CN115190800A - Application of BRD4 inhibitor - Google Patents

Application of BRD4 inhibitor Download PDF

Info

Publication number
CN115190800A
CN115190800A CN202180017690.XA CN202180017690A CN115190800A CN 115190800 A CN115190800 A CN 115190800A CN 202180017690 A CN202180017690 A CN 202180017690A CN 115190800 A CN115190800 A CN 115190800A
Authority
CN
China
Prior art keywords
compound
tumor
solid tumor
pharmaceutically acceptable
acceptable salt
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.)
Pending
Application number
CN202180017690.XA
Other languages
Chinese (zh)
Inventor
姚雪坤
张宋安
张威
高天宁
申丽
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 CN115190800A publication Critical patent/CN115190800A/en
Pending legal-status Critical Current

Links

Images

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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • A61K31/55131,4-Benzodiazepines, e.g. diazepam or clozapine
    • A61K31/55171,4-Benzodiazepines, e.g. diazepam or clozapine condensed with five-membered rings having nitrogen as a ring hetero atom, e.g. imidazobenzodiazepines, triazolam
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Landscapes

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

Abstract

Use of a BDR4 inhibitor for the manufacture of a medicament for the treatment of a tumour, in particular an advanced solid tumour or a refractory solid tumour, in a regimen and in a safe and effective dose for the treatment of an advanced solid tumour or a refractory solid tumour. The BDR4 inhibitor can effectively treat advanced solid tumors or refractory solid tumors, has definite curative effects on advanced or refractory breast cancer, colorectal cancer, prostate cancer, gastrointestinal tumors and the like, and has better clinical safety.

Description

Application of BRD4 inhibitor
The present application claims the priority of the prior patent application with application number 202010156960.2 entitled "use of a BDR4 inhibitor" filed on the national intellectual property office of china on the day 03/09 in 2020. This prior application is incorporated by reference in its entirety.
Technical Field
The invention belongs to the field of medicines, and particularly relates to application of a BRD4 inhibitor in preparation of a medicine for treating tumors, particularly advanced solid tumors or refractory solid tumors, and a method for treating tumors by using the inhibitor.
Background
Neoplastic disease is one of the most common diseases in the modern world and is also a common cause of non-natural death. Tumors include both benign and malignant types, which are at high risk, and most malignant tumors are life-threatening. At present, the method for treating malignant tumor comprises surgical excision and drug chemotherapy, and the existing treatment means hardly has good curative effect on a plurality of patients with middle and late stage tumor including Triple Negative Breast Cancer (TNBC) and colorectal cancer (CRC).
According to the latest data, about 1800 thousands of new tumor cases are globally seen in 2018, wherein the breast cancer accounts for 11.6% and is located the second world; the number of cancer deaths is about 960 million, the breast cancer accounts for 6.6 percent, and the breast cancer occupies the sixth place of the world. In China, the incidence rate of breast cancer is 45.29/100000, and the number of the affected people is fifth nationwide. Meanwhile, the breast cancer becomes the first high-incidence malignant tumor type of women in China, about 1.6 million women die of the breast cancer every year in China, and the breast cancer becomes the 5 th malignant tumor death reason of women in China. Triple Negative Breast Cancer (TNBC) refers to breast cancer in which Estrogen Receptor (ER), progesterone Receptor (PR) and human epidermal growth factor receptor (HER-2) are all negative, accounting for approximately 15% of all breast cancer cases. Triple negative breast cancer has a high incidence in certain ethnic groups, such as asians and african-americans. TNBC patients are characterized by youthfulness, familiarity, invasiveness, relapse, metastasis and the like. Research shows that 87% of patients in TNBC have negative Androgen Receptor (AR) expression, so that the TNBC is easy to relapse and distant metastasis occurs, compared with other types of breast cancer, the TNBC has a significantly higher risk of metastasis within 5 years, and metastasis and spread mainly occur in internal organs, particularly in lung and brain. Both disease-free 5-year survival (DFS) and Overall Survival (OS) were also significantly lower than non-triple-negative breast cancer. The existing breast cancer targeted drugs are ineffective to triple-negative breast cancer, compared with other types of breast cancer, the triple-negative breast cancer has slightly higher sensitivity to chemotherapy, but the prognosis of conventional chemotherapy is poor, the 5-year survival rate is extremely low, particularly, the curative effect of patients with breast cancer susceptibility gene (BRCA) mutation is worse, and the field has very high unmet medical needs.
Colorectal cancer, also known as large bowel cancer, is a heterogeneous malignancy that grows in the first six feet of the large intestine (colon) and the last 8-10 inches of the large intestine (rectum), involving various molecular pathways and genetic/epigenetic changes that trigger the sequential transformation of normal mucosa into adenomas and then into carcinoma. It is a common malignant tumor in gastrointestinal tract, has unobvious early symptoms, and shows symptoms of defecation habit change, hematochezia, diarrhea and constipation alternation, local abdominal pain and the like along with the increase of the tumor; at the late stage, general symptoms such as anemia and weight loss appear. The onset age of colorectal cancer is 40-60 years old in China, the average onset age is 48.3 years old, the foot of western people is 10-15 years old, more common in China and young people than Europe and America, and not less common in colorectal cancer patients below 30 years old. Colorectal cancer is extremely dangerous, and because early detection is not timely, the colorectal cancer is in the middle and late stages when more than 80% of patients are actually found in China, and because the optimal treatment opportunity is lost, the survival rate of the patients 5 years after diagnosis is very low; every 5 minutes in our country 1 person dies of colorectal cancer. Colorectal cancer is currently treated mainly by surgery, but the quality of life is also significantly reduced after surgery, including: decreased sexual function, decreased excretory function, increased mental disability, decreased social activity, and the like. Domestic investigation shows that the postoperative survival rate of early colorectal cancer reaches over 90-95%, while the survival rate of later colorectal cancer is only 5%. With the increasing incidence rate of colorectal cancer year by year, and the surgical treatment has reached a bottleneck, it is difficult to make a major breakthrough, especially for patients with limited physical conditions who cannot be treated by surgical resection and patients with postoperative recurrence and metastasis, the drug therapy is very important.
Bromodomain and extra-terminal domain, the Bromodomain and the super-terminal domain family (BET), is a class of proteins with two bromodomains and one super-terminal domain that can recognize acetylated lysines in histones. There are 4 BET family members in humans, BRD2, BRD3, BRD4 and BRDT. Among them, BRD4 (Bromodemin-associating protein 4) is very widely expressed in human body, and BRD4 protein is combined with RNA polymerase II and forward transcription elongation factor to participate in the transcription process of MYC, BCL2, BCL6 and other oncogenes, and then regulates cell transcription, marks mitosis of mammals, regulates cell cycle, and plays an important role in the processes of cancer generation, development, fibrosis and inflammation. High expression BRD4 is detected in cancer cells and fibrotic cells of blood system and solid tumors such as leukemia, lymphoma, breast cancer, gastrointestinal tumors and the like. By targeted inhibition of BRD4, tumor cell apoptosis, i.e., proliferation slowing, can be induced, thereby achieving the anti-tumor effect. BRD4 has therefore received widespread attention and development in recent years as an extremely potential anti-tumor target. BRD4 inhibitors have become a popular area of drug research for diseases such as solid tumors, leukemia, and fibrosis.
At present, no drug is available on the market at home and abroad for the BRD4 inhibitor, but small-molecule drugs of a plurality of pharmaceutical companies are in clinical stage. Such as Birabresib (MK-8628, OTX-015) by Merck, molibresib (GSK-525762A, GSK-525762, I-BET-762, GSK' 762) under development by GlaxoSmithKline (GSK), AZ5153 by Aslicon (AZ) in research, and the like. Molibresib has been reported to be useful in the treatment of solid and hematological cancers, including multiple myeloma, non-hodgkin's lymphoma, acute myeloid leukemia, small cell lung cancer, castration resistant prostate cancer, colorectal cancer, non-small cell lung cancer, breast cancer, including metastatic breast cancer, estrogen receptor positive (ER +) breast cancer, and Triple Negative Breast Cancer (TNBC). Birabresib (MK 8628) is undergoing clinical studies involving Acute Leukemia (AL) and Other Hematologic Malignancies (OHM), triple negative breast cancer, non-small cell lung cancer, castration-resistant prostate cancer, and pancreatic ductal carcinoma.
Gastrointestinal toxicity is the prominent toxic response of BDR4 inhibitors in preclinical toxicology studies, but the response varies widely between species, with both rats and dogs being poorly tolerated and the human intestinal wall being insensitive to this. MK-8628 is reported to have strong gastrointestinal toxicity in rats, but is relatively well tolerated in humans during the clinical phase. AZ was similar to the BET inhibitor AZ5153 studied. Common gastrointestinal adverse reactions include diarrhea, constipation, nausea, vomiting, dysgeusia, mucositis and the like. The blood toxicity is also one of the main clinical adverse reactions of the BDR4 inhibitor, and researches show that the BET inhibitor can inhibit miR17-92, so that BIM protein is remarkably up-regulated, BAX and BAK protein are directly or indirectly activated, and normal hematopoietic cells are killed by controlling the internal mitochondrial apoptosis process. In clinical research, the compounds OTX015 and MK-8628 have the blood system adverse reactions such as thrombocytopenia, anemia, neutropenia and the like.
The compound A is a BRD4 small-molecule inhibitor, can obviously inhibit the activity of BRD4 protein, is expected to be used for treating tumors, and has the structure shown as the following formula (I):
Figure PCTCN2021079672-APPB-000001
WO2019056950A1 discloses a preparation method and activity of compound a, and WO2020192637A1 further discloses a crystal form and use of compound a. At present, the compound A has no sufficient evaluation data of efficacy and safety, so the important risk and the exact efficacy of the compound A for clinical application are not clear. The invention discusses the effectiveness and safety of the compound A in treating tumors and provides reference for clinical medication.
Disclosure of Invention
Based on the above-mentioned drawbacks of the prior art, the first aspect of the present invention is to provide a compound a or a pharmaceutically acceptable salt thereof for use in preparing a medicament for treating tumors, preferably advanced solid tumors or refractory solid tumors, wherein the compound a has the following structure represented by formula (i):
Figure PCTCN2021079672-APPB-000002
in some embodiments, the solid tumor in the advanced solid tumor or refractory solid tumor is a gastrointestinal tumor, colorectal cancer, breast cancer, prostate cancer, or the like.
In some embodiments, the solid tumor in the advanced solid tumor or refractory solid tumor is colorectal cancer, triple negative breast cancer, androgen-independent prostate cancer that is highly infiltrated by tumor-associated macrophages.
In some embodiments, the subject having the tumor is a human.
In some embodiments, the medicament contains a therapeutically effective amount of compound a or a pharmaceutically acceptable salt thereof. The therapeutically effective amount is preferably from about 0.001 to about 1000mg.
In some embodiments, the administration mode of compound a or a pharmaceutically acceptable salt thereof is not particularly limited, and a representative administration mode may be oral administration, injection administration, topical administration, or in vitro administration. Correspondingly, the medicine is prepared into clinically acceptable preparations, such as oral preparations, injection preparations, topical preparations, external preparations and the like by using conventional auxiliary materials and processes. The medicament is in a single dose dosage form or a multi-dose dosage form. The dosage form contains from about 0.001mg to about 1000mg of compound a or a pharmaceutically acceptable salt thereof (based on compound a), preferably from about 1mg to about 500mg, or from about 5mg to about 400mg, or from about 10mg to about 300mg, or from about 10mg to about 250mg, or from about 10mg to about 200mg, or from about 20mg to about 200mg, more preferably from about 10mg to about 160mg, or from about 20mg to about 160mg, or from about 80mg to about 160mg; for example, about 10mg, about 20mg, about 40mg, about 60mg, about 80mg, about 100mg, about 120mg, about 140mg, about 160mg, more preferably about 10mg, about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, still more preferably about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, or still more preferably about 10mg or about 40mg.
In some embodiments, compound a or a pharmaceutically acceptable salt thereof may be used in combination with one or more of other targeted or chemotherapeutic drugs. The other targeted drugs or chemotherapeutic drugs refer to targeted drugs or chemotherapeutic drugs clinically used for treating tumor-related diseases.
In a second aspect, the present invention provides a method of treating a tumour, which method comprises administering to a subject or patient a therapeutically effective amount of compound a or a pharmaceutically acceptable salt thereof, wherein the tumour is an advanced solid tumour or a refractory solid tumour.
In some embodiments, the solid tumor in the advanced solid tumor or refractory solid tumor is a gastrointestinal tumor, breast cancer, colorectal cancer, or prostate cancer.
In some embodiments, the solid tumor in the advanced solid tumor or refractory solid tumor is colorectal cancer, triple negative breast cancer, androgen-independent prostate cancer that is highly infiltrated by tumor-associated macrophages.
In some embodiments, the subject or patient is a human.
A suitable dosage range for Compound A or a pharmaceutically acceptable salt thereof is from about 0.001mg/kg to about 1000mg/kg per day; preferably, from about 0.01mg/kg to about 100mg/kg. Preferably, compound a or a pharmaceutically acceptable salt thereof is administered at a dose of about 0.001mg to about 1000mg per day. Administered in single or divided doses.
In some embodiments, the administering is oral administration, injection administration, topical administration, or in vitro administration; or the compound A or the pharmaceutically acceptable salt thereof is prepared into clinically acceptable preparations and then is administered, wherein the preparations comprise oral preparations, injection preparations, local administration preparations and external preparations.
In some embodiments, compound a or a pharmaceutically acceptable salt thereof (based on compound a) is administered at a dose of about 0.001mg to about 1000mg, preferably about 1mg to about 500mg, or about 5mg to about 400mg, or about 10mg to about 300mg, or about 10mg to about 250mg, or about 10mg to about 200mg, or about 20mg to about 200mg per day; more preferably from about 10mg to about 160mg, or from about 20mg to about 160mg, or from about 80mg to about 160mg; for example, about 10mg, about 20mg, about 40mg, about 60mg, about 80mg, about 100mg, about 120mg, about 140mg, about 160mg, more preferably about 10mg, about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, still more preferably about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, administered in a single dose or divided doses.
In some embodiments, the dosing regimen for compound a or a pharmaceutically acceptable salt thereof is 28 days per cycle, once daily for 21 days, and 7 days off; preferably, compound a or a pharmaceutically acceptable salt thereof (based on compound a) is administered at about 20mg, about 40mg, about 80mg, about 120mg, about 160mg per administration.
In some embodiments, the compound a or a pharmaceutically acceptable salt thereof is used in combination with one or more of other targeted or chemotherapeutic drugs.
In a third aspect, the present invention aims to provide compound a or a pharmaceutically acceptable salt thereof for use in the treatment of a tumor, preferably an advanced solid tumor or a refractory solid tumor.
In some embodiments, the solid tumor in the advanced solid tumor or refractory solid tumor is a gastrointestinal tumor, colorectal cancer, breast cancer, or prostate cancer.
In some embodiments, the solid tumor in the advanced solid tumor or refractory solid tumor is colorectal cancer, triple negative breast cancer, androgen-independent prostate cancer that is highly infiltrated by tumor-associated macrophages.
In some embodiments, the subject having the tumor is a human.
In some embodiments, the compound a or a pharmaceutically acceptable salt thereof is administered after being formulated into a clinically acceptable formulation, including oral, injectable, topical formulations.
In some embodiments, compound a or a pharmaceutically acceptable salt thereof (based on compound a) is administered at a dose of about 0.001mg to about 1000mg, preferably about 1mg to about 500mg, or about 5mg to about 400mg, or about 10mg to about 300mg, or about 10mg to about 250mg, or about 10mg to about 200mg, or about 20mg to about 200mg per day; more preferably from about 10mg to about 160mg, or from about 20mg to about 160mg, or from about 80mg to about 160mg; for example, about 10mg, about 20mg, about 40mg, about 60mg, about 80mg, about 100mg, about 120mg, about 140mg, about 160mg, more preferably about 10mg, about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, still more preferably about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, administered in a single dose or divided doses.
In some embodiments, the dosing regimen for compound a or a pharmaceutically acceptable salt thereof is 28 days per cycle, once daily for 21 days, and 7 days off; preferably, compound a or a pharmaceutically acceptable salt thereof (based on compound a) is administered at about 20mg, about 40mg, about 80mg, about 120mg, about 160mg per administration.
In some embodiments, the compound a or a pharmaceutically acceptable salt thereof may be used in combination with one or more of other targeted or chemotherapeutic drugs.
The dosage of the compound A or the pharmaceutically acceptable salt thereof is calculated by the compound A.
In order to evaluate the curative effect and safety of the compound A or the pharmaceutically acceptable salt thereof on treating the tumor, the inventor of the application develops BDR4 in-vitro activity inhibition test, in-vivo tumor inhibition test and clinical phase I research, and the result shows that the compound A has definite action mechanism, can obviously inhibit the activity of BRD4 protein, and has obvious effect of inhibiting the tumor growth in a mouse in-vivo efficacy model of colon cancer, prostate cancer and three-negative breast cancer. The clinical application is intended for the treatment of advanced solid tumors, including but not limited to gastrointestinal tumors, colorectal cancer, triple negative breast cancer, prostate cancer, and the like. Therefore, the compound A can effectively treat advanced solid tumors, particularly gastrointestinal tumors, three-yin breast cancer, colorectal cancer and prostate cancer, and has good clinical safety.
Interpretation of terms:
"advanced solid tumor" refers to a cancer that has spread by local invasion or metastasis outside the site or organ of origin. The term "advanced solid tumors" includes locally advanced and metastatic solid tumors.
"refractory solid tumor" refers to a solid tumor whose condition has progressed even though an anti-tumor therapy has been applied to the patient. The anti-tumor therapy includes radiotherapy, chemotherapy drug therapy, targeted drug therapy, immune checkpoint drug therapy and the like. In some embodiments, the refractory solid tumor is a solid tumor for which standard treatment regimens are ineffective.
Drawings
FIG. 1 is a flow chart of a clinical phase I protocol.
Detailed Description
The following examples are given for better illustration of the present invention, but the present invention is not limited to the examples. Those skilled in the art can make insubstantial modifications and adaptations to the embodiments described above while remaining within the scope of the invention.
1. Test for inhibition of BRD4 Activity in vitro by Compound A
Example 1: BRD4 Biochemical Activity assay
1. Preparation of the experiment:
1) The BRD4-BD1 and BRD4-BD2 proteins from BPS were used for the experiments; polypeptides from anaspc; detection reagents from PerkinElmer corporation;
2) Experiments compounds were screened using the experimental principle of TR-FRET.
3) Related control Compounds
2. The experimental steps are as follows:
1) Preparation of compound plates:
preparation of the compound plates in the experiments was achieved by an Echo non-contact nano-liter sonic pipetting system:
the compound dilution was done by Echo non-contact nanoliter sonic pipetting system with 3-fold decreasing dilutions to 10 concentrations 20000, 6666.67, 2222.22, 740.74, 246.91, 82.305, 27.435, 9.145, 3.048, 1.016nM.
2) Preparation of reaction reagents:
the relevant reagents should be prepared on the day of the experiment:
a) Preparing 1 × assay buffer (test buffer solution);
b) Prepare 3 × experimental component solution:
1. taking out the reagent and putting the reagent on ice to naturally melt for later use;
2. the "solution A" (protein solution), "solution B" (polypeptide solution), and "solution C" (detection reagent solution) used in the experiment were prepared using 1 × assay buffer, and the respective components were allowed to form a 3 × solution in the reaction system of the experiment, and the amounts of the solutions A, B, and C were enough to be required for the experiment.
3) The experimental operation steps are as follows:
experimental plates i.e. plates containing compound gradient concentrations and corresponding DMSO solutions prepared before the experiment using the Echo non-contact nano-upgrade sonic pipetting system:
a) Taking out the experimental plate, adding 5 mu L/hole of 'solution A' (protein solution) into the 2 nd to 23 th columns of the experimental plate, adding 5 ul/hole of 1 × assay buffer into the 1 st and 24 th columns of the experimental plate, and taking the columns 1 and 24 as Min control in the experimental system;
b) Centrifuging for 1000 revolutions for 30 seconds;
c) Incubate the plate at 23 ℃ for 20 minutes;
d) After 20 minutes of incubation, "solution B" (polypeptide solution) was added at 5 μ L/well to columns 1-24 of the assay plate;
e) Centrifuging for 1000 revolutions for 30 seconds;
f) Incubate the plate at 23 ℃ for 20 minutes;
g) After 20 minutes of incubation, 5 μ L/well of "solution C" (detection reagent solution) was added to columns 1-24 of the assay plate;
h) Centrifuging for 1000 revolutions for 30 seconds;
i) Incubate the plate at 23 ℃ for 40 minutes;
j) The plates were read on an EnVision.
4) And (3) data analysis:
a) Converting the Z 'value of each experimental plate by using corresponding Max control (maximum control) and Min control (minimum control) of each experimental plate, and ensuring that the Z' value of each plate is greater than 0.5;
b) Signal from control Compound IC was calculated by XLFIT5 50 And ensure that it remained within 3 times the average of the historical data, the results are shown in table 5.
TABLE 1 BRD4 detection IC 50 Test results
Figure PCTCN2021079672-APPB-000003
5) And (4) conclusion:
the compound A has obvious inhibition effect on BRD4-BD1 and BRD4-BD 2.
2. In vivo efficacy testing in mouse transplantable tumor model
Example 2: in vivo efficacy research of compound A in human breast cancer MDA-MB-231 \ luc cell subcutaneous xenograft tumor model
1. Experimental animals and group administration
1.1 Experimental animals
The species is as follows: mouse
Strain: BALB/c nude mice
Week age and body weight: 6-8 weeks old, 18-22 g of body weight
Sex: female with a female
The supplier: shanghai Sphall-Bikai laboratory animals Co., ltd
1.2 methods of formulating test substances
Table 2 test substance preparation method
Figure PCTCN2021079672-APPB-000004
Note: BID: twice daily.
1.3 grouping and dosing regimens
TABLE 3 in vivo efficacy test animal grouping and dosing regimen
Figure PCTCN2021079672-APPB-000005
Note: PO: the product is administered orally.
2. Experimental methods and procedures
2.1 cell culture
Human breast cancer MDA-MB-231. Sup. U luc cell in vitro monolayer culture with RPMI-1640 medium (supplier: gibco; cat # 22400-089; production lot # 4868546) supplemented with 10% fetal bovine serum, 100U/ml penicillin and 100. Mu.g/ml streptomycin at 37 ℃ 5% CO 2 And (5) culturing. Passage was performed twice a week with conventional digestion treatment with pancreatin-EDTA. When the cells are in exponential growth phase, the cells are harvested, counted and inoculated.
2.2 tumor cell inoculation
0.2mL of 10X 10 6 Individual MDA-MB-231 \ luc cells were inoculated subcutaneously into the right dorsal back of each nude mouse (PBS: matrigel = 1. The average tumor volume reaches 100-150mm 3 The grouped drug administration is started.
2.3 tumor measurement and Experimental indices
The experimental index is to investigate whether the tumor growth is inhibited, delayed or cured. Tumor diameters were measured twice weekly using a vernier caliper. The formula for tumor volume is: v =0.5a × b 2 And a and b represent the major and minor diameters of the tumor, respectively.
The tumor suppressor therapeutic effect of the compound was evaluated as TGI (%) or relative tumor proliferation rate T/C (%).
TGI (%), reflecting the rate of tumor growth inhibition. Calculation of TGI (%): TGI (%) = [ 1- (average tumor volume at the end of administration of a certain treatment group-average tumor volume at the start of administration of the treatment group))/(average tumor volume at the end of treatment of the solvent control group-average tumor volume at the start of treatment of the solvent control group) × 100%.
The relative tumor proliferation rate T/C (%) was calculated as follows: T/C% = T RTV /C RTV ×100%(T RTV : treatment group RTV; c RTV : negative control group RTV).
Calculating Relative Tumor Volume (RTV) according to the tumor measurement result, wherein the calculation formula is RTV = V t /V 0 In which V is 0 When administered in groups (i.e. d) 0 ) Measurement of the mean tumor volume, V t Mean tumor volume at a certain measurement, T RTV And C RTV The same day data was taken.
2.4 statistical analysis
Statistical analysis, including mean and Standard Error (SEM) of tumor volume for each time point for each group. Treatment groups showed the best treatment effect at day 21 after dosing at the end of the trial, and therefore statistical analysis was performed based on this data to assess differences between groups. Comparisons between two groups were analyzed using T-test, comparisons between three or more groups were analyzed using one-way ANOVA, and if F-values were significantly different, the measurements were performed using the Games-Howell method. If there is no significant difference in F value, analysis is performed by the Dunnet (2-sized) method. All data analyses were performed with SPSS 17.0. Significant differences were considered with p < 0.05.
3. Results of the experiment
After 21 days of administration, the tumor inhibition rate TGI =54.85% and T/C =52.99% for compound a, compared to vehicle control, p =0.200; the animal weight has no obvious change and has good tolerance.
Example 3: in vivo efficacy research of compound A in human prostate cancer PC-3 cell subcutaneous xenograft tumor model
1. Design of experiments
The test substances are prepared according to the method shown in the table 2, and the animal grouping and administration scheme are shown in the table 3.
2. Laboratory animal
The species are as follows: mouse
Strain: BALB/c nude mice
Week age and body weight: 6-8 weeks old, weight 18-22 g
Sex: male(s)
The supplier: shanghai Sphall-Bikai laboratory animals Co., ltd
3. Experimental methods and procedures
3.1 cell culture
Human prostate cancer PC-3 cells were cultured in vitro as monolayers under conditions of 10% fetal bovine serum, 100U/mL penicillin and 100. Mu.g/mL streptomycin in F-12K medium (supplier: gibco; cat # 21127-022; manufacturing lot # 1868870) at 37 ℃ 5% CO 2 And (5) culturing. Passage was performed twice a week with conventional digestion treatment with pancreatin-EDTA. When the cells are in the exponential growth phase, the cells are harvested, counted and seeded.
3.2 tumor cell inoculation
0.1mL of 10X 10 6 One PC-3 cell was inoculated subcutaneously in the right back of each nude mouse. The average tumor volume reaches 100-150mm 3 The grouped administration is started.
3.3 tumor measurements, experimental indices and statistical analysis were tested with the MDA-MB-231 model.
4. Results of the experiment
After 21 days of administration, the test compound a had a significant tumor suppression effect compared to the vehicle control group (T/C =44.63%, TGI =58.4%, p = 0.033); the animals had good tolerance.
Example 4: in-vivo anti-tumor efficacy research of compound A in MC38 mouse colon cancer cell animal transplantation tumor model
1. Design of experiments
The test formulations are prepared according to the method shown in Table 2, and the animal groups and dosing schedules are shown in Table 4 below.
TABLE 4 in vivo efficacy test animal grouping and dosing regimen
Figure PCTCN2021079672-APPB-000006
2. Laboratory animal
The species are as follows: mouse
Strain: c57BL6 mice
Week age and body weight: 6-7 weeks old, weight 16-20 g
Sex: female
The supplier: shanghai Si Laike laboratory animals Limited liability company
3. Experimental methods and procedures
3.1 cell culture
Mouse colon cancer MC38 cells (and Biotechnology Co., ltd.) were cultured in vitro in a monolayer in DMEM medium (Gibco, cat # 12100) containing 10% fetal bovine serum, at 37 ℃ with 5% CO 2 The incubator of (2). Passage was performed with conventional digestion treatment with 0.25% pancreatin-EDTA. When the cells are in the exponential growth phase and the saturation is 80% -90%, collecting the cells, counting and inoculating.
3.2 tumor cell inoculation
0.1mL of 2X 10 5 The right dorsal aspect of each mouse was inoculated subcutaneously with individual MC38 cells until the mean tumor volume reached about 70mm 3 At times, random group dosing was performed according to tumor volume.
3.3 tumor measurements, experimental indices and statistical analysis were tested with the MDA-MB-231 model.
4. Conclusion of the experiment
After 20 days of administration, the 15mg/kg administration group of compound a had a relative tumor proliferation rate T/C =33.68%, tumor growth inhibition rate TGI =68.81%, and p <0.0001, compared to the vehicle control group; the relative tumor proliferation rate T/C =27.59%, TGI =75.21%, p <0.0001 in the 25mg/kg administration group; T/C =10.04%, TGI =93.46%, p < 0.0001% in the 50mg/kg dose group. The animals in each administration group have obvious tumor inhibiting effect and good tolerance.
3. Toxicology study
Example 5 Single dose toxicity test in rats
40 SD rats, female and male half, were divided into 2 groups of 20, 10/sex/group, vehicle control and Compound A. The single administration dose of the compound A group is 100mg/kg, the single administration volume is 10mL/kg, the administration concentration is 10mg/mL (maximum gavage concentration), the administration is carried out 2 times within 24h (8-hour interval), and the total administration dose is 200mg/kg. Vehicle control group was given an equal volume of vehicle.
During the test period, the animals in the compound A administration group did not show severe poisoning symptoms, and no abnormality was found in feeding, blood index, blood coagulation index and gross anatomical observation. The compound A of 200mg/kg may have reversible toxic effect on the digestive system of SD rats, and can cause the animals to salivate, urinate and defecate and the weight of females to slightly increase. The level of UREA in the serum of the male animals in the group to which the compound A is administered is reduced. Thus, under the conditions of this experiment, the Maximum Tolerated Dose (MTD) of compound A given by a single oral gavage in SD rats was 200mg/kg.
EXAMPLE 6 Single dose toxicity test in dogs
8 Beagle dogs, female and male, are divided into 4 groups according to the body weight, 2 dogs each group and female and male. The groups are as follows: vehicle control group, compound a low dose group, compound a medium dose group, and compound a high dose group. The single administration dose of the compound low, medium and high dose groups is 6mg/kg, 18mg/kg and 48mg/kg respectively, the single administration dose is 2 times (8-hour interval) within 24h, and the total administration dose is 12mg/kg, 36mg/kg and 96mg/kg respectively. Vehicle control group was given an equal volume of vehicle.
During the test period, animals in each administration group of the compound A do not have serious toxic symptoms, only a water sample is passed after the second administration on the administration day, and the food consumption of the animals in the high-dose group tends to be reduced 4-5 days after the administration. Compound a dose groups Beagle dogs showed a trend towards an increase in blood indicators WBC and # NEUT after dosing. Thus, under the conditions of this test, a single administration of Compound A gave a non-toxic response dose (NOAEL) to Beagle dogs of 36mg/kg; MTD was 96mg/kg.
Example 7 repeated administration of SD rats for 4 weeks recovery 4 weeks with TK toxicity test study
SD rats were divided into 4 groups of 46/group (30/group for main trial, 16/group for TK satellite), male and female halves. The groups are a solvent control group, a compound A low-dose group, a compound A medium-dose group and a compound A high-dose group. Compound A Low, medium, and high dose groups SD rats were orally gavaged 2,6, and 20mg/kg of Compound A (total dose 4, 12, 40 mg/kg. Day, respectively) 2 times daily (8 hours apart) for 4 weeks, and were discontinued for 4 weeks. Vehicle control group was given an equal volume of vehicle.
The results show that: during the administration period, animals of each group including the solvent control group have intermittent loose stool, the SD rats of the compound A medium-dose group and the high-dose group have intermittent salivation, and the individual SD rats of the high-dose group also have perianal filthy, spontaneous hypoactivity, sallowness, abnormal urine color and emaciation. The male animals in the high-dose group have reduced food intake and slow weight increase, and can recover after stopping taking the medicine. Clinical pathological examination indicates that the test sample has reversible toxic effects on blood system and blood coagulation system at high dose, specifically as rising NEUT and RETIC, lowering LYMPH and/or PLT, prolonging PT, and shortening TT. Histopathological examination finds that 12-40 mg/kg of the compound A has reversible toxic effect on the digestive system, the immune system, the respiratory system and the skeletal system of male and female SD rats, and is represented by rectal mucosa goblet cell reduction, spleen and bone marrow lymphocyte reduction, thymus weight/organ coefficient reduction, lung foam macrophage aggregation and femur and knee joint trabecula increase. Compound A at 40mg/kg has reversible toxic effects on the heart of male SD rats, manifested by monocyte infiltration or myocardial necrosis.
TK results show that after D1 administration, the AUC0-24h in the dose group of the compound A has obvious sex difference, and after D1 and D28 administration, the systemic exposure (AUC 0-24h and Cmax) of the rest dose groups has no obvious sex difference; the systemic exposure to compound A in females (AUC 0-24h and Cmax) increased dose-proportionally following administration of D1 and D28; the AUC0-24h increase rate of the male compound A is higher than the dose increase, and the Cmax is increased in dose proportion; after 2 times daily administration for 4 weeks, no accumulation of compound a was observed in plasma of both male and female SD rats.
Therefore, under the test conditions, the MTD was 40mg/kg/day. AUC0-24h at day 28 in male and female animals at this dose were 23115 and 18017h × nm, respectively. The NOAEL was 4mg/kg/day.
Example 8 repeated dosing of beagle dogs for 28 days of recovery with TK toxicity test study
Beagle dogs were randomly divided into 4 groups, 10 dogs/group, and male and female halves. The groups are vehicle control group, compound A low dose group, compound A medium dose group and compound A high dose group. Compound A Low, medium, and high dose groups were orally gavaged 2 times daily (8 hours apart) with 1,3 and 6mg/kg of Compound A (2 mg/kg, 6mg/kg, and 12mg/kg total daily dose, respectively) for 4 weeks, and discontinued for 4 weeks.
The results show that: during the administration, 3/10 cases (1 female and 2 male) of animals in the 12mg/kg compound A dose group are dying, and the death reason is related to the toxicity of the digestive system, the blood system, the immune system and the like of the test products. After administration of each dose of the compound A, the compound A has toxic effect on the digestive system of animals, and the toxic effect is manifested by vomit, abnormal stool, reduced appetite, slow weight increase or reduction of the animals after administration, and the toxic reaction of the animals in medium and high dose groups is obvious. High doses of compound a may also actuate hypomotility, wasting, tremor, gait abnormalities, ocular or nasal secretion abnormalities. High and/or medium doses of compound a affect Beagle dog hematological indicators (RETIC, EOS, BASO decrease), coagulation indicators (APTT prolongation, FIB increase) and blood biochemical indicators (TP, ALB, CREA, UREA decrease, ALT, AST, CHOL, TG, GLU increase), but the effect is reversible. Histopathological examination revealed that 2mg/kg of compound a had a reversible toxic effect on the Beagle canine immune system (thymus). 6-12 mg/kg of compound A has reversible toxic effect on the digestive system (pancreas, liver and digestive tract), immune system (thymus/spleen and intestinal tract-related lymph nodes), blood system (sternum bone marrow) and skin of the Beagle dog, the toxic effect on the reproductive system (testis and epididymis) of the Beagle dog is not completely recovered after the drug is stopped, and spleen extramedullary hematopoiesis can be seen in the recovery animals. 12mg/kg of Compound A has a reversible toxic effect on the Beagle dog respiratory system (trachea, lungs).
The TK results show that there was no significant sex difference in compound A systemic exposure (AUC 0-24h and Cmax) for each dose group after D1 and D28 administration; compound a exposure (AUC 0-24h and Cmax) did not significantly accumulate in each dose group after repeated dosing; the AUC0-24h increase for compound a was higher than the dose increase after D1 and D28 administration, and the Cmax increased dose proportionally.
Thus, under the conditions of this test, the lethal dose was 12mg/kg/day and HNSTD (the highest non-severely toxic dose) was 6mg/kg/day. At this dose, the dose was 9443.7 and 9149.9hnM on day 28 (AUC 0-24 h) in the male and female animals, respectively. The lowest dose at which adverse effects were observed (LOAEL) was 2mg/kg/day.
4. Clinical research
EXAMPLE 9 phase I clinical trial study
1. Scheme design
The trial is a phase i clinical study open, dose escalation and cohort expansion for patients with advanced solid tumors aimed at evaluating the safety, tolerability, pharmacokinetic profile and primary antitumor activity of compound a in patients with advanced solid tumors. The endpoint indicators include:
(1) Safety is as follows: AE. SAE, DLT occurrence and frequency; the Maximum Tolerated Dose (MTD) (if any), the phase II clinically recommended dose (RP 2D) and the dosing regimen for compound a were determined.
(2) Pharmacokinetic indexes are as follows: including but not limited to AUC 0-last 、AUC 0-∞ 、C max 、T max 、t 1/2 And CL/F and the like.
(3) The curative effect index is as follows: overall Remission Rate (ORR), progression Free Survival (PFS), disease Control Rate (DCR), duration of remission (DOR), and the like.
The study was conducted in two phases, the first (Stage I) being a Dose escalation (Dose evolution) and Dose expansion (Dose expansion) study and the second (Stage II) being a Cohort expansion (Extension cowart) study.
Stage one (Stage I):
dose escalation study:
referring to HNSTD/MTD dosage and in vivo efficacy test dosage in prophase toxicology test, designing clinical research dosage. The initial dose of compound a was set at 20 mg/day, and 5 dose groups were initially planned: 20. 40, 80, 120, 160mg. The preset highest ascending dose was 160 mg/day. The study will follow an "i3+3" dose escalation protocol.
Each dose level was first cycle (31 days), and subjects entered a continuous dosing phase on day 4 after a single dose on day 1 (day 1), with no DLT and no serious adverse events observed for 3 days. (the specific frequency of administration will be adjusted to the patient's tolerance, safety and PK parameters for the drug).
After completion of the first Cycle (Cycle 1) of administration, subjects were evaluated for safety, tolerability, pharmacokinetic profile, and antitumor activity. If the subjects were well tolerated and i agree, then the subsequent Cycle (Cycle 2-Cycle N) will continue to be administered every 28-day repeat Cycle until disease progression or intolerable toxicity occurs, or other causes terminate the study.
Dose amplification study:
according to the data such as safety, tolerance and effectiveness obtained by the dose-escalation study, a target dose group dose-expansion study and an exploration study of different dosing schedules (such as 21 days of administration and 7 days of withdrawal) can be carried out if necessary. The safety assessments will continue to be performed by the sponsor and investigator to determine the dosage level and dosing regimen to be administered during dose escalation and dose escalation based on the available data for past dosage levels.
Second Stage (Stage II):
after determination of the MTD, preliminary prediction of RP2D and appropriate dosing regimen in the first phase, cohort expansion studies will be performed in different indications to observe the safety, tolerability, pharmacokinetic profile and antitumor activity of the study drug.
The expansion phase may select populations of different tumor species, each of which is expected to account for 20-40 subjects in the group. The final cohort expanded sample size and expanded tumor size will be determined after full discussion with the investigator, taking into account the first stage and preclinical study data. Also, sponsors and researchers may consider grouping that prematurely ends a cohort on a patient-based basis of maximum benefit/risk ratio.
The above two stage I clinical experiments are planned to be included in 53-186 cases of malignant advanced solid tumor patients.
2. Inclusion and exclusion criteria
1) Inclusion criteria
The subjects eligible for the study must meet the following inclusion criteria:
1. the age is 18-75 years (including) without limitation.
2. The subject must have histologically or cytologically confirmed advanced or metastatic tumors and either no standard treatment regimen, or no efficacy or intolerance to standard treatment regimens, or be unconditionally treated and meet the tumor species requirements for the corresponding stage:
(1) Stage I: solid tumor species are not limited;
(2)Stage II:
①Arm 1:TNBC
②Arm 2:CRC
(3) arm 3: other possible tumor species
3. There is at least one measurable lesion, and the measurable definition is derived from RECIST version 1.1 criteria.
Ecog performance status score: 0 to 1 minute.
5. Survival time was expected to exceed 3 months.
6. Major organ function was within 7 days prior to treatment, meeting the following criteria (no blood transfusion, EPO, G-CSF or other medical support treatment received within 14 days prior to the first dose of study drug):
Figure PCTCN2021079672-APPB-000007
7. women should agree that contraceptive measures (e.g. intrauterine device [ IUD ], contraceptive or condom) must be taken during the study and within 6 months after the study; serum pregnancy test negative within 7 days prior to study enrollment and must be a non-lactating subject; the male should agree on subjects who must take contraceptive measures during the study and within 6 months after the end of the study period.
8. Subjects had informed consent to the study prior to the trial and voluntarily signed a written informed consent form.
2) Exclusion criteria
Subjects were excluded from the study without selection for any of the following:
1. chemotherapy, radiotherapy, biotherapy, endocrine therapy, targeted therapy, immunotherapy and other anti-tumor treatments are received within 4 weeks before the study drug is used for the first time, except for the following items: nitrosoureas (such as carmustine, lomustine, etc.) or mitomycin C are present within 6 weeks prior to the first use of the study drug; the oral administration of the fluorouracil and small molecule targeted drugs is carried out 2 weeks before the study drug is used for the first time or within 5 half-lives (based on long time) of the known drugs; the traditional Chinese medicine with anti-tumor indications is used within 2 weeks before the research medicine is used for the first time.
2. Other non-marketed clinical study medications were received within 4 weeks prior to the first use of the study medication.
3. Major organ surgery (not including needle biopsy) or significant trauma occurred within 4 weeks prior to the first use of study drug.
4. Systemic glucocorticoid (prednisone > 10 mg/day or equivalent doses of the same drug) or other immunosuppressive therapy is received within 14 days prior to the first study drug use; except for the following: treatment with topical, ocular, intra-articular, intranasal, and inhaled glucocorticoids; prophylactic treatment with glucocorticoids is carried out for a short period of time (e.g. to prevent contrast agent allergy).
5. Those who have used potent inhibitors or potent inducers of CYP3A4 within 14 days prior to the first use of the study drug.
6. Have previously been treated with BET inhibitors.
7. Adverse reactions of the previous antitumor therapy do not return to CTCAE grade 5.0 evaluation which is less than or equal to grade 1 (except toxicity which is judged by researchers such as alopecia and the like to have no safety risk).
8. Central nervous system metastasis or meningeal metastasis with clinical symptoms, or other evidence that central nervous system metastasis or meningeal metastasis in patients has not been controlled, is judged by researchers to be unsuitable for grouping.
9. Uncontrolled active infections (acute or chronic fungal, bacterial, viral or other infections).
10. There is a history of autoimmune disease, immunodeficiency disease, including positive HIV detection, or other acquired, congenital immunodeficiency disease, or a history of organ transplantation.
11. Active hepatitis b (hepatitis b virus titer > 1000 copies/ml or 200 IU/ml), allowing prophylactic antiviral treatment in addition to interferon; positive for hepatitis c antibody.
12. There is a history of serious cardiovascular disease, including but not limited to:
(1) Serious heart rhythm or conduction disorder, such as ventricular arrhythmia requiring clinical intervention, II-III degree atrioventricular block, etc., or other arrhythmia requiring administration of anti-arrhythmic drugs (except anticoagulant drugs, the blood coagulation function associated with administration of anticoagulant drugs must meet the selection criteria);
(2) The history of myocardial infarction, angina, angioplasty and coronary bridging surgery;
(3) QT/QTc interval prolongation of electrocardiogram at baseline (QTcF >480 ms);
(4) The baseline Echocardiography (ECHO) or multi-gate acquisition (MUGA) technology shows that the Left Ventricular Ejection Fraction (LVEF) is less than or equal to 50 percent;
(5) Heart failure, new york cardiology society (NYHA) graded class II and above;
(6) Poorly controlled hypertension (systolic pressure ≧ 150mmHg and/or diastolic pressure ≧ 95mmHg despite optimal treatment);
(7) Formerly or currently suffering from cardiomyopathy.
13. There is no way to swallow the drug orally, or there are conditions that are judged by researchers to severely affect gastrointestinal absorption.
14. There is a history of other serious systemic diseases that researchers judge are not appropriate for patients to participate in clinical trials.
15. Alcohol or drug dependence is known.
16. There has been a well-established history of neurological or psychiatric disorders, including epilepsy or dementia.
17. Pregnant or lactating women.
18. The investigator considered the subject to be unsuited to participate in the clinical study for other reasons.
3. Results of the study
At present, in the clinical test, the number of the tested subjects is small, the treatment period is short, and accurate estimation on the final result and the later-stage scheme is not enough. The Maximum Tolerated Dose (MTD) of the compound A is estimated to be 120 mg/d-160 mg/d and the expected curative dose is 80mg/d by comprehensively considering the tolerated dose, the non-clinical pharmacodynamic dose and the phase I clinical dose of the similar compound. Under test conditions, the compound A is administered at 80mg/d, 120mg/d and 160mg/d, and has effective therapeutic effect on patients with advanced solid tumors, especially gastrointestinal tumors, triple negative breast cancer, colorectal cancer and prostate cancer. In the study, if the dose is increased to 160mg/d, the group is still good in safety and tolerance, so that the researchers and the sponsor can discuss the decision whether to explore higher dose. The determination of higher doses will be based on available data for existing dose levels and cannot be accurately predicted at present.
It is expected that compound a will be safe and effective in the treatment of advanced solid tumors, especially gastrointestinal tumors, triple negative breast cancer, colorectal cancer, prostate cancer.
EXAMPLE 10 typical cases
A patient is male, is 62 years old, and is diagnosed in the rectal cancer IIIB stage in 2015, and is subjected to pre-excision radical operation of the rectal cancer under a laparoscope, postoperative auxiliary oxaliplatin +5-Fu and postoperative auxiliary pelvic radiotherapy; then, the treatment is carried out by oxaliplatin + capecitabine, FOLFIRI (irinotecan +5-Fu + calcium folinate), ximinokang + thalidomide, capecitabine single drug, recombinant anti-EGFR human-mouse chimeric monoclonal antibody injection and regorafenib; in 2017, two radiotherapy treatments were changed for left lung disease. The disease progress occurs and the treatment fails. Diagnosis at patient enrollment: stage IVA of rectal cancer.
The patient was given compound a orally at a dose of 20mg for the first time at 2020-5-13. Observing that no serious abnormality exists in two days, continuously taking the medicine at 2020-5-15 with the dose of 20 mg/day and the last medicine at 2020-7-10 for 2 cycles (28 days in each cycle). An enhanced CT examination was performed at the end of the second week and evaluated as SD (disease stability). Grade 1 hypoalbuminemia, grade 1 anemia, and a reduction in grade 1 white blood cell count occurred during dosing. Hypoproteinemia may not be associated with the study drug, the latter two may be associated with the study drug.
The full name and Chinese definition of English abbreviation in the specification are as follows:
abbreviations and terms English full scale Chinese explanation
AE Adverse event Adverse events
AUC Area under drug time curve Area under the curve of time of administration
AL Acute leukemia Acute leukemia
ALT Alanine aminotransferase Alanine aminotransferase
ANC Absolute neutrophil count Neutrophil count
APTT Activeated partial thromboplastin time Partial thromboplastin time
AR Androgen receptor Androgen receptor
AST Aspartate aminotransferase Aspartate aminotransferase
BET Bromodomain and extra-terminal domain Bromodomains and superterminal structures
BID Bis in die Twice daily
BRCA Breast cancer susceptibility gene Breast cancer susceptibility gene
BRD4 Bromodomain-containing protein 4 Bromodomain protein 4
CHL Chinese hamster lung cells Chinese hamster lung cells
CHO Chinese Hamster Ovary Chinese hamster ovary cells
CL Plasma clearance rate Clearance rate of blood plasma
Cmax Peak concentration Peak concentration
CR Complete remission Complete relief
CRC Colorectal cancer Colorectal cancer
DFS Disease-free survival Disease-free survival
DLBCL Diffuse large B-cell lymphoma Diffuse large B cell lymphoma
DLT Dose limited toxicity Dose limiting toxicity
EC50 50%effective concentration Half the effective concentration
ECHO Echocardiography Echocardiography
ER Estrogen receptor Estrogen receptors
GPCR G protein coupled receptor G-protein coupled receptors
HER2 Human epidermal growth factor receptor 2 Human epidermal growth factor receptor
HNSTD Maximum non serious toxic dose Highest non-severe toxic dose
ICF Informed consent form Informed consent
INR International Normalized Ratio International normalized ratio
IUD Intrauterine device Intrauterine device
LOAEL The lowest dose at which harmful effects are observed The lowest dose at which adverse effects are observed
MTD Maximum torlerate dose Maximum tolerated dose
MUGA Multiple uptake gated acquisition scan Multi-gate circuit detection
NOAEL No harmful effect dose level Dose level without harmful effect
NYHA New York Heart Association The new york heart disease association of america
OHM Other hematological malignancies Other hematological malignancies
OS Overall survival Overall life cycle
PD-L1 Programmed cell death-Ligand 1 Programmed death receptor-ligand 1
PR Partial remission Partial mitigation
PD progression disease Progression of disease
PR Progesterone receptor Progestagen receptors
PT Prothrombin time Prothrombin time
RECIST Response evaluation criteria in solid tumours Evaluation criteria for therapeutic efficacy of solid tumor
RP2D Recommended phase 2 dose Recommended dose for phase II clinical trial
SAE Serious adverse event Serious adverse events
SD Stable disease Stabilization of disease
TGI Tumor growth inhibition Tumor growth inhibition
T1/2 half life Half life period
Tmax Peak time Time to peak
TNBC Triple-Negative Breast Cancer Triple negative breast cancer
ULN Upper limit of normal Upper limit of normal value
Vdss Apparent volume of steady state distribution Volume of steady state apparent distribution

Claims (14)

  1. The application of a compound A or a pharmaceutically acceptable salt thereof in preparing a medicament for treating tumors, wherein the structure of the compound A is shown as the following formula (I):
    Figure PCTCN2021079672-APPB-100001
    the tumor is preferably an advanced solid tumor or a refractory solid tumor, more preferably the solid tumor in the advanced solid tumor or the refractory solid tumor is gastrointestinal tumor, colorectal cancer, breast cancer or prostate cancer, and further preferably the solid tumor in the advanced solid tumor or the refractory solid tumor is colorectal cancer, triple negative breast cancer or androgen-independent prostate cancer with high infiltration of tumor-associated macrophages; preferably, the subject having the tumor is a human.
  2. The use of claim 1, wherein: the medicine is prepared into clinically acceptable preparations, such as oral preparations, injection preparations, topical preparations and external preparations.
  3. Use according to claim 1 or 2, characterized in that: the medicament is in a single dose dosage form or a multi-dose dosage form; a therapeutically effective amount of compound a, or a pharmaceutically acceptable salt thereof, (based on the weight of compound a) in a dosage form of from about 0.001mg to about 1000mg, preferably from about 1mg to about 500mg, or from about 5mg to about 400mg, or from about 10mg to about 300mg, or from about 10mg to about 250mg, or from about 10mg to about 200mg, or from about 20mg to about 200mg, more preferably from about 10mg to about 160mg, or from about 20mg to about 160mg, or from about 80mg to about 160mg; for example, about 10mg, about 20mg, about 40mg, about 60mg, about 80mg, about 100mg, about 120mg, about 140mg, about 160mg, more preferably about 10mg, about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, still more preferably about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, or still more preferably about 10mg or about 40mg.
  4. Use according to any one of claims 1 to 3, wherein: the compound a or a pharmaceutically acceptable salt thereof may be used in combination with one or more other targeted or chemotherapeutic drugs.
  5. A method of treating a neoplasm, said method comprising administering to a subject or patient a therapeutically effective amount of compound a, or a pharmaceutically acceptable salt thereof, having the structure of formula (i):
    Figure PCTCN2021079672-APPB-100002
    the tumor is preferably an advanced solid tumor or a refractory solid tumor, further preferably the solid tumor in the advanced solid tumor or the refractory solid tumor is gastrointestinal tumor, breast cancer, colorectal cancer or prostate cancer, further preferably the solid tumor in the advanced solid tumor or the refractory solid tumor is colorectal cancer, triple negative breast cancer or androgen-independent prostate cancer with high infiltration of tumor-associated macrophages; preferably the subject or patient is a human.
  6. The method of claim 5, wherein said administration is oral administration, injection administration, topical administration, or in vitro administration; or the compound A or the pharmaceutically acceptable salt thereof is prepared into clinically acceptable preparations and then is administered, wherein the preparations comprise oral preparations, injection preparations, local administration preparations and external preparations.
  7. The method of any one of claims 5 to 6, wherein compound a or a pharmaceutically acceptable salt thereof (based on compound a) is administered at a daily dose of about 0.001mg to about 1000mg, preferably about 1mg to about 500mg, or about 5mg to about 400mg, or about 10mg to about 300mg, or about 10mg to about 250mg, or about 10mg to about 200mg, or about 20mg to about 200mg, more preferably about 10mg to about 160mg, or about 20mg to about 160mg, or about 80mg to about 160mg; for example, about 10mg, about 20mg, about 40mg, about 60mg, about 80mg, about 100mg, about 120mg, about 140mg, about 160mg, more preferably about 10mg, about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, still more preferably about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, administered in a single dose or divided doses.
  8. The method of any one of claims 5-7, wherein the dosing regimen of compound a or a pharmaceutically acceptable salt thereof is 28 days per cycle, once daily for 21 days, and 7 days off; preferably, compound a or a pharmaceutically acceptable salt thereof (based on compound a) is administered at about 20mg, about 40mg, about 80mg, about 120mg, about 160mg per administration.
  9. The method of any one of claims 5-8, wherein: the compound A or the pharmaceutically acceptable salt thereof is used in combination with one or more of other targeted drugs or chemotherapeutic drugs.
  10. Compound a or a pharmaceutically acceptable salt thereof, for use in the treatment of a tumor, preferably an advanced solid tumor or a refractory solid tumor, more preferably a solid tumor of said advanced solid tumor or refractory solid tumor is a gastrointestinal tumor, colorectal cancer, breast cancer or prostate cancer, further preferably a solid tumor of said advanced solid tumor or refractory solid tumor is a colorectal cancer, triple negative breast cancer, androgen-independent prostate cancer, which is highly infiltrated with tumor-associated macrophage cells; preferably the subject having said tumor is a human,
    wherein the structure of the compound A is shown as the following formula (I):
    Figure PCTCN2021079672-APPB-100003
  11. compound a or a pharmaceutically acceptable salt thereof as claimed in claim 10, wherein the compound a or a pharmaceutically acceptable salt thereof is administered after being formulated into clinically acceptable formulations including oral, injectable, topical and topical formulations.
  12. Compound a or a pharmaceutically acceptable salt thereof according to any one of claims 10 to 11, wherein compound a or a pharmaceutically acceptable salt thereof (based on compound a) is administered at a dose of from about 0.001mg to about 1000mg, preferably from about 1mg to about 500mg, or from about 5mg to about 400mg, or from about 10mg to about 300mg, or from about 10mg to about 250mg, or from about 10mg to about 200mg, or from about 20mg to about 200mg, more preferably from about 10mg to about 160mg, or from about 20mg to about 160mg, or from about 80mg to about 160mg per day; for example, about 10mg, about 20mg, about 40mg, about 60mg, about 80mg, about 100mg, about 120mg, about 140mg, about 160mg, more preferably about 10mg, about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, still more preferably about 20mg, about 40mg, about 80mg, about 120mg or about 160mg, administered in a single dose or divided doses.
  13. Compound a or a pharmaceutically acceptable salt thereof according to any one of claims 11-12, wherein the dosing regimen for compound a or a pharmaceutically acceptable salt thereof is 28 days per cycle, once daily for 21 days, and off for 7 days; preferably, compound a or a pharmaceutically acceptable salt thereof (based on compound a) is administered at about 20mg, about 40mg, about 80mg, about 120mg, about 160mg per administration.
  14. Compound a, or a pharmaceutically acceptable salt thereof, according to any one of claims 11-13, wherein: the compound A or the pharmaceutically acceptable salt thereof is used in combination with one or more of other targeted drugs or chemotherapeutic drugs.
CN202180017690.XA 2020-03-09 2021-03-09 Application of BRD4 inhibitor Pending CN115190800A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202010156960 2020-03-09
CN2020101569602 2020-03-09
PCT/CN2021/079672 WO2021180055A1 (en) 2020-03-09 2021-03-09 Use of brd4 inhibitor

Publications (1)

Publication Number Publication Date
CN115190800A true CN115190800A (en) 2022-10-14

Family

ID=77671222

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180017690.XA Pending CN115190800A (en) 2020-03-09 2021-03-09 Application of BRD4 inhibitor

Country Status (2)

Country Link
CN (1) CN115190800A (en)
WO (1) WO2021180055A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115894678B (en) * 2022-07-15 2023-10-20 新疆赛尔托马斯生物科技有限公司 Preparation method of stem cell cytokine and application of stem cell cytokine in preparation of medicines

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019056950A1 (en) * 2017-09-22 2019-03-28 南京明德新药研发股份有限公司 Thienodiazepine derivatives and application thereof
CN113646312A (en) * 2019-03-22 2021-11-12 石药集团中奇制药技术(石家庄)有限公司 Solid BRD4 inhibitor compound and preparation method and application thereof

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170027955A1 (en) * 2014-04-04 2017-02-02 Constellation Pharmaceuticals, Inc. Expression levels of bcl-xl, bcl2, bcl-w, and bad and cancer therapies
WO2016156905A1 (en) * 2015-04-03 2016-10-06 Oncoethix Gmbh Pharmaceutical doses for a bromodomain and extraterminal protein (bet) inhibitor
WO2016196065A1 (en) * 2015-05-29 2016-12-08 Genentech, Inc. Methods and compositions for assessing responsiveness of cancers to bet inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019056950A1 (en) * 2017-09-22 2019-03-28 南京明德新药研发股份有限公司 Thienodiazepine derivatives and application thereof
CN113646312A (en) * 2019-03-22 2021-11-12 石药集团中奇制药技术(石家庄)有限公司 Solid BRD4 inhibitor compound and preparation method and application thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
赵刚主编: "《常见肛肠病图谱》", 人民军医出版社, pages: 88 *

Also Published As

Publication number Publication date
WO2021180055A1 (en) 2021-09-16

Similar Documents

Publication Publication Date Title
EP3076972B1 (en) Cancer treatment with combination of plinabulin and taxane
EP2269604B1 (en) Treatment of solid kidney tumours with a rapamycin derivative
US20070281040A1 (en) Combination therapy of hedgehog inhibitors, radiation and chemotherapeutic agents
JP2012500180A5 (en)
US11576919B2 (en) Treatment of breast cancer using combination therapies comprising an ATP competitive AKT inhibitor, a CDK4/6 inhibitor, and fulvestrant
JP2015512388A (en) Combination therapy
CN115135649A (en) Pharmaceutical combinations comprising pyrido [1,2-a ] pyrimidone compounds
JP2022501394A (en) Pharmaceutical compositions of MDM2 inhibitors, and their use for the prevention and / or treatment of diseases
US20150072020A1 (en) Dexanabinol or a Derivative Thereof for Use in the Treatment of Cancer in Dose Ranges of 2-30 mg/kg
CN115190800A (en) Application of BRD4 inhibitor
CA2437768A1 (en) Targetted anti-tumor drug delivery systems
US20230038138A1 (en) Combination therapy for treating cancer
EP4159238A1 (en) Combined pharmaceutical composition of c-met kinase inhibitor and anti-pd-l1 antibody
KR20100126453A (en) Combination anti-cancer agents
TW202029961A (en) Use of ar antagonist combined with parp inhibitor in preparation of medicament for treating prostate cancer
TWI843217B (en) A drug combination and its application
WO2021228146A1 (en) Use of lsd1 inhibitor
CN117357528A (en) New application of kinase inhibitor
US20230404971A1 (en) Combination Comprising Abemaciclib and 6-(2,4-Dichlorophenyl)-5-[4-[(3S)-1-(3-Fluoropropyl)Pyrrolidin-3-yl]Oxyphenyl]-8,9-Dihydro-7H-Benzo[7]Annulene-2-Carboxylic Acid
CN117919234A (en) Application of naphthalamide compound in treatment of drug-resistant tumor
WO2021185234A1 (en) Combined pharmaceutical composition of compound as c-met kinase inhibitor and use thereof
TW202308641A (en) Methods and dosing regimens comprising a cdk inhibitor for the treatment of cancer
CN117940164A (en) Application of mitoxantrone liposome combined with anti-angiogenesis targeting drug in treatment of ovarian cancer
CN115300511A (en) Medical application of composition
CN114984027A (en) Application of saikosaponin A in preparation of medicine for relieving toxic and side effects of EGFR inhibitor

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