[go: up one dir, main page]

WO2024017406A1 - Utilisation d'un inhibiteur de la voie de signalisation mek/erk dans la préparation d'un médicament pour le traitement de néoplasmes myéloprolifératifs - Google Patents

Utilisation d'un inhibiteur de la voie de signalisation mek/erk dans la préparation d'un médicament pour le traitement de néoplasmes myéloprolifératifs 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
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/CN2023/116402
Other languages
English (en)
Chinese (zh)
Inventor
邓沱
胡婉钰
吾甫尔艾尼
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Second Xiangya Hospital of Central South University
Original Assignee
Second Xiangya Hospital of Central South University
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 Second Xiangya Hospital of Central South University filed Critical Second Xiangya Hospital of Central South University
Publication of WO2024017406A1 publication Critical patent/WO2024017406A1/fr
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • 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

L'invention concerne le domaine technique des médicaments, en particulier une utilisation d'un inhibiteur de la voie de signalisation MEK/ERK dans la préparation d'un médicament pour le traitement de néoplasmes myéloprolifératifs. L'inhibiteur de la voie de signalisation MEK/ERK est utilisé pour traiter des néoplasmes myéloprolifératifs, en offrant une nouvelle approche de traitement pour des patients atteints de néoplasmes myéloprolifératifs, et plus de choix pour les cliniciens et les patients. Pour des patients atteints de néoplasmes myéloprolifératifs qui sont résistants au ruxolitinib, l'inhibiteur de la voie de signalisation MEK/ERK peut fournir un traitement médicamenteux oral continu aux patients, sans avoir besoin d'une transplantation de moelle osseuse. L'inhibiteur de la voie de signalisation MEK/ERK peut être synthétisé chimiquement, et le coût est inférieur à celui d'une préparation biologique. De plus, les inhibiteurs dans la présente invention ont tous passé les essais cliniques de phase I, et montrent des effets secondaires légers et une bonne tolérance des patients cliniques.
PCT/CN2023/116402 2022-07-19 2023-09-01 Utilisation d'un inhibiteur de la voie de signalisation mek/erk dans la préparation d'un médicament pour le traitement de néoplasmes myéloprolifératifs Ceased WO2024017406A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210846544.4A CN115054695B (zh) 2022-07-19 2022-07-19 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
CN202210846544.4 2022-07-19

Publications (1)

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

Family

ID=83205933

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/116402 Ceased WO2024017406A1 (fr) 2022-07-19 2023-09-01 Utilisation d'un inhibiteur de la voie de signalisation mek/erk dans la préparation d'un médicament pour le traitement de néoplasmes myéloprolifératifs

Country Status (2)

Country Link
CN (1) CN115054695B (fr)
WO (1) WO2024017406A1 (fr)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015066432A1 (fr) * 2013-11-01 2015-05-07 Foundation Medicine, Inc. Méthode de traitement de post-néoplasies myéloprolifératives (nmp) et de la leucémie aiguë myéloïde post-nmp
US20160243150A1 (en) * 2013-10-03 2016-08-25 Duke University Compositions and Methods for Treating Cancer with JAK2 Activity
CN112843055A (zh) * 2021-01-15 2021-05-28 徐州医科大学 一种药物组合物及其在制备治疗靶向钙网蛋白突变类型的骨髓增殖性疾病药物中的应用

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015095831A1 (fr) * 2013-12-20 2015-06-25 Biomed Valley Discoveries, Inc. Traitements du cancer faisant appel à des associations d'inhibiteurs de l'erk et de la mtor
CN111870600B (zh) * 2020-07-13 2022-04-15 中南大学湘雅二医院 索拉非尼、瑞戈非尼及其类似物或衍生物的新应用
EP4225317A1 (fr) * 2020-10-08 2023-08-16 Novartis AG Utilisation d'un inhibiteur d'erk pour le traitement de la myélofibrose

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 (fr) * 2013-11-01 2015-05-07 Foundation Medicine, Inc. Méthode de traitement de post-néoplasies myéloprolifératives (nmp) et de la leucémie aiguë myéloïde post-nmp
CN112843055A (zh) * 2021-01-15 2021-05-28 徐州医科大学 一种药物组合物及其在制备治疗靶向钙网蛋白突变类型的骨髓增殖性疾病药物中的应用

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 (zh) 2022-09-16
CN115054695B (zh) 2024-06-25

Similar Documents

Publication Publication Date Title
WO2022228323A1 (fr) Application de ly3009120 dans la préparation de médicaments pour le traitement de néoplasmes myéloprolifératifs
KR20190077441A (ko) 리포솜성 이리노테칸, 옥살리플라틴, 5-플루오로우라실 (및 류코보린)을 포함하는 조합 요법을 사용하는 위암 치료
US20250325543A1 (en) EGFR TKIs FOR USE IN THE TREATMENT OF NON-SMALL LUNG CANCER
TW202038964A (zh) 組合療法
US20230226030A1 (en) Therapeutic combinations comprising a raf inhibitor for use in treating braf mutant nsclc
US20180169055A1 (en) Composition containing carboplatin and use
JP7640033B2 (ja) ピリド[1,2-a]ピリミジノン類似体の使用
US20230310394A1 (en) Sorafenib, regorafenib and novel use of analogue or derivative thereof
CN119384277A (zh) 使用经取代的嘧啶-4(3h)-酮和索托拉西布的组合疗法
TW201141473A (en) Combination therapy for small cell lung cancer
JP2021063014A (ja) 白血病治療薬
WO2024017403A1 (fr) Utilisation d'une pluralité de composés dans la préparation d'un médicament pour le traitement de néoplasmes myéloprolifératifs
CN110522753A (zh) 一种新型抗肿瘤药物组合物、制剂和应用
CN115054695B (zh) 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
CN111728974B (zh) 西奥罗尼用于小细胞肺癌的治疗
CN109568313B (zh) 一种抗肿瘤联合用药物及其在制备抗癌药物中的应用
CN115135326B (zh) 作为c-Met激酶抑制剂的化合物的联用药物组合物及其用途
JP6708634B2 (ja) 急性骨髄性白血病及び骨髄異形成症候群の併用療法iii
TW201244732A (en) Ezatiostat for treating multiple myeloma
TWI843217B (zh) 一種藥物組合及其應用
Wu et al. Inhibition of hepatocellular carcinoma progression by artesunate via modulation of the TLR4/MyD88/NF-κB signaling pathway
WO2024255883A1 (fr) Utilisation d'un inhibiteur d'akt dans la préparation d'un médicament pour la prévention ou le traitement du cancer du sein
WO2023280244A1 (fr) Utilisations du chiauranib et administration combinée de celui-ci pour le traitement du cancer du sein
TW202342044A (zh) Cdk4及6抑制劑及氟維司群用於治療先前以cdk4及6抑制劑治療的患者之荷爾蒙受體陽性、人類表皮生長因子受體2陰性之晚期或轉移性乳癌
WO2024230727A1 (fr) Composition pharmaceutique combinée de composé pyrrolo[2,3-c]pyridone et son utilisation

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

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 23842468

Country of ref document: EP

Kind code of ref document: A1