CN115244041A - Vinyl-substituted pyridines - Google Patents

Vinyl-substituted pyridines Download PDF

Info

Publication number
CN115244041A
CN115244041A CN202180020076.9A CN202180020076A CN115244041A CN 115244041 A CN115244041 A CN 115244041A CN 202180020076 A CN202180020076 A CN 202180020076A CN 115244041 A CN115244041 A CN 115244041A
Authority
CN
China
Prior art keywords
compound
mol
ethyl acetate
give
dissolved
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
CN202180020076.9A
Other languages
Chinese (zh)
Inventor
陆剑宇
贺辉君
胡国平
胡利红
丁照中
黎健
陈曙辉
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.)
Medshine Discovery Inc
Original Assignee
Medshine Discovery Inc
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 Medshine Discovery Inc filed Critical Medshine Discovery Inc
Publication of CN115244041A publication Critical patent/CN115244041A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • 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
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings

Landscapes

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

Abstract

Provides a vinyl substituted pyridine compound (I) and pharmaceutically acceptable salts thereof, and application of the compound and the pharmaceutically acceptable salts thereof in preparing medicaments for treating related diseases.

Description

Vinyl-substituted pyridines
The following priority is claimed in the present application:
CN202010163709.9, application date 2020, 03 month 10.
Technical Field
The invention relates to a vinyl substituted pyridine compound and a pharmaceutically acceptable salt thereof, and application of the compound and the pharmaceutically acceptable salt thereof in preparation of medicaments for treating related diseases.
Background
Tyrosine kinases can be classified as either growth factor receptor (e.g., axl, mer, EGFR, PDGFR, FGFR and erbB 2) or non-receptor (e.g., c-src and bcr-abl) kinases. Receptor-type tyrosine kinases make up about 20 different subfamilies. Non-receptor tyrosine kinases constitute a number of subfamilies. These tyrosine kinases have a variety of biological activities. Receptor tyrosine kinases are large enzymes that span the cell membrane, have an extracellular binding domain for growth factors, a transmembrane domain, and an intracellular portion that functions as a kinase that phosphorylates specific tyrosine residues in proteins and thus affects cell proliferation. Abnormal or inappropriate protein kinase activity can lead to an increase in disease states associated with such abnormal kinase activity.
Disruption of VEGF receptor signaling is a very attractive therapeutic target in cancer, as angiogenesis is a prerequisite for the growth of all solid tumors. Angiogenesis is an important component of certain normal physiological processes (e.g., embryogenesis and wound healing), but aberrant angiogenesis leads to several pathological conditions, particularly tumor growth. Recent studies have found that VEGF directly inhibits tumor immune responses and that anti-angiogenic therapy enhances tumor immunity through normalization of blood vessels. (Fukumura et al, "Enhancing cancer immunology using antimiangiogenics: opportunities and strains", nat Rev Clin Oncol.2018). VEGF-A (vascular endothelial growth factor A) is Sub>A key factor in promoting tumor neovascularization (angiogenesis). VEGF induces endothelial cell proliferation and migration through signaling by receptors, namely VEGFR-1 (Flt-1), VEGFR-2 (KDR) and VEGFR-3 (Flt-4). These signaling responses are heavily dependent on receptor dimerization and activation of intrinsic Receptor Tyrosine Kinase (RTK) activity. Binding of VEGF as a disulfide-linked homodimer stimulates receptor dimerization and activation of the RTK domain. Kinase activity autophosphorylates cytoplasmic receptor tyrosine residues, which then serve as binding sites for molecules involved in the propagation of signaling cascades. Although multiple pathways are probably elucidated for all three receptors, VEGFR-2 signaling is most widely studied, and its mitogenic response is thought to involve ERK-1 and ERK-2 mitogen-activated protein kinases.
Axl is a member of the TAM receptor family, which also includes Mer and Tyro3. These three are activated by a common ligand, growth arrest specific protein 6 (Gas 6), which normally prays for embryonic development in cell survival, migration and differentiation. Axl overexpression and signal transduction have been implicated in a number of malignancies in humans, such as colon, breast, glioma, thyroid, gastric, melanoma, lung and renal cell carcinoma.
Inhibition of Axl may more completely arrest the growth of the cancer, both exert complementary and overlapping effects, and therapeutic regimens targeting these two RTKs may be more effective than single targeting agents. In addition, therapeutic antibodies targeting Axl can block Axl function in the tumor stroma, and the additive effects of inhibiting Axl in combination with inhibiting VEGF receptors (VEGFRs) suggest that blocking Axl function is an effective means for promoting "anti-angiogenic therapy". Cancer cells employ a variety of mechanisms to evade tight cell regulation processes such as cell proliferation, apoptosis, and aging. Thus, many tumors escape the action of single kinase inhibitors. Sitravatinib enhances tumor immunity by simultaneously inhibiting VEGFR-2 and Axl signaling. Therefore, targeting VEGFRs/Axl is a potential treatment strategy for targeting cancer cells, and can avoid the overcoming of the independent inhibition effect of tumor cells on VEGFRs, axl and the like, so that the targeted VEGFRs/Axl becomes an advanced cancer treatment scheme.
Figure PCTCN2021079337-APPB-000001
Disclosure of Invention
The present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof,
Figure PCTCN2021079337-APPB-000002
wherein the content of the first and second substances,
ring a is selected from a 5-membered heteroaryl group, said 5-membered heteroaryl group comprising 2N heteroatoms;
alternatively, ring a is selected from 6-membered heteroaryl, said 6-membered heteroaryl comprising up to 2N atoms;
T 1 and T 2 Are each independently selected from C (R) 5 ) And N;
R 1 and R 3 Are each independently selected from H, F and CH 3
R 2 H, F and Cl;
R 4 each independently selected from H, C 1~6 Alkyl radical, C 1~6 Heteroalkyl group, C 3~6 Cycloalkyl, 3-to 6-membered heterocycloalkyl and-L-R 6 Said C is 1~6 Alkyl radical, C 1~6 Heteroalkyl group, C 3~6 Cycloalkyl, 3-to 6-membered heterocycloalkyl and-L-R 6 Optionally substituted with 1,2 or 3R;
R 5 selected from H and Cl;
R 6 selected from H, OH, 5-to 6-membered heterocycloalkyl and C 1~3 An alkyl group;
l is selected from-CH 2 -、-CH 2 -CH 2 -and
Figure PCTCN2021079337-APPB-000003
n is selected from 0, 1,2 and 3;
r is respectively and independently selected from F, cl, br, I, OH, CN, COOH and NH 2 、-NHCH 3 、-N(CH3) 2 、CH 3 、CH 2 CH 3 、CF 3 、-OCH 3 、-OCH 2 CH 3 、-O-CH(CH 3 ) 2 、-C(=O)OCH 3 、-C(=O)CH 3 and-C (= O) CH 2 CH 3
The 5-membered heteroaryl, 6-membered heteroaryl, C 1~6 Heteroalkyl and 3-to 6-membered heterocycloalkyl each independently contain 1,2, or 3 heteroatoms or groups of heteroatoms independently selected from O, S, N and NH.
In some embodiments of the invention, a compound of formula (I) as described above, or a pharmaceutically acceptable salt thereof, wherein,
ring a is selected from a 5-membered heteroaryl group, said 5-membered heteroaryl group comprising 2N heteroatoms;
alternatively, ring a is selected from 6-membered heteroaryl, said 6-membered heteroaryl comprising up to 2N atoms;
T 1 and T 2 Are each independently selected from C (R) 5 ) And N;
R 1 and R 3 Are each independently selected from H, F and CH 3
R 2 H, F and Cl; r 4 Each independently selected from H, C 1~6 Alkyl radical, C 1~6 Heteroalkyl group, C 3~6 Cycloalkyl and 3-to 6-membered heterocycloalkyl, said C 1~6 Alkyl radical, C 1~6 Heteroalkyl group, C 3~6 Cycloalkyl and 3-6 membered heterocycloalkyl optionally substituted with 1,2 or 3R;
R 5 selected from H and Cl;
n is selected from 0, 1,2 and 3;
r is independently selected from F, cl, br, I, OH, CN, COOH, NH2, -NHCH3, -N (CH 3) 2, CH3, CH2CH3, CF3, -OCH2CH3, -O-CH (CH 3) 2, -C (= O) OCH3, -C (= O) CH3 and-C (= O) CH2CH3.
The 5-membered heteroaryl, 6-membered heteroaryl, C 1~6 Heteroalkyl and 3-to 6-membered heterocycloalkyl each independently contain 1,2, or 3 heteroatoms or groups of heteroatoms independently selected from O, S, N and NH.
In some embodiments of the invention, the above compound or a pharmaceutically acceptable salt thereof, wherein ring a is selected from pyridyl, pyrazolyl, and imidazolyl, and the other variables are as defined herein.
In some embodiments of the invention, the compound or pharmaceutically acceptable salt thereof, wherein R is 4 Each independently selected from H, C 1~4 Alkyl radical, C 1~4 Heteroalkyl, 5-to 6-membered heterocycloalkyl and-L-R 6 Said, C 1~4 Alkyl radical, C 1~4 Heteroalkyl, 5-to 6-membered heterocycloalkyl and-L-R 6 Optionally substituted with 1,2 or 3R, the other variables being as defined herein.
In some embodiments of the invention, the compound or pharmaceutically acceptable salt thereof, wherein R is 4 Each independently selected from H,
Figure PCTCN2021079337-APPB-000004
Figure PCTCN2021079337-APPB-000005
The described
Figure PCTCN2021079337-APPB-000006
Figure PCTCN2021079337-APPB-000007
Optionally substituted with 1,2 or 3R, the other variables being as defined herein.
In some embodiments of the invention, the above compound or a pharmaceutically acceptable salt thereof, wherein R 4 Each independently selected from H,
Figure PCTCN2021079337-APPB-000008
Figure PCTCN2021079337-APPB-000009
Other variables are as defined herein.
In some embodiments of the invention, the above compound or a pharmaceutically acceptable salt thereof, wherein R 4 Each independently selected from H, C 1~4 Alkyl and C 1~4 Heteroalkyl group of said C 1~4 Alkyl and C 1~4 Heteroalkyl is optionally substituted with 1,2 or 3R, the other variables being as defined herein.
In some embodiments of the invention, the compound or pharmaceutically acceptable salt thereof, wherein R is 4 Each independently selected from H,
Figure PCTCN2021079337-APPB-000010
The above-mentioned
Figure PCTCN2021079337-APPB-000011
Optionally substituted with 1,2 or 3R, and the other variables are as defined herein.
In some embodiments of the invention, the compound or pharmaceutically acceptable salt thereof, wherein R is 4 Each independently selected from H,
Figure PCTCN2021079337-APPB-000012
Other variables are as defined herein.
In some embodiments of the invention, the compound or a pharmaceutically acceptable salt thereof, wherein the structural unit
Figure PCTCN2021079337-APPB-000013
Is selected from
Figure PCTCN2021079337-APPB-000014
The other variables are as defined herein.
In some embodiments of the invention, the compound or a pharmaceutically acceptable salt thereof, wherein the structural unit
Figure PCTCN2021079337-APPB-000015
Is selected from
Figure PCTCN2021079337-APPB-000016
Other variables are as defined herein.
In some embodiments of the invention, the compound or a pharmaceutically acceptable salt thereof, wherein the structural unit
Figure PCTCN2021079337-APPB-000017
Is selected from
Figure PCTCN2021079337-APPB-000018
Figure PCTCN2021079337-APPB-000019
Other variables are as defined herein.
In some embodiments of the invention, the compound or a pharmaceutically acceptable salt thereof, wherein the structural unit
Figure PCTCN2021079337-APPB-000020
Is selected from
Figure PCTCN2021079337-APPB-000021
Figure PCTCN2021079337-APPB-000022
Other variables are as defined herein.
In some embodiments of the invention, the compound or pharmaceutically acceptable salt thereof is selected from
Figure PCTCN2021079337-APPB-000023
Wherein the content of the first and second substances,
R 41 selected from H, C 1~4 Alkyl radical, C 1~4 Heteroalkyl, 6-membered heterocycloalkyl and-L-R 6 H, C as described 1~4 Alkyl radical, C 1~4 Heteroalkyl, 6-membered heterocycloalkyl and-L-R 6 Optionally substituted with 1,2 or 3R;
R 42 selected from H and-L-R 6 said-L-R 6 Optionally substituted with 1,2 or 3R;
R 6 selected from H, OH, 6-membered heterocycloalkyl and C 1~3 An alkyl group;
R 2 l and R are as defined herein.
In some embodiments of the invention, the compound or pharmaceutically acceptable salt thereof is selected from
Figure PCTCN2021079337-APPB-000024
R 1 、R 2 、R 3 And R 4 As defined herein.
The invention also provides a compound or a pharmaceutically acceptable salt thereof selected from
Figure PCTCN2021079337-APPB-000025
Figure PCTCN2021079337-APPB-000026
The invention also has some technical schemes which are formed by the random combination of the variables.
The invention also provides a pharmaceutical composition, which comprises a therapeutically effective amount of the compound or the pharmaceutically acceptable salt as an active ingredient and a pharmaceutically acceptable carrier.
The invention also provides application of the compound or the pharmaceutically acceptable salt or the composition in preparation of Axl and VEGFR-2 inhibitors.
In some embodiments of the invention, the above-described Axl and VEGFR-2 inhibitors are drugs for treating cancer.
Technical effects
The compounds of the invention are useful in the treatment of neoplastic diseases which respond to inhibition of protein tyrosine kinase activity, for example cancers which respond to inhibition of protein tyrosine kinase activity of growth factor receptors. The invention also provides novel compounds and methods for treating diseases responsive to inhibition of receptor-type tyrosine kinase signaling, e.g., cancers responsive to inhibition of VEGF receptor signaling and to inhibition of Axl kinase. The compound has excellent in vitro activity performance, and has good inhibitory activity on VEGFR2/KDR kinase and AXL kinase. The representative compounds of the present invention are greatly improved in permeability, more stable in metabolic stability, and also excellent in CYP inhibitory properties as compared to the prior art. The compound has good in-vivo PK property, and has better exposure and bioavailability. The compound has better drug forming property and better curative effect clinically.
Definitions and explanations
As used herein, the following terms and phrases are intended to have the following meanings, unless otherwise indicated. A particular term or phrase, unless otherwise specifically defined, should not be considered as indefinite or unclear, but rather construed according to ordinary meaning. When a trade name appears herein, it is intended to refer to its corresponding commodity or its active ingredient.
The term "pharmaceutically acceptable" as used herein is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The term "pharmaceutically acceptable salts" refers to salts of the compounds of the present invention, prepared from the compounds of the present invention found to have particular substituents, with relatively nontoxic acids or bases. When compounds of the present invention contain relatively acidic functional groups, base addition salts can be obtained by contacting such compounds with a sufficient amount of a base in neat solution or in a suitable inert solvent. Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amines or magnesium salts or similar salts. When compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting such compounds with a sufficient amount of acid, either in neat solution or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, hydrogen sulfate, hydroiodic acid, phosphorous acid, and the like; and salts of organic acids including acids such as acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, methanesulfonic, and the like; also included are salts of amino acids such as arginine and the like, and salts of organic acids such as glucuronic acid and the like. Certain specific compounds of the invention contain both basic and acidic functionalities and can thus be converted to any base or acid addition salt.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, which contains an acid or base, by conventional chemical methods. In general, such salts are prepared by the following method: prepared by reacting these compounds in free acid or base form with a stoichiometric amount of the appropriate base or acid, in water or an organic solvent or a mixture of the two.
The terms "optional" or "optionally" mean that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where said event or circumstance occurs and instances where it does not.
The term "substituted" means that any one or more hydrogen atoms on a particular atom is replaced with a substituent, and may include variations of deuterium and hydrogen, so long as the valency of the particular atom is normal and the substituted compound is stable. When the substituent is oxygen (i.e = O), it means that two hydrogen atoms are substituted.
The term "optionally substituted" means that it may or may not be substituted, and unless otherwise specified, the kind and number of substituents may be arbitrary on the basis of chemical realizability.
When any variable (e.g., R) occurs more than one time in the composition or structure of a compound, its definition in each case is independent. Thus, for example, if a group is substituted with 0 to 2R, the group may optionally be substituted with up to two R, and there are separate options for R in each case. Furthermore, combinations of substituents and/or variants thereof are permissible only if such combinations result in stable compounds.
When the number of one linking group is 0, e.g., - (CRR) 0 -, denotes that the linking group is a single bond, -C 0 alkyl-A indicates that the structure is actually-A.
When the number of a substituent is 0, it means that the substituent is absent, such as-A- (R) 0 Indicating that the structure is actually-a.
When a substituent is absent, it indicates that the substituent is absent, e.g., when X is absent in A-X, it indicates that the structure is actually A.
When one of the variables is selected from a single bond, it means that the two groups to which it is attached are directly linked, for example, in A-L-Z where L represents a single bond, it means that the structure is actually A-Z.
When a substituent is absent, it indicates that the substituent is absent, e.g., when X is absent in A-X, it indicates that the structure is actually A.
When no atom through which a substituent is attached to a substituted group is indicated in the listed substituents, such substituents may be bonded through any atom thereof, for example, a pyridyl group as a substituent may be attached to a substituted group through any one of carbon atoms on the pyridine ring.
When a substituent bond can be cross-linked to more than two atoms on a ring, such substituent may be bonded to any atom on the ring, e.g., a building block
Figure PCTCN2021079337-APPB-000027
Means that the substituent R can be substituted at any position on the cyclohexyl or cyclohexadiene.
When the listed linking groups do not indicate their direction of attachment, the direction of attachment is arbitrary, for example,
Figure PCTCN2021079337-APPB-000028
wherein the linking group L is-M-W-, in which case-M-W-can be formed by connecting the ring A and the ring B in the same direction as the reading sequence from left to right
Figure PCTCN2021079337-APPB-000029
The ring A and the ring B may be connected in the reverse direction of the reading sequence from left to right
Figure PCTCN2021079337-APPB-000030
Combinations of the linking groups, substituents, and/or variants thereof are permissible only if such combinations result in stable compounds.
Unless otherwise specified, when a group has one or more attachable sites, any one or more of the sites of the group may be attached to other groups by chemical bonds. When the chemical bond is not positioned and H atoms exist in the connectable sites, the number of the H atoms of the connectable sites is correspondingly reduced along with the number of the connected chemical bonds to become a group with corresponding valence number. The chemical bond linking said site to other groups may be a direct solid bond
Figure PCTCN2021079337-APPB-000031
Straight dotted line key
Figure PCTCN2021079337-APPB-000032
Or wavy lines
Figure PCTCN2021079337-APPB-000033
And (4) showing. For example-OCH 3 The straight solid line bond in (a) represents a bond to another group via an oxygen atom in the group;
Figure PCTCN2021079337-APPB-000034
the straight dotted bond in (3) represents the linkage to the other group through both ends of the nitrogen atom in the group;
Figure PCTCN2021079337-APPB-000035
the wavy line in (a) indicates that the phenyl group is bonded to other groups through the carbon atoms at the 1-and 2-positions in the phenyl group;
Figure PCTCN2021079337-APPB-000036
means that any of the available attachment sites on the piperidinyl group can be attached to another group via 1 bond, including at least
Figure PCTCN2021079337-APPB-000037
These 4 linkages, even though the-N-atom is depicted as H, are
Figure PCTCN2021079337-APPB-000038
Still comprise
Figure PCTCN2021079337-APPB-000039
This attachment is a group whose H at the site is reduced by 1 to the corresponding monovalent piperidinyl group, except when 1 bond is attached.
Unless otherwise specified, the number of atoms on a ring is generally defined as the number of ring members, e.g., "5 to 7 membered ring" means a "ring" around which 5 to 7 atoms are arranged.
Unless otherwise specified, the term "C 1~6 Alkyl "is intended to mean a straight or branched saturated hydrocarbon group consisting of 1 to 6 carbon atoms. Said C is 1~6 The alkyl group comprising C 1~5 、C 1~4 、C 1~3 、C 1~2 、C 2~6 、C 2~4 、C 6 And C 5 Alkyl, etc.; it may be monovalent (e.g., methyl), divalent (e.g., methylene), or multivalent (e.g., methine). C 1~6 Examples of alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, s-butyl and t-butyl), pentyl (including n-pentyl, isopentyl and neopentyl), hexyl, and the like.
Unless otherwise specified, the term "C 1~4 Alkyl "is intended to mean a straight or branched saturated hydrocarbon group consisting of 1 to 4 carbon atoms. Said C is 1~4 The alkyl group including C 1~2 、C 1~3 And C 2~3 Alkyl, etc.; it may be monovalent (e.g., methyl), divalent (e.g., methylene), or multivalent (e.g., methine). C 1~4 Examples of alkyl groups include, but are not limited to, methyl (Me), ethyl (Et), propyl (including n-propyl and isopropyl), butyl (including n-butyl, isobutyl, s-butyl and t-butyl), and the like.
The term "heteroalkyl," by itself or in combination with another term, means a stable straight or branched chain alkyl radical consisting of a number of carbon atoms and at least one heteroatom or heteroatom group, or combinations thereof. In some embodiments, the heteroatom is selected from B, O, N and S, wherein the nitrogen and sulfur atoms are optionally oxidized and the nitrogen heteroatom is optionally quaternized. In other embodiments, the heteroatom groups are selected from-C (= O) O-, -C (= O) -, -C (= S) -, -S (= O) 2 -、-C(=O)N(H)-、-N(H)-、-C(=NH)-、-S(=O) 2 N (H) -and-S (= O) N (H) -. In some embodiments, the heteroalkyl is C 1~6 A heteroalkyl group; in other embodiments, the heteroalkyl is C 1~4 A heteroalkyl group. The heteroatom or heteroatom group may be located at any internal position of the heteroalkyl group, including the position of attachment of the alkyl group to the remainder of the molecule, but the terms "alkoxy", "alkylamino" and "alkylthio" (or thioalkoxy) are used conventionally to refer to those alkyl groups that are attached to the remainder of the molecule through an oxygen atom, an amino group, or a sulfur atom, respectively. Examples of heteroalkyl groups include, but are not limited to, -OCH 3 、-OCH 2 CH 3 、-OCH 2 CH 2 CH 3 、-OCH 2 (CH 3 ) 2 、-CH 2 -CH 2 -O-CH 3 、-NHCH 3 、-N(CH 3 ) 2 、-NHCH 2 CH 3 、-N(CH 3 )(CH 2 CH 3 )、-CH 2 -CH 2 -NH- CH 3 、-CH 2 -CH 2 -N(CH 3 )-CH 3 、-SCH 3 、-SCH 2 CH 3 、-SCH 2 CH 2 CH 3 、-SCH 2 (CH 3 ) 2 、-CH 2 -S-CH 2 -CH 3 、-CH 2 -CH 2 、-S(=O)-CH 3 、-CH 2 -CH 2 -S(=O) 2 -CH 3 . Up to two heteroatoms may be consecutive, e.g. -CH 2 -NH-OCH 3
Unless otherwise specified, "C" is 3~6 Cycloalkyl "denotes a saturated cyclic hydrocarbon group consisting of 3 to 6 carbon atoms, being a monocyclic and bicyclic ring system, said C 3~6 Cycloalkyl radicals including C 3~5 、C 4~5 And C 5~6 Cycloalkyl groups and the like; it may be monovalent, divalent or polyvalent. C 3~6 Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like.
Unless otherwise specified, the term "3-6 membered heterocycloalkyl" by itself or in combination with other terms denotes a saturated cyclic group consisting of 3 to 6 ring atoms, 1,2,3 or 4 of which are heteroatoms independently selected from O, S and N, the remainder being carbon atoms, wherein the nitrogen atoms are optionally quaternized, the carbon, nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., C (= O), NO and S (O) p, p is 1 or 2). It includes monocyclic and bicyclic ring systems, wherein bicyclic ring systems include spiro, fused and bridged rings. Further, with respect to the "3-to 6-membered heterocycloalkyl", the heteroatom may occupy the position of the heterocycloalkyl linkage to the rest of the molecule. The 3-to 6-membered heterocycloalkyl group includes 4-to 6-membered, 5-to 6-membered, 4-membered, 5-membered, and 6-membered heterocycloalkyl groups and the like. Examples of 3-to 6-membered heterocycloalkyl include, but are not limited to, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrothienyl (including tetrahydrothien-2-yl and tetrahydrothien-3-yl, etc.), tetrahydrofuryl (including tetrahydrofuran-2-yl, etc.), tetrahydropyranyl, piperidinyl (including 1-piperidinyl, 2-piperidinyl, and 3-piperidinyl, etc.), piperazinyl (including 1-piperazinyl and 2-piperazinyl, etc.), morpholinyl (including 3-morpholinyl and 4-morpholinyl, etc.), dioxanyl, dithianyl, isoxazolidinyl, isothiazolidinyl, 1,2-oxazinyl, 1,2-thiazinyl, hexahydropyridazinyl, homopiperazinyl, homopiperidinyl, etc.
Unless otherwise specified, the term "5-6 membered heterocycloalkyl" by itself or in combination with other terms denotes a saturated cyclic group consisting of 5 to 6 ring atoms, 1,2,3 or 4 of which are heteroatoms independently selected from O, S and N, the remainder being carbon atoms, wherein the nitrogen atoms are optionally quaternized, the carbon, nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., C (= O), NO and S (O) p, p is 1 or 2). It includes monocyclic and bicyclic ring systems, wherein bicyclic ring systems include spiro, fused and bridged rings. Further, with respect to the "5-to 6-membered heterocycloalkyl", the heteroatom may occupy the position of the heterocycloalkyl linkage to the rest of the molecule. The 5-to 6-membered heterocycloalkyl group includes 5-and 6-membered heterocycloalkyl groups. Examples of 5-to 6-membered heterocycloalkyl include, but are not limited to, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, tetrahydrothienyl (including tetrahydrothien-2-yl and tetrahydrothien-3-yl, etc.), tetrahydrofuryl (including tetrahydrofuran-2-yl, etc.), tetrahydropyranyl, piperidinyl (including 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, etc.), piperazinyl (including 1-piperazinyl and 2-piperazinyl, etc.), morpholinyl (including 3-morpholinyl and 4-morpholinyl, etc.), dioxanyl, dithianyl, isoxazolidinyl, isothiazolidinyl, 1,2-oxazinyl, 1,2-thiazinyl, hexahydropyridazinyl, homopiperazinyl, homopiperidinyl, etc.
Unless otherwise specified, the term "6-membered heterocycloalkyl" by itself or in combination with other terms denotes a saturated cyclic group consisting of 6 ring atoms, 1,2,3 or 4 of which are heteroatoms independently selected from O, S and N, the remainder being carbon atoms, wherein the nitrogen atoms are optionally quaternized, the carbon, nitrogen and sulfur heteroatoms can be optionally oxidized (i.e., C (= O), NO and S (O) p, p is 1 or 2). It includes monocyclic and bicyclic ring systems, wherein bicyclic ring systems include spiro, fused and bridged rings. Further, with respect to the "6-membered heterocycloalkyl", the heteroatom may occupy the position of the attachment of the heterocycloalkyl to the rest of the molecule. Examples of 6-membered heterocycloalkyl include, but are not limited to, piperidinyl (including 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, and the like), piperazinyl (including 1-piperazinyl, 2-piperazinyl, and the like), morpholinyl (including 3-morpholinyl, 4-morpholinyl, and the like), dioxanyl, 1,2-oxazinyl, 1,2-thiazinyl, hexahydropyridazinyl, and the like.
Unless otherwise specified, the terms "5-6 membered heteroaromatic ring" and "5-6 membered heteroaryl" are used interchangeably herein, the term "5-6 membered heteroaryl" denotes a monocyclic group consisting of 5 to 6 ring atoms with a conjugated pi-electron system, 1,2,3 or 4 of which are heteroatoms independently selected from O, S and N, the remainder being carbon atoms, wherein the nitrogen atoms are optionally quaternized, and the carbon, nitrogen and sulfur heteroatoms are optionally oxidized (i.e., C (= O), NO and S (O) p, p being 1 or 2). The 5-to 6-membered heteroaryl group may be attached to the rest of the molecule through a heteroatom or a carbon atom. The 5-to 6-membered heteroaryl group includes 5-and 6-membered heteroaryl groups. Examples of the 5-to 6-membered heteroaryl group include, but are not limited to, pyrrolyl (including N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, etc.), pyrazolyl (including 2-pyrazolyl, 3-pyrazolyl, etc.), imidazolyl (including N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, etc.), oxazolyl (including 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, etc.), triazolyl (1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl, 4H-1,2,4-triazolyl, etc.), tetrazolyl, isoxazolyl (3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, etc.), thiazolyl (including 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, etc.), furyl (including 2-furyl, 3-furyl, etc.), thienyl (including 2-thienyl, 3-thienyl, etc.), pyridyl (including 2-pyridyl, pyrimidinyl, 4-pyridyl, etc.).
Unless otherwise specified, the terms "6-membered heteroaryl ring" and "6-membered heteroaryl" are used interchangeably herein, and the term "6-membered heteroaryl" denotes a monocyclic group consisting of 6 ring atoms with a conjugated pi-electron system, 1,2,3 or 4 of the ring atoms being heteroatoms independently selected from O, S and N, the remainder being carbon atoms, wherein the nitrogen atom is optionally quaternized, and the carbon, nitrogen and sulfur heteroatoms are optionally oxidized (i.e., C (= O), NO and S (O) p, p being 1 or 2). The 6-membered heteroaryl group may be attached to the rest of the molecule through a heteroatom or a carbon atom. Examples of the 6-membered heteroaryl group include, but are not limited to, pyridyl (including 2-pyridyl, 3-pyridyl, 4-pyridyl and the like), pyrazinyl or pyrimidinyl (including 2-pyrimidinyl, 4-pyrimidinyl and the like).
Unless otherwise specified, the terms "5-membered heteroaryl ring" and "5-membered heteroaryl" are used interchangeably herein, the term "5-membered heteroaryl" denotes a monocyclic group consisting of 5 ring atoms with a conjugated pi-electron system, 1,2,3 or 4 of the ring atoms being heteroatoms independently selected from O, S and N, the remainder being carbon atoms, wherein the nitrogen atom is optionally quaternized, and the carbon, nitrogen and sulfur heteroatoms are optionally oxidized (i.e., C (= O), NO and S (O) p, p being 1 or 2). The 5-membered heteroaryl group may be attached to the rest of the molecule through a heteroatom or a carbon atom. Examples of such 5-membered heteroaryl groups include, but are not limited to, pyrrolyl (including N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, and the like), pyrazolyl (including 2-pyrazolyl, 3-pyrazolyl, and the like), imidazolyl (including N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, and the like), oxazolyl (including 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, and the like), triazolyl (1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, 1H-1,2,4-triazolyl, and 4H-1,2,4-triazolyl, and the like), tetrazolyl, isoxazolyl (3-isoxazolyl, 4-isoxazolyl, and 5-isoxazolyl, and the like), thiazolyl (including 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, and the like), furyl (including 2-furyl, and 3-furyl, and the like), and the like.
The compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, examples of the present invention.
The solvent used in the present invention can be obtained commercially.
The invention employs the following abbreviations: FA represents formic acid; TFA represents trifluoroacetic acid; ACN represents acetonitrile; DIEA stands for diisopropylethylamine; p (o-tolyl) 3 Represents tris (o-methylphenyl) phosphine; TBAI represents tetrabutylammonium iodide; T3P represents 1-propylphosphoric anhydride; TBSCl represents tert-butyldimethylsilyl chloride; HATU stands for 2- (7-azabenzotriazole) -N, N' -tetramethyluronium hexafluorophosphate; HOBt represents 1-hydroxybenzotriazole; pyBOP represents benzotriazol-1-yl-oxytripyrrolidinophosphonium hexafluorophosphate; DMSO represents dimethyl sulfoxide; ATP stands for adenosine triphosphate.
The compounds are used according to the conventional naming principle in the field
Figure PCTCN2021079337-APPB-000040
The software names, and the commercial compounds are under the supplier catalog name.
Detailed Description
The present invention is described in detail below by way of examples, but is not meant to be limited to any of the disadvantages of the present invention. The compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, examples of the present invention. It will be apparent to those skilled in the art that various changes and modifications can be made in the specific embodiments of the invention without departing from the spirit and scope of the invention.
Example 1: compound 1
Figure PCTCN2021079337-APPB-000041
Step A: compound 1-1 (12g, 91.23mmol), 1-2 (14.05g, 91.23mmol), tetrakis (triphenylphosphine) palladium (5.27g, 4.56mmol), sodium carbonate (19.34g, 182.46mmol) were dissolved in 1,4-dioxane (100 mL) and water (20 mL), and the resulting mixture was replaced three times with nitrogen and stirred at 110 ℃ for 12 hours. The reaction solution was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =20/1 to 5/1) to give 1-3.MS (ESI) m/z of 124.0[ 2 ], [ M ] +H + ]。
And B, step B: compounds 1 to 3 (1g, 8.12mmol), 1 to 4 (2.8g, 8.12mmol), palladium acetate (0.18g, 0.812mmol), cesium carbonate (5.29g, 16.24mmol), tetrabutylammonium bromide (2.62g, 8.12mmol) were dissolved in N, N-dimethylacetamide (30 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 130 ℃ for 12 hours. The reaction solution was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. Separating the crude product by high performance liquid chromatography (
Figure PCTCN2021079337-APPB-000042
40g
Figure PCTCN2021079337-APPB-000043
Rapid silica gel column with 0-50% of ACN/H as eluent 2 O, flow rate 60 mL/min) to obtain 1-5.MS (ESI) m/z:387.9, [ M ] +H + ]。
Step C: a mixture of compounds 1 to 5 (0.2g, 0.516mmol), 1 to 6 (0.078g, 0.619mmol) and potassium carbonate (0.142g, 1.03mmol) in N-methylpyrrolidone (5 mL) was stirred with a microwave at 150 ℃ for 2 hours. The reaction was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. Separating the crude product by high performance liquid chromatography (
Figure PCTCN2021079337-APPB-000044
40g
Figure PCTCN2021079337-APPB-000045
Rapid silica gel column, eluent 0-50% ACN/H 2 O, flow rate 60 mL/min) to obtain 1-7.MS (ESI) m/z of 495.0[ 2 ], [ M ] +H + ]。
Step D: compounds 1-8 (0.3g, 1.34mmol) were dissolved in thionyl chloride (5 mL) and the mixture stirred at 25 ℃ for 12 h, concentrated under reduced pressure to give crude product, then dissolved with 1-7 (25mg, 50.55. Mu. Mol) in dichloromethane (5 mL), DIEA (26.13mg, 202.20. Mu. Mol) was added to the solution and the mixture stirred at 25 ℃ for 1 h. The reaction was quenched by the addition of water (50 mL) and extracted with dichloromethane (50 mL. Times.2). The combined organic phase is washed by saturated sodium chloride solution (50 mL), dried by anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain crude product 1-9.MS (ESI) m/z of 700.2[ M ] +H + ].
Step E: after compounds 1 to 9 (45mg, 64.31. Mu. Mol) were dissolved in methylene chloride (1 mL), trifluoroacetic acid (770 mg,6.75mmol,0.5 mL) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 0.5 hour. The reaction was quenched by adding aqueous sodium bicarbonate (30 mL) and extracted with ethyl acetate (50 mL. Times.2) and the mixture was combinedThe organic phase was washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was passed through (column information: phenomenex Synergi C18 150 x 25 x 10 μm; mobile phase: [ Water (0.225% by weight FA) -ACN)](ii) a 17% -41% of ACN and 8 minutes) to obtain the compound 1. 1 H NMR(400MHz,DMSO-d 6 )δ=10.38(s,1H),10.00(s,1H),8.53(d,J=2.0Hz,1H),8.48(d,J=5.6Hz,1H),7.88(dd,J=13.2,2.4Hz,1H),7.75(dd,J=8.0,2.0Hz,1H),7.72-7.57(m,5H),7.50(dd,J=8.8,1.2Hz,1H),7.39-7.33(m,1H),7.19(d,J=2.4Hz,1H),7.18-7.13(m,2H),6.80(dd,J=5.6,2.4Hz,1H),3.76(s,2H)3.41(br s,2H),3.24(s,3H),2.69-2.64(m,2H),1.47(br d,J=10.0Hz,4H).MS(ESI)m/z:600.1[M+H + ].
Example 2: compound 2
Figure PCTCN2021079337-APPB-000046
Figure PCTCN2021079337-APPB-000047
Step A: compound 2-1 (2g, 13.42mmol), 2-2 (2.11g, 13.42mmol), and potassium carbonate (3.71g, 26.85mmol) were dissolved in N, N-dimethylformamide (20 mL), and the resulting mixture was stirred at 80 ℃ for 3 hours. The reaction solution was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 2-3.MS (ESI) m/z of 269.7[ 2 ], [ M ] +H + ].
And B: compounds 2-3 (1.9g, 7.05mmol), 1-2 (1.19g, 7.75mmol), tetrakis (triphenylphosphine) palladium (814.33mg, 704.71. Mu. Mol), sodium carbonate (1.49g, 14.09mmol) were dissolved in 1,4-dioxane (20 mL) and water (4 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 80 ℃ for 3 hours. The reaction was quenched by addition of water (100 mL) and extracted with ethyl acetate (100 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solutionWashed (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =20/1 to 5/1) to give 2-4.MS (ESI) m/z of 261.8[ 2 ], [ M ] +H + ].
And C: compounds 2-4 (0.5g, 1.91mmol), 1-4 (0.66g, 1.91mmol), palladium acetate (42.97mg, 0.191mmol), triethylamine (387.39mg, 3.83mmol), tetrabutylammonium bromide (617.07mg, 1.91mmol), P (o-tolyl) 3 (582.61mg, 1.91mmol) was dissolved in toluene (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 90 ℃ for 12 hours. The reaction solution was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =20/1 to 5/1) to give 2-5.MS (ESI) m/z:526.1[ 2 ] M + H + ]。
Step D: compound 2-5 (90mg, 171.26mmol) was dissolved in ethanol (10 mL), followed by addition of iron powder (191.28mg, 3.43mmol), ammonium chloride (18.32mg, 0.342mmol) and stirring of the resulting mixture at 100 ℃ for 12 hours. The reaction solution was filtered, concentrated under reduced pressure to give a crude product, which was extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 2-6.MS (ESI) m/z:496.1, [ m ] +H + ]。
Step E: compounds 1-8 (0.3g, 1.34mmol) were dissolved in thionyl chloride (5 mL) and the resulting mixture was stirred at 25 ℃ for 12 hours. Concentration under reduced pressure gave crude acid chloride, 2-6 (80mg, 161.44. Mu. Mol) and crude acid chloride (78mg, 322.88. Mu. Mol) were dissolved in tetrahydrofuran (5 mL), DIEA (83.46mg, 645.75. Mu. Mol) was added to the solution, and the resulting mixture was stirred at 25 ℃ for 1 hour. The reaction solution was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product which was then separated by reverse phase column (
Figure PCTCN2021079337-APPB-000048
40g
Figure PCTCN2021079337-APPB-000049
Flash column of silica gel, eluent 0-50% ACN/0.1% 2 O, flow rate 60 mL/min) to obtain 2-7.MS (ESI) m/z:701.2[ 2 ], [ M + H ] + ]。
Step F: after compounds 2 to 7 (72mg, 102.75. Mu. Mol) were dissolved in methylene chloride (1 mL), trifluoroacetic acid (1.23g, 10.79mmol,0.8 mL) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 0.5 hour. The reaction was quenched by adding aqueous sodium bicarbonate (30 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was separated by HPLC (column information: phenomenex Synergi C18 150X 25X 10 μm; mobile phase: [ water (0.225% FA) -ACN%](ii) a 20 percent to 40 percent of ACN and 10 minutes) to obtain the formate of the compound 2. 1 H NMR(400MHz,DMSO-d 6 )δ=10.34(s,1H),10.03(s,1H),8.72(s,1H),8.61(d,J=2.0Hz,1H),8.19(s,1H),7.95(d,J=15.6Hz,1H),7.85-7.78(m,2H),7.72-7.63(m,4H),7.46-7.44(m,2H),7.37-7.32(m,1H),7.16(t,J=8.88Hz,2H),3.82(s,2H),3.43(t,J=8.8Hz,2H),3.25(s,3H),2.71(t,J=5.6Hz,2H),1.47(br d,J=2.8Hz,4H);MS(ESI)m/z:601.1[M+H + ]
Example 3: compound 3
Figure PCTCN2021079337-APPB-000050
Figure PCTCN2021079337-APPB-000051
Step A: compound 3-1 (3.4g, 23.13mmol), 3-2 (3.18g, 25.45mmol), cesium carbonate (11.31g, 34.70mmol), TBAI (1.71g, 4.63mmol) were dissolved in N, N-bisMethyl acetamide (15 mL) and the resulting mixture was stirred at 90 ℃ for 12 hours. The reaction was quenched by the addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude compound 3-3.MS (ESI) m/z:191.1[ 2 ] M + H + ].
And B: compound 3-3 (4.4g, 23.03mmol), TBSCl (4.17g, 27.64mmol), imidazole (3.14g, 46.07mmol) were dissolved in N, N-dimethylacetamide (15 mL), and the resulting mixture was stirred at 25 ℃ for 12 hours. The reaction was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL × 2), the combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude product, which was separated by reverse phase column (waters Xbridge BEH C18 250 × 50mm 10 μm; mobile phase [ water (0.05% ammonia v/v) -ACN](ii) a 65-80 percent of ACN and 20 minutes) to obtain 3-4.MS (ESI) m/z:304.9[ 2 ], [ M + H ] + ].
And C: compound 1-1 (10g, 76.03mmol), 2-2 (10g, 76.03mmol), potassium carbonate (21.01g, 152.05mmol) were dissolved in N, N-dimethylacetamide (20 mL), and the resulting mixture was stirred at 100 ℃ for 3 hours. The reaction solution was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =20/1,5/1) to give compound 3-5.MS (ESI) m/z:268.7[ 2 ], [ M + H ] + ].
Step D: compounds 3-5 (5g, 18.61mmol), 1-2 (3.44g, 22.34mmol), tetrakis (triphenylphosphine) palladium (2.15g, 1.86mmol), sodium carbonate (3.95g, 37.23mmol) were dissolved in dioxane (50 mL) and water (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 80 ℃ for 3 hours. The reaction was quenched by the addition of water (100 mL) and extracted with ethyl acetate (100 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =20/1,5/1) to give 3-6.MS (ESI) m/z:260.8[ 2 ], [ M ] +H + ].
Step E: compounds No. 3-6 (255.71mg, 982.69mmol), no. 3-4 (300mg, 982.69mmol), palladium acetate (22.06mg, 98.27. Mu. Mol), triethylamine (497.19mg, 4.91mmol), lithium bromide (256.02mg, 2.95mmol), P (o-tolyl) 3 (299.10mg, 982.69mmol) was dissolved in toluene (10 mL), and the resulting mixture was purged with nitrogen three times and stirred at 110 ℃ for 12 hours. The reaction solution was quenched by addition of water (100 mL) and extracted with ethyl acetate (100 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (25 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. Separating the column information of the crude product by reverse phase column chromatography, waters Xbridge BEH C18 250 x 50mm x 10 μm; mobile phase [ water (0.05% ammonia v/v) -ACN](ii) a 55 to 85 percent of ACN and purifying for 20 minutes to obtain 3 to 7.MS (ESI) m/z of 485.0[ 2 ] M + H + ].
Step F: compound 3-7 (200mg, 412.72. Mu. Mol) was dissolved in ethanol 10 mL) and water (2 mL), then iron powder (230.48mg, 4.13mmol), ammonium chloride (110.38mg, 2.06mmol) were added and the resulting mixture was stirred at 90 ℃ for 6 hours. The reaction mixture was filtered and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 3-8.MS (ESI) m/z:455.0[ 2 ], [ M ] +H + ].
Step G: compound 3-8 (90mg, 197.97. Mu. Mol) was dissolved in dichloromethane (5 mL) and 1-8 (88.3mg, 395.94. Mu. Mol), DIEA (127.93mg, 989.86. Mu. Mol) and T3P (377.94mg, 593.91. Mu. Mol) were added to the solution at 25 ℃ and the resulting mixture was stirred at 25 ℃ for 12 hours. Water (50 mL) was added to the reaction mixture and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude 3-9.MS (ESI) m/z of 660.2[ 2 ], [ M + H ] + ].
Step H: compound 3-9 (65mg, 98.52. Mu. Mol) was dissolved in methylene chloride (2 mL), and trifluoroacetic acid (1.54g, 13.51mmol) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 0.5 hour. The reaction solution is decompressed and concentrated to obtain a crude product. The crude product was passed through (Phenomenex Synergi C18 150 x 25 x 10 μm; mobile phase: [ water (0.225% FA) -ACN%](ii) a 17 percent to 50 percent of ACN and 10 minutes) to obtain the formate of the compound 3. 1H NMR (400 MHz),DMSO-d6)δ=10.39(s,1H),10.01(s,1H),8.40(d,J=5.6Hz,1H),8.22(s,1H),7.97(s,1H),7.87(dd,J=2.4,13.2Hz,1H),7.77(s,1H),7.70-7.60(m,2H),7.54(s,1H),7.50-7.46(m,1H),7.39-7.31(m,1H),7.21-7.11(m,2H),6.92(d,J=16.0Hz,1H),6.86(d,J=2.4Hz,1H),6.74(dd,J=2.4,5.6Hz,1H),4.12(t,J=5.6Hz,2H),3.72(t,J=5.6Hz,2H),1.47(br d,J=7.2Hz,4H);MS(ESI)m/z:546.1[M+H + ]。
Example 4: compound 4
Figure PCTCN2021079337-APPB-000052
Figure PCTCN2021079337-APPB-000053
Step A: compound 3-1 (1g, 6.80mmol), 4-1 (1.46g, 10.21mmol), cesium carbonate (3.33g, 10.21mmol) were dissolved in N, N-dimethylformamide (8 mL), and the resulting mixture was stirred at 60 ℃ for 1 hour. The reaction was quenched by the addition of water (20 mL) and extracted with ethyl acetate (30 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (50 mL. Times.3), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give compound 4-2, which was used directly in the next step.
And B, step B: compound 4-2 (1.43g, 6.83mmol), compound 4-3 (1.54g, 20.48mmol) were dissolved in DMSO (5 mL), and the resulting mixture was stirred at 60 ℃ for 5 hours. The reaction mixture was diluted with 1mol/L hydrochloric acid (10 mL) and ethyl acetate (20 mL), separated, the aqueous phase was washed with ethyl acetate (30 mL), the organic phase was discarded, the aqueous phase was adjusted to pH =8 using 1mol/L sodium hydroxide and extracted with ethyl acetate (30 mL × 3), the combined organic phases were washed with a saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 4-4.MS (ESI) m/z:247.8[ 2 ] M + H + ].
Step C: compound 4-4 (900mg, 3.63mmol) was dissolved in tetrahydrofuran (10 mL), di-tert-butyl dicarbonate (949.97mg, 4.35mmol) was added, and the resulting mixture was stirred at 20 ℃ for 12 hours. Concentrating under reduced pressure to obtain crude product. The crude product is separated and purified by silica gel column (ethyl acetate/petroleum ether =0% -50%) to obtain the compound 4-5.
And E, step E: compounds 3-6 (0.4g, 1.54mmol), 4-5 (1.07g, 3.07mmol), palladium acetate (69.02mg, 307.43. Mu. Mol), triethylamine (933.27mg, 9.22mmol), lithium bromide (534.01mg, 6.15mmol), P (o-tolyl) 3 (467.87mg, 1.54mmol) was dissolved in N, N-dimethylacetamide (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 125 ℃ for 12 hours. To the reaction mixture were added water (40 mL) and ethyl acetate (40 mL), the mixture was separated, the aqueous phase was extracted with ethyl acetate (40 mL. Times.3), and the combined organic phases were washed with a saturated sodium chloride solution (50 mL. Times.3), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a crude product. The crude product is purified by silica gel column separation (ethyl acetate/petroleum ether =20% -80%) to obtain 4-6.MS (ESI) m/z:528.3[ 2 ] M + H + ].
Step F: compound 4-6 (150mg, 284.34. Mu. Mol) was dissolved in ethanol (3 mL), followed by addition of iron powder (158.79mg, 2.84mmol), a saturated ammonium chloride solution (0.3 mL), and the resulting mixture was stirred at 80 ℃ for 2 hours. After filtration, the filtrate was collected, concentrated to remove the organic solvent, and then diluted with ethyl acetate (20 mL) and water (20 mL), the layers were separated, and the aqueous layer was extracted with ethyl acetate (30 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 4-7, which was used directly in the next step. MS (ESI) m/z:498.3[ 2 ] M + H + ].
Step G: compounds 1 to 8 (72.81mg, 326.20. Mu. Mol) were dissolved in methylene chloride (5 mL), oxalyl chloride (159.25mg, 1.25mmol) and N, N-dimethylformamide (1.83mg, 25.09. Mu. Mol) were added at 0 ℃ and reacted at 25 ℃ for half an hour. The solution was concentrated to give the corresponding acid chloride and dissolved in dichloromethane (5 mL), 4-7 (124.85mg, 250.92. Mu. Mol) and pyridine (99.24mg, 1.25mmol) were added to the solution at 0 deg.C, and the resulting mixture was stirred at 25 deg.C for half an hour. Water (20 mL) was added to the reaction mixture, and the aqueous phase was extracted with dichloromethane (20 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 4-8, which was used directly in the next step. MS (ESI) m/z:703.2[ 2 ], [ M + H ] + ].
Step H: compound 4-8 (150mg, 213.45. Mu. Mol) was dissolved in methylene chloride (5 mL), and trifluoroacetic acid (1 mL) was added to the reaction solution, and the resulting mixture was stirred at 20 ℃ for 1 hour. And concentrating the reaction solution under reduced pressure to obtain a crude product. The crude product was purified by preparative high performance liquid chromatography (column: waters Xbridge 150 × 25mm × 5 μm; mobile phase: [ water (0.05% ammonia v/v) -ACN](ii) a 38 percent to 68 percent of B percent for 10 minutes) to obtain a compound 4. 1 H NMR(400MHz,DMSO-d6)δ=10.39(s,1H),10.01(s,1H),8.40(d,J=5.6Hz,1H),7.99(s,1H),7.88(dd,J=2.4,13.2Hz,1H),7.77(s,1H),7.65(dd,J=5.2,9.1Hz,2H),7.54-7.47(m,2H),7.35(t,J=9.2Hz,1H),7.16(t,J=8.8Hz,2H),6.92(d,J=16.0Hz,1H),6.86(d,J=2.4Hz,1H),6.74(dd,J=2.4,5.6Hz,1H),4.14(t,J=6.0Hz,2H),3.35(t,J=5.6Hz,2H),3.22(s,3H),2.92(t,J=6.0Hz,2H),2.65(t,J=5.6Hz,2H),1.54-1.41(m,4H);MS(ESI)m/z:603.2[M+H + ]。
Example 5: compound 5
Figure PCTCN2021079337-APPB-000054
Step A: compound 5-1 (1.6 g, 8.25mmol) was dissolved in acetonitrile (20 mL), potassium carbonate (2.28g, 16.50mmol), potassium iodide (136.93mg, 824.86. Mu. Mol) and 5-2 (1.94g, 8.66mmol) were added, and the resulting mixture was stirred at 80 ℃ for 12 hours. Water (100 mL) was added to the reaction mixture to precipitate a white solid, which was filtered and the filter cake was collected and concentrated under reduced pressure to give compound 5-3.MS (ESI) m/z:338.2[ 2 ], [ M ] +H + ].
And B, step B: compounds 3-6 (0.4 g, 1.54mmol), 5-3 (1.04g, 3.08mmol), palladium acetate (69.15mg, 308.00. Mu. Mol), triethylamine (935.00mg, 9.24mmol), lithium bromide (401.25mg, 4.62mmol), P (o-tolyl) 3 (468.73mg, 1.54mmol) was dissolved in N, N-dimethylacetamide (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 125 ℃ for 36 hours. Water (40 mL) and ethyl acetate (40 mL) were added to the reaction mixture, the mixture was separated, the aqueous phase was extracted with ethyl acetate (40 mL. Times.3), the combined organic phases were washed with a saturated sodium chloride solution (50 mL. Times.3), and anhydrous sodium sulfate was addedDrying, filtering and concentrating under reduced pressure to obtain a crude product. The crude product is separated and purified by a silica gel column (ethyl acetate/petroleum ether =20% -80%) to obtain 5-4.MS (ESI) m/z:470.0[ 2 ], [ M + H ] + ].
And C: compound 5-4 (370mg, 788.13. Mu. Mol) was dissolved in ethanol (6 mL), then iron powder (440.17mg, 7.88mmol) and a saturated ammonium chloride solution (0.5 mL) were added, and the resulting mixture was stirred at 80 ℃ for 2 hours. After filtration, the filtrate was collected, concentrated to remove the organic solvent, and then diluted with ethyl acetate (20 mL) and water (20 mL), the mixture was separated, and the aqueous phase was extracted with ethyl acetate (30 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 5-5, and the crude product was used directly in the next step. MS (ESI) m/z of 440.3[ 2 ], [ M + H ] + ].
Step D: compounds 1-8 (211.27mg, 946.57. Mu. Mol) were dissolved in dichloromethane (10 mL), oxalyl chloride (462.11mg, 3.64mmol, 318.69. Mu.L) and N, N-dimethylformamide (5.32mg, 72.81. Mu. Mol) were added at 0 ℃ and reacted at 25 ℃ for half an hour. The solution was concentrated to give the corresponding acid chloride and dissolved in dichloromethane (5 mL), 5-5 (320mg, 728.13. Mu. Mol) and pyridine (287.98mg, 3.64mmol) were added to the solution at 0 deg.C and the resulting mixture was stirred at 25 deg.C for half an hour. Water (20 mL) was added to the reaction mixture, and the aqueous phase was extracted with dichloromethane (20 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 5-6, and the crude product was used directly in the next step. MS (ESI) m/z:645.2[ 2 ], [ M + H ] + ].
Step E: compound 5-6 (450mg, 698.04. Mu. Mol) was dissolved in methylene chloride (20 mL), and trifluoroacetic acid (2 mL) was added to the reaction solution, and the resulting mixture was stirred at 20 ℃ for 3 hours. The reaction solution is decompressed and concentrated to obtain a crude product. The crude product was purified by preparative high performance liquid chromatography (column: phenomenex Synergi C18: 25X 10 μm; mobile phase: [ water (0.225% FA) -ACN%](ii) a 8 percent to 38 percent of the compound B) for 10 minutes) to obtain the formate of the compound 5. 1 H NMR(400MHz,DMSO-d 6 )δ=10.40(br d,J=2.4Hz,1H),10.01(br s,1H),8.40(d,J=5.6Hz,1H),8.30(br s,1H),8.02(s,1H),7.91-7.81(m,2H),7.65(dd,J=5.0,9.2Hz,2H),7.58-7.44(m,2H),7.35(t,J=9.2Hz,1H),7.16(t,J=8.8Hz,2H),6.95(d,J=16.0Hz,1H),6.87(d,J=2.4Hz,1H),6.75(dd,J=2.4,5.6Hz,1H),4.22(br t,J=6.0Hz,2H),3.16-3.03(m,2H),1.55-1.38(m,4H);MS(ESI)m/z:545.3[M+H + ]。
Example 6: compound 6
Figure PCTCN2021079337-APPB-000055
Figure PCTCN2021079337-APPB-000056
Step A: compound 3-1 (7g, 47.63mmol) was dissolved in tetrahydrofuran (100 mL), sodium hydride (2.48g, 61.92mmol,60% content) was added portionwise at 0 ℃ and stirred at 0 ℃ for 30 minutes, 6-1 (9.93g, 59.53mmol) was added and the resulting mixture stirred at 25 ℃ for 12 hours. The reaction was quenched by careful addition of saturated ammonium chloride (50 mL) at 0 deg.C, separated, the aqueous phase extracted with ethyl acetate (50 mL. Times.3), the organic phases combined, washed with saturated sodium chloride (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give compound 6-2.
And B: mixing 6-2 (1.28g, 4.61mmol), 3-6 (1g, 3.84mmol), palladium acetate (86.28mg, 384.29. Mu. Mol), triethylamine (1.94g, 19.21mmol), lithium bromide (1.00g, 11.53mmol), P (o-tolyl) 3 (1.17g, 3.84mmol) was dissolved in N, N-dimethylacetamide (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 125 ℃ for 36 hours. Water (50 mL) and ethyl acetate (50 mL) were added to the reaction mixture, the layers were separated, the aqueous layer was extracted with ethyl acetate (50 mL. Times.3), the combined organic layers were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether/ethyl acetate =10/1 to 2/1) to give 6-3.MS (ESI) m/z:457.2[ 2 ], [ M ] +H + ].
And C: compound 6-3 (580mg, 1.27mmol) was dissolved in ethanol (7 mL), and then iron powder (709.47mg, 12.70mmol) and saturated ammonium chloride solution were addedLiquid (0.7 mL), and the resulting mixture was stirred at 80 ℃ for 10 h. After filtration, the filtrate was collected, concentrated to remove the organic solvent, and then diluted with ethyl acetate (30 mL) and water (30 mL), the layers were separated, and the aqueous layer was extracted with ethyl acetate (50 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 6-4, which was used directly in the next step. MS (ESI) m/z:427.1[ 2 ] M + H + ].
Step D: compound 1 to 8 (285.70mg, 1.28mmol) was dissolved in methylene chloride (5 mL), oxalyl chloride (624.87mg, 4.92mmol) and N, N-dimethylformamide (7.20mg, 98.46. Mu. Mol) were added at 0 ℃ and reacted at 25 ℃ for half an hour. The solution was concentrated to give the corresponding acid chloride and dissolved in dichloromethane (5 mL), 6-4 (420mg, 984.63. Mu. Mol) and pyridine (389.42mg, 4.92mmol) were added to the solution at 0 deg.C and the resulting mixture was stirred at 25 deg.C for half an hour. Water (30 mL) and methylene chloride (30 mL) were added to the reaction mixture, and the mixture was separated, and the aqueous phase was extracted with methylene chloride (50 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 6-5, which was used directly in the next step. MS (ESI) m/z of 632.3[ 2 ], M + H ], + ].
step E: compound 6-5 (700mg, 1.11mmol) was dissolved in methylene chloride (14 mL), and trifluoroacetic acid (7 mL) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 2 hours. The reaction solution is decompressed and concentrated to obtain a crude product. The crude product was purified by preparative high performance liquid chromatography (column: waters Xbridge BEH C18 25mm 5 μm; mobile phase: [ water (0.05% aqueous ammonia v/v) -ACN)](ii) a 36 percent to 66 percent of B percent and 10 minutes) to obtain a compound 6. 1 H NMR(400MHz,DMSO-d 6 )δ=12.94(br s,1H),10.38(s,1H),10.00(s,1H),8.40(d,J=5.6Hz,1H),8.09-7.76(m,3H),7.65(dd,J=5.2,9.2Hz,2H),7.56(d,J=16.0Hz,1H),7.49(dd,J=1.2,8.8Hz,1H),7.35(t,J=9.2Hz,1H),7.16(t,J=8.8Hz,2H),6.94(d,J=16.0Hz,1H),6.84(d,J=2.4Hz,1H),6.75(dd,J=2.4,5.6Hz,1H),1.53-1.43(m,4H);MS(ESI)m/z:502.3[M+H + ]。
Example 7: compound 7
Figure PCTCN2021079337-APPB-000057
Step A: compound 1-1 (3.5g, 26.61mmol), 7-1 (3.70g, 26.61mmol), potassium carbonate (7.36g, 53.22mmol) were dissolved in N, N-dimethylformamide (10 mL), and the resulting mixture was stirred at 100 ℃ for 12 hours. The reaction mixture was quenched by addition of water (100 mL) and extracted with ethyl acetate (100 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =20/1 to 5/1) to give 7-2.MS (ESI) m/z of 250.8[ 2 ], M + H + ].
And B: compound 7-2 (4.5g, 17.95mmol), 1-2 (3.32g, 21.55mmo), tetrakis (triphenylphosphine) palladium (2.07g, 1.80mmol), sodium carbonate (3.81g, 35.91mmol) were dissolved in 1,4-dioxane (50 mL) and water (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 80 ℃ for 3 hours. The reaction solution was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (25 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =1/0 to 1/2) to give 7-3.MS (ESI) m/z:243.0[ 2 ], [ M ] +H + ].
And C: compounds 7-3 (1g, 4.13mmol), 3-4 (1.26g, 4.13mmol), palladium acetate (92.68mg, 412.83. Mu. Mol), triethylamine (2.09g, 20.64mmol), lithium bromide (1.08g, 12.38mmol) were dissolved in toluene (15 mL), and the resulting mixture was replaced three times with nitrogen and stirred at 120 ℃ for 12 hours. Water (100 mL) was added to the reaction solution, and the mixture was extracted with ethyl acetate (100 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (25 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =1/0 to 1/2) to give 7-4.MS (ESI) m/z:467.0[ 2 ], [ M + H ] + ]。
Step D: compound 7-4 (1.5g, 3.21mmol) was dissolved in ethanol (10 mL) and water (2 mL), and then iron powder (1.80g, 32.15mmol), ammonium chloride (859.78mg, 16.07mmol) were added. The resulting mixture was at 90 deg.CStirred for 2 hours. The reaction mixture was filtered and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 7-5.MS (ESI) m/z:437.3[ 2 ], [ M + H ] + ]。
Step E: compounds 1 to 8 (500mg, 1.15mmol) were dissolved in methylene chloride (10 mL) and then 7 to 5 (330.09mg, 2.29mmol), T3P (2.91g, 4.58mmol,2.72mL,50% purity), and DIEA (888.00mg, 6.87mmol, 1.20mL) were added and reacted at 25 ℃ for 12 hours. Water (50 mL) was added to the reaction solution, and the mixture was extracted with methylene chloride (25 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (25 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 7-6.MS (ESI) m/z:563.1[ 2 ] M + H + ]。
Step F: compound 7-6 (550mg, 977.38. Mu. Mol) was dissolved in tetrahydrofuran (2 mL) and water (0.5 mL), then lithium hydroxide monohydrate (82.02mg, 1.95mmol) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 1 hour. The reaction mixture was concentrated under reduced pressure, water (10 mL) was added, the pH was adjusted to 6 with 1mol/L hydrochloric acid, and the mixture was extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 7-7.MS (ESI) m/z:549.1[ 2 ] M + H + ]。
Step G: compound 7-7 (536.29mg, 977.38. Mu. Mol) was dissolved in tetrahydrofuran (5 mL) and 7-8 (108.60mg, 977.38. Mu. Mol), T, was added 3 P (1.24g, 1.95mmol,50% ethyl acetate solution) and DIEA (505.26mg, 3.91mmol) were reacted at 25 ℃ for 12 hours. Water (50 mL) was added to the reaction solution, and the mixture was extracted with ethyl acetate (25 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (25 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 7-9.MS (ESI) m/z 642.1[ 2 ] M + H + ]。
Step H: compound 7-9 (480mg, 747.89. Mu. Mol) was dissolved in methylene chloride (2 mL), and trifluoroacetic acid (4.26g, 37.39mmol) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for half an hour. The reaction solution is concentrated under reduced pressure to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (column chromatography: phenomenex Luna C18 75 x 30 x 3 μm; mobile phase: [ water (0.225% FA) -A%CN](ii) a 13 percent to 35 percent of ACN) for 10 minutes) to obtain a compound 7. 1 H NMR(400MHz,DMSO-d 6 )δ=10.17(s,1H),10.04(s,1H),8.38(d,J=5.6Hz,1H),7.97(s,1H),7.77(s,1H),7.73(d,J=8.8Hz,2H),7.69-7.61(m,2H),7.50(d,J=16.0Hz,1H),7.22-7.07(m,4H),6.90(d,J=16.0Hz,1H),6.84(d,J=2.4Hz,1H),6.71(dd,J=2.4,5.6Hz,1H),5.12-4.78(m,1H),4.12(t,J=5.6Hz,2H),3.73(q,J=5.2Hz,2H),1.47(s,4H);MS(ESI)m/z:528.1[M+H + ]。
Example 8: compound 8
Figure PCTCN2021079337-APPB-000058
Step A: compound 3-1 (3g, 20.41mmol) was dissolved in N, N-dimethylformamide (50 mL), and 8-1 (3.12g, 22.45mmol), cesium carbonate (9.98g, 30.62mmol) and TBAI (1.51g, 4.08mmol) were added to the reaction system, and the resulting mixture was stirred at 90 ℃ for 4 hours. The reaction solution was quenched by addition of water (50 mL) and extracted with dichloromethane (25 mL. Times.2). The combined organic phases were washed twice with saturated sodium chloride solution (25 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was isolated by preparative high performance liquid chromatography (chromatography column: waters Xbridge BEH C18. About.50mm. About.10 μm; mobile phase [ water (0.05% ammonia v/v) -ACN](ii) a 15 percent to 40 percent of B percent, 20 minutes) to obtain 8 to 2.MS (ESI) m/z:206.8[ 2 ], [ M + H ] + ]。
And B: compounds 8-2 (1g, 4.88mmol), 3-6 (1.27g, 4.88mmol), palladium acetate (109.49mg, 487.68. Mu. Mol), P (o-tolyl) 3 (1.48g, 4.88mmol), triethylamine ((2.47g, 24.38mmol, 3.39mL) and lithium bromide (1.27g, 14.63mmol) were dissolved in toluene (10 mL), the resulting mixture was replaced with nitrogen three times and stirred at 110 ℃ for 12 hours. The reaction solution was quenched by addition of water (100 mL) and extracted with ethyl acetate (100 mL. Times.2.) the combined organic phases were washed twice with saturated sodium chloride solution (25 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude product which was separated by silica gel column chromatography (petroleum ether: ethyl acetate = 20; 1-0:1) to give 8-3.MS (ESI) m/z:384.9[ M ] H + H ] + ]。
And C: compound 8-3 (230mg, 598.40. Mu. Mol) was dissolved in water (2 mL) and ethanol (10 mL), and iron powder (334.20mg, 5.98mmol) and ammonium chloride (160.04mg, 2.99mmol) were added to the reaction system, and the resulting mixture was stirred at 90 ℃ for 3 hours. The reaction solution was filtered through celite, the filtrate was extracted with ethyl acetate (50 mL. Times.2), the combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 8-4.MS (ESI) m/z of 355.1[ 2 ], M + H ] + ]。
Step D: compound 8-4 (100mg, 282.19. Mu. Mol) was dissolved in tetrahydrofuran (5 mL), and 1-8 (125.97mg, 564.37. Mu. Mol), T3P (538.71mg, 846.56. Mu. Mol, 503.47. Mu.L, 50% pure write) and DIEA (182.35mg, 1.41mmol, 245.75. Mu.L) were added to the reaction system, and the resulting mixture was stirred at 25 ℃ for 12 hours. The reaction was quenched by the addition of water (50 mL) and extracted with dichloromethane (25 mL. Times.2). The combined organic phases were washed twice with saturated sodium chloride solution (25 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by high performance liquid chromatography (column information: phenomenex Gemini-NX C18 75X 30mm 3 μm; mobile phase: water (0.1% TFA) -ACN%](ii) a 30-40 percent of B percent and 7 minutes) to obtain a compound 8. 1 H NMR(400MHz,DMSO-d 6 )δ=10.45(s,1H),9.97(s,1H),8.54(d,J=6.4Hz,1H),8.06(s,1H),7.92(br dd,J=1.6,13.2Hz,1H),7.84(s,1H),7.70(br d,J=16.4Hz,1H),7.63(br dd,J=5.2,8.8Hz,2H),7.52(br s,1H),7.42(s,1H),7.34(br s,1H),7.16(br t,J=8.8Hz,2H),7.11-7.05(m,1H),6.98(d,J=16.4Hz,1H),4.28(br t,J=5.2Hz,2H),3.69(br d,J=5.2Hz,2H),3.25-3.21(m,3H),1.48(br d,J=13.2Hz,4H);MS(ESI)m/z:560.1[M+H + ].
Example 9: compound 9
Figure PCTCN2021079337-APPB-000059
Step A: compound 9-1 (4.75g, 30.21mmol), 2-2 (4.5g, 30.21mmol), and potassium carbonate (8.35g, 60.41mmol) were dissolved in N, N-dimethylformamide (100 mL), and the mixture was obtainedThe mixture was stirred at 80 ℃ for 12 hours. The reaction mixture was quenched by addition of water (300 mL) and extracted with ethyl acetate (100 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (200 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 9-2.MS (ESI) m/z:270.1[ 2 ] M + H + ].
And B: compounds 9-2 (7g, 25.96mmol), 1-2 (4.80g, 31.16mmol), tetrakis (triphenylphosphine) palladium (3.00g, 2.60mmol), sodium carbonate (5.50g, 51.93mmol) were dissolved in 1,4-dioxane (120 mL) and water (24 mL), and the resulting mixture was replaced three times with nitrogen and stirred at 84 ℃ for 12 hours. The reaction solution was quenched by addition of water (50 mL) and extracted with ethyl acetate (100 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (100 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =1/0 to 3/5) to give 9-3.MS (ESI) m/z:262.1[ 2 ], [ M ] +H + ].
Step C: compounds 9-3 (200mg, 765.67. Mu. Mol,;), 9-4 (539.47mg, 1.53mmol), palladium acetate (17.19mg, 76.57. Mu. Mol),; triethylamine (387.39mg, 3.83mmol, 532.86. Mu.L), lithium bromide (199.48mg, 2.30mmol), P (o-tolyl) 3 (233.04mg, 765.67. Mu. Mol) was dissolved in N, N-dimethylacetamide (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 70 ℃ for 12 hours. The reaction mixture was diluted with quenched ethyl acetate (30 mL) by adding water (30 mL), filtered through celite, and the filtrate was extracted with ethyl acetate (30 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (30 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =1/0 to 1/2) to give 9-5.MS (ESI) m/z:486.1[ 2 ], [ M ] +H + ]。
Step D: compound 9-5 (200mg, 411.88. Mu. Mol) was dissolved in ethanol (5 mL) and water (1 mL), then iron powder (230.01mg, 4.12mmol), ammonium chloride (110.16mg, 2.06mmol) were added and the resulting mixture was stirred at 90 ℃ for 1 hour. The reaction mixture was filtered and extracted with ethyl acetate (20 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 9-6.MS (ESI) m/z:456.1[ 2 ], [ M + H ] + ]。
And E, step E: compound 1-8 (82.79mg, 370.94. Mu. Mol) was dissolved in dichloromethane (3 mL) at zero degrees, oxalyl chloride (181.08mg, 1.43mmol), N-dimethylformamide ((2.09mg, 28.53. Mu. Mol) were added dropwise, and the resulting mixture was stirred at 25 ℃ for half an hour, concentrated under reduced pressure to give a crude acid chloride, which was then dissolved in dichloromethane (3 mL), pyridine (112.85mg, 1.43mmol) was added to the solution at zero degrees, 9-6 (130mg, 285.34. Mu. Mol) was dissolved in dichloromethane (3 mL) and added dropwise to the mixed solution, and the resulting mixture was stirred at 0 ℃ for ten minutes, then stirred at 25 ℃ for half an hour, water (10 mL) was added to the reaction solution to quench the reaction solution and dichloromethane (10 mL. Times.2) was extracted, the combined organic phases were washed with a saturated sodium chloride solution (20 mL. Times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 9-7.MS (ESI) m/z: 661M H2 + ]。
Step F: compound 9-7 (170mg, 257.27. Mu. Mol) was dissolved in methylene chloride (2 mL), and trifluoroacetic acid (2 mL) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 12 hours. The reaction solution is concentrated under reduced pressure to obtain a crude product. The crude product was separated by HPLC (column information: phenomenex luna C18: 40: 15 μm; mobile phase: [ water (0.1% TFA) -ACN%](ii) a 30% -60% of ACN% and 10 minutes) to obtain the trifluoroacetate of the compound 9. 1 H NMR(400MHz,DMSO-d 6 )δ=10.33(s,1H),10.01(s,1H),8.63(d,J=5.6Hz,1H),8.07(s,1H),7.87-7.77(m,2H),7.68-7.57(m,3H),7.49-7.43(m,1H),7.38-7.31(m,1H),7.15(t,J=8.8Hz,2H),6.94(d,J=6.0Hz,1H),6.72(d,J=15.6Hz,1H),4.13-4.09(m,2H),3.71(t,J=5.6Hz,2H),1.47(br d,J=4.0Hz,4H);MS(ESI)m/z:547.1[M+H + ]。
Example 10: compound 10
Figure PCTCN2021079337-APPB-000060
Figure PCTCN2021079337-APPB-000061
Step A: compound 10-1 (2g, 12.20mmol) was dissolved in N, N-dimethylformamide (20 mL), and 10-2 (3.88g, 24.39mmol) and potassium carbonate (7.95g, 24.39mmol) were added to the reaction, and the resulting mixture was stirred at 70 ℃ for 12 hours. Water (20 mL) was added to the reaction solution, and the mixture was extracted with ethyl acetate (20 mL. Times.2). The combined organic phases were washed twice with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product, which was isolated by silica gel column chromatography (petroleum ether: ethyl acetate =1:0-5/1/, v/v) to give 10-3.MS (ESI) m/z 303.0[ m ] +H + ].
And B: compound 10-3 (700mg, 2.31mmol), 1-2 (426.87mg, 2.77mmol), tetrakis (triphenylphosphine) palladium (266.90mg, 230.97. Mu. Mol), sodium carbonate (489.60mg, 4.62mmol) were dissolved in 1,4-dioxane (10 mL) and water (2 mL), and the resulting mixture was replaced three times with nitrogen and stirred at 85 ℃ for 12 hours. The reaction mixture was diluted with water (10 mL) and ethyl acetate (10 mL) and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product is separated by means of a column (
Figure PCTCN2021079337-APPB-000062
10g
Figure PCTCN2021079337-APPB-000063
Flash silica gel column, eluent is ethyl acetate/petroleum ether = 0-10%) and purification is carried out to obtain 10-4.MS (ESI) m/z:295.0[ 2 ], [ M + H ] + ].
Step C: compounds 10-4 (0.65g, 2.21mmol), 3-4 (1.35g, 4.41mmol), palladium acetate (49.52mg, 220.59. Mu. Mol), triethylamine (1.12g, 11.03mmol), tetrabutylammonium bromide (617.07mg, 1.91mmol), P (o-tolyl) 3 (671.40mg, 2.21mmol), lithium bromide (574.71mg, 6.62mmol) were dissolved in N, N-dimethylacetamide (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 125 ℃ for 12 hours. The reaction mixture was diluted with water (10 mL) and ethyl acetate (10 mL) and extracted with ethyl acetate (20 mL. Times.3).The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether: ethyl acetate =1/0 to 1/1) to give 10-5.MS (ESI) m/z of 519.0[ 2 ], [ M + H ] + ]。
Step D: compound 10-5 (270mg, 520.19. Mu. Mol) was dissolved in ethanol (10 mL) and water (2 mL), then iron powder (290.50mg, 5.20mmol), ammonium chloride (139.13mg, 2.60mmol) were added, and the resulting mixture was stirred at 90 ℃ for 1 hour. The reaction mixture was filtered, and the filtrate was extracted with ethyl acetate (20 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 10-6.MS (ESI) m/z:489.2[ 2 ], [ M + H ] + ]。
Step E: compounds 1 to 8 (94.93mg, 425.31. Mu. Mol) were dissolved in methylene chloride (5 mL), N-dimethylformamide (2.39mg, 32.72. Mu. Mol) was added to the reaction, oxalyl chloride (207.63mg, 1.64mmol) was added to the reaction system at 25 ℃ and the resulting mixture was stirred at 25 ℃ for 0.5 hour. Concentrating under reduced pressure to obtain crude acyl chloride, dissolving the crude acyl chloride in dichloromethane (5 mL), cooling to 0 ℃, adding pyridine (129.39mg, 1.64mmol) into a reaction system at the temperature, then dropwise adding 10-6 (160mg, 327.16 mu mol) of dichloromethane (1 mL) into the reaction system, stirring the reaction system for 10 minutes at 0 ℃ after the dropwise adding, and then heating to 25 ℃ and stirring for 30 minutes. The reaction solution was diluted with water (10 mL) and dichloromethane (20 mL), extracted with dichloromethane (20 mL. Times.3), the combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product, which was separated by a reverse phase column (
Figure PCTCN2021079337-APPB-000064
10g
Figure PCTCN2021079337-APPB-000065
Flash silica gel column, eluent is ethyl acetate/petroleum ether = 0-50%) and purification is carried out to obtain 10-7.MS (ESI) m/z 694.4[ 2 ], [ M + H ] + ]。
Step F: compound 10-7 (100mg, 144.04. Mu. Mol) was dissolved in methylene chloride (5 mL), and trifluoroacetic acid (1 mL) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 1 hour. The reaction solution is decompressed and concentrated to obtain a crude product. The crude product was separated by HPLC (column information: phenomenex Gemini-NX C18 75X 30mm 3 μm; mobile phase: [ water (0.1% TFA) -ACN%](ii) a 42% -52% of B%, and 7 minutes) to obtain the trifluoroacetate of the compound 10. 1 H NMR(400MHz,DMSO-d 6 )δ=10.40(d,J=2.4Hz,1H),10.00(s,1H),8.47-8.31(m,1H),8.18(d,J=22.8Hz,1H),7.92-7.88(m,1H),7.76(d,J=15.6Hz,1H),7.64(t,J=6.8Hz,3H),7.51(br d,J=8.8Hz,1H),7.43-7.31(m,2H),7.16(t,J=8.8Hz,2H),6.77-6.67(m,1H),6.63-6.54(m,1H),4.14(td,J=5.6,16.8Hz,2H),3.78-3.67(m,2H),1.47(br d,J=4.8Hz,4H);MS(ESI)m/z:601.1[M+H + ].
Example 11: compound 11
Figure PCTCN2021079337-APPB-000066
Step A: compound 11-1 (2g, 13.61mmol) was dissolved in N, N-dimethylformamide (10 mL), 2-bromoethanol (1.87g, 14.97mmol, 1.06mL), cesium carbonate (6.65g, 20.41mmol), and TBAI (1.01g, 2.72mmol) were added to the reaction system, and the resulting mixture was stirred at 80 ℃ for 12 hours. The reaction was quenched by the addition of water (50 mL) and extracted with dichloromethane (20 mL. Times.3). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude compound 11-2.
And B, step B: compound 11-2 (3g, 15.70mmol) was dissolved in N, N-dimethylformamide (20 mL), TBSCl (2.84g, 18.85mmol) and imidazole (2.14g, 31.41mmol) were added to the reaction system, and the resulting mixture was stirred at 25 ℃ for 12 hours. The reaction was quenched by the addition of water (20 mL) and ethyl acetate (50 mL) and extracted with ethyl acetate (50 mL. Times.3). The combined organic phases are washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product which is then separated by a reverse phase column (
Figure PCTCN2021079337-APPB-000067
10g
Figure PCTCN2021079337-APPB-000068
A rapid silica gel column, 0 to 10 percent of petroleum ether/ethyl acetate as eluent) to obtain 11-3.MS (ESI) m/z:305.1[ 2 ] M + H + ].
And C: compounds 3-6 (1g, 3.84mmol), 11-3 (2.35g, 7.69mmol), palladium acetate (86.28mg, 384.29. Mu. Mol), triethylamine (1.94g, 19.21mmol), lithium bromide (1.00g, 11.53mmol), P (o-tolyl) 3 (1.17g, 3.84mmol) was dissolved in N, N-dimethylacetamide (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 125 ℃ for 12 hours. The reaction mixture was diluted with water (10 mL) and ethyl acetate (10 mL) and extracted with ethyl acetate (20 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product is separated by means of a reverse phase column (
Figure PCTCN2021079337-APPB-000069
10g
Figure PCTCN2021079337-APPB-000070
Flash silica gel column, eluent is ethyl acetate/petroleum ether = 0-100%) and purification is carried out to obtain 11-4.MS (ESI) m/z of 485.2[ 2 ], [ M + H ] + ].
Step D: compound 11-4 (500mg, 1.03mmol) was dissolved in ethanol (10 mL) and water (2 mL), then iron powder (576.20mg, 10.32mmol), ammonium chloride (1551.91mg, 10.32mmol) were added, and the resulting mixture was stirred at 90 ℃ for 1 hour. The reaction mixture was filtered, and the filtrate was extracted with ethyl acetate (20 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 11-5.MS (ESI) m/z:455.2[ 2 ], [ M ] +H + ].
Step E: compound 1-8 (287.22mg, 1.29mmol) was dissolved in dichloromethane (10 mL) and added to the reactionN, N-dimethylformamide (7.24mg, 98.99. Mu. Mol), oxalyl chloride (628.19mg, 4.95mmol) was added to the reaction system at 25 ℃ and the resulting mixture was stirred at 25 ℃ for 0.5 hour. The crude acyl chloride is obtained by decompression concentration, the crude acyl chloride is dissolved in dichloromethane (10 mL), the temperature is reduced to 0 ℃, pyridine (391.49mg, 4.95mmol) is added into a reaction system at the temperature, then 11-5 (450mg, 989.86 mu mol) of dichloromethane (1 mL) solution is dripped into the reaction system, the dripped reaction system is stirred for 10 minutes at 0 ℃, and then the temperature is increased to 25 ℃ and stirred for 30 minutes. The reaction mixture was diluted with water (10 mL) and dichloromethane (20 mL) and extracted with dichloromethane (20 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product, which was isolated and purified by chromatography plates (petroleum ether/ethyl acetate = 1/2) to give 11-6.MS (ESI) m/z of 660.2[ 2 ], [ M + H ] + ]。
Step F: compound 11-6 (. 45g, 682.03. Mu. Mol) was dissolved in methylene chloride (10 mL), and trifluoroacetic acid (77.77mg, 682.03. Mu. Mol) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 5 hours. The reaction solution is decompressed and concentrated to obtain a crude product. The crude product was passed through (PPhenomenex Gemini-NX C18 75 + 30mm 3 μm; mobile phase: [ water (0.1% TFA) -ACN](ii) a 30-40 percent of B percent, and 7 minutes) to obtain the trifluoroacetate of the compound 11. 1 H NMR(400MHz,DMSO-d 6 )δ=10.44(s,1H),9.98(s,1H),8.58(d,J=6.4Hz,1H),7.92(dd,J=2.4,13.2Hz,1H),7.79-7.59(m,4H),7.58-7.38(m,3H),7.26-7.05(m,4H),6.62(d,J=2.4Hz,1H),4.17(br t,J=5.6Hz,2H),3.75(t,J=5.6Hz,2H),1.53-1.41(m,4H);MS(ESI)m/z:546.2[M+H + ]
Example 12: compound 12
Figure PCTCN2021079337-APPB-000071
Step A: compound 12-1 (5g, 34.02mmol), 2-bromoethanol (4.68g, 37.42mmol), cesium carbonate (16.63g, 51.03mmol), TBAI (2.51g, 6.80mmol) were dissolved in N, N-dimethylacetamide (50 mL) and water (20 mL), and the resulting mixture was stirred at 90 ℃For 12 hours. The reaction mixture was diluted with water (200 mL), extracted with ethyl acetate (100 mL. Times.2), and the combined organic layers were washed with saturated sodium chloride solution (100 mL. Times.2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 12-2.MS (ESI) m/z:192.03[ m ] +H + ]。
And B: compound 12-2 (6.5g, 34.03mmol), TBSCl (6.15g, 40.83mmol), imidazole (4.63g, 68.05mmol) were dissolved in N, N-dimethylformamide (50 mL), and the resulting mixture was stirred at 25 ℃ for 12 hours. The reaction mixture was diluted with water (200 mL), extracted with ethyl acetate (100 mL. Times.2), and the combined organic layers were washed with saturated sodium chloride solution (50 mL. Times.2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product. The crude product is separated by means of a silica gel column (
Figure PCTCN2021079337-APPB-000072
80g
Figure PCTCN2021079337-APPB-000073
A rapid silica gel column, the eluent is ethyl acetate/petroleum ether = 0-100%, and the flow rate is 60 mL/min) to obtain 12-3.MS (ESI) m/z:307.0[ alpha ], [ M ] +H + ]。
Step C: a mixture of compounds 12-3 (1g, 3.84mmol), 3-6 (2.34g, 7.68mmol), palladium acetate (86.28mg, 384.00. Mu. Mol), tetrakis (triphenylphosphine) palladium (1.17g, 3.84mmol), anhydrous lithium bromide (1.00g, 11.52mmol, 289.38. Mu.L), triethylamine (1.94g, 19.20mmol) dissolved in N, N-dimethylacetamide (10 mL) was replaced with nitrogen three times and stirred at 125 ℃ for 12 hours. The reaction mixture was diluted with 300mL of water, extracted with ethyl acetate (100 mL. Times.2), and the combined organic layers were washed with saturated sodium chloride solution (100 mL. Times.2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product. The residue was purified by silica gel column chromatography (petroleum ether/ethyl acetate =10/1 to 0/1) to obtain 12-4.MS (ESI) m/z of 485.3[ m ] +H + ]。
Step D: compound 12-4 (560mg, 1.16mmol) was dissolved in ethanol (5 mL) and water (1 mL), followed by addition of iron powder (645.41mg, 11.56 mmol), ammonium chloride (309.07mg, 5.78mmol) and the resulting mixture stirred at 90 ℃ for 2 hours. The reaction mixture was filtered and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 12-5.MS (ESI) m/z:455.2[ 2 ], [ M ] +H + ].
Step E Compound 1-8 (294.58mg, 1.32mmol) was dissolved in dichloromethane (5 mL), oxalyl chloride (698.01mg, 5.50mmol) and N, N-dimethylformamide (8.04mg, 109.98. Mu. Mol) were added at 0 deg.C and reacted at 25 deg.C for half an hour. The solution was concentrated to give the corresponding acid chloride and dissolved in dichloromethane (5 mL), and 12-5 (500mg, 1.10 mmol) and 434.99mg,5.50mmol of pyridine were added to the solution at 0 ℃. The resulting mixture was stirred at 25 ℃ for half an hour. Water (20 mL) was added to the reaction mixture, and the mixture was separated, and the aqueous phase was extracted with methylene chloride (30 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 12-6, which was used directly in the next step. MS (ESI) m/z:660.3[ 2 ], [ M + H ] + ].
Step F: compound 12-6 (450mg, 682.03. Mu. Mol) was dissolved in methylene chloride (4 mL), and trifluoroacetic acid (3.89g, 34.10 mmol) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 1 hour. The reaction solution is concentrated to obtain a crude product. Separation by high performance liquid chromatography (column information: phenomenex Luna C18 + 25mm + 10 μm; mobile phase: [ Water (0.225% FA) -ACN)](ii) a 18% -48% for 9 min) to obtain crude product, and separating by supercritical fluid chromatography (column information: DAICEL CHIRALCEL OD (250mm. About.30mm, 10 μm); mobile phase of ethanol in 0.1% ammonia water](ii) a 60-60% of B, 3.9min;100 min) to yield compound 12. 1 H NMR(400MHz,DMSO-d 6 )δ=10.37(s,1H),10.00(s,1H),8.39(d,J=5.6Hz,1H),8.14(s,1H),7.87(dd,J=2.4,13.2Hz,1H),7.69-7.61(m,3H),7.52-7.46(m,2H),7.42(s,1H),7.37-7.30(m,1H),7.16(t,J=8.8Hz,2H),7.07(d,J=16Hz,1H),7.01(d,J=2.4Hz,1H),6.67(dd,J=2.4,5.6Hz,1H),4.99(br t,J=5.2Hz,1H),4.00(t,J=5.6Hz,2H),3.66(q,J=5.2Hz,2H),1.47(br d,J=4.4Hz,4H);MS(ESI)m/z:546.0[M+H + ].
Example 13: compound 13
Figure PCTCN2021079337-APPB-000074
Step A: compounds 1-8 (2.63g, 11.80mmol) were dissolved in dichloromethane (15 mL), oxalyl chloride (2.50g, 19.67mmol) and N, N-dimethylformamide ((28.75mg, 393.34. Mu. Mol) were added at 0 deg.C and reacted at 25 deg.C for half an hour, the solution was concentrated to give the corresponding acid chloride and dissolved in tetrahydrofuran (15 mL), 2-2 ((2g, 15.73mmol) was added to the solution at 0 deg.C and the resulting mixture was stirred at 70 deg.C for two hours, water (300 mL) was added to the reaction solution, the liquid was separated, the aqueous phase was extracted with dichloromethane (100 mL. Times.2), the combined organic phase was washed with saturated sodium chloride solution (200 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 13-7, the crude product was used directly for the next step MS (ESI) m/z:333.1[ M H ], [ 333.1 ] + ]。
And B: compound 13-7 (2g, 6.02mmol) was dissolved in N, N-dimethylformamide (15 mL), sodium hydride (262.63mg, 6.57mmol,60% content) was slowly added at 0 deg.C, the mixture was reacted at 25 deg.C for half an hour, further compound 1-1 (719.69mg, 5.47mmol) was added to the reaction, and the resulting mixture was stirred at 120 deg.C for 12 hours, the reaction mixture was diluted with water (300 mL), extracted with ethyl acetate (150 mL. Times.2), and the combined organic layers were washed with a saturated sodium chloride solution (200 mL), dried over sodium sulfate, filtered, and concentrated under reduced pressure to give a crude product. The crude product is separated by means of a silica gel column (
Figure PCTCN2021079337-APPB-000075
40g
Figure PCTCN2021079337-APPB-000076
Flash silica gel column, eluent is ethyl acetate/petroleum ether = 0-100%) and purification is carried out to obtain 13-6.MS (ESI) m/z:444.2[ 2 ], [ M + H ] + ]。
Step C Compound 3-4 (500mg, 1.64mmol), cuprous iodide (31.19mg, 163.78. Mu. Mol), xantphos (94.77mg, 163.78. Mu. Mol)mol), palladium chloride (29.04mg, 163.78. Mu. Mol) was dissolved in triethylamine (50 mL), and the resulting mixture was replaced with nitrogen three times, and 13-1 (3.22g, 32.76mmol) was added to stir the resulting mixture at 65 ℃ for 12 hours. The reaction mixture was filtered through Celite, diluted with water (200 mL), extracted with ethyl acetate (100 mL. Times.2), and the combined organic layers were washed with saturated sodium chloride solution (200 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give 13-2.MS (ESI) m/z:323.2[ 2 ], [ M + H ] + ]。
Step D: compound 13-2 (2g, 6.20mmol), potassium carbonate (1.71g, 12.40mmol) was dissolved in methanol (20 mL), and the resulting mixture was stirred at 60 ℃ for 2 hours. The reaction mixture was diluted with water (300 mL), extracted with ethyl acetate (150 mL. Times.2), and the combined organic layers were washed with saturated sodium chloride solution (200 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product. The crude product is separated by means of a silica gel column (
Figure PCTCN2021079337-APPB-000077
24g
Figure PCTCN2021079337-APPB-000078
Purifying with fast silica gel column with ethyl acetate/petroleum ether = 0-100% as eluent to obtain 13-3.MS (ESI) m/z:251.2[ M ] +H + ]。
Step E: compound 13-4 (1.570g, 6.18mmol), cuprous chloride (15.81mg, 159.74. Mu. Mol), xantphos (92.43mg, 159.74. Mu. Mol) were dissolved in tetrahydrofuran (5 mL), the resulting mixture was replaced with nitrogen three times, a solution of potassium tert-butoxide in tetrahydrofuran (1M, 1.60mL) was added, 13-3 (400mg, 1.60mmol) and iodomethane (453.46mg, 3.19mmol) were further added, the mixture was replaced with nitrogen once, and the resulting mixture was stirred at 25 ℃ for 12 hours. After the reaction was completed, a saturated aqueous ammonium chloride solution was added to the reaction system and stirred for one hour to quench. The mixture was diluted with water (50 mL), extracted with ethyl acetate (100 mL), and the combined organic layers were washed with saturated sodium chloride solution (50 mL. Times.2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product. The crude product is subjected to thin layer chromatographyPurifying to obtain 13-5.MS (ESI) m/z:393.3[ 2 ], [ M ] +H + ]。
Step F: compound 13-5 (130mg, 331.28. Mu. Mol), compound 13-6 (176.44mg, 397.54. Mu. Mol), tetrakis (triphenylphosphine) palladium (38.28mg, 33.13. Mu. Mol), sodium carbonate (70.23mg, 662.56. Mu. Mol) were dissolved in 1,4-dioxane (1 mL) and water (0.4 mL), the mixture was replaced three times with nitrogen, and the resulting mixture was stirred at 85 ℃ for 4 hours. The reaction mixture was diluted with 50mL of water, extracted with 100mL of ethyl acetate (50 mL. Times.2), and the combined organic layers were washed with saturated sodium chloride solution (25 mL. Times.2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product. The crude product was purified by thin layer chromatography to give 13-9.MS (ESI) m/z of 674.3[ 2 ], M + H + ]。
Step G Compound 13-9 (100mg, 148.41. Mu. Mol) was dissolved in dichloromethane (1 mL), the resulting mixture was stirred at 25 ℃ for 0.5 hour by adding trifluoroacetic acid (846.07mg, 7.42mmol) to the reaction system, and the reaction was quenched by adding aqueous sodium bicarbonate (30 mL) to the reaction solution and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by high performance liquid chromatography (column information: water Xbridge BEH C18. Multidot.50mm. Multidot.10 μm; mobile phase: [ water (0.05% ammonia) -ACN)](ii) a ACN% 40% -60%,20 minutes) to obtain the compound 13. 1 H NMR(400MHz,DMSO-d 6 )δ=10.38(s,1H),10.01(s,1H),8.45(d,J=5.6Hz,1H),7.96(s,1H),7.87(dd,J=2.4,13.2Hz,1H),7.78(s,1H),7.67-7.61(m,2H),7.51-7.46(m,1H),7.34(t,J=8.8Hz,1H),7.16(t,J=8.8Hz,2H),6.81(dd,J=1.6,5.3Hz,2H),6.74(dd,J=2.4,5.7Hz,1H),4.92(br s,1H),4.11(t,J=5.6Hz,2H),3.73(t,J=5.6Hz,2H),2.45(d,J=1.2Hz,3H),1.52-1.42(m,4H)。MS(ESI)m/z:560.1[M+H + ].
Example 14: compound 14
Figure PCTCN2021079337-APPB-000079
Step A: mixing the compounds 2-4 (855.62)mg,3.28 mmol), 3-4 (1g, 3.28mmol), palladium acetate (73.54mg, 327.56. Mu. Mol), triethylamine (1.66g, 16.38mmol), P (o-tolyl) 3 (996.98mg, 3.28mmol), lithium bromide (853.41mg, 9.83mmol) were dissolved in toluene (30 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 110 ℃ for 12 hours. The reaction solution was quenched by addition of water (100 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether/ethyl acetate =10/1 to 0/1) to give 14-1.MS (ESI) m/z of 486.3[ 2 ], [ M + H ] + ]。
And B: compound 14-1 (300mg, 617.81. Mu. Mol) was dissolved in ethanol (10 mL) and water (2 mL), then iron powder (345.02mg, 6.18mmol) and ammonium chloride (165.24mg, 3.09mmol) were added, and the resulting mixture was stirred at 90 ℃ for 6 hours. The reaction mixture was filtered and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 14-2.MS (ESI) m/z:456.1[ 2 ], [ M + H ] + ]。
Step C Compound 14-2 (100mg, 219.49. Mu. Mol) was dissolved in dichloromethane (5 mL), and 1-8 (97.98mg, 438.98. Mu. Mol), T, was added 3 P (419.03mg, 658.47. Mu. Mol), DIEA (141.84mg, 1.10mmol). The mixture was stirred at 25 ℃ for 12 hours. The reaction solution was quenched by addition of water (50 mL) and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude 14-3.MS (ESI) m/z:661.1[ 2 ], [ M + H ] + ]。
Step D Compound 14-3 (30mg, 45.40. Mu. Mol) was dissolved in dichloromethane (2 mL), trifluoroacetic acid (709.19mg, 6.22mmol) was added to the reaction system, the resulting mixture was stirred at 25 ℃ for 1 hour, and the reaction mixture was concentrated under reduced pressure to give the crude product. The crude product was separated by HPLC (column information: XTTIMATE C18 150X 40mm X10 μm; mobile phase: [ water (0.05% ammonia) -ACN)](ii) a 32-62% of ACN percent and 10 minutes) to obtain the compound 14. 1 H NMR(400MHz,DMSO-d 6 )δ=10.32(s,1H),10.02(s,1H),8.62(s,1H),8.09(s,1H),7.87-7.77(m,3H),7.67-7.62(m,2H),7.44(dd,J=1.2,8.8Hz,1H),7.33(t,J=8.8Hz,1H),7.16(t,J=8.8Hz,2H),7.10(s,1H),6.97(d,J=16.0Hz,1H),4.94(t,J=5.2Hz,1H),4.16(t,J=5.6Hz,2H),3.74(q,J=5.2Hz,2H),1.47(br d,J=2.8Hz,4H);MS(ESI)m/z:547.1[M+H + ].
Example 15: compound 15
Figure PCTCN2021079337-APPB-000080
Step A: compound 15-1 (3g, 23.11mmol), compound 15-2 (4.07g, 23.11mmol), and potassium carbonate (6.40g, 46.32mmol) were dissolved in N, N-dimethylformamide (20 mL), and the resulting mixture was stirred at 100 ℃ for 12 hours. The reaction mixture was diluted with water (100 mL), extracted with ethyl acetate (100 mL. Times.2), and the combined organic layers were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give crude product which was separated by silica gel column (
Figure PCTCN2021079337-APPB-000081
120g
Figure PCTCN2021079337-APPB-000082
Purifying by a rapid silica gel column, wherein the eluent is ethyl acetate/petroleum ether = 0-100 percent to obtain 15-3.MS (ESI) m/z:284.7[ M ] +H + + ]。
And B: compound 15-3 (3g, 10.52mmol), 1-2 (1.94g, 12.63mmol), tetrakis (triphenylphosphine) palladium (1.22g, 1.05mmol), sodium carbonate (2.23g, 21.05mmol) were dissolved in 1,4-dioxane (30 mL) and water (6 mL), the resulting mixture was replaced three times with nitrogen and stirred at 84 ℃ for 4 hours. The reaction solution was quenched by addition of water (100 mL) and extracted with ethyl acetate (100 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product is purified by column chromatography (SiO 2, petroleum ether: ethyl acetate = 20/1-5/1) to obtain 15-4。MS(ESI)m/z:277.0[M+H + ]。
And C: compounds 15-4 (906.28mg, 3.28mmol), 3-4 (1g, 3.28mmol), palladium acetate (73.54mg, 327.56. Mu. Mol), triethylamine (1.66g, 16.38mmol), P (o-tolyl) 3 (997.00mg, 3.28mmol), lithium bromide (853.46mg, 9.83mmol) were dissolved in toluene (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 110 ℃ for 12 hours. The reaction mixture was quenched by addition of water (100 mL) and extracted with ethyl acetate (100 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether/ethyl acetate =10/1 to 0/1) to give 15-5.MS (ESI) m/z:501.2[ 2 ], [ M ] +H + ]。
Step D Compound 15-5 (400mg, 798.33. Mu. Mol) was dissolved in ethanol (10 mL) and water (2 mL), then iron powder (445.86mg, 7.98mmol), ammonium chloride (213.51mg, 3.99mmol) were added and the resulting mixture was stirred at 90 ℃ for 6 hours. The reaction mixture was filtered and extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give crude 15-6.MS (ESI) m/z:471.2[ 2 ], [ M ] +H + ]。
Step E Compound 1-8 (56.86mg, 254.74. Mu. Mol) was dissolved in dichloromethane (5 mL), oxalyl chloride (134.73mg, 1.06mmol) and N, N-dimethylformamide (1.55mg, 21.23. Mu. Mol) were added at 0 ℃ and reacted at 25 ℃ for half an hour. The solution was concentrated to give the corresponding acid chloride and dissolved in dichloromethane (10 mL), and 15-6 (100mg, 212.28. Mu. Mol) and pyridine (83.96mg, 1.06mmol) were added to the solution at 0 ℃. The resulting mixture was stirred at 25 ℃ for half an hour. Water (20 mL) was added to the reaction mixture, and the aqueous phase was extracted with dichloromethane (20 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (20 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 15-7, which was used directly in the next step. MS (ESI) m/z:676.3[ 2 ], [ M + H ] + ].
Step F Compound 15-7 (120mg, 177.45. Mu. Mol) was dissolved in methylene chloride (2 mL), and the resulting mixture was stirred at 25 ℃ for 1 hour with the addition of trifluoroacetic acid (2.77g, 24.31mmol) to the reaction system, and the reaction mixture wasConcentration under reduced pressure gave the crude product. The crude product was separated by HPLC (column information: XTTIMATE C18 150X 40mm X10 μm; mobile phase: [ water (0.05% ammonia) -ACN)](ii) a 37-67 percent of ACN percent and 10 minutes) to obtain a compound 15. 1 H NMR(400MHz,DMSO-d 6 )δ=10.34(s,1H),10.01(s,1H),8.40(d,J=5.6Hz,1H),8.06(d,J=2.4Hz,1H),7.97(s,1H),7.77(s,1H),7.68-7.62(m,3H),7.52(d,J=16Hz,1H),7.35(d,J=8.8Hz,1H),7.16(t,J=8.8Hz,2H),6.92(d,J=16Hz,1H),6.81(d,J=2.4Hz,1H),6.69(dd,J=2.4,5.6Hz,1H),4.91(br s,1H),4.12(t,J=5.6Hz,2H),3.73(br d,J=4.4Hz,2H),1.47(br d,J=5.6Hz,4H);MS(ESI)m/z:562.4[M+H + ].
Example 16: compound 16
Figure PCTCN2021079337-APPB-000083
Step A: compound 6 (300mg, 598.23. Mu. Mol), 16-1 (239.80mg, 1.79mmol), cesium carbonate (584.74mg, 1.79mmol), potassium iodide (9.93mg, 59.82. Mu. Mol) were dissolved in N, N-dimethylacetamide (2 mL), and the resulting mixture was stirred at 100 ℃ for 12 hours. The reaction mixture was diluted with water (10 mL) and ethyl acetate (20 mL), separated, the aqueous phase was extracted with ethyl acetate (20 mL × 2), and the combined organic layer was washed with saturated brine (20 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a crude product, which was separated by preparative thin layer chromatography (dichloromethane/methanol = 5/1) to give compound 16. 1 H NMR(400MHz,DMSO-d 6 )δ=10.40(s,1H),10.01(s,1H),8.39(d,J=5.6Hz,1H),8.07(s,1H),7.87(dd,J=2.2,13.2Hz,1H),7.78(s,1H),7.67-7.60(m,2H),7.54-7.45(m,2H),7.39-7.31(m,1H),7.20-7.12(m,2H),6.92(d,J=16.0Hz,1H),6.83(d,J=2.4Hz,1H),6.74(dd,J=2.4,5.6Hz,1H),4.16-4.05(m,1H),2.88(br d,J=11.2Hz,2H),2.24(s,3H),2.17-2.04(m,2H),2.03-1.89(m,4H),1.51-1.41(m,4H);MS(ESI)m/z:599.4[M+H + ]。
Example 17: compound 17
Figure PCTCN2021079337-APPB-000084
Step A: compound 6 (50mg, 99.70. Mu. Mol), 17-1 (33.28mg, 119.64. Mu. Mol), and cesium carbonate (97.46mg, 299.10. Mu. Mol) were dissolved in N, N-dimethylacetamide (2 mL), and the resulting mixture was stirred at 60 ℃ for 2 hours. The reaction mixture was diluted with water (10 mL) and ethyl acetate (20 mL), separated, the aqueous phase was extracted with ethyl acetate (20 mL × 2), and the combined organic layer was washed with saturated brine (20 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a crude product, which was separated by preparative thin layer chromatography (dichloromethane/methanol = 5/1) to give compound 17-2.MS (ESI) m/z:699.2[ 2 ], [ M + H ] + ]。
And B: compound 17-2 (70mg, 100.18. Mu. Mol) was dissolved in ethyl acetate (3 mL), a methanol solution of hydrochloric acid (4 mol/L,3 mL) was added to the reaction system, the resulting mixture was stirred at 20 ℃ for 30 minutes, and the reaction mixture was concentrated under reduced pressure to give a crude product. The crude product was separated by high performance liquid chromatography (column information: unisil 3-100 C18 Ultra 150 x 50mm x 3 μm; mobile phase: [ water (0.225% FA) -ACN%](ii) a 15 percent to 35 percent of B percent for 10 minutes) to obtain a compound 17-3.MS (ESI) m/z of 599.3[ 2 ], [ M + H ] + ]。
And C: compound 17-3 (97.71mg, 163.22. Mu. Mol) was dissolved in tetrahydrofuran (10 mL), 37 aqueous formaldehyde (49.01mg, 1.63mmol) and sodium triacetoxyborohydride (172.97mg, 816.10. Mu. Mol) were added, and the resulting mixture was stirred at 20 ℃ for 12 hours. Quenching the reaction with saturated sodium bicarbonate (5 mL), adding water (5 mL), separating, extracting the aqueous phase with ethyl acetate (20 mL × 3), washing the combined organic layer with saturated brine (20 mL), drying over anhydrous sodium sulfate, filtering, concentrating under reduced pressure to obtain crude product, separating by preparative high performance liquid chromatography (column information: unisil 3-100 C18 Ultra 150 × 50mm 3 μm; mobile phase [ Water (0.225% FA) -ACN](ii) a 15 percent to 35 percent of the compound B) for 10 minutes) to obtain the formate of the compound 17. 1 H NMR(400MHz,DMSO-d 6 )δ=10.41(s,1H),10.02(s,1H),8.40(d,J=5.6Hz,1H),8.27(s,1H),7.98(s,1H),7.88(dd,J=2.4,13.2Hz,1H),7.78(s,1H),7.65(dd,J=5.2,9.2Hz,2H),7.55-7.45(m,2H),7.35(t,J=9.2Hz,1H),7.16(t,J=8.8Hz,2H),6.92(d,J=16.0Hz,1H),6.86(d,J=2.4Hz,1H),6.74(dd,J=2.4,5.6Hz,1H),3.99(br d,J=7.2Hz,2H),2.92(br d,J=11.2Hz,2H),2.30(s,3H),2.12(br t,J=11.2Hz,2H),1.83(br dd,J=3.6,6.8Hz,1H),1.57-1.41(m,6H),1.28(q,J=10.8Hz,2H);MS(ESI)m/z:613.3[M+H + ]。
Example 18: compound 18
Figure PCTCN2021079337-APPB-000085
Step A: compound 6 (50mg, 99.70. Mu. Mol), 18-1 (34.96mg, 119.65. Mu. Mol), cesium carbonate (97.46mg, 299.10. Mu. Mol) were dissolved in N, N-dimethylacetamide (2 mL), and the resulting mixture was stirred at 60 ℃ for 2 hours. The reaction mixture was diluted with water (10 mL) and ethyl acetate (20 mL), separated, the aqueous phase was extracted with ethyl acetate (20 mL × 2), the combined organic layer was washed with saturated brine (20 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a crude product, which was separated by preparative thin layer chromatography (dichloromethane/methanol = 5/1) to give compound 18-2.MS (ESI) m/z:713.2[ 2 ], [ M ] +H + ]。
And B: compound 18-2 (70mg, 98.21. Mu. Mol) was dissolved in ethyl acetate (3 mL), a methanol solution of hydrochloric acid (4 mol/L,3 mL) was added to the reaction system, the resulting mixture was stirred at 20 ℃ for 30 minutes, and the reaction mixture was concentrated under reduced pressure to give a crude product. The crude product was separated by preparative high performance liquid chromatography (column information: unisil 3-100 C18 Ultra 150 x 50mm x 3 μm; mobile phase: [ water (0.225% FA) -ACN)](ii) a 15-35 percent of ACN for 10 minutes) to obtain the compound 18-3.MS (ESI) m/z:613.3[ 2 ], [ M + H ] + ]。
And C: compound 18-3 (100mg, 163.22. Mu. Mol) was dissolved in tetrahydrofuran (10 mL), 37 aqueous formaldehyde (49.01mg, 1.63mmol) and sodium triacetoxyborohydride (172.97mg, 816.10. Mu. Mol) were added, and the resulting mixture was stirred at 20 ℃ for 12 hours. The reaction was quenched with saturated sodium bicarbonate (5 mL), diluted with water (5 mL), separated, the aqueous phase extracted with ethyl acetate (20 mL. Times.3), mixed andthe organic layer was washed with saturated brine (20 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a crude product, which was separated by preparative high performance liquid chromatography (column information: unisil 3-100 c18 Ultra 150 × 50mm × 3 μm; mobile phase [ Water (0.225% FA) -ACN](ii) a 15% -35% of ACN% for 10 minutes) to obtain the formate of the compound 18. 1 H NMR(400MHz,DMSO-d 6 )δ=10.42(br s,1H),10.03(s,1H),8.40(d,J=5.6Hz,1H),8.31(s,1H),8.01(s,1H),7.88(dd,J=2.3,13.2Hz,1H),7.77(s,1H),7.68-7.62(m,2H),7.54-7.47(m,2H),7.35(t,J=9.0Hz,1H),7.16(t,J=9.0Hz,2H),6.92(d,J=15.9Hz,1H),6.86(d,J=2.4Hz,1H),6.74(dd,J=2.4,5.6Hz,1H),4.12(br t,J=7.1Hz,2H),2.93(br d,J=11.7Hz,2H),2.31(s,3H),2.16(br t,J=10.9Hz,2H),1.79-1.60(m,4H),1.54-1.39(m,4H),1.33-1.08(m,3H);MS(ESI)m/z:627.3[M+H + ]。
Example 19: compound 19
Figure PCTCN2021079337-APPB-000086
Step A: compound 6 (50mg, 99.70. Mu. Mol), 19-1 (35.08mg, 119.64. Mu. Mol), and cesium carbonate (97.46mg, 299.10. Mu. Mol) were dissolved in N, N-dimethylacetamide (2 mL), and the resulting mixture was stirred at 60 ℃ for 2 hours. The reaction mixture was diluted with water (10 mL) and ethyl acetate (20 mL), separated, the aqueous phase was extracted with ethyl acetate (20 mL × 2), the combined organic layer was washed with saturated brine (20 mL × 2), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a crude product, which was separated by preparative thin layer chromatography (dichloromethane/methanol = 5/1) to give compound 19-2.MS (ESI) m/z:714.2[ 2 ], [ M ] +H + ]。
And B: compound 19-2 (70mg, 98.07. Mu. Mol) was dissolved in ethyl acetate (3 mL), a methanol solution of hydrochloric acid (4 mol/L,3 mL) was added to the reaction system, the resulting mixture was stirred at 20 ℃ for 30 minutes, and the reaction mixture was concentrated under reduced pressure to give a crude product. The crude product was separated by high performance liquid chromatography (column information: unisil 3-100 C18 Ultra 150 x 50mm x 3 μm; mobile phase: [ water (0.225% FA) -ACN%];B%:15%-35%,10 min) to yield compound 19-3.MS (ESI) m/z:614.3[ M ] +H + ]。
And C: compound 19-3 (100mg, 162.96. Mu. Mol) was dissolved in tetrahydrofuran (10 mL), 37% aqueous formaldehyde (48.94mg, 1.63mmol) and sodium triacetoxyborohydride (172.69mg, 814.79. Mu. Mol) were added, and the resulting mixture was stirred at 20 ℃ for 12 hours. Quenching the reaction with saturated sodium bicarbonate (5 mL), adding water (5 mL), separating, extracting the aqueous phase with ethyl acetate (20 mL × 3), washing the combined organic layer with saturated brine (20 mL), drying over anhydrous sodium sulfate, filtering, and concentrating under reduced pressure to obtain crude product, which is separated by high performance liquid chromatography (column information: unisil 3-100 C18 Ultra 150 × 50mm × 3 μm; mobile phase [ Water (0.225% FA) -ACN](ii) a 15 percent to 35 percent of the compound B) for 10 minutes) to obtain the formate of the compound 19. 1 H NMR(400MHz,DMSO-d 6 )δ=10.40(s,1H),10.02(s,1H),8.40(d,J=5.6Hz,1H),8.23(s,1H),7.99(s,1H),7.88(dd,J=2.0,13.2Hz,1H),7.77(s,1H),7.65(dd,J=5.2,8.8Hz,2H),7.56-7.45(m,2H),7.35(t,J=9.2Hz,1H),7.16(t,J=8.8Hz,2H),6.91(d,J=16.0Hz,1H),6.87(d,J=2.0Hz,1H),6.74(dd,J=2.4,5.6Hz,1H),4.19(br t,J=6.4Hz,2H),2.70(br t,J=6.4Hz,2H),2.50-2.31(m,8H),2.20(s,3H),1.57-1.39(m,4H);MS(ESI)m/z:628.3[M+H + ]。
Example 20: compound 20
Figure PCTCN2021079337-APPB-000087
Step A: compound 20-1 (5.08g, 19.10mmol), compound 20-2 (5.02g, 21.00mmol), cesium carbonate (12.44g, 38.19mmol) were dissolved in N, N-dimethylacetamide (50 mL), and the resulting mixture was stirred at 60 ℃ for 12 hours. The reaction mixture was diluted with water (100 mL), extracted with ethyl acetate (100 mL × 2), and the combined organic layer was washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give a crude product, which was then separated with silica gel column (petroleum ether/ethyl acetate = 1:0-5:1) and purified to give compound 20-3.MS (ESI) m/z:425.3[ 2 ], [ M + H ] + ]。
And B: compound 20-3 (2.4 g, 5.66mmol) was dissolved in ethanol (20 mL), sodium borohydride (641.91mg, 16.97mmol) was added to the reaction system, the resulting mixture was stirred at 90 ℃ for 12 hours, hydrochloric acid (1 mol/L,10 mL) was added to quench the reaction, water (50 mL) was added, extraction was performed with ethyl acetate (50 mL. Times.2), and the combined organic layers were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give compound 20-4.MS (ESI) m/z:383.3[ 2 ], [ M ] +H + ]。
And C: compound 20-4 (2.9g, 7.59mmol) was dissolved in N, N-dimethylacetamide (15 mL), TBSCl (1.37g, 9.10mmol) and imidazole (1.03g, 15.17mmol) were added, and the resulting mixture was stirred at 25 ℃ for 12 hours. Diluting with water (50 mL), extracting with ethyl acetate (50 mL. Times.2), washing the mixed organic layer with saturated brine (80 mL), drying with anhydrous sodium sulfate, filtering, and concentrating under reduced pressure to obtain crude product, separating the crude product by preparative high performance liquid chromatography (column information: phenomenex luna C18 (250X 70mm,15 μm); mobile phase: [ water (0.1% TFA) -ACN: ]](ii) a 70 percent ACN-100 percent ACN for 30 minutes) to obtain 20-5 of compound. MS (ESI) m/z:497.4[ 2 ] M + H + ]。
Step D: compounds 3-6 (655.04mg, 2.52mmol), compounds 20-5 (1.25g, 2.52mmol), palladium acetate (56.51mg, 251.73. Mu. Mol), triethylamine (1.27g, 12.59mmol), lithium bromide (655.87mg, 7.55mmol), P (o-tolyl) 3 (766.18mg, 2.52mmol) was dissolved in N, N-dimethylacetamide (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 110 ℃ for 12 hours. Water (50 mL) was added to the reaction mixture, which was extracted with ethyl acetate (50 mL. Times.2), and the combined organic phases were washed with saturated sodium chloride solution (100 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether/ethyl acetate = 5:1) to give 20-6.MS (ESI) m/z:629.5, [ M ] +H + ].
Step E: compound 20-6 (1.5g, 2.39mmol) was dissolved in ethanol (15 mL), then iron powder (1.33g, 23.85mmol), an ammonium chloride solution (127.59mg, 2.39mmol,2.5mL water) were added and the resulting mixture was stirred at 80 ℃ for 12 hours. After filtration, the filtrate was collected, and water (50 mL) was added to the filtrate, followed by extraction with ethyl acetate (50 mL. Times.2). MergingThe organic phase was washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 20-7, and the crude product was used directly in the next step. MS (ESI) m/z of 599.5[ 2 ], [ M + H ] + ].
Step F: compounds 1-8 (339.14mg, 1.52mmol) were dissolved in dichloromethane (10 mL), oxalyl chloride (741.79mg, 5.84mmol) and N, N-dimethylformamide (8. Mu.L) were added at 0 ℃ and reacted at 25 ℃ for half an hour. The solution was concentrated to give the corresponding acid chloride and dissolved in dichloromethane (10 mL), 20-7 (700mg, 1.17mmol) and pyridine (462.27mg, 5.84mmol) were added to the solution at 0 deg.C and the resulting mixture was stirred at 25 deg.C for half an hour. Water (30 mL) was added to the reaction mixture, and the aqueous phase was extracted with dichloromethane (20 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 20-8, and the crude product was used directly in the next step. MS (ESI) m/z:804.2[ 2 ], [ M + H ] + ].
G: compound 20-8 (0.9g, 1.12mmol) was dissolved in dichloromethane (20 mL), and then a solution of hydrochloric acid in methanol (4 mol/L,5 mL) and ethyl acetate (5 mL) were added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 1 hour. And concentrating the reaction solution under reduced pressure to obtain a crude product. The crude product was separated by preparative high performance liquid chromatography (column information: phenomenex luna C18: 40mm: 15 μm; mobile phase: [ water (0.225% FA) -ACN%](ii) a 15% -45% of ACN and 10 minutes) to obtain a compound 20. 1 H NMR(400MHz,DMSO-d 6 )δ=10.36(s,1H),9.99(s,1H),8.39(d,J=5.6Hz,1H),8.00(s,1H),7.86(dd,J=2.4,13.2Hz,1H),7.66-7.59(m,3H),7.48(dd,J=1.2,8.8Hz,1H),7.38-7.31(m,1H),7.15(t,J=8.8Hz,2H),6.91-6.84(m,2H),6.71(dd,J=2.4,5.6Hz,1H),5.08(t,J=5.2Hz,1H),4.91(t,J=5.2Hz,1H),4.49(d,J=5.2Hz,2H),4.07(t,J=5.6Hz,2H),3.72(q,J=5.6Hz,2H),1.50-1.43(m,4H);MS(ESI)m/z:576.3[M+H + ].
Example 21: compound 21
Figure PCTCN2021079337-APPB-000088
Figure PCTCN2021079337-APPB-000089
Step A: mixing compound 3-6 (735.87mg, 2.83mmol), compound 20-3 (1.2g, 2.83mmol), palladium acetate (63.49mg, 283.00. Mu. Mol), triethylamine (1.43g, 14.15mmol), lithium bromide (736.80mg, 8.49mmol), P (o-tolyl) 3 (860.72mg, 2.83mmol) was dissolved in N, N-dimethylacetamide (10 mL), and the resulting mixture was replaced with nitrogen three times and stirred at 110 ℃ for 12 hours. Water (50 mL) was added to the reaction mixture, which was extracted with ethyl acetate (50 mL. Times.2), and the combined organic phases were washed with saturated sodium chloride solution (100 mL), dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure to give the crude product. The crude product was purified by silica gel column separation (petroleum ether/ethyl acetate = 1:1-0:1) to give 21-1.MS (ESI) m/z:557.4[ m + H ] + ]。
And B: compound 21-1 (1.36g, 2.44mmol) was dissolved in ethanol (25 mL), followed by addition of iron powder (1.21g, 21.60mmol), ammonium chloride solution (1.34g, 24.97mmol,2.5mL water) and stirring of the resulting mixture at 80 ℃ for 12 hours. After filtration, the filtrate was collected, and water (50 mL) was added to the filtrate, followed by extraction with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 21-2, which was used directly in the next step. MS (ESI) m/z of 527.4[ 2 ], [ M + H ] + ]。
And C: compounds 1-8 (330.56mg, 1.48mmol) were dissolved in dichloromethane (10 mL), oxalyl chloride (723.01mg, 5.70mmol) and N, N-dimethylformamide (8. Mu.L) were added at 0 ℃ and reacted at 25 ℃ for half an hour. The solution was concentrated to give the corresponding acid chloride and dissolved in dichloromethane (10 mL), 21-2 (600mg, 1.14mmol) and pyridine (450.56mg, 5.70mmol) were added to the solution at 0 deg.C and the resulting mixture was stirred at 25 deg.C for half an hour. Water (30 mL) was added to the reaction mixture, and the mixture was separated, and the aqueous phase was extracted with methylene chloride (20 mL. Times.3). The combined organic phases were washed with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 21-3, which was used directly in the next step. MS (ESI) m/z of 732.3[ 2 ], [ M + H ] + ]。
Step D:compound 21-3 (1g, 1.37mmol) was dissolved in methanol (5 mL) and water (5 mL), then lithium hydroxide (65.45mg, 2.73mmol) was added to the reaction solution, and the resulting mixture was stirred at 25 ℃ for 18 hours. The pH was adjusted to about 3 using hydrochloric acid (1 mol/L), water (50 mL) was added, and extraction was performed with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (100 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 21-4, which was used directly in the next step. MS (ESI) m/z of 590.2[ m ] +H + ]。
Step E: compound 21-4 (100mg, 169.62. Mu. Mol) was dissolved in N, N-dimethylacetamide (2 mL), ammonium chloride (18.15mg, 339.24. Mu. Mol), pyBOP (105.92mg, 203.55. Mu. Mol), DIEA (87.69mg, 678.49. Mu. Mol) and HOBt (27.50mg, 203.55. Mu. Mol) were added, and the resulting mixture was stirred at 25 ℃ for 12 hours. Water (20 mL) was added, and the mixture was extracted with ethyl acetate (20 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product, which was separated by high performance liquid chromatography (column information: phenomenex Luna C18 × 25mm × 10 μm; mobile phase [ water (0.225%; FA) -ACN](ii) a 22-52 percent of ACN percent and 11 minutes) to obtain the compound 21. 1 H NMR(400MHz,DMSO-d 6 )δ=10.39(s,1H),10.02(s,1H),8.42(d,J=5.6Hz,1H),8.23(s,1H),8.12(d,J=16.4Hz,1H),7.87(dd,J=2.4,13.2Hz,1H),7.68-7.60(m,2H),7.49(dd,J=1.2,8.8Hz,1H),7.43(s,1H),7.35(t,J=9.2Hz,1H),7.26(s,1H),7.19-7.11(m,2H),7.00(d,J=16.4Hz,1H),6.80(d,J=2.4Hz,1H),6.76(dd,J=2.4,5.6Hz,1H),4.16(t,J=5.2Hz,2H),3.78(t,J=5.2Hz,2H),1.49–1.41(m,4H);MS(ESI)m/z:589.4[M+H + ].
Example 22: compound 22
Figure PCTCN2021079337-APPB-000090
Step A: compound 21-4 (380mg, 644.56. Mu. Mol) was dissolved in acetonitrile (9 mL), methylamine hydrochloride (87.04mg, 1.29mmol), HATU (269.59mg, 709.02. Mu. Mol), DIEA (333.21mg, 2.58mmol) and N, N-dimethylacetamide (0.9 mL) were added, and the resulting mixture was stirred at 25 ℃ for 12 hours. Water (50 mL) was added, and the mixture was extracted with methylene chloride (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product, which was purified by high performance liquid chromatography (column information: waters Xbridge 150 × 25mm × 5 μm; mobile phase: [ water (10mM NH4HCO3) -ACN ]; ACN%:27% -57%,9 min) to give Compound 22.1H NMR (400mhz, dmso-d 6) δ =10.36 (s, 1H), 9.99 (s, 1H), 8.42 (d, J =5.6hz, 1h), 8.23 (s, 1H), 8.11 (d, J =16.4hz, 1h), 8.05 (d, J =4.8hz, 1h), 7.86 (dd, J =2.4,13.2hz, 1h), 7.67-7.60 (m, 2H), 7.49 (dd, J =1.2,8.8hz, 1h), 7.38-7.32 (m, 1H), 7.19-7.11 (m, 2H), 7.00 (d, J =16.4hz, 1h), 6.81 (d, J = 2.4h), 6.75 (1h, J =2.4, 5.1h), 7.00 (d, J =16.4h, 3.78, 3.8H, 18H, 3.8H, 18 hz, 3.8H, 1H); MS (ESI) m/z 603.2[ 2 ], [ M + H + ].
Example 23: compound 23
Figure PCTCN2021079337-APPB-000091
Figure PCTCN2021079337-APPB-000092
Step A: compound 21-4 (100mg, 169.62. Mu. Mol) was dissolved in N, N-dimethylacetamide (2 mL), hydroxylamine hydrochloride (23.57mg, 339.24. Mu. Mol), pyBOP (105.92mg, 203.55. Mu. Mol), DIEA (109.61mg, 848.11. Mu. Mol) and HOBt (27.50mg, 203.55. Mu. Mol) were added, and the resulting mixture was stirred at 25 ℃ for 12 hours. Water (20 mL) was added, and the mixture was extracted with ethyl acetate (20 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the crude product, which was separated by high performance liquid chromatography (column information: unisil 3-100 c18 Ultra 150 × 50mm 3 μm; mobile phase [ water (0.225%; FA) -ACN](ii) a 25% -45% of ACN% and 10 minutes) to obtain a compound 23. 1 H NMR(400MHz,DMSO-d 6 )δ=10.88(s,1H),10.37(s,1H),10.00(s,1H),8.94(d,J=1.2Hz,1H),8.43(d,J=5.6Hz,1H),8.23(s,1H),7.99(d,J=16.4Hz,1H),7.87(dd,J=2.4,13.2Hz,1H),7.70-7.60(m,2H),7.49(dd,J=1.6,8.8Hz,1H),7.40-7.32(m,1H),7.16(t,J=9.2Hz,2H),7.02(d,J=16.4Hz,1H),6.83-6.73(m,2H),4.98(t,J=5.6Hz,1H),4.16(t,J=5.2Hz,2H),3.78(q,J=5.2Hz,2H),1.51–1.44(m,4H);MS(ESI)m/z:605.4[M+H + ].
Example 24: compound 24
Figure PCTCN2021079337-APPB-000093
Step A: compound 21-4 (100mg, 169.62. Mu. Mol) was dissolved in N, N-dimethylacetamide (2 mL), hydroxyethylamine (20.72mg, 339.24. Mu. Mol), pyBOP (105.92mg, 203.55. Mu. Mol), DIEA (109.61mg, 848.11. Mu. Mol) and HOBt (27.50mg, 203.55. Mu. Mol) were added, and the resulting mixture was stirred at 25 ℃ for 12 hours. Water (50 mL) was added, and the mixture was extracted with ethyl acetate (50 mL. Times.2). The combined organic phases were washed with saturated sodium chloride solution (50 mL), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a crude product, which was separated by HPLC (column information: phenomenex Gemini-NX C18 75 x 30mm x 3 μm; mobile phase [ Water (0.225% FA) -ACN](ii) a 15% -45% of ACN and 7 minutes) to obtain a compound 24. 1 H NMR(400MHz,DMSO-d 6 )δ=10.42(s,1H),9.99(s,1H),8.50(d,J=6.0Hz,1H),8.27(s,1H),8.17(d,J=16.4Hz,1H),7.98(t,J=5.8Hz,1H),7.90(dd,J=2.4,13.2Hz,1H),7.67-7.60(m,2H),7.52(dd,J=1.2,8.8Hz,1H),7.40(t,J=8.8Hz,1H),7.19-7.11(m,3H),7.09(s,1H),6.96(s,1H),4.20(t,J=5.2Hz,2H),3.79(t,J=5.2Hz,2H),3.53-3.48(m,4H),1.52-1.42(m,4H);MS(ESI)m/z:633.1[M+H + ].
Experimental example 1: vascular endothelial growth factor receptor 2 (VEGFR-2/KDR) kinase Activity inhibition assay (in vitro evaluation)
Experimental materials:
KDR Kinase Enzyme System (KDR Kinase Enzyme System) was purchased from Promega. Nivo multi-label analyzer (PerkinElmer).
The experimental method comprises the following steps:
use ofThe kinase buffer in the kit dilutes the enzyme, substrate, ATP, and inhibitor. The test compound was diluted 5-fold with a riffle to 8 th concentration, i.e. from 50 μ M to 0.65nM with a final DMSO concentration of 5%, setting up a double-well experiment. mu.L of each concentration gradient of inhibitor, 2. Mu.L of KDR enzyme (1.5 ng), 2. Mu.L of a mixture of substrate and ATP (138.8. Mu.M ATP, 0.2. Mu.g/. Mu.l Poly E) were added to the plate 4 Y 1 ) The final concentration gradient of the compound was then 10. Mu.M diluted to 0.13nM. The reaction system was left at 25 ℃ for 60 minutes. After the reaction is finished, 5 mul ADP-Glo reagent is added into each hole, the reaction is continued for 40 minutes at 25 ℃,10 mul of kinase detection reagent is added into each hole after the reaction is finished, the chemiluminescence is read by a Perkinelmer Envision multi-label analyzer after the reaction is carried out for 30 minutes at 25 ℃, and the integration time is 0.5 second.
And (3) data analysis:
the raw data was converted to inhibition, IC, using the equation (sample-Min)/(Max-Min) 100% 50 Values of (d) can be obtained by curve fitting of four parameters (log (inhibition) vs. response-variable slope mode in GraphPad Prism).
The results are shown in Table 1.
The experimental conclusion is that: the compound has good inhibitory activity on VEGFR2/KDR kinase.
Experimental example 2: axl kinase Activity inhibition assay (in vitro evaluation)
Experimental materials:
AXL Kinase Enzyme System (AXL Kinase Enzyme System) was purchased from Promega. Nivo multi-label analyzer (PerkinElmer). The experimental method comprises the following steps:
the enzyme, substrate, ATP and inhibitor were diluted with the kinase buffer in the kit.
The test compounds were diluted 5-fold with a calandria to 8 th concentration, i.e. from 5 μ M to 0.065nM, with a DMSO concentration of 5%, setting up a double-well experiment. mu.L of each concentration gradient of inhibitor, 2. Mu.L of AXL enzyme (6 ng), 2. Mu.L of a mixture of substrate and ATP (89.37. Mu.M ATP, 0.2. Mu.g/. Mu.L Axltide) were added to the plate, at a final concentration gradient of compound of 1. Mu.M diluted to 0.013nM. The reaction system was left at 25 ℃ for 60 minutes. After the reaction, 5 μ L of ADP-Glo reagent is added into each hole, the reaction is continued for 40 minutes at 25 ℃,10 μ L of kinase detection reagent is added into each hole after the reaction is finished, and after the reaction is carried out for 30 minutes at 25 ℃, a Perkinelmer Envision multi-label analyzer is adopted to read chemiluminescence, and the integration time is 0.5 second.
And (3) data analysis:
the raw data was converted to inhibition, IC, using the equation (sample-Min)/(Max-Min) 100% 50 Values of (d) are obtained by curve fitting of four parameters (obtained from log (inhibition) vs. response-variable slope mode in GraphPad Prism). Table 1 provides the AXL enzymatic inhibitory activity of the compounds of the present invention.
The results are shown in Table 1.
TABLE 1 inhibition of kinase Activity test results
Test compounds Axl(nM) VEGFR2(nM)
Compound 1 23.16 25.84
Formate salt of Compound 2 36.62 44.12
Formate salt of Compound 3 11.34 37.37
Compound 4 19.14 31.59
Compound 6 37.98 96.48
Compound 7 20.17 76.98
Compound 8 13.25 /
Trifluoroacetic acid salt of Compound 9 34.23 74.28
Trifluoroacetic acid salt of Compound 11 33.07 42.69
Compound 12 11.48 74.94
Compound 13 97.25 224.2
Compound 14 25.38 46.9
Compound 15 17.09 /
Compound 16 8.5 17.09
Formic acid salt of Compound 17 12.44 20.54
Formate salt of Compound 18 / 37.17
Formate salt of Compound 19 13.89 20.51
Compound 20 / 33.56
Compound 21 1.79 19.47
Compound 22 4.22 12.76
Compound 23 / 48.71
Compound 24 / 14.81
"/" indicates that no test results have been obtained.
And (4) experimental conclusion: the compound has good inhibition activity on AXL and VEGFR2 kinase.
Experimental example 3: n-octanol/water partition coefficient pH7.4 (Log D) 7.4 )
Experimental materials:
DMSO stock solution 10Mm DMSO stock solutions of test compounds and controls (nadolol, meting, propranolol, quinidine, amitriptyline, chlorpromazine) were formulated
The buffer solution is 100mM phosphate buffer solution, pH7.4, DMSO% is 1%; n-octanol saturated buffer (pH 7.4) solution; buffer (pH7.4) saturated n-octanol solution
And (3) experimental operation:
1. test compounds (10 mM DMSO solution, 2. Mu.L/well) and QC samples (10 mM DMSO solution; 2. Mu.L/well) were added to 96-well polypropylene tubes from the stock solutions, respectively;
2. adding n-octanol saturated buffer solution (150. Mu.L/well) and buffer (pH7.4) saturated n-octanol (150. Mu.L/well) solution into each well;
3. stirring strongly for one minute, then shaking for one minute at 600 rpm and 28 ℃;
4. centrifuging at 2500 rpm for 10 min
5. N-octanol layer diluted Y times (200 times), buffer layer diluted X times (20 times)
6. The test mix concentration of the filtrate was determined by LCMS/MS.
The Log D value of a compound is calculated as follows:
Figure PCTCN2021079337-APPB-000094
O-S: area of peak of sample in normal Xin Chunceng
O-IS: peak area of internal standard in n Xin Chunceng
B-S: peak area of sample in buffer layer
B-IS: peak area of internal standard in buffer layer
Y: dilution factor of sample in positive Xin Chunceng
X: dilution factor of sample in buffer layer
Note: sample analysis was performed using a triple-pole quadrupole mass spectrometer. Injecting the diluted sample into a small column-shaped high performance liquid chromatography column by using an isochoric water loaded solvent, and then flushing the sample into a mass spectrometer by using a rapid gradient to obtain an organic elution solvent. The peak areas were corrected using a dilution factor and the ratio of the corrected peak areas was used to calculate the results (Log D values) in combination with internal standards.
The experimental results are as follows: see table 2.
TABLE 2 Log D 7.4 Test results
Test compounds Sitravatinib Formate salt of Compound 3
Log D 7.4 >4.0 3.65
And (4) experimental conclusion: log D 7.4 Is the oil-water distribution coefficient of the compound at pH7.4, representing the physicochemical properties of the compound, log D 7.4 About 3 is considered to have the best druggability. Therefore, compared with Sitravatinib, the compound provided by the invention has better physicochemical properties.
Experimental example 4: in vitro pharmacokinetic Studies
(1) MDR1-MDCK cell two-way permeability assessment experiment
The MDR1-MDCKII cell line authorized by Piet Borst laboratory of Dutch cancer research institute is used as an in vitro model of experiment, and is a Madin-Darby canine kidney cell transfected with human multi-drug resistance gene (MDR 1), and the cell can stably express efflux transporter P-gp, so that the method is suitable for screening P-gp substrates or inhibitors, and can predict the permeability of compounds with high efflux barrier in duodenum, blood brain barrier, hepatocyte nucleus, kidney unit and the like. The objective of this study was to determine the bi-directional permeability of the compounds of the invention through a model of MDR1-MDCK II cells using MDR1-MDCK II cells.
Experimental operation: the experimental standard conditions were as follows:
test concentration 2. Mu.M (DMSO. Ltoreq.1%);
-repeating: n =2;
-a direction: bidirectional transport, including both A → B and B → A directions;
-incubation time: single time point, 2.5 hours;
-a transport buffer: HBSS buffer containing 10mM Hepes, pH7.4;
-incubation conditions: 37 ℃,5% of CO 2
After incubation, the sample solutions taken from the dosing and receiving wells were immediately mixed with a cold acetonitrile solution containing an internal standard. The concentration of the test compound in all samples (including initial dosing solution, dosing well supernatant, receiving solution) was analyzed by LC/MS/MS method. And calculating parameters such as apparent permeability coefficient, external discharge ratio and the like.
The experimental results are as follows: table 3 lists the permeability coefficients of Sitravatinib and compound 3 in MDR1-MDCK II monolayers. The results indicate that compound 3 has moderate permeability, while Sitravatinib has low permeability.
TABLE 3 results of Permeability experiments
Figure PCTCN2021079337-APPB-000095
(2) Cytochrome P450 isoenzyme inhibitory study
The purpose of the experiment is as follows: the inhibitory effect of the test compounds on the activity of human liver microsomal cytochrome P450 isozymes (CYP 1A2, CYP2C9, CYP2C19, CYP2D6, and CYP3 A4) was measured.
Experimental operation: the test compound (10 mM) was first subjected to a gradient to prepare working solutions (100 Xfinal concentrations) having the following concentrations: 5. 1.5, 0.5, 0.15, 0.05, 0.015 and 0.005mM, and simultaneously preparing working solutions of each positive inhibitor of P450 isozymes (CYP 1A2, CYP2C9, CYP2C19, CYP2D6 and CYP3A 4) and a specific substrate mixture (5in 1) thereof; thawing human liver microsomes frozen in a refrigerator at-80 deg.C on ice, diluting with PB to obtain working solution (0.253 mg/ml) with certain concentration after human liver microsomes are completely dissolved; adding 20 mu L of substrate mixed solution into a reaction plate (20 mu L of PB is added into a Blank hole), simultaneously adding 158 mu L of human liver microsome working solution into the reaction plate, and placing the reaction plate on ice for later use; at this time, 2 μ l of each concentration of test compound (N = 1) and specific inhibitor (N = 2) was added to the corresponding well, and the group without inhibitor (test compound or positive inhibitor) was added to the corresponding organic solvent as a control sample (test compound control sample is 1; after pre-incubation in a 37 ℃ water bath for 10 minutes, 20. Mu.l of a coenzyme factor (NADPH) solution was added to the reaction plate, and incubated in a 37 ℃ water bath for 10 minutes; the reaction was stopped by adding 400. Mu.L of cold acetonitrile solution (internal standard 200ng/mL Tolbutamide and Labetalol); placing the reaction plate on a shaking table, and oscillating for 10 minutes; 4,000rpm for 20 minutes; adding 200 mu L of supernatant into 100 mu L of water for sample dilution; finally, sealing the plate, oscillating, shaking up, and performing LC/MS/MS detection.
The experimental results are as follows: the compound and Sitravatinib have no obvious inhibition effect on CYP1A2, and have moderate inhibition effect on CYP2C9 and CYP2C 19. The compound has weak inhibition and no inhibition on CYP2D6 and CYP3A4 (substrate is midazolam) respectively, while Sitravatinib has strong inhibition on CYP2D6 and CYP3A4 (substrate is midazolam).
TABLE 4 cytochrome P450 isoenzyme inhibitory test results
Figure PCTCN2021079337-APPB-000096
(3) Plasma protein binding Rate Studies
The purpose of the experiment is as follows: determination of the protein binding Rate of test Compounds in human, CD-1 mouse and SD rat plasma
And (3) experimental operation: a sample of 796. Mu.L of blank plasma from human, CD-1 mice and SD rats was added with 4. Mu.L of test compound working solution (400. Mu.M) or warfarin working solution (400. Mu.M) to give a final concentration of 2. Mu.M of both test compound and warfarin in the plasma sample. The samples were mixed thoroughly. The final concentration of organic phase DMSO is 0.5%; 50 μ L of test compound and warfarin plasma samples were pipetted into sample receiving plates (three in parallel) and corresponding volumes of blank plasma or buffer were immediately added such that the final volume of each sample well was 100 μ L, plasma: the volume ratio of dialysis buffer was 1:1, and 400. Mu.L of stop buffer was added to these samples, which were used as T0 samples for recovery and stability determination. Storing the T0 sample at 2-8 ℃ for subsequent treatment with other dialyzed samples; mu.L of test compound and warfarin plasma samples were added to the administration end of each dialysis well, and 150. Mu.L of blank dialysis buffer was added to the receiving end of the corresponding dialysis well. Then sealing the dialysis plate with a gas permeable membrane and placing in a moist, 5% CO 2 The incubator of (1) is incubated at 37 ℃ for 4-hr with shaking at about 100 rpm. After dialysis, 50. Mu.L of the dialyzed buffer sample and the dialyzed plasma sample were transferred to a new sample receiving plate. Corresponding blank plasma or buffer was added to the samples in corresponding volumes such that the final volume of each sample well was 100 μ L, plasma: the volume ratio of the dialysis buffer was 1:1. All samples were subjected to LC/MS/MS analysis after protein precipitation and analyzed by the formula: % Unbound =100 × fc/TC; % Bound =100-% unbonded; protein binding rates and Recovery were calculated as% Recovery =100 × (FC + TC)/T0.
And (4) experimental conclusion: both formate salt of compound 3 and Sitravatinib exhibited higher plasma protein binding rates. The protein binding rates of compound 3 in plasma of human, CD-1 mouse and SD rat were 99.7%, 99.7% and 99.8%, respectively, and the protein binding rates of Sitravatinib in plasma of human, CD-1 mouse and SD rat were 99.95%, 99.97% and NA, respectively (the values cannot be calculated, since no free compound was detected at the free end).
(4) Microsomal metabolic stability study
Purpose of the experiment: the stability of the test compounds in liver microsomes of human, CD-1 mice and SD rats was determined.
Experimental operation: firstly, preparing 8 96-well plates which are named as T0, T5, T10, T20, T30, T60, NCF60 and BLANK respectively; with the exception of BLANK (plus 10. Mu.L/well buffer), 10. Mu.L/well of the drug solution was added to each plate; the prepared microsomes were added to 7 plates (80. Mu.L/well) except for the T0 plate; add 10. Mu.L/well buffer to NCF60 plates, incubate in 37 degree water bath, start timing:
Figure PCTCN2021079337-APPB-000097
then, the prepared NADPH cofactor is dispensed into a 96 Kong Jian pore plate as a sample adding groove, each plate is placed into a water bath kettle at 37 ℃ by using a 96 channel pipettor, and 10 mu L/pore of each plate is incubated, so that the reaction is started:
Figure PCTCN2021079337-APPB-000098
adding 300 (mu L/hole) stop solutions (cold acetonitrile containing 100ng/mL tolbutamide and 100ng/mL labetalol as internal standards) at each time point to stop the reaction, and uniformly mixing; centrifuging for 20 minutes at 4000rpm of a centrifuge to precipitate protein; after centrifugation, 100. Mu.L/well of the supernatant was pipetted using a 96-channel pipette into a new plate with 300. Mu.L/well of HPLC water, mixed well and sent to the analyst for detection by LC/MS/MS.
The experimental results are as follows: the remaining percentage of formate salt of compound 3 incubated in human, CD-1 mouse and SD rat liver microsomes for 60 minutes was: 65.7%, 63.3% and 64.8%, the remaining percentage of Sitravatinib incubated in human, CD-1 mouse and SD rat liver microsomes for 60 minutes was: 77.3%, 10.2% and 34.4%. Compound 3 was metabolized at a moderate rate in all three species, sitravatinib was metabolized at a moderate rate in human and SD rat liver microsomes, and was metabolized more rapidly in CD-1 mouse liver microsomes.
The results of in vitro DMPK property evaluations are summarized in table 5:
TABLE 5 summary of in vitro DMPK Properties
Figure PCTCN2021079337-APPB-000099
And (4) experimental conclusion: the compound of the invention is obviously superior to the AXL & VEGFR2 inhibitor Sitravatinib in clinical stage III in liver microsome stability (MMS), plasma protein binding rate (PPB), drug interaction (DDI) and in vitro permeability, which means that the series of compounds have better metabolic stability; the improvement in permeability means that the compounds of the series will be more absorbable, while there is less risk of drug resistance due to active efflux; the series of compounds have lower CYP inhibitory activity, good drug interaction property, smaller safety risk of clinical combination with other drugs and stronger developability.
Experimental example 5: in vivo pharmacokinetic Studies
Purpose of the experiment: pharmacokinetics study of CD-1 mice after oral and intravenous injection of compound 3 or Sitravatinib
Experimental operation: an appropriate amount of formate salt of compound 3 or Sitravatinib was weighed, dissolved in a solvent 5% DMSO/95% (20% HP- β -CD), respectively, stirred and sonicated to prepare a clear solution of 0.4mg/mL, which was filtered through a microporous membrane for use. CD-1 male mice of 7-9 weeks old are selected and administered by intravenous injection, with the administration volume of 5mL/kg and the dosage of 2mg/kg.
Compound 3 or Sitravatinib was weighed out in an appropriate amount, dissolved in a solvent at a ratio of 5% DMSO/95% (20% HP- β -CD), respectively, and subjected to stirring and sonication to prepare a clear solution of 1 mg/mL. CD-1 male mice of 7-9 weeks old are selected and orally administered, with the administration volume of 10mL/kg and the dose of 10mg/kg.
Whole blood was collected for a certain period of time, plasma was prepared, drug concentration was analyzed by LC-MS/MS method, and drug parameters were calculated using Phoenix WinNonlin software (Pharsight, USA).
The experimental results are as follows:
after a single intravenous injection of 2mg/kg Sitravatinib and compound 3 in male CD-1 mice, the plasma Clearance (CL) was 7.21 and 6.18mL/min/kg, and the steady-state apparent distribution volume (Vd) ss ) 2.24 and 0.72L/kg, elimination half-life (T) 1/2 ) 3.87 and 1.58h, respectively, the area under the plasma concentration curve from point 0 to the last quantifiable time point (AUC) 0-last ) The values of (A) are 7284 and 9645nM · h, respectively.
After the male CD-1 mice are administrated with 10mg/kg of Sitravatinib and the compound 3 by single intragastric administration, the bioavailability is 65.8 percent and 109 percent respectively, and the AUC 0-last 24003 and 53755nM · h, respectively, peak concentration (C) max ) At 2530 and 10600nM, the time to peak occurred at 2.00 and 1.00h post-dose. The results of in vivo PK property evaluations are summarized in table 6.
TABLE 6 summary of PK Properties in vivo
Figure PCTCN2021079337-APPB-000100
And (4) experimental conclusion:
the compound of the invention has better oral maximum blood concentration C max Half-life, clearance (CL), AUC, and oral bioavailability.

Claims (14)

  1. A compound of formula (I) or a pharmaceutically acceptable salt thereof,
    Figure PCTCN2021079337-APPB-100001
    wherein the content of the first and second substances,
    ring a is selected from a 5-membered heteroaryl group, said 5-membered heteroaryl group comprising 2N heteroatoms;
    alternatively, ring a is selected from 6-membered heteroaryl, said 6-membered heteroaryl comprising up to 2N atoms;
    T 1 and T 2 Are each independently selected from C (R) 5 ) And N;
    R 1 and R 3 Are each independently selected from H, F and CH 3
    R 2 H, F and Cl;
    R 4 each independently selected from H, C 1~6 Alkyl radical, C 1~6 Heteroalkyl group, C 3~6 Cycloalkyl, 3-to 6-membered heterocycloalkyl and-L-R 6 Said C is 1~6 Alkyl radical, C 1~6 Heteroalkyl group, C 3~6 Cycloalkyl, 3-to 6-membered heterocycloalkyl and-L-R 6 Optionally substituted with 1,2 or 3R;
    R 5 selected from H and Cl;
    R 6 selected from H, OH, 5-to 6-membered heterocycloalkyl and C 1~3 An alkyl group;
    l is selected from-CH 2 -、-CH 2 -CH 2 -and
    Figure PCTCN2021079337-APPB-100002
    n is selected from 0, 1,2 and 3;
    r is respectively and independently selected from F, cl, br, I, OH, CN, COOH and NH 2 、-NHCH 3 、-N(CH3) 2 、CH 3 、CH 2 CH 3 、CF 3 、-OCH 3 、-OCH 2 CH 3 、-O-CH(CH 3 ) 2 、-C(=O)OCH 3 、-C(=O)CH 3 and-C (= O) CH 2 CH 3
    The 5-membered heteroaryl, 6-membered heteroaryl, C 1~6 The heteroalkyl and 3-to 6-membered heterocycloalkyl each independently contain 1,2, or 3 heteroatoms independently selected from O, S, N and NH orA heteroatom group.
  2. A compound according to claim 1, or a pharmaceutically acceptable salt thereof, wherein ring a is selected from pyridyl, pyrazolyl and imidazolyl.
  3. A compound or pharmaceutically acceptable salt thereof according to claim 1 or 2, wherein R 4 Each independently selected from H, C 1~4 Alkyl radical, C 1~4 Heteroalkyl, 5-to 6-membered heterocycloalkyl and-L-R 6 Said C is 1~4 Alkyl radical, C 1~4 Heteroalkyl, 5-to 6-membered heterocycloalkyl and-L-R 6 Optionally substituted with 1,2 or 3R.
  4. A compound or pharmaceutically acceptable salt thereof according to claim 3, wherein R 4 Each independently selected from H,
    Figure PCTCN2021079337-APPB-100003
    Figure PCTCN2021079337-APPB-100004
    Figure PCTCN2021079337-APPB-100005
    The above-mentioned
    Figure PCTCN2021079337-APPB-100006
    Figure PCTCN2021079337-APPB-100007
    Optionally substituted with 1,2 or 3R.
  5. A compound or pharmaceutically acceptable salt thereof according to claim 4, wherein R 4 Each independently selected from H,
    Figure PCTCN2021079337-APPB-100008
    Figure PCTCN2021079337-APPB-100009
  6. A compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein the building block
    Figure PCTCN2021079337-APPB-100010
    Is selected from
    Figure PCTCN2021079337-APPB-100011
    Figure PCTCN2021079337-APPB-100012
  7. A compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein the building block
    Figure PCTCN2021079337-APPB-100013
    Is selected from
    Figure PCTCN2021079337-APPB-100014
  8. A compound according to claim 7, or a pharmaceutically acceptable salt thereof, wherein the building block
    Figure PCTCN2021079337-APPB-100015
    Is selected from
    Figure PCTCN2021079337-APPB-100016
    Figure PCTCN2021079337-APPB-100017
  9. The compound according to any one of claims 1 to 5, wherein the compound is selected from the group consisting of
    Figure PCTCN2021079337-APPB-100018
    Wherein the content of the first and second substances,
    R 41 selected from H, C 1~4 Alkyl radical, C 1~4 Heteroalkyl, 6-membered heterocycloalkyl and-L-R 6 H, C as described 1~4 Alkyl radical, C 1~4 Heteroalkyl, 6-membered heterocycloalkyl and-L-R 6 Optionally substituted with 1,2 or 3R;
    R 42 selected from H and-L-R 6 said-L-R 6 Optionally substituted with 1,2 or 3R;
    R 6 selected from H, OH, 6-membered heterocycloalkyl and C 1~3 An alkyl group;
    R 2 l and R are as defined in claim 1.
  10. The compound according to any one of claims 1 to 5, wherein the compound is selected from the group consisting of
    Figure PCTCN2021079337-APPB-100019
    R 1 、R 2 、R 3 And R 4 As defined in claims 1-5.
  11. A compound or a pharmaceutically acceptable salt thereof selected from
    Figure PCTCN2021079337-APPB-100020
    Figure PCTCN2021079337-APPB-100021
  12. A pharmaceutical composition comprising a therapeutically effective amount of a compound or pharmaceutically acceptable salt according to any one of claims 1 to 11 as an active ingredient and a pharmaceutically acceptable carrier.
  13. Use of a compound or pharmaceutically acceptable salt according to any one of claims 1 to 11 or a composition according to claim 12 for the manufacture of an Axl and/or VEGFR-2 inhibitor.
  14. The use according to claim 13 wherein the Axl and/or VEGFR-2 inhibitor is a medicament for the treatment of cancer.
CN202180020076.9A 2020-03-10 2021-03-05 Vinyl-substituted pyridines Pending CN115244041A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2020101637099 2020-03-10
CN202010163709 2020-03-10
PCT/CN2021/079337 WO2021180006A1 (en) 2020-03-10 2021-03-05 Vinyl-substituted pyridine compounds

Publications (1)

Publication Number Publication Date
CN115244041A true CN115244041A (en) 2022-10-25

Family

ID=77670984

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180020076.9A Pending CN115244041A (en) 2020-03-10 2021-03-05 Vinyl-substituted pyridines

Country Status (2)

Country Link
CN (1) CN115244041A (en)
WO (1) WO2021180006A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102256493A (en) * 2008-10-29 2011-11-23 迪赛孚尔制药有限公司 Cyclopropane amides and analogs exhibiting anti-cancer and anti-proliferative activities
CN102964308A (en) * 2012-11-30 2013-03-13 中国药科大学 Novel pyrimidine compound, preparation method thereof, pharmaceutical composition containing novel pyrimidine compound and application of novel pyrimidine compound
CN107793363A (en) * 2016-09-06 2018-03-13 上海医药工业研究院 A kind of substituted aromatic amines base heteroaromatic class compound and its application as antineoplastic
CN108530464A (en) * 2017-03-02 2018-09-14 深圳海王医药科技研究院有限公司 A kind of multiple target point kinase inhibitor
WO2019148036A1 (en) * 2018-01-26 2019-08-01 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110862397A (en) * 2018-08-27 2020-03-06 北京赛特明强医药科技有限公司 Dioxane quinazoline and dioxane quinoline compounds, and preparation method and application thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102256493A (en) * 2008-10-29 2011-11-23 迪赛孚尔制药有限公司 Cyclopropane amides and analogs exhibiting anti-cancer and anti-proliferative activities
CN102964308A (en) * 2012-11-30 2013-03-13 中国药科大学 Novel pyrimidine compound, preparation method thereof, pharmaceutical composition containing novel pyrimidine compound and application of novel pyrimidine compound
CN107793363A (en) * 2016-09-06 2018-03-13 上海医药工业研究院 A kind of substituted aromatic amines base heteroaromatic class compound and its application as antineoplastic
CN108530464A (en) * 2017-03-02 2018-09-14 深圳海王医药科技研究院有限公司 A kind of multiple target point kinase inhibitor
WO2019148036A1 (en) * 2018-01-26 2019-08-01 Exelixis, Inc. Compounds for the treatment of kinase-dependent disorders

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
STEPHEN K.CHIA,ET AL.: "A phase-I study of lapatinib in combination with foretinib, a c-MET, AXL and vascular endothelial growth factor receptor inhibitor, in human epidermal growth factor receptor 2(HER-2)-positive metastatic breast cancer", BREAST CANCER RESEARCH, pages 1 - 8 *

Also Published As

Publication number Publication date
WO2021180006A1 (en) 2021-09-16

Similar Documents

Publication Publication Date Title
CN113365988B (en) SHP2 inhibitor and application thereof
CN110054618B (en) Pyrimidine or pyridine compound, preparation method and medical application thereof
TWI585088B (en) Imidazo[1,2-b]pyridazine analogues as kinase inhibitors
CN114846005B (en) SHP2 inhibitor and application thereof
JP5894714B1 (en) An activating mutant quinazoline inhibitor of epidermal growth factor receptor
CN110156786A (en) Pyrimido cycle compound and its preparation method and application
WO2013170770A1 (en) Acetylene derivatives having antitumor activity
EP2739618A1 (en) Quinazoline compounds as serine/threonine kinase inhibitors
CN105315285A (en) 2,4-disubstituted 7H-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medical uses thereof
JP2021517555A (en) Phenyl-2-hydroxy-acetylamino-2-methyl-phenyl compound
CN109689645A (en) Cyanoindole quinoline derivant as NIK inhibitor
JP2021536436A (en) A novel inhibitor prepared from quinoline derivatives
CN102548970A (en) Novel (6-oxo-1, 6-dihydro-pyrimidin-2-yl)-amide derivatives, preparation thereof, and pharmaceutical use thereof as AKT (PKB) phosphorylation inhibitors
WO2020182018A1 (en) Nitrogen heterocyclic compound, preparation method therefor and use thereof
CN115916765A (en) Pyridine or pyrimidine derivative and preparation method and application thereof
WO2019137201A1 (en) Heteroarylo tetrahydropyridine compound and preparation method, pharmaceutical composition, and application thereof
CN110066276A (en) Heteroaromatic compound, wherein mesosome, preparation method, pharmaceutical composition and application
WO2020135878A1 (en) Imidazopyridine derivative as fgfr and vegfr dual inhibitor
CN110903283B (en) Substituted quinazoline compound, pharmaceutical composition containing compound and application of compound
TW202227441A (en) Quinazoline compound and pharmaceutical composition thereof
CN115322158B (en) As KRASG12CSubstituted quinazoline compounds of protein inhibitor
CN115244041A (en) Vinyl-substituted pyridines
CN107286140A (en) Substituted aromatic amines base heteroaromatic class compound and its application as antineoplastic
JP2020526593A (en) Crystal form, salt form and production method of N-phenyl-2-aminopyrimidine compounds
JP7477846B2 (en) N-containing heteroaryl derivative and pharmaceutical composition for preventing or treating cancer containing the same as an active ingredient

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
WD01 Invention patent application deemed withdrawn after publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20221025