WO2021089005A1 - Use of fgfr inhibitor - Google Patents

Use of fgfr inhibitor Download PDF

Info

Publication number
WO2021089005A1
WO2021089005A1 PCT/CN2020/127211 CN2020127211W WO2021089005A1 WO 2021089005 A1 WO2021089005 A1 WO 2021089005A1 CN 2020127211 W CN2020127211 W CN 2020127211W WO 2021089005 A1 WO2021089005 A1 WO 2021089005A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
fgfr
cholangiocarcinoma
cancer
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2020/127211
Other languages
French (fr)
Chinese (zh)
Inventor
王彩霞
王玲玲
张阳
李桂霞
祁欢欢
赵晶
李筱
Original Assignee
石药集团中奇制药技术(石家庄)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 石药集团中奇制药技术(石家庄)有限公司 filed Critical 石药集团中奇制药技术(石家庄)有限公司
Priority to CN202080076195.1A priority Critical patent/CN114641293A/en
Publication of WO2021089005A1 publication Critical patent/WO2021089005A1/en

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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the invention belongs to the field of medicine. More specifically, the present invention relates to an FGFR inhibitor or a pharmaceutically acceptable salt thereof for the treatment of FGFR-related tumors, a pharmaceutical composition containing the same, a method for treating FGFR-related tumors using a drug containing the same, and the preparation thereof Use in drugs for treating FGFR-related tumors.
  • Fibroblast growth factor receptor is a receptor for fibroblast growth factor (FGF) signal transduction, and its family consists of four members (FGFR1-4). Through FGFR, FGF plays an important role in many physiological regulation processes such as cell proliferation, cell differentiation, cell migration and angiogenesis.
  • FGF signaling pathway high expression, gene amplification, gene mutation, chromosome recombination, etc.
  • pathological processes such as tumor cell proliferation, migration, invasion and angiogenesis
  • FGFR is in many tumors
  • FGFR2 fusion mutations occur in about 10-20% of patients with intrahepatic cholangiocarcinoma; FGFR3 gene changes in bladder cancer have a strong correlation with low-grade pathological and clinical low-stage cancers, more than 70%
  • the pathological low-grade non-invasive urothelial papilloma contains FGFR3 mutation; and high expression of FGFR1, FGFR2 and FGFR3 are found in gastric cancer tissues and gastric cancer cells. Therefore, FGFR has become an important therapeutic target, attracting a wide range of research and development interest.
  • inhibitors that target the kinase domain of the FGFR membrane can be divided into ATP-competitive inhibitors, non-ATP-competitive reversible inhibitors, and irreversible inhibitors.
  • non-ATP-competitive reversible inhibitors and irreversible inhibitors affect kinases.
  • the inhibitory activity is not affected by the high ATP concentration in the cell and in the body.
  • some FGFR-targeted drugs for the treatment of the above-mentioned tumor diseases have entered the clinical trial stage.
  • Balversa Erdafitinib
  • FGFR reversible inhibitor targeted drugs have become the world's first pandemic drug for metastatic urothelial cancer approved by the FDA.
  • FGFR1-4 inhibitors for example, BGJ-398, Debio-1347 and TAS-120, etc.
  • solid tumors such as urothelial cell carcinoma and liver cancer.
  • the structural formulas of the above drugs are as follows:
  • pan-FGFR pan-FGFR
  • irreversible inhibitors such as TAS-120 Patients who are resistant to BGJ-398 are still effective, showing the unique advantages of irreversible inhibitors. But so far, no FGFR1-4 irreversible inhibitor has been approved for marketing at home and abroad.
  • liver cancer Primary liver cancer
  • liver cancer is a relatively common malignant tumor and the fourth leading cause of cancer deaths in the world, posing a serious threat to human life and health.
  • Liver cancer mainly includes hepatocellular carcinoma (HCC), intrahepatic cholangiocarcinoma (ICC), and combined hepatocellular cholangiocarcinoma (c-CC-CC).
  • HCC hepatocellular carcinoma
  • ICC intrahepatic cholangiocarcinoma
  • c-CC-CC combined hepatocellular cholangiocarcinoma
  • Liver cancer has no obvious clinical symptoms in the early stage, and is often found in the middle and late stages, with high malignancy, high recurrence rate, poor treatment effect, and poor prognosis.
  • the clinical treatment of liver cancer mainly includes surgical treatment, transcatheter arterial chemoembolization (TACE), radiotherapy and chemotherapy.
  • TACE transcatheter arterial chemoembolization
  • Sorafenib is currently the only molecularly targeted drug approved for the treatment of advanced liver cancer, but its effect on liver cancer patients with liver function Child-Pugh grade B is still poor.
  • Urothelial cancer is a common malignant tumor worldwide, and it is also one of the most common clinical malignancies in urology in my country. Among them, 90%-95% of urothelial cancers are bladder cancer. Urothelial cell carcinoma is mostly non-muscular invasive at the first diagnosis, but it has a high recurrence rate; and with the increase in the number of recurrences, the malignancy of the tumor will increase and turn into a muscular invasive tumor. Patients with metastatic urothelial cancer with FGFR gene alterations have a poor prognosis and a low response rate to treatment. There are significant clinical needs that are far from being met in this type of patients.
  • Gastric cancer is also one of the most common malignant tumors in the world, with a relatively poor prognosis and a serious threat to human health. Due to the lack of a mature early screening system and the atypical symptoms of early gastric cancer and the low detection rate, most patients are already in the advanced stage when they are diagnosed.
  • Today's treatment measures for gastric cancer mainly include surgical treatment, systemic application of chemical drugs, radiotherapy and molecular targeted drug therapy. Targeted therapy is a drug treatment aimed at specific tumor targets.
  • clinical studies on targeted therapy of gastric cancer have few successes and many failures.
  • Cholangiocarcinoma is a malignant tumor of epithelial cells with different characteristics of cholangiocarcinoma. The incidence has increased by nearly 20% in the past 10 years, accounting for about 3% of gastrointestinal tumors and 10-15% of hepatobiliary malignancies. According to anatomical location, cholangiocarcinoma can be divided into intrahepatic cholangiocarcinoma and extrahepatic cholangiocarcinoma (eCCA), the latter is further divided into hilar cholangiocarcinoma (pCCA) and distal cholangiocarcinoma (distal cholangiocarcinoma) , DCCA).
  • pCCA hilar cholangiocarcinoma
  • DCCA distal cholangiocarcinoma
  • FGFR inhibitors have shown promise in clinical trials of cholangiocarcinoma.
  • the FGFR inhibitor BGJ-398 has obtained excellent phase II trial results in advanced cholangiocarcinoma with FGFR gene fusion, mutation, and amplification, and objective remission
  • the rate and disease control rate in FGFR gene fusion patients reached 18.8% and 83.3%.
  • Intrahepatic cholangiocarcinoma refers to malignant tumors of bile duct epithelium located in the liver at level 2 and above. It is also called intrahepatic cholangiocarcinoma. It belongs to a type of primary liver cancer (accounting for 10% to 15%). Can be attributed to a type of cholangiocarcinoma (about 10%). Intrahepatic cholangiocarcinoma is highly malignant, has strong invasion and lymph node metastasis characteristics, and is difficult to diagnose early. The prognosis of the patient is poor. The overall 5-year survival rate is less than 10%, and the median survival time after surgical resection is 36 months.
  • Intrahepatic cholangiocarcinoma is insensitive to traditional chemotherapy, radiotherapy and recent tumor immunotherapy. There is no standard treatment for cholangiocarcinoma patients who have failed chemotherapy with the first-line gemcitabine.
  • FGFR inhibitors are in different stages of clinical research on cholangiocarcinoma, such as BGJ-398 (reversible selective inhibitor), ARQ087 (ATP competitive inhibitor), TAS-120 (irreversible pan-FGFR inhibitor) Et al.
  • the first FGFR inhibitor reported in cholangiocarcinoma was BGJ-398.
  • Further studies found polyclonal secondary mutations in the kinase domain of FGFR2, including 3 cases The FGFR2V564F gene mutation exists in all patients.
  • TAS-120 is a highly selective and irreversible pan-FGFR inhibitor. Studies have shown that TAS-120 has clinical effects on BGJ-398-resistant intrahepatic cholangiocarcinoma patients, and can inhibit a variety of secondary FGFR2 mutations. However, there is no literature that discloses the cell-level inhibitory activity data of TAS-120. Currently, no targeted drugs for the treatment of intrahepatic cholangiocarcinoma have been approved for marketing.
  • WO2019034076A1 discloses FGFR inhibitors and their medical uses, including compound A (Example 2).
  • This patent application discloses the evaluation results of compound A on FGFR wild-type kinase in vitro inhibitory activity, the evaluation results on mutant kinase in vitro inhibitory activity and the pharmacokinetic evaluation results in mice, but it does not disclose which diseases Compound A can be used for. the treatment.
  • An object of the present invention is to provide a use of Fibroblast Growth Factor Receptor (FGFR) inhibitor compound A or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of FGFR-related tumors.
  • FGFR Fibroblast Growth Factor Receptor
  • Another object of the present invention is to provide an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof, which is used for the treatment of FGFR-related tumors.
  • Another object of the present invention is to provide a method for treating FGFR-related tumors, the method comprising administering to a subject or patient a drug containing a therapeutically effective amount of an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof.
  • Another object of the present invention is to provide a pharmaceutical composition for the treatment of FGFR-related tumors, which comprises an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof and optionally a pharmaceutically acceptable carrier.
  • a technical problem to be solved by the present invention is to provide a small molecule compound with excellent FGFR1-4 wild-type and mutant kinase inhibitory activity, and a pharmaceutical composition, use and treatment method thereof.
  • Another technical problem to be solved by the present invention is to provide a small molecule with excellent FGFR1-4 wild-type and mutant kinase inhibitory activity and excellent in vitro/in vivo anti-FGFR-related tumors (especially digestive or urinary system tumors) activity Compound and its pharmaceutical composition, use and treatment method.
  • Another technical problem to be solved by the present invention is to provide an excellent FGFR1-4 wild-type and mutant kinase inhibitory activity and excellent in vitro/in vivo anti-FGFR-related tumor (especially digestive or urinary system tumors) activity, and has Small molecule compounds with good safety and their pharmaceutical compositions, uses and treatment methods.
  • Another technical problem to be solved by the present invention is to provide an excellent FGFR1-4 wild-type and mutant kinase inhibitory activity and excellent in vitro/in vivo anti-FGFR-related tumor (especially digestive or urinary system tumors) activity, and has Small molecule compounds with excellent plasma stability and safety, and pharmaceutical compositions, uses and treatment methods thereof.
  • an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of FGFR-related tumors, wherein the compound A has The structure is as follows:
  • the FGFR-related tumor is one or more of digestive or urinary system tumors.
  • the FGFR-related tumor is any one of gastric cancer, liver cancer, urothelial cancer, cholangiocarcinoma, or any combination thereof.
  • the FGFR-related tumor is gastric cancer.
  • the gastric cancer is gastric cancer with FGFR2 gene amplification.
  • the FGFR-related tumor is liver cancer.
  • the liver cancer is a liver cancer with high FGFR4/FGF19 expression.
  • the liver cancer is a liver cancer with high FGFR3 expression.
  • the liver cancer is any one of hepatocellular carcinoma, intrahepatic cholangiocarcinoma, mixed liver cancer, or any combination thereof.
  • the FGFR-related tumor is urothelial carcinoma.
  • the urothelial cancer is bladder cancer.
  • the bladder cancer is a bladder cancer with high FGFR3 expression and FGFR3-TACC3 fusion.
  • the FGFR-related tumor is cholangiocarcinoma.
  • the cholangiocarcinoma is a cholangiocarcinoma with high FGFR2 expression.
  • the cholangiocarcinoma is any one of intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, distal cholangiocarcinoma, or any combination thereof.
  • the cholangiocarcinoma is intrahepatic cholangiocarcinoma.
  • the compound A or a pharmaceutically acceptable salt thereof is the only active ingredient in the drug.
  • the compound A or a pharmaceutically acceptable salt thereof is used in combination with one or more other targeted drugs or chemotherapeutics.
  • the drug is formulated into a clinically accepted formulation.
  • the preparation is an oral preparation, an injection preparation or a topical preparation.
  • the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.001 mg/kg to about 1000 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.01 mg/kg to about 100 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.02 mg/kg to about 50 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.03 mg/kg to about 20 mg/kg.
  • the medicament contains a therapeutically effective amount of Compound A or a pharmaceutically acceptable salt thereof.
  • the therapeutically effective amount is 0.001-1000 mg.
  • the therapeutically effective amount is 0.01-100 mg.
  • the therapeutically effective amount is 0.1-50 mg.
  • the therapeutically effective amount is 0.5-30 mg.
  • the compound A or a pharmaceutically acceptable salt thereof is administered in a single dose or in divided doses.
  • the drug is administered orally, by injection, topical, or in vitro. In a preferred embodiment, the drug is administered orally or by injection.
  • an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof for the treatment of FGFR-related tumors wherein the compound A has the following structure:
  • the FGFR-related tumor is one or more of digestive or urinary system tumors.
  • the FGFR-related tumor is any one of gastric cancer, liver cancer, urothelial cancer, cholangiocarcinoma, or any combination thereof.
  • the FGFR-related tumor is gastric cancer.
  • the gastric cancer is gastric cancer with FGFR2 gene amplification.
  • the FGFR-related tumor is liver cancer.
  • the liver cancer is a liver cancer with high FGFR4/FGF19 expression.
  • the liver cancer is a liver cancer with high FGFR3 expression.
  • the liver cancer is any one of hepatocellular carcinoma, intrahepatic cholangiocarcinoma, mixed liver cancer, or any combination thereof.
  • the FGFR-related tumor is urothelial carcinoma.
  • the urothelial cancer is bladder cancer.
  • the bladder cancer is a bladder cancer with high FGFR3 expression and FGFR3-TACC3 fusion.
  • the FGFR-related tumor is cholangiocarcinoma.
  • the cholangiocarcinoma is a cholangiocarcinoma with high FGFR2 expression.
  • the cholangiocarcinoma is any one of intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, distal cholangiocarcinoma, or any combination thereof.
  • the cholangiocarcinoma is intrahepatic cholangiocarcinoma.
  • the compound A or a pharmaceutically acceptable salt thereof is used as the sole active ingredient.
  • the compound A or a pharmaceutically acceptable salt thereof is used in combination with one or more other targeted drugs or chemotherapeutic drugs.
  • the compound A or a pharmaceutically acceptable salt thereof is formulated into a clinically acceptable preparation.
  • the preparation is an oral preparation, an injection preparation or a topical preparation.
  • the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.001 mg/kg to about 1000 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.01 mg/kg to about 100 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.02 mg/kg to about 50 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.03 mg/kg to about 20 mg/kg.
  • the compound A or a pharmaceutically acceptable salt thereof is formulated in a medicament in a therapeutically effective amount.
  • the therapeutically effective amount is 0.001-1000 mg.
  • the therapeutically effective amount is 0.01-100 mg.
  • the therapeutically effective amount is 0.1-50 mg.
  • the therapeutically effective amount is 0.5-30 mg.
  • the compound A or a pharmaceutically acceptable salt thereof is administered in a single dose or in divided doses.
  • the compound A or a pharmaceutically acceptable salt thereof is administered orally, by injection, topical, or in vitro. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered by oral administration or injection.
  • FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof Drug, wherein the compound A has the following structure:
  • the FGFR-related tumor is one or more of digestive or urinary system tumors.
  • the FGFR-related tumor is any one of gastric cancer, liver cancer, urothelial cancer, cholangiocarcinoma, or any combination thereof.
  • the FGFR-related tumor is gastric cancer.
  • the gastric cancer is gastric cancer with FGFR2 gene amplification.
  • the FGFR-related tumor is liver cancer.
  • the liver cancer is a liver cancer with high FGFR4/FGF19 expression.
  • the liver cancer is a liver cancer with high FGFR3 expression.
  • the liver cancer is any one of hepatocellular carcinoma, intrahepatic cholangiocarcinoma, mixed liver cancer, or any combination thereof.
  • the FGFR-related tumor is urothelial carcinoma.
  • the urothelial cancer is bladder cancer.
  • the bladder cancer is a bladder cancer with high FGFR3 expression and FGFR3-TACC3 fusion.
  • the FGFR-related tumor is cholangiocarcinoma.
  • the cholangiocarcinoma is a cholangiocarcinoma with high FGFR2 expression.
  • the cholangiocarcinoma is any one of intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, distal cholangiocarcinoma, or any combination thereof.
  • the cholangiocarcinoma is intrahepatic cholangiocarcinoma.
  • the compound A or a pharmaceutically acceptable salt thereof is administered as the sole active ingredient.
  • the compound A or a pharmaceutically acceptable salt thereof is administered in combination with one or more other targeted drugs or chemotherapeutic drugs.
  • the drug is formulated into a clinically accepted formulation.
  • the preparation is an oral preparation, an injection preparation or a topical preparation.
  • the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.001 mg/kg to about 1000 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.01 mg/kg to about 100 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.02 mg/kg to about 50 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.03 mg/kg to about 20 mg/kg.
  • the therapeutically effective amount is 0.001-1000 mg. In a preferred embodiment, the therapeutically effective amount is 0.01-100 mg. In a preferred embodiment, the therapeutically effective amount is 0.1-50 mg. In a preferred embodiment, the therapeutically effective amount is 0.5-30 mg.
  • the compound A or a pharmaceutically acceptable salt thereof is administered in a single dose or in divided doses.
  • the compound A or a pharmaceutically acceptable salt thereof is administered orally, by injection, topical, or in vitro. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered by oral administration or injection.
  • a pharmaceutical composition for the treatment of FGFR-related tumors which comprises an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof and optionally a pharmaceutically acceptable , wherein the compound A has the following structure:
  • the FGFR-related tumor is one or more of digestive or urinary system tumors.
  • the FGFR-related tumor is any one of gastric cancer, liver cancer, urothelial cancer, cholangiocarcinoma, or any combination thereof.
  • the FGFR-related tumor is gastric cancer.
  • the gastric cancer is gastric cancer with FGFR2 gene amplification.
  • the FGFR-related tumor is liver cancer.
  • the liver cancer is a liver cancer with high FGFR4/FGF19 expression.
  • the liver cancer is a liver cancer with high FGFR3 expression.
  • the liver cancer is any one of hepatocellular carcinoma, intrahepatic cholangiocarcinoma, mixed liver cancer, or any combination thereof.
  • the FGFR-related tumor is urothelial carcinoma.
  • the urothelial cancer is bladder cancer.
  • the bladder cancer is a bladder cancer with high FGFR3 expression and FGFR3-TACC3 fusion.
  • the FGFR-related tumor is cholangiocarcinoma.
  • the cholangiocarcinoma is a cholangiocarcinoma with high FGFR2 expression.
  • the cholangiocarcinoma is any one of intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, distal cholangiocarcinoma, or any combination thereof.
  • the cholangiocarcinoma is intrahepatic cholangiocarcinoma.
  • the compound A or a pharmaceutically acceptable salt thereof is the only active ingredient in the pharmaceutical composition.
  • the pharmaceutical composition also contains one or more other targeted drugs or chemotherapeutic drugs as active ingredients.
  • the pharmaceutical composition is formulated into a clinically accepted formulation.
  • the preparation is an oral preparation, an injection preparation or a topical preparation.
  • the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.001 mg/kg to about 1000 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.01 mg/kg to about 100 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.02 mg/kg to about 50 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.03 mg/kg to about 20 mg/kg.
  • the pharmaceutical composition contains a therapeutically effective amount of Compound A or a pharmaceutically acceptable salt thereof.
  • the therapeutically effective amount is 0.001-1000 mg.
  • the therapeutically effective amount is 0.01-100 mg.
  • the therapeutically effective amount is 0.1-50 mg.
  • the therapeutically effective amount is 0.5-30 mg.
  • the compound A or a pharmaceutically acceptable salt thereof is administered in a single dose or in divided doses.
  • the pharmaceutical composition is administered by oral administration, injection administration, topical administration or in vitro administration. In a preferred embodiment, the pharmaceutical composition is administered by oral administration or injection.
  • the present invention evaluated the in vitro kinase inhibitory activity of compound A against FGFR1-4 wild-type and mutant types and compared with The proliferation inhibitory activity of FGFR-related exemplary digestive or urinary system tumors (including gastric cancer, liver cancer, bladder cancer, and cholangiocarcinoma) models, and the inhibitory effect of compound A on tumor growth was further evaluated for several tumor xenograft models.
  • FGFR-related exemplary digestive or urinary system tumors including gastric cancer, liver cancer, bladder cancer, and cholangiocarcinoma
  • the present invention measured the plasma stability of compound A in the human and mouse plasma stability test, and the structure was similar to the reference compound (Example 8 in WO2019034076A1 (S configuration)) A comparison was made.
  • the results of plasma stability show that compound A has better stability in both mouse and human plasma.
  • irreversible inhibitors enhance their affinity with the target through covalent bonding with the target protein, which is the fundamental reason why irreversible inhibitors exhibit high biological activity.
  • affinity-enhancing effect of irreversible inhibitors will also occur on off-target sites, resulting in enhanced toxic and side effects [see Yang Bo et al. Research Progress on Small Molecule Covalent Inhibitors, Acta Pharmaceutical Sciences, 2014 , 49(2):158-165].
  • the present invention detects the off-target effects of compound A on a variety of important non-target kinases, and the results show that compound A has low off-target effects.
  • the present invention conducted safety pharmacology tests and acute toxicity tests on dogs and mice. The results show that compound A has no significant effect on the detection indicators, and the safety is good.
  • compound A has the activity of selectively inhibiting FGFR1-4, has good inhibitory activity on both wild-type and mutant FGFR1-4, and can significantly inhibit the abnormality of FGFR1-4.
  • Compound A refers to a compound with the following structure as an FGFR inhibitor:
  • “Pharmaceutically acceptable salt” refers to a conventional non-toxic salt formed by the reaction of Compound A of the present invention with an inorganic acid, organic acid, inorganic base or organic base.
  • the salt is within the scope of reliable medical judgment and is suitable for use in contact with human and animal tissues without excessive toxicity, irritation, allergic reactions or other problems or complications, and reasonable benefits/risks Commensurate.
  • a pharmaceutically acceptable base addition salt or acid addition salt can be obtained by contacting compound A with a sufficient amount of non-toxic acid or base in a pure solution or a suitable inert solvent.
  • “Pharmaceutically acceptable carrier” refers to a carrier suitable for formulating a pharmaceutical composition of Compound A or a pharmaceutically acceptable salt thereof or a clinically accepted preparation thereof.
  • the carrier is within the scope of reliable medical judgment and is suitable for use in contact with human and animal tissues without excessive toxicity, irritation, allergic reactions or other problems or complications, and reasonable benefits/risks Commensurate.
  • FGFR refers to fibroblast growth factor receptor, which is a receptor for fibroblast growth factor signaling, and its family consists of four members (FGFR1-4). Through FGFR, FGF plays an important role in many physiological regulation processes such as cell proliferation, cell differentiation, cell migration and angiogenesis.
  • FGFR-related tumors refer to tumors whose occurrence or development is closely related to the expression and activation of any one or any combination of FGFR1-4, including but not limited to gastric cancer, bladder cancer, urothelial cancer, liver cancer, Cholangiocarcinoma (eg intrahepatic cholangiocarcinoma).
  • Targeteted drug refers to a targeted drug that is clinically used to treat tumor-related diseases. It is endowed with a targeting ability to enable the active ingredient or its carrier to target a specific target lesion site, and accumulate or release the active ingredient at that site , To form a relatively high concentration, thereby improving the curative effect while inhibiting side effects and reducing damage to normal tissues and cells.
  • “Chemotherapeutic drugs” refer to drugs that can act on different links in the growth and reproduction of tumor cells, inhibit or kill tumor cells, and are clinically used to treat tumor-related diseases. It is currently one of the main methods of treating tumors.
  • Clinically accepted preparations are within the scope of reliable medical judgment, suitable for use in contact with human and animal tissues, without excessive toxicity, irritation, allergic reactions or other problems or complications, and A formulation with a reasonable benefit/risk ratio commensurate.
  • the clinically accepted preparations include oral preparations, injection preparations, and topical preparations.
  • the dosage and frequency of administration of the compound A or its pharmaceutically acceptable salt can be performed by conventional methods such as modeling, dose escalation studies, or clinical trials, and by considering the characteristics and severity of the disease to be treated. The degree, age, general condition and weight of the patient, as well as the specific compound administered, its pharmacokinetic properties, and the route of administration are determined.
  • a suitable daily dosage range of the compound A or a pharmaceutically acceptable salt thereof is from about 0.001 mg/kg to about 1000 mg/kg; preferably, from about 0.01 mg/kg to about 100 mg/kg; further Preferably, from about 0.02mg/kg to about 50mg/kg; still more preferably, from about 0.03mg/kg to about 20mg/kg (wherein, "kg” refers to the administration object (ie, the subject Or the weight of the patient).
  • the daily dose of compound A or its pharmaceutically acceptable salt is 0.001 mg-1000 mg, and more preferably, the daily dose of compound A or its pharmaceutically acceptable salt is 0.01-1000 mg.
  • the daily dose of Compound A or its pharmaceutically acceptable salt is 0.1-50 mg; More preferably, the daily dose of Compound A or its pharmaceutically acceptable salt is 0.5-30mg; More preferably, the daily dosage of Compound A or its pharmaceutically acceptable salt is 0.1mg, 0.5mg, 1mg, 2mg, 5mg, 10mg, 15mg, 20mg, 25mg, 30mg, 35mg, 36mg, 40mg, 45mg, 50mg, 55mg, 60mg, 70mg, 72mg, 100mg, 200mg, 500mg, 800mg, 1000mg, given in single or divided doses.
  • the compound A or a pharmaceutically acceptable salt thereof is included in the drug or pharmaceutical composition in a therapeutically effective amount.
  • the therapeutically effective amount is preferably 0.001-1000 mg, more preferably 0.01-100 mg, still more preferably 0.1-50 mg, still more preferably 0.5-30 mg, administered in a single dose or in divided doses.
  • the therapeutically effective amount, administration dose or administration dose of the compound A or a pharmaceutically acceptable salt thereof is calculated as compound A.
  • subjects or patients suffering from FGFR-related tumors can be human and non-human mammals such as mice, rats, guinea pigs, cats, dogs, cows, horses, sheep, pigs, monkeys, etc., more preferably humans. .
  • FGFR fibroblast growth factor receptor
  • liver cancer is hepatocellular carcinoma, intrahepatic cholangiocarcinoma, and mixed liver cancer.
  • the drug contains a therapeutically effective amount of the inhibitor, the therapeutically effective amount is preferably 0.001-1000 mg; it can be administered in a single dose or in divided doses.
  • the present invention also relates to the following embodiments:
  • cholangiocarcinoma includes intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma and distal cholangiocarcinoma; preferably intrahepatic cholangiocarcinoma.
  • the medicine contains a therapeutically effective amount of Compound A or a pharmaceutically acceptable salt thereof, and the therapeutically effective dose is preferably 0.001 mg-1000 mg, more preferably 0.01-100 mg, more preferably 0.1-50 mg, still more preferably 0.5-30 mg; it can be administered in a single dose or in divided doses.
  • a method for treating FGFR-related tumor diseases characterized in that a drug containing a therapeutically effective dose of Compound A or a pharmaceutically acceptable salt thereof is administered to a subject or patient.
  • the cholangiocarcinoma includes intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, and distal cholangiocarcinoma; preferably, it is intrahepatic cholangiocarcinoma.
  • the administration may be oral administration, injection administration, topical administration or in vitro administration, preferably oral administration or injection administration.
  • Example 1 Evaluation of FGFR1-4 wild-type kinase inhibitory activity in vitro
  • the 33 P-ATP membrane filtration experiment was used to determine the inhibitory effect of test compound A on each wild-type FGFR kinase.
  • Buffer conditions 20mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (Hepes) (pH 7.5), 10mM MgCl 2 , 1mM EGTA, 0.02% (v/v%) benzze 35 (Brij35) ), 0.02 mg/mL BSA, 0.1 mM Na 3 VO 4 , 2 mM DTT, 1% (v/v%) DMSO.
  • Test procedure At room temperature, the test compound was dissolved in DMSO to prepare a 10 mM solution for later use. Add the substrate (see Table 1) to the newly prepared reaction buffer, add the specific kinase (see Table 2) to the substrate solution, and mix gently. The sonic technology (Echo550) was used to transfer the compound dissolved in DMSO to the kinase reaction mixture. The initial concentration of the test compound in the reaction mixture was 10 ⁇ M, and a total of 10 concentrations were diluted by 4 times. After 15 minutes of incubation, 33 P-ATP (illuminance 0.01 ⁇ Ci/ ⁇ L final) was added to the reaction system to start the reaction.
  • kinase activity data is expressed as the ratio of the kinase activity of the test well (containing the test compound) and the blank well (only containing DMSO).
  • the IC 50 value is obtained by curve fitting with Prism4 software (GraphPad). The experimental results are shown in Table 3.
  • Table 3 Activity of test compounds against FGFR1-4 wild-type kinase (IC 50 , nM)
  • TAS-120 was purchased from Glpbio, catalog number GC191561.
  • the compound A of the present invention can significantly inhibit the activity of FGFR1-4 wild-type kinase, and the effect is better than that of the reference compound TAS-120.
  • Test method Thaw the frozen plasma for 10-20 minutes. After the plasma is completely thawed, place it in a centrifuge and centrifuge at 3220 ⁇ g (centrifugal force) for 5 minutes to remove suspended solids and sediments. Measure the plasma pH value, and adjust the pH to the range of 7.40 ⁇ 0.10 with 1% (v/v%) phosphoric acid solution or 1M sodium hydroxide solution. Prepare 96-well incubation plates, named T0 (0min), T10 (10min), T30 (30min), T60 (60min), T120 (120min).
  • the 33 P-ATP membrane filtration experiment was used to determine the inhibitory effect of test compound A on each mutant FGFR kinase.
  • Buffer conditions 20 mM Hepes (pH 7.5), 10 mM MgCl 2 , 1 mM EGTA, 0.02% Brij35, 0.02 mg/mL BSA, 0.1 mM Na 3 VO 4 , 2 mM DTT, 1% DMSO.
  • Test procedure Add the substrate (see Table 5) to the newly prepared reaction buffer, add the specific kinase (see Table 6) to the substrate solution, and mix gently.
  • the sonic technology (Echo550) was used to transfer the compound dissolved in DMSO to the kinase reaction mixture.
  • the initial concentration of the test compound in the reaction mixture was 10 ⁇ M, and a total of 10 concentrations were diluted by 4 times.
  • 33 P-ATP (illuminance 0.01 ⁇ Ci/ ⁇ L final) was added to the reaction system to start the reaction.
  • kinase activity data is expressed as the ratio of the kinase activity of the test well (containing the test compound) and the blank well (only containing DMSO), and the IC 50 value is obtained by curve fitting with Prism4 software (GraphPad). The experimental results are shown in Table 7.
  • BGJ-398 was purchased from Glpbio, catalog number GC10055.
  • the compound A of the present invention can significantly inhibit the activity of FGFR1-4 mutant kinase, and the effect is better than that of the reference compounds TAS-120 and BGJ-398.
  • Example 4 Evaluation of the proliferation inhibitory activity of human gastric cancer cells, bladder cancer cells and liver cancer cells
  • SNU-16 human gastric cancer cells
  • FGFR2 gastric cancer cells
  • RT112/84 human bladder cancer cells with high FGFR3 expression and FGFR3-TACC3 fusion
  • Hep3B human liver cancer cells
  • the medium of SNU-16 is PRMI-1640 medium (Invitrogen, catalog number 11875093) supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin solution, Hep3B cells and RT112/84 cells.
  • the medium is an EMEM medium (ATCC, catalog number: 30-2003) supplemented with a final concentration of 10% fetal bovine serum and a 1% penicillin/streptomycin solution.
  • Test procedure Digest the SNU-16, RT112/84 and Hep3B cells that have reached 80% cell fusion with trypsin, centrifuge and resuspend the count, and use the culture medium to make 100,000, 20,000 and 70,000 cells/mL SNU-16, RT112/84 and Hep3B cell suspension were added to a 96-well cell culture plate (90 ⁇ L/well) and placed in a cell culture incubator containing 5% CO 2 at 37°C. After the cells were cultured for 24 hours, the reference compound Epirubicin and the test compound A were dissolved in DMSO into a mother liquor with a concentration of 30 mM.
  • the final concentration of the test compound is 30 ⁇ M (as IC50 test The initial concentration), 5 times decreasing dilution 9 concentrations, 9 concentrations are: 30 ⁇ M, 6 ⁇ M, 1.2 ⁇ M, 0.24 ⁇ M, 0.048 ⁇ M, 0.0096 ⁇ M, 0.0019 ⁇ M, 0.0004 ⁇ M and 0.00008 ⁇ M, mix and centrifuge, set Cultured in a cell incubator containing 5% CO 2 at 37°C for 3 days.
  • Table 8 The inhibitory effect of compound A on the proliferation of SNU-16, RT112/84 and Hep3B cells (IC 50 , nM)
  • Epirubicin was purchased from TOCRIS, article number 3260, batch number 2A7193516.
  • Compound A has a significant inhibitory effect on the proliferation of tested SNU-16, RT112/84 and Hep3B cells. It can be seen that compound A has a significant inhibitory effect on the proliferation of tumor cells with abnormal FGFR expression, and the effect is better than that of the reference compound. Spectrum anticancer drug Epirubicin.
  • Example 5 Evaluation of inhibitory activity on the proliferation of human cholangiocarcinoma cells
  • the MTT method was used to determine the inhibitory effect of test compound A on the proliferation of human cholangiocarcinoma cells HuCCT1 with high FGFR2 expression and human intrahepatic cholangiocarcinoma cells RBE.
  • the cell culture medium used was PRMI-1640 medium (Gibco, lot number: 8119264) supplemented with fetal bovine serum at a final concentration of 10%.
  • Test procedure Inoculate a certain number of cells in logarithmic growth phase in a 96-well plate (100 ⁇ L/well), and add 100 ⁇ L of culture medium containing different concentration gradients of compound A or the control drug TAS-120 to each well after 24 hours of attachment. There are 3 multiple holes for each drug concentration, and corresponding blank holes (only medium) and normal holes (drug concentration is 0). After 72 hours of drug action, add MTT working solution (5mg/mL, 20 ⁇ L per well), act at 37°C for 4 hours, shake the plate to remove the supernatant, add 150 ⁇ L of DMSO (analytical purity); Wipe the plate clean, and detect the optical density (OD) at 550nm with a microplate reader.
  • MTT working solution 5mg/mL, 20 ⁇ L per well
  • Inhibition rate (%) ( normal OD value-OD value dosing hole ) / ( normal OD value-OD value blank hole ) ⁇ 100%
  • Compound A has a good inhibitory effect on the proliferation of HuCCT1 and RBE cells tested, while TAS-120 has almost no inhibitory effect on the proliferation of HuCCT1 and RBE cells, with significant differences.
  • Example 6 In vivo pharmacodynamic evaluation of human gastric cancer SNU-16 cell subcutaneous xenograft tumor BALB/c nude mouse model
  • the BALB/c nude mouse model of human gastric cancer SNU-16 xenograft tumors amplified by FGFR2 gene was used to determine the inhibitory effect of compound A on human gastric cancer.
  • SNU-16 cells were cultured in vitro under RPMI-1640 medium containing 10% fetal bovine serum and 2mM L-glutamine, cultured in an incubator containing 5% CO 2 at 37°C. Use pancreatin-EDTA for routine digestion and passage twice a week. When the cell saturation is 80%-90% and the number reaches the requirement, the cells are collected, counted, and the test is carried out according to the following steps.
  • the reference compound TAS-120 and the test compound A are dissolved in an aqueous solution containing 0.5% (w/v) methyl cellulose (MC) and 0.5% (v/v) Tween 80 into an appropriate solution, and administered at 10 mL/kg Volumetric gavage was given to mice for 28 consecutive days.
  • the tumor diameter was measured with vernier calipers twice a week.
  • TGI tumor growth inhibition rate
  • Example 7 In vivo pharmacodynamic evaluation of human bladder cancer RT112/84 cell subcutaneous xenograft tumor BALB/c nude mouse model
  • RT112/84 cells were cultured in a monolayer in vitro, and the culture conditions were EMEM medium (Gibco, catalog number 11140076) containing 10% fetal bovine serum, 1% NEAA (non-essential amino acids), and 2mM L-glutamine (Gibco, catalog number 11140076). Cultivation in an incubator containing 5% CO 2. Use pancreatin-EDTA for routine digestion and passage twice a week. When the cell saturation is 80%-90% and the number reaches the requirement, the cells are collected, counted, and the test is carried out according to the following steps.
  • the control compound TAS-120 and the test compound A were dissolved into an appropriate solution with an aqueous solution containing 0.5% (w/v) MC and 0.5% (v/v) Tween 80, and were administered to mice by gavage at a dose volume of 10 mL/kg , Continuous administration for 20 days.
  • the tumor diameter was measured with vernier calipers twice a week.
  • TGI tumor growth inhibition rate
  • Example 8 In vivo pharmacodynamic evaluation of human liver cancer LI-03-0332 subcutaneous xenograft model
  • a subcutaneous xenograft tumor model constructed by nude mice after inoculation with LI-03-0332 human liver cancer tissue with high expression of FGFR3 was used to determine the inhibitory effect of compound A on human liver cancer.
  • Tumor tissue The LI-03-0332 model of human liver cancer was originally established from clinical samples resected by surgery. The collection and use of specimens strictly abide by national, hospital and company-related ethical laws and regulations. The passage naming rule is: the tumor sample is inoculated into nude mice as the P0 generation, and the subsequent passage is the P1 generation, and so on, the recovered sample is named FP, and the tumor tissue used in this experiment is the FP9 generation.
  • Test procedure The LI-03-0332 tumor tissue was cut into 20-30mm 3 pieces after removing the necrotic tissue. After adding base glue, the liver cancer tumor tissue was subcutaneously inoculated on the right back of each mouse, a total of 149 mice were inoculated Mice. On the 22nd day after inoculation, when the measured average tumor volume reached 135mm 3 , random stratified grouping was used according to tumor volume and animal body weight. There were 6 mice in the solvent control group and 8 mice in each of the remaining groups.
  • the control compound TAS-120 and the test compound A were dissolved into an appropriate solution with an aqueous solution containing 0.5% (w/v) MC and 0.5% (v/v) Tween 80, and were administered to mice by gavage at a dose volume of 10 mL/kg , Continuous administration for 21 days.
  • the tumor diameter was measured with vernier calipers twice a week.
  • TGI tumor growth inhibition rate
  • Table 12 Effect of compound A on tumor volume (mean ⁇ SEM, mm 3 ) of LI-03-0332 xenograft tumor model
  • the solvent control group contains 6 animals, and the other groups contain 8 animals.
  • Test procedure Take an appropriate amount of 50 times working solution of test compound A (100% DMSO dissolved) into the test hole (the final concentration of compound A is 0.1 ⁇ M), then add the mixed solution of kinase and substrate; add the selected The concentration of ATP solution starts to react; the mixture of kinase and substrate and compound do not need to be pre-incubated before ATP is added.
  • the Functional Observation Combination Test was used to evaluate the effect of single intragastric administration of test compound A on the central nervous system function of SD rats. Divide 20 male and female rats into 4 groups, each of which has 5 male and female rats. Orally administered a vehicle (containing 0.5% (weight/volume, w/v) methylcellulose and 0.2% (w/v) Tween 80). Aqueous solution) or compound A at doses of 0.5, 1.2 and 3 mg/kg. The administration volume for all animals was 5 mL/kg. In this experiment, animals were observed for mortality and weighed. FOB observations were performed before the test, 2 hours after the administration, 8 hours after the administration, and 24 hours after the administration. Observation indicators include motor function, behavior change, coordination function, sensory/motor reflex and body temperature. As a result, no changes related to the test article were seen at each observation time point. Under the test conditions, there was no effect of test compound A on the central nervous system of rats.
  • test compound A Using telemetry, the effects of oral administration of test compound A on the cardiovascular parameters of awake beagle dogs were evaluated.
  • Female and male dogs were orally administered a control formulation (vehicle: an aqueous solution containing 0.5% (w/v) methylcellulose and 0.2% (w/v) Tween 80), and the test compound at a dose of 0.3, 0.6 or 2 mg/kg A.
  • the survival rate of the animals is observed every day.
  • Cage-side observations were conducted twice a day on non-dosing days, and detailed clinical observations were performed before and after each dosing.
  • the electrocardiogram, heart rate and blood pressure of all experimental animals were continuously recorded at least 2 hours before each administration to about 24 hours after administration.
  • test compound A A single oral gavage of the test compound A was given to SD rats to evaluate its effect on respiratory function. Divide 20 female and male rats into 4 groups, each group has 5 male and female, and the control formulation (vehicle: containing 0.5% (w/v) methyl cellulose and 0.2% (w/v) Tween 80 aqueous solution) was administered orally. Or 0.5, 1.2 or 3 mg/kg of test compound A. The administration volume for all animals was 5 mL/kg. Before administration, about 15 minutes of respiratory data (tidal volume, respiratory rate, ventilation volume per minute) were collected as the baseline of respiratory parameters before grouping.
  • the collection time points on the day of administration were before the administration, 2 hours after the administration, 8 hours after the administration, and 24 hours after the administration, and collected data for approximately 15 minutes.
  • the results showed that 24 hours after a single oral administration, the test article had no effect on the rat's respiratory rate, tidal volume and minute ventilation.
  • Example 11 Acute toxicity test
  • Test method 40 rats were randomly divided into 4 groups, 10 rats in each group, half male and half male, and compound A and the control solvent were administered by oral gavage.
  • the dose of compound A was 10, 40 or 400 mg/kg, respectively.
  • the solvent is an aqueous solution containing 0.5% (w/v) MC and 0.2% (w/v) Tween 80 to detect the maximum tolerated dose of compound A.
  • the administration volume is 10 mL/kg, and the observation period is 14 days.
  • Test results Only in the 400mg/kg dose group, abnormal yellow stools and a slight increase in total bilirubin were observed. There were no deaths, clinical symptoms, gross lesions, weight changes, food intake, and clinical tests (hematology, blood coagulation, serum biochemistry, urinalysis) related to the test product in the female and male animals in the other dose groups compared with the solvent control group. difference. Therefore, the maximum tolerated dose (MTD) for a single administration under the experimental conditions is 400 mg/kg.
  • MTD maximum tolerated dose
  • Test method 8 beagle dogs were randomly divided into 4 groups, 2 dogs in each group, half male and half male, and compound A and the control solvent were given by oral gavage.
  • the dose of compound A was 15, 50 or 250 mg/ kg
  • the solvent is an aqueous solution containing 0.5% (w/v) MC and 0.2% (w/v) Tween 80.
  • the administration volume is 5mL/kg.
  • the animals were fasted overnight before dosing. The observation period is 14 days. Both males and females undergo necropsy on the 21st day.
  • Test results A single dose of 15, 50, or 250 mg/kg of compound A was administered to beagle dogs. There were no mortality, clinical symptoms, body weight, food intake, hematology, blood coagulation, serum biochemistry, urine Fluid analysis parameters, gross pathological changes, and histopathological adverse reactions. Therefore, it is believed that the maximum tolerable dose (MTD) of male and female beagle dogs under this test condition is 250 mg/kg.
  • MTD tolerable dose
  • Compound A has a high tolerable dose in toxicity test of single intragastric administration and has good safety.

Abstract

Provided is the use of an FGFR inhibitor. More specifically, provided are an FGFR inhibitor for treating FGFR-related tumors or a pharmaceutically acceptable salt thereof, a pharmaceutical composition comprising same, a method for treating FGFR-related tumors by means of using a drug comprising same, and the use thereof in the preparation of a drug for treating FGFR-related tumors. In vitro and in vivo test results show that compound A has the effect of inhibiting the activity of digestive or urinary system tumors related to abnormal FGFR expression, can be used to develop a drug for treating diseases involving digestive or urinary system tumors, and has important clinical application values.

Description

一种FGFR抑制剂的用途Use of a FGFR inhibitor
相关申请的交叉引用Cross-references to related applications
本申请要求2019年11月8日提交的中国专利申请号201911086889.9和2020年4月22日提交的中国专利申请号202010321669.6的优先权,上述两篇专利申请的全部内容特此通过引用并入本文。This application claims the priority of the Chinese patent application number 201911086889.9 filed on November 8, 2019 and the Chinese patent application number 202010321669.6 filed on April 22, 2020. The entire contents of the above two patent applications are hereby incorporated by reference.
技术领域Technical field
本发明属于医药领域。更具体地,本发明涉及一种用于治疗FGFR相关肿瘤的FGFR抑制剂或其药学上可接受的盐,包含其的药物组合物,使用含有其的药物治疗FGFR相关肿瘤的方法以及其在制备治疗FGFR相关肿瘤的药物中的用途。The invention belongs to the field of medicine. More specifically, the present invention relates to an FGFR inhibitor or a pharmaceutically acceptable salt thereof for the treatment of FGFR-related tumors, a pharmaceutical composition containing the same, a method for treating FGFR-related tumors using a drug containing the same, and the preparation thereof Use in drugs for treating FGFR-related tumors.
背景技术Background technique
成纤维细胞生长因子受体(fibroblast growth factor receptor,FGFR)是成纤维细胞生长因子(fibroblast growth factor,FGF)信号传导的受体,其家族由四个成员(FGFR1-4)组成。FGF通过FGFR在细胞增殖、细胞分化、细胞迁移和血管生成等许多生理学调节过程中发挥重要作用。诸多研究证据表明,FGF信号通路异常(高表达、基因扩增、基因突变、染色体重组等)与肿瘤细胞增殖、迁移、入侵和血管形成等许多病理过程直接相关联,并且FGFR在多种肿瘤如非小细胞肺癌、乳腺癌、胃癌、膀胱癌、尿路上皮癌、子宫内膜癌、肝癌、前列腺癌、宫颈癌、结肠癌、食管癌、骨髓瘤等中均表现出过度表达或过度激活。例如,研究发现,FGFR2融合突变在约10-20%的肝内胆管癌患者中发生;FGFR3基因的改变在膀胱癌中与病理低分级、临床低分期癌有较强的相关性,超过70%的病理低级别非浸润性尿路上皮乳头状瘤含有FGFR3突变;并且在胃癌组织和胃癌细胞中均发现FGFR1、FGFR2和FGFR3高表达。因此,FGFR成为了一类重要治疗靶点,吸引了广泛的研发兴趣。Fibroblast growth factor receptor (FGFR) is a receptor for fibroblast growth factor (FGF) signal transduction, and its family consists of four members (FGFR1-4). Through FGFR, FGF plays an important role in many physiological regulation processes such as cell proliferation, cell differentiation, cell migration and angiogenesis. A lot of research evidence shows that abnormal FGF signaling pathway (high expression, gene amplification, gene mutation, chromosome recombination, etc.) is directly related to many pathological processes such as tumor cell proliferation, migration, invasion and angiogenesis, and FGFR is in many tumors such as Non-small cell lung cancer, breast cancer, gastric cancer, bladder cancer, urothelial cancer, endometrial cancer, liver cancer, prostate cancer, cervical cancer, colon cancer, esophageal cancer, myeloma, etc. all show overexpression or overactivation. For example, studies have found that FGFR2 fusion mutations occur in about 10-20% of patients with intrahepatic cholangiocarcinoma; FGFR3 gene changes in bladder cancer have a strong correlation with low-grade pathological and clinical low-stage cancers, more than 70% The pathological low-grade non-invasive urothelial papilloma contains FGFR3 mutation; and high expression of FGFR1, FGFR2 and FGFR3 are found in gastric cancer tissues and gastric cancer cells. Therefore, FGFR has become an important therapeutic target, attracting a wide range of research and development interest.
根据作用机制,靶向FGFR膜内激酶域的抑制剂可分为ATP竞争性抑制剂、非ATP竞争性可逆抑制剂和不可逆抑制剂,其中非ATP竞 争性可逆抑制剂和不可逆抑制剂对激酶的抑制活性不受细胞内和体内高ATP浓度的影响。近年来,一些以FGFR为靶点的治疗上述肿瘤疾病的药物已进入临床试验阶段,如Balversa(厄达替尼,Erdafitinib)成为被FDA批准的全世界首款针对转移性尿路上皮癌的泛FGFR可逆性抑制剂靶向药物。另外,其他FGFR1-4抑制剂(例如,BGJ-398、Debio-1347和TAS-120等)正处于尿路上皮细胞癌、肝癌等实体瘤不同临床阶段试验中。上述药物的结构式如下:According to the mechanism of action, inhibitors that target the kinase domain of the FGFR membrane can be divided into ATP-competitive inhibitors, non-ATP-competitive reversible inhibitors, and irreversible inhibitors. Among them, non-ATP-competitive reversible inhibitors and irreversible inhibitors affect kinases. The inhibitory activity is not affected by the high ATP concentration in the cell and in the body. In recent years, some FGFR-targeted drugs for the treatment of the above-mentioned tumor diseases have entered the clinical trial stage. For example, Balversa (Erdafitinib) has become the world's first pandemic drug for metastatic urothelial cancer approved by the FDA. FGFR reversible inhibitor targeted drugs. In addition, other FGFR1-4 inhibitors (for example, BGJ-398, Debio-1347 and TAS-120, etc.) are in different clinical trials for solid tumors such as urothelial cell carcinoma and liver cancer. The structural formulas of the above drugs are as follows:
Figure PCTCN2020127211-appb-000001
Figure PCTCN2020127211-appb-000001
然而,研究发现,泛FGFR(pan-FGFR)可逆抑制剂如BGJ-398在使用4-6个月后就会难以避免地产生耐药性,导致疗效降低,而TAS-120等不可逆抑制剂对BGJ-398耐药后的患者仍然有效,展现出了不可逆抑制剂的独特优势。但是迄今为止,尚未有FGFR1-4不可逆抑制剂被批准在国内外上市。However, studies have found that pan-FGFR (pan-FGFR) reversible inhibitors such as BGJ-398 will inevitably develop drug resistance after 4-6 months of use, resulting in reduced efficacy, while irreversible inhibitors such as TAS-120 Patients who are resistant to BGJ-398 are still effective, showing the unique advantages of irreversible inhibitors. But so far, no FGFR1-4 irreversible inhibitor has been approved for marketing at home and abroad.
原发性肝癌(简称肝癌)是比较常见的一种恶性肿瘤,是全球第4大因癌症死亡的原因,对人类的生命健康造成了严重威胁。肝癌主要包括肝细胞癌(hepatocellular carcinoma,HCC)、肝内胆管细胞癌(intrahepatic cholangiocarcinoma,ICC)以及混合性肝癌(combined hepatocellular cholangiocarcinoma,c-CC-CC)。肝癌在早期没有明显的临床症状,发现时常处于中晚期,其恶性程度大、复发率高、治疗效果欠佳、预后差。临床治疗肝癌的手段主要包括手术治疗、经肝动脉化疗栓塞术(transcatheter arterial chemoembolization,TACE)及放化疗等方法。然而,手术后患者的残余肝组织仍然存在患癌风险,其5 年复发风险超过70%。索拉非尼(Sorafenib)是目前唯一一种被批准用于治疗晚期肝癌的分子靶向药物,但其对肝功能Child-Pugh分级为B级的肝癌患者的疗效仍然较差。Primary liver cancer (referred to as liver cancer) is a relatively common malignant tumor and the fourth leading cause of cancer deaths in the world, posing a serious threat to human life and health. Liver cancer mainly includes hepatocellular carcinoma (HCC), intrahepatic cholangiocarcinoma (ICC), and combined hepatocellular cholangiocarcinoma (c-CC-CC). Liver cancer has no obvious clinical symptoms in the early stage, and is often found in the middle and late stages, with high malignancy, high recurrence rate, poor treatment effect, and poor prognosis. The clinical treatment of liver cancer mainly includes surgical treatment, transcatheter arterial chemoembolization (TACE), radiotherapy and chemotherapy. However, there is still a risk of cancer in the residual liver tissue of the patient after surgery, and the 5-year risk of recurrence exceeds 70%. Sorafenib is currently the only molecularly targeted drug approved for the treatment of advanced liver cancer, but its effect on liver cancer patients with liver function Child-Pugh grade B is still poor.
尿路上皮癌(urothelial cancer,UCC)是世界范围内常见的恶性肿瘤,也是我国泌尿外科临床最为常见的恶性肿瘤之一,其中90%-95%的尿路上皮癌为膀胱癌。尿路上皮细胞癌初诊时多为非肌层浸润性,但其具有高复发率;并且伴随复发次数的增加,肿瘤恶性程度会提高并转为肌层浸润性肿瘤。具有FGFR基因改变的转移性尿路上皮癌患者预后很差,对治疗的应答率低,该类患者中存在着远未满足的显著临床需求。几十年来,尿路上皮癌的治疗标准是基于顺铂(Cisplatin)的化疗方案。二线化疗药长春氟宁(Vinflunine)或紫杉烷类药物(taxanes)的疗效稍为改善,历史客观缓解率(objective response rate,ORR)也仅约为10%,中位总生存期(overall survival,OS)为7-9个月。晚期或转移性的尿路上皮癌对于近年来批准的PD-1/PD-L1检查点抑制剂,整体响应率也仅为15%至20%,中位OS约为10个月,因而许多患者并没有获益。Urothelial cancer (UCC) is a common malignant tumor worldwide, and it is also one of the most common clinical malignancies in urology in my country. Among them, 90%-95% of urothelial cancers are bladder cancer. Urothelial cell carcinoma is mostly non-muscular invasive at the first diagnosis, but it has a high recurrence rate; and with the increase in the number of recurrences, the malignancy of the tumor will increase and turn into a muscular invasive tumor. Patients with metastatic urothelial cancer with FGFR gene alterations have a poor prognosis and a low response rate to treatment. There are significant clinical needs that are far from being met in this type of patients. For decades, the standard of treatment for urothelial cancer has been based on Cisplatin (Cisplatin) chemotherapy. The curative effect of second-line chemotherapy drugs Vinflunine or taxanes is slightly improved, the historical objective response rate (ORR) is only about 10%, and the median overall survival (overall survival, OS) is 7-9 months. For advanced or metastatic urothelial cancer, the overall response rate of PD-1/PD-L1 checkpoint inhibitors approved in recent years is only 15% to 20%, and the median OS is about 10 months. Therefore, many patients Did not benefit.
胃癌(gastric cancer)也是全球常见的恶性肿瘤之一,预后相对较差,严重威胁人类健康。由于缺乏成熟的早期筛查体系,且早期胃癌症状不典型且发现率较低,大多患者确诊时为已为进展期。当今针对胃癌的治疗措施主要包括手术治疗、全身应用化学药物、放射治疗和分子靶向药物治疗。靶向治疗是针对肿瘤特有靶点的药物治疗,但是由于胃癌异质性强等原因,有关胃癌靶向治疗的临床研究成功的少、失败的多。Gastric cancer is also one of the most common malignant tumors in the world, with a relatively poor prognosis and a serious threat to human health. Due to the lack of a mature early screening system and the atypical symptoms of early gastric cancer and the low detection rate, most patients are already in the advanced stage when they are diagnosed. Today's treatment measures for gastric cancer mainly include surgical treatment, systemic application of chemical drugs, radiotherapy and molecular targeted drug therapy. Targeted therapy is a drug treatment aimed at specific tumor targets. However, due to the strong heterogeneity of gastric cancer and other reasons, clinical studies on targeted therapy of gastric cancer have few successes and many failures.
胆管癌(cholangiocarcinoma,CCA)是一种具有不同胆管细胞分化特征的上皮细胞恶性肿瘤,过去10年间发病率增加近20%,约占消化道肿瘤的3%和肝胆恶性肿瘤的10-15%。根据解剖学位置,胆管癌可分为肝内胆管癌和肝外胆管癌(extrahepatic cholangiocarcinoma,eCCA),后者进一步分为肝门部胆管癌(perihilar cholangiocarcinoma,pCCA)和远端胆管癌(distal cholangiocarcinoma,dCCA)。根治性手术是唯一的治愈手段,但疾病早期无特征性临床表现,约2/3的患者在初诊时已失去手术机会,5年生存率约10%。即使手术切除,胆管癌术后1年复发率仍高达50%。对于不可切除的局部进展期即转移性胆道肿瘤(包括 肝内外胆管癌、胆囊癌即壶腹癌等),标准一线化学治疗方案(吉西他滨联合顺铂)仅带来11.7个月的中位生存期,预后极差。但是,FGFR抑制剂在胆管癌临床试验中已展现出前景,例如FGFR抑制剂BGJ-398在FGFR基因融合、突变、扩增的进展期胆管癌中获得了极佳的II期试验结果,客观缓解率、疾病控制率在FGFR基因融合患者中达18.8%、83.3%。Cholangiocarcinoma (CCA) is a malignant tumor of epithelial cells with different characteristics of cholangiocarcinoma. The incidence has increased by nearly 20% in the past 10 years, accounting for about 3% of gastrointestinal tumors and 10-15% of hepatobiliary malignancies. According to anatomical location, cholangiocarcinoma can be divided into intrahepatic cholangiocarcinoma and extrahepatic cholangiocarcinoma (eCCA), the latter is further divided into hilar cholangiocarcinoma (pCCA) and distal cholangiocarcinoma (distal cholangiocarcinoma) , DCCA). Radical surgery is the only cure, but there are no characteristic clinical manifestations in the early stage of the disease. About two-thirds of patients have lost the opportunity of surgery at the first diagnosis, and the 5-year survival rate is about 10%. Even if surgically removed, the recurrence rate of cholangiocarcinoma 1 year after surgery is still as high as 50%. For unresectable locally advanced metastatic biliary tumors (including intrahepatic and extrahepatic cholangiocarcinoma, gallbladder cancer, ampullary carcinoma, etc.), the standard first-line chemotherapy regimen (gemcitabine combined with cisplatin) only brings a median survival of 11.7 months , The prognosis is extremely poor. However, FGFR inhibitors have shown promise in clinical trials of cholangiocarcinoma. For example, the FGFR inhibitor BGJ-398 has obtained excellent phase II trial results in advanced cholangiocarcinoma with FGFR gene fusion, mutation, and amplification, and objective remission The rate and disease control rate in FGFR gene fusion patients reached 18.8% and 83.3%.
肝内胆管癌是指位于肝内二级及以上的胆管上皮来源的恶性肿瘤,也称肝内胆管细胞癌,属于原发性肝癌的一种(占10%~15%),根据发生部位也可归属于胆管癌的一种(约占10%)。肝内胆管癌恶性程度高,具有极强的侵袭和淋巴结转移特性,且早期诊断困难,患者预后差,5年总体生存率低于10%,手术切除后的中位生存时间为36个月。目前唯一可治愈肝内胆管癌的方法仍是早期发现、手术切除,但切除后的复发率也很高。至于局部晚期不可切除和复发、转移的患者更是缺乏能明显改善预后的有效措施。肝内胆管癌对传统的化疗、放疗及近年来的肿瘤免疫治疗皆不敏感,对于一线药物吉西他滨化疗失败的胆管癌患者尚无标准治疗方法。文献报道,接受5-氟尿嘧啶(5-FU)和伊立替康、5-FU和奥沙利铂、5-FU和顺铂、5-FU或卡培他滨及舒尼替尼等各种方案的二线化疗的患者,疾病无进展生存期(progression-free survival,PFS)和总生存期(overall survival,OS)的中位数分别为3.2和6.7个月,各方案之间的PFS或OS无显著差异。对于肝内胆管癌的分子机制研究已有较多报道。Nakamura等人对260个胆管癌患者进行了全面的基因组分析,发现40%的病例呈现出FGFR基因的改变,其中FGFR2在肝内胆管癌中呈现高表达。使用二代测序技术也发现,13%-50%的肝内胆管癌患者携带有FGFR2基因融合,其中含有FGFR2-BICC1和FGFR2-AHCYL1基因融合的患者占13.6%。这些发现为肝内胆管癌的治疗提供了新的机会。Intrahepatic cholangiocarcinoma refers to malignant tumors of bile duct epithelium located in the liver at level 2 and above. It is also called intrahepatic cholangiocarcinoma. It belongs to a type of primary liver cancer (accounting for 10% to 15%). Can be attributed to a type of cholangiocarcinoma (about 10%). Intrahepatic cholangiocarcinoma is highly malignant, has strong invasion and lymph node metastasis characteristics, and is difficult to diagnose early. The prognosis of the patient is poor. The overall 5-year survival rate is less than 10%, and the median survival time after surgical resection is 36 months. At present, the only cure for intrahepatic cholangiocarcinoma is early detection and surgical resection, but the recurrence rate after resection is also high. As for patients with locally advanced unresectable, recurrence and metastasis, there is a lack of effective measures that can significantly improve the prognosis. Intrahepatic cholangiocarcinoma is insensitive to traditional chemotherapy, radiotherapy and recent tumor immunotherapy. There is no standard treatment for cholangiocarcinoma patients who have failed chemotherapy with the first-line gemcitabine. It is reported in the literature to accept various programs such as 5-fluorouracil (5-FU) and irinotecan, 5-FU and oxaliplatin, 5-FU and cisplatin, 5-FU or capecitabine and sunitinib In patients with second-line chemotherapy, the median disease progression-free survival (PFS) and overall survival (OS) were 3.2 and 6.7 months, respectively. There was no PFS or OS between the regimens. Significant differences. There have been many reports on the molecular mechanism of intrahepatic cholangiocarcinoma. Nakamura et al. conducted a comprehensive genomic analysis of 260 cholangiocarcinoma patients and found that 40% of the cases showed FGFR gene changes, and FGFR2 was highly expressed in intrahepatic cholangiocarcinoma. Using next-generation sequencing technology, it was also found that 13%-50% of patients with intrahepatic cholangiocarcinoma carried FGFR2 gene fusions, of which 13.6% of patients had FGFR2-BICC1 and FGFR2-AHCYL1 gene fusions. These findings provide new opportunities for the treatment of intrahepatic cholangiocarcinoma.
目前,已有几种FGFR抑制剂正处于胆管癌临床研究的不同阶段,例如BGJ-398(可逆选择性抑制剂)、ARQ087(ATP竞争性抑制剂)、TAS-120(不可逆泛FGFR抑制剂)等,最早报道在胆管癌中使用的FGFR抑制剂是BGJ-398。研究报道,3例接受BGJ-398治疗的FGFR2融合阳性肝内胆管癌患者产生临床获得性FGFR抑制剂耐药,进一步研究发现FGFR2激酶结构域中的多克隆次级突变,这其中就包括3例 患者中都存在的FGFR2V564F基因突变。TAS-120为高选择性不可逆的泛FGFR抑制剂。有研究表明,TAS-120对BGJ-398耐药的肝内胆管癌患者具有临床疗效,并且可以抑制多种FGFR2的继发突变。然而,并无文献公开TAS-120的细胞水平抑制活性数据。目前尚无治疗肝内胆管癌的靶向药物获批上市。At present, several FGFR inhibitors are in different stages of clinical research on cholangiocarcinoma, such as BGJ-398 (reversible selective inhibitor), ARQ087 (ATP competitive inhibitor), TAS-120 (irreversible pan-FGFR inhibitor) Et al. The first FGFR inhibitor reported in cholangiocarcinoma was BGJ-398. The study reported that 3 patients with FGFR2 fusion-positive intrahepatic cholangiocarcinoma who received BGJ-398 treatment developed clinically acquired FGFR inhibitor resistance. Further studies found polyclonal secondary mutations in the kinase domain of FGFR2, including 3 cases The FGFR2V564F gene mutation exists in all patients. TAS-120 is a highly selective and irreversible pan-FGFR inhibitor. Studies have shown that TAS-120 has clinical effects on BGJ-398-resistant intrahepatic cholangiocarcinoma patients, and can inhibit a variety of secondary FGFR2 mutations. However, there is no literature that discloses the cell-level inhibitory activity data of TAS-120. Currently, no targeted drugs for the treatment of intrahepatic cholangiocarcinoma have been approved for marketing.
WO2019034076A1公开了FGFR抑制剂及其医药用途,其中包括化合物A(实施例2)。该专利申请公开了化合物A对FGFR野生型激酶体外抑制活性评价结果、对突变型激酶体外抑制活性评价结果及在小鼠体内的药代动力学评价结果,但未公开化合物A具体可用于哪些疾病的治疗。WO2019034076A1 discloses FGFR inhibitors and their medical uses, including compound A (Example 2). This patent application discloses the evaluation results of compound A on FGFR wild-type kinase in vitro inhibitory activity, the evaluation results on mutant kinase in vitro inhibitory activity and the pharmacokinetic evaluation results in mice, but it does not disclose which diseases Compound A can be used for. the treatment.
发明内容Summary of the invention
【本发明的目的】[Object of the present invention]
本发明的一个目的在于提供一种成纤维细胞生长因子受体(FGFR)抑制剂化合物A或其药学上可接受的盐在用于制备治疗FGFR相关肿瘤的药物中的用途。An object of the present invention is to provide a use of Fibroblast Growth Factor Receptor (FGFR) inhibitor compound A or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of FGFR-related tumors.
本发明的另一个目的在于提供一种FGFR抑制剂化合物A或其药学上可接受的盐,其用于治疗FGFR相关肿瘤。Another object of the present invention is to provide an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof, which is used for the treatment of FGFR-related tumors.
本发明的另一个目的在于提供一种治疗FGFR相关肿瘤的方法,所述方法包括给予受试者或患者含有治疗有效量的一种FGFR抑制剂化合物A或其药学上可接受的盐的药物。Another object of the present invention is to provide a method for treating FGFR-related tumors, the method comprising administering to a subject or patient a drug containing a therapeutically effective amount of an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof.
本发明的再一个目的在于提供一种用于治疗FGFR相关肿瘤的药物组合物,其包含一种FGFR抑制剂化合物A或其药学上可接受的盐并任选地包含药学上可接受的载体。Another object of the present invention is to provide a pharmaceutical composition for the treatment of FGFR-related tumors, which comprises an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof and optionally a pharmaceutically acceptable carrier.
【本发明的技术方案】[Technical Solution of the Invention]
本发明所要解决的一个技术问题是提供一种具有优良的FGFR1-4野生型和突变型激酶抑制活性的小分子化合物及其药物组合物、用途和治疗方法。A technical problem to be solved by the present invention is to provide a small molecule compound with excellent FGFR1-4 wild-type and mutant kinase inhibitory activity, and a pharmaceutical composition, use and treatment method thereof.
本发明所要解决的另一个技术问题是提供一种具有优良的FGFR1-4野生型和突变型激酶抑制活性和优良的体外/体内抗FGFR相关肿瘤(特别是消化或泌尿系统肿瘤)活性的小分子化合物及其药物组合物、用途和治疗方法。Another technical problem to be solved by the present invention is to provide a small molecule with excellent FGFR1-4 wild-type and mutant kinase inhibitory activity and excellent in vitro/in vivo anti-FGFR-related tumors (especially digestive or urinary system tumors) activity Compound and its pharmaceutical composition, use and treatment method.
本发明所要解决的再一个技术问题是提供一种具有优良的FGFR1-4野生型和突变型激酶抑制活性和优良的体外/体内抗FGFR相关肿瘤(特别是消化或泌尿系统肿瘤)活性,并且具有良好安全性的小分子化合物及其药物组合物、用途和治疗方法。Another technical problem to be solved by the present invention is to provide an excellent FGFR1-4 wild-type and mutant kinase inhibitory activity and excellent in vitro/in vivo anti-FGFR-related tumor (especially digestive or urinary system tumors) activity, and has Small molecule compounds with good safety and their pharmaceutical compositions, uses and treatment methods.
本发明所要解决的再一个技术问题是提供一种具有优良的FGFR1-4野生型和突变型激酶抑制活性和优良的体外/体内抗FGFR相关肿瘤(特别是消化或泌尿系统肿瘤)活性,并且具有优良的血浆稳定性和安全性的小分子化合物及其药物组合物、用途和治疗方法。Another technical problem to be solved by the present invention is to provide an excellent FGFR1-4 wild-type and mutant kinase inhibitory activity and excellent in vitro/in vivo anti-FGFR-related tumor (especially digestive or urinary system tumors) activity, and has Small molecule compounds with excellent plasma stability and safety, and pharmaceutical compositions, uses and treatment methods thereof.
为解决以上技术问题,本申请的发明人在现有技术WO2019034076A1的基础上进行了进一步的试验,结果发现化合物A对野生型和突变型FGFR1-4均具有良好的抑制活性,能显著抑制与FGFR1-4异常表达相关的消化或泌尿系统肿瘤细胞及异种移植肿瘤模型的增殖,且具有较高的血浆稳定性和安全性,从而提示化合物A可用于开发治疗FGFR相关的消化或泌尿系统肿瘤疾病的药物。以上这些研究的结果导致完成本发明的技术方案。In order to solve the above technical problems, the inventor of the present application conducted further experiments on the basis of the prior art WO2019034076A1, and found that compound A has good inhibitory activity on both wild-type and mutant FGFR1-4, and can significantly inhibit FGFR1 -4 Proliferation of digestive or urinary system tumor cells and xenograft tumor models related to abnormal expression, and has high plasma stability and safety, which suggests that compound A can be used to develop treatments for FGFR-related digestive or urinary system tumor diseases drug. The results of the above studies have led to the completion of the technical solution of the present invention.
具体而言,在本发明的第一方面中,提供了一种FGFR抑制剂化合物A或其药学上可接受的盐在用于制备治疗FGFR相关肿瘤的药物中的用途,其中所述化合物A具有如下结构:Specifically, in the first aspect of the present invention, there is provided the use of an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of FGFR-related tumors, wherein the compound A has The structure is as follows:
Figure PCTCN2020127211-appb-000002
Figure PCTCN2020127211-appb-000002
在一个实施方案中,所述FGFR相关肿瘤是消化或泌尿系统肿瘤中的一种或多种。In one embodiment, the FGFR-related tumor is one or more of digestive or urinary system tumors.
在一个实施方案中,所述FGFR相关肿瘤是胃癌、肝癌、尿路上皮癌、胆管癌中的任何一种或其任何组合。In one embodiment, the FGFR-related tumor is any one of gastric cancer, liver cancer, urothelial cancer, cholangiocarcinoma, or any combination thereof.
在一个实施方案中,所述FGFR相关肿瘤是胃癌。在一个优选的实施方案中,所述胃癌是FGFR2基因扩增的胃癌。In one embodiment, the FGFR-related tumor is gastric cancer. In a preferred embodiment, the gastric cancer is gastric cancer with FGFR2 gene amplification.
在一个实施方案中,所述FGFR相关肿瘤是肝癌。在一个优选的实施方案中,所述肝癌是FGFR4/FGF19高表达的肝癌。在一个优选的实施方案中,所述肝癌是FGFR3高表达的肝癌。在一个优选的实施方 案中,所述肝癌是肝细胞癌、肝内胆管细胞癌、混合性肝癌中的任何一种或其任何组合。In one embodiment, the FGFR-related tumor is liver cancer. In a preferred embodiment, the liver cancer is a liver cancer with high FGFR4/FGF19 expression. In a preferred embodiment, the liver cancer is a liver cancer with high FGFR3 expression. In a preferred embodiment, the liver cancer is any one of hepatocellular carcinoma, intrahepatic cholangiocarcinoma, mixed liver cancer, or any combination thereof.
在一个实施方案中,所述FGFR相关肿瘤是尿路上皮癌。在一个优选的实施方案中,所述尿路上皮癌是膀胱癌。在一个优选的实施方案中,所述膀胱癌是FGFR3高表达及FGFR3-TACC3融合的膀胱癌。In one embodiment, the FGFR-related tumor is urothelial carcinoma. In a preferred embodiment, the urothelial cancer is bladder cancer. In a preferred embodiment, the bladder cancer is a bladder cancer with high FGFR3 expression and FGFR3-TACC3 fusion.
在一个实施方案中,所述FGFR相关肿瘤是胆管癌。在一个优选的实施方案中,所述胆管癌是FGFR2高表达的胆管癌。在一个优选的实施方案中,所述胆管癌是肝内胆管癌、肝门部胆管癌、远端胆管癌中的任何一种或其任何组合。在一个更优选的实施方案中,所述胆管癌是肝内胆管癌。In one embodiment, the FGFR-related tumor is cholangiocarcinoma. In a preferred embodiment, the cholangiocarcinoma is a cholangiocarcinoma with high FGFR2 expression. In a preferred embodiment, the cholangiocarcinoma is any one of intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, distal cholangiocarcinoma, or any combination thereof. In a more preferred embodiment, the cholangiocarcinoma is intrahepatic cholangiocarcinoma.
在一个实施方案中,所述化合物A或其药学上可接受的盐是所述药物中的唯一的活性成分。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is the only active ingredient in the drug.
在一个实施方案中,所述化合物A或其药学上可接受的盐与一种或多种其它靶向药物或化疗药物联合使用。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is used in combination with one or more other targeted drugs or chemotherapeutics.
在一个实施方案中,所述药物被制成临床接受的制剂。在一个优选的实施方案中,所述制剂是口服制剂、注射制剂或外用制剂。In one embodiment, the drug is formulated into a clinically accepted formulation. In a preferred embodiment, the preparation is an oral preparation, an injection preparation or a topical preparation.
在一个实施方案中,所述化合物A或其药学上可接受的盐以从约0.001mg/kg至约1000mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.01mg/kg至约100mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.02mg/kg至约50mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.03mg/kg至约20mg/kg的每日给药剂量范围给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.001 mg/kg to about 1000 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.01 mg/kg to about 100 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.02 mg/kg to about 50 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.03 mg/kg to about 20 mg/kg.
在一个实施方案中,所述药物含有治疗有效量的化合物A或其药学上可接受的盐。在一个优选的实施方案中,所述治疗有效量是0.001-1000mg。在一个优选的实施方案中,所述治疗有效量是0.01-100mg。在一个优选的实施方案中,所述治疗有效量是0.1-50mg。在一个优选的实施方案中,所述治疗有效量是0.5-30mg。In one embodiment, the medicament contains a therapeutically effective amount of Compound A or a pharmaceutically acceptable salt thereof. In a preferred embodiment, the therapeutically effective amount is 0.001-1000 mg. In a preferred embodiment, the therapeutically effective amount is 0.01-100 mg. In a preferred embodiment, the therapeutically effective amount is 0.1-50 mg. In a preferred embodiment, the therapeutically effective amount is 0.5-30 mg.
在一个实施方案中,所述化合物A或其药学上可接受的盐以单剂量给予或分剂量给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a single dose or in divided doses.
在一个实施方案中,所述药物通过口服给予、注射给予、局部给 予或体外给予。在一个优选的实施方案中,所述药物通过口服给予或注射给予。In one embodiment, the drug is administered orally, by injection, topical, or in vitro. In a preferred embodiment, the drug is administered orally or by injection.
在本发明的第二方面中,提供了一种FGFR抑制剂化合物A或其药学上可接受的盐,其用于治疗FGFR相关肿瘤,其中所述化合物A具有如下结构:In the second aspect of the present invention, there is provided an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof for the treatment of FGFR-related tumors, wherein the compound A has the following structure:
Figure PCTCN2020127211-appb-000003
Figure PCTCN2020127211-appb-000003
在一个实施方案中,所述FGFR相关肿瘤是消化或泌尿系统肿瘤中的一种或多种。In one embodiment, the FGFR-related tumor is one or more of digestive or urinary system tumors.
在一个实施方案中,所述FGFR相关肿瘤是胃癌、肝癌、尿路上皮癌、胆管癌中的任何一种或其任何组合。In one embodiment, the FGFR-related tumor is any one of gastric cancer, liver cancer, urothelial cancer, cholangiocarcinoma, or any combination thereof.
在一个实施方案中,所述FGFR相关肿瘤是胃癌。在一个优选的实施方案中,所述胃癌是FGFR2基因扩增的胃癌。In one embodiment, the FGFR-related tumor is gastric cancer. In a preferred embodiment, the gastric cancer is gastric cancer with FGFR2 gene amplification.
在一个实施方案中,所述FGFR相关肿瘤是肝癌。在一个优选的实施方案中,所述肝癌是FGFR4/FGF19高表达的肝癌。在一个优选的实施方案中,所述肝癌是FGFR3高表达的肝癌。在一个优选的实施方案中,所述肝癌是肝细胞癌、肝内胆管细胞癌、混合性肝癌中的任何一种或其任何组合。In one embodiment, the FGFR-related tumor is liver cancer. In a preferred embodiment, the liver cancer is a liver cancer with high FGFR4/FGF19 expression. In a preferred embodiment, the liver cancer is a liver cancer with high FGFR3 expression. In a preferred embodiment, the liver cancer is any one of hepatocellular carcinoma, intrahepatic cholangiocarcinoma, mixed liver cancer, or any combination thereof.
在一个实施方案中,所述FGFR相关肿瘤是尿路上皮癌。在一个优选的实施方案中,所述尿路上皮癌是膀胱癌。在一个优选的实施方案中,所述膀胱癌是FGFR3高表达及FGFR3-TACC3融合的膀胱癌。In one embodiment, the FGFR-related tumor is urothelial carcinoma. In a preferred embodiment, the urothelial cancer is bladder cancer. In a preferred embodiment, the bladder cancer is a bladder cancer with high FGFR3 expression and FGFR3-TACC3 fusion.
在一个实施方案中,所述FGFR相关肿瘤是胆管癌。在一个优选的实施方案中,所述胆管癌是FGFR2高表达的胆管癌。在一个优选的实施方案中,所述胆管癌是肝内胆管癌、肝门部胆管癌、远端胆管癌中的任何一种或其任何组合。在一个更优选的实施方案中,所述胆管癌是肝内胆管癌。In one embodiment, the FGFR-related tumor is cholangiocarcinoma. In a preferred embodiment, the cholangiocarcinoma is a cholangiocarcinoma with high FGFR2 expression. In a preferred embodiment, the cholangiocarcinoma is any one of intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, distal cholangiocarcinoma, or any combination thereof. In a more preferred embodiment, the cholangiocarcinoma is intrahepatic cholangiocarcinoma.
在一个实施方案中,所述化合物A或其药学上可接受的盐作为唯一的活性成分使用。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is used as the sole active ingredient.
在一个实施方案中,所述化合物A或其药学上可接受的盐与一种 或多种其它靶向药物或化疗药物联合使用。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is used in combination with one or more other targeted drugs or chemotherapeutic drugs.
在一个实施方案中,所述化合物A或其药学上可接受的盐被制成临床接受的制剂。在一个优选的实施方案中,所述制剂是口服制剂、注射制剂或外用制剂。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is formulated into a clinically acceptable preparation. In a preferred embodiment, the preparation is an oral preparation, an injection preparation or a topical preparation.
在一个实施方案中,所述化合物A或其药学上可接受的盐以从约0.001mg/kg至约1000mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.01mg/kg至约100mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.02mg/kg至约50mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.03mg/kg至约20mg/kg的每日给药剂量范围给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.001 mg/kg to about 1000 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.01 mg/kg to about 100 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.02 mg/kg to about 50 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.03 mg/kg to about 20 mg/kg.
在一个实施方案中,所述化合物A或其药学上可接受的盐以治疗有效量配制在药物中。在一个优选的实施方案中,所述治疗有效量是0.001-1000mg。在一个优选的实施方案中,所述治疗有效量是0.01-100mg。在一个优选的实施方案中,所述治疗有效量是0.1-50mg。在一个优选的实施方案中,所述治疗有效量是0.5-30mg。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is formulated in a medicament in a therapeutically effective amount. In a preferred embodiment, the therapeutically effective amount is 0.001-1000 mg. In a preferred embodiment, the therapeutically effective amount is 0.01-100 mg. In a preferred embodiment, the therapeutically effective amount is 0.1-50 mg. In a preferred embodiment, the therapeutically effective amount is 0.5-30 mg.
在一个实施方案中,所述化合物A或其药学上可接受的盐以单剂量给予或分剂量给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a single dose or in divided doses.
在一个实施方案中,所述化合物A或其药学上可接受的盐通过口服给予、注射给予、局部给予或体外给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐通过口服给予或注射给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered orally, by injection, topical, or in vitro. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered by oral administration or injection.
在本发明的第三方面中,提供了一种治疗FGFR相关肿瘤的方法,所述方法包括给予受试者或患者含有治疗有效量的一种FGFR抑制剂化合物A或其药学上可接受的盐的药物,其中所述化合物A具有如下结构:In the third aspect of the present invention, there is provided a method of treating FGFR-related tumors, the method comprising administering to a subject or patient a therapeutically effective amount of an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof Drug, wherein the compound A has the following structure:
Figure PCTCN2020127211-appb-000004
Figure PCTCN2020127211-appb-000004
在一个实施方案中,所述FGFR相关肿瘤是消化或泌尿系统肿瘤 中的一种或多种。In one embodiment, the FGFR-related tumor is one or more of digestive or urinary system tumors.
在一个实施方案中,所述FGFR相关肿瘤是胃癌、肝癌、尿路上皮癌、胆管癌中的任何一种或其任何组合。In one embodiment, the FGFR-related tumor is any one of gastric cancer, liver cancer, urothelial cancer, cholangiocarcinoma, or any combination thereof.
在一个实施方案中,所述FGFR相关肿瘤是胃癌。在一个优选的实施方案中,所述胃癌是FGFR2基因扩增的胃癌。In one embodiment, the FGFR-related tumor is gastric cancer. In a preferred embodiment, the gastric cancer is gastric cancer with FGFR2 gene amplification.
在一个实施方案中,所述FGFR相关肿瘤是肝癌。在一个优选的实施方案中,所述肝癌是FGFR4/FGF19高表达的肝癌。在一个优选的实施方案中,所述肝癌是FGFR3高表达的肝癌。在一个优选的实施方案中,所述肝癌是肝细胞癌、肝内胆管细胞癌、混合性肝癌中的任何一种或其任何组合。In one embodiment, the FGFR-related tumor is liver cancer. In a preferred embodiment, the liver cancer is a liver cancer with high FGFR4/FGF19 expression. In a preferred embodiment, the liver cancer is a liver cancer with high FGFR3 expression. In a preferred embodiment, the liver cancer is any one of hepatocellular carcinoma, intrahepatic cholangiocarcinoma, mixed liver cancer, or any combination thereof.
在一个实施方案中,所述FGFR相关肿瘤是尿路上皮癌。在一个优选的实施方案中,所述尿路上皮癌是膀胱癌。在一个优选的实施方案中,所述膀胱癌是FGFR3高表达及FGFR3-TACC3融合的膀胱癌。In one embodiment, the FGFR-related tumor is urothelial carcinoma. In a preferred embodiment, the urothelial cancer is bladder cancer. In a preferred embodiment, the bladder cancer is a bladder cancer with high FGFR3 expression and FGFR3-TACC3 fusion.
在一个实施方案中,所述FGFR相关肿瘤是胆管癌。在一个优选的实施方案中,所述胆管癌是FGFR2高表达的胆管癌。在一个优选的实施方案中,所述胆管癌是肝内胆管癌、肝门部胆管癌、远端胆管癌中的任何一种或其任何组合。在一个更优选的实施方案中,所述胆管癌是肝内胆管癌。In one embodiment, the FGFR-related tumor is cholangiocarcinoma. In a preferred embodiment, the cholangiocarcinoma is a cholangiocarcinoma with high FGFR2 expression. In a preferred embodiment, the cholangiocarcinoma is any one of intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, distal cholangiocarcinoma, or any combination thereof. In a more preferred embodiment, the cholangiocarcinoma is intrahepatic cholangiocarcinoma.
在一个实施方案中,所述化合物A或其药学上可接受的盐作为唯一的活性成分给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered as the sole active ingredient.
在一个实施方案中,所述化合物A或其药学上可接受的盐与一种或多种其它靶向药物或化疗药物联合给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in combination with one or more other targeted drugs or chemotherapeutic drugs.
在一个实施方案中,所述药物被制成临床接受的制剂。在一个优选的实施方案中,所述制剂是口服制剂、注射制剂或外用制剂。In one embodiment, the drug is formulated into a clinically accepted formulation. In a preferred embodiment, the preparation is an oral preparation, an injection preparation or a topical preparation.
在一个实施方案中,所述化合物A或其药学上可接受的盐以从约0.001mg/kg至约1000mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.01mg/kg至约100mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.02mg/kg至约50mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.03mg/kg至约20mg/kg的每日给药剂量范围给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.001 mg/kg to about 1000 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.01 mg/kg to about 100 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.02 mg/kg to about 50 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.03 mg/kg to about 20 mg/kg.
在一个实施方案中,所述治疗有效量是0.001-1000mg。在一个优选的实施方案中,所述治疗有效量是0.01-100mg。在一个优选的实施方案中,所述治疗有效量是0.1-50mg。在一个优选的实施方案中,所述治疗有效量是0.5-30mg。In one embodiment, the therapeutically effective amount is 0.001-1000 mg. In a preferred embodiment, the therapeutically effective amount is 0.01-100 mg. In a preferred embodiment, the therapeutically effective amount is 0.1-50 mg. In a preferred embodiment, the therapeutically effective amount is 0.5-30 mg.
在一个实施方案中,所述化合物A或其药学上可接受的盐以单剂量给予或分剂量给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a single dose or in divided doses.
在一个实施方案中,所述化合物A或其药学上可接受的盐通过口服给予、注射给予、局部给予或体外给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐通过口服给予或注射给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered orally, by injection, topical, or in vitro. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered by oral administration or injection.
在本发明的第四方面中,提供了一种用于治疗FGFR相关肿瘤的药物组合物,其包含一种FGFR抑制剂化合物A或其药学上可接受的盐并任选地包含药学上可接受的载体,其中所述化合物A具有如下结构:In the fourth aspect of the present invention, there is provided a pharmaceutical composition for the treatment of FGFR-related tumors, which comprises an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof and optionally a pharmaceutically acceptable , Wherein the compound A has the following structure:
Figure PCTCN2020127211-appb-000005
Figure PCTCN2020127211-appb-000005
在一个实施方案中,所述FGFR相关肿瘤是消化或泌尿系统肿瘤中的一种或多种。In one embodiment, the FGFR-related tumor is one or more of digestive or urinary system tumors.
在一个实施方案中,所述FGFR相关肿瘤是胃癌、肝癌、尿路上皮癌、胆管癌中的任何一种或其任何组合。In one embodiment, the FGFR-related tumor is any one of gastric cancer, liver cancer, urothelial cancer, cholangiocarcinoma, or any combination thereof.
在一个实施方案中,所述FGFR相关肿瘤是胃癌。在一个优选的实施方案中,所述胃癌是FGFR2基因扩增的胃癌。In one embodiment, the FGFR-related tumor is gastric cancer. In a preferred embodiment, the gastric cancer is gastric cancer with FGFR2 gene amplification.
在一个实施方案中,所述FGFR相关肿瘤是肝癌。在一个优选的实施方案中,所述肝癌是FGFR4/FGF19高表达的肝癌。在一个优选的实施方案中,所述肝癌是FGFR3高表达的肝癌。在一个优选的实施方案中,所述肝癌是肝细胞癌、肝内胆管细胞癌、混合性肝癌中的任何一种或其任何组合。In one embodiment, the FGFR-related tumor is liver cancer. In a preferred embodiment, the liver cancer is a liver cancer with high FGFR4/FGF19 expression. In a preferred embodiment, the liver cancer is a liver cancer with high FGFR3 expression. In a preferred embodiment, the liver cancer is any one of hepatocellular carcinoma, intrahepatic cholangiocarcinoma, mixed liver cancer, or any combination thereof.
在一个实施方案中,所述FGFR相关肿瘤是尿路上皮癌。在一个优选的实施方案中,所述尿路上皮癌是膀胱癌。在一个优选的实施方案中,所述膀胱癌是FGFR3高表达及FGFR3-TACC3融合的膀胱癌。In one embodiment, the FGFR-related tumor is urothelial carcinoma. In a preferred embodiment, the urothelial cancer is bladder cancer. In a preferred embodiment, the bladder cancer is a bladder cancer with high FGFR3 expression and FGFR3-TACC3 fusion.
在一个实施方案中,所述FGFR相关肿瘤是胆管癌。在一个优选的实施方案中,所述胆管癌是FGFR2高表达的胆管癌。在一个优选的实施方案中,所述胆管癌是肝内胆管癌、肝门部胆管癌、远端胆管癌中的任何一种或其任何组合。在一个更优选的实施方案中,所述胆管癌是肝内胆管癌。In one embodiment, the FGFR-related tumor is cholangiocarcinoma. In a preferred embodiment, the cholangiocarcinoma is a cholangiocarcinoma with high FGFR2 expression. In a preferred embodiment, the cholangiocarcinoma is any one of intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, distal cholangiocarcinoma, or any combination thereof. In a more preferred embodiment, the cholangiocarcinoma is intrahepatic cholangiocarcinoma.
在一个实施方案中,所述化合物A或其药学上可接受的盐是所述药物组合物中的唯一的活性成分。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is the only active ingredient in the pharmaceutical composition.
在一个实施方案中,所述药物组合物中还包含一种或多种其它靶向药物或化疗药物作为活性成分。In one embodiment, the pharmaceutical composition also contains one or more other targeted drugs or chemotherapeutic drugs as active ingredients.
在一个实施方案中,所述药物组合物被制成临床接受的制剂。在一个优选的实施方案中,所述制剂是口服制剂、注射制剂或外用制剂。In one embodiment, the pharmaceutical composition is formulated into a clinically accepted formulation. In a preferred embodiment, the preparation is an oral preparation, an injection preparation or a topical preparation.
在一个实施方案中,所述化合物A或其药学上可接受的盐以从约0.001mg/kg至约1000mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.01mg/kg至约100mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.02mg/kg至约50mg/kg的每日给药剂量范围给予。在一个优选的实施方案中,所述化合物A或其药学上可接受的盐以从约0.03mg/kg至约20mg/kg的每日给药剂量范围给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.001 mg/kg to about 1000 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.01 mg/kg to about 100 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.02 mg/kg to about 50 mg/kg. In a preferred embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a daily dosage range from about 0.03 mg/kg to about 20 mg/kg.
在一个实施方案中,所述药物组合物含有治疗有效量的化合物A或其药学上可接受的盐。在一个优选的实施方案中,所述治疗有效量是0.001-1000mg。在一个优选的实施方案中,所述治疗有效量是0.01-100mg。在一个优选的实施方案中,所述治疗有效量是0.1-50mg。在一个优选的实施方案中,所述治疗有效量是0.5-30mg。In one embodiment, the pharmaceutical composition contains a therapeutically effective amount of Compound A or a pharmaceutically acceptable salt thereof. In a preferred embodiment, the therapeutically effective amount is 0.001-1000 mg. In a preferred embodiment, the therapeutically effective amount is 0.01-100 mg. In a preferred embodiment, the therapeutically effective amount is 0.1-50 mg. In a preferred embodiment, the therapeutically effective amount is 0.5-30 mg.
在一个实施方案中,所述化合物A或其药学上可接受的盐以单剂量给予或分剂量给予。In one embodiment, the compound A or a pharmaceutically acceptable salt thereof is administered in a single dose or in divided doses.
在一个实施方案中,所述药物组合物通过口服给予、注射给予、局部给予或体外给予。在一个优选的实施方案中,所述药物组合物通过口服给予或注射给予。In one embodiment, the pharmaceutical composition is administered by oral administration, injection administration, topical administration or in vitro administration. In a preferred embodiment, the pharmaceutical composition is administered by oral administration or injection.
以上实施方案代表了本发明的示例性实施方案,但是本发明并不限于以上实施方案。另外,本发明的以上实施方案中的各个技术特征可以相互组合,从而构成一个或多个新的技术方案,这些新的技术方 案也落在本发明的范围内,只要这样的新的技术方案是在技术上可行的即可。The above embodiments represent exemplary embodiments of the present invention, but the present invention is not limited to the above embodiments. In addition, the various technical features in the above embodiments of the present invention can be combined with each other to form one or more new technical solutions. These new technical solutions also fall within the scope of the present invention, as long as such new technical solutions are What is technically feasible is fine.
【本发明的有益效果】[Beneficial effects of the present invention]
为证明本发明的化合物A是对FGFR相关肿瘤,特别是消化或泌尿系统肿瘤有效的FGFR1-4抑制剂,本发明评价了化合物A对FGFR1-4野生型和突变型的体外激酶抑制活性及与FGFR相关的示例性消化或泌尿系统肿瘤(包括胃癌、肝癌、膀胱癌、胆管癌)模型的增殖抑制活性,并进一步针对若干肿瘤异种移植模型评价了化合物A对肿瘤生长的抑制效果。In order to prove that the compound A of the present invention is an effective FGFR1-4 inhibitor against FGFR-related tumors, especially tumors of the digestive or urinary system, the present invention evaluated the in vitro kinase inhibitory activity of compound A against FGFR1-4 wild-type and mutant types and compared with The proliferation inhibitory activity of FGFR-related exemplary digestive or urinary system tumors (including gastric cancer, liver cancer, bladder cancer, and cholangiocarcinoma) models, and the inhibitory effect of compound A on tumor growth was further evaluated for several tumor xenograft models.
体外激酶活性试验和细胞试验结果显示,化合物A对FGFR1-4野生型、突变型均具有良好的体外激酶抑制活性;对FGFR2基因扩增的人胃癌细胞(SNU-16)、FGFR3高表达及FGFR3-TACC3融合的人膀胱癌细胞(RT112/84)、FGFR4/FGF19高表达的人肝癌细胞(Hep3B)、FGFR2高表达的人胆管癌细胞HuCCT1及人肝内胆管癌细胞RBE的增殖具有良好的抑制作用;对人胃癌SNU-16细胞皮下异种移植肿瘤模型、人膀胱癌RT112/84细胞皮下异种移植肿瘤模型及人肝癌LI-03-0332皮下异种移植模型具有显著的抑制效果。In vitro kinase activity test and cell test results show that compound A has good in vitro kinase inhibitory activity on FGFR1-4 wild type and mutant type; on human gastric cancer cells (SNU-16) with FGFR2 gene amplification, FGFR3 high expression and FGFR3 -TACC3 fusion human bladder cancer cells (RT112/84), FGFR4/FGF19 high-expressing human liver cancer cells (Hep3B), FGFR2 high-expressing human cholangiocarcinoma cells HuCCT1 and human intrahepatic cholangiocarcinoma cells RBE have good inhibition of proliferation Effect; It has a significant inhibitory effect on human gastric cancer SNU-16 cell subcutaneous xenograft tumor model, human bladder cancer RT112/84 cell subcutaneous xenograft tumor model and human liver cancer LI-03-0332 subcutaneous xenograft model.
另外,为了考察化合物A的成药性能,本发明在人和小鼠血浆稳定性试验中测定了化合物A的血浆稳定性,并与结构接近的参照化合物(WO2019034076A1中实施例8(S构型))进行了对比。血浆稳定性结果显示,化合物A在小鼠和人的血浆中均具有更好的稳定性。In addition, in order to investigate the pharmaceutical properties of compound A, the present invention measured the plasma stability of compound A in the human and mouse plasma stability test, and the structure was similar to the reference compound (Example 8 in WO2019034076A1 (S configuration)) A comparison was made. The results of plasma stability show that compound A has better stability in both mouse and human plasma.
再者,已知的是,相对于可逆抑制剂,不可逆抑制剂通过与靶蛋白的共价键结合增强了与靶标的亲和性,这是不可逆抑制剂表现出高生物活性的根本原因。然而如果脱靶,不可逆抑制剂这种亲和性增强作用也会同样发生在脱靶靶点上,从而也带来增强的毒副作用[参见杨波等,小分子共价抑制剂研究进展,药学学报,2014,49(2):158-165]。出于对不可逆抑制剂可能带来更大毒副作用的担心,本发明检测了化合物A对多种重要非靶点激酶的脱靶效果,结果表明化合物A脱靶效应较低。Furthermore, it is known that, relative to reversible inhibitors, irreversible inhibitors enhance their affinity with the target through covalent bonding with the target protein, which is the fundamental reason why irreversible inhibitors exhibit high biological activity. However, if off-target, the affinity-enhancing effect of irreversible inhibitors will also occur on off-target sites, resulting in enhanced toxic and side effects [see Yang Bo et al. Research Progress on Small Molecule Covalent Inhibitors, Acta Pharmaceutical Sciences, 2014 , 49(2):158-165]. Out of concerns that irreversible inhibitors may bring greater toxic and side effects, the present invention detects the off-target effects of compound A on a variety of important non-target kinases, and the results show that compound A has low off-target effects.
再进一步,为了评价化合物A在机体内的安全性,本发明针对犬和鼠进行了安全药理学试验和急性毒性试验。结果表明,化合物A对各检测指标无显著影响,安全性良好。Furthermore, in order to evaluate the safety of compound A in vivo, the present invention conducted safety pharmacology tests and acute toxicity tests on dogs and mice. The results show that compound A has no significant effect on the detection indicators, and the safety is good.
综上所述,本申请的发明人通过试验发现,化合物A具有选择性抑制FGFR1-4的活性,对野生型和突变型FGFR1-4均具有良好的抑制活性,能显著抑制与FGFR1-4异常表达相关的消化或泌尿系统肿瘤细胞及异种移植肿瘤模型的增殖,且具有较高的血浆稳定性和安全性。由此可见,化合物A具有抑制FGFR异常表达相关的消化或泌尿系统肿瘤的活性,可用于开发治疗消化或泌尿系统肿瘤疾病的药物,具有重要的临床应用价值。In summary, the inventors of the present application have found through experiments that compound A has the activity of selectively inhibiting FGFR1-4, has good inhibitory activity on both wild-type and mutant FGFR1-4, and can significantly inhibit the abnormality of FGFR1-4. Expression related to the proliferation of digestive or urinary system tumor cells and xenograft tumor models, and has high plasma stability and safety. It can be seen that compound A has the activity of inhibiting digestive or urinary system tumors related to abnormal expression of FGFR, can be used to develop drugs for the treatment of digestive or urinary system tumor diseases, and has important clinical application value.
【发明的详细描述】[Detailed description of the invention]
除非另有说明,否则本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。Unless otherwise stated, the following terms and phrases used herein are intended to have the following meanings. A specific term or phrase should not be considered uncertain or unclear without a special definition, but should be understood in its ordinary meaning.
“化合物A”是指一种作为FGFR抑制剂的具有如下结构的化合物:"Compound A" refers to a compound with the following structure as an FGFR inhibitor:
Figure PCTCN2020127211-appb-000006
Figure PCTCN2020127211-appb-000006
,其合成方法可参见WO2019034076A1的实施例2。For its synthesis method, please refer to Example 2 of WO2019034076A1.
“药学上可接受的盐”是指由本发明的化合物A与无机酸、有机酸、无机碱或有机碱反应形成的常规的无毒盐。所述盐在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。可以通过在纯的溶液或合适的惰性溶剂中用足够量的无毒的酸或碱与化合物A接触的方式获得药学上可接受的碱加成盐或酸加成盐。"Pharmaceutically acceptable salt" refers to a conventional non-toxic salt formed by the reaction of Compound A of the present invention with an inorganic acid, organic acid, inorganic base or organic base. The salt is within the scope of reliable medical judgment and is suitable for use in contact with human and animal tissues without excessive toxicity, irritation, allergic reactions or other problems or complications, and reasonable benefits/risks Commensurate. A pharmaceutically acceptable base addition salt or acid addition salt can be obtained by contacting compound A with a sufficient amount of non-toxic acid or base in a pure solution or a suitable inert solvent.
“药学上可接受的载体”是指适用于配制化合物A或其药学上可接受的盐的药物组合物或其临床接受的制剂的载体。所述载体在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。“FGFR”是指成纤维细胞生长因子受体,它是成纤维细胞生长因子信号传导的受体,其家族由四个成员(FGFR1-4)组成。FGF通过FGFR在细胞增殖、细胞分化、细胞迁移和血管生成等许多生理学调节过程中发挥重要作用。"Pharmaceutically acceptable carrier" refers to a carrier suitable for formulating a pharmaceutical composition of Compound A or a pharmaceutically acceptable salt thereof or a clinically accepted preparation thereof. The carrier is within the scope of reliable medical judgment and is suitable for use in contact with human and animal tissues without excessive toxicity, irritation, allergic reactions or other problems or complications, and reasonable benefits/risks Commensurate. "FGFR" refers to fibroblast growth factor receptor, which is a receptor for fibroblast growth factor signaling, and its family consists of four members (FGFR1-4). Through FGFR, FGF plays an important role in many physiological regulation processes such as cell proliferation, cell differentiation, cell migration and angiogenesis.
“FGFR相关肿瘤”是指其发生或发展与FGFR1-4中的任何一种或其任何组合的表达及激活密切相关的肿瘤,其包括但不限于胃癌、膀胱癌、尿路上皮癌、肝癌、胆管癌(例如肝内胆管癌)。"FGFR-related tumors" refer to tumors whose occurrence or development is closely related to the expression and activation of any one or any combination of FGFR1-4, including but not limited to gastric cancer, bladder cancer, urothelial cancer, liver cancer, Cholangiocarcinoma (eg intrahepatic cholangiocarcinoma).
“靶向药物”是指临床用于治疗肿瘤相关疾病的靶向药物,它被赋予了靶向能力,能够使有效成分或其载体瞄准特定的目标病变部位,并在该部位蓄积或释放有效成分,形成相对较高的浓度,从而在提高疗效的同时抑制毒副作用,减少对正常组织、细胞的伤害。"Targeted drug" refers to a targeted drug that is clinically used to treat tumor-related diseases. It is endowed with a targeting ability to enable the active ingredient or its carrier to target a specific target lesion site, and accumulate or release the active ingredient at that site , To form a relatively high concentration, thereby improving the curative effect while inhibiting side effects and reducing damage to normal tissues and cells.
“化疗药物”是指能作用在肿瘤细胞生长繁殖的不同环节上,抑制或杀死肿瘤细胞的临床用于治疗肿瘤相关疾病的药物,是目前治疗肿瘤的主要手段之一。"Chemotherapeutic drugs" refer to drugs that can act on different links in the growth and reproduction of tumor cells, inhibit or kill tumor cells, and are clinically used to treat tumor-related diseases. It is currently one of the main methods of treating tumors.
“临床接受的制剂”是指在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称的制剂。临床接受的制剂包括口服制剂、注射制剂、外用制剂等。"Clinically accepted preparations" are within the scope of reliable medical judgment, suitable for use in contact with human and animal tissues, without excessive toxicity, irritation, allergic reactions or other problems or complications, and A formulation with a reasonable benefit/risk ratio commensurate. The clinically accepted preparations include oral preparations, injection preparations, and topical preparations.
根据本发明,所述化合物A或其药学上可接受的盐的给予剂量和剂量频率可以通过常规方法诸如建模、剂量递增研究或临床试验的常规方法和通过考虑诸如待治疗疾病的特性和严重程度、患者的年龄、一般情况和体重,以及所给予的具体化合物,它的药物代谢动力学特性,以及给予途径等因素进行确定。所述化合物A或其药学上可接受的盐的合适的每日给药剂量范围为从约0.001mg/kg至约1000mg/kg;优选地,从约0.01mg/kg至约100mg/kg;进一步优选地,从约0.02mg/kg至约50mg/kg;更进一步优选地,从约0.03mg/kg至约20mg/kg(其中,“kg”指代的是给药对象(即,受试者或患者)的体重)。优选地,所述化合物A或其药学上可接受的盐的每日给药剂量为0.001mg-1000mg,进一步优选地,化合物A或其药学上可接受的盐的每日给药剂量为0.01-100mg;更进一步优选地,化合物A或其药学上可接受的盐的每日给药剂量为0.1-50mg;更进一步优选地,化合物A或其药学上可接受的盐的每日给药剂量为0.5-30mg;更进一步优选地,化合物A或其药学上可接受的盐的每日给药剂量为0.1mg、0.5mg、1mg、2mg、5mg、10mg、15mg、20mg、25mg、30mg、35mg、36mg、40mg、45mg、50mg、55mg、60mg、70mg、72mg、100mg、200mg、500mg、800mg、1000mg,以单剂量或分剂量给予。According to the present invention, the dosage and frequency of administration of the compound A or its pharmaceutically acceptable salt can be performed by conventional methods such as modeling, dose escalation studies, or clinical trials, and by considering the characteristics and severity of the disease to be treated. The degree, age, general condition and weight of the patient, as well as the specific compound administered, its pharmacokinetic properties, and the route of administration are determined. A suitable daily dosage range of the compound A or a pharmaceutically acceptable salt thereof is from about 0.001 mg/kg to about 1000 mg/kg; preferably, from about 0.01 mg/kg to about 100 mg/kg; further Preferably, from about 0.02mg/kg to about 50mg/kg; still more preferably, from about 0.03mg/kg to about 20mg/kg (wherein, "kg" refers to the administration object (ie, the subject Or the weight of the patient). Preferably, the daily dose of compound A or its pharmaceutically acceptable salt is 0.001 mg-1000 mg, and more preferably, the daily dose of compound A or its pharmaceutically acceptable salt is 0.01-1000 mg. 100mg; More preferably, the daily dose of Compound A or its pharmaceutically acceptable salt is 0.1-50 mg; More preferably, the daily dose of Compound A or its pharmaceutically acceptable salt is 0.5-30mg; More preferably, the daily dosage of Compound A or its pharmaceutically acceptable salt is 0.1mg, 0.5mg, 1mg, 2mg, 5mg, 10mg, 15mg, 20mg, 25mg, 30mg, 35mg, 36mg, 40mg, 45mg, 50mg, 55mg, 60mg, 70mg, 72mg, 100mg, 200mg, 500mg, 800mg, 1000mg, given in single or divided doses.
根据本发明,所述化合物A或其药学上可接受的盐以治疗有效量包含在所述药物或药物组合物中。所述治疗有效量优选为0.001-1000mg,进一步优选为0.01-100mg,又进一步优选为0.1-50mg,更进一步优选为0.5-30mg,以单剂量给予或分剂量给予。According to the present invention, the compound A or a pharmaceutically acceptable salt thereof is included in the drug or pharmaceutical composition in a therapeutically effective amount. The therapeutically effective amount is preferably 0.001-1000 mg, more preferably 0.01-100 mg, still more preferably 0.1-50 mg, still more preferably 0.5-30 mg, administered in a single dose or in divided doses.
根据本发明,所述化合物A或其药学上可接受的盐的治疗有效量、给予剂量或给药剂量,均以化合物A计。According to the present invention, the therapeutically effective amount, administration dose or administration dose of the compound A or a pharmaceutically acceptable salt thereof is calculated as compound A.
根据本发明,患有FGFR相关肿瘤的受试者或患者可以是人和非人哺乳动物如小鼠、大鼠、豚鼠、猫、狗、牛、马、羊、猪、猴等,更优选人。According to the present invention, subjects or patients suffering from FGFR-related tumors can be human and non-human mammals such as mice, rats, guinea pigs, cats, dogs, cows, horses, sheep, pigs, monkeys, etc., more preferably humans. .
【发明的其它实施方案】[Other embodiments of the invention]
本发明还涉及以下实施方案:The present invention also relates to the following embodiments:
1.一种成纤维细胞生长因子受体(FGFR)抑制剂在用于制备治疗FGFR相关肿瘤的药物中的用途。1. The use of a fibroblast growth factor receptor (FGFR) inhibitor in the preparation of a medicine for treating FGFR-related tumors.
2.根据方案1所述的用途,其特征在于,所述肿瘤包括胃癌、肝癌、尿路上皮癌。2. The use according to scheme 1, characterized in that the tumors include gastric cancer, liver cancer, and urothelial cancer.
3.根据方案2所述的用途,其特征在于,所述肝癌为肝细胞癌、肝内胆管细胞癌、混合性肝癌。3. The use according to scheme 2, characterized in that the liver cancer is hepatocellular carcinoma, intrahepatic cholangiocarcinoma, and mixed liver cancer.
4.根据方案2所述的用途,其特征在于,所述尿路上皮癌为膀胱癌。4. The use according to scheme 2, characterized in that the urothelial cancer is bladder cancer.
5.根据方案1至4任一项所述的用途,其特征在于,所述抑制剂可与靶向药物、化疗药物中的一种或多种联合使用。5. The use according to any one of schemes 1 to 4, characterized in that the inhibitor can be used in combination with one or more of targeted drugs and chemotherapeutic drugs.
6.根据方案1至4任一项所述的用途,其特征在于,所述药物制成临床接受的制剂,所述制剂优选口服制剂、注射制剂、外用制剂。6. The use according to any one of schemes 1 to 4, characterized in that the medicine is made into a clinically accepted preparation, and the preparation is preferably an oral preparation, an injection preparation, or an external preparation.
7.根据方案1至4任一项所述的用途,其特征在于,所述抑制剂的每日给药剂量范围为从约0.001mg/kg至约1000mg/kg;优选从约0.01mg/kg至约100mg/kg;可以单剂量施用或分剂量施用。7. The use according to any one of schemes 1 to 4, characterized in that the daily dose of the inhibitor ranges from about 0.001 mg/kg to about 1000 mg/kg; preferably from about 0.01 mg/kg To about 100 mg/kg; can be administered in a single dose or in divided doses.
8.根据方案1至4任一项所述的用途,其特征在于,所述药物含有治疗有效量的抑制剂,所述治疗有效量优选0.001-1000mg;可以单剂量施用或分剂量施用。8. The use according to any one of the schemes 1 to 4, characterized in that the drug contains a therapeutically effective amount of the inhibitor, the therapeutically effective amount is preferably 0.001-1000 mg; it can be administered in a single dose or in divided doses.
进一步地,本发明还涉及以下实施方案:Further, the present invention also relates to the following embodiments:
1.一种FGFR抑制剂化合物A或其药学上可接受的盐在用于制备治疗FGFR相关肿瘤的药物中的用途。1. The use of an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of FGFR-related tumors.
2.根据方案1所述的用途,其特征在于,所述肿瘤为胆管癌。2. The use according to scheme 1, characterized in that the tumor is cholangiocarcinoma.
3.根据方案2所述的用途,其特征在于,所述胆管癌包括肝内胆管癌、肝门部胆管癌和远端胆管癌;优选为肝内胆管癌。3. The use according to scheme 2, characterized in that the cholangiocarcinoma includes intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma and distal cholangiocarcinoma; preferably intrahepatic cholangiocarcinoma.
4.根据方案1或2所述的用途,其特征在于,所述药物还含有一种或多种其它靶向药物、化疗药物。4. The use according to scheme 1 or 2, characterized in that the medicine also contains one or more other targeted medicines and chemotherapeutics.
5.根据方案1或2所述的用途,其特征在于,所述药物制成临床接受的制剂,所述制剂优选口服制剂、注射制剂、外用制剂。5. The use according to scheme 1 or 2, characterized in that the medicine is made into a clinically accepted preparation, and the preparation is preferably an oral preparation, an injection preparation, or an external preparation.
6.根据方案1或2所述的用途,其特征在于,所述药物含有治疗有效量的化合物A或其药学上可接受的盐,所述治疗有效剂量优选为0.001mg-1000mg,进一步优选为0.01-100mg,更进一步优选为0.1-50mg,更进一步优选为0.5-30mg;可以单剂量施用或分剂量施用。6. The use according to scheme 1 or 2, characterized in that the medicine contains a therapeutically effective amount of Compound A or a pharmaceutically acceptable salt thereof, and the therapeutically effective dose is preferably 0.001 mg-1000 mg, more preferably 0.01-100 mg, more preferably 0.1-50 mg, still more preferably 0.5-30 mg; it can be administered in a single dose or in divided doses.
7.一种治疗FGFR相关肿瘤疾病的方法,其特征在于,给予受试者或患者含有治疗有效剂量的化合物A或其药学上可接受的盐的药物。7. A method for treating FGFR-related tumor diseases, characterized in that a drug containing a therapeutically effective dose of Compound A or a pharmaceutically acceptable salt thereof is administered to a subject or patient.
8.根据方案7所述的方法,其特征在于,所述肿瘤为胆管癌。8. The method according to scheme 7, wherein the tumor is cholangiocarcinoma.
9.根据方案8所述的方法,其特征在于,所述胆管癌包括肝内胆管癌、肝门部胆管癌和远端胆管癌;优选为肝内胆管癌。9. The method according to claim 8, wherein the cholangiocarcinoma includes intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, and distal cholangiocarcinoma; preferably, it is intrahepatic cholangiocarcinoma.
10.根据方案7-9任一项所述的方法,其特征在于,所述给予可以是口服给予、注射给予、局部给予或体外给予,优选为口服给予或注射给予。10. The method according to any one of the schemes 7-9, characterized in that the administration may be oral administration, injection administration, topical administration or in vitro administration, preferably oral administration or injection administration.
具体实施方式Detailed ways
为了进一步理解本发明,下面结合实施例对本发明的具体实施方案进行详细地描述。但是应当理解,这些描述的目的只是为了进一步说明本发明的特征和优点,而不构成对本发明的方面的任何限制。In order to further understand the present invention, specific embodiments of the present invention will be described in detail below in conjunction with examples. However, it should be understood that the purpose of these descriptions is only to further illustrate the features and advantages of the present invention, and does not constitute any limitation to the aspects of the present invention.
实施例1:FGFR1-4野生型激酶体外抑制活性评价Example 1: Evaluation of FGFR1-4 wild-type kinase inhibitory activity in vitro
采用 33P-ATP膜过滤法实验,测定受试化合物A对各野生型FGFR激酶的抑制作用。 The 33 P-ATP membrane filtration experiment was used to determine the inhibitory effect of test compound A on each wild-type FGFR kinase.
缓冲液条件:20mM 4-(2-羟乙基)-1-哌嗪乙磺酸(Hepes)(pH 7.5),10mM MgCl 2,1mM EGTA,0.02%(v/v%)苄泽35(Brij35),0.02mg/mL BSA,0.1mM Na 3VO 4,2mM DTT,1%(v/v%)DMSO。 Buffer conditions: 20mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (Hepes) (pH 7.5), 10mM MgCl 2 , 1mM EGTA, 0.02% (v/v%) benzze 35 (Brij35) ), 0.02 mg/mL BSA, 0.1 mM Na 3 VO 4 , 2 mM DTT, 1% (v/v%) DMSO.
试验步骤:室温下,将受试化合物溶解在DMSO中配置成10mM溶液待用。在新制备的反应缓冲液中加入底物(见表1),将特定的激 酶(见表2)加入底物溶液中,轻轻混合。利用声波技术(Echo550)将溶于DMSO中的化合物转移到激酶反应混合物中,反应混合物中受试化合物的起始浓度为10μM,4倍递减稀释共10个浓度。孵化15分钟后,将 33P-ATP(照度0.01μCi/μL final)加入反应体系中,启动反应。室温孵育激酶反应120分钟;将反应转移在P81离子交换纸(Whatman#3698-915)上。用0.75%(v/v%)的磷酸广泛清洗过滤膜,测量滤纸上残留的放射性磷酸化底物。激酶活性数据用测试孔(含有受试化合物)和空白孔(仅含有DMSO)的激酶活性的比值表示,通过Prism4软件(GraphPad)进行曲线拟合得到IC 50值,实验结果如表3所示。 Test procedure: At room temperature, the test compound was dissolved in DMSO to prepare a 10 mM solution for later use. Add the substrate (see Table 1) to the newly prepared reaction buffer, add the specific kinase (see Table 2) to the substrate solution, and mix gently. The sonic technology (Echo550) was used to transfer the compound dissolved in DMSO to the kinase reaction mixture. The initial concentration of the test compound in the reaction mixture was 10 μM, and a total of 10 concentrations were diluted by 4 times. After 15 minutes of incubation, 33 P-ATP (illuminance 0.01μCi/μL final) was added to the reaction system to start the reaction. Incubate the kinase reaction at room temperature for 120 minutes; transfer the reaction to P81 ion exchange paper (Whatman#3698-915). The filter membrane was extensively washed with 0.75% (v/v%) phosphoric acid, and the radioactive phosphorylated substrate remaining on the filter paper was measured. The kinase activity data is expressed as the ratio of the kinase activity of the test well (containing the test compound) and the blank well (only containing DMSO). The IC 50 value is obtained by curve fitting with Prism4 software (GraphPad). The experimental results are shown in Table 3.
表1:体外测试中底物的相关信息Table 1: Information about the substrate in the in vitro test
Figure PCTCN2020127211-appb-000007
Figure PCTCN2020127211-appb-000007
表2:体外测试中激酶和ATP的相关信息Table 2: Related information of kinases and ATP in in vitro tests
Figure PCTCN2020127211-appb-000008
Figure PCTCN2020127211-appb-000008
表3:受试化合物对FGFR1-4野生型激酶的活性(IC 50,nM) Table 3: Activity of test compounds against FGFR1-4 wild-type kinase (IC 50 , nM)
化合物Compound FGFR1FGFR1 FGFR2FGFR2 FGFR3FGFR3 FGFR4FGFR4
TAS-120TAS-120 0.7840.784 0.3970.397 1.3401.340 4.6504.650
化合物ACompound A 0.1550.155 0.0850.085 0.7900.790 0.2920.292
注:TAS-120购买自Glpbio,目录号GC191561。Note: TAS-120 was purchased from Glpbio, catalog number GC191561.
结论:本发明化合物A能显著抑制FGFR1-4野生型激酶的活性,且效果优于参照化合物TAS-120。Conclusion: The compound A of the present invention can significantly inhibit the activity of FGFR1-4 wild-type kinase, and the effect is better than that of the reference compound TAS-120.
实施例2:人和小鼠血浆稳定性试验Example 2: Human and mouse plasma stability test
试验方法:将冻存的血浆解冻10~20分钟,待血浆完全解冻后,将其置于离心机中以3220×g(离心力)离心5分钟,去除其中存在的悬浮物和沉淀物。测定血浆pH值,并用1%(v/v%)磷酸溶液或1M氢氧化钠溶液调节pH至7.40±0.10范围。准备96孔孵育板,分别命名为T0(0min)、T10(10min)、T30(30min)、T60(60min)、T120(120min)。向对应的孵育板中加入198μL的小鼠或人的空白血浆,然后将2μL受试化合物的工作液(DMSO溶液)加入到相应的孵育板中(每个样品准备两个平行孔)。将所有的样品在37℃水浴锅中进行孵育。受试化合物的最终孵育浓度为2μM,最终有机相含量为1.0%(v/v%)。每一个孵育时间点结束时,取出相应的孵育板,向每个对应的样品孔中加入400μL乙腈沉淀蛋白。将所有样品板封膜并摇匀后,以3220×g离心20分钟。取50μL上清液并加入100μL超纯水稀释,混匀后用LC/MS/MS的方法分析。Test method: Thaw the frozen plasma for 10-20 minutes. After the plasma is completely thawed, place it in a centrifuge and centrifuge at 3220×g (centrifugal force) for 5 minutes to remove suspended solids and sediments. Measure the plasma pH value, and adjust the pH to the range of 7.40±0.10 with 1% (v/v%) phosphoric acid solution or 1M sodium hydroxide solution. Prepare 96-well incubation plates, named T0 (0min), T10 (10min), T30 (30min), T60 (60min), T120 (120min). Add 198 μL of mouse or human blank plasma to the corresponding incubation plate, and then add 2 μL of the working solution of the test compound (DMSO solution) to the corresponding incubation plate (two parallel wells are prepared for each sample). Incubate all samples in a 37°C water bath. The final incubation concentration of the test compound was 2 μM, and the final organic phase content was 1.0% (v/v%). At the end of each incubation time point, take out the corresponding incubation plate, and add 400 μL of acetonitrile to each corresponding sample well to precipitate the protein. After sealing and shaking all sample plates, centrifuge at 3220×g for 20 minutes. Take 50μL of supernatant and add 100μL of ultrapure water to dilute, mix well and analyze by LC/MS/MS method.
实验结果如表4所示。The experimental results are shown in Table 4.
表4:血浆稳定性试验结果Table 4: Results of plasma stability test
Figure PCTCN2020127211-appb-000009
Figure PCTCN2020127211-appb-000009
结论:与表中的参照化合物(WO2019034076A1中的实施例8(S构型))相比,化合物A在小鼠和人的血浆中均具有更好的稳定性。Conclusion: Compared with the reference compound in the table (Example 8 (S configuration) in WO2019034076A1), compound A has better stability in both mouse and human plasma.
实施例3:突变型激酶体外抑制活性评价Example 3: Evaluation of in vitro inhibitory activity of mutant kinases
采用 33P-ATP膜过滤法实验,测定受试化合物A对各突变型FGFR激酶的抑制作用。 The 33 P-ATP membrane filtration experiment was used to determine the inhibitory effect of test compound A on each mutant FGFR kinase.
缓冲液条件:20mM Hepes(pH 7.5),10mM MgCl 2,1mM EGTA,0.02%Brij35,0.02mg/mL BSA,0.1mM Na 3VO 4,2mM DTT,1%DMSO。 Buffer conditions: 20 mM Hepes (pH 7.5), 10 mM MgCl 2 , 1 mM EGTA, 0.02% Brij35, 0.02 mg/mL BSA, 0.1 mM Na 3 VO 4 , 2 mM DTT, 1% DMSO.
试验步骤:在新制备的反应缓冲液中加入底物(见表5),将特定的激酶(见表6)加入底物溶液中,轻轻混合。利用声波技术(Echo550)将溶于DMSO中的化合物转移到激酶反应混合物中,反应混合物中受试化合物的起始浓度为10μM,4倍递减稀释共10个浓度。孵化15分 钟后,将 33P-ATP(照度0.01μCi/μL final)加入反应体系中,启动反应。室温孵育激酶反应120分钟,将反应转移在P81离子交换纸(Whatman#3698-915)上,用0.75%的磷酸广泛清洗过滤膜;测量滤纸上残留的放射性磷酸化底物。激酶活性数据用测试孔(含有受试化合物)和空白孔(仅含有DMSO)的激酶活性的比值表示,通过Prism4软件(GraphPad)进行曲线拟合得到IC 50值,实验结果如表7所示。 Test procedure: Add the substrate (see Table 5) to the newly prepared reaction buffer, add the specific kinase (see Table 6) to the substrate solution, and mix gently. The sonic technology (Echo550) was used to transfer the compound dissolved in DMSO to the kinase reaction mixture. The initial concentration of the test compound in the reaction mixture was 10 μM, and a total of 10 concentrations were diluted by 4 times. After 15 minutes of incubation, 33 P-ATP (illuminance 0.01μCi/μL final) was added to the reaction system to start the reaction. Incubate the kinase reaction at room temperature for 120 minutes, transfer the reaction to P81 ion exchange paper (Whatman#3698-915), and wash the filter membrane extensively with 0.75% phosphoric acid; measure the residual radioactive phosphorylated substrate on the filter paper. The kinase activity data is expressed as the ratio of the kinase activity of the test well (containing the test compound) and the blank well (only containing DMSO), and the IC 50 value is obtained by curve fitting with Prism4 software (GraphPad). The experimental results are shown in Table 7.
表5:底物的相关信息Table 5: Information about the substrate
Figure PCTCN2020127211-appb-000010
Figure PCTCN2020127211-appb-000010
表6:激酶和ATP的相关信息Table 6: Information about kinases and ATP
Figure PCTCN2020127211-appb-000011
Figure PCTCN2020127211-appb-000011
表7:受试化合物对FGFR突变型酶的活性(IC 50,nM) Table 7: Activity of test compounds on FGFR mutant enzyme (IC 50 , nM)
激酶Kinase TAS-120TAS-120 BGJ-398BGJ-398 化合物ACompound A
FGFR1(V561M)FGFR1(V561M) 605605 13101310 26.826.8
FGFR2(E565G)FGFR2(E565G) 1.371.37 7.247.24 0.120.12
FGFR2(N549H)FGFR2(N549H) 2.362.36 29.829.8 0.230.23
FGFR2(V564F)FGFR2(V564F) 255255 65206520 42.142.1
FGFR3(K650M)FGFR3(K650M) 1.991.99 2727 0.170.17
FGFR3(V555M)FGFR3(V555M) 22.822.8 888888 24.724.7
FGFR4(N535K)FGFR4(N535K) 80208020 1020010200 17.517.5
FGFR4(V550M)FGFR4(V550M) 93.993.9 39603960 4.244.24
注:BGJ-398购买自Glpbio,目录号GC10055。Note: BGJ-398 was purchased from Glpbio, catalog number GC10055.
结论:本发明化合物A能显著抑制FGFR1-4突变型激酶的活性,且效果优于参照化合物TAS-120和BGJ-398。Conclusion: The compound A of the present invention can significantly inhibit the activity of FGFR1-4 mutant kinase, and the effect is better than that of the reference compounds TAS-120 and BGJ-398.
实施例4:对人胃癌细胞、膀胱癌细胞和肝癌细胞增殖抑制活性评价Example 4: Evaluation of the proliferation inhibitory activity of human gastric cancer cells, bladder cancer cells and liver cancer cells
采用CellTiter-Glo TM活细胞检测试剂盒,测定受试化合物A对FGFR2基因扩增的人胃癌细胞(SNU-16)、FGFR3高表达及FGFR3-TACC3融合的人膀胱癌细胞(RT112/84)和FGFR4/FGF19高表达的人肝癌细胞(Hep3B)增殖的抑制作用。其中,SNU-16的培养基为添加终浓度为10%的胎牛血清、1%的青霉素/链霉素溶液的PRMI-1640培养基(Invitrogen,货号11875093),Hep3B细胞和RT112/84细胞的培养基为添加终浓度为10%胎牛血清、1%的青霉素/链霉素溶液的EMEM培养基(ATCC,货号:30-2003)。 CellTiter-Glo TM live cell detection kit was used to determine the effects of test compound A on human gastric cancer cells (SNU-16) with FGFR2 gene amplification, human bladder cancer cells with high FGFR3 expression and FGFR3-TACC3 fusion (RT112/84) and Inhibition of proliferation of human liver cancer cells (Hep3B) with high FGFR4/FGF19 expression. Among them, the medium of SNU-16 is PRMI-1640 medium (Invitrogen, catalog number 11875093) supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin solution, Hep3B cells and RT112/84 cells. The medium is an EMEM medium (ATCC, catalog number: 30-2003) supplemented with a final concentration of 10% fetal bovine serum and a 1% penicillin/streptomycin solution.
试验步骤:用胰酶消化已达到80%细胞融合的SNU-16、RT112/84和Hep3B细胞,离心重悬计数,用培养基分别制成100000、20000和70000个细胞/mL的SNU-16、RT112/84、Hep3B细胞悬液,加入96孔细胞培养板(90μL/孔),置于含5%CO 2的细胞培养箱中于37℃培养。细胞培养24小时后,参考化合物表阿霉素(Epirubicin)及受试化合物A用DMSO溶解成浓度为30mM的母液。用SNU-16、RT112/84和Hep3B的培养基将稀释好的化合物母液进行进一步稀释,并将稀释好的混合液分别转移至相应的细胞板中,受试化合物终浓度为30μM (作为IC50测试的起始浓度),5倍递减稀释9个浓度,9个浓度分别为:30μM、6μM、1.2μM、0.24μM、0.048μM、0.0096μM、0.0019μM、0.0004μM和0.00008μM,混匀离心,置于含5%CO 2的细胞培养箱中于37℃培养3天。取出96孔细胞培养板,加入CellTiterGlo(CTG,化学发光细胞活性检测试剂盒)试剂(100μL/孔),混匀离心,于室温孵育10分钟。使用Envision多标记分析仪读数。根据原始数据计算抑制率,抑制率计算公式为:抑制率%=(DMSO对照-待测孔)/(DMSO对照-培养基对照)×100。用XLFit统计软件进行数据分析,计算IC 50。实验结果如表8所示。 Test procedure: Digest the SNU-16, RT112/84 and Hep3B cells that have reached 80% cell fusion with trypsin, centrifuge and resuspend the count, and use the culture medium to make 100,000, 20,000 and 70,000 cells/mL SNU-16, RT112/84 and Hep3B cell suspension were added to a 96-well cell culture plate (90 μL/well) and placed in a cell culture incubator containing 5% CO 2 at 37°C. After the cells were cultured for 24 hours, the reference compound Epirubicin and the test compound A were dissolved in DMSO into a mother liquor with a concentration of 30 mM. Use SNU-16, RT112/84 and Hep3B medium to further dilute the diluted compound stock solution, and transfer the diluted mixture to the corresponding cell plate. The final concentration of the test compound is 30μM (as IC50 test The initial concentration), 5 times decreasing dilution 9 concentrations, 9 concentrations are: 30μM, 6μM, 1.2μM, 0.24μM, 0.048μM, 0.0096μM, 0.0019μM, 0.0004μM and 0.00008μM, mix and centrifuge, set Cultured in a cell incubator containing 5% CO 2 at 37°C for 3 days. Take out the 96-well cell culture plate, add CellTiterGlo (CTG, Chemiluminescence Cell Activity Detection Kit) reagent (100 μL/well), mix and centrifuge, and incubate at room temperature for 10 minutes. Use Envision multi-marker analyzer to read. Calculate the inhibition rate based on the original data. The calculation formula of the inhibition rate is: Inhibition rate %=(DMSO control-well to be tested)/(DMSO control-medium control)×100. Data analysis was performed using statistical software XLFit calculated IC 50. The experimental results are shown in Table 8.
表8:化合物A对SNU-16、RT112/84和Hep3B细胞增殖的抑制作用(IC 50,nM) Table 8: The inhibitory effect of compound A on the proliferation of SNU-16, RT112/84 and Hep3B cells (IC 50 , nM)
化合物Compound SNU-16SNU-16 RT112/84RT112/84 Hep3BHep3B
化合物ACompound A 5.05.0 2.32.3 5.15.1
EpirubicinEpirubicin 274.6274.6 75.975.9 451.9451.9
注:Epirubicin购买自TOCRIS,货号3260,批号2A7193516。Note: Epirubicin was purchased from TOCRIS, article number 3260, batch number 2A7193516.
结论:化合物A对受试的SNU-16、RT112/84和Hep3B细胞增殖均有显著的抑制作用,可见化合物A对FGFR异常表达的肿瘤细胞的增殖有显著抑制作用,且效果优于参照化合物广谱抗癌药物Epirubicin。Conclusion: Compound A has a significant inhibitory effect on the proliferation of tested SNU-16, RT112/84 and Hep3B cells. It can be seen that compound A has a significant inhibitory effect on the proliferation of tumor cells with abnormal FGFR expression, and the effect is better than that of the reference compound. Spectrum anticancer drug Epirubicin.
实施例5:对人胆管癌细胞增殖抑制活性评价Example 5: Evaluation of inhibitory activity on the proliferation of human cholangiocarcinoma cells
采用MTT法,测定受试化合物A对FGFR2高表达的人胆管癌细胞HuCCT1和人肝内胆管癌细胞RBE增殖的抑制作用。使用的细胞培养基为添加终浓度为10%的胎牛血清的PRMI-1640培养基(Gibco,批号:8119264)。The MTT method was used to determine the inhibitory effect of test compound A on the proliferation of human cholangiocarcinoma cells HuCCT1 with high FGFR2 expression and human intrahepatic cholangiocarcinoma cells RBE. The cell culture medium used was PRMI-1640 medium (Gibco, lot number: 8119264) supplemented with fetal bovine serum at a final concentration of 10%.
试验步骤:将处于对数生长期的细胞以一定数量接种于96孔板(100μL/孔),贴壁24h后每孔加入100μL含不同浓度梯度的化合物A或对照药TAS-120的培养液,每个药物浓度设3个复孔,并设相应的空白孔(只有培养基)及正常孔(药物浓度为0)。药物作用72小时后,加入MTT工作液(5mg/mL,每孔20μL),于37℃作用4小时,甩板去除上清液,加入DMSO(分析纯)150μL;微孔振荡器震荡混匀,将板擦拭干净,用酶标仪于550nm处检测光密度值(OD)。Test procedure: Inoculate a certain number of cells in logarithmic growth phase in a 96-well plate (100μL/well), and add 100μL of culture medium containing different concentration gradients of compound A or the control drug TAS-120 to each well after 24 hours of attachment. There are 3 multiple holes for each drug concentration, and corresponding blank holes (only medium) and normal holes (drug concentration is 0). After 72 hours of drug action, add MTT working solution (5mg/mL, 20μL per well), act at 37°C for 4 hours, shake the plate to remove the supernatant, add 150μL of DMSO (analytical purity); Wipe the plate clean, and detect the optical density (OD) at 550nm with a microplate reader.
采用下列公式计算细胞生长的抑制率:Use the following formula to calculate the inhibition rate of cell growth:
抑制率(%)=(OD值 正常孔-OD值 给药孔)/(OD值 正常孔-OD值 空白孔)×100% Inhibition rate (%) = ( normal OD value-OD value dosing hole ) / ( normal OD value-OD value blank hole ) × 100%
根据各浓度抑制率,用SPSS19.0计算药物半数抑制浓度IC 50。实验结果如表9所示。 The inhibition rate at each concentration was calculated by SPSS19.0 medicament half maximal inhibitory concentration IC 50. The experimental results are shown in Table 9.
表9:化合物A对HuCCT1、RBE细胞增殖的抑制作用(IC 50,μM) Table 9: Inhibitory effect of compound A on the proliferation of HuCCT1 and RBE cells (IC 50 , μM)
化合物Compound HuCCT1HuCCT1 RBERBE
化合物ACompound A 1.451±0.4641.451±0.464 1.068±0.1301.068±0.130
TAS-120TAS-120 >20>20 >200>200
结论:化合物A对受试的HuCCT1和RBE细胞增殖均有较好抑制作用,而TAS-120对HuCCT1和RBE细胞增殖几乎无抑制作用,具有显著性差异。Conclusion: Compound A has a good inhibitory effect on the proliferation of HuCCT1 and RBE cells tested, while TAS-120 has almost no inhibitory effect on the proliferation of HuCCT1 and RBE cells, with significant differences.
实施例6:人胃癌SNU-16细胞皮下异种移植肿瘤BALB/c裸小鼠模型的体内药效学评价Example 6: In vivo pharmacodynamic evaluation of human gastric cancer SNU-16 cell subcutaneous xenograft tumor BALB/c nude mouse model
采用FGFR2基因扩增的人胃癌SNU-16异种移植肿瘤BALB/c裸小鼠模型,测定了化合物A对人胃癌的抑制效果。The BALB/c nude mouse model of human gastric cancer SNU-16 xenograft tumors amplified by FGFR2 gene was used to determine the inhibitory effect of compound A on human gastric cancer.
细胞培养:将SNU-16细胞于体外培养,培养条件为含10%胎牛血清、2mM左旋谷酰胺的RPMI-1640培养基,在于37℃含5%CO 2的孵箱中培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%,数量达到要求时,收取细胞,计数,按如下步骤进行试验。 Cell culture: SNU-16 cells were cultured in vitro under RPMI-1640 medium containing 10% fetal bovine serum and 2mM L-glutamine, cultured in an incubator containing 5% CO 2 at 37°C. Use pancreatin-EDTA for routine digestion and passage twice a week. When the cell saturation is 80%-90% and the number reaches the requirement, the cells are collected, counted, and the test is carried out according to the following steps.
试验步骤:将0.2mL含5×10 6个SNU-16细胞及基底胶的细胞悬液(PBS∶基底胶(Matrigel)=1∶1(v/v))皮下接种于每只小鼠的右后背。接种后第13天,当肿瘤平均体积达到144mm 3时,以分层随机化法分组,每组6只小鼠。参照化合物TAS-120及受试化合物A用含0.5%(w/v)甲基纤维素(MC)和0.5%(v/v)吐温80的水溶液溶解成适当溶液,以10mL/kg给药容积灌胃给予小鼠,连续给药28天。每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a×b 2,a和b分别表示肿瘤的长径和短径。 Test procedure: 0.2 mL of a cell suspension containing 5×10 6 SNU-16 cells and base glue (PBS: Matrigel = 1:1 (v/v)) was subcutaneously inoculated on the right side of each mouse Back. On the 13th day after inoculation, when the average tumor volume reached 144 mm 3 , they were divided into groups by stratified randomization, with 6 mice in each group. The reference compound TAS-120 and the test compound A are dissolved in an aqueous solution containing 0.5% (w/v) methyl cellulose (MC) and 0.5% (v/v) Tween 80 into an appropriate solution, and administered at 10 mL/kg Volumetric gavage was given to mice for 28 consecutive days. The tumor diameter was measured with vernier calipers twice a week. The calculation formula of tumor volume is: V=0.5a×b 2 , a and b represent the long diameter and short diameter of the tumor, respectively.
化合物的抑瘤疗效用肿瘤生长抑制率(简称抑瘤率)TGI(%)表示。TGI(%)的计算:TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积))/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]×100%。The anti-tumor efficacy of the compound is expressed by the tumor growth inhibition rate (abbreviated as tumor inhibition rate) TGI (%). Calculation of TGI(%): TGI(%)=[(1-(Average tumor volume at the end of a certain treatment group-average tumor volume at the beginning of the treatment group))/(Average tumor volume at the end of treatment in the solvent control group Volume-average tumor volume at the start of treatment in the solvent control group)]×100%.
实验结果如表10所示。The experimental results are shown in Table 10.
表10:化合物A对SNU-16异种移植瘤模型瘤体积(平均值±SEM,mm 3,n=6)的影响 Table 10: Effect of Compound A on SNU-16 Xenograft Tumor Model Tumor Volume (Mean±SEM, mm 3 , n=6)
Figure PCTCN2020127211-appb-000012
Figure PCTCN2020127211-appb-000012
注:两组间比较用T-test进行分析,三组或多组间比较用one-way ANOVA进行分析,使用Prism进行数据分析。Note: The comparison between two groups is analyzed by T-test, the comparison between three or more groups is analyzed by one-way ANOVA, and Prism is used for data analysis.
结论:化合物A在10、20和40mg/kg剂量下每天一次(QD)给予,均具有显著的抑瘤效果,且抑瘤效果具有剂量依赖性趋势。给药28天,抑瘤率(TGI)分别为63%、86%和98%,与溶剂对照组相比p值均小于0.05。在同等剂量下,化合物A的抑瘤效果优于TAS-120,并且在40mg/kg的剂量下,二者的疗效差异显著,具有统计学意义(p<0.05)。Conclusion: Compound A administered once a day (QD) at doses of 10, 20 and 40 mg/kg, all have significant anti-tumor effect, and the anti-tumor effect has a dose-dependent trend. After 28 days of administration, the tumor inhibition rate (TGI) was 63%, 86%, and 98%, respectively, and the p values were all less than 0.05 compared with the solvent control group. At the same dose, the anti-tumor effect of compound A is better than that of TAS-120, and at a dose of 40 mg/kg, the therapeutic effect of the two is significantly different, which is statistically significant (p<0.05).
实施例7:人膀胱癌RT112/84细胞皮下异种移植肿瘤BALB/c裸小鼠模型的体内药效学评价Example 7: In vivo pharmacodynamic evaluation of human bladder cancer RT112/84 cell subcutaneous xenograft tumor BALB/c nude mouse model
采用FGFR3高表达及FGFR3-TACC3融合的人膀胱癌细胞RT112/84异种移植肿瘤BALB/c裸小鼠模型,测定了化合物A对人膀胱癌的抑制效果。Using the human bladder cancer cell RT112/84 xenograft BALB/c nude mouse model with high FGFR3 expression and FGFR3-TACC3 fusion, the inhibitory effect of compound A on human bladder cancer was determined.
细胞培养:将RT112/84细胞于体外单层培养,培养条件为含10%胎牛血清、1%NEAA(非必须氨基酸)、2mM左旋谷酰胺的EMEM培养基(Gibco,货号11140076),在于37℃含5%CO 2的孵箱中培养。一周两次用胰酶-EDTA进行常规消化处理传代。当细胞饱和度为80%-90%,数量达到要求时,收取细胞,计数,按如下步骤进行试验。 Cell culture: RT112/84 cells were cultured in a monolayer in vitro, and the culture conditions were EMEM medium (Gibco, catalog number 11140076) containing 10% fetal bovine serum, 1% NEAA (non-essential amino acids), and 2mM L-glutamine (Gibco, catalog number 11140076). Cultivation in an incubator containing 5% CO 2. Use pancreatin-EDTA for routine digestion and passage twice a week. When the cell saturation is 80%-90% and the number reaches the requirement, the cells are collected, counted, and the test is carried out according to the following steps.
试验步骤:将0.2mL含10×10 6个RT112/84细胞及基底胶的细胞悬液(PBS∶Matrigel=1∶1(v/v))皮下接种于每只小鼠的右后背。当肿瘤平均体积达到188mm 3时,以分层随机化法分组,每组8只小鼠。对照化合物TAS-120及受试化合物A用含0.5%(w/v)MC和0.5%(v/v)吐温80的水溶液溶解成适当溶液,以10mL/kg给药容积灌胃给予小鼠,连续给药20天。每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a×b 2,a和b分别表示肿瘤的长径和短径。 Test procedure: 0.2 mL of a cell suspension (PBS: Matrigel=1:1 (v/v)) containing 10×10 6 RT112/84 cells and base glue was subcutaneously inoculated on the right back of each mouse. When the average tumor volume reached 188 mm 3 , they were divided into groups by stratified randomization, with 8 mice in each group. The control compound TAS-120 and the test compound A were dissolved into an appropriate solution with an aqueous solution containing 0.5% (w/v) MC and 0.5% (v/v) Tween 80, and were administered to mice by gavage at a dose volume of 10 mL/kg , Continuous administration for 20 days. The tumor diameter was measured with vernier calipers twice a week. The calculation formula of tumor volume is: V=0.5a×b 2 , a and b represent the long diameter and short diameter of the tumor, respectively.
化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)表示。TGI(%)的计算:TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积))/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]×100%。The anti-tumor efficacy of the compound is expressed by the tumor growth inhibition rate TGI (%). Calculation of TGI(%): TGI(%)=[(1-(Average tumor volume at the end of a certain treatment group-average tumor volume at the beginning of the treatment group))/(Average tumor volume at the end of treatment in the solvent control group Volume-average tumor volume at the start of treatment in the solvent control group)]×100%.
实验结果如表11所示。The experimental results are shown in Table 11.
表11:化合物A对RT112/84异种移植瘤模型瘤体积(平均值±SEM,mm 3,n=8)的影响 Table 11: Effect of compound A on tumor volume of RT112/84 xenograft tumor model (mean ± SEM, mm 3 , n=8)
Figure PCTCN2020127211-appb-000013
Figure PCTCN2020127211-appb-000013
结论:化合物A在5、10和20mg/kg剂量下每天一次(QD)给予,均具有显著的抑瘤效果。给药20天,抑瘤率(TGI)分别为67%、79%和83%,与溶剂对照组相比p值均小于0.05。在20mg/kg的剂量下,化合物A与TAS-120具有相当的抑瘤效果。Conclusion: Compound A administered once a day (QD) at doses of 5, 10 and 20 mg/kg, all have significant anti-tumor effects. After 20 days of administration, the tumor inhibition rate (TGI) was 67%, 79%, and 83%, respectively, and the p values were all less than 0.05 compared with the solvent control group. At a dose of 20 mg/kg, compound A and TAS-120 have comparable anti-tumor effects.
实施例8:人肝癌LI-03-0332皮下异种移植模型的体内药效学评价Example 8: In vivo pharmacodynamic evaluation of human liver cancer LI-03-0332 subcutaneous xenograft model
采用FGFR3高表达的LI-03-0332人源肝癌组织接种后裸鼠构建的皮下移植瘤模型,测定了化合物A对人肝癌的抑制效果。A subcutaneous xenograft tumor model constructed by nude mice after inoculation with LI-03-0332 human liver cancer tissue with high expression of FGFR3 was used to determine the inhibitory effect of compound A on human liver cancer.
肿瘤组织:人源肝癌LI-03-0332模型的建立最初来源于外科手术 切除的临床样本。标本的采集使用严格遵守国家、医院以及公司有关伦理的法律法规。传代命名规则为:肿瘤样本接种于裸鼠后为P0代,继续传代为P1代,以此类推,复苏的样本命名为FP,本次实验中使用的肿瘤组织为FP9代。Tumor tissue: The LI-03-0332 model of human liver cancer was originally established from clinical samples resected by surgery. The collection and use of specimens strictly abide by national, hospital and company-related ethical laws and regulations. The passage naming rule is: the tumor sample is inoculated into nude mice as the P0 generation, and the subsequent passage is the P1 generation, and so on, the recovered sample is named FP, and the tumor tissue used in this experiment is the FP9 generation.
试验步骤:将LI-03-0332肿瘤组织剔除坏死组织后切成20-30mm 3的小块,加基底胶后,将肝癌肿瘤组织皮下接种于每只小鼠的右后背,共接种149只小鼠。在接种后第22天,测量肿瘤平均体积达到135mm 3时,依据肿瘤体积和动物体重采用随机分层分组方法分组,溶剂对照组6只小鼠,其余组别每组8只小鼠。对照化合物TAS-120及受试化合物A用含0.5%(w/v)MC和0.5%(v/v)吐温80的水溶液溶解成适当溶液,以10mL/kg给药容积灌胃给予小鼠,连续给药21天。每周两次用游标卡尺测量肿瘤直径。肿瘤体积的计算公式为:V=0.5a×b 2,a和b分别表示肿瘤的长径和短径。 Test procedure: The LI-03-0332 tumor tissue was cut into 20-30mm 3 pieces after removing the necrotic tissue. After adding base glue, the liver cancer tumor tissue was subcutaneously inoculated on the right back of each mouse, a total of 149 mice were inoculated Mice. On the 22nd day after inoculation, when the measured average tumor volume reached 135mm 3 , random stratified grouping was used according to tumor volume and animal body weight. There were 6 mice in the solvent control group and 8 mice in each of the remaining groups. The control compound TAS-120 and the test compound A were dissolved into an appropriate solution with an aqueous solution containing 0.5% (w/v) MC and 0.5% (v/v) Tween 80, and were administered to mice by gavage at a dose volume of 10 mL/kg , Continuous administration for 21 days. The tumor diameter was measured with vernier calipers twice a week. The calculation formula of tumor volume is: V=0.5a×b 2 , a and b represent the long diameter and short diameter of the tumor, respectively.
化合物的抑瘤疗效用肿瘤生长抑制率TGI(%)表示。TGI(%)的计算:TGI(%)=[(1-(某处理组给药结束时平均瘤体积-该处理组开始给药时平均瘤体积))/(溶剂对照组治疗结束时平均瘤体积-溶剂对照组开始治疗时平均瘤体积)]×100%。The anti-tumor efficacy of the compound is expressed by the tumor growth inhibition rate TGI (%). Calculation of TGI(%): TGI(%)=[(1-(Average tumor volume at the end of a certain treatment group-average tumor volume at the beginning of the treatment group))/(Average tumor volume at the end of treatment in the solvent control group Volume-average tumor volume at the start of treatment in the solvent control group)]×100%.
实验结果如表12所示。The experimental results are shown in Table 12.
表12:化合物A对LI-03-0332异种移植瘤模型瘤体积(平均值±SEM,mm 3)的影响 Table 12: Effect of compound A on tumor volume (mean ± SEM, mm 3 ) of LI-03-0332 xenograft tumor model
Figure PCTCN2020127211-appb-000014
Figure PCTCN2020127211-appb-000014
注:溶剂对照组含6只动物,其它组为8只动物。Note: The solvent control group contains 6 animals, and the other groups contain 8 animals.
结论:化合物A在5、10和20mg/kg剂量下每天一次(QD)给予, 均具有显著的抑瘤效果。给药21天,抑瘤率(TGI)分别为81%、79%和87%,与溶剂对照组相比p值均小于0.0001。在20mg/kg的剂量下,化合物A与TAS-120具有相当的抑瘤效果。Conclusion: Compound A administered once a day (QD) at doses of 5, 10, and 20 mg/kg, all have significant anti-tumor effects. After 21 days of administration, the tumor inhibition rate (TGI) was 81%, 79%, and 87%, respectively, and the p-values were all less than 0.0001 compared with the solvent control group. At a dose of 20 mg/kg, compound A and TAS-120 have comparable anti-tumor effects.
实施例9:对重要激酶的脱靶效果评价Example 9: Evaluation of off-target effects of important kinases
利用eurofins公司的Enzyme Profiling Services技术平台,分析了化合物A对多种非靶点激酶活性的抑制作用。化合物对每个激酶的检测均使用Eurofins标准的Kinase Profiler TM实验流程,该流程遵循相关的标准操作规程。蛋白激酶(除了ATM(h)和DNA-PK(h))测试以辐射量进行检测,而脂质激酶、ATM(h)、ATR/ATRIP(h)和DNA-PK(h)的测试以
Figure PCTCN2020127211-appb-000015
进行检测。
Using Eurofins' Enzyme Profiling Services technology platform, the inhibitory effect of compound A on the activity of a variety of non-target kinases was analyzed. The detection of the compound for each kinase uses the Eurofins standard Kinase Profiler TM experimental procedure, which follows the relevant standard operating procedures. Protein kinase (except ATM(h) and DNA-PK(h)) tests are carried out by the amount of radiation, while lipid kinase, ATM(h), ATR/ATRIP(h) and DNA-PK(h) are tested by
Figure PCTCN2020127211-appb-000015
Perform testing.
试验步骤:取适量测试化合物A的50倍工作液(100%DMSO溶解)到测试孔(化合物A的终浓度为0.1μM)中,然后加入混匀的激酶和底物混合溶液;加入选定的浓度的ATP溶液开始反应;激酶和底物的混合物与化合物在ATP加入前不需要预孵育。Test procedure: Take an appropriate amount of 50 times working solution of test compound A (100% DMSO dissolved) into the test hole (the final concentration of compound A is 0.1μM), then add the mixed solution of kinase and substrate; add the selected The concentration of ATP solution starts to react; the mixture of kinase and substrate and compound do not need to be pre-incubated before ATP is added.
实验结果如表13所示。The experimental results are shown in Table 13.
表13:脱靶效果评价结果Table 13: Evaluation results of off-target effect
Figure PCTCN2020127211-appb-000016
Figure PCTCN2020127211-appb-000016
结论:在0.1μM浓度下,化合物A对所检测的50余种激酶无明显的抑 制作用。Conclusion: At a concentration of 0.1 μM, compound A has no obvious inhibitory effect on more than 50 kinases tested.
实施例10:安全药理学评价Example 10: Safety pharmacology evaluation
使用功能性观察组合测试(FOB),评估受试化合物A单次灌胃给药对SD大鼠中枢神经系统功能的影响。将雌雄各20只大鼠分成4组,每组雌雄各5只,口服给予溶媒(含0.5%(重量/体积,w/v)甲基纤维素和0.2%(w/v)的吐温80的水溶液)或0.5、1.2和3mg/kg剂量的化合物A。所有动物给药体积为5mL/kg。本试验对动物进行死亡率观察和体重称量。试验前、给药后2小时,给药后8小时,和给药后24小时各进行了一次FOB观察。观察指标包括运动功能、行为改变、协调功能、感觉/运动反射和体温。结果,各观察时间点均未见与供试品相关的改变。在本试验条件下,未见受试化合物A对大鼠中枢神经系统的影响。The Functional Observation Combination Test (FOB) was used to evaluate the effect of single intragastric administration of test compound A on the central nervous system function of SD rats. Divide 20 male and female rats into 4 groups, each of which has 5 male and female rats. Orally administered a vehicle (containing 0.5% (weight/volume, w/v) methylcellulose and 0.2% (w/v) Tween 80). Aqueous solution) or compound A at doses of 0.5, 1.2 and 3 mg/kg. The administration volume for all animals was 5 mL/kg. In this experiment, animals were observed for mortality and weighed. FOB observations were performed before the test, 2 hours after the administration, 8 hours after the administration, and 24 hours after the administration. Observation indicators include motor function, behavior change, coordination function, sensory/motor reflex and body temperature. As a result, no changes related to the test article were seen at each observation time point. Under the test conditions, there was no effect of test compound A on the central nervous system of rats.
使用遥测方法,评价经口灌胃给予受试化合物A对清醒比格犬心血管参数的影响。雌雄犬经口给予对照制剂(溶媒:含0.5%(w/v)甲基纤维素和0.2%(w/v)吐温80的水溶液),以及0.3、0.6或2mg/kg剂量的受试化合物A。每天对动物进行存活率观察。在非给药日进行了每天2次的笼边观察,在每次给药前和给药后各进行一次详细临床观察。在每次给药前至少2小时至给药后约24小时连续记录所有实验动物的心电图、心率和血压。打印每次给药的给药前2个时间点(至少间隔30分钟)及给药后6个时间点(给药后0.5小时、1小时、2小时、4小时、12小时和24小时)的约30秒长度的心电图波形。结果显示,在本试验条件下,比格犬单次经口灌胃给予0.3、0.6和2mg/kg剂量的受试化合物A,给药后24小时内,未见供试品相关的心血管指标变化。Using telemetry, the effects of oral administration of test compound A on the cardiovascular parameters of awake beagle dogs were evaluated. Female and male dogs were orally administered a control formulation (vehicle: an aqueous solution containing 0.5% (w/v) methylcellulose and 0.2% (w/v) Tween 80), and the test compound at a dose of 0.3, 0.6 or 2 mg/kg A. The survival rate of the animals is observed every day. Cage-side observations were conducted twice a day on non-dosing days, and detailed clinical observations were performed before and after each dosing. The electrocardiogram, heart rate and blood pressure of all experimental animals were continuously recorded at least 2 hours before each administration to about 24 hours after administration. Print the 2 time points before administration (at least 30 minutes apart) and 6 time points after administration (0.5 hour, 1 hour, 2 hours, 4 hours, 12 hours and 24 hours after administration) for each administration An ECG waveform with a length of about 30 seconds. The results showed that under the test conditions, beagle dogs were given a single oral gavage of 0.3, 0.6 and 2 mg/kg of test compound A. Within 24 hours after administration, there were no cardiovascular indicators related to the test product. Variety.
对SD大鼠单次口服灌胃给予受试化合物A,评估其对呼吸功能的影响。将雌雄各20只大鼠分成4组,每组雌雄各5只,口服给予对照制剂(溶媒:含0.5%(w/v)甲基纤维素和0.2%(w/v)吐温80水溶液)或0.5、1.2或3mg/kg的受试化合物A。所有动物给药体积为5mL/kg。给药前,采集约15分钟的呼吸数据(潮气量、呼吸频率、每分钟通气量)作为分组前呼吸参数基线。给药当天采集时间点为给药前,给药后2小时,给药后8小时,和给药后24小时各采集一次大约连续15分钟的数据。结果显示,单次口服给药后24小时,未见供 试品对大鼠呼吸频率、潮气量和每分钟通气量有影响。A single oral gavage of the test compound A was given to SD rats to evaluate its effect on respiratory function. Divide 20 female and male rats into 4 groups, each group has 5 male and female, and the control formulation (vehicle: containing 0.5% (w/v) methyl cellulose and 0.2% (w/v) Tween 80 aqueous solution) was administered orally. Or 0.5, 1.2 or 3 mg/kg of test compound A. The administration volume for all animals was 5 mL/kg. Before administration, about 15 minutes of respiratory data (tidal volume, respiratory rate, ventilation volume per minute) were collected as the baseline of respiratory parameters before grouping. The collection time points on the day of administration were before the administration, 2 hours after the administration, 8 hours after the administration, and 24 hours after the administration, and collected data for approximately 15 minutes. The results showed that 24 hours after a single oral administration, the test article had no effect on the rat's respiratory rate, tidal volume and minute ventilation.
实验结果如表14所示。The experimental results are shown in Table 14.
表14:安全药理学试验结果Table 14: Results of safety pharmacology test
Figure PCTCN2020127211-appb-000017
Figure PCTCN2020127211-appb-000017
结论:化合物A对SD大鼠的中枢神经系统和呼吸系统、对比格犬的心血管系统均无影响,提示化合物A具有较高的安全性。Conclusion: Compound A has no effect on the central nervous system and respiratory system of SD rats and the cardiovascular system of Beagle dogs, suggesting that compound A has high safety.
实施例11:急性毒性试验Example 11: Acute toxicity test
1.大鼠单次灌胃给药毒性试验1. Toxicity test of single intragastric administration in rats
试验方法:将40只大鼠随机分成4组,每组10只,雌雄各半,分别单次经口灌胃给予化合物A和对照溶剂,化合物A的剂量分别为10、40或400mg/kg,溶剂为含0.5%(w/v)MC和0.2%(w/v)吐温80的水溶液,以检测化合物A的最大耐受剂量。给药体积是10mL/kg,观察期是14天。Test method: 40 rats were randomly divided into 4 groups, 10 rats in each group, half male and half male, and compound A and the control solvent were administered by oral gavage. The dose of compound A was 10, 40 or 400 mg/kg, respectively. The solvent is an aqueous solution containing 0.5% (w/v) MC and 0.2% (w/v) Tween 80 to detect the maximum tolerated dose of compound A. The administration volume is 10 mL/kg, and the observation period is 14 days.
试验结果:仅在400mg/kg剂量组观察到黄色异常粪便和总胆红素轻微升高。其他剂量组雌雄动物未见供试品相关的死亡、临床症状、肉眼病变,体重变化、摄食量、临床检验(血液学、血凝、血清生化、尿液分析)与溶剂对照组相比无显著差异。因此,在本试验条件下单次给药的最大耐受剂量(MTD)为400mg/kg。Test results: Only in the 400mg/kg dose group, abnormal yellow stools and a slight increase in total bilirubin were observed. There were no deaths, clinical symptoms, gross lesions, weight changes, food intake, and clinical tests (hematology, blood coagulation, serum biochemistry, urinalysis) related to the test product in the female and male animals in the other dose groups compared with the solvent control group. difference. Therefore, the maximum tolerated dose (MTD) for a single administration under the experimental conditions is 400 mg/kg.
2.犬单次灌胃给药毒性试验2. Toxicity test of single intragastric administration in dogs
试验方法:8只比格犬,随机分成4组,每组2只犬,雌雄各半,分别单次经口灌胃给予化合物A和对照溶剂,化合物A的剂量分别为15、50或250mg/kg,溶剂为含0.5%(w/v)MC和0.2%(w/v)吐温80的水溶液。给药体积为5mL/kg。动物给药前过夜禁食。观察期是14天。雌雄动物于第21天进行剖检。Test method: 8 beagle dogs were randomly divided into 4 groups, 2 dogs in each group, half male and half male, and compound A and the control solvent were given by oral gavage. The dose of compound A was 15, 50 or 250 mg/ kg, the solvent is an aqueous solution containing 0.5% (w/v) MC and 0.2% (w/v) Tween 80. The administration volume is 5mL/kg. The animals were fasted overnight before dosing. The observation period is 14 days. Both males and females undergo necropsy on the 21st day.
试验结果:比格犬单剂量灌胃给予15、50或250mg/kg的化合物 A,未产生供试品相关的死亡率、临床症状、体重、摄食量、血液学、血凝、血清生化、尿液分析参数、肉眼病变和组织病理学的有害性反应。因此认为本试验条件下雌雄比格犬最大耐受量为(MTD)为250mg/kg。Test results: A single dose of 15, 50, or 250 mg/kg of compound A was administered to beagle dogs. There were no mortality, clinical symptoms, body weight, food intake, hematology, blood coagulation, serum biochemistry, urine Fluid analysis parameters, gross pathological changes, and histopathological adverse reactions. Therefore, it is believed that the maximum tolerable dose (MTD) of male and female beagle dogs under this test condition is 250 mg/kg.
结论:化合物A单次灌胃给药毒性试验耐受剂量较高,具有良好的安全性。Conclusion: Compound A has a high tolerable dose in toxicity test of single intragastric administration and has good safety.
已经通过上述实施例对本发明的各个方面进行了例示。显然,上述实施例仅仅是为了清楚地举例说明的目的而给出,而并非是为了对本发明的范围做出任何限定。对于所属领域的普通技术人员来说,在上述举例说明的基础上还可以做出其它不同形式的变化或变动。这里无需也无法对所有的实施方式予以穷举。而由此所引伸出的显而易见的变化或修饰仍处于本发明创造的保护范围之中。The various aspects of the present invention have been exemplified by the above-mentioned embodiments. Obviously, the above-mentioned embodiments are only given for the purpose of clear illustration, and are not intended to limit the scope of the present invention in any way. For those of ordinary skill in the art, other changes or changes in different forms can be made on the basis of the above examples. It is unnecessary and impossible to list all the implementation methods here. The obvious changes or modifications derived from this are still within the scope of protection created by the present invention.

Claims (10)

  1. 一种FGFR抑制剂化合物A或其药学上可接受的盐在用于制备治疗FGFR相关肿瘤的药物中的用途,其中所述化合物A具有如下结构:Use of an FGFR inhibitor compound A or a pharmaceutically acceptable salt thereof in the preparation of a medicament for treating FGFR-related tumors, wherein the compound A has the following structure:
    Figure PCTCN2020127211-appb-100001
    Figure PCTCN2020127211-appb-100001
  2. 根据权利要求1所述的用途,其中所述FGFR相关肿瘤是消化或泌尿系统肿瘤中的一种或多种;优选为胃癌、肝癌、尿路上皮癌、胆管癌中的任何一种或其任何组合。The use according to claim 1, wherein the FGFR-related tumor is one or more of digestive or urinary system tumors; preferably any one of gastric cancer, liver cancer, urothelial cancer, cholangiocarcinoma, or any of them combination.
  3. 根据权利要求2所述的用途,其中所述肝癌为肝细胞癌、肝内胆管细胞癌、混合性肝癌中的任何一种或其任何组合;所述尿路上皮癌为膀胱癌;所述胆管癌为肝内胆管癌、肝门部胆管癌、远端胆管癌中的任何一种或其任何组合,优选为肝内胆管癌。The use according to claim 2, wherein the liver cancer is any one or any combination of hepatocellular carcinoma, intrahepatic cholangiocarcinoma, mixed liver cancer; the urothelial cancer is bladder cancer; the bile duct The cancer is any one or any combination of intrahepatic cholangiocarcinoma, hilar cholangiocarcinoma, and distal cholangiocarcinoma, and is preferably intrahepatic cholangiocarcinoma.
  4. 根据权利要求1-3中的任一项所述的用途,其中所述化合物A或其药学上可接受的盐是所述药物中的唯一的活性成分,或与一种或多种其它靶向药物或化疗药物联合使用。The use according to any one of claims 1 to 3, wherein the compound A or a pharmaceutically acceptable salt thereof is the only active ingredient in the drug, or is combined with one or more other targets Combination of drugs or chemotherapy drugs.
  5. 根据权利要求1-3中的任一项所述的用途,其中所述药物被制成临床接受的制剂,优选口服制剂、注射制剂或外用制剂。The use according to any one of claims 1 to 3, wherein the drug is prepared into a clinically accepted preparation, preferably an oral preparation, an injection preparation or an external preparation.
  6. 根据权利要求1-3中的任一项所述的用途,其中所述化合物A或其药学上可接受的盐以从约0.001mg/kg至约1000mg/kg,优选从约0.01mg/kg至约100mg/kg,进一步优选从约0.02mg/kg至约50mg/kg,更进一步优选从约0.03mg/kg至约20mg/kg的每日给药剂量范围给予。The use according to any one of claims 1-3, wherein the compound A or a pharmaceutically acceptable salt thereof is from about 0.001 mg/kg to about 1000 mg/kg, preferably from about 0.01 mg/kg to It is administered in a daily dosage range of about 100 mg/kg, more preferably from about 0.02 mg/kg to about 50 mg/kg, and still more preferably from about 0.03 mg/kg to about 20 mg/kg.
  7. 根据权利要求1-3中的任一项所述的用途,其中所述药物含有治疗有效量的化合物A或其药学上可接受的盐。The use according to any one of claims 1 to 3, wherein the medicament contains a therapeutically effective amount of Compound A or a pharmaceutically acceptable salt thereof.
  8. 根据权利要求7所述的用途,其中所述治疗有效量是0.001-1000mg,优选0.01-100mg,进一步优选0.1-50mg,更进一步优选0.5-30mg。The use according to claim 7, wherein the therapeutically effective amount is 0.001-1000 mg, preferably 0.01-100 mg, more preferably 0.1-50 mg, still more preferably 0.5-30 mg.
  9. 根据权利要求1-3中的任一项所述的用途,其中所述化合物A或其药学上可接受的盐以单剂量给予或分剂量给予。The use according to any one of claims 1-3, wherein the compound A or a pharmaceutically acceptable salt thereof is administered in a single dose or in divided doses.
  10. 根据权利要求1-3中的任一项所述的用途,其中所述药物通过口服给予、注射给予、局部给予或体外给予,优选通过口服给予或注射给予。The use according to any one of claims 1 to 3, wherein the drug is administered by oral administration, injection administration, topical administration or in vitro administration, preferably by oral administration or injection administration.
PCT/CN2020/127211 2019-11-08 2020-11-06 Use of fgfr inhibitor WO2021089005A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202080076195.1A CN114641293A (en) 2019-11-08 2020-11-06 Application of FGFR inhibitor

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201911086889.9 2019-11-08
CN201911086889 2019-11-08
CN202010321669 2020-04-22
CN202010321669.6 2020-04-22

Publications (1)

Publication Number Publication Date
WO2021089005A1 true WO2021089005A1 (en) 2021-05-14

Family

ID=75849783

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2020/127211 WO2021089005A1 (en) 2019-11-08 2020-11-06 Use of fgfr inhibitor

Country Status (2)

Country Link
CN (1) CN114641293A (en)
WO (1) WO2021089005A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023232012A1 (en) * 2022-05-30 2023-12-07 石药集团中奇制药技术(石家庄)有限公司 Pharmaceutical combination and pharmaceutical composition for treating cancer

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103958512A (en) * 2012-01-19 2014-07-30 大鹏药品工业株式会社 3,5-disubstituted alkynylbenzene compound and salt thereof
CN104245700A (en) * 2011-12-15 2014-12-24 拜耳制药股份公司 Disubstituted benzothienyl-pyrrolotriazines and their use as fgfr kinase inhibitors
WO2017215485A1 (en) * 2016-06-16 2017-12-21 中国科学院上海药物研究所 New compound having fgfr inhibitory activity and preparation and application thereof
WO2018121650A1 (en) * 2016-12-29 2018-07-05 南京明德新药研发股份有限公司 Fgfr inhibitor
WO2019034076A1 (en) * 2017-08-15 2019-02-21 南京明德新药研发股份有限公司 Fgfr inhibitor and medical application thereof
WO2019034075A1 (en) * 2017-08-15 2019-02-21 南京明德新药研发股份有限公司 Fgfr and egfr inhibitor
WO2020164603A1 (en) * 2019-02-15 2020-08-20 石药集团中奇制药技术(石家庄)有限公司 Fgfr inhibitor compound in solid form and preparation method therefor

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104245700A (en) * 2011-12-15 2014-12-24 拜耳制药股份公司 Disubstituted benzothienyl-pyrrolotriazines and their use as fgfr kinase inhibitors
CN103958512A (en) * 2012-01-19 2014-07-30 大鹏药品工业株式会社 3,5-disubstituted alkynylbenzene compound and salt thereof
WO2017215485A1 (en) * 2016-06-16 2017-12-21 中国科学院上海药物研究所 New compound having fgfr inhibitory activity and preparation and application thereof
WO2018121650A1 (en) * 2016-12-29 2018-07-05 南京明德新药研发股份有限公司 Fgfr inhibitor
WO2019034076A1 (en) * 2017-08-15 2019-02-21 南京明德新药研发股份有限公司 Fgfr inhibitor and medical application thereof
WO2019034075A1 (en) * 2017-08-15 2019-02-21 南京明德新药研发股份有限公司 Fgfr and egfr inhibitor
WO2020164603A1 (en) * 2019-02-15 2020-08-20 石药集团中奇制药技术(石家庄)有限公司 Fgfr inhibitor compound in solid form and preparation method therefor

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GAIA CRISTINA GHEDINI; ROBERTO RONCA; MARCO PRESTA; ARIANNA GIACOMINI: "Future applications of FGF/FGFR inhibitors in cancer", EXPERT REVIEW OF ANTICANCER THERAPY, vol. 18, no. 9, 1 January 2018 (2018-01-01), GB, pages 861 - 872, XP009527867, ISSN: 1473-7140, DOI: 10.1080/14737140.2018.1491795 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023232012A1 (en) * 2022-05-30 2023-12-07 石药集团中奇制药技术(石家庄)有限公司 Pharmaceutical combination and pharmaceutical composition for treating cancer

Also Published As

Publication number Publication date
CN114641293A (en) 2022-06-17

Similar Documents

Publication Publication Date Title
US11666574B2 (en) Combination therapy involving diaryl macrocyclic compounds
EP2034835B1 (en) Non-toxic anti-cancer drug combining ascorbate, magnesium and a naphthoquinone
EP3429582B1 (en) Combination therapy for proliferative diseases
AU2020248270B2 (en) Chiauranib for treatment of small cell lung cancer
WO2021089005A1 (en) Use of fgfr inhibitor
EP3429572B1 (en) Combination therapy for proliferative diseases
TW202045173A (en) Cancer treatment
CN116850289A (en) Application of PLK4 targeted drug in treatment of platinum drug resistant tumor
CN106389437A (en) Application of low-dose sildenafil as antitumor drug
WO2022152296A1 (en) Application of pyrido[1,2-a]pyrimidinone analog
WO2022007136A1 (en) Composition and use thereof in preparation of medication for treating cancer
AU2020276392B2 (en) Combination therapy for proliferative conditions
WO2017031914A1 (en) Pharmaceutical composition of ibrutinib
KR20140008645A (en) Pharmaceutical composition for the preventing or treating brain tumor or glioblastoma having resistance of temodal containing azathioprine as an active ingredient
WO2020083187A1 (en) Use of combination of ar antagonist and parp inhibitor in preparation of drug for treating prostatic cancer
CN113453671A (en) Combination therapy of a Raf inhibitor and a CDK4/6 inhibitor for the treatment of cancer
RU2784869C1 (en) Chiauranib for treating small cell lung cancer
TWI835050B (en) Application of a pyrido[1,2-a]pyrimidinone analogue
WO2023051606A1 (en) Medical use of shp2 inhibitor in combination with egfr-tki in treatment and prevention of tumor diseases
WO2022179608A1 (en) Use of multi-target protein kinase inhibitor
WO2021219137A1 (en) Aminopyridine derivative for treating diseases caused by met genetic abnormalities
KR101378410B1 (en) Pharmaceutical composition for the preventing or treating brain tumor or glioblastoma having resistance of Temodal containing Benzydamine as an active ingredient
US20230117545A1 (en) Pharmaceutical composition comprising protein kinase inhibitor and chemotherapeutic drug and use thereof
CN115006397A (en) Pharmaceutical application for preventing or treating tumor diseases
CN117731674A (en) Application of ursodeoxycholic acid berberine salt in preparing medicine for preventing and/or treating digestive tract inflammation

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: 20884825

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: 20884825

Country of ref document: EP

Kind code of ref document: A1