CN106589055B - Substituted cell acyl dipeptide compound and preparation method and application thereof - Google Patents

Substituted cell acyl dipeptide compound and preparation method and application thereof Download PDF

Info

Publication number
CN106589055B
CN106589055B CN201610960010.9A CN201610960010A CN106589055B CN 106589055 B CN106589055 B CN 106589055B CN 201610960010 A CN201610960010 A CN 201610960010A CN 106589055 B CN106589055 B CN 106589055B
Authority
CN
China
Prior art keywords
cancer
compound
group
nod1
optionally substituted
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
CN201610960010.9A
Other languages
Chinese (zh)
Other versions
CN106589055A (en
Inventor
刘刚
马瑶
董毅
王素华
王春廷
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 CN201610960010.9A priority Critical patent/CN106589055B/en
Publication of CN106589055A publication Critical patent/CN106589055A/en
Application granted granted Critical
Publication of CN106589055B publication Critical patent/CN106589055B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06078Dipeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06139Dipeptides with the first amino acid being heterocyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The invention discloses a substituted cell acyl dipeptide compound, a preparation method and application thereof, and further relates to a pharmaceutical composition and application of the compound. The substituted cell acyl dipeptide compound can antagonize inflammatory NF-kB and MAPKs signal transduction pathways mediated by human or murine immunocyte NOD1 and/or NOD2, inhibit the growth of tumors and inhibit the metastasis of tumors.

Description

Substituted cell acyl dipeptide compound and preparation method and application thereof
Technical Field
The invention relates to the field of medicines, in particular to a substituted cell acyl dipeptide compound, a preparation method and application thereof and application of an antagonist in preparing a medicine, and more particularly, the invention relates to a compound obtained by connecting paclitaxel or docetaxel and analogues thereof with muramyl dipeptide through an ether bond, a preparation method and application thereof and application of an antagonist containing the compound in preparing a medicine.
Background
NOD1 and NOD2 belong to the NOD-like receptors (N L Rs) family, are important intrinsic pattern recognition receptors (PPRs) in the body, and like Toll-like receptors (T L Rs), initiate downstream signaling pathways by recognizing exogenous pathogenic molecular patterns (PAMP) or endogenous injury-related molecular patterns (damageassapped molecular patterns (DAMAGeAssociation Patterns, JNMP), thereby initiating innate immune regulation and subsequent acquired immune and inflammatory responses.NOD 45 or NOD2 are expressed mainly in monocytes, macrophages, dendritic cells, neutrophils, epithelial cells and small amounts of T-lymphocytes.NOD 1 recognizes gram-negative degradation products of cell walls, NOD 37 recognizes negative/oligomeric dendritic cell, neutrophil-derived proteins (CDP), and bind to central regulatory domains of the receptor domains (CDRs), such as cDNA-binding domain, CDRs), receptor domains (CDRs), receptor domains of intracellular signaling domains (CDRs), receptor domains of CD-like, and receptor domains (CDRs-like), and are closely related to the receptor binding of intracellular signaling proteins (CDIRC-like) and are shown to promote the interaction of the receptor binding of intracellular CD-like receptor proteins (CDNOD-like) and are known to promote the binding of the central regulatory protein receptor binding of the receptor motif, receptor binding of the receptor motif, receptor binding of CD-like, receptor binding of the receptor motif, receptor binding of the receptor motif, receptor binding to the receptor binding of the receptor motif, receptor of the receptor of.
Studies have shown that the innate immune response initiated by PRRs is critical for the triggering of tumor-associated inflammation, playing a major role in the development of the development (or remodeling) of the tumor immune microenvironment and tumor growth and metastasis, while few studies have been made on the function of N L Rs in promoting or inhibiting tumors, and the relationship of NOD1 or NOD2 to tumors is unclear.
Thus, the NOD1/2 receptor has yet to be 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.
The inventors surprisingly found in experiments that NOD1 or NOD2 knockout mice have significantly reduced tumor weights and metastases relative to wild type mice inoculated with L ewis lung cancer, and thus, the inventors concluded that inhibition of the NOD1 or NOD2 signaling pathway is effective in inhibiting tumor growth or tumor metastasis.
Based on the above, the inventors developed a compound which is a conjugate obtained by connecting paclitaxel or docetaxel and analogues thereof with muramyl dipeptide through an ether bond, and strongly verified that the compound can antagonize inflammatory NF- κ B and MAPKs signaling pathways mediated by human or murine immune cells NOD1 and/or NOD2 through experiments, and has the efficacy of effectively inhibiting the growth of tumors and inhibiting the metastasis of tumors.
In a first aspect of the invention, 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 BDA0001142660300000021
wherein,
Figure BDA0001142660300000022
R1is optionally substituted aryl, optionally substituted alkyl or optionally substituted heteroalkyl,
R2is optionally substituted alkyl, optionally substituted heteroalkyl, hydroxy, cyano, amino, aldehyde or acetoxy,
R3is optionally substituted alkyl, optionally substituted heteroalkyl, halogen, hydroxy, cyano or amino,
ar is optionally substituted aryl or optionally substituted heteroaryl,
R10is halogen, hydroxyl, cyano or amino,
x is optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkenyl, O or S,
R9is an optionally substituted alkyl group, and is,
m and n are respectively and independently 0, 1,2 or 3.
According to the embodiment of the invention, the inventor unexpectedly finds that the compound can antagonize inflammatory NF-kB and MAPKs signal transduction pathways mediated by human or murine immune cells NOD1 and/or NOD2, inhibit the growth of tumors and inhibit the metastasis of the tumors through a plurality of experiments.
In some embodiments of the invention, R0Is composed of
Figure BDA0001142660300000031
Figure BDA0001142660300000032
R1Is optionally substituted phenyl, optionally substituted C1-9Alkyl or optionally substituted C1-9An alkoxy group,
R2is optionally substituted C1-9Alkyl, hydroxyl, cyano, amino, aldehyde or acetoxy,
R3is optionally substituted C1-9Alkyl, hydroxy, cyano or amino,
ar is optionally substituted aryl or optionally substituted heteroaryl,
R9is optionally substituted C1-6An alkyl group, a carboxyl group,
R10is halogen, hydroxyl, cyano or amino,
x is optionally substituted C1-6Alkyl, optionally substituted C1-6An alkenyl group, O or S, or a pharmaceutically acceptable salt thereof,
m and n are respectively and independently 0, 1,2 or 3.
In some embodiments of the invention, R0Is composed of
Figure BDA0001142660300000033
Figure BDA0001142660300000041
R1Is optionally substituted phenyl, optionally substituted C1-3Alkyl or optionally substituted C1-3An alkoxy group,
R2is optionally substituted C1-3Alkyl, hydroxyl, cyano, amino, aldehyde or acetoxy,
R3is optionally substituted C1-3Alkyl, hydroxy, cyano or amino,
ar is optionally substituted phenyl, optionally substituted naphthyl, optionally substituted azanaphthyl or optionally substituted thienyl,
R9is optionally takenSubstituted C1-6An alkyl group, a carboxyl group,
R10is halogen, hydroxyl, cyano or amino,
x is optionally substituted C1-3Alkyl, optionally substituted C1-3An alkenyl group, O or S, or a pharmaceutically acceptable salt thereof,
m and n are respectively and independently 0, 1 or 2.
In some embodiments of the invention, R0Is composed of
Figure BDA0001142660300000042
Figure BDA0001142660300000043
R1Is a phenyl group or a tert-butoxy group,
R2is a hydroxyl group or an acetoxy group,
R3is a hydroxyl group or an amino group,
ar is
Figure BDA0001142660300000044
The R is4、R5、R6、R7、R8Each independently of the other is H or Cl,
R9is a methyl group or a tertiary butyl group,
R10is a hydroxyl group, and the hydroxyl group,
x is O or-CH ═ CH-,
m and n are independently 0 or 1.
In some embodiments of the invention, the compounds proposed by the invention have the structure of one of the following:
Figure BDA0001142660300000051
Figure BDA0001142660300000061
Figure BDA0001142660300000071
Figure BDA0001142660300000081
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 compound can antagonize inflammatory NF-kB and MAPKs signal transduction pathways mediated by human or murine immune cells NOD1 and/or NOD2, inhibit the growth of tumors and inhibit the metastasis of tumors.
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 (carmustine), leucomycin (leucomycin), ciclopirox (ciclopirox), ciclovir (ciclopirox), ciclopiroxan), ciclopirox (ciclopirox), ciclovir (ciclovir), ciclovir (ciclopirox (ciclovir), ciclovir (ciclovir), ciclovir (ciclopirox (ciclovir), ciclovir (ciclovir), ciclopirox (ciclovir), ciclovir (ciclovir), ciclopirox (ciclovir), ciclopirox (ciclovir), ciclovir (ciclopirox (ciclovir), ciclovir (ciclopirox (ciclovir), ciclovir (ciclovir), ciclovir (ciclovir), ciclopirox (ciclovir), ciclovir (ciclovir), ciclovir (ciclovir), ciclovir (ciclovir), ciclovir (ciclovir), ciclovir (ciclovir), ciclovir (ciclovir), ciclovir (dox (ciclovir), ciclovir (dox (peripheral), ciclovir (peripheral), ciclovir (dox (peripheral), ciclovir (peripheral), ciclovir), ciclopirox (peripheral), ciclovir (peripheral), ciclovir (peripheral), ciclovir (peripheral), ciclovir), ciclopirox (peripheral), ciclovir (peripheral), ciclopirox (peripheral), ciclovir (peripheral), ciclovir (peripheral), ciclovir), ciclopirox (peripheral), ciclovir (peripheral), ciclopirox (peripheral), ciclovir (peripheral), ciclovir (peripheral), ciclopirox (peripheral), ciclovir (peripheral), ciclopirox (peripheral), ciclovir (peripheral), ciclopirox (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 drug can antagonize inflammatory NF-kB and MAPKs signal transduction pathways mediated by human or murine immune cells NOD1 and/or NOD2, inhibit the growth of tumors and inhibit the metastasis of tumors.
In a fourth aspect, the invention provides the use of a compound as defined above or a pharmaceutical composition as defined above in the manufacture of a medicament for antagonizing inflammatory NF-. kappa.B and MAPKs signalling pathways mediated by human or murine immune cells NOD1 and/or NOD 2.
In a fifth aspect, the present invention provides the use of an antagonist for the preparation of a medicament for the prevention or treatment of an immunoinflammatory disorder or tumor, wherein the antagonist is for antagonizing at least one of (1) NOD1, (2) NOD2, (3) NF-. kappa.B signaling pathway, and (4) MAPKs signaling pathway the inventors have found that both tumor weight and metastasis are significantly reduced after L ewis lung cancer vaccination of NOD1 or NOD2 knockout mice relative to wild type mice, and thus, the inventors have concluded that by antagonizing NOD1 or NOD2 signaling pathway, tumor growth or tumor metastasis can be effectively inhibited.
In some embodiments of the invention, the antagonist comprises a compound described above.
In some embodiments of the invention, the antagonist is for preventing or treating an immunoinflammatory disorder or tumor.
In some embodiments of the invention, the antagonist is used to prevent or treat 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 addition, in a sixth aspect of the present invention, the present invention provides methods for the preparation, isolation, purification and characterization of the above compounds.
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 is the result of PCR gene identification of NOD1 gene knock-out mice according to an embodiment of the present invention;
FIG. 2 shows the results of experiments on tumor growth or metastasis in NOD1 and NOD2 gene knock-out mice according to the present invention; wherein A is tumor weight, B is lung metastasis count, and C is lung metastasis picture;
FIG. 3 is the results of an antagonistic experiment of the compound DY-16-43 against NOD1 and NOD2 according to the example of the present invention,
wherein, A is the result of experiments of inhibition of compound DY-16-43 on hNOD1 and hNOD2, and B is the result of experiments of cytotoxicity of compound DY-16-43;
FIG. 4 is an experimental result that Compound DY-16-43 antagonizes the agonistic effect of C12-ie-DAP on human peripheral blood-derived macrophage NOD1 according to the present example;
FIG. 5 is an experimental result showing that Compound DY-16-43 antagonizes the agonistic effect of MDP on human peripheral blood-derived macrophage NOD2, according to the present invention;
FIG. 6 is a graph showing the results of experiments in which compound DY-16-43 inhibits the expression of NOD1/2 signaling pathway-related protein after stimulation with a stimulator, wherein A is the results of experiments in which C12-ie-DAP stimulates the expression of related protein, and B is the results of experiments in which MDP stimulates the expression of related protein, according to examples of the present invention;
FIG. 7 is an experimental result of the effect of compound DY16-43 on spontaneous metastasis of mouse L ewis lung cancer model (LL C) according to an embodiment of the present invention, wherein A is a graph of tumor volume increase change, B is tumor weight, C is the number of lung metastasis nodules in tumor-bearing mice, and D is a photograph of lung metastasis.
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 "alkoxy" means an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkoxy group contains 1 to 12 carbon atoms. In one embodiment, the alkoxy group contains 1 to 6 carbon atoms; in another embodiment, the alkoxy group contains 1 to 4 carbon atoms; in yet another embodiment, the alkoxy group contains 1 to 3 carbon atoms. The alkoxy group may be optionally substituted with one or more substituents described herein.
Examples of alkoxy groups include, but are not limited to, methoxy (MeO, -OCH)3) Ethoxy (EtO, -OCH)2CH3) 1-propoxy (n-PrO, n-propoxy, -OCH)2CH2CH3) 2-propoxy (i-PrO, i-propoxy, -OCH (CH)3)2) 1-butoxy (n-BuO, n-butoxy, -OCH)2CH2CH2CH3) 2-methyl-l-propoxy (i-BuO, i-butoxy, -OCH)2CH(CH3)2) 2-butoxy (s-BuO, s-butoxy, -OCH (CH)3)CH2CH3) 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC (CH)3)3) 1-pentyloxy (n-pentyloxy, -OCH)2CH2CH2CH2CH3) 2-pentyloxy (-OCH (CH)3)CH2CH2CH3) 3-pentyloxy-OCH (CH)2CH3)22-methyl-2-butoxy (-OC (CH))3)2CH2CH3) 3-methyl-2-butoxy (-OCH (CH)3)CH(CH3)2) 3-methyl-l-butoxy (-OCH)2CH2CH(CH3)2) 2-methyl-l-butoxy (-OCH)2CH(CH3)CH2CH3) And so on.
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. In one embodiment, 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 includeIncluding, but not limited to, vinyl (-CH ═ CH)2) Allyl (-CH)2CH=CH2) And so on.
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.Symposium Series,Edward B.Roche,ed.,BioreversibleCarriers in Drug Design,American Pharmaceutical Association and PergamonPress,1987,J.Rautio et al.,Prodrugs:Design and Clinical Applications,NatureReview Drug Discovery,2008,7,255-270,and S.J.Hecker et al.,Prodrugs ofPhosphates and Phosphonates,Journal of Medicinal Chemistry,2008,51,2328-2345。
"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.
As used herein, "pharmaceutically acceptable salts" refer to organic and inorganic salts of the compounds of the present 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, ascorbates, aspartates, benzoates, bisulfates, salts of gluconic acidSugar acid salts, hemisulfate salts, heptanoate salts, hexanoate salts, hydroiodide salts, 2-hydroxy-ethanesulfonate salts, lactobionate salts, lactate salts, laurate salts, lauryl sulfate salts, malate salts, methanesulfonate salts, 2-naphthalenesulfonate salts, nicotinate salts, nitrate salts, oleate salts, palmitate salts, embonate salts, pectate salts, persulfate salts, 3-phenylpropionate salts, picrate salts, pivalate salts, propionate salts, stearate salts, thiocyanate salts, p-toluenesulfonate salts, undecanoate salts, valerate salts, 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 conjugate obtained by connecting taxol or docetaxel and analogues thereof with Muramyl Dipeptide (MDP) simplifier (MDA analogue) through a non-cleavage connecting bridge (ether bond), and pharmaceutically acceptable salts, pharmaceutical preparations and compositions thereof can be used as antagonists of NOD1/2 signal transduction pathways, and can be used for treating human epidemic inflammatory diseases or tumors, such as rheumatoid arthritis, sjogren's syndrome, Wegener's granulomatosis, Behcet's disease, sarcoidosis, Takayasu's disease, reactive arthritis, osteoarthritis, AIDS, allergic diseases, rheumatic 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, colon cancer of the lung cancer, colon cancer of the pancreas, bladder cancer of the stomach, colon cancer of the head of the liver, liver, The treatment of lung cancer, skin cancer, melanoma, thyroid cancer, osteosarcoma, soft tissue sarcoma, head and neck cancer, central nervous system tumors, 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-responsive arthritis, 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 of the invention, 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 BDA0001142660300000181
wherein,
R0is composed of
Figure BDA0001142660300000182
Figure BDA0001142660300000183
R1Is optionally substituted aryl, optionally substituted alkyl or optionally substituted heteroalkyl,
R2is optionally substituted alkyl, optionally substituted heteroalkyl, hydroxy, cyano, amino, aldehyde or acetoxy,
R3is optionally substituted alkyl, optionally substituted heteroalkyl, halogen, hydroxy, cyano or amino,
ar is optionally substituted aryl or optionally substituted heteroaryl,
R10is halogen, hydroxyl, cyano or amino,
x is optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkenyl, O or S,
R9is an optionally substituted alkyl group, and is,
m and n are respectively and independently 0, 1,2 or 3.
In some embodiments of the invention, R0Is composed of
Figure BDA0001142660300000184
Figure BDA0001142660300000191
R1Is optionally substituted phenyl, optionally substituted C1-9Alkyl or optionally substituted C1-9An alkoxy group,
R2is optionally substituted C1-9Alkyl, hydroxyl, cyano, amino, aldehyde or acetoxy,
R3is optionally substituted C1-9Alkyl, hydroxy, cyano or amino,
ar is optionally substituted aryl or optionally substituted heteroaryl,
R9is optionally substituted C1-6An alkyl group, a carboxyl group,
R10is halogen, hydroxyl, cyano or amino,
x is optionally substituted C1-6Alkyl, optionallySubstituted C1-6An alkenyl group, O or S, or a pharmaceutically acceptable salt thereof,
m and n are respectively and independently 0, 1,2 or 3.
In some embodiments of the invention, R0Is composed of
Figure BDA0001142660300000192
Figure BDA0001142660300000193
R1Is optionally substituted phenyl, optionally substituted C1-3Alkyl or optionally substituted C1-3An alkoxy group,
R2is optionally substituted C1-3Alkyl, hydroxyl, cyano, amino, aldehyde or acetoxy,
R3is optionally substituted C1-3Alkyl, hydroxy, cyano or amino,
ar is optionally substituted phenyl, optionally substituted naphthyl, optionally substituted azanaphthyl or optionally substituted thienyl,
R9is optionally substituted C1-6An alkyl group, a carboxyl group,
R10is halogen, hydroxyl, cyano or amino,
x is optionally substituted C1-3Alkyl, optionally substituted C1-3An alkenyl group, O or S, or a pharmaceutically acceptable salt thereof,
m and n are respectively and independently 0, 1 or 2.
In some embodiments of the invention, R0Is composed of
Figure BDA0001142660300000194
Figure BDA0001142660300000201
R1Is a phenyl group or a tert-butoxy group,
R2is a hydroxyl group or an acetoxy group,
R3is a hydroxyl group or an amino group,
ar is
Figure BDA0001142660300000202
The R is4、R5、R6、R7、R8Each independently of the other is H or Cl,
R9is a methyl group or a tertiary butyl group,
R10is a hydroxyl group, and the hydroxyl group,
x is O or-CH ═ CH-,
m and n are independently 0 or 1.
In some embodiments of the invention, the compounds proposed by the invention have the structure of one of the following:
Figure BDA0001142660300000203
Figure BDA0001142660300000211
Figure BDA0001142660300000221
Figure BDA0001142660300000231
Figure BDA0001142660300000241
according to the embodiment of the invention, the inventor unexpectedly discovers through a large number of experiments that the compound can antagonize inflammatory NF-kB and MAPKs signal transduction pathways mediated by human or murine immunocyte NOD1 and/or NOD2, effectively inhibit the growth of tumors and inhibit the metastasis of the tumors.
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 compounds are 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 the immunoinflammatory disease or tumor include, but are not limited to, melphalan (melphalan), cyclophosphamide (cyclophosphamide), ifosfamide (ifosfamide), busulfan (busufan), carmustine (carmustine), amoxicillin (rituximab), ciclopirox (bleomycin), streptozotocin (streptozotocin), cisplatin (cissplastin), carboplatin (carboplatin), oxaliplatin (oxaliplatin), dacarbazine (leucomycin), amoxicillin (paclitaxel), cetuximab (neomycin), paclitaxel (cetib), paclitaxel (cetirizine), ciclopirox (bleomycin), ciclopirox (ritorinib), ciclopirox (morphine), ciclovir (morphine), ciclopirox (morphine), ciclovir (morphine), ciclovir (morphine (dox (morphine), ciclovir (dox), ciclovir (dox (ciclovir), ciclovir (ciclovir), ciclovir (dox (ciclovir), ciclovir (dox), or (dox), ciclovir), or (dox), or (dox), or (dox), or (dox), or (dox), or (dox), or (e), or dox), or (dox), or (e), or (or dox), or (dox), or (e), or (dox), or (e), or (e), or (e), or (e), or (e), or (e), or (or), or (e), or (or), or (e), or (e), or (or), or (e), or (or), or (or), or (e), or (or), or (e), or (e), or (or), or (e), or (or), or (e), or (or), or (e), or (or), or (e), or (or), or (e), or (or), or (e), or (or), or (e), or (e), or (or), or.
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 those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcellulose, hydroxypropylmethylcellulose and polyvinylpyrrolidone. Suitable emulsifiers include those described herein, including polyoxyethyleneSuitable 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(s) ((R))
Figure BDA0001142660300000331
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 compound disclosed herein and one or more finely powdered pharmaceutically acceptable excipients. 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.
Use of the antagonists of the invention for the preparation of a medicament
The present invention proposes the use of an antagonist for the preparation of a medicament for the prevention or treatment of an immunoinflammatory disorder or tumour, for antagonizing at least one of the following: (1) NOD 1; (2) NOD 2; (3) the NF- κ B signaling pathway; and (4) the MAPKs signaling pathway.
In some embodiments of the invention, the antagonist comprises at least one of a compound described above, a small interfering nucleotide encoding a gene that silences the NOD1 or the NOD2, an antisense nucleotide, or a protein, polypeptide, antibody, and active organic compound that antagonizes the NOD1 or the NOD 2.
In some embodiments of the invention, the antagonist is for preventing or treating an immunoinflammatory disorder or tumor.
In some embodiments of the invention, the antagonist is used to prevent or treat 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.
As can be appreciated by those skilled in the art, the antagonists can be used alone or in combination with other active ingredients or adjuvants to make a medicament. The antisense oligonucleotide is a single-stranded structure with the length of 14-30bp, and has more than 90% of identity with a matched nucleotide sequence in a coding gene of the NOD1 or NOD2 protein. The antibody can be a monoclonal antibody or a polyclonal antibody.
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, Beijing Bomijie reagents, Acros Organics, Alfa Aesar, Sigma-Aldrich and TCI, unless otherwise specified, and were used without purification. The solvents used in the experiments were purchased mainly from Beijing chemical plant and Shinga chemical Co., Ltd, except that THF, DMF and dichloromethane were further processed by solvent purification system of VAC corporation, USA, and all solvents were used without treatment. 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 chromatography column Kromasil C18 analytical column (4.6 μm,4.6mm × 50mm), available from DIKMA Inc. the mobile phase was acetonitrile containing 0.05% HCOOH and water.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 254nm ThermoFinnigan L CQ-AdvantaAnd (5) dividing 5% of eluent into a mass spectrometer, and performing electrospray ionization (ESI) by adopting a positive ion or negative ion scanning mode. The method is mainly used for reaction monitoring and primary determination of compound purity.
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 BDA0001142660300000381
BEH C18 column (1.7 μm,2.1mm × 50 mm.) mobile phase was acetonitrile containing 0.05% HCOOH and water linear gradient elution 5:95(v: v) acetonitrile-H2O to 95:5(v: v) acetonitrile-H2O, time is 3minutes, flow rate is 0.3m L/min, UV detection wavelength is 254nm, SQ mass spectrum detector adopts positive ion or negative ion scanning mode, and electrospray ionization source (ESI) 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.
High resolution Mass spectrometer Agilent L C/MSD TOF System chromatographic column Agilent ZORBAX SB-C18(Rapid resolution,3.5 μm, 2.130 mm) mobile phase MeOH H2O75: 25(v: v), containing 5 mmol/L formic acid, isocratic elution, time 5min, flow rate 0.40m L/min, mass spectrometry detection by positive ion scanning, electrospray ionization (ESI), mainly used to determine the exact molecular weight of the target compound.
Nuclear magnetic resonance apparatus: varian Mercury 300MHz,400MHz,500MHz,600MHz and Bruker Avance 35400MHz, solvent CDCl3,DMSO-d6,acetone-d6or methanol-d4。
Melting point apparatus: yanaco micro melting point apparatus, OptiMelt
Typical synthetic procedures for preparing the disclosed compounds of the invention are shown in the following synthetic schemes. Unless otherwise stated, each R1、R2Having the definition as described in the invention, R means R4、R5、R6、R7、R8And has the definitions as described herein.
Intermediate 1-3 synthetic schemes
Figure BDA0001142660300000391
General procedure for the selective alkylation of 2' -OH including paclitaxel and docetaxel reactions:
the first step of reaction (reaction a) is that paclitaxel and docetaxel are directly used as starting materials and are subjected to addition reaction with benzyl propiolate under the action of organic base (N-methylmorpholine) to obtain corresponding addition products 1-2 with an alkenyloxy structure, wherein the N-methylmorpholine and the benzyl propiolate are 1.05 equivalent, the reaction is carried out at room temperature, the reaction can be monitored through L C-Ms or T L C, the reaction time is 4 hours, and the intermediate 1-2 (white powder) can be obtained by directly purifying through column chromatography.
And (b) in the second step, removing a benzyl protecting group and simultaneously reducing unsaturated double bonds to obtain an intermediate 1-3 under the condition of catalytic hydrogenation, adjusting the reaction temperature to between room temperature and 50 ℃ according to the dosage of Pd/C, using ethyl acetate or methanol as a solvent, carrying out hydrogenation under normal pressure, monitoring the reaction by L C-Ms or T L C for 12 hours, and filtering out Pd/C and then concentrating to directly obtain the target intermediate 1-3 (white powder).
Intermediate 1-4 synthetic schemes
Summary the solid phase synthesis of a terminal carboxyl Muramyl Dipeptide (MDP) shorte (MDA analogue) was performed:
Figure BDA0001142660300000401
putting Wang resin into a solid phase reactor, adding DCM into the reactor, and dissolvingAfter 1H of resin swelling, draining off the solvent, adding 1.5 equivalents of Fmoc-L ys (Boc) -OH, 1.5 equivalents of HOBt,1.5 equivalents of DIC and DMF to the reactor, after completion of the detection reaction by the rocking bed reaction 8H (introduction of the first amino acid to the resin), draining off the reaction solution, washing twice with DCM and DMF, adding a 20% piperidine-containing DMF solution, removing the Fmoc protecting group, after 1H of reaction, draining off the reaction solution, washing three times with DCM and DMF, draining off the reaction solution, adding 1.5 equivalents of Fmoc-D-iso-Gln-OH, 1.5 equivalents of HOBt,1.5 equivalents of DIC and DMF, after 8H of rocking bed reaction (introduction of the second amino acid to the resin), detecting the reaction by the draining off the reaction solution, washing twice with DCM and DMF, adding a 20% piperidine-containing DMF, after 1H of reaction, draining off the equivalent of DCM and DMF, after 1.5H of the reaction solution, adding three equivalents of DCM and DMF, after 5% of DCM, after removal of the reaction solution, adding the reaction solution, after 5H of the detection reaction solution, adding three times with DCM and DMF, after 5% of the reaction solution, adding the reaction solution, after 5% of DCM and DMF, the reaction solution, adding three equivalents of DCM, the reaction solution, adding DCM and DMF, the reaction solution, adding three reaction solution, the reaction solution of DCM, the reaction solution of DCM and DMF, the reaction solution of DCM, the reaction solution of DCM, the reaction solution of the reaction, the reaction solution of DCM, the reaction, the2And (3) cracking the solution O for 2 hours, collecting lysate, washing the resin with DCM, merging filtrate and lysate, evaporating the solvent to dryness under reduced pressure, adding anhydrous ether under ice bath, allowing a large amount of precipitates to appear in the system, standing for 1 hour, taking out supernatant, continuously adding anhydrous ether, performing ultrasonic washing for multiple times, filtering, and draining to obtain a white solid, namely the target product. The next reaction was carried out without purification.
Intermediate 1-5 synthetic schemes
Summary the solid phase synthesis of the terminal amido Muramyl Dipeptide (MDP) simplifications (MDA analogs) was performed:
Figure BDA0001142660300000411
the synthesis of terminal amido MDP analogs was performed following the synthesis of intermediates 1-4 starting from Rink-amine based resin and will not be described here, except that the final cleavage was performed using 90% TFA/DCM solution.
Intermediate 1-6 synthetic schemes
A synthetic method is described that includes conjugation of paclitaxel and docetaxel to MDP analogs via a non-cleavable linking bridge:
Figure BDA0001142660300000412
dissolving the intermediate 1-3, 1.05 equivalents of HOSU and 1.05 equivalents of EDCI in DMSO, reacting at room temperature, completely converting into corresponding active ester, adding MDP derivative and N-methylmorpholine, reacting at room temperature, detecting by L C-Ms, and purifying by a reverse phase column to obtain white solid.
The invention will now be described with reference to specific examples, which are intended to be illustrative only and not to be limiting in any way.
Example 1
Synthesis of Compound 1 (accession number Z L T-03-14)
Figure BDA0001142660300000413
Paclitaxel (1mmol,853mg) is dissolved in 10m L dichloromethane, 1.05 equivalents, 110 mu L N-methylmorpholine and 160mg benzyl propiolate are added at room temperature, after 4 hours of room temperature reaction, L C-Ms detect complete reaction, and then column chromatography purification is directly carried out to obtain 912mg of intermediate 1-2a (white solid powder), and the yield is 90%.
507mg of intermediate 1-2a is weighed and dissolved in 10m L ethyl acetate, 507mg of Pd/C is added, the reaction is carried out at 50 ℃ under the hydrogen of one atmosphere, the progress of the reaction is monitored through L C-Ms, after the reaction is completed, the Pd/C is filtered, a filter cake is washed twice by ethyl acetate, the filtrates are combined and concentrated to obtain 460mg of intermediate 1-3a (white solid powder), the yield is 99%, no further purification is needed, and the intermediate is directly used for the subsequent reaction.
Weighing 1g (1.0mmol) Wang resin into a solid phase reactor, adding 10m L DCM into the reactor, swelling the resin for 1H, draining off the solvent, adding 1.5 equivalents (0.703g) Fmoc-L ys (Boc) -OH, 1.5 equivalents (0.202g) HOBt,1.5 equivalents (0.23m L) DIC and 10m L DMF into the reactor, introducing the first amino acid into the resin, shaking table reacting for 8H, draining off the reaction solution, washing twice with DCM and DMF, adding 20% piperidine-containing DMF solution, removing the Fmoc protecting group, draining off the reaction solution for 1H, draining off the reaction solution, washing three times with DCM and DMF, draining off the reaction solution, adding 1.5 equivalents (0.553g) Fmoc-D-iso-Gln-OH, draining off the reaction solution for 1.5 equivalents (0.5 equivalents) DCM, draining off the reaction solution, draining off the reaction2O solution, cracking for 2h, collecting lysate, washing resin with 20m L DCM, mixing filtrate with lysate, evaporating solvent under reduced pressure, adding anhydrous ether under ice bath, and making systemA large amount of precipitate appears in the solution, the solution is kept stand for 1h, supernatant is taken out, absolute ethyl ether is continuously added for ultrasonic washing for a plurality of times, and filtration and pumping-out are carried out to obtain 0.52g of white solid 1-4a with the yield of 84%. The next reaction was carried out without purification.
Weighed 0.05mmol,46mg of intermediate 1-3a, 1.0 equivalent (5.8mg) of HOSU, 1.0 equivalent (9.6mg) of EDC hydrochloride, dissolved in 0.5m L DMSO and reacted at room temperature, after completion of the reaction checked by L C-Ms, 31mg of intermediate 1-4a and 22. mu. L N-methylmorpholine were added, and after 30 minutes of reaction checked by completion of the reaction, purified directly by reverse phase column chromatography to give 57mg of Compound 1 (white powder) in 80% yield.1HNMR(400MHz,DMSO-d6)1.00(s,3H),1.02(s,3H),1.20-1.31(m,7H),1.42-1.53(m,6H),1.63-1.65(m,1H),1.66-1.77(m,2H),1.81(s,3H),1.90-2.03(m,1H),2.10-2.20(m,8H),2.26-2.34(m,1H),2.34-2.42(m,2H),2.85-2.94(m,2H),3.61(d,1H,J=6.7Hz),3.70-3.84(m,2H),3.95-4.20(m,5H),4.35-4.48(m,2H),4.67(s,1H),4.88-5.11(m,2H),5.31(t,1H,J=8.5Hz),5.40(d,1H,J=6.7Hz),5.89(t,1H,J=8.8Hz),6.31(s,1H),6.88(d,1H,J=15.9Hz),7.10(s,1H),7.16-7.24(m,1H),7.32-7.37(m,1H),7.37-7.44(m,5H),7.47-7.57(m,4H),7.62-7.80(m,5H),7.86(d,J=7.1Hz,3H),7.95(d,J=7.4Hz,3H),8.26(d,J=7.6Hz,1H),8.52(d,J=6.2Hz,1H),9.03(d,J=7.4Hz,1H).
Example 2
Synthesis of Compound 2 (accession number Z L T-03-15)
Figure BDA0001142660300000431
The title compound 2 was prepared according to the procedure described for example 1 using intermediate 1-3a to react with a p-methyl cinnamic acid substituted carboxy terminal MDP derivative.1HNMR(400MHz,DMSO-d6)1.00(s,3H),1.02(s,3H),1.20-1.33(m,7H),1.44-1.52(m,6H),1.61-1.66(m,1H),1.66-1.78(m,2H),1.80(s,3H),1.91-2.02(m,1H),2.07-2.20(m,8H),2.26-2.35(m,4H),2.35-2.42(m,2H),2.84-2.96(m,2H),3.60(d,1H,J=6.7Hz),3.70-3.85(m,2H),3.95-4.18(m,5H),4.38-4.45(m,2H),4.66(s,1H),4.90(d,J=9.3Hz,1H),4.95(1H,brs),5.30(t,1H,J=8.7Hz),5.40(d,1H,J=6.9Hz),5.88(t,1H,J=8.5Hz),6.31(s,1H),6.70(d,1H,J=15.7Hz),7.09(s,1H),7.17-7.25(m,3H),7.32(s,1H),7.37-7.42(m,4H),7.42-7.52(m,4H),7.52-7.58(m,1H),7.62-7.68(m,2H),7.74(t,J=7.1Hz,1H),7.86(d,J=7.5Hz,3H),7.94(d,J=7.5Hz,3H),8.22(d,J=7.8Hz,1H),8.32(d,J=6.2Hz,1H),9.02(d,J=7.4Hz,1H).
Example 3
Synthesis of Compound 3 (accession number Z L T-03-18)
Figure BDA0001142660300000432
The title compound 3 was prepared according to the procedure described for example 1 using intermediate 1-3a to react with a 3-4-difluorocinnamic acid substituted carboxy-terminal MDP derivative.1HNMR(400MHz,DMSO-d6)1.01(s,3H),1.03(s,3H),1.21-1.31(m,7H),1.42-1.55(m,6H),1.58-1.65(m,1H),1.70-1.82(m,5H),1.91-2.03(m,1H),2.11-2.20(m,8H),2.26-2.35(m,1H),2.35-2.42(m,2H),2.87-2.94(m,2H),3.60(d,1H,J=6.8Hz),3.72-3.84(m,2H),3.88-4.05(m,3H),4.06-4.19(m,2H),4.37-4.52(m,2H),4.68(s,1H),4.91(d,J=9.0Hz,1H),4.95-5.15(brs,1H),5.32(t,1H,J=8.6Hz),5.40(d,1H,J=6.7Hz),5.88(t,1H,J=8.5Hz),6.32(s,1H),6.83(d,1H,J=15.6Hz),7.02(s,1H),7.17-7.25(m,1H),7.35-7.52(m,9H),7.53-7.58(m,1H),7.60-7.80(m,5H),7.88(d,J=7.2Hz,3H),7.95(d,J=7.5Hz,3H),8.31(d,J=6.2Hz,1H),8.52(d,J=6.5Hz,1H),9.16(d,J=7.2Hz,1H).
Example 4
Synthesis of Compound 4 (accession number Z L T-03-19A)
Figure BDA0001142660300000441
The title compound 4 was prepared according to the procedure described for example 1 using intermediate 1-3a to react with a p-chlorocinnamic acid substituted carboxy terminal MDP derivative.1HNMR(400MHz,DMSO-d6)1.00(s,3H),1.02(s,3H),1.20-1.30(m,7H),1.43-1.53(m,6H),1.62-1.65(m,1H),1.67-1.77(m,2H),1.80(s,3H),1.92-2.02(m,1H),2.11(s,3H),2.16(s,5H),2.26-2.34(m,1H),2.35-2.42(m,2H),2.85-2.95(m,2H),3.60(d,1H,J=6.8Hz),3.70-3.85(m,2H),3.95-4.20(m,5H),4.35-4.45(m,2H),4.67(s,1H),4.90(d,J=9.4Hz,1H),4.98(1H,brs),5.30(t,1H,J=8.6Hz),5.39(d,1H,J=6.9Hz),5.88(t,1H,J=8.2Hz),6.31(s,1H),6.78(d,1H,J=15.8Hz),7.08(s,1H),7.16-7.24(m,1H),7.36(s,1H),7.37-7.43(m,5H),7.44-7.52(m,4H),7.54(d,J=7.0Hz,1H),7.59(d,J=8.4Hz,2H),7.65(t,J=7.3Hz,2H),7.70-7.76(m,1H),7.82-7.88(m,3H),7.89-7.99(m,3H),8.25(d,J=7.9Hz,1H),8.40(d,J=6.7Hz,1H),9.05(d,J=7.9Hz,1H).
Example 5
Synthesis of Compound 5 (accession number Z L T-03-19B)
Figure BDA0001142660300000451
The title compound 5 was prepared according to the procedure described for example 1 using intermediate 1-3a to react with a p-fluorocinnamic acid substituted carboxy-terminal MDP derivative.1HNMR(400MHz,DMSO-d6)0.99(s,3H),1.02(s,3H),1.15-1.30(m,7H),1.40-1.55(m,5H),1.55-1.68(m,2H),1.70-1.85(m,5H),1.92-2.02(m,1H),2.08-2.22(m,8H),2.26-2.35(m,1H),2.35-2.42(m,2H),2.85-2.95(m,2H),3.59(d,1H,J=6.8Hz),3.71-3.84(m,2H),3.90-4.05(m,3H),4.05-4.18(m,2H),4.35-4.45(m,2H),4.68(s,1H),4.90(d,J=9.4Hz,1H),5.08(1H,brs),5.30(t,1H,J=8.6Hz),5.39(d,J=7.0Hz,1H),5.87(t,J=8.7Hz,1H),6.31(s,1H),6.75(d,J=15.8Hz,1H),7.07(s,1H),7.16-7.30(m,3H),7.37-7.45(m,6H),7.45-7.52(m,2H),7.53-7.57(m,1H),7.60-7.70(m,4H),7.70-7.76(m,1H),7.84-7.94(m,6H),8.34(d,J=7.7Hz,1H),8.46(d,J=6.6Hz,1H),9.15(d,J=7.8Hz,1H).
Example 6
Synthesis of Compound 6 (accession number Z L T-03-20A)
Figure BDA0001142660300000452
The title compound 6 was prepared according to the procedure described for example 1 using intermediate 1-3a to react with a2, 4-difluorocinnamic acid substituted carboxy-terminal MDP derivative.1HNMR(400MHz,DMSO-d6)1.00(s,3H),1.02(s,3H),1.20-1.33(m,7H),1.45-1.53(m,6H),1.63-1.67(m,1H),1.66-1.78(m,2H),1.80(s,3H),1.90-2.01(m,1H),2.10-2.21(m,8H),2.26-2.35(m,1H),2.36-2.42(m,2H),2.84-2.94(m,2H),3.59(d,1H,J=7.2Hz),3.71-3.84(m,2H),3.96-4.21(m,5H),4.35-4.47(m,2H),4.68(s,1H),4.90(d,J=9.6Hz,1H),5.04(brs,1H),5.30(t,1H,J=8.5Hz),5.39(d,1H,J=7.0Hz),5.87(t,1H,J=8.8Hz),6.32(s,1H),6.85(d,1H,J=15.8Hz),7.07(s,1H),7.16-7.24(m,1H),7.32-7.37(m,1H),7.37-7.44(m,5H),7.45-7.51(m,3H),7.52-7.56(m,1H),7.62-7.67(m,3H),7.71-7.75(m,2H),7.87(d,J=7.5Hz,3H),7.94(d,J=7.7Hz,3H),8.32(d,J=7.3Hz,1H),8.57(d,J=6.8Hz,1H),9.14(d,J=8.2Hz,1H).
Example 7
Synthesis of Compound 7 (accession number Z L T-03-20B)
Figure BDA0001142660300000461
The title compound 7 was prepared according to the procedure described for example 1 using intermediate 1-3a to react with a para-bromocinnamic acid substituted carboxy terminal MDP derivative.1HNMR(400MHz,DMSO-d6)1.00(s,3H),1.02(s,3H),1.20-1.31(m,7H),1.42-1.52(m,6H),1.62-1.66(m,1H),1.66-1.77(m,2H),1.80(s,3H),1.92-2.02(m,1H),2.08-2.20(m,8H),2.26-2.34(m,1H),2.35-2.41(m,2H),2.84-2.96(m,2H),3.59(d,1H,J=6.8Hz),3.70-3.85(m,2H),3.97(d,J=7.8Hz,1H),4.02(d,J=7.9Hz,1H),4.05-4.17(m,3H),4.41(d,J=8.5Hz,2H),4.67(s,1H),4.90(d,J=9.4Hz,1H),4.96(1H,brs),5.29(t,1H,J=8.7Hz),5.39(d,1H,J=6.9Hz),5.87(t,1H,J=8.5Hz),6.30(s,1H),6.78(d,1H,J=15.8Hz),7.11(s,1H),7.17-7.23(m,1H),7.32(s,1H),7.35-7.45(m,5H),7.46-7.56(m,5H),7.58-7.69(m,4H),7.74(t,J=7.1Hz,1H),7.85(d,J=6.9Hz,3H),7.94(d,J=7.7Hz,2H),8.06(d,J=6.1Hz,1H),8.24(d,J=7.9Hz,1H),8.38(d,J=6.5Hz,1H),9.02(d,J=7.9Hz,1H),12.49(brs,1H).
Example 8
Synthesis of Compound 8 (accession number Z L T-03-43B)
Figure BDA0001142660300000462
According to the method described in example 1, inReaction of intermediate 1-3a with a 3-methoxycinnamic acid substituted carboxy-terminal MDP derivative affords the title compound 8.1HNMR(400MHz,DMSO-d6)1.00(s,3H),1.02(s,3H),1.20-1.34(m,7H),1.42-1.52(m,6H),1.62-1.66(m,1H),1.66-1.76(m,2H),1.80(s,3H),1.92-2.02(m,1H),2.06-2.20(m,8H),2.26-2.36(m,4H),2.35-2.44(m,2H),2.84-2.98(m,2H),3.59(d,1H,J=6.7Hz),3.70-3.85(m,5H),3.94-4.16(m,5H),4.39-4.45(m,2H),4.67(s,1H),4.90(d,J=9.8Hz,1H),4.99(1H,brs),5.29(t,1H,J=8.7Hz),5.39(d,1H,J=7.0Hz),5.87(t,1H,J=9.1Hz),6.30(s,1H),6.79(d,J=15.9Hz,1H),6.95(d,J=8.5Hz,1H),7.08-7.16(m,2H),7.16-7.22(m,1H),7.32(t,J=7.8Hz,1H),7.35-7.44(m,6H),7.46-7.51(m,2H),7.52-7.61(m,1H),7.65(t,J=7.5Hz,2H),7.73(t,J=7.1Hz,1H),7.86(d,J=7.3Hz,3H),7.94(d,J=7.7Hz,3H),8.26(d,J=7.9Hz,1H),8.37(d,J=5.4Hz,1H),9.07(d,J=4.5Hz,1H).
Example 9
Synthesis of Compound 9 (accession number Z L T-03-44B)
Figure BDA0001142660300000471
Example 4
Synthesis of Compound 4 (accession number Z L T-03-19A)
The title compound 9 was prepared according to the procedure described for example 1 using intermediate 1-3a to react with a 2-chlorocinnamic acid substituted carboxy-terminal MDP derivative.1HNMR(400MHz,DMSO-d6)0.99(s,3H),1.02(s,3H),1.20-1.31(m,7H),1.42-1.53(m,6H),1.63-1.65(m,1H),1.67-1.78(m,2H),1.80(s,3H),1.92-2.03(m,1H),2.11(s,3H),2.15(s,5H),2.26-2.35(m,1H),2.35-2.41(m,2H),2.84-2.94(m,2H),3.59(d,1H,J=7.2Hz),3.72-3.85(m,2H),3.95-4.21(m,5H),4.35-4.45(m,2H),4.67(s,1H),4.90(d,J=9.6Hz,1H),4.99(1H,brs),5.29(t,1H,J=8.8Hz),5.39(d,1H,J=7.2Hz),5.87(t,1H,J=8.4Hz),6.30(s,1H),6.84(d,1H,J=15.7Hz),7.10(s,1H),7.17-7.24(m,1H),7.36-7.43(m,6H),7.46-7.57(m,5H),7.64-7.75(m,5H),7.84-7.89(m,3H),7.92-7.99(m,3H),8.29(d,J=8.1Hz,1H),8.51(d,J=6.2Hz,1H),9.06(d,J=7.1Hz,1H).
Example 10
Synthesis of Compound 10 (accession number Z L T-03-45A)
Figure BDA0001142660300000481
The title compound 10 was prepared according to the procedure described for example 1 using intermediate 1-3a reacted with 7-quinolinecarboxylic acid substituted carboxy-terminal MDP derivative.1HNMR(400MHz,DMSO-d6)0.99(s,3H),1.02(s,3H),1.20-1.31(m,7H),1.42-1.52(m,6H),1.62-1.66(m,1H),1.66-1.78(m,2H),1.80(s,3H),1.92-2.01(m,1H),2.08-2.21(m,8H),2.25-2.34(m,1H),2.35-2.42(m,2H),2.84-2.96(m,2H),3.59(d,1H,J=7.0Hz),3.72-3.84(m,2H),3.95-4.04(m,3H),4.06-4.13(m,1H),4.18-4.24(m,1H),4.41(d,J=9.6Hz,1H),4.59-4.74(m,2H),4.90(d,J=9.5Hz,1H),5.02(1H,brs),5.26-5.35(m,1H),5.39(d,1H,J=7.0Hz),5.76-5.94(m,1H),6.31(s,1H),7.10(s,1H),7.24–7.13(m,1H),7.40(d,J=4.0Hz,4H),7.47(dd,J=14.0,6.1Hz,3H),7.55(t,J=7.2Hz,1H),7.65(t,J=7.5Hz,2H),7.73(t,J=7.0Hz,2H),7.80(d,J=6.7Hz,1H),7.88(t,J=7.4Hz,4H),7.94(d,J=7.7Hz,2H),8.09(d,J=8.1Hz,1H),8.16(d,J=8.5Hz,2H),8.49(d,J=7.7Hz,1H),8.58(d,J=8.5Hz,1H),8.93(d,J=7.4Hz,1H),9.13(d,J=7.8Hz,1H).
Example 11
Synthesis of Compound 11 (accession number Z L T-03-45B)
Figure BDA0001142660300000482
The title compound 11 was prepared according to the procedure described for example 1 using intermediate 1-3a to react with a p-hydroxycinnamic acid substituted carboxy terminal MDP derivative.1HNMR(400MHz,DMSO-d6)1.00(s,3H),1.02(s,3H),1.21-1.31(m,7H),1.42-1.53(m,6H),1.62-1.67(m,1H),1.66-1.78(m,2H),1.81(s,3H),1.92-2.01(m,1H),2.09-2.22(m,8H),2.27-2.35(m,1H),2.35-2.42(m,2H),2.85-2.95(m,2H),3.60(d,1H,J=7.0Hz),3.70-3.85(m,2H),3.95-4.16(m,5H),4.43(d,J=9.6Hz,2H),4.68(s,1H),4.90(d,J=9.5Hz,1H),4.99(1H,brs),5.30(t,1H,J=7.1Hz),5.39(d,1H,J=7.1Hz),5.76-5.96(m,2H),6.31(s,1H),6.53(d,J=15.6Hz,1H),6.79(d,J=8.3Hz,2H),7.10(s,1H),7.16-7.26(m,1H),7.31(d,J=15.7Hz,1H),7.35-7.45(m,6H),7.49(t,J=7.4Hz,2H),7.53–7.58(m,1H),7.65(t,J=7.6Hz,2H),7.74(t,J=7.2Hz,1H),7.84-8.05(m,7H),8.26(d,J=6.2Hz,2H),9.08(d,J=8.1Hz,1H),9.91(brs,1H).
Example 12
Synthesis of Compound 12 (accession number Zpr0224-01)
Figure BDA0001142660300000491
Intermediate 1-2b (white solid powder) was prepared according to the method described in example 1, using docetaxel and benzyl propiolate to react; preparing intermediates 1-3b (white solid powder) by catalytic hydrogenation of the intermediates 1-2 b; the title compound 12 was prepared by reacting intermediates 1-3b with a p-fluorocinnamic acid substituted carboxy-terminal MDP derivative.1HNMR(400MHz,DMSO-d6)0.96(s,6H),1.16-1.30(m,7H),1.36(s,9H),1.42-1.80(m,12H),1.95-2.03(m,1H),2.12-2.18(m,5H),2.21-2.30(m,1H),2.35-2.42(m,2H),2.97-3.07(m,2H),3.63(d,J=6.9Hz,1H),3.65-3.76(m,2H),3.92–4.08(m,3H),4.08–4.21(m,3H),4.36–4.43(m,1H),4.46(s,1H),4.79(s,1H),4.88(d,J=9.9Hz,1H),4.98(s,1H),5.03(d,J=7.0Hz,1H),5.08(s,1H),5.37(d,J=7.0Hz,1H),5.80(t,J=9.8Hz,1H),6.70(d,J=15.8Hz,1H),7.12(s,1H),7.16(t,J=7.3Hz,1H),7.22-7.34(m,5H),7.34-7.44(m,3H),7.56(d,J=8.1Hz,1H),7.56-7.63(m,4H),7.73(t,J=7.3Hz,1H),7.86(s,1H),7.94(d,J=7.6Hz,2H),8.11(d,J=7.5Hz,1H),8.23(d,J=8.0Hz,1H),8.34(d,J=6.8Hz,1H),12.49(brs,1H).
Example 13
Synthesis of Compound 13 (accession number Zpr0223-02)
Figure BDA0001142660300000501
The title compound 13 was prepared according to the procedure described for example 12, using intermediate 1-3b to react with quinoline-2-carboxylic acid substituted carboxy-terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.96(s,6H),1.22-1.45(m,16H),1.42-1.82(m,12H),1.92-2.02(m,1H),2.12-2.19(m,5H),2.20-2.30(m,1H),2.35-2.43(m,2H),2.97-3.05(m,2H),3.63(d,J=6.7Hz,1H),3.75-3.85(m,2H),3.95-4.05(m,3H),4.06-4.16(m,2H),4.16-4.22(m,1H),4.46(s,1H),4.59-4.67(m,1H),4.79(t,J=8.3Hz,1H),4.88(d,J=9.7Hz,1H),4.97(s,1H),5.02(d,J=5.9Hz,1H),5.09(s,1H),5.37(d,J=6.8Hz,1H),5.80(t,J=8.8Hz,1H),7.11(s,1H),7.16(t,J=7.1Hz,1H),7.29(d,J=6.9Hz,2H),7.37(t,J=7.4Hz,2H),7.42(s,1H),7.55(d,J=7.8Hz,1H),7.64(t,J=7.5Hz,2H),7.73(t,J=7.5Hz,2H),7.82–7.91(m,2H),7.94(d,J=7.9Hz,2H),8.10(t,J=9.4Hz,2H),8.16(d,J=8.5Hz,2H),8.39(d,J=8.0Hz,1H),8.59(d,J=8.5Hz,1H),8.93(d,J=7.3Hz,1H),12.50(brs,1H).
Example 14
Synthesis of Compound 14 (accession number Zpr0223-03)
Figure BDA0001142660300000502
The title compound 14 was prepared according to the procedure described for example 12 using intermediates 1-3b to react with a 2-thiopheneacrylic acid substituted carboxy-terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.96(s,6H),1.22-1.47(m,16H),1.42-1.81(m,12H),1.92-2.01(m,1H),2.12-2.18(m,5H),2.20-2.32(m,1H),2.35-2.45(m,2H),2.97-3.06(m,2H),3.63(d,J=7.0Hz,1H),3.66-3.76(m,2H),3.95-4.05(m,3H),4.09–4.19(m,3H),4.36(p,J=7.0Hz,1H),4.45(s,1H),4.79(t,J=8.6Hz,1H),4.88(d,J=10.2Hz,1H),4.97(s,1H),5.02(d,J=6.3Hz,1H),5.09(s,1H),5.37(d,J=7.2Hz,1H),5.80(t,J=8.6Hz,1H),6.50(d,J=15.5Hz,1H),7.07-7.12(t,J=4.2Hz,2H),7.16(t,J=7.4Hz,1H),7.25–7.33(m,3H),7.34-7.40(m,3H),7.51–7.59(m,2H),7.61(d,J=5.1Hz,1H),7.64(t,J=7.6Hz,2H),7.73(t,J=7.3Hz,1H),7.86(s,1H),7.94(d,J=7.6Hz,2H),8.09(d,J=7.6Hz,1H),8.20(d,J=8.1Hz,1H),8.37(d,J=6.7Hz,1H),12.50(brs,1H).
Example 15
Synthesis of Compound 15 (accession number Zpr0241-01)
Figure BDA0001142660300000511
The title compound 15 was prepared according to the procedure described for example 12 using intermediate 1-3b reacted with a2, 4-difluorocinnamic acid substituted carboxy-terminal MDP derivative.1HNMR(500MHz,DMSO-d6)0.96(s,6H),1.16-1.31(m,7H),1.36(s,9H),1.42-1.82(m,12H),1.92-2.02(m,1H),2.11-2.18(m,5H),2.21-2.31(m,1H),2.35-2.43(m,2H),2.98-3.06(m,2H),3.63(d,J=7.0Hz,1H),3.66-3.76(m,2H),3.95-4.05(m,3H),4.09–4.21(m,3H),4.40(p,J=7.0Hz,1H),4.45(s,1H),4.79(t,J=8.2Hz,1H),4.88(d,J=10.2Hz,1H),4.96(s,1H),5.02(d,J=6.8Hz,1H),5.09(s,1H),5.37(d,J=7.2Hz,1H),5.80(t,J=8.7Hz,1H),6.81(d,J=16.0Hz,1H),7.11(s,1H),7.13–7.21(m,2H),7.25–7.33(m,3H),7.33-7.40(m,3H),7.44(d,J=16.0Hz,1H),7.54(d,J=8.3Hz,1H),7.64(t,J=7.6Hz,2H),7.68–7.76(m,2H),7.85(s,1H),7.94(d,J=7.4Hz,2H),8.09(d,J=7.6Hz,1H),8.23(d,J=8.1Hz,1H),8.47(d,J=6.8Hz,1H),12.50(brs,1H).
Example 16
Synthesis of Compound 16 (accession number Zpr0241-02)
Figure BDA0001142660300000521
The title compound 16 was prepared according to the procedure described for example 12 using intermediate 1-3b reacted with a3, 4-difluorocinnamic acid substituted carboxy-terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.96(s,6H),1.16-1.32(m,7H),1.36(s,9H),1.42-1.84(m,12H),1.92-2.01(m,1H),2.11-2.19(m,5H),2.21-2.33(m,1H),2.35-2.42(m,2H),2.95-3.05(m,2H),3.61(d,J=7.0Hz,1H),3.65-3.75(m,2H),3.92-4.02(m,3H),4.07–4.19(m,3H),4.34–4.42(m,1H),4.43(s,1H),4.77(t,J=9.0Hz,1H),4.86(d,J=9.8Hz,1H),4.94(s,1H),5.00(d,J=6.8Hz,1H),5.07(s,1H),5.35(d,J=7.0Hz,1H),5.78(t,J=8.6Hz,1H),6.72(d,J=15.8Hz,1H),7.08(s,1H),7.15(t,J=7.3Hz,1H),7.23–7.32(m,3H),7.32–7.56(m,6H),7.59-7.66(m,3H),7.71(t,J=7.4Hz,1H),7.83(s,1H),7.93(d,J=8.1Hz,2H),8.07(d,J=7.4Hz,1H),8.20(d,J=8.0Hz,1H),8.32(d,J=6.8Hz,1H),12.48(brs,1H).
Example 17
Synthesis of Compound 17 (accession number Zpr0261-01)
Figure BDA0001142660300000522
The title compound 17 was prepared according to the procedure described for example 12 using intermediate 1-3b reacted with a 3-fluorocinnamic acid substituted carboxy-terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.96(s,6H),1.16-1.34(m,7H),1.36(s,9H),1.42-1.82(m,12H),1.91-2.01(m,1H),2.11-2.18(m,5H),2.21-2.31(m,1H),2.35-2.41(m,2H),2.95-3.05(m,2H),3.63(d,J=7.2Hz,1H),3.67-3.77(d,J=6.2Hz,2H),3.95-4.05(m,3H),4.21–4.08(m,3H),4.37-4.47(m,2H),4.79(t,J=7.6Hz,1H),4.88(d,J=10.1Hz,1H),4.96(s,1H),5.02(d,J=6.9Hz,1H),5.09(s,1H),5.37(d,J=7.2Hz,1H),5.80(t,J=7.8Hz,1H),6.82(d,J=15.8Hz,1H),7.11(s,1H),7.16(t,J=7.3Hz,1H),7.24–7.34(m,3H),7.35–7.46(m,5H),7.49–7.60(m,2H),7.61–7.68(m,3H),7.73(t,J=7.3Hz,1H),7.85(s,1H),7.94(d,J=7.2Hz,2H),8.09(d,J=7.7Hz,1H),8.23(d,J=8.1Hz,1H),8.34(d,J=6.8Hz,1H),12.50(brs,1H).
Example 18
Synthesis of Compound 18 (accession number Zpr0261-02)
Figure BDA0001142660300000531
The title compound 18 was prepared according to the procedure described for example 12 using intermediate 1-3b reacted with a 4-methylcinnamic acid substituted carboxy-terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.96(s,6H),1.16-1.36(m,7H),1.36(s,9H),1.42-1.84(m,12H),1.92-2.02(m,1H),2.11-2.17(m,5H),2.21-2.32(m,4H),2.35-2.42(m,2H),2.96-3.06(m,2H),3.63(d,J=7.1Hz,1H),3.66–3.81(m,2H),3.95-4.07(m,3H),4.08–4.20(m,3H),4.39(p,J=7.1Hz,1H),4.45(s,1H),4.79(t,J=8.8Hz,1H),4.88(d,J=10.5Hz,1H),4.96(s,1H),5.02(d,J=6.6Hz,1H),5.09(s,1H),5.37(d,J=7.1Hz,1H),5.80(t,J=8.4Hz,1H),6.69(d,J=15.8Hz,1H),7.10(s,1H),7.16(t,J=7.3Hz,1H),7.22(d,J=8.0Hz,2H),7.25–7.34(m,3H),7.34–7.42(m,3H),7.45(d,J=8.0Hz,2H),7.55(d,J=8.4Hz,1H),7.64(t,J=7.5Hz,2H),7.73(t,J=7.3Hz,1H),7.85(s,1H),7.94(d,J=7.2Hz,2H),8.09(d,J=7.7Hz,1H),8.21(d,J=8.1Hz,1H),8.31(d,J=6.7Hz,1H),12.50(s,1H).
Example 19
Synthesis of Compound 19 (accession number Zpr0261-03)
Figure BDA0001142660300000541
The title compound 19 was prepared according to the procedure described for example 12 using intermediate 1-3b reacted with a 2-fluoro-4-chlorocinnamic acid substituted carboxy-terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.96(s,6H),1.16-1.35(m,7H),1.36(s,9H),1.42-1.82(m,12H),1.91-2.01(m,1H),2.11-2.18(m,5H),2.21-2.31(m,1H),2.35-2.43(m,2H),2.96-3.06(m,2H),3.63(d,J=7.0Hz,1H),3.67-3.75(m,2H),3.95-4.05(m,3H),4.08–4.19(m,3H),4.37-4.47(m,2H),4.79(t,J=8.4Hz,1H),4.88(d,J=10.2Hz,1H),4.96(s,1H),5.02(d,J=7.2Hz,1H),5.09(s,1H),5.37(d,J=7.1Hz,1H),5.80(t,J=8.9Hz,1H),6.86(d,J=16.0Hz,1H),7.11(s,1H),7.16(t,J=7.3Hz,1H),7.26–7.36(m,3H),7.37(t,J=7.6Hz,3H),7.44(d,J=16.0Hz,1H),7.50-7.56(m,2H),7.61–7.80(m,4H),7.85(s,1H),7.94(d,J=7.2Hz,2H),8.09(d,J=7.6Hz,1H),8.23(d,J=8.1Hz,1H),8.50(d,J=6.7Hz,1H),12.49(brs,1H).
Example 20
Synthesis of Compound 20 (accession number Zpr0261-04)
Figure BDA0001142660300000542
Example 4
Synthesis of Compound 4 (accession number Z L T-03-19A)
The title compound 20 was prepared according to the procedure described for example 12 using intermediates 1-3b to react with p-hydroxycinnamic acid substituted carboxy terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.96(s,6H),1.16-1.36(m,7H),1.36(s,9H),1.42-1.82(m,12H),1.92-2.01(m,1H),2.11-2.19(m,5H),2.21-2.32(m,1H),2.35-2.42(m,2H),2.98-3.06(m,2H),3.63(d,J=7.0Hz,1H),3.80–3.66(m,2H),3.94-4.06(m,3H),4.19–4.08(m,3H),4.32-4.42(m,1H),4.45(s,1H),4.79(t,J=8.6Hz,1H),4.88(d,J=9.9Hz,1H),4.96(s,1H),5.02(d,J=6.0Hz,1H),5.09(s,1H),5.37(d,J=7.1Hz,1H),5.80(t,J=8.1Hz,1H),6.52(d,J=15.8Hz,1H),6.79(d,J=8.6Hz,2H),7.10(s,1H),7.16(t,J=7.3Hz,1H),7.26-7.34 7(m,3H),7.38(t,J=8.2Hz,4H),7.55(d,J=8.6Hz,1H),7.64(t,J=7.5Hz,2H),7.73(t,J=7.3Hz,1H),7.85(s,1H),7.94(d,J=7.2Hz,2H),8.08(d,J=7.7Hz,1H),8.20(dd,J=11.3,7.5Hz,2H),9.85(s,1H),12.49(s,1H).
Example 21
Synthesis of Compound 21 (accession number Zpr0261-05)
Figure BDA0001142660300000551
The title compound 21 was prepared according to the procedure described for example 12 using intermediate 1-3b reacted with a 2-naphthoxyacetic acid substituted carboxy-terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.96(s,6H),1.16-1.34(m,7H),1.36(s,9H),1.42-1.82(m,12H),1.91-2.02(m,1H),2.11-2.18(m,5H),2.21-2.31(m,1H),2.35-2.41(m,2H),2.95-3.05(m,2H),3.63(d,J=7.0Hz,1H),3.67-3.74(m,2H),3.95-4.05(m,3H),4.10-4.22(m,3H),4.45(t,J=7.0Hz,2H),4.74(s,2H),4.79(t,J=9.0Hz,1H),4.88(d,J=10.6Hz,1H),4.96(s,1H),5.02(d,J=7.2Hz,1H),5.09(s,1H),5.37(d,J=7.2Hz,1H),5.80(t,J=9.3Hz,1H),6.92(d,J=7.5Hz,1H),7.10(s,1H),7.16(t,J=7.3Hz,1H),7.25-7.45(m,6H),7.50-7.56(m,4H),7.64(t,J=7.5Hz,2H),7.73(t,J=7.4Hz,1H),7.78–7.92(m,2H),7.95(d,J=7.2Hz,2H),8.11(d,J=7.6Hz,1H),8.19–8.33(m,3H),12.50(s,1H).
Example 22
Synthesis of Compound 22 (accession number Zpr0276-01)
Figure BDA0001142660300000561
The title compound 22 was prepared according to the procedure described for example 12 using intermediates 1-3b to react with a benzoic acid substituted carboxy terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.95(s,6H),1.16-1.33(m,7H),1.34(s,9H),1.43-1.76(m,12H),1.91-2.01(m,1H),2.08-2.16(m,5H),2.20-2.27(m,1H),2.34-2.41(m,2H),2.95-3.05(m,2H),3.61(d,J=7.0Hz,1H),3.64-3.75(m,2H),3.94-4.04(m,3H),4.06-4.18(m,3H),4.40-4.50(m,2H),4.77(t,J=8.7Hz,1H),4.86(d,J=10.7Hz,1H),4.91-5.05(m,2H),5.07(s,1H),5.35(d,J=7.1Hz,1H),5.78(t,J=8.4Hz,1H),7.09-7.19(m,2H),7.24-7.32(m,3H),7.36(t,J=7.6Hz,2H),7.45(t,J=7.4Hz,2H),7.48-7.57(m,2H),7.63(t,J=7.5Hz,2H),7.71(t,J=7.3Hz,1H),7.81-7.88(m,3H),7.93(d,J=7.2Hz,2H),8.08(d,J=7.7Hz,1H),8.17(d,J=8.1Hz,1H),8.58(d,J=6.6Hz,1H),12.48(s,1H).
Example 23
Synthesis of Compound 23 (accession number Zpr0276-02)
Figure BDA0001142660300000562
The title compound 23 was prepared according to the procedure described for example 12 using intermediate 1-3b reacted with a 3-chlorobenzoic acid substituted carboxy terminal MDP derivative.1H NMR(500MHz,DMSO-d6)0.95(s,6H),1.22-1.30(m,3H),1.31-1.41(m,13H),1.42-1.77(m,12H),1.90-2.01(m,1H),2.10-2.19(m,5H),2.20-2.29(m,1H),2.33-2.41(m,2H),2.97-3.07(m,2H),3.61(d,J=7.0Hz,1H),3.66-3.76(m,2H),3.95-4.05(m,3H),4.08-4.16(m,3H),4.42-4.47(m,2H),4.77(t,J=8.4Hz,1H),4.86(d,J=10.3Hz,1H),4.91-5.05(m,2H),5.07(s,1H),5.35(d,J=7.2Hz,1H),5.78(t,J=8.4Hz,1H),7.12-7.18(m,2H),7.27(d,J=5.9Hz,3H),7.36(t,J=7.6Hz,2H),7.49(t,J=7.9Hz,1H),7.53(d,J=8.9Hz,1H),7.62-7.67(m,3H),7.71(t,J=7.4Hz,1H),7.82(d,J=7.9Hz,2H),7.93(dd,J=3.8,2.0Hz,3H),8.08(d,J=7.7Hz,1H),8.19(d,J=8.0Hz,1H),8.74(d,J=6.6Hz,1H),12.48(s,1H).
Example 24
Synthesis of Compound 24 (No. DY-16-16)
Figure BDA0001142660300000571
Prepared according to the method of example 12 by reacting the intermediate 1-3b with an amide-terminated MDP derivative substituted with 2-chloro-4-fluorocinnamic acidTo the title compound 24.1HNMR(400MHz,DMSO-d6)0.97(s,6H),1.16-1.31(m,7H),1.37(s,9H),1.42-1.82(m,12H),1.94-2.04(m,1H),2.10-2.18(m,5H),2.21-2.32(m,1H),2.35-2.44(m,2H),2.94-3.06(m,2H),3.63(d,J=5.9Hz,1H),3.67-3.75(m,2H),3.94–4.06(m,3H),4.08–4.18(m,3H),4.36–4.48(m,2H),4.75-4.83(m,1H),4.88(d,J=8.9Hz,1H),4.95(s,1H),5.01(d,J=7.4Hz,1H),5.09(s,1H),5.37(d,J=6.4Hz,1H),5.80(t,J=8.3Hz,1H),6.78(d,J=15.9Hz,1H),6.95(s,1H),7.10(s,1H),7.17(t,J=7.5Hz,1H),7.24–7.44(m,7H),7.54(d,J=8.5Hz,2H),7.61–7.70(m,3H),7.70–7.80(m,2H),7.81–7.90(m,2H),7.94(d,J=7.9Hz,2H),8.23(d,J=7.8Hz,1H),8.44(d,J=6.2Hz,1H).
Example 25
Synthesis of Compound No. 25 (DY-16-43)
Figure BDA0001142660300000572
The title compound 25 was prepared according to the procedure described for example 1 using intermediate 1-3a to react with a p-chlorobenzoic acid substituted amide terminated MDP derivative.1HNMR(400MHz,DMSO-d6)1.00(s,3H,16-H),1.03(s,3H,17-H),1.12-1.24(m,2H,27-H),1.23-1.33(m,5H,43-H and 28-H),1.40-1.46(m,1H,14-Ha),1.50(s,3H,19-H),1.57-1.61(m,2H,29-H),1.62-1.68(m,1H,6-Ha),1.70-1.76(m,2H,14-Hband 36a-H),1.80(s,3H,18-H),1.95-2.05(m,1H,36-Hb),2.11(s,3H,75-H),2.16(s,5H,21-Hand 35-H),2.26-2.34(m,1H,6-Hb),2.35-2.41(m,2H,23-H),2.80-3.00(m,2H,26-H),3.60(d,1H,J=6.4Hz,3-H),3.70-3.85(m,2H,22-H),3.97(d,1H,J=7.8Hz,20-Ha),4.03(d,1H,J=8.4Hz,20-Hb),4.07-4.17(m,3H,7-H,30-H and 37-H),4.35-4.45(m,2H,2-Hand 42-H),4.65(brs,1H,1-OH),4.87-4.94(m,2H,5-H and 7-OH),5.31(t,1H,J=9.1Hz,3’-H),5.40(d,1H,J=6.9Hz,2’-H),5.88(t,1H,J=8.8Hz,13-H),6.31(s,1H,10-H),6.76(d,1H,J=15.9Hz,46-H),6.94(s,1H,32-Ha),7.08(s,1H,39-Ha),7.16-7.24(m,1H,72-H),7.28(s,2H,32-Hb and 39-Hb),7.35-7.45(m,5H,47-H,71-H,73-H,50-H and 52-H),7.45-7.52(m,4H,62-H,63-H,65-H and 66-H),7.53-7.61(m,3H,64-H,49-H and 53-H),7.62-7.67(m,2H,57-H and 59-H),7.70-7.76(m,1H,58-H),7.78-7.90(m,4H,25-H,33-H,70-Hand 74-H),7.95(d,2H,J=6.8Hz,56-H and 60-H),8.19(d,1H,J=7.4Hz,40-H),8.35(d,1H,J=5.5Hz,44-H).8.97(d,1H,J=8.0Hz,67-H).
Example 26
Synthesis of Compound 26 (accession number WCT-218-1)
Figure BDA0001142660300000581
The title compound 26 was prepared according to the procedure described in example 1 by reacting paclitaxel with benzyl alkynylpropionate to prepare intermediates 1-2c, catalytically hydrogenating intermediates 1-2c to prepare intermediates 1-3c, and reacting intermediates 1-3c with p-chlorocinnamic acid substituted amide-terminated MDP derivatives.1H NMR(400MHz,DMSO)9.02(t,J=11.8Hz,1H),8.39(d,J=6.6Hz,1H),8.24(d,J=8.0Hz,1H),7.97(d,J=7.5Hz,2H),7.85(dt,J=14.9,7.2Hz,4H),7.77–7.70(m,1H),7.65(t,J=7.4Hz,2H),7.57(dd,J=14.9,7.7Hz,3H),7.49(t,J=9.7Hz,4H),7.45–7.37(m,5H),7.32(s,2H),7.22(d,J=4.0Hz,1H),7.12(s,1H),6.97(s,1H),6.77(d,J=15.8Hz,1H),6.32(s,1H),5.90(t,J=8.3Hz,1H),5.41(t,J=8.2Hz,1H),5.35(t,J=8.9Hz,1H),4.92(d,J=9.6Hz,2H),4.70(s,1H),4.42(dd,J=11.9,5.1Hz,2H),4.21–4.08(m,3H),4.02(dd,J=18.9,8.0Hz,2H),3.80(d,J=7.1Hz,2H),3.63(d,J=6.5Hz,1H),2.94(t,J=21.1Hz,2H),2.74(s,2H),2.61(t,J=6.4Hz,2H),2.39–1.95(m,12H),1.84–1.42(m,12H),1.32–1.16(m,7H),1.04(s,3H),1.02(s,3H).
Example 27
Synthesis of Compound 27 (accession number WCT-491-1)
Figure BDA0001142660300000591
The synthetic route of compound 27 (accession number WCT-491-1) is as follows:
Figure BDA0001142660300000592
10-DAB reacts with TES-Cl in pyridine, and TES protecting group is selectively introduced to OH at position 7. Protection of the OH group at the 10 position is achieved by reaction with 8 equivalents of acetyl chloride in pyridine. The title compound 27 was prepared by removing the TES protecting group at the OH position 7 in formic acid, introducing benzyl acrylate (reacted with benzyl propiolate according to the procedure described in example 1), and catalytic hydrogenation, condensation with MDA derivatives.1H NMR(400MHz,DMSO-d6)8.52(d,J=4.6Hz,1H),8.25(d,J=7.5Hz,1H),8.02(d,J=6.9Hz,2H),7.88(d,J=7.3Hz,1H),7.74–7.51(m,6H),7.44(d,J=16.1Hz,1H),7.37(d,J=8.3Hz,1H),7.30(s,2H),7.09(d,J=18.7Hz,1H),6.95(s,1H),6.84(t,J=13.8Hz,1H),6.28(d,J=23.2Hz,1H),5.40(d,J=7.1Hz,1H),4.94(t,J=14.4Hz,1H),4.65(s,1H),4.55(s,1H),4.41(d,J=6.0Hz,1H),4.23–3.97(m,6H),3.91(s,1H),3.76(d,J=5.8Hz,1H),3.66(d,J=7.1Hz,1H),2.98(s,2H),2.66(s,1H),2.17(d,J=29.2Hz,11H),1.99(d,J=16.3Hz,4H),1.65(d,J=32.3Hz,3H),1.49(d,J=25.2Hz,4H),1.28(dd,J=23.1,16.3Hz,8H),0.99(s,3H),0.97(s,3H).
Example 28
Synthesis of Compound 28 (accession number WCT-482-3)
Figure BDA0001142660300000601
The synthetic route for compound 28 (accession number WCT-482-3) is as follows:
Figure BDA0001142660300000602
TES protected at position 7, acetoxy protected at position 10, 10-DAB, according to the procedure described in example 1, was reacted with benzyl propiolate to give the corresponding benzyl acrylate substituted product. The title compound 28 was prepared by removing the TES protecting group under formic acid conditions, catalytic hydrogenation and condensation.1H NMR(400MHz,DMSO)8.53(d,J=6.5Hz,1H),8.25(d,J=8.1Hz,1H),8.02(d,J=7.6Hz,2H),7.90(d,J=8.6Hz,2H),7.73–7.50(m,5H),7.44(d,J=16.0Hz,1H),7.37(d,J=8.4Hz,1H),7.32(s,2H),7.13(s,1H),6.98(s,1H),6.86(d,J=16.0Hz,1H),6.36(s,1H),5.44(d,J=6.9Hz,1H),4.97–4.83(m,2H),4.59(s,1H),4.47–4.36(m,2H),4.19–4.09(m,3H),4.02(s,2H),3.87–3.78(m,1H),3.76–3.66(m,2H),3.03(d,J=5.9Hz,2H),2.41–2.27(m,3H),2.23–2.06(m,9H),2.05–1.89(m,4H),1.75–1.57(m,3H),1.50(s,4H),1.43–1.33(m,1H),1.32–1.20(m,7H),1.02(s,3H),0.98(s,3H).
Example 29
Synthesis of Compound 29 (accession number WCT-474-1)
Figure BDA0001142660300000611
The synthetic route for compound 29 (accession number WCT-474-1) is as follows:
Figure BDA0001142660300000612
after the protection of TES at position 7, 10-DAB reacts with benzyl propiolate according to the method described in example 1 to obtain the corresponding benzyl acrylate substituted product. The title compound 29 was prepared by removing the TES protecting group under formic acid conditions, catalytic hydrogenation and condensation.1H NMR(400MHz,DMSO-d6)8.52(d,J=6.4Hz,1H),8.24(d,J=8.1Hz,1H),8.01(d,J=7.1Hz,2H),7.89(d,J=6.4Hz,2H),7.72–7.53(m,5H),7.43(d,J=15.7Hz,1H),7.37(d,J=8.3Hz,1H),7.31(s,2H),7.12(s,1H),6.97(s,1H),6.84(t,J=12.5Hz,1H),5.48(d,J=12.1Hz,1H),4.92(t,J=9.5Hz,1H),4.83(s,1H),4.64(s,1H),4.48–4.33(m,2H),4.26–3.96(m,5H),3.91–3.66(m,3H),3.12–2.96(m,2H),2.37(s,2H),2.33–2.05(m,7H),2.03–1.94(m,1H),1.86(s,3H),1.69(dd,J=31.6,10.1Hz,3H),1.49(d,J=19.5Hz,4H),1.38(s,1H),1.33–1.17(m,7H),0.99(s,3H),0.96(s,3H).
Example 30
Synthesis of Compound 30 (accession number WCT-454-2)
Figure BDA0001142660300000621
Following the procedure described in example 1, (2R, 3S) -3-benzamide-2-hydroxy-phenylpropionic acid methyl ester was reacted with benzyl propiolate to give the corresponding benzyl acrylate substitution product. TES protecting group is removed under formic acid condition, and catalysis is carried outAfter hydrogenation and condensation, the title compound 30 was prepared.1H NMR(400MHz,DMSO-d6)8.79(d,J=7.8Hz,1H),8.52(s,1H),8.24(d,J=7.4Hz,1H),7.96–7.73(m,4H),7.67(d,J=7.8Hz,1H),7.61–7.19(m,13H),7.12(s,1H),6.97(s,1H),6.86(d,J=15.8Hz,1H),5.32(s,1H),4.52–4.29(m,2H),4.13(d,J=6.4Hz,2H),3.65(s,2H),3.47(s,3H),2.91(s,2H),2.29(s,2H),2.16(s,2H),1.99(s,1H),1.71(s,1H),1.58(s,1H),1.44(s,1H),1.36–0.99(m,9H).
Example 31
Synthesis of Compound 31 (accession number WCT-460-1)
Figure BDA0001142660300000622
The reaction of n-butyl (2R, 3S) -3-benzamide-2-hydroxy-phenylpropionate with benzyl propiolate according to the procedure described in example 1 gives the corresponding benzyl acrylate substitution product. The title compound 31 was prepared by removing the TES protecting group under formic acid conditions, catalytic hydrogenation and condensation.1H NMR(400MHz,DMSO-d6)8.79(d,J=8.4Hz,1H),8.52(d,J=6.6Hz,1H),8.25(d,J=7.9Hz,1H),7.87(d,J=7.9Hz,1H),7.82(d,J=7.4Hz,2H),7.77(s,1H),7.67(t,J=8.4Hz,1H),7.57–7.22(m,13H),7.12(s,1H),6.97(s,1H),6.86(d,J=16.0Hz,1H),5.28(t,J=8.0Hz,1H),4.44–4.37(m,1H),4.35(d,J=7.7Hz,1H),4.19–4.04(m,2H),3.87(t,J=6.3Hz,2H),3.64(s,2H),3.01–2.77(m,2H),2.29(s,2H),2.14(d,J=7.1Hz,2H),1.99(d,J=7.1Hz,1H),1.71(d,J=8.5Hz,1H),1.56(s,1H),1.42(d,J=9.1Hz,1H),1.35–1.06(m,11H),0.76(t,J=7.3Hz,3H).
Example 32
Mouse knockout of NOD1 and NOD2 genes reverses tumor growth and metastasis
Test materials:
the mouse is a wild type mouse C57/B6, male, 5-6w, purchased from Beijing Wintolite laboratory animal technology Ltd, and the NOD2 gene knockout mouse is introduced from US Jax L ab, cat No.005763, purified and bred in the laboratory, the NOD1 gene knockout mouse is constructed and bred by the laboratory by the CRISPR/CAS9 technology (FIG. 1 shows the PCR gene identification result of the NOD1 gene knockout mouse, wherein the symbol male indicates the male mouse, the symbol female indicates the female mouse, and WT indicates the wild type mouse), the tumor strain used in the test is non-small cell lung Cancer L ewis L ung Cancer (LL C), and the tumor strain is bred by the laboratory.
The test method comprises the following steps:
selecting L ewis lung cancer preserved C57B L/6 male tumor-bearing mice with good tumor growth and whole body condition, dislocation of cervical vertebra, taking out tumor mass under aseptic condition, cutting into tumor mass with diameter of 2-3mm with a scalpel, grinding with a homogenizer, centrifuging at 4 deg.C and 1000rpm for 7min, discarding supernatant, diluting cell precipitate at 1:20 (RPMI1640) to obtain homogenate, mixing well to 0.1m L/inoculating to healthy C57B L/6 WT and NOD1-/-、NOD2-/-Mice axilla subcutaneously, tumors naturally grow.
The day of inoculation was designated as D0, and the body weight of the mice and the natural growth of the tumor were measured and recorded. On day 28 post inoculation (D28), mice were sacrificed by cervical dislocation, tumors and lungs were dissected and dissected off, tumor weights were weighed, lungs were fixed in Bouin's fixative for 24h, photographs were taken, lung surface nodules were counted, and differences between groups were compared at t test.
And (3) test results:
as shown in FIG. 2, NOD1 or NOD2 knockout mice have significantly reduced tumor weights and metastases after inoculation with L ewis lung cancer, as compared to wild-type mice (WT), and are statistically different.
Example 33
In this example, the inventors specifically examined the antagonistic action of the compound of the present invention on NOD1 and NOD2, taking compound 25 (No. DY16-43) as an example.
Materials and methods
(1) Cells and reagents
The test substance: DY-16-43 (batch No. 20140313) was prepared as a 50mM stock solution in DMSO, and diluted with the medium to the corresponding concentration immediately before use.
HEK-Blue hNOD1cell, HEK-Blue hNOD2cell, HEK-Blue hT L R4cell, and 293-hNOD2 were purchased from InvivoGen (hNOD1 refers to humanized NOD1, hNOD2 refers to humanized NOD2)
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), p-p65(NOVUS, NBP1-77808), IKK α (CST, 2682S), IKK β (CST, 2370S), p-IKK α/β (CST, 2694S), Gobbronal conditioner Antibody RabbRabbIgG-H & L-HRP (Abcam, 6721), Rabbbscalpclonal Antibody reagent Antibody to RabbH & L-6752 (HRP-6752H & 6752).
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 reverse Transcription Kits,
Figure BDA0001142660300000641
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-MACS Column 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 Albumin was purchased from MP, CD14MicroBeads and L S-MACS Column were purchased from Miltenyl, RNeasy Mini Kit was purchased from Sigma, NaF and BSA were purchased from AmrestTMThe western EC L substrate, PVDF membrane and Precision Plus Protein Dual color standards were purchased from Bio-Rad, 3MM filter paper from Millipore, RIPA lysate, SDS-PAGE Gel Kit, L applying buffer, transmembrane buffer, electrophoresis buffer and 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 Y)MPUS),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), centrifuge tubes (Corning), petri dishes (Corning), and the like.
(3) Detection of HEK-Blue hNOD1 antagonist (HEK-Blue Detection)
HEK-Blue hNOD1/2 cells were cultured in DMEM medium (DMEM complete medium) containing 10% FBS, 50U/M L penicilin, 50 μ g/M L streptomycin, 50 μ g/M L Normocin, 2mM L-glutamine, cells in the logarithmic growth phase were collected, HEK-Blue Detection assay adjusted for cell concentration and seeded in 96-well plates (50000 cells/well), test compound gradient working solutions (final concentrations 10 μ M,5 μ M, 2.5 μ M, 1.25 μ M, 625nM, 312.5nM, 31.25nM, 3.125nM) and vehicle control 37 ℃, 5% CO2After incubation in the incubator for 3h, the positive stimulus C12-ie-DAP (final concentration 50ng/m L), 37 ℃ C, 5% CO was added2The 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% TCA and fixed for 1h at 4 ℃, deionized water is used for washing the plate for 5 times and 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 naturally dried, 150 mu L/well is added with Tris base and 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, and the OD value of a tested group is marked as (T) and the Growth rate% (Percentage Growth) ([ 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 (100ng/m L), the incubation time is 18h, and the detection wavelength is 650 nm.
(5) Culture and treatment of human peripheral blood-derived macrophages (PBMCs-derived macrophages)
Venous blood of 200m L of healthy volunteers is taken, Peripheral Blood Mononuclear Cells (PBMCs) are separated by a Ficoll density gradient centrifugation method, and CD14 is sorted by an immunomagnetic bead method+Monocyte, M-CSF induced in vitro to differentiate into macrophages, 1 × 106The density per well was inoculated in 12-well plates, DY-16-43 (final concentrations 10. mu.M and 1. mu.M, respectively) or positive control (posivetontrol, 1. mu.M) was added, after 1h of incubation, C12-ie-DAP (final concentration 500ng/M L) or MDP (final concentration 5. mu.g/M L) was added, after 90min the supernatant was aspirated, and the cell samples were lysed with Trizol.
(6) Reverse transcription and fluorescent quantitative PCR
After lysing cell samples with Trizol, RNA content was detected with RNAeasy mini kit (Qianen) for extraction and purification of total RNA. ultra-micro spectrophotometer Nanodrop photometry (Thermo), where the ratio of absorbance at wavelength 260nm to absorbance at wavelength 280nm (A260:280OD ratios) of 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, the final volume was 20. mu. L, and the samples were placed in 96-well or 384-well plates and subjected to a quantitative PCR reaction, I L-6, TNF-alpha and GAPDH primers and probes were purchased from Invitron (Hs 005631 _ m1, Hs 01124 _ g1, Hs 02799581. g1, the reaction system was 10. mu.7 Man external Master (Taq 2), Taq 351. mu. 25, Asd3526. mu. cDNA (Taq 3526), AsdH 3526. mu. 20. mu. multidot. cDNA2O。
The reaction parameters are ① 50 ℃ and 2min
② 95 ℃ for 10min
③ 95 degree centigrade, 15sec
④ 60 deg.C, 1min ③ to ④ 40cycles
GAPDH was used as an internal reference gene, and
Figure BDA0001142660300000651
the 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 or 12-well plate, adding test compound DY-16-43 (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, cracking cell with RIPA lysate, extracting protein, performing SDS-PAGE electrophoresis, loading 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 4 deg.C, rinsing at 1 × TBST for 3 times, adding diluted secondary antibody at room temperature for 1h, rinsing with 1 × ST, adding EC L luminescent substrate, and ChemiDocTMXRS+Exposure in the imager.
Test results
As shown in FIG. 3A, compound DY-16-43 has a significant antagonistic effect on hNOD1, L ogIC50Is-5.881; DY-16-43 has a certain inhibitory effect on hNOD2, and the inhibition rate is about 40% at a concentration of 10 mu M. No significant cytotoxicity was observed with DY-16-43 at the above concentrations tested, and the results are shown in FIG. 3B.
As can be seen from FIG. 4, DY-16-43 can significantly antagonize the agonistic action of C12-ie-DAP on human peripheral blood-derived macrophage NOD1, reduce the transcription of pro-inflammatory cytokines I L6 and TNF- α, and the inhibition rate on I L-6 can reach 70%. Positive compound (positive control) is a known NOD1 selective antagonist (structural formula I). As can be seen from FIG. 5, DY-16-43 can significantly antagonize the agonistic action of MDP on human peripheral blood-derived macrophage NOD2, reduce the transcription of pro-inflammatory cytokine I L6. Positive compound (positive control) is a known NOD2 selective antagonist (structural formula ii).
Figure BDA0001142660300000661
As can be seen from FIG. 6A, after stimulation of human peripheral blood-derived macrophages by C12-ie-DAP, the expression of phosphorylated Ikk α/β and phosphorylated p65 is increased, and the expression of IkappaB α is decreased, indicating that the NF-kappaB signal pathway is activated, the expression of phosphorylated JNK, p38 and ERK is increased, indicating that the MAPK signal pathway is activated, while after treatment of the cells by DY-16-43, the expression of phosphorylated Ikk α/β and phosphorylated p65 is decreased, the expression of IkappaB α is increased, the expression of phosphorylated JNK, p38 and ERK is decreased, and DY-16-43 can block NOD 1-mediated activation of NF-kappaB and MAPK signal pathway, as can be seen in FIG. 6B, after stimulation of 293-hNOD2 cells by MDP, the expression of IkappaB α is decreased, indicating that the NF-kappaB signal pathway is activated, and after stimulation of the phosphorylated IkappaB, the expression of ERK 6343, the expression of ERK is increased, indicating that the phosphorylated IkappaB 3943 and the expression of DY-16-6, the phosphorylation of the cells is blocked.
In conclusion, DY-16-43 can remarkably antagonize the NF-kB and MAPK signal pathway activation mediated by NOD1 and NOD2, and is an NOD1/2 inhibitor.
Example 34
In this example, the inventors specifically examined the effect of the compound of the present invention on tumor growth and metastasis in mice using compound 25 (No. DY16-43) as an example.
Effect of DY16-43 on spontaneous metastasis of mouse L ewis lung cancer model (LL C)
The test substance:
paclitaxel (PTX) (batch: JF20080401), DY-16-43 (batch: 20150309-159) were prepared as DMSO stock solutions of 20 ×, and diluted to the administration concentration with physiological saline containing 5% polyoxyethylene castor oil (cremophor E L) 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 tumors, homogenizing under aseptic condition, preparing tumor cell suspension, taking healthy C57B L/6 male mice, inoculating 0.1m L/mouse axilla subcutaneous tumor liquid, setting the day of inoculation as D0, and after 1 day of inoculation (D1), dividing the mice into 4 groups according to body weight, wherein the groups are ① solvent control group (control), ② DY-16-4330 mg/kg group, ③ Paclitaxel (PTX)12mg/kg group, ④ DY-16-43(30mg/kg) + PTX (12mg/kg), 10 animals in each group, the PTX 12mg/kg intermittent administration group animals are intravenously injected for 1 two days every 4 days for 7 times, DY-16-43 and the control group animals are intravenously injected for 1 time every 2 days, 14 times, and the DY-16-43-6 + PTX 6 groups are intravenously injected for 1 mg/kg, and DY-16 mg/kg of solvent is injected for 1 time every 4 days.
Weighing animal body weight every 2-3 days during administration, and measuring the length and short diameter of tumor with vernier caliper according to formula (1/2) × long diameter × (short diameter)2Tumor size was calculated. The experiment was terminated 28 days after the inoculation (D28), and the mice were sacrificed by cervical dislocation after blood sampling from the eyeballs, and the tumors and lungs were dissected out and weighed. The lung was fixed in Bouin's fixative for 24h and the number of surface transferred nodules on the lung was counted. And (4) counting the detection index results, and performing comparative analysis on the administration group and the control group.
And (3) test results:
as shown in FIG. 7, DY-16-43+ PTX and PTX both significantly inhibited the growth of primary tumor and increased the tumor volume more slowly than the control group (FIG. 7A). After the administration, the tumor weights of the three groups of animals are remarkably reduced compared with that of a control group, and the tumor weights are statistically significant (FIG. 7B), wherein the tumor inhibition effect of DY-16-43+ PTX is remarkably stronger than that of a PTX single administration group. DY-16-43+ PTX and PTX can both significantly reduce the number of lung metastasis nodules in tumor-bearing mice, and have statistical significance compared with a vehicle control group (FIG. 7C). Also, DY-16-43+ PTX was significantly stronger than PTX alone (FIG. 7C). Fig. 7D is a photograph of lung metastases.
Example 35
In this example, the inventors specifically examined the antagonistic action of the compounds of the present invention on NOD1 and NOD 2. The experimental procedure is as described in example 33. The results of the experiment are shown in table 1:
table 1: antagonism of the compounds of the examples of the invention against NOD1 and NOD2 in vitro
Figure BDA0001142660300000681
Figure BDA0001142660300000691
ND means not tested
The results show that the compounds have different degrees of antagonism on the activation of NOD1 and NOD2 on the screened substrates under the measured concentration, and L ogIC of the NOD1 antagonism activity is measured by selecting DY-16-43 and DY-16-16 with the strongest antagonism activity50L ogIC for NOD2 with values of 5.7 and 5.9, respectively50The values were 5.3 and 5.1, respectively.
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 (12)

1. A compound which is a compound shown in a formula (I) or a pharmaceutically acceptable salt of the compound shown in the formula (I),
Figure FDA0002437083100000011
wherein,
R0is composed of
Figure FDA0002437083100000012
R1Is a phenyl group or a tert-butoxy group,
R2is a hydroxyl group or an acetoxy group,
R3is a hydroxyl group or an amino group,
ar is
Figure FDA0002437083100000013
R4、R5、R6、R7、R8Are independently H, F, Cl, Br, methyl, methoxy or hydroxyl respectively,
R10is a hydroxyl group, and the hydroxyl group,
x is O or-CH ═ CH-,
m and n are respectively and independently 0 or 1.
2. The compound of claim 1, having the structure of one of:
Figure FDA0002437083100000021
Figure FDA0002437083100000031
Figure FDA0002437083100000041
Figure FDA0002437083100000051
3. a pharmaceutical composition comprising a compound of claim 1 or 2.
4. The pharmaceutical composition of claim 3, further comprising a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle, or combination thereof.
5. The pharmaceutical composition of claim 4, further comprising paclitaxel and/or docetaxel.
6. Use of a compound according to claim 1 or 2 or a pharmaceutical composition according to any one of claims 3 to 5 in the manufacture of a medicament for the prevention or treatment of an immunoinflammatory disorder or tumour.
7. Use according to claim 6, wherein the medicament is for the prevention or treatment of polycythemia vera, rheumatoid arthritis, sjogren's syndrome, wegener's granulomatosis, behcet's disease, sarcoidosis, takayasu's arteritis, reactive arthritis, AIDS, allergic diseases, rheumatoid arthritis, chronic fatigue, type II diabetes, lupus erythematosus or multiple sclerosis, colon cancer, rectal cancer, stomach cancer, pancreatic cancer, bladder cancer, gallbladder cancer, breast cancer, kidney cancer, liver cancer, lung cancer, skin cancer, osteosarcoma, soft tissue sarcoma, head and neck cancer, tumors of the central nervous system, ovarian cancer, uterine cancer, prostate cancer, acute myeloid leukemia or acute lymphocytic leukemia or metastases thereof.
8. Use of a compound according to claim 1 or 2 or a pharmaceutical composition according to any one of claims 3 to 5 in the manufacture of a medicament for antagonizing inflammatory NF-. kappa.B and MAPKs signalling pathways mediated by human or murine immune cells NOD1 and/or NOD 2.
9. Use of an antagonist for the manufacture of a medicament for the prevention or treatment of an immunoinflammatory disorder or a tumor, said antagonist comprising a compound of claim 1 or 2, for antagonizing at least one of:
(1)NOD1;
(2)NOD2;
(3) the NF- κ B signaling pathway; and
(4) MAPKs signaling pathways.
10. The use of claim 9, wherein the antagonist further comprises at least one of a small interfering nucleotide, an antisense nucleotide, or a protein, polypeptide that antagonizes the NOD1 or the NOD2 that silences the gene encoding the NOD1 or the NOD 2.
11. The use according to claim 10, wherein the antagonist is for the prevention or treatment of an immunoinflammatory disorder or a tumor.
12. The use according to claim 11, wherein the antagonist is for the prevention or treatment of polycythemia vera, rheumatoid arthritis, sjogren's syndrome, wegener's granulomatosis, behcet's disease, sarcoidosis, takayasu's arteritis, reactive arthritis, aids, allergic diseases, rheumatoid arthritis, chronic fatigue, type II diabetes, lupus erythematosus or multiple sclerosis, colon cancer, rectal cancer, gastric cancer, pancreatic cancer, bladder cancer, gallbladder cancer, breast cancer, kidney cancer, liver cancer, lung cancer, skin cancer, osteosarcoma, soft tissue sarcoma, head and neck cancer, tumors of the central nervous system, ovarian cancer, uterine cancer, prostate cancer, acute myeloid leukemia or acute lymphocytic leukemia or metastases thereof.
CN201610960010.9A 2016-11-03 2016-11-03 Substituted cell acyl dipeptide compound and preparation method and application thereof Active CN106589055B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201610960010.9A CN106589055B (en) 2016-11-03 2016-11-03 Substituted cell acyl dipeptide compound and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201610960010.9A CN106589055B (en) 2016-11-03 2016-11-03 Substituted cell acyl dipeptide compound and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN106589055A CN106589055A (en) 2017-04-26
CN106589055B true CN106589055B (en) 2020-07-28

Family

ID=58589600

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201610960010.9A Active CN106589055B (en) 2016-11-03 2016-11-03 Substituted cell acyl dipeptide compound and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN106589055B (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11957758B2 (en) 2017-09-07 2024-04-16 Shenzhen Salubris Pharmaceuticals Co. Ltd. Pharmaceutical composition of docetaxel conjugate and preparation method
CN111757888A (en) * 2017-09-07 2020-10-09 深圳信立泰药业股份有限公司 Application of docetaxel conjugate in preparation of medicines for preventing or treating various immune diseases
CN108392634A (en) * 2018-03-28 2018-08-14 清华大学 Purposes of the B7S1 inhibitor in preparing liver-cancer medicine
CN109453384A (en) * 2018-11-19 2019-03-12 山东大学 Application of the NOD1 in the product that preparation inhibits tumour SRC signal path
CN109512833B (en) * 2018-12-04 2020-10-30 天津医科大学总医院 Function and use of E2F6 inhibitors
WO2020223770A1 (en) * 2019-05-08 2020-11-12 Garvan Institute Of Medical Research Cancer stratification and treatment based on inhibition of nod-2
ES2827149A1 (en) * 2019-11-19 2021-05-19 Consejo Superior Investigacion PEPTIDES CONTAINING D-ALANINE (D-ALA) OR RELATED AMINOALCOHOLS (Machine-translation by Google Translate, not legally binding)
CN113125587B (en) * 2019-12-30 2022-05-24 成都百裕制药股份有限公司 Tofacitinib intermediate and detection method of enantiomer thereof
TW202340177A (en) 2021-12-30 2023-10-16 美商拜歐米富士恩股份有限公司 Pyrazine compounds as inhibitors of flt3

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0305559D0 (en) * 2003-03-11 2003-04-16 Teijin Ltd Compounds
CN1712399B (en) * 2004-06-24 2010-08-11 中国医学科学院药物研究所 Production and use for taxol and muramic acyl dipeptide conjugate substance of immune reinforcer
CN100418532C (en) * 2005-06-17 2008-09-17 吕志民 NF-kB compound inhibitor for treating various diseases
US9415061B2 (en) * 2008-04-01 2016-08-16 Innate Immunotherapeutics Limited Compositions and methods for treatment of neoplastic disease
JP5922104B2 (en) * 2010-05-27 2016-05-24 シェンヅェン サルブリス ファーマシューティカルズ カンパニー リミテッドShenzhen Salubris Pharmaceuticals Co., Ltd Chemical synthesis and anti-tumor and anti-tumor metastatic effects of dual functional conjugates

Also Published As

Publication number Publication date
CN106589055A (en) 2017-04-26

Similar Documents

Publication Publication Date Title
CN106589055B (en) Substituted cell acyl dipeptide compound and preparation method and application thereof
AU2019202922B2 (en) Compositions and methods of using the same for treatment of neurodegenerative and mitochondrial disease
TWI542594B (en) Proteasome inhibitors
EP3235819B1 (en) Pyrrolopyrimidine compounds useful as jak inhibitors
EP3560926B1 (en) 6-amino-7,9-dihydro-8h-purin-8-one compounds as brk inhibitors
CN101977915A (en) Macrocyclic serine protease inhibitors
WO2017191130A2 (en) Arginase inhibitors and their therapeutic applications
EA020488B1 (en) Derivatives of purine or deazapurine useful for the treatment of viral infections and other diseases
CN111542526B (en) Condensed imidazole derivatives substituted with tertiary hydroxyl groups as PI 3K-gamma inhibitors
CA3224105A1 (en) Nitrogen-containing heterocyclic compound, and preparation method therefor, intermediate thereof, and application thereof
CN106831614B (en) Substituted benzodiazacyclo compound and its preparation method and use
CN116348455A (en) Protein tyrosine phosphatase inhibitors and methods of use thereof
CN111971280A (en) Heteroaryl compounds, pharmaceutical compositions thereof and therapeutic uses thereof
US20230357182A1 (en) Solid forms of (s)-1-((2',6-bis(difluoromethyl)-[2,4'-bipyridin]-5-yl)oxy)-2,4-dimethylpentan-2-amine and salts thereof
EP4083032A1 (en) Pd-l1 antagonist compound
EP3774843B1 (en) Dipeptide piperidine derivatives
CN105960399B (en) Enzyme inhibitor epoxy ketone compound
CN112851648B (en) Application of brefeldin A ester derivatives in antitumor drugs
CN112851647B (en) Brefeldin A derivative and preparation method and application thereof
CN116178350A (en) Natural macrolide derivative, preparation method thereof and application of natural macrolide derivative as antitumor drug
TWI810547B (en) Pd-l1 antagonist compound
CN116970028A (en) Macrocyclic peptoid protease inhibitors and uses thereof
WO2022125613A1 (en) Phosphonates as inhibitors of enpp1 and cdnp
KR20240155293A (en) Solid form of (S)-1-((2',6-bis(difluoromethyl)-[2,4'-bipyridin]-5-yl)oxy)-2,4-dimethylpentan-2-amine and salts thereof
TW201700482A (en) Proteasome inhibitors

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