WO2023246371A1 - Small molecule compound having pyrimidothiophene structure and use thereof - Google Patents

Small molecule compound having pyrimidothiophene structure and use thereof Download PDF

Info

Publication number
WO2023246371A1
WO2023246371A1 PCT/CN2023/094038 CN2023094038W WO2023246371A1 WO 2023246371 A1 WO2023246371 A1 WO 2023246371A1 CN 2023094038 W CN2023094038 W CN 2023094038W WO 2023246371 A1 WO2023246371 A1 WO 2023246371A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
alkyl
add
reaction
synthesis
Prior art date
Application number
PCT/CN2023/094038
Other languages
French (fr)
Chinese (zh)
Inventor
樊梦阳
马大为
丁克
张红进
贾素云
Original Assignee
中国科学院基础医学与肿瘤研究所(筹)
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 中国科学院基础医学与肿瘤研究所(筹) filed Critical 中国科学院基础医学与肿瘤研究所(筹)
Publication of WO2023246371A1 publication Critical patent/WO2023246371A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings

Definitions

  • the present invention relates to the field of chemical drugs, and in particular to a class of small molecule compounds with a pyrimidothiophene structure and their applications.
  • the cyclin-dependent kinase family is a serine/threonine protein kinase that regulates transcription (CDK 7-13 and 19-20) or cell cycle progression (CDK 1-6 and 14-18 ).
  • CDK 7-13 and 19-20 serine/threonine protein kinase that regulates transcription
  • CDK 1-6 and 14-18 cell cycle progression
  • cell cycle dysregulation is a hallmark of cancer, and targeting cells
  • Drugs CDK4/6 inhibitors of cycle regulators have been launched and have achieved great success. Therefore, drugs targeting CDK family kinases are important tumor treatments.
  • CDK can combine with cyclin to form a heterodimer, in which CDK is the catalytic subunit and cyclin is the regulatory subunit.
  • Different cyclin-CDK complexes catalyze the phosphorylation of different substrates through CDK activity, thereby achieving different effects on the cell cycle.
  • the activity of CDK depends on the sequential expression of its positive regulatory subunit cyclin and the concentration of its negative regulatory subunit CKI (cyclin-dependent kinase inhibitor, CDK inhibitor).
  • CDK activity is also regulated by phosphorylation and dephosphorylation, as well as oncogenes and tumor suppressor genes.
  • CDK4/6 combines with cyclinD to promote the phosphorylation of retinoblastoma protein (Rb), and the activity of part of the transcription elongation factor E2F is released, which is used to synthesize a large amount of RNA and proteins to prepare for chromosome replication.
  • CDK2 combines with cyclinE to form the proteasome complex CDK2-cyclinE and is activated, which promotes further phosphorylation of Rb and induces the continued expression of the transcription factor E2F, thereby regulating cells to successfully pass through the G1 phase.
  • CDK2 After entering the S phase, CDK2 combines with cyclinA to form a complex CDK2-cyclinA, which participates in the S phase of the cell cycle and completes DNA replication. Subsequently, cyclinA/B and CDK1 form the CDK1-cyclinA/B complex, driving the cell cycle into the G2/M phase, synthesizing RNA and proteins to prepare for the formation of spindle fibers. Finally, the cells go through all the cell cycles of G1-S-G2-M in an orderly manner to complete cell mitosis. Almost all CDK activations occur through: (1) Binding to Cyclin; (2) T-Loop is phosphorylated by CDK Activating Kinase (CAK). CAK is a ternary complex consisting of CDK7, Cyclin H and RING-finger protein MAT1.
  • CDK7 The unique feature of CDK7 is that it is involved in the regulation of both transcription and cell cycle. It is ubiquitously expressed in various types of cancer, and its downregulation leads to reduced cell proliferation. CDK7 is considered a feasible cancer therapeutic target. High CDK7 expression has been detected in multiple cancer types and is associated with aggressive clinicopathological features and poor prognosis. CDK7 is amplified in hepatocellular carcinoma, gastric cancer, and colorectal cancer (CRC). Immunohistochemical analysis of 173 gastric cancer specimens showed that elevated CDK7 levels were related to tumor grade.
  • CDK7 protein is expressed in most oral squamous cells High expression in cell carcinoma specimens is associated with increased T stage and decreased disease-free survival rate, indicating that it can be used as a prognostic biomarker. CDK7 protein and mRNA levels are upregulated in cancerous breast tissue compared with adjacent normal breast tissue.
  • CDK2 is directly involved in cell cycle regulation and plays a key role.
  • the activity of CDK2 depends on the combination with cyclin E or cyclin A, regulating the transition of cells from G1 to S phase and the progression of S phase.
  • pan-CDK inhibitors have been discovered in the past decade.
  • first-generation CDK inhibitors exhibited strong side effects, which greatly hindered clinical development. Therefore, the development of drugs that can selectively inhibit the activity of CDK7 or CDK2 and exert pharmacological effects in the body is of great significance for the treatment of tumors.
  • the object of the present invention is to provide a small molecule compound with a pyrimidothiophene structure and its application.
  • a selective CDK7 or CDK2 inhibitor it can inhibit the kinase function of CDK in in vitro and in vivo experiments, and block The transcription of oncogenes and uncontrolled cell cycle progression achieve the effect of inhibiting tumor cell proliferation.
  • the first aspect of the present invention provides a compound represented by Formula I or Formula I' or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof,
  • X is S or P
  • X’ and X are each independently CH or N;
  • Y and Y’ are each independently C or N;
  • Z and Z' are each independently CR z or N; wherein, R z is selected from: H, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2. Nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 4-6 One-membered heterocyclyl;
  • R 1 is selected from the following substituted or unsubstituted groups: C3-C10 cycloalkyl, -C1-C6 alkylene, C3-C10 cycloalkyl, 4-10 membered N-containing heterocyclyl, -C1-C6 alkylene Alkyl 4-10 membered N-containing heterocyclic group;
  • R 2 is selected from: H, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-6 membered heterocyclyl;
  • R 4 and R' 4 are each independently selected from: None, hydrogen, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, Cyano, C1-C4 alkyl, C1-C4 alkoxy, C3-C6 cycloalkyl, -COOC1-C6 alkyl, -OCOC1-C6 alkyl, -CONHC1-C6 alkyl, -CON(C1-C6 Alkyl) 2 , -NHC(O)C1-C6 alkyl, -NHC(O)OC1-C6 alkyl, -NHS(O) 2 C1-C6 alkyl, -S(O) 2 C1-C6 alkyl , -S(O)N(C1-C6 alkyl) 2 , -S(O) 2 N(C1-C6 alkyl
  • Rp 3 and R 5 are each independently selected from: H, halogen, OH, NH 2 , cyano, C1-C4 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkyl, halogenated C1-C4 alkyl Oxygen;
  • R p-1 and R' p-1 are each independently selected from the following substituted or unsubstituted groups: C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 ring Alkyl or 3-6 membered heterocyclyl; wherein, the substitution means substitution by one or more Ra;
  • L 1 is a substituted or unsubstituted C1-C6 alkyl group, wherein the substitution means substitution by one or more Ra;
  • Rn 1 and Rn 2 are each independently H, C1-C6 alkyl, halogenated C1-C6 alkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl; wherein, the substitution means by one or multiple Ra substitutions;
  • Ra is each independently selected from: halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , Nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl , 3-6 membered heterocyclic group.
  • YR 4 is N.
  • X is P
  • Rp 1 and Rp 2 are each independently methyl, ethyl, or propyl.
  • R p-1 and R' p-1 are each independently methyl, ethyl, or propyl.
  • R' 1 is selected from the following substituted or unsubstituted groups: C1-C6 alkyl NRn 1 Rn 2 , C3-C10 cycloalkyl, -C1-C6 alkylene, C3-C10 cycloalkyl, 4- 10-membered N-containing heterocyclic group, -C1-C6 alkylene group, 4-10-membered N-containing heterocyclic group;
  • Rn 1 and Rn 2 are each independently H, C1-C6 alkyl, halogenated C1-C6 alkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl; wherein, the substitution means substitution by one or more Ra;
  • R' 2 is selected from: H, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, cyano, C1-C6 alkyl , C1-C6 alkoxy, C1-C6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-6 membered heterocyclyl;
  • W is selected from: phenyl, naphthyl, 5-6 membered monocyclic heteroaryl or 8-10 membered fused bicyclic heteroaryl, benzo 5-10 membered heterocyclyl, 5-6 membered monocyclic heteroaryl And 5-10 membered heterocyclyl;
  • R' 5 and R' 6 are each independently selected from: hydrogen, halogen, hydroxyl, nitro, cyano, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkoxy, -N(R 5-1a R 5-1b ), phenyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, - NHC(O)R 5-2 , -NHC(O)OR 5-3 , -NHS(O) 2 R 5-4 , -C(O)N(R 5-5a R 5-5b ), -S( O) 2 R 5-6 , -S(O)(NH)R 5-7 , -S(O) 2 N(R 5-8a R 5-8b ) or -P(O)R 5-9a R 5 -9b ; or when R' 5 and R' 6 are connected to adjacent or the same ring atom, R 5' and R 6' together with the ring
  • R 5-1a , R 5-1b , R 5-2 , R 5-3 , R 5-4 , R 5-5a , R 5-5b , R 5-6 , R 5-7 , R 5-8a , R 5-8b , R 5-9a and R 5-9b are each independently selected from: hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl; wherein, the C1-C6 alkyl, C3-C6 cycloalkyl The group is optionally substituted by one or more groups selected from the group consisting of: halogen, hydroxyl; or R 5-1a and R 5-1b , R 5-5a and R 5-5b , R 5-8a and R 5 -8b , R 5-9a and R 5-9b can together form a substituted or unsubstituted C3-C6 carbocyclic ring or a substituted or unsubstituted 4-6 membered heterocyclyl group with the atoms to which they are connected, where
  • Ra is each independently selected from: halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, Cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- 6-membered heterocyclic group.
  • Rp 3 , Rp 4 , Rz and R' 4 are defined as above.
  • the W ring is selected from the following substituted or unsubstituted groups: phenyl, naphthyl, pyridyl, pyrimidine base, indolyl, isoindolyl, indazolyl, quinolyl, Wherein, the substitution refers to substitution by one or more Ra, and Ra is defined as above.
  • R' 5 and R' 6 are each independently selected from: hydrogen, halogen, hydroxyl, nitro, cyano, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkoxy, -NH 2 , -NHCH 3 , -NHCH(CH 3 ) 2 , -C(O)NH2, -C(O)NHCH 3 , -C(O)CH(CH 3 ) 2 , -P(O)(CH 3 ) 2 , -P(O)(CH 2 CH 3 ) 2 , -S(O) 2 NH 2 , -S(O ) 2 NHCH 3 , -NHC(O)O CH 3 , -S(O)(NH)CH 3 , -NHC(O)O CH 3 , -S(O)(NH)CH 3 , -NHC(O)CH 3 , -C(O)NH CH 2 CH 2 OH.
  • R 1 is selected from the following substituted or unsubstituted groups: C3-C10 cycloalkyl, -C1-C6 alkylene, C3-C10 cycloalkyl, 4-10 membered N-containing heterocycle group, -C1-C6 alkylene group, 4-10 membered N-containing heterocyclic group; wherein, the substitution means substitution by one or more Ra, and Ra is defined as above.
  • R 1 is selected from: substituted or unsubstituted 5-6 membered N-containing monocyclic heterocyclic groups (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.) , substituted or unsubstituted 7-10 membered bicyclic or tricyclic (such as spirocyclic, fused ring, bridged ring) heterocyclic group, substituted or unsubstituted -(CH 2 ) 1-3 -(5-6 membered N-containing Monocyclic heterocyclyl (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.), substituted or unsubstituted -(CH 2 ) 1-3 -(7-10 membered bicyclic or tricyclic Ring (such as spiro ring, fused ring, bridged ring) heterocyclyl), wherein the
  • R 1 is selected from: Among them, Rq is selected from: H, F, Cl.
  • R 1 is selected from:
  • R' 1 is selected from: C1-C6 alkyl NH 2 , C1-C6 alkyl NHC1-C6 alkyl, substituted or unsubstituted 5-6 membered N-containing monocyclic heterocyclic group (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.), substituted or unsubstituted 7-10 membered bicyclic or tricyclic (such as spirocyclic, fused ring, bridged ring) heterocyclyl, substituted or untaken -(CH 2 ) 1-3 -(5-6 membered N-containing monocyclic heterocyclic group (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.), substituted or Not taken -(CH 2 ) 1-3 -(7-10 membered bicyclic or tricyclic (such as spir
  • R'1 is selected from:
  • Y, Y', Z, Z', R 2 , R 4 , R' 4 , Rp 3 , R 5 , Rp 4 , L 1 , Rn 1 and Rn 2 are the specific ones in the embodiment.
  • the compound is selected from any one of the following compounds:
  • the compound is the compound shown in the embodiment.
  • a second aspect of the present invention provides a pharmaceutical composition, wherein the pharmaceutical composition includes the compound as described in the first aspect, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof; and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition further includes a drug selected from the following group: PD-1 inhibitors (such as nivolumab, pembrolizumab, pidilizumab, cemiplimab or biosimilars of the above drugs, etc.) , PD-L1 inhibitors (such as durvalumab, atezolizumab, avelumab or biosimilars of the above drugs, etc.), CD20 antibodies (such as rituximab, obinumab Tocilizumab, ofatumumab, veltuzumab, tositumomab, 131I-tositumomab, itumomab, 90Y- itumomab, 90In- itumomab, ibritumomab tiuxetan, etc.), CD47 antibodies (such as Hu5F9-G4, CC-90002, TTI-621, TTI-622, OSE-172,
  • a method for preparing a pharmaceutical composition including the steps of: combining a pharmaceutically acceptable carrier with the compound described in the first aspect of the present invention or its stereoisomer or optical isomer. , pharmaceutically acceptable salts, prodrugs or solvates are mixed to form a pharmaceutical composition.
  • the compound of the present invention can be prepared into powders, tablets, granules, capsules, solutions, emulsions, suspensions, etc.
  • the third aspect of the present invention provides a compound as described in the first aspect, or a pharmaceutically acceptable salt thereof, immediately isomer, solvate or prodrug, or the use of the pharmaceutical composition as described in the second aspect in the preparation of anti-tumor drugs.
  • the tumor is selected from colorectal cancer, breast cancer, lung cancer, ovarian cancer or gastric cancer, preferably breast cancer.
  • Figure 1 shows the anti-proliferation curve of the compound of the present invention on the triple-negative breast cancer cell line MDA-MB-453.
  • Figure 2 shows the anti-proliferation curve of the compounds of the present invention on the ovarian cancer cell line OVCAR3.
  • Figure 3 shows the anti-tumor efficacy test of the compounds of the present invention on the nude mouse transplanted tumor model of the triple-negative breast cancer cell line MDA-MB-231.
  • substituents When a substituent is described by a conventional chemical formula written from left to right, the substituent also includes substituents that are chemically equivalent when the structural formula is written from right to left. For example, -CH 2 O- is equivalent to -OCH 2 -.
  • the term "about” when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value.
  • the expression “about 100” includes all values between 99 and 101 and between (eg, 99.1, 99.2, 99.3, 99.4, etc.).
  • the term “contains” or “includes” can be open, semi-closed and closed. In other words, the term also includes “consisting essentially of,” or “consisting of.”
  • alkyl includes straight or branched chain alkyl groups.
  • C1-C8 alkyl represents a straight-chain or branched alkyl group with 1-8 carbon atoms, preferably C1-C6 alkyl, more preferably C1-C3 alkyl.
  • alkyl include but are not limited to methyl, Ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, etc.
  • alkenyl includes straight or branched chain alkenyl groups.
  • C2-C8 alkenyl refers to a linear or branched alkenyl group with 2-8 carbon atoms, preferably C2-C6 alkenyl, and more preferably C2-C4 alkenyl.
  • alkenyl include but are not limited to vinyl, Allyl, 1-propenyl, isopropenyl, 1-butenyl, 2-butenyl, 1-pentenyl, 1-hexenyl or similar groups.
  • alkynyl includes straight or branched chain alkynyl groups.
  • C2-C8 alkynyl refers to a straight-chain or branched alkynyl group with 2-8 carbon atoms, preferably C2-C6 alkynyl, more preferably C2-C4 alkynyl
  • examples of alkynyl include but are not limited to ethynyl , propynyl, butynyl, pentynyl, hexynyl or similar groups.
  • cycloalkyl refers to a cyclic alkyl group containing a specific number of C atoms, such as "C3-C12 cycloalkyl” refers to a cyclic alkyl group having 3 to 12 (preferably 3, 4, 5, 6, 7 or 8) cycloalkyl carbon atoms. It may be a monocyclic ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or similar groups. Bicyclic forms, such as bridged or spirocyclic forms, are also possible. In the present invention, cycloalkyl is intended to include substituted cycloalkyl.
  • carbocyclyl refers to a saturated or partially saturated cyclic group containing a specified number of C atoms, such as "C3-C6 carbocyclyl” refers to a cyclic group having 3 to 6 (i.e., 3, 4, 5 , 6) Carbocyclic group of carbon atoms.
  • Bicyclic forms, such as bridged or spirocyclic forms, are also possible.
  • carbocyclyl is intended to include substituted carbocyclyl.
  • C1-C6 alkoxy refers to a straight or branched chain alkoxy group having 1 to 6 carbon atoms; which has the formula C1-C6 alkyl-O- or -C1-C5 alkyl -O-C1-C5 alkyl (e.g., -CH 2 -O-CH 2 CH 3 , -CH 2 -O-(CH 2 ) 2 CH 3 , -CH 2 CH 2 -O-CH 2 CH 3 ) Structure, for example, methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tert-butoxy, etc.
  • heterocyclyl refers to a saturated or partially saturated cyclic group having 1-3 heteroatoms selected from N, S and O
  • heterocyclyl refers to a cyclic group having 3 to 12-membered heterocyclyl.
  • the 3-12-membered heterocyclyl group is preferably a 3-8-membered heterocyclyl group, and more preferably is a 4-6-membered or 6-8-membered heterocyclyl group.
  • Specific examples may be oxetane, azetidine, tetrahydro-2H-pyranyl, piperidinyl, piperazinyl, tetrahydrofuranyl, morpholinyl, pyrrolidinyl, and the like.
  • aryl refers to an aromatic cyclic group containing no heteroatoms in the ring, which aryl group may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring where it is attached to the parent structure
  • the ring is an aryl ring.
  • phenyl i.e. six-membered aromatic ring
  • naphthyl etc.
  • Aryl groups may be optionally substituted or unsubstituted.
  • heteroaryl refers to a cyclic aromatic group having 1-3 heteroatoms selected from the group consisting of N, S and O
  • heteroaryl refers to a cyclic aromatic group having 5-6 heteroatoms.
  • Heteroaryl can be a single ring (for example, 5-6 members) or a condensed ring (for example, 8-10 members).
  • Specific examples may be pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyridone, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3)-triazolyl and (1 ,2,4)-triazolyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, indolyl, isoindolyl, indazolyl, quinolyl, wait.
  • the heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl ring, where the ring attached to the parent structure is the heteroaryl ring.
  • Heteroaryl groups may be optionally substituted or unsubstituted.
  • the substituent is preferably one or more groups independently selected from alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, alkylthio , alkylamino, halogen, amino, nitro, Hydroxy, mercapto, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylthio, oxo, amide, sulfonamide, formyl, formamide, carboxyl and carboxylate Key et al.
  • halogen or "halogen atom” refers to F, Cl, Br, and I. More preferably, the halogen or halogen atom is selected from F, Cl and Br.
  • -C1-C6 alkylene 4-10 membered N-containing heterocyclyl group refers to a C1-C6 alkylene group substituted by a 4-10 membered N-containing heterocyclyl group, preferably -C1-C3 alkylene
  • the base is a 4-6 membered N-containing heterocyclic group, for example, wait.
  • "-C1-C6 alkylene C3-C10 cycloalkyl” has a similar meaning.
  • substituted means that one or more hydrogen atoms on a specific group are replaced by a specific substituent.
  • Specific substituents are the substituents described accordingly in the foregoing text, or the substituents appearing in each embodiment.
  • a substituted group may have a substituent selected from a specific group at any substitutable position of the group, and the substituents may be the same or different at each position. It will be understood by those skilled in the art that combinations of substituents contemplated by the present invention are those that are stable or chemically achievable.
  • substituted or unsubstituted the groups described in the present invention can be substituted with substituents selected from the following group: deuterium, halogen, cyano, nitro, hydroxyl , amino, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, 3-12 membered heterocyclyl, C3-C12 cycloalkyl, 5-12 membered heteroaryl , C6-C12 aryl.
  • substituents selected from the following group: deuterium, halogen, cyano, nitro, hydroxyl , amino, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, 3-12 membered heterocyclyl, C3-C12 cycloalkyl, 5-12 membered heteroaryl , C6-C12 aryl.
  • the structural formulas described in the present invention are intended to include all isomeric forms (such as enantiomers, diastereomers and geometric isomers (or conformational isomers)): for example, containing asymmetric The R and S configurations of the center, the (Z) and (E) isomers of the double bond, etc. Therefore, individual stereochemical isomers of the compounds of the present invention or mixtures of their enantiomers, diastereomers or geometric isomers (or conformational isomers) are within the scope of the present invention.
  • tautomer means that structural isomers with different energies can exceed a low energy barrier and thereby convert into each other.
  • proton tautomers i.e., proton transfer
  • interconversion through proton migration such as 1H-indazole and 2H-indazole.
  • Valence tautomers involve interconversions through the recombination of some of the bonding electrons.
  • solvate refers to a complex in which a compound of the invention is coordinated with solvent molecules to form a complex in a specified proportion.
  • compounds of the present invention refers to compounds represented by Formula I, Formula I' or Formula II, and also includes pharmaceutically acceptable salts, stereoisomers, solvates or precursors of the compounds of Formula (A). medicine.
  • salts formed by the compounds in the present invention also belong to the scope of the present invention.
  • compounds in the present invention are understood to include salts thereof.
  • the term "salt” as used herein refers to an acidic or basic salt formed from an inorganic or organic acid and a base.
  • the compound of the present invention contains a basic moiety, it includes but is not limited to pyridine.
  • Zwitterions that may be formed when containing an acidic moiety, including but not limited to carboxylic acids, are included within the scope of the term "salt”.
  • Pharmaceutically acceptable (i.e., nontoxic, physiologically acceptable) salts are preferred, although other salts are also useful, for example, in isolation or purification steps during preparation.
  • the compounds of the present invention may form salts, for example, compound I can be obtained by reacting with a certain amount of, for example, an equivalent amount of acid or base, salting out in a medium, or by freeze-drying in an aqueous solution.
  • the compounds of the present invention contain basic moieties, including but not limited to amines or pyridine or imidazole rings, which may form salts with organic or inorganic acids.
  • Typical acids that can form salts include acetates (eg, with acetic acid or trihaloacetic acids, such as trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, and benzoates.
  • benzenesulfonate hydrogen sulfate, borate, butyrate, citrate, camphor salt, camphor sulfonate, cyclopentane propionate, diglycolate, dodecyl sulfate, Ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, enanthate, caproate, hydrochloride, hydrobromide, hydroiodide, glycolate (e.g., 2-hydroxyethanesulfonate), lactate, maleate, methanesulfonate, naphthalenesulfonate (e.g., 2-naphthalenesulfonate), nicotinate, nitrate, oxalic acid Salt, pectate, persulfate, phenylpropionate (such as 3-phenylpropionate), phosphate, picrate, pivalate, propionate, sal
  • Certain compounds of the invention may contain acidic moieties, including but not limited to carboxylic acids, which may form salts with various organic or inorganic bases.
  • Typical salts formed with bases include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, and salts formed with organic bases (such as organic amines), such as benzathine and dicyclohexylamine.
  • Hypamine salt with N,N-bis(dehydroabidyl)ethylenediamine
  • N-methyl-D-glucamine N-methyl-D-glucamide
  • tert-butyl Amines and salts formed with amino acids such as arginine, lysine, etc.
  • Basic nitrogen-containing groups can be combined with halide quaternary ammonium salts, such as small molecule alkyl halides (such as chlorides, bromides and iodides of methyl, ethyl, propyl and butyl), dialkyl sulfates (such as dimethyl sulfate, diethyl sulfate, dibutyl ester and dipentyl ester), long chain halides (such as decyl, dodecyl, tetradecyl and tetradecyl chlorides, bromides and iodide), aralkyl halides (such as benzyl and phenyl bromide), etc.
  • small halides such as chlorides, bromides and iodides of methyl, ethyl, propyl and butyl
  • dialkyl sulfates such as dimethyl sulfate, diethyl sulfate, dibutyl este
  • Prodrugs and solvates of the compounds of the present invention are also within the scope of the invention.
  • the term "prodrug” here refers to a compound that undergoes chemical transformation through metabolism or chemical processes to produce the compound, salt, or solvate of the present invention when treating related diseases.
  • Compounds of the present invention include solvates, such as hydrates.
  • the compounds, salts or solvates of the present invention may exist in tautomeric forms (eg amides and imine ethers). All such tautomers are part of the present invention.
  • All stereoisomers of the compounds are contemplated by the present invention.
  • the compounds of the invention may be independent stereoisomers that do not exist simultaneously with other isomers (e.g., have a specific activity as a pure or substantially pure optical isomer), or they may be mixtures, e.g. Racemates, or mixtures with all other stereoisomers or portions thereof.
  • the chiral center of the present invention has two configurations: S or R, which are defined as recommended by the International Union of Theoretical and Applied Chemistry (IUPAC) in 1974.
  • the racemic form can be resolved by physical means, For example, fractional crystallization, or separation and crystallization by derivatization into diastereoisomers, or separation by chiral column chromatography.
  • Individual optical isomers can be obtained from the racemate by suitable methods, including but not limited to traditional methods, such as salt formation with an optically active acid followed by recrystallization.
  • the weight content of the compounds in the present invention obtained by sequential preparation, separation and purification is equal to or greater than 90%, for example, equal to or greater than 95%, equal to or greater than 99% ("very pure" compounds), as described in the text List. Such "very pure” compounds of the invention are here also included as part of the invention.
  • Certain compounds of the present invention may exist in specific geometric or stereoisomeric forms.
  • the present invention encompasses all compounds, including their cis and trans isomers, R and S enantiomers, diastereomers, (D) isomers, (L) isomers, elimination Spin mixtures and other mixtures.
  • asymmetric carbon atoms can represent substituents, such as alkyl groups. All isomers, as well as mixtures thereof, are included in the present invention.
  • the mixture of isomers may contain the isomers in various ratios.
  • a mixture of only two isomers can have the following combinations: 50:50, 60:40, 70:30, 80:20, 90:10, 95:5, 96:4, 97:3, 98: All ratios of isomers 2, 99:1, or 100:0 are within the scope of the invention. Similar ratios, as well as ratios for more complex mixtures of isomers that are readily understood by those of ordinary skill in the art, are also within the scope of the present invention.
  • the present invention also includes isotopically labeled compounds that are equivalent to the original compounds disclosed herein. In practice, however, it often occurs that one or more atoms are replaced by atoms with a different atomic weight or mass number.
  • isotopes of compounds that may be included in the present invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 O respectively. , 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • the compounds of the present invention or enantiomers, diastereomers, isomers, or pharmaceutically acceptable salts or solvates, which contain isotopes or other isotope atoms of the above compounds are within the scope of the present invention.
  • Certain isotopically labeled compounds of the present invention such as radioactive isotopes of 3 H and 14 C, are also included and are useful in tissue distribution experiments of drugs and substrates. Tritium, or 3H , and carbon-14, or 14C , are relatively easy to prepare and detect. It is the first choice among isotopes.
  • heavier isotope substitutions such as deuterium, i.e.
  • Isotopically labeled compounds can be labeled in the usual manner by replacing readily available isotopically labeled reagents with non-isotopically labeled reagents, using The protocols disclosed in the examples can be prepared.
  • a synthesis of a specific enantiomer of the compound of the present invention it can be prepared by asymmetric synthesis, or derivatized with a chiral auxiliary, and the resulting diastereomeric mixture is separated and then the chiral auxiliary is removed. Pure enantiomer.
  • a suitable optically active acid or base can be used to form a diastereomeric salt with it, and then through separation, crystallization or chromatography, etc. After separation by conventional means, the pure enantiomers are obtained.
  • the compounds of the present invention may be provided with any number of substituents or functional groups to broaden their encompassing scope.
  • substituents or functional groups in general, whether the term “substituted” appears before or after the term “optional”, the general formula of the substituent included in the formulation of the present invention means that the substituent of the specified structure is used in place of the hydrogen radical. When multiple positions in a specific structure are substituted by multiple specific substituents, the substituents may be the same or different at each position.
  • substitution as used herein includes all permissible substitutions of organic compounds. Broadly speaking, permissible substituents include acyclic, cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic organic compounds.
  • heteroatoms such as nitrogen may have hydrogen substituents or any of the permissible organic compounds described above to supplement their valence. Furthermore, this invention is not intended to be limited in any way to the permitted substituted organic compounds.
  • the present invention considers that combinations of substituents and variable groups are excellent in the treatment of diseases in the form of stable compounds.
  • stable refers to a compound that is stable, detectable over a long enough period of time to maintain the structural integrity of the compound, and preferably effective over a long enough period of time, and is used herein for the above purposes.
  • each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (such as 0°C to 150°C, preferably 10°C to 100°C).
  • the reaction time is usually 0.1 hour to 60 hours, preferably 0.5 to 48 hours.
  • the compounds of the present invention can be prepared by the following steps
  • the corresponding intermediate is obtained through a nucleophilic substitution reaction with an indole derivative under the action of aluminum trichloride or a Suzuki coupling reaction with a boronic acid derivative.
  • the resulting intermediate is a substitution reaction with the corresponding amine under the action of an organic base to obtain the corresponding product, and finally the I series target compounds are obtained by removing the protecting group;
  • the corresponding intermediate compound is obtained through a substitution reaction with the corresponding amine under the action of an organic base; the obtained intermediate is subjected to Suzuki coupling with a boronic acid derivative The reaction obtains the corresponding product, and finally the II series target compounds are obtained by removing the protecting group.
  • compositions and methods of administration are provided.
  • the compound of the present invention has excellent CDK kinase inhibitory activity
  • the compound of the present invention or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug, as well as a pharmaceutical composition containing the compound of the present invention as the main active ingredient It can be used to prevent and/or treat (stabilize, alleviate or cure) CDK kinase related diseases such as tumors (such as colorectal cancer, breast cancer, lung cancer, ovarian cancer or gastric cancer).
  • the pharmaceutical composition of the present invention contains a compound of the present invention and a pharmaceutically acceptable excipient or carrier within a safe and effective amount.
  • the “safe and effective dose” refers to the amount of compound that is sufficient to significantly improve the condition without causing serious side effects.
  • the pharmaceutical composition contains 1-2000 mg of the compound of the present invention/dose, more preferably, it contains 10-200 mg of the compound of the present invention/dose.
  • the "dose" is a capsule or tablet.
  • “Pharmaceutically acceptable carrier” refers to one or more compatible solid or liquid filler or gel substances that are suitable for human use and must be of sufficient purity and low enough toxicity. "Compatibility” here means that the components of the composition can be blended with the compounds of the present invention and with each other without significantly reducing the efficacy of the compounds.
  • Examples of pharmaceutically acceptable carriers include cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid , magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as Tween ), wetting agents (such as sodium lauryl sulfate), colorants, flavorings, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
  • cellulose and its derivatives such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • gelatin such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.
  • the administration mode of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative administration modes include (but are not limited to): oral, parenteral (intravenous, intramuscular or subcutaneous).
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules.
  • the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or compatibilizers, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) Binders, for example, hydroxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and gum arabic; (c) Humectants, For example, glycerol; (d) disintegrants, such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) retarder, such as paraffin; (f) Absorption accelerators, such as quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glyceryl mono
  • Solid dosage forms such as tablets, dragees, capsules, pills and granules may be prepared using coatings and shell materials such as enteric casings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be released in a delayed manner in a certain part of the digestive tract. Examples of embedding components that can be used are polymeric substances and waxy substances. If necessary, the active compounds can also be in microencapsulated form with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures.
  • Liquid dosage forms may contain, in addition to the active compound, inert diluents conventionally employed in the art, such as water or other solvents.
  • Agents, solubilizers and emulsifiers for example, ethanol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, Corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances, etc.
  • compositions may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents.
  • Suspensions may contain, in addition to the active compound, suspending agents, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these substances and the like.
  • suspending agents for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these substances and the like.
  • compositions for parenteral injection may contain physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
  • the compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds (eg, CDK inhibitors).
  • other pharmaceutically acceptable compounds eg, CDK inhibitors.
  • the pharmaceutical composition When administered in combination, the pharmaceutical composition also includes one or more (2, 3, 4, or more) other pharmaceutically acceptable compounds (eg, CDK inhibitors).
  • one or more (2, 3, 4, or more) of the other pharmaceutically acceptable compounds may be used simultaneously, separately, or sequentially with the compound of the invention to prevent and/or treat CDK Diseases related to kinase activity or expression.
  • a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, and the dosage when administered is a pharmaceutically effective dosage.
  • a mammal such as a human
  • the daily dose is usually 1 to 2000 mg, preferably 20 to 500 mg.
  • the specific dosage should also take into account factors such as the route of administration and the patient's health condition, which are all within the skill of a skilled physician.
  • the compound of the present invention has a novel structure and has excellent CDK kinase inhibitory activity, especially CDK7 or CDK2;
  • the compounds of the present invention can be used as CDK kinase inhibitors, especially as highly selective inhibitors of CDK7 or CDK2.
  • the compound of the present invention has good pharmacokinetics and efficacy.
  • Trifluoroacetic acid (1 mL) was added to a solution (5 mL) of compound I-1-4 (110 mg, 210 ⁇ mol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain light yellow solid I-1 (25 mg).
  • Trifluoroacetic acid (1 mL) was added to a solution (5 mL) of compound I-2-1 (30 mg, 54 ⁇ mol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-2 (10 mg).
  • Trifluoroacetic acid (1 mL) was added to a solution (5 mL) of compound I-3-1 (30 mg, 57 ⁇ mol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain light yellow solid I-3 (15 mg).
  • Trifluoroacetic acid (1 mL) was added to a solution (5 mL) of compound I-4-2 (30 mg, 57 ⁇ mol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain light yellow solid I-4 (12 mg).
  • Trifluoroacetic acid (0.3 mL) was added to a solution (3 mL) of compound I-5-2 (80 mg, 57 ⁇ mol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain light yellow solid I-5 (30 mg).
  • Trifluoroacetic acid (0.1 mL) was added to a solution (1 mL) of compound I-6-6 (30 mg, 51 ⁇ mol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-6 (11 mg). LCMS(M+H):483.
  • Trifluoroacetic acid (0.1 mL) was added to a solution (1 mL) of compound I-7-8 (20 mg, 36 ⁇ mol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours.
  • the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product.
  • the crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-7 (9 mg).
  • Compound I-35 was synthesized by referring to the synthetic route of compound I-1 in Example 1. Compound 4-fluoro-6-bromoindole was replaced with 6-bromoindole as the starting material. The obtained compound I-35 was a light yellow solid. (11 mg), LCMS (M+H): 444.
  • Compound I-36 was synthesized by referring to the synthetic route of compound I-1 in Example 1, replacing (S)-3-aminopiperidine-1-carboxylic compound with trans-(4-aminocyclohexyl)carbamic acid tert-butyl ester. Acid tert-butyl ester, the obtained compound I-36 was a light yellow solid (12 mg), LCMS (M+H): 440.
  • Compound I-37 was synthesized by referring to the synthetic route of compound I-1 in Example 1, except that (S)-3 was replaced with compound 3-(aminomethyl)-3-methyl-1-azetidinecarboxylic acid tert-butyl ester. -Aminopiperidine-1-carboxylic acid tert-butyl ester, the obtained compound I-37 was a light yellow solid (12 mg), LCMS (M+H): 426.
  • Compound I-38 was synthesized by referring to the synthetic route of compound I-1 in Example 1, except that compound (R)-2-aminomethyl-1-N-Boc-pyrrolidine was replaced with (S)-3-aminopiperidine- 1-tert-butylcarboxylate, the obtained compound 1-38 was a light yellow solid (8 mg), LCMS (M+H): 426.
  • Compound I-40 was synthesized by referring to the synthetic route of compound I-1 in Example 1, except that compound 3-(aminomethyl)-3-fluoro-1-azetidinecarboxylic acid tert-butyl ester was replaced with (S)-3- Aminopiperidine-1-carboxylic acid tert-butyl ester, the obtained compound I-40 was a light yellow solid (7 mg), LCMS (M+H): 430.
  • Compound I-41 was synthesized by referring to the synthetic route of compound I-1 in Example 1, except that compound 1-tert-butoxycarbonyl-3-(aminomethyl)pyrrolidine was replaced with (S)-3-aminopiperidine-1- Tert-butyl carboxylate, the obtained compound I-41 was a light yellow solid (11 mg), LCMS (M+H): 426.
  • Compound I-42 was synthesized by referring to the synthetic route of compound I-1 in Example 1, replacing (S)-3-aminopiperidine with compound ((1R,3S)-3-aminocyclopentyl)carbamate tert-butyl ester. -1-tert-butylcarboxylate, the obtained compound I-42 was a light yellow solid (13 mg), LCMS (M+H): 426.
  • Compound I-43 was synthesized by referring to the synthetic route of compound I-1 in Example 1, replacing (S)-3-aminopiperidine with compound ((1S,3S)-3-aminocyclopentyl)carbamate tert-butyl ester. -1-tert-butylcarboxylate, the obtained compound I-43 was a light yellow solid (14 mg), LCMS (M+H): 426.
  • reaction solution is cooled to room temperature, added to 20 mL of water, and then extracted with ethyl acetate (15 mL*3), combined the organic phases, washed with 20 mL saturated brine, and the resulting organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain yellow oil II-30-1 (90 mg).
  • Compound I-44 was synthesized by referring to the synthetic route of compound I-7 in Example 1, replacing (S)-3-aminopiperidine-1 with compound (1S,3S)-3-aminocyclopentylcarbamate tert-butyl ester. -tert-butyl carboxylate, the obtained compound I-44 was a light yellow solid (10 mg), LCMS (M+H): 451.2.
  • Compound I-45 was synthesized by referring to the synthetic route of compound I-7 in Example 1, except that compound 5-amino-3,3-difluoropiperidine-1-carboxylic acid tert-butyl ester was replaced with (S)-3-aminopiperidine. -1-tert-butylcarboxylate, the obtained compound I-45 was a light yellow solid (9 mg), LCMS (M+H): 487.3.
  • Compound I-46 was synthesized by referring to the synthetic route of compound I-7 in Example 1, replacing (S)-3- with compound (3S,5S)-3-amino-5-fluoroperidine-1-carboxylic acid tert-butyl ester. Aminopiperidine-1-carboxylic acid tert-butyl ester, the obtained compound I-46 was a light yellow solid (13 mg), LCMS (M+H): 469.2.
  • compound I-47 was based on the synthetic route of compound I-7 in Example 1, except that compound (3S,5S)-3-amino-5-methylpiperidine-1-carboxylic acid tert-butyl ester was replaced with (S)-3. -Aminopiperidine-1-carboxylic acid tert-butyl ester, the obtained compound I-47 was a light yellow solid (11 mg), LCMS (M+H): 465.2.
  • Trifluoroacetic acid (0.2 mL) was added to a solution (2 mL) of compound I-1-3 (20 mg, 39 ⁇ mol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 10 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-8 (10 mg). LCMS(M+H):428.
  • Test Example 1 Biochemical test of compounds inhibiting CDK7/cyclinH/MANT1 kinase protein activity
  • CDK7/cyclinH/MNAT1 kinase protein complex and its substrate peptide CDK7/9tide in 50mM HEPES Prepare a 2X solution in a buffer system consisting of pH 7.5, 0.01% BRIJ-35, 10mM MgCl 2 and 1mM EGTA.
  • Detection mixture is prepared from EDTA (30mM), Eu-anti-ADP antibody (6nM) and ADP tracer in TR-FRET dilution buffer.
  • the 10uL enzyme reaction system contains 20ng CDK7/cyclinH/MNAT1 kinase protein complex, 200uM substrate peptide CDK7/9tide and 150uM ATP.
  • the compound to be tested was diluted 3 times downward starting from the highest concentration of 10uM, for a total of 11 concentration points. Set up 2 multiple holes. Calculate the inhibition rate of each well based on the fluorescence reading, draw a dose-dependent curve, and calculate the half inhibitory concentration IC 50 of the tested compound. The results are shown in Table 1 below:
  • Test Example 2 Biochemical test of compounds inhibiting CDK2/cyclinE kinase protein activity
  • GST-tagged CDK2/cyclinE kinase protein complex and Eu-anti-GST were prepared into a 2X solution in a buffer system consisting of 50mM HEPES pH 7.5, 0.01% BRIJ-35, 10mM MgCl 2 and 1mM EGTA.
  • AlexaFluor-labeled Tracer is prepared as a 2X solution in Kinase buffer.
  • the final incubation system contained 2.5nM CDK2/cyclinE kinase protein complex, 2nM Eu-anti-GST and 100nM AlexaFluor-labeled Tracer.
  • the compound to be tested was diluted 3 times downward starting from the highest concentration of 10uM, for a total of 11 concentration points. Set up 2 multiple holes. Calculate the inhibition rate of each well based on the fluorescence reading, draw a dose-dependent curve, and calculate the half inhibitory concentration IC 50 of the tested compound. The results are as follows in Table 3:
  • Test Example 3 Test of anti-proliferative activity of compounds on triple-negative breast cancer cell line MDA-MB-453
  • MDA-MB-453 cells were seeded in a 96-well culture plate at a density of 4000 cells/well: the outer circle of cells was filled with 200uL PBS to prevent the rapid evaporation of the edge medium from causing excessive differences in the culture conditions of the inner plate wells.
  • the leftmost column of the 60 internal wells is a blank well, which is filled with an equal volume of PBS without adding cells.
  • the remaining 54 wells are plated with cells using a row gun. Each well is 100 uL and placed in a carbon dioxide incubator for 24 hours at 37°C. Use complete culture medium to dilute the compound stock solution, and add 50uL of drug solution of corresponding concentration to each well, with a final concentration of up to 10uM.
  • Test Example 4 Test of anti-proliferative activity of compounds on ovarian cancer cell line OVCAR3
  • OVCAR3 cells were seeded in a 96-well culture plate at a density of 2000 cells/well: the outer circle of cells was filled with 200uL PBS to prevent the rapid evaporation of the edge medium from causing excessive differences in the culture conditions of the inner plate wells.
  • the leftmost column of the 60 internal wells is a blank well, which is filled with an equal volume of PBS without adding cells.
  • the remaining 54 wells are plated with cells using a row gun. Each well is 100 uL and placed in a carbon dioxide incubator for 24 hours at 37°C. Use complete culture medium to dilute the compound stock solution, add 50uL of drug solution of corresponding concentration to each well, the final concentration is up to 10uM, and dilute 3 times downwards.
  • liver microsomes used in the experiment.
  • composition of the experimental incubation system is the composition of the experimental incubation system:
  • liver microsomes Take out the liver microsomes from the -80°C refrigerator, place them in a 37°C water bath constant-temperature oscillator to pre-incubate for 3 minutes, thaw and set aside for use.
  • Control group (without ⁇ -NADPH): Take 30 ⁇ L of the NCF control group as the 0min sample, add 180 ⁇ L of precipitant containing internal standard, and then add 30 ⁇ L of NADPH to obtain the 0min sample; add 30 ⁇ L of magnesium chloride to the remaining NCF group, incubate for 60 minutes, and then add 180 ⁇ L contains internal standard precipitating agent.
  • Sample group Take 30 ⁇ L of the sample group as the 0min sample, add 180 ⁇ L of precipitant containing internal standard, and then add 30 ⁇ L of NADPH to obtain the 0min sample; add 30 ⁇ L of NADPH to each tube of the remaining sample group, incubate for 5, 15, 30, and 60 min, then add 180 ⁇ L Contains internal standard precipitant.
  • Positive control group Take the PB solution, add the calculated liver microsomes of the corresponding species, then add the positive control working solution, vortex to mix, and divide into tubes. Add NADPH solution for incubation, and add 180 ⁇ L of precipitating agent at 5 and 15 min. Divide into two other tubes, first add the internal standard precipitant, and then add NADPH to obtain the 0min sample.
  • Test Example 6 Selectivity test of compounds inhibiting the activity of CDK family kinase proteins
  • I-7 has good selectivity for inhibiting CDK7; II-3 also has good inhibitory activity against CDK7, but it also inhibits multiple other CDK family kinase members, including CDK12, CDK13, CDK9, and CDK18. .
  • Animal dietary status The animals are not fasting before administration and can drink water freely.
  • Compound preparation method 0.5% CMC-Na was used for intragastric administration (G1 group), and 5% DMSO+30% PEG400+65% water for injection was used for intravenous injection (G2 group).
  • G1 group sampling time points 10min, 30min, 1h, 2h, 4h, 6h, 8h, 24h after administration, a total of 8 points.
  • G2 group sampling time points 5min, 10min, 30min, 1h, 2h, 4h, 6h, 8h, 24h after administration, a total of 9 points.
  • Sample collection On the day of the experiment, collect approximately 0.1 mL of whole blood samples at each set time point and place them in EDTA-K2 anticoagulant tubes.
  • Sample processing Within 30 minutes after collection, whole blood samples were centrifuged for 5 minutes at 4°C and 12,000 rpm, and the plasma was separated. The upper plasma samples were collected into sample tubes. The plasma samples were frozen and stored in a -10 ⁇ -30°C refrigerator within 30 minutes. And transfer to -60 ⁇ -90°C refrigerator within 24 hours.
  • IV* refers to the intravenous route
  • PO* refers to the intragastric route
  • Test Example 8 Anti-tumor efficacy test of the compound on the nude mouse transplanted tumor model of the triple-negative breast cancer cell line MDA-MB-231
  • a transplanted tumor model of the human triple-negative breast cancer cell line MDA-MB-231 was constructed on nude mice to evaluate the anti-tumor efficacy of compound I-46.
  • the specific steps are as follows:
  • MDA-MB-231 cells were cultured in DMEM culture medium containing 10% fetal bovine serum at 37°C and 5% CO2 in a cell culture incubator. After the cells were full grown, they were divided into bottles and passaged every 2 to 3 days. Tumor cells in the logarithmic growth phase are used for in vivo tumor seeding.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present application discloses a small molecule compound having a pyrimidothiophene structure, and a use thereof. By means of inhibiting activity of the cyclin-dependent kinase (CDK) family, the small molecule compound can block the cell cycle process and hinder the transcription of oncogenes, thereby inhibiting tumor cell proliferation and achieving the purpose of treating tumors.

Description

具有嘧啶并噻吩结构的小分子化合物及其应用Small molecule compounds with pyrimidothiophene structure and their applications 技术领域Technical field
本发明涉及化学药物领域,特别涉及一类具有嘧啶并噻吩结构的小分子化合物及其应用。The present invention relates to the field of chemical drugs, and in particular to a class of small molecule compounds with a pyrimidothiophene structure and their applications.
背景技术Background technique
细胞周期蛋白依赖性激酶家族(cyclin-dependent kinase,CDK)是丝氨酸/苏氨酸蛋白激酶,可调节转录(CDK 7-13和19-20)或细胞周期进程(CDK 1-6和14-18)。一方面,已有确凿的证据支持过度激活癌症转录程序的遗传变异可作为潜在靶标,并激起了人们开发阻止过度转录疗法的热情;另一方面,细胞周期失调是癌症的标志,靶向细胞周期调控因子的药物CDK4/6抑制剂已经上市并获得了巨大成功。因此,以CDK家族激酶为靶点的药物是重要的肿瘤治疗手段。The cyclin-dependent kinase family (CDK) is a serine/threonine protein kinase that regulates transcription (CDK 7-13 and 19-20) or cell cycle progression (CDK 1-6 and 14-18 ). On the one hand, there is conclusive evidence supporting genetic variations that overactivate cancer transcriptional programs as potential targets and spurring enthusiasm for the development of therapies that block overtranscription. On the other hand, cell cycle dysregulation is a hallmark of cancer, and targeting cells Drugs CDK4/6 inhibitors of cycle regulators have been launched and have achieved great success. Therefore, drugs targeting CDK family kinases are important tumor treatments.
CDK可以和cyclin结合形成异二聚体,其中CDK为催化亚基,cyclin为调节亚基,不同的cyclin—CDK复合物,通过CDK活性,催化不同底物磷酸化,而实现对细胞周期不同时相的推进和转化作用。CDK的活性依赖于其正调节亚基cyclin的顺序性表达和其负调节亚基CKI(cyclin dependent kinase inhibitor,CDK抑制因子)的浓度。同时,CDK的活性还受到磷酸化和去磷酸化,以及癌基因和抑癌基因的调节。正常情况下,静默期的细胞在有丝分裂信号的刺激下,由G0期进入到有丝分裂Gl期,细胞内部活动逐渐上调。CDK4/6与cyclinD结合,促使视网膜母细胞瘤蛋白(Rb)磷酸化,部分转录延展因子E2F的活性得以释放,用于合成大量的RNA与蛋白质,为染色体复制做准备。G1后期,CDK2与cyclinE结合形成蛋白酶体复合物CDK2-cyclinE并活化,促使Rb进一步磷酸化,诱导转录因子E2F的持续表达,从而调控细胞顺利通过G1期。进入S期后,CDK2与cyclinA结合形成复合物CDK2-cyclinA参与到细胞周期S期的进程,完成DNA的复制。随后,cyclinA/B与CDK1形成CDK1-cyclinA/B复合物,驱动细胞周期进入G2/M期,合成RNA与蛋白质为纺锤丝的形成做准备。最终,细胞有序地经过G1-S-G2-M所有细胞周期完成细胞的有丝分裂。几乎所有的CDK激活都通过:(1)与细胞周期蛋白(Cyclin)结合;(2)T-Loop被CDK激活激酶(CDK Activating Kinase,CAK)磷酸化。CAK是一种三元复合物,由CDK7、细胞周期蛋白H(Cyclin H)与环-指蛋白MAT1(RING-finger protein MAT1)组成。CDK can combine with cyclin to form a heterodimer, in which CDK is the catalytic subunit and cyclin is the regulatory subunit. Different cyclin-CDK complexes catalyze the phosphorylation of different substrates through CDK activity, thereby achieving different effects on the cell cycle. The advancement and transformation of phases. The activity of CDK depends on the sequential expression of its positive regulatory subunit cyclin and the concentration of its negative regulatory subunit CKI (cyclin-dependent kinase inhibitor, CDK inhibitor). At the same time, CDK activity is also regulated by phosphorylation and dephosphorylation, as well as oncogenes and tumor suppressor genes. Under normal circumstances, cells in the silent phase enter the Gl phase of mitosis from the G0 phase under the stimulation of mitotic signals, and the internal activities of the cells gradually increase. CDK4/6 combines with cyclinD to promote the phosphorylation of retinoblastoma protein (Rb), and the activity of part of the transcription elongation factor E2F is released, which is used to synthesize a large amount of RNA and proteins to prepare for chromosome replication. In the late G1 phase, CDK2 combines with cyclinE to form the proteasome complex CDK2-cyclinE and is activated, which promotes further phosphorylation of Rb and induces the continued expression of the transcription factor E2F, thereby regulating cells to successfully pass through the G1 phase. After entering the S phase, CDK2 combines with cyclinA to form a complex CDK2-cyclinA, which participates in the S phase of the cell cycle and completes DNA replication. Subsequently, cyclinA/B and CDK1 form the CDK1-cyclinA/B complex, driving the cell cycle into the G2/M phase, synthesizing RNA and proteins to prepare for the formation of spindle fibers. Finally, the cells go through all the cell cycles of G1-S-G2-M in an orderly manner to complete cell mitosis. Almost all CDK activations occur through: (1) Binding to Cyclin; (2) T-Loop is phosphorylated by CDK Activating Kinase (CAK). CAK is a ternary complex consisting of CDK7, Cyclin H and RING-finger protein MAT1.
CDK7的独特之处在于其同时涉及转录与细胞周期的调控,它在各种类型的癌症中普遍表达,其下调导致细胞增殖减少,CDK7被认为是可行的癌症治疗靶标。人们已在多种癌症类型中检测到CDK7高表达,并与侵袭性临床病理特征和不良预后相关。CDK7在肝细胞癌、胃癌和结肠直肠癌(CRC)中扩增。对173份胃癌标本的免疫组织化学分析显示,CDK7水平升高与肿瘤分级有关。同样,CDK7蛋白在大部分口腔鳞状细 胞癌标本中高表达,与T分期增高、无疾病生存率降低有关,表明其可作为预后生物标志物。与相邻的正常乳腺组织相比,在癌性乳腺组织中的CDK7蛋白和mRNA水平上调。The unique feature of CDK7 is that it is involved in the regulation of both transcription and cell cycle. It is ubiquitously expressed in various types of cancer, and its downregulation leads to reduced cell proliferation. CDK7 is considered a feasible cancer therapeutic target. High CDK7 expression has been detected in multiple cancer types and is associated with aggressive clinicopathological features and poor prognosis. CDK7 is amplified in hepatocellular carcinoma, gastric cancer, and colorectal cancer (CRC). Immunohistochemical analysis of 173 gastric cancer specimens showed that elevated CDK7 levels were related to tumor grade. Similarly, CDK7 protein is expressed in most oral squamous cells High expression in cell carcinoma specimens is associated with increased T stage and decreased disease-free survival rate, indicating that it can be used as a prognostic biomarker. CDK7 protein and mRNA levels are upregulated in cancerous breast tissue compared with adjacent normal breast tissue.
从上面描述的细胞周期过程可以看出,CDK2直接参与细胞周期调控且起关键作用。CDK2的活性依赖于与cyclin E或cyclin A的结合,调控细胞G1-S期的转变以及S期的进程。研究表明,CDK2介导了CDK4/6抑制剂药物的耐药,并且CCNE扩增(cyclin E高表达)的肿瘤对CDK2敲除高度敏感。It can be seen from the cell cycle process described above that CDK2 is directly involved in cell cycle regulation and plays a key role. The activity of CDK2 depends on the combination with cyclin E or cyclin A, regulating the transition of cells from G1 to S phase and the progression of S phase. Studies have shown that CDK2 mediates resistance to CDK4/6 inhibitor drugs, and tumors with CCNE amplification (high expression of cyclin E) are highly sensitive to CDK2 knockout.
长期以来,发现CDK抑制剂一直是学术界与工业界追求的目标,过去十几年已经发现很多泛CDK抑制剂(pan-CDK inhibitors)。然而,由于缺乏选择性,第一代的CDK抑制剂表现出很强的副作用,大大阻碍了临床开发。因此,开发能够选择性抑制CDK7或CDK2活性并且能够在体内发挥药效的药物,对肿瘤的治疗具有重要意义。The discovery of CDK inhibitors has been a goal pursued by academia and industry for a long time. Many pan-CDK inhibitors (pan-CDK inhibitors) have been discovered in the past decade. However, due to lack of selectivity, first-generation CDK inhibitors exhibited strong side effects, which greatly hindered clinical development. Therefore, the development of drugs that can selectively inhibit the activity of CDK7 or CDK2 and exert pharmacological effects in the body is of great significance for the treatment of tumors.
发明内容Contents of the invention
为解决上述问题,本发明的目的在于提供一种嘧啶并噻吩结构的小分子化合物及其应用,其作为选择性CDK7或CDK2抑制剂能够在体外和体内实验中抑制CDK的激酶功能,并且阻断致癌基因的转录和失控的细胞周期进程,达到抑制肿瘤细胞增殖的效果。In order to solve the above problems, the object of the present invention is to provide a small molecule compound with a pyrimidothiophene structure and its application. As a selective CDK7 or CDK2 inhibitor, it can inhibit the kinase function of CDK in in vitro and in vivo experiments, and block The transcription of oncogenes and uncontrolled cell cycle progression achieve the effect of inhibiting tumor cell proliferation.
本发明的第一方面,提供一种式I或式I’所示的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,
The first aspect of the present invention provides a compound represented by Formula I or Formula I' or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof,
其中,X为S或P;Among them, X is S or P;
X’和X”各自独立地为CH或N;X’ and X” are each independently CH or N;
Y和Y’各自独立地为C或N;Y and Y’ are each independently C or N;
Z和Z’各自独立地为CRz或N;其中,Rz选自:H、卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、4-6元杂环基;Z and Z' are each independently CR z or N; wherein, R z is selected from: H, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2. Nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 4-6 One-membered heterocyclyl;
Rp1和Rp2各自独立地为C1-C6烷基或=O;Rp 1 and Rp 2 are each independently C1-C6 alkyl or =O;
R1选自取代或未取代的下组基团:C3-C10环烷基、-C1-C6亚烷基C3-C10环烷基、4-10元含N杂环基、-C1-C6亚烷基4-10元含N杂环基; R 1 is selected from the following substituted or unsubstituted groups: C3-C10 cycloalkyl, -C1-C6 alkylene, C3-C10 cycloalkyl, 4-10 membered N-containing heterocyclyl, -C1-C6 alkylene Alkyl 4-10 membered N-containing heterocyclic group;
R2选自:H、卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、3-6元杂环基;R 2 is selected from: H, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-6 membered heterocyclyl;
R4和R’4各自独立地选自:无、氢、卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C4烷基、C1-C4烷氧基、C3-C6环烷基、-COOC1-C6烷基、-OCOC1-C6烷基、-CONHC1-C6烷基、-CON(C1-C6烷基)2、-NHC(O)C1-C6烷基、-NHC(O)OC1-C6烷基、-NHS(O)2C1-C6烷基、-S(O)2C1-C6烷基、-S(O)N(C1-C6烷基)2、-S(O)2N(C1-C6烷基)2;其中,所述烷基、烷氧基、环烷基任选地被一个或多个Ra取代;R 4 and R' 4 are each independently selected from: None, hydrogen, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, Cyano, C1-C4 alkyl, C1-C4 alkoxy, C3-C6 cycloalkyl, -COOC1-C6 alkyl, -OCOC1-C6 alkyl, -CONHC1-C6 alkyl, -CON(C1-C6 Alkyl) 2 , -NHC(O)C1-C6 alkyl, -NHC(O)OC1-C6 alkyl, -NHS(O) 2 C1-C6 alkyl, -S(O) 2 C1-C6 alkyl , -S(O)N(C1-C6 alkyl) 2 , -S(O) 2 N(C1-C6 alkyl) 2 ; wherein, the alkyl, alkoxy, and cycloalkyl groups are optionally replaced by One or more Ra substitutions;
Rp3和R5各自独立地选自:H、卤素、OH、NH2、氰基、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷基、卤代C1-C4烷氧基;Rp 3 and R 5 are each independently selected from: H, halogen, OH, NH 2 , cyano, C1-C4 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkyl, halogenated C1-C4 alkyl Oxygen;
Rp4选自:H、卤素、C1-C6烷基、卤代C1-C6烷基、C1-C6烷氧基、卤代C1-C6烷氧基、=O;Rp 4 is selected from: H, halogen, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy, halogenated C1-C6 alkoxy, =O;
其中,Rp-1和R’p-1各各自独立地选自取代或未取代的下组基团:C1-C6烷基、C2-C6烯基、C2-C6炔基、C3-C6环烷基或3-6元杂环基;其中,所述取代是指被一个或多个Ra取代;Among them, R p-1 and R' p-1 are each independently selected from the following substituted or unsubstituted groups: C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 ring Alkyl or 3-6 membered heterocyclyl; wherein, the substitution means substitution by one or more Ra;
L1为取代或未取代的C1-C6烷基,其中,所述取代是指被一个或多个Ra取代;L 1 is a substituted or unsubstituted C1-C6 alkyl group, wherein the substitution means substitution by one or more Ra;
Rn1和Rn2各自独立地为H、C1-C6烷基、卤代C1-C6烷基、C3-C6环烷基、4-6元杂环基;其中,所述取代是指被一个或多个Ra取代;Rn 1 and Rn 2 are each independently H, C1-C6 alkyl, halogenated C1-C6 alkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl; wherein, the substitution means by one or multiple Ra substitutions;
Ra各自独立地选自:卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、3-6元杂环基。Ra is each independently selected from: halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , Nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl , 3-6 membered heterocyclic group.
在另一优选例中,当X为S时,Rp1为=O,也即 In another preferred example, when X is S, Rp 1 is=O, that is for
在另一优选例中,Y-R4为N。In another preferred embodiment, YR 4 is N.
在另一优选例中,X为P,Rp1和Rp2各自独立地为甲基、乙基、丙基。In another preferred example, X is P, and Rp 1 and Rp 2 are each independently methyl, ethyl, or propyl.
在另一优选例中,Rp-1和R’p-1各自独立地为甲基、乙基、丙基。In another preferred example, R p-1 and R' p-1 are each independently methyl, ethyl, or propyl.
在另一优选例中,提供一种式II化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,
In another preferred embodiment, a compound of formula II or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof is provided,
其中,R’1选自取代或未取代的下组基团:C1-C6烷基NRn1Rn2、C3-C10环烷基、-C1-C6亚烷基C3-C10环烷基、4-10元含N杂环基、-C1-C6亚烷基4-10元含N杂环基;Rn1和Rn2各自独立地为H、C1-C6烷基、卤代C1-C6烷基、C3-C6环烷基、4-6元杂环基;其中,所述取代是指被一个或多个Ra取代;Among them, R' 1 is selected from the following substituted or unsubstituted groups: C1-C6 alkyl NRn 1 Rn 2 , C3-C10 cycloalkyl, -C1-C6 alkylene, C3-C10 cycloalkyl, 4- 10-membered N-containing heterocyclic group, -C1-C6 alkylene group, 4-10-membered N-containing heterocyclic group; Rn 1 and Rn 2 are each independently H, C1-C6 alkyl, halogenated C1-C6 alkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl; wherein, the substitution means substitution by one or more Ra;
R’2选自:H、卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、3-6元杂环基;R' 2 is selected from: H, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, cyano, C1-C6 alkyl , C1-C6 alkoxy, C1-C6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-6 membered heterocyclyl;
W选自:苯基、萘基、5-6元单环杂芳基或8-10元稠合双环杂芳基、苯并5-10元杂环基、5-6元单环杂芳基并5-10元杂环基;W is selected from: phenyl, naphthyl, 5-6 membered monocyclic heteroaryl or 8-10 membered fused bicyclic heteroaryl, benzo 5-10 membered heterocyclyl, 5-6 membered monocyclic heteroaryl And 5-10 membered heterocyclyl;
R’5和R’6各自独立地选自:氢、卤素、羟基、硝基、氰基、C1-C6烷基、C3-C6环烷基、C1-C6烷氧基、-N(R5-1aR5-1b)、苯基、C1-C6卤代烷基、C1-C6卤代烷氧基、-NHC(O)R5-2、-NHC(O)OR5-3、-NHS(O)2R5-4、-C(O)N(R5-5aR5-5b)、-S(O)2R5-6、-S(O)(NH)R5-7、-S(O)2N(R5-8aR5-8b)或-P(O)R5-9aR5-9b;或者当R’5和R’6连接于相邻或相同环原子时,R5’和R6’与它们相连的环原子一起形成取代或未取代的C3-C6环烷基或4-6元杂环基;其中,所述取代是指被一个或多个Ra取代;R' 5 and R' 6 are each independently selected from: hydrogen, halogen, hydroxyl, nitro, cyano, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkoxy, -N(R 5-1a R 5-1b ), phenyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, - NHC(O)R 5-2 , -NHC(O)OR 5-3 , -NHS(O) 2 R 5-4 , -C(O)N(R 5-5a R 5-5b ), -S( O) 2 R 5-6 , -S(O)(NH)R 5-7 , -S(O) 2 N(R 5-8a R 5-8b ) or -P(O)R 5-9a R 5 -9b ; or when R' 5 and R' 6 are connected to adjacent or the same ring atom, R 5' and R 6' together with the ring atoms to which they are connected form a substituted or unsubstituted C3-C6 cycloalkyl or 4 -6-membered heterocyclyl; wherein the substitution means substitution by one or more Ra;
R5-1a、R5-1b、R5-2、R5-3、R5-4、R5-5a、R5-5b、R5-6、R5-7、R5-8a、R5-8b、R5-9a和R5-9b各自独立地选自:氢、C1-C6烷基、C3-C6环烷基;其中,所述C1-C6烷基、C3-C6环烷基任选地被一个或多个选自下组的基团取代:卤素、羟基;或者R5-1a和R5-1b、R5-5a和R5-5b、R5-8a和R5-8b、R5-9a和R5-9b可与它们连接的原子共同形成取代或未取代C3-C6碳环或取代或未取代4-6元杂环基,其中,所述取代是指被一个或多个Ra取代;R 5-1a , R 5-1b , R 5-2 , R 5-3 , R 5-4 , R 5-5a , R 5-5b , R 5-6 , R 5-7 , R 5-8a , R 5-8b , R 5-9a and R 5-9b are each independently selected from: hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl; wherein, the C1-C6 alkyl, C3-C6 cycloalkyl The group is optionally substituted by one or more groups selected from the group consisting of: halogen, hydroxyl; or R 5-1a and R 5-1b , R 5-5a and R 5-5b , R 5-8a and R 5 -8b , R 5-9a and R 5-9b can together form a substituted or unsubstituted C3-C6 carbocyclic ring or a substituted or unsubstituted 4-6 membered heterocyclyl group with the atoms to which they are connected, wherein the substitution refers to being replaced by One or more Ra substitutions;
Ra各自独立地选自:卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、3-6元杂环基。Ra is each independently selected from: halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, Cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- 6-membered heterocyclic group.
在另一优选例中,选自: In another preferred example, Selected from:
选自: 其中,Rp3、Rp4、Rz和R’4的定义如上所述。 Selected from: Among them, Rp 3 , Rp 4 , Rz and R' 4 are defined as above.
在另一优选例中,选自: In another preferred example, Selected from:
在另一优选例中,W环选自取代或未取代的下组基团:苯基、萘基、吡啶基、嘧啶 基、吲哚基、异吲哚基、吲唑基、喹啉基、 其中,所述取代是指被一个或多个Ra取代,Ra的定义如上所述。In another preferred embodiment, the W ring is selected from the following substituted or unsubstituted groups: phenyl, naphthyl, pyridyl, pyrimidine base, indolyl, isoindolyl, indazolyl, quinolyl, Wherein, the substitution refers to substitution by one or more Ra, and Ra is defined as above.
在另一优选例中,R’5和R’6各自独立地选自:氢、卤素、羟基、硝基、氰基、C1-C6烷基、C3-C6环烷基、C1-C6烷氧基、-NH2、-NHCH3、-NHCH(CH3)2、-C(O)NH2、-C(O)NHCH3、-C(O)CH(CH3)2、-P(O)(CH3)2、-P(O)(CH2CH3)2、-S(O)2NH2、-S(O)2NHCH3、-NHC(O)O CH3、-S(O)(NH)CH3、-NHC(O)CH3、-C(O)NH CH2CH2OH。In another preferred example, R' 5 and R' 6 are each independently selected from: hydrogen, halogen, hydroxyl, nitro, cyano, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkoxy, -NH 2 , -NHCH 3 , -NHCH(CH 3 ) 2 , -C(O)NH2, -C(O)NHCH 3 , -C(O)CH(CH 3 ) 2 , -P(O)(CH 3 ) 2 , -P(O)(CH 2 CH 3 ) 2 , -S(O) 2 NH 2 , -S(O ) 2 NHCH 3 , -NHC(O)O CH 3 , -S(O)(NH)CH 3 , -NHC(O)CH 3 , -C(O)NH CH 2 CH 2 OH.
在另一优选例中,选自: In another preferred example, Selected from:
在另一优选例中,R1选自取代或未取代的下组基团:C3-C10环烷基、-C1-C6亚烷基C3-C10环烷基、4-10元含N杂环基、-C1-C6亚烷基4-10元含N杂环基;其中,所述取代是指被一个或多个Ra取代,Ra的定义如上所述。In another preferred example, R 1 is selected from the following substituted or unsubstituted groups: C3-C10 cycloalkyl, -C1-C6 alkylene, C3-C10 cycloalkyl, 4-10 membered N-containing heterocycle group, -C1-C6 alkylene group, 4-10 membered N-containing heterocyclic group; wherein, the substitution means substitution by one or more Ra, and Ra is defined as above.
在另一优选例中,R1选自:取代或未取的5-6元含N单环杂环基(优选氮杂环丁基、四氢吡咯基、哌啶基、哌嗪基等)、取代或未取的7-10元双环或三环(如螺环、稠环、桥环)杂环基、取代或未取的-(CH2)1-3-(5-6元含N单环杂环基(优选氮杂环丁基、四氢吡咯基、哌啶基、哌嗪基等)、取代或未取的-(CH2)1-3-(7-10元双环或三环(如螺环、稠环、桥环)杂环基),其中,所述取代是指被一个或多个Ra取代,Ra的定义如上所述。In another preferred example, R 1 is selected from: substituted or unsubstituted 5-6 membered N-containing monocyclic heterocyclic groups (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.) , substituted or unsubstituted 7-10 membered bicyclic or tricyclic (such as spirocyclic, fused ring, bridged ring) heterocyclic group, substituted or unsubstituted -(CH 2 ) 1-3 -(5-6 membered N-containing Monocyclic heterocyclyl (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.), substituted or unsubstituted -(CH 2 ) 1-3 -(7-10 membered bicyclic or tricyclic Ring (such as spiro ring, fused ring, bridged ring) heterocyclyl), wherein the substitution means substitution by one or more Ra, and Ra is as defined above.
在另一优选例中,R1选自:其中,Rq选自:H、F、Cl。In another preferred example, R 1 is selected from: Among them, Rq is selected from: H, F, Cl.
在另一优选例中,选自: In another preferred example, Selected from:
在另一优选例中,R1选自: In another preferred example, R 1 is selected from:
在另一优选例中,R’1选自:C1-C6烷基NH2、C1-C6烷基NHC1-C6烷基、取代或未取的5-6元含N单环杂环基(优选氮杂环丁基、四氢吡咯基、哌啶基、哌嗪基等)、取代或未取的7-10元双环或三环(如螺环、稠环、桥环)杂环基、取代或未取的-(CH2)1-3-(5-6元含N单环杂环基(优选氮杂环丁基、四氢吡咯基、哌啶基、哌嗪基等)、取代或未取的 -(CH2)1-3-(7-10元双环或三环(如螺环、稠环、桥环)杂环基),其中,所述取代是指被一个或多个Ra取代,Ra的定义如上所述。In another preferred example, R' 1 is selected from: C1-C6 alkyl NH 2 , C1-C6 alkyl NHC1-C6 alkyl, substituted or unsubstituted 5-6 membered N-containing monocyclic heterocyclic group (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.), substituted or unsubstituted 7-10 membered bicyclic or tricyclic (such as spirocyclic, fused ring, bridged ring) heterocyclyl, substituted or untaken -(CH 2 ) 1-3 -(5-6 membered N-containing monocyclic heterocyclic group (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.), substituted or Not taken -(CH 2 ) 1-3 -(7-10 membered bicyclic or tricyclic (such as spirocyclic, fused ring, bridged ring) heterocyclyl), wherein the substitution means substitution by one or more Ra, Ra The definition is as above.
在另一优选例中,R’1选自: In another preferred example, R'1 is selected from:
在另一优选例中,Y、Y’、Z、Z’、R2、R4、R’4、Rp3、R5、Rp4、L1、Rn1和Rn2为实施例中各具体化合物所对应基团。In another preferred embodiment, Y, Y', Z, Z', R 2 , R 4 , R' 4 , Rp 3 , R 5 , Rp 4 , L 1 , Rn 1 and Rn 2 are the specific ones in the embodiment. The group corresponding to the compound.
在另一优选例中,所述化合物选自下述化合物中的任一种:










In another preferred embodiment, the compound is selected from any one of the following compounds:










在另一优选例中,所述化合物为实施例中所示化合物。In another preferred embodiment, the compound is the compound shown in the embodiment.
本发明第二方面,提供一种药物组合物,其中,所述药物组合物包括如第一方面所述的化合物,或者其药学上可接受的盐、立体异构体、溶剂化物或前药;和药学上可接受的载体。A second aspect of the present invention provides a pharmaceutical composition, wherein the pharmaceutical composition includes the compound as described in the first aspect, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof; and a pharmaceutically acceptable carrier.
在另一优选例中,所述药物组合物还包括选自下组的药物:PD-1抑制剂(如纳武单抗、派姆单抗、pidilizumab、cemiplimab或上述药物的生物类似药等)、PD-L1抑制剂(如度伐单抗、阿特珠单抗、阿维鲁单抗(avelumab)或上述药物的生物类似药等)、CD20抗体(如利妥昔单抗、奥滨尤妥珠单抗、奥法木单抗、veltuzumab,托西莫单抗、131I-托西莫单抗、替伊莫单抗、90Y-替伊莫单抗、90In-替伊莫单抗、替伊莫单抗(ibritumomab tiuxetan)等)、CD47抗体(如Hu5F9-G4、CC-90002、TTI-621、TTI-622、OSE-172、SRF-231、ALX-148、NI-1701、SHR-1603、IBI188、IMM01)、ALK抑制剂(如色瑞替尼、艾乐替尼、布加替尼、劳拉替尼、奥卡替尼)、PI3K抑制剂(如艾代拉里斯、Duvelisib、Dactolisib、Taselisib、Bimiralisib、Omipalisib、Buparlisib等)、BTK抑制剂(如依鲁替尼、Tirabrutinib、阿卡替尼、赞布替尼、Vecabrutinib等)、EGFR抑制剂(如阿法替尼、吉非替尼、厄洛替尼、拉帕替尼、达克替尼、埃克替尼、卡奈替尼、沙普替尼、Naquotinib、吡咯替尼、罗乐替尼、奥希替尼等)、VEGFR抑制剂(如索拉非尼、帕唑帕尼、瑞戈非尼、司曲替尼、Ningetinib、卡博替尼、舒尼替尼、多纳非尼等)、HDAC抑制剂(如Givinostat、Tucidinostat、伏立诺他、Fimepinostat、Droxinostat、恩替诺特、达西司特、Quisinostat、泰克地那林等)、CDK抑制剂(如帕博西尼、瑞博西尼、Abemaciclib、Milciclib、Trilaciclib、Lerociclib等)、MEK抑制剂(如司美替尼(AZD6244)、曲美替尼(GSK1120212)、PD0325901、U0126、Pimasertib(AS-703026)、PD184352(CI-1040)等)、mTOR抑制剂(如Vistusertib等)、SHP2抑制剂(如RMC-4630、JAB-3068、TNO155等),或其组合。In another preferred embodiment, the pharmaceutical composition further includes a drug selected from the following group: PD-1 inhibitors (such as nivolumab, pembrolizumab, pidilizumab, cemiplimab or biosimilars of the above drugs, etc.) , PD-L1 inhibitors (such as durvalumab, atezolizumab, avelumab or biosimilars of the above drugs, etc.), CD20 antibodies (such as rituximab, obinumab Tocilizumab, ofatumumab, veltuzumab, tositumomab, 131I-tositumomab, itumomab, 90Y- itumomab, 90In- itumomab, ibritumomab tiuxetan, etc.), CD47 antibodies (such as Hu5F9-G4, CC-90002, TTI-621, TTI-622, OSE-172, SRF-231, ALX-148, NI-1701, SHR-1603 , IBI188, IMM01), ALK inhibitors (such as ceritinib, alectinib, brigatinib, lorlatinib, occalatinib), PI3K inhibitors (such as idelalisib, Duvelisib, Dactolisib , Taselisib, Bimiralisib, Omipalisib, Buparlisib, etc.), BTK inhibitors (such as ibrutinib, Tirabrutinib, acalatinib, zanbrutinib, Vecabrutinib, etc.), EGFR inhibitors (such as afatinib, gefitinib, etc.) Ni, erlotinib, lapatinib, dacomitinib, icotinib, carnetinib, sapotinib, naquotinib, pyrotinib, loletinib, osimertinib, etc.), VEGFR inhibitors (such as sorafenib, pazopanib, regorafenib, sitrinib, Ningetinib, cabozantinib, sunitinib, donafenib, etc.), HDAC inhibitors (such as Givinostat , Tucidinostat, Vorinostat, Fimepinostat, Droxinostat, Entinostat, Dacilast, Quisinostat, Tectinaline, etc.), CDK inhibitors (such as Palbociclib, Ribociclib, Abemaciclib, Milciclib, Trilaciclib, Lerociclib, etc.), MEK inhibitors (such as selumetinib (AZD6244), trametinib (GSK1120212), PD0325901, U0126, Pimasertib (AS-703026), PD184352 (CI-1040), etc.), mTOR inhibitors (such as Vistusertib, etc.), SHP2 inhibitors (such as RMC-4630, JAB-3068, TNO155, etc.), or a combination thereof.
在另一优选例中,提供一种药物组合物的制备方法,包括步骤:将药学上可接受的载体与本发明所述第一方面所述的化合物或其立体异构体或光学异构体、药学上可接受的盐、前药或溶剂化物进行混合,从而形成药物组合物。In another preferred embodiment, a method for preparing a pharmaceutical composition is provided, including the steps of: combining a pharmaceutically acceptable carrier with the compound described in the first aspect of the present invention or its stereoisomer or optical isomer. , pharmaceutically acceptable salts, prodrugs or solvates are mixed to form a pharmaceutical composition.
在另一优选例中,本发明化合物可以制备成散剂、片剂、颗粒剂、胶囊剂、溶液剂、乳剂、混悬剂等。In another preferred embodiment, the compound of the present invention can be prepared into powders, tablets, granules, capsules, solutions, emulsions, suspensions, etc.
本发明第三方面,提供一种如第一方面所述的化合物,或其药学上可接受的盐、立 体异构体、溶剂化物或前药,或如第二方面所述的药物组合物在制备治疗抗肿瘤药物中的用途。The third aspect of the present invention provides a compound as described in the first aspect, or a pharmaceutically acceptable salt thereof, immediately isomer, solvate or prodrug, or the use of the pharmaceutical composition as described in the second aspect in the preparation of anti-tumor drugs.
在另一优选例中,所述肿瘤选自:结直肠癌、乳腺癌、肺癌、卵巢癌或胃癌,优选地为乳腺癌。In another preferred embodiment, the tumor is selected from colorectal cancer, breast cancer, lung cancer, ovarian cancer or gastric cancer, preferably breast cancer.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It should be understood that within the scope of the present invention, the above-mentioned technical features of the present invention and the technical features specifically described below (such as embodiments) can be combined with each other to form new or preferred technical solutions. Due to space limitations, they will not be described one by one here.
附图说明Description of the drawings
图1示出了本发明化合物对三阴乳腺癌细胞系MDA-MB-453的抗增殖曲线。Figure 1 shows the anti-proliferation curve of the compound of the present invention on the triple-negative breast cancer cell line MDA-MB-453.
图2示出了本发明化合物对卵巢癌细胞系OVCAR3的抗增殖曲线。Figure 2 shows the anti-proliferation curve of the compounds of the present invention on the ovarian cancer cell line OVCAR3.
图3示出了本发明化合物在三阴乳腺癌细胞系MDA-MB-231的裸鼠移植瘤模型上的抗肿瘤药效测试。Figure 3 shows the anti-tumor efficacy test of the compounds of the present invention on the nude mouse transplanted tumor model of the triple-negative breast cancer cell line MDA-MB-231.
具体实施方式Detailed ways
本发明人经过广泛而深入的研究,开发了一种嘧啶并噻吩结构化合物,其具有抑制CDK的激酶功能,阻断致癌基因的转录和失控的细胞周期进程,从而达到抑制肿瘤细胞增殖的效果。在此基础上,完成了本发明。After extensive and in-depth research, the inventor has developed a pyrimidothiophene structural compound, which has the ability to inhibit the kinase function of CDK, block the transcription of oncogenes and uncontrolled cell cycle progression, thereby achieving the effect of inhibiting tumor cell proliferation. On this basis, the present invention was completed.
在本发明中,除非特别指出,所用术语具有本领域技术人员公知的一般含义。In the present invention, unless otherwise specified, the terms used have their ordinary meanings known to those skilled in the art.
当通过从左向右书写的常规化学式描述取代基时,该取代基也同样包括从右向左书写结构式时所得到的在化学上等同的取代基。举例而言,-CH2O-等同于-OCH2-。When a substituent is described by a conventional chemical formula written from left to right, the substituent also includes substituents that are chemically equivalent when the structural formula is written from right to left. For example, -CH 2 O- is equivalent to -OCH 2 -.
如本文所用,在提到具体列举的数值中使用时,术语“约”意指该值可以从列举的值变动不多于1%。例如,如本文所用,表述“约100”包括99和101和之间的全部值(例如,99.1、99.2、99.3、99.4等)。As used herein, the term "about" when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value. For example, as used herein, the expression "about 100" includes all values between 99 and 101 and between (eg, 99.1, 99.2, 99.3, 99.4, etc.).
如本文所用,术语“含有”或“包括(包含)”可以是开放式、半封闭式和封闭式的。换言之,所述术语也包括“基本上由…构成”、或“由…构成”。As used herein, the term "contains" or "includes" can be open, semi-closed and closed. In other words, the term also includes "consisting essentially of," or "consisting of."
如本文所用,术语“烷基”包括直链或支链的烷基。例如C1-C8烷基表示具有1-8个碳原子的直链或支链的烷基,优选C1-C6烷基,更优选C1-C3烷基,烷基的实例包括但不限于甲基、乙基、丙基、异丙基、丁基、异丁基、叔丁基等。As used herein, the term "alkyl" includes straight or branched chain alkyl groups. For example, C1-C8 alkyl represents a straight-chain or branched alkyl group with 1-8 carbon atoms, preferably C1-C6 alkyl, more preferably C1-C3 alkyl. Examples of alkyl include but are not limited to methyl, Ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, etc.
如本文所用,术语“烯基”包括直链或支链的烯基。例如C2-C8烯基指具有2-8个碳原子的直链或支链的烯基,优选C2-C6烯基,更优选C2-C4烯基,烯基的实例包括但不限于乙烯基、烯丙基、1-丙烯基、异丙烯基、1-丁烯基、2-丁烯基、1-戊烯基、1-己烯基或类似基团。As used herein, the term "alkenyl" includes straight or branched chain alkenyl groups. For example, C2-C8 alkenyl refers to a linear or branched alkenyl group with 2-8 carbon atoms, preferably C2-C6 alkenyl, and more preferably C2-C4 alkenyl. Examples of alkenyl include but are not limited to vinyl, Allyl, 1-propenyl, isopropenyl, 1-butenyl, 2-butenyl, 1-pentenyl, 1-hexenyl or similar groups.
如本文所用,术语“炔基”包括直链或支链的炔基。例如C2-C8炔基是指具有2-8个碳原子的直链或支链的炔基,优选C2-C6炔基,更优选C2-C4炔基,炔基的实例包括但不限于乙炔基、丙炔基、丁炔基、戊炔基、己炔基或类似基团。 As used herein, the term "alkynyl" includes straight or branched chain alkynyl groups. For example, C2-C8 alkynyl refers to a straight-chain or branched alkynyl group with 2-8 carbon atoms, preferably C2-C6 alkynyl, more preferably C2-C4 alkynyl, examples of alkynyl include but are not limited to ethynyl , propynyl, butynyl, pentynyl, hexynyl or similar groups.
如本文所用,术语“环烷基”是指包含特定数目的C原子的环状烷基,如“C3-C12环烷基”指具有3-12个(优选3、4、5、6、7或8个)碳原子的环烷基。其可以是单环,例如环丙基、环丁基、环戊基、环己基、或类似基团。也可以是双环形式,例如桥环或螺环形式。本发明中,环烷基意在包含取代环烷基。As used herein, the term "cycloalkyl" refers to a cyclic alkyl group containing a specific number of C atoms, such as "C3-C12 cycloalkyl" refers to a cyclic alkyl group having 3 to 12 (preferably 3, 4, 5, 6, 7 or 8) cycloalkyl carbon atoms. It may be a monocyclic ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or similar groups. Bicyclic forms, such as bridged or spirocyclic forms, are also possible. In the present invention, cycloalkyl is intended to include substituted cycloalkyl.
如本文所用,术语“碳环基”是指包含特定数目的C原子饱和或部分饱和的环状基团,如“C3-C6碳环基”指具有3-6个(即3、4、5、6)碳原子的碳环基。其包括环烷基或环烯基,其可以是单环,例如环丙基、环丁基、环戊基、环己基、或类似基团。也可以是双环形式,例如桥环或螺环形式。本发明中,碳环基意在包含取代碳环基。As used herein, the term "carbocyclyl" refers to a saturated or partially saturated cyclic group containing a specified number of C atoms, such as "C3-C6 carbocyclyl" refers to a cyclic group having 3 to 6 (i.e., 3, 4, 5 , 6) Carbocyclic group of carbon atoms. This includes cycloalkyl or cycloalkenyl, which may be monocyclic, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or similar groups. Bicyclic forms, such as bridged or spirocyclic forms, are also possible. In the present invention, carbocyclyl is intended to include substituted carbocyclyl.
如本文所用,术语“C1-C6烷氧基”是指具有1-6个碳原子的直链或支链的烷氧基;其具有式C1-C6烷基-O-或-C1-C5烷基-O-C1-C5烷基(如,-CH2-O-CH2CH3、-CH2-O-(CH2)2CH3、-CH2CH2-O-CH2CH3)结构,例如,甲氧基、乙氧基、丙氧基、异丙氧基、丁氧基、异丁氧基、叔丁氧基等。As used herein, the term "C1-C6 alkoxy" refers to a straight or branched chain alkoxy group having 1 to 6 carbon atoms; which has the formula C1-C6 alkyl-O- or -C1-C5 alkyl -O-C1-C5 alkyl (e.g., -CH 2 -O-CH 2 CH 3 , -CH 2 -O-(CH 2 ) 2 CH 3 , -CH 2 CH 2 -O-CH 2 CH 3 ) Structure, for example, methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, tert-butoxy, etc.
如本文所用,“杂环基”是指具有1-3个选自N、S和O的杂原子的饱和或部分饱和的环状基团,“3-12元杂环基”是指具有3-12个原子的且其中1-3个原子为选自下组N、S和O的杂原子的饱和或部分饱和的环状基团。其可以是单环,也可以是双环形式,例如桥环或螺环形式。3-12元杂环基优选3-8元杂环基,更优选地为4-6元或6-8元杂环基。具体的实例可以为氧杂环丁烷、氮杂环丁烷、四氢-2H-吡喃基、哌啶基、哌嗪基、四氢呋喃基、吗啉基和吡咯烷基等。As used herein, "heterocyclyl" refers to a saturated or partially saturated cyclic group having 1-3 heteroatoms selected from N, S and O, and "3-12 membered heterocyclyl" refers to a cyclic group having 3 to 12-membered heterocyclyl. - a saturated or partially saturated cyclic group of 12 atoms, of which 1 to 3 atoms are heteroatoms selected from the group consisting of N, S and O. It can be a single ring or a double ring, such as a bridged ring or a spiro ring. The 3-12-membered heterocyclyl group is preferably a 3-8-membered heterocyclyl group, and more preferably is a 4-6-membered or 6-8-membered heterocyclyl group. Specific examples may be oxetane, azetidine, tetrahydro-2H-pyranyl, piperidinyl, piperazinyl, tetrahydrofuranyl, morpholinyl, pyrrolidinyl, and the like.
如本文所用,“芳基”是指环上不含杂原子的芳香族环基,所述芳基可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为芳基环。如苯基(即六元芳环)、萘基等。芳基可以是任选取代的或未取代的。As used herein, "aryl" refers to an aromatic cyclic group containing no heteroatoms in the ring, which aryl group may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring where it is attached to the parent structure The ring is an aryl ring. Such as phenyl (i.e. six-membered aromatic ring), naphthyl, etc. Aryl groups may be optionally substituted or unsubstituted.
如本文所用,“杂芳基”指具有1-3个原子为选自下组N、S和O的杂原子的环状芳香基,“5-6元杂芳基”指具有5-6个原子的且其中1-3个原子为选自下组N、S和O的杂原子的环状芳香基团。“杂芳基”可以是单环(例如5-6元),也可以是稠环形式(例如8-10元)。具体的实例可以为吡啶基、哒嗪基、嘧啶基、吡嗪基、吡啶酮、三嗪基、吡咯基、吡唑基、咪唑基、(1,2,3)-三唑基以及(1,2,4)-三唑基、四唑基、呋喃基、噻吩基、异恶唑基、噻唑基、恶唑基、吲哚基、异吲哚基、吲唑基、喹啉基、 等。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环。杂芳基可以是任选取代的或未取代的。当被取代时,取代基优选为一个或多个以下基团,其独立地选自烷基、氘代烷基、卤代烷基、烷氧基、卤代烷氧基、烯基、炔基、烷硫基、烷基氨基、卤素、氨基、硝基、 羟基、巯基、氰基、环烷基、杂环基、芳基、杂芳基、环烷硫基、氧代基、酰胺基、磺酰胺基、甲酰基、甲酰胺基、羧基和羧酸酯基等。As used herein, "heteroaryl" refers to a cyclic aromatic group having 1-3 heteroatoms selected from the group consisting of N, S and O, and "5-6 membered heteroaryl" refers to a cyclic aromatic group having 5-6 heteroatoms. Atomic cyclic aromatic groups of which 1 to 3 atoms are heteroatoms selected from the group consisting of N, S and O. "Heteroaryl" can be a single ring (for example, 5-6 members) or a condensed ring (for example, 8-10 members). Specific examples may be pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyridone, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3)-triazolyl and (1 ,2,4)-triazolyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl, indolyl, isoindolyl, indazolyl, quinolyl, wait. The heteroaryl ring may be fused to an aryl, heterocyclyl or cycloalkyl ring, where the ring attached to the parent structure is the heteroaryl ring. Heteroaryl groups may be optionally substituted or unsubstituted. When substituted, the substituent is preferably one or more groups independently selected from alkyl, deuterated alkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, alkylthio , alkylamino, halogen, amino, nitro, Hydroxy, mercapto, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylthio, oxo, amide, sulfonamide, formyl, formamide, carboxyl and carboxylate Key et al.
如本文所用,“卤素”或“卤原子”指F、Cl、Br、和I。更佳地,卤素或卤原子选自F、Cl和Br。As used herein, "halogen" or "halogen atom" refers to F, Cl, Br, and I. More preferably, the halogen or halogen atom is selected from F, Cl and Br.
如本文所用,“-C1-C6亚烷基4-10元含N杂环基”是指被4-10元含N杂环基取代的C1-C6亚烷基,优选-C1-C3亚烷基4-6元含N杂环基,例如, 等。“-C1-C6亚烷基C3-C10环烷基”具有类似含义。As used herein, "-C1-C6 alkylene 4-10 membered N-containing heterocyclyl group" refers to a C1-C6 alkylene group substituted by a 4-10 membered N-containing heterocyclyl group, preferably -C1-C3 alkylene The base is a 4-6 membered N-containing heterocyclic group, for example, wait. "-C1-C6 alkylene C3-C10 cycloalkyl" has a similar meaning.
在本发明中,术语“取代”指特定的基团上的一个或多个氢原子被特定的取代基所取代。特定的取代基为在前文中相应描述的取代基,或各实施例中所出现的取代基。除非特别说明,某个取代的基团可以在该基团的任何可取代的位点上具有一个选自特定组的取代基,所述的取代基在各个位置上可以是相同或不同的。本领域技术人员应理解,本发明所预期的取代基的组合是那些稳定的或化学上可实现的组合。In the present invention, the term "substituted" means that one or more hydrogen atoms on a specific group are replaced by a specific substituent. Specific substituents are the substituents described accordingly in the foregoing text, or the substituents appearing in each embodiment. Unless otherwise specified, a substituted group may have a substituent selected from a specific group at any substitutable position of the group, and the substituents may be the same or different at each position. It will be understood by those skilled in the art that combinations of substituents contemplated by the present invention are those that are stable or chemically achievable.
本发明所述的基团除非特别说明是“取代的或未取代的”,否则本发明的基团均可被选自下组的取代基所取代:氘、卤素、氰基、硝基、羟基、氨基、C1-C6烷基、C2-C6烯基、C2-C6炔基、C1-C6烷氧基、3-12元杂环基、C3-C12环烷基、5-12元杂芳基、C6-C12芳基。Unless otherwise specified as "substituted or unsubstituted", the groups described in the present invention can be substituted with substituents selected from the following group: deuterium, halogen, cyano, nitro, hydroxyl , amino, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C1-C6 alkoxy, 3-12 membered heterocyclyl, C3-C12 cycloalkyl, 5-12 membered heteroaryl , C6-C12 aryl.
在本发明中,术语“多个”独立指2、3、4、5个。In the present invention, the term "plurality" independently refers to 2, 3, 4, and 5.
除非特别说明,本发明所描述的结构式意在包括所有的同分异构形式(如对映异构,非对映异构和几何异构体(或构象异构体)):例如含有不对称中心的R、S构型,双键的(Z)、(E)异构体等。因此,本发明化合物的单个立体化学异构体或其对映异构体、非对映异构体或几何异构体(或构象异构体)的混合物都属于本发明的范围。Unless otherwise specified, the structural formulas described in the present invention are intended to include all isomeric forms (such as enantiomers, diastereomers and geometric isomers (or conformational isomers)): for example, containing asymmetric The R and S configurations of the center, the (Z) and (E) isomers of the double bond, etc. Therefore, individual stereochemical isomers of the compounds of the present invention or mixtures of their enantiomers, diastereomers or geometric isomers (or conformational isomers) are within the scope of the present invention.
如本文所用,术语“互变异构体”表示具有不同能量的结构同分异构体可以超过低能垒,从而互相转化。比如,质子互变异构体(即质子移变)包括通过质子迁移进行互变,如1H-吲唑与2H-吲唑。化合价互变异构体包括通过一些成键电子重组而进行互变。As used herein, the term "tautomer" means that structural isomers with different energies can exceed a low energy barrier and thereby convert into each other. For example, proton tautomers (i.e., proton transfer) include interconversion through proton migration, such as 1H-indazole and 2H-indazole. Valence tautomers involve interconversions through the recombination of some of the bonding electrons.
如本文所用,术语“溶剂合物”是指本发明化合物与溶剂分子配位形成特定比例的配合物。As used herein, the term "solvate" refers to a complex in which a compound of the invention is coordinated with solvent molecules to form a complex in a specified proportion.
活性成分active ingredients
如本文所用,“本发明化合物”指式I、式I’或式II所示的化合物,并且还包括及式(A)化合物的药学上可接受的盐、立体异构体、溶剂化物或前药。As used herein, "compounds of the present invention" refers to compounds represented by Formula I, Formula I' or Formula II, and also includes pharmaceutically acceptable salts, stereoisomers, solvates or precursors of the compounds of Formula (A). medicine.
本发明中的化合物可能形成的盐也是属于本发明的范围。除非另有说明,本发明中的化合物被理解为包括其盐类。在此使用的术语“盐”,指用无机或有机酸和碱形成酸式或碱式的盐。此外,当本发明中的化合物含一个碱性片段时,它包括但不限于吡 啶或咪唑,含一个酸性片段时,包括但不限于羧酸,可能形成的两性离子(“内盐”)包含在术语“盐”的范围内。药学上可接受的(即无毒,生理可接受的)盐是首选,虽然其他盐类也有用,例如可以用在制备过程中的分离或纯化步骤。本发明的化合物可能形成盐,例如,化合物I与一定量如等当量的酸或碱反应,在介质中盐析出来,或在水溶液中冷冻干燥得来。The possible salts formed by the compounds in the present invention also belong to the scope of the present invention. Unless otherwise stated, compounds in the present invention are understood to include salts thereof. The term "salt" as used herein refers to an acidic or basic salt formed from an inorganic or organic acid and a base. In addition, when the compound of the present invention contains a basic moiety, it includes but is not limited to pyridine. Zwitterions ("internal salts") that may be formed when containing an acidic moiety, including but not limited to carboxylic acids, are included within the scope of the term "salt". Pharmaceutically acceptable (i.e., nontoxic, physiologically acceptable) salts are preferred, although other salts are also useful, for example, in isolation or purification steps during preparation. The compounds of the present invention may form salts, for example, compound I can be obtained by reacting with a certain amount of, for example, an equivalent amount of acid or base, salting out in a medium, or by freeze-drying in an aqueous solution.
本发明中的化合物含有的碱性片段,包括但不限于胺或吡啶或咪唑环,可能会和有机或无机酸形成盐。可以成盐的典型的酸包括醋酸盐(如用醋酸或三卤代醋酸,如三氟乙酸)、己二酸盐、藻朊酸盐、抗坏血酸盐、天冬氨酸盐、苯甲酸盐、苯磺酸盐、硫酸氢盐、硼酸盐、丁酸盐、柠檬酸盐、樟脑盐、樟脑磺酸盐、环戊烷丙酸盐、二甘醇酸盐、十二烷基硫酸盐、乙烷磺酸盐、延胡索酸盐、葡庚糖酸盐、甘油磷酸盐、半硫酸盐、庚酸盐、己酸盐、盐酸盐、氢溴酸盐、氢碘酸盐、羟基乙磺酸盐(如,2-羟基乙磺酸盐)、乳酸盐、马来酸盐、甲磺酸盐、萘磺酸盐(如,2-萘磺酸盐)、烟酸盐、硝酸盐、草酸盐、果胶酸盐、过硫酸盐、苯丙酸盐(如3-苯丙酸盐)、磷酸盐、苦味酸盐、新戊酸盐、丙酸盐,水杨酸盐、琥珀酸盐、硫酸盐(如与硫酸形成的)、磺酸盐、酒石酸盐、硫氰酸盐、甲苯磺酸盐如对甲苯磺酸盐、十二烷酸盐等等。The compounds of the present invention contain basic moieties, including but not limited to amines or pyridine or imidazole rings, which may form salts with organic or inorganic acids. Typical acids that can form salts include acetates (eg, with acetic acid or trihaloacetic acids, such as trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, and benzoates. , benzenesulfonate, hydrogen sulfate, borate, butyrate, citrate, camphor salt, camphor sulfonate, cyclopentane propionate, diglycolate, dodecyl sulfate, Ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, enanthate, caproate, hydrochloride, hydrobromide, hydroiodide, glycolate (e.g., 2-hydroxyethanesulfonate), lactate, maleate, methanesulfonate, naphthalenesulfonate (e.g., 2-naphthalenesulfonate), nicotinate, nitrate, oxalic acid Salt, pectate, persulfate, phenylpropionate (such as 3-phenylpropionate), phosphate, picrate, pivalate, propionate, salicylate, succinate, Sulfates (such as formed with sulfuric acid), sulfonates, tartrates, thiocyanates, toluenesulfonates such as p-toluenesulfonate, dodecanoate, etc.
本发明的某些化合物可能含有的酸性片段,包括但不限于羧酸,可能会和各种有机或无机碱形成盐。典型的碱形成的盐包括铵盐、碱金属盐如钠、锂、钾盐,碱土金属盐如钙、镁盐和有机碱形成的盐(如有机胺),如苄星、二环已基胺、海巴胺(与N,N-二(去氢枞基)乙二胺形成的盐)、N-甲基-D-葡糖胺、N-甲基-D-葡糖酰胺、叔丁基胺,以及和氨基酸如精氨酸、赖氨酸等等形成的盐。碱性含氮基团可以与卤化物季铵盐,如小分子烷基卤化物(如甲基、乙基、丙基和丁基的氯化物、溴化物及碘化物),二烷基硫酸盐(如,硫酸二甲酯、二乙酯,二丁酯和二戊酯),长链卤化物(如癸基、十二烷基、十四烷基和十四烷基的氯化物、溴化物及碘化物),芳烷基卤化物(如苄基和苯基溴化物)等等。Certain compounds of the invention may contain acidic moieties, including but not limited to carboxylic acids, which may form salts with various organic or inorganic bases. Typical salts formed with bases include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, and salts formed with organic bases (such as organic amines), such as benzathine and dicyclohexylamine. , Hypamine (salt with N,N-bis(dehydroabidyl)ethylenediamine), N-methyl-D-glucamine, N-methyl-D-glucamide, tert-butyl Amines, and salts formed with amino acids such as arginine, lysine, etc. Basic nitrogen-containing groups can be combined with halide quaternary ammonium salts, such as small molecule alkyl halides (such as chlorides, bromides and iodides of methyl, ethyl, propyl and butyl), dialkyl sulfates (such as dimethyl sulfate, diethyl sulfate, dibutyl ester and dipentyl ester), long chain halides (such as decyl, dodecyl, tetradecyl and tetradecyl chlorides, bromides and iodide), aralkyl halides (such as benzyl and phenyl bromide), etc.
本发明中化合物的前药及溶剂合物也在涵盖的范围之内。此处术语“前药”是指一种化合物,在治疗相关疾病时,经过代谢或化学过程的化学转化而产生本发明中的化合物、盐、或溶剂合物。本发明的化合物包括溶剂合物,如水合物。Prodrugs and solvates of the compounds of the present invention are also within the scope of the invention. The term "prodrug" here refers to a compound that undergoes chemical transformation through metabolism or chemical processes to produce the compound, salt, or solvate of the present invention when treating related diseases. Compounds of the present invention include solvates, such as hydrates.
本发明中的化合物、盐或溶剂合物,可能存在的互变异构形式(例如酰胺和亚胺醚)。所有这些互变异构体都是本发明的一部分。The compounds, salts or solvates of the present invention may exist in tautomeric forms (eg amides and imine ethers). All such tautomers are part of the present invention.
所有化合物的立体异构体(例如,那些由于对各种取代可能存在的不对称碳原子),包括其对映体形式和非对映形式,都属于本发明的设想范围。本发明中的化合物独立的立体异构体可能不与其他异构体同时存在(例如,作为一个纯的或者实质上是纯的光学异构体具有特殊的活性),或者也可能是混合物,如消旋体,或与所有其他立体异构体或其中的一部分形成的混合物。本发明的手性中心有S或R两种构型,由理论与应用化学国际联合会(IUPAC)1974年建议定义。外消旋形式可通过物理方法解决, 例如分步结晶,或通过衍生为非对映异构体分离结晶,或通过手性柱色谱法分离。单个的光学异构体可通过合适的方法由外消旋体得到,包括但不限于传统的方法,例如与光学活性酸成盐后再结晶。All stereoisomers of the compounds (eg, those that may exist due to asymmetric carbon atoms for various substitutions), including their enantiomeric and diastereomeric forms, are contemplated by the present invention. The compounds of the invention may be independent stereoisomers that do not exist simultaneously with other isomers (e.g., have a specific activity as a pure or substantially pure optical isomer), or they may be mixtures, e.g. Racemates, or mixtures with all other stereoisomers or portions thereof. The chiral center of the present invention has two configurations: S or R, which are defined as recommended by the International Union of Theoretical and Applied Chemistry (IUPAC) in 1974. The racemic form can be resolved by physical means, For example, fractional crystallization, or separation and crystallization by derivatization into diastereoisomers, or separation by chiral column chromatography. Individual optical isomers can be obtained from the racemate by suitable methods, including but not limited to traditional methods, such as salt formation with an optically active acid followed by recrystallization.
本发明中的化合物,依次通过制备、分离纯化获得的该化合物其重量含量等于或大于90%,例如,等于或大于95%,等于或大于99%(“非常纯”的化合物),在正文描述列出。此处这种“非常纯”本发明的化合物也作为本发明的一部分。The weight content of the compounds in the present invention obtained by sequential preparation, separation and purification is equal to or greater than 90%, for example, equal to or greater than 95%, equal to or greater than 99% ("very pure" compounds), as described in the text List. Such "very pure" compounds of the invention are here also included as part of the invention.
本发明的化合物所有的构型异构体都在涵盖的范围之内,无论是混合物、纯的或非常纯的形式。在本发明化合物的定义包含顺式(Z)和反式(E)两种烯烃异构体,以及碳环和杂环的顺式和反式异构体。All configurational isomers of the compounds of the invention are included within the scope, whether in mixtures, pure or very pure form. The definition of compounds in the present invention includes both cis (Z) and trans (E) olefin isomers, as well as cis and trans isomers of carbocyclic and heterocyclic rings.
在整个说明书中,基团和取代基可以被选择以提供稳定的片段和化合物。Throughout the specification, groups and substituents may be selected to provide stable fragments and compounds.
特定官能团和化学术语定义都详细介绍如下。对本发明来说,化学元素与Periodic Table of the Elements,CAS version,Handbook of Chemistry and Physics,75th Ed.中定义的一致。特定官能团的定义也在其中描述。此外,有机化学的基本原则以及特定官能团和反应性在“Organic Chemistry”,Thomas Sorrell,University Science Books,Sausalito:1999,也有说明,其全部内容纳入参考文献之列。Definitions of specific functional groups and chemical terms are detailed below. For the purposes of this invention, chemical elements are as defined in the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Definitions of specific functional groups are also described therein. In addition, the basic principles of organic chemistry and specific functional groups and reactivities are described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausalito: 1999, the entire contents of which are incorporated by reference.
本发明的某些化合物可能存在于特定的几何或立体异构体形式。本发明涵盖所有的化合物,包括其顺式和反式异构体、R和S对映异构体、非对映体、(D)型异构体、(L)型异构体、外消旋混合物和其它混合物。另外不对称碳原子可表示取代基,如烷基。所有异构体以及它们的混合物,都包涵在本发明中。Certain compounds of the present invention may exist in specific geometric or stereoisomeric forms. The present invention encompasses all compounds, including their cis and trans isomers, R and S enantiomers, diastereomers, (D) isomers, (L) isomers, elimination Spin mixtures and other mixtures. In addition, asymmetric carbon atoms can represent substituents, such as alkyl groups. All isomers, as well as mixtures thereof, are included in the present invention.
按照本发明,同分异构体的混合物含有异构体的比率可以是多样的。例如,在只有两个异构体的混合物可以有以下组合:50:50,60:40,70:30,80:20,90:10,95:5,96:4,97:3,98:2,99:1,或100:0,异构体的所有比率都在本发明范围之内。本专业内一般技术人员容易理解的类似的比率,及为更复杂的异构体的混合物的比率也在本发明范围之内。According to the present invention, the mixture of isomers may contain the isomers in various ratios. For example, a mixture of only two isomers can have the following combinations: 50:50, 60:40, 70:30, 80:20, 90:10, 95:5, 96:4, 97:3, 98: All ratios of isomers 2, 99:1, or 100:0 are within the scope of the invention. Similar ratios, as well as ratios for more complex mixtures of isomers that are readily understood by those of ordinary skill in the art, are also within the scope of the present invention.
本发明还包括同位素标记的化合物,等同于原始化合物在此公开。不过实际上对一个或更多的原子被与其原子量或质量序数不同的原子取代通常会出现。可以列为本发明的化合物同位素的例子包括氢、碳、氮、氧、磷、硫、氟和氯同位素,分别如2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F和36Cl。本发明中的化合物,或对映体,非对映体,异构体,或药学上可接受的盐或溶剂化物,其中含有上述化合物的同位素或其他同位素原子都在本发明的范围之内。本发明中某些同位素标记化合物,例如3H和14C的放射性同位素也在其中,在药物和底物的组织分布实验中是有用的。氚,即3H和碳-14,即14C,它们的制备和检测比较容易。是同位素中的首选。此外,较重同位素取代如氘,即2H,由于其很好的代谢稳定性在某些疗法中有优势,例如在体内增加半衰期或减少用量,因此,在某些情况下可以优先考虑。同位素标记的化合物可以用一般的方法,通过用易得的同位素标记试剂替换为非同位素的试剂,用 批露在示例中的方案可以制备。The present invention also includes isotopically labeled compounds that are equivalent to the original compounds disclosed herein. In practice, however, it often occurs that one or more atoms are replaced by atoms with a different atomic weight or mass number. Examples of isotopes of compounds that may be included in the present invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes such as 2 H, 3 H, 13 C, 11 C, 14 C, 15 N, 18 O respectively. , 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl. The compounds of the present invention, or enantiomers, diastereomers, isomers, or pharmaceutically acceptable salts or solvates, which contain isotopes or other isotope atoms of the above compounds are within the scope of the present invention. Certain isotopically labeled compounds of the present invention, such as radioactive isotopes of 3 H and 14 C, are also included and are useful in tissue distribution experiments of drugs and substrates. Tritium, or 3H , and carbon-14, or 14C , are relatively easy to prepare and detect. It is the first choice among isotopes. In addition, heavier isotope substitutions such as deuterium, i.e. 2H , may have advantages in certain therapies due to their good metabolic stability, such as increased half-life in the body or reduced dosage, and therefore may be prioritized in certain circumstances. Isotopically labeled compounds can be labeled in the usual manner by replacing readily available isotopically labeled reagents with non-isotopically labeled reagents, using The protocols disclosed in the examples can be prepared.
如果要设计一个本发明的化合物特定的对映体的合成,它可以不对称合成制备,或用手性辅剂衍生化,将所产生的非对映混合物分离,再除去手性辅剂而得到纯的对映体。另外,如果分子中含有一个碱性官能团,如氨基酸,或酸性官能团,如羧基,可以用合适的光学活性的酸或碱的与之形成非对映异构体盐,再通过分离结晶或色谱等常规手段分离,然后就得到了纯的对映体。If one wants to design a synthesis of a specific enantiomer of the compound of the present invention, it can be prepared by asymmetric synthesis, or derivatized with a chiral auxiliary, and the resulting diastereomeric mixture is separated and then the chiral auxiliary is removed. Pure enantiomer. In addition, if the molecule contains a basic functional group, such as an amino acid, or an acidic functional group, such as a carboxyl group, a suitable optically active acid or base can be used to form a diastereomeric salt with it, and then through separation, crystallization or chromatography, etc. After separation by conventional means, the pure enantiomers are obtained.
如本文所述,本发明中的化合物可与任何数量取代基或官能团取而扩大其包涵范围。通常,术语“取代”不论在术语“可选”前面或后面出现,在本发明配方中包括取代基的通式,是指用指定结构取代基,代替氢自由基。当特定结构中的多个在位置被多个特定的取代基取代时,取代基每一个位置可以是相同或不同。本文中所使用的术语“取代”包括所有允许有机化合物取代。从广义上讲,允许的取代基包括非环状的、环状的、支链的非支链的、碳环的和杂环的,芳环的和非芳环的有机化合物。在本发明中,如杂原子氮可以有氢取代基或任何允许的上文所述的有机化合物来补充其价态。此外,本发明是无意以任何方式限制允许取代有机化合物。本发明认为取代基和可变基团的组合在以稳定化合物形式在疾病的治疗上是很好的。此处术语“稳定”是指具有稳定的化合物,在足够长的时间内检测足以维持化合物结构的完整性,最好是在足够长的时间内都在效,本文在此用于上述目的。As described herein, the compounds of the present invention may be provided with any number of substituents or functional groups to broaden their encompassing scope. In general, whether the term "substituted" appears before or after the term "optional", the general formula of the substituent included in the formulation of the present invention means that the substituent of the specified structure is used in place of the hydrogen radical. When multiple positions in a specific structure are substituted by multiple specific substituents, the substituents may be the same or different at each position. The term "substitution" as used herein includes all permissible substitutions of organic compounds. Broadly speaking, permissible substituents include acyclic, cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic organic compounds. In the present invention, heteroatoms such as nitrogen may have hydrogen substituents or any of the permissible organic compounds described above to supplement their valence. Furthermore, this invention is not intended to be limited in any way to the permitted substituted organic compounds. The present invention considers that combinations of substituents and variable groups are excellent in the treatment of diseases in the form of stable compounds. The term "stable" as used herein refers to a compound that is stable, detectable over a long enough period of time to maintain the structural integrity of the compound, and preferably effective over a long enough period of time, and is used herein for the above purposes.
本申请所涉及的化合物及其药学可接受的盐的代谢产物,以及可以在体内转变为本申请所涉及的化合物及其药学可接受的盐的结构的前药,也包含在本申请的权利要求中。The metabolites of the compounds involved in this application and their pharmaceutically acceptable salts, as well as the prodrugs that can be converted into the structures of the compounds involved in this application and their pharmaceutically acceptable salts in vivo, are also included in the claims of this application. middle.
化合物的制备方法Preparation methods of compounds
以下方案和实例中描述了制备式I的化合物的方法。原料和中间体从商业来源购买,由已知步骤制备,或以其他方式说明。在某些情况下,可以改变执行反应方案的步骤的顺序,以促进反应或避免不需要的副反应产物。Methods for preparing compounds of formula I are described in the following schemes and examples. Starting materials and intermediates were purchased from commercial sources, prepared by known procedures, or otherwise described. In some cases, the order of steps in performing a reaction scheme can be changed to facilitate the reaction or avoid undesired side reaction products.
通常,在制备流程中,各反应通常在惰性溶剂中,在室温至回流温度(如0℃~150℃,优选10℃~100℃)下进行。反应时间通常为0.1小时-60小时,较佳地为0.5-48小时。Generally, in the preparation process, each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (such as 0°C to 150°C, preferably 10°C to 100°C). The reaction time is usually 0.1 hour to 60 hours, preferably 0.5 to 48 hours.
优选地,本发明化合物可用如下步骤制得Preferably, the compounds of the present invention can be prepared by the following steps
合成路线一:
Synthesis route one:
以2,4-二氯噻吩并嘧啶为起始原料,通过与吲哚类衍生物在三氯化铝作用下发生亲核取代反应或通过与硼酸类衍生物发生Suzuki偶联反应得到相应的中间体化合物;所得中间体通过在有机碱的作用下与相应的胺发生取代反应得到相应的产物,最终通过将保护基脱除得到I系列目标化合物;Using 2,4-dichlorothienopyrimidine as the starting material, the corresponding intermediate is obtained through a nucleophilic substitution reaction with an indole derivative under the action of aluminum trichloride or a Suzuki coupling reaction with a boronic acid derivative. The resulting intermediate is a substitution reaction with the corresponding amine under the action of an organic base to obtain the corresponding product, and finally the I series target compounds are obtained by removing the protecting group;
合成路线二:
Synthesis route two:
以7-溴-2-氯噻吩并嘧啶为起始原料,通过在有机碱的作用下与相应的胺发生取代反应得到相应的中间体化合物;所得中间体通过与硼酸类衍生物发生Suzuki偶联反应得到相应的产物,最终通过将保护基脱除得到II系列目标化合物。Using 7-bromo-2-chlorothienopyrimidine as the starting material, the corresponding intermediate compound is obtained through a substitution reaction with the corresponding amine under the action of an organic base; the obtained intermediate is subjected to Suzuki coupling with a boronic acid derivative The reaction obtains the corresponding product, and finally the II series target compounds are obtained by removing the protecting group.
药物组合物和施用方法 Pharmaceutical compositions and methods of administration
由于本发明化合物具有优异的CDK激酶的抑制活性,因此本发明化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,以及含有本发明化合物为主要活性成分的药物组合物可用于预防和/或治疗(稳定、减轻或治愈)CDK激酶相关疾病例如,肿瘤(如结直肠癌、乳腺癌、肺癌、卵巢癌或胃癌)。Since the compound of the present invention has excellent CDK kinase inhibitory activity, the compound of the present invention or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug, as well as a pharmaceutical composition containing the compound of the present invention as the main active ingredient It can be used to prevent and/or treat (stabilize, alleviate or cure) CDK kinase related diseases such as tumors (such as colorectal cancer, breast cancer, lung cancer, ovarian cancer or gastric cancer).
本发明的药物组合物包含安全有效量范围内的本发明化合物及药学上可以接受的赋形剂或载体。其中“安全有效量”指的是:化合物的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg本发明化合物/剂,更佳地,含有10-200mg本发明化合物/剂。较佳地,所述的“一剂”为一个胶囊或药片。The pharmaceutical composition of the present invention contains a compound of the present invention and a pharmaceutically acceptable excipient or carrier within a safe and effective amount. The “safe and effective dose” refers to the amount of compound that is sufficient to significantly improve the condition without causing serious side effects. Usually, the pharmaceutical composition contains 1-2000 mg of the compound of the present invention/dose, more preferably, it contains 10-200 mg of the compound of the present invention/dose. Preferably, the "dose" is a capsule or tablet.
“药学上可接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明化合物以及它们之间相互掺和,而不明显降低化合物的药效。药学上可以接受的载体部分例子有纤维素及其衍生物(如羧甲基纤维素钠、乙基纤维素钠、纤维素乙酸酯等)、明胶、滑石、固体润滑剂(如硬脂酸、硬脂酸镁)、硫酸钙、植物油(如豆油、芝麻油、花生油、橄榄油等)、多元醇(如丙二醇、甘油、甘露醇、山梨醇等)、乳化剂(如吐温)、润湿剂(如十二烷基硫酸钠)、着色剂、调味剂、稳定剂、抗氧化剂、防腐剂、无热原水等。"Pharmaceutically acceptable carrier" refers to one or more compatible solid or liquid filler or gel substances that are suitable for human use and must be of sufficient purity and low enough toxicity. "Compatibility" here means that the components of the composition can be blended with the compounds of the present invention and with each other without significantly reducing the efficacy of the compounds. Examples of pharmaceutically acceptable carriers include cellulose and its derivatives (such as sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (such as stearic acid , magnesium stearate), calcium sulfate, vegetable oils (such as soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (such as propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (such as Tween ), wetting agents (such as sodium lauryl sulfate), colorants, flavorings, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
本发明化合物或药物组合物的施用方式没有特别限制,代表性的施用方式包括(但并不限于):口服、肠胃外(静脉内、肌肉内或皮下)。The administration mode of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative administration modes include (but are not limited to): oral, parenteral (intravenous, intramuscular or subcutaneous).
用于口服给药的固体剂型包括胶囊剂、片剂、丸剂、散剂和颗粒剂。在这些固体剂型中,活性化合物与至少一种常规惰性赋形剂(或载体)混合,如柠檬酸钠或磷酸二钙,或与下述成分混合:(a)填料或增容剂,例如,淀粉、乳糖、蔗糖、葡萄糖、甘露醇和硅酸;(b)粘合剂,例如,羟甲基纤维素、藻酸盐、明胶、聚乙烯基吡咯烷酮、蔗糖和阿拉伯胶;(c)保湿剂,例如,甘油;(d)崩解剂,例如,琼脂、碳酸钙、马铃薯淀粉或木薯淀粉、藻酸、某些复合硅酸盐、和碳酸钠;(e)缓溶剂,例如石蜡;(f)吸收加速剂,例如,季胺化合物;(g)润湿剂,例如鲸蜡醇和单硬脂酸甘油酯;(h)吸附剂,例如,高岭土;和(i)润滑剂,例如,滑石、硬脂酸钙、硬脂酸镁、固体聚乙二醇、十二烷基硫酸钠,或其混合物。胶囊剂、片剂和丸剂中,剂型也可包含缓冲剂。Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or compatibilizers, for example, Starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) Binders, for example, hydroxymethylcellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and gum arabic; (c) Humectants, For example, glycerol; (d) disintegrants, such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) retarder, such as paraffin; (f) Absorption accelerators, such as quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glyceryl monostearate; (h) adsorbents, such as kaolin; and (i) lubricants, such as talc, hard Calcium fatty acid, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage form may also contain buffering agents.
固体剂型如片剂、糖丸、胶囊剂、丸剂和颗粒剂可采用包衣和壳材制备,如肠衣和其它本领域公知的材料。它们可包含不透明剂,并且,这种组合物中活性化合物或化合物的释放可以延迟的方式在消化道内的某一部分中释放。可采用的包埋组分的实例是聚合物质和蜡类物质。必要时,活性化合物也可与上述赋形剂中的一种或多种形成微胶囊形式。Solid dosage forms such as tablets, dragees, capsules, pills and granules may be prepared using coatings and shell materials such as enteric casings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be released in a delayed manner in a certain part of the digestive tract. Examples of embedding components that can be used are polymeric substances and waxy substances. If necessary, the active compounds can also be in microencapsulated form with one or more of the above-mentioned excipients.
用于口服给药的液体剂型包括药学上可接受的乳液、溶液、悬浮液、糖浆或酊剂。除了活性化合物外,液体剂型可包含本领域中常规采用的惰性稀释剂,如水或其它溶 剂,增溶剂和乳化剂,例知,乙醇、异丙醇、碳酸乙酯、乙酸乙酯、丙二醇、1,3-丁二醇、二甲基甲酰胺以及油,特别是棉籽油、花生油、玉米胚油、橄榄油、蓖麻油和芝麻油或这些物质的混合物等。Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. Liquid dosage forms may contain, in addition to the active compound, inert diluents conventionally employed in the art, such as water or other solvents. Agents, solubilizers and emulsifiers, for example, ethanol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1,3-butanediol, dimethylformamide and oils, especially cottonseed oil, peanut oil, Corn germ oil, olive oil, castor oil and sesame oil or mixtures of these substances, etc.
除了这些惰性稀释剂外,组合物也可包含助剂,如润湿剂、乳化剂和悬浮剂、甜味剂、矫味剂和香料。Besides these inert diluents, the compositions may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring and perfuming agents.
除了活性化合物外,悬浮液可包含悬浮剂,例如,乙氧基化异十八烷醇、聚氧乙烯山梨醇和脱水山梨醇酯、微晶纤维素、甲醇铝和琼脂或这些物质的混合物等。Suspensions may contain, in addition to the active compound, suspending agents, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar or mixtures of these substances and the like.
用于肠胃外注射的组合物可包含生理上可接受的无菌含水或无水溶液、分散液、悬浮液或乳液,和用于重新溶解成无菌的可注射溶液或分散液的无菌粉末。适宜的含水和非水载体、稀释剂、溶剂或赋形剂包括水、乙醇、多元醇及其适宜的混合物。Compositions for parenteral injection may contain physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and non-aqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
本发明化合物可以单独给药,或者与其他药学上可接受的化合物(例如CDK抑制剂)联合给药。The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds (eg, CDK inhibitors).
联合给药时,所述药物组合物还包括与一种或多种(2种,3种,4种,或更多种)其他药学上可接受的化合物(例如CDK抑制剂)。该其他药学上可接受的化合物中的一种或多种(2种,3种,4种,或更多种)可与本发明的化合物同时、分开或顺序地用于预防和/或治疗CDK激酶的活性或表达量相关的疾病。When administered in combination, the pharmaceutical composition also includes one or more (2, 3, 4, or more) other pharmaceutically acceptable compounds (eg, CDK inhibitors). One or more (2, 3, 4, or more) of the other pharmaceutically acceptable compounds may be used simultaneously, separately, or sequentially with the compound of the invention to prevent and/or treat CDK Diseases related to kinase activity or expression.
使用药物组合物时,是将安全有效量的本发明化合物适用于需要治疗的哺乳动物(如人),其中施用时剂量为药学上认为的有效给药剂量,对于60kg体重的人而言,日给药剂量通常为1~2000mg,优选20~500mg。当然,具体剂量还应考虑给药途径、病人健康状况等因素,这些都是熟练医师技能范围之内的。When using the pharmaceutical composition, a safe and effective amount of the compound of the present invention is applied to a mammal (such as a human) in need of treatment, and the dosage when administered is a pharmaceutically effective dosage. For a person weighing 60 kg, the daily dose is The dosage is usually 1 to 2000 mg, preferably 20 to 500 mg. Of course, the specific dosage should also take into account factors such as the route of administration and the patient's health condition, which are all within the skill of a skilled physician.
本发明的主要优点在于:The main advantages of the present invention are:
1.本发明化合物结构新颖且具有优异的CDK激酶抑制活性,尤其是CDK7或CDK2;1. The compound of the present invention has a novel structure and has excellent CDK kinase inhibitory activity, especially CDK7 or CDK2;
2.本发明的化合物可以作为CDK激酶抑制剂,尤其是作为CDK7或CDK2的高选择性抑制剂。2. The compounds of the present invention can be used as CDK kinase inhibitors, especially as highly selective inhibitors of CDK7 or CDK2.
3.本发明化合物具有较好的药代动力学和药效。3. The compound of the present invention has good pharmacokinetics and efficacy.
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。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,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。The present invention will be further described below in conjunction with specific embodiments. It should be understood that these examples are only used to illustrate the invention and are not intended to limit the scope of the invention. Experimental methods without specifying specific conditions in the following examples usually follow conventional conditions or conditions recommended by the manufacturer. Unless otherwise stated, percentages and parts are by weight.
实施例Example
实施例1、化合物I-1的合成与表征
Example 1. Synthesis and characterization of compound I-1
步骤1:化合物I-1-2的合成Step 1: Synthesis of compound I-1-2
向I-1-1(3g,14.6mmol,3eq)的1,2-二氯乙烷溶液中(15mL)加入无水三氯化铝(1g,7.5mmol,1.5eq),氮气置换三次后将反应置于85℃油浴中搅拌0.5小时,然后再向上述反应液中加入7-溴吲哚(1g,5.1mmol,1eq)的1,2-二氯乙烷溶液(10mL),反应继续搅拌9.5小时。待反应完全,反应液冷却至室温,然后向其中加入50mL2-甲基四氢呋喃和30mL水,分离出有机相,用30mL的饱和食盐水洗涤一次,然后有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色粗品。向粗品中加入30mL甲基叔丁基醚进行打浆,经过滤得到淡黄色固体I-1-2(1.5g)。LCMS(M+H):364。Anhydrous aluminum trichloride (1g, 7.5mmol, 1.5eq) was added to the 1,2-dichloroethane solution (15mL) of I-1-1 (3g, 14.6mmol, 3eq), and nitrogen was replaced three times. The reaction was stirred in an 85°C oil bath for 0.5 hours, and then a solution of 7-bromoindole (1g, 5.1mmol, 1eq) in 1,2-dichloroethane (10mL) was added to the above reaction solution, and the reaction was continued to stir. 9.5 hours. When the reaction is complete, cool the reaction solution to room temperature, then add 50 mL of 2-methyltetrahydrofuran and 30 mL of water, separate the organic phase, wash it once with 30 mL of saturated brine, and then dry the organic phase over anhydrous sodium sulfate, filter, and reduce. Concentrate under pressure to obtain a pale yellow crude product. Add 30 mL of methyl tert-butyl ether to the crude product for pulping, and obtain light yellow solid I-1-2 (1.5 g) after filtration. LCMS(M+H):364.
步骤2:化合物I-1-3的合成Step 2: Synthesis of compound I-1-3
向封管中加入I-1-2(0.1g,274μmol,1eq)和1mL的N-甲基吡咯烷酮,再分别加入(S)-3-氨基哌啶-1-羧酸叔丁酯(165mg,0.83mmol,3eq)和N,N-二异丙基乙胺(180mg,1.4mmol,5eq),然后置于145℃油浴中搅拌5小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用30mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化得到淡黄色油状物I-1-3(150mg)。LCMS(M+H):528。Add I-1-2 (0.1g, 274μmol, 1eq) and 1mL of N-methylpyrrolidone to the sealed tube, and then add (S)-3-aminopiperidine-1-carboxylic acid tert-butyl ester (165mg, 0.83mmol, 3eq) and N,N-diisopropylethylamine (180mg, 1.4mmol, 5eq), then placed in a 145°C oil bath and stirred for 5 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 30mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography to obtain light yellow oily substance I-1-3 (150 mg). LCMS(M+H):528.
步骤3:化合物I-1-4的合成Step 3: Synthesis of compound I-1-4
向反应管中加入I-1-3(150mg,284μmol,1eq)、Pd(dppf)Cl2(21mg,28μmol,0.1eq)、1mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(66mg,0.85mmol,3eq)和三乙胺(150mg,1.5mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到淡黄色油状物I-1-4(110mg)。LCMS(M+H):526。 Add I-1-3 (150 mg, 284 μmol, 1 eq), Pd(dppf)Cl 2 (21 mg, 28 μmol, 0.1 eq), and 1 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (66 mg, 0.85 mmol, 3 eq) and triethylamine (150 mg, 1.5 mmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain light yellow oily substance I-1-4 (110 mg). LCMS(M+H):526.
步骤4:化合物I-1的合成Step 4: Synthesis of Compound I-1
向化合物I-1-4(110mg,210μmol,1eq)的二氯甲烷溶液中(5mL)加入三氟乙酸(1mL),然后于室温条件下搅拌5小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(FA)分离纯化得到淡黄色固体I-1(25mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.40(s,1H),8.14(d,J=8.0Hz,1H),7.90–7.84(m,2H),7.34(dd,J=12.0,8.0Hz,1H),7.10(td,J=8.0,2.4Hz,1H),6.94(d,J=4.0Hz,1H),4.00-3.95(m,1H),3.45(dd,J=12.0,4.0Hz,1H),3.47-3.31(m,1H),3.12–2.99(m,2H),2.06-2.03(m,2H),1.89(dd,J=12.0,4.0Hz,6H),1.86–1.76(m,1H),1.67-1.60(m,1H);LCMS(M+H):426。Trifluoroacetic acid (1 mL) was added to a solution (5 mL) of compound I-1-4 (110 mg, 210 μmol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain light yellow solid I-1 (25 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.40 (s, 1H), 8.14 (d, J = 8.0Hz, 1H), 7.90–7.84 (m, 2H), 7.34 (dd, J = 12.0, 8.0Hz, 1H),7.10(td,J=8.0,2.4Hz,1H),6.94(d,J=4.0Hz,1H),4.00-3.95(m,1H),3.45(dd,J=12.0,4.0Hz,1H ),3.47-3.31(m,1H),3.12–2.99(m,2H),2.06-2.03(m,2H),1.89(dd,J=12.0,4.0Hz,6H),1.86–1.76(m,1H) ),1.67-1.60(m,1H); LCMS(M+H):426.
实施例2、化合物I-2的合成与表征
Example 2. Synthesis and characterization of compound I-2
步骤1:化合物I-2-1的合成Step 1: Synthesis of compound I-2-1
向反应管中加入I-1-3(80mg,151μmol,1eq)、Pd(dppf)Cl2(11mg,15μmol,0.1eq)、1mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二乙基氧化膦(32mg,453μmol,3eq)和三乙胺(46mg,0.75mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到淡黄色油状物I-2-1(50mg)。LCMS(M+H):554。Add I-1-3 (80 mg, 151 μmol, 1 eq), Pd(dppf)Cl 2 (11 mg, 15 μmol, 0.1 eq), and 1 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Diethylphosphine oxide (32 mg, 453 μmol, 3 eq) and triethylamine (46 mg, 0.75 mmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain light yellow oily substance I-2-1 (50 mg). LCMS(M+H):554.
步骤2:化合物I-2的合成Step 2: Synthesis of Compound I-2
向化合物I-2-1(30mg,54μmol,1eq)的二氯甲烷溶液中(5mL)加入三氟乙酸(1mL),然后于室温条件下搅拌5小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到淡黄色固体I-2(10mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.36–8.27(m,2H),8.14(d,J=4.0Hz,1H),7.45(dd,J=12.0,8.0Hz,1H),7.31(td,J=8.0,2.4Hz,1H),7.10(d,J=4.0Hz,1H),4.27(s,1H),3.56(dd,J=12.0,4.0Hz,1H),3.42–3.33(m,1H),3.20–3.04(m,2H),2.32–2.04(m,6H),1.97–1.83(m,1H),1.81–1.69(m,1H),1.17-1.04(m,6H);LCMS(M+H):426。Trifluoroacetic acid (1 mL) was added to a solution (5 mL) of compound I-2-1 (30 mg, 54 μmol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-2 (10 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.36–8.27 (m, 2H), 8.14 (d, J = 4.0Hz, 1H), 7.45 (dd, J = 12.0, 8.0Hz, 1H), 7.31 (td, J=8.0,2.4Hz,1H),7.10(d,J=4.0Hz,1H),4.27(s,1H),3.56(dd,J=12.0,4.0Hz,1H),3.42–3.33(m,1H ),3.20–3.04(m,2H),2.32–2.04(m,6H),1.97–1.83(m,1H),1.81–1.69(m,1H),1.17-1.04(m,6H); LCMS(M +H):426.
实施例3、化合物I-3的合成与表征
Example 3. Synthesis and characterization of compound I-3
步骤1:化合物I-3-1的合成Step 1: Synthesis of compound I-3-1
向反应管中依次加入I-1-3(150mg,284μmol,1eq)、CuI(110mg,0.56mmol,2eq)、甲基亚磺酸钠(150mg,1.4mmol,5eq)和2mL的二甲基亚砜,氮气置换三次后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到淡黄色油状物I-3-1(60mg)。LCMS(M+H):528。To the reaction tube, add I-1-3 (150 mg, 284 μmol, 1 eq), CuI (110 mg, 0.56 mmol, 2 eq), sodium methylsulfinate (150 mg, 1.4 mmol, 5 eq) and 2 mL of dimethylsulfinate in sequence. sulfone, and after three replacements with nitrogen, the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain light yellow oily substance I-3-1 (60 mg). LCMS(M+H):528.
步骤2:化合物I-3的合成Step 2: Synthesis of Compound I-3
向化合物I-3-1(30mg,57μmol,1eq)的二氯甲烷溶液中(5mL)加入三氟乙酸(1mL),然后于室温条件下搅拌5小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(FA)分离纯化得到淡黄色固体I-3(15mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.48(d,J=8.0Hz,1H),8.19(s,1H),8.16(d,J=4.0Hz,1H),7.73(d,J=8.0Hz,1H),7.36(t,J=8.0Hz,1H),7.21(d,J=4.0Hz,1H),4.22(s,1H),3.57(dd,J=12.0,4.0Hz,1H),3.40-3.36(m,1H),3.28(s,3H),3.15-3.08(m,2H),2.28–2.17(m,1H),2.14-2.12(m,1H),1.94-1.90(m,1H),1.82-1.77(m,1H);LCMS(M+H):428。Trifluoroacetic acid (1 mL) was added to a solution (5 mL) of compound I-3-1 (30 mg, 57 μmol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain light yellow solid I-3 (15 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.48 (d, J = 8.0Hz, 1H), 8.19 (s, 1H), 8.16 (d, J = 4.0Hz, 1H), 7.73 (d, J = 8.0Hz ,1H),7.36(t,J=8.0Hz,1H),7.21(d,J=4.0Hz,1H),4.22(s,1H),3.57(dd,J=12.0,4.0Hz,1H),3.40 -3.36(m,1H),3.28(s,3H),3.15-3.08(m,2H),2.28–2.17(m,1H),2.14-2.12(m,1H),1.94-1.90(m,1H) ,1.82-1.77(m,1H);LCMS(M+H):428.
实施例4、化合物I-4的合成与表征
Example 4. Synthesis and characterization of compound I-4
步骤1:化合物I-4-1的合成Step 1: Synthesis of compound I-4-1
向I-1-1(1g,5mmol,3eq)的1,2-二氯乙烷溶液中(10mL)加入无水三氯化铝(0.34g,7.5mmol,2.5eq),氮气置换三次后将反应置于85℃油浴中搅拌0.5小时,然 后再向上述反应液中加入吲哚(200mg,1.7mmol,1eq)的1,2-二氯乙烷溶液(5mL),反应继续搅拌9.5小时。待反应完全,反应液冷却至室温,然后向其中加入30mL2-甲基四氢呋喃和20mL水,分离出有机相,用30mL的饱和食盐水洗涤一次,然后有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色粗品。向粗品中加入15mL甲基叔丁基醚进行打浆,经过滤得到淡黄色固体I-4-1(100mg)。LCMS(M+H):286。Anhydrous aluminum trichloride (0.34g, 7.5mmol, 2.5eq) was added to the 1,2-dichloroethane solution (10mL) of I-1-1 (1g, 5mmol, 3eq), and the nitrogen was replaced three times. The reaction was stirred in an 85°C oil bath for 0.5 hours, then Then, a solution (5 mL) of indole (200 mg, 1.7 mmol, 1 eq) in 1,2-dichloroethane was added to the above reaction solution, and the reaction was continued to stir for 9.5 hours. When the reaction is complete, cool the reaction solution to room temperature, then add 30 mL of 2-methyltetrahydrofuran and 20 mL of water to it, separate the organic phase, wash it once with 30 mL of saturated brine, and then dry the organic phase over anhydrous sodium sulfate, filter, and reduce. Concentrate under pressure to obtain a pale yellow crude product. 15 mL of methyl tert-butyl ether was added to the crude product for pulping, and light yellow solid I-4-1 (100 mg) was obtained after filtration. LCMS(M+H):286.
步骤2:化合物I-4-2的合成Step 2: Synthesis of compound I-4-2
向封管中加入I-4-1(100mg,350μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入3-氨基哌啶-1-羧酸叔丁酯(330mg,1.65mmol,5eq)和N,N-二异丙基乙胺(450mg,3.5mmol,10eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用50mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化得到淡黄色油状物I-4-2(150mg)。LCMS(M+H):450。Add I-4-1 (100 mg, 350 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add 3-aminopiperidine-1-carboxylic acid tert-butyl ester (330 mg, 1.65 mmol, 5 eq) respectively. ) and N,N-diisopropylethylamine (450 mg, 3.5 mmol, 10 eq), then placed in a 135°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 50mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography to obtain light yellow oily substance I-4-2 (150 mg). LCMS(M+H):450.
步骤3:化合物I-4的合成Step 3: Synthesis of Compound I-4
向化合物I-4-2(30mg,57μmol,1eq)的二氯甲烷溶液中(5mL)加入三氟乙酸(1mL),然后于室温条件下搅拌5小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(FA)分离纯化得到淡黄色固体I-4(12mg)。1H NMR(400MHz,Methanol-d4)δ=8.64(s,2H),8.42(d,J=8.0Hz,1H),7.60(dd,J=8.0,4.0Hz,1H),7.48(d,J=8.0Hz,1H),7.43-7.36(m,2H),4.69(s,1H),3.78–3.69(m,1H),3.50–3.40(m,1H),3.22–3.10(m,1H),2.43(d,J=13.3Hz,1H),2.26–2.14(m,1H),2.12–1.97(m,1H),1.94–1.81(m,1H),1.33-1.29(m,1H);LCMS(M+H):350。Trifluoroacetic acid (1 mL) was added to a solution (5 mL) of compound I-4-2 (30 mg, 57 μmol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain light yellow solid I-4 (12 mg). 1 H NMR (400MHz, Methanol-d 4 ) δ = 8.64 (s, 2H), 8.42 (d, J = 8.0Hz, 1H), 7.60 (dd, J = 8.0, 4.0Hz, 1H), 7.48 (d, J=8.0Hz,1H),7.43-7.36(m,2H),4.69(s,1H),3.78–3.69(m,1H),3.50–3.40(m,1H),3.22–3.10(m,1H) ,2.43(d,J=13.3Hz,1H),2.26–2.14(m,1H),2.12–1.97(m,1H),1.94–1.81(m,1H),1.33-1.29(m,1H); LCMS (M+H):350.
实施例5、化合物I-5的合成与表征
Example 5. Synthesis and characterization of compound I-5
步骤1:化合物I-5-1的合成Step 1: Synthesis of compound I-5-1
向I-1-1(290mg,1.4mmol,2eq)的1,2-二氯乙烷溶液中(0.5mL)加入无水三氯化铝(103mg,0.77mmol,1.1eq),氮气置换三次后将反应置于85℃油浴中搅拌0.5小时,然后再向上述反应液中加入5-氰基吲哚(100mg,0.77mmol,1eq)的1,2-二氯 乙烷溶液(0.5mL),反应继续搅拌9.5小时。待反应完全,反应液冷却至室温,然后向其中加入10mL2-甲基四氢呋喃和20mL水,分离出有机相,用20mL的饱和食盐水洗涤一次,然后有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色粗品。向粗品中加入5mL甲基叔丁基醚进行打浆,经过滤得到淡黄色固体I-5-1(100mg)。LCMS(M+H):311。Anhydrous aluminum trichloride (103 mg, 0.77 mmol, 1.1 eq) was added to a solution (0.5 mL) of I-1-1 (290 mg, 1.4 mmol, 2 eq) in 1,2-dichloroethane, and the nitrogen was replaced three times. The reaction was stirred in an 85°C oil bath for 0.5 hours, and then 1,2-dichloro of 5-cyanoindole (100 mg, 0.77 mmol, 1 eq) was added to the above reaction solution. Ethane solution (0.5 mL) and the reaction was continued to stir for 9.5 hours. When the reaction is complete, cool the reaction solution to room temperature, then add 10 mL of 2-methyltetrahydrofuran and 20 mL of water, separate the organic phase, wash it once with 20 mL of saturated brine, and then dry the organic phase over anhydrous sodium sulfate, filter, and reduce. Concentrate under pressure to obtain a pale yellow crude product. Add 5 mL of methyl tert-butyl ether to the crude product for pulping, and obtain light yellow solid I-5-1 (100 mg) after filtration. LCMS(M+H):311.
步骤2:化合物I-5-2的合成Step 2: Synthesis of compound I-5-2
向封管中加入I-5-1(100mg,322μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(S)-3-氨基哌啶-1-羧酸叔丁酯(130mg,0.65mmol,2eq)和N,N-二异丙基乙胺(120mg,0.93mmol,3eq),然后置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用50mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化得到淡黄色油状物I-5-2(150mg)。LCMS(M+H):475。Add I-5-1 (100 mg, 322 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (S)-3-aminopiperidine-1-carboxylic acid tert-butyl ester (130 mg, 0.65mmol, 2eq) and N,N-diisopropylethylamine (120mg, 0.93mmol, 3eq), and then placed in an oil bath at 130°C and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 50mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography to obtain light yellow oily substance I-5-2 (150 mg). LCMS(M+H):475.
步骤3:化合物I-5的合成Step 3: Synthesis of Compound I-5
向化合物I-5-2(80mg,57μmol,1eq)的二氯甲烷溶液中(3mL)加入三氟乙酸(0.3mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(FA)分离纯化得到淡黄色固体I-5(30mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.43(s,1H),7.83(d,J=4.0Hz,1H),7.78(d,J=8.0Hz,1H),7.58(s,1H),7.30–7.25(m,1H),6.99(d,J=8.0Hz,1H),6.96(d,J=8.0Hz,1H),3.78(s,1H),3.37(d,J=12.0Hz,1H),3.26(d,J=16.0Hz,1H),2.97(t,J=8.0Hz,1H),2.90-2.84(m,1H),2.01-1.97(m,2H),1.78-1.75(m,1H),1.61-1.55(m,1H);LCMS(M+H):375。Trifluoroacetic acid (0.3 mL) was added to a solution (3 mL) of compound I-5-2 (80 mg, 57 μmol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain light yellow solid I-5 (30 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.43 (s, 1H), 7.83 (d, J = 4.0Hz, 1H), 7.78 (d, J = 8.0Hz, 1H), 7.58 (s, 1H), 7.30 –7.25(m,1H),6.99(d,J=8.0Hz,1H),6.96(d,J=8.0Hz,1H),3.78(s,1H),3.37(d,J=12.0Hz,1H) ,3.26(d,J=16.0Hz,1H),2.97(t,J=8.0Hz,1H),2.90-2.84(m,1H),2.01-1.97(m,2H),1.78-1.75(m,1H ),1.61-1.55(m,1H); LCMS(M+H):375.
实施例6、化合物I-6的合成与表征
Example 6. Synthesis and characterization of compound I-6
步骤1:化合物I-6-2的合成Step 1: Synthesis of compound I-6-2
向三口瓶中加入乙烯基溴化镁的四氢呋喃溶液(102mL,102mmol,5eq),于-70℃条件下缓慢滴加I-6-1(5g,1.4mmol,2eq)的四氢呋喃溶液中(100mL),滴加完毕自然升温至室温,继续搅拌10小时。待反应完全,向反应液中加入400mL饱和氯化铵溶液,再用2N盐酸调节pH至2左右,用乙酸乙酯萃取(300mL*3),合并有机相,经无水硫酸钠干燥后过滤、减压浓缩得到棕黄色粗品。向粗品中加入80mL甲基叔丁基醚进行打浆,经过滤得到土黄色固体I-6-2(2.5g)。LCMS(M-H):238。Add the tetrahydrofuran solution (102mL, 102mmol, 5eq) of vinyl magnesium bromide to the three-necked flask, and slowly add the tetrahydrofuran solution (100mL) of I-6-1 (5g, 1.4mmol, 2eq) at -70°C. , after the dropwise addition was completed, the temperature was naturally raised to room temperature, and stirring was continued for 10 hours. When the reaction is complete, add 400mL saturated ammonium chloride solution to the reaction solution, then adjust the pH to about 2 with 2N hydrochloric acid, extract with ethyl acetate (300mL*3), combine the organic phases, dry over anhydrous sodium sulfate, and filter. Concentrate under reduced pressure to obtain brown crude product. 80 mL of methyl tert-butyl ether was added to the crude product for pulping, and khaki solid I-6-2 (2.5 g) was obtained after filtration. LCMS(M-H):238.
步骤2:化合物I-6-3的合成Step 2: Synthesis of compound I-6-3
向反应管中加入I-6-2(150mg,0.63mmol,2eq),二甲胺盐酸盐(420mg,6.2mmol,10eq)和N,N-二异丙基乙胺(1.6g,12mmol,20eq),再加入5mL的DMF,待搅拌溶解后向其中加入HATU(1.2g,3.3mmol,5eq),于常温下继续搅拌15小时。待反应完全,将反应液加入到100mL水中,用乙酸乙酯萃取(30mL*3),合并有机相后用50mL饱和食盐水洗涤,有机相经无水硫酸钠干燥后过滤、减压浓缩得到淡黄色油状物粗品I-6-3(200mg)。LCMS(M+H):253。Add I-6-2 (150mg, 0.63mmol, 2eq), dimethylamine hydrochloride (420mg, 6.2mmol, 10eq) and N,N-diisopropylethylamine (1.6g, 12mmol, 20eq), then add 5mL of DMF, add HATU (1.2g, 3.3mmol, 5eq) after stirring and dissolving, and continue stirring at room temperature for 15 hours. When the reaction is complete, add the reaction solution to 100mL of water, extract with ethyl acetate (30mL*3), combine the organic phases and wash with 50mL of saturated brine. The organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a light Yellow oil crude product I-6-3 (200 mg). LCMS(M+H):253.
步骤3:化合物I-6-4的合成Step 3: Synthesis of compound I-6-4
向I-1-1(300mg,1.5mmol,2eq)的1,2-二氯乙烷溶液中(6mL)加入无水三氯化铝(130mg,0.98mmol,1.2eq),氮气置换三次后将反应置于85℃油浴中搅拌0.5小时,然后再向上述反应液中加入I-6-3(200mg,0.79mmol,1eq)的1,2-二氯乙烷溶液(2mL),反应继续搅拌9.5小时。待反应完全,反应液冷却至室温,然后向其中加入30mL的2-甲基四氢呋喃和30mL水,分离出有机相,用20mL的饱和食盐水洗涤一次,然后有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色粗品。向粗品中加入5mL甲基叔丁基醚进行打浆,经过滤得到淡黄色固体I-6-4(60mg)。LCMS(M+H):421。Anhydrous aluminum trichloride (130 mg, 0.98 mmol, 1.2 eq) was added to a solution (6 mL) of I-1-1 (300 mg, 1.5 mmol, 2 eq) in 1,2-dichloroethane, and the nitrogen was replaced three times. The reaction was stirred in an 85°C oil bath for 0.5 hours, and then a 1,2-dichloroethane solution (2 mL) of I-6-3 (200 mg, 0.79 mmol, 1 eq) was added to the above reaction solution, and the reaction was continued to stir. 9.5 hours. When the reaction is complete, cool the reaction solution to room temperature, then add 30 mL of 2-methyltetrahydrofuran and 30 mL of water, separate the organic phase, wash it once with 20 mL of saturated brine, and then dry the organic phase over anhydrous sodium sulfate and filter. , concentrated under reduced pressure to obtain a pale yellow crude product. Add 5 mL of methyl tert-butyl ether to the crude product for pulping, and obtain light yellow solid I-6-4 (60 mg) after filtration. LCMS(M+H):421.
步骤4:化合物I-6-5的合成Step 4: Synthesis of compound I-6-5
向封管中加入I-6-4(50mg,119μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(S)-3-氨基哌啶-1-羧酸叔丁酯(71mg,355μmol,3eq)和N,N-二异丙基乙胺(76mg,0.59mmol,5eq),然后置于145℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用50mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物粗品I-6-5(40mg)。LCMS(M+H):585。Add I-6-4 (50 mg, 119 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (S)-3-aminopiperidine-1-carboxylic acid tert-butyl ester (71 mg, 355μmol, 3eq) and N,N-diisopropylethylamine (76mg, 0.59mmol, 5eq), and then placed in an oil bath at 145°C and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 50mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain crude product I-6-5 (40 mg) as a pale yellow oil. LCMS(M+H):585.
步骤5:化合物I-6-6的合成Step 5: Synthesis of Compound I-6-6
向反应管中加入I-6-5(40mg,68μmol,1eq)、Pd(dppf)Cl2(5mg,6.8μmol,0.1eq)、1mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(16mg,205μmol,3eq)和三乙胺(35mg,347μmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤, 所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到淡黄色油状物I-1-4(30mg)。LCMS(M+H):583。Add I-6-5 (40 mg, 68 μmol, 1 eq), Pd(dppf)Cl 2 (5 mg, 6.8 μmol, 0.1 eq), and 1 mL of N,N-dimethylformamide into the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (16 mg, 205 μmol, 3 eq) and triethylamine (35 mg, 347 μmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and the reaction was stirred in an oil bath at 130°C for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10mL of water, and then extract with ethyl acetate (10mL*3), combine the organic phases, and wash with 25mL of saturated brine. The obtained organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain a crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain light yellow oily substance I-1-4 (30 mg). LCMS(M+H):583.
步骤6:化合物I-6的合成Step 6: Synthesis of compound I-6
向化合物I-6-6(30mg,51μmol,1eq)的二氯甲烷溶液中(1mL)加入三氟乙酸(0.1mL),然后于室温条件下搅拌5小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到淡黄色固体I-6(11mg)。LCMS(M+H):483。Trifluoroacetic acid (0.1 mL) was added to a solution (1 mL) of compound I-6-6 (30 mg, 51 μmol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-6 (11 mg). LCMS(M+H):483.
实施例7、化合物I-7的合成与表征
Example 7. Synthesis and characterization of compound I-7
步骤1:化合物I-7-1的合成Step 1: Synthesis of compound I-7-1
向三口瓶中加入I-6-2(2.5g,10.4mmol,1eq),加入50mL的DMF溶解,于0℃条件下分批加入CDI(3.4g,21mmol,2eq),加料完毕继续搅拌0.5小时。然后向其中滴加氨水15g,100mmol,10eq),继续搅拌1小时。待反应完全,向反应液中加入100mL饱和氯化铵溶液,100mL饱和食盐水,100mL2-甲基四氢呋喃,分液得到有机相,经无水硫酸钠干燥后过滤、减压浓缩得到粗品。向粗品中加入50mL甲基叔丁基醚进行打浆,经过滤得到黄色固体I-7-1(2g)。LCMS(M+H):239。Add I-6-2 (2.5g, 10.4mmol, 1eq) to the three-necked flask, add 50mL of DMF to dissolve, add CDI (3.4g, 21mmol, 2eq) in batches at 0°C, and continue stirring for 0.5 hours after the addition is complete. . Then add dropwise ammonia 15g, 100mmol, 10eq) to it, and continue stirring for 1 hour. When the reaction is complete, add 100 mL of saturated ammonium chloride solution, 100 mL of saturated saline solution, and 100 mL of 2-methyltetrahydrofuran to the reaction solution. Separate the layers to obtain the organic phase, which is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain the crude product. 50 mL of methyl tert-butyl ether was added to the crude product for pulping, and yellow solid I-7-1 (2 g) was obtained after filtration. LCMS(M+H):239.
步骤2:化合物I-7-2的合成Step 2: Synthesis of compound I-7-2
向反应瓶中加入I-7-1(0.8g,3.4mmol,1eq),再加入20mL1,2-二氯乙烷溶解,然后向其中加入三氯氧磷(3.1g,20.1mmol,6eq),置换氮气后于60℃油浴中搅拌1小时。待反应完全后,将反应液加入到50mL水中,再加入50mL二氯甲烷,分液,有机相经50mL水洗涤,再经饱和碳酸钠溶液洗涤至无明显气泡产生,有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经20mL甲基叔丁基醚进行打浆,经过滤得到类白色固体I-7-2(0.56g)。Add I-7-1 (0.8g, 3.4mmol, 1eq) to the reaction bottle, then add 20mL of 1,2-dichloroethane to dissolve, then add phosphorus oxychloride (3.1g, 20.1mmol, 6eq), After replacing with nitrogen, the mixture was stirred in a 60°C oil bath for 1 hour. After the reaction is complete, add the reaction solution to 50 mL of water, then add 50 mL of methylene chloride, and separate the liquids. The organic phase is washed with 50 mL of water, and then with saturated sodium carbonate solution until no obvious bubbles are generated. The organic phase is washed with anhydrous sodium sulfate. Dry, filter and concentrate under reduced pressure to obtain crude product. The crude product was slurried with 20 mL of methyl tert-butyl ether, and filtered to obtain an off-white solid I-7-2 (0.56 g).
步骤3:化合物I-7-3的合成Step 3: Synthesis of compound I-7-3
向反应管中加入I-7-2(200mg,0.9mmol,1eq)、Pd(dppf)Cl2(66mg,90μmol,0.1 eq)、3mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(220mg,2.7mmol,3eq)和三乙胺(460mg,4.5mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到淡黄色油状物I-7-3(130mg)。LCMS(M+H):219。Add I-7-2 (200mg, 0.9mmol, 1eq) and Pd(dppf)Cl 2 (66mg, 90μmol, 0.1 eq), 3mL of N,N-dimethylformamide, replace it with nitrogen three times, and then add dimethylphosphine oxide (220mg, 2.7mmol, 3eq) and triethylamine (460mg, 4.5mmol, 5eq) in sequence, The nitrogen was replaced three times again, and the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain light yellow oily substance I-7-3 (130 mg). LCMS(M+H):219.
步骤4:化合物I-7-4的合成Step 4: Synthesis of compound I-7-4
向反应管中加入I-7-3(130mg,0.6mmol,1eq)、3mL的N,N-二甲基甲酰胺,氮气吹扫后向其中加入NBS(130mg,0.7mmol,1.2eq),将反应于常温搅拌10小时。待反应完全,将反应液加入到30mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,分别用25mL饱和硫代硫酸钠溶液、25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-7-4(180mg)。LCMS(M+H):297。Add I-7-3 (130 mg, 0.6 mmol, 1 eq) and 3 mL of N, N-dimethylformamide to the reaction tube. After purging with nitrogen, add NBS (130 mg, 0.7 mmol, 1.2 eq) to the reaction tube. The reaction was stirred at room temperature for 10 hours. When the reaction is complete, add the reaction solution to 30 mL of water, then extract with ethyl acetate (10 mL*3), combine the organic phases, and wash with 25 mL of saturated sodium thiosulfate solution and 25 mL of saturated brine, and the resulting organic phase is anhydrous. Dry over sodium sulfate, filter, and concentrate under reduced pressure to obtain light yellow oily substance I-7-4 (180 mg). LCMS(M+H):297.
步骤5:化合物I-7-5的合成Step 5: Synthesis of Compound I-7-5
向反应瓶中加入I-7-4(180mg,0.6mmol,1eq)、3mL的无水四氢呋喃,然后向其中加入Boc2O(260mg,1.2mmol,2eq)和DMAP(10mg,60μmol,0.1eq),将反应于常温搅拌12小时。待反应完全,将反应液加入到30mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,分别用25mL饱和硫代硫酸钠溶液、25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-7-5(100mg)。LCMS(M+H):397。Add I-7-4 (180 mg, 0.6 mmol, 1 eq) and 3 mL of anhydrous tetrahydrofuran to the reaction flask, and then add Boc 2 O (260 mg, 1.2 mmol, 2 eq) and DMAP (10 mg, 60 μmol, 0.1 eq). , the reaction was stirred at room temperature for 12 hours. When the reaction is complete, add the reaction solution to 30 mL of water, then extract with ethyl acetate (10 mL*3), combine the organic phases, and wash with 25 mL of saturated sodium thiosulfate solution and 25 mL of saturated brine, and the resulting organic phase is anhydrous. Dry over sodium sulfate, filter, and concentrate under reduced pressure to obtain light yellow oily substance I-7-5 (100 mg). LCMS(M+H):397.
步骤6:化合物I-7-6的合成Step 6: Synthesis of Compound I-7-6
向反应瓶中加入I-7-5(100mg,252μmol,1eq)、Bpin2(130mg,0.5mmol,2eq)、无水乙酸钾(74mg,0.76mmol,3eq)和Pd(dppf)Cl2(18mg,25μmol,0.1eq),再加入1mL的无水1,4-二氧六环,置换氮气后将反应置于80℃油浴中搅拌8小时。待反应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物I-7-6(100mg)。LCMS(M+H):445。Add I-7-5 (100mg, 252μmol, 1eq), Bpin 2 (130mg, 0.5mmol, 2eq), anhydrous potassium acetate (74mg, 0.76mmol, 3eq) and Pd(dppf)Cl 2 (18mg) to the reaction flask. , 25 μmol, 0.1 eq), then add 1 mL of anhydrous 1,4-dioxane, replace the nitrogen, and place the reaction in an 80°C oil bath and stir for 8 hours. When the reaction is complete, add the reaction solution to 20 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A yellow oily substance I-7-6 (100 mg) was obtained. LCMS(M+H):445.
步骤7:化合物I-7-7的合成Step 7: Synthesis of Compound I-7-7
向反应瓶中加入I-7-6(100mg,225μmol,1eq)、I-1-1(92mg,450μmol,2eq)、碳酸钠(50mg,472μmol,3eq),再加入2mL1,4-二氧六环和0.2mL水,搅拌均匀后加入Pd(dppf)Cl2(16mg,22μmol,0.1eq),置换氮气后将反应置于80℃油浴中搅拌10小时。待反应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物I-7-7(20mg)。LCMS(M+H):387。Add I-7-6 (100mg, 225μmol, 1eq), I-1-1 (92mg, 450μmol, 2eq), sodium carbonate (50mg, 472μmol, 3eq) to the reaction bottle, and then add 2mL of 1,4-dioxane ring and 0.2 mL of water, stir evenly, add Pd(dppf)Cl 2 (16 mg, 22 μmol, 0.1 eq), replace nitrogen, and place the reaction in an 80°C oil bath for 10 hours. When the reaction is complete, add the reaction solution to 20 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A yellow oily substance I-7-7 (20 mg) was obtained. LCMS(M+H):387.
步骤8:化合物I-7-8的合成 Step 8: Synthesis of compound I-7-8
向封管中加入I-7-7(20mg,52μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(S)-3-氨基哌啶-1-羧酸叔丁酯(31mg,155μmol,3eq)和N,N-二异丙基乙胺(33mg,256μmol,5eq),然后置于145℃油浴中搅拌5小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用50mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物粗品I-7-8(20mg)。LCMS(M+H):551。Add I-7-7 (20 mg, 52 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (S)-3-aminopiperidine-1-carboxylic acid tert-butyl ester (31 mg, 155 μmol, 3 eq) and N,N-diisopropylethylamine (33 mg, 256 μmol, 5 eq), and then placed in an oil bath at 145°C and stirred for 5 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 50mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain crude product I-7-8 (20 mg) as a pale yellow oil. LCMS(M+H):551.
步骤9:化合物I-7的合成Step 9: Synthesis of Compound I-7
向化合物I-7-8(20mg,36μmol,1eq)的二氯甲烷溶液中(1mL)加入三氟乙酸(0.1mL),然后于室温条件下搅拌5小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到淡黄色固体I-7(9mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.36(s,1H),8.23(d,J=12.0Hz,1H),8.14(d,J=4.0Hz,1H),7.40(d,J=8.0Hz,1H),7.07(d,J=4.0Hz,1H),4.23(s,1H),3.64(dd,J=12.0,4.0Hz,1H),3.45(d,J=12.0Hz,1H),3.25–3.09(m,2H),2.29–2.10(m,8H),1.99-1.90(m,1H),1.88–1.76(m,1H);LCMS(M+H):451。Trifluoroacetic acid (0.1 mL) was added to a solution (1 mL) of compound I-7-8 (20 mg, 36 μmol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-7 (9 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.36 (s, 1H), 8.23 (d, J = 12.0Hz, 1H), 8.14 (d, J = 4.0Hz, 1H), 7.40 (d, J = 8.0Hz ,1H),7.07(d,J=4.0Hz,1H),4.23(s,1H),3.64(dd,J=12.0,4.0Hz,1H),3.45(d,J=12.0Hz,1H),3.25 –3.09(m,2H),2.29–2.10(m,8H),1.99-1.90(m,1H),1.88–1.76(m,1H); LCMS(M+H):451.
实施例8、化合物I-13的合成与表征
Example 8. Synthesis and characterization of compound I-13
步骤1:化合物I-13-1的合成Step 1: Synthesis of Compound I-13-1
向封管中加入I-1-2(50mg,137μmol,1eq),加入0.5mL的N-甲基吡咯烷酮,再分别加入(2R,5S)-5-氨基-2-甲基哌啶-1-羧酸苄酯(100mg,400μmol,3eq)和N,N-二异丙基乙胺(90mg,0.7mmol,5eq),然后置于140℃油浴中搅拌8小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-13-1(80mg)。LCMS(M+H):576。Add I-1-2 (50 mg, 137 μmol, 1 eq) to the sealed tube, add 0.5 mL of N-methylpyrrolidone, and then add (2R, 5S)-5-amino-2-methylpiperidine-1- respectively. Benzyl carboxylate (100 mg, 400 μmol, 3 eq) and N, N-diisopropylethylamine (90 mg, 0.7 mmol, 5 eq) were then placed in a 140°C oil bath and stirred for 8 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10mL of water, and then extract with ethyl acetate (10mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oily substance I-13-1 (80 mg). LCMS(M+H):576.
步骤2:化合物I-13-2的合成 Step 2: Synthesis of compound I-13-2
向反应管中加入I-13-1(80mg,138μmol,1eq)、Pd(dppf)Cl2(10mg,13.7μmol,0.1eq)、0.6mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(32mg,410μmol,3eq)和三乙胺(70mg,0.69mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到淡黄色油状物I-13-2(50mg)。LCMS(M+H):574。Add I-13-1 (80 mg, 138 μmol, 1 eq), Pd(dppf)Cl 2 (10 mg, 13.7 μmol, 0.1 eq), and 0.6 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Then dimethylphosphine oxide (32 mg, 410 μmol, 3 eq) and triethylamine (70 mg, 0.69 mmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was placed in a 130°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain light yellow oily substance I-13-2 (50 mg). LCMS(M+H):574.
步骤3:化合物I-13的合成Step 3: Synthesis of Compound I-13
向化合物I-13-2(50mg,20.93μmol,1eq)中加入HBr/HOAc(1mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到淡黄色固体I-13(12mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.41(s,1H),8.12(d,J=8.0Hz,1H),7.79(d,J=4.0Hz,1H),7.75(s,1H),7.39–7.30(m,1H),7.08(td,J=8.0,2.4Hz,1H),6.92(d,J=8.0Hz,1H),3.89-3.88(m,1H),3.53(dd,J=12.0,4.0Hz,1H),3.38–3.27(m,1H),3.18(dd,J=16.0,4.0Hz,1H),1.91-1.86(m,9H),1.81–1.67(m,1H),1.32(d,J=4.0Hz,3H);LCMS(M+H):440。HBr/HOAc (1 mL) was added to compound I-13-2 (50 mg, 20.93 μmol, 1 eq), and then stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-13 (12 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.41 (s, 1H), 8.12 (d, J = 8.0Hz, 1H), 7.79 (d, J = 4.0Hz, 1H), 7.75 (s, 1H), 7.39 –7.30(m,1H),7.08(td,J=8.0,2.4Hz,1H),6.92(d,J=8.0Hz,1H),3.89-3.88(m,1H),3.53(dd,J=12.0 ,4.0Hz,1H),3.38–3.27(m,1H),3.18(dd,J=16.0,4.0Hz,1H),1.91-1.86(m,9H),1.81–1.67(m,1H),1.32( d, J=4.0Hz, 3H); LCMS (M+H): 440.
实施例9、化合物I-14的合成与表征
Example 9. Synthesis and characterization of compound I-14
步骤1:化合物I-14-1的合成Step 1: Synthesis of Compound I-14-1
向封管中加入I-1-2(100mg,274μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(2s,4r)-叔丁基2-氨基-6-氮杂螺[3.4]辛烷-6-甲酸酯(190mg,0.8mmol,3eq)和N,N-二异丙基乙胺(180mg,1.4mmol,5eq),然后置于150℃油浴中搅拌8小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-14-1(130mg)。LCMS(M+H):554。 Add I-1-2 (100 mg, 274 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (2s, 4r)-tert-butyl 2-amino-6-azaspiro [3.4 ] Octane-6-carboxylate (190 mg, 0.8 mmol, 3 eq) and N, N-diisopropylethylamine (180 mg, 1.4 mmol, 5 eq), and then placed in a 150°C oil bath and stirred for 8 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (10mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oily substance I-14-1 (130 mg). LCMS(M+H):554.
步骤2:化合物I-14-2的合成Step 2: Synthesis of Compound I-14-2
向反应管中加入I-14-1(130mg,234μmol,1eq)、Pd(dppf)Cl2(17mg,23μmol,0.1eq)、1mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(55mg,0.7mmol,3eq)和三乙胺(120mg,1.2mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物I-14-2(120mg)。LCMS(M+H):552。Add I-14-1 (130 mg, 234 μmol, 1 eq), Pd(dppf)Cl 2 (17 mg, 23 μmol, 0.1 eq), and 1 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (55 mg, 0.7 mmol, 3 eq) and triethylamine (120 mg, 1.2 mmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil I-14-2 (120 mg). LCMS(M+H):552.
步骤3:化合物I-14的合成Step 3: Synthesis of Compound I-14
向化合物I-14-2(120mg,20.93μmol,1eq)中加入二氯甲烷(5mL),再向其中加入三氟乙酸(0.5mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到淡黄色固体I-14(45mg)。1H NMR(400MHz,Deuterium Oxide)δ=7.99(d,J=8.0Hz,1H),7.72(d,J=8.0Hz,1H),7.61(s,1H),7.28(dd,J=14.0,7.2Hz,1H),6.94(t,J=8.0Hz,1H),6.76(d,J=4.0Hz,1H),3.86(p,J=8.0Hz,1H),3.33-3.28(m,4H),2.41(t,J=10.0Hz,2H),2.05(t,J=8.0Hz,2H),1.98(dd,J=12.0,8.0Hz,2H),1.89(d,J=12.0Hz,6H);LCMS(M+H):452。Dichloromethane (5 mL) was added to compound I-14-2 (120 mg, 20.93 μmol, 1 eq), trifluoroacetic acid (0.5 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-14 (45 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 7.99 (d, J = 8.0Hz, 1H), 7.72 (d, J = 8.0Hz, 1H), 7.61 (s, 1H), 7.28 (dd, J = 14.0, 7.2Hz,1H),6.94(t,J=8.0Hz,1H),6.76(d,J=4.0Hz,1H),3.86(p,J=8.0Hz,1H),3.33-3.28(m,4H) ,2.41(t,J=10.0Hz,2H),2.05(t,J=8.0Hz,2H),1.98(dd,J=12.0,8.0Hz,2H),1.89(d,J=12.0Hz,6H) ;LCMS(M+H):452.
实施例10、化合物I-15的合成与表征
Example 10. Synthesis and characterization of compound I-15
步骤1:化合物I-15-1的合成Step 1: Synthesis of Compound I-15-1
向封管中加入I-1-2(100mg,274μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(R)-叔-丁基3-氨基-1-氧杂-8-氮杂螺[4.5]癸烷-8-甲酸基酯(210mg,0.8mmol,3eq)和N,N-二异丙基乙胺(180mg,1.4mmol,5eq),然后置于150℃油浴中搅拌8小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙 酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-15-1(130mg)。LCMS(M+H):584。Add I-1-2 (100 mg, 274 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (R)-tert-butyl 3-amino-1-oxa-8-nitrogen respectively. Heterospiro[4.5]decane-8-carboxylate (210mg, 0.8mmol, 3eq) and N,N-diisopropylethylamine (180mg, 1.4mmol, 5eq), then placed in an oil bath at 150°C and stirred 8 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20 mL of water, and then add it with ethanol. Extract with ethyl acid (10 mL*3), combine the organic phases, and wash with 20 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain light yellow oily substance I-15-1 (130 mg). LCMS(M+H):584.
步骤2:化合物I-15-2的合成Step 2: Synthesis of Compound I-15-2
向反应管中加入I-15-1(130mg,223μmol,1eq)、Pd(dppf)Cl2(16mg,22μmol,0.1eq)、1mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(52mg,0.67mmol,3eq)和三乙胺(111mg,1.1mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物I-15-2(100mg)。LCMS(M+H):582。Add I-15-1 (130 mg, 223 μmol, 1 eq), Pd(dppf)Cl 2 (16 mg, 22 μmol, 0.1 eq), and 1 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (52 mg, 0.67 mmol, 3 eq) and triethylamine (111 mg, 1.1 mmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil I-15-2 (100 mg). LCMS(M+H):582.
步骤3:化合物I-15的合成Step 3: Synthesis of Compound I-15
向化合物I-15-2(100mg,172μmol,1eq)中加入二氯甲烷(5mL),再向其中加入三氟乙酸(0.5mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到淡黄色固体I-15(41mg)。1H NMR(400MHz,Deuterium Oxide)δ=7.79(d,J=8.0Hz,1H),7.62(d,J=4.0Hz,1H),7.39(s,1H),7.20(dd,J=12.0,8.0Hz,1H),6.80-6.76(m,1H),6.67(d,J=8.0Hz,1H),3.87-3.80(m,2H),3.50–3.40(m,1H),3.32–3.13(m,4H),2.04-1.99(m,1H),1.96–1.69(m,10H),1.56-1.51(m,1H);LCMS(M+H):482。Dichloromethane (5 mL) was added to compound I-15-2 (100 mg, 172 μmol, 1 eq), trifluoroacetic acid (0.5 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-15 (41 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 7.79 (d, J = 8.0Hz, 1H), 7.62 (d, J = 4.0Hz, 1H), 7.39 (s, 1H), 7.20 (dd, J = 12.0, 8.0Hz,1H),6.80-6.76(m,1H),6.67(d,J=8.0Hz,1H),3.87-3.80(m,2H),3.50–3.40(m,1H),3.32–3.13(m ,4H),2.04-1.99(m,1H),1.96–1.69(m,10H),1.56-1.51(m,1H); LCMS(M+H):482.
实施例11、化合物I-16的合成与表征
Example 11. Synthesis and characterization of compound I-16
步骤1:化合物I-16-1的合成Step 1: Synthesis of Compound I-16-1
向封管中加入I-1-2(100mg,274μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(S)-1-叔丁氧羰基-3-氨基吡咯烷(150mg,0.8mmol,3eq)和N,N-二异丙基乙胺(180mg,1.4mmol,5eq),然后置于135℃油浴中搅拌10小时。待反应完全,反 应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-16-1(80mg)。LCMS(M+H):514。Add I-1-2 (100 mg, 274 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (S)-1-tert-butoxycarbonyl-3-aminopyrrolidine (150 mg, 0.8 mmol, 3eq) and N,N-diisopropylethylamine (180mg, 1.4mmol, 5eq), and then placed in a 135°C oil bath and stirred for 10 hours. Wait until the reaction is complete, then The reaction solution was cooled to room temperature, and the reaction solution was added to 20 mL of water, and then extracted with ethyl acetate (10 mL*3). The organic phases were combined and washed with 20 mL of saturated brine. The resulting organic phase was dried over anhydrous sodium sulfate, filtered, and reduced. Concentrate under pressure to obtain light yellow oily substance I-16-1 (80 mg). LCMS(M+H):514.
步骤2:化合物I-16-2的合成Step 2: Synthesis of Compound I-16-2
向反应管中加入I-16-1(80mg,156μmol,1eq)、Pd(dppf)Cl2(11mg,15μmol,0.1eq)、1mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(30mg,395μmol,2.5eq)和三乙胺(46mg,455μmol,3eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物I-16-2(45mg)。LCMS(M+H):512。Add I-16-1 (80 mg, 156 μmol, 1 eq), Pd(dppf)Cl 2 (11 mg, 15 μmol, 0.1 eq), and 1 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (30 mg, 395 μmol, 2.5 eq) and triethylamine (46 mg, 455 μmol, 3 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil I-16-2 (45 mg). LCMS(M+H):512.
步骤3:化合物I-16的合成Step 3: Synthesis of Compound I-16
向化合物I-16-2(45mg,88μmol,1eq)中加入二氯甲烷(5mL),再向其中加入三氟乙酸(0.5mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到淡黄色固体I-16(21mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.23(d,J=8.0Hz,1H),8.21–8.10(m,2H),7.48(dd,J=16.0,8.0Hz,1H),7.29(td,J=8.0,2.4Hz,1H),7.01(d,J=8.0Hz,1H),4.53(s,1H),3.74-3.69(m,1H),3.57-3.54(m,2H),3.41(dd,J=12.0,4.0Hz,1H),2.53-2.44(m,1H),2.25–2.13(m,1H),2.00-1.92(m,6H).;LCMS(M+H):412。Dichloromethane (5 mL) was added to compound I-16-2 (45 mg, 88 μmol, 1 eq), trifluoroacetic acid (0.5 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-16 (21 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.23 (d, J = 8.0Hz, 1H), 8.21–8.10 (m, 2H), 7.48 (dd, J = 16.0, 8.0Hz, 1H), 7.29 (td, J=8.0,2.4Hz,1H),7.01(d,J=8.0Hz,1H),4.53(s,1H),3.74-3.69(m,1H),3.57-3.54(m,2H),3.41(dd ,J=12.0,4.0Hz,1H),2.53-2.44(m,1H),2.25–2.13(m,1H),2.00-1.92(m,6H).;LCMS(M+H):412.
实施例12、化合物I-17的合成与表征
Example 12. Synthesis and characterization of compound I-17
步骤1:化合物I-17-1的合成Step 1: Synthesis of Compound I-17-1
向封管中加入I-1-2(70mg,192μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入6-氨基-3-氮杂双环[3.1.0]己烷-3-羧酸叔丁酯(114mg,0.58mmol,3eq)和N,N- 二异丙基乙胺(124mg,0.96mmol,5eq),然后置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-17-1(62mg)。LCMS(M+H):526。Add I-1-2 (70 mg, 192 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add 6-amino-3-azabicyclo[3.1.0]hexane-3-carboxy respectively. Tert-butyl acid ester (114mg, 0.58mmol, 3eq) and N,N- Diisopropylethylamine (124 mg, 0.96 mmol, 5 eq) was then placed in a 130°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (10mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oily substance I-17-1 (62 mg). LCMS(M+H):526.
步骤2:化合物I-17-2的合成Step 2: Synthesis of Compound I-17-2
向反应管中加入I-17-1(62mg,119μmol,1eq)、Pd(dppf)Cl2(9mg,12μmol,0.1eq)、1mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(28mg,359μmol,3eq)和三乙胺(61mg,598μmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物I-17-2(30mg)。LCMS(M+H):524。Add I-17-1 (62 mg, 119 μmol, 1 eq), Pd(dppf)Cl 2 (9 mg, 12 μmol, 0.1 eq), and 1 mL of N,N-dimethylformamide into the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (28 mg, 359 μmol, 3 eq) and triethylamine (61 mg, 598 μmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil I-17-2 (30 mg). LCMS(M+H):524.
步骤3:化合物I-17的合成Step 3: Synthesis of Compound I-17
向化合物I-17-2(30mg,88μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到淡黄色固体I-17(11mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.39(d,J=8.0Hz,1H),8.18(s,1H),8.09(d,J=4.0Hz,1H),7.46(dd,J=16.0,8.0Hz,1H),7.26(td,J=8.0,4.0Hz,1H),7.06(s,1H),3.68(s,4H),2.81(t,J=2.4Hz,1H),2.20(s,2H),1.96(d,J=16.0Hz,6H);LCMS(M+H):424。Dichloromethane (2 mL) was added to compound I-17-2 (30 mg, 88 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-17 (11 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.39 (d, J = 8.0Hz, 1H), 8.18 (s, 1H), 8.09 (d, J = 4.0Hz, 1H), 7.46 (dd, J = 16.0, 8.0Hz,1H),7.26(td,J=8.0,4.0Hz,1H),7.06(s,1H),3.68(s,4H),2.81(t,J=2.4Hz,1H),2.20(s, 2H), 1.96 (d, J=16.0Hz, 6H); LCMS (M+H): 424.
实施例13、化合物I-18的合成与表征
Example 13. Synthesis and characterization of compound I-18
步骤1:化合物I-18-1的合成Step 1: Synthesis of Compound I-18-1
向封管中加入I-1-2(50mg,137μmol,1eq),加入0.5mL的N-甲基吡咯烷酮,再 分别加入3-[(叔丁氧基)羰基]-3-氮杂二环[3.1.0]己烷-2-羧酸酯(80mg,404μmol,3eq)和N,N-二异丙基乙胺(89mg,0.69mmol,5eq),然后置于150℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-18-1(30mg)。LCMS(M+H):526。Add I-1-2 (50 mg, 137 μmol, 1 eq) to the sealed tube, add 0.5 mL of N-methylpyrrolidone, and then Add 3-[(tert-butoxy)carbonyl]-3-azabicyclo[3.1.0]hexane-2-carboxylate (80mg, 404μmol, 3eq) and N,N-diisopropylethyl ester respectively. amine (89 mg, 0.69 mmol, 5 eq), and then placed in a 150°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10mL of water, and then extract with ethyl acetate (10mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oily substance I-18-1 (30 mg). LCMS(M+H):526.
步骤2:化合物I-18-2的合成Step 2: Synthesis of Compound I-18-2
向反应管中加入I-18-1(30mg,57μmol,1eq)、Pd(dppf)Cl2(4mg,6μmol,0.1eq)、1mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(13mg,171μmol,3eq)和三乙胺(29mg,287μmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物I-18-2(20mg)。LCMS(M+H):524。Add I-18-1 (30 mg, 57 μmol, 1 eq), Pd(dppf)Cl 2 (4 mg, 6 μmol, 0.1 eq), and 1 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (13 mg, 171 μmol, 3 eq) and triethylamine (29 mg, 287 μmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil I-18-2 (20 mg). LCMS(M+H):524.
步骤3:化合物I-18的合成Step 3: Synthesis of Compound I-18
向化合物I-18-2(20mg,38μmol,1eq)中加入二氯甲烷(1mL),再向其中加入三氟乙酸(0.1mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体I-18(5mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.54(d,J=8.0Hz,1H),8.36(s,1H),8.22(d,J=8.0Hz,1H),7.53(dd,J=12.0,8.0Hz,1H),7.38(td,J=8.0,2.4Hz,1H),7.23(d,J=4.0Hz,1H),3.89–3.75(m,2H),3.56-3.48(m,2H),2.25-2.20(m,1H),2.00-1.95(m,6H),1.51–1.38(m,2H);LCMS(M+H):424。Dichloromethane (1 mL) was added to compound I-18-2 (20 mg, 38 μmol, 1 eq), trifluoroacetic acid (0.1 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid I-18 (5 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.54 (d, J = 8.0Hz, 1H), 8.36 (s, 1H), 8.22 (d, J = 8.0Hz, 1H), 7.53 (dd, J = 12.0, 8.0Hz,1H),7.38(td,J=8.0,2.4Hz,1H),7.23(d,J=4.0Hz,1H),3.89–3.75(m,2H),3.56-3.48(m,2H), 2.25-2.20(m,1H),2.00-1.95(m,6H),1.51–1.38(m,2H); LCMS(M+H):424.
实施例14、化合物I-19的合成与表征
Example 14. Synthesis and characterization of compound I-19
步骤1:化合物I-19-1的合成 Step 1: Synthesis of Compound I-19-1
向封管中加入I-1-2(70mg,192μmol,1eq),加入0.5mL的N-甲基吡咯烷酮,再分别加入4-(氨甲基)-4-氟哌啶-1-羧酸叔丁酯(134mg,0.58mmol,3eq)和N,N-二异丙基乙胺(124mg,0.96mmol,5eq),然后置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-19-1(104mg)。LCMS(M+H):560。Add I-1-2 (70 mg, 192 μmol, 1 eq) to the sealed tube, add 0.5 mL of N-methylpyrrolidone, and then add 4-(aminomethyl)-4-fluoroperidine-1-carboxylic acid tert. Butyl ester (134 mg, 0.58 mmol, 3 eq) and N, N-diisopropylethylamine (124 mg, 0.96 mmol, 5 eq) were then placed in a 130°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10mL of water, and then extract with ethyl acetate (10mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oily substance I-19-1 (104 mg). LCMS(M+H):560.
步骤2:化合物I-19-2的合成Step 2: Synthesis of Compound I-19-2
向反应管中加入I-19-1(104mg,186μmol,1eq)、Pd(dppf)Cl2(14mg,19μmol,0.1eq)、2mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(44mg,0.56mmol,3eq)和三乙胺(94mg,0.93mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物I-19-2(34mg)。LCMS(M+H):558。Add I-19-1 (104 mg, 186 μmol, 1 eq), Pd(dppf)Cl 2 (14 mg, 19 μmol, 0.1 eq), and 2 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (44 mg, 0.56 mmol, 3 eq) and triethylamine (94 mg, 0.93 mmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil I-19-2 (34 mg). LCMS(M+H):558.
步骤3:化合物I-19的合成Step 3: Synthesis of Compound I-19
向化合物I-19-2(34mg,61μmol,1eq)中加入二氯甲烷(5mL),再向其中加入三氟乙酸(0.5mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体I-19(13mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.33(s,1H),8.21(s,1H),8.13(d,J=4.0Hz,1H),7.51(dd,J=16.0,8.0Hz,1H),7.30(td,J=8.0,2.4Hz,1H),7.01(d,J=4.0Hz,1H),3.72(d,J=24.0Hz,2H),3.42(d,J=16.0Hz,2H),3.32–3.20(m,2H),2.22(s,2H),2.02-1.88(m,8H);19F NMR(376MHz,Deuterium Oxide)δ-165.12;LCMS(M+H):458。Dichloromethane (5 mL) was added to compound I-19-2 (34 mg, 61 μmol, 1 eq), trifluoroacetic acid (0.5 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid I-19 (13 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.33 (s, 1H), 8.21 (s, 1H), 8.13 (d, J = 4.0Hz, 1H), 7.51 (dd, J = 16.0, 8.0Hz, 1H) ,7.30(td,J=8.0,2.4Hz,1H),7.01(d,J=4.0Hz,1H),3.72(d,J=24.0Hz,2H),3.42(d,J=16.0Hz,2H) ,3.32–3.20(m,2H),2.22(s,2H),2.02-1.88(m,8H); 19 F NMR (376MHz, Deuterium Oxide) δ-165.12; LCMS (M+H): 458.
实施例15、化合物I-20的合成与表征
Example 15. Synthesis and characterization of compound I-20
步骤1:化合物I-20-1的合成Step 1: Synthesis of Compound I-20-1
向封管中加入I-1-2(100mg,274μmol,1eq),加入0.5mL的N-甲基吡咯烷酮,再分别加入叔丁基(2R,4S)-4-氨基-2-甲基吡咯烷-1-甲酸叔丁酯(165mg,0.82mmol,3eq)和N,N-二异丙基乙胺(177mg,1.4mmol,5eq),然后置于145℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-20-1(162mg)。LCMS(M+H):528。Add I-1-2 (100mg, 274μmol, 1eq) to the sealed tube, add 0.5mL of N-methylpyrrolidone, and then add tert-butyl (2R,4S)-4-amino-2-methylpyrrolidine respectively. -1-tert-butyl formate (165 mg, 0.82 mmol, 3 eq) and N, N-diisopropylethylamine (177 mg, 1.4 mmol, 5 eq), and then placed in a 145°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10mL of water, and then extract with ethyl acetate (10mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oily substance I-20-1 (162 mg). LCMS(M+H):528.
步骤2:化合物I-20-2的合成Step 2: Synthesis of Compound I-20-2
向反应管中加入I-20-1(162mg,306μmol,1eq)、Pd(dppf)Cl2(22mg,31μmol,0.1eq)、2mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(72mg,0.92mmol,3eq)和三乙胺(155mg,1.5mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物I-20-2(74mg)。LCMS(M+H):526。Add I-20-1 (162 mg, 306 μmol, 1 eq), Pd(dppf)Cl 2 (22 mg, 31 μmol, 0.1 eq), and 2 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (72 mg, 0.92 mmol, 3 eq) and triethylamine (155 mg, 1.5 mmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil I-20-2 (74 mg). LCMS(M+H):526.
步骤3:化合物I-20的合成Step 3: Synthesis of Compound I-20
向化合物I-20-2(74mg,141μmol,1eq)中加入二氯甲烷(5mL),再向其中加入三氟乙酸(0.5mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体I-20(13mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.24(d,J=8.0Hz,1H),8.15(s,1H),8.09(d,J=4.0Hz,1H),7.48(dd,J=16.0,8.0Hz,1H),7.28(td,J=8.0,2.4Hz,1H),7.02(dd,J=8.0,4.0Hz,1H),4.60-4.56(m,1H),3.89-3.80(m,1H),3.72(dd,J=12.0,8.0Hz,1H),3.31(dd,J=12.0,8.0Hz,1H),2.78-2.71(m,1H),1.96(t,J=12.0Hz,6H),1.86-1.77(m,1H),1.52(d,J=4.0Hz,3H);LCMS(M+H):426。Dichloromethane (5 mL) was added to compound I-20-2 (74 mg, 141 μmol, 1 eq), trifluoroacetic acid (0.5 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid I-20 (13 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.24 (d, J = 8.0Hz, 1H), 8.15 (s, 1H), 8.09 (d, J = 4.0Hz, 1H), 7.48 (dd, J = 16.0, 8.0Hz,1H),7.28(td,J=8.0,2.4Hz,1H),7.02(dd,J=8.0,4.0Hz,1H),4.60-4.56(m,1H),3.89-3.80(m,1H ),3.72(dd,J=12.0,8.0Hz,1H),3.31(dd,J=12.0,8.0Hz,1H),2.78-2.71(m,1H),1.96(t,J=12.0Hz,6H) ,1.86-1.77(m,1H),1.52(d,J=4.0Hz,3H); LCMS(M+H):426.
实施例16、化合物I-23的合成与表征
Example 16. Synthesis and characterization of compound I-23
步骤1:化合物I-23-1的合成Step 1: Synthesis of Compound I-23-1
向封管中加入I-1-2(100mg,274μmol,1eq),加入0.5mL的N-甲基吡咯烷酮,再分别加入叔丁基(3R,4S)-3-氨基-4-氟吡咯烷-1-羧酸叔丁酯(168mg,0.82mmol,3eq)和N,N-二异丙基乙胺(177mg,1.4mmol,5eq),然后置于145℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-23-1(96mg)。LCMS(M+H):532。Add I-1-2 (100 mg, 274 μmol, 1 eq) to the sealed tube, add 0.5 mL of N-methylpyrrolidone, and then add tert-butyl (3R, 4S)-3-amino-4-fluoropyrrolidone- 1-tert-butylcarboxylate (168 mg, 0.82 mmol, 3 eq) and N, N-diisopropylethylamine (177 mg, 1.4 mmol, 5 eq) were then placed in a 145°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10mL of water, and then extract with ethyl acetate (10mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oily substance I-23-1 (96 mg). LCMS(M+H):532.
步骤2:化合物I-23-2的合成Step 2: Synthesis of Compound I-23-2
向反应管中加入I-23-1(96mg,180μmol,1eq)、Pd(dppf)Cl2(13mg,18μmol,0.1eq)、2mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(42mg,0.54mmol,3eq)和三乙胺(91mg,0.9mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物I-23-2(51mg)。LCMS(M+H):530。Add I-23-1 (96 mg, 180 μmol, 1 eq), Pd(dppf)Cl 2 (13 mg, 18 μmol, 0.1 eq), and 2 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (42 mg, 0.54 mmol, 3 eq) and triethylamine (91 mg, 0.9 mmol, 5 eq) were added in sequence, nitrogen was replaced three times again, and the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil I-23-2 (51 mg). LCMS(M+H):530.
步骤3:化合物I-23的合成Step 3: Synthesis of Compound I-23
向化合物I-23-2(51mg,96μmol,1eq)中加入二氯甲烷(3mL),再向其中加入三氟乙酸(0.3mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体I-23(23mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.10(d,J=8.0Hz,1H),8.01-7.99(m,2H),7.40(dd,J=12.0,8.0Hz,1H),7.20(td,J=8.0,4.0Hz,1H),6.93(d,J=8.0Hz,1H),5.43(dt,J=52.0,4.0Hz,1H),3.99–3.68(m,3H),3.46(t,J=10.0Hz,1H),1.92(d,J=12.0Hz,6H);19F NMR(376MHz,Deuterium Oxide)δ=-195.14;LCMS(M+H):430。Dichloromethane (3 mL) was added to compound I-23-2 (51 mg, 96 μmol, 1 eq), trifluoroacetic acid (0.3 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid I-23 (23 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.10 (d, J = 8.0Hz, 1H), 8.01-7.99 (m, 2H), 7.40 (dd, J = 12.0, 8.0Hz, 1H), 7.20 (td, J=8.0,4.0Hz,1H),6.93(d,J=8.0Hz,1H),5.43(dt,J=52.0,4.0Hz,1H),3.99–3.68(m,3H),3.46(t,J =10.0Hz, 1H), 1.92 (d, J = 12.0Hz, 6H); 19 F NMR (376MHz, Deuterium Oxide) δ = -195.14; LCMS (M+H): 430.
实施例17、化合物I-24的合成与表征
Example 17. Synthesis and characterization of compound I-24
步骤1:化合物I-24-1的合成Step 1: Synthesis of Compound I-24-1
向封管中加入I-1-2(100mg,274μmol,1eq),加入0.5mL的N-甲基吡咯烷酮,再分别加入叔丁基(3R,4R)-3-氨基-4-羟基吡咯烷-1-羧酸叔丁酯(166mg,0.82mmol,3eq)和N,N-二异丙基乙胺(177mg,1.4mmol,5eq),然后置于145℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物I-24-1(180mg)。LCMS(M+H):530。Add I-1-2 (100 mg, 274 μmol, 1 eq) to the sealed tube, add 0.5 mL of N-methylpyrrolidone, and then add tert-butyl (3R, 4R)-3-amino-4-hydroxypyrrolidine- 1-Carboxylic acid tert-butyl ester (166 mg, 0.82 mmol, 3 eq) and N, N-diisopropylethylamine (177 mg, 1.4 mmol, 5 eq) were then placed in a 145°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10mL of water, and then extract with ethyl acetate (10mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oily substance I-24-1 (180 mg). LCMS(M+H):530.
步骤2:化合物I-24-2的合成Step 2: Synthesis of Compound I-24-2
向反应管中加入I-24-1(180mg,339μmol,1eq)、Pd(dppf)Cl2(25mg,34μmol,0.1eq)、2mL的N,N-二甲基甲酰胺,氮气置换三次后再向其中依次加入二甲基氧化膦(79mg,1mmol,3eq)和三乙胺(172mg,1.7mmol,5eq),再次置换氮气三次,然后将反应置于130℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到10mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物I-24-2(144mg)。LCMS(M+H):528。Add I-24-1 (180 mg, 339 μmol, 1 eq), Pd(dppf)Cl 2 (25 mg, 34 μmol, 0.1 eq), and 2 mL of N,N-dimethylformamide to the reaction tube, and replace with nitrogen three times. Dimethylphosphine oxide (79 mg, 1 mmol, 3 eq) and triethylamine (172 mg, 1.7 mmol, 5 eq) were added to it in sequence, nitrogen was replaced three times again, and then the reaction was stirred in a 130°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 10 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil I-24-2 (144 mg). LCMS(M+H):528.
步骤3:化合物I-24的合成Step 3: Synthesis of Compound I-24
向化合物I-24-2(144mg,273μmol,1eq)中加入二氯甲烷(5mL),再向其中加入三氟乙酸(0.5mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体I-24(51mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.37(d,J=8.0Hz,1H),8.13(s,1H),8.10(d,J=8.0Hz,1H),7.47(dd,J=16.0,8.0Hz,1H),7.24(td,J=8.0,4.0Hz,1H),7.02(d,J=4.0Hz,1H),4.55(s,1H),4.34(d,J=4.0Hz,1H),3.89(dd,J=16.0,8.0Hz,1H),3.58–3.45(m,3H),1.97(dd,J=12.0,8.0Hz,6H);LCMS(M+H):428。 Dichloromethane (5 mL) was added to compound I-24-2 (144 mg, 273 μmol, 1 eq), trifluoroacetic acid (0.5 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid I-24 (51 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.37 (d, J = 8.0Hz, 1H), 8.13 (s, 1H), 8.10 (d, J = 8.0Hz, 1H), 7.47 (dd, J = 16.0, 8.0Hz,1H),7.24(td,J=8.0,4.0Hz,1H),7.02(d,J=4.0Hz,1H),4.55(s,1H),4.34(d,J=4.0Hz,1H) ,3.89(dd,J=16.0,8.0Hz,1H),3.58–3.45(m,3H),1.97(dd,J=12.0,8.0Hz,6H); LCMS(M+H):428.
实施例18、化合物I-9的合成与表征
Example 18. Synthesis and characterization of compound I-9
化合物I-9的合成参考实施例7化合物I-7的合成路线,将化合物I-9-1替换掉I-7-2作为起始原料,所得化合物I-9为淡黄色固体(3mg),LCMS(M+H):427。The synthesis of compound I-9 was based on the synthetic route of compound I-7 in Example 7. Compound I-9-1 was used as the starting material instead of I-7-2. The obtained compound I-9 was a light yellow solid (3 mg). LCMS(M+H):427.
实施例19、化合物I-10的合成与表征
Example 19. Synthesis and characterization of compound I-10
化合物I-10的合成参考实施例7化合物I-9的合成路线,将化合物I-10-1替换掉I-9-1作为起始原料,所得化合物I-10为淡黄色固体(4mg),LCMS(M+H):427。The synthesis of compound I-10 was based on the synthetic route of compound I-9 in Example 7. Compound I-10-1 was used as the starting material instead of I-9-1. The obtained compound I-10 was a light yellow solid (4 mg). LCMS(M+H):427.
实施例20、化合物I-11的合成与表征
Example 20. Synthesis and characterization of compound I-11
化合物I-11的合成参考实施例7化合物I-9的合成路线,将化合物I-11-1替换掉I-9-2作为起始原料,所得化合物I-11为淡黄色固体(5mg),LCMS(M+H):381。The synthesis of compound I-11 was based on the synthetic route of compound I-9 in Example 7. Compound I-11-1 was used as the starting material instead of I-9-2. The obtained compound I-11 was a light yellow solid (5 mg). LCMS(M+H):381.
实施例21、化合物I-12的合成与表征
Example 21. Synthesis and characterization of compound I-12
化合物I-12的合成参考实施例7化合物I-9的合成路线,将化合物I-12-1替换掉I-9-1作为起始原料,所得化合物I-12为淡黄色固体(6mg),LCMS(M+H):427。The synthesis of compound I-12 was based on the synthetic route of compound I-9 in Example 7. Compound I-12-1 was used as the starting material instead of I-9-1. The obtained compound I-12 was a light yellow solid (6 mg). LCMS(M+H):427.
实施例22、化合物I-35的合成与表征
Example 22. Synthesis and characterization of compound I-35
化合物I-35的合成参考实施例1化合物I-1的合成路线,将化合物4-氟-6-溴吲哚替换掉6-溴吲哚作为起始原料,所得化合物I-35为淡黄色固体(11mg),LCMS(M+H):444。Compound I-35 was synthesized by referring to the synthetic route of compound I-1 in Example 1. Compound 4-fluoro-6-bromoindole was replaced with 6-bromoindole as the starting material. The obtained compound I-35 was a light yellow solid. (11 mg), LCMS (M+H): 444.
实施例23、化合物I-36的合成与表征
Example 23. Synthesis and characterization of compound I-36
化合物I-36的合成参考实施例1化合物I-1的合成路线,将化合物反式-(4-氨基环己基)氨基甲酸叔丁酯替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-36为淡黄色固体(12mg),LCMS(M+H):440。Compound I-36 was synthesized by referring to the synthetic route of compound I-1 in Example 1, replacing (S)-3-aminopiperidine-1-carboxylic compound with trans-(4-aminocyclohexyl)carbamic acid tert-butyl ester. Acid tert-butyl ester, the obtained compound I-36 was a light yellow solid (12 mg), LCMS (M+H): 440.
实施例24、化合物I-37的合成与表征
Example 24. Synthesis and characterization of compound I-37
化合物I-37的合成参考实施例1化合物I-1的合成路线,将化合物3-(氨基甲基)-3-甲基-1-吖丁啶羧酸叔丁酯替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-37为淡黄色固体(12mg),LCMS(M+H):426。Compound I-37 was synthesized by referring to the synthetic route of compound I-1 in Example 1, except that (S)-3 was replaced with compound 3-(aminomethyl)-3-methyl-1-azetidinecarboxylic acid tert-butyl ester. -Aminopiperidine-1-carboxylic acid tert-butyl ester, the obtained compound I-37 was a light yellow solid (12 mg), LCMS (M+H): 426.
实施例25、化合物I-38的合成与表征
Example 25. Synthesis and characterization of compound I-38
化合物I-38的合成参考实施例1化合物I-1的合成路线,将化合物(R)-2-氨甲基-1-N-Boc-吡咯烷替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-38为淡黄色固体(8mg),LCMS(M+H):426。Compound I-38 was synthesized by referring to the synthetic route of compound I-1 in Example 1, except that compound (R)-2-aminomethyl-1-N-Boc-pyrrolidine was replaced with (S)-3-aminopiperidine- 1-tert-butylcarboxylate, the obtained compound 1-38 was a light yellow solid (8 mg), LCMS (M+H): 426.
实施例26、化合物I-39的合成与表征
Example 26. Synthesis and characterization of compound I-39
化合物I-39的合成参考实施例1化合物I-1的合成路线,将化合物(S)-2-氨甲基-1-N-Boc-吡咯烷替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-39为淡黄色固体(7mg),LCMS(M+H):426。Compound I-39 was synthesized by referring to the synthetic route of compound I-1 in Example 1, except that compound (S)-2-aminomethyl-1-N-Boc-pyrrolidine was replaced with (S)-3-aminopiperidine- 1-tert-butylcarboxylate, the obtained compound 1-39 was a light yellow solid (7 mg), LCMS (M+H): 426.
实施例27、化合物I-40的合成与表征
Example 27. Synthesis and characterization of compound I-40
化合物I-40的合成参考实施例1化合物I-1的合成路线,将化合物3-(氨基甲基)-3-氟-1-吖丁啶羧酸叔丁酯替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-40为淡黄色固体(7mg),LCMS(M+H):430。Compound I-40 was synthesized by referring to the synthetic route of compound I-1 in Example 1, except that compound 3-(aminomethyl)-3-fluoro-1-azetidinecarboxylic acid tert-butyl ester was replaced with (S)-3- Aminopiperidine-1-carboxylic acid tert-butyl ester, the obtained compound I-40 was a light yellow solid (7 mg), LCMS (M+H): 430.
实施例28、化合物I-41的合成与表征
Example 28. Synthesis and characterization of compound I-41
化合物I-41的合成参考实施例1化合物I-1的合成路线,将化合物1-叔丁氧羰基-3-(氨基甲基)吡咯烷替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-41为淡黄色固体(11mg),LCMS(M+H):426。Compound I-41 was synthesized by referring to the synthetic route of compound I-1 in Example 1, except that compound 1-tert-butoxycarbonyl-3-(aminomethyl)pyrrolidine was replaced with (S)-3-aminopiperidine-1- Tert-butyl carboxylate, the obtained compound I-41 was a light yellow solid (11 mg), LCMS (M+H): 426.
实施例29、化合物I-42的合成与表征
Example 29. Synthesis and characterization of compound I-42
化合物I-42的合成参考实施例1化合物I-1的合成路线,将化合物((1R,3S)-3-氨基环戊基)氨基甲酸叔丁酯替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-42为淡黄色固体(13mg),LCMS(M+H):426。Compound I-42 was synthesized by referring to the synthetic route of compound I-1 in Example 1, replacing (S)-3-aminopiperidine with compound ((1R,3S)-3-aminocyclopentyl)carbamate tert-butyl ester. -1-tert-butylcarboxylate, the obtained compound I-42 was a light yellow solid (13 mg), LCMS (M+H): 426.
实施例30、化合物I-43的合成与表征
Example 30. Synthesis and characterization of compound I-43
化合物I-43的合成参考实施例1化合物I-1的合成路线,将化合物((1S,3S)-3-氨基环戊基)氨基甲酸叔丁酯替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-43为淡黄色固体(14mg),LCMS(M+H):426。Compound I-43 was synthesized by referring to the synthetic route of compound I-1 in Example 1, replacing (S)-3-aminopiperidine with compound ((1S,3S)-3-aminocyclopentyl)carbamate tert-butyl ester. -1-tert-butylcarboxylate, the obtained compound I-43 was a light yellow solid (14 mg), LCMS (M+H): 426.
实施例31、化合物II-1的合成与表征
Example 31. Synthesis and characterization of compound II-1
步骤1:化合物II-1-2的合成Step 1: Synthesis of compound II-1-2
向封管中加入II-1-1(150mg,0.6mmol,1eq),加入1.5mL的N-甲基吡咯烷酮,再分别加入(S)-3-氨基哌啶-1-羧酸叔丁酯(482mg,2.4mmol,4eq)和N,N-二异丙基乙胺(311mg,2.4mmol,4eq),然后置于140℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到30mL水中,然后用乙酸乙酯萃取(20mL*3),合 并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物II-1-2(173mg)。LCMS(M+H):413。Add II-1-1 (150mg, 0.6mmol, 1eq) to the sealed tube, add 1.5mL of N-methylpyrrolidone, and then add (S)-3-aminopiperidine-1-carboxylic acid tert-butyl ester ( 482mg, 2.4mmol, 4eq) and N,N-diisopropylethylamine (311mg, 2.4mmol, 4eq), and then placed in a 140°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 30mL of water, and then extract with ethyl acetate (20mL*3). The organic phase was combined and washed with 20 mL of saturated brine. The resulting organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain light yellow oil II-1-2 (173 mg). LCMS(M+H):413.
步骤2:化合物II-1-3的合成Step 2: Synthesis of compound II-1-3
向反应管中加入II-1-2(80mg,194μmol,1eq)、2-氧代吲哚啉-6-硼酸(64mg,363μmol,2eq)、碳酸钠(41mg,387μmol,2eq)和2mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌6分钟,然后再向其中依次加入Pd(PPh3)2Cl2(14mg,19μmol,0.1eq)、tBuXphos(8mg,19μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-1-3(62mg)。LCMS(M+H):466。Add II-1-2 (80 mg, 194 μmol, 1 eq), 2-oxoindoline-6-boronic acid (64 mg, 363 μmol, 2 eq), sodium carbonate (41 mg, 387 μmol, 2 eq) and 2 mL of 1 to the reaction tube. , 4-dioxane, 0.2mL water, replace with nitrogen three times and stir for 6 minutes, then add Pd(PPh 3 ) 2 Cl 2 (14mg, 19μmol, 0.1eq), tBuXphos (8mg, 19μmol, 0.1) to it in sequence eq), replaced the nitrogen three times again, and then placed the reaction in a 100°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-1-3 (62 mg). LCMS(M+H):466.
步骤3:化合物II-1的合成Step 3: Synthesis of Compound II-1
向化合物II-1-3(10mg,273μmol,1eq)中加入二氯甲烷(1mL),再向其中加入三氟乙酸(0.1mL),然后于室温条件下搅拌3小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-1(3mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.92(s,1H),8.36(s,1H),7.51(dd,J=8.0,1.6Hz,1H),7.38–7.31(m,2H),4.24-4.17(m,1H),3.60(s,2H),3.56(dd,J=12.0,4.0Hz,1H),3.39–3.31(m,1H),3.21–3.05(m,2H),2.20–2.05(m,2H),1.93–1.73(m,2H);LCMS(M+H):366。Dichloromethane (1 mL) was added to compound II-1-3 (10 mg, 273 μmol, 1 eq), trifluoroacetic acid (0.1 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-1 (3 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.92 (s, 1H), 8.36 (s, 1H), 7.51 (dd, J = 8.0, 1.6Hz, 1H), 7.38–7.31 (m, 2H), 4.24- 4.17(m,1H),3.60(s,2H),3.56(dd,J=12.0,4.0Hz,1H),3.39–3.31(m,1H),3.21–3.05(m,2H),2.20–2.05( m,2H),1.93–1.73(m,2H); LCMS(M+H):366.
实施例32、化合物II-2的合成与表征
Example 32. Synthesis and characterization of compound II-2
步骤1:化合物II-2-2的合成Step 1: Synthesis of compound II-2-2
向反应瓶中加入II-2-1(100mg,403μmol,1eq)、Bpin2(250mg,1mmol,2.5eq)、无水乙酸钾(120mg,1.2mmol,3eq)和Pd(dppf)Cl2(50mg,68μmol,0.17eq),再加入1.5mL的无水1,4-二氧六环,置换氮气后将反应置于100℃油浴中搅拌10小时。待 反应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-2-2(83mg)。LCMS(M+H):296。Add II-2-1 (100mg, 403μmol, 1eq), Bpin 2 (250mg, 1mmol, 2.5eq), anhydrous potassium acetate (120mg, 1.2mmol, 3eq) and Pd(dppf)Cl 2 (50mg) to the reaction flask. , 68 μmol, 0.17 eq), then add 1.5 mL of anhydrous 1,4-dioxane, replace the nitrogen, and place the reaction in a 100°C oil bath and stir for 10 hours. treat When the reaction is complete, add the reaction solution to 20 mL of water, then extract with ethyl acetate (10 mL*3), combine the organic phases, and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain Yellow oil II-2-2 (83 mg). LCMS(M+H):296.
步骤2:化合物II-2-3的合成Step 2: Synthesis of Compound II-2-3
向反应管中加入II-2-2(80mg,271μmol,2.2eq)、II-1-2(50mg,121μmol,1eq)、碳酸钠(52mg,495μmol,4eq)和2mL的1,4-二氧六环、0.25mL水,氮气置换三次后搅拌6分钟,然后再向其中依次加入Pd(PPh3)2Cl2(9mg,12μmol,0.1eq)、tBuXphos(5mg,12μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-2-3(31mg)。LCMS(M+H):502。Add II-2-2 (80mg, 271μmol, 2.2eq), II-1-2 (50mg, 121μmol, 1eq), sodium carbonate (52mg, 495μmol, 4eq) and 2mL of 1,4-dioxygen to the reaction tube. Six rings, 0.25 mL water, replaced with nitrogen three times, stirred for 6 minutes, then added Pd(PPh 3 ) 2 Cl 2 (9 mg, 12 μmol, 0.1 eq), tBuXphos (5 mg, 12 μmol, 0.1 eq), and replaced again. Nitrogen was added three times, and the reaction was stirred in a 100 °C oil bath for 10 h. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-2-3 (31 mg). LCMS(M+H):502.
步骤3:化合物II-2的合成Step 3: Synthesis of Compound II-2
向化合物II-2-3(31mg,62μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-2(11mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.79(d,J=4.0Hz,1H),8.33(s,1H),7.56(d,J=8.0Hz,1H),7.46(d,J=8.0Hz,1H),7.17(s,1H),4.13-4.08(m,1H),3.51(dd,J=12.0,4.0Hz,1H),3.40–3.31(m,1H),3.13-3.07(m,2H),2.13-2.06(m,2H),1.88-1.82(m,1H),1.78-1.70(m,1H);19F NMR(376MHz,Deuterium Oxide)δ=-112.01;LCMS(M+H):402。Dichloromethane (2 mL) was added to compound II-2-3 (31 mg, 62 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-2 (11 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.79 (d, J = 4.0Hz, 1H), 8.33 (s, 1H), 7.56 (d, J = 8.0Hz, 1H), 7.46 (d, J = 8.0Hz ,1H),7.17(s,1H),4.13-4.08(m,1H),3.51(dd,J=12.0,4.0Hz,1H),3.40–3.31(m,1H),3.13-3.07(m,2H ), 2.13-2.06 (m, 2H), 1.88-1.82 (m, 1H), 1.78-1.70 (m, 1H); 19 F NMR (376MHz, Deuterium Oxide) δ = -112.01; LCMS (M+H): 402.
实施例33、化合物II-3的合成与表征
Example 33. Synthesis and characterization of compound II-3
步骤1:化合物II-3-2的合成Step 1: Synthesis of compound II-3-2
向反应瓶中加入II-3-1(100mg,420μmol,1eq)、Bpin2(270mg,1.1mmol,2.5eq)、 无水乙酸钾(120mg,1.2mmol,3eq)和Pd(dppf)Cl2(60mg,82μmol,0.2eq),再加入1mL的无水1,4-二氧六环,置换氮气后将反应置于100℃油浴中搅拌10小时。待反应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-3-2(85mg)。LCMS(M+H):286。Add II-3-1 (100mg, 420μmol, 1eq), Bpin 2 (270mg, 1.1mmol, 2.5eq), Anhydrous potassium acetate (120mg, 1.2mmol, 3eq) and Pd(dppf)Cl 2 (60mg, 82μmol, 0.2eq), then add 1mL of anhydrous 1,4-dioxane, replace nitrogen and place the reaction at Stir in an oil bath at 100°C for 10 hours. When the reaction is complete, add the reaction solution to 20 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A yellow oily substance II-3-2 (85 mg) was obtained. LCMS(M+H):286.
步骤2:化合物II-3-3的合成Step 2: Synthesis of Compound II-3-3
向反应管中加入II-3-2(80mg,280μmol,2.2eq)、II-1-2(60mg,145μmol,1eq)、碳酸钠(62mg,585μmol,4eq)和2mL的1,4-二氧六环、0.25mL水,氮气置换三次后搅拌6分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-3-3(45mg)。LCMS(M+H):492。Add II-3-2 (80mg, 280μmol, 2.2eq), II-1-2 (60mg, 145μmol, 1eq), sodium carbonate (62mg, 585μmol, 4eq) and 2mL of 1,4-dioxygen to the reaction tube. Six rings, 0.25mL water, replaced with nitrogen three times, stirred for 6 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10mg, 14μmol, 0.1eq), tBuXphos (6mg, 14μmol, 0.1eq), and replaced again. Nitrogen was added three times, and the reaction was stirred in a 100 °C oil bath for 10 h. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-3-3 (45 mg). LCMS(M+H):492.
步骤3:化合物II-3的合成Step 3: Synthesis of Compound II-3
向化合物II-3-3(45mg,92μmol,1eq)中加入二氯甲烷(3mL),再向其中加入三氟乙酸(0.3mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-3(28mg)。LCMS(M+H):392。Dichloromethane (3 mL) was added to compound II-3-3 (45 mg, 92 μmol, 1 eq), trifluoroacetic acid (0.3 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-3 (28 mg). LCMS(M+H):392.
实施例34、化合物II-4的合成与表征
Example 34. Synthesis and characterization of compound II-4
步骤1:化合物II-4-2的合成Step 1: Synthesis of compound II-4-2
向反应瓶中加入II-4-1(100mg,416μmol,1eq)、Bpin2(270mg,1.1mmol,2.5eq)、无水乙酸钾(120mg,1.2mmol,3eq)和Pd(dppf)Cl2(60mg,82μmol,0.2eq),再加入1mL的无水1,4-二氧六环,置换氮气后将反应置于100℃油浴中搅拌10小时。待反 应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-4-2(100mg)。LCMS(M+H):288。Add II-4-1 (100mg, 416μmol, 1eq), Bpin 2 (270mg, 1.1mmol, 2.5eq), anhydrous potassium acetate (120mg, 1.2mmol, 3eq) and Pd(dppf)Cl 2 ( 60 mg, 82 μmol, 0.2 eq), then add 1 mL of anhydrous 1,4-dioxane, replace the nitrogen, and place the reaction in a 100°C oil bath and stir for 10 hours. Wait to rebel If it should be complete, add the reaction solution to 20mL of water, then extract with ethyl acetate (10mL*3), combine the organic phases, wash with 25mL of saturated brine, and dry the resulting organic phase over anhydrous sodium sulfate, filter, and concentrate under reduced pressure to obtain Yellow oil II-4-2 (100mg). LCMS(M+H):288.
步骤2:化合物II-4-3的合成Step 2: Synthesis of compound II-4-3
向反应管中加入II-4-2(100mg,348μmol,1.8eq)、II-1-2(80mg,194μmol,1eq)、碳酸钠(70mg,660μmol,3.4eq)和2mL的1,4-二氧六环、0.25mL水,氮气置换三次后搅拌6分钟,然后再向其中依次加入Pd(PPh3)2Cl2(15mg,20μmol,0.1eq)、tBuXphos(8mg,19μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-4-3(51mg)。LCMS(M+H):494。Add II-4-2 (100mg, 348μmol, 1.8eq), II-1-2 (80mg, 194μmol, 1eq), sodium carbonate (70mg, 660μmol, 3.4eq) and 2mL of 1,4-bis to the reaction tube. Oxygen hexacyclic ring, 0.25mL water, replaced with nitrogen three times and stirred for 6 minutes, then added Pd(PPh 3 ) 2 Cl 2 (15mg, 20μmol, 0.1eq), tBuXphos (8mg, 19μmol, 0.1eq), and again The nitrogen was replaced three times, and the reaction was stirred in a 100°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-4-3 (51 mg). LCMS(M+H):494.
步骤3:化合物II-4的合成Step 3: Synthesis of Compound II-4
向化合物II-4-3(51mg,92μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-4(17mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.88(s,1H),8.35(s,1H),7.50(dd,J=8.0,4.0Hz,1H),7.39–7.31(m,2H),4.19-4.13(m,1H),3.50(dd,J=12.0,4.0Hz,1H),3.36–3.27(m,1H),3.17–3.02(m,2H),2.16–2.00(m,2H),1.88–1.69(m,2H),1.34(s,6H);LCMS(M+H):394。Dichloromethane (2 mL) was added to compound II-4-3 (51 mg, 92 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-4 (17 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.88 (s, 1H), 8.35 (s, 1H), 7.50 (dd, J = 8.0, 4.0Hz, 1H), 7.39–7.31 (m, 2H), 4.19- 4.13(m,1H),3.50(dd,J=12.0,4.0Hz,1H),3.36–3.27(m,1H),3.17–3.02(m,2H),2.16–2.00(m,2H),1.88– 1.69(m,2H),1.34(s,6H); LCMS(M+H):394.
实施例35、化合物II-5的合成与表征
Example 35. Synthesis and characterization of compound II-5
步骤1:化合物II-5-1的合成Step 1: Synthesis of Compound II-5-1
向反应管中加入3-甲基亚磺酰氨基苯硼酸(100mg,465μmol,1.1eq)、II-1-1(100 mg,402μmol,1eq)、碳酸钠(85mg,0.8mmol,2eq)和3mL的1,4-二氧六环、0.4mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(28mg,40μmol,0.1eq)、tBuXphos(17mg,40μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-5-1(90mg)。LCMS(M+H):340。Add 3-methylsulfenamidobenzeneboronic acid (100mg, 465μmol, 1.1eq), II-1-1 (100 mg, 402 μmol, 1 eq), sodium carbonate (85 mg, 0.8 mmol, 2 eq), 3 mL of 1,4-dioxane, and 0.4 mL of water. Replace nitrogen three times and stir for 5 minutes. Then add Pd(PPh) to them in sequence. 3 ) 2 Cl 2 (28 mg, 40 μmol, 0.1 eq), tBuXphos (17 mg, 40 μmol, 0.1 eq), replace nitrogen three times again, and then place the reaction in a 100°C oil bath and stir for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-5-1 (90 mg). LCMS(M+H):340.
步骤2:化合物II-5-2的合成Step 2: Synthesis of Compound II-5-2
向封管中加入II-5-1(90mg,265μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入3-氨基哌啶-1-羧酸叔丁酯(110mg,0.55mmol,2eq)和N,N-二异丙基乙胺(100mg,0.78mmol,3eq),然后置于140℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到30mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物II-5-2(33mg)。LCMS(M+H):504。Add II-5-1 (90 mg, 265 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add 3-aminopiperidine-1-carboxylic acid tert-butyl ester (110 mg, 0.55 mmol, 2 eq) respectively. ) and N,N-diisopropylethylamine (100 mg, 0.78 mmol, 3 eq), then placed in a 140°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 30mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oil II-5-2 (33 mg). LCMS(M+H):504.
步骤3:化合物II-5的合成Step 3: Synthesis of Compound II-5
向化合物II-5-2(10mg,20μmol,1eq)中加入二氯甲烷(1mL),再向其中加入三氟乙酸(0.1mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(FA)分离纯化得到黄色固体II-5(17mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.95(s,1H),8.45(s,1H),7.93(t,J=2.0Hz,1H),7.72(d,J=8.0Hz,1H),7.55(t,J=8.0Hz,1H),7.29(ddd,J=8.0,2.0,1.0Hz,1H),4.33-4.28(m,1H),3.57(dd,J=12.0,4.0Hz,1H),3.38-3.35(m,1H),3.20–3.04(m,5H),2.18–2.04(m,2H),1.94-1.85(m,1H),1.81-1.74(m,1H);LCMS(M+H):404。Dichloromethane (1 mL) was added to compound II-5-2 (10 mg, 20 μmol, 1 eq), trifluoroacetic acid (0.1 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain yellow solid II-5 (17 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.95 (s, 1H), 8.45 (s, 1H), 7.93 (t, J = 2.0Hz, 1H), 7.72 (d, J = 8.0Hz, 1H), 7.55 (t,J=8.0Hz,1H),7.29(ddd,J=8.0,2.0,1.0Hz,1H),4.33-4.28(m,1H),3.57(dd,J=12.0,4.0Hz,1H), 3.38-3.35(m,1H),3.20–3.04(m,5H),2.18–2.04(m,2H),1.94-1.85(m,1H),1.81-1.74(m,1H); LCMS(M+H) ):404.
实施例36、化合物II-6的合成与表征
Example 36. Synthesis and characterization of compound II-6
步骤1:化合物II-6-1的合成 Step 1: Synthesis of Compound II-6-1
向反应管中加入II-1-2(60mg,145μmol,1eq)、3-氨基苯硼酸(40mg,292μmol,2eq)、碳酸钠(41mg,387μmol,2eq)和2mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌6分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-6-1(61mg)。LCMS(M+H):426。Add II-1-2 (60 mg, 145 μmol, 1 eq), 3-aminophenylboronic acid (40 mg, 292 μmol, 2 eq), sodium carbonate (41 mg, 387 μmol, 2 eq) and 2 mL of 1,4-dioxane to the reaction tube. Ring, 0.2mL water, replaced with nitrogen three times and stirred for 6 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10mg, 14μmol, 0.1eq) and tBuXphos (6mg, 14μmol, 0.1eq), and replaced with nitrogen again. three times, then the reaction was stirred in a 100°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-6-1 (61 mg). LCMS(M+H):426.
步骤2:化合物II-6-2的合成Step 2: Synthesis of compound II-6-2
向反应管中加入II-6-1(60mg,141μmol,1eq)、N,N-二异丙基乙胺(36mg,279μmol,2eq)和0.5mL的无水四氢呋喃,然后再向其中滴加焦碳酸二甲酯(21mg,157μmol,1.1eq)的无水四氢呋喃溶液(0.5mL),将反应置于室温搅拌5小时。待反应完全,将反应液加入到15mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用15mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-6-2(51mg)。LCMS(M+H):484。Add II-6-1 (60 mg, 141 μmol, 1 eq), N, N-diisopropylethylamine (36 mg, 279 μmol, 2 eq) and 0.5 mL of anhydrous tetrahydrofuran to the reaction tube, and then add coke dropwise to it. A solution of dimethyl carbonate (21 mg, 157 μmol, 1.1 eq) in anhydrous tetrahydrofuran (0.5 mL) was stirred at room temperature for 5 hours. When the reaction is complete, add the reaction solution to 15 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 15 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A yellow oily substance II-6-2 (51 mg) was obtained. LCMS(M+H):484.
步骤3:化合物II-6的合成Step 3: Synthesis of Compound II-6
向化合物II-6-2(25mg,52μmol,1eq)中加入二氯甲烷(1mL),再向其中加入三氟乙酸(0.1mL),然后于室温条件下搅拌1小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-1(11mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.28(s,2H),8.00(s,1H),7.64(s,2H),7.48(s,2H),7.03(d,J=8.0Hz,2H),6.92(t,J=8.0Hz,2H),6.70(dd,J=8.0,1.2Hz,2H),3.62-3.56(m,2H),2.99(dd,J=12.0,4.0Hz,2H),2.87-2.84(m,2H),2.58–2.51(m,2H),2.46-2.40(m,2H),1.66–1.55(m,4H),1.38-1.31(m,2H),1.26–1.16(m,2H);LCMS(M+H):384。Dichloromethane (1 mL) was added to compound II-6-2 (25 mg, 52 μmol, 1 eq), trifluoroacetic acid (0.1 mL) was added thereto, and the mixture was stirred at room temperature for 1 hour. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-1 (11 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.28 (s, 2H), 8.00 (s, 1H), 7.64 (s, 2H), 7.48 (s, 2H), 7.03 (d, J = 8.0Hz, 2H) ,6.92(t,J=8.0Hz,2H),6.70(dd,J=8.0,1.2Hz,2H),3.62-3.56(m,2H),2.99(dd,J=12.0,4.0Hz,2H), 2.87-2.84(m,2H),2.58–2.51(m,2H),2.46-2.40(m,2H),1.66–1.55(m,4H),1.38-1.31(m,2H),1.26–1.16(m ,2H); LCMS(M+H):384.
实施例37、化合物II-7的合成与表征
Example 37. Synthesis and characterization of compound II-7
步骤1:化合物II-7-1的合成Step 1: Synthesis of compound II-7-1
向反应管中加入II-1-2(90mg,218μmol,1eq)、4-氨基苯硼酸频那醇酯(95mg,434μmol,2eq)、碳酸钠(70mg,660μmol,3eq)和1.5mL的1,4-二氧六环、0.3mL水,氮气置换三次后搅拌6分钟,然后再向其中依次加入Pd(PPh3)2Cl2(15mg,21μmol,0.1eq)、tBuXphos(10mg,23μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-7-1(61mg)。LCMS(M+H):426。Add II-1-2 (90mg, 218μmol, 1eq), 4-aminophenylboronic acid pinacol ester (95mg, 434μmol, 2eq), sodium carbonate (70mg, 660μmol, 3eq) and 1.5mL of 1, 4-Dioxane, 0.3mL water, replace with nitrogen three times and stir for 6 minutes, then add Pd(PPh 3 ) 2 Cl 2 (15mg, 21μmol, 0.1eq), tBuXphos (10mg, 23μmol, 0.1eq) to it in sequence ), replace nitrogen three times again, and then place the reaction in a 100°C oil bath and stir for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-7-1 (61 mg). LCMS(M+H):426.
步骤2:化合物II-7-2的合成Step 2: Synthesis of compound II-7-2
向反应管中加入II-7-1(40mg,94μmol,1eq)、N,N-二异丙基乙胺(36mg,279μmol,3eq)和0.5mL的无水四氢呋喃,然后再向其中滴加焦碳酸二甲酯(15mg,112μmol,1.2eq)的无水四氢呋喃溶液(0.3mL),将反应置于室温搅拌10小时。待反应完全,将反应液加入到15mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用15mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-7-2(36mg)。LCMS(M+H):484。Add II-7-1 (40 mg, 94 μmol, 1 eq), N, N-diisopropylethylamine (36 mg, 279 μmol, 3 eq) and 0.5 mL of anhydrous tetrahydrofuran to the reaction tube, and then add coke dropwise to it. A solution of dimethyl carbonate (15 mg, 112 μmol, 1.2 eq) in anhydrous tetrahydrofuran (0.3 mL) was stirred at room temperature for 10 hours. When the reaction is complete, add the reaction solution to 15 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 15 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A yellow oily substance II-7-2 (36 mg) was obtained. LCMS(M+H):484.
步骤3:化合物II-7的合成Step 3: Synthesis of Compound II-7
向化合物II-7-2(30mg,52μmol,1eq)中加入二氯甲烷(1mL),再向其中加入三氟乙酸(0.1mL),然后于室温条件下搅拌1小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-7(13mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.88(s,1H),8.31(s,1H),7.79(d,J=8.0Hz,2H),7.43(d,J=8.0Hz,2H),4.19–4.11(m,1H),3.77(s,3H),3.52(dd,J=12.0,4.0Hz,1H),3.32(d,J=12.0Hz,1H),3.11–3.01(m,2H),2.11-2.03(m,2H),1.86–1.68(m,2H);LCMS(M+H):384。Dichloromethane (1 mL) was added to compound II-7-2 (30 mg, 52 μmol, 1 eq), trifluoroacetic acid (0.1 mL) was added thereto, and the mixture was stirred at room temperature for 1 hour. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-7 (13 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.88 (s, 1H), 8.31 (s, 1H), 7.79 (d, J = 8.0Hz, 2H), 7.43 (d, J = 8.0Hz, 2H), 4.19 –4.11(m,1H),3.77(s,3H),3.52(dd,J=12.0,4.0Hz,1H),3.32(d,J=12.0Hz,1H),3.11–3.01(m,2H), 2.11-2.03(m,2H),1.86–1.68(m,2H); LCMS(M+H):384.
实施例38、化合物II-8的合成与表征
Example 38. Synthesis and characterization of compound II-8
步骤1:化合物II-8-1的合成Step 1: Synthesis of Compound II-8-1
向反应管中加入II-1-2(60mg,145μmol,1eq)、3-乙酰氨基苯硼酸(52mg,291μmol,2eq)、碳酸钠(46mg,434μmol,3eq)和1.5mL的1,4-二氧六环、0.3mL水,氮气置换三次后搅拌6分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃ 油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-8-1(31mg)。LCMS(M+H):468。Add II-1-2 (60 mg, 145 μmol, 1 eq), 3-acetylaminobenzene boronic acid (52 mg, 291 μmol, 2 eq), sodium carbonate (46 mg, 434 μmol, 3 eq) and 1.5 mL of 1,4-dibenzoic acid into the reaction tube. Oxygen hexacyclic ring, 0.3mL water, replaced with nitrogen three times and stirred for 6 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10mg, 14μmol, 0.1eq), tBuXphos (6mg, 14μmol, 0.1eq), and again Replace nitrogen three times, then place the reaction at 100°C Stir in oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-8-1 (31 mg). LCMS(M+H):468.
步骤2:化合物II-8的合成Step 2: Synthesis of Compound II-8
向化合物II-8-1(31mg,66μmol,1eq)中加入二氯甲烷(1mL),再向其中加入三氟乙酸(0.1mL),然后于室温条件下搅拌1小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-8(13mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.81(s,1H),8.45(s,1H),8.19(s,1H),7.51(d,J=8.0Hz,1H),7.40(t,J=8.0Hz,1H),7.13(d,J=8.0Hz,1H),4.26–4.16(m,1H),3.44–3.34(m,2H),3.01-2.95(m,1H),2.87(dd,J=12.0,8.0Hz,1H),2.18(s,3H),2.08-2.04(m,2H),1.84–1.64(m,2H);LCMS(M+H):368。Dichloromethane (1 mL) was added to compound II-8-1 (31 mg, 66 μmol, 1 eq), trifluoroacetic acid (0.1 mL) was added thereto, and the mixture was stirred at room temperature for 1 hour. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-8 (13 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.81 (s, 1H), 8.45 (s, 1H), 8.19 (s, 1H), 7.51 (d, J = 8.0Hz, 1H), 7.40 (t, J = 8.0Hz,1H),7.13(d,J=8.0Hz,1H),4.26–4.16(m,1H),3.44–3.34(m,2H),3.01-2.95(m,1H),2.87(dd,J =12.0,8.0Hz,1H),2.18(s,3H),2.08-2.04(m,2H),1.84–1.64(m,2H); LCMS(M+H):368.
实施例39、化合物II-9的合成与表征
Example 39. Synthesis and characterization of compound II-9
步骤1:化合物II-9-1的合成Step 1: Synthesis of Compound II-9-1
向反应管中加入3-磺酰叔丁胺基苯硼酸(124mg,481μmol,1.2eq)、II-1-1(100mg,402μmol,1eq)、碳酸钠(85mg,0.8mmol,2eq)和3mL的1,4-二氧六环、0.4mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(28mg,40μmol,0.1eq)、tBuXphos(17mg,40μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-9-1(119mg)。LCMS(M+H):382。Add 3-sulfonyl tert-butylaminophenylboronic acid (124 mg, 481 μmol, 1.2 eq), II-1-1 (100 mg, 402 μmol, 1 eq), sodium carbonate (85 mg, 0.8 mmol, 2 eq) and 3 mL of 1 to the reaction tube. 4-Dioxane, 0.4mL water, replace with nitrogen three times and stir for 5 minutes, then add Pd(PPh 3 ) 2 Cl 2 (28mg, 40μmol, 0.1eq), tBuXphos (17mg, 40μmol, 0.1eq) to it in sequence ), replace nitrogen three times again, and then place the reaction in a 100°C oil bath and stir for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain yellow oil II-9-1 (119 mg). LCMS(M+H):382.
步骤2:化合物II-9-2的合成 Step 2: Synthesis of compound II-9-2
向封管中加入II-9-1(110mg,288μmol,1eq),加入3mL的N-甲基吡咯烷酮,再分别加入3-氨基哌啶-1-羧酸叔丁酯(231mg,1.15mmol,4eq)和N,N-二异丙基乙胺(149mg,1.15mmol,4eq),然后置于140℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到30mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物II-9-2(88mg)。LCMS(M+H):546。Add II-9-1 (110 mg, 288 μmol, 1 eq) to the sealed tube, add 3 mL of N-methylpyrrolidone, and then add 3-aminopiperidine-1-carboxylic acid tert-butyl ester (231 mg, 1.15 mmol, 4 eq) respectively. ) and N,N-diisopropylethylamine (149 mg, 1.15 mmol, 4 eq), then placed in a 140°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 30mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oil II-9-2 (88 mg). LCMS(M+H):546.
步骤3:化合物II-9的合成Step 3: Synthesis of Compound II-9
向化合物II-9-2(20mg,37μmol,1eq)中加入二氯甲烷(1mL),再向其中加入三氟乙酸(0.5mL),然后于室温条件下搅拌24小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(FA)分离纯化得到黄色固体II-9(7mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.85(s,1H),8.68(s,1H),8.44(s,1H),7.97(d,J=8.0Hz,1H),7.90(dd,J=8.0,2.0Hz,1H),7.67(t,J=8.0Hz,1H),4.16-4.11(m,1H),3.61(dd,J=12.0,4.0Hz,1H),3.41-3.34(m,1H),3.12-3.05(m,2H),2.17-2.05(m,2H),1.94–1.82(m,1H),1.81–1.71(m,1H);LCMS(M+H):390。Dichloromethane (1 mL) was added to compound II-9-2 (20 mg, 37 μmol, 1 eq), trifluoroacetic acid (0.5 mL) was added thereto, and the mixture was stirred at room temperature for 24 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain yellow solid II-9 (7 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.85 (s, 1H), 8.68 (s, 1H), 8.44 (s, 1H), 7.97 (d, J = 8.0Hz, 1H), 7.90 (dd, J = 8.0,2.0Hz,1H),7.67(t,J=8.0Hz,1H),4.16-4.11(m,1H),3.61(dd,J=12.0,4.0Hz,1H),3.41-3.34(m,1H ),3.12-3.05(m,2H),2.17-2.05(m,2H),1.94–1.82(m,1H),1.81–1.71(m,1H); LCMS(M+H):390.
实施例40、化合物II-10的合成与表征
Example 40. Synthesis and characterization of compound II-10
步骤1:化合物II-10-2的合成Step 1: Synthesis of Compound II-10-2
向反应瓶中加入II-10-1(1g,4.9mmol,1eq)、碳酸铵(0.71g,7.4mmol,1.5eq)和50mL无水甲醇,待其完全溶解后加入醋酸碘苯(4g,12.4mmol,2.5eq),将反应置于室温下搅拌10小时。待反应完全,将反应液加入到100mL饱和食盐水中,然后用乙酸乙酯萃取(50mL*3),合并有机相,分别用60mL碳酸氢钠饱和溶液和60mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物II-10-2(0.8g)。LCMS(M+H):234。Add II-10-1 (1g, 4.9mmol, 1eq), ammonium carbonate (0.71g, 7.4mmol, 1.5eq) and 50mL anhydrous methanol to the reaction bottle. After it is completely dissolved, add iodobenzene acetate (4g, 12.4 mmol, 2.5eq), and the reaction was stirred at room temperature for 10 hours. When the reaction is complete, add the reaction solution to 100 mL of saturated brine, and then extract with ethyl acetate (50 mL*3). Combine the organic phases and wash them with 60 mL of saturated sodium bicarbonate solution and 60 mL of saturated brine. The resulting organic phase is Dry over sodium sulfate, filter, and concentrate under reduced pressure to obtain light yellow oil II-10-2 (0.8g). LCMS(M+H):234.
步骤2:化合物II-10-3的合成 Step 2: Synthesis of Compound II-10-3
向反应瓶中加入II-10-2(100mg,427μmol,1eq)、Bpin2(220mg,0.87mmol,2eq)、无水乙酸钾(130mg,1.3mmol,3eq)和Pd(dppf)Cl2(60mg,82μmol,0.2eq),再加入1mL的无水1,4-二氧六环,置换氮气后将反应置于100℃油浴中搅拌10小时。待反应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-10-3(70mg)。LCMS(M+H):282。Add II-10-2 (100mg, 427μmol, 1eq), Bpin 2 (220mg, 0.87mmol, 2eq), anhydrous potassium acetate (130mg, 1.3mmol, 3eq) and Pd(dppf)Cl 2 (60mg) to the reaction flask. , 82 μmol, 0.2 eq), then add 1 mL of anhydrous 1,4-dioxane, replace the nitrogen, and place the reaction in a 100°C oil bath and stir for 10 hours. When the reaction is complete, add the reaction solution to 20 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A yellow oily substance II-10-3 (70 mg) was obtained. LCMS(M+H):282.
步骤3:化合物II-10-4的合成Step 3: Synthesis of Compound II-10-4
向反应管中加入II-10-3(70mg,249μmol,2eq)、II-1-2(50mg,121μmol,1eq)、碳酸钠(52mg,490μmol,4eq)和2mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌6分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-10-4(34mg)。LCMS(M+H):488。Add II-10-3 (70mg, 249μmol, 2eq), II-1-2 (50mg, 121μmol, 1eq), sodium carbonate (52mg, 490μmol, 4eq) and 2mL of 1,4-dioxane to the reaction tube. Ring, 0.2mL water, replaced with nitrogen three times and stirred for 6 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10mg, 14μmol, 0.1eq) and tBuXphos (6mg, 14μmol, 0.1eq), and replaced with nitrogen again. three times, then the reaction was stirred in a 100°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-10-4 (34 mg). LCMS(M+H):488.
步骤4:化合物II-10的合成Step 4: Synthesis of Compound II-10
向化合物II-10-4(34mg,70μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-10(13mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.98(d,J=2.4Hz,1H),8.85(d,J=56.0Hz,1H),8.65(d,J=12.0Hz,1H),8.25(t,J=8.0Hz,1H),8.08–8.03(m,1H),7.85-7.79(m,1H),4.34-4.23(m,1H),3.67–3.57(m,1H),3.54(s,3H),3.37-3.33(m,1H),3.14-3.05(m,2H),2.21–2.03(m,2H),1.93–1.76(m,2H);LCMS(M+H):388。Dichloromethane (2 mL) was added to compound II-10-4 (34 mg, 70 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-10 (13 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.98 (d, J = 2.4Hz, 1H), 8.85 (d, J = 56.0Hz, 1H), 8.65 (d, J = 12.0Hz, 1H), 8.25 (t ,J=8.0Hz,1H),8.08–8.03(m,1H),7.85-7.79(m,1H),4.34-4.23(m,1H),3.67–3.57(m,1H),3.54(s,3H ),3.37-3.33(m,1H),3.14-3.05(m,2H),2.21–2.03(m,2H),1.93–1.76(m,2H); LCMS(M+H):388.
实施例41、化合物II-11的合成与表征
Example 41. Synthesis and characterization of compound II-11
步骤1:化合物II-11-2的合成Step 1: Synthesis of Compound II-11-2
向反应瓶中加入II-11-1(200mg,0.67mmol,1eq)、二甲基氧化膦(63mg,0.81mmol,1.2eq)、Pd(dppf)Cl2(50mg,67μmol,0.1eq)和1mL无水N,N-二甲基甲酰胺,置换氮气后再向其中加入三乙胺(200mg,1.98mmol,3eq),将反应置于90℃油浴中搅拌20小时。待反应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物II-11-2(200mg)。LCMS(M+H):249。Add II-11-1 (200mg, 0.67mmol, 1eq), dimethylphosphine oxide (63mg, 0.81mmol, 1.2eq), Pd(dppf)Cl 2 (50mg, 67μmol, 0.1eq) and 1mL to the reaction flask. Anhydrous N,N-dimethylformamide was replaced with nitrogen, then triethylamine (200 mg, 1.98 mmol, 3 eq) was added, and the reaction was stirred in a 90°C oil bath for 20 hours. When the reaction is complete, add the reaction solution to 20 mL of water, and then extract with ethyl acetate (15 mL*3). Combine the organic phases and wash with 20 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A light yellow oily substance II-11-2 (200 mg) was obtained. LCMS(M+H):249.
步骤2:化合物II-11-3的合成Step 2: Synthesis of Compound II-11-3
向反应瓶中加入II-11-2(200mg,0.81mmol,1eq)、Bpin2(410mg,1.6mmol,2eq)、无水乙酸钾(250mg,2.6mmol,3eq)和Pd(dppf)Cl2(180mg,246μmol,0.3eq),再加入2mL的无水1,4-二氧六环,置换氮气后将反应置于80℃油浴中搅拌10小时。待反应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-11-3(170mg)。LCMS(M+H):296。Add II-11-2 (200mg, 0.81mmol, 1eq), Bpin 2 (410mg, 1.6mmol, 2eq), anhydrous potassium acetate (250mg, 2.6mmol, 3eq) and Pd(dppf)Cl 2 ( 180 mg, 246 μmol, 0.3 eq), and then add 2 mL of anhydrous 1,4-dioxane. After replacing nitrogen, the reaction was placed in an 80°C oil bath and stirred for 10 hours. When the reaction is complete, add the reaction solution to 20 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A yellow oily substance II-11-3 (170 mg) was obtained. LCMS(M+H):296.
步骤3:化合物II-11-4的合成Step 3: Synthesis of Compound II-11-4
向反应管中加入II-11-3(170mg,0.58mmol,1.2eq)、II-1-2(120mg,481μmol,1eq)、碳酸钠(153mg,1.4mmol,3eq)和5mL的1,4-二氧六环、0.7mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(35mg,48μmol,0.1eq)、tBuXphos(20mg,48μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-11-4(100mg)。LCMS(M+H):338。Add II-11-3 (170mg, 0.58mmol, 1.2eq), II-1-2 (120mg, 481μmol, 1eq), sodium carbonate (153mg, 1.4mmol, 3eq) and 5mL of 1,4- Dioxane and 0.7mL water were replaced with nitrogen three times and stirred for 5 minutes, then Pd(PPh 3 ) 2 Cl 2 (35mg, 48μmol, 0.1eq) and tBuXphos (20mg, 48μmol, 0.1eq) were added in sequence. The nitrogen was replaced three times again, and the reaction was stirred in a 100°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-11-4 (100 mg). LCMS(M+H):338.
步骤4:化合物II-11-5的合成Step 4: Synthesis of Compound II-11-5
向封管中加入II-11-4(100mg,296μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(S)-3-氨基哌啶-1-羧酸叔丁酯(178mg,0.89mmol,3eq)和N,N-二异丙基乙胺(230mg,1.78mmol,6eq),然后置于150℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物II-11-15(70mg)。LCMS(M+H):502。Add II-11-4 (100 mg, 296 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (S)-3-aminopiperidine-1-carboxylic acid tert-butyl ester (178 mg, 0.89mmol, 3eq) and N,N-diisopropylethylamine (230mg, 1.78mmol, 6eq), and then placed in a 150°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain light yellow oil II-11-15 (70 mg). LCMS(M+H):502.
步骤5:化合物II-11的合成Step 5: Synthesis of Compound II-11
向化合物II-11-5(70mg,140μmol,1eq)中加入乙酸乙酯(4mL),再向其中加入4N盐酸乙酸乙酯溶液(2mL),然后于室温条件下搅拌8小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-11(13mg)。1H NMR(400MHz,Methanol-d4)δ=9.21(s,1H),8.60(s,1H),7.66(s,2H),7.31(s, 1H),4.28(s,1H),3.61(dd,J=12.0,4.0Hz,1H),3.11–2.99(m,2H),2.26–2.03(m,3H),1.97(dd,J=12.0,8.0Hz,6H),1.92–1.73(m,3H),1.34-1.29(m,2H);LCMS(M+H):402。Ethyl acetate (4 mL) was added to compound II-11-5 (70 mg, 140 μmol, 1 eq), and 4N hydrochloric acid ethyl acetate solution (2 mL) was added thereto, followed by stirring at room temperature for 8 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-11 (13 mg). 1 H NMR (400MHz, Methanol-d 4 ) δ = 9.21 (s, 1H), 8.60 (s, 1H), 7.66 (s, 2H), 7.31 (s, 1H),4.28(s,1H),3.61(dd,J=12.0,4.0Hz,1H),3.11–2.99(m,2H),2.26–2.03(m,3H),1.97(dd,J=12.0, 8.0Hz,6H),1.92–1.73(m,3H),1.34-1.29(m,2H); LCMS(M+H):402.
实施例42、化合物II-12的合成与表征
Example 42. Synthesis and characterization of compound II-12
步骤1:化合物II-12-2的合成Step 1: Synthesis of Compound II-12-2
向反应瓶中加入II-12-1(0.5g,1.67mmol,1eq)、二甲基氧化膦(160mg,2.05mmol,1.2eq)、Pd(dppf)Cl2(120mg,164μmol,0.1eq)和5mL无水N,N-二甲基甲酰胺,置换氮气后再向其中加入三乙胺(0.51g,5.05mmol,3eq),将反应置于90℃油浴中搅拌10小时。待反应完全,将反应液加入到50mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用30mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物II-12-2(380mg)。LCMS(M+H):249。Add II-12-1 (0.5g, 1.67mmol, 1eq), dimethylphosphine oxide (160mg, 2.05mmol, 1.2eq), Pd(dppf)Cl 2 (120mg, 164μmol, 0.1eq) and 5 mL of anhydrous N,N-dimethylformamide, replaced with nitrogen, then added triethylamine (0.51g, 5.05mmol, 3eq), and stirred the reaction in a 90°C oil bath for 10 hours. When the reaction is complete, add the reaction solution to 50 mL of water, and then extract with ethyl acetate (15 mL*3). Combine the organic phases and wash with 30 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A light yellow oily substance II-12-2 (380 mg) was obtained. LCMS(M+H):249.
步骤2:化合物II-12-3的合成Step 2: Synthesis of Compound II-12-3
向反应瓶中加入II-12-2(380mg,1.5mmol,1eq)、Bpin2(1g,3.9mmol,2.5eq)、无水乙酸钾(450mg,4.5mmol,3eq)和Pd(dppf)Cl2(200mg,273μmol,0.18eq),再加入5mL的无水1,4-二氧六环,置换氮气后将反应置于100℃油浴中搅拌10小时。待反应完全,将反应液加入到30mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-12-3(280mg)。LCMS(M+H):296。Add II-12-2 (380mg, 1.5mmol, 1eq), Bpin 2 (1g, 3.9mmol, 2.5eq), anhydrous potassium acetate (450mg, 4.5mmol, 3eq) and Pd(dppf)Cl 2 to the reaction flask. (200 mg, 273 μmol, 0.18 eq), and then add 5 mL of anhydrous 1,4-dioxane. After replacing nitrogen, the reaction was placed in a 100°C oil bath and stirred for 10 hours. When the reaction is complete, add the reaction solution to 30 mL of water, and then extract with ethyl acetate (15 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A yellow oily substance II-12-3 (280 mg) was obtained. LCMS(M+H):296.
步骤3:化合物II-12-4的合成Step 3: Synthesis of Compound II-12-4
向反应管中加入II-12-3(86mg,292μmol,2eq)、II-1-2(60mg,145μmol,1eq)、碳酸钠(62mg,590μmol,4eq)和2mL的1,4-二氧六环、0.3mL水,氮气置换三次后搅拌6分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙 酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-12-4(28mg)。LCMS(M+H):502。Add II-12-3 (86mg, 292μmol, 2eq), II-1-2 (60mg, 145μmol, 1eq), sodium carbonate (62mg, 590μmol, 4eq) and 2mL of 1,4-dioxane to the reaction tube. Ring, 0.3mL water, replaced with nitrogen three times and stirred for 6 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10mg, 14μmol, 0.1eq) and tBuXphos (6mg, 14μmol, 0.1eq), and replaced with nitrogen again. three times, then the reaction was stirred in a 100°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20 mL of water, and then add it with ethyl acetate. Ester extraction (20mL*3), combine the organic phases, wash with 20mL saturated brine, dry the obtained organic phase over anhydrous sodium sulfate, filter, and concentrate under reduced pressure to obtain the crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-12-4 (28 mg). LCMS(M+H):502.
步骤4:化合物II-12的合成Step 4: Synthesis of Compound II-12
向化合物II-12-4(28mg,56μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-12(13mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.95(s,1H),8.50(s,1H),7.55–7.44(m,3H),4.28-4.22(m,1H),3.55(dd,J=12.0,4.0Hz,1H),3.36–3.29(m,1H),3.15–3.02(m,2H),2.18–2.03(m,2H),1.91(d,J=12.0Hz,6H),1.88–1.71(m,2H);LCMS(M+H):402。Dichloromethane (2 mL) was added to compound II-12-4 (28 mg, 56 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-12 (13 mg). 1 H NMR (400MHz, Deuterium Oxide)δ=8.95(s,1H),8.50(s,1H),7.55–7.44(m,3H),4.28-4.22(m,1H),3.55(dd,J=12.0 ,4.0Hz,1H),3.36–3.29(m,1H),3.15–3.02(m,2H),2.18–2.03(m,2H),1.91(d,J=12.0Hz,6H),1.88–1.71( m,2H); LCMS(M+H):402.
实施例43、化合物II-13的合成与表征
Example 43. Synthesis and characterization of compound II-13
步骤1:化合物II-13-1的合成Step 1: Synthesis of Compound II-13-1
向反应管中加入II-1-2(80mg,194μmol,1eq)、2-(叔丁氧羰基氨基)吡啶-3-硼酸(70mg,294μmol,1.5eq)、碳酸钠(41mg,389μmol,2eq)和1.5mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(14mg,19μmol,0.1eq)、tBuXphos(8mg,19μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-13-1(21mg)。LCMS(M+H):427。Add II-1-2 (80 mg, 194 μmol, 1 eq), 2-(tert-butoxycarbonylamino)pyridine-3-boronic acid (70 mg, 294 μmol, 1.5 eq), and sodium carbonate (41 mg, 389 μmol, 2 eq) to the reaction tube. Add 1.5 mL of 1,4-dioxane and 0.2 mL of water, replace with nitrogen three times and stir for 5 minutes, then add Pd(PPh 3 ) 2 Cl 2 (14 mg, 19 μmol, 0.1 eq), tBuXphos ( 8 mg, 19 μmol, 0.1 eq), replace nitrogen three times again, and then place the reaction in a 100°C oil bath and stir for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-13-1 (21 mg). LCMS(M+H):427.
步骤2:化合物II-13的合成Step 2: Synthesis of Compound II-13
向化合物II-13-1(21mg,49μmol,1eq)中加入二氯甲烷(1mL),再向其中加入三氟乙酸(0.1mL),然后于室温条件下搅拌1小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(FA)分离纯化得到黄色固体II-13(3mg)。LCMS(M+H):326。Dichloromethane (1 mL) was added to compound II-13-1 (21 mg, 49 μmol, 1 eq), trifluoroacetic acid (0.1 mL) was added thereto, and the mixture was stirred at room temperature for 1 hour. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain yellow solid II-13 (3 mg). LCMS(M+H):326.
实施例44、化合物II-14的合成与表征
Example 44. Synthesis and characterization of compound II-14
步骤1:化合物II-14-1的合成Step 1: Synthesis of Compound II-14-1
向反应管中加入II-1-2(80mg,194μmol,1eq)、1-叔丁氧羰基吲哚-3-硼酸(101mg,387μmol,2eq)、碳酸钠(41mg,389μmol,2eq)和1.5mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(14mg,19μmol,0.1eq)、tBuXphos(8mg,19μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-14-1(30mg)。LCMS(M+H):450。Add II-1-2 (80mg, 194μmol, 1eq), 1-tert-butoxycarbonylindole-3-boronic acid (101mg, 387μmol, 2eq), sodium carbonate (41mg, 389μmol, 2eq) and 1.5mL to the reaction tube. 1,4-dioxane and 0.2mL water, replaced with nitrogen three times and stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (14mg, 19μmol, 0.1eq), tBuXphos (8mg, 19μmol) to it in sequence ,0.1eq), replace nitrogen three times again, and then place the reaction in a 100°C oil bath and stir for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-14-1 (30 mg). LCMS(M+H):450.
步骤2:化合物II-14的合成Step 2: Synthesis of Compound II-14
向化合物II-14-1(30mg,67μmol,1eq)中加入二氯甲烷(3mL),再向其中加入三氟乙酸(0.3mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(FA)分离纯化得到黄色固体II-14(12mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.77(s,1H),8.40(s,1H),7.96(s,1H),7.83(d,J=8.0Hz,1H),7.58(d,J=8.0Hz,1H),7.35–7.30(m,1H),7.25–7.20(m,1H),4.11-4.05(m,1H),3.49(dd,J=12.0,4.0Hz,1H),3.36–3.30(m,1H),3.07–2.98(m,2H),2.15–2.01(m,2H),1.88–1.67(m,2H);LCMS(M+H):350。Dichloromethane (3 mL) was added to compound II-14-1 (30 mg, 67 μmol, 1 eq), trifluoroacetic acid (0.3 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain yellow solid II-14 (12 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.77 (s, 1H), 8.40 (s, 1H), 7.96 (s, 1H), 7.83 (d, J = 8.0Hz, 1H), 7.58 (d, J = 8.0Hz,1H),7.35–7.30(m,1H),7.25–7.20(m,1H),4.11-4.05(m,1H),3.49(dd,J=12.0,4.0Hz,1H),3.36–3.30 (m,1H),3.07–2.98(m,2H),2.15–2.01(m,2H),1.88–1.67(m,2H); LCMS(M+H):350.
实施例45、化合物II-15的合成与表征
Example 45. Synthesis and characterization of compound II-15
步骤1:化合物II-15-1的合成Step 1: Synthesis of Compound II-15-1
向反应管中加入II-1-2(60mg,145μmol,1eq)、6-羟基吡啶-3-硼酸(25mg,180μmol,1.2eq)、碳酸钠(40mg,377μmol,2.5eq)和1mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机 相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-15-1(27mg)。LCMS(M+H):428。Add II-1-2 (60mg, 145μmol, 1eq), 6-hydroxypyridine-3-boronic acid (25mg, 180μmol, 1.2eq), sodium carbonate (40mg, 377μmol, 2.5eq) and 1mL of 1 to the reaction tube. 4-Dioxane, 0.2mL water, replace with nitrogen three times and stir for 5 minutes, then add Pd(PPh 3 ) 2 Cl 2 (10mg, 14μmol, 0.1eq), tBuXphos (6mg, 14μmol, 0.1eq) to it in sequence ), replace nitrogen three times again, and then place the reaction in a 100°C oil bath and stir for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic The phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-15-1 (27 mg). LCMS(M+H):428.
步骤2:化合物II-15的合成Step 2: Synthesis of Compound II-15
向化合物II-15-1(27mg,63μmol,1eq)中加入二氯甲烷(1mL),再向其中加入三氟乙酸(0.1mL),然后于室温条件下搅拌1小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(FA)分离纯化得到黄色固体II-15(10mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.75(s,1H),8.42(s,1H),8.30(d,J=4.0Hz,1H),8.04(s,1H),7.99(dd,J=12.0,2.4Hz,1H),6.61(d,J=8.0Hz,1H),4.06–3.99(m,1H),3.52(dd,J=12.0,4.0Hz,1H),3.37–3.30(m,1H),3.12–3.01(m,2H),2.17–2.05(m,2H),1.92–1.81(m,1H),1.78–1.69(m,1H);LCMS(M+H):328。Dichloromethane (1 mL) was added to compound II-15-1 (27 mg, 63 μmol, 1 eq), trifluoroacetic acid (0.1 mL) was added thereto, and the mixture was stirred at room temperature for 1 hour. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (FA) to obtain yellow solid II-15 (10 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.75 (s, 1H), 8.42 (s, 1H), 8.30 (d, J = 4.0Hz, 1H), 8.04 (s, 1H), 7.99 (dd, J = 12.0,2.4Hz,1H),6.61(d,J=8.0Hz,1H),4.06–3.99(m,1H),3.52(dd,J=12.0,4.0Hz,1H),3.37–3.30(m,1H ),3.12–3.01(m,2H),2.17–2.05(m,2H),1.92–1.81(m,1H),1.78–1.69(m,1H); LCMS(M+H):328.
实施例46、化合物II-16的合成与表征
Example 46. Synthesis and characterization of compound II-16
步骤1:化合物II-16-1的合成Step 1: Synthesis of Compound II-16-1
向反应管中加入II-1-2(60mg,145μmol,1eq)、3-甲基吲哚-6-硼酸频那醇酯(75mg,292μmol,2eq)、碳酸钠(62mg,585μmol,4eq)和1mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-16-1(31mg)。LCMS(M+H):464。Add II-1-2 (60mg, 145μmol, 1eq), 3-methylindole-6-boronic acid pinacol ester (75mg, 292μmol, 2eq), sodium carbonate (62mg, 585μmol, 4eq) and 1 mL of 1,4-dioxane and 0.2 mL of water were replaced with nitrogen three times and stirred for 5 minutes, then Pd(PPh 3 ) 2 Cl 2 (10 mg, 14 μmol, 0.1 eq), tBuXphos (6 mg, 14 μmol, 0.1 eq), replaced the nitrogen three times again, and then placed the reaction in a 100°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-16-1 (31 mg). LCMS(M+H):464.
步骤2:化合物II-16的合成Step 2: Synthesis of Compound II-16
向化合物II-16-1(27mg,63μmol,1eq)中加入乙酸乙酯(4mL),再向其中加入4N的盐酸乙酸乙酯溶液(4mL),然后于室温条件下搅拌5小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-16(9mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.90(s,1H),8.30(s,1H),7.90(s,1H),7.69(d,J=8.0Hz,1H),7.53(d,J=8.0Hz,1H),7.21(d,J=4.0Hz,1H),4.18–4.11(m,1H),3.53–3.48(m,1H),3.31-3.25(m,1H),3.15–3.00(m,3H),2.33(d,J=4.0Hz,3H),2.13–2.01(m,2H),1.85–1.71(m,2H);LCMS(M+H):364。Ethyl acetate (4 mL) was added to compound II-16-1 (27 mg, 63 μmol, 1 eq), 4N hydrochloric acid ethyl acetate solution (4 mL) was added thereto, and the mixture was stirred at room temperature for 5 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-16 (9 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.90 (s, 1H), 8.30 (s, 1H), 7.90 (s, 1H), 7.69 (d, J = 8.0Hz, 1H), 7.53 (d, J = 8.0Hz,1H),7.21(d,J=4.0Hz,1H),4.18–4.11(m,1H),3.53–3.48(m,1H),3.31-3.25(m,1H),3.15–3.00(m ,3H),2.33(d,J=4.0Hz,3H),2.13–2.01(m,2H),1.85–1.71(m,2H); LCMS(M+H):364.
实施47、化合物II-17的合成与表征
Implementation 47. Synthesis and characterization of compound II-17
步骤1:化合物II-17-2的合成Step 1: Synthesis of Compound II-17-2
向反应瓶中加入II-17-1(200mg,0.9mmol,1eq)、Bpin2(0.57g,2.2mmol,2.5eq)、无水乙酸钾(270mg,2.7mmol,3eq)和Pd(dppf)Cl2(130mg,178μmol,0.2eq),再加入3mL的无水1,4-二氧六环,置换氮气后将反应置于90℃油浴中搅拌10小时。待反应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(10mL*3),合并有机相,用25mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-17-2(170mg)。LCMS(M+H):269。Add II-17-1 (200mg, 0.9mmol, 1eq), Bpin 2 (0.57g, 2.2mmol, 2.5eq), anhydrous potassium acetate (270mg, 2.7mmol, 3eq) and Pd(dppf)Cl to the reaction flask. 2 (130 mg, 178 μmol, 0.2 eq), and then add 3 mL of anhydrous 1,4-dioxane. After replacing nitrogen, the reaction was placed in a 90°C oil bath and stirred for 10 hours. When the reaction is complete, add the reaction solution to 20 mL of water, and then extract with ethyl acetate (10 mL*3). Combine the organic phases and wash with 25 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A yellow oily substance II-17-2 (170 mg) was obtained. LCMS(M+H):269.
步骤2:化合物II-17-3的合成Step 2: Synthesis of Compound II-17-3
向反应管中加入II-17-2(80mg,298μmol,2eq)、II-1-2(60mg,145μmol,1eq)、碳酸钠(62mg,0.58mmol,4eq)和2mL的1,4-二氧六环、0.3mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-17-3(33mg)。LCMS(M+H):475。Add II-17-2 (80mg, 298μmol, 2eq), II-1-2 (60mg, 145μmol, 1eq), sodium carbonate (62mg, 0.58mmol, 4eq) and 2mL of 1,4-dioxygen to the reaction tube. Six rings, 0.3 mL water, replaced with nitrogen three times, stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10 mg, 14 μmol, 0.1 eq), tBuXphos (6 mg, 14 μmol, 0.1 eq), and replaced again. Nitrogen was added three times, and the reaction was stirred in a 100 °C oil bath for 10 h. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-17-3 (33 mg). LCMS(M+H):475.
步骤3:化合物II-17的合成Step 3: Synthesis of Compound II-17
向化合物II-17-3(33mg,88μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-17(12mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.78(s,1H),8.20(s,1H),7.94(s,1H),7.79(s,1H),7.60–7.51(m,2H),4.09-4.04(m,1H),3.48(dd,J=16.0,4.0Hz,1H),3.30(d,J=16.0Hz,1H),3.12-3.04(m, 2H),2.11-2.04(m,2H),1.88–1.70(m,2H);LCMS(M+H):375。Dichloromethane (2 mL) was added to compound II-17-3 (33 mg, 88 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-17 (12 mg). 1 H NMR (400MHz, Deuterium Oxide)δ=8.78(s,1H),8.20(s,1H),7.94(s,1H),7.79(s,1H),7.60–7.51(m,2H),4.09- 4.04(m,1H),3.48(dd,J=16.0,4.0Hz,1H),3.30(d,J=16.0Hz,1H),3.12-3.04(m, 2H), 2.11-2.04(m,2H),1.88–1.70(m,2H); LCMS(M+H):375.
实施例48、化合物II-18的合成与表征
Example 48. Synthesis and characterization of compound II-18
步骤1:化合物II-18-1的合成Step 1: Synthesis of Compound II-18-1
向反应管中加入II-1-2(60mg,145μmol,1eq)、3-甲酸乙酯-6-硼酸频那醇酯(92mg,292μmol,2eq)、碳酸钠(50mg,472μmol,3eq)和1mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-18-1(43mg)。LCMS(M+H):522。Add II-1-2 (60 mg, 145 μmol, 1 eq), 3-ethyl formate-6-boronic acid pinacol ester (92 mg, 292 μmol, 2 eq), sodium carbonate (50 mg, 472 μmol, 3 eq) and 1 mL to the reaction tube. 1,4-dioxane and 0.2mL water, replaced with nitrogen three times and stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10mg, 14μmol, 0.1eq), tBuXphos (6mg, 14μmol) to it in sequence ,0.1eq), replace nitrogen three times again, and then place the reaction in a 100°C oil bath and stir for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-18-1 (43 mg). LCMS(M+H):522.
步骤2:化合物II-18-2的合成Step 2: Synthesis of Compound II-18-2
向II-18-1(30mg,57μmol,1eq)中加入无水乙醇(0.5mL)和乙醇胺(0.5mL),然后于80℃油浴中搅拌10小时。待反应完全,将反应液直接减压浓缩得到黄色油状物II-18-2(30mg)。LCMS(M+H):537。Absolute ethanol (0.5 mL) and ethanolamine (0.5 mL) were added to II-18-1 (30 mg, 57 μmol, 1 eq), and then stirred in an 80°C oil bath for 10 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain yellow oil II-18-2 (30 mg). LCMS(M+H):537.
步骤2:化合物II-18的合成Step 2: Synthesis of Compound II-18
向化合物II-18-2(30mg,56μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-18(9mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.79(s,1H),8.23(s,1H),7.81(s,1H),7.65(d,J=8.0Hz,1H),7.46(dd,J=8.0,1.6Hz,1H),7.03(s,1H),4.04-3.99(m,1H),3.78(t,J=5.6Hz,2H),3.55(t,J=5.6Hz,2H),3.47(dd,J=12.0,4.0Hz,1H),3.29-3.25(m,1H),3.10–3.00(m,2H),2.09–1.97(m,2H),1.84–1.65(m,2H);LCMS(M+H):437。 Dichloromethane (2 mL) was added to compound II-18-2 (30 mg, 56 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-18 (9 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.79 (s, 1H), 8.23 (s, 1H), 7.81 (s, 1H), 7.65 (d, J = 8.0Hz, 1H), 7.46 (dd, J = 8.0,1.6Hz,1H),7.03(s,1H),4.04-3.99(m,1H),3.78(t,J=5.6Hz,2H),3.55(t,J=5.6Hz,2H),3.47( dd,J=12.0,4.0Hz,1H),3.29-3.25(m,1H),3.10–3.00(m,2H),2.09–1.97(m,2H),1.84–1.65(m,2H); LCMS( M+H):437.
实施例49、化合物II-19的合成与表征
Example 49. Synthesis and characterization of compound II-19
步骤1:化合物II-19-2的合成Step 1: Synthesis of Compound II-19-2
向反应瓶中加入II-19-1(200mg,0.72mmol,1eq)、碳酸铯(0.7g,2.15mmol,3eq)和1mL无水N,N-二甲基甲酰胺,然后再加入碘甲烷(200mg,1.41mmol,2eq),反应于室温条件下搅拌10小时。待反应完全,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用30mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物II-19-2(233mg)。LCMS(M+H):294。Add II-19-1 (200mg, 0.72mmol, 1eq), cesium carbonate (0.7g, 2.15mmol, 3eq) and 1mL anhydrous N,N-dimethylformamide to the reaction flask, then add methyl iodide ( 200mg, 1.41mmol, 2eq), the reaction was stirred at room temperature for 10 hours. When the reaction is complete, add the reaction solution to 20 mL of water, and then extract with ethyl acetate (15 mL*3). Combine the organic phases and wash with 30 mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A light yellow oily substance II-19-2 (233 mg) was obtained. LCMS(M+H):294.
步骤2:化合物II-19-3的合成Step 2: Synthesis of Compound II-19-3
向反应瓶中加入II-19-2(230mg,0.78mmol,1eq)、Bpin2(400mg,1.57mmol,2eq)、无水乙酸钾(230mg,2.3mmol,3eq)和Pd(dppf)Cl2(130mg,178μmol,0.2eq),再加入2mL的无水1,4-二氧六环,置换氮气后将反应置于85℃油浴中搅拌10小时。待反应完全,向反应液中加入10mL乙酸乙酯,过滤后经减压浓缩得到黑色油状物II-19-3(221mg)。LCMS(M+H):259。Add II-19-2 (230mg, 0.78mmol, 1eq), Bpin 2 (400mg, 1.57mmol, 2eq), anhydrous potassium acetate (230mg, 2.3mmol, 3eq) and Pd(dppf)Cl 2 ( 130 mg, 178 μmol, 0.2 eq), and then add 2 mL of anhydrous 1,4-dioxane. After replacing nitrogen, the reaction was placed in an 85°C oil bath and stirred for 10 hours. When the reaction is complete, 10 mL of ethyl acetate was added to the reaction solution, filtered, and concentrated under reduced pressure to obtain black oil II-19-3 (221 mg). LCMS(M+H):259.
步骤3:化合物II-19-4的合成Step 3: Synthesis of Compound II-19-4
向反应管中加入II-19-3(75mg,291μmol,2eq)、II-1-2(60mg,145μmol,1eq)、碳酸钠(46mg,434μmol,3eq)和1mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-19-4(51mg)。LCMS(M+H):447。Add II-19-3 (75mg, 291μmol, 2eq), II-1-2 (60mg, 145μmol, 1eq), sodium carbonate (46mg, 434μmol, 3eq) and 1mL of 1,4-dioxane to the reaction tube. Ring, 0.2mL water, replaced with nitrogen three times and stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10mg, 14μmol, 0.1eq) and tBuXphos (6mg, 14μmol, 0.1eq), and replaced with nitrogen again. three times, then the reaction was stirred in a 100°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-19-4 (51 mg). LCMS(M+H):447.
步骤4:化合物II-19的合成Step 4: Synthesis of Compound II-19
向化合物II-19-4(50mg,56μmol,1eq)中加入二氯甲烷(3mL),再向其中加 入三氟乙酸(0.3mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-19(33mg)。1H NMR(400MHz,Methanol-d4)δ=9.12(s,1H),8.67(s,1H),7.73(s,1H),4.48-4.42(m,1H),3.78(dd,J=16.0,4.0Hz,1H),3.59–3.52(m,1H),3.29-3.23(m,2H),2.43–2.26(m,2H),2.15–1.94(m,2H);LCMS(M+H):347。Dichloromethane (3 mL) was added to compound II-19-4 (50 mg, 56 μmol, 1 eq), and then Trifluoroacetic acid (0.3 mL) was added, and then stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-19 (33 mg). 1 H NMR (400MHz, Methanol-d 4 ) δ = 9.12 (s, 1H), 8.67 (s, 1H), 7.73 (s, 1H), 4.48-4.42 (m, 1H), 3.78 (dd, J = 16.0 ,4.0Hz,1H),3.59–3.52(m,1H),3.29-3.23(m,2H),2.43–2.26(m,2H),2.15–1.94(m,2H); LCMS(M+H): 347.
实施例50、化合物II-20的合成与表征
Example 50, Synthesis and Characterization of Compound II-20
步骤1:化合物II-20-2的合成Step 1: Synthesis of Compound II-20-2
向反应瓶中加入II-20-1(0.5g,1.79mmol,1eq)、碳酸铯(1.8g,5.52mmol,3eq)和10mL无水N,N-二甲基甲酰胺,然后再加入碘甲烷(0.61g,3.59mmol,2eq),反应于室温条件下搅拌10小时。待反应完全,将反应液加入到100mL水中,然后用乙酸乙酯萃取(45mL*3),合并有机相,用60mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到淡黄色油状物II-20-2(0.75g)。LCMS(M+H):322。Add II-20-1 (0.5g, 1.79mmol, 1eq), cesium carbonate (1.8g, 5.52mmol, 3eq) and 10mL anhydrous N,N-dimethylformamide to the reaction flask, then add methyl iodide (0.61g, 3.59mmol, 2eq), the reaction was stirred at room temperature for 10 hours. When the reaction is complete, add the reaction solution to 100mL of water, and then extract with ethyl acetate (45mL*3). Combine the organic phases and wash with 60mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure. A light yellow oily substance II-20-2 (0.75g) was obtained. LCMS(M+H):322.
步骤2:化合物II-20-3的合成Step 2: Synthesis of Compound II-20-3
向反应瓶中加入II-20-2(0.75g,2.3mmol,1eq)、Bpin2(1.2g,4.7mmol,2eq)、无水乙酸钾(0.68g,6.9mmol,3eq)和Pd(dppf)Cl2(300mg,410μmol,0.18eq),再加入6mL的无水1,4-二氧六环,置换氮气后将反应置于85℃油浴中搅拌10小时。待反应完全,向反应液中加入10mL乙酸乙酯,过滤后经减压浓缩得到黑色油状物II-20-3(0.61g)。LCMS(M+H):287。Add II-20-2 (0.75g, 2.3mmol, 1eq), Bpin 2 (1.2g, 4.7mmol, 2eq), anhydrous potassium acetate (0.68g, 6.9mmol, 3eq) and Pd (dppf) to the reaction flask. Cl 2 (300 mg, 410 μmol, 0.18 eq), and then add 6 mL of anhydrous 1,4-dioxane. After replacing nitrogen, the reaction was placed in an 85°C oil bath and stirred for 10 hours. When the reaction is complete, 10 mL of ethyl acetate was added to the reaction solution, filtered and concentrated under reduced pressure to obtain black oil II-20-3 (0.61g). LCMS(M+H):287.
步骤3:化合物II-20-4的合成Step 3: Synthesis of Compound II-20-4
向反应管中加入II-20-3(83mg,290μmol,2eq)、II-1-2(60mg,145μmol,1eq)、碳酸钠(46mg,434μmol,3eq)和1mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10 小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-20-4(60mg)。LCMS(M+H):475。Add II-20-3 (83mg, 290μmol, 2eq), II-1-2 (60mg, 145μmol, 1eq), sodium carbonate (46mg, 434μmol, 3eq) and 1mL of 1,4-dioxane to the reaction tube. Ring, 0.2mL water, replaced with nitrogen three times and stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10mg, 14μmol, 0.1eq) and tBuXphos (6mg, 14μmol, 0.1eq), and replaced with nitrogen again. three times, then the reaction was stirred in a 100°C oil bath for 10 Hour. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-20-4 (60 mg). LCMS(M+H):475.
步骤4:化合物II-20的合成Step 4: Synthesis of Compound II-20
向化合物II-20-4(60mg,127μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入15mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-20(23mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.94(s,1H),8.53(s,1H),7.49(s,1H),4.31-4.24(m,1H),3.80(p,J=4.0Hz,1H),3.58(dd,J=16.0,4.0Hz,1H),3.37-3.33(m,1H),3.11-3.04(m,2H),2.24–2.07(m,2H),1.95–1.73(m,2H),1.36(dd,J=4.0,2.4Hz,6H);LCMS(M+H):375。Dichloromethane (2 mL) was added to compound II-20-4 (60 mg, 127 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 15 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-20 (23 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.94 (s, 1H), 8.53 (s, 1H), 7.49 (s, 1H), 4.31-4.24 (m, 1H), 3.80 (p, J = 4.0Hz, 1H),3.58(dd,J=16.0,4.0Hz,1H),3.37-3.33(m,1H),3.11-3.04(m,2H),2.24–2.07(m,2H),1.95–1.73(m, 2H), 1.36 (dd, J=4.0, 2.4Hz, 6H); LCMS (M+H): 375.
实施例51、化合物II-22的合成与表征
Example 51. Synthesis and characterization of compound II-22
步骤1:化合物II-22-1的合成Step 1: Synthesis of Compound II-22-1
向封管中加入II-1-1(100mg,401μmol,1eq),加入2mL的N-甲基吡咯烷酮,再分别加入(S)-6,6-二甲基哌啶-3-胺二盐酸盐(161mg,0.8mmol,2eq)和N,N-二异丙基乙胺(104mg,0.8mmol,2eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到30mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(二氯甲烷:甲醇=1:0至10:1)得到黄色油状物II-22-1(173mg)。LCMS(M+H):342。Add II-1-1 (100 mg, 401 μmol, 1 eq) to the sealed tube, add 2 mL of N-methylpyrrolidone, and then add (S)-6,6-dimethylpiperidin-3-amine dihydrochloride respectively. salt (161 mg, 0.8 mmol, 2 eq) and N, N-diisopropylethylamine (104 mg, 0.8 mmol, 2 eq), and then placed in a 135°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 30mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (dichloromethane: methanol = 1:0 to 10:1) to obtain yellow oil II-22-1 (173 mg). LCMS(M+H):342.
步骤2:化合物II-22的合成Step 2: Synthesis of Compound II-22
向反应管中加入II-22-1(70mg,205μmol,1eq)、II-3-2(88mg,309μmol,2eq)、碳酸钠(109mg,1mmol,5eq)和3mL的1,4-二氧六环、0.3mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(14mg,19μmol,0.1eq)、tBuXphos(8mg,19μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体 II-22(11mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.70(s,1H),8.01(s,1H),7.25(s,1H),7.12(d,J=8.0Hz,1H),6.63(d,J=8.0Hz,1H),4.05-3.92(m,1H),3.42–3.33(m,1H),3.20–3.10(m,1H),1.83(s,1H),1.78-1.55(m,7H),1.38(d,J=28.0Hz,6H);LCMS(M+H):420。Add II-22-1 (70mg, 205μmol, 1eq), II-3-2 (88mg, 309μmol, 2eq), sodium carbonate (109mg, 1mmol, 5eq) and 3mL of 1,4-dioxane to the reaction tube. Ring, 0.3mL water, replaced with nitrogen three times and stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (14mg, 19μmol, 0.1eq), tBuXphos (8mg, 19μmol, 0.1eq), and replaced with nitrogen again. three times, then the reaction was stirred in a 100°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain a yellow solid II-22(11mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.70 (s, 1H), 8.01 (s, 1H), 7.25 (s, 1H), 7.12 (d, J = 8.0Hz, 1H), 6.63 (d, J = 8.0Hz,1H),4.05-3.92(m,1H),3.42–3.33(m,1H),3.20–3.10(m,1H),1.83(s,1H),1.78-1.55(m,7H),1.38 (d, J=28.0Hz, 6H); LCMS (M+H): 420.
实施例52、化合物II-24的合成与表征
Example 52. Synthesis and characterization of compound II-24
步骤1:化合物II-24-1的合成Step 1: Synthesis of Compound II-24-1
向封管中加入II-1-1(50mg,201μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(2s,4r)-叔丁基2-氨基-6-氮杂螺[3.4]辛烷-6-甲酸酯(136mg,0.6mmol,3eq)和N,N-二异丙基乙胺(130mg,1mmol,5eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-24-1(52mg)。LCMS(M+H):439。Add II-1-1 (50 mg, 201 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (2s, 4r)-tert-butyl 2-amino-6-azaspiro [3.4 ] Octane-6-carboxylate (136 mg, 0.6 mmol, 3 eq) and N,N-diisopropylethylamine (130 mg, 1 mmol, 5 eq), and then placed in an oil bath at 135°C and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain yellow oil II-24-1 (52 mg). LCMS(M+H):439.
步骤2:化合物II-24-2的合成Step 2: Synthesis of Compound II-24-2
向反应管中加入II-24-1(50mg,114μmol,1eq)、II-3-2(50mg,175μmol,1.5eq)、碳酸钠(40mg,377μmol,3eq)和2mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(8mg,11μmol,0.1eq)、tBuXphos(5mg,11μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-24-2(33mg)。LCMS(M+H):518。Add II-24-1 (50 mg, 114 μmol, 1 eq), II-3-2 (50 mg, 175 μmol, 1.5 eq), sodium carbonate (40 mg, 377 μmol, 3 eq) and 2 mL of 1,4-dioxygen to the reaction tube. Six rings, 0.2mL water, replaced with nitrogen three times, stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (8mg, 11μmol, 0.1eq), tBuXphos (5mg, 11μmol, 0.1eq), and replaced again. Nitrogen was added three times, and the reaction was stirred in a 100 °C oil bath for 10 h. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-24-2 (33 mg). LCMS(M+H):518.
步骤3:化合物II-24的合成Step 3: Synthesis of Compound II-24
向化合物II-24-2(33mg,64μmol,1eq)中加入二氯甲烷(2mL),再向其中加 入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-24(13mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.73(s,1H),8.15(s,1H),7.29(d,J=8.0Hz,1H),7.14(s,1H),6.76(d,J=8.0Hz,1H),4.08(p,J=8.0Hz,1H),3.36–3.26(m,4H),2.48(t,J=10.3Hz,2H),2.16–1.99(m,4H),1.65(dt,J=16.0,4.0Hz,4H);LCMS(M+H):418。Dichloromethane (2 mL) was added to compound II-24-2 (33 mg, 64 μmol, 1 eq), and then Trifluoroacetic acid (0.2 mL) was added, and then stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-24 (13 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.73 (s, 1H), 8.15 (s, 1H), 7.29 (d, J = 8.0Hz, 1H), 7.14 (s, 1H), 6.76 (d, J = 8.0Hz,1H),4.08(p,J=8.0Hz,1H),3.36–3.26(m,4H),2.48(t,J=10.3Hz,2H),2.16–1.99(m,4H),1.65( dt, J=16.0, 4.0Hz, 4H); LCMS (M+H): 418.
实施例53、化合物II-25的合成与表征
Example 53. Synthesis and characterization of compound II-25
步骤1:化合物II-25-1的合成Step 1: Synthesis of Compound II-25-1
向封管中加入II-1-1(100mg,401μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入2-氨基-7-BOC-7-氮杂螺[3.5]壬烷(193mg,0.8mmol,2eq)和N,N-二异丙基乙胺(104mg,0.8mmol,2eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-25-1(70mg)。LCMS(M+H):453。Add II-1-1 (100 mg, 401 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add 2-amino-7-BOC-7-azaspiro[3.5]nonane (193 mg). ,0.8mmol, 2eq) and N,N-diisopropylethylamine (104mg, 0.8mmol, 2eq), and then placed in a 135°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain yellow oil II-25-1 (70 mg). LCMS(M+H):453.
步骤2:化合物II-25-2的合成Step 2: Synthesis of Compound II-25-2
向反应管中加入II-25-1(70mg,154μmol,1eq)、II-3-2(70mg,245μmol,1.5eq)、碳酸钠(49mg,462μmol,3eq)和2mL的1,4-二氧六环、0.23mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(11mg,15μmol,0.1eq)、tBuXphos(7mg,15μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-25-2(23mg)。LCMS(M+H):532。 Add II-25-1 (70mg, 154μmol, 1eq), II-3-2 (70mg, 245μmol, 1.5eq), sodium carbonate (49mg, 462μmol, 3eq) and 2mL of 1,4-dioxygen to the reaction tube. Six rings, 0.23 mL water, replaced with nitrogen three times, stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (11 mg, 15 μmol, 0.1 eq), tBuXphos (7 mg, 15 μmol, 0.1 eq), and replaced again. Nitrogen was added three times, and the reaction was stirred in a 100 °C oil bath for 10 h. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-25-2 (23 mg). LCMS(M+H):532.
步骤3:化合物II-25的合成Step 3: Synthesis of Compound II-25
向化合物II-25-2(23mg,43μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-25(11mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.62(s,1H),7.96(s,1H),7.21(d,J=8.0Hz,1H),7.01(s,1H),6.58(d,J=8.0Hz,1H),3.82(s,1H),3.07(d,J=8.0Hz,4H),2.16(s,2H),1.80–1.53(m,10H);LCMS(M+H):432。Dichloromethane (2 mL) was added to compound II-25-2 (23 mg, 43 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-25 (11 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.62 (s, 1H), 7.96 (s, 1H), 7.21 (d, J = 8.0Hz, 1H), 7.01 (s, 1H), 6.58 (d, J = 8.0Hz, 1H), 3.82 (s, 1H), 3.07 (d, J = 8.0Hz, 4H), 2.16 (s, 2H), 1.80–1.53 (m, 10H); LCMS (M+H): 432.
实施例54、化合物II-26的合成与表征
Example 54. Synthesis and characterization of compound II-26
步骤1:化合物II-26-1的合成Step 1: Synthesis of Compound II-26-1
向封管中加入II-1-1(80mg,321μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(R)-叔丁基-3-氨基-1-氧杂-8-氮杂螺[4.5]癸烷-8-甲酸酯(160mg,0.6mmol,2eq)和N,N-二异丙基乙胺(210mg,1.6mmol,5eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-26-1(70mg)。LCMS(M+H):469。Add II-1-1 (80 mg, 321 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (R)-tert-butyl-3-amino-1-oxa-8-nitrogen respectively. Heterospiro[4.5]decane-8-carboxylate (160mg, 0.6mmol, 2eq) and N,N-diisopropylethylamine (210mg, 1.6mmol, 5eq) were then placed in an oil bath at 135°C and stirred 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain yellow oil II-26-1 (70 mg). LCMS(M+H):469.
步骤2:化合物II-26-2的合成Step 2: Synthesis of Compound II-26-2
向反应管中加入II-26-1(70mg,149μmol,1eq)、II-3-2(64mg,225μmol,1.5eq)、碳酸钠(47mg,443μmol,3eq)和2mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(10mg,14μmol,0.1eq)、tBuXphos(6mg,14μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯 =10:1至0:1)得到黄色油状物II-26-2(36mg)。LCMS(M+H):548。Add II-26-1 (70mg, 149μmol, 1eq), II-3-2 (64mg, 225μmol, 1.5eq), sodium carbonate (47mg, 443μmol, 3eq) and 2mL of 1,4-dioxygen to the reaction tube. Six rings, 0.2 mL water, replaced with nitrogen three times, stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (10 mg, 14 μmol, 0.1 eq), tBuXphos (6 mg, 14 μmol, 0.1 eq), and replaced again. Nitrogen was added three times, and the reaction was stirred in a 100 °C oil bath for 10 h. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate =10:1 to 0:1) to obtain yellow oil II-26-2 (36 mg). LCMS(M+H):548.
步骤3:化合物II-26的合成Step 3: Synthesis of Compound II-26
向化合物II-26-2(36mg,66μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-26(19mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.62(s,1H),7.93(s,1H),7.14(dd,J=8.0,4.0Hz,1H),7.09(d,J=1.6Hz,1H),6.56(d,J=8.0Hz,1H),4.03(s,1H),3.98-3.91(m,1H),3.56(dd,J=12.0,4.8Hz,1H),3.32–3.16(m,4H),2.17(dd,J=16.0,8.0Hz,1H),1.98-1.90(m,2H),1.87–1.78(m,3H),1.67–1.53(m,4H);LCMS(M+H):448。Dichloromethane (2 mL) was added to compound II-26-2 (36 mg, 66 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-26 (19 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.62 (s, 1H), 7.93 (s, 1H), 7.14 (dd, J = 8.0, 4.0Hz, 1H), 7.09 (d, J = 1.6Hz, 1H) ,6.56(d,J=8.0Hz,1H),4.03(s,1H),3.98-3.91(m,1H),3.56(dd,J=12.0,4.8Hz,1H),3.32–3.16(m,4H ),2.17(dd,J=16.0,8.0Hz,1H),1.98-1.90(m,2H),1.87–1.78(m,3H),1.67–1.53(m,4H); LCMS(M+H): 448.
实施例55、化合物II-27的合成与表征
Example 55. Synthesis and characterization of compound II-27
步骤1:化合物II-27-1的合成Step 1: Synthesis of Compound II-27-1
向封管中加入II-1-1(100mg,401μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入6-氨基-3-氮杂双环[3.1.0]己烷-3-羧酸叔丁酯(159mg,0.8mmol,2eq)和N,N-二异丙基乙胺(104mg,0.8mmol,2eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-27-1(100mg)。LCMS(M+H):411。Add II-1-1 (100 mg, 401 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add 6-amino-3-azabicyclo[3.1.0]hexane-3-carboxy respectively. Acid tert-butyl ester (159 mg, 0.8 mmol, 2 eq) and N, N-diisopropylethylamine (104 mg, 0.8 mmol, 2 eq) were then placed in a 135°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain yellow oil II-27-1 (100 mg). LCMS(M+H):411.
步骤2:化合物II-27-2的合成Step 2: Synthesis of Compound II-27-2
向反应管中加入II-27-1(100mg,243μmol,1eq)、II-3-2(70mg,243μmol,1eq)、碳酸钠(77mg,0.73mmol,3eq)和2mL的1,4-二氧六环、0.25mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(17mg,24μmol,0.1eq)、tBuXphos(10mg,24μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10 小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-27-2(34mg)。LCMS(M+H):490。Add II-27-1 (100 mg, 243 μmol, 1 eq), II-3-2 (70 mg, 243 μmol, 1 eq), sodium carbonate (77 mg, 0.73 mmol, 3 eq) and 2 mL of 1,4-dioxygen to the reaction tube. Six rings, 0.25mL water, replaced with nitrogen three times, stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (17mg, 24μmol, 0.1eq), tBuXphos (10mg, 24μmol, 0.1eq), and replaced again. nitrogen three times, then the reaction was placed in a 100°C oil bath and stirred for 10 Hour. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-27-2 (34 mg). LCMS(M+H):490.
步骤3:化合物II-27的合成Step 3: Synthesis of Compound II-27
向化合物II-27-2(34mg,70μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-27(13mg)。1H NMR(400MHz,Methanol-d4)δ=9.16(s,1H),8.67(s,1H),7.68-7.61(m,2H),7.08(d,J=8.0Hz,1H),3.59(s,4H),2.90-2.85(m,1H),2.21(s,2H),1.72–1.66(m,4H);LCMS(M+H):390。Dichloromethane (2 mL) was added to compound II-27-2 (34 mg, 70 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-27 (13 mg). 1 H NMR (400MHz, Methanol-d 4 ) δ = 9.16 (s, 1H), 8.67 (s, 1H), 7.68-7.61 (m, 2H), 7.08 (d, J = 8.0Hz, 1H), 3.59 ( s,4H),2.90-2.85(m,1H),2.21(s,2H),1.72–1.66(m,4H); LCMS(M+H):390.
实施例56、化合物II-28的合成与表征
Example 56. Synthesis and characterization of compound II-28
步骤1:化合物II-28-1的合成Step 1: Synthesis of Compound II-28-1
向封管中加入II-1-1(100mg,401μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入3-[(叔丁氧基)羰基]-3-氮杂二环[3.1.0]己烷-2-羧酸酯(119mg,0.6mmol,1.5eq)和N,N-二异丙基乙胺(155mg,1.2mmol,3eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-28-1(70mg)。LCMS(M+H):411。Add II-1-1 (100 mg, 401 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add 3-[(tert-butoxy)carbonyl]-3-azabicyclo[3.1 .0] Hexane-2-carboxylate (119mg, 0.6mmol, 1.5eq) and N,N-diisopropylethylamine (155mg, 1.2mmol, 3eq), then placed in a 135°C oil bath and stirred for 10 Hour. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain yellow oil II-28-1 (70 mg). LCMS(M+H):411.
步骤2:化合物II-28-2的合成Step 2: Synthesis of Compound II-28-2
向反应管中加入II-28-1(70mg,170μmol,1eq)、II-3-2(73mg,256μmol,1.5eq)、碳酸钠(54mg,0.51mmol,3eq)和2mL的1,4-二氧六环、0.2mL水,氮气置换三次 后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(12mg,17μmol,0.1eq)、tBuXphos(7mg,17μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-28-2(41mg)。LCMS(M+H):490。Add II-28-1 (70 mg, 170 μmol, 1 eq), II-3-2 (73 mg, 256 μmol, 1.5 eq), sodium carbonate (54 mg, 0.51 mmol, 3 eq) and 2 mL of 1,4-bis to the reaction tube. Oxygen hexacyclic ring, 0.2mL water, nitrogen replacement three times Stir for 5 minutes, then add Pd(PPh 3 ) 2 Cl 2 (12 mg, 17 μmol, 0.1 eq) and tBuXphos (7 mg, 17 μmol, 0.1 eq) in sequence, replace nitrogen three times again, and then place the reaction at 100°C. Stir in oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-28-2 (41 mg). LCMS(M+H):490.
步骤3:化合物II-28的合成Step 3: Synthesis of Compound II-28
向化合物II-28-2(41mg,84μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-28(9mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.81(s,1H),8.14(s,1H),7.34(d,J=8.0Hz,1H),7.12(s,1H),6.82(d,J=8.0Hz,1H),3.66–3.37(m,4H),2.01-1.95(m,1H),1.70–1.58(m,4H),1.28(t,J=8.0Hz,1H),1.15–1.06(m,1H);LCMS(M+H):390。Dichloromethane (2 mL) was added to compound II-28-2 (41 mg, 84 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-28 (9 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.81 (s, 1H), 8.14 (s, 1H), 7.34 (d, J = 8.0Hz, 1H), 7.12 (s, 1H), 6.82 (d, J = 8.0Hz,1H),3.66–3.37(m,4H),2.01-1.95(m,1H),1.70–1.58(m,4H),1.28(t,J=8.0Hz,1H),1.15–1.06(m , 1H); LCMS (M+H): 390.
实施例57、化合物II-29的合成与表征
Example 57, Synthesis and Characterization of Compound II-29
步骤1:化合物II-29-1的合成Step 1: Synthesis of Compound II-29-1
向封管中加入II-1-1(80mg,321μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(S)-1-叔丁氧羰基-3-氨基吡咯烷(120mg,0.6mmol,2eq)和N,N-二异丙基乙胺(210mg,1.6mmol,5eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-29-1(100mg)。LCMS(M+H):399。 Add II-1-1 (80 mg, 321 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (S)-1-tert-butoxycarbonyl-3-aminopyrrolidine (120 mg, 0.6 mmol, 2eq) and N,N-diisopropylethylamine (210mg, 1.6mmol, 5eq), and then placed in a 135°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain yellow oil II-29-1 (100 mg). LCMS(M+H):399.
步骤2:化合物II-29-2的合成Step 2: Synthesis of Compound II-29-2
向反应管中加入II-29-1(50mg,125μmol,1eq)、II-3-2(50mg,175μmol,1.5eq)、碳酸钠(40mg,377μmol,3eq)和2mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(9mg,13μmol,0.1eq)、tBuXphos(5mg,12μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-29-2(36mg)。LCMS(M+H):478。Add II-29-1 (50mg, 125μmol, 1eq), II-3-2 (50mg, 175μmol, 1.5eq), sodium carbonate (40mg, 377μmol, 3eq) and 2mL of 1,4-dioxygen to the reaction tube. Six rings, 0.2mL water, replaced with nitrogen three times, stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (9mg, 13μmol, 0.1eq), tBuXphos (5mg, 12μmol, 0.1eq), and replaced again. Nitrogen was added three times, and the reaction was stirred in a 100 °C oil bath for 10 h. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-29-2 (36 mg). LCMS(M+H):478.
步骤3:化合物II-29的合成Step 3: Synthesis of Compound II-29
向化合物II-29-2(36mg,75μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-29(15mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.80(s,1H),8.18(s,1H),7.37(d,J=8.0Hz,1H),7.29(s,1H),6.81(d,J=8.0Hz,1H),4.49-4.42(m,1H),3.63–3.34(m,4H),2.46-2.33(m,1H),2.20-2.08(m,1H),1.72–1.54(m,4H);LCMS(M+H):378。Dichloromethane (2 mL) was added to compound II-29-2 (36 mg, 75 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-29 (15 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.80 (s, 1H), 8.18 (s, 1H), 7.37 (d, J = 8.0Hz, 1H), 7.29 (s, 1H), 6.81 (d, J = 8.0Hz,1H),4.49-4.42(m,1H),3.63–3.34(m,4H),2.46-2.33(m,1H),2.20-2.08(m,1H),1.72–1.54(m,4H) ;LCMS(M+H):378.
实施例58、化合物II-30的合成与表征
Example 58. Synthesis and characterization of compound II-30
步骤1:化合物II-30-1的合成Step 1: Synthesis of Compound II-30-1
向封管中加入II-1-1(80mg,321μmol,1eq),加入1mL的N-甲基吡咯烷酮,再分别加入(2R,4S)-4-氨基-2-甲基吡咯烷-1-甲酸叔丁酯(84mg,417μmol,1.3eq)和N,N-二异丙基乙胺(124mg,0.96mmol,3eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15 mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-30-1(90mg)。LCMS(M+H):413。Add II-1-1 (80 mg, 321 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then add (2R, 4S)-4-amino-2-methylpyrrolidine-1-carboxylic acid. Tert-butyl ester (84 mg, 417 μmol, 1.3 eq) and N, N-diisopropylethylamine (124 mg, 0.96 mmol, 3 eq) were then placed in a 135°C oil bath and stirred for 10 hours. When the reaction is complete, the reaction solution is cooled to room temperature, added to 20 mL of water, and then extracted with ethyl acetate (15 mL*3), combined the organic phases, washed with 20 mL saturated brine, and the resulting organic phase was dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to obtain yellow oil II-30-1 (90 mg). LCMS(M+H):413.
步骤2:化合物II-30-2的合成Step 2: Synthesis of Compound II-30-2
向反应管中加入II-30-1(90mg,218μmol,1eq)、II-3-2(90mg,316μmol,1.5eq)、碳酸钠(69mg,0.65mmol,3eq)和3mL的1,4-二氧六环、0.3mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(15mg,21μmol,0.1eq)、tBuXphos(9mg,21μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-30-2(43mg)。LCMS(M+H):492。Add II-30-1 (90mg, 218μmol, 1eq), II-3-2 (90mg, 316μmol, 1.5eq), sodium carbonate (69mg, 0.65mmol, 3eq) and 3mL of 1,4-bis to the reaction tube. Oxygen hexacyclo and 0.3 mL water were replaced with nitrogen three times and stirred for 5 minutes. Then Pd(PPh 3 ) 2 Cl 2 (15 mg, 21 μmol, 0.1 eq) and tBuXphos (9 mg, 21 μmol, 0.1 eq) were added in sequence, and again The nitrogen was replaced three times, and the reaction was stirred in a 100°C oil bath for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-30-2 (43 mg). LCMS(M+H):492.
步骤3:化合物II-30的合成Step 3: Synthesis of Compound II-30
向化合物II-30-2(43mg,88μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-30(17mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.77(s,1H),8.11(s,1H),7.29(d,J=8.0Hz,1H),7.21(s,1H),6.77(d,J=8.0Hz,1H),4.47–4.38(m,1H),3.78-3.65(m,1H),3.60(dd,J=12.0,8.0Hz,1H),3.27(dd,J=16.0,8.0Hz,1H),2.68-2.57(m,1H),1.79-1.70(m,1H),1.69-1.57(m,4H),1.42(d,J=8.0Hz,3H);LCMS(M+H):392。Dichloromethane (2 mL) was added to compound II-30-2 (43 mg, 88 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-30 (17 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.77 (s, 1H), 8.11 (s, 1H), 7.29 (d, J = 8.0Hz, 1H), 7.21 (s, 1H), 6.77 (d, J = 8.0Hz,1H),4.47–4.38(m,1H),3.78-3.65(m,1H),3.60(dd,J=12.0,8.0Hz,1H),3.27(dd,J=16.0,8.0Hz,1H ), 2.68-2.57(m,1H),1.79-1.70(m,1H),1.69-1.57(m,4H),1.42(d,J=8.0Hz,3H); LCMS(M+H):392.
实施例59、化合物II-32的合成与表征
Example 59. Synthesis and characterization of compound II-32
步骤1:化合物II-32-1的合成Step 1: Synthesis of Compound II-32-1
向封管中加入II-1-1(100mg,401μmol,1eq),加入1mL的N-甲基吡咯烷酮,再 分别加入(3R,4S)-3-氨基-4-氟吡咯烷-1-甲酸叔丁酯(123mg,0.6mmol,1.5eq)和N,N-二异丙基乙胺(104mg,0.8mmol,2eq),然后置于135℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(15mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到黄色油状物II-32-1(56mg)。LCMS(M+H):417。Add II-1-1 (100 mg, 401 μmol, 1 eq) to the sealed tube, add 1 mL of N-methylpyrrolidone, and then Add (3R,4S)-3-amino-4-fluoropyrrolidine-1-carboxylic acid tert-butyl ester (123mg, 0.6mmol, 1.5eq) and N,N-diisopropylethylamine (104mg, 0.8mmol, 2eq), then placed in a 135°C oil bath and stirred for 10 hours. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (15mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered, and concentrated under reduced pressure to obtain yellow oil II-32-1 (56 mg). LCMS(M+H):417.
步骤2:化合物II-32-2的合成Step 2: Synthesis of Compound II-32-2
向反应管中加入II-32-1(50mg,120μmol,1eq)、II-3-2(51mg,316μmol,1.5eq)、碳酸钠(40mg,377μmol,3eq)和2mL的1,4-二氧六环、0.2mL水,氮气置换三次后搅拌5分钟,然后再向其中依次加入Pd(PPh3)2Cl2(8mg,12μmol,0.1eq)、tBuXphos(5mg,12μmol,0.1eq),再次置换氮气三次,然后将反应置于100℃油浴中搅拌10小时。待反应完全,反应液冷却至室温,将反应液加入到20mL水中,然后用乙酸乙酯萃取(20mL*3),合并有机相,用20mL饱和食盐水洗涤,所得有机相经无水硫酸钠干燥、过滤、减压浓缩得到粗品。粗品经硅胶柱层析分离纯化(石油醚:乙酸乙酯=10:1至0:1)得到黄色油状物II-32-2(14mg)。LCMS(M+H):496。Add II-32-1 (50mg, 120μmol, 1eq), II-3-2 (51mg, 316μmol, 1.5eq), sodium carbonate (40mg, 377μmol, 3eq) and 2mL of 1,4-dioxygen to the reaction tube. Six rings, 0.2mL water, replaced with nitrogen three times, stirred for 5 minutes, then added Pd(PPh 3 ) 2 Cl 2 (8mg, 12μmol, 0.1eq), tBuXphos (5mg, 12μmol, 0.1eq), and replaced again. Nitrogen was added three times, and the reaction was stirred in a 100 °C oil bath for 10 h. When the reaction is complete, cool the reaction solution to room temperature, add the reaction solution to 20mL of water, and then extract with ethyl acetate (20mL*3). Combine the organic phases and wash with 20mL of saturated brine. The resulting organic phase is dried over anhydrous sodium sulfate. , filtered and concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10:1 to 0:1) to obtain yellow oil II-32-2 (14 mg). LCMS(M+H):496.
步骤3:化合物II-32的合成Step 3: Synthesis of Compound II-32
向化合物II-32-2(14mg,28μmol,1eq)中加入二氯甲烷(2mL),再向其中加入三氟乙酸(0.2mL),然后于室温条件下搅拌3小时。待反应完全,向反应液加入10mL二氯甲烷,用10mL饱和碳酸氢钠溶液洗涤,有机相经减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到黄色固体II-32(5mg)。1H NMR(400MHz,Deuterium Oxide)δ=8.79(s,1H),8.08(s,1H),7.31(d,J=8.0Hz,1H),7.23(s,1H),6.80(d,J=8.0Hz,1H),5.37(d,J=52.0Hz,1H),3.90–3.61(m,4H),3.36(t,J=12.0Hz,1H),1.65(d,J=8.0Hz,4H);19F NMR(376MHz,Deuterium Oxide)δ=-195.98;LCMS(M+H):396。Dichloromethane (2 mL) was added to compound II-32-2 (14 mg, 28 μmol, 1 eq), trifluoroacetic acid (0.2 mL) was added thereto, and the mixture was stirred at room temperature for 3 hours. When the reaction is complete, add 10 mL of methylene chloride to the reaction solution, wash with 10 mL of saturated sodium bicarbonate solution, and concentrate the organic phase under reduced pressure to obtain a crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain yellow solid II-32 (5 mg). 1 H NMR (400MHz, Deuterium Oxide) δ = 8.79 (s, 1H), 8.08 (s, 1H), 7.31 (d, J = 8.0Hz, 1H), 7.23 (s, 1H), 6.80 (d, J = 8.0Hz,1H),5.37(d,J=52.0Hz,1H),3.90–3.61(m,4H),3.36(t,J=12.0Hz,1H),1.65(d,J=8.0Hz,4H) ; 19 F NMR (376MHz, Deuterium Oxide) δ = -195.98; LCMS (M+H): 396.
实施例60、化合物I-44的合成与表征
Example 60, Synthesis and Characterization of Compound I-44
化合物I-44的合成参考实施例1化合物I-7的合成路线,将化合物(1S,3S)-3-氨基环戊基氨基甲酸叔丁酯替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-44为淡黄色固体(10mg),LCMS(M+H):451.2。1H NMR(400MHz,Deuterium Oxide)δ8.38(s,1H),8.35(d,J=8.3Hz,1H),8.11(d,J=5.5Hz,1H),7.39(dd,1H),7.07(d,J=5.6Hz,1H),4.36(s,1H),3.93(ddd,J=14.4,7.3Hz,1H),2.48–2.33(m,3H),2.31–2.19(m,3H),2.14(d,J=13.6Hz,6H),1.90–1.76(m,2H).Compound I-44 was synthesized by referring to the synthetic route of compound I-7 in Example 1, replacing (S)-3-aminopiperidine-1 with compound (1S,3S)-3-aminocyclopentylcarbamate tert-butyl ester. -tert-butyl carboxylate, the obtained compound I-44 was a light yellow solid (10 mg), LCMS (M+H): 451.2. 1 H NMR (400MHz, Deuterium Oxide) δ8.38 (s, 1H), 8.35 (d, J = 8.3Hz, 1H), 8.11 (d, J = 5.5Hz, 1H), 7.39 (dd, 1H), 7.07 (d,J=5.6Hz,1H),4.36(s,1H),3.93(ddd,J=14.4,7.3Hz,1H),2.48–2.33(m,3H),2.31–2.19(m,3H), 2.14(d,J=13.6Hz,6H),1.90–1.76(m,2H).
实施例61、化合物I-45的合成与表征
Example 61. Synthesis and characterization of compound I-45
化合物I-45的合成参考实施例1化合物I-7的合成路线,将化合物5-氨基-3,3-二氟哌啶-1-甲酸叔丁酯替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-45为淡黄色固体(9mg),LCMS(M+H):487.3。Compound I-45 was synthesized by referring to the synthetic route of compound I-7 in Example 1, except that compound 5-amino-3,3-difluoropiperidine-1-carboxylic acid tert-butyl ester was replaced with (S)-3-aminopiperidine. -1-tert-butylcarboxylate, the obtained compound I-45 was a light yellow solid (9 mg), LCMS (M+H): 487.3.
实施例62、化合物I-46的合成与表征
Example 62. Synthesis and characterization of compound I-46
化合物I-46的合成参考实施例1化合物I-7的合成路线,将化合物(3S,5S)-3-氨基-5-氟哌啶-1-甲酸叔丁酯替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-46为淡黄色固体(13mg),LCMS(M+H):469.2。1H NMR(400MHz,Deuterium Oxide)δ8.27(s, 1H),8.04(d,J=5.5Hz,1H),7.89(s,1H),7.04(dd,J=8.4,3.2Hz,1H),6.88(d,J=5.5Hz,1H),5.43(d,J=44.5Hz,1H),4.56–4.41(m,1H),3.89–3.67(m,2H),3.39(dd,J=41.0,14.6Hz,1H),3.29–3.12(m,1H),2.42(s,1H),2.20–1.89(m,7H).Compound I-46 was synthesized by referring to the synthetic route of compound I-7 in Example 1, replacing (S)-3- with compound (3S,5S)-3-amino-5-fluoroperidine-1-carboxylic acid tert-butyl ester. Aminopiperidine-1-carboxylic acid tert-butyl ester, the obtained compound I-46 was a light yellow solid (13 mg), LCMS (M+H): 469.2. 1 H NMR(400MHz,Deuterium Oxide)δ8.27(s, 1H),8.04(d,J=5.5Hz,1H),7.89(s,1H),7.04(dd,J=8.4,3.2Hz,1H),6.88(d,J=5.5Hz,1H),5.43( d,J=44.5Hz,1H),4.56–4.41(m,1H),3.89–3.67(m,2H),3.39(dd,J=41.0,14.6Hz,1H),3.29–3.12(m,1H) ,2.42(s,1H),2.20–1.89(m,7H).
实施例63、化合物I-47的合成与表征
Example 63. Synthesis and characterization of compound I-47
化合物I-47的合成参考实施例1化合物I-7的合成路线,将化合物(3S,5S)-3-氨基-5-甲基哌啶-1-甲酸叔丁酯替换掉(S)-3-氨基哌啶-1-羧酸叔丁酯,所得化合物I-47为淡黄色固体(11mg),LCMS(M+H):465.2。1H NMR(400MHz,Deuterium Oxide)δ8.26(s,1H),8.19–7.92(m,2H),7.33(s,1H),7.01(d,J=5.5Hz,1H),4.15(s,1H),3.66(d,J=13.0Hz,1H),3.43(d,J=13.1Hz,1H),2.94(t,J=11.9Hz,1H),2.70(t,J=12.4Hz,1H),2.38–2.14(m,2H),2.10(d,J=13.4Hz,6H),1.51–1.29(m,1H),1.09(d,J=6.6Hz,3H).The synthesis of compound I-47 was based on the synthetic route of compound I-7 in Example 1, except that compound (3S,5S)-3-amino-5-methylpiperidine-1-carboxylic acid tert-butyl ester was replaced with (S)-3. -Aminopiperidine-1-carboxylic acid tert-butyl ester, the obtained compound I-47 was a light yellow solid (11 mg), LCMS (M+H): 465.2. 1 H NMR (400MHz, Deuterium Oxide) δ8.26 (s, 1H), 8.19–7.92 (m, 2H), 7.33 (s, 1H), 7.01 (d, J = 5.5Hz, 1H), 4.15 (s, 1H),3.66(d,J=13.0Hz,1H),3.43(d,J=13.1Hz,1H),2.94(t,J=11.9Hz,1H),2.70(t,J=12.4Hz,1H) ,2.38–2.14(m,2H),2.10(d,J=13.4Hz,6H),1.51–1.29(m,1H),1.09(d,J=6.6Hz,3H).
对比例1、化合物I-8的合成与表征
Comparative Example 1. Synthesis and characterization of compound I-8
步骤1:化合物I-8的合成Step 1: Synthesis of Compound I-8
向化合物I-1-3(20mg,39μmol,1eq)的二氯甲烷溶液中(2mL)加入三氟乙酸(0.2mL),然后于室温条件下搅拌10小时。待反应完全,将反应液直接减压浓缩得到粗品。粗品经pre-HPLC(HCl)分离纯化得到淡黄色固体I-8(10mg)。LCMS(M+H):428。Trifluoroacetic acid (0.2 mL) was added to a solution (2 mL) of compound I-1-3 (20 mg, 39 μmol, 1 eq) in dichloromethane, and the mixture was stirred at room temperature for 10 hours. When the reaction is complete, the reaction solution is directly concentrated under reduced pressure to obtain crude product. The crude product was separated and purified by pre-HPLC (HCl) to obtain light yellow solid I-8 (10 mg). LCMS(M+H):428.
测试例1、化合物抑制CDK7/cyclinH/MANT1激酶蛋白活性的生化测试Test Example 1. Biochemical test of compounds inhibiting CDK7/cyclinH/MANT1 kinase protein activity
CDK7/cyclinH/MNAT1激酶蛋白复合物和其底物肽CDK7/9tide在50mM HEPES  pH 7.5,0.01%BRIJ-35,10mM MgCl2和1mM EGTA构成的缓冲体系里制备成2X溶液。CDK7/cyclinH/MNAT1 kinase protein complex and its substrate peptide CDK7/9tide in 50mM HEPES Prepare a 2X solution in a buffer system consisting of pH 7.5, 0.01% BRIJ-35, 10mM MgCl 2 and 1mM EGTA.
Detection混合物由EDTA(30mM),Eu-anti-ADP抗体(6nM)和ADP tracer在TR-FRET稀释缓冲液中制备。Detection mixture is prepared from EDTA (30mM), Eu-anti-ADP antibody (6nM) and ADP tracer in TR-FRET dilution buffer.
在384孔板(Corning#4512)中依次按照以下步骤加入溶液或进行相应操作:Follow the steps below to add the solution or perform corresponding operations in a 384-well plate (Corning #4512):
1. 2.4uL-30mM HEPES1. 2.4uL-30mM HEPES
2. 2.5uL-4X ATP溶液2. 2.5uL-4X ATP solution
3. 100nL-100X被测化合物的DMSO溶液3. 100nL-100X DMSO solution of test compound
4. 5uL-2X CDK7/cyclinH/MNAT1激酶蛋白复合物和其底物肽CDK7/9tide溶液4. 5uL-2X CDK7/cyclinH/MNAT1 kinase protein complex and its substrate peptide CDK7/9tide solution
5.摇动孔板30秒5. Shake the plate for 30 seconds
6. 1000*g下离心1分钟6. Centrifuge at 1000*g for 1 minute
7.室温下反应60分钟7. React at room temperature for 60 minutes
8. 5uL–Detection混合物8. 5uL–Detection mixture
9.摇动孔板30秒9. Shake the plate for 30 seconds
10. 1000*g下离心1分钟10. Centrifuge at 1000*g for 1 minute
11.室温下孵育平衡60分钟11. Incubate at room temperature for 60 minutes to equilibrate.
12.读取荧光数值12. Read the fluorescence value
在10uL的酶反应体系中含有20ng CDK7/cyclinH/MNAT1激酶蛋白复合物,200uM底物肽CDK7/9tide和150uM ATP。被测化合物从最高浓度10uM开始向下依次3倍稀释,共11个浓度点。设置2个复孔。根据荧光读值计算每个孔的抑制率,绘制剂量依赖曲线,计算被测化合物的半数抑制浓度IC50,结果如下表1所示:The 10uL enzyme reaction system contains 20ng CDK7/cyclinH/MNAT1 kinase protein complex, 200uM substrate peptide CDK7/9tide and 150uM ATP. The compound to be tested was diluted 3 times downward starting from the highest concentration of 10uM, for a total of 11 concentration points. Set up 2 multiple holes. Calculate the inhibition rate of each well based on the fluorescence reading, draw a dose-dependent curve, and calculate the half inhibitory concentration IC 50 of the tested compound. The results are shown in Table 1 below:
表1

Table 1

被测化合物在1000nM或100nM和250nM或25nM浓度下的抑制率,结果如下表2所示:The inhibition rates of the tested compounds at concentrations of 1000nM or 100nM and 250nM or 25nM are shown in Table 2 below:
表2
Table 2
实验结果表明部分化合物表现出较好的CDK7酶活抑制能力。Experimental results show that some compounds show better ability to inhibit CDK7 enzyme activity.
测试例2、化合物抑制CDK2/cyclinE激酶蛋白活性的生化测试Test Example 2. Biochemical test of compounds inhibiting CDK2/cyclinE kinase protein activity
GST标记的CDK2/cyclinE激酶蛋白复合物和Eu-anti-GST在50mM HEPES pH 7.5,0.01%BRIJ-35,10mM MgCl2和1mM EGTA构成的缓冲体系里制备成2X溶液。GST-tagged CDK2/cyclinE kinase protein complex and Eu-anti-GST were prepared into a 2X solution in a buffer system consisting of 50mM HEPES pH 7.5, 0.01% BRIJ-35, 10mM MgCl 2 and 1mM EGTA.
AlexaFluor标记的Tracer在Kinase缓冲液中制备成2X溶液。 AlexaFluor-labeled Tracer is prepared as a 2X solution in Kinase buffer.
在384孔板(Greiner#784207)中依次按照以下步骤加入溶液或进行相应操作:Add the solution or perform corresponding operations according to the following steps in the 384-well plate (Greiner#784207):
1. 3.84uL Kinase缓冲液1. 3.84uL Kinase buffer
2. 160nL-100X被测化合物的DMSO溶液2. 160nL-100X DMSO solution of the test compound
3. 8.0uL-2X CDK2/cyclinE激酶蛋白复合物和Eu-anti-GST溶液3. 8.0uL-2X CDK2/cyclinE kinase protein complex and Eu-anti-GST solution
4. 4.0uL-4X AlexaFluor标记的Tracer溶液4. 4.0uL-4X AlexaFluor labeled Tracer solution
5.摇动孔板30秒5. Shake the plate for 30 seconds
6.室温下孵育平衡60分钟6. Incubate at room temperature for 60 minutes to equilibrate.
7.读取荧光数值7. Read the fluorescence value
在最终的孵育体系中含有2.5nM CDK2/cyclinE激酶蛋白复合物,2nM Eu-anti-GST和100nM AlexaFluor标记的Tracer。被测化合物从最高浓度10uM开始向下依次3倍稀释,共11个浓度点。设置2个复孔。根据荧光读值计算每个孔的抑制率,绘制剂量依赖曲线,计算被测化合物的半数抑制浓度IC50,结果如下表3:The final incubation system contained 2.5nM CDK2/cyclinE kinase protein complex, 2nM Eu-anti-GST and 100nM AlexaFluor-labeled Tracer. The compound to be tested was diluted 3 times downward starting from the highest concentration of 10uM, for a total of 11 concentration points. Set up 2 multiple holes. Calculate the inhibition rate of each well based on the fluorescence reading, draw a dose-dependent curve, and calculate the half inhibitory concentration IC 50 of the tested compound. The results are as follows in Table 3:
表3
table 3
被测化合物在1000nM或100nM浓度下的抑制率,结果如下表4所示:The inhibition rate of the tested compound at a concentration of 1000nM or 100nM is shown in Table 4 below:
表4
Table 4
实验结果表明部分化合物表现出较好的CDK2酶活抑制能力。Experimental results show that some compounds show better ability to inhibit CDK2 enzyme activity.
测试例3、化合物对三阴乳腺癌细胞系MDA-MB-453的抗增殖活性测试Test Example 3. Test of anti-proliferative activity of compounds on triple-negative breast cancer cell line MDA-MB-453
MDA-MB-453细胞以4000个/孔的密度接种于96孔培养板内:细胞外面一圈以200uL PBS填充,以防边缘培养基蒸发较快导致内部板孔的培养条件差异过大。内部60个孔的最左列为空白孔,不加细胞,以等体积的PBS填充,其余的54个孔以排枪进行细胞铺板,每孔100uL,放入二氧化碳培养箱内37℃培养24小时。使用完全培养基稀释化合物储备液,每孔加入50uL相应浓度的药物溶液,终浓度最高为10uM, 向下依次3倍稀释,共10个浓度点。设置2个复孔。药物处理3天后,每孔加入50uL Cell Tier Glo试剂(Promega#G7570),用酶标仪读取Lumi发光数值,计算每孔抑制率,绘制剂量依赖曲线,部分化合物的剂量依赖曲线如图1a-s所示,计算被测化合物抗增殖的半数有效浓度EC50,结果如下表5:MDA-MB-453 cells were seeded in a 96-well culture plate at a density of 4000 cells/well: the outer circle of cells was filled with 200uL PBS to prevent the rapid evaporation of the edge medium from causing excessive differences in the culture conditions of the inner plate wells. The leftmost column of the 60 internal wells is a blank well, which is filled with an equal volume of PBS without adding cells. The remaining 54 wells are plated with cells using a row gun. Each well is 100 uL and placed in a carbon dioxide incubator for 24 hours at 37°C. Use complete culture medium to dilute the compound stock solution, and add 50uL of drug solution of corresponding concentration to each well, with a final concentration of up to 10uM. Dilute the solution 3 times downwards, for a total of 10 concentration points. Set up 2 multiple holes. After 3 days of drug treatment, add 50uL Cell Tier Glo reagent (Promega#G7570) to each well, read the Lumi luminescence value with a microplate reader, calculate the inhibition rate of each well, and draw a dose dependence curve. The dose dependence curve of some compounds is shown in Figure 1a- As shown in s, calculate the half effective anti-proliferation concentration EC 50 of the tested compound. The results are as follows in Table 5:
表5
table 5
实验结果表明部分化合物表现出较强的抑制三阴乳腺癌细胞系MDA-MB-453增殖的能力。Experimental results showed that some compounds showed strong ability to inhibit the proliferation of triple-negative breast cancer cell line MDA-MB-453.
测试例4、化合物对卵巢癌细胞系OVCAR3的抗增殖活性测试Test Example 4. Test of anti-proliferative activity of compounds on ovarian cancer cell line OVCAR3
OVCAR3细胞以2000个/孔的密度接种于96孔培养板内:细胞外面一圈以200uL PBS填充,以防边缘培养基蒸发较快导致内部板孔的培养条件差异过大。内部60个孔的最左列为空白孔,不加细胞,以等体积的PBS填充,其余的54个孔以排枪进行细胞铺板,每孔100uL,放入二氧化碳培养箱内37℃培养24小时。使用完全培养基稀释化合物储备液,每孔加入50uL相应浓度的药物溶液,终浓度最高为10uM,向下依次3倍稀 释,共10个浓度点。设置2个复孔。药物处理5天后,每孔加入50uL Cell Tier Glo试剂(Promega#G7570),用酶标仪读取Lumi发光数值,计算每孔抑制率,绘制剂量依赖曲附图线,部分化合物的剂量依赖曲线如图2a-b所示,计算被测化合物抗增殖的半数有效浓度EC50,结果如下表6:OVCAR3 cells were seeded in a 96-well culture plate at a density of 2000 cells/well: the outer circle of cells was filled with 200uL PBS to prevent the rapid evaporation of the edge medium from causing excessive differences in the culture conditions of the inner plate wells. The leftmost column of the 60 internal wells is a blank well, which is filled with an equal volume of PBS without adding cells. The remaining 54 wells are plated with cells using a row gun. Each well is 100 uL and placed in a carbon dioxide incubator for 24 hours at 37°C. Use complete culture medium to dilute the compound stock solution, add 50uL of drug solution of corresponding concentration to each well, the final concentration is up to 10uM, and dilute 3 times downwards. Release, a total of 10 concentration points. Set up 2 multiple holes. After 5 days of drug treatment, add 50uL Cell Tier Glo reagent (Promega#G7570) to each well, read the Lumi luminescence value with a microplate reader, calculate the inhibition rate of each well, and draw a dose-dependent curve. The dose-dependent curves of some compounds are as follows: As shown in Figure 2a-b, the half effective anti-proliferation concentration EC 50 of the tested compound was calculated. The results are as follows in Table 6:
表6
Table 6
实验结果表明部分化合物表现出较强的抑制卵巢癌细胞系OVCAR3增殖的能力。Experimental results show that some compounds exhibit strong ability to inhibit the proliferation of ovarian cancer cell line OVCAR3.
测试例5、化合物的肝微粒体稳定性测试Test Example 5. Liver Microsome Stability Test of Compounds
实验所用的肝微粒体信息如下:
The information on liver microsomes used in the experiment is as follows:
实验温孵体系的构成:

The composition of the experimental incubation system:

实验步骤:Experimental steps:
(1)从-80℃冰箱中取出肝微粒体,置于37℃水浴恒温振荡器上预温孵3min,融化待用。(1) Take out the liver microsomes from the -80°C refrigerator, place them in a 37°C water bath constant-temperature oscillator to pre-incubate for 3 minutes, thaw and set aside for use.
(2)按照上面“实验温孵体系的构成”比例,制备温孵体系混合溶液(不含β-NADPH)。(2) Prepare the incubation system mixed solution (without β-NADPH) according to the ratio of the "Composition of Experimental Incubation System" above.
(3)取化合物储备液,用50%乙腈-水稀释成100μM的测试化合物工作溶液,待用。(3) Take the compound stock solution and dilute it with 50% acetonitrile-water to make a 100 μM working solution of the test compound, and set aside.
(4)对照组(不含β-NADPH):取NCF对照组30μL作为0min样品,加入180μL含内标沉淀剂,再加入30μL NADPH,得到0min样品;剩余NCF组加氯化镁30μL,孵育60min后加180μL含内标沉淀剂。(4) Control group (without β-NADPH): Take 30 μL of the NCF control group as the 0min sample, add 180 μL of precipitant containing internal standard, and then add 30 μL of NADPH to obtain the 0min sample; add 30 μL of magnesium chloride to the remaining NCF group, incubate for 60 minutes, and then add 180μL contains internal standard precipitating agent.
(5)样品组:取样品组30μL作为0min样品,加入180μL含内标沉淀剂,再加入30μLNADPH,得到0min样品;剩余样品组每管加NADPH 30μL,孵育5,15,30,60min后加入180μL含内标沉淀剂。(5) Sample group: Take 30 μL of the sample group as the 0min sample, add 180 μL of precipitant containing internal standard, and then add 30 μL of NADPH to obtain the 0min sample; add 30 μL of NADPH to each tube of the remaining sample group, incubate for 5, 15, 30, and 60 min, then add 180 μL Contains internal standard precipitant.
(6)阳性对照组:取PB溶液,加入计算好的对应种属肝微粒体,再加入阳性对照工作溶液,涡旋混匀,分管。加入NADPH溶液进行孵育,于5,15min加入180μL沉淀剂。另分装两管,先加入内标沉淀剂,再加入NADPH,得到0min样品。(6) Positive control group: Take the PB solution, add the calculated liver microsomes of the corresponding species, then add the positive control working solution, vortex to mix, and divide into tubes. Add NADPH solution for incubation, and add 180 μL of precipitating agent at 5 and 15 min. Divide into two other tubes, first add the internal standard precipitant, and then add NADPH to obtain the 0min sample.
(7)所有样品涡旋并离心。(7) Vortex and centrifuge all samples.
(8)取上清液150μL加入150μL水,涡旋混匀,LC-MS/MS进样分析。(8) Take 150 μL of the supernatant, add 150 μL of water, vortex to mix, and inject the sample for LC-MS/MS analysis.
用一级动力学公式计算化合物的半衰期(t1/2)和清除率(CL),结果如下表7:Use the first-order kinetic formula to calculate the half-life (t 1/2 ) and clearance (CL) of the compound. The results are as follows in Table 7:
表7

Table 7

实验结果表明部分化合物在小鼠和人源肝微粒体上表现出较好的稳定性。Experimental results show that some compounds show good stability on mouse and human liver microsomes.
测试例6、化合物抑制CDK家族激酶蛋白活性的选择性测试Test Example 6. Selectivity test of compounds inhibiting the activity of CDK family kinase proteins
表8

Table 8

实验结果表明I-7对CDK7的抑制具有很好的选择性;II-3对CDK7也具有很好的抑制活性,但同时也抑制多个其他CDK家族激酶成员,包括CDK12,CDK13,CDK9,CDK18。Experimental results show that I-7 has good selectivity for inhibiting CDK7; II-3 also has good inhibitory activity against CDK7, but it also inhibits multiple other CDK family kinase members, including CDK12, CDK13, CDK9, and CDK18. .
测试例7化合物在小鼠上的血浆药代动力学测试Test Example 7 Plasma Pharmacokinetic Test of Compounds in Mice
本测试例中,使用不同化合物进行小鼠药物代谢动力学实验,具体步骤如下:In this test example, different compounds were used to conduct pharmacokinetic experiments in mice. The specific steps are as follows:
动物信息:CD-1小鼠,雄性,20g左右,购自北京维通利华实验动物技术有限公司。Animal information: CD-1 mice, male, about 20 g, were purchased from Beijing Vitong Lihua Experimental Animal Technology Co., Ltd.
动物饮食状态:给药前动物不禁食,可自由饮水。Animal dietary status: The animals are not fasting before administration and can drink water freely.
化合物配置方式:灌胃(G1组)用0.5%CMC-Na配制,静注(G2组)用5%DMSO+30%PEG400+65%注射用水配制。Compound preparation method: 0.5% CMC-Na was used for intragastric administration (G1 group), and 5% DMSO+30% PEG400+65% water for injection was used for intravenous injection (G2 group).
灌胃给药(G1组):化合物以10mg/kg(10mL/kg)剂量灌胃给予实验动物(n=9);静注给药(G2组):化合物以1mg/kg(10mL/kg)剂量静注给予实验动物(n=9)。Oral administration (G1 group): the compound was administered to experimental animals (n=9) at a dose of 10 mg/kg (10 mL/kg); intravenous administration (G2 group): the compound was administered at a dose of 1 mg/kg (10 mL/kg) The dose was administered intravenously to experimental animals (n=9).
G1组采样时间点:给药后10min,30min,1h,2h,4h,6h,8h,24h共8个点。G1 group sampling time points: 10min, 30min, 1h, 2h, 4h, 6h, 8h, 24h after administration, a total of 8 points.
G2组采样时间点:给药后5min,10min,30min,1h,2h,4h,6h,8h,24h共9个点。G2 group sampling time points: 5min, 10min, 30min, 1h, 2h, 4h, 6h, 8h, 24h after administration, a total of 9 points.
样品收集:实验当天,分别于各设定时间点采集约0.1mL全血样品置于EDTA-K2抗凝管中。Sample collection: On the day of the experiment, collect approximately 0.1 mL of whole blood samples at each set time point and place them in EDTA-K2 anticoagulant tubes.
样品处理:全血样品在采集后30min内,于4℃,12000rpm条件下离心5min后分离血浆,收集上层血浆样品至样品管中,血浆样品于30min内冻存至-10~-30℃冰箱,并于24h内转移至-60~-90℃冰箱。Sample processing: Within 30 minutes after collection, whole blood samples were centrifuged for 5 minutes at 4°C and 12,000 rpm, and the plasma was separated. The upper plasma samples were collected into sample tubes. The plasma samples were frozen and stored in a -10~-30°C refrigerator within 30 minutes. And transfer to -60~-90℃ refrigerator within 24 hours.
样品检测和数据处理:LC/MS/MS检测,WinNonlin进行拟合和药代参数计算。结果见表9。 Sample detection and data processing: LC/MS/MS detection, WinNonlin for fitting and calculation of pharmacokinetic parameters. The results are shown in Table 9.
表9

Note:IV*为静脉注射途径,PO*为灌胃途径。
Table 9

Note: IV* refers to the intravenous route, and PO* refers to the intragastric route.
实验结果表明本发明化合物具有较好的药代动力学性质。Experimental results show that the compound of the present invention has good pharmacokinetic properties.
测试例8化合物在三阴乳腺癌细胞系MDA-MB-231的裸鼠移植瘤模型上的抗肿瘤药效测试Test Example 8 Anti-tumor efficacy test of the compound on the nude mouse transplanted tumor model of the triple-negative breast cancer cell line MDA-MB-231
本测试例中,在裸鼠上构建人三阴乳腺癌细胞系MDA-MB-231的移植瘤模型,评价化合物I-46的抗肿瘤药效,具体步骤如下:In this test example, a transplanted tumor model of the human triple-negative breast cancer cell line MDA-MB-231 was constructed on nude mice to evaluate the anti-tumor efficacy of compound I-46. The specific steps are as follows:
细胞培养:MDA-MB-231细胞培养在含10%胎牛血清的DMEM培养液、37℃,5%CO2细胞培养箱中,每隔2至3天待细胞长满后分瓶传代,将处于对数生长期的肿瘤细胞用于体内肿瘤的接种。Cell culture: MDA-MB-231 cells were cultured in DMEM culture medium containing 10% fetal bovine serum at 37°C and 5% CO2 in a cell culture incubator. After the cells were full grown, they were divided into bottles and passaged every 2 to 3 days. Tumor cells in the logarithmic growth phase are used for in vivo tumor seeding.
肿瘤细胞接种:采用无菌PBS+Matrigel(V:V=1:1)重悬MDA-MB-231细胞,浓度为5×107/mL,接种于试验动物的右侧胁肋部皮下,100μL/只,在肿瘤生长至平均体积为150mm3左右时分组给药,共4组,分别为溶媒对照组,每日一次灌胃5mg/kg组,每日一次灌胃10mg/kg组,每日一次灌胃20mg/kg组。Tumor cell inoculation: Use sterile PBS+Matrigel (V:V=1:1) to resuspend MDA-MB-231 cells at a concentration of 5×107/mL, and inoculate them subcutaneously into the right flank of the test animal, 100 μL/ Only, when the tumor grows to an average volume of about 150mm3, it is divided into four groups, namely the vehicle control group, the 5mg/kg group once a day, the 10mg/kg group once a day, and the 10mg/kg group once a day. stomach 20mg/kg group.
检测指标:肿瘤体积和体重,每周使用游标卡尺对肿瘤体积进行2~3次的测量,测量肿瘤的长径和短径,其体积计算公式为:体积=0.5×长径×短径2。结果见图3,说明化合物I-46具有较好的抗肿瘤药效。Detection indicators: tumor volume and weight. Use vernier calipers to measure the tumor volume 2 to 3 times a week. Measure the long and short diameters of the tumors. The volume calculation formula is: volume = 0.5 × long diameter × short diameter 2. The results are shown in Figure 3, indicating that compound I-46 has good anti-tumor efficacy.
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。 All documents mentioned in this application are incorporated by reference in this application to the same extent as if each individual document was individually incorporated by reference. In addition, it should be understood that after reading the above teaching content of the present invention, those skilled in the art can make various changes or modifications to the present invention, and these equivalent forms also fall within the scope defined by the appended claims of this application.

Claims (15)

  1. 一种式I或式I’所示的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,
    A compound represented by Formula I or Formula I' or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof,
    其中,X为S或P;Among them, X is S or P;
    X’和X”各自独立地为CH或N;X’ and X” are each independently CH or N;
    Y和Y’各自独立地为C或N;Y and Y’ are each independently C or N;
    Z和Z’各自独立地为CRz或N;其中,Rz选自:H、卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、4-6元杂环基;Z and Z' are each independently CR z or N; wherein, R z is selected from: H, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2. Nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 4-6 One-membered heterocyclyl;
    Rp1和Rp2各自独立地为C1-C6烷基或=O;Rp 1 and Rp 2 are each independently C1-C6 alkyl or =O;
    R1选自取代或未取代的下组基团:C3-C10环烷基、-C1-C6亚烷基C3-C10环烷基、4-10元含N杂环基、-C1-C6亚烷基4-10元含N杂环基;R 1 is selected from the following substituted or unsubstituted groups: C3-C10 cycloalkyl, -C1-C6 alkylene, C3-C10 cycloalkyl, 4-10 membered N-containing heterocyclyl, -C1-C6 alkylene Alkyl 4-10 membered N-containing heterocyclic group;
    R2选自:H、卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、3-6元杂环基;R 2 is selected from: H, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-6 membered heterocyclyl;
    R4和R’4各自独立地选自:无、氢、卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C4烷基、C1-C4烷氧基、C3-C6环烷基、-COOC1-C6烷基、-OCOC1-C6烷基、-CONHC1-C6烷基、-CON(C1-C6烷基)2、-NHC(O)C1-C6烷基、-NHC(O)OC1-C6烷基、-NHS(O)2C1-C6烷基、-S(O)2C1-C6烷基、-S(O)N(C1-C6烷基)2、-S(O)2N(C1-C6烷基)2;其中,所述烷基、烷氧基、环烷基任选地被一个或多个Ra取代;R 4 and R' 4 are each independently selected from: None, hydrogen, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, Cyano, C1-C4 alkyl, C1-C4 alkoxy, C3-C6 cycloalkyl, -COOC1-C6 alkyl, -OCOC1-C6 alkyl, -CONHC1-C6 alkyl, -CON(C1-C6 Alkyl) 2 , -NHC(O)C1-C6 alkyl, -NHC(O)OC1-C6 alkyl, -NHS(O) 2 C1-C6 alkyl, -S(O) 2 C1-C6 alkyl , -S(O)N(C1-C6 alkyl) 2 , -S(O) 2 N(C1-C6 alkyl) 2 ; wherein, the alkyl, alkoxy, and cycloalkyl groups are optionally replaced by One or more Ra substitutions;
    Rp3和R5各自独立地选自:H、卤素、OH、NH2、氰基、C1-C4烷基、C1-C4烷氧基、卤代C1-C4烷基、卤代C1-C4烷氧基;Rp 3 and R 5 are each independently selected from: H, halogen, OH, NH 2 , cyano, C1-C4 alkyl, C1-C4 alkoxy, halogenated C1-C4 alkyl, halogenated C1-C4 alkyl Oxygen;
    Rp4选自:H、卤素、C1-C6烷基、卤代C1-C6烷基、C1-C6烷氧基、卤代C1-C6烷氧基、 Rp 4 is selected from: H, halogen, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy, halogenated C1-C6 alkoxy,
    其中,Rp-1和R’p-1各各自独立地选自取代或未取代的下组基团:C1-C6烷基、C2-C6 烯基、C2-C6炔基、C3-C6环烷基或3-6元杂环基;其中,所述取代是指被一个或多个Ra取代;Wherein, R p-1 and R' p-1 are each independently selected from the following substituted or unsubstituted groups: C1-C6 alkyl, C2-C6 Alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl or 3-6 membered heterocyclyl; wherein the substitution means substitution by one or more Ra;
    L1为取代或未取代的C1-C6烷基,其中,所述取代是指被一个或多个Ra取代;L 1 is a substituted or unsubstituted C1-C6 alkyl group, wherein the substitution means substitution by one or more Ra;
    Rn1和Rn2各自独立地为H、C1-C6烷基、卤代C1-C6烷基、C3-C6环烷基、4-6元杂环基;其中,所述取代是指被一个或多个Ra取代;Rn 1 and Rn 2 are each independently H, C1-C6 alkyl, halogenated C1-C6 alkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl; wherein, the substitution means by one or multiple Ra substitutions;
    Ra各自独立地选自:卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、3-6元杂环基。Ra is each independently selected from: halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , Nitro, cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl , 3-6 membered heterocyclic group.
  2. 一种式II化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,
    a compound of formula II or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof,
    其中,R’1选自取代或未取代的下组基团:C1-C6烷基NRn1Rn2、C3-C10环烷基、-C1-C6亚烷基C3-C10环烷基、4-10元含N杂环基、-C1-C6亚烷基4-10元含N杂环基;Rn1和Rn2各自独立地为H、C1-C6烷基、卤代C1-C6烷基、C3-C6环烷基、4-6元杂环基;其中,所述取代是指被一个或多个Ra取代;Among them, R' 1 is selected from the following substituted or unsubstituted groups: C1-C6 alkyl NRn 1 Rn 2 , C3-C10 cycloalkyl, -C1-C6 alkylene, C3-C10 cycloalkyl, 4- 10-membered N-containing heterocyclic group, -C1-C6 alkylene group, 4-10-membered N-containing heterocyclic group; Rn 1 and Rn 2 are each independently H, C1-C6 alkyl, halogenated C1-C6 alkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl; wherein, the substitution means substitution by one or more Ra;
    R’2选自:H、卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、3-6元杂环基;R' 2 is selected from: H, halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, cyano, C1-C6 alkyl , C1-C6 alkoxy, C1-C6 haloalkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-6 membered heterocyclyl;
    W选自:苯基、萘基、5-6元单环杂芳基或8-10元稠合双环杂芳基、苯并5-10元杂环基、5-6元单环杂芳基并5-10元杂环基;W is selected from: phenyl, naphthyl, 5-6 membered monocyclic heteroaryl or 8-10 membered fused bicyclic heteroaryl, benzo 5-10 membered heterocyclyl, 5-6 membered monocyclic heteroaryl And 5-10 membered heterocyclyl;
    R’5和R’6各自独立地选自:氢、卤素、羟基、硝基、氰基、C1-C6烷基、C3-C6环烷基、C1-C6烷氧基、-N(R5-1aR5-1b)、苯基、C1-C6卤代烷基、C1-C6卤代烷氧基、-NHC(O)R5-2、-NHC(O)OR5-3、-NHS(O)2R5-4、-C(O)N(R5-5aR5-5b)、-S(O)2R5-6、-S(O)(NH)R5-7、-S(O)2N(R5-8aR5-8b)或-P(O)R5-9aR5-9b;或者当R’5和R’6连接于相邻或相同环原子时,R5’和R6’与它们相连的环原子一起形成取代或未取代的C3-C6环烷基或4-6元杂环基;其中,所述取代是指被一个或多个Ra取代;R' 5 and R' 6 are each independently selected from: hydrogen, halogen, hydroxyl, nitro, cyano, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkoxy, -N(R 5-1a R 5-1b ), phenyl, C1-C6 haloalkyl, C1-C6 haloalkoxy, - NHC(O)R 5-2 , -NHC(O)OR 5-3 , -NHS(O) 2 R 5-4 , -C(O)N(R 5-5a R 5-5b ), -S( O) 2 R 5-6 , -S(O)(NH)R 5-7 , -S(O) 2 N(R 5-8a R 5-8b ) or -P(O)R 5-9a R 5 -9b ; or when R' 5 and R' 6 are connected to adjacent or the same ring atom, R 5' and R 6' together with the ring atoms to which they are connected form a substituted or unsubstituted C3-C6 cycloalkyl or 4 -6-membered heterocyclyl; wherein the substitution means substitution by one or more Ra;
    R5-1a、R5-1b、R5-2、R5-3、R5-4、R5-5a、R5-5b、R5-6、R5-7、R5-8a、R5-8b、R5-9a和R5-9b各自独立地选自:氢、C1-C6烷基、C3-C6环烷基;其中,所述C1-C6烷基、C3-C6环烷基任选地被一个或多个选自下组的基团取代:卤素、羟基;或者R5-1a和R5-1b、R5-5a和R5-5b、R5-8a和R5-8b、R5-9a和R5-9b可与它们连接的原子共同形成取代或未取代C3-C6碳环或取代或未取 代4-6元杂环基,其中,所述取代是指被一个或多个Ra取代;R 5-1a , R 5-1b , R 5-2 , R 5-3 , R 5-4 , R 5-5a , R 5-5b , R 5-6 , R 5-7 , R 5-8a , R 5-8b , R 5-9a and R 5-9b are each independently selected from: hydrogen, C1-C6 alkyl, C3-C6 cycloalkyl; wherein, the C1-C6 alkyl, C3-C6 cycloalkyl The group is optionally substituted by one or more groups selected from the group consisting of: halogen, hydroxyl; or R 5-1a and R 5-1b , R 5-5a and R 5-5b , R 5-8a and R 5 -8b , R 5-9a and R 5-9b can form a substituted or unsubstituted C3-C6 carbocyclic ring or a substituted or unsubstituted carbon ring with the atoms to which they are connected. Generation 4-6 membered heterocyclyl, wherein said substitution means substitution by one or more Ra;
    Ra各自独立地选自:卤素、OH、NH2、NHCH3、NHCH2CH3、N(CH3)2、N(CH2CH3)2、硝基、氰基、C1-C6烷基、C1-C6烷氧基、C1-C6卤代烷基、C1-C6卤代烷氧基、C2-C6烯基、C2-C6炔基、C3-C6环烷基、3-6元杂环基。Ra is each independently selected from: halogen, OH, NH 2 , NHCH 3 , NHCH 2 CH 3 , N(CH 3 ) 2 , N(CH 2 CH 3 ) 2 , nitro, Cyano, C1-C6 alkyl, C1-C6 alkoxy, C1-C6 haloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3- 6-membered heterocyclic group.
  3. 如权利要求1所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,选自: The compound of claim 1 or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug, characterized in that, Selected from:
    选自: 其中,Rp3、Rp4、Rz和R’4的定义如权利要求1所述。 Selected from: Wherein, Rp 3 , Rp 4 , Rz and R' 4 are as defined in claim 1 .
  4. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,W环选自取代或未取代的下组基团:苯基、萘基、吡啶基、嘧啶基、吲哚基、异吲哚基、吲唑基、喹啉基、 其中,所述取代是指被一个或多个Ra取代,Ra 的定义如权利要求2所述。The compound of claim 2 or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, wherein the W ring is selected from the following substituted or unsubstituted groups: phenyl, naphthalene base, pyridyl, pyrimidinyl, indolyl, isoindolyl, indazolyl, quinolyl, Wherein, the substitution refers to being replaced by one or more Ra, Ra is as defined in claim 2.
  5. 如权利要求1所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,R1选自:取代或未取的5-6元含N单环杂环基(优选氮杂环丁基、四氢吡咯基、哌啶基、哌嗪基等)、取代或未取的7-10元双环或三环(如螺环、稠环、桥环)杂环基、取代或未取的-(CH2)1-3-(5-6元含N单环杂环基(优选氮杂环丁基、四氢吡咯基、哌啶基、哌嗪基等)、取代或未取的-(CH2)1-3-(7-10元双环或三环(如螺环、稠环、桥环)杂环基),其中,所述取代是指被一个或多个Ra取代,Ra的定义如权利要求1所述。The compound of claim 1 or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug, characterized in that R 1 is selected from: substituted or unsubstituted 5-6 membered N-containing monocyclic rings Heterocyclyl (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.), substituted or unsubstituted 7-10 membered bicyclic or tricyclic rings (such as spiro ring, fused ring, bridged ring) Heterocyclyl, substituted or unsubstituted -(CH 2 ) 1-3 -(5-6 membered N-containing monocyclic heterocyclyl (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl etc.), substituted or unsubstituted -(CH 2 ) 1-3 -(7-10 membered bicyclic or tricyclic (such as spirocyclic, fused ring, bridged ring) heterocyclyl), wherein the substitution means by One or more Ra are substituted, Ra is as defined in claim 1.
  6. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,R’1选自:C1-C6烷基NH2、C1-C6烷基NHC1-C6烷基、取代或未取的5-6元含N单环杂环基(优选氮杂环丁基、四氢吡咯基、哌啶基、哌嗪基等)、取代或未取的7-10元双环或三环(如螺环、稠环、桥环)杂环基、取代或未取的-(CH2)1-3-(5-6元含N单环杂环基(优选氮杂环丁基、四氢吡咯基、哌啶基、哌嗪基等)、取代或未取的-(CH2)1-3-(7-10元双环或三环(如螺环、稠环、桥环)杂环基),其中,所述取代是指被一个或多个Ra取代,Ra的定义如权利要求2所述。The compound of claim 2 or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug, characterized in that R' 1 is selected from: C1-C6 alkyl NH 2 , C1-C6 alkyl NHC1-C6 alkyl group, substituted or unsubstituted 5-6 membered N-containing monocyclic heterocyclic group (preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.), substituted or unsubstituted 7-10 membered bicyclic or tricyclic (such as spirocyclic, fused ring, bridged ring) heterocyclyl, substituted or unsubstituted -(CH 2 ) 1-3 -(5-6 membered N-containing monocyclic heterocyclyl (Preferably azetidinyl, tetrahydropyrrolyl, piperidinyl, piperazinyl, etc.), substituted or unsubstituted -(CH 2 ) 1-3 -(7-10 membered bicyclic or tricyclic ring (such as spirocyclic ring) , fused ring, bridged ring) heterocyclyl), wherein the substitution means substitution by one or more Ra, and Ra is as defined in claim 2.
  7. 如权利要求1所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,R1选自:其中,Rq选自:F、Cl。The compound of claim 1 or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug, characterized in that R 1 is selected from: Among them, Rq is selected from: F, Cl.
  8. 如权利要求1所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,R1选自: The compound of claim 1 or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug, characterized in that R 1 is selected from:
  9. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,R’1选自: The compound of claim 2 or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug, characterized in that R'1 is selected from:
  10. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,R’5和R’6各自独立地选自:氢、卤素、羟基、硝基、氰基、C1-C6烷基、C3-C6环烷基、C1-C6烷氧基、-NH2、-NHCH3、-NHCH(CH3)2、-C(O)NH2、-C(O)NHCH3、-C(O)CH(CH3)2、-P(O)(CH3)2、-P(O)(CH2CH3)2、-S(O)2NH2、-S(O)2NHCH3、-NHC(O)O CH3、-S(O)(NH)CH3、-NHC(O)CH3、-C(O)NH CH2CH2OH。The compound of claim 2 or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, wherein R' 5 and R' 6 are each independently selected from: hydrogen, halogen, hydroxyl , nitro, cyano, C1-C6 alkyl, C3-C6 cycloalkyl, C1-C6 alkoxy, -NH 2 , -NHCH 3 , -NHCH(CH 3 ) 2 , -C(O)NH2, -C(O)NHCH 3 , -C(O)CH(CH 3 ) 2 , -P(O)(CH 3 ) 2 , -P(O)(CH 2 CH 3 ) 2 , -S(O) 2 NH 2 , -S(O ) 2 NHCH 3 , -NHC(O)O CH 3 , -S(O)(NH)CH 3 , -NHC(O)CH 3 , -C(O)NH CH 2 CH 2 OH.
  11. 如权利要求2所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,选自: The compound of claim 2 or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug, characterized in that, Selected from:
  12. 如权利要求1或2所述的化合物或其药学上可接受的盐、立体异构体、溶剂化物或前药,其特征在于,所述化合物选自下述化合物中的任一种:










    The compound or its pharmaceutically acceptable salt, stereoisomer, solvate or prodrug according to claim 1 or 2, characterized in that the compound is selected from any one of the following compounds:










  13. 一种药物组合物,其特征在于,所述药物组合物包括如权利要求1-12中任一项所述的化合物,或者其药学上可接受的盐、立体异构体、溶剂化物或前药;和药学上可接受的载体。A pharmaceutical composition, characterized in that the pharmaceutical composition includes the compound according to any one of claims 1-12, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof ; and a pharmaceutically acceptable carrier.
  14. 如权利要求1-12中任一项所述的化合物,或其药学上可接受的盐、立体异构体、溶剂化物或前药,或如权利要求13所述的药物组合物在制备治疗抗肿瘤药物中的用途。The compound according to any one of claims 1 to 12, or a pharmaceutically acceptable salt, stereoisomer, solvate or prodrug thereof, or the pharmaceutical composition according to claim 13 for the preparation of a treatment for Use in oncology drugs.
  15. 如权利要求14所述的用途,其特征在于,所述肿瘤选自:结直肠癌、乳腺癌、肺癌、卵巢癌或胃癌,优选地为乳腺癌。 The use according to claim 14, wherein the tumor is selected from the group consisting of colorectal cancer, breast cancer, lung cancer, ovarian cancer or gastric cancer, preferably breast cancer.
PCT/CN2023/094038 2022-06-24 2023-05-12 Small molecule compound having pyrimidothiophene structure and use thereof WO2023246371A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210731231.4 2022-06-24
CN202210731231 2022-06-24

Publications (1)

Publication Number Publication Date
WO2023246371A1 true WO2023246371A1 (en) 2023-12-28

Family

ID=89379115

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/094038 WO2023246371A1 (en) 2022-06-24 2023-05-12 Small molecule compound having pyrimidothiophene structure and use thereof

Country Status (1)

Country Link
WO (1) WO2023246371A1 (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090118276A1 (en) * 2007-11-02 2009-05-07 Wyeth Thienopyrimidines, thienopyridines, and pyrrolopyrimidines as b-raf inhibitors
CN102712656A (en) * 2009-10-22 2012-10-03 韩国科学技术研究院 2,7-substituted thieno[3,2-d]pyrimidine compounds as protein kinase inhibitors
US20130012703A1 (en) * 2009-11-19 2013-01-10 Tae Bo Sim 2,4,7-substituted thieno[3,2-d]pyrimidine compounds as protein kinase inhibitors
CN104292242A (en) * 2014-09-18 2015-01-21 广州必贝特医药技术有限公司 Pyrantel compound and preparation, as well as preparation method and application of pyrantel compound and preparation
US20180153894A1 (en) * 2015-06-26 2018-06-07 Dana-Farber Cancer Institute, Inc. Fused bicyclic pyrimidine derivatives and uses thereof
CN112661745A (en) * 2020-07-24 2021-04-16 浙江同源康医药股份有限公司 Compounds useful as CDK7 kinase inhibitors and uses thereof
CN113226306A (en) * 2018-11-01 2021-08-06 希洛斯医药品股份有限公司 Methods of treating cancer in a patient identified by a biomarker using a non-covalent inhibitor of cyclin dependent kinase 7(CDK7)
CN113557017A (en) * 2018-12-28 2021-10-26 达纳-法伯癌症研究所股份有限公司 Inhibitors of cyclin-dependent kinase 7 and uses thereof
WO2022089444A1 (en) * 2020-10-28 2022-05-05 恒元生物医药科技(苏州)有限公司 Nitrogen-containing heterocyclic compound and application thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090118276A1 (en) * 2007-11-02 2009-05-07 Wyeth Thienopyrimidines, thienopyridines, and pyrrolopyrimidines as b-raf inhibitors
CN102712656A (en) * 2009-10-22 2012-10-03 韩国科学技术研究院 2,7-substituted thieno[3,2-d]pyrimidine compounds as protein kinase inhibitors
US20130012703A1 (en) * 2009-11-19 2013-01-10 Tae Bo Sim 2,4,7-substituted thieno[3,2-d]pyrimidine compounds as protein kinase inhibitors
CN104292242A (en) * 2014-09-18 2015-01-21 广州必贝特医药技术有限公司 Pyrantel compound and preparation, as well as preparation method and application of pyrantel compound and preparation
US20180153894A1 (en) * 2015-06-26 2018-06-07 Dana-Farber Cancer Institute, Inc. Fused bicyclic pyrimidine derivatives and uses thereof
CN113226306A (en) * 2018-11-01 2021-08-06 希洛斯医药品股份有限公司 Methods of treating cancer in a patient identified by a biomarker using a non-covalent inhibitor of cyclin dependent kinase 7(CDK7)
CN113557017A (en) * 2018-12-28 2021-10-26 达纳-法伯癌症研究所股份有限公司 Inhibitors of cyclin-dependent kinase 7 and uses thereof
CN112661745A (en) * 2020-07-24 2021-04-16 浙江同源康医药股份有限公司 Compounds useful as CDK7 kinase inhibitors and uses thereof
WO2022089444A1 (en) * 2020-10-28 2022-05-05 恒元生物医药科技(苏州)有限公司 Nitrogen-containing heterocyclic compound and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CHO HANNA, SHIN INJAE, YOON HOJONG, JEON EUNHYE, LEE JIWON, KIM YOUNGHOON, RYU SEONGSHICK, SONG CHIMAN, KWON NAM HOON, MOON YOUNGJ: "Identification of Thieno[3,2- d ]pyrimidine Derivatives as Dual Inhibitors of Focal Adhesion Kinase and FMS-like Tyrosine Kinase 3", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 64, no. 16, 26 August 2021 (2021-08-26), US , pages 11934 - 11957, XP093118556, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.1c00459 *

Similar Documents

Publication Publication Date Title
JP7446348B2 (en) Tetrahydro-pyrido[3,4-b]indole estrogen receptor modulators and uses thereof
JP6522807B2 (en) Benzoxazepine oxazolidinone compounds and methods of use
JP7018026B2 (en) Heteroarylestrogen receptor modulator and its use
RU2581834C1 (en) New piperidine compound or salt thereof
US20190152970A1 (en) TETRAHYDRO-PYRIDO[3,4-b]INDOLE ESTROGEN RECEPTOR MODULATORS AND USES THEREOF
CN109219604A (en) Tetrahydroisoquinoline estrogenic agents and application thereof
JP2020105191A (en) Optically active azabicyclo ring derivatives
TW202137979A (en) Anti-tumor effect potentiator using novel biphenyl compound
WO2023078451A1 (en) Compound used as cdk7 kinase inhibitor and use thereof
JP6554480B2 (en) Oxepan-2-yl-pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
WO2024002373A1 (en) Substituted pyrimidine-fused ring inhibitor, method for preparing same, and use thereof
WO2023246371A1 (en) Small molecule compound having pyrimidothiophene structure and use thereof
EP4353724A1 (en) Compound as cdk kinase inhibitor and use thereof
CA3234429A1 (en) Ras inhibitors, compositions and methods of use thereof
CN111372927A (en) Tricyclic compounds as histone methyltransferase inhibitors
JP2021501778A (en) Pyridopyrimidine compounds as mTORC1 / 2 double inhibitors
WO2023151635A1 (en) Compound based on quinazoline-substituted glutarimide skeleton and use thereof
WO2023245015A2 (en) Phenyl amide compounds and methods of use
WO2024008083A1 (en) Compound as cdk7 kinase inhibitor and use thereof

Legal Events

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

Ref document number: 23826020

Country of ref document: EP

Kind code of ref document: A1