WO2016098042A1 - Utilisation du céritinib (ldk -378) dans le traitement de troubles médiés par fes ou fer, en particulier des troubles prolifératifs - Google Patents

Utilisation du céritinib (ldk -378) dans le traitement de troubles médiés par fes ou fer, en particulier des troubles prolifératifs Download PDF

Info

Publication number
WO2016098042A1
WO2016098042A1 PCT/IB2015/059725 IB2015059725W WO2016098042A1 WO 2016098042 A1 WO2016098042 A1 WO 2016098042A1 IB 2015059725 W IB2015059725 W IB 2015059725W WO 2016098042 A1 WO2016098042 A1 WO 2016098042A1
Authority
WO
WIPO (PCT)
Prior art keywords
ceritinib
fes
fer
treatment
disease
Prior art date
Application number
PCT/IB2015/059725
Other languages
English (en)
Inventor
Laure Christina BOUCHEZ
Bertran GERRITS
Johannes Voshol
Enrico FUNHOFF
Natasa Zamurovic RIBRIOUX
Original Assignee
Novartis Ag
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 Novartis Ag filed Critical Novartis Ag
Publication of WO2016098042A1 publication Critical patent/WO2016098042A1/fr

Links

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
    • A61K31/495Heterocyclic 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Definitions

  • the present invention relates to use of ceritinib in disorder or a disease mediated by the activity of kinase. It also relates to pharmaceutical composition comprising ceritinib that is suitable for the treatment of such disorder or disease.
  • FES feline sarcoma
  • FER cytoplasmic non-receptor protein-tyrosine kinase family. Fes oncogene was first isolated from the tumour-causing feline retroviruses and once compared with cellular homologues from other species it was confirmed that it corresponds to the same gene. FER is closely related to FES (P Greer, Nature Reviews Molecular Cell Biology 3, 278-289, April 2002). In human, FES and FER are encoded by paralogous human FES and FER genes located at chromosome positions 15q26.1 and 5q21 , retrospectively (P Greer et al, Frontiers in Bioscience S4, 489-501 , 2012).
  • FES and FER proteins Beside the protein-tyrosine kinase domain, which is at carboxy-terminus, FES and FER proteins include also SRC-homology (SH2), coiled-coil (CC) and F-BAR (the Bin-Amphiphysin-Rvs) domains.
  • SH2 SRC-homology
  • CC coiled-coil
  • F-BAR the Bin-Amphiphysin-Rvs
  • Ceritinib (also named LDK378) was disclosed as compound 66 in Example 7 of WO2008/073687.
  • Chemical formula of ceritinib is 5-chloro-N2-(2-isopropoxy-5-methyl-4- (piperidin-4-yl)phenyl)-N4-[2-(propane-2-sulfonyl)-phenyl]-pyrimidine-2,4-diamine.
  • ALK anaplastic lymphoma kinase
  • ALK gene has been found to be rearranged, mutated, or amplified in a series of tumours, including non-small cell lung cancer.
  • FES and FES-related kinase comprise a unique subfamily of protein-tyrosine kinases (PTKs) that signal downstream of several classes of receptors for growth factors, cytokines and immunoglobulins, such as for example IL-6, FGF2, SCF and PDGF.
  • PTKs protein-tyrosine kinases
  • FES and FER kinases have been linked also to the regulation of cell-cell and cell-matrix interactions, as well as cell division, cell morphology, cell migration, and thus to tumorigenesis and development of metastasis in cancer. Therefore, it is postulated that inhibition of FES and FER kinases by ceritinib can be well employed in the treatment of a disorder or a disease mediated by the activity of FES and/or FER.
  • ceritinib for use in the treatment of a disorder or a disease mediated by the activity of FES and/or FER.
  • the disclosure also provides use of ceritinib for the manufacture of a medicament for the treatment of a disorder or a disease in a subject mediated by the activity of FES and/or FER; or a method for the treatment of a disorder or a disease mediated by the activity of FES and/or FER comprising the step of administering to a subject a therapeutically effective amount of ceritinib.
  • Second aspect of the disclosure is a pharmaceutical composition comprising a ceritinib for use in the treatment of a disorder or a disease mediated by the activity of FES and/or FER.
  • Third aspect of the disclosure is a method of modulating FES and/or FER activity in a subject, comprising the step of administering to a subject a therapeutically effective amount of ceritinib.
  • Figure 4 Effects of LDK378 on cell viability and apoptosis induction in LPS stimulated rat BM- derived macrophages after 5 h of treatment
  • FIG. 5 Effects of Ceritinib on TNFa protein level in LPS-stimulated rat BM derived macrophages after 5 h of treatment
  • ceritinib Functionality of ceritinib was assessed in a chemical proteomics experiment, where proteins that ceritinib binds to were identified. Surprisingly, the top hit of ceritinib chemical proteomics experiment, identified with the highest significance, was the non-receptor tyrosine kinase FER. Biochemical validation confirmed very potent inhibition of FER kinase by ceritinib (IC50 8 nM), as well as its related kinase FES (IC50 10nM). Based on the proven antitumor effect of ceritinib and the link between FES and FER kinases and certain tumor types, ceritinib can be utilized to treat a disorder or a disease mediated by the activity of FES and/or FER.
  • FER refers to tyrosine-protein kinase FER. Unless specifically stated otherwise, FER as used herein, refers to human gene and protein listed under accession numbers NM 005246.2 / NP_005237.2, respectively.
  • FES refers to tyrosine-protein kinase FES and all its isoforms 1 to 4. Unless specifically stated otherwise, FES as used herein, refers to human gene and protein listed under accession number NM_002005.3 / NP_001996.1 for tyrosine-protein kinase FES/Fps isoform 1 , NM_1 143783.1 / NP_001 137255.1 for tyrosine-protein kinase FES/Fps isoform 2, NM_001 143784.1 / NP_001 137256.1 for tyrosine-protein kinase FES/Fps isoform 3 and/or NM_001 143785.1 / NP_001 137257.1 for tyrosine-protein kinase FES/Fps isoform 4.
  • disorder or a disease mediated by FES and/or FER denotes an ailment, symptom, condition, disorder or a disease caused by an increased differential expression of the nucleic acid or polypeptide, of gene number of FES and/or FER, or function or activity of FES and/or FER in a cell or tissue compared to a control.
  • Disorder or a disease mediated by the activity of FES and/or FER is the ailment, symptom, condition, disorder or a disease can be alleviated, inhibited, prevented and/or ameliorated by inhibiting the increased differential expression of the nucleic acid or polypeptide, of gene number of FES and/or FER, or function or activity of FES and/or FER in a cell by a FES and/or FER inhibitor, such as Ceritinib, at least to the extent that the expression of the nucleic acid or polypeptide, of gene number of FES and/or FER, or function or activity of FES and/or FER in a cell or tissue is the same as in the control.
  • An FES and/or FER inhibitor can also be an antibody.
  • Disorder or a disease mediated by FES and/or FER can be a proliferative disorder or disease.
  • the term means cancer.
  • Cancer can mean presence of cells possessing characteristics typical of cancer-causing cells, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, and certain characteristic morphological features. Often, cancer cells will be in the form of a tumor, but such cells may exist alone or may circulate in the blood stream as independent cells, such as leukemic cells.
  • the cancer mediated by FES and/or FER can be hematological cancer or a solid tumor, particularly leukemia, lung, brain, colorectal or vascular (hemangioma) cancer.
  • Leukemia can be acute myeloid leukemia (AML) or chronic myelogenous leukemia.
  • Ceritinib can be used to target FES and FER in leukemias, because the studies identified them to be potential downstream effectors of growth or survival signaling in leukemias (A Craig, Frontiers in Bioscience 17, 861 - 875, 2012). Further studies showed that the tyrosine kinase FES is an essential effector of KITD816V proliferation signal.
  • Breitkopf et al identified FER as a potential target downstream of the BCR-ABL oncoprotein in chronic myelogenous leukemia (J Proteome Res, 2010).
  • the two kinases carry the BAR domain. Proteins with the BAR domain have been found to play a major role in remodeling cellular membranes linked with organelle biogenesis, membrane trafficking, (Frost A, et all, Cell, 137(2):191 -6, 2009).
  • the BAR domain superfamily of proteins is evolutionarily conserved with representative members present from yeast to man.
  • FER kinase is associated with cytoplasmic domain of N-cadherin (El Sayegh et al, Mol Biol Cell, 16, 5514-5527, 2005), as well as E-cadherin in adherence junction complexes of epithelial cells (Harder et al, Cell, 133, 1 1 18, 2008).
  • FER kinase By phosphorylating actin-interacting protein cortactin, FER kinase participates in the adherence junction interaction with cytoskeleton and regulating intercellular adhesion strength (Mege et al, Curr Opin Cell Biol, 18, 541 -548, 2006). FER kinase promotes breast cancer metastasis by regulating a6- and ⁇ -integrin-dependent cell adhesion and anoikis resistance (Ivanova et al, Oncogene. 2013 Dec 1 2;32(50) :5582-92). FER kinase is elevated in NSCLC tumors and is important for cellular invasion and metastasis (Ahn et al, Mol Cancer Res. 2013 Aug;1 1 (8):952-63).
  • FES absence attenuates tumor-associated angiogenesis and the metastasis-promoting functions of tumor-associated macrophages (Zhang et al., Cancer Res 201 1 ;71 :1465-1473).
  • FES kinase promotes mast cell recruitment to mammary tumors via the Stem Cell Factor/KIT receptor signaling axis (Kwok et al, Mol Cancer Res. 201 2 Jul;1 0(7) :881 - 91 ). Therefore, it is expected that Ceritinib can reduce, delay or prevent metastasis, by reducing invasiveness of tumors or by the effect on the cells of the tumor niche.
  • a “control” refers to the expression of the nucleic acid or polypeptide, of gene number of FES and/or FER, or function or activity of FES and/or FER in a normal cell.
  • the expression, function or the activity of FES and/or FER can be compared between the cancer cell and a normal cell, such as for example noncancerous cell from a healthy tissue, or a healthy subject.
  • Differential expression, function or activity means a measurable difference in expression, function or activity of FES and/or FER when compared to the control.
  • the differential expression can mean upregulation of FES and/or FER compared to the control, meaning for example increased DNA copy number of the FES and/or FER, their genomic DNA, cDNA or RNA sequence copies; increased gene expression, protein expression, mRNA expression; functional effect of the protein; functional effect of the FES and/or FER gene, cDNA or mRNA; protein, cDNA, gene or mRNA activity, or mutation status like for example frame-shift mutation, deletion, translocation, insertion, duplication, inversion, functional mutation, or combinations thereof that translates into increased activity of FES and/or FER, compared to the control.
  • a differentially mutated gene when applied to a gene, it can refer to the differential mutation status of a gene compared to a normal, wildtype gene in a normal cell.
  • a differentially mutated gene can be mutated as compared to the wildtype gene, which causes activation and increased function of the mutated gene. It can also refer to the differential production of the mRNA transcribed and/or translated from the gene or the protein product encoded by the gene.
  • a differentially expressed gene may be overexpressed (or upregulated) or underexpressed (or downregulated) as compared to the expression level of a normal or control cell.
  • increased differential expression or upregulation is an increase in gene expression and generally is at least 1 .25 fold or, alternatively, at least 1 .5 fold or, alternatively, at least 2 fold, or alternatively, at least 3 fold or alternatively, at least 4 fold expression over that detected in a normal or control counterpart cell or tissue.
  • the term "differentially expressed” also refers to where expression in a cancer cell or cancerous tissue is different compared to the expression in a control (for example control cell or normal tissue, e.g. non-cancerous cell or tissue) . Similar comparison can be applied when measuring if FES and/or FER are "activated” compared to the control.
  • Activated FES and/or FER would mean that the activity of the proteins is more pronounced in treated cell compared to control, for example phosphorylation level of a substrate is at least 1 .5 fold or, alternatively, at least 2 fold, or alternatively, at least 3 fold or alternatively, at least 4 fold higher in cancerous or treated cell compared to control.
  • Methods used in measuring the expression, function or activity of FES and/or FER are generally known in the art, and in any event would be applied the same to the cell and the control.
  • expression of the nucleic acid can be assessed by measuring levels of messenger RNA using a RT-PCR.
  • Levels of protein and phosphorylated protein can be monitored by using FES and/or FER specific antibodies.
  • Level of FES and/ or FER activity can be monitored by phosphorylation level of its direct protein substrates.
  • the disorder or a disease mediated by the activity of FES and/or FER is also an ailment, symptom, condition, disorder or a disease in a subject that can be alleviated, inhibited, prevented and/or ameliorated by administering Ceritinib at the dose that causes blood concentration to reach C i 0 o d of 10 ⁇ or less, preferably at 5 ⁇ , preferably C i 0 o d of 2 ⁇ or less. Therefore, a method of modulating FES and/or FER activity in a subject is also provided herein, the method comprising the step of administering to a subject a therapeutically effective amount of ceritinib.
  • a therapeutically effective amount of a compound of the present disclosure refers to an amount of ceritinib that will elicit the biological or medical response of a subject.
  • the effect can be reduction or inhibition of FES and/or FER, or the protein activity, or ameliorate symptoms, alleviate conditions, slow or delay disease progression etc.
  • a therapeutically effective amount refers to the amount of the compound of the present disclosure that, when administered to a subject, is effective to (1 ) at least partially alleviate, inhibit, prevent and/or ameliorate a condition, or a disorder or a disease (i) mediated by FES and/or FER and/or mediated by FES and/or FER activity, or (ii) characterized by activity (normal or abnormal) of FES and/or FER; or (2) reduce or inhibit the activity of FES and/or FER; or (3) reduce or inhibit the expression of FES and/or FER.
  • the therapeutically effective amount of Ceritinib would be in the range of 1 mg - 1500 mg, preferably from 100 mg to 1000 mg, more preferably between 150 mg and 750 mg.
  • the dose can be administered daily in a single or in divided doses.
  • the compound to be used in the treatment of the disorder or disease is to be administered to a subject, which refers to an animal.
  • a subject also refers to, for example, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like.
  • the subject is a primate.
  • the subject is a human.
  • Ceritinib can be formulated in a pharmaceutical composition in order to be used for the purposes described above, particularly for use in the treatment of a disorder or a disease mediated by FES and/or FER.
  • the composition can contain further pharmaceutical excipients.
  • the pharmaceutically acceptable excipients used can be for example filler, disintegrant, glidant, and/or lubricant, or mixture thereof. Further excipients like antioxidants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof can also be added.
  • the composition can comprise one or more fillers, for example microcrystalline cellulose, silicified microcrystalline cellulose, lactose (anhydrous, monohydrate), mannitol, sorbitol, calcium phosphate (dibasic anhydrous, dibasic hydrate, tribasic), isomalt, sucrose, Hydroxypropyl Cellulose Low-substituted; one or more disintegrants, for example Croscarmellose Sodium, Crospovidone, Sodium starch glycolate; further binders, such as for example starch,
  • Hypromellose Hydropropyl Cellulose Low-substituted, Hypromellose (Hydroxypropylmethyl cellulose), Copovidone (Copolyvidone), gelatine, polymethacrylates
  • one or more glidants for example Silica, colloidal silica, talc, hydrophobic colloidal silica, magnesium silicate
  • one or more lubricants for example Magnesium stearate, calcium stearate, zinc stearate, glyceryl mono fatty acid, glyceryl monostearate, glyceryl dibehenate, glyceyryl palmito stearic ester, or
  • polyoxyethylen glycol or mixtures thereof. They can be mixed in the composition in a powder form, as separately granulated granules, or dissolved in a fluid, which can then be substantially removed by drying throughout the process, or added after going through the process of briquetting or slugging, and optionally sieving.
  • the pharmaceutical composition can be formulated for example in a tablet or a capsule by tableting or filling the composition in a capsule, respectively.
  • the effective dosage of the compound to treat the disorder or disease mediated by FES and/or FER may vary depending on a pharmaceutical composition employed, the mode of administration, the condition being treated, and the severity of the condition being treated. Thus, a dosage regimen for the treatment would be selected in accordance with a variety of factors including the route of administration and the renal and hepatic function of the patient. A clinician or physician of ordinary skill can determine and prescribe the effective amount of the single therapeutic agents required to alleviate, counter or arrest the progress of the condition.
  • the linker was synthesized from the commercially available 4-chlorobutan-1 -ol after treatment with sodium azide in a solution of DMSO at 85°C.
  • the hydroxyl moiety was first transformed into its corresponding mesylate under standard conditions.
  • the iodine was inserted after nucleophilic substitution, using sodium iodide in acetone at room temperature.
  • the coupling of the linker with Ceritinib was successfully achieved in presence of potassium carbonate in DMF, after 24h of trituration.
  • the crude mixture was diluted with EtOAc (20 mL) and the organic phase was washed with 1 M HCI (2x 20 mL). The 2 layers were separated and a 1 M solution of NaOH was added to the aqueous phase until the pH was basic (pH ⁇ 10). The aqueous layer was extracted with EtOAc (3x 20 mL) and the organic phases were combined and washed successively with water (2x 20 mL), saturated NaCI (2x 20 mL), and dried over MgS0 4 . After filtration and evaporation under reduced pressure, the crude residue was quickly purified through a pad of celite to afford the desired compound 5 (90.6 mg, 94%) as a white solid and with high purity (>95%).
  • Rat biliopancreatic duct tissue was quickly removed during necropsy, washed in PBS, and immediately flash frozen in liquid N 2 to minimize degradation processes. Several ducts were pooled. Subsequently rat biliopancreatic duct tissue was thawed and homogenized in lysis buffer (50 mM Tris-HCI , pH 7.4, 100 mM NaCI, 2 mM MgCI 2 , 5 % glycerol, 0.8 % NP-40 and protease inhibitors) at a volume to weight ratio of 8. After clearing by centrifugation, the lysate was split in 4 equal samples, each containing approximately 10 mg of protein. Lysate samples were then incubated with either DMSO or 0.3, 3, or 30 ⁇ of ceritinib for 30 min at 4°C.
  • lysis buffer 50 mM Tris-HCI , pH 7.4, 100 mM NaCI, 2 mM MgCI 2 , 5 % gly
  • Sepharose-bound linker-ceritinib was added and incubation was continued for another 2 h at 4°C.
  • Sepharose beads were then separated from the unbound proteins by centrifugation, transferred to a spin column and washed with 10 ml of lysis buffer. Finally, bound proteins were eluted in sample buffer, separated by SDS-PAGE and visualized with colloidal Coomassie Blue for excision. Bands were excised from each lane at an equidistant space. Disulfide bonds from the protein were reduced using dithiotreitol and the free thiols that were generated alkylated with iodoacetamide. Proteins were subsequently digested in-gel by trypsin. Peptides were eluted from the gel pieces and labeled with iTRAQ reagent.
  • Peptides from bands of the same Mw across the gel were pooled and analyzed by LC-MS. Peptides were identified by querying against humanJPI database using the Mascot search algorithm and proteins were quantified by combining iTRAQ reporter ions across all identified peptides using an in-house analysis pipeline. Specific binders to Ceritinib were identified based on their depletion from the bound protein fraction in the samples containing the competitor compound relative to the DMSO control (Table 1 ). Kinase inhibitory potency of identified kinases were measured at Nanosys.
  • RNA isolation was performed immediately after. RNA was isolated using Rneasy plus mini kit (Qiagen; cat n°74134), according to the manufacturer's instruction. cDNA synthesis and quantitative PCR Total RNA was reverse transcribed to cDNA using the High Capacity cDNA Archive Kit (Applied Biosystems, cat n° 4368813) according to the manufacturer instructions, using the DNA Engine Dyad PTC-220 (BioRad).
  • cDNA was synthesized starting from 300 ng of total RNA and adding a mix preparation to a final volume of 100 ⁇ _.
  • Real-time PCR was performed using ABI prismTM 7900HT (Applied Biosystems) in combination with TaqMan® Fast Universal PCR Master Mix (Applied Biosystems, cat n°4352042). The PCR conditions were as follow: one cycle at 95°C for 20 sec, followed by 40 cycles of amplification: a denaturation step at 95°C for 1 s and annealing/elongation step at 60°C for 20 sec.
  • Human probe/primer sets was also used on rat RNA, purchased commercially from Applied Biosystems. 18S (Hs99999901_s1 ) was used as internal standard for real time PCR analysis.
  • the relative quantification of gene expression changes was performed using the standard curve method.
  • the standard curve is generated from 8 dilution series (10 fold dilution) constructed from a "reference" sample.
  • "Reference” sample was a commercial mRNA from rat liver (Ambion cat n° 09508186B). The standard curve was used to generate expression data expressed in molecule number.
  • mRNA expression of kinases Fert2 and Fes was higher in epithelial tissues such as duodenum as compared to liver and pancreas, and correlated well with Keratin 19 expression level.
  • the expression of Alk kinase, primary Ceritinib target was virtually absent from all the tissues analyzed.
  • NRC-1 Normal rat cholangiocyte
  • NRC-1 cells were cultivated in NRC-1 medium. Cells were maintained up to passage 36 into T75 collagen I coated flask (Biocoat). For splitting cells were washed with 2:1 PBS:Trypsin-EDTA (Gibco), and then incubated at 37°C/5 % C02 for approximately 10-15 min with 1 ml of trypsin-EDTA. Once all cells were detached, 9 ml of NRC-1 medium were added and cells were counted. Then 1 .10E6 cells were seeded into a T75 collagen I flask for 48 hours maintenance. Viability and apoptosis testing
  • NRC-1 were seeded at 8000 or 16000 cells/well in BD BioCoatTM Collagen I, 96-well plates (BD, #354650). Cells were grown for 2 days at 37°C, 5 % C02, at which point 8000 cells/well reached 70 % confluence, while 16000 cells/well reached 100 % confluence. Medium was then replaced with fresh NRC-1 medium containing compound dilutions (1 .3 ⁇ -100 ⁇ ) for 3 h or 24 hours. NRC-1 medium containing 1 % DMSO was used as control.
  • Cell viability after treatment was determined by measuring ATP content using CellTiter-Glo® Luminescent Cell Viability Assay (Promega), according to manufacturer's instruction.
  • Luminescent Cell Viability Assay Promega
  • cells were seeded at 20000 cells/well in BD BioCoatTM Collagen I, 96-well plates (BD, #354650). Cells were grown for 4 days at 37°C, 5 % C0 2 , at which point they were fully confluent. Cell seeding density corresponded to seeding for barrier function analysis using ECIS technology. Medium was then replaced with fresh NRC-1 medium containing Ceritinib dilutions (1 ⁇ -32 ⁇ ) for 24 hours.
  • NRC-1 medium containing 1 % DMSO was used as a control.
  • Treatment with 20 ⁇ taxol and 1 ⁇ staurosporine was used as a positive control for apoptosis induction.
  • Cell viability after treatment was determined by using CellTiter-Blue® Cell Viability Assay (Promega), according to manufacturer's instruction.
  • NRC-1 medium was removed from each well and replaced by 300 ⁇ of fresh NRC-1 medium.
  • baseline measurement at 37°C 5 % C0 2 was performed in order to allow barrier stabilization.
  • Cells were stimulated with 33 ⁇ of 10x compounds solution pre-diluted into DMEM-F12 (Gibco). During compounds injection, measurement was paused for few seconds.
  • Cells were transfected by electroporation with Amaxa 96-well plate system (Lonza), with P1 primary cell 96-well Nucleofector Kit (Lonza cat # V4SP-1096). Cells were grown up to 80 % confluence in T75 collagen I flask and trypsinized in order to prepare a cell suspension. From the cell suspension, after 5 minutes centrifugation at 800 rpm, a solution of 3.5x10E6 cells/ml was prepared in P1 complemented nucleofector solution, in order to have 0.7x10E5 cells/20 ⁇ in each well. Each condition was prepared in Eppendorf tubes with 300 nM of the corresponding siRNA. From these cells/siRNA mixes, 20 ⁇ replicates were dispensed into each well.
  • Electroporation was performed in the 96-well plate using the Amaxa electroporation machine
  • RNA extraction 50 ⁇ of electroporated cells were seeded per well, in duplicates, into collagen I coated 96 well plates containing 150 ⁇ /well of pre-warmed NRC-1 medium. Cells were grown for 72 hours at 37°C/5 % C0 2 in order to reach confluence and form barrier, then medium was washed-out and 150 ⁇ of RLT lysis buffer (Qiagen RNeasy 96 Kit #74181 ) were added into each well. RNA was extracted according to manufacturer's instruction.
  • Femurs were taken from rats. BM cells were isolated by flushing the femurs with HBSS
  • rat M-CSF recombinant rat M-CSF (Peprotech).
  • Cell suspensions were put then into 175 cm2 flasks (Falcon). After one day of culture, non-adherent cells and medium were harvested, centrifuged (1400 rpm, 5 minutes, room temperature) and only the cell-free supernatants were put back into the respective flasks for further six days. After differentiation to BM-derived macrophages, medium was removed and adherent cells were detached by cold PBS/2 mM EDTA
  • Viability of stimulated BM-derived macrophages after treatment was determined by using CellTiter-Blue® Cell Viability Assay (Promega), according to manufacturer's instruction.
  • Apoptosis induction was quantified by using Apo-ONE® Homogeneous Caspase-3/7 Assay (Promega), according to manufacturer's instruction.
  • a broad kinase inhibitor staurosporin was used as positive control for apoptosis induction.
  • IL-6 was under detection limit in this experiment. Ceritinib in different concentrations (without LPS addition) had no effects on TNFa release. In contrast, LPS induced a strong TNFa protein release compared to control and Ceritinib showed a dose-dependent decrease in LPS-induced TNFa levels after 5 h of treatment (Fig 5). Identical results were obtained after 6 hours of treatment.
  • Ceritinib was tested in BaF3 cells driven by FES or FER kinase.
  • the measured IC50 was 3.65 ⁇ and 1 .65 ⁇ in BaF3/FES and BaF3/FER line, respectively.

Abstract

La présente invention concerne l'utilisation de céritinib (LDK378) dans le traitement d'un trouble ou une maladie médiée par l'activité des kinases FES et/ou FER, en particulier le traitement de troubles prolifératifs. L'invention concerne en outre des compositions pharmaceutiques comprenant du céritinib qui sont appropriées pour le traitement de tels troubles ou maladies.
PCT/IB2015/059725 2014-12-19 2015-12-17 Utilisation du céritinib (ldk -378) dans le traitement de troubles médiés par fes ou fer, en particulier des troubles prolifératifs WO2016098042A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462094285P 2014-12-19 2014-12-19
US62/094,285 2014-12-19

Publications (1)

Publication Number Publication Date
WO2016098042A1 true WO2016098042A1 (fr) 2016-06-23

Family

ID=55069037

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2015/059725 WO2016098042A1 (fr) 2014-12-19 2015-12-17 Utilisation du céritinib (ldk -378) dans le traitement de troubles médiés par fes ou fer, en particulier des troubles prolifératifs

Country Status (1)

Country Link
WO (1) WO2016098042A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021195706A1 (fr) * 2020-03-31 2021-10-07 Children's Medical Research Institute Nouveaux inhibiteurs de la dynamine et leurs utilisations
CN114901289A (zh) * 2019-10-28 2022-08-12 奥斯陆大学 用于治疗alk阴性癌症和浆细胞介导的疾病的alk抑制剂

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008073687A2 (fr) 2006-12-08 2008-06-19 Irm Llc Composés et compositions inhibant la protéine kinase
WO2012082972A1 (fr) * 2010-12-17 2012-06-21 Novartis Ag Formes cristallines de la 5-chloro-n2-(2-isopropoxy-5-méthyl-4-pipéridin-4-yl-phényl)-n4[2-(propane-2-sulfonyl)-phényl]-pyrimidine-2,4-diamine
WO2014074580A1 (fr) * 2012-11-07 2014-05-15 Novartis Ag Polythérapie

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008073687A2 (fr) 2006-12-08 2008-06-19 Irm Llc Composés et compositions inhibant la protéine kinase
WO2012082972A1 (fr) * 2010-12-17 2012-06-21 Novartis Ag Formes cristallines de la 5-chloro-n2-(2-isopropoxy-5-méthyl-4-pipéridin-4-yl-phényl)-n4[2-(propane-2-sulfonyl)-phényl]-pyrimidine-2,4-diamine
WO2014074580A1 (fr) * 2012-11-07 2014-05-15 Novartis Ag Polythérapie

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
A CRAIG, FRONTIERS IN BIOSCIENCE, vol. 17, 2012, pages 861 - 875
AHN ET AL., MOL CANCER RES., vol. 11, no. 8, August 2013 (2013-08-01), pages 952 - 63
CONDORELLI ET AL., CURR MED CHEM, vol. 18, 2011, pages 2913 - 2920
E VOISSET ET AL., BLOOD, vol. 110, 2007, pages 2593 - 9
E VOISSET ET AL., LEUKEMIA, vol. 24, 2010, pages 721 - 8
E VOISSET ET AL: "FES kinases are required for oncogenic FLT3 signaling", LEUKEMIA., vol. 24, no. 4, 1 April 2010 (2010-04-01), US, pages 721 - 728, XP055249692, ISSN: 0887-6924, DOI: 10.1038/leu.2009.301 *
EI SAYEGH ET AL., MOL BIOL CELL, vol. 16, 2005, pages 5514 - 5527
FROST A, CELL, vol. 137, no. 2, 2009, pages 191 - 6
HARDER ET AL., CELL, vol. 133, 2008, pages 1118
IVANOVA ET AL., ONCOGENE, vol. 32, no. 50, 12 December 2013 (2013-12-12), pages 5582 - 92
J PROTEOME RES, 2010
JIANYONG CHEN ET AL: "LDK378: A Promising Anaplastic Lymphoma Kinase (ALK) Inhibitor", JOURNAL OF MEDICINAL CHEMISTRY, vol. 56, no. 14, 25 July 2013 (2013-07-25), US, pages 5673 - 5674, XP055249558, ISSN: 0022-2623, DOI: 10.1021/jm401005u *
KWOK ET AL., MOL CANCER RES., vol. 1 0, no. 7, July 2012 (2012-07-01), pages 881 - 91
MASANORI KAWAKAMI, INT J CLIN EXP PATHOL, vol. 6, 2013, pages 598 - 612
MEGE ET AL., CURR OPIN CELL BIOL, vol. 18, 2006, pages 541 - 548
P GREER ET AL., FRONTIERS IN BIOSCIENCE, vol. S4, 2012, pages 489 - 501
P GREER, NATURE REVIEWS MOLECULAR CELL BIOLOGY, vol. 3, April 2002 (2002-04-01), pages 278 - 289
XU ET AL., NATURE IMMUNOLOGY, vol. 13, no. 6, June 2012 (2012-06-01)
ZHANG ET AL., CANCER RES, vol. 71, 2011, pages 1465 - 1473

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114901289A (zh) * 2019-10-28 2022-08-12 奥斯陆大学 用于治疗alk阴性癌症和浆细胞介导的疾病的alk抑制剂
WO2021195706A1 (fr) * 2020-03-31 2021-10-07 Children's Medical Research Institute Nouveaux inhibiteurs de la dynamine et leurs utilisations

Similar Documents

Publication Publication Date Title
US20230346786A1 (en) Chiral diaryl macrocycles and uses thereof
EP3843850B1 (fr) Composés pyrazolo[3,4-b]pyridine utilisés en tant qu'inhibiteurs de kinases tam et met
Axelrod et al. Axl as a mediator of cellular growth and survival
US10202391B2 (en) Chemical compounds
US20190300511A1 (en) 2-aryl- and 2-heteroaryl-substituted 2-pyridazin-3(2h)-one compounds as inhibitors of fgfr tyrosine kinases
US10172835B2 (en) Anticancer agent composition
Maubach et al. NF-kappaB-inducing kinase in cancer
KR20160147934A (ko) 폐 선암종을 치료하는 방법
TW202019427A (zh) 以喹唑啉化合物作為有效成分之醫藥組成物
US20140018372A1 (en) Crystalline form of a indolinone derivative and its use
TW202136252A (zh) 化合物及其用途
Guo et al. Molecular characteristics of CTA056, a novel interleukin-2-inducible T-cell kinase inhibitor that selectively targets malignant T cells and modulates oncomirs
AU2013307383A1 (en) Aminoheteroaryl compounds as MTH1 inhibitors
EP3906234A1 (fr) Composés de quinoléine utilisés en tant qu'inhibiteurs de kinases tam et met
WO2016098042A1 (fr) Utilisation du céritinib (ldk -378) dans le traitement de troubles médiés par fes ou fer, en particulier des troubles prolifératifs
Chan et al. Discovery of 3-aminopyrazole derivatives as new potent and orally bioavailable AXL inhibitors
KR20190128660A (ko) 암의 치료에 이용하기 위한 usl-311
AU2014266278A1 (en) Therapeutic effect prediction method for colorectal cancer patient in whom expression of TK1 protein has increased
JP2019526617A (ja) 治療用化合物
Hilal Involvement of Interleukin-6 Induced PI3K/Akt/mTor Pathway in the Regulation of Telomerase and Alpha-Fetoprotein Expression in Hepatocellular Carci-noma
JP2024514958A (ja) ユビキチン特異的ペプチダーゼ22(usp22)の阻害剤、並びに疾患及び障害の治療のためのその使用
CN117561255A (zh) 作为mek抑制剂的3,4-二氢-2,7-萘啶-1,6(2h,7h)-二酮化合物
JP2022509257A (ja) 2,3-ジヒドロ-イソインドール-1-オン化合物を用いた組合せ療法及び様々な変異を有する患者を治療するための方法
TW201912160A (zh) 外顯子18及/或外顯子21突變型egfr之選擇性抑制劑
WO2008011799A1 (fr) Nouvelle utilisation d'inhibiteurs spécifiques de la tyrosine kinase c-ab1 non réceptrice

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: 15819882

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: 15819882

Country of ref document: EP

Kind code of ref document: A1