WO2010139180A1 - 作为蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘酰胺衍生物、其制备方法及应用 - Google Patents

作为蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘酰胺衍生物、其制备方法及应用 Download PDF

Info

Publication number
WO2010139180A1
WO2010139180A1 PCT/CN2010/000272 CN2010000272W WO2010139180A1 WO 2010139180 A1 WO2010139180 A1 WO 2010139180A1 CN 2010000272 W CN2010000272 W CN 2010000272W WO 2010139180 A1 WO2010139180 A1 WO 2010139180A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
diseases
hydrogen
trifluoromethyl
halogen
Prior art date
Application number
PCT/CN2010/000272
Other languages
English (en)
French (fr)
Other versions
WO2010139180A8 (zh
Inventor
鲁先平
李志斌
山松
余金迪
宁志强
Original Assignee
深圳微芯生物科技有限责任公司
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
Priority to CA2763822A priority Critical patent/CA2763822C/en
Priority to RU2011152113/04A priority patent/RU2497809C2/ru
Application filed by 深圳微芯生物科技有限责任公司 filed Critical 深圳微芯生物科技有限责任公司
Priority to DK10782884.0T priority patent/DK2439195T3/da
Priority to SI201030695T priority patent/SI2439195T1/sl
Priority to MX2011012752A priority patent/MX2011012752A/es
Priority to PL10782884T priority patent/PL2439195T3/pl
Priority to ES10782884.0T priority patent/ES2509615T3/es
Priority to KR1020117030659A priority patent/KR101421786B1/ko
Priority to BRPI1011994A priority patent/BRPI1011994B8/pt
Priority to JP2012513451A priority patent/JP5484568B2/ja
Priority to EP10782884.0A priority patent/EP2439195B1/en
Priority to UAA201115118A priority patent/UA103092C2/ru
Priority to AU2010256246A priority patent/AU2010256246B9/en
Priority to KR1020147000095A priority patent/KR20140014313A/ko
Publication of WO2010139180A1 publication Critical patent/WO2010139180A1/zh
Priority to ZA2011/09030A priority patent/ZA201109030B/en
Publication of WO2010139180A8 publication Critical patent/WO2010139180A8/zh
Priority to HRP20140717AT priority patent/HRP20140717T1/hr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/88Oxygen atoms
    • C07D239/90Oxygen atoms with acyclic radicals attached in position 2 or 3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • 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
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • C07D215/233Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/24Oxygen atoms attached in position 8
    • C07D215/26Alcohols; Ethers thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/24Oxygen atoms attached in position 8
    • C07D215/26Alcohols; Ethers thereof
    • C07D215/28Alcohols; Ethers thereof with halogen atoms or nitro radicals in positions 5, 6 or 7
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • Naphthylamide derivatives as protein kinase inhibitors and histone deacetylase inhibitors
  • the present invention relates to the synthesis, preparation and treatment of a naphthylamide derivative having both protein kinase inhibitory activity and histone deacetylase inhibitory activity in the treatment of diseases associated with abnormal protein kinase activity and abnormal histone deacetylase activity Clinical application.
  • Protein kinases are a class of enzymes that catalyze the phosphorylation of proteins, particularly phosphorylation of hydroxyl groups on specific tyrosine, serine and threonine residues in proteins. Protein kinases play a key role in regulating many cellular physiological processes, including metabolism, cell proliferation, cell differentiation, cell survival, environmental-host response, immune response, and angiogenesis. Many diseases are associated with abnormal cellular responses triggered by protein kinase regulation. These diseases include inflammation, autoimmune diseases, cancer, nervous system diseases and degenerative diseases, cardiovascular diseases, metabolic diseases, allergies, asthma and hormone-related diseases (Tan, SL., 2006, J. Immunol., 176: 2872-2879; Healy, A.
  • Protein kinases are generally divided into two classes, protein tyrosine kinases (PTKs) and serine-threonine kinases (STKs). Protein tyrosine kinases (PTKs) can be divided into two classes, non-transmembrane tyrosine kinases and transmembrane growth factor receptor tyrosine kinases (RTKs). At present, at least 19 different subfamilies of RTKs have been identified, such as epidermal growth factor receptor (EGFR), vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR) and fibroblast growth. Factor receptor (FGFR).
  • EGFR epidermal growth factor receptor
  • VEGFR vascular endothelial growth factor receptor
  • PDGFR platelet-derived growth factor receptor
  • FGFR fibroblast growth. Factor receptor
  • the epidermal growth factor receptor (EGFR) family comprises four transmembrane tyrosine kinase growth factor receptors, namely HER1, HER2, HER3 and HER4.
  • EGFR epidermal growth factor receptor
  • HER1, HER2, HER3 and HER4 transmembrane tyrosine kinase growth factor receptors
  • HER1, HER2, HER3 and HER4 transmembrane tyrosine kinase growth factor receptors
  • HER1, HER2, HER3 and HER4 transmembrane kinase growth factor receptors
  • VEGF vascular endothelial growth factor
  • EGFR overexpression is very common in non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • the activity of EGFR can be inhibited by blocking the extracellular ligand binding domain using an anti-EGFR antibody or by inhibiting the EGFR tyrosine kinase using a small molecule, thereby achieving the purpose of inhibiting downstream components of the EGFR pathway (Mendelsohn, ⁇ , 1997) , Clin. Can. Res., 3: 2707-2707).
  • VEGF vascular endothelial growth factor
  • VEGFR-2 Ligand specific receptors on endothelial cells binds.
  • VEGF-A binds to VEGFR-1 to cause migration of endothelial cells
  • binding to VEGFR-2 causes endothelial cell proliferation, invasion and survival
  • VEGFR-3 is thought to regulate lymphangiogenesis.
  • VEGF and VEGFR Binding of the -2 receptor results in activation and autophosphorylation of the intracellular tyrosine kinase domain and further triggers the generation of a series of signals within the cell (Parikh, AA., 2004, Hematol. Oncol. Clin. N. Am. , 18:951 -971 ) o
  • STKs Serine-threonine kinases
  • STKs are mainly present in cells. There are also a few receptor kinases of the STKs type. STKs are the most common form of cytosolic kinases, which perform their functions at the cytoplasmic site, rather than the organelles and cytoskeleton in the cytoplasm.
  • Glycogen synthase kinase-3 (GS-3) is a serine-threonine protein kinase comprising two isoforms, alpha and beta, each encoded by a unique gene. GSK-3 has been found to phosphorylate many regulatory proteins and regulate their activity. GSK-3 is associated with a variety of diseases, including diabetes, Alzheimer's disease, central nervous system disorders such as manic depressive disorder and neurodegenerative diseases, cardiac hypertrophy, etc. (Haq, et al., 2000, J. Cell Biol., 151: 117 ).
  • Aurora-2 is a serine-threonine protein kinase that is closely related to human cancers such as colon cancer, breast cancer and other solid tumors. It is thought to be involved in phosphorylation of proteins that regulate cell cycle. In particular, Aurora-2 plays a role in controlling the accurate separation of chromosomes during mitosis. Abnormal regulation of the cell cycle can lead to cell proliferation and other abnormalities.
  • Aurom-2 protein has been found to be overexpressed (Schumacher, et al., 1998, J. Cell Biol., 143: 1635-1646; Kimura et al., 1997, J. Biol. Chem. , 272: 13766-13771 )
  • CDKs Cell cycle-dependent kinases
  • CDK l-1 nine kinase subunits
  • CDK l-2 nine kinase subunits
  • Uncontrolled proliferation is a hallmark of cancer cells, and abnormal regulation of CDK function often occurs in many important solid tumors. People are particularly interested in CDK2 and CDK4 because their activity is often abnormally regulated in many human tumors.
  • Raf kinase is a downstream effector protein of tumor proteins. It is a key regulator of the signal transduction pathway from the cell surface to the nucleus. Inhibition of Raf kinase is closely related to inhibition of the growth of many human tumors in vivo and in vitro (Monia et al., 1996, Nat. Med., 2: 668-675).
  • serine-threonine protein kinases include protein kinases A, B and C. These kinases (ie, P A, PKB, and P C) play key roles in signal transduction pathways.
  • One of the objects of the present invention is to disclose a class of naphthamide derivatives having a protein kinase selective inhibitory activity and a histone acetylase inhibiting activity;
  • the second object of the present invention is to disclose a process for the preparation of the compounds described in this class
  • a third object of the present invention is to disclose the use of such a compound as a clinical application for the treatment of diseases associated with abnormalities in protein kinase activity and abnormal histone deacetylase activity.
  • Histone deacetylase (HDAC) proteins play a key role in regulating gene expression in vivo, altering the accessibility of transcription factors to genomic DNA.
  • HDAC removes the acetyl group of acetylated lysine residues in histones, resulting in nucleosome remodeling (Grunstein, M., 1997, Nature, 389: 349-352). Since HDAC proteins play a key role in gene expression, they are closely related to many cellular functions, including cell cycle regulation, cell proliferation, differentiation, gene program expression, and cancerogenesis (Ruijter, AJM, 2003, Biochem.
  • HDAC inhibitors can inhibit tumor growth in humans and animals, including lung cancer, gastric cancer, breast cancer, prostate cancer, and lymphoma (Dokmanovic, M., 2005, J. Cell Biochenm., 96: 293-304 ).
  • Mammalian HDACs can be classified into three classes based on sequence homology.
  • the first class consists of yeast Rpd3-like proteins (HDAC 1 , 2, 3, 8 and 11).
  • the second class consists of yeast HDA proteins (HDAC 4, 5, 6, 7, 9 and 10).
  • the third category consists of yeast SIR2-like proteins (SIRT 1, 2, 3, 4, 5, 6 and 7).
  • HDAC 1 The activity of HDAC 1 is associated with cell proliferation (a hallmark of cancer). Mammalian cells reduce HDAC1 expression by siRNA and have antiproliferative properties (Glaser, KB., 2003, Biochem. Biophys. Res. Comm., 310: 529-536). HDAC 1 knockout mice are embryonic lethal, resulting in changes in stem cell growth rate (Lagger, G, 2002, EMBO J., 21 : 2672-268. DcHDAC1 overexpressing mouse cells show 0 2 and M phase prolongation and growth The rate is reduced (Bartl. S., 1997, Mol. Cell Biol., 17: 5033-5043). Therefore, experimental data indicate that HDAC1 is closely related to cell cycle regulation and cell proliferation.
  • HDAC2 regulates the expression of many fetal cardiac protein isoforms. Lack of HDAC2 or chemical inhibition of histone deacetylase can prevent re-expression of embryonic genes and reduce ventricular hypertrophy. Anti-hypertrophy is associated with increased expression of the inositol polyphosphate-5-phosphatase f (Inpp5f)-encoding gene, which increases the proto-oncogene (Akt) and 3-phosphoinositol-dependent protein of thymoma virus kinase-1 inactivation activate glycogen synthase kinase 3 ⁇ (Gsk3p) 0 in contrast, transgenic mice of HDAC2 ventricular hypertrophy increases, which is related to inactivated Gsk3p.
  • Inpp5f inositol polyphosphate-5-phosphatase f
  • Akt proto-oncogene
  • Gsk3p glycogen synthase kinase 3 ⁇
  • HDAC2 is an important molecular target of HDAC inhibitors in the heart.
  • Both HDAC2 and Gsk3P are part of the regulatory pathway, which provides an attractive therapeutic target for the treatment of ventricular hypertrophy and heart failure (Trivedi, CM., 2007, Nat. Med,. 13: 324-331).
  • HDAC3 is most expressed in proliferating crypt cells of the normal small intestine. Silencing of HDAC3 expression in colon cancer cell lines results in inhibition of cell growth, decreased cell survival, and increased apoptosis.
  • HDAC3 gene silencing also selectively causes expression of alkaline phosphatase (a marker of colon cell maturation). Overexpression of HDAC3 inhibits basal transcription and butyrate-induced P21 transcription, while silencing HDAC3 stimulates the activity and expression of the P21 gene promoter.
  • HDAC6 is a subtype of the HDAC family that removes the acetyl group of alpha-tubulin and increases cell motility. Quantitative real-time reverse transcription polymerase chain reaction (PCR) and Western Blots analysis were performed on nine groups of oral squamous cell carcinoma (OSCC) cell lines and normal oral keratinocytes (NOKs), compared to NOKs, HDAC6 mR A and protein. The level of expression is elevated in all cancer cells. HDAC6 protein was detected in the cytoplasm of OSCC cell lines by immunofluorescence analysis. Similar to the OSCC cell line, HDAC6 is up-regulated in early human OSCC tumors with up to 74% mRNA and 51% protein.
  • PCR quantitative real-time reverse transcription polymerase chain reaction
  • NOKs normal oral keratinocytes
  • HDAC makes functional chromosome epigene silencing one of the main mechanisms of many pathological processes.
  • the function-related genes are inhibited or re-regulated by HDAC, resulting in phenotypic loss in terminal differentiation, maturation and growth control, and loss of tissue function.
  • tumor suppressor genes are often silenced during the development of cancer, and HDAC inhibitors are able to induce expression of these tumor suppressor genes, thereby inhibiting tumor cell growth and differentiation (Glaros S et al., 2007, Oncogene June 4 Epub ahead of print Mai, A, et al., 2007, Int J. Biochem Cell Bio., April 4, Epub ahead of print; Vincent A.
  • HDAC inhibitors overhabit the HDAC "hot spot" program on chromosome 6p21-22, inducing expression of the entire MHC II family of genes, further extending epigenetic regulation of immune recognition and immune response (Gialitakis M et al., 2007, Nucleic Acids Res., 34(l); 765-72).
  • HDAC inhibitors include: 1) short chain fatty acids such as butyric acid and phenylbutyric acid; 2) organic hydroxamic acids such as suberoylanilide hydroxamic acid (SAHA) and trichostatin A (TSA); 2-amino-8-oxo-9, 10-epoxydecanoyl (AOE) ring tetrapeptides such as trapoxin and HC-toxin; 4) 2-amino-8-oxo-9, 10-epoxy-free Cyclodecyl quinones, such as apicidin and FK228; 5) Benzamides, such as MS-275 (European Patent EP 0847992A1, US Patent 2002/0103192A1, World Patent 02/26696A1, World Patent 01/70675A2, World Patent 01/ 18171A2).
  • MS-275 European Patent EP 0847992A1, US Patent 2002/0103192A1, World Patent 02/26696A1, World Patent 01/70675A2, World Patent 01/ 18171A
  • HDAC is a promising drug target
  • the current SAHA developed by Merck is limited to the treatment of cutaneous T-cell lymphoma, but not for solid tumors. Therefore, it is necessary to continue to develop new compounds that have stronger HDAC inhibitory activity, stronger anticancer activity, better HDAC subselectivity and lower toxicity.
  • Targeted therapy has been highly regarded by anti-cancer drug developers. People want to design drugs that can reach the tumor accurately A specific target of the cell kills the tumor cell and at the same time does not harm the normal cell.
  • tumor cells are capable of growing and spreading using a variety of bioinitiators and pathways. Tumor cells are struck at a target that reorganizes and redeploys along new growth pathways. As a result, combined targeted therapies have been developed and are emerging as new paradigms for cancer treatment.
  • Several multi-target kinase inhibitors are currently under development, with Sorafenib and Sutent being approved for marketing in the United States.
  • Sorafenib (developed by Bayer) is the first drug to target both the RAF/ME/ERK pathway (associated with cell proliferation) and the VEGFR2/PDGFRP cascade (associated with angiogenesis). Approved for the treatment of advanced kidney cancer. Although these targeted therapies are effective in the treatment of some solid tumors, they are not ideal and have toxic side effects when treating other solid tumors.
  • the compound of the present invention binds to the anti-angiogenic and anti-proliferative activity of an RTK inhibitor and the activity of the HDAC inhibitor to induce differentiation, immunomodulation, hinder cell cycle, and promote apoptosis, and is intended for solid tumors. It has better curative effect while overcoming the toxic side effects of commercially available RTK inhibitors, such as hypertension, prolonged QT interval, thyroid degeneration, rash and skin discoloration, pain and the like.
  • Z is CH or N
  • R 2 and R 3 are each independently hydrogen, halogen, fluorenyl, decyloxy or trifluoromethyl;
  • X is a benzene ring or a pyridine ring
  • R 5 is one or more substituents selected from hydrogen, halogen, decyl, decyloxy or trifluoromethyl;
  • the compound is as shown in formula (I), wherein
  • R' R 2 and R 3 are each independently hydrogen, halogen, alkyl, decyloxy or trifluoromethyl;
  • X is a benzene ring or a pyridine ring
  • R 5 is one or more substituents selected from hydrogen, halogen, decyl, decyloxy or trifluoromethyl. More preferably, the compound is as shown in formula (I), wherein
  • R', R 2 and R 3 are each hydrogen or a decyloxy group
  • X is a benzene ring or a pyridine ring
  • R 5 is one or more substituents selected from hydrogen, halogen, alkyl, alkoxy or trifluoromethyl. More preferably, the compound is as shown in the formula (I),
  • R 1 , R 2 are each hydrogen or methoxy
  • R 3 is H
  • X is a benzene ring or a pyridine ring
  • R 5 is one or more substituents selected from hydrogen, halogen, decyl, decyloxy or trifluoromethyl. Optimally, Z is CH;
  • R' R 2 is hydrogen or methoxy, respectively
  • R 3 is H
  • X is a benzene ring or a pyridine ring
  • halogen described in the present invention is fluorine, chlorine, bromine or iodine
  • the "mercapto group" according to the present invention includes a linear, branched or cyclic fluorenyl group such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, ortho. Pentyl, isopentyl, n-hexyl, isohexyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, etc.;
  • nonyloxy group refers to a group formed by linking a sulfhydryl group to an oxygen atom, wherein the oxygen atom has a free bonding ability such as a methoxy group, an ethoxy group, a propoxy group, and a butoxy group.
  • Base pentyloxy, isopropoxy, tert-butoxy, cyclopropoxy, cyclohexyloxy, etc.;
  • the preparation method of the compound of the present invention is as follows:
  • the compound (ii) is condensed with the compound ⁇ ) to obtain the target compound (1).
  • the condensation reaction uses a peptide condensing agent as a catalyst, such as 1-ethyl-3-G-dimethylaminopropyl carbodiimide (EDC), N, N '-" dicyclohexylcarbodiimide (DCC). , ⁇ , ⁇ '-carbonyldiimidazole (CDI), etc.
  • EDC 1-ethyl-3-G-dimethylaminopropyl carbodiimide
  • DCC N, N '-" dicyclohexylcarbodiimide
  • CDI ⁇ , ⁇ '-carbonyldiimidazole
  • the solvent used in the reaction is a usual solvent such as benzene, toluene, tetrahydrofuran, dioxane, dichloromethane, chloroform, N,N-dimethylformamide or the like.
  • a base such as sodium hydroxide, triethylamine or pyridine may also be added as necessary.
  • Compound (II) can be produced by the following method:
  • the commercially available compound (V) is subjected to condensation reaction with the compound (VI) to obtain a compound ( ⁇ ).
  • the condensation reaction uses a peptide condensing agent as a catalyst, such as 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC), hydrazine, ⁇ '-dicyclohexylcarbodiimide (DCC). , ⁇ , ⁇ '-carbonyldiimidazole (CDI), etc.
  • EDC 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide
  • DCC ⁇ '-dicyclohexylcarbodiimide
  • CDI ⁇ , ⁇ '-carbonyldiimidazole
  • the solvent used in the reaction is a usual solvent such as benzene, toluene, tetrahydrofuran, dioxane, dichloromethane, chloroform, hydrazine, hydrazine-dimethylformamide or the like.
  • a base such as sodium hydroxide, triethylamine or pyridine may also be added as necessary.
  • the compound (VII) is dissolved in methanol, and catalytically hydrogenated using 5% palladium on carbon as a catalyst to obtain a compound (IIIa).
  • the reaction was carried out at room temperature.
  • An acid such as sulfuric acid may be added to the reaction system as necessary.
  • the compound of the formula (I) can be purified by a usual separation method such as extraction, recrystallization, column chromatography and the like.
  • the compound of the formula (I) has both protein kinase inhibitory activity and histone deacetylase inhibitory activity, and can be used for treating diseases associated with abnormal protein kinase activity and abnormal histone deacetylase activity, particularly for blood cancer. And solid tumors have excellent efficacy.
  • the compound of the formula (I) is processed into conventional pharmaceutical preparations such as tablets, capsules, powders, syrups, solutions, suspensions, injections, and ointments.
  • the preparation contains, as an active ingredient, a compound of the formula (I), and a pharmaceutically acceptable carrier, adjuvant or diluent.
  • the preparation usually contains 0.5 to 70% of the active ingredient, preferably 1 to 20%.
  • the pharmaceutically acceptable carrier, excipient or diluent of the present invention includes the Handbook of Pharmaceutical Excipients (American Pharmaceutical Association, October 1986) or the Handbook of Pharmaceutical, published by the Chemical Industry Press. Excipients, original fourth edition) listed carrier fillers, but are not limited to these carrier fillers.
  • the compound of the formula (I) according to the present invention can be administered clinically to mammals (including humans) by oral or injection, and is particularly preferably administered orally.
  • the dosage is 0.0001 ⁇ 200 mg/kg body weight per day, and the preferred dosage is 0.01 ⁇ 100 mg/kg body weight per day.
  • the optimal dosage is 0.1 ⁇ 50 mg/kg body weight per day. At the same time, the optimal dose is the individual. However, usually the dose is small at the beginning and then gradually increased.
  • Fig. 1 Inhibitory activity of compound 31 on transplanted tumor of human A549 lung cancer in nude mice, wherein Vehicle represents a vector, Sutent represents the existing drug sunitinib, and comp31 represents compound 31.
  • Fig. 2 Inhibitory activity of compound 31 on human HCT-8 intestinal cancer transplanted tumor in nude mice, wherein Vehicle represents a vector, Sutent represents the existing drug sunitinib, and comp31 represents compound 31.
  • Fig. 3 Inhibitory activity of compound 31 on transplanted tumor of human SMMC-7721 liver cancer nude mice, wherein Vehicle represents a vector, Sutent represents the existing drug sunitinib, and comp31 represents compound 31.
  • Figure 4 Inhibitory activity of compounds 33 and 34 on human HCT-8 intestinal cancer xenograft tumors, wherein Vehicle represents the vector, Sutent represents the existing drug sunitinib, comp33 represents compound 33, and comp34 represents compound 34.
  • Fig. 5 Inhibitory activities of compounds 33 and 37 against human HCT-8 intestinal cancer xenograft tumors, wherein Vehicle represents a vector, Sutent represents the existing drug sunitinib, comp33 represents compound 33, and comp37 represents compound 37.
  • Fig. 6 Inhibitory activities of compounds 33 and 37 on human SMMC-7721 liver cancer transplanted tumor in nude mice, wherein Vehicle represents a vector, Sutent represents the existing drug sunitinib, comp33 represents compound 33, and comp37 represents compound 37.
  • N-(2-Aminophenyl)-4-cyanobenzamide (237 mg, 1 mmol) was dissolved in 40 mL methanol then sulfuric acid (196 mg, 1 mmol) and 5% palladium carbon (0.20 g) . The mixture was stirred with hydrogen until the reaction was completed. The reaction solution was filtered through celite, and then filtered and evaporated. filter. The filtrate was concentrated in vacuo to give a white solid, 4-(aminomethyl)-N-(2-aminophenyl)benzamide (232 mg, yield 96%). LC-MS (m/z) 242 (M+1).
  • the amide 29.0 mg, 0.12 mmol was used as a starting material to give a brown solid (2-aminophenyl)-6-((2-(6,7-dimethoxyquinazoline).
  • 4-oxo) amide 1-naphthyl) methyl) nicotinamide (41.4 mg, yield 69%) c LC-MS ( m / z) 601 (m + 1) 0
  • 6-(7-methoxyquinoline 4-oxo)-1-naphthoic acid (34.5 mg, 0.1 mmol) and o-phenylenediamine (43.2 mg, 0.4 mmol) were used as a starting material in a similar procedure as in Example 16.
  • N-(2-Aminophenyl)-6-(7-methoxyquinoline-4-oxo)-1-naphthylamide (39.6 mg, yield 91%) was obtained as a brown solid.
  • 6-(7-methoxyquinoline ⁇ 4-oxo)-1-naphthoic acid (34.5 mg, 0.1 mmol) and 4-fluoro-o-phenylenediamine (15.1 mg, 0.12 mmol) were used as the starting material.
  • a similar procedure in the preparation of the brown solid N-(2-amino-4-fluorophenyl)-6-(7-methoxyquinoline-4-oxo)-1-naphthalenamide (33.1 mg, yield 73%) .
  • 6-(6,7-Dimethoxyquinoline-4-oxo)-1-naphthoic acid (37.5 mg, 0.1 mmol) and 4-(aminomethyl)-N-(2-aminophenyl)benzamide (28.9 mg, 0.12 mmol) was used as a starting material to give a brown solid N-(4-((2-aminophenyl)carbamoyl)benzyl)-6-(6,7- Dimethoxyquinoline ⁇ 4-oxo-1-naphthylamide (49.0 mg, yield 82%).
  • a brown solid was obtained by the similar procedure of Example 16 using 6-(quinoline ⁇ 4-oxo)-1-naphthoic acid (31.5 mg, 0.1 mmol) and o-phenylenediamine (43.2 mg, 0.4 mmol).
  • Microcrystalline cellulose 90 g
  • Talc powder 0.5 g
  • Preparation process Compound 31 is passed through a 100 mesh sieve, microcrystalline cellulose, sodium carboxymethyl starch and talc powder are passed through an 80 mesh sieve, and the prescribed amount of microcrystalline cellulose and sodium carboxymethyl starch are weighed and mixed uniformly. Compound 31 was mixed with an equal amount of addition method, and added with 4% povidone K30 absolute ethanol solution to prepare granules. After drying, a prescribed amount of talc powder was added and mixed, and the tablets were obtained.
  • Microcrystalline Cellulose - Lactose - Sodium Carboxymethyl Starch Microcrystalline Cellulose - Lactose - Sodium Carboxymethyl Starch:
  • Preparation process Compound 31 is passed through a 100 mesh sieve, and microcrystalline cellulose, lactose, sodium carboxymethyl starch and magnesium stearate are passed through an 80 mesh sieve to weigh the prescribed amount of microcrystalline cellulose, lactose and carboxymethyl starch. Sodium, mix well, then mix compound 31 with the same amount of addition method, add the prescription amount of magnesium stearate, mix and fill the capsule.
  • Mouse NIH-3T3 fibroblast engineered cell lines stably expressing human PDGFRP were used to evaluate PDGF-dependent cell proliferation.
  • PDGFR "NIH-3T3 cells were seeded in 96-well microtiter plates at a density of 5000 per well, and cultured overnight in serum-free medium after 24 hours. Add the test compound and PDGF BB (50 ng/ml), continue in serum-free The medium was cultured for 72 hours. The cell proliferation was measured by the MTS method (Promega) according to the instructions. The cells were cultured in a CO 2 incubator at 37 ° C for 2 hours, and then the absorbance at 490 nm was measured on an ELISA plate reader.
  • HUVEC cells were seeded in 96-well microplates at a density of 6000 per well, and cultured in serum-free medium for 2 hours after 24 hours.
  • the test compound and VEGF 165 50 ng/ml were added and incubation was continued for 72 hours in serum-free medium.
  • Cell proliferation was measured by the MTS method (Promega) according to the instructions. The cells were incubated at 37 ° C for 2 hours in a CO 2 incubator, and then the absorbance at 490 nm was measured on an ELISA plate reader.
  • the Discovery Kit tested the inhibitory activity of the inhibitor in vitro.
  • the principle of the experiment is as follows: A special substrate / «or cfe will remove an acetyl group under the action of histone deacetylase (the nuclear extract of HeLa cells, HDAC rich in various subtypes). Exposure of free amino groups, when added to Developei, produces an inducible fluorescence. This fluorescence has an excitation wavelength of 360 nm and an emission wavelength of 460 nm. The more deacetylated the substrate, the higher the induced fluorescence value.
  • the fluorescence value in the absence of inhibitor is the control; and in the presence of the inhibitor, the induced fluorescence value is reduced, and the fluorescence value in the absence of the enzyme (corresponding to the complete inhibition of the enzyme activity) is blank. Generally, the fluorescence value after suppression will be between the control and the blank. In the analysis, we take the blank as 0, and the control as 1 to calculate. The smaller the value, the higher the inhibitory activity.
  • Fluorescence values were read on a fluorescent microplate reader with an excitation wavelength of 369 nm and an emission wavelength of 451 nm.
  • HDAC human fluorescent protein
  • a regulatory element that selects the appropriate transcription factor to construct a reporter gene can be used to assess the selective inhibition of the HDAC subtype by the inhibitor.
  • HeLa cells were seeded into 96-well plates one day prior to transfection to achieve a 50-80% density fusion during transfection.
  • the reporter plasmid inserting the p21-promoter regulatory sequence upstream of the luciferase gene was separately transfected with FuGe ne 6 ( Roche ) reagent, according to Instructions for transfection.
  • FuGe ne 6 Roche
  • the expression plasmid of green fluorescent protein (GFP) was co-transfected.
  • DMSO Compound or solvent control
  • CS055 Sidabenamine, a HDAC inhibitor developed by Shenzhen Microcore Biotechnology Co., Ltd., has excellent antitumor activity and is undergoing clinical phase 2 trials.
  • the tumor cells were trypsinized, they were seeded in 96-well microplates at a density of 3000 per well, in 10% FBS. Incubate in complete medium for 24 hours. The test compound and solvent control were added, and the final compound concentration was 100 nmol/L to 100 ⁇ /L. Then, it was cultured in complete medium for 72 hours. The method of addition of MTS reagent instructions (Promega), incubated at 37 ° C C0 2 incubator for 2 hours, and then absorbance was read at 490nm on an ELISA plate reader.
  • MTS reagent instructions Promega
  • CS055 Sidabenamine, a HDAC inhibitor developed by Shenzhen Microcore Biotechnology Co., Ltd., has excellent antitumor activity and is undergoing clinical phase 2 trials.
  • Compound 31 was used to inhibit the transplanted tumor of human A549 lung cancer in nude mice. Nude mice, females, 14-16 g were used. After normal feeding for 3 days, if there was no abnormality, 50 patients were inoculated with 50 underarms. After lung cancer rearing observation, it was confirmed that the tumor cells were implanted and the tumor mass was larger than 6 mm, and the rats were randomly divided into 8 groups/groups, a total of 6 groups, among which 4 drug groups were administered in four groups, and the solvent control group was a positive control. Drug Sutent - group. Oral administration was given once a day, 24 times a day, and Compound 31 was administered at doses of 5, 10, 20 and 40 mg/kg body weight.
  • Compound 31 inhibited human HCT-8 intestinal cancer transplanted tumor in nude mice.
  • Nude mice, females, 18-20 g were used. After normal feeding for 3 days, if there was no abnormality, 50 patients underwent conventional HCT-8 colon cancer inoculation. After rear-feeding observation, it was confirmed that the tumor cells were implanted and the tumor mass was not less than 100 mm 3 , and the group was divided into 8 groups/groups, a total of 6 groups, wherein the test drug was administered in four groups, and the solvent control group was positive. Control drug Sutent-group. The same day oral medication, 1 time/day, a total of 24 times, the compound 31 was administered at doses of 2.5, 5, 10 and 20 mg/kg body weight.
  • Nude mice, females, 18-20 g were selected. After normal feeding for 3 days, if there was no abnormality, 50 patients were randomly vaccinated with SSMC7721 liver cancer and observed, and the tumor cells were implanted and the tumor volume was not less than 100 mm 3 . Grouped, 8 rats/group, a total of 6 groups, of which 4 drug concentrations were tested in four groups, one group of solvent control, and the positive control drug Sutent-group. Oral administration was given once a day, 24 times a day, and Compound 31 was administered at doses of 2.5, 5, 10 and 20 mg/kg body weight. Animal body weight was weighed twice a week and tumor volume was measured. After 24 times of administration, the animals were sacrificed the next day and the tumor was weighed. According to the formula:
  • Sutent 40 21.0 ⁇ 0.8 24.8 ⁇ 1.2 1.00 ⁇ 0.68 70.3 ⁇ 0.001 Compound 31 20 20.2 ⁇ 1.7 21.0 ⁇ 2.2 0.53 ⁇ 0.28 84.4 ⁇ 0.001 Compound 31 10 20.4 ⁇ 1.6 23.6 ⁇ 1.5 0.70 ⁇ 0.45 79.2 ⁇ 0.001 Compound 31 5 20.8 ⁇ 1.2 24.8 ⁇ 1.5 1.16 ⁇ 0.55 65.4 ⁇ 0.001 Compound 31 2.5 20.1 ⁇ 0.9 23.2 ⁇ 2.1 1.63 ⁇ 0.70 51.7 ⁇ 0.001 a group of 8 nude mice. b tumor growth inhibition rate.
  • Compound 33, 34 inhibited human HCT-8 colon cancer transplanted tumor in nude mice.
  • Nude mice, females, 18-20 g were used. After normal feeding for 3 days, if there was no abnormality, 50 patients were inoculated with HCT-8 under normal armpits. After intestinal cancer feeding observation, it was confirmed that the tumor cells were implanted and the tumor mass was not less than 100 mm 3 , and were randomly divided into 8 groups/groups, a total of 6 groups, of which the test drug compounds 33 and 34 were administered in two groups. The solvent control group and the positive control drug Sutent group. Oral administration on the same day, 1 time / day, a total of 20 times. Animal body weight was weighed twice a week and tumor volume was measured.
  • Compound 33, 37 inhibited human HCT-8 colon cancer xenografts in nude mice. Nude mice, females, 18-20 g were used. After normal feeding for 3 days, if there was no abnormality, 50 patients were inoculated with HCT-8 under normal armpits. After intestinal cancer feeding observation, it was confirmed that the tumor cells were implanted and the tumor mass was not less than 100 mm 3 , and were randomly divided into 8 groups/groups, a total of 6 groups, among which the drug concentration of each of the test drug compounds 33 and 37 was four groups. The solvent control group and the positive control drug Sutent group. Oral administration on the same day, compound 33 two dose groups 2 times / day, interval 6 hours, the other groups 1 time / day, a total of 20 days.
  • Compound 33, 37 inhibited human SSMC7721 liver cancer xenografts in nude mice. Nude mice, females, 18-20 g were observed. After normal feeding for 3 days, if there was no abnormality, 50 patients were randomly inoculated with human SSMC7721 liver cancer. When the tumor cells were implanted and the tumor mass was not less than 100 mm 3 , the rats were randomly divided into 8 groups/groups, 6 groups in total. Among them, the test drug compounds 33 and 37 were administered in two groups, and the solvent control group was used. Positive control drug Sutent-group. Oral administration on the same day, once a day, 30 times in total. Animal body weight was weighed twice a week and tumor volume was measured.

Description

作为蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘酰胺衍生物
其制备方法及应用 技术领域
本发明涉及同时具有蛋白激酶抑制活性和组蛋白去乙酰化酶抑制活性的萘酰胺衍生物的 合成、 制备及其在治疗与蛋白激酶活性异常以及组蛋白去乙酰化酶活性异常相关的疾病方面 的临床应用。
背景技术
蛋白激酶是催化蛋白磷酸化的一类酶, 特别是催化蛋白中特定的酪氨酸、 丝氨酸和苏氨 酸残基上的羟基的磷酸化。蛋白激酶在调节许多细胞生理过程中都起到关键作用, 包括代谢、 细胞增殖、 细胞分化、 细胞存活、 环境 -主体反应、 免疫应答和血管生成。 许多疾病都与由蛋 白激酶调节所引发的异常的细胞反应有关。 这些疾病包括炎症、 自身免疫性疾病、 癌症、 神 经***疾病和祌经退化性疾病、心血管疾病、代谢病、过敏、哮喘以及与激素相关疾病(Tan, S-L.,2006, J. Immunol., 176: 2872-2879; Healy, A. ea al.,2006, J. Immunol., 177: 1886-1893; Salek-Ardakani, S. et al.,2005, J. Immunol., 175: 7635-7641 ; Kim, J. et al.,2004 J. Clin. Invest, 114: 823-827)。 因此, 人们一直致力于寻求能够有效地治疗这些疾病的蛋白 激酶抑制剂。
蛋白激酶通常分为两类, 即蛋白酪氨酸激酶 (PTKs) 和丝氨酸 -苏氨酸激酶 (STKs)。 蛋白酪氨酸激酶 (PTKs) 可分为两类, 即非跨膜酪氨酸激酶和跨膜生长因子受体酪氨酸 激酶(RTKs)。目前,至少己经确定了 RTKs的 19种不同的亚族,如表皮生长因子受体(EGFR), 血管内皮生长因子受体 (VEGFR), 血小板源生长因子受体 (PDGFR) 和纤维母细胞生长因 子受体 (FGFR)。
表皮生长因子受体 (EGFR) 家族包含了四种跨膜酪氨酸激酶生长因子受体, 即 HER1、 HER2、 HER3和 HER4。 当一组特定的配体与受体结合后, 促进了 EGFR的二聚, 并且导致 受体中酪氨酸残基的自磷酸化 (Arteaga, C-L.,2001 , Curr. Opin. Oncol., 6: 491498)。 一旦 受体自磷酸化后, EGFR转导通路的的几个下游信号就被激活了。 EGFR的信号转导通路与肿 瘤生成的过程密切相关, 包括细胞循环进程、 细胞凋亡的抑制、 肿瘤细胞的运动、 入侵和转 移。 EGFR的激活也刺激了血管内皮生长因子 (VEGF), 它是血管生成的主要诱导体 (Petit, A-M. et al.,1997, Am. J. Pathol., 151 : 1523-1530)。 在实验模型中, EGFR介导的信号转导 通路的下调与肿瘤的发生密切相关 (Wikstrand, C-J. et al.,1998, J Natl Cancer Inst, 90: 799-800)。在许多人体肿瘤中可以观察到突变导致的 EGFR蛋白的连续扩增激活和过度表达, 括胸部、 肺部、 卵巢和肾脏的肿瘤。 这些突变是肿瘤侵润的决定因素 (Wikstrand, C-J. et al., 1998, J Natl Cancer Inst, 90: 799-800 ) o EGFR过度表达在非小细胞肺癌 (NSCLC ) 中 是非常常见的。 EGFR的活性可以通过使用抗 -EGFR抗体进行阻断胞外配体结合域或通过使 用小分子抑制 EGFR 酪氨酸激酶来进行抑制, 从而达到抑制 EGFR通路下游组分的目的 ( Mendelsohn,丄, 1997, Clin. Can. Res., 3: 2707-2707)。
几乎所有的实体瘤和间质瘤在低氧情况下都会分泌血管内皮生长因子 (VEGF)。 它对血 管内皮是高度专一的, 并且对血管增殖和渗透都能起到调节作用。 VEGF 水平的过度表达与 微脉管密度的增加、癌症复发和存活率降低均密切相关(Parikh, A-A., 2004;, Hematol. Oncol. Clin. Ν· Am., 18:951-971 Parikh, A-A., 2004;, Hematol. Oncol. Clin. N. 18:951 -971 0 对 VEGF受体来说有 6种不同的配体: VEGF-A到 E和胎盘生长因子。 配体与内皮细胞上的 特定受体 (主要是 VEGFR-2 ) 相结合。 VEGF-A与 VEGFR-1的结合引起内皮细胞的迁移, 与 VEGFR-2的结合引起内皮细胞增殖、 渗透和存活, VEGFR-3被认为调节淋巴血管生成。 VEGF与 VEGFR-2受体的结合导致了细胞内酪氨酸激酶区域的活化和自磷酸化,并进一步触 发了细胞内一连串信号的产生 (Parikh, A-A., 2004, Hematol. Oncol. Clin. N. Am. , 18:951 -971 ) o
丝氨酸 -苏氨酸激酶(STKs)主要存在于细胞内。也存在少数几种 STKs类型的受体激酶。 STKs是胞液激酶最常见的形式,它们在细胞质部位执行它们的功能, 而不是细胞质中的细胞 器和细胞骨架。
糖原合成酶激酶 -3 (GS -3 ) 是一种丝氨酸 -苏氨酸蛋白激酶, 包含了 α和 β两种异构形 式, 这两种异构形式各自以独特的基因编码。 人们发现 GSK-3能够使许多调控蛋白磷酸化, 并可以调节它们的活性。 GSK-3与多种疾病相关, 包括糖尿病、 老年痴呆症、 中枢神经*** 障碍如狂躁抑郁障碍和神经退化疾病、心肌肥大等(Haq, et al., 2000, J. Cell Biol., 151 : 117)。
Aurora-2 是一种丝氨酸 -苏氨酸蛋白激酶, 它与人体癌症密切相关, 如结肠癌、 ***癌和 其它实体瘤。人们认为它与调节细胞循环的蛋白磷酸化有关。尤其,有丝***过程中, Aurora-2 在控制染色体的准确分离上起了作用。 细胞周期的异常调节可以导致细胞增殖和其它异常。 在人体结肠癌组织中, 人们发现 Aurom-2蛋白被过度表达(Schumacher, et al., 1998, J. Cell Biol. , 143: 1635-1646; Kimura et al., 1997, J. Biol. Chem., 272: 13766-13771 )„
细胞周期依赖性激酶 (CDKs) 是一种丝氨酸 -苏氨酸蛋白激酶, 它调节哺乳动物的细胞 ***。 目前, 人们已经确定了九个激酶亚基 (CDK l-9)。 每一个激酶都和一个特定的调节对 象相结合, 一起构成了活性催化位点。 增殖失控是癌细胞的一个特点, 在许多重要的实体瘤 中, 常常发生 CDK功能的异常调节。 人们对 CDK2和 CDK4尤其感兴趣, 因为在许多人体 肿瘤中, 它们的活动常常被异常地调节。
Raf激酶是一种 肿瘤蛋白的下游效应蛋白。它是由细胞表面到细胞核的信号转导通路 的一个关键调节器。抑制 Raf激酶与在体内和体外抑制许多人体肿瘤的生长密切相关(Monia et al.,1996, Nat. Med., 2: 668-675)。
其它的丝氨酸-苏氨酸蛋白激酶包括蛋白激酶 A、 B和 C。 这些激酶 (即 P A, PKB和 P C) 在信号转导通路中起着关键作用。
人们一直致力于寻求具有蛋白激酶抑制活性并能治疗与蛋白激酶活性异常相关疾病的小 分子化合物。 文献报道的化合物有环状化合物 (美国专利 US 7,151 ,096)、 双环化合物 (美 国专利 US 7, 189,721 )、 三环化合物 (美国专利 US 7, 132,533)、 (2-羟基吲哚基 -3-亚甲基) 乙酸衍生物 (美国专利 US 7,214,700)、 3- (4-酰胺吡咯基 -2-基亚甲基) -2-吲哚满酮衍生物 (美国专利 US 7, 179,910)、稠合吡唑衍生物(美国专利 US7,166,597)、胺基呋咱化合物(美 国专利 US 7, 157,476)、 吡咯取代的的 2-吲哚满酮化合物 (美国专利 US 7,125,905)、 *** 化合物(美国专利 US 7, 115,739)、吡唑基胺基取代的喹唑啉化合物(美国专利 US 7,098,330 ) 和吲唑化合物 (美国专利 US 7,041 ,687) 等。 有几个蛋白激酶抑制剂已被 FDA批准用于癌 症治疗, 如 Glivec、 Sutent和 Sorafenib。 临床使用表明, 与传统的化疗相比, 这些药物优势 明显。 由此激发人们基于机理对治疗方法进行改进, 优化化合物分子骨架, 以期发现具有更 好口服生物利用度、 更高的抗癌活性和更低毒性的新化合物。
发明内容
本发明目的之一在于公开一类具有蛋白激酶选择性抑制活性和同时具有组蛋白考乙酰化 酶抑制活性的萘酰胺衍生物;
本发明目的之二在于公开这一类所述的化合物的制备方法;
本发明目的之三在于公开这一类所述的化合物作为治疗与蛋白激酶活性异常以及组蛋白 去乙酰化酶活性异常相关的疾病方面的临床应用。
组蛋白去乙酰化酶 (HDAC ) 蛋白在调控体内的基因表达方面起着关键作用, 它改变转 录因子到染色体组 DNA的可达性。尤其是 HDAC去除组蛋白中乙酰化赖氨酸残基的乙酰基, 从而导致核小体重构 (Grunstein, M., 1997, Nature, 389: 349-352)。 由于 HDAC蛋白在基因表 达中起着关键作用, 所以它们与许多细胞功能密切相关, 包括细胞周期调控、 细胞增殖、 分 化、 基因程序化表达和癌症发生(Ruijter, A-J-M, 2003, Biochem. J., 370: 737-749; Grignani, R, 1998, Nature, 391 : 815-818; Lin, R-J.,1998, 391 : 811-814; Marks, P-A., 2001, Nature Reviews Cancer, l : 194)o 由组蛋白去乙酰化酶误调所致的异常去乙酰化与许多临床疾病密切相关, 如 鲁-泰(Rubinstein-Taybi )二氏综合征、 脆性 X染色体综合症、 神经退化性疾病、 心血管与代 谢疾病、类风湿、 白血病和其它各种癌症 ( Langley B et al., 2005, Current Drug Targets - CNS & Neurological Disorders, 4: 41-50)。 试验表明, HDAC抑制剂能够抑制人类及动物体内的肿瘤 的生长, 包括肺癌、 胃癌、 乳腺癌、 ***癌和淋巴瘤等 (Dokmanovic, M.,2005, J. Cell Biochenm., 96: 293-304)。
依据序列同源性, 哺乳动物的 HDACs 可以分为三类。 第一类由酵母 Rpd3-类蛋白组成 ( HDAC 1 , 2, 3, 8和 11 )。 第二类由酵母 HDA 类蛋白组成(HDAC 4, 5, 6, 7, 9和 10)。 第三 类由酵母 SIR2-类蛋白组成 ( SIRT 1, 2, 3, 4, 5, 6和 7 )。
HDAC 1的活性和细胞增殖(癌症的标志)相关。 哺乳动物细胞通过 siRNA降低 HDAC1 表达,具有抗增殖性(Glaser, K-B., 2003, Biochem. Biophys. Res. Comm., 310: 529-536)。 HDAC 1 基因敲除鼠是胚胎致死的,其结果导致干细胞生长速度的改变(Lagger, G, 2002, EMBO J., 21 : 2672-268 DcHDACl过度表达的老鼠细胞显示 02和 M期延长及生长速度降低(Bartl. S., 1997, Mol. Cell Biol., 17: 5033-5043 )。 因此, 试验数据表明, HDAC1与细胞周期调控及细胞增殖密 切相关。
HDAC2调控许多胎心肌蛋白异构体的表达。 HDAC2缺乏或是通过化学方法抑制组蛋白 去乙酰化酶可以阻止胚胎基因的再表达, 减少心室肥大。 抗肥大和肌醇多磷酸盐 -5-磷酸酶 f ( Inpp5f) 编码基因表达增加有关, 这种增加导致胸腺瘤滤过性病毒的原癌基因 (Akt) 和 3- 磷酸肌醇-依赖的蛋白激酶 -1失活, 激活了糖原合成酶激酶 3β ( Gsk3p ) 0 相反, HDAC2转基 因鼠心室肥大增加,这与失活的 Gsk3p有关。通过化学方法抑制活化的 Gsk3p使得 HDAC2- 缺乏的成人对心室肥大的刺激变得敏感。 这些结果表明, 在心脏中 HDAC2是 HDAC抑制剂 的一个重要的分子靶标。 HDAC2和 Gsk3P都是调控通路的组成部分, 这为治疗心室肥大和 心力衰竭提供了极具吸引力的治疗靶标 (Trivedi, C-M., 2007, Nat. Med,. 13: 324-331 )。 HDAC3在正常小肠的增殖隐窝细胞中表达最多。在结肠癌细胞系中 HDAC3表达沉默导 致细胞生长抑制、 细胞存活降低和细胞凋亡增加。 沉默 HDAC2表达可以观察到类似的效果, 而对于 HDAC1 , 效果则没有那么显著。 HDAC3基因沉默也选择性的引起碱性磷酸酯酶的表 达 (结肠细胞成熟的标志)。 HDAC3的过度表达抑制基础转录及丁酸酯诱导的 P21转录, 而 沉默 HDAC3则刺激 P21基因启动子的活性和表达。 这些发现表明 HDAC3是在人体结肠癌 中进行下调的基因, 是结肠细胞成熟和 P21表达的一种新型的调节剂 (Wilson, A-J., 2006, J. Biol. Chem., 281 : 13548-13558 )。
HDAC6是 HDAC家族的一个亚型, 它脱去 α-微管蛋白的乙酰基, 增加细胞运动性。 在 九组口腔鳞状细胞癌 (OSCC) 的细胞系和正常的口腔角化细胞 (NOKs) 上运用定量实时逆 转录聚合酶链反应和 Western Blots技术分析, 与 NOKs相比, HDAC6 mR A和蛋白表达水 平在所有的癌细胞中都升高了。 通过免疫荧光法分析, 在 OSCC细胞系的细胞质中检测到了 HDAC6蛋白。和 OSCC细胞系相似, 在早期的人体 OSCC肿瘤中 HDAC6上调明显, mRNA 达 74%, 蛋白达 51 %。 通过对临床变量的分析, 人们发现临床肿瘤的发展阶段和 HDAC6的 表达状态相关。 分析表明, HDAC6的表达水平在肿瘤早期(I和 II期)和晚期(III和 IV期) 存在显著差异 (P=0.014)。 这些结果表明 HDAC6 的表达可能和肿瘤的恶性程度有关, 这也 为设计新的治疗方法提供了线索 (Sakuma^ T., 2006, Int. J. Oncol., 29: 117-124)。
HDAC使功能染色体表观基因沉默是许多病理过程的主要机制之一。 其中, 功能相关基 因被 HDAC抑制或重调,导致在终末分化、成熟和生长控制中表型缺失,并且丧失组织功能。 例如, 肿瘤抑制基因经常在癌症的发展过程中被沉默, HDAC抑制剂能够诱导这些肿瘤抑制 基因的表达, 从而抑制肿瘤细胞生长和分化(Glaros S et al., 2007, Oncogene June 4 Epub ahead of print; Mai, A, et al., 2007, Int J. Biochem Cell Bio., April 4, Epub ahead of print; Vincent A. et al., 2007, Oncogene, April 30, Epub ahead of print; our unpublished results )。 结构基因 (如与 Friedreich's运动失调相关的 FXN基因和与脊柱肌肉萎缩相关的 SMN基因) 的抑制可以被 HDAC抑制剂逆转,使得 FXN和 SMN基因重新表达,并恢复组织功能(Herman D et al., 2006, Nature Chemical Biology, 2(10):551-8; Avila AM et al., 2007, J Clinic Investigation, 117(3)659-71; de Bore J, 2006, Tissue Eng. 12(10):2927-37)o HDAC抑制剂在染色体 6p21-22重调 HDAC "热 点"程序, 诱导整个 MHC II家族基因表达, 进一步延伸了免疫识别和免疫响应的表观基因 调控 (Gialitakis M et al., 2007, Nucleic Acids Res., 34(l);765-72)。
目前已经确定了几类 HDAC抑制剂, 包括: 1 ) 短链脂肪酸, 如丁酸和苯丁酸; 2) 有机 羟肟酸, 如 suberoylanilide hydroxamic acid (SAHA)和 trichostatin A (TSA); 3)含有 2-胺基 -8-氧 -9, 10-环氧癸酰基 (AOE) 的环四肽, 如 trapoxin和 HC-toxin; 4) 不含 2-氨基 -8-氧 -9, 10-环氧癸酰基的环四肽, 如 apicidin 和 FK228; 5) 苯酰胺类, 如 MS-275 (欧洲专利 EP 0847992A1 , 美国专利 2002/0103192A1 , 世界专利 02/26696A1 , 世界专利 01/70675A2, 世 界专利 01/18171A2 )。 尽管 HDAC是一个极具前景的药物靶标, 但目前 Merck公司研制的 SAHA仅仅局限于对皮肤 T细胞淋巴瘤的治疗, 而对实体瘤疗效并不明显。 因此, 有必要继 续研制新的化合物, 使其具有更强的 HDAC抑制活性、 更强的抗癌活性、 更好的 HDAC亚 型选择性和更低的毒性。
靶向治疗一直为抗癌药物研发人员所推崇。 人们希望设计药物, 它既能准确的到达肿瘤 细胞的某个特定靶标并杀死肿瘤细胞, 同时又对正常细胞又没有伤害。 然而, 肿瘤细胞能够 运用多种生物引发器和通路来进行生长和传播。 在一个靶点对肿瘤细胞进行打击, 它们会沿 着新的生长通路重组和重新部署。 由此人们发展了组合靶向治疗, 并正在成为癌症治疗的新 范例。几个多靶点激酶抑制剂现正在研发中, 其中 Sorafenib和 Sutent己经在美国获得批准上 市。 Sorafenib (Bayer公司开发) 是第一个同时以 RAF/ME /ERK通路 (与细胞增殖有关) 和 VEGFR2/PDGFRP级联信号通路 (与血管生成有关) 为靶点的药物, 该药在 2005年 12月 获得批准用于治疗晚期肾癌。 尽管这些靶向治疗药物在治疗一些实体瘤时是有效的, 但是在 治疗其它实体瘤时, 疗效并不理想且具有毒副作用。
本发明所述的化合物, 结合了 RTK抑制剂的抗血管生成和抗增殖活性以及 HDAC抑制 剂的所具有的诱导分化、 免疫调节、 阻碍细胞周期、 促使细胞凋亡的活性, 旨在对实体瘤具 有更好的疗效, 同时克服市售的 RTK抑制剂的毒副作用, 如高血压、 QT间期延长、 甲状腺 退化、 皮疹和皮肤变色、 疼痛等。
本发明所述的化合物, 其化学结构如通式 (I ) 所示:
Figure imgf000007_0001
其中,
Z为 CH或 N;
R' . R2和 R3 分别为氢、 卤素、 垸基、 垸氧基或三氟甲基;
R4
Figure imgf000007_0002
X为苯环或吡啶环;
R5为一个或多个取代基, 选自氢、 卤素、 垸基、 垸氧基或三氟甲基;
包括其游离形式、 盐的形式、 对映异构体、 非对映异构体或水合物。
较优地, 所述化合物如式 (I) 所示, 其中,
Z为 CH;
R' R2和 R3 分别为氢、 卤素、 烷基、 垸氧基或三氟甲基;
R4
Figure imgf000008_0001
X为苯环或吡啶环;
R5为一个或多个取代基, 选自氢、 卤素、 垸基、 垸氧基或三氟甲基。 再优地, 所述化合物如式 (I) 所示, 其中,
Z为 CH;
R'、 R2和 R3 分别为氢或垸氧基;
R4
Figure imgf000008_0002
X为苯环或吡啶环;
R5为一个或多个取代基, 选自氢、 卤素、 烷基、 烷氧基或三氟甲基。 更优地, 所述化合物如式 (I) 所示, 中,
Z为 CH;
R1 , R2 分别为氢或甲氧基;
R3为 H;
R4
Figure imgf000008_0003
X为苯环或吡啶环;
R5为一个或多个取代基, 选自氢、 卤素、 垸基、 垸氧基或三氟甲基。 最优地, Z为 CH;
R' R2 分别为氢或甲氧基;
R3为 H;
R4
Figure imgf000008_0004
X为苯环或吡啶环;
R5为 1^或?。
本发明所述的 "卤素", 为氟、 氯、 溴、 碘;
本发明所述的 "垸基", 包括直链、 支链或环状垸基, 如甲基、 乙基、 正丙基、 异丙基、 正丁基、 异丁基、 特丁基、 正戊基、 异戊基、 正己基、 异己基、 环丙基、 环丁基、 环戊基、 环己基等;
本发明所述的 "垸氧基", 是指垸基与氧原子相连所形成的基团, 其中, 氧原子具有自由 成键能力, 如甲氧基、 乙氧基、 丙氧基、 丁氧基、 戊氧基、 异丙氧基、 特丁氧基、 环丙氧基、 环己基氧基等;
本发明所述的化合物的制备方法如下:
Figure imgf000009_0001
( II ) ( III ) ( I ) 将化合物(ii)与化合物 π)进行缩合反应得到目标化合物 (1)。 该缩合反应以肽缩合 剂为催化剂,如 1-乙基 -3- G-二甲胺丙基)碳二亚胺(EDC),N, N '-」二环己基碳二亚胺(DCC), Ν, Ν'-碳酰二咪唑 (CDI) 等。 反应温度为 0〜80QC, 反应时间为 4- 72小时。 反应所用溶剂为 常用溶剂, 如苯、 甲苯、 四氢呋喃、 二氧六环、 二氯甲垸、 氯仿、 N,N-二甲基甲酰胺等。 必 要时, 也可以加入碱如氢氧化钠、 三乙胺或吡啶。
化合物 (II) 可以通过如下方法制备:
Figure imgf000009_0002
将市售的 6-羟基萘酸、 碳酸铯和具有指定取代基的 4-氯喹啉 ( IV) 于 DMSO溶剂中加 热得到萘酸 (11)。 反应温度为 130~140°C, 反应时间为 3~24小时,
化合物 (III ) 可直接购得或通过如下方法制备- NC-X-COOH
Figure imgf000009_0003
( V ) (VI ) (VII )
Figure imgf000010_0001
(VII ) ( Ilia )
将市售的化合物 (V ) 与化合物 (VI) 进行缩合反应得到化合物 (νπ)。 该缩合反应以 肽缩合剂为催化剂, 如 1-乙基 -3- (3-二甲胺丙基)碳二亚胺 (EDC), Ν, Ν '-二环己基碳二亚 胺 (DCC ), Ν, Ν'-碳酰二咪唑 (CDI) 等。 反应温度为 0~60°C, 反应时间为 2~72小时。 反 应所用溶剂为常用溶剂, 如苯、 甲苯、 四氢呋喃、 二氧六环、 二氯甲烷、 氯仿、 Ν, Ν-二甲基 甲酰胺等。 必要时, 也可以加入碱如氢氧化钠、 三乙胺或吡啶。
将化合物 (VII) 溶于甲醇, 以 5%钯碳为催化剂, 催化氢化得到化合物 (IIIa)。 该反应 在室温进行。 必要时, 可在反应体系中加入酸, 如硫酸。
通式(I)所述的化合物可以采用常见的分离方法进行纯化, 如萃取、重结晶、柱层析等。 通式 (I)所述的化合物同时具有蛋白激酶抑制活性和组蛋白去乙酰化酶抑制活性, 可以 用于治疗与蛋白激酶活性异常以及组蛋白去乙酰化酶活性异常相关的疾病, 尤其对血癌和实 体瘤具有优异疗效。
通式 (I) 所述的化合物被加工成常用的药用制剂, 如片剂、 胶囊、 粉剂、 糖浆、 液剂、 悬浮剂、 针剂、 膏剂。 该制剂含有作为活性组分的通式 (I) 的化合物, 以及药用载体、 辅料 或稀释剂。 该制剂中通常含有 0.5~70%的活性成分, 较佳含量为 1~20%。
本发明所说的药用载体、 辅料或稀释剂, 包括《药用赋形剂手册》(美国药学协会, 1986 年 10月)或化学工业出版社出版的《药用辅料手册》(Handbook of Pharmaceutical Excipients, 原著第四版) 所列的载体填料, 但并不局限于这些载体填料。
本发明所述的通式(I)化合物, 在临床上可以通过口服或注射方式对哺乳动物(包括人) 进行用药, 其中尤以口服方式最佳。 用药剂量为每日 0.0001~200 mg/kg体重, 较佳用药剂量 为每日 0.01~100 mg/kg体重, 最佳用药剂量为每日 0.1~50 mg/kg体重, 同时, 最佳剂量视个 体而定, 通常开始时剂量较小, 然后逐渐增加用量。
本发明所述的代表性化合物如表 1所示。 化合物编号与实施例部分中的 "实施例编号" 相一致, 即表 1中化合物 1的合成在"实施例 1 "中得到描述,表 1中化合物 44的合成在"实 施例 44" 中得到描述。
表 1 本发明代表性化合物
,7- ) -1 -萘酰胺
Figure imgf000010_0002
N-(2-氨基 ·4-氟苯基 )-6-
17 (6,7-二甲氧基喹唑啉 "4-氧) -1-萘 酰胺
Ν-(2-氨基 "4-甲基苯基) -6·
18 (6,7-二甲氧基喹唑啉 氧 )-1-萘 酰胺
N-(2-氨基 ·4-甲氧基苯基) -6-
19 (6,7-二甲氧基喹唑啉 4-氧) -1-萘 酰胺
N-(2-氨基斗氯苯基 )-6-
20 (6,7-二甲氧基喹唑啉 ·4·氧) -1-萘 酰胺
N-(2-氨基 "4-溴苯基 )"6·
21 (6,7-二甲氧基喹唑啉 " -氧) -1-萘 酰胺
N-(2-氨基 · -三氟甲基苯基 )-6-
22 (6,7-二甲氧基喹唑啉 ·4-氧) -1 -萘 酰胺
Figure imgf000011_0001
N-(4-((2-氨基苯基)氨基甲酰基) - 苄基) -6·(6,7-二甲氧基喹唑啉 氧) -1-萘酰胺
Figure imgf000012_0001
N-(4-«2-氨基 "4-氟苯基) - 氨基甲酰基)苄基) (6,7-二甲氧 基喹唑啉 4-氧) -1-萘酰胺
N-(2-氨基苯基) -6-((2-(6,7- 二甲氧基喹唑啉 ·4·氧 )-1-萘酰胺) 甲基)烟酰胺
Figure imgf000012_0002
N-(2-氨基 · ·氟苯基 )-6- ((2-(6,7-二甲氧基喹唑啉 " -氧) -1- 萘酰胺)甲基)烟酰胺
Ν-(3-((2-氨基苯基)氨基甲酰基) - 苄基) -6·(6,7-二甲氧基喹唑啉 氧) -1-萘酰胺
Figure imgf000012_0003
Figure imgf000013_0001
Figure imgf000014_0001
N-(2-氨基苯基) -6-(8-甲基喹啉
•4-氧) -1-萘酰胺
Figure imgf000015_0001
N-(2-氨基苯基) -6-(7-氯喹啉
•4-氧) -1-萘酰胺
N-(2-氨基苯基) -6-(8-
(三氟甲基)喹啉 "4-氧) -1 -萘酰胺
N-(4-((2-氨基苯基)氨基甲酰基) - 苄基) -6-(7-氯喹啉 ·4-氧) -1-萘酰胺
Figure imgf000015_0002
N-(4-((2-氨基苯基)氨基甲酰基) - 苄基) -6-(8- (三氟甲基)喹啉- 4-氧) -1-萘酰胺
CF3
附图说明
图 1 化合物 31对人 A549肺癌裸鼠移植性肿瘤的抑制活性,其中 Vehicle表示载体, Sutent 表示现有药物舒尼替尼, comp31表示化合物 31。
图 2 化合物 31对人 HCT-8肠癌裸鼠移植性肿瘤的抑制活性, 其中 Vehicle表示载体, Sutent表示现有药物舒尼替尼, comp31表示化合物 31。 图 3 化合物 31对人 SMMC-7721肝癌裸鼠移植性肿瘤的抑制活性,其中 Vehicle表示载 体, Sutent表示现有药物舒尼替尼, comp31表示化合物 31。
图 4 化合物 33和 34对人 HCT-8肠癌裸鼠移植性肿瘤的抑制活性,其中 Vehicle表示载 体, Sutent表示现有药物舒尼替尼, comp33表示化合物 33, comp34表示化合物 34。
图 5 化合物 33和 37对人 HCT-8肠癌裸鼠移植性肿瘤的抑制活性,其中 Vehicle表示载 体, Sutent表示现有药物舒尼替尼, comp33表示化合物 33, comp37表示化合物 37。
图 6 化合物 33和 37对人 SMMC-7721肝癌裸鼠移植性肿瘤的抑制活性, 其中 Vehicle 表示载体, Sutent表示现有药物舒尼替尼, comp33表示化合物 33, comp37表示化合物 37。 具体实施方式
下面结合实例进一步阐明本发明的内容,但本发明的保护范围并不仅仅局限于这些实例。 本发明所述的百分比除特别注明外, 均为重量百分比。 说明书中所描述的数值范围, 如计量 单位、 反应条件、 化合物物理状态或百分比, 均是为了提供明白无误的书面参考。 本领域熟 练技术人员在实践本专利时, 使用在此范围之外或有别于单个数值的温度、 浓度、 数量、 碳 原子数等, 仍然可以得到预期的结果。
实施例 1
6-(6,7-二甲氧基喹唑啉 4-氧) -1-萘酸的制备
Figure imgf000016_0001
将 6-羟基 -1-萘酸 ( 1.43 g, 7.6 mmol)溶于 38 mL DMSO, 然后加入碳酸铯 (7.5 g, 22.9 mmol) 和 4-氯 -6,7-二甲氧基-喹唑啉 (2.05 g, 9.14 mmol)。该反应液在 140°C下加热 3小时。 反应结束后冷却至室温。 加入 40 mL水稀释。 用 2N盐酸中和至 pH=6.5。 将析出的固体过 滤、 水洗、 干燥。 用甲醇重结晶得棕色固体 6-(6,7-二甲氧基喹唑啉 · -氧) -1-萘酸 (1.68 g, 产率 59% )。 LC-MS (m/z) 377 (M+1)0
实施例 2
6-(7-甲氧基喹啉 4-氧) -1-萘酸的制备
Figure imgf000016_0002
以 6-羟基 -1-萘酸 (1 ,43 g, 7.6 mmol) 和 4-氯 -7-甲氧基喹啉 (1.77 g, 9.14 mmol) 为原 料, 按照实施例 1中的类似步骤制得棕色固体 6-(7-甲氧基喹啉 4-氧) -1-萘酸 (1.73 g, 产率 66% )。 LC-MS (m/z) 346 (M+1) o
实施例 3
6-(6,7-二甲氧基喹啉 4-氧) -1-萘酸的制备
Figure imgf000017_0001
以 6-羟基 -1-萘酸 (1.43 g, 7.6 mmol) 和 4-氯- 6,7-二甲氧基喹啉 (2.04 g, 9.14 mmol)为 原料, 按照实施例 1中的类似步骤制得棕色固体 6-(6,7-二甲氧基喹啉 4-氧) -1-萘酸(1.95 g, 产率 68% )。 LC-MS (m/z) 376 (M+1) o
实施例 4
6- (喹啉 _ -氧) -1-萘酸的制备
Figure imgf000017_0002
以 6-羟基 -1-萘酸 (1.43 g, 7.6 mmol) 和 4-氯喹啉 (1.49 g, 9.14 mmol)为原料,按照实施 例 1中的类似步骤制得棕色固体 6- (喹啉 "4-氧) -1-萘酸(1.24 g,产率 52%)。 LC-MS (m/z) 316 (M+1)。
实施例 5
6-(8-甲基喹啉斗氧 )-1-萘酸的制备
Figure imgf000017_0003
以 6-羟基 -1-萘酸 (1.43 g, 7.6 mmol) 和 4-氯 -8-甲基喹啉 (1.62 g, 9.14 mmol)为原料, 按照实施例 1中的类似步骤制得棕色固体 6-(8-甲基喹啉 氧 )-1-萘酸 (1.25 g, 产率 55%)( LC-MS (m/z) 330 (M+1)。
实施例 6
6-(7-氯喹啉 -4-氧) -1-萘酸的制备
Figure imgf000017_0004
以 6-羟基 -1-萘酸 (1.43 g, 7.6 mmol) 和 4,7-二氯喹啉 (1.81 g, 9.14 mmol)为原料, 按照 实施例 1中的类似步骤制得棕色固体 6-(7-氯喹啉 ~4-氧) -1-萘酸(1.57 g,产率 59%)。 LC-MS (m/z) 350 (M+1)。 实施例 7
6-(8- (三氟甲基)喹啉 4-氧) -1-萘酸的制备
Figure imgf000018_0001
以 6-羟基 -1-萘酸 (1.43 g, 7.6 mmol) 和 4-氯 -8- (三氟甲基)喹啉 (2.12 g, 9.14 mmol)为原 料, 按照实施例 1中的类似步骤制得棕色固体 6-(8- (三氟甲基)喹啉 "4-氧) -1-萘酸(1.43 g, 产 率 49%)。 LC-MS (m/z) 384 (M+1).
实施例 8
4- (氨基甲基) -N-(2-氨基苯基)苯酰胺的制备
Figure imgf000018_0002
将 4-氰基苯甲酸 (294 mg, 2 mmol) 溶于 8 ml DMF中, 然后加入 1-乙基 -3- (3-二甲胺 丙基) 碳二亚胺盐酸盐 (768 mg, 4 mmol) 、 1-羟基苯并*** (324 mg, 2.4 mmol) 、 三乙胺 (808 mg, 8 mmol) 和邻苯二胺 (432 mg, 4 mmol)。混合物在室温下搅拌 20小时。然后用 400 ml盐水稀释。 真空抽滤收集固体。 固体用水洗涤。 真空干燥得灰色固体 N-(2-氨基苯基) -4-氰 基苯酰胺 (364 mg, 产率 77%)。 LC-MS (m/z) 238 (M+1)。
将 N-(2-氨基苯基) -4-氰基苯酰胺 (237 mg, 1 mmol) 溶于 40 mL 甲醇中, 然后加入硫酸 (196 mg, 1 mmol) 和 5%钯碳 (0.20 g)。 混合物通氢气搅拌直至反应完成。 反应液经硅藻土 过滤,滤液用 1 N NaOH溶液 (2 ml)中和。过滤。滤液真空浓缩得灰色固体 4- (氨基甲基) -N-(2- 氨基苯基)苯酰胺 (232 mg, 产率 96%)。 LC-MS (m/z) 242 (M+1)。
实施例 9
4- (氣基甲基) -N-(2-氨基 4-氟苯基)苯酰胺的制备
Figure imgf000018_0003
以 4-氰基苯甲酸 (294 mg, 2 mmol) 和 4-氟邻苯二胺 (302 mg, 2.4 mmol) 为原料, 按 照实施例 8中的类似步骤制得棕色固体 4- (氨基甲基) -N-(2-氨基 4-氟苯基)苯酰胺 (186 mg, 产 率 72%)。 LC-MS (m/z) 260 (M+1)。
实施例 10
4- (氨基甲基) -N-(2-氨基 4-甲基苯基)苯酰胺的制备
Figure imgf000018_0004
以 4-氰基苯甲酸 (294 mg, 2 mmol) 和 4-甲基邻苯二胺 (293 mg, 2.4 mmol)为原料,按 照实施例 8中的类似步骤制得灰色固体 4- (氨基甲基) -N-(2-氨基 甲基苯基)苯酰胺 (173 mg, 产率 68%)。 LC-MS (m/z) 256 (M+1)。
实施例 11
4- (氨基甲基) -N-(2-氨基 ·4-甲氧基苯基)苯酰胺的制备
Figure imgf000019_0001
以 4-氰基苯甲酸 (294 mg, 2 mmol) 和 4-甲氧基邻苯二胺 (331 mg, 2.4 mmol)为原料, 按照实施例 8中的类似步骤制得灰色固体 4- (氨基甲基) -N-(2-氨基 4-甲氧基苯基)苯酰胺 (192 mg, 产率 71 %)。 LC-MS (m/z) 272 (M+1)。
实施例 12
4- (氨基甲基) -N-(2-氨基 4-三氟甲基苯基)苯酰胺的制备
Figure imgf000019_0002
以 4-氰基苯甲酸 (294 mg, 2 mmol) 和 4-三氟甲基邻苯二胺 (422 mg, 2.4 mmol)为原 料, 按照实施例 8中的类似步骤制得灰色固体 4- (氨基甲基) -N-(2-氨基 4-三氟甲基苯基)苯酰 胺 (195 mg, 产率 63%)。 LC-MS (m/z) 310 (M+1)。
实施例 13
3- (氨基甲基) -N-(2-氨基苯基)苯酰胺的制备
Figure imgf000019_0003
以 3-氰基苯甲酸 (294 mg, 2 mmol) 和 邻苯二胺 (432 mg, 4 mmol)为原料, 按照实施 例 8中的类似步骤制得灰色固体 3- (氨基甲基) -N-(2-氨基苯基)苯酰胺 (140 mg, 产率 58%)。
LC-MS (m/z) 242 (M+1)0
实施例 14
6- (氨基甲基) -N-(2-氨基苯基)烟酰胺的制备
Figure imgf000019_0004
以 6-氰基烟酸 (296 mg, 2 mmol) 和 邻苯二胺 (864 mg, 8 mmol)为原料,按照实施例 8 中的类似步骤制得灰色固体 6- (氨基甲基) -N-(2-氨基苯基)烟酰胺 (157 mg, 产率 65%)丄 C-MS (m/z) 243 (M+1)0
实施例 15
Figure imgf000020_0001
ns#搬*¾i^蝴 w®糊 s ώ%68H(2-99(寸ζ/) (dυ/E6 - ----- - (ΗνΉ1)VHl 98PH pp)H7- p 1) 9ε(ΗVHl· p 92I·,-sJZeeeeJN =--- -- - --,· -- -,
Figure imgf000021_0001
¾郴糊 Q;9)9 (Η--
9pp) S 3 Ή)H0〇Ηε) oε95 =ss X- -. - - -.
l)(HVHXgL 9p)Z L(HV ίX20HV ΉΙH 989p) 9(.·sJNJ =NJN--- - - - -. - - - -. -. -.
6hH. -
Z.
(i) (/Ο 6) iΊHNqI 6V)(H Ή1(H) o(Hh g8VH..NE§NU9ssJJZ---- 。 , - - - - -. - - OAV 08Ϊ62/固z.io0 z
Figure imgf000022_0001
6Hv 8(H νΉΙ)8(Hv6 s(HiH SIp) n8( gp)v..sJ NJZ =---. - -.. - - - - - - Szi)ί ) S (/_Z319)ic0CszltN - -
20112)/-6UJ - δ 4.01 (s, 6H, 2 χ OCH3), 5.72 (s, 2H, benzene-NH2), 6.92 (d, J= 8.5 Hz, 1 H, Ar-H), 7.42 (s, 1 H, Ar-H), 7.59-7.65 (m, 3H, Ar-H), 7.90-7.96 (m, 2H, Ar-H), 7.98 (s, 1 H, Ar-H), 8.10 (d, J= 8.3 Hz, 1 H, Ar-H), 8.17 (d, J= 7.3 Hz, 1 H, Ar-H), 8.39 (d, J= 9.2 Hz, 1 H, Ar-H), 8.54 (s, 1 H, Ar-H), 9.90 (s, 1 H, benzene-NH). LC-MS (m/z) 535 (M+1)。
实施例 23
N-(4-((2-氨基苯基)氨基甲酰基)苄基) -6-(6,7-二甲氧基喹唑啉 4-氧) -1-萘酰胺的制备
Figure imgf000023_0001
以 6-(6,7-二甲氧基喹唑啉 4-氧) -1-萘酸 (37.6 mg, 0.1 mmol) 和 4- (氨基甲基) -N-(2-氨 基苯基)苯酰胺 (28.9 mg, 0.12 mmol)为原料, 按照实施例 16 中的类似步骤制得棕色固体 N-(4-((2-氨基苯基)氨基甲酰基)苄基) -6-(6,7-二甲氧基喹唑啉 4-氧) -1-萘酰胺 (43.1 mg, 产率 72%) LC-MS (m/z) 600 (M+1 )0
实施例 24
N-(4-((2-氨基 ·4-氟苯基)氨基甲酰基)苄基) -6- (6,7-二甲氧基喹唑啉 ~ -氧) -1-萘酰胺的制备
Figure imgf000023_0002
以 6-(6,7-二甲氧基喹唑啉 -4-氧) -1-萘酸 (37.6 mg, 0.1 mmol) 和 4- (氨基甲基) -N-(2-氨 基 -4-氟苯基)苯酰胺 (31.1 mg, 0.12 mmol)为原料, 按照实施例 16中的类似步骤制得棕色固 体 N-(4-((2-氨基 ~4-氟苯基)氨基甲酰基)苄基) -6-(6,7-二甲氧基喹唑啉 4-氧) -1-萘酰胺 (46.3 mg, 产率 75%)。 LC-MS (m/z) 618 (M+1)0
实施例 25
N-(2-氨基苯基) -6-((2-(6,7-二甲氧基喹唑啉 ·4-氧) -
1-萘酰胺)甲基)烟酰胺的制备
Figure imgf000024_0001
以 6-(6,7-二甲氧基喹唑啉 4-氧) -1-萘酸 (37.6 mg, 0.1 mmol) 和 6- (氨基甲基) -N-(2-氨 基苯基)烟酰胺 (29.0 mg, 0.12 mmol)为原料, 按照实施例 16 中的类似步骤制得棕色固体 ^(2-氨基苯基)-6-((2-(6,7-二甲氧基喹唑啉4-氧)-1-萘酰胺)甲基)烟酰胺(41.4 mg, 产率 69%) c LC-MS (m/z) 601 (M+1)0
实施例 26
N-(2-氨基 -4-氟苯基 )-6-((2-(6,7-二甲氧基喹唑啉 -4-氧) - 1 -萘酰胺)甲基)烟酰胺的制备
Figure imgf000024_0002
以 6-(6,7-二甲氧基喹唑啉 · -氧) -1-萘酸 (37.6 mg, 0.1 mmol) 和 6- (氨基甲基) - N-(2-氨 基 -4-氟苯基)烟酰胺 (31.2 mg, 0.12 mmol)为原料, 按照实施例 16中的类似步骤制得棕色固 体 N-(2-氨基 -4-氟苯基 )-6-((2-(6,7-二甲氧基喹唑啉 氧) -1-萘酰胺)甲基)烟酰胺 (43.3 mg, 产 率 77%)。 LC-MS (m/z) 619 (M+1) o
实施例 27
N-(3-((2-氨基苯基)氨基甲酰基)苄基) -6-(6,7-二甲氧基喹唑啉 ·4-氧) -1-萘酰胺的制备
Figure imgf000024_0003
以 6-(6,7-二甲氧基喹唑啉 -4-氧) -1-萘酸 (37.6 mg, 0.1 mmol) 和 3- (氨基甲基) -N-(2-氨 基苯基)苯酰胺 (28.9 mg, 0.12 mmol)为原料, 按照实施例 16 中的类似步骤制得棕色固体 N-(3-((2-氨基苯基)氨基甲酰基)苄基) -6-(6,7-二甲氧基喹唑啉 氧 )-1-萘酰胺 (48.5 mg, 产率 81 %)。 LC-MS (m/z) 600 (M+1 )0
实施例 28
N-(4-((2-氨基 · -甲基苯基)氨基甲酰基)苄基) -6- (6,7-二甲氧基喹唑啉 氧 )-1-萘酰胺的制备
Figure imgf000025_0001
以 6- (6,7-二甲氧基喹唑啉 -4-氧) -1-萘酸 (37.6 mg, 0.1 mmol) 和 4- (氨基甲基) -N-(2-氨 基 -4-甲基苯基)苯酰胺 (30.6 mg, 0.12 mmol)为原料, 按照实施例 16中的类似步骤制得棕色 固体 N-(4-((2-氨基 -4-甲基苯基)氨基甲酰基)苄基) -6-(6,7-二甲氧基喹唑啉 ·4-氧) -1-萘酰胺 (52.7 mg, 产率 86%)。 LC-MS (m/z) 614 (M+1)。
实施例 29
N-(4-((2-氨基 4-甲氧基苯基)氨基甲酰基)苄基) -6- (6,7-二甲氧基喹唑啉 4-氧) -1-萘酰胺的制备
Figure imgf000025_0002
以 6-(6,7-二甲氧基喹唑啉 -4-氧) -1-萘酸 (37.6 mg, 0.1 mmol) 和 4- (氨基甲基) -N-(2-氨 基 -4-甲氧基苯基)苯酰胺 (32.5 mg, 0.12 mmol)为原料, 按照实施例 16中的类似步骤制得棕 色固体 N-(4-((2-氨基 4-甲氧基苯基)氨基甲酰基)苄基) -6-(6,7-二甲氧基喹唑啉 4-氧) -1-萘酰 胺 (51.6 mg, 产率 82%)。 LC-MS (m/z) 630 (M+1)。
实施例 30
N-(4-((2-氨基 -4-三氟甲基苯基)氨基甲酰基)苄基) -6- (6,7-二甲氧基喹唑啉 氧 )-1-萘酰胺的制备
Figure imgf000025_0003
以 6-(6,7-二甲氧基喹唑啉 -4-氧) -1-萘酸 (37.6 mg, 0.1 mmol) 和 4- (氨基甲基) -N-(2-氨 基 -4-三氟甲基苯基)苯酰胺 (37.1 mg, 0.12 mmol)为原料, 按照实施例 16中的类似步骤制得 棕色固体 N-(4-((2-氨基 4-三氟甲基苯基)氨基甲酰基)苄基) -6-(6,7-二甲氧基喹唑啉 _4-氧) -1 - 萘酰胺 (46.7 mg, 产率 70%)。 LC-MS (m/z) 668 (M+1)。
实施例 31
N-(2-氨基苯基) -6-(7-甲氧基喹啉 4-氧) -1-萘酰胺的制备
Figure imgf000026_0001
以 6-(7-甲氧基喹啉 4-氧) -1-萘酸 (34.5 mg, 0.1 mmol) 和 邻苯二胺 (43.2 mg, 0.4 mmol)为原料,按照实施例 16中的类似步骤制得棕色固体 N-(2-氨基苯基) -6-(7-甲氧基喹啉 4- 氧) -1-萘酰胺 (39.6 mg, 产率 91%)。 1H NMR (DMSO-d6) δ 3.95 (s, 3H, -OCH3), 4.97 (s, 2H, benzene-NH2), 6.60 (d, J= 5.2 Hz, 1 H, Ar-H), 6.64 (t, J= 7.6 Hz, 1 H, Ar-H), 6.82 (d, J= 7.8 Hz, 1 H, Ar-H), 6.99 (t, J= 7.4 Hz, 1 H, Ar-H), 7.31 (dd, J= 2.5 and 9.1 Hz, 1 H, Ar-H), 7.38 (d, J= 7.6 Hz, 1 H, Ar-H), 7.45 (d, J= 2.4 Hz, 1 H, Ar-H), 7.57 (dd, J= 2.4 and 9.2 Hz, 1 H, Ar-H), 7.65 (t, J= 7.8 Hz, 1 H, Ar-H), 7.87-7.88 (m, 2H, Ar-H), 8.07 (d, J= 8.2 Hz, 1 H, Ar-H), 8.25 (d, J= 9.2 Hz, 1 H, Ar-H), 8.43 (d, J= 9.2 Hz, 1 H, Ar-H), 8.65 (d, J= 5.2 Hz, 1 H, Ar-H), 9.84 (s, 1 H, benzene-NH). LC-MS (m/z) 436 (M+1)。
实施例 32
N-(2-氨基 ·4-氟苯基 )-6-(7-甲氧基喹啉 4-氧) -1-萘酰胺的制备
Figure imgf000026_0002
以 6-(7-甲氧基喹啉 ~4-氧) -1-萘酸 (34.5 mg, 0.1 mmol) 和 4-氟邻苯二胺 (15.1 mg, 0.12 mmol)为原料, 按照实施例 16中的类似步骤制得棕色固体 N-(2-氨基 4-氟苯基 )-6-(7-甲 氧基喹啉 ·4-氧) -1-萘酰胺 (33.1 mg, 产率 73%)。 H NMR (DMSO-d6) δ 3.95 (s, 3Η, -OCH3), 5.27 (s, 2H, benzene-NH2), 6.41 (td, J= 2.5 and 8.4 Hz, 1 H, Ar-H), 6.57-6.61 (m, 2H, Ar-H), 7.30-7.36 (m, 2H, Ar-H), 7.45 (d, J= 2.2 Hz, 1 H, Ar-H), 7.56 (dd, J= 2.2 and 9.2 Hz, 1 H, Ar-H), 7.65 (t, J= 7.6 Hz, 1 H, Ar-H), 7.87-7.91 (m, 2H, Ar-H), 8.07 (d, J= 8.3 Hz, 1 H, Ar-H), 8.24 (d, J= 9.1 Hz, 1 H, Ar-H), 8.43 (d, J= 9.2 Hz, 1 H, Ar-H), 8.65 (d, J= 5.1 Hz, 1 H, Ar-H), 9.75 (s, 1 H, benzene-NH). LC-MS (m/z) 454 (M+1)。
实施例 33
N-(4-((2-氨基苯基)氨基甲酰基)苄基) -6-(7-甲氧基喹啉 4-氧) -1-萘酰胺的制备 O/AVeIsoz 08一 6u/od/zooiz is
Figure imgf000027_0001
fi¾«¾繮撇¾香¾¾ 1«猢 £ H(%ΙΝs 9ώ9s(((39 (寸))ϋ?- ----。 -, ίX 98ρ) _ε(H Ή)Vι(H Ή1) 89(V Ή)9(ΗνV6HΙ 6-·-·N =JSSSJJ---- - -· " - " - - · - -.
(HV Ή1) 87-(HV Ή)LHVΗε ΉΙ L L() ε98s(HVι)(Hv···JsJsJEJs----- " - - - - - - -· " - p) s(HV) εε?vh8HH ε8p)(HV ΉΙX 9gp)o g∞· =JsZJN = =-- - - - - - -·· - - -. - (HNqHI)6HN0(3 ΉΙ)26(hN ε6(H ΉΙH 9V ΉΙ) V g-··-95z9sussZ-- - . - - . - -. - - " -.
) i(i 0 (/)〇_ 9ωζ- 。" O/AV2οϊοί 08ϊ6u/ud/sl iooso
Figure imgf000028_0001
以 6-(6,7-二甲氧基喹啉 4-氧) -1-萘酸 (37.5 mg, 0.1 mmol) 和 4- (氨基甲基) -N-(2-氨基 苯基)苯酰胺 (28.9 mg, 0.12 mmol)为原料, 按照实施例 16 中的类似步骤制得棕色固体 N-(4-((2-氨基苯基)氨基甲酰基)苄基) -6-(6,7-二甲氧基喹啉 ·4-氧) -1-萘酰胺 (49.0 mg, 产率 82%)。 H NMR (DMSO-de) δ 3.93 (s, 3H, -OCH3), 3.95 (s, 3H, -OCH3), 4,63 (d, J= 5.6 Hz, 2H, -CH2), 4.90 (s, 2H, benzene-NH2), 6.56-6.59 (m, 2H, Ar-H), 6.78 (d, J= 7.6 Hz, 1 H, Ar-H), 6.96 (t, J= 8.1 Hz, 1 H, Ar-H), 7.17 (d, J= 7.6 Hz, 1 H, Ar-H), 7,42 (s, 1 H, Ar-H), 7.53-7.55 (m, 4H, Ar-H), 7.62 (t, J= 8.0 Hz, 1 H, Ar-H), 7.71 (d, J= 6.8 Hz, 1 H, Ar-H), 7.87 (s, 1 H, Ar-H), 7.98-8.06 (m, 3H, Ar-H), 8.39 (d, J= 9.2 Hz, 1 H, Ar-H), 8.49 (d, J= 5.2 Hz, 1 H, Ar-H), 9.26 (t, J= 6.0 Hz, 1 H, -CONH), 9.66 (s, 1 H, benzene-NH). LC-MS (m/z) 599 (M+1)。
实施例 38
N-(2-氨基苯基) -6-((2-(6,7-二甲氧基喹啉 ~4-氧) -1-萘酰胺)甲基)烟酰胺的制备
Figure imgf000029_0001
以 6-(6,7-二甲氧基喹啉 -4-氧) -1-萘酸 (37.5 mg, 0.1 mmol) 和 6- (氨基甲基) -N-(2-氨基 苯基)烟酰胺 (29.0 mg, 0.12 mmol)为原料,按照实施例 16中的类似步骤制得棕色固体 N-(2- 氨基苯基) -6-((2-(6,7-二甲氧基喹啉 4-氧) -1-萘酰胺)甲基)烟酰胺 (47.9 mg, 产率 80%)。 1H NMR (DMSO-de) δ 3.93 (s, 3Η, -OCH3), 3.95 (s, 3H, -OCH3), 4.73 (d, J= 5.6 Hz, 2H, -CH2), 4.97 (s, 2H, benzene-NH2), 6.57 (m, 2H, Ar-H), 6.77 (d, J= 6.4 Hz, 1 H, Ar-H), 6,98 (t, J= 8.1 Hz, 1 H, Ar-H), 7.16 (d, J= 5.6 Hz, 1 H, Ar-H), 7.42 (s, 1 H, Ar-H), 7.55-7.63 (m, 4H, Ar-H), 7.62 (t, J= 8.0 Hz, 1 H, Ar-H), 7,76 (d, J= 6.8 Hz, 1 H, Ar-H), 7.88 (s, 1 H, Ar-H), 8.06 (s, 1 H, Ar-H), 8,33 (s, 1 H, Ar-H), 8.45-8.48 (m, 2H, Ar-H), 9.12 (s, 1 H, Ar-H), 9.30 (t, J= 6.0 Hz, 1 H, -CONH), 9.80 (s, 1 H, benzene-NH). LC-MS (m/z) 600 (M+1)。
实施例 39
N-(2-氨基苯基) -6- (喹啉 ·4-氧) -1-萘酰胺的制备
Figure imgf000029_0002
以 6- (喹啉 ~4-氧) -1-萘酸 (31.5 mg, 0.1 mmol) 和 邻苯二胺 (43.2 mg, 0.4 mmol)为原料, 按照实施例 16中的类似步骤制得棕色固体 N-(2-氨基苯基) -6- (喹啉 4-氧) -1-萘酰胺 (35.6 mg, 产率 88%) H NMR (DMSO-de) 54.97 (s, 2H, benzene-NH2), 6.65 (t, J= 7.3 Hz, 1 H, Ar-H), 6.75 (d, J= 5.1 Hz, 1 H, Ar-H), 6,82 (d, J= 7.8 Hz, 1 H, Ar-H), 7.00 (t, J= 7.1 Hz, 1 H, Ar-H), ) L9(V Ή_H 36 p ε2pp) 6V ΉH 8ε9ζ(Hι 9ρ)··ΕJNeeJN-- - - -,. -., - - - (HXV ΉΙ 6p) sHVH( s) 808VΗ 36ε?νp(Hε8·JNJZJ =--- - -. - - - - - -. -,
(HNqHI) 986(VHHg) εVHp 8(H Ήι 26) S 8p,·u9 UsJZJ =Z =--- - -. - - -. - -.. -
Figure imgf000030_0001
N-(2-氨基苯基) -6-(8- (三氟甲基)喹啉 -4-氧) -1-萘酰胺的制备
Figure imgf000031_0001
以 6-(8-三氟甲基喹啉 · -氧) -1-萘酸 (39.8 mg, 0.1 mmol) 和 邻苯二胺 (43.2 mg, 0.4 mmol)为原料, 按照实施例 16中的类似步骤制得棕色固体 N-(2-氨基苯基) -6-(8-三氟甲基喹 啉 _4-氧) -1-萘酰胺 (38.3 mg, 产率 81%)。 1H NMR (DMSO-d6) δ 4.98 (s, 2Η, benzene-NH2), 6,65 (t, J= 7.3 Hz, 1 H, Ar-H), 6.83 (d, J= 7.6 Hz, 1 H, Ar-H), 6.89 (d, J= 5.2 Hz, 1 H, Ar-H), 7.00 (t, J= 7.2 Hz, 1 H, Ar-H), 7,38 (d, J= 7.5 Hz, 1 H, Ar-H), 7.62 (dd, J= 2.4 and 9.2 Hz, 1 H, Ar-H), 7.68 (t, J= 7.7 Hz, 1 H, Ar-H), 7.83 (t, J= 7.9 Hz, 1 H, Ar-H), 7.90 (d, J= 7.0 Hz, 1 H, Ar-H), 7.97 (d, J= 2.3 Hz, 1 H, Ar-H), 8.10 (d, J= 8.3 Hz, 1 H, Ar-H), 8.29 (d, J= 7.1 Hz, 1 H, Ar-H), 8,47 (d, J= 9,2 Hz, 1 H, Ar-H), 8.70 (d, J= 7.8 Hz, 1 H, Ar-H), 8.87 (d, J= 5.2 Hz, 1 H, Ar-H), 9.86 (s, 1 H, benzene-NH). LC-MS (m/z) 474 (M+1 )。
实施例 43
N-(4-((2-氨基苯基)氨基甲酰基)苄基) -6-(7-氯喹啉 氧) -1-萘酰胺的制备
Figure imgf000031_0002
以 6-(7-氯喹啉 ·4-氧) -1-萘酸 (35.0 mg, 0.1 mmol) 和 4- (氨基甲基) -N-(2-氨基苯基)苯酰 胺 (28.9 mg, 0.12 mmol)为原料, 按照实施例 16中的类似步骤制得棕色固体 N-(4-((2-氨基 苯基)氨基甲酰基)苄基) -6-(7-氯喹啉 ~4-氧) -1-萘酰胺 (42.4mg, 产率 74%)。1H NMR (DMSO-d6) δ 4.64 (d, J= 5.8 Hz, 2H, -CH2), 4.87 (s, 2H, benzene-NH2), 6.60 (t, J= 7.0 Hz, 1 H, Ar-H), 6.75-6.79 (m, 2H, Ar-H), 6.97 (t, J= 7.5 Hz, 1 H, Ar-H), 7,18 (d, J= 7.7 Hz, 1 H, Ar-H), 7.53-7.59 (m, 3H, Ar-H), 7.66 (t, J= 8.0 Hz, 1 H, Ar-H), 7.70-7.74 (m, 2H, Ar-H), 7.92 (d, J= 2.0 Hz, 1 H, Ar-H), 7.99 (d, J= 7.9 Hz, 2H, Ar-H), 8.06 (d, J= 8,2 Hz, 1 H, Ar-H), 8.13 (s, 1 H, Ar-H), 8.39-8.42 (m, 2H, Ar-H), 8,75 (d, J= 5.1 Hz, 1 H, Ar-H), 9.22 (t, J= 5.6 Hz, 1 H, -CONH), 9.62 (s, 1 H, benzene-NH). LC-MS (m/z) 573 (M+1)。
实施例 44
N-(4-((2-氨基苯基)氨基甲酰基)苄基) -6-(8- (三氟甲基)喹啉 氧) -1-萘酰胺的制备
Figure imgf000032_0001
以 6-(8-三氟甲基喹啉 -4-氧) -1-萘酸 (38.3 mg, 0.1 mmol) 和 6- (氨基甲基) -N-(2-氨基苯 基)烟酰胺 (29.0 mg, 0.12 mmol)为原料,按照实施例 16中的类似步骤制得棕色固体 N-(4-((2- 氨基苯基)氨基甲酰基)苄基) -6-(8- (三氟甲基)喹啉 4-氧) -1-萘酰胺 (47.3mg, 产率 78%)。 1H NMR (DMSO-de) δ 4.64 (d, J= 5.6 Hz, 2H, -CH2), 4.87 (s, 2H, benzene-NH2), 6.60 (t, J= 7.2 Hz, 1 H, Ar-H), 6.78 (d, J= 7.8 Hz, 1 H, Ar-H), 6,89 (d, J= 5.1 Hz, 1 H, Ar-H), 6.97 (t, J= 7.2 Hz, 1 H, Ar-H), 7.18 (d, J= 7.9 Hz, 1 H, Ar-H), 7.53-7.66 (m, 4H, Ar-H), 7.74 (d, J= 6.9 Hz, 1 H, Ar-H), 7.83 (t, J= 7.9 Hz, 1 H, Ar-H), 7.95-8.08 (m, 4H, Ar-H), 8.29 (d, J= 7.0 Hz, 1 H, Ar-H), 8.42 (d, J= 9.1 Hz, 1 H, Ar-H), 8.69 (d, J= 8,3 Hz, 1 H, Ar-H), 8.86 (d, J= 5.0 Hz, 1 H, Ar-H), 9.22 (t, J= 5.5 Hz, 1 H, -CONH), 9.61 (s, 1 H, benzene-NH)丄 C-MS (m/z) 607(M+1)。
实施例 45 片剂的制备
处方 ( 1000片):
化合物 31 : 5 g
微晶纤维素: 90 g
羧甲基淀粉钠: 5 g
4%聚维酮 K30无水乙醇溶液: 50 g
滑石粉: 0.5 g
制备工艺: 将化合物 31过 100目筛, 将微晶纤维素、羧甲基淀粉钠和滑石粉过 80目筛, 称取处方量的微晶纤维素和羧甲基淀粉钠, 混合均匀, 然后将化合物 31与其按等量递加法混 匀, 加入 4%聚维酮 K30无水乙醇溶液适量制粒, 干燥后加入处方量滑石粉混匀, 压片即得。
实施例 46 胶囊剂的制备
处方 ( 1000粒):
化合物 31 :
微晶纤维素- 乳糖- 羧甲基淀粉钠:
硬脂酸镁:
制备工艺: 将化合物 31过 100目筛, 将微晶纤维素、 乳糖、 羧甲基淀粉钠和硬脂酸镁过 80 目筛, 称取处方量的微晶纤维素、 乳糖和羧甲基淀粉钠, 混合均匀, 然后将化合物 31 与 其按等量递加法混匀, 加入处方量硬脂酸镁混匀, 灌装胶囊即得。
实施例 47 注射液的制备
处方: 化合物 31 : 1.00 mg
药用 DMSO: 0.10 ml
药用乙醇: 1.00 ml
制备工艺: 将化合物 31溶于药用 DMSO, 加入药用乙醇即得。
实施例 48
通式 (I) 化合物 PDGF和 VEGF配体依赖性细胞增殖试验
(一) PDGF依赖性细胞增殖:
采用稳定表达人 PDGFRP的小鼠 NIH-3T3成纤维工程细胞株来评价 PDGF依赖的细胞增 殖。 PDGFR "NIH-3T3细胞以 5000每孔的密度分种于 96孔微孔板, 24小时后换成无血清培 养基培养过夜。 加入待测化合物和 PDGF BB (50ng/ml), 继续在无血清培养基中培养 72小 时。 细胞增殖情况根据说明用 MTS方法 (Promega) 进行检测。 在 C02培养箱 37°C培养 2 小时, 然后在 ELISA酶标仪上测量 490nm的吸收值。
(二) VEGF依赖性细胞增殖:
HUVEC细胞以 6000每孔的密度分种于 96孔微孔板, 24小时后换成无血清培养基中培 养 2小时。 加入待测化合物和 VEGF 165 (50ng/ml), 继续在无血清培养基中培养 72小时。 细胞增殖情况根据说明用 MTS方法(Promega)进行检测。 在 C02培养箱 37°C培养 2小时, 然后在 ELISA酶标仪上测量 490nm的吸收值。
实验结果见表 2。
表 2
样品 GI50 nM GI50 n
(化合物) (PDGF 配体依赖细胞增殖) (VEGF配体依赖细胞增殖)
16 48 3
17 40 3
18 15 7
19 11 23
20 23 6
21 19 5
22 372 3
23 148 18
25 69 13
31 46 5
32 20 2
33 300 8
34 248 90
35 5 1
36 3 2
37 159 4 38 74 25
39 32 107
40 1000 1000
41 479 105
42 48 1000
43 1000 288
44 1000 1000
实施例 49
通式 (I ) 化合物体外抑制 HDAC总酶活性和
体外抑制 HDAC亚型活性的试验。
一、 体外 HDAC总酶活性的测定:
体外 HDAC总酶活性的测定是我们采用 BIOMOL公司的 HDAC 7¾or/ e/r/c
Figure imgf000034_0001
Discovery Kit对抑制剂的体外抑制活性进行测试。实验的原理如下:一种特殊的底物 /«or cfe 在组蛋白去乙酰化酶(实验中采用 HeLa细胞的核提取物, 富含多种亚型的 HDAC ) 的作 用下会去掉一个乙酰基, 暴露出游离的氨基, 当加入 Developei "后会产生一种可诱导的荧光。 这种荧光的激发波长为 360nm, 发射波长为 460nm。 底物去乙酰化越充分, 诱导的荧光值就 越高, 在无抑制剂情况下的荧光值为对照; 而当有抑制剂存在的时候, 被诱导的荧光值会降 低, 而无酶时 (相当于酶活性完全被抑制) 的荧光值为空白。 而一般抑制后的荧光值会介于 对照和空白之间。 在分析时我们将空白作为 0, 对照作为 1进行计算, 值越小则说明抑制活 性越高。
1. 将实验缓冲液, 稀释后的 trichostatin A及被测试的抑制剂加入微孔板适当的孔中。 下表中 列出了不同实验类型每种试剂的用量。
Figure imgf000034_0002
2. 将稀释的 HeLa核蛋白提取物加入那些除标记为"空白"以外的所有孔中。
3. 让稀释后的 F/MOr cfe Z ^™底物和微孔板中的样品平衡到 25°C。
4. 将稀释后的底物 (25 μΐ ) 加入每一个孔中并混合均匀以启动 HDAC反应。
5. 在室温反应 30分钟, 然后加入 i/or ife Zj^™显影剂 (50 μΐ ) 终止反应。 将微孔板在室 温 ( 25°C ) 孵育 10-15分钟。
6. 在激发波长为 369纳米, 发射波长为 451纳米的荧光酶标仪上读取荧光值。
二、 报告基因对抑制剂 HDAC亚型选择性的测定:
HDAC不同亚型可以与不同的转录因子结合, 参与不同基因的表达调控中。 选择合适转 录因子的调控元件构建成报告基因, 可以用来评估抑制剂对 HDAC亚型的选择性抑制作用。 在转染前一天将 HeLa细胞种入 96孔板使转染时密度达到 50-80%的融合。在荧光素酶基因上 游***了 p21-启动子调控序列的报告基因质粒分别使用 FuGene6 ( Roche )转染试剂, 根据操 作说明进行转染。 同时为了校正转染效率, 共转染了绿色荧光蛋白 (GFP ) 的表达质粒。 在 转染 24小时后加入化合物或溶剂对照(DMSO)。 再过 24小时后收集并裂解细胞, 按照操作 说明使用荧光素酶 (Promega ) 检测试剂盒对荧光素酶的量进行评估。 同时进一步测定 GFP 的含量, 对转染效率进行校正。
实验结果见表 3。
表 3
样品 30μΜ下 HDAC总酶抑制活性 10μΜ下 HDAC I类 (Ρ21报告基因) (化合物) ( %) 诱导倍数
CS055 50.4 33
16 8.6 1.3
17 22.5 1.1
18 17.1 1.1
19 21.9 1.4
20 21.9 1.5
21 18.6 1.1
22 17 1.1
23 49.4 11.3
25 47.9 12.1
31 10.1 1.6
32 21.7 1.8
33 39.1 2.8
34 38.8 5.0
35 19.3 1.2
36 14.4 1.2
37 35.9 3.0
38 39.3 3.1
39 15.9 1.2
40 22.2 1.3
41 19.3 1.1
42 6.2 1.3
43 38.7 6.1
44 35.1 3.2
CS055: 西达本胺, 深圳微芯生物科技有限责任公司研制的 HDAC抑制剂, 具有优异的抗肿瘤活性, 正进行临床 2期试验。
实施例 50
通式 (I ) 化合物对肿瘤细胞增殖的抑制试验。
将肿瘤细胞用胰酶消化后, 以 3000个每孔的密度分种于 96孔微孔板, 在含 10%FBS的 完全培养基里培养 24小时。 加入待测化合物和溶剂对照, 最终化合物浓度为 100 nmol/L到 100 μπιοΙ/L.然后在完全培养基里培养 72小时。根据说明书的方法加入 MTS试剂(Promega), 在 37°C C02培养箱中培养 2小时, 然后在 ELISA酶标仪上读取 490nm的吸收值。
实验结果见表 4。
表 4
样品 G O MM G o MM G\50 iM Gl50 μΜ Glso MM (化合物) (A-498) (A549) ( Bel-7402 ) ( HCT-8 ) ( MCF-7)
CS055 12.08 11.15 18.93 7.711 3.865
16 30.0 30.0 30.0 12.3 30.0
17 30.0 30.0 30.0 3.0 30.0
18 nd nd nd nd nd
19 nd nd nd nd nd
20 nd nd nd nd nd
21 nd nd nd nd nd
22 nd nd nd nd nd
23 14.7 30.0 30.0 5.7 4.3
25 14.7 30.0 30.0 4.9 6.1
31 9.5 17.3 30.0 6.6 10.2
32 7.5 8.3 17.3 6.6 15.9
33 1.9 2.1 2.8 1.5 2.0
34 7.9 11.2 17.7 5.5 5.2
35 9.1 7.7 19.5 8.9 13.3
36 4.2 7.4 12.1 4.1 8.9
37 30.0 30.0 30.0 30.0 30.0
38 6.9 30.0 30.0 8.0 9.4
39 nd nd nd nd nd
40 nd nd nd nd nd
41 nd nd nd nd nd
42 nd nd nd nd nd
43 nd nd nd nd nd
44 nd nd nd nd nd nd*: 未测定;
CS055: 西达本胺, 深圳微芯生物科技有限责任公司研制的 HDAC抑制剂, 具有优异的抗肿瘤活性, 正进行临床 2期试验。
实施例 51
化合物 31对人 A549肺癌裸鼠移植性肿瘤的抑制试验 选用裸鼠, 雌性, 14-16g, 经正常饲养观察 3天如无异常, 以 50只常规腋下接种人 A549 肺癌后饲养观察, 确认瘤细胞着床增殖且瘤块直径大于 6mm时随机分组, 8只 /组, 共 6组, 其中受试药物 4个给药浓度共四组, 溶剂对照一组, 阳性对照药 Sutent—组。 即日口服给药, 1次 /日, 共 24次, 化合物 31的给药剂量为 5、 10、 20和 40mg/kg体重。 每周 2次称量动物 体重及测量肿瘤体积。 给药 24次后次日处死动物剥取肿瘤称瘤重, 按公式: [ (对照组平均 瘤重 -实验组平均瘤重) /对照组平均瘤重] χ 100%, 计算各组肿瘤抑制率。 实验结果见表 5和 图 1。
表 5
剂量 体重 (g) 瘤重
分组 a TGI (%) b P
(mg kg) 起始 结束 (g)
溶剂 - 20.3±0. 9 25·4± 2. 3 4.20±0·75 - -
Sutent 40 20.3士 1. 4 24.4±2.3 2.06±0.71 50.9 <0.001 化合物 31 40 20·0±0·9 22.6±2.4 1.06±0.54 74.8 <0.001 化合物 31 20 20.6± 1 ·1 24.2 ±0.7 1.50±0.41 64.3 <0.001 化合物 31 10 19.9± 1.3 25.1 ± 1.3 2.13±0.51 49.4 <0.001 化合物 31 5 21.1 ±0·6 24.6± 1.3 2.20±0.57 47.6 <0.001 a每组 8只裸鼠。 b肿瘤生长抑制率。
实施例 52
化合物 31对人 HCT-8肠癌裸鼠移植性肿瘤的抑制试验 选用裸鼠,雌性, 18-20g,经正常饲养观察 3天如无异常,以 50只常规腋下接种人 HCT-8 肠癌后饲养观察, 确认瘤细胞着床增殖且瘤块体积不小于 100mm3时随机分组, 8只 /组, 共 6组, 其中受试药物 4个给药浓度共四组, 溶剂对照一组, 阳性对照药 Sutent—组。 即日口 服 药, 1次 /日, 共 24次, 化合物 31的给药剂量为 2.5、 5、 10和 20mg/kg体重。 每周 2次 称量动物体重及测量肿瘤体积。 给药 20次后次日处死动物剥取肿瘤称瘤重, 按公式: [ (对 照组平均瘤重 -实验组平均瘤重) /对照组平均瘤重 ]χ 100%, 计算各组肿瘤抑制率。 实验结果 见表 6和图 2。
表 6
剂量 体重 (g) 瘤重
分组 a TGI (%) b P
(mg/kg)
起始 结束 (g)
溶剂 - 20.8± 1. 0 22.1 ±2. 1 4.78 ± 1.99 - -
Sutent 40 21.5±0. 7 22·4± 1 ·1 0.23±0.07 95.3 <0.001 化合物 31 20 20.5± 1.3 22.5± 1.6 0.19±0.06 96.1 <0.001 化合物 31 10 20.7± 1.1 23.7 ±0.8 0.46±0.15 90.3 <0.001 化合物 31 5 21.6± 1.4 24.8 ± 1.5 0.78±0.25 83.8 <0.001 化合物 31 2.5 20.3±0.8 24.5 ± 1.1 2.18± 1.28 54.5 <0.001 只裸鼠。 b肿瘤生长抑制率。
实施例 53 化合物 31对人 SSMC7721肝癌裸鼠移植性肿瘤的抑制试验
选用裸鼠, 雌性, 18-20g, 经正常饲养观察 3 天如无异常, 以 50 只常规腋下接种人 SSMC7721肝癌后饲养观察,确认瘤细胞着床增殖且瘤块体积不小于 100mm3时随机分组, 8 只 /组, 共 6组, 其中受试药物 4个给药浓度共四组, 溶剂对照一组, 阳性对照药 Sutent—组。 即日口服给药, 1次 /日, 共 24次, 化合物 31的给药剂量为 2.5、 5、 10和 20mg/kg体重。 每 周 2次称量动物体重及测量肿瘤体积。 给药 24次后次日处死动物剥取肿瘤称瘤重, 按公式:
[ (对照组平均瘤重 -实验组平均瘤重) /对照组平均瘤重] χ ΐοο%, 计算各组肿瘤抑制率。 实验 结果见表 7和图 3。
表 7
剂量 体重 (g) 瘤重
分组 a TGI (%) b P
(mg/kg) 起始 结束 (g)
溶剂 - 20.8±0.8 25.1 ± 1.5 3.36±0.41 - -
Sutent 40 21.0±0.8 24.8 ± 1.2 1.00±0.68 70.3 <0.001 化合物 31 20 20.2 ± 1.7 21.0±2.2 0.53±0.28 84.4 <0.001 化合物 31 10 20.4 ± 1.6 23.6± 1.5 0.70±0.45 79.2 <0.001 化合物 31 5 20.8± 1.2 24.8 ± 1.5 1.16±0.55 65.4 <0.001 化合物 31 2.5 20.1 ±0.9 23.2±2.1 1.63 ±0.70 51.7 <0.001 a每组 8只裸鼠。 b肿瘤生长抑制率。
实施例 54
化合物 33、 34对人 HCT-8肠癌裸鼠移植性肿瘤的抑制试验 选用裸鼠,雌性, 18-20g,经正常饲养观察 3天如无异常, 以 50只常规腋下接种人 HCT-8 肠癌后饲养观察, 确认瘤细胞着床增殖且瘤块体积不小于 100mm3时随机分组, 8只 /组, 共 6组,其中受试药物化合物 33、 34各 2个给药浓度共四组,溶剂对照一组, 阳性对照药 Sutent 一组。 即日口服给药, 1次 /日, 共 20次。 每周 2次称量动物体重及测量肿瘤体积。 给药 20 次后次日处死动物剥取肿瘤称瘤重, 按公式: [ (对照组平均瘤重 -实验组平均瘤重) /对照组 平均瘤重 ]χ 100%, 计算各组肿瘤抑制率。 实验结果见表 8和图 4。
表 8
剂量 体重 (g) 瘤重
分组 a TGI (%) b P
(mg/kg)
起始 结束 (g)
溶剂 - 19.4± 1.6 21.2±2.4 4.08±0.95 - -
Sutent 40 20.6± 1.2 22.1 ± 1.5 0.44 ±0.15 89.1 <0.001 化合物 33 60 19.4±0.8 21 ·4± 1 ·5 1.98±0.61 51.5 <0.001 化合物 33 30 19.0± 1.3 21.1 ±2.2 2.31 ±0.43 43.3 <0.001 化合物 34 60 19.6± 1.1 21.6±2·3 2.74 ±0.77 32.7 <0.001 化合物 34 30 19.7± 1.2 21.2± 1 ·9 3.95 ±0.73 3.07 >0.05
每组 8只裸鼠。 b肿瘤生长抑制率。 实施例 55
化合物 33、 37对人 HCT-8肠癌裸鼠移植性肿瘤的抑制试验 选用裸鼠,雌性, 18-20g,经正常饲养观察 3天如无异常, 以 50只常规腋下接种人 HCT-8 肠癌后饲养观察, 确认瘤细胞着床增殖且瘤块体积不小于 100mm3时随机分组, 8只 /组, 共 6组,其中受试药物化合物 33、 37各 2个给药浓度共四组,溶剂对照一组, 阳性对照药 Sutent 一组。 即日口服给药, 化合物 33两个剂量组 2次 /日, 间隔 6小时, 其余各组 1次 /日, 共 20 天。 每周 2次称量动物体重及测量肿瘤体积。 给药 20天后次日处死动物剥取肿瘤称瘤重, 按 公式: [ (对照组平均瘤重 -实验组平均瘤重) /对照组平均瘤重] χΐοο%, 计算各组肿瘤抑制率。 实验结果见表 9和图 5。
表 9
剂量 体重 (g) 瘤重
分组 a TGI (%) b P
(mg/kg) 起始 结束 (g)
溶剂 - 21.1 ±0.7 23·4± 1 ·5 6.13±0.28 - -
Sutent 40 21.3±0.6 23.7±0.8 0.29±0.08 95.3 <0.001 化合物 33 60X 2 20.1 ±0.9 19.0± 1.8 0.45±0.05 92.6 <0.001 化合物 33 30 X 2 21.1 ± 1.2 22.6± 1.6 0.73 ±0.36 88.1 <0.001 化合物 37 60 20.8±0.8 24.1 ±2.1 3.36 ±0.80 45.1 <0.001 化合物 37 30 20.6±0.8 23·6±2·2 3.89± 1.19 36.5 <0.001 a每组 8只裸鼠。 b肿瘤生长抑制率。
实施例 56
化合物 33、 37对人 SSMC7721肝癌裸鼠移植性肿瘤的抑制试验 选用裸鼠, 雌性, 18-20g, 经正常饲养观察 3 天如无异常, 以 50 只常规腋下接种人 SSMC7721肝癌后饲养观察,确认瘤细胞着床增殖且瘤块体积不小于 100mm3时随机分组, 8 只 /组, 共 6组, 其中受试药物化合物 33、 37各 2个给药浓度共四组, 溶剂对照一组, 阳性 对照药 Sutent—组。 即日口服给药, 1次 /日, 共 30次。 每周 2次称量动物体重及测量肿瘤体 积。 给药 30次后次日处死动物剥取肿瘤称瘤重, 按公式: [ (对照组平均瘤重-实验组平均瘤 重) /对照组平均瘤重 ]x l00%, 计算各组肿瘤抑制率。 实验结果见表 10和图 6。
表 10
剂量 体重 (g) 瘤重
分组 a TGI (%) b P
(mg/kg) 起始 结束 (g)
溶剂 - 21.1 ±0.4 24.5 ± 1.6 2.25±0.85 - -
Sutent 40 21.2± 1.1 24.0 ±0.6 0.88±0.39 61.1 <0.001 化合物 33 60 21.4± 1.3 25.4±2.8 1.48±0.89 34.4 >0.05
化合物 33 30 20.8±0.5 24.0 ± 1.7 1.63±0.47 27.8 >0.05
化合物 37 60 21.4士 0.6 24.3± 1.1 1.28±0.51 43.3 <0.05
化合物 37 30 20.7± 1.2 25.3±0.9 1.45 ±0.58 35.6 <0.05
每组 8只裸鼠。 b肿瘤生长抑制率。

Claims

权 利 要 求
1 . 一种具有通式 (I) 的化合物:
Figure imgf000040_0001
(I)
中,
Z为 CH或 N;
R1. R2和 R3 分别为氢、 卤素、 烷基、 烷氧基或三氟甲基;
R4
Figure imgf000040_0002
X为苯环或吡啶环;
R5为一个或多个取代基, 选自氢、 卤素、 垸基、 烷氧基或三氟甲基; 包括其游离形式、 盐的形式、 对映异构体、 非对映异构体或水合物。
2. 如权利要求 1所述的化合物, 其特征在于所述化合物为- Z为 CH;
R1. R2和 R3 分别为氢、 卤素、 垸基、 烷氧基或三氟甲基;
R4
Figure imgf000040_0003
X为苯环或吡啶环;. .
R5为一个或多个取代基, 选自氢、 卤素、 烷基、 垸氧基或三氟甲基。
3. 如权利要求 1所述的化合物, 其特征在于所述化合物为: Z为 CH;
R1 R2和 R3 分别为氢或垸氧基
R4
Figure imgf000041_0001
X为苯环或吡啶环;
R5为一个或多个取代基, 选自氢、 卤素、 烷基、 烷氧基或三氟甲基。
4. 如权利要求 1所述的化合物,.其特征在于所述化合物为- Z为 CH;
I 1、 R2 分别为氢或甲氧基;
R3为 H;
R4
Figure imgf000041_0002
X为苯环或吡啶环;
R5为一个或多个取代基, 选自氢、 卤素、 垸基、 垸氧基或三氟甲基。
5. 如权利要求 1所述的化合物, 其特征在于所述化合物为:
Z为 CH;
R1 , R2 分别为氢或甲氧基;
R3为 H;
R4
Figure imgf000041_0003
X为苯环或吡啶环;
R5为 H或F。
6. .通式 (I) 的化合物的制备方法, 该方法是将通式 (Π) 化合物
Figure imgf000042_0001
(II)
与通式 (III) 化合物
H2N-R4 (ΠΙ)
在有机溶剂和肽缩合剂存在下进行缩合反应得到通式 (I) 化合物, 其中,
Z为 CH或 N;
R R2和 R3 分别为氢、 卤素、 垸基、 垸氧基或三氟甲基;
R4
Figure imgf000042_0002
X为苯环或吡啶环;
R5为一个或多个取代基, 选自氢、 卤素、 烷基、 垸氧基或三氟甲基。
7. 如权利要求 6所述的化合物的制备方法, 其特征在于: 所述肽缩合剂选自 1-乙基 -3- (3-二甲胺丙基) 碳二亚胺 (EDC:)、 N, Ν '-二环己基碳二亚胺 (DCC) 或 Ν, Ν'-碳酰二咪唑 (CDI)。
8. 如权利要求 6的所述的化合物的制备方法,其特征在于:所述有机溶剂选自苯、 甲苯、 四氢呋喃、 1,4-二氧六环、 二氯甲烷、 氯仿或 N, N-二甲基甲酰胺。
9.一种用于治疗与蛋白激酶活性异常或组蛋白去乙酰化酶活性异常相关的疾病的药用制 剂, 其特征在于, 该制剂包括作为活性组分的权利要求 1所述通式 (I) 的化合物以及药用载 体、 辅料或稀释剂。
10. 如权利要求 9所述的药用制剂, 其特征在于: 所述制剂为片剂、 胶囊、 粉剂、 糖浆、 液剂、 悬浮剂、 针剂、 膏剂。
11 . 如权利要求 1所述的化合物在制备用于治疗炎症、 自身免疫性疾病、 癌症、 神经系 统疾病和神经退化性疾病、 过敏、 哮喘、 心血管疾病及代谢病, 或与激素相关的疾病的药物 中的应用。
12. 如权利要求 9所述的药用制剂在制备用于治疗炎症、 自身免疫性疾病、 癌症、 神经 ***疾病和神经退化性疾病、 过敏、 哮喘、 心血管疾病及代谢病, 或与激素相关的疾病的药 物中的应用。
13.如权利要求 9所述的药用制剂,其特征在于:所述制剂的剂型规格为 0.0001~200 mg。
PCT/CN2010/000272 2009-06-04 2010-03-05 作为蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘酰胺衍生物、其制备方法及应用 WO2010139180A1 (zh)

Priority Applications (16)

Application Number Priority Date Filing Date Title
AU2010256246A AU2010256246B9 (en) 2009-06-04 2010-03-05 Naphthalene carboxamide derivatives as inhibitors of protein kinase and histone deacetylase, preparation methods and uses thereof
KR1020117030659A KR101421786B1 (ko) 2009-06-04 2010-03-05 단백질 키나제 및 히스톤 디아세틸라제의 억제제로서 나프탈렌 카르복스아미드 유도체, 그 제조 방법 및 용도
DK10782884.0T DK2439195T3 (da) 2009-06-04 2010-03-05 Naftalenkarboxamidderivater som proteinkinasehæmmere og histondeacetlylase, fremstillingsmetoder samt anvendelser deraf
SI201030695T SI2439195T1 (sl) 2009-06-04 2010-03-05 Derivati naftalen karboksamida kot inhibitorji protein kinaze in histon deacetilaze, pripravljalni postopki in njihova uporaba
MX2011012752A MX2011012752A (es) 2009-06-04 2010-03-05 Derivados de naftalenocarboxamida como inhibidores de proteina quinasa y de histona desacetilasa, procedimientos de preparacion y usos de los mismos.
PL10782884T PL2439195T3 (pl) 2009-06-04 2010-03-05 Pochodne naftalenokarboksyamidu jako inhibitory kinazy białkowej i deacetylazy histonowej, sposoby ich wytwarzania oraz ich zastosowania
ES10782884.0T ES2509615T3 (es) 2009-06-04 2010-03-05 Derivados de naftaleno carboxamida como inhibidores de proteína cinasa e histona desacetilasa, procedimientos de preparación y usos de los mismos
CA2763822A CA2763822C (en) 2009-06-04 2010-03-05 Naphthalene carboxamide derivatives as inhibitors of protein kinase and histone deacetylase, preparation methods and uses thereof
BRPI1011994A BRPI1011994B8 (pt) 2009-06-04 2010-03-05 derivados de naftaleno carboxamida como inibidores de proteína quinase e histona desacetilase, métodos de preparação e usos
EP10782884.0A EP2439195B1 (en) 2009-06-04 2010-03-05 Naphthalene carboxamide derivatives as inhibitors of protein kinase and histone deacetylase, preparation methods and uses thereof
JP2012513451A JP5484568B2 (ja) 2009-06-04 2010-03-05 プロテインキナーゼおよびヒストンデアセチラーゼの阻害剤としてのナフタレンカルボキサミド誘導体、その製造方法および用途
UAA201115118A UA103092C2 (ru) 2009-06-04 2010-03-05 Производные нафталинкарбоксамида как ингибиторы протеинкиназы и гистондеацетилазы, способ их получения и применения
RU2011152113/04A RU2497809C2 (ru) 2009-06-04 2010-03-05 Производные нафталинкарбоксамида в качестве ингибиторов протеинкиназы и гистондеацетилазы, способы их получения и применение
KR1020147000095A KR20140014313A (ko) 2009-06-04 2010-03-05 단백질 키나제 및 히스톤 디아세틸라제의 억제제로서 나프탈렌 카르복스아미드 유도체, 그 제조 방법 및 용도
ZA2011/09030A ZA201109030B (en) 2009-06-04 2011-12-08 Naphthalene carboxamide derivatives as inhibitors of protein kinase and histone deacetylase,preparation methods and uses thereof
HRP20140717AT HRP20140717T1 (hr) 2009-06-04 2014-07-25 Derivati naftalen karboksamida kao inhibitori protein kinaze i histon deacetilaze, postupci njihove pripreme i njihove uporabe

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN200910143978.2 2009-06-04
CN200910143978 2009-06-04

Publications (2)

Publication Number Publication Date
WO2010139180A1 true WO2010139180A1 (zh) 2010-12-09
WO2010139180A8 WO2010139180A8 (zh) 2012-01-05

Family

ID=43261686

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2010/000272 WO2010139180A1 (zh) 2009-06-04 2010-03-05 作为蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘酰胺衍生物、其制备方法及应用

Country Status (18)

Country Link
EP (1) EP2439195B1 (zh)
JP (1) JP5484568B2 (zh)
KR (2) KR101421786B1 (zh)
CN (1) CN101906076B (zh)
AU (1) AU2010256246B9 (zh)
BR (1) BRPI1011994B8 (zh)
CA (1) CA2763822C (zh)
DK (1) DK2439195T3 (zh)
ES (1) ES2509615T3 (zh)
HR (1) HRP20140717T1 (zh)
MX (1) MX2011012752A (zh)
PL (1) PL2439195T3 (zh)
PT (1) PT2439195E (zh)
RU (1) RU2497809C2 (zh)
SI (1) SI2439195T1 (zh)
UA (1) UA103092C2 (zh)
WO (1) WO2010139180A1 (zh)
ZA (1) ZA201109030B (zh)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103351336A (zh) * 2013-06-28 2013-10-16 华侨大学 作为蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘甲酰胺衍生物及其制备方法
CN103896836A (zh) * 2014-03-20 2014-07-02 华侨大学 N-苯基-6-[(7-氯喹啉-4-氧基)酚醚]-2-萘甲酰胺及其制备方法
CN103923004A (zh) * 2014-04-04 2014-07-16 华侨大学 一种萘甲酰胺衍生物及其制备和应用
CN104030980A (zh) * 2014-06-10 2014-09-10 华侨大学 N-(3-甲氧基-4-氯苯基)-4-[(7-氯-4-喹啉)氨基]苯甲酰胺及其制备和应用
WO2018187894A1 (zh) * 2017-04-10 2018-10-18 师健友 一种用于***疾病以及具有抗菌、抗病毒和抗炎作用的药物
RU2784869C1 (ru) * 2019-03-25 2022-11-30 Шэньчжэнь Чипскрин Байосайенсиз Ко., Лтд. Чиаураниб для лечения мелкоклеточного рака легкого
WO2022247844A1 (zh) * 2021-05-26 2022-12-01 深圳微芯生物科技股份有限公司 包含蛋白激酶抑制剂的药物组合物及其医药用途

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011057661A (ja) * 2009-08-14 2011-03-24 Bayer Cropscience Ag 殺虫性カルボキサミド類
CN102260210B (zh) * 2011-05-30 2012-06-27 王立强 蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘酰胺衍生物的制备方法
CN102603627B (zh) * 2011-05-31 2013-02-06 王立强 作为蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘酰胺衍生物及其制备方法
CN102850236B (zh) * 2011-06-27 2016-05-18 国药一心制药有限公司 新型苯甲酰胺类组蛋白去乙酰化酶抑制剂及其应用
CN103420923B (zh) * 2012-05-18 2015-08-19 国药一心制药有限公司 4-氨基喹唑啉异羟肟酸类化合物及作为抗肿瘤药物应用
CN103288728A (zh) * 2013-05-17 2013-09-11 华侨大学 一种萘甲酰胺衍生物、其制备方法以及应用
CN103432077A (zh) * 2013-08-21 2013-12-11 北京淦航医药科技有限公司 西达苯胺固体分散制剂
CN104418867B (zh) * 2013-08-26 2016-12-28 上海汇伦生命科技有限公司 作为PI3K/mTOR抑制剂的化合物,其制备方法和用途
CN103724260A (zh) * 2013-12-18 2014-04-16 华侨大学 一种苯甲酰胺衍生物及其制备方法和应用
CN104860885B (zh) 2014-02-24 2017-11-17 中国科学院上海药物研究所 萘酰胺类化合物、其制备方法和用途
CN105399685B (zh) * 2014-09-16 2018-05-22 深圳微芯生物科技有限责任公司 作为选择性jak3和/或jak1激酶抑制剂的芳杂环化合物的制备方法及其应用
CN104447534A (zh) * 2014-12-04 2015-03-25 厦门大学 6-[(7-氯喹啉-4-氧基)酚醚]-2-萘甲酰胺类衍生物及其制备方法和用途
CN107868044B (zh) * 2016-09-27 2020-10-16 深圳微芯生物科技股份有限公司 一种非溶剂化晶体及其制备方法与应用
CN109985039A (zh) * 2017-12-29 2019-07-09 深圳微芯生物科技股份有限公司 用于治疗白血病的联合用药物及其在制备用于治疗急性髓性白血病的药物中的用途
CN110833544B (zh) * 2018-08-17 2022-08-09 深圳微芯生物科技股份有限公司 组蛋白去乙酰化酶抑制剂与蛋白激酶抑制剂之组合及其制药用途
CN109908143B (zh) * 2018-12-18 2021-11-19 厦门大学附属第一医院 西奥罗尼在制备治疗急性髓系白血病药物的新用途
TWI797426B (zh) * 2019-03-25 2023-04-01 大陸商深圳微芯生物科技股份有限公司 西奧羅尼用於小細胞肺癌的治療
CN113440615A (zh) * 2020-03-24 2021-09-28 深圳微芯生物科技股份有限公司 包含蛋白激酶抑制剂和化疗药物的药物组合物及其用途
US20230382871A1 (en) * 2020-09-23 2023-11-30 Shanghai Institute Of Materia Medica, Chinese Academy Of Sciences Csf1r kinase inhibitor and use thereof

Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0847992A1 (en) 1996-09-30 1998-06-17 Mitsui Chemicals, Inc. Benzamide derivatives, useful as cell differentiation inducers
WO2001018171A2 (en) 1999-09-08 2001-03-15 Sloan-Kettering Institute For Cancer Research Novel class of cytodifferentiating agents and histone deacetylase inhibitors, and methods of use thereof
WO2001070675A2 (en) 2000-03-24 2001-09-27 Methylgene, Inc. Inhibitors of histone deacetylase
WO2002026696A1 (en) 2000-09-29 2002-04-04 Prolifix Limited Carbamic acid compounds comprising an amide linkage as hdac inhibitors
US20020103192A1 (en) 2000-10-26 2002-08-01 Curtin Michael L. Inhibitors of histone deacetylase
WO2005021553A1 (en) * 2003-08-29 2005-03-10 Pfizer Inc. Naphthalene carboxamides and their derivatives useful as new anti-angiogenic agents
US7041687B2 (en) 2002-01-25 2006-05-09 Vertex Pharmaceuticals Incorporated Indazole compounds useful as protein kinase inhibitors
US7098330B2 (en) 2000-09-15 2006-08-29 Vertex Pharmaceuticals Incorporated Pyrazolylamine substituted quinazoline compounds useful as protein kinase inhibitors
US7125905B2 (en) 2000-02-15 2006-10-24 Agouron Pharmaceuticals, Inc. Pyrrole substituted 2-indolinone protein kinase inhibitors
US7132533B2 (en) 2003-01-09 2006-11-07 Agouron Pharmaceuticals, Inc. Tricyclic compounds protein kinase inhibitors for enhancing the efficacy of anti-neoplastic agents and radiation therapy
US7151096B2 (en) 2003-03-05 2006-12-19 Irm Llc Cyclic compounds and compositions as protein kinase inhibitors
US7157476B2 (en) 2003-08-20 2007-01-02 Vertex Pharmaceuticals Incorporated Aminofurazan compounds useful as protein kinase inhibitors
US7166597B2 (en) 2000-08-22 2007-01-23 Glaxo Group Limited Fused pyrazole derivatives being protein kinase inhibitors
US7179910B2 (en) 2001-02-15 2007-02-20 Agouron Pharmaceuticals, Inc. 3-(4-amidopyrrol-2-ylmethlidene)-2-indolinone derivatives as protein kinase inhibitors
US7189721B2 (en) 1997-05-07 2007-03-13 Sugen Inc. Bicyclic protein kinase inhibitors
CN1933839A (zh) * 2004-01-23 2007-03-21 安进公司 化合物和使用方法
US7214700B2 (en) 2000-05-02 2007-05-08 Sugen Inc. (2-Oxindol-3-ylidenyl) acetic acid derivatives and their use as protein kinase inhibitors

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4405602B2 (ja) * 1998-04-16 2010-01-27 バイエル・シエーリング・ファーマ アクチエンゲゼルシャフト ヒストン脱アセチル化酵素阻害剤
CN101851173A (zh) * 2001-09-14 2010-10-06 梅特希尔基因公司 组蛋白脱乙酰化酶抑制剂
TWI319387B (en) * 2002-04-05 2010-01-11 Astrazeneca Ab Benzamide derivatives
EP2060565A1 (en) * 2007-11-16 2009-05-20 4Sc Ag Novel bifunctional compounds which inhibit protein kinases and histone deacetylases

Patent Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0847992A1 (en) 1996-09-30 1998-06-17 Mitsui Chemicals, Inc. Benzamide derivatives, useful as cell differentiation inducers
US7189721B2 (en) 1997-05-07 2007-03-13 Sugen Inc. Bicyclic protein kinase inhibitors
WO2001018171A2 (en) 1999-09-08 2001-03-15 Sloan-Kettering Institute For Cancer Research Novel class of cytodifferentiating agents and histone deacetylase inhibitors, and methods of use thereof
US7125905B2 (en) 2000-02-15 2006-10-24 Agouron Pharmaceuticals, Inc. Pyrrole substituted 2-indolinone protein kinase inhibitors
WO2001070675A2 (en) 2000-03-24 2001-09-27 Methylgene, Inc. Inhibitors of histone deacetylase
US7214700B2 (en) 2000-05-02 2007-05-08 Sugen Inc. (2-Oxindol-3-ylidenyl) acetic acid derivatives and their use as protein kinase inhibitors
US7166597B2 (en) 2000-08-22 2007-01-23 Glaxo Group Limited Fused pyrazole derivatives being protein kinase inhibitors
US7098330B2 (en) 2000-09-15 2006-08-29 Vertex Pharmaceuticals Incorporated Pyrazolylamine substituted quinazoline compounds useful as protein kinase inhibitors
US7115739B2 (en) 2000-09-15 2006-10-03 Vertex Pharmaceuticals Incorporated Triazole compounds useful as protein kinase inhibitors
WO2002026696A1 (en) 2000-09-29 2002-04-04 Prolifix Limited Carbamic acid compounds comprising an amide linkage as hdac inhibitors
US20020103192A1 (en) 2000-10-26 2002-08-01 Curtin Michael L. Inhibitors of histone deacetylase
US7179910B2 (en) 2001-02-15 2007-02-20 Agouron Pharmaceuticals, Inc. 3-(4-amidopyrrol-2-ylmethlidene)-2-indolinone derivatives as protein kinase inhibitors
US7041687B2 (en) 2002-01-25 2006-05-09 Vertex Pharmaceuticals Incorporated Indazole compounds useful as protein kinase inhibitors
US7132533B2 (en) 2003-01-09 2006-11-07 Agouron Pharmaceuticals, Inc. Tricyclic compounds protein kinase inhibitors for enhancing the efficacy of anti-neoplastic agents and radiation therapy
US7151096B2 (en) 2003-03-05 2006-12-19 Irm Llc Cyclic compounds and compositions as protein kinase inhibitors
US7157476B2 (en) 2003-08-20 2007-01-02 Vertex Pharmaceuticals Incorporated Aminofurazan compounds useful as protein kinase inhibitors
WO2005021553A1 (en) * 2003-08-29 2005-03-10 Pfizer Inc. Naphthalene carboxamides and their derivatives useful as new anti-angiogenic agents
CN1933839A (zh) * 2004-01-23 2007-03-21 安进公司 化合物和使用方法

Non-Patent Citations (37)

* Cited by examiner, † Cited by third party
Title
ARTEAGA, C-L., CURR. OPIN. ONCOL., vol. 6, 2001, pages 491 - 498
AVILA AM ET AL., J CLINIC INVESTIGATION, vol. 117, no. 3, 2007, pages 659 - 71
BARTL. S., MOL. CELL BIOL., vol. 17, 1997, pages 5033 - 5043
DE BORE J, TISSUE ENG., vol. 12, no. 10, 2006, pages 2927 - 37
DOKMANOVIC, M., J. CELL BIOCHENM., vol. 96, 2005, pages 293 - 304
DU, JUAN ET AL.: "Molecular modeling studies of vascular endothelial growth factor receptor tyrosine kinase inhibitors using QSAR and docking.", JOURNAL OF MOLECULAR GRAPHICS AND MODELLING, vol. 27, 5 November 2008 (2008-11-05), pages 642 - 654, XP025815659 *
GIALITAKIS M ET AL., NUCLEIC ACIDS RES., vol. 34, no. 1, 2007, pages 765 - 72
GLAROS S ET AL., ONCOGENE, 4 June 2007 (2007-06-04)
GLASER, K-B., BIOCHEM. BIOPHYS. RES. COMM., vol. 310, 2003, pages 529 - 536
GRIGNANI, F., NATURE, vol. 391, 1998, pages 815 - 818
GRUNSTEIN, M., NATURE, vol. 389, 1997, pages 349 - 352
HAQ ET AL., J. CELL BIOL., vol. 151, 2000, pages 117
HARMANGE, JEAN-CHRISTOPHE ET AL.: "Naphthamides as novel and potent vascular endothelial growth factor receptor tyrosine kinase inhibitors: design, synthesis, and evaluation.", J. MED. CHEM., vol. 51, 2008, pages 1649 - 1667, XP002668409 *
HEALY, A., J. IMMUNOL., vol. 177, 2006, pages 1886 - 1893
HERMAN D ET AL., NATURE CHEMICAL BIOLOGY, vol. 2, no. 10, 2006, pages 551 - 8
KIM, J. ET AL., J. CLIN. INVEST., vol. 114, 2004, pages 823 - 827
KIMURA ET AL., J. BIOL. CHEM., vol. 272, 1997, pages 13766 - 13771
LAGGER, G., EMBO J., vol. 21, 2002, pages 2672 - 2681
LANGLEY B ET AL., CURRENT DRUG TARGETS-CNS & NEUROLOGICAL DISORDERS, vol. 4, 2005, pages 41 - 50
LIN, R-J., NATURE, vol. 391, 1998, pages 811 - 814
MAI, A ET AL., INT J. BIOCHEM CELL BIO.
MARKS, P-A., NATURE REVIEWS CANCER, vol. 1, 2001, pages 194
MENDELSOHN, J., CLIN. CAN. RES., vol. 3, 1997, pages 2707 - 2707
MONIA ET AL., NAT. MED., vol. 2, 1996, pages 668 - 675
PARIKH, A-A., HEMATOL. ONCOL. CLIN. N. AM., vol. 18, 2004, pages 951 - 971
PETIT, A-M. ET AL., AM. J. PATHOL., vol. 151, 1997, pages 1523 - 1530
RUIJTER, A-J-M., BIOCHEM. J., vol. 370, 2003, pages 737 - 749
SAKUMA, T, INT. J. ONCOL., vol. 29, 2006, pages 117 - 124
SALEK-ARDAKANI, S. ET AL., J. IMMUNOL., vol. 175, 2005, pages 7635 - 7641
SCHUMACHER ET AL., J. CELL BIOL., vol. 143, 1998, pages 1635 - 1646
See also references of EP2439195A4
TAN, S-L., J. IMMUNOL., vol. 176, 2006, pages 2872 - 2879
TRIVEDI, C-M., NAT. MED, vol. 13, 2007, pages 324 - 331
VINCENT A. ET AL., ONCOGENE, 30 April 2007 (2007-04-30)
WIKSTRAND, C-J. ET AL., J NATL CANCER INST., vol. 90, 1998, pages 799 - 800
WIKSTRAND, C-J., J NATL CANCER INST., vol. 90, 1998, pages 799 - 800
WILSON, A-J., J BIOL. CHEM., vol. 281, 2006, pages 13548 - 13558

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103351336A (zh) * 2013-06-28 2013-10-16 华侨大学 作为蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘甲酰胺衍生物及其制备方法
CN103896836A (zh) * 2014-03-20 2014-07-02 华侨大学 N-苯基-6-[(7-氯喹啉-4-氧基)酚醚]-2-萘甲酰胺及其制备方法
CN103923004A (zh) * 2014-04-04 2014-07-16 华侨大学 一种萘甲酰胺衍生物及其制备和应用
CN104030980A (zh) * 2014-06-10 2014-09-10 华侨大学 N-(3-甲氧基-4-氯苯基)-4-[(7-氯-4-喹啉)氨基]苯甲酰胺及其制备和应用
WO2018187894A1 (zh) * 2017-04-10 2018-10-18 师健友 一种用于***疾病以及具有抗菌、抗病毒和抗炎作用的药物
RU2784869C1 (ru) * 2019-03-25 2022-11-30 Шэньчжэнь Чипскрин Байосайенсиз Ко., Лтд. Чиаураниб для лечения мелкоклеточного рака легкого
WO2022247844A1 (zh) * 2021-05-26 2022-12-01 深圳微芯生物科技股份有限公司 包含蛋白激酶抑制剂的药物组合物及其医药用途

Also Published As

Publication number Publication date
CN101906076B (zh) 2013-03-13
EP2439195A4 (en) 2012-10-31
AU2010256246B2 (en) 2013-04-11
SI2439195T1 (sl) 2014-12-31
BRPI1011994B8 (pt) 2021-05-25
AU2010256246A1 (en) 2012-01-12
BRPI1011994B1 (pt) 2020-04-07
WO2010139180A8 (zh) 2012-01-05
ZA201109030B (en) 2013-02-27
JP5484568B2 (ja) 2014-05-07
KR20120016659A (ko) 2012-02-24
AU2010256246B9 (en) 2014-01-30
DK2439195T3 (da) 2014-09-22
UA103092C2 (ru) 2013-09-10
BRPI1011994A2 (pt) 2016-05-10
PL2439195T3 (pl) 2015-02-27
HRP20140717T1 (hr) 2014-11-21
CA2763822C (en) 2014-12-09
CA2763822A1 (en) 2010-12-09
RU2497809C2 (ru) 2013-11-10
EP2439195A1 (en) 2012-04-11
CN101906076A (zh) 2010-12-08
JP2012528800A (ja) 2012-11-15
MX2011012752A (es) 2012-03-07
PT2439195E (pt) 2014-09-10
KR101421786B1 (ko) 2014-07-22
ES2509615T3 (es) 2014-10-17
EP2439195B1 (en) 2014-07-16
KR20140014313A (ko) 2014-02-05

Similar Documents

Publication Publication Date Title
WO2010139180A1 (zh) 作为蛋白激酶抑制剂和组蛋白去乙酰化酶抑制剂的萘酰胺衍生物、其制备方法及应用
US8211901B2 (en) Naphthamide derivatives as multi-target protein kinase inhibitors and histone deacetylase inhibitors
WO2017101803A1 (zh) 一种新型egfr和alk激酶的双重抑制剂
CN107531633B (zh) 5-芳香炔基取代的苯甲酰胺类化合物及其制备方法、药物组合物和用途
US9353062B2 (en) Substituted quinolines as bruton&#39;s tyrosine kinases inhibitors
CN111051300B (zh) 作为组蛋白脱乙酰基酶1和/或2(hdac1-2)的选择性抑制剂的新杂芳基酰胺衍生物
JPWO2006104161A1 (ja) c−Met自己リン酸化阻害作用を有するチエノピリジン誘導体、キノリン誘導体、およびキナゾリン誘導体
JP6609308B2 (ja) キナーゼ阻害剤としての置換マクロサイクル
JP6916562B2 (ja) 化合物、その薬学的に許容される塩、溶媒和物、立体異性体及び互変異性体、並びに薬物組成物、過剰増殖性障害治療剤、過剰増殖性障害予防剤、薬物、癌治療剤、癌予防剤、及びキナーゼシグナル伝達調節剤
JP7345901B2 (ja) キナーゼ阻害剤として有用な置換大員環類
CN110563697A (zh) 2-吡啶甲酰胺类化合物的制备及应用
Mularski et al. The p53 stabilizing agent CP-31398 and multi-kinase inhibitors. Designing, synthesizing and screening of styrylquinazoline series
WO2020221006A1 (zh) 一种bet蛋白抑制剂、其制备方法及用途
CN107151233B (zh) 含腙的嘧啶类衍生物及其用途
CN107311933B (zh) 一类苯并咪唑衍生物,及其制备方法和用途
WO2009014941A1 (en) 3-(4-amidopyrrol-2-ylmethlidene)-2-indolinone derivatives as multi-target protein kinase inhibitors and histone deacetylase inhibitors
US20200222367A1 (en) Xpa inhibitor compounds and their use
US20230000876A1 (en) Treating cancers with a cyclin-dependent kinase inhibitor
US11801243B2 (en) Bromodomain inhibitors for androgen receptor-driven cancers
US8158656B2 (en) 2-indolinone derivatives as multi-target protein kinase inhibitors and histone deacetylase inhibitors
WO2014190872A1 (zh) 一种含硒化合物及其医药用途
JP2011520891A (ja) 強力かつ選択的なヒストン脱アセチル化酵素阻害剤としての6−アミノニコチンアミド誘導体
Deng et al. Design, Synthesis and Biological Evaluation of 5-Amino-1h-Pyrazole-4-Carboxamide Derivatives as Pan-Fgfr Covalent Inhibitors

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10782884

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2763822

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2012513451

Country of ref document: JP

Ref document number: MX/A/2011/012752

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2010256246

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2010782884

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: A201115118

Country of ref document: UA

ENP Entry into the national phase

Ref document number: 20117030659

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 7/CHENP/2012

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2011152113

Country of ref document: RU

ENP Entry into the national phase

Ref document number: 2010256246

Country of ref document: AU

Date of ref document: 20100305

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI1011994

Country of ref document: BR

ENP Entry into the national phase

Ref document number: PI1011994

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20111130