WO2013107225A1 - N-((4-氯-3-三氟甲基)苯基)-n'-(2-氟-4-((2-羟甲基氨基甲酰基)-4-吡啶基氧)苯基)脲及其作为抗癌药物的应用 - Google Patents

N-((4-氯-3-三氟甲基)苯基)-n'-(2-氟-4-((2-羟甲基氨基甲酰基)-4-吡啶基氧)苯基)脲及其作为抗癌药物的应用 Download PDF

Info

Publication number
WO2013107225A1
WO2013107225A1 PCT/CN2012/085791 CN2012085791W WO2013107225A1 WO 2013107225 A1 WO2013107225 A1 WO 2013107225A1 CN 2012085791 W CN2012085791 W CN 2012085791W WO 2013107225 A1 WO2013107225 A1 WO 2013107225A1
Authority
WO
WIPO (PCT)
Prior art keywords
acid
cancer
phenyl
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/CN2012/085791
Other languages
English (en)
French (fr)
Inventor
张世喜
方垂
谭玉婷
Original Assignee
湖南有色凯铂生物药业有限公司
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
Application filed by 湖南有色凯铂生物药业有限公司 filed Critical 湖南有色凯铂生物药业有限公司
Publication of WO2013107225A1 publication Critical patent/WO2013107225A1/zh

Links

Classifications

    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen 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
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides

Definitions

  • the present invention relates to N-((4-chloro-3-trifluoromethyl)phenyl)-N,- which is used as a kinase inhibitor such as VEGFR-2.
  • the signal begins with the binding of epidermal growth factor (EGF) to the epidermal growth factor receptor (EGFR) on the cell membrane, and the EGFR at its phosphorylation site SH2 and the adaptor protein GRB2 (growth factor) Receptor-bound protein 2) Binding, recruitment of guanylate exchange factor SOS protein and Ras binding to adjacent inner cell membrane, GTP replaces Ras-bound GDP and stimulates Ras, Ras is one of the key signal transduction centers, Multiple upstream and downstream signal transductions.
  • EGF epidermal growth factor
  • EGFR epidermal growth factor receptor
  • the intensified Ras binds to the cellular paddle/threonine kinase Raf, Raf rephosphorylates MAPK kinase (MAPKK, also known as MEK), and MEK activates ERK1/2 (extracel lular signal regulated kinase, ERK). After activation, ERK enters the nucleus and directly activates transcription factors to initiate biological processes such as cell growth, differentiation and apoptosis. Once this pathway is over-activated, accelerated cell proliferation and prolonged cell survival can lead to tumor formation and development (Kolch W. Biochem J, 2000, 351: 289-305; Gi shizky ML. Annu. Rep. Med. Chem. 1995, 30: 247-253).
  • the signaling proteins in the signaling pathway cascade may be activated or inhibited by different upstream proteins to form an extremely complex network regulatory structure.
  • Raf and ERK on the Raf/MEK/ERK signaling pathway can also exert their signal transduction regulation in a Ras-independent manner; Ras activity can also be increased by a decrease in GTP hydrolysis caused by mutation of the tumor suppressor gene NF1. .
  • epidermal growth factor receptor EGFR
  • PI3K phosphatidylinositol 3-kinase
  • Ras and PI3K together regulate molecular lactation
  • mTOR animal rapamycin target protein
  • S6 kinase and 4EBP1 are phosphorylated by mTOR to initiate ribosomal protein synthesis (Hay N, Sonenberg N) (2004) . Genes Dev 18 (16): 1926 _ 45 ).
  • the protein kinase gene co-expresses more than 518 protein kinases, of which the receptor kinase has 20 subfamilies of 58 types, and the cell paddle kinase has 10 subfamilies of 32 types; among the more than 50 known oncogenes, most of them are protein kinases. Gene (Manning et al. Sicence, 2002, 298, 1912).
  • Cancer genes are usually in a state of inhibition, but when they are mutated or abnormally activated, such as chemical poisons, long-term ultraviolet light, or radioactive substances, oncogenes overexpress kinases, or express persistently activated kinases, resulting in Cells that are out of control, such as cell growth, differentiation, and apoptosis, produce cancer cells (Croce CM. N Engl J Med. 2008, 358 (5): 502 - 11 ).
  • Raf kinases in the Raf/MEK/ERK signaling pathway are overexpressed in many cancer cells, and more than 60% of human malignant melanoma, 12% of colorectal cancer, and 14% of ovarian cancers have activating mutations in B-raf. This mutation is also present in tumors of the thyroid and lungs; on the other hand, there is abnormally high C-raf activity in 50% of kidney cancers and almost 100% of liver cancer cells, although C-raf does not have mutations. (Brose, MS et al. Cancer Res. 2000, 63: 6997-7000; Davies, H. Nature, 2002, 417: 949-954; Yuen, ST et al. Cancer Res. 2002, 62: 6451-6455).
  • the B-raf mutant kinase B-raf-V600E which is 500 times more active than wild-type kinase, forms a heterodimer with C_raf and continues to activate downstream ERK and protein kinase mTOR (Sridhar SS, Hedley D, Siu LL. Mol. Cancer Ther. 2005, 4 (4) : 677 - 85 ).
  • RAF kinase There are three subtypes of RAF kinase, including A-raf, B-raf and C-raf (also known as Raf-1), which are highly homologous and enjoy high sequences.
  • Sorafenib developed by Bayer, was the first treatment of advanced kidney approved in 2005.
  • the cancer RAF kinase inhibitor was approved in 2007 as a treatment for metastatic liver cancer.
  • sorafenib actually antagonizes multiple kinases, which inhibit cancer cell growth by simultaneously antagonizing RAF kinase and several angiogenic growth factor receptors (eg, VEGFR, PDGFR, etc.). And transfer (Keating GM, Santoro A. Drugs 2009, 69 (2): 223-240).
  • Renal cancer and liver cancer are highly metastatic cancers.
  • the main pathological type of renal cancer is clear cel l carcinoma, and its pathogenesis is related to mutation of VHL tumor suppressor gene.
  • VHL encoded amino acid protein
  • HIF-la hypoxia-inducible factor-la
  • HIF -loc re-stimulates the expression and synthesis of VEGF and PDGF- ⁇ and other cell growth factors, and participates in neovascularization of tumor tissues.
  • most kidney cancers have EGFR expression, and the degree of expression is related to prognosis.
  • HIF-lodl over-activated TGF-oc produces autocrine effect and binds to EGFR, thereby promoting cell proliferation and survival. Regardless of the combination of growth factors and their receptors, it is usually required to function through the Raf/MEK/ERK pathway (Gunaratnam L, et al. J Biol Chem. 2003, 278: 44966-44974).
  • VEGF vascular endothelial growth factor
  • VAGFR receptor for vascular endothelial growth factor
  • MMPs matrix metalloproteinases
  • Growth factors such as PDGF and FGF secreted by cancer cells help to form new blood vessels by recruiting and stimulating the growth of fibroblasts, smooth muscle cells, and adventitial cells to form new cancer cell vascular stroma.
  • this new tumor blood vessel tends to be irregular in structure, imperfect vascular matrix, and high permeability. Therefore, tumor cells can penetrate into the blood, spread and attach to other parts of the body to grow and proliferate to form metastasis. .
  • Clinical studies have shown that inhibiting this process can effectively prevent tumor growth and metastasis, and prolong patient life. (Folkman J. Scientific American 1996, 275 (3): 150 - 4).
  • VEGFR-1 Flt-1
  • VEGFR-2 KDR, or Flk_l
  • PDGF platelet-derived growth factor
  • FGF FGF receptor FGFR1-4, etc.
  • VEGFR-3 the structurally similar VEGFR-3 (Flt-4) receptor is mainly involved in the formation of lymphangiogenesis (Lymphangiogenesis), which plays an important role in the process of cancer metastasis through the lymphatic system. Role, these receptors can be used as targets against solid tumor growth and metastasis
  • imatinib inhibits cell growth and division by antagonizing over-expressed bcr-abl kinase in leukemia cancer cells; it also antagonizes c-kit kinase activity and clinically displays its gastrointestinal tract
  • the efficacy of stromal cell tumors has been approved as a therapeutic drug for gastrointestinal stromal tumors.
  • Bristol-Myers Squibb and Novartis developed two other chronic bone marrow cancer treatments, Dasatinib and Nilotinib, in 2006 and 2007, respectively. Patients who are fighting imatinib also have good results.
  • Gefitinib developed by AstraZeneca in the UK, was approved for marketing in Sakamoto in August 2002. It is a first-line treatment for non-small cell lung cancer. Gefitinib is especially effective for smoking non-small cell lung cancer in Asian men. . In 2004, Genentech and 0SI Pharmaceutical jointly developed erlotinib, which is more widely used as a first-line treatment, maintenance therapy and sequential in non-small cell lung cancer in seven or eighty countries around the world. Therapeutic drugs.
  • Both gefitinib and erlotinib are epidermal growth factor receptor (EGFR) inhibitors that antagonize the binding of EGFR receptors to epidermal growth factor (EGF) on the cell surface transmembrane and inhibit EGFR-mediated Raf/MEK/ Signal channels such as ERK (Raymond E, Faivre S, Armand J. Drugs 2000, 60 Suppl 1 : 15-23; discussion 41 - 2 ). Same The principle, erlotinib is also effective in inhibiting the growth of pancreatic tumor cells, becoming the first FDA-approved targeted drug for the treatment of pancreatic cancer.
  • EGFR epidermal growth factor receptor
  • Sunitinib As a small molecule inhibitor of tyrosine kinase for the treatment of renal cancer (RCC), sunitinib delays the growth and metastasis of cancer cells by antagonizing the vascular endothelial growth factor receptor (VGEFR) and inhibiting the formation of neovascularization in cancer cells. Sunitinib also inhibits the activity of c-kit kinase and is approved for the treatment of patients with gastrointestinal stromal tumors of imatinib.
  • VGEFR vascular endothelial growth factor receptor
  • Sorafenib developed by Bayer, fights cancer growth and metastasis by simultaneously antagonizing multiple pathways such as RAF kinase, vascular endothelial growth factor receptor (VGEFR) and platelet-derived growth factor receptor (PDGFR- ⁇ ).
  • VGEFR vascular endothelial growth factor receptor
  • PDGFR- ⁇ platelet-derived growth factor receptor
  • the FDA has approved the treatment of advanced kidney cancer and metastatic liver cancer.
  • the efficacy of sorafenib in metastatic liver cancer is a major breakthrough in the field of liver cancer treatment. Liver cancer is recognized as one of the most difficult and most metastatic cancers (Escudier B, et al. (January 2007). N. Engl. J Med. 356 (2): 125 - 34; Keating GM, Santoro A. Drugs 2009, 69 (2): 223 - 240).
  • the mammalian target of rapamycin is a multifunctional silk/threonine kinase in the PI3K/AKT signaling pathway and is directly associated with cancer cell growth, division, survival and metastasis ( Rubio-Viqueira, B , Hidalgo M. Curr. Op in. Invest ig. Drugs 2006, 7 : 501 - 512 ).
  • the role of mTOR kinase in renal cell metastasis is particularly pronounced. Uncontrolled mTOR causes an increase in the concentration of HIF-loc in cells. HIF-loc induces the synthesis of VGEF and promotes angiogenesis in cancer cells. On the other hand, many renal cancer cells are due to VHL.
  • HIF-loc Mutation or loss of tumor suppressor genes resulting in a decrease in HIF-loc decomposition also increases the concentration of HIF-loc (Thomas GV. et al. Nature Medicine 2006, 12: 122-127).
  • the first clinical mTOR inhibitor was Temsirol imus, a rapamycin derivative developed by Wyeth Pharmaceuticals of the United States. In 2007, it was approved by the FDA as a therapeutic drug for kidney cancer.
  • Lapatinib is a dual inhibitor of small molecule tyrosine kinase that inhibits both epidermal growth factor receptor (EGFR) and ErbB2 (HER-2/neu) receptors, with approximately 30% of breast cancers. The patient develops an excess of HER-2/neu receptor due to the HER-2/neu proto-oncogene.
  • EGFR epidermal growth factor receptor
  • HER-2/neu ErbB2
  • lapatinib was approved by the FDA as a combination therapy for breast cancer, and in 2010 it was approved as a first-line drug for ER+/EGFR+/HER2+ triple-positive breast cancer patients (Wood ER et al. Cancer research 64 (18):
  • kinases Due to the high homology and diversity of kinases, most of the small molecule anticancer drugs currently being developed are multi-kinase inhibitors. Fortunately, unlike previous concerns, lower kinase selectivity does not affect drugs. Therapeutic effects; in contrast, preferred multiple kinase inhibitors may be more conducive to the treatment of complex, diverse forms of cancer, and may be versatile.
  • macromolecular monoclonal antibodies are usually specific inhibitors targeting only a single target in the cell signaling pathway, and several clinical examples have demonstrated the efficacy of specific monoclonal antibodies in the treatment of cancer.
  • trastuzumab was approved by the US FDA for breast cancer treatment for breast cancer patients overexpressing HER-2/neu (Hudi s, CA. N Engl J Med. 2007, 357 ( 1) : 39 - 51 ).
  • Bevacizumab specifically binds to vascular endothelial growth factor (VGEF), inhibits the formation of cancerous blood vessels, and promotes the penetration of chemotherapeutic drugs into cancer tissues (Los M et al. The Oncologist 2007, 12 (4) : 443 - 50). Subsequently, panitumumab and cetuximab were also approved for metastatic colorectal cancer. Unlike bevacizumab, panitumumab and cetuximab inhibit the cell-growth signaling pathway by binding to the epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • Cetuximab (IgGl) and panitumumab (IgG2) belong to different subtypes of immunoglobulins, and they have subtle differences in anticancer mechanism.
  • the West produced by Squibb Topoxib can also treat head and neck cancer.
  • VEGF Vascular endothelial growth factor
  • Tumor blood vessels are highly sensitive to VEGF.
  • VEGF mRNA concentration is significantly higher than normal cells.
  • These tumors include lung cancer (Mattern et al Br. J Cancer 1996, 73, 93, 1), thyroid cancer (Vigl ietto et al. Oncogene 1995, 11, 1569), breast cancer (Brown et al. Human Pathol. 1995, 26, 86), gastrointestinal cancer ( Brown et al. Cancer Res. 1993, 53, 4727; Suzuki et al. Cancer Res. 1996, 56, 3004), Kidney cancer and bladder cancer (Brown et al. Am. JPalhol.
  • angiogenesis inhibitors such as VEGFR-2 and PDGFR- ⁇ inhibitors
  • Anticancer drugs have become a hot area and have yielded new, promising clinical trial results.
  • W0-2004007458 discloses a group of 2 amidamine nicotinamide derivatives as VEGFR, PDGFR and Kit inhibitors for the treatment of non-squamous non-small cell lung cancer in clinical phase III trials, other indications including parenchymal cancer, gastrointestinal Cancer, colorectal cancer, endocrine cancer, breast cancer and lung cancer.
  • W0-2004113304 discloses a group of carbazole, benzisoxazole and benzothiazole derivatives as CSFR-PDGFR, Flt3, Kit, VEGFR-1, VEGFR-2, VEGFR-3 inhibitors, used in clinical phase III trials.
  • the second phase is used for non-small cell lung cancer, breast cancer, colorectal cancer, and other indications include kidney cancer, acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS).
  • AML acute myeloid leukemia
  • MDS myelodysplastic syndrome
  • W0-2000043366 discloses a group of quinoline-urea derivatives as Kit, PDGFR family receptors, VEGFR-K VEGFR-2, VEGFR-3 inhibitors, for the treatment of renal cancer in a clinical phase III trial, and other clinical trials including Breast cancer, colorectal cancer, gastrointestinal cancer, non-small cell lung cancer and liver cancer.
  • W0-200102369 discloses a group of carbazole derivatives as CSFR_1, PDGF, VEGF_1, VEGF-2 and VEGF-3 inhibitors for the treatment of renal cancer in clinical phase III trials, phase II clinical trials including mesothelioma, blood vessels Sarcoma, adrenal cortical adenocarcinoma and liver cancer.
  • W0-200232872 discloses a novel group of quinoline-urea derivatives as VEGFR_2, VEGFR-3, FGFR 1-4 and RET inhibitors for the treatment of thyroid cancer in a clinical phase III trial, and a phase II clinical trial including liver cancer. Endometrial cancer, melanoma, kidney cancer and glioma.
  • W0-2003082272 discloses a group of arylamine benzimidazoles as Raf, VEGFR-2, PDGFR- ⁇ and Kit inhibitors for the treatment of melanoma.
  • the present invention relates to N-((4-chloro-3-trifluoromethyl)phenyl)-N,-(2-fluoro-4-((2-hydroxymethylcarbamoyl)-4-pyridyloxy) a phenyl) urea or a pharmaceutically acceptable salt thereof, and a method of treating a patient, the method comprising administering to the patient an effective amount of N-((4-chloro-3-trifluoromethyl)benzene which inhibits VEGFR-2 kinase base) - ⁇ '-(2-Fluoro-4-((2-hydroxymethylcarbamoyl)-4-pyridyloxy)phenyl)urea or a pharmaceutically acceptable salt thereof.
  • the present invention relates to ⁇ -((4-chloro-3-trifluoromethyl)phenyl)-indole,-(2-fluoro-4-((2-hydroxymethylcarbamoyl)-4-pyridyloxy) a phenyl) urea or a pharmaceutically acceptable salt thereof, selected from the group consisting of:
  • an acid salt of a cationic organic and inorganic base selected from the group consisting of an alkali metal cation, an alkaline earth metal cation, an ammonium ion, an aliphatic substituted ammonium ion or an aromatic substituted ammonium ion.
  • the pharmaceutical composition for treating cancer of the invention comprises N-((4-chloro-3-trifluoromethyl)phenyl)-N,-(2-fluoro-4-((2-hydroxymethylamino) Acyl: --4-pyridyloxy)phenyl)urea or a pharmaceutically acceptable salt thereof, and a physiologically suitable carrier.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can be used for the manufacture of a medicament for inhibiting VEGFR-2 kinase-mediated cancer cell growth.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof can also be used for the preparation of a medicament for treating cancer diseases.
  • the cancer of the present invention includes parenchymal cancer, kidney cancer, lung cancer, breast cancer, liver cancer, ovarian cancer, pancreatic cancer, thyroid cancer, bladder cancer, leukemia, melanoma, gastric cancer, colorectal cancer, endocrine cancer, acute myeloid Leukemia (AML:), myelodysplastic syndrome (MDS:), mesothelioma, angiosarcoma, adrenal cortical adenocarcinoma, endometrial cancer, and glioma.
  • AML acute myeloid Leukemia
  • MDS myelodysplastic syndrome
  • mesothelioma mesothelioma
  • angiosarcoma angiosarcoma
  • endometrial cancer endometrial cancer
  • glioma Especially for the treatment of kidney cancer, liver cancer, lung cancer, colorectal cancer, stomach cancer, breast cancer and angiosarcoma.
  • the pharmaceutically acceptable salt is preferably N-((4-chloro-3-trifluoromethyl)phenyl)-N,-(2-fluoro-4-((2-hydroxymethylcarbamoyl):-4)
  • a pharmaceutically acceptable acid addition salt of pyridyloxy)phenyl)urea is preferably N-((4-chloro-3-trifluoromethyl)phenyl)-N,-(2-fluoro-4-((2-hydroxymethylcarbamoyl):-4)
  • a pharmaceutically acceptable acid addition salt of pyridyloxy)phenyl)urea for example, an acid addition salt, preferably an acid addition salt with an organic or inorganic acid, preferably a pharmaceutically acceptable salt.
  • Suitable inorganic acids are, for example, hydrohalic acids such as hydrochloric acid, sulfuric acid or phosphoric acid.
  • Suitable organic acids are, for example, carboxylic acids, phosphonic acids, sulfonic acids or sulfamic acids, such as acetic acid, propionic acid, caprylic acid, capric acid, dodecanoic acid, glycolic acid, lactic acid, 2-hydroxybutyric acid, gluconic acid, Fumaric acid, succinic acid, adipic acid, pimelic acid, suberic acid, azelaic acid, malic acid, tartaric acid, citric acid, glucaric acid, galactose diacid, amino acid, such as glutamic acid, day Aspartic acid, N-methylglycine, acetaminoacetic acid, N-acetylaspartamide, N-acetylcysteine, pyruvic acid, acetoacetic acid, phosphoserine, 2- or 3-glycerophosphate, Malay Acid, hydroxymaleic acid, methyl maleic acid, cyclohexylcarboxy
  • the present invention relates to N-((4-chloro-3-trifluoromethyl)phenyl)-N,-(2-fluoro-4-((2-hydroxymethylamino)) which is used as a VEGFR-2 kinase inhibitor Acyl: )-4-pyridyloxy)phenyl)urea, and its use in the manufacture of a medicament for the treatment of a disease mediated by VEGFR-2 kinase.
  • the patient is a mammal, typically a human.
  • the compounds of the invention are useful for treating cancer in a mammal, preferably a human cancer, including but not limited to parenchymal, renal, lung, breast, liver, ovarian, pancreatic, thyroid, bladder, leukemia, melanoma, Gastric cancer, colorectal cancer, endocrine cancer, acute myeloid leukemia (AML:), myelodysplastic syndrome (MDS:), mesothelioma, angiosarcoma, adrenal cortical adenocarcinoma, endometrial cancer, and glioma.
  • the compounds of the invention are also useful in the treatment of inflammatory diseases including rheumatoid arthritis, retinopathy (including diabetic retinal neuropathy and macular degeneration), cardiovascular diseases and metabolic diseases.
  • Phenyl)urea or a pharmaceutically acceptable salt thereof has valuable pharmacological properties as described above.
  • a basic salt of an organic acid and an inorganic acid selected from the group consisting of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, methanesulfonic acid, trifluorosulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, 1-naphthalene Sulfonic acid, 2-naphthalenesulfonic acid, acetic acid, trifluoroacetic acid, malic acid, tartaric acid, citric acid, lactic acid, oxalic acid, succinic acid, fumaric acid, maleic acid, benzoic acid, salicylic acid, phenylacetic acid and almond Acid; and
  • an acid salt of a cationic organic and inorganic base selected from the group consisting of alkali metal cations, alkaline earth metal cations, ammonium ions, aliphatic substituted ammonium ions and aromatic substituted ammonium ions.
  • the pharmaceutically acceptable salt of oxy)phenyl)urea is a pharmaceutically acceptable acid addition salt.
  • acid addition salts preferably acid addition salts with organic or inorganic acids, especially pharmaceutically acceptable salts.
  • Suitable inorganic acids are selected from the group consisting of hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid and the like.
  • Suitable organic acids are selected from the group consisting of carboxylic acids, phosphonic acids, sulfonic acids or sulfamic acids, such as methanesulfonic acid, trifluorosulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, 1-naphthalenesulfonic acid, 2-naphthalenesulfonic acid, Acetic acid, trifluoroacetic acid, malic acid, tartaric acid, citric acid, lactic acid, oxalic acid, succinic acid, fumaric acid, maleic acid, benzoic acid, salicylic acid, phenylacetic acid, mandelic acid, propionic acid, octanoic acid, citric acid , dodecanoic acid, glycolic acid, 2-hydroxybutyric acid, gluconic acid, succinic acid, adipic acid, pimelic acid, suberic acid, azelaic acid, glucaric acid,
  • the compounds of the present invention may be administered alone or in combination with other anticancer drugs, such as compounds that inhibit tumor angiogenesis, such as protease inhibitors, epidermal growth factor receptor kinase inhibitors, vascular endothelial growth factor receptor kinase inhibitors, and the like; Toxic drugs, such as antimetabolites, such as anti-metabolites of purines and pyrimidine analogs; anti-mitotic drugs such as microtubule stabilizing drugs and anti-mitotic alkaloids; platinum coordination complexes; antitumor antibiotics; deuterated agents such as nitrogen mustard and Nitrosoureas; endocrines, such as adrenal corticosteroids, androgens, antiandrogens, estrogens, antiestrogens, aromatase inhibitors, gonadotropin releasing hormone agonists, and somatostatin Analogs, as well as compounds that target enzymes or receptors that are overexpressed and/or otherwise associated with specific metabolic pathways that are up-regulated in tumor cells, such as
  • the compounds of the invention may also be administered with radiation therapy, immunotherapy, surgery, or a combination thereof. Treatments for maintaining the state of a patient after tumor reduction or even chemopreventive treatment (e.g., in the case of a patient at risk:) are also possible.
  • the compounds of the invention are useful not only for the prophylactic and preferably therapeutic: treatment of humans, but also for the treatment of other warm-blooded animals, such as commercially useful warm-blooded animals, such as rodents such as mice, rabbits or Rat, or guinea pig.
  • the present invention also encompasses the inclusion of N-((4-chloro-3-trifluoromethyl)phenyl)-N,-(2-fluoro-4-((2-hydroxymethylcarbamoyl:)-4-pyridine)
  • a pharmaceutical composition of a base oxygen) phenyl) urea and a physiologically acceptable carrier is included.
  • the compounds of the invention may be administered by injection, inhalation or spraying or rectally, orally, dermally, parenterally, or in unit dosage form.
  • injectable administration includes intravenous, intramuscular, subcutaneous and parenteral injections, as well as the use of infusion techniques.
  • Skin administration includes topical or transdermal administration.
  • One or more compounds may be co-existed with one or more pharmaceutically acceptable non-toxic carriers, as well as other active ingredients as appropriate.
  • Oral compositions can be prepared according to any suitable method known in the art of pharmaceutical composition manufacture.
  • the composition may contain one or more of the following agents: diluents, sweeteners, perfumes, colorants and preservatives.
  • Tablets contain the active ingredients in admixture with pharmaceutically acceptable non-toxic excipients suitable for tablet manufacture.
  • the excipients are, for example, inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating agents and disintegrating agents such as corn starch or alginic acid; binders such as magnesium stearate, hard Fatty acid or talcum powder.
  • the tablets may be uncoated or they may be coated by known techniques to delay their disintegration and absorption in the gastrointestinal tract, providing a long lasting effect.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed.
  • the compounds can also be prepared in solid, immediate release form.
  • the oral preparation may also be a hard gelatin capsule in which the active ingredient is mixed with an inert solid diluent such as calcium carbonate, calcium phosphate or kaolin, or a soft gelatin capsule in which the active ingredient is mixed with water or, for example, peanut oil, liquid paraffin or olive Oil and other oils are mixed.
  • an inert solid diluent such as calcium carbonate, calcium phosphate or kaolin
  • a soft gelatin capsule in which the active ingredient is mixed with water or, for example, peanut oil, liquid paraffin or olive Oil and other oils are mixed.
  • Aqueous suspensions containing the active substance in admixture with excipients suitable for the manufacture of aqueous suspensions may also be employed.
  • the excipient is a suspending agent, such as sodium carboxymethylcellulose, methylcellulose, hydroxypropyl-methylcellulose, sodium alginate, polyvinylpyrrolidone, tragacanth and gum arabic;
  • the dispersing or wetting agent may be a natural phospholipid, such as lecithin, or a condensation product of an epoxy oxime with a fatty acid, such as polyoxyethylene stearate, or a condensation product of an epoxy oxime with a long chain fatty alcohol, for example A condensation product of heptahexylethylene hexadecanol, or an epoxy oxime with a partial ester of a fatty acid with hexitol, such as polyoxyethylene sorbitan monooleate.
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl or n-propyl p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents.
  • preservatives for example ethyl or n-propyl p-hydroxybenzoate
  • coloring agents for example, ethyl or n-propyl p-hydroxybenzoate
  • coloring agents for example, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents.
  • sweetening agents for example, sucrose or saccharin.
  • the active ingredient is mixed with a dispersing or wetting agent, a suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium tartrate
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium EDTA, sodium EDTA, sodium EDTA, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate, sodium bicarbonate
  • the form of the compound may also be a non-aqueous liquid preparation such as an oily suspension, which can be formulated by suspending the active ingredient in a vegetable oil such as peanut oil, olive oil, sesame oil or peanut oil or a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol.
  • a thickening agent such as beeswax, hard paraffin or cetyl alcohol.
  • an antioxidant such as ascorbic acid.
  • the pharmaceutical composition of the present invention may also be in the form of an oil-in-water emulsion.
  • the oil phase may be a vegetable oil such as olive oil or peanut oil or a mineral oil such as liquid beeswax, or a mixture thereof.
  • Suitable emulsifiers may be natural gums such as tragacanth and gum arabic, or natural phospholipids such as soy lecithin or lecithin; partial esters of fatty acids with anhydrohexitol, such as sorbitan oleate; A condensation product of a partial ester with epoxy oxime, such as polyoxyethylene sorbitan monooleate.
  • the emulsion may also contain Sweeteners and spices.
  • Sugar candies and tinctures can also be formulated with sweetening agents such as glycerin, polypropylene glycol, sorbitol or sucrose. Such preparations may also contain emollients, preservatives, and perfumes and colorants.
  • compositions of the invention are prepared in a manner known per se, for example by conventional mixing, granulating, shaping, dissolving or lyophilizing processes.
  • compositions Preference is given to using solutions of the active ingredient, in addition to suspensions or dispersions, in particular isotonic aqueous solutions, dispersions or suspensions, for example, lyophilized solutions containing only active substances or containing active substances with carriers such as mannitol
  • solutions of the active ingredient in addition to suspensions or dispersions, in particular isotonic aqueous solutions, dispersions or suspensions, for example, lyophilized solutions containing only active substances or containing active substances with carriers such as mannitol
  • the pharmaceutical compositions may be sterilized and/or contain excipients such as preservatives, stabilizers, wetting agents and/or emulsifiers, solubilizers, salts for regulating osmotic pressure and/or buffers, and are known per se
  • the method is prepared, for example, by a conventional dissolution or lyophilization method.
  • the solution or suspension may contain a viscosity increasing substance such as sodium carboxymethylcellulose, carboxymethyl
  • the suspension in oil contains vegetable oil, synthetic oil or semi-synthetic oil conventionally used for injection purposes as an oily component. Mention may be made, in particular, of liquid fatty acid esters comprising as long as a fatty acid component having from 8 to 22, in particular from 12 to 22, carbon atoms, for example lauric acid, tridecanoic acid, myristic acid, Pentadecanoic acid, palmitic acid, heptadecanoic acid, stearic acid, arachidic acid, didecanoic acid or the corresponding unsaturated acid, such as oleic acid, oleic acid, erucic acid, eicosanic acid or linoleic acid An antioxidant such as vitamin E, beta-carotene or 3,5-di-tert-butyl-4-hydroxytoluene is optionally added.
  • liquid fatty acid esters comprising as long as a fatty acid component having from 8 to 22, in particular from 12 to 22, carbon atoms, for example lauri
  • the alcohol component of these fatty acid esters has up to 6 carbon atoms and is a mono- or poly-hydric alcohol such as a mono-, di- or tri-alcohol such as methanol, ethanol, propanol, butanol or pentanol or Their isomers, but especially ethylene glycol and glycerol.
  • fatty acid esters which may be mentioned are: ethyl oleate, isopropyl myristate, isopropyl palmitate, etc., but especially vegetable oils such as cottonseed oil, almond oil, olive oil, sesame oil, Soybean oil and more especially peanut oil.
  • the preparation of the injectable compositions is carried out in a conventional manner under sterile conditions, and the introduction into a container such as an ampoule or vial and a container is also carried out under aseptic conditions in a conventional manner.
  • compositions for oral administration can be obtained, for example, by the following methods: mixing the active ingredient with one or more solid carriers, granulating the resulting mixture, if appropriate, processing the mixture or granules (if necessary, adding additional Excipients:) into the core of a tablet or dragee.
  • Suitable carriers are, in particular, fillers, for example sugars such as lactose, sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates such as tricalcium phosphate or calcium hydrogen phosphate, and also binders such as starches such as corn, Wheat, rice or potato starch, methylcellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone, and/or, if desired, a disintegrant, such as mentioned above Starch, as well as carboxymethyl starch, crosslinked polyvinylpyrrolidone, alginic acid or a salt thereof, such as sodium alginate.
  • fillers for example sugars such as lactose, sucrose, mannitol or sorbitol, cellulose preparations and/or calcium phosphates such as tricalcium phosphate or calcium hydrogen phosphate, and also binders such as starches such as corn, Wheat, rice or potato starch,
  • a fluidity regulator and a lubricant such as silicic acid, talc, stearic acid or a salt thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol or a derivative thereof.
  • a lubricant such as silicic acid, talc, stearic acid or a salt thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol or a derivative thereof.
  • the dragee core may have a suitable coating, optionally an enteric coating, especially a concentrated sugar solution, which may contain gum arabic, talc, polyvinylpyrrolidone, polyethylene glycol and/or titanium dioxide.
  • a coating solution in a suitable organic solvent or solvent mixture, or for the preparation of an enteric coating using a solution of a suitable cellulosic product, such as cellulose acetate phthalate or hydroxypropyl methylcellulose Phthalates.
  • Colorants or pigments may be added to the tablets or dragee coatings, for example for identification purposes or to show different dosages of the active ingredients.
  • compositions for oral administration are also hard gelatin capsules and soft, sealed capsules consisting of gelatin and a plasticizer such as glycerol or sorbitol.
  • the hard gelatine capsules may contain the active ingredient in the form of granules, for example, in the form of granules mixed with fillers such as corn starch, binders and/or glidants such as talc or magnesium stearate and optionally stabilizing agents.
  • the active ingredient is preferably dissolved or suspended in a suitable liquid vehicle such as a fatty oil, a paraffin oil or a liquid polyethylene glycol or a fatty acid ester of ethylene glycol or propylene glycol, as well as stabilizers and Detergents, such as polyoxyethylene sorbitan fatty acid ester detergents.
  • a suitable liquid vehicle such as a fatty oil, a paraffin oil or a liquid polyethylene glycol or a fatty acid ester of ethylene glycol or propylene glycol, as well as stabilizers and Detergents, such as polyoxyethylene sorbitan fatty acid ester detergents.
  • Suitable rectal-administered pharmaceutical compositions are, for example, suppositories which consist of a mixture of the active ingredient and a suppository base.
  • Suitable suppository bases are, for example, natural or synthetic triglycerides, streptammonium, polyethylene glycol or higher chain sterols.
  • aqueous solutions for example aqueous solutions of the active ingredient in the form of a water-soluble salt, or aqueous injection suspensions, containing a viscosity increasing substance such as sodium carboxymethylcellulose, sorbitol and/or are suitable. Or dextran, and if necessary, stabilizers.
  • the active ingredient optionally together with the excipient may also be in the form of a lyophilizate which may be brought into solution by the addition of a suitable solvent before parenteral administration.
  • Solutions for parenteral administration can also be used in the form of infusions.
  • Preferred preservatives are, for example, antioxidants such as ascorbic acid or microbicides such as sorbic acid or benzoic acid.
  • the daily oral dose is preferably from 0.01 to 200 mg/kg body weight.
  • Injections including intravenous, intramuscular, subcutaneous and parenteral injections, and application of the input technique, are preferably administered at a dose of 0.01 to 200 mg/kg body weight per day.
  • the daily dose for rectal administration is preferably from 0.01 to 200 mg/kg body weight.
  • the daily dose for external use is 1 to 4 times a day, preferably 0.1-200 mg per time.
  • the daily dose for inhalation is preferably 0.01-10 mg/kg body weight.
  • the optimal treatment regimen ie, the treatment regimen within a certain number of days, and N-((4-chloro-3-trifluoromethyl)phenyl)-N,-(2-fluoro-4-
  • the number of daily administrations of ((2-hydroxymethylcarbamoyl)-4-pyridyloxy)phenyl)urea or a pharmaceutically acceptable salt thereof can be determined by a person skilled in the art using conventional therapeutic tests.
  • the specific record level of a particular patient depends on a number of factors, including the activity of the particular compound used, the age of the patient, the patient's weight, the overall health of the patient, the patient's gender, the patient's diet, the time of administration, the route of administration, the rate of discharge, the drug The combination and severity of the current condition.
  • the compound of the present invention can be produced by a known compound (or a starting material which can be obtained from a known compound:), for example, by a general method as described later.
  • the activity of each compound known to VEGFR-2 kinase can be determined by a usual method described later.
  • the following examples are illustrative only and not limiting.
  • the biological activity of the compound of the present invention for inhibiting tumor growth was measured by the following method.
  • HepG2 tumor tissue of human liver cancer was cut into 1.5 mm 3 in the vigorous growth stage, and inoculated subcutaneously in the right axilla of nude mice under aseptic conditions.
  • the nude mice xenografts were measured for the diameter of the transplanted tumor using a vernier caliper, and the animals were randomly divided into groups after the tumors were grown to 60 to 200 mm 3 .
  • Dynamic observation of the test using the method of measuring the diameter of the tumor Anti-tumor effect. The number of tumor diameters measured is 3 times a week, and each measurement is also required to weigh the mouse.
  • the reference drug docetaxel injection was administered intraperitoneally at a dose of 30 mg/kg, once a week.
  • N-((4-chloro-3-trifluoromethyl)phenyl)-N,-(2-fluoro-4-((2-carbamoyl)-4-pyridyloxy)phenyl)urea (lg And paraformaldehyde (320 mg) was dissolved in 16 mL of tetrahydrofuran and 8 mL of water, and 20 mg of potassium carbonate was added thereto, and the mixture was stirred at 30 ° C under microwave for 30 min.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Epidemiology (AREA)
  • Hematology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本发明涉及一种用作VEGFR-2激酶抑制剂的 N-((4-氯-3-三氟甲基)苯基)-N'(2-氟-4-((2-羟甲基氨基甲酰基)-4-吡啶基氧)苯基)脲,及其用于制备治疗VEGFR-2所介导疾病的药物中的用途。

Description

N- ( (4-氯 -3-三氟甲基)苯基) - ( 2-氟 -4- ( ( 2-羟甲基氨基甲酰基) -4- 吡啶基氧) 苯基) 脲及其作为抗癌药物的应用 本申请要求于 2012年 1月 17日提交中国专利局,申请号为 201210012640.5, 发明名称为 "N- ( (4-氯 -3-三氟甲基) 苯基) -N,- (2-氟 -4- ( (2-羟甲基氨基甲酰 基 )-4-吡啶基氧)苯基)脲及其作为抗癌药物的应用"的中国专利申请的优先权, 其全部内容通过引用结合在本申请中。 发明领域
本发明涉及用作 VEGFR-2等激酶抑制剂的 N- ( (4-氯 -3-三氟甲基)苯基) -N,-
(2-氟 -4- ( (2-羟甲基氨基甲酰基: )-4-吡啶基氧) 苯基) 脲, 及其用于制备治疗 VEGFR-2等激酶所介导疾病的药物中的用途。
背景技术 随着对肿瘤分子生物学的深入研究, 通过对多种致癌蛋白激酶进行抑制的靶 向治疗法已成为抗肿瘤治疗的重要组成部分, 并已在各类肿瘤的治疗中获得了 重大进展。 在细胞中, 蛋白激酶通过对上游蛋白、 下游蛋白及自身的磷酸化来 传达及放大信息, 控制细胞的生长、 分化和凋亡。 目前已知, 所有真核细胞中 均存在 Raf/MEK/ERK这一信号通路, 其通过 Ras、 Raf、 MEK及 ERK的特异性级 联磷酸化将信号由细胞外传入细胞核内,大约 30%的肿瘤细胞存在这一通路的上 调。 在 Raf/MEK/ERK信号通路中, 信号起始于表皮生长因子 (EGF) 与细胞膜上 表皮生长因子受体 (EGFR) 的结合, EGFR在其磷酸化的位置 SH2部位与衔接蛋 白质 GRB2 (growth factor receptor-bound protein 2 ) 结合, 招募鸟苷酸交 换因子 S0S蛋白与 Ras结合在相邻的内细胞膜上, GTP替代与 Ras结合的 GDP而 激化 Ras , Ras是关键的信号转导中心之一, 参入多个上游和下游的信号转导。 在研究得最多的通路中, 激化后的 Ras与细胞桨丝 /苏氨酸激酶 Raf 结合, Raf 再磷酸化 MAPK激酶(MAPKK,又称 MEK), MEK激活 ERK1/2 (extracel lular signal regulated kinase, ERK) 。 ERK被激活后, 进入细胞核内并直接激活转录因子, 启动细胞的生长、 分化和凋亡等生物学过程。 一旦该通路发生过度的激活, 细 胞增殖的加速与细胞生存期的延长可导致肿瘤的形成及发展(Kolch W. Biochem J, 2000, 351 : 289-305; Gi shizky ML. Annu. Rep. Med. Chem. 1995, 30 : 247-253)。
需要指出的是, 随环境因素的不同, 信号通路级联反应中的信号蛋白都可 能被不同的上游蛋白激活或抑制而形成一个极其复杂的网络调控结构。 例如, Raf/MEK/ERK信号通路上的 Raf和 ERK也可通过不依赖 Ras的方式发挥其信号转 导调节作用; Ras的活性也可以通过肿瘤抑制基因 NF1的突变而引起 GTP水解的 减少来提高。 除了在 Raf/MEK/ERK信号通路上的关键作用外, 表皮生长因子受 体(EGFR)还激活 PI3K/AKT通道上的磷脂酰肌醇 3-激酶 (PI3K) , Ras和 PI3K 再一起调控分子哺乳动物雷帕霉素靶蛋白 (mTOR) 的作用。 mTOR是控制细胞生 理作用的另一多功能细胞桨丝 /苏氨酸激酶, 作为下游的底物, S6 kinase (S6K) 和 4EBP1被 mTOR磷酸化后启动核糖体蛋白的合成(Hay N, Sonenberg N (2004) . Genes Dev 18 (16): 1926 _ 45 ) 。
蛋白激酶基因共表达 518多个蛋白激酶, 其中受体激酶有 20亚科分 58类 型, 细胞桨激酶有 10亚科分 32类型; 在已知的 50多种致癌基因中, 大部分是 蛋白激酶基因 (Manning et al. Sicence, 2002, 298, 1912 ) 。 癌症基因通常处 于被抑制的状态, 但当它们发生突变或者被异常激活后, 例如化学毒物、 长期 紫外光照或放射性物质的作用下, 癌基因会过量地表达激酶, 或表达持续活化 的激酶,导致细胞生长、分化和凋亡等重要环节的失控而产生癌细胞(Croce CM. N Engl J Med. 2008 , 358 (5): 502 - 11 ) 。 针对细胞信号通路上失控的激酶或 生长因子等作为药物靶点, 研发小分子或大分子抑制剂, 对抗癌细胞生长与转 移已成为当今抗癌药物研发的重要方向 (Novel anticancer agents, Academic Press, 2006, editors: Alex A. Adijel ; John K. Buolamwini ) 。
Raf/MEK/ERK信号通路上的 Raf激酶在许多癌症细胞中被过量表达, 超过 60%的人类恶性黑色素瘤、 12%的大肠癌、 14%的卵巢癌中都发现 B-raf 的激活突 变, 这种突变还存在于甲状腺和肺部的肿瘤中; 另一方面, 在 50%的肾癌和几乎 100%的肝癌细胞中存在异常高的 C-raf 活性, 虽然其中 C-raf 并没有发生突变 (Brose, M. S. et al. Cancer Res. 2000, 63 : 6997-7000; Davies , H. Nature, 2002, 417 : 949-954 ; Yuen, S. T. et al. Cancer Res. 2002, 62 : 6451-6455)。 B-raf 突变后的激酶 B-raf-V600E, 其活性是野生型激酶的 500倍, 它能与 C_raf 形 成异源二聚体, 持续激活下游的 ERK及蛋白激酶 mTOR (Sridhar SS, Hedley D, Siu LL. Mol. Cancer Ther. 2005, 4 (4) : 677 - 85 ) 。 RAF激酶存在三种亚型, 包括 A-raf、 B-raf 和 C-raf (也称 Raf- 1 ) , 它们高度同源, 享有很高的序列 相似性, 所以小分子 RAF 激酶抑制剂通常会同时拮抗 B-ref、 C-raf 及 B-raf-V600E o 由拜耳研发的索拉非尼 (Sorafenib ) 是于 2005年批准的首个治 疗晚期肾癌的 RAF激酶抑制剂, 2007年又被批准为转移性肝癌的治疗药物。 进 一步的研究使拜耳的科学家们认为, 索拉非尼实际上能拮抗多重激酶, 它通过 同时拮抗 RAF激酶以及几种血管生成细胞生长因子受体(例如 VEGFR、 PDGFR等) 来抑制癌细胞的生长与转移 (Keating GM, Santoro A. Drugs 2009, 69 (2): 223 - 240 ) 。 肾癌及肝癌均是转移性极强的癌症, 其中肾癌的主要病理类型是 透明细胞癌(clear cel l carcinoma),其发病机制与 VHL抑癌基因的突变相关。 一旦 VHL 基因发生突变,即便在正常非缺氧生理状态下,其编码的氨基酸蛋白 (pVHL)也会导致缺氧诱导因子 -loc (hypoxia-inducible factor-la, HIF-la) 的 异常激活; HIF-loc再刺激 VEGF与 PDGF- β等细胞生长因子的表达合成,参与肿瘤 组织的新生血管生成。此外,大多数的肾癌具 EGFR表达,且表达程度与预后相关, HIF-lodl过激活 TGF-oc产生自分泌作用与 EGFR结合,从而促进细胞增殖与生存。 无论何种生长因子与其受体的结合,通常均需通过 Raf/MEK/ERK 通路发挥作用 (Gunaratnam L, et al. J Biol Chem. 2003, 278 : 44966-44974)。
早在 1971年, Judah Folkman即提出血管生成理论 (Angiogenesis )来解 释肿瘤的增长与转移 (metastasis ) 。 他们的实验显示血管内皮细胞生长因子 抑制齐 ij, 内皮抑素 (endostatin) 与人血管抑素 (angiostatin) 可以抑制小鼠 上肿瘤的增长。他认为肿瘤细胞在自身分裂繁殖到一定程度后(1-2立方毫米), 必须借助新生的肿瘤血管提供养分与氧气来帮助其进一步增长与扩散(Folkman J, Klagsbrun M. Science 1987, 235 (4787): 442 - 7 ) 。 在血管形成过程中, 癌细胞分泌出血管内皮细胞生长因子(VEGF)与细胞摸上的受体(VEGFR)结合, 刺激血管内皮细胞的生长、 分裂、 繁殖; 同时细胞释放出蛋白水解酶 (proteolytic enzymes ) 和基质金属蛋白酶 (MMP ) 降解周围基质, 细胞得以 前移、 增长、 形成管道化血管环和新基底膜, 最终形成新生血管。 文献进一步 显示, 多种不同的生长因子同时参与新生肿瘤血管的形成; 除 VEGF外, 还有血 小板衍生生长因子(Platelet-derived growth factor, PDGF) 和成纤维细胞生 长因子 (Fibroblast Growth Factor, FGF) 等。 癌细胞分泌的 PDGF和 FGF等 生长因子通过招募和刺激纤维细胞、 平滑肌细胞、 外膜细胞的生长形成新的癌 细胞血管基质, 帮助新生血管的形成。 与正常血管不同, 这种新生肿瘤血管往 往结构不规整, 血管基质不完善, 渗透性高, 所以肿瘤细胞得以渗透到血液中, 扩散并附着在身体的其它部位上生长繁殖形成癌症转移 (metastasis ) 。 临床 研究证明, 抑制这一过程能有效地阻止肿瘤的增长和转移, 延长患者寿命 (Folkman J. Scientific American 1996, 275 (3): 150 - 4 ) 。 已知参与癌 细胞新生血管形成的生长因子受体有 VEGFR-l(Flt-l ), VEGFR-2(KDR,或 Flk_l ), 血小板衍生生长因子(PDGF) 受体 PDGFR-oc及 PDGFR-β , 和成纤维细胞生长因子
(FGF) 受体 FGFR1-4等; 另一方面, 结构相似的 VEGFR-3 (Flt-4 )受体主要参 与新生淋巴管的形成 (Lymphangiogenesis ) , 在癌症通过淋巴系统转移的过程 中扮演重要的角色, 这些受体都可以作为对抗实质肿瘤增长与转移的靶点
(Steven A. Stacker. Lymphangiogenesis in Cancer Metastasis, Springer. 2009 pp. 27 _ ) 。
综上所述, 最近二十年来人们对癌症的发生、 生长、 存活及转移在分子层 面上的认识取得了非凡的进步, 更重要的是, 多个临床研究实例都证明了用小 分子或大分子药物抑制激酶或生长因子来治疗癌症的疗效; 研究血管生成 (Angiogenesi s ) 的不懈努力也终于证明了抑制血管内皮细胞生长因子或者受 体的临床疗效, 从阻抗癌细胞周围血管形成的角度来延缓癌症的增长与扩散, 延长患者寿命。
2001年 5月, 美国 FDA批准了第一个酪氨酸激酶小分子抑制剂伊马替尼 ( Imatinib ) 为慢性粒细胞白血病 (慢性骨髓血癌, CML ) 的治疗用药 ( Gambacorti-Passerini C. Lancet Oncology 2008 , 9 (600) : 600 ) , 伊马替 尼首次证明了激酶抑制剂治疗癌症的可行性, 同时伊马替尼显示了相对于化学 疗法较低的药物毒性, 提高了患者的生活品质。 在分子层面, 伊马替尼通过拮 抗白血病癌细胞中过度表达的 bcr-abl 激酶, 抑制细胞的生长和分裂; 它同时 也拮抗 c-kit激酶的活性, 并在临床上显示其对胃肠道间质细胞瘤的疗效, 被 批准为胃肠道间质细胞瘤的治疗用药。 几年后, 百时美-施贵宝和诺华又分别于 2006年和 2007年开发了另外两个慢性骨髓血癌治疗用药-达沙替尼(Dasatinib ) 和尼罗替尼 (Ni lotinib ) , 而且两者对抗伊马替尼的病患也有良好的疗效。
由英国阿斯利康公司研发的吉非替尼 (Gefitinib )于 2002年 8月批准在 曰本上市, 作为非小细胞肺癌一线治疗药物吉非替尼尤其对亚洲吸烟男性非小 细胞肺癌患者非常有效。 2004 年, 基因泰克和 0SI 制药联合开发了厄洛替尼 (erlotinib ) , 它的适用范围更加广泛, 在全球七、 八十个国家中被用作非小 细胞肺癌一线治疗、 维持治疗和序贯治疗的药物。 吉非替尼和厄洛替尼均为表 皮生长因子受体 (EGFR) 抑制剂, 拮抗细胞表面跨膜上 EGFR受体与表皮生长因 子(EGF)的结合而抑制 EGFR转导的 Raf/MEK/ERK等信号通道(Raymond E, Faivre S, Armand J. Drugs 2000, 60 Suppl 1 : 15 - 23 ; discussion 41 - 2 ) 。 同样 的原理, 厄洛替尼也能有效地抑制胰腺肿瘤细胞的生长, 成为 FDA批准的第一 个治疗胰腺癌的靶向药物。
2006年, 辉瑞与 Sugene共同开发了舒尼替尼 (Sunitinib ) 。 作为治疗肾 癌 (RCC) 的酪氨酸激酶小分子抑制剂, 舒尼替尼通过拮抗血管内皮细胞生长因 子受体 (VGEFR) , 抑制癌细胞新生血管的形成来延缓癌细胞的生长与转移。 舒 尼替尼同时也能抑制 c-kit激酶的活性, 被批准为抗伊马替尼胃肠道间质细胞 瘤患者的治疗用药。
由拜耳研发的索拉非尼 (Sorafenib ) 通过同时拮抗 RAF激酶、 血管内皮 细胞生长因子受体 (VGEFR) 及血小板衍生生长因子受体 (PDGFR-β ) 等多重途 径对抗癌症的生长与转移, 被 FDA批准为晚期肾癌及转移性肝癌的治疗药物。 索拉非尼对转移性肝癌的疗效是肝癌治疗领域的重大突破, 肝癌是公认最难治 疗, 转移性最强的癌症之一 (Escudier B, et al. (January 2007) . N. Engl. J. Med. 356 (2): 125 - 34; Keating GM, Santoro A. Drugs 2009, 69 (2) : 223 - 240 ) 。
哺乳动物雷帕霉素靶蛋白 (mTOR)是 PI3K/AKT信号通道中一个多功能丝 / 苏氨酸激酶, 与癌细胞的生长、 分裂、 存活及转移都有直接的关联 ( Rubio-Viqueira, B, Hidalgo M. Curr. Op in. Invest ig. Drugs 2006, 7 : 501 - 512 ) 。 mTOR激酶在肾癌转移中的作用尤其明显, 失控的 mTOR引 起细胞内 HIF-loc浓度的提高, HIF-loc再诱发 VGEF的合成, 促进癌细胞血管形 成; 另一方面, 许多肾癌细胞由于 VHL肿瘤抑制基因的突变或失去引起 HIF-loc 分解的减少也提高 HIF-loc的浓度 (Thomas GV. et al. Nature Medicine 2006, 12 : 122 - 127 ) 。 第一个临床用 mTOR抑制剂为美国惠氏制药研发的雷帕霉素衍 生物西罗莫司 (Temsirol imus ) , 2007年被 FDA批准为肾癌的治疗用药。
拉帕替尼 (Lapatinib ) 是小分子酪氨酸激酶双重抑制剂, 它能同时抑制 表皮生长因子受体 (EGFR) 和 ErbB2 (HER-2/neu) 受体, 大约有 30%左右的乳 腺癌病患由于 HER-2/neu原癌基因而产生过量的 HER-2/neu受体。 2007年, 拉 帕替尼被 FDA批准为乳腺癌组合疗法用药, 2010年又被批准为 ER+/EGFR+/HER2+ 三重阳性乳腺癌患者的一线用药(Wood ER et al. Cancer research 64 (18):
6652 - 9 ) 。
由于激酶的高度同源性及多样性, 目前研发的小分子抗癌药物大多为多重 激酶抑制剂, 幸运的是与以前担心的不同, 较低的激酶选择性并没有影响药物 的疗效; 相反, 优选的多重激酶抑制剂可能更利于对抗成因复杂, 形式多样的 癌症, 并可一药多用。
与小分子激酶抑制剂多重性不同的是, 大分子单克隆抗体通常是只针对细 胞信号通道中单一靶点的特异抑制剂, 多个临床实例也已证明了特异性单克隆 抗体治疗癌症的疗效。 1998年, 曲妥珠单抗 (Trastuzumab ) 获得美国 FDA批 准为乳腺癌治疗用药, 适用于过量表达 HER-2/neu的乳腺癌病患 (Hudi s, CA. N Engl J Med. 2007 , 357 (1) : 39 - 51 ) 。 2004 年, FDA 批准了贝伐单抗 (Bevacizumab )与标准化疗药物结合治疗转移性大肠癌 (mCRC)和非小细胞肺癌 (NSCLC ) 。 贝伐单抗特异性地结合血管内皮细胞生长因子 (VGEF) , 抑制癌 新生血管的形成, 并能促进化疗药物对癌症组织的渗透 (Los M et al. The Oncologist 2007, 12 (4) : 443 - 50 ) 。 继后, 帕尼单抗 (Panitumumab ) 和西 妥昔单抗 (cetuximab ) 也被批准为转移性大肠癌用药。 与贝伐单抗不同, 帕尼 单抗和西妥昔单抗通过与表皮生长因子受体 (EGFR) 的结合来抑制癌细胞生长 分裂的信号通道。 西妥昔单抗 (IgGl ) 与帕尼单抗 (IgG2)分属于免疫球蛋白的 不同亚型, 它们在抗癌机理上存在细微的差别, 除对大肠癌的疗效外, 施贵宝 公司生产的西妥昔单抗也可治疗头颈癌。
上述临床实例全面证明了用激酶抑制剂拮抗癌细胞失控的信号通道, 治疗 不同癌症的可行性。 然而, 癌症是最复杂的疾病之一, 身体各个器官都可能通 过各种机理形成结构组织各异的癌症, 很多癌症是由多个基因的同时突变造成, 而且同类癌症也可由不同的癌变原因形成等。 癌症成因机理的多元性, 形式结 构的多样性使它的治疗一直以来充满着各种挑战, 癌症专家不得不普遍利用多 种药物的不同组合来对抗肿瘤顽疾; 另一方面, 癌症也会对现有药物产生抗药 性, 所以有必要不断地开发结构新颖、 功能各异、 药效更好的新型靶向药物, 根据信号通路和肿瘤遗传学信息进行药物的优选组合, 提高癌症治疗的疗效。
血管内皮生长因子(VEGF)是肿瘤血管生成过程中最重要的细胞生长因子, 肿瘤血管对 VEGF高度敏感, 在很多肿瘤细胞中 VEGF mRNA浓度显著地高于正常 细胞, 这些肿瘤包括肺癌(Mattern et al. Br. J Cancer 1996, 73, 93, 1) , 甲 状腺癌(Vigl ietto et al. Oncogene 1995, 11, 1569) , 乳腺癌(Brown et al. Human Pathol. 1995, 26, 86), 胃腸癌(Brown et al. Cancer Res. 1993, 53, 4727 ; Suzuki et al. Cancer Res. 1996, 56, 3004) , 肾癌和膀胱癌 (Brown et al. Am. JPalhol. 1993, 143L 1255) , 卵巢癌(Olson et al. Cancer Res. 1994, 54, 1255) , 宫颈癌(Guidi et al. J Nat 'l Cancer 30 Inst. 1995, 87, 12137)、 以及血管 肉瘤 (Hashimoto et al. Lab. Invest. 1995, 73, 859)和多种颅内肿瘤 (Plate et al. Nature 1992, 359, 845 ; Phi l l ips et al. Int. J Oncol. 1993, 2, 913 ; Berkman et al. J Clin. Invest. , 1993, 91 ; 153)。 所以继贝伐单抗、 舒尼替 尼、 索拉非尼作为血管形成抑制剂成功地应用于临床以来, 研发新型血管形成 抑制剂(如 VEGFR-2及 PDGFR-β抑制剂等)作为广谱抗癌药物已成为十分热门的 领域, 并已取得了新的、 前景看好的临床试验结果。
W0-2004007458公开了一组 2_垸胺烟酰胺衍生物作为 VEGFR、 PDGFR and Kit抑制剂, 在临床三期试验中用于非鳞状非小细胞肺癌的治疗, 其它适应症包 括实质癌, 胃腸癌, 结肠直肠癌,内分泌癌, 乳腺癌及肺癌。
W0-2004113304公开了一组吲唑、苯并异唑及苯并噻唑衍生物作为 CSFR- PDGFR、 Flt3、 Kit, VEGFR- 1、 VEGFR-2, VEGFR-3抑制剂, 在临床三期试验中用 于治疗肝癌, 二期临床用于非小细胞肺癌, 乳腺癌,结肠直肠癌, 其它适应症还 包括肾癌, 急性髓样白血病 (AML)和骨髓增生异常综合征 (MDS)。
W0-2000043366 公开了一组喹啉-脲衍生物作为 Kit、 PDGFR 科受体、 VEGFR-K VEGFR-2, VEGFR-3抑制剂, 在临床三期试验中用于治疗肾癌, 其它临 床实验包括乳腺癌, 结肠直肠癌, 胃腸癌, 非小细胞肺癌和肝癌等。
W0-200102369公开了一组吲唑衍生物作为 CSFR_1、 PDGF、 VEGF_1、 VEGF-2 及 VEGF-3抑制剂,在临床三期试验中用于治疗肾癌,二期临床实验包括間皮癌, 血管肉瘤, 肾上腺皮质腺癌和肝癌等。
W0-200232872公开了一组新型喹啉-脲衍生物作为 VEGFR_2、 VEGFR-3, FGFR 1-4及 RET抑制剂, 在临床三期试验中用于甲状腺癌的治疗, 二期临床实 验包括肝癌, 子宫内膜癌, 黑素瘤, 肾癌及神经胶质瘤。
W0-2003082272 公开了一组芳胺苯并咪唑类化合物作为 Raf, VEGFR-2, PDGFR-β及 Kit抑制剂用于治疗黑素瘤。
中国专利 CN101475513A, CN101260106A和 CN101735215A也公开了三种 raf激 酶抑制剂双芳基脲类化合物在治疗癌症上的用途。 发明简述
本发明涉及 N- ( (4-氯 -3-三氟甲基)苯基) -N,- (2-氟 -4- ( (2-羟甲基氨基甲 酰基: )-4-吡啶基氧) 苯基) 脲或其药学上可接受的盐, 以及治疗患者的方法, 该 方法包括向患者施用抑制 VEGFR-2激酶有效量的 N- ( (4-氯 -3-三氟甲基)苯基) -Ν'- (2-氟 -4- ( (2-羟甲基氨基甲酰基) -4-吡啶基氧) 苯基)脲或其药学上可接受 的盐。
本发明涉及 Ν- ( (4-氯 -3-三氟甲基)苯基) -Ν,- (2-氟 -4- ( (2-羟甲基氨基甲 酰基: )-4-吡啶基氧) 苯基) 脲或其药学上可接受的盐, 这些盐选自:
a)有机酸和无机酸的碱式盐,所述有机酸和无机酸选自盐酸、氢溴酸、硫酸, 磷酸、 甲磺酸、 三氟磺酸、 苯磺酸、 对甲苯磺酸、 1-萘磺酸、 2-萘磺酸、 乙酸, 三氟乙酸、 苹果酸、 酒石酸、 柠檬酸、 乳酸、 草酸、 琥珀酸、 富马酸、 马来酸, 苯甲酸、 水杨酸、 苯基乙酸或杏仁酸; 和
b)含阳离子的有机和无机碱的酸式盐, 所述阳离子选自碱金属阳离子、碱土 金属阳离子、 铵离子、 脂族取代的铵离子或芳族取代的铵离子。
本发明用于治疗癌症的药物组合物,包含 N- ( (4-氯 -3-三氟甲基)苯基) -N,- (2-氟 -4- ( (2-羟甲基氨基甲酰基: )-4-吡啶基氧)苯基)脲或其药学上可接受的盐, 和生理上合适的载体。
本发明所述的化合物或其药学上可接受的盐可用于制造抑制 VEGFR-2激酶 介导的癌性细胞生长的药物的用途。
本发明所述的化合物或其药学上可接受的盐还可以用在制备治疗癌症疾病 药物上的用途。
本发明所述的癌症包括实质癌、 肾癌、 肺癌、 乳腺癌、 肝癌、 卵巢癌、 胰 腺癌、 甲状腺癌、 膀胱癌、 白血病、 黑素瘤、 胃癌, 结肠直肠癌、 内分泌癌、 急 性髓样白血病 (AML:)、 骨髓增生异常综合征 (MDS:)、 间皮癌、 血管肉瘤、 肾上腺 皮质腺癌、 子宫内膜癌及神经胶质瘤等。 尤其对肾癌、 肝癌、 肺癌、 结肠直肠 癌、 胃癌、 乳腺癌及血管肉瘤等的治疗有效。
药学上可接受的盐优选 N- ( (4-氯 -3-三氟甲基) 苯基) -N,- (2-氟 -4- ( (2- 羟甲基氨基甲酰基: )-4-吡啶基氧)苯基)脲的可药用的酸加成盐。例如酸加成盐, 优选与有机或无机酸形成的酸加成盐, 优选可药用盐。 合适的无机酸有例如氢 卤酸如盐酸、 硫酸或磷酸。 合适的有机酸有例如羧酸、 膦酸、 磺酸或氨基磺酸, 例如乙酸、 丙酸、 辛酸、 癸酸、 十二垸酸、 羟基乙酸、 乳酸、 2-羟基丁酸、 葡糖 酸、 富马酸、 丁二酸、 己二酸、 庚二酸、 辛二酸、 壬二酸、 苹果酸、 酒石酸、 柠檬酸、 葡糖二酸、 半乳糖二酸、 氨基酸, 例如谷氨酸、 天冬氨酸、 N-甲基甘 氨酸、 乙酰氨基乙酸、 N-乙酰基天冬酰胺、 N-乙酰基半胱氨酸、 丙酮酸、 乙酰 乙酸、 磷酸丝氨酸、 2-或 3-甘油磷酸、 马来酸、 羟基马来酸、 甲基马来酸、 环己 垸甲酸、 苯甲酸、 水杨酸、 1-或 3-羟基萘 -2-甲酸、 3, 4, 5-三甲氧基苯甲酸、 2- 苯氧基苯甲酸、 2-乙酰氧基苯甲酸、 4-氨基水杨酸、 邻苯二甲酸、 苯基乙酸、 葡 糖醛酸、 半乳糖醛酸、 甲垸-或乙垸-磺酸、 2-羟基乙垸磺酸、 乙垸 -1, 2-二磺酸、 苯磺酸、 2-萘磺酸、 1, 5-萘二磺酸、 N-环己基氨基磺酸、 N-甲基-、 N-乙基-或 N-丙基 -氨基磺酸, 或其它有机质子酸, 例如抗坏血酸。
本发明涉及用作 VEGFR-2激酶抑制剂的 N- ( (4-氯 -3-三氟甲基) 苯基) -N,- (2-氟 -4- ( (2-羟甲基氨基甲酰基: )-4-吡啶基氧) 苯基) 脲, 及其用于制备治疗 VEGFR-2激酶所介导疾病的药物中的用途。
所述患者为哺乳动物, 一般为人。
本发明的化合物可用于治疗哺乳动物癌症, 优选人类癌症, 包括但不限于 实质癌、 肾癌、 肺癌、 乳腺癌、 肝癌、 卵巢癌、 胰腺癌、 甲状腺癌、 膀胱癌、 白血病、 黑素瘤、 胃癌, 结肠直肠癌、 内分泌癌、 急性髓样白血病 (AML:)、 骨髓 增生异常综合征 (MDS:)、 间皮癌、 血管肉瘤、 肾上腺皮质腺癌、 子宫内膜癌及神 经胶质瘤。 本发明的化合物也可用于治疗炎性疾病, 包括类风湿性关节炎、 视 网膜病(包括糖尿病性视网膜神经病和黄斑变性)、 心血管疾病和代谢疾病。
本发明的 N- ( (4-氯 -3-三氟甲基)苯基) -N,- (2-氟 -4- ( (2-羟甲基氨基甲酰 基 )-4-吡啶基氧) 苯基)脲或其药学上可接受的盐具有如上所述的有价值的药理 学性质。
有机酸和无机酸的碱式盐, 所述有机酸和无机酸选自盐酸, 氢溴酸, 硫酸, 磷酸, 甲磺酸, 三氟磺酸, 苯磺酸, 对甲苯磺酸, 1-萘磺酸, 2-萘磺酸, 乙酸, 三氟乙酸, 苹果酸, 酒石酸, 柠檬酸, 乳酸, 草酸, 琥珀酸, 富马酸, 马来酸, 苯甲酸, 水杨酸, 苯基乙酸和杏仁酸; 和
b)含阳离子的有机和无机碱的酸式盐, 所述阳离子选自碱金属阳离子, 碱土 金属阳离子, 铵离子, 脂族取代的铵离子和芳族取代的铵离子。
本发明所述的 N- ( (4-氯 -3-三氟甲基)苯基) -N,- (2-氟 -4- ( (2-羟甲基氨基 甲酰基 )-4-吡啶基氧) 苯基) 脲药学上可接受的盐为可药用的酸加成盐。 例如酸 加成盐, 优选与有机或无机酸形成的酸加成盐, 尤其是可药用盐。 合适的无机 酸选自盐酸, 氢溴酸, 硫酸, 磷酸等。 合适的有机酸选自羧酸、 膦酸、 磺酸或 氨基磺酸, 例如甲磺酸、 三氟磺酸、 苯磺酸、 对甲苯磺酸、 1-萘磺酸、 2-萘磺酸、 乙酸、 三氟乙酸、 苹果酸、 酒石酸、 柠檬酸、 乳酸、 草酸、 琥珀酸、 富马酸、 马来酸, 苯甲酸、 水杨酸、 苯基乙酸、 杏仁酸、 丙酸、 辛酸、 癸酸、 十二垸酸、 羟基乙酸、 2-羟基丁酸、 葡糖酸、 丁二酸、 己二酸、 庚二酸、 辛二酸、 壬二酸、 葡糖二酸、 半乳糖二酸, 或者氨基酸, 例如谷氨酸、 天冬氨酸、 N-甲基甘氨酸、 乙酰氨基乙酸、 N-乙酰 基天冬酰胺、 N-乙酰基半胱氨酸、 丙酮酸、 乙酰乙酸、 磷酸丝氨酸、 2-或 3-甘油磷酸、 羟基马来酸、 甲基马来酸、 环己垸甲酸、 1-或 3-羟基萘 -2-甲酸、 3, 4, 5-三甲氧基苯甲酸、 2-苯氧基苯甲酸、 2-乙酰氧基苯甲 酸、 4-氨基水杨酸、 邻苯二甲酸、 苯基乙酸、 葡糖醛酸、 半乳糖醛酸、 甲垸-或 乙垸-磺酸、 2-羟基乙垸磺酸、 乙垸 -1, 2-二磺酸、 1, 5-萘二磺酸、 N-环己基氨 基磺酸、 N-甲基-、 N-乙基-或 N-丙基 -氨基磺酸, 或其它有机质子酸, 例如抗坏 血酸。
本发明的化合物可以单独施用或与其它抗癌药组合施用, 例如抑制肿瘤血 管生成的化合物, 例如蛋白酶抑制剂、 表皮生长因子受体激酶抑制剂、 血管内 皮生长因子受体激酶抑制剂等; 细胞毒性药物, 例如抗代谢物, 如嘌呤和嘧啶 类似物抗代谢物; 抗有丝分裂药如微管稳定药和抗有丝分裂生物碱; 铂配位复 合物; 抗肿瘤抗生素; 垸化剂, 例如氮芥和亚硝基脲; 内分泌物, 例如肾上腺 类皮质激素类、 雄激素类、 抗雄激素类、 雌激素类、 抗雌激素类、 芳香酶抑制 剂、 促性腺激素释放激素激动剂和促生长素抑制素类似物, 以及靶向于被过量 表达和 /或在其它方面与肿瘤细胞中被上调的特定代谢通路有关的酶或受体的化 合物, 例如 ATP和 GTP磷酸二酯酶抑制剂、 蛋白激酶抑制剂, 例如丝氨酸、 苏 氨酸和酪氨酸激酶抑制剂,例如 Abelson蛋白酪氨酸激酶和各种生长因子、它们 的受体和其激酶抑制剂, 例如表皮生长因子受体激酶抑制剂、 血管内皮生长因 子受体激酶抑制剂、 成纤维细胞生长因子抑制剂、 胰岛素样生长因子受体抑制 剂和血小板源生长因子受体激酶抑制剂等; 甲硫氨酸氨基肽酶抑制剂、 蛋白酶 体抑制剂、 环加氧酶抑制剂, 例如环加氧酶 -1或 -2抑制剂, 和组蛋白脱乙酰基 酶抑制剂。
本发明的化合物还可以与放射疗法、 免疫疗法、 手术治疗或其组合一起施 用。 用于在肿瘤减轻或甚至化学预防治疗后维持患者状态的治疗 (例如在危险中 的患者的情况:)也是可能的。
本发明的化合物不仅用于 (预防性地和优选治疗性地:)治疗人,而且用于治疗 其它温血动物, 例如商业上有用的温血动物, 例如啮齿类动物, 如 小鼠、 兔或 大鼠, 或豚鼠。
本发明还包括包含 N- ( (4-氯 -3-三氟甲基)苯基) -N,- (2-氟 -4- ( (2-羟甲基 氨基甲酰基: )-4-吡啶基氧) 苯基) 脲和生理学上认可的载体的药物组合物。
本发明化合物可通过注射、 吸入或喷洒或直肠, 经口、 皮肤、 胃肠外给予, 或以单位制剂剂型给予。 "注射给予"包括静脉、 肌内、 皮下和胃肠外注射, 以 及应用输液技术。 皮肤给药包括外用或透皮给予。 一种或多种化合物可与一种 或多种药学上认可的无毒载体, 以及视需要而定的其他活性成分共存。
口用组合物可按照任何药物组合物制造领域已知的合适方法制备。 为了改 善制剂口感, 所述组合物可含一种或多种以下试剂: 稀释剂, 甜味剂, 香料, 着色剂和防腐剂。 片剂含有活性成分, 它们与药学上认可的、 适合片剂生产的 无毒赋形剂混合。 所述赋形剂例如惰性稀释剂, 例如碳酸钙, 碳酸钠, 乳糖, 磷酸钙或磷酸钠; 粒化剂和崩解剂, 例如玉米淀粉或藻酸; 黏合剂, 例如硬脂 酸镁, 硬脂酸或滑石粉。 片剂可以没有包衣, 也可以用已知技术进行包裹, 以 延迟其在胃肠道内的崩解与吸收, 提供长期的持续作用。 例如, 可采用诸如一 硬脂酸甘油酯或二硬脂酸甘油酯之类延时物质。 所述化合物也可以制成固体, 快释形式。
口服制剂还可以是硬明胶胶囊, 其中的活性成分与例如碳酸钙、 磷酸钙或 高岭土等惰性固体稀释剂相混合, 或者是软明胶胶囊, 其中的活性成分与水或 例如花生油、 液体石蜡或橄榄油等油混合。
也可使用含有活性物质与适合制造水性悬浮液的赋形剂混合的水性悬浮 液。 所述赋形剂是悬浮剂, 例如羧甲基纤维素钠, 甲基纤维素, 羟基丙基 -甲基 纤维素, 藻酸钠, 聚乙烯基吡咯垸酮, 西黄蓍胶和阿拉伯树胶; 分散剂或润湿 剂可以是天然磷脂, 例如卵磷脂, 或环氧乙垸与脂肪酸的缩合产物, 例如硬脂 酸聚氧乙烯酯, 或环氧乙垸与长链脂肪醇的缩合产物, 例如十七氧乙烯十六垸 醇, 或环氧乙垸与脂肪酸与己糖醇所成偏酯的缩合产物, 例如单油酸聚氧乙烯 山梨醇酯。 水性悬浮液还可含有一种或多种防腐剂, 例如对羟基苯甲酸乙酯或 正丙酯, 一种或多种着色剂, 一种或多种香料, 和一种或多种甜味剂, 例如蔗 糖或糖精。
适合加水成为水性悬浮液的可分散粉末或颗粒中, 活性成分与分散剂或润 湿剂, 悬浮剂和一种或多种防腐剂混合。 合适的分散剂或润湿剂和悬浮剂可以 上文所述为例。 还可以含其他赋形剂, 例如甜味剂, 香料和着色剂。
化合物的形式还可以是非水性液体制剂, 例如油性悬浮液, 这可以通过将 活性成分悬浮在花生油、 橄榄油、 芝麻油或花生油等植物油或诸如液体石蜡等 矿物油中来配制。 该油性悬浮液可含增稠剂, 例如蜂蜡、 硬石蜡或鲸蜡醇。 为 了改善口感, 可加入上述甜味剂和香料。 所述组合物可通过添加诸如抗坏血酸 等抗氧化剂来保质。
本发明药物组合物的形式还可以是水包油乳液。 油相可以是诸如橄榄油或 花生油等植物油或例如液体蜂蜡等矿物油, 或它们的混合物。 合适的乳化剂可 以是西黄蓍胶和阿拉伯树胶等天然树胶, 或天然磷脂, 例如大豆卵磷脂或卵磷 脂; 脂肪酸与脱水己糖醇形成的偏酯, 例如但油酸脱水山梨醇酯; 所述偏酯与 环氧乙垸的缩合产物, 例如单油酸聚氧乙烯脱水山梨醇酯。 所述乳液还可含有 甜味剂和香料。
也可用例如甘油、 聚丙二醇、 山梨醇或蔗糖等甜味剂配制糖桨和酏剂。 这 类制剂还可含有润药, 防腐剂和香料及着色剂。
本发明的药物组合物以本身已知的方法制备,例如通过常规的混合、制粒、 成型、 溶解或冻干方法制备。
优选使用活性成分的溶液, 另外还可使用混悬液或分散体, 尤其是等张的 水溶液、 分散体或混悬液, 例如在仅含有活性物质或含有活性物质与载体例如 甘露醇的冻干组合物的情况下, 这些剂型可以在使用前制备。 药物组合物可以 被灭菌和 /或包含赋形剂, 例如防腐剂、 稳定剂、 湿润剂和 /或乳化剂、 增溶剂、 调节渗透压的盐和 /或缓冲剂, 并以本身已知的方法制备, 例如通过常规的溶解 或冻干方法制备。 所述的溶液或混悬液可以包含 增粘物质例如羧甲基纤维素 钠、 羧甲基纤维素、 葡聚糖、 聚乙烯吡咯垸酮或明胶或增溶剂例如 Tween 80。
在油中的混悬液包含常规用于注射目的的植物油、 合成油或半合成油作为 油性组分。可提及的如尤其是液体脂肪酸酯, 其包含具有 8至 22个、 尤其是 12 至 22个碳原子的长链脂肪酸作为酸组分, 例如月桂酸、 十三垸酸、 肉豆蔻酸、 十五垸酸、 棕榈酸、 十七垸酸、 硬脂酸、 花生酸、 二十二垸酸或相应的不饱和 酸, 例如油酸、 反油酸、 芥酸、 巴西烯酸或亚油酸, 任选地加入抗氧化剂, 例 如维生素E、 β -胡萝卜素或 3, 5-二-叔丁基 -4-羟基甲苯。 这些脂肪酸酯的醇组 分具有最多 6个碳原子, 为单-或多-元醇, 例如单-、 二-或三-元醇, 例如甲醇、 乙醇、 丙醇、 丁醇或戊醇或它们的异构体, 但是尤其是乙二醇和甘油。 因此, 可以提及的脂肪酸酯的例子有: 油酸乙酯、 肉豆蔻酸异丙酯、 棕榈酸异丙酯等, 但是尤其是植物油, 例如棉子油、 杏仁油、 橄榄油、 芝麻油、 豆油和更尤其是 花生油。
注射组合物的制备在无菌条件下以常规方法进行, 将其引入到例如安瓿或 小瓶中和容器的密封也是在无菌条件下以常规方法进行的。
用于口服施用的药物组合物可以例如通过以下方法获得: 将活性成分与一 种或多种固体载体混合, 将得到的混合物制粒, 如果合适, 将混合物或颗粒加 工 (如果需要, 酌情加入另外的赋形剂:)成片剂或糖衣丸的芯。
合适的载体尤其是填充剂, 例如糖如乳糖、 蔗糖、 甘露醇或山梨糖醇、 纤 维素制品和 /或磷酸钙例如磷酸三钙或磷酸氢钙, 还有粘合剂,例如淀粉如玉米、 小麦、 米或马铃薯淀粉、 甲基纤维素、 羟丙基甲基纤维素、 羧甲 基纤维素钠和 /或聚乙烯吡咯垸酮, 和 /或如果需要, 崩解剂, 例如以上提到的淀粉, 还有羧甲 基淀粉、 交联聚乙烯吡咯垸酮、 海藻酸或其盐, 例如藻酸钠。 另外的赋形剂尤 其是流动性调节剂和润滑剂, 例如硅酸、 滑石粉、 硬脂酸或其盐, 例如硬脂酸 镁或硬脂酸钙, 和 /或聚乙二醇或其衍生物。
糖衣丸芯可以具有合适的包衣, 任选肠溶衣, 尤其是使用浓糖溶液, 所述 浓糖溶液可以含有阿拉伯胶、 滑石粉、 聚乙烯吡咯垸酮、 聚乙二醇和 /或二氧化 钛, 或使用在合适有机溶剂或溶剂混合物中的包衣溶液, 或者对于肠溶衣的制 备, 使用合适的纤维素制品的溶液, 例如醋酸纤维素邻苯二甲酸酯或羟丙基甲 基纤维素邻苯二甲酸酯。 可以向片剂或糖衣丸包衣中加入着色剂或色素, 例如 为了鉴别目的或为了显示活性成分的不同剂量。
用于口服施用的药物组合物还有硬明胶胶囊和由明胶和增塑剂例如甘油或 山梨醇组成的软密封胶囊。 硬明胶胶囊可以含有颗粒形式的活性成分, 例如混 合有填充剂如玉米淀粉、 粘合剂和 /或助流剂如滑石粉或硬脂酸镁以及任选地稳 定剂的颗粒形式的活性成分。 在软胶囊中, 活性成分优选溶解或混悬在合适的 液体赋形剂中, 例如脂肪油、 石蜡油或液体聚乙二醇或者乙二醇或丙二醇的脂 肪酸酯, 同样可以加入稳定剂和清洁剂, 例如聚氧乙烯脱水山梨醇脂肪酸酯类 的清洁剂。
合适的可直肠施用的药物组合物有例如栓剂, 其由活性成分与栓剂基质的 混合物组成。 合适的栓剂基质有例如天然或合成的甘油三酯、 链垸烃、 聚乙二 醇或高级链垸醇。
对于胃肠外施用, 尤其适合的是水溶性形式、 例如水溶性盐形式的活性成 分的水性溶液, 或水性注射混悬液, 其包含增粘物质, 例如羧甲基纤维素钠、 山梨醇和 /或葡聚糖, 以及如果需要, 还含有稳定剂。 任选地与赋形剂一起的活 性成分也可以是冻干物的形式, 其可以在胃肠外施用前通过加入合适的溶剂被 制成溶液。
用于例如胃肠外施用的溶液也可以以输液形式被使用。 优选的防腐剂有例 如抗氧化剂如抗坏血酸或杀微生物剂如山梨酸或苯甲酸。
在本文中所有使用 N- ( (4-氯 -3-三氟甲基)苯基) -N,- (2-氟 -4- ( (2-羟甲基 氨基甲酰基: )-4-吡啶基氧)苯基)脲的治疗方案中,每日口服剂量以 0.01-200mg/kg 体重为佳。 注射, 包括静脉、 肌内、 皮下和胃肠外注射和应用输入技术, 的每 日剂量以 0.01-200mg/kg体重为佳。 直肠给药的日剂量以 0.01-200mg/kg体重为 佳。外用的每日剂量以每日 1至 4次, 没每次 0.1-200mg为佳。吸入的每日剂量 以 0.01-10mg/kg体重为佳。
本领域技术人员将看出, 具体的给药方式取决于多种因素, 这些都是在常 规给药时常考虑的。 然而, 还可以看出, 特定患者的具体剂量取决于包括所用 具体化合物的活性、 患者年龄、 患者体重、 患者总体健康状况、 患者性别、 患 者饮食、 给药时间、 给药途径、 排出速度、 药物组合和当前所治病情的严重程 度等多种因素。 本领域技术人员还将发现, 最佳治疗程式, 即一定天数内的治 疗方式和 N- ( (4-氯 -3-三氟甲基)苯基) -N,- (2-氟 -4- ( (2-羟甲基氨基甲酰基) -4- 吡啶基氧) 苯基) 脲或其药学上认可的盐的日给药次数, 本领域技术人员可用 常规治疗试验来确定。
显然, 具体患者的具体记录水平取决于多种因素, 包括所用具体化合物的 活性、 患者年龄、 患者体重、 患者总体健康状况、 患者性别、 患者饮食、 给药 时间、 给药途径、 排出速度、 药物组合和当前所治病情的严重程度。
本发明化合物可由已知化合物 (或以可由已知化合物制得的原料作为起始 物:), 通过例如后文所述的一般方法来制备。 可以后文所述的常用方法测定各化 合物已知 VEGFR-2激酶的活性。 以下实施例仅作说明, 而非限定。
一般方法
本发明的化合
用反应式制备的。
Figure imgf000015_0001
本发明所述化合物抑制肿瘤生长的生物活性按以下方法测定。
取生长旺盛期的人肝癌 HepG2瘤组织剪切成 1.5 mm3左右,在无菌条件下, 接种于裸小鼠右侧腋窝皮下。 裸小鼠移植瘤用游标卡尺测量移植瘤直径, 待肿 瘤生长至 60〜200mm3后将动物随机分组。使用测量瘤径的方法, 动态观察受试 物抗肿瘤的效应。 肿瘤直径的测量次数为每周 3次, 每次测量同时还需称鼠重。 对照品多西他赛注射液腹腔注射给药, 给药剂量分别为 30mg/kg, 每周 1 次。 N- ( (4-氯 -3-三氟甲基) 苯基) -N,- (2-氟 -4- ( (2-羟甲基氨基甲酰基) -4-吡啶基 氧) 苯基) 脲和对照品索拉非尼给药剂量分别为 30mg/kg和 10mg/kg, 每日 1 次。阴性对照(蓖麻油聚氧乙烯醚: 95%乙醇: 0.9%生理盐水 =10%: 10%: 80%) 腹腔给药, 每日 1次。 连续给药 21天。 实验结果如表 1。 表 1. 本发明化合物对人肝癌 HepG2裸小鼠移植瘤的治疗作用
Figure imgf000016_0002
* P<0.05 ; **P<0.01 具体实施方式
以下通过具体实施方式进一
解为对本发明保护范围的限制。 实施例 1
4-氯吡啶 -2-甲酰胺的制备
Figure imgf000016_0001
将 4-氯吡啶 -2-甲酰氯 (20g) 溶于甲苯 (200mL), 室温, 2 hr内滴加甲醇 (200mL),然后向反应液中加入水和乙酸乙酯,静置分层,有机层用无水 Na2S04 干燥, 减压蒸出溶剂的淡黄色油状物。 用甲醇 (200mL) 溶解, 然后加入甲苯 (200mL), 加入氨水 (200mL), 室温搅拌过夜。 分层, 有机层减压蒸出溶剂, 硅胶柱纯化得白色固体 (14.3g, 80.4%)。 实施例 2
2-氨基甲酰基 -4- ( -氟 -4-氨基) 苯氧基) 吡啶的制备
Figure imgf000017_0001
将 4-氯吡啶 -2-甲酰胺(2.5g)和 3-氟 -4-氨基苯酚(2.25g)溶于 DMF ( 16mL), 然后加入 t-BuOK ( 1.9g), 165°C搅拌 75分钟, 减压蒸出溶剂, 硅胶柱纯化得 白色固体 (1.8g, 45.6%)。 实施例 3
N- ( (4-氯 -3-三氟甲基) 苯基) -N,- (2-氟 -4- ( (2-氨基甲酰基) -4-吡啶基氧) 苯基) 脲的制备
Figure imgf000017_0002
2-氨基甲酰基 -4- ( (3-氟 -4-氨基) 苯氧基) 吡啶 (3.5g) 溶于预算一直乙酸 乙酯 (20mL), 然后加入 3-三氟甲基 -4-氯苯基异氰酸酯 (2.9g), 60°C搅拌 4小 时,减压蒸出溶剂,硅胶柱纯化得白色固体(1.2g, 18.1%)。 lHNMR (DMSO-d6): δ=7.18 (d, 1H), δ=7.20 (m, 1H), 5=7.32 (m, 1H), 5=7.40 (d, 1H), 5=7.61 (d, 2H), 5=7.72 (s, 1H), 5=8.18 (m, 3H), 5=8.52 (d, 1H), 5=8.73 (s, 1H), 5=9.51 (s, lH m/e=469.26。 实施例 4
N- ( (4-氯 -3-三氟甲基) 苯基) -N,- (2-氟 -4- ( (2-羟甲基氨基甲酰基) -4-吡 啶基氧) 苯基) 脲制备
N- ( (4-氯 -3-三氟甲基) 苯基) -N,- (2-氟 -4- ( (2-氨基甲酰基) -4-吡啶基氧) 苯基)脲(lg)和多聚甲醛(320mg)溶于 16mL四氢呋喃和 8mL水, 加入 20mg 碳酸钾, 100°C, 微波条件下, 搅拌 30min, 减压蒸出溶剂, 硅胶柱纯化, 得白 色固体。 1HNMR (DMSO-d6): δ=4·69 (d, 2Η), δ=7.05 (d, 1H), δ=7.18 (m, 1H), 5=7.31 (d, 1H), 5=7.45 (s, 1H), 5=7.61 (s, 2H), 5=8.11 (m, 2H), 5=8.53 (d, 1H), 5=8.75 (s, 1H), 5=9.26 (m, 1H), 5=9.53 (s, 1H)。 m/e=498.81。

Claims

权利要求
1、 式 (I) N- ( (4-氯 -3-三氟甲基) 苯基) -Ν,- (2-氟 -4- ( (2-羟甲基氨基甲酰 基) -4-吡啶基氧) 苯基) 脲或其药学上可接受的盐:
Figure imgf000018_0001
式 (1)。
2、 权利要求 1所述的化合物, 它是式 (I)化合物或其药学上可接受的盐, 选 自:
a)有机酸和无机酸的碱式盐,所述有机酸和无机酸选自盐酸、氢溴酸、硫酸, 磷酸、 甲磺酸、 三氟磺酸、 苯磺酸、 对甲苯磺酸、 1-萘磺酸、 2-萘磺酸、 乙酸, 三氟乙酸、 苹果酸、 酒石酸、 柠檬酸、 乳酸、 草酸、 琥珀酸、 富马酸、 马来酸, 苯甲酸、 水杨酸、 苯基乙酸或杏仁酸; 和
b)含阳离子的有机和无机碱的酸式盐, 所述阳离子选自碱金属阳离子、碱土 金属阳离子、 铵离子、 脂族取代的铵离子或芳族取代的铵离子。
3、 用于治疗癌症的药物组合物, 包含权利要求 1所述的化合物或其药学上 可接受的盐, 和生理上合适的载体。
4、 权利要求 1所述的化合物或其药学上可接受的盐用于制造抑制 VEGFR-2 等激酶介导疾病的药物的用途。
5、 权利要求 1所述的化合物或其药学上可接受的盐用于制造抑制 VEGFR-2 等激酶介导的癌性细胞生长和转移的药物的用途。
6、 权利要求 1所述的化合物或其药学上可接受的盐在制备治疗癌症疾病药 物上的用途。
7、 权利要求 6所述的用途, 其中所述的癌症是实质癌、 肾癌、 肺癌、 乳腺 癌、 肝癌、 卵巢癌、 胰腺癌、 甲状腺癌、 膀胱癌、 白血病、 黑素瘤、 胃癌, 结肠 直肠癌、 内分泌癌、 急性髓样白血病 (AML)、 骨髓增生异常综合征 (MDS)、 间皮 癌、 血管肉瘤、 肾上腺皮质腺癌、 子宫内膜癌及神经胶质瘤。
8、 权利要求 6所述的用途, 其中所述癌症是肾癌、 肝癌、 肺癌、 结肠直肠 癌、 胃癌、 乳腺癌及血管肉瘤。
PCT/CN2012/085791 2012-01-17 2012-12-04 N-((4-氯-3-三氟甲基)苯基)-n'-(2-氟-4-((2-羟甲基氨基甲酰基)-4-吡啶基氧)苯基)脲及其作为抗癌药物的应用 WO2013107225A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201210012640.5 2012-01-17
CN2012100126405A CN102885814A (zh) 2012-01-17 2012-01-17 一种化合物及其作为抗癌药物的应用

Publications (1)

Publication Number Publication Date
WO2013107225A1 true WO2013107225A1 (zh) 2013-07-25

Family

ID=47529635

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2012/085791 WO2013107225A1 (zh) 2012-01-17 2012-12-04 N-((4-氯-3-三氟甲基)苯基)-n'-(2-氟-4-((2-羟甲基氨基甲酰基)-4-吡啶基氧)苯基)脲及其作为抗癌药物的应用

Country Status (2)

Country Link
CN (1) CN102885814A (zh)
WO (1) WO2013107225A1 (zh)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103508944A (zh) * 2012-06-25 2014-01-15 湖南有色凯铂生物药业有限公司 N-((4-氯-3-三氟甲基)苯基)-n’-((2-氟-4-(2-氨基甲酰基)-4-吡啶基氧)苯基)脲的晶型a及其制备方法
CN104288147A (zh) * 2013-07-17 2015-01-21 湖南有色凯铂生物药业有限公司 芳香脲类化合物在制备治疗实体瘤药物中的用途
CN112294971B (zh) * 2020-02-20 2022-02-01 深圳市泰力生物医药有限公司 具有改进的溶解性的尼洛替尼组合物
CN112159351B (zh) * 2020-09-21 2021-12-07 广州南鑫药业有限公司 一种多靶点抗肿瘤药物的制备方法

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011130728A1 (en) * 2010-04-17 2011-10-20 Bayer Healthcare Llc Synthetic metabolites of fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention diseases and conditions

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BRPI0610048A2 (pt) * 2005-05-27 2010-05-25 Bayer Healthcare Ag terapia de combinação que compreende um composto de diaril uréia e uma pi3, akt quinase ou inibidores de mtor (rapamicinas) para o tratamento de cáncer
UA91385C2 (ru) * 2005-11-10 2010-07-26 Баер Шеринг Фарма Акциэнгезельшафт Диарилмочевина для лечения легочной гипертензии

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011130728A1 (en) * 2010-04-17 2011-10-20 Bayer Healthcare Llc Synthetic metabolites of fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention diseases and conditions

Also Published As

Publication number Publication date
CN102885814A (zh) 2013-01-23

Similar Documents

Publication Publication Date Title
JP5649132B2 (ja) N−[4−(1−シアノシクロペンチル)フェニル]−2−(4−ピリジルメチル)アミノ−3−ピリジンカルボキサミドの塩
TWI693225B (zh) 一種egfr抑制劑的鹽、晶型及其用途
WO2014040242A1 (zh) 3-氯-及3-甲氧基-n-甲基-2-吡啶酰胺化合物及其作为抗癌药物的应用
CN103102342B (zh) 氨基喹唑啉类衍生物及其盐和使用方法
CN103565653B (zh) 取代的吡唑酮化合物及其使用方法和用途
TW201806598A (zh) 治療癌症及骨癌疼痛之方法
KR20160048807A (ko) 과증식성 질병의 치료에서 사용하기 위한 mek 억제제 및 erk 억제제의 조합
CN108503650B (zh) 二噁烷并喹唑啉类化合物或其药用盐或其水合物及其作为酪氨酸激酶抑制剂的应用
CN102731385A (zh) 3-氯-及3-甲氧基-n-甲基-2-吡啶酰胺化合物及其作为抗癌药物的应用
CN103420986A (zh) 取代的喹啉化合物及其使用方法和用途
TW202200581A (zh) Sik—3抑制劑及其用途
WO2013107225A1 (zh) N-((4-氯-3-三氟甲基)苯基)-n&#39;-(2-氟-4-((2-羟甲基氨基甲酰基)-4-吡啶基氧)苯基)脲及其作为抗癌药物的应用
CN102643229A (zh) N-((4-氯-3-三氟甲基)苯基)-n’-((2-氟-4-(2-氨基甲酰基)-4-吡啶基氧)苯基)脲及其作为抗癌药物的应用
KR20100029097A (ko) 안트라닐아미드 피리딘우레아와 벤즈아미드 유도체의 상승작용적 조합물
CN102775385A (zh) N-取代苯基-n’-取代杂环脲类化合物及其作为抗癌药物的应用
CN108148071B (zh) 氨基喹唑啉衍生物的盐及其用途
JP6239103B2 (ja) チロシンキナーゼ阻害活性を有する物質ならびにその調製方法および使用
WO2013037292A1 (zh) 取代的芳香脲类化合物及其作为抗癌药物的应用
WO2013037293A1 (zh) N-吲哚-1-酰胺类化合物及作为抗癌药物的应用
WO2015182625A1 (ja) Ras活性阻害薬及びその用途
WO2014040243A1 (zh) N-取代苯基-n&#39;-取代杂环脲类化合物及其作为抗癌药物的应用
CN106176757B (zh) 一种化合物与替吉奥联合在制备治疗增生性疾病中的药物中的用途
WO2023239945A1 (en) Btological activities of 5-(2-(4-(4-fluoro-2-methyl-1h-indol-5- yloxy)-6-methoxyquinolin-7-yloxy)ethyl)-5-azaspiro[2.4]-heptan- 7-ol crystalline, phosphoric acid salt and its enantiomers
TWI462738B (zh) N-〔4-(1-氰基環戊基)苯基〕-2-(4-吡啶甲基)胺基-3-吡啶甲醯胺的鹽
CN108864101A (zh) 氘代sgx523及其用途

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: 12865935

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12865935

Country of ref document: EP

Kind code of ref document: A1