CN117338941A - Pharmaceutical composition containing TKIs inhibitor and SCD1 inhibitor and anti-tumor application thereof - Google Patents

Pharmaceutical composition containing TKIs inhibitor and SCD1 inhibitor and anti-tumor application thereof Download PDF

Info

Publication number
CN117338941A
CN117338941A CN202311239865.9A CN202311239865A CN117338941A CN 117338941 A CN117338941 A CN 117338941A CN 202311239865 A CN202311239865 A CN 202311239865A CN 117338941 A CN117338941 A CN 117338941A
Authority
CN
China
Prior art keywords
inhibitor
tkis
scd1
tumor
cancer
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
CN202311239865.9A
Other languages
Chinese (zh)
Inventor
丁志文
王鲁
董立巍
赵一鸣
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.)
Fudan University Shanghai Cancer Center
Original Assignee
Fudan University Shanghai Cancer Center
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 Fudan University Shanghai Cancer Center filed Critical Fudan University Shanghai Cancer Center
Priority to CN202311239865.9A priority Critical patent/CN117338941A/en
Publication of CN117338941A publication Critical patent/CN117338941A/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/45Non condensed piperidines, e.g. piperocaine having oxo groups directly attached to the heterocyclic ring, e.g. cycloheximide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/575Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids substituted in position 17 beta by a chain of three or more carbon atoms, e.g. cholane, cholestane, ergosterol, sitosterol
    • 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 invention relates to the technical field of medicines, in particular to application of a combination of a tyrosine kinase inhibitor and a stearoyl-CoA desaturase (SCD 1) inhibitor in malignant tumor treatment. Tyrosine kinase inhibitors are the first-line drug for systemic treatment of malignant tumors, and their resistance is the main cause affecting the effective rate of treatment. The invention provides a pharmaceutical composition for enhancing the curative effect of a tyrosine kinase inhibitor on treating liver malignant tumors, which comprises the tyrosine kinase inhibitor and an SCD1 inhibitor. After the tyrosine kinase inhibitor is combined with the SCD1 inhibitor, the proliferation of malignant tumor cells and the growth of xenogeneic planting tumors can be effectively inhibited. The invention can provide a sensitization individuation scheme for first-line treatment of malignant tumors by using tyrosine kinase inhibitors, and has better clinical application prospect.

Description

Pharmaceutical composition containing TKIs inhibitor and SCD1 inhibitor and anti-tumor application thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a medicine composition containing TKIs inhibitor and SCD1 inhibitor and an anti-tumor application thereof.
Background
Tyrosine kinase inhibitors (TKIs inhibitors) are a class of multi-kinase inhibitors that inhibit tyrosine kinase activity and inhibit cell proliferation. Various small molecule multi-kinase inhibitors for solid tumors have been developed clinically, including donafanib for treating liver cancer, sorafenib (Sorafenib) for treating liver cancer and kidney cancer, regorafenib for treating colorectal cancer, liver cancer, gastrointestinal stromal tumors, and the like. However, the effect of tyrosine kinase inhibitors on the treatment of tumors is not very desirable and the objective remission rate and overall survival rate of patients are not significantly improved. Taking a tyrosine kinase inhibitor for treating liver cancer as an example, SHARP research opens the way for treating liver cancer by using the tyrosine kinase inhibitor, but objective remission rate of Sorafenib (Sorafenib) which is a first-line medicine for treating liver cancer is only 12.4% (Llovet, J.M.et al N Engl J Med,359,378-390, doi:10.1056/NEJMoa 0708857). Mainly derived from drug resistance of liver cancer cells to tyrosine kinase inhibitors. New excision of the new generation of TKK was achieved by Duonani Donafinib (Qin, bi et al 2021) (Qin, S., F.Bi, S.Gu, et al, (2021), "Donafenib Versus Sorafenib in First-Line Treatment of Unresectable or Metastatic Hepatocellular Carcinoma: A random, open-Label, parallel-Controlled Phase II-III three," J Clin Oncol 39 (27): 3002-3011.DOI: 10.1200/JCO.21.00163), lenvartinib (Kudo, finn et al 2018) (Kudo, M., R.S.Finn, S.Qin, et al (2018), "Lenvatinib versus sorafenib in first-line treatment of patients with unresectable hepatocellular carcinoma: a randomised phase 3.3 non-input three," Lancet 391 (10126): 1163-1173.DOI: 10.1016/S0-6736 (18) 30207-1) so that the rate of HCC could not be increased gradually from 2% to 18.014. However, with the popularization of TKIs in clinical applications, the therapeutic effects thereof show increasingly obvious individuation differences, and at the same time, natural resistance or acquired resistance are unavoidable. TKIs resistance becomes a bottleneck limiting its clinical application, and how to solve and prevent resistance is also a key to the development of TKIs in the future.
At present, the combined application of TKIs and other target medicaments is probably a strategy for effectively overcoming TIKs resistance. The search for a drug combination which has high safety and can sensitize TKIs has important application value. The reprogramming of the lipid metabolism of tumor cells not only provides energy for uncontrolled growth of tumor cells, but also is involved in the therapeutic resistance of tumor cells. stearoyl-CoA desaturase (SCD) is the rate-limiting enzyme in the synthesis of monounsaturated fatty acids. It synthesizes monounsaturated fatty acyl coenzyme A by introducing a first double bond at cis-9 position of catalytic saturated fatty acyl coenzyme A. There are mainly two types of human tissue: SCD1 and SCD5, wherein SCD1 is widely expressed in adipose tissue, brain, liver, lung, heart, etc., SCD5 is limited in expression in adult tissues, principally limited to brain and pancreas (Ascenzi, de vitamins et al 2021) (Francesca Ascenzi, et al. (2021) "SCD1, autophagy and cancer: implications for therapy" J Exp Clin Cancer Res.2021Aug 24;40 (1): 265.doi:10.1186/s13046-021-02067-6. Studies have shown that SCD1 is capable of promoting tumor cell proliferation, tumor growth and metastasis. Whereas SCD1, which is highly expressed in tumors, is significantly associated with tumor invasion and poor prognosis. Furthermore, SCD1 is able to promote tumor cells to acquire or maintain dry characteristics, including chemotherapy resistance and self-renewal, etc., suggesting that SCD1 may be involved in tumor cell resistance.
At present, no curative effect research for combining lipid metabolism medicaments with TKIs treatment is known clinically.
Disclosure of Invention
In order to solve the problem that TKIs have natural drug resistance in middle and late malignant tumors, the invention provides the following technical scheme:
in a first aspect of the present invention, there is provided a pharmaceutical composition for enhancing anti-tumor effect of TKIs comprising a TKIs inhibitor and an SCD1 inhibitor;
further, in the pharmaceutical composition, the mass ratio of the TKIs inhibitor to the SCD1 inhibitor is 1:0.1-10; further, in the pharmaceutical composition, the mass ratio of the TKIs inhibitor to the SCD1 inhibitor is 1:1;
in a second aspect, the invention provides a pharmaceutical preparation for enhancing anti-tumor effect of TKIs, which consists of the pharmaceutical composition and a pharmaceutically acceptable carrier;
further, the carrier refers to a system for changing the mode of entering the human body, the distribution in the human body, the release speed and the like in the pharmaceutical field, and mainly comprises additives except active ingredients, wherein the additives are selected from any one or more of a filler, a coating agent, a sweetener, a colorant, a preservative and a propellant;
further, the filler is selected from one or more of starch, sucrose, lactose, calcium salt, sodium bicarbonate, citric acid and glycerol;
further, the coating agent is selected from any one or more of gelatin, gluten and mucilage;
further, the sweetener is selected from any one or more of sucrose, lactose and dextran;
further, the flavoring agent is selected from any one or more of natural spice and artificial spice;
further, the colorant is selected from any one or more of azo and monoazo dyes and triphenylmethane dyes;
further, the buffering agent is selected from any one or more of phosphate, borate and acetate;
further, the preservative is selected from any one or more of sorbic acid and benzyl alcohol;
further, the propellant is selected from any one or more of compressed gas and fluorocarbon;
further, the administration mode of the pharmaceutical preparation is selected from any one or more of oral administration, sublingual administration, rectal administration, skin mucosa administration, inhalation administration and injection administration; still further, the administration by injection is subcutaneous injection, intramuscular injection or intravenous drip;
further, the oral preparation is selected from any one or more of tablets, capsules, pills, granules, powder, inclusion compound, emulsion, syrup, sol, suspension, mixture and solution;
further, the injection is selected from any one or more of injection solution, injection emulsion, injection liposome, injection microcapsule, injection microsphere, injection water or oily suspension and injection nanoparticle;
further, the pharmaceutical formulation is a sustained or controlled release formulation; further, the slow release or controlled release preparation is a preparation for targeted release at a specific part; further, the slow release or controlled release preparation is selected from any one or more of tablets, coated tablets, capsules, micropills, suppositories and injections;
furthermore, the application amount of each medicine can be adjusted according to the administration route, the age and weight of a patient, the type and severity of the treated tumor and the like, and the daily dose of the medicine composition is 0.001-1000 mg/kg body weight; preferably, the daily dose of the pharmaceutical composition is 0.01-100 mg/kg body weight; more preferably, the daily dosage of the pharmaceutical composition of the invention is 0.1-50 mg/kg body weight; still further, the medicament is administered in a single or multiple administrations; further, the medicine is continuously administered or intermittently administered according to the course of treatment;
in a third aspect, the invention provides the pharmaceutical composition or the pharmaceutical preparation, and the application thereof in preparing antitumor drugs;
in a fourth aspect, the present invention provides a combination of an SCD1 inhibitor and a TKIs inhibitor for use in the manufacture of an anti-tumour medicament; further, the combination is the simultaneous or sequential administration of an effective amount of TKIs and SCD1 inhibitors; still further, the sequentially administering an effective amount of a TKIs and an SCD1 inhibitor is first inhibited with a TKIs, then an SCD1 inhibitor, or first an SCD1 inhibitor, then a TKIs inhibitor;
further, the SCD1 inhibitors are substances that inhibit SCD1 activity, including but not limited to: a939572, MK-8245, mf-438, cvt-11127,4-picolinic acid 2-phenylhydrazine (PluriSIn #1 (NSC 14613)), malic acid (steruicc acid), xen723,3β -arachidoylamino-7α,12α -dihydroxy-5β -cholan-24-oic acid (or (4 r) -4- ((3 s,5s,7r,10s,12s,13r,17 r) -7, 12-dihydroxy-3-eicosanamido-10, 13-dimethylhexadecyl-1 h-cyclopenta [ a ] phenanthren-17-yl) pentanoic acid (Aramchol), a gene expression vector shRNA that inhibits SCD1 function, a gene expression vector microRNA that inhibits SCD1 function, preferably, the SCD1 inhibitor is selected from a939572, MK-8245 or Aramchol;
further, the tyrosine kinase inhibitor is selected from any one or more of Sorafenib (Sorafenib, BAY 43-9006), dorafinib (Donafenib, CM-4307), lenvatinib (E7080), cabozantinib (BMS-907351), regorafenib (Regorafenib, BAY-734506), vatalanib (PTK 787), sunitinib (SU 11248), semaxanib (SU 5416), erlotinib (Erlotinib, OSI-774), lenvatinib (Lapatinib, GW-572016), fruquintinib (HMPL-013), anlotinib (AL 3818), axitinib (AG 013136), pazopanib (GW 786034), tivozanib (AV-951) or Apatinib (Apinib, YN968D 1); preferably, the tyrosine kinase inhibitor is selected from one or more of Sorafenib (BAY 43-9006), lenvantinib (E7080), regorafenib (BAY-734506) and Donafenib (CM-4307);
further, the composition or formulation includes SCD1 inhibitor a939572 or MK8245, and TKIs inhibitor Sorafenib;
further, the compositions or formulations are the SCD1 inhibitors Aramchol and the TKIs inhibitor Donafenib.
Further, the tumor is a solid tumor, and is selected from any one or more of hepatocellular carcinoma, cholangiocellular carcinoma, hepatic mixed cell carcinoma, gallbladder carcinoma, pancreatic cancer, ampulla malignant tumor, esophageal cancer, gastric cancer, colon cancer, rectal cancer, lung cancer, renal cancer, parathyroid cancer, thyroid cancer, lung cancer, ovarian cancer, cervical cancer or gastrointestinal stromal tumor; preferably, the tumor is selected from any one or more of hepatocellular carcinoma, cholangiocellular carcinoma, hepatic mixed cell carcinoma, gallbladder carcinoma, renal carcinoma, colon carcinoma, rectal cancer, parathyroid cancer or gastrointestinal stromal tumor; further preferably, the solid tumor p53 gene is sequenced to wild type;
further, the anti-tumor effect is that the SCD1 inhibitor has sensitization effect on the anti-tumor effect of the TKIs inhibitor; further, the anti-tumor effect is that the SCD1 inhibitor can increase the lipid peroxidation level of the tumor cells induced by TKIs and promote the death of the tumor cells caused by TKIs.
Drawings
Other features, objects and advantages of the present invention will become more apparent upon reading of the detailed description of non-limiting embodiments, given with reference to the accompanying drawings.
FIG. 1 TKIs inhibitors are capable of inducing iron death in tumor cells. (A) Cell viability of hepatoma cell lines (JHH 1, hepG2, hep3B and HUH 7) and colon cancer cell lines (HT 29 and HCT 116) at different concentrations of TKIs inhibitor (Sorafenib, sorafenib; lenvantinib, lenvatinib, regorafenib, regorafenib, apatinib, apatinib; erlotinib, erlotinib; donafanib, dorafinib), respectively. (B) Lipid peroxidation of liver cancer cell lines (JHH 1, hepG2, hep3B and HUH 7) and colon cancer cell lines (HT 29 and HCT 116) at different concentrations of TKIs inhibitor (Sorafenib, sorafenib; lenvantinib, lenvatinib, regorafenib, regrafenib, apatinib, apatinib; erlotinib, erlotinib; donafanib, dorafinib), respectively. (C) Clone formation experiments prove that the TKIs inhibitor Sorafneib has long-acting inhibition effect on liver cancer cell proliferation;
FIG. 2.TKIs inhibitors are capable of affecting the expression of SCD1 in tumor cells. (A) The venn plot shows that the transcriptome analysis was significantly different for 13 genes given Sorafenib treatment in HepG2, HUH7 and subcutaneous tumor-bearing models, respectively. (B) Schematic diagram of liver fatty acid synthesis from head, black font is metabolite, italic grey font is catalytic enzyme. (C) as a catalytic enzyme, SCD1 catalyzes a reaction scheme. (D) Protein expression levels of fatty acid anabolic enzymes in JHH1, hepG2 cells, with significant downregulation of SCD1 protein levels, were induced by iron death inducer RSL 3. (E) The TKIs inhibitor Sorafenib can obviously reduce the level of SCD1 protein in tumor cells.
Figure 3. Administration of therapeutic doses of SCD1 inhibitors did not affect tumor cell survival. (A) Tumor cells JHH1 and HepG2 were treated for 48 hours with different concentrations of the SCD1 inhibitor MK8245, respectively, and cell survival was examined by ATP. (B) Tumor cells JHH1 and HepG2 were treated with SCD1 inhibitor a939572 at different concentrations for 48 hours, respectively, and ATP was used to examine cell survival. (C) Tumor cells JHH1 and HepG2 were treated with the SCD1 inhibitor armchol at different concentrations for 48 hours, respectively, and ATP was used to examine cell survival.
FIG. 4 shows that the SCD1 inhibitor can promote the anti-tumor effect of TKIs inhibitor in human liver cancer cell line. (A) The SCD1 inhibitor (A939572, MK 8245) can obviously inhibit the survival of JHH1 and HepG2 cells in combination with Sorafenib, and the iron death inhibitor Ferrosin-1 can reverse the cell death induced by Sorafenib single drug or combination. ATP detects cell survival. (B) Plate cloning experiments prove that the SCD1 inhibitor (A939572, MK 8245) combined with Sorafenib can inhibit proliferation of JHH1 and HepG2 cells for a long time, and the iron death inhibitor Ferrosin-1 can reverse cell death induced by Sorafenib single drug or combined drug. (C) statistical histogram of FIG. 4B plate cloning experiments. (D) In JHH1 and HepG2 cells, SCD1 inhibitor (A939572, MK 8245) can obviously promote lipid peroxidation level induced by Sorafenib, and iron death inhibitor Ferrostatin-1 can inhibit lipid peroxidation level induced by Sorafenib singly or in combination. (E) Flag-SCD1 is overexpressed in JHH1 and HepG2 cells, and can inhibit the antitumor effect of Sorafenib.
FIG. 5 shows that the SCD1 inhibitor can promote the anti-tumor effect of TKIs inhibitor in human liver cancer organoids. (A) schematic diagram of liver cancer organoids construction. (B) In human liver cancer organoids 187, the SCD1 inhibitor Aramchol can significantly promote the anti-tumor effect of dorafinib Donafanib. (C) In human liver cancer organoids 188, the SCD1 inhibitor Aramchol can obviously promote the anti-tumor effect of Donafanib. (D) FIG. 5B, statistical histogram of C cell survival experiments. (E) In human liver cancer organoids 187, 188, the SCD1 inhibitor armchol was able to significantly promote Donafenib-induced lipid peroxidation levels.
Figure 6 in vivo studies show that SCD1 inhibitor alone has no significant effect of inhibiting tumor growth. (A) Schematic diagrams of subcutaneous tumor-bearing models of HepG2 cell nude mice (different doses of armchol) were constructed. (B) different doses of Aramchol had no significant effect on nude mice body weight. (C) Different doses of Aramchol had no significant effect on the subcutaneous tumor weight of nude mice. (D) Subcutaneous tumor images of nude mice after treatment with different doses of armchol. (E) Different doses of Aramchol had no obvious effect on the subcutaneous tumor volume of nude mice. (F-H) different doses of Aramchol have no obvious effect on the blood glutamic pyruvic transaminase ALT (F), the blood glutamic oxaloacetic transaminase AST (G) and the blood triglyceride TG (H) of nude mice. (I) After treatment with different doses of armchol, liver, spleen, kidney, lung and heart images of nude mice with tumors were obtained subcutaneously. (J) Different doses of Aramchol have no obvious effect on the weight of the important organs of the nude mice. (K) After treatment with different doses of armchol, subcutaneous tumor-bearing nude mice were HE stained with liver, spleen, kidney, lung, heart paraffin sections.
Fig. 7 shows that SCD1 inhibitors are able to promote the anti-tumor effect of TKIs inhibitors in vivo. (A) Schematic of HepG2 cell nude mice subcutaneous tumor-bearing model (Donafenib single drug or Donafenib combined Aramchol administration) was constructed. (B) Subcutaneous tumor images of nude mice following Donafenib alone or in combination with armchol. (C) The SCD1 inhibitor Aramchol combined with TKIs inhibitor Aramchol can obviously inhibit the weight of tumor. (D) The SCD1 inhibitor Aramchol combined with TKIs inhibitor Aramchol can obviously inhibit tumor volume. (E) SCD1, ki67 immunohistochemical images in nude mice subcutaneous tumors following Donafenib single or Donafenib combined with Aramchol. (F) The SCD1 inhibitor Aramchol combined with TKIs inhibitor Aramchol can obviously inhibit the expression of SCD1 in tumors. (G) The SCD1 inhibitor Aramchol in combination with TKIs inhibitor Aramchol can significantly inhibit Ki67 expression in tumors.
Advantageous effects
The TKIs inhibitor and SCD1 inhibitor composition provided by the invention can obviously inhibit the growth of malignant tumors, effectively sensitize the treatment effect of TKIs, improve the drug resistance of TKIs and is easy to clinically use and popularize.
Detailed Description
In order to make the objects, technical solutions and advantages of the embodiments of the present invention more clear, the technical solutions of the embodiments of the present invention will be clearly and completely described below with reference to the accompanying drawings of the embodiments of the present invention. It will be apparent that the described embodiments are some, but not all, embodiments of the invention. All other embodiments, which can be made by a person skilled in the art without creative efforts, based on the described embodiments of the present invention fall within the protection scope of the present invention.
In the invention, the tyrosine kinase inhibitor Sorafenib, lenvatinib, regorafenib, apatinib, erlotinib, donafenib is purchased from Selleck, and the SCD1 inhibitors MK8245, A939572 and Aramchol are purchased from ABclonal; iron death inhibitor Ferrositin-1 was purchased from Selleck. CellTiter-Glo used to measure cell viability was purchased from Promega; cell counting-Lite 3D Luminescent Cell Viability Assay for detecting organoid cell viability was purchased from Vazyme; the reagent C11-BODIPY for detecting lipid peroxidation is purchased from ABclonal; the reagent used for detecting lipid peroxidation, liperflo, is available from Dojindo; the Flag-SCD1 plasmid was purchased from LoGENBIO.
Antibodies used by Western Blot in the invention, ACC was purchased from CST, cat# 3676; FASN is available from abclon, cat No. a19050; SCD1 was purchased from ABclonal, cat No. A16429; FADS2 is available from ABclonal under the designation A10270; GCLM is available from ABclonal under the designation A11444; GCLC was purchased from ABclonal under the trade designation A4499; GPX4 is available from ABclonal under the designation A11243; SLC7A11 is purchased from Proteintech under accession number 26864-1-AP; GAPDH was purchased from abclon, cat No. AC001.
In the invention, liver cancer cell lines HepG2, hep3B, HUH7, HCT116 and HT29 are purchased from cell line institute of Chinese sciences, and JHH1 is purchased from Hangzhou Qiano biology company.
Example 1: tyrosine kinase inhibitors are capable of inducing pig death in tumor cells.
Cell viability detection: effect of different concentrations of tyrosine kinase inhibitors on tumor cell survival. Tumor cells (hepatoma cells JHH1, hepG2, hep3B, HUH7; colon cancer cells: HT29, HCT 116) were transferred into 96-well plates, 1X 105 cells per well, respectively. Each tumor cell was divided into 4 groups of 3 replicates each.
(1) Sorafenib concentration gradient of0,5μmol/L,10μmol/L,20μmol/L;
(2) The concentration gradient of Lenretinib is 0,5 mu mol/L,10 mu mol/L and 20 mu mol/L;
(3) The concentration gradient of the Regorafinib is 0,5 mu mol/L,10 mu mol/L and 20 mu mol/L;
(4) Apatinib concentration gradient was 0, 5. Mu. Mol/L, 10. Mu. Mol/L, 20. Mu. Mol/L;
(5) The concentration gradient of the Erlotinib is 0,5 mu mol/L,10 mu mol/L and 20 mu mol/L;
(6) Donafenib, JHH1, hepG2, HUH7, HT29 cells, concentration gradient 0, 5. Mu. Mol/L, 10. Mu. Mol/L, 20. Mu. Mol/L; hep3B and HCT116 cells were present at a concentration gradient of 0, 1. Mu. Mol/L, 2. Mu. Mol/L, 5. Mu. Mol/L.
After 48 hours of drug stimulation, the medium was discarded, serum-free medium containing 50% CellTiter-Glo reagent was added, incubated for 25 minutes at 37℃in a cell incubator, and intracellular ATP content was measured using an enzyme-labeled instrument to determine cell viability. Each of the above tumor cell viability assay experiments was repeated 3 times. As shown in fig. 1A, the tyrosine kinase inhibitor is shown in the following formula20μmol/LCan inhibit the survival of tumor cells to a certain extent.
Lipid peroxidation assay: effect of different concentrations of tyrosine kinase inhibitors on the peroxidized lipid level of tumor cells. Tumor cells (hepatoma cells JHH1, hepG2, hep3B, HUH; colon cancer cells: HT29, HCT 116) were respectively transferred into 6-well plates, and after the cells had adhered to the walls, each cell was divided into 7 groups, each group was replaced with a medium containing the following drugs or tyrosine kinase inhibitors, respectively:
(1)DMSO;
(2)Sorafenib 10μmol/L;
(3) Lenretinib, JHH1, hepG2, hep3B cells 10. Mu. Mol/L, HUH7, HT29, HCT116 cells 20. Mu. Mol/L;
(4) Regorafenib, hepG2 cells 5. Mu. Mol/L, JHH1, hep3B, HUH7, HT29, HCT116 cells 10. Mu. Mol/L;
(5)Apatinib 20μmol/L;
(6)Erlotinib 20μmol/L;
(7) Donafinib, hep3B, HCT116 cells 5. Mu. Mol/L, JHH1, hepG2, HUH7, HT29 cells 10. Mu. Mol/L.
After 48 hours of drug treatment, the adherent cells were digested, washed once with PBS, resuspended in a 1.5 ml centrifuge tube, the supernatant was centrifuged off, stained with 20mmol/L C-BODIPY, incubated in a 37℃cell incubator for 1 hour, detected by flow cytometry, at a wavelength greater than 580 nm of non-peroxidized C11, and at a wavelength of 505-550 nm of oxidized C11. The above experiment was repeated 3 times. As shown in fig. 1B, tyrosine kinase inhibitors can promote lipid peroxidation levels in tumor cells to some extent.
Plate cloning experiments: the adherent JHH1 and HepG2 cells were digested and resuspended after log phase growth, 5X 103 was transferred to 6-well plate, and after cell adherence, DMEM medium containing the following drugs was added: DMSO or Sorafenib 2.5. Mu. Mol/L. The culture medium with the medicine is changed once in 2-3 days, and the culture is carried out for 14 days. After pre-cooling the PBS and fixing the cells for 10 minutes with 4% paraformaldehyde, washing 1 time with PBS, staining with crystal violet for 15 minutes, washing 2 times with PBS, and photographing under an optical microscope. Plate cloning experiments were repeated 3 times and the photographed pictures were analyzed by Image J software to count the number of cell colonies. As shown in fig. 1C, tyrosine kinase inhibitors can inhibit proliferation of tumor cells for a certain period of time.
Example 2: tyrosine kinase inhibitors are capable of inhibiting the tumor cell fatty acid metabolizing enzyme SCD1.
Analyzing the data GSE96793, GSE96794 and GSE121153 in the GEO public database, selecting the differentially expressed genes in each data set, taking the intersection of the differentially expressed genes as a Venn diagram, and recruiting 13 genes as shown in FIG. 2A, wherein the genes are respectively: lyz, tpp1, sema6a, slc35b3, scd1, cthrc1, tsc22d3, timp1, serrine2, kdm4d, plac8, wipi1, zfyve26. Wherein the Scd1 gene is transcribed and translated to form a protein Scd1 which is a fatty acid metabolism-related metabolic enzyme. FIG. 2B is a schematic diagram of the de novo synthesis of intracellular fatty acids, the metabolizing enzyme SCD1 being capable of catalyzing the synthesis of monounsaturated fatty acids from saturated fatty acids during fatty acid synthesis. As shown in FIG. 2C, SCD1 catalyzes primarily palmitic acid (C16:0) to palmitoleic acid (C16:1) and stearic acid (C18:0) to oleic acid (C18:1).
Western Blot detects protein levels of fatty acid anabolism-related enzymes. Liver cancer cells JHH1 and HepG2 are respectively transferred into a 6-well plate, after the cells grow to 90% confluence, DMSO or an iron death inducer RSL3 mu mol/L is added, the cells are respectively cultured for 12 hours or 24 hours, 100 microliter of Western and IP cell lysate (Biyun, P0013) is used for collecting proteins, the proteins are collected after ultrasound for 12000 revolutions and centrifuged for 20 minutes, SDS high-temperature denaturation is added after quantification, 40 microgram of proteins are taken, after SDS-PAGE electrophoresis, membrane transfer, 5% BSA is used for sealing, and Odyssey scanning imaging is carried out after primary antibody incubation. As shown in fig. 2D, iron death inducer RSL3 was able to inhibit SCD1 protein levels in tumor cells. Liver cancer cells JHH1 and HepG2 are respectively transferred into a 6-well plate, after the cells grow to 90% confluence, DMSO or Sorafenib 20 mu mol/L is added, and the cells are respectively cultured for 12 hours or 24 hours, and the protein level is detected under the same Western Blot conditions. As shown in fig. 2E, sorafenib was able to inhibit SCD1 protein levels in tumor cells.
Example 3: SCD1 inhibitors alone do not significantly inhibit tumor cell survival.
Cell viability detection: effect of SCD1 inhibitors at different concentrations on tumor cell survival. Tumor cells JHH1, hepG2 or HCT116 were each transferred into 96-well plates, 1×105 cells per well. Each tumor cell was divided into 7 groups of 3 replicates each, each group was replaced with medium containing either the following drugs or tyrosine kinase inhibitors:
(1) MK8245 concentration gradients of 0,0.1. Mu. Mol/L, 1. Mu. Mol/L, 5. Mu. Mol/L, 10. Mu. Mol/L, 20. Mu. Mol/L, 50. Mu. Mol/L;
(2) A939572 has a concentration gradient of 0,0.1. Mu. Mol/L, 1. Mu. Mol/L, 5. Mu. Mol/L, 10. Mu. Mol/L, 20. Mu. Mol/L, 50. Mu. Mol/L;
(3) (3) Aramchol concentration gradient was 0,0.1. Mu. Mol/L, 1. Mu. Mol/L, 5. Mu. Mol/L, 10. Mu. Mol/L, 20. Mu. Mol/L.
48 hours after changing to the drug-containing medium, the medium was discarded, a serum-free medium containing 50% CellTiter-Glo reagent was added, and the cells were incubated in a 37℃cell incubator for 25 minutes, and the intracellular ATP content was detected using an enzyme-labeled instrument to determine the cell viability. Each of the above tumor cell viability assay experiments was repeated 3 times. As shown in fig. 3A, SCD1 inhibitor MK8245 alone did not significantly inhibit tumor cell survival. As shown in fig. 3B, SCD1 inhibitor a939572 alone failed to significantly inhibit tumor cell survival. As shown in fig. 3C, SCD1 inhibitor MK8245 alone did not significantly inhibit tumor cell survival.
Example 4: in vitro cell line experiments prove that the tyrosine kinase inhibitor combined with the SCD1 inhibitor can obviously inhibit the survival of tumor cells.
The tyrosine kinase inhibitor and the SCD1 inhibitor obviously inhibit the activity of tumor cells, and the tumor cells JHH1 or HepG2 are respectively transferred into 96-well plates, wherein each well is 1 multiplied by 105 cells. Each tumor cell was divided into 7 groups of 3 replicates each, each group was replaced with medium containing either the following drugs or tyrosine kinase inhibitors:
(1)DMSO;
(2)Sorafenib 10μmol/L;
(3)Sorafenib 10μmol/L,Ferrostatin-1 20μmol/L;
(4)Sorafenib 10μmol/L,A929572 10μmol/L;
(5)Sorafenib 10μmol/L,A929572 10μmol/L,Ferrostatin-1 20μmol/L;
(6)Sorafenib 10μmol/L,MK8245 10μmol/L;
(7)Sorafenib 10μmol/L,MK8245 10μmol/L,Ferrostatin-1 20μmol/L;
after 48 hours from the replacement of the culture medium containing the drug, the culture medium was discarded, and the intracellular ATP content was detected by an ELISA reader to determine the cell viability, as in the cell viability assay described above. Each of the above tumor cell viability assay experiments was repeated 3 times. As shown in FIG. 4A, the SCD1 inhibitor significantly promoted the anti-tumor effect of the tyrosine kinase inhibitor, and the iron death inhibitor Ferrostatin-1 was able to reverse cell death caused by combination therapy.
The tyrosine kinase inhibitor combined with SCD1 inhibitor can inhibit tumor cell proliferation for a long period, and the adherent JHH1 and HepG2 cells are subjected to digestion and resuspension after growth log phase, and 5×10 is taken 3 After cells attach to 6-well plates, DMEM medium containing the following drugs was added:
(1)DMSO;
(2)Sorafenib 2.5μmol/L;
(3)Sorafenib 2.5μmol/L,Ferrostatin-1 5μmol/L;
(4)Sorafenib 2.5μmol/L,A929572 2.5μmol/L;
(5)Sorafenib 2.5μmol/L,A929572 2.5μmol/L,Ferrostatin-1 5μmol/L;
(6)Sorafenib 2.5μmol/L,MK8245 2.5μmol/L;
(7)Sorafenib 2.5μmol/L,MK8245 2.5μmol/L,Ferrostatin-1 5μmol/L;
the culture medium with the medicine is changed once in 2-3 days, and the culture is carried out for 14 days. The steps of fixing, staining, observing and photographing by an optical microscope are the same as the experiment, repeating for 3 times, and analyzing and counting the number of cell colonies by using Image J software through photographed pictures. As shown in fig. 4B-C, SCD1 inhibitor can significantly promote the long-acting anti-tumor effect of tyrosine kinase inhibitor, and Ferrostatin-1 inhibitor can inhibit the anti-tumor effect of combination therapy.
Liperflo assay detects lipid peroxidation levels in tumor cells: tumor cells JHH1 or HepG2 were respectively transferred into 6-well plates, and after the cells had adhered to the walls and grown to a density of 90%, each cell was divided into 7 groups of 3 replicates each, each group was replaced with a medium containing the following drugs or tyrosine kinase inhibitors, respectively:
(1)DMSO;
(2)Sorafenib 10μmol/L;
(3)Sorafenib 10μmol/L,Ferrostatin-1 20μmol/L;
(4)Sorafenib 10μmol/L,A929572 10μmol/L;
(5)Sorafenib 10μmol/L,A929572 10μmol/L,Ferrostatin-1 20μmol/L;
(6)Sorafenib 10μmol/L,MK8245 10μmol/L;
(7)Sorafenib 10μmol/L,MK8245 10μmol/L,Ferrostatin-1 20μmol/L;
cells were treated with drug for 48h, incubated with 5. Mu.M Liperfluo (syn chemical L248) for 30min at 37℃and then flow cytometry was performed, with excitation light at 488nm and emission light at 550nm. As shown in fig. 4D, SCD1 inhibitors were able to significantly increase tyrosine kinase inhibitor-induced lipid peroxidation levels of tumor cells.
JHH1 or HepG2 was transferred into 6-well plates, respectively, and after the cells adhered to the walls and the growth density reached 30%, each cell was divided into 4 groups, and the PC3.1A plasmid was transferred from outside in groups 1 and 2, the Flag-SCD1 plasmid was transferred from outside in groups 3 and 4, and the transfection reagent was Lipo8000. DMSO was added to groups 1 and 3, and Sorafenib 20. Mu. Mol/L was added to group 2. After 48 hours of drug treatment, the medium was discarded, serum-free medium containing 50% CellTiter-Glo reagent was added, incubated at 37℃for 20 minutes in a cell incubator, and intracellular ATP content was measured using an enzyme-labeled instrument to determine cell viability. The above experiment was repeated 3 times. As shown in fig. 4E, exogenous supplementation with SCD1 can inhibit the anti-tumor effect of tyrosine kinase inhibitors.
Example 5: in vitro organoid experiments prove that the tyrosine kinase inhibitor combined with the SCD1 inhibitor can obviously inhibit the survival of tumor cells.
Human liver cancer organoids are constructed: fresh human liver cancer tissue samples were minced and incubated in digestion buffer (DMEM (Gibco) containing 0.1mg/mL DNase I (Sigma), 4mg/mL collagenase D (Roche), 2X 10-6M Y27632 (Sigma-Aldrich), 100. Mu.g/mL Primocin (InvivoGen)) at 37℃for 30-90 min. The suspension was filtered through a cell filter (100 μm) and centrifuged at 1000rpm for 5 minutes. Washed twice with pre-chilled DMEM/F12 (GIBCO, usa) and then mixed with Matrigel (BD Transduction Laboratories). 150. Mu.L of cold Matrigel was mixed with 200. Mu.L of the cell suspension and placed in a 6-well suspension plate for 30 minutes at 37 ℃. After gelation, 2mL of medium was added to each well. The organoid medium contained advanced DMEM/F12,1% penicillin/streptomycin, 1% GlutaMAX-I, 100 μg/mL Primocin, 10×10-3M HEPES, 1:50B27 supplement (without vitamin A), 1.25X10-3-M N-acetyl-l-cysteine,1ng/mL recombinant human FGF-basic,50ng/mL mouse recombinant EGF,100ng/mL recombinant human FGF10,10×10-6M forskolin, 25ng/mL recombinant human HGF, 5×10-6M A8301, 10×10-6M Y27632, 10% (volume fraction) Rspo-1 conditioned medium, 30% (volume fraction) Wnt3a conditioned medium, and 5% (volume fraction) Noggin conditioned medium. As shown in FIG. 5A, two p53 wild type human liver cancer organoids, HCC-187PDO and HCC-188PDO, were constructed together.
6000 cells were seeded in 96-well suspension culture plates, organoids were observed under the mirror in real time, and after organoids were formed, each organoid was divided into 4 groups, each group was replaced with a medium containing the following drugs or tyrosine kinase inhibitors, respectively:
(1)DMSO;
(2)Aramchol 10μmol/L;
(3)Donafenib 10μmol/L;
(4)Donafenib 10μmol/L,Aramchol 10μmol/L。
cells were treated with drug for 5d and organoid morphology and organoid numbers were observed under an optical microscope. As shown in fig. 5B-C, SCD1 inhibitors significantly promoted the antitumor effect of tyrosine kinase inhibitors.
6000 cells were seeded in 96-well suspension culture plates, organoids were observed under the mirror in real time, and after organoids were formed, each organoid was divided into 4 groups, each group was replaced with a medium containing the following drugs or tyrosine kinase inhibitors, respectively:
(1)DMSO;
(2)Aramchol 10μmol/L;
(3)Donafenib 10μmol/L;
(4)Donafenib 10μmol/L,Aramchol 10μmol/L。
cells were treated with drug for 5D, medium was discarded, serum-free medium containing 50% CellCounting-Lite 3D reagent was added, incubated for 25 min at 37 degrees celsius in a cell incubator, and intracellular ATP content was detected using an enzyme-labeled instrument to determine cell viability. The above experiment was repeated 3 times. As shown in fig. 5D, SCD1 inhibitors significantly promoted the antitumor effect of tyrosine kinase inhibitors.
The growth of organoids was observed under the mirror in real time by seeding 60000 cells in 6-well suspension plates, and after organoids were formed, each organoid was divided into 4 groups, each group was replaced with medium containing the following drugs or tyrosine kinase inhibitors, respectively:
(1)DMSO;
(2)Aramchol 10μmol/L;
(3)Donafenib 10μmol/L;
(4)Donafenib 10μmol/L,Aramchol 10μmol/L。
cells were treated with drug for 5d, medium was discarded, and intracellular MDA levels were detected using the MDA Assay Kit (Dojindo, M496). As shown in fig. 5E, SCD1 inhibitors significantly promoted tyrosine kinase inhibitor-induced lipid peroxidation levels of tumor cells.
Example 6: the experiment of tumor-bearing under the body endothelium shows that the single SCD1 inhibitor has no obvious effect of inhibiting the growth of tumor.
Constructing a nude mouse subcutaneous tumor-bearing model, and evaluating the safety of an SCD1 inhibitor Aramchol in vivo: 1X 107HepG2 cells were resuspended and then planted subcutaneously in hind limbs of 6-week-old male nude mice, 15 nude mice were added, and the mice were randomly divided into 3 groups after day 7, each group being given the following drugs:
(1) Peanut oil, 50 μl, intraperitoneally injected, 1/day;
(2) Aramchol,5mg/kg, intraperitoneal injection, 1/day;
(3) Aramchol,10mg/kg, i.p. injection, 1/day.
Mice were sacrificed by day 28 following continuous dosing, blood samples, tissue samples, etc. were left. Fig. 6A is a schematic diagram showing the construction of a nude mouse subcutaneous tumor-bearing model. And statistically analyzing the weight, the weight of the planted tumor, the size, the serum biochemical index, the important visceral conditions and the like of the nude mice. As shown in fig. 6B, the SCD1 inhibitor armchol did not affect the body weight of subcutaneous tumor-bearing nude mice. As shown in fig. 6C, the SCD1 inhibitor armchol alone did not affect the weight of subcutaneous tumors. Fig. 6D shows images of groups of subcutaneous tumors. As shown in fig. 6E, the SCD1 inhibitor armchol alone did not affect the volume of subcutaneous tumor. As shown in FIGS. 6F-H, the SCD1 inhibitor Aramchol did not affect blood ALT, blood TG levels in subcutaneous tumor-bearing nude mice, but was able to reduce blood AST levels. FIG. 6I is a photograph showing the major viscera of each group of subcutaneous tumor-bearing nude mice. As shown in FIG. 6J, the SCD1 inhibitor Aramchol did not affect the weight of the important organs of the subcutaneous tumor-bearing nude mice. As shown in fig. 6K, the important organs of each group of subcutaneous tumor-bearing nude mice were stained with hematoxylin-eosin (HE) after paraffin fixation and slicing, and no significant difference was observed under a microscope.
Example 7: the tumor-bearing experiment under the body endothelium proves that the tyrosine kinase inhibitor and the SCD1 inhibitor can obviously inhibit the growth of tumors.
Constructing a nude mouse subcutaneous tumor-bearing model, and evaluating the anti-tumor effect of an SCD1 inhibitor Aramchol combined with a tyrosine kinase inhibitor Donafenib: will be 1X 10 7 HepG2 cells were resuspended and then planted subcutaneously in the hind limbs of 6-week-old male nude mice, 15 nude mice in total, and randomly divided into 3 groups after day 7, each group being given the following drugs:
(1) Peanut oil, 50 μl, intraperitoneal injection, 1/day, physiological saline, 100 μl, intragastric administration, 1/day;
(2) Peanut oil, 50 μl, intraperitoneal injection, 1/day, donafenib,10mg/kg, lavage, 1/day;
(3) Aramchol,10mg/kg, intraperitoneal injection, 1/day, donafanib, 10mg/kg, gastric lavage, 1/day.
Mice were sacrificed by day 28 following continuous dosing, blood samples, tissue samples, etc. were left. Fig. 7A is a schematic diagram showing the construction of a nude mouse subcutaneous tumor-bearing model. And statistically analyzing the weight, the weight of the planted tumor, the size, the serum biochemical index, the important visceral conditions and the like of the nude mice. Fig. 7B shows images of groups of subcutaneous tumors. FIG. 7C shows that the SCD1 inhibitor Aramchol in combination with the tyrosine kinase inhibitor Donafenib significantly reduced the weight of subcutaneous tumors. FIG. 7D shows that the SCD1 inhibitor Aramchol in combination with the tyrosine kinase inhibitor Donafenib significantly reduced the volume of subcutaneous tumors. As shown in FIGS. 7E-G, immunohistochemical detection of SCD1, ki67 expression in each group of subcutaneous tumors was performed as follows: endogenous peroxidase is inactivated by 3% hydrogen peroxide, nonspecific signals are blocked by 1% BSA, and an SCD1 antibody and a Ki67 antibody are respectively used as the primary antibody. The SCD1 inhibitor Aramchol in combination with the tyrosine kinase inhibitor Donafenib can significantly reduce the expression of SCD1 and Ki67 in subcutaneous tumors.
Furthermore, it should be understood that although the present disclosure describes embodiments, not every embodiment is provided with a separate embodiment, and the description is provided for clarity only, and those skilled in the art will recognize that the embodiments of the disclosure may be combined appropriately to form other embodiments that will be understood by those skilled in the art.

Claims (16)

1. A pharmaceutical composition for enhancing anti-tumor effect of TKIs, which is characterized by comprising a TKIs inhibitor and an SCD1 inhibitor; further, in the pharmaceutical composition, the mass ratio of the TKIs inhibitor to the SCD1 inhibitor is 1:0.1-10; further, in the pharmaceutical composition, the mass ratio of the TKIs inhibitor to the SCD1 inhibitor is 1:1.
2. The pharmaceutical composition of claim 1, wherein the SCD1 inhibitor is a substance that inhibits SCD1 activity, including but not limited to: a939572, MK-8245, MF-438, CVT-11127,4-picolinic acid 2-phenylhydrazine (PluriSIn#1 (NSC 14613)), malic acid (Sterculic acid), XEN723,3β -arachidyl-7α,12α -dihydroxy-5β -cholan-24-oic acid (or (4 r) -4- ((3 s,5s,7r,10s,12s,13r,17 r) -7, 12-dihydroxy-3-eicosanamido-10, 13-dimethylhexadecyl-1 h-cyclopenta [ a ] phenanthren-17-yl) pentanoic acid (Aramchol), a gene expression vector shRNA that inhibits SCD1 function, a gene expression vector microRNA that inhibits SCD1 function, preferably the SCD1 inhibitor is selected from A939572, MK-8245 or Aramchol.
3. The pharmaceutical composition according to claim 1, wherein the TKIs inhibitor is selected from Sorafenib (Sorafenib, BAY 43-9006), dorafinib (donafinib, CM-4307), lenvatinib (Lenvatinib, E7080), cabozantinib (Cabozantinib, BMS-907351), regorafenib (Regorafenib, BAY-734506), watanib (Vatalanib, PTK 787), sunitinib (Sunitinib, SU 11248), semaxanib (Semaxanib, SU 5416), erlotinib (Erlotinib, OSI-774), lenvatinib (Lapatinib, GW-57578), furatinib (fluquintinib, HMPL-013), an Luoti ni (anlatinb, AL 3818), acitinib (Axitinib, 013136), pazotinib (AG) or one of the several types of papanib, panatib (Apatinib, ap 951-96D, ap 1 or any of the other types; preferably, the tyrosine kinase inhibitor is selected from one or more of Sorafenib (Sorafenib, BAY 43-9006), lenvatinib (Lenvatinib, E7080), regorafenib (Regorafenib, BAY-734506) and dorafinib (Donafenib, CM-4307).
4. A pharmaceutical composition according to any one of claims 1 to 3, wherein the composition comprises the SCD1 inhibitor a939572, MK8245 or 3β -arachidylamino-7α,12α -dihydroxy-5β -cholan-24-oic acid (or (4 r) -4- ((3 s,5s,7r,10s,12s,13r,17 r) -7, 12-dihydroxy-3-eicosanamido-10, 13-dimethylhexadecyl-1 h-cyclopenta [ a ] phenanthren-17-yl) pentanoic acid (Aramchol), and the TKIs inhibitor Sorafenib (Sorafenib), dorafenib (Donafenib) or Regorafenib (Regorafenib).
5. The pharmaceutical composition according to claim 1, wherein the tumor is a solid tumor selected from any one or more of hepatocellular carcinoma, cholangiocellular carcinoma, hepatocellular carcinoma, gallbladder carcinoma, pancreatic cancer, ampulla malignancy, esophageal cancer, gastric cancer, colon cancer, rectal cancer, lung cancer, renal cancer, parathyroid cancer, thyroid cancer, lung cancer, ovarian cancer, cervical cancer, or gastrointestinal stromal tumor; preferably, the tumor is selected from any one or more of hepatocellular carcinoma, cholangiocellular carcinoma, hepatic mixed cell carcinoma, gallbladder carcinoma, renal carcinoma, colon carcinoma, rectal cancer, parathyroid cancer or gastrointestinal stromal tumor; further preferably, the solid tumor p53 gene is sequenced to wild type.
6. The pharmaceutical composition according to claim 5, wherein the anti-tumor effect is that the SCD1 inhibitor has a sensitization effect on the anti-tumor effect of the TKIs inhibitor; further, the anti-tumor effect is that the SCD1 inhibitor can increase the lipid peroxidation level of the tumor cells induced by TKIs and promote the death of the tumor cells caused by TKIs.
7. A pharmaceutical preparation for enhancing anti-tumor effect of TKIs, which is characterized by comprising the pharmaceutical composition of claims 1-6 and a pharmaceutically acceptable carrier.
8. The pharmaceutical preparation for enhancing anti-tumor effect of TKIs according to claim 7, wherein the carrier is a system for changing the mode of drug entering human body, distribution in human body, release rate, etc. in the pharmaceutical field, and is mainly additive other than active ingredient, selected from any one or more of filler, coating agent, sweetener, colorant, antiseptic, and propellant;
further, the filler is selected from any one or more of starch, sucrose, lactose, calcium salt, sodium bicarbonate, citric acid and glycerol;
further, the coating agent is selected from any one or more of gelatin, gluten and mucilage;
further, the sweetener is selected from any one or more of sucrose, lactose and dextran;
further, the flavoring agent is selected from any one or more of natural spice and artificial spice;
further, the colorant is selected from any one or more of azo and monoazo dyes and triphenylmethane dyes;
further, the buffer is selected from any one or more of phosphate, borate and acetate;
further, the preservative is selected from any one or more of sorbic acid and benzyl alcohol;
further, the propellant is selected from any one or more of compressed gas and fluorocarbon.
9. The pharmaceutical preparation for enhancing anti-tumor effect of TKIs according to claim 7 or 8, wherein the administration mode of the pharmaceutical preparation is selected from any one or more of oral administration, sublingual administration, rectal administration, skin mucosa administration, inhalation administration, injection administration; further, the administration by injection is subcutaneous injection, intramuscular injection or intravenous drip.
10. The pharmaceutical preparation for enhancing anti-tumor effect of TKIs according to claim 9, wherein the preparation is an oral preparation or an injection; further, the oral preparation is selected from any one or more of tablets, capsules, pills, granules, powder, inclusion compound, emulsion, syrup, sol, suspension, mixture and solution; further, the injection is selected from any one or more of injection solution, injection emulsion, injection liposome, injection microcapsule, injection microsphere, injection water or oily suspension and injection nanoparticle.
11. The pharmaceutical preparation for enhancing an antitumor effect of TKIs according to claim 7, 8 or 10, wherein the pharmaceutical preparation is a sustained-release or controlled-release preparation; further, the slow release or controlled release preparation is a preparation for targeted release at a specific part; further, the slow release or controlled release preparation is selected from any one or more of tablets, coated tablets, capsules, micropills, suppositories and injections.
12. The pharmaceutical preparation for enhancing anti-tumor effect of TKIs according to claim 7, 8 or 10, wherein the administration amount of each drug is adjusted according to the administration route, age, body weight of the patient, kind or severity of tumor to be treated, and the daily dose of the pharmaceutical preparation is 0.001 to 1000mg/kg body weight based on the pharmaceutical composition; preferably, the daily dose is 0.01-100 mg/kg body weight; more preferably, the daily dose is 0.1 to 50mg/kg body weight; still further, the pharmaceutical formulation is administered in a single or multiple administrations; still further, the pharmaceutical formulation is administered continuously or intermittently according to a course of treatment.
13. Use of a pharmaceutical composition according to any one of claims 1 to 6 or a pharmaceutical formulation according to any one of claims 7 to 12 for the preparation of an anti-tumour agent.
14. The use according to claim 13, wherein the anti-tumor effect is that the SCD1 inhibitor has a sensitization effect on the anti-tumor effect of the TKIs inhibitor; further, the anti-tumor effect is that the SCD1 inhibitor can increase the lipid peroxidation level of the tumor cells induced by TKIs and promote the death of the tumor cells caused by TKIs.
Use of a combination of an scd1 inhibitor and a TKIs inhibitor for the preparation of an anti-tumor medicament; further, in the use of the TKIs inhibitor in combination with an SCD1 inhibitor, the TKIs inhibitor: the mass ratio of the SCD1 inhibitor is 1:0.1-10; still further, in the use of the TKIs inhibitor in combination with an SCD1 inhibitor, the TKIs inhibitor: the mass ratio of the SCD1 inhibitor is 1:1.
16. The use according to claim 15, wherein the combination is the simultaneous or sequential administration of an effective amount of TKIs and SCD1 inhibitor; further, the sequential administration of an effective amount of a TKIs and an SCD1 inhibitor is with TKIs inhibition followed by SCD1 inhibitor or with SCD1 inhibitor followed by TKIs inhibitor.
CN202311239865.9A 2023-09-22 2023-09-22 Pharmaceutical composition containing TKIs inhibitor and SCD1 inhibitor and anti-tumor application thereof Pending CN117338941A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311239865.9A CN117338941A (en) 2023-09-22 2023-09-22 Pharmaceutical composition containing TKIs inhibitor and SCD1 inhibitor and anti-tumor application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311239865.9A CN117338941A (en) 2023-09-22 2023-09-22 Pharmaceutical composition containing TKIs inhibitor and SCD1 inhibitor and anti-tumor application thereof

Publications (1)

Publication Number Publication Date
CN117338941A true CN117338941A (en) 2024-01-05

Family

ID=89360353

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311239865.9A Pending CN117338941A (en) 2023-09-22 2023-09-22 Pharmaceutical composition containing TKIs inhibitor and SCD1 inhibitor and anti-tumor application thereof

Country Status (1)

Country Link
CN (1) CN117338941A (en)

Similar Documents

Publication Publication Date Title
Duan et al. Desmoplasia suppression by metformin-mediated AMPK activation inhibits pancreatic cancer progression
Jiang et al. Increased serotonin signaling contributes to the Warburg effect in pancreatic tumor cells under metabolic stress and promotes growth of pancreatic tumors in mice
Samid et al. Selective growth arrest and phenotypic reversion of prostate cancer cells in vitro by nontoxic pharmacological concentrations of phenylacetate.
WO2016145974A1 (en) Use of gene engineering bacteria vpn 20009-m in preparation of medicaments for preventing and treating cancer metastasis
Gao et al. Inhibition of indoleamine 2, 3-dioxygenase enhances the therapeutic efficacy of immunogenic chemotherapeutics in breast cancer
CN105899223A (en) Treatment of metastatic prostate cancer
Tian et al. HYD-PEP06 suppresses hepatocellular carcinoma metastasis, epithelial–mesenchymal transition and cancer stem cell-like properties by inhibiting PI3K/AKT and WNT/β-catenin signaling activation
WO2021239817A1 (en) Therapeutic combinations comprising agonists of ferroptosis for treating proliferative disorders
JP2021527651A (en) Combination therapy with C / EBP alpha saRNA
CN109646680B (en) Combined medicine for treating KRAS mutant intestinal cancer
Gnocchi et al. Inhibition of LPAR6 overcomes sorafenib resistance by switching glycolysis into oxidative phosphorylation in hepatocellular carcinoma
EP2589385A1 (en) Triple combination for the treatment of cancer
Zhang et al. CDK14 inhibition reduces mammary stem cell activity and suppresses triple negative breast cancer progression
Kinoh et al. Nanomedicines blocking adaptive signals in cancer cells overcome tumor TKI resistance
Wang et al. The antitumor activity of betulinic acid-loaded nanoliposomes against colorectal cancer in vitro and in vivo via glycolytic and glutaminolytic pathways
CN106928229B (en) Application of tryptanthrin and derivatives thereof in preparation of hIDO2 inhibitor
CN117338941A (en) Pharmaceutical composition containing TKIs inhibitor and SCD1 inhibitor and anti-tumor application thereof
Subramanian et al. Synthetic high-density lipoprotein nanoconjugate targets neuroblastoma stem cells, blocking migration and self-renewal
Jiao et al. The metabolic landscape of breast cancer and its therapeutic implications
CN110215516A (en) It is a kind of to inhibit the treatment of CDK5 synergetic immunity in the application inhibited in breast cancer
CN110464845A (en) A kind of combination medicine combination and its application for Breast cancer immunotherapy
CN112933239A (en) Application of reagent for activating endogenous PD-1 of tumor cells in preparation of antitumor drugs
Romero-Trejo et al. The systemic administration of neural stem cells expressing an inducible and soluble form of growth arrest specific 1 inhibits mammary gland tumor growth and the formation of metastases
CN111419832A (en) Pharmaceutical composition and application thereof in preparing medicines for treating tumors
Lv et al. CSF1R inhibition reprograms tumor-associated macrophages to potentiate anti-PD-1 therapy efficacy against colorectal cancer

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