CN111732585A - Cyclic sulfonamide ring-substituted pyridone pyrrole compound and synthesis method and application thereof - Google Patents

Cyclic sulfonamide ring-substituted pyridone pyrrole compound and synthesis method and application thereof Download PDF

Info

Publication number
CN111732585A
CN111732585A CN201910228878.3A CN201910228878A CN111732585A CN 111732585 A CN111732585 A CN 111732585A CN 201910228878 A CN201910228878 A CN 201910228878A CN 111732585 A CN111732585 A CN 111732585A
Authority
CN
China
Prior art keywords
compound
alkyl
group
hydroxy
heterocyclyl
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
CN201910228878.3A
Other languages
Chinese (zh)
Inventor
张翱
谭文福
刘晓华
张宇
宋子兰
王娟
杨君
黄文静
李阳
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Fudan University
Shanghai Institute of Materia Medica of CAS
Original Assignee
Fudan University
Shanghai Institute of Materia Medica of CAS
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 Fudan University, Shanghai Institute of Materia Medica of CAS filed Critical Fudan University
Priority to CN201910228878.3A priority Critical patent/CN111732585A/en
Publication of CN111732585A publication Critical patent/CN111732585A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Emergency Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Endocrinology (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to a cyclic sulfonamide ring substituted pyridone pyrrole compound and a synthesis method and application thereof. Specifically, the compound has a structure shown in a formula I, wherein the definition of each group and substituent is described in the specification. The invention also discloses a preparation method of the compound and application of the compound in preventing and/or treating diseases related to Hedgehog signal channel abnormality.

Description

Cyclic sulfonamide ring-substituted pyridone pyrrole compound and synthesis method and application thereof
Technical Field
The invention relates to the field of medicaments, in particular to a cyclic sulfonamide ring-substituted pyridone pyrrole compound and a synthesis method and application thereof.
Background
Basal Cell Carcinoma (BCC) is a type of keratinocyte tumor, which is frequently found in the head and one of the most common tumors in humans, accounting for over 90% of skin cancers. Intensive studies of molecular mechanisms have gradually revealed heterogeneous (heterologous) features of basal cell carcinogenesis, progression. The research finds that the main molecular pathological changes driving the occurrence and development of basal cell carcinoma are the mutation or amplification of key signal molecules of a Hedgehog (Hh) signal pathway and the mutation of proteins such as MYCN, P53, PIK3CA, RAS and the like. The Hh signaling pathway is a highly conserved signaling pathway that plays an important role in important physiological phenomena such as embryonic development and tissue remodeling. Activation of the Hh signaling pathway is a cascade of reactions involving spatially and temporally ordered actions of a variety of proteins. Ligand Hh binds to Patched (Ptch) receptor of 12 times transmembrane domain, relieves the inhibition effect of Ptch on G-protein coupled receptor smoothened (smo) in the signal path, leads the protein to be translocated to clium at key signal center on cell membrane, leads the complex of Sufu and Gli to be translocated to the clium, further separates the Sufu from Gli, relieves the inhibition effect of the Sufu on Gli, finally promotes Gli with transcriptional activity to enter nucleus, binds to GACCACCCA area in promoter area at the upstream of transcriptional target gene, stimulates the expression of Gli1, Ptch1, Bcl-2 and the like, thereby playing a role in the pathological physiological phenomena of proliferation, survival, metastasis and the like of a plurality of tumor target genes.
In 2012 and 2015, two Smo protein-targeting Hh signaling pathway small molecule inhibitors, Vismodegib (GDC-0449, Vismodegib) and sonegib (LDE-225, Sonidegib), were approved by FDA for marketing, respectively, for the treatment of basal cell carcinoma in which ptch mutation leads to ligand-independent Hh aberrant activation, with a 30% remission rate of Vismodegib on patients with metastatic basal cell carcinoma in which ptch mutation leads to Hh aberrant activation and a 44% remission rate of sonegib on patients with locally advanced basal cell carcinoma. And primary resistance to subpopulations of basal cell carcinomas due to abnormal genetic alterations of SUFU, SMO, and Gli 2. Meanwhile, the patients lose the curative effect by the SMO mutation at the D473H point, the Gli2 amplification, the alternative activation of the PI3K signal path and the drug resistance caused by the ccnd1 amplification within 3 months of using the vismodegib.
The transcription factor Gli is the very end signal molecule of the Hh pathway, and its protein family mediates the activity of all Hh pathways. In vertebrates, the Gli protein family has at least 3 alleles: gli1, Gli2, Gli 3. Among them, Gli1 and Gli2 are major gene activators of Hh pathway and are responsible for activating transcription of target genes, while Gli3 plays a role in inhibiting transcription. Researches show that Gli1/2 is highly expressed in tumors including basal cell carcinoma and the like, a downstream transcription factor Gli of a targeted Hh signal path is likely to become an ideal means for treating basal cell carcinoma, and the Gli inhibitor not only can overcome primary drug resistance of a Smo inhibitor, but also can overcome secondary drug resistance and improve clinical effective rate, but no Gli inhibitor with excellent in vivo and in vitro activity is successfully developed so far.
Disclosure of Invention
The invention provides a pyridone pyrrole compound containing cyclic sulfamide substitution, and pharmaceutically acceptable salt or pharmaceutically acceptable solvate thereof. The compound is a novel Hedgehog (Hh) signal pathway inhibitor, has a good inhibition effect on transcription factor Gli, and has the potential of further researching and treating Hh pathway abnormality related diseases.
In a first aspect of the invention, a cyclic sulfonamide substituted pyridone pyrrole compound shown in formula I, or an isomer, a pharmaceutically acceptable salt, a pharmaceutically acceptable solvate or hydrate thereof is provided,
Figure BDA0002006087980000021
wherein:
n is an integer selected from the group consisting of: 0.1, 2 and 3;
x is selected from the group consisting of: CH. N;
l is selected from the group consisting of: o, NR5、C(O)、-(CH2)n-、-S(O)m-、-O(CR6R7)n-、-NR5(CR7)n-;
R1、R2、R3、R4Each independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6 alkyl,Halogenated C1-C6 alkyl, deuterated C1-C6 alkyl, C2-C6 alkenyl, C1-C6 alkoxy, halogenated C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, halogen, amino, nitro, hydroxy, cyano, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, -OR8、-C(O)R8、-C(O)OR8、-S(O)mR8、-NR9R10、-C(O)NR9R10、-NR9C(O)R10、-NR9S(O)mR10(ii) a Wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogenated C1-C6 alkyl, halogen, amino, nitro, cyano, hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, halogenated C3-C8 cycloalkyl, heterocyclyl, - (C1-C6 alkylene) hydroxy-substituted heterocyclyl, (C1-C6 alkyl) -substituted heterocyclyl, C6-C10 aryl, heteroaryl, -OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R5Selected from the group consisting of: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
R6and R7The same or different and each is independently selected from the group consisting of: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, halogen, amino, nitro, hydroxy, cyano, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, -OR8、-C(O)R8、-C(O)OR8、-S(O)mR8、-NR9R10、-C(O)NR9R10、-NR9C(O)R10、-NR9S(O)mR10
Or, R6And R7C adjacent thereto may form a substituted or unsubstituted 3-to 8-membered cycloalkyl or heterocyclyl group, whereinThe substituent means being substituted with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, halogen, amino, nitro, hydroxy, cyano, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R8Selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, C2-C6 alkenyl, hydroxyl, amino, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclic, C6-C10 aryl, heteroaryl; wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, amino, nitro, cyano, hydroxy, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R9And R10The same or different, and each is independently selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, hydroxy, amino, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12Wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, hydroxy, amino, nitro, cyano, C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, heterocyclyl, (C1-C6 alkyl) -substituted heterocyclyl, C6-C10 aryl, heteroaryl;
R11and R12The same or different, and each is independently selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, hydroxy, amino, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, hydroxy, amino, nitro, cyano, C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
m is an integer selected from the group consisting of: 0.1, 2;
wherein said "heterocyclyl" is a 4-8 membered heterocyclyl containing 1,2 or 3 heteroatoms selected from N, O, S;
the "heteroaryl" is a 4-10 membered heteroaryl containing 1,2 or 3 heteroatoms selected from N, O, S.
In another preferred embodiment, n is an integer selected from the group consisting of: 0.1, 2 and 3;
x is selected from the group consisting of: CH. N;
l is selected from the group consisting of: o, NR5
R1、R2、R3、R4Each independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6 alkyl, halogenated C1-C6 alkyl, deuterated C1-C6 alkyl, C1-C6 alkoxy, halogenated C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, halogen, amino, nitro, hydroxy, cyano, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, -OR8、-C(O)R8、-C(O)OR8、-S(O)mR8、-NR9R10、-C(O)NR9R10、-NR9C(O)R10、-NR9S(O)mR10(ii) a Wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogenated C1-C6 alkyl, halogen, amino, nitro, cyano, hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, halogenated C3-C8 cycloalkyl, heterocyclyl, - (C1-C6 alkylene) hydroxy-substituted heterocyclyl, (C1-C6 alkyl) -substituted heterocyclyl, C6-C10 aryl, heteroaryl, -OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R5Selected from the group consisting of: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
R8selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, C2-C6 alkenyl, hydroxyl, amino, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclic, C6-C10 aryl, heteroaryl; wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, amino, nitro, cyano, hydroxy, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R9And R10The same or different, and each is independently selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, hydroxy, amino, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12Wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, hydroxy, amino, nitro, cyano, C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, heterocyclyl, (C1-C6 alkyl) -substituted heterocyclyl, C6-C10 aryl, heteroaryl;
R11and R12Are the same or different and are each independentlySelected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, hydroxy, amino, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, hydroxy, amino, nitro, cyano, C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
m is an integer selected from the group consisting of: 0.1, 2;
wherein said "heterocyclyl" is a 4-8 membered heterocyclyl containing 1,2 or 3 heteroatoms selected from N, O, S;
the "heteroaryl" is a 4-10 membered heteroaryl containing 1,2 or 3 heteroatoms selected from N, O, S.
In another preferred embodiment, L is selected from the group consisting of: o, NR5
R5Selected from the group consisting of: hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl.
In another preferred embodiment, R1、R2、R3、R4Each independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6 alkyl, halogen, C3-C8 cycloalkyl, C6-C10 aryl, -C (O) OR8、-NR9R10、-C(O)NR9R10(ii) a Wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogenated C1-C6 alkyl, halogen, hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, halogenated C3-C8 cycloalkyl, heterocyclyl, - (C1-C6 alkylene) hydroxy-substituted heterocyclyl, (C1-C6 alkyl) -substituted heterocyclyl, -NR11R12、-C(O)NR11R12
R8Selected from the group consisting of: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, C3-C8 cycloalkyl;
R9and R10The same or different, and each is independently selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, C3-C8 cycloalkyl, heterocyclyl, (C1-C6 alkyl)) Substituted heterocyclyl, C6-C10 aryl, heteroaryl;
R11and R12The same or different, and each is independently selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, hydroxy, C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl;
m is an integer selected from the group consisting of: 0.1 and 2.
In another preferred embodiment, L is O.
In another preferred embodiment, L is NR5
R5Selected from the group consisting of: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
R4selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6 alkyl, halogenated C1-C6 alkyl, deuterated C1-C6 alkyl, C3-C8 cycloalkyl, heterocyclic, C6-C10 aryl, heteroaryl; wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogenated C1-C6 alkyl, halogen, C3-C8 cycloalkyl, and halogenated C3-C8 cycloalkyl.
In another preferred embodiment, the compound is selected from the compounds listed in table 1.
In a second aspect of the present invention, there is provided a process for the preparation of a compound according to the first aspect of the present invention, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof, selected from the group consisting of:
the method comprises the following steps:
Figure BDA0002006087980000061
removing a protecting group R' from the compound of the formula A under the condition of NaOH solution to obtain a compound of a formula I,
wherein, n, X, L, R1、R2、R3、R4As defined in the first aspect of the invention;
R' is Ts;
the second method comprises the following steps:
Figure BDA0002006087980000062
cyclizing the compound of the formula B under the condition of NaOH solution to remove the protecting group R' to obtain the compound of the formula I,
wherein, n, X, L, R1、R2、R3、R4As defined in the first aspect of the invention;
r' is Ts;
the third method comprises the following steps:
Figure BDA0002006087980000071
cyclizing the compound of the formula C under the condition of NaOH solution to remove the protecting group R' to obtain the compound of the formula I,
wherein, n, X, L, R1、R2、R3、R4As defined in the first aspect of the invention;
r' is Ts.
In a third aspect of the invention, there is provided a pharmaceutical composition comprising a pharmaceutically acceptable carrier and one or more compounds of the first aspect of the invention, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof.
In a fourth aspect of the present invention, there is provided a use of the pharmaceutical composition according to the third aspect of the present invention for the preparation of a formulation for the prevention and/or treatment of a disease associated with an abnormal Hedgehog signaling pathway.
In another preferred embodiment, the disease associated with an abnormality in the Hedgehog signaling pathway is selected from the group consisting of: cancer, inflammation, chronic liver disease, diabetes, cardiovascular disease.
In another preferred embodiment, the cancer is selected from the group consisting of: basal cell carcinoma, medulloblastoma, rhabdomyosarcoma, acute leukemia, acute lymphocytic leukemia, acute myelogenous leukemia, acute t-cell leukemia, cholangiocarcinoma, bladder carcinoma, brain carcinoma, breast carcinoma, bronchial carcinoma, cervical carcinoma, chondrosarcoma, chordoma, choriocarcinoma, chronic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, colon carcinoma, colorectal carcinoma, craniopharyngioma, cystadenocarcinoma, diffuse large B-cell lymphoma, embryonic carcinoma, endometrial carcinoma, endothelial sarcoma, epithelial carcinoma, erythroleukemia, esophageal carcinoma, estrogen receptor positive breast carcinoma, primary thrombocytosis, Ewing's sarcoma, fibrosarcoma, follicular lymphoma, germ cell testicular carcinoma, glioma, glioblastoma, glioma, Liver cancer, stomach cancer, large intestine cancer, leiomyosarcoma, liposarcoma, lymphoma (hodgkin and non-hodgkin), thyroid cancer, and malignant and hyperproliferative disorders of the bladder, breast, colon, lung, ovary, pancreas, prostate, skin and/or uterus.
In another preferred embodiment, the cardiovascular disease is selected from the group consisting of: dyslipidemia, arrhythmia, atherosclerosis, hypertension and coronary heart disease (e.g. asymptomatic myocardial ischemia, angina pectoris, myocardial infarction, ischemic heart failure and sudden cardiac death).
In a fifth aspect of the present invention, there is provided a Gli inhibitor comprising a pharmaceutically acceptable carrier and an inhibitory effective amount of one or more compounds of the first aspect of the present invention, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Detailed Description
The present inventors have made extensive and intensive studies for a long time and have unexpectedly prepared a cyclic sulfonamide ring-substituted pyridone pyrrole compound having an excellent inhibitory activity against transcription factor Gli. On this basis, the inventors have completed the present invention.
Term(s) for
In the present invention, unless otherwise specified, the terms used have the ordinary meanings well known to those skilled in the art.
In the present invention, the term "halogen" means F, Cl, Br or I.
In the present invention, "C1-C6 alkyl" means a straight or branched chain alkyl group including 1 to 6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, neopentyl, tert-pentyl, or the like.
In the present invention, the term "C3-C8 cycloalkyl" refers to a cyclic alkyl group having 3 to 8 carbon atoms in the ring, including, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like.
In the present invention, the term "aromatic ring" or "aryl" has the same meaning, and is preferably "C6-C10 aryl". The term "C6-C10 aryl" refers to an aromatic ring group having 6 to 10 carbon atoms, such as phenyl, naphthyl, and the like, that does not contain heteroatoms in the ring.
In the present invention, the term "heteroaromatic ring" or "heteroaryl" has the same meaning and refers to a heteroaromatic group containing one to more heteroatoms. For example, "C3-C10 heteroaryl" refers to an aromatic heterocycle containing 1 to 4 heteroatoms selected from oxygen, sulfur and nitrogen and 3 to 10 carbon atoms. Non-limiting examples include: furyl, thienyl, pyridyl, pyrazolyl, pyrrolyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, imidazolyl, tetrazolyl, and the like. The heteroaryl ring may be fused to an aryl, heterocyclyl, or cycloalkyl ring, wherein the ring joined to the parent structure is a heteroaryl ring. Heteroaryl groups may be optionally substituted or unsubstituted.
In the present invention, the term "heterocyclyl" is a 4-8 membered heterocyclyl containing 1,2 or 3 heteroatoms selected from N, O, S, including (but not limited to) the following groups:
Figure BDA0002006087980000091
in the present invention, the term "halo" means substituted by halogen.
In the present invention, the term "deuterated" means substituted by deuterium.
In the present invention, the term "C2-C6 alkenyl group" means a straight or branched chain alkenyl group having 2 to 6 carbon atoms containing one double bond, including, but not limited to, ethenyl, propenyl, butenyl, isobutenyl, pentenyl, hexenyl and the like.
In the present invention, the term "C2-C6 alkynyl" refers to a straight or branched alkynyl group having 2 to 6 carbon atoms containing one triple bond, including, but not limited to, ethynyl, propynyl, butynyl, isobutynyl, pentynyl, hexynyl, and the like.
In the present invention, the term "C1-C6 alkoxy" means a straight or branched chain alkoxy group having 1 to 6 carbon atoms, including, but not limited to, methoxy, ethoxy, propoxy, isopropoxy, butoxy and the like. C1-C4 alkoxy is preferred.
In the present invention, the term "substituted" means that one or more hydrogen atoms on a specified group are replaced with a specified substituent. Particular substituents are those described correspondingly in the foregoing, or as appearing in the examples. Unless otherwise specified, a certain substituted group may have one substituent selected from a specific group at any substitutable site 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 the combinations of substituents contemplated by the present invention are those that are stable or chemically achievable. Such substituents are for example (but not limited to): halogen, hydroxyl, carboxyl (-COOH), C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-to 12-membered heterocyclyl, aryl, heteroaryl, C1-C8 aldehyde, C2-C10 acyl, C2-C10 ester, amino, C1-C6 alkoxy, C1-C10 sulfonyl and the like.
Compound (I)
The invention provides a cyclic sulfamide substituted pyridone pyrrole compound shown in formula I, or an isomer, a pharmaceutically acceptable salt, a pharmaceutically acceptable solvate or hydrate thereof,
Figure BDA0002006087980000101
wherein, n, X, L, R1、R2、R3、R4As defined above.
In another preferred embodiment, in the compound, n, X, L and R1、R2、R3、R4Each of which is a group corresponding to a specific compound described in table 1.
In another preferred embodiment, the compound is preferably the compound prepared in the examples.
In another preferred embodiment, the compound is selected from the compounds listed in table 1.
TABLE 1
Figure BDA0002006087980000102
Figure BDA0002006087980000111
Figure BDA0002006087980000121
As used herein, the term "pharmaceutically acceptable salt" refers to a salt of a compound of the present invention with an acid or base that is suitable for use as a pharmaceutical. Pharmaceutically acceptable salts include inorganic and organic salts. One preferred class of salts is that formed by reacting a compound of the present invention with an acid. Suitable acids for forming the salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, and the like; organic acids such as formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid and the like; and amino acids such as proline, phenylalanine, aspartic acid, glutamic acid, etc.
Another preferred class of salts are those of the compounds of the invention with bases, for example alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., magnesium or calcium salts), ammonium salts (e.g., lower alkanolammonium salts and other pharmaceutically acceptable amine salts), for example methylamine salts, ethylamine salts, propylamine salts, dimethylamine salts, trimethylamine salts, diethylamine salts, triethylamine salts, tert-butylamine salts, ethylenediamine salts, hydroxyethylamine salts, dihydroxyethylamine salts, triethanolamine salts, and amine salts formed from morpholine, piperazine, lysine, respectively.
The term "solvate" refers to a complex of a compound of the present invention coordinated to molecules of a solvent including, but not limited to, ethanol, isopropanol, diethyl ether, acetone, in a specific ratio. "hydrate" refers to a complex formed by the coordination of a compound of the present invention with water.
Preparation method
The process for the preparation of the compounds of formula I according to the invention is described in more detail below, but these particular processes do not limit the invention in any way. The compounds of the present invention may also be conveniently prepared by optionally combining various synthetic methods described in the present specification or known in the art, and such combinations may be readily carried out by those skilled in the art to which the present invention pertains.
Typically, the process for the preparation of the compounds of the present invention is as follows, wherein the starting materials and reagents used are commercially available without specific reference.
The preparation method comprises the following steps:
Figure BDA0002006087980000131
(i) substituting the compound a with chlorosulfonyl isocyanate to obtain a compound b;
(ii) deprotecting a compound obtained by substituting a compound b with dihaloalkane c under a TFA condition to obtain a compound d;
(iii) compounds d with halogenated alkanes R1X1And (4) removing a protecting group from the cyclized compound under the condition of a sodium hydroxide solution to obtain a compound e.
In the formulae (III), (III) L, R1、R2、R3、R4N is as defined above;X1、X2The same or different and each is independently selected from chlorine, bromine, iodine.
The second preparation method comprises the following steps:
Figure BDA0002006087980000141
(i) carrying out substitution reaction on the compound a and the compound f to obtain a compound g;
(ii) and (3) removing a protecting group of the compound g under the condition of sodium hydroxide solution and cyclizing to obtain a compound h.
In the formulae (III), (III) L, R2、R3、R4N is as defined above.
The preparation method comprises the following steps:
Figure BDA0002006087980000142
(i) substituting compound f of the compound i and then removing protecting groups to obtain a compound j;
(ii) compounds j and R8OH is condensed to obtain a compound k;
(iii) compounds k and R9R10OH is condensed to obtain a compound l.
In the formulae (III), (III) L, R4、R8、R9、R10N is as defined above.
The preparation method comprises the following steps:
Figure BDA0002006087980000151
(i) the compound a and 2- (chlorosulfonyl) methyl acetate are substituted to obtain a compound o;
(ii) cyclizing the compound o and dihalogenated alkane c to obtain a compound p;
(iii) compound p in NaBH4And CaCl2Reducing the obtained compound under the condition of (1) and removing a protecting group by NaOH to obtain a compound q.
In the formulae (III), (III) L, R1、R2、R3、R4N is as defined above; x1、X2The same or different and each is independently selected from chlorine, bromine, iodine.
The preparation method comprises the following steps:
Figure BDA0002006087980000152
and performing electrophilic fluorine substitution on the compound r and 1-fluoro-2, 6-dichloropyridine tetrafluoroborate to obtain a compound s.
Pharmaceutical compositions and methods of administration
The invention also provides a pharmaceutical composition, which comprises a pharmaceutically acceptable carrier and one or more compounds, or isomers, pharmaceutically acceptable salts, or pharmaceutically acceptable solvates or hydrates thereof.
The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof in a safe and effective amount range and a pharmacologically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 1-2000mg of a compound of the invention per dose, more preferably, 10-1000mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"pharmaceutically acceptable carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of intermixing with and with the compounds of the present invention without significantly diminishing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g., sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g., stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g., soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (e.g., propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (e.g., propylene glycol, glycerin, mannitol, sorbitol, etc.), and the like
Figure BDA0002006087980000161
) Wetting agents (e.g., sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
The medicine composition is injection, capsule, tablet, pill, powder or granule.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration.
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 extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings 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 delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may 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. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, 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 comprise 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 nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The compound can be independently administered or combined with other pharmaceutically acceptable compounds (such as antitumor drugs).
The treatment methods of the present invention can be administered alone or in combination with other therapeutic means or agents.
When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) to be treated, wherein the administration dose is a pharmaceutically-considered effective administration dose, and for a human body with a weight of 60kg, the daily administration dose is usually 1 to 2000mg, preferably 50 to 1000 mg. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.
Compared with the prior art, the invention has the following main advantages:
(1) the compound is different from the existing Hedgehog signal pathway inhibitor and is a novel structure type inhibitor acting on the downstream of a Hedgehog pathway;
(2) the compound has excellent inhibition effect on a Hedgehog signal path and/or a transcription factor Gli, and the IC of a plurality of compounds50Values below 1nM and better than the positive compound;
(3) the compound has better inhibition effect on various cell types, especially on SKM-1 cells, and the proliferation inhibition effect of the compounds S32 and S33 on SKM-1 cells is better than that of positive compounds;
(4) the compound pair Ptch +/-; the P53-/-medulloblastoma has obvious inhibiting effect, and the compounds S1, S15 and S16 have the tumor inhibiting effect equivalent to that of positive compounds, and have better safety than positive compounds.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, molecular cloning is generally performed according to conventional conditions such as Sambrook et al: the conditions described in the laboratory Manual (New York: Cold Spring Harbor laboratory Press,1989), or according to the manufacturer's recommendations. Unless otherwise indicated, percentages and parts are by weight.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention. The preferred embodiments and materials described herein are intended to be exemplary only.
1H-NMR measurement with a Varian MercuryAMX300, 400 or 500 model apparatusAll solvents are redistilled before use, and the used anhydrous solvents are obtained by drying according to a standard method; all reactions were carried out under nitrogen protection and TLC tracking except for the indication, and the post-treatment was carried out by washing with saturated sodium chloride aqueous solution and drying with anhydrous sodium sulfate; purification of the product except for the indication silica gel (200 and 300 mesh) column chromatography was used; wherein the silica gel (200-300 mesh) is produced by Qingdao ocean factory, and the GF-254 thin layer silica gel plate is produced by Yangttai Jiangyou silica gel development company Limited.
Preparation example 1 preparation of compound S1
Figure BDA0002006087980000181
Reference method for the Synthesis of Compound 1-1 (J.Med.chem.2017,60,3828-3850.)
Synthesis of Compounds 1-2:
chlorosulfonyl isocyanate (1.1eq) dissolved in CH2Cl2T-butanol (1.1eq) was added with pre-cooling at 0 ℃ and then stirred for half an hour. In another reaction flask, compound 1-1(1eq) was dissolved in CH2Cl2To this solution, triethylamine (1.2eq) was added, and the reaction mixture was added dropwise and stirred at room temperature for 2 hours. Quenching with water after the reaction is completed, extracting with dichloromethane for three times, combining organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with a sample, loading onto a column, and performing reaction on the mixture with CH2Cl2MeOH is 100:1 to 30:1, and the compound 1-2 is obtained.
Synthesis of Compounds 1-3:
compound 1-2(1eq) was dissolved in acetonitrile, and 1, 2-dibromoethane (1.2eq) and potassium carbonate (2.2eq) were added, followed by heating under reflux and stirring overnight. Quenching with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2EA [ ethyl acetate ]]Getting compound 1-3 with 10: 1.
Synthesis of Compounds 1-4:
the compounds 1-3(1eq) were dissolved in dichloromethane, TFA (trifluoroacetic acid, 20eq) was added dropwise and reacted at room temperature for 2 h. After the reaction is completed, the reaction solution is decompressed and steamed in a rotary mode, residues are dissolved by dichloromethane, saturated sodium bicarbonate solution is washed, combined organic phases are washed by saturated salt solution, anhydrous sodium sulfate is dried, the crude product 1-4 is obtained through decompression and rotary steaming, and the next step is directly carried out.
Synthesis of compound S1:
the compound 1-4(1eq) was dissolved in DMF, and potassium carbonate (1.5eq) and iodomethane (1.2eq) were added to react at room temperature for 1 hour. After the reaction is completed, adding water to wash, extracting with ethyl acetate, washing the organic phase with water for 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, and carrying out reduced pressure rotary evaporation to obtain a crude product which is directly put into the next step.
Dissolving the crude product in 1, 4-dioxane, adding 3M NaOH solution, and reacting at 90 deg.C for half an hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S1.
1H NMR(400MHz,DMSO-d6)12.08(s,1H),7.43–7.36(m,2H),7.35(s,1H),7.30(t,J=2.8Hz,1H),7.22(dd,J=8.9,2.9Hz,1H),7.11(td,J=9.2,5.6Hz,1H),7.05–6.97(m,2H),6.29(t,J=2.4Hz,1H),3.88(t,J=6.3Hz,2H),3.55(s,3H),3.49(t,J=6.3Hz,2H),2.73(s,3H).
Preparation example 2 preparation of compound S2
Figure BDA0002006087980000201
Synthesis of compound S2:
compound 1-4(1eq) was dissolved in DMF and cesium carbonate (2eq) and 2-bromoethyl methyl ether (1.2eq) were added and reacted at 60 ℃ for 3 h. Washing with water, extracting with ethyl acetate, washing organic phase with water for 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2MeOH is 40:1 to 30:1 to obtain the intermediate.
The intermediate is dissolved in 1, 4-dioxane, 3M NaOH solution is added, and the reaction is carried out for half an hour at 90 ℃. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S2.
1H NMR(400MHz,CDCl3)11.10(s,1H),7.49(d,J=2.8Hz,1H),7.31(t,J=2.8Hz,1H),7.26(dd,J=8.9,2.9Hz,1H),7.22(s,1H),6.95–6.85(m,3H),6.74(dddd,J=9.2,7.7,2.9,1.6Hz,1H),6.46(dd,J=2.9,2.0Hz,1H),3.84(t,J=6.3Hz,2H),3.73(s,3H),3.71–3.63(m,4H),3.41(s,3H),3.37(dd,J=5.5,4.6Hz,2H).
Preparation example 3 preparation of compound S3
Figure BDA0002006087980000202
Synthesis of compound S3:
compound 1-4(1eq) was dissolved in DMF and cesium carbonate (2eq) and (2-bromoethyl) trimethylammonium bromide (1.2eq) were added and reacted at 60 ℃ for 3 h. Washing with water, extracting with ethyl acetate, washing organic phase with water for 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2MeOH is 40:1 to 30:1 to obtain the intermediate.
The intermediate is dissolved in 1, 4-dioxane, 3M NaOH solution is added, and the reaction is carried out for half an hour at 90 ℃. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S3.
1H NMR(400MHz,CD2Cl2)11.21(s,1H),7.48(d,J=2.8Hz,1H),7.33(t,J=2.8Hz,1H),7.26(dd,J=8.9,2.9Hz,1H),7.24(s,1H),6.99–6.90(m,3H),6.80(dddd,J=9.2,7.9,2.9,1.6Hz,1H),6.46(dd,J=2.9,2.1Hz,1H),3.86(t,J=6.3Hz,2H),3.70(s,3H),3.66(t,J=6.3Hz,2H),3.34(t,J=6.4Hz,2H),2.76(t,J=6.1Hz,2H),2.42(s,6H).
Preparation example 4 preparation of compound S4
Figure BDA0002006087980000211
Synthesis of Compound 4-1:
compound 1-2(1eq) was dissolved in acetonitrile, and 1, 3-dibromopropane (1.2eq) and potassium carbonate (2.2eq) were added, followed by heating under reflux and stirring overnight. Quenching with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2EA [ ethyl acetate ]]Obtaining the compound 4-1 by 10: 1.
Synthesis of Compound 4-2:
compound 4-1(1eq) was dissolved in dichloromethane, TFA (trifluoroacetic acid, 20eq) was added dropwise, and the reaction was carried out at room temperature for 2 h. And after the reaction is completed, carrying out reduced pressure rotary evaporation on the reaction liquid, dissolving the residue by using dichloromethane, washing the residue by using a saturated sodium bicarbonate solution, combining organic phases, washing the combined organic phases by using saturated salt, drying the combined organic phases by using anhydrous sodium sulfate, carrying out reduced pressure rotary evaporation to obtain a crude product 4-2, and directly putting the crude product into the next step.
Synthesis of Compounds 4-3:
the compound 4-2(1eq) was dissolved in DMF, and potassium carbonate (1.5eq) and iodomethane (1.2eq) were added to react at room temperature for 1 hour. After the reaction is completed, adding water to wash, extracting with ethyl acetate, washing the organic phase with water for 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, and carrying out reduced pressure rotary evaporation to obtain a crude product 4-3 which is directly put into the next step.
Synthesis of compound S4:
dissolving the compound 4-3 in 1, 4-dioxane, adding 3M NaOH solution, and reacting at 90 deg.C for half an hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S4.
1H NMR(400MHz,DMSO-d6)12.08(s,1H),7.48–7.40(m,2H),7.35(s,1H),7.30(dt,J=5.2,2.9Hz,2H),7.23(td,J=9.2,5.6Hz,1H),7.11–7.04(m,1H),6.86(d,J=8.8Hz,1H),6.29(dd,J=2.8,2.0Hz,1H),3.69(t,J=5.7Hz,2H),3.56(s,5H),2.87(s,3H),1.97–1.87(m,2H).
Preparation example 5 preparation of Compound S5
Figure BDA0002006087980000221
Synthesis of reference example S2, starting from 4-2, gave compound S5.
1H NMR(400MHz,CDCl3)11.30(s,1H),7.60(d,J=2.7Hz,1H),7.32–7.28(m,2H),7.21(s,1H),6.97–6.87(m,2H),6.84(d,J=8.7Hz,1H),6.80–6.73(m,1H),6.49–6.45(m,1H),3.83–3.74(m,4H),3.73(s,3H),3.63(t,J=5.4Hz,2H),3.47(t,J=5.4Hz,2H),3.38(s,3H),1.96(p,J=6.0Hz,2H).
Preparation example 6 preparation of Compound S6
Figure BDA0002006087980000222
Synthesis of reference example S3, starting from 4-2, gave compound S6.
1H NMR(400MHz,CD2Cl2)10.67(s,1H),7.59(d,J=2.7Hz,1H),7.36–7.31(m,2H),7.23(s,1H),7.04–6.93(m,2H),6.91–6.87(m,1H),6.87–6.81(m,1H),6.47(dd,J=2.9,2.2Hz,1H),3.77(dt,J=13.9,5.7Hz,4H),3.69(s,3H),3.60(s,2H),2.89(s,2H),2.53(s,6H),1.99(p,J=6.2Hz,2H).
Preparation example 7 preparation of compound S7
Figure BDA0002006087980000231
Synthesis of compound S7:
compound 4-2(1eq) was dissolved in DMF, and potassium carbonate (2eq) and 1, 1-difluoro-2-iodoethane (1.2eq) were added to react at 60 ℃ for 3 hours. Washing with water, extracting with ethyl acetate, washing organic phase with water for 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2MeOH is 40:1 to 30:1 to obtain the intermediate.
The intermediate is dissolved in 1, 4-dioxane, 3M NaOH solution is added, and the reaction is carried out for half an hour at 90 ℃. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S7.
1H NMR(400MHz,CDCl3)10.85(s,1H),7.56(d,J=2.7Hz,1H),7.29(t,J=2.9Hz,2H),7.19(s,1H),6.98–6.86(m,2H),6.82(d,J=8.7Hz,1H),6.77(td,J=10.6,9.9,2.2Hz,1H),6.44(dd,J=2.9,2.1Hz,1H),5.97(tt,J=55.8,4.5Hz,1H),3.77(q,J=5.7,4.9Hz,4H),3.71(s,3H),3.57(td,J=13.6,4.5Hz,2H),1.96(p,J=5.6Hz,2H).
Preparation example 8 preparation of Compound S8
Figure BDA0002006087980000232
Synthesis of referential example S3 using 4-2 as a starting material and 2- (4-morpholine) ethyl bromide instead of (2-bromoethyl) trimethyl ammonium bromide gave compound S8.
1H NMR(400MHz,CDCl3)10.85(s,1H),7.56(d,J=2.7Hz,1H),7.29(t,J=2.9Hz,2H),7.19(s,1H),6.98–6.86(m,2H),6.82(d,J=8.7Hz,1H),6.77(td,J=10.6,9.9,2.2Hz,1H),6.44(dd,J=2.9,2.1Hz,1H),5.97(tt,J=55.8,4.5Hz,1H),3.77(q,J=5.7,4.9Hz,4H),3.71(s,3H),3.57(td,J=13.6,4.5Hz,2H),1.96(p,J=5.6Hz,2H).
Preparation example 9 preparation of compound S9
Figure BDA0002006087980000241
Synthesis of referential example S3 using 4-2 as a starting material and 3-bromomethyl-3-hydroxymethyl-1-oxetane instead of (2-bromoethyl) trimethylammonium bromide gave compound S9.
1H NMR(400MHz,CDCl3)10.99(s,1H),7.57(d,J=2.7Hz,1H),7.29(dt,J=5.8,3.1Hz,2H),7.20(s,1H),6.99–6.88(m,2H),6.84(d,J=8.7Hz,1H),6.79(td,J=9.1,2.8Hz,1H),6.44(t,J=2.5Hz,1H),4.54(d,J=6.5Hz,2H),4.46(d,J=6.5Hz,2H),4.02(d,J=6.0Hz,2H),3.81–3.73(m,4H),3.72(s,3H),3.65(s,2H),3.01(t,J=6.1Hz,1H),2.02(dt,J=11.0,5.9Hz,2H).
Preparation example 10 preparation of compound S10
Figure BDA0002006087980000251
Synthesis of Compound 10-1:
compound 4-2(1eq) was dissolved in DMF, and cesium carbonate (2eq) and 1, 2-dibromoethane (1.1eq) were added and reacted at 60 ℃ for 2 hours. Washing with water, extracting with ethyl acetate, washing organic phase with water for 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2MeOH is 40:1 to 30:1, to give intermediate 10-1.
Synthesis of Compound 10-2:
3-Methoxyazetidine hydrochloride (2eq) was dissolved in DMF, potassium carbonate (3eq) and intermediate 10-1(1eq) were added, and reaction was carried out at 70 ℃ for 2 h. Washing with water, extracting with ethyl acetate, washing organic phase with water for 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2MeOH is 40:1 to 30:1, to obtain intermediate 10-2.
Synthesis of compound S10:
dissolving the compound 10-2 in 1, 4-dioxane, adding 3M NaOH solution, and reacting at 90 ℃ for half an hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S10.
1H NMR(400MHz,CDCl3)10.99(s,1H),7.57(d,J=2.7Hz,1H),7.29(dt,J=5.8,3.1Hz,2H),7.20(s,1H),6.99–6.88(m,2H),6.84(d,J=8.7Hz,1H),6.79(td,J=9.1,2.8Hz,1H),6.44(t,J=2.5Hz,1H),4.54(d,J=6.5Hz,2H),4.46(d,J=6.5Hz,2H),4.02(d,J=6.0Hz,2H),3.81–3.73(m,4H),3.72(s,3H),3.65(s,2H),3.01(t,J=6.1Hz,1H),2.95–2.80(m,1H),2.02(dt,J=11.0,5.9Hz,2H).
Preparation of Compound S11 of preparation example 11
Figure BDA0002006087980000261
Synthesis of compound S11:
3-hydroxymethyl pyridineAlkane (2eq) was dissolved in DMF, potassium carbonate (3eq) and intermediate 10-1(1eq) were added, and reaction was carried out at 70 ℃ for 2 h. Washing with water, extracting with ethyl acetate, washing organic phase with water for 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2MeOH is 40:1 to 30:1 to obtain the intermediate.
The intermediate is dissolved in 1, 4-dioxane, 3M NaOH solution is added, and the reaction is carried out for half an hour at 90 ℃. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S11.
1H NMR(400MHz,CD2Cl2)11.17(s,1H),7.60(d,J=2.7Hz,1H),7.35–7.29(m,2H),7.25(s,1H),7.05–6.93(m,2H),6.88(d,J=8.3Hz,1H),6.87–6.81(m,1H),6.46(dd,J=2.8,2.0Hz,1H),3.87–3.81(m,2H),3.81–3.73(m,4H),3.71(s,3H),3.69–3.57(m,2H),3.29(t,J=6.0Hz,2H),2.61(d,J=14.1Hz,1H),2.20–2.08(m,1H),2.04–1.95(m,2H),1.93–1.82(m,1H).
Preparation example 12 preparation of compound S12
Figure BDA0002006087980000262
Synthesis of compound S12 in reference example S11 using 10-1 as the starting material and N-methyl-2-hydroxyethylamine instead of 3-hydroxymethylpyrrolidine.
1H NMR(400MHz,CD2Cl2)7.60(d,J=2.7Hz,1H),7.37–7.31(m,2H),7.24(s,1H),7.05–6.93(m,2H),6.88(d,J=8.7Hz,1H),6.84(tdd,J=8.0,2.9,1.4Hz,1H),6.46(dd,J=2.8,1.9Hz,1H),3.78(dt,J=15.7,4.4Hz,6H),3.71(d,J=4.8Hz,5H),3.16(t,J=6.1Hz,2H),3.06–2.98(m,2H),2.70(s,3H),1.99(p,J=5.7Hz,2H).
Preparation example 13 preparation of compound S13
Figure BDA0002006087980000271
Synthesis of reference example S11, using 10-1 as the starting material and N-methylpiperazine instead of 3-hydroxymethylpyrrolidine, gave compound S13.
1H NMR(400MHz,CD2Cl2)7.60(d,J=2.7Hz,1H),7.36(t,J=2.8Hz,1H),7.31(dd,J=8.8,2.7Hz,1H),7.23(s,1H),7.04–6.93(m,2H),6.88(dd,J=8.9,0.8Hz,1H),6.86–6.81(m,1H),6.45(dd,J=2.8,2.1Hz,1H),3.75(t,J=5.7Hz,2H),3.71(s,5H),3.40(s,3H),2.97(d,J=40.1Hz,6H),2.74(d,J=23.0Hz,5H).
Preparation example 14 preparation of compound S14
Figure BDA0002006087980000272
Synthesis of Compound 14-1:
compound 1-2(1eq) was dissolved in acetonitrile, and 1, 4-dibromobutane (1.2eq) and potassium carbonate (2.2eq) were added, followed by heating under reflux and stirring overnight. Quenching with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2EA [ ethyl acetate ]]Obtaining the compound 14-1 by 10: 1.
Synthesis of Compound 14-2:
compound 14-1(1eq) was dissolved in dichloromethane, TFA (trifluoroacetic acid, 20eq) was added dropwise and reacted at room temperature for 2 h. And after the reaction is completed, carrying out reduced pressure rotary evaporation on the reaction liquid, dissolving the residue by using dichloromethane, washing the residue by using a saturated sodium bicarbonate solution, combining organic phases, washing the combined organic phases by using saturated salt, drying the combined organic phases by using anhydrous sodium sulfate, carrying out reduced pressure rotary evaporation to obtain a crude product 14-2, and directly putting the crude product into the next step.
Synthesis of Compound 14-3:
compound 14-2(1eq) was dissolved in DMF and added with potassium carbonate (1.5eq) and iodomethane (1.2eq) and reacted at room temperature for 1 h. After the reaction is completed, adding water to wash, extracting with ethyl acetate, washing the organic phase with water for 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, and carrying out reduced pressure rotary evaporation to obtain a crude product 14-3 which is directly put into the next step.
Synthesis of compound S14:
dissolving compound 14-3 in 1, 4-dioxane, adding 3M NaOH solution, and reacting at 90 deg.CShould be half an hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S14.
1H NMR(400MHz,DMSO-d6)12.08(s,1H),7.48–7.40(m,2H),7.35(s,1H),7.30(dt,J=5.2,2.9Hz,2H),7.23(td,J=9.2,5.6Hz,1H),7.11–7.04(m,1H),6.86(d,J=8.8Hz,1H),6.29(dd,J=2.8,2.0Hz,1H),3.69(t,J=5.7Hz,2H),3.56(s,5H),2.87(s,3H),1.97–1.87(m,2H),1.23(p,J=7Hz,2H).
Preparation example 15 preparation of compound S15
Figure BDA0002006087980000281
Synthesis of Compound 15-1:
dissolving the compound 1-1(1eq) in dichloromethane, adding triethylamine (3eq), dropwise adding 3-chloropropane sulfonyl chloride (2eq) while stirring at room temperature, and reacting at room temperature for 0.5 h. Quenching with water after the reaction is completed, extracting with dichloromethane for three times, combining organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with a sample, loading onto a column, and performing reaction on the mixture with CH2Cl2EA [ ethyl acetate ]]Obtaining the compound 15-1 by 10: 1.
Synthesis of compound S15:
the compound 15-1 is dissolved in 1, 4-dioxane, and 3M NaOH solution is added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S15.
1H NMR(400MHz,CDCl3)10.90(s,1H),7.48(d,J=2.8Hz,1H),7.31(d,J=2.6Hz,1H),7.26(dd,J=8.9,2.8Hz,1H),7.22(s,1H),6.96–6.83(m,3H),6.78–6.71(m,1H),6.48(t,J=2.1Hz,1H),3.80(t,J=6.6Hz,2H),3.73(s,3H),3.42(t,J=7.6Hz,2H),2.56(p,J=7.0Hz,2H).
Preparation of Compound S16 of preparation example 16
Figure BDA0002006087980000291
Synthesis of Compound 16-1:
compound 16-1 was obtained by substituting 4-chloro-1-butylsulfonyl chloride for 3-chloropropanesulfonyl chloride using compound 1-1 as a starting material, referring to the synthesis of example 15-1.
Synthesis of compound S16:
compound S16 was obtained by synthesizing compound 16-1 as the starting material in referential example S15.
1H NMR(400MHz,CDCl3)10.48(s,1H),7.55(d,J=2.7Hz,1H),7.30(dd,J=3.0,2.3Hz,1H),7.28–7.25(m,1H),7.22(s,1H),6.98–6.88(m,2H),6.85(dd,J=8.8,0.9Hz,1H),6.82–6.75(m,1H),6.50(dd,J=2.9,1.9Hz,1H),3.81–3.74(m,2H),3.72(s,3H),3.29–3.21(m,2H),2.36(dq,J=8.0,6.0Hz,2H),1.94(p,J=5.7Hz,2H).
Preparation of Compound S17 of preparation example 17
Figure BDA0002006087980000301
Synthesis of Compound 17-1:
compound 17-1 was obtained by substituting 5-chloro-1-pentylsulfonyl chloride for 3-chloropropanesulfonyl chloride using compound 1-1 as a starting material according to the synthesis in example 15-1.
Synthesis of compound S17:
compound S17 was obtained by synthesizing compound 17-1 as the starting material in referential example S15.
1H NMR(400MHz,CDCl3)10.48(s,1H),7.55(d,J=2.7Hz,1H),7.30(dd,J=3.0,2.3Hz,1H),7.28–7.25(m,1H),7.22(s,1H),6.98–6.88(m,2H),6.85(dd,J=8.8,0.9Hz,1H),6.82–6.75(m,1H),6.50(dd,J=2.9,1.9Hz,1H),3.81–3.74(m,2H),3.72(s,3H),3.29–3.21(m,2H),2.36(dq,J=8.0,6.0Hz,2H),1.94(p,J=5.7Hz,2H),1.23(p,J=7Hz,2H).
Preparation example 18 preparation of compound S18
Figure BDA0002006087980000302
Synthesis of Compound 18-1:
compound 1-1(1eq) was dissolved in dichloromethane, triethylamine (2eq) was added, methyl 2- (chlorosulfonyl) acetate (1.1eq) was added dropwise with stirring at room temperature, and the reaction was carried out at room temperature for 0.5 h. Quenching with water after the reaction is completed, extracting with dichloromethane for three times, combining organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with a sample, loading onto a column, and performing reaction on the mixture with CH2Cl2EA [ ethyl acetate ]]Obtaining the compound 18-1 by the ratio of 4: 1.
Synthesis of Compound 18-2:
compound 18-1(1eq) was dissolved in DMF, potassium carbonate (3eq) was added and stirred at 70 ℃ and 1, 2-dibromoethane (1.2eq) was dissolved in DMF and the reaction solution was added dropwise to the dropping funnel. Washing with water after reaction, extracting with ethyl acetate twice, combining organic phases, washing with water 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2EA [ ethyl acetate ]]Obtaining the compound 18-2 by the ratio of 4: 1.
Synthesis of compound S18:
adding a compound of CaCl2(2eq) and NaBH4(4eq) was dissolved in dry tetrahydrofuran and stirred at room temperature for 1 h. Dissolving the compound 18-2(1eq) in tetrahydrofuran, adding the reaction solution dropwise at 0 ℃, and reacting for 2 hours at room temperature after the addition. After the reaction is completed, quenching the mixture by water, extracting the mixture twice by ethyl acetate, combining organic phases, washing the combined organic phases by saturated salt water, drying the organic phases by anhydrous sodium sulfate, decompressing and carrying out rotary evaporation, and directly putting the obtained intermediate into the next step.
The intermediate is dissolved in 1, 4-dioxane, 3M NaOH solution is added, and the reaction is carried out for half an hour at 90 ℃. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S18.
1H NMR(400MHz,DMSO-d6)12.09(s,2H),7.45–7.32(m,6H),7.33–7.26(m,2H),7.24–7.16(m,2H),7.16–6.97(m,2H),7.02(s,2H),6.96(d,J=8.8Hz,2H),6.30(s,2H),5.29(ddd,J=6.0,4.5,1.1Hz,2H),3.89–3.66(m,5H),3.70(s,3H),3.63(q,J=7.4,6.8Hz,1H),3.54(d,J=1.2Hz,5H),2.06(s,1H).
Preparation of Compound S19 of preparation example 19
Figure BDA0002006087980000311
Synthesis of Compound 19-1:
dissolving the compound 18-1(1eq) in DMF, adding potassium carbonate (3eq) and stirring at 70 ℃, dissolving 1-bromo-3-chloropropane (1.2eq) in DMF and dropwise adding the reaction solution into a dropping funnel. Washing with water after reaction, extracting with ethyl acetate twice, combining organic phases, washing with water 5 times, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2EA [ ethyl acetate ]]Getting compound 19-1 with 4: 1.
Synthesis of Compound 19-2:
adding a compound of CaCl2(2eq) and NaBH4(4eq) was dissolved in dry tetrahydrofuran and stirred at room temperature for 1 h. The compound 19-1(1eq) was dissolved in tetrahydrofuran, and the reaction mixture was added dropwise at 0 ℃ and reacted at room temperature for 2 hours. Quenching the mixture with water after the reaction is completed, extracting the mixture twice with ethyl acetate, combining organic phases, washing the combined organic phases with saturated salt water, drying the organic phases with anhydrous sodium sulfate, decompressing and carrying out rotary evaporation to obtain a crude product 19-2, and directly feeding the crude product to the next step.
Synthesis of compound S19:
dissolving the compound 19-2 in 1, 4-dioxane, adding 3M NaOH solution, and reacting at 90 ℃ for half an hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S19.
1H NMR(400MHz,CD3CN)10.45(s,1H),7.50(d,J=2.7Hz,1H),7.33–7.27(m,3H),7.13–7.05(m,2H),6.96–6.89(m,2H),6.41(t,J=2.6Hz,1H),4.04(dd,J=11.8,5.6Hz,1H),3.87–3.73(m,2H),3.62(s,4H),3.34(td,J=10.3,6.0Hz,1H),2.38–2.30(m,1H),2.15–2.03(m,1H),1.95–1.86(m,2H).
Preparation example 20 preparation of compound S20
Figure BDA0002006087980000321
The synthesis of compound 20-1 is referred to the patented procedure (WO2017177955A 1).
Synthetic reference methods for compound 20-2 (j.med. chem.2017,60, 3828-3850.).
Synthesis of Compound 20-3:
dissolving compound 20-1(1eq), 20-2(1.2eq) and palladium tetratriphenylphosphine (0.1eq) in 1, 4-dioxane, adding 2M Na2CO3The solution is reacted for 1h at 90 ℃ by microwave. After the reaction is completed, the reaction product is filtered, the filtrate is decompressed and rotary evaporated, and then the sample is mixed and loaded on a column, and the PE: EA is 1:1, thus obtaining the compound 20-3.
Synthesis of Compounds 20-4:
the compound 20-4 was obtained by synthesizing the compound 15-1 of reference example using the compound 20-3 as a starting material.
Synthesis of compound S20:
dissolving the compound 20-4 in 1, 4-dioxane, adding 3M NaOH solution, and reacting at 90 ℃. And (3) after the reaction is completed, spin-drying the reaction liquid, adding water to dissolve the reaction liquid, extracting the reaction liquid for 2 times by using anhydrous ether, adding 1M HCl solution into the residual water phase under stirring to adjust the pH value to 3-4, separating out a large amount of solids, and performing suction filtration and drying to obtain a compound S20.
1H NMR(400MHz,CDCl3)14.49(s,1H),13.25(s,1H),7.42(d,J=2.8Hz,1H),7.36(dd,J=8.9,2.7Hz,1H),7.27(s,1H),7.20(d,J=2.1Hz,1H),7.01–6.86(m,3H),6.77(t,J=8.7Hz,1H),3.84(t,J=6.6Hz,2H),3.77(s,3H),3.44(t,J=7.6Hz,2H),2.59(p,J=7.0Hz,2H).
Preparation of Compound S21 of preparation example 21
Figure BDA0002006087980000331
Synthesis of compound S21:
compound S20(1eq) was dissolved in dichloromethane, HOBT (1-hydroxybenzotriazole, 1.2eq) and EDCI (1-ethyl- (3-dimethylaminopropyl) carbonyldiimine hydrochloride, 1.2eq) were added, and after stirring at room temperature for 10min triethylamine (1.5eq) and NH were added4Cl (1.5eq), cellStir warm overnight. Washing with water after reaction, extracting with dichloromethane twice, combining organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and purifying with CH2Cl2MeOH is 30:1 to 15:1, and then the compound S21 is obtained.
1H NMR(400MHz,CD3OD)7.43(d,J=2.8Hz,1H),7.35–7.28(m,2H),7.05–6.87(m,4H),6.78(t,J=8.9Hz,1H),3.82(t,J=6.5Hz,2H),3.64(s,3H),3.45(t,J=7.5Hz,2H),2.51(p,J=7.0Hz,2H).
Preparation of Compound S22 of preparation example 22
Figure BDA0002006087980000341
Synthesis of compound S22:
s20 is used as raw material, ethylamine is used to replace NH4Cl, Synthesis of reference example S21, Compound S22 was obtained.
1H NMR(400MHz,CDCl3)11.01(s,1H),7.53(d,J=2.8Hz,1H),7.26(s,1H),7.22(dd,J=8.9,2.8Hz,1H),7.01–6.84(m,5H),6.80–6.74(m,1H),3.81(t,J=6.6Hz,2H),3.71(s,3H),3.58–3.48(m,2H),3.43(t,J=7.6Hz,2H),2.57(p,J=7.0Hz,2H),1.27(t,J=7.2Hz,3H).
Preparation of Compound S23 of preparation example 23
Figure BDA0002006087980000342
Synthesis of compound S23:
s20 is used as raw material, 1- (3-aminopropyl) -4-methyl piperazine is used for replacing NH4Cl, Synthesis of reference example S21, Compound S23 was obtained.
1H NMR(400MHz,CDCl3)11.14(s,1H),8.10(s,1H),7.48(d,J=2.8Hz,1H),7.25–7.18(m,2H),7.02(s,1H),6.97–6.84(m,3H),6.77(dddd,J=9.1,7.7,3.0,1.6Hz,1H),3.81(t,J=6.6Hz,2H),3.67(s,3H),3.54(q,J=6.1Hz,2H),3.43(t,J=7.5Hz,2H),3.03–2.60(m,10H),2.54(q,J=7.1Hz,2H),2.34(s,3H),1.89(p,J=6.8,6.0Hz,2H).
Preparation of Compound S24 of preparation example 24
Figure BDA0002006087980000351
Synthesis of Compound 24-1:
using 20-3 as raw material, substituting 4-chloro-1-butylsulfonyl chloride for 3-chloropropane sulfonyl chloride, and obtaining a compound 24-1 by the synthesis in reference example 20-4.
Synthesis of compound S24:
synthesis of example S20 was repeated using 24-1 as a starting material to obtain Compound S24.
1H NMR(400MHz,CDCl3)14.47(s,1H),13.24(s,1H),7.53(d,J=2.7Hz,1H),7.33(dd,J=8.8,2.7Hz,1H),7.26(s,1H),7.21(d,J=2.2Hz,1H),6.99(td,J=9.0,5.4Hz,1H),6.92(ddd,J=10.8,8.3,3.0Hz,1H),6.87(dd,J=8.8,0.8Hz,1H),6.84–6.78(m,1H),3.78(s,5H),3.32–3.23(m,2H),2.43–2.32(m,2H),1.98(p,J=6.2Hz,2H).
Preparation of Compound S25 of preparation example 25
Figure BDA0002006087980000352
Synthesis of compound S25:
compound S25 was obtained by the synthesis of reference example S21 using S24 as a starting material.
1H NMR(400MHz,CDCl3)12.54(s,1H),8.30(s,1H),7.57(d,J=2.6Hz,1H),7.29(d,J=6.8Hz,2H),7.19(d,J=2.1Hz,1H),7.02–6.76(m,5H),3.75(t,J=5.5Hz,2H),3.69(s,3H),3.28(t,J=6.1Hz,2H),2.36(dt,J=12.4,5.5Hz,2H),1.96(dt,J=11.4,5.8Hz,2H).
Preparation of Compound S26 of preparation example 26
Figure BDA0002006087980000361
Synthesis of compound S26:
compound S26 was obtained by the synthesis of reference example S22 using S24 as a starting material.
1H NMR(400MHz,CDCl3)11.21(s,1H),7.56(d,J=2.7Hz,1H),7.28–7.25(m,2H),7.14(t,J=5.7Hz,1H),7.01(d,J=2.2Hz,1H),6.98–6.89(m,2H),6.85(d,J=8.8Hz,1H),6.83–6.76(m,1H),3.83–3.70(m,5H),3.53(p,J=7.3Hz,2H),3.31–3.22(m,2H),2.36(p,J=6.2Hz,2H),1.96(p,J=5.8Hz,2H),1.27(t,J=7.2Hz,3H).
Preparation of Compound S27 of preparation example 27
Figure BDA0002006087980000362
Synthesis of compound S27:
compound S27 was obtained by the synthesis of reference example S23 using S24 as a starting material.
1H NMR(400MHz,CDCl3)11.14(s,1H),8.10(s,1H),7.48(d,J=2.8Hz,1H),7.25–7.18(m,2H),7.02(s,1H),6.97–6.84(m,3H),6.77(dddd,J=9.1,7.7,3.0,1.6Hz,1H),3.81(t,J=6.6Hz,2H),3.67(s,3H),3.54(q,J=6.1Hz,2H),3.43(t,J=7.5Hz,2H),3.03–2.60(m,10H),2.54(q,J=7.1Hz,2H),2.34(s,3H),1.89(p,J=6.8,6.0Hz,2H),1.23(p,J=7Hz,2H).
Preparation of Compound S28 of preparation example 28
Figure BDA0002006087980000371
S20 is used as raw material, and ethanol is used for replacing NH4Cl, Synthesis of reference example S21, Compound S28 was obtained.
1H NMR(400MHz,CDCl3)10.38(s,1H),7.39(d,J=2.8Hz,1H),7.24(d,J=2.8Hz,2H),7.18(s,1H),7.08(d,J=2.2Hz,1H),6.93–6.83(m,3H),6.79–6.71(m,1H),4.39(q,J=7.1Hz,2H),3.78(t,J=6.5Hz,2H),3.71(s,3H),3.40(t,J=7.6Hz,2H),2.55(p,J=6.9Hz,2H),1.39(t,J=7.1Hz,3H).
Preparation of Compound S29 of preparation example 29
Figure BDA0002006087980000372
Compound S29 was obtained by the synthesis of reference example S28 using S24 as a starting material.
1H NMR(400MHz,CDCl3)10.36(s,1H),7.45(d,J=2.6Hz,1H),7.26(s,2H),7.15(s,1H),7.08(d,J=2.2Hz,1H),6.97–6.85(m,2H),6.83–6.74(m,2H),4.37(q,J=7.1Hz,2H),3.73(t,J=5.5Hz,2H),3.69(s,3H),3.26–3.19(m,2H),2.33(p,J=6.4Hz,2H),1.92(p,J=5.7Hz,2H),1.38(t,J=7.1Hz,3H).
Preparation of Compound S30 of preparation example 30
Figure BDA0002006087980000381
Synthesis of Compound 30-2:
dissolving the compound 30-1(1eq) in dichloromethane, adding 1-fluoro-2, 6-dichloropyridine tetrafluoroborate (1.5eq), stirring at room temperature for 10h, adding water for quenching, extracting with dichloromethane twice, combining organic phases, washing with saturated common salt water, drying with anhydrous sodium sulfate, mixing with a sample, loading onto a column, and obtaining the compound 30-2 by using PE (polyethylene) EA (1: 1).
Synthesis of Compound 30-3:
dissolving compound 30-2(1eq) in DMF, precooling at 0 ℃ and N2NaH (1.5eq) was added under protection, p-toluenesulfonyl chloride (1.5eq) was added after stirring for 10min, and the reaction solution was allowed to cool to room temperature for 1 h. After the reaction is completed, water is added for quenching under stirring, and solid is separated out, filtered, and the filter cake is dried to obtain a crude product which is directly put into the next step.
The crude product (1eq) was dissolved in tetrahydrofuran, p-toluenesulfonic acid (0.5eq) and NBS (N-bromosuccinimide, 1eq) were added with stirring, and stirred at room temperature for 1 h. And after the reaction is completed, carrying out rotary drying on the reaction liquid under reduced pressure, mixing the sample and loading the mixture on a column, and obtaining the compound 30-3 by taking PE (polyethylene) and EA as 4: 1.
Synthesis of Compounds 30-4:
compound 30-4 was obtained by synthesizing compound 20-3 of reference example using compound 30-3 as a starting material.
Synthesis of Compounds 30-5:
dissolving the compound 30-4(1eq) in dichloromethane, adding triethylamine (3eq), stirring at room temperatureDropwise adding 3-chloropropane sulfonyl chloride (2.2eq) and reacting for 0.5h at room temperature. Quenching with water after the reaction is completed, extracting with dichloromethane for three times, combining organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with a sample, loading onto a column, and performing reaction on the mixture with CH2Cl2EA [ ethyl acetate ]]Getting compound 30-5 with 10: 1.
Synthesis of compound S30:
dissolving the compound 30-5 in 1, 4-dioxane, adding 3M NaOH solution, and reacting at 90 ℃. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S30.
1H NMR(500MHz,CDCl3)10.75(s,1H),7.37(t,J=2.4Hz,1H),7.29(d,J=8.3Hz,1H),7.10–7.04(m,2H),6.98(td,J=9.0,5.4Hz,1H),6.87(dd,J=10.4,8.3Hz,2H),6.80–6.75(m,1H),3.80(t,J=6.6Hz,2H),3.70(s,3H),3.41(t,J=7.6Hz,2H),2.55(p,J=7.0Hz,2H).
Preparation of Compound S31 of preparation example 31
Figure BDA0002006087980000391
Synthesis of Compound 31-1:
the compound 30-4(1eq) was dissolved in dichloromethane, triethylamine (3eq) was added, 4-chlorobutylsulfonyl chloride (2.2eq) was added dropwise with stirring at room temperature, and the reaction was carried out at room temperature for 0.5 h. Quenching with water after the reaction is completed, extracting with dichloromethane for three times, combining organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with a sample, loading onto a column, and performing reaction on the mixture with CH2Cl2EA is 10:1, and the compound 31-1 is obtained.
Synthesis of compound S31:
the compound 31-1 was dissolved in 1, 4-dioxane, and 3M NaOH solution was added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S31.
1H NMR(500MHz,CDCl3)11.13(s,1H),7.45(t,J=2.4Hz,1H),7.30–7.28(m,1H),7.09(t,J=3.1Hz,1H),7.08–7.00(m,2H),6.89(ddd,J=10.8,8.3,3.0Hz,1H),6.83–6.76(m,2H),3.80–3.74(m,2H),3.70(s,3H),3.27–3.20(m,2H),2.35(p,J=6.1Hz,2H),1.95–1.92(m,2H).
Preparation of Compound S32 of preparation example 32
Figure BDA0002006087980000401
Synthetic reference methods for compound 32-1 (j.med. chem.2017,60, 8369-8384).
Synthesis of Compound 32-2:
compound 32-1(1eq) was dissolved in dimethyl sulfoxide, trans 4-methylcyclohexylamine (3eq) was added, and the mixture was heated to 85 ℃ to react for 5 hours. After the reaction is completed, water is used for washing, ethyl acetate is used for extracting twice, the combined organic phase is washed by water for 3 times, then is washed by saturated salt solution, is dried by anhydrous sodium sulfate and is mixed with a sample and loaded on a column, and the PE: EA is 4:1, so that the compound 32-2 is obtained.
Synthesis of Compound 32-3:
mixing the compound 32-2(1eq), iron powder (2eq) and NH4Cl (5eq) dissolved in EtOH THF H2Heating the mixture to 80 ℃ for reaction for 3 hours in a mixed solvent of O-5: 1: 1. After the reaction is completed, the filtration is carried out, the filtrate is decompressed and dried in a spinning mode, and a crude product 32-3 is obtained and is directly put into the next step.
Synthesis of Compound 32-4:
dissolving the compound 32-3(1eq) in dichloromethane, adding triethylamine (2eq), dropwise adding 3-chloropropylsulfonyl chloride (1.5eq) at room temperature under stirring, and reacting at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 32-4 with PE: EA being 3: 1.
Synthesis of compound S32:
the compound 32-4 was dissolved in 1, 4-dioxane, and 3M NaOH solution was added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S32.
1H NMR(400MHz,CDCl3)11.61(s,1H),7.32(t,J=2.7Hz,1H),7.26(dd,J=8.7,2.7Hz,1H),7.15(d,J=2.6Hz,1H),6.99(s,1H),6.71(d,J=8.8Hz,1H),6.22(dd,J=2.8,2.1Hz,1H),3.77–3.69(m,5H),3.40–3.32(m,2H),3.25–3.15(m,1H),2.54–2.43(m,2H),2.02(m,2H),1.75–1.66(m,2H),1.30(s,1H),1.02(dt,J=27.7,13.2Hz,4H),0.90(d,J=6.5Hz,3H).
Preparation of Compound S33 of preparation example 33
Figure BDA0002006087980000411
Synthesis of Compound 33-1:
the compound 32-3(1eq) was dissolved in dichloromethane, triethylamine (2eq) was added, 4-chlorobutylsulfonyl chloride (1.5eq) was added dropwise with stirring at room temperature, and the reaction was carried out at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 33-1 with PE: EA being 3: 1.
Synthesis of compound S33:
the compound 33-1 was dissolved in 1, 4-dioxane, and 3M NaOH solution was added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S33.
1H NMR(400MHz,CDCl3)11.10(s,1H),7.30(t,J=2.8Hz,1H),7.24(dd,J=8.7,2.6Hz,1H),7.14(d,J=2.6Hz,1H),6.99(s,1H),6.67(d,J=8.8Hz,1H),6.24(t,J=2.4Hz,1H),3.73(s,4H),3.70–3.65(m,2H),3.27–3.15(m,3H),2.32(p,J=6.3Hz,2H),2.02(d,J=11.2Hz,2H),1.90(d,J=6.3Hz,2H),1.71(d,J=12.0Hz,2H),1.31(dd,J=6.8,3.6Hz,1H),1.02(dt,J=26.4,12.9Hz,4H),0.90(d,J=6.5Hz,3H).
Preparation of Compound S34 of preparation example 34
Figure BDA0002006087980000421
Synthesis of Compound 34-1:
compound 32-1(1eq) was dissolved in dimethyl sulfoxide, 4-difluorocyclohexylamine (3eq) was added, and the mixture was heated to 85 ℃ to react for 5 hours. After the reaction is completed, water is used for washing, ethyl acetate is used for extracting twice, the combined organic phase is washed by water for 3 times, then is washed by saturated salt solution, is dried by anhydrous sodium sulfate and is mixed with a sample and loaded on a column, and the compound 34-1 is obtained by PE: EA is 4: 1.
Synthesis of Compound 34-2:
mixing compound 34-1(1eq), iron powder (2eq), NH4Cl (5eq) dissolved in EtOH THF H2Heating the mixture to 80 ℃ for reaction for 3 hours in a mixed solvent of O-5: 1: 1. After the reaction is completed, the filtration is carried out, the filtrate is decompressed and dried in a spinning mode, and a crude product 34-2 is obtained and is directly put into the next step.
Synthesis of Compound 34-3:
the compound 34-2(1eq) was dissolved in dichloromethane, triethylamine (2eq) was added, 3-chloropropylsulfonyl chloride (1.5eq) was added dropwise with stirring at room temperature, and the reaction was carried out at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 34-3 with PE: EA being 3: 1.
Synthesis of compound S34:
the compound 34-3 was dissolved in 1, 4-dioxane, and 3M NaOH solution was added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S34.
1H NMR(400MHz,CDCl3)11.61(s,1H),7.32(t,J=2.7Hz,1H),7.26(dd,J=8.7,2.7Hz,1H),7.15(d,J=2.6Hz,1H),6.99(s,1H),6.71(d,J=8.8Hz,1H),6.22(dd,J=2.8,2.1Hz,1H),3.77–3.69(m,5H),3.40–3.32(m,2H),3.25–3.15(m,1H),2.54–2.43(m,2H),2.02(m,2H),1.75–1.66(m,2H),1.46(m,4H).
Preparation of Compound S35 of preparation example 35
Figure BDA0002006087980000431
Synthesis of Compound 35-1:
compound 34-2(1eq) was dissolved in dichloromethane, triethylamine (2eq) was added, 4-chlorobutylsulfonyl chloride (1.5eq) was added dropwise with stirring at room temperature, and the reaction was carried out at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 35-1 with PE: EA being 3: 1.
Synthesis of compound S35:
the compound 35-1 was dissolved in 1, 4-dioxane, and 3M NaOH solution was added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S35.
1H NMR(400MHz,CDCl3)11.10(s,1H),7.30(t,J=2.8Hz,1H),7.24(dd,J=8.7,2.6Hz,1H),7.14(d,J=2.6Hz,1H),6.99(s,1H),6.67(d,J=8.8Hz,1H),6.24(t,J=2.4Hz,1H),3.73(s,4H),3.70–3.65(m,2H),3.27–3.15(m,3H),2.32(p,J=6.3Hz,2H),2.02(d,J=11.2Hz,2H),1.90(d,J=6.3Hz,2H),1.71(d,J=12.0Hz,2H),1.46(m,4H).
Preparation of Compound S36 of preparation example 36
Figure BDA0002006087980000441
Synthesis of Compound 36-1:
the compound 32-1(1eq) was dissolved in dimethyl sulfoxide, and (2, 2-difluorocyclopropyl) methylamine (3eq) was added thereto, followed by heating to 85 ℃ for 5 hours. After the reaction is completed, water is used for washing, ethyl acetate is used for extracting twice, the combined organic phase is washed by water for 3 times, then is washed by saturated salt solution, is dried by anhydrous sodium sulfate and is mixed with a sample and loaded on a column, and the PE: EA is 4:1, so that the compound 36-1 is obtained.
Synthesis of Compound 36-2:
mixing compound 36-1(1eq), iron powder (2eq), NH4Cl (5eq) solutionIn EtOH THF H2Heating the mixture to 80 ℃ for reaction for 3 hours in a mixed solvent of O-5: 1: 1. After the reaction is completed, the filtration is carried out, the filtrate is decompressed and dried in a spinning mode, and a crude product 36-2 is obtained and is directly put into the next step.
Synthesis of Compound 36-3:
dissolving the compound 36-2(1eq) in dichloromethane, adding triethylamine (2eq), dropwise adding 3-chloropropylsulfonyl chloride (1.5eq) at room temperature under stirring, and reacting at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 36-3 with PE: EA being 3: 1.
Synthesis of compound S36:
the compound 36-3 is dissolved in 1, 4-dioxane, and 3M NaOH solution is added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S36.
1H NMR(400MHz,CDCl3)11.61(s,1H),7.32(t,J=2.7Hz,1H),7.26(dd,J=8.7,2.7Hz,1H),7.15(d,J=2.6Hz,1H),6.99(s,1H),6.71(d,J=8.8Hz,1H),6.22(dd,J=2.8,2.1Hz,1H),3.73(s,3H)3.69–3.59(m,2H),3.40–3.32(m,2H),3.11–2.86(m,2H),2.54–2.43(m,2H),1.62–1.55(m,1H),0.95–0.88(m,1H),0.75–0.70(m,1H).
Preparation of Compound S37 of preparation example 37
Figure BDA0002006087980000451
Synthesis of Compound 37-1:
dissolving the compound 36-2(1eq) in dichloromethane, adding triethylamine (2eq), dropwise adding 4-chlorobutylsulfonyl chloride (1.5eq) while stirring at room temperature, and reacting at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 37-1 with PE: EA being 3: 1.
Synthesis of compound S37:
the compound 37-1 was dissolved in 1, 4-dioxane, and 3M NaOH solution was added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S37.
1H NMR(400MHz,CDCl3)11.10(s,1H),7.30(t,J=2.8Hz,1H),7.24(dd,J=8.7,2.6Hz,1H),7.14(d,J=2.6Hz,1H),6.99(s,1H),6.67(d,J=8.8Hz,1H),6.24(t,J=2.4Hz,1H),3.73(s,3H),3.70–3.65(m,2H),3.27–3.15(m,2H),3.11–2.86(m,2H),2.32(p,J=6.3Hz,2H),2.02(d,J=11.2Hz,2H),1.62–1.55(m,1H),0.95–0.88(m,1H),0.75–0.70(m,1H).
Preparation of Compound S38 of preparation example 38
Figure BDA0002006087980000461
Synthesis of Compound 38-1:
the compound 32-1(1eq) was dissolved in tetrahydrofuran, trans-4-methylcyclohexylamine (3eq) was added, and the mixture was heated to 85 ℃ to react for 5 hours. After the reaction is completed, water is used for washing, ethyl acetate is used for extracting twice, the combined organic phase is washed by water for 3 times, then is washed by saturated salt solution, is dried by anhydrous sodium sulfate and is mixed with a sample and loaded on a column, and the PE: EA is 4:1, so that the compound 38-1 is obtained.
Synthesis of Compound 38-2:
mixing compound 38-1(1eq), iron powder (2eq), NH4Cl (5eq) dissolved in EtOH THF H2Heating the mixture to 80 ℃ for reaction for 3 hours in a mixed solvent of O-5: 1: 1. After the reaction is completed, the filtration is carried out, the filtrate is decompressed and dried in a spinning mode, and a crude product 38-2 is obtained and is directly put into the next step.
Synthesis of Compound 38-3:
dissolving the compound 38-2(1eq) in dichloromethane, adding triethylamine (2eq), dropwise adding 3-chloropropylsulfonyl chloride (1.5eq) at room temperature under stirring, and reacting at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 38-3 with PE: EA being 3: 1.
Synthesis of compound S38:
the compound 38-3 was dissolved in 1, 4-dioxane, and 3M NaOH solution was added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S38.
1H NMR(400MHz,CDCl3)10.90(s,1H),7.48(d,J=2.8Hz,1H),7.31(d,J=2.6Hz,1H),7.26(dd,J=8.9,2.8Hz,1H),7.22(s,1H),6.96–6.83(m,3H),6.78–6.71(m,1H),6.48(t,J=2.1Hz,1H),3.80(t,J=6.6Hz,2H),3.73(s,3H),3.68–3.64(m,1H),3.40–3.32(m,2H),2.54–2.43(m,2H),2.02(m,2H),1.75–1.66(m,2H),1.33–1.29(m,1H),1.02(dt,J=27.7,13.2Hz,4H),0.90(d,J=6.5Hz,3H).
Preparation of Compound S39 of preparation example 39
Figure BDA0002006087980000471
Synthesis of Compound 39-1:
the compound 38-2(1eq) was dissolved in dichloromethane, triethylamine (2eq) was added, 4-chlorobutylsulfonyl chloride (1.5eq) was added dropwise with stirring at room temperature, and the reaction was carried out at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 39-1 with PE: EA being 3: 1.
Synthesis of compound S39:
the compound 39-1 was dissolved in 1, 4-dioxane, and 3M NaOH solution was added to react at 90 ℃ for 1 hour. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S39.
1H NMR(400MHz,CDCl3)10.48(s,1H),7.55(d,J=2.7Hz,1H),7.30(dd,J=3.0,2.3Hz,1H),7.28–7.25(m,1H),7.22(s,1H),6.98–6.88(m,2H),6.85(dd,J=8.8,0.9Hz,1H),6.82–6.75(m,1H),6.50(dd,J=2.9,1.9Hz,1H),3.81–3.74(m,2H),3.72(s,3H),3.70–3.65(m,1H),3.27–3.15(m,2H),2.32(p,J=6.3Hz,2H),1.98(p,J=5.7Hz,2H),1.90(d,J=6.3Hz,2H),1.71(d,J=12.0Hz,2H),1.31(dd,J=6.8,3.6Hz,1H),1.02(dt,J=26.4,12.9Hz,4H),0.90(d,J=6.5Hz,3H).
Preparation of Compound S40 of preparation example 40
Figure BDA0002006087980000481
Synthetic reference methods for compound 40-1 (j.med. chem.2017,60, 8369-8384.).
Synthesis of Compound 40-2:
dissolving compound 30-3(1eq), 40-1(1.2eq) and palladium tetratriphenylphosphine (0.1eq) in 1, 4-dioxane, adding 2M Na2CO3The solution is reacted for 1h at 90 ℃ by microwave. After the reaction is completed, the reaction product is filtered, the filtrate is decompressed and rotary evaporated, and then the sample is mixed and loaded on a column, and the compound 40-2 is obtained by PE: EA being 1: 1.
Synthesis of Compound 40-3:
the compound 40-2(1eq) was dissolved in dimethyl sulfoxide, trans 4-methylcyclohexylamine (3eq) was added, and the mixture was heated to 85 ℃ to react for 5 hours. After the reaction is completed, water is used for washing, ethyl acetate is used for extracting twice, the combined organic phase is washed by water for 3 times, then is washed by saturated salt solution, is dried by anhydrous sodium sulfate and is mixed with a sample and loaded on a column, and the PE: EA is 4:1, so that the compound 40-3 is obtained.
Synthesis of Compound 40-4:
mixing compound 40-3(1eq), iron powder (2eq), NH4Cl (5eq) dissolved in EtOH THF H2Heating the mixture to 80 ℃ for reaction for 3 hours in a mixed solvent of O-5: 1: 1. And after the reaction is completed, carrying out suction filtration, carrying out rotary drying on the filtrate under reduced pressure, extracting the obtained residue for three times by washing DCM, combining organic phases, washing the combined organic phases by saturated salt water, drying the combined organic phases by anhydrous sodium sulfate, carrying out rotary drying to obtain a compound 40-4, and directly putting the compound into the next step.
Synthesis of Compounds 40-5:
the compound 40-4(1eq) was dissolved in dichloromethane, triethylamine (2eq) was added, 3-chloropropylsulfonyl chloride (1.5eq) was added dropwise with stirring at room temperature, and the reaction was carried out at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 40-5 with PE: EA being 3: 1.
Synthesis of compound S40:
the compound 40-5 is dissolved in 1, 4-dioxane, and 3M NaOH solution is added to react for 1 hour at 90 ℃. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S40.
1H NMR(400MHz,CDCl3)11.61(s,1H),7.26(dd,J=8.7,2.7Hz,1H),7.15(d,J=2.6Hz,1H),7.05(t,J=3.1Hz,1H),6.99(s,1H),6.71(d,J=8.8Hz,1H),3.77–3.69(m,5H),3.40–3.32(m,2H),3.25–3.15(m,1H),2.54–2.43(m,2H),2.02(m,2H),1.75–1.66(m,2H),1.30(s,1H),1.02(dt,J=27.7,13.2Hz,4H),0.90(d,J=6.5Hz,3H).
Preparation of Compound S41 of preparation example 41
Figure BDA0002006087980000491
Synthesis of Compound 41-1:
the compound 40-4(1eq) was dissolved in dichloromethane, triethylamine (2eq) was added, 4-chlorobutylsulfonyl chloride (1.5eq) was added dropwise with stirring at room temperature, and the reaction was carried out at room temperature for 0.5 h. After the reaction is completed, quenching the mixture by water, extracting the mixture for three times by dichloromethane, combining organic phases, washing the organic phases by saturated salt solution, drying the organic phases by anhydrous sodium sulfate, mixing the dried organic phases with a sample, and loading the mixture on a column to obtain the compound 41-1 with PE: EA being 3: 1.
Synthesis of compound S41:
the compound 41-1 is dissolved in 1, 4-dioxane, and 3M NaOH solution is added to react for 1 hour at 90 ℃. Washing with water after reaction, extracting with ethyl acetate for three times, mixing organic phases, washing with saturated salt water, drying with anhydrous sodium sulfate, mixing with sample, loading onto column, and collecting CH2Cl2MeOH 60:1 to 30:1, to obtain compound S41.
1H NMR(400MHz,CDCl3)11.10(s,1H),7.24(dd,J=8.7,2.6Hz,1H),7.14(d,J=2.6Hz,1H),7.05(t,J=3.1Hz,1H),6.99(s,1H),6.67(d,J=8.8Hz,1H),3.73(s,4H),3.70–3.65(m,2H),3.27–3.15(m,3H),2.32(p,J=6.3Hz,2H),2.02(d,J=11.2Hz,2H),1.90(d,J=6.3Hz,2H),1.71(d,J=12.0Hz,2H),1.31(dd,J=6.8,3.6Hz,1H),1.02(dt,J=26.4,12.9Hz,4H),0.90(d,J=6.5Hz,3H).
Biological evaluation
The present invention is further described and explained below in conjunction with test examples, which are not intended to limit the scope of the present invention.
Experimental example 1: example compounds Activity assays for in vitro inhibition of the Hedgehog signaling pathway
Gli Luciferase Activity assay (Dual-Luciferase Reporter, Promega, E1960)
To verify the level of inhibition of the Hedgehog pathway by the compounds provided by the present invention, evaluation was performed by a Gli-luciferase (Gli-luciferase) reporter assay for Hedgehog signaling. The NIH3T3 cell strain transfected by a luciferase reporter gene (Gli-reporter) comprises a Gli-1 response fluorescent protein reporter gene, and the inhibition effect of a compound on a Hedgehog signal path is detected through the strength of fluorescent protein.
The specific experimental operation steps are as follows:
after logarithmic growth cell light II cells were inoculated to a 96-well plate and cultured for 24 hours, the culture medium was changed to a starvation medium (1% FBS, 0.4mg/ml G418, 0.15mg/ml Zeocin DMEM medium), Shh CM or SAG and the drug were added thereto, respectively, and the experiment was terminated after 36 hours. After aspiration of the culture medium, the cells were washed twice with pre-chilled 1 XPBS and then treated according to the protocol of the fluorescence double-Reporter kit (Promega Dual-Luciferase Reporter assay). The luminescence value of sea cucumber fluorescein (Renilla) and the luminescence value of firefly fluorescein (Luciferase) are detected by using a Thermo enzyme-labeling instrument, and the relative transcription activity of the reporter gene is expressed by adopting Luciferase/Renilla value.
And (4) analyzing results:
Figure BDA0002006087980000501
inhibition of Gli luciferase activity and IC50Evaluation experiment
Table 2: inhibitory Activity of some Compounds of the examples on Hedgehog
Figure BDA0002006087980000502
Figure BDA0002006087980000511
As can be seen from table 2 above: the compounds have excellent Hedgehog inhibitory activity, and the IC of 8 compounds50Below 1nM, the compounds S4, S16, S22, S32, S33 all had greater activity than the positive control compound ABBV-075.
Experimental example 2: inhibition of acute monocytic leukemia cell MV-4-11 proliferation activity by partial compound
The growth inhibition of MV-4-11 cells was measured by the CCK-8 method. The method comprises the following specific steps: cells in logarithmic growth phase are inoculated to a 96-well culture plate according to proper density, each well is 90 mu L, after overnight culture, drugs with different concentrations are added for 72h, each concentration is provided with three or more wells, and a solvent control with corresponding concentration and a cell-free zeroing well are arranged. After the reaction, 10 mu LCCK-8 is added into each hole, and after incubation for 4h in an incubator, the optical density (OD value) at the wavelength of 450nm is measured by a SpectraMax 190 microplate reader.
The inhibition (%) of the tumor cell growth by the compound was calculated using the following formula:
inhibition (%) - (OD control well-OD administration well)/OD control well X100%
Table 3: inhibition of MV-4-11 cell proliferation by compounds
Figure BDA0002006087980000512
As can be seen from table 3 above: the compound has a certain proliferation inhibition effect on MV-4-11 cells, wherein the inhibition rate of 15 compounds on the MV-4-11 cells under the concentration of 1000nM is more than 70%, the proliferation inhibition effect of the compounds S1, S4 and S16 on the MV-4-11 cells is strong, and IC is IC5049nM and 56 respectivelynM、21.8nM。
Experimental example 3: inhibition of human myelodysplastic syndrome cell SKM-1 proliferation activity by partial compound
The growth inhibition of SKM-1 cells is detected by a CCK-8 method. The method comprises the following specific steps: cells in logarithmic growth phase are inoculated to a 96-well culture plate according to proper density, each well is 90 mu L, after overnight culture, drugs with different concentrations are added for 72h, each concentration is provided with three or more wells, and a solvent control with corresponding concentration and a cell-free zeroing well are arranged. After the reaction, 10 mu LCCK-8 is added into each hole, and after incubation for 4h in an incubator, the optical density (OD value) at the wavelength of 450nm is measured by a SpectraMax 190 microplate reader.
The inhibition (%) of the tumor cell growth by the compound was calculated using the following formula:
inhibition (%) - (OD control well-OD administration well)/OD control well X100%
Table 4: inhibition of SKM-1 cell proliferation by compounds
Figure BDA0002006087980000521
As can be seen from table 4 above: the compound has better proliferation inhibition effect on SKM-1 cells, wherein the inhibition rate of 22 compounds on SKM-1 cells under the concentration of 100nM is more than 70%, and the inhibition activity of the compounds S32 and S33 on SKM-1 cells is better than that of positive control ABBV-075 and IC 33507.1nM and 6.3nM, respectively.
Experimental example 4: in vivo efficacy test of Compound mice
Growth inhibition of compound on subcutaneous transplantation tumor of medulloblastoma mouse
(1) Dose setting
The S15 and S16 doses were 12.5mg/kg, and the S1 doses were 25 mg/kg. The dosage of the positive contrast medicament ABBV-075 is 12.5 mg/kg.
The structural formula of ABBV-075 is as follows:
Figure BDA0002006087980000531
(2) animal(s) production
Nude mice, female, 6-7 weeks old, weight 25 + -1 g, purchased from Shanghai Spiral-BikeKa laboratory animals Co., Ltd, quality certification number: no. 2008001685287. Producing license numbers: SCXK (Shanghai) 2013-0016. Use license number for laboratory animals at the university of fudan: SYXK (Shanghai) 2015-0023. Number of animals per group: each group had 5.
(3) Tumor mass
Spontaneous medulloblastoma was isolated from ptch +/-p 53-/-mice, inoculated subcutaneously into nude mice, and used after 1 passage in nudemice mice.
(4) Experimental methods
Using Ptch +/-; p53-/-knockout mice establish medulloblastoma models according to reported methods. Ptch +/-genotype mice were crossed with P53-/-genotype mice. Obtaining the genotype Ptch +/-; p53-/-mice, which spontaneously develop medulloblastoma in their cerebellum. Detecting the genotype of the filial generation, Ptch +/-; p53-/-genotype mice spontaneously develop medulloblastoma in their cranium. Taking in-situ medulloblastoma, transplanting allogenic under aseptic condition to the right side armpit subcutaneous of a female nude mouse (Shanghai Xipul-Bikai laboratory animals Co., Ltd.) with age of 6-7 weeks, taking out a tumor block when the transplanted tumor grows to a proper size, shearing the tumor block into about 1.5mm under aseptic condition, and inoculating the tumor block under the left side armpit of the nude mouse by subcutaneous puncture. When it grows to 150mm3The drug is administered in groups at random. The S15 and S16 doses are 12.5mg/kg, the S1 dose is 25mg/kg, and the administration is carried out twice a day and continuously for 21 days by adopting two administration groups of oral administration (ig) and intraperitoneal injection (ip). The dose of the positive control drug ABBV-075 is 12.5mg/kg, and the positive control drug ABBV-075 is orally taken twice a day and continuously taken for 21 days. Tumor volume was measured once three days. Relative Tumor Volume (RTV) calculation: RTV ═ Vt/V0Where Vt represents the volume of the tumor measured per day, V0Denotes tumor body measured on the first day, V ═ a × b2) And/2, wherein a represents the measured length of the tumor mass and b represents the measured width of the tumor mass. Calculating the tumor growth inhibition rate: TGI ═ 1- (Vt)Administration set-V0 administration group)/(VtControl group-V0 control group)
Table 5: in vivo efficacy data of compound S1/S15/S16
Figure BDA0002006087980000541
As can be seen from table 5 above: compound S125 mg/kg/group, administered orally or intraperitoneally twice daily for 21 consecutive days for Ptch +/-; the growth of subcutaneous transplantable tumor of P53-/-medulloblastoma mouse had very significant inhibitory effect, with TGI percentages obtained at day 21 of 70.28% and 81.02%, respectively.
Compound s1512.5mg/kg/group, administered orally or intraperitoneally twice daily for 21 consecutive days for Ptch +/-; the growth of subcutaneous transplanted tumors of P53-/-medulloblastoma mice was extremely significantly inhibited, with the TGI percentages obtained at day 21 being 77.27% and 84.82%, respectively.
Compound s1612.5mg/kg/group, administered orally or intraperitoneally twice daily for 21 consecutive days for Ptch +/-; the growth of subcutaneous transplanted tumors of P53-/-medulloblastoma mice was extremely significantly inhibited, with the TGI percentages obtained at day 21 being 76.33% and 81.64%, respectively.
Compound s1512.5mg/kg oral administration vs Ptch +/-; the inhibition effect of P53-/-medulloblastoma is slightly weaker than that of the positive compound ABBV-07512.5 mg/kg oral administration, and the positive compound ABBV-07512.5 mg/kg oral administration is stronger in S1512.5mg/kg intraperitoneal injection. However, the positive compound caused weight loss in mice after 21 days of oral administration, while compound S15 did not cause weight loss in mice both orally and intraperitoneally, indicating that compound S15 is safer than ABBV-075.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (10)

1. A cyclic sulfamide substituted pyridone pyrrole compound shown in formula I, or an isomer, a pharmaceutically acceptable salt, a pharmaceutically acceptable solvate or hydrate thereof,
Figure FDA0002006087970000011
wherein:
n is an integer selected from the group consisting of: 0.1, 2 and 3;
x is selected from the group consisting of: CH. N;
l is selected from the group consisting of: o, NR5、C(O)、-(CH2)n-、-S(O)m-、-O(CR6R7)n-、-NR5(CR7)n-;
R1、R2、R3、R4Each independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6 alkyl, halogenated C1-C6 alkyl, deuterated C1-C6 alkyl, C2-C6 alkenyl, C1-C6 alkoxy, halogenated C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, halogen, amino, nitro, hydroxy, cyano, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, -OR8、-C(O)R8、-C(O)OR8、-S(O)mR8、-NR9R10、-C(O)NR9R10、-NR9C(O)R10、-NR9S(O)mR10(ii) a Wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogenated C1-C6 alkyl, halogen, amino, nitro, cyano, hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, halogenated C3-C8 cycloalkyl, heterocyclyl, - (C1-C6 alkylene) hydroxy-substituted heterocyclyl, (C1-C6 alkyl) -substituted heterocyclyl, C6-C10 aryl, heteroaryl, -OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R5Selected from the group consisting of: hydrogenC1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
R6and R7The same or different and each is independently selected from the group consisting of: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, halogen, amino, nitro, hydroxy, cyano, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, -OR8、-C(O)R8、-C(O)OR8、-S(O)mR8、-NR9R10、-C(O)NR9R10、-NR9C(O)R10、-NR9S(O)mR10
Or, R6And R7And C adjacent thereto may form a substituted or unsubstituted 3-8 membered cycloalkyl or heterocyclyl group, wherein the substitution means being substituted with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, halogen, amino, nitro, hydroxy, cyano, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R8Selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, C2-C6 alkenyl, hydroxyl, amino, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclic, C6-C10 aryl, heteroaryl; wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, amino, nitro, cyano, hydroxy, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R9And R10The same or different, and each is independently selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, hydroxy, amino, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12Wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, hydroxy, amino, nitro, cyano, C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, heterocyclyl, (C1-C6 alkyl) -substituted heterocyclyl, C6-C10 aryl, heteroaryl;
R11and R12The same or different, and each is independently selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, hydroxy, amino, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, hydroxy, amino, nitro, cyano, C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
m is an integer selected from the group consisting of: 0.1, 2;
wherein said "heterocyclyl" is a 4-8 membered heterocyclyl containing 1,2 or 3 heteroatoms selected from N, O, S;
the "heteroaryl" is a 4-10 membered heteroaryl containing 1,2 or 3 heteroatoms selected from N, O, S.
2. The compound of claim 1, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof,
n is an integer selected from the group consisting of: 0.1, 2 and 3;
x is selected from the group consisting of: CH. N;
l is selected from the group consisting of: o, NR5
R1、R2、R3、R4Each independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6 alkyl, halogenated C1-C6 alkyl, deuterated C1-C6 alkyl, C1-C6 alkoxy, halogenated C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, halogen, amino, nitro, hydroxy, cyano, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, -OR8、-C(O)R8、-C(O)OR8、-S(O)mR8、-NR9R10、-C(O)NR9R10、-NR9C(O)R10、-NR9S(O)mR10(ii) a Wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogenated C1-C6 alkyl, halogen, amino, nitro, cyano, hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, halogenated C3-C8 cycloalkyl, heterocyclyl, - (C1-C6 alkylene) hydroxy-substituted heterocyclyl, (C1-C6 alkyl) -substituted heterocyclyl, C6-C10 aryl, heteroaryl, -OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R5Selected from the group consisting of: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
R8selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, C2-C6 alkenyl, hydroxyl, amino, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclic, C6-C10 aryl, heteroaryl; wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, amino, nitro, cyano, hydroxy, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12
R9And R10The same or different, and each is independently selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, hydroxy, amino, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, OR11、-C(O)R11、-C(O)OR11、-S(O)mR11、-NR11R12、-C(O)NR11R12、-NR11C(O)R12、-NR11S(O)mR12Wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, hydroxy, amino, nitro, cyano, C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, heterocyclyl, (C1-C6 alkyl) -substituted heterocyclyl, C6-C10 aryl, heteroaryl;
R11and R12The same or different, and each is independently selected from the group consisting of substituted or unsubstituted: hydrogen, C1-C6 alkyl, hydroxy, amino, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl, wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogen, hydroxy, amino, nitro, cyano, C1-C6 alkoxy, - (C1-C6 alkylene) hydroxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
m is an integer selected from the group consisting of: 0.1, 2;
wherein said "heterocyclyl" is a 4-8 membered heterocyclyl containing 1,2 or 3 heteroatoms selected from N, O, S;
the "heteroaryl" is a 4-10 membered heteroaryl containing 1,2 or 3 heteroatoms selected from N, O, S.
3. The compound of claim 1, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof,
l is O.
4. The compound of claim 1, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof,
l is NR5
R5Selected from the group consisting of: hydrogen, C1-C6 alkyl, halogenated C1-C6 alkyl, - (C1-C6 alkylene) hydroxy, C1-C6 alkoxy, halogenated C1-C6 alkoxy, C3-C8 cycloalkyl, heterocyclyl, C6-C10 aryl, heteroaryl;
R4selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6 alkyl, halogenated C1-C6 alkyl, deuterated C1-C6 alkyl, C3-C8 cycloalkyl, heterocyclic, C6-C10 aryl, heteroaryl; wherein said substitution means substitution with one or more substituents selected from the group consisting of: C1-C6 alkyl, halogenated C1-C6 alkyl, halogen, C3-C8 cycloalkyl, and halogenated C3-C8 cycloalkyl.
5. The compound of claim 1, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof, wherein the compound is selected from the group consisting of:
Figure FDA0002006087970000041
Figure FDA0002006087970000051
Figure FDA0002006087970000061
6. a process for preparing a compound of claim 1, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof, selected from the group consisting of:
the method comprises the following steps:
Figure FDA0002006087970000071
removing a protecting group R' from the compound of the formula A under the condition of NaOH solution to obtain a compound of a formula I,
wherein, n, X, L, R1、R2、R3、R4As defined in claim 1;
r' is Ts;
the second method comprises the following steps:
Figure FDA0002006087970000072
cyclizing the compound of the formula B under the condition of NaOH solution to remove the protecting group R' to obtain the compound of the formula I,
wherein, n, X, L, R1、R2、R3、R4As defined in claim 1;
r' is Ts;
the third method comprises the following steps:
Figure FDA0002006087970000073
cyclizing the compound of the formula C under the condition of NaOH solution to remove the protecting group R' to obtain the compound of the formula I,
wherein, n, X, L, R1、R2、R3、R4As defined in claim 1;
r' is Ts.
7. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and one or more compounds of claim 1, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof.
8. Use of a pharmaceutical composition according to claim 7 for the preparation of a formulation for the prevention and/or treatment of a disease associated with an abnormal Hedgehog signaling pathway.
9. The use of claim 8, wherein the disease associated with an abnormal Hedgehog signaling pathway is selected from the group consisting of: cancer, inflammation, chronic liver disease, diabetes, cardiovascular disease.
10. A Gli inhibitor comprising a pharmaceutically acceptable carrier and an inhibitory effective amount of one or more compounds of claim 1, or an isomer, a pharmaceutically acceptable salt, or a pharmaceutically acceptable solvate or hydrate thereof.
CN201910228878.3A 2019-03-25 2019-03-25 Cyclic sulfonamide ring-substituted pyridone pyrrole compound and synthesis method and application thereof Pending CN111732585A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910228878.3A CN111732585A (en) 2019-03-25 2019-03-25 Cyclic sulfonamide ring-substituted pyridone pyrrole compound and synthesis method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910228878.3A CN111732585A (en) 2019-03-25 2019-03-25 Cyclic sulfonamide ring-substituted pyridone pyrrole compound and synthesis method and application thereof

Publications (1)

Publication Number Publication Date
CN111732585A true CN111732585A (en) 2020-10-02

Family

ID=72645742

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910228878.3A Pending CN111732585A (en) 2019-03-25 2019-03-25 Cyclic sulfonamide ring-substituted pyridone pyrrole compound and synthesis method and application thereof

Country Status (1)

Country Link
CN (1) CN111732585A (en)

Similar Documents

Publication Publication Date Title
WO2021129824A1 (en) New-type k-ras g12c inhibitor
KR102499780B1 (en) Heterocyclic compound serving as fgfr4 inhibitor
CN114929704A (en) Spiro-containing quinazoline compounds
KR20170069199A (en) Indazole compounds as fgfr kinase inhibitor, preparation and use thereof
CN112661745A (en) Compounds useful as CDK7 kinase inhibitors and uses thereof
CN112824420B (en) Compounds useful as EGFR kinase inhibitors and uses thereof
WO2012031563A1 (en) Heterocyclic amino berbamine derivatives, preparation method and use thereof
TWI832539B (en) Compounds used as CDK7 kinase inhibitors and their applications
CN108699030B (en) Substituted amino six-membered nitrogen heterocyclic compound and preparation and application thereof
WO2023016562A1 (en) Polycyclic compound and use thereof
JP7338896B2 (en) MDM2 inhibitor, its preparation method, pharmaceutical composition and application
WO2017181974A1 (en) Five-membered heterocyclic compound, preparation method therefor, pharmaceutical composition and use
CN102432622B (en) 4-amino oxadiazole epipodophyllotoxin derivative and preparation method and application thereof
CN105130884B (en) 5 methyl 2 (1H) Pyridione derivatives and its production and use
CN112243437A (en) Acryloyl group-containing nuclear transport modulators and uses thereof
CN106397408A (en) 5-methyl-2(1H) pyridone derivative and preparation method and application thereof
CN109476650B (en) Five-membered heterocyclic compound and preparation method, pharmaceutical composition and application thereof
CN111732585A (en) Cyclic sulfonamide ring-substituted pyridone pyrrole compound and synthesis method and application thereof
KR20240021239A (en) Compounds used as CDK kinase inhibitors and their uses
JP5856971B2 (en) Crystalline CDC7 inhibitor salt
CN114380805A (en) Substituted benzo or pyrido pyrimidine amine inhibitor and preparation method and application thereof
CN113754659A (en) Spiro-containing quinazoline compounds
CN105175326B (en) 5 methyl 2 (1H) Pyridione derivatives and its production and use
TWI831325B (en) Naphthyridine derivatives as ATR inhibitors and preparation methods thereof
WO2024008083A1 (en) Compound as cdk7 kinase inhibitor and use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20201002

WD01 Invention patent application deemed withdrawn after publication