CN106831614B - Substituted benzodiazacyclo compound and its preparation method and use - Google Patents

Substituted benzodiazacyclo compound and its preparation method and use Download PDF

Info

Publication number
CN106831614B
CN106831614B CN201710012668.1A CN201710012668A CN106831614B CN 106831614 B CN106831614 B CN 106831614B CN 201710012668 A CN201710012668 A CN 201710012668A CN 106831614 B CN106831614 B CN 106831614B
Authority
CN
China
Prior art keywords
compound
synthesis
methyl
optionally substituted
cancer
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
CN201710012668.1A
Other languages
Chinese (zh)
Other versions
CN106831614A (en
Inventor
刘刚
马瑶
王素华
杨景舒
刘子节
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.)
Tsinghua University
Original Assignee
Tsinghua University
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 Tsinghua University filed Critical Tsinghua University
Priority to CN201710012668.1A priority Critical patent/CN106831614B/en
Publication of CN106831614A publication Critical patent/CN106831614A/en
Application granted granted Critical
Publication of CN106831614B publication Critical patent/CN106831614B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D243/00Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms
    • C07D243/06Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4
    • C07D243/10Heterocyclic compounds containing seven-membered rings having two nitrogen atoms as the only ring hetero atoms having the nitrogen atoms in positions 1 and 4 condensed with carbocyclic rings or ring systems
    • C07D243/141,4-Benzodiazepines; Hydrogenated 1,4-benzodiazepines
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention discloses a substituted benzodiazacyclo compound, a preparation method and application thereof, and further relates to a pharmaceutical composition and application of the compound. The substituted benzodiazepine compound can selectively antagonize or synergically agonize the activation of NOD1/2 signaling pathway to prevent or treat immunoinflammatory diseases or tumors, has the stability of a polypeptide enzyme, and cannot be degraded by the polypeptide enzyme in a body.

Description

Substituted benzodiazacyclo compound and its preparation method and use
Technical Field
The invention relates to the field of medicines, in particular to a substituted benzodiazepine ring compound and a preparation method and application thereof.
Background
The NOD1/2 protein is a pattern recognition receptor in cytoplasm, recognizes bacterial cell walls entering cells and degradation products thereof, and mediates NF-kB and Mitogen Activated Protein Kinase (MAPK) signaling pathways. The NOD1 receptor is also induced to produce type 1 interferon via interferon-stimulated gene factor 3(ISGF3) signaling pathway, and NOD2 receptor recognizes ssRNA and viral genomic ssRNA, activating interferon regulatory factor 3 via mitochondrial anti-viral signal protein (MAV) signaling pathway (IRF 3). They are involved in the immune response of pathogenic microorganisms such as antibacterial, antiviral and antiparasitic agents.
However, the prevention or treatment of immunoinflammatory disorders or tumors by selectively antagonizing the activation of the NOD1/2 signaling pathway has yet to be extensively studied.
Disclosure of Invention
The present invention is directed to solving, at least to some extent, one of the technical problems in the related art. Therefore, the invention aims to provide a substituted benzodiazacycle compound and a preparation method and application thereof.
The present invention is based on the discovery by the inventors of the following facts and problems:
n-acetyl Muramyl Dipeptide (MDP) is a minimum structural glycopeptide fragment with immunoadjuvant activity, is firstly found in the cell wall of mycobacteria, can effectively increase the humoral immune response and cellular immune response of the organism to antigens by nonspecifically activating human immune cells (such as macrophages, bone marrow mononuclear cells, neutrophils, T and B lymphocytes and the like), and can play a role in nonspecific infection resistance (pneumoconiosis, escherichia coli, pseudomonas aeruginosa, mononucleosis listeria monocytogenes, candida albicans and the like), tumor resistance (fibrosarcoma, hepatoma and the like), immune regulation and the like.
Figure GDA0001986101440000011
Chemical Structure of MDP of formula (a)
For example, Murabutide as a conjugate of MDP molecules and fatty alkanes can improve the nonspecific antibacterial and antiviral abilities of host immune systems, induce the body to release CSF, regulate the functions of macrophages and the like, Mifamurtide (MTP-PE) as a linker uses alanine to link MDP and phosphatidyl aminoethanol to form a conjugate, is currently marketed for adjuvant therapy of osteosarcoma, and has the main side effects of fever and shivering, MDP-L ys (L) is a conjugate obtained by connecting MDP molecules to fatty alkanes through lysine (L ys), can improve the levels of various cytokines (such as CSF, I L-1, I L-6 and TNF- α) in vivo, has stronger anti-infection and anti-tumor activities, is used as an enhancer on the radiotherapy and chemotherapy, but has the advantages of MDP molecules, such as the discovery of the commercial MDP structural analogs, the discovery of MDP structural analogs with fewer side effects, the discovery of MDP molecules, the replacement of MDP molecules into novel lipophilic derivatives, the MDP-acyl aminoethanol-amino acid derivatives, and the like, and has the advantages of MDP-acyl amino acid residues, and the MDP-alanine-amino acid-lysine-amino acid-lysine-amino-lysine-amino-lysine-amino-5-arginine-for the same, and-arginine-for the same, and-arginine.
Figure GDA0001986101440000021
Chemical structure of Mifamotide of formula (b)
Figure GDA0001986101440000022
Chemical Structure of MDP-C of formula (C)
Figure GDA0001986101440000023
Chemical Structure of MDP derivative of formula (d)
However, the inventors have found that the results of the above invention are based on a linear structure, with the possibility of instability of the polypeptide enzyme.
In view of the above, the inventors propose a substituted benzodiazepine compound, and a preparation method and use thereof. The inventor realizes more excellent therapeutic value by simulating or isosteering the biological activity or pharmacological activity of the compound and improving the enzyme stability. More unexpectedly, the inventor finds that the compound selectively antagonizes the activation of NOD1/2 signaling pathway to achieve the effect of preventing or treating the immunoinflammatory diseases or tumors through a large number of experimental results. Clearly, as the biological function of NOD1/2 is not fully understood, the therapeutic significance of NOD1/2 selective antagonists of the present invention will not be limited to immunoinflammatory diseases or tumors.
In a first aspect, the invention features a compound. According to an embodiment of the present invention, it is a compound represented by formula (I) or a stereoisomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof,
Figure GDA0001986101440000031
wherein R is1Is H, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl or optionally substituted heterocycloalkyl;
R2is OR6Or NR7R8
R3Is optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl,
Figure GDA0001986101440000032
R4、R5Each independently is H or alkyl;
R6is H or alkyl;
R7、R8each independently of the other being H, optionally substituted alkyl or with R2The nitrogen atom in (a) forms an optionally substituted heterocycloalkyl group;
R9is optionally substituted aryl, optionally substituted heteroarylOptionally substituted alkyl or optionally substituted alkenyl;
R10is H, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl or optionally substituted heterocycloalkyl;
R11is optionally substituted styryl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl or optionally substituted heterocycloalkyl;
y is N, O, S or alkyl;
x is N, O, S or alkyl;
X1、X2、X3、X4each independently is H, halogen, methyl, hydroxy, amino, cyano, alkoxy, trifluoromethyl or mercapto, and alkyl derivatives of these substituents;
n is 1,2 or 3.
In some embodiments of the invention, R1Is optionally substituted C1-6Alkyl, optionally substituted C1-6Heteroalkyl, optionally substituted C4-6Cycloalkyl or forming optionally substituted C with X4-6Heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl or
Figure GDA0001986101440000041
R2Is OR6Or NR7R8
R3Is optionally substituted C1-6Alkyl, optionally substituted C1-6Heteroalkyl, optionally substituted C4-6Cycloalkyl or optionally substituted C4-6A heterocycloalkyl group,
Figure GDA0001986101440000042
R4、R5Each independently is H or C1-6An alkyl group;
R6is H or C1-6An alkyl group;
R7、R8are each independently H, C1-20Alkyl or with R2Wherein the nitrogen atom forms an optionally substituted C4-6A heterocycloalkyl group;
R9is optionally substituted aryl, optionally substituted heteroaryl, optionally substituted C1-6Alkyl or optionally substituted C1-6An alkenyl group;
R10is H, optionally substituted C1-3 alkyl, optionally substituted C1-6Heteroalkyl or optionally substituted C4-6A heterocycloalkyl group; r11Is optionally substituted styryl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl;
y is N, O, S or alkyl;
x is N, O, S or alkyl;
X1、X2、X3、X4each independently is H, halogen, methyl, hydroxy, amino, cyano, alkoxy, trifluoromethyl or mercapto or an alkyl derivative of these substituents;
n is 1,2 or 3.
In some embodiments of the invention, R1Is optionally substituted C1-6Alkyl, optionally substituted C1-6Heteroalkyl, optionally substituted C4-6Cycloalkyl or forming optionally substituted C with X4-6Heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl or
Figure GDA0001986101440000043
R2Is OR6Or NR7R8
R3Is optionally substituted C1-6Alkyl, optionally substituted C1-6Heteroalkyl, optionally substituted C4-6Cycloalkyl or optionally substituted C4-6A heterocycloalkyl group,
Figure GDA0001986101440000051
R4、R5Each independently is H or C1-3An alkyl group;
R6is H or C1-3An alkyl group;
R7、R8are each independently H, C1-18Alkyl or with R2Wherein the nitrogen atom forms an optionally substituted C4-6A heterocycloalkyl group;
R9is optionally substituted phenyl, optionally substituted naphthyl, optionally substituted thienyl, optionally substituted furyl, optionally substituted C1-3Alkyl or optionally substituted C1-3An alkenyl group;
R10is H, optionally substituted C1-3 alkyl, optionally substituted C1-6Heteroalkyl or optionally substituted C4-6A heterocycloalkyl group;
R11is optionally substituted styryl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl or optionally substituted heterocycloalkyl;
y is N, O or C1-3An alkyl group;
x is N, O;
X1、X2、X3、X4each independently is H, methyl or halogen;
n is 1 or 2.
In some embodiments of the invention, R1Is methyl, naphthyl, dibutylamino,
Figure GDA0001986101440000052
R2Is OR6Or NR7R8
R3Is composed of
Figure GDA0001986101440000053
R4、R5Each independently is H or methyl;
R6is H or methyl;
R7、R8each independently is H, methyl, ethyl, n-propyl, cyclopropyl, n-butyl, tert-butyl, n-dodecyl, n-hexadecyl, n-octadecyl or with R2Nitrogen atom formation in
Figure GDA0001986101440000054
Figure GDA0001986101440000055
Or bridged or spiro rings;
R9is composed of
Figure GDA0001986101440000056
Figure GDA0001986101440000061
Or a vinyl group;
R10is H,
Figure GDA0001986101440000062
R11Is methyl, benzyl, p-trifluorophenyl alkenyl, 3,4, 5-trimethoxyphenyl, vinyl, 3-methyl-2 butenyl, p-trifluoromethylphenyl, naphthyl, 3, 5-bistrifluoromethylphenyl, cyclopropyl, p-cyanophenyl,
Figure GDA0001986101440000063
Figure GDA0001986101440000064
Y is methylene or O;
X1、X2、X3、X4each independently is H, methyl or Cl;
n is 1 or 2.
In some embodiments of the invention, the compounds proposed by the invention have the structure of one of the following:
Figure GDA0001986101440000065
Figure GDA0001986101440000071
Figure GDA0001986101440000081
Figure GDA0001986101440000091
Figure GDA0001986101440000101
Figure GDA0001986101440000111
Figure GDA0001986101440000121
Figure GDA0001986101440000131
Figure GDA0001986101440000141
Figure GDA0001986101440000151
Figure GDA0001986101440000161
in a second aspect of the invention, a pharmaceutical composition is provided. According to an embodiment of the invention, the pharmaceutical composition comprises the above compound. The inventor finds that the pharmaceutical composition can effectively achieve the effect of preventing or treating the immunoinflammatory diseases or tumors by selectively antagonizing or synergistically agonizing the activation of NOD1/2 signaling pathway, has the stability of the polypeptidase, and cannot be degraded by the polypeptidase in the body.
In some embodiments of the invention, the pharmaceutical composition further comprises a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle, or combination thereof.
In some embodiments of the invention, the pharmaceutical composition further comprises an additional agent for preventing or treating an immunoinflammatory disorder or tumor.
In some embodiments of the present invention, the other drugs for preventing or treating the immunoinflammatory disease or tumor include, but are not limited to, melphalan (melphalan), cyclophosphamide (cyclophosphamide), ifosfamide (ifosfamide), busulfan (busufan), carmustine (carmustine), amoxicillin (luminfine), ciclopirox (bleomycin), ciclopirox (ciclopirox), ciclopirox-or (ciclopirox), ciclopirox (ciclopirox), ciclovir (ciclopirox), ciclopirox (e), ciclopirox (e), ciclopirox (ciclopirox), ciclopiroxan), ciclopirox (ciclopirox), ciclopiroxan (ciclopiroxan), ciclopirox (ciclopirox), ciclopirox (e), ciclopirox (ciclopirox), ciclopirox (e), ciclopirox (e), ciclopirox (ciclopirox), ciclopirox (e), ciclopiroxan), ciclopirox (ciclopirox), ciclopirox (e), ciclopirox (e), ciclopirox (e), ciclopirox (ciclopirox), ciclopirox (e), ciclopirox (ciclopirox), ciclopirox (e), ciclopirox (ciclopirox), ciclopiroxan (e), ciclopirox (or, ciclopirox (ciclopirox), ciclopiroxan (ciclopirox), ciclopirox (e), ciclopirox), ciclopiroxan (ib (ciclopirox), ciclopirox (e), ciclopirox (ciclopirox), ciclopiroxan (e), ciclopirox (ciclopiroxan (e), ciclopirox (e), ciclopirox (e), ciclopirox (or, ciclopirox (e (ciclopirox (e), ciclopirox (ciclopirox), ciclopirox (e), ciclopirox-ribbon (e), ciclopirox (e), ciclopirox (or, ciclopirox (e), ciclopirox (e), ciclopirox (e), ciclopirox (e), ciclopirox (e), ciclopirox (ciclopirox-insulin (or, ciclopirox (tablet (ciclopirox (tablet (ciclopirox (e), ciclopirox (e), ciclopirox (tablet (or, ciclopirox (tablet (e), ciclopirox (e), ciclopirox (or, ciclopirox (tablet (ciclopirox), ciclopirox (or, ciclopirox (tablet (e), ciclopirox (peripheral, ciclopirox (e), ciclopirox (or, ciclopirox (tablet (e), ciclopiroxan), ciclopirox (tablet (or, ciclopirox (e), ciclopiroxan), or, ciclopirox), ciclopiroxan), or, ciclopirox-ribbon (peripheral (.
In a third aspect of the invention, the invention proposes the use of a compound as described above or a pharmaceutical composition as described above for the preparation of a medicament for the prevention or treatment of an immunoinflammatory disorder or a tumour. The inventor finds that the medicament can effectively achieve the effect of preventing or treating the immunoinflammatory diseases or tumors by selectively antagonizing or synergistically agonizing the activation of NOD1/2 signaling pathway, has the stability of the polypeptide enzyme, and is not degraded by the polypeptide enzyme in the body.
In some embodiments of the invention, the medicament is for preventing or treating rheumatoid arthritis, sjogren's syndrome, wegener's granulomatosis, behcet's disease, sarcoidosis, takayasu's arteritis, reactive arthritis, osteoarthritis, aids, allergic diseases, rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus or multiple sclerosis, colon cancer, rectal cancer, gastric adenocarcinoma, pancreatic cancer, bladder cancer, gallbladder cancer, breast cancer, renal cell carcinoma, liver cancer, hepatocellular carcinoma, lung cancer, skin cancer, melanoma, thyroid cancer, osteosarcoma, soft tissue sarcoma, head and neck cancer, central nervous system tumor, glioma, glioblastoma, ovarian cancer, uterine cancer, endometrial cancer, prostate cancer, acute myeloid leukemia or acute lymphocytic leukemia or metastatic cancers thereof, Polycythemia vera, primary hemo-allergic arthritis, osteoarthritis, AIDS, allergic diseases, rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus or multiple sclerosis.
In a fourth aspect, the present invention provides the use of a compound as described above or a pharmaceutical composition as described above in the manufacture of a medicament for selectively antagonizing or agonizing the activation of the NOD1/2 signalling pathway.
In addition, in a fifth aspect of the invention, methods of preparation, isolation, purification and characterization of the above compounds are presented.
Additional aspects and advantages of the invention will be set forth in part in the description which follows and, in part, will be obvious from the description, or may be learned by practice of the invention.
Drawings
The above and/or additional aspects and advantages of the present invention will become apparent and readily appreciated from the following description of the embodiments, taken in conjunction with the accompanying drawings of which:
FIG. 1 shows the results of experiments on the antagonism of NOD1 and NOD2 by Compound 13 according to the examples of the present invention,
wherein A is the result of antagonism of hNOD1/2 by compound 13,
b is the cytotoxicity result of compound 13;
FIG. 2 is a graph showing the results of experiments on the antagonism of C12-ie-DAP-activated NOD1 pathway by Compound 13 according to the example of the present invention on macrophages derived from human peripheral blood,
wherein A is the result of the effect of compound 13 on C12-ie-DAP activation of NOD1 to cause the transcription of pro-inflammatory cytokine I L-6,
b is the result of the effect of compound 13 on C12-ie-DAP activation of NOD1 to cause the transcription of the pro-inflammatory cytokine TNF-alpha;
FIG. 3 is a graph showing the results of experiments on the antagonism of MDP-activated NOD2 pathway by Compound 13 on macrophages derived from human peripheral blood in accordance with the present invention,
wherein A is the result of the effect of compound 13 on MDP activation of NOD2 to cause transcription of pro-inflammatory cytokine I L-6,
b is the result of the effect of compound 13 on MDP-activated NOD 2-induced transcription of the pro-inflammatory cytokine TNF-alpha;
FIG. 4 shows the results of the effect of Compound 13 on the expression of proteins involved in the NOD1 and NOD2 signaling pathways activated by C12-ie-DAP and MDP according to the present invention,
wherein A is the result of the experiment on the expression change of related protein of the compound 13 after C12-ie-DAP stimulation activates NOD1 pathway,
b is the result of experiment of expression change of related protein of compound 13 after NOD2 pathway is stimulated and activated by MDP;
FIG. 5 shows the effect of compound 13 on the growth of tumors in mouse L ewis lung cancer model (LL C),
wherein A is the picture of the tumor at the end of the experiment,
b is a tumor volume change curve chart,
c is a graph of the change result of tumor weight,
d is a tumor weight inhibition rate result graph;
FIG. 6 shows the results of selective antagonism of NOD1/2 by Compound 13 according to the examples of the present invention.
Detailed Description
The following describes embodiments of the present invention in detail. The following examples are illustrative only and are not to be construed as limiting the invention. The examples, where specific techniques or conditions are not indicated, are to be construed according to the techniques or conditions described in the literature in the art or according to the product specifications. The reagents or instruments used are not indicated by the manufacturer, and are all conventional products commercially available.
Definitions and general terms
Reference will now be made in detail to certain embodiments of the invention, examples of which are illustrated by the accompanying structural and chemical formulas. The invention is intended to cover alternatives, modifications and equivalents, which may be included within the scope of the invention as defined by the appended claims. Those skilled in the art will recognize that many methods and materials similar or equivalent to those described herein can be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated documents, patents, and similar materials differ or contradict this application (including but not limited to defined terminology, application of terminology, described techniques, and the like), this application controls.
It will be further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination.
The articles "a," "an," and "the" as used herein are intended to include "at least one" or "one or more" unless otherwise indicated or clearly contradicted by context. Thus, as used herein, the articles refer to articles of one or more than one (i.e., at least one) object. For example, "a component" refers to one or more components, i.e., there may be more than one component contemplated for use or use in embodiments of the described embodiments.
The term "patient" as used herein refers to humans (including adults and children) or other animals. In some embodiments, "patient" refers to a human.
The term "comprising" is open-ended, i.e. includes the elements indicated in the present invention, but does not exclude other elements.
The term "treating" any disease or condition, as used herein, means all that can slow, halt, arrest, control or halt the progression of the disease or condition, but does not necessarily mean that all the symptoms of the disease or condition have disappeared, and also includes prophylactic treatment of the symptoms, particularly in patients susceptible to such disease or disorder. In some of these embodiments, refers to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.
The term "therapeutically effective amount" or "therapeutically effective dose" as used herein refers to an amount of a compound of the invention that is capable of eliciting a biological or medical response (e.g., reducing or inhibiting enzyme or protein activity, or ameliorating symptoms, alleviating a disorder, slowing or delaying the progression of a disease, or preventing a disease, etc.) in a subject.
The terms "administration" and "administering" as used herein shall be understood as providing a compound of the invention or a prodrug of a compound of the invention to a subject in need thereof. It will be appreciated that those skilled in the art will treat patients suffering from neurological and psychiatric disorders at present, or prophylactically treat patients suffering from such disorders, by administering an effective amount of a compound of the invention.
The term "composition" as used herein refers to a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. The meaning of such terms in relation to pharmaceutical compositions includes products comprising the active ingredient(s) and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from mixing, complexation or aggregation of any two or more of the ingredients, or from decomposition of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention include any composition prepared by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
The compounds of the invention may be optionally substituted with one or more substituents, as described herein, in compounds of the general formula above, or as specifically exemplified, sub-classes, and classes of compounds encompassed by the invention.
In general, the term "substituted" means that one or more hydrogen atoms in a given structure are replaced with a particular substituent. Unless otherwise indicated, a substituted group may have one substituent substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, then the substituents may be substituted, identically or differently, at each substitutable position.
In addition, it should be noted that, unless otherwise explicitly indicated, the description of the present invention as "independently" is to be understood in a broad sense and may mean that specific items expressed between the same symbols in different groups do not affect each other, or that specific items expressed between the same symbols in the same groups do not affect each other.
In the various parts of this specification, substituents of the disclosed compounds are disclosed in terms of group type or range. It is specifically intended that the invention includes each and every independent subcombination of the various members of these groups and ranges. For example, the term "C1-6Alkyl "means in particular independently disclosed methyl, ethyl, C3Alkyl radical, C4Alkyl radical, C5Alkyl and C6An alkyl group.
In each of the parts of the invention, linking substituents are described. Where the structure clearly requires a linking group, the markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the markush group definition for the variable recites "alkyl" or "aryl," it is understood that the "alkyl" or "aryl" represents an attached alkylene group or arylene group, respectively.
The term "alkyl" or "alkyl group" as used herein, denotes a saturated straight or branched chain monovalent hydrocarbon radical, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. Unless otherwise specified, alkyl groups contain 1-20 carbon atoms. In one embodiment, the alkyl group contains 1 to 12 carbon atoms; in another embodiment, the alkyl group contains 3 to 12 carbon atoms; in another embodiment, the alkyl group contains 1 to 6 carbon atoms; in yet another embodiment, the alkyl group contains 1 to 4 carbon atoms.
Examples of alkyl groups include, but are not limited to, methyl (Me, -CH)3) Ethyl group (Et, -CH)2CH3) N-propyl (n-Pr, -CH)2CH2CH3) Isopropyl group (i-Pr, -CH (CH)3)2) N-butyl (n-Bu, -CH)2CH2CH2CH3) Isobutyl (i-Bu, -CH)2CH(CH3)2) Sec-butyl (s-Bu, -CH (CH)3)CH2CH3) Tert-butyl (t-Bu, -C (CH)3)3) N-pentyl (-CH)2CH2CH2CH2CH3) 2-pentyl (-CH (CH)3)CH2CH2CH3) 3-pentyl (-CH (CH)2CH3)2) 2-methyl-2-butyl (-C (CH)3)2CH2CH3) 3-methyl-2-butyl (-CH (CH)3)CH(CH3)2) 3-methyl-1-butyl (-CH)2CH2CH(CH3)2) 2-methyl-1-butyl (-CH)2CH(CH3)CH2CH3) N-hexyl (-CH)2CH2CH2CH2CH2CH3) 2-hexyl (-CH (CH)3)CH2CH2CH2CH3) 3-hexyl (-CH (CH)2CH3)(CH2CH2CH3) 2-methyl-2-pentyl (-C (CH))3)2CH2CH2CH3) 3-methyl-2-pentyl (-CH (CH)3)CH(CH3)CH2CH3) 4-methyl-2-pentyl (-CH (CH)3)CH2CH(CH3)2) 3-methyl-3-pentyl (-C (CH)3)(CH2CH3)2) 2-methyl-3-pentyl (-CH (CH)2CH3)CH(CH3)2)2, 3-dimethyl-2-butyl (-C (CH)3)2CH(CH3)2)3, 3-dimethyl-2-butyl (-CH (CH)3)C(CH3)3) N-heptyl, n-octyl, and the like.
The term "alkyl derivative of a substituent" means that the substituent may be substituted by an alkyl derivative, for example, an alkyl derivative of halogen may be an alkyl substituted by halogen and an alkyl derivative of cyano may be an alkyl substituted by cyano, wherein the term "alkyl" has the definition as described herein.
The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon radical having at least one carbon-carbon sp2Double bonds, which include the positioning of "cis" and "trans", or the positioning of "E" and "Z". Wherein said alkenyl group may be optionally substituted with one or more substituents as described herein. At one endIn embodiments, the alkenyl group contains 2 to 12 carbon atoms; in another embodiment, the alkenyl group contains 3 to 12 carbon atoms; in another embodiment, the alkenyl group contains 2 to 6 carbon atoms; in yet another embodiment, the alkenyl group contains 2 to 4 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (-CH ═ CH)2) Allyl (-CH)2CH=CH2) And so on.
The term "cycloalkyl" denotes a monovalent or polyvalent saturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 ring carbon atoms. In one embodiment, cycloalkyl groups contain from 7 to 12 ring carbon atoms; in yet another embodiment, cycloalkyl contains 3 to 8 ring carbon atoms; in yet another embodiment, cycloalkyl contains 3 to 6 ring carbon atoms. The cycloalkyl groups may be independently unsubstituted or substituted with one or more substituents described herein.
The terms "heterocyclyl" and "heterocycle" are used interchangeably herein and refer to a saturated or partially unsaturated, non-aromatic, monovalent or polyvalent, monocyclic, bicyclic, or tricyclic ring containing from 3 to 12 ring atoms, wherein at least one ring atom is selected from nitrogen, sulfur, or oxygen. Unless otherwise specified, heterocyclyl may be carbon-or nitrogen-based, and-CH2-the group may optionally be replaced by-C (═ O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atom of the ring may optionally be oxidized to an N-oxygen compound. Examples of heterocyclyl groups include, but are not limited to: oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1, 3-dioxolanyl, dithiocyclopentyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, dithianyl, thiaxanyl, homopiperazinyl, homopiperidinyl, diazepanyl, oxepanyl, thietanyl, oxazepanyl, and pyrazolinyl
Figure GDA0001986101440000221
Radical diaza
Figure GDA0001986101440000222
Radical, sulfur nitrogen hetero
Figure GDA0001986101440000223
Radicals, indolinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 1, 3-benzodioxolyl, 2-oxa-5-azabicyclo [2.2.1]Hept-5-yl. In heterocyclic radicals of-CH2Examples of-groups substituted by-C (═ O) -include, but are not limited to, 2-oxopyrrolidinyl, oxo-1, 3-thiazolidinyl, 2-piperidinonyl, 3, 5-dioxopiperidinyl and pyrimidinedione. Examples of the sulfur atom in the heterocyclic group being oxidized include, but are not limited to, sulfolane group, 1-dioxothiomorpholinyl group. Wherein said heterocyclyl group may be optionally substituted by one or more substituents as described herein.
The term "heterocycloalkyl" refers to a monovalent or polyvalent saturated monocyclic, bicyclic, or tricyclic ring system containing 3 to 12 ring atoms, wherein at least one ring atom is selected from nitrogen, sulfur, or oxygen atoms. Wherein said heterocycloalkyl group may be optionally substituted with one or more substituents as described herein.
The term "halogen" refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
The term "aryl" denotes a monocyclic, bicyclic or tricyclic carbocyclic ring system containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring is aromatic and has one or more attachment points to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring". In one embodiment, aryl is a carbocyclic ring system consisting of 6 to 10 ring atoms and containing at least one aromatic ring therein. Examples of the aryl group may include phenyl, naphthyl and anthracenyl. Wherein the aryl group may independently be optionally substituted with one or more substituents described herein.
The term "heteroaryl" denotes a monocyclic, bicyclic or tricyclic ring containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein at least one ring is aromatic and at least one aromatic ring contains one or more heteroatoms and has one or more attachment points to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic ring" or "heteroaromatic compound". Wherein said heteroaryl group is optionally substituted with one or more substituents as described herein. In one embodiment, heteroaryl is a 5-12 atom heteroaryl comprising 1,2,3, or 4 heteroatoms independently selected from O, S and N; in another embodiment, heteroaryl is 5-6 atom consisting of 1,2,3, or 4 heteroatoms independently selected from O, S and N.
Examples of heteroaryl groups include, but are not limited to, 2-furyl, 3-furyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, oxadiazolyl (e.g., 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl), oxadiazolyl (e.g., 1,2,3, 4-oxadiazolyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, isothiazolyl, 2-thiadiazolyl (e.g., 1,3, 4-thiadiazolyl, 1,2, 3-thiadiazolyl, 1,2, 5-thiadiazolyl) Thiatriazolyl (e.g., 1,2,3, 4-thiatriazolyl), tetrazolyl (e.g., 2H-1,2,3, 4-tetrazolyl, 1H-1,2,3, 4-tetrazolyl), triazolyl (e.g., 2H-1,2, 3-triazolyl, 1H-1,2, 4-triazolyl, 4H-1,2, 4-triazolyl), 2-thienyl, 3-thienyl, 1H-pyrazolyl (e.g., 1H-pyrazol-3-yl, 1H-pyrazol-4-yl, 1H-pyrazol-5-yl), 1,2, 3-thiadiazolyl, 1,3, 4-thiadiazolyl, 1,2, 5-thiadiazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl, 4-pyridazinyl), 2-pyrazinyl, triazinyl (e.g., 1,3, 5-triazine), tetrazinyl (e.g., 1,2,4, 5-tetrazine, 1,2,3, 5-tetrazine); the following bicyclic rings are also included, but are in no way limited to these: benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolyl (e.g., 2-quinolyl, 3-quinolyl, 4-quinolyl), isoquinolyl (e.g., 1-isoquinolyl, 3-isoquinolyl, or 4-isoquinolyl), imidazo [1,2-a ] pyridyl, pyrazolo [1,5-a ] pyrimidinyl, imidazo [1,2-b ] pyridazinyl, [1,2,4] triazolo [4,3-b ] pyridazinyl, [1,2,4] triazolo [1,5-a ] pyrimidinyl, [1,2,4] triazolo [1,5-a ] pyridyl, and the like.
The term "prodrug", as used herein, represents a compound that is converted in vivo to a compound of formula (I). 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 prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C)1-C24) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can 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. For a complete discussion of prodrugs, reference may be made to the following: T.Higuchi and V.Stella, Pro-drugs as Novel delivery systems, Vol.14of the A.C.S.Symphosium Series, Edward B.Roche, ed., Bioredeployers in Drug designs, American Pharmaceutical Association and PergammonPress, 1987, J.Rautio et al, Prodrug: Design and Clinical Applications, Nature review delivery, 2008,7,255 and 270, S.J.Hecker et al, Prodrugs of pharmaceuticals and phosphates, Journal of chemical Chemistry,2008,51,2328 and 2345.
"metabolite" refers to the product of a particular compound or salt thereof obtained by metabolism in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assay methods as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a sufficient period of time.
Used in the inventionThe "pharmaceutically acceptable salts" of (a) refer to organic and inorganic salts of the compounds of the invention. Pharmaceutically acceptable salts are well known in the art, as are: berge et al, descriptive acceptable salts in detail in J. pharmaceutical Sciences,1977,66:1-19. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, salts of inorganic acids formed by reaction with amino groups such as hydrochlorides, hydrobromides, phosphates, sulfates, perchlorates, and salts of organic acids such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, or those obtained by other methods described in the literature above, such as ion exchange. Other pharmaceutically acceptable salts include adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, cyclopentylpropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, malates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, palmitates, pamoates, pectinates, persulfates, 3-phenylpropionates, picrates, pivalates, propionates, stearates, thiocyanates, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained with appropriate bases include alkali metals, alkaline earth metals, ammonium and N+(C1-4Alkyl radical)4A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C1-8Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules that is water.
When the solvent is water, the term "hydrate" may be used. In some embodiments, a molecule of a compound of the present invention may be associated with a molecule of water, such as a monohydrate; in other embodiments, one molecule of the compound of the present invention may be associated with more than one molecule of water, such as a dihydrate, and in still other embodiments, one molecule of the compound of the present invention may be associated with less than one molecule of water, such as a hemihydrate. It should be noted that the hydrates of the present invention retain the biological effectiveness of the compound in its non-hydrated form.
The term "treating" any disease or condition, as used herein, means all that can slow, halt, arrest, control or halt the progression of the disease or condition, but does not necessarily mean that all the symptoms of the disease or condition have disappeared, and also includes prophylactic treatment of the symptoms, particularly in patients susceptible to such disease or disorder. In some of these embodiments, refers to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one clinical symptom thereof). In other embodiments, "treating" or "treatment" refers to moderating or improving at least one physical parameter, including physical parameters that may not be perceived by the patient. In other embodiments, "treating" or "treatment" refers to modulating the disease or disorder, either physically (e.g., stabilizing a perceptible symptom) or physiologically (e.g., stabilizing a parameter of the body), or both. In other embodiments, "treating" or "treatment" refers to preventing or delaying the onset, occurrence, or worsening of a disease or disorder.
The terms "administration" and "administering" of a compound as used herein shall be understood as providing a compound of the invention or a prodrug of a compound of the invention to a subject in need thereof. It will be appreciated that those skilled in the art will treat patients suffering from neurological and psychiatric disorders at present, or prophylactically treat patients suffering from such disorders, by administering an effective amount of a compound of the invention.
The term "composition" as used herein refers to a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts. The meaning of such terms in relation to pharmaceutical compositions includes products comprising the active ingredient(s) and the inert ingredient(s) that make up the carrier, as well as any product which results, directly or indirectly, from mixing, complexation or aggregation of any two or more of the ingredients, or from decomposition of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention include any composition prepared by admixing a compound of the present invention and a pharmaceutically acceptable carrier.
Description of the Compounds of the invention
The invention discloses a substituted benzodiazacyclic compound, pharmaceutically acceptable salts thereof, a pharmaceutical preparation and a composition thereof, which can be used as an antagonist of NOD1/2 signal transduction pathway and can be used for treating human inflammatory diseases or tumors, such as rheumatoid arthritis, sicca syndrome, Wegener's granulomatosis, Behcet's disease, sarcoidosis, Takayasu's disease, reactive arthritis, osteoarthritis, AIDS, allergic diseases, rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus or multiple sclerosis, colon cancer, rectal cancer, stomach cancer, gastric adenocarcinoma, pancreatic cancer, bladder cancer, gall bladder cancer, breast cancer, kidney cancer, renal cell cancer, liver cancer, hepatocellular carcinoma, lung cancer, skin cancer, melanoma, thyroid cancer, osteosarcoma, soft tissue sarcoma, head and neck cancer, central nervous system tumor, The treatment of glioma, glioblastoma, ovarian cancer, uterine cancer, endometrial cancer, prostate cancer, acute myeloid leukemia or acute lymphocytic leukemia or their metastatic cancers, polycythemia vera, primary hemotopathy, osteoarthritis, AIDS, allergic diseases, rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus or multiple sclerosis has potential uses.
In a first aspect, the invention features a compound. According to an embodiment of the present invention, it is a compound represented by formula (I) or a stereoisomer, nitrogen oxide, solvate, metabolite, pharmaceutically acceptable salt or prodrug thereof,
Figure GDA0001986101440000261
wherein R is1Is H, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl or optionally substituted heterocycloalkyl;
R2is OR6Or NR7R8
R3Is optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl,
Figure GDA0001986101440000262
R4、R5Each independently is H or alkyl;
R6is H or alkyl;
R7、R8each independently of the other being H, optionally substituted alkyl or with R2The nitrogen atom in (a) forms an optionally substituted heterocycloalkyl group;
R9is optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl or optionally substituted alkenyl;
R10is H, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl or optionally substituted heterocycloalkyl;
R11is optionally substituted styryl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroarylOptionally substituted heteroalkyl, optionally substituted cycloalkyl or optionally substituted heterocycloalkyl;
y is N, O, S or alkyl;
x is N, O, S or alkyl;
X1、X2、X3、X4each independently is H, halogen, methyl, hydroxy, amino, cyano, alkoxy, trifluoromethyl or mercapto, and alkyl derivatives of these substituents;
n is 1,2 or 3.
In some embodiments of the invention, R1Is optionally substituted C1-6Alkyl, optionally substituted C1-6Heteroalkyl, optionally substituted C4-6Cycloalkyl or forming optionally substituted C with X4-6Heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl or
Figure GDA0001986101440000263
R2Is OR6Or NR7R8
R3Is optionally substituted C1-6Alkyl, optionally substituted C1-6Heteroalkyl, optionally substituted C4-6Cycloalkyl or optionally substituted C4-6A heterocycloalkyl group,
Figure GDA0001986101440000271
R4、R5Each independently is H or C1-6An alkyl group;
R6is H or C1-6An alkyl group;
R7、R8are each independently H, C1-20Alkyl or with R2Wherein the nitrogen atom forms an optionally substituted C4-6A heterocycloalkyl group;
R9is optionally substituted aryl, optionally substituted heteroaryl, optionally substituted C1-6Alkyl or optionally substituted C1-6An alkenyl group;
R10is H, optionally substituted C1-3 alkyl, orOptionally substituted C1-6Heteroalkyl or optionally substituted C4-6A heterocycloalkyl group; r11Is optionally substituted styryl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl;
y is N, O, S or alkyl;
x is N, O, S or alkyl;
X1、X2、X3、X4each independently is H, halogen, methyl, hydroxy, amino, cyano, alkoxy, trifluoromethyl or mercapto or an alkyl derivative of these substituents;
n is 1,2 or 3.
In some embodiments of the invention, R1Is optionally substituted C1-6Alkyl, optionally substituted C1-6Heteroalkyl, optionally substituted C4-6Cycloalkyl or forming optionally substituted C with X4-6Heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl or
Figure GDA0001986101440000272
R2Is OR6Or NR7R8
R3Is optionally substituted C1-6Alkyl, optionally substituted C1-6Heteroalkyl, optionally substituted C4-6Cycloalkyl or optionally substituted C4-6A heterocycloalkyl group,
Figure GDA0001986101440000273
R4、R5Each independently is H or C1-3An alkyl group;
R6is H or C1-3An alkyl group;
R7、R8are each independently H, C1-18Alkyl or with R2Wherein the nitrogen atom forms an optionally substituted C4-6A heterocycloalkyl group;
R9is optionally substituted phenyl, optionally substituted naphthyl, optionally substituted thienyl, optionally substituted furyl, optionally substituted C1-3Alkyl or optionally substituted C1-3An alkenyl group;
R10is H, optionally substituted C1-3 alkyl, optionally substituted C1-6Heteroalkyl or optionally substituted C4-6A heterocycloalkyl group;
R11is optionally substituted styryl, optionally substituted alkenyl, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted cycloalkyl or optionally substituted heterocycloalkyl;
y is N, O or C1-3An alkyl group;
x is N, O;
X1、X2、X3、X4each independently is H, methyl or halogen;
n is 1 or 2.
In some embodiments of the invention, R1Is methyl, naphthyl, dibutylamino,
Figure GDA0001986101440000281
R2Is OR6Or NR7R8
R3Is composed of
Figure GDA0001986101440000282
R4、R5Each independently is H or methyl;
R6is H or methyl;
R7、R8each independently is H, methyl, ethyl, n-propyl, cyclopropyl, n-butyl, tert-butyl, n-dodecyl, n-hexadecyl, n-octadecyl or with R2Nitrogen atom formation in
Figure GDA0001986101440000283
Figure GDA0001986101440000284
Or bridged or spiro rings;
R9is composed of
Figure GDA0001986101440000285
Figure GDA0001986101440000286
Or a vinyl group;
R10is H,
Figure GDA0001986101440000287
R11Is methyl, benzyl, p-trifluorophenyl alkenyl, 3,4, 5-trimethoxyphenyl, vinyl, 3-methyl-2 butenyl, p-trifluoromethylphenyl, naphthyl, 3, 5-bistrifluoromethylphenyl, cyclopropyl, p-cyanophenyl,
Figure GDA0001986101440000288
Figure GDA0001986101440000289
Y is methylene or O;
X1、X2、X3、X4each independently is H, methyl or Cl;
n is 1 or 2.
In some embodiments of the invention, the compounds proposed by the invention have the structure of one of the following:
Figure GDA0001986101440000291
Figure GDA0001986101440000301
Figure GDA0001986101440000311
Figure GDA0001986101440000321
Figure GDA0001986101440000331
Figure GDA0001986101440000341
Figure GDA0001986101440000351
Figure GDA0001986101440000361
Figure GDA0001986101440000371
Figure GDA0001986101440000381
Figure GDA0001986101440000391
the inventor finds that the compound can effectively achieve the effect of preventing or treating the immunoinflammatory diseases or tumors by selectively antagonizing or synergistically agonizing the activation of NOD1/2 signaling pathway, has the stability of the polypeptide enzyme, and cannot be degraded by the polypeptide enzyme in the body.
It should be noted that the phrase "selectively antagonizes activation of the NOD1/2 signaling pathway" as used herein means that the compounds of the present invention are capable of antagonizing activation of the NOD1/2 signaling pathway without affecting other potential receptors in the cell.
It should be noted that the term "stability of the polypeptide enzyme" as used above refers to the ability of the compound not to be degraded by the polypeptide enzyme in the body.
Stereoisomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs of the compounds of formula (I) are included within the scope of the present invention unless otherwise indicated.
The compounds of the present disclosure may contain asymmetric or chiral centers and thus may exist in different stereoisomeric forms. The present invention contemplates that all stereoisomeric forms of the compounds of formula (I), including but not limited to diastereomers, enantiomers, atropisomers and geometric (or conformational) isomers, and mixtures thereof, such as racemic mixtures, are integral to the invention.
In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not specified, then all stereoisomers of that structure are contemplated as within this invention and are included as disclosed compounds in this invention. When stereochemistry is indicated by a solid wedge (solid wedge) or dashed line representing a particular configuration, then the stereoisomers of the structure are so well-defined and defined.
The compounds of formula (I) may exist in different tautomeric forms and all such tautomers are included within the scope of the invention.
The compounds of formula (I) may be present in the form of salts. In one embodiment, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be compatible chemically and/or toxicologically with the other ingredients comprising the formulation and/or the mammal being treated therewith. In another embodiment, the salts need not be pharmaceutically acceptable salts and may be intermediates useful in the preparation and/or purification of compounds of formula (I) and/or in the isolation of enantiomers of compounds of formula (I).
Pharmaceutically acceptable acid addition salts may be formed from the disclosed compounds of the invention by the action of an inorganic or organic acid, for example, acetate, aspartate, benzoate, benzenesulfonate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlorotheyl salt, citrate, edisylate, fumarate, glucoheptonate, gluconate, glucuronate, hippurate, hydroiodide, isethionate, lactate, lactobionate, lauryl sulfate, malate, maleate, malonate, mandelate, methanesulfonate, methylsulfate, naphthoate, naphthalenesulfonate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/biphosphate/dihydrogen phosphate, phosphate, Polysilonolactates, propionates, stearates, succinates, sulfosalicylates, tartrates, tosylates and trifluoroacetates.
Pharmaceutically acceptable base addition salts may be formed from the disclosed compounds by reaction with an inorganic or organic base.
Inorganic bases from which salts can be derived include, for example, ammonium salts and metals of groups I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts can be derived include primary, secondary and tertiary amines, and substituted amines include naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Some organic amines include, for example, isopropylamine, benzathine (benzathine), choline salts (cholinate), diethanolamine, diethylamine, lysine, meglumine (meglumine), piperazine, and tromethamine.
The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety, by conventional chemical methods. In general, such salts can be prepared by reacting the free acid forms of these compounds with a stoichiometric amount of the appropriate base (e.g., Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate, etc.), or by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are usually carried out in water or an organic solvent or a mixture of both. Generally, where appropriate, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile. In, for example, "Remington's Pharmaceutical Sciences", 20 th edition, Mack Publishing Company, Easton, Pa., (1985); and "handbook of pharmaceutically acceptable salts: properties, Selection and application (Handbook of pharmaceutical salts: Properties, Selection, and Use) ", Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002) may find some additional lists of suitable salts.
In addition, the compounds disclosed herein, including their salts, may also be obtained in the form of their hydrates or in the form of solvents containing them (e.g., ethanol, DMSO, etc.), for their crystallization. The compounds disclosed herein may form solvates with pharmaceutically acceptable solvents (including water), either inherently or by design; thus, the present invention is intended to include both solvated and unsolvated forms of the disclosed compounds.
Any formulae given herein are also intended to represent the non-isotopically enriched forms as well as the isotopically enriched forms of these compounds. Isotopically enriched compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H、3H、11C、13C、14C、15N、17O、18O、18F、31P、32P、35S、36Cl and125I。
in another aspect, the compounds of the invention include isotopically enriched compounds as defined herein, e.g. wherein a radioisotope, e.g. is present3H、14C and18those compounds of F, or in which a non-radioactive isotope is present, e.g.2H and13those of C. The isotopically enriched compounds can be used for metabolic studies (use)14C) Reaction kinetics study (using, for example2H or3H) Detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution determination, or may be used in radiotherapy of a patient.18F-enriched compoundsIs particularly desirable for PET or SPECT studies. Isotopically enriched compounds of formula (I) can be prepared by conventional techniques known to those skilled in the art or by the procedures and examples described in the present specification using a suitable isotopically labelled reagent in place of the original used unlabelled reagent.
In addition, heavier isotopes are, in particular, deuterium (i.e.,2substitution of H or D) may provide certain therapeutic advantages resulting from greater metabolic stability. For example, increased in vivo half-life or decreased dosage requirements or improved therapeutic index. It is to be understood that deuterium in the present invention is to be considered as a substituent of the compound of formula (I). The concentration of such heavier isotopes, particularly deuterium, can be defined by isotopic enrichment factors. The term "isotopic enrichment factor" as used herein refers to the ratio between the isotopic and natural abundance of a given isotope. If a substituent of a compound of the invention is designated as deuterium, the compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation). Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g. D2O, acetone-d6、DMSO-d6Those solvates of (a).
In another aspect, the invention relates to intermediates for the preparation of compounds of formula (I).
In another aspect, the invention relates to methods for the preparation, isolation and purification of compounds of formula (I).
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention. In one embodiment, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle or combination thereof. In another embodiment, the pharmaceutical composition may be in a liquid, solid, semi-solid, gel, or spray dosage form.
Pharmaceutical compositions, formulations and administration of the compounds of the invention
The present invention provides a pharmaceutical composition comprising a compound disclosed herein, for example, as set forth in the examples. According to a specific example of the present invention, the pharmaceutical composition may further comprise a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or combination thereof.
The present invention provides methods of treating, preventing or ameliorating a disease or condition comprising administering a safe and effective amount of a combination comprising a compound of the present disclosure and one or more therapeutically active agents. Wherein the combination comprises one or more other agents for the prophylaxis or treatment of an immunoinflammatory disorder or tumour.
The other drugs for preventing or treating immunoinflammatory diseases or tumors include, but are not limited to, melphalan (melphalan), cyclophosphamide (cyclophosphamide), ifosfamide (ifosfamide), busulfan (busufan), carmustine (carmustine), ciclopirox (bleomycin), streptozotocin (streptozocin), cisplatin (cissplatin), carboplatin (carboplatin), oxaliplatin (oxaliplatin), dacarbazine (dacarbazine), cetuximab (paclitaxel), cetrimide (paclitaxel), cetrimidine (paclitaxel), ciclopirox (ciclopirox-L), ciclopiroxan (ciclopirox-alone), ciclopirox-or (ciclopirox-a), ciclovir (ciclopirox-a), ciclopirox (ciclopirox-dol (ciclopirox), ciclovir (ciclopirox), ciclopirox (ciclopirox-ne), ciclopiroxan (ciclopirox-r), ciclopirox (ciclopiroxan), ciclopiroxan (ciclopirox (ciclopiroxan), ciclopiroxan (ciclopirox-or (ciclopirox-alone, ciclopirox-L), ciclopirox-L (ciclopirox), ciclopirox-L), ciclopirox (ciclopirox-L ), ciclopiroxan), ciclopirox-or (ciclopiroxan), ciclopirox-L (ciclopirox-L, ciclopiroxan), ciclopirox-or (tablet, ciclopirox-L, ciclopirox (tablet, ciclopirox-or tablet, ciclopirox (tablet, ciclopirox-tablet, ciclopirox (tablet, ciclopirox), or tablet, ciclopirox), ciclopirox (tablet, or tablet (tablet, or tablet, and tablet, or tablet (tablet, or tablet of an for example, or tablet, or tablet for example, or tablet for example, or tablet for treating an for example, or tablet for treating immunoinflammatory disease, or injection for treating immunoinflammatory disease, or injection, for treating immunoinflammatory disease, for treating an for treating immunoinflammatory disease, for example, for treating an immune, for treating immune, or injection, for treating an, for treating immunoinflammatory disease, or injection, for treating an, or injection, for treating an immune, or injection, for treating an, or injection, for treating immune, or injection, for treating an, or injection, for treating immunoinflammatory disease, for treating immune, for treating an, for treating immune, or injection, for treating immune, for treating an immune, for treating an, for treating immune, for treating an, for treating immune, for treating an, or injection, for treating an, or for treating an, for treating an immune or treating immune, for treating an, for treating immune, or immune, for treating immune, or treating immune.
The amount of compound in the pharmaceutical compositions disclosed herein is that amount which is effective to detect antagonism of the NOD1/2 receptor in the biological sample or patient. The dosage of the active ingredient in the composition of the present invention may vary, however, the amount of the active ingredient must be such that a suitable dosage form is obtained. The active ingredient may be administered to patients (animals and humans) in need of such treatment at dosages that provide optimal pharmaceutical efficacy. The selected dosage depends on the desired therapeutic effect, on the route of administration and on the duration of the treatment. The dosage will vary from patient to patient depending on the nature and severity of the disease, the weight of the patient, the particular diet of the patient, the concurrent use of drugs, and other factors that will be recognized by those skilled in the art. The dosage range is generally about 0.5mg to 1.0g per patient per day and may be administered in a single dose or in multiple doses. In one embodiment, the dosage range is from about 0.5mg to 500mg per patient per day; from about 0.5mg to 200mg per patient per day in another embodiment; and in yet another embodiment from about 5mg to 50mg per patient per day.
It will also be appreciated that certain compounds of the invention may be present in free form and used in therapy, or if appropriate in the form of a pharmaceutically acceptable derivative thereof. Pharmaceutically acceptable derivatives include pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any additional adduct or derivative that upon administration to a patient in need thereof provides, directly or indirectly, a compound of the present invention or a metabolite or residue thereof.
The medicaments or pharmaceutical compositions disclosed herein may be prepared and packaged in bulk (bulk) form, wherein a safe and effective amount of the compound of formula (I) may be extracted and then administered to a patient in the form of a powder or syrup. Typically, the administration to a patient is at a dosage level of between 0.0001 and 10mg/kg body weight per day to achieve effective antagonism of NOD1/2 receptors. Alternatively, the pharmaceutical compositions disclosed herein can be prepared and packaged in unit dosage forms, wherein each physically discrete unit contains a safe and effective amount of a compound of formula (I). When prepared in unit dosage form, the disclosed pharmaceutical compositions can generally contain, for example, from 0.5mg to 1g, or from 1mg to 700mg, or from 5mg to 100mg of the disclosed compounds.
When the pharmaceutical composition of the invention contains one or more other active ingredients in addition to the compound of the invention, the compound weight ratio of the compound of the invention to the second active ingredient may vary and depends on the effective dose of each ingredient. Generally, an effective dose of each is used. Thus, for example, when a compound of the invention is mixed with another agent, the weight ratio of the compound of the invention to the other agent typically ranges from about 1000:1 to about 1:1000, e.g., from about 200:1 to about 1: 200. Mixtures of the compounds of the invention with other active ingredients are generally also within the above-mentioned ranges, but in each case an effective dose of each active ingredient should be used.
As used herein, "pharmaceutically acceptable excipient" means a pharmaceutically acceptable material, mixture or vehicle, which is compatible with the dosage form or pharmaceutical composition to be administered. Each excipient, when mixed, must be compatible with the other ingredients of the pharmaceutical composition to avoid interactions that would substantially reduce the efficacy of the disclosed compounds and which would result in a pharmaceutical composition that is not pharmaceutically acceptable when administered to a patient. Furthermore, each excipient must be pharmaceutically acceptable, e.g., of sufficiently high purity.
Suitable pharmaceutically acceptable excipients will vary depending on the particular dosage form selected. In addition, pharmaceutically acceptable excipients may be selected for their specific function in the composition. For example, certain pharmaceutically acceptable excipients may be selected to aid in the production of a uniform dosage form. Certain pharmaceutically acceptable excipients may be selected to aid in the production of stable dosage forms. Certain pharmaceutically acceptable excipients may be selected to facilitate carrying or transporting the disclosed compounds from one organ or portion of the body to another organ or portion of the body when administered to a patient. Certain pharmaceutically acceptable excipients may be selected that enhance patient compliance.
Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, taste masking agents, colorants, anti-caking agents, humectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants and buffers. The skilled artisan will recognize that certain pharmaceutically acceptable excipients may provide more than one function, and provide alternative functions, depending on how many such excipients are present in the formulation and those other excipients are present in the formulation.
Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing L approved), and The Handbook of Pharmaceutical Excipients (The American Pharmaceutical Association and The Pharmaceutical Press).
Various carriers for The preparation of incompatible pharmaceutically acceptable compositions and known techniques for their preparation are disclosed in Remington, The Science and Practice of Pharmacy,21st edition,2005, ed.D.B.Troy, &lTtTtranslation = L "&gTtL &lTt/T &gTtippincott Williams & Wilkins, Philadelphia, and Encyclopedia of pharmaceutical Technology, eds.J.Swarbrick and J.C.Boylan,1988-1999, Mardecekker, New York, The contents of each of which are incorporated herein by reference.
The pharmaceutical compositions disclosed herein are prepared using techniques and methods known to those skilled in the art. Some commonly used methods in the art are described in Remington's Pharmaceutical Sciences (Mack publishing company).
Thus, in another aspect, the invention relates to a process for preparing a pharmaceutical composition comprising a compound of the present disclosure and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or combination thereof, which process comprises admixing the ingredients. Pharmaceutical compositions comprising the disclosed compounds may be prepared by mixing, for example, at ambient temperature and atmospheric pressure.
The compounds disclosed herein are generally formulated in a dosage form suitable for administration to a patient by a desired route. For example, dosage forms include those suitable for the following routes of administration: (1) oral administration, such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets and cachets; (2) parenteral administration, such as sterile solutions, suspensions, and reconstituted powders; (3) transdermal administration, such as transdermal patches; (4) rectal administration, e.g., suppositories; (5) inhalation, such as aerosols, solutions, and dry powders; and (6) topical administration, such as creams, ointments, lotions, solutions, pastes, sprays, foams and gels.
In one embodiment, the compounds disclosed herein may be formulated in oral dosage forms. In another embodiment, the compounds disclosed herein may be formulated in an inhalation dosage form. In another embodiment, the compounds disclosed herein can be formulated for nasal administration. In yet another embodiment, the compounds disclosed herein can be formulated for transdermal administration. In yet another embodiment, the compounds disclosed herein may be formulated for topical administration.
The pharmaceutical compositions provided by the present invention may be provided as compressed tablets, milled tablets, chewable lozenges, fast-dissolving tablets, double-compressed tablets, or enteric-coated, sugar-coated or film-coated tablets. Enteric coated tablets are compressed tablets coated with a substance that is resistant to the action of gastric acid but dissolves or disintegrates in the intestine, thereby preventing the active ingredient from contacting the acidic environment of the stomach. Enteric coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalate. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which can help to mask unpleasant tastes or odors and prevent oxidation of the tablet. Film-coated tablets are compressed tablets covered with a thin layer or film of a water-soluble substance. Film coatings include, but are not limited to, hydroxyethyl cellulose, sodium carboxymethyl cellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coatings are endowed with the same general characteristics as sugar coatings. A tabletted tablet is a compressed tablet prepared over more than one compression cycle, including a multi-layer tablet, and a press-coated or dry-coated tablet.
Tablet dosage forms may be prepared from the active ingredient in powder, crystalline or granular form, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled release polymers, lubricants, diluents and/or colorants. Flavoring and sweetening agents are particularly useful in forming chewable tablets and lozenges.
The pharmaceutical composition provided by the present invention may be provided in soft or hard capsules, which may be prepared from gelatin, methylcellulose, starch or calcium alginate. The hard gelatin capsules, also known as Dry Fill Capsules (DFC), consist of two segments, one inserted into the other, thus completely encapsulating the active ingredient. Soft Elastic Capsules (SEC) are soft, spherical shells, such as gelatin shells, which are plasticized by the addition of glycerol, sorbitol or similar polyols. The soft gelatin shell may contain a preservative to prevent microbial growth. Suitable preservatives are those as described herein, including methyl and propyl parabens, and sorbic acid. The liquid, semi-solid and solid dosage forms provided by the present invention may be encapsulated in a capsule. Suitable liquid and semi-solid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils or triglycerides. Capsules containing such solutions may be as described in U.S. patent nos.4,328,245; 4,409,239 and 4,410,545. The capsules may also be coated as known to those skilled in the art to improve or maintain dissolution of the active ingredient.
The pharmaceutical compositions provided herein may be provided in liquid and semi-solid dosage forms, including emulsions, solutions, suspensions, elixirs and syrups. Emulsions are two-phase systems in which one liquid is dispersed throughout another in the form of globules, which can be either oil-in-water or water-in-oil. Emulsions may include pharmaceutically acceptable non-aqueous liquids and solvents, emulsifiers and preservatives. Suspensions may include a pharmaceutically acceptable suspending agent and a preservative. The aqueous alcoholic solution may comprise pharmaceutically acceptable acetals, such as di (lower alkyl) acetals of lower alkyl aldehydes, e.g. acetaldehyde diethyl acetal; and water-soluble solvents having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, hydroalcoholic solutions. Syrups are concentrated aqueous solutions of sugars, such as sucrose, and may also contain preservatives. For liquid dosage forms, for example, a solution in polyethylene glycol may be diluted with a sufficient amount of a pharmaceutically acceptable liquid carrier, such as water, for precise and convenient administration.
Other useful liquid and semi-solid dosage forms include, but are not limited to, those comprising the active ingredients provided herein and a secondary mono-or poly-alkylene glycol, including: 1, 2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, where 350, 550, 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations may further include one or more antioxidants, such as Butylated Hydroxytoluene (BHT), Butylated Hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
Dosage unit formulations for oral administration may be microencapsulated, where appropriate. They may also be prepared as extended or sustained release compositions, for example by coating or embedding the particulate material in a polymer, wax or the like.
The oral pharmaceutical composition provided by the invention can also be provided in the form of liposome, micelle, microsphere or nano system. Micellar dosage forms can be prepared using the methods described in U.S. Pat. No.6,350,458.
The pharmaceutical compositions provided herein can be provided as non-effervescent or effervescent granules and powders for reconstitution into liquid dosage forms. Pharmaceutically acceptable carriers and excipients used in non-effervescent granules or powders may include diluents, sweeteners and wetting agents. Pharmaceutically acceptable carriers and excipients used in effervescent granules or powders may include organic acids and sources of carbon dioxide.
Coloring and flavoring agents may be used in all of the above dosage forms.
The disclosed compounds may also be conjugated to soluble polymers as targeted drug carriers. Such polymers include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamide-phenol, polyhydroxyethylaspartamidephenol or polyoxyethylene polylysine substituted with palmitoyl residues. In addition, the disclosed compounds may be combined with a class of biodegradable polymers used in achieving controlled release of a drug, such as polylactic acid, poly-caprolactone, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates, and crosslinked or amphipathic block copolymers of hydrogels.
The pharmaceutical compositions provided by the present invention may be formulated into immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed release forms.
The pharmaceutical compositions provided by the present invention may be co-formulated with other active ingredients that do not impair the intended therapeutic effect, or with substances that supplement the intended effect.
The pharmaceutical compositions provided by the present invention may be administered parenterally by injection, infusion or implantation for local or systemic administration. Parenteral administration as used herein includes intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous administration.
The pharmaceutical compositions provided herein can be formulated in any dosage form suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems and solid forms suitable for solution or suspension in a liquid prior to injection. Such dosage forms may be prepared according to conventional methods known to those skilled in The art of pharmaceutical Science (see Remington: The Science and Practice of Pharmacy, supra).
Pharmaceutical compositions intended for parenteral administration may include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives to inhibit microbial growth, stabilizers, solubility enhancers, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, thickening agents, pH adjusting agents, and inert gases.
Suitable aqueous carriers include, but are not limited to: water, saline, normal saline or Phosphate Buffered Saline (PBS), sodium chloride injection, Ringers injection, isotonic glucose injection, sterile water injection, dextrose and lactated Ringers injection. Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and the medium chain triglycerides of coconut oil, and palm seed oil. Water-miscible vehicles include, but are not limited to, ethanol, 1, 3-butanediol, liquid polyethylene glycols (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerol, N-methyl-2-pyrrolidone, N-dimethylacetamide, and dimethylsulfoxide.
Suitable antimicrobial agents or preservatives include, but are not limited to, phenol, cresol, mercurial, benzyl alcohol, chlorobutanol, methyl and propyl parabens, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl and propyl parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerol and glucose. Suitable buffers include, but are not limited to, phosphate and citrate. Suitable antioxidants are, for example, those
Suitable local anesthetics include, but are not limited to, procaine hydrochloride, suitable suspending and dispersing agents are those described herein, including sodium carboxymethylcellulose, hydroxypropylmethylcellulose, and polyvinylpyrrolidone, suitable emulsifying agents include those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate, suitable sequestering or chelating agents include, but are not limited to, edta, suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid, suitable complexing agents include, but are not limited to, cyclodextrins, including α -cyclodextrin, β -cyclodextrin, hydroxypropyl- β -cyclodextrin, sulfobutyl ether- β -cyclodextrin, and sulfobutyl ether 7- β -cyclodextrin (r) (r)
Figure GDA0001986101440000481
CyDex,Lenexa,KS)。
The pharmaceutical compositions provided herein may be formulated for single or multiple dose administration. The single dose formulations are packaged in ampoules, vials or syringes. The multi-dose parenteral formulation must contain a bacteriostatic or fungistatic concentration of the antimicrobial agent. All parenteral formulations must be sterile, as is known and practiced in the art.
In one embodiment, the pharmaceutical composition is provided as a ready-to-use sterile solution. In another embodiment, the pharmaceutical compositions are provided as sterile dried soluble products, including lyophilized powders and subcutaneous injection tablets, which are reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a ready-to-use sterile suspension. In yet another embodiment, the pharmaceutical composition is formulated as a sterile, dry, insoluble product that is reconstituted with a carrier prior to use. In yet another embodiment, the pharmaceutical composition is formulated as a sterile emulsion ready for use.
The pharmaceutical composition may be formulated as a suspension, solid, semi-solid, or thixotropic liquid for depot administration for implantation. In one embodiment, the disclosed pharmaceutical compositions are dispersed in a solid internal matrix surrounded by an outer polymeric membrane that is insoluble in body fluids but allows diffusion therethrough of the active ingredient in the pharmaceutical composition.
Suitable internal matrices include polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinyl chloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, silicone carbonate copolymers, hydrogels of hydrophilic polymers such as esters of acrylic and methacrylic acid, collagen, crosslinked polyvinyl alcohol, and partially hydrolyzed polyvinyl acetate of the class of copolymers.
Suitable outer polymeric films include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubber, polydimethylsiloxane, neoprene, chlorinated polyethylene, polyvinyl chloride, copolymers of chlorinated ethylene and vinyl acetate, vinylidene chloride, ethylene and propylene, ionomers polyethylene terephthalate, butyl rubber chlorohydrin rubber, ethylene/vinyl alcohol copolymers, ethylene/vinyl acetate/vinyl alcohol terpolymers, and ethylene/ethyleneoxyethanol copolymers.
In another aspect, the disclosed pharmaceutical compositions may be formulated in any dosage form suitable for administration to a patient by inhalation, such as a dry powder, aerosol, suspension, or solution composition. In one embodiment, the disclosed pharmaceutical compositions may be formulated in a dosage form suitable for inhalation administration to a patient as a dry powder. In yet another embodiment, the disclosed pharmaceutical compositions may be formulated in a dosage form suitable for inhalation administration to a patient via a nebulizer. Dry powder compositions for delivery to the lung by inhalation typically comprise a finely powdered form of the disclosed compounds andone or more pharmaceutically acceptable excipients in the form of a fine powder. Pharmaceutically acceptable excipients that are particularly suitable for use as dry powders are known to those skilled in the art and include lactose, starch, mannitol, and mono-, di-and polysaccharides. Fine powders may be prepared, for example, by micronization and milling. Generally, the size-reduced (e.g., micronized) compound may pass through a D of about 1 to 10 microns50Values (e.g., measured by laser diffraction).
Aerosols can be formulated by suspending or dissolving the disclosed compounds in a liquefied propellant. Suitable propellants include chlorinated hydrocarbons, hydrocarbons and other liquefied gases. Representative propellants include: trichlorofluoromethane (propellant 11), dichlorofluoromethane (propellant 12), dichlorotetrafluoroethane (propellant 114), tetrafluoroethane (HFA-134a), 1-difluoroethane (HFA-152a), difluoromethane (HFA-32), pentafluoroethane (HFA-12), heptafluoropropane (HFA-227a), perfluoropropane, perfluorobutane, perfluoropentane, butane, isobutane and pentane. Aerosols comprising the compounds disclosed herein are typically administered to a patient via a Metered Dose Inhaler (MDI). Such devices are known to those skilled in the art
The aerosol may contain additional pharmaceutically acceptable excipients that may be used by MDIs, such as surfactants, lubricants, co-solvents, and other excipients, to improve the physical stability of the formulation, to improve valve characteristics, to improve solubility, or to improve taste.
Pharmaceutical compositions suitable for transdermal administration may be prepared as discrete patches intended to remain in intimate contact with the epidermis of the patient for an extended period of time. For example, the active ingredient may be delivered from a patch agent by iontophoresis, as generally described in Pharmaceutical Research,3(6),318 (1986).
Pharmaceutical compositions suitable for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. For example, ointments, creams and gels may be formulated with a water or oil base, and suitable thickeners and/or gelling agents and/or solvents. Such bases may include, water, and/or oils such as liquid paraffin and vegetable oils (e.g., peanut oil or castor oil), or solvents such as polyethylene glycol. Thickeners and gelling agents used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycol, lanolin, beeswax, carbopol and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifiers.
Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilizing agents, dispersing agents, suspending agents or thickening agents.
Powders for external use may be formed in the presence of any suitable powder base, for example talc, lactose or starch. Drops may be formulated with an aqueous or non-aqueous base containing one or more dispersing agents, solubilising agents, suspending agents or preservatives.
Topical formulations may be administered by application to the affected area one or more times per day; an occlusive dressing covering the skin is preferably used. Adhesive depot systems allow for continuous or extended administration.
Use of the Compounds and compositions of the invention
The compounds or pharmaceutical compositions disclosed in the present invention can be used for the preparation of a medicament for the treatment, prevention, amelioration, control or alleviation of immunoinflammatory disorders or tumors in mammals, particularly humans, and for the preparation of other medicaments that antagonize the NOD1/2 receptor.
The compounds or pharmaceutical compositions of the present invention may be applied to, but are in no way limited to, the prevention, treatment, or alleviation of immunoinflammatory disorders or tumors by administering to a patient an effective amount of a compound or pharmaceutical composition of the present invention. The immunoinflammatory disease or tumor further includes, but is not limited to, rheumatoid arthritis, sjogren's syndrome, wegener's granulomatosis, behcet's disease, sarcoidosis, takayasu's arteritis, reactive arthritis, osteoarthritis, aids, allergic diseases, rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus or multiple sclerosis, colon cancer, rectal cancer, gastric adenocarcinoma, pancreatic cancer, bladder cancer, gallbladder cancer, breast cancer, kidney cancer, renal cell carcinoma, liver cancer, hepatocellular carcinoma, lung cancer, skin cancer, melanoma, thyroid cancer, osteosarcoma, soft tissue sarcoma, head and neck cancer, central nervous system tumor, glioma, glioblastoma, ovarian cancer, uterine cancer, endometrial cancer, prostate cancer, acute myelogenous leukemia or acute lymphocytic leukemia or their metastatic cancers, Polycythemia vera, primary hemo-allergic arthritis, osteoarthritis, AIDS, allergic diseases, rheumatoid arthritis, allergic asthma, chronic fatigue, type II diabetes, hay fever, lupus erythematosus or multiple sclerosis.
In addition to being beneficial for human therapy, the compounds and pharmaceutical compositions of the present invention may also find application in veterinary therapy for pets, animals of the introduced species and mammals in farm animals. Examples of other animals include horses, dogs, and cats. Herein, the compound of the present invention includes pharmaceutically acceptable derivatives thereof.
Method of treatment
In one embodiment, the presently disclosed methods of treatment comprise administering to a patient in need thereof a safe and effective amount of a compound of the present invention or a pharmaceutical composition comprising a compound of the present invention. Various embodiments of the present disclosure include methods of treating the above-mentioned diseases by administering to a patient in need thereof a safe and effective amount of a disclosed compound or a pharmaceutical composition comprising a disclosed compound.
In one embodiment, the disclosed compounds or pharmaceutical compositions comprising the disclosed compounds may be administered by any suitable route of administration, including systemic and topical administration. Systemic administration includes oral, parenteral, transdermal and rectal administration. Typical parenteral administration refers to administration by injection or infusion, including intravenous, intramuscular, and subcutaneous injection or infusion. Topical administration includes application to the skin and intraocular, otic, intravaginal, inhalation, and intranasal administration. In one embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered orally. In another embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered by inhalation. In yet another embodiment, the presently disclosed compounds or compositions comprising the presently disclosed compounds may be administered intranasally.
In one embodiment, a disclosed compound or a pharmaceutical composition comprising a disclosed compound may be administered once or several times at different time intervals over a specified period of time according to a dosing regimen. For example, once, twice, three times or four times daily. In one embodiment, the administration is once daily. In yet another embodiment, the administration is twice daily. The administration may be carried out until the desired therapeutic effect is achieved or the desired therapeutic effect is maintained indefinitely. Suitable dosing regimens for the disclosed compounds or pharmaceutical compositions comprising the disclosed compounds depend on the pharmacokinetic properties of the compound, such as dilution, distribution and half-life, which can be determined by the skilled person. In addition, suitable dosing regimens for the compounds or pharmaceutical compositions comprising the disclosed compounds, including the duration of the regimen, will depend on the condition being treated, the severity of the condition being treated, the age and physical condition of the patient being treated, the medical history of the patient being treated, the nature of concurrent therapy, the desired therapeutic effect, and other factors within the knowledge and experience of the skilled artisan. Such a skilled artisan will also appreciate that adjustments to the subject's response to the dosage regimen, or the need for changes in the subject's patient over time, may be required.
The compounds disclosed herein may be administered simultaneously, or before or after, one or more other therapeutic agents. The compounds of the invention may be administered separately from the other therapeutic agents, by the same or different routes of administration, or together with them in pharmaceutical compositions.
For an individual of about 50-70kg, the disclosed pharmaceutical compositions and combinations may be in unit dosage form containing from about 1-1000mg, or from about 1-500mg, or from about 1-250mg, or from about 1-150mg, or from about 0.5-100mg, or from about 1-50mg of the active ingredient. The therapeutically effective amount of the compound, pharmaceutical composition or combination thereof will depend on the species, weight, age and condition of the individual, the disease (disorder) or illness (disease) being treated, or the severity thereof. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each active ingredient to prevent, treat or inhibit the progression of the disease (disorder) or condition (disease).
The above cited dose profiles have been demonstrated in vitro and in vivo tests using beneficial mammals (e.g., mice, rats, dogs, monkeys) or isolated organs, tissues and specimens thereof. The compounds disclosed herein are used in vitro in the form of solutions, e.g. aqueous solutions, and also enterally, parenterally, especially intravenously, in vivo, e.g. in the form of suspensions or aqueous solutions.
In one embodiment, a therapeutically effective dose of a compound of the present disclosure is from about 0.1mg to about 2,000mg per day. The pharmaceutical composition thereof should provide a dose of the compound of about 0.1mg to about 2,000 mg. In a particular embodiment, the pharmaceutical dosage unit form is prepared to provide from about 1mg to about 2,000mg, from about 10mg to about 1,000mg, from about 20mg to about 500mg, or from about 25mg to about 250mg of the principal active ingredient or a combination of principal ingredients per dosage unit form. In a particular embodiment, the pharmaceutical dosage unit form is prepared to provide about 10mg,20mg,25mg,50mg,100mg,250mg,500mg,1000mg or 2000mg of the primary active ingredient.
In addition, the compounds disclosed herein may be administered in the form of a prodrug. In the present invention, a "prodrug" of a disclosed compound is a functional derivative that, when administered to a patient, is ultimately released in vivo. When administering the compounds disclosed herein in the form of a prodrug, one skilled in the art can practice one or more of the following: (a) altering the in vivo onset time of the compound; (b) altering the duration of action of the compound in vivo; (c) altering the in vivo delivery or distribution of the compound; (d) altering the in vivo solubility of the compound; and (e) overcoming side effects or other difficulties faced by the compounds. Typical functional derivatives useful for preparing prodrugs comprise variants of the compounds which are cleaved in vivo either chemically or enzymatically. These variants, which involve the preparation of phosphates, amides, esters, thioesters, carbonates and carbamates, are well known to those skilled in the art.
General synthetic procedure
To illustrate the invention, the following examples are set forth. It is to be understood that the invention is not limited to these embodiments, but is provided as a means of practicing the invention.
In general, the compounds of the present invention may be prepared by the methods described herein, wherein the substituents are as defined in formula (I), unless otherwise indicated. The following reaction schemes and examples serve to further illustrate the context of the invention.
Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare a number of other compounds of the invention, and other methods for preparing the compounds of the invention are considered to be within the scope of the invention. For example, the synthesis of those non-exemplified compounds according to the present invention can be successfully accomplished by those skilled in the art by modification, such as appropriate protection of interfering groups, by the use of other known reagents in addition to those described herein, or by some routine modification of reaction conditions. In addition, the reactions disclosed herein or known reaction conditions are also recognized as being applicable to the preparation of other compounds of the present invention.
Reagents for the experiments were purchased from Beijing coupling technologies, Inc., Bailingwei technologies, Inc., Acros Organics, Alfa Aesar, Sigma-Aldrich, and TCI, unless otherwise specified, and used without purification. The solvents used in the experiments were purchased mainly from Beijing chemical plant and Shigaku chemical Co., Ltd, and were used without further treatment except for THF and DMF which were further treated by the solvent purification system of VAC corporation. GF254 thin layer chromatography silica gel plate, GF254 silica gel thick preparation plate and silica gel powder (60-100 mesh, 160-.
HP L C-MS Analyzer HP L C Analyzer Agilent 1100HP L C System, Agilent G1312A Pump, Agilent G1314A UV Detector, Agilent G1313A Autosampler, Agilent G1316A column incubator and diverter valve the column is Kromasil C18 analytical column (4.6 μm,4.6mm × 50mm), available from DIKMA Inc. the mobile phase is acetonitrile and water containing 0.05% HCOOH.Linear gradient elution 5:95(v: v) acetonitrile-H2O to 95:5(v: v) acetonitrile-H2O, time 5minutes, flow rate 1m L/min. UV detection wavelength 254 nm.ThermoFinniigigan L CQ-Advaage Mass spectrometer, 5% of the eluent enters into the Mass spectrometer in either positive ion or negative ion scanning mode, electrospray ionization (electrospray ionization) system is used to monitor the purity of the primary reaction compounds and primary ion source.
UP L C-MS Analyzer-Acquity UP L C-MS System from Waters including binary solvent manager, sample manager, column manager, PDA detector, and SQ Mass Spectroscopy
Figure GDA0001986101440000531
BEH C18 column (1.7 μm,2.1mm × 50 mm). mobile phase is acetonitrile and water containing 0.05% HCOOH.Linear gradient elution 5:95(v: v) acetonitrile-H2O to 95:5(v: v) acetonitrile-H2O, time 3minutes, flow rate 0.3m L/min. UV detection wavelength 254 nm. SQ mass spectrometer adopts positive ion or negative ion scanning mode, electrospray ionization source (ESI). The method is mainly used for reaction monitoring and preliminary determination of compound purity.
HP L C analyzer, Agilent 1260HP L C system, Agilent G1311C quaternary pump, Agilent G4212B ultraviolet detector, Agilent G1367E high-performance autosampler, Agilent G1316A column incubator chiral analysis column DAICE L CHIRA L PAK AD-H,250 × 4.6.6 mm,5 μ M (produced by DAICE L of Daxylon Japan.) mobile phase is n-hexane/isopropanol, isocratic elution UV detection wavelength 254 nm.
The high resolution mass spectrometer is an Agilent L C/MSD TOF system, a chromatographic column is an Agilent ZORBAX SB-C18(Rapid resolution,3.5 mu m,2.1 × 30mm), a mobile phase is MeOH, H2O, 75:25(v: v), contains 5 mmol/L formic acid, is eluted at an equal rate for 5min, has a flow rate of 0.40m L/min, and is used for mass spectrometry in a positive ion scanning mode and an electrospray ion source (ESI) mainly used for determining the accurate molecular weight of a target compound.
Nuclear magnetic resonance apparatus: varian Mercury 300MHz,400MHz,500MHz,600MHz and Bruker Avance 400MHz, solvent CDCl3, DMSO-d6, acetone-d6or methanol-d 4.
Melting point apparatus: yanaco micro melting point apparatus, OptiMelt melting point apparatus, were not calibrated.
Typical synthetic procedures for preparing the disclosed compounds of the invention are shown in the following synthetic schemes. Unless otherwise stated, each R1、R2、R3、R4、R5、X1、X2、X3、X4And Y has the definition as described in the present invention.
Synthesis scheme 1
Figure GDA0001986101440000532
Compound (A) to (B)6) Can be prepared according to scheme 1 in the above figure: the compound (1) reacts with concentrated sulfuric acid and potassium nitrate under the heating condition to obtain an intermediate (2) (ii) a Intermediate (A)2) Reacting with substituted phenol under heating to obtain an intermediate (3) (ii) a Under the action of N, N-diisopropylcarbodiimide, intermediate (A)3) Condensation reaction to obtain intermediate (4) (ii) a Intermediate (A)4) Cyclizing under the action of glacial acetic acid and iron powder to obtain an intermediate (5) (ii) a Under the action of NaH, intermediate (5) Amino group in (1) by R3Substitution to give the compound (A)6)。
The compounds of the present invention are described below with reference to specific examples, which are intended to be illustrative only and not to be limiting in any way.
Example 1
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (2- ((3-fluorobenzyl) oxa) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (1)
Figure GDA0001986101440000541
Step (a): synthesis of 5-chloro-2, 4-dinitrobenzoic acid (1-a)
Figure GDA0001986101440000542
While stirring at room temperature, 3-chlorobenzoic acid (10.0 g,63.9mmol) was dissolved in 120m L concentrated sulfuric acid, potassium nitrate (16.5 g,163.2 mmol) was added in 15 min, the reaction solution was reacted at 80 ℃ for 30min, at 110 ℃ for 2h and at 120 ℃ for 2h in this order, the reaction solution was poured into 660 g of ice water, filtered, and the resulting white solid was recrystallized from a mixed solvent of ethanol and water to give 8.08 g of pale yellow crystals (1-a) in 51.3% yield.1H NMR(400 MHz,DMSO-d6)14.21(brs,1H),8.85(s,1H),8.28(s,1H).13C NMR(100 MHz,DMSO-d6)163.78,147.95,145.70,132.86,132.06,130.52,122.05.
Step (b): synthesis of 5- (3- (dimethylamino) phenoxy) -2, 4-dinitrobenzoic acid (1-b)
Figure GDA0001986101440000543
Adding the compound 1-a (4.877 g,19.78 mmol) into 36m L of water, adding sodium bicarbonate (3.556 g,42.33 mmol,2.14eq) and 3, 5-dimethylphenol (2.872g,20.97mmol,1.06eq), heating to 110 ℃, refluxing for 2 hours until the raw materials disappear, cooling, performing ice-water bath, adjusting the pH to 3-4, filtering, draining and drying to obtain the compound 1-b with the yield of 60%.
Step (c): synthesis of (R) -dimethyl-2- (5- (3- (dimethylamino) phenoxy) -2, 4-dinitrobenzoylamino) pentanediacid ester (1-c)
Figure GDA0001986101440000551
Sequentially adding the compound 1-b (4.546g,13.1mmol), D-glutamic acid dimethyl ester hydrochloride (2.909g,13.75mmol, 1.05e q), N, N-diisopropylcarbodiimide DIC (3.06m L, 19.65mmol, 1.5eq) into 120m L THF, stirring at room temperature for 4h, filtering, evaporating, adding dichloromethane, washing with water, washing with saturated common salt water, drying, filtering and evaporating,and (4) performing column chromatography to obtain the compound 1-c with the yield of 70-85%.1H NMR(400MHz,DMSO-d6)9.15(d,J=7.7Hz,1H),8.85(s,1H),7.32-7.28(m,1H),6.96(s,1H),6.69(dd,J=8.4,2.2Hz,1H),6.57-6.56(m,1H),6.47(dd,J=7.9,2.0Hz,1H),4.43(td,J=8.8,5.3Hz,1H),3.58(d,J=8.1Hz,6H),2.92(s,6H),2.41–2.37(m,2H),2.05(td,J=13.3,7.7Hz,1H),1.85(dt,J=14.3,8.3Hz,1H)。
Step (d): synthesis of (R) -methyl 3- (8-amino-7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate methyl ester (1-d)
Figure GDA0001986101440000552
Adding the compound 1-C (250 mg,0.464mmol) into 10m L glacial acetic acid, adding iron powder (520mg,9.38mmol,20eq), heating to 90 ℃, reacting for 1.5-2.5h, detecting by L C-MS until the target product is a main point (about 75%), cooling, adding 20m L DCM, passing through kieselguhr, evaporating the filtrate to dryness, and performing column chromatography to obtain the compound 1-d with the yield of 30-40%.1H NMR(400MHz,DMSO-d6)10.15(s,1H),8.05(d,J=5.2Hz,1H),7.15-7.11(m,1H),7.00(s,1H),6.48(dd,J=8.3,2.2Hz,1H),6.39(s,1H),6.39-6.37(m,1H),6.18(dd,J=7.9,2.0Hz,1H),5.70(s,2H),3.69-3.64(m,1H),3.56(s,3H),2.88(s,6H),2.44–2.39(m,2H),2.04–1.97(m,1H),1.80(dt,J=14.2,7.3Hz,1H)。
A step (e): synthesis of 2- ((3-fluorobenzyl) oxy) acetic acid (1-e)
Figure GDA0001986101440000561
Under the protection of argon, adding an ice-water bath, adding sodium hydrogen (880mg, 60% and 22mmol) into 30m L THF, dropwise adding a compound of m-fluorobenzyl alcohol (10mmol and 1eq) under the protection of the ice-water bath, stirring the ice-water bath for 1h, dropwise adding a 2-bromoacetic acid (1.11g and 8mmol and 0.8eq)/20m L THF solution, naturally heating to room temperature and stirring overnight after dropwise adding, carrying out methanol quenching reaction, extracting impurities from methyl tert-butyl ether, adjusting the pH of a water phase to 2-3 by using 6N HCl, extracting by using ethyl acetate, drying, filtering and evaporating to dryness to obtain a compound 1-e, wherein the yield is 71.3%.
Step (f): synthesis of the title Compound 1
Dissolving the compound 1-e (0.2mmol,2eq) in 4m L dichloromethane, dropwise adding oxalyl chloride (0.4mmol, 4eq) and 6 mu L DMF under an ice water bath, transferring to room temperature for reaction for 1h, allowing raw materials to disappear (detecting by adding methanol), evaporating the solvent to dryness, dissolving in 1m L THF to obtain an intermediate THF solution, using 100% of the intermediate THF solution directly for the next step reaction, mixing the THF solution with 59(0.1mmol) and triethylamine (0.5mmol,5eq) in 2m L THF, reacting for 1h, and performing silica gel column chromatography to obtain the compound 1 with the yield of 64.1%.1H NMR(400MHz,DMSO-d6)10.43(s,1H),9.40(s,1H),8.45(d,J=5.3Hz,1H),8.16(s,1H),7.34–7.28(m,1H),7.23–7.09(m,5H),6.57(dd,J=8.4,2.0Hz,1H),6.43(s,1H),6.28(dd,J=7.9,1.7Hz,1H),4.62(s,2H),4.27–4.18(m,2H),3.75-3.70(m,1H),3.57(s,3H),2.89(s,6H),2.45-2.38(m,2H),2.04(dt,J=14.5,6.8Hz,1H),1.82(td,J=14.7,7.8Hz,1H)。
Example 2
Synthesis of methyl (R) -methyl 3- (8- (2- ((3-bromobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (2)
Figure GDA0001986101440000562
Step (a): synthesis of 2- ((3-bromobenzyl) oxy) acetic acid (2-a)
Figure GDA0001986101440000563
M-bromobenzyl alcohol and 2-bromoacetic acid were reacted according to the synthesis of compound 1-e described in example 1 to give 2-a.
Step (b): synthesis of the title Compound 2
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 2-a, compound 2 was obtained in 69% yield.1H NMR(400MHz,DMSO-d6)10.43(s,1H),9.40(s,1H),8.44(d,J=5.3Hz,1H),8.15(s,1H),7.56(s,1H),7.48(d,J=7.9Hz,1H),7.33(d,J=7.6Hz,1H),7.22(m,2H),7.14(s,1H),6.57(d,J=8.4Hz,1H),6.43(s,1H),6.29(d,J=9.7Hz,1H),4.60(s,2H),4.27–4.18(m,2H),3.72–3.69(m,1H),3.57(s,3H),2.89(s,6H),2.45–2.36(m,2H),2.04(dt,J=14.6,6.9Hz,1H),1.82(dt,J=12.2,6.2Hz,1H)。
Example 3
Synthesis of methyl (R) -methyl 3- (8- (2- ((4-bromobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (3)
Figure GDA0001986101440000571
Step (a): synthesis of 2- ((3-bromobenzyl) oxy) acetic acid (3-a)
Figure GDA0001986101440000572
P-bromobenzyl alcohol and 2-bromoacetic acid were reacted according to the synthesis of compound 1-e described in example 1 to give 3-a.
Step (b): synthesis of the title Compound 3
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 3-a, compound 3 was obtained in 64% yield.1H NMR(400MHz,DMSO-d6)10.44(s,1H),9.38(s,1H),8.45(d,J=5.3Hz,1H),8.17(s,1H),7.42(d,J=8.3Hz,2H),7.27(d,J=8.3Hz,2H),7.22(t,J=8.2Hz,1H),7.16(s,1H),6.59(dd,J=8.4,2.0Hz,1H),6.43(t,J=2.0Hz,1H),6.27(dd,J=7.9,1.8Hz,1H),4.57(s,2H),4.26–4.17(m,2H),3.72–3.69(m,1H),3.57(s,3H),2.90(s,6H),2.45–2.378(m,2H),2.09–2.00(m,1H),1.82(td,J=14.7,7.9Hz,1H)。
Example 4
Synthesis of methyl (R) -methyl 3- (8- (2- ((4-p-chlorobenzyl alcohol) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (4)
Figure GDA0001986101440000581
Step (a): synthesis of 2- ((4-chlorobenzyl) oxy) acetic acid (4-a)
Figure GDA0001986101440000582
P-chlorobenzyl alcohol and 2-bromoacetic acid were reacted according to the method for synthesizing compound 1-e described in example 1 to give 4-a.
Step (b): synthesis of the title Compound 4
Prepared according to the method for synthesizing compound 1 described in example 1, using 1-d and 4-a, compound 4 was obtained in 52.7% yield.1H NMR(400MHz,DMSO-d6)10.44(s,1H),9.38(s,1H),8.45(d,J=5.3Hz,1H),8.17(s,1H),7.34(d,J=8.4Hz,2H),7.29(d,J=8.5Hz,2H),7.22(t,J=8.2Hz,1H),7.15(s,1H),6.59(dd,J=8.4,1.9Hz,1H),6.43(s,1H),6.27(dd,J=7.9,1.6Hz,1H),4.59(s,2H),4.26–4.17(m,2H),3.72–3.69(m,1H),3.53(s,3H),2.90(s,6H),2.45–2.40(m,2H),2.04(dt,J=20.8,7.1Hz,1H),1.82(td,J=14.6,7.6Hz,1H)。
Example 5
Synthesis of methyl (R) -methyl 3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (5)
Figure GDA0001986101440000583
Step (a): synthesis of 2- (benzyloxy) acetic acid (5-a)
Figure GDA0001986101440000584
Benzyl alcohol was reacted with 2-bromoacetic acid according to the synthesis of compound 1-e described in example 1 to give 5-a.
Step (b): synthesis of the title Compound 5
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 5-a, compound 5 was obtained in 28.2% yield.1H NMR(400MHz,DMSO-d6)10.43(s,1H),9.40(s,1H),8.45(d,J=5.2Hz,1H),8.18(s,1H),7.33–7.20(m,6H),7.15(s,1H),6.59(dd,J=8.3,1.7Hz,1H),6.45(s,1H),6.30(dd,J=7.8,1.3Hz,1H),4.60(s,2H),4.25–4.16(m,2H),3.75-3.71(m,1H),3.57(s,3H),2.90(s,6H),2.45-2.40(m,2H),2.04(td,J=14.2,6.9Hz,1H),1.83(td,J=14.6,7.7Hz,1H)。
Example 6
Synthesis of (R) -3- (8- (2- (phenoxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N, N-diethylpropionamide (6)
Figure GDA0001986101440000591
Compound 5(1mmol) was dissolved in 6m L THF and 3m L H2And adding L iOH (3mmol,3eq) into the O mixed solution, reacting at room temperature for 0.5h until the reaction is completed, evaporating the reaction solution to dryness, adding water for dilution, adjusting the pH to acidity by using 1M hydrochloric acid, precipitating a solid, filtering, drying to obtain an intermediate product with the yield of 82%, dissolving the intermediate product (0.5mmol), DIC (1mmol,2eq) and HoSu (1mmol,2eq) in a THF solution, stirring at room temperature overnight, adding diethylamine (1.5mmol,3eq) for reacting for 0.5h, evaporating the solvent to dryness, and performing column chromatography to separate to obtain the compound 6 with the yield of 12%.1H NMR(400MHz,DMSO-d6)10.40(s,1H),9.39(s,1H),8.44(d,J=5.3Hz,1H),8.17(s,1H),7.33–7.20(m,6H),7.14(s,1H),6.59(dd,J=8.4,2.1Hz,1H),6.45-6.44(m,1H),6.29(dd,J=7.9,1.8Hz,1H),4.60(s,2H),4.25–4.16(m,2H),3.75-3.70(m,1H),3.26–3.17(m,4H),2.90(s,6H),2.41-2.37(m,2H),2.05–1.97(m,1H),1.80(td,J=14.9,7.7Hz,1H),1.06(t,J=7.0Hz,3H),0.96(t,J=7.0Hz,3H)。
Example 7
Synthesis of (R) -2- (benzyloxy) -N- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-3- (3-oxo-3- (piperidin-1-yl) propyl) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) acetamide (7)
Figure GDA0001986101440000592
Following the synthesis of compound 6 as described in example 6, using 5Was prepared with piperidine to give compound 7 in 46% yield.1H NMR(400MHz,DMSO-d6)10.40(s,1H),9.40(s,1H),8.43(d,J=5.0Hz,1H),8.17(s,1H),7.34–7.20(m,6H),7.13(s,1H),6.59(d,J=8.2Hz,1H),6.45(s,1H),6.30(d,J=7.8Hz,1H),4.60(s,2H),4.20(d,J=2.2Hz,2H),3.72–3.69(m,1H),3.36(s,4H),2.90(s,6H),2.39(t,J=7.1Hz,2H),2.01–1.98(m,1H),1.82–1.77(m,1H),1.53(s,2H),1.44(s,2H),1.37(s,2H)。
Example 8
Synthesis of methyl (R) -methyl 3- (8- (2- (benzyloxy) acetamido) -7- (2-chloro-5- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (8)
Step (a): synthesis of 2-chloro-5- (dimethylamino) phenol (8-a)
Figure GDA0001986101440000601
Dissolving 3, 5-dimethylphenol (2g, 0.014577mol) in an aqueous solution (15M L) of sodium hydroxide (1.447g, 0.03617mol), adding sodium hypochlorite (12.5M L, 0.0146mol) dropwise in an ice-water bath, reacting at 0 ℃ for 1h, adjusting the pH to 7 with 6M hydrochloric acid, extracting with ethyl acetate, washing with saturated saline, drying, filtering, evaporating, and carrying out column chromatography to obtain 195mg of a compound 8-a.
Step (b): synthesis of 5- (2-chloro-5- (dimethylamino) phenoxy) -2, 4-dinitrobenzoic acid (8-b)
Figure GDA0001986101440000602
According to the method for synthesizing the compound 1-b described in example 1, the compound 1-a and 8-a were reacted to obtain the compound 8-b in a yield of 51%.
Step (c): synthesis of (R) -dimethyl-2- (5- (2-chloro-5- (dimethylamino) phenoxy) -2, 4-dinitrobenzoylamino) pentanedionate (8-c)
Figure GDA0001986101440000603
Compound 8-b was reacted with D-glutamic acid dimethyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to obtain compound 8-c with a yield of 90%.
Step (d): synthesis of (R) -methyl 3- (8-amino-7- (2-chloro-5- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate methyl ester (8-d)
Figure GDA0001986101440000611
Compound 8-c was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 8-d in 29% yield.
A step (e): synthesis of the title Compound 8
Prepared according to the method for the synthesis of compound 1 described in example 1, using 8-d and 5-a, compound 8 was obtained in 34% yield.1H NMR(400MHz,DMSO-d6)10.41(s,1H),9.50(s,1H),8.45(d,J=5.3Hz,1H),8.19(s,1H),7.40-7.37(m,3H),7.27-7.22(m,3H),6.91(s,1H),6.68(dd,J=9.0,2.9Hz,1H),6.62(d,J=2.8Hz,1H),4.66(s,2H),4.30–4.21(m,2H),3.72-3.67(m,1H),3.56(s,3H),2.91(s,6H),2.44–2.40(m,2H),2.03(dt,J=14.6,6.5Hz,1H),1.81(td,J=14.6,7.6Hz,1H)。
Example 9
Synthesis of (R) -3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-octadecylpropanamide (9)
Figure GDA0001986101440000612
Prepared according to the synthesis of compound 6 described in example 6, using 5 and octadecanamine, compound 9 was obtained in 56% yield.1H NMR(400MHz,DMSO-d6)10.40(s,1H),9.38(s,1H),8.43(d,J=5.3Hz,1H),8.19(s,1H),7.75(t,J=5.5Hz,1H),7.32(dd,J=7.0,2.3Hz,2H),7.28-7.20(m,4H),7.15(s,1H),6.59(dd,J=8.4,2.1Hz,1H),6.44-6.43(m,1H),6.29(dd,J=7.9,1.8Hz,1H),4.60(s,2H),4.24–4.15(m,2H),3.67-3.62(m,1H),2.98-2.93(m,2H),2.90(s,6H),2.20-2.16(m,2H),2.00(td,J=13.6,7.0Hz,1H),1.79(td,J=13.9,6.9Hz,1H),1.28–1.17(m,32H),0.85(t,J=6.8Hz,3H)。
Example 10
Synthesis of (R) -3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-dodecylpropionamide (10)
Figure GDA0001986101440000621
Prepared according to the synthesis of compound 6 described in example 6, using 5 and dodecylamine, compound 10 was obtained in 83% yield.1H NMR(400MHz,DMSO-d6)10.39(s,1H),9.37(s,1H),8.42(d,J=5.3Hz,1H),8.18(s,1H),7.75(t,J=5.5Hz,1H),7.31(dd,J=7.0,2.2Hz,2H),7.26–7.19(m,4H),7.14(s,1H),6.58(dd,J=8.3,2.1Hz,1H),6.43-6.42(m,1H),6.28(dd,J=7.9,1.8Hz,1H),4.59(s,2H),4.24–4.14(m,2H),3.66-3.61(m,1H),2.98-2.93(m,2H),2.89(s,6H),2.19-2.16(m,2H),1.99(td,J=14.3,7.5Hz,1H),1.78(td,J=14.5,7.4Hz,1H),1.31-1.16(m,20H),0.84(t,J=6.7Hz,3H)。
Example 11
Synthesis of (R) -3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-hexadecylpropionamide (11)
Figure GDA0001986101440000622
Prepared according to the synthesis of compound 6 described in example 6, using 5 with hexadecylamine, compound 11 was obtained in 61% yield.1H NMR(400MHz,DMSO-d6)10.39(s,1H),9.37(s,1H),8.42(d,J=5.3Hz,1H),8.18(s,1H),7.75(t,J=5.5Hz,1H),7.31(dd,J=6.9,2.2Hz,2H),7.28–7.17(m,4H),7.14(s,1H),6.58(dd,J=8.3,2.0Hz,1H),6.43(m,1H),6.28(dd,J=7.9,1.7Hz,1H),4.59(s,2H),4.26–4.12(m,2H),3.64(m,1H),2.95(m,2H),2.89(s,6H),2.17(m,2H),2.06–1.93(m,1H),1.78(td,J=14.2,7.0Hz,1H),1.23(m,28H),0.85(t,J=6.7Hz,3H)。
Example 12
Synthesis of (R) -3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N, N-dimethylpropanamide (12)
Figure GDA0001986101440000631
Prepared according to the synthesis of compound 6 described in example 6, using 5 and dimethylamine, compound 12 was obtained in 64% yield.1H NMR(400MHz,DMSO-d6)10.39(s,1H),9.38(s,1H),8.42(d,J=5.2Hz,1H),8.16(s,1H),7.32(dd,J=6.9,2.4Hz,2H),7.27–7.20(m,4H),7.13(s,1H),6.58(dd,J=8.4,2.0Hz,1H),6.45-6.44(m,1H),6.29(dd,J=7.9,1.8Hz,1H),4.59(s,2H),4.24–4.15(m,2H),3.75-3.70(m,1H),2.91(s,3H),2.89(s,6H),2.77(s,3H),2.40-2.37(m,2H),2.04–1.95(m,1H),1.79(td,J=14.6,7.3Hz,1H)。
Example 13
Synthesis of (R) -3- (8- (2- (benzyloxy) acetylamino) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-propanoic acid propionamide (13)
Figure GDA0001986101440000632
Prepared according to the procedure described for the synthesis of compound 6 in example 6, using 5 and n-propylamine, compound 13 was obtained in 70% yield.1H NMR(400MHz,DMSO-d6)10.39(s,1H),9.38(s,1H),8.43(d,J=5.1Hz,1H),8.17(s,1H),7.77(t,J=4.7Hz,1H),7.32–7.19(m,6H),7.13(s,1H),6.59–6.57(m,1H),6.44(s,1H),6.28(d,J=7.8Hz,1H),4.59(s,2H),4.24–4.15(m,2H),3.68-3.63(m,1H),2.96–2.93(m,2H),2.89(s,6H),2.19(t,J=6.6Hz,2H),2.04–1.97(m,1H),1.78(dt,J=21.2,7.3Hz,1H),1.31(dt,J=14.0,7.0Hz,2H),0.75(t,J=7.3Hz,3H)。
Example 14
Synthesis of methyl (R) -methyl 3- (8- (2- ((2-chlorobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (14)
Figure GDA0001986101440000641
Step (a): synthesis of 2- ((2-benzyl chloride) oxy) acetic acid (14-a)
Figure GDA0001986101440000642
According to the synthetic method of the compound 1-e described in the embodiment 1, the reaction of the parachlorobenzyl alcohol and the 2-bromoacetic acid is carried out to obtain 14-a.
Step (b): synthesis of the title Compound 14
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 14-a, compound 14 was prepared in 86% yield.1H NMR(400MHz,DMSO-d6)10.41(s,1H),9.39(s,1H),8.43(d,J=5.2Hz,1H),8.16(s,1H),7.48(d,J=7.3Hz,1H),7.42(d,J=7.8Hz,1H),7.34-7.30(m,1H),7.23–7.18(m,2H),7.13(s,1H),6.56(dd,J=8.2,1.3Hz,1H),6.41(s,1H),6.27(d,J=7.9Hz,1H),4.69(s,2H),4.32–4.24(m,2H),3.74-3.70(m,1H),3.56(s,3H),2.88(s,6H),2.44-2.33(m,2H),2.02(dt,J=14.8,7.4Hz,1H),1.81(td,J=14.8,7.6Hz,1H)。
Example 15
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (2- ((4-fluorobenzyl) oxa) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (15)
Figure GDA0001986101440000643
Step (a): synthesis of 2- ((4-Benzylfluoro) oxy) acetic acid (15-a)
Figure GDA0001986101440000644
P-fluorobenzyl alcohol and 2-bromoacetic acid were reacted according to the synthesis of compound 1-e described in example 1 to give 15-a.
Step (b): synthesis of the title Compound 15
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 15-a, compound 15 was obtained in 68% yield.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.36(s,1H),8.43(d,J=5.4Hz,1H),8.16(s,1H),7.38-7.34(m,2H),7.23-7.19(m,1H),7.14(s,1H),7.08-7.03(m,2H),6.58(dd,J=8.3,2.1Hz,1H),6.43-6.42(m,1H),6.27(dd,J=7.9,1.9Hz,1H),4.57(s,2H),4.23-4.15(m,2H),3.74-3.70(m,1H),3.56(s,3H),2.89(s,6H),2.44–2.39(m,2H),2.06-2.00(m,1H),1.82(td,J=14.6,7.9Hz,1H)。
Example 16
Synthesis of methyl (R) -methyl 3- (8- (2- ((3-chlorobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (16)
Figure GDA0001986101440000651
Step (a): synthesis of 2- ((3-benzyl chloride) oxy) acetic acid (16-a)
Figure GDA0001986101440000652
M-chlorobenzyl alcohol and 2-bromoacetic acid were reacted according to the method for synthesizing compound 1-e described in example 1 to give 16-a.
Step (b): synthesis of the title Compound 16
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 16-a, compound 16 was obtained in 56% yield.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.40(s,1H),8.44(d,J=5.3Hz,1H),8.15(s,1H),7.42(s,1H),7.36-7.32(m,1H),7.30–7.29(m,2H),7.23-7.18(m,1H),7.14(s,1H),6.57(dd,J=8.3,2.1Hz,1H),6.43-6.42(m,1H),6.29(dd,J=7.9,1.9Hz,1H),4.61(s,2H),4.27–4.18(m,2H),3.75-3.70(m,1H),3.57(s,3H),2.89(s,6H),2.45–2.40(m,2H),2.07–2.00(m,1H),1.82(td,J=14.7,7.9Hz,1H)。
Example 17
Synthesis of methyl (R) -methyl 3- (8- (2- ((2-chloro-4-fluorobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (17)
Figure GDA0001986101440000661
Step (a): synthesis of 2- ((2-chloro-4-fluorobenzyl) oxy) acetic acid (17-a)
Figure GDA0001986101440000662
According to the synthesis of compound 1-e described in example 1, 2-chloro-4-fluorobenzyl alcohol and 2-bromoacetic acid were reacted to give 17-a.
Step (b): synthesis of the title Compound 17
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 17-a, compound 17 was obtained in 50% yield.1H NMR(400MHz,DMSO-d6)10.41(s,1H),9.36(s,1H),8.43(d,J=5.4Hz,1H),8.15(s,1H),7.54-7.50(m,1H),7.42-7.39(m,1H),7.21-7.17(m,1H),7.13(s,1H),7.10-7.05(m,1H),6.56(dd,J=8.4,2.1Hz,1H),6.40-6.39(m,1H),6.25(dd,J=7.9,1.8Hz,1H),4.65(s,2H),4.31–4.22(m,2H),3.74-3.69(m,1H),3.56(s,3H),2.88(s,6H),2.44–2.39(m,2H),2.08–1.97(m,1H),1.81(td,J=14.7,8.0Hz,1H)。
Example 18
Synthesis of methyl (R) -methyl 3- (8- (2- ((3, 4-difluorobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (18)
Figure GDA0001986101440000663
Step (a): synthesis of 2- ((3, 4-fluorobenzyl) oxy) acetic acid (18-a)
Figure GDA0001986101440000664
According to the synthesis method of the compound 1-e in the embodiment 1, the reaction of the parachlorobenzyl alcohol and the 2-bromoacetic acid is carried out to obtain 18-a.
Step (b): synthesis of the title Compound 18
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 18-a, compound 18 was obtained in 76% yield.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.37(s,1H),8.44(d,J=5.2Hz,1H),8.14(s,1H),7.42–7.37(m,1H),7.33-7.26(m,1H),7.21–7.17(m,2H),7.13(s,1H),6.56(dd,J=8.3,1.8Hz,1H),6.40(s,1H),6.26(dd,J=7.9,1.5Hz,1H),4.57(s,2H),4.25–4.17(m,2H),3.74-3.69(m,1H),3.56(s,3H),2.88(s,6H),2.44-2.37(m,2H),2.03(dt,J=14.3,6.9Hz,1H),1.81(td,J=14.4,7.5Hz,1H)。
Example 19
Synthesis of (R) -3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-tert-butylpropanamide (19)
Figure GDA0001986101440000671
Prepared according to the synthesis of compound 6 described in example 6, using 5 with tert-butylamine, compound 19 was obtained in 20% yield.1H NMR(400MHz,DMSO-d6)10.38(s,1H),9.37(s,1H),8.38(d,J=5.2Hz,1H),8.17(s,1H),7.36(s,1H),7.32–7.25(m,5H),7.20(t,J=8.2Hz,1H),7.13(s,1H),6.59–6.57(m,1H),6.43(s,1H),6.26(dd,J=7.8,1.3Hz,1H),4.59(s,2H),4.23–4.15(m,2H),3.68-3.63(m,1H),2.89(s,6H),2.15-2.11(m,2H),1.96(dt,J=13.1,8.7Hz,1H),1.75(dt,J=20.2,6.5Hz,1H),1.17(s,9H)。
Example 20
Synthesis of methyl (R) -methyl 3- (8- (2- (benzyloxy) acetamido) -7- (2-chloro-3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (20)
Figure GDA0001986101440000672
Step (a): synthesis of 2-chloro-3- (dimethylamino) phenol (20-a)
Figure GDA0001986101440000681
Dissolving 3, 5-dimethylphenol (2g, 0.014577mol) in an aqueous solution (15M L) of sodium hydroxide (1.447g, 0.03617mol), adding sodium hypochlorite (12.5M L, 0.0146mol) dropwise in an ice-water bath, reacting at 0 ℃ for 1h, adjusting the pH to 7 with 6M hydrochloric acid, extracting with ethyl acetate, washing with saturated saline, drying, filtering, evaporating, and carrying out column chromatography to obtain 102mg of a compound 20-a.
Step (b): synthesis of 5- (2-chloro-3- (dimethylamino) phenoxy) -2, 4-dinitrobenzoic acid (20-b)
Figure GDA0001986101440000682
Following the synthesis of compound 1-b as described in example 1, compound 1-a was reacted with 20-a to give compound 20-b in 55% yield.
Step (c): synthesis of (R) -dimethyl-2- (5- (2-chloro-3- (dimethylamino) phenoxy) -2, 4-dinitrobenzoylamino) pentanedionate (20-c)
Figure GDA0001986101440000683
Compound 20-b was reacted with D-glutamic acid dimethyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to obtain compound 20-c in a yield of 95%.
Step (d): synthesis of (R) -methyl 3- (8-amino-7- (2-chloro-3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate methyl ester (20-d)
Figure GDA0001986101440000684
Compound 20-c was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 20-d with a yield of 32%.
A step (e): synthesis of the title Compound 20
Prepared according to the method for the synthesis of compound 1 described in example 1, using 20-d and 5-a, compound 20 was obtained in 34% yield.1H NMR(400MHz,DMSO-d6)10.43(s,1H),9.46(s,1H),8.44(d,J=5.1Hz,1H),8.20(s,1H),7.34(dd,J=14.6,7.0Hz,3H),7.25–7.19(m,3H),7.09(d,J=8.1Hz,1H),6.96(s,1H),6.89(d,J=8.0Hz,1H),4.63(s,2H),4.27–4.19(m,2H),3.74-3.69(m,1H),3.56(s,3H),2.78(s,6H),2.413-2.40(m,2H),2.03(td,J=14.2,6.7Hz,1H),1.81(td,J=14.8,7.7Hz,1H)。
Example 21
Synthesis of methyl (R) -methyl 3- (8- (2- (benzyloxy) acetamido) -7- (4-chloro-3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (21)
Figure GDA0001986101440000691
Step (a): synthesis of 4-chloro-3- (dimethylamino) phenol (21-a)
Figure GDA0001986101440000692
3, 5-dimethylphenol (2g, 0.014577mol) is dissolved in an aqueous solution (15M L) of sodium hydroxide (1.447g, 0.03617mol), sodium hypochlorite (12.5M L, 0.0146mol) is added dropwise in an ice-water bath, after the addition is finished, the reaction is carried out at 0 ℃ for 1h, the pH is adjusted to be neutral by 6M hydrochloric acid, ethyl acetate is used for extraction, saturated salt water is used for washing, drying, filtering and drying are carried out, and column chromatography is carried out to obtain 21-a (195 mg).
Step (b): synthesis of 5- (4-chloro-3- (dimethylamino) phenoxy) -2, 4-dinitrobenzoic acid (21-b)
Figure GDA0001986101440000693
According to the method for synthesizing the compound 1-b described in example 1, the compound 1-a and 21-a were reacted to obtain the compound 21-b with a yield of 51%.
Step (c): synthesis of (R) -dimethyl-2- (5- (4-chloro-3- (dimethylamino) phenoxy) -2, 4-dinitrobenzoylamino) pentanedionate (21-c)
Figure GDA0001986101440000701
Compound 21-b was reacted with D-glutamic acid dimethyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to obtain compound 21-c with a yield of 90%.
Step (d): synthesis of (R) -methyl 3- (8-amino-7- (4-chloro-3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate methyl ester (21-d)
Figure GDA0001986101440000702
Compound 21-c was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 21-d in 29% yield.
A step (e): synthesis of the title Compound 21
Prepared according to the method for the synthesis of compound 1 described in example 1, using 21-d and 5-a, compound 21 was obtained in 61% yield.1H NMR(400MHz,DMSO-d6)10.46(s,1H),9.37(s,1H),8.45(d,J=5.2Hz,1H),8.14(s,1H),7.40(d,J=8.7Hz,1H),7.31-7.26(m,5H),7.20(s,1H),6.87(d,J=2.6Hz,1H),6.61(dd,J=8.7,2.6Hz,1H),4.57(s,2H),4.22–4.14(m,2H),3.76-3.71(m,1H),3.56(s,3H),2.71(s,6H),2.42(dd,J=11.5,6.9Hz,2H),2.04(td,J=14.3,6.9Hz,1H),1.83(td,J=14.8,7.8Hz,1H).
Example 22
Synthesis of methyl (R) -methyl 3- (8- (2- (benzyloxy) acetamido) -7- (3-chloro-5- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (22)
Figure GDA0001986101440000711
Step (a): synthesis of 2-amino-3-chloro-5-nitrophenol (22-a)
Figure GDA0001986101440000712
2-amino-5-nitrophenol (12g,0.0779mol) was dissolved in 430m L acetonitrile, stirred to dissolve, NCS (10.8g,0.081mol) was added, the mixture was heated to 80 ℃ to react for 2 hours, the solvent was evaporated to dryness, and column chromatography was performed to obtain compound 22-a (10.3g) with a yield of 70%.
Step (b): synthesis of 3-chloro-5-nitrophenol (22-b)
Figure GDA0001986101440000713
Adding the compound 22-a (11g, 0.05836mol) into 187m L ethanol, adding concentrated sulfuric acid (5.4m L, 98 percent and 0.0986mol), heating to reflux for 30min, adding sodium nitrite solid in batches, reacting for 30min after use, refluxing for 1h, evaporating the solvent after no raw material exists, adding water and ethyl acetate, separating liquid, washing with water, washing with sodium bicarbonate solution, washing with saturated salt solution, drying, filtering, evaporating, and performing column chromatography to obtain the compound 22-b (6.3g) with the yield of 59.5 percent.
Step (c): synthesis of 3-amino-5-chlorophenol (22-c)
Figure GDA0001986101440000714
Dissolving the compound 22-b (2g, 0.01153mol) in 35m L ethanol and 35m L acetic acid, heating to 85 ℃, adding iron powder (6.4g,0.1153mol) in batches, reacting for 2h until no raw material exists, filtering, evaporating to dryness, adding water, adjusting the pH value to 8-9 with sodium bicarbonate, extracting with dichloromethane, drying, filtering, evaporating to dryness, and performing column chromatography to obtain the compound 22-c (1.15g) with the yield of 59%.
Step (d): synthesis of 3-chloro-5- (dimethylamino) phenol (22-d)
Figure GDA0001986101440000721
Adding the compound 22-C (1.435g,10mmol) into 6m L DMF, heating to 80 ℃, dropwise adding dimethyl sulfate (2.4m L, 25mmol), reacting for 1h, adding T L C without raw materials into reaction liquid, adding the reaction liquid into 30m L water, adjusting the pH value to 7-8 with sodium bicarbonate solution, extracting with ethyl acetate, washing with saturated salt water, drying, filtering and evaporating to dryness, and carrying out column chromatography to obtain the compound 22-d (0.412g), wherein the yield is 24%.
A step (e): synthesis of 5- (3-chloro-5- (dimethylamino) phenoxy) -2, 4-dinitrobenzoic acid (22-e)
Figure GDA0001986101440000722
Following the synthesis of compound 1-b as described in example 1, compound 1-a was reacted with 22-d to give compound 22-e in 85% yield.
Step (f): synthesis of (R) -dimethyl-2- (5- (3-chloro-5- (dimethylamino) phenoxy) -2, 4-dinitrobenzoylamino) pentanedionate (22-f)
Figure GDA0001986101440000723
Compound 22-e was reacted with D-glutamic acid dimethyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to give compound 22-f in 100% yield.
Step (g): synthesis of methyl (R) -methyl 3- (8-amino-7- (3-chloro-5- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (22-g)
Figure GDA0001986101440000724
Compound 22-f was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 22-g with a yield of 32%.
A step (h): synthesis of the title Compound 22
Prepared according to the method for synthesizing the compound 1 in the example 1 by adopting 22-g and 5-a, the yield is 68 percent。1H NMR(400MHz,DMSO-d6)10.45(s,1H),9.35(s,1H),8.45(d,J=5.2Hz,1H),8.14(s,1H),7.30-7.28(m,5H),7.20(s,1H),6.55(s,1H),6.36(s,1H),6.28(s,1H),4.58(s,2H),4.23–4.14(m,2H),3.77-3.72(m,1H),3.56(s,3H),2.89(s,6H),2.45–2.41(m,2H),2.04(td,J=14.5,7.2Hz,1H),1.83(td,J=14.8,7.7Hz,1H)。
Example 23
Synthesis of methyl (R) -methyl 3- (8- (2- (benzyloxy) acetamido) -2, 5-dioxo-7- (2,4, 5-trichloro-3- (dimethylamino) phenoxy) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (23)
Figure GDA0001986101440000731
Step (a): synthesis of 2,4, 5-trichloro-3- (dimethylamino) phenol (23-a)
Figure GDA0001986101440000732
Adding the compound 22-d (400mg,2.33mmol) into 12m L acetonitrile, adding NCS (311mg,2.33mmol), reacting at room temperature for 3h, heating to react for 30min, supplementing NCS (31mg,0.233mol) until no raw material exists, evaporating the solvent, and performing column chromatography to obtain the compound 23-a (63 mg).
Step (b): synthesis of 2, 4-dinitro-5- (2,4, 5-trichloro-3- (dimethylamino) phenoxy) benzoic acid (23-b)
Figure GDA0001986101440000733
According to the method for synthesizing the compound 1-b described in example 1, the compound 1-a and 23-a were reacted to obtain the compound 23-b with a yield of 64%.
Step (c): synthesis of (R) -dimethyl-2- (2, 4-dinitro-5- (2,4, 5-trichloro-3- (dimethylamino) phenoxy) benzamido) pentanediacid ester (23-c)
Figure GDA0001986101440000741
Compound 23-b was reacted with D-glutamic acid dimethyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to obtain compound 23-c in a yield of 91%.
Step (d): synthesis of (R) -methyl 3- (8-amino-2, 5-dioxo-7- (2,4, 5-trichloro-3- (dimethylamino) phenoxy) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (23-d)
Figure GDA0001986101440000742
Compound 23-c was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 23-d in 26% yield.
A step (e): synthesis of the title Compound 23
Prepared according to the method for synthesizing compound 1 described in example 1, using 23-d and 5-a, yield 38%.1H NMR(400MHz,DMSO-d6)10.48(s,1H),9.42(s,1H),8.48(d,J=4.8Hz,1H),8.13(s,1H),7.33–7.25(m,6H),7.12(s,1H),4.61(s,2H),4.25–4.16(m,2H),3.77-3.72(m,1H),3.56(s,3H),2.87(s,6H),2.42(s,2H),2.07-2.01(m,1H),1.85-1.80(m,1H)。
Example 24
Synthesis of methyl (R) -methyl 3- (8- (2- (benzyloxy) acetamido) -7- (2, 3-dichloro-5- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (24)
Figure GDA0001986101440000743
Step (a): synthesis of 2, 3-dichloro-5- (dimethylamino) phenol (24-a)
Figure GDA0001986101440000751
Adding the compound 22-d (400mg,2.33mmol) into 12m L acetonitrile, adding NCS (311mg,2.33mmol), reacting at room temperature for 3h, heating to react for 30min, supplementing NCS (31mg,0.233mol) until no raw material exists, evaporating the solvent, and performing column chromatography to obtain the compound 24-a (98 mg).
Step (b): synthesis of 5- (2, 3-dichloro-5- (dimethylamino) phenoxy) -2, 4-dinitrobenzoic acid (24-b)
Figure GDA0001986101440000752
According to the method for synthesizing the compound 1-b described in example 1, the compound 1-a and 24-a were reacted to obtain the compound 24-b in a yield of 77%.
Step (c): synthesis of (R) -dimethyl-2- (5- (2, 3-dichloro-5- (dimethylamino) phenoxy) -2, 4-dinitrobenzamido) pentanediate (24-c)
Figure GDA0001986101440000753
Compound 24-b was reacted with D-glutamic acid dimethyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to obtain compound 24-c with a yield of 98%.
Step (d): synthesis of (R) -methyl 3- (8-amino-7- (2, 3-dichloro-5- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate methyl ester (24-d)
Figure GDA0001986101440000754
Compound 24-c was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 24-d in 25% yield.
A step (e): synthesis of the title Compound 24
Prepared according to the method for synthesizing compound 1 described in example 1, using 24-d and 5-a, yield 24%.1H NMR(400MHz,DMSO-d6)10.43(s,1H),9.46(s,1H),8.45(d,J=4.7Hz,1H),8.17(s,1H),7.36-7.34(m,2H),7.25-7.24(m,3H),6.95(s,1H),6.85(s,1H),6.58(s,1H),4.64(s,2H),4.28–4.19(m,2H),3.72-3.67(m,1H),3.56(s,3H),2.91(s,6H),2.45-2.37(m,2H),2.06-2.00(m,1H),1.86–1.79(m,1H)。
Example 25
Synthesis of methyl (R) -methyl 3- (8- (2- (benzyloxy) acetamido) -2, 5-dioxo-7- (2,3, 4-trichloro-5- (dimethylamino) phenoxy) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (25)
Figure GDA0001986101440000761
Step (a): synthesis of 2,3, 4-trichloro-5- (dimethylamino) phenol (25-a)
Figure GDA0001986101440000762
Adding compound 22-d (400mg,2.33mmol) into 12m L acetonitrile, adding NCS (311mg,2.33mmol), reacting at room temperature for 3h, heating to react for 30min, adding NCS (31mg,0.233mol) until no raw material is available, evaporating solvent, and performing column chromatography to obtain compound 25-a (63 mg).
Step (b): synthesis of 2, 4-dinitro-5- (2,3, 4-trichloro-5- (dimethylamino) phenoxy) benzoic acid (25-b)
Figure GDA0001986101440000763
Following the synthesis of compound 1-b as described in example 1, compound 1-a was reacted with 25-a to give compound 25-b in 88% yield.
Step (c): synthesis of (R) -dimethyl-2- (2, 4-dinitro-5- (2,3, 4-trichloro-5- (dimethylamino) phenoxy) benzamido) pentanediacid ester (25-c)
Figure GDA0001986101440000771
Compound 25-b was reacted with D-glutamic acid dimethyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to obtain compound 25-c in 84% yield.
Step (d): synthesis of (R) -methyl 3- (8-amino-2, 5-dioxo-7- (2,3, 4-trichloro-5- (dimethylamino) phenoxy) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (25-d)
Figure GDA0001986101440000772
Compound 25-c was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 25-d in 26% yield.
A step (e): synthesis of the title Compound 25
Prepared according to the method for the synthesis of compound 1 described in example 1, using 25-d and 5-a, in 36% yield.1H NMR(400MHz,DMSO-d6)10.47(s,1H),9.46(s,1H),8.46(d,J=5.5Hz,1H),8.14(s,1H),7.33–7.31(m,2H),7.25-7.23(m,3H),7.02(d,J=11.0Hz,2H),4.62(s,2H),4.26-4.18(m,2H),3.73-3.69(m,1H),3.56(s,3H),2.74(s,6H),2.41(dd,J=11.0,7.3Hz,2H),2.03(dt,J=14.2,6.7Hz,1H),1.82(td,J=14.6,7.8Hz,1H)。
Example 26
Synthesis of (R) -3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-cyclopropylpropionamide (26)
Figure GDA0001986101440000773
Prepared according to the procedure for the synthesis of compound 6 described in example 6, using 5 and cyclopropylamine, compound 26 is obtained in 64% yield.1H NMR(400MHz,DMSO-d6)10.38(s,1H),9.37(s,1H),8.39(d,J=5.2Hz,1H),8.16(s,1H),7.85(d,J=3.6Hz,1H),7.32–7.25(m,5H),7.23-7.19(m,1H),7.13(s,1H),6.58(dd,J=8.6,1.5Hz,1H),6.44(s,1H),6.30–6.28(m,1H),4.59(s,2H),4.23–4.15(m,2H),3.68-3.63(m,1H),2.89(s,6H),2.57–2.53(m,1H),2.15-2.12(m,2H),1.98(dt,J=13.9,7.0Hz,1H),1.76(td,J=14.6,7.6Hz,1H),0.57-0.51(m,2H),0.33-0.26(m,2H)。
Example 27
Synthesis of (R) -2- (benzyloxy) -N- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-3- (3-oxo-3- (pyrrolidin-1-yl) propyl) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) acetamide (27)
Figure GDA0001986101440000781
Prepared according to the procedure for the synthesis of compound 6 described in example 6, using 5 and tetrahydropyrrole, compound 27 is obtained in 95% yield.1H NMR(400MHz,DMSO)10.38(s,1H),9.38(s,1H),8.43(d,J=5.0Hz,1H),8.16(s,1H),7.33–7.26(m,5H),7.24-7.19(m,1H),7.13(s,1H),6.58(d,J=8.3Hz,1H),6.44(s,1H),6.29(d,J=7.9Hz,1H),4.60(s,2H),4.24–4.15(m,2H),3.76-3.72(m,1H),3.34(s,2H),3.23(t,J=6.7Hz,2H),2.89(s,6H),2.36-2.33(m,2H),2.01(td,J=13.6,6.8Hz,1H),1.85–1.78(m,3H),1.76-1.69(m,2H)。
Example 28
Synthesis of (R) -2- ((4-chlorobenzyl) oxa) -N- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-3- (3-oxo-3- (piperidin-1-yl) propyl) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) acetamide (28)
Figure GDA0001986101440000782
Prepared according to the procedure for the synthesis of compound 6 described in example 6, using 4 and piperidine, compound 28 was obtained in 93% yield.1H NMR(400MHz,DMSO-d6)10.38(s,1H),9.36(s,1H),8.41(d,J=5.1Hz,1H),8.15(s,1H),7.33(d,J=8.1Hz,2H),7.28(d,J=8.2Hz,2H),7.23-7.18(m,1H),7.14(s,1H),6.58(d,J=8.3Hz,1H),6.42(s,1H),6.26(d,J=7.9Hz,1H),4.58(s,2H),4.25–4.16(m,2H),3.74-3.69(m,1H),3.36-3.34(m,4H),2.89(s,6H),2.41-2.37(m,2H),2.04–1.95(m,1H),1.79(td,J=14.5,7.4Hz,1H),1.54-1.53(m,2H),1.43-1.36(m,4H)。
Example 29
Synthesis of methyl (R) -methyl 3- (7- (2-chloro-3- (dimethylamino) phenoxy) -8- (2- ((4-chlorobenzyl) oxa) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (29)
Figure GDA0001986101440000791
Prepared according to the method for the synthesis of compound 1 described in example 1, using 4-a and 20-d, yield 75%.1H NMR(400MHz,DMSO-d6)10.43(s,1H),9.44(s,1H),8.43(d,J=5.1Hz,1H),8.18(s,1H),7.36-7.34(m,3H),7.24(d,J=8.0Hz,2H),7.09(d,J=8.1Hz,1H),6.97(s,1H),6.87(d,J=8.0Hz,1H),4.62(s,2H),4.28–4.20(m,2H),3.73-3.69(m,1H),3.56(s,3H),2.77(s,6H),2.41-2.39(m,2H),2.02(dt,J=14.2,7.1Hz,1H),1.81(td,J=14.7,7.7Hz,1H)。
Example 30
Synthesis of methyl (R) -methyl 3- (7- (4-chloro-3- (dimethylamino) phenoxy) -8- (2- ((4-chlorobenzyl) oxa) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (30)
Figure GDA0001986101440000792
Prepared according to the method for the synthesis of compound 1 described in example 1, using 4-a and 21-d, compound 30 was obtained in 73% yield.1H NMR(400MHz,DMSO-d6)10.45(s,1H),9.36(s,1H),8.44(d,J=5.1Hz,1H),8.13(s,1H),7.39(d,J=8.7Hz,1H),7.34–7.29(m,4H),7.20(s,1H),6.86(s,1H),6.59(dd,J=8.2,1.2Hz,1H),4.57(s,2H),4.23–4.15(m,2H),3.76-3.71(m,1H),3.56(s,3H),2.72(s,6H),2.44-2.42(m,2H),2.08–2.00(m,1H),1.87-1.78(m,1H)。
Example 31
Synthesis of (R) -3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N, N-dibutylpropanamide (31)
Figure GDA0001986101440000801
Prepared according to the method for synthesizing compound 6 described in example 6, using 5 with di-n-butylamine, compound 31 was obtained in 53% yield.1H NMR(400MHz,DMSO-d6)10.41(s,1H),9.39(s,1H),8.44(d,J=5.3Hz,1H),8.17(s,1H),7.33–7.25(m,5H),7.24-7.19(m,1H),7.12(s,1H),6.58(dd,J=8.4,2.1Hz,1H),6.44-6.43(m,1H),6.28(dd,J=7.9,1.9Hz,1H),4.59(s,2H),4.24–4.15(m,2H),3.70-3.66(m,1H),3.23–3.10(m,4H),2.89(s,6H),2.46-2.31(m,2H),2.05–1.96(m,1H),1.79(td,J=13.9,7.3Hz,1H),1.46–1.39(m,2H),1.38-1.30(m,2H),1.25-1.22(m,2H),1.18–1.13(m,2H),0.86(t,J=7.3Hz,3H),0.81(t,J=7.3Hz,3H)。
Example 32
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-8- (2- ((4- (trifluoromethyl) benzyl) oxa) acetamido) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (32)
Figure GDA0001986101440000802
Step (a): synthesis of 2- ((4- (trifluoromethyl) benzyl) oxy) acetic acid (32-a)
Figure GDA0001986101440000803
P-trifluoromethylbenzyl alcohol and 2-bromoacetic acid were reacted according to the method for synthesizing the compound 1-e described in example 1 to obtain 32-a with a yield of 55%.
Step (b): synthesis of the title Compound 32
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 32-a, compound 32 was obtained in 66% yield.1H NMR(400MHz,DMSO-d6)10.43(s,1H),9.41(s,1H),8.44(d,J=5.0Hz,1H),8.16(s,1H),7.57(d,J=8.1Hz,2H),7.52(d,J=8.0Hz,2H),7.22-7.18(m,1H),7.16(s,1H),6.58–6.56(m,1H),6.43(s,1H),6.27–6.25(m,1H),4.70(s,2H),4.30–4.21(m,2H),3.75-3.70(m,1H),3.56(s,3H),2.87(s,6H),2.44-2.40(m,2H),2.03(td,J=14.5,7.3Hz,1H),1.82(td,J=14.7,7.7Hz,1H)。
Example 33
(R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (2- ((4-methoxybenzyl) oxa) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate methyl ester (33)
Figure GDA0001986101440000811
Step (a): synthesis of 2- ((4- (methoxy) benzyl) oxy) acetic acid (33-a)
Figure GDA0001986101440000812
P-methoxybenzyl alcohol and 2-bromoacetic acid were reacted according to the method for synthesizing the compound 1-e described in example 1 to obtain 33-a with a yield of 46%.
Step (b): synthesis of the title Compound 33
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 33-a, compound 33 was obtained in 59% yield.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.35(s,1H),8.43(d,J=5.4Hz,1H),8.17(s,1H),7.25–7.20(m,3H),7.14(s,1H),6.77(d,J=8.6Hz,2H),6.59(dd,J=8.4,2.1Hz,1H),6.45-6.44(m,1H),6.29(dd,J=7.9,1.9Hz,1H),4.51(s,2H),4.19–4.11(m,2H),3.74–3.71(m,1H),3.70(s,3H),3.56(s,3H),2.89(s,6H),2.44-2.37(m,2H),2.03(dt,J=14.2,6.4Hz,1H),1.81(td,J=14.6,7.8Hz,1H)。
Example 34
Synthesis of methyl (R) -methyl 3- (8- (2- ((4-cyanobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (34)
Figure GDA0001986101440000821
Step (a): synthesis of 2- ((4- (cyano) benzyl) oxy) acetic acid (34-a)
Figure GDA0001986101440000822
P-cyanobenzaldehyde (2.62g,20mmol) was added to EtOH, and NaBH was added portionwise in an ice water bath4(0.912g,24mmol), reaction at room temperature for 1h, after reaction is complete, post-treatment, columnChromatography gave 2.57g of intermediate, which was reacted with 2-bromoacetic acid according to the synthesis of compound 1-e described in example 1 to give 34-a in 47% yield.
Step (b): synthesis of the title Compound 34
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 34-a, compound 34 was obtained in 59% yield.1H NMR(400MHz,DMSO)10.43(s,1H),9.40(s,1H),8.44(d,J=5.4Hz,1H),8.14(s,1H),7.69(d,J=8.2Hz,2H),7.49(d,J=8.2Hz,2H),7.22-7.18(m,1H),7.14(s,1H),6.57(dd,J=8.4,2.1Hz,1H),6.40-6.39(m,1H),6.25(dd,J=7.9,1.9Hz,1H),4.69(s,2H),4.30–4.21(m,2H),3.74-3.70(m,1H),3.56(s,3H),2.88(s,6H),2.44–2.39(m,2H),2.03(dt,J=15.2,7.0Hz,1H),1.82(td,J=14.6,8.0Hz,1H)。
Example 35
Synthesis of methyl (R) -methyl 3- (8- (2- ((4-chlorobenzyl) oxa) acetamido) -2, 5-dioxo-7- (3- (pyrrolidin-1-yl) phenoxy) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (35)
Figure GDA0001986101440000823
Step (a): synthesis of 3- (pyrrolidin-1-yl) phenol (35-a)
Figure GDA0001986101440000831
Adding M-hydroxyaniline (1.09g, 10mmol) into 1, 4-dibromobutane (2.4M L,20 mmol), heating to 130 ℃, reacting for 12h, stopping the reaction when the raw materials are completely reacted, cooling, adding ethyl acetate, water, separating, washing an ethyl acetate phase with 1M HCl, combining water phases, adjusting the pH value to 6-7 with sodium hydroxide, extracting with ethyl acetate, washing with saturated salt water, drying, filtering, evaporating to dryness, and performing column chromatography to obtain the compound 35-a (0.737g) with the yield of 45%.
Step (b): synthesis of 2, 4-dinitro-5- (3- (pyrrolidin-1-yl) phenoxy) benzoic acid (35-b)
Figure GDA0001986101440000832
Following the synthesis of compound 1-b as described in example 1, compound 1-a was reacted with 35-a to give compound 35-b in 87% yield.
Step (c): synthesis of (R) -dimethyl-2- (2, 4-dinitro-5- (3- (pyrrolidin-1-yl) phenoxy) benzamido) pentanediate (35-c)
Figure GDA0001986101440000833
Compound 35-b was reacted with D-glutamic acid dimethyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to obtain compound 35-c in a yield of 72%.
Step (d): synthesis of methyl (R) -methyl 3- (8-amino-2, 5-dioxo-7- (3- (pyrrolidin-1-yl) phenoxy) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (35-d)
Figure GDA0001986101440000834
Compound 35-c was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 35-d in 33% yield.
A step (e): synthesis of the title Compound 35
Prepared according to the synthesis of compound 1 described in example 1, using 35-d and 4-a, in 65% yield.1H NMR(400MHz,DMSO-d6))10.44(s,1H),9.37(s,1H),8.45(d,J=5.4Hz,1H),8.17(s,1H),7.31(q,J=8.6Hz,4H),7.21–7.17(m,2H),6.41(dd,J=8.3,1.6Hz,1H),6.23–6.20(m,2H),4.59(s,2H),4.26–4.18(m,2H),3.72–3.69(m,1H),3.57(s,3H),3.19(t,J=6.3Hz,4H),2.45–2.40(m,2H),2.09–2.00(m,1H),1.95(t,J=6.4Hz,4H),1.82(dt,J=14.4,7.8Hz,1H)。
Example 36
Synthesis of (R) -3- (8- (2- ((4-chlorobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-propylpropanamide (36)
Figure GDA0001986101440000841
Prepared according to the procedure described for the synthesis of compound 6 in example 6, using 4 and n-propylamine, compound 36 was obtained in 44% yield.1H NMR(400MHz,DMSO-d6)10.40(s,1H),9.36(s,1H),8.43(d,J=5.3Hz,1H),8.15(s,1H),7.77(t,J=5.6Hz,1H),7.33(d,J=8.5Hz,2H),7.28(d,J=8.5Hz,2H),7.23-7.19(m,1H),7.13(s,1H),6.58(dd,J=8.4,2.0Hz,1H),6.43-6.42(m,1H),6.25(dd,J=7.9,1.7Hz,1H),4.58(s,2H),4.25–4.16(m,2H),3.68-3.63(m,1H),2.96–2.91(m,2H),2.90(m,6H),2.20-2.17(m,2H),2.00(dt,J=13.4,6.8Hz,1H),1.78(td,J=14.3,7.0Hz,1H),1.36–1.27(m,2H),0.75(t,J=7.4Hz,3H)。
Example 37
Synthesis of (R) -3- (8- (2- ((4-bromobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-propylpropionamide (37)
Figure GDA0001986101440000842
Prepared according to the procedure described for the synthesis of compound 6 in example 6, using 3 and n-propylamine, compound 37 was obtained in 54% yield.1H NMR(400MHz,DMSO-d6)10.40(s,1H),9.36(s,1H),8.43(d,J=5.3Hz,1H),8.16(s,1H),7.77(t,J=5.5Hz,1H),7.41(d,J=8.3Hz,2H),7.26(d,J=8.3Hz,2H),7.23-7.18(m,1H),7.13(s,1H),6.58(dd,J=8.4,2.0Hz,1H),6.42(s,1H),6.25(dd,J=7.9,1.7Hz,1H),4.57(s,2H),4.25–4.16(m,2H),3.68-3.63(m,1H),2.96–2.92(m,2H),2.90(s,6H),2.20-2.17(m,2H),1.99(td,J=13.8,7.2Hz,1H),1.78(td,J=14.0,6.6Hz,1H),1.36–1.27(m,2H),0.75(t,J=7.4Hz,3H)。
Example 38
Synthesis of (R) -3- (7- (3- (dimethylamino) phenoxy) -8- (2- ((4-fluorobenzyl) oxa) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-propylpropionamide (38)
Figure GDA0001986101440000851
Prepared according to the procedure described for the synthesis of compound 6 in example 6, using 15 and n-propylamine, compound 38 was obtained in 49% yield.1H NMR(400MHz,DMSO-d6)10.40(s,1H),9.36(s,1H),8.43(d,J=5.2Hz,1H),8.15(s,1H),7.77(t,J=5.5Hz,1H),7.37-7.34(m,2H),7.23-7.19(m,1H),7.12(s,1H),7.08-7.03(m,2H),6.58(dd,J=8.4,2.1Hz,1H),6.43-6.42(m,1H),6.26(dd,J=7.9,2.0Hz,1H),4.57(s,2H),4.23–4.15(m,2H),3.68-3.63(m,1H),2.96-2.91(m,2H),2.89(s,6H),2.20-2.16(m,2H),1.99(td,J=13.7,6.6Hz,1H),1.78(td,J=14.9,7.6Hz,1H),1.36-1.27(m,2H),0.75(t,J=7.4Hz,3H)。
Example 39
Synthesis of (R) -2- (benzyloxy) -N- (7- (3- (dimethylamino) phenoxy) -3- (3- (4-methylpiperazin-1-yl) -3-propionyl) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) acetamide (39)
Figure GDA0001986101440000852
Prepared according to the procedure described for the synthesis of compound 6 in example 6, using 5 and N-methylpiperazine, compound 39 was obtained in 42% yield.1H NMR(400MHz,DMSO-d6)10.40(s,1H),9.39(s,1H),8.43-8.42(m,1H),8.16(s,1H),7.33–7.20(m,6H),7.12(s,1H),6.58(d,J=9.2Hz,1H),6.44(s,1H),6.29(d,J=7.9Hz,1H),4.59(s,2H),4.24–4.15(m,2H),3.73-3.68(m,1H),3.38(d,J=0.8Hz,4H),2.89(s,6H),2.41-2.38(m,2H),2.23–2.14(m,7H),2.01–1.96(m,1H),1.82–1.75(m,1H)。
Example 40
Synthesis of (R) -2- (benzyloxy) -N- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-3- (3-oxo-3- (piperazin-1-yl) propyl) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) acetamide (40)
Figure GDA0001986101440000861
Prepared according to the method for the synthesis of compound 6 described in example 6, using 5 and piperazine, compound 40 was obtained in 35% yield.1H NMR(400MHz,DMSO-d6)10.39(s,1H),9.39(s,1H),8.42(d,J=4.2Hz,1H),8.16(s,1H),7.31-7.13(m,6H),7.13(s,1H),6.58(dd,J=7.6,1.5Hz,1H),6.44(s,1H),6.30–6.28(m,1H),4.59(s,2H),4.24–4.15(m,2H),3.72-3.70(m,1H),3.43-3.38(m,4H),2.89(s,6H),2.62-2.58(m,4H),2.40–2.36(m,2H),2.02-1.97(m,2H),1.82–1.76(m,1H)。
EXAMPLE 41
Synthesis of methyl (R) -methyl 3- (8- (2- ((2, 4-dichlorobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (41)
Figure GDA0001986101440000862
Step (a): synthesis of 2- ((2, 4-dichlorobenzyl) oxy) acetic acid (41-a)
Figure GDA0001986101440000863
According to the method for synthesizing the compound 1-e described in example 1,2, 4-dichlorobenzyl alcohol and 2-bromoacetic acid were reacted to obtain 41-a with a yield of 85%.
Step (b): synthesis of the title Compound 41
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 41-a, compound 41 was obtained in 53% yield.1H NMR(400MHz,DMSO-d6)10.43(s,1H),9.38(s,1H),8.44(d,J=5.6Hz,1H),8.15(s,1H),7.58(s,1H),7.48(d,J=8.7Hz,1H),7.26(d,J=7.4Hz,1H),7.21-7.17(m,1H),7.13(s,1H),6.56(dd,J=8.0,0.8Hz,1H),6.40(d,J=0.7Hz,1H),6.24(dd,J=7.2,0.8Hz,1H),4.66(s,2H),4.28(s,2H),3.74-3.69(m,1H),3.56(s,3H),2.88(s,6H),2.43–2.40(m,2H),2.05–1.99(m,1H),1.84–1.7(m,1H)。
Example 42
Synthesis of methyl (R) -methyl 3- (8- (2- ((3, 4-dichlorobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (42)
Figure GDA0001986101440000871
Step (a): synthesis of 2- ((3, 4-dichlorobenzyl) oxy) acetic acid (42-a)
Figure GDA0001986101440000872
According to the method for synthesizing the compound 1-e described in example 1,3, 4-dichlorobenzyl alcohol and 2-bromoacetic acid were reacted to obtain 42-a in a yield of 68%.
Step (b): synthesis of the title Compound 42
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 42-a, yield 95%.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.39(s,1H),8.44(d,J=4.4Hz,1H),8.14(s,1H),7.60(s,1H),7.49(d,J=8.3Hz,1H),7.31(dd,J=8.3,1.1Hz,1H),7.22-7.17(m,1H),7.13(s,1H),6.56(dd,J=8.3,1.8Hz,1H),6.41-6.40(m,1H),6.26(dd,J=8.1,1.7Hz,1H),4.60(s,2H),4.27–4.19(m,2H),3.76-3.69(m,1H),3.56(s,3H),2.88(s,6H),2.44–2.40(m,2H),2.06–1.99(m,1H),1.84-1.79(m,1H)。
Example 43
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-8- (2- (thiophene-3-methoxy) acetamido) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (43)
Figure GDA0001986101440000881
Step (a): synthesis of 2- (thiophene-3-methoxy) acetic acid (43-a)
Figure GDA0001986101440000882
3-Methanothiophene and 2-bromoacetic acid were reacted according to the synthesis of compound 1-e described in example 1 to give 43-a in 62% yield.
Step (b): synthesis of the title Compound 43
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 43-a, compound 43 is obtained in 63% yield.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.33(s,1H),8.44(d,J=5.3Hz,1H),8.16(s,1H),7.46(dd,J=4.9,2.9Hz,1H),7.40-7.39(m,1H),7.23-7.19(m,1H),7.14(s,1H),7.06(dd,J=4.9,1.0Hz,1H),6.58(dd,J=8.4,2.2Hz,1H),6.46-6.45(m,1H),6.28(dd,J=7.9,2.0Hz,1H),4.59(s,2H),4.21-4.13(m,2H),3.75-3.70(m,1H),3.56(s,3H),2.91(s,6H),2.44–2.40(m,2H),2.03(dt,J=15.0,6.7Hz,1H),1.82(td,J=14.5,8.0Hz,1H)。
Example 44
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (2- (furan-3-methoxy) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (44)
Figure GDA0001986101440000883
Step (a): synthesis of 2- (Furan-3-methoxy) acetic acid (111)
Figure GDA0001986101440000884
According to the method for synthesizing the compound 1-e described in example 1, 3-methanofuran and 2-bromoacetic acid were reacted to obtain 44-a with a yield of 73%.
Step (b): synthesis of the title Compound 44
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 44-a, compound 44 was obtained in 63% yield.1H NMR(400MHz,DMSO-d6)10.41(s,1H),9.28(s,1H),8.43(d,J=5.3Hz,1H),8.15(s,1H),7.64-7.59(m,2H),7.23-7.19(m,1H),7.12(s,1H),6.58(dd,J=8.4,2.1Hz,1H),6.45-6.44(m,2H),6.28(dd,J=7.9,1.9Hz,1H),4.46(s,2H),4.18–4.10(m,2H),3.74-3.69(m,1H),3.56(s,3H),2.90(s,6H),2.44–2.39(m,2H),2.03(td,J=14.6,6.7Hz,1H),1.81(td,J=15.0,8.0Hz,1H)。
Example 45
Synthesis of methyl (S) -methyl 3- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-8- (2- ((4- (trifluoromethyl) benzyl) oxa) acetamido) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (45)
Figure GDA0001986101440000891
Step (a): synthesis of (S) -dimethyl-2- (5- (3- (dimethylamino) phenoxy) -2, 4-dinitrobenzoylamino) pentanediacid ester (45-a)
Figure GDA0001986101440000892
Compound 1-b was reacted with L-glutamic acid dimethyl ester hydrochloride according to the method for the synthesis of compound 1-c described in example 1 to give compound 45-a in 55% yield.
Step (b): synthesis of methyl (S) -methyl 3- (8-amino-7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (45-b)
Figure GDA0001986101440000893
Compound 45-a was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 45-b in 30% yield.
Step (c): synthesis of the title Compound 45
Prepared according to the synthesis of compound 1 described in example 1, using 45-b and 32-a, in 65% yield.1H NMR(400MHz,DMSO-d6)10.44(s,1H),9.42(s,1H),8.45(d,J=4.7Hz,1H),8.16(s,1H),7.57(d,J=8.2Hz,2H),7.52(d,J=7.9Hz,2H),7.22-7.18(m,1H),7.15(s,1H),6.57(dd,J=8.1,1.1Hz,1H),6.44(s,1H),6.27–6.26(m,1H),4.70(s,2H),4.30–4.22(m,2H),3.75-3.70(m,1H),3.56(s,3H),2.88(s,6H),2.44-2.40(m,2H),2.03(td,J=14.7,7.6Hz,1H),1.86–1.79(m,1H)。
Example 46
Synthesis of (S) -3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-propylpropanamide (46)
Figure GDA0001986101440000901
Step (a): (S) -methyl 3- (8- (2- (benzyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (46-a)
Figure GDA0001986101440000902
Prepared according to the synthesis method of the compound 1 in the example 1 by adopting 45-b and 5-a, and the compound 46-a is obtained with the yield of 100 percent.
Step (b): synthesis of the title Compound 46
Prepared according to the procedure described for the synthesis of compound 6 in example 6, using 46-a and n-propylamine, compound 46 was obtained in 75% yield.1H NMR(400MHz,DMSO-d6)10.40(s,1H),9.38(s,1H),8.43(d,J=4.9Hz,1H),8.17(s,1H),7.77(t,J=4.9Hz,1H),7.33–7.26(m,5H),7.23-7.19(m,1H),7.12(s,1H),6.58(d,J=8.2Hz,1H),6.44(s,1H),6.28(d,J=7.9Hz,1H),4.59(s,2H),4.24–4.15(m,2H),3.68-3.63(m,1H),2.96-2.91(m,2H),2.89(s,6H),2.20-2.17(m,2H),2.02-1.97(m,1H),1.78(td,J=14.4,7.5Hz,1H),1.34-1.29(m,2H),0.75(t,J=7.4Hz,3H)。
Example 47
Synthesis of methyl (S) -methyl 3- (8- (2- ((2-chloro-4-fluorobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (47)
Figure GDA0001986101440000911
According to the synthesis of Compound 1 described in example 1, thePrepared from 45-b and 17-a to give 47 in 31% yield.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.37(s,1H),8.44(d,J=4.2Hz,1H),8.15(s,1H),7.54-7.50(m,1H),7.42-7.40(m,1H),7.21-7.17(m,1H),7.13(s,1H),7.10-7.06(m,1H),6.56(d,J=7.9Hz,1H),6.39(s,1H),6.25(d,J=7.6Hz,1H),4.65(s,2H),4.31–4.22(m,2H),3.74-3.69(m,1H),3.56(s,3H),2.88(s,6H),2.46–2.37(m,2H),2.03(td,J=13.2,6.5Hz,1H),1.86–1.77(m,1H)。
Example 48
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (2- (naphthylacetamide-2-methoxy) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (48)
Figure GDA0001986101440000912
Step (a): synthesis of 2- (Naphthylacetamide-2-methoxy) acetic acid (48-a)
Figure GDA0001986101440000913
According to the synthesis method of the compound 1-e described in example 1, naphthalenemethanol and 2-bromoacetic acid were reacted to give 48-a with a yield of 71%.
Step (b): synthesis of the title Compound 48
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 48-a, compound 48 was obtained in 47% yield.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.45(s,1H),8.43(d,J=5.2Hz,1H),8.18(s,1H),7.87–7.79(m,3H),7.69-7.67(m,1H),7.51–7.45(m,3H),7.23-7.19(m,1H),7.13(s,1H),6.59(d,J=8.5Hz,1H),6.44(s,1H),6.31(d,J=8.0Hz,1H),4.78(s,2H),4.31–4.23(m,2H),3.74-3.69(m,1H),3.56(s,3H),2.85(s,6H),2.44–2.39(m,2H),2.03(dt,J=14.8,6.8Hz,1H),1.81(dt,J=15.1,7.3Hz,1H)。
Example 49
Synthesis of methyl (R) -methyl 3- (2, 5-dioxo-7-phenoxy-8- (2- ((4- (trifluoromethyl) benzyl) oxa) acetamido) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (49)
Figure GDA0001986101440000921
Step (a): synthesis of 2, 4-dinitro-5-phenoxybenzoic acid (49-a)
Figure GDA0001986101440000922
Compound 1-a was reacted with phenol according to the synthesis method of compound 1-b described in example 1 to give compound 49-a with a yield of 100%.
Step (b): synthesis of (R) -dimethyl-2- (2, 4-dinitro-5-phenoxybenzamido) pentanedioate (49-b)
Figure GDA0001986101440000923
Compound 49-a was reacted with D-glutamic acid dimethyl ester hydrochloride according to the method for synthesizing compound 1-c described in example 1 to obtain compound 49-b with a yield of 100%.
Step (c): synthesis of (R) -methyl 3- (8-amino-2, 5-dioxo-7-phenoxy) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate methyl ester (49-c)
Figure GDA0001986101440000924
Compound 49-b was reacted with iron powder according to the synthesis of compound 1-d described in example 1 to give compound 49-c in 18% yield.
Step (d): synthesis of the title Compound 49
Prepared according to the method for the synthesis of compound 1 described in example 1, using 49-c and 32-a, in 46% yield.1H NMR(400MHz,DMSO-d6)10.46(s,1H),9.44(s,1H),8.46(d,J=4.9Hz,1H),8.16(s,1H),7.60(d,J=7.9Hz,2H),7.52(d,J=7.9Hz,2H),7.44-7.40(m,2H),7.23-7.20(m,1H),7.14(s,1H),7.08-7.06(m,2H),4.68(s,2H),4.29–4.20(m,2H),3.76-3.71(m,1H),3.56(s,3H),2.44-2.40(m,2H),2.05–1.97(m,1H),1.82(td,J=14.2,7.2Hz,1H)。
Example 50
Synthesis of methyl (R) -methyl 3- (8- (2- ((4- (tert-butyl) benzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (50)
Figure GDA0001986101440000931
Step (a): synthesis of 2- ((4- (tert-butyl) benzyl) oxy) acetic acid (50-a)
Figure GDA0001986101440000932
P-tert-butylbenzyl alcohol and 2-bromoacetic acid were reacted according to the method for synthesizing the compound 1-e described in example 1 to obtain 50-a with a yield of 64%.
Step (b): synthesis of the title Compound 50
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 50-a, compound 50 was obtained in 49% yield.1H NMR(400MHz,DMSO-d6)10.44(s,1H),9.41(s,1H),8.45(d,J=5.2Hz,1H),8.18(s,1H),7.25-7.18(m,5H),7.13(s,1H),6.60(dd,J=8.4,1.7Hz,1H),6.48(s,1H),6.31(dd,J=8.0,1.1Hz,1H),4.55(s,2H),4.23–4.15(m,2H),3.74-3.69(m,1H),3.56(s,3H),2.90(s,6H),2.44–2.39(m,2H),2.05–1.98(m,1H),1.82(dt,J=14.4,7.2Hz,1H),1.20(s,9H)。
Example 51
Synthesis of (R) -3- (8- (2- (phenoxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N-propanoic acid butanamide (51)
Figure GDA0001986101440000941
Prepared according to the method for the synthesis of compound 6 described in example 6, using 5 and n-butylamine, compound 51 was obtained in 37% yield.1H NMR(400MHz,DMSO-d6)10.41(s,1H),9.39(s,1H),8.44(d,J=4.7Hz,1H),8.18(s,1H),7.77(t,J=4.8Hz,1H),7.32–7.20(m,6H),7.14(s,1H),6.59(d,J=7.9Hz,1H),6.44(s,1H),6.29(d,J=7.4Hz,1H),4.60(s,2H),4.25–4.16(m,2H),3.67–3.63(m,1H),2.98(m,2H),2.90(s,6H),2.18(t,J=6.2Hz,2H),2.00(dd,J=13.1,6.7Hz,1H),1.79(dd,J=13.8,6.9Hz,1H),1.31–1.24(m,2H),1.20-1.15(m,2H),0.79(t,J=7.1Hz,3H)。
Example 52
Synthesis of methyl (R) -methyl 3- (8- (2- ((4-chlorobenzyl) oxa) acetamido) -7- (3- (dimethylamino) phenoxy) -1-methyl-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (52)
Figure GDA0001986101440000942
Dissolve compound 3(100mg,0.168mmol) in DMF and add K2CO3(45mg,0.326mmol) and iodomethane (10 μ L, 0.168mmol) were stirred at room temperature overnight until the starting material disappeared, the solvent was evaporated and column chromatography was carried out to give compound 52 in 37% yield.1HNMR(400MHz,DMSO-d6)9.48(s,1H),8.57(d,J=5.5Hz,1H),8.31(s,1H),7.35(d,J=7.9Hz,2H),7.29(d,J=8.4Hz,2H),7.25-7.21(m,1H),7.05(s,1H),6.60(dd,J=7.8,1.2Hz,1H),6.45(s,1H),6.31(dd,J=7.5,0.7Hz,1H),4.60(s,2H),4.22(s,2H),3.80-3.75(m,1H),3.55(s,3H),3.28(s,3H),2.90(s,6H),2.41-2.36(m,2H),2.06–1.98(m,1H),1.89–1.82(m,1H)。
Example 53
Preparation of methyl (R) -methyl 3- (8- (2- (allyloxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (53)
Figure GDA0001986101440000951
Step (a): synthesis of 2- (allyloxy) acetic acid (53-a)
Figure GDA0001986101440000952
Allyl alcohol and 2-bromoacetic acid were reacted according to the synthesis of compound 1-e described in example 1 to give 53-a.
Step (b): synthesis of the title Compound 53
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 53-a, compound 53 was obtained in 78% yield.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.28(s,1H),8.43(d,J=5.0Hz,1H),8.16(s,1H),7.22-7.18(m,1H),7.13(s,1H),6.57(d,J=8.3Hz,1H),6.44(s,1H),6.27(d,J=7.7Hz,1H),5.86(ddd,J=22.0,10.4,5.2Hz,1H),5.26(d,J=17.2Hz,1H),5.14(d,J=10.4Hz,1H),4.13(d,J=1.6Hz,2H),4.05(d,J=4.9Hz,2H),3.74-3.70(m,1H),3.56(s,3H),2.90(s,6H),2.44-2.37(m,2H),2.03(dt,J=14.7,7.6Hz,1H),1.81(td,J=14.7,7.7Hz,1H)。
Example 54
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-8- (4-phenylbutanolamine) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (54)
Figure GDA0001986101440000953
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and phenylbutyric acid, compound 54 was obtained in 83% yield.1H NMR(400MHz,DMSO-d6)10.33(s,1H),9.64(s,1H),8.40(d,J=5.0Hz,1H),7.99(s,1H),7.29-7.25(m,2H),7.21-7.17(m,4H),7.09(s,1H),6.54(d,J=8.2Hz,1H),6.42(s,1H),6.27(d,J=7.7Hz,1H),3.73-3.68(m,1H),3.56(s,3H),2.88(s,6H),2.57(t,J=7.6Hz,2H),2.46–2.43(m,4H),2.05-2.00(m,1H),1.87-1.80(m,3H)。
Example 55
Synthesis of methyl (R, E) -methyl 3- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-8- (3- (4- (trifluoromethyl) phenyl) acrylamide) -2,3,4, 5-tetrahydro-1H-benzo [ E ] [1,4] diazepin-3-yl) propanoate (55)
Figure GDA0001986101440000961
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and p-trifluoromethylcinnamic acid to give compound 55 with a yield of 37.8%.1H NMR(400MHz,DMSO-d6)10.42(s,1H),9.99(s,1H),8.42(d,J=4.8Hz,1H),8.25(s,1H),7.86-7.80(m,4H),7.69(d,J=15.7Hz,1H),7.45(d,J=15.7Hz,1H),7.23(t,J=8.2Hz,1H),7.08(s,1H),6.59(d,J=8.7Hz,1H),6.50(s,1H),6.35(d,J=8.0Hz,1H),3.75-3.71(m,1H),3.56(s,3H),2.91(s,6H),2.44-2.42(m,2H),2.08–1.9(m,1H),1.82(dt,J=21.6,7.4Hz,1H)。
Example 56
Synthesis of 3- ((R) -8- (2- (phenoxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) -N- (2-hydroxypropyl) propionamide (56)
Figure GDA0001986101440000962
Prepared according to the procedure described for the synthesis of compound 6 in example 6, using 5 and 2-hydroxypropylamine, compound 56 is obtained in 99% yield.1H NMR(400MHz,DMSO-d6)10.40(s,1H),9.38(s,1H),8.43(d,J=4.9Hz,1H),8.17(s,1H),7.76(t,J=5.5Hz,1H),7.32(d,J=7.0Hz,2H),7.27-7.20(m,4H),7.13(s,1H),6.58(d,J=8.0Hz,1H),6.44(s,1H),6.29(d,J=7.9Hz,1H),4.60(s,2H),4.24–4.16(m,2H),3.67(dd,J=12.6,5.9Hz,1H),3.60–3.53(m,1H),2.93(d,J=6.4Hz,2H),2.89(s,6H),2.21(t,J=6.9Hz,2H),1.99(dd,J=13.1,6.2Hz,1H),1.78(td,J=14.4,6.9Hz,1H),0.93(d,J=5.8Hz,3H)。
Example 57
Synthesis of (R) -N- (7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-3- (3-oxo-3- (piperidin-1-yl) propyl) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (57)
Figure GDA0001986101440000971
Prepared according to the procedure for the synthesis of compound 6 described in example 6, using 32 and piperidine, compound 57 is obtained in 76% yield.1H NMR(400MHz,DMSO-d6)10.41(s,1H),9.42(s,1H),8.43(d,J=4.9Hz,1H),8.16(s,1H),7.57(d,J=8.0Hz,2H),7.52(d,J=8.0Hz,2H),7.20(t,J=8.2Hz,1H),7.14(s,1H),6.57(d,J=8.6Hz,1H),6.44(s,1H),6.26(d,J=8.0Hz,1H),4.70(s,2H),4.30–4.22(m,2H),3.71(dd,J=12.4,6.4Hz,1H),3.35(s,4H),2.88(s,6H),2.39(t,J=7.2Hz,2H),2.04–1.95(m,1H),1.83–1.75(m,1H),1.53(d,J=4.8Hz,2H),1.43(s,2H),1.36(s,2H)。
Example 58
Synthesis of methyl (R, E) -methyl 3- (8- (3- (4-chloro-2-fluorophenyl) acrylamide) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ E ] [1,4] diazepin-3-yl) propionate (58)
Figure GDA0001986101440000972
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 4-chloro-2-fluorocinnamic acid to give compound 58 in 44% yield.1H NMR(400MHz,DMSO-d6)10.41(s,1H),10.04(s,1H),8.42(d,J=5.0Hz,1H),8.22(s,1H),7.74(t,J=8.2Hz,1H),7.59(dd,J=20.5,13.3Hz,2H),7.41–7.37(m,2H),7.22(t,J=8.1Hz,1H),7.08(s,1H),6.58(d,J=8.1Hz,1H),6.48(s,1H),6.34(d,J=8.0Hz,1H),3.73(dd,J=12.4,6.5Hz,1H),3.56(s,3H),2.91(s,6H),2.42(s,2H),2.08–1.96(m,1H),1.81(td,J=15.2,8.0Hz,1H)。
Example 59
Synthesis of methyl (R, E) -methyl 3- (8- (butyl-2-enamide) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ E ] [1,4] diazepin-3-yl) propanoate (59)
Figure GDA0001986101440000981
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and crotonic acid, compound 59 was obtained in 61% yield.1H NMR(400MHz,DMSO-d6)10.36(s,1H),9.67(s,1H),8.40(d,J=5.2Hz,1H),8.09(s,1H),7.20(t,J=8.2Hz,1H),7.07(s,1H),6.88–6.79(m,1H),6.55(d,J=8.4Hz,1H),6.46(d,J=15.7Hz,2H),6.29(d,J=7.0Hz,1H),3.73-3.69(m,1H),3.55(s,3H),2.89(s,6H),2.40(dd,J=11.2,6.2Hz,2H),2.01(tt,J=13.5,6.7Hz,2H),1.84(d,J=6.9Hz,3H).
Example 60
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (3-methyl-2-enebutenamide) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (60)
Figure GDA0001986101440000982
Prepared according to the method for the synthesis of compound 1 described in example 1, using 1-d and 3-methyl-2-butenoic acid to give compound 60 in 48% yield.1H NMR(400MHz,DMSO-d6)10.29(s,1H),9.45(s,1H),8.35(d,J=4.8Hz,1H),8.10(s,1H),7.21-7.18(m,1H),7.07(s,1H),6.55(d,J=8.0Hz,1H),6.44(s,1H),6.28(d,J=7.5Hz,1H),6.20(s,1H),3.71–3.69(m,1H),3.56(s,3H),2.90(s,6H),2.41-2.40(m,2H),2.15(s,3H),2.03(dt,J=11.3,6.9Hz,1H),1.85(s,3H),1.82-1.77(m,1H)。
Example 61
Synthesis of (R) -N- (7- (3- (dimethylamino) phenoxy) -3- (3- (4-fluoropiperidin-1-yl) -3-propionyl) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (61)
Figure GDA0001986101440000991
Prepared according to the synthetic method for compound 6 described in example 6, using 32 and 4-fluoropiperidine hydrochloride, compound 61 is obtained in 36.2% yield.1H NMR(400MHz,DMSO-d6)10.38(s,1H),9.40(s,1H),8.40(s,1H),8.15(s,1H),7.57(d,J=7.7Hz,2H),7.52(d,J=7.2Hz,2H),7.18(dd,J=16.9,8.7Hz,2H),6.57(d,J=8.3Hz,1H),6.43(s,1H),6.26(d,J=7.6Hz,1H),4.70(s,2H),4.29–4.21(m,2H),3.73-3.72(m,1H),3.55-3.51(m,2H),3.43–3.37(m,3H),2.87(s,6H),2.43(t,J=6.4Hz,2H),2.03–1.99(m,3H),1.81-1.79(m,3H)。
Example 62
Synthesis of (R) -N- (3- (3- (4, 4-difluoropiperidin-1-yl) -3-propionyl) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) -2- ((4- (trifluoromethyl) benzyl) oxa) acetamide (62)
Figure GDA0001986101440000992
Prepared according to the procedure described for the synthesis of compound 6 in example 6, using 32 and 4, 4-difluoropiperidine hydrochloride, compound 62 was obtained in 20% yield.1H NMR(400MHz,DMSO-d6)10.39(s,1H),9.40(s,1H),8.39(d,J=3.3Hz,1H),8.15(s,1H),7.57(d,J=7.5Hz,2H),7.52(d,J=7.1Hz,2H),7.18(dd,J=16.6,8.2Hz,2H),6.57(d,J=8.0Hz,1H),6.43(s,1H),6.26(d,J=7.7Hz,1H),4.70(s,2H),4.30–4.21(m,2H),3.75-3.71(m,1H),3.52-3.51(m,4H),2.87(s,6H),2.47(s,2H),2.04–1.97(m,3H),1.88–1.78(m,3H)。
Example 63
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (N-methyl-2- ((4- (trifluoromethyl) benzyl) oxy) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (63)
Figure GDA0001986101440001001
Step (a): synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (methylamino) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (63-a)
Figure GDA0001986101440001002
Dissolving compound 1-d (50mg, 1eq) in DCM, adding 37% formaldehyde solution (3.3 μ L,1eq) and catalytic amount of AcOH in sequence, reacting at room temperature for 2 hours, adding NaBH3CN (38.1mg, 5eq) continues to react for 1 hour, water is added to quench the reaction after the reaction is completed, DCM is used for extraction, and column chromatography separation is carried out to obtain 45mg of a compound 63-a with the yield of 87%.
Step (b): synthesis of the title Compound 63
Prepared according to the method for synthesizing compound 1 described in example 1 using 63-a and 32-a, yield 23.22%.1HNMR(400MHz,DMSO-d6)10.39(s,1H),9.40(s,1H),8.39(d,J=3.3Hz,1H),8.15(s,1H),7.57(d,J=7.5Hz,2H),7.52(d,J=7.1Hz,2H),7.18(dd,J=16.6,8.2Hz,2H),6.57(d,J=8.0Hz,1H),6.43(s,1H),6.26(d,J=7.7Hz,1H),4.70(s,2H),4.30–4.21(m,2H),3.75-3.71(m,1H),3.52-3.51(m,4H),2.87(s,6H),2.47(s,2H),2.04–1.97(m,3H),1.88–1.78(m,3H)。
Example 64
Synthesis of (R) -N- (3- (3-amino-3-propionyl) -7- (3, 5-dimethylphenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) -3,4, 5-trimethoxybenzamide (64)
Figure GDA0001986101440001011
Step (a): synthesis of 5- (3, 5-dimethylphenoxy) -2, 4-dinitrobenzoic acid (64-a)
Figure GDA0001986101440001012
1.66g (1.0eq) of the compound 1-a, 1.2g (2.2eq) of NaHCO30.87g (1.09eq) of 3, 5-dimethylphenol is dissolved in 25m L of distilled water, the mixture is heated and refluxed at 110 ℃ for 2 hours, then the mixture is cooled to room temperature, the pH value of a solution system is adjusted to 1-2 by hydrochloric acid under the ice bath condition, a large amount of light yellow solid is precipitated, the mixture is filtered and dried by suction to obtain the light yellow solid, the light yellow solid is recrystallized by using a mixed solvent of EtOH and distilled water (the volume ratio is 1:2), the crystals are fully precipitated from a mother solution, and then the mixture is filtered and dried by suction to obtain 1.8 g of light yellow solid, namely the target product (64-a), the yield is 81.0 percent, and the m.p. ═ 194 and 196 ℃.
Step (b): synthesis of Compound 64-b
Figure GDA0001986101440001013
3.32g (1.0eq) of compound 64-a, 2.11g (1.0eq) of HCl, H-D-Glu (OMe) -OMe and 2.36m L (1.5eq) DIC were dissolved in 100m L THF and stirred at room temperature for 4 hours3The DCM phase was extracted with aqueous and distilled water, collected and evaporated to dryness under reduced pressure to give 4.25g of a yellow oil, the title product (64-b) in 87.0% yield.
Step (c): synthesis of Compound 64-c
Figure GDA0001986101440001021
4.25g of compound 64-b, 4.25g Pd/C and 8.5g HCOONH4Dissolving in 100m of mixed solvent of L THF and EtOH in equal proportion, stirring at room temperature for 2 hours, filtering to remove Pd/C after reaction, evaporating the filtrate under reduced pressure, diluting the residue with DCM, and then sequentially diluting with dilute hydrochloric acid and saturated NaHCO3The aqueous solution and distilled water were used to extract the DCM phase, the DCM phase was collected and evaporated to dryness under reduced pressure, the residue was dissolved in 100m L AcOH and heated under reflux at 90 ℃ for 5 hours, after the reaction was complete, the solvent was evaporated to dryness under reduced pressure to obtain a brownish red solid, which was purified by silica gel column chromatography to obtain 1.79g of a yellow solid, i.e. the target product (64-c), with a yield of 52.0%.
A step (d); synthesis of Compound 64-d
Figure GDA0001986101440001022
Dissolving 1.79mg (1.0eq) of compound 64-c in 100m L MeOH, dropwise adding L iOH saturated aqueous solution, adjusting the pH value of the solution system to 13-14, stirring at room temperature for 20 minutes, after the reaction is finished, adjusting the pH value of the solution system to 6-7 by using 2N hydrochloric acid, evaporating the solvent under reduced pressure, dissolving the residue in THF, sequentially adding 1.07m L (1.5eq) DIC and 779mg (1.5eq) HOSu, reacting at room temperature for 12 hours, filtering to remove insoluble matters (DIU), introducing dry ammonia gas into the filtrate under ice bath conditions for 30 minutes, after the reaction is finished, evaporating the solvent under reduced pressure, and purifying the residue by silica gel column chromatography to obtain 741mg of yellow solid, namely the target product (64-d), wherein the yield is 43.0%.
1H-NMR(300MHz,DMSO-d6):5.72(2H,s,1-H),7.00(1H,s,2-H),6.42(1H,s,3-H),6.58(2H,s,4and 5-H),7.33(1H,s,6-H),2.38(6H,s,7and 8-H),8.06(1H,d,J=5.4Hz,9-H),3.62(1H,m,10-H),10.17(1H,s,11-H),1.74(1H,s,12-Ha),1.95(1H,s,12-Hb),2.16(2H,m,13-H),6.74(2H,s,14-H).
ESI-MS:383.04[M+H]+.
HR-MS(TOF):observed for 383.1724[M+H]+,calculated for 383.1720
[M+H]+,C20H22N4O4.
A step (e): synthesis of the title Compound 64
50mg (1.0eq) of the compound 64-d is dissolved in 10m L THF, 36.1 mu L (2.0eq) TEA is added, 36mg (1.2eq) of 3,4, 5-trimethoxy-benzenecarbonoyl chloride is added into the solution system under ice bath condition, the solution is heated and refluxed at 70 ℃ for 2 hours, after the reaction is finished, the solvent is evaporated, and the residue is purified by silica gel column chromatography to obtain 24mg of white solid, namely the target product (64) with the yield of 32%.
1H-NMR(300MHz,DMSO-d6):9.84(1H,s,1-H),7.25(1H,s,2-H),6.77(1H,s,3-H),6.54(2H,s,4and 5-H),7.58(1H,s,6-H),2.22(6H,s,7and 8-H),8.51(1H,d,J=5.4Hz,9-H),3.71(1H,m,10-H),10.47(1H,s,11-H),1.82(1H,s,12-Ha),2.04(1H,s,12-Hb),2.16(2H,m,13-H),6.77(1H,s,14-Ha),7.28(1H,s,14-Hb),7.10(2H,s,15and 16-H),3.80(6H,s,17and19-H),3.71(3H,s,18-H).
ESI-MS:577.53[M+H]+,1153.30[2M+H]+.
HR-MS(TOF):observed for 577.2303[M+H]+,calculated for 577.2293[M+H]+,C30H32N4O8.
Example 65
Synthesis of (R) -N- (3- (3-amino-3-propionyl) -7-methoxy-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) butanamide (65)
Figure GDA0001986101440001031
Step (a): synthesis of 5-methoxy-2, 4-dinitrobenzoic acid (65-a)
Figure GDA0001986101440001032
3.0g (1.0eq) of the compound 1-a and 2.4g (3.5eq) of KOH are dissolved in 40m L MeOH, the reaction is carried out at room temperature for 12 hours, then the reaction is finished, the solvent is evaporated under reduced pressure, the residue is diluted by 50m L distilled water, the pH value of the solution system is adjusted to 1-2 by hydrochloric acid under the ice bath condition, a large amount of light yellow solid is precipitated, the light yellow solid is obtained by filtration and pumping drying, the light yellow solid is recrystallized by 60m L distilled water, the crystals are fully precipitated, and then the light yellow solid is filtered and pumped drying, so that 2.6 g of light yellow needle-shaped crystals are obtained, namely the target product 65-a, the yield is 85.0%, and the m.p. is 188-.
Step (b): synthesis of Compound 65-b
Figure GDA0001986101440001041
2.42g (1.0eq) of compound 65-a, 2.11g (1.0eq) of HCl, H-D-Glu (OMe) -OMe and 2.36m L (1.5eq) DIC were dissolved in 100m L THF and stirred at room temperature for 4 hours3The DCM phase was extracted with aqueous and distilled water, collected and evaporated to dryness under reduced pressure to give 3.63g of a yellow oil, the title product (65-b) in 91.0% yield.
Step (c): synthesis of Compound 65-c
Figure GDA0001986101440001042
3.63g of compound 65-b, 3.63g Pd/C and 7.26g HCOONH4Dissolved in 100m L THF and EtOH, mixing the solvent in equal proportion, and stirring the mixture for 2 hours at room temperature. After the reaction, Pd/C was removed by filtration, the filtrate was evaporated to dryness under reduced pressure, and the residue was diluted with DCM, then diluted with dilute hydrochloric acid, and saturated NaHCO in this order3The aqueous solution and distilled water were used to extract the DCM phase, the DCM phase was collected and evaporated to dryness under reduced pressure, the residue was dissolved in 100m L AcOH and heated under reflux at 90 ℃ for 5 hours, after the reaction was complete, the solvent was evaporated to dryness under reduced pressure to give a brownish red solid which was purified by silica gel column chromatography to give 1.54g of a yellow solid, the target product (65-c), with a yield of 55.0%.
Step (d): synthesis of Compound 65-d
Figure GDA0001986101440001051
Dissolving 1.54mg (1.0eq) of compound 65-c in 100m L MeOH, dropwise adding L iOH saturated aqueous solution, adjusting the pH value of the solution system to 13-14, stirring at room temperature for 20 minutes, after the reaction is finished, adjusting the pH value of the solution system to 6-7 by using 2N hydrochloric acid, evaporating the solvent under reduced pressure, dissolving the residue in DMF, sequentially adding 1.55g of DCC (1.5eq) and 866mg of HOSu (1.5eq), reacting at room temperature for 12 hours, filtering to remove insoluble matters (DCU), introducing dry ammonia gas into the filtrate under ice bath conditions for 30 minutes, after the reaction is finished, evaporating the solvent under reduced pressure, and purifying the residue by silica gel column chromatography to obtain 571mg of yellow solid, namely the target product (65-d), wherein the yield is 39.0%.
1H-NMR(300MHz,DMSO-d6):5.51(2H,s,1-H),7.27(1H,s,2-H),6.73(1H,s,3-H),3.77(3H,s,4-H),8.00(1H,d,J=4.8Hz,5-H),3.58(1H,m,6-H),9.99(1H,s,7-H),1.76(1H,m,8-Ha),1.95(1H,m,8-Hb),2.16(2H,m,9-H),6.27(1H,s,10-Ha),7.04(1H,s,10-Hb).
ESI-MS:293.14[M+H]+.
HR-MS(TOF):observed for 293.1246[M+H]+,calculated for 293.1244[M+H]+,C13H16N4O4.
A step (e): synthesis of the title Compound 65
55mg (1.0eq) of the compound 65-d is dissolved in 10m L THF, 78 mu L (3.0eq) TEA and 39 mu L (2.0eq) butyryl chloride are added into the solution system, the mixture is heated and refluxed at 70 ℃ for 2 hours, after the reaction is finished, the solvent is evaporated, and the residue is purified by silica gel column chromatography to obtain 31mg of a white solid, namely the target product (65) with the yield of 46%.
1H-NMR(300MHz,DMSO-d6):9.25(1H,s,1-H),7.99(1H,s,2-H),6.53(1H,s,3-H),3.87(3H,s,4-H),8.38(1H,d,J=5.4Hz,5-H),3.60(1H,m,6-H),10.12(1H,s,7-H),1.78(1H,m,8-Ha),1.99(1H,m,8-Hb),2.18(2H,t,J=7.2Hz,9-H),7.25(2H,s,10-H),2.42(2H,t,J=7.2Hz,11-H),1.18(2H,m,12-H),0.86(2H,t,J=7.8Hz,13-H).
ESI-MS:363.08[M+H]+,725.00[2M+H]+.
HR-MS(TOF):observed for 363.1670[M+H]+,calculated for 363.1663[M+H]+,C17H22N4O5.
Example 66
Synthesis of (R) -N- (3- (3-amino-3-propionyl) -7-methoxy-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) furan-2-carboxamide (66)
Figure GDA0001986101440001061
70mg (1.0eq) of the compound 65-d is dissolved in 10m L THF, 33.3 mu L (2.0eq) TEA and 23.5 mu L (1.0eq) 2-furoyl chloride are added into the solution system, and the mixture is heated and refluxed at 70 ℃ for 2 hours to react, after the reaction is finished, the solvent is evaporated, and the residue is purified by silica gel column chromatography to obtain 23mg of white solid, namely the target product (66) with the yield of 24%.
1H-NMR(300MHz,DMSO-d6):9.17(1H,s,1-H),7.96(1H,s,2-H),6.73(1H,s,3-H),3.93(3H,s,4-H),8.45(1H,d,J=5.1Hz,5-H),3.67(1H,m,6-H),10.31(1H,s,7-H),1.80(1H,m,8-Ha),2.00(1H,m,8-Hb),2.20(2H,t,J=7.8Hz,9-H),7.28(2H,s,10-H),7.37(1H,m,11-H),6.75(1H,m,12-H),7.98(1H,dd,J=8.1and 0.9Hz,13-H).
ESI-MS:387.18[M+H]+,773.55[2M+H]+.
HR-MS(TOF):observed for 387.1317[M+H]+,calculated for 387.1299[M+H]+,C18H18N4O6.
Example 67
Synthesis of (R) -N- (3- (3-amino-3-propionyl) -7-methoxy-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) -5-nitrofuran-2-carboxamide (67)
Figure GDA0001986101440001062
50mg (1.0eq) of the compound 65-d was dissolved in 10m L THF, 47.2. mu. L (2.0eq) TEA and 59.5mg (2.0eq) of 5-nitro-2-furoyl chloride were added to the solution system, and the mixture was heated under reflux at 70 ℃ for 2 hours.
1H-NMR(300MHz,DMSO-d6):9.83(1H,s,1-H),7.27(1H,s,2-H),6.75(1H,s,3-H),3.92(3H,s,4-H),8.50(1H,d,J=5.4Hz,5-H),3.68(1H,m,6-H),10.34(1H,s,7-H),1.81(1H,m,8-Ha),2.03(1H,m,8-Hb),2.21(2H,m,9-H),7.36(1H,s,10-Ha),7.72(1H,s,10-Hb),7.69(1H,d,J=4.2Hz,11-H),7.83(1H,d,J=4.2Hz,12-H).
ESI-MS:431.95[M+H]+.
HR-MS(TOF):observed for 432.1166[M+H]+,calculated for 432.1150[M+H]+,C18H17N5O8.
Example 68
Synthesis of methyl (R) -methyl 3- (8-acrylamido-7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (68)
Figure GDA0001986101440001071
41.2mg (1.0eq) of the compound 1-d is dissolved in 10m L DCM, 27.7 mu L (2.0eq) TEA is added, 9.7 mu L (1.2eq) acryloyl chloride is added to the solution system under ice bath conditions, stirring is carried out for 15 minutes, after the reaction is finished, the solvent is evaporated, and the residue is purified by silica gel column chromatography to obtain 18mg of light yellow solid, namely the target product (68) with the yield of 38.6%.
1H-NMR(300MHz,DMSO-d6):9.90(1H,s,1-H),7.06(1H,s,2-H),8.10(1H,s,3-H),6.45(1H,br.s,4-H),6.55(1H,d,J=8.4Hz,5-H),7.20(1H,t,J=8.4Hz,6-H),6.25(1H,d,J=8.4Hz,7-H),2.89(6H,s,8and 9-H),8.40(1H,d,J=5.1Hz,10-H),3.70(1H,m,11-H),10.36(1H,s,12-H),1.81(1H,m,13-Ha),2.05(1H,m,13-Hb),2.40(2H,t,J=7.2Hz,14-H),3.55(3H,s,15-H),6.77(1H,dd,J=10.2and 16.8Hz,16-H),5.76(1H,d,J=10.2Hz,17-Ha),6.29(1H,d,J=16.8Hz,17-Hb).
ESI-MS:467.33[M+H]+,933.09[2M+H]+.
HR-MS(TOF):observed for 467.1923[M+H]+,calculated for 467.1925[M+H]+,C24H26N4O6.
Example 69
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (4-fluorobenzamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (69)
Figure GDA0001986101440001081
70mg (1.0eq) of the compound 1-d is dissolved in 10m L DCM, 47.2 μ L (2.0eq) TEA is added, 30.5 μ L (1.5eq) 4-fluorobenzoyl chloride is added to the solution system under ice bath conditions, stirring is carried out for 20 minutes, after the reaction is finished, the solvent is evaporated, and the residue is purified by silica gel column chromatography to obtain 21mg of a yellow solid, namely the target product (69) with the yield of 23%.
1H-NMR(300MHz,DMSO-d6):9.98(1H,s,1-H),7.23(1H,s,2-H),7.72(1H,s,3-H),6.53(1H,br.s,4-H),6.60(1H,m,5-H),7.20(1H,m,6-H),6.36(1H,d,J=6.3Hz,7-H),2.89(6H,s,8and 9-H),8.48(1H,d,J=5.1Hz,10-H),3.76(1H,m,11-H),10.45(1H,s,12-H),1.85(1H,m,13-Ha),2.02(1H,m,13-Hb),2.43(2H,m,14-H),3.57(3H,s,15-H),7.93(2H,m,16and 17-H),7.34(2H,t,J=8.7Hz,18and 19-H).
ESI-MS:535.27[M+H]+1069.14[2M+H]+.
HR-MS(TOF):observed for 535.1991[M+H]+,calculated for 535.1987[M+H]+,C28H27FN4O6.
Example 70
Synthesis of methyl (R) -methyl 3- (8- (cyclopropylcarboxamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (70)
Figure GDA0001986101440001082
60mg (1.0eq) of the compound 1-d is dissolved in 10m L DCM, 41.6 mu L (2.0eq) TEA is added, 41.6 mu L (2.0eq) cyclopropylcarbonyl chloride is added to the solution system under ice bath conditions, stirring is carried out for 30 minutes, after the reaction is finished, the solvent is evaporated, and the residue is purified by silica gel column chromatography to obtain 21mg of a yellow solid, namely the target product (70) with the yield of 29.2%.
1H-NMR(300MHz,DMSO-d6):9.95(1H,s,1-H),7.99(1H,s,2-H),7.08(1H,s,3-H),6.69(1H,br.s,4-H),6.75(1H,br.s,5-H),7.27(1H,t,J=7.8Hz,6-H),6.44(1H,br.s,7-H),2.94(6H,s,8and 9-H),8.38(1H,d,J=4.8Hz,10-H),3.69(1H,m,11-H),10.31(1H,s,12-H),1.80(1H,m,13-Ha),2.02(1H,m,13-Hb),2.40(2H,t,J=7.5Hz,14-H),3.55(3H,s,15-H),1.22(1H,br.s,16-H),0.78(4H,br.s,17and 18-H).
ESI-MS:481.46[M+H]+,961.15[2M+H]+.
HR-MS(TOF):observed for 481.2084[M+H]+,calculated for 481.2082[M+H]+,C25H28N4O6.
Example 71
Synthesis of methyl (R) -methyl 3- (7-methoxy-2, 5-dioxo-8- (4- (trifluoromethyl) benzamido) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (71)
Figure GDA0001986101440001091
Weighing about 31mg (1eq) of intermediate 65-c into a 25m L round-bottom flask, adding 5m L THF for dissolving, and adding 1.2eq of 4-trifluorobenzoyl chloride and 21 mu L (1.5eq) Et sequentially under the condition of ice-water bath with stirring3And N, heating to room temperature, monitoring by L C-MS, and finishing the reaction after about 2 hours.
Adding an appropriate amount of methanol to quench the reaction, evaporating the reaction solution to dryness under reduced pressure, adding an appropriate amount of distilled water, extracting with DCM for 3 times, and collecting an organic layer to obtain a crude product of the target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-sand 300-mesh silica gel, and the mobile phase is DCM and MeOH which are 100:4, so as to obtain the pure product of the target compound.
1H NMR(400MHz,DMSO)10.40(s,1H),10.02(s,1H),8.56(d,J=5.4,1H),8.32(dd,J=14.0,6.3,2H),8.06(d,J=8.1,1H),7.86(t,J=8.1,1H),7.78(s,1H),7.42(s,1H),3.96(s,3H),3.80(dd,J=13.4,6.3,1H),3.64(s,3H),2.52(dd,J=14.8,6.4,2H),2.18–2.07(m,1H),1.92(td,J=14.5,7.9,1H).HRMS(ESI):m/z(M+H+)calcd forC22H21O6N3F,480.1377,found:480.1380.
Example 72
Synthesis of methyl (R) -methyl 3- (8- (((9H-fluoren-3-yl) methyl) amino) -7-methoxy-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (72)
Figure GDA0001986101440001101
Taking intermediate 65-c 31mg (1eq) in a 25m L round-bottom flask, adding 5m L THF for dissolving, and adding 2eq aldehyde group component and 25mg (4eq) NaBH in sequence under stirring at room temperature3CN, heating to reflux, monitoring by L C-MS, and finishing the reaction after about 24 hours.
Evaporating the reaction solution to dryness under reduced pressure, adding appropriate amount of distilled water, extracting with DCM for 3 times, and collecting organic layer to obtain crude product of target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-sand 300-mesh silica gel, and the mobile phase is DCM and MeOH which are 100:4, so as to obtain the pure product of the target compound.
1H NMR(300MHz,DMSO)9.98(s,1H),8.01(d,J=5.2,1H),7.87–7.83(m,2H),7.56(d,J=3.8,2H),7.39–7.26(m,3H),7.06(s,1H),6.45(t,J=6.1,1H),6.17(s,1H),4.47–4.30(m,2H),3.89(s,2H),3.85(s,3H),3.63–3.55(m,1H),3.53(s,3H),3.32(s,3H),2.39(dd,J=13.0,6.5,2H),2.06–1.90(m,1H),1.84–1.69(m,1H).HRMS(ESI):m/z(M+H+)calcdfor C28H28O5N3,486.2024,found:486.2021.
Example 73
Synthesis of methyl (R) -methyl 3- (8- (2-naphthylamido) -7-methoxy-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (73)
Figure GDA0001986101440001102
Prepared according to the procedure for the synthesis of compound 71 in example 71, using 65-c and β -naphthoyl chloride, to provide compound 73.
1H NMR(300MHz,DMSO)10.31(s,1H),9.76(s,1H),8.47(d,J=5.4,1H),8.21(d,J=9.1,1H),8.10–7.98(m,3H),7.77(d,J=7.0,1H),7.59(t,J=3.8,3H),7.32(s,1H),3.84(s,3H),3.77–3.69(m,1H),3.56(s,3H),2.44(s,2H),2.13–1.96(m,1H),1.91–1.75(m,1H).13C NMR(150MHz,DMSO)174.40,173.76,173.46,171.98,168.77,157.46,155.11,144.66,136.23,132.59,131.00,130.40,124.49,123.66,121.94,119.78,119.10,118.40,52.85,52.56,49.26,39.22,32.21,32.12,29.40,28.25,23.46,21.44,19.50,13.81,12.97.HRMS(ESI):m/z(M+H+)calcd for C25H24O6N3,462.1660,found:462.1658.
Example 74
Synthesis of methyl (R) -methyl 3- (8- (3, 5-bis (trifluoromethyl) benzamido) -7-methoxy-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (74)
Figure GDA0001986101440001111
Prepared according to the procedure for the synthesis of compound 71 in example 71, using 65-c and p-trifluoromethylbenzoyl chloride, to provide compound 74.
1H NMR(300MHz,DMSO)10.34(s,1H),10.29(s,1H),8.55(s,2H),8.49(d,J=5.4,1H),8.37(s,1H),7.62(s,1H),7.35(s,1H),3.87(s,3H),3.72(d,J=7.2,1H),3.56(s,3H),2.44–2.40(m,2H),2.10–1.98(m,1H),1.92–1.74(m,1H).HRMS(ESI):m/z(M+H+)calcd forC23H20O6N3F6,548.1251,found:548.1252.
Example 75
Synthesis of methyl (R) -methyl 3- (8- ((2-ethylbutyl) amino) -2, 5-dioxo-7-phenoxy-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (75)
Figure GDA0001986101440001112
Prepared according to the synthesis of compound 72 described in example 72, using 49-c and 2eq aldehyde components.
1H NMR(300MHz,DMSO)10.21(s,1H),8.07(d,J=4.9,1H),7.40(t,J=8.0,2H),7.17–7.11(m,1H),7.03–7.00(m,3H),6.35(s,1H),5.96(t,J=5.7,1H),3.71(s,1H),3.59(s,3H),3.01(t,J=6.2,2H),2.49–2.42(m,2H),7.17–7.11(m,1H),2.11–2.04(m,1H),1.65–1.57(m,1H),1.35–1.26(m,3.3,4H),0.89–0.83(m,6H).HRMS(ESI):m/z(M+H+)calcdfor C25H32O5N3,454.2336,found:454.2331.
Example 76
Synthesis of methyl (R) -methyl 3- (8- (3- (4-fluorophenyl) ureido) -2, 5-dioxo-7-phenoxy-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (76)
Figure GDA0001986101440001121
Weighing about 31mg (1eq) of intermediate 49-C into a 25m L round-bottom flask, adding 5m L THF for dissolving, adding 1.2eq p-fluorophenyl isocyanate in turn under the condition of ice-water bath while stirring, heating to room temperature, monitoring by L C-MS, and finishing the reaction after about 24 hours.
Evaporating the reaction solution to dryness under reduced pressure, adding appropriate amount of distilled water, extracting with DCM for 3 times, and collecting organic layer to obtain crude product of target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-sand 300-mesh silica gel, and the mobile phase is DCM and MeOH which are 100:4, so as to obtain the pure product of the target compound.
1H NMR(300MHz,DMSO)10.38(s,1H),9.47(s,1H),8.78(s,1H),8.34(s,1H),8.17(s,1H),7.46(d,J=6.8,4H),7.27–7.04(m,6H),3.78–3.64(m,1H),3.56(s,3H),2.46–2.37(m,2H),2.11–1.95(m,1H),1.88–1.75(m,1H).HRMS(ESI):m/z(M+H+)calcd for269H24O6N4F,507.1674,found:507.1678.
Example 77
Synthesis of methyl (R) -methyl 3- (8- (cyclopropylcarboxamido) -7- (3, 4-dimethylphenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (77)
Figure GDA0001986101440001131
Step (a): synthesis of 5- (3, 4-Dimethylphenylmethoxy) -2, 4-dinitrobenzoic acid (77-a)
Figure GDA0001986101440001132
2.46g of 5-chloro-2, 4-dinitrobenzoic acid and 1.34g (1.1eq) of 3, 4-dimethylphenol are weighed into a 100ml round-bottomed flask, about 30ml of distilled water is added, and ultrasonic wave is used for assisting dissolution. While stirring at room temperature, 1.85g (2.2eq) NaHCO was slowly added3Continuously stirring at room temperature until no bubbles are generated, heating to 110 ℃ at 100 ℃, monitoring the reaction by L C-MS, after about 1h, finishing the reaction, cooling the reaction solution to room temperature, adding dilute hydrochloric acid to adjust the pH value to 1-2, so that a large amount of yellow solid is separated out, performing vacuum filtration to obtain a target substance, and directly performing vacuum filtration without purificationThen the next reaction is carried out.
Step (b): synthesis of 5- (3, 4-dimethyl) phenoxy-2, 4-dinitrobenzoic acid D glutamic acid dimethyl ester amide (77-b)
Figure GDA0001986101440001133
Weighing 332mg of 77-a, putting the weighed 77-a into a 100m L round-bottom flask, sequentially adding 30m L DCM and 3-5 drops of DMF, carrying out ultrasonic dissolution assistance, stirring at room temperature, slowly adding 0.25m L (3eq) oxalyl chloride, heating to reflux when no more gas is generated, reacting for about 1-2 hours, cooling the reaction liquid to room temperature, evaporating the solvent under reduced pressure, adding 30m L DCM, adding 0.42m L (3eq) Et3N and 0.32g (1.5eq) of D-dimethyl glutamate under the condition of ice water bath, gradually heating to room temperature, monitoring the reaction by L C-MS, and finishing the reaction after about 1-3 hours.
Adding appropriate amount of distilled water, extracting for 3 times, and collecting organic layer to obtain crude product of target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-300 meshes of silica gel, and the mobile phase is DCM and MeOH which is 100:1, so as to obtain the pure product of the target compound.
Step (c): 4-amino-5- (3, 4-dimethyl) benzyloxy-1, 4-benzodiazepine-2-5-dione (77-c)
Figure GDA0001986101440001141
Weighing 489mg of 77-b in a 100m L round-bottom flask, sequentially adding 10m L THF and 10m L EtOH, performing ultrasonic assisted dissolution, stirring at room temperature, slowly adding 400mg Pd/C and 800mg ammonium formate, heating to 40 ℃, reacting for about 3-5 hours, cooling the reaction solution to room temperature, performing reduced pressure filtration, collecting filtrate, evaporating the solvent to dryness under reduced pressure, directly taking 1mol of the solvent to the 50m L round-bottom flask without purification, dissolving 15m L glacial acetic acid, reacting at 90 ℃, monitoring by L C-MS, reacting for about 2 hours, and evaporating the solvent to dryness under reduced pressure to obtain a crude target compound.
Separating and purifying by silica gel column chromatography, wherein the stationary phase is 200-300 mesh silica gel, and the mobile phase is DCM and MeOH which is 100:4, so as to obtain the pure product of the target compound
1H NMR(400MHz,DMSO)10.15(s,1H),8.04(d,J=5.1,1H),7.12(d,J=8.2,1H),6.94(s,1H),6.81(d,J=2.1,1H),6.70(dd,J=8.2,2.3,1H),6.40(s,1H),5.72(s,2H),3.69–3.64(m,1H),3.57(s,3H),2.44–2.39(m,2H),2.19(d,J=5.2,6H),2.04–1.97(m,1H),1.84–1.74(m,1H).
Step (d): synthesis of target Compound 77
Weighing about 30mg (1eq) of intermediate 77-c into a 25m L round-bottom flask, adding 5m L THF for dissolving, and adding 1.2eq of cyclopropyl formyl chloride and 21 mu L (1.5eq) Et sequentially under the condition of ice-water bath with stirring3And N, heating to room temperature, monitoring by L C-MS, and finishing the reaction after about 2 hours.
Adding an appropriate amount of methanol to quench the reaction, evaporating the reaction solution to dryness under reduced pressure, adding an appropriate amount of distilled water, extracting with DCM for 3 times, and collecting an organic layer to obtain a crude product of the target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-sand 300-mesh silica gel, and the mobile phase is DCM and MeOH which are 100:4, so as to obtain the pure product of the target compound.
1H NMR(300MHz,DMSO)10.31(s,1H),9.97(s,1H),8.39(d,J=5.4,1H),8.03(s,1H),7.21(d,J=8.2,1H),7.00(s,1H),6.94(d,J=2.3,1H),6.85(dd,J=8.0,2.4,1H),3.70(dd,J=12.3,6.8,1H),3.57(s,3H),2.09–1.94(m,1H),0.81(d,J=7.4,4H).
HRMS(ESI):m/z(M+H+)calcd for C25H28O6N3,466.1973,found:466.1973.
Example 78
Synthesis of methyl (R) -methyl 3- (8- (2- (phenoxy) acetamido) -7- (3, 4-dimethylphenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (78)
Figure GDA0001986101440001151
Prepared according to the method for the synthesis of compound 1 in example 1, using 77-c and 5-a, compound 78 is obtained.1H NMR(300MHz,DMSO)10.42(s,1H),9.39(s,1H),8.43(d,J=5.4,1H),8.17(s,1H),7.35–7.25(m,5H),7.19(d,J=8.0,1H),7.07(s,1H),6.94–6.87(m,1H),6.81(dd,J=8.9,3.1,1H),4.59(s,2H),4.20(s,2H),3.75–3.68(m,1H),3.56(s,3H),2.41(d,J=12.2,2H),2.22(s,6H),2.07–1.99(m,1H),1.87–1.74(m,1H).
HRMS(ESI):m/z(M+H+)calcd for C30H32O7N3,546.2235,found:546.2239.
Example 79
Synthesis of (R) -3- (8- (cyclopropylcarboxamido) -7- (3, 4-dimethylphenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionic acid (79)
Figure GDA0001986101440001152
Weighing about 47mg of compound 77 into a 25m L round-bottomed flask, adding 5m L THF for dissolving, adding 2.5eq L iOH in 5m L aqueous solution under stirring at room temperature, continuing the reaction at room temperature, and monitoring by L C-MS, wherein the reaction is finished after about 15 mm.
Adding a proper amount of dilute hydrochloric acid to quench the reaction, evaporating THF to dryness under reduced pressure, adding a proper amount of distilled water, and performing ultrasonic treatment to obtain solid powder which is separated out, thereby obtaining a crude product of the target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-mesh silica gel with 300 meshes, and the mobile phase is DCM, MeOH and HOAc which are 100:5:2, so as to obtain the pure product of the target compound.
1H NMR(400MHz,DMSO)12.085(s,1H),10.28(s,1H),9.96(s,1H),8.43(d,J=4.8,1H),8.02(s,1H),7.20(d,J=8.2,1H),6.99(s,1H),6.93(d,J=2.3,1H),6.84(dd,J=8.2,2.5,1H),3.69–3.64(m,1H),2.30(t,J=7.4,2H),2.23(d,J=3.4,6H),2.18(dd,J=12.4,6.7,1H),1.99–1.94(m,1H),1.79–1.74(m,1H),0.82–0.79(m,4H).13C NMR(150MHz,dmso)174.03,172.63,170.97,166.88,153.53,144.09,138.25,132.65,132.31,131.95,130.72,121.12,120.89,117.08,117.04,113.96,30.09,23.31,19.48,18.66,14.17,7.86,7.77.
HRMS(ESI):m/z(M+H+)calcd for C24H26O6N3,452.1816,found:452.1814.
Example 80
Synthesis of methyl (R) -methyl 3- (8- (3-cyanobenzoylamino) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (80)
Figure GDA0001986101440001161
Prepared according to the procedure for the synthesis of compound 71 in example 71, using 1-d and p-cyanobenzoyl chloride, compound 80 is obtained.1H NMR(300MHz,DMSO)10.46(s,1H),10.21(s,1H),8.50(s,1H),8.20–8.03(m,3H),7.76–7.69(m,2H),7.26–7.14(m,2H),6.54–6.50(m,1H),6.46–6.40(m,1H),6.29–6.25(m,1H),3.78(s,1H),2.87(s,6H),2.15–1.95(m,1H),1.95–1.70(m,1H).13C NMR(150MHz,DMSO)173.38,171.55,167.34,164.53,157.37,152.42,146.27,135.74,135.62,133.07,132.62,132.51,131.92,130.54,130.28,123.93,119.45,118.66,117.66,111.94,108.60,106.40,103.38,51.82,51.39,40.41,30.15,23.64.
HRMS(ESI):m/z(M+H+)calcd for C29H28O6N5,542.2034,found:542.2033.
Example 81
Synthesis of methyl (R) -methyl 3- (7- (3- (dimethylamino) phenoxy) -8- (2- (dodecylamino) acetamido) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (81)
Figure GDA0001986101440001162
Prepared according to the procedure for the synthesis of compound 71 in example 71, using 1-d and 2- (dodecylamino) acetyl chloride, compound 81 is obtained.1H NMR(300MHz,DMSO)10.46(s,1H),8.46(d,J=5.0,1H),8.28(s,1H),7.24(t,J=8.2,1H),7.17(s,1H),6.60(dd,J=8.5,2.0,1H),6.47(s,1H),6.30(d,J=7.9,1H),3.76(dd,J=12.4,6.6,1H),3.61(s,3H),2.95(s,6H),2.49–2.44(m,4H),2.15–2.01(m,1H),1.96–1.74(m,1H),1.37–1.18 1.28(m,24H),0.90(t,J=6.3,3H).
HRMS(ESI):m/z(M+H+)calcd for C35H52O6N5,638.3912,found:638.3908.
Example 82
Synthesis of methyl 3- ((3R) -8- ((norbornen-2-ylmethyl) amino) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (82)
Figure GDA0001986101440001171
Compound 82 was prepared according to the synthesis of compound 72 in example 72, using 1-d and 2eq aldehyde components.1H NMR(300MHz,DMSO)10.09(s,1H),8.04(s,1H),7.17(t,J=8.1,1H),6.99(d,J=1.9,1H),6.55(d,J=7.7,1H),6.47(s,1H),6.35–6.32(m,1H),6.26(d,J=7.7,1H),6.18(s,1H),6.00(s,1H),3.67(s,1H),3.56(s,3H),2.90(s,7H),2.78(s,3H),2.43(s,3H),2.08–2.00(m,1H),1.87–1.74(m,2H),1.38–1.28(m,1H),1.21(d,J=8.8,2H),0.54(d,J=12.8,1H).
HRMS(ESI):m/z(M+H+)calcd for C29H35O5N4,519.2602,found:519.2603.
Example 83
Synthesis of (R) -3- (8- (2- (phenoxy) acetamido) -7- (3- (dimethylamino) phenoxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanamide (83)
Figure GDA0001986101440001172
Weighing about 50mg of compound 5 into a 25m L round-bottomed flask, adding 5m L THF for dissolving, adding 2.5eq L iOH in 5m L aqueous solution under stirring at room temperature, continuing the reaction at room temperature, and after about 15 mm, finishing the reaction by L C-MS monitoring.
Adding a proper amount of dilute hydrochloric acid to quench the reaction, evaporating THF to dryness under reduced pressure, adding a proper amount of distilled water, and performing ultrasonic treatment to obtain solid powder which is separated out, thereby obtaining a hydrolysate of the THF.
Dissolving the mixture in 10m L THF, adding 1.5eqHOSu and 1.5eqDIC while stirring at room temperature, reacting at room temperature overnight, detecting the next day, and converting the substrate into an active ester intermediate, adding 3 eqammonia water to the reaction solution while stirring at room temperature, monitoring by L C-MS, and finishing the reaction after about 30 min.
Adding a large amount of methanol to quench the reaction, evaporating the reaction solution to dryness under reduced pressure, adding a proper amount of distilled water, extracting with DCM for 3 times, and collecting an organic layer to obtain a crude product of the target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-sand 300-mesh silica gel, and the mobile phase is DCM and MeOH which are 100:5, so as to obtain the pure product of the target compound.
1H NMR(400MHz,DMSO)10.40(s,1H),9.38(s,1H),8.43(d,J=5.5,1H),8.17(s,1H),7.34–7.20(m,7H),7.13(s,1H),6.73(s,1H),6.60–6.57(m,1H),6.45(t,J=2.3,1H),6.31–6.29(m,1H),4.60(s,2H),4.20(d,J=2.8,2H),3.65(s,1H),2.89(s,6H),2.17(d,J=6.8,2H),2.00–1.95(m,1H),1.81–1.75(m,1H).
HRMS(ESI):m/z(M+H+)calcd for C29H32O6N5,546.2347,found:546.2349.
Example 84
Synthesis of methyl (R) -methyl 3- (8-acetamido-7- (dibutylamino) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (84)
Figure GDA0001986101440001181
Step (a): synthesis of 5- (dibutylamino) -2, 4-dinitrobenzoic acid (84-a)
Figure GDA0001986101440001182
2.46g1-a was weighed into a 100m L round bottom flask, about 30m L THF was added and dissolved by sonication, 3.5m L (2.5eq) Et was added slowly in sequence with stirring at room temperature3N, 1.85m L (1.1eq) dibutylamin, continuing the reaction at room temperature, monitoring the reaction by L C-MS, after about 3H, adding dilute hydrochloric acid to adjust the pH value to acidity, decompressing and evaporating THF, adding a proper amount of DCM and H2Extracting with O for 3 times, collecting organic layer, and evaporating solvent under reduced pressureThe target product is obtained and directly subjected to the next reaction without purification.
1H NMR(300MHz,CDCl3)8.52(s,4H),7.19(s,1H),3.28(t,J=7.2,4H),1.64–1.52(m,4H),1.29(dd,J=14.8,7.3,4H),0.89(t,J=7.3,6H).
Step (b): synthesis of 5-dibutylamino-2, 4-dinitrobenzoic acid, D-glutamic acid dimethyl ester amide (84-b)
Figure GDA0001986101440001191
Weighing 297mg 84-a in a 100m L round-bottom flask, sequentially adding 30m L DCM and 3-5 drops of DMF, performing ultrasonic dissolution, stirring at room temperature, slowly adding 0.25m L (3eq) oxalyl chloride, heating to reflux when no more gas is generated, reacting for about 1-2 h, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, adding 30m L DCM, and adding 0.42m L (3eq) Et in an ice-water bath3And (3) gradually heating 0.32g (1.5eq) of D-dimethyl glutamate to room temperature, monitoring the reaction by L C-MS, and finishing the reaction after about 1-3 hours.
Adding appropriate amount of distilled water, extracting for 3 times, and collecting organic layer to obtain crude product of target compound.
Separating and purifying by silica gel column chromatography, wherein the stationary phase is 200-300 mesh silica gel, and the mobile phase is DCM and MeOH which is 100:2.5, so as to obtain the pure product of the target compound
1H NMR(400MHz,CDCl3)8.60(s,1H),7.03(s,1H),6.69(d,J=7.5,1H),4.86–4.81(m,1H),3.81(s,3H),3.68(s,3H),3.29–3.25(m,4H),2.60–2.51(m,2H),2.38–2.33(m,1H),2.20–2.13(m,1H),1.63–1.55(m,4H),1.35–1.28(m,4H),.
Step (c): synthesis of 4-amino-5-dibutylamino-1, 4-benzodiazepine-2-5-dione (84-c)
Figure GDA0001986101440001192
497mg of 84-b is weighed into a 100m L round-bottom flask, 10m L THF and 10m L EtOH are sequentially added into the flask, ultrasonic dissolution is carried out, stirring is carried out under the condition of room temperature, 400mg Pd/C and 800mg ammonium formate are slowly added into the flask while stirring is carried out, the temperature is increased to 40 ℃, reaction is carried out for about 3-5 hours, the reaction liquid is cooled to room temperature, reduced pressure filtration is carried out, filtrate is collected, the solvent is evaporated to dryness under reduced pressure, about 1mol of the solvent is directly taken into the 50m L round-bottom flask without purification, 15m L glacial acetic acid is dissolved, 0.15m L distilled water is added into a reaction container, the reaction is carried out at 90 ℃, L C-MS monitoring is carried out, after about 2 hours, the reaction is finished.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-sand 300-mesh silica gel, and the mobile phase is DCM and MeOH which are 100:4, so as to obtain the pure product of the target compound.
Step (d): synthesis of target Compound 84
Prepared according to the procedure for the synthesis of compound 71 in example 71, using 84-c and acetyl chloride, compound 81 is obtained.1HNMR(400MHz,DMSO)10.34(s,1H),9.05(s,1H),8.38(d,J=5.3,1H),8.00(s,1H),7.55(s,1H),3.73–3.68(m,1H),3.56(s,3H),2.86–2.82(m,4H),2.47–2.39(m,2H),2.15(s,3H),2.09–1.96(m,1H),1.88–1.76(m,1H),1.35–1.23(m,8H),0.84(t,J=6.9,6H).13C NMR(150MHz,dmso)172.87,170.95,168.40,167.36,138.72,136.17,133.86,124.46,120.89,110.91,54.39,51.26,50.92,29.68,28.87,24.35,23.18,19.92,13.81.
HRMS(ESI):m/z(M+H+)calcd for C23H35O5N4,447.2602,found:447.2599.
Example 85
Synthesis of methyl (R) -methyl 3- (8- (cyclopropylcarboxamido) -7-morpholinyl-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (85)
Figure GDA0001986101440001201
Step (a): synthesis of 5-morpholine-2, 4-dinitrobenzoic acid (85-a)
Figure GDA0001986101440001202
2.46g of 5-chloro-2, 4-dinitrobenzoic acid are weighed into a 100m L round-bottomed flask, about 30m L THF are added, ultrasound assistedDissolve, add slowly 3.5m L (2.5eq) Et in sequence with stirring at room temperature3N, 0.96Ml (1.1eq) morpholine, continuing the reaction at room temperature, monitoring the reaction by L C-MS, after about 3H, adding dilute hydrochloric acid to adjust the pH value to acidity, evaporating THF under reduced pressure, adding a proper amount of DCM and H2And (3) extracting for 3 times, collecting an organic layer, and evaporating the solvent to dryness under reduced pressure to obtain a target substance, wherein the target substance is directly subjected to the next reaction without purification.
Step (b): synthesis of 5-morpholinyl-2, 4-dinitrobenzoic acid D glutamic acid dimethyl ester amide (85-b)
Figure GDA0001986101440001203
Weighing 297mg of 85-a in a 100 round-bottom flask, sequentially adding 30m of L DCM and 3-5 drops of DMF (dimethyl formamide), performing ultrasonic dissolution assistance, stirring at room temperature, slowly adding 0.25m of L (3eq) of oxalyl chloride, heating to reflux when no more gas is generated, reacting for about 1-2 hours, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, adding 30m of L DCM, and adding 0.42m of L (3eq) of Et under the condition of ice-water bath3And (3) gradually heating 0.32g (1.5eq) of D-dimethyl glutamate to room temperature, monitoring the reaction by L C-MS, and finishing the reaction after about 1-3 hours.
Adding appropriate amount of distilled water, extracting for 3 times, and collecting organic layer to obtain crude product of target compound.
Separating and purifying by silica gel column chromatography, wherein the stationary phase is 200-300 mesh silica gel, and the mobile phase is DCM and MeOH which is 100:2.5, so as to obtain the pure product of the target compound
1H NMR(300MHz,CDCl3)8.65(s,1H),7.08(s,1H),6.86(d,J=7.5,1H),4.84–4.77(m,7.9,1H),3.88–3.84(m,4H),3.81(s,3H),3.67(s,3H),3.32–3.28(m,4H),2.61–2.50(m,2H),2.39–2.29(m,1H),2.21–2.11(m,1H).
Step (c): synthesis of 4-amino-5-morpholinyl-1, 4-benzodiazepine-2-5-dione (85-c)
Figure GDA0001986101440001211
Weighing 454mg 85-b into a 100m L round-bottom flask, sequentially adding 10m L THF and 10m L EtOH, performing ultrasonic assisted dissolution, stirring at room temperature, slowly adding 400mg Pd/C and 800mg ammonium formate, heating to 40 ℃, reacting for about 3-5 hours, cooling the reaction solution to room temperature, performing reduced pressure filtration, collecting filtrate, evaporating the solvent under reduced pressure, directly dissolving 15m L glacial acetic acid in about 1mol in a 50m L round-bottom flask without purification, adding 0.15m L distilled water into a reaction container, reacting at 90 ℃, monitoring by L C-MS, reacting for about 2 hours, finishing the reaction, and evaporating the solvent under reduced pressure to obtain a crude target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-300 meshes of silica gel, and the mobile phase is DCM and MeOH which is 100:6, so as to obtain the pure product of the target compound.
1H NMR(300MHz,DMSO)10.09(s,1H),8.01(d,J=5.0,1H),7.22(s,1H),6.31(s,1H),5.57(s,2H),3.75(s,4H),3.57(s,3H),2.83–2.69(m,4H),2.42(s,2H),2.05–1.97(m,1H),1.86–1.75(m,1H).
Step (d): synthesis of target Compound 85
Prepared according to the method for the synthesis of compound 71 in example 71, using 85-c and cyclopropylcarbonyl chloride, compound 85 is obtained.1H NMR(400MHz,DMSO)10.29(s,1H),9.32(s,1H),8.38(d,J=5.3,1H),7.89(s,1H),7.46(s,1H),3.87–3.79(m,4H),3.72–3.67(m,1H),3.56(s,3H),2.89–2.84(m,2H),2.78–2.72(m,2H),2.45–2.39(m,2H),2.14–2.07(m,1H),2.05–1.98(t,J=14.8,1H),1.86–1.77(m,1H),0.83(d,J=6.0,4H).
HRMS(ESI):m/z(M+H+)calcd for C21H27O6N4,431.1925,found:431.1933.
Example 86
Synthesis of methyl (R) -methyl 3- (7-morpholinyl-2, 5-dioxo-8- (2-phenylacetamido) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (86)
Figure GDA0001986101440001221
Prepared according to the procedure for the synthesis of compound 85 in example 85, using 85-c and phenylacetyl chloride, to provide compound 86.1HNMR(400MHz,DMSO)10.34(s,1H),8.94(s,1H),8.39(d,J=5.4,1H),7.98(s,1H),7.44(s,1H),7.40(d,J=4.3,4H),7.36–7.31(m,1H),3.83(q,J=15.1,2H),3.71–3.66(m,1H),3.56(s,3H),3.55–3.46(m,4H),2.68–2.62(m,2H),2.61–2.55(m,2H),2.44–2.36(m,2H),2.05–1.99(m,1H),1.85–1.78(m,1H).13C NMR(150MHz,dmso)172.87,170.97,169.35,167.28,138.00,136.06,135.13,133.78,129.47,128.83,127.11,121.80,121.31,111.97,66.02,51.84,51.30,50.88,43.83,29.64,23.17.
HRMS(ESI):m/z(M+H+)calcd for C25H29O6N3,481.2082,found:481.2081.
Example 87
Synthesis of (R) -N- (3- (3- (dodecylamino) -3-propionyl) -7-morpholinyl-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) cyclopropylcarboxamide (87)
Figure GDA0001986101440001222
Prepared according to the method for the synthesis of compound 6 in example 6, using 85 and dodecylamine, compound 87 is obtained.1HNMR(400MHz,DMSO)10.25(s,1H),9.29(s,1H),8.35(d,J=5.3,1H),7.89(s,1H),7.72(t,J=5.6,1H),7.44(s,1H),3.83–3.79(m,4H),3.63–3.58(m,1H),2.96–2.91(m,2H),2.86–2.83(m,2H),2.76–2.71(m,2H),2.16(t,J=7.1,2H),2.12–2.05(m,1H),2.01–1.91(m,1H),1.80–1.73(m,1H),1.31–1.16(m,20H),0.85–0.81(m,8H).13C NMR(150MHz,dmso)174.81,174.05,173.82,170.13,141.02,139.04,136.36,124.21,124.02,115.49,68.81,54.74,53.97,41.08,34.07,33.99,31.81,31.79,31.78,31.73,31.48,31.42,29.06,26.57,24.87,17.51,16.73,10.63,10.50.
HRMS(ESI):m/z(M+H+)calcd for C32H50O5N5,584.3806,found:584.3806.
Example 88
Synthesis of N- ((3R) -7-morpholinyl-2, 5-dioxo-3- (3-oxo-3- ((1,2,3, 4-tetrahydronaphthaleneacetamido-1-yl) amino) propyl) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) cyclopropylcarboxamide (88)
Figure GDA0001986101440001231
Prepared according to the method for the synthesis of compound 6 in example 6, using 85 and 1,2,3, 4-tetrahydro-1-naphthylamine, compound 88 is obtained.1H NMR(400MHz,DMSO)10.29(s,1H),9.33(d,J=2.3Hz,1H),8.42–8.33(m,1H),8.20(d,J=8.6Hz,1H),7.92(d,J=4.2Hz,1H),7.47(d,J=7.4Hz,1H),7.23–6.84(m,4H),4.90(s,1H),3.93–3.73(m,4H),3.75–3.56(m,1H),2.96–2.82(m,2H),2.80–2.72(m,2H),2.26(t,J=7.1Hz,2H),2.22–2.01(m,3H),1.99–1.72(m,4H),1.73–1.49(m,2H),0.85(d,J=5.7Hz,4H).
HRMS(ESI):m/z(M+H+)calcd for C30H36O5N5,546.2711,found:546.2708.
Example 89
Synthesis of (R) -N- (3- (3- ((2- (cyclohexyl-1-en-1-yl) ethyl) amino) -3-propionyl) -7-morpholinyl-2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) cyclopropylcarboxamide (89)
Figure GDA0001986101440001232
Prepared according to the method for the synthesis of compound 6 in example 6, using 85 and 2- (1-cyclohexenyl) ethylamine to provide compound 89.1H NMR(400MHz,DMSO)10.26(s,1H),9.30(s,1H),8.37(d,J=5.3,1H),7.90(s,1H),7.70(s,1H),7.45(s,1H),5.28(s,1H),3.87–3.78(m,4H),3.65–3.60(m,1H),3.06–3.01(dd,m,2H),2.89–2.82(m,2H),2.79–2.71(m,2H),2.19–2.15(m,2H),2.13–2.07(m,1H),2.02–1.97(m,1H),1.92–1.87(m,4H),1.85–1.81(m,2H),1.76–1.74(m,1H),1.52–1.42(m,4H),0.83(d,J=6.1,4H).13C NMR(150MHz,dmso)173.37,172.55,172.37,168.68,139.57,137.57,136.19,134.89,123.11,122.73,122.49,114.04,67.48,53.27,38.91,38.28,32.56,28.90,25.96,25.15,23.71,23.25,16.05,9.17,9.05.
HRMS(ESI):m/z(M+H+)calcd for C28H38O5N5,524.2867,found:524.2869.
Example 90
Synthesis of methyl (R, Z) -methyl 3- (8- (3- (2-chloro-4-fluorophenyl) acrylamido) -7- (naphthylacetyl-1-yloxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionate (90)
Figure GDA0001986101440001241
Step (a): synthesis of 5- (1) -Naphthalenemethoxy-2, 4-dinitrobenzoic acid (90-a)
Figure GDA0001986101440001242
2.46g of 1-a, 1.58g (1.1eq) of 1-naphthol were weighed into a 100m L round bottom flask, about 30m L of distilled water was added, ultrasonic solubilization was performed, and 1.85g (2.2eq) of NaHCO was slowly added with stirring at room temperature3And continuously stirring at room temperature until no bubbles are generated, heating to 100-110 ℃, monitoring the reaction by L C-MS, after about 1h, finishing the reaction, cooling the reaction solution to room temperature, adding dilute hydrochloric acid to adjust the pH value to 1-2, separating out yellow solid, performing reduced pressure suction filtration to obtain a target substance, and directly performing the next reaction without purification.
1H NMR(300MHz,DMSO)8.85(s,1H),8.09(d,J=7.7,1H),7.97(d,J=8.2,2H),7.70–7.55(m,3H),7.45(d,J=7.4,1H),7.04(s,1H).
Step (b): synthesis of dimethyl 5-naphthyloxy-2, 4-dinitrobenzoic acid D-glutamic acid amide (90-b)
Figure GDA0001986101440001243
Weighing 354mg of 90-a into a 100m L round-bottom flask, sequentially adding 30m L DCM and 3-5 drops of DMF, carrying out ultrasonic dissolution assistance, stirring at room temperature, slowly adding 0.25m L (3eq) oxalyl chloride, heating to reflux when no more gas is generated, reacting for about 1-2 h, cooling the reaction solution to room temperature, evaporating the solvent under reduced pressure, adding 30m L DCM, and adding 0.42m L (3eq) Et in an ice water bath3N, 0.32g (1.5eq) of D-glutamic acid dimethyl esterAnd (3) gradually heating the ester to room temperature, monitoring the reaction by L C-MS, and finishing the reaction after about 1-3 h.
Adding appropriate amount of distilled water, extracting for 3 times, and collecting organic layer to obtain crude product of target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-300 meshes of silica gel, and the mobile phase is DCM and MeOH which is 100:1, so as to obtain the pure product of the target compound.
1H NMR(300MHz,CDCl3)8.79(s,1H),7.94(d,J=7.1,2H),7.84(d,J=8.3,1H),7.62–7.47(m,3H),7.29–7.20(m,1H),6.92(s,1H),6.81(d,J=7.4,1H),4.66–4.59(m,1H),3.65(s,3H),3.54(s,3H),2.43–2.36(m,2H),2.24–2.14(m,1H),2.06–1.99(m,1H).
Step (c), synthesizing 4-amino-5- α naphthylmethoxy-1, 4-benzodiazepine-2-5-dione (90-c)
Figure GDA0001986101440001251
Weighing 511mg 90-b, putting the weighed 511mg 90-b into a 100m L round-bottom flask, sequentially adding 10m L THF and 10m L EtOH, performing ultrasonic assisted dissolution, stirring at room temperature, slowly adding 400mg Pd/C and 800mg ammonium formate, heating to 40 ℃, reacting for about 3-5 hours, cooling the reaction solution to room temperature, performing reduced pressure filtration, collecting filtrate, and performing reduced pressure evaporation on the solvent to obtain the target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-300 meshes of silica gel, and the mobile phase is DCM and MeOH which is 100:4, so as to obtain the pure product of the target compound.
1H NMR(300MHz,DMSO)10.20(s,1H),8.13(d,J=9.3,1H),8.04(d,J=5.2,1H),7.99(d,J=9.1,1H),7.71(d,J=8.2,1H),7.62–7.53(m,2H),7.47(t,J=7.9,1H),6.93(d,J=6.3,2H),6.48(s,1H),5.91(s,2H),3.67(d,J=5.7,1H),3.56(s,3H),2.40(d,J=7.1,2H),2.00(s,1H),1.80(d,J=7.5,1H).
Step (d): synthesis of target Compound 90
Weighing intermediate 90-c 1eq to 25m L round-bottomed flask, 5m L THF was added to dissolve the mixture, and 1.2eq of (Z) -3- (2-chloro-4-fluorophenyl) acryloyl chloride and 1.5eq of Et were added to the flask under stirring in an ice-water bath3And N, heating to room temperature, monitoring by L C-MS, and finishing the reaction after about 2 hours.
Adding an appropriate amount of methanol to quench the reaction, evaporating the reaction solution to dryness under reduced pressure, adding an appropriate amount of distilled water, extracting with DCM for 3 times, and collecting an organic layer to obtain a crude product of the target compound.
And (4) performing silica gel column chromatography separation and purification, wherein the stationary phase is 200-sand 300-mesh silica gel, and the mobile phase is DCM and MeOH which are 100:4, so as to obtain the pure product of the target compound.
1H NMR(400MHz,DMSO)10.45(s,1H),10.20(s,1H),8.38(d,J=5.3,1H),8.32(s,1H),8.05(t,J=6.9,2H),7.89–7.80(m,3H),7.65–7.55(m,4H),7.37–7.31(m,2H),7.21(d,J=7.5,1H),6.91(s,1H),3.74–3.69(m,1H),3.55(s,3H),2.44–2.37(m,2H),2.07–1.98(m,1H),1.84–1.74(m,1H).13C NMR(150MHz,dmso)175.97,174.07,169.83,166.98,166.40,164.73,154.19,147.62,138.16,137.75,137.6(d,J=5.4),135.49,135.40,132.52(d,J=9.3),132.38(d,J=3.5),131.13,130.07,129.69,129.40,129.33,128.25,127.82,124.73(d,J=9.75),122.65,120.41(d,J=24.9),119.65,118.83,118.53(d,J=21.6),117.01,54.41,53.99,32.76,26.26.
HRMS(ESI):m/z(M+H+)calcd for C32H26O6N3ClF,602.1489,found:602.1494.
Example 91
Synthesis of methyl (R) -methyl 3- (8- (cyclopropylcarboxamido) -7- (naphthylacetyl-1-yloxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanoate (91)
Figure GDA0001986101440001261
Prepared according to the procedure for the synthesis of compound 90 of example 90, using 90-c and cyclopropylcarbonyl chloride, give compound 91.1H NMR(400MHz,DMSO)10.35(s,1H),10.16(s,1H),8.36(d,J=5.3,1H),8.08(d,J=5.2,2H),8.03(d,J=7.4,1H),7.83(d,J=8.2,1H),7.62–7.53(m,3H),7.14(d,J=7.5,1H),6.91(s,1H),3.71–3.66(m,1H),3.55(s,3H),2.41–2.37(m,4.6,2H),2.22–2.17(m,1H),2.05–1.96(m,1H),1.82–1.73(m,1H),0.84–0.80(m,4H).
HRMS(ESI):m/z(M+H+)calcd for C27H26O6N3,488.1816,found:488.1824.
Example 92
Synthesis of (R, Z) -3- (8- (3- (2-chloro-4-fluorophenyl) acrylamido) -7- (naphthylacetyl-1-yloxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propionic acid (92)
Figure GDA0001986101440001271
Compound 90(1mmol) was dissolved in 6m L THF and 3m L H2And adding L iOH (3mmol,3eq) into the O mixed solution, reacting at room temperature for 0.5h, completely reacting, evaporating the reaction solution to dryness, adding water for diluting, adjusting the pH value to acidity by using 1M hydrochloric acid, separating out a solid, filtering, and drying to obtain the target compound 92.1H NMR(400MHz,DMSO)12.09(s,1H),10.46(s,1H),10.21(s,1H),8.39(d,J=5.3,1H),8.33(s,1H),8.06(t,J=7.9,2H),7.91–7.80(m,3H),7.65–7.56(m,4H),7.38–7.32(m,2H),7.22(d,J=7.4,1H),6.92(s,1H),3.73-3.69(m,1H),2.32(t,J=7.8,2H),2.04–1.96(m,1H),1.82–1.74(m,1H).
HRMS(ESI):m/z(M+H+)calcd for C31H24O6N3ClF,588.1332,found:588.1341.
Example 93
Synthesis of (R) -N- (3- (3-amino-3-propionyl) -7- (naphthylacetyl-1-yloxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) cyclopropylcarboxamide (93)
Figure GDA0001986101440001272
Prepared according to the method for synthesizing compound 6 in example 6, using 91 with ammonia water, compound 93 is obtained.1H NMR(300MHz,DMSO)10.33(s,1H),10.16(s,1H),8.36(d,J=4.8,1H),8.13–7.98(m,3H),7.83(d,J=8.3,1H),7.67–7.49(m,3H),7.23(s,1H),7.15(d,J=7.2,1H),6.91(s,1H),6.72(s,1H),3.66–3.60(m,1H),2.20–2.13(m,3H),2.03–1.84(m,1H),1.79–1.69(m,1H),0.83(s,4H).
HRMS(ESI):m/z(M+H+)calcd for C26H25O5N4,473.1819,found:473.1823.
Example 94
Synthesis of (R, Z) -N- (3- (3-amino-3-propionyl) -7- (naphthylacetyl-1-yloxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) -3- (2-chloro-4-fluorophenyl) acrylamide (94)
Figure GDA0001986101440001281
Prepared according to the synthesis of compound 6 in example 6, using 90 with ammonia to afford compound 94.1H NMR(400MHz,DMSO)10.43(s,1H),10.19(s,1H),8.38(d,J=5.2,1H),8.31(s,1H),8.05(t,J=9.2,2H),7.89–7.80(m,3H),7.65–7.53(m,4H),7.37–7.30(m,2H),7.22(d,J=7.7,2H),6.91(s,1H),6.72(s,1H),3.68–3.63(m,1H),2.16(t,J=7.2,2H),1.99–1.94(m,1H),1.77–1.72(m,1H).13C NMR(150MHz,dmso)173.69,171.05,166.68,163.85,163.28,161.61,151.10,144.47,135.02,134.63,134.45(d,J=10.9),132.39,132.32,129.40(d,J=9.3),129.37,129.26(d,J=3.1),128.01,126.95,126.57,126.29,126.22,125.15,124.67,121.63(d,J=6.3),117.29(d,J=25),116.51,115.68,115.41(d,J=21.75),113.85,51.33,30.83,23.41.
HRMS(ESI):m/z(M+H+)calcd for C31H25O5N4ClF,587.1492,found:587.1495.
Example 95
Synthesis of (R, Z) -3- (2-chloro-4-fluorophenyl) -N- (3- (3- (diethylamino) -3-propionyl) -7- (naphthylacetyl-1-yloxy) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-8-yl) acrylamide (95)
Figure GDA0001986101440001282
Prepared according to the synthesis of compound 6 of example 6, using 90 and diethylamine, compound 95 was obtained.1H NMR(400MHz,DMSO)10.42(s,1H),10.20(s,1H),8.38(d,J=5.4,1H),8.31(s,1H),8.08–8.03(m,2H),7.89–7.70(m,3H),7.65–7.55(m,4H),7.36–7.32(m,2H),7.21(d,J=7.5,1H),6.90(s,1H),3.75–3.68(m,1H),3.24–3.18(m,4H),2.36(t,J=7.3,2H),2.03–1.95(m,1H),1.81–1.72(m,1H),1.03(t,J=7.1,3H),0.94(t,J=7.1,3H).13C NMR(150MHz,dmso)171.21,170.45,166.72,163.86,163.29,161.62,151.11,144.52,135.04,134.64,134.49,134.41,132.33,129.45,129.39,129.29,129.26,128.03,126.96,126.60,126.31,126.22,125.15,124.72,121.72,121.59,117.39,117.22,116.50,115.77,115.49,115.35,113.88,55.94,28.38,23.66,14.09,13.11.
HRMS(ESI):m/z(M+H+)calcd for C35H33O5N4ClF,643.2118,found:643.2116.
Example 96
Synthesis of methyl (96) 3- (8- (3- (2-chloro-4-fluorophenyl) acrylamido) -7- (di-n-butylamino) -2, 5-dioxo-2, 3,4, 5-tetrahydro-1H-benzo [ E ] [1,4] diazepin-3-yl) propanoate
Figure GDA0001986101440001291
Prepared according to the procedure for the synthesis of compound 84 in example 84 using 84-c and (Z) -3- (2-chloro-4-fluorophenyl) acryloyl chloride, to provide compound 96.1H NMR(400MHz,DMSO)10.42(s,1H),9.42(s,1H),8.43(d,J=5.1,1H),8.18(s,1H),8.05–8.00(m,6.5,1H),7.84(d,J=15.5,1H),7.64–7.56(m,2H),7.41–7.37(m,1H),7.28(d,J=15.5,1H),3.77–3.72(m,1H),3.57(d,J=5.9,3H),2.89(t,J=7.0,4H),2.44(t,J=9.2,2H),2.10–2.00(m,1H),1.88–1.79(m,1H),1.35–1.23(m,8H),0.82(t,J=7.1,6H).
HRMS(ESI):m/z(M+H+)calcd for C30H37O5N4ClF,587.2431,found:587.2426.
Example 97
Synthesis of (R) -N-heptyl-3- (7-morpholinyl-2, 5-dioxo-8- (2-phenylacetamido) -2,3,4, 5-tetrahydro-1H-benzo [ e ] [1,4] diazepin-3-yl) propanamide (97)
Figure GDA0001986101440001292
Prepared according to the method for the synthesis of compound 6 in example 6, using 86 and heptylamine, compound 96 is obtained.1H NMR(400MHz,DMSO)10.30(s,1H),8.91(s,1H),8.35(d,J=5.3,1H),7.97(s,1H),7.71(d,J=5.5,1H),7.45–7.35(m,5H),7.34–7.26(m,1H),3.81(q,J=15.2,2H),3.62–3.57(m,1H),3.52–3.46(m,4H),2.93(dd,J=12.8,6.6,2H),2.65–2.54(m,4H),2.15(t,J=7.1,2H),2.00–1.92(m,1H),1.79–1.71(m,1H),1.29–1.15(m,10H),0.82(t,J=7.0,3H).HRMS(ESI):m/z(M+H+)calcd for C31H42O5N5,564.3180,found:564.3182.
Example 98
Examination of the antagonistic action of WYZ035-31D (Compound 13 prepared in example 13) on NOD1 and NOD2
Materials and methods
(1) Cells and reagents
The test substance: WYZ035-31D was formulated as a 20mM DMSO stock solution and diluted with medium to the appropriate concentration just prior to use.
Cell: HEK-Blue hNOD1cell, HEK-Blue hNOD2cell, and 293-hNOD2, all from InvivoGen.
β -actin (Abcam, ab6276), I κ B α (CST, 9242S), JNK (CST, 9252S), p-JNK (CST, 9251S), p38(CST, 9212S), p-p38(CST, 4631S), ERK (Santa cruz, sc-94), p-ERK (Santa cruz, sc-7383), RIP2(CST, 4142), p-RIP2(CST, 4364), Goat polyclone semicolone semiconductive antibody to Rabbit IgG & L-HRP (Abcam, ab6721), rain polyclone semicolone semiconductive antibody to Mouse IgG-H & L-6728 (Abcam, 6728).
Primers and probes I L-6 (Hs00985639_ m1), TNF-alpha (Hs01113624_ g1) and GAPDH (Hs02758991_ g1) were purchased from Invitrogen.
Reagents hNOD1 agonist C12-ie-DAP (edition of a lauroyl group to the glutaminic acid derivative of ie-DAP), hNOD2 agonist Muramyl Dipeptide (MDP), hT L R4 agonist lipopolysaccharide (standard lipopolysaccharides, L PS-EK), HEK-Blue Detection, QUANTI-Blue, Normocin, Blastidin and Zeocin are all available from InvivoGen, DMEM medium, RPMI1640 medium, bovine serum, penilin-Streptomyces, PBS, EDTA, Trizol, High-capicin recovery transcription Kits,
Figure GDA0001986101440001301
gene Expression Master Mix was purchased from Invitrogen, Ficoll lymphocyte isolate was purchased from GE, Bovine Serum Albumin was purchased from MP, CD14MicroBeads and L S-MACSCcolumn were purchased from Miltenyl, RNeasy Mini Kit was purchased from QIAGEN, Phosphatase inhibitor Cocktail, protease inhibitor, Na3VO4, SRB, TCA, Tris-base and glacial acetic acid were purchased from Sigma, NaF and BSA were purchased from Amresco, Skim Milk was purchased from BD, Clarity Master Mix was purchased from Invitrogen, Ficoll lymphocyte isolate was purchased from GE, Bovine Serum was purchased from MP, CD14MicroBeads and L S-MACSCcolumn were purchased from Miltenyl, RNeasy Mini Kit was purchased from Sigma, NaF and BSA were purchased from Amresco, Skim MilkTMThe western EC L substrate, PVDF membrane and Precision plus protein Dual color standards were purchased from Bio-Rad, the 3MM filter paper from Millipore, the RIPA lysate, SDS-PAGE Gel Kit, L applying buffer, the transmembrane buffer, the electrophoresis buffer and the Western blot membrane regenerant were purchased from kang, century.
(2) Instrument and consumable
Microplate reader (PO L ARstar Omega, USA), horizontal centrifuge (Eppendorf), microscope (O L YMPUS), MACS Separator (Miltenyl), CO2Cell incubator (Thermo fisher), Nanodrop spectrophotometry (Thermo fisher), fluorescent quantitative PCR instrument (ABI), PCR instrument (Eppendorf), electrophoresis instrument (Bio-Rad), transfer tank (Bio-Rad), ChemiDocTMXRS+Imager (Bio-Rad), blood count plate (Shanghai Biochemical instruments Co., Ltd.), shaker, etc. 96-well plates (Costar, USA), 6-well plates (Costar, USA), 12-well plates (Costar, USA), centrifuge tubes (Corning), petri dishes (Corning), and the like.
(3) Detection of HEK-Blue hNOD1 antagonist (HEK-Blue Detection)
HEK-Blue hNOD1 cells cultured in a medium containing10% FBS, 50U/M L penicilin, 50. mu.g/M L streptomycin, 50. mu.g/M L Normocin, 2mM L-glutamine in DMEM medium (DMEM complete medium) cells were collected in the logarithmic growth phase, HEK-Blue Detection assay adjusted for cell concentration and seeded in 96well plates (50000 cells/well), test compound gradient working solutions (final concentrations 10. mu.M, 5. mu.M, 2.5. mu.M, 1.25. mu.M, 625nM, 312.5nM, 31.25nM, 3.125nM, 0.3125nM, respectively) and vehicle control were added, 37 ℃, 5% CO2After 3h incubation in the incubator, the positive stimulus C12-ie-DAP (final concentration 50ng/m L) was added at 37 ℃ with 5% CO2The incubation was continued in the incubator for 20h and the OD was measured at 655 nm.
The positive control group OD value was designated as C, and the test group OD value was designated as (T).
Antagonism Rate% (Inhibit Rate) [ (C-T)/C]× 100, computing IC50I.e., [ (C-T)/C]× 100 at 50;
after the detection is finished, an SRB method is adopted to examine the influence of the compound on the cell growth, 50 mu L/well is added with 80% TCA4 ℃ for fixing for 1h, deionized water is used for washing the plate for 5 times and is naturally dried, 0.4% SRB is added into 100 mu L/well for dyeing, the plate is placed at room temperature for 10min, 100 mu L/well 1% acetic acid is used for washing the plate for 5 times and is naturally dried, 150 mu L/well is added with Tris base and is oscillated for about 5min, the OD value is detected at 515nm of an enzyme labeling instrument, the OD value of a solvent control group is marked as C, the OD value of a tested group is marked as (T), and the growth rate% (Percentagegrowth) ([ T/C ] × 100.
(4) Detection of HEK-Blue hNOD2 antagonist (HEK-Blue Detection)
The method is the same as the detection of an HEK-Blue hNOD1 antagonist, the cell density is 25000 cells/well, the positive stimulus is MDP (50ng/m L), the incubation time is 18h, and the detection wavelength is 650 nm.
(5) Culture and treatment of human peripheral blood-derived macrophages (hPBMCs-derived macrophages)
Taking venous blood of healthy volunteers 200M L, separating peripheral blood mononuclear cells (hPBMCs) by Ficoll density gradient centrifugation, sorting CD14+ mononuclear cells by immunomagnetic bead method, inducing and differentiating M-CSF in vitro into macrophages, and taking 1 × 106Per well density was inoculated into 12 well plates and compounds WYZ035-31D (final concentrations 10. mu.M and 1. mu.M, respectively) or positive pairs were addedAfter incubation for 1h, C12-ie-DAP (final concentration 500ng/M L) or MDP (final concentration 5. mu.g/M L) was added in the control (1. mu.M), and after 90min the supernatant was aspirated and the cell sample was lysed with Trizol.
(6) Reverse transcription and fluorescent quantitative PCR
After lysing cell samples with Trizol, total RNA was extracted and purified using RNAeasy mini kit (Qianen) and RNA content was measured using a Nanodrop spectrophotometry (Thermo), the ratio of absorbance at wavelength 260 to absorbance at wavelength 280 (A260:280OD ratios) for each sample was greater than 1.8, 100ng of RNA was reverse transcribed into cDNA, 1. mu. L cDNA was added to the corresponding primers and probes to a final volume of 20. mu. L, and the samples were placed in a 96-well plate and subjected to a quantitative PCR reaction using a Fluorogenic PCR instrument, L-6, TNF-alpha and GAPDH primers and probes were purchased from Invitrogen (Cat. Hs. 00989 _ m1, Cat 01124 _ g1, Hs02758991_ g 1. the reaction system was 10. mu.7 Man Taq Master Mix (2x), Taq 1. mu. L, Taq 3520. mu.H 3526, As5631. mu.20. mu.H 2. mu.32 cDNA2O。
The reaction parameters are ① 50 ℃ and 2min
②95℃,10min
③95℃,15sec
④60℃,1min③to④40cycles
GAPDH was used as reference gene, 2-△ctThe relative expression amount is calculated, and the antagonism rate is calculated.
(7)Western Blotting
Collecting cells in logarithmic growth phase, inoculating into 6-well plate, adding test compound WYZ035-31D (final concentration of 10 μ M and 1 μ M), NOD1 or NOD2 positive antagonist (final concentration of 1 μ M), 37 deg.C, and 5% CO2Incubating in incubator for 1h, adding stimulus C12-ie-DAP (final concentration of 500ng/m L) or MDP (final concentration of 5 μ g/m L), incubating for 30min or 60min, lysing cell with RIPA lysate, extracting protein, performing SDS-PAGE electrophoresis with sample loading of 15 μ L/well, transferring protein to PVDF membrane, sealing with 5% skimmed milk powder at room temperature for 2h, adding diluted primary antibody at appropriate ratio, rinsing at 4 deg.C overnight at 1 × TBST for 3 times, adding diluted secondary antibody at appropriate ratio, incubating at room temperature for 1h, rinsing with 1 × TBST, and separatingThereafter, EC L luminescent substrate, ChemiDoc, was addedTMXRS+Exposure in the imager.
Test results
As shown in FIG. 1A, the compound WYZ035-31D has obvious antagonism to hNOD1/2 signal path, L ogIC50Respectively-5.627 and-5.379. No significant cytotoxicity was observed in WYZ035-31D under the above experimental concentration conditions, and the results are shown in FIG. 1
As can be seen from FIG. 2, WYZ035-31D can significantly antagonize the agonistic action of C12-ie-DAP on NOD1 signal pathway of human peripheral blood-derived macrophage, and decrease the transcription of pro-inflammatory cytokine I L-6 and TNF-alpha. Positive compound (positive control) is known NOD1 selective antagonist (structural formula I). As can be seen from FIG. 3, WYZ035-31D can significantly antagonize the agonistic action of MDP on NOD2 signal pathway of human peripheral blood-derived macrophage, and decrease the transcription of pro-inflammatory cytokine I L-6 and TNF-alpha. positive compound (positive control) is known NOD2 selective antagonist (structural formula ii).
Figure GDA0001986101440001331
As can be seen from FIG. 4(A), the expression of phosphorylated RIP2 was increased and the expression of IkB α was decreased after C12-ie-DAP-stimulated human peripheral blood-derived macrophages were activated, compared to the vehicle Control (Control) group, the expression of phosphorylated RIP2 was increased and the expression of IkB α was decreased, indicating that the NF-kB signaling pathway was activated, the expression of phosphorylated JNK, p38 and ERK was increased and indicating that the MAPK signaling pathway was activated, whereas the expression of phosphorylated JNK, p38 and ERK was decreased after WYZ 035-31D-treated cells compared to the C12-ie-DAP alone group, it was found that the expression of phosphorylated RIP2 was decreased and the expression of IkB α was increased and the expression of phosphorylated JNK was decreased, both JNK 38 and ERK were decreased, and WYZ035-31D blocked NOD 1-mediated activation of NF-kB and MAPK signaling pathway, it was found that FIG. 4(B) shows that 293-hNONkD 3 cells were increased after MDP-stimulated, and activated by MDP-19, and WYZ-7, whereas the phosphorylation of cells were decreased and the phosphorylation of the MDK-7-stimulated and the phosphorylation of the MDK-stimulated cells compared to the vehicle Control (WYZ-7, and the phosphorylation of the MDK-7, the phosphorylation pathways, and the phosphorylation of the MDK-7, the MDK-7.
WYZ035-31D can remarkably antagonize NF-kB and MAPK signal channel activation mediated by NOD1 and NOD2, and is NOD1/2 antagonist.
Example 99
Antagonism of NOD1 and NOD2 in vitro by the Compounds prepared in examples 1-54
Table 1 details the percent antagonism of the compounds of the examples to the NOD1/2 signaling pathway, where 10 μ M represents the percent antagonism to the NOD1/2 pathway at a test compound concentration of 10 μ M, and 5 μ M represents the percent antagonism to the NOD1/2 pathway at a test compound concentration of 5 μ M. L omiC50The value represents the log of the concentration of the test compound at a percent antagonism of 50%; "ND" means not detected.
TABLE 1 antagonism of NOD1 and NOD2 in vitro by the compounds of the examples
Figure GDA0001986101440001332
Figure GDA0001986101440001341
Figure GDA0001986101440001351
Figure GDA0001986101440001361
The results show that 13 compounds provided by the invention have NOD1/2 antagonistic activity of more than 80% at 5 mu M, wherein the antagonistic activity L ogIC50 values of the compounds 3,4, 45, 52, 57, 61, 62 on NOD1/2 channels are all below 1 mu M, the antagonistic activity of 17 compounds on NOD1/2 is between 50% and 80%, and the antagonistic activity of 33 compounds on NOD1/2 is lower than 50%, and further analysis shows a structure-activity relationship.
Example 100
Effect of WYZ035-31D (Compound 13 prepared in example 13) on tumor growth in the mouse L ewis Lung cancer model (LL C)
Test article
PTX (batch: JF20080401), WYZ035-31D were all prepared as DMSO stock solutions of 20 ×, and were diluted with physiological saline containing 5% polyoxyethylene castor oil (Cremophor E L) and 5% Tween20, respectively, to the administration concentration immediately before use.
Experimental animals and tumor strains:
the male C57B L/6 mouse is 6-8 weeks old, purchased from Beijing Wintonli Hua laboratory animal technology Limited company with license number SCXK (Jing) 2012-0001, and raised in Qinghua university animal center, and the tumor strain used in the experiment is L ewis lung cancer transplantation tumor of the mouse, which is preserved by passage in the laboratory.
The test method comprises the following steps:
l ewis lung cancer protected C57B L/6 male mice, after dying by cervical dislocation, dissecting and taking out tumor, homogenating under aseptic condition to prepare tumor cell suspension, taking healthy C57B L/6 male mice, inoculating subcutaneous tumor liquid in axilla, 0.1m L/mouse, setting the day of inoculation as D0, after inoculation on day 1 (D1), dividing the mice into 4 groups according to body weight, which are ① solvent Control group (Control), ② WYZ035-31D 20mg/kg group, ③ Paclitaxel (PTX)12mg/kg group, ④ WYZ035-31D (20mg/kg) + PTX (12mg/kg), PTX 12mg/kg intermittent group animals are intravenously injected every 4 days for 1 time, 3 times, WYZ035-31D and Control group animals are injected 1 time daily, 10 times, WYZ035-31D (20mg/kg) + PTX) are injected every 4 days, and intraperitoneal injection is 12mg/kg, 1 time for each day, WYZ035-31D (20 mg/kg).
During the administration, the weight of the animals is measured every 2 days, and the length and the short diameter of the tumor are measured by a vernier caliper according to the formula (1/2) × long diameter × (short diameter)2Tumor size was calculated. The experiment was terminated on day 11 after inoculation (D11), and the mice were sacrificed by cervical dislocation after blood sampling from the eyeballs, dissected, removed the tumor, and weighed. Comparative analysis of the administered group and the control group was performed.
And (3) test results:
compared with the vehicle control group, the tumor volume growth rate of the WYZ035-31D (20mg/kg) and PTX (12mg/kg) group administered alone was slower than that of the control group, but there was no statistical significance, and the WYZ035-31D (20mg/kg) + PTX (12mg/kg) group significantly inhibited the growth of primary tumor and had statistical significance (as shown in FIG. 5). Moreover, the tumor inhibiting effect of the combined group of WYZ035-31D (20mg/kg) + PTX (12mg/kg) is obviously higher than that of the combined group of WYZ035-31D (20mg/kg) and PTX (12mg/kg), and the statistical significance is achieved. Fig. 5A is a photograph of the tumor at the time of the experimental result.
Example 101
Determination of Selective antagonistic Activity of Compound 13NOD1/2 of the examples of the invention
Materials and methods
(1) Cells and reagents
The test substance: WYZ035-31D was formulated as a 20mM DMSO stock solution and diluted with medium to the appropriate concentration just prior to use.
HEK-Blue hNOD1cell, HEK-Blue hNOD2cell, HEK-Blue hT L R2cell, HEK-Blue hT L R4cell and HEK-Dual hTNF- α cell were purchased from InvivoGen.
Reagents hNOD1 agonist C12-ie-DAP (addition of a lauroyl group to the glutamic acid derivative of ie-DAP), hNOD2 agonist Muramyl Dipeptide (MDP), hT L R4 agonist lipopolysaccharide (stabilized lipopolysaccharides, L PS-EK), hT L R2 agonist FS L-1 (synthesized lipoprotein), hTNF α agonist rhTNF α (Recombinant human TNF- α), HEK-Blue detection, QUANTI-Blue, Normolin, Blastin, Zeocin and HEK-Blue Selection Tris are all available from Invivo corporation, DMEM medium, fetal calf serum, peptide-Streptomycin, SRPBS, SRB, Sigma-acetate and Sigma-ice-acetate;
(2) instrument and consumable
Microplate reader (PO L ARstar Omega, USA), horizontal centrifuge (Eppendorf), microscope (O L YMPUS), MACS Separator (Miltenyl), CO2A cell culture box (Thermo fisher), a blood counting chamber (Shanghai refining biochemical instruments, Inc.), a shaker, etc. 96-well plates (Costar, USA), centrifuge tubes (Corning), petri dishes (Corning), and the like.
(3) Detection of HEK-Blue hNOD1 antagonist (HEK-Blue Detection)
HEK-Blue hNOD1 cells were cultured in 50. mu.g/m L streptomyces containing 10% FBS, 50U/m L penicillinin DMEM medium (DMEM complete medium) with 50. mu.g/m L Normocin and 2mM L-glutamine cells in logarithmic growth phase were collected, HEK-Blue Detection solution was used to adjust cell concentration, and the cells were plated in 96-well plates (50000 cells/well) and tested compound gradient working solution and vehicle control were added, 5% CO at 37 ℃ and2after 3h incubation in the incubator, the positive stimulus C12-ie-DAP (final concentration 50ng/m L) was added at 37 ℃ with 5% CO2The incubation was continued in the incubator for 20h and the OD was measured at 655 nm.
The positive control group OD value was designated as C, and the test group OD value was designated as (T).
Antagonism Rate% (Inhibit Rate) [ (C-T)/C]× 100, computing IC50I.e., [ (C-T)/C]× 100 at 50;
after the detection is finished, an SRB method is adopted to examine the influence of the compound on the cell growth, 50 mu L/well is added with 80% TCA4 ℃ for fixing for 1h, deionized water is used for washing the plate for 5 times and is naturally dried, 0.4% SRB is added into 100 mu L/well for dyeing, the plate is placed at room temperature for 10min, 100 mu L/well 1% acetic acid is used for washing the plate for 5 times and is naturally dried, 150 mu L/well is added with Tris base and is oscillated for about 5min, the OD value is detected at 515nm of an enzyme labeling instrument, the OD value of a solvent control group is marked as C, the OD value of a tested group is marked as (T), and the growth rate% (Percentagegrowth) ([ T/C ] × 100.
(4) Detection of HEK-Blue hNOD2 antagonist (HEK-Blue Detection)
The method is the same as the detection of an HEK-Blue hNOD1 antagonist, the cell density is 25000 cells/well, the positive stimulus is MDP (50ng/m L), the incubation time is 18h, and the detection wavelength is 650 nm.
(5) Detection of HEK-Blue hT L R2 antagonist (QUANTI-Blue)
HEK-Blue hT L R2 cells were cultured in DMEM medium (DMEM complete medium) containing 10% FBS, 50U/m L penicillin, 50 μ g/m L streptomycin, 50 μ g/m L Normocin, 2mM L-glutamine, cells were collected in logarithmic growth phase, DMEM complete medium was used to adjust cell concentration, and seeded in 96well plates (30000 cells/well), test compound gradient working medium and vehicle control were added, 37 ℃, 5% CO2After 3h incubation in the incubator, positive stimulus FS L-1 (final concentration 1ng/m L), 37 ℃ with 5% CO was added2The incubation was continued in the incubator.
After 20h, 20 μ L cell supernatants were taken in 96well-plate containing 180 μ L QUANTI-Blue at 37 ℃ with 5% CO2The incubation was developed for 1h in the incubator and then the OD was measured at 650 nm.
The positive control group OD value was designated as C, and the test group OD value was designated as (T).
Antagonism Rate% (Inhibit Rate) [ (C-T)/C]× 100, computing IC50I.e., [ (C-T)/C]× 100 at 50.
After the detection is finished, an SRB method is adopted to examine the influence of the compound on the cell growth, 50 mu L/well is added with 80% TCA4 ℃ for fixing for 1h, deionized water is used for washing the plate for 5 times and is naturally dried, 0.4% SRB is added into 100 mu L/well for dyeing, the plate is placed at room temperature for 10min, 100 mu L/well 1% acetic acid is used for washing the plate for 5 times and is naturally dried, 150 mu L/well is added with Tris base and is oscillated for about 5min, the OD value is detected at 515nm of an enzyme labeling instrument, the OD value of a solvent control group is marked as C, the OD value of a tested group is marked as (T), and the growth rate% (Percentagegrowth) ([ T/C ] × 100.
(6) Detection of HEK-Blue hT L R4 antagonist (QUANTI-Blue)
The method is the same as the detection of an antagonist of HEK-Blue hT L R2, the cell density is 25000 cells/well, the positive stimulus is L PS-EK (1ng/m L), the incubation time is 20h, and the detection wavelength is 650 nm.
(7) Detection of HEK-Blue hTNF α antagonist (QUANTI-Blue)
The method is the same as the detection of an antagonist of HEK-Blue hT L R2, the cell density is 50000 per hole, the positive stimulus is rhTNF α (5ng/m L), the incubation time is 20h, and the detection wavelength is 650 nm.
Test results
As shown in FIG. 6, Compound 13 of the present example has very good antagonistic activity against the NOD1/2 pathway, L omiC50-5.627, -5.379, respectively, and have essentially no antagonistic activity against the T L R2, T L R4, TNF- α pathways, so compound 13 has activity selectively antagonizing the NOD1/2 pathway.
In the description herein, references to the description of the term "one embodiment," "some embodiments," "an example," "a specific example," or "some examples," etc., mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the invention. In this specification, the schematic representations of the terms used above are not necessarily intended to refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples. Furthermore, various embodiments or examples and features of different embodiments or examples described in this specification can be combined and combined by one skilled in the art without contradiction.
Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that variations, modifications, substitutions and alterations can be made to the above embodiments by those of ordinary skill in the art within the scope of the present invention.

Claims (10)

1. A compound which is a compound shown in a formula (I) or a stereoisomer and a pharmaceutically acceptable salt of the compound shown in the formula (I),
Figure FDA0002386159700000011
wherein R is1Is methyl, naphthyl, dibutylamino,
Figure FDA0002386159700000012
R2Is OR6Or NR7R8
R3Is composed of
Figure FDA0002386159700000013
Figure FDA0002386159700000014
R4、R5Each independently is H or C1-3An alkyl group;
R6is H or C1-3An alkyl group;
R7、R8each independently is H, methyl, ethyl, n-propyl, cyclopropyl, n-butyl, tert-butyl, n-dodecyl, n-hexadecyl, n-octadecyl or with R2Nitrogen atom formation in
Figure FDA0002386159700000015
Figure FDA0002386159700000016
Or bridged or spiro rings;
R9is composed of
Figure FDA0002386159700000017
Figure FDA0002386159700000018
Or a vinyl group;
R10is H, methyl,
Figure FDA0002386159700000021
R11Is methyl, benzyl, p-trifluorophenyl alkenyl, 3,4, 5-trimethoxyphenyl, vinyl, 3-methyl-2 butenyl, p-trifluoromethylphenyl, naphthyl, 3, 5-bistrifluoromethylphenyl, cyclopropyl, p-cyanophenyl,
Figure FDA0002386159700000022
Figure FDA0002386159700000023
Y is N, O, S or C1-3An alkyl group;
x is N or O;
X1、X2、X3、X4each independently is H, halogen, methyl, hydroxy, amino, cyano, alkoxy, trifluoromethyl or mercapto;
n is 1,2 or 3.
2. The compound of claim 1, wherein,
R4、R5each independently is H or methyl;
R6is H or methyl;
y is methylene or O;
X1、X2、X3、X4each independently is H, methyl or Cl;
n is 1 or 2.
3. The compound of claim 1, having the structure of one of:
Figure FDA0002386159700000024
Figure FDA0002386159700000031
Figure FDA0002386159700000041
Figure FDA0002386159700000051
Figure FDA0002386159700000061
Figure FDA0002386159700000071
Figure FDA0002386159700000081
Figure FDA0002386159700000091
Figure FDA0002386159700000101
Figure FDA0002386159700000111
Figure FDA0002386159700000121
4. a pharmaceutical composition comprising a compound according to any one of claims 1 to 3.
5. The pharmaceutical composition of claim 4, further comprising a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle, or combination thereof.
6. The pharmaceutical composition of claim 5, further comprising an additional agent for preventing or treating an immunoinflammatory disorder or tumor.
7. The pharmaceutical composition according to claim 6, wherein the other drugs for preventing or treating immunoinflammatory diseases or tumors include but are not limited to melphalan, cyclophosphamide, ifosfamide, busulfan, carmustine, lomustine, streptozotocin, cisplatin, carboplatin, oxaliplatin, dacarbazine, temozolomide, procarbazine, methotrexate, fluorouracil, cytarabine, gemcitabine, mercaptopurine, fludarabine, vinblastine, vincristine, vinorelbine, paclitaxel, docetaxel, topotecan, irinotecan, etoposide, trabectedin, dactinomycin, doxorubicin, epirubicin, daunorubicin, mitoxantrone, bleomycin, mitomycin C, ixabepilone, tamoxifene, flutamide, gonadorelin analogs, megestrol, prednisone, dexamethasone, methylprednisolone, sardine, interferon α, folinic acid, rapamycin, cetocitinib, cetrimitinib, valacyclotinib, valsartan, valacyclotinib, valsartan, valtinib, valsartan, valacyclotinib, valsartan, valtinib, valsartan-145, valsartan, valtinib, valacyclotinib, valtinib hydrochloride, valsartan, valacyclotinib, valtinib, valdecovatinib, valtinib, valdecovatinib, valacyclotinib, valdecovatinib, valtinib, valdecovatinib, valtinib, valdecovatinib, valdecovatib, valdecovatinib, valdecovatib, valdecovatinib, valdecovatib, valdecovatinib, valdecovatib, valdeco.
8. Use of a compound according to any one of claims 1 to 3 or a pharmaceutical composition according to any one of claims 4 to 7 in the manufacture of a medicament for the prevention or treatment of an immunoinflammatory disorder or tumour.
9. Use according to claim 8, wherein the medicament is for the prevention or treatment of rheumatoid arthritis, sjogren's syndrome, wegener's granulomatosis, behcet's disease, sarcoidosis, takayasu's arteritis, reactive arthritis, osteoarthritis, aids, allergic diseases, rheumatoid arthritis, chronic fatigue, type II diabetes, hay fever, lupus erythematosus or multiple sclerosis, colon cancer, rectal cancer, gastric adenocarcinoma, pancreatic cancer, bladder cancer, gallbladder cancer, breast cancer, kidney cancer, renal cell carcinoma, liver cancer, hepatocellular carcinoma, lung cancer, skin cancer, melanoma, thyroid cancer, osteosarcoma, soft tissue sarcoma, head and neck cancer, central nervous system tumor, glioma, glioblastoma, ovarian cancer, uterine cancer, endometrial cancer, prostate cancer, acute myeloid leukemia or acute lymphocytic leukemia or their metastatic cancers, Polycythemia vera, primary hemo-stress arthritis.
10. Use of a compound according to any one of claims 1 to 3 or a pharmaceutical composition according to any one of claims 4 to 7 in the manufacture of a medicament for selectively antagonizing the activation of the NOD1/2 signalling pathway.
CN201710012668.1A 2017-01-09 2017-01-09 Substituted benzodiazacyclo compound and its preparation method and use Active CN106831614B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201710012668.1A CN106831614B (en) 2017-01-09 2017-01-09 Substituted benzodiazacyclo compound and its preparation method and use

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201710012668.1A CN106831614B (en) 2017-01-09 2017-01-09 Substituted benzodiazacyclo compound and its preparation method and use

Publications (2)

Publication Number Publication Date
CN106831614A CN106831614A (en) 2017-06-13
CN106831614B true CN106831614B (en) 2020-07-28

Family

ID=59117434

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710012668.1A Active CN106831614B (en) 2017-01-09 2017-01-09 Substituted benzodiazacyclo compound and its preparation method and use

Country Status (1)

Country Link
CN (1) CN106831614B (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3668844A1 (en) 2017-08-15 2020-06-24 Bayer Aktiengesellschaft 4-oxo-2,3,4,5-tetrahydro-1h-1,5-benzodiazepine-7-carboxamides
CN113354590A (en) * 2020-03-05 2021-09-07 宁波康柏睿格医药科技有限公司 Quinazolinone compound for antagonizing NOD1/2 receptor signal pathway
CN113968792A (en) * 2020-07-24 2022-01-25 郭峰 Novel method for synthesizing 2-chloro-5-aminophenol
AU2021382771A1 (en) * 2020-11-17 2023-07-06 Ningbo Combireg Pharmaceutical Technology Co., Ltd. Deuterated 1,4-benzodiazepine-2,5-dione compound and use thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105693634A (en) * 2016-03-17 2016-06-22 清华大学 Compound and application thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105693634A (en) * 2016-03-17 2016-06-22 清华大学 Compound and application thereof

Also Published As

Publication number Publication date
CN106831614A (en) 2017-06-13

Similar Documents

Publication Publication Date Title
CA2800079C (en) Macrocyclic compounds as trk kinase inhibitors
JP2022087180A (en) Silanol-based therapeutic payloads
EP3802534B1 (en) Tricyclic heterocyclic compounds as sting activators
TWI639602B (en) Tricyclic gyrase inhibitors
CN106831614B (en) Substituted benzodiazacyclo compound and its preparation method and use
CN106589055B (en) Substituted cell acyl dipeptide compound and preparation method and application thereof
CN111018861B (en) Pyrrolopyrimidine compounds
AU2015326936B2 (en) Treatments for autoimmune disease
CN104884458A (en) Fused heterocyclic compounds as protein kinase inhibitors
CN111050765B (en) Spiro compounds and methods of making and using the same
TW201422624A (en) Tricyclic fused thiophene derivatives as JAK inhibitors
SG176461A1 (en) Imidazo[1,2-b]pyridazine and pyrazolo[1,5-a]pyrimidine derivatives and their use as protein kinase inhibitors
CN107787322B (en) Tricyclic compounds and their use as phosphodiesterase inhibitors
US20200039994A1 (en) Heteroaryl amide compounds as sting activators
ES2837018T3 (en) 6,7,8,9-tetrahydro-5H-pyrido [2,3-d] azepine D3 receptor ligands
CN111004214B (en) Pyridylpiperidine derivatives and use thereof
CN110240557B (en) Pyrrolidine amide derivatives and use thereof
CN112312899A (en) Cancer therapy targeting cancer stem cells
US20050215556A1 (en) Macrocyclic kinase inhibitors
JP2021535909A (en) Highly active STING protein agonist compound
CA3015166C (en) 6,7-dihydro-5h-pyrazolo[5,1-b][1,3]oxazine-2-carboxamide compounds
CN111320633A (en) Pyrrole/imidazo six-membered heteroaromatic ring compound and preparation method and medical application thereof
CN112851647B (en) Brefeldin A derivative and preparation method and application thereof
CN112851648B (en) Application of brefeldin A ester derivatives in antitumor drugs
TW202136269A (en) Fused bicyclic derivatives, preparation method and medical use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant