CN112920240A - Galactose-containing nitrogen-containing aromatic ring derivative and use thereof - Google Patents

Galactose-containing nitrogen-containing aromatic ring derivative and use thereof Download PDF

Info

Publication number
CN112920240A
CN112920240A CN202110100504.0A CN202110100504A CN112920240A CN 112920240 A CN112920240 A CN 112920240A CN 202110100504 A CN202110100504 A CN 202110100504A CN 112920240 A CN112920240 A CN 112920240A
Authority
CN
China
Prior art keywords
radical
cancer
atoms
alkylene
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202110100504.0A
Other languages
Chinese (zh)
Other versions
CN112920240B (en
Inventor
金传飞
钟文和
张英俊
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.)
Guangdong HEC Pharmaceutical
Original Assignee
Sunshine Lake Pharma Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sunshine Lake Pharma Co Ltd filed Critical Sunshine Lake Pharma Co Ltd
Priority to CN202110100504.0A priority Critical patent/CN112920240B/en
Publication of CN112920240A publication Critical patent/CN112920240A/en
Application granted granted Critical
Publication of CN112920240B publication Critical patent/CN112920240B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/20Carbocyclic rings
    • C07H15/203Monocyclic carbocyclic rings other than cyclohexane rings; Bicyclic carbocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H15/00Compounds containing hydrocarbon or substituted hydrocarbon radicals directly attached to hetero atoms of saccharide radicals
    • C07H15/26Acyclic or carbocyclic radicals, substituted by hetero rings

Abstract

The invention belongs to the technical field of medicines, and relates to a galactose-containing nitrogen-containing aromatic ring derivative, application thereof and a pharmaceutical composition containing the compound. The invention also relates to a method for preparing the compounds and the pharmaceutical compositions, and application of the compounds and the pharmaceutical compositions in preventing or treating tumors, inflammatory diseases, autoimmune diseases and other diseases, particularly tumors.

Description

Galactose-containing nitrogen-containing aromatic ring derivative and use thereof
Technical Field
The invention belongs to the technical field of medicines, and relates to a galactose-containing nitrogen-containing aromatic ring derivative, a pharmaceutical composition, a using method and application thereof. In particular, the invention relates to a nitrogenous aromatic ring derivative containing galactose and a pharmaceutical composition thereof, and also relates to a method for preparing the compound and the pharmaceutical composition, and application of the compound and the pharmaceutical composition in preventing or treating tumors, inflammatory diseases, autoimmune diseases and other diseases. In particular, the compound and the pharmaceutical composition thereof are used for preventing or treating tumors.
Background
Medically, cancer (cancer) refers to a malignant tumor that originates in epithelial tissue, and is the most common type of malignant tumor. The general term "cancer" is used to generally refer to all malignant tumors. Cancer has the biological characteristics of abnormal cell differentiation and proliferation, uncontrolled growth, infiltrative and metastatic properties, and the like, and the occurrence of the cancer is a multi-factor and multi-step complex process which is divided into three processes of carcinogenesis, cancer promotion and evolution. The etiology of malignant tumors is not completely understood, but is closely related to smoking, infection, environmental pollution, occupational exposure, genetic factors, and unreasonable diet. The currently well-defined cancer-related factors can be divided into two major categories, exogenous and endogenous: 1. exogenous factors- -lifestyle habits, environmental pollution and occupational, natural and biological factors, chronic irritation and trauma, iatrogenic factors; 2. endogenous factors-genetic factors, immune factors, endocrine factors.
Cancer is one of the major diseases that endanger human health, disrupt family and social harmony, is a major health problem worldwide and remains a major cause of death. Since 2010, the cancer is the second cause of human death, second only to cardiovascular and cerebrovascular diseases. Global cancer incidence is growing at an alarming rate as shown by globoc 2014, data published by the international cancer research institution under WHO at month 2 2014, with a total of about 1,400 million new cancer cases, of which about 820 million people die, worldwide in 2012. The incidence of cancer is getting more severe and the incidence and mortality rate are continuously rising, and according to the conjecture, with the growth and aging of population, the development of society and the establishment of bad living habits, new cases of cancer in the world can reach the level of 2,200 ten thousand per year in the next 20 years, and the number of deaths caused by cancer can rise dramatically to 1,300 ten thousand per year. About 60% of newly added cancer cases worldwide occur in developing countries, and annual mortality accounts for 70% of the world as well. WHO data shows that the number of newly diagnosed cancer cases in China in 2012 is 307 thousands, which accounts for about 21.8% of the world; data of '2013 annual report for registration of Chinese tumor' shows that in 2013, more than 309 ten thousands of new cancer cases are added in China, and the probability of getting cancer in life of a person is 22% according to the average life of the person being 74 years old, which means that the cancer is a common disease in China.
Cancer is a group of diseases characterized by abnormal cell differentiation and proliferation, which, in advanced stages, spread to other areas of the body, including bones and vital organs. Despite significant advances in the feasibility of multiple therapeutic regimens for treating cancer, currently available chemotherapies remain unsatisfactory and the prognosis for the majority of patients diagnosed with cancer remains unattractive. Therefore, the research and development of new anti-tumor drugs (or anti-cancer drugs) are of great significance.
Glycolipids are important components of cell membranes, participate in recognition activities of cell membranes, and have significant antitumor activity. The galactose-containing nitrogen-containing aromatic ring derivative has unexpected excellent antitumor activity, and the structure and the activity of the derivative are not reported so far.
Disclosure of Invention
The following is a summary of some aspects of the invention only and is not intended to be limiting. These aspects and others are described more fully below. All references in this specification are incorporated herein by reference in their entirety. When the disclosure of the present specification differs from the cited documents, the disclosure of the present specification controls.
The invention aims to overcome the defects in the prior art and provide a nitrogen-containing aromatic ring derivative containing galactose and a pharmaceutical composition with unexpected excellent antitumor activity, and a using method and application thereof.
The invention has the advantages that: the invention discovers and synthesizes the nitrogen-containing aromatic ring derivative containing galactose for the first time, has stronger inhibitory activity to tumor cells, and can be used for treating tumors. The compound has stable property, good safety and little toxic and side effect, and has the advantages of pharmacodynamics and pharmacokinetics, such as good brain/plasma ratio (brain plasma ratio), good bioavailability or good metabolic stability and the like, thereby having better clinical application prospect.
The invention also provides processes for the preparation of such compounds and pharmaceutical compositions containing them.
In one aspect, the invention relates to a compound that is a compound of formula (I) or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, pharmaceutically acceptable salt of a compound of formula (I) or a prodrug thereof,
Figure BDA0002915655280000021
wherein, R' and R4、R5、R6、R7、R8、R9Y, Z and V have the meanings as described in the invention.
In some embodiments, R' is
Figure BDA0002915655280000022
Wherein R is1、R2a、R2b、R3And X has the meaning as described in the present invention.
In some embodiments, V is-NH-, -O-, or-S-.
In some embodiments, X is CRxOr N; wherein R isxHave the meaning as described in the present invention.
In some embodiments, Y is CRyOr N; wherein R isyHave the meaning as described in the present invention.
In some embodiments, Z is CH or N.
In some embodiments, R1、R2a、R2b、R3And RxEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy), -S (═ O)2C1-C6Alkyl radical, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino or hydroxy substituted C1-C6An alkyl group.
In some embodiments, R4、R5、R6And RyEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy), -S (═ O)2C1-C6Alkyl radical, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino or hydroxy substituted C1-C6An alkyl group.
In some embodiments, R7And R8Each independently is H, D, -OH, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C3-6Cycloalkyl, heterocyclic group consisting of 3 to 8 atoms, C6-10Aryl, heteroaryl of 5 to 10 atoms, C1-C6Alkoxy radical C1-6Alkylene radical, C3-6Cycloalkyl radical C1-6Alkylene, (heterocyclic group consisting of 3 to 8 atoms) C1-6Alkylene radical, C6-10Aryl radical C1-6Alkylene or (heteroaryl of 5 to 10 atoms) C1-6An alkylene group; orR is7、R8And the nitrogen atom to which they are attached, together form a heterocyclic group of 3 to 8 atoms.
In other embodiments, R9Is C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C3-6Cycloalkyl, heterocyclic group consisting of 3 to 8 atoms, C6-10Aryl, heteroaryl of 5 to 10 atoms, C1-C6Alkoxy radical C1-6Alkylene radical, C3-6Cycloalkyl radical C1-6Alkylene, (heterocyclic group consisting of 3 to 8 atoms) C1-6Alkylene radical, C6-10Aryl radical C1-6Alkylene or (heteroaryl of 5 to 10 atoms) C1-6An alkylene group.
In some embodiments, R1、R2a、R2b、R3And RxEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy), -S (═ O)2C1-C4Alkyl radical, C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C4Alkylamino or hydroxy substituted C1-C4An alkyl group.
In other embodiments, R1、R2a、R2b、R3And RxEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3、-S(=O)2CH3Methyl, ethyl, n-propylAlkyl, isopropyl, tert-butyl, vinyl, -CHF2、-CF3Methoxy, -OCF3Methylamino, dimethylamino or hydroxymethyl.
In some embodiments, R4、R5、R6And RyEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy), -S (═ O)2C1-C4Alkyl radical, C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C4Alkylamino or hydroxy substituted C1-C4An alkyl group.
In other embodiments, R4、R5、R6And RyEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3、-S(=O)2CH3Methyl, ethyl, n-propyl, isopropyl, tert-butyl, vinyl, -CHF2、-CF3Methoxy, -OCF3Methylamino, dimethylamino or hydroxymethyl.
In some embodiments, R7And R8Each independently is H, D, -OH, C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C3-6Cycloalkyl, heterocyclic group consisting of 3 to 6 atoms, C6-10Aryl, heteroaryl of 5-6 atoms、C1-C4Alkoxy radical C1-4Alkylene radical, C3-6Cycloalkyl radical C1-4Alkylene, (heterocyclic group consisting of 3 to 6 atoms) C1-4Alkylene radical, C6-10Aryl radical C1-4Alkylene or (heteroaryl of 5 to 6 atoms) C1-4An alkylene group; or R7、R8And the nitrogen atom to which they are attached, together form a heterocyclic group of 3 to 6 atoms.
In other embodiments, R7And R8Each independently H, D, -OH, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, 2-methylpropyl, 1-methylpropyl, ethenyl, propenyl, allyl, ethynyl, propynyl, propargyl, -CHF2、-CF3Methoxy, ethoxy, isopropoxy, -OCF3Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuryl, piperidinyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, thienyl, thiazolyl, oxazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, methoxymethylene, methoxyethylene, C3-6Cycloalkyl methylene, C3-6Cycloalkylethylene radical, C3-6Cycloalkylpropylene, (heterocyclyl) methylene of 3-6 atoms, (heterocyclyl) ethylene of 3-6 atoms, phenylmethylene, phenylethylene, phenylpropylene, pyridylmethylene, pyrimidylmethylene, pyrrolylmethylene, pyrazolyl methylene, triazolylmethylene or tetrazolylmethylene; or R7、R8And the nitrogen atom to which they are attached form an azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, or morpholinyl group.
In some embodiments, R9Is C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C3-6Cycloalkyl, heterocyclic group consisting of 3 to 6 atoms, C6-10Aryl, heteroaryl of 5 to 6 atoms, C1-C4Alkoxy radical C1-4Alkylene radical, C3-6Cycloalkyl radical C1-4Alkylene, (heterocyclic group consisting of 3 to 6 atoms) C1-4Alkylene radical, C6-10Aryl radical C1-4Alkylene or (heteroaryl of 5 to 6 atoms) C1-4An alkylene group.
In other embodiments, R9Is methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, 2-methylpropyl, 1-methylpropyl, ethenyl, propenyl, allyl, ethynyl, propynyl, propargyl, -CHF2、-CF3Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuryl, piperidinyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, thienyl, thiazolyl, oxazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, methoxymethylene, methoxyethylene, C3-6Cycloalkyl methylene, C3-6Cycloalkylethylene radical, C3-6Cycloalkylpropylene, (heterocyclyl) methylene of 3-6 atoms, (heterocyclyl) ethylene of 3-6 atoms, phenylmethylene, phenylethylene, phenylpropylene, pyridylmethylene, pyrimidylmethylene, pyrrolylmethylene, pyrazolyl methylene, triazolylmethylene or tetrazolylmethylene.
In some embodiments, the compound of the present invention, which is a compound of formula (II) or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt of a compound of formula (II), or a prodrug thereof,
Figure BDA0002915655280000041
wherein R is3、R4、R6、R7、R8And R9Have the meaning as described in the present invention.
In some embodiments, the compound of the present invention, which is a compound of formula (III) or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt of a compound of formula (III), or a prodrug thereof,
Figure BDA0002915655280000042
wherein R is3、R4、R6、R7、R8And R9Have the meaning as described in the present invention.
In some embodiments, the compound of the present invention is a compound having one of the following structures or a stereoisomer, geometric isomer, tautomer, nitrogen oxide, hydrate, solvate, metabolite, pharmaceutically acceptable salt of the compound having one of the following structures, or a prodrug thereof:
Figure BDA0002915655280000043
Figure BDA0002915655280000051
in another aspect, the present invention relates to a pharmaceutical composition comprising a compound disclosed herein.
In some embodiments, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable excipient, carrier, adjuvant, or any combination thereof.
In yet another aspect, the present invention relates to the use of a compound or pharmaceutical composition disclosed herein for the preparation of a medicament for the prevention or treatment of a tumor.
In some embodiments, the tumor of the invention is a solid tumor, a B cell lymphoma, and/or a hematologic and immune system tumor.
In some embodiments, the solid tumor according to the present invention is gastric cancer, intestinal cancer, liver cancer, kidney cancer, lung cancer, brain cancer, laryngeal cancer, nasopharyngeal cancer, esophageal cancer, gallbladder cancer, head and neck cancer, squamous carcinoma, cancer of the lymphatic system, thyroid cancer, bladder cancer, ovarian cancer, cervical cancer, endometrial cancer, testicular cancer, prostate cancer, genitourinary tract cancer, breast cancer, small cell lung cancer, lung adenocarcinoma, pancreatic cancer, colon cancer, rectal cancer, glioblastoma, and/or monocytic leukemia.
In other embodiments, the B cell lymphoma according to the present invention is diffuse large B cell lymphoma, small lymphocytic tumor, mantle cell lymphoma, multiple myeloma, and/or chronic lymphocytic leukemia.
In still other embodiments, the hematologic and immune system tumors of the present invention are acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myeloid leukemia, and/or chronic myeloid leukemia.
In a further aspect, the invention relates to the use of a compound or pharmaceutical composition disclosed herein for the preparation of a medicament for the prevention or treatment of an inflammatory disorder or an autoimmune disease.
In some embodiments, the inflammatory disorder or autoimmune disease described herein is asthma, psoriasis, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, a heteroimmune disease, idiopathic thrombocytopenic purpura, immune complex-mediated vasculitis, and/or autoimmune-mediated hemolytic anemia.
In yet another aspect, the invention relates to methods for the preparation, isolation and purification of compounds of formula (I), formula (II) or formula (III).
Biological test results show that the compound has a remarkable inhibition effect on cell proliferation and unexpectedly excellent antitumor activity, so that the compound provided by the invention can be used as a better antitumor drug.
Any embodiment of any aspect of the invention may be combined with other embodiments, as long as they do not contradict. Furthermore, in any embodiment of any aspect of the invention, any feature may be applicable to that feature in other embodiments, so long as they do not contradict.
Detailed description of the invention
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. One 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 following definitions, as used herein, should be applied unless otherwise indicated. For the purposes of the present invention, the chemical elements are in accordance with the CAS version of the periodic Table of the elements, and the handbook of chemistry and Physics, 75 th edition, 1994. In addition, general principles of Organic Chemistry can be referred to as described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, and "March's Advanced Organic Chemistry" by Michael B.Smith and Jerry March, John Wiley & Sons, New York:2007, the entire contents of which are incorporated herein by reference.
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 one or to more than one (i.e., to at least one) of the objects. 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 "stereoisomers" refers to compounds having the same chemical structure, but differing in the arrangement of atoms or groups in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric (cis/trans) isomers, atropisomers, and the like.
The term "tautomer" or "tautomeric form" refers to structural isomers having different energies that can interconvert by a low energy barrier (low energy barrier). If tautomerism is possible (e.g., in solution), then the chemical equilibrium of the tautomer can be reached. For example, proton tautomers (also known as proton transfer tautomers) include interconversions by proton migration, such as keto-enol isomerization and imine-enamine isomerization.
When tautomerism (e.g., keto-enol tautomerism) of the compounds of the present invention or prodrugs thereof exists, it is intended to claim both their individual forms (e.g., keto or enol forms), respectively, and mixtures thereof in any proportion. The same applies to their stereoisomers, e.g., enantiomers, diastereomers, conformers (rotamers), geometric (cis/trans) isomers, atropisomers, and the like.
If desired, the tautomers can be separated according to methods known in the art (e.g., liquid chromatography). The same applies to their enantiomers, for example, using chiral stationary phase separation. Furthermore, enantiomers can be separated by conversion to diastereomers, i.e. coupling with enantiomerically pure auxiliary compounds, followed by separation of the resulting diastereomers and cleavage of the auxiliary residues. Alternatively, any enantiomer of a compound of the invention may be obtained by stereoselective synthesis using optically pure starting materials.
"pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of patients without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio, and which are effective for their intended use.
The term "optionally substituted with … …" is used interchangeably with the term "unsubstituted or substituted with …", i.e., the structure is unsubstituted or substituted with one or more substituents described herein, including but not limited to D, F, Cl, Br, I, N3,-CN,-NO2,-NH2,-OH,-SH,-COOH,-C(=O)NH2,-C(=O)NHCH3,-C(=O)N(CH3)2-C (═ O) -alkyl, -C (═ O) -alkoxy, -NHS (═ O)2-alkyl, -N (alkyl) S (═ O)2-alkyl, -S (═ O)2Alkyl, alkoxy, alkylthio, alkylamino, alkenyl, alkynyl, haloalkyl, haloalkoxy, hydroxy-substituted alkyl, cyano-substituted alkyl, amino-substituted alkyl, cycloalkyl, heterocyclyl, aryl, heteroaryl, alkoxy-alkylene, cycloalkyl-alkylene, heterocyclyl-alkylene, aryl-alkylene, heteroaryl-alkylene, and the like.
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-C6Alkyl "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 terms "halogen" and "halo" are used interchangeably herein to refer to fluorine (F), chlorine (Cl), bromine (Br), or iodine (I).
The term "alkyl" or "alkyl group" as used herein, denotes a saturated, straight or branched chain, monovalent hydrocarbon group containing 1 to 20 carbon atoms, wherein the alkyl group may be optionally substituted with one or more substituents as described herein. In some embodiments, the alkyl group contains 1 to 6 carbon atoms; in other embodiments, the alkyl group contains 1 to 4 carbon atoms; in still other embodiments, the alkyl group contains 1 to 3 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(2-methylpropyl)), sec-butyl (s-Bu, -CH (CH)3)CH2CH3(1-methylpropyl)), tert-butyl (t-Bu, -C (CH)3)3) And so on.
The term "alkylene" refers to a saturated divalent hydrocarbon radical resulting from the removal of two hydrogen atoms from a saturated straight or branched chain hydrocarbon. Unless otherwise specified, the alkylene group contains 1 to 10 carbon atoms. In some embodiments, the alkylene group contains 1 to 6 carbon atoms; in other embodiments, the alkylene group contains 1 to 4 carbon atoms; in still other embodiments, the alkylene group contains 1 to 2 carbon atoms. Examples include, but are not limited to, methylene (-CH)2-) ethylene (-CH2CH2-) propylene (-CH)2CH2CH2-) isopropylidene (-CH (CH)3)CH2-) and the like. Said alkylene group is optionallySubstituted with one or more substituents as described herein.
The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. one carbon-carbon sp2A double bond, wherein the alkenyl group may be optionally substituted with one or more substituents described herein, including the positioning of "cis" and "trans", or the positioning of "E" and "Z". In one embodiment, the alkenyl group contains 2 to 8 carbon atoms; in another embodiment, the alkenyl group contains 2 to 6 carbon atoms; in yet another embodiment, the alkenyl group contains 2 to 4 carbon atoms. Examples of alkenyl groups include, but are not limited to, vinyl (-CH ═ CH)2) Allyl (-CH)2CH=CH2) 1-propenyl (i.e., propenyl, -CH ═ CH-CH)3) And so on.
The term "alkynyl" denotes a straight or branched chain monovalent hydrocarbon radical containing 2 to 12 carbon atoms, wherein there is at least one site of unsaturation, i.e. a carbon-carbon sp triple bond, wherein said alkynyl radical may optionally be substituted with one or more substituents as described herein. In one embodiment, alkynyl groups contain 2-8 carbon atoms; in another embodiment, alkynyl groups contain 2-6 carbon atoms; in yet another embodiment, alkynyl groups contain 2-4 carbon atoms. Examples of alkynyl groups include, but are not limited to, ethynyl (-C.ident.CH), propargyl (-CH)2C.ident.CH), 1-propynyl (i.e., propynyl, -C.ident.C-CH)3) And so on.
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 some embodiments, alkoxy groups contain 1 to 6 carbon atoms; in other embodiments, the alkoxy group contains 1 to 4 carbon atoms; in still other embodiments, alkoxy groups contain 1-3 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 toThen, methoxy (MeO, -OCH)3) Ethoxy (EtO, -OCH)2CH3) 1-propoxy (n-propoxy, n-PrO, n-propoxy, -OCH2CH2CH3) 2-propoxy (isopropoxy, 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) And so on.
The term "alkylthio" means an alkyl group attached to the rest of the molecule through a sulfur atom, wherein the alkyl group has the meaning as described herein. Unless otherwise specified, the alkylthio group contains 1 to 12 carbon atoms. In some embodiments, the alkylthio group contains 1 to 6 carbon atoms; in other embodiments, the alkylthio group contains 1 to 4 carbon atoms; in still other embodiments, the alkylthio group contains 1 to 3 carbon atoms. The alkylthio group may be optionally substituted with one or more substituents described herein. Examples of alkylthio groups include, but are not limited to, methylthio (MeS, -SCH)3) Ethylthio (EtS, -SCH)2CH3) And so on.
The term "alkylamino" or "alkylamino" denotes an amino group independently substituted with one or two alkyl groups, respectively, including "N-alkylamino" and "N, N-dialkylamino" wherein the alkyl groups have the meaning as described herein. Suitable alkylamino groups can be monoalkylamino or dialkylamino, and such examples include, but are not limited to, N-methylamino (methylamino), N-ethylamino (ethylamino), N-dimethylamino (dimethylamino), N-diethylamino (diethylamino), and the like. The alkylamino group is optionally substituted with one or more substituents described herein.
The term "hydroxy-substituted alkyl" denotes an alkyl group substituted with one or more hydroxy (-OH) groups, whereinThe alkyl group has the meaning as described in the present invention. Unless otherwise specified, the hydroxy-substituted alkyl group contains 1 to 12 carbon atoms. In some embodiments, the hydroxy-substituted alkyl group contains 1 to 6 carbon atoms, e.g., hydroxy-substituted C1-C6An alkyl group; in other embodiments, the hydroxy-substituted alkyl group contains 1 to 4 carbon atoms, e.g., hydroxy-substituted C1-C4An alkyl group; in still other embodiments, the hydroxy-substituted alkyl group contains 1 to 3 carbon atoms, e.g., hydroxy-substituted C1-C3An alkyl group. Examples include, but are not limited to, hydroxymethyl, hydroxyethyl (e.g., 2-hydroxyethyl), 2-hydroxy-1-propyl, 3-hydroxy-1-propyl, 2, 3-dihydroxypropyl, and the like.
The term "haloalkyl" or "haloalkoxy" means an alkyl or alkoxy group substituted with one or more halogen atoms, wherein the alkyl and alkoxy groups have the meaning as described herein, examples of which include, but are not limited to, -CHF2、-CF3、-CHFCH2F、-CF2CHF2、-CH2CHF2、-CH2CF3、-CH2CF2CHF2、-OCHF2、-OCF3、-OCHFCH2F、-OCF2CHF2、-OCH2CHF2、-OCH2CF3、-OCH2CF2CHF2And the like. In some embodiments, C1-C6The haloalkyl group containing a fluorine-substituted C1-C6An alkyl group; in other embodiments, C1-C4The haloalkyl group containing a fluorine-substituted C1-C4An alkyl group; in still other embodiments, C1-C2The haloalkyl group containing a fluorine-substituted C1-C2An alkyl group.
The term "cycloalkyl" denotes a saturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 ring carbon atoms. In some embodiments, cycloalkyl groups contain 3 to 10 ring carbon atoms, e.g., C3-10A cycloalkyl group; in other embodiments, cycloalkyl groups contain 3 to 8 ring carbon atomsSeeds, e.g. C3-8A cycloalkyl group; in still other embodiments, cycloalkyl groups contain 3-6 ring carbon atoms, e.g., C3-6A cycloalkyl group. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like. Wherein, as described in the present invention, C3-8Cycloalkyl radicals including C3-6A cycloalkyl group; said C3-6Cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl. The cycloalkyl group may be optionally substituted with one or more substituents described herein.
The term "heterocyclyl" refers to a saturated or partially unsaturated monocyclic, bicyclic, or tricyclic ring system containing 3 to 12 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen atoms; wherein the heterocyclic group is non-aromatic and does not contain any aromatic ring. Unless otherwise specified, heterocyclyl may be carbon-or nitrogen-based, and-CH2-the group may optionally be replaced by-C (═ O) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atoms of the ring may optionally be oxidized to the N-oxide. The term "heterocyclyl" may be used interchangeably with the term "heterocycle". As described herein, the heterocyclyl group may consist of 3 to 8 atoms or 3 to 6 atoms, optionally selected from C, N, O or S and at least one atom being N, O or S; wherein the heterocyclic group consisting of 3 to 8 atoms includes a heterocyclic group consisting of 3 to 6 atoms; the heterocyclic group consisting of 3 to 6 atoms includes a heterocyclic group consisting of 3 to 5 atoms. Specifically, the heterocyclic group consisting of 3 to 6 atoms includes, but is not limited to, ethylene oxide, aziridinyl, azetidinyl, oxetanyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, thiazolidinyl, pyrazolidinyl, pyrazolinyl, oxazolidinyl, imidazolidinyl, piperidinyl, piperazinyl, morpholinyl, and the like. The heterocyclyl group may be optionally substituted with one or more substituents described herein.
The term "aryl" denotes monocyclic, bicyclic and tricyclic carbocyclic ring systems containing 6 to 14 ring atoms, or 6 to 12 ring atoms, or 6 to 10 ring atoms, wherein at least one ring is aromaticAnd one or more attachment points are attached to the rest of the molecule. The term "aryl" may be used interchangeably with the terms "aromatic ring" or "aromatic ring". Examples of the aryl group may include phenyl, indenyl, 2, 3-dihydro-1H-indenyl, naphthyl and anthryl. The aryl group may be optionally substituted with one or more substituents described herein. Unless otherwise stated, the group "C6-10Aryl "represents an aryl group containing from 6 to 10 ring carbon atoms.
The term "heteroaryl" denotes monocyclic, bicyclic and tricyclic ring systems 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 ring contains 1, 2,3 or 4 ring heteroatoms selected from nitrogen, oxygen, sulfur, and wherein the heteroaryl has one or more attachment points to the rest of the molecule. when-CH is present in the heteroaryl group2When it is a group, -CH2-the group may optionally be replaced by-C (═ O) -. Unless otherwise indicated, the heteroaryl group may be attached to the rest of the molecule (e.g., the main structure in the formula) via any reasonable site, which may be C or N. The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic ring" or "heteroaromatic compound". The heteroaryl group may be optionally substituted with one or more substituents described herein. In some embodiments, heteroaryl is 5-10 atom consisting of heteroaryl, meaning that heteroaryl contains 1-9 ring carbon atoms and 1, 2,3, or 4 ring heteroatoms selected from O, S and N; in other embodiments, heteroaryl is 5-6 atom heteroaryl, meaning that heteroaryl contains 1-5 ring carbon atoms and 1, 2,3, or 4 ring heteroatoms selected from O, S and N; examples of heteroaryl groups consisting of 5 to 6 atoms include, but are not limited to, furyl, imidazolyl, isoxazolyl, oxazolyl, pyrrolyl, pyrazolyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, thienyl, thiazolyl, triazolyl, tetrazolyl, and the like.
The term "j-k atoms" means that the cyclic group consists of j-k ring atoms including carbon atoms and/or heteroatoms such as O, N, S, P; j and k are each independently any non-zero natural number, and k > j; the term "j-k" includes j, k and any natural number therebetween. For example, "3 to 8 atoms", "3 to 6 atoms", "5 to 10 atoms" or "5 to 6 atoms" means that the cyclic group consists of 3 to 8 (i.e., 3,4,5, 6, 7 or 8), 3 to 6 (i.e., 3,4,5 or 6), 5 to 10 (i.e., 5, 6, 7, 8, 9 or 10) or 5 to 6 (i.e., 5 or 6) ring atoms including carbon atoms and/or heteroatoms such as O, N, S, P.
The terms "alkoxyalkylene", "cycloalkylalkylene", "heterocycloalkylenealkylene", "arylalkylene", "heteroarylalkylene" mean that the alkoxy, cycloalkyl, heterocyclyl, aryl or heteroaryl groups are each independently linked to the rest of the molecule through an alkylene group, wherein the alkoxy, cycloalkyl, heterocyclyl, aryl, heteroaryl and alkylene groups all have the meaning described herein. For example, examples of arylalkylene groups include, but are not limited to, phenylmethylene, phenylethylene, phenylpropylene, and the like. The alkoxyalkylene, cycloalkylalkylene, heterocyclylalkylene, arylalkylene, heteroarylalkylene are each independently optionally substituted with one or more substituents described herein.
The term "prodrug", as used herein, represents a compound that is converted in vivo to a compound of formula (I), formula (II) or formula (III). 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)1-24) 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.
"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, description of the scientific 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. 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. 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-C8Sulfonates 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, ethanolamine, or mixtures thereof. 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 one embodiment, a molecule of a compound of the present invention may be associated with a molecule of water, such as a monohydrate; in another embodiment, one molecule of the compound of the present invention may be associated with more than one molecule of water, such as a dihydrate; in yet another embodiment, 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" or "treatment" as used herein refers, in some embodiments, to ameliorating a 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 "prevent" or "prevention" refers to a reduction in the risk of acquiring a disease or disorder (i.e., arresting the development of at least one clinical symptom of a disease in a subject that may be facing or predisposed to facing such a disease, but who has not yet experienced or exhibited symptoms of the disease).
The term "therapeutically effective amount" means an amount of a compound that, when administered to a subject to treat a disease, is sufficient to effect treatment of the disease. The "therapeutically effective amount" may vary with the compound, the disease and the severity, as well as the condition, age, weight, sex, etc., of the subject to be treated.
Unless otherwise indicated, all suitable isotopic variations, stereoisomers, tautomers, solvates, metabolites, pharmaceutically acceptable salts and prodrugs thereof, of the compounds of the present invention are encompassed within the scope of the present 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.
Nitroxides of the compounds of the present invention are also included within the scope of the present invention. The nitroxides of the compounds of the present invention may be prepared by oxidation of the corresponding nitrogen-containing basic species using a common oxidizing agent (e.g. hydrogen peroxide) in the presence of an acid such as acetic acid at elevated temperature, or by reaction with a peracid in a suitable solvent, for example peracetic acid in dichloromethane, ethyl acetate or methyl acetate, or 3-chloroperoxybenzoic acid in chloroform or dichloromethane.
The compounds of formula (I), formula (II) or formula (III) may be present in the form of a salt. In some embodiments, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal being treated therewith. In other embodiments, the salt need not be a pharmaceutically acceptable salt, and may be an intermediate useful in the preparation and/or purification of a compound of formula (I), formula (II) or formula (III) and/or in the isolation of an enantiomer of a compound of formula (I), formula (II) or formula (III).
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 invention relates to intermediates for the preparation of compounds of formula (I), formula (II) or formula (III).
Pharmaceutical compositions, formulations and administration of the compounds of the invention
The invention provides a pharmaceutical composition, which comprises a compound shown as a formula (I), a formula (II) or a formula (III) or an individual stereoisomer, a racemic or non-racemic mixture of isomers or a pharmaceutically acceptable salt or solvate thereof. In some embodiments of the invention, the pharmaceutical composition further comprises at least one pharmaceutically acceptable carrier, adjuvant or vehicle, and optionally, other therapeutic and/or prophylactic ingredients.
Suitable carriers, adjuvants and excipients are well known to those skilled in the art and are described in detail, for example, in Ansel h.c.et al, Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems (2004) Lippincott, Williams & Wilkins, philidelphia; gennaro a.r.et al, Remington: the Science and Practice of Pharmacy (2000) Lippincott, Williams & Wilkins, Philadelphia; and Rowe R.C., Handbook of Pharmaceutical Excipients (2005) Pharmaceutical Press, Chicago.
It will also be appreciated that certain compounds of the invention may be present in free form or, if appropriate, in the form of a pharmaceutically acceptable derivative thereof, when used in therapy. Some non-limiting embodiments of 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.
As used herein, "pharmaceutically acceptable excipient" means a pharmaceutically acceptable material, mixture or vehicle that 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 in order to avoid interactions that would substantially reduce the efficacy of the compounds of the invention when administered to a patient and interactions that would result in a pharmaceutical composition that is not pharmaceutically acceptable. 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 a compound of the invention 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 which other excipients are present in the formulation.
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. The compounds of the invention are preferably administered orally.
In some embodiments, the compounds disclosed herein can be formulated in oral dosage forms. In other embodiments, the compounds disclosed herein may be formulated in an inhalation dosage form. In other embodiments, the compounds disclosed herein can be formulated for nasal administration. In still other embodiments, the compounds disclosed herein can be formulated for transdermal administration. In still other embodiments, the compounds disclosed herein may be formulated for topical administration.
In practice, the compounds of the present invention may be combined as the active ingredient in intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier can take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). In preparing the compositions for oral dosage form, any of the usual pharmaceutical media may be employed, such as water, glycols, alcohols, oils, preservatives, flavoring agents, coloring agents and the like. In the case of oral liquid preparations, any conventional pharmaceutical media can be used, for example, elixirs, solutions and suspensions; or carriers such as sugars, starches, diluents, disintegrating agents, lubricants, granulating agents, binders, microcrystalline cellulose and the like. In the case of oral solid preparations, the compositions may be presented, for example, as powders, hard and soft capsules, and as tablets. Solid oral dosage forms are preferred over liquid dosage forms.
The pharmaceutical compositions provided by the present invention may be provided as compressed tablets, milled tablets, chewable lozenges, fast-dissolving tablets, double-compressed tablets, enteric-coated tablets, sugar-coated or film-coated tablets.
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 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).
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.
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.
In one embodiment, a compound of the invention or a pharmaceutical composition comprising a compound of the invention 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 compounds of the invention or pharmaceutical compositions comprising the compounds of the invention depend on the pharmacokinetic properties of the compound, such as absorption, distribution and half-life, which can be determined by the skilled person. In addition, the appropriate dosage regimen, including the duration of the regimen, of the compound of the invention or of the pharmaceutical composition containing the compound of the invention depends 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 appropriate dosage regimens may be required to be adjusted for the individual patient's response to the dosage regimen, or as the individual patient needs to change over time.
The compounds of the present invention may be administered simultaneously, or before or after one or more other additional therapeutic agents. The compounds of the invention may be administered separately from other additional therapeutic agents, by the same or different routes of administration, or in the same pharmaceutical composition.
The present invention also relates to a compound or pharmaceutical composition for inhibiting abnormal cell growth in a mammal comprising an amount of a compound of the present invention or a pharmaceutically acceptable salt or solvate or prodrug thereof and an amount of an additional anticancer therapeutic, wherein the amount of the compound, salt, solvate or prodrug together with the amount of the additional anticancer therapeutic is effective to inhibit abnormal cell growth. Many anti-cancer therapeutic agents are currently known in the art. In one embodiment, the anti-cancer therapeutic is an antibody selected from the group consisting of: zaolizumab, trastuzumab, bevacizumab, disitumumab, lintuzumab, ibritumomab, rituximab, cetuximab, tiximumab, IGF 1R-specific antibody, CD 40-specific antibody, WX G250, and chTNT-1/B. In another embodiment, the anti-cancer therapeutic is a chemotherapeutic agent selected from the group consisting of: enzymes, alkylating agents, anti-hormones, anti-androgens, anti-metabolites, intercalating antibiotics (intercalating antibiotics), mitotic inhibitors, growth factor inhibitors, topoisomerase inhibitors, cell cycle inhibitors, angiogenesis inhibitors, and biological response modifiers. In yet another embodiment, the anti-cancer therapeutic is an inhibitor of another protein kinase, such as Vegfr1, Vegfr2, Vegfr3 (also known as Flt-4), Flt-3, Akt, Axl, Aurora A, Aurora B, dyrk2, epha2, fgfr3, igf1r, IKK2, JNK3, KDR, MEK, MET, Plk1, PDK1, RSK1, TAK1, Src, ZA, Zap70, cKit, bRaf, EGFR, PDGFR, Jak2, PI3K, NPM-Alk, c-Abl, BTK, FAK, LimK, and Erk.
The term "therapeutically effective amount" as used herein refers to the total amount of each active ingredient sufficient to show meaningful patient benefit. When the active ingredient alone is used for separate administration, the term refers only to that ingredient. When used in combination, the term refers to the combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, sequentially or simultaneously. By this is meant that, when properly administered, any amount of the compound is sufficient to achieve the desired prophylactic or therapeutic effect in the patient to which it is administered. Generally, it will depend on the condition to be prevented or treated and the route of administration. Substantially satisfactory results are obtained when a daily dose of from about 0.01mg to about 100mg per kg of animal body weight is administered, preferably a single daily dose. For large mammals, the total daily dosage is from about 0.1mg to about 1000mg, preferably from about 0.2mg to about 50 mg. In the case of a 70kg adult, the total daily dose is approximately about 0.2mg to 200 mg. The dosage regimen may be adjusted to provide the optimum therapeutic response. The amount administered, the route of administration and further treatment regimens may be determined by the treating clinician, depending on, for example: age, sex, general condition of the patient, and the nature and severity of the disease/condition to be treated.
The compounds of the invention anduse of pharmaceutical composition
In one aspect, the present invention provides compounds and pharmaceutical compositions having excellent antitumor activity and low toxicity (e.g., acute toxicity, chronic toxicity, genetic toxicity, reproductive toxicity, cardiotoxin, drug interaction, and carcinogenicity). Also, the compound or the pharmaceutical composition is excellent in oral absorbability, action sustainability, stability and pharmacokinetics. Furthermore, the compounds or pharmaceutical compositions are less affected by plasma components. Therefore, the compound or the pharmaceutical composition of the present invention can be safely used in mammals (e.g., human, monkey, cow, horse, dog, cat, rabbit, rat, mouse, etc.) for the prevention and/or treatment of tumors.
In some embodiments, the tumor of the invention is a solid tumor, a B cell lymphoma, and/or a hematologic and immune system tumor.
In some embodiments, the solid tumor according to the present invention is gastric cancer, intestinal cancer, liver cancer, kidney cancer, lung cancer, brain cancer, laryngeal cancer, nasopharyngeal cancer, esophageal cancer, gallbladder cancer, head and neck cancer, squamous carcinoma, cancer of the lymphatic system, thyroid cancer, bladder cancer, ovarian cancer, cervical cancer, endometrial cancer, testicular cancer, prostate cancer, genitourinary tract cancer, breast cancer, small cell lung cancer, lung adenocarcinoma, pancreatic cancer, colon cancer, rectal cancer, glioblastoma, and/or monocytic leukemia.
In other embodiments, the B cell lymphoma according to the present invention is diffuse large B cell lymphoma, small lymphocytic tumor, mantle cell lymphoma, multiple myeloma, and/or chronic lymphocytic leukemia.
In still other embodiments, the hematologic and immune system tumors of the present invention are acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myeloid leukemia, and/or chronic myeloid leukemia.
In another aspect, the compounds or pharmaceutical compositions of the invention are used for the prevention and/or treatment of an inflammatory disorder or an autoimmune disease.
In some embodiments, the inflammatory disorder or autoimmune disease described herein is asthma, psoriasis, multiple sclerosis, rheumatoid arthritis, systemic lupus erythematosus, a heteroimmune disease, idiopathic thrombocytopenic purpura, immune complex-mediated vasculitis, and/or autoimmune-mediated hemolytic anemia.
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.
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 invention may be prepared by the methods described herein, wherein the substituents are as defined for formula (I), formula (II) or formula (III), unless otherwise specified. 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.
The examples described below, unless otherwise indicated, are all temperatures set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company and were used without further purification unless otherwise indicated. General reagents were purchased from Shantou Wen Long chemical reagent factory, Guangdong Guanghua chemical reagent factory, Guangzhou chemical reagent factory, Tianjin Haojian Yunyu chemical Co., Ltd, Tianjin Shucheng chemical reagent factory, Wuhan Xin Huayuan scientific and technological development Co., Ltd, Qingdao Tenglong chemical reagent Co., Ltd, and Qingdao Kaolingyi factory.
The anhydrous tetrahydrofuran, dioxane, toluene and ether are obtained through reflux drying of metal sodium. The anhydrous dichloromethane and chloroform are obtained by calcium hydride reflux drying. Ethyl acetate, petroleum ether, N-hexane, N, N-dimethylacetamide and N, N-dimethylformamide were used as they were previously dried over anhydrous sodium sulfate.
The following reactions are generally carried out under positive pressure of nitrogen or argon or by sleeving a dry tube over an anhydrous solvent (unless otherwise indicated), the reaction vial being stoppered with a suitable rubber stopper and the substrate being injected by syringe. The glassware was dried.
The column chromatography is performed using a silica gel column. Silica gel (300 and 400 meshes) was purchased from Qingdao oceanic chemical plants.
1H NMR spectra were recorded using a Bruker 400MHz or 600MHz NMR spectrometer.1H NMR Spectrum in CDC13、DMSO-d6、CD3OD or acetone-d6TMS (0ppm) or chloroform (7.26ppm) was used as a reference standard for the solvent (in ppm). When multiple peaks occur, the following abbreviations will be used: s (singlets, singlet), d (doublets ), t (triplets, triplet), q (quatets, quartets), m (multiplets ), br (broadended, broad), brs (broadended singlets, broad singlet), dd (doublets ), ddd (doublets, doublets), ddles of doublets), ddt (doublets of doublets, doublets), dt (doublets ), dq (doublets of doublets, doublets), td (triplets of doublets, triplet), tt (triplet of triplets). Coupling constant J, expressed in Hertz (Hz).
The conditions for determining low resolution Mass Spectrometry (MS) data were: agilent 6120 four-stage rod HPLC-M (column model: Zorbax SB-C18, 2.1X 30mm,3.5 micron, 6min, flow rate 0.6 mL/min. mobile phase 5% -95% (CH with 0.1% formic acid)3CN) in (H containing 0.1% formic acid)2O) by electrospray ionization (ESI) at 210nm/254nm, with UV detection.
Pure compounds were detected by UV at 210nm/254nm using Agilent 1260pre-HPLC or Calesep pump 250pre-HPLC (column model: NOVASEP 50/80mm DAC).
The following acronyms are used throughout the invention:
DBU 1, 8-diazabicycloundec-7-ene mM, mol/L, millimoles per liter
CDC13Deuterated chloroform mu M, nmol/L micromol per liter
DMSO nM, nmol/L nanomole per liter
DMSO-d6Deuterated dimethyl sulfoxide M, mol/L, mol per liter
EA ethyl acetate mg
DCM dichloromethane g
RT, RT Room temperature kg
min mL, mL
h μ L, μ L
Mmol millimoles of DIPEA N, N-diisopropylethylamine
Pd/C palladium/carbon 4A molecular sieve 1A is 10-10m
The following synthetic schemes describe the steps for preparing the presently disclosed compounds, wherein R is, unless otherwise indicated3、R4And R6Having the definitions set out in the present invention.
Synthesis scheme 1
Formula (A), (B) and9) Shown inThe compound can be prepared by the following processes: formula (A), (B) and1) Reacting the compound with trichloroacetonitrile under alkaline condition (such as DBU) to obtain a compound shown in a formula (A)2) A compound shown in the specification; then formula (A), (B), (C), (2) A compound of the formula3) The compound shown in the formula (I) undergoes glycosylation reaction to obtain a compound shown in the formula (II)4) The compounds shown. Formula (A), (B) and4) The nitro group of the compound is reduced to obtain the compound of the formula (A)5) The compounds shown. Formula (A), (B) and5) Reacting the compound with triphosgene under alkaline condition (such as DIPEA) to obtain the compound shown in the formula (A)6) A compound shown in the specification; formula (A), (B) and6) A compound of the formula7) Nucleophilic addition reaction of the compound shown in the formula (I) to obtain a compound shown in the formula (I)8) The compounds shown. Formula (A), (B) and8) The compound shown in the formula (I) is deacetylated to obtain the compound shown in the formula (II)9) The target product shown.
Figure BDA0002915655280000151
Synthesis scheme 2
Formula (A), (B) and9’) The compound shown can be prepared by the following steps: formula (A), (B) and1) Reacting the compound with trichloroacetonitrile under alkaline condition (such as DBU) to obtain a compound shown in a formula (A)2) A compound shown in the specification; then formula (A), (B), (C), (2) A compound of the formula3’) The compound shown in the formula (I) undergoes glycosylation reaction to obtain a compound shown in the formula (II)4’) The compounds shown. Formula (A), (B) and4’) The nitro group of the compound is reduced to obtain the compound of the formula (A)5’) The compounds shown. Formula (A), (B) and5’') Reacting the compound with triphosgene under alkaline condition (such as DIPEA) to obtain the compound shown in the formula (A)6’) A compound shown in the specification; formula (A), (B) and6’) A compound of the formula7) Nucleophilic addition reaction of the compound shown in the formula (I) to obtain a compound shown in the formula (I)8’) The compounds shown. Formula (A), (B) and8’) The compound shown in the formula (I) is deacetylated to obtain the compound shown in the formula (II)9’) The target product shown.
Figure BDA0002915655280000161
The compounds, pharmaceutical compositions and uses thereof provided by the present invention are further illustrated below in connection with the examples.
Examples
Example 14 Synthesis of- (3-chloro-4- (3- (4- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000162
Step 1) (2R,3S,4S,5R,6R) -2- (acetoxymethyl) -6- (2,2, 2-trichloro-1-iminoethoxy) tetra Synthesis of hydrogen-2H-pyran-3, 4, 5-triyltriacetate
Figure BDA0002915655280000163
Adding (2R,3S,4S,5R) -2- (acetoxymethyl) -6-hydroxytetrahydro-2H-pyran-3, 4, 5-triyltriacetate (3.6g,10.3mmol), trichloroacetonitrile (10.4mL,104mmol), 4A molecular sieve (3g) and dichloromethane (30mL) into a 100mL single-neck round-bottom flask, stirring in ice bath for 0.5H under the protection of nitrogen, then adding DBU (0.32mL,2.1mmol), and stirring in ice bath for reaction for 0.5H; the reaction was stopped, filtered, the filtrate was collected, spin-dried under reduced pressure, and purified by column chromatography (petroleum ether/ethyl acetate (v/v) ═ 4/1) to give the title compound as a white solid (4.56g, 89.5%).
Step 2) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-nitrophenoxy) tetrahydro-2H-pyran-3, synthesis of 4, 5-Tritriacetic acid ester
Figure BDA0002915655280000171
Adding (2R,3S,4S,5R,6R) -2- (acetoxymethyl) -6- (2,2, 2-trichloro-1-iminoethoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (1.41g,2.86mmol), 4-nitrophenol (0.8g,5.75mmol), 4A molecular sieve (5g) and dichloromethane (20mL) into a 100mL single-neck round-bottom flask, stirring for 30 minutes at-25 ℃ under nitrogen protection, then dropwise adding boron trifluoride diethyl ether (0.45mL,3.5mmol), and continuing stirring for half an hour; the reaction was stopped, quenched by addition of triethylamine (0.5mL,3.5mmol), the solvent was removed by rotary drying under reduced pressure, and purified by column chromatography (petroleum ether/ethyl acetate (v/v) ═ 2/1) to give the title compound as a pale yellow sticky substance (1.2g, 90%).
MS(ESI,pos.ion)m/z:492.5[M+Na]+
1H NMR(400MHz,CDCl3)δ(ppm)8.22–8.20(m,2H),7.09–7.07(m,2H),5.52(dd,J=10.4,7.9Hz,1H),5.48(d,J=3.1Hz,1H),5.18(d,J=7.9Hz,1H),5.14(dd,J=10.4,3.4Hz,1H),4.23–4.17(m,2H),4.14(d,J=5.8Hz,1H),2.19(s,3H),2.07(s,6H),2.02(s,3H).
Step 3) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-aminophenoxy) tetrahydro-2H-pyran-3, synthesis of 4, 5-Tritriacetic acid ester
Figure BDA0002915655280000172
(2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-nitrophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyl triacetate (1.13g,2.41mmol), Pd/C (240mg), tetrahydrofuran (10mL) and methanol (20mL) were charged at 25 ℃ into a 100mL single-necked round-bottomed flask, reacted under a hydrogen balloon for 13 hours to stop the reaction, and purified by column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1/1) to give the title compound as a pale yellow solid (1.02g, 96.6%).
MS(ESI,pos.ion)m/z:440.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)6.85–6.83(m,2H),6.61–6.59(m,2H),5.43(dd,J=10.4,8.0Hz,2H),5.07(dd,J=10.5,3.4Hz,1H),4.87(d,J=8.0Hz,1H),4.23(dd,J=11.2,6.8Hz,1H),4.15(dd,J=11.2,6.6Hz,1H),3.98(t,J=6.7Hz,1H),3.53(s,2H),2.18(s,3H),2.08(s,3H),2.05(s,3H),2.00(s,3H).
Step 4) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-isocyanatophenoxy) tetrahydro-2H-py-ridine Synthesis of pyran-3, 4, 5-triyltriacetanoate
Figure BDA0002915655280000173
(2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-aminophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyl triacetate (0.51g,1.16mmol), DIPEA (0.41mL,2.3mmol) and dichloromethane (10mL) were added to a 100mL single neck round bottom flask at 0 deg.C, followed by dropwise addition of triphosgene (115mg,0.387mmol, dissolved in 10mL dichloromethane), stirring was continued for 2 hours, the reaction was stopped, and the solvent was dried under reduced pressure to give the title compound as a pale yellow viscous substance (0.54g, 100%) which was directly charged to the next reaction.
Step 5) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4- (3- (4- ((6-carbamoyl-7-methoxy) Synthesis of quinolinyl-4-yl) oxy) -2-chlorophenyl) urea) phenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate
Figure BDA0002915655280000174
(2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-isocyanatophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyl triacetate (0.54g,1.16mmol) and dichloromethane (20mL) were added to a 100mL single-neck round-bottomed flask at 0 deg.C, then 4- (4-amino-3-chlorophenoxy) -7-methoxyquinoline-6-carboxamide (400mg,1.16mmol) was added and transferred to 25 deg.C for 24 hours to stop the reaction, spin-dried under reduced pressure, and purified by column chromatography (dichloromethane/methanol (v/v) ═ 40/1) to give the title compound as a reddish brown solid (0.433g, 46.1%).
MS(ESI,pos.ion)m/z:809.0[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)9.28(s,2H),8.66(d,J=5.2Hz,1H),8.32(d,J=8.8Hz,2H),7.88(s,1H),7.60(s,1H),7.52(d,J=8.9Hz,2H),7.15(d,J=2.6Hz,1H),7.05(dd,J=9.1,2.6Hz,1H),6.96(d,J=8.9Hz,2H),6.51(d,J=5.3Hz,1H),6.09(s,1H),5.49–5.45(m,2H),5.12(dd,J=10.4,3.3Hz,1H),4.98(d,J=8.0Hz,1H),4.24–4.18(m,2H),4.15(s,3H),4.06(t,J=6.6Hz,1H),2.19(s,3H),2.09(s,3H),2.07(s,3H),2.02(s,3H).
Step 6)4- (3-chloro-4- (3- (4- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) tetrahydro- Synthesis of 2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000181
(2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4- (3- (4- ((6-carbamoyl-7-methoxyquinolin-4-yl) oxy) -2-chlorophenyl) urea) phenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.43g,0.53mmol) and methanolic ammonia (15mL,7M) were added to a 100mL single neck round bottom flask at 25 deg.C and the reaction was allowed to continue stirring for 48 hours, the reaction was stopped, filtered, and the filter cake was dried in vacuo to give the title compound as a tan solid (0.17g, 51.1%).
MS(ESI,pos.ion)m/z:641.1[M+H]+.
Example Synthesis of 24- (3-chloro-4- (3- (3-fluoro-4- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000182
Step 1) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-fluoro-4-nitrophenoxy) tetrahydro-2H-pyri-dine Synthesis of pyran-3, 4, 5-triyltriacetanoate
Figure BDA0002915655280000183
The title compound was prepared as described in example 1, step 2 by reacting (2R,3S,4S,5R,6R) -2- (acetoxymethyl) -6- (2,2, 2-trichloro-1-iminoethoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (1.82g,3.69mmol), 2-fluoro-4-nitrophenol (1.16g,7.38mmol), 4A molecular sieve (5g) and boron trifluoride etherate (0.57mL,4.4mmol) in dichloromethane (20mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a pale yellow solid (1.28g, 70.8%).
MS(ESI,pos.ion)m/z:510.0[M+Na]+
1H NMR(400MHz,CDCl3)δ(ppm)8.03(t,J=3.4Hz,1H),8.02–8.00(m,1H),7.32–7.28(m,1H),5.55(dd,J=10.4,7.9Hz,1H),5.47(d,J=2.6Hz,1H),5.14–5.10(m,2H),4.25–4.16(m,2H),4.09(t,J=6.5Hz,1H),2.19(s,3H),2.09(s,3H),2.06(s,3H),2.02(s,3H).
Step 2) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-amino-2-fluorophenoxy) tetrahydro-2H-pyri-dine Synthesis of pyran-3, 4, 5-triyltriacetanoate
Figure BDA0002915655280000191
The title compound was prepared as described in example 1, step 3, by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-fluoro-4-nitrophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyl triacetate (1.18g,2.42mmol), Pd/C (0.24g) in tetrahydrofuran (10mL) and methanol (20mL) and purifying the crude by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1/1) to give the title compound as a white solid (1.0g, 90.4%).
MS(ESI,pos.ion)m/z:458.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)7.00(t,J=8.8Hz,1H),6.41(dd,J=12.1,2.6Hz,1H),6.34–6.31(m,1H),5.46–5.42(m,2H),5.06(dd,J=10.5,3.4Hz,1H),4.73(d,J=8.0Hz,1H),4.23(dd,J=11.2,6.7Hz,1H),4.14(dd,J=11.2,6.8Hz,1H),3.92(t,J=6.7Hz,1H),3.64(s,2H),2.18(s,3H),2.11(s,3H),2.03(s,3H),2.00(s,3H).
Step 3) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-fluoro-4-isocyanatophenoxy) tetrahydro- Synthesis of 2H-pyran-3, 4, 5-triyltriacetate
Figure BDA0002915655280000192
The title compound was prepared as described in example 1, step 4, by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-amino-2-fluorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.23g,0.5mmol), triphosgene (50mg,0.2mmol) and DIPEA (0.18mL,1.0mmol) in dichloromethane (10mL) to give the title compound as a pale yellow sticky mass (0.242g, 99.6%).
Step 4) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4- (3- (4- ((6-carbamoyl-7-methoxy) Combinations of quinolinyl-4-yl) oxy) -2-chlorophenyl) urea) -2-fluorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate Become into
Figure BDA0002915655280000193
The title compound was prepared as described in example 1, step 5 by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-fluoro-4-isocyanatophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.242g,0.5mmol), 4- (4-amino-3-chlorophenoxy) -7-methoxyquinoline-6-carboxamide (172mg,0.5mmol) in dichloromethane (20mL) and purifying the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 50/1) to give the title compound as a reddish brown solid (0.324g, 78.2%).
MS(ESI,pos.ion)m/z:827.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)9.34(s,1H),8.77(s,1H),8.64(d,J=5.3Hz,1H),8.33–8.30(m,2H),8.21(d,J=3.6Hz,1H),7.57(s,1H),7.44(dd,J=12.6,2.3Hz,1H),7.11(t,J=8.8Hz,1H),7.03(d,J=8.9Hz,1H),6.93(dd,J=7.4,2.4Hz,2H),6.40(d,J=5.3Hz,1H),6.19(d,J=3.7Hz,1H),5.49–5.43(m,2H),5.08(dd,J=10.5,3.4Hz,1H),4.83(d,J=8.0Hz,1H),4.24(dd,J=11.2,6.6Hz,1H),4.17(s,3H),4.15–4.12(m,1H),3.95(t,J=6.8Hz,1H),2.18(s,3H),2.12(s,3H),2.04(s,3H),2.01(s,3H).
Step 5)4- (3-chloro-4- (3- (3-fluoro-4- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) Synthesis of tetrahydro-2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000201
The title compound was prepared as described in example 1, step 6 by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4- (3- (4- ((6-carbamoyl-7-methoxyquinolin-4-yl) oxy) -2-chlorophenyl) urea) -2-fluorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyl triacetate (0.405g,0.49mmol) in methanolic ammonia (15mL,7M) and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 5/1) to give the title compound as a tan solid (0.24g, 75%).
MS(ESI,pos.ion)m/z:659.1[M+H]+.
Example 34 Synthesis of- (3-chloro-4- (3- (3-chloro-4- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000202
Step 1) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-chloro-4-nitrophenoxy) tetrahydro-2H-pyri-dine Synthesis of pyran-3, 4, 5-triyltriacetanoate
Figure BDA0002915655280000203
The title compound was prepared as described in example 1, step 2 by reacting (2R,3S,4S,5R,6R) -2- (acetoxymethyl) -6- (2,2, 2-trichloro-1-iminoethoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (1.72g,3.49mmol), 2-chloro-4-nitrophenol (0.73g,4.2mmol), 4A molecular sieve (5g) and boron trifluoride etherate (0.54mL,4.2mmol) in dichloromethane (20mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a white solid (1.16g, 65.8%).
MS(ESI,pos.ion)m/z:526.0[M+Na]+
1H NMR(600MHz,CDCl3)δ(ppm)8.31(d,J=2.7Hz,1H),8.13(dd,J=9.1,2.7Hz,1H),7.27(d,J=9.4Hz,1H),5.62(dd,J=10.5,7.9Hz,1H),5.49(d,J=2.9Hz,1H),5.13(dd,J=10.5,3.4Hz,1H),5.10(d,J=7.9Hz,1H),4.25(dd,J=11.4,7.1Hz,1H),4.18(dd,J=11.4,5.9Hz,1H),4.13(t,J=6.8Hz,1H),2.20(s,3H),2.09(s,3H),2.08(s,3H),2.03(s,3H).
Step 2) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-amino-2-chlorophenoxy) tetrahydro-2H-pyrazine Synthesis of pyran-3, 4, 5-triyltriacetanoate
Figure BDA0002915655280000211
(2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-chloro-4-nitrophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (1.14g,2.25mmol), iron powder (0.63g,11mmol), ammonium chloride (0.24g,4.5mmol), tetrahydrofuran (10mL), ethanol (10mL), and water (2mL) were added to a 100mL single-neck round-bottom flask, and the reaction was stirred at 75 ℃ in an oil bath for 14 hours; the reaction was stopped, filtered, the filtrate was spin-dried under reduced pressure, and purified by column chromatography (dichloromethane/methanol (v/v) ═ 50/1) to give the title compound as a pale yellow solid (0.95g, 89.2%).
MS(ESI,pos.ion)m/z:474.2[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)7.04(d,J=8.7Hz,1H),6.69(d,J=2.7Hz,1H),6.48(dd,J=8.7,2.7Hz,1H),5.50(dd,J=10.5,8.0Hz,1H),5.43(d,J=2.8Hz,1H),5.08(dd,J=10.5,3.4Hz,1H),4.79(d,J=8.0Hz,1H),4.24(dd,J=11.2,6.8Hz,1H),4.14(dd,J=11.2,6.6Hz,1H),3.95(t,J=6.4Hz,1H),2.18(s,3H),2.11(s,3H),2.05(s,3H),2.01(s,3H).
Step 3) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-fluoro-4-isocyanatophenoxy) tetrahydro- Synthesis of 2H-pyran-3, 4, 5-triyltriacetate
Figure BDA0002915655280000212
This title compound was prepared as described in example 1, step 4, by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4-amino-2-fluorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.46g,0.98mmol), triphosgene (100mg,0.33mmol) and DIPEA (0.35mL,2.0mmol) in dichloromethane (20mL) to give the title compound as a pale yellow sticky mass (0.49g, 100%).
Step 4) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4- (3- (4- ((6-carbamoyl-7-methoxy) Combinations of quinolinyl-4-yl) oxy) -2-chlorophenyl) urea) -2-chlorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate Become into
Figure BDA0002915655280000213
The title compound was prepared as described in example 1, step 5 by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-chloro-4-isocyanatophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.49g,0.98mmol), 4- (4-amino-3-chlorophenoxy) -7-methoxyquinoline-6-carboxamide (0.337g,0.98mmol) in dichloromethane (30mL) and purifying the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 50/1) to give the title compound as a tan solid (0.416g, 50.3%).
1H NMR(400MHz,CDCl3)δ(ppm)9.29(s,2H),8.65(d,J=5.3Hz,1H),8.32(s,1H),8.29(d,J=9.0Hz,1H),7.97(d,J=3.3Hz,1H),7.63(d,J=2.5Hz,1H),7.58(s,1H),7.41(dd,J=8.9,2.5Hz,1H),7.14(d,J=8.9Hz,1H),7.07(d,J=2.7Hz,1H),7.01(dd,J=9.0,2.7Hz,1H),6.47(d,J=5.4Hz,1H),6.09(d,J=3.0Hz,1H),5.53(dd,J=10.5,8.0Hz,1H),5.45(d,J=3.0Hz,1H),5.10(dd,J=10.5,3.4Hz,1H),4.87(d,J=8.0Hz,1H),4.24(dd,J=11.2,6.9Hz,1H),4.17(d,J=6.3Hz,1H),4.14(s,3H),4.01(t,J=6.8Hz,1H),2.18(s,3H),2.10(s,3H),2.05(s,3H),2.01(s,3H).
Step 5)4- (3-chloro-4- (3- (3-chloro-4- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) Synthesis of tetrahydro-2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000221
The title compound was prepared as described in example 1, step 6 by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (4- (3- (4- ((6-carbamoyl-7-methoxyquinolin-4-yl) oxy) -2-chlorophenyl) urea) -2-chlorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyl triacetate (0.405g,0.48mmol) in methanolic ammonia (15mL,7M) and the crude product was purified by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 5/1) to give the title compound as a brown solid (0.182g, 56.1%).
MS(ESI,pos.ion)m/z:675.2[M+H]+
1H NMR(400MHz,DMSO-d6)δ(ppm)9.45(s,1H),8.69(d,J=7.5Hz,2H),8.41(s,1H),8.27(d,J=9.0Hz,1H),7.87(s,1H),7.73(d,J=6.8Hz,2H),7.56(d,J=2.4Hz,1H),7.53(s,1H),7.30(dd,J=9.1,2.4Hz,1H),7.22(s,2H),6.58(d,J=5.2Hz,1H),4.88(d,J=7.6Hz,2H),3.64–3.57(m,3H),3.51(d,J=8.4Hz,1H).
Example Synthesis of 44- (3-chloro-4- (3- (4-fluoro-3- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000222
Step 1) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-fluoro-5-nitrophenoxy) tetrahydro-2H-pyri-dine Synthesis of pyran-3, 4, 5-triyltriacetanoate
Figure BDA0002915655280000223
The title compound was prepared as described in example 1, step 2 by reacting (2R,3S,4S,5R,6R) -2- (acetoxymethyl) -6- (2,2, 2-trichloro-1-iminoethoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (1.23g,2.5mmol), 2-fluoro-5-nitrophenol (0.47g,3.0mmol), 4A molecular sieve (5g) and boron trifluoride etherate (0.39mL,3.0mmol) in dichloromethane (20mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a pale yellow sticky mass (1.04g, 85.3%).
MS(ESI,pos.ion)m/z:510.0[M+Na]+
1H NMR(400MHz,CDCl3)δ(ppm)8.16(dd,J=6.9,2.7Hz,1H),8.03–7.99(m,1H),7.25(t,J=9.2Hz,1H),5.55(dd,J=10.4,7.9Hz,1H),5.48(d,J=3.3Hz,1H),5.13(dd,J=10.5,3.4Hz,1H),5.07(d,J=7.9Hz,1H),4.26–4.19(m,2H),4.14–4.11(m,1H),2.20(s,3H),2.11(s,3H),2.10(s,3H),2.02(s,3H).
Step 2) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (5-amino-2-fluorophenoxy) tetrahydro-2H-pyri-dine Synthesis of pyran-3, 4, 5-triyltriacetanoate
Figure BDA0002915655280000231
The title compound was prepared as described in example 1, step 3 by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-fluoro-5-nitrophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (1.04g,2.13mmol), Pd/C (0.21g) in tetrahydrofuran (10mL) and methanol (20mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 1/1) to give the title compound as a white solid (0.83g, 85%).
MS(ESI,pos.ion)m/z:458.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)6.86(dd,J=10.5,8.8Hz,1H),6.53(dd,J=6.8,2.8Hz,1H),6.34(dt,J=8.7,3.3Hz,1H),5.47(dd,J=10.5,8.0Hz,1H),5.43(d,J=2.7Hz,1H),5.08(dd,J=10.5,3.4Hz,1H),4.89(d,J=8.0Hz,1H),4.25(dd,J=11.2,6.6Hz,1H),4.14(dd,J=11.3,6.8Hz,1H),3.98(dd,J=7.1,6.3Hz,1H),2.18(s,3H),2.09(s,3H),2.05(s,3H),2.01(s,3H).
Step 3) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-fluoro-5-isocyanatophenoxy) tetrahydro- Synthesis of 2H-pyran-3, 4, 5-triyltriacetate
Figure BDA0002915655280000232
This title compound was prepared as described in example 1, step 4, by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (5-amino-2-fluorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.53g,1.16mmol), triphosgene (114mg,0.384mmol) and DIPEA (0.41mL,2.3mmol) in dichloromethane (10mL) to give the title compound as a pale yellow sticky mass (0.556g, 99.3%).
Step 4) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (5- (3- (4- ((6-carbamoyl-7-methoxy) Combinations of quinolinyl-4-yl) oxy) -2-chlorophenyl) urea) -2-fluorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate Become into
Figure BDA0002915655280000233
The title compound was prepared as described in example 1, step 5 by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-fluoro-5-isocyanatophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.56g,1.16mmol), 4- (4-amino-3-chlorophenoxy) -7-methoxyquinoline-6-carboxamide (395mg,1.15mmol) in dichloromethane (30mL) and purifying the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 40/1) to give the title compound as a tan solid (0.378g, 39.4%).
MS(ESI,pos.ion)m/z:827.0[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)9.33(s,1H),8.74(s,1H),8.63(d,J=5.3Hz,1H),8.36(s,1H),8.32(d,J=9.2Hz,1H),8.17(d,J=3.3Hz,1H),7.57(s,1H),7.31(ddd,J=9.8,8.0,3.2Hz,3H),7.04–7.00(m,1H),6.93(s,1H),6.56(s,1H),6.39(d,J=5.3Hz,1H),5.51(dd,J=10.4,7.9Hz,1H),5.46(d,J=3.0Hz,1H),5.10(dd,J=10.5,3.4Hz,1H),4.96(d,J=8.0Hz,1H),4.38(dd,J=11.3,7.0Hz,1H),4.17(s,3H),4.12(dd,J=11.4,6.0Hz,1H),4.03(t,J=6.5Hz,1H),2.21(s,3H),2.11(s,3H),2.07(s,3H),2.02(s,3H).
Step 5)4- (3-chloro-4- (3- (4-fluoro-3- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) Synthesis of tetrahydro-2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000241
This step was prepared by the method described in example 1, step 6, i.e. (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (5- (3- (4- ((6-carbamoyl-7-methoxyquinolin-4-yl) oxy) -2-chlorophenyl) urea) -2-fluorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.372g,0.45mmol) was reacted in methanolic ammonia (15mL,7M) to give the title compound as a brown solid (0.121g, 40.8%).
MS(ESI,pos.ion)m/z:659.2[M+H]+.
Example Synthesis of 54- (3-chloro-4- (3- (4-chloro-3- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) tetrahydro-2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000242
Step 1) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-chloro-5-nitrophenoxy) tetrahydro-2H-pyri-dine Synthesis of pyran-3, 4, 5-triyltriacetanoate
Figure BDA0002915655280000243
The title compound was prepared as described in example 1, step 2 by reacting (2R,3S,4S,5R,6R) -2- (acetoxymethyl) -6- (2,2, 2-trichloro-1-iminoethoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (1.23g,2.5mmol), 2-chloro-5-nitrophenol (0.52g,3.0mmol), 4A molecular sieve (5g) and boron trifluoride etherate (0.39mL,3.0mmol) in dichloromethane (20mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) ═ 3/1) to give the title compound as a pale yellow solid (1.11g, 87.9%).
MS(ESI,pos.ion)m/z:525.9[M+Na]+
1H NMR(400MHz,CDCl3)δ(ppm)8.11(d,J=2.5Hz,1H),7.93(dd,J=8.8,2.5Hz,1H),7.55(d,J=8.8Hz,1H),5.61(dd,J=10.5,8.0Hz,1H),5.50(d,J=3.2Hz,1H),5.14(dd,J=10.5,3.4Hz,1H),5.09(d,J=7.9Hz,1H),4.25(dd,J=9.0,5.6Hz,1H),4.19(dt,J=7.0,3.6Hz,2H),2.19(s,3H),2.13(s,3H),2.09(s,3H),2.02(s,3H).
Step 2) (2R,3S,4S,5R,6S) -2- (Acetyloxymethyl) -6- (5-amino-2-chloro)Phenoxy) tetrahydro-2H-pyridine Synthesis of pyran-3, 4, 5-triyltriacetanoate
Figure BDA0002915655280000251
The title compound was prepared as described in example 1, step 3, by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-chloro-5-nitrophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyl triacetate (1.0g,1.98mmol), iron powder (0.56g,9.99mmol), ammonium chloride (0.214g,4.0mmol) in tetrahydrofuran (10mL), ethanol (10mL) and water (2mL) and purifying the crude product by silica gel column chromatography (petroleum ether/ethyl acetate (v/v) 1/1) to give the title compound as a pale yellow solid (0.86g, 91.8%).
MS(ESI,pos.ion)m/z:474.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)7.09(d,J=8.5Hz,1H),6.55(d,J=2.6Hz,1H),6.36(dd,J=8.5,2.6Hz,1H),5.54(dd,J=10.5,8.0Hz,1H),5.46(d,J=2.7Hz,1H),5.09(dd,J=10.5,3.4Hz,1H),4.90(d,J=8.0Hz,1H),4.29(dd,J=11.3,6.7Hz,1H),4.14(dd,J=11.3,6.6Hz,1H),4.02(t,J=6.6Hz,1H),3.70(s,2H),2.19(s,3H),2.09(s,3H),2.07(s,3H),2.02(s,3H).
Step 3) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-chloro-5-isocyanatophenoxy) tetrahydro- Synthesis of 2H-pyran-3, 4, 5-triyltriacetate
Figure BDA0002915655280000252
The title compound was prepared as described in example 1, step 4, by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (5-amino-2-chlorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.56g,1.18mmol), triphosgene (117mg,0.394mmol) and DIPEA (0.42mL,2.4mmol) in dichloromethane (10mL) to give the title compound as a pale yellow sticky mass (0.589g, 99.7%).
Step 4) (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (5- (3- (4- ((6-carbamoyl-7-methoxy) Combinations of quinolinyl-4-yl) oxy) -2-chlorophenyl) urea) -2-chlorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate Become into
Figure BDA0002915655280000253
The title compound was prepared as described in example 1, step 5 by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (2-chloro-5-isocyanatophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyltriacetate (0.589g,1.18mmol), 4- (4-amino-3-chlorophenoxy) -7-methoxyquinoline-6-carboxamide (405mg,1.18mmol) in dichloromethane (30mL) and purifying the crude product by silica gel column chromatography (dichloromethane/methanol (v/v) ═ 40/1) to give the title compound as a tan solid (0.585g, 58.8%).
MS(ESI,pos.ion)m/z:843.1[M+H]+
1H NMR(400MHz,CDCl3)δ(ppm)9.32(s,1H),8.94(s,1H),8.63(d,J=5.3Hz,1H),8.43(s,1H),8.33(d,J=9.7Hz,1H),8.16(s,1H),7.56(s,1H),7.44(s,1H),7.24(s,2H),6.93–6.91(m,2H),6.52(s,1H),6.39(d,J=5.3Hz,1H),5.57(dd,J=10.4,8.0Hz,1H),5.47(d,J=3.0Hz,1H),5.12(dd,J=10.5,3.3Hz,1H),5.01(d,J=8.0Hz,1H),4.39(dd,J=10.7,6.5Hz,1H),4.16(s,3H),4.12–4.10(m,2H),2.20(s,3H),2.10(s,3H),2.06(s,3H),2.02(s,3H).
Step 5)4- (3-chloro-4- (3- (4-chloro-3- (((2S,3R,4S,5R,6R) -3,4, 5-trihydroxy-6- (hydroxymethyl) Synthesis of tetrahydro-2H-pyran-2-yl) oxy) phenyl) urea) phenoxy) -7-methoxyquinoline-6-carboxamide
Figure BDA0002915655280000261
The title compound was prepared as described in example 1, step 6 by reacting (2R,3S,4S,5R,6S) -2- (acetoxymethyl) -6- (5- (3- (4- ((6-carbamoyl-7-methoxyquinolin-4-yl) oxy) -2-chlorophenyl) urea) -2-chlorophenoxy) tetrahydro-2H-pyran-3, 4, 5-triyl triacetate (0.574g,0.68mmol) in methanolic ammonia (15mL,7M) to give the title compound as a white solid (0.201g, 43.6%).
MS(ESI,pos.ion)m/z:675.0[M+H]+.
Biological assay
EXAMPLE A Activity test of the Compound of the present invention for inhibition of cell proliferation
Purpose of experiment
Determination of IC of Compounds on cell proliferation Using the CTG method50The ability of the compounds of the present invention to inhibit cell proliferation was evaluated.
Experimental test cell lines
Figure BDA0002915655280000262
Remarking:
ATCC, American type culture collection; SIBS, Shanghai Institutes for Biological Sciences; ZHFU: subsidiary Zhongshan Hospital of the university of Fudan.
2. The cell culture conditions were: 37 ℃ and 5% CO2And 95% humidity.
Experimental methods
Cells in exponential growth phase were collected and counted for viable cells using a Vi-Cell XR Cell counter. The collected cells were adjusted to an appropriate concentration using DMEM medium (Thermo scientific, product No.: SH30243.01) or RPMI1640 medium (Thermo scientific, product No.: SH30809.01) used for culturing the cells. 90. mu.l of the collected cells were added to each well of a 96-well cell culture plate (corning, product No. 3610) at a final cell concentration of 1,000-3,000 cells/well, placed at 37 ℃ with 5% CO2After 24 hours incubation in an incubator, test compounds are added. Test compounds were dissolved in DMSO to 20mM stock. By usingStock solution and DMSO are prepared into 2.5-fold serial gradient dilution solution, and diluted into 9 gradients, and then each gradient dilution solution is diluted by 100 times by using culture medium. Finally, 10. mu.l of the dilution was added to each well, 3 wells for each test compound concentration, and 10. mu.l of 0.1% DMSO was added to each of the 3 wells as a solvent control. Standing at 37 deg.C for 5% CO2Incubate for 72 hours. After 72 hours of drug treatment, 50. mu.l (1/2 culture volume) of CTG solution previously thawed and equilibrated to room temperature was added to each cell well according to the instructions of the CTG kit (Promega, product No. G7572), mixed by a microplate shaker for 2 minutes, left at room temperature for 10 minutes, and the fluorescence signal value was measured by an Envision2104 plate reader. Cell viability using the formula: Vsample/Vvehicle control × 100% calculation. Where vsampe is the reading for the drug treated group and Vvehicle control is the mean value for the solvent control group. Sigmoidal dose-survival curves were plotted using a non-linear regression model using GraphPad Prism 5.0 software and IC calculated50The value is obtained. See table a for experimental results.
Table a results of experiments on the activity of the compounds of the present invention on inhibition of cell proliferation
Figure BDA0002915655280000271
Remarking: lenvatinib refers to Lenvatinib, and the structure is
Figure BDA0002915655280000272
The experimental results show that: the compound has a remarkable inhibition effect on cell proliferation and has unexpected excellent antitumor activity.
Example B pharmacokinetic evaluation of the Compounds of the invention after intravenous injection or gavage in rats and dogs
The present invention evaluates the pharmacokinetic studies of the compounds of the invention in rats and/or dogs for details in the animals shown in table 1.
Table 1 information sheet of the subject animals of the present invention
Figure BDA0002915655280000273
Test method
The compounds of the invention were administered to the animals in the form of 10% DMSO + 10% Kolliphor HS15+ 78% Saline + 2% (2% HCl) solution or 78% Saline + 2% (2% HCl) + 20% PEG400 solution, and the animals were fasted for 12h before administration and allowed free access to water. For the group administered by intravenous injection, the dose was 1mg/kg (rat) or 0.5mg/kg (dog), and blood was taken intravenously (blood volume taken was about 0.2mL) at the following time points after administration: 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24h (dog) or 0.083, 0.25, 0.5, 1.0, 2.0, 5.0, 7.0 and 24h (rat), EDTA-K is added into the blood sampling tube in advance2Can be used as anticoagulant. For the gavage group, 5mg/kg (rat) or 2.5mg/kg (dog) was administered, and intravenous blood sampling (blood volume taken about 0.2mL) was performed at the following time points after administration: 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24h (dog) or 0.25, 0.5, 1.0, 2.0, 5.0, 7.0 and 24h (rat), EDTA-K is added into the blood sampling tube in advance2Can be used as anticoagulant. Blood samples were centrifuged at 12,000rpm for 2 minutes, plasma was collected and stored at-20 ℃ or-70 ℃.
The plasma samples collected above were processed (frozen plasma was thawed at room temperature, vortexed for 15s, 10-20. mu.L of plasma was taken, 120-150. mu.L of acetonitrile solution containing the internal standard was added, vortexed for 5min, centrifuged at 4,000rpm for 5min, 100. mu.L of supernatant was taken, and 120-150. mu.L of methanol/water (v/v. 1/1) was added and mixed) and then the concentration of the compounds in the plasma was analyzed by LC/MS/MS.
The analysis result shows that the compound of the invention has better pharmacokinetic property in rats and/or dogs. The compound of the invention has good drug property and good clinical application prospect.
In the description herein, references to the description of the term "one embodiment," "an embodiment," "some embodiments," "an example," "a specific example" or "some examples" or the like are intended to 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, a schematic representation of the above terms does not necessarily refer to the same embodiment, implementation, or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments, implementations, or examples. Furthermore, the various examples, embodiments, or examples described in this specification, as well as features of various examples, embodiments, or examples, may be combined and combined by one skilled in the art without contradiction.
Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that variations, modifications, substitutions and alterations can be made to the above embodiments by those of ordinary skill in the art within the scope of the present invention.

Claims (10)

1.A compound which is a compound represented by formula (I) or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt, or a prodrug thereof,
Figure FDA0002915655270000011
wherein:
r' is
Figure FDA0002915655270000012
V is-NH-, -O-or-S-;
x is CRxOr N;
y is CRyOr N;
z is CH or N;
R1、R2a、R2b、R3and RxEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy), -S (═ O)2C1-C6Alkyl radical, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino or hydroxy substituted C1-C6An alkyl group;
R4、R5、R6and RyEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C6Alkyl), -C (═ O) - (C)1-C6Alkoxy), -S (═ O)2C1-C6Alkyl radical, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C1-C6Alkylthio radical, C1-C6Alkylamino or hydroxy substituted C1-C6An alkyl group;
R7and R8Each independently is H, D, -OH, C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C3-6Cycloalkyl, heterocyclic group consisting of 3 to 8 atoms, C6-10Aryl, heteroaryl of 5 to 10 atoms, C1-C6Alkoxy radical C1-6Alkylene radical, C3-6Cycloalkyl radical C1-6Alkylene, (heterocyclic group consisting of 3 to 8 atoms) C1-6Alkylene radical, C6-10Aryl radical C1-6Alkylene or (heteroaryl of 5 to 10 atoms) C1-6An alkylene group; or R7、R8And the nitrogen atom to which they are attached form a heterocyclic group of 3 to 8 atoms;
R9is C1-C6Alkyl radical, C2-C6Alkenyl radical, C2-C6Alkynyl, C1-C6Haloalkyl, C3-6Cycloalkyl, heterocyclic group consisting of 3 to 8 atoms, C6-10Aryl, heteroaryl of 5 to 10 atoms, C1-C6Alkoxy radical C1-6Alkylene radical, C3-6Cycloalkyl radical C1-6Alkylene, (heterocyclic group consisting of 3 to 8 atoms) C1-6Alkylene radical, C6-10Aryl radical C1-6Alkylene or (heteroaryl of 5 to 10 atoms) C1-6An alkylene group.
2. The compound of claim 1, wherein R1、R2a、R2b、R3And RxEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy), -S (═ O)2C1-C4Alkyl radical, C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C4Alkylamino or hydroxy substituted C1-C4An alkyl group;
R4、R5、R6and RyEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-(C1-C4Alkyl), -C (═ O) - (C)1-C4Alkoxy), -S (═ O)2C1-C4Alkyl radical, C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C1-C4Alkylthio radical, C1-C4Alkylamino or hydroxy substituted C1-C4An alkyl group.
3. A compound according to claim 1 or 2, wherein R1、R2a、R2b、R3And RxEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3、-S(=O)2CH3Methyl, ethyl, n-propyl, isopropyl, tert-butyl, vinyl, -CHF2、-CF3Methoxy, -OCF3Methylamino, dimethylamino, or hydroxymethyl;
R4、R5、R6and RyEach independently is H, D, F, Cl, Br, I, -CN, -NO2、-NH2、-OH、-SH、-COOH、-C(=O)NH2、-C(=O)NHCH3、-C(=O)N(CH3)2、-C(=O)-CH3、-C(=O)-OCH3、-S(=O)2CH3Methyl, ethyl, n-propyl, isopropyl, tert-butyl, vinyl, -CHF2、-CF3Methoxy, -OCF3Methylamino, dimethylamino or hydroxymethyl.
4. The compound of claim 1, wherein R7And R8Each independently is H, D, -OH, C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C1-C4Alkoxy radical, C1-C4Haloalkoxy, C3-6Cycloalkyl, heterocyclic group consisting of 3 to 6 atoms, C6-10Aryl, heteroaryl of 5 to 6 atoms, C1-C4Alkoxy radical C1-4Alkylene radical, C3-6Cycloalkyl radical C1-4Alkylene, (heterocyclic group consisting of 3 to 6 atoms) C1-4Alkylene radical, C6-10Aryl radical C1-4Alkylene or (heteroaryl of 5 to 6 atoms) C1-4An alkylene group; or R7、R8And the nitrogen atom to which they are attached form a heterocyclic group of 3 to 6 atoms;
R9is C1-C4Alkyl radical, C2-C4Alkenyl radical, C2-C4Alkynyl, C1-C4Haloalkyl, C3-6Cycloalkyl, heterocyclic group consisting of 3 to 6 atoms, C6-10Aryl, heteroaryl of 5 to 6 atoms, C1-C4Alkoxy radical C1-4Alkylene radical, C3-6Cycloalkyl radical C1-4Alkylene, (heterocyclic group consisting of 3 to 6 atoms) C1-4Alkylene radical, C6-10Aryl radical C1-4Alkylene or (heteroaryl of 5 to 6 atoms) C1-4An alkylene group.
5. A compound according to claim 1 or 4, wherein R7And R8Each independently H, D, -OH, methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, 2-methylpropyl, 1-methylpropyl, ethenyl, propenyl, allyl, ethynyl, propynyl, propargyl, -CHF2、-CF3Methoxy, ethoxy, isopropoxy, -OCF3Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, thienyl, thiazolyl, oxazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, methoxyimino, morpholinyl, pyrrolyl, oxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, thienyl, and the likeMethyl, methoxyethylene, C3-6Cycloalkyl methylene, C3-6Cycloalkylethylene radical, C3-6Cycloalkylpropylene, (heterocyclyl) methylene of 3-6 atoms, (heterocyclyl) ethylene of 3-6 atoms, phenylmethylene, phenylethylene, phenylpropylene, pyridylmethylene, pyrimidylmethylene, pyrrolylmethylene, pyrazolyl methylene, triazolylmethylene or tetrazolylmethylene; or R7、R8And the nitrogen atom to which they are attached form an azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, or morpholinyl group;
R9is methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, 2-methylpropyl, 1-methylpropyl, ethenyl, propenyl, allyl, ethynyl, propynyl, propargyl, -CHF2、-CF3Cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, azetidinyl, pyrrolidinyl, tetrahydrofuryl, piperidinyl, piperazinyl, morpholinyl, phenyl, indenyl, naphthyl, pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, furanyl, thienyl, thiazolyl, oxazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, methoxymethylene, methoxyethylene, C3-6Cycloalkyl methylene, C3-6Cycloalkylethylene radical, C3-6Cycloalkylpropylene, (heterocyclyl) methylene of 3-6 atoms, (heterocyclyl) ethylene of 3-6 atoms, phenylmethylene, phenylethylene, phenylpropylene, pyridylmethylene, pyrimidylmethylene, pyrrolylmethylene, pyrazolyl methylene, triazolylmethylene or tetrazolylmethylene.
6. The compound of claim 1, which is a compound having one of the following structures or a stereoisomer, a geometric isomer, a tautomer, a nitrogen oxide, a hydrate, a solvate, a metabolite, a pharmaceutically acceptable salt, or a prodrug thereof of a compound having one of the following structures:
Figure FDA0002915655270000031
7. a pharmaceutical composition comprising a compound of any one of claims 1-6; and
the pharmaceutical composition optionally further comprises a pharmaceutically acceptable excipient, carrier, adjuvant, or any combination thereof.
8. Use of a compound according to any one of claims 1 to 6 or a pharmaceutical composition according to claim 7 for the manufacture of a medicament for the prophylaxis or treatment of a tumour.
9. The use according to claim 8, wherein the tumor is a solid tumor, a B-cell lymphoma and/or a hematological and immune system tumor.
10. The use according to claim 9, wherein,
wherein the solid tumor is gastric cancer, intestinal cancer, liver cancer, kidney cancer, lung cancer, brain cancer, laryngeal cancer, nasopharyngeal cancer, esophageal cancer, gallbladder cancer, head and neck cancer, squamous carcinoma, cancer of the lymphatic system, thyroid cancer, bladder cancer, ovarian cancer, cervical cancer, endometrial cancer, testicular cancer, prostate cancer, genitourinary tract cancer, breast cancer, small cell lung cancer, lung adenocarcinoma, pancreatic cancer, colon cancer, rectal cancer, glioblastoma and/or monocytic leukemia;
wherein the B cell lymphoma is diffuse large B cell lymphoma, small lymphocytic tumor, mantle cell lymphoma, multiple myeloma and/or chronic lymphocytic leukemia;
wherein the blood and immune system tumor is acute lymphatic leukemia, chronic lymphatic leukemia, acute myelogenous leukemia and/or chronic myelogenous leukemia.
CN202110100504.0A 2021-01-26 2021-01-26 Galactose-containing nitrogen-containing aromatic ring derivative and use thereof Active CN112920240B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110100504.0A CN112920240B (en) 2021-01-26 2021-01-26 Galactose-containing nitrogen-containing aromatic ring derivative and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110100504.0A CN112920240B (en) 2021-01-26 2021-01-26 Galactose-containing nitrogen-containing aromatic ring derivative and use thereof

Publications (2)

Publication Number Publication Date
CN112920240A true CN112920240A (en) 2021-06-08
CN112920240B CN112920240B (en) 2022-04-29

Family

ID=76165929

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110100504.0A Active CN112920240B (en) 2021-01-26 2021-01-26 Galactose-containing nitrogen-containing aromatic ring derivative and use thereof

Country Status (1)

Country Link
CN (1) CN112920240B (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6143764A (en) * 1995-11-07 2000-11-07 Kirin Beer Kabushiki Kaisha Quinoline and quinazoline derivatives inhibiting platelet-derived growth factor receptor autophosphorylation and pharmaceutical compositions containing the same
WO2010123591A2 (en) * 2009-01-09 2010-10-28 The Uab Research Foundation Small molecule inhibitors of nads, namnat, and nmnat
CN108530455A (en) * 2017-03-01 2018-09-14 北京赛特明强医药科技有限公司 Aromatic ring Lian dioxanes simultaneously quinazoline compounds or pharmaceutical salts or hydrate and the application as tyrosine kinase inhibitor of urea substitution

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6143764A (en) * 1995-11-07 2000-11-07 Kirin Beer Kabushiki Kaisha Quinoline and quinazoline derivatives inhibiting platelet-derived growth factor receptor autophosphorylation and pharmaceutical compositions containing the same
WO2010123591A2 (en) * 2009-01-09 2010-10-28 The Uab Research Foundation Small molecule inhibitors of nads, namnat, and nmnat
CN108530455A (en) * 2017-03-01 2018-09-14 北京赛特明强医药科技有限公司 Aromatic ring Lian dioxanes simultaneously quinazoline compounds or pharmaceutical salts or hydrate and the application as tyrosine kinase inhibitor of urea substitution

Also Published As

Publication number Publication date
CN112920240B (en) 2022-04-29

Similar Documents

Publication Publication Date Title
JP6139782B2 (en) Substituted pyrazolopyrimidine compounds, and pharmaceutically acceptable salts thereof, and solvates, stereoisomers, and tautomers thereof, and pharmaceutical compositions containing them
RU2719428C2 (en) Indazole compounds as fgfr kinase inhibitors, preparation and use thereof
EP2947086B1 (en) Novel fused pyrimidine compound or salt thereof
US10874670B2 (en) Substituted fused heteroaromatic compounds as kinase inhibitors and the use thereof
JP2019509990A (en) 3,5-disubstituted pyrazoles useful as checkpoint kinase 1 (CHK1) inhibitors, and their preparation and use
EP3556761B1 (en) Pyrrolo-aromatic heterocyclic compound, preparation method therefor, and medical use thereof
AU2020342189A1 (en) 3, 5-disubstituted pyrazole compounds as kinase inhibitors and uses thereof
EP3475285B1 (en) Mechanistic target of rapamycin signaling pathway inhibitors and therapeutic applications thereof
CN112920240B (en) Galactose-containing nitrogen-containing aromatic ring derivative and use thereof
CN112898360B (en) Nitrogen-containing aromatic ring derivative containing glucose and application thereof
JP7329052B2 (en) Fluorine-containing substituted benzothiophene compounds and pharmaceutical compositions and applications thereof
CN111848572B (en) Amide compound and preparation method and application thereof
CN112250666B (en) Substituted pyrimidines and their use
US20090202571A1 (en) Bioreductively-activated prodrugs
EP3875452A1 (en) Monoacylglycerol lipase inhibitors
CN114026097B (en) Substituted pyrazoloquinazolinone compounds and uses thereof
CN116655638B (en) Deuterated PRMT5 inhibitors
JPWO2018159613A1 (en) Antitumor effect enhancer using pyrazolo [3,4-d] pyrimidine compound
WO2022258043A1 (en) Gemcitabine anticancer derivative and anticancer pharmaceutical use
RU2801306C2 (en) New phosphate ether compound having a pyrrolopyrimidine skeleton, or a pharmaceutically acceptable salt thereof
KR20220015433A (en) A novel carbonate compound having a pyrrolopyrimidine skeleton or a pharmaceutically acceptable salt thereof
AU2022306467A1 (en) Compound of the 7a,8,9,10,11,11a-hexahydro-1h,7h-pyrano[2,3-c]xanthene type, method of preparation thereof, intermediates thereof and therapeutic applications thereof
CN117222649A (en) Pyrrolopyrimidine derivatives, process for their preparation and their use
TW202322805A (en) Heterocyclic compounds with cyclin-dependent kinase inhibition activity, a preparation method and medical use thereof
EP4365175A1 (en) Novel benzopyran derivative and use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
CP01 Change in the name or title of a patent holder
CP01 Change in the name or title of a patent holder

Address after: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Patentee after: Guangdong Dongyangguang Pharmaceutical Co.,Ltd.

Address before: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Patentee before: SUNSHINE LAKE PHARMA Co.,Ltd.