CN115607548A - Application of tyrosine kinase inhibitor in combination with neoadjuvant chemotherapeutic drugs for treating tumors - Google Patents

Application of tyrosine kinase inhibitor in combination with neoadjuvant chemotherapeutic drugs for treating tumors Download PDF

Info

Publication number
CN115607548A
CN115607548A CN202210837289.7A CN202210837289A CN115607548A CN 115607548 A CN115607548 A CN 115607548A CN 202210837289 A CN202210837289 A CN 202210837289A CN 115607548 A CN115607548 A CN 115607548A
Authority
CN
China
Prior art keywords
use according
cyclophosphamide
her4
compound
administration
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
CN202210837289.7A
Other languages
Chinese (zh)
Inventor
宋尔卫
陈凯
谭璐媛
庞正
刘翼龙
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.)
Jiangsu Hengrui Medicine Co Ltd
Original Assignee
Jiangsu Hengrui Medicine 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 Jiangsu Hengrui Medicine Co Ltd filed Critical Jiangsu Hengrui Medicine Co Ltd
Publication of CN115607548A publication Critical patent/CN115607548A/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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

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

Abstract

The present disclosure relates to the use of tyrosine kinase inhibitors in combination with neoadjuvant chemotherapeutic drugs for the treatment of tumors. In particular, the disclosure relates to the use of a tyrosine kinase inhibitor for the preparation of a medicament for the treatment of HER2 negative, HER4 high expressing (HER 2-/HER4 high expressing) tumors. Specifically, the disclosure relates to an application of a tyrosine kinase inhibitor compound shown in formula (I) or a pharmaceutically acceptable salt thereof in preparing a medicament for treating HER2 negative and HER4 high-expression tumors.

Description

Application of tyrosine kinase inhibitor in combination with neoadjuvant chemotherapeutic drugs for treating tumors
Technical Field
The disclosure belongs to the field of medicine, and relates to application of a tyrosine kinase inhibitor in preparation of a medicine for treating HER2 negative and HER4 high-expression tumors.
Background
Malignant tumor is a serious disease which endangers the life and health of people. In recent years, with the rapid development of tumor biology and related disciplines, specific anti-tumor drugs aiming at abnormal signal system targets in tumor cells are the focus of new drug development. Meanwhile, the combination of multiple antitumor drugs for treating tumor diseases is also a hot spot of scientific research.
Breast cancer is the most common female malignancy, accounting for 30% of all women's newly diagnosed tumors. Wherein HR + HER 2-type breast cancer accounts for 55-70% of the total proportion of breast cancer. However, the response rate of the chemotherapy for the breast cancer is not high, and a specific treatment target is lacked, so that endocrine therapy aiming at hormone receptors is the main treatment method at present. However, due to tumor heterogeneity, endocrine drug resistance pathway activation and other reasons, tumor recurrence and metastasis still appear in about 30% of patients after treatment, and recurrence and metastasis caused by poor treatment effect are important causes of death of the breast cancer patients. Therefore, the method for improving the treatment effect of HR + HER 2-type breast cancer is important for improving the treatment level of the whole breast cancer and reducing the overall mortality of breast cancer patients.
Receptor tyrosine kinases are a class of transmembrane proteins involved in signal transduction and are expressed in a variety of cells to regulate cell growth, differentiation and neovascularization. Research shows that over 50% of proto-oncogenes and oncogene products have tyrosine kinase activity, and abnormal expression of the proto-oncogenes and oncogene products leads to tumorigenesis and is also closely related to tumor invasion and metastasis, tumor angiogenesis, and tumor chemotherapy resistance. WO2011029265 discloses an effective tyrosine kinase inhibitor and a preparation method thereof, the structure of which is shown as formula I,
Figure BDA0003749090810000011
the compound has obvious drug effect advantage. The dimaleate salt form of this compound is described in CN102933574a, which has improved physicochemical, pharmacokinetic and bioavailability properties, the compound of formula I has been demonstrated to be effective in HER2 positive (HER 2 +) breast cancer, but there have been no relevant clinical findings for HER2 negative (HER 2-) breast cancer.
Disclosure of Invention
The disclosure provides an application of a compound shown in a formula (I) or a medicinal salt thereof in preparing a medicament for treating HER2 negative and HER4 high-expression tumors, wherein the tumors are selected from lung cancer, liver cancer, colorectal cancer or breast cancer,
Figure BDA0003749090810000021
in some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is used in combination with a neoadjuvant chemotherapeutic agent. The disclosure also provides a method of treating a HER2 negative, HER4 high expressing tumor comprising administering to a patient a compound of formula (I) or a pharmaceutically acceptable salt thereof.
The disclosure also provides a method of treating a HER2 negative, HER4 high expressing tumor comprising administering to a patient a compound of formula (I) or a pharmaceutically acceptable salt thereof and a neoadjuvant chemotherapeutic agent.
The disclosure also provides a compound shown in formula (I) or a pharmaceutically acceptable salt thereof for treating HER2 negative and HER4 high-expression tumors.
The disclosure also provides a compound shown in formula (I) or a pharmaceutically acceptable salt thereof for treating HER2 negative and HER4 high-expression tumors, and the compound is combined with a neoadjuvant chemotherapeutic drug.
The disclosure also provides a neoadjuvant chemotherapeutic drug for treating HER2 negative and HER4 high-expression tumors, which is combined with the compound shown in the formula (I) or the pharmaceutically acceptable salt thereof.
In some embodiments, the HER2 negative, HER4 high expressing tumor is a HR positive tumor, wherein the tumor is selected from lung cancer, liver cancer, colorectal cancer, or breast cancer.
In some embodiments, the HER2 negative, HER4 high expressing tumor is an ER positive tumor, wherein the tumor is selected from lung cancer, liver cancer, colorectal cancer, or breast cancer.
In some embodiments, the HER 2-negative, HER 4-high expressing tumor is a non-small cell lung cancer or a breast cancer.
In some embodiments, the HER 2-negative, HER 4-high expressing tumor is a breast cancer.
In some embodiments, the HER2 negative, HER4 high expressing tumor is HR positive breast cancer.
In some embodiments, the HER 2-negative, HER 4-high expressing tumor is an ER-positive breast cancer.
In some embodiments, the breast cancer is stage II/III (28 days prior to screening for visual evidence, staging using AJCC cancer staging manual (eighth edition)) breast cancer.
In some embodiments, the pharmaceutically acceptable salt of the compound of formula (I) is selected from the group consisting of hydrochloride, phosphate, hydrogen phosphate, sulfate, hydrogen sulfate, sulfite, acetate, oxalate, malonate, valerate, glutamate, oleate, palmitate, stearate, laurate, borate, p-toluenesulfonate, methanesulfonate, isethionate, maleate. Malate, tartrate, benzoate, pamoate, salicylate, vanillate, mandelate, succinate, gluconate, lactobionate and laurylsulfonate.
In some embodiments, the pharmaceutically acceptable salt of the compound of formula (I) is a maleate salt.
In some embodiments, the pharmaceutically acceptable salt of the compound of formula (I) is a dimaleate salt.
In some embodiments, the pharmaceutically acceptable salt of the compound of formula (I) and crospovidone form a pharmaceutical composition.
In some embodiments, the pharmaceutical composition further comprises a binder in an amount of 0.5 to 15% by weight based on the total weight of the composition.
In some embodiments, the binder in the pharmaceutical composition is selected from one or more of hydroxypropyl methylcellulose, hydroxypropyl cellulose, sodium hydroxymethyl cellulose, polyvinylpyrrolidone, and methylcellulose.
In some embodiments, the pharmaceutical composition further comprises a filler in an amount of 5% to 80% based on the total weight of the composition.
In some embodiments, the filler in the pharmaceutical composition is selected from one or more of microcrystalline cellulose, calcium bicarbonate, mannitol, pregelatinized starch, and lactose.
In some embodiments, the pharmaceutical composition further comprises a lubricant in an amount of 0.5% to 5% based on the total weight of the composition.
In some embodiments, the lubricant in the pharmaceutical composition is selected from one or more of talc, magnesium stearate, zinc stearate, glyceryl behenate, sodium lauryl sulfate, hydrogenated vegetable oil, colloidal silicon dioxide.
In some embodiments, the neoadjuvant chemotherapeutic agent is selected from the group consisting of an anthracycline, a taxane, cyclophosphamide, and 5-fluorouracil in combination with one or more other drugs.
In some embodiments, the neoadjuvant chemotherapeutic agent is selected from any combination of:
(1) Anthracyclines, cyclophosphamide and taxanes;
(2) Anthracyclines and cyclophosphamide;
(3) Taxoids and cyclophosphamide;
(4) 5-fluorouracil, anthracyclines and cyclophosphamide.
In some embodiments, the neoadjuvant chemotherapeutic agent is an anthracycline, cyclophosphamide, or a taxane.
In some embodiments, the neoadjuvant chemotherapeutic agent is an anthracycline and cyclophosphamide.
In some embodiments, the neoadjuvant chemotherapeutic agent is a taxane and cyclophosphamide.
In some embodiments, the neoadjuvant chemotherapeutic agent is 5-fluorouracil, an anthracycline, and cyclophosphamide.
In some embodiments, the anthracycline is selected from the group consisting of epirubicin, pirarubicin, and doxorubicin hydrochloride liposomal injection (doxorubicin hydrochloride liposomal injection).
In some embodiments, the taxane is selected from the group consisting of paclitaxel, albumin paclitaxel, and docetaxel.
In some embodiments, the neoadjuvant chemotherapeutic agent is selected from any combination of:
(1) Doxorubicin hydrochloride liposomal injections, cyclophosphamide, and docetaxel;
(2) Epirubicin, cyclophosphamide and docetaxel;
(3) Pirarubicin, cyclophosphamide and docetaxel;
(4) Doxorubicin hydrochloride liposome injection, cyclophosphamide and albumin paclitaxel;
(5) Epirubicin, cyclophosphamide and albumin paclitaxel;
(6) Pirarubicin, cyclophosphamide and albumin paclitaxel.
In some embodiments, the neoadjuvant chemotherapeutic agent is doxorubicin hcl liposome injection, cyclophosphamide, and docetaxel.
In some embodiments, the neoadjuvant chemotherapeutic agent is epirubicin, cyclophosphamide, and docetaxel.
In some embodiments, the neoadjuvant chemotherapeutic agent is pirarubicin, cyclophosphamide and docetaxel.
In some embodiments, the neoadjuvant chemotherapeutic agent is doxorubicin hydrochloride liposomal injection, cyclophosphamide, and albumin paclitaxel.
In some embodiments, the neoadjuvant chemotherapeutic agent is epirubicin, cyclophosphamide, and albumin paclitaxel.
In some embodiments, the neoadjuvant chemotherapeutic agent is pirarubicin, cyclophosphamide, and albumin paclitaxel.
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in a dose of 100mg to 1000mg once daily.
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered at a dose of 240 to 560mg once daily.
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in a dose of 320 to 480mg once daily.
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in an amount selected from the group consisting of 240mg, 320mg, and 400mg, once daily.
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered at a dose of 400mg once daily.
The dosage of the compound represented by the formula (I) or the pharmaceutically acceptable salt thereof described in the present disclosure is calculated based on the compound represented by the formula (I).
<xnotran> , (I) 100mg 1000mg, 100mg, 105mg, 110mg, 115mg, 120mg, 125mg, 130mg, 135mg, 140mg, 145mg, 150mg, 155mg, 160mg, 165mg, 170mg, 175mg, 180mg, 185mg, 190mg, 195mg, 200mg, 205mg, 210mg, 215mg, 220mg, 225mg, 230mg, 235mg, 240mg, 245mg, 250mg, 255mg, 260mg, 265mg, 270mg, 275mg, 280mg, 285mg, 290mg, 295mg, 300mg, 305mg, 310mg, 315mg, 320mg, 325mg, 330mg, 335mg, 340mg, 345mg, 350mg, 355mg, 360mg, 365mg, 370mg, 375mg, 380mg, 385mg, 390mg, 395mg, 400mg, 405mg, 410mg, 415mg, 420mg, 425mg, 430mg, 435mg, 440mg, 445mg, 450mg, 455mg, 460mg, 465mg, 470mg, 475mg, 480mg, 485mg, 490mg, 495mg, 500mg, 505mg, 510mg, 515mg, 520mg, 525mg, 530mg, 535mg, 540mg, 545mg, 550mg, 555mg, 560mg, 565mg, 570mg, 575mg, 580mg, 585mg, 590mg, 595mg, 600mg, 605mg, 610mg, 615mg, 620mg, 625mg, 630mg, 635mg, 640mg, 645mg, 650mg, 655mg, 660mg, 665mg, 670mg, 675mg, 680mg, 685mg, 690mg, 695mg, 700mg, 705mg, 710mg, 715mg, 720mg, 725mg, 730mg, 735mg, 740mg, 745mg, 750mg, 755mg, 760mg, 765mg, 770mg, 775mg, 780mg, 785mg, 790mg, 795mg, 800mg, 805mg, 810mg, 815mg, 820mg, 825mg, 830mg, 835mg, 840mg, 845mg, 850mg, 855mg, 860mg, 865mg, 870mg, 875mg, 880mg, 885mg, 890mg, 895mg, 900mg, 905mg, 910mg, 915mg, 920mg, 925mg, 930mg, 935mg, 940mg, 945mg, 950mg, 955mg, 960mg, 965mg, 970mg, 975mg, 980mg, 985mg, 990mg, 995mg, 1000mg. </xnotran>
For asians, the daily dosage may range between 240 and 400 mg.
The compound of formula (I) or a pharmaceutically acceptable salt thereof of the present disclosure may be used once daily.
An alternative embodiment of the compounds of formula (I) of the present disclosure or pharmaceutically acceptable salts thereof is administration after meal consumption.
An alternative embodiment of the compound of formula (I) or a pharmaceutically acceptable salt thereof of the present disclosure is administered within 30 minutes after meal consumption.
An alternative embodiment of the compound of formula (I) or a pharmaceutically acceptable salt thereof of the present disclosure is taken within 30 minutes after breakfast.
In some embodiments, the crospovidone is present in an amount of 2% to 20% based on the total weight of the composition.
In some embodiments, the crospovidone is present in an amount of 4% to 15% based on the total weight of the composition.
In some embodiments, the crospovidone is present in an amount of 6% to 10% based on the total weight of the composition.
In some embodiments, the dose of the doxorubicin hydrochloride liposomal injection is 20mg/m 2 ~100mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the liposomal doxorubicin hydrochloride injection is at a dose of 30mg/m 2 ~60mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the composition is prepared byThe dosage of the doxorubicin hydrochloride liposome injection is 30mg/m 2 Or 50mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of epirubicin is 60mg/m 2 ~120mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of epirubicin is 75mg/m 2 ~100mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of epirubicin is 100mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of pirarubicin is 30mg/m 2 ~80mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of pirarubicin is 30mg/m 2 ~50mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of pirarubicin is 50mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of cyclophosphamide is 400mg/m 2 ~1000mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of cyclophosphamide is 500mg/m 2 Or 600mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of docetaxel is 60mg/m 2 ~120mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the docetaxel is administered at a dose of 75mg/m 2 ~100mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the docetaxel is administered at a dose of 75mg/m 2 Or 100mg/m 2 The frequency of administration was 1 time every 3 weeks.
In some embodiments, the dose of albumin paclitaxel is 60mg/m 2 ~140mg/m 2 The frequency of administration was 1 time per week.
In some embodiments, the dose of albumin paclitaxel is 80mg/m 2 ~120mg/m 2 (ii) a The frequency of administration was 1 time per week.
In some embodiments, the dose of albumin paclitaxel is 80mg/m 2 Or 120mg/m 2 (ii) a The frequency of administration was 1 time per week.
The modes of administration of the combinations of the present disclosure are selected from simultaneous administration, independently formulated and co-administered, or independently formulated and administered sequentially.
The route of administration of the combination of the present disclosure is selected from oral, parenteral, transdermal, including but not limited to intravenous, subcutaneous, intramuscular.
In some embodiments, the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered orally.
In some embodiments, the neoadjuvant chemotherapeutic agent is administered intravenously.
The present disclosure further relates to the use of a compound of formula (I) or a pharmaceutically acceptable salt thereof, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof may be administered once daily, twice daily, three times daily, once every two days, once every three days, twice weekly and once weekly, in combination with a neoadjuvant chemotherapeutic agent, in the manufacture of a medicament for the treatment of a neoplastic disease.
In the embodiment of the present disclosure, the combination optionally further comprises other components, including but not limited to other antitumor agents, etc.
The compounds of formula (I) or pharmaceutically acceptable salts thereof, and neoadjuvant chemotherapeutic agents, as described in this disclosure, may be administered alone or in combination with one or more therapeutic agents.
The compound of formula (I) or a pharmaceutically acceptable salt thereof, and various neoadjuvant chemotherapeutic agents of the present disclosure may be administered simultaneously or separately in sequential order. In addition, the compound represented by the formula (I) or a pharmaceutically acceptable salt thereof and various neoadjuvant chemotherapeutic agents of the present disclosure may also be administered in combination in the same formulation or in separate and distinct formulations.
The high expression of HER4 as described in the present disclosure can be understood as the increase of the HER4 mRNA expression level of ER + HER 2-breast cancer cell line in breast cancer cell lines by 4-5 times compared to other molecular subtype breast cancer cell lines; or in an immunohistochemical experiment of a patient specimen, the HER4 expression amount is scored, and the HER4 protein expression SCORE of the ER + HER 2-breast cancer is the highest in 4 molecular subtypes, such as pathologically confirmed HER4 immunohistochemistry 2+, 3+ or immunohistochemical SCORE SCORE > =4.
HER2 negativity as described in the present disclosure may be understood as immunohistochemistry ≦ 2+ and/or FISH negativity.
The term "combination" as used in this disclosure is intended to mean a mode of administration in which at least one dose of a compound of formula (I) or a pharmaceutically acceptable salt thereof and at least one group of neoadjuvant chemotherapeutic agents are administered over a period of time, wherein the administered agents all exhibit pharmacological effects. The time period may be within one administration cycle.
In some embodiments, one dosing cycle is within 4 weeks.
In some embodiments, one dosing cycle is within 3 weeks.
In some embodiments, one dosing cycle is within 2 weeks.
In some embodiments, one dosing cycle is within 1 week.
In some embodiments, one dosing cycle is within 24 hours.
In some embodiments, one dosing cycle is within 12 hours.
The compound of formula (I) or a pharmaceutically acceptable salt thereof and the neoadjuvant chemotherapeutic agent may be administered simultaneously or sequentially. Such term includes treatments wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof and the neoadjuvant chemotherapeutic agent are administered by the same route of administration or different routes of administration.
The mode of administration of the combinations of the present disclosure is selected from simultaneous administration, separate formulation and co-administration, or separate formulation and sequential administration.
The present disclosure provides a synergistic therapeutic effect of administering a compound represented by formula (I) or a pharmaceutically acceptable salt thereof in combination with a neoadjuvant chemotherapeutic agent or a pharmaceutically acceptable salt thereof, thereby enhancing antitumor activity.
Drawings
FIG. 1 shows the results of the analysis of the mRNA expression levels of HER4 in different molecular subtypes in the data set GEO (GSE 81540) and TCGA breast cancer patients;
FIG. 2 is data correlating the amount of HER4 expression in ER + HER 2-breast cancer with prognosis;
FIG. 3 is a graph showing the correlation between the expression level of HER4 and DFS;
FIG. 4-1 is an analysis result of the expression level of HER4 in ER + HER 2-breast cancer cell lines MCF7 and T47D;
FIG. 4-2 shows the results of analysis of the expression level of HER4 in MCF-7 and T47D;
FIGS. 4 to 3 are results of analysis of the expression level of HER4 in 3 kinds of cells in the form of a thermogram;
FIG. 5 shows the results of the inhibition of the cloning ability of MCF-7 and T47D cells by Compound 1;
FIGS. 6 and 7 are data showing the effect of Compound 1 in inhibiting tumor growth in a mouse MCF7 breast cancer xenograft model;
Detailed Description
Example 1: investigation of HER4 expression in different molecular subtypes of breast cancer and correlation with DFS
Research and design: selecting a data set GEO (GSE 81540) with the sample size of more than 1000 and TCGA breast cancer patients from a public database, analyzing the mRNA expression quantity of HER4 in different molecular subtypes, and finding that the HER4 is highly expressed in an ER positive breast cancer patient sample (p is less than 0.001); meanwhile, the expression level of HER4 in the Luminal A type of ER + HER 2-is higher than that of other molecular types (p < 0.001) through molecular typing analysis (see figure 1), and the expression level of HER4 (median is a cut-off value with high and low expression level) is negatively correlated with prognosis in ER + HER 2-breast cancer through K-M curve analysis (see figure 2), which indicates that HER4 is a potential key molecule influencing the prognosis of ER + HER 2-breast cancer, and the targeted HER4 can possibly play an anti-tumor role.
220 cases of pathological sections of breast cancer patients diagnosed in 2014 by a breast tumor center of a memorial hospital of Zhongshan university Sun Yixian were collected, and IHC staining of HER4 was performed in 4 molecular typing species distribution. HER4 protein expression levels were classified according to staining intensity and positive cell ratio, and it was found that HER4 expression levels were the highest in ER + HER 2-breast cancer (p < 0.001), HER4 protein expression levels were lower in HER 2-positive breast cancer, and HER4 protein was hardly expressed in Triple Negative Breast Cancer (TNBC). HER4 staining intensity was divided into high and low expression using median, with higher HER4 expression and poorer DFS in ER + HER 2-breast cancer (see fig. 3).
And (4) conclusion: HER4 is expressed at a higher level in ER + HER 2-breast cancer, and the expression level of HER4 is negatively correlated with DFS.
Example 2: cell proliferation inhibition assay of compound 1 on HER 4-highly expressed MCF7 and T47D
Experiment design: the expression of known targets EGFR, HER2 and HER4 of the compound of formula I was detected in a common cell line of breast cancer, and ER-positive HER 2-negative (ER + HER 2-) breast cancer cell lines MCF7 and T47D with only HER4 high expression were found (see FIG. 4-1), suggesting that pan-HER TKIs have the potential to exert antitumor effect on the ER-positive HER 2-negative MCF-7 and T47D cell lines through HER 4. Further, in MCF-7 and T47D cell lines, qPCR was performed using the highly expressed ER gene as a positive control, and a cell line BT474 in which HER4 was expressed in an amount comparable to that of ER and higher than HER2+ in MCF-7 and T47D was found (see FIG. 4-2); the results of cellular mRNA sequencing of MCF-7, T47D and BT474 were also downloaded from the Oncomine database and analyzed in the form of heatmap for HER4 expression in 3 cells, consistent with the qPCR results (see fig. 4-3). This revealed that HER4 was highly expressed in MCF-7 and T47D. MCF-7 and T47D of HER4 with high expression are selected for subsequent experiments.
Experimental drugs and cell sources:
compound 1: which is the dimaleate salt of a compound of formula I:
Figure BDA0003749090810000081
produced and supplied by Henry pharmaceutical Co., ltd, jiangsu.
Cell lines used in this disclosure were all purchased from ATCC.
The experimental process comprises the following steps: MCF-7 and T47D breast cancer ER + HER2 cell lines with high HER4 expression are planted in a 96-well plate by 3000 cells/well, compound 1 is added after 24 hours, the cell activity is detected by a CCK8 experiment, the positive cell control is HER2+ (HER 2 positive) BT-474 (BT 474 is HER2 positive, and Pan-HER TKIs belong to the classification of the current clinical indications of the Pan-HER TKIs), and the negative cell control is 231 cell strains.
The experimental results are as follows:
TABLE 1 IC of Compound 1 for cellular proliferation inhibition of HER 4-highly expressed MCF7 and T47D 50 Value of
Figure BDA0003749090810000082
And (4) conclusion: compound 1 significantly inhibited cell proliferation of ER + HER2 negative cell lines MCF7 (highly expressed HER 4) and T47D (highly expressed HER 4) and MCF-7 and T47D clonogenic capacity was reduced after 48 hours of compound 1 treatment (see fig. 5).
Example 3: drug effect experiment of compound 1 in inhibiting mouse MCF7 (HER 4 high expression) breast cancer xenograft
The experimental process comprises the following steps: construction of the MCF7 xenograft tumor model was performed using 4-week-old NOD-SCID mice (purchased from Schlekschada laboratory animals Co., ltd., hunan) divided into 3 groups including a drug solvent control group, a compound 1 low dose group (10 mg/kg), a compound 1 high dose group (20 mg/kg), and 6 mice per group. The right fat pad of the 3 rd pair of fat pads in each mouse was injected with 100ul PBS resuspended 1 x 10^7 MCF7 (HER 4 high expression) tumor cells. The intragastric administration was started when the tumor grew to a diameter of 0.5 mm. Tumor major and minor diameters were measured every 3 days, tumor volume was calculated, and after 21 days of administration, mice were sacrificed. Tumors were collected and tumor size was measured.
And (4) conclusion: compound 1 was able to significantly inhibit tumor growth in the mouse MCF7 breast cancer xenograft model and was concentration dependent (see fig. 6 and 7).
Example 4: a randomized, double-blind, controlled, phase II clinical study (NCT 04872985) of a compound of formula (I) or a pharmaceutically acceptable salt thereof in combination with neoadjuvant chemotherapy for HR +/HER2-, HER 4-highly expressed breast cancer
1. Purpose of study and endpoint index
1.1 Purpose of study
The curative effect and safety of the compound of the formula (I) in combination with a new auxiliary chemotherapeutic medicament for treating HR +/HER 2-HER 4 high-expression breast cancer are explored, the relationship between a biomarker and the curative effect is explored, and the characteristics of treatment-sensitive people are determined.
1.2 end Point index
1.2.1 Primary endpoint index
Complete remission rate of pathology (pCR): ypT0/isN, i.e., no evidence of malignant histology or only an in situ cancer component was found in the primary tumor, and no metastasis to the axillary lymph nodes.
1.2.2 Secondary endpoint indicator
(1) Breast pCR: ypT0/is, i.e., no evidence of malignant histology or only an in situ cancer component is found in the primary tumor.
(2) Objective Remission Rate (ORR): namely, evaluating according to a solid tumor curative effect evaluation standard (RECIST) version 1.1, and displaying the proportion of the number of patients with optimal remission of CR or PR in the total number of evaluable patients after the new adjuvant therapy is evaluated by the imaging;
(3) Event free lifetime (EFS): time from start of enrollment to occurrence of postoperative recurrence, metastasis or death event (whichever occurs first);
(4) Overall Survival (OS): time from start of enrollment to total cause of death;
(5) Rate of change of cell proliferation index (Ki 67): changes in Ki67 expression in tumor tissue before and after neoadjuvant therapy;
(6) HER4 expression levels correlated with therapeutic efficacy;
(7) Correlation of other peripheral blood and tissue biomarkers with treatment efficacy and safety;
(8) Adverse Events (AE), incidence of Severe Adverse Events (SAE).
2. Design of research
2.1 Overall design
The study is a prospective, random, control, single-center, double-blind, phase II clinical trial study. The study planned enrollment of patients in the memorial hospital at Zhongshan university Sun Yixian. Patients in the group meeting the inclusion criteria are randomly divided into 2 groups, and each group contains 62 people (wherein the sample size is determined by adopting 1:1 parallel design, the pCR rate of the breast cancer neoadjuvant chemotherapy is estimated to be 13% according to the research result related to the prior HR +/HER2 neoadjuvant therapy, the pCR rate of the breast cancer neoadjuvant chemotherapy is assumed to be 30% according to the research result of I-SPY2, the pCR rate of the test group is assumed to be 30%, the single-side alpha of the type I error is assumed to be 0.1, the beta value of the type II error is assumed to be 0.17, namely, the testing efficiency Power is 83%, the sample size is calculated to obtain 57 cases in each group, 10% of clinical test falling or missing is considered, and finally, 62 cases in each group are calculated, and 124 cases are required in total in the group). The following treatments were received, respectively:
test groups: epirubicin + cyclophosphamide + compound of formula (I) 4 cycle, sequential docetaxel + compound of formula (I) 4 cycle
Control group: epirubicin + cyclophosphamide + placebo 4 cycles, sequential docetaxel + placebo 4 cycles.
2.2 study drug and dosing regimen
2.2.1 study drugs:
compound 1: it is a dimaleate salt of the compound of formula I, and is prepared into tablets, the dosage of which is calculated by the compound of formula I and is produced and provided by Jiangsu Henry medicine GmbH, and the specification is selected from 40mg, 60mg and 200mg.
2.2.2 dosing regimen:
test groups: all patients received 4 cycles of intravenous chemotherapy with epirubicin (100 mg/m2, q3 w) and cyclophosphamide (600 mg/m2, q3 w), followed by 4 cycles of docetaxel (100 mg/m2, q3 w); all patients received the compound of formula (I) (400mg, qd) oral drug therapy simultaneously.
Control group: all patients received 4 cycles of intravenous chemotherapy with epirubicin (100 mg/m2, q3 w) and cyclophosphamide (600 mg/m2, q3 w), followed by 4 cycles of docetaxel (100 mg/m2, q3 w); all patients received placebo (400mg, qd) oral drug therapy simultaneously.
Compound 1: the administration is 1 time 400mg per day, and oral administration is carried out within 30 minutes after breakfast, and the continuous administration is 1 cycle for 21 days.
Epirubicin: 100mg/m2, q3w, administered intravenously on day 1, with a cycle of every 3 weeks. 4 cycles (C1-C4) before chemotherapy, all drugs are not limited to import/home versions in view of practical clinical application. Wherein epirubicin allows for replacement with either pirarubicin (30 mg/m2, q3w 4) or liposomal doxorubicin (30 mg/m2, q3w 4).
Cyclophosphamide: 600mg/m2, q3w, administered intravenously on day 1, with a cycle of every 3 weeks. Chemotherapy was given 4 cycles (C1-C4) prior to chemotherapy.
Docetaxel: 100mg/m2, q3w, intravenously administered on day 1, with one cycle every 3 weeks. Docetaxel was allowed to replace with albumin paclitaxel (120 mg/m2, q3w 4) given 4 cycles (C5-C8) post chemotherapy.
The dosage can be adjusted according to the adverse reaction condition of the subject. Depending on the type and timing of the clinical adverse event, compound 1 can be discontinued. Multiple pauses were allowed to adjust the dose of compound 1 multiple times in the trial, and compound 1 was adjusted in a gradient of 400mg, 320mg, 240mg. After adjustment of the dosing, if the subject still had clinically uncontrollable adverse events as described above, the investigator allowed to reduce the dose by one step again when resuming dosing after a pause, with a dose downregulation of a minimum of 240mg.
2.2.3 grouping criteria
(1) The age is more than or equal to 18 years old;
(2) Patients with histologically or cytologically confirmed breast cancer, stages II-III (28 days before screening for visual evidence, staged using AJCC cancer staging Manual (eighth edition));
(3) Clinically judging the indication or need of new auxiliary chemotherapy;
(4) Pathologically confirmed diagnosis is ER positive (more than or equal to 10 percent) and/or PR positive (more than or equal to 1 percent) and Her2 negative (immunohistochemistry is less than or equal to 2+ and/or FISH negative);
(5) Pathologically confirmed HER4 immunohistochemistry 2+, 3+ or immunohistochemistry SCORE SCORE ≥ 4
(6) ECOG physical condition score of 0-1;
(7) There is sufficient organ and bone marrow function: serum creatinine is less than or equal to 1.5 times of the upper limit of normal values, glutamic-pyruvic transaminase and glutamic-oxalacetic transaminase are less than or equal to 2.5 times of the upper limit of normal values, total bilirubin is less than or equal to 1.5 times of the upper limit of normal values, the white blood cell count is more than or equal to 3.0 multiplied by 10^9/L, the neutrophil count is more than or equal to 1.5 multiplied by 10^9/L, the platelet count is more than or equal to 100 multiplied by 10^9/L, and hemoglobin is more than or equal to 90g/L;
(8) Has not received any anti-tumor drug treatment, radiotherapy and surgery treatment before;
(9) Women in the fertile age must have taken reliable contraceptive measures and have been subjected to pregnancy tests (serum or urine) 7 days prior to enrollment and the results are negative and willing to use the appropriate method of contraception during the test period and 24 weeks after the last dose.
2.3 data analysis and statistics method
Statistical analysis was performed on the data of this study using R software (R software, http:// www.R-project. Org), pythom software (https:// www.pythom.com /), SPSS 24.0 software (IBM SPSS Inc, chicago, IL, USA).
Therapeutic effect analysis statistical method
The primary efficacy index pCR population and percentage were calculated and their 95% confidence intervals were calculated using the method of cloner Pearson.
The same statistical method was used for the secondary efficacy indices ORR, ki 67. For the secondary efficacy indexes EFS and OS, a Kaplan-Meier method is adopted to estimate a survival function and a median DFS value, a Greenwood formula is used to estimate the error of the survival function, and a 95% confidence interval of the median EFS and OS value is calculated. The Log-rank test was used to compare the EFS, OS survival functions of the two groups. And analyzing the influence of other factors such as layering factors on the result by adopting a Cox proportional risk model. Different sensitivity analyses are defined, and the influence of different deletion rules and hierarchical analysis on the result is discussed.

Claims (20)

1. An application of a compound shown in a formula (I) or a medicinal salt thereof in preparing a medicament for treating HER2 negative and HER4 high-expression tumors, wherein the tumors are selected from lung cancer, liver cancer, colorectal cancer or breast cancer,
Figure FDA0003749090800000011
2. the use according to claim 1, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is used in combination with a neoadjuvant chemotherapeutic agent.
3. Use according to claim 1 or 2, wherein the HER2 negative HER4 high expressing tumor is a HR positive tumor, preferably an ER positive tumor, wherein the tumor is selected from lung cancer, liver cancer, colorectal cancer or breast cancer.
4. Use according to any one of claims 1 to 3, wherein the HER2 negative, HER4 high expressing tumor is breast cancer.
5. The use according to any one of claims 1 to 4, wherein the HER2 negative, HER4 high expressing tumor is HR positive breast cancer.
6. The use according to any one of claims 1 to 5, wherein the HER2 negative, HER4 high expressing tumor is an ER positive breast cancer.
7. Use according to any one of claims 1 to 6, wherein the pharmaceutically acceptable salt of the compound of formula (I) is a maleate salt, preferably a dimaleate salt.
8. Use according to any one of claims 2 to 7, wherein the neoadjuvant chemotherapeutic is selected from the group consisting of anthracyclines, taxanes, cyclophosphamide and 5-fluorouracil in combination with one or more.
9. Use according to any one of claims 2 to 8, wherein the neoadjuvant chemotherapeutic agent is selected from any combination of:
(1) Anthracyclines, cyclophosphamide and taxanes;
(2) Anthracyclines and cyclophosphamide;
(3) Taxoids and cyclophosphamide;
(4) 5-fluorouracil, anthracyclines and cyclophosphamide.
10. The use according to claim 8 or 9, wherein the anthracycline is selected from the group consisting of epirubicin, pirarubicin, and doxorubicin hydrochloride liposomal injection.
11. The use according to claim 8 or 9, wherein the taxoid is selected from paclitaxel, albumin paclitaxel and docetaxel.
12. Use according to any one of claims 2 to 11, wherein the neoadjuvant chemotherapeutic agent is selected from any combination of:
(1) Doxorubicin hydrochloride liposomal injections, cyclophosphamide, and docetaxel;
(2) Epirubicin, cyclophosphamide and docetaxel;
(3) Pirarubicin, cyclophosphamide and docetaxel;
(4) Doxorubicin hydrochloride liposome injection, cyclophosphamide and albumin paclitaxel;
(5) Epirubicin, cyclophosphamide, and albumin paclitaxel;
(6) Pirarubicin, cyclophosphamide and albumin paclitaxel.
13. The use according to any one of claims 1 to 12, wherein the compound of formula (I) or a pharmaceutically acceptable salt thereof is administered in a dose of 100mg to 1000mg, preferably 240mg to 560mg, more preferably 320mg to 480mg, once daily.
14. The use according to claim 13, wherein the dose of the compound of formula (I) or a pharmaceutically acceptable salt thereof is selected from the group consisting of 240mg, 320mg and 400mg; most preferably 400mg; the administration frequency was once daily.
15. The method of any one of claims 8 to 14The use of (1), wherein the dosage of the doxorubicin hydrochloride liposome injection is 20mg/m 2 ~60mg/m 2 (ii) a Preferably 30mg/m 2 ~50mg/m 2 (ii) a The frequency of administration was 1 time every 3 weeks.
16. Use according to any one of claims 8 to 14, wherein the epirubicin is administered at a dose of 60mg/m 2 ~120mg/m 2 (ii) a Preferably 75mg/m 2 ~100mg/m 2 (ii) a The frequency of administration was 1 time every 3 weeks.
17. The use according to any one of claims 8 to 14, wherein the dose of pirarubicin is 30mg/m 2 ~80mg/m 2 (ii) a Preferably 30mg/m 2 ~50mg/m 2 (ii) a The frequency of administration was 1 time every 3 weeks.
18. The use according to any one of claims 8 to 17, wherein the dose of cyclophosphamide is 400mg/m 2 ~1000mg/m 2 (ii) a Preferably 500mg/m 2 Or 600mg/m 2 (ii) a The frequency of administration was 1 time every 3 weeks.
19. The use according to any one of claims 8 to 18, wherein the dose of docetaxel is 60mg/m 2 ~120mg/m 2 (ii) a Preferably 75mg/m 2 ~100mg/m 2 (ii) a The frequency of administration was 1 time every 3 weeks.
20. The use according to any one of claims 8 to 18, wherein the dose of albumin paclitaxel is 60mg/m 2 ~140mg/m 2 (ii) a Preferably 80mg/m 2 ~120mg/m 2 (ii) a The frequency of administration was 1 time per week.
CN202210837289.7A 2021-07-15 2022-07-15 Application of tyrosine kinase inhibitor in combination with neoadjuvant chemotherapeutic drugs for treating tumors Pending CN115607548A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021108015128 2021-07-15
CN202110801512 2021-07-15

Publications (1)

Publication Number Publication Date
CN115607548A true CN115607548A (en) 2023-01-17

Family

ID=84857406

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210837289.7A Pending CN115607548A (en) 2021-07-15 2022-07-15 Application of tyrosine kinase inhibitor in combination with neoadjuvant chemotherapeutic drugs for treating tumors

Country Status (1)

Country Link
CN (1) CN115607548A (en)

Similar Documents

Publication Publication Date Title
RU2689779C2 (en) Duocarmicin adc showing improved anti-tumour activity in vivo
US12011449B2 (en) Therapeutic combinations comprising a c-RAF inhibitor
CN110494166B (en) Combination therapy
TW202038957A (en) Combination of antibody-drug conjugate and kinase inhibitor
TWI762784B (en) Use of cdk4/6 inhibitor in combination with egfr inhibitor for preparation of medicament for treating tumor diseases
WO2019109938A1 (en) Use of parp inhibitor in treating chemotherapy-resistant ovarian cancer or breast cancer
KR20200014298A (en) Treatment of HER2-positive cancer
JP2021505571A (en) Compositions and Methods for Treating Peripheral T-Cell Lymphoma and Cutaneous T-Cell Lymphoma
TW202133857A (en) Combination therapies for treatment of breast cancer
CN111184863A (en) Use of a combination of a tyrosine kinase inhibitor, a CDK4/6 inhibitor and a SERD for the preparation of a medicament for the treatment of tumours
WO2022111618A1 (en) Combined pharmaceutical composition of anti-pd-l1 antibody and c-met kinase inhibitor for treating lung cancer
CN115607548A (en) Application of tyrosine kinase inhibitor in combination with neoadjuvant chemotherapeutic drugs for treating tumors
WO2021057764A1 (en) Use of pd-1 antibody in combination with taxoid compound in preparation of drugs for treating triple-negative breast cancer
CN110585429B (en) Application of tyrosine kinase inhibitor combined with monoclonal antibody and taxol medicaments in treating tumor diseases
AU2005279344A1 (en) Use of midostaurin for treating gastrointestinal stromal tumors
US9566334B2 (en) Combinations of a PI3K/AKT inhibitor compound with an HER3/EGFR inhibitor compound and use thereof in the treatment of a hyperproliferative disorder
WO2018133838A1 (en) Uses of egfr/her2 inhibitor combined with pyrimidine-type anti-metabolic drug
JP2016008215A (en) Use of eribulin and s-1(or 5-fu) as combination therapy for cancer treatment
TWI822897B (en) Use of anti-pd-1 antibody in combination with famitinib for preparation of medicament for treating tumor diseases
CN114533886B (en) Pharmaceutical composition for treating advanced breast cancer
WO2024148307A1 (en) Raf inhibitor and erk1/2 and/or shp2 inhibitor combination therapy
WO2022234110A1 (en) Gamma-aminobutyric acid derivatives for use in cancer therapy
WO2023281413A1 (en) Methods and dosing regimens comprising pf-06873600 for the treatment of cancer
WO2024148291A1 (en) Raf inhibitor and kras g12c inhibitor combination therapy
CN113395969A (en) Application of CDK4/6 inhibitor-containing composition and anastrozole in preparation of drugs for treating tumor diseases

Legal Events

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