WO2024017406A1 - Use of mek/erk signal pathway inhibitor in preparation of drug for treating myeloproliferative neoplasms - Google Patents

Use of mek/erk signal pathway inhibitor in preparation of drug for treating myeloproliferative neoplasms Download PDF

Info

Publication number
WO2024017406A1
WO2024017406A1 PCT/CN2023/116402 CN2023116402W WO2024017406A1 WO 2024017406 A1 WO2024017406 A1 WO 2024017406A1 CN 2023116402 W CN2023116402 W CN 2023116402W WO 2024017406 A1 WO2024017406 A1 WO 2024017406A1
Authority
WO
WIPO (PCT)
Prior art keywords
hel
cells
drug
mek
apoptosis
Prior art date
Application number
PCT/CN2023/116402
Other languages
French (fr)
Chinese (zh)
Inventor
邓沱
胡婉钰
吾甫尔艾尼
Original Assignee
中南大学湘雅二医院
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 中南大学湘雅二医院 filed Critical 中南大学湘雅二医院
Publication of WO2024017406A1 publication Critical patent/WO2024017406A1/en

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/166Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the carbon of a carboxamide group directly attached to the aromatic ring, e.g. procainamide, procarbazine, metoclopramide, labetalol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • 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
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with 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

Definitions

  • the present invention relates to the field of medical technology, and in particular to the use of MEK/ERK signaling pathway inhibitors in the preparation of drugs for the treatment of myeloproliferative tumors.
  • Myeloproliferative neoplasms refer to a group of tumor diseases caused by the clonal proliferation of one or more lines of relatively mature bone marrow cells. The clinical manifestations are the proliferation of one or more blood cells, accompanied by enlargement of the liver, spleen or lymph nodes.
  • WHO World Health Organization
  • the classification of bone marrow tumors including polycythemia vera (PV), primary myelofibrosis (PMF), and essential thrombocythemia (ET). Included in the category of Philadelphia-negative classic myeloproliferative neoplasms.
  • Myeloproliferative neoplasms are clonal hematopoietic stem cell diseases.
  • the main disease driver gene mutations include JAK2/V617F, CALR, and MPL mutations.
  • JAK2/V617F mutation is the most common type and can be found in 95% of PV and 50-60% of cases. ET and 55-65% of patients with PMF.
  • the activation of genes leads to the activation of the JAK-STAT pathway, which leads to the occurrence of diseases.
  • Ruxolitinib a JAK1/JAK2 inhibitor
  • Ruxolitinib has been approved by the FDA as first-line treatment for intermediate and high-risk bone marrow fibrosis.
  • MF Myelofibrosis
  • Hydroxyurea HU
  • phase II and phase III clinical trials suggest that RUX can reduce spleen volume and symptoms in patients with intermediate and high-risk MF and PV compared with optimal therapy.
  • ruxolitinib there are many problems during the use of ruxolitinib.
  • the results of the COMFORT and RESPONSE clinical trials showed that MF patients treated with ruxolitinib had more severe anemia. What’s more serious is that long-term use of type I JAK inhibitors such as RUX can induce the occurrence of drug resistance.
  • Bone marrow transplantation is the only cure for myeloproliferative neoplasms, but there are still issues that need to be addressed. The choice of transplantation modality and regimen is uncertain, and the choice of allogeneic or haploidentical transplantation is unclear. Furthermore, when choosing transplantation, transplantation-related mortality and the long-term nature of myeloproliferative neoplasms must be considered. At present, bone marrow transplantation is mainly used to treat high-risk myelofibrosis patients, but the timing of bone marrow transplantation for patients with other types of myeloproliferative neoplasms needs further exploration and research to confirm. Bone marrow transplantation is expensive, and in the current medical environment, this treatment is not an option for most patients.
  • ruxolitinib is a landmark drug in the treatment of myeloproliferative tumors
  • ruxolitinib currently has a narrow scope of application, and myelosuppression as a common side effect limits its application in the main indication MF.
  • Ruxolitinib cannot reduce the burden of mutated genes, which means that ruxolitinib treatment cannot achieve molecular-level remission of the disease and cannot fundamentally treat myeloproliferative tumors.
  • the limited number of therapeutic drugs is a major challenge.
  • the purpose of the present invention is to address the problems existing in the current treatment of myeloproliferative neoplasms, and to treat patients with myeloproliferative neoplasms, especially myeloproliferative neoplasms that are resistant to ruxolitinib. People provide an effective, safe and reliable medicine.
  • the present invention provides the use of MEK/ERK signaling pathway inhibitors in preparing drugs for treating myeloproliferative tumors.
  • the myeloproliferative tumor is a myeloproliferative tumor that is resistant to chemotherapy drugs.
  • the chemotherapeutic drug is a chemotherapeutic drug for treating myeloproliferative tumors, and the chemotherapeutic drug includes at least one of ruxolitinib and fizotinib.
  • the myeloproliferative tumor is polycythemia vera, essential thrombocythemia or myelofibrosis
  • the myelofibrosis is primary myelofibrosis or myelofibrosis secondary to polycythemia vera. and at least one of myelofibrosis secondary to essential thrombocythemia.
  • the MEK/ERK signaling pathway inhibitor is at least one of Trematinib, Cobimetinib, Binimetinib, Selumetinib, Mirdametinib, Refametinib, TIC10, Ulixertinib, PD184352, and Pimasertib.
  • Trametinib (GSK1120212, JTP-74057, Mekinist) is a highly specific and effective MEK1/2 inhibitor that was approved by the US FDA in 2013. It is suitable for surgically unresectable melanoma or patients carrying BRAF V600E or V600K mutations. Treatment of adult patients with metastatic melanoma. On January 8, 2014, the FDA approved the combination of dabrafenib and trametinib for the treatment of patients with BRAF V600E/K mutant metastatic melanoma. On May 1, 2018, the FDA approved the combined use of dabrafenib/trametinib as adjuvant therapy.
  • Cobimetinib (GDC-0973, RG7420) is an effective and highly selective MEK1 inhibitor that was approved by the FDA on November 10, 2015. It is used in combination with vemurafenib to treat migratory melanoma with BRAF V600E or V600K mutations. NCT03695380 is currently recruiting for a Phase I clinical trial for the treatment of ovarian tumors.
  • the molecular formula of Cobimetinib is C 21 H 21 F 3 IN 3 O 2 , the molecular weight is 531.31, and the structural formula is shown in Formula II.
  • Binimetinib is a reversible inhibitor of mitogen-activated extracellular signal-regulated kinase 1 (MEK1) and cytokine MEK2 activity.
  • MEK protein is an upstream regulator of the extracellular signal-related kinase (ERK) pathway.
  • binimetinib inhibits extracellular signal-related kinase (ERK) phosphorylation immunoassays in cells as well as viability and MEK-dependent phosphorylation of BRAF-mutant human melanoma cells. Binimetinib also inhibits ERK phosphorylation and tumor growth in BRAF mutant mouse xenograft models.
  • Binimetinib is C 17 H 15 BrF 2 N 4 O 3 , the molecular weight is 441.23, and the structural formula is shown in Formula III.
  • Selumetinib also known as AZD6244 and Y-142886, is a highly efficient, non-ATP competitive inhibitor of MEK1/2 and ERK1/2. Selumetinib also has significant efficacy in multiple tumor models, significantly inhibiting ERK activity, inhibiting tumor growth, and inhibiting lung metastasis.
  • the U.S. FDA announced the approval of Koselugo (Selumetinib) capsules for the treatment of pediatric patients 2 years old and older with neurofibromatosis type I (NF1). This is the first FDA-approved treatment for NF1.
  • the drug is specifically indicated for the treatment of pediatric patients with symptomatic, inoperable plexiform neurofibromas.
  • the molecular formula of Selumetinib is C 17 H 15 BrClFN 4 O 3 ,
  • Mirdametinib is an oral, small molecule inhibitor of MEK1 and MEK2.
  • Mirdametinib The European Commission (EC) and the US FDA have granted Mirdametinib (formerly known as PD-0325901) orphan drug designation for the treatment of neurofibromatosis type 1 (NF1).
  • the molecular formula of Mirdametinib is C 16 H 14 F 3 IN 2 O 4 , the molecular weight is 482.19, and the structural formula is shown in Formula V.
  • Refametinib (RDEA119) is a potent, non-ATP competitive, highly selective inhibitor of MEK1 and MEK2 with IC50 of 19nM and 47nM respectively.
  • Refametinib is an oral MEK inhibitor with antitumor activity in combination with sorafenib for the treatment of patients with RAS-mutated hepatocellular carcinoma (HCC).
  • HCC RAS-mutated hepatocellular carcinoma
  • CLIN CANCER RES once published an article reporting the efficacy of Refametinib alone and Refametinib combined with Sorafenib in the treatment of patients with unresectable or metastatic HCC with RAS mutations.
  • Refametinib is currently undergoing a Phase II clinical study in advanced biliary tract cancer.
  • the molecular formula of Refametinib is C 19 H 20 F 3 IN 2 O 5 S, the molecular weight is 572.34, and the structural formula is shown in Formula
  • TIC10 (ONC201) inhibits Akt and ERK activity, induces TNF-related apoptosis-inducing ligand (TRAIL) through FoxO3a, can penetrate the blood-brain barrier, has super stability, and improved pharmacokinetic properties.
  • TIC10 causes tumor cell cell surface Significant and long-lasting expression of TRAIL on the face. In HCT116p53-/- cells, TIC10 also caused TRAIL-mediated apoptosis. Furthermore, TIC10 simultaneously inactivates Akt and ERK, leading to nuclear translocation of Foxo3a and subsequent upregulation of TRAIL.
  • TIC10 has TRAIL-dependent antitumor effects, causing tumor-specific cell death through TRAIL-mediated direct and bystander effects. This molecule has been found to be effective against a variety of solid tumors and has already carried out clinical phase 1 and 2 trials. Currently, it is difficult to advance clinical trials due to compound patent disputes.
  • the molecular formula of TIC10 is C 24 H 26 N 4 O, the molecular weight is 386.49, and the structural formula is shown in Formula VII.
  • Ulixertinib (BVD-523, VRT752271) is a potent reversible ERK1/ERK2 inhibitor with an IC50 of ERK2 ⁇ 0.3nM and can be taken orally. Ulixertinib has shown promising results in clinical trials in patients with advanced solid tumors.
  • the molecular formula of Ulixertinib is C 21 H 22 Cl 2 N 4 O 2 , the molecular weight is 433.33, and the structural formula is shown as Formula VIII.
  • PD184352 (CI-1040) is an ATP non-competitive MEK1/2 inhibitor with an IC50 of 17nM in cell assays and is 100 times more selective for MEK1/2 than MEK5.
  • PD184352(CI-1040) selectively induces apoptosis.
  • PD184352 is the first MEK to enter clinical trials Inhibitors have been stopped in phase II clinical trials due to problems such as poor solubility, short half-life, low oral bioavailability, and large individual differences.
  • the molecular formula of PD184352 is C 17 H 14 ClF 2 IN 2 O 2 , the molecular weight is 478.67, and the structural formula is shown in Formula IX.
  • Pimasertib (AS-703026, MSC1936369B, SAR 245509) is a highly selective, ATP-noncompetitive, orally available MEK1/2 allosteric inhibitor with an IC50 of 5nM-2 ⁇ M in MM cell lines. Pimasertib has been studied in more than 10 Phase 1/2 clinical trials in approximately 900 patients with various tumor types. Pimasertib, in combination with DAY101, is indicated for the treatment of patients ⁇ 12 years of age with recurrent, progressive, or refractory solid tumors with MAPK pathway aberrations. The molecular formula of Pimasertib is C 15 H 15 FIN 3 O 3 , the molecular weight is 431.20, and the structural formula is shown in Formula X.
  • the treatment includes inhibiting the proliferation of tumor cells and/or promoting the growth of tumor cells. Apoptosis.
  • the present invention also provides the use of MEK/ERK signaling pathway inhibitors in preparing drugs that inhibit the proliferation of HEL cells and/or promote the apoptosis of HEL cells.
  • the HEL cells include non-drug-resistant HEL cells and/or drug-resistant HEL cells.
  • Trematinib, Cobimetinib, Binimetinib, Selumetinib, Mirdametinib, Refametinib, Ulixertinib, PD184352, and Pimasertib cannot promote apoptosis of HEL cells, and only TIC10 can promote apoptosis.
  • Trematinib and TIC10 can inhibit the proliferation of HEL cells
  • Pimasertib has a weak inhibitory effect on HEL cell proliferation, and the other inhibitors have no inhibitory effect.
  • Trematinib, Cobimetinib, Binimetinib, Selumetinib, Mirdametinib, Refametinib, TIC10, Ulixertinib, PD184352, and Pimasertib can all inhibit the proliferation of drug-resistant HEL cells.
  • Refametinib can promote the apoptosis of drug-resistant HEL cells. Death. This shows that Refametinib has a more significant therapeutic effect on drug-resistant myeloproliferative tumors.
  • MEK/ERK signaling pathway inhibitors have a strong inhibitory effect on the proliferation of drug-resistant HEL cells, and some inhibitors can also promote cell apoptosis. This indicates that MEK/ERK signaling pathway inhibitors have a significant therapeutic effect on drug-resistant myeloproliferative tumors.
  • the medicine also contains other pharmaceutical ingredients for treating myeloproliferative tumors and pharmaceutically acceptable excipients.
  • the pharmaceutical dosage form is an oral preparation or an injection preparation.
  • the present invention provides the use of MEK/ERK signaling pathway inhibitors in the preparation of drugs for the treatment of myeloproliferative tumors.
  • the myeloproliferative tumors are polycythemia vera, essential thrombocythemia, or myelofibrosis and ruxolitinib-resistant myeloproliferative tumors, and the myelofibrosis is primary myelofibrosis, secondary myelofibrosis, or ruxolitinib-resistant myeloproliferative tumors.
  • Myelofibrosis arising from polycythemia vera or secondary to essential thrombocythemia.
  • the technical effects of the present invention are:
  • MEK/ERK signaling pathway inhibitors in the treatment of myeloproliferative tumors provides a new treatment approach for patients with myeloproliferative tumors and provides more options for clinicians and patients. select.
  • MEK/ERK signaling pathway inhibitors can provide patients with continued oral drug therapy and avoid bone marrow transplantation.
  • MEK/ERK signaling pathway inhibitors can be chemically synthesized and the cost is lower than biological agents.
  • the MEK/ERK signaling pathway inhibitors under application have all passed Phase I clinical trials, and some have successfully passed Phase II and III clinical trials. They will be used in clinical treatments in the future and have good clinical application prospects.
  • Figure 1 shows Example 1: The establishment results of ruxolitinib-resistant myeloproliferative tumor cell model HEL RE ;
  • FIG. 2 shows the results of Example 2: MEK/ERK signaling pathway inhibitors treated myeloproliferative tumor cells, and the cell proliferation was detected by the CellTiter-Lumi TM luminescence method; a is the result of HEL cell proliferation treated with Trematinib; b is the result of HEL cells treated with Cobimetinib. Cell proliferation results; c is Binimetinib-treated HEL cell proliferation results; d is Selumetinib-treated HEL cell proliferation results; e is Mirdametinib-treated HEL cell proliferation results; f is Refametinib-treated HEL cell proliferation results; g is TIC10-treated HEL.
  • the cell proliferation result chart; h is the HEL cell proliferation result chart treated with Ulixertinib; i is the HEL cell proliferation result chart treated with PD184352; j is the HEL cell proliferation result chart treated with Pimasertib;
  • FIG. 3 shows the results of Example 3: MEK/ERK signaling pathway inhibitors treated myeloproliferative tumor cells, using AnnexinV-PI staining to detect cell apoptosis by flow cytometry; a is the result of Trematinib treatment of HEL cell apoptosis; b is the result of apoptosis of HEL cells treated with Trematinib; Cobimetinib treated HEL cell apoptosis results; c is Binimetinib treated HEL cell apoptosis results; d is Selumetinib treated HEL cell apoptosis results; e is Mirdametinib treated HEL cell apoptosis results; f is Refametinib treated HEL cell apoptosis Result graph; g is the graph of the apoptosis result of HEL cells treated with TIC10; h is the graph of the apoptosis result of HEL cells treated with U
  • Figure 4 shows Example 4: MEK/ERK signaling pathway inhibitors treated ruxolitinib-resistant myeloproliferative tumor cells, and the results of detecting cell proliferation by CellTiter-Lumi TM luminescence assay; a shows the proliferation of HEL RE cells treated with Trematinib.
  • Result chart b is the result chart of HEL RE cell proliferation treated with Cobimetinib; c is the result chart of HEL RE cell proliferation treated with Binimetinib; d is Selumetinib The result of HEL RE cell proliferation after treatment; e is the result of HEL RE cell proliferation treated with Mirdametinib; f is the result of HEL RE cell proliferation treated with Refametinib; g is the result of HEL RE cell proliferation treated with TIC10; h is the result of HEL RE cell proliferation treated with Ulixertinib.
  • Figure i is the result of HEL RE cell proliferation treated with PD184352; j is the result of HEL RE cell proliferation treated with Pimasertib;
  • FIG. 5 shows Example 5: MEK/ERK signaling pathway inhibitors treated ruxolitinib-resistant myeloproliferative tumor cells, and the results of flow cytometry detection of cell apoptosis after staining with AnnexinV-PI; a is Trematinib treatment of HEL RE Cell apoptosis results; b is Cobimetinib-treated HEL RE cell apoptosis results; c is Binimetinib-treated HEL RE cell apoptosis results; d is Selumetinib-treated HEL RE cell apoptosis results; e is Mirdametinib-treated HEL RE cell apoptosis f is the result of apoptosis of HEL RE cells treated with Refametinib; g is the result of apoptosis of HEL RE cells treated with TIC10; h is the result of apoptosis of HEL RE cells treated with Ulixertin
  • the present invention discloses the application of MEK/ERK signaling pathway inhibitors in the preparation of drugs for the treatment of myeloproliferative tumors.
  • Persons skilled in the art can learn from the contents of this article and appropriately improve the process parameters for implementation. It should be noted that all similar substitutions and modifications are obvious to those skilled in the art, and they are deemed to be included in the present invention.
  • the methods and applications of the present invention have been described through preferred embodiments. Relevant persons can obviously make modifications or appropriate changes and combinations to the methods and applications described herein without departing from the content, spirit and scope of the present invention to achieve and Apply the technology of this invention.
  • the inhibitory effect of MEK/ERK signaling pathway inhibitors on myeloproliferative tumor cells is clarified through a cell line model.
  • the present invention establishes ruxolitinib resistance based on two commonly used human myeloproliferative tumor cell lines containing JAK2-V617F mutations, namely HEL cells (Human erythroleukemia cell line, human erythroleukemia cells).
  • Drug Cell Model HEL RE The drug resistance model is constructed using Start adding ruxolitinib at the IC50 concentration of the cells and slowly increase it to a high concentration to keep the cells from being killed. Compare the IC50 to verify whether the model is successfully constructed.
  • the present invention uses increasing concentrations of the MEK/ERK signaling pathway to inhibit The myeloproliferative tumor cell lines were treated with the agent, and the cell proliferation was detected by CellTiter-Lumi TM luminescence method. The results show that some MEK/ERK signaling pathway inhibitors can successfully inhibit the proliferation of HEL cells.
  • the present invention uses increasing concentrations of MEK/ERK signaling pathway inhibitors to treat myeloproliferative tumor cell lines, and uses AnnexinV-PI staining to detect cell apoptosis using flow cytometry.
  • the results show that some MEK/ERK signaling pathway inhibitors can promote the apoptosis of HEL cells.
  • MEK/ERK signaling pathway inhibitors can be used to treat myeloproliferative tumor diseases.
  • the present invention uses increasing concentrations of MEK/ERK signaling pathway inhibitors to treat ruxolitinib-resistant myeloproliferative tumor cells, and detect cell proliferation through the CellTiter-Lumi TM luminescence method. The results showed that inhibitors of the MEK/ERK signaling pathway could inhibit the proliferation of HEL RE cells.
  • the present invention uses increasing concentrations of MEK/ERK signaling pathway inhibitors to treat ruxolitinib-resistant myeloproliferative tumor cells, and uses AnnexinV-PI staining to detect the apoptosis of the cells by flow cytometry.
  • the results show that some MEK/ERK signaling pathway inhibitors can promote the apoptosis of HEL RE cells.
  • MEK/ERK signaling pathway inhibitors can be used to treat ruxolitinib-resistant myeloproliferative tumor diseases.
  • the present invention provides the use of some MEK/ERK signaling pathway inhibitors in the preparation of drugs that inhibit the proliferation of HEL cells and promote the apoptosis of HEL cells.
  • the present invention provides the use of MEK/ERK signaling pathway inhibitors in the preparation of drugs for treating myeloproliferative tumors.
  • the myeloproliferative tumors are polycythemia vera, essential thrombocythemia and myelofibrosis (including primary myelofibrosis, myelofibrosis secondary to polycythemia vera and myelofibrosis secondary to primary myelofibrosis). Thrombocytosis, myelofibrosis) and drug-resistant myeloproliferative neoplasms.
  • the drug-resistant myeloproliferative neoplasm is a ruxolitinib-resistant myeloproliferative neoplasm.
  • the drug-resistant myeloproliferative neoplasm is drug-resistant polycythemia vera, drug-resistant myelofibrosis (including primary myelofibrosis, secondary myelofibrosis Polycythemia myelofibrosis and myelofibrosis secondary to essential thrombocythemia) and drug-resistant essential thrombocythemia.
  • the drugs are Trematinib, Cobimetinib, Binimetinib, Selumetinib, Mirdametinib, Refametinib, TIC10, Ulixertinib, PD184352, and Pimasertib.
  • the medicine also includes pharmaceutically acceptable excipients.
  • the medicine can be in any dosage form in the current pharmaceutical field, including oral preparations or injection preparations.
  • Each pharmaceutical dosage form can be prepared by selecting appropriate acceptable excipients according to the actual needs of the dosage form, which is a conventional dosage form preparation technology in this field. Such as making capsules, tablets, injection powders, etc.
  • Example 1 Establishment of two common ruxolitinib-resistant cell models (HEL RE ).
  • HEL Human erythroleukemia cell line
  • ruxolitinib-resistant HEL cells were cultured in RPMI medium (Gibco) containing 20% heat-inactivated fetal calf serum (Gibco) and 1% penicillin/streptomycin.
  • the ruxolitinib-resistant HEL model is the HEL RE model.
  • the construction method is to start adding ruxolitinib at a concentration lower than the IC50 of the original cells and slowly increase it to a high concentration to maintain the cells from being killed.
  • Our starting concentration is 0.1 ⁇ M, and the drug is added when cells proliferate.
  • the drug addition gradient is 1.25 times increments, and the final concentration is 2.0 ⁇ M.
  • Stable drug-resistant cells were obtained after 4-6 weeks.
  • Ruxolitinib and MEK/ERK signaling pathway inhibitors were purchased from Selleck Company, dissolved in DMSO, the concentration of the mother solution was 10mM, and frozen at -80°C. The working solution was diluted to the specified multiple with RPMI medium before processing the cells. Ruxolitinib is specifically ruxolitinib phosphate.
  • the above cell lines were cultured at 3000 cells/100 cells per well. ⁇ L system culture, add increasing concentrations of ruxolitinib (HEL cell concentration gradient: 0, 0.1, 0.3, 1, 3, 10 ⁇ M; or 0, 0.001, 0.003, 0.01, 0.03, 0.1 ⁇ M), and DMSO is added until equal. quantity. Set up 4 parallel replicate groups and set up 3 blank wells (wells containing culture medium without cells). After 48 hours, the cell proliferation was detected by CellTiter-Lumi TM luminescence method (Beyotime). Multifunctional microplate reader reading, IC50 is calculated by GraphPadprism.
  • cell proliferation rate (Luminescence value of drug-added group - average Luminescence value of blank wells) / (Luminescence value of DMSO control group - average Luminescence value of blank wells) ⁇ 100%.
  • the method to evaluate whether the model is successfully constructed is to compare the IC50 of drug-resistant cells and original cells.
  • the ratio is the drug resistance index. If the ratio is greater than 3, the construction is successful.
  • the IC50 of HEL cells is 0.801 ⁇ M
  • the IC50 of HEL RE cells is 12.27 ⁇ M
  • its drug resistance index is 15.3, indicating the successful construction of the drug resistance model HEL RE .
  • the proliferation rate results in the figure are shown as the mean ⁇ standard deviation.
  • the comparison of the proliferation rates of the two groups in the figure is using t test (***p ⁇ 0.001), and the IC50 is shown as the mean.
  • Example 2 Some MEK/ERK signaling pathway inhibitors can inhibit the proliferation of myeloproliferative tumor cells.
  • the method was to use increasing concentrations of ruxolitinib and MEK/ERK signaling pathway inhibitors to treat the HEL original cell line, and then use CellTiter- The Lumi TM luminescence method was used to detect cell proliferation.
  • the method was the same as in Example 1. The results are shown in Figure 2.
  • Figure 2 reflects that in HEL cells, the drug concentrations (average proliferation rate % ⁇ standard deviation) in the ruxolitinib-treated group are: 0 ⁇ M (100 ⁇ 1.47) (not shown in the figure), 0.1 ⁇ M (81.0 ⁇ 1.35), 0.3 ⁇ M (54.9 ⁇ 4.35), 1 ⁇ M (42.7 ⁇ 4.05), 3 ⁇ M (34.7 ⁇ 1.14), 10 ⁇ M (29.4 ⁇ 0.497);
  • Figure 2a reflects that in HEL cells, the drug concentration (average proliferation rate) of the Trematinib treatment group is: 0 ⁇ M ( 100 ⁇ 9.58) (not shown in the figure), 0.0001 ⁇ M (93.91 ⁇ 2.37), 0.0003 ⁇ M (98.17 ⁇ 3.44), 0.01 ⁇ M (84.82 ⁇ 6.67), 0.03 ⁇ M (83.96 ⁇ 9.48), 0.1 ⁇ M (85.99 ⁇ 6.9) .
  • FIG. 2b reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Cobimetinib treatment group are: 0 ⁇ M (100 ⁇ 6.6) (not shown in the figure), 0.001 ⁇ M (98.53 ⁇ 4.64), 0.003 ⁇ M (105.51 ⁇ 7.14), 0.01 ⁇ M (99.78 ⁇ 5.17), 0.03 ⁇ M (99.66 ⁇ 1.93), 0.1 ⁇ M (96.59 ⁇ 8.57). This suggests that Cobimetinib cannot inhibit the proliferation of HEL cells.
  • Figure 2c reflects that the drug concentrations (average proliferation rate) of the Binimetinib treatment group are: 0 ⁇ M (100 ⁇ 2.65) (not shown in the figure), 0.1 ⁇ M (111 ⁇ 13.9), 0.3 ⁇ M (105 ⁇ 17.2), 1 ⁇ M (115 ⁇ 22.9) , 3 ⁇ M (107 ⁇ 14.4), 10 ⁇ M (92.4 ⁇ 2.44). This suggests that Binimetinib cannot inhibit the proliferation of HEL cells.
  • Figure 2d reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Selumetinib treatment group are: 0 ⁇ M (100 ⁇ 1.62) (not shown in the figure), 0.1 ⁇ M (107 ⁇ 17.6), 0.3 ⁇ M (117 ⁇ 16.3), 1 ⁇ M (110 ⁇ 26.3), 3 ⁇ M (103 ⁇ 14.5), 10 ⁇ M (88.8 ⁇ 2.69). This suggests that Selumetinib cannot inhibit the proliferation of HEL cells.
  • Figure 2e reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Mirdametinib treatment group are: 0 ⁇ M (100 ⁇ 1.73) (not shown in the figure), 0.1 ⁇ M (107 ⁇ 17.6), 0.3 ⁇ M (117 ⁇ 16.3), 1 ⁇ M (110 ⁇ 26.3), 3 ⁇ M (103 ⁇ 14.5), 10 ⁇ M (88.8 ⁇ 2.69). This suggests that Mirdametinib cannot inhibit the proliferation of HEL cells.
  • Figure 2f reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Refametinib treatment group are: 0 ⁇ M (100 ⁇ 2.63) (not shown in the figure), 0.1 ⁇ M (96.0 ⁇ 7.44), 0.3 ⁇ M (99.8 ⁇ 4.36), 1 ⁇ M (93.4 ⁇ 4.31), 3 ⁇ M (89.1 ⁇ 7.23), 10 ⁇ M (84.6 ⁇ 8.89). This suggests that Refametinib cannot inhibit the proliferation of HEL cells.
  • Figure 2g reflects that in HEL cells, the drug concentrations (average proliferation rate) of the TIC10 treatment group are: 0 ⁇ M (100 ⁇ 4.31) (not shown in the figure), 0.1 ⁇ M (113 ⁇ 15.0), 0.3 ⁇ M (116 ⁇ 14.9), 1 ⁇ M (80.3 ⁇ 3.43), 3 ⁇ M (19.6 ⁇ 2.64), 10 ⁇ M (11.1 ⁇ 7.15).
  • TIC10 can inhibit the proliferation of HEL cells. This effect increases as the drug concentration increases, and its inhibitory effect strengthens as the concentration increases.
  • Figure 2h reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Ulixertinib treatment group are: 0 ⁇ M (100 ⁇ 2.58) (not shown in the figure), 0.1 ⁇ M (98.9 ⁇ 8.68), 0.3 ⁇ M (109 ⁇ 5.36), 1 ⁇ M (96.1 ⁇ 5.13), 3 ⁇ M (100 ⁇ 4.61), 10 ⁇ M (95.2 ⁇ 5.83). This suggests that Ulixertinib cannot inhibit the proliferation of HEL cells.
  • Figure 2i reflects that in HEL cells, the drug concentrations (average proliferation rate) of the PD184352 treatment group are: 0 ⁇ M (100 ⁇ 4.46) (not shown in the figure), 0.1 ⁇ M (105 ⁇ 16.7), 0.3 ⁇ M (118 ⁇ 21.2), 1 ⁇ M (103 ⁇ 22.7), 3 ⁇ M (99.0 ⁇ 20.3), 10 ⁇ M (82.3 ⁇ 6.50). This suggests that PD184352 cannot inhibit the proliferation of HEL cells.
  • Figure 2j reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Pimasertib treatment group are: 0 ⁇ M (100 ⁇ 3.07) (not shown in the figure), 0.1 ⁇ M (89.0 ⁇ 1.42), 0.3 ⁇ M (95.0 ⁇ 7.62), 1 ⁇ M (86.9 ⁇ 1.71), 3 ⁇ M (82.8 ⁇ 2.13), 10 ⁇ M (77.3 ⁇ 2.52).
  • the comparison of proliferation rates between the two groups in the figure uses t test (*p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001).
  • the HEL original cell line was treated with ruxolitinib and MEK/ERK signaling pathway inhibitors for 24 hours (concentration: 0, 0.1, 0.3, 1, 3, 10 ⁇ M), and supplemented with DMSO to equal amounts.
  • Three parallel replicate groups were set up, and cell apoptosis was detected by flow cytometry after AnnexinV and PI staining.
  • Apoptosis rate early apoptotic cell ratio (Annexin V + /PI - ) + late apoptotic cell and necrotic cell ratio (AnnexinV + /PI + ).
  • the drug concentrations (average apoptosis rate % ⁇ standard deviation) of Trematinib, Cobimetinib, TIC10, Ulixertinib, PD184352, and Pimasertib in the ruxolitinib-treated group of HEL cells in the control group are: 0 ⁇ M (7.26 ⁇ 0.360), 0.1 ⁇ M (7.39 ⁇ 0.0600), 0.3 ⁇ M (7.40 ⁇ 0.820), 1 ⁇ M (8.58 ⁇ 0.490), 3 ⁇ M (7.12 ⁇ 0.180), 10 ⁇ M (6.04 ⁇ 0.700); Binimetinib, Selumetinib, Mirdametinib, Refametinib control HEL cell ruxolitinib treatment group
  • the drug concentrations (average apoptosis rate % ⁇ standard deviation) are: 0 ⁇ M (2.30 ⁇ 0.720), 0.1 ⁇ M (2.30 ⁇ 0.210), 0.3 ⁇ M (1.90 ⁇ 0.360), 1 ⁇ M (2.4 ⁇ 0.5
  • Figure 3a reflects that in HEL cells, the drug concentrations (average apoptosis rate) in the Trematinib treatment group are: 0 ⁇ M (6.94 ⁇ 0.690) (not shown in the figure), 0.1 ⁇ M (7.09 ⁇ 1.35), 0.3 ⁇ M (6.32 ⁇ 0.780), 1 ⁇ M (6.11 ⁇ 1.19), 3 ⁇ M (6.83 ⁇ 1.83), 10 ⁇ M (6.41 ⁇ 0.240). This suggests that Trematinib cannot promote apoptosis of HEL cells.
  • Figure 3b reflects that in HEL cells, the drug concentrations (average apoptosis rate) of the Cobimetinib treatment group are: 0 ⁇ M (5.09 ⁇ 0.060) (not shown in the figure), 0.1 ⁇ M (5.35 ⁇ 0.500), 0.3 ⁇ M (6.53 ⁇ 0.370), 1 ⁇ M (6.31 ⁇ 0.670), 3 ⁇ M (6.61 ⁇ 1.490), 10 ⁇ M (5.64 ⁇ 0.470). This suggests that Cobimetinib cannot promote apoptosis of HEL cells.
  • the drug concentrations (apoptosis rate) in the Binimetinib treatment group are 0 ⁇ M (1.20 ⁇ 0.320) (not shown in the figure), 0.1 ⁇ M (1.21 ⁇ 0.110), 0.3 ⁇ M (1.55 ⁇ 0.310), 1 ⁇ M (1.73 ⁇ 0.06), 3 ⁇ M (1.39 ⁇ 0.29), 10 ⁇ M (2.19 ⁇ 0.07), which suggests that Binimetinib cannot promote the apoptosis of myeloproliferative tumor cells.
  • the drug concentrations (average apoptosis rate) in the Selumetinib-treated group of HEL cells are: 0 ⁇ M (1.91 ⁇ 0.51) (not shown in the figure), 0.1 ⁇ M (2.24 ⁇ 0.0580), 0.3 ⁇ M (2.14 ⁇ 0.748), 1 ⁇ M ( 2.45 ⁇ 0.473), 3 ⁇ M (1.91 ⁇ 0.225), 10 ⁇ M (2.40 ⁇ 0.260), which suggests that Selumetinib cannot promote the apoptosis of myeloproliferative tumor cells.
  • Figure 3e reflects that in HEL cells, the drug concentrations (average apoptosis rate) of Mirdametinib treatment group are: 0 ⁇ M (1.46 ⁇ 0.146) (not shown in the figure), 0.1 ⁇ M (1.99 ⁇ 0.367), 0.3 ⁇ M (2.23 ⁇ 0.432), 1 ⁇ M (2.45 ⁇ 0.473), 3 ⁇ M (1.91 ⁇ 0.225), 10 ⁇ M (2.40 ⁇ 0.260). This suggests that Mirdametinib cannot promote apoptosis of HEL cells.
  • Figure 3f reflects that in HEL cells, the drug concentrations (average apoptosis rate) of the Refametinib treatment group are: 0 ⁇ M (2.08 ⁇ 0.467) (not shown in the figure), 0.1 ⁇ M (2.78 ⁇ 1.02), 0.3 ⁇ M (2.21 ⁇ 0.369), 1 ⁇ M (2.06 ⁇ 0.457), 3 ⁇ M (2.39 ⁇ 0.713), 10 ⁇ M (1.79 ⁇ 0.745). This suggests that Refametinib cannot promote apoptosis of HEL cells.
  • Figure 3g reflects that in HEL cells, the drug concentrations (average apoptosis rate) of the TIC10 treatment group are: 0 ⁇ M (3.69 ⁇ 0.192) (not shown in the figure), 0.1 ⁇ M (3.19 ⁇ 0.516), 0.3 ⁇ M (5.63 ⁇ 2.50), 1 ⁇ M (4.39 ⁇ 0.2880), 3 ⁇ M (5.16 ⁇ 0.266), 10 ⁇ M (9.43 ⁇ 0.445).
  • TIC10 can promote the apoptosis of HEL cells, and its effect increases with increasing concentration.
  • the drug concentrations (average apoptosis rate) of the Ulixertinib treatment group are: 0 ⁇ M (6.00 ⁇ 0.717) (not shown in the figure), 0.1 ⁇ M (5.56 ⁇ 1.22), 0.3 ⁇ M (6.22 ⁇ 1.86), 1 ⁇ M (5.72 ⁇ 0.619), 3 ⁇ M (5.99 ⁇ 1.73), 10 ⁇ M (6.12 ⁇ 0.903). This suggests that Ulixertinib cannot promote apoptosis of HEL cells.
  • Figure 3i reflects that in HEL cells, the drug concentration (average apoptosis rate) of the PD184352 treatment group was: 0 ⁇ M (3.86 ⁇ 0.198) (not shown in the figure), 0.1 ⁇ M (4.65 ⁇ 0.959), 0.3 ⁇ M (4.90 ⁇ 0.494), 1 ⁇ M (4.72 ⁇ 0.161), 3 ⁇ M (4.19 ⁇ 0.709), 10 ⁇ M (4.77 ⁇ 0.902). This suggests that PD184352 cannot promote apoptosis of HEL cells.
  • Figure 3j reflects that in HEL cells, the drug concentrations (average apoptosis rate) of the Pimasertib treatment group are: 0 ⁇ M (3.98 ⁇ 0.115) (not shown in the figure), 0.1 ⁇ M (4.61 ⁇ 0.535), 0.3 ⁇ M (5.74 ⁇ 1.68), 1 ⁇ M (1.37 ⁇ 0.290), 3 ⁇ M (6.37 ⁇ 0.272), 10 ⁇ M (4.89 ⁇ 0.182).
  • t test (*p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001).
  • MEK/ERK signaling pathway inhibitors can inhibit the proliferation of drug-resistant myeloproliferative tumor cells.
  • the method was to use increasing concentrations of ruxolitinib and MEK/ERK signaling pathway inhibitors to treat HEL RE through CellTiter-Lumi TM Cell proliferation was detected by luminescence.
  • the method is the same as in Example 1, and the results are shown in Figure 4.
  • Figure 4 reflects that in HEL RE cells, the drug concentrations (average proliferation rate % ⁇ standard deviation) of the ruxolitinib treatment group are: 0 ⁇ M (100 ⁇ 3.79) (not shown in the figure), 0.1 ⁇ M (105 ⁇ 2.08), 0.3 ⁇ M (105 ⁇ 3.60), 1 ⁇ M (104 ⁇ 9.46), 3 ⁇ M (109 ⁇ 4.40), 10 ⁇ M (96.0 ⁇ 3.69);
  • Figure 4a reflects that in HEL RE cells, the drug concentration (average proliferation rate) of the Trematinib treatment group is: 0 ⁇ M (100 ⁇ 2.27) (not shown in the figure), 0.001 ⁇ M (99.38 ⁇ 2.75), 0.003 ⁇ M (94.51 ⁇ 3.41), 0.01 ⁇ M (67.59 ⁇ 6.26), 0.03 ⁇ M (51.61 ⁇ 2.5), 0.1 ⁇ M (40.45 ⁇ 4.5).
  • FIG. 4b reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the Cobimetinib treatment group are: 0 ⁇ M (100 ⁇ 2.22) (not shown in the figure), 0.001 ⁇ M (94.82 ⁇ 3.95), 0.003 ⁇ M (101.27 ⁇ 4.23), 0.01 ⁇ M (98.06 ⁇ 2.53), 0.03 ⁇ M (83.06 ⁇ 2.48), 0.1 ⁇ M (58.69 ⁇ 3.51). This suggests that Cobimetinib can effectively inhibit the proliferation of HEL RE cells.
  • Figure 4c reflects that the drug concentrations (average proliferation rate) of the Binimetinib treatment group are: 0 ⁇ M (100 ⁇ 4.10) (not shown in the figure), 0.1 ⁇ M (67.9 ⁇ 3.44), 0.3 ⁇ M (53.8 ⁇ 1.41), 1 ⁇ M (50.4 ⁇ 3.27) , 3 ⁇ M (43.1 ⁇ 2.18), 10 ⁇ M (37.6 ⁇ 1.24).
  • 0 ⁇ M 100 ⁇ 4.10 (not shown in the figure)
  • 0.1 ⁇ M 67.9 ⁇ 3.44
  • 0.3 ⁇ M 53.8 ⁇ 1.411
  • 1 ⁇ M 50.4 ⁇ 3.27
  • 3 ⁇ M (43.1 ⁇ 2.18) 10 ⁇ M (37.6 ⁇ 1.24).
  • Figure 4d reflects the HEL RE fine In cells, the drug concentrations (average proliferation rate) of the Selumetinib-treated group were: 0 ⁇ M (100 ⁇ 1.45) (not shown in the figure), 0.1 ⁇ M (77.1 ⁇ 1.86), 0.3 ⁇ M (62.0 ⁇ 3.90), 1 ⁇ M (54.9 ⁇ 1.63) , 3 ⁇ M (48.2 ⁇ 2.63), 10 ⁇ M (41.0 ⁇ 3.57). This suggests that Selumetinib can inhibit the proliferation of HEL RE cells, and the inhibition rate increases with increasing drug concentration.
  • Figure 4e reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the Mirdametinib treatment group are: 0 ⁇ M (100 ⁇ 1.70) (not shown in the figure), 0.1 ⁇ M (54.8 ⁇ 2.74), 0.3 ⁇ M (57.9 ⁇ 4.50) , 1 ⁇ M (44.2 ⁇ 3.38), 3 ⁇ M (39.6 ⁇ 0.380), 10 ⁇ M (32.5 ⁇ 3.73).
  • 0 ⁇ M 100 ⁇ 1.70
  • 0.1 ⁇ M 54.8 ⁇ 2.74
  • 0.3 ⁇ M 57.9 ⁇ 4.50
  • 1 ⁇ M (44.2 ⁇ 3.38) 1 ⁇ M (44.2 ⁇ 3.38)
  • 3 ⁇ M (39.6 ⁇ 0.380) 3 ⁇ M (32.5 ⁇ 3.73).
  • Figure 4f reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the Refametinib treatment group are: 0 ⁇ M (100 ⁇ 3.85) (not shown in the figure), 0.1 ⁇ M (66.0 ⁇ 3.12), 0.3 ⁇ M (61.2 ⁇ 2.05), 1 ⁇ M (50.6 ⁇ 2.09), 3 ⁇ M (43.3 ⁇ 1.23), 10 ⁇ M (36.6 ⁇ 2.83).
  • 0 ⁇ M 100 ⁇ 3.85
  • 0.1 ⁇ M 66.0 ⁇ 3.12
  • 0.3 ⁇ M 61.2 ⁇ 2.05
  • 1 ⁇ M 50.6 ⁇ 2.09
  • 3 ⁇ M (43.3 ⁇ 1.23) 10 ⁇ M (36.6 ⁇ 2.83).
  • Figure 4g reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the TIC10 treatment group are: 0 ⁇ M (100 ⁇ 6.41) (not shown in the figure), 0.1 ⁇ M (101 ⁇ 7.19), 0.3 ⁇ M (105 ⁇ 3.69), 1 ⁇ M (62.5 ⁇ 5.82), 3 ⁇ M (31.3 ⁇ 1.45), 10 ⁇ M (19.6 ⁇ 1.01). This suggests that TIC10 can inhibit the proliferation of HEL RE cells, and this effect increases with increasing drug concentration.
  • Figure 4h reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of Ulixertinib treatment group are: 0 ⁇ M (100 ⁇ 1.65) (not shown in the figure), 0.1 ⁇ M (95.6 ⁇ 5.27), 0.3 ⁇ M (91.4 ⁇ 3.49), 1 ⁇ M (58.5 ⁇ 14.1), 3 ⁇ M (48.4 ⁇ 7.37), 10 ⁇ M (37.5 ⁇ 6.71). This suggests that Ulixertinib can inhibit the proliferation of HEL RE cells, and this effect increases with increasing drug concentration.
  • Figure 4i reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the PD184352 treatment group are: 0 ⁇ M (100 ⁇ 4.73) (not shown in the figure), 0.1 ⁇ M (81.8 ⁇ 1.27), 0.3 ⁇ M (79.6 ⁇ 3.79), 1 ⁇ M (52.2 ⁇ 1.24), 3 ⁇ M (42.7 ⁇ 0.720), 10 ⁇ M (33.9 ⁇ 0.700).
  • PD184352 can inhibit the proliferation of HEL RE cells, and this effect increases with increasing drug concentration.
  • Figure 4j reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the Pimasertib treatment group are: 0 ⁇ M (100 ⁇ 3.71) (not shown in the figure), 0.1 ⁇ M (59.3 ⁇ 1.78), 0.3 ⁇ M (53.8 ⁇ 0.410), 1 ⁇ M (42.2 ⁇ 3.46), 3 ⁇ M (36.5 ⁇ 1.66), 10 ⁇ M (30.3 ⁇ 2.22).
  • the proliferation rate of the two groups was compared using t test (*p ⁇ 0.05, **p ⁇ 0.01,***p ⁇ 0.001).
  • Example 5 Some MEK/ERK signaling pathway inhibitors can promote the apoptosis of drug-resistant myeloproliferative tumor cells.
  • the method was to treat HEL RE with increasing concentrations of ruxolitinib and MEK/ERK signaling pathway inhibitors, and stain with AnnexinV and PI.
  • the apoptosis of cells was detected by flow cytometry. The method is the same as in Example 3, and the results are shown in Figure 5.
  • the drug concentrations (average apoptosis rate % ⁇ standard deviation) of TIC10, Ulixertinib, PD184352, and Pimasertib control HEL RE cells in the ruxolitinib-treated group were: 0 ⁇ M (2.15 ⁇ 1.01), 0.1 ⁇ M (2.73 ⁇ 0.748), 0.3 ⁇ M ( 2.11 ⁇ 0.989), 1 ⁇ M (2.73 ⁇ 0.748), 3 ⁇ M (2.09 ⁇ 0.982), 10 ⁇ M (1.900 ⁇ 0.751);
  • Figure 5a reflects that in HEL RE cells, the drug concentration (average apoptosis rate) of the Trematinib treatment group is: 0 ⁇ M ( 7.13 ⁇ 1.46) (not shown in the figure), 0.1 ⁇ M (9.42 ⁇ 2.48), 0.3 ⁇ M (6.49 ⁇ 1.58), 1 ⁇ M (9.56 ⁇ 1.39), 3 ⁇ M (8.01 ⁇ 2.04), 10 ⁇ M (7.72 ⁇ 0.167).
  • FIG. 5b reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) in the Cobimetinib treatment group are: 0 ⁇ M (4.87 ⁇ 1.37) (not shown in the figure), 0.1 ⁇ M (3.53 ⁇ 1.10), 0.3 ⁇ M (1.88 ⁇ 0.160) , 1 ⁇ M (4.09 ⁇ 1.46), 3 ⁇ M (2.90 ⁇ 0.344), 10 ⁇ M (2.36 ⁇ 0.414). This suggests that Cobimetinib cannot promote apoptosis of HEL RE cells.
  • the drug concentrations (apoptosis rate) of the Binimetinib treatment group are 0 ⁇ M (0.250 ⁇ 0.0620) (not shown in the figure), 0.1 ⁇ M (0.220 ⁇ 0.0300), 0.3 ⁇ M (0.160 ⁇ 0.0410), 1 ⁇ M (0.270 ⁇ 0.0360), 3 ⁇ M (0.240 ⁇ 0.0180), 10 ⁇ M (0.220 ⁇ 0.111), which suggests that binimetinib cannot promote apoptosis of drug-resistant myeloproliferative tumor cells.
  • the drug concentrations (average apoptosis rate) in the Selumetinib-treated group of HEL RE cells are: 0 ⁇ M (0.290 ⁇ 0.110) (not shown in the figure), 0.1 ⁇ M (0.230 ⁇ 0.0160), 0.3 ⁇ M (0.270 ⁇ 0.0790), 1 ⁇ M (0.220 ⁇ 0.0400), 3 ⁇ M (0.310 ⁇ 0.0290), 10 ⁇ M (0.320 ⁇ 0.00800), which suggests that Selumetinib cannot promote the apoptosis of drug-resistant myeloproliferative tumor cells.
  • Figure 5e reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) of Mirdametinib treatment group are: 0 ⁇ M (0.270 ⁇ 0.0250) (not shown in the figure), 0.1 ⁇ M (0.220 ⁇ 0.0180), 0.3 ⁇ M (0.370 ⁇ 0.0640) , 1 ⁇ M (0.410 ⁇ 0.0340), 3 ⁇ M (0.140 ⁇ 0.141), 10 ⁇ M (0.330 ⁇ 0.0220). This suggests that Mirdametinib cannot promote apoptosis in drug-resistant myeloproliferative tumor cells.
  • Figure 5f reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) in the Refametinib treatment group are: 0 ⁇ M (0.320 ⁇ 0.0670) (not shown in the figure), 0.1 ⁇ M (0.320 ⁇ 0.0590), 0.3 ⁇ M (0.330 ⁇ 0.0660) , 1 ⁇ M (0.330 ⁇ 0.0710), 3 ⁇ M (0.560 ⁇ 0.0420), 10 ⁇ M (0.800 ⁇ 0.0810).
  • This suggests that Refametinib can promote apoptosis of drug-resistant myeloproliferative tumor cells, and its effect increases with increasing concentration.
  • Figure 5g reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) in the TIC10 treatment group are: 0 ⁇ M (2.450 ⁇ 0.169) (not shown in the figure), 0.1 ⁇ M (2.35 ⁇ 0.286), 0.3 ⁇ M (1.89 ⁇ 0.253) , 1 ⁇ M (2.03 ⁇ 0.271), 3 ⁇ M (2.29 ⁇ 0.210), 10 ⁇ M (2.80 ⁇ 0.684).
  • TIC10 cannot promote apoptosis in drug-resistant myeloproliferative tumor cells.
  • the drug concentrations (average apoptosis rate) in the Ulixertinib treatment group are: 0 ⁇ M (2.62 ⁇ 0.210) (not shown in the figure), 0.1 ⁇ M (2.28 ⁇ 0.450), 0.3 ⁇ M (2.53 ⁇ 0.382) , 1 ⁇ M (2.39 ⁇ 0.399), 3 ⁇ M (3.11 ⁇ 0.891), 10 ⁇ M (3.29 ⁇ 0.284).
  • 0 ⁇ M (2.62 ⁇ 0.210) not shown in the figure
  • 0.1 ⁇ M (2.28 ⁇ 0.450 0.3 ⁇ M (2.53 ⁇ 0.382)
  • 1 ⁇ M (2.39 ⁇ 0.399) 1 ⁇ M (2.39 ⁇ 0.399
  • 10 ⁇ M (3.29 ⁇ 0.284) the drug concentrations (average apoptosis rate) in the Ulixertinib treatment group are: 0 ⁇ M (2.62 ⁇ 0.210) (not shown in the figure), 0.1 ⁇ M (2.28 ⁇ 0.450), 0.3 ⁇ M (2.53 ⁇
  • Figure 5i reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) of the PD184352 treatment group are: 0 ⁇ M (1.07 ⁇ 0.532) (not shown in the figure), 0.1 ⁇ M (0.650 ⁇ 0.157), 0.3 ⁇ M (0.990 ⁇ 0.153) , 1 ⁇ M (0.870 ⁇ 0.0560), 3 ⁇ M (0.970 ⁇ 0.0600), 10 ⁇ M (1.46 ⁇ 0.0250). This suggests that PD184352 cannot promote apoptosis of drug-resistant myeloproliferative tumor cells.
  • Figure 5j reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) in the Pimasertib treatment group are: 0 ⁇ M (1.55 ⁇ 0.372) (not shown in the figure), 0.1 ⁇ M (1.45 ⁇ 0.528), 0.3 ⁇ M (1.63 ⁇ 0.284) , 1 ⁇ M (1.43 ⁇ 0.189), 3 ⁇ M (1.60 ⁇ 0.217), 10 ⁇ M (1.77 ⁇ 0.167).
  • the comparison of apoptosis rates between the two groups in the figure uses t test (*p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001).

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 a use of an MEK/ERK signal pathway inhibitor in the preparation of a drug for treating myeloproliferative neoplasms. The MEK/ERK signaling pathway inhibitor is used for treating myeloproliferative neoplasms, providing a new treatment approach for patients with myeloproliferative neoplasms, and more choices for clinicians and patients. For patients with myeloproliferative neoplasms who are resistant to ruxolitinib, the MEK/ERK signaling pathway inhibitor can provide continuous oral drug treatment for the patients, without needing bone marrow transplantation. The MEK/ERK signal pathway inhibitor can be chemically synthesized, and the cost is lower than that of a biological preparation. In addition, the inhibitors in the present specification have all passed phase I clinical trial, and have light side reactions and good tolerance of clinical patients.

Description

MEK/ERK信号通路抑制剂在制备治疗骨髓增殖性肿瘤的药物中的应用Application of MEK/ERK signaling pathway inhibitors in the preparation of drugs for the treatment of myeloproliferative tumors
本申请要求于2022年7月19日提交中国专利局、申请号为202210846544.4、发明名称为“MEK/ERK信号通路抑制剂在制备治疗骨髓增殖性肿瘤的药物中的应用”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。This application requires priority for the Chinese patent application submitted to the China Patent Office on July 19, 2022, with the application number 202210846544.4 and the invention title "Application of MEK/ERK signaling pathway inhibitors in the preparation of drugs for the treatment of myeloproliferative tumors" rights, the entire contents of which are incorporated herein by reference.
技术领域Technical field
本发明涉及医药技术领域,特别涉及MEK/ERK信号通路抑制剂在制备治疗骨髓增殖性肿瘤的药物中的应用。The present invention relates to the field of medical technology, and in particular to the use of MEK/ERK signaling pathway inhibitors in the preparation of drugs for the treatment of myeloproliferative tumors.
背景技术Background technique
骨髓增殖性肿瘤(Myeloproliferative neoplasmas,MPNs)是指分化相对成熟的一系或多系骨髓细胞克隆性增殖所致的一组肿瘤性疾病。在临床表现为一种或多种血细胞增生,伴肝、脾或淋巴结肿大。2016年世界卫生组织(WHO)对骨髓肿瘤进行分类修订,将真性红细胞增多症(polycythemiavera,PV)、原发性骨髓纤维化(primary myelofibrosis,PMF)、原发性血小板增多症(essentialthrombocythemia,ET)归入费城阴性经典骨髓增殖性肿瘤(Philadelphia-negative classical骨髓增殖性肿瘤)的范畴。骨髓增殖性肿瘤为克隆性造血干细胞疾病,主要的疾病驱动基因突变包括JAK2/V617F、CALR、MPL突变,其中JAK2/V617F突变是最常见的类型,可见于95%的PV、50-60%的ET和55-65%的PMF患者。基因的活化带来JAK-STAT通路的激活从而导致疾病的发生。全球每年新增骨髓增殖性肿瘤患者约20万,给医疗卫生系统带来沉重的负担。Myeloproliferative neoplasms (MPNs) refer to a group of tumor diseases caused by the clonal proliferation of one or more lines of relatively mature bone marrow cells. The clinical manifestations are the proliferation of one or more blood cells, accompanied by enlargement of the liver, spleen or lymph nodes. In 2016, the World Health Organization (WHO) revised the classification of bone marrow tumors, including polycythemia vera (PV), primary myelofibrosis (PMF), and essential thrombocythemia (ET). Included in the category of Philadelphia-negative classic myeloproliferative neoplasms. Myeloproliferative neoplasms are clonal hematopoietic stem cell diseases. The main disease driver gene mutations include JAK2/V617F, CALR, and MPL mutations. Among them, JAK2/V617F mutation is the most common type and can be found in 95% of PV and 50-60% of cases. ET and 55-65% of patients with PMF. The activation of genes leads to the activation of the JAK-STAT pathway, which leads to the occurrence of diseases. There are approximately 200,000 new patients with myeloproliferative neoplasms worldwide every year, placing a heavy burden on the medical and health systems.
在芦可替尼(Ruxolitinib,RUX)面世之前,骨髓增殖性肿瘤的常用治疗药物包括羟基脲及聚乙二醇-重组干扰素-α2a。羟基脲只能缓解症状却不能抑制克隆性造血,长期使用可能会增加骨髓增生异常综合征和急性髓系白血病的风险。而干扰素则因存在较高的毒副反应限制了其使用。芦可替尼作为JAK1/JAK2抑制剂,被FDA批准一线用于对中、高危骨髓纤 维化(myelofibrosis,MF),并作为二线药物用于羟基脲(Hydroxyurea,HU)耐药或不能耐受的PV患者。二期、三期临床试验结果提示,与最佳疗法相比,RUX能够减少中、高危MF和PV患者的脾脏体积和减轻症状。但是芦可替尼在使用过程中也存在许多问题,COMFORT和RESPONSE临床试验结果显示,接受芦可替尼治疗的MF患者贫血更重。更严重的是,长时间使用RUX等I型JAK抑制剂可诱导耐药的发生,在接受治疗1年的MF病人中,超过40%病人出现耐药,在临床研究中也发现了几种JAK抑制剂之间的交叉耐药。2019年8月,美国FDA批准了新型口服JAK2选择性抑制剂Fedratinib用于成人中、高危原发性或继发性(PV后或ET后)MF,其中包括以前接受过芦可替尼治疗的患者。FDA同时予以黑框警示Fedratinib可能引起脑病,包括韦尼克脑病的风险。为了进一步评估Fedratinib的有效性及安全性,新的多中心IIIb期临床实验(NCT03755518)正在进行。目前的JAK抑制剂不能显著减少突变等位基因负荷,因此其治疗潜力有限。骨髓移植是唯一治愈骨髓增殖性肿瘤的方法,但是仍然有一些问题需要解决。移植方式和方案的选择还不确定,选择同种异体移植还是单倍体同种移植尚不清楚。此外,当选择移植时,必须考虑到移植相关死亡率和骨髓增殖性肿瘤的长期性。目前骨髓移植主要用于治疗高危的骨髓纤维化患者,但其他类型的骨髓增殖性肿瘤患者选择骨髓移植的时机需要进一步探讨和研究证实。骨髓移植费用昂贵,在目前的医疗环境下,对大多数患者来说无法选择该治疗手段。Before the advent of Ruxolitinib (RUX), commonly used therapeutic drugs for myeloproliferative tumors included hydroxyurea and polyethylene glycol-recombinant interferon-α2a. Hydroxyurea can only relieve symptoms but cannot inhibit clonal hematopoiesis. Long-term use may increase the risk of myelodysplastic syndrome and acute myeloid leukemia. However, the use of interferon is limited due to its high toxic and side effects. Ruxolitinib, a JAK1/JAK2 inhibitor, has been approved by the FDA as first-line treatment for intermediate and high-risk bone marrow fibrosis. Myelofibrosis (MF), and as a second-line drug for PV patients who are resistant or intolerant to hydroxyurea (Hydroxyurea, HU). The results of phase II and phase III clinical trials suggest that RUX can reduce spleen volume and symptoms in patients with intermediate and high-risk MF and PV compared with optimal therapy. However, there are many problems during the use of ruxolitinib. The results of the COMFORT and RESPONSE clinical trials showed that MF patients treated with ruxolitinib had more severe anemia. What’s more serious is that long-term use of type I JAK inhibitors such as RUX can induce the occurrence of drug resistance. Among MF patients who received treatment for one year, more than 40% of patients developed drug resistance. Several JAK inhibitors have also been found in clinical studies. Cross-resistance between inhibitors. In August 2019, the US FDA approved the new oral JAK2 selective inhibitor Fedratinib For adults with intermediate- to high-risk primary or secondary (post-PV or post-ET) MF, including patients previously treated with ruxolitinib. The FDA also issued a black box warning that Fedratinib may cause encephalopathy, including the risk of Wernicke's encephalopathy. To further evaluate the efficacy and safety of fedratinib, a new multicenter phase IIIb clinical trial (NCT03755518) is ongoing. Current JAK inhibitors are unable to significantly reduce mutant allele burden and therefore have limited therapeutic potential. Bone marrow transplantation is the only cure for myeloproliferative neoplasms, but there are still issues that need to be addressed. The choice of transplantation modality and regimen is uncertain, and the choice of allogeneic or haploidentical transplantation is unclear. Furthermore, when choosing transplantation, transplantation-related mortality and the long-term nature of myeloproliferative neoplasms must be considered. At present, bone marrow transplantation is mainly used to treat high-risk myelofibrosis patients, but the timing of bone marrow transplantation for patients with other types of myeloproliferative neoplasms needs further exploration and research to confirm. Bone marrow transplantation is expensive, and in the current medical environment, this treatment is not an option for most patients.
尽管芦可替尼是骨髓增殖性肿瘤治疗的里程碑药物,但芦可替尼目前适用的范围较窄,骨髓抑制作为常见的副反应限制了其在主要适应症MF中的应用。芦可替尼不能减少突变基因的负荷,这意味着芦可替尼治疗不能使疾病达到分子水平的缓解,无法从根本上治疗骨髓增殖性肿瘤。尤其是在出现芦可替尼耐药后,治疗药物有限是目前面临的重大挑战。Although ruxolitinib is a landmark drug in the treatment of myeloproliferative tumors, ruxolitinib currently has a narrow scope of application, and myelosuppression as a common side effect limits its application in the main indication MF. Ruxolitinib cannot reduce the burden of mutated genes, which means that ruxolitinib treatment cannot achieve molecular-level remission of the disease and cannot fundamentally treat myeloproliferative tumors. Especially after the emergence of ruxolitinib resistance, the limited number of therapeutic drugs is a major challenge.
发明内容Contents of the invention
有鉴于此,本发明的目的在于针对目前骨髓增殖性肿瘤治疗中存在的问题,对骨髓增殖性肿瘤病人尤其是对芦可替尼耐药的骨髓增殖性肿瘤病 人提供一种有效、安全、可靠的药物。In view of this, the purpose of the present invention is to address the problems existing in the current treatment of myeloproliferative neoplasms, and to treat patients with myeloproliferative neoplasms, especially myeloproliferative neoplasms that are resistant to ruxolitinib. People provide an effective, safe and reliable medicine.
为了实现上述发明目的,本发明提供以下技术方案:In order to achieve the above-mentioned object of the invention, the present invention provides the following technical solutions:
本发明提供了MEK/ERK信号通路抑制剂在制备治疗骨髓增殖性肿瘤的药物中的应用。The present invention provides the use of MEK/ERK signaling pathway inhibitors in preparing drugs for treating myeloproliferative tumors.
作为优选,所述骨髓增殖性肿瘤为对化疗药物具有耐药性的骨髓增殖性肿瘤。Preferably, the myeloproliferative tumor is a myeloproliferative tumor that is resistant to chemotherapy drugs.
作为优选,所述化疗药物为治疗骨髓增殖性肿瘤的化疗药物,所述化疗药物包括芦可替尼、菲卓替尼中的至少一种。Preferably, the chemotherapeutic drug is a chemotherapeutic drug for treating myeloproliferative tumors, and the chemotherapeutic drug includes at least one of ruxolitinib and fizotinib.
作为优选,所述骨髓增殖性肿瘤为真性红细胞增多症、原发性血小板增多症或骨髓纤维化,所述骨髓纤维化为原发性骨髓纤维化、继发于真性红细胞增多症的骨髓纤维化和继发于原发性血小板增多症的骨髓纤维化中的至少一种。Preferably, the myeloproliferative tumor is polycythemia vera, essential thrombocythemia or myelofibrosis, and the myelofibrosis is primary myelofibrosis or myelofibrosis secondary to polycythemia vera. and at least one of myelofibrosis secondary to essential thrombocythemia.
作为优选,MEK/ERK信号通路抑制剂为Trematinib、Cobimetinib、Binimetinib、Selumetinib、Mirdametinib,Refametinib、TIC10、Ulixertinib、PD184352、Pimasertib中的至少一种。Preferably, the MEK/ERK signaling pathway inhibitor is at least one of Trematinib, Cobimetinib, Binimetinib, Selumetinib, Mirdametinib, Refametinib, TIC10, Ulixertinib, PD184352, and Pimasertib.
Trametinib(GSK1120212,JTP-74057,Mekinist)是一种高特异性的,有效的MEK1/2抑制剂,2013年获美国FDA批准上市,适用于携带BRAF V600E或V600K突变的手术不可切除性黑色素瘤或转移性黑色素瘤成人患者的治疗。2014年1月8日,FDA批准了dabrafenib和trametinib联合治疗BRAF V600E/K突变型转移性黑色素瘤患者。2018年5月1日,FDA批准联合使用dabrafenib/trametinib作为辅助治疗。根据COMBI-AD3期研究结果,手术切除后BRAF V600E突变的III期黑色素瘤,使其成为首个口服化疗方案,可预防淋巴结阳性,BRAF突变黑色素瘤的癌症复发。Trametinib的分子式为C26H23FIN5O4,分子量为615.39,结构式如式I所示。
Trametinib (GSK1120212, JTP-74057, Mekinist) is a highly specific and effective MEK1/2 inhibitor that was approved by the US FDA in 2013. It is suitable for surgically unresectable melanoma or patients carrying BRAF V600E or V600K mutations. Treatment of adult patients with metastatic melanoma. On January 8, 2014, the FDA approved the combination of dabrafenib and trametinib for the treatment of patients with BRAF V600E/K mutant metastatic melanoma. On May 1, 2018, the FDA approved the combined use of dabrafenib/trametinib as adjuvant therapy. It is the first oral chemotherapy regimen to prevent cancer recurrence in node-positive, BRAF-mutated melanoma after surgical resection of BRAF V600E-mutated stage III melanoma, according to results from the Phase 3 COMBI-AD study. The molecular formula of Trametinib is C 26 H 23 FIN 5 O 4 , the molecular weight is 615.39, and the structural formula is shown in Formula I.
Cobimetinib(GDC-0973,RG7420)是一种有效的高选择性MEK1抑制剂,于2015年11月10日获FDA批准上市,与威罗菲尼联合治疗BRAF V600E或V600K突变的迁移性黑色素瘤。目前NCT03695380正在进行卵巢肿瘤治疗的I期临床试验招募。Cobimetinib的分子式为C21H21F3IN3O2,分子量为531.31,结构式如式II所示。
Cobimetinib (GDC-0973, RG7420) is an effective and highly selective MEK1 inhibitor that was approved by the FDA on November 10, 2015. It is used in combination with vemurafenib to treat migratory melanoma with BRAF V600E or V600K mutations. NCT03695380 is currently recruiting for a Phase I clinical trial for the treatment of ovarian tumors. The molecular formula of Cobimetinib is C 21 H 21 F 3 IN 3 O 2 , the molecular weight is 531.31, and the structural formula is shown in Formula II.
Binimetinib是有丝分裂原激活的细胞外信号调节激酶1(MEK1)和细胞因子的可逆抑制剂MEK2活动。MEK蛋白是细胞外信号相关激酶(ERK)的上游调节因子途径。在体外,Binimetinib抑制细胞中的细胞外信号相关激酶(ERK)磷酸化免疫测定以及BRAF-突变型人黑素瘤细胞的活力和MEK依赖性磷酸化。Binimetinib还抑制BRAF突变小鼠体内ERK磷酸化和肿瘤生长异种移植模型。FDA批准Binimetinib与Encorafenib联合用于治疗患有BRAF V600E或V600K突变的不可切除或转移性黑色素瘤患者。Binimetinib分子式为C17H15BrF2N4O3,分子量为441.23,结构式如式III所示。
Binimetinib is a reversible inhibitor of mitogen-activated extracellular signal-regulated kinase 1 (MEK1) and cytokine MEK2 activity. MEK protein is an upstream regulator of the extracellular signal-related kinase (ERK) pathway. In vitro, binimetinib inhibits extracellular signal-related kinase (ERK) phosphorylation immunoassays in cells as well as viability and MEK-dependent phosphorylation of BRAF-mutant human melanoma cells. Binimetinib also inhibits ERK phosphorylation and tumor growth in BRAF mutant mouse xenograft models. The FDA approved binimetinib in combination with encorafenib for the treatment of patients with unresectable or metastatic melanoma who have BRAF V600E or V600K mutations. The molecular formula of Binimetinib is C 17 H 15 BrF 2 N 4 O 3 , the molecular weight is 441.23, and the structural formula is shown in Formula III.
Selumetinib又称为AZD6244、Y-142886,是一种高效、非ATP竞争性MEK1/2、ERK1/2抑制剂。在多种肿瘤模型中,Selumetinib也具有明显的效力,显著抑制ERK活性,抑制肿瘤生长,并抑制肺转移。2020年4月10日,美国FDA宣布批准Koselugo(Selumetinib)胶囊用于治疗2岁及以上的I型神经纤维瘤(NF1)儿童患者。这是FDA批准的首款NF1治疗药物。该药具体适应症为:用于治疗患有症状性、无法手术的丛状神经纤维瘤的儿童患者。Selumetinib的分子式为C17H15BrClFN4O3
Selumetinib, also known as AZD6244 and Y-142886, is a highly efficient, non-ATP competitive inhibitor of MEK1/2 and ERK1/2. Selumetinib also has significant efficacy in multiple tumor models, significantly inhibiting ERK activity, inhibiting tumor growth, and inhibiting lung metastasis. On April 10, 2020, the U.S. FDA announced the approval of Koselugo (Selumetinib) capsules for the treatment of pediatric patients 2 years old and older with neurofibromatosis type I (NF1). This is the first FDA-approved treatment for NF1. The drug is specifically indicated for the treatment of pediatric patients with symptomatic, inoperable plexiform neurofibromas. The molecular formula of Selumetinib is C 17 H 15 BrClFN 4 O 3 ,
Mirdametinib是一种口服小分子MEK1和MEK2抑制剂。Mirdametinib欧盟委员会(EC)及美国FDA已授予Mirdametinib(前称PD-0325901)治疗1型神经纤维瘤病(NF1)的孤儿药资格。Mirdametinib的分子式为C16H14F3IN2O4,分子量为482.19,结构式如式V所示。
Mirdametinib is an oral, small molecule inhibitor of MEK1 and MEK2. Mirdametinib The European Commission (EC) and the US FDA have granted Mirdametinib (formerly known as PD-0325901) orphan drug designation for the treatment of neurofibromatosis type 1 (NF1). The molecular formula of Mirdametinib is C 16 H 14 F 3 IN 2 O 4 , the molecular weight is 482.19, and the structural formula is shown in Formula V.
Refametinib(RDEA119)是一种有效的,非ATP竞争性的,高选择性的MEK1和MEK2抑制剂,IC50分别为19nM和47nM。Refametinib是一种口服MEK抑制剂,与Sorafenib联合使用治疗RAS突变的肝细胞癌(HCC)的患者时具有抗肿瘤活性。CLIN CANCER RES曾发表了一篇文章,报道了Refametinib单药以及Refametinib联合Sorafenib治疗RAS突变无法切除或转移性HCC患者的疗效。Refametinib目前正在开展晚期胆道癌的II期临床研究。Refametinib的分子式为C19H20F3IN2O5S,分子量为572.34,结构式如式VI所示。
Refametinib (RDEA119) is a potent, non-ATP competitive, highly selective inhibitor of MEK1 and MEK2 with IC50 of 19nM and 47nM respectively. Refametinib is an oral MEK inhibitor with antitumor activity in combination with sorafenib for the treatment of patients with RAS-mutated hepatocellular carcinoma (HCC). CLIN CANCER RES once published an article reporting the efficacy of Refametinib alone and Refametinib combined with Sorafenib in the treatment of patients with unresectable or metastatic HCC with RAS mutations. Refametinib is currently undergoing a Phase II clinical study in advanced biliary tract cancer. The molecular formula of Refametinib is C 19 H 20 F 3 IN 2 O 5 S, the molecular weight is 572.34, and the structural formula is shown in Formula VI.
TIC10(ONC201)抑制Akt和ERK活性,通过FoxO3a诱导TNF-related apoptosis-inducing ligand(TRAIL),可以穿透血脑屏障,具有超强的稳定性,和改善的药代动力学性能。TIC10引起肿瘤细胞的细胞表 面上TRAIL的显著和长期的表达。在HCT116p53-/-细胞中,TIC10也导致TRAIL介导的细胞凋亡。此外,TIC10同时灭活Akt和ERK,从而导致Foxo3a的核易位和随后TRAIL的上调。在肿瘤异种移植小鼠中,TIC10具有TRAIL依赖性的抗肿瘤效果,通过TRAIL介导的直接和旁观者效应,引起肿瘤特异性的细胞死亡。该分子发现对多种实体肿瘤有效,已开展临床1、2期实验,目前由于化合物专利纠纷临床试验推进困难。TIC10的分子式为C24H26N4O,分子量为386.49,结构式如式VII所示。
TIC10 (ONC201) inhibits Akt and ERK activity, induces TNF-related apoptosis-inducing ligand (TRAIL) through FoxO3a, can penetrate the blood-brain barrier, has super stability, and improved pharmacokinetic properties. TIC10 causes tumor cell cell surface Significant and long-lasting expression of TRAIL on the face. In HCT116p53-/- cells, TIC10 also caused TRAIL-mediated apoptosis. Furthermore, TIC10 simultaneously inactivates Akt and ERK, leading to nuclear translocation of Foxo3a and subsequent upregulation of TRAIL. In tumor xenograft mice, TIC10 has TRAIL-dependent antitumor effects, causing tumor-specific cell death through TRAIL-mediated direct and bystander effects. This molecule has been found to be effective against a variety of solid tumors and has already carried out clinical phase 1 and 2 trials. Currently, it is difficult to advance clinical trials due to compound patent disputes. The molecular formula of TIC10 is C 24 H 26 N 4 O, the molecular weight is 386.49, and the structural formula is shown in Formula VII.
Ulixertinib(BVD-523,VRT752271)是有效的可逆ERK1/ERK2抑制剂,其抑制ERK2的IC50<0.3nM,可口服。Ulixertinib在针对晚期实体肿瘤患者的临床试验中效果良好。Ulixertinib的分子式为C21H22Cl2N4O2,分子量为433.33,结构式如式VIII所示。
Ulixertinib (BVD-523, VRT752271) is a potent reversible ERK1/ERK2 inhibitor with an IC50 of ERK2<0.3nM and can be taken orally. Ulixertinib has shown promising results in clinical trials in patients with advanced solid tumors. The molecular formula of Ulixertinib is C 21 H 22 Cl 2 N 4 O 2 , the molecular weight is 433.33, and the structural formula is shown as Formula VIII.
PD184352(CI-1040)是一种ATP非竞争性的MEK1/2抑制剂,细胞试验中IC50为17nM,对MEK1/2的选择性比MEK5高100倍。PD184352(CI-1040)可选择性地诱导凋亡。PD184352是最早进入临床试验的MEK 抑制剂,因其溶解性差、半衰期短、口服生物利用度低、个体差异大等问题停止于Ⅱ期临床试验。PD184352的分子式为C17H14ClF2IN2O2,分子量为478.67,结构式如式IX所示。
PD184352 (CI-1040) is an ATP non-competitive MEK1/2 inhibitor with an IC50 of 17nM in cell assays and is 100 times more selective for MEK1/2 than MEK5. PD184352(CI-1040) selectively induces apoptosis. PD184352 is the first MEK to enter clinical trials Inhibitors have been stopped in phase II clinical trials due to problems such as poor solubility, short half-life, low oral bioavailability, and large individual differences. The molecular formula of PD184352 is C 17 H 14 ClF 2 IN 2 O 2 , the molecular weight is 478.67, and the structural formula is shown in Formula IX.
Pimasertib(AS-703026,MSC1936369B,SAR 245509)是一种高度选择性的ATP非竞争性的可口服的MEK1/2变构抑制剂,在MM细胞系中IC50为5nM-2μM。Pimasertib已在大约900个患有各种肿瘤类型的患者中进行了10多项1/2期临床试验。Pimasertib与DAY101联合使用,用于治疗≥12岁且患有复发性、进行性或难治性实体瘤且MAPK途径异常的患者。Pimasertib的分子式为C15H15FIN3O3,分子量为431.20,结构式如式X所示。
Pimasertib (AS-703026, MSC1936369B, SAR 245509) is a highly selective, ATP-noncompetitive, orally available MEK1/2 allosteric inhibitor with an IC50 of 5nM-2μM in MM cell lines. Pimasertib has been studied in more than 10 Phase 1/2 clinical trials in approximately 900 patients with various tumor types. Pimasertib, in combination with DAY101, is indicated for the treatment of patients ≥12 years of age with recurrent, progressive, or refractory solid tumors with MAPK pathway aberrations. The molecular formula of Pimasertib is C 15 H 15 FIN 3 O 3 , the molecular weight is 431.20, and the structural formula is shown in Formula X.
作为优选,所述治疗包括抑制肿瘤细胞的增殖和/或促进肿瘤细胞的 凋亡。Preferably, the treatment includes inhibiting the proliferation of tumor cells and/or promoting the growth of tumor cells. Apoptosis.
本发明还提供了MEK/ERK信号通路抑制剂在制备抑制HEL细胞的增殖和/或促进HEL细胞凋亡的药物中的应用。The present invention also provides the use of MEK/ERK signaling pathway inhibitors in preparing drugs that inhibit the proliferation of HEL cells and/or promote the apoptosis of HEL cells.
其中,所述HEL细胞包括非耐药性HEL细胞和/或耐药性的HEL细胞。Wherein, the HEL cells include non-drug-resistant HEL cells and/or drug-resistant HEL cells.
实验表明,对于非耐药性HEL细胞,Trematinib、Cobimetinib、Binimetinib、Selumetinib、Mirdametinib,Refametinib、Ulixertinib、PD184352、Pimasertib均不能促进HEL细胞的凋亡,仅TIC10可促进其凋亡。其中,Trematinib和TIC10能够抑制HEL细胞的增殖,Pimasertib有较弱的抑制HEL细胞增殖的作用,其余抑制剂没有抑制作用。Experiments have shown that for non-drug-resistant HEL cells, Trematinib, Cobimetinib, Binimetinib, Selumetinib, Mirdametinib, Refametinib, Ulixertinib, PD184352, and Pimasertib cannot promote apoptosis of HEL cells, and only TIC10 can promote apoptosis. Among them, Trematinib and TIC10 can inhibit the proliferation of HEL cells, Pimasertib has a weak inhibitory effect on HEL cell proliferation, and the other inhibitors have no inhibitory effect.
对于耐药性HEL细胞,Trematinib、Cobimetinib、Binimetinib、Selumetinib、Mirdametinib,Refametinib、TIC10、Ulixertinib、PD184352、Pimasertib均可抑制耐药性HEL细胞的增殖,其中,仅Refametinib可促进耐药性HEL细胞的凋亡。说明,Refametinib对耐药性骨髓增殖性肿瘤的治疗效果更为显著。For drug-resistant HEL cells, Trematinib, Cobimetinib, Binimetinib, Selumetinib, Mirdametinib, Refametinib, TIC10, Ulixertinib, PD184352, and Pimasertib can all inhibit the proliferation of drug-resistant HEL cells. Among them, only Refametinib can promote the apoptosis of drug-resistant HEL cells. Death. This shows that Refametinib has a more significant therapeutic effect on drug-resistant myeloproliferative tumors.
以上结果显示,MEK/ERK信号通路抑制剂对耐药性HEL细胞的增殖具有较强的抑制作用,部分抑制剂还可促进细胞的凋亡。这表明MEK/ERK信号通路抑制剂对耐药性骨髓增殖性肿瘤的治疗效果显著。The above results show that MEK/ERK signaling pathway inhibitors have a strong inhibitory effect on the proliferation of drug-resistant HEL cells, and some inhibitors can also promote cell apoptosis. This indicates that MEK/ERK signaling pathway inhibitors have a significant therapeutic effect on drug-resistant myeloproliferative tumors.
作为优选,所述药物还包含用于治疗骨髓增殖性肿瘤的其他药物成分以及药学上可接受的辅料。Preferably, the medicine also contains other pharmaceutical ingredients for treating myeloproliferative tumors and pharmaceutically acceptable excipients.
作为优选,药物的剂型为口服制剂或注射制剂。Preferably, the pharmaceutical dosage form is an oral preparation or an injection preparation.
由上述技术方案可知,本发明提供了MEK/ERK信号通路抑制剂在制备治疗骨髓增殖性肿瘤的药物中的应用。所述骨髓增殖性肿瘤为真性红细胞增多症、原发性血小板增多症或骨髓纤维化以及芦可替尼耐药性的骨髓增殖性肿瘤,所述骨髓纤维化为原发性骨髓纤维化、继发于真性红细胞增多症的骨髓纤维化或继发于原发性血小板增多症的骨髓纤维化。本发明具有的技术效果为:It can be seen from the above technical solutions that the present invention provides the use of MEK/ERK signaling pathway inhibitors in the preparation of drugs for the treatment of myeloproliferative tumors. The myeloproliferative tumors are polycythemia vera, essential thrombocythemia, or myelofibrosis and ruxolitinib-resistant myeloproliferative tumors, and the myelofibrosis is primary myelofibrosis, secondary myelofibrosis, or ruxolitinib-resistant myeloproliferative tumors. Myelofibrosis arising from polycythemia vera or secondary to essential thrombocythemia. The technical effects of the present invention are:
MEK/ERK信号通路抑制剂用于骨髓增殖性肿瘤的治疗给广大骨髓增殖性肿瘤的患者提供了新的治疗途径,给临床医生及患者提供了更多选 择。对于芦可替尼耐药的骨髓增殖性肿瘤患者,MEK/ERK信号通路抑制剂能为患者提供继续的口服药物治疗,免于接受骨髓移植。MEK/ERK信号通路抑制剂可以化学合成,成本较生物制剂要低。且申请中的MEK/ERK信号通路抑制剂均已经通过I期临床试验,部分已顺利通过II、III期临床试验,将来用于临床治疗,具有较好的临床应用前景。The use of MEK/ERK signaling pathway inhibitors in the treatment of myeloproliferative tumors provides a new treatment approach for patients with myeloproliferative tumors and provides more options for clinicians and patients. select. For patients with ruxolitinib-resistant myeloproliferative tumors, MEK/ERK signaling pathway inhibitors can provide patients with continued oral drug therapy and avoid bone marrow transplantation. MEK/ERK signaling pathway inhibitors can be chemically synthesized and the cost is lower than biological agents. Moreover, the MEK/ERK signaling pathway inhibitors under application have all passed Phase I clinical trials, and some have successfully passed Phase II and III clinical trials. They will be used in clinical treatments in the future and have good clinical application prospects.
附图说明Description of drawings
图1示实施例1:芦可替尼耐药的骨髓增殖性肿瘤细胞模型HELRE的建立结果图;Figure 1 shows Example 1: The establishment results of ruxolitinib-resistant myeloproliferative tumor cell model HEL RE ;
图2示实施例2:MEK/ERK信号通路抑制剂处理骨髓增殖性肿瘤细胞,通过CellTiter-LumiTM发光法检测细胞增殖的结果图;a为Trematinib处理HEL细胞增殖结果图;b为Cobimetinib处理HEL细胞增殖结果图;c为Binimetinib处理HEL细胞增殖结果图;d为Selumetinib处理HEL细胞增殖结果图;e为Mirdametinib处理HEL细胞增殖结果图;f为Refametinib处理HEL细胞增殖结果图;g为TIC10处理HEL细胞增殖结果图;h为Ulixertinib处理HEL细胞增殖结果图;i为PD184352处理HEL细胞增殖结果图;j为Pimasertib处理HEL细胞增殖结果图;Figure 2 shows the results of Example 2: MEK/ERK signaling pathway inhibitors treated myeloproliferative tumor cells, and the cell proliferation was detected by the CellTiter-Lumi TM luminescence method; a is the result of HEL cell proliferation treated with Trematinib; b is the result of HEL cells treated with Cobimetinib. Cell proliferation results; c is Binimetinib-treated HEL cell proliferation results; d is Selumetinib-treated HEL cell proliferation results; e is Mirdametinib-treated HEL cell proliferation results; f is Refametinib-treated HEL cell proliferation results; g is TIC10-treated HEL. The cell proliferation result chart; h is the HEL cell proliferation result chart treated with Ulixertinib; i is the HEL cell proliferation result chart treated with PD184352; j is the HEL cell proliferation result chart treated with Pimasertib;
图3示实施例3:MEK/ERK信号通路抑制剂处理骨髓增殖性肿瘤细胞,使用AnnexinV-PI染色后流式检测细胞凋亡的结果图;a为Trematinib处理HEL细胞凋亡结果图;b为Cobimetinib处理HEL细胞凋亡结果图;c为Binimetinib处理HEL细胞凋亡结果图;d为Selumetinib处理HEL细胞凋亡结果图;e为Mirdametinib处理HEL细胞凋亡结果图;f为Refametinib处理HEL细胞凋亡结果图;g为TIC10处理HEL细胞凋亡结果图;h为Ulixertinib处理HEL细胞凋亡结果图;i为PD184352处理HEL细胞凋亡结果图;j为Pimasertib处理HEL细胞凋亡结果图;Figure 3 shows the results of Example 3: MEK/ERK signaling pathway inhibitors treated myeloproliferative tumor cells, using AnnexinV-PI staining to detect cell apoptosis by flow cytometry; a is the result of Trematinib treatment of HEL cell apoptosis; b is the result of apoptosis of HEL cells treated with Trematinib; Cobimetinib treated HEL cell apoptosis results; c is Binimetinib treated HEL cell apoptosis results; d is Selumetinib treated HEL cell apoptosis results; e is Mirdametinib treated HEL cell apoptosis results; f is Refametinib treated HEL cell apoptosis Result graph; g is the graph of the apoptosis result of HEL cells treated with TIC10; h is the graph of the apoptosis result of HEL cells treated with Ulixertinib; i is the graph of the apoptosis result of HEL cells treated with PD184352; j is the graph of the apoptosis result of HEL cells treated with Pimasertib;
图4示实施例4:MEK/ERK信号通路抑制剂处理芦可替尼耐药的骨髓增殖性肿瘤细胞,通过CellTiter-LumiTM发光法检测细胞增殖的结果图;a为Trematinib处理HELRE细胞增殖结果图;b为Cobimetinib处理HELRE细胞增殖结果图;c为Binimetinib处理HELRE细胞增殖结果图;d为Selumetinib 处理HELRE细胞增殖结果图;e为Mirdametinib处理HELRE细胞增殖结果图;f为Refametinib处理HELRE细胞增殖结果图;g为TIC10处理HELRE细胞增殖结果图;h为Ulixertinib处理HELRE细胞增殖结果图;i为PD184352处理HELRE细胞增殖结果图;j为Pimasertib处理HELRE细胞增殖结果图;Figure 4 shows Example 4: MEK/ERK signaling pathway inhibitors treated ruxolitinib-resistant myeloproliferative tumor cells, and the results of detecting cell proliferation by CellTiter-Lumi TM luminescence assay; a shows the proliferation of HEL RE cells treated with Trematinib. Result chart; b is the result chart of HEL RE cell proliferation treated with Cobimetinib; c is the result chart of HEL RE cell proliferation treated with Binimetinib; d is Selumetinib The result of HEL RE cell proliferation after treatment; e is the result of HEL RE cell proliferation treated with Mirdametinib; f is the result of HEL RE cell proliferation treated with Refametinib; g is the result of HEL RE cell proliferation treated with TIC10; h is the result of HEL RE cell proliferation treated with Ulixertinib. Figure; i is the result of HEL RE cell proliferation treated with PD184352; j is the result of HEL RE cell proliferation treated with Pimasertib;
图5示实施例5:MEK/ERK信号通路抑制剂处理芦可替尼耐药的骨髓增殖性肿瘤细胞,使用AnnexinV-PI染色后流式检测细胞凋亡的结果图;a为Trematinib处理HELRE细胞凋亡结果图;b为Cobimetinib处理HELRE细胞凋亡结果图;c为Binimetinib处理HELRE细胞凋亡结果图;d为Selumetinib处理HELRE细胞凋亡结果图;e为Mirdametinib处理HELRE细胞凋亡结果图;f为Refametinib处理HELRE细胞凋亡结果图;g为TIC10处理HELRE细胞凋亡结果图;h为Ulixertinib处理HELRE细胞凋亡结果图;i为PD184352处理HELRE细胞凋亡结果图;j为Pimasertib处理HELRE细胞凋亡结果图。Figure 5 shows Example 5: MEK/ERK signaling pathway inhibitors treated ruxolitinib-resistant myeloproliferative tumor cells, and the results of flow cytometry detection of cell apoptosis after staining with AnnexinV-PI; a is Trematinib treatment of HEL RE Cell apoptosis results; b is Cobimetinib-treated HEL RE cell apoptosis results; c is Binimetinib-treated HEL RE cell apoptosis results; d is Selumetinib-treated HEL RE cell apoptosis results; e is Mirdametinib-treated HEL RE cell apoptosis f is the result of apoptosis of HEL RE cells treated with Refametinib; g is the result of apoptosis of HEL RE cells treated with TIC10; h is the result of apoptosis of HEL RE cells treated with Ulixertinib; i is the result of apoptosis of HEL RE cells treated with PD184352 Figure; j shows the apoptosis results of HEL RE cells treated with Pimasertib.
具体实施方式Detailed ways
本发明公开了MEK/ERK信号通路抑制剂在制备治疗骨髓增殖性肿瘤的药物中的应用,本领域技术人员可以借鉴本文内容,适当改进工艺参数实现。特别需要指出的是,所有类似的替换和改动对本领域技术人员来说是显而易见的,它们都被视为包括在本发明。本发明的方法及应用已经通过较佳实施例进行了描述,相关人员明显能在不脱离本发明内容、精神和范围内对本文所述的方法和应用进行改动或适当变更与组合,来实现和应用本发明技术。The present invention discloses the application of MEK/ERK signaling pathway inhibitors in the preparation of drugs for the treatment of myeloproliferative tumors. Persons skilled in the art can learn from the contents of this article and appropriately improve the process parameters for implementation. It should be noted that all similar substitutions and modifications are obvious to those skilled in the art, and they are deemed to be included in the present invention. The methods and applications of the present invention have been described through preferred embodiments. Relevant persons can obviously make modifications or appropriate changes and combinations to the methods and applications described herein without departing from the content, spirit and scope of the present invention to achieve and Apply the technology of this invention.
在本发明中,通过细胞系模型来明确MEK/ERK信号通路抑制剂对骨髓增殖性肿瘤细胞(耐药与非耐药)的抑制作用。In the present invention, the inhibitory effect of MEK/ERK signaling pathway inhibitors on myeloproliferative tumor cells (drug-resistant and non-drug-resistant) is clarified through a cell line model.
在一些实施方案中,本发明基于常用的两种含JAK2-V617F突变的人源骨髓增殖性肿瘤细胞系,即HEL细胞(Human erythroleukemia cell line,人红白血病细胞),建立了芦可替尼耐药细胞模型HELRE。耐药模型的构建方法为使用低于细胞IC50浓度开始加芦可替尼,缓慢递增至高浓度,维持细胞不被杀灭,通过比较IC50来验证模型的构建是否成功。In some embodiments, the present invention establishes ruxolitinib resistance based on two commonly used human myeloproliferative tumor cell lines containing JAK2-V617F mutations, namely HEL cells (Human erythroleukemia cell line, human erythroleukemia cells). Drug Cell Model HEL RE . The drug resistance model is constructed using Start adding ruxolitinib at the IC50 concentration of the cells and slowly increase it to a high concentration to keep the cells from being killed. Compare the IC50 to verify whether the model is successfully constructed.
在一些实施方案中,本发明使用递增浓度的MEK/ERK信号通路抑制 剂处理骨髓增殖性肿瘤细胞株,通过CellTiter-LumiTM发光法检测细胞的增殖。结果显示,部分MEK/ERK信号通路抑制剂可成功抑制HEL细胞的增殖。In some embodiments, the present invention uses increasing concentrations of the MEK/ERK signaling pathway to inhibit The myeloproliferative tumor cell lines were treated with the agent, and the cell proliferation was detected by CellTiter-Lumi TM luminescence method. The results show that some MEK/ERK signaling pathway inhibitors can successfully inhibit the proliferation of HEL cells.
在一些实施方案中,本发明使用递增浓度的MEK/ERK信号通路抑制剂处理骨髓增殖性肿瘤细胞株,使用AnnexinV-PI染色后流式检测细胞的凋亡情况。结果显示,部分MEK/ERK信号通路抑制剂可促进HEL细胞的凋亡。In some embodiments, the present invention uses increasing concentrations of MEK/ERK signaling pathway inhibitors to treat myeloproliferative tumor cell lines, and uses AnnexinV-PI staining to detect cell apoptosis using flow cytometry. The results show that some MEK/ERK signaling pathway inhibitors can promote the apoptosis of HEL cells.
由此可见,部分MEK/ERK信号通路抑制剂可用于治疗骨髓增殖性肿瘤疾病。It can be seen that some MEK/ERK signaling pathway inhibitors can be used to treat myeloproliferative tumor diseases.
进一步的,在一些实施方案中,本发明使用递增浓度的MEK/ERK信号通路抑制剂处理芦可替尼耐药的骨髓增殖性肿瘤细胞,通过CellTiter-LumiTM发光法检测细胞的增殖。结果显示,MEK/ERK信号通路抑制剂可抑制HELRE细胞的增殖。Further, in some embodiments, the present invention uses increasing concentrations of MEK/ERK signaling pathway inhibitors to treat ruxolitinib-resistant myeloproliferative tumor cells, and detect cell proliferation through the CellTiter-Lumi TM luminescence method. The results showed that inhibitors of the MEK/ERK signaling pathway could inhibit the proliferation of HEL RE cells.
在一些实施方案中,本发明使用递增浓度的MEK/ERK信号通路抑制剂处理芦可替尼耐药的骨髓增殖性肿瘤细胞,使用AnnexinV-PI染色后流式检测细胞的凋亡情况。结果显示,部分MEK/ERK信号通路抑制剂可促进HELRE细胞的凋亡。In some embodiments, the present invention uses increasing concentrations of MEK/ERK signaling pathway inhibitors to treat ruxolitinib-resistant myeloproliferative tumor cells, and uses AnnexinV-PI staining to detect the apoptosis of the cells by flow cytometry. The results show that some MEK/ERK signaling pathway inhibitors can promote the apoptosis of HEL RE cells.
由此可见,MEK/ERK信号通路抑制剂可用于治疗芦可替尼耐药的骨髓增殖性肿瘤疾病。It can be seen that MEK/ERK signaling pathway inhibitors can be used to treat ruxolitinib-resistant myeloproliferative tumor diseases.
进一步的,本发明提供了部分MEK/ERK信号通路抑制剂在制备抑制HEL细胞的增殖、促进HEL细胞的凋亡的药物中的应用。Furthermore, the present invention provides the use of some MEK/ERK signaling pathway inhibitors in the preparation of drugs that inhibit the proliferation of HEL cells and promote the apoptosis of HEL cells.
综上所述,本发明提供了MEK/ERK信号通路抑制剂在制备治疗骨髓增殖性肿瘤的药物中的应用。In summary, the present invention provides the use of MEK/ERK signaling pathway inhibitors in the preparation of drugs for treating myeloproliferative tumors.
进一步的,所述骨髓增殖性肿瘤为真性红细胞增多症、原发性血小板增多症和骨髓纤维化(包括原发性骨髓纤维化、继发于真性红细胞增多症的骨髓纤维化和继发于原发性血小板增多症的骨髓纤维化)和具有耐药性的骨髓增殖性肿瘤。Further, the myeloproliferative tumors are polycythemia vera, essential thrombocythemia and myelofibrosis (including primary myelofibrosis, myelofibrosis secondary to polycythemia vera and myelofibrosis secondary to primary myelofibrosis). Thrombocytosis, myelofibrosis) and drug-resistant myeloproliferative neoplasms.
在一些实施方案中,所述具有耐药性的骨髓增殖性肿瘤为芦可替尼耐药性的骨髓增殖性肿瘤。 In some embodiments, the drug-resistant myeloproliferative neoplasm is a ruxolitinib-resistant myeloproliferative neoplasm.
在一些实施方案中,所述具有耐药性的骨髓增殖性肿瘤为具有耐药性的真性红细胞增多症、具有耐药性的骨髓纤维化(包括原发性骨髓纤维化、继发于真性红细胞增多症的骨髓纤维化和继发于原发性血小板增多症的骨髓纤维化)以及具有耐药性的原发性血小板增多症。In some embodiments, the drug-resistant myeloproliferative neoplasm is drug-resistant polycythemia vera, drug-resistant myelofibrosis (including primary myelofibrosis, secondary myelofibrosis Polycythemia myelofibrosis and myelofibrosis secondary to essential thrombocythemia) and drug-resistant essential thrombocythemia.
其中,所述药物为Trematinib,Cobimetinib,Binimetinib,Selumetinib,Mirdametinib,Refametinib,TIC10,Ulixertinib,PD184352,Pimasertib。Among them, the drugs are Trematinib, Cobimetinib, Binimetinib, Selumetinib, Mirdametinib, Refametinib, TIC10, Ulixertinib, PD184352, and Pimasertib.
进一步的,所述药物还包括药学上可接受的辅料。Furthermore, the medicine also includes pharmaceutically acceptable excipients.
所述药物可以为当前药品领域任何剂型,包括口服制剂或注射制剂。The medicine can be in any dosage form in the current pharmaceutical field, including oral preparations or injection preparations.
各药物剂型可根据该剂型实际需要选取合适的可接受辅料来制备,这属于本领域常规的剂型制备技术。如制成胶囊剂、片剂、注射粉剂等。Each pharmaceutical dosage form can be prepared by selecting appropriate acceptable excipients according to the actual needs of the dosage form, which is a conventional dosage form preparation technology in this field. Such as making capsules, tablets, injection powders, etc.
本发明中所用试剂或仪器均可由市场购得。All reagents or instruments used in the present invention can be purchased from the market.
下面结合实施例,进一步阐述本发明:The present invention will be further described below in conjunction with the examples:
实施例1、两种常见芦可替尼耐药细胞模型的建立(HELRE)。Example 1. Establishment of two common ruxolitinib-resistant cell models (HEL RE ).
一、材料与方法1. Materials and methods
1、细胞系1. Cell line
HEL(Human erythroleukemia cell line)、芦可替尼耐药的HEL细胞均培养于含20%热灭活胎牛血清(Gibco)及1%青霉素/链霉素的RPMI培养基(Gibco)。HEL (Human erythroleukemia cell line) and ruxolitinib-resistant HEL cells were cultured in RPMI medium (Gibco) containing 20% heat-inactivated fetal calf serum (Gibco) and 1% penicillin/streptomycin.
芦可替尼耐药的HEL模型即HELRE模型,构建方法为使用低于原始细胞IC50浓度开始加芦可替尼,缓慢递增至高浓度,维持细胞不被杀灭。我们的起始浓度为0.1μM,细胞出现增殖就加药,加药梯度为1.25倍递增,终浓度为2.0μM。4-6周后获得稳定的耐药细胞。The ruxolitinib-resistant HEL model is the HEL RE model. The construction method is to start adding ruxolitinib at a concentration lower than the IC50 of the original cells and slowly increase it to a high concentration to maintain the cells from being killed. Our starting concentration is 0.1 μM, and the drug is added when cells proliferate. The drug addition gradient is 1.25 times increments, and the final concentration is 2.0 μM. Stable drug-resistant cells were obtained after 4-6 weeks.
2、抑制剂2. Inhibitors
芦可替尼及MEK/ERK信号通路抑制剂均购自Selleck公司,溶于DMSO,母液浓度为10mM,冻存于-80℃,工作液采用RPMI培养基稀释至指定倍数后处理细胞。芦可替尼具体为磷酸芦可替尼。Ruxolitinib and MEK/ERK signaling pathway inhibitors were purchased from Selleck Company, dissolved in DMSO, the concentration of the mother solution was 10mM, and frozen at -80°C. The working solution was diluted to the specified multiple with RPMI medium before processing the cells. Ruxolitinib is specifically ruxolitinib phosphate.
3、体外抑制试验3. In vitro inhibition test
为了检测抑制剂的抗增殖效应,上述细胞系以每孔3000个细胞/100 μL体系培养,加入递增浓度的芦可替尼(HEL细胞浓度梯度:0,0.1,0.3,1,3,10μM;或0,0.001,0.003,0.01,0.03,0.1μM),DMSO补齐至等量。设4个平行重复组,并设3个空白孔(不含细胞的培养液孔)。48小时后通过CellTiter-LumiTM发光法(碧云天)检测细胞的增殖。多功能酶标仪读数,IC50通过GraphPadprism计算得来。In order to detect the anti-proliferative effect of inhibitors, the above cell lines were cultured at 3000 cells/100 cells per well. μL system culture, add increasing concentrations of ruxolitinib (HEL cell concentration gradient: 0, 0.1, 0.3, 1, 3, 10 μM; or 0, 0.001, 0.003, 0.01, 0.03, 0.1 μM), and DMSO is added until equal. quantity. Set up 4 parallel replicate groups and set up 3 blank wells (wells containing culture medium without cells). After 48 hours, the cell proliferation was detected by CellTiter-Lumi TM luminescence method (Beyotime). Multifunctional microplate reader reading, IC50 is calculated by GraphPadprism.
细胞增殖率计算公式:细胞增殖率=(加药组Luminescence值-空白孔平均Luminescence值)/(DMSO对照组Luminescence值-空白孔平均Luminescence值)×100%。Calculation formula for cell proliferation rate: cell proliferation rate = (Luminescence value of drug-added group - average Luminescence value of blank wells) / (Luminescence value of DMSO control group - average Luminescence value of blank wells) × 100%.
评价模型是否构建成功的方法是比较耐药细胞与原始细胞的IC50,比值为耐药指数,大于3即为构建成功。The method to evaluate whether the model is successfully constructed is to compare the IC50 of drug-resistant cells and original cells. The ratio is the drug resistance index. If the ratio is greater than 3, the construction is successful.
二、结果分析2. Result analysis
图1中,HEL细胞IC50为0.801μM,HELRE细胞IC50为12.27μM,其耐药指数为15.3,提示耐药模型HELRE的成功构建。图中增殖率结果显示为平均数±标准差,图中两组增殖率的比较采用t检验(***p<0.001),IC50显示为均数。In Figure 1, the IC50 of HEL cells is 0.801 μM, the IC50 of HEL RE cells is 12.27 μM, and its drug resistance index is 15.3, indicating the successful construction of the drug resistance model HEL RE . The proliferation rate results in the figure are shown as the mean ± standard deviation. The comparison of the proliferation rates of the two groups in the figure is using t test (***p<0.001), and the IC50 is shown as the mean.
实施例2、部分MEK/ERK信号通路抑制剂可抑制骨髓增殖性肿瘤细胞的增殖Example 2. Some MEK/ERK signaling pathway inhibitors can inhibit the proliferation of myeloproliferative tumor cells.
为了检测MEK/ERK信号通路抑制剂对骨髓增殖性肿瘤耐药细胞增殖能力的影响,方法为分别使用递增浓度的芦可替尼及MEK/ERK信号通路抑制剂处理HEL原始细胞株,通过CellTiter-LumiTM发光法检测细胞的增殖,方法同实施例1,结果见图2。In order to detect the effect of MEK/ERK signaling pathway inhibitors on the proliferation of drug-resistant myeloproliferative tumor cells, the method was to use increasing concentrations of ruxolitinib and MEK/ERK signaling pathway inhibitors to treat the HEL original cell line, and then use CellTiter- The Lumi TM luminescence method was used to detect cell proliferation. The method was the same as in Example 1. The results are shown in Figure 2.
图2反映了在HEL细胞中,芦可替尼处理组药物浓度(平均增殖率%±标准差)为:0μM(100±1.47)(图中未显示)、0.1μM(81.0±1.35)、0.3μM(54.9±4.35)、1μM(42.7±4.05)、3μM(34.7±1.14)、10μM(29.4±0.497);图2a反映在HEL细胞中,Trematinib处理组药物浓度(平均增殖率)为:0μM(100±9.58)(图中未显示)、0.0001μM(93.91±2.37)、0.0003μM(98.17±3.44)、0.01μM(84.82±6.67)、0.03μM(83.96±9.48)、0.1μM(85.99±6.9)。这提示Trematinib可抑制HEL细胞增殖的作用,效果 弱于芦可替尼。图2b反映在HEL细胞中,Cobimetinib处理组药物浓度(平均增殖率)为:0μM(100±6.6)(图中未显示)、0.001μM(98.53±4.64)、0.003μM(105.51±7.14)、0.01μM(99.78±5.17)、0.03μM(99.66±1.93)、0.1μM(96.59±8.57)。这提示Cobimetinib不能抑制HEL细胞的增殖。图2c反映Binimetinib处理组药物浓度(平均增殖率)为:0μM(100±2.65)(图中未显示)、0.1μM(111±13.9)、0.3μM(105±17.2)、1μM(115±22.9)、3μM(107±14.4)、10μM(92.4±2.44)。这提示Binimetinib不能抑制HEL细胞的增殖。图2d反映了在HEL细胞中,Selumetinib处理组药物浓度(平均增殖率)为:0μM(100±1.62)(图中未显示)、0.1μM(107±17.6)、0.3μM(117±16.3)、1μM(110±26.3)、3μM(103±14.5)、10μM(88.8±2.69)。这提示Selumetinib不能抑制HEL细胞的增殖。图2e反映了在HEL细胞中,Mirdametinib处理组药物浓度(平均增殖率)为:0μM(100±1.73)(图中未显示)、0.1μM(107±17.6)、0.3μM(117±16.3)、1μM(110±26.3)、3μM(103±14.5)、10μM(88.8±2.69)。这提示Mirdametinib不能抑制HEL细胞的增殖。图2f反映在HEL细胞中,Refametinib处理组药物浓度(平均增殖率)为:0μM(100±2.63)(图中未显示)、0.1μM(96.0±7.44)、0.3μM(99.8±4.36)、1μM(93.4±4.31)、3μM(89.1±7.23)、10μM(84.6±8.89)。这提示Refametinib不能抑制HEL细胞的增殖。图2g反映在HEL细胞中,TIC10处理组药物浓度(平均增殖率)为:0μM(100±4.31)(图中未显示)、0.1μM(113±15.0)、0.3μM(116±14.9)、1μM(80.3±3.43)、3μM(19.6±2.64)、10μM(11.1±7.15)。这提示TIC10能抑制HEL细胞的增殖,该效应随药物浓度增加而增加,其抑制效果随浓度的增加而加强。图2h反映在HEL细胞中,Ulixertinib处理组药物浓度(平均增殖率)为:0μM(100±2.58)(图中未显示)、0.1μM(98.9±8.68)、0.3μM(109±5.36)、1μM(96.1±5.13)、3μM(100±4.61)、10μM(95.2±5.83)。这提示Ulixertinib不能抑制HEL细胞的增殖。图2i反映在HEL细胞中,PD184352处理组药物浓度(平均增殖率)为:0μM(100±4.46)(图中未显示)、0.1μM(105±16.7)、0.3μM(118±21.2)、1μM(103±22.7)、3μM(99.0±20.3)、10μM(82.3 ±6.50)。这提示PD184352不能抑制HEL细胞的增殖。图2j反映在HEL细胞中,Pimasertib处理组药物浓度(平均增殖率)为:0μM(100±3.07)(图中未显示)、0.1μM(89.0±1.42)、0.3μM(95.0±7.62)、1μM(86.9±1.71)、3μM(82.8±2.13)、10μM(77.3±2.52)。这提示Pimasertib有较弱的抑制HEL细胞增殖的作用,效果弱于芦可替尼。图中两组增殖率的比较采用t检验(*p<0.05,**p<0.01,***p<0.001)。Figure 2 reflects that in HEL cells, the drug concentrations (average proliferation rate % ± standard deviation) in the ruxolitinib-treated group are: 0 μM (100 ± 1.47) (not shown in the figure), 0.1 μM (81.0 ± 1.35), 0.3 μM (54.9±4.35), 1μM (42.7±4.05), 3μM (34.7±1.14), 10μM (29.4±0.497); Figure 2a reflects that in HEL cells, the drug concentration (average proliferation rate) of the Trematinib treatment group is: 0μM ( 100±9.58) (not shown in the figure), 0.0001μM (93.91±2.37), 0.0003μM (98.17±3.44), 0.01μM (84.82±6.67), 0.03μM (83.96±9.48), 0.1μM (85.99±6.9) . This suggests that Trematinib can inhibit the proliferation of HEL cells. Weaker than ruxolitinib. Figure 2b reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Cobimetinib treatment group are: 0 μM (100±6.6) (not shown in the figure), 0.001 μM (98.53±4.64), 0.003 μM (105.51±7.14), 0.01 μM (99.78±5.17), 0.03μM (99.66±1.93), 0.1μM (96.59±8.57). This suggests that Cobimetinib cannot inhibit the proliferation of HEL cells. Figure 2c reflects that the drug concentrations (average proliferation rate) of the Binimetinib treatment group are: 0μM (100±2.65) (not shown in the figure), 0.1μM (111±13.9), 0.3μM (105±17.2), 1μM (115±22.9) , 3μM (107±14.4), 10μM (92.4±2.44). This suggests that Binimetinib cannot inhibit the proliferation of HEL cells. Figure 2d reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Selumetinib treatment group are: 0 μM (100±1.62) (not shown in the figure), 0.1 μM (107±17.6), 0.3 μM (117±16.3), 1μM (110±26.3), 3μM (103±14.5), 10μM (88.8±2.69). This suggests that Selumetinib cannot inhibit the proliferation of HEL cells. Figure 2e reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Mirdametinib treatment group are: 0 μM (100±1.73) (not shown in the figure), 0.1 μM (107±17.6), 0.3 μM (117±16.3), 1μM (110±26.3), 3μM (103±14.5), 10μM (88.8±2.69). This suggests that Mirdametinib cannot inhibit the proliferation of HEL cells. Figure 2f reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Refametinib treatment group are: 0μM (100±2.63) (not shown in the figure), 0.1μM (96.0±7.44), 0.3μM (99.8±4.36), 1μM (93.4±4.31), 3μM (89.1±7.23), 10μM (84.6±8.89). This suggests that Refametinib cannot inhibit the proliferation of HEL cells. Figure 2g reflects that in HEL cells, the drug concentrations (average proliferation rate) of the TIC10 treatment group are: 0μM (100±4.31) (not shown in the figure), 0.1μM (113±15.0), 0.3μM (116±14.9), 1μM (80.3±3.43), 3μM (19.6±2.64), 10μM (11.1±7.15). This suggests that TIC10 can inhibit the proliferation of HEL cells. This effect increases as the drug concentration increases, and its inhibitory effect strengthens as the concentration increases. Figure 2h reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Ulixertinib treatment group are: 0 μM (100±2.58) (not shown in the figure), 0.1 μM (98.9±8.68), 0.3 μM (109±5.36), 1 μM (96.1±5.13), 3μM (100±4.61), 10μM (95.2±5.83). This suggests that Ulixertinib cannot inhibit the proliferation of HEL cells. Figure 2i reflects that in HEL cells, the drug concentrations (average proliferation rate) of the PD184352 treatment group are: 0μM (100±4.46) (not shown in the figure), 0.1μM (105±16.7), 0.3μM (118±21.2), 1μM (103±22.7), 3μM (99.0±20.3), 10μM (82.3 ±6.50). This suggests that PD184352 cannot inhibit the proliferation of HEL cells. Figure 2j reflects that in HEL cells, the drug concentrations (average proliferation rate) of the Pimasertib treatment group are: 0μM (100±3.07) (not shown in the figure), 0.1μM (89.0±1.42), 0.3μM (95.0±7.62), 1μM (86.9±1.71), 3μM (82.8±2.13), 10μM (77.3±2.52). This suggests that Pimasertib has a weak inhibitory effect on HEL cell proliferation, and its effect is weaker than ruxolitinib. The comparison of proliferation rates between the two groups in the figure uses t test (*p<0.05, **p<0.01, ***p<0.001).
实施例3、部分MEK/ERK信号通路抑制剂可促进骨髓增殖性肿瘤细胞的凋亡Example 3. Some MEK/ERK signaling pathway inhibitors can promote apoptosis of myeloproliferative tumor cells
一、材料与方法1. Materials and methods
1、细胞系与抑制剂同实施例1。1. Cell lines and inhibitors are the same as in Example 1.
2、细胞凋亡检测2. Cell apoptosis detection
为了检测抑制剂的促凋亡效应,分别使用芦可替尼及MEK/ERK信号通路抑制剂处理HEL原始细胞株24小时(浓度:0,0.1,0.3,1,3,10μM),补齐DMSO至等量。设3个平行重复组,使用AnnexinV和PI染色后流式细胞术检测细胞的凋亡情况。In order to detect the pro-apoptotic effect of the inhibitor, the HEL original cell line was treated with ruxolitinib and MEK/ERK signaling pathway inhibitors for 24 hours (concentration: 0, 0.1, 0.3, 1, 3, 10 μM), and supplemented with DMSO to equal amounts. Three parallel replicate groups were set up, and cell apoptosis was detected by flow cytometry after AnnexinV and PI staining.
细胞凋亡率计算公式:细胞凋亡率=早期凋亡细胞比率(Annexin V+/PI-)+晚期凋亡细胞及坏死细胞比率(AnnexinV+/PI+)。Apoptosis rate calculation formula: Apoptosis rate = early apoptotic cell ratio (Annexin V + /PI - ) + late apoptotic cell and necrotic cell ratio (AnnexinV + /PI + ).
二、结果分析2. Result analysis
图3中,Trematinib、Cobimetinib、TIC10、Ulixertinib、PD184352、Pimasertib对照的HEL细胞芦可替尼处理组药物浓度(平均凋亡率%±标准差)为:0μM(7.26±0.360)、0.1μM(7.39±0.0600)、0.3μM(7.40±0.820)、1μM(8.58±0.490)、3μM(7.12±0.180)、10μM(6.04±0.700);Binimetinib,Selumetinib,Mirdametinib,Refametinib对照的HEL细胞芦可替尼处理组药物浓度(平均凋亡率%±标准差)为:0μM(2.30±0.720)、0.1μM(2.30±0.210)、0.3μM(1.90±0.360)、1μM(2.4±0.50)、3μM(2.00±0.480)、10μM(2.30±0.720)。图3a反映在HEL细胞中,Trematinib处理组药物浓度(平均凋亡率)为:0μM(6.94±0.690)(图中未显示)、0.1μM(7.09±1.35)、0.3μM(6.32±0.780)、1μM(6.11±1.19)、3μM (6.83±1.83)、10μM(6.41±0.240)。这提示Trematinib不能促进HEL细胞的凋亡。图3b反映在HEL细胞中,Cobimetinib处理组药物浓度(平均凋亡率)为:0μM(5.09±0.060)(图中未显示)、0.1μM(5.35±0.500)、0.3μM(6.53±0.370)、1μM(6.31±0.670)、3μM(6.61±1.490)、10μM(5.64±0.470)。这提示Cobimetinib不能促进HEL细胞的凋亡。图3c中Binimetinib处理组药物浓度(凋亡率)为0μM(1.20±0.320)(图中未显示)、0.1μM(1.21±0.110)、0.3μM(1.55±0.310)、1μM(1.73±0.06)、3μM(1.39±0.29)、10μM(2.19±0.07),这提示Binimetinib不能促进骨髓增殖性肿瘤细胞的凋亡。图3d中,HEL细胞Selumetinib处理组药物浓度(平均凋亡率)为:0μM(1.91±0.51)(图中未显示)、0.1μM(2.24±0.0580)、0.3μM(2.14±0.748)、1μM(2.45±0.473)、3μM(1.91±0.225)、10μM(2.40±0.260),这提示Selumetinib不能促进骨髓增殖性肿瘤细胞的凋亡。图3e反映在HEL细胞中,Mirdametinib处理组药物浓度(平均凋亡率)为:0μM(1.46±0.146)(图中未显示)、0.1μM(1.99±0.367)、0.3μM(2.23±0.432)、1μM(2.45±0.473)、3μM(1.91±0.225)、10μM(2.40±0.260)。这提示Mirdametinib不能促进HEL细胞的凋亡。图3f反映在HEL细胞中,Refametinib处理组药物浓度(平均凋亡率)为:0μM(2.08±0.467)(图中未显示)、0.1μM(2.78±1.02)、0.3μM(2.21±0.369)、1μM(2.06±0.457)、3μM(2.39±0.713)、10μM(1.79±0.745)。这提示Refametinib不能促进HEL细胞的凋亡。图3g反映在HEL细胞中,TIC10处理组药物浓度(平均凋亡率)为:0μM(3.69±0.192)(图中未显示)、0.1μM(3.19±0.516)、0.3μM(5.63±2.50)、1μM(4.39±0.2880)、3μM(5.16±0.266)、10μM(9.43±0.445)。这提示TIC10能促进HEL细胞的凋亡,其效应随浓度的增加而增强。图3h反映在HEL细胞中,Ulixertinib处理组药物浓度(平均凋亡率)为:0μM(6.00±0.717)(图中未显示)、0.1μM(5.56±1.22)、0.3μM(6.22±1.86)、1μM(5.72±0.619)、3μM(5.99±1.73)、10μM(6.12±0.903)。这提示Ulixertinib不能促进HEL细胞的凋亡。图3i反映在HEL细胞中,PD184352处理组药物浓度(平均凋亡率)为:0μM(3.86±0.198)(图中未显示)、0.1μM(4.65± 0.959)、0.3μM(4.90±0.494)、1μM(4.72±0.161)、3μM(4.19±0.709)、10μM(4.77±0.902)。这提示PD184352不能促进HEL细胞的凋亡。图3j反映在HEL细胞中,Pimasertib处理组药物浓度(平均凋亡率)为:0μM(3.98±0.115)(图中未显示)、0.1μM(4.61±0.535)、0.3μM(5.74±1.68)、1μM(1.37±0.290)、3μM(6.37±0.272)、10μM(4.89±0.182)。这提示Pimasertib不能促进HEL细胞的凋亡。图中两组凋亡率的比较采用t检验(*p<0.05,**p<0.01,***p<0.001)。In Figure 3, the drug concentrations (average apoptosis rate % ± standard deviation) of Trematinib, Cobimetinib, TIC10, Ulixertinib, PD184352, and Pimasertib in the ruxolitinib-treated group of HEL cells in the control group are: 0 μM (7.26 ± 0.360), 0.1 μM (7.39 ±0.0600), 0.3μM (7.40±0.820), 1μM (8.58±0.490), 3μM (7.12±0.180), 10μM (6.04±0.700); Binimetinib, Selumetinib, Mirdametinib, Refametinib control HEL cell ruxolitinib treatment group The drug concentrations (average apoptosis rate %±standard deviation) are: 0μM (2.30±0.720), 0.1μM (2.30±0.210), 0.3μM (1.90±0.360), 1μM (2.4±0.50), 3μM (2.00±0.480) , 10μM (2.30±0.720). Figure 3a reflects that in HEL cells, the drug concentrations (average apoptosis rate) in the Trematinib treatment group are: 0 μM (6.94±0.690) (not shown in the figure), 0.1 μM (7.09±1.35), 0.3 μM (6.32±0.780), 1μM (6.11±1.19), 3μM (6.83±1.83), 10μM (6.41±0.240). This suggests that Trematinib cannot promote apoptosis of HEL cells. Figure 3b reflects that in HEL cells, the drug concentrations (average apoptosis rate) of the Cobimetinib treatment group are: 0 μM (5.09±0.060) (not shown in the figure), 0.1 μM (5.35±0.500), 0.3 μM (6.53±0.370), 1μM (6.31±0.670), 3μM (6.61±1.490), 10μM (5.64±0.470). This suggests that Cobimetinib cannot promote apoptosis of HEL cells. In Figure 3c, the drug concentrations (apoptosis rate) in the Binimetinib treatment group are 0 μM (1.20±0.320) (not shown in the figure), 0.1 μM (1.21±0.110), 0.3 μM (1.55±0.310), 1 μM (1.73±0.06), 3μM (1.39±0.29), 10μM (2.19±0.07), which suggests that Binimetinib cannot promote the apoptosis of myeloproliferative tumor cells. In Figure 3d, the drug concentrations (average apoptosis rate) in the Selumetinib-treated group of HEL cells are: 0 μM (1.91 ± 0.51) (not shown in the figure), 0.1 μM (2.24 ± 0.0580), 0.3 μM (2.14 ± 0.748), 1 μM ( 2.45±0.473), 3μM (1.91±0.225), 10μM (2.40±0.260), which suggests that Selumetinib cannot promote the apoptosis of myeloproliferative tumor cells. Figure 3e reflects that in HEL cells, the drug concentrations (average apoptosis rate) of Mirdametinib treatment group are: 0 μM (1.46±0.146) (not shown in the figure), 0.1 μM (1.99±0.367), 0.3 μM (2.23±0.432), 1μM (2.45±0.473), 3μM (1.91±0.225), 10μM (2.40±0.260). This suggests that Mirdametinib cannot promote apoptosis of HEL cells. Figure 3f reflects that in HEL cells, the drug concentrations (average apoptosis rate) of the Refametinib treatment group are: 0 μM (2.08±0.467) (not shown in the figure), 0.1 μM (2.78±1.02), 0.3 μM (2.21±0.369), 1μM (2.06±0.457), 3μM (2.39±0.713), 10μM (1.79±0.745). This suggests that Refametinib cannot promote apoptosis of HEL cells. Figure 3g reflects that in HEL cells, the drug concentrations (average apoptosis rate) of the TIC10 treatment group are: 0 μM (3.69±0.192) (not shown in the figure), 0.1 μM (3.19±0.516), 0.3 μM (5.63±2.50), 1μM (4.39±0.2880), 3μM (5.16±0.266), 10μM (9.43±0.445). This suggests that TIC10 can promote the apoptosis of HEL cells, and its effect increases with increasing concentration. As reflected in Figure 3h, in HEL cells, the drug concentrations (average apoptosis rate) of the Ulixertinib treatment group are: 0 μM (6.00±0.717) (not shown in the figure), 0.1 μM (5.56±1.22), 0.3 μM (6.22±1.86), 1μM (5.72±0.619), 3μM (5.99±1.73), 10μM (6.12±0.903). This suggests that Ulixertinib cannot promote apoptosis of HEL cells. Figure 3i reflects that in HEL cells, the drug concentration (average apoptosis rate) of the PD184352 treatment group was: 0 μM (3.86±0.198) (not shown in the figure), 0.1 μM (4.65± 0.959), 0.3μM (4.90±0.494), 1μM (4.72±0.161), 3μM (4.19±0.709), 10μM (4.77±0.902). This suggests that PD184352 cannot promote apoptosis of HEL cells. Figure 3j reflects that in HEL cells, the drug concentrations (average apoptosis rate) of the Pimasertib treatment group are: 0 μM (3.98±0.115) (not shown in the figure), 0.1 μM (4.61±0.535), 0.3 μM (5.74±1.68), 1μM (1.37±0.290), 3μM (6.37±0.272), 10μM (4.89±0.182). This suggests that Pimasertib cannot promote apoptosis of HEL cells. The comparison of apoptosis rates between the two groups in the figure uses t test (*p<0.05, **p<0.01, ***p<0.001).
实施例4、MEK/ERK信号通路抑制剂可抑制耐药骨髓增殖性肿瘤细胞的增殖。Example 4. MEK/ERK signaling pathway inhibitors can inhibit the proliferation of drug-resistant myeloproliferative tumor cells.
为了检测MEK/ERK信号通路抑制剂对骨髓增殖性肿瘤耐药细胞增殖能力的影响,方法为分别使用递增浓度的芦可替尼及MEK/ERK信号通路抑制剂处理HELRE,通过CellTiter-LumiTM发光法检测细胞的增殖。方法同实施例1,结果见图4。In order to detect the effect of MEK/ERK signaling pathway inhibitors on the proliferation of drug-resistant myeloproliferative tumor cells, the method was to use increasing concentrations of ruxolitinib and MEK/ERK signaling pathway inhibitors to treat HEL RE through CellTiter-Lumi TM Cell proliferation was detected by luminescence. The method is the same as in Example 1, and the results are shown in Figure 4.
图4反映了在HELRE细胞中,芦可替尼处理组药物浓度(平均增殖率%±标准差)为:0μM(100±3.79)(图中未显示)、0.1μM(105±2.08)、0.3μM(105±3.60)、1μM(104±9.46)、3μM(109±4.40)、10μM(96.0±3.69);图4a反映在HELRE细胞中,Trematinib处理组药物浓度(平均增殖率)为:0μM(100±2.27)(图中未显示)、0.001μM(99.38±2.75)、0.003μM(94.51±3.41)、0.01μM(67.59±6.26)、0.03μM(51.61±2.5)、0.1μM(40.45±4.5)。这提示Trematinib能有效抑制HELRE细胞的增殖。图4b反映在HELRE细胞中,Cobimetinib处理组药物浓度(平均增殖率)为:0μM(100±2.22)(图中未显示)、0.001μM(94.82±3.95)、0.003μM(101.27±4.23)、0.01μM(98.06±2.53)、0.03μM(83.06±2.48)、0.1μM(58.69±3.51)。这提示Cobimetinib能有效抑制HELRE细胞的增殖。图4c反映Binimetinib处理组药物浓度(平均增殖率)为:0μM(100±4.10)(图中未显示)、0.1μM(67.9±3.44)、0.3μM(53.8±1.41)、1μM(50.4±3.27)、3μM(43.1±2.18)、10μM(37.6±1.24)。这提示Binimetinib能抑制HELRE细胞的增殖,抑制率随药物浓度的增加而增强。图4d反映了在HELRE细 胞中,Selumetinib处理组药物浓度(平均增殖率)为:0μM(100±1.45)(图中未显示)、0.1μM(77.1±1.86)、0.3μM(62.0±3.90)、1μM(54.9±1.63)、3μM(48.2±2.63)、10μM(41.0±3.57)。这提示Selumetinib能抑制HELRE细胞的增殖,抑制率随药物浓度的增加而增强。图4e反映了在HELRE细胞中,Mirdametinib处理组药物浓度(平均增殖率)为:0μM(100±1.70)(图中未显示)、0.1μM(54.8±2.74)、0.3μM(57.9±4.50)、1μM(44.2±3.38)、3μM(39.6±0.380)、10μM(32.5±3.73)。这提示Mirdametinib能抑制HELRE细胞的增殖,抑制率随药物浓度的增加而增强。图4f反映在HELRE细胞中,Refametinib处理组药物浓度(平均增殖率)为:0μM(100±3.85)(图中未显示)、0.1μM(66.0±3.12)、0.3μM(61.2±2.05)、1μM(50.6±2.09)、3μM(43.3±1.23)、10μM(36.6±2.83)。这提示Refametinib能抑制HELRE细胞的增殖,抑制率随药物浓度的增加而增强。图4g反映在HELRE细胞中,TIC10处理组药物浓度(平均增殖率)为:0μM(100±6.41)(图中未显示)、0.1μM(101±7.19)、0.3μM(105±3.69)、1μM(62.5±5.82)、3μM(31.3±1.45)、10μM(19.6±1.01)。这提示TIC10能抑制HELRE细胞的增殖,该效应随药物浓度增加而增加。图4h反映在HELRE细胞中,Ulixertinib处理组药物浓度(平均增殖率)为:0μM(100±1.65)(图中未显示)、0.1μM(95.6±5.27)、0.3μM(91.4±3.49)、1μM(58.5±14.1)、3μM(48.4±7.37)、10μM(37.5±6.71)。这提示Ulixertinib能抑制HELRE细胞的增殖,该效应随药物浓度增加而增加。图4i反映在HELRE细胞中,PD184352处理组药物浓度(平均增殖率)为:0μM(100±4.73)(图中未显示)、0.1μM(81.8±1.27)、0.3μM(79.6±3.79)、1μM(52.2±1.24)、3μM(42.7±0.720)、10μM(33.9±0.700)。这提示PD184352能抑制HELRE细胞的增殖,该效应随药物浓度增加而增加。图4j反映在HELRE细胞中,Pimasertib处理组药物浓度(平均增殖率)为:0μM(100±3.71)(图中未显示)、0.1μM(59.3±1.78)、0.3μM(53.8±0.410)、1μM(42.2±3.46)、3μM(36.5±1.66)、10μM(30.3±2.22)。这提示Pimasertib能抑制HELRE细胞的增殖,该效应随药物浓度增加而增加。图中两组增殖率的比较采用t检验(*p<0.05,**p< 0.01,***p<0.001)。Figure 4 reflects that in HEL RE cells, the drug concentrations (average proliferation rate % ± standard deviation) of the ruxolitinib treatment group are: 0 μM (100 ± 3.79) (not shown in the figure), 0.1 μM (105 ± 2.08), 0.3μM (105±3.60), 1μM (104±9.46), 3μM (109±4.40), 10μM (96.0±3.69); Figure 4a reflects that in HEL RE cells, the drug concentration (average proliferation rate) of the Trematinib treatment group is: 0μM (100±2.27) (not shown in the figure), 0.001μM (99.38±2.75), 0.003μM (94.51±3.41), 0.01μM (67.59±6.26), 0.03μM (51.61±2.5), 0.1μM (40.45± 4.5). This suggests that Trematinib can effectively inhibit the proliferation of HEL RE cells. Figure 4b reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the Cobimetinib treatment group are: 0 μM (100±2.22) (not shown in the figure), 0.001 μM (94.82±3.95), 0.003 μM (101.27±4.23), 0.01μM (98.06±2.53), 0.03μM (83.06±2.48), 0.1μM (58.69±3.51). This suggests that Cobimetinib can effectively inhibit the proliferation of HEL RE cells. Figure 4c reflects that the drug concentrations (average proliferation rate) of the Binimetinib treatment group are: 0μM (100±4.10) (not shown in the figure), 0.1μM (67.9±3.44), 0.3μM (53.8±1.41), 1μM (50.4±3.27) , 3μM (43.1±2.18), 10μM (37.6±1.24). This suggests that Binimetinib can inhibit the proliferation of HEL RE cells, and the inhibition rate increases with increasing drug concentration. Figure 4d reflects the HEL RE fine In cells, the drug concentrations (average proliferation rate) of the Selumetinib-treated group were: 0 μM (100±1.45) (not shown in the figure), 0.1 μM (77.1±1.86), 0.3 μM (62.0±3.90), 1 μM (54.9±1.63) , 3μM (48.2±2.63), 10μM (41.0±3.57). This suggests that Selumetinib can inhibit the proliferation of HEL RE cells, and the inhibition rate increases with increasing drug concentration. Figure 4e reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the Mirdametinib treatment group are: 0 μM (100 ± 1.70) (not shown in the figure), 0.1 μM (54.8 ± 2.74), 0.3 μM (57.9 ± 4.50) , 1μM (44.2±3.38), 3μM (39.6±0.380), 10μM (32.5±3.73). This suggests that Mirdametinib can inhibit the proliferation of HEL RE cells, and the inhibition rate increases with increasing drug concentration. Figure 4f reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the Refametinib treatment group are: 0μM (100±3.85) (not shown in the figure), 0.1μM (66.0±3.12), 0.3μM (61.2±2.05), 1μM (50.6±2.09), 3μM (43.3±1.23), 10μM (36.6±2.83). This suggests that Refametinib can inhibit the proliferation of HEL RE cells, and the inhibition rate increases with the increase of drug concentration. Figure 4g reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the TIC10 treatment group are: 0μM (100±6.41) (not shown in the figure), 0.1μM (101±7.19), 0.3μM (105±3.69), 1μM (62.5±5.82), 3μM (31.3±1.45), 10μM (19.6±1.01). This suggests that TIC10 can inhibit the proliferation of HEL RE cells, and this effect increases with increasing drug concentration. Figure 4h reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of Ulixertinib treatment group are: 0 μM (100±1.65) (not shown in the figure), 0.1 μM (95.6±5.27), 0.3 μM (91.4±3.49), 1μM (58.5±14.1), 3μM (48.4±7.37), 10μM (37.5±6.71). This suggests that Ulixertinib can inhibit the proliferation of HEL RE cells, and this effect increases with increasing drug concentration. Figure 4i reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the PD184352 treatment group are: 0μM (100±4.73) (not shown in the figure), 0.1μM (81.8±1.27), 0.3μM (79.6±3.79), 1μM (52.2±1.24), 3μM (42.7±0.720), 10μM (33.9±0.700). This suggests that PD184352 can inhibit the proliferation of HEL RE cells, and this effect increases with increasing drug concentration. Figure 4j reflects that in HEL RE cells, the drug concentrations (average proliferation rate) of the Pimasertib treatment group are: 0μM (100±3.71) (not shown in the figure), 0.1μM (59.3±1.78), 0.3μM (53.8±0.410), 1μM (42.2±3.46), 3μM (36.5±1.66), 10μM (30.3±2.22). This suggests that Pimasertib can inhibit the proliferation of HEL RE cells, and this effect increases with increasing drug concentration. In the figure, the proliferation rate of the two groups was compared using t test (*p<0.05, **p< 0.01,***p<0.001).
实施例5、部分MEK/ERK信号通路抑制剂可促进耐药骨髓增殖性肿瘤细胞的凋亡Example 5. Some MEK/ERK signaling pathway inhibitors can promote the apoptosis of drug-resistant myeloproliferative tumor cells.
为了检测MEK/ERK信号通路抑制剂对骨髓增殖性肿瘤耐药细胞存活能力的影响,方法为分别使用递增浓度的芦可替尼及MEK/ERK信号通路抑制剂处理HELRE,使用AnnexinV和PI染色后流式检测细胞的凋亡情况。方法同实施例3,结果见图5。In order to detect the effect of MEK/ERK signaling pathway inhibitors on the survival of drug-resistant myeloproliferative tumor cells, the method was to treat HEL RE with increasing concentrations of ruxolitinib and MEK/ERK signaling pathway inhibitors, and stain with AnnexinV and PI. The apoptosis of cells was detected by flow cytometry. The method is the same as in Example 3, and the results are shown in Figure 5.
图5中,Trematinib、Cobimetinib对照的HELRE细胞芦可替尼处理组药物浓度(平均凋亡率%±标准差)为:0μM(8.36±0.636)、0.1μM(12.29±0.642)、0.3μM(13.14±1.241)、1μM(10.82±1.245)、3μM(9.74±0.255)、10μM(10.07±0.804);Binimetinib,Selumetinib,Mirdametinib,Refametinib对照的HELRE细胞芦可替尼处理组药物浓度(平均凋亡率%±标准差)为:0μM(0.300±0.0240)、0.1μM(0.290±0.0800)、0.3μM(0.330±0.0430)、1μM(0.290±0.0800)、3μM(0.280±0.0560)、10μM(0.360±0.0260)。TIC10、Ulixertinib、PD184352、Pimasertib对照的HELRE细胞芦可替尼处理组药物浓度(平均凋亡率%±标准差)为:0μM(2.15±1.01)、0.1μM(2.73±0.748)、0.3μM(2.11±0.989)、1μM(2.73±0.748)、3μM(2.09±0.982)、10μM(1.900±0.751);图5a反映在HELRE细胞中,Trematinib处理组药物浓度(平均凋亡率)为:0μM(7.13±1.46)(图中未显示)、0.1μM(9.42±2.48)、0.3μM(6.49±1.58)、1μM(9.56±1.39)、3μM(8.01±2.04)、10μM(7.72±0.167)。这提示Trematinib不能促进耐药骨髓增殖性肿瘤细胞凋亡。图5b反映在HELRE细胞中,Cobimetinib处理组药物浓度(平均凋亡率)为:0μM(4.87±1.37)(图中未显示)、0.1μM(3.53±1.10)、0.3μM(1.88±0.160)、1μM(4.09±1.46)、3μM(2.90±0.344)、10μM(2.36±0.414)。这提示Cobimetinib不能促进HELRE细胞的凋亡。图5c中Binimetinib处理组药物浓度(凋亡率)为0μM(0.250±0.0620)(图中未显示)、0.1μM(0.220±0.0300)、0.3μM(0.160±0.0410)、1μM(0.270±0.0360)、3μM(0.240±0.0180)、10μM(0.220 ±0.111),这提示Binimetinib不能促进耐药骨髓增殖性肿瘤细胞的凋亡。图5d中,HELRE细胞Selumetinib处理组药物浓度(平均凋亡率)为:0μM(0.290±0.110)(图中未显示)、0.1μM(0.230±0.0160)、0.3μM(0.270±0.0790)、1μM(0.220±0.0400)、3μM(0.310±0.0290)、10μM(0.320±0.00800),这提示Selumetinib不能促进耐药骨髓增殖性肿瘤细胞的凋亡。图5e反映在HELRE细胞中,Mirdametinib处理组药物浓度(平均凋亡率)为:0μM(0.270±0.0250)(图中未显示)、0.1μM(0.220±0.0180)、0.3μM(0.370±0.0640)、1μM(0.410±0.0340)、3μM(0.140±0.141)、10μM(0.330±0.0220)。这提示Mirdametinib不能促进耐药骨髓增殖性肿瘤细胞凋亡。图5f反映在HELRE细胞中,Refametinib处理组药物浓度(平均凋亡率)为:0μM(0.320±0.0670)(图中未显示)、0.1μM(0.320±0.0590)、0.3μM(0.330±0.0660)、1μM(0.330±0.0710)、3μM(0.560±0.0420)、10μM(0.800±0.0810)。这提示Refametinib能促进耐药骨髓增殖性肿瘤细胞凋亡,其效应随浓度的增加而增强。图5g反映在HELRE细胞中,TIC10处理组药物浓度(平均凋亡率)为:0μM(2.450±0.169)(图中未显示)、0.1μM(2.35±0.286)、0.3μM(1.89±0.253)、1μM(2.03±0.271)、3μM(2.29±0.210)、10μM(2.80±0.684)。这提示TIC10不能促进耐药骨髓增殖性肿瘤细胞凋亡。图5h反映在HELRE细胞中,Ulixertinib处理组药物浓度(平均凋亡率)为:0μM(2.62±0.210)(图中未显示)、0.1μM(2.28±0.450)、0.3μM(2.53±0.382)、1μM(2.39±0.399)、3μM(3.11±0.891)、10μM(3.29±0.284)。这提示Ulixertinib不能促进耐药骨髓增殖性肿瘤细胞凋亡。图5i反映在HELRE细胞中,PD184352处理组药物浓度(平均凋亡率)为:0μM(1.07±0.532)(图中未显示)、0.1μM(0.650±0.157)、0.3μM(0.990±0.153)、1μM(0.870±0.0560)、3μM(0.970±0.0600)、10μM(1.46±0.0250)。这提示PD184352不能促进耐药骨髓增殖性肿瘤细胞凋亡。图5j反映在HELRE细胞中,Pimasertib处理组药物浓度(平均凋亡率)为:0μM(1.55±0.372)(图中未显示)、0.1μM(1.45±0.528)、0.3μM(1.63±0.284)、1μM(1.43±0.189)、3μM(1.60±0.217)、10μM(1.77±0.167)。这提示Pimasertib不能促进耐药 骨髓增殖性肿瘤细胞凋亡。图中两组凋亡率的比较采用t检验(*p<0.05,**p<0.01,***p<0.001)。In Figure 5, the drug concentrations (average apoptosis rate % ± standard deviation) in the ruxolitinib-treated group of HEL RE cells compared with Trematinib and Cobimetinib are: 0 μM (8.36 ± 0.636), 0.1 μM (12.29 ± 0.642), 0.3 μM ( 13.14±1.241), 1μM (10.82±1.245), 3μM (9.74±0.255), 10μM (10.07±0.804); Binimetinib, Selumetinib, Mirdametinib, Refametinib control HEL RE cell ruxolitinib treatment group drug concentration (average apoptosis Rate% ± standard deviation) are: 0μM (0.300±0.0240), 0.1μM (0.290±0.0800), 0.3μM (0.330±0.0430), 1μM (0.290±0.0800), 3μM (0.280±0.0560), 10μM (0.360±0.0260) ). The drug concentrations (average apoptosis rate % ± standard deviation) of TIC10, Ulixertinib, PD184352, and Pimasertib control HEL RE cells in the ruxolitinib-treated group were: 0 μM (2.15 ± 1.01), 0.1 μM (2.73 ± 0.748), 0.3 μM ( 2.11±0.989), 1μM (2.73±0.748), 3μM (2.09±0.982), 10μM (1.900±0.751); Figure 5a reflects that in HEL RE cells, the drug concentration (average apoptosis rate) of the Trematinib treatment group is: 0μM ( 7.13±1.46) (not shown in the figure), 0.1μM (9.42±2.48), 0.3μM (6.49±1.58), 1μM (9.56±1.39), 3μM (8.01±2.04), 10μM (7.72±0.167). This suggests that Trematinib cannot promote apoptosis in drug-resistant myeloproliferative tumor cells. Figure 5b reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) in the Cobimetinib treatment group are: 0 μM (4.87±1.37) (not shown in the figure), 0.1 μM (3.53±1.10), 0.3 μM (1.88±0.160) , 1μM (4.09±1.46), 3μM (2.90±0.344), 10μM (2.36±0.414). This suggests that Cobimetinib cannot promote apoptosis of HEL RE cells. In Figure 5c, the drug concentrations (apoptosis rate) of the Binimetinib treatment group are 0 μM (0.250±0.0620) (not shown in the figure), 0.1 μM (0.220±0.0300), 0.3 μM (0.160±0.0410), 1 μM (0.270±0.0360), 3μM (0.240±0.0180), 10μM (0.220 ±0.111), which suggests that binimetinib cannot promote apoptosis of drug-resistant myeloproliferative tumor cells. In Figure 5d, the drug concentrations (average apoptosis rate) in the Selumetinib-treated group of HEL RE cells are: 0 μM (0.290±0.110) (not shown in the figure), 0.1 μM (0.230±0.0160), 0.3 μM (0.270±0.0790), 1 μM (0.220±0.0400), 3μM (0.310±0.0290), 10μM (0.320±0.00800), which suggests that Selumetinib cannot promote the apoptosis of drug-resistant myeloproliferative tumor cells. Figure 5e reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) of Mirdametinib treatment group are: 0 μM (0.270±0.0250) (not shown in the figure), 0.1 μM (0.220±0.0180), 0.3 μM (0.370±0.0640) , 1μM (0.410±0.0340), 3μM (0.140±0.141), 10μM (0.330±0.0220). This suggests that Mirdametinib cannot promote apoptosis in drug-resistant myeloproliferative tumor cells. Figure 5f reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) in the Refametinib treatment group are: 0μM (0.320±0.0670) (not shown in the figure), 0.1μM (0.320±0.0590), 0.3μM (0.330±0.0660) , 1μM (0.330±0.0710), 3μM (0.560±0.0420), 10μM (0.800±0.0810). This suggests that Refametinib can promote apoptosis of drug-resistant myeloproliferative tumor cells, and its effect increases with increasing concentration. Figure 5g reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) in the TIC10 treatment group are: 0 μM (2.450±0.169) (not shown in the figure), 0.1 μM (2.35±0.286), 0.3 μM (1.89±0.253) , 1μM (2.03±0.271), 3μM (2.29±0.210), 10μM (2.80±0.684). This suggests that TIC10 cannot promote apoptosis in drug-resistant myeloproliferative tumor cells. As reflected in Figure 5h, in HEL RE cells, the drug concentrations (average apoptosis rate) in the Ulixertinib treatment group are: 0 μM (2.62±0.210) (not shown in the figure), 0.1 μM (2.28±0.450), 0.3 μM (2.53±0.382) , 1μM (2.39±0.399), 3μM (3.11±0.891), 10μM (3.29±0.284). This suggests that Ulixertinib cannot promote apoptosis in drug-resistant myeloproliferative tumor cells. Figure 5i reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) of the PD184352 treatment group are: 0 μM (1.07±0.532) (not shown in the figure), 0.1 μM (0.650±0.157), 0.3 μM (0.990±0.153) , 1μM (0.870±0.0560), 3μM (0.970±0.0600), 10μM (1.46±0.0250). This suggests that PD184352 cannot promote apoptosis of drug-resistant myeloproliferative tumor cells. Figure 5j reflects that in HEL RE cells, the drug concentrations (average apoptosis rate) in the Pimasertib treatment group are: 0 μM (1.55±0.372) (not shown in the figure), 0.1 μM (1.45±0.528), 0.3 μM (1.63±0.284) , 1μM (1.43±0.189), 3μM (1.60±0.217), 10μM (1.77±0.167). This suggests that Pimasertib cannot promote drug resistance Apoptosis of myeloproliferative neoplasms. The comparison of apoptosis rates between the two groups in the figure uses t test (*p<0.05, **p<0.01, ***p<0.001).
以上仅是本发明的优选实施方式,应当指出,对于本技术领域的普通技术人员来说,在不脱离本发明原理的前提下,还可以做出若干改进和润饰,这些改进和润饰也应视为本发明的保护范围。 The above are only the preferred embodiments of the present invention. It should be pointed out that for those of ordinary skill in the art, several improvements and modifications can be made without departing from the principles of the present invention. These improvements and modifications should also be regarded as It is the protection scope of the present invention.

Claims (10)

  1. MEK/ERK信号通路抑制剂在制备治疗骨髓增殖性肿瘤的药物中的应用。Application of MEK/ERK signaling pathway inhibitors in the preparation of drugs for the treatment of myeloproliferative tumors.
  2. 根据权利要求1所述的应用,其特征在于,所述骨髓增殖性肿瘤为对化疗药物具有耐药性的骨髓增殖性肿瘤。The application according to claim 1, wherein the myeloproliferative tumor is a myeloproliferative tumor that is resistant to chemotherapy drugs.
  3. 根据权利要求2所述的应用,其特征在于,所述化疗药物为治疗骨髓增殖性肿瘤的化疗药物,所述化疗药物包括芦可替尼和/或菲卓替尼。The application according to claim 2, wherein the chemotherapeutic drug is a chemotherapeutic drug for treating myeloproliferative tumors, and the chemotherapeutic drug includes ruxolitinib and/or fizotinib.
  4. 根据权利要求1所述的应用,其特征在于,所述骨髓增殖性肿瘤为真性红细胞增多症、原发性血小板增多症或骨髓纤维化,所述骨髓纤维化为原发性骨髓纤维化、继发于真性红细胞增多症的骨髓纤维化和继发于原发性血小板增多症的骨髓纤维化中的至少一种。The application according to claim 1, wherein the myeloproliferative tumor is polycythemia vera, essential thrombocythemia or myelofibrosis, and the myelofibrosis is primary myelofibrosis, secondary myelofibrosis. At least one of myelofibrosis arising from polycythemia vera and myelofibrosis secondary to essential thrombocythemia.
  5. 根据权利要求1所述的应用,其特征在于,MEK/ERK信号通路抑制剂为Trematinib、Cobimetinib、Binimetinib、Selumetinib、Mirdametinib、Refametinib、TIC10、Ulixertinib、PD184352、Pimasertib中的至少一种。The application according to claim 1, wherein the MEK/ERK signaling pathway inhibitor is at least one of Trematinib, Cobimetinib, Binimetinib, Selumetinib, Mirdametinib, Refametinib, TIC10, Ulixertinib, PD184352, and Pimasertib.
  6. 根据权利要求1所述的应用,其特征在于,所述治疗包括抑制肿瘤细胞的增殖和/或促进肿瘤细胞的凋亡。The application according to claim 1, wherein the treatment includes inhibiting the proliferation of tumor cells and/or promoting the apoptosis of tumor cells.
  7. MEK/ERK信号通路抑制剂在制备抑制HEL细胞的增殖和/或促进HEL细胞凋亡的药物中的应用。The application of MEK/ERK signaling pathway inhibitors in the preparation of drugs that inhibit the proliferation of HEL cells and/or promote the apoptosis of HEL cells.
  8. 根据权利要求7所述的应用,其特征在于,所述HEL细胞包括非耐药性HEL细胞和/或耐药性的HEL细胞。The application according to claim 7, wherein the HEL cells include non-drug-resistant HEL cells and/or drug-resistant HEL cells.
  9. 根据权利要求1至8中任一项所述的应用,其特征在于,所述药物还包含用于治疗骨髓增殖性肿瘤的其他药物成分以及药学上可接受的辅料。The application according to any one of claims 1 to 8, characterized in that the medicine also contains other pharmaceutical ingredients for treating myeloproliferative tumors and pharmaceutically acceptable excipients.
  10. 根据权利要求9所述的应用,其特征在于,所述药物的剂型为口服制剂或注射制剂。 The application according to claim 9, characterized in that the dosage form of the drug is an oral preparation or an injection preparation.
PCT/CN2023/116402 2022-07-19 2023-09-01 Use of mek/erk signal pathway inhibitor in preparation of drug for treating myeloproliferative neoplasms WO2024017406A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210846544.4A CN115054695A (en) 2022-07-19 2022-07-19 Application of MEK/ERK signal pathway inhibitor in preparation of medicine for treating myeloproliferative tumors
CN202210846544.4 2022-07-19

Publications (1)

Publication Number Publication Date
WO2024017406A1 true WO2024017406A1 (en) 2024-01-25

Family

ID=83205933

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/116402 WO2024017406A1 (en) 2022-07-19 2023-09-01 Use of mek/erk signal pathway inhibitor in preparation of drug for treating myeloproliferative neoplasms

Country Status (2)

Country Link
CN (1) CN115054695A (en)
WO (1) WO2024017406A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015066432A1 (en) * 2013-11-01 2015-05-07 Foundation Medicine, Inc. Method of treating post-myeloproliferative neoplasms (mpns) and post-mpn acute myeloid leukemia
US20160243150A1 (en) * 2013-10-03 2016-08-25 Duke University Compositions and Methods for Treating Cancer with JAK2 Activity
CN112843055A (en) * 2021-01-15 2021-05-28 徐州医科大学 Pharmaceutical composition and application thereof in preparation of medicines for treating myeloproliferative diseases of targeted calreticulin mutation type

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111870600B (en) * 2020-07-13 2022-04-15 中南大学湘雅二医院 New application of sorafenib, regorafenib and analogues or derivatives thereof
WO2022074600A1 (en) * 2020-10-08 2022-04-14 Novartis Ag Use of an erk inhibitor for the treatment of myelofibrosis

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160243150A1 (en) * 2013-10-03 2016-08-25 Duke University Compositions and Methods for Treating Cancer with JAK2 Activity
WO2015066432A1 (en) * 2013-11-01 2015-05-07 Foundation Medicine, Inc. Method of treating post-myeloproliferative neoplasms (mpns) and post-mpn acute myeloid leukemia
CN112843055A (en) * 2021-01-15 2021-05-28 徐州医科大学 Pharmaceutical composition and application thereof in preparation of medicines for treating myeloproliferative diseases of targeted calreticulin mutation type

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
STIVALA, S. ET AL.: "Targeting compensatory MEK/ERK activation increases JAK inhibitor efficacy in myeloproliferative neoplasms", THE JOURNAL OF CLINICAL INVESTIGATION, vol. 129, no. 4, 1 April 2019 (2019-04-01), pages 1596 - 1611, XP055869761, ISSN: 0021-9738, DOI: 10.1172/JCI98785 *

Also Published As

Publication number Publication date
CN115054695A (en) 2022-09-16

Similar Documents

Publication Publication Date Title
WO2022228323A1 (en) Application of ly3009120 in preparation of drugs for treatment of myeloproliferative neoplasms
US10888559B2 (en) Quinoline derivatives for non-small cell lung cancer
AU2017354903B2 (en) Treating gastric cancer using combination therapies comprising liposomal irinotecan, oxaliplatin, 5-fluoruracil (and leucovorin)
US20230310394A1 (en) Sorafenib, regorafenib and novel use of analogue or derivative thereof
AU2016279096B2 (en) Composition containing carboplatin and use
WO2024017403A1 (en) Use of plurality of compounds in preparation of drug for treating myeloproliferative neoplasms
TW201141473A (en) Combination therapy for small cell lung cancer
WO2020192506A1 (en) Chiauranib for treatment of small cell lung cancer
WO2024017406A1 (en) Use of mek/erk signal pathway inhibitor in preparation of drug for treating myeloproliferative neoplasms
CN108778336A (en) A kind of VEGFR inhibitor combines the purposes in the drug for preparing treatment gastric cancer with PARP inhibitor
US11890292B2 (en) Compositions, methods, systems and/or kits for preventing and/or treating neoplasms
US20210369709A1 (en) EGFR TKIs FOR USE IN THE TREATMENT OF NON-SMALL CELL LUNG CANCER
TW202342047A (en) Combination therapy using substituted pyrimidin-4(3h)-ones and sotorasib
US20230226030A1 (en) Therapeutic combinations comprising a raf inhibitor for use in treating braf mutant nsclc
US9867831B2 (en) Combination treatment of acute myeloid leukemia and myelodysplastic syndrome
TW201244732A (en) Ezatiostat for treating multiple myeloma
TW202038964A (en) Combination therapy
TWI843217B (en) A drug combination and its application
Ashish et al. Anaplastic Lymphoma Kinase (ALK) Mutation-Targeting Treatment With Alectinib in Lung Adenocarcinoma and Primary Cutaneous Marginal Zone B-Cell Lymphoma
WO2023280244A1 (en) Uses of chiauranib and combined administration thereof for treating breast cancer
CN113274394B (en) Pharmaceutical composition for treating tyrosine kinase inhibitor drug-resistant non-small cell lung cancer
WO2022199656A1 (en) Pharmaceutical combination, kit containing same, and use thereof
JP2021063014A (en) Leukemia therapeutic agent
US9956225B2 (en) Treatment of myelodysplastic syndrome
WO2021185234A1 (en) Combined pharmaceutical composition of compound as c-met kinase inhibitor and use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23842468

Country of ref document: EP

Kind code of ref document: A1