WO2009029998A1 - Composés rétrométaboliques - Google Patents

Composés rétrométaboliques Download PDF

Info

Publication number
WO2009029998A1
WO2009029998A1 PCT/AU2008/001326 AU2008001326W WO2009029998A1 WO 2009029998 A1 WO2009029998 A1 WO 2009029998A1 AU 2008001326 W AU2008001326 W AU 2008001326W WO 2009029998 A1 WO2009029998 A1 WO 2009029998A1
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
pyrimidin
ylamino
alkyl
cyanomethylcarbamoyl
Prior art date
Application number
PCT/AU2008/001326
Other languages
English (en)
Inventor
Christopher John Burns
James T Palmer
Michelle Mcnally
Original Assignee
Cytopia Research Pty Ltd
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
Priority claimed from AU2007906387A external-priority patent/AU2007906387A0/en
Application filed by Cytopia Research Pty Ltd filed Critical Cytopia Research Pty Ltd
Publication of WO2009029998A1 publication Critical patent/WO2009029998A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • This invention relates to phenylaminopyrimidine compounds that are retrometabolic drugs or metabolites thereof, pharmaceutical compositions thereof and uses for such compounds and compositions.
  • These compounds are also inhibitors of protein kinases including JAK kinases, in particular JAK2 kinases and can be used in the treatment of kinase associated diseases such as vascular diseases including pulmonary arterial hypertension (PAH) hypertension and ischemia; immunological or inflammatory diseases including asthma and Chronic Obstructive Pulmonary Disease (COPD); and hyperproliferative diseases including cancers for example lung cancer.
  • vascular diseases including pulmonary arterial hypertension (PAH) hypertension and ischemia
  • immunological or inflammatory diseases including asthma and Chronic Obstructive Pulmonary Disease (COPD)
  • COPD Chronic Obstructive Pulmonary Disease
  • cancers for example lung cancer.
  • Pulmonary arterial hypertension is a pulmonary vascular disease affecting the pulmonary arterioles resulting in an elevation in pulmonary artery pressure and pulmonary vascular resistance but with normal or only mildly elevated left-sided filling pressures.
  • PAH is caused by a constellation of diseases that affect the pulmonary vasculature.
  • PAH can be caused by or associated with collagen vascular disorders such as systemic sclerosis (scleroderma), uncorrected congenital heart disease, liver disease, portal hypertension, HIV infection, Hepatitis C, certain toxins, splenectomy, hereditary hemorrhagic teleangiectasia, and primary genetic abnormalities.
  • a mutation in the bone morphogenetic protein type 2 receptor has been identified as a cause of familial primary pulmonary hypertension (PPH). It is estimated that 6% of cases of PPH are familial, and that the rest are "sporadic.” The incidence of PPH is estimated to be approximately 1 case per 1 million population. Secondary causes of PAH have a much higher incidence.
  • the pathologic signature of PAH is the plexiform lesion of the lung which consists of obliterative endothelial cell proliferation and vascular smooth muscle cell hypertrophy in small precapillary pulmonary arterioles. PAH is a progressive disease associated with a high mortality. Patients with PAH may develop right ventricular (RV) failure. The extent of RV failure predicts outcome.
  • RV right ventricular
  • PGI2 may have additional beneficial effects on vascular remodelling. In most cases, incremental dosing is needed because of apparent tachyphylaxis/resistance. The mechanism for this resistance is not known.
  • Selective endothelin type A receptor antagonists are currently in development for the treatment of PAH.
  • Sildenafil a phosphodiesterase type V (PDE-V) inhibitor has recently been approved for the treatment of PAH.
  • PDE-V inhibition results in an increase in cyclic GMP which leads to vasodilation of the pulmonary vasculature.
  • Treprostinil an analogue of PGI2
  • Iloprost another prostacyclin analogue
  • PPH primary pulmonary hypertension
  • idiopathic PAH scleroderma
  • mixed connective tissue disease systemic lupus erythematosus
  • HIV infection toxins such as phentermine/fenfluramine, congenital heart disease, Hepatitis C, liver cirrhosis, chronic thrombo-embolic pulmonary artery hypertension (distal or inoperable), hereditary hemorrhagic teleangiectasia, and splenectomy.
  • JAKs are kinases which phosphorylate a group of proteins called Signal Transduction and Activators of Transcription or STATs. When phosphorylated, STATs dimerize, translocate to the nucleus and activate expression of genes which lead to proliferation of endothelial cells and smooth muscle cells, and cause hypertrophy of cardiac myocytes.
  • STATs Signal Transduction and Activators of Transcription or STATs.
  • STATs dimerize, translocate to the nucleus and activate expression of genes which lead to proliferation of endothelial cells and smooth muscle cells, and cause hypertrophy of cardiac myocytes.
  • JAKl a group of proteins
  • JAK2 Another protein with high homology to JAKs is designated Tyk2.
  • Tyk2 Another protein with high homology to JAKs is designated Tyk2.
  • pulmonary arterial endothelial cells PAECs
  • a promitogenic agent or protein is an agent or protein that induces or contributes to the induction of cellular proliferation. Therefore, one effect of JAK2 inhibition would be to decrease proliferation of endothelial cells or other cells, such as smooth muscle cells.
  • a contemplated effect of a JAK2 inhibitor would be to decrease the proliferation of endothelial cells or other cells which obstruct the pulmonary arteriolar lumen. By decreasing the obstructive proliferation of cells, a JAK2 inhibitor could be an effective treatment of PAH.
  • Potent and specific inhibitors of JAK2 will also be useful in vascular disease (additionally to those described above) such as hypertension, hypertrophy, cardiac ischemia, heart failure (including systolic heart failure and diastolic heart failure), migraine and related cerebrovascular disorders, stroke, Raynaud's phenomenon, POEMS syndrome, Prinzmetal's angina, vasculitides, such as Takayasu's arteritis and Wegener's granulomatosis, peripheral arterial disease, heart disease and PAH.
  • vascular disease such as hypertension, hypertrophy, cardiac ischemia, heart failure (including systolic heart failure and diastolic heart failure), migraine and related cerebrovascular disorders, stroke, Raynaud's phenomenon, POEMS syndrome, Prinzmetal's angina, vasculitides, such as Takayasu's arteritis and Wegener's granulomatosis, peripheral arterial disease, heart disease and PAH.
  • Hypertension is a disease largely of the vasculature with consequences including hypertrophy, heart failure, coronary heart disease and aortic disease. Hypertension usually occurs as result of resistance in the blood vessels to blood flow. The resistance may, for example, be due to structural or functional changes to the blood vessels (eg atherosclerosis, arteriolosclerosis and arteriolitis). Hypertension can be defined by an elevated diastolic and/or systolic blood pressure.
  • Hypertrophy is the enlarging of an organ, myocardial hypertrophy, for example, is enlarging of the heart which may be due to myocardial valve damage or hypertension. Hypertrophic damage may lead, for example, to myocardial infarction, congestive heart failure and cardiomyopathy.
  • Ischemia is the deficiency of an organ in oxygenated blood.
  • the deficiency of oxygenated blood may, for example, be due to functional constriction or obstruction of a blood vessel.
  • Ischemic heart disease is often caused by a reduction in coronary blood flow relative to myocardial demand. The reduction in blood flow may occur for a number of reasons, and typically occurs as a result of artherosclerosis.
  • Heart Failure is a clinical syndrome resulting from disturbances in cardiac output or from increased venous pressure.
  • the disturbances in cardiac output or increases in venous pressure may be due to dilated cardio myopathy, myocardial fibrosis, deposition of an amyloid, constrictive periocarditis, hypertension, hypertrophy and/or ischemia.
  • JAK/STAT pathway has been implicated in a number of other diseases.
  • a review of the JAK/STAT literature offers strong support to the hypothesis that this pathway is important for the recruitment and marshalling of the host immune response to environmental insults, such as viral and bacterial infection.
  • inhibitors of JAKs could be used for immunological and inflammatory diseases including organ transplants, asthma and Chronic Obstructive Pulmonary Disease as well as autoimmune diseases such as systemic lupus erythematosus, mixed connective tissue disease, scleroderma, autoimmune vasculitides, multiple sclerosis, rheumatoid arthritis, Crohns disease, Type I diabetes and autoimmune thyroid disorders.
  • autoimmune diseases such as systemic lupus erythematosus, mixed connective tissue disease, scleroderma, autoimmune vasculitides, multiple sclerosis, rheumatoid arthritis, Crohns disease, Type I diabetes and autoimmune thyroid disorders.
  • Asthma is a complex disorder characterized by local and systemic allergic inflammation and reversible airway obstruction. Asthma symptoms, especially shortness of breath, are a consequence to airway obstruction, and death is almost invariably due to asphyxiation.
  • Airway Hyper Responsiveness (AHR), and mucus hyper secretion by goblet cells are two of the principle causes of airway obstruction in asthma patients. Intriguingly recent work in animal experimental models of asthma has underscored the importance of IL- 13 as a key player in the pathology of asthma.
  • IL- 13 acts independently of IL-4 and may be capable of inducing the entire allergic asthma phenotype, without the induction of IgE (i.e. in a non-atopic fashion).
  • This and other models have pointed to an important second tier mechanism for elicitating the pathophysiology of asthma, that is not dependent on the production of IgE by resident B-cells or the presence of eonisophils.
  • a direct induction of AHR by IL- 13 represents an important process that is likely to be an excellent target for intervention by new therapies.
  • a contemplated effect of a JAK2 inhibitor to the lungs would result in the suppression of the local release of IL- 13 mediated IgE production, and therefore reduction in histaminine release by mast cells and eosinophils.
  • This and other consequences of the absence of IL- 13 indicate that many of the effects of asthma may be alleviated through administration of a JAK2 inhibitor to the lungs.
  • COPD Chronic Obstructive Pulmonary Disease
  • Current clinical guidelines define COPD as a disease state characterized by airflow limitation which is not fully reversible. The airflow limitation is usually both progressive and associated with an abnormal inflammatory response of the lungs to noxious particles and gases, particularly cigarette smoke and pollution.
  • IL- 13 and COPD Several studies have pointed to an association between increased production of IL- 13 and COPD, lending support to the proposition that the potential alleviation of asthma symptoms by use of a JAK2 inhibitor, may also be achieved in COPD.
  • COPD patients have a variety of symptoms including cough, shortness of breath, and excessive production of sputum.
  • COPD includes several clinical respiratory syndromes including chronic bronchitis and emphysema.
  • Chronic bronchitis is a long standing inflammation of the bronchi which causes increased production of mucus and other changes. The patient's symptoms are cough and expectoration of sputum. Chronic bronchitis can lead to more frequent and severe respiratory infections, narrowing and plugging of the bronchi, difficult breathing and disability.
  • Emphysema is a chronic lung disease which affects the alveoli and/or the ends of the smallest bronchi.
  • the lung looses its elasticity and therefore these areas of the lungs become enlarged. These enlarges areas trap stale air and do not effectively exchange it with fresh air. This results in difficult breathing and may result in insufficient oxygen being delivered to the blood.
  • the predominant symptom in patients with emphysema is shortness of breath.
  • the central role played by the JAK family of protein tyrosine kinases in the cytokine dependent regulation of both proliferation and end function of several important cell types indicates that agents capable of inhibiting the JAK kinases are useful in the prevention and chemotherapeutic treatment of disease states dependent on these enzymes.
  • JAK2 inhibitors will also be useful in myeloproliferatve disorders (MPD) such as polycythemia vera (PCV).
  • MPD myeloproliferatve disorders
  • PCV polycythemia vera
  • JAK kinase inhibitors for the treatment of viral diseases and metabolic diseases is indicated.
  • the consequence of the wide ranging systemic actions of the JAK2, and the consequential systemic effect of a JAK2 inhibitor means that where it is desired to target a localised disorder such as cardiovascular disease, PAH, asthma, COPD, or lung cancer it is highly desirable to deliver a JAK2 inhibitor to the organ in a manner that does not enable significant quantities of the active drug to enter other organs.
  • a localised disorder such as cardiovascular disease, PAH, asthma, COPD, or lung cancer
  • Topical administration of drugs has typically been applied to lung and airway disorders, eye diseases and skin disorders.
  • skin disorders direct topical application of a drug which does not have a high degree of transdermal penetration is a suitable solution.
  • eye diseases unique problems are encountered in delivering drugs as the eyelids, tear- flow and cornea itself act as an effective protection system for the eye from foreign matter.
  • systemically administered medications reach the eye in very small quantites due to the low permeability of the blood-retinal barrier.
  • a type of therapy in which an active drug is metabolised into an inactive molecule after leaving the site of therapeutic need can be used to treat lung disorders.
  • Such therapies are known as soft drugs, ante drugs or retrometabolic drugs.
  • Retrometabolic drugs can be distinguished from prodrugs as the prodrug concept is the administration of an inactive molecule, which is systemically activated, compared with a soft drug where the compound administered is active, but is rapidly metabolised into an inactive molecule once it leaves the site of required activity.
  • the molecule is desirably relatively stable in the target tissue, but readily and simply degradable to an
  • Soft drugs can themselves be formulated as prodrugs, which are enzymatically activated in vivo and then subsequently enzymatically deactivated.
  • the challenges in developing a therapeutically appropriate JAK2 inhibitor for use in treatment of PAH include developing an active drug which specifically inhibits JAK2 and which has metabolic weak points resulting in a rapid and predictable degradation to an inactive metabolite.
  • the compound is required to be relatively stable in the target tissue, and degradation ideally occurs at a site distant to the required target.
  • R 1 is independently selected from halogen, R 2 , OR 2 , R 4 , CN, NO 2 , R 2 R 4 , SO 2 R 4 , NR 2 SO 2 R 3 , COR 4 , NR 2 COR 3 , NR 2 COR 4 , R 2 CN, R 2 OH 5 R 2 OR 3 and OR 5 R 4 ;
  • R 2 is substituted or unsubstituted C 1-6 alkyl or C 1-6 alkylene;
  • R 3 is R 2 or substituted or unsubstituted aryl
  • R 4 is selected from NHR 2 , N(R 2 ) 2 , morpholino, thiomorpholino, thiomorpholino- 1- oxide, thiomorpholino- 1,1 -dioxide, NR 2 -piperazine, 4-hydroxy piperidine, 3-hydroxy pyrrolidine, 3-hydroxypyrrole or piperidine, each of which may be optionally substituted with C 1-8 alkyl wherein up to 4 carbon atoms are optionally replaced with CO, O, S, C(O)S, C(O)O or NR Y and/or optionally substituted with halogen, C 4-10 lactone, COSR Y or COOR Y ;
  • R 5 is substituted or unsubstituted C 2-4 alkylene
  • R 6 , R 7 , R 8 , R 9 and R 10 are independently selected from H, R X CN, halogen, substituted or unsubstituted C 1-6 alkyl, NR Y SO 2 R Y and SO 2 N(R Y ) 2 ; where R 7 and R 8 are optionally joined with the carbon atoms to which they are attached to form a C 4-6 substituted or unsubstituted aryl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocyclyl;
  • R ⁇ is H or substituted or unsubstituted d- ⁇ alkyl
  • R x is C 1-6 alkyl wherein up to 3 carbon atoms are optionally replaced with CO
  • R 11 is selected from H, halogen, C 1-6 alkyl , C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkyl, N(R Y ) 2 , NHR Y , COR Y NO 2 , C00R ⁇ , CON(R Y ) 2 , OC 1-6 alkyl, CN, CH 2 F, CHF 2 and CF 3 ;
  • R 12 is H, P(O)(OR 2 ) 2 , OR 13 , COOR 13 , COSR 13 , CONHR 4 or CON(R 4 ) 2 ;
  • R 13 is selected from H, C 1-6 alkyl, C 3-6 cycloalkyl or heterocyclyl, each of which may be optionally substituted with halogen, CN, C 1-6 alkyl OC 1-6 alkyl, or heterocyclyl; m is 1 to 3; and n is 1 or 2, wherein the heterocyclyl contains 1 to 3 heteroatoms independently selected from O, N and S; with the proviso that when R is H, then R is morpholino, thiomorpholino, thiomorpholino-1 -oxide, thiomorpholino- 1,1 -dioxide, NR 2 -piperazine, 4-hydroxy piperidine, 3-hydroxy pyrrolidine, 3-hydroxypyrrole or piperidine, each of which is substituted with at least one C 1-8 alkyl wherein 1 to 4 carbon atoms are replaced with CO, O, S, C(O)S or C(O)O and/or optionally substituted with halogen, C 4-10 lactone, COSR
  • R 11 and n are as defined above and X is a leaving group with compounds of formulae III and IV
  • R 1 , R 6 -R 10 , R 12 and m are as defined above and M is a metal.
  • the compounds of formula I are retrometabolic drugs or metabolites thereof, and kinase inhibitors, preferably JAK inhibitors, more preferably JAK2 inhibitors. These compounds are useful in the treatment of a kinase associated disease, preferably a JAK kinase associated disease such as vascular diseases including cardiovascular diseases and pulmonary vascular diseases, for example, PAH; immunological diseases such as asthma and COPD; and hyperproliferative diseases such as lung cancer.
  • a kinase associated disease preferably a JAK kinase associated disease
  • vascular diseases including cardiovascular diseases and pulmonary vascular diseases, for example, PAH
  • immunological diseases such as asthma and COPD
  • hyperproliferative diseases such as lung cancer.
  • a retrometabolic drug or metabolites thereof comprising the compound of formula I defined above.
  • a retrometabolic drug or metabolites thereof comprising the compound of formula I defined above.
  • a kinase inhibitor comprising the compound formula I defined above.
  • a kinase inhibitor as a retrometabolic drug.
  • the compound of formula I may also be administered in the form of a pharmaceutical composition together with a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising the compound of formula I defined above and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition also comprises one or more additional therapeutic agents.
  • the compound of formula I may be contained within or attached to an implant, such as a drug eluting stent.
  • an implant such as a drug eluting stent.
  • the compound when the compound is used for the treatment of PAH, the compound may be contained within or attached to a pulmonary artery stent, which may act locally, or be released from the stent into the pulmonary circulation where the compound exerts its therapeutic activity in the pulmonary vasculature.
  • the compound of formula I may be contained within a pressurised pack with a suitable propellant.
  • an inhaler for oral inhalation such as a "Diskhaler"
  • an implant which comprises the compound of formula I defined above.
  • a method for the treatment of a kinase associated disease such as a vascular or immunological disease which comprises administering an effective amount of the compound of formula I or a pharmaceutical composition defined above to a subject in need thereof.
  • use of the compound of formula I or a pharmaceutical composition as defined above in the manufacture of a medicament for use in the treatment of a kinase associated disease state such as a vascular, hyperproliferative disease or immunological or inflammatory disease.
  • a kinase associated disease state such as a vascular, hyperproliferative or immunological or inflammatory disease.
  • a method for the treatment of a kinase associated disease such as a vascular or immunological or inflammatory disease which comprises administering an effective amount of a kinase inhibitor which acts as a retrometabolic drug to a subject in need thereof.
  • a kinase inhibitor which acts as a retrometabolic drug in the treatment of a kinase associated disease such as a vascular, hyperproliferative or immunological or inflammatory disease.
  • a kinase associated disease a kinase inhibitor which acts as a retrometabolic drug for use in the treatment of a kinase associated disease such as a vascular, hyperproliferative or immunological or inflammatory disease.
  • Figure 1 shows a Western Blot of incremental concentrations of compound 29 incubated with rat lung microvascular endothelial cells with IL-6 at 10 ng/ml and the STAT3 phosphorylation at tyrosine 705 was determined compared with total STAT3 (+positive control; - negative control; v vehicle); and
  • Figure 2 shows a graph of the results from duplicate experiments described for Figure 1 , comparing the effect of concentration of compound 29 on IL-6 induced phosphorylation of STAT3.
  • the present invention relates to compounds of formula I that inhibit kinases, in particular JAK kinases such as JAK2 kinases and are useful in the treatment of kinase associated diseases such as vascular diseases, hyperproliferative diseases or immunological diseases.
  • JAK kinases such as JAK2 kinases
  • kinase associated diseases such as vascular diseases, hyperproliferative diseases or immunological diseases.
  • R 1 is independently selected from halogen, R 2 , OR 2 , R 4 , CN, NO 2 , R 2 R 4 , SO 2 R 4 ,
  • R 2 is substituted or unsubstituted C 1-6 alkyl or C 1-6 alkylene;
  • R 3 is R 2 or substituted or unsubstituted aryl;
  • R 4 is selected from NHR 2 , N(R 2 ) 2 , morpholino, thiomorpholino, thiomorpholino-1- oxide, thiomorpholino- 1 , 1 -dioxide, NR 2 -piperazine, 4-hydroxy piperidine, 3-hydroxy pyrrolidine, 3-hydroxypyrrole or piperidine, each of which may be optionally substituted with C 1-8 alkyl wherein up to 4 carbon atoms are optionally replaced with CO, O, S, C(O)S, C(O)O or NR Y and/or optionally substituted with halogen, C 4-10 lactone, COSR Y or C00R ⁇ ; R 5 is substituted or unsubstituted C 2-4 alkylene;
  • R 6 , R 7 , R 8 R 9 and R 10 are independently selected from H, R X CN, halogen, substituted or unsubstituted Ci. 6 alkyl, NR Y SO 2 R Y and SO 2 N(R Y ) 2 ; where R 7 and R 8 are optionally joined with the carbon atoms to which they are attached to form a C 4-6 substituted or unsubstituted aryl, substituted or unsubstituted cycloalkyl or substituted or unsubstituted heterocyclyl;
  • R ⁇ is H or substituted or unsubstituted Cj-ealkyl
  • R x is C 1-6 alkyl wherein up to 3 carbon atoms are optionally replaced with CO,
  • R 13 is selected from H, C 1-6 alkyl, C 3-6 cycloalkyl or heterocyclyl, each of which may be optionally substituted with halogen, CN, C] -6 alkyl OC 1-6 alkyl, or heterocyclyl; m is 1 to 3; and n is 1 or 2, 5 wherein the heterocyclyl contains 1 to 3 heteroatoms independently selected from
  • R 1 is morpholino, thiomorpholino, thiomorpholino-1 -oxide, thiomorpholino- 1, 1 -dioxide, NR 2 -piperazine, 4-hydroxy piperidine, 3-hydroxy pyrrolidine, 3-hydroxypyrrole or piperidine, each of which is O substituted with at least one C 1-S alkyl wherein 1 to 4 carbon atoms are replaced with CO,
  • the at least one substituent on morpholino, thiomorpholino, thiomorpholino-1 -oxide, thiomorpholino-1, 1 -dioxide, NR 2 -piperazine, 4-hydroxy piperidine, 3-hydroxy pyrrolidine, 3-hydroxypyrrole or piperidine contains at least one C(O)S, C(O)O, halogen, C 4-10 lactone, COSR Y or C00R ⁇ group.
  • R 1 is preferably morpholino or piperidine substituted with Ci- ⁇ alkyl, wherein up to 4 carbon atoms are optionally replaced with CO, O, S, C(O)S, C(O)O or NH and/or optionally substituted with F, C 4-10 lactone, COSC 1-4 alkyl or CO 2 C 1-4 alkyl.
  • R 1 is in the 4-position.
  • R 6 , R 9 and R 10 are preferably H.
  • R 7 is preferably SO 2 NHR Y or NHS0 2 R Y in which R ⁇ is C 1-6 alkyl.
  • R 8 is preferably R X CN in which R x is C 1-6 alkyl wherein up to 2 atoms can be optionally replaced with CO, NR Y SO and/or substituted with C]. 6 alkyl or NHSO 2 R Y wherein R ⁇ is H or C 1-6 alkyl. Alternatively, R 7 and R 8 are optionally joined together with carbon atoms to which they are attached to form optionally substituted heterocyclyl.
  • R 11 is preferably methyl or trifluromethyl and is in the 5 position.
  • R 12 is preferably in the 3 or 5 position and is OR 13 ,COOR 13 , COSR 13 or P(O)(OR 2 ) 2 wherein R 2 is C ⁇ alkyl and R 13 is H, C 1-6 alkyl, C 3-6 cycloalkyl or heterocyclyl each of which may be optionally substituted with halogen, C 1-6 alkyl or OC 1-6 alkyl.
  • R 1 is morpholino or piperidine substituted with C ⁇ alkyl, wherein up to 4 carbon atoms are optionally replaced with CO, O, S, C(O)S, C(O)O or NH and/or optionally substituted with F, C 4-10 lactone, COSC 1-4 alkyl or CO 2 C 1-4 alkyl;
  • R 7 is S0 2 NHR ⁇ or NHSO 2 R Y ;
  • R ⁇ is H or C 1-6 alkyl
  • R 8 is R X CN wherein R x is C 1-6 alkyl wherein up to 2 atoms can be optionally replaced with CO, NR Y S0 and/or substituted with C 1-6 alkyl or NHSO 2 R Y ; or
  • R 11 is C ⁇ alkyl or trifluoromethyl
  • R 12 is OR 13 , COOR 13 , COSR 13 or P(O)(OR 2 ) 2 wherein R 2 is C 1-4 alkyl and R 13 is H, C 1-6 alkyl, C 3-6 cycloalkyl or heterocyclyl, each of which may be optionally substituted with halogen, C 1-6 alkyl or OC 1-6 alkyl.
  • the compounds of formula I have the formula Ia
  • R' a is morpholino or piperidine substituted with Q-salkyl, wherein up to 4 carbon atoms are optionally replaced with CO, O, S, C(O)S, C(O)O or NH and/or optionally substituted with F, C 4-10 lactone, C0SC 1-4 alkyl or CO 2 C 1-4 alkyl;
  • R 7 a is S0 2 NHR Y or NHS0 2 R Y in which R ⁇ is C 1-6 alkyl
  • R 8 a is R X CN in which R x is C 1-6 alkyl wherein up to 2 atoms can be optionally replaced with CO, NR Y SO and/or substituted with Ci -6 alkyl or NHSO 2 R Y wherein R ⁇ is H or C 1-6 alkyl;
  • R a and R a are optionally joined together with the carbon atoms to which they are attached to form optionally substituted heterocyclyl;
  • R n a is C 1-4 alkyl or trifluoromethyl;
  • R 12 is OR 13 , COOR 13 , COSR 13 or P(O)(OR 2 ) 2 wherein R 2 is C 1-4 alkyl and R 13 is H, Ci- ⁇ alkyl, C 3-6 cycloalkyl or heterocyclyl, each of which may be optionally substituted with halogen, Ci -6 alkyl or OC 1-6 alkyl.
  • the compound is selected from the following: methyl 5-(4-(4-(cyanomethylcarbamoyl)phenyl) ⁇ yrimidin-2-ylamino)-2- morpholinobenzoate; ethyl 5-(4-(4-(cyanomethylcarbamoyl)phenyl)pyrimidin-2-ylamino)-2-morpholinobenzoate; propyl 5-(4-(4-(cyanomethylcarbamoyl)phenyl)pyrimidin-2-ylamino)-2- morpholinobenzoate; isopropyl 5 ⁇ (4 ⁇ (4-(cyanomethylcarbamoyl)phenyl)pyrimidin-2-ylammo)-2- mo ⁇ holinobenzoate; tert-butyl 5-(4-(4-(cyanomethylcarbamoyl) ⁇ henyl)pyrimidin-2-ylamino)-2- morpholinobenzoate ; cyclopropyl 5 ⁇ (
  • C 1-6 alkyl refers to straight chain or branched chain hydrocarbon groups having from 1 to 6 carbon atoms. Examples include ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, neopentyl and hexyl.
  • Ci -6 alkylene refers to the divalent equivalents of Ci- ⁇ alkyl defined above.
  • C 2-6 alkenyl refers to straight chain or branched chain hydrocarbon groups having at least one double bond of either E or Z stereochemistry where applicable and 2 to 6 carbon atoms. Examples include vinyl, 1-propenyl, 1- and 2-butenyl and 2- methyl-2-propenyl .
  • C 2-6 alkynyl refers to straight chain or branched chain hydrocarbon groups having at least one triple bond and 2 to 4 carbon atoms. Examples include ethynyl, 1- or 2-propynyl, 1-, 2- or 3- butynyl and methyl-2-propynyl.
  • C3 -6 cycloalkyl refers to non-aromatic cyclic hydrocarbon groups having from 3 to 6 carbon atoms. Examples include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • aryl refers to single, polynuclear, conjugated or fused residues of aromatic hydrocarbons. Examples include phenyl, biphenyl, terphenyl, quaterphenyl, naphthyl, tetrahydronaphthyl, anthracenyl, dihydroanthracenyl, benzanthracenyl, dibenxanthracenyl and phenanthrenyl.
  • heterocyclyl refers to saturated or unsaturated, monocyclic or polycyclic hydrocarbon groups containing at least one heteroatom atom selected from the group consisting of nitrogen, sulphur and oxygen.
  • Suitable heterocyclyls include N-containing heterocyclic groups, such as, unsaturated 3 to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms, for example, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl or tetrazolyl; saturated 3 to 6-membered heteromonocyclic groups containing 1 to 4 nitrogen atoms, such as, pyrrolidinyl, imidazolidinyl, piperidino or piperazinyl; unsaturated condensed heterocyclic groups containing 1 to 5 nitrogen atoms, such as indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl or tetrazolopyridazin
  • nitrogen atoms such as, benzothiazolyl or benzothiadiazolyl.
  • Preferred heterocyclyls are N-containing heterocyclyls such as morpholino, piperidinyl and l,2-benzisothiazol-3(2H)-one 1,1 -dioxide.
  • halogen refers to fluorine, chlorine, bromine and iodine, preferably fluorine.
  • optionally substituted refers to a group that may or may not be further substituted with one or more groups selected from C 1-6 alkyl, CF 3 , C 3-6 cycloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heterocyclyl, halo, haloC 1-6 alkyl, haloC 3-6 cycloalkyl, haloC 2- ⁇ alkenyl, haloC 2-6 alkynyl, haloaryl, haloheterocyclyl, hydroxy, C 1-6 alkoxy, C 2-6 alkenyloxy, C 2-6 alkynyloxy, aryloxy, heterocyclyloxy, carboxy, haloC 1-6 alkoxy, haloC 2-6 alkenyloxy, haloC 2 - 6 alkynyloxy, haloaryloxy, nitro, nitroC 1-6 ,alkyl, nitroC 2-6 alkenyl
  • Preferred optional substituents are selected from the group consisting of C 1-4 alkyl, CF 3 , C 3-6 cycloalkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heterocyclyl, halo, haloaryl, haloheterocyclyl, hydroxy, C 1-4 alkoxy, aryloxy, carboxy, amino, arylacyl, heterocyclacyl, acylamino, acyloxy, arylsulphonyl and cyano.
  • the leaving group may be any suitable known type such as those disclosed in J. March, "Advanced Organic Chemistry: Reactions, Mechanisms and Structure” 4 th Edition, pp 352-357, John Wiley & Sons, New York, 1992 which is incorporated herein by reference.
  • the leaving group is halogen, more preferably chlorine.
  • the compounds of the invention may also be prepared as salts which are pharmaceutically acceptable, but it will be appreciated that non-pharmaceutically acceptable salts also fall within the scope of the present invention, since these are useful as intermediates in the preparation of pharmaceutically acceptable salts.
  • pharmaceutically acceptable salts include salts of pharmaceutically acceptable cations such as sodium, potassium, lithium, calcium, magnesium, ammonium and alkylammonium; acid addition salts of pharmaceutically acceptable inorganic acids such as hydrochloric, orthophosphoric, sulphuric, phosphoric, nitric, carbonic, boric, sulfamic and hydrobromic acids; or salts of pharmaceutically acceptable organic acids such as acetic, propionic, butyric, tartaric, maleic, hydroxymaleic, fumaric, citric, lactic, mucic, gluconic, benzoic, succinic, oxalic, phenylacetic, methanesulphonic, trihalomethanesulphonic, toluenesulphonic,
  • the salts may be formed by conventional means, such as by reacting the free base form of the compound with one or more equivalents of the appropriate acid in a solvent or medium in which the salt is insoluble, or in a solvent such as water which is removed in vacuo or by freeze drying or by exchanging the anions of an existing salt for another anion on a suitable ion exchange resin.
  • the compound can be used as a purified enantiomer or diastereomer, or as a mixture of any ratio of stereoisomers. It is however preferred that the mixture comprises at least 70%, 80%, 90%, 95%, 97.5% or 99% of the preferred isomer.
  • This invention also encompasses prodrugs of the compounds of formula I.
  • This invention also encompasses methods of treating or preventing disorders that can be treated or prevented by the inhibition of protein kinases, such as JAK comprising administering drugs or prodrugs of compounds of the invention.
  • compounds of formula I having free amino, amido, hydroxy or carboxylic acid groups can be converted into prodrugs.
  • Prodrugs include compounds wherein an amino acid residue, or a polypeptide chain of two or more (eg, two, three or four) amino acid residues which are covalently joined through peptide bonds to free amino, hydroxy and carboxylic acid groups of compounds of the invention.
  • the amino acid residues include the 20 naturally occurring amino acids commonly designated by three letter symbols and also include, 4- hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvlin, beta- alanine, gamma-aminobutyric acid, citrulline, homocysteine, homoserine, ornithine and methioine sulfone.
  • Prodrugs also include compounds wherein carbonates, carbamates, amides and alkyl esters which are covalently bonded to the above substituents of compounds of the present invention through the carbonyl carbon prodrug sidechain.
  • Prodrugs also include phosphate derivatives of compounds (such as acids, salts of acids, or esters) joined through a phosphorus-oxygen bond to a free hydroxyl of compounds of formula I.
  • Prodrugs may also include N-oxides, and S-oxides of the appropriate nitrogen and sulphur atoms in compounds of formula I.
  • Compounds of the general formula I are generally prepared from a dichloropyrimidine .
  • the first step of the process typically begins with a cross-coupling reaction between a dichloropyrimidine and a suitably functionalised coupling partner.
  • Typical coupling partners are organoboronic acids or esters (Suzuki coupling: see for example Miyaura, N. and Suzuki, Chem Rev. 1995, 95 2457), organostannanes (Stille coupling: see for example Stille, J.K., Angew. Chem., Int. Ed. Engl, 1986, 25, 508), Grignard reagents (Kumada coupling: Kumada, M.; Tamao, K.; Sumitani, K. Org. Synth.
  • Suzuki coupling is the preferred coupling method and is typically performed in a solvent such as DME, THF, DMF, ethanol, propanol, toluene, acetonitrile or 1,4-dioxane, with or without added water, in the presence of a base such as sodium or potassium carbonate, lithium hydroxide, caesium carbonate, sodium hydroxide, potassium fluoride or potassium phosphate.
  • a base such as sodium or potassium carbonate, lithium hydroxide, caesium carbonate, sodium hydroxide, potassium fluoride or potassium phosphate.
  • the reaction may be carried out at elevated temperatures and the palladium catalyst employed may be selected from Pd(PPh 3 ) 4 , Pd(OAc) 2 , [PdCl 2 (dppf)], Pd 2 (dba) 3 /P(t-Bu) 3 .
  • the second step of the process involves a nucleophilic aromatic substitution reaction of the derived above with a suitably substituted aniline.
  • the nucleophilic aromatic substitution is typically carried out by addition of the aniline to monohalo heterocyclic intermediate obtained from the first in a solvent such as ethanol, n-propanol, isopropanol, tert-butanol, dioxane, THF, DMF, toluene or xylene.
  • the reaction is typically performed at elevated temperature in the presence of an acid such as HCl or p-toluenesulfonic acid or in the presence of base such as a non-nucleophilic base such as triethylamine or diisopropylethylamine, or an inorganic base such as potassium carbonate or sodium carbonate.
  • an acid such as HCl or p-toluenesulfonic acid
  • base such as a non-nucleophilic base such as triethylamine or diisopropylethylamine, or an inorganic base such as potassium carbonate or sodium carbonate.
  • the aniline substituent may be introduced through a transition metal catalysed amination reaction.
  • Typical catalysts for such transformations include Pd(OAc) 2 / P(t-Bu) 3 , Pd 2 (dba) 3 /BINAP and Pd(OAc) 2 /BINAP.
  • These reactions are typically carried out in solvents such as toluene or dioxane, in the presence of bases such as caesium carbonate or sodium or potassium tert-butoxide at temperatures ranging from room temperature to reflux (e.g. Hartwig, J.F., Angew. Chem. Int. Ed. 1998, 37, 2046).
  • the anilines employed in the first step of the synthesis of these compounds are obtained commercially or are prepared using methods well known to those skilled in the art.
  • reaction step may be further derivatised using techniques known to those skilled in the art.
  • derivatisation of the mono-halo intermediate may be undertaken prior to displacement of the halo substituent.
  • Those skilled in the art will appreciate that the order of the reactions described for the syntheses above may be changed in certain circumstances and that certain functionalities may need to be derivatised (i.e. protected) in certain instances for the reactions described above to proceed with reasonable yield and efficiency.
  • the types of protecting functionality are well-known to those skilled in the art and are described for example in Greene (Greene, T., Wuts, P. (1999) Protective Groups in Organic Synthesis. Wiley-Interscience; 3rd edition.).
  • Retrometabolic drugs can be distinguished from pro-drugs which are administered as inactive drugs and then systematically activated. Retrometabolic drugs are desirably relatively stable in the target tissue, but readily and simply degradable to an inactive drug in other organs or tissues.
  • Retrometabolic drugs can themselves be formulated as prodrugs, which are enzymatically activated in vivo and then subsequently enzymatically deactivated. Without wishing to be bound by any theory, it is believed that the retrometabolic activity of the compounds of formula I is provided by including a substituent on the periphery of the compound which has metabolic weak spot. In a preferred embodiment this is provided by substituents R 1 and/or R 12 which may be attached to the phenyl ring on the right hand side of the compounds of formula I. Specific examples of such substituents include:
  • the compounds of formula I have activity against protein kinases, particularly the
  • JAK kinases and most particularly JAK2 kinases are JAK2 inhibitors.
  • a JAK2 inhibitor is any compound that selectively inhibits the activity of JAK2.
  • One activity of JAK2 is to phosphorylate a STAT protein. Therefore an example of an effect of a JAK2 inhibitor is to decrease the phosphorylation of one or more STAT proteins.
  • the inhibitor may inhibit the phosphorylated form of JAK2 or the non-phosphorylated form of JAK2.
  • the present invention also provides the use of kinase inhibitors such as JAK kinase inhibitors, in particular JAK2 inhibitors as retrometabolic drugs.
  • kinase inhibitors such as JAK kinase inhibitors, in particular JAK2 inhibitors as retrometabolic drugs.
  • compositions comprising at least one of the compounds of the formula I and a pharmaceutically acceptable carrier.
  • the carrier must be “pharmaceutically acceptable” means that it is compatible with the other ingredients of the composition and is not deleterious to a subject.
  • compositions of the present invention may contain other therapeutic agents as described below, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavours, etc.) according to techniques such as those well known in the art of pharmaceutical formulation (See, for example, Remington: The Science and Practice of Pharmacy, 21st Ed., 2005, Lippincott Williams & Wilkins).
  • the compounds of the invention may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, intra(trans)dermal, or intracisternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray or insufflation; topically, such as in the form of a cream or ointment ocularly in the form of a solution or suspension; vaginally in the form of pessaries, tampons or creams; or rectally such as in the form of suppositories; in dosage unit formulations containing nontoxic, pharmaceutically acceptable vehicles or diluents.
  • parenterally such as by subcutaneous, intravenous, intramuscular, intra(trans)dermal, or intracisternal injection or infusion techniques
  • the compounds may, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • compositions for the administration of the compounds of the invention may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. These methods generally include the step of bringing the compound of formula I into association with the carrier which constitutes one or more accessory ingredients.
  • the pharmaceutical compositions are prepared by uniformly and intimately bringing the compound of formula I into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • compositions containing the compound of formula I may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents such as sweetening agents, flavoring agents, coloring agents and preserving agents, e.g. to provide pharmaceutically stable and palatable preparations.
  • Tablets contain the compound of formula I in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated to form osmotic therapeutic tablets for control release.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the compound of formula I is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the compound of formula I is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxy- propylmethylcellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monoo
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n-propyl, p-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • preservatives for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • coloring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • flavoring agents for example ethyl, or n-propyl, p-hydroxybenzoate
  • sweetening agents such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the compound of formula I in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the compound of formula I in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, sorbitol, glycerol, glycerol, glycerol, glycerol, glycerol, glycerol, sorbitol, sorbitol, gly
  • the pharmaceutical compositions of the invention may also be in the form of oil-in- water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally- occurring gums, for example gum acacia or gum tragacanth, naturally-occurring phosphatides, for example soy bean, lecithin, and esters or partial esters derived from fatty acids and hexitol anhydrides, for example sorbitan monooleate, and condensation products of the said partial esters with ethylene oxide, for example poly oxy ethylene sorbitan monooleate.
  • the emulsions may also contain sweetening and flavoring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • the pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectable formulations.
  • the active compound may be administered by any of the methods and formulations employed in the art for administration to the respiratory tract.
  • the active compound may be administered in the form of a solution or a suspension or as a dry powder.
  • Solutions and suspensions will generally be aqueous, for example prepared from water alone (for example sterile or pyrogen-free water) or water and a physiologically acceptable co-solvent (for example ethanol, propylene glycol or polyethylene glycols such as PEG 400).
  • a physiologically acceptable co-solvent for example ethanol, propylene glycol or polyethylene glycols such as PEG 400.
  • Such solutions or suspensions may additionally contain other excipients for example preservatives (such as benzalkonium chloride), solubilising agents/surfactants such as polysorbates (eg. Tween 80, Span 80, benzalkonium chloride), buffering agents, isotonicity-adjusting agents (for example sodium chloride), absorption enhancers and viscosity enhancers.
  • Suspensions may additionally contain suspending agents (for example microcrystalline cellulose and carboxymethyl cellulose sodium).
  • Solutions or suspensions are applied directly to the nasal cavity by conventional means, for example with a dropper, pipette or spray.
  • the formulations may be provided in single or multidose form. In the latter case a means of dose metering is desirably provided.
  • a dropper or pipette this may be achieved by the subject administering an appropriate, predetermined volume of the solution or suspension.
  • a spray this may be achieved for example by means of a metering atomising spray pump.
  • Administration to the respiratory tract may also be achieved by means of an aerosol formulation in which the compound is provided in a pressurised pack with a suitable propellant, such as a chlorofluorocarbon (CFC), for example dichlorodifluoromethane, trichlorofluoromethane or dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant such as a chlorofluorocarbon (CFC), for example dichlorodifluoromethane, trichlorofluoromethane or dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • CFC chlorofluorocarbon
  • the aerosol may conveniently also contain a surfactant such as lecithin.
  • the dose of active compound may be controlled by provision of a metered valve,
  • the active compound may be provided in the form of a dry powder, for example a powder mix of the compound in a suitable powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidine (PVP).
  • a powder base such as lactose, starch, starch derivatives such as hydroxypropylmethyl cellulose and polyvinylpyrrolidine (PVP).
  • PVP polyvinylpyrrolidine
  • the powder carrier will form a gel in the nasal cavity.
  • the powder composition may be presented in unit dose form, for example in capsules or cartridges of eg. gelatin, or blister packs from which the powder may be administered by means of an inhaler.
  • the active compound will generally have a small particle size, for example of the order of 5 microns or less. Such a particle size may be obtained by means known in the art, for example by micronisation.
  • formulations adapted to give sustained release of the active compound may be employed.
  • the active compound may be administered by oral inhalation as a free-flow powder via a "Diskhaler” (trade mark of Glaxo Group Ltd) or a meter dose aerosol inhaler.
  • the compounds of the present invention may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • compositions suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or sprays containing in addition to the active ingredient such carriers as are known in the art to be appropriate.
  • creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention are employed.
  • topical application shall include mouthwashes and gargles.
  • the active compound may be in the form of a solution or suspension in a suitable sterile aqueous or non-aqueous vehicle.
  • Additives for instance buffers, preservatives including bactericidal and fungicidal agents, such as phenyl mercuric acetate or nitrate, benzalkonium chloride, or chlorohexidine and thickening agents such as hypromellose may also be included.
  • the compounds of the present invention can also be administered in the form of liposomes.
  • liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multilamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolisable lipid capable of forming liposomes can be used.
  • the present compositions in liposome form can contain, in addition to a compound of the present invention, stabilisers, preservatives, excipients and the like.
  • the preferred lipids are the phospholipids and phosphatidyl cholines, both natural and synthetic. Methods to form liposomes are known in the art.
  • Efficacy of this class of compounds may be applicable to drug eluting stents.
  • Potential applications of drug eluting stents with these compounds include pulmonary artery stenosis, pulmonary vein stenosis, as well as coronary artery stenosis.
  • Drug eluting stents may also be used in saphenous vein grafts or arterial grafts or conduits.
  • Drug eluting stents that release this class of compounds may also be applicable for treating stenoses of the aorta or peripheral arteries, such as the iliac artery, the femoral artery or the popliteal artery.
  • the compound may be bound to the drug eluting stent by any of various methods known in the field. Examples of such methods include polymers, phosphoryl choline, and ceramics.
  • the compound may also be impregnated into a bioabsorbable stent.
  • the active compounds may also be presented for use in the form of veterinary compositions, which may be prepared, for example, by methods that are conventional in the art.
  • veterinary compositions include those adapted for:
  • oral administration external application, for example drenches (e.g. aqueous or nonaqueous solutions or suspensions); tablets or boluses; powders, granules or pellets for admixture with feed stuffs; pastes for application to the tongue;
  • drenches e.g. aqueous or nonaqueous solutions or suspensions
  • tablets or boluses e.g. aqueous or nonaqueous solutions or suspensions
  • pastes for application to the tongue for example drenches (e.g. aqueous or nonaqueous solutions or suspensions); tablets or boluses; powders, granules or pellets for admixture with feed stuffs; pastes for application to the tongue;
  • parenteral administration for example by subcutaneous, intramuscular or intravenous injection, e.g. as a sterile solution or suspension; or (when appropriate) by intramammary injection where a suspension or solution is introduced in the udder via the teat;
  • topical applications e.g. as a cream, ointment or spray applied to the skin;
  • compositions and method of the present invention may further comprise other therapeutically active compounds as noted herein which are usually applied in the treatment of the above mentioned pathological conditions. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles.
  • the combination of therapeutic agents may act synergistically to effect the treatment or prevention of the various disorders described above. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
  • Examples of other therapeutic agents include the following: endothelin receptor antagonists (eg ambrisentan, bosentan, sitaxsentan), PDE-V inhibitors (eg sildenafil, tadalafil, vardenafil), Calcium channel blockers (eg amlodipine, felodipine, varepamil, diltiazem, menthol), prostacyclin, treprostinil, iloprost, beraprost, nitric oxide, oxygen, heparin, warfarin, diuretics, digoxin, cyclosporins (e.g., cyclosporin A), CTLA4-Ig, antibodies such as ICAM-3, anti-IL-2 receptor (Anti-Tac), anti-CD45RB, anti-CD2, anti-CD3 (OKT-3), anti-CD4, anti-CD80 5 anti-CD86, agents blocking the interaction between CD40 and gp39, such as antibodies specific for CD40 and/or gp39
  • the compounds of formula I may be used in the treatment of kinase associated diseases including JAK kinase associated diseases such as vascular or immunological diseases.
  • treatment means affecting a subject, tissue or cell to obtain a desired pharmacological and/or physiological effect and include: (a) preventing the disease from occurring in a subject that may be predisposed to the disease, but has not yet been diagnosed as having it; (b) inhibiting the disease, i.e., arresting its development; or (c) relieving or ameliorating the effects of the disease, i.e., cause regression of the effects of the disease.
  • subject refers to any animal having a disease which requires treatment with the compound of formula I.
  • mammals including, but not limited to, cows, sheep, goats, horses, dogs, cats, guinea pigs, rats or other bovine, ovine, equine, canine, feline, rodent or murine species can be treated.
  • the invention can also be practiced in other species, such as avian species (e.g., chickens).
  • avian species e.g., chickens.
  • administering should be understood to mean providing a compound of the invention to a subject in need of treatment.
  • kinase associated diseases refers to a disorder or disorders that directly or indirectly result from or are aggravated by aberrant kinase activity, in particular JAK activity and/or which are alleviated by inhibition of one or more of these kinase enzymes.
  • the kinase associated disease state involves one or more of the JAK kinases, JAKl, JAK2, JAK3 or TYK2.
  • the diseases involves JAK2 kinase.
  • Such diseases include, but are not limited to, those listed in the Table below. Activation of the JAK/STAT pathway in various pathologies
  • immunological and inflammatory disease refers to an immunological, inflammatory or autoimmune disease, including but not limited to rheumatoid arthritis, polyarthritis, rheumatoid spondylitis, osteoarthritis, gout, asthma, bronchitis, allergic rhinitis, chronic obstructive pulmonary disease, cystic fibrosis, inflammatory bowl disease, irritable bowl syndrome, mucous colitis, ulcerative colitis, diabrotic colitis, Crohn's disease, autoimmune thyroid disorders , gastritis, esophagitis, hepatitis, pancreatitis, nephritis, psoriasis, eczema, acne vulgaris, dermatitis, hives, multiple sclerosis, Alzheimer's disease, Motor Neurone Disease (Lou Gehrig's disease), disease, Paget's disease, sepsis, conjunctivitis, neranl catarrh,
  • hypoproliferative diseases includes cancer and myeloproliferative disease states such as cellular-proliferative disease states, including but not limited to: Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hanlartoma, inesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lympho
  • vascular disease refers to diseases including but not limited to cardiovascular diseases, hypertension, hypertrophy, hypercholesterolemia, hyperlipidemia, thrombotic disorders, stroke, Raynaud's phenomenon, POEMS syndrome, angina, ischemia, migraine, peripheral arterial disease, heart failure, restenosis, atherosclerosis, left ventricular hypertrophy, myocardial infarction, ischemic diseases of heart, kidney, liver and brain, and pulmonary arterial hypertension.
  • Preferred diseases for JAK2 selective inhibitors include immunological and inflammatory diseases such as asthma, COPD, auto-immune diseases such as atopic dermatitis, asthma, rheumatoid arthritis, Crohn's disease, psoriasis, Crouzon syndrome, achondroplasia, systemic lupus erythematosus, scleroderma, mixed connective tissue disease, vasculitis, thanatophoric dysplasia and diabetes; vascular diseases such as hypertension, hypertrophy, stroke, Raynaud's phenomenon, POEMS syndrome, angina, ischemia, migraine, peripheral arterial disease, heart failure, restenosis, atherosclerosis and pulmonary arterial hypertension; hyperproliferative disorders such as cancer for example prostate cancer, colon cancer, breast cancer, head and neck cancer, leukemia and lymphoma.
  • the compounds of the present invention or metabolites thereof and kinase inhibitors in general are also retrometabolic drugs they are particularly useful in a the treatment
  • terapéuticaally effective amount refers to the amount of the compound of formula I that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician.
  • an appropriate dosage level will generally be about 0.01 to 500 mg per kg patient body weight per day which can be administered in single or multiple doses.
  • the dosage level will be about 0.1 to about 250 mg/kg per day; more preferably about 0.5 to about 100 mg/kg per day.
  • a suitable dosage level may be about 0.01 to 250 mg/kg per day, about 0.05 to 100 mg/kg per day, or about 0.1 to 50 mg/kg per day. Within this range the dosage may be 0.05 to 0.5, 0.5 to 5 or 5 to 50 mg/kg per day.
  • compositions are preferably provided in the form of tablets containing 1.0 to 1000 milligrams of the active ingredient, particularly 1.0, 5.0, 10.0, 15.0. 20.0, 25.0, 50.0, 75.0, 100.0, 150.0, 200.0, 250.0, 300.0, 400.0, 500.0, 600.0, 750.0, 800.0, 900.0, and 1000.0 milligrams of the active ingredient.
  • the dosage may be selected, for example to any dose within any of these ranges, for therapeutic efficacy and/or symptomatic adjustment of the dosage to the patient to be treated.
  • the compounds will preferably be administered on a regimen of 1 to 4 times per day, preferably once or twice per day.
  • the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • the compounds of the invention may be prepared by methods well known to those skilled in the art, and as described in the synthetic and experimental procedures shown below for selected compounds. Definition of Abbreviations:
  • iV-(4-(2-chloropyrimidm-4-yl)phenyl)methanesulfonamide 750 mg 2.64 mmol
  • potassium carbonate 730 mg, 2 eq
  • the reaction mixture was stirred for several minutes before adding bromoacetonitrile (368 ⁇ L, 2 eq).
  • the reaction mixture was then stirred at room temperature for 48 hour.
  • the crude reaction mixture was then concentrated in vacuo and the residue taken up in ethyl acetate (200 mL) and washed with water (2 x 100 mL), brine (100 mL) and then dried (Na 2 SO 4 ).
  • Intermediate 6 can be prepared from 4-bromosaccharin via conversion to the corresponding pinacol boronate and subsequent Suzuki coupling (analogous to that described above) as outlined in the general scheme below.
  • isothiazol-3-one may be alkylated using appropriate alkyl halides and procedures similar to those described above for alkylation of N-(4-(2-chloropyrimidin-4-yl)phenyl)methanesulfonamide (Intermediate 4).
  • anilines which cannot be purchased from commercial sources can generally be prepared by reaction of appropriate secondary amines to the required nitrofluorobenzene and subsequent reduction of the nitro moeity.
  • Elaborated anilines may be coupled to the chlorides described above under acidic conditions (as per example 1) or Buchwald-Hartwig coupling conditions.
  • anilines may be coupled using the described procedures and then further elaborated subsequently using methods familiar to those skilled in the art (as indicated in various examples below).
  • Ethyl isonipecoate (764 ⁇ L, 4.96 mmol), l-fluoro-4-nitrobenzene (1.03 g, 6.55 mmol, 1.3 eq) and potassium carbonate (1.42 g, 10.27 mmol, 2.1 eq) were placed in a round bottomed flask, suspended in 1,4-dioxane (15 mL) and heated at 100°C under a nitrogen atmosphere for 108 hours. The reaction mixture was diluted with water (40 mL) and allowed to stand at room temperature. The product solidified and was collected by filtration and washed with diethyl ether to remove the excess l-fluoro-4-nitrobenzene starting material.
  • Ethyl 1- (4-nitrophenyl)piperidine-4-carboxylate was thus obtained as a yellow solid (582 mg, 42% yield) and used without further purification.
  • a round bottomed flask was charged with ethyl l-(4-nitrophenyl)piperidine-4-carboxylate (582 mg, 2.09 mmol) in 1:1 ethyl acetate methanol (20 mL : 20 mL).
  • the reaction vessel was evacuated and backfilled with nitrogen three times before adding 10% Pd/C (29 mg, 5% w/w).
  • the reaction vessel was again evacuated and backfilled with nitrogen three times.
  • the reaction was then placed under a hydrogen balloon and stirred at room temperature overnight.
  • the reaction vessel was evacuated and backfilled with nitrogen three times before adding 10% Pd/C (27 mg). The reaction vessel was again evacuated and backfilled with nitrogen three times. The reaction was then placed under a hydrogen balloon and stirred at room temperature overnight. The hydrogen balloon was then replaced and the mixture stirred at room temperature for a further 3 h. The reaction mixture was then filtered through Celite and concentrated in vacuo to afford the desired aniline as a beige solid. Formation of the crude product was confirmed by LC-MS and the product was then used without further purification or characterization. LC-MS: rt 0.89 min m/z 219.4 [M-H] ' . eneral scheme for preparation of fluoromethylthioesters
  • N-(4-(2-chloropyrimidin-4-yl)phenyl)methanesulfonamide 50 mg, 0.18 mmol
  • (l-(4- aminophenyl)piperidin-4-yl)methanol 54 mg, 0.26 mmol, 1.4 eq
  • />-toluene sulfonic acid monohydrate 32 mg, 0.95 eq
  • dioxane 4 mL
  • the reaction mixture was then cooled to room temperature and concentrated in vacuo.
  • the residue was taken up in ethyl acetate (30 mL) and water (10 mL). The two phases were mixed and separated.
  • the flask was again evacuated and backfilled with nitrogen six times before being placed to the level of the solvent in a thermostatted oil bath heated to 100°C. A blast shield was placed around the reaction and it was stirred at this temperature for 48 hrs before being allowed to cool to room temperature.
  • the reaction mixture was diluted with ethyl acetate (30 mL) and filtered through a sintered funnel. The residue was washed with ethyl acetate (30 mL) and diethyl ether (30 mL) and the combined organics were evaporated to dryness in vacuo to give the crude product which was purified by trituration with DCMMeOH 90:10 and subsequent washing with DCM or alternatively by silica column chromatography.
  • iV-(4-(2-Chloropyrimidin-4-yl)phenyl)methanesulfonamide 151 mg, 0.53 mmol
  • 5-amino- 2-morpholinobenzoic acid 142 mg, 0.64 mmol, 1.2 eq
  • j»-toluene sulphonic acid monohydrate 98 mg, 0.97 eq
  • the reagents were dissolved in 1,4-dioxane (8 mL) and the reaction mixture was heated at 100 0 C overnight.
  • iV-(4-(2-chloropyrimidin-4-yl)phenyl)methanesulfonamide (77 mg, 0.27 mmol), 5-amino- 2-morpholinobenzoic acid (73 mg, 0.33 mmol, 1.2 eq) and /Holuene sulphonic acid monohydrate (49 mg, 0.97 eq) were all placed in a round bottomed flask fitted with a condenser and CaCl 2 drying tube.
  • the reagents were dissolved in 1,4-dioxane (4 mL) and absolute ethanol (2 mL) and the reaction mixture was heated at 12O 0 C overnight. The reaction mixture was then cooled to room temperature, concentrated in vacuo.
  • iV-(4-(2-chloropyrimidin-4-yl)phenyl)methanesulfonamide 755 mg, 2.66 mmol
  • 5-amino- 2-morpholinobenzoic acid 714 mg, 3.21 mmol, 1.2 eq
  • /7-toluene sulphonic acid monohydrate 490 mg, 2.57 mmol , 0.97 eq
  • 1,4-Dioxane 40 mL was added and the reaction mixture was heated at 100 0 C for 72 h.
  • n-Propanol (15 mL) was added to the reaction mixture and the reaction continued at 12O 0 C for a further 48 h (note: power was cut to reaction for ⁇ 16 hours during this time) .
  • the reaction mixture was then allowed to stir at room temperature overnight.
  • the resulting precipitate was collected by filtration and the crude product was then washed with methanol ( ⁇ 3 rnL) and dried under reduced pressure.
  • the resulting solid was then taken up in ethyl acetate (150 niL) and washed with water (50 mL).
  • the organic phase was dried (Na 2 SO 4 ) and concentrated in vacuo to afford the propyl ester product (447 mg, yellow solid) in sufficient purity for subsequent reactions.
  • reaction mixture was stirred at room temperature overnight before being diluted with ethyl acetate (20 mL) and washed with diluted brine solution (25 mL), bicarbonate solution (25 mL) and saturated brine solution (20 mL).
  • the organic phase was then dried (Na 2 SO 4 ) and concentrated in vacuo.
  • the residue was triturated with methanol ( ⁇ 2 mL), the suspension was sonicated and then allowed to stand overnight.
  • Ethyl l-(4-aminophenyl)piperidine-4-carboxylate, iV-(4-(2-chloropyrimidin-4- yl)phenyl)methanesulfonamide and p-tohien ⁇ sulfonic acid monohydrate (61 mg, 0.32 mmol, 0.84 eq) were placed in a reaction vessel, suspended in dioxane (4 mL) and heated at 100°C under a nitrogen atmosphere for 66 hours. The reaction mixture was transferred to a polypropylene tube and centrifuged. The liquid was decanted and the residue washed with methanol (2 x 3 mL).
  • Example 8 Compound 35 - 5-(4- ⁇ 4-[l-(cyanomethyl-amino)-2,2,2-trifluoro-ethyl]- phenyl ⁇ -pyrimidin-2-ylamino)-2-morpholin-4-yl-thiobenzoic acid S-fluoromethyl ester
  • Preparation of substituted trifluoroethylamines can be accomplished as indicated by several different paths.
  • path 1 the reductive animation of 4-bromo- 1,1,1, -trifluoroacetophenone with aminoacetonitrile, followed by optical resolution methods well known in the art, can afford either enantiomer of A.
  • Subsequent conversion of the bromide to boronate B is achieved by palladium-catalyzed cross-coupling with bis-pinacolatodiboron, and Suzuki- type coupling with 2,4-dichloropyrimidine, followed by elaboration via nucleophilic heteroaromatic displacement of the 2-chloride with the relevant aniline.
  • path 2 cross- imination of 4-methoxy- 1,1,1, -trifluoroacetophenone with benzophenone imine followed by reduction and cleavage under hydrogenolysis conditions would be expected to yield the free trifluoroethylbenzylamine, which upon conversion to the triflate and Pd-catalyzed cross-coupling as indicated in path 1, followed by alkylation using bromoacetonitrile, would yield B.
  • a modification to this route would be available through reduction of a benzyl imine (formed by the reaction of benzylamine/TiCl 4 ) and reduction of the intermediate with NaCNBH 3 to give the same intermediates.
  • Path 3 offers an alternative method to introduce the trifiuoromethyl group on an aldimine formed by the reaction of 4- bromobenzaldehyde and aminoacetonitrile, using the Motherwell-Storey reagent C as described.
  • Path 2 Path 3 (for preparation of C see. Motherwell, W.B., Storey, L.J., J. Fluorine Chem. (2005) 126(4), 489-496.;
  • Example 9 Compounds 37 and 38 - 5-[4-(2-ethyl-l,l,3-trioxo-2,3-dihydro-lH-l ⁇ 6 - benzo [d] isothiazol-6-yl)-py rimidin-2-ylamino] -2-morpholin-4-yl-thiobenzoic acid S- fluoromethyl ester and 5-[4-(2-Cyanomethyl-l,l,3-trioxo-2,3-dihydro-lH-l ⁇ 6 - benzo[ ⁇ /]isothiazol-6-yl)-pyrimidm-2-ylammo]-2-morphoIin-4-yl-thiobenzoic acid S- fluoromethyl ester
  • JAK kinase domains were produced using the following procedures:
  • JAKl The kinase domain of human JAKl was amplified from U937mRNA using the polymerase chain reaction with the following primers: XHOI-Jl 5'-CCG CTC GAG ACT GAA GTG GAC CCC ACA CAT-3' [SEQ. ID. NO.
  • the JAKl PCR products were cloned into the pDest20 destination vector (Gibco).
  • the JAKl plasmid was then transformed into competent DHlOBac cells (Gibco), and the recombinant baculovirus was prepared via Sf9 insect cell transfection.
  • the JAK2 PCR products were cloned into the pDest20 destination vector (Gibco).
  • the pDest20 destination vector Gibco.
  • JAK2 plasmid was then transformed into competent DHlOBac cells (Gibco), and the recombinant baculovirus was prepared via Sf9 insect cell transfection.
  • the kinase domain of human JAK3 was amplified from U937mRNA using the polymerase chain reaction with the following primers: XHOI- J3 5'-CCGCTC GAGTATGCC TGC CAAGACCCCACG-S' [SEQ. ID. NO.9]
  • the JAK3 PCR products were cloned into the pDest20 destination expression vector (Gibco).
  • the JAK3 plasmid was then transformed into competent DHlOBac cells (Gibco), and the recombinant baculovirus was prepared via Sf9 insect cell transfection.
  • Baculovirus preparations from each of the JAK family members were infected into one litre of Sf9 ⁇ Spodoptera frugiperda) cells (Invitrogen) grown in SF900II serum free medium (Invitrogen) to a cell density of approximately 2 x 10 6 cells/ml. Cells were infected with virus at a cell culture to virus stock ratio of 20:1. Cells were harvested and lysed 48 hours post infection. The GST-tagged JAK kinase domains were purified by affinity chromatography on a GSH agarose column (Scientifix).
  • Kinase assays were performed in 384 well Optiplates (Packard) using an Alphascreen Protein Tyrosine KinasePlOO detection kit
  • the compounds were pre-incubated with affinity purified PTK domain in the presence of phosphotyrosine assay buffer (1OmM HEPES, pH 7.5, 10OmM MgCl 2 , 25mM NaCl, 20OmM sodium vanadate and 0.1% Tween 20) for 20 minutes.
  • the compounds were then incubated with substrate in the presence of either 80 or 625um ATP for 60 or 90 minutes.
  • the substrate used was either susbtrate-1 with the sequence biotin-EGPWLEEEEEA YGWMDF-NH 2 [SEQ. ID. NO.
  • the coding region encompassing nucleotides 1-487 of TEL was amplified by PCR using the oligonucleotides 5TEL (5' -GGA GGA TCC TGA TCT CTC TCG CTG TGA GAC- 3') [SEQ ID NO 14] and 3TEL (5' -AGGC GTC GAC TTC TTC TTC ATG GTT CTG-3') [SEQ ID NO 15] and U937 mRNA as a template.
  • a BamHI restriction site was incorporated into the 5TEL primer, and a Sal I restriction site was incorporated into the 3TEL primer.
  • JAK2 The regions encompassing the kinase domain of JAK2 (nucleotides 2994- 3914; JAK2F 5'-ACGC GTC GAC GGT GCC TTT GAA GAC CGG GAT-3' [SEQ ID NO 16]; JAK2R 5'-ATA GTT TAG CGG CCG CTC AGA ATG AAG GTC ATT T-3') [SEQ ID NO 17] and JAK3 (nucleotides 2520-3469; JAK3F 5'-GAA GTC GAC TAT GCC TGC CAA GAC CCC ACG ATC TT-3') [SEQ ID NO 18] were generated by PCR using Taq DNA polymerase (Gibco/BRL) and U937 mRNA as a template.
  • Taq DNA polymerase Gabco/BRL
  • a Sal I restriction site was incorporated into the forward primer of JAK2 and JAK3, a Not I site was incorporated into the JAK2 reverse primer and a Xba I site was added to the reverse primer of JAK3.
  • a TEL/Jak2 fusion was generated by digestion of the TELPCR product with BamH I/Sal I restriction enzymes, digestion of the JAK2 PCR product with Sal I/Not I restriction enzymes, followed by ligation and subcloning of the ligation product into the mammalian expression Vector pTRE 2 (Clontech), which was prepared by digestion with BamH I- Not I restriction enzymes, to give the the TEL/Jak2 fusion plasmid pTELJAK2.
  • the TEL/Jak3 fusion was prepared by ligation of the JAK3 Sal I/Not I cleaved kinase domain PCR product with the BamH I/Sal I restriction digested TEL product, followed by ligation of the ligation product into the BamH I/Not I digested pTRE2, to give the TEL/Jak3 fusion plasmid pTELJAK3.
  • the growth factor dependant myelomonocytic cell line BaF3 bearing the pTET-off plasmid (Clontech) was transfected with either pTELJAK2 or pTELJAK3, and the transfected cells were selected for growth-factor independent cell growth.
  • the BaF3 wild-type cells were cultured in DMEM containing 10% FCS, 10% WEHI 3B conditioned medium.
  • the BaF3 TELJAK cells (BafT_J2 or BafT_J2) were cultured in DMEM 10% Tet-System Approved FBS (without WEHI 3B conditioned medium).
  • Cell suspensions were prepared by harvesting cells from culture, (the cells used in this test were in late log phase growth with high viability.) Cells were diluted in the appropriate growth medium, as described above, to l.lx final concentration (from 50,000 cell/mL to 200,000 cell/mL, depending on cell line).
  • Rat lung microvascular endothelial cells were grown in serum free media for 24 hours then exposed to IL-6 10 ng/ml with varying concentrations of compound 29. The cells were harvested after 30 minutes of exposure to IL-6 and the amount of STAT3 phosphorylation at tyrosine 705 was determined relative to total STAT3 with Western blots.
  • An example of a Western blot from this experiment is shown in Figure 1.
  • the primary antibody against pSTAT3 was specific for phosphyorylation at Y705.
  • the antibodies against pSTAT3 and STAT3 were from Cell Signaling (MA). Densitometric analysis was performed with Image J. Non-linear regression was performed with Graph Pad Prizm to obtain sigmoidal curves and calculate the EC50. Results shown in Figure 2 are from duplicate experiments.
  • Compound 29 was incubated at 37°C with human liver microsomes at a concentration of 1 ⁇ M. The reaction was initiated by the addition of an NADPH-regenerating system and quenched at various time points over the incubation period by the addition of acetonitrile. The relative loss of parent compound and formation of metabolic products was determined by LC/MS using a Micromass Q-TQF mass spectrometer.
  • Compound 29 exhibited rapid degradation in the incubation matrix containing human liver microsomes devoid of the cofactors required for CYP450-mediated metabolism. At the point of initiation of the assay, concentrations of test compound could not be detected, although a putative P-28 metabolite was detected. This indicates that compound 29 underwent non-NADPH dependent degradation in the microsomal matrix during the 10 minute incubation period which was likely to be due to hydrolysis of the ester bond by microsomal hydrolytic enzymes.
  • the compounds of formula I can be tested in the dog model of pulmonary hypertension as described in Gust, R and Schuster, D. P. Experimental Lung Research, 27:1-12, 2001. They can also be tested in a rabbit model of monocrotaline induced pulmonary hypertension.
  • the compounds of formula I can also be tested in humans with pulmonary arterial hypertension.
  • the effect of the compounds of formula I can be tested in humans with pulmonary arterial hypertension by measurement of its acute effects on cardiopulmonary hemodynamics.
  • the effect of the compounds on right ventricular pressures, pulmonary artery pressures, pulmonary vascular resistance, and cardiac output may be determined.
  • the effect of the compounds on the six minute walk time, and maximal oxygen consumption may be determined in humans with PAH.
  • the effect of the compounds on quality of life (as measured by a questionnaire), hospitalization, and survival may be determined in humans with PAH.
  • PAH may be caused by genetic abnormalities (i.e., primary or familial PAH) or secondary causes such as scleroderma, uncorrected congenital heart disease, mixed collagen vascular disorder, hepatitis C, or other liver disease, HIV infection, or hereditary hemorrhagic teleangiectasia.
  • the effect of the compounds may also be tested on human endothelial cells, fibroblasts and/or smooth muscle cell lines: for example, determination of IC50 for STAT3 phosphorylation in human pulmonary artery smooth muscle cell lines. Cell lines from other species, ie, the rat may also be examined.
  • rat pulmonary artery rings preconstricted with phenylephrine, or endothelin, or serotonin, or vasopressin, angiotensin II, or KCL may be studied to determine the dose response to the compounds for vasorelaxation.
  • Other vasoconstrictors may be examined.
  • hypoxia induced pulmonary hypertension might include study of rats, such as the Fawn-Hooded rat exposed to low oxygen (i.e., 5 percent oxygen).
  • Another model of hypoxia induced pulmonary hypertension might include the fetal calf maintained in a high altitude chamber.
  • the effect of the compounds may be examined in transgenic models of pulmonary hypertension: i.e., the BMPR2 knockout mouse treated with IL6, the caveolinl knock out mouse, or the vasoactive intestinal peptide knockout mouse.
  • the effect of the compounds on histopathologic changes that occur in both human and animal models of PAH may be measured.
  • the compounds may decrease the extent of plexiform lesions in the pulmonary arterioles of diseased lungs.
  • the plexiform lesion consists of endothelial cells, smooth muscle cells, and fibroblasts which proliferate and obstruct to a varying degree, the pulmonary arteriolar lumen.
  • the effect of the compounds can also be evaluated in asthma models. Asthma is restricted to human species, but animal models are often used to investigate particular aspects of this human disease. Bronchial biopsies and bronchoalveolar lavage (BAL) fluid recovered from patients with asthma have been shown to contain an increased number of activated T cells, B cells, eosinophils and mast cells. Many patients with asthma are sensitized and have specific immunoglogulin E (IgE) antibodies to one or more inhalant allergens. Atopy is, considered to be a major cause of asthma. In atopic individuals, inhalation of allergens preferentially induces a T-helper 2 cell (Th2) response.
  • IgE immunoglogulin E
  • mice are sensitized by itraperitoneal (ip) injection of ovalbumin (OVA), often together with a Th2 skewed adjuvant, such as alum.
  • OVA ovalbumin
  • C57/BL6 mice are actively sensitized on day 0 by ip injection of lO ⁇ g of OVA absorbed onto 1 mg of alum. From day 14-21 the mice are exposed daily to aerosolized OVA over a 30 minute period. On day 22, airway inflammation is apparent.
  • BAL fluid recovered from these animals demonstrate an increase in peri-bronchiolar space consisting of mixed cellular infiltrates of mononuclear cells and eosinophils.
  • OVA-specif ⁇ c IgE antibodies can be demonstrated in the serum of sensitized animals.
  • the mononuclear cell population consists mainly of cells of Th2 phenotype secreting cytokines IL-4 and IL- 5.
  • IL-4 promotes isotype switching of B cells towards IgE synthesis and IL-5 influences the production, maturation and activation of eosinophils.
  • AU publications mentioned in this specification are herein incorporated by reference. Any discussion of documents, acts, materials, devices, articles or the like which has been included in the present specification is solely for the purpose of providing a context for the present invention. It is not to be taken as an admission that any or all of these matters form part of the prior art base or were common general knowledge in the field relevant to the present invention as it existed in Australia or elsewhere before the priority date of each claim of this application.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Transplantation (AREA)
  • Virology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des composés de phénylaminopyrimidine qui sont des médicaments rétrométaboliques ou des métabolites de ces derniers. Ces composés sont des inhibiteurs des protéines kinases, dont les JAK kinases, en particulier les JAK2 kinases, et peuvent s'utiliser pour le traitement de maladies associées aux kinases telles que les maladies vasculaires, dont l'hypertension artérielle et pulmonaire, l'hypertrophie et l'ischémie; les affections immunologiques et respiratoire dont l'asthme et la maladie pulmonaire obstructive chronique, et les maladies prolifératives telles que le cancer, notamment le cancer du poumon.
PCT/AU2008/001326 2007-09-06 2008-09-05 Composés rétrométaboliques WO2009029998A1 (fr)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US97051907P 2007-09-06 2007-09-06
US60/970,519 2007-09-06
AU2007906387 2007-11-22
AU2007906387A AU2007906387A0 (en) 2007-11-22 Retrometabolic compounds

Publications (1)

Publication Number Publication Date
WO2009029998A1 true WO2009029998A1 (fr) 2009-03-12

Family

ID=40428376

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2008/001326 WO2009029998A1 (fr) 2007-09-06 2008-09-05 Composés rétrométaboliques

Country Status (1)

Country Link
WO (1) WO2009029998A1 (fr)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011101806A1 (fr) 2010-02-17 2011-08-25 Debiopharm S.A. Composés bicycliques et utilisations associées en tant qu'inhibiteurs mixtes de c-src/jak
EP2648728A2 (fr) * 2010-12-06 2013-10-16 Cephalon, Inc. Inhibiteur de la janus kinase 2 (jak2) pour le traitement du lupus
WO2014000032A1 (fr) * 2012-06-29 2014-01-03 Ym Biosciences Australia Pty Ltd Composés phénylamino-pyrimidine bicycliques et leurs utilisations
WO2014013014A1 (fr) 2012-07-18 2014-01-23 Fundació Privada Centre De Regulació Genòmica (Crg) Inhibiteurs de jak pour l'activation de populations de cellules souches épidermiques
WO2014060493A2 (fr) * 2012-10-18 2014-04-24 Bayer Pharma Aktiengesellschaft Dérivés n-(pyridin-2-yl)pyrimidin-4-amine contenant un groupe sulfone
WO2014060375A2 (fr) * 2012-10-18 2014-04-24 Bayer Pharma Aktiengesellschaft Dérivés 5-fluoro-n-(pyridin-2-yl)pyridin-2-amine contenant un groupe sulfone
WO2014114274A1 (fr) * 2013-01-28 2014-07-31 苏州泽璟生物制药有限公司 Composé d'aminopyrimidine phényle deutérié et composition pharmaceutique le contenant
WO2015019365A1 (fr) 2013-08-07 2015-02-12 Cadila Healthcare Limited N-cyanométhylamides comme inhibiteurs de la janus kinase
US9233934B2 (en) 2007-03-12 2016-01-12 Ym Biosciences Australia Pty Ltd Phenyl amino pyrimidine compounds and uses thereof
US9650361B2 (en) 2012-11-15 2017-05-16 Bayer Pharam Aktiengesellschaft N-(pyridin-2-yl)pyrimidin-4-amine derivatives containing a sulfoximine group
US9650340B2 (en) 2012-11-15 2017-05-16 Bayer Pharma Aktiengesellschaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfoximine group
US9669034B2 (en) 2011-05-24 2017-06-06 Bayer Intellectual Property Gmbh 4-aryl-N-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group
US9770445B2 (en) 2013-07-04 2017-09-26 Bayer Pharma Aktiengesellschaft Sulfoximine substituted 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives and their use as CDK9 kinase inhibitors
US9790189B2 (en) 2014-04-01 2017-10-17 Bayer Pharma Aktiengesellschaft Disubstituted 5-fluoro pyrimidine derivatives containing a sulfondiimine group
US9856242B2 (en) 2014-03-13 2018-01-02 Bayer Pharma Aktiengesellscaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfone group
US9884849B2 (en) 2014-10-16 2018-02-06 Bayer Pharma Aktiengesellschaft Fluorinated benzofuranyl-pyrimidine derivatives containing a sulfoximine group
US9902716B2 (en) 2014-10-16 2018-02-27 Bayer Pharma Aktiengesellschaft Fluorinated benzofuranyl-pyrimidine derivatives containing a sulfone group
WO2018041989A1 (fr) 2016-09-02 2018-03-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de diagnostic et de traitement de la maladie coeliaque réfractaire de type 2
US9963464B2 (en) 2014-04-11 2018-05-08 Bayer Pharma Aktiengesellschaft Macrocyclic compounds
WO2018185266A1 (fr) 2017-04-06 2018-10-11 Inventiva Nouveaux composés inhibiteurs de l'interaction yap/taz-tead et leur utilisation dans le traitement du mésothéliome malin
JP2019522011A (ja) * 2016-07-26 2019-08-08 深▲セン▼市塔吉瑞生物医▲薬▼有限公司Shenzhen TargetRx,Inc. プロテインチロシンキナーゼの活性を阻害するためのアミノピリミジン系化合物
RU2712226C2 (ru) * 2015-06-26 2020-01-27 Сучжоу Зелген Биофармасьютикалс Ко., Лтд Фениламинопиримидин или полиморфная форма соли фениламинопиримидина
EP3632908A1 (fr) 2018-10-02 2020-04-08 Inventiva Inhibiteurs de l'interaction yap / taz-tead et leur utilisation dans le traitement du cancer
WO2020201362A2 (fr) 2019-04-02 2020-10-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes de prédiction et de prévention du cancer chez des patients ayant des lésions prémalignes
WO2020212395A1 (fr) 2019-04-16 2020-10-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Utilisation d'inhibiteurs de jak pour le traitement d'états douloureux impliquant des canaux nav1.7
USRE48285E1 (en) 2014-06-12 2020-10-27 Sierra Oncology, Inc. N-(cyanomethyl)-4-(2-(4-morpholinophenylamino)pyrimidin-4-yl)benzamide
CN112239384A (zh) * 2020-08-07 2021-01-19 浙江理工大学 一种硫酯化合物的制备方法
US11242356B2 (en) 2017-03-28 2022-02-08 Bayer Aktiengesellschaft PTEFb inhibiting macrocyclic compounds
US11254690B2 (en) 2017-03-28 2022-02-22 Bayer Pharma Aktiengesellschaft PTEFb inhibiting macrocyclic compounds
US11701347B2 (en) 2018-02-13 2023-07-18 Bayer Aktiengesellschaft Use of 5-fluoro-4-(4-fluoro-2-methoxyphenyl)-N-{4-[(S-methylsulfonimidoyl)methyl]pyridin-2-yl}pyridin-2-amine for treating diffuse large B-cell lymphoma
WO2023222565A1 (fr) 2022-05-16 2023-11-23 Institut National de la Santé et de la Recherche Médicale Procédés d'évaluation de l'épuisement de cellules souches hématopoïétiques induites par une inflammation chronique

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002079197A1 (fr) * 2001-03-29 2002-10-10 Vertex Pharmaceuticals Incorporated Inhibiteurs de kinases n-terminales c-jun (jnk) et autres proteineskinases
WO2004041789A1 (fr) * 2002-11-01 2004-05-21 Vertex Pharmaceuticals Incorporated Composes utiles comme inhibiteurs de proteines jak et autres proteines kinase

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002079197A1 (fr) * 2001-03-29 2002-10-10 Vertex Pharmaceuticals Incorporated Inhibiteurs de kinases n-terminales c-jun (jnk) et autres proteineskinases
WO2004041789A1 (fr) * 2002-11-01 2004-05-21 Vertex Pharmaceuticals Incorporated Composes utiles comme inhibiteurs de proteines jak et autres proteines kinase

Cited By (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9238628B2 (en) 2007-03-12 2016-01-19 YM Biosicences Australia PTY LTD Phenyl amino pyrimidine compounds and uses thereof
US9233934B2 (en) 2007-03-12 2016-01-12 Ym Biosciences Australia Pty Ltd Phenyl amino pyrimidine compounds and uses thereof
WO2011101806A1 (fr) 2010-02-17 2011-08-25 Debiopharm S.A. Composés bicycliques et utilisations associées en tant qu'inhibiteurs mixtes de c-src/jak
EP2648728A2 (fr) * 2010-12-06 2013-10-16 Cephalon, Inc. Inhibiteur de la janus kinase 2 (jak2) pour le traitement du lupus
EP2648728A4 (fr) * 2010-12-06 2014-06-04 Cephalon Inc Inhibiteur de la janus kinase 2 (jak2) pour le traitement du lupus
US9962389B2 (en) 2011-05-24 2018-05-08 Bayer Intellectual Property Gmbh 4-aryl-N-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group
US9669034B2 (en) 2011-05-24 2017-06-06 Bayer Intellectual Property Gmbh 4-aryl-N-phenyl-1,3,5-triazin-2-amines containing a sulfoximine group
AU2013201780B2 (en) * 2012-06-29 2015-04-02 Ym Biosciences Australia Pty Ltd Phenyl Amino Pyrimidine Bicyclic Compounds And Uses Thereof
WO2014000032A1 (fr) * 2012-06-29 2014-01-03 Ym Biosciences Australia Pty Ltd Composés phénylamino-pyrimidine bicycliques et leurs utilisations
CN104583216A (zh) * 2012-06-29 2015-04-29 Ym生物科学澳大利亚股份有限公司 苯基氨基嘧啶双环化合物及其用途
US8809359B2 (en) 2012-06-29 2014-08-19 Ym Biosciences Australia Pty Ltd Phenyl amino pyrimidine bicyclic compounds and uses thereof
WO2014013014A1 (fr) 2012-07-18 2014-01-23 Fundació Privada Centre De Regulació Genòmica (Crg) Inhibiteurs de jak pour l'activation de populations de cellules souches épidermiques
US9670161B2 (en) 2012-10-18 2017-06-06 Bayer Pharma Aktiengesellschaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfone group
WO2014060493A2 (fr) * 2012-10-18 2014-04-24 Bayer Pharma Aktiengesellschaft Dérivés n-(pyridin-2-yl)pyrimidin-4-amine contenant un groupe sulfone
WO2014060375A3 (fr) * 2012-10-18 2014-08-07 Bayer Pharma Aktiengesellschaft Dérivés 5-fluoro-n-(pyridin-2-yl)pyridin-2-amine contenant un groupe sulfone
CN104854091A (zh) * 2012-10-18 2015-08-19 拜耳药业股份公司 含砜基团的5-氟-n-(吡啶-2-基)吡啶-2-胺衍生物
WO2014060375A2 (fr) * 2012-10-18 2014-04-24 Bayer Pharma Aktiengesellschaft Dérivés 5-fluoro-n-(pyridin-2-yl)pyridin-2-amine contenant un groupe sulfone
JP2015534960A (ja) * 2012-10-18 2015-12-07 バイエル ファーマ アクチエンゲゼルシャフト スルホン基を含んでいる5−フルオロ−n−(ピリジン−2−イル)ピリジン−2−アミン誘導体
US9708293B2 (en) 2012-10-18 2017-07-18 Bayer Pharma Aktiengesellschaft N-(pyridin-2-yl)pyrimidin-4-amine derivatives containing a sulfone group
WO2014060493A3 (fr) * 2012-10-18 2014-08-07 Bayer Pharma Aktiengesellschaft Dérivés n-(pyridin-2-yl)pyrimidin-4-amine contenant un groupe sulfone
US9877954B2 (en) 2012-11-15 2018-01-30 Bayer Pharma Aktiengesellschaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfoximine group
US9650361B2 (en) 2012-11-15 2017-05-16 Bayer Pharam Aktiengesellschaft N-(pyridin-2-yl)pyrimidin-4-amine derivatives containing a sulfoximine group
US9650340B2 (en) 2012-11-15 2017-05-16 Bayer Pharma Aktiengesellschaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfoximine group
CN103965114A (zh) * 2013-01-28 2014-08-06 苏州泽璟生物制药有限公司 氘代的苯基氨基嘧啶化合物以及包含该化合物的药物组合物
WO2014114274A1 (fr) * 2013-01-28 2014-07-31 苏州泽璟生物制药有限公司 Composé d'aminopyrimidine phényle deutérié et composition pharmaceutique le contenant
RU2633694C2 (ru) * 2013-01-28 2017-10-17 Сучжоу Зельген Биофармасьютикалс Ко., Лтд. Дейтерированный фениламинопиримидин и фармацевтическая композиция, содержащая такое соединение
CN103965114B (zh) * 2013-01-28 2016-01-06 苏州泽璟生物制药有限公司 氘代的苯基氨基嘧啶化合物以及包含该化合物的药物组合物
CN104955811A (zh) * 2013-01-28 2015-09-30 苏州泽璟生物制药有限公司 氘代的苯基氨基嘧啶化合物以及包含该化合物的药物组合物
US9604935B2 (en) 2013-01-28 2017-03-28 Suzhou Zelgen Biopharmaceuticals Co., Ltd. Deuterated phenyl amino pyrimidine compound and pharmaceutical composition containing the same
US9770445B2 (en) 2013-07-04 2017-09-26 Bayer Pharma Aktiengesellschaft Sulfoximine substituted 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives and their use as CDK9 kinase inhibitors
TWI562990B (en) * 2013-08-07 2016-12-21 Cadila Healthcare Ltd Novel compounds as inhibitors of janus kinase
CN105612154A (zh) * 2013-08-07 2016-05-25 卡迪拉保健有限公司 作为詹纳斯激酶的抑制剂的n-氰基甲基酰胺
WO2015019365A1 (fr) 2013-08-07 2015-02-12 Cadila Healthcare Limited N-cyanométhylamides comme inhibiteurs de la janus kinase
KR101665301B1 (ko) 2013-08-07 2016-10-11 카딜라 핼쓰캐어 리미티드 야누스 키나제의 억제제로서 n-시아노메틸아미드
JP2016525071A (ja) * 2013-08-07 2016-08-22 カディラ ヘルスケア リミテッド ヤーヌスキナーゼのインヒビターとしてのn−シアノメチルアミド
KR20160030580A (ko) * 2013-08-07 2016-03-18 카딜라 핼쓰캐어 리미티드 야누스 키나제의 억제제로서 n-시아노메틸아미드
US9556148B2 (en) 2013-08-07 2017-01-31 Cadila Healthcare Limited N-cyanomethylamides as inhibitors of janus kinase
US9856242B2 (en) 2014-03-13 2018-01-02 Bayer Pharma Aktiengesellscaft 5-fluoro-N-(pyridin-2-yl)pyridin-2-amine derivatives containing a sulfone group
US9790189B2 (en) 2014-04-01 2017-10-17 Bayer Pharma Aktiengesellschaft Disubstituted 5-fluoro pyrimidine derivatives containing a sulfondiimine group
US9963464B2 (en) 2014-04-11 2018-05-08 Bayer Pharma Aktiengesellschaft Macrocyclic compounds
USRE49445E1 (en) 2014-06-12 2023-03-07 Sierra Oncology, Inc. N-(cyanomethyl)-4-(2-(4-morpholinophenylamino)pyrimidin-4-yl)benzamide
USRE48285E1 (en) 2014-06-12 2020-10-27 Sierra Oncology, Inc. N-(cyanomethyl)-4-(2-(4-morpholinophenylamino)pyrimidin-4-yl)benzamide
US9884849B2 (en) 2014-10-16 2018-02-06 Bayer Pharma Aktiengesellschaft Fluorinated benzofuranyl-pyrimidine derivatives containing a sulfoximine group
US9902716B2 (en) 2014-10-16 2018-02-27 Bayer Pharma Aktiengesellschaft Fluorinated benzofuranyl-pyrimidine derivatives containing a sulfone group
RU2712226C2 (ru) * 2015-06-26 2020-01-27 Сучжоу Зелген Биофармасьютикалс Ко., Лтд Фениламинопиримидин или полиморфная форма соли фениламинопиримидина
JP2020183432A (ja) * 2016-07-26 2020-11-12 深▲セン▼市塔吉瑞生物医▲薬▼有限公司Shenzhen TargetRx,Inc. プロテインチロシンキナーゼの活性を阻害するためのアミノピリミジン系化合物
JP2019522011A (ja) * 2016-07-26 2019-08-08 深▲セン▼市塔吉瑞生物医▲薬▼有限公司Shenzhen TargetRx,Inc. プロテインチロシンキナーゼの活性を阻害するためのアミノピリミジン系化合物
JP7156715B2 (ja) 2016-07-26 2022-10-19 深▲セン▼市塔吉瑞生物医▲薬▼有限公司 プロテインチロシンキナーゼの活性を阻害するためのアミノピリミジン系化合物
US11111233B2 (en) 2016-07-26 2021-09-07 Shenzhen Targetrx, Inc. Amino pyrimidine compound for inhibiting protein tyrosine kinase activity
WO2018041989A1 (fr) 2016-09-02 2018-03-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés de diagnostic et de traitement de la maladie coeliaque réfractaire de type 2
US11242356B2 (en) 2017-03-28 2022-02-08 Bayer Aktiengesellschaft PTEFb inhibiting macrocyclic compounds
US11254690B2 (en) 2017-03-28 2022-02-22 Bayer Pharma Aktiengesellschaft PTEFb inhibiting macrocyclic compounds
US11691986B2 (en) 2017-03-28 2023-07-04 Bayer Aktiengesellschaft PTEFB inhibiting macrocyclic compounds
WO2018185266A1 (fr) 2017-04-06 2018-10-11 Inventiva Nouveaux composés inhibiteurs de l'interaction yap/taz-tead et leur utilisation dans le traitement du mésothéliome malin
US11701347B2 (en) 2018-02-13 2023-07-18 Bayer Aktiengesellschaft Use of 5-fluoro-4-(4-fluoro-2-methoxyphenyl)-N-{4-[(S-methylsulfonimidoyl)methyl]pyridin-2-yl}pyridin-2-amine for treating diffuse large B-cell lymphoma
WO2020070181A1 (fr) 2018-10-02 2020-04-09 Inventiva Inhibiteurs de l'interaction yap/taz-tead et leur utilisation dans le traitement du cancer
EP3632908A1 (fr) 2018-10-02 2020-04-08 Inventiva Inhibiteurs de l'interaction yap / taz-tead et leur utilisation dans le traitement du cancer
WO2020201362A2 (fr) 2019-04-02 2020-10-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Méthodes de prédiction et de prévention du cancer chez des patients ayant des lésions prémalignes
WO2020212395A1 (fr) 2019-04-16 2020-10-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Utilisation d'inhibiteurs de jak pour le traitement d'états douloureux impliquant des canaux nav1.7
CN112239384A (zh) * 2020-08-07 2021-01-19 浙江理工大学 一种硫酯化合物的制备方法
CN112239384B (zh) * 2020-08-07 2023-05-12 浙江理工大学 一种硫酯化合物的制备方法
WO2023222565A1 (fr) 2022-05-16 2023-11-23 Institut National de la Santé et de la Recherche Médicale Procédés d'évaluation de l'épuisement de cellules souches hématopoïétiques induites par une inflammation chronique

Similar Documents

Publication Publication Date Title
WO2009029998A1 (fr) Composés rétrométaboliques
US20220388967A1 (en) Phenyl Amino Pyrimidine Compounds and Uses Thereof
EP2114900B1 (fr) Composés à base de thiopyrimidine et ses utilisations
EP2215094B1 (fr) Composés hétérocycliques à teneur en n
AU2016200866B2 (en) Phenyl amino pyrimidine compounds and uses thereof
AU2013201306B2 (en) Phenyl Amino Pyrimidine Compounds and Uses Thereof

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: 08783061

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: 08783061

Country of ref document: EP

Kind code of ref document: A1