CN112574183B - PD-1 inhibitor and preparation method and application thereof - Google Patents

PD-1 inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN112574183B
CN112574183B CN201910936108.4A CN201910936108A CN112574183B CN 112574183 B CN112574183 B CN 112574183B CN 201910936108 A CN201910936108 A CN 201910936108A CN 112574183 B CN112574183 B CN 112574183B
Authority
CN
China
Prior art keywords
compound
cancer
formula
acid
synthesis
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.)
Active
Application number
CN201910936108.4A
Other languages
Chinese (zh)
Other versions
CN112574183A (en
Inventor
宋志春
何东伟
包金远
张孝清
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.)
Nanjing Huawe Medicine Technology Group Co Ltd
Original Assignee
Nanjing Huawe Medicine Technology Group Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nanjing Huawe Medicine Technology Group Co Ltd filed Critical Nanjing Huawe Medicine Technology Group Co Ltd
Priority to CN201910936108.4A priority Critical patent/CN112574183B/en
Publication of CN112574183A publication Critical patent/CN112574183A/en
Application granted granted Critical
Publication of CN112574183B publication Critical patent/CN112574183B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no 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, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no 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, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D211/60Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D319/00Heterocyclic compounds containing six-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D319/101,4-Dioxanes; Hydrogenated 1,4-dioxanes
    • C07D319/141,4-Dioxanes; Hydrogenated 1,4-dioxanes condensed with carbocyclic rings or ring systems
    • C07D319/161,4-Dioxanes; Hydrogenated 1,4-dioxanes condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • 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
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/10Spiro-condensed systems

Abstract

The invention relates to a compound shown in formula I, and racemate, stereoisomer, tautomer, isotopic marker, nitrogen oxide, solvate, polymorph, metabolite, ester, prodrug or pharmaceutically acceptable salt thereof, a pharmaceutical composition containing the compound, a preparation method thereof, and medical application thereof, wherein the structure of the formula I is as follows:

Description

PD-1 inhibitor and preparation method and application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to a PD-1 inhibitor, and a preparation method and application thereof.
Background
PD-1 is known as programmed death receptor 1, is an important immunosuppressive molecule and is a member of the CD28 superfamily. PD-L1, collectively referred to as programmed death receptor-ligand 1, has been shown to interact with CD 80. The interaction of PD-L1/CD80 with expressing immune cells has been shown to be inhibitory.
The immunotherapy based on PD1/PDL1 is a new generation immunotherapy which is currently spotlighted, aims to utilize the immune system of the human body to resist tumors, induces apoptosis by blocking a PD-1/PD-L1 signal channel, and has the potential of treating various types of tumors. Currently, there is a need to develop PD-1 inhibitors with good activity and suitable for druggability.
Disclosure of Invention
In order to solve the problems in the prior art, the invention provides a compound shown as formula I, and racemate, stereoisomer, tautomer, isotopic marker, nitrogen oxide, solvate, polymorph, metabolite, ester, prodrug or pharmaceutically acceptable salt thereof:
Figure BDA0002221622080000011
wherein, R is1Selected from the following groups optionally substituted with one, two or more R:
Figure BDA0002221622080000012
the R is2Selected from H or the following optionally substituted with one, two or more R: (C)1-C12) Aliphatic hydrocarbon radical, optionally containing one, two or more heteroatoms (C)1-C12) An aliphatic hydrocarbon group;
each R3Each independently selected from halo or-W-Rs optionally substituted by one, two or more R;
said Rs is selected from H, C3-12Cycloalkyl, 3-to 12-membered heterocyclyl, C6-20Aryl or 5-14 membered heteroaryl;
n is selected from 0, 1,2, 3 and 4;
r is selected from halogen, CN, OH, SH, NH2COOH, O, NH ═ OH (N ═ or selected from anyOptionally substituted with one, two or more R': (C)1-C12) Aliphatic hydrocarbon radical, optionally containing one, two or more heteroatoms (C)1-C12) Aliphatic hydrocarbon radical, C3-12Cycloalkyl, 3-12 membered heterocyclyl, C6-20Aryl or 5-14 membered heteroaryl; or two substituents R attached to the same atom form a 3-12 membered heterocyclic group;
r' is selected from halogen, CN, OH, SH, NH2、COOH、=O;
The R is4、R5Selected from H or optionally substituted with one, two or more R: (C)1-C12) Aliphatic hydrocarbon radical, optionally containing one, two or more heteroatoms (C)1-C12) Aliphatic hydrocarbon radical, C3-12Cycloalkyl, 3-12 membered heterocyclyl, C6-20Aryl or 5-14 membered heteroaryl; or R4、R5Together with the N atom to which they are attached form an N-containing 3-12 membered heterocycloalkyl or N-containing 5-14 membered heteroaryl unsubstituted or optionally substituted with one, two or more R;
the-W-is selected from- (C)1-C12) Aliphatic hydrocarbon group-, -O- (C)1-C12) Aliphatic hydrocarbyl-, - (C)1-C12) Aliphatic hydrocarbon radical-O-, -S- (C)1-C12) Aliphatic hydrocarbyl-, - (C)1-C12) Aliphatic hydrocarbon radical-S-, -NH- (C)1-C12) Aliphatic hydrocarbyl-, - (C)1-C12) Aliphatic hydrocarbyl-NH-.
In accordance with an embodiment of the present invention,
said "optionally comprising one, two or more heteroatoms of (C)1-C12) Aliphatic hydrocarbon groups ", the heteroatoms may be selected from sulphur, nitrogen, oxygen, phosphorus and silicon, optionally the heteroatoms are inserted in the aliphatic hydrocarbon groups, optionally C-C bonds and C-H bonds; for example, may be selected from (C)1-C12) Aliphatic hydrocarbyloxy, (C)1-C12) Aliphatic hydrocarbyl mercapto group, (C)1-C6) Aliphatic hydrocarbyloxy group (C)1-C6) Aliphatic hydrocarbyl mercapto group, (C)1-C6) Aliphatic hydrocarbyloxy (C)1-C6) FatHydrocarbyl radical, (C)1-C6) Aliphatic hydrocarbyl mercapto (C)1-C6) Aliphatic hydrocarbon group, N- (C)1-C3) Aliphatic hydrocarbyl amino group (C)1-C6) Aliphatic hydrocarbon group, N-di- (C)1-C3) Aliphatic hydrocarbyl amino group (C)1-C6) An aliphatic hydrocarbon group;
said (C)1-C12) The aliphatic hydrocarbon group may be selected from (C)1-C12) Alkyl, (C)2-C12) Alkenyl, (C)2-C12) Alkynyl groups, in some embodiments, can be selected from (C)1-C6) Alkyl, (C)2-C6) Alkenyl, (C)2-C6) Alkynyl.
In accordance with an embodiment of the present invention,
the R is1Selected from:
Figure BDA0002221622080000021
preferably, said R is1Is selected from
Figure BDA0002221622080000022
Figure BDA0002221622080000023
Figure BDA0002221622080000031
Said R is2Selected from the group consisting of methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, ethenyl, 1-propenyl, 2-propenyl, 1-methylethenyl, 1-butenyl, 1-ethylethenyl, 1-methyl-2-propenyl, 2-butenyl, 3-butenyl, 2-methyl-1-propenyl, 2-methyl-2-propenyl, 1-pentenyl, 1-hexenyl, ethynyl, 1-propynyl,2-propynyl, 1-butynyl, 1-methyl-2-propynyl, 3-butynyl, 1-pentynyl, 1-hexynyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, methoxy, ethoxy, propoxy, butoxy, pentoxy, methoxymethyl, ethoxymethyl, propoxymethyl, methoxyethyl, ethoxyethyl, propoxyethyl, methoxypropyl, ethoxypropyl, propoxypropyl, N-methylaminomethyl, N-methylaminoethyl, N-ethylaminoethyl, N-dimethylaminomethyl, N-dimethylaminoethyl, N-diethylaminoethyl, amino, N-dimethylamino, N-diethylamino;
the R is3Selected from halogen, -O- (C)1-C12) An aliphatic hydrocarbon group,
Figure BDA0002221622080000032
Figure BDA0002221622080000033
Preferably, said R is3Selected from halogen,
Figure BDA0002221622080000034
Figure BDA0002221622080000035
-O-(C1-C6) An aliphatic hydrocarbon group;
more preferably, R3Selected from halogen,
Figure BDA0002221622080000036
Figure BDA0002221622080000041
Methoxy, ethoxy, propoxy, butoxy, pentoxy;
the-NR4R5Is selected from
Figure BDA0002221622080000042
Figure BDA0002221622080000043
Wherein m is selected from 0, 1,2, 3;
r is selected from halogen, CN, OH, SH, NH2COOH, ═ O, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, ethenyl, 1-propenyl, 2-propenyl, 1-methylethenyl, 1-butenyl, 1-ethylethenyl, 1-methyl-2-propenyl, 2-butenyl, 3-butenyl, 2-methyl-1-propenyl, 2-methyl-2-propenyl, 1-pentenyl, 1-hexenyl, ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 1-methyl-2-propynyl, 3-butynyl, 1-pentynyl, 1-hexynyl, cyclopropyl, cyclobutyl, cyclopentyl, Cyclohexyl, methoxy, ethoxy, propoxy, butoxy, pentoxy, methoxymethyl, ethoxymethyl, propoxymethyl, methoxyethyl, ethoxyethyl, propoxyethyl, methoxypropyl, ethoxypropyl, propoxypropyl, N-methylaminomethyl, N-methylaminoethyl, N-ethylaminoethyl, N-dimethylaminomethyl, N-dimethylaminoethyl, N-diethylaminoethyl, amino, N-dimethylamino, N-diethylamino, tetrahydropyrrolyl, piperidinyl, pyridinyl, pyrazinyl, pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl;
the halogen is selected from F, Cl, Br and I;
according to an embodiment of the invention, the structure of formula I is further selected from the following formulae II, III, IV, V, VI, VII:
Figure BDA0002221622080000044
Figure BDA0002221622080000051
in the formula II, the formula III, the formula IV, the formula V, the formula VI and the formula VII, R1、R2、R3、R4、R5W, n are as defined above for formula I.
According to an embodiment of the present invention, in the compound represented by formula I and its racemate, stereoisomer, tautomer, isotopic label, nitrogen oxide, solvate, polymorph, metabolite, ester, prodrug, or pharmaceutically acceptable salt thereof, illustrative, non-limiting specific examples of the compound represented by formula I are as follows:
Figure BDA0002221622080000052
Figure BDA0002221622080000061
Figure BDA0002221622080000071
Figure BDA0002221622080000081
the invention also provides a preparation method of the compound shown in the formula I (including the compounds shown in the formulas II to VII) and racemate, stereoisomer, tautomer, isotopic marker, nitrogen oxide, solvate, polymorphic substance, metabolite, ester, prodrug or pharmaceutically acceptable salt thereof, but not limited to the methods described below. All starting materials are prepared or purchased directly according to the general rules of the target molecule and by protocols in these routes, methods well known to those of ordinary skill in the art of organic chemistry. The compounds of the invention can be synthesized by combining the methods described below with synthetic methods known in the art of synthetic organic chemistry or variations thereon as recognized by those skilled in the art.
In a first embodiment, the preparation of the compounds of the invention comprises the following steps:
Figure BDA0002221622080000082
wherein R is1、R2、R3、R4、R5W, n are as defined above for formula I.
According to an embodiment of the invention, said first variant, a reductive amination reaction is used; in some embodiments, compound M-1 is reacted with compound M-2 in the presence of a weak acid, a reducing agent to produce a compound of formula I; the weak acid can be selected from acetic acid, oxalic acid, diluted hydrochloric acid and the like, the reducing agent can be selected from sodium cyanoborohydride, sodium borohydride, sodium triacetoxyborohydride and the like, the reaction solvent for the reaction can be selected from acetonitrile, DMF and the like, and the reaction temperature can be 0-50 ℃.
In a second embodiment, the preparation of the compounds of the invention comprises the following steps:
Figure BDA0002221622080000083
wherein R is1、R2、R3W, n are as defined above for formula I; x is selected from halogens, such as F, Cl, Br, I.
According to an embodiment of the invention, said second scheme, compound M-1 is synthesized using a Suzuki coupling reaction; in some embodiments, the compounds L-1 and R1-X reacts under the action of a palladium catalyst and a ligand to form a compound M-1; the palladium catalyst may be Pd (PPh)3)2、PdCl2(PPh3)2、PdCl2(MeCN)2Etc.; the reaction can be carried out in the presence of a base, wherein the base can be potassium carbonate, potassium phosphate, potassium acetate and the like; the solvent for the reaction can be selected from 1, 4-dioxane, N, N-dimethylformamide and the like, and the reaction temperature can be 0-100 ℃.
In a third embodiment, the preparation of the compounds of the invention comprises the steps of:
Figure BDA0002221622080000091
wherein R is2、R3W, n are as defined above for formula I.
According to an embodiment of the present invention, said third scheme, compound L-1(W is-CH) is synthesized using Mitsunobu reaction (Mitsunobu reaction)2-O-), in some embodiments, compound K-1 and compound K-2 are reacted under the action of triphenylphosphine (PPh3) and diethyl azodicarboxylate (DEAD) to form compound L-1; the solvent for the reaction is preferably tetrahydrofuran, and the reaction temperature may be-20 to 50 ℃.
In a fourth embodiment, the preparation of the compounds of the invention comprises the steps of:
Figure BDA0002221622080000092
wherein R is1,R2、R3W, n are as defined above for formula I; x is selected from halogens, such as F, Cl, Br, I.
According to the fourth embodiment of the present invention, compound N-1 is obtained by Suzuki coupling reaction of compound K-1, and Mitsunobu reaction is further performed with compound K-2 to produce compound M-1(W is-CH)2-O-). The Suzuki coupling reaction and the Mitsunobu reaction are carried out under the same reaction conditions of the two and three phases of the scheme
The present invention further provides a pharmaceutical composition comprising a compound of formula I as described herein and racemates, stereoisomers, tautomers, isotopic labels, nitrogen oxides, solvates, polymorphs, metabolites, esters, prodrugs or pharmaceutically acceptable salts thereof.
In some embodiments, the pharmaceutical compositions of the present invention further comprise a therapeutically effective amount of a compound of formula I of the present invention and racemates, stereoisomers, tautomers, isotopic labels, nitrogen oxides, solvates, polymorphs, metabolites, esters, prodrugs thereof, or pharmaceutically acceptable salts thereof, and a pharmaceutically acceptable carrier.
The carrier in the pharmaceutical composition is "acceptable" in that it is compatible with (and preferably capable of stabilizing) the active ingredient of the composition and is not deleterious to the subject being treated. One or more solubilizing agents may be used as pharmaceutical excipients for the delivery of the active compound.
The compounds of the invention inhibit PD-1/PD-L1 protein/protein, thereby causing PD-L1 blockade. Blocking PD-L1 can enhance the immune response to cancer cells and infectious diseases in mammals including humans.
The invention further provides application of the compound shown in the formula I, racemate, stereoisomer, tautomer, isotopic marker, nitrogen oxide, solvate, polymorph, metabolite, ester, prodrug or pharmaceutically acceptable salt thereof or the pharmaceutical composition in preparing medicines for preventing and/or treating diseases related to PD-1/PD-L1 signal pathways, wherein the diseases related to PD-1/PD-L1 signal pathways are selected from cancer, infectious diseases and autoimmune diseases.
The invention further provides the use of the compound of formula I, and racemates, stereoisomers, tautomers, isotopic labels, nitrogen oxides, solvates, polymorphs, metabolites, esters, prodrugs or pharmaceutically acceptable salts thereof, or the pharmaceutical composition thereof, for the manufacture of a medicament for the prevention and/or treatment of cancer related disorders. Examples of cancers include diseases in which growth can be inhibited using the compounds of the invention, including cancers that are typically responsive to immunotherapy. Non-limiting examples of preferred cancers for treatment include melanoma (e.g., metastatic malignant melanoma), renal cancer (e.g., clear cell carcinoma), prostate cancer (e.g., hormone refractory prostate cancer), breast cancer, colon cancer, and lung cancer (e.g., non-small cell lung cancer). Examples of other cancers include bone cancer; pancreatic cancer; skin cancer; head and neck cancer; cutaneous or intraocular malignant melanoma; uterine cancer; ovarian cancer; rectal cancer; cancer of the anal region; gastric cancer; testicular cancer; uterine cancer; endometrial cancer of fallopian tube cancer; cervical cancer; vaginal cancer; vulvar cancer; hodgkin's disease; non-hodgkin lymphoma; esophageal cancer; small bowel cancer; cancer of the endocrine system; thyroid cancer; parathyroid cancer; adrenal cancer; soft tissue sarcoma; cancer of the urethra; penile cancer; chronic or acute leukemias, including acute myeloleukemia, chronic myeloleukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia; a solid tumor in children; lymphocytic lymphomas; bladder cancer; renal or urethral cancer; renal pelvis cancer; central Nervous System (CNS) neoplasms; primary CNS lymphoma; tumor angiogenesis; spinal axis tumors; brain stem glioma; pituitary adenoma; kaposi's sarcoma; epidermoid carcinoma; squamous cell carcinoma; t cell lymphoma; environmentally induced cancers, including cancers induced by asbestos; and combinations of said cancers. The invention is also applicable to the treatment of metastatic cancer, particularly metastatic cancer expressing PD-L1.
The present invention also provides a method of treating cancer, which comprises using the compound of formula I of the present invention and racemates, stereoisomers, tautomers, isotopic labels, nitrogen oxides, solvates, polymorphs, metabolites, esters, prodrugs or pharmaceutically acceptable salts thereof or said pharmaceutical composition.
The methods of the invention may comprise administering a compound of the invention alone, as well as in combination with one or more other chemotherapeutic agents. Administration of multiple drugs can be simultaneous or sequential.
Interpretation of terms:
unless otherwise indicated, the definitions of groups and terms described in the specification and claims of the present application, including definitions thereof as examples, exemplary definitions, preferred definitions, definitions described in tables, definitions of specific compounds in the examples, and the like, may be arbitrarily combined and coupled with each other. The definitions of the groups and the structures of the compounds in such combinations and after the combination should fall within the scope of the present specification.
Where a range of numerical values is recited in the specification and claims herein, and where the range of numerical values is defined as an "integer," it is understood that the two endpoints of the range are recited and each integer within the range is recited. For example, "an integer of 0 to 6" should be understood to describe each integer of 0, 1,2, 3,4, 5, and 6. "more" means three or more.
The term "halogen" refers to F, Cl, Br and I. In other words, F, Cl, Br, and I may be described as "halogen" in the present specification.
The optional substitution with a substituent described herein covers the absence of substitution as well as substitution with one or more substituents, e.g., "optionally substituted with one, two or more R" means that it may be unsubstituted (unsubstituted) or substituted with one, two or more R.
The term "aliphatic hydrocarbon group" includes saturated or unsaturated, straight-chain or branched chain or cyclic hydrocarbon groups, the type of the aliphatic hydrocarbon group may be selected from alkyl, alkenyl, alkynyl and the like, the number of carbon atoms of the aliphatic hydrocarbon group is preferably 1 to 12, and may also be 1 to 10, and further preferably ranges from 1 to 6, and specifically may include but is not limited to the following groups: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, ethenyl, 1-propenyl, 2-propenyl, 1-methylethenyl, 1-butenyl, 1-ethylethenyl, 1-methyl-2-propenyl, 2-butenyl, 3-butenyl, 2-methyl-1-propenyl, 2-methyl-2-propenyl, 1-pentenyl, 1-hexenyl, ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 1-methyl-2-propynyl, 3-butynyl, 1-pentynyl, 1-hexynyl, cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl;
the "aliphatic hydrocarbon group" may optionally comprise one, two or more heteroatoms (or be construed as an optional insertion of heteroatoms into the aliphatic hydrocarbon group, optionally a C-C bond and a C-H bond). Suitable heteroatoms will be apparent to those skilled in the art and include, for example, sulfur, nitrogen, oxygen, phosphorus and silicon. The heteroatom containing aliphatic hydrocarbyl group may be selected from the following groups: (C)1-C6) Aliphatic hydrocarbyloxy, (C)1-C6) Aliphatic hydrocarbyl mercapto group, (C)1-C6) Aliphatic hydrocarbyloxy (C)1-C6) Aliphatic hydrocarbon group, (C)1-C6) Aliphatic hydrocarbyl mercapto group (C)1-C6) Aliphatic hydrocarbon group, N- (C)1-C3) Aliphatic hydrocarbyl amino group (C)1-C6) Aliphatic hydrocarbon group, N-di- (C)1-C3) Aliphatic hydrocarbyl amino group (C)1-C6) Aliphatic hydrocarbon groups which may be mentioned are, for example, methoxy, ethoxy, propoxy, butoxy, pentoxy, methoxymethyl, ethoxymethyl, propoxymethyl, methoxyethyl, ethoxyethyl, propoxyethyl, methoxypropyl, ethoxypropyl, propoxypropyl, N-methylaminomethyl, N-methylaminoethyl, N-ethylaminoethyl, N-dimethylaminomethyl, N-dimethylaminoethyl, N-diethylaminoethyl; the "aliphatic hydrocarbon group" moiety contained in the other groups is as explained above.
The term "C3-12Cycloalkyl "is understood to mean a saturated or unsaturated, monovalent monocyclic or bicyclic ring having 3 to 12 carbon atoms, preferably" C3-10Cycloalkyl groups ". The term "C3-10Cycloalkyl "is understood to mean a saturated or unsaturated monovalent monocyclic or bicyclic ring having 3,4, 5,6, 7, 8,9 or 10 carbon atoms. Said C is3-10Cycloalkyl groups may be monocyclic, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl or cyclodecyl, or bicyclic, such as tetralin or decalin.
The term "3-12 membered heterocyclic group" means a saturated or unsaturated monovalent monocyclic or bicyclic ring comprising 1-5 heteroatoms independently selected from N, O and S, the heteroatom-containing group having no aromatic character, said 3-12 membered heterocyclic group, preferably "3-10 membered heterocyclic group". The term "3-10 membered heterocyclyl" means a saturated monovalent monocyclic or bicyclic ring comprising 1-5, preferably 1-3 heteroatoms selected from N, O and S. The heterocyclic group may be attached to the rest of the molecule through any of the carbon atoms or the nitrogen atom (if present). In particular, the heterocyclic group may include, but is not limited to: 4-membered rings such as azetidinyl, oxetanyl; 5-membered rings such as tetrahydrofuranyl, tetrahydrothienyl, dioxolyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, pyrrolinyl; or a 6-membered ring such as tetrahydropyranyl, piperidinyl, morpholinyl, dithianyl, thiomorpholinyl, piperazinyl, or trithianyl; or a 7-membered ring such as diazepanyl. Optionally, the heterocyclic group may be benzo-fused. The heterocyclyl group may be bicyclic, for example but not limited to a 5,5 membered ring, such as a hexahydrocyclopenta [ c ] pyrrol-2 (1H) -yl ring, or a 5,6 membered bicyclic ring, such as a hexahydropyrrolo [1,2-a ] pyrazin-2 (1H) -yl ring. The nitrogen atom containing ring may be partially unsaturated, i.e. it may contain one or more double bonds, such as but not limited to 2, 5-dihydro-1H-pyrrolyl, 4H- [1,3,4] thiadiazinyl, 4, 5-dihydrooxazolyl or 4H- [1,4] thiazinyl, or it may be benzo-fused, such as but not limited to dihydroisoquinolinyl. According to the present invention, the 3-12 membered heterocyclic group may be further selected from the following groups:
Figure BDA0002221622080000111
the term "C6-20Aryl "is to be understood as preferably meaning a mono-, bi-or tricyclic hydrocarbon ring of monovalent or partially aromatic character having 6 to 20 carbon atoms, preferably" C6-14Aryl ". The term "C6-14Aryl "is understood as preferably meaning a monocyclic, bicyclic or tricyclic hydrocarbon ring of monovalent or partial aromaticity having 6, 7, 8,9, 10, 11, 12, 13 or 14 carbon atoms (" C)6-14Aryl "), in particular a ring having 6 carbon atoms (" C6Aryl "), such as phenyl; or biphenyl, or is a ring having 9 carbon atoms ("C9Aryl), such as indanyl or indenyl, or a ring having 10 carbon atoms ("C10Aryl radicals), such as tetralinyl, dihydronaphthyl or naphthyl, or rings having 13 carbon atoms ("C13Aryl radicals), such as the fluorenyl radical, or a ring having 14 carbon atoms ("C)14Aryl), such as anthracenyl.
The term "5-14 membered heteroaryl" is understood to include such monovalent monocyclic, bicyclic or tricyclic aromatic ring systems: which has 5,6, 7, 8,9, 10, 11, 12, 13 or 14 ring atoms, in particular 5 or 6 or 9 or 10 carbon atoms, and which contains 1 to 5, preferably 1 to 3 heteroatoms independently selected from N, O and S and, in addition, can be benzo-fused in each case. In particular, heteroaryl is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl, thia-4H-pyrazolyl and the like and their benzo derivatives, such as benzofuryl, benzothienyl, benzoxazolyl, benzisoxazolyl, benzimidazolyl, benzotriazolyl, indazolyl, indolyl, isoindolyl and the like; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like, and benzo derivatives thereof, such as quinolyl, quinazolinyl, isoquinolyl, and the like; or azocinyl, indolizinyl, purinyl and the like and benzo derivatives thereof; or cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, naphthyridinyl, pteridinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and the like.
Unless otherwise indicated, heterocyclyl or heteroaryl includes all possible isomeric forms thereof, e.g. positional isomers thereof. Thus, for some illustrative, non-limiting examples, pyridyl or pyridinylene includes pyridin-2-yl, pyridinylene-2-yl, pyridin-3-yl, pyridinylene-3-yl, pyridin-4-yl, and pyridinylene-4-yl; thienyl or thienylene includes thien-2-yl, thien-3-yl and thien-3-yl.
Unless otherwise indicated, "C" as contained in other groups of the invention (e.g., N-containing 3-12 membered heterocycloalkyl or N-containing 5-14 membered heteroaryl)3-12Cycloalkyl group "," 3-12 membered heterocyclic group "," C6-20Aryl "," 5-to 14-membered heteroaryl "are equivalent to the above explanations.
Depending on their molecular structure, the compounds of the invention may be chiral and may therefore exist in various enantiomeric forms. These compounds may thus be present in racemic or optically active form. The compounds of the invention or intermediates thereof may be separated into enantiomeric compounds by chemical or physical methods well known to those skilled in the art, or used in this form for synthesis. In the case of racemic amines, diastereomers are prepared from mixtures by reaction with optically active resolving agents. Examples of suitable resolving agents are optically active acids such as tartaric acid in the R and S forms, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid, suitable N-protected amino acids (e.g. N-benzoylproline or N-benzenesulfonylproline) or various optically active camphorsulfonic acids. Chromatographic enantiomeric resolution can also advantageously be carried out with the aid of optically active resolving agents, such as dinitrobenzoylphenylglycine, cellulose triacetate or other carbohydrate derivatives or chirally derivatized methacrylate polymers, which are immobilized on silica gel. Suitable eluents for this purpose are aqueous or alcoholic solvent mixtures, for example hexane/isopropanol/acetonitrile.
Those skilled in the art will appreciate that not all nitrogen-containing heterocycles can form N-oxides, as the nitrogen needs to have available lone pairs of electrons for oxidation to an oxynitride; those skilled in the art will recognize nitrogen-containing heterocycles that are capable of forming N-oxides. Those skilled in the art will also recognize that tertiary amines are capable of forming N-oxides. Synthetic methods for preparing N-oxides of heterocycles and tertiary amines are well known to those skilled in the art and include the oxidation of heterocycles and tertiary amines with peroxy acids such as peracetic and m-chloroperbenzoic acid (MCPBA), hydrogen peroxide, alkyl hydroperoxides such as t-butyl hydroperoxide, sodium perborate, and dioxiranes (dioxiranes) such as dimethyldioxirane. These methods for preparing N-oxides have been widely described and reviewed in the literature.
Pharmaceutically acceptable salts may be acid addition salts of the compounds of the invention having sufficient basicity, for example having a nitrogen atom in the chain or ring, for example with the following inorganic acids: for example hydrochloric acid, hydrofluoric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, pyrosulfuric acid, phosphoric acid or nitric acid, or hydrogen sulfates, or acid addition salts with organic acids such as: such as formic acid, acetic acid, acetoacetic acid, pyruvic acid, trifluoroacetic acid, propionic acid, butyric acid, caproic acid, heptanoic acid, undecanoic acid, lauric acid, benzoic acid, salicylic acid, 2- (4-hydroxybenzoyl) benzoic acid, camphoric acid, cinnamic acid, cyclopentanepropionic acid, digluconic acid, 3-hydroxy-2-naphthoic acid, nicotinic acid, pamoic acid, pectinic acid, persulfuric acid, 3-phenylpropionic acid, picric acid, pivalic acid, 2-hydroxyethanesulfonic acid, itaconic acid, sulfamic acid, trifluoromethanesulfonic acid, dodecylsulfuric acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, 2-naphthalenesulfonic acid, naphthalenedisulfonic acid, camphorsulfonic acid, citric acid, tartaric acid, stearic acid, lactic acid, oxalic acid, malonic acid, succinic acid, malic acid, adipic acid, alginic acid, maleic acid, fumaric acid, D-gluconic acid, succinic acid, malic acid, adipic acid, alginic acid, maleic acid, fumaric acid, D-gluconic acid, and mixtures thereof, Mandelic acid, ascorbic acid, glucoheptylic acid, glycerophosphoric acid, aspartic acid, sulfosalicylic acid, hemisulfuric acid or thiocyanic acid.
In addition, another suitable pharmaceutically acceptable salt of a compound of the invention which is sufficiently acidic is an alkali metal salt (e.g., sodium or potassium salt), an alkaline earth metal salt (e.g., calcium or magnesium salt), an ammonium salt, or a salt with an organic base which affords a physiologically acceptable cation, such as a salt with: sodium ions, potassium ions, N-methylglucamine, dimethylglucamine, ethylglucamine, lysine, dicyclohexylamine, 1, 6-hexanediamine, ethanolamine, glucosamine, meglumine, sarcosine, serinol, trihydroxymethylaminomethane, aminopropanediol, 1-amino-2, 3, 4-butanetriol. By way of example, the pharmaceutically acceptable salts include salts of the group-COOH with: sodium ion, potassium ion, calcium ion, magnesium ion, N-methylglucamine, dimethylglucamine, ethylglucamine, lysine, dicyclohexylamine, 1, 6-hexanediamine, ethanolamine, glucosamine, meglumine, sarcosine, serinol, trihydroxymethylaminomethane, aminopropanediol, 1-amino-2, 3, 4-butanetriol.
In addition, the basic nitrogen-containing groups may be quaternized with the following agents: lower alkyl halides such as methyl, ethyl, propyl and butyl chlorides, bromides and iodides; dialkyl sulfates such as dimethyl sulfate, diethyl sulfate, dibutyl sulfate, and diamyl sulfate; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; aralkyl halides such as benzyl and phenethyl bromide, and the like. By way of example, pharmaceutically acceptable salts include hydrochloride, sulfate, nitrate, bisulfate, hydrobromide, acetate, oxalate, citrate, methanesulfonate, formate, or meglumine salts and the like.
Since the compound of the present invention may exist at a plurality of salt-forming sites, the "pharmaceutically acceptable salt" includes not only the salt formed at 1 salt-forming site among the compounds of the present invention but also the salt formed at 2,3 or all of the salt-forming sites among them. For this purpose, the molar ratio of the "pharmaceutically acceptable salt" of the compound of formula (I) to the cation of the acid (anion) or base required for salt formation may vary within wide limits, and may be, for example, 4:1 to 1:4, such as 3:1, 2:1, 1:2, 1:3, etc.
According to the present invention, the pharmaceutically acceptable anion includes anions selected from the group consisting of those generated by ionization of inorganic or organic acids. The "inorganic acid" includes, but is not limited to, hydrochloric acid, hydrofluoric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, pyrosulfuric acid, phosphoric acid, or nitric acid. The "organic acid" includes, but is not limited to, formic acid, acetic acid, acetoacetic acid, pyruvic acid, trifluoroacetic acid, propionic acid, butyric acid, caproic acid, heptanoic acid, undecanoic acid, lauric acid, benzoic acid, salicylic acid, 2- (4-hydroxybenzoyl) benzoic acid, camphoric acid, cinnamic acid, cyclopentanepropionic acid, digluconic acid, 3-hydroxy-2-naphthoic acid, nicotinic acid, pamoic acid, pectinic acid, persulfuric acid, 3-phenylpropionic acid, picric acid, pivalic acid, 2-hydroxyethanesulfonic acid, itaconic acid, sulfamic acid, trifluoromethanesulfonic acid, dodecylsulfuric acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, methanesulfonic acid, 2-naphthalenesulfonic acid, naphthalenedisulfonic acid, camphorsulfonic acid, citric acid, tartaric acid, stearic acid, lactic acid, oxalic acid, malonic acid, succinic acid, malic acid, adipic acid, alginic acid, maleic acid, fumaric acid, maleic acid, and mixtures thereof, Fumaric acid, D-gluconic acid, mandelic acid, ascorbic acid, glucoheptonic acid, glycerophosphoric acid, aspartic acid, sulfosalicylic acid, hemisulfuric acid or thiocyanic acid.
Depending on the position and nature of the various substituents, the compounds of the present invention may also contain one or more asymmetric centers. Asymmetric carbon atoms may exist in either the (R) or (S) configuration, with only one asymmetric center yielding a racemic mixture and multiple asymmetric centers yielding a diastereomeric mixture. In some cases, asymmetry may also exist due to hindered rotation about a particular bond, for example, the central bond connects two substituted aromatic rings of a particular compound. Also, the substituents may exist in cis or trans isomeric forms.
The compounds of the invention also include all possible stereoisomers of each, either as a single stereoisomer or as any mixture of said stereoisomers (e.g. the R-or S-isomers, or the E-or Z-isomers) in any proportion. Separation of single stereoisomers (e.g. single enantiomers or single diastereomers) of the compounds of the invention may be achieved by any suitable prior art method (e.g. chromatography, particularly for example chiral chromatography).
The term "tautomer" refers to an isomer of a functional group resulting from the rapid movement of an atom in two positions in a molecule. The compounds of the invention may exhibit tautomerism. Tautomeric compounds may exist in two or more interconvertible species. Prototropic tautomers result from the migration of a covalently bonded hydrogen atom between two atoms. Tautomers generally exist in equilibrium, and attempts to isolate a single tautomer usually result in a mixture whose physicochemical properties are consistent with the mixture of the compounds. The position of equilibrium depends on the chemical properties within the molecule. For example, in many aliphatic aldehydes and ketones such as acetaldehyde, the keto form predominates; whereas in phenol the enol type predominates. The present invention encompasses all tautomeric forms of the compounds.
In the present invention, reference to compounds also includes isotopically-labeled compounds, which are identical to those shown in formula I, but wherein one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of H, C, N, O, S, F and Cl, such as2H、3H、13C、11C、14C、15N、18O、17O、32P、35S、18F and36and (4) Cl. Containing isotopes of the above and/or other isotopes of other atomsThe compounds of the invention, prodrugs thereof, or pharmaceutically acceptable salts of said compounds or of said prodrugs are within the scope of the invention. Certain isotopically-labeled compounds of the present invention, for example, incorporate a radioactive isotope (such as3H and14C) the compounds of (a) are useful in drug and/or substrate tissue distribution assays. Tritium (i.e. tritium3H) And carbon 14 (i.e.14C) Isotopes are particularly preferred for their ease of preparation and detectability. Again, with heavier isotopes such as deuterium (i.e. deuterium)2H) Substitutions may provide certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and may therefore be preferred in certain circumstances. The compounds of the invention as claimed may be particularly limited to replacement by deuterium or tritium. Furthermore, the absence of hydrogen in the substituents indicating the term deuterium or tritium alone is not meant to exclude deuterium or tritium, but may equally well comprise deuterium or tritium.
The term "effective amount" or "therapeutically effective amount" refers to an amount of a compound of the present invention sufficient to effect the intended use, including but not limited to the treatment of a disease as defined below. The therapeutically effective amount may vary depending on the following factors: the intended application (in vitro or in vivo), or the subject and disease condition being treated, such as the weight and age of the subject, the severity of the disease condition and the mode of administration, etc., can be readily determined by one of ordinary skill in the art. The specific dosage will vary depending on the following factors: the particular compound selected, the dosage regimen to be followed, whether to administer in combination with other compounds, the timing of administration, the tissue to be administered and the physical delivery system carried.
The term "solvate" is those forms of the compounds of the present invention which form complexes in the solid or liquid state by coordination with solvent molecules. Hydrates are a particular form of solvates in which the coordination is with water. In the present invention, the preferred solvate is a hydrate. Further, pharmaceutically acceptable solvates (hydrates) of the compounds of general formula I according to the invention refer to co-crystals and clathrates of compound I with one or more molecules of water or other solvents in stoichiometric amounts. Solvents that may be used for the solvate include, but are not limited to: water, methanol, ethanol, ethylene glycol and acetic acid.
The term "prodrug", or "prodrug" refers to a compound that is converted in vivo to a compound of the general formula or a particular compound. Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrugs of the present invention may be esters, and in the present invention esters may be used as prodrugs, which are esters of benzene, aliphatic (C1-24) esters, acyloxymethyl esters, carbonates, carbamates and amino acids. For example, a compound of the present invention comprises a hydroxy/carboxy group, i.e., it may be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent.
Advantageous effects
1) The invention provides a novel compound with a general formula I, which has obvious inhibition activity of PD-1/PD-L1 protein-protein interaction. The PD-1/PD-L1 inhibitory activity of some compounds is obviously better than that of a positive control drug;
2) the compound has obvious inhibition effect on 4 tumor cells, the tumor cell inhibition activity of some compounds is obviously superior to that of a positive control medicament, and especially, the inhibition activity on MC-38 and SUM149 is far greater than that of the positive control medicament;
3) the compound of the invention has appropriate half-life and excellent bioavailability, is suitable for patent medicine and has wide medicinal prospect.
Detailed Description
The technical solution of the present invention will be further described in detail with reference to specific embodiments. It is to be understood that the following examples are only illustrative and explanatory of the present invention and should not be construed as limiting the scope of the present invention. All the technologies realized based on the above-mentioned contents of the present invention are covered in the protection scope of the present invention.
Unless otherwise specified, the raw materials and reagents used in the following examples are all commercially available products or can be prepared by known methods.
EXAMPLE 1 Synthesis of Compound I-1
Figure BDA0002221622080000151
Synthesis of Compound I-1-1: adding 6.01g of 6-bromo-1, 4-benzooxane, 11.90g of NBS, 0.15g of AIBN and 80ml of carbon tetrachloride into a 250ml single-neck bottle, replacing nitrogen for 3 times and protecting the nitrogen, reacting for 18.0h at 80 ℃, stopping the reaction, cooling to room temperature, carrying out suction filtration, leaching a filter cake by using 30ml of carbon tetrachloride and draining, adding 100ml of water into a filtrate, stirring, carrying out liquid separation to retain an organic phase, drying by using anhydrous sodium sulfate, concentrating under reduced pressure to dryness to obtain a compound I-1-1, and directly putting the compound I-1-1 into the next step.
Synthesis of Compound I-1-2: adding 100ml of acetone into the compound I-1-1 to dissolve, adding 20.90g of sodium iodide in batches, replacing nitrogen for 3 times, carrying out nitrogen protection, reacting for 2.0h at 60 ℃, basically completely reacting the raw materials, stopping the reaction, carrying out reduced pressure concentration to remove the solvent, adding 150ml of saturated sodium sulfite aqueous solution, extracting with 50ml of ethyl acetate 2, combining organic phases, drying by anhydrous sodium sulfate, carrying out reduced pressure concentration to dryness, and carrying out column chromatography purification to obtain 3.02g of compound I-1-2, namely a light yellow oily liquid.
Synthesis of Compounds I-1-3: a100 ml three-necked flask was charged with 2.04g of 2-methyl-3- (hydroxymethyl) phenylboronic acid pinacol ester, 1.62g of 5-chloro-2, 4-dihydroxybenzaldehyde, 3.12g of triphenylphosphine and 30ml of tetrahydrofuran, and the mixture was purged with nitrogen for 3 times and then purged with nitrogen, and then cooled to-10 ℃. Diluting 2.02g DIEA with 3ml tetrahydrofuran, slowly adding dropwise to the reaction solution, controlling the temperature below 0 ℃, moving to room temperature to react for 12.0h after dropwise adding, stopping the reaction, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 1.74g of off-white solid, namely the compound I-1-3.
Synthesis of Compounds I-1-4: adding 1.44g of compound I-1-3, 0.70g of 3-cyanobenzyl bromide, 0.98g of potassium carbonate and 15ml of DMF into a 100ml single-neck bottle, reacting at 50 ℃ for 2.0h, stopping the reaction, cooling to room temperature, slowly adding the reaction solution into 100ml of water, precipitating a large amount of solid, carrying out suction filtration, leaching a filter cake with water, drying by pumping, and drying by a forced air oven to obtain 1.50g of compound I-1-4.
Compounds I-1-5Synthesizing: a250 ml single-necked flask was charged with 0.90g of Compound I-1-4, 1.05g of Compound I-1-2, 1.16g of potassium carbonate, 0.16g of Pd (dppf) Cl2And (3) replacing nitrogen by 30ml of DMF and 5ml of water, carrying out nitrogen protection, reacting at 90 ℃ for 2.0h, stopping the reaction, cooling to room temperature, pouring the reaction solution into 200ml of water, extracting by using 60ml of 2 ethyl acetate, combining organic phases, drying by using anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 0.80g of light yellow oily liquid, namely the compound I-1-5.
Synthesis of Compound I-1: adding 250mg of compound I-1-5, 90mg of high proline, 60mg of sodium cyanoborohydride, 6ml of DMF and glacial acetic acid 1d into a 100ml single-neck bottle, reacting at 50 ℃ for 12.0h, stopping the reaction, cooling to room temperature, pouring the reaction liquid into 50ml of water, extracting with ethyl acetate (25ml of 2), combining organic phases, drying with anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 12mg of compound I-1. MS M/z 638.1(M +1),1H NMR(400MHz,d6-DMSO)δ:9.85(s,1H),8.79-8.78(m,1H),8.14-8.13(m,1H),7.85-7.83(m,1H),7.56-7.55(m,1H),7.43-7.41(m,1H),7.26-7.22(m,2H),7.20-7.11(m,1H),6.80-6.78(m,2H),6.65-6.62(m,1H),6.26-6.24(m,1H),5.48-5.38(m,2H),5.28-5.26(m,2H),4.39-4.03(m,4H),3.09-3.06(m,1H),2.23(s,3H),2.11-2.09(m,2H),1.88-1.82(m,2H),1.70-1.56(m,4H)ppm。
EXAMPLE 2 Synthesis of Compound I-2
Figure BDA0002221622080000161
Synthesis of Compound I-2-1: a250 ml single-neck flask was charged with 1.80g of Compound I-1-2, 2.51g of 2-methyl-3- (hydroxymethyl) phenylboronic acid pinacol ester, 2.33g of potassium carbonate, 0.31g of Pd (dppf) Cl2Replacing nitrogen with 30ml of DMF and 5ml of water for 3 times under the protection of nitrogen, reacting at 90 ℃ for 2.0 hours, stopping the reaction, cooling to room temperature, pouring the reaction liquid into 200ml of water, extracting with 60ml of ethyl 2 acetate, combining organic phases, drying with anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 1.60g of light yellow oily liquid, namely the compound I-2-1.
Synthesis of Compound I-2-2: a100 ml three-necked flask was charged with 1.02g of Compound I-2-1, 0.81g of 5-chloro-2, 4-dihydroxybenzaldehyde, 1.56g of triphenylphosphine and 15ml of tetrahydrofuran, and then the mixture was purged with nitrogen for 3 times under nitrogen protection, and then cooled to-10 ℃. Diluting 1.01g of DIEA with 3ml of tetrahydrofuran, slowly dropwise adding the diluted DIEA into the reaction solution, controlling the temperature below 0 ℃, moving the reaction solution to room temperature to react for 12.0h after the dropwise adding is finished, stopping the reaction, concentrating the dry solvent under reduced pressure, adding 15ml of ethanol, crystallizing at 0 ℃, carrying out suction filtration, leaching the filter cake with glacial ethanol, and carrying out suction filtration to obtain 0.87g of off-white solid, namely the compound I-2-2.
Synthesis of Compounds I-1-5: and adding 0.80g of compound I-2-2, 0.57g of 3-cyanobenzyl bromide, 0.54g of potassium carbonate and 15ml of DMF into a 100ml single-neck bottle, reacting at 50 ℃ for 2.0h, stopping the reaction, cooling to room temperature, slowly adding the reaction solution into 100ml of water, precipitating a large amount of solid, carrying out suction filtration, leaching a filter cake with water, drying by pumping, and drying by a forced air oven to obtain 0.82g of compound I-1-5.
Synthesis of Compound I-2: adding 250mg of compound I-1-5140 mg of cis-4-hydroxy-L-proline, 60mg of sodium cyanoborohydride, 6ml of DMF and 1d of glacial acetic acid into a 100ml single-neck bottle, reacting at 50 ℃ for 12.0h, stopping the reaction, cooling to room temperature, pouring the reaction solution into 50ml of water, extracting with 25ml of ethyl acetate 2, combining organic phases, drying with anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 15mg of compound I-2. MS M/z 640.1(M +1),1H NMR(400MHz,d6-DMSO)δ:9.86(s,1H),8.78-8.77(m,1H),8.17-8.16(m,1H),7.88-7.85(m,1H),7.53-7.51(m,1H),7.44-7.42(m,1H),7.28-7.25(m,2H),7.20-7.16(m,1H),6.87-6.79(m,2H),6.68-6.65(m,1H),6.24-6.22(m,1H),5.50-5.39(m,2H),5.29-5.20(m,4H),4.08-4.05(m,2H),3.69(s,1H),3.52-3.41(m,1H),3.22-3.18(m,1H),2.26(s,3H),2.10-2.05(m,2H),1.83-1.76(m,2H)ppm。
EXAMPLE 3 Synthesis of Compound I-3
Figure BDA0002221622080000171
Synthesis method of Compound I-3 referring to the preparation of the Synthesis method of Compound I-2, cis-4-hydroxy-L-proline is replaced by L-proline to obtain 13mg Compound I-3. MS M/z 624.2(M +1),1H NMR(400MHz,d6-DMSO)δ:10.05(s,1H),8.77-8.75(m,1H),8.18-8.15(m,1H),7.82-7.80(m,1H),7.55-7.53(m,1H),7.45-7.43(m,1H),7.29-7.26(m,2H),7.22-7.18(m,1H),6.88-6.80(m,2H),6.69-6.62(m,1H),6.25-6.23(m,1H),5.55-5.42(m,2H),5.19-5.04(m,4H),4.09-4.02(m,2H),3.42-3.31(m,1H),2.23(s,3H),2.11-2.02(m,2H),1.95-1.82(m,2H),1.44-1.32(m,2H)ppm。
EXAMPLE 4 Synthesis of Compound I-4
Figure BDA0002221622080000172
Synthesis of Compound I-4 the compound I-4 was prepared by reference to the synthesis of Compound I-2, substituting cis-4-hydroxy-L-proline for serinol to give 15mg of Compound I-4. MS M/z 600.2(M +1),1H NMR(400MHz,d6-DMSO)δ:8.79-8.76(m,1H),8.18-8.15(m,1H),7.83-7.80(m,1H),7.55-7.52(m,1H),7.43-7.41(m,1H),7.27-7.22(m,2H),7.20-7.17(m,1H),6.88-6.77(m,2H),6.65-6.63(m,1H),6.26-6.22(m,1H),5.54-5.40(m,2H),5.26-5.23(m,4H),3.99-3.86(m,2H),3.69-3.62(m,2H),3.58-3.42(m,4H),3.26-3.17(m,1H),2.26(s,3H),2.05(s,1H)ppm。
EXAMPLE 5 Synthesis of Compound I-5
Figure BDA0002221622080000181
Synthesis method of Compound I-5 the compound I-5 was prepared by reference to the synthesis method of Compound I-2, substituting cis-4-hydroxy-L-proline for ethanolamine to obtain 15mg of Compound I-5. MS M/z 570.2(M +1),1H NMR(400MHz,d6-DMSO)δ:8.77-8.74(m,1H),8.19-8.17(m,1H),7.85-7.82(m,1H),7.56-7.53(m,1H),7.41-7.40(m,1H),7.26-7.24(m,2H),7.21-7.18(m,1H),6.89-6.78(m,2H),6.66-6.62(m,1H),6.28-6.26(m,1H),5.64-5.58(m,2H),5.28-5.22(m,4H),3.82-3.79(m,2H),3.69(s,1H),3.55-3.47(m,2H),2.86-2.77(m,2H),2.26(s,3H),2.02(s,1H)ppm
EXAMPLE 6 Synthesis of Compound I-6
Figure BDA0002221622080000182
Synthesis of Compound I-6-1: a100 ml three-necked flask was charged with 1.02g of Compound I-2-1, 0.73g of 4-hydroxy-2, 6-dimethoxybenzaldehyde, 1.61g of triphenylphosphine and 15ml of tetrahydrofuran, and then the mixture was purged with nitrogen for 3 times under nitrogen protection, and the temperature was lowered to-10 ℃. Diluting 1.03g of DIEA with 3ml of tetrahydrofuran, slowly dropwise adding the diluted solution to the reaction solution, controlling the temperature below 0 ℃, moving the solution to room temperature for reaction for 12.0h after dropwise adding, stopping the reaction, concentrating the solution under reduced pressure, drying the solution, and purifying the solution by column chromatography to obtain 0.97g of a white-like solid, namely the compound I-6-1.
Synthesis of Compound I-6: adding 250mg of compound I-6-1, 115mg of high proline, 75mg of sodium cyanoborohydride, 6ml of DMF and glacial acetic acid 1d into a 100ml single-neck bottle, reacting at 50 ℃ for 12.0h, stopping the reaction, cooling to room temperature, pouring the reaction liquid into 50ml of water, extracting with ethyl acetate (25ml of 2), combining organic phases, drying with anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 12mg of compound I-6. MS M/z 532.6(M +1),1H NMR(400MHz,d6-DMSO)δ:10.12(s,1H),7.86-7.83(m,1H),7.46-7.43(m,1H),7.26-7.20(m,3H),6.89-6.85(m,1H),6.28-6.25(m,2H),5.67-5.55(m,2H),5.18-5.12(m,2H),3.86(s,6H),3.69-3.62(m,2H),3.22-3.15(m,1H),2.55-2.48(m,2H),2.26(s,3H),2.02-1.98(m,2H),1.55-1.32(m,4H)ppm。
EXAMPLE 7 Synthesis of Compound I-7
Figure BDA0002221622080000191
Synthesis methods of Compounds I-7-1 and I-7 referring to the synthesis method preparation of Compound I-2, 3-cyanobenzyl bromide was replaced with 2-cyano-4-bromomethylpyridine, and cis-4-hydroxy-L-proline was replaced with L-proline to give 12mg of Compound I-7. MS M/z 625.1(M +1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),8.78-8.76(m,1H),8.38-8.35(m,1H),8.12-8.07(m,1H),7.49-7.45(m,1H),7.28-7.25(m,2H),7.21-7.16(m,1H),6.87-6.82(m,2H),6.68-6.64(m,1H),6.28-6.26(m,1H),5.56-5.48(m,2H),5.18-5.07(m,4H),4.08-4.04(m,2H),3.48-3.37(m,1H),2.26(s,3H),2.18-2.07(m,2H),1.93-1.88(m,2H),1.46-1.35(m,2H)ppm。
EXAMPLE 8 Synthesis of Compound I-8
Figure BDA0002221622080000192
Synthesis of Compound I-8: adding 300mg of compound I-7 and 10ml of tetrahydrofuran into a 100ml single-mouth bottle, adding 1ml of 50% hydroxylamine aqueous solution, reacting at room temperature for 3.0h, stopping the reaction, removing the solvent by decompression and rotary separation, and purifying by column chromatography to obtain 12mg of compound I-8. MS M/z 658.2(M +1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),8.79-8.77(m,1H),8.12-8.07(m,1H),7.99-7.95(m,1H),7.48-7.46(m,1H),7.26-7.23(m,2H),7.20-7.14(m,1H),7.08(s,2H),6.88-6.83(m,2H),6.69-6.66(m,1H),6.29-6.27(m,1H),5.57-5.48(m,2H),5.19-5.08(m,4H),4.06-4.03(m,2H),3.49-3.35(m,1H),2.26(s,3H),2.19-2.08(m,2H),2.03(s,1H),1.96-1.88(m,2H),1.47-1.38(m,2H)ppm。
EXAMPLE 9 Synthesis of Compound I-9
Figure BDA0002221622080000193
Synthesis of Compound I-9 reference was made to the synthesis of Compound I-8 to obtain 16mg of Compound I-9. MS M/z 657.1(M +1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),8.76-8.74(m,1H),8.16-8.14(m,1H),7.86-7.84(m,1H),7.56-7.53(m,1H),7.48-7.46(m,1H),7.28-7.25(m,2H),7.24-7.19(m,1H),7.06(s,2H),6.89-6.85(m,2H),6.68-6.66(m,1H),6.27-6.26(m,1H),5.57-5.48(m,2H),5.17-5.08(m,4H),4.04-4.01(m,2H),3.46-3.33(m,1H),2.27(s,3H),2.14-2.06(m,2H),2.05(s,1H),1.97-1.84(m,2H),1.46-1.35(m,2H)ppm。
EXAMPLE 10 Synthesis of Compound I-10
Figure BDA0002221622080000201
Synthesis of Compound I-10 reference was made to the synthesis of Compound I-2, which was prepared by substituting cis-4-hydroxy-L-proline for L-serine to give 15mg of Compound I-10. MS M/z 614.1(M +1),1H NMR(400MHz,d6-DMSO)δ:10.08(s,1H),8.76-8.71(m,1H),8.14-8.12(m,1H),7.84-7.80(m,1H),7.56-7.52(m,1H),7.44-7.41(m,1H),7.28-7.24(m,2H),7.22-7.18(m,1H),6.87-6.78(m,2H),6.66-6.63(m,1H),6.25-6.21(m,1H),5.51-5.43(m,2H),5.22-5.20(m,4H),3.95-3.84(m,2H),3.64-3.61(m,1H),3.56-3.44(m,2H),3.24-3.18(m,1H),2.26(s,3H),2.09(s,1H)ppm。
EXAMPLE 11 Synthesis of Compound I-11
Figure BDA0002221622080000202
Synthesis methods of Compounds I-11-1 and I-11 reference was made to the synthesis method of Compound I-2 by substituting 5-chloro-2, 4-dihydroxybenzaldehyde for 3-chloro-4-hydroxybenzaldehyde and cis-4-hydroxy-L-proline for L-serine to give 12mg of Compound I-11. MS M/z 482.9(M +1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),7.69-7.65(m,1H),7.46-7.43(m,2H),7.34-7.31(m,1H),7.09-7.07(m,1H),7.01-6.82(m,4H),6.34-6.32(m,1H),5.81-5.78(m,2H),5.18-5.16(m,2H),4.92-4.90(m,1H),4.02-3.73(m,2H),3.80-3.76(m,2H),3.51-3.46(m,1H),2.68(s,3H)ppm。
EXAMPLE 12 Synthesis of Compound I-12
Figure BDA0002221622080000211
Synthesis methods of Compounds I-12-1 and I-12 reference was made to the synthesis of Compound I-2 by substituting 5-chloro-2, 4-dihydroxybenzaldehyde for 3-fluoro-4-hydroxybenzaldehyde and cis-4-hydroxy-L-proline for L-serine to give 15mg of Compound I-12. MS M/z 466.5(M +1),1H NMR(400MHz,d6-DMSO)δ:10.08(s,1H),7.70-7.68(m,1H),7.49-7.47(m,1H),7.36-7.32(m,1H),7.11-7.08(m,1H),6.95-6.83(m,5H),6.37-6.32(m,1H),5.80-5.78(m,2H),5.14-5.10(m,2H),4.94-4.93(m,1H),4.05-3.77(m,2H),3.81-3.78(m,2H),3.52-3.48(m,1H),2.69(s,3H)ppm。
EXAMPLE 13 Synthesis of Compound I-13
Figure BDA0002221622080000212
Synthesis methods of Compounds I-13-1 and I-13 preparation of reference Compound I-2 was carried out by substituting 5-chloro-2, 4-dihydroxybenzaldehyde for 3, 5-difluoro-4-hydroxybenzaldehyde and cis-4-hydroxy-L-proline for L-proline to give 15mg of Compound I-13. MS M/z 494.5(M +1),1H NMR(400MHz,d6-DMSO)δ:10.05(s,1H),7.69-7.68(m,1H),7.48-7.47(m,1H),7.34-7.32(m,1H),7.11-7.08(m,1H),6.97-6.96(m,1H),6.83-6.77(m,3H),5.83-5.77(m,2H),5.16-5.11(m,2H),3.62-3.61(m,2H),3.22-3.21(m,1H),2.69(s,3H),2.42-2.38(m,2H),1.95-1.73(m,2H),1.66-1.54(m,2H)ppm。
EXAMPLE 14 Synthesis of Compound I-14
Figure BDA0002221622080000221
Synthesis methods of Compounds I-14-1 and I-14 reference was made to the synthesis of Compound I-2 by substituting 5-chloro-2, 4-dihydroxybenzaldehyde for 2-methoxy-4-hydroxybenzaldehyde to give 15mg of Compound I-14. MS M/z 504.5(M +1),1H NMR(400MHz,d6-DMSO)δ:10.05(s,1H),7.85-7.82(m,1H),7.57-7.52(m,1H),7.38-7.26(m,2H),7.12-7.09(m,1H),6.99-6.96(m,1H),6.85-6.83(m,1H),6.66-6.56(m,2H),5.84-5.79(m,2H),5.35-5.33(m,1H),5.16-5.11(m,2H),3.83(s,3H),3.74-3.62(m,3H),3.22-3.16(m,1H),2.69(s,3H),2.14-1.82(m,4H)ppm。
EXAMPLE 15 Synthesis of Compound I-15
Figure BDA0002221622080000222
Synthesis method of Compound I-15-1 referring to the preparation of Compound I-6, L-homoproline was replaced with (2S,4S) -4-amino-1- (tert-butoxycarbonyl) pyrrolidine dicarboxylic acid to give 108mg of Compound I-15-1, which was directly administered to the next step.
Synthesis of Compound I-15: adding the I-15-1 and 10ml dichloromethane into a 50ml reaction bottle, adding 2ml trifluoroacetic acid, reacting at room temperature for 2.0h, performing reduced pressure spin drying, and purifying a prepared liquid phase to obtain 12mg of a compound I-15. MS M/z 533.6(M +1),1H NMR(400MHz,d6-DMSO)δ:10.09(s,1H),9.98-9.97(m,1H),7.70-7.67(m,1H),7.48-7.45(m,1H),7.35-7.27(m,1H),7.12-7.08(m,1H),6.96-6.95(m,1H),6.85-6.83(m,1H),6.29-6.26(m,2H),5.83-5.79(m,2H),5.15-5.13(m,2H),3.93-3.92(m,2H),3.78(s,6H),3.05-2.78(m,4H),2.68(s,3H),2.11-1.85(m,3H)ppm。
EXAMPLE 16 Synthesis of Compound I-16
Figure BDA0002221622080000231
Synthesis of Compound I-16 the compound I-16 was prepared by reference to the Synthesis of Compound I-6, substituting L-homoproline for carboxymethylhydroxylamine hemihydrochloride to give 13mg of Compound I-16. MS M/z 494.5(M +1),1H NMR(400MHz,d6-DMSO)δ:9.98(s,1H),9.96-9.95(m,1H),7.75-7.72(m,1H),7.67-7.66(m,1H),7.38-7.26(m,1H),7.16-7.09(m,1H),6.93-6.90(m,1H),6.83-6.81(m,1H),6.26-6.24(m,2H),5.83-5.79(m,2H),5.18-5.16(m,2H),4.86-4.85(m,2H),3.96-3.88(m,2H),3.78(s,6H),2.67(s,3H)ppm。
EXAMPLE 17 Synthesis of Compound I-17
Figure BDA0002221622080000232
Synthesis method of compound I-17Prepared according to the synthetic method of the compound I-1 by replacing L-homoproline with methyl 2- (aminooxy) acetate to obtain 15mg of the compound I-17. MS M/z 614.1(M +1),1H NMR(400MHz,d6-DMSO)δ:9.96(s,1H),7.99-7.88(m,1H),7.84-7.68(m,3H),7.49-7.46(m,2H),7.35-7.31(m,1H),7.18-7.09(m,2H),6.99-6.96(m,1H),6.88-6.86(m,1H),6.57-6.52(m,1H),5.83-5.78(m,2H),5.15-5.13(m,4H),4.85-4.81(m,2H),3.93-3.90(m,2H),3.86(s,3H),2.69(s,3H)ppm
EXAMPLE 18 Synthesis of Compound I-18
Figure BDA0002221622080000233
Synthesis method of Compound I-18-1 referring to the preparation of the Synthesis method of Compound I-2, cis-4-hydroxy-L-proline is replaced by N' -tert-butoxycarbonyl-L-2, 3-diaminopropionic acid methyl ester hydrochloride to obtain 150mg of Compound I-18-1.
Synthesis of Compound I-18-2: taking a 10ml single-mouth bottle, adding 150mg of compound I-18-1 and 5ml of dichloromethane, dissolving, dropwise adding 1ml of trifluoroacetic acid, reacting at room temperature for 3.0h, stopping reaction, removing the solvent by rotary removal under reduced pressure to obtain a white-like solid, and directly putting the white-like solid into the next step.
Synthesis of Compound I-18: and adding 10ml of tetrahydrofuran, 100mg of triethylamine and 100mg of carbonyl diimidazole into the solid, reacting at room temperature for 2.0 hours, stopping the reaction, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 10mg of a compound I-18. MS M/z 639.1(M +1),1H NMR(400MHz,d6-DMSO)δ:10.02(s,1H),8.05-7.98(m,1H),7.90-7.88(m,1H),7.79-7.69(m,3H),7.47-7.46(m,2H),7.33-7.32(m,1H),7.16-7.09(m,2H),6.97-6.95(m,1H),6.84-6.82(m,1H),6.53-6.50(m,1H),5.82-5.79(m,2H),5.17-5.08(m,5H),4.43-4.41(m,2H),3.78-3.51(m,2H),2.68(s,3H)ppm。
EXAMPLE 19 Synthesis of Compound I-19
Figure BDA0002221622080000241
Synthesis of Compound I-19-1: taking a 100ml reaction bottle, adding 1.90g of 4-bromocatechol, 3.45g of potassium carbonate, 3.80g of methyl bromoacetate and 50ml of DMF, reacting at 50 ℃ for 5.0h, stopping the reaction, cooling to room temperature, adding 300ml of water, extracting with 150ml of x 2 ethyl acetate, combining organic phases, drying by anhydrous sodium sulfate, and concentrating under reduced pressure to obtain 3.25g of compound I-19-1.
Synthesis of Compound I-19-2: adding 3.25g of compound I-19-1, 1.25g of potassium tert-butoxide and 50ml of tetrahydrofuran into a 100ml reaction flask, heating to 70 ℃ for reaction for 1.0h, stopping the reaction, cooling to room temperature, adding 100ml of water, extracting with 50ml of 2 ethyl acetate, combining organic phases, drying by anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 1.50g of compound I-19-2.
Synthesis of Compound I-19-3: adding 1.50g of compound I-19-2, 30ml of ethanol and 5ml of hydrochloric acid into a 100ml reaction bottle, heating to reflux, reacting for 3.0h, stopping reaction, cooling to room temperature, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 1.00g of compound I-19-3.
Synthesis of Compound I-19-4: adding 850mg of compound I-19-3, 708mg of dimethylamine hydrochloride, 550mg of sodium cyanoborohydride, 10ml of DMF and 4d of glacial acetic acid into a 100ml single-neck bottle, reacting at 50 ℃ for 12.0h, stopping the reaction, cooling to room temperature, pouring the reaction solution into 100ml of water, extracting with 50ml of ethyl acetate by 2, combining organic phases, drying by anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 765mg of compound I-19-4.
Synthesis methods of Compounds I-19-5 and I-19 reference the synthesis method of Compound I-1, and 12mg of Compound I-19 was obtained by substituting Compound I-1-2 for I-19-4. MS M/z 697.2(M +1),1H NMR(400MHz,d6-DMSO)δ:10.02(s,1H),7.99-7.97(m,1H),7.78-7.69(m,3H),7.48-7.35(m,4H),7.18-7.14(m,1H),6.95-6.94(m,1H),6.86-6.82(m,1H),6.55-6.53(m,1H),5.17-5.16(m,4H),4.13-3.88(m,4H),3.69-3.62(m,3H),3.21-3.20(m,1H),2.65(s,3H),2.43-2.32(m,2H),2.26(s,6H),1.85-1.82(m,2H),1.73-1.58(m,4H)ppm。
EXAMPLE 20 Synthesis of Compound I-20
Figure BDA0002221622080000251
Synthesis of Compound I-20-1: adding 4.50g of compound I-19-3, 80ml of toluene, 0.35g of p-toluenesulfonic acid and 4.25g of ethanol into a 100ml reaction bottle, heating to reflux reaction for 3.0h, stopping reaction, cooling to room temperature, concentrating under reduced pressure, drying, and purifying by column chromatography to obtain 5.32g of compound I-20-1.
Synthesis methods of Compounds I-20-2, I-20-3, I-20-4 and I-20-5 reference the synthesis method of Compound I-2 to prepare by substituting Compound I-1-2 for I-20-1 to obtain 1.02g of Compound I-20-5.
Synthesis of Compound I-20-6: a250 ml reaction flask was taken, and 1.02g of Compound I-20-5, 100ml of THF and 15ml of 6N hydrochloric acid were added to react at room temperature for 2.0 hours, the reaction was stopped, the pH was adjusted to 6.0-7.0 with saturated sodium bicarbonate, extraction was performed with ethyl acetate (200 ml. times.2), the organic phases were combined, washed with saturated brine (100ml), dried over anhydrous sodium sulfate, concentrated under reduced pressure to dryness, and purified by column chromatography to obtain 0.43g of Compound I-20-6.
Synthesis of Compound I-20: adding 250mg of compound I-20-6, 84mg of glycine, 76mg of sodium cyanoborohydride, 6ml of DMF and 1d of glacial acetic acid into a 100ml single-mouth bottle, reacting at 50 ℃ for 12.0h, stopping the reaction, pouring the reaction liquid into 50ml of water, extracting with ethyl acetate 25ml of x 2, combining organic phases, drying by anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 15mg of compound I-20. MS M/z 727.2(M +1),1H NMR(400MHz,d6-DMSO)δ:10.08(s,1H),9.85(s,1H),7.97-7.92(m,1H),7.78-7.69(m,3H),7.49-7.35(m,4H),7.17-7.16(m,1H),6.95-6.92(m,1H),6.87-6.85(m,1H),6.56-6.53(m,1H),5.54-5.52(m,1H),5.17-5.16(m,4H),4.17-3.88(m,4H),3.63-3.62(m,3H),3.49-3.46(m,2H),3.22-3.20(m,1H),2.69(s,3H),2.41-2.38(m,2H),1.86-1.81(m,2H),1.76-1.59(m,4H)ppm。
EXAMPLE 21 Synthesis of Compound I-21
Figure BDA0002221622080000261
Synthesis of Compound I-21-1: taking a 100ml reaction bottle, adding 4.50g of the compound I-19-3, 80ml of toluene, 0.35g of p-toluenesulfonic acid and 3.45g of ethylene glycol, heating to reflux reaction for 3.0h, stopping the reaction, cooling to room temperature, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 4.32g of the compound I-21-1.
Synthesis methods of Compounds I-21-2 and I-21 reference the synthesis method of Compound I-1, substituting Compound I-1-2 for I-21-1 to obtain 16mg of Compound I-21. MS M/z 712.2(M +1),1H NMR(400MHz,d6-DMSO)δ:10.05(s,1H),7.90-7.89(m,1H),7.79-7.69(m,3H),7.48-7.32(m,4H),7.16-7.14(m,1H),6.97-6.95(m,1H),6.84-6.82(m,1H),6.53-6.50(m,1H),5.17-5.16(m,4H),4.18-4.16(m,4H),3.85-3.74(m,4H),3.63-3.60(m,2H),3.22-3.20(m,1H),2.68(s,3H),2.39-2.25(m,2H),1.87-1.46(m,6H)ppm。
EXAMPLE 22 Synthesis of Compound I-22
Figure BDA0002221622080000262
Synthesis method of Compound I-22-1 the compound I-1-4 was prepared by substituting 3-cyanobenzylbromide with 2-cyano-4-bromomethylpyridine to give 1.02g of Compound I-22-1.
Synthesis methods of Compounds I-22-2 and I-22 reference the synthesis method of Compound I-1, and Compound I-1-2 was replaced with I-19-4 to give 19mg of Compound I-22. MS M/z 698.2(M +1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),8.86-8.85(m,1H),8.41-8.40(m,1H),8.25-8.24(m,1H),7.68-7.66(m,1H),7.48-7.32(m,4H),6.97-6.95(m,1H),6.84-6.82(m,1H),6.53-6.50(m,1H),5.93-5.91(m,2H),5.16(s,2H),4.11-3.86(m,4H),3.63-3.62(m,3H),3.22-3.20(m,1H),2.68(s,3H),2.29-2.18(m,2H),2.26(s,6H),1.88-1.49(m,6H)ppm。
EXAMPLE 23 Synthesis of Compound I-23
Figure BDA0002221622080000271
Synthesis of Compound I-23 reference was made to the synthesis of Compound I-22, L-homoproline was replaced with cis-4-hydroxy-L-proline to give 12mg of Compound I-23. MS (Mass Spectrometry)m/z=700.2(M+1),1H NMR(400MHz,d6-DMSO)δ:10.08(s,1H),8.86-8.84(m,1H),8.41-8.40(m,1H),8.25-8.23(m,1H),7.68-7.66(m,1H),7.48-7.32(m,4H),6.97-6.95(m,1H),6.84-6.82(m,1H),6.53-6.50(m,1H),5.93-5.91(m,2H),5.38-5.36(m,1H),5.16(s,2H),4.11-3.86(m,4H),3.69-3.62(m,4H),3.22-3.20(m,1H),2.68(s,3H),2.25-2.14(m,2H),2.26(s,6H),2.09-1.84(m,2H)ppm。
EXAMPLE 24 Synthesis of Compound I-24
Figure BDA0002221622080000272
Synthesis of Compound I-24 reference was made to the synthesis of Compound I-22 by substituting L-homoproline for N- [2- (aminooxy) ethyl]Acetamide to give 12mg of Compound I-24. MS M/z 673.2(M +1),1H NMR(400MHz,d6-DMSO)δ:9.96(s,1H),9.17(s,1H),8.86-8.84(m,1H),8.41-8.40(m,1H),8.23-8.20(m,1H),7.68-7.66(m,1H),7.48-7.32(m,4H),6.97-6.95(m,1H),6.84-6.82(m,1H),6.53-6.50(m,1H),5.93-5.91(m,2H),5.43-5.42(m,2H),5.16(s,2H),4.11-3.86(m,6H),3.62-3.60(m,1H),2.67(s,3H),2.26(s,6H),1.83-1.80(m,3H)ppm。
EXAMPLE 25 Synthesis of Compound I-25
Figure BDA0002221622080000281
Synthesis of Compound I-25-1: adding 4.50g of 2-bromo-5, 6,8, 9-tetrahydro-benzo-7-cycloheptanone, 80ml of toluene, 0.35g of p-toluenesulfonic acid and 3.50g of ethylene glycol into a 100ml reaction bottle, heating to reflux for 3.0h, stopping reaction, cooling to room temperature, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 4.52g of a compound I-25-1.
Synthesis methods of Compound I-25-2 and Compound I-25-3 the compound I-1 was prepared by referring to the synthesis method of Compound I-1, and Compound I-25-1 was substituted for Compound I-2 to obtain 1.20g of Compound I-25-3.
Synthesis of Compound I-25-4: a250 ml reaction flask was taken, and 1.20g of Compound I-25-3, 100ml of THF and 10ml of 6N hydrochloric acid were added thereto, and the reaction was allowed to proceed at room temperature for 2.0 hours, and then stopped, the pH was adjusted to 6.0-7.0 with saturated sodium bicarbonate, and the mixture was extracted with ethyl acetate (200 ml. times.2), and the organic phases were combined, washed with saturated brine (100ml), dried over anhydrous sodium sulfate and concentrated under reduced pressure to dryness to obtain 0.86g of Compound I-25-4.
Synthesis of Compound I-25: adding 100mg of compound I-25-4 and 10ml of methanol into a 100ml reaction bottle, cooling to 0 ℃, adding 15mg of sodium borohydride, reacting at room temperature for 3.0h, stopping the reaction, adding 2ml of saturated ammonium chloride, quenching, concentrating under reduced pressure, drying, and purifying by column chromatography to obtain 15mg of compound I-25. MS M/z 668.2(M +1),1H NMR(400MHz,d6-DMSO)δ:10.02(s,1H),7.89-7.68(m,5H),7.48-7.27(m,5H),7.16(m,1H),6.53-6.52(m,1H),5.38-5.37(m,1H),5.16(s,4H),4.48(s,1H),3.69-3.62(m,3H),3.39(s,1H),3.22-3.20(m,1H),2.91-2.80(m,4H),2.60-2.37(m,5H),2.09-1.85(m,2H),1.88-1.63(m,4H)ppm。
EXAMPLE 26 Synthesis of Compound I-26
Figure BDA0002221622080000282
Synthesis of Compound I-26: adding 200mg of compound I-25-3, 73mg of dimethylamine hydrochloride, 61mg of sodium cyanoborohydride, 5ml of DMF and glacial acetic acid 1d into a 100ml reaction bottle, reacting at 50 ℃ for 12.0h, stopping the reaction, cooling to room temperature, pouring the reaction solution into 50ml of water, extracting with ethyl acetate (25ml of 2), combining organic phases, drying with anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 12mg of compound I-26. MS M/z 695.3(M +1),1H NMR(400MHz,d6-DMSO)δ:10.05(s,1H),7.89-7.68(m,5H),7.48-7.27(m,5H),7.16-7.15(m,1H),6.53-6.50(m,1H),5.38-5.35(m,1H),5.16-5.14(s,4H),4.48(s,1H),3.69-3.62(m,3H),3.39(s,1H),3.22-3.20(m,1H),2.91-2.80(m,4H),2.60-2.37(m,5H),2.09-1.85(m,2H),1.88-1.63(m,4H)ppm。
EXAMPLE 27 Synthesis of Compound I-27
Figure BDA0002221622080000291
Synthesis of Compound I-27 reference was made to the synthesis of Compound I-26, substituting dimethylamine hydrochloride for pyrrolidine to give 12mg of Compound I-27. MS M/z 721.3(M +1),1H NMR(400MHz,d6-DMSO)δ:10.08(s,1H),7.89-7.68(m,5H),7.48-7.27(m,5H),7.16-7.14(m,1H),6.53-6.50(m,1H),5.38-5.36(m,1H),5.16(s,4H),3.69-3.62(m,3H),3.22-3.20(m,1H),2.91-2.80(m,4H),2.60-2.34(m,10H),2.09-1.85(m,2H),1.59-1.52(m,8H)ppm。
EXAMPLE 28 Synthesis of Compound I-28
Figure BDA0002221622080000292
Synthesis of Compound I-28 preparation of Compound I-2 by substituting Compound I-1-2 for I-19-4, 5-chloro-2, 4-dihydroxybenzaldehyde for 4-hydroxy-2, 6-dimethoxybenzaldehyde gave 18mg of Compound I-28. MS M/z 588.8(M +1),1H NMR(400MHz,d6-DMSO)δ:10.08(s,1H),7.81-7.80(m,1H),7.69-7.67(m,1H),7.47-7.38(m,4H),7.16-7.14(m,1H),6.94-6.86(m,2H),6.54-6.52(m,1H),5.15(s,2H),4.16(s,1H),3.82-3.80(m,2H),3.72-3.71(m,6H),3.62-3.60(m,1H),2.92-2.70(m,7H),2.60-2.58(m,3H),2.43-2.40(m,1H),2.27-2.24(m,6H),2.03-1.75(m,3H),1.78-1.52(m,4H)ppm。
EXAMPLE 29 Synthesis of Compound I-29
Figure BDA0002221622080000301
Synthesis of Compound I-29 preparation of reference to the Synthesis of Compound I-25, cis-4-hydroxy-L-proline was replaced by L-proline to give 15mg of Compound I-29. MS M/z 670.2(M +1),1H NMR(400MHz,d6-DMSO)δ:10.02(s,1H),7.89-7.86(m,1H),7.79-7.68(m,3H),7.48-7.32(m,4H),7.16-7.15(m,1H),6.95-6.85(m,2H),7.33-7.31(m,1H),5.78(s,1H),5.17-5.15(m,4H),4.61-4.60(m,1H),4.20-3.94(m,4H),3.62-3.60(m,2H),3.21-3.20(m,1H),2.60-2.39(m,5H),1.86-1.60(m,2H),1.55-1.46(m,4H)ppm。
EXAMPLE 30 Synthesis of Compound I-30
Figure BDA0002221622080000302
Synthesis of Compound I-30-1: and (2) adding 1.20g of compound I-19-3, 2.00g of methoxy formyl methylene triphenylphosphine and 50ml of dichloromethane into a 100ml reaction bottle, reacting at room temperature for 6.0h, stopping the reaction, adding 50ml of water, separating, retaining an organic phase, performing reduced pressure spin drying, and performing column chromatography purification to obtain 1.05g of compound I-30-1.
Synthesis of Compound I-30-2: adding 1.05g of compound I-30-1, 0.10g of Pd/C and 20ml of methanol into a 100ml reaction bottle, replacing hydrogen for 3 times, carrying out hydrogen protection, reacting at 50 ℃ for 5.0h, stopping the reaction, cooling to room temperature, carrying out auxiliary filtration by using kieselguhr, concentrating under reduced pressure to dry, and directly putting into the next step.
Synthesis of Compound I-30-3: adding the compound I-30-2 and 20ml of methanol solution into a 100ml reaction bottle, adding 0.28g of sodium hydroxide and 1ml of water, reacting for 5.0h at room temperature, stopping the reaction, concentrating under reduced pressure to remove most of the solvent, adding 20ml of water, adjusting the pH to 3.0-4.0 by using dilute hydrochloric acid, adding ethyl acetate (25ml of 2), extracting, combining organic phases, drying by using anhydrous sodium sulfate, and performing rotary drying under reduced pressure to obtain 0.85g of the compound I-30-3.
Synthesis methods of Compounds I-30-4 and I-30 reference the synthesis method of Compound I-1, and Compound I-30-3 was substituted for Compound I-1-2 to obtain 14mg of Compound I-30. MS M/z 712.3(M +1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),11.78(s,1H),7.89-7.86(m,1H),7.79-7.68(m,3H)7.48-7.32(m,4H),7.16-7.15(m,1H),6.95-6.85(m,2H),7.33-7.31(m,1H),5.17-5.15(m,4H),3.98-3.70(m,4H),3.62-3.60(m,2H),3.21-3.20(m,1H),2.83-2.80(m,1H),2.60-2.39(m,5H),2.26-2.00(m,2H),1.86-1.60(m,2H),1.55-1.46(m,4H)ppm。
EXAMPLE 31 Synthesis of Compound I-31
Figure BDA0002221622080000311
Synthesis method of Compound I-31-2 the compound is prepared by referring to the synthesis method of Compound I-25-4, and cis-4-hydroxy-L-proline is replaced by L-homoproline.
Synthesis of Compound I-31 referring to Compound I-30-1, 17mg of Compound I-31 was obtained. MS M/z 724.7(M +1),1H NMR(400MHz,d6-DMSO)δ:9.85(s,1H),8.12-8.08(m,1H),7.99-7.85(m,1H),7.78-7.60(m,2H),7.26-7.19(m,4H),7.10-6.91(m,3H),6.42-6.40(m,1H),6.08(s,1H),5.52-5.41(m,4H),4.68-4.60(m,4H),3.82(s,3H),3.62-3.58(m,2H),3.09-3.02(m,1H),2.71(s,3H),2.55-2.40(m,2H),1.86-1.62(m,2H),1.60-1.42(m,4H)ppm。
EXAMPLE 32 Synthesis of Compound I-32
Figure BDA0002221622080000312
Synthesis of Compound I-32 referring to the preparation of Compound I-26, dimethylamine hydrochloride was replaced by N, N, N' -trimethylethylenediamine to give 12mg of Compound I-32. MS M/z 754.5(M +1),1H NMR(400MHz,d6-DMSO)δ:9.86(s,1H),8.13-8.09(m,1H),7.95-7.83(m,1H),7.76-7.62(m,2H),7.28-7.17(m,4H),7.11-6.92(m,3H),6.43-6.40(m,1H),5.50-5.43(m,4H),4.18-4.10(m,4H),3.72-3.61(m,3H),3.13-3.08(m,1H),2.68(s,3H),2.55-2.45(m,2H),2.50-2.43(m,4H),2.26(s,9H),1.88-1.67(m,2H),1.55-1.40(m,4H)ppm
EXAMPLE 33 Synthesis of Compound I-33
Figure BDA0002221622080000321
Synthesis method of Compound I-33 reference was made to the synthesis method of Compound I-26, substituting dimethylamine hydrochloride for ethanolamine to obtain 12mg of Compound I-33. MS M/z 713.4(M +1),1H NMR(400MHz,d6-DMSO)δ:9.88(s,1H),8.10-8.07(m,1H),7.92-7.85(m,1H),7.78-7.64(m,2H),7.26-7.16(m,4H),7.10-6.91(m,3H),6.42-6.40(m,1H),5.48-5.41(m,4H),4.15-4.02(m,4H),3.81(s,1H),3.77-3.65(m,3H),3.55-3.42(m,2H),3.15-3.09(m,1H),2.82-2.74(m,2H),2.68(s,3H),2.55-2.41(m,2H),2.08(s,1H),1.87-1.68(m,2H),1.58-1.44(m,4H)ppm。
EXAMPLE 34 Synthesis of Compound I-34
Figure BDA0002221622080000322
Synthesis of Compound I-34-3 reference was made to the Synthesis of Compound I-18 to give 630mg of Compound I-34-3.
Synthesis methods of Compounds I-34-4 and I-34 reference to the Synthesis methods of Compounds I-25-4 and I-26, N, N, N' -trimethylethylenediamine was used to obtain 10mg of Compound I-34. MS M/z 755.3(M +1),1H NMR(400MHz,d6-DMSO)δ:10.05(s,1H),8.15-8.10(m,1H),7.98-7.86(m,1H),7.77-7.66(m,2H),7.27-7.19(m,4H),7.13-6.95(m,3H),6.45-6.41(m,1H),6.09(m,1H),5.54-5.47(m,4H),5.22-5.18(m,1H),4.62-4.59(m,2H),4.16-4.10(m,4H),3.74-3.52(m,3H),2.68(s,3H),2.56-2.43(m,4H),2.27(s,9H)ppm。
EXAMPLE 35 Synthesis of Compound I-35
Figure BDA0002221622080000331
Synthesis of Compound I-35 the compound I-1 was prepared by substituting 5-bromo-2, 2-difluoro-1, 3-benzodioxole for Compound I-1-2 and L-homoproline for L-proline to give 15mg of Compound I-35. MS M/z 648.4(M +1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),8.13-8.08(m,1H),7.96-7.87(m,1H),7.79-7.68(m,2H),7.28-7.17(m,4H),7.15-6.93(m,3H),6.46-6.43(m,1H),5.38-5.31(m,4H),3.72-3.64(m,2H),3.18-3.12(m,1H),2.69(s,3H),2.45-2.35(m,2H),2.02-1.83(m,2H),1.64-1.45(m,2H)ppm。
EXAMPLE 36 Synthesis of Compound I-36
Figure BDA0002221622080000332
Synthesis of Compound I-36 the compound I-1 was prepared by the same method as that for Compound I-1, substituting 5-bromo-2, 2-difluoro-1, 3-benzodioxole for Compound I-1-2 to give 13mg of Compound I-36. MS M/z 662.1(M +1),1H NMR(400MHz,d6-DMSO)δ:10.08(s,1H),8.10-8.06(m,1H),7.95-7.88(m,1H),7.76-7.65(m,2H),7.26-7.15(m,4H),7.17-6.92(m,3H),6.44-6.40(m,1H),5.34-5.30(m,4H),3.74-3.62(m,2H),3.16-3.11(m,1H),2.69(s,3H),2.55-2.42(m,2H),2.05-1.86(m,2H),1.62-1.44(m,4H)ppm。
EXAMPLE 37 Synthesis of Compound I-37
Figure BDA0002221622080000341
Synthesis of Compound I-37-1: taking a 100ml reaction bottle, adding 4.0g of 4-bromo-2-nitrofluorobenzene, 2.40g N, N-dimethylethylenediamine and 20ml of ethanol, reacting at 75 ℃ for 12.0h, stopping the reaction, cooling to room temperature, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 3.05g of a compound I-37-1.
Synthesis of Compound I-37-2: adding 3.05g of compound I-37-1, 0.6g of Pd/C and 30ml of ethanol into a 100ml reaction bottle, reacting at 50 ℃ for 6.0h, stopping the reaction, cooling to room temperature, filtering with diatomite as an auxiliary agent, concentrating under reduced pressure to dryness, and purifying by column chromatography to obtain 2.50g of compound I-37-2.
Synthesis of Compound I-37-3: adding 2.50g of compound I-37-2, 10ml of water and 2ml of sulfuric acid into a 100ml reaction bottle, stirring, dissolving, cooling to 0 ℃, and slowly dropwise adding 0.67g of NaNO2Adding 2ml of water, keeping the temperature at 0 ℃ for reaction for 2.0h after the dropwise addition is finished, stopping the reaction, performing suction filtration and drying to obtain 1.20g of a compound I-37-3.
Synthesis method of Compound I-37 reference was made to the synthesis method of Compound I-1, where Compound I-1-2 was replaced with I-37-3 to give 12mg of Compound I-37. MS M/z 694.2(M +1),1H NMR(400MHz,d6-DMSO)δ:10.05(s,1H),8.22-8.03(m,3H),7.93-7.84(m,1H),7.78-7.69(m,1H),7.65-7.62(m,2H),7.26-7.19(m,3H),7.11-7.09(m,1H),6.44-6.40(m,1H),5.55-5.49(m,2H),5.23-5.16(m,4H),3.78-3.62(m,4H),3.16-3.08(m,1H),2.89(s,6H),2.68(s,3H),2.55-2.46(m,2H),1.88-1.69(m,2H),1.58-1.46(m,4H)ppm。
EXAMPLE 38 Synthesis of Compound I-38
Figure BDA0002221622080000351
Synthesis of Compound I-38 reference was made to the Synthesis of Compound I-37, substituting isopropylamine with N, N-dimethylethylenediamine to give 16mg of Compound I-38.
MS m/z=666.5(M+1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),8.55-8.54(m,1H),8.48-8.46(m,1H),8.18-8.10(m,2H),7.99-7.96(m,1H),7.78-7.69(m,2H),7.26-7.19(m,3H),6.86-6.83(m,1H),5.25-5.19(m,4H),4.14-4.08(m,1H),3.78-3.62(m,2H),3.11-3.09(m,1H),2.68(s,3H),2.53-2.47(m,2H),1.86-1.69(m,2H),1.62-1.55(m,6H),1.48-1.42(m,4H)ppm。
EXAMPLE 39 Synthesis of Compound I-39
Figure BDA0002221622080000352
Synthesis method of Compound I-39 reference was made to the Synthesis method of Compound I-37, substituting ethanolamine for N, N-dimethylethylenediamine to give 16mg of Compound I-39.
MS m/z=653.4(M+1),1H NMR(400MHz,d6-DMSO)δ:10.07(s,1H),8.25-8.06(m,3H),7.96-7.88(m,1H),7.79-7.66(m,1H),7.64-7.60(m,2H),7.28-7.17(m,3H),7.12-7.08(m,1H),6.49-6.46(m,1H),5.25-5.13(m,4H),5.07(s,1H),4.08-4.02(m,2H),3.78-3.62(m,4H),3.13-3.05(m,1H),2.68(s,3H),2.44-2.33(m,2H),1.98-1.85(m,2H),1.57-1.49(m,2H)ppm。
EXAMPLE 40 Synthesis of Compound I-40
Figure BDA0002221622080000361
Synthesis of Compound I-40-1: adding 3.00g of 5-bromo-2-indanone, 2.88g of dimethylamine hydrochloride, 2.41g of sodium cyanoborohydride, 15ml of DMF and glacial acetic acid 5d into a 100ml reaction bottle, reacting at 50 ℃ for 12.0h, stopping the reaction, cooling to room temperature, pouring the reaction liquid into 150ml of water, extracting by using ethyl acetate (75ml of 2), combining organic phases, drying by using anhydrous sodium sulfate, concentrating under reduced pressure to dryness, and purifying by using column chromatography to obtain 2.50g of a compound I-40-1.
Synthesis method of Compound I-40 reference was made to the synthesis of Compound I-1, where Compound I-1-2 was replaced with I-40-1, to obtain 13mg of Compound I-40.
MS m/z=665.3(M+1),1H NMR(400MHz,d6-DMSO)δ:10.04(s,1H),8.11-8.08(m,1H),7.89-7.86(m,1H),7.79-7.70(m,1H),7.66-7.40(m,3H),7.28-7.18(m,4H),7.11-7.07(m,1H),6.46-6.41(m,1H),5.26-5.16(m,4H),3.74-3.61(m,2H),3.25-3.17(m,1H),3.13-3.04(m,1H),,3.01-2.88(m,4H),2.68(s,3H),2.54-2.45(m,2H),2.26(s,6H),1.84-1.68(m,2H),1.52-1.43(m,4H)ppm。
EXAMPLE 41 Synthesis of Compound I-41
Figure BDA0002221622080000371
Synthesis of Compound I-41 referring to the preparation of Compound I-40, dimethylamine hydrochloride was replaced with N-methyl-L-alanine to give 16mg of Compound I-41.
MS m/z=723.5(M+1),1H NMR(400MHz,d6-DMSO)δ:10.04(s,1H),9.86(s,1H),8.16-8.09(m,1H),7.88-7.87(m,1H),7.76-7.74(m,1H),7.67-7.45(m,3H),7.27-7.17(m,4H),7.10-7.08(m,1H),6.43-6.41(m,1H),5.26-5.16(m,4H),3.75-3.62(m,3H),3.22-3.17(m,1H),3.11-3.05(m,1H),3.02-2.89(m,4H),2.68(s,3H),2.57-2.46(m,2H),2.26(s,3H),1.87-1.69(m,2H),1.55-1.41(m,4H),1.25-1.11(m,3H)ppm。
EXAMPLE 42 Synthesis of Compound I-42
Figure BDA0002221622080000372
Synthesis of Compound I-42-1: taking a 100ml reaction bottle, adding 2.0g of 6-bromo-2H-1, 4-benzoxazine-3 (4H) -ketone, 3.50g of Lawson reagent and 30ml of toluene, heating to reflux reaction for 5.0H, stopping the reaction, cooling to room temperature, adding 30ml of water, separating, concentrating under reduced pressure organic phase, and purifying by column chromatography to obtain 1.20g of a compound I-42-1.
Synthesis of Compound I-42-2: taking a 100ml reaction bottle, adding 1.20g of compound I-42-1, 1.20g of dimethylamine hydrochloride, 1.47g of triethylamine and 30ml of ethanol, reacting at 50 ℃ for 12.0h, stopping the reaction, cooling to room temperature, concentrating under reduced pressure, drying, and carrying out column chromatography to obtain 0.65g of compound I-42-2.
Synthesis methods of Compounds I-42-3 and I-42 reference the synthesis method of Compound I-1, and Compound I-1-2 was replaced with I-42-2 to obtain 20mg of Compound I-42.
MS m/z=680.3(M+1),1H NMR(400MHz,d6-DMSO)δ:10.05(s,1H),8.13-8.07(m,1H),7.88-7.85(m,1H),7.76-7.71(m,1H),7.65-7.43(m,3H),7.24-7.19(m,4H),7.12-7.08(m,1H),6.47-6.41(m,1H),5.22-5.13(m,4H),3.92-3.88(m,2H),3.72-3.60(m,2H),3.13-3.04(m,1H),2.98(s,6H),2.68(s,3H),2.56-2.47(m,2H),1.83-1.69(m,2H),1.55-1.41(m,4H)ppm。
EXAMPLE 43 Synthesis of Compound I-43
Figure BDA0002221622080000381
Synthesis of Compound I-43 reference was made to the synthesis of Compound I-42, substituting Compound I-1-4 for Compound I-22-1 to give 12mg of Compound I-43.
MS m/z=681.3(M+1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),8.56-8.53(m,1H),8.46-8.42(m,1H),8.16-8.10(m,2H),7.98-7.95(m,1H),7.76-7.67(m,2H),7.28-7.13(m,3H),6.88-6.85(m,1H),5.26-5.16(m,4H),3.93-3.88(m,2H),3.77-3.62(m,2H),3.11-3.10(m,1H),2.98(s,6H),2.68(s,3H),2.54-2.46(m,2H),1.86-1.71(m,2H),1.49-1.43(m,4H)ppm。
EXAMPLE 44 Synthesis of Compound I-44
Figure BDA0002221622080000382
Synthesis of Compound I-44 the compound I-44 was prepared by reference to the synthesis of Compound I-42, substituting L-homoproline for L-proline to give 12mg of Compound I-44.
MS m/z=666.1(M+1),1H NMR(400MHz,d6-DMSO)δ:9.86(s,1H),8.15-8.08(m,1H),7.89-7.82(m,1H),7.77-7.73(m,1H),7.63-7.49(m,3H),7.26-7.18(m,4H),7.11-7.04(m,1H),6.48-6.43(m,1H),5.21-5.16(m,4H),3.93-3.89(m,2H),3.71-3.60(m,2H),3.11-3.07(m,1H),2.98(s,6H),2.69(s,3H),2.40-2.37(m,2H),1.89-1.79(m,1H),1.65-1.42(m,3H)ppm。
EXAMPLE 45 Synthesis of Compound I-45
Figure BDA0002221622080000391
Synthesis methods of Compound I-45-1, Compound I-45-2 and Compound I-45 preparation reference was made to the Synthesis method of Compound I-42, dimethylamine was replaced by N, N, N' -trimethylethylenediamine, L-homoproline was replaced by L-proline to give 15mg of Compound I-45.
MS m/z=724.2(M+1),1H NMR(400MHz,d6-DMSO)δ:9.88(s,1H),8.54-8.51(m,1H),8.43-8.42(m,1H),8.14-8.12(m,2H),7.99-7.98(m,1H),7.77-7.69(m,2H),7.25-7.18(m,3H),6.85-6.82(m,1H),5.22-5.17(m,4H),3.92-3.87(m,2H),3.72-3.63(m,2H),3.13-3.11(m,1H),2.98(s,3H),2.68-2.62(m,5H),2.42-2.31(m,4H),2.26(s,6H),1.89-1.77(m,1H),1.55-1.45(m,3H)ppm。
EXAMPLE 46 Synthesis of Compound I-46
Figure BDA0002221622080000392
Synthesis of Compound I-46-1: adding 3.20g of 4-bromocatechol, 5.12g of 3-bromopropylene, 5.84g of potassium carbonate and 20ml of DMF into a 100ml reaction bottle, reacting at 50 ℃ for 6.0h, stopping the reaction, cooling to room temperature, adding 200ml of water, extracting with 100ml of 2 ethyl acetate, combining organic phases, drying by anhydrous sodium sulfate, performing reduced pressure spin drying, and purifying by column chromatography to obtain 3.50g of a compound I-46-1.
Synthesis of Compound I-46-2: adding 3.00g of compound I-46-1, 1.80g of phenylmethylenebis (tricyclohexylphosphorus) ruthenium dichloride, 5ml of dichloromethane and 20ml of benzene into a 100ml reaction bottle, reacting at 55 ℃ for 17.0h, stopping the reaction, cooling to room temperature, removing the solvent by rotary removal under reduced pressure, and purifying by column chromatography to obtain 1.80g of compound I-46-2.
Synthesis methods of Compounds I-46-3 and I-46 reference the synthesis method of Compound I-1, and Compound I-46-2 is substituted with Compound I-1-2 to obtain 12mg of Compound I-46.
MS m/z=667.7(M+1),1H NMR(400MHz,d6-DMSO)δ:10.06(s,1H),8.52-8.51(m,1H),8.44-8.42(m,1H),8.12-8.10(m,2H),7.96-7.95(m,1H),7.75-7.69(m,2H),7.24-7.17(m,3H),6.89-6.86(m,1H),6.05-5.95(m,2H),5.22-5.17(m,4H),4.73-4.68(m,4H),3.72-3.62(m,2H),3.13-3.09(m,1H),2.68(s,3H),2.53-2.48(m,2H),1.88-1.73(m,2H),1.46-1.42(m,4H)ppm。
EXAMPLE 47 Synthesis of Compound I-47
Figure BDA0002221622080000401
Synthesis of Compound I-47 the compound I-47 was prepared by reference to the synthesis of Compound I-46, substituting L-proline for L-homoproline to give 13mg of Compound I-47.
MS m/z=653.5(M+1),1H NMR(400MHz,d6-DMSO)δ:9.86(s,1H),8.56-8.54(m,1H),8.46-8.42(m,1H),8.10-8.07(m,2H),7.93-7.91(m,1H),7.78-7.74(m,2H),7.26-7.19(m,3H),6.88-6.87(m,1H),6.06-5.96(m,2H),5.20-5.15(m,4H),4.75-4.66(m,4H),3.73-3.61(m,2H),3.12-3.09(m,1H),2.68(s,3H),2.43-2.32(m,2H),1.88-1.77(m,2H),1.56-1.43(m,2H)ppm。
EXAMPLE 48 Synthesis of Compound I-48
Figure BDA0002221622080000402
Synthesis of Compound I-48-1: adding 0.60g of compound I-46-2, 20ml of methanol and 0.12g of Pd/C into a 100ml reaction bottle, replacing hydrogen for 3 times, reacting at 50 ℃ for 12.0h under hydrogen, stopping the reaction, cooling to room temperature, carrying out auxiliary filtration by using kieselguhr, and directly putting the filtrate into the next step after reduced pressure spin-drying.
Synthesis methods of Compound I-48-2 and Compound I-48 reference was made to the Synthesis methods of Compound I-46-3 and Compound I-46 to prepare by substituting Compound I-46-2 for I-48-1 and L-proline for L-homoproline to give 12mg of Compound I-48.
MS m/z=655.4(M+1),1H NMR(400MHz,d6-DMSO)δ:9.88(s,1H),8.53-8.50(m,1H),8.48-8.47(m,1H),8.11-8.09(m,2H),7.98-7.96(m,1H),7.79-7.76(m,2H),7.28-7.22(m,3H),6.89-6.88(m,1H),5.18-5.16(m,4H),4.08-3.99(m,4H),3.74-3.62(m,2H),3.11-3.10(m,1H),2.68(s,3H),2.41-2.32(m,2H),1.88-1.71(m,6H),1.54-1.42(m,2H)ppm。
EXAMPLE 49 Synthesis of Compound I-49
Figure BDA0002221622080000411
Synthesis of Compound I-49 preparation was carried out by reference to the synthesis of Compound I-7, substituting L-proline for L-homoproline to give 15mg of Compound I-49.
MS m/z=639.2(M+1),1H NMR(400MHz,d6-DMSO)δ:9.85(s,1H),8.76-8.75(m,1H),8.13-8.12(m,1H),7.84-7.82(m,1H),7.55-7.53(m,1H),7.42-7.41(m,1H),7.21-7.20(m,1H),7.15-7.11(m,1H),6.82-6.79(m,2H),6.63-6.61(m,1H),6.22-6.20(m,1H),5.42-5.37(m,2H),5.22-5.21(m,2H),4.35-4.05(m,4H),3.08-3.07(m,1H),2.26(s,3H),2.12-2.09(m,2H),1.86-1.83(m,2H),1.68-1.56(m,4H)ppm。
EXAMPLE 50 Synthesis of Compound I-50
Figure BDA0002221622080000412
Synthesis of Compound I-50 preparation was carried out by reference to the synthesis of Compound I-7, substituting L-proline for cis-4-hydroxy-L-proline to give 12mg of Compound I-50.
MS m/z=641.1(M+1),1H NMR(400MHz,d6-DMSO)δ:9.87(s,1H),8.77-8.76(m,1H),8.16-8.14(m,1H),7.89-7.86(m,1H),7.54-7.53(m,1H),7.42-7.41(m,1H),7.26-7.24(m,1H),7.20-7.17(m,1H),6.88-6.78(m,2H),6.67-6.66(m,1H),6.26-6.24(m,1H),5.51-5.38(m,2H),5.28-5.23(m,4H),4.07-4.06(m,2H),3.69(s,1H),3.53-3.42(m,1H),3.22-3.19(m,1H),2.26(s,3H),2.11-2.04(m,2H),1.84-1.77(m,2H)ppm。
Evaluation test of biological Activity
Example 51 evaluation of inhibitory Activity of the Compound of the present invention against PD-1/PD-L1 protein-protein interaction
These effects show that the compounds of the present invention have significant inhibitory effects on PD 1/PD-L1.
The specific test method is as follows:
purpose and principle of experiment
HTRF (Homogeneous time-Resolved Fluorescence) is a technique for detecting analytes in a pure liquid phase system. The technology is based on two technologies of Fluorescence Resonance Energy Transfer (FRET) and Time-Resolved Fluorescence (TRF), and opens a high-throughput drug screening instrument.
Experimental materials and instruments
HTRF kits are purchased from Cisbio (CAT #63ADK000 CPACPEG) and comprise Anti-Tag 1-captate, Anti-Tag2-XL665/d2, Tag1-PD-L1, Tag2-PD-1, Dilution Buffer, Detection Buffer and other reagents required by experiments. SpectraMax i3X multifunctional microplate reader was purchased from Molecular Devices. 384 shallow well plates, available from Nunc (CAT # 264706).
Test Compounds-Compounds from examples 1 to 50.
Experimental procedure
Detection was performed using HTRF kit. And setting a negative control group, a positive control group and an administration group. PD-1 recombinant protein and PD-L1 recombinant protein were diluted to 250nM and 25nM, respectively, with Dilution Buffer. 100 μ M DMSO-solubilized small molecule compounds were diluted with Dilution Buffer to 1 μ M and 0.1 μ M. To 384 wells, 2ul of diluted test compound, 4ul of diluted PD-1 and 4ul of diluted PDL-1 were added in sequence. Mixing, and standing at room temperature for 15 min. Diluting anti-Tag1-Eu with Detection buffer3+(1:25) and anti-Tag2-XL665(1:100), and then mixing the diluted detection reagent in equal volume, and adding 10. mu.l of antibody mixture to each reaction well. And (5) sealing the membrane and incubating for 2h at room temperature. Fluorescence signals (320nm stimulation, 665nm, 615nm emission) were detected with an envision (perkinelmer) instrument.
Data analysis
Inhibition ratio (ER)positive―ER sample)/(ERpositive―ERnegative) Data analysis was performed at 100%.
Results of the experiment
The inhibitory activity of the compounds of examples 1-50 was initially screened (100nM &10nM) using the Kit PD1/PD-L1 Binding Assay Kit (Cisbio/Cat #63ADK000CPAPEG) -HTRF technique, with PD-1/PD-L1inhibitor 1(Selleck, Cat # S7911) as a positive reference compound.
TABLE 1 protein level Activity of Compounds to inhibit PD-1/PD-L1 protein-protein interaction
Figure BDA0002221622080000421
Figure BDA0002221622080000431
Experimental results show that the compound has remarkable inhibitory activity of PD-1/PD-L1 protein-protein interaction. The PD-1/PD-L1 inhibitory activity of some compounds is significantly better than that of the positive control drug. This suggests that such compounds of the invention may be useful as immune checkpoint PD-1/PD-L1 inhibitors.
Example 52 cytotoxicity assays
Application of CCK-8 detection kit to detection of cytotoxicity IC of compound of embodiment of the invention on 4 tumor cells50Value experiment material and instrument four tumor cell strains
MDA-MB-231 human breast cancer cell line, ordered from ATCC in usa; MC-38 mouse colon cancer cell line, ordered from seiki (shanghai) bioengineering limited; HT29 human colon cancer cell line, ordered in Shanghai cell resource center of Chinese academy of sciences; SUM149 human breast cancer cell line, ordered from Shanghai cell resource center of Chinese academy of sciences.
Reagent and consumable
Cell Counting Kit-8(Cat # D3100L4057), ordered from Shanghai Liji Biotech Co., Ltd; 96-well plates (Cat #3599) ordered from Corning Costar; fetal bovine serum (Cat #10099-141), DMEM medium (Cat #11995-040) and F-12 medium (Cat #31765-035) were ordered into GIBCO. Desktop Microplate reader SpectraMax M5 Microplate, ordered from Molecular Devices.
Experimental procedure
Reagent preparation
Preparation of culture Medium
Cell lines Culture medium
HT29 DMEM+10%FBS
MDA-MB-231 RPMI1640+10%FBS
MC-38 DMEM+10%FBS
SUM149 F12+5μg/ml insulin+1μg/ml hydrocortisone+10%FBS
Preparing a stock solution of a substance to be detected: compounds were diluted with DMSO to a final concentration of 10 nM.
IC50 value test (CCK-8 test)
Cells in the logarithmic growth phase were collected, counted, resuspended in complete medium, adjusted to the appropriate concentration (as determined by the cell density optimization assay) and seeded into 96-well plates with 100. mu.l of cell suspension per well. Cells were incubated at 37 ℃ and 100% relative humidity, 5% CO2Incubate in incubator for 24 hours.
The test compound was diluted with the medium to the set corresponding effect concentration and the cells were added at 25. mu.l/well. Compound effect concentration was started at 100 μ M, diluted in 4-fold gradient, for 10 concentration points.
Cells were incubated at 37 ℃ and 100% relative humidity, 5% CO2Incubate for 72 hours in the incubator.
10. mu.l of CCK-8 was added directly to the cell culture medium and incubated in an incubator at 37 ℃ for 2-4 hours.
After gentle shaking, the absorbance at a wavelength of 450nm was measured on a SpectraMax M5 Microplate, and the inhibition was calculated using the absorbance at 650nm as a reference.
Data processing
The inhibition rate of the drug on the growth of tumor cells was calculated according to the following formula: tumor cell growth inhibition ratio [ (Ac-As)/(Ac-Ab) ] -. 100%
As: OA of the sample (cell + CCK-8+ test Compound)
Ac: OA of negative control (cell + CCK-8+ DMSO)
Ab: OA of positive control (cell + CCK-8+ DMSO)
The experimental result shows that the compound has obvious inhibition effect on 4 tumor cells, and the table 2 shows that the tumor cell inhibition activity of some compounds is obviously superior to that of a positive control medicament, and particularly, the inhibition activity on MC-38 and SUM149 is far greater than that of the positive control medicament.
Table 2: half inhibitory concentration IC of compound on proliferation of 4 tumor cells50(μM)
BMS-1166 I-12 I-50
HT29 0.025 0.010 0.012
MDA-MB-231 0.039 0.23 0.015
MC-38 0.58 0.012 0.005
SUM149 0.69 0.25 0.025
Example 53 pharmacokinetic experiments
Pharmacokinetic study experimental material for compound PD-1 administered to SD rat by single intravenous injection or oral gavage
SD rats, male and female, SPF grade.
Preparation of test article
5% DMSO + 10% Solutol + 85% sterile water for injection is used for intravenous injection; 5% DMSO + 10% Solutol + 85% sterile water for injection was used orally.
Experimental groups the compounds of the examples of the present invention were administered in groups according to the administration modes of groups 1 to 5, respectively, below.
Figure BDA0002221622080000441
Mode of administration
The weight was measured before administration, and the amount administered was calculated based on the body weight. Administration is by intravenous injection or intragastric administration.
Time point of blood sampling
IV: before administration, 5min, 15min, 30min, 1h, 2h, 4h, 6h, 8h and 24h after administration. PO: before administration, 15min, 30min, 1h, 2h, 4h, 6h, 8h, 10h and 24h after administration.
Sample collection and disposal
Blood is collected via jugular vein or other suitable method, each sample is collected about 0.20mL, heparin sodium is anticoagulated, the blood sample is placed on ice after collection, and plasma is centrifugally separated within 2 hours (centrifugal force: 6800g, 6 minutes, 2-8 ℃). The collected plasma sample is stored in a refrigerator at minus 80 ℃ before analysis, and the residual plasma sample after analysis is continuously stored in the refrigerator at minus 80 ℃ for temporary storage, wherein the storage life is one month.
Biological analysis and data processing
And (3) detecting the blood concentration of the sample, and calculating pharmacokinetic parameters such as AUC (0-T), T1/2, Cmax, Tmax, MRT and the like by using Phoenix WinNonlin through the blood concentration data of different time points. When calculating the drug-induced parameters, the concentration before administration is calculated according to 0; BLQ (including "No peak") before Cmax is calculated as 0; BLQ (including "No peak") appearing after Cmax does not participate in the calculation uniformly.
Results of the experiment
The pharmacokinetics and oral bioavailability of the compounds of the present invention were studied in rats after a single intravenous and oral administration. Experiments prove that: the compound of the invention shows good pharmacokinetic characteristics on rats, and has appropriate half-life period and excellent bioavailability.
The embodiments of the present invention have been described above. However, the present invention is not limited to the above embodiment. Any modification, equivalent replacement, or improvement made within the spirit and principle of the present invention should be included in the protection scope of the present invention.

Claims (12)

1. A compound of formula I or a pharmaceutically acceptable salt thereof:
Figure FDA0003644931310000011
wherein, R is1Is selected from
Figure FDA0003644931310000012
Figure FDA0003644931310000013
The R is2Is selected from H or (C)1-C12) An alkyl group;
each R3Each independently selected from halogen,
Figure FDA0003644931310000014
Figure FDA0003644931310000015
-O-(C1-C12) An alkyl group;
n is selected from 1,2, 3 and 4;
the R is4、R5Selected from H or optionally substituted with one, two or more R: (C)1-C12) Alkyl, said R is selected from halogen, CN, OH,
or said-NR4R5Is selected from
Figure FDA0003644931310000021
Figure FDA0003644931310000022
Figure FDA0003644931310000023
Wherein m is selected from 0, 1,2, 3;
the-W-is selected from- (C)1-C12) Alkyl-, - (C)1-C12) alkyl-O-.
2. A compound of formula I, or a pharmaceutically acceptable salt thereof, according to claim 1, wherein: said (C)1-C12) Alkyl is selected from (C)1-C6) An alkyl group.
3. A compound of formula I or a pharmaceutically acceptable salt thereof according to claim 1 or 2, wherein R is2Selected from methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl.
4. A compound of formula I or a pharmaceutically acceptable salt thereof according to claim 1 or 2,
R3selected from halogen,
Figure FDA0003644931310000024
Figure FDA0003644931310000025
Methoxy, ethoxy, propoxy, butoxy, pentoxy.
5. The compound of formula I or a pharmaceutically acceptable salt thereof according to claim 1 or 2, wherein the structure of formula I is further selected from the group consisting of formula II, formula III, formula IV, formula V, formula VI, formula VII:
Figure FDA0003644931310000026
Figure FDA0003644931310000031
in the formula II, the formula III, the formula IV, the formula V, the formula VI and the formula VII, R1、R2、R3、R4、R5W is as defined for formula I in claim 1 or 2.
6. The following compounds or pharmaceutically acceptable salts thereof:
Figure FDA0003644931310000032
Figure FDA0003644931310000041
Figure FDA0003644931310000051
7. the process according to claim 1, wherein the preparation of the compound of formula I or a pharmaceutically acceptable salt thereof is selected from the following preparation schemes: in a first aspect, the method comprises the steps of:
Figure FDA0003644931310000061
wherein R is1、R2、R3、R4、R5W, n is as defined for formula I in claim 1;
in a second aspect, the method comprises the steps of:
Figure FDA0003644931310000062
wherein R is1、R2、R3W, n is as defined for formula I in claim 1; x is selected from halogen;
in a third aspect, the method comprises the steps of:
Figure FDA0003644931310000063
wherein R is2、R3W, n is as defined for formula I in claim 1;
in a fourth aspect, the method comprises the steps of:
Figure FDA0003644931310000064
wherein R is1,R2、R3W, n is as defined for formula I in claim 1; x is selected from halogen.
8. A pharmaceutical composition comprising a compound of any one of claims 1-6, or a pharmaceutically acceptable salt thereof.
9. Use of a compound according to any one of claims 1 to 6 or a pharmaceutically acceptable salt thereof or a pharmaceutical composition according to claim 8 for the manufacture of a medicament for the prevention and/or treatment of a disease associated with the PD-1/PD-L1 signalling pathway.
10. The use of claim 9, wherein the disease associated with the PD-1/PD-L1 signaling pathway is selected from the group consisting of cancer, infectious disease, and autoimmune disease.
11. Use according to claim 10, wherein the cancer is selected from: melanoma, renal cancer, prostate cancer, breast cancer, colon cancer, lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head and neck, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, testicular cancer, carcinoma of the fallopian tubes, cancer of the cervix, cancer of the vagina, cancer of the vulva, hodgkin's disease, non-hodgkin's lymphoma, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, chronic or acute leukemia, solid tumor of the child, lymphocytic lymphomas, cancer of the bladder, carcinoma of the renal pelvis, primary CNS lymphoma, spinal axis tumor, brain stem glioma, kaposi's sarcoma, epidermoid carcinoma, squamous cell carcinoma, T-cell lymphoma.
12. Use according to claim 10, wherein the cancer is selected from metastatic malignant melanoma, clear cell carcinoma, hormone refractory prostate cancer, non-small cell lung cancer, acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia.
CN201910936108.4A 2019-09-29 2019-09-29 PD-1 inhibitor and preparation method and application thereof Active CN112574183B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910936108.4A CN112574183B (en) 2019-09-29 2019-09-29 PD-1 inhibitor and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910936108.4A CN112574183B (en) 2019-09-29 2019-09-29 PD-1 inhibitor and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN112574183A CN112574183A (en) 2021-03-30
CN112574183B true CN112574183B (en) 2022-07-08

Family

ID=75110765

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910936108.4A Active CN112574183B (en) 2019-09-29 2019-09-29 PD-1 inhibitor and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN112574183B (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117750950A (en) * 2021-07-30 2024-03-22 Jl生物疗法公司 Composition containing benzyl ethanolamine derivative as active ingredient for preventing or treating metabolic diseases

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105705489A (en) * 2013-09-04 2016-06-22 百时美施贵宝公司 Compounds useful as immunomodulators
CN106536515A (en) * 2014-04-14 2017-03-22 百时美施贵宝公司 Compounds useful as immunomodulators
US20180016260A1 (en) * 2016-07-14 2018-01-18 Incyte Corporation Heterocyclic compounds as immunomodulators
CN108368090A (en) * 2015-10-15 2018-08-03 百时美施贵宝公司 Compound as immunomodulator
CN109689640A (en) * 2016-07-08 2019-04-26 百时美施贵宝公司 It can be used as the compound of immunomodulator
CN109721527A (en) * 2017-10-27 2019-05-07 广州丹康医药生物有限公司 A kind of novel anti-PD-L1 compound, its application and the composition containing it
CN110092779A (en) * 2018-01-29 2019-08-06 广州丹康医药生物有限公司 A kind of substituted phenyl compound and its application

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105705489A (en) * 2013-09-04 2016-06-22 百时美施贵宝公司 Compounds useful as immunomodulators
CN106536515A (en) * 2014-04-14 2017-03-22 百时美施贵宝公司 Compounds useful as immunomodulators
CN108368090A (en) * 2015-10-15 2018-08-03 百时美施贵宝公司 Compound as immunomodulator
CN109689640A (en) * 2016-07-08 2019-04-26 百时美施贵宝公司 It can be used as the compound of immunomodulator
US20180016260A1 (en) * 2016-07-14 2018-01-18 Incyte Corporation Heterocyclic compounds as immunomodulators
CN109721527A (en) * 2017-10-27 2019-05-07 广州丹康医药生物有限公司 A kind of novel anti-PD-L1 compound, its application and the composition containing it
CN110092779A (en) * 2018-01-29 2019-08-06 广州丹康医药生物有限公司 A kind of substituted phenyl compound and its application

Also Published As

Publication number Publication date
CN112574183A (en) 2021-03-30

Similar Documents

Publication Publication Date Title
WO2021129820A1 (en) Spiro ring-containing quinazoline compound
KR102436430B1 (en) Inhibitors of Menin-MLL Interaction
CN113767103B (en) Novel spirocyclic K-Ras G12C inhibitors
CN108623615B (en) Macrocyclic derivatives of pyrazolo [3,4-d ] pyrimidin-3-one, pharmaceutical compositions and uses thereof
JP5628603B2 (en) Tetracyclic compound
WO2021129824A1 (en) New-type k-ras g12c inhibitor
JP2023519815A (en) Spiro ring-containing quinazoline compounds
JP6522646B2 (en) Substituted 4,5,6,7-tetrahydro-pyrazolo [1,5-α] pyrazine derivatives as ROS1 inhibitors and 5,6,7,8-tetrahydro-4H-pyrazolo [1,5-α] [1,1 4] Diazepine derivatives
EP3862348A1 (en) Isoindoline compound, preparation method, pharmaceutical composition and use thereof
CN110330479A (en) A kind of antitumoral compounds and application thereof as AXL inhibitor
JP2017508779A5 (en)
CN112430234A (en) Novel KRAS G12C protein inhibitor and preparation method and application thereof
CN113387962A (en) Pyrazolo [3,4-d ] pyrimidine-3-one derivative, pharmaceutical composition and application thereof
EP2928466B1 (en) Use of maleimide derivatives for preventing and treating leukemia
EP3954694A1 (en) Ubiquitin-specific protease inhibitors, and preparation method therefor and application thereof
CN110343101B (en) Estrogen receptor degrading agents for the treatment of breast cancer
WO2020192750A1 (en) Thienoheterocyclic derivative, preparation method therefor and medical use thereof
CN112574183B (en) PD-1 inhibitor and preparation method and application thereof
CN115028644A (en) SOS1 inhibitor heterocyclic compounds
CN113754659A (en) Spiro-containing quinazoline compounds
WO2022078480A1 (en) Triheterocyclic derivative, and pharmaceutical composition and application thereof
CN112538084B (en) Novel KRAS G12C protein inhibitor and preparation method and application thereof
EP4219504A1 (en) Macrocyclic tlr7 agonist, preparation method therefor, pharmaceutical composition and use thereof
JP2024508769A (en) Pyrimidine compounds as Wee-1 inhibitors
CN115490686A (en) Nitrogen-containing heterocyclic compound

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
GR01 Patent grant
GR01 Patent grant