22(s,2H),3.58-3.57(m,1H),2.62-2.53(m,1H),1.89-1.75(m,7H),1.30-1.04(m,4H).
实施例19:
Figure PCTCN2022094496-appb-000032
中间体19a的合成
将7-溴苯并呋喃(600mg,3.00mmol)溶于乙二醇二甲醚(50mL)中,接着加入联硼酸频那醇酯(928mg,3.70mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(163mg,0.20mmol)和乙酸钾(892mg,9.10mmol),体系在氮气保护下升温至100℃,反应4小时,TLC监测原料完全反应。反应液降至室温,粗品经柱层析分离纯化(石油醚:乙酸乙酯=100:1-50:1),得到19a(450mg,产率62%)。
中间体19b的合成
将19a(450mg,1.84mmol)溶解于乙二醇二甲醚(50mL)和水(10mL)中,接着加入5-氟-4-碘吡啶-2-胺(350mg,1.47mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(110mg,0.15mmol)和碳酸钾(405mg,2.94mmol),体系在氮气保护下升温至100℃,反应4小时,TLC监测原料完全反应。反应液降至室温,粗品经柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到19b(300mg,产率71%)。
中间体19c的合成
(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(292mg,1.20mmol)溶于二氯甲烷(50mL)中,接着加入吡啶(474mg,6.00mmol)和二氯亚砜(242mg,2.04mmol),室温反应4小时,接着将19b(300mg,1.32mmol)加入到上述反应液中,室温继续反应过夜。TLC监测原料无剩余。用水(30mL)稀释反应液,萃取收集有机相,减压浓缩,粗品经柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到19c(200mg,产率39%)。
中间体19d的合成
将19c(200mg,0.47mmol)溶于二氯甲烷(10mL)中,接着加入三氟乙酸(1mL),室温反应1.5小时,TLC监测原料完全反应。反应液中加入饱和碳酸氢钠(30mL),二氯甲烷萃取(20mL×3),合并有机相,饱和氯化钠洗涤(20mL×2),无水硫酸钠干燥,减压除去溶剂,得到19d(128mg,产率83%)。
终产物19的合成
将19d(128mg,0.39mmol)溶解于二氯甲烷(10mL)中,接着加入乙酸酐(48mg,0.47mmol)和三乙胺(47mg,0.47mmol),室温反应1.5小时,TLC监测原料完全反应。反应液中加入饱和碳酸氢钠(30mL),二氯甲烷萃取(20mL×3),合并有机相,饱和氯化钠洗涤(20mL×2),无水硫酸钠干燥,减压除去溶剂,粗品经柱层析纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物19(117mg,产率76%)。MS m/z(ESI):396.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.68(s,1H),8.48(s,1H),8.41(d,J=5.4Hz,1H),8.09(s,1H),7.83-7.78(m,2H),7.46(d,J=7.2Hz,1H),7.41(dd,J=7.2Hz,J=7.2Hz,1H),7.09(s,1H),3.58-3.56(m,1H),2.64-2.60(m,1H),1.90-1.84(m,1H),1.78-1.76(m,6H),1.31-1.25(m,3H),1.10-1.06(m,1H).
实施例20:
Figure PCTCN2022094496-appb-000033
中间体20a的合成:
将2,6-二氟吡啶-3-硼酸(550mg,3.14mmol)和化合物5-氟-4-碘吡啶-2-胺(898mg,3.77mmol)溶解于1,4-二氧六环(15mL)中,依次加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(219mg,0.30mmol)和碳酸钾(1.30g,9.42mmol),再加入水(6mL)。将体系用氮气保护,在100℃下搅拌,反应5小时,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,粗品经柱层析纯化(石油醚:乙酸乙酯=4:1-2:3),得到20a(700mg,产率99%)。
中间体20b的合成:
将20a(700mg,3.14mmol)溶解于乙腈(50mL)中,接着加入化合物(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(778mg,3.20mmol),四甲基氯代脲六氟磷酸酯(898mg,3.20mmol)和N-甲基咪唑(0.87mL,11.00mmol)整个体系在室温下搅拌15小时,TLC检测原料无剩余。浓缩溶剂后,柱层析分离纯化(石油醚:乙酸乙酯=4:1-2:3),得到20b(1.40g,产率99%)
终产物20的合成:
将20b(1.40g,3.14mmol)溶解于二氯甲烷(50mL)中,在室温下加入5mL的三氟乙酸,反应4小时,TLC检测原料无剩余。饱和碳酸钠水溶液将反应体系洗涤至弱碱性,二氯甲烷萃取(30mL×3),合并有机相,无水硫酸钠干燥。将溶剂浓缩至50mL,再加入2mL的三乙胺和2mL的乙酸酐,室温搅拌30分钟,TLC监测原料无剩余。用无水碳酸钠溶液洗涤,再用二氯甲烷萃取(30mL×3),合并有机相,减压浓缩除去溶剂,粗品经柱层析分离,得到终产物20(750mg,收率61%)。MS m/z(ESI):393.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.74(s,1H),8.50(s,1H),8.40(dd,J=16.8Hz,J=7.8Hz,1H),8.26(d,J=6.0Hz,1H),7.79(d,J=7.8Hz,1H),7.41(dd,J=7.8Hz,J=2.4Hz,1H),3.60-3.55(m,1H),2.64-2.60(m,1H),1.91-1.88(m,1H),1.78-1.76(m,6H),1.29-1.27(m,3H),1.09-1.06(m,1H).
实施例21:
Figure PCTCN2022094496-appb-000034
中间体21a的合成:
将5-氟-4-碘吡啶-2-胺(1.00g,4.20mmol)溶解于乙二醇二甲醚(20mL)和水(4mL)中,接着加入吲唑-4-硼酸(0.68g,4.20mmol),[1,1'-双(二苯基膦基)二茂铁]二氯化钯(0.34g,0.42mmol)和碳酸钾(1.74g,12.60mmol),用氮气置换三次,使整个体系处于氮气的氛围下.体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。减压浓缩,经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到21a(0.77g,产率80%)。
中间体21b的合成:
将21a(0.75g,3.30mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.80g,3.30mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(1.56g,4.10mmol)和N,N-二异丙基乙胺(0.85g,6.60mmol),整个体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到21b(0.91g,产率60%)。
中间体21c的合成:
将21b(0.91g,2.00mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入2mL的三氟乙酸,整个体系在室温下搅拌过夜,TLC检测原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,用二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1)后,得到21c(0.61g,产率86%)。
终产物21的合成:
将21c(0.61g,1.72mmol)溶解于二氯甲烷(35mL)中,接着加入乙酸酐(0.53g,5.20mmol)和三乙胺(0.52g,5.20mmol),体系在室温下搅拌,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物21(0.32g,产率47%)。MS m/z(ESI):396.1[M+H] +
1H NMR(600MHz,CDCl 3)δ8.60(s,1H),8.27(s,1H),8.14(s,1H),7.63(d,J=7.8Hz,1H),7.51(d,J=7.8Hz,1H),7.39(d,J=6.6Hz,1H),7.15(d,J=7.2Hz,1H),3.89-3.87(m,1H),2.54-2.50(m,1H),2.29(d,J=12.0Hz,1H),1.79-1.77(m,6H),1.44-1.28(m,3H),1.12-1.09(m,1H).
实施例22:
Figure PCTCN2022094496-appb-000035
中间体22a的合成:
将5-氟-4-碘吡啶-2-胺(1.00g,4.20mmol)溶解于乙二醇二甲醚(20mL)和水(4mL)中,接着加入吲哚-4-硼酸(0.68g,4.20mmol),[1,1'-双(二苯基膦基)二茂铁]二氯化钯(0.34g,0.42mmol)和碳酸钾(1.74g,12.60mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。减压浓缩,经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到22a(0.78g,产率82%)。
中间体22b的合成:
将22a(0.78g,3.40mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.83g,3.40mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(1.56g,4.10mmol)和N,N-二异丙基乙胺(0.88g,6.80mmol),整个体系在室温下搅拌过夜,TLC监测原料无剩余.向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到22b(0.92g,产率59%)。
中间体22c的合成:
将22b(0.92g,2.03mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入2mL的三氟乙酸,整个体系在室温下搅拌过夜,TLC检测原料无剩余.向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1)后,得到22c(0.61g,产率85%)。
终产物22的合成:
将22c(0.61g,1.73mmol)溶解于二氯甲烷(35mL)中,接着加入乙酸酐(0.53g,5.20mmol)和三乙胺(0.52g,5.20mmol),体系在室温下搅拌,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物22(0.35g,产率51%)。MS m/z(ESI):395.1[M+H] +
1H NMR(600MHz,DMSO-d 6)δ11.38(s,1H),10.59(s,1H),8.42(s,1H),8.36(d,J=6.0Hz,1H),7.77(d,J=7.8Hz,1H),7.54(d,J=8.4Hz,1H),7.46(d,J=3.0Hz,1H),7.23(d,J=8.4Hz,1H),7.15(d,J=7.2Hz,1H),6.41(s,1H),3.60-3.54(m,1H),2.64-2.60(m,1H),1.89(d,J=12.0Hz,1H),1.77-1.76(m,6H),1.34-1.23(m,3H),1.10-1.08(m,1H).
实施例23:
Figure PCTCN2022094496-appb-000036
中间体23a的合成:
将5-氟-4-碘吡啶-2-胺(1.00g,4.20mmol)溶解于N,N-二甲基甲酰胺(20mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(1.32g,5.40mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(2.39g,6.29mmol)和N,N-二甲基乙基胺(2.08mL,12.6mmol),整个体系在室温下搅拌过夜,TLC检测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到23a(806mg,产率41%)。
中间体23b的合成:
将23a(806mg,1.74mmol)溶解于二氯甲烷(10mL)中,接着冰水浴下加入5mL的三氟乙酸,整个体系在室温下搅拌2小时,TLC检测原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,用二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,得到23b(770mg,粗品)。
中间体23c的合成:
将23b(770mg,粗品)溶解于二氯甲烷(10mL)中,接着加入三乙胺(883μL,6.35mmol),乙酸酐(297μL,3.18mmol)。体系在室温下搅拌,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(乙酸乙酯),得到23c(490mg,产率69%)。
中间体23d的合成:
将23c(100mg,0.25mmol)溶解于1,4二氧六环(10mL)和水(5mL)中,接着加入4-(4,4,5,5-四甲基-1,3,2-二氧杂硼硼烷-2-基)-1H-吡咯并[2,3-b]吡啶-1-羧酸叔丁酯(167mg,0.49mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯(15mg,0.02mmol)和碳酸钾(68mg,0.49mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。反应直接经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到23d(122mg,产率99%)。
终产物23的合成:
将23d(122mg,0.24mmol)溶解于二氯甲烷(10mL)中,接着冰水浴下加入5mL的三氟乙酸,整个体系在室温下搅拌2小时,TLC检测原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,用二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到终产物23(57mg,产率59%)。MS m/z(ESI):396.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ11.97(s,1H),10.70(s,1H),8.42(s,1H),8.41(s,1H),8.36(d,J=4.8Hz,1H),7.79(d,J=7.8Hz,1H),7.63-7.62(m,1H),7.23(d,J=4.2Hz,1H),6.47(s,1H),3.58-3.56(m,1H),2.62-2.61(m,1H),1.99(d,J=4.8Hz,1H),1.91-1.89(m,6H),1.39-1.21(m,3H),1.20-1.09(m,1H).
实施例24:
Figure PCTCN2022094496-appb-000037
中间体24a的合成:
将(1-甲基-1H-吡咯并[2,3-b]吡啶-4-基)硼酸(176mg,1.00mmol)和23a(461mg,1.00mmol)溶解于20mL 1,4-二氧六环中,依次加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(73mg,0.10mmol)和碳酸钾(414mg,3.00mmol)。将体系用氮气保护,在100℃下搅拌反应4.5小时,TLC监测原料反应完毕。停止反应并向反应体系中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=4:1-石油醚:乙酸乙酯=1:1),得到24a(400mg,产率86%)。
终产物24的合成:
将24a(400mg,0.86mmol)溶解到二氯甲烷(25mL)中,在室温下加入三氟乙酸(3mL),室温下反应4小时,TLC监测原料无剩余,用碳酸钠饱和溶液洗涤至弱碱性,二氯甲烷萃取水相(30mL×3),合并有机相,无水硫酸钠干燥。浓缩溶剂至体积为15mL,加入三乙胺(2mL)和乙酸酐(2mL)在室温反应30分钟,TLC监测原料无剩余,饱和碳酸钠溶液洗涤反应体系,浓缩溶剂,经过硅胶柱层析分离(二氯甲烷:甲醇=50:1-10:1),得到终产物24(169mg,收率48%)。MS m/z(ESI):410.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.69(s,1H),8.50(s,1H),8.41(s,1H),8.40(s,1H),7.77(d,J=7.8Hz,1H),7.68(d,J=3.6Hz,1H),7.26(d,J=4.8Hz,1H),6.47(d,J=2.4Hz,1H),3.89(s,3H),3.58-3.56(m,1H),2.64-2.60(m,1H),1.91-1.89(m,1H),1.79-1.77(m,6H),1.30-1.26(m,3H),1.09-1.07(m,1H).
实施例25:
Figure PCTCN2022094496-appb-000038
终产物25的合成:
将5b(0.61g,1.69mmol)溶解于二氯甲烷(35mL)中,接着加入甲基磺酰氯(0.29g,2.54mmol)和三乙胺(0.34g,3.38mmol),体系在室温下搅拌,TLC跟踪监测直至原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物25(0.41g,产率55%)。MS m/z(ESI):440.1[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.60(s,1H),8.33(s,1H),8.08(d,J=4.8Hz,1H),7.34(d,J=7.2Hz,1H),7.10(d,J=7.8Hz,2H),6.92(d,J=8.4Hz,1H),3.80(s,3H),3.13-3.11(m,1H),2.92(s,3H),2.62-2.57(m,1H),2.03(d,J=12Hz,1H),2.25(d,J=12Hz,1H),1.77-1.73(m,2H),1.36-1.11(m,4H).
实施例26:
Figure PCTCN2022094496-appb-000039
中间体26a的合成:
将5-氟-4-碘吡啶-2-胺(0.50g,2.10mmol)溶解于乙二醇二甲醚(20mL)和水(4mL)中,接着加入4,5-二氟-2-甲氧基苯基硼酸(0.39g,2.10mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯(0.15g,0.21mmol)和碳酸钾(0.87g,6.3mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。反应直接经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到26a(0.40g,产率75%)。
中间体26b的合成:
将26a(0.30g,1.18mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.35g,1.42mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(0.54g,1.42mmol)和N,N-二甲基乙基胺(0.30g,2.36mmol),整个体系在室温下搅拌过夜,TLC检测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到26b(0.33g,产率58%)。
中间体26c的合成:
将26b(0.20g,0.42mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入的三氟乙酸(2mL),整个体系在室温下搅拌过夜,TLC检测直至原料无剩余。向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1),得到26c(0.11g,产率70%)。
终产物26的合成:
将26c(0.11g,0.29mmol)溶解于二氯甲烷(35mL)中,接着加入甲磺酰氯(0.40g,0.35mmol)和三乙胺(44mg,0.46mmol)。体系在室温下搅拌,TLC跟踪监测直至原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物26(40mg,产率30%)。MS m/z(ESI):458.1[M+H] +
1H NMR(600MHz,CDCl 3)δ8.59(s,1H),8.31(s,1H),8.14(d,J=10.8Hz,1H),7.17(d,J=9.0Hz,1H),6.85-6.82(m,1H),5.46(s,1H),3.88(s,1H),3.83(s,3H),2.82(s,1H),2.48(s,1H),2.27-2.25(m,1H),2.05-1.94(m,5H),1.45-1.16(m,4H).
实施例27:
Figure PCTCN2022094496-appb-000040
终产物27的合成:
将5b(0.61g,1.69mmol)溶解于二氯甲烷(35mL)中,接着加入噻吩磺酰氯(0.46g,2.53mmol)和三乙胺(0.34g,3.38mmol)。体系在室温下搅拌过夜,TLC跟踪监测直至原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物27(0.52g,产率61%)。MS m/z(ESI):508.1[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.55(s,1H),8.31(s,1H),8.03(s,1H),7.96(d,J=7.2Hz,1H),7.89(s,1H),7.57(s,1H),7.32(d,J=7.2Hz,1H),7.15(s,1H),7.08(d,J=11.4Hz,1H),6.89(d,J=8.4Hz,1H),4.02(s,3H),3.04-3.03(m,1H),1.78-1.62(m,5H),1.17-1.08(m,4H).
实施例28:
Figure PCTCN2022094496-appb-000041
终产物28的合成:
将5b(79mg,0.22mmol)溶解于乙腈(5mL)中,加入碳酸钠(47mg,0.44mmol),和苄溴(37mg,0.22mmol)。体系处于室温反应过夜。TLC监控显示无原料剩余。浓缩除去溶剂,粗品经薄板层析分离纯化(二氯甲烷:甲醇=10:1),得到终产物28(30mg,产率30%)。MS:(m/z,ESI):451.2[M+H] +
1H NMR(600MHz,CD 3OD)δ8.16(s,1H),8.09(d,J=4.8Hz,1H),7.37-7.32(m,4H),7.30-7.26(m,2H),6.93(d,J=11.4Hz,1H),6.80(dd,J=8.4Hz,1H),3.86(s,2H),3.81(s,3H),2.71-2.67(m,1H),2.51-2.47(m,1H),2.20-2.18(m,1H),2.06-2.04(m,1H),1.89-1.85(m,2H),1.49-1.39(m,3H),1.21-1.16(m,1H).
实施例29:
Figure PCTCN2022094496-appb-000042
终产物29的合成:
将5b(0.61g,1.69mmol)溶解于N,N-二甲基甲酰胺(35mL)中,接着加入溴乙基甲基醚(0.26g,1.86mmol)和碳酸钾(0.47g,3.38mmol)。体系在室温下搅拌,TLC监测原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物29(0.42g,产率59%)。MS m/z(ESI):420.2[M+H] +
1H NMR(600MHz,CDCl 3)δ8.25(d,J=5.4Hz,1H),8.12(s,1H),7.25(d,J=6.6Hz,1H),6.75-6.70(m,2H),3.80(s,3H),3.57(t,J=4.8Hz,2H),3.36(s,3H),2.90(t,J=4.8Hz,2H),2.70(s,1H),2.37(t,J=5.4Hz,1H),2.26(d,J=12.0Hz,1H),2.05-1.90(m,3H),1.52-1.22(m,4H).
实施例30:
Figure PCTCN2022094496-appb-000043
终产物30的合成:
将5b(155mg,0.43mmol)溶解于乙腈(5mL)中,加入三乙胺(86mg,0.86mmol),溴乙醇(54mg,0.43mmol)和碘化钠(10mg,0.06mmol)。体系处于60℃反应12小时。TLC监控显示无原料剩余。降温后,浓缩除去溶剂。粗品经薄板层析分离纯化(二氯甲烷:甲醇:三乙胺=40:2:1),得到终产物30(30mg,产率17%)。MS m/z(ESI):406.2[M+H] +
1H NMR(600MHz,CD 3OD)δ8.18(s,1H),8.08(d,J=4.8Hz,1H),7.28(d,J=7.2Hz,1H),6.92(d,J=11.4Hz,1H),6.79(d,J=8.4,1H),3.82-3.80(m,5H),3.20-3.25(m,1H),3.20-3.18(m,2H),2.65-2.62(m,1H),2.31-2.29(m,1H),2.18-2.16(m,1H),1.99-1.91(m,2H),1.70-1.64(m,1H),1.53-1.34(m,3H).
实施例31:
Figure PCTCN2022094496-appb-000044
终产物31的合成:
将5b(79mg,0.22mmol)溶解于二氧六环(5mL)中,再加入化合物2-溴-6-甲氧基吡啶(40mg,0.22mmol),三(二亚苄基丙酮)二钯(20mg,0.022mmol),4,5-双二苯基膦-9,9-二甲基氧杂蒽(12.7mg,0.022mmol)和碳酸铯(215mg,0.66mmol)。用氮气置换三次,使整个体系处于氮气的氛围下。体系处于120℃反应2小时。TLC监控显示原料无剩余。降温后,减压浓缩,粗品经薄板层析进行分离纯化(二氯甲烷:甲醇=10:1),得到终产物31(30mg,产率29%)。MS:(m/z,ESI):469.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.31(s,1H),8.07(d,J=5.4Hz,1H),7.32(d,J=7.2Hz,1H),7.24(d,J=7.8Hz,1H),7.08(d,J=11.4Hz,1H),6.89(d,J=8.4Hz,1H),6.33(d,J=7.2Hz,1H),5.97(d,J=7.8Hz,1H),5.80(d,J=7.8Hz,1H),3.76(s,3H),3.72(s,3H),3.63-3.61(m,1H),2.64-2.62(m,1H),2.06-2.04(m,1H),1.97-1.95(m,1H),1.79-1.77(m,2H),1.36-1.30(m,3H),1.11-1.09(m,1H).
实施例32:
Figure PCTCN2022094496-appb-000045
终产物32的合成:
将5b(61mg,0.17mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入氰乙酸(16mg,0.18mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(69mg,0.18mmol)和N,N-二异丙基乙胺(83uL,0.50mmol),整个体系在室温下搅拌,反应10小时,TLC监测跟踪直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物32(50mg,产率71%)。MS m/z(ESI):429.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.58(s,1H),8.31(s,1H),8.20(d,J=7.8Hz,1H),8.06(d,J=5.4Hz,1H),7.32(t,J=7.2Hz,1H),7.08(d,J=11.4Hz,1H),6.90(t,J=8.4Hz,1H),3.77(s,3H),3.56(m,3H),2.61-2.57(m,1H),1.89-1.76(m,4H),1.30-1.07(m,4H).
实施例33:
Figure PCTCN2022094496-appb-000046
终产物33的合成:
将5b(0.16g,0.43mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入1-氰基-1-环丙烷羧酸(72mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物33(96mg,产率49%)。MS m/z(ESI):455.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.31(s,1H),8.05(d,J=4.8Hz,1H),8.09(d,J=7.8Hz,1H),7.32(dd,J=7.8Hz,J=7.2Hz,1H),7.09(d,J=11.4Hz,1H),6.90(dd,J=8.4Hz,J=7.8Hz,1H),3.77(s,3H),3.68-3.56(m,1H),3.59-3.55(m,1H),1.81-1.67(m,4H),1.53-1.46(m,5H)),1.28-1.23(m,3H).
实施例34:
Figure PCTCN2022094496-appb-000047
终产物34的合成:
将5b(0.16g,0.43mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入1-氰基环丁烷羧酸(81mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物34(119mg,产率59%)。MS m/z(ESI):469.2[M+H] +
1H NMR(600MHz,DMSO-d6)δ10.56(s,1H),8.31(s,1H),8.05(d,J=4.8Hz,1H),8.09(d,J=7.8Hz,1H),7.34-7.31(m,1H),7.09(s,1H),6.91-6.88(m,1H),3.77(s,3H),3.68-3.56(m,1H),2.59-2.55(m,1H),1.81-1.67(m,4H),1.53-1.46(m,5H),1.30-1.23(m,5H).
实施例35:
Figure PCTCN2022094496-appb-000048
中间体35a的合成:
将2,2'-二溴二乙醚(12.85g,55.60mmol)溶解于N,N-二甲基甲酰胺(10mL)中,接着加入氰乙酸甲酯(5.00g,50.45mmol)和1,8-二氮杂双环[5.4.0]十一碳-7-烯(11.50g,75.68mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在85℃下搅拌,反应4小时,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,经柱层析分离纯化(石油谜:乙酸乙酯=1:1),得到35a(5.00g,产率59%)。
中间体35b的合成:
将35a(2.37g,14.00mmol)溶解于乙醇(36mL)和水(5mL)中,接着加入氢氧化钠(2.24g,56.00mmol),整个体系在室温下搅拌4小时,TLC检测原料无剩余。向反应液中加入水(100mL),碳酸氢钠水溶液调PH至8,乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,得到35b(1.50g,产率69%)。
终产物35的合成:
将35b(123mg,0.79mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入(1S,3R)-3-氨基-N-(5-氟-4-(4-氟-2-甲氧基苯基)吡啶-2-基)环己酰胺(260mg,0.72mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(548mg,1.44mmol)和N,N-二异丙基乙胺(357μL,2.16mmol),整个体系在室温下搅拌,反应10小时,TLC监测原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物35(214mg,产率60%)。MS m/z(ESI):499.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.58(s,1H),8.33(s,1H),8.17(d,J=7.8Hz,1H),8.08(d,J=6.0Hz,1H),7.35-7.33(m,1H),7.11-7.09(m,1H),6.93-6.89(m,1H),3.88-3.86(m,2H),3.78(s,3H),3.66-3.63(m,1H),3.52-3.51(m,2H),2.63(m,1H),1.97-1.96(m,4H),1.85-1.75(m,4H),1.38-1.21(m,4H).
实施例36:
Figure PCTCN2022094496-appb-000049
终产物36的合成:
将三光气(33mg,0.11mmol)溶解于二氯甲烷(2mL)中,接着滴加5b(61mg,0.17mmol)和三乙胺(22mg,0.22mmol)溶解于二氯甲烷(2mL)的溶液,整个体系在室温下搅拌,反应2小时,接着加入甲醇溶液(2mL),继续反应3小时,TLC监测直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物36(30mg,产率42%)。MS m/z(ESI):420.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.31(s,1H),8.06(d,J=5.4Hz,1H),7.33(d,J=7.8Hz,1H),7.10-7.07(m,2H),6.90(d,J=8.4Hz,1H),3.77(s,3H),3.48(s,3H),3.32(m,1H),2.56(m,1H),1.87-1.71(m,4H),1.31-1.06(m,4H).
实施例37:
Figure PCTCN2022094496-appb-000050
终产物37的合成:
将三光气(33mg,0.11mmol)溶解于二氯甲烷(2mL)中,接着滴加溶解有5b(61mg,0.17mmol) 和三乙胺(22mg,0.22mmol)的二氯甲烷溶液,整个体系在室温下搅拌,反应2小时,接着加入33%甲胺醇溶液3mL,继续反应3小时,TLC监测直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物37(15mg,产率21%)。MS m/z(ESI):419.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.31(s,1H),8.06(d,J=5.4Hz,1H),7.33(t,J=8.4Hz,1H),7.10-7.07(m,1H),6.91-6.88(m,1H),5.78(d,J=8.4Hz,1H),5.56(d,J=4.8Hz,1H),3.76(s,3H),3.36-3.34(m,1H),2.65-2.55(m,1H),2.51(s,3H),1.88-1.73(m,4H),1.28-0.82(m,4H).
实施例38:
Figure PCTCN2022094496-appb-000051
终产物38的合成:
将5b(0.61g,1.69mmol)溶解于二氯甲烷(35mL)中,接着加入二甲氨基甲酰氯(0.27g,2.54mmol)和三乙胺(0.34g,3.38mmol)。体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=100:1-40:1),得到终产物38(0.38g,产率53%)。MS m/z(ESI):433.1[M+H] +
1H NMR(600MHz,CDCl 3)δ8.28-8.25(m,2H),8.11(s,1H),7.27-7.24(m,2H),6.76-6.70(m,2H),3.81(s,3H),3.76-3.73(m,1H),2.88(s,6H),2.47-2.43(m,1H),2.30(d,J=12.0Hz,1H),2.02-1.90(m,3H),1.47-1.12(m,4H).
实施例39:
Figure PCTCN2022094496-appb-000052
中间体39a的合成:
将5b(0.12g,0.33mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入(S)-2-((叔丁氧羰基)氨基)-2-(4-羟基苯基)乙酸(0.10g,0.36mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(0.15g,0.40mmol),N,N-二异丙基乙胺(0.09g,0.66mmol),室温反应8小时,TLC监测原料无剩余。在反应液中加入水(20mL),乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥。有机相直接柱层析分离(二氯甲烷:甲醇=10:1),得到39a(0.12g,产率60%)。
终产物39的合成:
将39a(0.12g,0.20mmol)溶解于二氯甲烷(20mL)中,加入三氟乙酸(4mL),室温反应4小时,TLC监测原料无剩余。将反应液用水洗涤(20mL×3),合并水相,水相用碳酸钠调节pH为8-9,二氯甲烷萃取(30mL×3),合并有机相,无水硫酸钠干燥。减压蒸除有机相,得到终产物39(0.04g,产率39%)。MS m/z(ESI):511.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),9.23(s,1H),8.32(s,1H),8.08(d,J=5.4Hz,1H),7.90-7.89(m,1H),7.36-7.34(m,1H),7.17-7.15(m,2H),7.12-7.10(m,1H),6.93-6.91(m,1H),6.68-6.66(m,2H),4.19(s,1H),4.04(d,J=7.2Hz,2H),3.80(s,3H),3.58-3.57(m,1H),2.59-2.57(m,1H),1.80-1.76(m,4H),1.33-1.25(m,4H).
实施例40:
Figure PCTCN2022094496-appb-000053
终产物40的合成:
将5b(0.20g,0.55mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入N,N-二甲基甘氨酸(67mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物40(100mg,产率41%)。MS m/z(ESI):447.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.31(s,1H),8.06(d,J=4.2Hz,1H),7.58(d,J=8.4Hz,1H),7.32(dd,J=7.8Hz,J=7.2Hz,1H),7.08(d,J=10.8Hz,1H),6.90(dd,J=7.8Hz,J=7.8Hz,1H),3.76(s,3H),3.65-3.63(m,1H),2.84-2.77(m,2H),2.66-2.60(m,1H),2.16(s,6H),1.85-1.84(m,1H),1.75-1.69(m,3H),1.41-1.35(m,1H),1.32-1.23(m,3H).
实施例41:
Figure PCTCN2022094496-appb-000054
化合物41的合成:
将三光气(0.027g,0.09mmol)溶于无水四氢呋喃(20mL)中,加入3-甲基异恶唑-5-胺(0.027g,0.28mmol)的四氢呋喃溶液,三乙胺(0.028g,0.28mmol),室温反应6小时。在反应液中加入5b(0.10g,0.28mmol)的四氢呋喃溶液,三乙胺(0.028g,0.28mmol),加热至70℃,反应6小时。TLC监测无原料剩余,停止反应,降至室温,反应液柱层析分离(石油醚:乙酸乙酯=1:1),得到终产物41(45mg,产率33%)。MS m/z(ESI):486.2[M+H] +
1H NMR(600MHz,CDCl 3)δ8.65(s,1H),8.29(s,1H),8.12(s,1H),6.76-6.71(m,2H),6.23(s,2H),5.43(s,1H),4.03-4.01(m,1H),3.81(s,3H),2.54-2.53(m,1H),2.35(s,3H),2.36-2.24(m,2H),2.05-1.94(m,3H),1.56-1.50(m,3H).
实施例42:
Figure PCTCN2022094496-appb-000055
终产物42的合成:
将5b(100mg,0.28mmol)溶解于二氯甲烷(10mL)中,然后加入三氟乙酸酐(56mg,0.55mmol),和三乙胺(56mg,0.55mmol),室温反应1.5小时,TLC监测原料无剩余。反应液中加入饱和碳酸氢钠(30mL),二氯甲烷萃取(20mL×3),合并有机相,饱和氯化钠洗涤(20mL×2),无水硫酸钠干燥,减压除去溶剂,粗品经柱层析纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物42(100mg,产率78%)。MS m/z(ESI):458.1[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.59(s,1H),9.33(d,J=8.4Hz,1H),8.31(s,1H),8.05(d,J=6Hz,1H),7.32(dd,J=7.8Hz,J=7.2Hz,1H),7.08(m,1H),6.89(m,1H),3.76(s,3H),3.70-3.68(m,1H),2.63-2.59(m,1H),1.86-1.83(m,1H),1.80-1.76(m,2H),1.51-1.44(m,1H),1.33-1.24(m,4H).
实施例43:
Figure PCTCN2022094496-appb-000056
化合物43的合成:
将5b(0.11g,0.30mmol),2-吡啶羧酸(0.04g,0.33mmol)溶于N,N-二甲基甲酰胺(15mL)中,加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(0.14g,0.36mmol),N,N-二异丙基乙胺(0.08g,0.6mmol),室温反应8小时,TLC监测原料无剩余。在反应液中加入水(30mL),析出固体,过滤,得到终产物43(0.07g,50%)。MS m/z(ESI):467.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.63(s,1H),8.63-8.62(m,2H),8.31(s,1H),8.07(d,J=5.4Hz,1H),8.01(d,J=7.8Hz,1H),7.98-7.95(m,1H),7.58-7.56(m,1H),7.34-7.31(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),3.89-3.87(m,1H),3.77(s,3H),2.71-2.63(m,1H),1.92-1.91(m,1H),1.82-1.77(m,3H),1.61-1.55(m,1H),1.45-1.28(m,3H).
实施例44:
Figure PCTCN2022094496-appb-000057
终产品化合物44的合成:
将5b(188mg,0.52mmol)和1-乙基-3-甲基-1H-吡唑-4-羧酸(161mg,1.04mmol)溶解于N,N-二甲基甲酰胺(5mL)中,依次加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(395mg,1.04mmol)和二异丙基乙胺(0.26mL,1.56mmol),将反应体系在室温反应15小时,TLC检测原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,粗品经柱层析纯化(石油醚:乙酸乙酯=4:1-2:1),得到终产物44(230mg,收率88%)。MS m/z(ESI):498.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.63(s,1H),8.34(s,1H),8.22(d,J=8.4Hz,1H),8.09(d,J=6.0Hz,1H),7.35(m,1H),7.11(m,1H),6.92(m,1H),6.59(s,1H),4.38(dd,J=7.2Hz,J=3.0Hz,2H),3.79(s,4H),2.15(s,3H),2.00(d,J=11.4Hz,1H),1.84-1.80(m,4H),1.49-1.46(m,2H),δ1.32-1.30(m,5H).
实施例45:
Figure PCTCN2022094496-appb-000058
终产物45的合成:
将5b(50mg,0.138mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入羟乙酸(16mg,0.21mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(80mg,0.21mmol)和N,N-二异丙基乙胺(69μL,0.42mmol),整个体系在室温下搅拌,反应10小时,TLC监测直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物45(30mg,产率52%)。MS m/z(ESI):420.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.31(s,1H),8.06(d,J=5.4Hz,1H),7.56(d,J=9.0Hz,1H),7.34-7.31(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),5.38-5.36(m,1H),3.77-3.75(m,5H),3.66-3.63(m,1H),2.61-2.57(m,1H),1.83-1.68(m,4H),1.34-1.26(m,4H).
实施例46:
Figure PCTCN2022094496-appb-000059
终产物46的合成:
将5b(0.20g,0.55mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入乙氧基乙酸(68mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物46(96mg,产率39%)。MS m/z(ESI):448.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.58(s,1H),8.33(s,1H),8.07(d,J=5.4Hz,1H),7.59(d,J=8.4Hz,1H),7.35-7.33(m,1H),7.11-7.09(m,1H),6.93-6.90(m,1H),3.78-3.76(m,5H),3.69-3.67(m,1H),3.47-3.44(m,2H),2.63-2.61(m,1H),1.84-1.69(m,4H),1.47-1.43(m,1H),1.30-1.28(m,6H).
实施例47:
Figure PCTCN2022094496-appb-000060
终产物47的合成:
将45(0.20g,0.48mmol)溶解于二氯甲烷(30mL)中,接着加入乙酰氯(45mg,0.57mmol),三乙胺(97mg,0.96mmol),体系在室温下搅拌1.5小时,TLC监测原料无剩余。减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物47(100mg,产率45%)。MS m/z(ESI):462.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.58(s,1H),8.33(s,1H),8.07(d,J=5.4Hz,1H),7.59(d,J=8.4Hz,1H),7.35-7.34(m,1H),7.33-7.11(m,1H),7.09-6.90(m,1H),4.39(s,2H),3.78(s,3H),3.65-3.63(m,1H),2.66-2.60(m,1H),2.58(s,3H),1.85-1.84(m,1H),1.75-1.69(m,3H),1.39-1.24(m,3H),1.16-1.14(m,1H).
实施例48:
Figure PCTCN2022094496-appb-000061
终产物48的合成:
将5b(0.20g,0.55mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入L-乳酸(58.5mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物48(96mg,产率40%)。MS m/z(ESI):434.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.57(s,1H),8.31(s,1H),8.05(d,J=6Hz,1H),7.51(d,J=8.4Hz,1H),7.34-7.31(m,1H),7.10-7.07(m,1H),6.91-6.88(m,1H),5.38(d,J=5.4Hz,1H),3.92-3.88(m,1H),3.77(s,3H),3.62-3.59(m,1H),2.60-2.56(m,1H),1.83-1.81(m,1H),1.77-1.72(m,2H),1.41-1.35(m,1H),1.31-1.26(m,4H),1.23(d,J=13.2Hz,3H).
实施例49:
Figure PCTCN2022094496-appb-000062
终产物49的合成:
将5b(0.20g,0.55mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入D-乳酸(58.5mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物49(100mg,产率42%)。MS m/z(ESI):434.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.57(s,1H),8.30(s,1H),8.05(d,J=4.8Hz,1H),7.51(d,J=7.8Hz,1H),7.33(t,J=7.8Hz,1H),7.08(d,J=11.4Hz,1H),6.89(t,J=7.8Hz,1H),5.38(d,J=5.4Hz,1H),3.92-3.88(m,1H),3.77(s,3H),3.62-3.59(m,1H),2.60-2.56(m,1H),1.81(d,J=12Hz,1H),1.77-1.72(m,2H),1.67(d,J=11.4Hz,1H),1.41-1.35(m,1H),1.31-1.25(m,4H),1.23(s,3H).
实施例50:
Figure PCTCN2022094496-appb-000063
终产物50的合成:
将5b(50mg,0.138mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入3-羟丙酸(19mg,0.21mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(79mg,0.21mmol)和N,N-二异丙基乙胺(69μL,0.42mmol),整个体系在室温下搅拌,反应10小时,TLC监测直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物50(32mg,产率53%)。MS m/z(ESI):434.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.31(s,1H),8.06-8.05(d,J=5.4Hz,1H),7.57-7.55(d,J=9.0Hz,1H),7.34-7.31(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),5.38-5.36(t,J=5.4Hz,1H),3.77-3.75(m,5H),3.66-3.63(m,1H),2.61-2.56(m,3H),1.83-1.68(m,4H),1.34-1.26(m,4H).
实施例51:
Figure PCTCN2022094496-appb-000064
终产物51的合成:
将5b(50mg,0.138mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入1-羟基环丙烷羧酸(21mg,0.21mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(79mg,0.21mmol)和N,N-二异丙基乙胺(69μL,0.42mmol),整个体系在室温下搅拌,反应10小时,TLC监测直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物51(36mg,产率59%)。MS m/z(ESI):446.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.61(s,1H),8.34(s,1H),8.09-8.08(m,1H),7.73-7.71(m,1H),7.37-7.34(m,1H),7.13-7.10(m,1H),6.95-6.91(m,1H),3.80(s,3H),2.61-2.55(m,2H),1.85-1.72(m,4H),1.50-1.47(m,1H),1.32-1.30(m,3H),1.00-0.98(m,2H),0.81-0.80(m,2H).
实施例52:
Figure PCTCN2022094496-appb-000065
终产物52的合成:
将5b(50mg,0.138mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入3-氧杂环丁烷羧酸(21mg,0.21mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(79mg,0.21mmol)和N,N-二异丙基乙胺(69μL,0.42mmol),整个体系在室温下搅拌,反应10小时,TLC监测直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物52(36mg,产率59%)。MS m/z(ESI):446.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.59(s,1H),8.33(s,1H),8.07-8.06(m,1H),7.98-7.97(m,1H),7.35-7.32(m,1H),7.10-7.08(m,1H),6.93-6.91(m,1H),4.61-4.56(m,2H),3.78(s,3H),3.69-3.67(m,2H),3.42-3.40(m,1H),2.69-2.55(m,2H),1.89-1.87(m,1H),1.77-1.75(m,3H),1.29-1.25(m,3H),1.18-1.09(m,1H).
实施例53:
Figure PCTCN2022094496-appb-000066
终产物53的合成:
将5b(50mg,0.138mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入1-甲基环丙烷-1-羧酸(21mg,0.21mmol)、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(79mg,0.21mmol)和N,N-二异丙基乙胺(69μL,0.42mmol),整个体系在室温下搅拌,反应10小时,TLC监测直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物53(40mg,产率65%)。MS m/z(ESI):444.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.57(s,1H),8.32(s,1H),8.08-8.06(m,1H),7.34-7.32(m,1H),7.24-7.22(m,1H),7.11-7.09(m,1H),6.93-6.91(m,1H),3.78(s,3H),3.66-3.58(m,1H),2.59-2.50(m,1H),1.80-1.69(m,4H),1.47-1.44(m,1H),1.30-1.22(m,6H),0.92-0.90(m,2H),0.46-0.44(m,2H).
实施例54:
Figure PCTCN2022094496-appb-000067
终产物54的合成:
将5b(50mg,0.138mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入1-氟环丙烷羧酸(22mg,0.21mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(79mg,0.21mmol)和N,N-二异丙基乙胺(69μL,0.42mmol),整个体系在室温下搅拌,反应10小时,TLC监测直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物54(37mg,产率60%)。MS m/z(ESI):448.2[M+H] +
1H NMR(600MHz,CDCl 3)δ8.23-8.20(m,1H),8.13(s,1H),7.86(s,1H),7.27-7.25(m,1H),6.80-6.70(m,2H),6.40-6.25(m,1H),4.00-3.85(m,1H),3.82(s,3H),2.50-2.40(m,1H),2.35-2.25(m,1H),2.10-1.90(m,3H),1.55-1.45(m,3H),1.40-1.30(m,2H),1.25-1.15(m,3H).
实施例55:
Figure PCTCN2022094496-appb-000068
终产物55的合成:
将5b(50mg,0.138mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入1-三氟甲基环丙烷-1-甲酸(32mg,0.21mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(79mg,0.21mmol)和N,N-二异丙基乙胺(69μL,0.42mmol),整个体系在室温下搅拌,反应10小时,TLC监测直至原料无剩余。向反应液中加入水(50mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物55(45mg,产率65%)。MS m/z(ESI):498.2[M+H] +
1H NMR(600MHz,CDCl 3)δ8.23-8.20(m,1H),8.13(s,1H),7.86(s,1H),7.27-7.25(m,1H),6.79-6.69(m,2H),6.05-5.95(m,1H),4.00-3.86(m,1H),3.82(s,3H),2.50-2.35(m,1H),2.30-2.20(m,1H),2.05-1.90(m,3H),1.56-1.35(m,5H),1.21-1.15(m,3H).
实施例56:
Figure PCTCN2022094496-appb-000069
中间体56a的合成:
将甘氨酸(500mg,6.70mmol)溶于二氧六环(10mL)和水(10mL),加入三乙胺(1.40g,14.40mmol)和二碳酸二叔丁酯(1.74g,8.00mmol)。体系处于室温反应过夜。TLC监控显示原料反应完全。将体系浓缩,加入碳酸钠水溶液(100mL),乙酸乙酯洗涤(100mL×3),将水相用稀盐酸调pH为3,乙酸乙酯萃取(100mL×3),合并有机相,无水硫酸钠干燥。过滤后,滤液浓缩,得到56a(1.00g,产率85%)。
中间体56b的合成:
将56a(46mg,0.26mmol)溶于N,N-二甲基甲酰胺(3mL)中,加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(125mg,0.33mmol)以及二异丙基乙胺(57mg,0.44mmol),混合物搅拌10分钟后,加入5b(78mg,0.22mmol)。体系处于室温反应过夜。TLC监控显示原料反应完全。将体系中加入水(100mL),乙酸乙酯萃取(100mL×3),合并有机相后,饱和食盐水洗涤(50mL×3)。无水硫酸钠干燥,过滤后滤液浓缩。粗品用薄板层析分离纯化(二氯甲烷:甲醇=10:1),得到56b(80mg,产率70%)。
终产物56的合成:
将56b(80mg,0.22mmol)溶解于二氯甲烷(4mL)中,加入三氟乙酸(1mL)。体系处于室温反应2小时,TLC监控显示反应完全。将体系浓缩,粗品经薄板层析进行分离纯化(二氯甲烷:甲醇=10:1),得到终产物56(40mg,产率43%)。MS m/z(ESI):419.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.59(s,1H),8.31(s,1H),8.05(d,J=6.0Hz,1H),7.94(br,2H),7.32-7.30(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),3.76(s,3H),3.64-3.59(m,1H),3.15(d,J=4.8Hz,2H),2.62-2.59(m,1H),1.89-1.87(m,1H),1.79-1.77(m,3H),1.34-1.28(m,3H),1.18-1.10(m,1H).
实施例57:
Figure PCTCN2022094496-appb-000070
终产物57的合成:
将化合物56(205mg,0.49mmol)溶解于二氯甲烷(35mL)中,接着加入乙酸酐(149mg,1.47mmol)和三乙胺(148mg,1.47mmol),体系在室温下搅拌4小时,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到终产物57(138mg,产率61%)。MS m/z(ESI):461.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.60(s,1H),8.33(s,1H),8.07(d,J=6.0Hz,1H),7.95-7.92(m,2H),7.35-7.33(m,1H),7.11(d,J=1.8Hz,1H),6.93-6.90(m,1H),4.09(s,2H),3.79(s,3H),3.58-3.56(m,1H),2.59-2.50(m,1H),1.86-1.73(m,4H),1.83(s,3H),1.39-1.14(m,4H).
实施例58:
Figure PCTCN2022094496-appb-000071
终产物58的合成:
将5b(0.20g,0.55mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入N-乙酰-L-亮氨酸(112mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物58(190mg,产率67%)。MS m/z(ESI):517.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.60(s,1H),8.33(s,1H),8.07(d,J=6.0Hz,1H),7.94(dd,J=10.2,Hz,J=8.4Hz,2H),7.35(dd,J=7.8Hz,J=7.2Hz,1H),7.11-7.10(m,1H),6.93-6.90(m,1H),4.24-4.23(m,1H),3.79(s,3H),3.58-3.56(m,1H),2.59-2.50(m,1H),1.83-1.71(m,7H),1.53-1.51(m,1H),1.38-1.36(m,2H),1.31-1.28(m,6H),0.91(m,6H).
实施例59:
Figure PCTCN2022094496-appb-000072
终产物59的合成:
将5b(200mg,0.55mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入N-乙酰-D-亮氨酸(112mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物59(181mg,产率64%)。MS m/z(ESI):517.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.59(s,1H),8.33(s,1H),8.07(d,J=6.0Hz,1H),7.93(dd,J=10.2,Hz,J=8.4Hz,2H),7.34(dd,J=7.8Hz,J=7.2Hz,1H),7.11-7.10(m,1H),6.92-6.89(m,1H),4.24-4.23(m,1H),3.80(s,3H),3.57-3.55(m,1H),2.59-2.50(m,1H),1.83-1.71(m,7H),1.53-1.51(m,1H),1.38-1.36(m,2H),1.30-1.27(m,6H),0.91(m,6H).
实施例60:
Figure PCTCN2022094496-appb-000073
中间体60a的合成:
将5b(500mg,1.385mmol)溶解于N,N-二甲基甲酰胺(15mL)中,接着加入1-Boc-4-哌啶甲酸(380mg,1.66mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(787mg,2.07mmol)和N,N-二异丙基乙胺(687uL,4.15mmol),整个体系在室温下搅拌,反应10小时,TLC监测原料无剩余。向反应液中加入水(50mL),搅拌30分钟过滤,无水硫酸钠干燥,得到60a(623mg,产率79%)。
中间体60b的合成:
将60a(623mg,1.09mmol)溶解于二氯甲烷(20mL)中,接着加入三氟乙酸(5mL),整个体系在室温下搅拌2小时,TLC检测原料无剩余。向反应液中加入水(100mL),碳酸氢钠调PH至8,乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,得到60b(500mg,产率97%)。
终产物60的合成:
将60b(300mg,0.63mmol)溶解于甲醇(10mL)中,接着加入1mL甲醛水溶液、三乙酰氧基硼氢化钠(269mg,1.27mmol),1滴乙酸。整个体系在室温下搅拌,反应10小时,TLC监测原料无剩余。向反应液中加入水(50mL),碳酸氢钠调PH至8,乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,得到终产物60(214mg,产率70%)。MS m/z(ESI):487.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.59(s,1H),8.34(s,1H),8.08(d,J=4.8Hz,1H),7.68(d,J=7.8Hz,1H),7.35(d,J=7.8Hz,1H),7.11(d,J=11.4Hz,1H),6.93(d,J=8.4Hz,1H),3.79(s,3H),3.58-3.56(m,1H),2.76-2.74(m,2H),2.62-2.58(m,1H),2.51(s,3H),1.99-1.96(m,1H),1.86-1.84(m,1H),1.81-1.72(m,5H),1.57-1.53(m,4H),1.31-1.24(m,3H),1.18-1.08(m,1H).
实施例61:
Figure PCTCN2022094496-appb-000074
终产物61的合成:
将5b(0.07g,0.19mmol)溶解于N,N-二甲基甲酰胺(10mL)中,加入环丙基乙酸(0.02g,0.21mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(0.09g,0.23mmol)和N,N-二异丙基乙胺(0.05g,0.38mmol),室温反应8小时,TLC监测原料无剩余。在反应液中加入水(20mL),乙酸乙酯萃取(15mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,柱层析分离纯化(二氯甲烷:甲醇=20:1),得到终产物61(0.04g,产率47%)。MS m/z(ESI):444.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.58(s,1H),δ8.33(s,1H),8.07(d,J=5.4Hz,1H),7.63(d,J=8.4Hz,1H),7.35-7.33(m,1H),7.11-7.01(m,1H),6.93-6.90(m,1H),3.79(s,3H),3.60-3.57(m,1H),2.60-2.61(m,1H),1.94-1.93(m,2H),1.88-1.75(m,4H),1.32-1.23(m,4H),1.10-1.08(m,1H),0.54-0.45(m,2H),0.19-0.15(m,2H).
实施例62:
Figure PCTCN2022094496-appb-000075
终产物62的合成:
将5b(0.07g,0.19mmol)溶于二氯甲烷(20mL)中,加入环丙基甲酸(0.02g,0.21mmol),1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(0.07g,0.38mmol)和4-二甲氨基吡啶(2.3mg,0.019mmol),室温反应8小时,TLC监测原料无剩余。将反应液依次用水、碳酸氢钠水溶液洗涤,无水硫酸钠干燥。有机相直接柱层析分离(二氯甲烷:甲醇=50:1),得到终产物62(0.04g,产率48%)。MS m/z(ESI):430.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.58(s,1H),8.32(s,1H),8.08(d,J=5.4Hz,1H),7.99(d,J=8.4Hz,1H),7.36-7.33(m,1H),7.11-7.09(m,1H),6.93-6.90(m,1H),3.79(s,3H),3.60-3.58(m,1H),2.59-2.50(m,1H),1.89-1.77(m,4H),1.50-1.47(m,1H),1.34-1.26(m,3H),1.10-1.08(m,1H),0.64-0.60(m,4H).
实施例63:
Figure PCTCN2022094496-appb-000076
终产物63的合成:
将5b(0.10g,0.28mmol)溶解于N,N-二甲基甲酰胺(25mL)中,接着加入2-环丙基-2-羰基乙酸(35mg,0.31mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(118mg,0.31mmol)和N,N-二异丙基乙胺(0.07g,0.55mmol)。体系在室温下搅拌,TLC监测直至原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=100:1-40:1)得到终产物63(54mg,产率43%)。MS m/z(ESI):458.1[M+H] +
1H NMR(600MHz,CDCl 3)δ8.55(s,1H),8.32(d,J=6.0Hz,1H),8.11(s,1H),7.28-7.25(m,1H),6.94(d,J=8.4Hz,1H),6.77-6.71(m,2H),3.86-3.82(m,1H),3.81(s,3H),3.10-3.08(m,1H),2.48-2.46(m,1H),2.25(d,J=12.0Hz,1H),2.04-1.95(m,3H),1.53-1.47(m,3H),1.26-1.24(m,1H),1.18-1.15(m,4H).
实施例64:
Figure PCTCN2022094496-appb-000077
终产物64的合成:
将5b(0.16g,0.44mmol)溶解于二氯甲烷(20mL)中,加入三乙胺(0.05g,0.53mmol),冰浴下加入丙烯酰氯(0.05g,0.53mmol),室温反应4小时,TLC监测原料无剩余。在反应液中加入水(30mL),二氯甲烷萃取(20mL×3),合并有机相,无水硫酸钠干燥,减压浓缩,粗品经柱层析分离(二氯甲烷:甲醇=50:1),得到终产物64(0.10g,产率55%)。MS m/z(ESI):416.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.59(s,1H),8.31(s,1H),8.06(d,J=6.0Hz,1H),8.02(d,J=7.8Hz,1H),7.34-7.31(m,1H),7.10-7.07(m,1H),6.91-6.89(m,1H),6.18-6.14(m,1H),6.06-6.03(m,1H),5.55-5.53(m,1H),3.77(s,3H),3.65-3.63(m,1H),2.61-2.60(m,1H),1.90-1.77(m,4H),1.34-1.08(m,4H).
实施例65:
Figure PCTCN2022094496-appb-000078
终产物65的合成:
将5b(180mg,0.50mmol)和丙炔酸(70mg,1.00mmol)溶解于N,N-二甲基甲酰胺(20mL)中,依次加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(380mg,1.00mmol)和N,N-二异丙基乙胺(0.25mL,1.50mmol),将反应体系在室温反应15小时,TLC检测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,粗品经柱层析纯化(石油醚:乙酸乙酯=4:1-2:1),得到终产物65(200mg,收率97%)。MS m/z(ESI):414.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.60(s,1H),8.73(d,J=7.8Hz,1H),8.33(s,1H),8.08(d,J=5.4Hz,1H),7.35(dd,J=8.4Hz,J=7.8Hz,1H),7.11(dd,J=11.4Hz,J=2.4Hz,1H),6.92-6.90(m,1H),4.12(s,1H),3.79(s,3H),3.64-3.62(m,1H),2.61-2.57(m,1H),1.85-1.74(m,4H),1.35-1.23(m,4H).
实施例66:
Figure PCTCN2022094496-appb-000079
终产物66的合成:
将5b(0.25g,0.69mmol)溶于N,N-二甲基甲酰胺(20mL)中,加入反式-4-二甲基胺基巴豆酸盐酸盐(0.13g,0.76mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(0.32g,0.83mmol),N,N-二异丙基乙胺(0.27g,2.07mmol),室温反应8小时,TLC监测原料无剩余。在反应液中加入水(30mL),乙酸乙酯萃取(20mL×5),合并有机相,无水硫酸钠干燥。有机相直接柱层析分离(二氯甲烷:甲醇=10:1),得到终产物66(0.18g,产率55%)。MS m/z(ESI):472.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.61(s,1H),8.33(s,1H),8.08(d,J=6.0Hz,1H),8.01(d,J=6.0Hz,1H),7.35-7.33(m,1H),7.11-7.09(m,1H),6.93-6.90(m,1H),6.56-6.51(m,1H),6.04(d,J=15.0Hz,1H),3.78(s,3H),3.66(m,1H),3.14(s,2H),2.64-2.60(m,1H),2.25(s,6H),1.91-1.90(m,1H),1.84-1.78(m,3H),1.35-1.13(m,4H).
实施例67:
Figure PCTCN2022094496-appb-000080
终产物67的合成:
将26c(99mg,0.26mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入1-氰基-1-环丙烷羧酸(44mg,0.40mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(152mg,0.40mmol)和N,N-二异丙基乙胺(131μL,0.79mmol),整个体系在室温下搅拌过夜,TLC检测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物67(50mg,产率41%)。MS m/z(ESI):473.2[M+H] +
1H NMR(600MHz,CDCl 3)δ8.32-8.31(m,1H),8.13(s,1H),7.18-7.15(m,1H),6.84-6.81(m,1H),6.38-6.37(m,1H),3.90-3.85(m,1H),3.79(s,3H),2.49-2.47(m,1H),2.27-2.25(m,1H),2.01-1.94(m,3H),1.70-1.62(m,2H),1.60-48(m,5H),1.32-1.23(m,1H).
实施例68:
Figure PCTCN2022094496-appb-000081
终产物68的合成:
将26c(99mg,0.26mmol)溶解于N,N-二甲基甲酰胺(5mL)中,接着加入羟乙酸(20mg,0.26mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(152mg,0.40mmol)和N,N-二异丙基乙胺(131μL, 0.79mmol),整个体系在室温下搅拌过夜,TLC检测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物68(50mg,产率44%)。MS m/z(ESI):438.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.60(s,1H),8.35(s,1H),8.08(d,J=6.0Hz,1H),7.58-7.56(m,1H),7.53-7.51(m,1H),7.37-7.34(m,1H),5.38(t,J=6.0Hz,1H),3.82(s,5H),3.74-3.63(m,1H),2.69-2.61(m,1H),1.85-1.70(m,4H),1.33-1.22(m,4H).
实施例69:
Figure PCTCN2022094496-appb-000082
中间体69a的合成:
将5b(0.07g,0.19mmol)溶于无水乙醇(20mL)中,加入三乙胺(0.02g,0.19mmol),二硫化碳(0.02g,0.26mmol),室温反应30分钟,在反应液中加入二碳酸二叔丁酯(0.04g,0.19mmol),4-二甲氨基吡啶(2.3mg,0.019mmol),室温反应4小时,TLC监测原料无剩余。减压蒸除乙醇,残留物直接柱层析分离(二氯甲烷:甲醇=50:1),得到69a(0.03g,产率39%)。
终产物69的合成:
将69a(0.03g,0.07mmol)溶于无水乙醇(10mL)中,加入2.0M的氨乙醇溶液(0.004g,0.28mmol),加热至90℃,封管反应4小时,TLC监测原料无剩余。减压蒸除溶剂,残留物用无水乙醇(10mL)溶解,加入40%二氯乙醛水溶液(0.005g,0.07mmol),加热至90℃反应8小时,TLC监测原料无剩余。减压蒸除无水乙醇,残留物直接柱层析分离(二氯甲烷:甲醇=25:1),得到终产物69(0.02g,产率64%)。MS m/z(ESI):445.1[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.67(s,1H),8.33(s,1H),8.33-8.08(m,1H),7.50(d,J=7.8Hz,1H),7.35-7.33(m,1H),7.11(d,J=2.4Hz,1H),7.09(d,J=2.4Hz,1H),6.99-6.90(m,1H),6.58(d,J=4.2Hz,1H),3.79(s,3H),3.52-3.50(m,1H),2.63(m,1H),2.14-2.12(m,1H),2.02-2.00(m,1H),1.82-1.78(m,2H),1.36-1.30(m,3H),1.14-1.12(m,1H).
实施例70:
Figure PCTCN2022094496-appb-000083
中间体70a的合成:
将2a(160mg,0.68mmol)溶解于四氢呋喃(10mL)中,接着加入氯甲酸苯酯(191mg,1.22mmol),碳酸钾(187mg,1.35mmol),使整个体系搅拌过夜,TLC监测原料无剩余。反应直接经柱层析分离纯化(石油醚:乙酸乙酯=10:1-2:1),得到70a(200mg,产率83%)。
中间体70b的合成:
将(R)-1-叔丁氧羰基-3-氨基哌啶(1.00g,5.00mmol)溶解于二氯甲烷(30mL)中,接着加入三乙胺 (1.52g,15.0mmol),乙酸酐(701μL,7.50mmol)。体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠水溶液洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,得到70b(1.17g,产率97%)。
中间体70c的合成:
将70b(1.17g,4.83mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入10mL的三氟乙酸,整个体系在室温下搅拌3小时。减压除去溶剂,得到70c(1.50g)直接用于下一步。
终产物70的合成:
将70a(235mg,0.66mmol)溶解于四氢呋喃(10mL)中,接着加入70c(339mg,1.32mmol)和三乙胺(458μL,3.30mmol)。体系在室温下搅拌10小时,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,柱层析分离纯化(二氯甲烷:甲醇=40:1-20:1),得到终产物70(80mg,产率30%)。MS m/z(ESI):405.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ9.22(s,1H),8.25(s,1H),7.84(d,J=7.2Hz,1H),7.75(d,J=5.4Hz,1H),7.33(t,J=7.8Hz,1H),7.10(d,J=11.4Hz,1H),6.92(t,J=7.8Hz,1H),3.93(d,J=12.6Hz,1H),3.83(d,J=12.6Hz,1H),3.80(s,3H),3.64-3.59(m,1H),2.98-2.93(m,1H),2.79-2.76(m,1H),1.82-1.79(m,4H),1.71-1.69(m,1H),1.45-1.36(m,2H).
实施例71:
Figure PCTCN2022094496-appb-000084
中间体71a的合成:
将1a(0.40g,1.58mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.38g,1.58mmol),2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(0.72g,1.90mmol)和N,N-二异丙基乙胺(0.41g,3.16mmol),整个体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到71a(0.45g,产率60%)。
中间体71b的合成:
将71a(0.45g,0.94mmol)溶解于二氯甲烷(30mL)中,接着冰水浴下加入2mL的三氟乙酸,整个体系在室温下搅拌过夜,TLC检测原料无剩余.向反应液中加入水(100mL),再用饱和碳酸氢钠水溶液调节PH=9-10,二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-8:1)后,得到71b(0.31g,产率87%)。
终产物71的合成:
将71b(0.31g,0.82mmol)溶解于二氯甲烷(35mL)中,接着加入乙酸酐(0.25g,2.47mmol)和三乙胺(0.25g,2.47mmol),体系在室温下搅拌,TLC监测原料无剩余。向反应液中加入水(50mL),二氯甲烷萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得到终产物71(0.18g,产率52%)。MS m/z(ESI):420.1[M+H] +1H NMR(600MHz,CDCl 31H NMR(600MHz,DMSO-d 6)δ10.69(s,1H),8.41(s,1H),8.05(s,1H),7.78(d,J=7.8Hz,1H),7.25(d,J=7.2Hz,1H),7.09(d,J=2.4Hz,1H),6.91-6.88(m,1H),3.76(s,3H),3.57-3.54(m,1H),2.62-2.59(m,1H),1.86-1.84(m,1H),1.76-1.74(m,6H),1.31-1.23(m,3H),1.07-1.05(m,1H).
实施例72:
Figure PCTCN2022094496-appb-000085
中间体72a的合成:
将6-溴-1-甲基-1H-吲唑(211mg,1.00mmol),双联频哪醇二硼烷(508mg,2.00mmol),乙酸钾(294mg,3.00mmol)和[1,1'-双(二苯基膦基)二茂铁]二氯化钯(73mg,0.10mmol)溶解到1,4-二氧六环(20mL)中。将反应体系用氮气保护并在100℃反应4小时,TLC检测无原料剩余,停止加热。向反应液中加入水(100mL),乙酸乙酯萃取(100mL×3),合并有机相,饱和氯化钠洗涤(100mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚-石油醚:乙酸乙酯=4:1),得到72a(240mg,产率93%)。
中间体72b的合成:
将72a(258mg,1.00mmol)溶解于二乙二醇二甲醚(40mL)中,室温下加入5-氟-4碘吡啶-2-胺(286mg,1.20mmol),[1,1'-双(二苯基膦基)二茂铁]二氯化钯(73mg,0.10mmol),碳酸钾(414mg,3.00mmol)和水10mL,将反应体系在80℃下搅拌4小时,TLC检测原料无剩余,停止反应。向反应液中加入水(100mL),乙酸乙酯萃取(100mL×3),合并有机相,饱和氯化钠洗涤(100mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚-石油醚:乙酸乙酯=1:1),得到72b(175mg,产率72%)。
中间体72c的合成:
将72b(175mg,0.72mmol)和(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(350mg,1.44mmol)溶解于N,N-二甲基甲酰胺(20mL)中,依次加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(548mg,1.44mmol)和二异丙基乙胺(0.36mL,2.16mmol),将反应体系在室温反应4小时,TLC检测原料无剩余,向反应液中加入水(50mL),乙酸乙酯萃取(100mL×3),合并有机相,饱和氯化钠洗涤(100mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离纯化(石油醚:乙酸乙酯=4:1-2:1),得到72c(200mg,收率56%)。
终产物72的合成:
将72c(200mg,0.40mmol)溶解到二氯甲烷(50mL)中,接着加入三氟乙酸(5.00mL)室温下搅拌4小时,TLC检测原料无剩余。向体系内加入饱和碳酸钠水溶液,将反应体系的pH值调节至弱碱性。分液后,二氯甲烷萃取(100mL×3),合并有机相浓缩至体积约为50mL,加入三乙胺(2.00mL)和乙酸酐(2.00mL)室温下反应30分钟后,加入碳酸钠水溶液洗涤有机相。分液,再用二氯甲烷萃取(20mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,经柱层析分离(乙酸乙酯),得到终产物72(135mg,产率83%)。MS m/z(ESI):410.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.64(s,1H),8.44(s,1H),8.36(s,1H),8.14(s,1H),7.96(s,1H),7.90(s,1H),7.79(d,J=7.8Hz,1H),7.33(d,J=7.8Hz,1H),4.12(s,3H),3.59-3.58(m,1H),2.99-2.95(m,1H),1.91(s,3H),1.82-1.78(m,4H),1.14-1.12(m,4H).
实施例73:
Figure PCTCN2022094496-appb-000086
终产物73的合成:
将23c(101mg,0.25mmol)溶解于1,4二氧六环(10mL)和水(5mL)中,接着加入1-异丙基-4-(4,4,5,5-四甲基-1,3,2-二氧杂硼硼烷-2-基)-1H-吡咯并[2,3-b]吡啶(106mg,0.37mmol),[1,1'-双(二苯基膦)二茂铁]二氯化钯(15mg,0.02mmol)和碳酸钾(68mg,0.49mmol),用氮气置换三次,使整个体系处于氮气的氛围下。体系在100℃下回流搅拌,反应4小时,TLC监测原料无剩余。反应直接经柱层析分离纯化(二氯甲烷:甲醇=50:1-10:1),得终产物73(60mg,产率55%)。MS m/z(ESI):438.2[M+H] +
1H NMR(600MHz,DMSO-d6)δ10.70(s,1H),8.51(s,1H),8.41-8.38(m,2H),7.83(d,J=3.6Hz,1H),7.79(d,J=7.8Hz,1H),7.25-7.24(m,1H),6.50-6.49(m,1H),5.17-5.15(m,1H),3.58-3.56(m,1H),2.64-2.60(m,1H),1.90-1.88(m,1H),1.77-1.76(m,6H),1.51(s,3H),1.49(s,3H),1.40-1.25(m,3H),1.15-1.05(m,1H).
实施例74:
Figure PCTCN2022094496-appb-000087
中间体74a的合成:
将2,3-二氯吡啶-4-硼酸(1.05g,5.50mmol)溶解于二氧六环(30mL)和水(5mL)中,接着加入5-氟-4-碘-吡啶-2-胺(1.00g,4.20mmol),1,1’-双(二苯基膦)二茂铁]二氯化钯二氯甲烷络合物(0.34g,0.42mmol),碳酸钾(1.74g,12.6mmol),在氮气保护下加热至100℃反应8小时,TLC监测原料无剩余。停止加热,降至室温。将反应液直接进行柱层析分离(正己烷:乙酸乙酯=1:1),得到中间体74a(0.40g,产率37%)。
中间体74b的合成:
将74a(0.13g,0.50mmol)溶解于乙腈(10mL)中,接着加入(1S,3R)-3-[(叔丁氧羰基)氨基]环己烷甲酸(0.16g,0.65mmol),N,N,N',N'-四甲基氯甲脒六氟磷酸盐(0.17g,0.60mmol)和N-甲基咪唑(0.14g,1.75mmol),室温反应12小时,TLC监测原料无剩余。反应液直接柱层析分离纯化(正己烷:乙酸乙酯=2:1),得到74b(0.12g,产率50%)。
中间体74c的合成:
将74b(0.12g,0.25mmol)溶解于二氯甲烷(20ml)中,加入三氟乙酸4mL,室温反应4小时,TLC监测原料无剩余。将反应液用水洗涤(30mL×3),合并水相,水相用碳酸钠调节pH为8-9,二氯甲烷萃取(30mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥。减压浓缩除去溶剂,得到74c(0.08g,产率84%)。
终产物74的合成:
将74c(0.08g,0.21mmol)溶解于二氯甲烷(20mL)中,接着加入乙酸酐(0.04g,0.42mmol),三乙胺(0.04g,0.42mmol),室温反应2小时,TLC监测原料无剩余。反应液直接进行柱层析分离(二氯甲烷:甲醇=50:1),得到终产物74(0.02g,产率22%)。MS m/z(ESI):426.2[M+H] +.
1H NMR(600MHz,DMSO-d6)δ10.81(s,1H),8.55-8.54(m,2H),8.18(d,J=5.4Hz,1H),7.80(d,J=7.8Hz,1H),7.6(d,J=5.4Hz,1H),3.60-3.54(m,1H),2.64-2.60(m,1H),1.90-1.88(m,1H),1.77-1.76(m,6H),1.40-1.20(m,3H),1.19-1.05(m,1H).
实施例76:
Figure PCTCN2022094496-appb-000088
中间体76a的合成:
将5b(500mg,1.385mmol)溶解于N,N-二甲基甲酰胺(15mL)中,接着加入1-[(叔丁氧基)羰基]-3-氰基氮杂环丁烷-3-羧酸(375mg,1.66mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(787mg,2.07mmol)和N,N-二异丙基乙胺(687uL,4.15mmol),整个体系在室温下 搅拌,反应10小时,TLC监测原料无剩余。向反应液中加入水(50mL),搅拌30分钟过滤,无水硫酸钠干燥,得到76a(497mg,产率63%)。
终产物76的合成:
将76a(497mg,0.87mmol)溶解于二氯甲烷(20mL)中,接着加入三氟乙酸(5mL),整个体系在室温下搅拌2小时,TLC检测原料无剩余。向反应液中加入水(100mL),碳酸氢钠调PH至8,乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,进行柱层析分离(二氯甲烷:甲醇=40:1),得到终产物76(396mg,产率97%)。MS m/z(ESI):470.2[M+H] +1H NMR(600MHz,CD 3OD)δ8.38(s,1H),7.75(d,J=5.4Hz,1H),7.50-7.42(m,1H),7.04(d,J=10.8Hz,1H),6.96-6.86(m,1H),4.60-4.32(m,4H),3.93-3.76(m,4H),2.70(m,1H),2.23(m,1H),2.00(m,3H),1.71-1.46(m,3H),1.43-1.27(m,1H).
实施例77:
Figure PCTCN2022094496-appb-000089
终产物77的合成:
将76(296mg,0.63mmol)溶解于甲醇(10mL)中,接着加入1mL甲醛水溶液、三乙酰氧基硼氢化钠(249mg,1.27mmol),1滴乙酸。整个体系在室温下搅拌,反应10小时,TLC监测原料无剩余。向反应液中加入水(50mL),碳酸氢钠调PH至8,乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,得到终产物77(186mg,产率61%)。MS m/z(ESI):484.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.56(s,1H),8.32(s,1H),8.05(d,J=4.8Hz,1H),8.10(d,J=7.8Hz,1H),7.34-7.31(m,1H),7.09(s,1H),6.91-6.88(m,1H),3.79(s,3H),3.68-3.56(m,1H),2.98-2.73(m,4H),2.59-2.55(m,1H),2.42(s,3H),1.81-1.67(m,4H),1.30-1.09(m,4H).
实施例78:
Figure PCTCN2022094496-appb-000090
中间体78a的合成:
将5b(500mg,1.385mmol)溶解于N,N-二甲基甲酰胺(15mL)中,接着加入1-[(叔丁氧基)羰基]-3-氰基氮杂环丁烷-3-羧酸(375mg,1.66mmol)、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(787mg,2.07mmol)和N,N-二异丙基乙胺(687uL,4.15mmol),整个体系在室温下搅拌,反应10小时,TLC监测原料无剩余。向反应液中加入水(50mL),搅拌30分钟过滤,无水硫酸钠干燥,得到78a(504mg,产率61%)。
终产物78的合成:
将78a(504mg,0.84mmol)溶解于二氯甲烷(20mL)中,接着加入三氟乙酸(5mL),整个体系在室温下搅拌2小时,TLC检测原料无剩余。向反应液中加入水(100mL),碳酸氢钠调PH至8,乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,进行柱层析分离(二氯甲烷:甲醇=40:1),得到终产物78(384mg,产率92%)。MS m/z(ESI):498.7[M+H] +1H NMR(600MHz,CD3OD)δ8.32(s,1H),8.07(d,J=8.4Hz,1H),7.35-7.33(m,1H),7.11-7.10(m,1H),7.08-6.91(m,1H),3.78(s,3H),3.78-3.66(m,1H),3.30-3.28(m,2H),2.99-2.97(m,2H),2.62-2.52(m,1H),2.30-2.22(m,4H),1.79-1.77(m,4H),1.46-1.43(m,1H),1.29-1.26(m,3H).
实施例79:
Figure PCTCN2022094496-appb-000091
终产物79的合成:
将5b(0.16g,0.43mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入N,N-二乙基甘氨酸盐酸盐(109mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物40(116mg,产率57%)。MS m/z(ESI):475.3[M+H] +
1H NMR(600MHz,CDCl 3)δ8.23(d,J=7.8Hz,1H).8.13(s,1H),8.07(s,1H),7.40(m,1H),7.277.22(m,1H),6.87-6.60(m,2H),3.94-3.84(m,1H),3.81(s,3H),3.01(s,2H),2.54(m,4H),2.45(m,1H),2.22(m,1H),1.96(m,3H),1.56-1.36(m,3H),1.26-1.12(m,1H),1.01(m,6H).
实施例80:
Figure PCTCN2022094496-appb-000092
中间体80a的合成:
将5b(0.30g,0.83mmol)溶解于二氯甲烷(10mL)中,接着在0℃下加入三乙胺(420mg,4.15mmol)和氯乙酰氯(186mg,1.66mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(20mL),乙酸乙酯萃取(10mL×3),合并有机相,饱和氯化钠洗涤(10mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(正己烷:乙酸乙酯=1:0-0:1),得到80a(200mg,产率55%)。
终产物80的合成:
将80a(200mg,0.46mmol)溶解于N,N-二甲基甲酰胺(10mL)中,再加入硫代乙酸钾(78.25mg,0.69mmol),室温反应2小时,TLC监测原料无剩余。将反应液加入到1M HCl水溶液(10mL)中,再加入水(10mL)和二氯甲烷(15mL)。二氯甲烷萃取(10mL×3),合并有机相,饱和氯化钠洗涤(10mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(正己烷:乙酸乙酯=1:0-0:1),得到终产物80(200mg,产率91%)。MS m/z(ESI):478.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.58(s,1H),8.33(s,1H),8.07(d,J=5.4Hz,1H),7.59(d,J=8.4Hz,1H),7.35-7.34(m,1H),7.33-7.11(m,1H),7.09-6.90(m,1H),4.39(s,2H),3.78(s,3H),3.65-3.63(m,1H),2.66-2.60(m,1H),2.58(s,3H),1.85-1.69(m,4H),1.24-1.14(m,4H).
实施例81:
Figure PCTCN2022094496-appb-000093
终产物81的合成:
将80(220mg,0.46mmol)溶解于甲醇(5mL)中,再加入碳酸钾(317mg,2.30mmol)室温下反应20分钟,再升温至55℃反应40分钟。TLC监测原料无剩余。向反应液中加入水(20mL),乙酸乙酯萃取(10mL×3),合并有机相,饱和氯化钠洗涤(10mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(正己烷:乙酸乙酯=1:0-0:1),得到终产物81(41mg,产率20%)。MS m/z(ESI):436.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.55(s,1H),8.31(s,1H),8.07(d,J=5.4Hz,1H),7.56(d,J=9.0Hz,1H),7.35-7.32(m,1H),7.09-7.07(m,1H),6.91-6.88(m,1H),3.89-3.84(m,4H),3.66-3.63(m,1H),3.24-3.22(m,2H),2.61-2.57(m,1H),1.83-1.68(m,4H),1.34-1.26(m,4H).
实施例82:
Figure PCTCN2022094496-appb-000094
终产物82的合成:
将78(300mg,0.60mmol)溶解于甲醇(10mL)中,接着加入1mL甲醛水溶液、三乙酰氧基硼氢化钠(235mg,1.20mmol),1滴乙酸。整个体系在室温下搅拌,反应10小时,TLC监测原料无剩余。 向反应液中加入水(50mL),碳酸氢钠调PH至8,乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,减压除去溶剂,得到终产物82(205mg,产率67%)。MS m/z(ESI):512.2[M+H] +
1H NMR(600MHz,DMSO-d 6)δ10.28(s,1H),8.28(s,1H),8.22-8.09(m,1H),8.05(d,J=8.4Hz,1H),7.32-7.30(m,1H),7.09-7.02(m,1H),6.91-6.87(m,1H),3.80(s,3H),3.75-3.66(m,1H),3.57-3.52(m,1H),3.16-2.96(m,2H),2.81-2.59(m,4H),2.50-2.48(m,2H),2.43-2.31(m,3H),2.00-1.74(m,4H),1.52-1.48(m,1H),1.41-1.22(m,3H).
实施例83:
Figure PCTCN2022094496-appb-000095
终产物49的合成:
将5b(0.20g,0.55mmol)溶解于N,N-二甲基甲酰胺(30mL)中,接着加入2-羟基异丁酸(68mg,0.65mmol),2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(178mg,0.47mmol)和N,N-二异丙基乙胺(111mg,0.86mmol),体系在室温下搅拌过夜,TLC监测原料无剩余。向反应液中加入水(100mL),乙酸乙酯萃取(50mL×3),合并有机相,饱和氯化钠洗涤(50mL×2),无水硫酸钠干燥,然后减压除去溶剂,粗品经柱层析分离纯化(二氯甲烷:甲醇=50:1-20:1),得到终产物83(81mg,产率33%)。MS m/z(ESI):448.2[M+H] +
1H NMR(600MHz,CDCl 3)δ8.31(s,1H),8.24(d,J=8.4Hz,1H),8.12(s,1H),7.26-7.24(m,1H),6.76-6.73(m,2H),6.63(d,J=12.6Hz,1H),3.90-3.82(m,1H),3.81(s,3H),2.65(s,1H),2.56-2.42(m,1H),2.26-2.23(m,1H),2.00-1.89(m,3H),1.61-1.49(m,2H),1.45(d,J=4.8Hz,6H),1.30-1.18(m,1H).
实施例75、84-90:
根据实施例1-72的方法,选择对应的原料合成化合物75、84-90,其结构分别如下:
Figure PCTCN2022094496-appb-000096
试验例1:本申请的化合物对CDK9、CDK1、CDK2、CDK4、CDK5、CDK6和CDK7的抑制效果试验
1.实验目的
检测化合物在CDK1/2/4/5/6/7/9激酶上的抑制效果,并拟出有效的IC 50值。
2.CDKs检测家族
CDK1/CDK2/CDK4/CDK5/CDK6/CDK7/CDK9
表1:体外测试中激酶、底物和ATP的相关信息
Figure PCTCN2022094496-appb-000097
3.检测流程
3.1化合物稀释
将化合物用DMSO稀释11个浓度,3倍稀释,待测化合物最高浓度为10μM。
3.2酶反应
利用声波技术(Echo)将溶于DMSO中的化合物(50nL)转移到酶反应板中。取5μL CDK酶稀释液加入到酶反应板中,离心后室温孵育10分钟。取5μL底物预混液加入板中,每孔中底物和ATP终浓度见表1。离心后30℃反应120分钟。
3.3终止反应和信号检测
每孔加入10μL终止液,离心后在室温下孵育120分钟后,再在4℃下孵育过夜。在Envision仪器上使用HTRF程序读取信号值,并进行数据分析,IC 50(在50%最大效应时的抑制浓度)值以nM表示。结果见表2。
表2本申请化合物对CDK1、2、4、5、6、7和9的抑制效果(IC 50,nM)
Figure PCTCN2022094496-appb-000098
以上测试,说明本申请化合物对CDK9具有选择性的抑制作用。
试验例2:CDK9抑制剂对MV 4-11细胞增殖的体外抑制作用
1.试验目的
考察合成化合物对人白血病细胞增殖的体外抑制作用。
2.试验原理
MTT商品名噻唑蓝,是一种能接受氢原子的染料的四唑盐。活细胞的线粒体中的琥珀脱氢酶能使外源性的MTT还原为难溶的蓝紫色结晶物并沉积在细胞中,而死细胞无此功能。二甲基亚砜能溶解细胞中的蓝紫色复合物,用酶联免疫检测仪在490-550nm波长处测定其光吸收值,可间接反映细胞数量。在一定细胞数范围内,MTT结晶物形成的量与细胞数成正比。将待测药物依次稀释成不同浓度,加入96孔板,药物作用一定时间后,测定其OD值,OD值的大小能反映活细胞的数量,用SPSS19.0计算其IC 50值。
3.试验仪器
371型CO 2培养箱:Thermo
IX70-142型倒置荧光显微镜:Olympus
HFsafe-1500型生物安全柜:上海力申科学仪器有限公司
Varloskan flash酶标仪:Thermo公司
精密电子天平:梅特勒AL204型
4.试验材料:
4.1细胞及培养基
细胞名称 细胞来源 培养基 培养基厂家
MV 4-11 上海细胞库 10%IMDM Gibco
4.2试验材料
名称 规格 生产厂家
胎牛血清 500mL/瓶 Cellmo
PBS 2L/袋 Solarbio
DMSO 500mL/瓶 光复
MTT 5g/瓶 Amresco
4.3试剂配制
5mg/mL MTT工作液:称取MTT 0.5g溶于100mL PBS中,0.22μm微孔滤膜过滤除菌,置于4℃冰箱(两周内使用)或-20℃长期保存。
5.试验方法
5.1铺板
悬浮细胞:离心重悬后计数。用完全培养基配成一定密度的细胞悬液后,吹打均匀接种于96孔板,每孔100μL,之后于CO 2培养箱培养。
5.2药物配制
称取适量药物加入计算量DMSO溶解,分装,-20℃保存;配制浓度10mM
5.3加药
将化合物的10mM浓度储备液稀释成不同浓度(8个浓度)化合物的DMSO溶液(3倍稀释,20×终浓度),分别取各浓度化合物的DMSO溶液(10μL),使用细胞培养基(90μL)进行稀释,配成工作液(2×终浓度),取各浓度化合物的工作液(100μL)加于接种细胞的96孔板中(1×终浓度,最高终浓度为1000nM),于CO 2培养箱中继续培养。
6.检测
取出96孔板,显微镜观察细胞长满程度。采用MTT方法检测。
MTT法:每孔加入MTT 20μL,于培养箱培养约4h后,弃去孔内液体,每孔加入150μL DMSO,置于震荡仪震荡5-10min,用酶标仪在波长550nm处检测。
7.数据分析用SPSS19.0统计软件,计算药物的IC 50值。
化合物细胞增殖抑制结果见表3:
表3本申请化合物细胞增殖抑制试验数据
化合物编号 IC 50(μM) 化合物 IC 50(μM)
3 0.120 47 0.013
5 0.037 48 0.026
6 0.179 49 0.072
7 0.043 50 0.037
8 0.045 51 0.013
13 0.117 52 0.008
15 0.009 53 0.010
17 0.025 54 0.012
18 0.176 55 0.014
19 0.026 56 0.020
20 0.067 57 0.100
22 0.036 58 0.086
23 0.023 59 0.006
24 0.119 60 0.012
25 0.034 61 0.048
26 0.089 62 0.006
32 0.011 63 0.096
33 0.017 64 0.009
34 0.021 65 0.006
35 0.032 66 0.034
36 0.044 67 0.067
37 0.052 68 0.050
38 0.010 69 0.028
40 0.008 70 0.259
41 0.045 71 0.016
42 0.017 77 0.021
43 0.022 80 0.067
44 0.010 82 0.021
45 0.034 83 0.014
46 0.017    
实验结论:据表中数据,本申请化合物对MV 4-11细胞具有较强的抑制作用,体外细胞抑制活性基本都均在300nM以下,最优可达几nM。
试验例3:化合物对骨髓瘤和淋巴瘤细胞增殖的体外抑制作用
1.试验仪器
371型CO 2培养箱:Thermo
IX70-142型倒置荧光显微镜:Olympus
HFsafe-1500型生物安全柜:上海力申科学仪器有限公司
Varloskan flash酶标仪:Thermo公司
精密电子天平:梅特勒MS105型
TD6离心机:长沙湘锐离心机有限公司
2.试验材料:
2.1细胞及培养基
Figure PCTCN2022094496-appb-000099
Figure PCTCN2022094496-appb-000100
注:细胞培养环境均为37℃、5%CO 2
2.2试验材料
名称 规格 生产厂家
胎牛血清 500mL/瓶 兰州民海公司
PBS 500mL Gibco
DMSO 500mL/瓶 科密欧化学试剂有限公司
MTT 5g/瓶 Amresco
3.试验方法:
将处于对数生长期的细胞以一定数量接种于96孔板(100μL/孔),贴壁细胞贴壁24h后每孔加入100μL含不同浓度受试化合物的培养液,悬浮细胞接种当天即加入100μL含不同浓度受试化合物的培养液,每个药物浓度设3个复孔,并设相应的空白孔(只有培养基)及正常孔(药物浓度为0)。药物作用72小时后,加入MTT工作液(5mg/mL),每孔20μL;37℃作用4小时,去除上清液,加入DMSO(分析纯)150μL;微孔振荡器震荡混匀,将板擦拭干净,酶标仪550nm处检测光密度值(OD)。
采用下列公式计算细胞生长的抑制率:抑制率(%)=(OD值 正常孔-OD值 给药孔)/(OD值 正常孔-OD值 白孔)×100%
根据各浓度抑制率,用SPSS19.0计算药物半数抑制浓度IC 50
化合物细胞增殖抑制结果见表4:
表4本申请化合物细胞增殖抑制试验数据(IC 50,nM)
Figure PCTCN2022094496-appb-000101
RPMI-8826:人多发性骨髓瘤细胞;MM.1S:人多发性骨髓瘤细胞;SU-DHL-4:人弥漫大B淋巴瘤细胞;Jeko-1:人套细胞淋巴瘤细胞。
实验结论:据表中数据,本申请化合物对两种骨髓瘤细胞系和两种淋巴瘤细胞系均具有较强的抑制作用,体外细胞抑制活性均在100nM以下。
试验例4:药物体内抑瘤活性考察-本申请化合物在人急性髓细胞性白血病MV 4-11细胞皮下异种移植肿瘤模型中的体内药效学
细胞培养:含10%胎牛血清(FBS)的IMDM培养基,37℃,5%CO 2
NOD-SCID鼠,雌性,6-8周,体重约18-22克,每只小鼠在右后背皮下接种0.1mL(1×10 8个)MV 4-11细胞。当肿瘤体积平均值达到150立方毫米时,开始给药,给药剂量及方式见下表所示。肿瘤体积每周测量2次,体积以立方毫米计量,当溶剂组平均瘤体积生长至800立方毫米以上时,结束给药,以比较受试化合物组与溶剂组之间平均瘤体积的差异。化合物的抑瘤疗效用TGI(%)评价。TGI(%),反映肿瘤生长抑制率。
溶剂组及给药组的溶剂组成:DMSO:HP-β-CD(0.5g/mL):水比例为2%:20%:78%(v/v/v)
TGI(%)的计算:TGI(%)=[1-(某处理组给药结束时平均瘤体积-该处理组开始时平均瘤体积)/(溶剂对照组给药结束时平均瘤体积-溶剂对照组开始时平均瘤体积)]×100%。
结果见表5-表8。
表5体内抑瘤试验数据
Figure PCTCN2022094496-appb-000102
表6体内抑瘤试验数据
组别 动物只数 给药方式 给药剂量 给药天数 TGI(%)
溶剂组 5 qd,p.o. -- 9 --
化合物33 5 qd,p.o. 5mg/kg 9 56.9
化合物67 5 qd,p.o. 5mg/kg 9 67.8
表7体内抑瘤试验数据
组别 动物只数 给药方式 给药剂量 给药天数 TGI(%)
溶剂组 5 qd,p.o. -- 16 --
化合物45 5 qd,p.o. 5mg/kg 16 62.9
表8体内抑瘤试验数据
组别 动物只数 给药方式 给药剂量 给药天数 TGI(%)
溶剂组 5 qd,p.o. -- 7 --
化合物68 5 qd,p.o. 5mg/kg 7 78.1
试验结论:
本申请化合物在人急性髓细胞性白血病MV 4-11细胞皮下异种移植肿瘤模型中展示出良好的体内药效,具有显著的抑瘤作用。
试验例5:药物体内抑瘤活性考察-本申请化合物在人早幼粒急性白血病HL-60细胞皮下异种移植肿瘤模型中的体内药效。
细胞培养:含20%胎牛血清(FBS)的IMDM培养基,37℃,5%CO 2
NU/NU小鼠,雌性,6-8周,体重约18-22克,每只小鼠在右前肢腋部皮下接种HL-60细胞悬液0.1mL(约含细胞1×10 7个)。当肿瘤体积平均值达到150立方毫米时,开始给药,给药剂量及给药方式见下表所示。肿瘤体积每周测量2-3次,体积以立方毫米计量,当溶剂组平均瘤体积生长至800立方毫米以上时,结束给药,以比较受试化合物组与溶剂组之间平均瘤体积的差异。化合物的抑瘤疗效用TGI(%)评价。TGI(%),反映肿瘤生长抑制率。
溶剂组及给药组的溶剂组成:DMSO:HP-β-CD(0.5g/mL):水比例为2%:20%:78%(v/v/v)
TGI(%)的计算:TGI(%)=[1-(某处理组给药结束时平均瘤体积-该处理组开始时平均瘤体积)/(溶剂对照组给药结束时平均瘤体积-溶剂对照组开始时平均瘤体积)]×100%。
结果见表9。
表9体内抑瘤试验数据
组别 动物只数 给药方式 给药剂量 给药天数 TGI(%)
溶剂组 5 qd,p.o. -- 9 --
化合物45 5 qd,p.o. 5mg/kg 9 58.0
化合物67 5 qd,p.o. 5mg/kg 9 67.8
化合物68 5 qd,p.o. 5mg/kg 9 90.2
实验结论:
本申请化合物在人早幼粒急性白血病HL-60细胞皮下异种移植肿瘤模型中展示出良好的体内药效。开始给药9天后,本申请化合物具有显著的抑瘤作用。
试验例6:体外hERG抑制活性考察
1.试验目的:
快速激活的人延迟整流外向钾电流(IKr)主要由hERG离子通道介导,参与人类心肌细胞复极化。药物阻断这一电流是导致临床上出现QT间期延长综合征,甚至急性心律紊乱乃至猝死的主要原因。利用全细胞膜片钳技术,在稳定表达hERG通道的CHO-K1细胞株上检测化合物对hERG通道的阻断作用并且测定该化合物的半数抑制浓度IC 50。以其作为综合性心脏安全性评估的一部分,对其在心脏毒性的安全性体外筛选中进行初步的评价。
2.试验方法:
此试验包括以下几个方面:
●利用手动膜片钳技术在稳定表达hERG通道的CHO-K1细胞株上记录hERG电流;
●根据hERG尾电流计算每个浓度的抑制率;
●每个化合物测试5个浓度,推算IC 50值;
●每个浓度测试3个细胞;
●一个阳性对照药物。
采用全细胞膜片钳技术记录hERG电流。取细胞悬液加于细胞槽中,置于正置显微镜载物台上。待细胞贴壁后,用细胞外液灌流,流速为1–2mL/min。玻璃微电极由微电极拉制仪两步拉制,其入水电阻值为2-5MΩ。建立全细胞记录后,保持钳制电位为-80mV。给予电压刺激时去极化至+60mV,然后复极化至-50mV引出hERG尾电流。所有记录均在电流稳定后进行。胞外灌流给药从低浓度开始,每个浓度5-10min至电流稳定,再给下一个浓度。测试化合物的半数抑制浓度(IC 50)由Logistic方程最佳拟合得出。
Amitripyline是使用最为广泛的阻断hERG电流工具药物之一,故在本次研究中作为阳性对照药物。
3.结果见表10:
表10在CHO-K1稳定细胞株上所记录到的化合物对hERG电流的IC 50数值
Figure PCTCN2022094496-appb-000103
在上述试验中,阳性对照药物Amitriptyline对hERG电流抑制的IC 50结果与试验方的历史结果相一致,同时也与文献报道的结果相符合,表明本试验的结果可信。上述试验结果表明,所测化合物在最高测试浓度也无法达到对hERG电流的半数抑制,因此无法测出IC 50,说明在本试验的检测浓度范围内本申请化合物对hERG通道没有明显的抑制作用,可一定程度反映本申请化合物具有较低或无心脏毒性,对药物安全性评估具有积极意义。
试验例7:药物对小鼠的毒性考察
试验用动物:ICR小鼠(5周),雌雄各半
溶剂组及给药组的溶剂组成:DMSO:HP-β-CD(0.5g/mL):水比例为2%:20%:78%(v/v/v)
试验方法:将ICR小鼠按照体重均衡分组,共7组,每组雌雄各5只。给药方式为灌胃,每天一次,连续给药7天,给药剂量及结果见下表。
试验结果:
Figure PCTCN2022094496-appb-000104
试验结论:随着剂量递增,对照药物BAY1251152呈现出剂量依赖的毒性,动物死亡数量增多;化合物45在与对照药物同等剂量下,均未出现试验动物死亡事件,可见,化合物45在雌性和雄性动物中的耐受性明显优于对照药物BAY1251152。
尽管已出于清楚理解的目的通过说明及实例相当详细地描述前述发明,但根据本申请的教导,显而易见的是一般本领域技术人员可在不背离随附权利要求的精神或范围的情况下对其进行某些变化及修改。

Claims (17)

  1. 一种如式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药在制备用于治疗血液肿瘤的药物中的用途,
    Figure PCTCN2022094496-appb-100001
    其中,
    X选自Cl、F;优选F;
    R 1选自取代或未取代的芳基,或取代或未取代的杂芳基;R 1中的所述“取代”是指被1个、2个、3个、4个或5个各自独立地选自-F、-Cl、-Br、-NH 2、-OH、-SH、-CN、-NO 2、-N 3、-C≡CH、-COOH、-R 3、-(CH 2) wO(CH 2) nR 3、-(CH 2) wNH(CH 2) nR 3、-(CH 2) wNR 3(CH 2) nR 4、-(CH 2) wS(CH 2) nR 3、-(CH 2) wC(O)(CH 2) nR 3、-(CH 2) wC(O)O(CH 2) nR 3、-(CH 2) wOC(O)(CH 2) nR 3、-(CH 2) wC(O)NH(CH 2) nR 3、-(CH 2) wNHC(O)(CH 2) nR 3、-(CH 2) wC(O)NR 3(CH 2) nR 4、-(CH 2) wNR 3C(O)(CH 2) nR 4、-(CH 2) wOS(O) 2(CH 2) nR 3或-(CH 2) wS(O) 2O(CH 2) nR 3的基团所取代;其中,w、n每次出现时各自独立地选自0、1、2、3或4;
    R 3和R 4分别独立地选自取代或未取代的芳基,取代或未取代的杂芳基,取代或未取代的C 1-6烷基,取代或未取代的C 1-6卤代烷基,取代或未取代的C 2-6烯基,取代或未取代的C 2-6炔基,取代或未取代的C 1-6烷氧基,取代或未取代的C 1-6卤代烷氧基,取代或未取代的环烷基,取代或未取代的杂环烷基,或者当R 3、R 4共同连接至同一个氮原子时,R 3、R 4和共同连接的氮原子组成取代或未取代的杂环烷基;R 3和R 4中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-NH 2、-OH、-SH、-CN、-NO 2、-N 3、-C≡CH、-COOH、C 1-6烷基、C 1-6卤代烷基、C 1-6烷氧基、C 1-6卤代烷氧基等的基团所取代;
    A环选自取代或未取代的环烷基、取代或未取代的杂环烷基;A环中的所述“取代”是指被1个、2个、3个、4个或5个各自独立地选自-F、-Cl、-Br、OH、NH 2、SH、CN、NO 2、-N 3、-C≡CH、COOH、
    R 5、OR 5、-NHR 5、-NR 5R 6、-SR 5、-NHCOR 5、-CONHR 5、-NHS(O) 2R 5、-S(O) 2NHR 5、-NR 5S(O) 2R 6、-S(O) 2NR 5R 6的基团所取代,或A环结构中1个、2个或多个-CH 2-基团可任选地被-C(O)-基团替代;其中,R 5和R 6独立地为C 1-6烷基、C 1-6卤代烷基;
    R 2选自H、R 7、-(CH 2) xR 7、-(CH 2) xNH(CH 2) yR 7、-(CH 2) xO(CH 2) yR 7、-(CH 2) xNR 7(CH 2) yR 8、-(CH 2) xC(O)(CH 2) yH、-(CH 2) xC(O)(CH 2) yR 7、-(CH 2) xS(O) 2(CH 2) yR 7、-(CH 2) xC(O)C(O)(CH 2) yR 7、-(CH 2) xS(O) 2NH 2、-(CH 2) xNHS(O) 2H、-(CH 2) xS(O) 2NH(CH 2) yR 7、-(CH 2) xNHS(O) 2(CH 2) yR 7、-(CH 2) xS(O) 2NR 7(CH 2) yR 8、-(CH 2) xNR 7S(O) 2(CH 2) yR 8、-(CH 2) xC(O)O(CH 2) yR 7、-(CH 2) xOC(O)(CH 2) yR 7、-(CH 2) xC(O)NH 2、-(CH 2) xNHC(O)H、-(CH 2) xC(O)NH(CH 2) yR 7、-(CH 2) xNHC(O)(CH 2) yR 7、-(CH 2) xC(O)NR 7(CH 2) yR 8或-(CH 2) xNR 7C(O)(CH 2) yR 8;其中,1个、2个或多个-CH 2-基团可任选地被-C(O)-基团替代;x、y每次出现时各自独立地选自0、1、2、3或4;
    R 7和R 8独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、-R 10-NH-R 9、-R 10-C(O)-R 9、-R 10-NHC(O)-R 9、-R 10-C(O)NH-R 9、-R 10-S-R 9、-R 10-S(O)-R 9、-R 10-S-C(O)-R 9、环烷基、杂环烷基、芳基、杂芳基、-R 10-芳基、-R 10-杂芳基、-O-R 10-芳基、-O-R 10-杂芳基、-R 10-O-芳基、-R 10-O-杂芳基、-环烷基-芳基、-环烷基-杂芳基、-杂环烷基-芳基、-杂环烷基-杂芳基、C 2-6烯烃以及C 2-6炔烃,或者当R 7和R 8共同连接至同一个氮原子时,R 7和R 8与共同连接的氮原子组成取代或未取代的杂环烷基;其中R 9为C 1-6烷基,R 10为C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-NH 2、-SH、-CN、-NO 2、-N 3、-C≡CH、-COOH、C 1-6烷基、C 1-6烷氧基、C 1-6卤代烷基、C 1-6卤代烷氧基、-NHCN、-NHCONH 2、NHC(O)CH 3、N(CH 3) 2、N(C 2H 5) 2、-SC(O)CH 3、-OC(O)-C 1-6烷基等的基团所取代;
    上面未定义环原子数的所述的芳基优选包含6~10个碳原子,环烷基优选包含3~6个碳原子,杂芳基优选为5~10元杂芳基,且杂环烷基优选为3~8元杂环基;杂芳基或杂环烷基优选含有1个、2个或3个各自独立地选自N、O或S的杂原子,其余为碳原子;
    优选地,所述同位素衍生物为氘代形式的衍生物。
  2. 用于治疗血液肿瘤的药物组合物,其包含权利要求1中所定义的如式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,并任选地包含药学上可接受的载体。
  3. 用于治疗个体的血液肿瘤的方法,其包括给予所述个体权利要求1中所定义的如式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药,或者权利要求2所述的用于治疗血液肿瘤的药物组合物。
  4. 根据权利要求1所述的用途或权利要求2所述的药物组合物或权利要求3所述的方法,其中所述血液肿瘤为恶性血液肿瘤,优选为复发或难治性血液肿瘤;优选地,所述血液肿瘤选自白血病、淋巴瘤和骨髓瘤,优选为复发或难治性白血病、淋巴瘤和骨髓瘤;更优选地,所述白血病、淋巴瘤和骨髓瘤是CDK9相关的白血病、淋巴瘤和骨髓瘤。
  5. 根据权利要求4所述的用途或药物组合物或方法,其中所述白血病为急性白血病,优选为急性髓性白血病,更优选为复发或难治性急性髓性白血病,进一步优选为CDK9相关的复发或难治性急性髓性白血病;和/或所述淋巴瘤为非霍奇金淋巴瘤,优选为弥漫大B细胞淋巴瘤和套细胞淋巴瘤,更优选为CDK9相关的弥漫大B细胞淋巴瘤和套细胞淋巴瘤。
  6. 根据权利要求4所述的用途或药物组合物或方法,其中所述骨髓瘤选自多发性骨髓瘤;优选地,所述骨髓瘤选自CDK9相关的多发性骨髓瘤。
  7. 根据权利要求1-6任一项所述的用途或药物组合物或方法,其中A环选自取代或未取代的4-6元环烷基,取代或未取代的4-6元杂环烷基,取代或未取代的5-6元环烷基,或取代未取代的5-6元杂环烷基;优选为取代或未取代的5-6元环烷基,或取代未取代的5-6元杂环烷基;进一步优选为取代或未取代的5-6环烷基;或者,所述A环选自环己烷基、四氢吡咯基、哌啶基、哌嗪基、环戊烷基或吗啉基;优选为环己烷基、环戊烷基或四氢吡咯基;所述“取代”是指被1个、2个、3个、4个或5个各自独立地选自-F、-Cl、-Br、OH、NH 2、SH、CN、R 5、OR 5的基团所取代,其中,R 5为C 1-6烷基或C 1-6烷氧基。
  8. 根据权利要求1-7中任一项所述的用途或药物组合物或方法,其中所述式(I)所示化合物具有式(II)或式(III)所示结构:
    Figure PCTCN2022094496-appb-100002
    优选地,其具有式(IIa)或式(IIIa)所示结构:
    Figure PCTCN2022094496-appb-100003
  9. 根据权利要求1-8中任一项所述的用途或药物组合物或方法,其中R 1选自取代或未取代的6-10元芳基,或取代或未取代的5-10元杂芳基;所述杂芳基含有1或2个各自独立地选自N或O的杂原子;所述取代基的数量选自1、2或3个;进一步地,R 1选自取代或未取代的苯环、吡啶环、吲哚环、吲唑环、苯并呋喃环、吡咯并吡啶环;优选取代或未取代的苯环、吡啶环、吲哚环、苯并呋喃环、吡咯并吡啶环;进一步优选取代的苯环、吡啶环和未取代的吲哚环、苯并呋喃环、吡咯并吡啶环;更进一步优选取代的苯环;所述R 1上的取代基分别独立地选自-F、-Cl、-OH、-NH 2、-R 3、-(CH 2) wO(CH 2) nR 3或-(CH 2) wOC(O)(CH 2) nR 3;w和n各自独立的选自0、1或2;优选地,所述R 1上的取代基分别独立地选自F-、-Cl、-OH、-R 3、-(CH 2) wO(CH 2) nR 3;进一步优选地,所述R 1上的取代基分别独立地选自-F-、-OH、-R 3、-(CH 2) wO(CH 2) nR 3;当R 1为取代的苯环时,取代基选自-F、-OH或烷氧基,优选1或2个氟原子取代和1个-OH或烷氧基取代,优选1或2个氟原子取代和1个烷氧基取代。
  10. 根据权利要求1-9中任一项所述的用途或药物组合物或方法,其中R 3和R 4分别独立地选自取代或未取代的6元芳基,取代或未取代的5-6元杂芳基,取代或未取代的C 1-3烷基,取代或未取代的C 1-3烷氧基,取代或未取代的C 3-6环烷基,取代或未取代的C 3-6杂环烷基,或者当R 3、R 4共同连接至同一个氮原子时,R 3、R 4和共同连接的氮原子组成3-7元取代或未取代的杂环烷基;所述杂环烷基含有1个或2独立地选自N、O或S的杂原子;R 3和R 4中所述的“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-CH 3、-C 2H 5、-OCH 3、-OC 2H 5的取代基所取代;优选地,R 3和R 4分别独立地选自取代或未取代的C 1-3烷基、取代或未取代的C 1-3烷氧基;进一步地,R 3和R 4分别独立地选自取代或未取代的苯环、吡啶环、甲基、乙基、丙基、异丙基、甲氧基、乙氧基、丙氧基、异丙氧基、环丙基、环丁基、环戊基或环己基,R 3和R 4中所述的“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-CH 3、-C 2H 5、-OCH 3、-OC 2H 5的取代基所取代;优选地,R 3和R 4分别独立地选自取代或未取代的甲基、乙基、异丙基、甲氧基、 乙氧基、异丙氧基、环丙基、吡啶环;进一步优选地,R 3和R 4分别独立地选自取代或未取代的甲基、乙基、异丙基、甲氧基、乙氧基、异丙氧基;更进一步优选地,R 3和R 4分别独立地选自取代或未取代的甲基、甲氧基。
  11. 根据权利要求1-10中任一项所述的用途或药物组合物或方法,其中R 2选自R 7、-(CH 2) xR 7、-(CH 2) xNH(CH 2) yR 7、-(CH 2) xC(O)(CH 2) yR 7、-(CH 2) xS(O) 2(CH 2) yR 7、-(CH 2) xC(O)C(O)(CH 2) yR 7、-(CH 2) xC(O)O(CH 2) yR 7、-(CH 2) xC(O)NH(CH 2) yR 7、-(CH 2) xC(O)NR 7(CH 2) yR 8或-(CH 2) xNR 7C(O)(CH 2) yR 8;进一步优选地,R 2选自R 7、-(CH 2) xR 7、-(CH 2) xC(O)(CH 2) yR 7、-(CH 2) xS(O) 2(CH 2) yR 7、-(CH 2) xC(O)C(O)(CH 2) yR 7、-(CH 2) xC(O)O(CH 2) yR 7、-(CH 2) xC(O)NH(CH 2) yR 7;更进一步优选地,R 2选自R 7、-(CH 2) xR 7、-(CH 2) xC(O)(CH 2) yR 7;更进一步优选地,R 2选自-(CH 2) xC(O)(CH 2) yR 7
  12. 根据权利要求1-11中任一项所述的用途或药物组合物或方法,其中R 7和R 8各自独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、-R 10-NH-R 9、-R 10-C(O)-R 9、-R 10-NHC(O)-R 9、-R 10-C(O)NH-R 9、-R 10-S-R 9、-R 10-S-C(O)-R 9、C 3-6环烷基、3-6元杂环烷基、C 6-10芳基、5-10元杂芳基、-R 10-C 6-10芳基、-R 10-5-10元杂芳基、-O-R 10-C 6-10芳基、-O-R 10-5-10元杂芳基、-R 10-O-C 6-10芳基、-R 10-O-5-10元杂芳基、C 2-6烯烃以及C 2-6炔烃,或者当R 7和R 8共同连接至同一个氮原子时,R 7和R 8与共同连接的氮原子组成取代或未取代的3-6元杂环烷基;其中R 9为C 1-6烷基,R 10为C 1-6亚烷基、C 2-6亚烯基或C 2-6亚炔基;R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-NH 2、-SH、-CN、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基、C 1-3卤代烷氧基、-NHCN、-NHCONH 2、NHC(O)CH 3、N(CH 3) 2、N(C 2H 5) 2、-SC(O)CH 3、-OC(O)-C 1-6烷基等的基团所取代;优选地,R 7独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、-R 10-NHC(O)-R 9、C 3-6环烷基、3-6元杂环烷基、C 6-10芳基、5-10元杂芳基、C 2-6烯烃以及C 2-6炔烃,或者当R 7和R 8共同连接至同一个氮原子时,R 7和R 8与共同连接的氮原子组成取代或未取代的3-6元杂环烷基;进一步优选地,R 7独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、C 3-6环烷基、3-6元杂环烷基、C 6-10芳基、5-10元杂芳基;进一步优选地,R 7独立地选自取代或未取代的R 9、OR 9、-R 10-O-R 9、C 3-6环烷基、3-6元杂环烷基;进一步优选地,R 7独立地选自取代或未取代的R 9;R 7中所述的“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-NH 2、-SH、-CN、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基、C 1-3卤代烷氧基、-NHCN、-NHCONH 2、NHC(O)CH 3、N(CH 3) 2、N(C 2H 5) 2、-SC(O)CH 3、-OC(O)-C 1-6烷基等的基团所取代;优选地,R 7中所述的“取代”是指1个、2个或3个各自独立地选自-F、-OH、-NH 2、-SH、-CN、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基、NHC(O)CH 3、N(CH 3) 2、-OC(O)-C 1-6烷基等的基团所取代;进一步优选地,R 7中所述的“取代”是指1个、2个或3个各自独立地选自-F、-OH、-NH 2、-CN、C 1-3烷基、C 1-3烷氧基等的基团所取代;进一步优选地,R 7中所述的“取代”是指1个、2个或3个各自独立地选自-OH、-CN等的基团所取代;进一步优选地,R 7中所述的“取代”是指1个各自独立地选自-OH、-CN等的基团所取代;进一步优选地,R 7中所述的“取代”是指1个各自独立地选自-OH基团所取代。
  13. 根据权利要求1-12中任一项所述的用途或药物组合物或方法,其中R 7和R 8各自独立地选自取代或未取代的甲基、乙基、丙基、异丙基、丁基、戊基、甲氧基、乙氧基、丙氧基、异丙氧基、-CH 2OCH 3、-CH 2OCH 2CH 3、-CH 2CH 2OCH 3、-CH 2CH 2OCH 2CH 3、环丙基、环丁基、环戊基、环己基、氮杂环丙基、氮杂环丁基、氮杂环戊基、氮杂环己基、氧杂环丙基、氧杂环丁基、氧杂环戊基、氧杂环己基、苯基、吡啶基、吡唑基、噁唑基、异噁唑基、噻吩基、噻唑基、苯甲基、苯乙基、乙烯基、丙烯基、乙炔基或丙炔基;优选地,R 7独立地选自取代或未取代的甲基、乙基、甲氧基、乙氧基、环丙基、环丁基、氮杂环丁基、氮杂环己基、氧杂环丁基、氧杂环己基、苯基、吡啶基、吡唑基、异噁唑基、噻吩基、噻唑基、苯甲基、乙烯基、丙烯基或乙炔基;进一步优选地,R 7独立地选自取代或未取代的甲基、乙基、甲氧基、环丙基、环丁基、氮杂环己基、氧杂环丁基、氧杂环己基、吡啶基、吡唑基、异噁唑基、乙烯基、丙烯基或乙炔基;更进一步优选地,R 7独立地选自取代或未取代的甲基、乙基、环丙基、氮杂环己基、氧杂环丁基、吡唑基、乙烯基、丙烯基或乙炔基;更进一步优选地,R 7独立地选自取代或未取代的甲基、乙基、环丙基;更进一步优选地,R 7独立地选自取代或未取代的甲基;R 7和R 8中的所述“取代”是指被1个、2个或3个各自独立地选自-F、-Cl、-Br、-OH、-NH 2、-SH、-CN、C 1-3烷基、C 1-3烷氧基、C 1-3卤代烷基、C 1-3卤代烷氧基、-NHCN、-NHCONH 2、NHC(O)CH 3、N(CH 3) 2、N(C 2H 5) 2、-SC(O)CH 3、-OC(O)-C 1-6烷基等的基团所取代。
  14. 根据权利要求1-13中任一项所述的用途或药物组合物或方法,其中所述式(I)所示化合物具有如下结构:
    Figure PCTCN2022094496-appb-100004
    Figure PCTCN2022094496-appb-100005
    Figure PCTCN2022094496-appb-100006
    Figure PCTCN2022094496-appb-100007
  15. 根据权利要求1-14任一项所述的用途或药物组合物或方法,其中所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药是所述药物或药物组合物中的唯一的活性成分,或者所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药或所述药物组合物被作为唯一治疗剂给予所述个体。
  16. 根据权利要求1-14任一项所述的用途或药物组合物或方法,其中所述式(I)所示化合物或其可药用盐、其立体异构体、同位素衍生物或前药与一种或多种其它靶向药或化疗药联合使用,或者所述药物或所述药物组合物还包含一种或多种其它靶向药或化疗药作为活性成分。
  17. 根据权利要求1-16任一项所述的用途或药物组合物或方法,其中所述药物或药物组合物被配制成临床可接受的制剂,所述制剂优选为口服制剂或注射制剂。
PCT/CN2022/094496 2021-05-24 2022-05-23 周期蛋白依赖性激酶9抑制剂的用途 WO2022247796A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110582506 2021-05-24
CN202110582506.8 2021-05-24

Publications (1)

Publication Number Publication Date
WO2022247796A1 true WO2022247796A1 (zh) 2022-12-01

Family

ID=84115363

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/094496 WO2022247796A1 (zh) 2021-05-24 2022-05-23 周期蛋白依赖性激酶9抑制剂的用途

Country Status (2)

Country Link
CN (1) CN115381824A (zh)
WO (1) WO2022247796A1 (zh)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102471310A (zh) * 2009-09-04 2012-05-23 诺瓦提斯公司 用于治疗增生性疾病的联吡啶化合物
CN102482265A (zh) * 2009-09-04 2012-05-30 诺瓦提斯公司 用于治疗增殖性疾病的吡嗪基吡啶化合物
CN102498107A (zh) * 2009-09-04 2012-06-13 诺瓦提斯公司 作为激酶抑制剂的杂芳基化合物
CN105189481A (zh) * 2013-03-13 2015-12-23 艾伯维公司 吡啶cdk9激酶抑制剂
CN107873028A (zh) * 2015-06-29 2018-04-03 阿斯利康(瑞典)有限公司 用作cdk9抑制剂的多环酰胺衍生物
WO2021050824A1 (en) * 2019-09-11 2021-03-18 Prelude Therapeutics Incorporated Cdk inhibitors and their use as pharmaceuticals
WO2021115335A1 (zh) * 2019-12-09 2021-06-17 石药集团中奇制药技术(石家庄)有限公司 作为周期蛋白依赖性激酶9抑制剂的化合物及其应用

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102471310A (zh) * 2009-09-04 2012-05-23 诺瓦提斯公司 用于治疗增生性疾病的联吡啶化合物
CN102482265A (zh) * 2009-09-04 2012-05-30 诺瓦提斯公司 用于治疗增殖性疾病的吡嗪基吡啶化合物
CN102498107A (zh) * 2009-09-04 2012-06-13 诺瓦提斯公司 作为激酶抑制剂的杂芳基化合物
CN105189481A (zh) * 2013-03-13 2015-12-23 艾伯维公司 吡啶cdk9激酶抑制剂
CN107873028A (zh) * 2015-06-29 2018-04-03 阿斯利康(瑞典)有限公司 用作cdk9抑制剂的多环酰胺衍生物
WO2021050824A1 (en) * 2019-09-11 2021-03-18 Prelude Therapeutics Incorporated Cdk inhibitors and their use as pharmaceuticals
WO2021115335A1 (zh) * 2019-12-09 2021-06-17 石药集团中奇制药技术(石家庄)有限公司 作为周期蛋白依赖性激酶9抑制剂的化合物及其应用

Also Published As

Publication number Publication date
CN115381824A (zh) 2022-11-25

Similar Documents

Publication Publication Date Title
EP3555070B1 (en) Amine-substituted heterocyclic compounds as ehmt2 inhibitors and methods of use thereof
JP6932394B2 (ja) 含窒素複素環化合物、製造方法、中間体、医薬組成物及び応用
CN111484477B (zh) 一种苯并吡啶酮杂环化合物及其用途
JP6877407B2 (ja) Ntrk関連障害の治療に有用な化合物および組成物
WO2021115335A1 (zh) 作为周期蛋白依赖性激酶9抑制剂的化合物及其应用
CA2988816A1 (en) Ezh2 inhibitors for treating lymphoma
TW201823249A (zh) Menin-mll相互作用之稠合二環抑制劑
JP2018502159A (ja) Btk阻害剤
WO2022166890A1 (zh) 取代的哒嗪苯酚类衍生物
WO2022253101A1 (zh) 作为parp7抑制剂的哒嗪酮类化合物
WO2018001332A1 (zh) 具有突变型异柠檬酸脱氢酶抑制活性的化合物、其制备方法及用途
CN111718332B (zh) 2-取代吡唑氨基-4-取代氨基-5-嘧啶甲酰胺类化合物、组合物及其应用
WO2021129841A1 (zh) 用作ret激酶抑制剂的化合物及其应用
US11760751B2 (en) Benzo 2-azaspiro[4.4]nonane compound and use thereof
TWI782523B (zh) 用作ret激酶抑制劑的化合物及其應用
WO2022247796A1 (zh) 周期蛋白依赖性激酶9抑制剂的用途
US20230303534A1 (en) Preparation method for novel rho-related protein kinase inhibitor and intermediate in preparation method
WO2022247785A1 (zh) 一种周期蛋白依赖性激酶9抑制剂的用途
WO2022222911A1 (zh) 嘧啶酮化合物及其用途
WO2017008681A1 (zh) 酰胺类衍生物、其制备方法及其在医药上的用途
JP2021531331A (ja) チアジアゾール誘導体及びgls1阻害剤としてのその使用
JP2021517570A (ja) イミダゾピロロン化合物及びその使用
CN114276328B (zh) 作为小分子免疫抑制剂的化合物、其制备方法及其应用
JP2024519543A (ja) Parp7阻害剤としてのピリダジノン化合物
CN115677831A (zh) 拟肽类stat降解药物、组合物及应用

Legal Events

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

Ref document number: 22810521

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE