WO2015186137A1 - 1h-1,8- naphthyridin-2ones as anti proliferative compounds - Google Patents

1h-1,8- naphthyridin-2ones as anti proliferative compounds Download PDF

Info

Publication number
WO2015186137A1
WO2015186137A1 PCT/IN2014/000777 IN2014000777W WO2015186137A1 WO 2015186137 A1 WO2015186137 A1 WO 2015186137A1 IN 2014000777 W IN2014000777 W IN 2014000777W WO 2015186137 A1 WO2015186137 A1 WO 2015186137A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
benzamide
naphthyridin
equiv
trifluoromethyl
Prior art date
Application number
PCT/IN2014/000777
Other languages
French (fr)
Inventor
Amala Kompella
Venugopala Krishna Gampa
Srinivasulu GANGANAMONI
Balakrishna Reddy SIRIGIREDDY
Kali Satya Bhujanga Rao Adibhatla
Venkaiah Chowdary Nannapaneni
Original Assignee
Natco Pharma Limited
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=52589723&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2015186137(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to CN201480079519.1A priority Critical patent/CN106573929A/en
Priority to KR1020177000113A priority patent/KR20170016921A/en
Priority to AP2016009604A priority patent/AP2016009604A0/en
Priority to BR112016028674A priority patent/BR112016028674A2/en
Priority to AU2014396394A priority patent/AU2014396394A1/en
Priority to MX2016015350A priority patent/MX2016015350A/en
Priority to JP2017516226A priority patent/JP2017516867A/en
Priority to SG11201610149YA priority patent/SG11201610149YA/en
Priority to US15/316,832 priority patent/US9765072B2/en
Priority to EA201692371A priority patent/EA201692371A1/en
Priority to EP14838923.2A priority patent/EP3152205A1/en
Priority to CA2950250A priority patent/CA2950250A1/en
Application filed by Natco Pharma Limited filed Critical Natco Pharma Limited
Publication of WO2015186137A1 publication Critical patent/WO2015186137A1/en
Priority to IL249118A priority patent/IL249118A0/en
Priority to MA39524A priority patent/MA39524A1/en
Priority to PH12016502432A priority patent/PH12016502432A1/en
Priority to HK17104965.4A priority patent/HK1231469A1/en
Priority to US15/681,278 priority patent/US10047088B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • 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
    • A61K31/4353Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic 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 ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid

Definitions

  • the present invention relates to novel antiproliferative lH-1, 8-naphthyridin-2-ones of the general formula (I) or pharmaceutically acceptable salts thereof:
  • variable groups are as defined herein, and their preparation and use in therapeutic treatment of disorders related to inhibition of tyrosine kinases in warm blooded animals.
  • Protein tyrosine kinases are large family of proteins which plays a central role in the regulation of several disorders, particularly in the management of proliferative disorders. Deregulation of tyrosine kinase activity has emerged as a major mechanism by which cancer cells evade normal physiologic constraints on growth, proliferation and survival. An important mechanism leading to tyrosine kinase deregulation is mutation.
  • Chronic myeloid leukaemia (CML) is a chronic myelodysplastic hematopoietic stem cell disorder syndrome. 95% of the CML are resulting from a reciprocal translocation between chromosome-9 and chromosome-22 of Philadelphia chromosome.
  • the fusion gene produces a 210 KDa mutant protein in which the first exon of c-ABL has been replaced by BCR sequences, encoding either 927 or 902 amino acid.
  • the BCR-ABL chimeric gene product has a tyrosine kinase activity several fold higher than its normal counterpart and correlates with the disease phenotype.
  • Tyrosine kinase forms a significant share of all onco proteins taking a centre stage as possible targets for cancer therapy.
  • the anticancer drug Gleevec/ Glivec/ Imatinib Mesylate (Novartis STI571) is a block buster drug for the treatment of CML and c-kit positive metastatic GIST. Gleevec selectively and effectively inhibits the kinase activity of BCR-ABL fusion protein, which is responsible for the constitutive proliferative signaling. While Imatinib is therapeutically highly effective, with improving prospects over time for sustained remission and potential to severely limit or eliminate disease progression and transformation, a good number of patients either fail or respond sub optimally to Imatinib. Disease eradication may not be possible with Imatinib.
  • Distinct patterns of resistance have evolved with the use of Imatinib, and Abl kinase mutations, which alter Imatinib binding or favour kinase conformations inaccessible to Imatinib, Dasatinib and Nilotinib the available alternate Abl kinase inhibitors and restore hematologic and cytogenetic remission in the majority of patients with primary failure or acquired resistance in chronic phase.
  • Imatinib and Abl kinase mutations, which alter Imatinib binding or favour kinase conformations inaccessible to Imatinib, Dasatinib and Nilotinib the available alternate Abl kinase inhibitors and restore hematologic and cytogenetic remission in the majority of patients with primary failure or acquired resistance in chronic phase.
  • ABL kinase mutations generally cluster into four main categories and are associated with particular numbered amino acid residues: ATP binding loop (p-loop), particularly Y253 and E255 mutants; T315 mutants; M351 mutants; and activation loop (a-loop), particularly H396 mutants.
  • ATP binding loop p-loop
  • Y253 and E255 mutants T315 mutants
  • M351 mutants activation loop
  • a-loop activation loop
  • Omacetaxine (homoharringtonine) is approved by FDA for the treatment of adult patients with chronic or accelerated phase chronic myeloid leukemia (CML) with resistance and/or intolerance to two or more tyrosine kinase inhibitors (TKIs). However, it is administered subcutaneously with non-specific mechanism of action.
  • Other drug candidates include rebastinib (WO 2008/046003) and the Ponatinib (WO 2007/075869).
  • Ponatinib (ICLUSIG) is an approved as oral drug candidate and it is developed by Ariad pharmaceuticals for the treatment of chronic myeloid leukemia (CML) and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL).
  • CML chronic myeloid leukemia
  • Ph+ Philadelphia chromosome-positive acute lymphoblastic leukemia
  • Ponatinib was intended to target not only native BCR-ABL, but also its isoforms that carry mutations that confer resistance to treatment with existing tyrosine kinase inhibitors, including especially the T315I mutation for which no effective therapy exists.
  • the Food and Drug Administration temporarily suspended sales of the drug in the U.S. in 2013 because of "the risk of life-threatening blood clots and severe narrowing of blood vessels".
  • the present invention relates to a new family of lH-1, 8-naphthyridin-2-ones which are potent inhibitors of Abl tyrosine kinase and their mutated forms, including the T315I mutant.
  • the compounds of the present invention are devoid of some of the short comings of the existing drug products.
  • Figure- 1 shows x-ray powder diffraction patterns (XRPDs) for NRC 21 T
  • Figure-2 shows x-ray powder diffraction patterns (XRPDs) for NRC 21T hydrochloride salt
  • Figure-3 shows Anti-proliferative activity of NRC-21T on various Leukemial cell lines
  • Figure-4 shows Anti-Invasive property of NRC-21 T on Baf3/T3151 Cell line
  • Figure-5 shows Establishment of Antagonism of NRC21T in T315I induced tumour in Nude Mice
  • Figure-5A shows Establishment of Antagonism of NRC21T in T315I induced tumour in nude mice, pictures of tumour
  • Figure-5B shows Establishment of Antagonism of NRC21T in T315I induced tumour in nude mice, pictures of spleen
  • FIG. 6A shows Survival time study of NRC21T
  • Figure-6B shows Pictures of spleen under survival time study
  • the present invention relates to a compound of formula (I)
  • LI is Linker selected from unsaturated carbon bond; preferably carbon-carbon triple bond or carbon-carbon double bond;
  • L2 is a linker selected from NHC(O)-, C(0)NH-;
  • Ring A includes, but are not limited to substituted 5 or 6 membered aryl or heteroaryl ring.
  • ring A groups include:
  • the present invention provides a process for the preparation of compounds of formula (I) by palladium catalyzed Sonogashira coupling of 6-substituted naphthyridin-2-ones (II) with iodobenzamides (III).
  • the schematic representation is as follows:
  • Scheme-1 Several illustrative overall synthetic approaches to the preparation of two key intermediates II and III, based on known transformations, are illustrated in schemes 2 to 5.
  • Scheme-2 Represents preparation of intermediates-II: (First set)
  • Compound of formula (I) can be prepared as per above schemes.
  • Compound of formula (I) can be converted into an N-oxides, or its pharmaceutically acceptable salts.
  • suitable inorganic acids are, halogen acids, such as hydrochloric acid, sulfuric acid, or phosphoric acid.
  • Suitable organic acids are, for example, carboxylic, phosphonic, sulfonic or sulfamic acids, for example acetic acid, propionic acid, octanoic acid, decanoic acid, dodecanoic acid, glycolic acid, lactic acid, fumaric acid, succinic acid, adipic acid, pimelic acid, suberic acid, azelaic acid, malic acid, tartaric acid, citric acid, oxalic acid, amino acids, such as arginine or lysine, maleic acid, hydroxymaleic acid, methylmaleic acid, cyclohexanecarboxylic acid, adamantanecarboxylic acid, benzoic acid, -phenoxy benzoic acid, 2-acetoxy benzoic acid, salicylic acid, 4-aminosalicylic acid, phthalic acid, phenylacetic acid, mandelic acid, cinnamic acid, methane- or ethane-
  • salts for isolation or purification purposes it is also possible to use pharmaceutically unacceptable salts, for example picrates or perchlorates.
  • pharmaceutically acceptable salts or free compounds are employed (where applicable in the form of pharmaceutical preparations), and these are therefore preferred.
  • the compounds of the formula (I) or N-oxide or pharmaceutically acceptable salts thereof inhibit to varying degrees the receptor and non-receptor tyrosine kinases of all, which play a role in growth regulation and transformation in mammalian cells, including human cells.
  • the receptor tyrosine kinases may be kinases of the EGF family, e.g.
  • ErbB2 kinase HER-2
  • ErbB3 kinase ErbB4 kinase
  • insulin-like growth factor receptor kinase IGF-1 kinase
  • members of the PDGF-receptor tyrosine kinase family such as PDGF-a and PDGF- ⁇ receptor kinase, JAK-2, CSF-1 -receptor kinase, Phosphatidylinositol 3-kinases (PI-3-kinases or PDKs), AKT, CDK, mTOR, Kit-receptor kinase, Flt-3, Flt-4, FGFR-1, FGFR-3, FGFR-4, c-Met, RON, c-Ret, ALK and VEGF- receptor kinase.
  • PI-3-kinases or PDKs Phosphatidylinositol 3-kinases
  • the non-receptor tyrosine kinases may be kinases such as Abl/ Bcr-abl Kinase, Arg, kinases from the Src family, c-Src kinase, c-Yes. Lck, Lyn and Fyn.
  • the compounds of the present invention have been found to inhibit especially Abl/Bcr-Abl kinase, including mutant forms; Lyn and Lck kinases.
  • the compounds of the formula (I) or N-oxide or pharmaceutically acceptable salts thereof inhibit to varying degrees the mutant forms of Abl/Bcr-Abl kinase which include the mutants of the P-loop of the kinase i.e., L284V, G250E, Q225H, Y253F & E255K; the C-helix mutants of the kinase i.e., D276G and E279H; The ATP binding region mutants of the kinase i.e., V299L, T315I and F317L; SH2-contact mutant of the kinase i.e., M351T; substrate binding region mutant of the kinase i.e., F359V; the A-loop mutants of the kinase i.e.,L384M, H395P, H396R and G398R; and the C-terminal lobe mutant kinas
  • the compounds of the present invention have been found to inhibit especially the important mutants of abl/ Bcr-Abl kinases i.e., Q252H, Y253F, M351T, H396P and more particularly the compounds of formula (I) inhibit the highly resistant form of the mutated kinase i.e., the T3151 mutant.
  • the compounds of the present invention relates furthermore to a method for the treatment of a neoplastic disease or disorders dependent on tyrosine kinases especially chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), myelodysplastic syndrome, melanoma, germ cell tumours, gastrointestinal stromal tumour (GIST), non- small cell lung cancer (NSCLC), mastocytosis, neuroblastoma, glioblastoma, astrocytoma, hepatocellular carcinoma, renal cell cancer, breast cancer, cutaneous systemic sclerosis, prostate and colorectal cancer and other solid tumours, diabetes remissions.
  • CML chronic myelogenous leukemia
  • CLL chronic lymphocytic leukemia
  • ALL acute lymphoblastic leukemia
  • AML acute myeloid leukemia
  • MBE myelodysplastic
  • the present invention relates furthermore to a method for the treatment of a neoplastic disease which responds to an inhibition of a protein kinase activity, which comprises administering a compound of formula (I) or a N-oxide or a pharmaceutically acceptable salt thereof, in a quantity effective against said disease, to a warm-blooded animal requiring such treatment.
  • a method of treatment of proliferative disorders especially leukemia irrespective of etiology of the disorder, which respond to inhibition of the aforementioned tyrosine kinases, particularly the Abl/Bcr-Abl tyrosine kinase and one or more of its severe mutated forms.
  • the treatment comprises administering a compound of formula (I) or an N-oxide or a pharmaceutically acceptable salt thereof, in a quantity effective against said disease, to a warm-blooded animal requiring such treatment.
  • the biological efficacy of the compounds of the present invention has been established by In vitro efficacy evaluation on BCR-abl positive cell line K.562 and mutant cell lines Baf3/T315i, M351T, E255K and WT; Matrigel invasion assay; Determination of MTD of NRC21T; Establishment of Antagonism of NRC21T in T315I induced tumour in Nude Mice; Establishment of Survival time of NRC21T in SCID Mice.
  • the compounds of formula (I) according to the present invention shows therapeutic efficacy especially against disorders dependent on TK, especially in proliferative diseases.
  • the present invention also relates to pharmaceutical compositions comprising an affective amount of compound of formula (I) or a N-oxide or a pharmaceutically acceptable salt especially an amount effective in the prevention or therapy of one of the above mentioned diseases, of the active ingredient together with pharmaceutically acceptable carriers that are suitable for topical, enteral, for example oral or rectal, or parental administration, and may be inorganic or organic, solid or liquid.
  • pharmaceutical compositions of the present invention may contain one or more excipients or adjuvants. Selection of excipients and the amounts to use may be readily determined by the formulation scientist based upon experience and consideration of standard procedures and reference works in the field.
  • Diluents increase the bulk of a solid pharmaceutical composition, and may make a pharmaceutical dosage form containing the composition easier for the patient and care giver to handle.
  • Diluents for solid compositions include, for example, microcrystalline cellulose (e.g. Avicel®), microfine cellulose, lactose, starch, pregelatinized starch, calcium carbonate, calcium sulfate, sugar, dextrates, dextrin, dextrose, dibasic calcium phosphate dihydrate, tribasic calcium phosphate, kaolin, magnesium carbonate, magnesium oxide, maltodextrin, mannitol, polymethacrylates (e.g. Eudragit®), potassium chloride, powdered cellulose, sodium chloride, sorbitol and talc.
  • microcrystalline cellulose e.g. Avicel®
  • microfine cellulose lactose
  • starch pregelatinized starch
  • calcium carbonate calcium sulfate
  • sugar dextrates
  • dextrin
  • Solid pharmaceutical compositions that are compacted into a dosage form, such as capsules may include excipients whose functions include helping to bind the active ingredient and other excipients together after compression.
  • Binders for solid pharmaceutical compositions include acacia, alginic acid, carbomer (e.g. carbopol), carboxymethylcellulose sodium, dextrin, ethyl cellulose, gelatin, guar gum, hydrogenated vegetable oil, hydroxyethyl cellulose, hydroxypropyl cellulose (e.g. Klucel(R)), hydroxypropyl methyl cellulose (e.g. Methocel(R)), liquid glucose, magnesium aluminum silicate, maltodextrin, methylcellulose, polymethacrylates, povidone (e.g. Kollidon(R), Plasdone(R)), pregelatinized starch, sodium alginate and starch.
  • carbomer e.g. carbopol
  • carboxymethylcellulose sodium dextrin
  • the dissolution rate of a compacted solid pharmaceutical composition in the patient's stomach may be increased by the addition of a disintegrant to the composition.
  • Disintegrants include alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium (e.g. Ac-Di-Sol®, Primellose®), colloidal silicon dioxide, croscarmellose sodium, crospovidone (e.g. Kollidon®, Polyplasdone®), guar gum, magnesium aluminum silicate, methyl cellulose, microcrystalline cellulose, polacrilin potassium, powdered cellulose, pregelatinized starch, sodium alginate, sodium starch glycolate (e.g. Explotab®) and starch.
  • alginic acid include alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium (e.g. Ac-Di-Sol®, Primellose®), colloidal silicon dioxide, croscarmellose sodium, crospovidone (e.g. Kollidon®, Polyplasdone®
  • Glidants can be added to improve the flowability of a non-compacted solid composition and to improve the accuracy of dosing.
  • Excipients that may function as glidants include colloidal silicon dixoide, magnesium trisilicate, powdered cellulose, starch, talc and tribasic calcium phosphate.
  • a dosage form such as a capsule
  • the composition is subjected to pressure from a punch and dye.
  • Some excipients and active ingredients have a tendency to adhere to the surfaces of the punch and dye, which can cause the product to have pitting and other surface irregularities.
  • a lubricant can be added to the composition to reduce adhesion and ease the release of the product from the dye.
  • Lubricants include magnesium stearate, calcium stearate, glyceryl monostearate, glyceryl palmitostearate, hydrogenated castor oil, hydrogenated vegetable oil, mineral oil, polyethylene glycol, sodium benzoate, sodium lauryl sulfate, sodium stearyl fumarate, stearic acid, talc and zinc stearate.
  • Flavouring agents and flavour enhancers make the dosage form more palatable to the patient.
  • Common flavouring agents and flavour enhancers for pharmaceutical products include maltol, vanillin, ethyl vanillin, menthol, citric acid, fumaric acid, ethyl maltol, and tartaric acid.
  • Solid compositions may also be dyed using any pharmaceutically acceptable colorant to improve their appearance and/or facilitate patient identification.
  • reaction mass was filtered through a pad of silica gel at room temperature. The filtrate was concentrated and the compound was collected by filtration using propionitrile solvent at 0-5°C (60% yield).
  • Example-2 4-MethyI-N-[4-[(4-methyIpiperazin-l-yl) methyl]-3-(trifluoromethyl) phenyl]-3-[(E)- 2-(7-oxo-8H-l, 8-naphthyridin-3-yl) vinyl] benzamide (Development code: NRC20T)
  • Example-6 N-[3, 5-Bis (trifluoromethyl) phenyI]-4-methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3- yl)ethynyl]benzamide benzamide (Development code: NRC16T)
  • Example-10 In vitro efficacy evaluation:
  • Ponatinib was used as reference drug candidate for a comparative efficacy and safety evaluation.
  • a laboratory sample of the drug substance Ponatinib was synthesized in house employing the procedure disclosed in US8114874.
  • the experimental compounds and the standard reference drug (Ponatinib) were dissolved in cell culture medium and DMSO at a concentration of lOmM for in vitro studies.
  • the stock solution was further diluted with the same cell culture medium and used in concentrations of 0.1 nm to ⁇ .
  • Cell proliferation by MTT assay was done as follows. 1000 to 10,000 cells were seeded per well in 96-well plate and different concentrations of Pon-21T ranging from ⁇ to 0.1 nM were added in triplicates. After incubating the cells with NRC-21T for the required time period 24-72hrs, 15 ⁇ 1 of 5mg/ml MTT was added and incubated for additional 4 hours at 37°C and 5% C0 2 .
  • T315i cells (3X10 5 ) were suspended in 300 ⁇ 1 of serum- free medium and placed in the upper compartment of transwell chambers pre-coated with the matrigel (Millipore, Catalog No. ECM550, USA). The lower compartment of the chamber was filled with 500 ⁇ serum-medium (10% FBS) and the cells were allowed to migrate for 72hrs. After incubation, the cells were fixed and stained with the dye provided along with the kit and quantified using Elisa plate reader at 560nm. In Vitro matrigel assay of T315i cells in the presence of specified concentration of NRC-21T were represented in Table 2 and Figure-4.
  • NRC21T is a safer experimental drug than Ponatinib. Also NRC21T has comparable IC 50 values with respect to Ponatinib. Thus, the experimental drug of this invention, NRC21T is established as superior candidate in terms of safety and efficacy.
  • Group - 1 Positive control (3 Male + 3 Female)
  • Group - III Ponatinib (5 Male + 5 Female) (10 mg/kg, p.o)

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Diabetes (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Emergency Medicine (AREA)
  • Obesity (AREA)
  • Oncology (AREA)
  • Immunology (AREA)
  • Endocrinology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

The present invention relates to novel antiproliferative1H-1, 8-naphthyridin-2-ones of the general formula (I) or pharmaceutically acceptable salts thereof: In which the variable groups are as defined herein, and their preparation and use in therapeutic treatment of disorders related to inhibition of tyrosine kinases in warm blooded animals. The compounds can overcome imatinib induced drug resistance.

Description

FIELD OF THE INVENTION:
The present invention relates to novel antiproliferative lH-1, 8-naphthyridin-2-ones of the general formula (I) or pharmaceutically acceptable salts thereof:
Figure imgf000002_0001
Formula (I);
In which the variable groups are as defined herein, and their preparation and use in therapeutic treatment of disorders related to inhibition of tyrosine kinases in warm blooded animals. BACKGROUND OF THE INVENTION:
Protein tyrosine kinases are large family of proteins which plays a central role in the regulation of several disorders, particularly in the management of proliferative disorders. Deregulation of tyrosine kinase activity has emerged as a major mechanism by which cancer cells evade normal physiologic constraints on growth, proliferation and survival. An important mechanism leading to tyrosine kinase deregulation is mutation. Chronic myeloid leukaemia (CML) is a chronic myelodysplastic hematopoietic stem cell disorder syndrome. 95% of the CML are resulting from a reciprocal translocation between chromosome-9 and chromosome-22 of Philadelphia chromosome. Break point cluster region (BCR) sequences of chromosome-22 on translocation juxtaposes with the c-ABL tyrosine kinase of chromosome-9. The fusion gene produces a 210 KDa mutant protein in which the first exon of c-ABL has been replaced by BCR sequences, encoding either 927 or 902 amino acid. Another BCR-ABL fusion protein of 185 KDa containing BCR sequences from exon 1 fused to exon 2-11 of c-ABL, is found in 10% of adult ALL patients. The BCR-ABL chimeric gene product has a tyrosine kinase activity several fold higher than its normal counterpart and correlates with the disease phenotype. Tyrosine kinase forms a significant share of all onco proteins taking a centre stage as possible targets for cancer therapy. The anticancer drug Gleevec/ Glivec/ Imatinib Mesylate (Novartis STI571) is a block buster drug for the treatment of CML and c-kit positive metastatic GIST. Gleevec selectively and effectively inhibits the kinase activity of BCR-ABL fusion protein, which is responsible for the constitutive proliferative signaling. While Imatinib is therapeutically highly effective, with improving prospects over time for sustained remission and potential to severely limit or eliminate disease progression and transformation, a good number of patients either fail or respond sub optimally to Imatinib. Disease eradication may not be possible with Imatinib.
Distinct patterns of resistance have evolved with the use of Imatinib, and Abl kinase mutations, which alter Imatinib binding or favour kinase conformations inaccessible to Imatinib, Dasatinib and Nilotinib the available alternate Abl kinase inhibitors and restore hematologic and cytogenetic remission in the majority of patients with primary failure or acquired resistance in chronic phase. In the advanced disease and Philadelphia chromosome (Ph)+ ALL, responses are more limited and relapse is common.
ABL kinase mutations generally cluster into four main categories and are associated with particular numbered amino acid residues: ATP binding loop (p-loop), particularly Y253 and E255 mutants; T315 mutants; M351 mutants; and activation loop (a-loop), particularly H396 mutants. Modelling of Imatinib and other kinase inhibitors with the crystal structure of the catalytic region of the ABL kinase suggests that mutations may interrupt critical drug contact points or induce or favour a conformation of the Abl kinase in which drug binding is reduced or precluded. Now termed the "gatekeeper" position, mutations at threonine 315 confer resistance both to Imatinib and "second generation" Abl kinase inhibitors Nilotinib and Dasatinib.
Thus there is an unmet need with regard to treatment of patients having the T315I mutation. Omacetaxine (homoharringtonine) is approved by FDA for the treatment of adult patients with chronic or accelerated phase chronic myeloid leukemia (CML) with resistance and/or intolerance to two or more tyrosine kinase inhibitors (TKIs). However, it is administered subcutaneously with non-specific mechanism of action. Other drug candidates include rebastinib (WO 2008/046003) and the Ponatinib (WO 2007/075869). Ponatinib (ICLUSIG) is an approved as oral drug candidate and it is developed by Ariad pharmaceuticals for the treatment of chronic myeloid leukemia (CML) and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). Ponatinib was intended to target not only native BCR-ABL, but also its isoforms that carry mutations that confer resistance to treatment with existing tyrosine kinase inhibitors, including especially the T315I mutation for which no effective therapy exists. However the Food and Drug Administration temporarily suspended sales of the drug in the U.S. in 2013 because of "the risk of life-threatening blood clots and severe narrowing of blood vessels". This suspension was partially lifted subsequently with revised prescribing information, a new "Black Box Warning" and a "Risk Evaluation and Mitigation Strategy" in place. Thus there is a need for newer selective tyrosine kinase inhibitors which are orally active, safer than existing therapies particularly with regard to decrease in risk of life-threatening blood clots and severe narrowing of blood vessels and efficacious against the kinase mutations, including the T315I mutant. The present invention relates to a new family of lH-1, 8-naphthyridin-2-ones which are potent inhibitors of Abl tyrosine kinase and their mutated forms, including the T315I mutant. The compounds of the present invention are devoid of some of the short comings of the existing drug products.
BRIEF DESCRIPTION OF THE DRAWINGS:
Figure- 1 shows x-ray powder diffraction patterns (XRPDs) for NRC 21 T
Figure-2 shows x-ray powder diffraction patterns (XRPDs) for NRC 21T hydrochloride salt
Figure-3 shows Anti-proliferative activity of NRC-21T on various Leukemial cell lines Figure-4 shows Anti-Invasive property of NRC-21 T on Baf3/T3151 Cell line
Figure-5 shows Establishment of Antagonism of NRC21T in T315I induced tumour in Nude Mice Figure-5A shows Establishment of Antagonism of NRC21T in T315I induced tumour in nude mice, pictures of tumour
Figure-5B shows Establishment of Antagonism of NRC21T in T315I induced tumour in nude mice, pictures of spleen
Figure-6A shows Survival time study of NRC21T
Figure-6B shows Pictures of spleen under survival time study
DETAILED DESCRIPTION OF THE INVENTION:
The present invention relates to a compound of formula (I)
Figure imgf000005_0001
Formula (I)
or a pharmaceutically acceptable salts thereof; wherein,
LI is Linker selected from unsaturated carbon bond; preferably carbon-carbon triple bond or carbon-carbon double bond;
L2 is a linker selected from NHC(O)-, C(0)NH-;
Ring A includes, but are not limited to substituted 5 or 6 membered aryl or heteroaryl ring.
Illustrative examples of ring A groups include:
Figure imgf000006_0001
Figure imgf000007_0001
Figure imgf000008_0001
Compounds of formula I (a-u)
The present invention provides a process for the preparation of compounds of formula (I) by palladium catalyzed Sonogashira coupling of 6-substituted naphthyridin-2-ones (II) with iodobenzamides (III). The schematic representation is as follows:
Figure imgf000008_0002
Scheme-1 Several illustrative overall synthetic approaches to the preparation of two key intermediates II and III, based on known transformations, are illustrated in schemes 2 to 5. Scheme-2: Represents preparation of intermediates-II: (First set)
Me
Figure imgf000009_0001
II (L1 =
Scheme-3: Represents preparation of intermediates-II: (Second set)
Figure imgf000010_0001
II (L1
Scheme-4: Re resents preparation of intermediate-Ill and compound of formula (I)
Figure imgf000010_0002
Compound of formula (I) can be prepared as per above schemes. Compound of formula (I) can be converted into an N-oxides, or its pharmaceutically acceptable salts. For example, suitable inorganic acids are, halogen acids, such as hydrochloric acid, sulfuric acid, or phosphoric acid. Suitable organic acids are, for example, carboxylic, phosphonic, sulfonic or sulfamic acids, for example acetic acid, propionic acid, octanoic acid, decanoic acid, dodecanoic acid, glycolic acid, lactic acid, fumaric acid, succinic acid, adipic acid, pimelic acid, suberic acid, azelaic acid, malic acid, tartaric acid, citric acid, oxalic acid, amino acids, such as arginine or lysine, maleic acid, hydroxymaleic acid, methylmaleic acid, cyclohexanecarboxylic acid, adamantanecarboxylic acid, benzoic acid, -phenoxy benzoic acid, 2-acetoxy benzoic acid, salicylic acid, 4-aminosalicylic acid, phthalic acid, phenylacetic acid, mandelic acid, cinnamic acid, methane- or ethane- sulfonic acid, 2-hydroxyethanesulfonic acid, ethane- 1 ,2-disulfonic acid, benzenesulfonic acid, 2-naphthalenesulfonic acid, 1,5-naphthalene-disulfonic acid, 2-, 3- or 4- methylbenzenesulfonic acid, methylsulfuric acid, ethylsulfuric acid, dodecylsulfuric acid, N-cyclohexylsulfamic acid, N-methyl-, N-ethyl- or N-propyl-sulfamic acid, or other organic protonic acids, such as ascorbic acid.
For isolation or purification purposes it is also possible to use pharmaceutically unacceptable salts, for example picrates or perchlorates. For therapeutic use, only pharmaceutically acceptable salts or free compounds are employed (where applicable in the form of pharmaceutical preparations), and these are therefore preferred.
The compounds of the formula (I) or N-oxide or pharmaceutically acceptable salts thereof inhibit to varying degrees the receptor and non-receptor tyrosine kinases of all, which play a role in growth regulation and transformation in mammalian cells, including human cells. The receptor tyrosine kinases may be kinases of the EGF family, e.g. ErbB2 kinase (HER-2), ErbB3 kinase, ErbB4 kinase; insulin-like growth factor receptor kinase (IGF-1 kinase), especially members of the PDGF-receptor tyrosine kinase family, such as PDGF-a and PDGF-β receptor kinase, JAK-2, CSF-1 -receptor kinase, Phosphatidylinositol 3-kinases (PI-3-kinases or PDKs), AKT, CDK, mTOR, Kit-receptor kinase, Flt-3, Flt-4, FGFR-1, FGFR-3, FGFR-4, c-Met, RON, c-Ret, ALK and VEGF- receptor kinase. The non-receptor tyrosine kinases may be kinases such as Abl/ Bcr-abl Kinase, Arg, kinases from the Src family, c-Src kinase, c-Yes. Lck, Lyn and Fyn. The compounds of the present invention have been found to inhibit especially Abl/Bcr-Abl kinase, including mutant forms; Lyn and Lck kinases. The compounds of the formula (I) or N-oxide or pharmaceutically acceptable salts thereof inhibit to varying degrees the mutant forms of Abl/Bcr-Abl kinase which include the mutants of the P-loop of the kinase i.e., L284V, G250E, Q225H, Y253F & E255K; the C-helix mutants of the kinase i.e., D276G and E279H; The ATP binding region mutants of the kinase i.e., V299L, T315I and F317L; SH2-contact mutant of the kinase i.e., M351T; substrate binding region mutant of the kinase i.e., F359V; the A-loop mutants of the kinase i.e.,L384M, H395P, H396R and G398R; and the C-terminal lobe mutant kinase i.e., F486S.
The compounds of the present invention have been found to inhibit especially the important mutants of abl/ Bcr-Abl kinases i.e., Q252H, Y253F, M351T, H396P and more particularly the compounds of formula (I) inhibit the highly resistant form of the mutated kinase i.e., the T3151 mutant.
The compounds of the present invention relates furthermore to a method for the treatment of a neoplastic disease or disorders dependent on tyrosine kinases especially chronic myelogenous leukemia (CML), chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), myelodysplastic syndrome, melanoma, germ cell tumours, gastrointestinal stromal tumour (GIST), non- small cell lung cancer (NSCLC), mastocytosis, neuroblastoma, glioblastoma, astrocytoma, hepatocellular carcinoma, renal cell cancer, breast cancer, cutaneous systemic sclerosis, prostate and colorectal cancer and other solid tumours, diabetes remissions.
The present invention relates furthermore to a method for the treatment of a neoplastic disease which responds to an inhibition of a protein kinase activity, which comprises administering a compound of formula (I) or a N-oxide or a pharmaceutically acceptable salt thereof, in a quantity effective against said disease, to a warm-blooded animal requiring such treatment. In particular the present invention relates to a method of treatment of proliferative disorders especially leukemia, irrespective of etiology of the disorder, which respond to inhibition of the aforementioned tyrosine kinases, particularly the Abl/Bcr-Abl tyrosine kinase and one or more of its severe mutated forms. The treatment comprises administering a compound of formula (I) or an N-oxide or a pharmaceutically acceptable salt thereof, in a quantity effective against said disease, to a warm-blooded animal requiring such treatment.
The biological efficacy of the compounds of the present invention has been established by In vitro efficacy evaluation on BCR-abl positive cell line K.562 and mutant cell lines Baf3/T315i, M351T, E255K and WT; Matrigel invasion assay; Determination of MTD of NRC21T; Establishment of Antagonism of NRC21T in T315I induced tumour in Nude Mice; Establishment of Survival time of NRC21T in SCID Mice.
On the basis of these studies, the compounds of formula (I) according to the present invention shows therapeutic efficacy especially against disorders dependent on TK, especially in proliferative diseases.
The present invention also relates to pharmaceutical compositions comprising an affective amount of compound of formula (I) or a N-oxide or a pharmaceutically acceptable salt especially an amount effective in the prevention or therapy of one of the above mentioned diseases, of the active ingredient together with pharmaceutically acceptable carriers that are suitable for topical, enteral, for example oral or rectal, or parental administration, and may be inorganic or organic, solid or liquid. In addition to the active ingredient(s), the pharmaceutical compositions of the present invention may contain one or more excipients or adjuvants. Selection of excipients and the amounts to use may be readily determined by the formulation scientist based upon experience and consideration of standard procedures and reference works in the field. Diluents increase the bulk of a solid pharmaceutical composition, and may make a pharmaceutical dosage form containing the composition easier for the patient and care giver to handle. Diluents for solid compositions include, for example, microcrystalline cellulose (e.g. Avicel®), microfine cellulose, lactose, starch, pregelatinized starch, calcium carbonate, calcium sulfate, sugar, dextrates, dextrin, dextrose, dibasic calcium phosphate dihydrate, tribasic calcium phosphate, kaolin, magnesium carbonate, magnesium oxide, maltodextrin, mannitol, polymethacrylates (e.g. Eudragit®), potassium chloride, powdered cellulose, sodium chloride, sorbitol and talc.
Solid pharmaceutical compositions that are compacted into a dosage form, such as capsules may include excipients whose functions include helping to bind the active ingredient and other excipients together after compression. Binders for solid pharmaceutical compositions include acacia, alginic acid, carbomer (e.g. carbopol), carboxymethylcellulose sodium, dextrin, ethyl cellulose, gelatin, guar gum, hydrogenated vegetable oil, hydroxyethyl cellulose, hydroxypropyl cellulose (e.g. Klucel(R)), hydroxypropyl methyl cellulose (e.g. Methocel(R)), liquid glucose, magnesium aluminum silicate, maltodextrin, methylcellulose, polymethacrylates, povidone (e.g. Kollidon(R), Plasdone(R)), pregelatinized starch, sodium alginate and starch.
The dissolution rate of a compacted solid pharmaceutical composition in the patient's stomach may be increased by the addition of a disintegrant to the composition. Disintegrants include alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium (e.g. Ac-Di-Sol®, Primellose®), colloidal silicon dioxide, croscarmellose sodium, crospovidone (e.g. Kollidon®, Polyplasdone®), guar gum, magnesium aluminum silicate, methyl cellulose, microcrystalline cellulose, polacrilin potassium, powdered cellulose, pregelatinized starch, sodium alginate, sodium starch glycolate (e.g. Explotab®) and starch.
Glidants can be added to improve the flowability of a non-compacted solid composition and to improve the accuracy of dosing. Excipients that may function as glidants include colloidal silicon dixoide, magnesium trisilicate, powdered cellulose, starch, talc and tribasic calcium phosphate.
When a dosage form such as a capsule is made by the compaction of a powdered composition, the composition is subjected to pressure from a punch and dye. Some excipients and active ingredients have a tendency to adhere to the surfaces of the punch and dye, which can cause the product to have pitting and other surface irregularities. A lubricant can be added to the composition to reduce adhesion and ease the release of the product from the dye. Lubricants include magnesium stearate, calcium stearate, glyceryl monostearate, glyceryl palmitostearate, hydrogenated castor oil, hydrogenated vegetable oil, mineral oil, polyethylene glycol, sodium benzoate, sodium lauryl sulfate, sodium stearyl fumarate, stearic acid, talc and zinc stearate.
?
Flavouring agents and flavour enhancers make the dosage form more palatable to the patient. Common flavouring agents and flavour enhancers for pharmaceutical products that may be included in the composition of the present invention include maltol, vanillin, ethyl vanillin, menthol, citric acid, fumaric acid, ethyl maltol, and tartaric acid. Solid compositions may also be dyed using any pharmaceutically acceptable colorant to improve their appearance and/or facilitate patient identification.
The details of the present invention are provided in the Examples given below which are provided for illustrative purposes only and are not intended to limit the scope of the invention in any way. EXPERIMENTAL SECTION:
Example-1
4-Methyl-N-[4-[(4-methylpiperazin-l-yl) methyl]-3-(trifluoromethyl) phenyI]-3-[2- (7-oxo-8H-l, 8-naphthyridin-3-yI) ethynyl]benzamide (Development code: NRC21T)
Figure imgf000016_0001
To a solution of 2-amino-5-bromo pyridine in DMF, trifluoro acetic acid (1.1 equiv) was added at room temperature, followed by addition of N-Iodo succinimide (1.1 equiv) and the reaction mixture was heated at 50°C for 180min. After completion of the reaction, reaction mass was cooled to room temperature and the product was precipitated by adding the reaction mixture to water. After neutralization with sodium thiosulfate and IN NaOH the title compound was collected by filtration as a brown solid with 90% yield.
1.2 (E)-tert-butyl 3-(2-amino-5-bromopyridin-3-yl) acrylate
Figure imgf000017_0001
PropionMe, P(0-toI)3, 90°C,
Pd(OAC)2
To a 2-amino-5-bromo-3-iodo pyridine (1 equiv), tert-butyl acrylate (2 equiv), and isopropanol (2 equivalents), Ethyl nitrite (3 equiv) was added propionitrile (10 equiv) and then DMF (10 equiv). The solution was de-oxygenated with nitrogen gas for 15 minutes. The mixture was treated with Pd(OAc)2 (O.lequiv) and P(0-tol)3 (0.2 equiv) then heated to 90°C for 16h then filtered through a pad of silica gel. The filtrate was concentrated, diluted with water, extracted with ethyl acetate and the organic layer was concentrated under vacuum below 60°C. The compound was collected by filtration using hexane as solvent (70% yield). ESI MS m/z 299 (100%).
1.3 6-Bromo-lH-l, 8-naphthyridin-2-one
Figure imgf000017_0002
reflux
To a solution of (E)-tert-butyl 3-(2-amino-5-bromopyridin-3-yl) acrylate (1 equiv) in anhydrous methanol was treated with sodium methoxide (4.9M, 5 equiv) under nitrogen gas atmosphere. The solution was heated at reflux temperature for 3h then cooled to room temperature. The mixture was cooled in an ice water bath and treated with water under rapid stirring to give a precipitate. The solid was filtered and washed with water. Dried under reduced pressure to give an off-white solid (80% yield). ESI MS m/z 225 (100%).
1.4 6-(2-Trimethylsilylethynyl)-lH-l, 8-naphthyridin-2-one
Figure imgf000018_0001
PropionMe, 90°C
A mixture of 6-bromo-lH-l, 8-naphthyridin-2-one (1 equiv), Pd(PPh3)2C12 (0.1 equiv), Cul (0.15 equiv), (i-Pr)2EtN (4 equiv) in propionitrile was deoxygenated with nitrogen gas for 15 minutes. Then trimethyl silyl acetylene (2 equiv) was added and heated to
90°C for lOh. After reaction mass was filtered through a pad of silica gel at room temperature. The filtrate was concentrated and the compound was collected by filtration using propionitrile solvent at 0-5°C (60% yield).
1.5 6-Ethynyl-lH-l, 8-naphthyridin-2-one
Figure imgf000018_0002
To a solution of 6-(2-trimethylsilylethynyl)-lH-l, 8-naphthyridin-2-one (1 equiv) in THF (lOtimes) was slowly added 1M TBAF solution in THF (1.1 equiv) at room temperature under nitrogen gas atmosphere for 15minutes and stirred for 15 minutes. Product formation was observed by quenching the reaction mass in to the water. Product was collected by filtration at 0-5°C. For further purification, recrystallization was done in acetone solvent (80% yield).
1.6 4-Nitro-2-(trifIuoromethyl) benzoic acid (SCI)
Figure imgf000019_0001
Under Nitrogen gas atmosphere, a mechanically stirred mixture of 2-trifluoromethyl benzoic acid (1 equiv) and Conc.H2S04 (22 equiv) was cooled in an ice bath to 0-5°C. Then fuming nitric acid (9.8 equiv) was added drop wise at 0-5°C for 60min. The ice bath was removed and stirring continued for 120 min at room temperature. After completion of reaction the reaction mixture was poured into ice water, stirred for 60min at room temperature. Filtered the suspension, washed with chilled water and obtained the crude title compound. To remove the regio isomer the crude product was crystallized from water (45% yield). M.P: 137-142°C
1.7 (4-Methylpiperazin-l-yI)-[4-nitro-2-(trifluoromethyI) phenyl] methanone (SC2)
Figure imgf000019_0002
To an ice-cooled solution of 4-nitro-2-(trifluoromethyl) benzoic acid (1 equiv), CH2C12 (15times) and DMF (0.5 equiv) under nitrogen atmosphere, oxalylchloride (2 equiv) was added drop wise. After 4hrs, the resulting solution was concentrated in vacuum. The residue was dissolved in CH2C12 and added drop wise to an ice cooled solution of N- methyl piperazine (2.1 equiv) in CH2C12. After stirring for 3h, the mixture was diluted with CH2C12 and washed with water, 3 portions of 10% solution of Na2C03, water and brine. The organic phase was concentrated to get the title compound as an oil (96% yield).
1.8 [4-Amino-2-(trifIuoromethyI) phenyl]-(4-methyIpiperazin-l-yl)methanone (SC3)
Figure imgf000020_0001
To a solution of (4-methylpiperazin-l-yl)-[4-nitro-2-(trifluoromethyl) phenyl]methanone (1 equiv) in methanol (3 times) was added 10%Pd/C under nitrogen atmosphere. Then slowly added 66% aq. ammonium formate solution (5 equiv) at room temperature (Exothermic). After stirring for 120min, filtered through a pad of silica gel. The filtrate was concentrated and diluted with water, extracted with CH2C12 and washed with water. After CH2C12 concentration compound was collected by filtration using hexane solvent (90% yield).
1.9 4-[(4-Methylpiperazin-l-yl) methyl] -3-(trifluoromethyl) aniline (SC4)
Figure imgf000020_0002
To a solution of vitride (3 equiv) in toluene (6times) under nitrogen atmosphere was slowly added lot wise [4-amino-2-(trifluoromethyl) phenyl] -(4-methylpiperazin-l- yl)methanone (1 equiv) at room temperature during 2h (exothermic). After stirring for 4h at 65°C, slowly added 8% aq. NaOH solution (12 equiv) during lh at room temperature. Resulting solution was stirred for 30minutes, extracted with toluene, combined toluene layers were dried over Na2S04 and concentrated. Crystallization form boiling hexane afforded the title compound (50% yield). 1.10 3-iodo-4-methyl-N-[4-[(4-methylpiperazin-l-yl) methyl]-3-(trifluoromethyl) phenyl] benzamide (SC5)
Figure imgf000021_0001
To a solution of 4-[(4-methylpiperazin-l-yl)methyl]-3-(trifluoromethyl)aniline (0.7 equiv) in THF (4times) under nitrogen atmosphere was added 3-Iodo-4-methyl benzoyl chloride (1 equiv, prepared from the reaction of 3-iodo-4-methylbenzoic acid and SOCl2) in THF at room temperature for 30minutes followed by drop wise addition of (i-Pr)2EtN (2 equiv) and 4-DMAP (0.2 equiv). After stirring at ambient temperature for 120min, the reaction mixture was quenched with water, extracted with ethyl acetate. After drying over Na2S04, concentrated the ethyl acetate layer to provide the crude product. Acetone was added to this crude product and was converted to HC1 salt using IPA-HC1 (85% yield).
1.11 4 Methyl-N-[4-[(4-methylpiperazin-l-yl) methyl] -3-(trifluoromethyl) phenyl]-3- [2-(7-oxo-8H-l, 8-naphthyridin-3-yI) ethynyl] benzamide (Development code: NRC21T)
Figure imgf000021_0002
A mixture of 3-Iodo-4-methyl-N-[4-[(4-methylpiperazin-l-yl)methyl]-3-(trifluoromethyl) phenyl]benzamide (0.8 equiv, prepared as per 1, SC5), 6-ethynyl-lH- l,8-naphthyridin-2- one (1 equiv, prepared as per 1.5), Pd(PPh3)2Cl2 (0.1 equiv), Cul (0.15 equiv), (i-Pr)2EtN (4 equiv), in DMF under nitrogen atmosphere was deoxygenated with Nitrogen gas for 30minutes. Reaction mass was heated to 80°C for 5h. Filtered through a pad of silica gel at room temperature and filtrate was concentrated, diluted with water and methanol (1 :2 mixture). The compound was collected by filtration and dried. The XRD is depicted in Figure-1. The compound was subjected to salt formation with cone. HCl in methanol medium to yield titled compound as hydrated dihydrochloride salt (70% yield). The XRD is depicted in Figure-2.
1HNMR (400MHz,DMSOD6) 512.430 (s, 1H), 10.678 (s, 1H), 8.742 (s,lH), 8.397 (s, 1H), 8.288 (s, 1H), 8.156 (d, 2H), 7.958 (d, 2H), 7.893 (s, 1H), 7.537 (d, 1H), 6.645 (d, 1H), 3.946 (bs, 2H), 3.169-3.474 (bs, 8H), 2.794 (s, 3H), 2.586 (s, 3H). ESI MS m/z - 560.3(M+H) +.
Example-2: 4-MethyI-N-[4-[(4-methyIpiperazin-l-yl) methyl]-3-(trifluoromethyl) phenyl]-3-[(E)- 2-(7-oxo-8H-l, 8-naphthyridin-3-yl) vinyl] benzamide (Development code: NRC20T)
Figure imgf000023_0001
2.1 6-[2-Trimethylsilylvinyl]-lH-l, 8-naphthyridin-2-one
Figure imgf000023_0002
A mixture of 6-bromo-lH-l, 8-naphthyridin-2-one (1 equiv, which was prepared as per prepared as per- 1.3), Pd (PPh3)2Cl2 (0.1 equiv), Cul (0.15 equiv), (i-Pr)2EtN (4 equiv) in DMF (15 times) was deoxygenated with nitrogen for 15 minutes. Then vinyl trimethyl silane (2 equiv) was added and heated to 125°C for lOh. The reaction was monitored by thin layer chromatography. Filtered through a pad of silica gel at room temperature. The filtrate was concentrated and the residue was purified by silica gel chromatography (eluted with 20 to 30% ethyl acetate/hexane) to provide the title compound as a solid (30% yield). ESI MS m/z -245.22(M+H)+.
2.2 4-Methyl-N-[4-[(4-methylpiperazin-l-yl) methyl]-3-(trifluoromethyI) phenyl]-3- [(E)-2-(7-oxo-8H-l, 8-naphthyridin-3-yl) vinyl] benzamide (Development code: NRC20T)
Figure imgf000024_0001
A mixture of 3-iodo-4-methyl-N-[4-[(4-methylpiperazin-l-yl)methyl]-3-(trifluoromethyl) phenyl]benzamide (1 equiv, prepared as per example SC5), 6-[2-trimethylsilylvinyl]-lH- l,8-naphthyridin-2-one (1 equiv,), Pd(OAC)2 (0.1 equiv), P(0-tol)3 (0.2 equiv), (i- Pr)2EtN (4 equiv), in DMF was deoxygenated with Nitrogen gas for 30minutes. Then heated to 90°C for 7h. Filtered through a pad of silica gel at room temperature, filtrate was concentrated, and the residue was purified by silica gel chromatography (eluted with 10% methanol/methylene chloride) to provide the title compound as a solid. The compound was subjected to hydrochloride salt formation using methanol and IPA-HC1.
'HNMR (400MHz,DMSOD6) 512.283 (s, 1H), 10.772-10.802 (s, 1H), 8.822 (s,lH), 8.521 (s, 1H), 8.367 (s, 2H), 8.233 (s, 1H), 8.042 (s, 1H), 7.950-7.974 (d, 1H), 7.835- 7.854 (d, 1H), 7.577-7.618 (d, 1H), 7.409-7.465 (d, 2H), 6.611-6.634 (d, 1H), 4.155 (bs, 2H), 3.075-3.550 (bs, 8H), 2.806 (s, 3H), 2.529 (s, 3H). ESI MS m/z -562.2(M+H)+.
ExampIe-3:
N-[4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyl] phenyl]-4-[(4-methyl piperazin-l-yl) methyI]-3-(trifIuoromethyl)benzamide (Development code: NRC19T)
Figure imgf000025_0001
3.1 Methyl 4-methyl-3-(trifluoromethyl) benzoate
Figure imgf000025_0002
H3
To a mixture of 4-methyl-3-(trifluoromethyl) benzoic acid (1 equiv), K2C03 (1.5 equiv) in acetone (15 times) was added Mel (1.5 equiv) at room temperature. Stirring was continued for 24h. Reaction was monitored by thin layer chromatography. The salts were filtered and resulting filtrate was concentrated, diluted with water, extracted with ethyl acetate. Concentration of organic layer afforded the pale yellow oil (95% yield). ESI MS m/z -219 (M+IT l)+.
3.2 Methyl 4-(bromomethyI)-3-(trifIuoromethyl) benzoate
Figure imgf000025_0003
24
Ί To a solution of methyl-4-methyl-3-(trifluoromethyl) benzoate (1 equiv) in chloroform was added N-bromosuccinimide (1.1 equiv) and benzoyl peroxide (0.01 equiv). The reaction mixture was heated at reflux overnight (18h). It was then cooled to room temperature, washed with water, dried over Na2S0 and concentrated. It was purified by silica gel chromatography (eluted with 1% ethyl acetate/hexane) to provide the title compound (65% yield). ESI MS m/z -297.
3.3 Methyl 4-[(4-methylpiperazin-l-yl) methyl] -3-(trifluoromethyl) benzoate
Figure imgf000026_0001
To a solution of methyl 4-(bromomethyl)-3-(trifluoromethyl) benzoate (1 equiv) in chloroform (6times) was added to N-methylpiperazine (3 equiv) at room temperature. The resulting solution was stirred for 3hrs, the reaction mass was washed with water and concentrated with chloroform to get the product. (95% yield). ESI MS m/z - 317 (M+H)
3.4 4-[(4-Methylpiperazin-l-yl) methyl] -3-(trifluoromethyl) benzoic acid
Figure imgf000026_0002
To a solution of methyl 4-[(4-methylpiperazin-l-yl) methyl]-3-(trifluoromethyl) benzoate (1 equiv) in ethanol (lOtimes) was added 2N NaOH solution (2 equiv) at room temperature. The resulting solution was stirred for 3h. The reaction mixture was concentrated and acidified with aqueous 2N HCl until a white solid formed. The compound was collected by filtration at 0-5°C. (85% yield). ESI MS m/z -303.3 (M+H)+.
3.5 N-(3-iodo-4-methyl-phenyI)-4-[(4-methylpiperazin-l-yl) methyl] -3-(trifluoro methyl) benzamide
Figure imgf000027_0001
To a solution of 3-iodo-4-methyl aniline (0.7 equiv) in THF (lOtimes) under nitrogen gas atmosphere was added 4-[(4-methylpiperazin-l-yl)methyl]-3-(trifluoromethyl)benzoyl chloride (1 equiv, prepared from the reaction of 4-[(4-methylpiperazin-l-yl)methyl]-3- (trifluoromethyl)benzoic acid and SOCl2) in THF (4times) at room temperature for 30minutes followed by drop wise addition of (i-Pr)2EtN (4 equiv) and 4-DMAP (0.2 equiv). After stirring at ambient temperature for 120min, the reaction mixture was quenched with water and extracted with ethyl acetate. The ethyl acetate layer was concentrated to provide the crude product. Acetone was added to this crude product and isolated as HCl salt by employing IPA-HC1 (40% yield). ESI MS m/z -518 (M+H) +.
3.6 N-[4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyl] phenyl]-4-[(4- methyl piperazin-l-yl) methyl]-3-(trifluoromethyl)benzamide (Development code: NRC19T)
Figure imgf000028_0001
A mixture of N-(3-iodo-4-methyl-phenyl)-4-[(4-methylpiperazin-l-yl)methyl]-3- (trifluoro methyl)benzamide (0.8 equiv, prepared as per 3.5), 6-ethynyl-lH-l,8- naphthyridin-2-one (1 equiv, prepared as per 1.5), Pd(PPh3)2C-2 (0.1 equiv), Cul (0.15 equiv), (i-Pr^EtN (4 equiv), in DMF (20times) was deoxygenated with nitrogen gas for 30minutes. Heated to 80°C for 5h and filtered through a pad of silica gel at room temperature. The filtrate was concentrated, diluted with water and stirred for lh. The compound was collected by filtration. Methanol was added to the above wet compound and the product was isolated as the HC1 salt by employing IPA-HC1 (40% yield).
1HNMR(400MHz,DMSOD6) (512.403 (s, 1H), 10.626 (s, 1H), 8.716-8.721 (s,lH), 8.384- 8.390 (s, 1H), 8.338-8.348 (d, 2H), 8.143 (s, 1H), 8.063-8.069 (s, 1H), 7.945-7.969 (d, 1H), 7.706-7.732 (d, 1H), 7.342-7.364 (d, 1H), 6.636-6.659 (d, IH), 4.114 (bs, 2H), 2.894-3.502 (bs, 8H), 2.790 (s, 3H), 2.482 (s, 3H). ESI MS m/z -560.28 (M+H) +. Example-4:
4-Methyl-N-[3-(4-methyIimidazol-l-yI)-5-(trifluoromethyI) phenyl] -3- [2-(7-oxo-8H- 1, 8-naphthyridin-3-yl)ethynyl]benzamide Development code: NRC18T)
Figure imgf000029_0001
4.1 3-Iodo-4-methyl-N-[3-(4-methylimidazoI-l-yl)-5- trifluoromethyl) phenyl] benzamide
Figure imgf000029_0002
To a solution of 3-(4-methylimidazol-l-yl)-5-(trifluoromethyl)aniline (0.7 equiv, prepared according to literature known methods) in THF under nitrogen atmosphere was added 3-Iodo-4-methyl benzoyl chloride (1 equiv, prepared from the reaction of 3-iodo-4- methylbenzoic acid and SOCl2) in THF at room temperature during 30minutes followed by drop wise addition of (i-Pr)2EtN (2 equiv) and 4-DMAP (0.2 equiv). After stirring at ambient temperature over 3h, the reaction mixture was quenched with water. The resulting mixture was stirred at 0-5°C for 60min. Compound was collected by filtration. This compound was further purified by silica gel chromatography (eluted with 2% methanol/ chloroform) to provide the title compound (85% yield). ESI MS m/z - 486.2 (M+H)+.
4.2 4-Methyl-N-[3-(4-methylimidazol-l-yI)-5-(trifluoromethyl) phenyl]-3-[2-(7-oxo- 8H-1, 8-naphth ridin-3-yl)ethynyl]benzamide (Development code: NRC18T)
Figure imgf000030_0001
A mixture of 3-iodo-4-methyl-N-[3-(4-methylimidazol-l-yl)-5-(trifluoromethyl)phenyl] benzamide (0.8 equiv, prepared as per 4.1), 6-ethynyl-lH-l,8-naphthyridin-2-one (1 equiv, prepared as per 1.5), Pd(PPh3)2Cl2 (0.1 equiv), Cul (0.15 equiv), (i-Pr)2EtN (4 equiv), in DMF (20 times) was deoxygenated with nitrogen gas for 30minutes. Heated to 80°C for 5h, filtered through a pad of silica gel at room temperature. The filtrate was concentrated, diluted with water, stirred for lh and filtered. Acetone was added to above wet compound and the product was isolated as the HC1 salt employing IPA-HC1 (20% yield).
1HNMR(400MHz,DMSOD6) 512.440 (s, IH), 11.034 (s, IH), 9.668 (s,lH), 8.736-8.741 (s,lH), 8.634 (s, IH), 8.392-8.397 (s, IH), 8.320 (s, IH), 8.252 (s,lH), 7.950-8.051 (m, 4H), 7.557-7.577 (d, IH), 6.647-6.671 (d, IH), 2.591 (s, 3H), 2.377 (s, 3H). ESI MS m/z -528.27 (M+H)+.
ExampIe-5:
4-Methyl-N- [3- [(4-methylpiperazin-l -yl) methyl] -5-(trifluoromethyl) phenyl] -3- [2- (7-OXO-8H-1, 8-naphthyridin-3-yl)ethynyl]benzamide (Development code: NRC17T)
Figure imgf000031_0001
5.1 3-Iodo-4-methyl-N-[3-[(4-methyIpiperazin-l-yl) methyl]-5-(trifluoromethyI) phenyl] benzamide
Figure imgf000031_0002
To a solution of 3-[(4-methylpiperazin-l-yl)methyl]-5-(trifluoromethyl)aniline (0.7 equiv, prepared according to literature methods) in THF under nitrogen atmosphere was added 3-Iodo-4-methyl benzoyl chloride (1 equiv, prepared from the reaction of 3-iodo-4- methylbenzoic acid and SOCl2) in THF at room temperature during 30minutes followed by drop wise addition of (i-Pr)2EtN (2 equiv) and 4-DMAP (0.2 equiv). After stirring at ambient temperature over 3h, the reaction mixture was quenched with water. The resulting mixture was extracted with ethyl acetate and formed product was isolated as HC1 salt in acetone. ESI MS m/z - (M+l) +. 5.2 4-MethyI-N-[3-[(4-methylpiperazin-l-yl) methyI]-5-(trifluoromethyl) phenyl]-3- [2-(7-oxo-8H-l, 8-naphthyridin-3-yI)ethynyl]benzamide (Development code: NRC17T
Figure imgf000032_0001
A mixture of 3-iodo-4-methyl-N-[3-[(4-methylpiperazin-l-yl)methyl]-5-(trifluoromethyl) phenyljbenzamide (0.8 equiv, prepared as per 5.1), 6-ethynyl-lH-l,8-naphthyridin-2-one (1 equiv, prepared as per 1.5), Pd(PPh3)2Cl2 (0.1 equiv), Cul (0.15 equiv), (i-Pr)2EtN (4 equiv), in DMF (20 times) was deoxygenated with Nitrogen gas for 30minutes. Heated to 80°C for 5h, filtered through a pad of silica gel at room temperature. The filtrate was concentrated, diluted with water, stirred for lh and filtered. Methanol was added and the product was isolated as the HC1 salt employing IPA-HC1 (25% yield).
1HNMR(400MHz,DMSOD6) 512.432 (s, 1H), 10.796 (s, lH), 8.741 (s,lH), 8.399 (s,lH), 8.322 (s, 1H), 8.222 (s, 2H), 7.964 (d, 2H), 7.814 (s,lH), 7.542- (d, 1H), 6.647 (d, 1H), 4.363 (s, 2H), 3.394 (bs, 8H), 2.818 (s, 3H), 2.589 (s, 3H). ESI MS m/z - (M+l)+.
Example-6: N-[3, 5-Bis (trifluoromethyl) phenyI]-4-methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3- yl)ethynyl]benzamide benzamide (Development code: NRC16T)
Figure imgf000033_0001
N-[3, 5-Bis (trifluoromethyl) phenyl]-3-iodo-4-methyl-benzamide
Figure imgf000033_0002
To a solution of 3,5-bis trifluoro methyl aniline (0.7 equiv) in THF (6times) under Nitrogen gas atmosphere was added 3-Iodo-4-meyhyl benzoyl chloride (1 equiv, prepared from the reaction of 3-iodo-4-methylbenzoic acid and SOCl2) in THF at room temperature during 30minutes followed by drop wise addition of (i-Pr)2EtN (2 equiv) and 4-DMAP (0.2 equiv). After stirring at ambient temperature over 3h, the reaction mixture was quenched with water. The resulting mixture was extracted with ethyl acetate and concentrated. Acetone was added to the resulting residue and formed HC1 salt using IPA- HC1 (85% yield). ESI MS m/z - 474 (M+H) +.
6.2 N-[3, 5-Bis (trifluoromethyl) phenyl]-4-methyl-3-[2-(7-oxo-8H-l, 8- naphthyridin-3-yl) ethynyl]benzamide (Development code: NRC16T)
Figure imgf000034_0001
A mixture of N-[3,5-bis(trifluoromethyl)phenyl]-3-iodo-4-methylbenzamide (0.8 equiv, prepared as per 6.1), 6-ethynyl-lH-l,8-naphthyridin-2-one (1 equiv, prepared as per 1.5), Pd(PPh3)2Cl2 (0.1 equiv), Cul (0.15 equiv), (i-Pr)2EtN (4 equiv), in DMF was deoxygenated with nitrogen gas for 30minutes. Heated to 80°C for 9h, filtered through a pad of silica gel at room temperature. The filtrate was concentrated, diluted with water, stirred for lh and filtered. Methanol was added and the compound was filtered as HCl salt by treatment with IP A-HC1 (60% yield).
1HNMR(400MHz,DMSOD6) 512.429 (s, 1H), 10.874 (s, 1H), 8.746 (s,lH), 8.545 (s,2H), 8.402 (s, 1H), 8.223 (s, 1H), 7.956-7.979 (d, 2H), 7.839 (s, 1H), 7.563-7.582 (d, 1H), 6.646-6.668 (d, 1H), 2.594 (s, 3H). ESI MS m/z -516.13 (M+H)+. Example-7:
N-[4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyl] phenyl]-3, 5-bis (trifluoro methyl)benzamide benzamide (Development code: NRC15T)
Figure imgf000035_0001
Figure imgf000035_0002
To a solution of 3-Iodo-4- methyl aniline (0.7 equiv) in THF (6times) under nitrogen atmosphere was added 3,5-bis trifluoromethylbenzoyl chloride (1 equiv, prepared from the reaction of 3,5-bis trifluoromethylbenzoic acid and SOCl2) in THF at room temperature for 30minutes followed by drop wise addition of (i-Pr)2EtN (4 equiv) and 4- DMAP (0.2 equiv). After stirring at ambient temperature for 3h, the reaction mixture was quenched with water. The resulting mixture was extracted with ethyl acetate, concentrated and the compound was collected by adding hexane and filtration (44% yield).
7.2 N-[4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl)ethynyl] phenyl] -3,5-Bis (trifluoro methyl)benzamide (Development code: NRC15T)
Figure imgf000036_0001
A mixture of N-(3-iodo-4-methyl-phenyl)-3,5-bis(trifluoromethyl)benzamide (0.8 equiv, prepared as per 7.1), 6-ethynyl-lH-l,8-naphthyridin-2-one (1 equiv, prepared as per example 1.5), Pd(PPh3)2Cl2 (0.1 equiv), Cul (0.15 equiv), (i-Pr)2EtN (4 equiv), in DMF was deoxygenated with nitrogen gas for 30minutes. Heated to 65°C for 3h and filtered through a pad of silica gel at room temperature. The filtrate was diluted with water and extracted with ethyl acetate. Ethyl acetate layer was concentrated and formed HCl salt in acetone solvent. 1HNMR (400MHz, DMSO D6) 512.403 (s, 1H), 10.743 (s, 1H), 8.716 (s, 1H), 8.636 (s, 2H), 8.382 (s, 2H), 8.039 (s, 1H), 7.942 (d, 1H), 7.701 (d, 1H), 7.365 (d, 1H), 6.635 (d, 1H), 2.490 (s, 3H). ESI MS mix - (M+H) +.
ExampIe-8:
4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyI]-N-[3-(trifIuoromethyl) phenyl] benzamide benzamide (Development code: NRC14T)
Figure imgf000037_0001
8.1 3-Iodo-4-methyl-N-[3-(trifluoromethyl) phenyl] benzamide
Figure imgf000037_0002
To a solution of 3-trifluoromethyl aniline (0.7 equiv) in THF under nitrogen atmosphere was added 3-Iodo-4-meyhyl benzoyl chloride (1 equiv, prepared from the reaction of 3- iodo-4-methylbenzoic acid and SOCl2) in THF at room temperature during 30minutes followed by drop wise addition of (i-Pr)2EtN (2 equiv) and 4-DMAP (0.2 equiv). After stirring to ambient temperature over 3h, the reaction mixture was quenched in to water. The resulting mixture was extracted with ethyl acetate and concentrated. The compound was collected by filtration using hexane solvent (49.2% yield). ESI MS m/z - (M+l) +.
8.2 4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyl]-N-[3-(trifluoromethyl) phenyl] benzamide (Development code: NRC14T)
Figure imgf000038_0001
A mixture of 3-iodo-4-methyl-N-[3-(trifluoromethyl)phenyl]benzamide (0.8 equiv, prepared as per 8.1), 6-ethynyl-lH-l,8-naphthyridin-2-one (1 equiv, prepared as per 1.5), Pd(PPh3)2Cl2 (0.1 equiv), Cul (0.15 equiv), (i-Pr)2EtN (4 equiv), in DMF (20 times) was deoxygenated with nitrogen gas for 30minutes. Heated to 65°C for 3h, filtered through a pad of silica gel at room temperature and filtrate was diluted with water. The compound was collected by filtration and HC1 salt formation. 1HNMR(400MHz,DMSOD6) 512.426 (s, 1H), 10.595 (s, 1H), 8.742 (s,lH), 8.395 (s,lH), 8.260 (s, 1H), 8.192 (s, 1H), 8.079 (d, 1H), 7.935 (d, 2H), 7.596 (t, 1H), 7.533 (d, 1H), 7.459 (d, 1H), 6.644 (d, 1H), 2.587 (s, 3H). ESI MS m/z - (M+H)+.
Example-9:
N-[4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl)ethynyl]pbj (trifluoromethyl) benzamide benzamide (Development code: NRC13T)
Figure imgf000038_0002
9.1 N-(3-iodQ-4-methyl-phenyl)-3-(trifluoromethyl)benzamide
Figure imgf000039_0001
To a solution of 3-iodo-4- methyl aniline (0.7 equiv) in THF under nitrogen atmosphere was added 3-trifluoro methyl benzoyl chloride (1 equiv, prepared from the reaction of 3- trifluoromethyl benzoic acid and SOCl2) in THF at room temperature during 30minutes followed by drop wise addition of (i-Pr)2EtN (4 equiv) and 4-DMAP (0.2 equiv). After stirring at ambient temperature for 3h, the reaction mixture was quenched with water. The resulting mixture was extracted with ethyl acetate, concentrated and the compound was isolated by adding n-hexane.
9.2 N-[4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl)ethynyl] phenyl] -3-(trifluoro methyl) benzamide (Development code: NRC13T)
Figure imgf000039_0002
A mixture of N-(3-iodo-4-methyl-phenyl)-3-(trifluoromethyl)benzamide (0.8 equiv, prepared as per 9.1), 6-ethynyl-lH-l,8-naphthyridin-2-one (1 equiv, prepared as per 1.5), Pd(PPh3)2Ci2 (0.1 equiv), Cul (0.15 equiv), (i-Pr)2EtN (4 equiv), in DMF was deoxygenated with nitrogen gas for 30minutes. Heated to 65°C for 3h, filtered through a pad of silica gel at room temperature and the filtrate was diluted with water. The resulting mixture was stirred for lh and the compound was collected by filtration and HCl salt using IP A-HC1.
1HNMR(400MHz>DMSOD6) 512.400 (s, 1H), 10.523 (s, 1H), 8.718 (s,lH), 8.381 (s,lH), 8.317 (s, 1H), 8.272 (d, 1H), 8.055 (s, 1H), 7.940 (d, 2H), 7.786 (t, 1H), 7.704 (d, 1H), 7.343 (d,lH), 6.635 (d, 1H), 2.484 (s, 3H). ESI MS m/z - (M+H) +.
Example-10: In vitro efficacy evaluation:
All the experimental compounds were evaluated for in vitro efficacy employing standard cell lines used for the screening of anti-cancer compounds.
Ponatinib was used as reference drug candidate for a comparative efficacy and safety evaluation. A laboratory sample of the drug substance Ponatinib was synthesized in house employing the procedure disclosed in US8114874.
In Vitro studies of NRC-21T on BCR-abl positive cell line K562 and mutant cell lines Baf3/T315i, M351T, E255K and WT.
The experimental compounds and the standard reference drug (Ponatinib) were dissolved in cell culture medium and DMSO at a concentration of lOmM for in vitro studies. The stock solution was further diluted with the same cell culture medium and used in concentrations of 0.1 nm to ΙΟμηι. Cell proliferation by MTT assay was done as follows. 1000 to 10,000 cells were seeded per well in 96-well plate and different concentrations of Pon-21T ranging from ΙΟμπι to 0.1 nM were added in triplicates. After incubating the cells with NRC-21T for the required time period 24-72hrs, 15μ1 of 5mg/ml MTT was added and incubated for additional 4 hours at 37°C and 5% C02. After 4 hrs, formazan crystals were dissolved in solubilizing buffer overnight at 37°C. Absorbance was measured on Elisa reader at dual wavelength of 570-630-nm. By MTT assay the IC50 values of the NRC21T were computed. IC 50 values obtained by MTT assay were tabulated in Table- 1A & Table- IB and cells of control, Ponatinib and NRC21T was photo graphed under inverted microscope and presented. (Figure-3)
Table 1A: IC50 values obtained from MTT assay
Figure imgf000041_0001
Table-IB: IC50 values obtained from MTT assay
Figure imgf000041_0002
Example-11:
Matrigel invasion assay:
The in- vitro invasiveness of BaF3/T315i mutant Leukemia cancer cells in the presence of specified concentration of NRC21T were assessed.T315i cells (3X105) were suspended in 300μ1 of serum- free medium and placed in the upper compartment of transwell chambers pre-coated with the matrigel (Millipore, Catalog No. ECM550, USA). The lower compartment of the chamber was filled with 500 μΐ serum-medium (10% FBS) and the cells were allowed to migrate for 72hrs. After incubation, the cells were fixed and stained with the dye provided along with the kit and quantified using Elisa plate reader at 560nm. In Vitro matrigel assay of T315i cells in the presence of specified concentration of NRC-21T were represented in Table 2 and Figure-4.
Table 2: Anti-Invasive property of NRC-21T on T315i cell line
Figure imgf000042_0001
Example-12
Determination of MTD of NRC21T [The MTD study was carried out as per OECD guidelines 420]
The study was carried out using 5 (2 Male + 3 Female) Swiss Albino Mice weighing 18- 30 grams. All the animals were fasted for 3 hours prior to the oral administration of the drug. The sample was administered immediately to all the animals according to their body weight. After administration of the experimental drug all the animals were observed for ½hr, lhr, 2hr, 4 hours and mortality was observed for 14 days. At the end of 14 days, all the surviving animals were autopsied and stomach was cut opened and observed for absorption of the drug through the GIT.
> NRC21T: MTD > 2000 mg.kg, p.o (Single dose 14 days observation)
> Ponatinib: MTD = 50 mg/kg, p.o (Single dose 14 days observation)
Conclusion: Since the MTD of NRC21T is more than 2000 mg/kg, p.o according to ICH guidelines it is inferred that NRC21T is a safer experimental drug than Ponatinib. Also NRC21T has comparable IC50 values with respect to Ponatinib. Thus, the experimental drug of this invention, NRC21T is established as superior candidate in terms of safety and efficacy.
Example-12 Establishment of Antagonism of NRC21T in T315I induced tumour in Nude Mice: [Clackson etal. 2009, cancer cells November6; 16(5): 401-412]:
The study was carried out with 18 Nude Mice (9 Male + 9 Female). Weighing of Nude mice was taken initially before inoculation of cell line and made into groups as follows:
Group - 1: Positive control (3 Male + 3 Female)
Group - II: NRC21T (3 Male + 3 Female)
Group - III: Ponatinib (3 Male + 3 Female)
The cell line was inoculated into Nude Mice subcutaneously to the right hind limb flank at a strength of 1 X 106 cells/0.2 ml. Animals were observed daily for the appearance of tumour. The tumour volume was measured using the formula ½ 1 X w2 (1 = length of tumour & w = width of tumour). When the mean tumour volume was recorded above 400 mm , the treatment with the above drugs was started. The above drugs were administered orally daily for 30 days. Weights of Nude Mice were taken daily before dosing and tumour measurements were done on alternative days using digital Vernier caliper. Surviving animals were sacrificed after the dosing was complete for 30 days and the organs (tumour with skin and spleen) were collected.
The study was depicted in Figure-5, Firgure-5A and Figure-5B.
Figure imgf000044_0001
Example-13
Establishment of Survival time of NRC21T in SCID Mice [Clackson etal. 2009, cancer cells November 6; 16(5): 401-412]
5
The study was carried out using 30 SCID Mice (15 Male + 15 Female). All the SCID Mice were injected with T315I cell Line intravenously to the tail vein at a strength of 1 X 106 cells/0.1 ml. grouping as follows: Group - 1 : Positive Control (5 Male + 5 Female) (Vehicle treated)
Group - II : NRC21 T (5 Male + 5 Female) (200 mg/kg, p.o)
Group - III : Ponatinib (5 Male + 5 Female) (10 mg/kg, p.o)
After 72 hours of injection administration of drugs to the respective groups were started. All the animals in the groups were administered with the respective drugs for 30 days. In case of mortality during the study spleen and liver were collected and sent for histopathology. Animals in the moribund state were sacrificed and spleen and liver were collected and sent for histology. The study results depicted in Figure-6A and Figure- 6B.
Results:
Survival Time
Figure imgf000045_0001

Claims

We claim:
1. A compound of formula I:
Figure imgf000046_0001
Formula (I)
Wherein:
LI is Linker selected from unsaturated carbon bond; preferably carbon-carbon triple bond or carbon-carbon double bond;
L2 is a linker selected from NHC(O)-, C(0)NH-;
Ring A includes, but are not limited to substituted 5 or 6 membered aryl or heteroaryl ring;
or N-oxide or pharmaceutically acceptable salts thereof;
wherein Ring A is selected from the group consisting of;
Figure imgf000046_0002
2. The compound of formula is selected from the group consisting of: 4-Methyl-N- [4- [(4-methylpiperazin- 1 -yl) methyl] -3 -(trifluoromethyl) phenyl]-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl)ethynyl]benzamide or a pharmaceutically acceptable salt thereof,
4-Methyl-N- [4- [(4-methylpiperazin- 1 -yl) methyl] -3 -(trifluoromethyl) phenyl]-3-[(E)-2-(7-oxo-8H-l, 8-naphthyridin-3-yl) vinyl] benzamide or a pharmaceuticaly acceptable salts thereof,
N-[4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyl] phenyl]-4- [(4-methylpiperazin-l-yl) methyl] -3 -(trifluoromethyl)benzamide or a pharmaceuticaly acceptable salts thereof,
4-Methyl-N-[3-(4-methylimidazol-l-yl)-5-(trifluoromethyl) phenyl]-3-[2- (7-oxo-8H-l, 8-naphthyridin-3-yl)ethynyl]benzamide or a
pharmaceuticaly acceptable salts thereof,
4- Methyl-N-[3-[(4-methylpiperazin- 1 -yl) methyl]-5-(trifluoromethyl) phenyl]-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl)ethynyl]benzamide or a pharmaceuticaly acceptable salts thereof,
N-[3, 5-Bis (trifluoromethyl) phenyl]-4-methyl-3-[2-(7-oxo-8H-l, 8- naphthyridin-3-yl)ethynyl]benzamide benzamide or a pharmaceuticaly acceptable salts thereof,
N-[4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyl] phenyl]-3,
5- bis (trifluoromethyl)benzamide benzamide or a pharmaceuticaly acceptable salts thereof,
4-Methyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyl] -N- [3- (trifluoromethyl) phenyl] benzamide benzamide or a pharmaceuticaly acceptable salts thereof,
N-[4rMethyl-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl)ethynyl]phenyl]-3- (trifluoromethyl)benzamide benzamide or a pharmaceuticaly acceptable salts thereof.
3. The compound, 4-Methyl-N-[4-[(4-methylpiperazin-l-yl) methyl]-3- (trifluoromethyl) phenyl]-3-[2-(7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyljbenzamide or a pharmaceutically acceptable salt thereof.
4. A process for the synthesis of a compound of the formula I
Figure imgf000048_0001
Formula (I) or N-oxide or pharmaceutically acceptable salts thereof, wherein LI, L2 and ring A are as
defined in claim 1, characterized in that a compound of formula II
Figure imgf000048_0002
H
Ώ
wherein Ll is as defined for a compound of formula I, is reacted with an compound of formula III
Figure imgf000048_0003
wherein L2 and A is as defined for a compound of formula I.
5. A process for preparing 4-Methyl-N-[4-[(4-methylpiperazin-l-yl) methyl]-3- (trifluoromethyl) phenyl]-3-[2-(7-oxo-8H-l, 8-naphthyridin-3- yl)ethynyl]benzamide or a pharmaceutically acceptable salt thereof , 3-halo-4- methyl-N- [4- [(4-methylpiperazin- 1 -yl)methyl] -3 -
(trifluoromethyl)phenyl]benzamide is reacted with an 6-ethynyl-lH- 1,8- naphthyridin-2-one in presence of a suitable catalyst and solvent;
and optionally the obtained compound is converted into a pharmaceutically acceptable salt.
6. A pharmaceutical composition comprising the compound of formula I according to claim 1 or N-oxide or a pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier.
7. A pharmaceutical composition comprising the compound of formula according to claim 2 or a pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier.
8. A pharmaceutical composition comprising the 4-Methyl-N-[4-[(4- methylpiperazin-l-yl) methyl]-3-(trifluoromethyl) phenyl]-3-[2-(7-oxo-8H-l, 8- naphthyridin-3-yl) ethynyl] benzamide or a pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier.
9. A method for the treatment of leukemias which comprises administering a compound of formula I according to claim 1 or N-oxide or a pharmaceutically acceptable salt thereof.
10. A method for the treatment of leukemias which comprises administering a 4- Methyl-N- [4- [(4-methylpiperazin- 1-yl) methyl]-3-(trifluoromethyl) phenyl]-3-[2- (7-oxo-8H-l, 8-naphthyridin-3-yl) ethynyl] benzamide or a pharmaceutically acceptable salt thereof.
PCT/IN2014/000777 2014-06-06 2014-12-15 1h-1,8- naphthyridin-2ones as anti proliferative compounds WO2015186137A1 (en)

Priority Applications (17)

Application Number Priority Date Filing Date Title
KR1020177000113A KR20170016921A (en) 2014-06-06 2014-12-15 1h-1, 8-naphthyridin-2-ones as antiproliferative compounds
EA201692371A EA201692371A1 (en) 2014-06-06 2014-12-15 1H-1,8-NAFTYRIDIN-2-ONE AS ANTIPROLIFERATIVE COMPOUNDS
US15/316,832 US9765072B2 (en) 2014-06-06 2014-12-15 1H-1,8-naphthyridin-2ones as anti proliferative compounds
BR112016028674A BR112016028674A2 (en) 2014-06-06 2014-12-15 ? 1h-1,8-naphthyridin-2-ones as antiproliferative compounds?
AU2014396394A AU2014396394A1 (en) 2014-06-06 2014-12-15 1H-1,8- naphthyridin-2ones as anti proliferative compounds
MX2016015350A MX2016015350A (en) 2014-06-06 2014-12-15 1h-1,8- naphthyridin-2ones as anti proliferative compounds.
JP2017516226A JP2017516867A (en) 2014-06-06 2014-12-15 1H-1,8-naphthyridin-2-one as an antiproliferative compound
SG11201610149YA SG11201610149YA (en) 2014-06-06 2014-12-15 1h-1,8- naphthyridin-2ones as anti proliferative compounds
AP2016009604A AP2016009604A0 (en) 2014-06-06 2014-12-15 1h-1,8- naphthyridin-2ones as anti proliferative compounds
CN201480079519.1A CN106573929A (en) 2014-06-06 2014-12-15 1h-1,8- naphthyridin-2ones as anti proliferative compounds
CA2950250A CA2950250A1 (en) 2014-06-06 2014-12-15 1h-1,8- naphthyridin-2ones as anti proliferative compounds
EP14838923.2A EP3152205A1 (en) 2014-06-06 2014-12-15 1h-1,8- naphthyridin-2ones as anti proliferative compounds
IL249118A IL249118A0 (en) 2014-06-06 2016-11-22 1h-1,8- naphthyridin-2ones as anti proliferative compounds
MA39524A MA39524A1 (en) 2014-06-06 2016-12-05 1h-1,8-naphthyridin-2-ones used as antiproliferative compounds
PH12016502432A PH12016502432A1 (en) 2014-06-06 2016-12-06 1h-1,8-naphthyridin-2-ones as anti proliferative compounds
HK17104965.4A HK1231469A1 (en) 2014-06-06 2017-05-17 1h-1,8- naphthyridin-2ones as anti proliferative compounds 1h-18-
US15/681,278 US10047088B2 (en) 2014-06-06 2017-08-18 1H-1,8-naphthyridin-2-ones as anti proliferative compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN2781/CHE/2014 2014-06-06
IN2781CH2014 2014-06-06

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/316,832 A-371-Of-International US9765072B2 (en) 2014-06-06 2014-12-15 1H-1,8-naphthyridin-2ones as anti proliferative compounds
US15/681,278 Division US10047088B2 (en) 2014-06-06 2017-08-18 1H-1,8-naphthyridin-2-ones as anti proliferative compounds

Publications (1)

Publication Number Publication Date
WO2015186137A1 true WO2015186137A1 (en) 2015-12-10

Family

ID=52589723

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2014/000777 WO2015186137A1 (en) 2014-06-06 2014-12-15 1h-1,8- naphthyridin-2ones as anti proliferative compounds

Country Status (18)

Country Link
US (2) US9765072B2 (en)
EP (1) EP3152205A1 (en)
JP (1) JP2017516867A (en)
KR (1) KR20170016921A (en)
CN (1) CN106573929A (en)
AP (1) AP2016009604A0 (en)
AU (1) AU2014396394A1 (en)
BR (1) BR112016028674A2 (en)
CA (1) CA2950250A1 (en)
CL (1) CL2016003102A1 (en)
EA (1) EA201692371A1 (en)
HK (1) HK1231469A1 (en)
IL (1) IL249118A0 (en)
MA (1) MA39524A1 (en)
MX (1) MX2016015350A (en)
PH (1) PH12016502432A1 (en)
SG (1) SG11201610149YA (en)
WO (1) WO2015186137A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107663151A (en) * 2016-07-28 2018-02-06 江苏豪森药业集团有限公司 The intermediate synthetic method of methanesulfonic acid fluorine imatinib
WO2018068739A1 (en) * 2016-10-13 2018-04-19 深圳市塔吉瑞生物医药有限公司 Alkynyl heterocyclic compound for inhibiting protein kinase activity

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9765072B2 (en) * 2014-06-06 2017-09-19 Natco Pharma Limited 1H-1,8-naphthyridin-2ones as anti proliferative compounds

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007075869A2 (en) 2005-12-23 2007-07-05 Ariad Pharmaceuticals, Inc. Bicyclic heteroaryl compounds
WO2007133560A2 (en) * 2006-05-08 2007-11-22 Ariad Pharmaceuticals, Inc. Acetylenic heteroaryl compounds
WO2008046003A2 (en) 2006-10-11 2008-04-17 Deciphera Pharmaceuticals, Llc Kinase inhibitors useful for the treatment of myleoproliferative diseases and other proliferative diseases
US20130040949A1 (en) * 2009-12-29 2013-02-14 Dana-Farber Cancer Institute, Inc. Type ii raf kinase inhibitors

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101490053B (en) * 2006-05-08 2013-09-11 阿里亚德医药股份有限公司 Bicyclic heteroaryl compounds
JP5676650B2 (en) 2010-01-29 2015-02-25 ハンミ・サイエンス・カンパニー・リミテッドHanmi Science Co., Ltd. Thieno [3,2-d] pyrimidine derivatives having protein kinase inhibitory activity
US9765072B2 (en) * 2014-06-06 2017-09-19 Natco Pharma Limited 1H-1,8-naphthyridin-2ones as anti proliferative compounds

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007075869A2 (en) 2005-12-23 2007-07-05 Ariad Pharmaceuticals, Inc. Bicyclic heteroaryl compounds
US8114874B2 (en) 2005-12-23 2012-02-14 Ariad Pharmaceuticals, Inc. Substituted acetylenic imidazo[1,2-B]pyridazine compounds as kinase inhibitors
WO2007133560A2 (en) * 2006-05-08 2007-11-22 Ariad Pharmaceuticals, Inc. Acetylenic heteroaryl compounds
WO2008046003A2 (en) 2006-10-11 2008-04-17 Deciphera Pharmaceuticals, Llc Kinase inhibitors useful for the treatment of myleoproliferative diseases and other proliferative diseases
US20130040949A1 (en) * 2009-12-29 2013-02-14 Dana-Farber Cancer Institute, Inc. Type ii raf kinase inhibitors

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
DENG X ET AL: "Broad spectrum alkynyl inhibitors of T315I Bcr-Abl", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, AMSTERDAM, NL, vol. 20, no. 14, 15 July 2010 (2010-07-15), pages 4196 - 4200, XP027104017, ISSN: 0960-894X, [retrieved on 20100519] *
HUANG WEI-SHENG ET AL: "Discovery of 3-[2-(Imidazo[1,2-b]pyridazin-3-yl)ethynyl]-4-methyl-N-{ 4-[(4-methylpiperazin-1-yl)methyl]-3-(trifluoromethyl)phenyl}benzamide (AP24534), a Potent, Orally Active Pan-Inhibitor of Breakpoint Cluster Region-Abelson (BCR-ABL) Kinase Including the T315I Gatekeeper Mutant", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, vol. 53, no. 12, 24 June 2010 (2010-06-24), pages 4701 - 4719, XP002615777, ISSN: 0022-2623, [retrieved on 20100601], DOI: 10.1021/JM100395Q *
TIANJUN ZHOU ET AL: "Structural Mechanism of the Pan-BCR-ABL Inhibitor Ponatinib (AP24534): Lessons for Overcoming Kinase Inhibitor Resistance", CHEMICAL BIOLOGY & DRUG DESIGN, vol. 77, no. 1, 30 November 2010 (2010-11-30), pages 1 - 11, XP055065379, ISSN: 1747-0277, DOI: 10.1111/j.1747-0285.2010.01054.x *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107663151A (en) * 2016-07-28 2018-02-06 江苏豪森药业集团有限公司 The intermediate synthetic method of methanesulfonic acid fluorine imatinib
CN107663151B (en) * 2016-07-28 2021-11-26 江苏豪森药业集团有限公司 Intermediate synthesis method of flumatinib mesylate
WO2018068739A1 (en) * 2016-10-13 2018-04-19 深圳市塔吉瑞生物医药有限公司 Alkynyl heterocyclic compound for inhibiting protein kinase activity
CN108473476A (en) * 2016-10-13 2018-08-31 深圳市塔吉瑞生物医药有限公司 Alkynyl heterocycle compound for inhibiting protein kinase activity
CN108473476B (en) * 2016-10-13 2021-02-19 深圳市塔吉瑞生物医药有限公司 Alkynyl heterocyclic compounds for inhibiting protein kinase activity

Also Published As

Publication number Publication date
BR112016028674A2 (en) 2017-08-22
JP2017516867A (en) 2017-06-22
KR20170016921A (en) 2017-02-14
PH12016502432A1 (en) 2017-03-06
US9765072B2 (en) 2017-09-19
AP2016009604A0 (en) 2016-12-31
MX2016015350A (en) 2017-05-10
AU2014396394A1 (en) 2016-12-08
EA201692371A1 (en) 2017-04-28
CN106573929A (en) 2017-04-19
CA2950250A1 (en) 2015-12-10
US10047088B2 (en) 2018-08-14
SG11201610149YA (en) 2017-01-27
HK1231469A1 (en) 2017-12-22
IL249118A0 (en) 2017-01-31
US20170114057A1 (en) 2017-04-27
MA39524A1 (en) 2018-01-31
US20170349588A1 (en) 2017-12-07
EP3152205A1 (en) 2017-04-12
CL2016003102A1 (en) 2017-06-23

Similar Documents

Publication Publication Date Title
US8486951B2 (en) Cyclopropane amides and analogs exhibiting anti-cancer and anti-proliferative activities
US10005761B2 (en) Compounds and compositions as protein kinase inhibitors
KR102166002B1 (en) Indazole compounds as fgfr kinase inhibitor, preparation and use thereof
US8933103B2 (en) Pyridone derivatives
AU2012208388C1 (en) Diarylacetylene hydrazide containing tyrosine kinase inhibitors
JP2019527725A (en) 4-substituted aminoisoquinoline derivatives
WO2012000304A1 (en) Heterocyclic alkynyl benzene compounds and medical compositions and uses thereof
WO2013170770A1 (en) Acetylene derivatives having antitumor activity
CA2786424A1 (en) Raf kinase inhibitors
WO2013097280A1 (en) Quinoline and cinnoline compounds and use thereof
US20140221374A1 (en) Raf kinase inhibitors
US10047088B2 (en) 1H-1,8-naphthyridin-2-ones as anti proliferative compounds
CN105992767A (en) Wnt pathway modulators
JP2023021316A (en) Pyridinamine-pyridone and pyrimidinamine-pyridone compounds
WO2017140269A1 (en) Substituted amino six-membered nitric heterocyclic ring compound and preparation and use thereof
JP2023551116A (en) Succinate and its crystalline forms as therapeutic agents
WO2021121146A1 (en) Crystal form a of aminopyrimidine mesylate compound, preparation method therefor, and application thereof
US9902709B2 (en) Polysubstituted pyridine compound, preparation method, use and pharmaceutical composition
TW200538180A (en) New compounds
KR101663335B1 (en) 6-1--1--4--2-3-5-2--4----2---2--3- novel polymorphic forms of 6-1-methyl-1h-pyrazol-4-yl-2-3-5-2-morpholin-4-yl-ethoxy-pyrimidin-2-yl-benzyl-2h-pyridazin-3-one dihydrogenphosphate and processes of manufacturing thereof
KR102382641B1 (en) Novel TYRO 3 inhibitory compounds, preparation method thereof, pharmaceutical composition for use in preventing or treating TYRO 3 relating diseases containing the same as an active ingredient
US11806350B2 (en) Prevention and/or treatment of CNS disorders
TW201209050A (en) Compounds and compositions as protein kinase inhibitors
US20150232452A1 (en) Novel raf kinase inhibitors

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: 14838923

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 249118

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/015350

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2950250

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: NC2016/0004987

Country of ref document: CO

Ref document number: 39524

Country of ref document: MA

ENP Entry into the national phase

Ref document number: 2017516226

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 15316832

Country of ref document: US

Ref document number: 12016502432

Country of ref document: PH

ENP Entry into the national phase

Ref document number: 2014396394

Country of ref document: AU

Date of ref document: 20141215

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016028674

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 201692371

Country of ref document: EA

ENP Entry into the national phase

Ref document number: 20177000113

Country of ref document: KR

Kind code of ref document: A

REEP Request for entry into the european phase

Ref document number: 2014838923

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2014838923

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 14376

Country of ref document: GE

ENP Entry into the national phase

Ref document number: 112016028674

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20161206