WO2013190320A1 - Imidazopyridines comme inhibiteurs de la kinase aurora et/ou de la flt3 - Google Patents

Imidazopyridines comme inhibiteurs de la kinase aurora et/ou de la flt3 Download PDF

Info

Publication number
WO2013190320A1
WO2013190320A1 PCT/GB2013/051634 GB2013051634W WO2013190320A1 WO 2013190320 A1 WO2013190320 A1 WO 2013190320A1 GB 2013051634 W GB2013051634 W GB 2013051634W WO 2013190320 A1 WO2013190320 A1 WO 2013190320A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
solvate
acceptable salt
compounds
Prior art date
Application number
PCT/GB2013/051634
Other languages
English (en)
Inventor
Julian Blagg
Vassilios Bavetsias
Andrew S. Moore
Spyridon Linardopoulos
Original Assignee
Cancer Research Technology 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
Application filed by Cancer Research Technology Limited filed Critical Cancer Research Technology Limited
Publication of WO2013190320A1 publication Critical patent/WO2013190320A1/fr

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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Definitions

  • the present invention relates to pharmaceutically active compounds. More specifically, the present invention relates to compounds that are inhibitors of Aurora kinase enzyme activity. Compounds of the invention also act as inhibitors of FMS-like tyrosine kinase 3 (FLT3) activity. The present invention also relates to processes for the preparation of these compounds, to pharmaceutical compositions comprising them, and to their use in the treatment of proliferative disorders, such as cancer, as well as other diseases or conditions in which Aurora kinase and/or FLT3 activity is implicated.
  • FLT3 FMS-like tyrosine kinase 3
  • Proliferative diseases such as cancer
  • cancer are characterised by uncontrolled and unregulated cellular proliferation. Precisely what causes a cell to proliferate in an uncontrolled and unregulated manner has been the focus of intense research over recent decades.
  • Aurora kinases a family of three serine-threonine kinases designated as A, B, and C, play key and distinct roles in different stages of mitosis. 1"3 At the early stages of mitosis, Aurora-A forms a complex with the targeting protein for Xklp2 (TPX2) that regulates centrosome maturation and mitotic spindle assembly. 4 ' 5 Aurora-B forms complexes with the inner centromere protein (INCENP), survivin and borealin thereby regulating chromosome condensation, chromosome alignment, mitotic checkpoint and cytokinesis.
  • TPX2 Xklp2
  • ICENP inner centromere protein
  • This particular compound (known as CCT137690) is a potent and orally bioavailable inhibitor of Aurora kinases that inhibits the growth of a SW620 human colon carcinoma xenograft in vivo with concomitant biomarker modulation consistent with target engagement. 26
  • It is therefore an object of the present invention to provide orally bioavailable inhibitors of Aurora kinase enzyme activity are suitable for preclinical and clinical evaluation.
  • FLT3 is a trans-membrane kinase that belongs to the class III receptor tyrosine kinase (RTK) family. Binding of FLT3-ligand (FL) to its receptor leads to homodimerisation, autophosphorylation and subsequent activation of downstream signalling pathways 34 . High levels of FLT3 expression have been found in acute myeloid leukaemia (AML) blasts, and two major classes of mutations, i.e. internal-tandem duplications (ITDs) and tyrosine kinase domain (TKD) point mutations, have been identified in AML patients 34 ' 35 .
  • AML acute myeloid leukaemia
  • ITDs internal-tandem duplications
  • TKD tyrosine kinase domain
  • the present invention provides a compound, or a pharmaceutically acceptable salt or solvate thereof as defined herein.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the invention as defined herein, or a pharmaceutically acceptable salt or solvate thereof, and one or more pharmaceutically acceptable excipients.
  • the present invention relates to a compound of the invention as defined herein, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition as defined herein, for use in therapy.
  • the present invention relates to a compound of the invention as defined herein, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition as defined herein, for use in the treatment of diseases or conditions in which Aurora kinase and/or FLT3 activity is implicated.
  • the present invention relates to the use of a compound of the invention as defined herein, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for use in the treatment of diseases or conditions in which Aurora kinase and/or FLT3 activity is implicated.
  • the present invention relates to a method of treating a disease or condition in which Aurora kinase and/or FLT3 activity is implicated, said method comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound of the invention as defined herein, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition as defined herein.
  • the present invention provides a compound, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition as defined herein, for use in the treatment of a proliferative disorder, such as cancer.
  • a proliferative disorder such as cancer.
  • the cancer is a human cancer.
  • the present invention provides the use of a compound, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for use in the treatment of a proliferative disorder, such as cancer.
  • a proliferative disorder such as cancer.
  • the cancer is a human cancer.
  • the present invention provides a method of treating a proliferative disorder, such as cancer, said method comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition as defined herein.
  • a proliferative disorder such as cancer
  • the cancer is a human cancer.
  • the present invention provides a compound, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition as defined herein, for use in the production of an Aurora kinase and/or FLT3 inhibitory effect.
  • the present invention provides the use of a compound, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for use in the production of an Aurora kinase and/or FLT3 inhibitory effect.
  • the present invention provides a method of producing an Aurora kinase and/or FLT3 inhibitory effect in vitro, said method comprising administering an effective amount of a compound, or a pharmaceutically acceptable salt or solvate thereof.
  • the present invention provides a method of producing an Aurora kinase and/or FLT3 inhibitory effect in vivo, said method comprising administering an effective amount of a compound, or a pharmaceutically acceptable salt or solvate thereof.
  • the present invention provides a method of inhibiting cell proliferation in vitro or in vivo, said method comprising contacting a cell with an effective amount of a compound as defined herein, or a pharmaceutically acceptable salt or solvate thereof.
  • the present invention further provides a method of synthesising a compound, or a pharmaceutically acceptable salt or solvate thereof, as defined herein.
  • the present invention provides a compound, or a pharmaceutically acceptable salt or solvate thereof, obtainable by, or obtained by, or directly obtained by a method of synthesis as defined herein.
  • the present invention provides novel intermediates as defined herein which are suitable for use in any one of the synthetic methods set out herein.
  • references to "treating” or “treatment” include prophylaxis as well as the alleviation of established symptoms of a condition.
  • “Treating” or “treatment” of a state, disorder or condition therefore includes: (1 ) preventing or delaying the appearance of clinical symptoms of the state, disorder or condition developing in a human that may be afflicted with or predisposed to the state, disorder or condition but does not yet experience or display clinical or subclinical symptoms of the state, disorder or condition, (2) inhibiting the state, disorder or condition, i.e., arresting, reducing or delaying the development of the disease or a relapse thereof (in case of maintenance treatment) or at least one clinical or subclinical symptom thereof, or (3) relieving or attenuating the disease, i.e., causing regression of the state, disorder or condition or at least one of its clinical or subclinical symptoms.
  • a “therapeutically effective amount” means the amount of a compound that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease.
  • the “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated.
  • the present invention provides a compound of formula I shown below:
  • R 2 is a 5-membered heteroaryl ring comprising at least one nitrogen atom and at least one oxygen atom in the ring and wherein said heteroaryl ring is optionally substituted by methyl, ethyl, CF 3 or CHF 2 ;
  • the compounds of the present invention demonstrate improved metabolic stability (demonstrated by improved human microsomal stability) and a wider therapeutic index against hERG relative to Example 39 of WO2009/001021 .
  • Particular compounds of the invention include, for example, compounds of the formula I, or pharmaceutically acceptable salts thereof, wherein, unless otherwise stated, each of R and R 2 has any of the meanings defined hereinbefore or in any of paragraphs (1 ) to (10) hereinafter:-
  • R 2 is a 5-membered heteroaryl ring comprising at least one nitrogen atom and at least one oxygen atom in the ring and wherein said heteroaryl ring is optionally substituted by methyl, CF 3 or CHF 2 ;
  • R 2 is a 5-membered heteroaryl ring comprising at least one nitrogen atom and at least one oxygen atom in the ring and wherein said heteroaryl ring is optionally substituted by methyl;
  • R 2 is a 5-membered heteroaryl ring comprising one nitrogen atom and one oxygen atom in the ring and wherein said heteroaryl ring is optionally substituted by one methyl;
  • R 2 is a group of the formula:
  • R a is hydrogen, methyl, ethyl, CF 3 or CHF 2 ;
  • R 2 is a group of the formula:
  • R a is hydrogen, methyl, CF 3 or CHF 2
  • R 2 is a group of the formula:
  • R a is hydrogen or methyl
  • R a is hydrogen
  • R is chloro
  • R 2 is as defined in any one of paragraphs (3) to (10) above, and is especially as defined in paragraphs (8) to (10) above.
  • R a is hydrogen or methyl, especially methyl.
  • Particular compounds of the present invention include any one of the following:
  • a suitable pharmaceutically acceptable salt of a compound of the invention is, for example, an acid-addition salt of a compound of the invention which is sufficiently basic, for example, an acid-addition salt with, for example, an inorganic or organic acid, for example hydrochloric, hydrobromic, sulfuric, phosphoric, trifluoroacetic, formic, citric or maleic acid.
  • the present invention also encompasses compounds of the invention as defined herein which comprise one or more isotopic substitutions.
  • H may be in any isotopic form, including 1 H, 2 H(D), and 3 H (T);
  • C may be in any isotopic form, including 12 C, 13 C, and 14 C; and the like.
  • N-oxides may also form N- oxides.
  • a reference herein to a compound of the formula I that contains an amine function also includes the N-oxide.
  • one or more than one nitrogen atom may be oxidised to form an N-oxide.
  • Particular examples of N-oxides are the N-oxides of a nitrogen atom of a nitrogen- containing heterocycle.
  • N-Oxides can be formed by treatment of the corresponding amine with an oxidizing agent such as hydrogen peroxide or a per-acid (e.g.
  • N-oxides can be made by the procedure of L. W. Deady (Syn. Comm. 1977, 7, 509-514) in which the amine compound is reacted with m-chloroperoxybenzoic acid (MCPBA), for example, in an inert solvent such as dichloromethane.
  • MCPBA m-chloroperoxybenzoic acid
  • the compounds of the invention may be administered in the form of a pro-drug which is broken down in the human or animal body to release a compound of the invention.
  • a pro-drug may be used to alter the physical properties and/or the
  • a pro-drug can be formed when the compound of the invention contains a suitable group or substituent to which a property-modifying group can be attached.
  • Examples of pro-drugs include in vivo cleavable amide derivatives that may be formed at an amino group in a compound of the invention.
  • the present invention includes those compounds of the formula I as defined hereinbefore when made available by organic synthesis and when made available within the human or animal body by way of cleavage of a pro-drug thereof. Accordingly, the present invention includes those compounds of the formula I that are produced by organic synthetic means and also such compounds that are produced in the human or animal body by way of metabolism of a precursor compound, that is a compound of the formula I may be a synthetically-produced compound or a
  • a suitable pharmaceutically acceptable pro-drug of a compound of the formula I is one that is based on reasonable medical judgement as being suitable for
  • pro-drug Various forms of pro-drug have been described, for example in the following documents :- a) Methods in Enzvmoloav, Vol. 42, p. 309-396, edited by K. Widder, et al. (Academic Press, 1985);
  • the in vivo effects of a compound of the formula I may be exerted in part by one or more metabolites that are formed within the human or animal body after administration of a compound of the formula I. As stated hereinbefore, the in vivo effects of a compound of the formula I may also be exerted by way of metabolism of a precursor compound (a pro-drug).
  • compounds of formula I may also be covalently linked (at any suitable position) to other groups such as, for example, solubilising moieties (for example, PEG polymers), moieties that enable them to be bound to a solid support (such as, for example, biotin-containing moieties), and targeting ligands (such as antibodies or antibody fragments).
  • solubilising moieties for example, PEG polymers
  • moieties that enable them to be bound to a solid support such as, for example, biotin-containing moieties
  • targeting ligands such as antibodies or antibody fragments
  • protecting groups see one of the many general texts on the subject, for example, 'Protective Groups in Organic Synthesis' by Theodora Green (publisher: John Wiley & Sons).
  • Protecting groups may be removed by any convenient method described in the literature or known to the skilled chemist as appropriate for the removal of the protecting group in question, such methods being chosen so as to effect removal of the protecting group with the minimum disturbance of groups elsewhere in the molecule.
  • reactants include, for example, groups such as amino, carboxy or hydroxy it may be desirable to protect the group in some of the reactions mentioned herein.
  • a suitable protecting group for an amino or alkylamino group is, for example, an acyl group, for example an alkanoyl group such as acetyl, an alkoxycarbonyl group, for example a methoxycarbonyl, ethoxycarbonyl or f-butoxycarbonyl group, an arylmethoxycarbonyl group, for example benzyloxycarbonyl, or an aroyl group, for example benzoyl.
  • the deprotection conditions for the above protecting groups necessarily vary with the choice of protecting group.
  • an acyl group such as an alkanoyl or alkoxycarbonyl group or an aroyl group may be removed by, for example, hydrolysis with a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • a suitable base such as an alkali metal hydroxide, for example lithium or sodium hydroxide.
  • an acyl group such as a ferf-butoxycarbonyl group may be removed, for example, by treatment with a suitable acid as hydrochloric, sulfuric or phosphoric acid or trifluoroacetic acid and an arylmethoxycarbonyl group such as a benzyloxycarbonyl group may be removed, for example, by hydrogenation over a catalyst such as palladium-on-carbon, or by treatment with a Lewis acid for example BF 3 .OEt 2 .
  • a suitable alternative protecting group for a primary amino group is, for example, a phthaloyl group which may be removed by treatment with an alkylamine, for example dimethylaminopropylamine, or with hydrazine.
  • the compounds of the present invention may be prepared by using the general synthetic techniques described in WO2007/072017 and WO2009/001021 , the entire contents of which are incorporated herein by reference.
  • the present invention provides a method of synthesising a compound of the formula I, or a pharmaceutically acceptable salt or solvate thereof, the method comprising:
  • R 2 each have any one of the meanings set out hereinbefore; with 3-(4-methylpiperazin-1 -yl)benzaldehyde in the presence of a suitable reducing agent;
  • the reaction between the compound of formula A and 3-(4- methylpiperazin-1 -yl)benzaldehyde takes place in the presence of a suitable solvent.
  • a suitable solvent Any suitable solvent or solvent mixture may be used for this reaction.
  • suitable solvents include DMSO, water, DMF, and alcohols e.g. EtOH.
  • the reaction proceeds in the presence of a suitable reducing agent, such as aqueous Na 2 S 2 0 4 . 26
  • a suitable reducing agent such as aqueous Na 2 S 2 0 4 . 26
  • the reaction may also be carried out an elevated temperature, for example a temperature within the range of 50 to 190°C may be used (depending on the nature of the solvent).
  • the process defined herein may further comprise the step of subjecting the compound of formula I to a salt exchange, particularly in situations where the compound of formula I is formed as a mixture of different salt forms.
  • the salt exchange suitably comprises immobilising the compound of formula I on a suitable solid support or resin, and eluting the compounds with an appropriate acid to yield a single salt of the compound of formula I.
  • the compounds of formula A can be prepared by processes known in the art.
  • a pharmaceutical composition which comprises a compound of the invention as defined hereinbefore, or a pharmaceutically acceptable salt or solvate thereof, in association with a pharmaceutically acceptable diluent or carrier.
  • compositions of the invention may be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder) or for parenteral administration (for example as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular, intraperitoneal or intramuscular dosing or as a suppository for rectal dosing).
  • oral use for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or
  • compositions of the invention may be obtained by conventional procedures using conventional pharmaceutical excipients, well known in the art.
  • compositions intended for oral use may contain, for example, one or more colouring, sweetening, flavouring and/or preservative agents.
  • An effective amount of a compound of the present invention for use in therapy of proliferative disease is an amount sufficient to symptomatically relieve in a warm-blooded animal, particularly a human the symptoms of infection, to slow the progression of infection, or to reduce in patients with symptoms of infection the risk of getting worse.
  • the amount of active ingredient that is combined with one or more excipients to produce a single dosage form will necessarily vary depending upon the host treated and the particular route of administration.
  • a formulation intended for oral administration to humans will generally contain, for example, from 0.5 mg to 0.5 g of active agent (more suitably from 0.5 to 100 mg, for example from 1 to 30 mg) compounded with an appropriate and convenient amount of excipients which may vary from about 5 to about 98 percent by weight of the total composition.
  • the size of the dose for therapeutic or prophylactic purposes of a compound of the formula I will naturally vary according to the nature and severity of the conditions, the age and sex of the animal or patient and the route of administration, according to well known principles of medicine.
  • a daily dose in the range for example, 0.1 mg/kg to 30 mg/kg body weight is received, given if required in divided doses.
  • a parenteral route is employed.
  • a dose in the range for example, 0.1 mg/kg to 30 mg/kg body weight will generally be used.
  • a dose in the range for example, 0.05 mg/kg to 25 mg/kg body weight will be used.
  • Oral administration may also be suitable, particularly in tablet form.
  • unit dosage forms will contain about 0.5 mg to 0.5 g of a compound of this invention.
  • the compounds of the invention are inhibitors of Aurora kinase activity and/or FLT3.
  • the present invention provides a method of inhibiting Aurora kinase activity and/or FLT3 in a cell, the method comprising administering to said cell compound of formula I as defined herein, or a pharmaceutically acceptable salt or solvate thereof.
  • the present invention provides a method of inhibiting Aurora kinase activity and/or FLT3 in vitro or in vivo, said method comprising contacting a cell with an effective amount of a compound, or a pharmaceutically acceptable salt or solvate thereof, as defined herein.
  • the present invention provides a method of inhibiting Aurora kinase activity and/or FLT3 in a human or animal subject in need of such inhibition, the method comprising administering to said subject an effective amount of a compound of formula I as defined herein, or a pharmaceutically acceptable salt or solvate thereof.
  • the Aurora kinase may be Aurora kinase A, B or C.
  • the present invention provides a compound of Formula I, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition as defined herein for use in therapy.
  • the present invention provides a compound of formula I as defined herein, or a pharmaceutically acceptable salt or solvate thereof for use in the treatment of disease or condition associated with Aurora kinase activity and/or FLT3 activity.
  • the present invention provides the use of a compound of formula I as defined herein, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for use in the treatment of disease or condition associated with Aurora kinase activity and/or FLT3 activity.
  • the present invention provides a method of treating a proliferative disorder in a human or animal subject, the method comprising administering to said subject a therapeutically acceptable amount of a compound of formula I as defined herein, or a pharmaceutically acceptable salt or solvate thereof.
  • the present invention provides a compound of formula I as defined herein, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of a proliferative disorder.
  • the present invention provides the use of a compound of formula I as defined herein, or a pharmaceutically acceptable salt or solvate thereof, in the manufacture of a medicament for use in the treatment of a proliferative disorder.
  • the present invention provides a compound, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition as defined herein for use in the treatment of cancer.
  • the present invention provides the use of a compound, or a pharmaceutically acceptable salt or solvate thereof, as defined herein in the manufacture of a medicament for use in the treatment of cancer.
  • the present invention provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient a therapeutically effective amount of a compound, or a pharmaceutically acceptable salt or solvate thereof, or a pharmaceutical composition as defined herein.
  • the compounds of the invention may be useful, for example, for the treatment of colorectal, breast, lung, prostate, pancreatic or bladder and renal cancer or leukaemias or lymphomas.
  • the compounds of the present invention are useful for the treatment of leukaemias.
  • High expression of Aurora kinases has been demonstrated in leukaemia (cell lines and patient cohorts).
  • 27"30 In addition, internal tandem duplication of the F/.T3 gene ⁇ FLT3-YTD) results in constitutive FLT3 kinase activation.
  • FLT3- ⁇ JD occurs in 20-35% of adults and 15% of children with AML conferring a poor prognosis in both age groups.
  • the compounds are useful for treating leukaemias such as acute myeloid leukaemia (AML), myelodysplastic syndrome (MDS), chronic lymphocytic leukaemia (CLL) and multiple myeloma.
  • leukaemias such as acute myeloid leukaemia (AML), myelodysplastic syndrome (MDS), chronic lymphocytic leukaemia (CLL) and multiple myeloma.
  • AML acute myeloid leukaemia
  • MDS myelodysplastic syndrome
  • CLL chronic lymphocytic leukaemia
  • multiple myeloma multiple myeloma.
  • the compounds of the present invention are also envisaged to be useful for the treatment of neuroblastoma,
  • the compounds of the present invention are expected to be of particular benefit in patients that have failed treatment with standard therapies. It predicted that the compounds of the present invention will also be of value for the treatment of older patients (e.g. over 60 years old) with leukaemia (e.g. AML) because such patients are expected to benefit for aurora kinase inhibition.
  • leukaemia e.g. AML
  • the compounds of the present invention are also expected to be of value in the treatment of children with leukaemia (e.g. newly diagnosed FLT3-mutated AML and infant AML), as well as neuroblastomas.
  • leukaemia e.g. newly diagnosed FLT3-mutated AML and infant AML
  • neuroblastomas e.g. neuroblastomas
  • the compounds of the invention or pharmaceutical composition comprising the active compound may be administered to a subject by any convenient route of administration, whether systemically/ peripherally or topically (i.e. at the site of desired action).
  • Routes of administration include, but are not limited to, oral (e.g., by ingestion); buccal; sublingual; transdermal (including, e.g., by a patch, plaster, etc.); transmucosal (including, e.g., by a patch, plaster, etc.); intranasal (e.g., by nasal spray); ocular (e.g., by eyedrops); pulmonary (e.g., by inhalation or insufflation therapy using, e.g., via an aerosol, e.g., through the mouth or nose); rectal (e.g., by suppository or enema); vaginal (e.g., by pessary); parenteral, for example, by injection, including subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, sub
  • the compounds of the invention may be administered alone as a monotherapy or may administered in combination with one or more additional therapeutic agents.
  • the selection of the one or more additional therapeutic agents will of course vary depending on the disease or condition to be treated and its severity.
  • antiproliferative treatment may be applied as a sole therapy or may involve, in addition to the compound of the invention, conventional surgery or radiotherapy or chemotherapy.
  • chemotherapy may include one or more of the following categories of anti-tumour agents:-
  • antiproliferative/antineoplastic drugs and combinations thereof as used in medical oncology, such as alkylating agents (for example cis-platin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustard, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblast
  • cytostatic agents such as antioestrogens (for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHRH antagonists or LHRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5 -reductase such as finasteride;
  • antioestrogens for example tamoxifen, fulvestrant, toremifene, raloxifene, droloxifene and iodoxyfene
  • antiandrogens for example
  • anti-invasion agents for example c-Src kinase family inhibitors like 4-(6-chloro- 2,3-methylenedioxyanilino)-7-[2-(4-methylpiperazin-1 -yl)ethoxy]-5-tetrahydropyran-4- yloxyquinazoline (AZD0530; International Patent Application WO 01/94341 ), A/-(2-chloro- 6-methylphenyl)-2- ⁇ 6-[4-(2-hydroxyethyl)piperazin-1 -yl]-2-methylpyrimidin-4- ylamino ⁇ thiazole-5-carboxamide (dasatinib, BMS-354825; J. Med.
  • anti-invasion agents for example c-Src kinase family inhibitors like 4-(6-chloro- 2,3-methylenedioxyanilino)-7-[2-(4-methylpiperazin-1 -yl)ethoxy]-5-tetrahydro
  • inhibitors of growth factor function include growth factor antibodies and growth factor receptor antibodies (for example the anti-erbB2 antibody trastuzumab [HerceptinTM], the anti-EGFR antibody panitumumab, the anti-erbB1 antibody cetuximab [Erbitux, C225] and any growth factor or growth factor receptor antibodies disclosed by Stern et al. Critical reviews in oncology/haematology, 2005, Vol.
  • inhibitors also include tyrosine kinase inhibitors, for example inhibitors of the epidermal growth factor family (for example EGFR family tyrosine kinase inhibitors such as A/-(3-chloro-4-fluorophenyl)-7-methoxy-6-(3- morpholinopropoxy)quinazolin-4-amine (gefitinib, ZD1839), A/-(3-ethynylphenyl)-6,7- bis(2-methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774) and 6-acrylamido-A/-(3- chloro-4-fluorophenyl)-7-(3-morpholinopropoxy)-quinazolin-4-amine (CI 1033), erbB2 tyrosine kinase inhibitors such as lapatinib); inhibitors of the hepatocyte growth factor family; inhibitors of the hepatocyte growth factor family; inhibitor
  • antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, [for example the anti-vascular endothelial cell growth factor antibody bevacizumab (AvastinTM) and for example, a VEGF receptor tyrosine kinase inhibitor such as vandetanib (ZD6474), vatalanib (PTK787), sunitinib (SU1 1248), axitinib (AG-013736), pazopanib (GW 786034) and 4-(4-fluoro-2-methylindol-5-yloxy)-6- methoxy-7-(3-pyrrolidin-1 -ylpropoxy)quinazoline (AZD2171 ; Example 240 within WO 00/47212), compounds such as those disclosed in International Patent Applications W097/22596, WO 97/30035, WO 97/32856 and WO 98/13354 and compounds that work by other mechanisms (for example linomide
  • an endothelin receptor antagonist for example zibotentan (ZD4054) or atrasentan;
  • antisense therapies for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense;
  • (ix) gene therapy approaches including, for example, using the compounds of the invention in combination with oncolytic adenoviruses, approaches to replace aberrant genes such as aberrant p53 or aberrant BRCA1 or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or radiotherapy such as multi-drug resistance gene therapy; and
  • (x) immunotherapy approaches including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies.
  • cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor
  • Such conjoint/combination treatment may be achieved by way of the simultaneous, sequential or separate dosing of the individual components of the treatment.
  • Such combination products employ the compounds of this invention within the dosage range described hereinbefore and the other pharmaceutically-active agent within its approved dosage range.
  • a combination suitable for use in the treatment of a disease or condition in which protein kinase activity is implicated as defined herein comprising a compound of the invention as defined hereinbefore, or a pharmaceutically acceptable salt or solvate thereof, and another therapeutic agent (e.g. an anti-tumour agent).
  • a disease or condition in which protein kinase activity is implicated as defined herein e.g. cancer
  • a pharmaceutically acceptable salt or solvate thereof e.g. an anti-tumour agent
  • a combination suitable for use in the treatment of a cancer comprising a compound of the invention as defined hereinbefore, or a pharmaceutically acceptable salt or solvate thereof, and any one of the anti-tumour agents listed under (i) - (ix) above.
  • a pharmaceutical composition which comprises a compound of the invention, or a pharmaceutically acceptable salt or solvate thereof in combination with one or more additional therapeutic agents (for example, an anti-tumour agent selected from one listed under (i) - (ix) herein above), in association with a pharmaceutically acceptable diluent or carrier.
  • additional therapeutic agents for example, an anti-tumour agent selected from one listed under (i) - (ix) herein above
  • the compounds of the present invention are expected to be particularly useful as part of a combination therapy with the existing standard of care for the treatment of older patients (i.e. patients over 60 years old), as such patients may well benefit for Aurora kinase inhibition (regardless of their FLT3 status).
  • the compounds of the present invention are also expected to be particularly useful as part of combination therapy with the existing standard of care for the treatment of children suffering from with leukaemia (e.g. AML) or neuroblastoma.
  • leukaemia e.g. AML
  • neuroblastoma e.g. AML
  • Figure 1 shows the efficacy of Example 1 against MOLM-13 human tumour xenografts in athymic mice: Top two pictures (from left to right): Mean tumour volumes ⁇ SEM, and Relative tumour volumes ⁇ SEM. Bottom two pictures (from left to right): Mouse body weights, and final tumour weights.
  • Flash column chromatography was performed using Merck silica gel 60 (0.025 - 0.04 mm). Column chromatography was also performed on a FlashMaster personal unit using isolute Flash silica columns or a Biotage SP1 purification system using Biotage Flash silica cartridges. Preparative TLC was performed on Analtech or Merck plates. Ion exchange chromatography was performed using acidic Isolute Flash SCX-II cartridges. 1 H NMR spectra were recorded on a Bruker Avance-500. Samples were prepared as solutions in a deuterated solvent and referenced to the appropriate internal non-deuterated solvent peak or tetramethylsilane.
  • Analytical separation was carried out at 30°C either on a Merck Chromolith SpeedROD column (RP-18e, 50 x 4.6 mm) using a flow rate of 2 mL/min in a 3.5 minute gradient elution with detection at 254 nm or on a Merck Purospher STAR column (RP-18e, 30 x 4 mm) using a flow rate of 1 .5 mL/min in a 3.5 minute gradient elution with detection at 254 nm.
  • the mobile phase was a mixture of methanol (solvent A) and water (solvent B) both containing formic acid at 0.1 %.
  • LC-HRMS analysis was performed on an Agilent 1200 series HPLC and diode array detector coupled to a 6520 Quadrupole-Time of flight mass spectrometer with dual multimode APCI/ESI source.
  • Analytical separation was carried out at 30°C on a Merck Purospher STAR column (RP-18e, 30 x 4 mm) using a flow rate of 1 .5 mL/min in a 4 minute gradient elution with detection at 254 nm.
  • the mobile phase was a mixture of methanol (solvent A) and water (solvent B) both containing formic acid at 0.1 %.
  • This compound was also produced in bulk quantities ranging from 0.070 g to 2.0 g and in yields ranging from 54% to 77%. The above method was used but the crude product was purified by column chromatography eluting with a gradient of methanol (10 to 20%) in dichloromethane.
  • ferf-Butyl 4-(isoxazol-3-ylmethyl)piperazine-1 -carboxylate (335 mg, 1 .24 mmol) was dissolved in dichloromethane (8.5 mL) and trifluoroacetic acid (1 .7 mL) was added. The reaction was stirred for 2 h. The solvents were evaporated and the residue was azeotroped twice with toluene and dried in vacuum over sodium hydroxide for 4.5 h.
  • Aurora kinase assays Aurora kinase IC 50 values were determined as previously described. 26 - 33
  • Mouse liver microsomal stability Compounds (10 ⁇ ) were incubated with male CD1 mouse liver microsomes (1 mg.mL "1 ) protein in the presence of NADPH (1 mM), UDPGA (2.5 mM) and MgCI 2 (3 mM) in phosphate buffered saline (10 mM) at 37 °C. Incubations were conducted for 0 and 30 minutes. Control incubations were generated by the omission of NADPH and UDPGA from the incubation reaction. The percentage compound remaining was determined after analysis by LCMS.
  • Human liver microsomal stability Compounds (10 ⁇ ) were incubated with mixed gender pooled human liver microsomes (1 mg.mL "1 ) protein in the presence of NADPH (1 mM), UDPGA (2.5 mM) and MgCI 2 (3 mM) in phosphate buffered saline (10 mM) at 37 °C. Incubations were conducted for 0 and 30 minutes. Control incubations were generated by the omission of NADPH and UDPGA from the incubation reaction. The percentage compound remaining was determined after analysis by LCMS.
  • hERG inhibition All hERG percentage inhibitions at 10 ⁇ compound concentration were determined by Millipore in a high-throughput cell-based electrophysiology assay for inhibition of hERG tail current 38 , and values are reported as a mean of multiple determinations. 0.3% DMSO aqueous vehicle negative control gave 7- 16% inhibition. Cisapride (1 ⁇ ) positive control gave 96-104% inhibition. hERG IC 50 values were determined by Millipore. 38 Also hERG IC 50 values were determined at Cyprotex pic measuring hERG tail-currents by whole-cell voltage-clamping. 39
  • Aurora kinase activity Aurora kinase activity, cell activity, microsomal stability, and hERG inhibition
  • results are mean values of two independent determinations or mean ( ⁇ SD) for n>2 unless specified otherwise.
  • Results are mean values for samples run in triplicate.
  • MLM/HLM Percentage of parent compound metabolised after a 30 min incubation.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne des composés de formule I : dans laquelle R1 et R2 sont tels que définis dans la description, ou l'un de ses sels ou solvates pharmaceutiquement acceptables. Les composés de formule I sont des inhibiteurs de la kinase aurora et/ou de la FLT3. La présente invention concerne également des procédés pour la préparation de ces composés, des compositions pharmaceutiques les contenant, et leur utilisation dans le traitement de troubles prolifératifs, tels que le cancer, ainsi que d'autres maladies ou états impliquant l'activité de la kinase aurora et/ou de la FLT3.
PCT/GB2013/051634 2012-06-21 2013-06-21 Imidazopyridines comme inhibiteurs de la kinase aurora et/ou de la flt3 WO2013190320A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1211019.3A GB201211019D0 (en) 2012-06-21 2012-06-21 Inhibitor compounds
GB1211019.3 2012-06-21

Publications (1)

Publication Number Publication Date
WO2013190320A1 true WO2013190320A1 (fr) 2013-12-27

Family

ID=46641295

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2013/051634 WO2013190320A1 (fr) 2012-06-21 2013-06-21 Imidazopyridines comme inhibiteurs de la kinase aurora et/ou de la flt3

Country Status (2)

Country Link
GB (1) GB201211019D0 (fr)
WO (1) WO2013190320A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016124939A1 (fr) * 2015-02-06 2016-08-11 Cancer Research Technology Limited Composés inhibiteurs de l'autotaxine
WO2020185856A1 (fr) * 2019-03-11 2020-09-17 The Children's Medical Center Corporation Méthodes pour accroître la production de plaquettes
CN114502555A (zh) * 2019-10-15 2022-05-13 常州千红生化制药股份有限公司 用于治疗增殖性疾病和疾患的4-(咪唑并[1,2-a]吡啶-3-基)-N-(吡啶-3-基)嘧啶-2-胺的衍生物

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007072017A2 (fr) * 2005-12-22 2007-06-28 The Institute Of Cancer Research Inhibiteurs d'enzymes
WO2009001021A1 (fr) * 2007-06-26 2008-12-31 Chroma Therapeutics Ltd. Dérivés d'imidazopyridine utiles comme inhibiteurs d'enzymes pour le traitement de maladies de prolifération des cellules et auto-immunes

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007072017A2 (fr) * 2005-12-22 2007-06-28 The Institute Of Cancer Research Inhibiteurs d'enzymes
WO2009001021A1 (fr) * 2007-06-26 2008-12-31 Chroma Therapeutics Ltd. Dérivés d'imidazopyridine utiles comme inhibiteurs d'enzymes pour le traitement de maladies de prolifération des cellules et auto-immunes

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BAVETSIAS ET AL.: "Optimization of Imidazo[4,5- b ]pyridine-Based Kinase Inhibitors: Identification of a Dual FLT3/Aurora Kinase Inhibitor as an Orally Bioavailable Preclinical Development Candidate for the Treatment of Acute Myeloid Leukemia", J. MED. CHEM., vol. 55, no. 20, 8 October 2012 (2012-10-08), pages 8721 - 8734, XP055067890, ISSN: 0022-2623, DOI: 10.1021/jm300952s *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016124939A1 (fr) * 2015-02-06 2016-08-11 Cancer Research Technology Limited Composés inhibiteurs de l'autotaxine
CN107428752A (zh) * 2015-02-06 2017-12-01 癌症研究科技有限公司 自家趋化素抑制性化合物
US10654846B2 (en) 2015-02-06 2020-05-19 Cancer Research Technology Limited Autotaxin inhibitory compounds
US11453666B2 (en) 2015-02-06 2022-09-27 Cancer Research Technology Limited Autotaxin inhibitory compounds
WO2020185856A1 (fr) * 2019-03-11 2020-09-17 The Children's Medical Center Corporation Méthodes pour accroître la production de plaquettes
CN114502555A (zh) * 2019-10-15 2022-05-13 常州千红生化制药股份有限公司 用于治疗增殖性疾病和疾患的4-(咪唑并[1,2-a]吡啶-3-基)-N-(吡啶-3-基)嘧啶-2-胺的衍生物

Also Published As

Publication number Publication date
GB201211019D0 (en) 2012-08-01

Similar Documents

Publication Publication Date Title
US11548896B2 (en) Heterocyclic compounds as RET kinase inhibitors
US11661423B2 (en) Heterocyclic compounds as RET kinase inhibitors
AU2018214431B2 (en) Derivatives of N-cycloalkyl/heterocycloalkyl-4-(imidazo [1,2-a]pyridine)pyrimidin-2-amine as therapeutic agents
US11274098B2 (en) Tricyclic compounds for use in treatment of proliferative disorders
WO2018167269A1 (fr) Composés utiles pour traiter ou prévenir un trouble induit par prmt5
EP3521286B1 (fr) Dérivés de 1,4-dihydrodioxine fusionnés en tant qu'inhibiteurs de facteur de transcription de choc thermique 1
EP2892889A1 (fr) Composés inhibiteurs
DK2864328T3 (en) PHARMACEUTICAL ACTIVE RELATIONS
AU2016375851B2 (en) Novel pyrrolo[3,2-c]pyridine-6-amino derivatives
WO2013190320A1 (fr) Imidazopyridines comme inhibiteurs de la kinase aurora et/ou de la flt3
EP3997088A1 (fr) Dérivés de 4-(imidazo [l, 2-a] pyridine-3-yl)-n-(pyridinyl) pyrimidine-2-amine en tant qu'agents thérapeutiques

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: 13731471

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13731471

Country of ref document: EP

Kind code of ref document: A1