CN112457331A - Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof - Google Patents

Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof Download PDF

Info

Publication number
CN112457331A
CN112457331A CN201910848960.6A CN201910848960A CN112457331A CN 112457331 A CN112457331 A CN 112457331A CN 201910848960 A CN201910848960 A CN 201910848960A CN 112457331 A CN112457331 A CN 112457331A
Authority
CN
China
Prior art keywords
substituted
unsubstituted
group
reaction
compound
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
CN201910848960.6A
Other languages
Chinese (zh)
Inventor
王喆
潜安然
李德亮
张基勇
曾志宏
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.)
Shanghai Longwood Biopharmaceuticals Co Ltd
Original Assignee
Shanghai Longwood Biopharmaceuticals Co Ltd
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 Shanghai Longwood Biopharmaceuticals Co Ltd filed Critical Shanghai Longwood Biopharmaceuticals Co Ltd
Priority to CN201910848960.6A priority Critical patent/CN112457331A/en
Priority to CN202080063271.5A priority patent/CN114364685A/en
Priority to PCT/CN2020/114264 priority patent/WO2021047556A1/en
Publication of CN112457331A publication Critical patent/CN112457331A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4355Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • 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
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • A61P5/16Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4 for decreasing, blocking or antagonising the activity of the thyroid hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Immunology (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Neurology (AREA)
  • Urology & Nephrology (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Obesity (AREA)
  • Neurosurgery (AREA)
  • Pulmonology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Emergency Medicine (AREA)
  • Transplantation (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides a nitrogen-containing heterocyclic compound, and a preparation method, a pharmaceutical composition and application thereof, and particularly provides a compound shown as a formula I below, or optical isomers thereofA body, hydrate, solvate, or pharmaceutically acceptable salt thereof; wherein, the definition of each group is described in the specification. The compounds of formula I are useful for treating diseases associated with the PD-1/PD-L1 signaling pathway.

Description

Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof
Technical Field
The invention relates to the field of small molecule medicines, and particularly provides a small molecule compound which can be used for treating diseases related to a PD-1/PD-L1 signal pathway.
Background
The immune system plays a crucial role in controlling and curing many diseases, such as various cancers, diseases caused by viruses, and the like. Cancer cells often multiply rapidly by evading or suppressing the immune system in some way. One such way is to alter the expression of activating and inhibitory molecules on immune cells. Blocking the suppressive immune checkpoint, like PD-1, has proven to be a very effective method of suppressing cancer cells.
PD-1 is programmed cell death protein-1, also known as CD 279. It is expressed mainly in activated T cells and B cells and functions to inhibit the activation of cells, which is a normal homeostatic mechanism of the immune system, and PD-1 is a protective wall of our human body because excessive T/B cell activation causes autoimmune diseases. PD-1 is a type I transmembrane glycoprotein composed of 268 amino acids, and its structure mainly includes an outer immunoglobulin variable region, a hydrophobic transmembrane region, and an intracellular region. The intracellular region contains two phosphorylation sites, located in the immunoreceptor tyrosine inhibitory motif and immunoreceptor tyrosine switch motif, which also demonstrates that PD-1 is capable of back-regulating T cell receptor-mediated signaling. PD-1 has two ligands, PD-L1 and PD-L2, which differ in the manner of expression. PD-L1, which is up-regulated in many tumor cells, binds to PD-1 on T cells, inhibits T cell proliferation and activation, leaves T cells in an inactive state, and ultimately induces immune escape.
PD-1/PD-L1 exerts a reverse immunomodulatory effect. When PD-1 binds to PD-L1, tyrosine in the tyrosine-converting motif domain of the immune receptor of T cells can be phosphorylated, and the phosphorylated tyrosine can bind to phosphatase protein tyrosinase 2 and protein tyrosinase 1. This can block activation of extracellular signal-regulated kinase, and can block activation of phosphoinositide 3-kinase (PI3K) and serine-threonine protein kinase (Akt), thereby inhibiting T lymphocyte proliferation and secretion of related cytokines. While the PD-1/PD-L1 signal inhibits the activation and proliferation of T cells, the secretion of cytokines interleukin 2, interferon gamma and IL-10 can be realized. In addition, PD-1/PD-L1 signal has similar immune function to B cell, and when PD-1 binds to B cell antigen receptor, PD-1 cytoplasm region acts with tyrosinase containing protein tyrosinase 2 binding site, thereby preventing B cell activation.
PD-1/PD-L1-based immunotherapy is a new generation of immunotherapy that is of great interest. In recent years, a series of surprising research results prove that the PD-1/PD-L1 inhibitor has strong antitumor activity on various tumors. PD-1/PD-L1 antibody inhibitors that are currently marketed are Ninolumab from BMS, Lamboluzumab from Merck, and Atezolizumab from Roche. In addition to this, there are many studied PD-1/PD-L1 antibody inhibitors, including Cure Tech's Pidilizumab, GSK's AMP-224, and AstraZeneca's MEDI-4736.
Although tumor immunotherapy is considered as a new generation of revolution for targeted post-treatment cancer therapy. However, currently marketed and researched PD-1 single drug has its own defects, including injection administration, oral administration, instability in vivo, easy protease decomposition, easy immune cross reaction, difficult purification, high production cost, etc. Therefore, small molecule inhibitors of the PD-1/PD-L1 interaction are better candidates for tumor immunotherapy.
In view of the foregoing, there is a pressing need in the art to develop novel small molecule inhibitors of the PD-1/PD-L1 interaction.
Disclosure of Invention
The invention aims to provide a novel small molecule inhibitor of PD-1/PD-L1 interaction.
In a first aspect of the present invention, there is provided a compound represented by the following formula I, or an optical isomer, hydrate, solvate, or a pharmaceutically acceptable salt thereof:
Figure BDA0002196260460000021
wherein n, m, p and q are each independently selected from 0,1, 2,3 or 4;
L1、L2selected from the group consisting of: a bond, substituted or unsubstituted C1-C4 alkylene, substituted or unsubstituted C2-C4 alkenylene, substituted or unsubstituted C2-C4 alkynylene, -S-, -O-, substituted or unsubstituted-NH-, -S (O)2-, substituted or unsubstituted-NHC (O) NH-, or,
Figure BDA0002196260460000022
Substituted or unsubstituted
Figure BDA0002196260460000023
Substituted or unsubstituted
Figure BDA0002196260460000024
Substituted or unsubstituted
Figure BDA0002196260460000025
Figure BDA0002196260460000026
Is a group having the structure shown below:
Figure BDA0002196260460000027
Figure BDA0002196260460000028
wherein the content of the first and second substances,
Z1selected from the group consisting of: o, S, NRf, N-O-Rf; wherein, said Rf is selected from the group consisting of: H. substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C8Cycloalkyl, substituted or unsubstituted C6-C10Aryl, substituted or unsubstituted C6-C10Heteroaryl, cyano, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstituted C1-C6Alkyl, -C (═ O) -substituted or unsubstituted C3-C10Cycloalkyl, -C (═ O) -substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6An alkynyl group;
Z2、Z3、Z4each independently selected from the group consisting of: n, CH2、N-O、SO、SO2C (═ O), NRa, CRa; wherein Ra is selected from the group consisting of: H. chloro, bromo, fluoro, iodo, cyano, halogen, hydroxy, nitro, NRf, substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C8Cycloalkyl, substituted or unsubstituted C6-C10Aryl, substituted or unsubstituted C6-C10Heteroaryl, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstituted C1-C6Alkyl, -C (═ O) -substituted or unsubstituted C3-C10Cycloalkyl, substituted or unsubstituted C2-C6Alkenyl, substituted or unsubstituted C2-C6Alkynyl, -C (═ O) -substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6An alkynyl group;
Y1、Y2、Y3each independently selected from the group consisting of: CH. CH (CH)2、NH、NRa、N、N-O、CF、CRa、C(Ra)2O, S, SO or SO2
Figure BDA0002196260460000031
Is a single bond or a double bond;
and is
Figure BDA0002196260460000032
Is an aromatic or non-aromatic moiety;
r is H, substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C6-C10Aryl, cyano, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstituted C1-C6Alkyl, -C (═ O) -substituted or unsubstituted C3-C10Cycloalkyl, -C (═ O) -substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6Alkynyl, or- (L)1a)r-(L2a)s-(L3a)s-, in which,
each L1aEach independently is a group selected from the group consisting of; chemical bond, substituted or unsubstituted C1-C7Alkylene, substituted or unsubstituted C2-C4 alkenylene, substituted or unsubstituted C2-C4 alkynylene, -S-, -O-, substituted or unsubstituted-NH-, -S (O)2-、
L2aSelected from the group consisting of: substituted or unsubstituted C6-C12 arylene, substituted or unsubstituted 5-12 membered heteroarylene having 1-3 heteroatoms, substituted or unsubstituted C3-C8 cycloalkylene, substituted or unsubstituted 5-10 membered heterocyclylene having 1-3 heteroatoms;
L3aselected from the group consisting of: H. substituted or unsubstituted C1-C10 alkyl, C1-C10 aryl, -CN, hydroxy, amino, carboxy, -CO-NH-SO2-Rg、-NH-SO2-Rg、-SO2-NH-CO-Rg
r is 1,2,3, 4, 5, 6;
s is 0,1, 2 respectively;
rd, Re are each independently selected from the group consisting of: H. substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C10Cycloalkyl, substituted or unsubstituted C6-C10An aryl group;
Figure BDA0002196260460000033
selected from the group consisting of: substituted or unsubstituted 5-12 membered heteroaryl, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-12 membered heterocyclyl, substituted or unsubstituted 5-12 membered C5-C12 cyclyl, wherein the 5-12 membered heteroaryl and 5-12 membered heterocyclyl have 1-4 heteroatoms selected from B, P, N, O, S, wherein P, N, O as ring-forming atoms may be oxo and one or more ring-forming carbon atoms may be replaced by carbonyl; or the said
Figure BDA0002196260460000034
Is absent; or the said
Figure BDA0002196260460000035
Is equal to
Figure BDA0002196260460000036
Figure BDA0002196260460000037
Each independently a ring-forming divalent group selected from the group consisting of:
Figure BDA0002196260460000038
Figure BDA0002196260460000039
Figure BDA00021962604600000310
wherein, the bonding position of the ring can be N or C;
R1、R2、R3and R4Each independently selected from the group consisting of: H. halogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C8 cycloalkyl, oxo (i.e., ═ O), ═ NRf, -CN, hydroxy, NRdRe (e.g., amino), substituted or unsubstituted C1-C6 amine, substituted or unsubstituted- (C1-C6 alkylene) -NH- (C1-C6 alkylene), or a pharmaceutically acceptable salt thereof,Carboxyl, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-12 membered heteroaryl having 1-3 heteroatoms, substituted or unsubstituted 5-12 membered heterocyclyl having 1-4 heteroatoms, substituted or unsubstituted
Figure BDA0002196260460000041
Substituted or unsubstituted
Figure BDA0002196260460000042
Wherein, Rb, Rc and RzEach independently selected from the group consisting of: H. substituted or unsubstituted C1-C8An alkyl group; or said Rb and Rc taken together with the adjacent N atom form a substituted or unsubstituted 5-10 membered heterocyclic group having 1-3 heteroatoms selected from N, S and O; or (L)1a)r-(L2a)s-(L3a)s-;
Unless otherwise specified, "substituted" means substituted with one or more (e.g., 2,3, 4, etc.) substituents selected from the group consisting of: halogen: including but not limited to-F, Cl, Br, -CH2Cl、-CHCl2、-CCl3、-CH2F、-CHF2、-CF3Oxo, -CN, hydroxy, amino, C1-C6 alkylamino, carboxy, -NHAc, a group selected from the group consisting of unsubstituted or substituted with one or more substituents selected from the group consisting of: C1-C6 alkyl, C1-C6 alkoxy, C6-C10 aryl, C3-C8 cycloalkyl, halogenated C6-C10 aryl, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O, 5-10 membered heterocyclyl having 1-3 heteroatoms selected from N, S and O; the substituents are selected from the following group: halogen, hydroxy, carboxy, cyano, C1-C6 alkoxy, C1-C6 alkylamino;
in the formulae above, any of the heteroatoms is selected from the group consisting of: B. p, N, S and O.
In another preferred embodiment, the compound has the structure shown in formula II below:
Figure BDA0002196260460000043
wherein n, m, p and q are each independently selected from 0,1, 2,3 or 4;
L1、L2selected from the group consisting of: a bond, substituted or unsubstituted C1-C4 alkylene, substituted or unsubstituted C2-C4 alkenylene, substituted or unsubstituted C2-C4 alkynylene, -S-, -O-, substituted or unsubstituted-NH-, -S (O)2-, substituted or unsubstituted-NHC (O) NH-, or,
Figure BDA0002196260460000044
Substituted or unsubstituted
Figure BDA0002196260460000045
Substituted or unsubstituted
Figure BDA0002196260460000046
Substituted or unsubstituted
Figure BDA0002196260460000047
Figure BDA0002196260460000048
Is a group having the structure shown below:
Figure BDA0002196260460000049
Figure BDA0002196260460000051
wherein the content of the first and second substances,
Z1selected from the group consisting of: o, S, NRf, N-O-Rf; wherein, said Rf is selected from the group consisting of: H. substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C8Cycloalkyl, substituted or unsubstituted C6-C10Aryl, substituted or unsubstituted C6-C10Heteroaryl, cyano, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstitutedC of (A)1-C6Alkyl, -C (═ O) -substituted or unsubstituted C3-C10Cycloalkyl, -C (═ O) -substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6An alkynyl group;
Z2、Z3、Z4each independently selected from the group consisting of: n, CH2、N-O、SO、SO2C (═ O), NRa, CRa; wherein Ra is selected from the group consisting of: H. chloro, bromo, fluoro, iodo, cyano, halogen, hydroxy, nitro, NRf, substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C8Cycloalkyl, substituted or unsubstituted C6-C10Aryl, substituted or unsubstituted C6-C10Heteroaryl, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstituted C1-C6Alkyl, -C (═ O) -substituted or unsubstituted C3-C10Cycloalkyl, substituted or unsubstituted C2-C6Alkenyl, substituted or unsubstituted C2-C6Alkynyl, -C (═ O) -substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6An alkynyl group;
Y1、Y2、Y3each independently selected from the group consisting of: CH. CH (CH)2、NH、NRa、N、N-O、CF、CRa、C(Ra)2O, S, SO or SO2
Figure BDA0002196260460000052
Is a single bond or a double bond;
and is
Figure BDA0002196260460000053
Are aromatic or non-aromatic segments.
R1、R2、R3And R4Each independently selected from the group consisting of: H. halogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C8 cycloalkyl, oxo (i.e., ═ O), ═ NRf, -CN, hydroxy, NRdRe (e.g., amino), substituted or unsubstituted C1-C6 amine, substituted or unsubstituted- (C1-C6 alkylene) -NH- (C1-C6 alkylene), carboxy, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-12 membered heteroaryl having 1-3 heteroatoms, substituted or unsubstituted 5-12 membered heterocyclic having 1-4 heteroatoms, substituted or unsubstituted
Figure BDA0002196260460000054
Substituted or unsubstituted
Figure BDA0002196260460000055
Or (L)1a)r-(L2a)s-(L3a)s-
In another preferred embodiment, the
Figure BDA0002196260460000056
Has a structure represented by the following formula:
Figure BDA0002196260460000061
wherein the content of the first and second substances,
X6、X7、X8、X9、X10and X11Each independently selected from the group consisting of: n, CR, respectively;
R6selected from the group consisting of: H. halogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C8 cycloalkyl, oxo (i.e., ═ O), ═ NRf, -CN, hydroxy, NRdRe (e.g., amino), substituted or unsubstituted C1-C6 amine, substituted or unsubstituted- (C1-C6 alkylene) -NH- (C1-C6 alkylene), carboxy, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-to 12-membered heteroaryl having 1 to 3 heteroatoms, substituted or unsubstituted having 1 to 4 heteroatoms5-12 membered heterocyclic group of hetero atom, substituted or unsubstituted
Figure BDA0002196260460000062
Substituted or unsubstituted
Figure BDA0002196260460000063
Or (L)1a)r-(L2a)s-(L3a)s-,-C0-8-O-R8,-C0-8-C(O)OR8,-C0-8-OC(O)OR8,-C0-8-NR8R9,-C0-8-N(R8)C(O)R9,-C0-8-C(O)NR8R9
R8And R9Each independently selected from the group consisting of: H. hydroxy, substituted or unsubstituted C1-C10 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C8 cycloalkyl, oxo (i.e., ═ O), ═ NRf, -CN, hydroxy, NRdRe (e.g., amino), substituted or unsubstituted C1-C6 amine, substituted or unsubstituted- (C1-C6 alkylene) -NH- (C1-C6 alkylene), carboxy, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-to 12-membered heteroaryl having 1 to 3 heteroatoms, substituted or unsubstituted 5-to 12-membered heterocyclyl having 1 to 4 heteroatoms, substituted or unsubstituted
Figure BDA0002196260460000064
Substituted or unsubstituted
Figure BDA0002196260460000065
Or (L)1a)r-(L2a)s-(L3a)s-;
In another preferred embodiment, R is6Is R1(ii) a Or is
Figure BDA0002196260460000066
Wherein Rb and Rc are each independently selected from the group consisting of: H. substituted or unsubstituted C1-C8An alkyl group; or said Rb and Rc taken together with the adjacent N atom form a substituted or unsubstituted 3-10 membered heterocyclic group having 1-3 heteroatoms selected from N, S and O.
In another preferred embodiment, the
Figure BDA0002196260460000067
Is an optionally substituted monovalent radical formed from a ring selected from the group consisting of:
Figure BDA0002196260460000068
4. the compound of claim 1, wherein the compound of formula I is selected from the group consisting of formulae Id-1, Id-2, and Id-3, or an optical isomer, hydrate, solvate, or pharmaceutically acceptable salt thereof:
Figure BDA0002196260460000071
wherein each group is as defined above.
In another preferred embodiment, the ring is
Figure BDA0002196260460000072
And/or
Figure BDA0002196260460000073
Having a substituent represented by formula IV below:
Figure BDA0002196260460000074
wherein each L is4Independently selected from the group consisting of: substituted or unsubstituted C1-C4 alkylene, -S-, -O-, -NRa-, -S (O) -, -S (O)2-; preferably a substituted or unsubstituted C1-C4 alkylene group, with the proviso that each L4The co-formed structure is chemically stable;
Figure BDA0002196260460000075
selected from the group consisting of: substituted or unsubstituted C5-C10 cycloalkyl, substituted or unsubstituted 3-10 membered heterocyclyl having 1-3 heteroatoms selected from B, P, N, S and O; preferably, said
Figure BDA0002196260460000076
Is a 3-8 membered nitrogen containing heterocyclic group;
each R is5Each independently selected from the group consisting of: substituted or unsubstituted C1-C6 alkyl, -CN, hydroxy, amino, carboxy; wherein the substituents are selected from the group consisting of: halogen, hydroxy, carboxy, cyano, C1-C6 alkoxy.
In another preferred embodiment, the compound is selected from the following table;
Figure BDA0002196260460000081
Figure BDA0002196260460000091
Figure BDA0002196260460000101
Figure BDA0002196260460000111
Figure BDA0002196260460000121
Figure BDA0002196260460000131
Figure BDA0002196260460000141
Figure BDA0002196260460000151
Figure BDA0002196260460000163
in a second aspect of the invention, there is provided a process for the preparation of a compound of formula I as defined in claim 1, said process comprising a step selected from the group consisting of those depicted in schemes 1,2 or 3:
synthesis scheme 1
Figure BDA0002196260460000161
(e) Taking halide 1-1 and appropriate coupling reagent 1-2 (such as boric acid, boric acid ester, tin reagent or Grignard reagent) as basic raw materials, and obtaining an intermediate compound 1-3 through coupling reaction (such as Suzuki, Stille or Kumada coupling) catalyzed by palladium or copper;
(f) taking the intermediate 1-3 as a raw material, and reacting with carboxylic acid 1-4 under the action of a condensing agent (such as HATU, EDCI or HBTU) to obtain an amide intermediate 1-5;
(g) taking the intermediate 1-5 as a raw material, and removing a protecting group (Boc) under an acidic condition to obtain an intermediate 1-6;
(h) taking the intermediate 1-6 as a raw material, and carrying out nucleophilic substitution reaction with a halide under an alkaline condition, or carrying out reductive amination reaction with aldehyde or ketone under the action of a reducing agent to obtain a target compound I;
synthesis scheme 2:
Figure BDA0002196260460000162
(e) taking carboxylic ester 2-1 as a raw material, and carrying out an aminolysis reaction with amine 2-2 under the catalysis of Lewis acid to obtain an intermediate compound 2-3;
(f) taking the intermediate 2-3 and a proper coupling reagent 2-4 (such as boric acid, boric acid ester, a tin reagent or a Grignard reagent) as basic raw materials, and obtaining an intermediate compound 2-5 through a coupling reaction (such as Suzuki, Stille or Kumada coupling) catalyzed by palladium or copper;
(g) taking the intermediate 2-5 as a raw material, and removing a protecting group (Boc) under an acidic condition to obtain an intermediate 2-6;
(h) taking the intermediate 2-6 as a raw material, and carrying out nucleophilic substitution reaction with a halide under an alkaline condition, or carrying out reductive amination reaction with aldehyde or ketone under the action of a reducing agent to obtain a target compound I;
(b) carrying out reductive amination reaction in the synthesis scheme 3 to obtain a target compound I;
synthesis scheme 3:
Figure BDA0002196260460000171
the method 3 comprises the following steps:
(e) carrying out Suzuki coupling on the raw materials of the boron ester 3-1 and the halide 3-2 under the catalysis of palladium to obtain an intermediate compound 3-3;
(f) carboxylic acid 3-4 and a compound 3-5 are taken as raw materials, and a cyclization reaction is carried out under the action of a proper dehydrating agent to obtain an intermediate compound 3-3;
(g) taking aldehyde 3-6 as a raw material, and carrying out cyclization reaction under the action of a proper oxidant to obtain an intermediate 3-3;
(h) using the intermediate 3-3 and a proper coupling reagent 3-7 (such as boric acid, boric acid ester, a tin reagent or a Grignard reagent) as basic raw materials, and performing coupling reaction (such as Suzuki, Stille or Kumada coupling) catalyzed by palladium or copper to obtain a target compound I;
said Cy is
Figure BDA0002196260460000172
Is composed of
Figure BDA0002196260460000173
Y1、Y2、Z1、Z2、Z3R ofThe definition is the same as above.
In a third aspect of the present invention, there is provided a pharmaceutical composition comprising (1) a compound according to the first aspect of the present invention or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof; (2) a pharmaceutically acceptable carrier.
In a fourth aspect of the present invention, there is provided a use of a compound according to the first aspect of the present invention or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, or a pharmaceutical composition according to the second aspect of the present invention, for the preparation of a pharmaceutical composition for the prevention and/or treatment of a disease associated with an activity or an expression amount of PD-1/PD-L1.
In a fifth aspect of the invention, there is provided a PD-1/PD-L1 inhibitor, said inhibitor comprising a compound according to the first aspect of the invention, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof.
In another preferred embodiment, the pharmaceutical composition is used for treating a disease selected from the group consisting of: cancer, infectious disease, autoimmune disease.
In another preferred embodiment, the cancer is selected from the group consisting of: pancreatic cancer, bladder cancer, colorectal cancer, breast cancer, prostate cancer, kidney cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, gastric cancer, esophageal cancer, melanoma, neuroendocrine cancer, central nervous system cancer, brain cancer, bone cancer, soft tissue sarcoma, non-small cell lung cancer, small cell lung cancer or colon cancer, skin cancer, lung cancer, urologic tumors, hematologic tumors, glioma, digestive system tumors, reproductive system tumors, lymphoma, nervous system tumors, brain tumors, head and neck cancer.
In another preferred embodiment, the cancer is selected from the group consisting of: acute Lymphocytic Leukemia (ALL), Acute Myelogenous Leukemia (AML), Chronic Lymphocytic Leukemia (CLL), Small Lymphocytic Lymphoma (SLL), myelodysplastic syndrome (MDS), myeloproliferative disorder (MPD), Chronic Myelogenous Leukemia (CML), Multiple Myeloma (MM), non-hodgkin's lymphoma (NHL), Mantle Cell Lymphoma (MCL), follicular lymphoma, Waldestrom Macroglobulinemia (WM), T-cell lymphoma, B-cell lymphoma or diffuse large B-cell lymphoma (DLBCL).
In another preferred embodiment, the infectious disease is selected from bacterial infection and viral infection.
In another preferred embodiment, the autoimmune disease is selected from organ-specific autoimmune disease, systemic autoimmune disease.
In another preferred embodiment, the organ-specific autoimmune disease comprises chronic lymphocytic thyroiditis, hyperthyroidism, insulin-dependent diabetes mellitus, myasthenia gravis, ulcerative colitis, pernicious anemia with chronic atrophic gastritis, goodpasture's syndrome, primary biliary cirrhosis, multiple sclerosis, acute idiopathic polyneuritis.
In another preferred embodiment, the systemic autoimmune disease comprises rheumatoid arthritis, systemic lupus erythematosus, systemic vasculitis, scleroderma, pemphigus, dermatomyositis, mixed connective tissue disease, autoimmune hemolytic anemia.
In another preferred embodiment, the pharmaceutical composition is also used for improving the T cell function of Chronic Hepatitis B (CHB) patients.
In another preferred embodiment, the inhibitor further comprises at least one therapeutic agent selected from the group consisting of: nivolumab, pembrolizumab, atezolizumab, or ipilimumab.
In a sixth aspect of the present invention, there is provided a method for inhibiting the PD-1/PD-L1 interaction in vitro, comprising the steps of: contacting a compound according to the first aspect of the invention, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof, with PD-L1 protein.
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 inventor has extensively and intensively studied and found a PD-1/PD-L1 interaction inhibitor with excellent inhibitory effect. On this basis, the inventors have completed the present invention.
Definition of
As used herein, the term "alkyl" includes straight or branched chain alkyl groups. E.g. C1-C8Alkyl represents a straight or branched chain alkyl group having 1 to 8 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, and the like.
As used herein, the term "alkenyl" includes straight or branched chain alkenyl groups. E.g. C2-C6Alkenyl means a straight or branched alkenyl group having 2 to 6 carbon atoms, such as vinyl, allyl, 1-propenyl, isopropenyl, 1-butenyl, 2-butenyl, or the like.
As used herein, the term "alkynyl" includes straight or branched chain alkynyl groups. E.g. C2-C6Alkynyl means straight or branched chain alkynyl having 2 to 6 carbon atoms, such as ethynyl, propynyl, butynyl, or the like.
As used herein, the term "C3-C10Cycloalkyl "refers to cycloalkyl groups having 3 to 10 carbon atoms. It may be a single ring, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or the like. It may also be in the form of a double ring, for example a bridged or spiro ring.
As used herein, the term "C1-C8Alkylamino "is defined as being substituted by C1-C8The amino group substituted by the alkyl can be mono-substituted or di-substituted; for example, methylamino, ethylamino, propylamino, isopropylamino, butylamino, isobutylamino, tert-butylamino, dimethylamino, diethylamino, dipropylamino, diisopropylamino, dibutylamino, diisobutylamino, di-tert-butylamino and the like.
As used herein, the term "C1-C8Alkoxy "means a straight or branched chain alkoxy group having 1 to 8 carbon atoms; for example, methoxy, ethoxy, propoxy, isopropoxyAnd (c) a group, butoxy group, isobutoxy group, tert-butoxy group, and the like.
As used herein, the term "3-10 membered heterocycloalkyl having 1-3 heteroatoms selected from the group consisting of N, S and O" refers to a saturated or partially saturated cyclic group having 3-10 atoms and wherein 1-3 atoms are heteroatoms selected from the group consisting of N, S and O. It may be monocyclic or may be in the form of a double ring, for example a bridged or spiro ring. Specific examples may be oxetane, azetidine, tetrahydro-2H-pyranyl, piperidinyl, tetrahydrofuranyl, morpholinyl, pyrrolidinyl, and the like.
As used herein, the term "C6-C10Aryl "means an aryl group having 6 to 10 carbon atoms, for example, phenyl or naphthyl and the like.
As used herein, the term "5-10 membered heteroaryl having 1-3 heteroatoms selected from the group consisting of N, S and O" refers to a cyclic aromatic group having 5-10 atoms and wherein 1-3 atoms are heteroatoms selected from the group consisting of N, S and O. It may be a single ring or a condensed ring form. Specific examples may be pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl, (1,2,3) -triazolyl and (1,2,4) -triazolyl, tetrazolyl, furyl, thienyl, isoxazolyl, thiazolyl, oxazolyl and the like.
Unless specifically stated to be "substituted or unsubstituted", the groups of the present invention may be substituted with a substituent selected from the group consisting of: halogen, nitrile group, nitro group, hydroxyl group, amino group, C1-C6Alkyl-amino, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Alkoxy, halo C1-C6Alkyl, halo C2-C6Alkenyl, halo C2-C6Alkynyl, halo C1-C6Alkoxy, allyl, benzyl, C6-C12Aryl radical, C1-C6alkoxy-C1-C6Alkyl radical, C1-C6Alkoxy-carbonyl, phenoxycarbonyl, C2-C6Alkynyl-carbonyl, C2-C6Alkenyl-carbonyl, C3-C6Cycloalkyl-carbonyl, C1-C6Alkyl-sulfonyl, and the like.
As used herein, "halogen" or "halogen atom" refers to F, Cl, Br, and I. More preferably, the halogen or halogen atom is selected from F, Cl and Br. "halogenated" means substituted with an atom selected from F, Cl, Br, and I.
Unless otherwise specified, the structural formulae depicted herein are intended to include all isomeric forms (e.g., enantiomers, diastereomers and geometric isomers (or conformational isomers)): for example, R, S configuration containing an asymmetric center, (Z), (E) isomers of double bonds, and the like. Thus, individual stereochemical isomers of the compounds of the present invention or mixtures of enantiomers, diastereomers or geometric isomers (or conformers) thereof are within the scope of the present invention.
As used herein, the term "tautomer" means that structural isomers having different energies may exceed the low energy barrier, thereby converting with each other. For example, proton tautomers (i.e., proton transmutations) include interconversion by proton shift, such as 1H-indazoles and 2H-indazoles. Valence tautomers include interconversion by recombination of some of the bonding electrons.
As used herein, the term "solvate" refers to a complex of a compound of the present invention coordinated to solvent molecules in a specific ratio.
As used herein, the term "hydrate" refers to a complex formed by the coordination of a compound of the present invention with water.
Active ingredient
As used herein, "compounds of the invention" refers to compounds of formula I, and also includes various crystalline forms, pharmaceutically acceptable salts, hydrates, or solvates of the compounds of formula I.
Preferred compounds of the present invention include compounds 1-360 (including various classes of R configuration and/or S configuration stereoisomers of each compound, and/or E-/Z-cis-trans isomers).
In another preferred embodiment, the pharmaceutically acceptable salts include salts formed in combination with inorganic acids, organic acids, alkali metal ions, alkaline earth metal ions, or organic bases capable of providing physiologically acceptable cations, as well as ammonium salts.
In another preferred embodiment, the inorganic acid is selected from hydrochloric acid, hydrobromic acid, phosphoric acid or sulfuric acid; the organic acid is selected from methanesulfonic acid, p-toluenesulfonic acid, trifluoroacetic acid, lycic acid, tartaric acid maleate, fumaric acid, citric acid or lactic acid; the alkali metal ions are selected from lithium ions, sodium ions and potassium ions; the alkaline earth metal ions are selected from calcium ions and magnesium ions; the organic base capable of providing a physiologically acceptable cation is selected from methylamine, dimethylamine, trimethylamine, piperidine, morpholine or tris (2-hydroxyethyl) amine.
All such salts within the scope of the present invention may be prepared by conventional methods. During the preparation of the compounds of formula I and solvates and salts thereof, different crystallization conditions may occur as polycrystals or co-crystals.
Preparation of Compounds of formula I
For the preparation of the compounds of the general formula I according to the invention, the compounds of the general formula I according to the invention can be obtained by the following process 1 or 2, depending on the structure of the general formula I.
The method 1 comprises the following steps:
Figure BDA0002196260460000201
(a) taking halide 1-1 and appropriate coupling reagent 1-2 (such as boric acid, boric acid ester, tin reagent or Grignard reagent) as basic raw materials, and obtaining an intermediate compound 1-3 through coupling reaction (such as Suzuki, Stille or Kumada coupling) catalyzed by palladium or copper;
(b) taking the intermediate 1-3 as a raw material, and reacting with carboxylic acid 1-4 under the action of a condensing agent (such as HATU, EDCI or HBTU) to obtain an amide intermediate 1-5;
(c) taking the intermediate 1-5 as a raw material, and removing a protecting group (Boc) under an acidic condition to obtain an intermediate 1-6;
(d) taking the intermediate 1-6 as a raw material, and carrying out nucleophilic substitution reaction with a halide under an alkaline condition, or carrying out reductive amination reaction with aldehyde or ketone under the action of a reducing agent to obtain a target compound I;
Figure BDA0002196260460000202
the method 2 comprises the following steps:
(c) taking carboxylic ester 2-1 as a raw material, and carrying out an aminolysis reaction with amine 2-2 under the catalysis of Lewis acid to obtain an intermediate compound 2-3;
(d) taking the intermediate 2-3 and a proper coupling reagent 2-4 (such as boric acid, boric acid ester, a tin reagent or a Grignard reagent) as basic raw materials, and obtaining an intermediate compound 2-5 through a coupling reaction (such as Suzuki, Stille or Kumada coupling) catalyzed by palladium or copper;
(e) taking the intermediate 2-5 as a raw material, and removing a protecting group (Boc) under an acidic condition to obtain an intermediate 2-6;
(f) taking the intermediate 2-6 as a raw material, and carrying out nucleophilic substitution reaction with a halide under an alkaline condition, or carrying out reductive amination reaction with aldehyde or ketone under the action of a reducing agent to obtain a target compound I;
Figure BDA0002196260460000211
the method 3 comprises the following steps:
(i) using carboxylic ester 3-1 as a raw material, and carrying out Buchwald-Hartwig coupling or Ullman coupling with amine 3-2 under the catalysis of palladium or copper to obtain an intermediate compound 3-3;
(j) taking the intermediate 3-3 and a proper coupling reagent 3-4 (such as boric acid, boric acid ester, a tin reagent or a Grignard reagent) as basic raw materials, and obtaining an intermediate compound 3-5 through a coupling reaction (such as Suzuki, Stille or Kumada coupling) catalyzed by palladium or copper;
(k) taking the intermediate 3-5 as a raw material, and removing a protecting group (Boc) under an acidic condition to obtain an intermediate 3-6;
(l) Taking the intermediate 3-6 as a raw material, and carrying out nucleophilic substitution reaction with a halide under an alkaline condition, or carrying out reductive amination reaction with aldehyde or ketone under the action of a reducing agent to obtain a target compound I;
said Cy is
Figure BDA0002196260460000212
Is composed of
Figure BDA0002196260460000213
Y1、Y2、Z1、Z2、Z3And R is as defined above.
In addition, the starting materials and intermediates in the above reactions are readily available, and the reactions in each step can be readily synthesized according to reported literature or by conventional methods in organic synthesis to those skilled in the art. The compounds of formula I may exist in the form of solvates or non-solvates, and crystallization using different solvents may give different solvates.
Pharmaceutical compositions and methods of administration
The compound of the present invention and various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates thereof, and pharmaceutical compositions containing the compound of the present invention as a main active ingredient can be used for preventing and/or treating (stabilizing, alleviating or curing) diseases (e.g., cancer, infectious diseases, autoimmune diseases) associated with the PD-1/PD-L1 interaction, because the compound of the present invention has excellent inhibitory activity of the PD-1/PD-L1 interaction.
The pharmaceutical compositions of the present invention comprise a safe and effective amount of a compound of the present invention in combination with a pharmaceutically 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, 10-200mg 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. "compatibility" is defined hereinIt is meant that the components of the composition can be admixed with the compounds of the invention and with each other 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, glycerol, mannitol, sorbitol, etc.), emulsifiers (e.g. tween, etc.)
Figure BDA0002196260460000221
) Wetting agents (e.g., sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
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, parenteral (intravenous, intramuscular or subcutaneous).
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.
The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds (e.g., other anti-cancer agents).
When administered in combination, the pharmaceutical composition further comprises one or more (2, 3, 4, or more) other pharmaceutically acceptable compounds. One or more (2, 3, 4, or more) of the other pharmaceutically acceptable compounds may be used simultaneously, separately or sequentially with the compounds of the invention for the prevention and/or treatment of a disease associated with the PD-1/PD-L1 interaction.
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 20 to 500 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.
The main advantages of the invention include:
(1) the compound has high inhibitory activity on the interaction of PD-1/PD-L1, has strong binding capacity with PD-L1 protein, and has the capacity of relieving the inhibition of IFN gamma by PD-L1.
(2) The compound of the invention has better solubility; the toxicity to normal cells is very low and can therefore be applied to the treated subject over a wide dose range.
(3) Compared with the prior art, the compound of the invention has better solubility and good drug forming property, and the compound of the invention has good bioavailability in vivo experiments, and in addition, compared with the prior art, the compound of the invention can be easily prepared into pharmaceutically acceptable salts, thereby being beneficial to further preparation.
(4) The in vivo efficacy research shows that the compound can obviously inhibit the growth of subcutaneous tumors no matter on the tumor volume or the tumor weight, and can obviously increase the number of lymphocytes in the blood and the spleen of a mouse.
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. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Unless otherwise indicated, percentages and parts are percentages and parts by weight.
The test materials and reagents used in the following examples are commercially available without specific reference.
General materials and test methods:
the instruments and materials involved in the examples are described below:
the NMR spectrum was obtained by analysis with a Bruker AV-400(400MHz) NMR spectrometer.
Chemical shifts are reported in ppm units using tetramethylsilane as an internal standard (CDC 1)3Delta 7.26 ppm). The data reported are the chemical shifts and their split and coupling constants (s: singlet; d: doublet; t: triplet; q: quartet; br: broad; m: multiplet).
Mass spectrometry data were analyzed using a liquid mass spectrometer from the Finnigen advanced LCQ company (Finnigan LCQ Advantage), all reactions operated under anhydrous and oxygen-free conditions under dry argon protection, except for other requirements. The solid organometallic compound was stored in an argon protected dry box.
The tetrahydrofuran and the diethyl ether are obtained by distillation, and metal sodium and benzophenone are added into the tetrahydrofuran and the diethyl ether during the distillation. Methylene chloride, pentane and hexane were treated with calcium hydride.
The special raw materials and intermediates involved in the present invention are provided by custom-made processing of Tianjin Changsen pharmaceutical Co., Ltd, and all other chemical reagents are purchased from reagent suppliers such as Shanghai chemical reagent company, Aldrich company (Aldrich), Acros company (Acros). If the intermediates or products needed by the reaction in the synthesis process are not enough for the next experiment, the synthesis is repeated for a plurality of times until the intermediates or products are enough.
The raw materials and reagents related to the invention can be obtained by commercial or customized processing and purchase except for special instructions.
The compounds of the invention may contain one or more asymmetric centres and so the series of compounds may be in racemic or single enantiomeric form. The compound prepared by the invention is a heterocyclic compound with the purity of higher than 95%, and the structure table of each final productCharacterized by MS or/and hydrogen nuclear magnetic resonance (1H NMR) analysis. The synthesis of the various compounds and intermediates of the invention is illustrated by the examples below.
EXAMPLE 1 Synthesis of LW1005-001 Compound
Figure BDA0002196260460000241
Step 1-1:
Figure BDA0002196260460000242
compound 1(51g, Journal of Medicinal Chemistry,2019,62,276-287) and 2-methyl-3-bromobenzoic acid (95.56g) were successively added to PPA (500g) and reacted at 140 ℃ for 6 hours with mechanical stirring. After the reaction is completed, pouring ice water into the reaction bottle, pouring out after dilution, pulping for 30min, and filtering. Filtering to obtain solid, adding 500ml water, adding sodium hydroxide solid under mechanical stirring, adjusting pH to 6-8, and filtering. The solid filter cake is dried at 55 ℃ to obtain 80g of off-white solid product. MS-APCI:305[ M + H]+.
Step 1-2:
Figure BDA0002196260460000243
3(2.0g), DMAP (805mg) was added to DMF (30mL), the solution was cloudy, Boc2O was added, the solid was dissolved and the reaction was heated at 40 ℃ overnight. TLC showed 1 small amount remaining. The reaction solution was spin-dried with an oil pump. The solid was slurried with EA/HEP and filtered to give 1.6g of the product as a white solid. MS-APCI of 405.2[ M + H ]]+
Step 1-3:
Figure BDA0002196260460000244
4(202mg), 5(230mg, WO2018119286), Pd (dppf) Cl2/DCM (19.5mg) and Na were added at room temperature2CO3(21.2mg) was placed in the reactionThe flask was degassed under vacuum, and dioxane/H2O (5mL/1mL) was injected via syringe, and the reaction was heated at 100 ℃ for 4H after degassing again. Pouring the reaction solution into water, extracting with EA, drying, spin-drying the EA layer, pulping with EA/HEP (1:1), filtering to obtain 100mg crude product, and separating and purifying 50mg to obtain 9.0mg LW1005-001 as yellow solid. MS-APCI 559.2[ M + H ]]+
1H NMR(400MHz,DMSO-d6)δ11.85(d,J=6.1Hz,1H),10.55-10.28(m,1H),9.08(s,1H),8.52(s,1H),8.16(s,1H),8.0-8.05(m,2H),7.51(q,J=7.1,6.4Hz,2H),7.46–7.32(m,2H),7.25(d,J=6.1Hz,1H),7.06(s,1H),6.87(d,J=7.1Hz,1H),5.51(s,1H),4.68(d,J=25.9Hz,2H),4.47(d,J=27.3Hz,2H),2.45(s,3H),2.34-2.29(m,1H),2.06(s,3H),2.02-2.01(m,1H),1.90-1.86(m,1H),1.52-1.46(m,1H).
EXAMPLE 2 Synthesis of the Compound LW1005-002
Figure BDA0002196260460000251
Step 2-1:
Figure BDA0002196260460000252
compound 3(10g) was dissolved in DMF (150ml), and 6(14.3 g; Macromolecules, 2015, 48, 1688-1702) and Cs2CO3(21.4g) were added to react at room temperature overnight. The reaction was complete by TLC. Filtration, addition of water to the filtrate followed by extraction with EtOAc, spin drying of the organic phase and slurrying with HEP: EtOAc 10:1 afforded 11g of 7 as a white solid. MS-APCI:587.1[ M + H ] +
1H NMR(400MHz,Chloroform-d)δ8.05(dd,J=7.9,1.3Hz,1H),7.75(dd,J=8.0,1.3Hz,1H),7.54(dt,J=6.7,1.5Hz,4H),7.49(d,J=7.3Hz,1H),7.41–7.35(m,2H),7.34–7.27(m,4H),7.22(t,J=7.9Hz,1H),6.58(d,J=7.3Hz,1H),4.26(t,J=4.7Hz,2H),4.05(dd,J=5.5,4.1Hz,2H),2.92(s,3H),1.05(s,9H).
Step 2-2:
Figure BDA0002196260460000253
7(11g) was dissolved in THF (110mL) at room temperature, 1NTBAF/THF (20.7mL) was added and the reaction was allowed to proceed for 2 hours. TLC showed the starting material reaction was complete. The reaction was spun dry and chromatographed (DCM: MeOH ═ 20:1) to give 4.5g of product 8 as a white solid. MS-APCI 349.1[ M + H ]]+
Step 2-3:
Figure BDA0002196260460000254
8(350mg), Bpin2(304.8mg), Pd (dppf) Cl2(81.7mg) and KOAc (196mg) were placed in a reaction flask at room temperature, degassed under vacuum, and dioxane (5mL) was injected via a syringe, degassed again, and then heated at 100 ℃ for reaction overnight. TLC showed the reaction was complete. The reaction was extracted with water and EtOAc, the organic phase washed with saturated NaCl, dried, spun-dried and chromatographed (DCM: MeOH ═ 20:1) to give 350mg of compound 9 as a grey solid. MS-APCI 397.1[ M + H ]]+
Step 2-4:
Figure BDA0002196260460000261
9(350mg), 10(364mg, WO2018119286), Pd (dppf) Cl2/DCM (71.9mg) and Na at room temperature2CO3(186.6mg) was placed in a reaction flask, degassed under vacuum, and dioxane/H2O (3.6mL, 5:1) was injected via syringe, and the reaction was heated at 100 ℃ for 4 hours after degassing again. TLC showed the starting material reaction was complete. The reaction was extracted with water and EtOAc, and the organic phase was washed with saturated NaCl, dried, spun-dried and passed through a column (DCM: MeOH ═ 20:1) to give 45mg of the compound LW1005-002 as a yellow solid.
MS-APCI:603.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ9.33(s,1H),8.88(d,J=2.0Hz,1H),8.47(d,J=8.2Hz,1H),8.19(d,J=1.6Hz,1H),8.13–8.00(m,2H),7.77(d,J=7.3Hz,1H),7.51(t,J=7.7Hz,1H),7.37(dd,J=14.2,7.4Hz,2H),7.19(d,J=5.8Hz,1H),6.92(dd,J=7.2,4.3Hz,2H),4.90(t,J=5.4Hz,1H),4.72(d,J=4.5Hz,1H),4.23(s,1H),4.11(t,J=5.4Hz,2H),3.82(q,J=13.8Hz,2H),3.68(q,J=5.5Hz,2H),2.80–2.72(m,1H),2.67(d,J=8.0Hz,2H),2.43(s,3H),2.39–2.30(m,1H),2.10(s,3H),2.03(dd,J=13.4,6.9Hz,1H),1.61–1.55(m,1H)
EXAMPLE 3 Synthesis of Compound LW1005-003
Figure BDA0002196260460000262
Step 3-1:
Figure BDA0002196260460000263
3(1.38g) was added to DMF (40ml) at room temperature, Cs was added2CO3(2.95g) and Compound 11(1.45 g; Organic Letters,2018,20,6938-6942) were reacted at room temperature overnight. Water is added into reaction liquid, stirring and filtering are carried out, and the crude solid is pulped by EA/HEP (1:10) and filtered to obtain 1.33g of product. MS-APCI 491.1[ M + H ]]+
Step 3-2:
Figure BDA0002196260460000264
12(1.33g) was added to 30ml of dioxane, and Pd (dppf) Cl2(0.22g), KOAc (0.53g), bis (pinacolato) borate (0.76g) were added to the mixture, and the mixture was reacted at 100 ℃ for 3 hours while purging with nitrogen. And (3) post-treatment: adding water, filtering, extracting with EA, passing through a column with HEP (high efficiency particulate) and EA being 3:1, and spin-drying to obtain 1.14g of the product. MS-APCI 539.2[ M + H ]]+
Step 3-3:
Figure BDA0002196260460000271
13(1.14g) was added to 30mL of dioxane/6mL of water, and Pd (dppf) Cl2.DCM (0.17g), Na2CO3(0.45g) and 10(875mg, WO2018119286), purged with nitrogen and reacted at 100 ℃ for 4 h. And (3) post-treatment: water was added for filtration, EA extracted, DCM/MeOH 20:1 column filtered, spin dried to give 0.9g of product. MS-APCI 745.2[ M + H ]]+
Step 3-4:
Figure BDA0002196260460000272
14(100mg) was dissolved in tetrahydrofuran (10mL) and 4N HCl/dioxane (2mL) was added dropwise, r, t. TLC detection reaction was complete, and some of them were preparative separated to yield 20mg of product as a yellow solid. MS-APCI 631.2[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.56-10.31(m,1H),9.08(s,1H),8.52(s,1H),8.18(s,1H),8.06(dd,J=7.9,1.4Hz,2H),7.86(d,J=7.4Hz,1H),7.52(t,J=7.7Hz,1H),7.45–7.35(m,2H),7.25(d,J=6.0Hz,1H),7.06(s,1H),6.94(d,J=7.3Hz,1H),5.51(s,1H),4.77-4.62(m,2H),4.50-4.43(m,2H),4.06(t,J=7.2Hz,2H),3.70-3.65(m,3H),3.42(t,J=6.4Hz,2H),2.44(s,3H),2.34-2.30(m,1H),2.06(s,3H),2.02-1.96(m,1H),1.87-1.82(m,1H),1.72(p,J=7.3Hz,2H),1.44(p,J=6.6Hz,2H).
Example 4 Synthesis of Compound LW1005-004
Figure BDA0002196260460000273
Step 4-1:
Figure BDA0002196260460000274
15(1.0g) was dissolved in DMF (10mL), and imidazole (0.64g), TBSCl (1.1g) was added to the solution to react at room temperature overnight. TLC plate reaction was complete. The reaction solution was washed with water, EA extracted, and the EA layer was spin-dried and passed through a column to obtain 1.6g of a pale yellow oily product.
Step 4-2:
Figure BDA0002196260460000281
LAH (0.33g) was added to THF (15mL), compound 16(1.6g in 5mL THF) was added dropwise at 0 deg.C and reacted for 1 hour at 0 deg.C. TLC showed the reaction was complete. To the reaction mixture was added 0.33mL of water, 0.33mL of NaOH (10%), and 0.33mL of water at 0 deg.C, followed by filtration and spin-drying of the filtrate to give 1.3g of a pale yellow oily product.
Step 4-3:
Figure BDA0002196260460000282
17(0.5g) and TEA (0.41g) were dissolved in DCM (5mL), and MsCl (0.35g) was added dropwise at 0 ℃ to react at 0 ℃ for 1 hour. TLC showed the reaction was complete. The reaction was washed with water, extracted with DCM and spin dried to give 0.66g of a pale yellow oily product.
Step 4-4:
Figure BDA0002196260460000283
18(0.66g),3(0.48g),Cs2CO3(1.5g) was added to DMF (20mL) and reacted at room temperature overnight. The reaction solution was poured into water and a solid precipitated. Filtration and spin-drying of the solid gave 0.5g of compound 19 as a white solid. MS-APCI 531.1[ M + H ]]+
And 4-5:
Figure BDA0002196260460000284
19(200mg), 5(142.6mg, WO2018119286), Pd (dppf) Cl at room temperature2/DCM (31mg) and Na2CO3(80.6mg) was placed in a reaction flask, vacuum degassed, and dioxane/H was injected via syringe2O (6mL,5:1), degassed again and then heated at 100 ℃ for reaction overnight. The reaction was washed with water and chromatographed (DCM: MeOH ═ 20:1) to give 120mg of product 20.MS-APCI:785.4[ M + H ]]+
And 4-6:
Figure BDA0002196260460000285
20(120mg) was dissolved in THF (3mL), 4N HCl/dioxane (0.3mL) was added and the reaction was carried out at room temperature for 3After 0min, a solid precipitated. And (5) filtering. Preparation of the solid isolation gave 40mg of the product as a yellow solid. MS-APCI 671.3[ M + H ]]+
1H NMR(400MHz,DMSO-d6)δ10.61-10.46(m,1H),9.11(s,1H),8.55(s,1H),8.06(dd,J=7.9,1.4Hz,2H),7.97(s,1H),7.79(d,J=7.3Hz,1H),7.52(t,J=7.7Hz,1H),7.44(t,J=7.8Hz,1H),7.38(dd,J=7.7,1.4Hz,1H),7.27(d,J=6.3Hz,1H),7.12(s,1H),6.92(d,J=7.3Hz,1H),4.71-4.66(m,2H),4.50-4.44(m,2H),3.90(d,J=7.2Hz,2H),3.68-3.50(m,2H),3.17-3.11(m,1H),2.45(s,3H),2.37-3.19(m,1H),2.03-1.90(m,1H),1.82-1.75(m,3H),1.72-1.68(m,1H),1.57-1.51(m,2H),1.12-1.01(m,4H).
EXAMPLE 5 Synthesis of the Compound LW1005-005
Figure BDA0002196260460000291
Step 5-1:
Figure BDA0002196260460000292
7(1g), Bpin2(605mg), Pd (dppf) Cl was added at room temperature2(180mg) and KOAc (431.2mg) were placed in a reaction flask, vacuum degassed, and dioxane (15mL) injected via syringe, and the reaction was heated at 100 ℃ for 2h after degassing again. TLC showed the reaction was complete. Adding water to the reaction solution, extracting with EtOAc, washing the organic phase with saturated NaCl, drying, spin-drying, and passing through a column to obtain 900mg of a yellow solid 21.MS-APCI:635[ M + H ]]+
Step 5-2:
Figure BDA0002196260460000293
21(102mg), 10(82.4mg, WO2018119286), Pd (dppf) Cl2/DCM (16.3mg) and Na2CO3(42.4mg) were placed in a reaction flask at room temperature, degassed under vacuum, and the syringe was filled with dioxane/H2O (5mL/1mL), and the reaction was heated to 100 ℃ overnight after degassing again. TLC showed the reaction was complete. The reaction was washed with water, extracted with EA and passed through a column (DCM: MeOH: 20:1) to give 80mg of the product as a yellow solidAnd (3) a solid. MS-APCI:842[ M + H]+
Step 5-3:
Figure BDA0002196260460000301
22(500mg), BzCl (146.6mg), TEA (211.5mg) were added to DCM (5mL) and reacted overnight at room temperature. TLC showed the starting material reaction was complete. The reaction was poured into water, extracted with DCM, dried and applied to a column (EA/HEP ═ 1:1) and spin dried to give 350mg of a yellow solid. MS-APCI:945[ M + H ]]+
Step 5-4:
Figure BDA0002196260460000302
23(350mg) was dissolved in DCM (5mL), TBAF (1mL,1M in THF) was added and the reaction was run for 10min at RT and was checked by TLC for completion. The reaction was washed with brine and then passed through a column (DCM: MeOH ═ 20:1) to give 200mg of the product as a yellow solid. MS-APCI 707.3[ M + H ]]+
Step 5-5:
Figure BDA0002196260460000303
24(100mg) was dissolved in DMF (2mL), Dess-martin (120mg) was added at zero degrees and the reaction was continued overnight at zero degrees. TLC showed approximately 20% starting material remaining. Adding saturated NaHCO into reaction liquid3Quenching, EA extraction, spin-drying the EA layer, and directly feeding the crude product to the next step. MS-APCI 705.2[ M + H ]]+
And 5-6:
Figure BDA0002196260460000304
25(50mg),26(12.3mg), TEA (21.2mg) were added to DMF (2mL), reacted at room temperature for 1 hour, and NaBH was added3CN (6.9mg), and reacted at room temperature overnight. Washing the reaction solution with water, extracting with EA, drying, and separating the crude product to obtain 10mg of product 27And a yellow solid. MS-APCI 804.3[ M + H ]]+
And 5-7:
Figure BDA0002196260460000305
27(10mg) was dissolved in MeOH (2mL), K2CO3(3mg) was added and the reaction was allowed to proceed at room temperature for 1 hour, and TLC indicated that 27 was complete. The reaction solution was spun dry, dissolved in THF/MeOH/H2O (1:1:1), and LiOH2O (1mg) was reacted at room temperature for 10 minutes. The reaction was complete by TLC. The reaction solution was prepared and separated to obtain 5.5mg of a yellow solid product. MS-APCI 686.3[ M + H ]]+
1H NMR(400MHz,DMSO-d6)δ10.57-10.47(m,1H),9.80-9.68(m,1H),9.03(s,1H),8.48(s,1H),8.22-8.45(m,1H),8.08–8.01(m,2H),7.82(d,J=7.5Hz,1H),7.50(t,J=7.7Hz,1H),7.37(d,J=7.4Hz,1H),7.21(d,J=6.0Hz,1H),7.04(d,J=7.4Hz,1H),7.03–6.92(m,2H),4.78–4.62(m,2H),4.53–4.34(m,2H),4.36–4.15(m,6H),3.32–3.24(m,2H),2.42(s,3H),2.34–2.24(m,2H),2.03(s,3H),2.00–1.91(m,1H),1.87–1.79(m,1H),1.49–1.40(m,1H),1.37–1.31(m,1H).
EXAMPLE 6 Synthesis of the Compound LW1005-006
Figure BDA0002196260460000311
Step 6-1:
Figure BDA0002196260460000312
25(50mg),28(7.8mg), TEA (21.5mg) were added to DMF (2mL) and reacted at room temperature for 1 hour, followed by addition of NaBH3CN (6.7mg), and reacted at room temperature overnight. The reaction solution was washed with water, extracted with EA, dried, and the crude product was isolated to yield 10mg of product 29 as a yellow solid. MS-APCI 762.3[ M + H ]]+
Step 6-2:
Figure BDA0002196260460000313
29(10mg) in MeOH (2mL) and K added2CO3(5.4mg) was reacted at room temperature for 1 hour, and TLC showed completion of the reaction of the starting materials. Preparation and isolation of the reaction mixture yielded 3mg of LW1005-006 as a yellow solid. MS-APCI 658.3[ M + H ]]+
1H NMR(400MHz,DMSO-d6)δ10.65(s,1H),10.63-9.66(m,1H),9.08(s,1H),8.53(s,1H),8.14(s,1H),8.06(d,J=7.9Hz,1H),8.03(d,J=6.2Hz,1H),7.85(d,J=7.4Hz,1H),7.54(t,J=7.7Hz,1H),7.42(dd,J=9.9,7.4Hz,2H),7.24(d,J=6.1Hz,1H),7.06(dd,J=7.5,3.7Hz,2H),4.74-4.62(m,2H),4.58–4.43(m,2H),4.40-4.29(m,4H),3.99-3.85(m,3H),3.33-3.31(m,3H),2.45(s,3H),2.34-2.31(m,1H),2.05(s,3H),2.00-1.95(m,1H),1.89-1.81(m,1H),1.30-1.21(m,3H).
EXAMPLE 7 Synthesis of Compound LW1005-007
Figure BDA0002196260460000321
Step 7-1:
Figure BDA0002196260460000322
25(90mg),30(180mg), TEA (254.5mg) was added to DMF (2mL), reacted at room temperature for 1 hour, and NaBH was added3CN (79mg), and reacted at room temperature overnight. The reaction was washed with water, extracted with EA, dried and purified by TLC plate to give 40mg of product 31 as a yellow solid. MS-APCI 832.3[ M + H ]]+
Step 7-2:
Figure BDA0002196260460000323
31(40mg) in MeOH (2mL) and K added2CO3(13.3mg) was reacted at room temperature for 1 hour, heated at 40 ℃ for 10 minutes, and TLC showed that the reaction was complete 2. Isolation of the reaction solution yielded 8.2mg of LW1005-007 as a yellow solid. MS-APCI 714.3[ M ]+H]+
1H NMR(400MHz,DMSO-d6)δ10.56–10.36(m,1H),9.24–9.17(m,1H),9.06(s,1H),8.51(s,1H),8.22(s,1H),8.08–8.04(m,2H),7.89(d,J=7.4Hz,1H),7.53(t,J=7.8Hz,1H),7.40(d,J=6.9Hz,2H),7.24(d,J=5.9Hz,1H),7.09(d,J=7.2Hz,1H),6.97(s,1H),5.58–5.43(s,1H),4.73–4.62(m,2H),4.50-4.38(m,2H),3.75-3.64(m,4H),3.34-3.27(m,4H),3.15-2.94(m,5H),2.44(s,3H),2.15-2.10(m,2H),2.07(s,3H),2.04-1.94(m,1H),1.78-1.67(m,2H).
EXAMPLE 8 Synthesis of the Compound LW1005-008
Figure BDA0002196260460000324
Step 8-1:
Figure BDA0002196260460000331
raw materials 8(19g) and NIS (14.7g) were mixed, MeCN (200mL) was added, and 1mLCF was added dropwise3COOH, stirred in an oil bath at 55 ℃ for 2.5 hours. Filtration afforded 10.5g of a white solid. MS-APCI:475[ M + H ]]+
Step 8-2:
Figure BDA0002196260460000332
mixing the raw materials 32(10g), Zn (CN)2(2.5g),Pd2(dba)3(900mg), DPPf (1.2g) in a 500mL single-necked flask, N2The mixture was displaced 3 times, and 200ml of DMF was injected and stirred at 85 ℃ for 2 hours. After completion of the reaction, the reaction solution was poured into water and filtered to obtain 6.3g of a white solid. MS-APCI 374[ M + H ]]+
Step 8-3:
Figure BDA0002196260460000333
starting material 33(5.0g) was dispersed in DCM (250ml) and stirred overIn the process, Dess-Martin (11.3g) is added, the reaction is carried out for 30min at room temperature, a TLC point plate is completely reacted, then saturated sodium bicarbonate solution (250ml x 2) is sequentially added and stirred, the water phase is separated, 10% sodium thiosulfate (250ml x 2) is added into the organic phase and stirred, the mixture is kept stand, extracted, dried and concentrated to obtain orange solid 4.0g, and the orange solid is directly put into the next step. MS-APCI:372[ M + H ]]+
Step 8-4:
Figure BDA0002196260460000334
starting material 34(4.0g),35(3.53g) was dissolved in DCM (200mL) and Et was added3N (2.4g), one hour after reaction NaBH (OAc) was added3(2.25g) and reacted at room temperature for 1 hour, TLC showed complete reaction, 200ml of water was added, stirred, separated, extracted, dried, concentrated and purified by column chromatography to give 3.5g of pale yellow solid. MS-APCI 541[ M + H ]]+
And 8-5:
Figure BDA0002196260460000335
36(1080mg),5(756mg,WO2018119286),Pd(dppf)Cl2/DCM(167.4mg),Na2CO3(530mg) in a reaction flask, N2Degassing protection, injecting 15mL of dioxane/3mL of water into a reaction flask, and reacting for 2 hours at 90 ℃. TLC plate 36 was completely reacted. The reaction was washed with water, extracted with EA, dried, spun-dried, and passed through a column (DCM: MeOH ═ 30:1) to afford 1.0g of the product as a yellow solid. MS-APCI 795.3[ M + H ]]+
And 8-6:
Figure BDA0002196260460000341
compound 37(3.9g) was dissolved in THF (50mL) and added TFA (10mL) and stirred at room temperature overnight. The reaction was complete by TLC. The reaction was spun off THF and the TFA was pumped dry to afford the crude yellow solid. Further purification on column (DCM: MeOH ═ 10:1) afforded 3.5g of product. MS (Mass Spectrometry)-APCI:739.3[M+H]+
1H NMR(400MHz,DMSO-d6)δ10.57(s,1H),9.06(s,1H),8.85(s,1H),8.51(s,1H),8.11–8.09(m,2H),7.57(t,J=7.7Hz,1H),7.49–7.34(m,2H),7.24(d,J=6.1Hz,1H),7.02(s,1H),4.77-4.64(m,2H),4.53-4.38(m,3H),3.72-3.64(m,2H),3.35-3.26(m,2H),3.20–2.91(m,4H),2.44(s,3H),2.16-2.10(m,2H),2.07(s,3H),2.01-1.95(m,1H),1.95-1.83(m,2H),1.80-1.66(m,2H).
EXAMPLE 9 Synthesis of the Compound LW1005-009
Figure BDA0002196260460000342
Step 9-1:
Figure BDA0002196260460000343
starting material 5(5.0g, WO2018119286) was dissolved in DCM (100mL), Et3N (3.2g) was added, stirring was carried out, BzCl (3.0g) was added, and the reaction was carried out at room temperature for 3 hours. After the reaction is finished, washing with saturated salt water, DCM extracting, drying, concentrating and purifying by column chromatography to obtain 3.7g of a yellow solid MS-APCI (American Petroleum institute) -565.3 [ M + H ] (M + H)]+
Step 9-2:
Figure BDA0002196260460000344
starting material 38(1.97g), starting material 33(3.3g), Pd (dppf) Cl2/DCM (431mg), Na2CO3(1.12g) were mixed, purged with nitrogen, and then Dioxane/H2O (4:1) (50mL) was added to the mixture to conduct a reaction at 95 ℃ for 3 hours. After the reaction is finished, washing with water, extracting with EA, drying, concentrating, and purifying by column chromatography to obtain 2.7g of yellow solid. MS-APCI 732.3[ M + H ]]+Step 9-3:
Figure BDA0002196260460000351
39(200mg) was dissolved in DCM (20mL), DMP (406mg) was added at room temperature,the reaction was carried out at room temperature for 10 minutes. TLC spot plate reaction is complete. The reaction solution is firstly saturated NaHCO3Washing with 10% Na2S2O3And (6) washing. The aqueous phase was extracted with DCM. The DCM layers were combined, dried, spun dried and used directly in the next reaction. MS-APCI 730.3[ M + H ]]+
Step 9-4:
Figure BDA0002196260460000352
40(150mg) in DCM (15mL) was added 41(52mg), TEA (103mg), reacted at room temperature for 1 hour, and NaBH (OAc) was added3(216.2mg) and reacted at room temperature overnight. TLC spot plate reaction is complete. Saturated NaHCO of reaction solution3Washed, extracted with DCM, dried and passed through the column to give the product 100mg as a yellow solid. MS-APCI 815.3[ M + H ]]+Step 9-5:
Figure BDA0002196260460000353
compound 6(100mg) was dissolved in 10mL THF, and 2d NaOMe (5N in MeOH) was added dropwise at room temperature. The reaction was carried out for 5 minutes. Solid was precipitated. TLC point plate raw material reaction is complete. The reaction solution was neutralized with 4N HCl/dioxane. Isolation by spin-drying gave 19mg of product as a yellow solid. MS-APCI 711.3[ M + H ]]+
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),8.87(s,1H),8.71(s,1H),8.47(d,J=8.4Hz,1H),8.20(d,J=1.7Hz,1H),8.08(dd,J=6.7,4.1Hz,2H),7.54(t,J=7.6Hz,1H),7.42(d,J=7.7Hz,1H),7.36(t,J=7.8Hz,1H),7.19(d,J=5.7Hz,1H),6.93(d,J=7.6Hz,1H),4.75-4.70(m,1H),4.57-4.53(m,1H),4.26-4.21(m,1H),4.18-4.14(m,2H),3.82(q,J=13.8Hz,2H),3.51-3.82(m,3H),2.77-2.72(m,3H),2.67-2.64(m,1H),2.60-2.53(m,3H),2.43(s,3H),2.39-2.36(m,1H),2.09(s,3H),2.02-1.98(m,1H),1.71-1.65(m,2H),1.60-1.55(m,1H).
Example 10 Synthesis of Compound LW1005-010
Figure BDA0002196260460000361
Step 10-1:
Figure BDA0002196260460000362
compound 43(306mg, FCH group),44(633.6 mg; Organic Process Research)&Development,2018,22,97-102.) and Na2S2O5(570mg) was added to DMF (3mL) and the reaction was carried out overnight at 110 ℃ under protection of N2 and checked by LCMS. After the reaction was completed, the reaction solution was washed with water, EA extracted, and passed through a column to obtain 360mg of a pale yellow solid. MS-APCI:332[ M + H]+.1H NMR(400MHz,Chloroform-d)δ8.04(d,J=5.8Hz,1H),7.74(dd,J=8.0,1.3Hz,1H),7.38(dd,J=7.7,1.4Hz,1H),7.20(t,J=7.8Hz,1H),7.02(d,J=5.8Hz,1H),4.20(s,3H),3.61(s,3H),2.29(s,3H).
Step 10-2:
Figure BDA0002196260460000363
45(260mg) was added to HBr/AcOH (4mL,1:1), reacted at 85 ℃ for 1.5h and the reaction was complete by TLC. The reaction solution was spin-dried and used directly in the next reaction. MS-APCI:318[ M + H ]]+.
Step 10-3:
Figure BDA0002196260460000364
46(344mg),6(471mg) and Cs2CO3(710mg) was added to DMF (3mL) and reacted at room temperature overnight. The reaction was checked by TLC. After the reaction is completed, the reaction solution is filtered, the filtrate is washed with water, EA is extracted, drying and spin-drying are carried out, thus obtaining a light yellow solid, and EA/HEP (1:1) is used for pulping and filtering to obtain 400mg of a white solid. MS-APCI of 600.2[ M + H ]]+Step 10-4:
Figure BDA0002196260460000371
47(340mg), B at room temperature2pin2(173mg),Pd(dppf)Cl2the/DCM (46.5mg) and KOAc (111.7mg) were placed in a reaction flask, degassed under vacuum, and dioxane (5mL) was injected via syringe, and the reaction was heated at 100 ℃ for 4 hours after degassing again. TLC showed 6 small amount remaining. The reaction was extracted with EtOAc and the organic phase was washed with saturated NaCl, dried, spun-dried and chromatographed to give 250mg of a pale yellow solid. MS-APCI 648.2[ M + H ]]+
Step 10-5:
Figure BDA0002196260460000372
48(150mg), 10(95.7mg, WO2018119286), Pd (dppf) Cl2/DCM (18.8mg) and Na were added at room temperature2CO3(48.8mg) was placed in a reaction flask, vacuum degassed, and the dioxane/H was injected via syringe2O (6mL,5:1), degassed again and then heated at 100 ℃ for 3 hours. TLC showed the starting material reaction was complete. The reaction solution was washed with water, extracted with EA, and passed through a column to obtain 150mg of a yellow solid. MS-APCI 854.2[ M + H ]]+
Step 10-6:
Figure BDA0002196260460000373
9(150mg) was dissolved in THF (2mL) at room temperature, and TBAF/THF (0.2mL,1M) was added to react at room temperature for 2 hours. Solid precipitated from the reaction solution. Filtration and cake solids dissolved in MeOH and passed through a cation exchange column afforded 20.7mg of a yellow solid. MS-APCI 616.2[ M + H ]]+
1H NMR(400MHz,DMSO-d6)δ9.33(s,1H),8.87(d,J=2.0Hz,1H),8.46(dd,J=8.2,1.3Hz,1H),8.19(d,J=2.0Hz,1H),8.06(d,J=5.8Hz,1H),7.51(d,J=7.3Hz,1H),7.48–7.40(m,2H),7.39–7.30(m,2H),7.19(d,J=5.8Hz,1H),6.95(dd,J=7.5,1.3Hz,1H),6.68(d,J=7.3Hz,1H),4.85(t,J=5.4Hz,1H),4.73(d,J=4.5Hz,1H),4.23(s,1H),4.06(t,J=5.7Hz,2H),3.82(q,J=13.7Hz,2H),3.65(q,J=5.6Hz,2H),3.57(s,3H),2.75(dd,J=9.7,6.1Hz,1H),2.67(q,J=7.6Hz,1H) 2.39(dd, J ═ 9.6,3.7Hz,1H),2.13(s,3H),2.03(dd, J ═ 13.3,6.8Hz,1H),1.88(s,3H), 1.66-1.49 (m,1H), synthesis of example 11 compound LW1005-011
Figure BDA0002196260460000381
Step 11-1:
Figure BDA0002196260460000382
substrate 51(200mg,1.858 mmol; CN108373476), 2(340mg, 1.858mmol) was added to a three-necked flask containing polyphosphoric acid (8mL), and the reaction was stirred in an oil bath at 140 ℃ for 3 hours and checked by TLC. And after the reaction is completed, adding a sodium hydroxide aqueous solution (2M) into the reaction solution, adjusting the pH value to 6-7, extracting with ethyl acetate, combining organic layers, drying with anhydrous sodium sulfate, and carrying out column chromatography. 370mg of a white solid was obtained in a yield of 76.3%. MS-APCI:305[ M + H ] +; 1H NMR (400MHz, DMSO-d6) δ 9.38(d, J ═ 1.0Hz,1H),8.60(d, J ═ 5.4Hz,1H),8.28(dd, J ═ 5.4,1.0Hz,1H),7.87(dd, J ═ 8.0,1.3Hz,1H),7.77(dd, J ═ 7.8,1.3Hz,1H),7.38(m,1H),2.63(s,3H).
Step 11-2:
Figure BDA0002196260460000383
substrate 53(2.88g,9.44mmol) and m-CPBA (3.83g, 0.0188mol) were dissolved in DCM (40mL), stirred overnight at RT and the reaction checked by TLC. After the reaction is completed, adjusting the pH value to 6-7 by using a sodium hydroxide aqueous solution (2M), extracting by using DCM, drying an organic layer by using anhydrous sodium sulfate, and carrying out column chromatography to obtain a brown yellow solid 2.23g, wherein the yield is 73.5%. MS-APCI 321[ M + H ]]+
Step 11-3:
Figure BDA0002196260460000384
compound 4(2.2g, 6.94 m)mol) was added to a single-necked flask containing acetic anhydride (30mL,0.38mol), and then the reaction solution was heated at 140 ℃ with stirring for 2 hours, followed by TLC detection. After completion of the reaction, the reaction solution was cooled to 80 ℃ and then methanol (12mL) and water (3.6mL) were added to the reaction solution, and stirred at 80 ℃ for 30 minutes. The reaction was cooled to room temperature, a large amount of solid precipitated, the solid was filtered, rinsed with ethyl acetate (10mL x 3), and dried under reduced pressure to give 1.6g of a tea white solid with a yield of 71.8%. MS-APCI 321[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ11.80(s,1H),7.82(d,J=7.6Hz,1H),7.70(d,J=6.6Hz,1H),7.42(t,J=6.4Hz,1H),7.33(t,J=7.8Hz,1H),7.00(d,J=6.9Hz,1H),2.61(s,3H).
Step 11-4:
Figure BDA0002196260460000391
compound 55(1.3g, 4.05mmol),6(2.94g, 8.10mmol) and cesium carbonate (2.6g, 8.10mmol) were added to a single vial of DMF (30mL) and the reaction was stirred at room temperature overnight and checked by TLC. After the reaction was completed, the reaction solution was suspended to dryness, extracted with ethyl acetate/water, dried over anhydrous sodium sulfate, and subjected to column chromatography to obtain a tea white solid 2.3g, with a yield of 94%. MS-APCI [ M + H ]]+1H NMR(400MHz,DMSO-d6)δ7.84(d,J=8.1Hz,1H),7.77(d,J=7.2Hz,1H),7.72(d,J=7.6Hz,1H),7.48–7.32(m,7H),7.28(t,J=7.4Hz,4H),7.06(d,J=7.1Hz,1H),4.25(t,J=4.8Hz,2H),3.94(t,J=4.9Hz,2H),2.62(s,3H).
Step 11-5:
Figure BDA0002196260460000392
compound 56(500mg, 0.83mmol) and TBAF (650mg, 2.5mmol) were added to a single-necked flask containing THF (10mL), and then the reaction was stirred at room temperature for 15 minutes and checked by TLC. After the reaction is completed, the reaction solution is suspended to be dry, extracted by ethyl acetate/water, dried by anhydrous sodium sulfate and subjected to column chromatography to obtain 340mg of tea white solid, and the yield is more than 100 percent (containing TBAF). MS-APCI [ M + H ]]+1H NMR(400MHz,DMSO-d6)δ7.82(d,J=8.0Hz,1H),7.68(dd,J=7.5,5.4Hz,2H),7.33(t,J=7.9Hz,1H),7.02(d,J=7.2Hz,1H),4.90(t,J=5.5Hz,1H),4.10(t,J=5.5Hz,2H),3.70(q,J=5.5Hz,2H),2.60(s,3H).
Step 11-6:
Figure BDA0002196260460000393
mixing compound 57(240mg, 0.657mmol), B2Pin2(333mg, 1.31mmol), potassium acetate (129mg, 1.31mmol) and Pd (dppf)2Cl2DCM (54mg, 0.0657mmol) was added to a single vial charged with 1,4-dioxane (10mL) under nitrogen, and then the reaction was stirred at 100 ℃ for 3 hours and checked by TLC. After the reaction is completed, the reaction liquid is cooled, filtered, mixed with silica gel and subjected to column chromatography to obtain a tea white solid 150mg with the yield of 55.3%. LC-MS 413MS-APCI [ M + H ]]+1H NMR(400MHz,DMSO-d6)δ7.79(dd,J=7.4,1.5Hz,1H),7.74(dd,J=7.7,1.5Hz,1H),7.65(d,J=7.2Hz,1H),7.37(t,J=7.5Hz,1H),7.01(d,J=7.2Hz,1H),4.90(s,1H),4.09(t,J=5.5Hz,2H),3.70(d,J=5.0Hz,2H),2.67(s,3H),1.34(s,12H).
Step 11-7:
Figure BDA0002196260460000394
compound 7(150mg, 0.364mmol), 9(150mg, 0.364mmol), sodium carbonate (265mg, 0.91mmol) and Pd (dppf)2Cl2.DCM (30mg, 0.0364mmol) were added to a single vial charged with 1,4-dioxane (10mL) and water (2mL) under nitrogen, and the reaction was stirred at 100 ℃ for 3 hours and checked by TLC. After the reaction was completed, the reaction solution was cooled, filtered, and subjected to silica gel mixing, column chromatography, and reverse phase column chromatography to obtain a pale new green solid 6 mg. MS-APCI [ M + H ]]+,1H NMR(400MHz,DMSO-d6)δ9.10(s,1H),8.54(s,1H),8.14(d,J=8.0Hz,1H),8.02(d,J=6.0Hz,1H),7.74(d,J=7.7Hz,1H),7.66(d,J=7.1Hz,1H),7.48(t,J=7.7Hz,1H),7.41(t,J=7.8Hz,1H),7.34(d,J=7.4Hz,1H),7.23(d,J=6.0Hz,1H),7.06(d,J=7.0Hz,1H),7.03(d,J=7.2Hz,1H),4.67(br,2H),4.48(br,1H),4.09(t,J=5.6Hz,2H),3.70(t,J=5.6Hz,2H),2.41–2.25(m,4H),2.08(s,3H),2.03–1.95(m,1H),1.92–1.82(m,1H).
EXAMPLE 12 Synthesis of the Compound LW1005-012
Figure BDA0002196260460000401
Step 12-1:
Figure BDA0002196260460000402
compound 54(1g, 1.66mmol) and NCS (221mg, 1.66mmol) were added to a single-necked flask containing DMF (15mL), and the reaction was stirred at 60 ℃ for 45 minutes and checked by TLC. After the reaction was completed, the reaction solution was suspended to dryness, extracted with ethyl acetate/water, dried over anhydrous sodium sulfate, and subjected to column chromatography to obtain 600mg of an oily substance with a yield of 56.6%. LC-MS:637[ M + H ]]+1H NMR(400MHz,Chloroform-d)δ7.72(dd,J=8.1,1.3Hz,1H),7.64(dd,J=7.8,1.3Hz,1H),7.57–7.52(m,4H),7.50(s,1H),7.40–7.27(m,6H),7.19(t,J=7.9Hz,1H),4.23(dd,J=5.4,3.9Hz,2H),4.07(dd,J=5.5,3.9Hz,2H),2.72(s,3H),1.09(s,9H).
Step 12-2:
Figure BDA0002196260460000403
compound 57(680mg, 1066mmol) and TBAF (128mL, 1M THF solution) were added to a single vial of THF (10mL), and the reaction was stirred at room temperature for 1 hour and checked by TLC. After the reaction was completed, the reaction solution was suspended to dryness, extracted with ethyl acetate/water, dried over anhydrous sodium sulfate, and subjected to column chromatography to obtain a tea white solid 230mg with a yield of 54%. LC-MS 399[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ8.02(s,1H),7.84(dd,J=8.0,1.2Hz,1H),7.74(dd,J=7.8,1.2Hz,1H),7.34(t,J=7.9Hz,1H),4.93(t,J=5.6Hz,1H),4.11(t,J=5.4Hz,2H),3.70(q,J=5.5Hz,2H),2.61(s,3H).
Step 11-3:
Figure BDA0002196260460000411
compound 58(230mg, 0.575mmol), B2Pin2(154mg, 0.606mmol), potassium acetate (141mg, 1.43mmol) and Pd (dppf)2Cl2DCM (47mg, 0.0575mmol) was added to a single vial charged with 1,4-dioxane (10mL) under nitrogen, and the reaction was stirred at 110 ℃ for 2h and checked by TLC. After the reaction is completed, the reaction solution is cooled, filtered, mixed with silica gel and subjected to column chromatography to obtain a brown yellow oily substance of 210mg with the yield of 81.7%. LC-MS:447[ M + H]+
Step 12-4:
Figure BDA0002196260460000412
compound 59(210mg, 0.47mmol), 10(194mg, 0.47mmol), sodium carbonate (125mg, 1.18mmol) and Pd (dppf)2Cl2.DCM (38mg, 0.047mmol) were added to a single vial charged with 1,4-dioxane (8mL) and water (2mL) under nitrogen, and the reaction was stirred at 100 ℃ for 3 hours and checked by TLC. After the reaction is completed, cooling the reaction liquid, filtering, mixing a sample with silica gel, performing column chromatography, and then preparing LW1005-066 by using a reverse phase column. LC-MS:653[ M + H]+1H NMR(400MHz,DMSO-d6)δ10.66(s,1H),9.11(s,1H),8.54(s,1H),8.09(s,1H),8.01(d,J=9.8Hz,2H),7.81(dd,J=7.9,1.3Hz,1H),7.50(t,J=7.7Hz,1H),7.43(t,J=7.8Hz,1H),7.37(dd,J=7.6,1.3Hz,1H),7.25(d,J=6.2Hz,1H),7.17–7.04(m,1H),4.78–4.59(m,3H),4.57–4.39(m,2H),4.12(t,J=5.6Hz,2H),3.70(t,J=5.4Hz,2H),3.57(s,1H),3.14(s,1H),2.37–2.26(m,4H),2.07(s,3H),2.04–1.95(m,1H),1.93–1.81(m,1H).
EXAMPLE 13 Synthesis of the Compound LW1005-013
Figure BDA0002196260460000413
Step 13-1:
Figure BDA0002196260460000414
compound 7(1.5g, 2.55mmol) and NCS (400mg, 3.06mmol) were added to a single vial of DMF (15mL), and the reaction was stirred at 60 ℃ for 45 minutes and checked by TLC. After completion of the reaction, the reaction solution was suspended, stirred with ethyl acetate/water (1:1,50mL), filtered and dried to give 1.2g of a white solid with a yield of 75.7%. LC-MS 621[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ8.13(s,1H),8.05(d,J=7.9Hz,1H),7.91(d,J=8.0Hz,1H),7.49–7.26(m,11H),4.26(d,J=5.3Hz,2H),3.95(t,J=4.5Hz,2H),2.80(s,3H),0.98(s,9H).
Step 13-2:
Figure BDA0002196260460000421
compound 60(500mg, 0.804mmol), B2Pin2(408mg, 1.608mmol), potassium acetate (197mg, 2.01mmol) and Pd (dppf)2Cl2.DCM (66mg, 0.0804mmol) were charged into a single vial containing 1,4-dioxane (20mL) under nitrogen, and the reaction was then stirred at 110 ℃ for 4 hours and monitored by LC-MS. After the reaction is completed, the reaction solution is cooled, filtered, mixed with silica gel and subjected to column chromatography to obtain a white solid of 518mg with the yield of 96.3%. LC-MS 669.2[ M + H ] +.
Step 13-3:
Figure BDA0002196260460000422
compound 61(510mg, 0.762mmol), 10(315mg, 0.762mmol), sodium carbonate (242mg, 2.287mmol) and Pd (dppf)2Cl2.DCM (62mg, 0.0762mmol) were added to a single vial charged with 1,4-dioxane (15mL) and water (5mL) under nitrogen, and the reaction was stirred at 100 ℃ for 3 hours and monitored by LC-MS. After the reaction is completed, the reaction solution is cooled, filtered, mixed with silica gel and subjected to column chromatography to obtain a light yellow solid 267mgThe yield thereof was found to be 40%. LC-MS 875[ M + H ]]+.
Step 13-4:
Figure BDA0002196260460000423
compound 62(267mg, 0.305mmol) and TBAF (0.311mL,1M THF solution,0.311mmol) were added to a single vial with THF (3mL), and the reaction was stirred at room temperature for 1 hour and checked by TLC. After the reaction is completed, suspending the reaction solution, mixing the sample, carrying out column chromatography, adding distilled water and methanol into the obtained solid, and freeze-drying to obtain a light yellow solid. LC-MS:637[ M + H ]]+;1H NMR(400MHz,DMSO-d6)δ9.33(s,1H),8.87(d,J=2.0Hz,1H),8.47(d,J=8.1Hz,1H),8.19(s,1H),8.10–8.04(m,3H),7.53(t,J=7.8Hz,1H),7.41(d,J=7.5Hz,1H),7.36(t,J=7.9Hz,1H),7.19(d,J=5.7Hz,1H),6.92(d,J=7.6Hz,1H),4.92(t,J=5.5Hz,1H),4.72(s,1H),4.30–4.17(m,1H),4.12(t,J=5.6Hz,2H),3.82(d,J=10.8Hz,1H),3.68(q,J=5.5Hz,2H),2.79–2.71(m,1H),2.70–2.61(m,1H),2.46–2.35(m,4H),2.10(s,3H),2.06–1.97(m,1H),1.67–1.51(m,1H).
EXAMPLE 14 Synthesis of Compound LW1005-014
Figure BDA0002196260460000431
Step 14-1:
Figure BDA0002196260460000432
compound 1(1g, 4.69mmol) and triethylamine (0.66mL, 4.69mmol) were added to a single-necked flask containing acetonitrile (15mL) and stirred at 0 ℃. Then, a solution of p-ABSA (1.15g, 4.78mmol) in acetonitrile (15mL) was added dropwise to the reaction mixture, and after the addition, the mixture was warmed to room temperature and stirred for 1 hour, followed by TLC detection. After the reaction is completed, the reaction solution is filtered, rinsed by ethyl acetate, the organic layer is suspended and stirred, and column chromatography is carried out to obtain 1.03g of light yellow oily matter with the yield of 91.8%. LC-MS 140[ M-Boc +2H]+1H NMR(400MHz,DMSO-d6)δ3.85(t,J=6.2Hz,2H),2.66(t,J=6.2Hz,2H),1.48(s,9H).
Step 14-2:
Figure BDA0002196260460000433
compound 64(640mg, 2.99mmol) and Rh2(OAc)4(33mg, 0.075mmoL) was put in a single-necked flask containing 1, 2-dichloroethane (5mL), the reaction mixture was stirred under reflux at 81 ℃ and a solution of 2(1g,4.18mmoL) in 1, 2-dichloroethane (10mL) was added dropwise to the reaction mixture, and after the addition, the mixture was stirred under reflux overnight. After the reaction is completed, the reaction solution is suspended to be dry, and is stirred and subjected to column chromatography to obtain 650mg of white solid with the yield of 51.1%. LC-MS:425[ M + H]+1H NMR(400MHz,DMSO-d6)δ11.02(s,1H),8.91(s,1H),7.67(d,J=8.0Hz,1H),7.49–7.44(m,1H),7.21(t,J=7.9Hz,1H),3.76(t,J=6.4Hz,2H),2.64(t,J=6.3Hz,2H),2.44(s,3H),1.46(s,9H).
Step 14-3:
Figure BDA0002196260460000434
triphenylphosphine (123mg, 0.47mmol), elemental iodine (120mg, 0.47mmol) and triethylamine (0.13mL,0.94mmol) were added sequentially to a single vial with DCM (10mL) and stirred at room temperature for 10 min. Then, 66(100mg, 0.235mmol) in DCM (5mL) was added dropwise to the above reaction solution, and after the addition was completed, stirring was continued at room temperature for 2 hours, and the reaction was checked by TLC. After the reaction is completed, the reaction solution is suspended to be dry, silica gel is stirred and subjected to column chromatography to obtain 37mg of white solid with the yield of 38.8 percent. LC-MS 307[ M-Boc +2H]+1H NMR(400MHz,DMSO-d6)δ7.90(d,J=7.7Hz,1H),7.85(d,J=7.9Hz,1H),7.35(t,J=7.8Hz,1H),4.13(t,J=6.4Hz,2H),3.21(t,J=6.4Hz,2H),1.49(s,9H).
Step 14-4:
Figure BDA0002196260460000441
compound 67(290mg, 0.172mmol) and TFA (0.16mL,2.136mmol) were added sequentially to DCM (10mL), stirred at RT for 2h and the reaction monitored by TLC. After the reaction is completed, adding a saturated sodium bicarbonate solution to adjust the pH value to 7-8, extracting with ethyl acetate, drying with anhydrous sodium sulfate, and carrying out column chromatography to obtain a white solid 170mg with a yield of 78%. LC-MS 307[ M + H ]]+;1H NMR(400MHz,DMSO-d6)δ7.89(d,J=7.8Hz,1H),7.82(d,J=8.0Hz,1H),7.58(s,1H),7.33(s,1H),3.55(td,J=7.2,2.6Hz,2H),3.11(t,J=7.2Hz,2H).
Step 14-5:
Figure BDA0002196260460000442
compound 68(100mg, 0.326mmol), O-TBS-bromoethanol (0.14mL,0.651mmol) and NaH (26mg, 0.651mmol) were added sequentially to DMF (5mL) under nitrogen, stirred at room temperature for 2h and monitored by TLC. After the reaction was complete, the solvent was suspended, followed by ethyl acetate/water extraction, drying of the organic layer over anhydrous sodium sulfate, and column chromatography to give 70mg of a white solid with a yield of 46.1%. LC-MS 465[ M + H ] +;
step 14-6:
Figure BDA0002196260460000443
compound 69(70mg, 0.15mmol), 5(104mg, 0.23mmol), sodium carbonate (40mg, 3.015mmol) and Pd (dppf)2Cl2.DCM (12mg, 0.015mmol) were added to a single vial charged with 1,4-dioxane (4mL) and water (1mL) under nitrogen, and the reaction was then stirred at 100 ℃ for 3 hours and monitored by LC-MS. After the reaction is completed, the reaction solution is cooled, filtered, mixed with silica gel and subjected to column chromatography to obtain a light yellow solid 70mg with the yield of 64.9%. LC-MS 719[ M + H ]]+.
Step 14-7:
Figure BDA0002196260460000444
compound 70(110mg, 0.162 mmo)l) and TBAF (0.1mL,0.107mmol) were added to a single vial containing 1,4-dioane (2mL) under nitrogen, and the reaction was stirred at room temperature for 24h and checked by TLC. After the reaction was completed, purification was carried out by reverse phase column preparation, followed by freeze-drying to obtain 1.1mg of a pale yellow solid. LC-MS of 605[ M + H]+;1H NMR(400MHz,DMSO-d6)δ9.32(s,1H),8.88(d,J=2.1Hz,1H),8.46(d,J=8.1Hz,1H),8.20(s,1H),8.07(d,J=5.8Hz,1H),7.93(dd,J=7.8,1.4Hz,1H),7.47(t,J=7.7Hz,1H),7.39–7.28(m,2H),7.19(d,J=5.8Hz,1H),6.91(d,J=7.4Hz,1H),4.73(d,J=5.6Hz,2H),4.24(s,1H),3.89–3.73(m,4H),3.57(q,J=5.8Hz,2H),3.49(t,J=6.5Hz,2H),3.18(t,J=7.2Hz,2H),2.80–2.72(m,1H),2.68(dd,J=3.9,2.0Hz,1H),2.44–2.38(m,1H),2.36(s,3H),2.34–2.32(m,1H),2.08(s,3H),2.05–1.95(m,1H),1.66–1.52(m,1H).
Example 15 Synthesis of Compound LW1005-015
Figure BDA0002196260460000451
Step 15-1:
Figure BDA0002196260460000452
raw material 5(5.0g, WO2018119286) was dissolved in DCM (100mL), imidazole (3.2g) was added, stirring was uniform, TBDPSCl (5.9g) was added, and reaction was carried out at room temperature for 3 hours. After the reaction is finished, washing with saturated salt water, DCM extracting, drying, concentrating and purifying by column chromatography to obtain yellow solid 3.7g
Step 15-2:
Figure BDA0002196260460000453
the raw materials 71(1.97g), 33(3.3g), Pd (dppf) Cl2/DCM (431mg) and Na2CO3(1.12g) were mixed, purged with nitrogen, and Dioxane/H2O (4:1) (50mL) was added to conduct a reaction at 95 ℃ for 3 hours. After the reaction is finished, washing with water, extracting with EA, drying, concentrating, and purifying by column chromatography to obtain 2.7g of yellow solid. MS-APCI:866[ M + H ] +
Step 15-3:
Figure BDA0002196260460000454
compound 72(140mg, 0.162mmol) was added to a single vial containing DCM (15mL) and Dess-Martin Reagent (240mg, 0.567mmol) was added to a plastic tube containing DMF (3mL) with shaking once. Then, the Dess-Martin Reagent/DMF solution is dripped into the compound 1/DCM solution within 1-2 minutes, and the reaction solution is stirred for 10 minutes at room temperature. After completion of the reaction, saturated sodium bicarbonate (5mL) and 10% sodium thiosulfate solution (5mL) were added to the reaction solution, extracted with DCM, dried over anhydrous sodium sulfate, and evaporated to remove a portion of the solvent until about 15mL of the solvent remained. The reaction mixture was used in the next reaction without further purification. MS-APCI:864[ M + H ] +
Step 15-4:
Figure BDA0002196260460000461
3-azetidinecarboxylic acid (23mg, 0.23mmol) was dissolved in AcOH (0.25mL), and the solution was added dropwise to a solution of compound 73 above in DCM, followed by triethylamine (0.3mL), and the reaction was stirred at room temperature for 1 hour. Then, sodium triacetoxyborohydride (49mg, 0.23mmol) was added to the reaction solution, and the reaction solution was stirred at room temperature overnight. Quenching with water, extracting with DCM/water, drying organic layer with anhydrous sodium sulfate, and performing column chromatography to obtain light yellow solid 78mg with two-step yield of 50.7%. LC-MS 949[ M + H ]]+
Step 15-5:
Figure BDA0002196260460000462
compound 74(78mg, 0.082mmol), TBAF (1M in THF,0.1mL, 0.1mmol) was added to a single vial with THF (2mL), and the reaction was stirred at room temperature overnight. After completion of the reaction, the reaction mixture was suspended and water (20mL) was added) Stirring, filtering to obtain residue, and subjecting to reverse phase column to obtain 17.4mg of light yellow solid. LC-MS:711[ M + H]+1H NMR(400MHz,DMSO-d6)δ9.08(s,1H),8.82(s,1H),8.52(s,1H),8.25–8.14(br,1H),8.10(d,J=7.9Hz,1H),8.05(s,1H),7.57(t,J=7.8Hz,1H),7.48–7.38(m,2H),7.25(s,1H),7.05(s,1H),4.79–4.61(m,2H),4.53–4.41(m,1H),4.40–4.19(m,5H),3.71–3.51(m,5H),3.37–3.26(m,2H),2.44(s,3H),2.33(s,1H),2.06(s,3H),2.01(br,1H),1.87(s,1H).
Example 16 Synthesis of Compound LW1005-016
Figure BDA0002196260460000463
Step 16-1:
Figure BDA0002196260460000464
73(300mg, 1.0eq) was added to the reaction flask and 20mL of DCM was dissolved. To the above solution was added a solution of 3-hydroxyacridine hydrochloride (109.9mg, 2eq) in DMSO (0.5mL), followed by dropwise addition of Et3N (35.1mg, 1.0eq) and stirring at room temperature for 1 h. Sodium triacetoxyborohydride (147.2mg, 2.0eq) was added and stirred at room temperature overnight. TLC to monitor the reaction, the reaction liquid is washed by saturated sodium bicarbonate, dried, concentrated and chromatographed to obtain yellow solid 200 mg. MS-APCI of 921[ M + H]+
Step 16-2:
Figure BDA0002196260460000471
75(150mg, 1.0eq) was added to the flask, dissolved in THF, and a TBAF/THF solution (1M in THF, 2mL) was added dropwise and stirred at room temperature for 1 h. TLC to monitor the reaction, concentration, column chromatography, preparation, ion exchange column to obtain 25.60mg of bright yellow solid. MS-APCI 683[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),8.88(d,J=2.0Hz,1H),8.73(s,1H),8.47(d,J=7.7Hz,1H),8.20(s,1H),8.07(d,J=6.7Hz,2H),7.54(t,J=7.9Hz,1H),7.43(d,J=7.5Hz,1H),7.36(t,J=7.9Hz,1H),7.19(d,J=5.8Hz,1H),6.93(d,J=7.5Hz,1H),4.28–4.17(m,1H),4.09–3.98
(m,2H),3.90–3.74(m,1H),3.61–3.50(m,2H),2.78–2.71(m,7H),2.42(s,3H),2.41–2.29(m,2H),2.09(s,3H),2.08–1.95(m,1H),1.65–1.51(m,1H).
EXAMPLE 17 Synthesis of the Compound LW1005-017
Figure BDA0002196260460000472
Step 17-1:
Figure BDA0002196260460000473
(R) -pyrrolidine-3-carboxylic acid (44mg, 0.384mmol) was dissolved in AcOH (0.25mL), and the solution was added dropwise to a solution of compound 73(150mg) in DCM, followed by triethylamine (0.3mL) and the reaction stirred at room temperature for 1 h. Then, sodium triacetoxyborohydride (163mg, 0.768mmol) was added to the reaction solution, and the reaction solution was stirred at room temperature overnight. Quenching with water, extracting with DCM/water, drying organic layer with anhydrous sodium sulfate, and performing column chromatography to obtain light yellow solid 75mg with two-step yield of 40.6%. LC-MS 963[ M + H ]]+
Step 17-2:
Figure BDA0002196260460000481
compound 76(75mg, 0.078mmol), TBAF (1M in THF,0.1mL, 0.1mmol) was added to a single vial of THF (2mL), and the reaction was stirred at room temperature overnight. After completion of the reaction, the reaction mixture was suspended, and water (20mL) was added thereto, followed by stirring, filtration and reverse phase column chromatography to obtain 12.57mg of a pale yellow solid. LC-MS:725[ M + H]+1H NMR(400MHz,DMSO-d6)δ9.06(s,1H),8.86(s,1H),8.52(s,1H),8.22(br,1H),8.13–8.04(m,2H),7.57(t,J=7.7Hz,1H),7.47–7.37(m,2H),7.24(s,1H),7.03(s,1H),4.79–4.61(m,3H),4.53–4.39(m,3H),4.10–3.61(m,7H),3.44–3.38(m,2H),3.34–3.29(m,2H),2.44(s,3H),2.34(s,1H),2.07(s,3H),2.00(br,1H),1.86(s,1H).
EXAMPLE 18 Synthesis of Compounds LW1005-018
Figure BDA0002196260460000482
Step 18-1:
Figure BDA0002196260460000483
8(1.72g of 1.0eq) and Et3N (1.00g of 2.0eq) were added to a reaction flask containing 30mL of DCM and 1mL of DMF, the temperature was lowered to 0 ℃ and BzCl/DCM solution was added dropwise, after the addition was complete, the temperature was raised to room temperature and the mixture was stirred overnight. TLC detection, reaction completion, reaction liquid washed with saturated sodium bicarbonate solution and saturated brine, dried and concentrated. The solid obtained was purified with EA: the PE solution was beaten for 2h at 1:10(200 Ml). And (4) carrying out suction filtration, washing a filter cake by using PE, and drying to obtain 1.39g of white solid. MS-APCI of 453[ M + H ]]+
1H NMR(400MHz,DMSO-d6)δ8.04–7.91(m,4H),7.87(dt,J=8.1,1.7Hz,2H),7.71–7.62(m,1H),7.52(t,J=7.7Hz,2H),7.36(td,J=7.9,3.6Hz,2H),6.95(d,J=7.3Hz,1H),4.60(t,J=5.7,4.1Hz,2H),4.48(t,J=5.0Hz,2H),2.78(s,3H).
Step 18-2:
Figure BDA0002196260460000484
adding 77(200mg) into a reaction flask, protecting with N2, adding 5ml of OCl3, heating to 85 ℃, stirring for 2h, spin-drying excess POCl3, and cooling to-20 ℃. 20mL of ammonia water was diluted to 50mL with ice, added dropwise to the reaction flask, and then warmed to room temperature with white smoke and stirred for 30 min. Extracting with ethyl acetate for 2 times, washing organic phase with brine, drying, concentrating, and performing column chromatography. 67.2mg of a pale yellow solid was obtained. MS-APCI:452[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ9.14(s,2H),8.20(s,1H),8.06–7.89(m,4H),7.72–7.63(m,1H),7.54(t,J=7.7Hz,2H),7.40(t,J=7.9Hz,1H),4.75(t,2H),4.66(t,J=4.8Hz,2H),2.81(s,3H).
Step 18-3:
Figure BDA0002196260460000491
78(200mg 1.0eq), 5(240mg 1.2eq), Na2CO3(120mg 2.5eq) and Pd (dppf)2Cl2(50mg 0.1eq) were charged into a reaction flask, protected with N2, dissolved by adding 1,4-dioxane/H2O (10ml/2ml), heated to 100 ℃ and stirred for 3H. Monitoring the reaction by LC-MS, cooling, washing the reaction solution with water, drying, concentrating, and performing column chromatography. 14.06mg of a yellow solid was obtained. MS-APCI of 602[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),8.87(d,J=2.0Hz,1H),8.46(d,J=8.2Hz,1H),8.34(s,1H),8.20(d,J=1.9Hz,1H),8.10–8.03(m,2H),7.82(d,J=7.4Hz,1H),7.54(t,J=7.7Hz,1H),7.46–7.32(m,2H),7.20(d,J=5.8Hz,1H),7.11(d,J=7.1Hz,1H),6.92(d,J=7.3Hz,1H),4.31(t,2H),4.23(s,1H),3.82(d,J=10.9Hz,2H),3.76(t,2H),2.79–2.61(m,3H),2.45(s,4H),2.38(dd,J=9.8,3.6Hz,2H),2.09(s,3H),2.02(dt,J=14.3,7.2Hz,2H),1.62–1.54(m,1H).
Example 19 Synthesis of Compound LW1005-019
Figure BDA0002196260460000492
Step 19-1:
Figure BDA0002196260460000493
77(1.0g) was added to a reaction flask, protected by N2, POCl3(15mL) was added, the temperature was raised to 100 deg.C, stirred for 2h, excess POCl3 was spin dried, and the temperature was lowered to-20 deg.C. NH (NH)2OH (50% aqueous solution) (1.46g, 10eq) was added dropwise to the flask, with white smoke evolved, and the mixture was warmed to room temperature and stirred for 30 min. TLC monitored the reaction was complete, DCM extracted 2 times (poor solubility), the organic phase was washed with brine, dried and concentrated. To get yellow2g of a colored solid. MS-APCI 468[ M + H [ ]]+
Step 19-2:
Figure BDA0002196260460000494
79(2.0g, 1.0eq) and imidazole (1.45g, 10.0eq) were added to a reaction flask, DMF was added to disperse, TBSCl was added, and stirring was carried out at RT for 1 h. TLC monitoring reaction is finished, EA/water extraction is carried out, organic phase is washed by brine, dried, concentrated and column chromatography is carried out, and golden yellow solid 2.65g is obtained. MS-APCI of 582[ M + H]+
Step 19-3:
Figure BDA0002196260460000501
80(300mg, 1.0eq) was dispersed in methanol, K2CO3(200mg, 3.0eq) was added, and the mixture was stirred at room temperature overnight. TLC monitoring reaction, reaction liquid ethyl acetate/water extraction, water phase back extraction for 2 times, organic phase washing with brine, drying and concentrating, column chromatography to obtain light yellow solid 200 mg. MS-APCI:478[ M + H ]]+
Step 19-4:
Figure BDA0002196260460000502
81(177.5mg 1.0eq), 5(256.2mg 1.5eq), K3PO4(196.9mg 2.5eq) and Pd (PPh3)4(42.8mg 0.1eq) were added to a reaction flask in sequence, protected with N2, dissolved by adding 1,4-dioxane/H2O (10ml/2ml), heated to 100 ℃ and stirred for 3H. TLC monitoring reaction end, cooling, reaction liquid washing, drying, concentrating, column chromatography, golden yellow solid 187.6 mg. MS-APCI:732[ M + H ]]+
Step 19-5:
Figure BDA0002196260460000503
82(72.5mg) was added to the reaction flask, and dissolved in 2mL of methanol,hydrogen chloride methanol solution (4.567mol/L0.4ml) was added dropwise thereto, and the mixture was stirred at room temperature for 30 min. TLC monitored the reaction was complete and the reaction was run straight to yield 23.15mg of a bright yellow solid. MS-APCI 618[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ9.33(s,1H),9.03(s,1H),8.87(d,J=2.0Hz,1H),8.47(d,J=8.2Hz,1H),8.22–8.16(m,2H),8.07(d,J=5.8Hz,1H),7.97–7.90(m,1H),7.45(t,J=7.7Hz,1H),7.39–7.27(m,3H),7.21(dd,J=11.0,6.6Hz,2H),6.91(d,J=7.4Hz,1H),6.12(d,J=7.4Hz,1H),4.85(s,1H),4.74(s,1H),4.22(s,1H),3.89–3.78(m,4H),3.68(d,J=5.3Hz,2H),2.75(dd,J=9.6,6.2Hz,2H),2.37(s,3H),2.09(s,3H),2.02(dt,J=14.5,7.1Hz,2H).
EXAMPLE 20 Synthesis of the Compounds LW1005-020
Figure BDA0002196260460000511
Step 20-1:
Figure BDA0002196260460000512
compound 79(120mg, 0.256mmol),83(48mg, 0.307mmol) and NaH (32mg,0.768mmol) were added to a single vial with DMF (6mL), and the reaction was stirred at room temperature for 3 hours and checked by TLC. After the reaction was completed, the reaction solution was suspended to dryness, extracted with ethyl acetate/water, and the organic layer was dried over anhydrous sodium sulfate and subjected to column chromatography to obtain 70mg of a pale yellow solid. LC-MS of 582[ M + H]+
Step 20-2:
Figure BDA0002196260460000513
compound 84(70mg, 0.12mmol), 5(70mg), potassium phosphate (95mg, 0.45mmol) and Pd (PPh)3)4(17mg, 0.015mmol) was added to a single-neck flask containing 1,4-dioxane (4mL) and water (1mL) under nitrogen, the reaction was stirred at 100 ℃ for 3 hours and the reaction was monitored by LC-MS. After the reaction is completed, cooling the reaction liquid, filtering, and stirring with silica gelAnd performing column chromatography to obtain a light yellow solid 30mg, and then performing reverse phase column chromatography to obtain a light yellow solid 11.5 mg. LC-MS 734[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.69(s,1H),9.15(s,1H),8.97(d,J=2.0Hz,1H),8.91(d,J=2.0Hz,1H),8.58(s,1H),8.39(d,J=2.1Hz,1H),8.01(dd,J=7.9,1.4Hz,1H),7.97–7.89(m,2H),7.54–7.42(m,2H),7.41–7.32(m,2H),7.31–7.27(m,1H),7.21–7.14(m,1H),6.38(d,J=7.3Hz,1H),5.06(s,2H),4.79–4.61(m,2H),4.56–4.40(m,1H),3.93(t,J=5.4Hz,1H),3.65(t,J=5.2Hz,1H),3.57(s,1H),3.43(s,0H),3.33(s,1H),3.15(s,1H),2.38(s,3H),2.35–2.29(m,1H)2.03(s,3H),1.93–1.83(m,1H).
EXAMPLE 21 Synthesis of the Compound LW1005-021
Figure BDA0002196260460000521
Step 21-1:
Figure BDA0002196260460000522
the starting materials 77(18g) and NIS (10.7g) were added sequentially to MeCN (230mL), 5mL of dropwise TFA was added dropwise, and the mixture was stirred in an oil bath at 55 ℃ for 1.5 hours. After the TLC detection reaction, cooling, filtering and drying are carried out to obtain 13.8g of yellow solid. MS-APCI 579[ M + H ]]+.
Step 21-2:
Figure BDA0002196260460000523
compound 85(1.0g,1.73mmol) was added to POCl3(15ml), reacted at 90 ℃ for 1 hour, and concentrated to dryness for further use. Reacting NH2OMe/HCl (10g) was dissolved in 6mL of water, NaOH (4.79g) was added, stirred for 10 minutes, filtered, the filtrate was cooled in an ice bath and poured into the reacted residue, stirred in an ice bath to give a solid, which was filtered off and dried to give crude 1g of solid. MS-APCI of 608[ M + H ]]+.
Step 21-3:
Figure BDA0002196260460000524
compound 86(3g,4.9mmol) was dissolved in MeOH/THF (30mL/30mL), 5N sodium methoxide solution (2.5mL,12.3mmol) was added, stirred at room temperature, and monitored by TLC spot plate. After the reaction, water was added to quench, the solvent was dried, a solid appeared, filtered, and the solid was dried to obtain 2.7g of a pale yellow solid. MS-APCI 504[ M + H ]]+.
Step 21-4:
Figure BDA0002196260460000525
starting material 87(7.8g,15.5mmol), Zn (CN)2(1.09g,9.28mmol),Xantphos-PdCl2(1.17g,1.55mmol),Cs2CO3(7.56g,23.2mmol) in a 250mL three-necked flask, N2The replacement is carried out for 3 times, 150mLDMF is injected, the mixture is stirred for 2 hours at the temperature of 90 ℃, and the reaction is detected to be finished by LCMS. Filtration, concentration of the filtrate to dryness and slurrying of the solid with DCM/MeOH ═ 100mL/2mL gave 4.5g of a pale yellow solid. MS-APCI:403[ M + H ]]+.
Step 21-5:
Figure BDA0002196260460000531
with compound 88(80mg,0.198mmol), 5(76mg,0.2mmol), Pd (dppf)2Cl2(17mg, v) and Na2CO3(53mg,0.5mmol) in a reaction flask, N2Degassing protection, injecting 3mL of dioxane/0.6mL of water into a reaction flask, reacting at 90 ℃ for 2 hours, and completely reacting on a TLC point plate. The reaction solution was washed with water, extracted with ethyl acetate, dried, spun-dried, and passed through a column to give 23mg of a yellow solid. MS-APCI 657[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ9.29(s,1H),8.84(d,J=1.9Hz,1H),8.43(d,J=8.2Hz,1H),8.21(s,1H),8.16(s,1H),8.04(d,J=5.8Hz,1H),7.91(d,J=7.8Hz,1H),7.46(t,J=7.7Hz,1H),7.32(dd,J=11.5,7.6Hz,2H),7.16(d,J=5.8Hz,1H),6.88(d,J=7.5Hz,1H),4.94(s,1H),4.70(s,1H),4.19(s,1H),3.96(t,J=5.1Hz,2H),3.86–3.66(m,7H),2.72(dd,J=9.7,6.1Hz,1H),2.63(q,J=7.6Hz,1H),2.37–2.29(m,4H),2.06(s,3H),1.99(dd,J=13.7,7.0Hz,1H),1.59–1.50(m,1H).
EXAMPLE 22 Synthesis of Compound LW1005-022
Figure BDA0002196260460000532
Charge 77(1.0g) to POCl3(10mL) at 100 ℃ for 1 hour, concentrating to dryness, adding methylamine water solution to the residue, stirring in ice bath to obtain green solid, filtering, and drying to obtain 960mg of solid. MS-APCI 592[ M + H ]]+.
Step 22-2:
Figure BDA0002196260460000533
starting materials 89(960mg) and K2CO3(770mg) MeOH (20mL) was added in turn, stirred at 55 deg.C, monitored by TLC until the reaction was complete, filtered and dried to give 900mg of a green solid. MS-APCI:488[ M + H ]]+.
Step 22-3:
Figure BDA0002196260460000541
mixing the raw materials 90(48mg), ZnCN2(22mg),Pd2(dba)3(11mg), dppf (15mg) was mixed in a 50mL single-necked flask, N2Protecting, adding 5ml DMF, stirring at 95 ℃ for 16 hours, and detecting by TLC to finish the reaction. And filtering the reaction solution, washing the filtrate with water, and extracting with EA. Column chromatography gave 28mg of a green solid. MS-APCI 387[ M + H ]]+.
Step 22-4:
Figure BDA0002196260460000542
compound 1(100mg,0.156mmol), 5(71mg,0.187mmol), Pd (dppf)2Cl2(13mg) andNa2CO3(51mg,0.48mmol) in a reaction flask, N2Degassing protection, injecting 3mL of dioxane/0.6mL of water into a reaction flask, reacting at 90 ℃ for 2 hours, and completely reacting on a TLC point plate. The reaction was washed with water, extracted with EA, dried, spun-dried, and purified on a reverse phase column (DCM: MeOH ═ 10:1) to give 25mg of a yellow solid. MS-APCI 641[ M + H ]]+1H NMR(400MHz,Chloroform-d)δ9.10(s,1H),8.79(s,1H),8.59(d,J=8.3Hz,1H),8.13(d,J=5.9Hz,1H),8.07(d,J=7.7Hz,1H),7.97(s,1H),7.61(s,1H),7.43–7.31(m,2H),6.98(d,J=5.9Hz,1H),6.90(d,J=7.6Hz,1H),4.39(s,1H),4.24(s,2H),3.95(s,2H),3.85(s,2H),3.65(s,3H),2.98–2.89(m,1H),2.77–2.72(m,2H),2.68–2.62(m,2H),2.47(s,3H),2.44–2.38(m,1H),2.26–2.19(m,1H)2.18(s,3H),1.54–1.45(m,1H).
Example 23 Synthesis of Compounds LW1005-023
Figure BDA0002196260460000543
Step 23-1:
Figure BDA0002196260460000551
92(9.1g,1.0 eq; EP2848622) and Cs2CO3(41.56g,3.0eq) were added to a reaction flask, DMF was added to dissolve, 2-bromoethanol (6.03mL,2.0eq) was added dropwise, and stirring was carried out at 50 ℃ for 18 h. And (5) monitoring by LC-MS, and finishing the reaction. DMF was spin-dried and slurried with 100mL of water for 1 h. Suction filtration was carried out and the filter cake was washed with 50mL of water and 100mL of n-heptane. 9.0g of a coffee-colored solid was obtained. MS-APCI of 258[ M + H ]]+
Step 23-2:
Figure BDA0002196260460000552
93(9.0g,1.0eq), 94(13.46g,1.3 eq; Chem,2019,5, 929-. And (5) monitoring by LC-MS, and finishing the reaction. Reaction ofWashing with water, washing with brine, drying, concentrating, and performing column chromatography to obtain solid 10.1 g. MS-APCI 348[ M + H ]]+
Step 23-3:
Figure BDA0002196260460000553
95(5.0g,1.0eq) and Et3N (6mL,3.0eq) were added to dichloromethane, BzCl was added dropwise at RT, and stirring was carried out for 2h at RT. TLC monitored the reaction completion. The reaction solution is washed by saturated sodium bicarbonate solution and saturated brine, dried, concentrated and subjected to column chromatography. 6.4g of a pale yellow to off-white solid was obtained. MS-APCI:452[ M + H ]]+
Step 23-4:
Figure BDA0002196260460000554
96(11.0g,1.0eq) was dissolved in acetonitrile, NIS was added, TFA was added dropwise at RT and after addition, stirring was carried out at 55 ℃ for 3 hours, whereupon a large amount of solid precipitated. TLC monitoring reaction completion, suction filtration, filter cake washing with n-heptane, filtrate spin drying, EA/water extraction, 10% Na2S2O3 solution washing, drying, concentration, and filter cake mixing column chromatography. 13g of an off-white solid was obtained. MS-APCI:578[ M + H ]]+
Step 23-5:
Figure BDA0002196260460000555
97(1.0g) was added to the reaction flask, 5mL of POCl3 was added, and the mixture was stirred at 90 ℃ for 1.5 h. Spin-drying, and pumping to form foam solid. 30% methylamine in water (20mL, 100eq) was added and DCM dissolved and stirred at rt for 15 min. TLC monitors the complete conversion of the raw material, liquid separation, organic phase brine washing, drying, concentration and column chromatography to obtain 1.0g of coffee solid. MS-APCI:591[ M + H ]]+
Step 23-6:
Figure BDA0002196260460000561
98(1.0g 1.0eq) was dispersed in methanol/THF, K2CO3(700mg 3.0eq) was added as a solid and stirred at room temperature for 3 h. TLC showed a large amount of starting material remaining, supplemented with K2CO3(700mg 3.0eq), and stirring was continued overnight at room temperature, with TLC showing essentially complete reaction of starting material. Spin-dry, extract with DCM/water, wash the organic phase with brine, dry, concentrate, and column chromatographe. 700mg of a coffee-colored solid was obtained. MS-APCI:487[ M + H ]]+
Step 23-7:
Figure BDA0002196260460000562
99(2.7g 1.0eq), Zn (CN)2(299.4mg 0.46eq), Cs2CO3(2.17g 1.2eq), Xantphos-PdCl2(418mg 0.1eq) were charged into a reaction flask, protected with N2, dissolved in DMF and stirred at 90 ℃ for 2 h. Monitoring by LC-MS, reaction completion, spin-drying DMF, DCM/water extraction, washing organic phase with brine, drying, concentrating, and column chromatography. 2.2g of a coffee-colored solid was obtained. MS-APCI of 386[ M + H ]]+
Step 23-8:
Figure BDA0002196260460000563
100(300mg 1.0eq), 5(352.5mg 1.2eq), Na2CO3(164.6mg 2.0eq), Pd (dppf)2Cl2(41mg,0.1eq) were charged into a reaction flask, protected with N2, dissolved in 1, 4-dioxane/water and stirred at 90 ℃ for 2 h. And (5) monitoring by LC-MS, and finishing the reaction. Extracting the reaction solution with DCM/water, washing the organic phase with brine, drying, concentrating, and performing column chromatography to obtain a light yellow solid of 40 mg. MS-APCI of 640[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ8.87–8.81(m,1H),8.43(d,J=7.1Hz,1H),8.23–8.13(m,3H),8.07–8.00(m,1H),7.71–7.64(m,1H),7.54–7.48(m,1H),7.41(d,J=9.0Hz,1H),7.31(s,1H),7.24–7.12(m,2H),6.88(s,1H),4.20(s,1H),4.04(d,J=5.2Hz,2H),3.79(q,J=13.5Hz,3H),3.34–3.30(m,3H),2.72(s,1H),2.63(s,2H),2.41–2.32(m,1H),2.20–2.14(m,3H),2.10–2.04(m,3H),1.99(s,1H).
EXAMPLE 24 Synthesis of the Compound LW1005-024
Figure BDA0002196260460000571
Step 24-1:
Figure BDA0002196260460000572
compound 97(1.5g,2.59mmol) was added to POCl3(30ml), react for 2 hours at 90 ℃, and concentrate to dryness for later use. Reacting NH2OMe/HCl (22g) was dissolved in 10mL of water, NaOH (10.36g) was added, stirred for 10 minutes, filtered, the filtrate was cooled in an ice bath and poured into the residue after reaction 1, stirred in an ice bath to give a solid, which was filtered off and dried to give crude 1.5g of solid. MS-APCI 607[ M + H ]]+.
Step 24-2:
Figure BDA0002196260460000573
compound 101(300mg,0.49mmol) was dissolved in MeOH/THF (30mL/30mL), K2CO3(28mg,0.98mmol) was added, stirred at room temperature, and monitored by TLC spot plate. After the reaction was completed, the solvent was spin-dried, and a solid appeared, which was filtered and dried to obtain 200mg of a pale yellow solid. MS-APCI 503[ M + H ]]+.
Step 24-3:
Figure BDA0002196260460000574
starting material 102(135mg,0.268mmol), Zn (CN)2(16mg,0.137mmol),Xantphos-PdCl2(20mg,0.0268mmol),Cs2CO3(131mg,0.402mmol) in a 50mL three-necked flask, N2And (3) replacing for 3 times, injecting 10mLDMF, stirring for 2 hours at 90 ℃, and finishing LC-MS detection reaction. Filtering, concentrating the filtrate to dryness, mixing the sample, and performing column chromatography to obtain 4.5g of light yellow solid. MS-APCI 402[ M + H ]]+.
Step 24-4:
Figure BDA0002196260460000575
compound 103(100mg,0.249mmol), 5(100mg,0.261mmol), Pd (dppf)2Cl2(21mg) and Na2CO3(79mg,0.746mmol) in a reaction flask, N2Degassing protection, injecting 3mL of dioxane/0.6mL of water into a reaction flask, reacting at 90 ℃ for 2 hours, and completely reacting on a TLC point plate. The reaction was washed with water, extracted with EA, dried, spun-dried, and passed through a column (DCM: MeOH ═ 10:1) to afford 11mg of a yellow solid. MS-APCI:656[ M + H]+1H NMR(400MHz,DMSO-d6)δ9.29(s,1H),8.84(d,J=1.9Hz,1H),8.44(d,J=8.1Hz,1H),8.16(s,1H),8.08(s,1H),8.04(d,J=5.8Hz,1H),7.66(d,J=7.8Hz,1H),7.39(t,J=7.7Hz,1H),7.36(s,1H),7.31(t,J=7.9Hz,1H),7.20(d,J=7.5Hz,1H),7.16(d,J=5.8Hz,1H),6.88(d,J=7.4Hz,1H),4.92(s,1H),4.69(s,1H),4.20(s,1H),3.92(t,J=5.2Hz,2H),3.78(d,J=16.2Hz,5H),3.69(t,J=4.3Hz,2H),2.72(dd,J=9.6,6.1Hz,1H),2.63(q,J=7.6Hz,1H),2.36(dd,J=9.7,3.7Hz,1H),2.15(s,3H),2.07(s,3H),2.00(dq,J=13.9,7.3Hz,1H),1.60–1.50(m,1H).
Example 25 Synthesis of the Compound LW1005-025
Figure BDA0002196260460000581
Step 25-1:
Figure BDA0002196260460000582
compound 103(50mg, 0.124mmol) was dissolved in DCM (120mL) and Dess-Martin Reagent (158mg, 0.373mmol) was added to the above solution all at once, stirred at room temperature for 30min and the reaction monitored by TLC. After the reaction was completed, water was added to quench the reaction, and then the reaction solution was washed with saturated sodium bicarbonate, 10% sodium thiosulfate solution and saturated brine in this order, the organic layer was dried over anhydrous sodium sulfate, filtered, and the organic layer was poured into250mL single-necked bottles. To the above solution were added piperidine methyl 4-carboxylate (0.05mL, 0.373mmol), acetic acid (1 drop) and sodium borohydride acetate (79mg, 0.373mmol) in that order, stirred at room temperature for 2 hours and monitored by TLC for reaction. After the reaction is completed, water is added to quench the reaction, then the reaction solution is washed by saturated sodium bicarbonate and saturated saline solution in sequence, an organic layer is dried by anhydrous sodium sulfate, and column chromatography is carried out to obtain 20mg of light yellow solid. MS-APCI 527[ M + H ]]+.
Step 25-2:
Figure BDA0002196260460000583
starting material 104(230mg,0.436mmol) was dissolved in 10mL THF/H2O (2:1), LiOH (21mg, 0.872mmol) was added and the reaction was allowed to react at room temperature for 2 hours and monitored by TLC. After completion of the reaction, TFA (0.08mL,1.135mmol) was added to adjust the solution to neutral, THF was spun off, water was added and the solid precipitated, filtered and dried to 200mg of a pale yellow solid. MS-APCI 513[ M + H ]]+.
Step 25-3:
Figure BDA0002196260460000591
compound 105(200mg), 5(148mg), Pd (dppf)2Cl2(34mg) and Na2CO3(170mg) were placed in a reaction flask, degassed with N2, 3mL of dioxane/0.6mL of water was poured into the flask, the reaction was carried out at 90 ℃ for 2 hours, and the TLC spot plate reaction was complete. The reaction was washed with water, extracted with EA, dried, spun-dried, and passed through a column (DCM: MeOH ═ 10:1) to give 22.2mg of a yellow solid. MS-APCI:767[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ9.29(s,1H),8.84(d,J=1.8Hz,1H),8.43(d,J=8.1Hz,1H),8.16(s,1H),8.12(s,1H),8.04(d,J=5.8Hz,1H),7.66(d,J=7.8Hz,1H),7.39(t,J=7.7Hz,1H),7.35(s,1H),7.31(t,J=7.8Hz,1H),7.19(d,J=7.6Hz,1H),7.16(d,J=5.8Hz,1H),6.88(d,J=7.5Hz,1H),4.71(s,1H),4.20(s,1H),3.93(t,J=6.2Hz,2H),3.87–3.74(m,6H),2.82(d,J=11.0Hz,2H),2.73(dd,J=9.7,6.1Hz,1H),2.62(dt,J=17.7,6.9Hz,3H),2.38(dd,J=9.7,3.5Hz,1H),2.24–2.12(m,4H),2.11–2.02(s,5H),2.01–1.94(m,1H),1.80–1.64(m,2H),1.62–1.39(m,3H).
Example 26 Synthesis of Compounds LW1005-026
Figure BDA0002196260460000592
Step 26-1:
Figure BDA0002196260460000593
compound 97(1.3g) was added to POCl3(15mL), the mixture was heated to 90 ℃ and reacted for 1 hour. TLC point plate raw material is reacted completely. The reaction mixture was poured into a remaining 5mL portion, and the mixture was added dropwise to aqueous ammonia (100mL) through a pipette to precipitate a yellow solid. Filtration and spin-drying of the solid gave 1.0g of product 3 as a dark grey solid. MS-APCI:578[ M + H ]]+
Step 26-2:
Figure BDA0002196260460000594
compound 104(1.0g) was suspended in MeOH (15mL) and K was added at room temperature2CO3(478 mg). After stirring the suspension for 1 hour, LCMS checked reaction completion. And (4) spin-drying the reaction solution. The remaining solid was dissolved in DMF. The DMF solution was poured into water and a solid precipitated. Filtration and drying of the solid gave 600mg of a grey solid. MS-APCI:474[ M + H]+
Step 26-3:
Figure BDA0002196260460000601
starting material 107(300mg), Zn (CN)2(116mg),Xantphos-PdCl2(80mg),Cs2CO3(200mg) in a 50mL three-necked flask, N2And (3) replacing for 3 times, injecting 10mLDMF, stirring for 2 hours at 90 ℃, and finishing LC-MS detection reaction. Filtering, concentrating the filtrate, mixing, and performing column chromatography to obtain 160mg light yellowA colored solid. MS-APCI 402[ M + H ]]+.
Step 26-4:
Figure BDA0002196260460000602
mixing Compound 108(35mg), 5(40mg), Pd (dppf)2Cl2(12mg) and Na2CO3(22mg) in a reaction flask, N2Degassing protection, injecting 3mL of dioxane/0.6mL of water into a reaction flask, reacting at 90 ℃ for 2 hours, and completely reacting on a TLC point plate. The reaction was washed with water, extracted with EA, dried, spun-dried, and passed through a column (DCM: MeOH ═ 10:1) to give 12mg of a yellow solid. MS-APCI 627[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.57-10.1(m,2H),9.74-9.53(m,2H),9.05(s,1H),9.01(s,1H),8.51(s,1H),8.24(s,1H),8.15(dd,J=7.8,1.4Hz,1H),8.08(s,1H),7.61(t,J=7.7Hz,1H),7.50(dd,J=7.6,1.5Hz,1H),7.41(t,J=7.8Hz,1H),7.23(d,J=5.8Hz,1H),7.02(d,J=7.4Hz,1H),5.50(s,1H),5.18(s,1H),4.75-4.58(m,2H),4.51-4.37(m,3H),3.83-3.77(m,3H),3.35-3.25(m,3H),2.43(s,3H),2.07(s,3H),2.03-1.98(m,1H),1.91-1.81(m,2H).
EXAMPLE 27 Synthesis of the Compound LW1005-027
Figure BDA0002196260460000603
Step 27-1:
Figure BDA0002196260460000611
taking raw material 109(6.8g, 1.0 eq; Organic Syntheses,2007,84,262-271) and adding into DMAc (70ml), after dissolving, adding potassium carbonate (9.12g, 2.0eq), dropwise adding methyl iodide (7g, 1.5eq) and reacting at room temperature overnight; and (3) treatment: water (50ml), ammonia (20ml), EA (100ml x 3) was added for extraction, column: elution with ethyl acetate/n-heptane 15/85 gave 6.05g of a yellow solid (yield: 83.4%). LCMS found 221[ M + H ]]+1H NMR(400MHz,CDCl3)δ7.94(d,J=6.1Hz,1H),7.39(d,J=0.9Hz,1H),7.26(s,1H),6.97–6.83(m,1H),4.11(s,3H),4.04(s,3H),3.91(s,3H).
Step 27-2:
Figure BDA0002196260460000612
adding raw material 110(5g, 1.0eq) into acetic acid (82g, 30eq), adding 47% aqueous solution of hydrobromic acid (55g, 30eq), dissolving, heating to 95 deg.C, and reacting for 2 hr; and (3) treatment: concentration to give 9g (yield: quantitative) of pale yellow solid, which was directly fed to the next step. LCMS found 193[ M + H ]]+
Step 27-3:
Figure BDA0002196260460000613
adding 111(4.36g, 1.0eq) of raw material into 90ml of methanol for insolubilization, adding 5g of concentrated sulfuric acid (1.0 eq), and carrying out reflux reaction at 85 ℃ in an external bath overnight; and (3) treatment: under ice bath, 70ml of saturated sodium bicarbonate is added dropwise, the pH value is 7-8, a solid is precipitated, the solid is filtered, rinsed with water and dried to obtain 4.43g of a white solid (yield: 94.8%). LCMS found 207[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ11.02(s,1H),7.24(s,1H),7.20(t,J=6.5Hz,1H),6.58(d,J=7.4Hz,1H),3.93(s,3H),3.81(s,3H).
Step 27-4:
Figure BDA0002196260460000614
adding a raw material 112(4.2g, 1.0eq) into DMAc (50ml), under the protection of argon, adding cesium carbonate (13.3g, 2.0eq), adding a silanol raw material 8(9.62g, 1.3eq), and reacting at room temperature overnight; and (3) treatment: adding water (100ml) and 1M hydrochloric acid (80ml) to adjust the pH value to 5-6; EA extraction 3 times, column chromatography: 20% EA/n-heptane rinse, concentrate to give 4.5g of white solid, yield: 45.4 percent. LCMS found 489[ M + H ] +; 1H NMR (400MHz, DMSO-d6) δ 7.54(d, J ═ 7.5Hz,1H),7.45(d, J ═ 7.3Hz,4H),7.39(t, J ═ 7.5Hz,2H), 7.32-7.23 (m,5H),6.66(d, J ═ 7.5Hz,1H),4.13(t, J ═ 5.0Hz,2H),3.95(s,3H),3.86(t, J ═ 5.0Hz,2H),3.83(s,3H),0.93(s,9H).
Step 27-5:
Figure BDA0002196260460000621
dissolving compound 113(300mg) and compound 114(149mg) in 15mL of THF under nitrogen protection, adding 2.5mL of NaHMDS (2M) dropwise in a salt bath, and reacting for 20min under heat preservation after dropwise addition; and detecting the reaction by LC-MS. After the reaction is completed, water is added for quenching, EA extraction is carried out, organic phases are combined, brine is used for washing, drying is carried out, and 160mg of light yellow solid is obtained after rotary dry mixing and column chromatography. LC-MS found:699[ M + H ] +
Step 27-6:
Figure BDA0002196260460000622
compound 115(160mg) was dissolved in 2ml of methanol, 100mg of 10% Pd/C was added, and after the addition, hydrogen substitution was performed, reaction was performed at room temperature for 30min, and the reaction was checked by TLC. After the reaction is completed, adding diatomite and filtering, taking filtrate and spin-drying to obtain 140mg of gray solid crude product, and directly putting the crude product into the next step. LC-MS found:669[ M + H ] +
Step 27-7:
Figure BDA0002196260460000623
dissolving compound 113(55mg) and compound 116(75mg) in 3.8mL THF, nitrogen protection, and ice salt bath, adding 0.45mL NaHMDS (2M) dropwise, after adding dropwise, keeping the temperature for 1h, and detecting the reaction by LC-MS.
After the reaction is completed, water is added for quenching, EA extraction is carried out, organic phases are combined, salt washing and drying are carried out, and 53mg of yellow solid is obtained after rotary dry mixing and column chromatography. LC-MS found 1125[ M + H ]]+
Step 27-8:
Figure BDA0002196260460000624
compound 117(53mg) was dissolved in 2.5ml of THF, 2.5ml of triethylamine trihydrofluoride salt was added, and after the addition was completed, the reaction was carried out at room temperature for 2 hours, and the reaction was checked by TLC. After completion of the reaction, THF was removed by rotary evaporation at low temperature, and the resulting residue was concentrated to give 6.1mg of a white solid by means of a reverse phase column. LC-MS found 1125[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ9.85(s,1H),7.55(s,1H),7.43(d,J=7.5Hz,1H),7.39(d,J=7.9Hz,1H),7.30(t,J=7.8Hz,1H),7.02(d,J=7.4Hz,1H),6.62(d,J=7.5Hz,1H),4.00(t,J=5.7Hz,2H),3.95(s,3H),3.62(t,J=5.6Hz,2H),1.96(s,4H),1.24(s,1H).
Example 28 Synthesis of Compound LW1005-028
Figure BDA0002196260460000631
Step 28-1:
Figure BDA0002196260460000632
118(7.6g,51.0mmol,1.0eq) is added into a reaction bottle in turn, anhydrous THF is steamed, and inert gas is used for protection; cooling the dry ice-acetone to-78 ℃; n-BuLi (30.5mL,76.5mmol,1.5eq,2.5M) was added dropwise; stirring for 30min at low temperature; introducing carbon dioxide dried by concentrated sulfuric acid and anhydrous calcium chloride into the reaction solution; reacting for 3 hours, and slowly heating to 0 ℃ below zero; adding saturated solution of ammonium chloride for quenching; adding water and ethyl acetate; separating out an organic phase; the aqueous phase was washed once with ethyl acetate; adjusting the pH value of the water phase to 2; a large amount of solid was precipitated, filtered and dried to obtain 9.4g of solid with a yield of 95.5%. LCMS found:194[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ8.15(d,J=6.0Hz,1H),7.60(s,1H),7.39(dd,J=6.0,0.9Hz,1H),4.02(s,3H).
Step 28-2:
Figure BDA0002196260460000633
compound 119(9.0g, 46.6mmol, 1.0eq), ethanol (270mL) and concentrated sulfuric acid (11.5mL) were added to a reaction flask and heated to reflux; stirring overnight, evaporating ethanol, cooling the residue with ice water bath, and adding water dropwise to precipitate solid; filtration and drying gave 7.4g of solid in 76.7% yield. LCMS found 208[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ11.68(s,1H),7.61(d,J=2.3Hz,1H),7.50(d,J=7.5Hz,1H),6.73(d,J=7.4Hz,1H),4.33(q,J=7.1Hz,2H),1.31(td,J=7.1,2.9Hz,3H).
Step 28-3:
Figure BDA0002196260460000634
120(7.0g, 33.8mmol, 1.0eq), 70mL DMAc, cesium carbonate (22.0g,67.5mmol,2.0eq) and compound 5(14.7g, 40.5mmol, 1.2eq) were added to a reaction flask, stirred at room temperature for 2h, water and ethyl acetate were added, ethyl acetate was separated, the aqueous phase was extracted once with ethyl acetate, the organic phases were combined, washed with saturated sodium chloride, and spin-dried to give compound 121, which was directly fed to the next step without purification. LCMS: found 490[ M + H [ ]]+
Step 28-4:
Figure BDA0002196260460000641
crude compound 121 from the previous step, ethanol (70mL) and water (18mL) were added to the reaction flask and LiOH solid (1.22g, 20.9mmol, 1.5eq) was added. Stirring at room temperature for 0.5h, concentrating ethanol, and adding water and ethyl acetate to the residue; separating out ethyl acetate; water phase ice water bath, adjusting pH to 5, and separating out a large amount of solid; filtration and drying gave 10.3g of solid in 66.1% yield (two steps). LCMS found:462[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ7.82(d,J=7.5Hz,1H),7.54(d,J=0.9Hz,1H),7.48–7.34(m,7H),7.29(t,J=7.3Hz,4H),6.80(dd,J=7.5,0.9Hz,1H),4.19(t,J=4.9Hz,2H),3.88(t,J=4.9Hz,2H),0.92(s,9H).
Step 28-5:
Figure BDA0002196260460000642
substrate 123(600 mg; WO2018119286),124(1g), TsOH (365mg) was added to isopropanol (10mL) and stirred at 85 ℃ overnight. TLC detection of the disappearance of starting material, spin drying of the reaction, water washing, EtOAc extraction, column chromatography to yield 350mg of a pale yellow solid. LC-MS 540[ M + H ]]+
Step 28-6:
Figure BDA0002196260460000643
125(190mg) was added to 4N HCl/dioxane (1ml) at room temperature, and stirred at room temperature for 30 minutes. TLC detection raw material reaction is complete, reaction liquid is spin-dried, and 190mg of gray crude solid is obtained and is directly used for the next reaction. LC-MS 440[ M + H]+
Step 28-7:
Figure BDA0002196260460000644
126(150mg), 122(102mg), and HATU (108mg) were dissolved in DMF (1ml) at room temperature, followed by addition of DIEA (85mg), and the reaction was continued at room temperature overnight. The reaction was checked by TLC. After completion of the reaction, the reaction solution was washed with water, extracted with EtOAc, and the organic phase was washed with saturated NaCl, dried, spin-dried, and passed through a column to obtain 70mg of a yellow solid. LC-MS 883[ M + H ] +
Step 28-8:
Figure BDA0002196260460000645
127(70mg) was dissolved in THF (0.5mL) at room temperature, and 0.1mL of TBAF/THF (1N) was added thereto and stirred for 1 hour. TLC detection raw material disappeared, the reaction liquid was directly passed through the column, and the crude compound obtained was passed through the cation exchange resin column to obtain 7.4mg of a yellow solid. LC-MS 645[ M + H ] +; 1H NMR (400MHz, DMSO-d6) δ 10.10(s,1H),9.31(s,1H),8.87(d, J ═ 2.0Hz,1H),8.43(d, J ═ 8.2Hz,1H),8.19(s,1H),8.06(d, J ═ 5.7Hz,1H),7.78(s,1H),7.73(d, J ═ 7.5Hz,1H),7.42(d, J ═ 7.9Hz,1H),7.33(t, J ═ 7.5Hz,2H),7.18(d, J ═ 5.8Hz,1H),7.09(d, J ═ 7.4Hz,1H),6.88(d, J ═ 7.5Hz,1H),6.79(d, J ═ 4H, 7.79, 4H), 3.3.7.7, 3H, 3, 6.0Hz,1H),2.67(d, J ═ 7.0Hz,1H), 2.44-2.37 (m,1H),2.09(s,3H),1.99(m,4H),1.59(m,1H),1.24(s,2H).
EXAMPLE 29 Synthesis of the Compound LW1005-029
Figure BDA0002196260460000651
Step 29-1:
Figure BDA0002196260460000652
126(120mg), 113(86.5mg), was dissolved in THF (2mL) under ice-bath, NaHMDS (0.7mL) was added under protection of N2, and the reaction was continued for 1 hour in ice-bath. TLC shows that the raw material reaction is complete, saturated NH4Cl is added to quench the reaction, EA extraction is carried out, and the obtained product passes through a column to obtain LW1005-011 and 380mg of yellow solid. LC-MS 896[ M + H ]]+
Step 29-2:
Figure BDA0002196260460000653
substrate 128(90mg) was added to a mixture of THF and TEA/3HF (2mL,1:1) and reacted for 2 hours. TLC detected the starting material disappeared, the reaction was adjusted to pH 9 with saturated NaHCO3, extracted with EtOAc, spun dried and isolated to afford 12.4mg of a yellow solid. LC-MS 658[ M + H ]]+;1H NMR(400MHz,DMSO-d6)δ10.98(d,J=94.5Hz,1H),9.88(s,1H),9.09(s,1H),8.52(s,1H),8.18(d,J=8.3Hz,1H),8.05(d,J=5.9Hz,1H),7.56(s,1H),7.38(ddd,J=30.9,13.6,7.3Hz,4H),7.26–7.17(m,1H),7.08(t,J=6.5Hz,1H),6.97(d,J=7.6Hz,1H),6.62(d,J=7.4Hz,1H),4.67(s,2H),4.47(s,1H),4.01(s,2H),3.95(s,3H),3.71–3.60(m,4H),2.93(m,1H),2.34-2.32(s,1H),2.07(s,3H),2.00(s,3H),1.95-1.85(m,1H),1.21-1.15(m,3H).
Example 30 Synthesis of Compound LW1005-030
Figure BDA0002196260460000661
Step 30-1:
Figure BDA0002196260460000662
adding compound 128(1g,1 eq; WO2012154213), CuI (0.03g,0.05eq), palladium (0.12g,0.05eq) bis (triphenylphosphine) chloride into a 50ml reaction flask, replacing with nitrogen for 3 times, adding anhydrous DMF (20ml), and leaving the system to be reddish brown and clear; triethylamine (1.7g,5.0eq) was added, the system was clear brown yellow, nitrogen was substituted 3 times, and light was avoided, and alkyne starting material 3(0.52g,1.1eq) was added, reacted at room temperature, and the reaction was checked by TLC. After the reaction was completed, the reaction solution was poured into an aqueous solution of ice-cold ammonium chloride, extraction was performed 2 times with MTBE, the organic layer was washed 5 times with saturated sodium chloride, and column chromatography was performed to obtain 1.03g of an oily substance. LC-MS 309[ M + H ]]+1H NMR(400MHz,Chloroform-d)δ7.49(dd,J=8.0,1.4Hz,1H),7.40(dd,J=7.7,1.3Hz,1H),6.97(t,J=7.8Hz,1H),2.57(s,3H),1.06(t,J=7.9Hz,9H),0.70(q,J=7.8Hz,6H).
Step 30-2:
Figure BDA0002196260460000663
potassium phosphate (13g,2eq), palladium acetate (0.75g,0.03eq) were charged in a 500ml reaction flask, nitrogen gas was substituted 3 times, 130(10.5g,1.3 eq; LabNetwork) dioxane solution (75ml) was added, compound 129(9.5g,1eq) dioxane solution (75ml) was added, water (30ml) was added, the reaction was carried out for 2 hours in an external bath at 100 ℃ and the system was blackish brown, and TCL tracing reaction was completed. After the reaction was completed, water was added to quench, MTBE was extracted, the organic layer was washed with saturated brine, concentrated, and column chromatography was performed to obtain 9.8g of a liquid. LC-MS 366[ M + H]+1H NMR(400MHz,Chloroform-d)δ7.86(dd,J=7.8,1.7Hz,1H),7.55(dd,J=7.7,1.4Hz,1H),7.40–7.32(m,2H),7.23(t,J=7.7Hz,1H),7.06(dd,J=7.6,1.4Hz,1H),2.22(s,3H),2.19(s,3H),1.09(t,J=7.9Hz,9H),0.72(q,J=7.9Hz,6H).
Step 30-3:
Figure BDA0002196260460000664
the starting material 131(10.17g,1eq) was added to THF (50ml) to clear the system brown-yellow, and TBAF (17.55g,2eq) in THF (50ml) was added dropwise and reacted at room temperature for 30 minutes. After the reaction was complete, the column was directly packed with a sample to obtain 6.62g of a solid. LC-MS:252[ M + H ]]+1H NMR(400MHz,Chloroform-d)δ7.54(dd,J=7.8,1.4Hz,1H),7.40–7.32(m,2H),7.22(t,J=7.6Hz,1H),7.08(dd,J=7.7,1.4Hz,1H),3.32(s,1H),2.19(s,3H),2.16(s,3H).
Step 30-4:
Figure BDA0002196260460000671
compound 132(4.6g,1eq), 133(6.57g,1.15 eq; WO2014063199), palladium bistriphenylphosphine chloride (0.64g,0.05eq), CuI (0.15g,0.05eq), DIPEA (11.83g,5eq) were charged into a reaction flask, and the mixture was replaced with nitrogen for 3 times, DMF was added, and the mixture was replaced with nitrogen, and the system was clear in reddish brown color, and then reacted at 55 ℃ for 3 hours in an external bath. After the reaction was completed, 1000ml of water was added in ice bath, 500 × 2 times of EA extraction was performed, 2 times of saturated brine washing was performed, and column chromatography was performed to obtain 6.7 g. LCMS found 435[ M ]]+1H NMR(400MHz,Chloroform-d)δ8.26(d,J=1.5Hz,1H),8.05(d,J=1.5Hz,1H),7.91–7.83(m,2H),7.46–7.36(m,3H),7.19(dd,J=7.5,1.4Hz,1H),7.00(s,1H),3.96(s,3H),2.26(s,3H),2.24(s,3H).
Step 30-5:
Figure BDA0002196260460000672
compound 134(6.76g,1eq) was added to methanol (250ml) and isopropyl acetate (600ml) followed by zinc dust (10ml)g,10eq), ammonium chloride (8.3g,10eq), stirred at room temperature for 1 hour. Filtering, rinsing the solid with isopropyl acetate, adding EA into the solid, pulping, filtering, concentrating the filtrate, and performing column chromatography to obtain 1.6g of solid. LCMS found406:408 ═ 3:1[ M + H]+1H NMR(400MHz,DMSO-d6)δ8.27(d,J=1.5Hz,1H),7.92(d,J=1.5Hz,1H),7.85–7.69(m,1H),7.41(d,J=7.2Hz,2H),7.26–7.12(m,1H),6.96(t,J=7.7Hz,1H),6.68(d,J=7.9Hz,1H),6.34(d,J=7.4Hz,1H),4.92(s,2H),3.89(s,3H),2.19(s,3H),1.77(s,3H).
Step 30-6:
Figure BDA0002196260460000673
compound 135(500mg,1eq), palladium acetate (30mg, 0.1eq), BINAP (100mg,0.2eq), and cesium carbonate (1000mg,2.5eq) were charged in a 50ml reaction flask, and replaced with nitrogen 3 times, and a toluene solution (20ml) of chlorine starting material 123(700mg) was added, replaced with nitrogen, and reacted overnight at 100 ℃ in an external bath. After the reaction was complete, filtration and EA rinse were carried out and the filtrate was concentrated through the column to give 800mg of a yellow solid. LCMS found 633[ M + H ]]+
Step 30-7:
Figure BDA0002196260460000681
compound 136(550mg) was added to THF (5ml) and methanol (4ml), and sodium borohydride (100mg), lithium chloride (100mg) were added under ice-bath to react at room temperature overnight. After the reaction is completed, ammonium chloride aqueous solution is added under ice bath, EA is extracted for 2 times, and the mixture is concentrated and is subjected to column chromatography to obtain 450mg of yellow solid. LCMS found 605[ M + H ]]+
Step 30-8:
Figure BDA0002196260460000682
compound 137(420mg) was added to DCM (8ml), and activated manganese dioxide (430mg) was added to conduct a reaction under reflux overnight. After the reaction was complete, celite was added, the mixture was filtered, DCM was rinsed, the filtrate was concentrated, and the residue was filtered through a column to give 320mg of a yellow solid. LCMS found 603[ M + H ] +
Step 30-9:
Figure BDA0002196260460000683
compound 138(290mg) was added to DCM (9mL), followed by addition of a solution of R-3-carboxypyrrolidine (114mg) in AcOH (0.25mL) and triethylamine (0.4mL), reaction at room temperature for 1 hour, addition of sodium borohydride acetate (600mg), and stirring at room temperature overnight. After the reaction is completed, adding saturated ammonium chloride to quench the reaction, separating liquid, extracting once by DCM, drying by organic phase sodium sulfate, and passing through a column to obtain 100mg of light yellow solid. LCMS found 702[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ12.90(s,1H),10.57(s,1H),10.13(s,1H),9.05(s,1H),8.50(s,1H),8.23(s,1H),8.07(d,J=6.0Hz,1H),7.84(d,J=1.5Hz,1H),7.83(s,1H),7.67(d,J=1.5Hz,1H),7.49(t,J=7.7Hz,1H),7.43(s,1H),7.38(t,J=7.8Hz,1H),7.31(d,J=7.5Hz,1H),7.22(d,J=5.9Hz,1H),7.01(d,J=7.4Hz,1H),5.49(s,1H),4.61~4.65(m,2H),4.41~4.55(m,2H),2.27(s,3H),2.08(s,3H),1.29(d,J=46.4Hz,1H).
Synthesis of the Compounds LW1005-031 of example 31
Figure BDA0002196260460000691
Step 31-1:
Figure BDA0002196260460000692
compound 135(510mg,1eq), 122(500mg,1.05eq), HATU (825mg,2eq), DIEA (295mg,2.1eq) was added to a reaction flask, DMF (25ml) was added, clear, stirred at room temperature overnight; and (3) post-treatment: water (30ml) was added, EA was extracted 2 times, and the mixture was washed with saturated brine 1 time, and then the mixture was subjected to column chromatography to give 1.37g of a yellow solid. LCMS found849[ M + H ] +
Step 31-2:
Figure BDA0002196260460000693
adding 139(910mg,1eq) of raw material into freshly distilled THF (10ml), adding lithium borohydride (100mg,5eq) under ice bath, reacting at room temperature for 6 hours, and tracking by TLC; after the reaction was completed, an aqueous solution of ammonium chloride was added in an ice bath, EA was extracted 3 times, washed with saturated saline 1 time, concentrated, and passed through a column to obtain 690mg of a yellow solid. LCMS found:821[ M + H ]]+
Step 31-3:
Figure BDA0002196260460000694
compound 140(490mg,1eq) was added to DCM (45ml) and was slightly cloudy, DMP (380mg,1.5eq) was added and the reaction was run for 40 min at RT followed by TLC. After the reaction is completed, adding saturated sodium sulfite, stirring for 5 minutes, adding saturated sodium bicarbonate, and stirring for 5 minutes; DCM was extracted 2 times, washed 1 time with saturated brine, and the column was packed with a sample to give 319mg of a yellow solid. LCMS found 819[ M + H ]]+
Step 31-4:
Figure BDA0002196260460000701
after adding compound 141(100mg,1eq), a solution of R-3-carboxypyrrolidine (41mg) in AcOH (0.25mL) and triethylamine (0.4mL) in this order to DCM (5mL) and reacting at room temperature for 1 hour, sodium borohydride acetate (120mg) was added and reacted at room temperature overnight, and the reaction was checked by TCL. After the reaction is safe, saturated ammonium chloride is added, liquid separation is carried out, DCM is carried out for 2 times, and the mixture is mixed with a sample and concentrated to pass through a column to obtain 130mg of white solid. LCMS found 918[ M + H ]]+
Step 31-5:
Figure BDA0002196260460000702
compound 142(140mg) was dissolved in THF (4mL), a 1M solution of TBAF/THF (0.2mL) was added to the solution, the reaction was carried out at room temperature for 30 minutes, and the reaction was checked by TCL. Reaction ofAfter completion, the sample was directly applied to the column to give 80mg of a yellow solid. LCMS found 680[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.14(s,1H),7.84(d,J=7.9Hz,1H),7.78(s,1H),7.74(d,J=7.5Hz,1H),7.62(s,1H),7.49(t,J=7.7Hz,1H),7.46–7.39(m,2H),7.35(t,J=7.6Hz,1H),7.28(d,J=7.5Hz,1H),7.11(d,J=7.5Hz,1H),6.79(d,J=7.4Hz,1H),4.89(s,1H),4.40(s,1H),4.06(t,J=5.7Hz,2H),3.66(d,J=5.4Hz,2H),2.23(s,3H),1.97(s,3H).
EXAMPLE 32 Synthesis of the Compound LW1005-032
Figure BDA0002196260460000703
Step 32-1:
Figure BDA0002196260460000704
compound 143(5.6g, 300mmol), K2CO3(5.5g, 40mmol) and 123(5.2g, 20mmol) were added sequentially to a single vial of DMF (100mL) and then stirred in an oil bath at 120 ℃ overnight and the reaction checked by TLC. After the reaction is completed, the organic solvent is suspended, stirred and washed by adding water, filtered, dried, mixed with a sample and subjected to column chromatography to obtain 3.1g of brown solid. LCMS found 414[ M + H ]]+
Step 32-2:
Figure BDA0002196260460000711
raw material 144(1g,2.4mmol), bis-pinacolato borate (711mg,2.8mmol), PdCl2(dppf) DCM (195mg,0.24mmol), KOAc (470mg, 4.8mmol) were mixed in a 25mL single vial, displaced 3 times with N2, injected with 10mL of Dioxide, stirred at 95 ℃ for 16 hours, washed with water, extracted, concentrated, purified over column to give 850mg of a solid. LCMS found:462[ M + H ]]+
Step 32-3:
Figure BDA0002196260460000712
raw material 145(248mg,0.54mmol), raw material 33(100mg,0.27mmol), PdCl2(dppf)2DCM (22mg,0.027mmol), Na2CO3(57mg,0.54mmol) were mixed, nitrogen was replaced, Dioxane/H2O (8mL) was added, the reaction was carried out at 95 ℃ for 3 hours, and water washing was carried out to obtain 47mg of a white solid. LCMS found 629[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ9.19(d,J=2.4Hz,1H),8.72(s,1H),8.65–8.58(m,1H),8.16–8.06(m,2H),7.63(d,J=5.7Hz,1H),7.56(t,J=7.8Hz,1H),7.50–7.38(m,2H),7.27(d,J=8.0Hz,1H),7.14(d,J=7.4Hz,1H),5.60–5.44(m,1H),4.79–4.64(m,2H),4.47(dd,J=31.1,4.5Hz,2H),4.16(t,J=5.4Hz,2H),3.70(d,J=5.2Hz,2H),3.32(s,2H),3.21–3.10(m,1H),2.44(s,3H),2.35–2.29(m,1H),2.05–1.97(m,1H),1.93–1.84(m,1H),1.82(s,3H).
EXAMPLE 33 Synthesis of Compound LW1005-033
Figure BDA0002196260460000713
Step 33-1:
Figure BDA0002196260460000714
compound 146(1.0g) was dissolved in DCM (30mL), 147(1.16g), TEA (1.32g) were added, and after 1 hour of reaction at RT NaBH (OAc)3(3.31g) was added in portions. The reaction was 1 hour and the TLC spot plate reaction was complete. The reaction solution was washed with water, extracted with DCM, spin-dried and passed through a column to give 1.0g of a yellow solid product. LCMS found 306[ M + H ]]+
Step 33-2:
Figure BDA0002196260460000721
compound 148(1.0g), 149(0.92g) and TsOH (0.75g) were added to isopropanol (30mL) and reacted at 85 ℃ overnight. TLC showed the reaction was complete. Adding water into the reaction solution, removing isopropanol by rotation, and extracting impurities by EA. Adding NaHCO3 aqueous solution into water phase, adjusting pH to 7, precipitating solid,extraction with DCM, dissolution of the solid and spin-drying of DCM gave 1.24g of a yellow solid. LCMS found:455[ M + H [ ]]+
Step 33-3:
Figure BDA0002196260460000722
compound 33(1g), bis-pinacolato borate (815mg), PdCl2(dppf), DCM (180mg), and KOAc (420mg) were mixed in a 25mL single-necked flask, and N2 was substituted 3 times, 10mL of Dioxide was injected, and the mixture was stirred at 95 ℃ for 16 hours, washed with water, extracted, concentrated, and purified by column chromatography to obtain 900mg of a solid. LCMS found 422[ M + H ]]+
Step 33-3:
Figure BDA0002196260460000723
150(119mg), 151(100mg), Pd (dppf) Cl2/DCM (19.4mg) and Na2CO3(50mg) were placed in a reaction flask at room temperature, degassed under vacuum, and the reaction was heated at 100 ℃ for 4 hours after syringe injection of dioxane/H2O (6mL,5:1) and degassing again. TLC monitored the starting material reaction was complete. The reaction was washed with water and passed through a column (DCM: MeOH ═ 20:1) to give 50mg of crude product, which was isolated as 20mg of product 5 as a yellow solid. LCMS found 670[ M + H ]]+
Step 33-4:
Figure BDA0002196260460000724
compound 152(10mg) was dissolved in THF/H2O (1.3mL,1:0.3), and LiOH/H2O (1.3mg) was added to the solution to react at room temperature for 1 hour, after which TLC starting material was completely reacted. The reaction solution was prepared and separated to obtain 5mg of a yellow solid product. LCMS found:656[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.36(s,1H),9.07(s,1H),8.73(s,1H),8.50(s,1H),8.15(s,1H),8.09(d,J=7.7Hz,1H),8.03(s,1H),7.56(t,J=7.8Hz,18H),7.42(dd,J=7.9,4.9Hz,2H),7.29–7.19(m,2H),7.08(s,1H),5.33(t,J=4.8Hz,1H),4.68(s,2H),4.16(s,2H),3.69(t,J=5.3Hz,2H),3.19–3.00(m,2H),2.44(s,3H),2.28–2.15(m,2H),2.05(s,3H),2.03–1.91(m,2H),1.49-1.43(m,1H).
Example 34 Synthesis of Compound LW1005-034
Figure BDA0002196260460000731
Step 34-1:
Figure BDA0002196260460000732
compound 43(8g) was dissolved in acetic acid (100mL), cooled in an ice bath, and compound 2,2, 2-trichloroacetimidate methyl ester (16mL) was added dropwise thereto, followed by TLC detection overnight at room temperature. After the reaction was completed, the reaction mixture was poured into ice water, extracted with EA, washed with saturated brine, dried, and applied to a column to obtain 10.5g of a white solid. LCMS found 280[ M + H ]]+
Step 34-2:
Figure BDA0002196260460000733
dissolving compound 153(10.4g) in ethanol/water (110 mL; 10:1), adding sodium carbonate (5.9g), heating to 75 ℃ and reacting for 24 h; the reaction was checked by TLC. After the reaction was complete, the solvent was suspended and directly stirred and passed through the column to give 4.8g of a white solid. LCMS found 236[ M + H ] +
Step 34-3:
Figure BDA0002196260460000734
the starting material 154(4.5g,1eq) was added to 40% aqueous hydrobromic acid (45ml) and acetic acid (50ml) and reacted for 15 minutes in an external bath at 86 ℃ with detection by LC-MS. After the reaction is completed, the mixture is subjected to external bath at 40 ℃, and then is subjected to oil pump drying to obtain 3.16g of reddish brown solid. LCMS found 222[ M + H ] +
Step 34-4:
Figure BDA0002196260460000735
raw material 155(3g,1eq) was added to DMAc (100ml), and the mixture was clarified, bromine raw material (10g,2eq) and cesium carbonate (14g,3eq) were added to react at room temperature for 1 hour, and the reaction was checked by LC-MS. After the reaction was completed, ice water was added, EA (100mL) was added and extracted once, and the organic phase was sampled and passed through a column to obtain 2.2g of a white solid. LCMS found: 504[ M + H ] + step 34-5:
Figure BDA0002196260460000741
adding a raw material 126(200mg,2eq) into freshly distilled THF (5mL), cooling to about-15 ℃, under the protection of argon, dropwise adding a 2M NaHMDS solution (1mL,4eq) for reaction for 5 minutes, dropwise adding a raw material 156(100mg,1eq) THF (3mL), naturally heating for reaction for 2 hours, and detecting the reaction by TLC. After the reaction is completed, the mixture is directly stirred and passed through a column to obtain 104mg of light yellow solid. LCMS found:897[ M + H ] +
Step 34-6:
Figure BDA0002196260460000742
the starting material 157(50mg) was dissolved in THF (1mL), and 1M TBAF/THF (0.1mL) was added to the solution, followed by reaction at room temperature for 30 minutes and detection by TLC. After the reaction was completed, ethyl acetate/water was added for extraction, and a reversed phase column was used to prepare 11mg of a yellow solid. LCMS found 659.2[ M + H ]]+1H NMR(400MHz,DMSO-d6)δ10.31(s,1H),9.32(s,1H),8.88(s,1H),8.43(d,J=8.1Hz,1H),8.20(s,1H),8.15(s,1H),8.07(d,J=5.7Hz,1H),7.67–7.54(m,2H),7.41–7.27(m,2H),7.18(d,J=5.8Hz,1H),7.07(d,J=7.6Hz,1H),6.89(d,J=7.5Hz,1H),6.73(d,J=7.4Hz,1H),4.86(s,1H),4.74(s,1H),4.23(s,1H),4.07(d,J=7.8Hz,5H),3.84(q,J=13.7Hz,2H),3.65(s,2H),2.77(t,J=7.9Hz,1H),2.69(d,J=8.1Hz,1H),2.41(d,J=10.0Hz,1H),2.10(s,3H),2.04(s,4H),1.59(s,1H),.
TABLE 2 Synthesis of the following Compounds from the corresponding starting materials according to the synthetic route LW1005-002
Figure BDA0002196260460000743
Figure BDA0002196260460000751
TABLE 3 Synthesis of the following Compounds from the corresponding starting materials according to the synthetic route LW1005-032
Figure BDA0002196260460000752
TABLE 4 Synthesis of the following Table Compounds from the corresponding starting materials according to the synthetic route LW1005-016
Figure BDA0002196260460000753
TABLE 5 Synthesis of the following Compounds from the corresponding starting materials according to the synthetic route LW1005-013
Figure BDA0002196260460000754
Figure BDA0002196260460000761
TABLE 6 Synthesis of the following Table Compounds from the corresponding starting materials according to the synthetic route LW1005-026
Figure BDA0002196260460000762
Figure BDA0002196260460000771
Figure BDA0002196260460000781
TABLE 7 Synthesis of the following Table Compounds from the corresponding starting materials according to the synthetic route LW1005-024
Figure BDA0002196260460000782
Figure BDA0002196260460000791
Figure BDA0002196260460000801
TABLE 8 Synthesis of the following Compounds from the corresponding starting materials according to the synthetic route LW1005-025
Figure BDA0002196260460000802
Figure BDA0002196260460000811
TABLE 8 Synthesis of the following Table Compounds from the corresponding starting materials according to the synthetic route LW1005-008
Figure BDA0002196260460000812
Figure BDA0002196260460000821
Biological assay
Example A: PD-1/PD-L1 homogeneous time-resolved fluorescence (HTRF) binding assay
The assay was performed in standard black 384 well polystyrene plates with a final volume of 20. mu.L. Firstly, the methodInhibitors were serially diluted in DMSO and added to the plate wells, followed by the addition of the other reaction components. The final concentration of DMSO in the assay was 1%. The assay was performed in PBS buffer (pH7.4) containing 0.05% Tween-20 and 0.1% BSA at 25 ℃. Recombinant human PD-L1 protein (19-238) with a His tag at the C-terminus was purchased from Acro biosystems (PD 1-H5229). Recombinant human PD-1 protein (25-167) with an Fc marker at the C-terminus was also purchased from Acrobiosystems (PD 1-H5257). PD-L1 and PD-1 protein were diluted in assay buffer and 0.1. mu.l of the solution was extracted and added to the plate well. The plates were centrifuged and the proteins were preincubated with inhibitors for 40 minutes. After incubation, 0.1. mu.l of HTRF detection buffer containing europium-blocked labeled anti-human IgG (Perkinelmer-AD0212) Fc specific and anti-His
Figure BDA0002196260460000823
Allophycocyanin (APC, Perkinelmer-AD0059H) conjugated antibody. After centrifugation, the plates were incubated at 25 ℃ for 60 minutes. The data were read (665nm/620nm ratio) in a PHERAStar FS plate reader. The final concentrations in the assay were-3 nM PD1, 10nM PD-L1, 1nM europium anti-human IgG and 20nM anti-His-allophycocyanin. The activity data were fitted using GraphPad Prism 5.0 software to give IC50 values for the inhibitors.
The compound IC50 values illustrated in the examples are represented in the following manner: IC 50: 100nM or less; 100-1000 nM; 1000nM +++, 2
Data for the example compounds obtained using the PD-1/PD-L1 Homogeneous Time Resolved Fluorescence (HTRF) binding assay described in example a are provided in table 1.
Figure BDA0002196260460000822
Figure BDA0002196260460000831
Figure BDA0002196260460000841
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 compound of formula I, or an optical isomer, hydrate, solvate, or pharmaceutically acceptable salt thereof:
Figure RE-FDA0002336517440000011
wherein n, m, p and q are each independently selected from 0,1, 2,3 or 4;
L1、L2selected from the group consisting of: a bond, substituted or unsubstituted C1-C4 alkylene, substituted or unsubstituted C2-C4 alkenylene, substituted or unsubstituted C2-C4 alkynylene, -S-, -O-, substituted or unsubstituted-NH-, -S (O)2-, substituted or unsubstituted-NHC (O) NH-, or,
Figure RE-FDA0002336517440000012
Substituted or unsubstituted
Figure RE-FDA0002336517440000013
Substituted or unsubstituted
Figure RE-FDA0002336517440000014
Substituted or unsubstituted
Figure RE-FDA0002336517440000015
Figure RE-FDA0002336517440000016
Is a group having the structure shown below:
Figure RE-FDA0002336517440000017
Figure RE-FDA0002336517440000018
wherein the content of the first and second substances,
Z1selected from the group consisting of: o, S, NRf, N-O-Rf; wherein, said Rf is selected from the group consisting of: H. substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C8Cycloalkyl, substituted or unsubstituted C6-C10Aryl, substituted or unsubstituted C6-C10Heteroaryl, cyano, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstituted C1-C6Alkyl, -C (═ O) -substituted or unsubstituted C3-C10Cycloalkyl, -C (═ O) -substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6An alkynyl group;
Z2、Z3、Z4each independently selected from the group consisting of: n, CH2、N-O、SO、SO2C (═ O), NRa, CRa; wherein Ra is selected from the group consisting of: H. chloro, bromo, fluoro, iodo, cyano, halogen, hydroxy, nitro, NRf, substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C8Cycloalkyl, substituted or unsubstituted C6-C10Aryl, substituted or unsubstituted C6-C10Heteroaryl, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstituted C1-C6Alkyl, -C (═ O) -substituted or unsubstituted C3-C10Cycloalkyl, substituted or unsubstituted C2-C6Alkenyl, substituted or unsubstituted C2-C6Alkynyl, -C (═ O) -Substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6An alkynyl group;
Y1、Y2、Y3each independently selected from the group consisting of: CH. CH (CH)2、NH、NRa、N、N-O、CF、CRa、C(Ra)2O, S, SO or SO2
Figure RE-FDA0002336517440000021
Is a single bond or a double bond;
and is
Figure RE-FDA0002336517440000022
Is an aromatic or non-aromatic moiety;
r is H, substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C6-C10Aryl, cyano, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstituted C1-C6Alkyl, -C (═ O) -substituted or unsubstituted C3-C10Cycloalkyl, -C (═ O) -substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6Alkynyl, or- (L)1a)r-(L2a)s-(L3a)s-, in which,
each L1aEach independently is a group selected from the group consisting of; chemical bond, substituted or unsubstituted C1-C7Alkylene, substituted or unsubstituted C2-C4 alkenylene, substituted or unsubstituted C2-C4 alkynylene, -S-, -O-, substituted or unsubstituted-NH-, -S (O)2-、
L2aSelected from the group consisting of: substituted or unsubstituted C6-C12 arylene, substituted or unsubstituted 5-12 membered heteroarylene having 1-3 heteroatoms, substituted or unsubstituted C3-C8 cycloalkylene, substituted or unsubstituted 5-10 membered heterocyclylene having 1-3 heteroatoms;
L3aselected from the group consisting of: H.substituted or unsubstituted C1-C10 alkyl, C1-C10 aryl, -CN, hydroxy, amino, carboxy, -CO-NH-SO2-Rg、-NH-SO2-Rg、-SO2-NH-CO-Rg
r is 1,2,3, 4, 5, 6;
s is 0,1, 2 respectively;
rd, Re are each independently selected from the group consisting of: H. substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C10Cycloalkyl, substituted or unsubstituted C6-C10An aryl group;
Figure RE-FDA0002336517440000023
selected from the group consisting of: substituted or unsubstituted 5-12 membered heteroaryl, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-12 membered heterocyclyl, substituted or unsubstituted 5-12 membered C5-C12 cyclyl, wherein the 5-12 membered heteroaryl and 5-12 membered heterocyclyl have 1-4 heteroatoms selected from B, P, N, O, S, wherein P, N, O as ring-forming atoms may be oxo and one or more ring-forming carbon atoms may be replaced by carbonyl; or the said
Figure RE-FDA0002336517440000024
Is absent; or the said
Figure RE-FDA0002336517440000025
Is equal to
Figure RE-FDA0002336517440000026
Figure RE-FDA0002336517440000027
Each independently a ring-forming divalent group selected from the group consisting of:
Figure RE-FDA0002336517440000028
Figure RE-FDA0002336517440000029
Figure RE-FDA00023365174400000210
wherein, the bonding position of the ring can be N or C;
R1、R2、R3and R4Each independently selected from the group consisting of: H. halogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C8 cycloalkyl, oxo (i.e., ═ O), ═ NRf, -CN, hydroxy, NRdRe (e.g., amino), substituted or unsubstituted C1-C6 amine, substituted or unsubstituted- (C1-C6 alkylene) -NH- (C1-C6 alkylene), carboxy, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-to 12-membered heteroaryl having 1 to 3 heteroatoms, substituted or unsubstituted 5-to 12-membered heterocyclyl having 1 to 4 heteroatoms, substituted or unsubstituted
Figure RE-FDA0002336517440000031
Substituted or unsubstituted
Figure RE-FDA0002336517440000032
Wherein, Rb, Rc and RzEach independently selected from the group consisting of: H. substituted or unsubstituted C1-C8An alkyl group; or said Rb and Rc taken together with the adjacent N atom form a substituted or unsubstituted 5-10 membered heterocyclic group having 1-3 heteroatoms selected from N, S and O; or (L)1a)r-(L2a)s-(L3a)s-;
Unless otherwise specified, "substituted" means substituted with one or more (e.g., 2,3, 4, etc.) substituents selected from the group consisting of: halogen: including but not limited to-F, Cl, Br, -CH2Cl、-CHCl2、-CCl3、-CH2F、-CHF2、-CF3Oxo, -CN, hydroxy, amino, C1-C6 alkylamino, carboxy,-NHAc, a group selected from the group consisting of unsubstituted or substituted with one or more substituents selected from the group consisting of: C1-C6 alkyl, C1-C6 alkoxy, C6-C10 aryl, C3-C8 cycloalkyl, halogenated C6-C10 aryl, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O, 5-10 membered heterocyclyl having 1-3 heteroatoms selected from N, S and O; the substituents are selected from the following group: halogen, hydroxy, carboxy, cyano, C1-C6 alkoxy, C1-C6 alkylamino;
in the formulae above, any of the heteroatoms is selected from the group consisting of: B. p, N, S and O.
2. The compound of claim 1, or an optical isomer, hydrate, solvate, or pharmaceutically acceptable salt thereof, having the structure of formula II:
Figure RE-FDA0002336517440000033
wherein n, m, p and q are each independently selected from 0,1, 2,3 or 4;
L1、L2selected from the group consisting of: a bond, substituted or unsubstituted C1-C4 alkylene, substituted or unsubstituted C2-C4 alkenylene, substituted or unsubstituted C2-C4 alkynylene, -S-, -O-, substituted or unsubstituted-NH-, -S (O)2-, substituted or unsubstituted-NHC (O) NH-, or,
Figure RE-FDA0002336517440000034
Substituted or unsubstituted
Figure RE-FDA0002336517440000035
Substituted or unsubstituted
Figure RE-FDA0002336517440000036
Substituted or unsubstituted
Figure RE-FDA0002336517440000037
Figure RE-FDA0002336517440000038
Is a group having the structure shown below:
Figure RE-FDA0002336517440000039
Figure RE-FDA0002336517440000041
wherein the content of the first and second substances,
Z1selected from the group consisting of: o, S, NRf, N-O-Rf; wherein, said Rf is selected from the group consisting of: H. substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C8Cycloalkyl, substituted or unsubstituted C6-C10Aryl, substituted or unsubstituted C6-C10Heteroaryl, cyano, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstituted C1-C6Alkyl, -C (═ O) -substituted or unsubstituted C3-C10Cycloalkyl, -C (═ O) -substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6An alkynyl group;
Z2、Z3、Z4each independently selected from the group consisting of: n, CH2、N-O、SO、SO2C (═ O), NRa, CRa; wherein Ra is selected from the group consisting of: H. chloro, bromo, fluoro, iodo, cyano, halogen, hydroxy, nitro, NRf, substituted or unsubstituted C1-C6Alkyl, substituted or unsubstituted C3-C8Cycloalkyl, substituted or unsubstituted C6-C10Aryl, substituted or unsubstituted C6-C10Heteroaryl, -C (═ O) -NRdRe, -C (═ O) -substituted or unsubstituted C1-C6Alkoxy, -C (═ O) -substituted or unsubstituted C1-C6Alkyl, -C (O) -substituted or unsubstituted C3-C10Cycloalkyl, substituted or unsubstituted C2-C6Alkenyl, substituted or unsubstituted C2-C6Alkynyl, -C (═ O) -substituted or unsubstituted C2-C6Alkenyl, -C (═ O) -substituted or unsubstituted C2-C6An alkynyl group;
Y1、Y2、Y3each independently selected from the group consisting of: CH. CH (CH)2、NH、NRa、N、N-O、CF、CRa、C(Ra)2O, S, SO or SO2
Figure RE-FDA0002336517440000042
Is a single bond or a double bond;
and is
Figure RE-FDA0002336517440000043
Are aromatic or non-aromatic segments.
R1、R2、R3And R4Each independently selected from the group consisting of: H. halogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C8 cycloalkyl, oxo (i.e., ═ O), ═ NRf, -CN, hydroxy, NRdRe (e.g., amino), substituted or unsubstituted C1-C6 amine, substituted or unsubstituted- (C1-C6 alkylene) -NH- (C1-C6 alkylene), carboxy, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-to 12-membered heteroaryl having 1 to 3 heteroatoms, substituted or unsubstituted 5-to 12-membered heterocyclyl having 1 to 4 heteroatoms, substituted or unsubstituted
Figure RE-FDA0002336517440000044
Substituted or unsubstituted
Figure RE-FDA0002336517440000045
Or (L)1a)r-(L2a)s-(L3a)s-。
3. The compound of claim 1, wherein said compound is
Figure RE-FDA0002336517440000046
Has a structure represented by the following formula:
Figure RE-FDA0002336517440000051
wherein the content of the first and second substances,
X6、X7、X8、X9、X10and X11Each independently selected from the group consisting of: n, CR, respectively;
R6selected from the group consisting of: H. halogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C8 cycloalkyl, oxo (i.e., ═ O), ═ NRf, -CN, hydroxy, NRdRe (e.g., amino), substituted or unsubstituted C1-C6 amine, substituted or unsubstituted- (C1-C6 alkylene) -NH- (C1-C6 alkylene), carboxy, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-to 12-membered heteroaryl having 1 to 3 heteroatoms, substituted or unsubstituted 5-to 12-membered heterocyclyl having 1 to 4 heteroatoms, substituted or unsubstituted
Figure RE-FDA0002336517440000052
Substituted or unsubstituted
Figure RE-FDA0002336517440000053
Or (L)1a)r-(L2a)s-(L3a)s-,-C0-8-O-R8,-C0-8-C(O)OR8,-C0-8-OC(O)OR8,-C0-8-NR8R9,-C0-8-N(R8)C(O)R9,-C0-8-C(O)NR8R9
R8And R9Each independently selected from the group consisting of: H. hydroxy, substituted or unsubstituted C1-C10 alkyl, substituted or unsubstituted C2-C6 alkenyl, substituted or unsubstituted C2-C6 alkynyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C3-C8 cycloalkyl, oxo (i.e., ═ O), ═ NRf, -CN, hydroxy, NRdRe (e.g., amino), substituted or unsubstituted C1-C6 amine, substituted or unsubstituted- (C1-C6 alkylene) -NH- (C1-C6 alkylene), carboxy, substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted 5-to 12-membered heteroaryl having 1 to 3 heteroatoms, substituted or unsubstituted 5-to 12-membered heterocyclyl having 1 to 4 heteroatoms, substituted or unsubstituted
Figure RE-FDA0002336517440000054
Substituted or unsubstituted
Figure RE-FDA0002336517440000055
Or (L)1a)r-(L2a)s-(L3a)s-。
4. The compound of claim 1, wherein the compound of formula I is selected from the group consisting of formulae Id-1, Id-2, and Id-3, or an optical isomer, hydrate, solvate, or pharmaceutically acceptable salt thereof:
Figure RE-FDA0002336517440000061
wherein each group is as defined above.
5. The compound of claim 1, or an optical isomer, hydrate, solvate, or pharmaceutically acceptable salt thereof, wherein said ring is
Figure RE-FDA0002336517440000062
And/or
Figure RE-FDA0002336517440000063
Having a substituent represented by formula IV below:
Figure RE-FDA0002336517440000064
wherein each L is4Independently selected from the group consisting of: substituted or unsubstituted C1-C4 alkylene, -S-, -O-, -NRa-, -S (O) -, -S (O)2-; preferably a substituted or unsubstituted C1-C4 alkylene group, with the proviso that each L4The co-formed structure is chemically stable;
Figure RE-FDA0002336517440000065
selected from the group consisting of: substituted or unsubstituted C5-C10 cycloalkyl, substituted or unsubstituted 3-10 membered heterocyclyl having 1-3 heteroatoms selected from B, P, N, S and O; preferably, said
Figure RE-FDA0002336517440000066
Is a 3-8 membered nitrogen containing heterocyclic group;
each R is5Each independently selected from the group consisting of: substituted or unsubstituted C1-C6 alkyl, -CN, hydroxy, amino, carboxy; wherein the substituents are selected from the group consisting of: halogen, hydroxy, carboxy, cyano, C1-C6 alkoxy.
Preferably, the compounds are selected from the following table;
Figure RE-FDA0002336517440000071
Figure RE-FDA0002336517440000081
Figure RE-FDA0002336517440000091
Figure RE-FDA0002336517440000101
Figure RE-FDA0002336517440000111
Figure RE-FDA0002336517440000121
Figure RE-FDA0002336517440000131
Figure RE-FDA0002336517440000141
Figure RE-FDA0002336517440000151
6. a process for the preparation of a compound of formula I according to claim 1, comprising a step selected from the group consisting of schemes 1,2 and 3:
synthesis scheme 1
Figure RE-FDA0002336517440000152
(a) Taking halide 1-1 and appropriate coupling reagent 1-2 (such as boric acid, boric acid ester, tin reagent or Grignard reagent) as basic raw materials, and obtaining an intermediate compound 1-3 through coupling reaction (such as Suzuki, Stille or Kumada coupling) catalyzed by palladium or copper;
(b) taking the intermediate 1-3 as a raw material, and reacting with carboxylic acid 1-4 under the action of a condensing agent (such as HATU, EDCI or HBTU) to obtain an amide intermediate 1-5;
(c) taking the intermediate 1-5 as a raw material, and removing a protecting group (Boc) under an acidic condition to obtain an intermediate 1-6;
(d) taking the intermediate 1-6 as a raw material, and carrying out nucleophilic substitution reaction with a halide under an alkaline condition, or carrying out reductive amination reaction with aldehyde or ketone under the action of a reducing agent to obtain a target compound I;
synthesis scheme 2:
Figure RE-FDA0002336517440000153
(a) taking carboxylic ester 2-1 as a raw material, and carrying out an aminolysis reaction with amine 2-2 under the catalysis of Lewis acid to obtain an intermediate compound 2-3;
(b) taking the intermediate 2-3 and a proper coupling reagent 2-4 (such as boric acid, boric acid ester, a tin reagent or a Grignard reagent) as basic raw materials, and obtaining an intermediate compound 2-5 through a coupling reaction (such as Suzuki, Stille or Kumada coupling) catalyzed by palladium or copper;
(c) taking the intermediate 2-5 as a raw material, and removing a protecting group (Boc) under an acidic condition to obtain an intermediate 2-6;
(d) taking the intermediate 2-6 as a raw material, and carrying out nucleophilic substitution reaction with a halide under an alkaline condition, or carrying out reductive amination reaction with aldehyde or ketone under the action of a reducing agent to obtain a target compound I;
(a) carrying out reductive amination reaction in the synthesis scheme 3 to obtain a target compound I;
synthesis scheme 3:
Figure RE-FDA0002336517440000161
the method 3 comprises the following steps:
(a) carrying out Suzuki coupling on the raw materials of the boron ester 3-1 and the halide 3-2 under the catalysis of palladium to obtain an intermediate compound 3-3;
(b) carboxylic acid 3-4 and a compound 3-5 are taken as raw materials, and a cyclization reaction is carried out under the action of a proper dehydrating agent to obtain an intermediate compound 3-3;
(c) taking aldehyde 3-6 as a raw material, and carrying out cyclization reaction under the action of a proper oxidant to obtain an intermediate 3-3;
(d) using the intermediate 3-3 and a proper coupling reagent 3-7 (such as boric acid, boric acid ester, a tin reagent or a Grignard reagent) as basic raw materials, and performing coupling reaction (such as Suzuki, Stille or Kumada coupling) catalyzed by palladium or copper to obtain a target compound I;
said Cy is
Figure RE-FDA0002336517440000162
Is composed of
Figure RE-FDA0002336517440000163
Y1、Y2、Z1、Z2、Z3And R is as defined above.
7. A pharmaceutical composition comprising (1) a compound of claim 1 or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof; (2) a pharmaceutically acceptable carrier.
8. Use of a compound according to claim 1 or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof, or a pharmaceutical composition according to claim 7, for the preparation of a pharmaceutical composition for the prophylaxis and/or treatment of diseases associated with the activity or expression of PD-1/PD-L1.
9. A PD-1/PD-L1 inhibitor, wherein said inhibitor comprises a compound of claim 1, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof.
10. A method of inhibiting the PD-1/PD-L1 interaction in vitro comprising the steps of: contacting a compound of claim 1, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof, with PD-L1 protein.
CN201910848960.6A 2019-09-09 2019-09-09 Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof Pending CN112457331A (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN201910848960.6A CN112457331A (en) 2019-09-09 2019-09-09 Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof
CN202080063271.5A CN114364685A (en) 2019-09-09 2020-09-09 Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof
PCT/CN2020/114264 WO2021047556A1 (en) 2019-09-09 2020-09-09 Nitrogen-containing heterocyclic compound, and preparation method therefor, pharmaceutical composition comprising same and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910848960.6A CN112457331A (en) 2019-09-09 2019-09-09 Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof

Publications (1)

Publication Number Publication Date
CN112457331A true CN112457331A (en) 2021-03-09

Family

ID=74807414

Family Applications (2)

Application Number Title Priority Date Filing Date
CN201910848960.6A Pending CN112457331A (en) 2019-09-09 2019-09-09 Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof
CN202080063271.5A Pending CN114364685A (en) 2019-09-09 2020-09-09 Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202080063271.5A Pending CN114364685A (en) 2019-09-09 2020-09-09 Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof

Country Status (2)

Country Link
CN (2) CN112457331A (en)
WO (1) WO2021047556A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021147940A1 (en) * 2020-01-21 2021-07-29 上海华汇拓医药科技有限公司 Pd-1/pd-l1 inhibitor, preparation method therefor, and use thereof

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117871740B (en) * 2024-03-11 2024-05-10 炉霍雪域俄色有限责任公司 Russian color fruit raw pulp liquid chromatography quality detection method

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018119224A1 (en) * 2016-12-22 2018-06-28 Incyte Corporation Tetrahydro imidazo[4,5-c]pyridine derivatives as pd-l1 internalization inducers
WO2018119266A1 (en) * 2016-12-22 2018-06-28 Incyte Corporation Benzooxazole derivatives as immunomodulators
WO2018195321A1 (en) * 2017-04-20 2018-10-25 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
CN109678796A (en) * 2017-10-19 2019-04-26 上海长森药业有限公司 PD-1/PD-L1 micromolecular inhibitor and its preparation method and application

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111039942B (en) * 2018-10-12 2023-04-14 上海长森药业有限公司 Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018119224A1 (en) * 2016-12-22 2018-06-28 Incyte Corporation Tetrahydro imidazo[4,5-c]pyridine derivatives as pd-l1 internalization inducers
WO2018119266A1 (en) * 2016-12-22 2018-06-28 Incyte Corporation Benzooxazole derivatives as immunomodulators
WO2018195321A1 (en) * 2017-04-20 2018-10-25 Gilead Sciences, Inc. Pd-1/pd-l1 inhibitors
CN109678796A (en) * 2017-10-19 2019-04-26 上海长森药业有限公司 PD-1/PD-L1 micromolecular inhibitor and its preparation method and application

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021147940A1 (en) * 2020-01-21 2021-07-29 上海华汇拓医药科技有限公司 Pd-1/pd-l1 inhibitor, preparation method therefor, and use thereof
CN114650993A (en) * 2020-01-21 2022-06-21 上海华汇拓医药科技有限公司 PD-1/PD-L1 inhibitor and preparation method and application thereof

Also Published As

Publication number Publication date
WO2021047556A1 (en) 2021-03-18
CN114364685A (en) 2022-04-15

Similar Documents

Publication Publication Date Title
CN111039942B (en) Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof
CN106459009B (en) Hepatitis C virus inhibitor and its pharmaceutical applications
TW202043217A (en) Bcl-2 inhibitors
CN114615981A (en) KRAS G12D inhibitors
CN112778276A (en) Compound as SHP2 inhibitor and application thereof
CN114364676B (en) Novel tricyclic aromatic heterocyclic compound, preparation method, pharmaceutical composition and application thereof
KR20240029051A (en) KRAS G12D inhibitors and uses thereof
CA3075880A1 (en) Tetrahydro-imidazo quinoline compositions as cbp/p300 inhibitors
CN113874015B (en) Thienopyridine inhibitors of RIPK2
CN114364685A (en) Nitrogen-containing heterocyclic compound, and preparation method, pharmaceutical composition and application thereof
CN113637013A (en) Preparation and application of biaryl ring linked aromatic heterocyclic derivative as immunomodulator
CN114364673A (en) Aromatic heterocyclic compound containing tricyclic structure, and preparation method and application thereof
CN113045569A (en) Compounds useful as RET kinase inhibitors and uses thereof
CN114364683A (en) Preparation and application of aromatic heterocyclic derivative as immunomodulator
TW202118756A (en) Heterocyclic compounds as bcr-abl inhibitors
CN112300161B (en) Compound for treating and/or preventing hepatitis B virus infection and preparation method and application thereof
CN112521372B (en) Apoptosis protein inhibitor and preparation method and application thereof
EP4159736A1 (en) Novel tricyclic aromatic heterocyclic compound and preparation method therefor, pharmaceutical composition and use thereof
WO2023016529A1 (en) Naphthyridine derivative as atr inhibitor and method for preparing same
WO2024083256A1 (en) Pan-kras degrading agent, and preparation method therefor and use thereof
WO2023083330A1 (en) Drak2 inhibitor, and preparation method therefor and use thereof
CN115677831A (en) Peptidomimetic STAT degradation drug, composition and application
CN115960083A (en) Heterocyclic compounds, their preparation and use
WO2023232130A1 (en) Heterocyclic compound ccr4 inhibitor and user thereof
WO2023232135A1 (en) Pde4b inhibitor and use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination