WO2019039390A1 - Agent thérapeutique contre la leucémie myélomonocytaire juvénile - Google Patents

Agent thérapeutique contre la leucémie myélomonocytaire juvénile Download PDF

Info

Publication number
WO2019039390A1
WO2019039390A1 PCT/JP2018/030471 JP2018030471W WO2019039390A1 WO 2019039390 A1 WO2019039390 A1 WO 2019039390A1 JP 2018030471 W JP2018030471 W JP 2018030471W WO 2019039390 A1 WO2019039390 A1 WO 2019039390A1
Authority
WO
WIPO (PCT)
Prior art keywords
alk
tyrosine kinase
myelomonocytic leukemia
juvenile myelomonocytic
fusion gene
Prior art date
Application number
PCT/JP2018/030471
Other languages
English (en)
Japanese (ja)
Inventor
秀城 村松
友介 奥野
典寛 村上
Original Assignee
国立大学法人名古屋大学
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 国立大学法人名古屋大学 filed Critical 国立大学法人名古屋大学
Priority to JP2019537597A priority Critical patent/JPWO2019039390A1/ja
Publication of WO2019039390A1 publication Critical patent/WO2019039390A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • 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
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/15Medicinal preparations ; Physical properties thereof, e.g. dissolubility
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids

Definitions

  • the present invention relates to means useful for the treatment, diagnosis and the like of juvenile myelomonocytic leukemia (JMML). More specifically, it relates to a JMML therapeutic agent, a novel fusion gene involved in JMML, and its use.
  • JMML juvenile myelomonocytic leukemia
  • Juvenile myelomonocytic leukemia is a rare myelodysplasia of the infancy and childhood characterized by proliferation of myelomonocytic cells and hypersensitivity to granulocyte-macrophage colony stimulating factor (GM-CSF). / Myeloproliferative disease (MDS / MPD) (Non-patent Document 1). More than 80% of JMML cases are characterized as having mutually exclusive somatic / germline mutations in classical RAS pathway related genes such as PTPN11, NF1, NRAS, KRAS, CBL (non-patent literature) 2, 3).
  • Second hit mutations of SETBP 1 (Non-patent documents 4 and 5) and other genes (Non-patent documents 6 and 7), gene expression profiles (Non-patent documents 8 and 9), and methylation profiles of promoter regions of several genes (Non-patent documents 10 and 11) are identified as prognostic factors.
  • SETBP 1 Non-patent documents 4 and 5
  • other genes Non-patent documents 6 and 7
  • gene expression profiles Non-patent documents 8 and 9
  • methylation profiles of promoter regions of several genes (Non-patent documents 10 and 11) are identified as prognostic factors.
  • the interrelationship between these biomarkers has not yet been clarified.
  • Helsmoortel, H. H. et al. LIN28B overexpressions define a novel fetal-like subgroup of juvenile myelomonocytic leukemia. Blood 127, 1163-72 (2016). Bresolin, S. et al. Gene expression-based classification as an independent predictor of clinical outcome in juvenile myelomonocytic leukemia. J Clin Oncol 28, 1919-27 (2010). Olk-Batz, C. et al. Aberrant DNA methylation characterization juvenile myelomonocytic leukemia with poor outcome. Blood 117, 4871-80 (2011). Sakaguchi, H. et al. Aberrant DNA Methylation Is Associated with a Poor Outcome in Juvenile Myelomonocytic Leukemia. PLoS One 10, e0145394 (2015).
  • JMML cases are generally resistant to conventional chemotherapy, and allogeneic hematopoietic stem cell transplantation is the only definitive treatment, but overall survival at 5 years is poor at around 50%, and new molecules Targeted treatment is required.
  • JMML is known to have somatic or germline gene mutations (formation of fusion genes) of RAS pathway related genes such as PTPN11, NF1, NRAS, KRAS, CBL in 70% or more cases of JMML.
  • the three types of fusion genes identified are those with mutations in receptor tyrosine kinase genes and are extremely characteristic.
  • the three types of fusion genes are new therapeutic targets for JMML, and their utility or significance is great.
  • the existence of a fusion gene accompanied by a mutation in receptor tyrosine kinase gene can be evaluated as an extremely important finding in developing a future therapeutic strategy for JMML.
  • crizotinib which is an ALK tyrosine kinase inhibitor and is used for the treatment of non-small cell lung cancer, suppressed the growth of newly identified fusion gene positive cells in vitro. Moreover, crizotinib also improved the patient prognosis clinically. Intriguingly, crizotinib also showed antiproliferative effects in fusion gene negative JMML as well. As a result of further examination, tumor growth inhibitory effect was also recognized by ALK tyrosine kinase inhibitors other than crizotinib. These experimental results show that an agent called ALK tyrosine kinase inhibitor, which is a line with a known therapeutic agent for JMML, can be an effective molecular-targeted therapeutic agent for JMML.
  • JAK2 tyrosine kinase inhibitors can also be molecular targeted therapeutic agents for JMML, and have important implications for therapeutic strategies.
  • a therapeutic agent for juvenile myelomonocytic leukemia which comprises an ALK tyrosine kinase inhibitor or a JAK2 tyrosine kinase inhibitor.
  • ALK tyrosine kinase inhibitor is a tyrosine kinase inhibitor for ALK and ROS1.
  • a test method for juvenile myelomonocytic leukemia which comprises the following steps (1) to (3): (1) preparing a sample containing leukemia cells isolated from a patient with juvenile myelomonocytic leukemia; (2) detecting the presence or absence of a fusion gene of ALK or ROS1 or a fusion protein encoded by the fusion gene in the sample; (3) determining that the fusion gene or the fusion protein is detected to be compatible with treatment with an ALK tyrosine kinase inhibitor. [9] A fusion gene of ALK or ROS1 as a cause of juvenile myelomonocytic leukemia.
  • a method of screening a substance effective for the treatment of juvenile myelomonocytic leukemia which comprises the following steps (i) to (iii): (i) preparing a cell that expresses a fusion gene of ALK or ROS1; (ii) culturing the cells in the presence of a test substance; (iii) measuring the number of viable cells or the number of colonies to determine the efficacy of the test substance.
  • a method for treating juvenile myelomonocytic leukemia comprising administering a therapeutically effective amount of an ALK tyrosine kinase inhibitor or a JAK2 tyrosine kinase inhibitor to a patient with juvenile myelomonocytic leukemia.
  • JMML juvenile myelomonocytic leukemia
  • NS / MPD Noonan syndrome-related myeloproliferative disorder
  • LOH loss of heterozygosity.
  • A-c Structure of the detected fusion protein.
  • TBL1XR1-ROS1 identified by UPN106.
  • MAM MAM domain
  • LDL-R low density lipoprotein receptor class A domain
  • TM transmembrane domain
  • TPR tetratricopeptide repeat domain
  • RanBD1 Ran binding domain
  • LisH lis homologous domain
  • F-box like F-box like domain
  • FN Type III Fibronectin type III domain.
  • CD34 + cells (1 ⁇ 10 3 per culture) from each mutation positive patient, 0 , 200, 500 nM crizotinib was added to the medium of cytokine (+) or ( ⁇ ) and cultured for 2 weeks.
  • CFU-GM Granulocyte macrophage colony forming unit. *, p ⁇ 0.05; ,, p ⁇ 0.01.
  • Correlation between established risk factors in JMML Co-occurrence and exclusivity between clinical features, genetic changes, expression and methylation profiles, and outcomes. Gray lines indicate each factor associated with JMML, and circles indicate that the two crossed factors occur statistically simultaneously or exclusively.
  • the first aspect of the present invention relates to a therapeutic agent for juvenile myelomonocytic leukemia (JMML) and its use.
  • “Therapeutic agent” refers to a drug that exhibits a therapeutic or prophylactic effect on a target disease or condition (ie, JMML).
  • Therapeutic effects include alleviation of symptoms or concomitant symptoms characteristic of the target disease / pathology (reduction of symptoms), prevention or delay of deterioration of symptoms, and the like. The latter can be regarded as one of the preventive effects in terms of preventing the aggravation.
  • a typical preventive effect is to prevent or delay the recurrence of symptoms characteristic of the target disease / pathology. As long as it exhibits some therapeutic effect or preventive effect or both for the target disease / pathology, it is a therapeutic agent for the target disease / pathology.
  • ALK Advanced Lymphoma Kinase
  • ALK tyrosine kinase inhibitors have shown efficacy against JMML in vitro and in vivo.
  • ALK Anaplastic Lymphoma Kinase
  • An ALK fusion gene (EML4-ALK, TFG-ALK, KIF5B-ALK, etc.) in which the ALK gene and other genes are fused is found in some cases of non-small cell lung cancer, etc.
  • ALK tyrosine kinase is constitutively aberrantly activated to cause canceration.
  • Several therapeutic agents (ALK tyrosine kinase inhibitors) that target the ALK fusion gene have been developed and are clinically applied (for example, application of crizotinib to treatment of non-small cell lung cancer).
  • ALK tyrosine kinase inhibitors include crizotinib (Crizotinib), alectinib (Alectinib), ceritinib (Ceritinib), and TAE684. It is preferable based on the fact that a ROS1 (c-ros on cogene 1) fusion gene (TBL1XR1-ROS1) positive was found in cases of JMML and the fact that crizotinib, which is an inhibitor for ALK and ROS1, showed efficacy for JMML. Uses an ALK tyrosine kinase inhibitor that also exhibits an inhibitory effect on ROS1.
  • tyrosine kinase inhibitors against ALK and ROS1 are employed as ALK tyrosine kinase inhibitors.
  • the “tyrosine kinase inhibitors against ALK and ROS1” show at least an inhibitory activity against ALK and ROS1, and the presence or absence of the inhibitory activity against other tyrosine kinases is not particularly limited.
  • Examples of ALK tyrosine kinase inhibitors falling under “tyrosine kinase inhibitors against ALK and ROS1” include crizotinib, seritinib and lolatinib.
  • ROS1 is a type of receptor tyrosine kinase and, like ALK, belongs to the insulin receptor family.
  • ROS1 fusion genes CD74-ROS1, SLC34A2-ROS1, etc.
  • the tyrosine kinase domain of ROS1 shows high homology to the tyrosine kinase domain of ALK.
  • crizotinib which is an ALK tyrosine kinase inhibitor, exhibited a growth inhibitory action, ie, a drug effect not only on novel fusion gene-positive JMML cells, but also on novel fusion gene-negative JMML cells.
  • the subject (JMML patient) of the therapeutic agent of the present invention is not particularly limited, and the therapeutic agent of the present invention can be widely used for the treatment of JMML.
  • ALK fusion gene for example, DCTN1-ALK fusion gene, RANBP2-ALK fusion gene
  • ROS1 fusion gene for example, TBL1XR1-ROS1 fusion gene
  • a JAK2 tyrosine kinase inhibitor is used based on the fact that the JAK2 tyrosine kinase inhibitor, Luxolitinib (Ruxolitinib) has shown efficacy against JMML in vitro.
  • Luxoritinib selectively acts on JAK1 and JAK2 subtypes.
  • JAK2 tyrosine kinase inhibitors are luxoritinib, pacritinib (Pacritinib), varisitinib (Baricitinib).
  • pharmacologically acceptable salts of ALK tyrosine kinase inhibitors or JAK2 tyrosine kinase inhibitors may be used.
  • “Pharmaceutically acceptable salt” is not particularly limited, and use of various salts, for example, acid addition salts, metal salts, ammonium salts, organic amine addition salts, amino acid addition salts and the like is conceivable. .
  • Examples of acid addition salts include mineral acid salts such as hydrochlorides, sulfates, nitrates, phosphates, hydrobromides, acetates, maleates, fumarates, citrates, benzenesulfonates, Organic acid salts such as benzoate, malate, oxalate, methanesulfonate, tartrate and the like can be mentioned.
  • Examples of metal salts include alkali metal salts such as sodium salts, potassium salts and lithium salts, alkaline earth metal salts such as magnesium salts and calcium salts, aluminum salts and zinc salts.
  • Examples of ammonium salts include salts such as ammonium and tetramethyl ammonium.
  • organic amine addition salts include morpholine addition salts and piperidine addition salts.
  • amino acid addition salts include glycine addition salts, phenylalanine addition salts, lysine addition salts, aspartic acid addition salts and glutamic acid addition salts.
  • Two or more active ingredients may be used in combination.
  • other active ingredients are mixed. May be
  • any component that can usually be used in medicine may be appropriately blended with the therapeutic agent of the present invention, as long as the effects and pharmaceutical stability of the present invention are not impaired.
  • Ingredients which can be compounded are not particularly limited, and include, for example, carrier components or additives, and as carrier components or additives in a solid preparation, for example, excipients, disintegrants, binders, lubricants, antioxidants , Coating agents, coloring agents, flavoring agents, surfactants, plasticizers, sweeteners, flavoring agents, disintegrating aids, foaming agents, adsorbents, preservatives, preservatives, wetting agents, antistatic agents, etc. .
  • a carrier component or additive in the liquid preparation for example, a solvent, a pH adjuster, a refreshing agent, a suspending agent, an antifoamer, a thickener, a solubilizer, a surfactant, an antioxidant, a coloring agent Agents, sweeteners, flavoring agents, preservatives / antimicrobial agents, chelating agents, solubilizers or solubilizers, stabilizers, fluidizers, emulsifiers, thickeners, buffers, isotonic agents, dispersions Agents and the like.
  • a solvent for example, a solvent, a pH adjuster, a refreshing agent, a suspending agent, an antifoamer, a thickener, a solubilizer, a surfactant, an antioxidant, a coloring agent Agents, sweeteners, flavoring agents, preservatives / antimicrobial agents, chelating agents, solubilizers or solubilizers, stabilizers, fluidizers, e
  • the dosage form for formulation is also not particularly limited.
  • dosage forms include tablets (including uncoated tablets, sugar-coated tablets, intraoral fast disintegrating tablets, intraoral fast dissolving tablets, chewable tablets, effervescent tablets, troches, drops, film-coated tablets, etc.), pills, granules, Fine granules, powders, hard capsules, soft capsules, syrups, injections, external preparations and suppositories.
  • it may be any of a solid preparation, a semi-solid preparation, and a liquid preparation (for example, a decoction or a dip).
  • dosage form is a tablet.
  • dosage forms such as intraoral fast disintegrating tablets, intraoral fast dissolving tablets and chewable tablets, which can be easily taken without water when feeling symptoms of diarrhea, and block off unpleasant taste
  • Dosage forms such as sugar-coated tablets and film-coated tablets may be particularly preferred.
  • the therapeutic agents of the present invention can be manufactured by or in a manner conventional in the art.
  • it can be prepared by mixing the powdery active ingredient with a pharmaceutically acceptable carrier component (excipient etc.) and compression molding (direct compression method), and the drop agent is a mold
  • a pharmaceutically acceptable carrier component excipient etc.
  • compression molding direct compression method
  • the drop agent is a mold
  • powder granules such as granules, various granulation methods (extrusion granulation method, pulverization granulation method, dry consolidation granulation method, fluidized bed granulation method, rolling granulation method, high speed stirring It can be prepared by granulation method etc.).
  • Tablets can also be prepared by combining the above granulation method and tableting method (wet tableting method etc.) as appropriate (indirect compression method).
  • capsules can be prepared by filling powders (powders, granules, etc.) into capsules (soft or hard capsules) by a conventional method.
  • the tablets may be coated and formed into sugar-coated tablets or film-coated tablets.
  • the tablet may be a single layer tablet or a laminated tablet such as a double layer tablet.
  • the solution is prepared by dissolving or dispersing each component in a carrier component such as an aqueous medium (purified water, purified hot water, purified water containing ethanol, etc.), heating, filtering, clothing or sterilizing as necessary, and placing in a predetermined container It can be prepared by filling and sterilizing.
  • the amount of active ingredient in the therapeutic agent of the present invention generally varies depending on the dosage form, the amount of active ingredient is set, for example, within the range of about 0.01% by weight to about 95% by weight so as to achieve the desired dose.
  • the therapeutic agent of the present invention can be administered orally or parenterally (intravenous, intraarterial, subcutaneous, intradermal, intramuscular, or intraperitoneal injection, transdermal, transnasal, transmucosal, etc.) depending on its dosage form. Applies to Also, systemic and local administration may be employed. These administration routes are not mutually exclusive, and two or more optionally selected can be used in combination (for example, intravenous injection or the like simultaneously with oral administration or after a predetermined time has elapsed).
  • the therapeutic agent of the present invention contains the active ingredient in an amount necessary to obtain the expected therapeutic effect (ie, a therapeutically effective amount). Although the amount of the active ingredient in the therapeutic agent of the present invention generally varies depending on the dosage form, the amount of the active ingredient is set, for example, within the range of about 0.1% by weight to about 99% by weight so as to achieve the desired dose.
  • the dose of the therapeutic agent of the present invention is set so as to obtain the expected therapeutic effect.
  • the condition, age, sex, and weight of the patient, etc. are generally considered. Those skilled in the art can set appropriate dosages in consideration of these matters.
  • the dosage for crizotinib, 500 mg per day for adults and 280 mg / m 2 per child can be mentioned.
  • the administration schedule for example, once to several times a day, once every two days, or once every three days can be adopted. In preparation of the administration schedule, the patient's medical condition, the duration of the effect of the active ingredient and the like can be considered.
  • the therapeutic agent of the present invention may be used in combination with other agents.
  • the therapeutic agent of the present invention is added to standard chemotherapy for JMML to exert additive or synergistic effects.
  • Various combination modes are assumed, and the type and number of drugs used in combination, the administration schedule and the like are not particularly limited.
  • a representative example of the drug used in combination is an anticancer drug, a molecule-targeted therapeutic drug, cell therapy and the like may be used in combination.
  • anti-cancer agents examples include alkylating agents (eg cyclophosphamide, ifosfamide, melphalan), anti-metabolites (eg methotrexate, 6-mercaptopurine, cytarabine, fludarabine, clofarabine), anti-cancer antibiotics (eg Daunorubicin, doxorubicin, pirarubicin, idarubicin, mitoxantrone, plant alkaloids (eg vincristine, vinblastine, vindesine, etoposide), demethylating agents (eg azacitidine).
  • molecule-targeted therapeutic agents include MEK inhibitors, JAK inhibitors, GM-CSF inhibitors and the like.
  • CAR-T therapy and the like can be mentioned.
  • the present application also provides a method of treating JMML, which comprises administering a therapeutically effective amount of an ALK tyrosine kinase inhibitor or a JAK2 tyrosine kinase inhibitor to a JMML patient.
  • a JMML inspection method which includes the following steps (1) to (3). (1) preparing a sample containing leukemia cells isolated from a patient with juvenile myelomonocytic leukemia (2) in the sample, the presence or absence of a fusion gene of ALK or ROS1 or a fusion protein encoded by the fusion gene Detecting (3) determining that the fusion gene or the fusion protein is detected as compatible with treatment with an ALK tyrosine kinase inhibitor
  • a sample to be used for the test is prepared.
  • a sample containing leukemia cells isolated from JMML patients is used.
  • the type and origin of the sample are not particularly limited as long as they contain leukemia cells.
  • a cell fraction (bone marrow cells) prepared from bone marrow or a cell fraction (blood cells) prepared from blood such as peripheral blood is used as a sample.
  • Samples are prepared prior to the practice of the present invention. That is, the test method of the present invention does not include the step of isolating (collecting) bone marrow and the like for preparing a sample from a patient.
  • step (2) the ALK fusion gene (eg, DCTN1-ALK fusion gene or RANBP2-ALK fusion gene) or ROS1 fusion gene (eg, TBL1XR1-ROS1 fusion gene) in the sample, or the fusion gene (eg, DCTN1-ALK fusion gene)
  • the fusion gene eg, DCTN1-ALK fusion gene
  • genomic DNA or mRNA is to be detected.
  • telomere sequence analysis is not particularly limited.
  • RT-PCR reverse transcription-polymerase chain reaction
  • PCR method PCR method
  • PCR-RFLP restriction fragment length polymorphism
  • PCR-SSCP single strand conformation polymorphism
  • RNA sequence analysis target sequence analysis
  • FISH Fluorescence in situ hybridization, whole genome analysis, Invader (registered trademark, Third Wave Technologies) method
  • LAMP Loop-Mediated Isothermal Amplification
  • CGH Comparative Genomic Hybridization
  • dot hybridization Northern hybridization, etc.
  • Means of detection can be used. Extraction and purification of genomic DNA and mRNA can be performed by known methods. A variety of kits for preparation are also commercially available, and they may be used.
  • the DCTN1-ALK fusion gene, the TBL1XR1-ROS1 fusion gene and the RANBP2-ALK fusion gene are novel fusion genes derived from JMML patients, which were found by the present inventors.
  • the breakpoints and structures of these fusion genes are shown in FIG. Based on the information, primers and probes to be used for detection of each fusion gene can be designed and prepared. In addition, specific examples of primers that can be used for detection means (RT-PCR method and PCR method described above) utilizing nucleic acid amplification reaction will be described later.
  • the fusion protein can be detected by immunoassay.
  • Immunological assays allow for rapid and sensitive detection. Also, the operation is simple.
  • an antibody against a fusion protein is used, and the fusion protein is detected with the binding ability (binding amount) of the antibody as an index.
  • immunoassays include Western blotting, immunohistochemistry, fluorescence immunoassay (FIA), enzyme immunoassay (EIA), radioimmunoassay (RIA), flow cytometry (FCM) Immunoprecipitation, immunochromatography, ELISA, etc.
  • step (3) based on the detection results of step (2), the suitability for treatment with ALK tyrosine kinase inhibitor (that is, whether or not it is a patient to be treated) is evaluated. Specifically, when a fusion gene or fusion protein to be detected in step (2) is detected, it is determined as "compatible with treatment with an ALK tyrosine kinase inhibitor".
  • a genetic abnormality characterized by the formation of the above-mentioned fusion gene hereinafter referred to as "the genetic abnormality of the present invention” and the like as an index for determining a therapeutic course or obtaining supplementary information or basis therefor. Is sometimes used).
  • the determination here can be performed automatically / mechanically without depending on the judgment of a person having expert knowledge such as a doctor or a laboratory technician, as is clear from the determination criteria.
  • the examination method of the present invention is carried out before or after the start of treatment. If the test method of the present invention is performed before the start of treatment and the results are used, more appropriate treatment becomes possible. That is, the present invention can be used as a means for formulating and determining a treatment plan. On the other hand, if the test method of the present invention is performed after the start of treatment, for example, the present invention can be used to change the treatment policy. That is, the present invention is also useful as a means to provide a scientific basis for reviewing a treatment plan. The test method of the present invention may be carried out several times sequentially to monitor the treatment effect and revise / revision the treatment policy.
  • the present invention further provides a method of screening for a substance effective for the treatment of JMML, based on the results of identification of a fusion gene as a new therapeutic target in JMML.
  • the substance identified by the screening method of the present invention is particularly a therapeutic agent for patients who are positive for ALK fusion gene (eg, DCTN-ALK fusion gene, RANBP2-ALK fusion gene) or ROS1 fusion gene (eg, TBL1XR1-ROS1 fusion gene) or It is promising as a therapeutic drug candidate.
  • ALK fusion gene eg, DCTN-ALK fusion gene, RANBP2-ALK fusion gene
  • ROS1 fusion gene eg, TBL1XR1-ROS1 fusion gene
  • the following steps (i) to (iii) are performed.
  • (ii) culturing the cell in the presence of a test substance culturing the cell in the presence of a test substance;
  • cells expressing an ALK fusion gene for example, DCTN1-ALK fusion gene, RANBP2-ALK fusion gene
  • a ROS1 fusion gene for example, TBL1XR1-ROS1 fusion gene
  • the prepared cells are cultured in the presence of the test substance.
  • the number of cells to be used is not particularly limited, and can be determined in consideration of detection sensitivity, experimental equipment, and the like. For example, 1 ⁇ 10 2 to 1 ⁇ 10 6 cells can be used in one screening operation.
  • the amount (addition amount) of the test substance in the culture solution can be set arbitrarily, but the addition amount may be set within a range that does not have a fatal effect when normal cells are cultured under the same conditions. Those skilled in the art can set appropriate addition amounts by preliminary experiments.
  • the culture time is set so that the action and effect of the test substance can be sufficiently evaluated, but is not particularly limited.
  • the culture time can be set in the range of 10 minutes to January.
  • the culture time in the subsequent screening can be set based on the time required for the substance to exhibit the action / effect.
  • organic compounds or inorganic compounds of various molecular sizes can be used as the test substance.
  • organic compounds include nucleic acids, peptides, proteins, lipids (simple lipids, complex lipids (phosphoglycerides, sphingolipids, glycosylglycerides, cerebrosides, etc.), prostaglandins, isoprenoids, terpenes, steroids, polyphenols, catechins, vitamins
  • the test substance may be derived from natural products or synthetically, In the latter case, an efficient screening system can be constructed using, for example, a combinatorial synthesis technique. Extracts, culture supernatants, etc. may be used as a test substance, and existing drugs, nutritional foods, food additives, etc. may be used as test substances etc. Testing by simultaneously adding two or more types of test substances Interactions between substances, synergy, etc. may be examined.
  • step (iii) following step (ii) the number of viable cells or the number of colonies after culture is measured to determine the cell growth inhibitory activity (cytotoxic activity) of the test substance, that is, the effectiveness.
  • the cell growth inhibitory activity cytotoxic activity
  • cells cultured in the presence of the test substance (test group) and cells cultured in the absence of the test substance (control group) are prepared, and the number of surviving cells or the number of colonies is measured for each group and compared. Do. From the comparison results, the extent to which the cell viability / number of colonies has changed as a result of the presence of the test substance is determined.
  • the test substance is effective against JMML when the number of living cells / colony number in the test group is small (cell viability is low) as compared to the control group, ie, when the test substance shows cell growth inhibitory activity. It can be determined that there is. In one aspect, in this case, it is determined that the test substance is effective against a novel fusion gene positive case. If a significant decrease in the survival rate is observed in the test group, it can be determined that the efficacy of the test substance is particularly high. It is also possible to determine the efficacy of the test substance by comparing the number of cells before and after culture in the test group, instead of setting a control group. However, setting the control group as described above can give more reliable results.
  • a test group in which cells are cultured in the presence of a cytokine (for example, GM-CSF can be used) and a test (cultured with no cytokine) group in which cells are cultured in the absence And determine based on the number of viable cells / number of colonies in both test groups.
  • a control group in which the cells are cultured in the absence of the test substance for each, and to make a judgment after comparing and evaluating the test group and the control group.
  • the substance (screening product) selected by the screening method of the present invention is a strong candidate (lead compound) as an active ingredient of a medicine for JMML (particularly, a case where a novel fusion gene is positive).
  • a medicine for JMML particularly, a case where a novel fusion gene is positive.
  • the selected substance has sufficient medicinal effects, it can be used as it is as an active ingredient of a medicine.
  • the drug does not have sufficient medicinal effects, it can be used as an active ingredient of a medicine after being modified such as chemical modification to enhance its medicinal effects.
  • similar modifications may be made for the purpose of further enhancing the efficacy.
  • the present invention further provides three novel fusion genes responsible for the onset of JMML, ie, a DCTN1-ALK fusion gene, a TBL1XR1-ROS1 fusion gene, and a RANBP2-ALK fusion gene.
  • the DCTN1-ALK fusion gene is A breakpoint is present in exon 28, a breakpoint is present in intron 19 in the ALK gene, and has a structure in which up to exon 26 of the DCTN gene and exon 20 or later of the ALK gene are linked.
  • the TBL1XR1-ROS1 fusion gene has a breakpoint in intron 12 in the TBL1XR1 gene, a breakpoint in exon 36 in the ROS1 gene, and a portion of the intron 12 of the TBL1XR1 gene and the ROS1 gene It has a structure in which exons 36 and beyond are linked.
  • the RANBP2-ALK fusion gene has a breakpoint at intron 18 in the RANBP2 gene, a breakpoint at intron 19 in the ALK gene, and the exon 18 of the RANBP2 gene is linked to the exon 20 and beyond of the ALK gene It has the following structure.
  • a typical genomic nucleotide sequence of a DCTN1-ALK fusion gene is a cDNA nucleotide sequence as SEQ ID NO: 1, a typical genomic nucleotide sequence of a TBL1XR1-ROS1 fusion gene as SEQ ID NO: 2, and a cDNA nucleotide sequence as SEQ ID NO: 3.
  • the typical genomic nucleotide sequence of the RANBP2-ALK fusion gene is shown in SEQ ID NO: 5
  • the cDNA nucleotide sequence is shown in SEQ ID NO: 6, respectively.
  • the present invention also provides a fusion protein which is the expression product of a novel fusion gene.
  • a DCTN-ALK fusion protein encoded by a DCTN1-ALK fusion gene a DCTN1-ALK fusion gene
  • a TBL1XR1-ROS1 fusion protein encoded by a TBL1XR1-ROS1 fusion gene and a RANBP2-ALK fusion protein encoded by a RANBP2-ALK fusion gene
  • the DCTN1-ALK fusion protein comprises a coiled coil domain from RANBP2, a kinase domain from ALK.
  • the TBL1XR1-ROS1 fusion protein contains an FN type III domain from TBL1XR1, a kinase domain from ROS1.
  • the RANBP2-ALK fusion protein contains a TRP domain derived from RANBP2, a kinase domain derived from ALK.
  • a typical amino acid sequence of a DCTN1-ALK fusion protein is shown in SEQ ID NO: 7
  • a typical amino acid sequence of a TBL1XR1-ROS1 fusion protein is shown in SEQ ID NO: 8
  • a typical amino acid sequence of a RANBP2-ALK fusion protein is shown in SEQ ID NO: 9 It shows each.
  • the fusion gene and fusion protein of the present invention are useful, for example, as a therapeutic target or a diagnostic marker, or as an indicator for judging the suitability of a therapeutic agent.
  • the fusion gene and fusion protein of the present invention can be prepared in an isolated state, for example, by separating and purifying from a patient who is positive for the corresponding fusion gene. Alternatively, they may be prepared by chemical synthesis, genetic engineering techniques, etc. based on the sequence information disclosed herein.
  • ⁇ PCR based target deep sequence analysis Gene regions of PTP N11, NRAS, KRAS, CBL, NF1, SETBP1, JAK3, SH3BP1 using Quick Taq HS DyeMix (TOYOBO) from DNA obtained from 86 JMML patients and 6 NS / MPD patients Is amplified and purified using QIAquick PCR Purification Kit (QIAGEN), NotI (Fermentas), T4 DNA ligase (Takara Bio), and then analyzed for analysis using Illumina pair-end library protocol (Illumina) I created a rally. After that, sequencing and analysis were performed in the same procedure as exome analysis.
  • QIAGEN QIAquick PCR Purification Kit
  • NotI Fermentas
  • T4 DNA ligase Takara Bio
  • RNA sequence analysis> From RNA obtained from 117 JMML patients and 12 NS / MPD patients, a library for RNA sequence analysis was created using NEBNext Ultra RNA Prep Kit for Illumina (New England Biolabs). Next generation sequencing was performed using HiSeq 2500 (Illumina), and the resulting sequence data was analyzed using Tophat-fusion software to detect fusion genes.
  • LIN28B was most significantly upregulated in hypermethyl profile Identified.
  • a fusion gene related to receptor tyrosine kinase is reported to be a fusion of a kinase domain of tyrosine kinase and a domain related to dimer formation of a fusion partner, which is expected to lead to gain of function
  • the TBL1XR1-ROS1 fusion protein also retains the kinase domain of ROS1 and the domain associated with dimerization of TBL1XR1, strongly suggesting that this fusion leads to gain of function.
  • RanBP2-ALK tumor growth inhibition in vitro effect of crizotinib in positive patients (1) a method RanBP2-ALK fusion gene positive patients (UPN168) of CD34-positive cells extracted from the bone marrow (1 ⁇ 10 3 cells) were MethoCult TM H4434 classic (cytokines (+)) or MethoCult TM H4230 (cytokines (-)) (STEMCELL Technologies Inc.) crizotinib diluted in DMSO (Sigma-Aldrich, Inc.) to (ALK / ROS1 / MET inhibitor Pfizer) to 0,20, The cells were cultured for 2 weeks in a medium supplemented with 80, 200 and 500 nM. Two weeks later, the number of colony forming unit-granulocyte macrophage (CFU-GM) and burst-forming unit-erythroid (BFU-E) in each medium was counted.
  • CFU-GM colony forming unit-granulocyte macrophage
  • BFU-E
  • RANBP2-ALK cDNA primer set (design position RANBP2 exon 15, ALK exon 20, product size 671 bp)> Forward (RANBP2-ALK forward primer): ATTTTTCACAGAGAGGCAGAAAGACATTGA (SEQ ID NO: 10) Reverse (RANBP2-ALK reverse primer): GTCTTGCCAGCAAAGCAGTAG (SEQ ID NO: 11)
  • crizotinib exhibited a tumor suppressor effect in vitro, suggesting that it may be effective in this case.
  • crizotinib 280 mg / m 2 / day
  • the dose of crizotinib was determined based on the results of a phase I trial of crizotinib for pediatric patients with refractory solid tumors and anaplastic large cell lymphoma (Mosse, YP et al. Safety and activity of crizotinib for paediatric patients with refractory Lancet Oncol 14, 472-80 (2013)) solid tumours or anaplastic large-cell lymphoma: a Children's Oncology Group phase 1 consortium study.
  • RNA extracted from peripheral blood mononuclear cells collected on the 135th day was negative.
  • ALK break apart FISH and monosomy 7 FISH were similarly confirmed to be negative, and it was judged that molecular biological solution was achieved.
  • bone marrow transplantation was performed from HLA-matched siblings, and disease-free survival was confirmed 11 months after transplantation.
  • crizotinib was clinically useful in RANBP2-ALK positive JMML cases. The remaining two cases with the ALK-ROS1 fusion gene did not receive crizotinib and died of tumor progression.
  • crizotinib inhibited JMML colony formation in a concentration-dependent manner. This effect was stronger in the KRAS and PTPN11 gene mutation positive cases than in the CBL mutation cases. Crizotinib is negative for ALK fusion gene, suggesting that classical RAS gene mutation-positive cases may suppress tumor growth.
  • the hypermethylation profile group showed an "AML-type" methylation and expression profile with poor prognosis, even though blasts did not increase to the level of AML at diagnosis.
  • the various clinical biomarkers reported so far may reflect different aspects of the same poor prognosis group of JMML patients; clinical features (age, HbF levels, platelet count) and LIN28B expression It suggests the possibility that it can be used as a surrogate marker for this poor prognosis group.
  • ALK tyrosine kinase inhibitor other than crizotinib and luxoritinib JK2 tyrosine kinase inhibitor
  • method PTPN11 p.D61 V mutation positive and CD34 positive cells (1 ⁇ 10 3 ) as MethoCult TM H4434 classic (cytokine (+)) or Metho Cult TM H4230 (cytokine (-)) (STEMCELL Technologies) in a medium supplemented with alectinib, seritinib or TAE 684 or luxolitinib diluted with DMSO (Sigma-Aldrich) at a predetermined concentration Incubated for 2 weeks. As a control, a test group to which crizotinib was added was provided. Two weeks later, the number of colony forming unit-granulocyte macrophage (CFU-GM) in each medium was counted.
  • CFU-GM colony forming unit-granulocyte macrophage
  • the therapeutic agents of the present invention provide new therapeutic strategies for JMML.
  • the present invention can contribute to the improvement of the treatment result of JMML, the improvement of prognosis and the like.
  • New fusion genes identified from JMML cases are useful not only as therapeutic targets but also as test / diagnostic markers for JMML.
  • SEQ ID NO: 10 Description of artificial sequences: RANBP2-ALK forward primer
  • SEQ ID NO: 11 Description of artificial sequences: RANBP2-ALK reverse primer

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Molecular Biology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Immunology (AREA)
  • Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Analytical Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Zoology (AREA)
  • Microbiology (AREA)
  • Urology & Nephrology (AREA)
  • Wood Science & Technology (AREA)
  • Biophysics (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Cell Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Le problème que cherche à résoudre la présente invention est de fournir un nouvel agent thérapeutique à ciblage moléculaire contre la leucémie myélomonocytaire juvénile, et l'utilisation de ce dernier. L'invention concerne un agent thérapeutique contre la leucémie myélomonocytaire juvénile contenant un inhibiteur de la tyrosine kinase ALK ou un inhibiteur de la tyrosine kinase JAK2. L'invention concerne également un nouveau gène de fusion qui régule la leucémie myélomonocytaire juvénile.
PCT/JP2018/030471 2017-08-25 2018-08-17 Agent thérapeutique contre la leucémie myélomonocytaire juvénile WO2019039390A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2019537597A JPWO2019039390A1 (ja) 2017-08-25 2018-08-17 若年性骨髄単球性白血病治療薬

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2017-162389 2017-08-25
JP2017162389 2017-08-25

Publications (1)

Publication Number Publication Date
WO2019039390A1 true WO2019039390A1 (fr) 2019-02-28

Family

ID=65438724

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2018/030471 WO2019039390A1 (fr) 2017-08-25 2018-08-17 Agent thérapeutique contre la leucémie myélomonocytaire juvénile

Country Status (2)

Country Link
JP (1) JPWO2019039390A1 (fr)
WO (1) WO2019039390A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111411157A (zh) * 2020-04-29 2020-07-14 中国医学科学院血液病医院(中国医学科学院血液学研究所) 一种用于评估幼年型粒单核细胞白血病预后的全基因组dna甲基化检测方法

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2017502994A (ja) * 2014-01-17 2017-01-26 ノバルティス アーゲー Shp2の活性を阻害するための1−(トリアジン−3−イル/ピリダジン−3−イル)−ピペリジン/ピペラジン誘導体およびその組成物

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2017502994A (ja) * 2014-01-17 2017-01-26 ノバルティス アーゲー Shp2の活性を阻害するための1−(トリアジン−3−イル/ピリダジン−3−イル)−ピペリジン/ピペラジン誘導体およびその組成物

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CAYE A ET AL.: "Juvenile myelomonocytic leukemia displays mutations in components of the RAS pathway and the PRC2 network", NAT GENET., vol. 47, no. 11, 2015, pages 1334 - 1340, XP055577900, ISSN: 1061-4036 *
HAYASHI A ET AL., CRIZOTINIB TREATMENT FOR PEDIATRIC ACUTE MYELOBLASTIC LEUKEMIA WITH RANBP2-ALK FUSION GENE, vol. 56, no. 9, 2015, pages 1694, ISSN: 0485-1439 *
MAESAKO Y ET AL., MAJOR CYTOGENETIC RESPONSE AT 3 MONTHS OF CRIZOTINIB TREATMENT IN RANBP-ALK ACUTE MYELOID LEUKEMIA, vol. 55, no. 9, pages 1551, ISSN: 0485-1439 *
SALETTA ET AL.: "Molecular profiling of childhood cancer : Biomarkers and novel therapies", BBA CLINICAL, vol. 1, 2014, pages 59 - 77, XP055501424, ISSN: 2214-6474 *
TENRI MED BULL., vol. 18, no. 2, 2015, pages 80 - 85, ISSN: 1344-1817 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111411157A (zh) * 2020-04-29 2020-07-14 中国医学科学院血液病医院(中国医学科学院血液学研究所) 一种用于评估幼年型粒单核细胞白血病预后的全基因组dna甲基化检测方法

Also Published As

Publication number Publication date
JPWO2019039390A1 (ja) 2020-07-30

Similar Documents

Publication Publication Date Title
JP7323393B2 (ja) ファルネシルトランスフェラーゼ阻害剤を用いて癌患者を治療する方法
US20220090205A1 (en) Methods for diagnosing and treating cancer by means of the expression status and mutational status of nrf2 and downstream target genes of said gene
JP2019507754A (ja) 少なくとも2つの非必須アミノ酸を欠くダイエタリー製品
JP2013543008A (ja) Braf阻害剤による治療方法
AU2017306588B2 (en) Use of mitochondrial activity inhibitors for the treatment of poor prognosis acute myeloid leukemia
US20230000856A1 (en) Novel uses of crenolanib
EP3436601A1 (fr) Niveau d'expression de la cytidine désaminase dans un cancer en tant que nouvelle cible thérapeutique
JP5948332B2 (ja) Mll再構成白血病の治療法
WO2019039390A1 (fr) Agent thérapeutique contre la leucémie myélomonocytaire juvénile
WO2015066432A1 (fr) Méthode de traitement de post-néoplasies myéloprolifératives (nmp) et de la leucémie aiguë myéloïde post-nmp
US20150119446A1 (en) Cul4b as predictive biomarker for cancer treatment
CA2855356A1 (fr) Biomarqueurs de la sensibilite a des inhibiteurs du proteasome
US20220002396A1 (en) Methods of treating cancer with farnesyltransferase inhibitors
Reuther Myeloproliferative neoplasms: Molecular drivers and therapeutics
US20220218694A1 (en) Crenolanib for treating flt3 mutated proliferative disorders associated mutations
US20210251980A1 (en) Crenolanib for treating flt3 mutated proliferative disorders associated mutations
WO2023002983A1 (fr) Procédé de détermination de la sensibilité à un inhibiteur de fgfr
WO2017139470A1 (fr) Classification et traitement du cancer gastrique
US20210145823A1 (en) Crenolanib for treating flt3 mutated proliferative disorders associated mutations
WO2021255518A1 (fr) Compositions et méthodes de traitement de la leucémie myéloïde aiguë
Zlobine Host immunity determines outcome of eukaryotic initiation factor-4A inhibition in breast cancer
LYMPHOCYTIC PROTEOMIC EXPLORATION OF CELL SURFACE LANDSCAPE REVEALS NEW LEUKEMIA ASSOCIATED FEATURES
Sammarelli et al. Stem Cells and Growth Factors
Ullrich et al. Freie Vorträge
TW201030338A (en) Method for predicting therapeutic effect of combination chemotherapy

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: 18847950

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019537597

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18847950

Country of ref document: EP

Kind code of ref document: A1