AU2011328237A1 - Pyridine compounds and aza analogues thereof as TYK2 inhibitors - Google Patents

Pyridine compounds and aza analogues thereof as TYK2 inhibitors Download PDF

Info

Publication number
AU2011328237A1
AU2011328237A1 AU2011328237A AU2011328237A AU2011328237A1 AU 2011328237 A1 AU2011328237 A1 AU 2011328237A1 AU 2011328237 A AU2011328237 A AU 2011328237A AU 2011328237 A AU2011328237 A AU 2011328237A AU 2011328237 A1 AU2011328237 A1 AU 2011328237A1
Authority
AU
Australia
Prior art keywords
methyl
pyrazol
pyrimidin
amino
amine
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
AU2011328237A
Inventor
Katie Ellard
Alison Jones
Rosemary Lynch
Jeremy Major
Nigel Ramsden
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cellzome Ltd
Original Assignee
Cellzome 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
Application filed by Cellzome Ltd filed Critical Cellzome Ltd
Publication of AU2011328237A1 publication Critical patent/AU2011328237A1/en
Abandoned legal-status Critical Current

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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • A61P5/40Mineralocorticosteroids, e.g. aldosterone; Drugs increasing or potentiating the activity of mineralocorticosteroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • 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
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms 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
    • C07D417/12Heterocyclic 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 linked by a chain containing hetero atoms as chain links
    • 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
    • C07D417/14Heterocyclic 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 three or more hetero rings

Abstract

The present invention relates to compounds of formula (I), wherein R

Description

WO 2012/062704 1 PCT/EP2011/069553 PYRIDINE COMPOUNDS AND AZA ANALOGUES THEREOF AS TYK2 INHIBITORS 5 The present invention relates to a novel class of kinase inhibitors, including pharmaceutically acceptable salts, prodrugs and metabolites thereof, which are useful for modulating protein kinase activity for modulating cellular activities such as signal transduction, proliferation, and 10 cytokine secretion. More specifically the invention provides compounds which inhibit, regulate and/or modulate kinase activity, in particular TYK2 activity, and signal transduction pathways relating to cellular activities as mentioned above. Furthermore, the present invention relates to pharmaceutical compositions comprising said compounds, for example for the treatment or prevention of an immunological, inflammatory, autoimmune, or allergic 15 disorder or disease or a transplant rejection or a Graft-versus host disease. Kinases catalyze the phosphorylation of proteins, lipids, sugars, nucleosides and other cellular metabolites and play key roles in all aspects of eukaryotic cell physiology. Especially, protein kinases and lipid kinases participate in the signaling events which control the activation, 20 growth, differentiation and survival of cells in response to extracellular mediators or stimuli such as growth factors, cytokines or chemokines. In general, protein kinases are classified in two groups, those that preferentially phosphorylate tyrosine residues and those that preferentially phosphorylate serine and/or threonine residues. The tyrosine kinases include membrane-spanning growth factor receptors such as the epidermal growth factor receptor 25 (EGFR) and cytosolic non-receptor kinases such as Janus kinases (JAK). Inappropriately high protein kinase activity is involved in many diseases including cancer, metabolic diseases, autoimmune or inflammatory disorders. This effect can be caused either directly or indirectly by the failure of control mechanisms due to mutation, overexpression or 30 inappropriate activation of the enzyme. In all of these instances, selective inhibition of the kinase is expected to have a beneficial effect. One group of kinases that has become a recent focus of drug discovery is the Janus kinase (JAK) family of non-receptor tyrosine kinases. In mammals, the family has four members, WO 2012/062704 2 PCT/EP2011/069553 JAKI, JAK2, JAK3 and Tyrosine kinase 2 (TYK2). Each protein has a kinase domain and a catalytically inactive pseudo-kinase domain. The JAK proteins bind to cytokine receptors through their amino-terminal FERM (Band-4.1, ezrin, radixin, moesin) domains. After the binding of cytokines to their receptors, JAKs are activated and phosphorylate the receptors, 5 thereby creating docking sites for signalling molecules, especially for members of the signal transducer and activator of transcription (STAT) family (Yamaoka et al., 2004. The Janus kinases (JAKs). Genome Biology 5(12): 253). In mammals, JAKI, JAK2 and TYK2 are ubiquitously expressed. By contrast, the expression 10 of JAK3 is predominantly in hematopoietic cells and it is highly regulated with cell development and activation (Musso et al., 1995. J. Exp. Med. 181(4):1425-3 1). The study of JAK-deficient cell lines and gene-targeted mice has revealed the essential, nonredundant functions of JAKs in cytokine signalling. JAKI knockout mice display a 15 perinatal lethal phenotype, probably related to the neurological effects that prevent them from sucking (Rodig et al., 1998. Cell 93(3):373-83). Deletion of the JAK2 gene results in embryonic lethality at embryonic day 12.5 as a result of a defect in erythropoiesis (Neubauer et al., 1998. Cell 93(3):397-409). Interestingly, JAK3 deficiency was first identified in humans with autosomal recessive severe combined immunodeficiency (SCID) (Macchi et al., 20 1995. Nature 377(6544):65-68). JAK3 knockout mice too exhibit SCID but do not display non-immune defects, suggesting that an inhibitor of JAK3 as an immunosuppressant would have restricted effects in vivo and therefore presents a promising drug for immunosuppression (Papageorgiou and Wikman 2004, Trends in Pharmacological Sciences 25(11):558-62). 25 The role of TYK2 in the biological response to cytokines was first characterized using a mutant human cell line that was resistant to the effects of Type I interferons (IFNs) and the demonstration that IFNa responsiveness could be restored by genetic complementation of TYK2 (Velazquez et al, 1992. Cell 70, 313-322). Further in vitro studies implicated TYK2 in the signaling pathways of multiple other cytokines involved in both innate and adaptive 30 immunity. Analysis of TYK-2_ mice however revealed less profound immunological defects than were anticipated (Karaghiosoff et al., 2000. Immunity 13, 549-560; Shimoda et al., 2000. Immunity 13, 561-671). Surprisingly, TYK2 deficient mice display merely reduced responsiveness to IFNa/0 and signal normally to interleukin 6 (IL-6) and interleukin 10 (IL 10), both of which activate TYK2 in vitro. In contrast, TYK2 was shown to be essential for WO 2012/062704 3 PCT/EP2011/069553 IL-12 signaling with the absence of TYK2 resulting in defective STAT4 activation and the failure of T cells from these mice to differentiate into IFNy- producing Th1 cells. Consistent with the involvement of TYK2 in mediating the biological effects of Type I IFNs and IL-12, TYK2-/- mice were more susceptible to viral and bacterial infections. 5 Thus far only a single patient with an autosomal recessive TYK2 deficiency has been described (Minegishi et al., 2006. Immunity 25, 745-755). The homozygous deletion of four base pairs (GCTT at nucleotide 550 in the TYK2 gene) and consequent frameshift mutation in the patient's coding DNA introduced a premature stop codon and resulted in the truncation of 10 the TYK2 protein at amino acid 90. The phenotype of this null mutation in human cells was much more severe than predicted by the studies in murine cells lacking TYK2. The patient displayed clinical features reminiscent of the primary immunodeficiency hyper-IgE syndrome (HIES) including recurrent skin abscesses, atopic dermatitis, highly elevated serum IgE levels and susceptibility to multiple opportunistic infections. Contrary to reports in TYK2-/- mice, 15 signaling by a wide variety of cytokines was found to be impaired thus highlighting non redundant roles for human TYK2 in the function of Type I IFNs, IL-6, IL-10, IL-12 and IL 23. An imbalance in T helper cell differentiation was also observed, with the patient's T cells exhibiting an extreme skew towards the development of IL-4 producing Th2 cells and impaired Th1 differentiation. Indeed, these cytokine signaling defects could be reponsible for 20 many of the clinical manifestations described, for example atopic dermatitis and elevated IgE levels (enhanced Th2), increased incidence of viral infections (IFN defect), infection with intracellular bacteria (IL-12/Thl defect) and extracellular bacteria (IL-6 and IL-23/Thl7 defect). 25 Emerging evidence from genome-wide association studies suggests that single nucleotide polymorphisms (SNPs) in the TYK2 gene significantly influence autoimmune disease susceptibility. Less efficient TYK2 variants are associated with protection against systemic lupus erythematosus (SLE) (TYK2 rs2304256 and rs12720270, Sigurdsson et al., 2005. Am. J. Hum. Genet. 76, 528-537; Graham et al., 2007. Rheumatology 46, 927-930; Hellquist et al., 30 2009. J. Rheumatol. 36, 1631-1638; Jarvinen et al., 2010. Exp. Dermatol. 19, 123-131) and multiple sclerosis (MS) (rs34536443, Ban et al., 2009. Eur. J. Hum. Genet. 17, 1309-1313; Mero et al., 2009. Eur. J. Hum. Genet. 18, 502-504). Whereas predicted gain-of-function mutations increase susceptibility to inflammatory bowel disease (IBD) (rs280519 and rs2304256, Sato et al., 2009. J. Clin. Immunol. 29, 815-825). In support of the involvement of WO 2012/062704 4 PCT/EP2011/069553 TYK2 in immunopathologic disease processes, it has been shown that B1O.DI mice harbouring a missense mutation in the pseudokinase domain of TYK2 that results in the absence of encoded TYK2 protein are resistant to both autoimmune arthritis (CIA) and experimental autoimmune encephalomyelitis (EAE) (Shaw et al., 2003. PNAS 100, 11594 5 11599; Spach et al., 2009. J. Immunol. 182, 7776-7783). Furthermore, a recent study showed that TYK2 -/- mice were completely resistant to MOG-induced EAE (Oyamada et al., 2009. J. Immunol. 183, 7539-7546). In these mice resistance was accompanied by a lack of CD4 T cells infiltrating the spinal cord, a failure to signal through IL-12R and IL-23R and hence the inability to upregulate encephalitogenic levels of IFNy and IL-17. 10 The non-receptor tyrosine kinase TYK2 plays essential roles in both innate and adaptive immunity. A lack of TYK2 expression manifests in the attenuated signaling of multiple pro inflammatory cytokines and a profound imbalance in T helper cell differentiation. Furthermore, evidence from genetic association studies supports that TYK2 is a shared 15 autoimmune disease susceptibility gene. Taken together, these reasons suggest TYK2 as a target for the treatment of inflammatory and auto-immune diseases. Several JAK family inhibitors have been reported in the literature which may be useful in the medical field (Ghoreschi et al., 2009. Immunol Rev, 228:273-287). It is expected that a 20 selective TYK2 inhibitor that inhibits TYK2 with greater potency than JAK2 may have advantageous therapeutic properties, because inhibition of JAK2 can cause anemia (Ghoreschi et al., 2009. Nature Immunol. 4, 3 56-360). TYK2 inhibitors are described in European patent application with application N' 25 10168056.9. Phenylaminopyrimidines as JAK2 kinase selective compounds are known from WO-A 2008/109943. Pyrimidinyl-thiophene kinase modulators are known from WO-A 2007/053776. TYK2 inhibitors are known from DE-A 102009001438, DE-A 102009015070 and WO-A 30 2011/113802. Similar pyrimidine compounds are known from N. Hebert et al., Internationnal Journal of Mass Spectrometry and Ion Processes 79 (1986), 45-56 and K. Takagi et al. Chimica Therapeutica 1974-9, N' 1, p. 14 to 18.
WO 2012/062704 5 PCT/EP2011/069553 Even though TYK2 inhibitors are known in the art there is a need for providing additional TYK2 inhibitors having at least partially more effective pharmaceutically relevant properties, like activity, selectivity especially over JAK2 kinase, and ADMET properties. 5 Thus, an object of the present invention is to provide a new class of compounds as TYK2 inhibitors which preferably show selectivity over JAK2 and may be effective in the treatment or prophylaxis of disorders associated with TYK2. 10 Accordingly, the present invention provides compounds of formula (I) N H x2 X1
R
2 or a pharmaceutically acceptable salt, prodrug or metabolite thereof, wherein 15
R
1 is T'; C(O)R 4 ; C(O)N(R 4 a)R 4 ; or C(O)OR 4 ;
R
4 is T'; or C 1
.
6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more R , which are the same or different; 20
R
4 a is H; or C 1
.
6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more halogen, which are the same or different; R is T 2 ; halogen; CN; C(O)OR6; OR6; C(O)R 6 ; C(O)N(RR 6 a); S(O) 2
N(RR
6 a); 25 S(O)N(R 6 Ra); S(O) 2
R
6 ; S(O)R 6 ; N(R 6
)S(O)
2 N(R6aR6 ); N(R 6 )S(O)N(R6aR6 ); SR 6 ; N(R 6
R
6 a); OC(O)R6; N(R6)C(O)R 6 a; N(R 6
)S(O)
2
R
6 a; N(R 6
)S(O)R
6 a; N(R 6
)C(O)N(R
6 aR6);
N(R
6
)C(O)OR
6 a; or OC(O)N(RR 6 a); R6, R 6a, R6b are independently selected from the group consisting of H; T 2 ; and C1.6 alkyl, 30 wherein C 1
.
6 alkyl is optionally substituted with one or more halogen, which are the same or different; WO 2012/062704 6 PCT/EP2011/069553 TI is C 3 .7 cycloalkyl; or 5 membered aromatic heterocyclyl, wherein T' is optionally substituted with one or more R 7 , which are the same or different; 5 R 7 is T 2 ; halogen; CN; C(O)OR; OR 8 ; oxo (=0), where the ring is at least partially saturated; C(O)R; C(O)N(RR a); S(O) 2 N(RR a); S(O)N(RR a); S(O) 2 R; S(O)R;
N(RS)S(O)
2 N(R'aR"); N(R )S(O)N(R aR"); SR'; N(RR a); OC(O)R; N(RS)C(O)R a;
N(RS)S(O)
2 R a; N(RS)S(O)R a; N(R 5 )C(O)N(RaR"); N(R )C(O)ORa; OC(O)N(RR8a); or C1.6 alkyl, wherein C1.6 alkyl is optionally substituted with one or more R 9 , which are the 10 same or different; R , R 8a, R8b are independently selected from the group consisting of H; T 2 ; or C1.6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more R1, which are the same or different; 15 R9 is T 2 ; halogen; CN; C(O)ORSC; ORSC; C(O)RC; C(O)N(RcR Id); S(O) 2 N(RCR d); S(O)N(RcR Id); S(O) 2 RC; S(O)RC; N(RC)S(O) 2 N(R'dR'e); N(R)S(O)N(R'dR e); SR8C; N(RcR 8d); OC(O)RC; N(Rc)C(O)R d; N(RC)S(O) 2 R d; N(RC)S(O)R d N(RC)C(O)N(R'dR'e); N(RC)C(O)OR d; and OC(O)N(RCR d); 20 R c, R d, Re are independently selected from the group consisting of H; T 2 ; or C1.6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more halogen, which are the same or different; 25 T 2 is phenyl; C 3 .7 cycloalkyl; or 4 to 7 membered heterocyclyl, wherein T2 is optionally substituted with one or more R", which are the same or different; R" is halogen; CN; C(O)OR ; OR ; oxo (=0), where the ring is at least partially saturated;
C(O)R
12 ; C(O)N(R 2 R1 2 a); S(O) 2 N(R1 2 R1 2 a); S(O)N(R1 2 R1 2 a); S(O)2R 12
S(O)R
12 ; 30 N(R 12
)S(O)
2 N(R1 2 aR 2 b); N(R 12
)S(O)N(R
2 aR 2 b); SR; 12 N(R1 2 R1 2 a); OC(O)R 12 ;
N(R
2 )C(O)R1 2 a; N(Rl 2 )S(O)2R1 2 a; N(R 2 )S(O)R1 2 a; N(R 2 )C(O)N(R1 2 aR 2 b).
N(R
12 )C(O)OR1 2 a; OC(O)N(R1 2 R1 2 a); or C1.6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more R , which are the same or different; WO 2012/062704 7 PCT/EP2011/069553 R10 13 14 14 R , R are independently selected from the group consisting of halogen; CN; C(O) OR OR 14; C(O)R 14; C(O)N(R14 R1 4 a); S(O) 2 N(R R1 4 a); S(O)N(R 14R 14a); S(O) 2 R 14; S(O)R14; N(R 14)S(O) 2 N(R1 4 aR14b); N(R 14)S(O)N(R1 4 aR 4 b); SR 14; N(R1 4 R1 4 a); OC(O)R14; N(R 4)C(O)R1 4 a; N(R 4)S(O) 2 R 14a; N(R 4)S(O)R 14a; N(R 14)C(O)N(R1 4 aR4b); 5 N(R 14)C(O)OR1 4 a; and OC(O)N(R 14R 14a 12 12a 12b 14 14a 14b R , R , R2, R , R1, R are independently selected from the group consisting of H; and C1.6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more halogen, which are the same or different; 10
X
1 is N and X 2 is C(R 15 ); or X 1 is C(R 16 ) and X 2 is C(R 15 ); or X 1 is N and X 2 is N;
R
15 is C(R 17
)
3 ; 15 R3, R16 are independently from the group consisting of H; and halogen; each R "7 is independently selected from the group consisting of H; and halogen;
R
2 is phenyl; or 4 to 7 membered aromatic heterocyclyl, wherein R 2 is optionally substituted 20 with one or more R 18 , which are the same or different;
R
1 8 is T 3 ; halogen; CN; C(O)OR"; OR"; C(O)R"; C(O)N(R 9
R
9 a); S(O) 2
N(R
9 R1 9 a
S(O)N(R
9 R1 9 a); S(O) 2 R 9; S(O)R 9; N(R 9)S(O) 2 N(R1 9 aRl9b); N(R 9)S(O)N(R19aR19b); SR 9 N(R R 19a) OC(O)R 1; N(R 9)C(O)R1 9 a; N(R 9)S(0) 2 R9a; N(R 9)S(O)R19a. 25 N(R 9)C(O)N(R1 9 aR 19b); N(R 9)C(O)OR1 9 a; OC(O)N(Rl9R1 9 a); or C1.6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more R 20 , which are the same or different; R 1, R 19a, R19b are independently selected from the group consisting of H; T 3 ; or C1.6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more R2, which are the same or 30 different;
T
3 is phenyl; C 3 .7 cycloalkyl; or 4 to 7 membered heterocyclyl, wherein T3 is optionally substituted with one or more R , which are the same or different; WO 2012/062704 8 PCT/EP2011/069553
R
22 is halogen; CN; C(O)OR ; OR ; oxo (=0), where the ring is at least partially saturated;
C(O)R
23 ; C(O)N(R 23 2 3 a); S(O) 2
N(R
23
R
23 a); S(O)N(R 23
R
23 a); S(O)R 2 3 ; S(O)R 23 ;
N(R
23
)S(O)
2
N(R
23 aR 23 b); N(R 23
)S(O)N(R
2 3 aR 23 b); SR 23 ; N(R 23
R
23 a); OC(O)R 2 3 ;
N(R
23
)C(O)R
23 a; N(R 23 ) S(O) 2
R
23 a; N(R 23
)S(O)R
23 a; N(R 23
)C(O)N(R
23 aR 23 b); 5 N(R 23
)C(O)OR
23 a; OC(O)N(R 23
R
2 3 a); or C1.6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more R 24 , which are the same or different; R 20, R2, R 2 4 are independently selected from the group consisting of halogen; CN; C(O)OR2;
OR
25 ; C(O)R 25 ; C(O)N(R5R 2 5a); S(O) 2 N(R5R 2 5a); S(O)N(R 25
R
2 5a); S(O) 2
R
25 ; S(O)R 25 ; 10 N(R 25
)S(O)
2
N(R
2 5aR 2 5b); N(R 25 )S(O)N(R5aR 2 5b); SR25; N(R5R 2 5a); OC(O)R 25 ;
N(R
25
)C(O)R
2 5a; N(R 2 5 )S(O)2R 2 5a; N(R 2 5
)S(O)R
2 5a; N(R 25
)C(O)N(R
2 5aR 2 5b).
N(R
25
)C(O)OR
2 5a; and OC(O)N(R R25a
R
2 , R 23 a, R 23 b, R, R 2 a, R 2 b are independently selected from the group consisting of H; T 4 ; 26 15 and C 1
.
6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more R , which are the same or different;
T
4 is phenyl; C 3 .7 cycloalkyl; or 4 to 7 membered heterocyclyl, wherein T4 is optionally substituted with one or more R 7 , which are the same or different; 20
R
27 is halogen; CN; C(O)OR28; OR28; oxo (=0), where the ring is at least partially saturated; C(O)R28; C(O)N(R 2 8
R
2 8 a); S(O) 2
N(R
2 8
R
28 a); S(O)N(R 2 8
R
28 a); S(O) 2 R28; S(O)R28. N(R28)S(O) 2
N(R
2 8 aR28b); N(R28)S(O)N(R 2 aR 28 b); SR28; N(R 2 8
R
28 a); OC(O)R28. N(R2 )C()R 28 a; N(R28)S(O) 2 R2a; N(R2)S(O)R ; N(R2)C(O)N(R 2 8 aR2 ) 25 N(R2 )C(O)OR 2 8 a; OC(O)N(R 2 8
R
2 8 a); or C1.6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more R 29 , which are the same or different; R 26, R29 are independently selected from the group consisting of halogen; CN; C(O)OR ; OR3 ; C(O)R3 ; C(O)N(R3 R 3 0a); S(O) 2 N(R R 3 0a); S(O)N(R3 R30a); S(O) 2 R3 ; S(O)R30; 30 N(R3 )S(O) 2
N(R
3 0aR30b); N(R3 )S(O)N(R 3 aR 3 0b); SR3 ; N(R 3
OR
3 0a); OC(O)R30; N(R3 )C(O)R 3 0a; N(R3 )S(O) 2 R30a; N(R3 )S(O)R30a; N(R3 )C(O)N(R 3 OaR30b); N(R3 )C(O)OR 3 0a; and OC(O)N(R3 R30a WO 2012/062704 9 PCT/EP2011/069553 R28, R28a, R28b, R3 , R30a, R30b are independently selected from the group consisting of H; and C1.6 alkyl, wherein C 1
.
6 alkyl is optionally substituted with one or more halogen, which are the same or different, 5 provided that the following compounds are excluded: H N NN H NS N N N N BrH (1) / , (2) ,(3) IO, H C N N H N CO2Et 0 (4) (3))10 N S\ N N N H OH (6) F ,(7) 10 Compound (1) is known from EP 1 654 706 A as compound [165-1]. Compound (2) is known from WO 2010/055077 A as compound A41. Compound (3) is known from M.E. Swarbrick et al., Bioorganic & Medicinal Chemistry Letters 19 (2009), 4504-4508 as compound 40. Compound (4) is known from GB 2 056 449 A as compound of example 5. Compound (5) is 15 described by the Chemical Abstracts Service with accession number 1185523-72-1. Compound (6) is described by the Chemical Abstracts Service with accession number 861211-24-7. Compound (7) excluded from the scope of the present invention is described in N. Hebert et al., International Journal of Mass Spectrometry and Ion Processes 79 (1986), 45-56; J.C. Blais 20 et al., International Journal of Mass Spectrometry and Ion Processes (1989), 88(1), 29-43 WO 2012/062704 10 PCT/EP2011/069553 In case a variable or substituent can be selected from a group of different variants and such variable or substituent occurs more than once the respective variants can be the same or different. 5 Within the meaning of the present invention the terms are used as follows: "Alkyl" means a straight-chain or branched hydrocarbon chain. Each hydrogen of an alkyl carbon may be replaced by a substituent as further specified. 10 "C 1
.
4 alkyl" means an alkyl chain having 1 - 4 carbon atoms, e.g. if present at the end of a molecule: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, or e.g. CH 2 -, -CH 2
-CH
2 -, -CH(CH 3 )-, -CH 2
-CH
2
-CH
2 -, -CH(C 2
H
5 )-, -C(CH 3
)
2 -, when two moieties of a molecule are linked by the alkyl group. Each hydrogen of a C 1 4 alkyl carbon may be replaced by a substituent as further specified. 15
"C
1
.
6 alkyl" means an alkyl chain having 1 - 6 carbon atoms, e.g. if present at the end of a molecule: C 14 alkyl, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl; tert-butyl, n-pentyl, n-hexyl, or e.g. -CH 2 -, -CH 2
-CH
2 -, -CH(CH 3 )-, -CH 2
-CH
2
-CH
2 -, -CH(C 2
H
5 )-, C(CH 3
)
2 -, when two moieties of a molecule are linked by the alkyl group. Each hydrogen of a 20 C 1 6 alkyl carbon may be replaced by a substituent as further specified.
"C
3 .7 cycloalkyl" or "C 3 .7 cycloalkyl ring" means a cyclic alkyl chain having 3 - 7 carbon atoms, e.g. cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl. Preferably, cyloalkyl refers to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or 25 cycloheptyl. Each hydrogen of a cycloalkyl carbon may be replaced by a substituent as further specified. The term "C 3
.
5 cycloalkyl" or "C 3
.
5 cycloalkyl ring" is defined accordingly. "Halogen" means fluoro, chloro, bromo or iodo. It is generally preferred that halogen is fluoro or chloro. 30 "4 to 7 membered heterocyclyl" or "4 to 7 membered heterocycle" means a ring with 4, 5, 6 or 7 ring atoms that may contain up to the maximum number of double bonds (aromatic or non-aromatic ring which is fully, partially or un-saturated) wherein at least one ring atom up to 4 ring atoms are replaced by a heteroatom selected from the group consisting of sulfur 35 (including -S(O)-, -S(O) 2 -), oxygen and nitrogen (including =N(O)-) and wherein the ring is linked to the rest of the molecule via a carbon or nitrogen atom. Examples for a 4 to 7 WO 2012/062704 11 PCT/EP2011/069553 membered heterocycles are azetidine, oxetane, thietane, furan, thiophene, pyrrole, pyrroline, imidazole, imidazoline, pyrazole, pyrazoline, oxazole, oxazoline, isoxazole, isoxazoline, thiazole, thiazoline, isothiazole, isothiazoline, thiadiazole, thiadiazoline, tetrahydrofuran, tetrahydrothiophene, pyrrolidine, imidazolidine, pyrazolidine, oxazolidine, isoxazolidine, 5 thiazolidine, isothiazolidine, thiadiazolidine, sulfolane, pyran, dihydropyran, tetrahydropyran, imidazolidine, pyridine, pyridazine, pyrazine, pyrimidine, piperazine, piperidine, morpholine, tetrazole, triazole, triazolidine, tetrazolidine, diazepane, azepine or homopiperazine. Accordingly, the term "4 to 7 membered aromatic heterocyclyl" or "4 to 7 membered 10 aromatic heterocycle" refers to a 4 to 7 membered heterocyclic ring which is aromatic. "5 membered aromatic heterocyclyl" or "5 membered aromatic heterocycle" means a heterocycle derived from cyclopentadienyl, where at least one carbon atom is replaced by a heteroatom selected from the group consisting of sulfur (including -S(O)-, -S(0) 2 -), oxygen 15 and nitrogen (including =N(O)-). Examples for such heterocycles are furan, thiophene, pyrrole, imidazole, pyrazole, oxazole, isoxazole, thiazole, isothiazole, thiadiazole, triazole, tetrazole. Preferred compounds of formula (I) are those compounds in which one or more of the 20 residues contained therein have the meanings given below, with all combinations of preferred substituent definitions being a subject of the present invention. With respect to all preferred compounds of the formula (I) the present invention also includes all tautomeric and stereoisomeric forms and mixtures thereof in all ratios, and their pharmaceutically acceptable salts. 25 In preferred embodiments of the present invention, the substituents mentioned below independently have the following meaning. Hence, one or more of these substituents can have the preferred or more preferred meanings given below. 30 Preferably, X 1 is N and X 2 is C(R 15 ). Further preferred is XI=X 2 =N. Preferably, R 1 is T 1 ; or C(O)R 4 . Preferably, R 4 is T 1
.
WO 2012/062704 12 PCT/EP2011/069553 Preferably, T' is cyclopropyl; thiophenyl; isothiazolyl; or pyrazolyl, wherein T is optionally substituted with one or more R 7 (preferably one R 7 ), which are the same or different. 5 Preferably, R 1 is C(O)T 1 , T' is C 3 .7 cycloalkyl, wherein T' is optionally substituted with one or more R 7 (preferably unsubstituted), which are the same or different. More preferably, T' is cyclopropyl. Preferably, R 1 is T', T' is 5 membered aromatic heterocyclyl, wherein T' is optionally 10 substituted with one or more R 7 (preferably one R 7 ), which are the same or different. Preferably, T' is isothiazolyl; or pyrazolyl, wherein T is optionally substituted with one or more R 7 , which are the same or different. 15 Preferably, T' is R 7 ,RI ,R7 NN S , more preferably, , wherein R 7 is defined as indicated above. Preferably, T' is cyclopropyl; or pyrazolyl, wherein T is unsubstituted or substituted with one 20 or more R 7 , which are the same or different. Preferably, R 7 is methyl; CH 2
CH
2 ORSC; CH 2
C(O)T
2 ; or CH 2 C(O)N(R'cRd). Also preferably,
R
7 is methyl; CH 2
CH
2 ORSC or CH 2
C(O)T
2 . RC is preferably H. T 2 is preferably morpholin-4 yl; pyrrolidin-lyl or piperidin-1-yl. Independent of each other, RC and R8d are preferably H 25 or Ci4 alkyl. More preferably, T 2 is morpholin-4-yl. More preferably, RC is H and R'd is ethyl. Even more preferably, R 7 is methyl. Even more preferably, R 7 is CH 2 C(O)N(RcR8d). Preferably, R 15 is CH 3 . 30 Preferably, R 3 is H.
WO 2012/062704 13 PCT/EP2011/069553 Preferably, R 2 is phenyl; pyridyl; pyrimidyl; pyrazolyl; oxadiazolyl; thiazolyl; or thiadiazolyl, wherein R 2 is substituted with one or more R 1 8, which are the same or different. More preferably, R 2 is phenyl, wherein R 2 is substituted with one or more R18. 5 Preferably, R 2 is substituted with one or two R 1 8 . Preferably, R18 is halogen; CN; S(O) 2 N(R1 9 R1 9 a); S(O) 2 R 9; NHS(O) 2 R1 9 a; OR 9 C(O)N(R1 9 aR 9 ); SR19; C 1
.
4 alkyl, unsubstituted or substituted with one or more halogen, which are the same or different; NHC(O)R 9 ; or C(O)R 19 . More preferably, R 18 is halogen; 10 OCH 3 ; S(O) 2
CH
3 ; NHS(O) 2
CH
3 ; or CH 2
NHS(O)
2
CH
3 . More preferably, R 18 is F; Cl; CN; OR 1; S(O) 2 R 9; CI 4 alkyl, unsubstituted or substituted with one R2; S(O) 2
NHR
1 9 ;
S(O)
2
N(CH
3 )R 9; NHS(O) 2 R 9; C(O)NHR 9; C(O)N(CH 3 )R 9; NHC(O)R 9; CF 3 ; SR 9; T , optionally substituted with one substituent selected from the group consisting of methyl and NH2 ;
C(O)T
3 or S(O) 2
T
3 , wherein T 3 is optionally substituted with one substituent selected 15 from the group consisting of methyl and methylcarbonyl; NHC(O)T 3 ; or NHC(O)NHR1 9 a. Even more preferably R 18 is F; Cl; CN; OH; OC1.4 alkyl; S(O)2-C1.4 alkyl; C 1
.
4 alkyl;
CH
2
S(O)
2
CH
3 ; S(O) 2
NH
2 ; S(O) 2
N(CH
3
)
2 ; S(0) 2 NHCI4 alkyl; NH 2 ; NHS(O) 2
CH
3 ; C(O)NH 2 ; C(O)NH-C1.4 alkyl; C(O)N(CH 3
)
2 ; NHC(O)CH 3 ; CF 3 ; SCH 3 ; cyclohexyloxy; hydroxyethyloxy; hydroxyethylaminocarbonyl; methoxyethyloxy; pyrrolidin-1 20 ylcarbonylmethyloxy; aminocarbonylmethylaminocarbonyl; pyrrolidin- 1 -ylcarbonyl; morpholin-4-ylcarbonyl; cyclopentylaminocarbonyl; tetrahydrofuranaminocarbonyl; hydroxyethylaminosulfonyl; methoxyethylaminosulfonyl; aminocarbonylmethylamino 333 sulfonyl; S(O) 2 T or C(O)T 3 , wherein T 3 is optionally substituted with methyl or methylcarbonyl; aminoethylaminocarbonyl; NHC(O)NH-C 1 .4 alkyl; pyrrolidin-1 25 ylcarbonylamino; aminoethyloxy; phenyl; cyclobutyloxy; cyclopentyloxy; cyclopropyloxy; cyclopropylaminocarbonyl; cylobutylaminocarbonyl; 3,3,3 -trifluorpropyloxy; or cyanomethylsulfonylamino. Even more preferably, R18 is F; methyl; ethyl; iso-propyl; OR 9 C(O)NHR 9; or S(O) 2
NHR
9 , wherein R19 is methyl; ethyl; iso-propyl; propyl; or
CH
2
CH
2 OH. 30 Preferably, R 2 is WO 2012/062704 14 PCT/EP2011/069553 F
R
18 or R 18 , wherein R" is defined as indicated above. More preferably, when R 2 is F R 18
R
18 is OR 19 . 5 Compounds of formula (I) in which some or all of the above-mentioned groups have the preferred meanings are also an object of the present invention. Further preferred compounds of the present invention are selected from the group consisting 10 of N-(4-(3-fluorophenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; 4-(2-fluoro-4-methoxyphenyl)-5-methyl-N-(1-methyl-iH-pyrazol-4-yl)pyrimidin-2-amine; N-(4-(2-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; N-(4-(4-chlorophenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; 15 N-(5-methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)cyclopropanecarboxamide; N-(5-methyl-4-(p-tolyl)pyrimidin-2-yl)cyclopropanecarboxamide; N-(4-(4-methoxyphenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; N-(4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; 4-(3-fluoro-4-methoxyphenyl)-5-methyl-N-(1-methyl-iH-pyrazol-4-yl)pyrimidin-2-amine; 20 5-methyl-N-(1-methyl-iH-pyrazol-4-yl)-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-amine; 4-(3-fluoro-4-methoxyphenyl)-5-methyl-N-(1H-pyrazol-4-yl)pyrimidin-2-amine; 5-methyl-4-(4-(methylsulfonyl)phenyl)-N-(1H-pyrazol-4-yl)pyrimidin-2-amine; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)- 1 morpholinoethanone; 25 2-(4-((5 -methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)ethanol; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)ethanol; WO 2012/062704 15 PCT/EP2011/069553 2-(4-((5 -methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1H-pyrazol- I-yl)-I morpholinoethanone; N-(3 -(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzyl)methane sulfonamide; 5 N-methyl-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; N-methyl-3 -(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 4-(2-fluoro-4-(methylsulfonyl)phenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 10 amine; N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)methanesulfonamide; 4-(3 -fluorophenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(4-fluorophenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 15 4-(2-methoxyphenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(4-methoxyphenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 3 -(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(4-(trifluoromethyl)phenyl)pyrimidin-2-amine; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(3 -(methylthio)phenyl)pyrimidin-2-amine; 20 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(4-(methylthio)phenyl)pyrimidin-2-amine; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-phenylpyrimidin-2-amine; 4-(3,4-difluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(3,5-difluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzonitrile; 25 4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; 4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzenesulfonamide; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(3 -(methylsulfonyl)phenyl)pyrimidin-2-amine; 4-(4-(ethylsulfonyl)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(4-(isopropylsulfonyl)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 30 4-(3,4-dimethoxyphenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(3 -chloro-4-methoxyphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(4-chloro-3 -fluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(4-methoxy-3 -(trifluoromethyl)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin 2-amine; WO 2012/062704 16 PCT/EP2011/069553 4-(3 -fluoro-4-methylphenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; N-(2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)acetamide; 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(pyridin-4-yl)pyrimidin-2-amine; 5 2'-methoxy-5-methyl-N-(1-methyl-1H-pyrazol-4-yl)-[4,5'-bipyrimidin]-2-amine; 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(1H-pyrazol-4-yl)pyrimidin-2-amine; 5-methyl-4-(4-(5-methyl- 1,3,4-oxadiazol-2-yl)phenyl)-N-(1-methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; 4-(4-ethoxy-3 -fluorophenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 10 4-(3 -fluoro-4-propoxyphenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(3 -fluoro-4-isopropoxyphenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(4-(cyclohexyloxy)-3 -fluorophenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 2-(2-fluoro-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 15 yl)phenoxy)ethanol; 4-(3 -fluoro-4-(2-methoxyethoxy)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin 2-amine; 2-(2-fluoro-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenoxy)- 1 (pyrrolidin- 1 -yl)ethanone; 20 N-methyl-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; N,N-dimethyl-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; N-ethyl-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; N-isopropyl-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; 4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N-propylbenzamide; 25 N-(2-hydroxyethyl)-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; N-(2-amino-2-oxoethyl)-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; (4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)(pyrrolidin- 1 30 yl)methanone; (4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)(morpholino)methanone; N-cyclopentyl-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; WO 2012/062704 17 PCT/EP2011/069553 4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N-(tetrahydrofuran-3 yl)benzamide; 2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(thiazol-2-yl)pyrimidin-2-amine; 5 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(thiazol-5-yl)pyrimidin-2-amine; 5-(4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)- 1,3,4-thiadiazol 2-amine; N,N-dimethyl-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 10 N-ethyl-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; N-isopropyl-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N 15 propylbenzenesulfonamide; N-(2-hydroxyethyl)-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; N-(2-methoxyethyl)-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 20 2-(4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenylsulfonamido)acetamide; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(4-(pyrrolidin- 1 -ylsulfonyl)phenyl)pyrimidin-2 amine; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(4-(morpholinosulfonyl)phenyl)pyrimidin-2 25 amine; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(4-((4-methylpiperazin- 1 yl)sulfonyl)phenyl)pyrimidin-2-amine; 1-(4-((4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)sulfonyl)piperazin- 1 -yl)ethanone; 30 N-cyclopentyl-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N-(tetrahydrofuran-3 yl)benzenesulfonamide; WO 2012/062704 18 PCT/EP2011/069553 N-(2-methoxyethyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; N-(2-aminoethyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 5 (4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)(4 methylpiperazin- 1 -yl)methanone; 1-(4-(4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzoyl)piperazin- 1 yl)ethanone; N-(4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)-3-methylisothiazol-5-amine; 10 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)-N isopropylacetamide; N-cyclopropyl-2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H pyrazol- 1 -yl)acetamide; N-(2-aminoethyl)-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 15 yl)benzenesulfonamide; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)acetamide; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)-N methylacetamide; 20 N-ethyl-2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)acetamide; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)-N,N dimethylacetamide; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl)-1 25 (piperazin- 1 -yl)ethanone; 2-(4-((4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 -yl)- 1 (piperidin- 1 -yl)ethanone; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)- 1 (pyrrolidin- 1 -yl)ethanone; 30 1 -ethyl-3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)urea; 1 -isopropyl-3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)urea; 1-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)-3 -propylurea; 1-(2-hydroxyethyl)-3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)urea; WO 2012/062704 19 PCT/EP2011/069553 N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)pyrrolidine- 1 carboxamide; 4-(3-fluoro-4-methoxyphenyl)-5-methyl-N-(thiophen-3-yl)pyrimidin-2-amine; N-(2-(2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 5 yl)phenoxy)ethyl)acetamide; 4-(4-(2-aminoethoxy)-3 -fluorophenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 1 -methyl-3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)urea; 1,1 -dimethyl-3-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 10 yl)phenyl)urea; 2-(3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)ureido)acetamide; N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)morpholine-4 carboxamide; 15 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(p-tolyl)pyrimidin-2-amine; 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(m-tolyl)pyrimidin-2-amine; 4-([1,1 '-biphenyl]-4-yl)-5-methyl-N-(1-methyl- IH-pyrazol-4-yl)pyrimidin-2-amine; 4-(3 -fluoro-4-(methylsulfonyl)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 20 4-(4-ethoxyphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 2-(4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenoxy)ethanol; 4-(3 -fluoro-4-(methylthio)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 2-(2-fluoro-4-(5 -methyl-2-((3 -methylisothiazol-5 -yl)amino)pyrimidin-4-yl)phenoxy)ethanol; 25 N-cyclopentyl-4-(5-methyl-2-((3-methylisothiazol-5-yl)amino)pyrimidin-4 yl)benzenesulfonamide; N-ethyl-4-(5-methyl-2-((3-methylisothiazol-5-yl)amino)pyrimidin-4-yl)benzamide; 3-methyl-N-(5-methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)isothiazol-5-amine; 4-(4-ethylphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 30 4-(4-isopropylphenyl)-5-methyl-N-(1 -methyl- IH-pyrazol-4-yl)pyrimidin-2-amine; 2-(4-((4-(3-fluoro-4-(2-hydroxyethoxy)phenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol 1 -yl)- 1 -morpholinoethanone; N-cyclopentyl-4-(5 -methyl-2-((1-(2-morpholino-2-oxoethyl)- 1 H-pyrazol-4 yl)amino)pyrimidin-4-yl)benzenesulfonamide; WO 2012/062704 20 PCT/EP2011/069553 N-ethyl-4-(5-methyl-2-((1-(2-morpholino-2-oxoethyl)- 1H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 2-methoxy-5-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzonitrile; 2-methoxy-5-(5-methyl-2-(( 1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; 5 4-(3 -fluoro-4-methoxyphenyl)-5-methyl-N-(1 -((methylsulfonyl)methyl)- 1H-pyrazol-4 yl)pyrimidin-2-amine; N-ethyl-2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 4-(4-(1H-pyrazol-3 -yl)phenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 10 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 -yl)- 1 -(4 methylpiperazin- 1 -yl)ethanone; 1-(3,5-dimethylpiperazin- 1 -yl)-2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2 yl)amino)- 1H-pyrazol- 1 -yl)ethanone; 1-(1,4-diazepan- 1 -yl)-2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino) 15 1H-pyrazol- 1 -yl)ethanone; 2-(4-((4-(4-ethylphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)- 1 morpholinoethanone; 2-(4-((4-(4-i sopropylphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)- 1 morpholinoethanone; 20 N-isopropyl-4-(5 -methyl-2-((1-(2-morpholino-2-oxoethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin 4-yl)benzamide; N-isopropyl-4-(5 -methyl-2-((1-(2-oxo-2-(pyrrolidin- 1 -yl)ethyl)- 1 H-pyrazol-4 yl)amino)pyrimidin-4-yl)benzamide; 4-(4-(sec-butoxy)-3 -fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 25 amine; 4-(3 -fluoro-4-i sobutoxyphenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(4-cyclobutoxy-3 -fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 4-(4-(cyclopentyloxy)-3 -fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 30 amine; N-(tert-butyl)-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; N-(sec-butyl)-4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; N-isobutyl-4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; WO 2012/062704 21 PCT/EP2011/069553 N-(2-hydroxypropyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 2-(4-((4-(4-ethylphenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1-yl)-1 -(piperidin- 1 yl)ethanone; 5 2-(4-((4-(4-isopropylphenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1-yl)-1 -(piperidin 1 -yl)ethanone; N-ethyl-4-(5 -methyl-2-((1-(2-oxo-2-(piperidin- 1 -yl)ethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin 4-yl)benzamide; N-isopropyl-4-(5 -methyl-2-((1-(2-oxo-2-(piperidin- 1 -yl)ethyl)- 1 H-pyrazol-4 10 yl)amino)pyrimidin-4-yl)benzamide; 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl)-1 morpholinoethanone; 2-(4-((4-(3 -fluoro-4-isopropoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl)-1 morpholinoethanone; 15 N-ethyl-4-(5 -methyl-2-((1-(2-oxo-2-(pyrrolidin- 1 -yl)ethyl)- 1 H-pyrazol-4 yl)amino)pyrimidin-4-yl)benzamide; 1-(2-fluoro-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenoxy)propan-2-ol; 4-(4-cyclopropoxy-3 -fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 20 amine; N-cyclopropyl-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; N-cyclobutyl-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; 2-(4-((4-(3 -fluoro-4-propoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)- 1 morpholinoethanone; 25 4-(3 -fluoro-4-(3,3,3 -trifluoropropoxy)phenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; N-ethyl-2-(4-((4-(4-ethylphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)acetamide; 4-(5 -methyl-2-((1-(2-oxo-2-(piperidin- 1 -yl)ethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 30 4-(5 -methyl-2-((1-(2-morpholino-2-oxoethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; N-ethyl-4-(2-((1-(2-(ethylamino)-2-oxoethyl)- 1 H-pyrazol-4-yl)amino)-5 -methylpyrimidin-4 yl)benzamide; WO 2012/062704 22 PCT/EP2011/069553 4-(2-((1-(2-(ethylamino)-2-oxoethyl)- 1H-pyrazol-4-yl)amino)-5-methylpyrimidin-4-yl)-N isopropylbenzamide; N-ethyl-2-(4-((5 -methyl-4-(4-sulfamoylphenyl)pyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)acetamide; 5 N-ethyl-2-(4-((5 -methyl-4-(4-(morpholinosulfonyl)phenyl)pyrimidin-2-yl)amino)- 1H pyrazol- 1 -yl)acetamide; N-ethyl-2-(4-((5 -methyl-4-(4-(N-(tetrahydrofuran-3 -yl) sulfamoyl)phenyl)pyrimidin-2 yl)amino)- 1H-pyrazol- 1 -yl)acetamide; N-ethyl-2-(4-((4-(4-isopropylphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 10 yl)acetamide; N-ethyl-2-(4-((5-methyl-4-(3 -(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)acetamide; 2-(4-((5-methyl-4-(3 -(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1H-pyrazol- 1-yl)-1 (piperidin- 1 -yl)ethanone; 15 2-(4-((5 -methyl-4-(4-(morpholino sulfonyl)phenyl)pyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl)-1 morpholinoethanone; 4-(5 -methyl-2-((1-(2-morpholino-2-oxoethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N (tetrahydrofuran-3 -yl)benzenesulfonamide; 2-(4-((5 -methyl-4-(3 -(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl)-1 20 morpholinoethanone; 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)-N ethylacetamide; N-ethyl-2-(4-((4-(3 -fluoro-4-isopropoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol 1-yl)acetamide; 25 N-ethyl-2-(4-((4-(3 -fluoro-4-propoxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)acetamide; N-ethyl-2-(4-((4-(3 -fluoro-4-hydroxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)acetamide; 2-(4-((5 -methyl-4-(4-(morpholino sulfonyl)phenyl)pyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl)-1 30 (piperidin- 1 -yl)ethanone; 4-(5 -methyl-2-((1-(2-oxo-2-(piperidin- 1 -yl)ethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N (tetrahydrofuran-3 -yl)benzenesulfonamide; 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl)-1 (piperidin- 1 -yl)ethanone; WO 2012/062704 23 PCT/EP2011/069553 2-(4-((4-(3-fluoro-4-isopropoxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- I-yl)-I (piperidin- 1 -yl)ethanone; 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- I-yl)-I (pyrrolidin- 1 -yl)ethanone; 5 2-(4-((4-(3-fluoro-4-isopropoxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- I-yl)-I (pyrrolidin- 1 -yl)ethanone; 4-(3 -fluoro-4-(2-methoxypropoxy)phenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin 2-amine; 4-(4-ethoxy-3 -fluorophenyl)-N-(1 -isopropyl- 1H-pyrazol-4-yl)-5-methylpyrimidin-2-amine; 10 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(4-methyl-3 -(methylsulfonyl)phenyl)pyrimidin-2 amine; N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)ethanesulfonamide; N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)propane- 1 15 sulfonamide; N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)propane-2 sulfonamide; (R)-N-(sec-butyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 20 (S)-N-(sec-butyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; (R)- 1 -(2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenoxy)propan-2-ol; (S)- 1-(2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 25 yl)phenoxy)propan-2-ol; (S)-4-(4-(sec-butoxy)-3 -fluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; (R)-4-(4-(sec-butoxy)-3 -fluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 30 2-(4-((4-(3 -fluoro-4-hydroxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 -yl)- 1 (pyrrolidin- 1 -yl)ethanone; 2-(4-((4-(3 -fluoro-4-hydroxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 -yl)- 1 (piperidin- 1 -yl)ethanone; 4-(4-ethoxy-3-fluorophenyl)-5-methyl-N-(1H-pyrazol-4-yl)pyrimidin-2-amine; WO 2012/062704 24 PCT/EP2011/069553 N-(4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)-2-methylthiazol-5-amine; 4-(4-(tert-butoxy)-3 -fluorophenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; N-(2-methoxypropyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 5 yl)benzamide; 4-(4-ethoxy-3 -fluorophenyl)-5-methyl-N-(1 -(2-morpholinoethyl)- 1H-pyrazol-4-yl)pyrimidin 2-amine; 3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 -yl)propan 1-01; 10 4-(4-ethoxy-3 -fluorophenyl)-N-(1-ethyl-i H-pyrazol-4-yl)-5 -methylpyrimidin-2-amine; 4-(4-ethoxy-3 -fluorophenyl)-5 -methyl-N-(1 -propyl- 1 H-pyrazol-4-yl)pyrimidin-2-amine; 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)acetonitrile; 4-(4-ethoxy-3 -fluorophenyl)-N-(1 -(2-methoxyethyl)- 1 H-pyrazol-4-yl)-5 -methylpyrimidin-2 15 amine; (S)-3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)propane-1,2-diol; (R)-3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)propane-1,2-diol; 20 4-(3,5-difluoro-4-methoxyphenyl)-5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 4-(4-methoxy-3 -(methyl sulfonyl)phenyl)-5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin 2-amine; 4-(4-ethoxy-3 -fluorophenyl)-N-(1 -(4-methoxybenzyl)- 1 H-pyrazol-4-yl)-5 -methylpyrimidin 25 2-amine; N-(4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)benzenesulfonamide; N-(1 -(2-aminoethyl)- 1 H-pyrazol-4-yl)-4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2 amine; 30 3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)-N ethylpropanamide; 3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)- 1 morpholinopropan- I-one; WO 2012/062704 25 PCT/EP2011/069553 1-(4-((4-(4-ethoxy-3 -fluorophenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1-yl)-2 methylpropan-2-ol; 1 -cyano-N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)methanesulfonamide; and 5 2-hydroxy-N-(4-(5-methyl-2-((1-methyl-iH-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)ethanesulfonamide. Prodrugs of the compounds of the present invention are also within the scope of the present invention. 10 "Prodrug" means a derivative that is converted into a compound according to the present invention by a reaction with an enzyme, gastric acid or the like under a physiological condition in the living body, e.g. by oxidation, reduction, hydrolysis or the like, each of which is carried out enzymatically. Examples of a prodrug are compounds, wherein the amino group 15 in a compound of the present invention is acylated, alkylated or phosphorylated to form, e.g., eicosanoylamino, alanylamino, pivaloyloxymethylamino or wherein the hydroxyl group is acylated, alkylated, phosphorylated or converted into the borate, e.g. acetyloxy, palmitoyloxy, pivaloyloxy, succinyloxy, fumaryloxy, alanyloxy or wherein the carboxyl group is esterified or amidated. These compounds can be produced from compounds of the present invention 20 according to well-known methods. Metabolites of compounds of formula (I) are also within the scope of the present invention. The term "metabolites" refers to all molecules derived from any of the compounds according 25 to the present invention in a cell or organism, preferably mammal. Preferably the term relates to molecules which differ from any molecule which is present in any such cell or organism under physiological conditions. 30 The structure of the metabolites of the compounds according to the present invention will be obvious to any person skilled in the art, using the various appropriate methods. Where tautomerism, e.g. keto-enol tautomerism, of compounds of general formula (I) may occur, the individual forms, e.g. the keto and enol form, are comprised separately and together WO 2012/062704 26 PCT/EP2011/069553 as mixtures in any ratio. The same applies for stereoisomers, e.g. enantiomers, cis/trans isomers, conformers and the like. If desired, isomers can be separated by methods well known in the art, e.g. by liquid 5 chromatography. The same applies for enantiomers by using e.g. chiral stationary phases. Additionally, enantiomers may be isolated by converting them into diastereomers, i.e. coupling with an enantiomerically pure auxiliary compound, subsequent separation of the resulting diastereomers and cleavage of the auxiliary residue. Alternatively, any enantiomer of a compound of formula (I) may be obtained from stereoselective synthesis using optically 10 pure starting materials. The compounds of formula (I) may exist in crystalline or amorphous form. Furthermore, some of the crystalline forms of the compounds of formula (I) may exist as polymorphs, which are included within the scope of the present invention. Polymorphic forms of 15 compounds of formula (I) may be characterized and differentiated using a number of conventional analytical techniques, including, but not limited to, X-ray powder diffraction (XRPD) patterns, infrared (IR) spectra, Raman spectra, differential scanning calorimetry (DSC), thermogravimetric analysis (TGA) and solid state nuclear magnetic resonance (ssNMR). 20 In case the compounds according to formula (I) contain one or more acidic or basic groups, the invention also comprises their corresponding pharmaceutically or toxicologically acceptable salts, in particular their pharmaceutically utilizable salts. Thus, the compounds of the formula (I) which contain acidic groups can be used according to the invention, for 25 example, as alkali metal salts, alkaline earth metal salts or as ammonium salts. More precise examples of such salts include sodium salts, potassium salts, calcium salts, magnesium salts or salts with ammonia or organic amines such as, for example, ethylamine, ethanolamine, triethanolamine or amino acids. Compounds of the formula (I) which contain one or more basic groups, i.e. groups which can be protonated, can be present and can be used according 30 to the invention in the form of their addition salts with inorganic or organic acids. Examples for suitable acids include hydrogen chloride, hydrogen bromide, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acids, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethylacetic acid, malonic acid, succinic acid, pimelic acid, WO 2012/062704 27 PCT/EP2011/069553 fumaric acid, maleic acid, malic acid, sulfaminic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to the person skilled in the art. If the compounds of the formula (I) simultaneously contain acidic and basic groups in the molecule, the invention also includes, in addition to the salt forms mentioned, 5 inner salts or betaines (zwitterions). The respective salts according to the formula (I) can be obtained by customary methods which are known to the person skilled in the art like, for example by contacting these with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with other salts. The present invention also includes all salts of the compounds of the formula (I) which, owing to low physiological 10 compatibility, are not directly suitable for use in pharmaceuticals but which can be used, for example, as intermediates for chemical reactions or for the preparation of pharmaceutically acceptable salts. Throughout the invention, the term "pharmaceutically acceptable" means that the 15 corresponding compound, carrier or molecule is suitable for administration to humans. Preferably, this term means approved by a regulatory agency such as the EMEA (Europe) and/or the FDA (US) and/or any other national regulatory agency for use in animals, preferably in humans. 20 The present invention furthermore includes all solvates of the compounds according to the invention. According to the present invention "JAK" comprises all members of the JAK family (e.g. JAK1, JAK2, JAK3, and TYK2). 25 According to the present invention, the expression "JAK1" or "JAKI kinase" means "Janus kinase 1". The human gene encoding JAKI is located on chromosome lp3l.3. According to the present invention, the expression "JAK2" or "JAK2 kinase" means "Janus 30 kinase 2".The human gene encoding JAK2 is located on chromosome 9p24. According to the present invention, the expression "JAK3" or "JAK3 kinase" means "Janus kinase 3". The gene encoding JAK3 is located on human chromosome 19pl3.1 and it is predominantly in hematopoietic cells. 35 WO 2012/062704 28 PCT/EP2011/069553 According to the present invention, the expression "TYK2" or "TYK2 kinase" means "Protein-Tyrosine kinase 2".The JAK3 and TYK2 genes are clustered on chromosome 19p13.1 and 19pl3.2, respectively. 5 As shown in the examples, compounds of the invention were tested for their selectivity for TYK2 over JAK2 kinases. As shown, all tested compounds bind TYK2 more selectively than, JAK2 (see table 9 below). Consequently, the compounds of the present invention as mentioned above are considered to 10 be useful for the prevention or treatment of diseases and disorders associated with TYK2, for example immunological, inflammatory, autoimmune, or allergic disorders, transplant rejection, Graft-versus-Host-Disease or proliferative diseases such as cancer. In a preferred embodiment, the compounds of the present invention are selective TYK2 15 inhibitors. The compounds of the present invention may be further characterized by determining whether they have an effect on TYK2, for example on its kinase activity (Fridman et al 2010. J. Immunology 2010 184(9):5298-307). 20 A cell-based assay was described to assess the inhibitory activity of small molecule drugs toward TYK2-dependent signal transduction (Bacon et al 1995. PNAS 92, 7307-7311; W02009155551). 25 The present invention provides pharmaceutical compositions comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof as active ingredient together with a pharmaceutically acceptable carrier, optionally in combination with one or more other pharmaceutical compositions. 30 "Pharmaceutical composition" means one or more active ingredients, and one or more inert ingredients that make up the carrier, as well as any product which results, directly or indirectly, from combination, complexation or aggregation of any two or more of the ingredients, or from dissociation of one or more of the ingredients, or from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical WO 2012/062704 29 PCT/EP2011/069553 compositions of the present invention encompass any composition made by admixing a compound of the present invention and a pharmaceutically acceptable carrier. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the 5 therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, including but not limited to peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered orally. Saline and aqueous dextrose are preferred carriers when the pharmaceutical composition is administered 10 intravenously. Saline solutions and aqueous dextrose and glycerol solutions are preferably employed as liquid carriers for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor 15 amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsions, tablets, pills, capsules, powders, sustained release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, 20 sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" by E.W. Martin. Such compositions will contain a therapeutically effective amount of the therapeutic, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration. 25 A pharmaceutical composition of the present invention may comprise one or more additional compounds as active ingredients like one or more compounds of formula (I) not being the first compound in the composition or other JAK inhibitors. Further bioactive compounds may be steroids, leukotriene antagonists, cyclosporine or rapamycin. 30 The compounds of the present invention or pharmaceutically acceptable salt(s) thereof and the other pharmaceutically active agent(s) may be administered together or separately and, when administered separately, this may occur separately or sequentially in any order. When combined in the same formulation it will be appreciated that the two compounds must be WO 2012/062704 30 PCT/EP2011/069553 stable and compatible with each other and the other components of the formulation. When formulated separately they may be provided in any convenient formulation, conveniently in such manner as are known for such compounds in the art. 5 It is further included within the present invention that the compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a compound of formula (I) is administered in combination with another drug or pharmaceutically active agent and/or that the pharmaceutical composition of the invention further comprises such a drug or pharmaceutically active agent. 10 In this context, the term "drug or pharmaceutically active agent" includes a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician. 15 "Combined" or "in combination" or "combination" should be understood as a functional coadministration, wherein some or all compounds may be administered separately, in different formulations, different modes of administration (for example subcutaneous, intravenous or oral) and different times of administration. The individual compounds of such combinations may be administered either sequentially in separate pharmaceutical 20 compositions as well as simultaneously in combined pharmaceutical compositions. For example, in rheumatoid arthritis therapy, combination with other chemotherapeutic or antibody agents is envisaged. Suitable examples of pharmaceutically active agents which may be employed in combination with the compounds of the present invention and their salts for 25 rheumatoid arthritis therapy include: immunosuppresants such as amtolmetin guacil, mizoribine and rimexolone; anti-TNFa agents such as etanercept, infliximab, Adalimumab, Anakinra, Abatacept, Rituximab; tyrosine kinase inhibitors such as leflunomide; kallikrein antagonists such as subreum; interleukin 11 agonists such as oprelvekin; interferon beta 1 agonists; hyaluronic acid agonists such as NRD-101 (Aventis); interleukin 1 receptor 30 antagonists such as anakinra; CD8 antagonists such as amiprilose hydrochloride; beta amyloid precursor protein antagonists such as reumacon; matrix metalloprotease inhibitors such as cipemastat and other disease modifying anti-rheumatic drugs (DMARDs) such as methotrexate, sulphasalazine, cyclosporin A, hydroxychoroquine, auranofin, aurothioglucose, gold sodium thiomalate and penicillamine.
WO 2012/062704 31 PCT/EP2011/069553 In particular, the treatment defined herein may be applied as a sole therapy or may involve, in addition to the compounds of the invention, conventional surgery or radiotherapy or chemotherapy. Accordingly, the compounds of the invention can also be used in combination 5 with existing therapeutic agents for the treatment proliferative diseases such as cancer. Suitable agents to be used in combination include: (i) antiproliferative/antineoplastic drugs and combinations thereof, as used in medical oncology such as alkylating agents (for example cis-platin, carboplatin, cyclophosphamide, 10 nitrogen mustard, melphalan, chlorambucil, busulphan and nitrosoureas); antimetabolites (for example antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, hydroxyurea and gemcitabine); antitumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, daunomycin, epirubicin, idarubicin, mitomycin-C, dactinomycin and mithramycin); antimitotic agents (for example 15 vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like paclitaxel and taxotere); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, topotecan and camptothecins); (ii) cytostatic agents such as antioestrogens (for example tamoxifen, toremifene, raloxifene, 20 droloxifene and iodoxyfene), oestrogen receptor down regulators (for example fulvestrant), antiandrogens (for example bicalutamide, flutamide, nilutamide and cyproterone acetate), LHIRH antagonists or LHIRH agonists (for example goserelin, leuprorelin and buserelin), progestogens (for example megestrol acetate), aromatase inhibitors (for example as anastrozole, letrozole, vorazole and exemestane) and inhibitors of 5a-reductase such as 25 finasteride; (iii) anti-invasion agents (for example c-Src kinase family inhibitors like 4-(6-chloro- 2,3 methylenedioxyanilino)-7- [2-(4-methylpiperazin- 1 -yl)ethoxy] -5 -tetrahydropyran- 4-yloxy quinazoline (AZD0530) and N-(2-chloro-6-methylphenyl)-2- { 6- [4-(2 30 hydroxyethyl)piperazin-1-yl]-2-methylpyrimidin- 4-ylamino }thiazole-5 -carboxamide (dasatinib, BMS-354825), and metalloproteinase inhibitors like marimastat and inhibitors of urokinase plasminogen activator receptor function); WO 2012/062704 32 PCT/EP2011/069553 (iv) inhibitors of growth factor function: for example such inhibitors include growth factor antibodies and growth factor receptor antibodies (for example the anti-erbB2 antibody trastuzumab [HerceptinTM] and the anti-erbBl antibody cetuximab [C225]); such inhibitors also include, for example, tyrosine kinase inhibitors, for example inhibitors of the epidermal 5 growth factor family (for example EGFR family tyrosine kinase inhibitors such as N-(3 chloro-4-fluorophenyl)-7-methoxy-6-(3-morpholinopropoxy)quinazolin-4-amine (gefitinib, ZD 1839), A/-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)quinazolin-4-amine (erlotinib, OSI-774) and 6-acrylamido-A/-(3-chloro-4-fluorophenyl)-7-(3- morpholinopropoxy) quinazolin-4-amine (CI 1033) and erbB2 tyrosine kinase inhibitors such as lapatinib), 10 inhibitors of the hepatocyte growth factor family, inhibitors of the platelet-derived growth factor family such as imatinib, inhibitors of serine/threonine kinases (for example Ras/Raf signalling inhibitors such as farnesyl transferase inhibitors, for example sorafenib (BAY 43 9006)) and inhibitors of cell signalling through MEK and/or Akt kinases; 15 (v) antiangiogenic agents such as those which inhibit the effects of vascular endothelial growth factor, for example the anti-vascular endothelial cell growth factor antibody bevacizumab (AvastinTM) and VEGF receptor tyrosine kinase inhibitors such as 4-(4-bromo 2-fiuoroanilino)-6-methoxy-7-( 1 -methylpiperidin-4-ylmethoxy)quinazoline (ZD6474; Example 2 within WO 01/32651), 4-(4-fluoro-2-methylindol-5-yloxy)-6-methoxy-7-(3 20 pyrrolidin-1-ylpropoxy)quinazoline (AZD2171; Example 240 within WO 00/47212), vatalanib (PTK787; WO 98/35985) and SUl 1248 (sunitinib; WO 01/60814), and compounds that work by other mechanisms (for example linomide, inhibitors of integrin avP3 function and angiostatin); 25 (vi) vascular damaging agents such as combretastatin A4 and compounds disclosed in International Patent Application WO 99/02166; (vii) antisense therapies, for example those which are directed to the targets listed above, such as ISIS 2503, an anti-ras antisense agent; 30 (viii) gene therapy approaches, including approaches to replace aberrant genes such as aberrant p53 or aberrant BRCAl or BRCA2, GDEPT (gene-directed enzyme pro-drug therapy) approaches such as those using cytosine deaminase, thymidine kinase or a bacterial nitroreductase enzyme and approaches to increase patient tolerance to chemotherapy or WO 2012/062704 33 PCT/EP2011/069553 radiotherapy such as multi-drug resistance gene therapy; and (ix) immunotherapeutic approaches, including ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, 5 approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies. Further combination treatments are described in WO-A 2009/008992 and WO-A 10 2007/107318, incorporated herein by reference. Accordingly, the individual compounds of such combinations may be administered either sequentially in separate pharmaceutical compositions as well as simultaneously in combined pharmaceutical compositions. 15 The pharmaceutical compositions of the present invention include compositions suitable for oral, rectal, topical, parenteral (including subcutaneous, intramuscular, and intravenous), ocular (ophthalmic), pulmonary (nasal or buccal inhalation), or nasal administration, although the most suitable route in any given case will depend on the nature and severity of the 20 conditions being treated and on the nature of the active ingredient. They may be conveniently presented in unit dosage form and prepared by any of the methods well-known in the art of pharmacy. In practical use, the compounds of formula (I) can be combined as the active ingredient in 25 intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral (including intravenous). In preparing the compositions for oral dosage form, any of the usual pharmaceutical media may be employed, such as water, glycols, oils, alcohols, flavoring agents, preservatives, coloring 30 agents and the like in the case of oral liquid preparations, such as, for example, suspensions, elixirs and solutions; or carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations such as powders, hard and soft capsules and tablets, with the solid oral preparations being preferred over the liquid preparations.
WO 2012/062704 34 PCT/EP2011/069553 Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit form in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be coated by standard aqueous or non-aqueous techniques. Such 5 compositions and preparations should contain at least 0.1 percent of active compound. The percentage of active compound in these compositions may, of course, be varied and may conveniently be between about 2 percent to about 60 percent of the weight of the unit. The amount of active compound in such therapeutically useful compositions is such that an effective dosage will be obtained. The active compounds can also be administered 10 intranasally, for example, as liquid drops or spray. The tablets, pills, capsules, and the like may also contain a binder such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid; a lubricant such as magnesium stearate; and a 15 sweetening agent such as sucrose, lactose or saccharin. When a dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier such as fatty oil. Various other materials may be present as coatings or to modify the physical form of the dosage unit. For instance, tablets may be coated with shellac, sugar or both. A syrup or elixir 20 may contain, in addition to the active ingredient, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and a flavoring such as cherry or orange flavor. Compounds of formula (I) may also be administered parenterally. Solutions or suspensions of these active compounds can be prepared in water suitably mixed with a surfactant such as 25 hydroxypropyl-cellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or 30 dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, WO 2012/062704 35 PCT/EP2011/069553 water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils. Any suitable route of administration may be employed for providing a mammal, especially a 5 human, with an effective dose of a compound of the present invention. For example, oral, rectal, topical, parenteral, ocular, pulmonary, nasal, and the like may be employed. Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like. Preferably compounds of formula (I) are administered orally. 10 The effective dosage of active ingredient employed may vary depending on the particular compound employed, the mode of administration, the condition being treated and the severity of the condition being treated. Such dosage may be ascertained readily by a person skilled in the art. 15 A therapeutically effective amount of a compound of the present invention will normally depend upon a number of factors including, for example, the age and weight of the animal, the precise condition requiring treatment and its severity, the nature of the formulation, and the route of administration. However, an effective amount of a compound of formula (I) for 20 the treatment of an inflammatory disease, for example rheumatoid arthritis (RA), will generally be in the range of 0.1 to 100 mg/kg body weight of recipient (mammal) per day and more usually in the range of 1 to 10 mg/kg body weight per day. Thus, for a 70 kg adult mammal, the actual amount per day would usually be from 70 to 700 mg and this amount may be given in a single dose per day or more usually in a number (such as two, three, four, five or 25 six) of sub-doses per day such that the total daily dose is the same. An effective amount of a pharmaceutically acceptable salt, prodrug or metabolite thereof, may be determined as a proportion of the effective amount of the compound of formula (I) per se. It is envisaged that similar dosages would be appropriate for treatment of the other conditions referred to above. 30 As used herein, the term "effective amount" means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician.
WO 2012/062704 36 PCT/EP2011/069553 Furthermore, the term "therapeutically effective amount" means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder. The term also includes within its 5 scope amounts effective to enhance normal physiological function. Another aspect of the present invention is a compound of the present invention or a pharmaceutically acceptable salt thereof for use as a medicament as mentioned above. 10 Another aspect of the present invention is a compound of the present invention or a pharmaceutically acceptable salt thereof for use in a method of treating or preventing a disease or disorder associated with TYK2 as mentioned above. In the context of the present invention, a disease or disorder associated with TYK2 is defined 15 as a disease or disorder where TYK2 is involved. In a preferred embodiment, wherein the diseases or disorder is associated with TYK2 is an immunological, inflammatory, autoimmune, or allergic disorder or disease of a transplant rejection or a Graft-versus host disease. 20 Consequently, another aspect of the present invention is a compound or a pharmaceutically acceptable salt thereof of the present invention for use in a method of treating or preventing an immunological, inflammatory, autoimmune, or allergic disorder or disease of a transplant rejection or a Graft-versus host disease. 25 Inflammation of tissues and organs occurs in a wide range of disorders and diseases and in certain variations, results from activation of the cytokine family of receptors. Exemplary inflammatory disorders associated with activation of TYK2 include, in a non-limiting manner, skin inflammation due radiation exposure, asthma, allergic inflammation and chronic 30 inflammation. In a preferred embodiment, the inflammatory disease is an eye disease.
WO 2012/062704 37 PCT/EP2011/069553 Dry eye syndrome (DES, also known as keratoconjunctivitis sicca) is one of the most common problems treated by eye physicians. Sometimes DES is referred to as dysfunctional tear syndrome (Jackson, 2009. Canadian Journal Ophthalmology 44(4), 385-394). DES affects up to 10% of the population between the ages of 20 to 45 years, with this percentage 5 increasing with age. Although a wide variety of artificial tear products are available, these products provide only transitory relief of symptoms. As such, there is a need for agents, compositions and therapeutic methods to treat dry eye. As used herein, "dry eye disorder" is intended to encompass the disease states summarized in 10 a recent official report of the Dry Eye Workshop (DEWS), which defined dry eye as "a multifactorial disease of the tears and ocular surface that results in symptoms of discomfort, visual disturbance, and tear film instability with potential damage to the ocular surface. It is accompanied by increased osmolality of the tear film and inflammation of the ocular surface." (Lemp, 2007. "The Definition and Classification of Dry Eye Disease: Report of the Definition 15 and Classification Subcommittee of the International Dry Eye Workshop", The Ocular Surface, 5(2), 75-92). Dry eye is also sometimes referred to as keratoconjunctivitis sicca. In some embodiments, the treatment of the dry eye disorder involves ameliorating a particular symptom of dry eye disorder, such as eye discomfort, visual disturbance, tear film instability, tear hyperosmolarity, and inflammation of the ocular surface. 20 Uveitis is the most common form of intraocular inflammation and remains a significant cause of visual loss. Current treatments for uveitis employs systemic medications that have severe side effects and are globally immunosuppressive. Clinically, chronic progressive or relapsing forms of non-infectious uveitis are treated with topical and/or systemic corticosteroids. In 25 addition, macrolides such as cyclosporine and rapamycin are used, and in some cases cytotoxic agents such as cyclophosphamide and chlorambucil, and antimetabolites such as azathioprine, methotrexate, and leflunomide (Srivastava et al., 2010. Uveitis: Mechanisms and recent advances in therapy. Clinica Chimica Acta, doi:10.1016/j.cca.2010.04.017). 30 Further eye diseases, combination treatments and route of administration are described for example in WO-A 2010/039939, which is hereby incorporated herein by reference. According to the present invention, an autoimmune disease is a disease which is at least partially provoked by an immune reaction of the body against own components, for example WO 2012/062704 38 PCT/EP2011/069553 proteins, lipids or DNA. Examples of organ-specific autoimmune disorders are insulin dependent diabetes (Type I) which affects the pancreas, Hashimoto's thyroiditis and Graves' disease which affect the thyroid gland, pernicious anemia which affects the stomach, Cushing's disease and Addison's disease which affect the adrenal glands, chronic active 5 hepatitis which affects the liver; polycystic ovary syndrome (PCOS), celiac disease, psoriasis, inflammatory bowel disease (IBD) and ankylosing spondylitis. Examples of non-organ specific autoimmune disorders are rheumatoid arthritis, multiple sclerosis, systemic lupus and myasthenia gravis. 10 Type I diabetes ensues from the selective aggression of autoreactive T-cells against insulin secreting beta-cells of the islets of Langerhans. In a preferred embodiment, the autoimmune disease is selected from the group consisting of rheumatoid arthritis (RA), inflammatory bowel disease (IBD; Crohns's disease and ulcerative 15 colitis), psoriasis, systemic lupus erythematosus (SLE), and multiple sclerosis (MS). Rheumatoid arthritis (RA) is a chronic progressive, debilitating inflammatory disease that affects approximately 1% of the world's population. RA is a symmetric polyarticular arthritis that primarily affects the small joints of the hands and feet. In addition to inflammation in the 20 synovium, the joint lining, the aggressive front of tissue called pannus invades and destroys local articular structures (Firestein 2003, Nature 423:356-361). Inflammatory bowel disease (IBD) is characterized by a chronic relapsing intestinal inflammation. IBD is subdivided into Crohn's disease and ulcerative colitis phenotypes. 25 Crohn disease involves most frequently the terminal ileum and colon, is transmural and discontinuous. In contrast, in ulcerative colitis, the inflammation is continuous and limited to rectal and colonic mucosal layers. In approximately 10% of cases confined to the rectum and colon, definitive classification of Crohn's disease or ulcerative colitis cannot be made and are designated 'indeterminate colitis.' Both diseases include extraintestinal inflammation of the 30 skin, eyes, or joints. Neutrophil-induced injuries may be prevented by the use of neutrophils migration inhibitors (Asakura et al., 2007, World J Gastroenterol. 13(15):2145-9). Psoriasis is a chronic inflammatory dermatosis that affects approximately 2% of the population. It is characterized by red, scaly skin patches that are usually found on the scalp, WO 2012/062704 39 PCT/EP2011/069553 elbows, and knees, and may be associated with severe arthritis. The lesions are caused by abnormal keratinocyte proliferation and infiltration of inflammatory cells into the dermis and epidermis (Schon et al., 2005, New Engl. J. Med. 352:1899-1912). 5 Systemic lupus erythematosus (SLE) is a chronic inflammatory disease generated by T cell mediated B-cell activation, which results in glomerulonephritis and renal failure. Human SLE is characterized at early stages by the expansion of long-lasting autoreactive CD4+ memory cells (D'Cruz et al., 2007, Lancet 369(9561):587-596). 10 Multiple sclerosis (MS) is an inflammatory and demyelating neurological disease. It has bee considered as an autoimmune disorder mediated by CD4+ type 1 T helper cells, but recent studies indicated a role of other immune cells (Hemmer et al., 2002, Nat. Rev. Neuroscience 3, 291-301). 15 Transplant rejection (allograft transplant rejection) includes, without limitation, acute and chronic allograft rejection following for example transplantation of kidney, heart, liver, lung, bone marrow, skin and cornea. It is known that T cells play a central role in the specific immune response of allograft rejection. Hyperacute, acute and chronic organ transplant rejection may be treated. Hyperacute rejection occurs within minutes of transplantation. Acute 20 rejection generally occurs within six to twelve months of the transplant. Hyperacute and acute rejections are typically reversible where treated with immunosuppressant agents. Chronic rejection, characterized by gradual loss of organ function, is an ongoing concern for transplant recipients because it can occur anytime after transplantation. 25 Graft-versus-host disease (GVDH) is a major complication in allogeneic bone marrow transplantation (BMT). GVDH is caused by donor T cells that recognize and react to recipient differences in the histocompatibility complex system, resulting in significant morbidity and mortality. 30 In a further preferred embodiment, the disease or disorder associated with TYK2 is a proliferative disease, especially cancer as mentioned above. Diseases and disorders associated especially with TYK2 are proliferative disorders or diseases, especially cancer.
WO 2012/062704 40 PCT/EP2011/069553 Therefore, another aspect of the present invention is a compound or a pharmaceutically acceptable salt thereof of the present invention for use in a method of treating or preventing a proliferative disease, especially cancer. 5 Cancer comprises a group of diseases characterized by uncontrolled growth and spread of abnormal cells. All types of cancers generally involve some abnormality in the control of cell growth, division and survival, resulting in the malignant growth of cells. Key factors contributing to said malignant growth of cells are independence from growth signals, 10 insensitivity to anti-growth signals, evasion of apoptosis, limitless replicative potential, sustained angiogenesis, tissue invasion and metastasis, and genome instability (Hanahan and Weinberg, 2000. The Hallmarks of Cancer. Cell 100, 57-70). Typically, cancers are classified as hematological cancers (for example leukemias and 15 lymphomas) and solid cancers such as sarcomas and carcinomas (for example cancers of the brain, breast, lung, colon, stomach, liver, pancreas, prostate, ovary). The TYK2 inhibitors of the present invention may also be useful in treating certain malignancies, including skin cancer and hematological malignancy such as lymphomas and 20 leukemias. Yet another aspect of the present invention is the use of a compound of the present invention or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment or prophylaxis of diseases and disorders associated with TYK2. 25 Yet another aspect of the present invention is the use of a compound of the present invention or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing an immunological, inflammatory, autoimmune, or allergic disorder or disease or a transplant rejection or a Graft-versus host disease. 30 Yet another aspect of the present invention is the use of a compound of the present invention or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for treating or preventing a proliferative disease, especially cancer.
WO 2012/062704 41 PCT/EP2011/069553 In the context of these uses of the invention, diseases and disorders associated with TYK2 are as defined above. Yet another aspect of the present invention is a method for treating, controlling, delaying or 5 preventing in a mammalian patient in need thereof one or more conditions selected from the group consisting of diseases and disorders associated with TYK2, wherein the method comprises the administration to said patient a therapeutically effective amount of a compound according to present invention or a pharmaceutically acceptable salt thereof 10 Yet another aspect of the present invention is a method for treating, controlling, delaying or preventing in a mammalian patient in need thereof one or more conditions selected from the group consisting of an immunological, inflammatory, autoimmune, or allergic disorder or disease or a transplant rejection or a Graft-versus host disease, wherein the method comprises the administration to said patient a therapeutically effective amount of a compound according 15 to present invention or a pharmaceutically acceptable salt thereof. Yet another aspect of the present invention is a method for treating, controlling, delaying or preventing in a mammalian patient in need thereof a proliferative disease, especially cancer, wherein the method comprises the administration to said patient a therapeutically effective 20 amount of a compound according to present invention or a pharmaceutically acceptable salt thereof In the context of these methods of the invention, diseases and disorders associated with TYK2 are as defined above. 25 As used herein, the term "treating" or "treatment" is intended to refer to all processes, wherein there may be a slowing, interrupting, arresting, or stopping of the progression of a disease, but does not necessarily indicate a total elimination of all symptoms. 30 Exemplary routes for the preparation of compounds of the present invention are described below. It is clear to a practitioner in the art to combine or adjust such routes especially in combination with the introduction of activating or protective chemical groups.
WO 2012/062704 42 PCT/EP2011/069553 A general route for the preparation of compounds according to present invention is outlined in Schemes 1 and 2. CI NH2 N IkIN N -N Nl N
R
3 cl R 3 R2 R 3 R2
NH
2 HN'R N N N N
R
3 R2 R 3 XR2 5 (i) R 2
B(OH)
2 , Pd(dppf)(Cl 2 ).DCM, EtOH, 2M Na 2
CO
3 , Dioxane, 130 0 C, 30min; (ii) anuonia, dioxane, 160 0 C, 1h; (iii) R 1 C(O)Cl, triethylamine, ACN, 1h. Scheme 1 CI CI N -N - N N RI3 cl R 3 R2 CI HNT HNT-R7 N N ____N _ _ -N N R 3 rR2
R
3 R2
R
3 R2 10 (i) R 2
B(OH)
2 , Pd(dppf)(C1 2 ).DCM, EtOH, 2M Na 2
CO
3 , Dioxane, 130 0 C, 30min; (ii) HN-T', IPA, HCl (4M dioxane), 150 0 C, 1h, microwave; (iii) R 7 C(O)Cl, K 2
CO
3 , Nal, ACN, 80 0 C, 1h. Scheme 2 15 Examples Analytical Methods NMR spectra were obtained on a Brucker dpx400. LCMS (methods A and B) was carried out 20 on an Agilent 1100 using a Gemini C18, 3 x 30 mm, 3 micron column. Solvents used were water and acetonitrile with 0.1% formic acid and with an injection volume of 3 p.L. Wavelengths were 254 and 210 nm. LCMS method C was carried out on a Waters uPLC SQD using the same solvents. Photodiode array detection was between 210 and 400 nm.
WO 2012/062704 43 PCT/EP2011/069553 Methods D-G were carried out on an Agilent 1200 series IPLC with detection at 254 nm. Water (with 0.07% TFA) and methanol were the solvents used. Method A 5 Column: Phenomenex Gemini-C 18, 3 x 30mm, 3microns. Flow rate: 1.2 mL/min Table 1 Time (min) Water (%) ACN (%) 0 95 5 3 5 95 4.5 5 95 4.6 95 5 5 STOP Method B 10 Column: Phenomenex Gemini-C18, 4.6 x 150mm, microns. Flow rate: 1.0 mL/min Table 2 Time (min) Water (%) ACN (%) 0.00 95.0 5.0 11.00 5.0 95.0 13.00 5.0 95.0 13.01 95.0 5.0 14.00 STOP Method C 15 Column: Waters Acquity UPLC BEH C18, 2.1 x 30 mm, 1.7 microns. Flow rate: 0.5 mL/min Table 3 Time (min) Water ACN (%) (%) 0.00 95.0 5.0 0.20 95.0 5.0 1.00 5.0 95.0 1.50 5.0 95.0 WO 2012/062704 44 PCT/EP2011/069553 1.70 95.0 5.0 2.70 95.0 5.0 3.00 STOP Method D Column: Ultimate AQ-C18 column, 4.6x250 mm, 5 microns. Flow rate: 1.0 mL/min at 30'C. 5 Table 4 Time (min) Methanol (%) Water (%) 0 40 60 3 40 60 5 60 40 7 80 20 8 95 5 15 95 5 17 STOP Method E Column: Ultimate AQ-C18 column, 4.6x250 mm, 5 microns. Flow rate: 1.0 mL/min at 30'C. 10 Table 5 Time (min) Methanol (%) Water (%) 0 20 80 5 20 80 8 65 35 10 95 5 14 95 5 17 STOP Method F Column: Waters Novapak C18, 3.9 x 150 mm, 4 microns. Flow rate: 1.0 mL/min. Gradient as described in Table 4 (Method D) 15 Method G Column: Waters Novapak C18, 3.9 x 150 mm, 4 microns. Flow rate: 1.0 mL/min. Gradient as described in Table 5 (Method E) 20 Table 6: Abbreviations ACN Acetonitrile WO 2012/062704 45 PCT/EP2011/069553 Ar Aryl aq Aqueous br Broad Boc Tert-Butoxycarbonyl BuLi Butyllithium d Doublet dba Dibenzylideneacetone DCM Dichloromethane dd Double doublet ddd Double doublet of doublets DDQ 2,3-Dichloro-5,6-dicyano-1,4-benzoquinone DEAD Diethyl azodicarboxylate DIAD Diisopropyl azodicarboxylate DIPEA Diisopropylethylamine DME 1,2-Dimethoxyethane DMF NN'-Dimethylformamide DMF-DMA N,N'-dimethylformamide dimethylacetal DMSO NN'-dimethylsulfoxide DP Drug pulldown dppf 1,1 '-Bis(diphenylphosphino)ferrocene dt Doublet of triplets DTT Dithiothreitol EDC 1 -Ethyl-3-(3-dimethylaminopropyl)carbodiimide EDTA Ethylenediaminetetraacetic acid EtOAc Ethyl acetate EtOH Ethanol eq Equivalents g Grams h Hours HATU O-(7-Azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate HCl Hydrochloric acid
H
2 0 Water WO 2012/062704 46 PCT/EP2011/069553
H
2 S Hydrogen sulfide HOBt 1 -Hydroxybenzotriazole HPLC High performance liquid chromatography
IC
50 50% inhibition concentration IPA Isopropyl alcohol iPr Isopropyl L Litres LC-MS Liquid chromatography mass spectroscopy m Multiplet M Molar MeOH Methanol Mesyl Methanesulfonyl chloride mg Milligrams MgSO 4 Magnesium Sulphate min Minutes mL Millilitres mm Millimetres mmol Millimoles mol% Molar percent [tL Microlitres nm Nanometres NMR Nuclear magnetic resonance PBS Phosphate buffered saline Prep. Preparative q Quartet rpm Revolutions per minute rt Room temperature RT Retention time s Singlet sat. Saturated t Triplet td Triplet of doublets tdd Triple doublet of doublets WO 2012/062704 47 PCT/EP2011/069553 tert Tertiary TFA Trifluoroacetic acid Tf Tetrahydrofuran tt Triplet of triplets Xantphos 4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene Experimental Procedure A 5 Step (i) 2,4-dichloro-5-methylpyrimidine (300 mg, 1.8 mmol), (4-(methylsulfonyl)phenyl)boronic acid (0.36 mL, 1.8 mmol), [1,1'-Bis(diphenylphosphino)ferrocene]dichloropalladium(II), complex with dichloromethane (44 mg, 0.054 mmol) and sodium carbonate (2M aqueous solution, 2.7 mL, 5.4 mmol) in ethanol (1 mL) and dioxane (3 mL) were heated to 130'C for 10 30 min. The residue was cooled to rt and partitioned between EtOAc and water. The phases were separated and the aqueous phase was extracted with EtOAc. The combined organic extracts were washed with brine, dried (MgSO 4 ) and the solvents removed in vacuo to afford a brown oil. This was purified by flash column chromatography to afford 2-chloro-5-methyl 4-(4-(methyl sulfonyl)phenyl)pyrimidine. 15 Step (ii) 2-chloro-5 -methyl-4-(4-(methylsulfonyl)phenyl)pyrimidine (112 mg, 0.4 mmol) and ammonia (5 mL, 0.06 mmol) in dioxane (0.75 mL) were heated to 160'C for 1 h. The mixture was concentrated in vacuo to afford 5-methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-amine as 20 a white solid which was used in the next step without further purification. Step (iii) Cyclopropanecarbonyl chloride (84 [tL, 0.91 mmol) was added to a stirred solution of 5 methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-amine (120 mg, 0.46 mmol) and 25 triethylamine (53 tL, 0.91 mmol) in ACN (10 mL). The mixture was stirred for a further 1 h then the solvent was removed in vacuo and water then DCM were added. The organic phase was separated and the solvent was removed in vacuo to afford a yellow oil which was purified by prep. HPLC to afford N-(5-methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2 yl)cyclopropanecarboxamide.
WO 2012/062704 48 PCT/EP2011/069553 Procedure B General Procedure for the Synthesis of 4-Amino-1-N-alkylated-pyrazoles Step (i) 5 A solution of 4-nitropyrazole (300mg, 2.65mmol), potassium carbonate (2eq) and the alkylating reagent (1.1 eq) in acetonitrile (10mL) was heated to 60'C for 18 h. After cooling to rt the mixture was diluted with EtOAc and washed with water. The organic phase was collected, dried (MgSO 4 ) and concentrated in vacuo. 10 Step (ii) The crude residue was dissolved in methanol (10mL), palladium on carbon (50mg) was added and the reaction was stirred under a balloon of hydrogen for 18 h. The resulting mixture was filtered through Celite and the filtrate concentrated in vacuo to give the desired product. 15 Procedure C Step (i) 2,4-dichloro-5-methylpyrimidine (300 mg, 1.8 mmol), (4-(methylsulfonyl)phenyl)boronic acid (0.36 mL, 1.8 mmol), [1,1'-Bis(diphenylphosphino)ferrocene]dichloropalladium(II), complex with dichloromethane (44 mg, 0.054 mmol) and sodium carbonate (2M aqueous 20 solution, 2.7 mL, 5.4 mmol) in ethanol (1 mL) and dioxane (3 mL) were heated to 130'C for 30 min. The residue was cooled to rt and partitioned between EtOAc and water. The phases were separated and the aqueous phase was extracted with EtOAc. The combined organic extracts were washed with brine, dried (MgSO 4 ) and the solvents removed in vacuo to afford a brown oil. This was purified by flash column chromatography to afford 2-chloro-5-methyl 25 4-(4-(methyl sulfonyl)phenyl)pyrimidine. Step (ii) 2-chloro-5 -methyl-4-(4-(methylsulfonyl)phenyl)pyrimidine (70 mg, 0.25 mmol), 1-methyl 1H-pyrazol-4-amine (26 mg, 0.27 mmol), prepared according to Procedure B, IPA (3 mL) and 30 HCl (4M in dioxane, 100[tL, 0.4 mmol) were heated to 150'C for 1 h in the microwave. The solvent was removed in vacuo and the brown oil purified by prep HPLC to afford 5-methyl N-(1-methyl-iH-pyrazol-4-yl)-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-amine. Procedure D WO 2012/062704 49 PCT/EP2011/069553 General procedure for alkylation of N-(1H-pyrazol-4-yl)pyrimidin-2-amines N-(1H-pyrazol-4-yl)pyrimidin-2-amine (1.1 eq), alkyl chloride (leq) potassium carbonate (2eq), sodium iodide (catalytic) and ACN were heated to 80'C for 1 h. The mixture was cooled to rt and EtOAc then water were added. The organic phase was separated, washed 5 with brine, dried (MgSO 4 ) and concentrated in vacuo to afford a yellow oil. Petrol was added and the resulting precipitate was separated by filtration to give the desired product as a white solid. 10 Procedure E Step (i) HCl (4M in dioxane, 200 mL) was added slowly to a stirred solution of 1-methyl-1H-pyrazol 4-amine (52 g, 0.53 mol, prepared according to Procedure B) and cyanamide (29 g, 0.69 mol) in dioxane (50 mL) then the mixture was heated to 95'C overnight. The reaction was cooled 15 to room temperature and the solvent removed in vacuo. The residue was washed with ether (100 mL) to afford 1-(1-methyl-1H-pyrazol-4-yl)guanidine hydrochloride (93 g , ~ 100%) as a yellow solid which was used in the next step without further purification. Step (ii) 20 Ethyl propionate (35 mL, 0.31 mol) was added dropwise to a stirred solution of sodium methoxide (28 g, 0.51 mol) in anhydrous DMF (300 mL) then the mixture was stirred for 20 min before the dropwise addition of methyl formate (14 mL, 0.23 mol). The reaction was stired for 30 min before 1-(1-methyl-1H-pyrazol-4-yl)guanidine hydrochloride (38 g, 0.21 mol) was added and the reaction heated at 1 00 0 C overnight. The mixture was concentrated in 25 vacuo, diluted with water and adjusted to pH6-7 by addition of 4M hydrochloric acid. The precipitate was collected and washed with H 2 0 (100 mL) and ethyl acetate (50 mL) to afford 5-methyl-2-(1-methyl-iH-pyrazol-4-ylamino)pyrimidin-4-ol (35 g, 79%) as a grey solid which was used in next step without further purification. 30 Step (iii) 5-methyl-2-(1-methyl-iH-pyrazol-4-ylamino)pyrimidin-4-ol (35 g, 0.17 mol) was dissolved in POC1 3 (25 mL) and the mixture was heated at 95'C for 5 h then concentrated in vacuo, poured onto ice water (100 mL) and adjusted to pH7-8 by addition of saturated aqueous NaHCO 3 . The precipitate was collected by filtration, dried in vacuo and purified by column WO 2012/062704 50 PCT/EP2011/069553 chromatography (silica, 3-5% v/v MeOH in DCM) to afford 4-chloro-5-methyl-N-(1-methyl 1H-pyrazol-4-yl)pyrimidin-2-amine (15.0 g, 39%) as a white solid. Procedure F 5 Step (i) IM NaOH (aq) (0.93 mol) was added to a stirred solution of ethyl 2-(4-(4-(3-fluoro-4 methoxyphenyl)-5 -methylpyrimidin-2-ylamino)- 1 H-pyrazol- 1 -yl)acetate (115 mg, 0.31 mmol, prepared according to procedure B step (i)) in MeOH (10 mL) and the mixture was stirred at rt for 3 h. After the reaction was complete, the solvent was removed in vacuo and 10 the residue was acidified by addition of dilute HCl and extracted with EtOAc. The combined organic extracts were dried (Na 2
SO
4 ) and concentrated in vacuo to afford 2-(4-(4-(3-fluoro-4 methoxyphenyl)-5-methyl pyrimidin-2-ylamino)- 1 H-pyrazol- 1 -yl)acetic acid (100 mg, 91%) as a white solid. 15 Step (ii) HATU (99 mg, 0.26 mmol) and triethylamine (52 mg, 0.51 mmol) were added to a stirred solution of 2-(4-(4-(3-fluoro-4-methoxyphenyl)-5-methyl pyrimidin-2-ylamino)-1H-pyrazol 1-yl)acetic acid (60 mg, 0.17 mmol, 1 eq) in ACN (25 mL) and the mixture was stirred at rt for 0.5 h before addition of cyclopropylamine (12 mg, 0.20 mmol). After 3 h the solvent was 20 removed in vacuo, the residue was diluted with saturated aqueous Na 2
CO
3 and extracted with EtOAc. The combined organic extracts were dried (Na 2
SO
4 ) ,concentrated in vacuo and purified by column chromatography (silica, 0-2%, v/v MeOH in DCM) to afford 2-(4-(4-(3 fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-ylamino)- 1 H-pyrazol- 1 -yl)-N cyclopropylacetamide (21 mg, 3 1%) as a yellow solid. 25 Procedure G A mixture of 2-fluoro-4-(5-methyl-2-(1-methyl-iH-pyrazol-4-ylamino)pyrimidin-4-yl)phenol (60 mg, 0.2 mmol), cyclobutyl bromide (32 mg, 0.24 mmol) and Cs 2
CO
3 (130 mg, 0.4 mmol) in acetonitrile (4 mL) was heated at 160'C in microwave reactor for 30 min. The mixture was 30 partitioned between water and ethyl acetate, the organic layer was washed with brine, dried (Na 2
SO
4 ), concentrated and purifed by preparative TLC (5% v/v MeOH in DCM) to afford 4 (4-cyclobutoxy-3-fluorophenyl)-5-methyl-N-(1-methyl-iH-pyrazol-4-yl)pyrimidin-2-amine (40 mg, 56%) as a white solid. 35 Procedure H WO 2012/062704 51 PCT/EP2011/069553 A mixture of 2-fluoro-4-(5-methyl-2-(1-methyl-iH-pyrazol-4-ylamino)pyrimidin-4-yl)phenol (60 mg, 0.2 mmol), propylene oxide (58 mg, 1.0 mmol), lithium chloride (25 mg, 0.6 mmol) and triethylamine (2 mg, 0.02 mmol) in ethanol was heated at 60'C in a sealed tube overnight. The mixture was concentrated in vacuo, dissolved in ethyl acetate, washed with brine, dried 5 (Na 2
SO
4 ), concentrated and purifed by preparative TLC (5% v/v MeOH in DCM) to obtain 1 (2-fluoro-4-(5-methyl-2-(1-methyl-iH-pyrazol-4-ylamino)pyrimidin-4-yl)phenoxy)propan-2 ol (20 mg, 28%) as a light yellow solid. Procedure I 10 Step (i) TFA (2 mL) was added to a solution of tert-butyl 4-(5-methyl-2-(1-methyl-1H-pyrazol-4 ylamino)pyrimidin-4-yl)phenylcarbamate (420 mg, 1.1 mmol) in DCM (5 mL). The mixture was stirred at rt overnight then concentrated in vacuo. The residue was suspended in 5% NaHCO 3 (aq) and extracted twice with DCM. The combined organic layer was washed with 15 brine, dried (Na 2
SO
4 ), and concentrated to give the crude product which was purifed by column chromatography (silica, 5% v/v MeOH in DCM) to afford 4-(4-aminophenyl)-5 methyl-N-(1-methyl-1H-pyrazol-4-yl)pyrimidin-2-amine (250 mg, 79%) as a white solid. Step (ii) 20 Ethylsulfonyl chloride (46 mg, 0.36 mmol) was added to a stirred solution of 4-(4 aminophenyl)-5-methyl-N-(1-methyl-iH-pyrazol-4-yl)pyrimidin-2-amine (100 mg, 0.36 mmol) in pyridine (4 mL) and the reaction was stirred at rt for 3 h. The pyridine was removed under reduced pressure and the residue was extracted with DCM, washed with water and brine, dried (Na 2
SO
4 ) and concentrated to give the crude product which was purified by 25 preparative TLC (5% v/v MeOH in DCM) to afford N-(4-(5-methyl-2-(1-methyl-1H-pyrazol 4-ylamino)pyrimidin-4-yl)phenyl) ethanesulfonamide (35 mg, 26%). Procedure J Step (i) 30 Phenylchloroformate (209 mg, 1.3 mmol) was added dropwise to a stirred mixture of 4-(4 aminophenyl)-5-methyl-N-(1-methyl-iH-pyrazol-4-yl)pyrimidin-2-amine (250 mg, 0.89 mmol, prepared according to Procedure I(i)) and NaHCO 3 (224 mg, 2. 7 mmol) in THF (10 mL) and the mixture was stirred at rt for 3 h. The solvent was removed in vacuo and the residue was purified by column chromatography (silica, 5% v/v MeOH in DCM) to afford WO 2012/062704 52 PCT/EP2011/069553 phenyl 4-(5-methyl-2-(1-methyl-iH-pyrazol-4-ylamino)pyrimidin-4-yl)phenylcarbamate (320 mg, 90%) as a white solid. Step (ii) 5 A mixture of 4-(5-methyl-2-(1-methyl-iH-pyrazol-4-ylamino)pyrimidin-4 yl)phenylcarbamate (80 mg, 0.2 mmol) and 2-methoxyethanamine (30 mg, 0.4 mmol) in THF (3 mL) was heated to 120'C for 15 min in a microwave reactor. The solvent was removed in vacuo and the residue was extracted with ethyl acetate. The combined organic layer was washed with water, dried (Na 2
SO
4 ) and the crude product was purifed by column 10 chromatography (silica, 3% v/v MeOH in DCM) to afford 1-(2-methoxyethyl)-3-(4-(5 methyl-2-(1-methyl-iH-pyrazol-4-ylamino)pyrimidin-4-yl)phenyl)urea (30 mg, 40%) as a white solid. Procedure K 15 A mixture of 2-chloro-4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidine (70 mg, 0.28 mmol), 3-methylisothiazol-5-amine (39 mg, 0.34 mmol), Cs 2
CO
3 (182 mg, 0.56 mmol), Pd 2 dba 3 (28 mg, 0.03 mmol) and Xantphos (35 mg, 0.06 mmol) in dioxane (14 mL) was purged with nitrogen and then heated at 130'C for 5 min in a microwave reactor. The solvent was removed in vacuo and the residue was dissolved in EtOAc. The organic layer was washed 20 with water and brine, dried (Na 2
SO
4 ) and concentrated to afford a crude residue which was purified by column chromatography (silica, 1-2% v/v MeOH in DCM) to afford 4-(3-fluoro 4-methoxyphenyl)-5-methyl-N-(3-methylisothiazol-5-yl)pyrimidin-2-amine (18 mg, 20%) as a brown solid. 25 Procedure L Step (i) To a solution of pentafluorophenol (5.0 g, 27 mmol) in DCM (220 mL) was added triethylamine (11 mL, 82 mmol). A solution of 4-bromophenylsulfonyl chloride (6.9 g, 27 mmol) in DCM (50 mL) was then added and the reaction stirred at rt overnight. The mixture 30 was washed with 5% citric acid (2x100 mL) and brine (2x50 mL), dried (Na 2
SO
4 ) filtered and concentrated to afford perfluorophenyl 4-bromobenzenesulfonate (10.2 g, 93.6%) as a white solid. Step (ii) WO 2012/062704 53 PCT/EP2011/069553 A mixture of perfluorophenyl 4-bromobenzenesulfonate (3.3 g, 8.2 mmol), bis(pinacolato)diboron (2.29 g, 9.0 mmol), Pd(dppf)C1 2 (669 mg, 0.82 mmol), dppf (454 mg, 0.82 mmol), and sodium acetate (4.0 g, 49 mmol) in dioxane (116 mL) was heated to reflux under a nitrogen atmosphere for 16 h. The reaction was then cooled and the solid precipitate 5 removed by filtration. The filtrate was concentrated to give a crude product which was boiled in hexane (40 mL). Any further solid that precipitated was removed by filtration, then the filtrate was collected and concentrated in vacuo to give perfluorophenyl 4-(4,4,5,5 tetramethyl-1,3,2-dioxaborolan-2-yl)benzenesulfonate (980 mg, 27%) as a pale red solid. 10 Step (iii) A mixture of perfluorophenyl 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2 yl)benzenesulfonate (890 mg, 2.0 mmol), 4-chloro-5-methyl-N-(1-methyl-iH-pyrazol-4 yl)pyrimidin-2-amine (340 mg, 1.5 mmol, prepared by Procedure E), Pd(dppf)C1 2 (163 mg, 1.5 mmol) and Na 2
B
4 0 7 (2.4 g, 12 mmol) in dioxane (23 mL) and ethanol (4.5 mL) was 15 heated to 120'C for 15 min in a microwave reactor. The resulting mixture was concentrated under reduced pressure to give a residue which was purifed by column chromatography (silica, 2% v/v MeOH in DCM) to afford perfluorophenyl 4-(5-methyl-2-(1-methyl-1H pyrazol-4-ylamino)pyrimidin-4-yl)benzenesulfonate (700 mg, 91%). 20 Step (iv) A mixture of perfluorophenyl 4-(5-methyl-2-(1-methyl-iH-pyrazol-4-ylamino)pyrimidin-4 yl)benzenesulfonate (30 mg, 0.087 mmol), morpholine (9 mg, 0.10 mmol) and triethylamine (2 mL) in dioxane (8 mL) was heated to 120'C for 15 min in a microwave reactor. The mixture was concentrated to give a residue which was purified by preparative fHPLC to 25 provide 5-methyl-N-(1-methyl-iH-pyrazol-4-yl)-4-(4-(morpholinosulfonyl)phenyl)pyrimidin 2-amine (11.6 mg, 48%). Procedure M Step (i) 30 n-BuLi (2.0 M in hexane, 2.1 mL, 4.2 mmol) was added dropwise over 5 min to a stirred solution of 2-bromopyridine (675 mg, 4.2 mmol) in dry ether (15 mL) at -45'C and the reaction was stirred at this temperature for a further 1 h. A suspension of 2-chloro-5 methylpyrimidine (500 mg, 3.9 mmol) in ether (30 mL) was added dropwise over 5 min, the reaction was stirred a further 15 min at -45'C, warmed to 0 0 C and stirred for 1 h. The reaction WO 2012/062704 54 PCT/EP2011/069553 was quenched by addition of water (100 mg) in THF (3 mL) and then DDQ (1.32 g, 5.8 mmol) in THF (5 mL) was added. The reaction was warmed to rt and stirred for a further 15 min. It was then cooled to 0 0 C and hexane (10 mL) was added followed by 3M aqueous sodium hydroxide (2.5 mL, 7.5 mmol). The suspension was stirred for 5 min, extracted with 5 ethyl acetate, concentrated in vacuo then purified by column chromatography to give a mixture of the deisred 2-chloro-5-methyl-4-(pyridin-2-yl)pyrimidine and starting material 2 chloro-5-methylpyrimidine. The mixture was dissolved in EtOAc and extracted with IM HCl. The aqueous layer was collected and made basic by addition of NaOH solution and extracted with EtOAc. The organics were concentrated in vacuo afford 2-chloro-5-methyl-4-(pyridin-2 10 yl)pyrimidine (220 mg, 28%) as a white solid. Step (ii) N-methyl-4-aminopyrazole (62 mg, 0.64 mmol) and 12M HCl (0.15 mL, 1.7 mmol) were added to a stirred solution of 2-chloro-5-methyl-4-(pyridin-2-yl)pyrimidine (100mg, 0.49 15 mmol), in IPA (3 mL) then the mixture was heated to 160'C for 45 min in a microwave reactor. The reaction was quenched by addition of water and made basic with sodium hydroxide solution. The resulting mixture was extracted with EtOAc and the combined extracts were dried, concentrated and purified by preparative TLC (5% v/v MeOH in DCM) followed by an ethanol rinse to afford 5-methyl-N-(1-methyl-1H-pyrazol-4-yl)-4-(pyridin-2 20 yl)pyrimidin-2-amine (28 mg, 22%) as a yellow solid. Procedure N Step (i) A mixture of 2-chloro-4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidine (300 mg, 1.2 25 mmol) and concentrated aqueous ammonia (2 mL) in EtOH (10 mL) was heated to 160'C in a sealed tube overnight. The solvent was removed in vacuo and the residue purified by column chromatography (silica, 0-10% v/v MeOH in DCM) to afford 4-(3-fluoro-4-methoxyphenyl) 5-methylpyrimidin-2-amine (150 mg, 54%) as a white solid. 30 Step (ii) Perbromomethane (1.07 g, 3.2 mmol) was added to a stirred mixture of 2-acetamidoacetic acid (377 mg, 3.2 mmol) in pyridine (10 mL) at 0 0 C under N 2 . The reaction was stirred for 10 min then triphenylphosphine (845 mg, 3.2 mmol) was added portionwise and the whole mixture was stirred for 1 h at rt. 4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-amine WO 2012/062704 55 PCT/EP2011/069553 (150 mg, 0.64 mmol) was added and the whole mixture was stirred overnight. The solvent was removed in vacuo, the residue was diluted with water and extracted twice with EtOAc. The combined organic layer was dried (Na 2
SO
4 ), concentrated and purified by column chromatography (silica, 0-5%, v/v MeOH in DCM) to afford 2-acetamido-N-[4-(3-fluoro-4 5 methoxyphenyl)-5-methylpyrimidin-2-yl]acetamide (60 mg, 28%) as a white solid. Step (iii) A mixture of 2-acetamido-N-[4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl] acetamide (40 mg, 0.12 mmol) and Lawesson's Reagent (243 mg, 0.60 mmol) in xylene (20 10 mL) was heated to 150'C overnight. The solvent was removed in vacuo and the residue was purified by preparative HPLC and then preparative TLC (5% v/v MeOH in DCM) to afford 4 (3-fluoro-4-methoxyphenyl)-5-methyl-N-(2-methyl- 1,3-thiazol-5-yl)pyrimidin-2-amine (2.0 mg, 5%) as a white solid. 15 Example 1: N-(4-(3-fluorophenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide 0 HN N N F Synthesised according to Procedure A using (3-fluorophenyl)boronic acid. 1H NMR (400 20 MVUlz, CDCl 3 ) 6 8.62 (s, 1H), 8.54 (s, 1H), 7.49 (m, 2H), 7.38 - 7.31 (m, 1H), 7.23 - 7.12 (m, 1H), 2.35 (s, 3H), 1.74 (m, 1H), 1.25 - 1.14 (m, 2H), 1.00 - 0.83 (m, 2H); LCMS method B, (ES+) 272, RT = 8.37 min.
WO 2012/062704 56 PCT/EP2011/069553 Example 2: 4-(2-fluoro-4-methoxyphenyl)-5-methyl-N-(]-methyl-]H-pyrazol-4-yl)pyrimidin-2 amine
N
HN F N N Synthesised according to Procedure C using (2-fluoro-4-methoxyphenyl)boronic acid. 1 H NMR (400 MHz, d 6 -DMSO) 6 9.36 (s, 1H), 8.34 (s, 1H), 7.80 (s, 1H), 7.46-7.43 (m, 2H), 7.00 - 6.94 (m, 2H), 3.85 (s, 3H), 3.78 (s, 3H), 2.02 (s, 3H); LCMS method B, (ES+) 314, RT = 8.79 min. Example 3: N-(4-(2-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)cyclopropane carboxamide 0 HN F N N Synthesised according to Procedure A using (2-fluoro-4-methoxyphenyl)boronic acid. 1 H NMR (400 MHz, d 6 -DMSO) 6 10.76 (s, 1H), 8.52 (s, 1H), 7.37 (s, 1H), 6.95 - 6.86 (m, 2H), 3.79 (s, 3H), 2.18 - 1.89 (m, 4H), 1.32 - 1.04 (m, 1H), 0.90 - 0.54 (m, 4H); LCMS method B, (ES+) 302, RT = 8.47 min.
WO 2012/062704 57 PCT/EP2011/069553 Example 4: N-(4-(4-chlorophenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide 0 HN N N CI Synthesised according to Procedure A using (4-chlorophenyl)boronic acid. 1H NMR (400 MHz, d 6 -DMSO) 6 10.81 (s, 1H), 8.58 (m, 1H), 7.73-7.71 (m, 2H), 7.60-7.58 (m, 2H), 2.29 2.28 (m, 3H), 2.14-2.07 (m, 1H), 0.82-0.78 (m, 4H); LCMS method B, (ES+) 288, RT = 9.09 min. Example 5: N-(5-methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2 yl)cyclopropanecarboxamide 0 HN N N 0 Synthesised according to Procedure A using (4-(methylsulfonyl)phenyl)boronic acid. 1 H NMR (400 MHz, d 6 -DMSO) 6 10.86 (s, 1H), 8.64 (m, 1H), 8.07 (d, J= 8.6 Hz, 2H), 7.93 (d, J= 8.6 Hz, 2H), 3.29 (s, 3H), 2.29 (s, 3H), 2.13 - 2.10 (m, 1H), 0.85 - 0.77 (m, 4H); LCMS method A, (ES+) 332, RT = 0.89min. Example 6: N-(5-methyl-4-(p-tolyl)pyrimidin-2-yl)cyclopropanecarboxamide 0 HN 1 N N Synthesised according to Procedure A using p-tolylboronic acid. 1H NMR (400 MHz, d 6
-
WO 2012/062704 58 PCT/EP2011/069553 DMSO) 6 10.74 (s, 1H), 8.53 (m, 1H), 7.59 (d, 2H), 7.32 (d, 2H), 2.38 (s, 3H), 2.29 (s, 3H), 2.16-2.10 (m, 1H), 0.82-0.77 (m, 4H); LCMS method B, (ES+) 268, RT = 8.70 min. Example 7: N-(4-(4-methoxyphenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide 0 HN N N Synthesised according to Procedure A using (4-methoxyphenyl)boronic acid. 1H NMR (400 MVUlz, d 6 -DMSO) 6 10.73 (s, 1H), 8.51 (s, 1H), 7.71 (d, 2H), 7.06 (d, 2H), 3.84 (s, 3H), 2.32 (s, 3H), 2.14 (m, 1H), 0.91 - 0.71 (m, 4H); LCMS method B, (ES+) 284, RT = 7.88 min. Example 8: N-(4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2 yl)cyclopropanecarboxamide 0 HN N N 01 10 F Synthesised according to Procedure A using (3-fluoro-4-methoxyphenyl)boronic acid. 1 H NMR (400 MVUlz, d 6 -DMSO) 6 10.76 (s, 1H), 8.55-8.53 (m, 1H), 7.64-7.54 (m, 2H), 7.32-7.26 (m, 1H), 3.94-3.89 (m, 3H), 2.35-2.30 (m, 3H), 2.14-2.09 (m, 1H), 0.83-0.78 (m, 4H); LCMS method B, (ES+) 302, RT = 8.26 min.
WO 2012/062704 59 PCT/EP2011/069553 Example 9: 4-(3-fluoro-4-methoxyphenyl)-5-methyl-N-(]-methyl-]H-pyrazol-4-yl)pyrimidin-2 amine
N
HN N N F O 0 Synthesised according to Procedure C using (3-fluoro-4-methoxyphenyl)boronic acid. 1 H NMR (400 MVUlz, d 6 -DMSO) 6 9.32 (s, 1H), 8.32 (s, 1H), 7.83 (s, 1H), 7.59-7.49 (m, 3H), 7.33-7.28 (m, 1H), 3.93 (s, 3H), 3.79 (s, 3H), 2.23 (m, 3H); LCMS method B, (ES+) 314, RT = 8.53 min. Example 10: 5-methyl-N-(]-methyl-]H-pyrazol-4-yl)-4-(4-(methylsulfonyl)phenyl)pyrimidin-2 amine
N
HN N N 0 Synthesised according to Procedure C using (4-(methylsulfonyl)phenyl)boronic acid. 1 H NMR (400 MHz, d 6 -DMSO) 6 9.43 (s, 1H), 8.41 (s, 1H), 8.08 (d, 2H), 7.94 (d, 2H), 7.83 (s, 1H), 7.49 (s, 1H), 3.79 (s, 3H), 3.31(s, 3H), 2.20 (s, 3H); LCMS method B, (ES+) 344, RT = 7.10 min. Example 11: 4-(3-fluoro-4-methoxyphenyl)-5-methyl-N-(]H-pyrazol-4-yl)pyrimidin-2-amine HN N N F C 0 WO 2012/062704 60 PCT/EP2011/069553 Synthesised according to Procedure C using (3-fluoro-4-methoxyphenyl)boronic acid in step (i) and 1H-pyrazol-4-amine hydrochloride in Step (ii). 1H NMR (400 Miz, d 6 -DMSO) 6 12.40 (s, 1H), 9.30 (s, 1H), 8.32 (s, 1H), 7.87 (br s, 1H), 7.60 - 7.53 (m, 3H), 7.31 (m, 1H), 3.93 (s, 3H), 2.23 (s, 3H); LCMS method A, (ES+) 300, RT = 0.97 min. Example 12: 5-methyl-4-(4-(methylsulfonyl)phenyl)-N-(]H-pyrazol-4-yl)pyrimidin-2-amine NH HN Z N N -IIS"C 0 Synthesised according to Procedure C using (4-(methylsulfonyl)phenyl)boronic acid in Step (i) and 1H-pyrazol-4-amine hydrochloride in Step (ii). 1 H NMR (400 Miz, d 6 -DMSO) 6 12.41 (s, 1H), 9.41 (s, 1H), 8.41 (s, 1H), 8.09 - 8.05 (m, 2H), 7.94 (br d, 2H), 7.87 (br s, 1H), 7.58 (br s, 1H), 3.32 (s, 3H), 2.14 (s, 3H); LCMS method A, (ES+) 330, RT = 0.87 min. Example 13: 2-(4-((4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)amino)-]H-pyrazol ]-yl)-]-morpholinoethanone 0 N N 0 N HN N N F) 0 The title product was synthesised according to Procedure D using 4-(3-fluoro-4 methoxyphenyl)-5-methyl-N-(1H-pyrazol-4-yl)pyrimidin-2-amine (see Example 11) and 2 chloro-1-morpholinoethanone as alkylating agent. 1H NNMR (400 MVUlz, d 6 -DMSO) 6 9.42 (s, 1H), 8.38 (s, 1H), 7.89 (s, 1H), 7.71 - 7.48 (m, 3H), 7.35 (t, J= 8.7 Hz, 1H), 5.12 (s, 2H), 3.97 (s, 3H), 3.70 - 3.43 (m, 8H), 2.28 (s, 3H); LCMS method B, (ES+) 428, RT = 7.72 min.
WO 2012/062704 61 PCT/EP2011/069553 Example 14: 2-(4-((5-methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)-]H-pyrazol ]-yl)ethanol H N OH NN N N S 0 Synthesised according to Procedures B and C. 1 H NMR (400 MHz, d 6 -DMSO) 6 9.43 (s, 1H), 8.41 (s, 1H), 8.14 - 8.03 (m, 2H), 7.95 (d, J= 8.1 Hz, 2H), 7.89 (s, 1H), 7.53 (s, 1H), 4.87 (t, J = 5.5 Hz, 1H), 4.08 (t, J= 5.5 Hz, 2H), 3.70 (m, 2H), 3.31 (s, 3H), 2.21 (s, 3H); LCMS method A, (ES+) 374, RT = 0.82 min. Example 15: 2-(4-((4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)amino)-]H-pyrazol ]-yl)ethanol HN OH HN N N FO 0 Synthesised according to Procedures B and C. 1 H NMR (400 MHz, d 6 -DMSO) 6 9.31 (s, 1H), 8.32 (s, 1H), 7.95 - 7.82 (m, 1H), 7.66 - 7.46 (m, 3H), 7.29 (t, J= 8.7 Hz, 1H), 4.85 (br s, 1H), 4.07 (t, J= 5.6 Hz, 2H), 3.92 (s, 3H), 3.70 (t, J= 5.6 Hz, 2H), 2.22 (s, 3H); LCMS method B, (ES+) 344, RT = 7.56 min.
WO 2012/062704 62 PCT/EP2011/069553 Example 16: 2-(4-((5-methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)-]H-pyrazol ]-yl)-]-morpholinoethanone 0 N N 0 HN N N 0 The title product was synthesised according to Procedure D using 5-methyl-4-(4 (methylsulfonyl)phenyl)-N-(1H-pyrazol-4-yl)pyrimidin-2-amine (see Example 12) and 2 chloro-1-morpholinoethanone as alkylating agent. 1H NMR (400 MVUlz, d 6 -DMSO) 6 9.48 (s, 1H), 8.42 (s, 1H), 8.07 (d, 2H), 7.94 (d, 2H), 7.83 (s, 1H), 7.56 (s, 1H), 5.08 (s, 2H), 3.66-3.52 (m, 4H), 3.52-3.39 (m, 4H), 3.31 (s, 3H), 2.20 (s, 3H); LCMS method B, (ES+) 458, RT = 6.64 min. Example 17: N-(3-(5-methyl-2-((]-methyl-]H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzy)methanesulfonamide N-N N NH N O%-NH 0 Synthesised according to Procedure C using ((3-(methylsulfonamidomethyl)phenyl)boronic acid. 1 H NNMR (400 MVUlz, d 6 -DMSO) 6 9.37 (s, 1H), 8.35 (s, 1H), 7.87 (s, 1H), 7.67 - 7.46 (m, 6H), 4.25 (d, 2H), 3.78 (s, 3H), 2.89 (s, 3H), 2.19 (s, 3H); LCMS method C, (ES+) 374, RT = 0.87 min.
WO 2012/062704 63 PCT/EP2011/069553 Example 18: N-methyl-4-(5-methyl-2-((]-methyl-]H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide H NN N NN NH Synthesised according to Procedure C using (4-(N-methylsulfamoyl)phenyl)boronic acid. 1 H NMR (400 MHz, d 6 -DMSO) 6 9.42 (s, 1H), 8.39 (s, 1H), 7.90 (s, 4H), 7.83 (s, 1H), 7.60 7.56 (m, 1H), 7.48 (s, 1H), 3.78 (s, 3H), 2.48 (s, 3H), 2.19 (s, 3H); LCMS method C, (ES+) 360, RT = 0.87 min. Example 19: N-methyl-3-(5-methyl-2-((]-methyl-]H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide H N N NN SI N'NH 0 \ Synthesised according to Procedure C using (3-(N-methylsulfamoyl)phenyl)boronic acid. 1 H NMR (400 MHz, d 6 -DMSO) 6 9.46 (s, 1H), 8.40 (s, 1H), 8.10 - 8.08 (m, 1H), 7.98 - 7.96 (m, 1H), 7.91 - 7.88 (m, 2H), 7.76 - 7.62 (m, 1H), 7.62 - 7.58 (m, 1H), 7.47 (s, 1H), 3.78 (s, 3H), 2.47 (s, 3H), 2.22 (s, 3H); LCMS method C, (ES+) 360, RT = 0.88 min. Examples 20-219 are given in Table 7 together with an indication of the procedures used to prepare them. The order of events is not implied and a person skilled in the art would understand that more than one order of events could lead to the desired product. Table 7 Example Synthetic Analytical RT MS Procedure Method (min) ES+ WO 2012/062704 64 PCT/EP2011/069553 NN N N'N 20 Nz- N N Z NBC A 2.19 362 HN H N-N F
I>
0 HN N N/N H N N 214 BCE A 12.11 396 N-Ns N HNAN 22 BCE E 15.27 284 N-N F HN N 23 BCE D 12.80 284 F N-N N HN I N 24 BCE D 12.11 296 CF3 N-N HNN 25 HN NBCE D 12.29 296 N-N N (N N-N 0 NH 2 HNAN 27 C3BCE D 13.49 334 N -N WO 2012/062704 65 PCT/EP2011/069553 HN N 28 BCE D 13.37 312 N-N N 29 BCE D 13.23 312 N-N HNN 30 BCE D 12.54 266 N-N N' HN N 31 BCE D 13.19 302 N-N F N' I HN N F~ 32 BCE D 13.30 302 N-N F N HN N 33 BCE D 12.25 291 CN N-N N' HNAN 34 BCE D 10.10 309 0 N-N
NH
2 HNN N N 35 1 NH 2 BCE D 9.93 345 N-N O WO 2012/062704 66 PCT/EP2011/069553 HNN 36 BCE B 7.25 344 N-N O S 0 NA HN N 37 HN ,i- BCE D 11.54 358 N-N O HNA N N 38 BCE D 12.05 372 N-N HNA 39 0 BCE D 11.70 326 N-N 0 N' HNA N 40 BCE D 13.22 330 :-1 0 N-N CI N' HN N 41 BCE D 13.62 318 'CI N-N F HN N 42 BCE D 13.36 364 N-N CF 3 N HN N 43 BCE D 13.46 298 N-N F WO 2012/062704 67 PCT/EP2011/069553 N HNAN 44 ABCE D 11.43 341 N-N F HNN N N 45 N BCE D 4.75 267 N-N N HN N N 46 NtO BCE D 11.39 298 N-N HN 47 H NH BCEI D 5.09 256 N-N N' 48 BCE D 12.30 348 N-N N-N N' HN N 49 BCG D 13.17 328 N-N F HN N 50 BCG D 13.60 342 N-N F N H N ljl N 51 BCG D 13.41 342 N-N F WO 2012/062704 68 PCT/EP2011/069553 HNAN 52 BCG D 14.28 382 N-N F N HN N 53 BCG D 11.45 344 N-N F N' HNAN 54 BCG D 12.56 358 N-N F NN 55 | N BCFG D 12.16 411 N-N F 0 N' 56 H BCF D 10.84 323 N-N 0 N 57 BCF D 11.16 337 N-N 0 N HNAN 58 H BCF D 11.55 337 N-N 0 N HNAN 59 H BCF D 12.09 351 N -N N N-N 0 WO 2012/062704 69 PCT/EP2011/069553 N', HN N 60 BCF D 12.20 351 N-N 0 N HN N 61 H BCF D 10.17 353 N-N 0 N HN N O 62 2H BCF D 9.16 366 Z /11! N,, ,NH 2 N-N 0 N HN N 63 N BCF D 12.06 363 N-N 0 N HN N O 64 N BCF D 11.03 379 - N N-N 0 N HN N 65 H BCF D 12.84 377 7z I N N-N O N HN N 66 H BCF D 11.40 379 N-N O N' HN N 67 BCEF D 10.82 327 0 N-N F NH 2 WO 2012/062704 70 PCT/EP2011/069553 HN N HNA-N -N 68 BEM D 12.76 273 N-N HN 69 H BEM D 12.76 273 N-N N 70 BCE D 11.58 365 SNH2 N-N N-N N HN N ' 71 o BCE D 11.95 373 N N-N N HN N 72 Oj 7' BEL D 11.78 373 S.. N- NH N-N N HN N 73 NH BEL D 12.28 387 S.., N-N NH N N N 74 O BEL D 12.35 387 \ N N-N H N 75 H'No BEL D 10.64 389 SN ,,OH N-N H WO 2012/062704 71 PCT/EP2011/069553 N 76 O BEL D 11.55 403 \S N N-N O H N HNAN ~ 77 Oj N BEFL D 10.18 402 S% N NH 2 N-N H 0 N HN N 78 0 BEL D 12.39 399 N-N O N N HN N 79 0 BEL D 12.00 415 SN N-N N N HN N 80 0 BEL D 10.00 428 N") N-N N HN HN N- a5; 0 81 N BEL D 11.55 456 N-N N N HN N 82 O BEL D 12.86 413 NN N-N H N HN N 83 O O BEL D 11.49 415 N-NO / - WO 2012/062704 72 PCT/EP2011/069553 N " HN N 84 H BCF D 11.36 367 N-N 0 N HN N 85 2H BCF D 7.23 352 N-N 0 N ' HN NN 86 N BCF D 5.58 392 N-N 0 N 0 87 NN BCF D 10.43 420 N-N 0 N HN N 88 CK D 12.45 331 S OMe N F N'" HN N 89 - OMe BCF D 12.54 399 N-N F NH N HN N 7 OMe 90 NF BCF D 12.23 397
NH
WO 2012/062704 73 PCT/EP2011/069553 N HNAN 91 O BEL E 13.13 388 SN
NH
2 N-N H N' HN N 92 OMe BCF D 11.43 357 N-N F
NH
2 0 N' 1 HN N 93 7 / OMe BCF D 11.81 371 N-N F NH O HN N 94 OMe BCF D 12.18 385 N-N F NH O \ N' HN N 95 7 / OMe BCF D 11.77 385 N-N F N O N HN JN 96 OMe BCF D 10.36 426 N-N F N NH 0 __ _ WO 2012/062704 74 PCT/EP2011/069553 N HN N I 97 OMe BCF D 12.71 425 N-N F N N HN N 98 / OMe BCF D 12.30 411 N-N F No N 99 HN N N NH BCIJ D 11.16 352 N-N H N HN N '-1;z 0 100 HN N N-NH BCIJ D 11.82 366 N-N H N HN N O 101 BCIJ D 11.88 366 NN N-N H H 102 N 1OH BCIJ D 9.65 368 H, N N-N HN O 103 BCIJ D 11.67 378 N 104 HN N CK D 13.77 316 OMe S F WO 2012/062704 75 PCT/EP2011/069553 N N HN N 105 H BCG D 11.61 385 N-N F 0 N HN N 106 / NH2 BCGI E 13.09 343 N-N F N HN N O 0 107 N NH BCIJ D 10.29 338 N-N H N HN N 0 108 .. BCIJ D 12.98 352 N-N N HN N O 109 BCIJ E 12.82 381 SN N NH2 H H" N-N 0 N HN N O 110 BCIJ D 10.98 394 N N N-N H O N HN N 111 BCE D 13.09 280 N-N N HN N 112 BCE D 13.12 280
N-N
WO 2012/062704 76 PCT/EP2011/069553 N N HN 4,N 113 BCE D 13.91 342 N-N N HN N 114 BCE D 11.38 362 7 /
SO
2 Me N-N F N HN N 115 BCG D 12.90 310 N-N N HN N 116 BCG D 10.55 326 N-N N HNJN 117 SN BCE D 13.43 330 r~l SMe N-N F N HN N 118 OH BCGK D 11.44 361 -- N F N HN N 119 H LK E 14.84 430 S 0 0 N HN N 120 N CFK E 14.12 354 0 WO 2012/062704 77 PCT/EP2011/069553 N 121 HN N CK D 10.40 361 N-N N HN N 123 BCE E 15.88 308 N-N N HN N 124 N-N F BCFG D 10.48 457 N HN N NN 125 N-N OBCFL D 12.20 526 N N 7N-': HN N 126 N-N 0 BCF E 13.99 450 N ____0 WO 2012/062704 78 PCT/EP2011/069553 N HN N 127 BCE D 12.19 321 0 N-N CN N' HN N 128 H BCEFL D 10.27 339 0 N-N \ H 2 N 0 N' HN N 129 / OMe BC D 12.24 392 N-N F N -' N HN N 130 H BCF E 14.62 355 N-N F 0 N HNN N . 131 N BCEI D 12.21 332 NH N-N N HN N 132 /OMe BCF E 13.65 440 N-N F N N HNN OMe 133 N FZe BCF E 13.69 454 N-N F 0
\-
WO 2012/062704 79 PCT/EP2011/069553 N HN '"N 134 OMe BCF C 0.87 440 N-N F NH oN HN 135 BCF D 12.88 407 N-N N/-\ N HN N 136 BCF D 13.26 421 N-N
N/
N HN N 137 BCF D 11.31 464 N-N 0 N/N N HN NH 138 BCF D 11.85 448 N-N 0 OH N 139 0 BCG D 13.73 356 N-N F WO 2012/062704 80 PCT/EP2011/069553 N HN N 140 | BCG D 13.89 356 O N-N F N HN N 141 HN BCG D 13.64 354 N-N F N HN N 142 BCG D 13.92 368 N-N F N -' HN N 143 H BCF D 12.71 365 zN N-N O N HN N 144 H BCF D 12.49 365 " I N N-N 0 N HN N 145 H BCF D 12.66 365 N-N 0 N HN N 146 H BCF D 10.77 367 .- N OH N-N 0 HN 147 BCF G 9.34 405 N-N
O$N
WO 2012/062704 81 PCT/EP2011/069553 N HN N 148 BCE G 9.46 419 N-N N HN N 149 BCF G 8.64 448 N-N 0 ONo N HN N 150 BCF G 8.79 462 N-N 0 ONo N HN N 151 BCFG D 12.48 441 N-N F N HN N 152 F 0 BCFG D 12.86 455 N-N F N HN NH 153 BCF G 8.44 434 N-N 0 O No WO 2012/062704 82 PCT/EP2011/069553 N HN N 154 | BCH F 7.23 358 N-N F OH N HN N 155 BCG D 13.29 340 N-N F N HN N 156 H BCF G 8.55 349 N-N 0 N HN N 157 H BCF D 12.40 363 N-N HN N 158 O BCFG D 13.10 455 N-N F 0 N \ N' 1 HNAN F 159 O F BCG C 1.09 396 N-N F HN 160 BCF G 8.03 365 N-N NH O \- WO 2012/062704 83 PCT/EP2011/069553 N HN N x NH 2 161 S BCF G 8.26 456 N-N 00 No N HN N x NH 2 162 S BCF G 7.69 458 N-N 00
N/-
N HN N 163 BCF F 8.37 408 N-N 0 NH N HN lilNH 164 BCF F 6.70 422 N-N 0 NH N HN N 165 (I NH2 BCF G 7.83 416 N-N 00 NH N HN N 0 166 N, BCFL G 8.51 486 N-N 0 0 N H o \- WO 2012/062704 84 PCT/EP2011/069553 N ~ HN NH 167 o S BCFL F 8.33 486 N-N K, NH N HN N 168 BCF G 8.22 379 N-N NH N HN N 169 BCF G 8.19 415 N-N &o NH N HN N 170 BCF G 8.53 455 N-N S5e No N HN N' O 171 BCFL G 8.43 528 N HN N 172 o BCFL G 8.24 528 N-N o/ -0 WO 2012/062704 85 PCT/EP2011/069553 N HN N 173 BCF F 8.13 457 N-N N/-\ N HN N 174 OBCFG F 7.71 399 N-N F NH N HN N 175 O BCFG F 7.94 413 N-N F NH N HN N 176 O BCFG G 8.08 413 N-N F NH N HN N 177 OH BCF G 8.27 371 N-N F NH HN N HN 0 178s BCFL G 8.73 526 N-N O No WO 2012/062704 86 PCT/EP2011/069553 N " HN Ii N I lz H 179 O N BCFL F 8.56 526 N-N o K' No N HN N 180 BCFG F 7.98 439 N-N F No N HN N 181 BCFG G 8.14 453 N-N F No N HN N 182 BCFG F 9.03 425 N-N F No N HN N 183 BCFG F 8.00 439 N-N F N 184 BCG F 7.97 372 N-N F O WO 2012/062704 87 PCT/EP2011/069553 N HN N 185 BCDG G 8.32 356 N-N F N HN N 186 BC G 8.58 358 N-N O-S N N N 187 HN N 0+0 BCEI G 8.39 373 N N-N N HN N 188 HN N BCEI G 8.62 387 N N-N N N N 189 HN N O\ O BCEI F 8.59 387 N N N HN N 190 I H BCF F 7.42 365 x N N-N 0 N HN N 191 H BCF G 7.41 365 N-N 0 N HN N 192 |N BCH F 8.75 358 N-N F OH WO 2012/062704 88 PCT/EP2011/069553 N HN N 193 BCH F 7.20 358 N-N F OH N HN N 194 BCG F 8.40 356 N-N F N HN N 195 BCG G 8.42 356 N-N F N HN N 196 OH BCF G 8.37 397 N-N F O No N HN N 197 OH BCF G 8.56 411 N-N F O No N 198 HN N BCG F 8.96 314 0 N-NH F N HN N 199 CN F 7.81 331 N F N HN N 200 I K BCG G 8.36 356 N-N F WO 2012/062704 89 PCT/EP2011/069553 N HN N 201 H BCFG G 8.60 381 lo N N-N 0 N HN N 202 0 BCDG F 8.80 427 0 N N-N F N HN N 203 BCDG F 7.73 372 HO N-N F N HN N 204 BCDG F 8.16 342 N-N F N HN N 205 BCDG G 8.35 356 N-N F N HN N 206 BCDG G 9.03 353 N-N F NC- N N HN N 207 BCDG G 9.19 372 / 7) ; 0 O N-N F N HN N 208 0 BCDG G 8.81 388 N-N F HO OH WO 2012/062704 90 PCT/EP2011/069553 N HN N 209 0 BCDG F 8.81 388 N-N F HO' OH N HN NF 210 BCE G 8.05 332 N-N F N HN N 211 x BCE F 8.27 374 N-N O=Sl / 0 N HN N 212 BCDG G 8.48 434 N-N F N HN N' 00 213 BCEI F 8.74 421 N HNHN N HN N HN N 214 / BCDG F 7.14 357
H
2 N N-N F N HN N 216 BCFG F 9.07 455 0 N-N F \- 4 N-N
F
WO 2012/062704 91 PCT/EP2011/069553 N HN N 217 0 BCDG G 7.91 386 N-N F HO N HN N 218 ONBCEI G 8.34 384 N-N NN 219 HN N BCEI F 8.09 389 N OH N-N H Biology Assays Determination of the effect of the compounds according to the invention on TYK2 The compounds of the present invention as described in the previous examples were tested in a Kinobeads T M assay as described for ZAP-70 (WO-A 2007/137867). Briefly, test compounds (at various concentrations) and the affinity matrix with the immobilized aminopyrido pyrimidine ligand 24 were added to cell lysate aliquots and allowed to bind to the proteins in the lysate sample. After the incubation time the beads with captured proteins were separated from the lysate. Bound proteins were then eluted and the presence of TYK2 and JAK2 was detected and quantified using specific antibodies in a dot blot procedure and the Odyssey infrared detection system. Dose response curves for individual kinases were generated and
IC
50 values calculated. KinobeadsTM assays have been previously described (WO-A 2007/137867; WO-A 2006/134056). Protocols Washing of affinity matrix The affinity matrix was washed two times with 15iL of 1x DP buffer containing 0.2% NP40 (IGEPAL@ CA-630, Sigma, #13021) and then resuspended in 1xDP buffer containing 0.2% WO 2012/062704 92 PCT/EP2011/069553 NP40 (3% beads slurry). 5xDP buffer: 250mM Tris-HCl pH 7.4, 25% Glycerol, 7.5mM MgCl 2 , 750mM NaCl, 5mM Na 3
VO
4 ; filter the 5xDP buffer through a 0.22pim filter and store in aliquots at -80'C. The 5xDP buffer is diluted with H 2 0 to 1xDP buffer containing 1mM DTT and 25mM NaF. Preparation of test compounds Stock solutions of test compounds were prepared in DMSO. In a 96 well plate 30pL solution of diluted test compounds at 5mM in DMSO were prepared. Starting with this solution a 1:3 dilution series (9 steps) was prepared. For control experiments (no test compound) a buffer containing 2% DMSO was used. Cell culture and preparation of cell lysates Molt4 cells (ATCC catalogue number CRL-1582) and Ramos cells (ATCC catalogue number CRL-1596) were grown in IL Spinner flasks (Integra Biosciences, #182101) in suspension in RPMI 1640 medium (Invitrogen, #21875-034) supplemented with 10% Fetal Bovine Serum (Invitrogen) at a density between 0.15 x 106 and 1.2 x 106 cells/mL. Cells were harvested by centrifugation, washed once with 1 x PBS buffer (Invitrogen, #14190-094) and cell pellets were frozen in liquid nitrogen and subsequently stored at -80'C. Cells were homogenized in a Potter S homogenizer in lysis buffer: 50mM Tris-HCl, 0.8% NP40, 5% glycerol, 150mM NaCl, 1.5mM MgCl 2 , 25 mM NaF, 1mM sodium vanadate, 1mM DTT, pH 7.5. One complete EDTA-free tablet (protease inhibitor cocktail, Roche Diagnostics, 1873580) per 25mL buffer was added. The material was dounced 10 times using a mechanized POTTER S, transferred to 50mL falcon tubes, incubated for 30 minutes on ice and spun down for 10 minutes at 20,000 g at 4'C (10,000 rpm in Sorvall SLA600, precooled). The supernatant was transferred to an ultracentrifuge (UZ)-polycarbonate tube (Beckmann, 355654) and spun for Ihour at 100.000g at 4'C (33.500 rpm in Ti50.2, precooled). The supernatant was transferred again to a fresh 50mL falcon tube, the protein concentration was determined by a Bradford assay (BioRad) and samples containing 50mg of protein per aliquot were prepared. The samples were immediately used for experiments or frozen in liquid nitrogen and stored frozen at -80'C. Dilution of cell lysate Cell lysate (approximately 50mg protein per plate) was thawed in a water bath at room temperature and then stored on ice. To the thawed cell lysate 1xDP 0.8% NP40 buffer containing protease inhibitors (1 tablet for 25mL buffer; EDTA-free protease inhibitor WO 2012/062704 93 PCT/EP2011/069553 cocktail; Roche Diagnostics 1873580) was added in order to reach a final protein concentration of 10mg/mL total protein. The diluted cell lysate was stored on ice. Mixed Molt4/Ramos lysate was prepared by combining one volume of Molt4 lysate and two volumes of Ramos lysate (ratio 1:2). Incubation of lysate with test compound and affinity matrix To a 96 well filter plate (Multiscreen HTS, BV Filter Plates, Millipore #MSBVN1250) were added per well: 100[tL affinity matrix (3 % beads slurry), 3 [L of compound solution, and 50[tL of diluted lysate. Plates were sealed and incubated for 3 hours in a cold room on a plate shaker (Heidolph tiramax 1000) at 750rpm. Afterwards the plate was washed 3 times with 230[tL washing buffer (1xDP 0.4% NP40). The filter plate was placed on top of a collection plate (Greiner bio-one, PP-microplate 96 well V-shape, 65120) and the beads were then eluted with 20[tL of sample buffer (100 mM Tris, pH 7.4, 4% SDS, 0.00025% bromophenol blue, 20% glycerol, 50 mM DTT). The eluate was frozen quickly at -80'C and stored at -20'C. Detection and quantification of eluted kinases The kinases in the eluates were detected and quantified by spotting on nitrocellulose membranes and using a first antibody directed against the kinase of interest and a fluorescently labelled secondary antibody (anti-rabbit IRDye TM antibody 800 (Licor, # 926-32211). The Odyssey Infrared Imaging system from LI-COR Biosciences (Lincoln, Nebraska, USA) was operated according to instructions provided by the manufacturer (Schutz-Geschwendener et al., 2004. Quantitative, two-color Western blot detection with infrared fluorescence. Published May 2004 by LI-COR Biosciences, www.licor.com). After spotting of the eluates the nitrocellulose membrane (BioTrace NT; PALL, #BTNT30R) was first blocked by incubation with Odyssey blocking buffer (LICOR, 927-40000) for 1 hour at room temperature. Blocked membranes were then incubated for 16 hours at the temperature shown in table 8 with the first antibody diluted in Odyssey blocking buffer (LICOR #927 40000). Afterwards the membrane was washed twice for 10 minutes with PBS buffer containing 0.2% Tween 20 at room temperature. The membrane was then incubated for 60 minutes at room temperature with the detection antibody (anti-rabbit IRDye TM antibody 800, Licor, # 926-32211) diluted in Odyssey blocking buffer (LICOR #927-40000). Afterwards the membrane was washed twice for 10 minutes each with 1 x PBS buffer containing 0.2% Tween 20 at room temperature. Then the membrane was rinsed once with PBS buffer to remove WO 2012/062704 94 PCT/EP2011/069553 residual Tween 20. The membrane was kept in PBS buffer at 4'C and then scanned with the Odyssey instrument. Fluorescence signals were recorded and analysed according to the instructions of the manufacturer. Table 8: Sources and dilutions of antibodies Target kinase Primary antibody Temperature Secondary antibody (dilution) of primary (dilution) incubation Jak2 Cell signaling #3230 Room Licor anti-rabbit 800 (1:100) temperature (1:15000) TYK2 Upstate #06-638 Room Licor anti-rabbit 800 (1:1000) temperature (1:5000) Results Table 9: Inhibition values (IC 50 in p.M) as determined in the KinobeadTM assay Activity level: A < 0.1piM; 0.1pM < B < 1pM; 1pM < C < 10piM; D >10pM). Example No. TYK2 IC 50 (pM) JAK2 IC 50 (pM) 1 B D 2 B D 3 B D 4 B D 5 B D 6 B D 7 B D 8 A D 9 A C 10 A C 11 A B 12 A B 13 A B WO 2012/062704 95 PCT/EP2011/069553 14 A B 15 A B 16 A C 17 B C 18 A B 19 B C Furthermore, the following Examples inhibited TYK2 with activity level A and JAK2 with activity level B: 36, 37, 49, 50, 51, 53, 54, 58, 60, 61, 71, 72, 75, 76, 77, 79, 83, 84, 88, 89, 91, 93, 94, 95, 96, 106, 114, 116, 117, 118, 120, 124, 137, 144, 146, 152, 154, 158, 159, 166, 5 171, 175, 176, 178, 183, 184, 185, 187, 191, 192, 193, 195, 198, 202, 203, 204, 207, 208, 209, 216, 218, 219. Furthermore, the following Examples inhibited TYK2 with activity level A and JAK2 with activity level C: 29, 35, 56, 59, 62, 64, 67, 85, 90, 92, 97, 98, 111, 115, 121, 122, 123, 125, 10 126, 129, 132, 133, 134, 135, 136, 138, 147, 148, 149, 150, 151, 153, 160, 161, 162, 163, 164, 167, 168, 172, 174, 179, 180, 181, 182, 188, 189, 190, 196, 206, 214, 215. All other examples inhibited TYK2 with activity level B and typically inhibited JAK2 with activity level C or D. 15

Claims (21)

  1. 2. The compound of claim 1, wherein X 1 is N and X 2 is C(R 15 ).
  2. 3. The compound of claim 1 or 2, R 1 is T'; or C(O)R 4 . 15 4. The compound of any one of claims I to 3, wherein R 4 is T 1 .
  3. 5. The compound of any one of claims 1 to 4, wherein T' is cyclopropyl; thiophenyl; isothiazolyl; or pyrazolyl and wherein Ti is optionally substituted with one or more R7 which are the same or different. 20 WO 2012/062704 101 PCT/EP2011/069553
  4. 6. The compound of any one of claims I to 5, wherein R 1 is C(O)T 1 , T' is C 3 .7 cycloalkyl and wherein T' is optionally substituted with one or more R 7 , which are the same or different. 5 7. The compound of any one of claims 1 to 5, wherein R 1 is T', T' is 5 membered aromatic heterocyclyl and wherein T' is optionally substituted with one or more R 7 , which are the same or different.
  5. 8. The compound of claim 7, wherein T' is isothiazolyl; or pyrazolyl and wherein T' is 10 optionally substituted with one or more R 7 , which are the same or different.
  6. 9. The compound of claim 8, wherein T' is R 7 R 7 N N or wherein R 7 is defined as indicated in claim 1. 15
  7. 10. The compound of any one of claims 1 to 9, wherein T' is cyclopropyl; or pyrazolyl and wherein T is unsubstituted or substituted with one or more R 7 , which are the same or different. 20 11. The compound of any one of claims I to 10, wherein R 7 is methyl; CH 2 CH 2 ORSC; CH 2 C(O)T 2 ; or CH 2 C(O)N(RCR8d).
  8. 12. The compound of any one of claims I to 11, wherein R 15 is CH 3 . 25 13. The compound of any one of claims I to 12, wherein R 3 is H.
  9. 14. The compound of any one of claims I to 13, wherein R 2 is phenyl; pyridyl; pyrimidyl; pyrazolyl; oxadiazolyl; thiazolyl; or thiadiazolyl and wherein R2 is substituted with one or more R 18 , which are the same or different. 30
  10. 15. The compound of claim 14, wherein R2 is phenyl and wherein R2 is substituted with one or more R 18 , which are the same or different. WO 2012/062704 102 PCT/EP2011/069553
  11. 16. The compound of any one of claims 1 to 15, wherein R 8 is halogen; CN; S(O) 2 N(R1 9 R1 9 a); S(O) 2 R"; NHS(O) 2 R1 9 a; OR"; C(O)N(R1 9 aR 9 ); SR 19 ; Ci4 alkyl, unsubstituted or substituted with one or more halogen, which are the same or different; 5 NHC(O)R 19 ; or C(O)R 19 .
  12. 17. The compound of any one of claims 1 to 16, wherein R 18 is halogen; OCH 3 ; S(O) 2 CH 3 ; NHS(O) 2 CH 3 ; or CH 2 NHS(0) 2 CH 3 . 10 18. The compound of any one of claims I to 17, wherein R2 is F R 18 or R 18 wherein R 18 is defined as indicated in any one of claims I to 17.
  13. 19. The compound of any one of claims I to 18 selected from the group consisting of 15 N-(4-(3-fluorophenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; 4-(2-fluoro-4-methoxyphenyl)-5-methyl-N-(1-methyl-iH-pyrazol-4-yl)pyrimidin-2 amine; N-(4-(2-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; 20 N-(4-(4-chlorophenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; N-(5-methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)cyclopropanecarboxamide; N-(5-methyl-4-(p-tolyl)pyrimidin-2-yl)cyclopropanecarboxamide; N-(4-(4-methoxyphenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; N-(4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)cyclopropanecarboxamide; 25 4-(3-fluoro-4-methoxyphenyl)-5-methyl-N-(i-methyl-iH-pyrazol-4-yl)pyrimidin-2 amine; 5-methyl-N-(i-methyl-iH-pyrazol-4-yl)-4-(4-(methylsulfonyl)phenyl)pyrimidin-2 amine; 4-(3-fluoro-4-methoxyphenyl)-5-methyl-N-(1H-pyrazol-4-yl)pyrimidin-2-amine; 30 5-methyl-4-(4-(methylsulfonyl)phenyl)-N-(1H-pyrazol-4-yl)pyrimidin-2-amine; WO 2012/062704 103 PCT/EP2011/069553 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)-i -morpholinoethanone; 2-(4-((5 -methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)ethanol; 5 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)ethanol; 2-(4-((5 -methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)-1- morpholinoethanone; N-(3 -(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzyl)methane 10 sulfonamide; N-methyl-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; N-methyl-3 -(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 15 4-(2-fluoro-4-(methylsulfonyl)phenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)methanesulfonamide; 4-(3 -fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 20 4-(4-fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(2-methoxyphenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(4-methoxyphenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 3 -(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; 5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(4-(trifluoromethyl)phenyl)pyrimidin-2 25 amine; 5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(3 -(methylthio)phenyl)pyrimidin-2-amine; 5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(4-(methylthio)phenyl)pyrimidin-2-amine; 5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-phenylpyrimidin-2-amine; 4-(3,4-difluorophenyl)-5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 30 4-(3,5-difluorophenyl)-5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzonitrile; 4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; 4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; WO 2012/062704 104 PCT/EP2011/069553 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(3 -(methylsulfonyl)phenyl)pyrimidin-2 amine; 4-(4-(ethylsulfonyl)phenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 5 4-(4-(isopropylsulfonyl)phenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 4-(3,4-dimethoxyphenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(3 -chloro-4-methoxyphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 10 4-(4-chloro-3 -fluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 4-(4-methoxy-3 -(trifluoromethyl)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; 4-(3 -fluoro-4-methylphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 15 amine; N-(2-fluoro-4-(5-methyl-2-((1 -methyl- IH-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)acetamide; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(pyridin-4-yl)pyrimidin-2-amine; 2'-methoxy-5-methyl-N-(1 -methyl- IH-pyrazol-4-yl)-[4,5'-bipyrimidin]-2-amine; 20 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(1H-pyrazol-4-yl)pyrimidin-2-amine; 5-methyl-4-(4-(5-methyl- 1,3,4-oxadiazol-2-yl)phenyl)-N-(1 -methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; 4-(4-ethoxy-3 -fluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 25 4-(3 -fluoro-4-propoxyphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 4-(3 -fluoro-4-isopropoxyphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 4-(4-(cyclohexyloxy)-3 -fluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4 30 yl)pyrimidin-2-amine; 2-(2-fluoro-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenoxy)ethanol; 4-(3 -fluoro-4-(2-methoxyethoxy)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; WO 2012/062704 105 PCT/EP2011/069553 2-(2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenoxy)- 1 -(pyrrolidin- 1 -yl)ethanone; N-methyl-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 5 N,N-dimethyl-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; N-ethyl-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; N-isopropyl-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 10 4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N propylbenzamide; N-(2-hydroxyethyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; N-(2-amino-2-oxoethyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin 15 4-yl)benzamide; (4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)(pyrrolidin 1 -yl)methanone; (4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)(morpholino)methanone; 20 N-cyclopentyl-4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N-(tetrahydrofuran 3-yl)benzamide; 2-fluoro-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)benzamide; 25 5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(thiazol-2-yl)pyrimidin-2-amine; 5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(thiazol-5 -yl)pyrimidin-2-amine; 5 -(4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)- 1,3,4 thiadiazol-2-amine; N,N-dimethyl-4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 30 yl)benzenesulfonamide; N-ethyl-4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; N-isopropyl-4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; WO 2012/062704 106 PCT/EP2011/069553 4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N propylbenzenesulfonamide; N-(2-hydroxyethyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 5 N-(2-methoxyethyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 2-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenylsulfonamido)acetamide; 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(4-(pyrrolidin- 1 10 ylsulfonyl)phenyl)pyrimidin-2-amine; 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(4-(morpholinosulfonyl)phenyl)pyrimidin 2-amine; 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(4-((4-methylpiperazin- 1 yl)sulfonyl)phenyl)pyrimidin-2-amine; 15 1-(4-((4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)sulfonyl)piperazin- 1 -yl)ethanone; N-cyclopentyl-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)-N-(tetrahydrofuran 20 3 -yl)benzenesulfonamide; N-(2-methoxyethyl)-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; N-(2-aminoethyl)-4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 25 (4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)(4 methylpiperazin- 1 -yl)methanone; 1-(4-(4-(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzoyl)piperazin- 1 -yl)ethanone; N-(4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)-3 -methylisothiazol-5 30 amine; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)-N-isopropylacetamide; N-cyclopropyl-2-(4-((4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)amino) IH-pyrazol- 1 -yl)acetamide; WO 2012/062704 107 PCT/EP2011/069553 N-(2-aminoethyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)acetamide; 5 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)-N-methylacetamide; N-ethyl-2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1H pyrazol- 1 -yl)acetamide; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 10 yl)-N,N-dimethylacetamide; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)-1 -(piperazin- 1 -yl)ethanone; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)-1 -(piperidin- 1 -yl)ethanone; 15 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)-1 -(pyrrolidin- 1 -yl)ethanone; 1 -ethyl-3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)urea; 1 -isopropyl-3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 20 yl)phenyl)urea; 1-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)-3 propylurea; 1-(2-hydroxyethyl)-3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)urea; 25 N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)pyrrolidine- 1 -carboxamide; 4-(3-fluoro-4-methoxyphenyl)-5-methyl-N-(thiophen-3-yl)pyrimidin-2-amine; N-(2-(2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenoxy)ethyl)acetamide; 30 4-(4-(2-aminoethoxy)-3 -fluorophenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; 1 -methyl-3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)urea; WO 2012/062704 108 PCT/EP2011/069553 1,1 -dimethyl-3-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)urea; 2-(3 -(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)ureido)acetamide; 5 N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)morpholine-4-carboxamide; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(p-tolyl)pyrimidin-2-amine; 5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)-4-(m-tolyl)pyrimidin-2-amine; 4-([1,1 '-biphenyl]-4-yl)-5-methyl-N-(1 -methyl- IH-pyrazol-4-yl)pyrimidin-2-amine; 10 4-(3 -fluoro-4-(methylsulfonyl)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; 4-(4-ethoxyphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 2-(4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenoxy)ethanol; 4-(3 -fluoro-4-(methylthio)phenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin 15 2-amine; 2-(2-fluoro-4-(5 -methyl-2-((3 -methylisothiazol-5 -yl)amino)pyrimidin-4 yl)phenoxy)ethanol; N-cyclopentyl-4-(5-methyl-2-((3-methylisothiazol-5-yl)amino)pyrimidin-4 yl)benzenesulfonamide; 20 N-ethyl-4-(5-methyl-2-((3-methylisothiazol-5-yl)amino)pyrimidin-4-yl)benzamide; 3-methyl-N-(5-methyl-4-(4-(methylsulfonyl)phenyl)pyrimidin-2-yl)isothiazol-5 amine; 4-(4-ethylphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 4-(4-isopropylphenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin-2-amine; 25 2-(4-((4-(3-fluoro-4-(2-hydroxyethoxy)phenyl)-5-methylpyrimidin-2-yl)amino)- iH pyrazol- 1 -yl)- 1 -morpholinoethanone; N-cyclopentyl-4-(5 -methyl-2-((1-(2-morpholino-2-oxoethyl)- 1 H-pyrazol-4 yl)amino)pyrimidin-4-yl)benzenesulfonamide; N-ethyl-4-(5 -methyl-2-((1-(2-morpholino-2-oxoethyl)- 1 H-pyrazol-4 30 yl)amino)pyrimidin-4-yl)benzamide; 2-methoxy-5 -(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzonitrile; 2-methoxy-5 -(5 -methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; WO 2012/062704 109 PCT/EP2011/069553 4-(3 -fluoro-4-methoxyphenyl)-5-methyl-N-(1 -((methylsulfonyl)methyl)- 1H-pyrazol 4-yl)pyrimidin-2-amine; N-ethyl-2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 5 4-(4-(1H-pyrazol-3 -yl)phenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)- 1 -(4-methylpiperazin- 1 -yl)ethanone; 1-(3,5-dimethylpiperazin- 1 -yl)-2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 10 methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)ethanone; 1-(1,4-diazepan- 1 -yl)-2-(4-((4-(3 -fluoro-4-methoxyphenyl)-5 -methylpyrimidin-2 yl)amino)- 1H-pyrazol- 1 -yl)ethanone; 2-(4-((4-(4-ethylphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)- 1 morpholinoethanone; 15 2-(4-((4-(4-i sopropylphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 -yl)- 1 morpholinoethanone; N-isopropyl-4-(5 -methyl-2-((1-(2-morpholino-2-oxoethyl)- 1 H-pyrazol-4 yl)amino)pyrimidin-4-yl)benzamide; N-isopropyl-4-(5 -methyl-2-((1-(2-oxo-2-(pyrrolidin- 1 -yl)ethyl)- 1 H-pyrazol-4 20 yl)amino)pyrimidin-4-yl)benzamide; 4-(4-(sec-butoxy)-3 -fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin 2-amine; 4-(3 -fluoro-4-i sobutoxyphenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin-2 amine; 25 4-(4-cyclobutoxy-3 -fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin 2-amine; 4-(4-(cyclopentyloxy)-3 -fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; N-(tert-butyl)-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 30 yl)benzamide; N-(sec-butyl)-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; N-isobutyl-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; WO 2012/062704 110 PCT/EP2011/069553 N-(2-hydroxypropyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 2-(4-((4-(4-ethylphenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1-yl)-1 (piperidin- 1 -yl)ethanone; 5 2-(4-((4-(4-i sopropylphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl)-1 (piperidin- 1 -yl)ethanone; N-ethyl-4-(5 -methyl-2-((1-(2-oxo-2-(piperidin- 1 -yl)ethyl)- 1 H-pyrazol-4 yl)amino)pyrimidin-4-yl)benzamide; N-isopropyl-4-(5 -methyl-2-((1-(2-oxo-2-(piperidin- 1 -yl)ethyl)- 1 H-pyrazol-4 10 yl)amino)pyrimidin-4-yl)benzamide; 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl) 1 -morpholinoethanone; 2-(4-((4-(3 -fluoro-4-isopropoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol 1-yl)-1 -morpholinoethanone; 15 N-ethyl-4-(5 -methyl-2-((1-(2-oxo-2-(pyrrolidin- 1 -yl)ethyl)- 1 H-pyrazol-4 yl)amino)pyrimidin-4-yl)benzamide; 1-(2-fluoro-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenoxy)propan-2-ol; 4-(4-cyclopropoxy-3 -fluorophenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4 20 yl)pyrimidin-2-amine; N-cyclopropyl-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; N-cyclobutyl-4-(5 -methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 25 2-(4-((4-(3 -fluoro-4-propoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)- 1 -morpholinoethanone; 4-(3 -fluoro-4-(3,3,3 -trifluoropropoxy)phenyl)-5 -methyl-N-(1-methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; N-ethyl-2-(4-((4-(4-ethylphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 30 yl)acetamide; 4-(5 -methyl-2-((1-(2-oxo-2-(piperidin- 1 -yl)ethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin 4-yl)benzenesulfonamide; 4-(5 -methyl-2-((1-(2-morpholino-2-oxoethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzenesulfonamide; WO 2012/062704 111 PCT/EP2011/069553 N-ethyl-4-(2-((1-(2-(ethylamino)-2-oxoethyl)- 1H-pyrazol-4-yl)amino)-5 methylpyrimidin-4-yl)benzamide; 4-(2-((1-(2-(ethylamino)-2-oxoethyl)- 1H-pyrazol-4-yl)amino)-5-methylpyrimidin-4 yl)-N-isopropylbenzamide; 5 N-ethyl-2-(4-((5 -methyl-4-(4-sulfamoylphenyl)pyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)acetamide; N-ethyl-2-(4-((5 -methyl-4-(4-(morpholinosulfonyl)phenyl)pyrimidin-2-yl)amino)- 1H pyrazol- 1 -yl)acetamide; N-ethyl-2-(4-((5 -methyl-4-(4-(N-(tetrahydrofuran-3 -yl) sulfamoyl)phenyl)pyrimidin-2 10 yl)amino)- 1H-pyrazol- 1 -yl)acetamide; N-ethyl-2-(4-((4-(4-isopropylphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)acetamide; N-ethyl-2-(4-((5-methyl-4-(3 -(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1H pyrazol- 1 -yl)acetamide; 15 2-(4-((5-methyl-4-(3 -(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)-1 -(piperidin- 1 -yl)ethanone; 2-(4-((5-methyl-4-(4-(morpholinosulfonyl)phenyl)pyrimidin-2-yl)amino)- 1H-pyrazol 1-yl)-1 -morpholinoethanone; 4-(5 -methyl-2-((1-(2-morpholino-2-oxoethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin-4-yl) 20 N-(tetrahydrofuran-3 -yl)benzenesulfonamide; 2-(4-((5 -methyl-4-(3 -(methylsulfonyl)phenyl)pyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)-1 -morpholinoethanone; 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl) N-ethylacetamide; 25 N-ethyl-2-(4-((4-(3 -fluoro-4-isopropoxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H pyrazol- 1 -yl)acetamide; N-ethyl-2-(4-((4-(3 -fluoro-4-propoxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1H pyrazol- 1 -yl)acetamide; N-ethyl-2-(4-((4-(3 -fluoro-4-hydroxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1H 30 pyrazol- 1 -yl)acetamide; 2-(4-((5-methyl-4-(4-(morpholinosulfonyl)phenyl)pyrimidin-2-yl)amino)- 1H-pyrazol 1-yl)-1 -(piperidin- 1 -yl)ethanone; 4-(5 -methyl-2-((1-(2-oxo-2-(piperidin- 1 -yl)ethyl)- 1 H-pyrazol-4-yl)amino)pyrimidin 4-yl)-N-(tetrahydrofuran-3 -yl)benzenesulfonamide; WO 2012/062704 112 PCT/EP2011/069553 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1-yl) 1 -(piperidin- 1 -yl)ethanone; 2-(4-((4-(3-fluoro-4-isopropoxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol 1-yl)-1 -(piperidin- 1 -yl)ethanone; 5 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1-yl) 1 -(pyrrolidin- 1 -yl)ethanone; 2-(4-((4-(3-fluoro-4-isopropoxyphenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol 1-yl)-1 -(pyrrolidin- 1 -yl)ethanone; 4-(3 -fluoro-4-(2-methoxypropoxy)phenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4 10 yl)pyrimidin-2-amine; 4-(4-ethoxy-3 -fluorophenyl)-N-(1 -isopropyl- 1H-pyrazol-4-yl)-5-methylpyrimidin-2 amine; 5-methyl-N-(1-methyl-i H-pyrazol-4-yl)-4-(4-methyl-3 (methylsulfonyl)phenyl)pyrimidin-2-amine; 15 N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)ethanesulfonamide; N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)propane 1-sulfonamide; N-(4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4-yl)phenyl)propane 20 2-sulfonamide; (R)-N-(sec-butyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; (S)-N-(sec-butyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 25 (R)- 1 -(2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenoxy)propan-2-ol; (S)- 1-(2-fluoro-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenoxy)propan-2-ol; (S)-4-(4-(sec-butoxy)-3 -fluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4 30 yl)pyrimidin-2-amine; (R)-4-(4-(sec-butoxy)-3 -fluorophenyl)-5-methyl-N-(1 -methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; 2-(4-((4-(3 -fluoro-4-hydroxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)- 1 -(pyrrolidin- 1 -yl)ethanone; WO 2012/062704 113 PCT/EP2011/069553 2-(4-((4-(3 -fluoro-4-hydroxyphenyl)-5 -methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)-1 -(piperidin- 1 -yl)ethanone; 4-(4-ethoxy-3-fluorophenyl)-5-methyl-N-(1H-pyrazol-4-yl)pyrimidin-2-amine; N-(4-(3-fluoro-4-methoxyphenyl)-5-methylpyrimidin-2-yl)-2-methylthiazol-5-amine; 5 4-(4-(tert-butoxy)-3 -fluorophenyl)-5-methyl-N-(1-methyl-i H-pyrazol-4-yl)pyrimidin 2-amine; N-(2-methoxypropyl)-4-(5-methyl-2-((1-methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)benzamide; 4-(4-ethoxy-3 -fluorophenyl)-5-methyl-N-(1 -(2-morpholinoethyl)- 1H-pyrazol-4 10 yl)pyrimidin-2-amine; 3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1 yl)propan- 1 -ol; 4-(4-ethoxy-3 -fluorophenyl)-N-(1-ethyl-i H-pyrazol-4-yl)-5 -methylpyrimidin-2 amine; 15 4-(4-ethoxy-3 -fluorophenyl)-5 -methyl-N-(1 -propyl- 1 H-pyrazol-4-yl)pyrimidin-2 amine; 2-(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)acetonitrile; 4-(4-ethoxy-3 -fluorophenyl)-N-(1 -(2-methoxyethyl)- 1 H-pyrazol-4-yl)-5 20 methylpyrimidin-2-amine; (S)-3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)propane-1,2-diol; (R)-3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1 yl)propane- 1,2-diol; 25 4-(3,5-difluoro-4-methoxyphenyl)-5 -methyl-N-(1 -methyl-i H-pyrazol-4-yl)pyrimidin 2-amine; 4-(4-methoxy-3 -(methylsulfonyl)phenyl)-5 -methyl-N-(1 -methyl-i H-pyrazol-4 yl)pyrimidin-2-amine; 4-(4-ethoxy-3 -fluorophenyl)-N-(1 -(4-methoxybenzyl)- 1 H-pyrazol-4-yl)-5 30 methylpyrimidin-2-amine; N-(4-(5-methyl-2-((1 -methyl-i H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)benzenesulfonamide; N-(1 -(2-aminoethyl)- 1 H-pyrazol-4-yl)-4-(4-ethoxy-3 -fluorophenyl)-5 methylpyrimidin-2-amine; WO 2012/062704 114 PCT/EP2011/069553 3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1-yl) N-ethylpropanamide; 3 -(4-((4-(4-ethoxy-3 -fluorophenyl)-5-methylpyrimidin-2-yl)amino)- 1H-pyrazol- 1-yl) 1 -morpholinopropan- 1-one; 5 1-(4-((4-(4-ethoxy-3 -fluorophenyl)-5 -methylpyrimidin-2-yl)amino)- 1 H-pyrazol- 1-yl) 2-methylpropan-2-ol; 1 -cyano-N-(4-(5 -methyl-2-((1-methyl- 1H-pyrazol-4-yl)amino)pyrimidin-4 yl)phenyl)methanesulfonamide; and 2-hydroxy-N-(4-(5-methyl-2-((1-methyl-iH-pyrazol-4-yl)amino)pyrimidin-4 10 yl)phenyl)ethanesulfonamide.
  14. 20. A compound of any one of claims 1 to 19 or a pharmaceutically acceptable salt thereof 15 21. A compound or a pharmaceutically acceptable salt thereof of any one of claims I to 20 for use as a medicament.
  15. 22. A compound or a pharmaceutically acceptable salt thereof of any one of claims 1 to 20 for use in a method for treating or preventing diseases or disorders associated with 20 TYK2.
  16. 23. A compound or a pharmaceutically acceptable salt thereof of any one of claims 1 to 20 for use in a method for treating or preventing an immunological, inflammatory, autoimmune, or allergic disorder or disease of a transplant rejection or a Graft-versus 25 host disease.
  17. 24. A compound or a pharmaceutically acceptable salt thereof of any one of claims 1 to 20 for use in a method of treating or preventing a proliferative disease. 30 25. A compound of claim 24, wherein the disease is cancer.
  18. 26. A pharmaceutical composition comprising a compound or a pharmaceutically acceptable salt thereof of any of claims 1 to 20 together with a pharmaceutically WO 2012/062704 115 PCT/EP2011/069553 acceptable carrier, optionally in combination with one or more other pharmaceutical compositions.
  19. 27. Method for treating, controlling, delaying or preventing in a mammalian patient in 5 need thereof one or more conditions selected from the group consisting of diseases and disorders associated with TYK2, wherein the method comprises the administration to said patient a therapeutically effective amount of a compound of any one of claims 1 to 20 or a pharmaceutically acceptable salt thereof 10 28. Method for treating, controlling, delaying or preventing in a mammalian patient in need thereof one or more conditions selected from the group consisting of an immunological, inflammatory, autoimmune, or allergic disorder or disease or a transplant rejection or a Graft-versus host disease, wherein the method comprises the administration to said patient a therapeutically effective amount of a compound of any 15 one of claims 1 to 20 or a pharmaceutically acceptable salt thereof
  20. 29. Method for treating, controlling, delaying or preventing in a mammalian patient in need thereof a proliferative disease, wherein the method comprises the administration to said patient a therapeutically effective amount of a compound of any one of claims 20 1 to 20 or a pharmaceutically acceptable salt thereof.
  21. 30. The method of claim 29, wherein the proliferative disease is cancer.
AU2011328237A 2010-11-09 2011-11-07 Pyridine compounds and aza analogues thereof as TYK2 inhibitors Abandoned AU2011328237A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP10190460.5 2010-11-09
EP10190460 2010-11-09
PCT/EP2011/069553 WO2012062704A1 (en) 2010-11-09 2011-11-07 Pyridine compounds and aza analogues thereof as tyk2 inhibitors

Publications (1)

Publication Number Publication Date
AU2011328237A1 true AU2011328237A1 (en) 2013-05-23

Family

ID=43587413

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2011328237A Abandoned AU2011328237A1 (en) 2010-11-09 2011-11-07 Pyridine compounds and aza analogues thereof as TYK2 inhibitors

Country Status (6)

Country Link
EP (1) EP2638018A1 (en)
JP (1) JP2014500254A (en)
CN (1) CN103298794A (en)
AU (1) AU2011328237A1 (en)
CA (1) CA2815330A1 (en)
WO (1) WO2012062704A1 (en)

Families Citing this family (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012142329A1 (en) * 2011-04-12 2012-10-18 Myrexis, Inc. Compositions and therapeutic uses of ikk-related kinase epsilon and tankbinding kinase 1 inhibitors
US9266838B2 (en) 2011-08-15 2016-02-23 University Of Utah Research Foundation Substituted (E)-N′-(1-phenylethylidene)benzohydrazide analogs as histone demethylase inhibitors
DE102011112978A1 (en) 2011-09-09 2013-03-14 Merck Patent Gmbh benzonitrile derivatives
CN104169272A (en) 2011-12-23 2014-11-26 赛尔佐姆有限公司 Pyrimidine-2,4-diamine derivatives as kinase inhibitors
RU2641895C2 (en) * 2012-05-24 2018-01-23 Целльзом Лимитид Heterocyclic pyrimidine analogues as tyk2 inhibitors
KR102233252B1 (en) * 2012-11-08 2021-03-26 브리스톨-마이어스 스큅 컴퍼니 ALKYL-AMIDE-SUBSTITUTED PYRIDYL COMPOUNDS USEFUL AS MODULATORS OF IL-12, IL-23 AND/OR IFNα RESPONSES
EP2981252A4 (en) * 2013-04-04 2017-02-22 Olivia Newton-John Cancer Research Institute Methods of treating diseases characterized by excessive wnt signalling
ES2739814T3 (en) * 2013-06-19 2020-02-04 Univ Utah Res Found Compounds of (3- (5-Chloro-2-hydroxyphenyl) -1-benzoyl-1H-pyrazole substituted as histone demethylase inhibitors
RS57662B1 (en) 2013-12-11 2018-11-30 Biogen Ma Inc Biaryl compounds useful for the treatment of human diseases in oncology, neurology and immunology
JP6879740B2 (en) 2013-12-13 2021-06-02 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド How to Treat Lymphatic Plasma Cell Lymphoma
JP6878004B2 (en) 2013-12-13 2021-05-26 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド How to Treat Lymphatic Plasma Cell Lymphoma
US10112957B2 (en) 2014-10-22 2018-10-30 Dana-Farber Cancer Institute, Inc. Thiazolyl-containing compounds for treating proliferative diseases
CA2995430C (en) * 2015-09-25 2023-07-11 Dizal (Jiangsu) Pharmaceutical Co., Limited Compounds and methods for inhibiting jak
CN106928216A (en) * 2015-12-31 2017-07-07 中国科学院上海药物研究所 Compound, Preparation Method And The Use with ERK kinase inhibiting activities
US20190086392A1 (en) 2016-03-21 2019-03-21 Inserm (Institut National De La Sante Et De La Recherch Medicale) Methods for diagnosis and treatment of solar lentigo
TWI825663B (en) * 2016-10-14 2023-12-11 美商林伯士拉克許米公司 Tyk2 inhibitors and uses thereof
CN106588885B (en) * 2016-11-10 2019-03-19 浙江大学 2- replaces aromatic ring-pyridine derivatives and preparation and application
JP7249950B2 (en) 2017-03-27 2023-03-31 カーデュリオン・ファーマシューティカルズ・リミテッド・ライアビリティ・カンパニー Heterocyclic compound
GB201705263D0 (en) * 2017-03-31 2017-05-17 Probiodrug Ag Novel inhibitors
CN113365632B (en) 2018-09-25 2024-03-08 卡都瑞恩医药有限公司 Aminopyrimidine compounds
US11066404B2 (en) 2018-10-11 2021-07-20 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
WO2020092015A1 (en) 2018-11-02 2020-05-07 University Of Rochester Therapeutic mitigation of epithelial infection
WO2020118683A1 (en) * 2018-12-14 2020-06-18 Lynk Pharmaceuticals Co. Ltd. Benzamides of pyrazolyl-amino-pyrimidinyl derivatives, and compositions and methods thereof
WO2020168197A1 (en) 2019-02-15 2020-08-20 Incyte Corporation Pyrrolo[2,3-d]pyrimidinone compounds as cdk2 inhibitors
WO2020180959A1 (en) 2019-03-05 2020-09-10 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as cdk2 inhibitors
US11919904B2 (en) 2019-03-29 2024-03-05 Incyte Corporation Sulfonylamide compounds as CDK2 inhibitors
WO2020206588A1 (en) * 2019-04-08 2020-10-15 Lynk Pharmaceuticals Co., Ltd. Benzethers and anilines of pyrazolyl-amino-pyrimidinyl derivatives, and compositions and methods thereof
IL287665A (en) 2019-04-30 2022-07-01 Celgene Corp Combination therapies comprising apremilast and tyk2 inhibitors
US11440914B2 (en) 2019-05-01 2022-09-13 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US11447494B2 (en) 2019-05-01 2022-09-20 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
CN112142743A (en) * 2019-06-28 2020-12-29 广州诺诚健华医药科技有限公司 Heterocyclic compound, preparation method and application thereof in medicine and pharmacology
CN110330484B (en) * 2019-07-18 2022-08-26 中国药科大学 Substituted phenyl pyrimidine derivative as JAK kinase inhibitor or pharmaceutically acceptable salt thereof, preparation method and application
CA3144420A1 (en) * 2019-08-09 2021-02-18 The National Institutes of Pharmaceutical R&D Co., Ltd. Bridged heterocyclyl-substituted pyrimidine compound, preparation method therefor, and pharmaceutical use thereof
CN116348458A (en) 2019-08-14 2023-06-27 因赛特公司 Imidazolylpyrimidinylamine compounds as CDK2 inhibitors
WO2021072232A1 (en) 2019-10-11 2021-04-15 Incyte Corporation Bicyclic amines as cdk2 inhibitors
CN110734428A (en) * 2019-10-24 2020-01-31 嘉兴特科罗生物科技有限公司 small molecule compounds
CN110862380A (en) * 2019-10-24 2020-03-06 嘉兴特科罗生物科技有限公司 Small molecule compound
CN110627775A (en) * 2019-10-24 2019-12-31 嘉兴特科罗生物科技有限公司 Small molecule compound
TWI782599B (en) * 2020-07-02 2022-11-01 美商輝瑞股份有限公司 Aminopyrimidinyl derivatives
EP3944859A1 (en) 2020-07-30 2022-02-02 Assistance Publique Hôpitaux de Paris Method for treating immune toxicities induced by immune checkpoint inhibitors
CN111961037B (en) * 2020-09-17 2021-09-21 嘉兴特科罗生物科技有限公司 Pharmaceutical compound as JAK kinase inhibitor
WO2023076161A1 (en) 2021-10-25 2023-05-04 Kymera Therapeutics, Inc. Tyk2 degraders and uses thereof
EP4206196A1 (en) * 2021-12-29 2023-07-05 Almirall S.A. Pyrimidine substituted derivatives as tyk2 inhibitors
WO2023208244A1 (en) * 2022-04-29 2023-11-02 南京明德新药研发有限公司 Macrocyclic compound and use thereof
WO2024041397A1 (en) * 2022-08-22 2024-02-29 Hangzhou Highlightll Pharmaceutical Co., Ltd Jak1/jak2/tyk2 inhibitors for topical treatment of dermatological diseases
WO2023151724A1 (en) * 2023-05-19 2023-08-17 凌科药业(杭州)有限公司 Crystal form of pyrazolyl-amino-pyrimidinyl derivative, and preparation method therefor and use thereof

Family Cites Families (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3029871A1 (en) 1979-08-08 1981-02-26 Searle & Co 8-SUBSTITUTED 7-PHENYL-1,2,4-TRIAZOLO ANGLE CLAMP ON 4.3-C ANGLE CLAMP ON PYRIMIDINE-5-AMINE AND AMIDE
NZ337750A (en) 1997-02-12 2001-08-31 Univ Michigan Protein markers for lung cancer and use thereof
GB9714249D0 (en) 1997-07-08 1997-09-10 Angiogene Pharm Ltd Vascular damaging agents
KR100838617B1 (en) 1999-02-10 2008-06-16 아스트라제네카 아베 Quinazoline derivatives as angiogenesis inhibitors
EE05330B1 (en) 1999-11-05 2010-08-16 Astrazeneca Ab Quinazoline derivatives as VEGF inhibitors
MY130363A (en) 2000-02-15 2007-06-29 Sugen Inc "pyrrole substituted 2-indolinone protein kinase inhibitors"
MY130778A (en) * 2001-02-09 2007-07-31 Vertex Pharma Heterocyclic inhibitiors of erk2 and uses thereof
WO2002083667A2 (en) * 2001-04-13 2002-10-24 Vertex Pharmaceuticals Incorporated Inhibitors of c-jun n-terminal kinases (jnk) and other protein kinases
JP4741948B2 (en) * 2002-08-14 2011-08-10 バーテックス ファーマシューティカルズ インコーポレイテッド Protein kinase inhibitors and their use
ATE450530T1 (en) * 2003-03-13 2009-12-15 Vertex Pharma COMPOSITIONS FOR USE AS PROTEIN KINASE INHIBITORS
KR20060060000A (en) 2003-08-05 2006-06-02 코닌클리케 필립스 일렉트로닉스 엔.브이. Video encoding and decoding methods and corresponding devices
WO2006008874A1 (en) * 2004-05-21 2006-01-26 Banyu Pharmaceutical Co., Ltd. SELECTIVE INHIBITORS AGAINST Cdk4 AND Cdk6 HAVING AMINOTHIAZOLE SKELETON
PL1891446T3 (en) 2005-06-14 2013-08-30 Cellzome Gmbh Process for the identification of novel enzyme interacting compounds
JP2009519218A (en) 2005-11-03 2009-05-14 エスジーエックス ファーマシューティカルズ、インコーポレイテッド Pyrimidinyl-thiophene kinase modulator
GB0605691D0 (en) 2006-03-21 2006-05-03 Novartis Ag Organic Compounds
EP1862802B1 (en) 2006-06-01 2008-12-10 Cellzome Ag Methods for the identification of ZAP-70 interacting molecules and for the purification of ZAP-70
DK2152701T3 (en) 2007-03-12 2016-02-15 Ym Biosciences Australia Pty Phenylaminopyrimidinforbindelser and uses thereof
US20090274698A1 (en) 2007-07-06 2009-11-05 Shripad Bhagwat Combination anti-cancer therapy
BRPI0820544A2 (en) * 2007-11-16 2015-06-16 Incyte Corp 4-pyrazolyl-n-arylpyrimidin-2-amines and pyrazolyl-n-heteroarylpyrimidin-2-amines as janus kinase inhibitors
WO2009155551A1 (en) 2008-06-20 2009-12-23 Genentech, Inc. Triazolopyridine jak inhibitor compounds and methods
CL2009001884A1 (en) 2008-10-02 2010-05-14 Incyte Holdings Corp Use of 3-cyclopentyl-3- [4- (7h-pyrrolo [2,3-d] pyrimidin-4-yl) -1h-pyrazol-1-yl) propanonitrile, janus kinase inhibitor, and use of a composition that understands it for the treatment of dry eye.
EP2356107A1 (en) 2008-11-13 2011-08-17 Bayer Cropscience Ag Substituted (pyridyl)-azinylamine derivatives as fungicides
DE102009001438A1 (en) 2009-03-10 2010-09-16 Bayer Schering Pharma Aktiengesellschaft New carbonylamino-substituted anilino-pyrimidine compounds are tyrosine kinase-2 inhibitors, useful for treating diseases associated with inflammatory conditions e.g. bronchitis, rheumatoid arthritis, psoriasis and Guillain Barre syndrome
DE102009015070A1 (en) 2009-03-30 2010-10-14 Bayer Schering Pharma Aktiengesellschaft New phenyl-pyrimidin-2-yl-amine compounds are tyrosine kinase 2 inhibitors useful for treating e.g. rheumatoid arthritis, Crohn's disease, asthma, multiple sclerosis, adult respiratory distress syndrome, allergic alveolitis and uveitis
JP2013522267A (en) 2010-03-17 2013-06-13 エフ.ホフマン−ラ ロシュ アーゲー Imidazopyridine compounds, compositions, and methods of use

Also Published As

Publication number Publication date
JP2014500254A (en) 2014-01-09
EP2638018A1 (en) 2013-09-18
CA2815330A1 (en) 2012-05-18
CN103298794A (en) 2013-09-11
WO2012062704A1 (en) 2012-05-18

Similar Documents

Publication Publication Date Title
AU2011328237A1 (en) Pyridine compounds and aza analogues thereof as TYK2 inhibitors
AU2012288892B2 (en) Heterocyclyl pyrimidine analogues as JAK inhibitors
AU2012357038B2 (en) Pyrimidine-2,4-diamine derivatives as kinase inhibitors
US20130143915A1 (en) Triazolopyridines as tyk2 inhibitors
AU2013265307B2 (en) Heterocyclyl pyrimidine analogues as TYK2 inhibitors
US20120040955A1 (en) Fluoro substituted pyrimidine compounds as jak3 inhibitors
MX2008005717A (en) Aminopyrimidines useful as kinase inhibitors.
EA015751B1 (en) Compounds and compositions as protein kinase inhibitors
US9249129B2 (en) Morpholino substituted urea derivatives as mTOR inhibitors
CA2763730A1 (en) Heterocyclylaminopyrimidines as kinase inhibitors
AU2012311504A1 (en) Pyrazolo[4,3-C]pyridine derivatives as kinase inhibitors
CA2771675A1 (en) Ortho substituted pyrimidine compounds as jak inhibitors
US20080108608A1 (en) Pyrimidines With Tie2 (Tek) Activity
WO2012143320A1 (en) (7h-pyrrolo[2,3-d]pyrimidin-2-yl)amine compounds as jak3 inhibitors
WO2013017479A1 (en) Pyrazolo[4,3-c]pyridine derivatives as jak inhibitors
EP3030561B1 (en) N-cyanomethylamides as inhibitors of janus kinase
RU2564419C1 (en) Heterocyclic analogues of pyrimidines as jak inhibitors
NZ720726A (en) Novel carboxamides, method for the production thereof, pharmaceutical preparations comprising them, and use thereof for producing medicaments

Legal Events

Date Code Title Description
MK1 Application lapsed section 142(2)(a) - no request for examination in relevant period