[go: up one dir, main page]

CN114948964B - Use of multi-target protein kinase inhibitors - Google Patents

Use of multi-target protein kinase inhibitors Download PDF

Info

Publication number
CN114948964B
CN114948964B CN202210178601.6A CN202210178601A CN114948964B CN 114948964 B CN114948964 B CN 114948964B CN 202210178601 A CN202210178601 A CN 202210178601A CN 114948964 B CN114948964 B CN 114948964B
Authority
CN
China
Prior art keywords
compound
use according
biliary tract
tract cancer
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN202210178601.6A
Other languages
Chinese (zh)
Other versions
CN114948964A (en
Inventor
徐雯
谢颖
魏苗苗
马玉秀
史慧静
孙静
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
CSPC Zhongqi Pharmaceutical Technology Shijiazhuang Co Ltd
Original Assignee
CSPC Zhongqi Pharmaceutical Technology Shijiazhuang Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by CSPC Zhongqi Pharmaceutical Technology Shijiazhuang Co Ltd filed Critical CSPC Zhongqi Pharmaceutical Technology Shijiazhuang Co Ltd
Publication of CN114948964A publication Critical patent/CN114948964A/en
Application granted granted Critical
Publication of CN114948964B publication Critical patent/CN114948964B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Landscapes

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

Abstract

The present application relates to the use of a multi-target protein kinase inhibitor or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment or prophylaxis of a condition associated with one or more of RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases, particularly biliary tract cancer, and methods of treatment thereof. The compound shown in the formula A or pharmaceutically acceptable salt thereof has good inhibitory activity on various kinases, has good inhibitory activity on human bile duct cancer cells and intrahepatic bile duct cancer cells in vitro, shows good safety in clinical phase I tests, can control disease progression of patients with advanced or metastatic biliary tract malignant tumors which fail or are intolerant to primary treatment to a certain extent in clinical phase II tests, and has clinical potential for treating biliary tract cancers.

Description

Use of multi-target protein kinase inhibitors
Citation of related application
The present application claims the priority of an inventive patent application filed in china at 25/2/2021 entitled "use of multi-target protein kinase inhibitor", application number 202110209218.8, the entire contents of which are incorporated herein by reference.
Technical Field
The application belongs to the field of medicines, and in particular relates to application of a multi-target protein kinase inhibitor in preparing medicines for treating or preventing related diseases caused by one or more than one of RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases, especially application in preparing medicines for treating biliary tract cancer and a method for treating the diseases.
Background
Protein tyrosine kinases (Protein tyrosine kinases, PTKs) are a very important member of the protein kinase family, and the PTKs transfer the gamma-phosphate group on adenosine triphosphate to the protein tyrosine residue of a substrate, complete information transmission among cells by phosphorylating phenolic hydroxyl groups, and play a vital role in the processes of cell development, regulation, differentiation, migration, apoptosis and the like of tumor cells. If PTKs are out of control during the regulation process, the correct activation of downstream signal channels is affected, and then the regulation function of cell proliferation is disturbed, so that a plurality of diseases are caused, for example, the tyrosine kinase is excessively high to phosphorylate receptors and activate downstream signals, so that cells are over-transformed, proliferate, resist apoptosis, promote cell survival and further form malignant tumors.
Epidermal growth factor receptor (Epidermal Growth Factor Receptor, EGFR), fibroblast growth factor receptor (Fibroblast Growth Factor Receptors, FGFRs), platelet-derived growth factor receptor (Platelet-derived Growth Factor Receptor, PDGFR), and RET protein encoded by RET (Rearranged during Transfection) protooncogene are important members of PTKs, and are important targets for tumor treatment.
EGFR includes EGFR (ErbB-1), type 2 human EGFR HER-2 (ErbB-2), type 3 human EGFR HER3 (ErbB-3) and type 4 human EGFR HER4 (ErbB-4), wherein EGFR and HER-2 are the most closely related targets of EGFR family members. EGFR has been shown to exhibit over-expression, gene mutation or gene fusion in a variety of tumors such as lung cancer, stomach cancer, epidermoid carcinoma, kidney cancer, ovarian cancer, and the like.
FGFR mainly comprises four subtypes FGFR1/2/3/4, and the four subtypes are over-expressed or over-activated in the modes of gene amplification, mutation, fusion or ligand induction and the like, and have important roles in tumor cell proliferation, invasion and migration and tumor angiogenesis. The research shows that FGFRs show mutation, overexpression or overactivation in various tumors such as lung cancer, gastric cancer, cholangiocarcinoma, colorectal cancer, liver cancer and the like.
Biliary tract cancer (Biliary Tract Carcinoma, BTC) is a malignant tumor with invasive, genetic heterogeneity, including gallbladder cancer (Gallbladder Carcinoma, GBC), bile duct cancer (Cholangio Carcinoma, CC), ampullate cancer, etc., bile duct cancer including intrahepatic and extrahepatic bile duct cancer. BTC has a low incidence of about 3% of digestive tract tumors, with primary gallbladder cancer accounting for more than about 50% of BTC. The early stages of BTC have no subjective symptoms, and are difficult to pay attention before jaundice appears, about 60-70% of BTC patients are diagnosed in a stage of incapacitation or metastasis, the prognosis is poor, and the 5-year survival rate is less than 5%. BTC has a high recurrence rate and a high distant metastasis rate, and adjuvant therapy such as chemotherapy, radiotherapy, etc. is generally required to prolong the survival time of patients or improve symptoms as much as possible. At present, chemotherapy is mainly adopted for advanced BTC, and oxaliplatin and 5-fluorouracil (5-FU) are combined, oxaliplatin/irinotecan Li Tikang/5-FU and irinotecan and capecitabine are combined, but the curative effect is poor.
Peup Mi Jiati Ni (pemigatinib, pemazyre) is a selective FGFR inhibitor, and is approved by the United states food and drug administration to be marketed in 4/17/2020, and is used for adult treatment of advanced FGFR2 gene fusion type cholangiocarcinoma which is treated previously but has poor effect, becomes a targeting drug in the treatment history of first-stage cholangiocarcinoma, and terminates the era of cholangiocarcinoma chemotherapy-only method. However, in clinical trials, toxic side effects of pe Mi Jiati are also observed during the course of treatment, most commonly hyperphosphatemia, alopecia areata, diarrhea, nail toxicity, parageusia, nausea, constipation, stomatitis, dry eyes, vomiting, joint pain, etc., and another potential toxicity is ocular toxicity.
Tumors are often related to imbalance of regulation of multiple signal transduction pathways and multiple targets, and single-target drugs cannot necessarily achieve expected therapeutic effects, and are also limited in application by drug toxic and side effects and drug resistance, so that multi-target drugs become a new drug research direction. Compared with single-target drugs, multi-target drugs can act on multiple targets related to tumors, even though the activity of the multi-target drugs against the single targets is possibly reduced compared with that of the single-target drugs, the multi-target drugs can benefit from the synergistic effect generated by multi-target regulation, so that the total effect can be larger than the sum of the single effects, thereby obtaining better curative effect and generating smaller adverse reaction.
CN106660970B discloses compounds of general formula a, in particular compounds of formula 1 (example 22), which are multi-target inhibitors of protein tyrosine kinase activities such as RET, KDR, EGFR, FGFR, FLT-1, etc., and no related research reports on the use of the compounds in human biliary tract cancer treatment are available.
Disclosure of Invention
The application aims to solve the technical problem of providing a multi-target protein kinase inhibitor shown in a formula A or pharmaceutically acceptable salt thereof, in particular to the application of a compound 1 or pharmaceutically acceptable salt thereof in preparing medicines for treating or preventing one or more than one related diseases caused by RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinase.
In particular, in a first aspect of the application there is provided the use of a multi-target protein kinase inhibitor as shown in formula A, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment or prophylaxis of a condition associated with one or more of the RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases,
wherein,,
R 1 is-O (CH) 2 ) n R 3 Wherein n is 3, 4 or 5, R 3 is-NR e R f Wherein R is e And R is f Each at each occurrence independently of one another selected from methyl, or R e And R is f Together form- (CH) 2 ) 4 -;
Wherein R is 2 Is that
In one embodiment, R in the compound of formula A 1 Selected from the group consisting of
In one embodiment, the compound of formula A is compound 1,
in one embodiment, the related disorder is a related disorder caused by one or more of RET, KDR, EGFR, FGFR1/2/3, FLT-1, HER-2, LCK, ephA1, SRC kinase.
In one embodiment, the related disorder is a neoplastic disease; preferably, the neoplastic disease is a solid tumor; further preferably, the solid tumor is biliary tract cancer.
In one embodiment, the biliary tract cancer is caused by one or more than one of RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases.
In one embodiment, the biliary tract cancer is advanced biliary tract cancer.
In one embodiment, the biliary tract cancer is locally advanced biliary tract cancer.
In one embodiment, the biliary tract cancer is locally advanced biliary tract cancer that is not surgically resectable.
In one embodiment, the biliary tract cancer is metastatic biliary tract cancer.
In one embodiment, the biliary tract cancer is a biliary tract cancer that fails or is intolerant of treatment.
In one embodiment, the biliary tract cancer is biliary tract cancer that failed or was intolerant via first line therapy.
In one embodiment, the biliary tract cancer is gallbladder cancer or biliary tract cancer.
In one embodiment, the cholangiocarcinoma is intrahepatic cholangiocarcinoma and/or extrahepatic cholangiocarcinoma; preferably intrahepatic cholangiocarcinoma.
In one embodiment, the compound of formula a or a pharmaceutically acceptable salt thereof, in particular compound 1 or a pharmaceutically acceptable salt thereof, is used as the sole active ingredient for the preparation of the medicament.
In one embodiment, a compound of formula a or a pharmaceutically acceptable salt thereof, particularly compound 1 or a pharmaceutically acceptable salt thereof, is used in combination with one or more of the other therapeutic agents to prepare the medicament. The other therapeutic agents include, but are not limited to, targeted drugs or chemotherapeutic drugs, which are clinically used for treating tumor-related diseases.
In a second aspect of the application there is provided a multi-target protein kinase inhibitor as shown in formula A, or a pharmaceutically acceptable salt thereof, for use in the treatment or prophylaxis of a condition associated with one or more of the RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases,
wherein,,
R 1 is-O (CH) 2 ) n R 3 Wherein n is 3, 4 or 5, R 3 is-NR e R f Wherein R is e And R is f Each at each occurrence independently of one another selected from methyl, or R e And R is f Together form- (CH) 2 ) 4 -;
Wherein R is 2 Is that
In one embodiment, R in the compound of formula A 1 Selected from the group consisting of
In one embodiment, the compound of formula A is compound 1,
in one embodiment, the related disorder is a related disorder caused by one or more of RET, KDR, EGFR, FGFR1/2/3, FLT-1, HER-2, LCK, ephA1, SRC kinase.
In one embodiment, the related disorder is a neoplastic disease; preferably, the neoplastic disease is a solid tumor; further preferably, the solid tumor is biliary tract cancer.
In one embodiment, the biliary tract cancer is caused by one or more than one of RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases.
In one embodiment, the biliary tract cancer is advanced biliary tract cancer.
In one embodiment, the biliary tract cancer is locally advanced biliary tract cancer.
In one embodiment, the biliary tract cancer is locally advanced biliary tract cancer that is not surgically resectable.
In one embodiment, the biliary tract cancer is metastatic biliary tract cancer.
In one embodiment, the biliary tract cancer is a biliary tract cancer that fails or is intolerant of treatment.
In one embodiment, the biliary tract cancer is biliary tract cancer that failed or was intolerant via first line therapy.
In one embodiment, the biliary tract cancer is gallbladder cancer or biliary tract cancer.
In one embodiment, the cholangiocarcinoma is intrahepatic cholangiocarcinoma and/or extrahepatic cholangiocarcinoma; preferably intrahepatic cholangiocarcinoma.
In one embodiment, the compound of formula a or a pharmaceutically acceptable salt thereof, in particular compound 1 or a pharmaceutically acceptable salt thereof, is used as the sole active ingredient for the preparation of a medicament.
In one embodiment, a compound of formula a or a pharmaceutically acceptable salt thereof, particularly compound 1 or a pharmaceutically acceptable salt thereof, is used in combination with one or more of the other therapeutic agents in the manufacture of a medicament, or for the treatment or prevention of a related disorder caused by one or more of the RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases. The other therapeutic agents include, but are not limited to, targeted drugs or chemotherapeutic drugs, which are clinically used for treating tumor-related diseases.
In a third aspect of the application there is provided the use of a multi-target protein kinase inhibitor of formula A or a pharmaceutically acceptable salt thereof as a medicament,
wherein,,
R 1 is-O (CH) 2 ) n R 3 Wherein n is 3, 4 or 5, R 3 is-NR e R f Wherein R is e And R is f Each at each occurrence independently of one another selected from methyl, or R e And R is f Together form- (CH) 2 ) 4 -;
Wherein R is 2 Is that
In one embodiment, R in the compound of formula A 1 Selected from the group consisting of
In one embodiment, the compound of formula A is compound 1,
in one embodiment, the medicament is for treating a related disorder caused by one or more of RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases.
In one embodiment, the related disorder is a related disorder caused by one or more of RET, KDR, EGFR, FGFR1/2/3, FLT-1, HER-2, LCK, ephA1, SRC kinase.
In one embodiment, the related disorder is a neoplastic disease; preferably, the neoplastic disease is a solid tumor; further preferably, the solid tumor is biliary tract cancer.
In one embodiment, the biliary tract cancer is caused by one or more than one of RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases.
In one embodiment, the biliary tract cancer is advanced biliary tract cancer.
In one embodiment, the biliary tract cancer is locally advanced biliary tract cancer.
In one embodiment, the biliary tract cancer is locally advanced biliary tract cancer that is not surgically resectable.
In one embodiment, the biliary tract cancer is metastatic biliary tract cancer.
In one embodiment, the biliary tract cancer is a biliary tract cancer that fails or is intolerant of treatment.
In one embodiment, the biliary tract cancer is biliary tract cancer that failed or was intolerant via first line therapy.
In one embodiment, the biliary tract cancer is gallbladder cancer or biliary tract cancer.
In one embodiment, the cholangiocarcinoma is intrahepatic cholangiocarcinoma and/or extrahepatic cholangiocarcinoma; preferably intrahepatic cholangiocarcinoma.
In one embodiment, the compound of formula a or a pharmaceutically acceptable salt thereof, in particular compound 1 or a pharmaceutically acceptable salt thereof, is used as the sole active ingredient for the preparation of the medicament.
In one embodiment, a compound of formula a or a pharmaceutically acceptable salt thereof, particularly compound 1 or a pharmaceutically acceptable salt thereof, is used in combination with one or more of the other therapeutic agents to prepare the medicament. The other therapeutic agents include, but are not limited to, targeted drugs or chemotherapeutic drugs, which are clinically used for treating tumor-related diseases.
In a fourth aspect of the application there is provided a method of treating or preventing a related disorder caused by one or more of RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases, the method comprising administering to a subject or patient a therapeutically effective amount of a multi-target protein kinase inhibitor of formula A or a pharmaceutically acceptable salt thereof,
wherein,,
R 1 is-O (CH) 2 ) n R 3 Wherein n is 3, 4 or 5, R 3 is-NR e R f Wherein R is e And R is f Each at each occurrence independently of one another selected from methyl, or R e And R is f Together form- (CH) 2 ) 4 -;
Wherein R is 2 Is that
In one embodiment, R in the compound of formula A 1 Selected from the group consisting of
In one embodiment, the compound of formula A is compound 1,
in one embodiment, the related disorder is a related disorder caused by one or more of RET, KDR, EGFR, FGFR1/2/3, FLT-1, HER-2, LCK, ephA1, SRC kinase.
In one embodiment, the related disorder is a neoplastic disease; preferably, the neoplastic disease is a solid tumor; further preferably, the solid tumor is biliary tract cancer.
In one embodiment, the biliary tract cancer is caused by one or more than one of RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases.
In one embodiment, the biliary tract cancer is advanced biliary tract cancer.
In one embodiment, the biliary tract cancer is locally advanced biliary tract cancer.
In one embodiment, the biliary tract cancer is locally advanced biliary tract cancer that is not surgically resectable.
In one embodiment, the biliary tract cancer is metastatic biliary tract cancer.
In one embodiment, the biliary tract cancer is a biliary tract cancer that fails or is intolerant of treatment.
In one embodiment, the biliary tract cancer is biliary tract cancer that failed or was intolerant via first line therapy.
In one embodiment, the biliary tract cancer is gallbladder cancer or biliary tract cancer.
In one embodiment, the cholangiocarcinoma is intrahepatic cholangiocarcinoma and/or extrahepatic cholangiocarcinoma; preferably intrahepatic cholangiocarcinoma.
In one embodiment, the compound of formula a or a pharmaceutically acceptable salt thereof, in particular compound 1 or a pharmaceutically acceptable salt thereof, is used as the sole active ingredient for the preparation of a medicament.
In one embodiment, a compound of formula a or a pharmaceutically acceptable salt thereof, particularly compound 1 or a pharmaceutically acceptable salt thereof, is used in combination with one or more of the other therapeutic agents in the manufacture of a medicament or for the treatment or prevention of a related disorder caused by one or more of the RET, KDR, EGFR, FGFR, FLT, HER, LCK, ephA, SRC kinases. The other therapeutic agents include, but are not limited to, targeted drugs or chemotherapeutic drugs, which are clinically used for treating tumor-related diseases.
In a fifth aspect of the present application, there is provided a kit comprising a compound of formula a or a pharmaceutically acceptable salt thereof, in particular compound 1 or a pharmaceutically acceptable salt thereof, as described above (first, second, third, fourth aspects).
In one embodiment, the kit comprises one or more containers comprising a compound of formula a or a pharmaceutically acceptable salt thereof, particularly compound 1 or a pharmaceutically acceptable salt thereof.
In one embodiment, the kit comprises the aforementioned (first, second, third, fourth) medicament.
In the present application, the compound represented by formula a, particularly the pharmaceutically acceptable salt of compound 1, as described in the preceding (first, second, third, fourth, fifth) aspects is not particularly limited, and for example, the pharmaceutically acceptable salt of the compound represented by formula a includes, but is not limited to, hydrochloride, sulfate, oxalate, maleate, or malate; preferably hydrochloride, sulfate or maleate; further preferred is hydrochloride; even more preferably dihydrochloride; even more preferred is the anhydrous form of the dihydrochloride salt or a hydrate thereof.
In some embodiments, the pharmaceutically acceptable salt of the compound of formula A as described above (first, second, third, fourth, fifth) is the dihydrochloride salt of compound 1 (compound of formula 2),
in some embodiments, the compound of formula 2 is in crystalline form.
In some embodiments, the crystalline form of the compound of formula 2 (form I), using Cu-ka radiation, has a powder X-ray diffraction pattern in terms of 2θ degrees (°) with characteristic diffraction peaks (±0.2°): 12.4, 18.8, 20.3, 24.6.
Alternatively, using Cu-ka radiation, a powder X-ray diffraction pattern expressed in terms of 2θ angles (°) has characteristic diffraction peaks (±0.2°): 9.8, 12.4, 18.8, 20.3, 24.6.
Alternatively, using Cu-ka radiation, a powder X-ray diffraction pattern expressed in terms of 2θ angles (°) has characteristic diffraction peaks (±0.2°): 8.1, 9.8, 12.4, 18.8, 20.3, 24.6, 29.9.
Alternatively, using Cu-ka radiation, a powder X-ray diffraction pattern expressed in terms of 2θ angles (°) has characteristic diffraction peaks (±0.2°): 8.1, 9.8, 12.4, 18.8, 19.3, 20.3, 24.6, 28.6, 29.9.
Alternatively, using Cu-ka radiation, a powder X-ray diffraction pattern expressed in terms of 2θ angles (°) has characteristic diffraction peaks (±0.2°): 8.1, 9.8, 12.4, 16.1, 18.8, 19.3, 20.3, 24.6, 28.6, 29.9, 30.9.
Alternatively, using Cu-ka radiation, an X-ray powder diffraction pattern substantially as shown in figure 1 or figure 2 is provided.
In some embodiments, the crystalline form of the compound of formula 2 (form I) has a DSC profile with endothermic peaks at 231.0±5 ℃ and 284.2±5 ℃, respectively.
In some embodiments, the crystalline form of the compound of formula 2 (form I) begins to decompose with a TGA profile at 205.6±5 ℃.
In some embodiments, the aforementioned (first, second, third, fourth, fifth, the same below) medicament is formulated into a clinically acceptable formulation, including, but not limited to, an oral formulation, an injectable formulation, a topical formulation, or an external formulation; preferably an oral formulation; further preferred are tablets and capsules. In some embodiments, the aforementioned medicament comprises a therapeutically effective amount of a compound of formula A or a pharmaceutically acceptable salt thereof, administered in a daily dosage ranging from about 0.001mg/kg to about 1000mg/kg, preferably from about 0.01mg/kg to 800mg/kg, or from 0.1mg/kg to 600mg/kg, or from 1mg/kg to 500mg/kg, or from 2mg/kg to 400mg/kg, or from 3mg/kg to 300mg/kg, or from 4mg/kg to 200mg/kg, or from 5mg/kg to 100mg/kg, or from 10mg/kg to 50mg/kg. Preferably, the compound shown in the formula A or the pharmaceutically acceptable salt thereof is compound 1 or the pharmaceutically acceptable salt thereof.
In some embodiments, the aforementioned medicament contains 0.1-5000mg, preferably 1-1000mg, or 10-800mg, or 20-700mg, or 22-650mg, or 25-650mg, or 50-600mg, or 85-500mg, or 86-500mg, or 90-500mg, or 100-450mg, or 100-400mg, or 150-350mg, or 170-350mg, or 173-350mg, or 175-350mg, or 200-300mg, or 150-200mg of the compound represented by formula A or a pharmaceutically acceptable salt thereof per unit of preparation. The dosages are calculated for the compounds of formula A. Preferably, the compound shown in the formula A or the pharmaceutically acceptable salt thereof is compound 1 or the pharmaceutically acceptable salt thereof.
In one embodiment, the aforementioned medicament contains 0.1-5000mg, preferably 1-1000mg, or 10-800mg, or 25-700mg, or 50-600mg, or 80-500mg, or 90-500mg, or 100-450mg, or 100-400mg, or 150-350mg, or 200-300mg, or 150-200mg of dihydrochloride of the compound represented by formula A per formulation unit; illustrative examples include 25mg, 50mg, 100mg, 150mg, or 200mg. The dosages are calculated as the dihydrochloride salt of the compound represented by formula a in anhydrous form. Preferably, the dihydrochloride of the compound represented by the formula a is dihydrochloride of the compound 1. Further preferred is a crystalline form of the compound of formula 2; still more preferred is form I of the compound of formula 2.
In one embodiment, the aforementioned medicament comprises a therapeutically effective amount of a compound of formula a or a pharmaceutically acceptable salt thereof, administered in the following amounts: 20mg-1500mg per dose; preferably, 50mg to 1200mg per administration; preferably, 100mg to 1000mg per administration; preferably, 200mg to 800mg per administration; further preferably, 250mg to 700mg per administration; further preferably, 300mg to 600mg per administration; further preferably, 300mg to 550mg per administration; further preferably, 300mg to 500mg per administration; further preferably, 300mg to 450mg per administration; exemplary doses of administration include 20mg, 22mg, 25mg, 40mg, 43mg, 45mg, 50mg, 80mg, 85mg, 86mg, 90mg, 100mg, 150mg, 170mg, 173mg, 175mg, 180mg, 200mg, 250mg, 259mg, 260mg, 300mg, 302mg, 310mg, 340mg, 346mg, 350mg, 400mg, 380mg, 385mg, 389mg, 390mg, 400mg, 430mg, 432mg, 440mg, 450mg, 475mg, 500mg, 518mg, 520mg, 550mg, 560mg, 562mg, 600mg, 605mg, 610mg, 648mg, 650mg, 690mg, 691mg, 700mg, 750mg, 778mg, 800mg, or 900mg per administration. The doses are calculated as compound a. Preferably, the compound shown in the formula A or the pharmaceutically acceptable salt thereof is compound 1 or the pharmaceutically acceptable salt thereof.
In one embodiment, the aforementioned medicament comprises a therapeutically effective amount of a dihydrochloride salt of a compound represented by formula a, which is administered in the following amounts: 25mg-900mg per dose; preferably, 200mg to 800mg per administration; further preferably, 300mg to 700mg per administration; further preferably, 300mg to 600mg per administration; further preferably, 400mg to 600mg per administration; further preferably, 450mg to 600mg per administration; exemplary doses of administration include 25mg, 50mg, 100mg, 200mg, 300mg, 350mg, 400mg, 450mg, 500mg, 550mg, 600mg, 650mg, 700mg, 750mg, 800mg, or 900mg per administration. The doses are calculated as the dihydrochloride salt of compound a in anhydrous form. Preferably, the dihydrochloride of the compound represented by the formula a is dihydrochloride of the compound 1. Further preferred is a crystalline form of the compound of formula 2; still more preferred is form I of the compound of formula 2.
In one embodiment, the aforementioned medicament is administered once daily, twice daily, three times daily or four times daily at a daily dosing frequency; preferably once daily.
In one embodiment, the aforementioned medicament comprises a therapeutically effective amount of a compound of formula a or a pharmaceutically acceptable salt thereof, administered at a daily dose of 20mg to 1500mg; preferably 50mg to 1200mg; further preferably 100mg to 1000mg; further preferably 200mg to 800mg; further preferably 250mg to 700mg; further preferably 300mg to 600mg; further preferably 300mg to 550mg; further preferably 300mg to 500mg; further preferably 300mg to 450mg; exemplary daily administration doses include 20mg, 22mg, 25mg, 40mg, 43mg, 45mg, 50mg, 80mg, 85mg, 86mg, 90mg, 100mg, 150mg, 170mg, 173mg, 175mg, 180mg, 200mg, 250mg, 259mg, 260mg, 300mg, 302mg, 310mg, 340mg, 346mg, 350mg, 400mg, 380mg, 385mg, 389mg, 390mg, 400mg, 430mg, 432mg, 440mg, 450mg, 475mg, 500mg, 518mg, 520mg, 550mg, 560mg, 562mg, 600mg, 605mg, 610mg, 648mg, 650mg, 690mg, 691mg, 700mg, 750mg, 778mg, 800mg or 900mg. The doses are calculated as compound a. Preferably, the compound shown in the formula A or the pharmaceutically acceptable salt thereof is compound 1 or the pharmaceutically acceptable salt thereof. Wherein the daily administration frequency is once, twice, three times or more.
In one embodiment, the aforementioned medicament comprises a therapeutically effective amount of the dihydrochloride salt of the compound represented by formula a, which is administered in a daily dose of 25mg to 900mg; preferably 200mg to 800mg; further preferably 300mg to 700mg; further preferably 300mg to 600mg; further preferably 400mg to 600mg; further preferably 450mg to 600mg; exemplary daily dosing amounts include daily dosing of 25mg, 50mg, 100mg, 200mg, 300mg, 350mg, 400mg, 450mg, 500mg, 550mg, 600mg, 650mg, 700mg, 750mg, 800mg, or 900mg. The doses are calculated as the dihydrochloride salt of compound a in anhydrous form. Preferably, the dihydrochloride of the compound represented by the formula a is dihydrochloride of the compound 1. Further preferred is a crystalline form of the compound of formula 2; still more preferred is form I of the compound of formula 2. Wherein the daily administration frequency is once, twice, three times or more.
In one embodiment, the aforementioned medicament comprises a therapeutically effective amount of a compound of formula a or a pharmaceutically acceptable salt thereof, in an amount and frequency of administration of: the medicine is administrated once a day, and 20mg-1500mg is administrated each time; preferably, the administration is once daily, each time 50mg-1200mg; preferably, the administration is once daily, each time 100mg-1000mg; preferably, the administration is once daily, each time 200mg-800mg; further preferably, the administration is once daily, 250mg to 700mg per administration; further preferably, the administration is once daily, 300mg to 600mg per administration; further preferably, the administration is once daily, 300mg to 550mg each time; exemplary dosages and frequencies of administration include, once daily, 20mg, 22mg, 25mg, 40mg, 43mg, 45mg, 50mg, 80mg, 85mg, 86mg, 90mg, 100mg, 150mg, 170mg, 173mg, 175mg, 180mg, 200mg, 250mg, 259mg, 260mg, 300mg, 302mg, 310mg, 340mg, 346mg, 350mg, 400mg, 380mg, 385mg, 389mg, 390mg, 400mg, 430mg, 432mg, 440mg, 450mg, 475mg, 500mg, 518mg, 520mg, 550mg, 560mg, 562mg, 600mg, 605mg, 610mg, 648mg, 650mg, 690mg, 691mg, 700mg, 750mg, 778mg, 800mg, or 900mg. The doses are calculated as compound a. Preferably, the compound shown in the formula A or the pharmaceutically acceptable salt thereof is compound 1 or the pharmaceutically acceptable salt thereof.
In one embodiment, the aforementioned medicament comprises a therapeutically effective amount of the dihydrochloride salt of the compound represented by formula a, at a dosage and frequency of: the medicine is administrated once a day, and 25mg-900mg is administrated each time; preferably, the administration is once daily, each time 200mg-800mg; further preferably, the administration is once daily, 300mg to 700mg per administration; further preferably, the administration is once daily, 300mg to 600mg per administration; further preferably, the administration is once daily, each time 400mg-600mg; further preferably, the administration is once daily, each time 450mg-600mg; exemplary dosages and frequencies of administration include once daily administration of 25mg, 50mg, 100mg, 200mg, 300mg, 350mg, 400mg, 450mg, 500mg, 550mg, 600mg, 650mg, 700mg, 750mg, 800mg, or 900mg per administration. The doses are calculated as the dihydrochloride salt of compound a in anhydrous form. Preferably, the dihydrochloride of the compound represented by the formula a is dihydrochloride of the compound 1. Further preferred is a crystalline form of the compound of formula 2; still more preferred is form I of the compound of formula 2.
In one embodiment, the aforementioned medicament comprises a therapeutically effective amount of a compound of formula a or a pharmaceutically acceptable salt thereof, which is administered at a frequency of: once daily for 21 days, and stopping for 7 days, wherein every 28 days is a period. Preferably, the compound shown in the formula A or the pharmaceutically acceptable salt thereof is compound 1 or the pharmaceutically acceptable salt thereof.
In one embodiment, the aforementioned drugs are administered orally, by injection, topically or in vitro. In a preferred embodiment, the medicament is administered orally or by injection; preferably, the administration is by oral administration.
In some embodiments, the compound of formula a as described above (first, second, third, fourth, fifth) or a pharmaceutically acceptable salt thereof is a dihydrochloride salt of compound 1; preferably a crystalline form of the compound of formula 2; further preferred is form I of the compound of formula 2.
In one embodiment, the compound of formula a or a pharmaceutically acceptable salt thereof, in particular compound 1 or a pharmaceutically acceptable salt thereof, as described above (first, second, third, fourth, fifth) may be administered in a single dose or in divided doses; preferably, the administration is in a single dose.
Definition and description
The following terms and phrases used herein are intended to have the following meanings unless otherwise indicated. A particular phrase or terminology, unless otherwise specifically defined, should not be construed as being ambiguous or otherwise clear, but rather should be construed in a generic sense. When trade names are presented herein, it is intended to refer to their corresponding commercial products or active ingredients thereof.
In the present application, the therapeutically effective dose and dose frequency of the compound of formula a or a pharmaceutically acceptable salt thereof, particularly compound 1 or a pharmaceutically acceptable salt thereof, can be determined by methods such as modeling, dose escalation studies or clinical trials and by considering factors such as the nature and severity of the disease to be treated, the age, general condition and weight of the patient, and the particular compound administered, its pharmacokinetic properties, and route of administration.
The term "chemotherapeutic agent" as used herein refers to a chemical agent for treating tumors, including but not limited to cytotoxic, antimetabolite, antibiotic, alkaloid, hormonal, and other agents.
The term "targeted drug" as used herein refers to a drug or formulation thereof for targeted therapy that prevents cancer cells from growing or proliferating by interfering with specific molecules required for cancer or tumor proliferation, including but not limited to small molecule targeted drugs including but not limited to protein kinase inhibitors, such as protein tyrosine kinase inhibitors, and large molecule targeted drugs including but not limited to monoclonal antibodies, antibody conjugated drugs, and immunotherapeutic drugs.
The dosage of the compound represented by the formula a or a pharmaceutically acceptable salt thereof, particularly the compound 1 or a pharmaceutically acceptable salt thereof according to the present application may be calculated as a free base form, or may be calculated as a specific form for practical use, unless otherwise specified, and if it contains a hydrate, it may be calculated as a unit form other than water, or may be calculated as the hydrate form. For example, if the hydrochloride salt of compound 1 is used, then the hydrochloride salt of compound 1 is used; when the hydrate of the hydrochloride of compound 1 is used, the hydrochloride of compound 1 may be used, or the hydrate of the hydrochloride of compound 1 may be used.
The dosage of the compound shown as the formula A or the pharmaceutically acceptable salt thereof, particularly the compound 1 or the pharmaceutically acceptable salt thereof, can float within the clinically acceptable range, for example, 100mg per administration, and the administration dosage can float up and down within the range of 10% of the dosage, namely, 90mg to 110mg.
The first-line treatment of the application is not limited to the first-line chemotherapy scheme approved for treating biliary tract cancer, and also comprises systemic drug treatments such as non-first-line chemotherapy scheme, immunotherapy, targeted therapy and the like which are first used for treating biliary tract cancer after diagnosis, and the failure of the first-line treatment also comprises the occurrence of disease progression within 6 months of auxiliary chemotherapy.
The term "administered alone" as used herein means without the need for combination with other anti-tumor agents, but does not preclude the use of adjunctive agents that do not have anti-tumor effects.
The terms "2θ", "2θ angle" or "2θ angle" as used herein refer to diffraction angles in degrees or degrees, and the error range of 2θ may be ±0.5°, ±0.4°, ±0.3°, ±0.2°, or ±0.1°, unless otherwise specified; in some embodiments of the application, the error range of 2θ is ±0.2°.
The term "substantially as shown in the figures" means that at least 50%, or at least 60%, or at least 70%, or at least 80%, or at least 90%, or at least 95%, or at least 96%, or at least 97%, or at least 98%, or at least 99% of the peaks in the X-ray powder diffraction pattern are shown in the figure. Further, as the content of a certain crystal form in a product gradually decreases, some diffraction peaks ascribed to the crystal form in the powder X-ray diffraction pattern thereof may be reduced due to factors of the detection sensitivity of the instrument.
The term "characteristic diffraction peak" refers to a diffraction peak useful in representing the crystalline form in an X-ray powder diffraction pattern, which is related to the peak position, peak shape, and relative peak intensity of the diffraction peak, e.g., a small angle peak, sharp peak shape, and a diffraction peak having a relative peak intensity of at least 3% or more, or at least 5% or more, or at least 10% or more, or at least 20% or more, or at least 30% or more, or at least 40% or more, or at least 50% or more, or at least 60% or more, or at least 70% or more, or at least 75% or more.
The application has one or more of the following beneficial technical effects: (1) The compound 1 or the pharmaceutically acceptable salt thereof has good inhibitory activity (less than 100 nM) on a plurality of protein kinases; (2) The compound 1 or the pharmaceutically acceptable salt thereof has good inhibitory activity on human bile duct cancer cells and human intrahepatic bile duct cancer cells; (3) The compound 1 or the pharmaceutically acceptable salt thereof (for example, dihydrochloride of the compound 1) shows better tolerance in clinical phase I test, has fewer adverse reaction types and has low occurrence rate of more than grade 3 side reaction, which indicates that the clinical safety is better; (4) The compound 1 or a pharmaceutically acceptable salt thereof (e.g., dihydrochloride salt of compound 1) can control the disease progression of patients with advanced or metastatic biliary tract malignancy who fail or are intolerant of first-line treatment to a certain extent in clinical phase II test, and the effect thereof is superior to that of chemotherapy drugs commonly used in clinic. Has clinical potential for treating biliary tract cancer.
Drawings
Fig. 1: XRPD pattern of crystalline form I of dihydrochloride salt of compound 1 from preparative example 2;
fig. 2: XRPD pattern post long term stability test of form I of the dihydrochloride salt of compound 1 from preparation 2 in test example 3.
Detailed Description
The technical scheme of the application will be further described in detail below with reference to specific preparation examples and examples. The following preparations and examples are illustrative of the present application and should not be construed as limiting the scope of the application. Those skilled in the art will appreciate that various modifications and adaptations of the embodiments described above are possible and can be made without departing from the scope of the application. Unless otherwise indicated, the starting materials, reagents (e.g., organic solvents, electrodeless solvents, kinases, substrates, antibodies, buffers, reaction solutions, etc.), used in the following preparation examples and examples are all commercially available or may be prepared or formulated by known methods or reagent specifications.
In the following preparation examples, the analytical detection conditions were as follows:
1. content of
Detection instrument: agilent 1260 (LC 1260-3-DAD) high performance liquid chromatograph
Column: c18 4.6X105 mm,5 μm
Test conditions: a wavelength of 252nm; column temperature 45 ℃.
2. Solubility (Water, pH2.0 buffer)
Detection instrument: agilent 1260 high performance liquid chromatograph
Detection medium: purified water, pH2.0 phosphate-disodium hydrogen phosphate buffer
Preparing a reference substance solution: taking a proper amount of the compound of formula 1 as a reference substance, precisely weighing, adding a solvent to completely dissolve the compound, diluting the compound to prepare a solution of 100 mug/mL, precisely weighing 10uL, and measuring the content of the compound 1 in the reference substance solution by using HPLC.
3. X-ray powder diffraction (X-Ray Powder Diffraction, XRPD)
Detection instrument: bruker D2 PHASER powder X-ray diffractometer
Test conditions:
type of light pipe: a Cu target, a ceramic X-ray tube;
x-ray wavelength: the composition of cukα,1.5406;
voltage current: 30kV,10mA;
scanning range: 3-40 degrees 2 theta;
scanning total time: for 40min;
scanning speed: 0.5 seconds/step;
sample dosage: 3mg of
And (3) acquisition software: diffrac Plus XRD Commander
Analysis software: MDI Jade 6.0.
4. Differential scanning calorimeter-thermogravimetric analysis (Differential Scanning Calorimetry-Thermogravimetric Analysis, DSC-TGA)
Detection instrument: NETZSCH STA449F3 synchronous thermal analyzer
The testing method comprises the following steps: the sample (about 3 mg) was weighed and placed in an alumina crucible for testing, and the sample was heated from 20℃to 340℃at a heating rate of 10K/min under a condition of 20mL/min of dry nitrogen (shielding gas).
Instrument control software: NETZSCH-protein
Analysis software: protein Analysis.
5. Chlorides (CPS)
Detection instrument: dynamics-900 ion chromatograph
Column: dionex Ion Pac AS11-HC anion chromatographic column (specification: 4X 250 mm)
Experimental operation:
sample solution preparation: taking a proper amount of the test sample, precisely weighing, dissolving with a leaching solution (12.5 mmol/L sodium hydroxide solution) and quantitatively diluting to obtain a solution containing about 0.5mg of the test sample in each 1mL, and shaking uniformly to obtain the test sample solution.
Preparing a reference substance solution: proper amount of sodium chloride (corresponding to 18mg of chloride ions) is taken, precisely weighed, placed in a 250mL measuring flask, dissolved by leacheate, fixed in volume and shaken uniformly to serve as a reference substance solution.
Assay: precisely measuring 10 mu L of each of the reference solution and the sample solution, respectively injecting into an ion chromatograph, recording the chromatograms, and calculating the chloride ion content according to an external standard method by using the peak area.
6. Nuclear magnetic hydrogen spectrum
Instrument model: bruker Advance 600 type nuclear magnetic resonance spectrometer
Measurement conditions: with DMSO-d 6 As solvent, the test was performed at room temperature (-25 ℃).
7. Determination of solubility in biological menstruum
The formulation procedure of the biological vehicle media (SGF, feSSIF and FaSSIF) for the solubility determination is shown in table 1 below:
TABLE 1
The testing method comprises the following steps: and adding the sample to be tested into a biological solvent medium to prepare a solution or suspension with the target concentration of 10 mg/mL. The resulting solution or suspension was continuously shaken at 37℃at 200 rpm. The suspension was filtered at 0.5 hours and the concentration of compound in the filtrate was determined using HPLC.
Preparation example 1: preparation of Compound 1
Using the procedure in CN106660970B (example 22), compound 1 was prepared at 15g.
Preparation example 2: preparation of dihydrochloride salt of Compound 1
Compound 1 (10 g,21.58 mmol) obtained in preparation example 1 was weighed into a eggplant-type bottle, methanol solvent (110 mL) was added, the temperature was raised to 55±5 ℃, hydrochloric acid (3.7 mL,44.4 mmol) was added dropwise to the solution, stirring was carried out for 20 minutes, 200mL of ethyl acetate was slowly added, the temperature was lowered to 5±5 ℃, stirring was carried out for 2±1 hour, suction filtration was carried out, and the filter cake was washed with ethyl acetate (20 mL), thus obtaining dihydrochloride (11 g) of compound 1 in white color, yield 94.8%.
1 H-NMR(600MHz,DMSO-d 6 )δ:15.41(s,1H),11.78(s,1H),10.44(s,1H),8.80(s,1H),8.42(s,1H),7.78(dd,J=9.6Hz,J=2.4Hz,1H),7.58-7.52(m,2H),7.47(s,1H),4.24(t,J=6Hz,2H),4.03(s,3H),3.15-3.14(m,2H),2.76-2.75(m,6H),1.90-1.88(m,4H).
The chloride ion content was determined by ion chromatography and the stoichiometric ratio of the hydrochloride salt was calculated (see table 2 below) and it was concluded that the alkali/acid ratio of the hydrochloride salt was 1:2.
TABLE 2
The obtained hydrochloride sample was taken for X-ray powder diffraction, which showed good crystallinity, and was designated as form I of dihydrochloride, whose XRPD characterization pattern is shown in fig. 1, and the main diffraction peak data is shown in table 3. Samples were taken for DSC-TGA testing, with two endothermic peaks, endothermic peak 1: having an endothermic peak onset at 219.1 ℃ and peak around 231.0 ℃; endothermic peak 2: an endothermic peak at 235.1℃was initiated, a peak was reached around 284.2℃and decomposition occurred around 205 ℃. The PLM plot shows that the crystalline particles are in a regular morphology.
TABLE 3 XRPD diffraction peak data for form I of dihydrochloride obtained in preparation example 2
Test example 1: solubility test
The sample of preparation 2 was taken and subjected to solubility test in water and pH2.0 buffer, and the test results are shown in Table 4 below:
table 4 solubility results
Results: the sample of preparation 2 has high solubility in both water and pH2.0 buffer, and although the solubility of preparation 2 is reduced compared with the solubility in water under acidic conditions, the sample still has a solubility of >10mg/mL, which meets the preparation requirements.
According to the general requirement of the drug formulation on the solubility of the bulk drug, the solubility of the solid oral formulation in water is required to be more than 0.1g/L, and the solubility of the drug is required to be more than 10g/L for solution preparations such as injection or oral liquid, and more importantly, the solubility of the drug is required to meet the clinically required dosage concentration. Based on the solubility results of Table 4, the dihydrochloride of preparation example 2 can be considered for preparing solid oral dosage forms, and can be further considered for preparing solution preparations such as injections or oral liquids.
Test example 2: solubility test in biological menstruum
A sample (20 mg) of preparation 2 was weighed, and a different biological vehicle (2 mL) was added to conduct a solubility test, and the results are shown in Table 5.
Table 5 solubility of samples in different biological vehicles
Examples Salt Time FaSSIF(pH6.5) FeSSIF(pH5.0) SGF(pH1.2)
Preparation example 2 Dihydrochloride salt 0.5h Dissolving and clearing the liquid in the solvent,>10 5.07 dissolving and clearing the liquid in the solvent,>10
results: the sample obtained in the preparation example 2 can keep good solubility in different biological solvents and has high dissolution speed.
Test example 3: long term stability test
Taking a proper amount of salt sample obtained in preparation example 2, placing a polyethylene film seal for 5 months at 40+/-2 ℃ and 75+/-5% RH, and carrying out long-term test, wherein the result is as follows:
stability results and Crystal form detection results of the samples of Table 6
Table 7 XRPD data sheet of crystalline sample of preparative example 2 after long term stability testing
XRPD patterns of crystalline form I of the dihydrochloride salt of compound 1 from preparation example 2 after long term stability testing are shown in fig. 2.
Results: the dihydrochloride salt of the compound 1 obtained in the preparation example 2 can maintain chemical stability and crystal form stability, and accords with the storage regulations as a raw material medicine.
Test example 4: solid stability test
The dihydrochloride salt of preparation 2 was taken and placed under 40 ℃/75% RH (open) conditions for 7 days, respectively, and stability test was performed, and the results are shown in Table 8.
TABLE 8 stability results and Crystal form detection results
Results: the dihydrochloride of the compound 1 obtained in the preparation example 2 can maintain chemical stability and crystal form stability after being placed for 7 days under the condition of 40 ℃/75% RH (opening), so that the sample obtained in the preparation example 2 has better thermal stability and meets the requirement of being stored as a bulk drug.
Test example 5: mechanical stability test
The appropriate amount of dihydrochloride in preparation example 2 was taken and subjected to mechanical grinding for 5 minutes, and then subjected to X-ray powder diffraction, and the result shows that the crystal form was unchanged.
Example 1 in vitro enzymatic assay
1.1 test for inhibition of kinases such as RET, KDR, EGFR, FGFR1 and FLT-1 by Compounds
The test method comprises the following steps: enzyme substrate 20. Mu.g/mL Poly (Glu, tyr) was reacted using ELISA method 4:1 Coating an ELISA plate, adding enzyme, a sample, ATP (5 mu M) and the like for reaction, detecting substrate phosphorylation by using anti-phosphotyrosine monoclonal antibody (4G 10), and then adding horseradish peroxidase (HRP) -marked goat anti-mouse Ig G and tetramethyl benzidine (TMB) for color development to detect the substrate phosphorylation degree; simultaneously setting a control hole without tyrosine kinase and a control hole with corresponding DMSO concentration; 0.18 g M H is added 2 SO 4 The reaction was stopped at 50. Mu.L/well, read with microplate reader, colorimetric reaction, and Optical Density (OD) was observed 450nm Values.
Inhibition ratio = (negative control OD value-compound well OD value)/(negative control OD value-no enzyme control well OD value) ×100%
The relative inhibition of tyrosine kinase proteins by the compounds was determined.
Calculating half inhibition concentration IC according to each concentration inhibition rate by LOGIT method 50 . Each of the above experiments was repeated 3 times to determine the average IC of the 3 experiments 50 The value serves as a final indicator of inhibition. The test results are shown in Table 9.
Table 9 results of in vitro enzymatic assay of Compound 1 (IC 50 ,nM)
Compounds of formula (I) RET KDR EGFR FGFR1 FLT-1
Compound 1 <20 <20 <100 <20 <50
1.2 test of Compounds for inhibition of kinases such as FGFR2/3, LCK, ephA1 and the like
The test method comprises the following steps: 2. Mu.L of a 25 Xcompound solution was added to each well using the Caliper method, wherein 2. Mu.L of a reaction solution containing 25% DMSO was added to the enzyme-free control well and the negative control well; mu.L of 5 Xkinase solution was added to each well, and 10. Mu.L of reaction solution was added to the enzyme-free control well; centrifuging the detection plate at 1000rpm to mix uniformly; mixing 2.6XATP solution with 2.6Xsubstrate solution in equal volume to obtain 1.3XATP-substrate solution, and adding 38 μl ATP-substrate solution per well; centrifuging the detection plate at 1000rpm to mix uniformly; placing the detection plate at 30 ℃ for reaction for 1 hour; the reaction was stopped by adding 20. Mu.L of 35mM EDTA per well. Calipers read conversion.
Inhibition = (negative control well conversion-compound well conversion)/(negative control well conversion-no enzyme control well conversion) ×100%
Prism 5.0 IC 50 The values were fitted and the results are shown in Table 10.
Table 10 results of in vitro enzymatic assay of the sample of preparation example 2 (IC 50 ,nM)
Compounds of formula (I) FGFR2 FGFR3 LCK EphA1
PREPARATION EXAMPLE 2 sample <50 <50 <20 <100
1.3 test for inhibition of HER-2 kinase by Compounds
The test method comprises the following steps: using the ADP Glo method, 1. Mu.L of a 10 Xcompound solution was added to each well, and 1. Mu.L of a reaction solution containing 10% DMSO was added to each of the enzyme-free control well and the negative control well; mu.L of 2.5 Xkinase solution was added to each well, and 4. Mu.L of reaction solution was added to the enzyme-free control wells; centrifuging the detection plate at 1000rpm to mix uniformly; mixing the 4X ATP solution with the 4X substrate solution in equal volume to obtain 2X ATP-substrate solution, and adding 5 μl of 2X ATP-substrate solution into each well; centrifuging the detection plate at 1000rpm to mix uniformly; placing the detection plate at 30 ℃ for reaction for 1 hour; 10. Mu.LADP-Glo reagent was added to each well and left at 27℃for 40 minutes; mu.L of Detection reagent was added to each well and left at 27℃for 30 minutes; envision reads the luminescence signal.
Inhibition = (negative control well read-compound well read)/(negative control well read-no enzyme control well read) ×100%
Prism 5.0 IC 50 The values were fitted and the results are shown in Table 11.
TABLE 11 in vitro enzymatic assay results for the preparation of the sample of example 2 (IC 50 ,nM)
Compounds of formula (I) HER-2
PREPARATION EXAMPLE 2 sample <50
Conclusion: compound 1 and dihydrochloride (preparation 2) have good inhibition activity on RET, KDR, EGFR, FGFR1/2/3, FLT-1, LCK, HER-2, ephA1 and other targets. Similarly, other pharmaceutically acceptable salts of compound 1 have similar effects as above.
Example 2 in vitro cytological assays
Cell source: human bile duct cancer cell HuCCT1: purchased from commercial urban and north na-Chuan-xiong biotechnology limited; human hepatobiliary carcinoma cells RBE: purchased from Shanghai life sciences institute of China academy of sciences.
Cell culture conditions: the culture Medium is RPMI Medium 1640 Medium containing 10% FBS, and the culture environment is 37 ℃ and 5% CO 2
The inhibitory effect of dihydrochloride (preparation example 2) of compound 1 on proliferation of human bile duct cancer cells HuCCT1 and human intrahepatic bile duct cancer cells RBE was measured using the MTT method.
The test steps are as follows: cells in the logarithmic growth phase were seeded in 96-well plates (100. Mu.L/well) in a defined amount, and after 24h of adherence, 100. Mu.L of compound 1 with different concentration gradients was added to each well, 3 multiplex wells were set for each concentration, and corresponding blank wells (medium only) and normal wells (drug concentration 0) were set. After 72 hours of drug action, MTT working solution (5 mg/mL) was added, 20. Mu.L per well; acting for 4 hours at 37 ℃, removing supernatant by throwing a plate, and adding 150 mu L of DMSO; the microwells were mixed by shaking and the Optical Density (OD) was measured at 550nm with an ELISA reader.
The inhibition of cell growth was calculated using the following formula:
inhibition (%) = (OD value Normal hole -OD value Drug delivery hole ) /(OD value) Normal hole -OD value Blank hole )×100%
Based on the inhibition ratios of the concentrations, the half-maximal inhibition concentration IC of the drug was calculated using SPSS19.0 50
Table 12: inhibition of HuCCT1 and RBE cell proliferation (IC) 50 ,μM)
Cell species HuCCT1 RBE
Inhibitory Activity <2 <10
Conclusion: the dihydrochloride salt of compound 1 (preparation 2) has good inhibition effect on HuCCT1 and RBE cell proliferation. Similarly, other pharmaceutically acceptable salts of compound 1 have similar effects as above.
EXAMPLE 3 phase I clinical Studies
1.1 main study purposes
The safety and tolerability of the dihydrochloride salt of compound 1 (preparation 2) in patients with advanced solid tumors was examined, the maximum tolerated dose was determined, and a safe dose was recommended for the subsequent study phase.
1.2 research methods
1.2.1 case inclusion criteria
The case inclusion criteria used in this study are as follows:
(1) Inclusion criteria:
1) Voluntarily participating in clinical trial and signing informed consent;
2) Age 18-75 years (containing boundary values), unlimited in sex;
3) Patients with recurrent, unresectable, locally advanced or metastatic advanced solid tumors, as confirmed by histopathology or cytology, failed or intolerance to standard treatment, judged by researchers to be suitable for treatment with small molecule tyrosine kinase inhibitors;
4) The interval between last chemotherapy, radiotherapy, targeted therapy and surgery before the group is at least 4 weeks;
5) ECOG score: 0 to 1 minute;
6) Expected lifetime is at least 3 months;
7) Laboratory checks meet the following criteria:
neutrophil Count (ANC) 1.5X10-9/L
Platelet count (PLT) is not less than 100X 10-9/L
Hemoglobin (Hb). Gtoreq.90 g/L (no transfusion within 14 days prior to screening test);
an upper limit of 2.5 times normal for glutamic-pyruvic transaminase (ALT) and aspartate Aminotransferase (AST) (e.g., an upper limit of 5.0 times normal for patients with liver metastasis);
the total bilirubin is less than or equal to 1.5 times of the upper limit of the normal value;
serum creatinine is less than or equal to 1.5 times of the upper limit of the normal value;
8) Male and female child-bearing subjects agree to take effective contraceptive measures during treatment and 6 months after treatment is completed; female subjects are negative in serum or urine pregnancy tests within 7 days prior to study entry into the group and must be non-lactating subjects.
(2) Exclusion criteria
1) Other drug clinical trials were enrolled within 4 weeks prior to group entry and group receiving treatment;
2) Inability to swallow, chronic diarrhea, and ileus, there are a number of factors that affect drug administration and absorption;
3) Exclusion was performed in compliance with any one of the following criteria:
-QTc) value female not less than 470ms, male not less than 450ms; or congenital long QT syndrome, taking drugs known to prolong QT interval, family history of long QT syndrome
Resting the electrocardiogram shows any significant clinical abnormality in rhythm, conduction or morphology, requiring clinical intervention.
4) Urine routine prompts that urine protein is more than or equal to++ and 24 hours urine protein is more than 1.0 g;
5) According to the judgment of researchers, there are serious diseases which seriously jeopardize the safety of patients or affect the completion of the study of patients (such as poorly controlled hypertension (systolic pressure is more than or equal to 140mmHg or diastolic pressure is more than or equal to 90mmHg, although optimal drug treatment is performed), diabetes mellitus and the like);
6) Symptomatic metastatic brain or meningeal tumors within 4 weeks prior to entry into the study;
7) Adverse events (except hair loss and pigmentation) that did not return to grade 1 were treated last time when the group was entered;
8) Surgical major surgery was performed within 4 weeks prior to group entry or had not been fully recovered from any previous invasive procedure;
9) Coagulation dysfunction (INR >1.5 or Prothrombin Time (PT) > uln+4 seconds or APTT >1.5 ULN): those with a bleeding tendency (e.g., active peptic ulcers) or undergoing thrombolytic or anticoagulant therapy;
10 A researcher judges that the lung infection or pneumonia or interstitial pneumonia exists, and the lung infection or pneumonia or interstitial pneumonia is unsuitable for a researcher;
11 With active hepatitis B virus or hepatitis C virus infection
If HBsAg positive, add HBV DNA (measurement result is higher than upper limit of normal range of the study center);
If the detection result of the HCV antibody is positive, the HCV RNA is additionally detected (the detection result is higher than the upper limit of the normal range of the research center).
12 Human immunodeficiency virus infected (HIV positive) or suffering from other acquired, congenital immunodeficiency diseases or history of organ transplantation;
13 Other antineoplastic therapies (including various radiotherapy, chemotherapy, immunotherapy, targeted therapy, traditional Chinese medicine therapy, etc.) need to be combined;
14 A well-defined history of neurological or psychiatric disorders, including epilepsy or dementia;
15 A) the investigator considered unsuitable for the participating investigator.
16 An echocardiographic left ventricular ejection fraction of less than 50%.
(3) Drop/reject criteria:
1) Subjects withdraw informed consent, asking for withdrawal;
2) Judging the disease progress by researchers;
3) Subject still cannot tolerate toxicity after dose adjustment;
4) The subject had a gestational event in the study;
5) The investigator thought that withdrawal from the study was necessary.
1.2.2 safety evaluation criteria
Safety index: including vital signs and physical examinations, electrocardiogram and echocardiogram examinations, laboratory examinations, adverse events, and the like.
Maximum Tolerated Dose (MTD) evaluation: the previous dose resulting from the end of dose escalation due to DLT events.
1.2.3 methods of treatment
The study included subjects with advanced solid tumors aged 18-75 years. The dose ramp-up phase was performed starting from a 25mg dose and divided into 8 groups, the grouping being detailed in table 13. Each subject was subjected to single and multiple dosing tolerability and pharmacokinetic studies. 7 days after single administration, multiple administration periods were entered. Multiple doses were given every 28 days for a period of 7 days with 21 days of drug withdrawal until disease progression or intolerable toxic response occurred. Each subject received only one corresponding dose, and the trial started at a low dose and the next dose group was restarted after all subject tolerance observations in one dose group ended.
Table 13 dose escalation protocol
The maximum tolerated dose (or 800 mg) group obtained during the up-dosing phase was subjected to a multicentric, open-ended extension study to evaluate the efficacy, safety and pharmacokinetic profile of compound 1 dihydrochloride in patients with advanced solid tumors. Subjects who met the inclusion criteria entered the first week dosing period directly, followed by 21 days of continuous dosing, 7 days of rest, 28 days being one dosing period, until disease progression or intolerable toxic responses occurred.
1.3 results of the study
Up to 2020-12-6, a total of 32 subjects with advanced solid tumors were enrolled.
1.3.1 Security
The adverse reactions in the dosage group of 600mg and below are mostly of grade 1-2, and the most common adverse reactions are rash, diarrhea, nausea, vomiting, prolongation of the QT interval of an electrocardiogram and the like. Only at 800mg there were 2 Dose Limiting Toxicity (DLT) events, 1 with grade 3 appetite decline and 1 with grade 3 electrocardiogram QT interval prolongation.
1.4 comparison with like products
Referring to the results of clinical studies of several related protein kinase inhibitors disclosed in the prior art (see table 14 below), comparing the safety difference between the dihydrochloride salt of compound 1 of the present application and the existing protein kinase inhibitors, it can be found that the 3-level and above toxic reaction of dihydrochloride salt of compound 1 is less and has better safety in a wider administration dosage range.
Table 14 safety evaluation of related protein kinase inhibitors
Example 4 phase II clinical study
1.1 purpose of investigation
The dihydrochloride salt of compound 1 (preparation 2) was evaluated for clinical efficacy, safety and pharmacokinetic profile in subjects with advanced biliary malignancy.
1.2 research methods
1.2.1 Integrated study design
The study was started within 28 days (D-28 to D-1) before the test drug was taken after informed consent was obtained from the subjects themselves.
The initial administration dose is 600mg, and the medicine is orally taken once a day, and after the administration of the medicine on the first day, the medicine is continuously administered for 21 days, the rest is carried out for 7 days, and one administration period is 28 days until the disease progresses or intolerable toxic reaction occurs; and the related safety inspection and curative effect evaluation and PK blood sample collection are completed during the whole research period according to the flow requirements. If the analysis of the integrated data during the course of the study found that the subject was intolerant to the dose, the sponsor and the investigator post-discussed down-regulates a dose group as the recommended starting dose.
If toxic responses occur during treatment, the treatment is conducted on the basis of recommended dosing guidelines or by researchers based on their own clinical experience and updated study data during the course of the trial, and comprehensive assessment of the risk benefit of the subject and the current condition of the subject is made to determine how to conduct the treatment. The possible adjustments to the drug include down-dosing levels (450 mg (dose level 1) and 300mg (dose level 2), discontinuation of the drug and discontinuation of the treatment, the dosing frequency was still 21 days of continuous dosing, 7 days of rest, and 28 days as a cycle after adjustment of the dose level in the study, efficacy evaluations (comprehensive evaluation for target, non-target and new lesions) were performed every 2 cycles (i.e., 8 weeks) for data analysis.
1.2.2 selection of test population
The case inclusion criteria used in this study are as follows:
(1) Inclusion criteria:
1) Voluntarily participating in clinical trial and signing informed consent;
2) A histologically or cytologically confirmed, non-surgically resectable, locally advanced or metastatic biliary malignancy subject (including intrahepatic, extrahepatic cholangiocarcinoma, and gallbladder carcinoma), a subject who failed first line therapy (including progression of the disease within 6 months of adjuvant chemotherapy), or who was intolerant; and at least one measurable lesion is present (according to RECIST 1.1);
3) Age 18-75 years (containing boundary values);
4) ECOG score: 0 to 1 minute;
5) Blood routine tests were met (no transfusion in 14 days):
hemoglobin (HB) is more than or equal to 90g/L;
absolute value of neutrophil (ANC). Gtoreq.1.5X10) 9 /L;
Platelet count ≡75×10 9 /L;
6) The blood coagulation function test accords with: international Normalized Ratio (INR) < 1.5 or partially activated prothrombin time (APTT) < 1.5 XULN;
7) The biochemical examination accords with:
total Bilirubin (TBIL) is less than or equal to 1.5 XULN; alanine Aminotransferase (ALT) and aspartic acid Aminotransferase (AST) are less than or equal to 2.5 XULN; for subjects with tumor involving liver, it is acceptable to have TBIL less than or equal to 3 XULN, AST less than or equal to 5 XULN, ALT less than or equal to 5 XULN;
serum creatinine (Cr) less than or equal to 1.5 XULN;
Serum albumin not less than 28g/L;
8) Male and female child-bearing subjects agree to take effective contraceptive measures during treatment and 6 months after treatment is completed; female subjects of childbearing age are negative in serum or urine pregnancy tests within 7 days prior to study entry into the group and must be non-lactating subjects.
(2) Exclusion criteria
1) Ampulla cancer;
2) Other drug clinical trials were enrolled within 4 weeks prior to dosing and the group received treatment;
3) The interval first administration time at the end of the final antitumor treatment is required to satisfy the following time interval, otherwise, the following time interval is excluded:
those receiving any anti-tumor therapy (including but not limited to chemotherapy, radiation therapy, targeted therapy, etc.) for ≡4 weeks (those receiving palliative local radiation therapy for pain relief for ≡2 weeks);
a traditional Chinese medicine therapist who receives approved indications for cancer is not less than 2 weeks;
4) Urine protein is more than or equal to++ and the urine protein ration is more than 1g in 24 hours;
5) Other malignant tumors, except for already cured carcinoma in situ of the cervix, skin cancer other than melanoma, superficial bladder tumor and early esophageal and gastrointestinal malignant tumors limited to mucosal layers, which have been resected endoscopically, appear or are present in the meantime within 5 years;
6) Subjects who received any solid organ, bone marrow transplant, except for those that do not require immunosuppression (e.g., cornea transplant, hair transplant);
7) At the time of group entry, there was no more than 1 grade of toxicity (except for alopecia and pigmentation) due to any previous treatment and researchers judged that drug safety evaluation was affected;
8) The electrocardiographic examination is in accordance with any one of the following:
QT interval (QTc) value ∈450ms; or congenital long QT syndrome, a family history of long QT syndrome;
resting the electrocardiogram shows any significant clinical abnormality in rhythm, conduction or morphology, requiring clinical intervention;
9) Echocardiography examining left ventricular ejection fraction <50%;
10 At the discretion of the researcher, there are concomitant diseases that seriously jeopardize the safety of the subject, or affect the completion of the study by the subject, including but not limited to:
controlling poorly high blood pressure (systolic pressure. Gtoreq.150 mmHg or diastolic pressure. Gtoreq.100 mmHg after treatment);
cardiovascular/cerebrovascular diseases of clinical significance, such as: cerebral stroke (pre-dose <6 months), myocardial infarction (pre-dose <6 months), unstable angina, congestive heart failure (. Gtoreq.NYHA class II) or severe arrhythmia;
interstitial pneumonia requiring steroid treatment or severe infections requiring systemic treatment, judged by researchers to be unsuitable for study participation;
pericardial effusion, or pleural or peritoneal effusions that remain poorly controlled after 4 weeks of supportive treatment;
In the presence of abnormal thyroid function and in the case of treated thyroid function which is not maintained within normal limits, or in the case of untreated and symptomatic cases, with the exception of low T3/T4 syndrome;
11 High risk factors for gastrointestinal tract diseases such as gastrointestinal perforation, abdominal fistula, etc., such as: overactive peptic ulcers, inflammatory bowel disease (including ulcerative colitis and crohn's disease), abdominal fistulas, gastrointestinal perforations, intestinal obstruction, intraperitoneal abscesses, and the like occur within 28 days prior to administration;
12 Spinal cord metastasis, meningeal metastasis, and brain metastasis (except brain metastasis subjects who were asymptomatic prior to administration or were stable for 4 weeks or more after treatment prior to administration);
13 A history of liver disease is known (or not found in the past) and is in accordance with any one of:
HBsAg positive and HBV DNA>2000IU/mL (or>1×10 4 copy/mL);
HCV antibody positive and HCV RNA positive;
cirrhosis;
14 Human Immunodeficiency Virus (HIV) antibody positive;
15 Surgical procedures or not yet recovered from any previous invasive procedure within 4 weeks prior to administration, except for biliary stents, bile drainage, etc.;
16 A) the investigator considers unsuitable participation.
1.2.3 efficacy evaluation criteria
Reference is made to "standard for efficacy evaluation of solid tumors 1.1 edition (New Response Evaluation Criteria in Solid Tumors: revised RECIST Version 1.1.1). The method comprises the following steps:
[ Mitigation criteria ]
(1) Target lesion assessment
Complete Remission (CR): all target lesions disappear and all pathological lymph nodes (including target nodes and non-target nodes) short diameters must be reduced to < 10mm.
Partial Relief (PR): the sum of target lesion diameters is reduced by at least 30% from baseline levels.
Disease Progression (PD): the sum of diameters is increased by at least 20% relative to the minimum sum of diameters of all measured target lesions throughout the experimental study (baseline value if the baseline measurement is minimal); in addition, it must be satisfied that the absolute value of the sum of diameters increases by at least 5mm (the appearance of one or more new lesions is also considered as disease progression).
Disease Stabilization (SD): the target lesions did not decrease to PR nor did they increase to PD levels, which was intermediate between them, and the minimum of the sum of diameters was considered as a reference.
(2) Non-target lesion assessment
Complete Remission (CR): all non-target lesions disappeared and the tumor markers restored to normal levels. All lymph nodes were of non-pathological size (minor diameter < 10 mm).
Incomplete remission/non-disease progression: the presence of one or more non-target lesions and/or the continued presence of tumor marker levels exceeds normal levels.
Disease progression: well-defined progression of existing non-target lesions occurs. And (3) injection: the appearance of one or more new lesions is also considered as disease progression.
(3) New lesions
Lesions that have been detected in follow-up but not found in baseline examination will be considered new lesions and suggested disease progression. For example, a subject who has a visceral lesion found in a baseline examination, who has a metastatic lesion found when he is performing a CT or MRI craniocerebral examination, will be considered the basis for disease progression even though he is not performing a craniocerebral examination at the time of the baseline examination.
If a new lesion is ambiguous, such as due to its small morphology, further treatment and follow-up evaluations are required to confirm whether it is a new lesion. If the repeated examination confirms that it is a new lesion, the time for disease progression should be counted from the time of its initial discovery.
The assessment of FDG-PET lesions generally requires additional testing for additional confirmation, and it is reasonable to evaluate progression in combination with FDG-PET examination and additional CT examination results (especially for new suspected diseases). New lesions may be identified by FDG-PET examination, performed according to the following procedure: the baseline FDG-PET test results were negative and the following FDG-PET test was positive, indicating disease progression.
No baseline FDG-PET examination was performed, and subsequent FDG-PET examination results were positive: disease progression is demonstrated if the new lesions found by the follow-up FDG-PET positive examination match the CT examination.
If the positive examination result of the follow-up FDG-PET shows that the new lesion can not be confirmed by CT examination result, CT examination is needed to be carried out again for confirmation (if the positive examination result of the follow-up FDG-PET shows that the disease progress time is abnormal from the early FDG-PET examination).
If the positive examination result of the follow-up FDG-PET coincides with an existing lesion examined by CT, and the lesion does not progress in the imaging examination, the disease does not progress.
1.2.4 effectiveness and Security indicators
1.2.4.1 effectiveness index
The curative effect index comprises: objective Remission Rate (ORR), progression Free Survival (PFS), total survival (OS), disease Control Rate (DCR), duration of remission (DOR);
ORR: from the day of first dosing, including the percentage of total remission (CR) and Partial Remission (PR) subjects in all subjects in the group;
PFS: the time period between the date of the first administration and the date of tumor progression (PD assessed by imaging diagnosis) or the date of total death (priority over the preexisting);
OS: the time from the first administration day until death due to any cause (the blind subject was the last time to follow-up; the subjects still survived at the end of the study, the end of follow-up day;
DCR: defined as the percentage of subjects including CR, PR or disease Stabilization (SD) to all subjects in the group from the day of first administration;
DOR: tumors begin at a time when they first evaluate as CR or PR to a time when they first evaluate as PD or die of any cause.
1.2.4.2 Security assessment
Safety assessment includes assessment of vital signs and physical examinations, electrocardiography, laboratory examinations, adverse events, and the like.
1.2.5 pharmacokinetic assessment
PK blood samples were collected at any time point on day of each cycle for the detection of drug concentration of compound 1 for subjects receiving at least one drug treatment. Descriptive statistical analysis of drug concentration was performed and corresponding curves were drawn.
1.2.6 statistical treatments
1.2.6.1 efficacy analysis
Main endpoint index: PFS adopts Kaplan-Meier method to estimate median time and 95% CI, and draws corresponding survival curve.
Secondary efficacy index: OS, DOR estimate median time and 95% CI by Kaplan-Meier method, and draw corresponding survival curve. ORR, DCR, etc., given percentages and 95% ci calculated using the Clopper-Pearson method.
1.2.6.2 Security analysis
Calculating the incidence of adverse events (Treatment Emergent Adverse Event, TEAE)/drug-related TEAE, serious adverse events/reactions, TEAE/drug-related TEAE resulting in discontinuation of the drug or discontinuation of the treatment during the treatment;
1.2.6.3 pharmacokinetic analysis
Descriptive statistical analysis of drug concentration was performed and corresponding curves were drawn.
1.3 results of the study
The experimental object: the study included 16 subjects (including 12 cases of intrahepatic cholangiocarcinoma, 1 case of portal cholangiocarcinoma, 1 case of common bile duct carcinoma, and 2 cases of gallbladder carcinoma).
The results of the patient's efficacy assessment are shown in tables 15-1 and 15-2 below:
TABLE 15-1
Overall optimal efficacy Number of examples Incidence rate
Complete Remission (CR) 0 0.0%
Partial alleviation (PR) 1 6.25%
Disease Stabilization (SD) 6 37.5%
Disease Progression (PD) 9 56.25%
TABLE 15-2
Objective Remission Rate (ORR) 6.25%
Disease Control Rate (DCR) 43.75%
Analysis of results: from the above table data, it is shown that the dihydrochloride salt of compound 1 of the present application can control the disease progression to some extent in patients with advanced or metastatic biliary tract malignancy who failed or are intolerant of first-line therapy, particularly in patients with intrahepatic cholangiocarcinoma. Disease Progression Free Survival (PFS) and duration of remission (DOR) results are better, and still further follow-up of patients, PFS and DOR results will continue to be prolonged.
Typical cases:
the patients, 66 years old, 11 months in 2020, were right hepatectomy + cholecystectomy, post-operative pathology indicated intrahepatic cholangiocarcinoma (adenocarcinoma), stage T2NxM1 IV, chronic cholecystitis. Imaging examination of 28 days of 2021, 5, after surgery with single drug capecitabine adjuvant chemotherapy suggests disease progression and discontinuation. The 2021 month 6 replacement was gemcitabine+cisplatin chemotherapy, which was discontinued in the first treatment session due to intolerance of toxic response. The CT plain scan+enhanced examination at 9.3.2021 shows that the right half liver is missing after operation, several low-density nodules in the remaining liver, the larger ones are about 14mm in diameter, and considering metastasis, the stage is T2NxM1 (liver metastasis) stage IV and ECOG is 1 minute.
The product was administered 1 time daily (600 mg each time, orally, and continuously administered for 21 days and 7 days, i.e., 28 days as a cycle) from day 9 and 8 of 2021, and the first imaging effect was evaluated as SD for 8 weeks, and PR was evaluated for 13 weeks, 16 weeks and 24 weeks after treatment. Prompting: in cases where none of the clinically usual anticancer drugs (capecitabine, gemcitabine+cisplatin chemotherapy) show good therapeutic effects, the dihydrochloride salt of compound 1 of the present application shows a partial remission effect on the disease.
Based on the results of the above experiments, compound 1 and its pharmaceutically acceptable salt (e.g., dihydrochloride) are multi-target protein kinase inhibitors, which have good inhibitory activity against various protein kinases, and further, have inhibitory activity against several protein kinases related to biliary tract cancer at a level of 100nM or less, and will exhibit good therapeutic effects against biliary tract cancer caused by different genetic mutations, and compound 1 and its pharmaceutically acceptable salt (e.g., dihydrochloride) exhibit good antitumor activity in vitro tumor cell inhibitory activity evaluation experiments.
According to the results of the phase I clinical study of the compound, the pharmaceutically acceptable salt of the compound 1, represented by the dihydrochloride of the compound 1, has better safety.
According to the results of the phase ii clinical study of the compound, the pharmaceutically acceptable salt of compound 1, represented by the dihydrochloride salt of compound 1, is capable of controlling to some extent the disease progression in patients with advanced or metastatic biliary tract malignancy who fail or are intolerant of first-line therapy, particularly in patients with intrahepatic cholangiocarcinoma. The effect is better than that of the chemotherapy drugs commonly used in clinic, and has clinical potential for treating biliary tract cancer.
Thus, considering the preclinical in vitro test, the clinical trial situation of the same class of drugs, and the safety and preliminary efficacy results of phase I and the clinical study results of phase ii of compound 1 or a pharmaceutically acceptable salt thereof (e.g., dihydrochloride), compound 1 or a pharmaceutically acceptable salt thereof (e.g., dihydrochloride) of the present application has potential for clinical application in the treatment of biliary tract cancer.
The whole course and chinese names of abbreviations used in the present specification are as follows:
abbreviations and terms English holonomy scale Chinese paraphrasing
ALT Alanine aminotransferase Alanine aminotransferase
AST Aspartate aminotransferase Aspartate aminotransferase
AE Adverse event Adverse events
HB Haemoglobin Hemoglobin (hemoglobin)
ULN Upper Limit of Normal Upper limit of normal value
OS Overall survival Total life cycle
ORR Objective Response Rate Objective remission rate
PK Pharmacokinetic Pharmacokinetics of drugs
DLT Dose-Limiting Toxicit Dose limiting toxicity
MTD Maximum tolerated dose Maximum tolerated dose
CR Complete remission Complete alleviation of
PR Partial remission Partial mitigation
PD Progression disease Disease progression
QD Quaque die Once a day
SD Stable disease Disease stabilization
DCR Disease Control Rate Disease control rate
DOR Duration of Response Duration of remission
CT computer tomography Computed tomography
TEAE Treatment Emergent Adverse Event Adverse events during treatment

Claims (34)

1. The use of compound 1 or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment or prophylaxis of biliary tract cancer,
2. the use according to claim 1, wherein the biliary tract cancer is advanced biliary tract cancer.
3. The use according to claim 2, wherein the biliary tract cancer is locally advanced biliary tract cancer.
4. The use according to claim 2, wherein the biliary tract cancer is locally advanced biliary tract cancer which cannot be surgically resected.
5. The use according to claim 1, wherein the biliary tract cancer is metastatic biliary tract cancer.
6. The use according to any one of claims 1 to 5, wherein the biliary tract cancer is a biliary tract cancer that fails or is intolerant of treatment.
7. The use according to any one of claims 1 to 5, wherein the biliary tract cancer is biliary tract cancer that failed or was intolerant via first line therapy.
8. The use according to any one of claims 1 to 5, wherein the biliary tract cancer is gallbladder cancer or biliary tract cancer.
9. The use according to claim 8, wherein the cholangiocarcinoma is intrahepatic cholangiocarcinoma and/or extrahepatic cholangiocarcinoma.
10. The use according to claim 9, wherein the cholangiocarcinoma is intrahepatic cholangiocarcinoma.
11. The use according to any one of claims 1 to 5, wherein the pharmaceutically acceptable salt of compound 1 is a hydrochloride, sulfate, oxalate, maleate or malate salt.
12. The use according to claim 11, wherein the pharmaceutically acceptable salt of compound 1 is the dihydrochloride salt.
13. The use according to claim 12, characterized in that the dihydrochloride salt is in crystalline form, using Cu-ka radiation, the powder X-ray diffraction pattern expressed in terms of 2Θ angles has characteristic diffraction peaks at: 12.4 + -0.2 deg., 18.8 + -0.2 deg., 20.3 + -0.2 deg., 24.6 + -0.2 deg..
14. The use according to any one of claims 1 to 5, wherein said compound 1 or a pharmaceutically acceptable salt thereof is used as sole active ingredient for the preparation of said medicament.
15. The use according to any one of claims 1 to 5, wherein compound 1 or a pharmaceutically acceptable salt thereof is used in combination with one or more of the other therapeutic agents for the preparation of the medicament.
16. The use according to any one of claims 1 to 5, wherein the medicament is formulated as an oral formulation, an injectable formulation, a topical formulation or an external formulation.
17. The use according to claim 16, wherein the medicament is formulated for oral administration.
18. The use according to claim 17, wherein the medicament is formulated as tablets and capsules.
19. The use according to any one of claims 1 to 5, wherein the medicament comprises 10 to 500mg of compound 1 or a pharmaceutically acceptable salt thereof per unit of preparation.
20. The use according to any one of claims 1 to 5, wherein the medicament contains 10 to 500mg of the dihydrochloride salt of compound 1 per unit of preparation.
21. The use according to claim 20, wherein the medicament comprises 25mg, 50mg, 100mg, 150mg, 200mg or 250mg of the dihydrochloride salt of compound 1 per unit of preparation.
22. The use according to any one of claims 1 to 5, wherein the medicament comprises a therapeutically effective amount of compound 1 or a pharmaceutically acceptable salt thereof, in an amount of: 200mg to 800mg per administration, said dose being calculated as compound 1.
23. The use according to any one of claims 1 to 5, wherein the medicament comprises a therapeutically effective amount of the dihydrochloride salt of compound 1, which is administered in the following amounts: 200 mg-800 mg of the medicine is administrated each time; the doses were calculated as the dihydrochloride salt of compound 1 in anhydrous form.
24. The use according to any one of claims 1 to 5, wherein the medicament comprises a therapeutically effective amount of the dihydrochloride salt of compound 1, which is administered in the following amounts: 25mg, 50mg, 100mg, 200mg, 300mg, 350mg, 400mg, 450mg, 500mg, 550mg, 600mg, 650mg, 700mg, 750mg or 800mg per administration; the doses were calculated as the dihydrochloride salt of compound 1 in anhydrous form.
25. The use according to claim 23, wherein the medicament is administered once daily, twice daily, three times daily or four times daily at a daily dosing frequency.
26. The use according to any one of claims 1 to 5, wherein the medicament comprises a therapeutically effective amount of compound 1 or a pharmaceutically acceptable salt thereof, administered at a daily dose of from 100mg to 1000mg; the dose is calculated as compound 1, wherein the daily dosing frequency is one, two, three or more times.
27. The use according to any one of claims 1 to 5, wherein the medicament comprises a therapeutically effective amount of the dihydrochloride salt of compound 1, which is administered in a daily dose of 25mg to 900mg; the dose is calculated as the dihydrochloride salt of compound 1 in anhydrous form, wherein the daily dosing frequency is once, twice, three times or more.
28. The use according to claim 27, wherein the medicament comprises a therapeutically effective amount of the dihydrochloride salt of compound 1, which is administered daily in the following amounts: 25mg, 50mg, 100mg, 200mg, 300mg, 350mg, 400mg, 450mg, 500mg, 550mg, 600mg, 650mg, 700mg, 750mg, 800mg or 900mg; the dose is calculated as the dihydrochloride salt of compound 1 in anhydrous form, wherein the daily dosing frequency is once, twice, three times or more.
29. The use according to any one of claims 1 to 5, wherein the medicament comprises a therapeutically effective amount of compound 1 or a pharmaceutically acceptable salt thereof, in an amount and frequency of administration of: the administration is once daily, 200mg to 800mg per administration, the dose being calculated as compound 1.
30. The use according to any one of claims 1 to 5, wherein the medicament comprises a therapeutically effective amount of the dihydrochloride salt of compound 1, in an amount and frequency of administration of: the medicine is administrated once a day, and 200 mg-800 mg is administrated each time; the doses were calculated as the dihydrochloride salt of compound 1 in anhydrous form.
31. The use according to any one of claims 1 to 5, wherein the medicament comprises a therapeutically effective amount of the dihydrochloride salt of compound 1, in an amount and frequency of administration of: once daily, 300mg, 350mg, 400mg, 450mg, 500mg, 550mg, 600mg, 650mg, 700mg, 750mg, or 800mg per dose; the doses were calculated as the dihydrochloride salt of compound 1 in anhydrous form.
32. The use according to any one of claims 1 to 5, wherein the medicament is administered at a dosage frequency of: once daily for 21 days, and stopping for 7 days, wherein every 28 days is a period.
33. The use according to any one of claims 1 to 5, wherein the medicament is administered orally, by injection, topically or in vitro.
34. The application of the compound 1 or the pharmaceutically acceptable salt thereof in preparing a kit for treating or preventing biliary tract cancer,
CN202210178601.6A 2021-02-25 2022-02-25 Use of multi-target protein kinase inhibitors Active CN114948964B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021102092188 2021-02-25
CN202110209218 2021-02-25

Publications (2)

Publication Number Publication Date
CN114948964A CN114948964A (en) 2022-08-30
CN114948964B true CN114948964B (en) 2023-10-03

Family

ID=82976092

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210178601.6A Active CN114948964B (en) 2021-02-25 2022-02-25 Use of multi-target protein kinase inhibitors

Country Status (2)

Country Link
CN (1) CN114948964B (en)
WO (1) WO2022179608A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024131727A1 (en) * 2022-12-19 2024-06-27 正大天晴药业集团股份有限公司 Use of sultam compound in treating biliary tract cancer

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1211239A (en) * 1996-02-13 1999-03-17 曾尼卡有限公司 Quinazoline derivatives as VEGF inhibitors
CN1231662A (en) * 1996-09-25 1999-10-13 曾尼卡有限公司 Quinazoline derivatives and pharmaceutical compositions containing them
CN1391562A (en) * 1999-09-21 2003-01-15 阿斯特拉曾尼卡有限公司 Quinazoline derivatives as pharmaceuticals
CN105330653A (en) * 2014-08-11 2016-02-17 石药集团中奇制药技术(石家庄)有限公司 Quinazoline derivatives

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8658655B2 (en) * 2011-03-24 2014-02-25 Fox Chase Cancer Center Inhibition of activated cdc42-associated kinase 1

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1211239A (en) * 1996-02-13 1999-03-17 曾尼卡有限公司 Quinazoline derivatives as VEGF inhibitors
CN1231662A (en) * 1996-09-25 1999-10-13 曾尼卡有限公司 Quinazoline derivatives and pharmaceutical compositions containing them
CN1391562A (en) * 1999-09-21 2003-01-15 阿斯特拉曾尼卡有限公司 Quinazoline derivatives as pharmaceuticals
CN105330653A (en) * 2014-08-11 2016-02-17 石药集团中奇制药技术(石家庄)有限公司 Quinazoline derivatives

Also Published As

Publication number Publication date
WO2022179608A1 (en) 2022-09-01
CN114948964A (en) 2022-08-30

Similar Documents

Publication Publication Date Title
CN110092775B (en) Crystalline forms of a targeted CDK4/6 kinase inhibitor
CN105985323A (en) Novel epidermal growth factor receptor inhibitor and application thereof
CN112010839B (en) Crystalline forms of a targeted silk/threonine kinase inhibitor
CN110198716A (en) Treatment of cancer
CN106999484A (en) Treat the quinoline of non-small cell lung cancer
CN114641293A (en) Application of FGFR inhibitor
CN114948964B (en) Use of multi-target protein kinase inhibitors
CN115768416A (en) Tucatinib in combination with trastuzumab and oxaliplatin-based chemotherapy for HER2-positive cancers
CN109476634B (en) Crystal of salt of quinazoline derivative
KR102686264B1 (en) Chiauranib for the treatment of small cell lung cancer
WO2024097848A1 (en) Malonate and glycolate salts of an egfr inhibitor
CN112638385A (en) Quinoline derivatives for the treatment of brain tumors
AU2021350982B2 (en) Salt of arylaminoquinazoline-containing compound, and preparation method therefor and use thereof
CN115135326B (en) Combination pharmaceutical composition of compounds as c-Met kinase inhibitors and use thereof
JP7381115B2 (en) Compositions and their application in the preparation of medicines for cancer treatment
TWI535724B (en) New polymorphic forms of icotinib phosphate and uses thereof
RU2784869C1 (en) Chiauranib for treating small cell lung cancer
RU2827402C2 (en) Pharmaceutical composition containing pyridine compound and use thereof in ros1-positive non-small cell lung cancer
CN117177756A (en) Pharmaceutical composition of multi-target protein kinase inhibitor, application and preparation method thereof
CN115040519A (en) Use of EGFR inhibitors for the treatment of rare or non-classical mutations
Kasai et al. Tumor Lysis Syndrome in a Patient with BRAFV600E Mutated Colon Cancer Treated with Cetuximab and Encorafenib
US9505774B2 (en) Polymorphic forms of icotinib maleate and uses thereof
AU2023372570A1 (en) Malonate and glycolate salts of an egfr inhibitor
CN116063288A (en) Crystal form of succinate, preparation method and application thereof
CN104487443B (en) Crystal formation of Conmana maleate and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant