WO2012069852A1 - Pharmaceutical compounds as inhibitors of sphingosine kinase - Google Patents

Pharmaceutical compounds as inhibitors of sphingosine kinase Download PDF

Info

Publication number
WO2012069852A1
WO2012069852A1 PCT/GB2011/052334 GB2011052334W WO2012069852A1 WO 2012069852 A1 WO2012069852 A1 WO 2012069852A1 GB 2011052334 W GB2011052334 W GB 2011052334W WO 2012069852 A1 WO2012069852 A1 WO 2012069852A1
Authority
WO
WIPO (PCT)
Prior art keywords
tetramethyl
tetrahydronaphthalen
pyridin
butyl
tert
Prior art date
Application number
PCT/GB2011/052334
Other languages
French (fr)
Inventor
Timothy Harrison
Frank Burkamp
Linda Jordan
Mark Bell
Dominic Janssen
Hugues Miel
Mary Mcfarland
Original Assignee
Almac Discovery Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Almac Discovery Limited filed Critical Almac Discovery Limited
Publication of WO2012069852A1 publication Critical patent/WO2012069852A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/04Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/24Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D213/28Radicals substituted by singly-bound oxygen or sulphur atoms
    • C07D213/30Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/76Nitrogen atoms to which a second hetero atom is attached
    • 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
    • C07D239/26Heterocyclic 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 only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • 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
    • C07D239/34One oxygen atom
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • C07D403/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention relates to compounds that are useful as inhibitors of the activity of one or more isoforms of sphingosine kinase. Particularly, although not exclusively, the present invention also relates to pharmaceutical compositions comprising these compounds and to the use of these compounds in the treatment and/or prevention of cancers, hyperproliferative, inflammatory, angiogenic, immune and viral infectious diseases. BACKGROUND TO THE INVENTION
  • Sphingolipids not only represent the major component of the cellular membrane but also serve as a reservoir for important signaling molecules with pleiotropic effects on important cellular processes.
  • Sphingolipid metabolites including ceramide, sphingosine and sphingosine-1 phosphate (S1 P) have emerged as a new class of potent bioactive messengers directly involved in numerous pathological conditions and disorders including cancer, hyperproliferative, inflammatory and immune disorders, angiogenesis, and viral infectious diseases. Consequently, the possibilities of harnessing the sphingolipid metabolic pathways represent a new avenue for the treatment and prevention of a variety of diseases related to undesirable sphingosine kinase (SphK) activity and S1 P homeostasis.
  • SphK undesirable sphingosine kinase
  • Ceramide results from the hydrolysis of sphingomyelin in response to various stimuli including inflammatory cytokines, chemokines, growth factors and hormones. Ceramide is further hydrolysed by ceramidase to produce sphingosine. Sphingosine in turn is rapidly phosphorylated by sphingosine kinase to produce S1 P.
  • ceramide and sphingosine exert antiproliferative effects such as cell growth inhibition, apoptosis induction and/or modulation of senescence.
  • S1 P is a growth promoter and survival factor, acting by up-regulating several anti-apoptotic pathways including PI3K/AKT or nuclear factor- ⁇ (NF-KB).
  • PI3K/AKT PI3K/AKT
  • NF-KB nuclear factor- ⁇
  • SphK sphingosine kinase
  • Both enzymes phosphorylate sphingosine to produce S1 P.
  • the functions of SphK1 and 2 are at least partly redundant as mice lacking either isoforms maintain nearly normal levels of S1 P and lack severe phenotypes.
  • the SphK1 and 2 double knock-out mouse is embryonic lethal due to incomplete maturation of the vascular system and brain.
  • Overexpression of mice SphK1 in NHI-3T3 fibroblasts induces transformation via a Ras-dependant signaling pathway, hence demonstrating the oncogenic function of the enzyme.
  • sphingosine kinases are ubiquitously expressed in cells with increased expression of SphK1 commonly reported in tumor tissues.
  • SphK may be activated by a variety of proliferative factors including Tumor Necrosis Factor-alpha (TNFoc), protein kinase C (PKC), serum or Platelet-Derived Growth Factors (PDGF).
  • TNFoc Tumor Necrosis Factor-alpha
  • PKC protein kinase C
  • PDGF Platelet-Derived Growth Factors
  • S1 P can then act as an intramolecular messenger and/or extracellular ligand for specific G-protein coupled receptors; namely S1 PR1 , S1 PR2, S1 PR3, S1 PR4, S1 PR5.
  • S1 P The intracellular action of S1 P is pro-survival and pro-growth whilst signaling through the S1 P receptors elicits various cellular processes important to tumorigenesis and cancer progression including proliferation, survival, differentiation, transformation and vascularization / angiogenesis.
  • SphK activity SphK1 , SphK2
  • SphK up-regulation correlates in the clinical setting with higher clinical grades and poor patient outcome.
  • down-regulation of SphK (SphK1 , SphK2) by siRNA, inhibition by a dominant-negative SphK mutant or treatment with small molecule inhibitors has demonstrated strong anti-proliferative effects and has been shown to block
  • Cancers that can be treated, managed and/or ameliorated include, but are not restricted to, benign, pre- malignant, metastatic and malignant cancers. Such cancers may include, without limitations, solid tumors as well as hematopoietic cancers.
  • SphK1 overexpression typically impairs treatment efficacy by decreasing the ceramide / S1 P ratio.
  • SphK1 , SphK2 down- regulation of SphK (SphK1 , SphK2) by siRNA or pharmacologic inhibition induces ceramide elevation with concomitant loss of S1 P and apoptosis induction hence overcoming the resistance phenotype.
  • MDR-associated protein MDR-associated protein
  • Pgp P-gylcoprotein
  • SphK inhibitors alone or in combination therapies could provide a new strategy for the treatment and management of therapeutic-resistant cancers. While maintaining and/or enhancing efficacy of current single agent treatment or combination therapies, combinations with SphK (SphK1 , SphK2) inhibitors may provide a better therapeutic profile by improving efficacy and/or reducing unwanted, adverse effects.
  • Treatments that can be used in combination with SphK (SphK1 , SphK2) inhibitors include, but or not restricted to, chemo- and radiotherapy, cancer vaccines, cytokines, immunotherapeutics (including antibodies or immunomodulators), anti- angiogenic agents, hormone therapies and gene therapies.
  • hypoxia Another condition known to promote tumor progression and treatment failure in which the SphK / S1 P pathway is involved is hypoxia. Hypoxia exists in 50-60% of solid tumors. To circumvent periods of insufficient nutrient and oxygen supply, cells evolve
  • hypoxia-inducible factor HIF-1 oc
  • the SphK/S1 P pathway is a strong modulator of HIF- 1 a activity under hypoxic conditions.
  • HIF-1 a positively regulates the expression of SphK1 under hypoxia.
  • Increased expression and activation of SphK1 and S1 P signaling through the S1 P receptors has been correlated with
  • SphK1 SphK2 activity may therefore provide a novel approach to treat hypoxic tumors.
  • Angiogenesis refers to a state in the body whereby various stimuli including growth factors promote the formation of new blood vessels. Excessive angiogenesis is commonly associated with disease progression and/or the production of undesirable effects. For instance, signaling of S1 P through the S1 P1 receptor stimulates NFKB production and promotes the formation of vascular endothelial growth factor (VEGF), COX-2, cytokines and adhesion molecules, all known contributors to VEGF (VEGF), COX-2, cytokines and adhesion molecules, all known contributors to VEGF.
  • VEGF vascular endothelial growth factor
  • angiogenesis In addition, SphK regulates the expression of endothelial isoforms of nitric oxide synthase which have also been implicated in modulating angiogenesis.
  • treatment of mice with anti-S1 P mAb blocked endothelial cell migration, capillary formation, inhibited VEGF- and FGF-induced blood vessel formation and arrested tumor- associated angiogenesis.
  • the S1 P mAb also prevented the release of pro-angiogenic cytokines (IL-8, IL-6) from tumor cells.
  • SphK SphK1 , SphK2
  • S1 P production in vascular endothelial cells can lead to reduction of angiogenesis and provide a novel strategy to treat and/or prevent disorders arising from abnormal angiogenesis.
  • disorders include, but are not limited to, cancer, hemangiomas, arthritis, psoriasis, atherosclerosis, myocardial angiogenesis, diabetic retinopathy and/or macular degeneration.
  • Sphk SphK1 , SphK2
  • S1 P are also emerging as important mediators in
  • SphK1 has been shown to be activated by cytokines including tumors necrosis factor-alpha (TNF-alpha) and interleukinl -beta (IL1 -beta).
  • TNF-alpha tumors necrosis factor-alpha
  • IL1 -beta interleukinl -beta
  • the activation of SphK1 in this pathway has been shown to be, at least in part, required for mediating TNF-alpha and IL1 -beta inflammatory responses in cells, including induction of cyclo-oxygenease 2 (COX2).
  • COX2 cyclo-oxygenease 2
  • SphKs and S1 P have also been implicated in various immune cell functions including mast cell degranulation, migration of neutrophils, maturation and migration of lymphocytes, and T-cell development.
  • Sphingosine kinase (SphK1 , SphK2) inhibitors may be used as single agent or administered in conjunction with known immunomodulators or anti- inflammatory agents.
  • immune disorders include, but are not restricted to ankylosing spondylitis, chronic fatigue immune dysfunction syndrome, Crohn's disease, multiple sclerosis, rheumatoid arthritis, autoimmune hepatitis, polyglandular syndromes, organ transplantation, post-surgical organ failure.
  • inflammatory diseases include, but are not limited to, inflammatory bowel disease, asthma, allergy, septic shock, inflammatory kidney disease, post-surgical organ failure, organ transplantation, multiple sclerosis, skin inflammation and chronic inflammation as a result of viral or bacteria infections.
  • SphK SphK1 , SphK2
  • S1 P are involved in various other pathological states such as diabetes, viral infections including HIV and Hepatitis C as well as diverse hyperproliferative disorders in addition to cancer including, but not limited to, psoriasis which is characterised by local keratinocyte hyperproliferation, mesangial disorders resulting from the abnormal proliferation of mesangial cells in the kidney as well as various vascular disorders such as atherosclerosis and restenosis both characterised by hyperproliferation of vascular smooth muscle cells.
  • psoriasis which is characterised by local keratinocyte hyperproliferation
  • mesangial disorders resulting from the abnormal proliferation of mesangial cells in the kidney well as various vascular disorders such as atherosclerosis and restenosis both characterised by hyperproliferation of vascular smooth muscle cells.
  • vascular disorders such as atherosclerosis and restenosis
  • SphK inhibitors are expected to be of use in the prevention, therapy and/or management of these diseases.
  • Examples of SphK1 inhibitors are provided in WO 2009/156041 and DE 10 2008/029734. These documents provide specific thiazolo-piperidine compounds. It would be useful if there were compounds available that showed improved activity, differential activity between different types of enzymes, differential activity between the sphingosine isoforms, better stability, greater ease of formulation, and an improved side- effect profile. It is an object of the present invention to address these needs in the art, to address the shortcomings of the prior art compounds, or to at least provide the public with a useful alternative.
  • the present invention provides a compound according to formula I
  • RrR 8 are independently hydrogen, lower alkyl, phenyl or benzyl and where X is O, CRR', or NR' where R and R' are independently hydrogen or lower alkyl and where for structures c and d Ri and R 2 may together form an optionally lower alkyl substituted 5 or 6 membered ring and where R 3 is optionally OR" where R" is alkyl or aryl;
  • X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 are independently selected from nitrogen atom or carbon atom optionally substituted with halogen, oxo, cyano, amino, aryl, heteroaryl, optionally substituted lower alkoxyl and optionally substituted lower alkyl,
  • X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 is an oxo-substituted carbon atom at least one of the others is a nitrogen atom;
  • X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 are nitrogen atoms; and wherein Q is a heteroatom-containing alkyl, substituted alkyl, optionally substituted cycloalkyi, optionally substituted aryl, or optionally substituted heterocyclyl containing side chain bound through a heteroatom or directly through a carbon atom; and pharmaceutically acceptable salts, stereoisomers and tautomers thereof.
  • Ri , R 2 , R 5 and R 6 are independently hydrogen, lower alkyl or lower cycloalkyi. More preferably Ri , R 2 , R5 and R 6 are each methyl, or and R 2 are both methyl and R 5 and R 6 are both hydrogen, or R 5 and R 6 are both methyl and Ri and R 2 are both hydrogen. In preferred embodiments where substructure 'a' is present R 3 is benzyloxy and R 4 is hydrogen. In preferred embodiments where substructure 'b' is present, Ri , R 2 , R 7 and R 8 are independently hydrogen, lower alkyl or lower cycloalkyi.
  • Ri , R 2 , R 7 and R 8 are each methyl, or R 7 and R 8 are both methyl and Ri and R 2 are both hydrogen. In particularly preferred compounds R 7 and R 8 are both methyl. In these preferred embodiments, R 3 , R 4 , R 5 and R 6 are each hydrogen.
  • the preferred gem-dimethyl groups may be joined to form a cyclopropyl group or other lower cycloalkyi group.
  • Ri , R 3 , and R 4 are independently hydrogen, lower alkyl or lower cycloalkyi, Ri and R 4 may cooperate to form a lower cycloalkyi group, and R 2 is lower alkyl or lower cycloalkyi; where X is preferably O or NR' where R' is H, lower alkyl or lower cycloalkyi and where R 3 and R 2 may cooperate to form a 5 or 6 membered heterocyclic ring.
  • R 2 is a branched lower alkyl group.
  • substructure 'c' wherein Ri R 2 , R 3 , and R 4 are defined as above.
  • the compound is not morpholine-2-carboxylic acid [6-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)-pyridin-2-yl]-amide or a compound selected from compounds of formula (I I)
  • R 9 and R ⁇ R and R 2 , R 2 and R 3 , R 3 and R 4 , R 4 and R 5 , R 5 and R 6 , R 6 and R 7 , R 7 and R 8 , R 0 and R , R and R 2 , R 2 and R 3 , R 4 and R 5 , R 5 and R 6 , R 6 and R 7 may also in each case together form a cycloalkyl having 3, 4, 5, 6 or 7 C atoms or Het having 3, 4, 5, 6 or 7 ring atoms,
  • R 8 , R 8' represent H, Dor A;
  • R 9 , R 9' represent H, D, A, OR 18 , NR 8 R 18 , F, CI, Br, CN or Het;
  • Mi, M 2 , M 3 , M 4 represent CR 19 or N;
  • Yi, Y 2 represent CR 19 or N;
  • V represents C(R 9 )(R 19' ), NR 9 or is absent;
  • W represents [C(R 9 )(R 9' )] P Z, CO-[C(R 9 )(R 9' )] p Z, [C(R 9 )(R 9' )] P N(R 9 )-Z, CO-N(R 19 ) [C(R 9 )(R 9' )] p Z, N(R 9 )-CO-[C(R 9 )(R 9' )] p Z, CO-0-[C(R 9 )(R 9' )] p Z, C(0)OR 18 , OR 18 , H or D;
  • V, W and Y 2 may also in each case together form a cycloalkyl having 3, 4, 5, 6 or 7 C atoms, in which preferably 1 , 2, 3, 4, 5, 6 or 7 H atoms can be replaced with F, CI, Br, CN and/or OH, OR 19 , OC(0)R 19 , NR 9 C(0)OZ, C(0)OR 19 , C(0)N(R 9 )(R 19' ) or N(R 9 )(R 19' ), or form Het having 3, 4, 5, 6 or 7 ring atoms, wherein Het is preferably a saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms which can be unsubstituted or singly, doubly or triply substituted with Hal, F, CI, Br, CN, A, OR 18 , W, SR 8 , N0 2 , N(R 9 )(R 19' ),
  • Z represents Het, Ar or A
  • A represents unbranched or branched alkyl having 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 C atoms, in which 1 , 2, 3, 4, 5, 6 or 7 H atoms can be replaced with F, CI, Br, CN and/or OH, OR 19 , OC(0)R 19 , NR 9 C(0)OZ, C(0)OR 19 , C(0)N(R 9 )(R 19' ) or N(R 9 )(R 19' );
  • Ar represents phenyl, naphthyl or biphenyl substituted singly, doubly or triply with Hal, F, CI, Br, CN, A, OR 18 , W, SR 8 , N0 2 , N(R 9 )(R 19' ), NR 8 COOZ, OCONHZ, NR 8 S0 2 Z,
  • Het represents, in each case independently of one another, a mononuclear, binuclear or trinuclear saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms which can be unsubstituted or singly, doubly or triply substituted with Hal, F, CI, Br, CN, A, OR 18 , W, SR 18 , N0 2 , N(R 9 )(R 19' ), NR 8 COOZ, OCONHZ, NR 8 S0 2 Z,
  • n 1 , 2 or 3
  • n, o are 0, 1 or 2
  • p 0, 1 , 2, 3 or 4
  • the compound is not a compound selected from the group: morpholine-2-carboxylic acid [6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)- pyridin-2-yl]-amide
  • Q is not a heteroatom-containing optionally substituted aryl.
  • one or more of X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 is an oxo- substituted carbon atom, methyl substituted carbon atom, or fluorine substituted carbon atom.
  • one or more of X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 is a carbon atom or nitrogen atom.
  • X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 are carbon atoms or nitrogen atoms.
  • the ring formed by X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 is aromatic or doubly unsaturated, more preferably the ring is aromatic.
  • the groups X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 are selected so as to form, together with the atoms to which they are bonded, the following ring structures:
  • the groups X 1 , X 2 , X 3 , X 4 , X 5 , and X 6 are selected so as to form, together with the atoms to which they are bonded, a pyrimidine ring.
  • a pyrimidine ring is selected so as to form, together with the atoms to which they are bonded, a pyrimidine ring.
  • the Inventors have found that surprisingly the presence of a pyrimidine group at this position combines good inhibition activity with improved physical properties of the compounds of the invention, such as their solubility, and that certain compounds having the pyrimidine ring present therefore have good pharmacokinetic properties, and are preferred.
  • at least one of the heteroatoms in Q is a nitrogen atom.
  • Q contains at least two heteroatoms, more preferably two nitrogen atoms.
  • Q may be selected from:
  • R10- 14 are selected from hydrogen or optionally substituted lower alkyl, or may combine with other R groups in Q to form optionally substituted cycloalkyl or optionally substituted heterocycloalkyl rings which may be aromatic or non-aromatic.
  • R 0 may, together with a nitrogen atom to which it is bound, form a guanidino group. In other preferred embodiments R 0 may be a double bonded nitrogen atom and any other R x moiety bound to the same atom is absent.
  • one or more aromatic carbon atoms of the benzylamino group may be replaced with N, for example to give those two pyridines shown in the structures above. Where two aromatic carbon atoms are replaced by nitrogen they are preferably arranged to give a pyridazine ring, although pyrimidine and pyrazine arrangements are also available in certain embodiments of the invention.
  • Particulary preferred are compounds of the invention having optionally substituted guanidino or amidino moiety containing Q groups.
  • the compounds of the invention have a guanidine or amidino group forming part of the side-chain 'Q' moiety.
  • the guanidine or amidino group forming part of the side-chain 'Q' moiety may be optionally substituted, preferably with cyano, OH or OR" where R" is optionally substituted lower alkyl.
  • alkyi group refers to an aliphatic group containing at least carbon and hydrogen and containing 1 to 15 carbon atoms, such as 1 to 10 carbon atoms.
  • Attachment to the alkyi group occurs through a carbon atom.
  • Cn alkyi refers to an aliphatic group containing n carbon atoms.
  • a C1 -C10 alkyi group contains 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms.
  • An alkyi group may be straight chained or it may be branched.
  • An alkyi group may contain no ring structures or it may contain one or more rings.
  • the alkyi group is a lower alkyi group, that is one having between 1 and 6 carbon atoms whether branched or straight chain.
  • a "cycloalkyi" group contains at least one ring. It is understood that attachment to a cycloalkyi group is via a ring of the cycloalkyi group.
  • Each ring may contain 3 to 10 atoms, such as 4 to 8 or 5 to 7 atoms.
  • Each ring may be independently selected to contain just carbon atoms or to contain both carbon atoms and from 1 to 4 heteroatoms selected from O, N and S.
  • cyclo-heteroalkyl groups i.e.
  • cycloalkyi groups that contain one or more heteroatoms
  • attachment to the cycloalkyi group may occur either through a carbon atom or, if one or more heteroatoms are contained in a ring, attachment may also occur through a heteroatom contained in a ring.
  • a cycloalkyi group may be mono-cyclic or bi-cyclic.
  • a "C n cycloalkyi" group contains n carbon atoms. All n carbon atoms may be contained in the ring(s) of the cycloalkyi group or one or more of the carbons may not be contained in the ring(s) and may instead form one or more chains branching from the ring.
  • a Cn alkyl group is joined to a separate C m alkyl group containing m carbon atoms to form, for example, a heterocycle, the two alkyl groups contain a total number of m + n carbon atoms.
  • an alkyl group may be saturated or unsaturated.
  • the alkyl group may be an alkenyl group (i.e. contain a carbon-carbon double bond) and / or an alkynyl group (i.e. contain a carbon-carbon triple bond). If the alkyl group is unsaturated, it may contain at least 2 carbon atoms. It is understood that any unsaturated portions of an alkyl group are non-aromatic (aromatic groups fall within the scope of the definition of "aryl'). Any part of the alkyl group may be unsaturated, for example the straight, branched or cyclic portion of an alkyl group may contain a carbon-carbon double bond or a carbon-carbon triple bond. Attachment to an unsaturated alky group may occur through the unsaturated part of the alkyl group or may occur through the unsaturated part of the group.
  • an unsaturated alkyl group may contain 1 to 4 carbon-carbon double bonds or 1 to 3 carbon-carbon triple bonds or 1 to 4 of a combination of carbon-carbon double bonds and carbon-carbon triple bonds.
  • An alkyl group may be substituted with one or more heteroatoms or it may be
  • the substituents are independently selected from one another unless they form a part of a particular functional group (e.g. an amide, amidino, or guanidino group).
  • the heteroatom substituents may in turn be substituted with further carbon-containing groups.
  • the C n or C m prefix that defines the substituted alkyl group refers to the total number of carbons contained in the group, i.e. including the carbon atoms contained in any substituted heteroatomic groups, and the total alkyl group contains 1 to 15 carbon atoms as defined previously.
  • the alkyl group may, for example, contain one or more of CN, C0 2 H, CONH 2 , CONHR, CONR a R b , C0 2 R, NH 2 , NHR, NR a R b , OH, OR, SH, SR, F, CI, Br and I, wherein each R, R a and R b are independently selected groups (e.g. alkyl / aryl groups) attached to the atom to which the group joins through a carbon atom of each group, including wherein R a and R b form a heterocycle that includes the heteroatom to which they are attached.
  • a group containing two C m -C n alkyl moieties that form a cycle that includes, for example, the heteroatom to which they are attached may contain from C 2m to C 2n carbon atoms.
  • unsubstituted saturated alkyl groups containing no cyclic structures include methyl, ethyl, n-propyl, sec-propyl, n-butyl, sec-butyl, te/t-butyl, pentyl (branched or unbranched), hexyl (branched or unbranched), heptyl (branched or unbranched), octyl (branched or unbranched), nonyl (branched or unbranched), and decyl (branched or unbranched).
  • Examples of unsubstitued saturated cyclic alkyl groups include cyclopropyl, cylcobutyl, cyclopentyl and cyclohexyl.
  • Examples of unsaturated alkyl groups include ethenyl, propenyl, butenyl, 2-methybutenyl and cyclohexenyl.
  • aryl group refers to a group containing at least one ring that is aromatic.
  • aryl group is stated as being substituted at a particular position, attachment of the position to the aryl group is onto the aromatic ring of the aryl group itself rather than the position being joined to the aryl group through any non-aromatic side-chain of the aryl group.
  • R is an aryl group in CR
  • the C is attached to the aromatic part of the aryl group.
  • Each ring may be independently selected to contain only carbon atoms or to contain both carbon atoms and from 1 to 4 heteroatoms selected from O, N and S.
  • heteroaryl groups i.e. aryl groups that contain one or more heteroatoms
  • attachment to the aryl group may occur either through a carbon atom or, if one or more heteroatoms are contained in a ring, attachment may also occur through a heteroatom contained in a ring.
  • heteroatoms contained in a ring of a heteroaryl group may be substituted, for example forming an /V-oxide.
  • the aromatic group may be mono-cyclic or bi-cyclic, wherein one or both of the rings of a bi-cyclic system is aromatic.
  • aryl groups include acridinyl, phenyl, carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, benzotriazolyl, furanyl, naphthyl, thienyl, thiazolyl, benzothienyl, benzofuranyl, quinolinyl, isoquinolinyl, oxazolyl, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetrahydroquinolinyl, benzimidazolyl and melaminyl.
  • heterocycle includes within its scope both cycloalkyl groups containing one or more heteroatoms within the ring system and aryl groups containing one or more heteroatoms within the ring system.
  • halo refers to a group selected from chlorine, fluorine, bromine and iodine.
  • each tautomeric form is intended to fall within the scope of the invention.
  • each enantiomer or diastereoisomer is intended to fall within the scope of the invention.
  • one enantiomer or diasterioisomer has more desirable characteristics than the related molecule(s)
  • the one having desirable characteristics is preferred.
  • the compounds of the present invention may be provided as a pro-drug.
  • Prodrugs are transformed, generally in vivo, from one form to the active forms of the drugs described herein.
  • a prodrug may be formed by protecting an amine as a physiological hydrolyzable amide.
  • the compounds of the present invention may be provided in the form of their pharmaceutically acceptable salts or as co-crystals.
  • the compounds may be provided having protonated amine groups.
  • pharmaceutically acceptable salt refers to ionic compounds formed by the addition of an acid to a base. The term refers to such salts that are considered in the art as being suitable for use in contact with a patient, for example in vivo and
  • compositions are generally chosen for their non-toxic, non-irritant characteristics.
  • co-crystal refers to a multi- component molecular crystal, which may comprise non-ionic interactions.
  • salts and co-crystals may be prepared by ion exchange chromatography or by reacting the free base or acidic form of a compound with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid or base in one or more suitable solvents.
  • Salts known in the art to be generally suitable for use in contact with a patient include salts derived from inorganic and / or organic acids, including the hydrobromide, hydrochloride, sulphate, bisulphate, nitrate, acetate, oxalate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate and tartrate. These may include cations based on the alkali and alkaline earth metals, such as sodium, potassium, calcium and magnesium, as well as ammonium,
  • the compounds of the present invention may sometimes exist as zwitterions, which are considered as part of the invention.
  • the compounds of the present invention are useful in the treatment of medical conditions associated with undesirable SphK activity and S1 P homeostasis and to treat a wide range of diseases where SphK acitivity and S1 P has been shown to play a key role including cancers, hyperproliferative, inflammatory, angiogenic, immune and viral infectious diseases.
  • the present invention provides compounds according to the invention for the treatment of a disease or disorder involving associated with SphK activity and S1 P homeostasis.
  • the present invention provides the use of those compounds of the invention having selectivity for Sphkl over Sphk2 for the treatment of a disease or disorder involving or associated with SphK1 activity and S1 P homeostasis.
  • the present invention provides the use of those compounds of the invention having selectivity for Sphk2 over Sphkl for the treatment of a disease or disorder involving or associated with SphK2 activity and S1 P homeostasis.
  • the compounds of the invention are for the treatment of cancers including benign, pre-malignant, metastatic and malignant cancers.
  • the cancers to be treated may include solid tumors as well as hematopoietic cancers.
  • the compounds of the invention are for the treatment of solid cancers.
  • the Inventors have found that surprisingly certain of the compounds developed in the work detailed herein demonstrate an effect not only in inhibiting Sphkl but also a marked inhibition of Sphk2.
  • Such 'dual active' compounds are preferred compounds of the invention and preferred compounds in this regard are detailed below as Examples 39, 41 , 47, 90 and 96.
  • Some compounds of the invention in general show Sphk2 activity and these compounds are also preferred in certain embodiments of the invention.
  • the compounds of the present invention may be administered separately, sequentially, simultaneously, concurrently or may be chronologically staggered with one or more standard therapeutics..
  • the present invention also provides a pharmaceutical composition suitable for clinical use.
  • a pharmaceutical composition may comprise a pharmaceutical carrier and, dispersed therein, a therapeutically effective amount of the compounds of the invention.
  • the composition may be solid or liquid.
  • the pharmaceutical carrier is generally chosen based on the type of administration being used and the pharmaceutical carrier may for example be solid or liquid.
  • the compounds of the invention may be in the same phase or in a different phase than the pharmaceutical carrier.
  • compositions may be formulated according to their particular use and purpose by mixing, for example, excipient, binding agent, lubricant, disintegrating agent, coating material, emulsifier, suspending agent, solvent, stabilizer, absorption enhancer and / or ointment base.
  • the composition may be suitable for oral, injectable, rectal or topical administration.
  • the pharmaceutical composition may be administered orally, such as in the form of tablets, coated tablets, hard or soft gelatine capsules, solutions, emulsions, or suspensions.
  • Administration can also be carried out rectally, for example using suppositories, locally or percutaneously, for example using ointments, creams, gels or solution, or parenterally, for example using injectable solutions.
  • the compounds of the present invention may be admixed with pharmaceutically inert, inorganic or organic excipients.
  • suitable excipients include lactose, mize starch or derivatives thereof, talc or stearic acid or salts thereof.
  • suitable excipients for use with soft gelatine capsules include, for example, vegetable oils, waxes, fats and semi-solid or liquid polyols.
  • excipients include, for example, water, polyols, saccharose, invert sugar and glucose.
  • excipients include, for example, water, alcohols, polyols, glycerine and vegetable oil.
  • excipients include, for example, natural or hardened oils, waxes, fats and semi-solid or liquid polyols.
  • compositions may also contain preserving agents, solublizing agents, stabilizing agents, wetting agents, emulsifiers, sweeteners, colorants, odorants, buffers, coating agents and / or antioxidants.
  • the second drug may be provided in pharmaceutical composition with the present invention or may be provided separately.
  • a pharmaceutical formulation for oral administration may, for example, be granule, tablet, sugar coated tablet, capsule, pill, suspension or emulsion.
  • a sterile aqueous solution may be provided that may contain other substances including, for example, salts and / or glucose to make to solution isotonic.
  • the therapeutic agent may also be administered in the form of a suppository or pessary, or may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder.
  • Sphkl /Sphk2 of greater than 10 ie the activity in inhibiting Sphkl is 10-fold greater than the inhibition of Sphk2.
  • those compounds where the selectivity is much higher than 10, eg 50 or higher are also preferred.
  • those compounds that demonstrate selectivity for Sphk2 over Sphkl of around 10 times or greater are also preferred.
  • the present invention provides the use of the preferred compounds having selectivity as described herein for inhibiting Sphkl or Sphk2.
  • these enzymes are inhibited by the use of a compound of the invention having specificity for that subtype such that the other subtype is relatively less inhibited.
  • DCM Dichloromethane
  • DIPEA Diisopropylethylamine
  • DMA N,N- Dim ethyl acetamide
  • DMAP 4-Dimethylaminopyridine
  • DME Dimethyl ether
  • DMF /V,/V-Dimethylformamide
  • DMSO Dimethylsulfoxide
  • EDCI 1 -Ethyl-3-(3'-dimethylaminopropyl)carbodiimide
  • EtOAc Ethyl acetate
  • h Hour: HATU: 0-(7-Azabenzotriazol-1 -yl)-A/,/V,/V',/V- tetramethyluronium hexafluorophosphate
  • HCI Hydrochloric acid
  • HOBt 1 -
  • THF Tetrahydrofuran
  • TMSCI Trimethylsilyl chloride
  • H NMR spectra were recorded at ambient temperature using a Varian Unity Inova (400MHz) spectrometer with a triple resonance 5mm probe, a Bruker Avance DRX (400MHz) with a 5mm inverse detection triple resonance TXI probe, a Bruker Avance (500MHz) spectrometer with a 5mm QNP probe or a Bruker Avance DPX (300MHz) spectrometer with a standard 5mm dual frequency probe. Chemical shifts are expressed in ppm relative to tetramethylsilane.
  • Method A The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler.
  • the spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered.
  • a purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridgeTM Prep C18 5 ⁇ OBDTM 19 x 50mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide.
  • Method B The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler.
  • the spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered.
  • a purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridgeTM Prep C18 5 ⁇ OBDTM 19 x 50mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide.
  • Method C The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler.
  • the spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered. A purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridgeTM Prep C18 5 ⁇ OBDTM 30 x 100mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide.
  • Method D The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler.
  • the spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered.
  • a purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridgeTM Prep C18 5 ⁇ OBDTM 30 x 100mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide. Gradient - Time Flow ml/min %A %B
  • Method E The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler.
  • the spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered.
  • a purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridgeTM Prep C18 5 ⁇ OBDTM 30 x 100mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide.
  • Method H The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler.
  • the spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered.
  • a purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridgeTM Prep C18 5 ⁇ OBDTM 19 x 50mm column at room temperature. Mobile phase - A) Water 0.1 % formic acid B) Acetonitrile/Water 95: 5 with 0.1 % formic acid.
  • LCMS High Pressure Liquid Chromatography Mass Spectrometry
  • Method F The system consists of an Agilent Technologies 6140 single quadrupole mass spectrometer linked to an Agilent Technologies 1290 Infinity LC system with UV diode array detector and autosampler.
  • the spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode.
  • An LCMS experiment is performed on each sample submitted using the following conditions: LC Column - Zorbax Eclipse Plus C18 RRHD 1 .8micron 50 x 2.1 mm maintained at 40 ⁇ .
  • the system consists of ThermoFinnigan LCQ Advantage Mass Spectrometer with the Surveyor LC system and 200 position autosampler.
  • the spectrometer has an electrospray source operating in positive and negative ion mode.
  • An LC-MS experiment is performed on each sample submitted using the following conditions: LC Column - Luna 3micron Ci 8 50 x 2 mm; Mobile phase -A) Water 0.1 % Formic Acid B) Acetonitrile 0.1 % Formic Acid
  • microwave experiments were carried out using a Biotage InitiatorTM Sixty, which uses a single-mode resonator and dynamic field tuning, both of which give reproducibility and control. Temperature from 60-250 °C can be achieved, and pressures of up to 20bar can be reached.
  • Sphingosine kinase 1 (SPHK1 ) or 2 (SPHK2) were purchased from Invitrogen (lot number 417324B and 683074A respectively). Unless otherwise indicated, all other reagents were purchased from Sigma.
  • the kinase reaction was performed in white polystyrene 384-well plates in 1 x reaction buffer containing 40 mM Tris (pH 7.5), 20 mM MgCI 2 , 0.1 img/mL BSA and 1 mM DTT.
  • SPHK1 (0.26 units) or SPHK2 (0.53 units) were incubated in the presence or absence of the appropriate concentration of the inhibitor (5 ⁇ ) for 30min at room temperature.
  • Inhibitors were dissolved in a 10 mM DMSO stock solution and serially diluted prior the assay to give a fourteen point half log dose response.
  • the Kinase reaction was then initiated by the addition of Ultrapure ATP (20 ⁇ ) (Promega) and Sphingosine (75 ⁇ ) (Echelon) and allowed to incubate at room temperature for 1 hour (total reaction volume of 15 ⁇ ). Sphingosine kinase inhibition was subsequently determined using the ADP-GloTM Kinase Assay as described by the manufacturer (Promega). Curve fitting was performed using the GraphPad Prism software and IC 5 o values determined using the non-linear regression curve fitting model. IC 50 values for the inhibitors of the invention represent the average of at least duplicate experiments.
  • Example 1 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDphenvDpiperazine di(trifluoroacetate)
  • Step 1 6-(3-bromophenyl)- 1 , 1 ,4,4-tetramethyl-1 ,2,3,4-tetrahydronaphthalene:
  • Step 2 tert-butyl 4-(3-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine- 1 -carboxylate:
  • Step 3 1 -(3-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)phenyl)piperazine di( trifluoroacetate):
  • Example 2 1 -(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-4- yl)piperidin-4-amine di(trifluoroacetate)
  • reaction mixture was heated at 100 °C for 18 h, cooled to room temperature, poured onto brine (100 ml), extracted with ethyl acetate, dried over magnesium sulphate, filtered and concentrated in vacuo to afford the title compound as a clear oil, which was used without further purification.
  • Step 3 1-(2-(5,5,8,8-tetramethyl-5fi ,8-tetrahydronaphthalen-2-yl)pyridin-4-yl)pfa 4-amine di(trifluoroacetate): Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), te/t-butyl (1 -(2-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-4-yl)piperidin-4-yl)carbamate (105 mg, 0.23 mmol) was reacted to afford the title compound as a white solid (68 mg, 83%).
  • Example 3 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-4- vDpiperazine di(trifluoroacetate)
  • Step 1 tert-butyl 4-(2-chloropyridin-4-yl)piperazine-1-carboxylate:
  • Step 1 tert-butyl 8-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]decane-2-carboxylate:
  • Step 2 tert-butyl 8-(2-(5A8,8-tetramethyl-5A 7,8-tetrahydronaphthalen-2-yl)pyrid ⁇ yl) -2, 8-diazaspiro[4.5]decane-2-carboxylate:
  • Example 5 1 -(4-(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-4- vDphenvDcvclobutanamine di(trifluoroacetate)
  • Step 1 tert-butyl (1-(4-(2-chloropyridin-4-yl)phenyl)cyclobutyl)carbamate:
  • Step 2 tert-butyl (1-(4-(2-(5,5A8-tetramethyl-5A 7,8-tetrahydronaphthalen-2 ⁇
  • Step 1 tert-butyl 2'-chloro-5,6-dihydro-[4,4'-bipyridine]- 1(2H)-carboxylate:
  • Step 2 tert-butyl 2'-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)-5,6-dihydro- [4,4'-bipyridine]-1(2H)-carboxylate:
  • Step 1 3-bromo-5-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridine:
  • Step 2 tert-butyl 5-(5A8,8-tetramethyl-5A 7,8-tetrahydronaphthalen-2-yl)- ⁇
  • Step 3 5-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)- 1 ',2',3',6'-tetrahydro- 3,4'-bipyridine di(trifluoroacetate):
  • Step 1 tert-butyl 4-(6-(5 ! 5 ! 8 ! 8-tetramethyl-5 ⁇ ,8-tetrahydronaphthalen-2-yl)pyrffl ⁇ yl)benzylcarbamate:
  • Step 2 (4-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine di(trifluoroacetate):
  • Step 1 tert-butyl 4-(4-(5 ! 5 ! 8 ! 8-tetramethyl-5 ⁇ ,8-tetrahydronaphthalen-2-yl)pyrid ⁇ yl)benzylcarbamate:
  • Step 2 (4-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine di(trifluoroacetate):
  • Example 10 1 -(6-(5,5,8,8-tetramethyl-5,6 J,8-tetrahvdronaphthalen-2-yl)pyridin-2- yl)piperidin-4-amine
  • Step 1 4,4,5,5-Tetramethyl-2-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)- 1 , 3, 2-dioxaborolane:
  • Step 2 2-bromo-6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridine:
  • Example 1 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-2-yl) piperazine
  • Example 12 6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)-1 '.2'.3'.6'- tetrahvdro-2,4'-bipyridine
  • Step 1 tert-butyl 6-(5,5 ⁇ -tetramethyl-5 ⁇ ⁇ -tetrahydronaphthalen-2-yl)-5 6'-dihydro- [2,4'-bipyridine]- 1 '(2'H)-carboxylate:
  • Example 13 1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2-yl)Dyridin-2- yl)phenyl)cvclobutanamine dihydrochloride
  • Step 1 tert-butyl 1-(4-(6-(5,5 ⁇ -tetramethyl-5 ⁇ J ⁇ -tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)cyclobutylcarbamate:
  • Step 2 1-(4-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) phenyl)cyclobutanamine hydrochloride:
  • Step 1 tert-butyl 8-(6-(5,5 ⁇ etra et y ⁇ -5 ⁇ etra ydronap t a ⁇ en-2-y ⁇ )pyridin-2- yl) -2, 8-diazaspiro[4.5]decane-2-carboxylate:
  • the reaction mixture was heated at 90 °C for 18 h under nitrogen. After cooling to RT, the mixture was filtered through celite and washed with EtOAc. The combined filtrates were concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si0 2 , gradient 0 to 25 % EtOAc in cyclohexane) to afford the title compound as a yellow foam (34.2 mg, 31 %).
  • Step 2 8-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8- diazaspiro[4.5]decane dihydrochloride:
  • Example 15 1 -(4-(5,5,8,8-tetramethyl-5,67,8-tetrahvdronaphthalen-2-yl)pyridin-2- yl)piperidin-4-amine
  • Step 1 2-chloro-4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridine:
  • Step 2 1-(4-(5,5,8,8-tetramethyl-5fi ,8-tetrahydronaphthalen-2-yl)pyridin
  • Example 16 1 -(4-(5,5,8,8-tetramethyl-5,6 J,8-tetrahvdronaphthalen-2-yl)pyridin-2-yl) piperazine
  • Step 1 1-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) piperazine:
  • Example 17 4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)-1 ',2',3',6'- tetrahvdro-2,4'-bipyridine dihydrochloride
  • Step 1 tert-butyl 4-(5 ! 5A8-tetramethyl-5 ! 6 ! 8-tetrahydronaphthalen-2-yl)-5' ! 6'-dihydro- [2,4'-bipyridine]- 1 '(2'H)-carboxylate:
  • Step 2 4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)- 1 ',2',3',6'-tetrahydro- 2, 4 '-bipyridine dihydrochloride:
  • Example 18 1 -(4-(4-(5,5,8,8-tetramethyl-5,6 J,8-tetrahvdronaphthalen-2-yl)pyridin-2- vDphenvDcvclobutanamine dihvdrochloride
  • Step 1 tert-butyl (1-(4-(4-(5,5A8-tetramethyl-5A7,8-tetrahydronaphthalen-2-y ⁇
  • Step 2 1-(4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) phenyl)cyclobutanamine dihydrochloride:
  • Example 21 1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyrimidin-2- yl)piperidin-4-amine dihydrochloride
  • Step 1 2-chloro-4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidine: To a solution of 2,4-dichloropyrimidine (0.603 g, 4.05 mmol) and 4,4,5, 5-tetramethyl-2- (5,5,8, 8-tetramethyl-5, 6,7, 8-tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (1 .4 g, 4.45 mmol) in DMF (12 ml) was added 2M Na 2 C0 3 (aq) (8.10 ml, 16.20 mmol). Nitrogen was bubbled through the mixture for 5 min.
  • Step 2 tert-butyl (1-(4-(5,5,8,8-tetramethyl-5 ⁇ ,8-tetrahydronaphthalen-2-yl)pyrimidin- 2-yl)piperidin-4-yl)carbamate:
  • tert-butyl 8-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane-2-carboxylate tert- butyl piperidin-4-ylcarbamate (59.9 mg, 0.299 mmol) and 2-chloro-4-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine (75 mg, 0.249 mmol) were reacted to afford the title compound (55.4 mg, 48 %).
  • Step 3 1 -(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)piperidin-4-amine dihydrochloride: Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl (1 -(4-(5, 5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyr
  • Step 1 tert-butyl 4-(4-(5,5,8,8-tetramethyl-5 ⁇ ,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)piperazine- 1 -carboxylate:
  • tert-butyl 8-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane-2-carboxylate tert- butyl piperazine-1 -carboxylate (55.7 mg, 0.299 mmol) and 2-chloro-4-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine (75 mg, 0.249 mmol) were reacted to afford the title compound (40.5 mg, 36 %).
  • Step 2 2-(piperazin- 1 -yl) -4-(5, 5, 8, 8-tetramethyl-5, 6, 7, 8-tetrahydronaphthalen-2-yl) pyrimidine dihydrochloride:
  • Example 24 2-(1 -methyl-1 ,2.3.6-tetrahvdropyridin-4-yl)-4-(5.5.8.8-tetramethyl-5.6.7.8- tetrahvdronaphthalen-2-yl)pyrimidine
  • Example 25 1 -(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2-yl)Dyrimidin-2- vDphenvDcyclobutanamine dihydrochloride
  • Step 1 tert-butyl (1-(4-(4-(5,5A8-tetramethyl-5,6 ,8-tetrahydronaphthalen-2- yl)pyrimidin-2-yl)phenyl)cyclobutyl)carbamate:
  • Step 2 1-(4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)phenyl)cyclobutanamine dihydrochloride:
  • Step 1 tert-butyl 8-(4-(5,5,8,8-tetramethyl-5 ⁇ ,8-tetrahydronaphthalen-2-yl)pyrimidin yl) -2, 8-diazaspiro[4.5]decane-2-carboxylate:
  • Step 2 8-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)-2,8- diazaspiro[4.5]decane dihydrochloride:
  • Example 30 1 -(2-(5,5,8,8-tetramethyl-5,67,8-tetrahvdronaphthalen-2-yl)pyrimidin-4- yl)piperidin-4-amine dihydrochloride
  • Step 1 tert-butyl (1 -(2-chloropyrimidin-4-yl)piperidin-4-yl)carbamate:
  • Step 2 tert-butyl (1-(2-(5,5A8-tetramethyl-5A 7,8-tetrahydronaphthalen-2-yl)pyr ⁇ 4-yl)piperidin-4-yl)carbamate:
  • Step 1 tert-butyl 4-(2-chloropyrimidin-4-yl)piperazine- 1-carboxylate:
  • Step 2 tert-butyl (1-(2-(5,5 ⁇ -tetramethyl-5 ⁇ ⁇ -tetrahydronaphthalen-2-yl)pyr ⁇ 4-yl)piperidin-4-yl)carbamate:
  • Step 2 2-(4-Bromo-2-isopropoxyphenyl)propan-2-ol: To a solution of methylmagnesium chloride (3M in THF, 4.88 ml, 14.65 mmol) in THF (4 ml) was slowly added a solution of methyl 4-bromo-2-isopropoxybenzoate (1 g, 3.66 mmol) in THF (4 ml). After stirring at 60 °C for 7 h, the mixture was added to 2N HCI (10 ml) and then partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were dried (MgS0 4 ) and concentrated in vacuo to afford the title compound as a brown oil (1 .0 g, 100%).
  • Step 4 1-(6-(3-isopropoxy-4-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride: To a solution of tert-butyl 4-(6-(4-(2-hydroxypropan-2-yl)-3-isopropoxyphenyl)pyridin-2- yl)piperazine-1 -carboxylate (0.1 g, 0.219 mmol) in DCM (1 ml) at 0 °C, was added triethylsilane (0.1 15 ml, 0.720 mmol) and borontrifluoride diethyletherate (0.088 ml, 0.713 mmol).
  • Step 3 tert-butyl 4-(6-(3-(2-hydroxypropan-2-yl)-4-isopropoxyphenyl)pyridin-2- yl)piperazine- 1 -carboxylate: Following the procedure for tert-butyl 4-(6-(4-(2-hydroxypropan-2-yl)-3- isopropoxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate, 2-(5-bromo-2- isopropoxyphenyl)propan-2-ol (250 mg, 0.915 mmol) was reacted to afford the title compound as an off-white foam (139 mg, 50%).
  • Step 4 1-(6-(4-isopropoxy-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride: Following the procedure for 1 -(6-(3-isopropoxy-4-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride, tert-butyl 4-(6-(3-(2-hydroxypropan-2-yl)-4-isopropoxyphenyl)pyridin-2- yl)piperazine-1 -carboxylate (0.1 1 g, 0.241 mmol) was reacted to afford the title compound as a yellow solid (78 mg, 78%).
  • Step 1 tert-butyl 4-(6-chloropyrazin-2-yl)piperazine-1 -carboxylate;
  • Te -butyl 4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrazin-2- yl)piperazine-1 -carboxylate (0.15 g, 0.33 mmol) was added to a solution of 4N HCI in dioxane (2 ml). The resulting solution was heated at 40 °C for 30 min then cooled to room temperature. The resulting precipitate was filtered and dried at ambient to give the title compound (0.1 18 g, 84%).
  • Example 35 1 -(5-(5,5,8,8-tetramethyl-5,6 ,7,8-tetrahvdronaphthalen-2-yl)pyridin-3- vDpiperazine trihvdrochloride
  • Step 1 tert-butyl 4-(5-bromopyridin-3-yl)piperazine-1 -carboxylate: A solution of 3,5-dibromopyridine (0.6 g, 2.53 mmol), tert-butyl piperazine-1 -carboxylate (0.71 g, 3.8 mmol), Pd 2 dba 3 (0.07 g, 0.076 mmol), cesium carbonate (0.82 g, 2.53 mmol) and xantphos (0.132 g, 0.228 mmol) in dioxane (10 ml) was heated at 1 15 °C for 15 h. The solution was cooled to room temperature and concentrated in vacuo.
  • Step 2 tert-butyl 4-(5-(5,5 ⁇ etra et y ⁇ -5 ⁇ etra ydronap t a ⁇ en-2-y ⁇ )pyridin-3- yl)piperazine- 1 -carboxylate:
  • tert-butyl 4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperazine-1 -carboxylate tert-butyl 4-(5- bromopyridin-3-yl)piperazine-1 -carboxylate (0.2 g, 0.58 mmol) was reacted to give the title compound (0.15 g, 57%).
  • Step 3 ⁇ -(5-(5,5 ⁇ etra et y ⁇ -5 ⁇ etra ydronap t a ⁇ en-2-y ⁇ )pyridin-3- yl)piperazine trihydrochloride:
  • Example 36 1 -(5-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-3- yl)piperidin-4-amine trihydrochloride
  • Step 1 tert-butyl 1-(5-bromopyridin-3-yl)piperidin-4-ylcarbamate:
  • Step 2 tert-butyl 1-(5-(5 -tetramethyl-5fi -tetrahydronaphthalen-2-yl)pyrM ⁇ yl)piperidin-4-ylcarbamate:
  • tert-butyl 4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperazine-1 -carboxylate tert-butyl 1-(5- bromopyridin-3-yl)piperidin-4-ylcarbamate (0.2 g, 0.56 mmol) was reacted to give the title compound (0.125 g, 48%).
  • Step 3 1-(5-(5 -tetramethyl-5fi -tetrahydronaphthalen-2-yl)pyrti ⁇
  • Example 37 4-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDpyrimidine dihydrochloride
  • Step 1 tert-butyl 4-(6-chloropyrimidin-4-yl)piperazine- 1-carboxylate:
  • Step 2 tert-butyl 4-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-4- yl)piperazine- 1 -carboxylate:
  • tert-butyl 4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperazine-1 -carboxylate tert-butyl 4-(6- chloropyrimidin-4-yl)piperazine-1 -carboxylate (0.19 g, 0.63 mmol) was reacted to give the title compound (0.20 g, 72%).
  • Step 3 4-(piperazin-1-yl)-6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidine dihydrochloride:
  • Step 1 tert-butyl 1-(6-chloropyrimidin-4-yl)piperidin-4-ylcarbamate:
  • Step 2 tert-butyl 1-(6-(5,5,8,8-tetramethyl-5fi ,8-tetrahydronaphthalen-2-yl)pyrimidin-4- yl)piperidin-4-ylcarbamate:
  • Step 3 1-(6-(5,5,8,8-tetramethyl-5fi ,8-tetrahydronaphthalen-2-yl)pyrimidin-4- yl)piperidin-4-amine dihydrochloride:
  • Step 1 tert-butyl 1-(6-chloropyrazin-2-yl)piperidin-4-ylcarbamate:
  • Step 2 tert-butyl 1-(6-(5 ⁇ -tetramethyl-5 ⁇ ⁇ -tetrahydronaphthalen-2-yl)pyrazin-2- yl)piperidin -4 -ylcarbamate
  • tert-butyl 4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperazine-1 -carboxylate tert-butyl 1 -(6- chloropyrazin-2-yl)piperidin-4-ylcarbamate (0.15g, 0.48mmol) was reacted to give the title compound (0.15 g, 71 %).
  • Step 3 1-(6-(5 ⁇ -tetramethyl-5 ⁇ ⁇ -tetrahydronaphthalen-2-yl)pyrazin-2- yl)piperidin-4-amine dihydrochloride:
  • Example 40 1 -(2-methoxyethyl)-4-(6-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen- 2-yl)pyridin-2-yl)piperazine dihydrochloride
  • the product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo.
  • the resultant residue was subjected to flash chromatography (KP-NH, Si0 2 , gradient 0 to 50 % ethyl acetate in cyclohexane) to afford the title compound as a yellow oil.
  • the oil was dissolved in dioxane and 4M HCI in dioxane was added. The solution was stirred at room temperature for 5 min. before being concentrated in vacuo.
  • the residue was triturated with diethyl ether to give the target compound as a yellow solid (121 mg, 52%).
  • Example 42 A/,A/-dimethyl-2-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2- yl)pyridin-2-yl)piperazin-1 -yl)acetamide
  • Example 44 A/.A/-dimethyl-2-(4-(6-(5.5.8.8-tetramethyl-5.6.7.8-tetrahvdronaphthalen-2- yl)pyridin-2-yl)piperazin-1 -vDethanamine trihydrochloride
  • the product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo.
  • the resultant residue was subjected to flash chromatography (KP-NH, Si0 2 , gradient 0 to 50 % ethyl acetate in cyclohexane) to afford the title compound as a clear oil.
  • the oil was dissolved in dioxane and 4M HCI in dioxane was added. The solution was stirred at room temperature for 5 min. before being concentrated in vacuo.
  • the residue was triturated with diethyl ether to give the target compound as a yellow solid (24 mg, 34%).
  • Example 45 3-hvdroxy-2-(hvdroxymethyl)-2-methyl-1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahvdronaphthalen-2-yl)pyridin-2-yl)piperazin-1 -yl)propan-1 -one hydrochloride
  • Example 46 4-(piperidin-4-yloxy)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDpyrimidine di(trifluoroacetate)
  • Step 1 tert-butyl 4-((2-chloropyrimidin-4-yl)oxy)piperidine- 1-carboxylate:
  • Step 2 tert-butyl 4-((2-(5 ! 5A8-tetramethyl-5 ⁇ ! 8-tetrahydronaphthalen-2-yl)pyrim 4-yl)oxy)piperidine- 1 -carboxylate:
  • Step 3 4-(piperidin-4-yloxy)-2-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidine di(trifluoroacetate):
  • Step 1 tert-butyl 4-((4-chloropyrimidin-2-yl)oxy)piperidine- 1 -carboxylate:
  • Step 2 tert-butyl 4-((4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin- 2-yl)oxy)piperidine- 1 -carboxylate:
  • Example 48 2-methyl-4-(piperazin-1 -yl)-6-(5,5, 8,8-tetramethyl-5, 6,7,8- tetrahvdronaphthalen-2-yl)pyrimidine di(trifluoroacetate)
  • Step 1 tert-butyl 4-(6-chloro-2-methylpyrimidin-4-yl)piperazine-1-carboxylate:
  • Step 2 tert-butyl 4-(2-methyl-6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidin-4-yl)piperazine- 1 -carboxylate:
  • Step 3 2-(piperidin-4-yloxy)-4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidine di(trifluoroacetate): Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), te/t-butyl 4-(2-methyl-6-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperazine-1 -carboxylate (120 mg , 0.26 mmol) was reacted to afford the title compound as a white solid (82 mg, 87%).
  • Step 2 tert-butyl 4-(6-(4-hydroxy-3-isopropylphenyl)pyridin-2-yl)piperazine-1- carboxylate:
  • Step 3 tert-butyl 4-(6-(4-isobutoxy-3-isopropylphenyl)pyridin-2-yl)piperazine-1- carboxylate: Following the procedure for Methyl 4-bromo-2-isopropoxybenzoate, tert-butyl 4-(6-(4- hydroxy-3-isopropylphenyl)pyridin-2-yl)piperazine-1 -carboxylate (150 mg, 0.377 mmol) and 1 -bromo-2-methylpropane (0.062 ml, 0.566 mmol) were reacted to afford the title compound as a yellow solid (37.7 mg, 22%).
  • Step 4 1-(6-(4-isobutoxy-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride: Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl 4-(6-(4-isobutoxy-3- isopropylphenyl)pyridin-2-yl)piperazine-1 -carboxylate (37.7 mg, 0.083 mmol) was reacted to afford the title compound as a yellow solid (20.9 mg, 59%).
  • Step 2 tert-butyl 4-(6-(3-(tert-butyl)-4-hydroxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
  • Step 3 tert-butyl 4-(6-(3-(tert-butyl)-4-isopropoxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
  • Step 4 1-(6-(3-(tert-butyl)-4-isopropoxyphenyl)pyridin-2-yl)piperazine dihydrochloride: Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl 4-(6-(3-(tert-butyl)-4- isopropoxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate (60 mg, 0.132 mmol) was reacted to afford the title compound as a yellow solid (35.6 mg, 63%).
  • Step 1 tert-butyl 4-(6-(3-(tert-butyl)-4-isobutoxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
  • Step 2 1-(6-(3-(tert-butyl)-4-isobutoxyphenyl)pyridin-2-yl)piperazine dihydrochloride: Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl 4-(6-(3-(tert-butyl)-4- isobutoxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate (43.6 mg, 0.093 mmol) was reacted to afford the title compound as a yellow solid (25.0 mg, 61 %).
  • Example 52 1 -(6-(2-(benzyloxy)-1 .1 ,3,3-tetramethyl-2,3-dihvdro-1 H-inden-5-yl)pyridin-
  • Step 1 1 , 1 ,3,3-tetramethyl- 1H-inden-2(3H)-one:
  • Step 3 5-bromo- 1, 1,3,3-tetramethyl-2,3-dihydro-1H-inden-2-ol:
  • Step 4 2-(benzyloxy)-5-bromo- 1 , 1 ,3,3-tetramethyl-2,3-dihydro- 1 H-indene:
  • Step 5 2-(2-(benzyloxy)- 1, 1 ,3,3-tetramethyl-2,3-dihydro-1 H-inden-5-yl)-4,4,5,5- tetramethyl- 1,3,2-dioxaborolane:
  • Nitrogen was bubbled through a suspension of 2-(benzyloxy)-5-bromo-1 ,1 ,3,3- tetramethyl-2,3-dihydro-1 H-indene (1 .1 g, 3.06 mmol), PdCI 2 (dppf) CH 2 CI 2 (0.250 g, 0.306 mmol), potassium acetate (0.9 g, 9.18 mmol) and 4,4,4',4', 5,5,5', 5'-octamethyl- 2,2'-bi(1 ,3,2-dioxaborolane) (1 .166 g, 4.59 mmol) in DMF (10 ml) for 10 minutes. The mixture was heated at 100 ' ⁇ for 15h.
  • Step 6 tert-butyl 1-(6-(2-(benzyloxy)-1 , 1,3,3-tetramethyl-2,3-dihydro- 1H-inden-5- yl)pyridin-2-yl)piperidin-4-ylcarbamate:
  • Step 7 1-(6-(2-(benzyloxy)-1, 1 ,3 etramethyl-2,3-dihydro-1H-inden-5-yl)pyridin-2- yl)piperidin-4-amine 1 -(6-(2-(benzyloxy)-1 ,1 ,3,3-tetramethyl-2,3- dihydro-1 H-inden-5-yl)pyridin-2-yl)piperidin-4-ylcarbamate (50 mg, 0.09 mmol) was added to a 4N HCI solution in dioxane (2ml_, 8mmol). The resulting solution was stirrred at 50 °C for 15 min then concentrated in vacuo to afford the title compound (48 mg, 100%).
  • Step 1 4-bromo-2-(tert-butyl)- 1-isopropoxybenzene:
  • Step 2 2-(3-(tert-butyl)-4-isopropoxyphenyl)-4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolane: To a solution of 4-bromo-2-(tert-butyl)-1 -isopropoxybenzene (1 1 .8 g, 43.5 mmol) in DMF (150 ml) was added 4,4,4',4', 5,5, 5', 5'-octamethyl-2,2'-bi(1 ,3,2-dioxaborolane) (16.57 g, 65.3 mmol) and potassium acetate (12.81 g, 131 mmol).
  • Step 4 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin- 1 -yl)ethanol: To a solution of 2-(piperazin-1 -yl)ethanol (42.7mg, 0.328 mmol) in THF (0.5 ml), in a sealable vial, was added DIPEA (0.086 ml, 0.492 mmol). A solution of 4-(3-(tert-butyl)-4- isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) in THF (0.500 ml) was added and the vial was sealed.
  • Example 54 cis-A/ l -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)cvclohexane-1 ,4- diamine
  • Example 58 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-4-methyl-1 A- diazepane
  • Step 1 2-(4-isopropoxy-3-isopropylphenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane: Following the procedure for 2-(3-(tert-butyl)-4-isopropoxyphenyl)-4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolane, 4-bromo-1 -isopropoxy-2-isopropylbenzene (1 .67 g, 6.49 mmol) was reacted to afford the title compound (686 mg, 35%).
  • Step 3 tert-butyl 4-(4-(4-isopropoxy-3-isopropylphenyl)pyrimidin-2-yl)piperazine-1- carboxylate:
  • Step 4 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin- 1-yl)pyrimidine dihydrochloride: To a solution of tert-butyl 4-(4-(4-isopropoxy-3-isopropylphenyl)pyrimidin-2-yl)piperazine- 1 -carboxylate (56.3 mg, 0.128 mmol) in dioxane (0.5 ml) was added HCI (4M in dioxane, 1 ml, 4.00 mmol). The mixture was stirred at room temperature for 1 h, and then further HCI (4M in dioxane, 0.5 ml, 2.00 mmol) was added. After 1 h at room temperature the mixture was concentrated in vacuo . The resultant solid was slurried in Et 2 0, then lyophilized o/n to afford the title compound as a white solid (19 mg, 36%).
  • Step 1 tert-butyl 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine- 1- carboxylate:
  • Step 2 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(piperazin- 1 -yl)pyrimidine dihydrochloride: Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin- 2-yl)piperazine-1 -carboxylate (39.4 mg, 0.087 mmol) was reacted to afford the title compound as a white solid (16 mg, 44%).
  • Example 66 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-amine dihydrochloride
  • Step 1 tert-butyl 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine- 1- carboxylate:
  • Step 1 4-bromo-2-(tert-butyl)- 1-(cyclopentyloxy)benzene:
  • Step 2 2-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)-4,4,5,5-tetramethyl- 1,3,2- dioxaborolane:
  • Step 3 4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)-2-chloropyrimidine:
  • Step 4 1-(4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)pyrimidin-2-yl)piperidin-4-am dihydrochloride:
  • Example 68 1 -(2-(3-isopropoxy-4-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine dihydrochloride
  • Step 1 tert-butyl (1 -(2-chloropyrimidin-4-yl)piperidin-4-yl)carbamate:
  • Step 2 tert-butyl (1-(2-(3-isopropoxy-4-isopropylphenyl)pyrimidin-4-yl)piperidin-4- yl) carbarn ate:
  • Step 3 1 -(2-(3-isopropoxy-4-isopropylphenyl)pyrimidin-4-yl)piperidin
  • Step 1 tert-butyl 4-(6-(3-hydroxy-4-isopropylphenyl)pyridin-2-yl)piperazine-1- carboxylate:
  • Step 2 tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2-yl)piperazine-1- carboxylate:
  • Example 70 1 -(6-(4-isopropyl-3-propoxyphenyl)pyridin-2-yl)piperazine dihydrochloride
  • Step 1 tert-butyl 4-(6-(4-isopropyl-3-propoxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
  • Step 1 tert-butyl 4-(6-(3-(cyclopentyloxy)-4-isopropylphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
  • Example 72 1 -(6-(4-isopropyl-3-(prop-1 -en-2-yl)phenyl)pyridin-2-yl)piperazine dihydrochloride
  • Step 1 tert-butyl 4-(6-(4-isopropyl-3-(((trifluoromethyl)sulfonyl)oxy)phenyl)pyridin-2- yl)piperazine- 1 -carboxylate:
  • Step 1 tert-butyl 4-(6-(3,4-diisopropylphenyl)pyridin-2-yl)piperazine- 1 -carboxylate:
  • Step 1 tert-butyl 4-(6-(3-isopropyl-4-propoxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
  • Example 75 1 -(6-(4-(cvclopentyloxy)-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride
  • Step 1 tert-butyl 4-(6-(4-(cyclopentyloxy)-3-isopropylphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
  • Example 76 (rac)-1 -(6-(3-isopropyl-4-((1 .1 ,1 -trifluoropropan-2-yl)oxy)phenyl)pyridin-2- vDpiperazine dihydrochloride
  • Step 1 (rac)-tert-butyl 4-(6-(3-isopropyl-4-((1 , 1, 1-trifluoropropan-2-yl)oxy)phenyl)pyridin- 2-yl)piperazine- 1 -carboxylate:
  • Step 2 (rac)-1 -(6-(3-isopropyl-4-((1 , 1, 1-trifluoropropan-2-yl)oxy)phenyl)pyridin-2- yl)piperazine dihydrochloride:
  • Step 1 (rac)-tert-butyl 4-(6-(4-(sec-butoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
  • Step 2 (rac)-1-(6-(4-(sec-butoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine
  • Step 1 tert-butyl 4-(6-(3-isopropyl-4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazin 1-carboxylate:
  • Example 82 1 -(6-(3-(tert-butyl)-4-(2.2.2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazine dihydrochloride
  • Step 1 tert-butyl 4-(6-(3-(ten-butyl)-4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2- yl)piperazine- 1 -carboxylate:
  • tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate tert-butyl 4-(6-(3-(tert-butyl)-4-hydroxyphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.292 mmol) and 1 ,1 ,1 -trifluoro-2-iodoethane (0.043 ml, 0.437 mmol) were reacted to afford the title compound as a yellow oil (51 .2 mg, 36%).
  • Step 2 1 -(6-(3-(ten-butyl)-4-(2,2,2-trifluoroethoxy)phenyl)pyrid ⁇
  • Example 83 1 -(2-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine dihydrochloride
  • Step 1 tert-butyl (1-(2-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4- yl) carbarn ate:

Abstract

The present invention relates to compounds that are useful as inhibitors of the activity of one or more isoforms of sphingosine kinase. Particularly, although not exclusively, the present invention also relates to pharmaceutical compositions comprising these compounds and to the use of these compounds in the treatment and/or prevention of cancers, hyperproliferative, inflammatory, angiogenic, immune and viral infectious diseases.

Description

PHARMACEUTICAL COMPOUNDS AS INHIBITORS OF SPHINGOSINE KINASE
The present invention relates to compounds that are useful as inhibitors of the activity of one or more isoforms of sphingosine kinase. Particularly, although not exclusively, the present invention also relates to pharmaceutical compositions comprising these compounds and to the use of these compounds in the treatment and/or prevention of cancers, hyperproliferative, inflammatory, angiogenic, immune and viral infectious diseases. BACKGROUND TO THE INVENTION
The past few years have seen an upsurge of interest in the field of sphingolipids.
Sphingolipids not only represent the major component of the cellular membrane but also serve as a reservoir for important signaling molecules with pleiotropic effects on important cellular processes. Sphingolipid metabolites including ceramide, sphingosine and sphingosine-1 phosphate (S1 P), have emerged as a new class of potent bioactive messengers directly involved in numerous pathological conditions and disorders including cancer, hyperproliferative, inflammatory and immune disorders, angiogenesis, and viral infectious diseases. Consequently, the possibilities of harnessing the sphingolipid metabolic pathways represent a new avenue for the treatment and prevention of a variety of diseases related to undesirable sphingosine kinase (SphK) activity and S1 P homeostasis.
Ceramide results from the hydrolysis of sphingomyelin in response to various stimuli including inflammatory cytokines, chemokines, growth factors and hormones. Ceramide is further hydrolysed by ceramidase to produce sphingosine. Sphingosine in turn is rapidly phosphorylated by sphingosine kinase to produce S1 P.
Cells maintain a tightly-regulated and dynamic balance of distinct sphingolipid metabolites. Evidence demonstrates that ceramide and sphingosine exert antiproliferative effects such as cell growth inhibition, apoptosis induction and/or modulation of senescence. On the contrary, S1 P is a growth promoter and survival factor, acting by up-regulating several anti-apoptotic pathways including PI3K/AKT or nuclear factor-κΒ (NF-KB). Thus, the dynamic balance between intracellular levels of ceramide and/or sphingosine and S1 P, as well as the signaling that control this balance, are critical factors that dictate cell fate (i.e death vs survival). Altering the sphingolipid rheostat thus represents a promising approach to effective therapy.
A complex network of enzymes regulates the balance between the different sphingolipid metabolites in mammalian cells. However, a key enzyme in the sphingolipid metabolic pathway is sphingosine kinase (Sphk), which is responsible for the production of S1 P. Therefore, SphK represents a target for drug discovery and for the development of new therapeutics to treat a wide range of diseases where sphingosine pathway members have been shown to play a role. To date, two isoforms of sphingosine kinase have been identified and characterised, namely sphingosine kinase 1 and sphingosine kinase 2 (herein referred to as SphK1 and SphK2 respectively). Both enzymes phosphorylate sphingosine to produce S1 P. The functions of SphK1 and 2 are at least partly redundant as mice lacking either isoforms maintain nearly normal levels of S1 P and lack severe phenotypes. In contrast, the SphK1 and 2 double knock-out mouse is embryonic lethal due to incomplete maturation of the vascular system and brain. Overexpression of mice SphK1 in NHI-3T3 fibroblasts induces transformation via a Ras-dependant signaling pathway, hence demonstrating the oncogenic function of the enzyme. In humans, sphingosine kinases are ubiquitously expressed in cells with increased expression of SphK1 commonly reported in tumor tissues. SphK may be activated by a variety of proliferative factors including Tumor Necrosis Factor-alpha (TNFoc), protein kinase C (PKC), serum or Platelet-Derived Growth Factors (PDGF). Overexpression of SphK (SphK1 , SphK2) leads to increased S1 P generation. S1 P can then act as an intramolecular messenger and/or extracellular ligand for specific G-protein coupled receptors; namely S1 PR1 , S1 PR2, S1 PR3, S1 PR4, S1 PR5. The intracellular action of S1 P is pro-survival and pro-growth whilst signaling through the S1 P receptors elicits various cellular processes important to tumorigenesis and cancer progression including proliferation, survival, differentiation, transformation and vascularization / angiogenesis. Additional studies also implicate SphK activity (SphK1 , SphK2) in adhesion and migration of cancer cells, properties associated with metastatic potential and advanced state disease. Accordingly, SphK up-regulation correlates in the clinical setting with higher clinical grades and poor patient outcome. Importantly however, down-regulation of SphK (SphK1 , SphK2) by siRNA, inhibition by a dominant-negative SphK mutant or treatment with small molecule inhibitors has demonstrated strong anti-proliferative effects and has been shown to block
tumorigenesis and angiogenesis as well as metastasis in mice xeno- or allograft cancer models. Together these findings highlight the critical role SphK (SphK1 , SphK2) and S1 P signaling plays in the development and progression of cancer. Cancers that can be treated, managed and/or ameliorated include, but are not restricted to, benign, pre- malignant, metastatic and malignant cancers. Such cancers may include, without limitations, solid tumors as well as hematopoietic cancers.
One of the main obstacles involved in cancer therapy is the development of drug resistance. In this regard, numerous studies have reported a correlation between SphK activity and resistance to chemotherapy or irradiation. SphK1 overexpression typically impairs treatment efficacy by decreasing the ceramide / S1 P ratio. Interestingly, down- regulation of SphK (SphK1 , SphK2) by siRNA or pharmacologic inhibition induces ceramide elevation with concomitant loss of S1 P and apoptosis induction hence overcoming the resistance phenotype. Up-regulation of the drug-efflux pumps such as the MDR-associated protein (MRP-1 ) and P-gylcoprotein (Pgp) may represent an alternative mechanism for the development of multi-drug resistance (MDR) phenotypes in cancer. In a recent study performed in brain tumor-derived endothelial cells, overexpression of SphK1 was shown to enhance the expression of the drug-efflux- pumps at the mRNA and protein levels. S1 P was also shown to stimulate the transport activity of Pgp via activation of the S1 P1 and S1 P3 receptors. These findings
demonstrate the important role SphK (SphK1 , SphK2) play in the development and maintenance of MDR. Conseqently, SphK inhibitors alone or in combination therapies could provide a new strategy for the treatment and management of therapeutic-resistant cancers. While maintaining and/or enhancing efficacy of current single agent treatment or combination therapies, combinations with SphK (SphK1 , SphK2) inhibitors may provide a better therapeutic profile by improving efficacy and/or reducing unwanted, adverse effects. Treatments that can be used in combination with SphK (SphK1 , SphK2) inhibitors include, but or not restricted to, chemo- and radiotherapy, cancer vaccines, cytokines, immunotherapeutics (including antibodies or immunomodulators), anti- angiogenic agents, hormone therapies and gene therapies.
Another condition known to promote tumor progression and treatment failure in which the SphK / S1 P pathway is involved is hypoxia. Hypoxia exists in 50-60% of solid tumors. To circumvent periods of insufficient nutrient and oxygen supply, cells evolve
mechanisms to adapt to such conditions in which the hypoxia-inducible factor (HIF-1 oc) is core. Recent studies indicate that the SphK/S1 P pathway is a strong modulator of HIF- 1 a activity under hypoxic conditions. Likewise, HIF-1 a positively regulates the expression of SphK1 under hypoxia. Increased expression and activation of SphK1 and S1 P signaling through the S1 P receptors has been correlated with
vascularization/angiogenesis as well as enhanced survival via activation of the
AKT/GSK33 pathway. Enhanced survival following activation of the p42/44 MAPK signaling as a result of SphK2 activation has also been reported under hypoxia in A549 cells. These findings demonstrate the implication of the SphK / S1 P pathway in mediating hypoxia-related molecular changes that allow cancer cells to adapt and survive hypoxic tumor environments. Interfering with S1 P levels by inhibiting SphK
(SphK1 , SphK2) activity may therefore provide a novel approach to treat hypoxic tumors.
Recent evidence also implicates up-regulation of SphK (SphK1 , SphK2) activity and S1 P production in angiogenesis. Angiogenesis refers to a state in the body whereby various stimuli including growth factors promote the formation of new blood vessels. Excessive angiogenesis is commonly associated with disease progression and/or the production of undesirable effects. For instance, signaling of S1 P through the S1 P1 receptor stimulates NFKB production and promotes the formation of vascular endothelial growth factor (VEGF), COX-2, cytokines and adhesion molecules, all known contributors to
angiogenesis. In addition, SphK regulates the expression of endothelial isoforms of nitric oxide synthase which have also been implicated in modulating angiogenesis. In vivo, treatment of mice with anti-S1 P mAb blocked endothelial cell migration, capillary formation, inhibited VEGF- and FGF-induced blood vessel formation and arrested tumor- associated angiogenesis. The S1 P mAb also prevented the release of pro-angiogenic cytokines (IL-8, IL-6) from tumor cells. Thus inhibiting SphK (SphK1 , SphK2) activity and S1 P production in vascular endothelial cells can lead to reduction of angiogenesis and provide a novel strategy to treat and/or prevent disorders arising from abnormal angiogenesis. Such disorders include, but are not limited to, cancer, hemangiomas, arthritis, psoriasis, atherosclerosis, myocardial angiogenesis, diabetic retinopathy and/or macular degeneration.
Sphk (SphK1 , SphK2) and S1 P are also emerging as important mediators in
inflammatory responses and as regulators of immune cell functions. SphK1 has been shown to be activated by cytokines including tumors necrosis factor-alpha (TNF-alpha) and interleukinl -beta (IL1 -beta). The activation of SphK1 in this pathway has been shown to be, at least in part, required for mediating TNF-alpha and IL1 -beta inflammatory responses in cells, including induction of cyclo-oxygenease 2 (COX2). In addition to their role in inflammatory signaling, SphKs and S1 P have also been implicated in various immune cell functions including mast cell degranulation, migration of neutrophils, maturation and migration of lymphocytes, and T-cell development. Based on the direct involvement of S1 P in regulating specific inflammatory and autoimmune conditions, inhibiting SphK (SphK1 , SphK2) activity and reducing S1 P levels provides a novel approach for preventing, treating, managing, and/or ameliorating inflammation and autoimmune disorders. Sphingosine kinase (SphK1 , SphK2) inhibitors may be used as single agent or administered in conjunction with known immunomodulators or anti- inflammatory agents. Examples of immune disorders include, but are not restricted to ankylosing spondylitis, chronic fatigue immune dysfunction syndrome, Crohn's disease, multiple sclerosis, rheumatoid arthritis, autoimmune hepatitis, polyglandular syndromes, organ transplantation, post-surgical organ failure. Examples of inflammatory diseases include, but are not limited to, inflammatory bowel disease, asthma, allergy, septic shock, inflammatory kidney disease, post-surgical organ failure, organ transplantation, multiple sclerosis, skin inflammation and chronic inflammation as a result of viral or bacteria infections.
In addition to the above, SphK (SphK1 , SphK2) and S1 P are involved in various other pathological states such as diabetes, viral infections including HIV and Hepatitis C as well as diverse hyperproliferative disorders in addition to cancer including, but not limited to, psoriasis which is characterised by local keratinocyte hyperproliferation, mesangial disorders resulting from the abnormal proliferation of mesangial cells in the kidney as well as various vascular disorders such as atherosclerosis and restenosis both characterised by hyperproliferation of vascular smooth muscle cells. Growing evidence implicates SphK (SphK1 , SphK2) activation in the pathogenesis and/or progression of all these disorders. SphK inhibitors are expected to be of use in the prevention, therapy and/or management of these diseases. In spite of the growing interest in the sphingolipid-mediated signaling and the recognised implications thereof in various serious clinical disorders and conditions, as described above, only a few examples of inhibitors of the enzymes of this pathway have been reported. Examples of SphK1 inhibitors are provided in WO 2009/156041 and DE 10 2008/029734. These documents provide specific thiazolo-piperidine compounds. It would be useful if there were compounds available that showed improved activity, differential activity between different types of enzymes, differential activity between the sphingosine isoforms, better stability, greater ease of formulation, and an improved side- effect profile. It is an object of the present invention to address these needs in the art, to address the shortcomings of the prior art compounds, or to at least provide the public with a useful alternative.
SUMMARY OF THE INVENTION
In a first aspect the present invention provides a compound according to formula I
Figure imgf000007_0001
Formula I
wherein:
Figure imgf000007_0002
where RrR8 are independently hydrogen, lower alkyl, phenyl or benzyl and where X is O, CRR', or NR' where R and R' are independently hydrogen or lower alkyl and where for structures c and d Ri and R2 may together form an optionally lower alkyl substituted 5 or 6 membered ring and where R3 is optionally OR" where R" is alkyl or aryl;
X1 , X2, X3, X4, X5, and X6 are independently selected from nitrogen atom or carbon atom optionally substituted with halogen, oxo, cyano, amino, aryl, heteroaryl, optionally substituted lower alkoxyl and optionally substituted lower alkyl,
where if one of X1 , X2, X3, X4, X5, and X6 is an oxo-substituted carbon atom at least one of the others is a nitrogen atom;
wherein not more than three of X1 , X2, X3, X4, X5, and X6 are nitrogen atoms; and wherein Q is a heteroatom-containing alkyl, substituted alkyl, optionally substituted cycloalkyi, optionally substituted aryl, or optionally substituted heterocyclyl containing side chain bound through a heteroatom or directly through a carbon atom; and pharmaceutically acceptable salts, stereoisomers and tautomers thereof.
In preferred embodiments where substructure 'a' is present, Ri , R2, R5 and R6 are independently hydrogen, lower alkyl or lower cycloalkyi. More preferably Ri , R2, R5 and R6 are each methyl, or and R2 are both methyl and R5 and R6 are both hydrogen, or R5 and R6 are both methyl and Ri and R2 are both hydrogen. In preferred embodiments where substructure 'a' is present R3 is benzyloxy and R4 is hydrogen. In preferred embodiments where substructure 'b' is present, Ri , R2, R7 and R8 are independently hydrogen, lower alkyl or lower cycloalkyi. More preferably Ri , R2, R7 and R8 are each methyl, or R7 and R8 are both methyl and Ri and R2 are both hydrogen. In particularly preferred compounds R7 and R8 are both methyl. In these preferred embodiments, R3, R4, R5 and R6 are each hydrogen.
In the preferred embodiments discussed above where substructure 'a' or 'b' is present the preferred gem-dimethyl groups may be joined to form a cyclopropyl group or other lower cycloalkyi group. In preferred embodiments where substructure 'c' or 'd' is present, Ri , R3, and R4 are independently hydrogen, lower alkyl or lower cycloalkyi, Ri and R4 may cooperate to form a lower cycloalkyi group, and R2 is lower alkyl or lower cycloalkyi; where X is preferably O or NR' where R' is H, lower alkyl or lower cycloalkyi and where R3 and R2 may cooperate to form a 5 or 6 membered heterocyclic ring. In preferred embodiments where substructure 'c' or 'd' is present and where X is O or NR', R2 is a branched lower alkyl group. In other embodiments 'c' or 'd' is present, X is optionally substituted C=C. In other preferred embodiments X is optionally substituted C=C and R3 and R4 cooperate to form optionally substituted C=C.
In particularly preferred embodiments
Figure imgf000009_0001
is selected from substructures 'c' and 'd' wherein Ri , R2, R3, and R4 are defined as above.
In one particularly preferred embodiment
Figure imgf000009_0002
is substructure 'c' wherein Ri R2, R3, and R4 are defined as above.
In another particularly preferred embodiment
Figure imgf000009_0003
is substructure 'd' wherein Ri , R2, R3, and R4 are defined as above.
In preferred embodiments the compound is not morpholine-2-carboxylic acid [6-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)-pyridin-2-yl]-amide or a compound selected from compounds of formula (I I)
Figure imgf000009_0004
in which, in each case independently of one another:
R1 , R2, R3, R4, R5, R6, R7, R8, R9, R 0, R1 1 , R 2, R 3, R 4, R 5, R 6, R 7 represent H, D (deuterium), A, OR18, CN, F, CI and NR 8R18 ; wherein R and R2, R3 and R4, R5 and R6, R 0 and R , R2 and R 3, R 4 and R5, R 6 and R7 may also in each case together form =0 (carbonyl oxygen);
wherein R9 and R\ R and R2, R2 and R3, R3and R4, R4 and R5, R5and R6, R6 and R7, R7 and R8, R 0 and R , R and R2, R2 and R 3, R 4 and R5, R5 and R 6, R 6 and R7may also in each case together form a cycloalkyl having 3, 4, 5, 6 or 7 C atoms or Het having 3, 4, 5, 6 or 7 ring atoms,
wherein R 0 and R 9 if Y = CR 9, R and R2, R3 and R9 if Y2 = CR9, R4 and R9 if Yi = CR19, R5 and R 6, R7 and R 9 if Y2 = CR19 may also in each case together form a C=C double bond with the single bond and the C atoms to which they are attached;
R8, R 8' represent H, Dor A;
R9, R 9' represent H, D, A, OR18, NR8R18, F, CI, Br, CN or Het;
Mi, M2, M3, M4 represent CR19 or N;
Yi, Y2 represent CR19 or N;
V represents C(R9)(R19'), NR9 or is absent;
W represents [C(R 9)(R9')]PZ, CO-[C(R 9)(R9')]pZ, [C(R 9)(R9')]PN(R9)-Z, CO-N(R19) [C(R9)(R9')]pZ, N(R 9)-CO-[C(R 9)(R9')]pZ, CO-0-[C(R 9)(R9')]pZ, C(0)OR18, OR18, H or D;
wherein V, W and Y2 may also in each case together form a cycloalkyl having 3, 4, 5, 6 or 7 C atoms, in which preferably 1 , 2, 3, 4, 5, 6 or 7 H atoms can be replaced with F, CI, Br, CN and/or OH, OR19, OC(0)R19, NR9C(0)OZ, C(0)OR19, C(0)N(R9)(R19') or N(R 9)(R19'), or form Het having 3, 4, 5, 6 or 7 ring atoms, wherein Het is preferably a saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms which can be unsubstituted or singly, doubly or triply substituted with Hal, F, CI, Br, CN, A, OR18, W, SR 8, N02, N(R9)(R19'),
NR8COOZ, OCONHZ, NR8S02Z, S02N(R8)Z, S(0)mZ, COZ, CHO, COZ, =S, =NH, =NA, Oxy (-0 ) and/or =0 (carbonyl oxygen);
Z represents Het, Ar or A;
A represents unbranched or branched alkyl having 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 C atoms, in which 1 , 2, 3, 4, 5, 6 or 7 H atoms can be replaced with F, CI, Br, CN and/or OH, OR19, OC(0)R19, NR9C(0)OZ, C(0)OR19, C(0)N(R9)(R19') or N(R9)(R19');
and/or in which one or two CH2 groups can be replaced having O, S, SO, S02, CO, COO, NR18, NR 8CO, CONR 8, a cycloalkyl having 3, 4, 5, 6 or 7 C atoms, CH=CH groups and/or CH≡CH groups; or a cycloalkyl having 3, 4, 5, 6 or 7 C atoms, in which preferably 1 , 2, 3, 4, 5, 6 or 7 H atoms can be replaced with F, CI, Br, CN and/or OH, OR19, OC(0)R19, NR 9C(0)OZ, C(0)OR19, C(0)N(R 9)(R19') or N(R 9)(R19');
Ar represents phenyl, naphthyl or biphenyl substituted singly, doubly or triply with Hal, F, CI, Br, CN, A, OR18, W, SR 8, N02, N(R 9)(R19'), NR 8COOZ, OCONHZ, NR 8S02Z,
S02N(R 8)Z, S(0)mZ, COZ, CHO, COZ;
Het represents, in each case independently of one another, a mononuclear, binuclear or trinuclear saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms which can be unsubstituted or singly, doubly or triply substituted with Hal, F, CI, Br, CN, A, OR18, W, SR18, N02, N(R 9)(R19'), NR 8COOZ, OCONHZ, NR 8S02Z,
S02N(R 8)Z, S(0)mZ, COZ, CHO, COZ, =S, =NH, =NA, Oxy (-0 ) and/or =0 (carbonyl oxygen);
m is1 , 2 or 3,
n, o are 0, 1 or 2,
p is 0, 1 , 2, 3 or 4
with the proviso compounds of the formula (I) in which
(a) V is absent, and
(b) is W = C(0)-CH2-Het
are excluded;
In preferred embodiments the compound is not a compound selected from the group: morpholine-2-carboxylic acid [6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)- pyridin-2-yl]-amide
1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)phenyl)piperazine
1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4-amine 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) piperazine 1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) piperazine 1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperidin-4- amine
2-(piperazin-1 -yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl) pyrimidine
1 - (2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperidin-4- amine
4-(piperazin-1 -yl)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine
2- (piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrazine 1 -(5-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyridin-3-yl)piperazine 1 -(5-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyridin-3-yl)piperidin-4-am 3-(4-(6-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperazin-1 - yl)propan-1 -ol.
In one embodiment Q is not a heteroatom-containing optionally substituted aryl.
In certain preferred embodiments one or more of X1 , X2, X3, X4, X5, and X6 is an oxo- substituted carbon atom, methyl substituted carbon atom, or fluorine substituted carbon atom.
In particularly preferred embodiments one or more of X1, X2, X3, X4, X5, and X6 is a carbon atom or nitrogen atom. In particularly preferred embodiments X1 , X2, X3, X4, X5, and X6 are carbon atoms or nitrogen atoms.
Preferably the ring formed by X1 , X2, X3, X4, X5, and X6 is aromatic or doubly unsaturated, more preferably the ring is aromatic.
In preferred embodiments the groups X1 , X2, X3, X4, X5, and X6 are selected so as to form, together with the atoms to which they are bonded, the following ring structures:
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000013_0002
where Q is as herein defined and wherein the resulting rings may be additionally substituted in accordance with the selections for groups X1 , X2, X3, X4, X5, and X6. Preferred are the compounds having the above rings with no additional substitution.
In particularly preferred embodiments the groups X1 , X2, X3, X4, X5, and X6 are selected so as to form, together with the atoms to which they are bonded, a pyrimidine ring. Without wishing to be bound by theory, the Inventors have found that surprisingly the presence of a pyrimidine group at this position combines good inhibition activity with improved physical properties of the compounds of the invention, such as their solubility, and that certain compounds having the pyrimidine ring present therefore have good pharmacokinetic properties, and are preferred. In preferred embodiments at least one of the heteroatoms in Q is a nitrogen atom.
In preferred embodiments Q contains at least two heteroatoms, more preferably two nitrogen atoms. In preferred embodiments Q may be selected from:
Figure imgf000014_0001
Figure imgf000014_0002
Figure imgf000014_0003
Where R10- 14 are selected from hydrogen or optionally substituted lower alkyl, or may combine with other R groups in Q to form optionally substituted cycloalkyl or optionally substituted heterocycloalkyl rings which may be aromatic or non-aromatic.
In preferred embodiments R 0 may, together with a nitrogen atom to which it is bound, form a guanidino group. In other preferred embodiments R 0 may be a double bonded nitrogen atom and any other Rx moiety bound to the same atom is absent.
In some embodiments one or more aromatic carbon atoms of the benzylamino group may be replaced with N, for example to give those two pyridines shown in the structures above. Where two aromatic carbon atoms are replaced by nitrogen they are preferably arranged to give a pyridazine ring, although pyrimidine and pyrazine arrangements are also available in certain embodiments of the invention. Particulary preferred are compounds of the invention having optionally substituted guanidino or amidino moiety containing Q groups. In particularly preferred embodiments the compounds of the invention have a guanidine or amidino group forming part of the side-chain 'Q' moiety. In certain other embodiments the guanidine or amidino group forming part of the side-chain 'Q' moiety may be optionally substituted, preferably with cyano, OH or OR" where R" is optionally substituted lower alkyl.
In preferred embodiments the moiety Q is selected from the following structures:
Figure imgf000015_0001
Figure imgf000015_0002
Figure imgf000015_0003
Figure imgf000015_0004
Figure imgf000016_0001
or structural isomers thereof, optionally additionally substituted with, for example, hydroxyl, amino, halogen, lower alkyi, lower alkoxy and cyano. Preferred are the compounds having the above Q groups, more preferably with no additional substitution.
The term "alkyi group" refers to an aliphatic group containing at least carbon and hydrogen and containing 1 to 15 carbon atoms, such as 1 to 10 carbon atoms.
Attachment to the alkyi group occurs through a carbon atom.
A "Cn alkyi" group refers to an aliphatic group containing n carbon atoms. For example, a C1 -C10 alkyi group contains 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 carbon atoms.
An alkyi group may be straight chained or it may be branched.
An alkyi group may contain no ring structures or it may contain one or more rings.
In preferred embodiments the alkyi group is a lower alkyi group, that is one having between 1 and 6 carbon atoms whether branched or straight chain. For example, a "cycloalkyi" group contains at least one ring. It is understood that attachment to a cycloalkyi group is via a ring of the cycloalkyi group. Each ring may contain 3 to 10 atoms, such as 4 to 8 or 5 to 7 atoms. Each ring may be independently selected to contain just carbon atoms or to contain both carbon atoms and from 1 to 4 heteroatoms selected from O, N and S. For cyclo-heteroalkyl groups (i.e. cycloalkyi groups that contain one or more heteroatoms), attachment to the cycloalkyi group may occur either through a carbon atom or, if one or more heteroatoms are contained in a ring, attachment may also occur through a heteroatom contained in a ring. For example, a cycloalkyi group may be mono-cyclic or bi-cyclic.
Thus, a "Cn cycloalkyi" group contains n carbon atoms. All n carbon atoms may be contained in the ring(s) of the cycloalkyi group or one or more of the carbons may not be contained in the ring(s) and may instead form one or more chains branching from the ring.
If a Cn alkyl group is joined to a separate Cm alkyl group containing m carbon atoms to form, for example, a heterocycle, the two alkyl groups contain a total number of m + n carbon atoms.
An alkyl group may be saturated or unsaturated. Thus, the alkyl group may be an alkenyl group (i.e. contain a carbon-carbon double bond) and / or an alkynyl group (i.e. contain a carbon-carbon triple bond). If the alkyl group is unsaturated, it may contain at least 2 carbon atoms. It is understood that any unsaturated portions of an alkyl group are non-aromatic (aromatic groups fall within the scope of the definition of "aryl'). Any part of the alkyl group may be unsaturated, for example the straight, branched or cyclic portion of an alkyl group may contain a carbon-carbon double bond or a carbon-carbon triple bond. Attachment to an unsaturated alky group may occur through the unsaturated part of the alkyl group or may occur through the unsaturated part of the group.
For example, an unsaturated alkyl group may contain 1 to 4 carbon-carbon double bonds or 1 to 3 carbon-carbon triple bonds or 1 to 4 of a combination of carbon-carbon double bonds and carbon-carbon triple bonds. An alkyl group may be substituted with one or more heteroatoms or it may be
unsubstituted (i.e. not contain any heteroatoms). If more than one hetero-substituent is present, the substituents are independently selected from one another unless they form a part of a particular functional group (e.g. an amide, amidino, or guanidino group).
The heteroatom substituents may in turn be substituted with further carbon-containing groups. In this case, the Cn or Cm prefix that defines the substituted alkyl group refers to the total number of carbons contained in the group, i.e. including the carbon atoms contained in any substituted heteroatomic groups, and the total alkyl group contains 1 to 15 carbon atoms as defined previously.
Accordingly, if the alkyl group is substituted, it may, for example, contain one or more of CN, C02H, CONH2, CONHR, CONRaRb, C02R, NH2, NHR, NRaRb, OH, OR, SH, SR, F, CI, Br and I, wherein each R, Ra and Rb are independently selected groups (e.g. alkyl / aryl groups) attached to the atom to which the group joins through a carbon atom of each group, including wherein Ra and Rb form a heterocycle that includes the heteroatom to which they are attached. A group containing two Cm-Cn alkyl moieties that form a cycle that includes, for example, the heteroatom to which they are attached may contain from C2m to C2n carbon atoms.
Examples of unsubstituted saturated alkyl groups containing no cyclic structures include methyl, ethyl, n-propyl, sec-propyl, n-butyl, sec-butyl, te/t-butyl, pentyl (branched or unbranched), hexyl (branched or unbranched), heptyl (branched or unbranched), octyl (branched or unbranched), nonyl (branched or unbranched), and decyl (branched or unbranched).
Examples of unsubstitued saturated cyclic alkyl groups include cyclopropyl, cylcobutyl, cyclopentyl and cyclohexyl. Examples of unsaturated alkyl groups include ethenyl, propenyl, butenyl, 2-methybutenyl and cyclohexenyl.
The term "aryl group" refers to a group containing at least one ring that is aromatic.
Where an aryl group is stated as being substituted at a particular position, attachment of the position to the aryl group is onto the aromatic ring of the aryl group itself rather than the position being joined to the aryl group through any non-aromatic side-chain of the aryl group. For example, when R is an aryl group in CR , the C is attached to the aromatic part of the aryl group. Each ring may be independently selected to contain only carbon atoms or to contain both carbon atoms and from 1 to 4 heteroatoms selected from O, N and S. For heteroaryl groups (i.e. aryl groups that contain one or more heteroatoms), attachment to the aryl group may occur either through a carbon atom or, if one or more heteroatoms are contained in a ring, attachment may also occur through a heteroatom contained in a ring.
It is noted that the heteroatoms contained in a ring of a heteroaryl group may be substituted, for example forming an /V-oxide.
For example, the aromatic group may be mono-cyclic or bi-cyclic, wherein one or both of the rings of a bi-cyclic system is aromatic.
Examples of aryl groups include acridinyl, phenyl, carbazolyl, cinnolinyl, quinoxalinyl, pyrrazolyl, benzotriazolyl, furanyl, naphthyl, thienyl, thiazolyl, benzothienyl, benzofuranyl, quinolinyl, isoquinolinyl, oxazolyl, isoxazolyl, indolyl, pyrazinyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, tetrahydroquinolinyl, benzimidazolyl and melaminyl.
It is noted that the term "heterocycle" includes within its scope both cycloalkyl groups containing one or more heteroatoms within the ring system and aryl groups containing one or more heteroatoms within the ring system.
The term "halo" refers to a group selected from chlorine, fluorine, bromine and iodine.
For completeness, it is also noted that certain chemical formulae used herein define delocalized systems. This definition is known in the art as a definition of aromaticity and may indicate the presence of, for example, a mono-, di- or tri-cyclic system that contains (4n+2) electrons where n is an integer. In other words, these systems may display Huckel aromaticity.
Each tautomeric form is intended to fall within the scope of the invention. Where the compound of the invention may have one or more chiral centres, each enantiomer or diastereoisomer is intended to fall within the scope of the invention.
Additionally, where one enantiomer or diasterioisomer has more desirable characteristics than the related molecule(s), the one having desirable characteristics is preferred.
In addition, the compounds of the present invention may be provided as a pro-drug. Prodrugs are transformed, generally in vivo, from one form to the active forms of the drugs described herein. For example, a prodrug may be formed by protecting an amine as a physiological hydrolyzable amide.
In addition, the compounds of the present invention may be provided in the form of their pharmaceutically acceptable salts or as co-crystals. For example, the compounds may be provided having protonated amine groups. The term "pharmaceutically acceptable salt" refers to ionic compounds formed by the addition of an acid to a base. The term refers to such salts that are considered in the art as being suitable for use in contact with a patient, for example in vivo and
pharmaceutically acceptable salts are generally chosen for their non-toxic, non-irritant characteristics.
The term "co-crystal" refers to a multi- component molecular crystal, which may comprise non-ionic interactions.
Pharmaceutically acceptable salts and co-crystals may be prepared by ion exchange chromatography or by reacting the free base or acidic form of a compound with stoichiometric amounts or with an excess of the desired salt-forming inorganic or organic acid or base in one or more suitable solvents.
Salts known in the art to be generally suitable for use in contact with a patient include salts derived from inorganic and / or organic acids, including the hydrobromide, hydrochloride, sulphate, bisulphate, nitrate, acetate, oxalate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate and tartrate. These may include cations based on the alkali and alkaline earth metals, such as sodium, potassium, calcium and magnesium, as well as ammonium,
tetramethylammonium, tetraethylammonium. Further reference is made to the number of literature sources that survey suitable pharmaceutically acceptable salts, for example the Handbook of pharmaceutical salts published by lUPAC.
In addition, the compounds of the present invention may sometimes exist as zwitterions, which are considered as part of the invention.
The compounds of the present invention are useful in the treatment of medical conditions associated with undesirable SphK activity and S1 P homeostasis and to treat a wide range of diseases where SphK acitivity and S1 P has been shown to play a key role including cancers, hyperproliferative, inflammatory, angiogenic, immune and viral infectious diseases.
In a further aspect the present invention provides compounds according to the invention for the treatment of a disease or disorder involving associated with SphK activity and S1 P homeostasis. In one preferred embodiment the present invention provides the use of those compounds of the invention having selectivity for Sphkl over Sphk2 for the treatment of a disease or disorder involving or associated with SphK1 activity and S1 P homeostasis. In another preferred embodiment the present invention provides the use of those compounds of the invention having selectivity for Sphk2 over Sphkl for the treatment of a disease or disorder involving or associated with SphK2 activity and S1 P homeostasis.
In particularly preferred embodiments the compounds of the invention are for the treatment of cancers including benign, pre-malignant, metastatic and malignant cancers. The cancers to be treated may include solid tumors as well as hematopoietic cancers. In preferred embodiments the compounds of the invention are for the treatment of solid cancers.
The Inventors have found that surprisingly certain of the compounds developed in the work detailed herein demonstrate an effect not only in inhibiting Sphkl but also a marked inhibition of Sphk2. Such 'dual active' compounds are preferred compounds of the invention and preferred compounds in this regard are detailed below as Examples 39, 41 , 47, 90 and 96. Some compounds of the invention in general show Sphk2 activity and these compounds are also preferred in certain embodiments of the invention. With regard to combination therapy the compounds of the present invention may be administered separately, sequentially, simultaneously, concurrently or may be chronologically staggered with one or more standard therapeutics..
The present invention also provides a pharmaceutical composition suitable for clinical use.
In particular, a pharmaceutical composition may comprise a pharmaceutical carrier and, dispersed therein, a therapeutically effective amount of the compounds of the invention. The composition may be solid or liquid. The pharmaceutical carrier is generally chosen based on the type of administration being used and the pharmaceutical carrier may for example be solid or liquid. The compounds of the invention may be in the same phase or in a different phase than the pharmaceutical carrier.
Pharmaceutical compositions may be formulated according to their particular use and purpose by mixing, for example, excipient, binding agent, lubricant, disintegrating agent, coating material, emulsifier, suspending agent, solvent, stabilizer, absorption enhancer and / or ointment base. The composition may be suitable for oral, injectable, rectal or topical administration.
For example, the pharmaceutical composition may be administered orally, such as in the form of tablets, coated tablets, hard or soft gelatine capsules, solutions, emulsions, or suspensions. Administration can also be carried out rectally, for example using suppositories, locally or percutaneously, for example using ointments, creams, gels or solution, or parenterally, for example using injectable solutions.
For the preparation of tablets, coated tablets or hard gelatine capsules, the compounds of the present invention may be admixed with pharmaceutically inert, inorganic or organic excipients. Examples of suitable excipients include lactose, mize starch or derivatives thereof, talc or stearic acid or salts thereof. Suitable excipients for use with soft gelatine capsules include, for example, vegetable oils, waxes, fats and semi-solid or liquid polyols. For the preparation of solutions and syrups, excipients include, for example, water, polyols, saccharose, invert sugar and glucose. For injectable solutions, excipients include, for example, water, alcohols, polyols, glycerine and vegetable oil. For suppositories and for local and percutaneous application, excipients include, for example, natural or hardened oils, waxes, fats and semi-solid or liquid polyols.
The pharmaceutical compositions may also contain preserving agents, solublizing agents, stabilizing agents, wetting agents, emulsifiers, sweeteners, colorants, odorants, buffers, coating agents and / or antioxidants.
For combination therapies, the second drug may be provided in pharmaceutical composition with the present invention or may be provided separately. Thus, a pharmaceutical formulation for oral administration may, for example, be granule, tablet, sugar coated tablet, capsule, pill, suspension or emulsion. For parenteral injection for, for example, intravenous, intramuscular or subcutaneous use, a sterile aqueous solution may be provided that may contain other substances including, for example, salts and / or glucose to make to solution isotonic. The therapeutic agent may also be administered in the form of a suppository or pessary, or may be applied topically in the form of a lotion, solution, cream, ointment or dusting powder.
In certain laboratory and clinical settings it is useful to have selectivity between 2 or more subtypes of a particular target protein. In a situation where, for example, different subtypes of an enzyme are made by a cell in response to different conditions or stimuli, the preferential inhibition of one subtype can leave a pathway involving other subtypes within the cell relatively unaffected by treatment. Given this advantage, although without wishing to be bound by theory, the Inventors consider those compounds of the invention showing selectivity between Sphkl and Sphk2 to be particularly advantageous and therefore preferred. Particularly preferred are those showing a selectivity of
Sphkl /Sphk2 of greater than 10, ie the activity in inhibiting Sphkl is 10-fold greater than the inhibition of Sphk2. Especially preferred are those compounds where the selectivity is much higher than 10, eg 50 or higher. Also preferred are those compounds that demonstrate selectivity for Sphk2 over Sphkl of around 10 times or greater. In another embodiment the present invention provides the use of the preferred compounds having selectivity as described herein for inhibiting Sphkl or Sphk2. In one preferred embodiment these enzymes are inhibited by the use of a compound of the invention having specificity for that subtype such that the other subtype is relatively less inhibited.
The present invention will now be further described. In the following passages different aspects of the invention are defined in more detail. Each aspect so defined may be combined with any other aspect or aspects unless clearly indicated to the contrary. In particular, any feature indicated as being preferred or advantageous may be combined with any other feature or features indicated as being preferred or advantageous. EXAMPLES
The present invention will now be described in relation to several examples.
The following exemplary compounds were synthesised according to the methods described. Their IC50 values were then determined as described below and are represented in the following table, in which the compound numbers correspond to the numbers in the examples.
Example
Number IC50 in SphK1 IC50 in SphK2
1 +++ +
2 ++ +
3 ++ +
4 ++ +
5 ++ +
6 ++ +
7 ++ +
8 ++ +
9 ++ +
10 +++ +
1 1 ++ +
12 ++ +
13 ++ + 14 +++ +
15 ++ +
16 +++ +
17 ++ +
18 ++ +
19 + +
20 + +
21 +++ +
22 +++ +
23 + +
24 ++ +
25 ++ +
26 +++ +
27 ++ +
28 + +
29 + +
30 +++ +
31 ++ +
32 ++ +
33 ++ +
34 ++ +
35 ++ +
36 ++ +
37 +++ +
38 +++ +
39 ++ ++
40 ++ +
41 ++ ++
42 ++ +
43 + +
44 ++ +
45 ++ + 46 +++ +
47 ++ ++
48 + +
49 ++ +
50 ++ +
51 ++ +
52 ++ +
53 + n.d.
54 + n.d.
55 + n.d.
56 ++ n.d.
57 ++ n.d.
58 + n.d.
59 + n.d.
60 ++ n.d.
61 + n.d.
62 + n.d.
63 ++ n.d.
64 ++ +
65 ++ +
66 +++ +
67 ++ +
68 ++ +
69 + n.d.
70 + n.d.
71 ++ n.d.
72 ++ n.d.
73 ++ n.d.
74 + n.d.
75 + n.d.
76 + +
77 ++ + 78 ++ n.d.
79 ++ n.d.
80 + n.d.
81 + n.d.
82 + n.d.
83 ++ +
84 ++ +
85 + n.d.
86 + +
87 ++ +
88 ++ +
89 ++ +
90 +++ ++
91 ++ +
92 +++ +
93 +++ n.d.
94 +++ n.d.
95 + +
96 +++ ++
97 +++ n.d.
98 ++ n.d.
99 ++ n.d.
100 + n.d.
101 ++ n.d.
102 ++ n.d.
103 ++ n.d.
104 ++ +
105 ++ +
106 ++ +
107 ++ +
108 ++ + Key
+ Ιΰ50 > 10μΜ
++ 1 μΜ < IC50 < 10μΜ
Figure imgf000028_0001
Abbreviations
AcOH: Acetic acid; nBuLi: n-Butyllithium; BINAP: 2,2'-bis(diphenylphosphino)-1 ,1 '- binaphthyl; COMU: (1 -Cyano-2-ethoxy-2-oxoethylidenaminooxy)dimethylamino- morpholino-carbenium hexafluorophosphate; DBU: 1 ,8-Diazabicyclo[5.4.0]undec-7-ene;
DCM: Dichloromethane; DIPEA: Diisopropylethylamine; DMA: N,N- Dim ethyl acetamide;
DMAP: 4-Dimethylaminopyridine; DME: Dimethyl ether; DMF: /V,/V-Dimethylformamide;
DMSO: Dimethylsulfoxide; EDCI: 1 -Ethyl-3-(3'-dimethylaminopropyl)carbodiimide;
EtOAc: Ethyl acetate; h:Hour: HATU: 0-(7-Azabenzotriazol-1 -yl)-A/,/V,/V',/V- tetramethyluronium hexafluorophosphate; HCI: Hydrochloric acid; HOBt: 1 -
Hydroxybenzotriazole; HPLC: High Pressure Liquid Chromatography; IMS: Industrial methylated spirits: M: Molar; MeOH: Methanol; Min: Minutes; NMP: /V-Methyl-2- pyrrolidone; NMR: Nuclear Magnetic Resonance; Oxyma: Ethyl
(hydroxyimino)cyanoacetate;; RT: Room temperature; SCX: SCX- strong cation exchange; TBAF: Tetra-n-butylammonium fluoride; TEA: Triethylamine; TFA:
Trifluoroacetic acid; THF: Tetrahydrofuran; TMSCI: Trimethylsilyl chloride
General Experimental Conditions
H NMR spectra were recorded at ambient temperature using a Varian Unity Inova (400MHz) spectrometer with a triple resonance 5mm probe, a Bruker Avance DRX (400MHz) with a 5mm inverse detection triple resonance TXI probe, a Bruker Avance (500MHz) spectrometer with a 5mm QNP probe or a Bruker Avance DPX (300MHz) spectrometer with a standard 5mm dual frequency probe. Chemical shifts are expressed in ppm relative to tetramethylsilane.
Preparative HPLC
Method A: The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler. The spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered. A purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridge™ Prep C18 5μηι OBD™ 19 x 50mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide.
Gradient - Time Flow ml/min %A %B
0.00 20.0 50 50
3.00 20 12 88
5.00 20.0 12 88
7.00 20.0 0 100
8.0 20.0 0 100
8.20 20.0 50 50
Detection - MS, UV
MS ionisation method - Multimode (positive and negative ion)
Total experiment time - 10 mins (approx)
Method B: The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler. The spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered. A purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridge™ Prep C18 5μηι OBD™ 19 x 50mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide.
Gradient - Time Flow ml/min %A %B
0.00 20.0 95 5
7.00 20.0 0 100
8.00 20.0 0 100
8.20 20.0 95 5 Detection - MS, UV
MS ionisation method - Multimode (positive and negative ion)
Total experiment time - 10 mins (approx)
Method C: The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler. The spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered. A purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridge™ Prep C18 5μηι OBD™ 30 x 100mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide.
Gradient - Time Flow ml/min %A %B
0.00 60.0 95 5
14.00 60.0 0 100
16.00 60.0 0 100
16.50 60.0 95 5
Detection - MS, UV
MS ionisation method - Multimode (positive and negative ion)
Total experiment time - 18 mins (approx) Method D: The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler. The spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered. A purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridge™ Prep C18 5μηι OBD™ 30 x 100mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide. Gradient - Time Flow ml/min %A %B
0.00 60.0 68 32
5.00 60.0 59 41
9.00 60.0 59 41
14.00 60.0 0 100
16.00 60.0 0 100
16.50 60.0 68 32
Detection - MS, UV
MS ionisation method - Multimode (positive and negative ion)
Total experiment time - 18 mins (approx)
Method E: The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler. The spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered. A purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridge™ Prep C18 5μηι OBD™ 30 x 100mm column at room temperature. Mobile phase - A) Water 0.1 % ammonium hydroxide B) Acetonitrile/Water 95: 5 with 0.1 % ammonium hydroxide.
Gradient - Time Flow ml/min %A %B
0.00 60.0 61 39
5.00 60.0 34 66
9.00 60.0 34 66
14.00 60.0 0 100
16.00 60.0 0 100
16.50 60.0 61 39
Detection - MS, UV
MS ionisation method - Multimode (positive and negative
Total experiment time - 18 mins (approx) Method H: The system consists of a Agilent Technologies 6120 single quadrupole mass spectrometer linked to a Agilent Technologies 1200 Preparative LC system with Multiple Wavelength detector and autosampler. The spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. Fraction collection is mass-triggered. A purification experiment is performed on each sample submitted using the following conditions: LC Column - Waters XBridge™ Prep C18 5μηι OBD™ 19 x 50mm column at room temperature. Mobile phase - A) Water 0.1 % formic acid B) Acetonitrile/Water 95: 5 with 0.1 % formic acid.
Gradient - Time Flow ml/min %A %B
0.00 20.0 95 5
7.00 20.0 0 100
9.00 20.0 0 100
9.20 20.0 95 5
Detection - MS, UV
MS ionisation method - Multimode (positive and negative ion)
Total experiment time - 10 mins (approx)
High Pressure Liquid Chromatography Mass Spectrometry (LCMS) experiments to determine retention times (RT) and associated mass ions were performed using one of the following methods.
Method F: The system consists of an Agilent Technologies 6140 single quadrupole mass spectrometer linked to an Agilent Technologies 1290 Infinity LC system with UV diode array detector and autosampler. The spectrometer has a multimode ionization source (electrospray and atmospheric pressure chemical ionizations) operating in positive and negative ion mode. An LCMS experiment is performed on each sample submitted using the following conditions: LC Column - Zorbax Eclipse Plus C18 RRHD 1 .8micron 50 x 2.1 mm maintained at 40^. Mobile phase - A) Water 0.1 % Formic Acid B) Acetonitrile 0.1 % Formic Acid. Gradient - Time Flow ml/min %A %B
0.00 1 .0 95 5
1 .80 1 .0 0 100
2.20 1 .0 0 100
2.21 1 .0 95 5
2.50 1 .0 95 5
Method G/ The system consists of ThermoFinnigan LCQ Advantage Mass Spectrometer with the Surveyor LC system and 200 position autosampler. The spectrometer has an electrospray source operating in positive and negative ion mode. An LC-MS experiment is performed on each sample submitted using the following conditions: LC Column - Luna 3micron Ci8 50 x 2 mm; Mobile phase -A) Water 0.1 % Formic Acid B) Acetonitrile 0.1 % Formic Acid
Gradient - Time Flow ml/min %A %B
0.00 0.6 95 5
7.00 0.6 5 95
8.00 0.6 5 95
8.20 0.6 95 5
1 1 .00 0.6 95 5
Split - Ι ΟΟμΙ/min split to the ESI source with inline Surveyor DAD detection.
Detection - MS, UV
MS ionisation method - Electrospray (positive and negative ion)
Total experiment time - 1 1 mins (approx)
Unless quoted otherwise, microwave experiments were carried out using a Biotage Initiator™ Sixty, which uses a single-mode resonator and dynamic field tuning, both of which give reproducibility and control. Temperature from 60-250 °C can be achieved, and pressures of up to 20bar can be reached.
Unless otherwise indicated, the nomenclature of structures was assigned using
ChemDraw Ultra 12.0 (CambridgeSoft). Names highlighted by a superscript asterisk were named using "structure=name" from ChemBioDraw 1 1 (CambridgeSoft). Sphingosine Kinase Assay testing
Sphingosine kinase 1 (SPHK1 ) or 2 (SPHK2) were purchased from Invitrogen (lot number 417324B and 683074A respectively). Unless otherwise indicated, all other reagents were purchased from Sigma. The kinase reaction was performed in white polystyrene 384-well plates in 1 x reaction buffer containing 40 mM Tris (pH 7.5), 20 mM MgCI2, 0.1 img/mL BSA and 1 mM DTT. SPHK1 (0.26 units) or SPHK2 (0.53 units) (5μΙ) were incubated in the presence or absence of the appropriate concentration of the inhibitor (5μΙ) for 30min at room temperature. Inhibitors were dissolved in a 10 mM DMSO stock solution and serially diluted prior the assay to give a fourteen point half log dose response. The Kinase reaction was then initiated by the addition of Ultrapure ATP (20μΜ) (Promega) and Sphingosine (75μΜ) (Echelon) and allowed to incubate at room temperature for 1 hour (total reaction volume of 15μΙ). Sphingosine kinase inhibition was subsequently determined using the ADP-Glo™ Kinase Assay as described by the manufacturer (Promega). Curve fitting was performed using the GraphPad Prism software and IC5o values determined using the non-linear regression curve fitting model. IC50 values for the inhibitors of the invention represent the average of at least duplicate experiments.
Example 1 : 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDphenvDpiperazine di(trifluoroacetate)
Figure imgf000034_0001
Step 1: 6-(3-bromophenyl)- 1 , 1 ,4,4-tetramethyl-1 ,2,3,4-tetrahydronaphthalene:
To a solution of 1 ,3-dibromobenzene (0.3 g, 1 .272 mmol) in DMF (10 ml) in a sealable vial was added 4,4,5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen- 2-yl)-1 ,3,2-dioxaborolane (0.400 g, 1 .272 mmol) and 2M (aq.) sodium carbonate (2.54 ml, 5.09 mmol). Nitrogen was bubbled through the suspension and
[1 ,1 '-Bis(diphenylphosphino)ferrocene]dichloropalladium(ll) CH2Cl2 (0.052 g, 0.064 mmol) added. The resulting mixture was heated for 16 hours at 80 °C, cooled to room temperature, poured onto brine (100 ml), extracted with ethyl acetate, dried over magnesium sulphate, filtered and concentrated in vacuo to afford the title compound as a brown oil, which was used without further purification. LCMS (Method F): RT = 2.06 min, M+H+ = 344.
Step 2: tert-butyl 4-(3-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine- 1 -carboxylate:
To a solution of 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4-tetrahydronaphthalene (215mg, 0.626 mmol) in dioxane (5 ml) in a sealable vial, was added te/t-butyl piperazine-1 -carboxylate (1 17 mg, 0.626 mmol), cesium carbonate (490 mg, 1 .503 mmol) and (9,9-dimethyl-9H-xanthene-4,5-diyl)bis(diphenylphosphine) (27.2 mg, 0.047 mmol). Nitrogen was bubbled through the suspension and
tris(dibenzylideneacetone)dipalladium(0) (14.34 mg, 0.016 mmol) added. The resulting mixture was heated for 16 hours at 100 °C, cooled to room temperature, poured onto brine (100 ml), extracted with ethyl acetate, dried over magnesium sulphate, filtered and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 50 % ethyl acetate in cyclohexane) to afford the title compound as a yellow oil (52 mg, 18%).
LCMS (Method G): RT = 9.71 min, M+H+ = 449; 1H NMR (500 MHz, CDCI3): 7.41 (1 H, d), 7.28 (1 H, d), 7.23-7.25 (2H, m), 7.01 -7.02 (2H, m), 6.82 (1 H, ddd), 3.52 (4H, dd), 3.1 1 (4H, dd), 1 .64 (4H, s), 1 .41 (9H, s), 1 .26 (6H, s), 1 .24 (6H, s).
Step 3: 1 -(3-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)phenyl)piperazine di( trifluoroacetate):
To a solution of te/t-butyl 4-(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine-1 -carboxylate (51 mg, 0.1 1 mmol) in dichloromethane (1 ml_) was added trifluoroacetic acid (1 ml). The yellow solution was stirred at room temperature for 5 minutes before being concentrated in vacuo. The resulting solid was triturated in diethyl ether to give the salt of the title compound as a yellow solid (21 mg, 53%).
LCMS (Method F): RT = 1 .13 min, M+H+ = 349; 1H NMR (500 MHz, MeOH-d4): 7.50 (1 H, d), 7.39 (1 H, d), 7.32-7.34 (2H, m), 7.18-7.20 (2H, m), 7.01 (1 H, dd), 3.47-3.49 (4H, m), 3.40-3.43 (4H, m), 1 .78 (4H, s), 1 .35 (6H, s), 1 .33 (6H, s).
Example 2: 1 -(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-4- yl)piperidin-4-amine di(trifluoroacetate)
Figure imgf000036_0001
Step 1: 1-(2-bromopyridin-4-yl)piperidin-4-amine:
To a solution of 2-chloro-4-iodopyridine (0.5 g, 2.088 mmol) in dioxane (15 ml) in a sealable vial, was added terf-butyl piperidin-4-ylcarbamate (0.418 g, 2.088 mmol), cesium carbonate (1 .633 g, 5.01 mmol) and (9,9-dimethyl-9H-xanthene-4,5- diyl)bis(diphenylphosphine) (0.091 g, 0.157 mmol). Nitrogen was bubbled through the suspension and tris(dibenzylideneacetone)dipalladium(0) (48.1 mg, 0.052 mmol) added. The reaction mixture was heated at 100 °C for 18 h, cooled to room temperature, poured onto brine (100 ml), extracted with ethyl acetate, dried over magnesium sulphate, filtered and concentrated in vacuo to afford the title compound as a clear oil, which was used without further purification.
LCMS (Method G): RT = 4.05 min, M+H+ = 312. Step 2: tert-butyl (1-(2-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-4- yl)piperidin -4 -yl) carbamate:
To a solution of 1 -(2-bromopyridin-4-yl)piperidin-4-amine (325 mg, 1 .042 mmol) in DMF (8 ml) in a sealable vial, was added 4,4, 5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (328 mg, 1 .042 mmol) and 2M (aq.) sodium carbonate (2.085 ml, 4.17 mmol). Nitrogen was bubbled through the suspension and [1 ,1 ' Bis(diphenylphosphino)ferrocene]dichloropalladium(ll) CH2CI2 (0.052 g, 0.064 mmol) added. The resulting mixture was heated for 16 hours at 80 °C, cooled to room temperature, poured onto brine (100 ml), extracted with ethyl acetate, dried over magnesium sulphate, filtered and concentrated in vacuo to afford the title compound as a clear oil (105 mg, 28%).
LCMS (Method F): RT = 1 .91 min, M+H+ = 464; 1H NMR (500 MHz, CDCI3): 8.27 (1 H, d), 7.78 (1 H, d), 7.68 (1 H, d), 7.53 (1 H, d), 6.88 (1 H, s), 6.54 (1 H, dd), 4.42 (1 H, s), 3.81 (2H, d), 3.63 (1 H, s), 2.93-2.95 (3H, m), 1 .70 (2H, m), 1 .64 (4H, s) 1 .48 (9H, s). 1 .23 (6H, s). 1 .19 (6H, s).
Step 3: 1-(2-(5,5,8,8-tetramethyl-5fi ,8-tetrahydronaphthalen-2-yl)pyridin-4-yl)pfa 4-amine di(trifluoroacetate): Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), te/t-butyl (1 -(2-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-4-yl)piperidin-4-yl)carbamate (105 mg, 0.23 mmol) was reacted to afford the title compound as a white solid (68 mg, 83%).
LCMS (Method F): RT = 0.76 min, M+H+ = 364; 1H NMR (500 MHz, MeOH-d4): 8.16 (1 H, d), 7.77 (1 H, s), 7.60 (2H, s), 7.35 (1 H, d), 7.23 (1 H, dd), 4.49 (2H, d), 3.57-3.60 (1 H, m), 3.33 (2H, d), 2.26 (2H, d), 1 .78 (4H, s), 1 .74 (2H, d), 1 .39 (6H, s), 1 .35 (6H, s).
Example 3: 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-4- vDpiperazine di(trifluoroacetate)
Figure imgf000037_0001
Step 1: tert-butyl 4-(2-chloropyridin-4-yl)piperazine-1-carboxylate:
To a solution of 2-chloro-4-iodopyridine (500 mg, 2.088 mmol) in toluene (15 ml) in a sealable vial, was added te/t-butyl piperazine-1 -carboxylate (389 mg, 2.088 mmol) and sodium te/t-butoxide (281 mg, 2.92 mmol). Nitrogen was bubbled through the suspension and [1 ,1 ' Bis(diphenylphosphino)ferrocene]dichloropalladium(ll) CH2CI2 (0.052 g, 0.064 mmol) added. The resulting mixture was heated for 18 hours at 100 'C, cooled to room temperature, poured onto brine (100 ml), extracted with ethyl acetate, dried over magnesium sulphate, filtered and concentrated in vacuo to afford the title compound as a clear oil, which was used without further purification.
LCMS (Method F): RT = 1 .01 min, M+H+ = 298. Step 2: tert-butyl 4-(3-(5, 5, 8, 8-tetramethyl-5, 6, 7, 8-tetrahydronaphthalen-2-yl)pyridin-4- yl)piperazine- 1 -carboxylate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, 1 -(2-chloropyridin-4-yl)piperidin-4-amine (31 1 mg, 1 .044 mmol) and 4,4,5, 5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 , 3,2- dioxaborolane (328 mg, 1 .044 mmol) were reacted to afford the title compound as a clear oil (256 mg, 55%).
LCMS (Method F): RT = 1 .82 min, M+H+ = 450. Step 3: 1 -(3-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-4- yl) piper azine di( trifluoroacetate) :
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-4-yl)piperazine di(trifluoroacetate), te/t-butyl 4-(3-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)phenyl)piperazine-1 -carboxylate (100 mg, 0.22 mmol) was reacted to afford the title compound as a white solid (64 mg, 83%).
LCMS (Method F): RT = 0.74 min, M+H+ = 350; 1H NMR (500 MHz, MeOH-d4): 8.27 (1 H, d), 7.80 (1 H, s), 7.77 (1 H, s), 7.61 (2H, s), 7.43 (1 H, d), 7.28 (1 H, dd), 4.10 (4H, dd), 3.46 (4H, dd), 1 .80 (4H, s), 1 .40 (6H, s), 1 .35 (6H, s).
Example 4: 8-(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2-yl)Dyridin-4-yl)-2,8- diazaspiro[4.51decane di(trifluoroacetate)
Figure imgf000038_0001
Step 1: tert-butyl 8-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]decane-2-carboxylate:
Following the procedure for te/t-butyl 4-(2-chloropyridin-4-yl)piperazine-1 -carboxylate, 2- chloro-4-iodopyridine (0.15 g, 0.63 mmol) and te/t-butyl 2,8-diazaspiro[4.5]decane-2- carboxylate (151 mg, 0.63 mmol) were reacted to afford the title compound as a yellow oil (162 mg, 77%).
LCMS (Method F): RT = 1 .1 1 min, M+H+ = 352.
Step 2: tert-butyl 8-(2-(5A8,8-tetramethyl-5A 7,8-tetrahydronaphthalen-2-yl)pyrid^^ yl) -2, 8-diazaspiro[4.5]decane-2-carboxylate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, tert-butyl 8-(2-chloropyridin-4-yl)-2,8-diazaspiro[4.5]decane-2- carboxylate (90 mg, 0.256 mmol) and 4,4,5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (80 mg, 0.256 mmol) were reacted to afford the title compound as a yellow oil (45 mg, 44%).
LCMS (Method F): RT = 1 .83 min, M+H+ = 504. Step 3: 8-(2-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-4-yl)-2,8- diazaspiro[4.5]decane di(trifluoroacetate):
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), tert-butyl 8-(2-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyridin-4-yl)-2,8-diazaspiro[4.5]decane-2-carboxylate (45 mg, 0.09 mmol) was reacted to afford the title compound as a white solid (10 mg, 38%). LCMS (Method F): RT = 0.82 min, M+H+ = 404; 1H NMR (500 MHz, MeOH-d4): 8.13 (1 H, d), 7.76 (1 H, s), 7.59 (2H, m), 7.30 (1 H, d), 7.18 (1 H, dd), 3.89-3.93 (2H, m), 3.78-3.82 (2H, m), 3.48 (2H, dd), 3.23 (2H, m), 2.10 (2H, dd), 1 .84-1 .88 (2H, m), 1 .79 (4H, s), 1 .39 (6H, s), 1 .35 (6H, s).
Example 5: 1 -(4-(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-4- vDphenvDcvclobutanamine di(trifluoroacetate)
Figure imgf000039_0001
Step 1: tert-butyl (1-(4-(2-chloropyridin-4-yl)phenyl)cyclobutyl)carbamate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, 2-chloro-4-iodopyridine (0.5 g, 2.01 mmol) and tert-butyl (1 -(4- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)cyclobutyl)carbamate (780 mg, 2.01 mmol) were reacted to afford the title compound as a brown oil (0.50 mg, 67%).
LCMS (Method F): RT = 1 .61 min, M+H+ = 359.
Step 2: tert-butyl (1-(4-(2-(5,5A8-tetramethyl-5A 7,8-tetrahydronaphthalen-2^
4-yl)phenyl)cyclobutyl)carbamate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, tert-butyl (1 -(4-(2-chloropyridin-4-yl)phenyl)cyclobutyl)carbamate (250 mg, 0.70 mmol) and 4,4,5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (219 mg, 0.7 mmol) were reacted to afford the title compound as a clear oil (150 mg, 42%).
LCMS (Method G): RT = 8.43 min, M+H+ = 51 1 . Step 3: 1-(4-(2-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-4- yl)phenyl)cyclobutanamine di(trifluoroacetate):
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), tert-butyl (1 -(4-(2-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyridin-4-yl)phenyl)cyclobutyl)carbamate (150 mg, 0.29 mmol) was reacted to afford the title compound as a off-white solid (60 mg, 50%).
LCMS (Method F): RT = 1 .1 1 min, M+H+ = 41 1 ; 1H NMR (500 MHz, MeOH-d4): 8.82 (1 H, d), 8.49 (1 H, d), 8.18 (1 H, dd), 8.15 (2H, d), 8.02 (1 H, d), 7.76-7.82 (3H, m), 7.66 (1 H, d), 2.83-2.89 (2H, m), 2.70-2.76 (2H, m), 2.28-2.36 (1 H, m), 2.00-2.28 (1 H, m), 1 .81 (4H, s), 1 .42 (6H, s), 1 .37 (6H, s).
Example 6: 2'-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2-yl)-1 ,2,3,6-tetrahvdro-
4.4'-bipyridine di(trifluoroacetate)
Figure imgf000040_0001
Step 1: tert-butyl 2'-chloro-5,6-dihydro-[4,4'-bipyridine]- 1(2H)-carboxylate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, 2-chloro-4-iodopyridine (0.4 g, 1 .67 mmol) and tert-butyl 4- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (0.517 g, 1 .67 mmol) were reacted to afford the title compound as a clear oil (0.35 mg, 71 %).
LCMS (Method G): RT = 6.62 min, M+H+ = 295.
Step 2: tert-butyl 2'-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)-5,6-dihydro- [4,4'-bipyridine]-1(2H)-carboxylate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, tert-butyl 2'-chloro-5,6-dihydro-[4,4'-bipyridine]-1 (2H)-carboxylate (245 mg, 0.83 mmol) and 4,4,5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (261 mg, 0.83 mmol) were reacted to afford the title compound as a brown oil (181 mg, 49%).
LCMS (Method F): RT = 1 .87 min, M+H+ = 447.
Step 3: 2'-(5!5A8-tetramethyl-5^ !8-tetrahydronaphthalen-2-yl)-1!2,3
bipyridine di(trifluoroacetate):
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), te/t-butyl 2'-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)-5,6-dihydro-[4,4'-bipyridine]-1 (2H)-carboxylate (181 mg, 0.40 mmol) was reacted to afford the title compound as a off-white solid (43 mg, 31 %).
LCMS (Method F): RT = 0.89 min, M+H+ = 347; 1H NMR (500 MHz, MeOH-d4): 8.74 (1 H, d), 8.22 (1 H, d), 7.96 (1 H, dd), 7.92 (1 H, d), 7.73 (1 H, dd), 7.64 (1 H, d), 6.89 (1 H, s), 4.04 (2H, d), 3.57 (2H, dd), 2.98 (2H, d), 1 .80 (4H, s), 1 .40 (6H, s), 1 .36 (6H, s).
Example 7: 5-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2-yl)-1 '.2'.3'.6'- tetrahydro-3,4'-bipyridine di(trifluoroacetate)
Figure imgf000041_0001
Step 1: 3-bromo-5-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridine:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, 3,5-dibromopyridine (1 .0 g, 4.22 mmol) and 4,4,5,5-tetramethyl- 2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (1 .327 g, 4.22 mmol) were reacted to afford the title compound as a clear oil (1 .2 g, 83%).
LCMS (Method F): RT = 1 .94 min, M+H+ = 346.
Step 2: tert-butyl 5-(5A8,8-tetramethyl-5A 7,8-tetrahydronaphthalen-2-yl)-^
[3,4'-bipyridine]- 1 '(2'H)-carboxylate:
Following the procedure for for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, 3-bromo-5-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridine (140 mg, 0.41 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan- 2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (0.126 g, 0.41 mmol) were reacted to afford the title compound as a yellow oil (102 mg, 55%).
LCMS (Method F): RT = 1 .90 min, M+H+ = 447.
Step 3: 5-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)- 1 ',2',3',6'-tetrahydro- 3,4'-bipyridine di(trifluoroacetate):
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), tert-butyl 5-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)-5',6'-dihydro-[3,4'-bipyridine]-1 '(2'H)-carboxylate (102 mg, 0.22 mmol) was reacted to afford the title compound as a off-white solid (51 mg, 68%). LCMS (Method F): RT = 0.94 min, M+H+ = 347; 1H NMR (500 MHz, MeOH-d4): 8.92 (1 H, s), 8.78 (1 H, s), 8.49 (1 H, s), 7.70 (1 H, d), 7.52-7.56 (2H, m), 6.53 (1 H, s), 3.97 (2H, d), 3.55 (2H, dd), 2.95 (2H, d), 1 .78 (4H, s), 1 .40 (6H, s), 1 .35 (6H, s).
Example 8: (4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-2- vDphenvDmethanamine di(trifluoroacetate)
Figure imgf000042_0001
Step 1: tert-butyl 4-(6-(5!5!8!8-tetramethyl-5^ ,8-tetrahydronaphthalen-2-yl)pyrffl^^ yl)benzylcarbamate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, 2-bromo-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridine (56 mg, 0.163 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan- 2-yl)benzylcarbamate (54.2 mg, 0.163 mmol) were reacted to afford the title compound as a brown oil (56 mg, 73%).
LCMS (Method F): RT = 2.01 min, M+H+ = 471 .
Step 2: (4-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine di(trifluoroacetate):
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), tert-butyl 4-(6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)benzylcarbamate (56 mg , 0.12 mmol) was reacted to afford the title compound as a white solid (23 mg, 38%).
LCMS (Method F): RT = 1 .20 min, M+H+ = 371 ; 1H NMR (500 MHz, MeOH-d4): 8.23 (2H, d), 8.10 (1 H, d), 7.95 (1 H, dd), 7.81 -7.86 (3H, m), 7.61 (2H, d), 7.48 (1 H, d), 4.20 (2H, s), 1 .77 (4H, s), 1 .37 (6H, s), 1 .32 (6H, s).
Example 9: (4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-2- vDphenvDmethanamine di(trifluoroacetate)
Figure imgf000043_0001
Step 1: tert-butyl 4-(4-(5!5!8!8-tetramethyl-5^ ,8-tetrahydronaphthalen-2-yl)pyrid^^^ yl)benzylcarbamate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, 2-chloro-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridine (77mg, 0.22 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)benzylcarbamate (74.2 mg, 0.22 mmol) were reacted to afford the title compound as a yellow oil (71 mg, 68%).
LCMS (Method F): RT = 1 .87 min, M+H+ = 471 .
Step 2: (4-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine di(trifluoroacetate):
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), tert-butyl 4-(6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)benzylcarbamate (71 mg , 0.15 mmol) was reacted to afford the title compound as a yellow solid (43 mg, 52%).
LCMS (Method F): RT = 1 .09 min, M+H+ = 371 ; 1H NMR (500 MHz, MeOH-d4): 8.76 (1 H, d), 8.35 (1 H, s), 8.14 (2H, d), 8.05 (1 H, d), 7.90 (1 H, d), 7.72-7.74 (3H, m), 7.61 (1 H, d), 4.28 (2H, s), 1 .80 (4H, s), 1 .41 (6H, s), 1 .36 (6H, s).
Example 10: 1 -(6-(5,5,8,8-tetramethyl-5,6 J,8-tetrahvdronaphthalen-2-yl)pyridin-2- yl)piperidin-4-amine
Figure imgf000044_0001
Step 1: 4,4,5,5-Tetramethyl-2-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)- 1 , 3, 2-dioxaborolane:
To a solution of 6-bromo-1 ,1 ,4,4-tetramethyl-1 ,2,3,4-tetrahydronaphthalene (3 g, 1 1 .23 mmol) in DMF (45 ml) was added 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1 ,3,2- dioxaborolane) (4.28 g, 16.84 mmol) and potassium acetate (3.31 g, 33.7 mmol).
Nitrogen was bubbled through the mixture, and PdCI2(dppf) CH2CI2 (0.917 g, 1 .123 mmol) was added. The mixture was heated at 80 °C for 6 h under nitrogen. After cooling to RT, the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 10 % EtOAc in cyclohexane) to afford the title compound as a yellow solid (2.49 g, 70 %).
1H NMR (500 MHz, CDCI3): 7.70-7.69 (1 H, d), 7.51 -7.49 (1 H, dd), 7.25-7.24 (1 H, d), 1 .61 (4H, s), 1 .25 (12H, s), 1 .24 (6H, s), 1 .21 (6H, s).
Step 2: 2-bromo-6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridine:
To a solution of 2,6-dibromopyridine (1 .028 g, 4.34 mmol) in DMF (15 ml) was added 4,4,5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ,3,2- dioxaborolane (1 .5 g, 4.77 mmol) and 2M Na2C03 (aq) (8.68 ml, 17.36 mmol). Nitrogen was bubbled through the mixture for 10 min. PdCI2(dppf)CH2CI2 (0.177 g, 0.217 mmol) was added, and the reaction mixture heated at 80 <Ό for 4 h under nitrogen. After cooling to RT, the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 20 % EtOAc in cyclohexane) to afford the title compound as a yellow oil (1 .39 g, 93%).
LCMS (Method F): RT = 1 .98 min, M+H+ = 346. Step 3: 1-(6-(5,5,8,8-tetramethyl-5^ ,8-tetrahydronaphthalen-2-yl)pyridin
4-amine:
A solution of 2-bromo-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridine (50 mg, 0.145 mmol) and piperidin-4-amine (0.031 ml, 0.290 mmol) in NMP (0.5 ml) was heated at 200 'Ό in a CEM microwave reactor for 30 min. The mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (KP-NH Si02, gradient 0 to 100 % EtOAc in cyclohexane) to afford the title compound (15.7 mg, 29%). LCMS (Method F): RT = 1 .16 min, M+H+ = 364; 1H NMR (500 MHz, MeOH-d4): 8.12-8.1 1 (1 H, d), 7.59-7.58 (1 H, d), 7.45-7.41 (2H, m), 6.98 (1 H, br s), 6.90-6.89 (1 H, dd), 4.33- 4.31 (2H, m), 3.00-2.90 (3H, m), 1 .96-1 .94 (2H, m), 1 .77 (4H, s), 1 .48-1 .40 (2H, m), 1 .36 (6H, s), 1 .33 (6H, s).
Example 1 1 : 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-2-yl) piperazine
Figure imgf000045_0001
Following the procedure for 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)piperidin-4-amine, 2-bromo-6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridine (50 mg, 0.145 mmol) and piperazine (62.5 mg, 0.726 mmol) were reacted to afford the title compound as a yellow gum (7.7 mg, 14 %).
LCMS (Method F): RT = 1 .14 min, M+H+ = 350; 1H NMR (500 MHz, MeOH-d4): 7.99 (1 H, d), 7.75-7.72 (1 H, dd), 7.63-7.60 (1 H, dd), 7.40-7.39 (1 H, d), 7.16-7.15 (1 H, d), 6.74-6.72 (1 H, d), 3.64-3.62 (4H, m), 3.00-2.98 (4H, m), 1 .76 (4H, s), 1 .36 (6H, s), 1 .33 (6H, s).
Example 12: 6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)-1 '.2'.3'.6'- tetrahvdro-2,4'-bipyridine
Figure imgf000046_0001
Step 1: tert-butyl 6-(5,5^-tetramethyl-5^ ^-tetrahydronaphthalen-2-yl)-5 6'-dihydro- [2,4'-bipyridine]- 1 '(2'H)-carboxylate:
To a solution of 2-bromo-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl) pyridine (75 mg, 0.218 mmol) in DMF (1 ml) was added tert-butyl 4-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (74.1 mg, 0.240 mmol) and 2M Na2C03 (aq) (0.436 ml, 0.871 mmol). Nitrogen was bubbled through the mixture for 10 min. PdCI2(dppf)CH2CI2 (8.9 mg, 10.89 μηιοΙ) was added, and the reaction mixture heated at 90 °C for 72 h under nitrogen. After cooling to RT, the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 25 % EtOAc in cyclohexane) to afford the title compound as a solid (63 mg, 64 %).
1H NMR (500 MHz, CDCI3): 7.91 (1 H, s), 7.72-7.70 (1 H, dd), 7.63-7.60 (1 H, dd), 7.50- 7.49 (1 H, d), 7.34-7.32 (1 H, d), 7.23-7.21 (1 H, d), 6.67 (1 H, s), 4.10-4.08 (2H, m), 3.65- 3.55 (2H, m), 2.70-2.65 (2H, m), 1 .65 (4H, s), 1 .43 (9H, s), 1 .29 (6H, s), 1 .24 (6H, s). Step 2: 6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)- 1 ',2',3',6'-tetrahydro- 2,4'-bipyridine:
To a solution of tert-butyl 6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-5',6'- dihydro-[2,4'-bipyridine]-1 '(2'H)-carboxylate (63 mg, 0.141 mmol) in dioxane (1 ml), was added 4M HCI in dioxane (0.5 ml, 2.00 mmol). The reaction was stirred for 1 h at RT. Et20 (approx. 2 ml) was added to the resultant suspension, and the mixture slurried for 15 min. The suspension was filtered, the solid washed with Et20 and then dried in vacuo o/n. Further purification was carried out, using preparative HPLC (Method A and B), to afford the title compound as a white solid (9.6 mg, 20 %).
LCMS (Method F): RT = 1 .09 min, M+H+ = 347; 1H NMR (500 MHz, MeOH-d4): 7.94 (1 H, d), 7.69-7.66 (2H, m), 7.58-7.56 (1 H, d), 7.33 (1 H, d), 7.31 (1 H, d), 6.70-6.66 (1 H, m), 3.50-3.48 (2H, m), 3.05-3.03 (2H, t), 2.66-2.61 (2H, m), 1 .65 (4H, s), 1 .26 (6H, s), 1 .22 (6H, s).
Example 13: 1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2-yl)Dyridin-2- yl)phenyl)cvclobutanamine dihydrochloride
Figure imgf000047_0001
Step 1: tert-butyl 1-(4-(6-(5,5^^-tetramethyl-5^J^-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)cyclobutylcarbamate:
To a solution of tert-butyl 1 -(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)
phenyl)cyclobutylcarbamate (178.9 mg, 0.479 mmol) and 2-bromo-6-(5, 5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridine (150 mg, 0.436 mmol) in DMF (2 ml) was added 2M Na2C03 (aq) (0.44 ml, 1 .743 mmol). Nitrogen was bubbled through the mixture for 5 min. PdCI2(dppf)CH2CI2 (17.8 mg, 0.022 mmol) was added, and the reaction mixture was heated at 80 °C for 18 h under nitrogen. After cooling to RT, the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 30 % EtOAc in cyclohexane) to afford the title compound as a white solid (107 mg, 48 %).
LCMS (Method F): RT = 2.13 min, M+H+ = 51 1 .
Step 2: 1-(4-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) phenyl)cyclobutanamine hydrochloride:
tert-Butyl 1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) phenyl)cyclobutylcarbamate (107 mg, 0.210 mmol) was dissolved in DCM (2 ml) and MeOH (2 ml), and loaded onto a 5 g SCX cartridge. After 5 h at RT, the cartridge was washed with MeOH, and the compound eluted with 7N NH3 in MeOH. The product containing fractions were concentrated in vacuo and the residue dissolved in DCM
(approx. 1 ml). TFA (approx. 1 ml) was added, and the mixture stirred at RT for 15 min. The mixture was concentrated in vacuo, and DCM was added and subsequently removed in vacuo (three times) to remove TFA. The residue was dissolved in DCM (2 ml) and basified with saturated aq. NaHC03. After filtration through a phase separator cartridge, the filtrates were concentrated in vacuo. The resultant residue was subjected to flash chromatography (KP-NH Si02, gradient 0 to 100 % EtOAc in cyclohexane). The isolated material was dissolved in 1 ml dioxane and 4M HCI in dioxane (1 .0 ml) was added, then the solvent was concentrated in vacuo and lyophilized o/n to afford the title compound as an off-white solid (24.1 mg, 24 %).
LCMS (Method F): RT = 1 .30 min, M+H+ = 41 1 ; 1H NMR (500 MHz, MeOH-d4): 8.62-8.59 (1 H, m), 8.23-8.15 (4H, m), 8.00 (1 H, d), 7.86-7.84 (2H, m), 7.79-7.77 (1 H, dd), 7.67-
7.66 (1 H, d), 2.90-2.85 (2H, m), 2.76-2.70 (2H, m), 2.36-2.30 (1 H, m), 2.08-2.02 (1 H, m), 1 .82 (4H, s), 1 .43 (6H, s), 1 .38 (6H, s).
Example 14: 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2-yl)Dyridin-2-yl)-2,8- diazaspiro[4.51decane dihvdrochloride
Figure imgf000048_0001
Step 1: tert-butyl 8-(6-(5,5^ etra et y\-5^ etra ydronap t a\en-2-y\)pyridin-2- yl) -2, 8-diazaspiro[4.5]decane-2-carboxylate:
To a solution of tert-butyl 2,8-diazaspiro[4.5]decane-2-carboxylate (62.8 mg, 0.261 mmol) and 2-bromo-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridine (75 mg, 0.218 mmol) in dioxane (1 ml) was added Xantphos (12.60 mg, 0.022 mmol) and K3PO4 (102 mg, 0.479 mmol). Pd2dba3 (1 1 .97 mg, 0.013 mmol) was added, and nitrogen was bubbled through the mixture for 5 min. The reaction mixture was heated at 90 °C for 18 h under nitrogen. After cooling to RT, the mixture was filtered through celite and washed with EtOAc. The combined filtrates were concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 25 % EtOAc in cyclohexane) to afford the title compound as a yellow foam (34.2 mg, 31 %).
1H NMR (500 MHz, CDCI3): 7.86 (1 H, m), 7.67-7.65 (1 H, d), 7.46-7.43 (1 H, m), 7.30-7.29 (1 H, d), 6.98-6.97 (1 H, m), 6.54-6.51 (1 H, m), 3.75-3.60 (2H, m), 3.54-3.41 (2H, m), 3.40-3.32 (2H, m), 3.23 (1 H, s), 3.14 (1 H, s), 1 .72-1 .69 (2H, t), 1 .67-1 .57 (4H, m), 1 .64 (4H, s), 1 .40 (9H, s), 1 .27 (6H, s), 1 .23 (6H, s).
Step 2: 8-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8- diazaspiro[4.5]decane dihydrochloride:
To a solution of tert-butyl 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane-2-carboxylate (34.2 mg, 0.068 mmol) in dioxane (0.5 ml) was added 4M HCI in dioxane (0.5 ml, 2.000 mmol). The reaction was stirred for 1 h at RT. Et20 (approx. 2 ml) was added to the resultant suspension and the mixture slurried for 15 min. The supernatant liquid was removed by pipette. The Et20 slurry was repeated twice. The solid residue was dissolved in MeOH (1 ml) / H20 (1 ml) and lyophilised o/n to afford the title compound as a yellow solid (20.8 mg, 64%).
LCMS (Method F): RT = 1 .14 min, M+H+ = 404; 1H NMR (500 MHz, MeOH-d4): 8.00-7.97 (1 H, m), 7.61 (1 H, d), 7.48-7.47 (1 H, d), 7.42-7.40 (1 H, dd), 7.27-7.25 (1 H, d), 7.07-7.06 (1 H, d). 3.77-3.73 (2H, m), 3.69-3.64 (2H, m), 3.38-3.35 (2H, t), 3.15 (2H, s), 2.00-1 .97 (2H, t), 1 .85-1 .77 (4H, m), 1 .67 (4H, s), 1 .26 (6H, s), 1 .23 (6H, s).
Example 15: 1 -(4-(5,5,8,8-tetramethyl-5,67,8-tetrahvdronaphthalen-2-yl)pyridin-2- yl)piperidin-4-amine
Figure imgf000049_0001
Step 1: 2-chloro-4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridine:
To a solution of 2-chloro-4-iodopyridine (1 .385 g, 5.79 mmol) in DMF (20 ml) was added 4,4,5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ,3,2- dioxaborolane (2 g, 6.36 mmol) and 2M Na2C03 (aq) (1 1 .6 ml, 23.14 mmol). Nitrogen was bubbled through the mixture for 5 min. PdCI2(dppf) CH2CI2 (0.236 g, 0.289 mmol) was added and the reaction mixture heated at 60 °C for 2 h under nitrogen. After cooling to RT the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 20 % EtOAc in cyclohexane) to afford the title compound as a pale yellow solid (1 .34 g, 77 %).
1H NMR (500 MHz, CDCI3): 8.40-8.39 (1 H, d), 7.53-7.51 (2H, m), 7.44-7.40 (2H, m), 7.38-7.36 (1 H, dd), 1 .73 (4H, s), 1 .34 (6H, s), 1 .32 (6H, s).
Step 2: 1-(4-(5,5,8,8-tetramethyl-5fi ,8-tetrahydronaphthalen-2-yl)pyridin
4-amine:
Following the procedure for 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)piperidin-4-amine, 2-chloro-4-(5,5, 8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridine (50 mg, 0.167 mmol) and piperidin-4-amine (0.035 ml, 0.334 mmol) were reacted to afford the title compound as a white solid (30.6 mg, 50 %).
LCMS (Method F): RT = 0.90 min, M+H+ = 364; 1H NMR (500 MHz, MeOH-d4): 8.00-7.99 (1 H, d), 7.73-7.72 (1 H, dd), 7.60-7.57 (1 H, dd), 7.40-7.38 (1 H, d), 7.1 1 -7.09 (1 H, d), 6.76-6.74 (1 H, d), 4.48-4.46 (2H, m), 2.99-2.94 (3H, m), 1 .97-1 .94 (2H, m), 1 .76 (4H, s), 1 .50-1 .43 (2H, m), 1 .36 (6H, s), 1 .33 (6H, s).
Example 16: 1 -(4-(5,5,8,8-tetramethyl-5,6 J,8-tetrahvdronaphthalen-2-yl)pyridin-2-yl) piperazine
Figure imgf000050_0001
Step 1: 1-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) piperazine:
Following the procedure for 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)piperidin-4-amine, 2-chloro-4-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridine (50 mg, 0.167 mmol) and piperazine (28.7 mg, 0.334 mmol) were reacted to afford the title compound as a yellow solid (12.7 mg, 22 %). LCMS (Method F): RT = 0.98 min, M+H+ = 350; 1H NMR (500 MHz, MeOH-d4): 8.15-8.14 (1 H, d), 7.60 (s, 1 H), 7.45 (2H, s), 6.99-6.96 (2H, m), 3.60-3.58 (4H, m), 3.03-3.01 (4H, m), 1 .77 (4H, s), 1 .36 (6H, s), 1 .33 (6H, s). Example 17: 4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)-1 ',2',3',6'- tetrahvdro-2,4'-bipyridine dihydrochloride
Figure imgf000051_0001
Step 1: tert-butyl 4-(5!5A8-tetramethyl-5!6 !8-tetrahydronaphthalen-2-yl)-5'!6'-dihydro- [2,4'-bipyridine]- 1 '(2'H)-carboxylate:
Following the procedure for tert-butyl 6-(5,5,8, 8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)-5',6'-dihydro-[2,4'-bipyridine]-1 '(2'H)-carboxylate, 2-chloro-4- (5,5,8, 8-tetramethyl-5, 6,7, 8-tetrahydronaphthalen-2-yl)pyridine (75 mg, 0.250 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-5,6-dihydropyridine-1 (2H)- carboxylate (85 mg, 0.275 mmol) were reacted to afford the title compound as a solid (54 mg, 48 %).
1H NMR (500 MHz, CDCI3): 8.52-8.51 (1 H, d), 7.48-7.46 (2H, m), 7.36-7.31 (2H, m), 7.27-7.26 (1 H, dd), 6.57 (1 H, br s), 4.09 (2H, br m), 3.61 (2H, br m), 2.64 (2H, br m), 1 .66 (4H, s), 1 .43 (9H, s), 1 .28 (6H, s), 1 .26 (6H, s).
Step 2: 4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)- 1 ',2',3',6'-tetrahydro- 2, 4 '-bipyridine dihydrochloride:
Following the procedure for 6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)- 1 ',2',3',6'-tetrahydro-2,4'-bipyridine dihydrochloride, tert-butyl 4-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)-5',6'-dihydro-[2,4'-bipyridine]-1 '(2'H)-carboxylate (54 mg, 0.121 mmol) was reacted to afford the title compound as a white solid (27.4 mg, 59 %).
LCMS (Method F): RT = 0.99 min, M+H+ = 347; 1H NMR (500 MHz, MeOH-d4): 8.74-8.73 (1 H, d), 8.37 (1 H, d), 8.29-8.28 (1 H, dd), 7.96 (1 H, d), 7.81 -7.78 (1 H, dd), 7.64-7.62 (1 H, d), 6.93-6.91 (1 H, m), 4.10-4.09 (2H, m), 3.61 (2H, t), 3.08-3.06 (2H, m), 1 .80 (4H, s), 1 .42 (6H, s), 1 .37 (6H, s). Example 18: 1 -(4-(4-(5,5,8,8-tetramethyl-5,6 J,8-tetrahvdronaphthalen-2-yl)pyridin-2- vDphenvDcvclobutanamine dihvdrochloride
Figure imgf000052_0001
Step 1: tert-butyl (1-(4-(4-(5,5A8-tetramethyl-5A7,8-tetrahydronaphthalen-2-y^
2-yl)phenyl)cyclobutyl)carbamate:
Following the procedure for tert-butyl 1 -(4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)phenyl)cyclobutylcarbamate, tert-butyl (1 -(4- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)cyclobutyl)carbamate (103 mg, 0.275 mmol) and 2-chloro-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridine (75 mg, 0.250 mmol) were reacted to afford the title compound (104.9 mg, 82 %).
1H NMR (500 MHz, CDCI3): 8.64-8.63 (1 H, d), 7.95-7.93 (2H, d), 7.82 (1 H, s), 7.54 (1 H, s), 7.49-7.48 (2H, m), 7.38 (2H, s), 7.35-7.34 (1 H, dd), 5.03 (1 H, br s), 2.53-2.50 (4H, m), 2.09-2.03 (1 H, m), 1 .83-1 .78 (1 H, m), 1 .67 (4H, s), 1 .29 (9H, br s), 1 .30 (6H, s), 1 .27 (6H, s).
Step 2: 1-(4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) phenyl)cyclobutanamine dihydrochloride:
Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl (1 -(4-(4-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)phenyl)cyclobutyl)carbamate (104.9 mg, 0.205 mmol) was reacted to afford the title compound as an off-white solid (74.1 mg, 81 %).
LCMS (Method F): RT = 1 .07 min, M+H+ = 41 1 ; 1H NMR (500 MHz, MeOH-d4): 8.70-8.69 (1 H, d), 8.45 (1 H, d), 8.20-8.19 (1 H, dd), 8.09-8.07 (2H, m), 7.88 (1 H, d), 7.77-7.75 (2H, m), 7.72-7.70 (1 H, dd), 7.53-7.51 (1 H, d), 2.79-2.73 (2H, m), 2.62-2.58 (2H, m), 2.23- 2.17 (1 H, m), 1 .98-1 .89 (1 H, m), 1 .69 (4H, s), 1 .30 (6H, s), 1 .25 (6H, s).
Example 19: (7S.8aS)-2-(4-(5.5.8.8-tetramethyl-5.6.7.8-tetrahydronaphthalen-2- yl)pyridin-2-yl)octahvdropyrrolo[1 ,2-a1pyrazin-7-ol
Figure imgf000053_0001
Following the procedure for 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)piperidin-4-amine, 2-chloro-4-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridine (50 mg, 0.167 mmol) and (7S,8aS)-tert-butyl 7- hydroxyhexahydropyrrolo[1 ,2-a]pyrazine-2(1 H)-carboxylate (101 mg, 0.417 mmol) were reacted to afford the title compound as an orange solid (21 .8 mg, 32 %).
LCMS (Method F): RT = 1 .01 min, M+H+ = 406; 1H NMR (500 MHz, MeOH-d4): 8.13-8.12 (1 H, d), 7.60 (1 H, s), 7.44 (2H, s), 6.98 (1 H, s), 6.94-6.92 (1 H, dd), 4.42-4.38 (2H, m), 4.27-4.23 (1 H, m), 3.15-3.04 (3H, m), 2.82-2.77 (1 H, m), 2.45-2.40 (2H, m), 2.34-2.27 (1 H, m), 2.23-2.16 (1 H, m), 1 .77 (4H, s), 1 .36 (6H, s), 1 .33 (6H, s). -(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)-2.4'-bipyridine
Figure imgf000053_0002
Following the procedure for tert-butyl 1 -(4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)phenyl)cyclobutylcarbamate, 2-chloro-4-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridine (250 mg, 0.834 mmol) and pyridin-4-ylboronic acid (1 13 mg, 0.917 mmol) were reacted to afford the title compound as an off-white solid (5.5 mg, 2 %).
LCMS (Method F): RT = 1 .57 min, M+H+ = 343; 1H NMR (500 MHz, CDCI3): 8.84-8.81 (3H, m), 8.46-8.45 (2H, d), 8.09 (1 H, s), 7.67-7.66 (1 H, d), 7.60-7.59 (1 H, d), 7.50-7.44 (2H, m), 1 .75 (4H, s), 1 .37 (6H, s), 1 .34 (6H, s).
Example 21 : 1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyrimidin-2- yl)piperidin-4-amine dihydrochloride
Figure imgf000054_0001
Step 1: 2-chloro-4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidine: To a solution of 2,4-dichloropyrimidine (0.603 g, 4.05 mmol) and 4,4,5, 5-tetramethyl-2- (5,5,8, 8-tetramethyl-5, 6,7, 8-tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (1 .4 g, 4.45 mmol) in DMF (12 ml) was added 2M Na2C03 (aq) (8.10 ml, 16.20 mmol). Nitrogen was bubbled through the mixture for 5 min. PdCI2(dppf)CH2CI2 (0.331 g, 0.405 mmol) was added, and the reaction mixture was heated at 80 °C for 18 h under nitrogen. After cooling to RT, the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 20 % EtOAc in cyclohexane) to afford the title compound as a pale yellow oil (596 mg, 49%).
1H NMR (500 MHz, CDCI3): 8.62-8.61 (1 H, d), 8.06 (1 H, d), 7.85-7.83 (1 H, m), 7.63-7.62 (1 H, d), 7.47-7.46 (1 H, d), 1 .75 (4H, s), 1 .38 (6H, s), 1 .34 (6H, s).
Step 2: tert-butyl (1-(4-(5,5,8,8-tetramethyl-5^ ,8-tetrahydronaphthalen-2-yl)pyrimidin- 2-yl)piperidin-4-yl)carbamate:
Following the procedure for tert-butyl 8-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane-2-carboxylate, tert- butyl piperidin-4-ylcarbamate (59.9 mg, 0.299 mmol) and 2-chloro-4-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine (75 mg, 0.249 mmol) were reacted to afford the title compound (55.4 mg, 48 %).
1H NMR (500 MHz, CDCI3): 8.26-8.25 (1 H, d), 7.88 (1 H, d), 7.71 -7.69 (1 H, dd), 7.34-7.32 (1 H, d), 6.82-6.81 (1 H, d), 4.72-4.69 (2H, m), 4.40 (1 H, br s), 3.67 (1 H, br s), 3.07-3.01 (2H, m), 2.00-1 .95 (2H, m), 1 .65 (4H, s), 1 .39 (9H, s), 1 .36-1 .29 (2H, m), 1 .27 (6H, s), 1 .24 (6H, s).
Step 3: 1 -(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)piperidin-4-amine dihydrochloride: Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl (1 -(4-(5, 5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyr
(55.4 mg, 0.1 19 mmol) was reacted to afford the title compound as an off-white solid (29.1 mg, 56 %).
LCMS (Method F): RT = 1 .07 min, M+H+ = 365; 1H NMR (500 MHz, MeOH-d4): 8.25-8.24 (1 H, d), 8.10 (1 H, d), 7.91 -7.89 (1 H, dd), 7.47-7.45 (1 H, d), 7.43-7.42 (1 H, d), 4.76-4.72 (2H, m), 3.47-3.44 (1 H, m), 3.29-3.23 (2H, m), 2.17-2.14 (2H, m), 1 .68-1 .63 (6H, m), 1 .28 (6H, s), 1 .25 (6H, s).
Example 22: 2-(piperazin-1 -yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl) pyrimidine dihydrochloride
Figure imgf000055_0001
Step 1: tert-butyl 4-(4-(5,5,8,8-tetramethyl-5^ ,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)piperazine- 1 -carboxylate:
Following the procedure for tert-butyl 8-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane-2-carboxylate, tert- butyl piperazine-1 -carboxylate (55.7 mg, 0.299 mmol) and 2-chloro-4-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine (75 mg, 0.249 mmol) were reacted to afford the title compound (40.5 mg, 36 %).
1H NMR (500 MHz, CDCI3): 8.28-8.27 (1 H, d), 7.89-7.88 (1 H, d), 7.71 -7.69 (1 H, dd), 7.34-7.33 (1 H, d), 6.87-6.86 (1 H, d), 3.84-3.82 (4H, m), 3.47-3.45 (4H, m), 1 .65 (4H, s), 1 .43 (9H, s), 1 .27 (6H, s), 1 .24 (6H, s).
Step 2: 2-(piperazin- 1 -yl) -4-(5, 5, 8, 8-tetramethyl-5, 6, 7, 8-tetrahydronaphthalen-2-yl) pyrimidine dihydrochloride:
Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride tert-butyl 4-(4-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazine-1 -carboxylate (40.5 mg, 0.090 mmol) was reacted to afford the title compound as an off-white solid (17.1 mg, 45 %).
LCMS (Method F): RT = 1 .06 min, M+H+ = 351 ; 1H NMR (500 MHz, MeOH-d4): 8.45-8.43 (1 H, d), 8.19-8.18 (1 H, d), 8.00-7.98 (1 H, dd), 7.55-7.52 (2H, m), 4.26-4.24 (4H, m), 3.47-3.45 (4H, m), 1 .79 (4H, s), 1 .38 (6H, s), 1 .35 (6H, s).
Example 23: 2-(pyridin-4-yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDpyrimidine
Figure imgf000056_0001
To a solution of 2-chloro-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidine (240 mg, 0.798 mmol) in DMF (5 ml) was added pyridin-4-yl-boronic acid (108 mg, 0.878 mmol) and 2M Na2C03 (aq.) (1 .596 ml, 3.19 mmol). Nitrogen was bubbled through the mixture for 5 min. PdCI2(dppf)CH2CI2 (65.2 mg, 0.080 mmol) was added, and the reaction mixture heated at 80 °C for 18 h. After cooling to RT the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 50 % EtOAc in cyclohexane) to afford the title compound as a colourless oil (83.9 mg, 31 %).
LCMS (Method F): RT = 1 .70 min, M+H+ = 344; 1H NMR (500 MHz, MeOH-d4): 8.91 -8.90 (1 H, d), 8.75-8.74 (2H, m), 8.51 -8.49 (2H, m), 8.30-8.29 (1 H, d), 8.06-8.04 (1 H, dd), 7.95-7.94 (1 H, d), 7.55-7.54 (1 H, d), 1 .79 (4H, s), 1 .42 (6H, s), 1 .36 (6H, s).
Example 24: 2-(1 -methyl-1 ,2.3.6-tetrahvdropyridin-4-yl)-4-(5.5.8.8-tetramethyl-5.6.7.8- tetrahvdronaphthalen-2-yl)pyrimidine
Figure imgf000057_0001
To a solution of 2-(pyridin-4-yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl) pyrimidine (75 mg, 0.218 mmol) in acetone (1 ml) was added Mel (0.034 ml, 0.546 mmol). After stirring at RT for 18 h further Mel (0.017 ml, 0.273 mmol) was added. After stirring for 6 h at RT the mixture was concentrated in vacuo. The crude mixture was re- suspended in MeOH (1 ml) and NaBH4 (41 .3 mg, 1 .092 mmol) was added. After stirring at RT for 18 h the mixture was concentrated in vacuo. The residue was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, the combined organic extracts were dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 100 % EtOAc in
cyclohexane, increasing to 20% MeOH in EtOAc) to afford the title compound as an off- white solid (55.2 mg, 70%).
LCMS (Method F): RT = 1 .09 min, M+H+ = 362; 1H NMR (500 MHz, MeOH-d4): 8.71 -8.70 (1 H, d), 8.22 (1 H, d), 7.96-7.94 (1 H, d), 7.75-7.73 (1 H, d), 7.51 -7.49 (1 H, d), 7.31 -7.29 (1 H, m), 3.33-3.31 (2H, m), 2.90-2.88 (2H, m), 2.83-2.80 (2H, m), 2.48 (3H, s), 1 .78 (4H, s), 1 .38 (6H, s), 1 .35 (6H, s).
Example 25: 1 -(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2-yl)Dyrimidin-2- vDphenvDcyclobutanamine dihydrochloride
Figure imgf000057_0002
Step 1: tert-butyl (1-(4-(4-(5,5A8-tetramethyl-5,6 ,8-tetrahydronaphthalen-2- yl)pyrimidin-2-yl)phenyl)cyclobutyl)carbamate:
Following the procedure for tert-butyl 1 -(4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)phenyl)cyclobutylcarbamate, tert-butyl (1 -(4- (4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)phenyl)cyclobutyl)carbamate (102 mg, 0.274 mmol) and 2-chloro-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine (75 mg, 0.249 mmol) were reacted to afford the title compound (66.1 mg, 52 %).
1H NMR (500 MHz, CDCI3): 8.73-8.72 (1 H, d), 8.48-8.47 (2H, d), 8.1 1 (1 H, d), 7.88-7.86 (1 H, dd), 7.51 -7.47 (3H, m), 7.41 -7.40 (1 H, d), 5.04 (1 H, br s), 2.58-2.46 (4H, m), 2.10- 2.02 (1 H, m), 1 .86-1 .80 (1 H, m), 1 .68 (4H, s), 1 .36-1 .25 (21 H, m).
Step 2: 1-(4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)phenyl)cyclobutanamine dihydrochloride:
Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl (1 -(4-(4-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)phenyl)cyclobutyl) carbamate (66.1 mg, 0.129 mmol) was reacted to afford the title compound as an off- white solid (47.6 mg, 82 %).
LCMS (Method F): RT = 1 .18 min, M+H+ = 412; 1H NMR (500 MHz, MeOH-d4): 8.79-8.78 (1 H, d), 8.54-8.52 (2H, d), 8.23 (1 H, d), 7.99-7.97 (1 H, dd), 7.90-7.89 (1 H, d), 7.65-7.63 (2H, d), 7.47-7.45 (1 H, d), 2.81 -2.75 (2H, m), 2.61 -2.55 (2H, m), 2.23-2.17 (1 H, m), 1 .99- 1 .92 (1 H, m), 1 .69 (4H, s), 1 .31 (6H, s), 1 .26 (6H, s).
Example 26: 8-(4-(5,5,8,8-tetramethyl-5,6J,8-tetrahvdronaphthalen-2-yl)pyrimidin-2-yl)- 2.8-diazaspiro[4.51decane dihydrochloride
Figure imgf000058_0001
Step 1: tert-butyl 8-(4-(5,5,8,8-tetramethyl-5^ ,8-tetrahydronaphthalen-2-yl)pyrimidin yl) -2, 8-diazaspiro[4.5]decane-2-carboxylate:
Following the procedure for tert-butyl 8-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane-2-carboxylate, tert- butyl 2,8-diazaspiro[4.5]decane-2-carboxylate (71 .9 mg, 0.299 mmol) and 2-chloro-4- (5,5,8, 8-tetramethyl-5, 6,7, 8-tetrahydronaphthalen-2-yl)pyrimidine (75 mg, 0.249 mmol) were reacted to afford the title compound as a yellow oil (45.6 mg, 36 %). 1H NMR (500 MHz, CDCI3): 8.27-8.26 (1 H, d), 7.90-7.89 (1 H, d), 7.70-7.68 (1 H, d), 7.34- 7.32 (1 H, d), 6.83-6.80 (1 H, m), 4.07-3.90 (2H, m), 3.80-3.63 (2H, m), 3.42-3.32 (2H, dt), 3.24-3.15 (2H, d), 1 .73-1 .70 (2H, t), 1 .65 (4H, s), 1 .60-1 .50 (4H, m), 1 .40 (9H, s), 1 .27 (6H, s), 1 .24 (6H, s).
Step 2: 8-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)-2,8- diazaspiro[4.5]decane dihydrochloride:
Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl 8-(4-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)-2,8-diazaspiro[4.5]decane- 2-carboxylate (45.6 mg, 0.090 mmol) was reacted to afford the title compound as a yellow solid (26.5 mg, 61 %).
LCMS (Method F): RT = 1 .14 min, M+H+ = 405; 1H NMR (500 MHz, MeOH-d4): 8.21 -8.19 (1 H, d), 8.12 (1 H, d), 7.93-7.91 (1 H, dd), 7.48-7.45 (2H, m), 4.08-3.98 (2H, m), 3.90-3.82 (2H, m), 3.40-3.37 (2H, t), 3.17 (2H, s), 2.03-2.00 (2H, t), 1 .84-1 .80 (4H, m), 1 .68 (4H, s), 1 .27 (6H, s), 1 .25 (6H, s).
Example 27: 2-(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyrimidin-2- yl)piperazin-1 -vDethanol
Figure imgf000059_0001
Following the procedure for 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)piperidin-4-amine, 2-chloro-4-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidine (75 mg, 0.249 mmol) and 2-(piperazin-1 -yl)ethanol (61 .2 μΙ, 0.499 mmol) were reacted to afford the title compound as an off-white solid (21 .4 mg, 22%).
LCMS (Method F): RT = 1 .09 min, M+H+ = 395; 1H NMR (500 MHz, MeOH-d4): 8.24-8.23 (1 H, d), 7.96 (1 H, d), 7.73-7.71 (1 H, dd), 7.34-7.32 (1 H, d), 6.99-6.98 (1 H, d), 3.87-3.83 (4H, m), 3.67-3.65 (2H, t), 2.62-2.54 (6H, m), 1 .65 (4H, s), 1 .24 (6H, s), 1 .21 (6H, s). Example 28: N-(pyridin-4-ylmethyl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-
Figure imgf000060_0001
Following the procedure for 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)piperidin-4-amine, 2-chloro-4-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidine (50 mg, 0.166 mmol) and pyridin-4-ylmethanamine (0.034 ml, 0.332 mmol) were reacted to afford the title compound as an orange solid (20.2 mg, 33%).
LCMS (Method F): RT = 1 .17 min, M+H+ = 373; 1H NMR (500 MHz, MeOH-d4): 8.47-8.46 (2H, dd), 8.30-8.29 (1 H, d), 7.96 (1 H, br s), 7.73-7.69 (1 H, m), 7.48-7.47 (2H, d), 7.42- 7.40 (1 H, d), 7.14-7.12 (1 H, d), 4.73 (2H, s), 1 .74 (4H, s), 1 .31 (6H, s), 1 .28 (6H, s).
Example 29: 2-(Dyridin-3-yl)-4-(5.5.8.8-tetramethyl-5.6.7.8-tetrahvdronaDhthalen-2- vDpyrimidine
Figure imgf000060_0002
Step 1: 2-(pyridin-3-yl)4-(5A8,8-tetramethyl-5A7,8-tetrahydronapM
pyrimidine:
Following the procedure for tert-butyl 1 -(4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)phenyl)cyclobutylcarbamate, 2-chloro-4-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine (200 mg, 0.665 mmol), and pyridin-3-yl-boronic acid (90 mg, 0.731 mmol) were reacted to afford the title compound as an off-white solid (84.4 mg, 37%).
LCMS (Method F): RT = 1 .78 min, M+H+ = 344; 1H NMR (500 MHz, CDCI3): 9.82 (1 H, d), 9.01 -9.00 (1 H, d), 8.85-8.83 (1 H, d), 8.77-8.75 (1 H, dd), 8.16 (1 H, d), 7.95-7.93 (1 H, dd), 7.67-7.66 (1 H, d), 7.62-7.60 (1 H, dd), 7.50-7.49 (1 H, d), 1 .75 (4H), 1 .39 (6H, s), 1 .34 (6H, s).
Example 30: 1 -(2-(5,5,8,8-tetramethyl-5,67,8-tetrahvdronaphthalen-2-yl)pyrimidin-4- yl)piperidin-4-amine dihydrochloride
Figure imgf000061_0001
Step 1: tert-butyl (1 -(2-chloropyrimidin-4-yl)piperidin-4-yl)carbamate:
To a solution of 2,4-dichloropyrimidine (250 mg, 1 .678 mmol) and tert-butyl piperidin-4- ylcarbamate (370 mg, 1 .846 mmol) in DMF (2.5 ml) was added Et3N (0.351 ml, 2.52 mmol) at RT. After stirring at RT for 18 h the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant solid was slurried in Et20, filtered and dried under vacuum to afford the title compound as a white solid (272 mg, 52%).
1H NMR (500 MHz, CDCI3): 8.06-8.04 (1 H, d), 6.43-6.42 (1 H, d), 4.55-4.20 (3H, br m), 3.85-3.70 (1 H, br m), 3.12-3.05 (2H, dt), 2.12-2.02 (2H, m), 1 .48 (9H, s), 1 .43-1 .35 (2H, ddd).
Step 2: tert-butyl (1-(2-(5,5A8-tetramethyl-5A 7,8-tetrahydronaphthalen-2-yl)pyr^ 4-yl)piperidin-4-yl)carbamate:
Following the procedure for 2-bromo-6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridine, tert-butyl (1 -(2-chloropyrimidin-4-yl)piperidin-4- yl)carbamate (100 mg, 0.320 mmol) and 4,4,5,5-tetramethyl-2-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (1 1 1 mg, 0.352 mmol) were reacted to afford the title compound as a white foam (81 .3 mg, 55%).
1H NMR (500 MHz, CDCI3): 8.25-8.23 (2H, m), 8.02-8.00 (1 H, dd), 7.32-7.30 (1 H, d), 6.34-6.33 (1 H, d), 4.42-4.34 (3H, m), 3.69 (1 H, br s), 3.05-2.99 (2H, m), 2.01 -1 .99 (2H, m), 1 .64 (4H, s), 1 .35-1 .31 (2H, dd), 1 .29 (6H, s), 1 .24 (6H, s). Step 3: 1 -(2-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-4- yl)piperidin-4-amine dihydrochloride:
Following the procedure for 4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)- 1 \2',3',6'-tetrahydro-2,4'-bipyridine hydrochloride, tert-butyl (1 -(2-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperidin-4-yl)carbamate (81 mg, 0.174 mmol) was reacted to afford the title compound as a white solid (32.6 mg, 43 %).
LCMS (Method F): RT = 0.75 min, M+H+ = 365; 1H NMR (500 MHz, MeOH-d4): 8.25-8.24 (1 H, d), 8.17 (1 H, d), 7.99-7.97 (1 H, dd), 7.64-7.63 (1 H, d), 7.19-7.17 (1 H, d), 5.41 (1 H, br s), 4.45 (1 H, br s), 3.65-3.60 (1 H, m), 3.55-3.35 (2H, br m), 2.31 -2.29 (2H, d), 1 .80 (4H, s), 1 .79-1 .70 (2H, m), 1 .40 (6H, s), 1 .36 (6H, s).
Example 31 : 4-(piperazin-1 -yl)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDpyrimidine dihydrochloride
Figure imgf000062_0001
Step 1: tert-butyl 4-(2-chloropyrimidin-4-yl)piperazine- 1-carboxylate:
Following the procedure for tert-butyl (1 -(2-chloropyrimidin-4-yl)piperidin-4-yl)carbamate, 2,4-dichloropyrimidine (250 mg, 1 .678 mmol) and tert-butyl piperazine-1 -carboxylate (344 mg, 1 .846 mmol) were reacted to afford the title compound as a white solid
(169 mg, 34%).
1H NMR (500 MHz, CDCI3): 8.10-8.09 (1 H, d), 6.42-6.41 (1 H, d), 3.72-3.64 (4H, m), 3.58- 3.53 (4H, m), 1 .51 (9H, s).
Step 2: tert-butyl (1-(2-(5,5^-tetramethyl-5^ ^-tetrahydronaphthalen-2-yl)pyr^ 4-yl)piperidin-4-yl)carbamate:
Following the procedure for 2-bromo-6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridine, tert-butyl 4-(2-chloropyrimidin-4-yl)piperazine-1 - carboxylate (100 mg, 0.335 mmol) and 4,4,5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (1 16 mg, 0.368 mmol) were reacted to afford the title compound as a colourless oil (90.5 mg, 60%). 1H NMR (500 MHz, CDCI3): 8.38-8.37 (1 H, d), 8.35 (1 H, d), 8.12-8.10 (1 H, dd), 7.42-7.40 (1 H, d), 6.42-6.41 (1 H, d), 3.78-3.74 (4H, br m), 3.60-3.58 (4H, m), 1 .74 (4H, s), 1 .39 (6H, s), 1 .33 (6H, s). Step 3: 4-(piperazin- 1 -yl)-2-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidine dihydrochloride:
Following the procedure for 4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)- 1 ',2',3',6'-tetrahydro-2,4'-bipyridine dihydrochloride, tert-butyl 4-(2-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperazine-1 -carboxylate (90 mg, 0.200 mmol) was reacted to afford the title compound as a white solid (32.6 mg, 54%).
LCMS (Method F): RT = 0.71 min, M+H+ = 351 ; 1H NMR (500 MHz, MeOH-d4): 8.25-8.24 (1 H, d), 8.09 (1 H, d), 7.92-7.90 (1 H, dd), 7.54-7.52 (1 H, d), 7.13-7.1 1 (1 H, d), 4.24 (4H, br s), 3.41 -3.39 (4H, t), 1 .70 (4H, s), 1 .30 (6H, s), 1 .26 (6H, s). Example 32: 1 -(6-(3-isopropoxy-4-isopropylphenyl)pyridin-2-yl)piperazine
dihydrochloride
Figure imgf000063_0001
Step 1: Methyl 4-bromo-2-isopropoxybenzoate:
To a solution of methyl 4-bromo-2-hydroxybenzoate (3 g, 12.98 mmol) in DMF (15 ml) was added K2C03 (2.153 g, 15.58 mmol), Kl (0.022 g, 0.130 mmol) and 2-iodopropane (1 .95 ml, 19.48 mmol). After stirring at 70 °C for 18 h, the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo to afford the title compound as a brown liquid (3.0 g, 85%).
1H NMR (500 MHz, CDCI3): 7.57-7.56 (1 H, d), 7.05 (1 H, d), 7.03-7.01 (1 H, dd), 4.53-4.46 (1 H, sept), 3.79 (3H, s), 1 .32-1 .30 (6H, d).
Step 2: 2-(4-Bromo-2-isopropoxyphenyl)propan-2-ol: To a solution of methylmagnesium chloride (3M in THF, 4.88 ml, 14.65 mmol) in THF (4 ml) was slowly added a solution of methyl 4-bromo-2-isopropoxybenzoate (1 g, 3.66 mmol) in THF (4 ml). After stirring at 60 °C for 7 h, the mixture was added to 2N HCI (10 ml) and then partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were dried (MgS04) and concentrated in vacuo to afford the title compound as a brown oil (1 .0 g, 100%).
1H NMR (500 MHz, CDCI3): 7.19-7.18 (1 H, d), 7.05-7.02 (2H, m), 4.72-4.65 (1 H, sept), 1 .58 (6H, s), 1 .43-1 .42 (6H, d). Step 3: tert-butyl 4-(6-(4-(2-hydroxypropan-2-yl)-3-isopropoxyphenyl)pyridin-2- yl)piperazine- 1 -carboxylate:
To a solution of 2-(4-bromo-2-isopropoxyphenyl)propan-2-ol (250 mg, 0.915 mmol) in DMF (3 ml) was added 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1 ,3,2-dioxaborolane) (349 mg, 1 .373 mmol) and potassium acetate (269 mg, 2.75 mmol)). Nitrogen was bubbled through the mixture for 10 min and PdCI2(dppf)CH2CI2 (74.7 mg, 0.092 mmol) was added. Nitrogen was bubbled for a further 5 min, and then the mixture was heated at 80 °C for 6 h. After cooling to RT, tert-butyl 4-(6-bromopyridin-2-yl)piperazine-1 - carboxylate (209 mg, 0.610 mmol), PdCI2(dppf)CH2CI2 (74.7 mg, 0.092 mmol) and 2M Na2C03 (aq) (1 .830 ml, 3.66 mmol) were added. After stirring at 80 °C for 18 h the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 30 % EtOAc in cyclohexane) to afford the title compound as an off- white solid (129 mg, 46%).
1H NMR (500 MHz, CDCI3): 7.55 (1 H, d), 7.51 -7.47 (1 H, dd), 7.41 -7.39 (1 H, dd), 7.29- 7.27 (1 H, d), 7.03-7.01 (1 H, d), 6.55-6.53 (1 H, d), 4.81 -4.76 (1 H, sept), 3.57-3.48 (8H, m), 1 .56 (6H, s), 1 .43 (9H, s), 1 .41 -1 .40 (6H, d).
Step 4: 1-(6-(3-isopropoxy-4-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride: To a solution of tert-butyl 4-(6-(4-(2-hydroxypropan-2-yl)-3-isopropoxyphenyl)pyridin-2- yl)piperazine-1 -carboxylate (0.1 g, 0.219 mmol) in DCM (1 ml) at 0 °C, was added triethylsilane (0.1 15 ml, 0.720 mmol) and borontrifluoride diethyletherate (0.088 ml, 0.713 mmol). The mixture was stirred for 18 h, gradually warming to RT, then quenched with sat. aq. NaHC03 (1 ml). The two layers were separated and the aqueous layer was extracted with DCM. The combined organic extracts were washed with water, filtered through a phase separator cartridge and concentrated in vacuo. The residue was dissolved in 4M HCI in dioxane (2 ml). The reaction was stirred for 5 min at RT. Et20 (approx. 2 ml) was added to the resultant suspension, and the mixture slurried for 15 min. The supernatant liquid was removed by pipette. The Et20 slurry was repeated twice and the residue was dissolved in MeOH (1 ml) / H20 (1 ml) and lyophilized o/n to afford the title compound as a yellow solid (72.6 mg, 80%).
LCMS (Method F): RT = 1 .01 min, M+H+ = 340; 1H NMR (500 MHz, MeOH-d4): 8.02-7.99 (1 H, dd), 7.31 -7.20 (5H, m), 4.72-4.66 (1 H, sept), 3.94-3.92 (4H, m), 3.40-3.36 (4H, m), 3.33-3.25 (1 H, sept), 1 .29-1 .28 (6H, d), 1 .15-1 .13 (6H, d).
Example 33: 1 -(6-(4-isopropoxy-3-isopropylphenyl)pyridin-2-yl)piperazine
dihvdrochloride
Figure imgf000065_0001
Step 1: Methyl 5-bromo-2-isopropoxybenzoate:
Following the procedure for methyl 4-bromo-2-isopropoxybenzoate, methyl 5-bromo-2- hydroxybenzoate (3 g, 12.98 mmol)) was reacted to afford the title compound as a colourless liquid (3.14 g, 89%).
1H NMR (500 MHz, CDCI3): 7.80-7.79 (1 H, d), 7.44-7.42 (1 H, dd), 6.80-6.78 (1 H, d), 4.51 -4.43 (1 H, sept), 3.81 (3H, s), 1 .30-1 .28 (6H, d).
Step 2: 2-(5-bromo-2-isopropoxyphenyl)propan-2-ol:
Following the procedure for 2-(4-Bromo-2-isopropoxyphenyl)propan-2-ol, methyl 5- bromo-2-isopropoxybenzoate (1 g, 3.66 mmol)) was reacted to afford the title compound as a yellow oil (0.93 g, 93%).
1H NMR (500 MHz, CDCI3): 7.43-7.42 (1 H, d), 7.31 -7.27 (1 H, dd), 6.78-6.76 (1 H, d), 4.71 -4.64 (1 H, sept), 1 .58 (6H, s), 1 .41 -1 .40 (6H, d). Step 3: tert-butyl 4-(6-(3-(2-hydroxypropan-2-yl)-4-isopropoxyphenyl)pyridin-2- yl)piperazine- 1 -carboxylate: Following the procedure for tert-butyl 4-(6-(4-(2-hydroxypropan-2-yl)-3- isopropoxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate, 2-(5-bromo-2- isopropoxyphenyl)propan-2-ol (250 mg, 0.915 mmol) was reacted to afford the title compound as an off-white foam (139 mg, 50%).
1H NMR (500 MHz, CDCI3): 7.92-7.91 (1 H, d), 7.79-7.77 (1 H, dd), 7.49-7.46 (1 H, dd), 7.00-6.99 (1 H, d), 6.90-6.89 (1 H, d), 6.51 -6.49 (1 H, d), 4.74-4.68 (1 H, sept), 3.57-3.50 (8H, m), 1 .60 (6H, s), 1 .42 (9H, s), 1 .38-1 .37 (6H, d).
Step 4: 1-(6-(4-isopropoxy-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride: Following the procedure for 1 -(6-(3-isopropoxy-4-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride, tert-butyl 4-(6-(3-(2-hydroxypropan-2-yl)-4-isopropoxyphenyl)pyridin-2- yl)piperazine-1 -carboxylate (0.1 1 g, 0.241 mmol) was reacted to afford the title compound as a yellow solid (78 mg, 78%).
LCMS (Method F): RT = 0.98 min, M+H+ = 340; 1H NMR (500 MHz, MeOH-d4): 8.20-8.17 (1 H, dd), 7.69-7.68 (2H, m), 7.37-7.34 (2H, m), 7.18-7.16 (1 H, d), 4.84-4.77 (1 H, sept), 4.07-4.04 (4H, t), 3.52-3.50 (4H, t), 3.44-3.35 (1 H, sept), 1 .42-1 .41 (6H, d), 1 .31 -1 .30 (6H, d). Example 34: 2-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6 ,7,8-tetrahydronaphthalen-2- vDpyrazine dihydrochloride
Figure imgf000066_0001
Step 1: tert-butyl 4-(6-chloropyrazin-2-yl)piperazine-1 -carboxylate;
A solution of 2,6 dichloropyrazine (1 .0 g, 6.71 mmol), tert-butyl 4-(6-chloropyrazin-2- yl)piperazine-1 -carboxylate (1 .25 g, 6.71 mmol) and triethylamine (0.68 g, 6.71 mmol) in acetonitrile (10 ml) was heated in a microwave vial at 120 °C for 2 h. The solution was then diluted with ethyl acetate, washed with a saturated aqueous solution of ammonium chloride, dried over magnesium sulphate, filtered and concentrated in vacuo to afford the title compound (1 .86 g, 93%).
1H NMR (500 MHz, CDCI3): 7.98 (s, 1 H), 7.83 (s, 1 H), 3.61 (m, 4H), 3.56 (s, 4H), 1 .48 (s, 9H). Step 2: tert-butyl 4-(6-(5,5 -tetramethyl-5 frtetrahydronaphthalen-2-yl)pyrazin-2- yl)piperazine- 1 -carboxylate:
A solution of 4,4,5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)-1 ,3,2-dioxaborolane (0.4 g, 1 .27 mmol), tert-butyl 4-(6-chloropyrazin-2-yl)piperazine- 1 -carboxylate (0.19 g, 0.636 mmol), 2N sodium carbonate (2 ml) and
PdCI2(dppf) CH2CI2 (0.037 g, 0.045 mmol) was heated at 140 °C under microwave irradiation for 2h. After cooling to room temperature, the solution was diluted with DCM and filtered through a phase separator cartridge. The filtrate was concentrated in vacuo. The residue was purified by flash chromatography using a Biotage SP4 instrument to afford the title compound (0.18 g, 64%).
1H NMR (500 MHz, CDCI3): 8.35 (s, 1 H), 8.07 (s, 1 H), 7.93 (s, 1 H), 7.75 (d, 1 H), 7.43 (d, 1 H), 3.69 (m, 4H), 3.62 (m, 4H), 1 .74 (s, 4H), 1 .52 (s, 9H), 1 .37 (s, 6H), 1 .33 (s, 6H). Step 3 : 2-(piperazin-1-yl)-6-(5!5A8-tetramethyl-5^ ,8-tetrahydronaphthalen-2- yl)pyrazine dihydrochloride:
Te -butyl 4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrazin-2- yl)piperazine-1 -carboxylate (0.15 g, 0.33 mmol) was added to a solution of 4N HCI in dioxane (2 ml). The resulting solution was heated at 40 °C for 30 min then cooled to room temperature. The resulting precipitate was filtered and dried at ambient to give the title compound (0.1 18 g, 84%).
LCMS (Method F): RT = 1 .06 min, M+H+ = 361 ; 1H NMR (500 MHz, DMSO-d6): 9.57 (s, 2H), 8.56 (s, 1 H), 8.37 (s, 1 H), 7.99 (s, 1 H), 7.81 (d, 1 H), 7.45 (s, 1 H), 3.95 (s, 4H), 3.2 (s, 4H), 1 .68 (s, 4H), 1 .32 (s, 6H), 1 .27 (s, 6H).
Example 35: 1 -(5-(5,5,8,8-tetramethyl-5,6 ,7,8-tetrahvdronaphthalen-2-yl)pyridin-3- vDpiperazine trihvdrochloride
Figure imgf000067_0001
Step 1: tert-butyl 4-(5-bromopyridin-3-yl)piperazine-1 -carboxylate: A solution of 3,5-dibromopyridine (0.6 g, 2.53 mmol), tert-butyl piperazine-1 -carboxylate (0.71 g, 3.8 mmol), Pd2dba3 (0.07 g, 0.076 mmol), cesium carbonate (0.82 g, 2.53 mmol) and xantphos (0.132 g, 0.228 mmol) in dioxane (10 ml) was heated at 1 15 °C for 15 h. The solution was cooled to room temperature and concentrated in vacuo. The residue was redissolved in DCM and water. The organic layer was filtered through a phase separator cartridge. The filtrate was concentrated in vacuo. The resulting residue was purified by chromatography using a Biotage SP4 instrument to afford the title compound (0.44 g, 51 %).
1H NMR (500 MHz, CDCI3): 8.21 (s, 1 H), 8.15 (s, 1 H), 7.30 (s, 1 H), 3.59 (m, 4H), 3.19 (m, 4H), 1 .49 (s, 9H).
Step 2: tert-butyl 4-(5-(5,5^ etra et y\-5^ etra ydronap t a\en-2-y\)pyridin-3- yl)piperazine- 1 -carboxylate:
Following the procedure for tert-butyl 4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperazine-1 -carboxylate, tert-butyl 4-(5- bromopyridin-3-yl)piperazine-1 -carboxylate (0.2 g, 0.58 mmol) was reacted to give the title compound (0.15 g, 57%).
1H NMR (500 MHz, CDCI3): 8.34 (s, 1 H), 8.27 (s, 1 H), 7.47 (s, 1 H), 7.40 (s, 1 H), 7.31 (m, 2H), 3.62 (m, 4H), 3.22 (m, 4H), 1 .69 (s, 4H), 1 .43 (s, 9H), 1 ,34 (s, 6H). 1 .32 (s, 6H).
Step 3 : ^-(5-(5,5β etra et y\-5β etra ydronap t a\en-2-y\)pyridin-3- yl)piperazine trihydrochloride:
Following the procedure for 2-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyrazine dihydrochloride, tert-butyl 4-(5-(5,5,8,8-tetramethyl- 5, 6,7,8-tetrahydronaphthalen-2-yl)pyridin-3-yl)piperazine-1 -carboxylate (0.15 g, 0.33 mmol) was reacted to give the title compound (0.107 g, 76%).
LCMS (Method F): RT = 0.94 min, M+H+ = 350; 1H NMR (500 MHz, DMSO-d6): 9.73 (s, 2H), 8.57 (s, 1 H), 8.47 (s, 1 H), 8.16 (s, 1 H), 7.76 (s, 1 H), 7.60 (d, 1 H), 7.50 (s, 1 H), 3.81 (m, 4H), 3.39 (m, 4H), 1 .68 (s, 4H), 1 .34 (s, 6H), 1 .32 (s, 6H).
Example 36: 1 -(5-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-3- yl)piperidin-4-amine trihydrochloride
Figure imgf000069_0001
Step 1: tert-butyl 1-(5-bromopyridin-3-yl)piperidin-4-ylcarbamate:
Following the procedure for tert-butyl 4-(5-bromopyridin-3-yl)piperazine-1 -carboxylate,
3,5-dibromopyridine (0.6 g, 2.53 mmol) was reacted to give the title compound (0.44 g,
49%).
1H NMR (500 MHz, CDCI3): 8.20 (s, 1 H), 8.09 (s, 1 H), 7.28 (s, 1 H), 4.50 (br s, 1 H), 3.63 (m, 3H), 2.92 (m, 2H), 2.07 (m, 2H), 1 .52 (m, 2H), 1 .46 (s, 9H).
Step 2 : tert-butyl 1-(5-(5 -tetramethyl-5fi -tetrahydronaphthalen-2-yl)pyrM^ yl)piperidin-4-ylcarbamate:
Following the procedure for tert-butyl 4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperazine-1 -carboxylate, tert-butyl 1-(5- bromopyridin-3-yl)piperidin-4-ylcarbamate (0.2 g, 0.56 mmol) was reacted to give the title compound (0.125 g, 48%).
1H NMR (500 MHz, CDCI3): 8.29 (s, 1 H), 8.26 (s, 1 H), 7.47 (s, 1 H), 7.40 (s, 1 H), 7.31 (m, 2H), 4.50 (br s, 1 H), 3.70 (m, 3H), 2.95 (m, 2H), 2.10 (m, 2H), 1 ,73 (s, 4H), 1 .54 (m, 2H), 1 .43 (s, 9H), 1 ,34 (s, 6H). 1 .32 (s, 6H).
Step 3 : 1-(5-(5 -tetramethyl-5fi -tetrahydronaphthalen-2-yl)pyrti^^^
yl)piperidin-4-amine trihydrochloride:
Following the procedure for 2-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyrazine dihydrochloride, tert-butyl 1 -(5-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-3-yl)piperidin-4-ylcarbamate (0.10 g, 0.216 mmol) was reacted to give the title compound (0.082 g, 88%).
LCMS (Method F): RT = 0.78 min, M+H+ = 364; 1H NMR (500 MHz, DMSO-d6): 8.48 (s, 1 H), 8.42 (s, 1 H), 8.37 (s, 2H), 8.1 1 (s, 1 H), 7.73 (s, 1 H), 7.59 (d, 1 H), 7.50 (d, 1 H), 4.13 (m, 2H), 3.40 (m, 1 H), 3.07 (m, 2H), 2.06 (m, 2H), 1 .71 (s, 4H), 1 .70 (m, 2H), 1 .33 (s, 6H), 1 .28 (s, 6H).
Example 37 : 4-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDpyrimidine dihydrochloride
Figure imgf000070_0001
Step 1: tert-butyl 4-(6-chloropyrimidin-4-yl)piperazine- 1-carboxylate:
A solution of 4,6-dichloropyrimidine (0.5 g, 3.36 mmol), tert-butyl piperazine-1 - carboxylate (0.69 g, 3.69 mmol) and triethylamine (0.34 g, 3.36 mmol) in acetonitrile (10ml) was heated in a microwave vial at 120 °C for 2 h. The solution was then diluted with ethyl acetate, washed with a saturated solution of ammonium chloride, dried over magnesium sulphate, filtered and concentrated in vacuo to give the title compound (0.82 g, 91 %).
1H NMR (500 MHz, CDCI3): 8.39 (s, 1 H), 6.50 (s, 1 H), 3.65 (m, 4H), 3.52 (m, 4H), 1 .48 (s, 9H).
Step 2: tert-butyl 4-(6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-4- yl)piperazine- 1 -carboxylate:
Following the procedure for tert-butyl 4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperazine-1 -carboxylate, tert-butyl 4-(6- chloropyrimidin-4-yl)piperazine-1 -carboxylate (0.19 g, 0.63 mmol) was reacted to give the title compound (0.20 g, 72%).
1H NMR (500 MHz, CDCI3): 8.62 (s, 1 H), 7.87 (s, 1 H), 7.58 (d, 1 H), 7.32 (d, 1 H), 6.75 (s, 1 H), 3.64 (m, 4H), 3.49 (m, 4H), 1 .64, (s, 4H), 1 .43 (s, 9H), 1 ,28 (s, 6H), 1 .26 (s, 6H).
Step 3 : 4-(piperazin-1-yl)-6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidine dihydrochloride:
Following the procedure for 2-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyrazine dihydrochloride, tert-butyl 4-(6-(5,5,8,8-tetramethyl- 5^ ^-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperazine-1-carboxylate (0.10 g, 0.222 mmol) was reacted to give the title compound (0.086 g, 92%).
LCMS (Method F): RT = 0.86 min, M+H+ = 351 ; 1H NMR (500 MHz, DMSO-d6): 9.86 (s, 2H), 8.84 (s, 1 H), 8.47 (s, 1 H), 8.06 (s, 1 H), 7.87 (d, 1 H), 7.58 (d, 1 H), 7.50 (s, 1 H), 4.22 (m, 4H), 3.27 (m, 4H), 1 .69 (s, 4H), 1 .35 (s, 6H), 1 .29 (s, 6H). Example 38: 1 -(6-(5,5,8,8-tetramethyl-5,6 J,8-tetrahvdronaphthalen-2-yl)pyrimidin-4- yl)piperidin-4-amine dihydrochloride
Figure imgf000071_0001
Step 1: tert-butyl 1-(6-chloropyrimidin-4-yl)piperidin-4-ylcarbamate:
Following the procedure for tert-butyl 4-(6-chloropyrimidin-4-yl)piperazine-1 -carboxylate,
4,6-dichloropyrimidine (0.5 g, 3.36 mmol) was reacted to give the title compound (0.97 g,
93%).
1H NMR (500 MHz, CDCI3): 8.36 (s, 1 H), 6.52 (s, 1 H), 4.54 (br s , 1 H), 4.30 (br s, 2H), 3.75 (br s, 1 H), 3.04 (m, 2H), 2.05 (m, 2H), 1 .48 (s, 9H), 1 .39 (m, 2H).
Step 2: tert-butyl 1-(6-(5,5,8,8-tetramethyl-5fi ,8-tetrahydronaphthalen-2-yl)pyrimidin-4- yl)piperidin-4-ylcarbamate:
Following the procedure for tert-butyl 4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperazine-1 -carboxylate, tert-butyl (1 -(6- chloropyrimidin-4-yl)piperidin-4-yl)carbamate (0.2 g, 0.63 mmol) was reacted to give the title compound (0.19 g, 64%).
1H NMR (500 MHz, CDCI3): 8.70 (s, 1 H), 7.96 (s, 1 H), 7.67 (d, 1 H), 7.41 (d, 1 H), 6.86 (s, 1 H), 4.40-4.50 (m, 3H), 3.75 (br s, 1 H), 3.1 1 (m, 2H), 2.10 (m, 2H) 1 .68, (s, 4H), 1 .43 (s, 9H), 1 .39 (M, 1 H), 1 ,30 (s, 6H), 1 .28 (s, 6H).
Step 3 : 1-(6-(5,5,8,8-tetramethyl-5fi ,8-tetrahydronaphthalen-2-yl)pyrimidin-4- yl)piperidin-4-amine dihydrochloride:
Following the procedure for 2-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyrazine dihydrochloride tert-butyl 1 -(6-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperidin-4-ylcarbamate (0.1 10 g, 0.237mmol) was reacted to give the title compound (0.082 g, 79%).
LCMS (Method F): RT = 0.86 min, M+H+ = 351 ; 1H NMR (500 MHz, DMSO-d6): 8.78 (s, 1 H), 8.58 (s, 2H), 8.47 (s, 1 H), 8.05 (s, 1 H), 7.87 (d, 1 H), 7.58 (d, 1 H), 7.47 (s, 1 H), 4.70 (br s, 2H), 3.3-3.7 (m, 4H), 2.15 (m, 2H), 1 .69 (s, 4H), 1 .68 (m, 1 H), 1 .35 (s, 6H), 1 .29 (s, 6H), 1 .27 (m, 1 H). Example 39: 1 -(6-(5,5,8,8-tetramethyl-5,6 ,7,8-tetrahvdronaphthalen-2-yl)pyrazin-2- yl)piperidin-4-amine trihydrochloride
Figure imgf000072_0001
Step 1: tert-butyl 1-(6-chloropyrazin-2-yl)piperidin-4-ylcarbamate:
Following the procedure for tert-butyl 1 -(6-chloropyrazin-2-yl)piperidin-4-ylcarbamate,
2,6-dichloropyrazine (1 g, 6.71 mmol) was reacted to give the title compound (1 .91 g,
91 %).
1H NMR (500 MHz, CDCI3): 8.0 (s, 1 H), 7.80 (s, 1 H), 4.53 (brs, 1 H), 4.25 (m, 2H), 3.70 (m, 1 H), 3.07 (m, 2H), 2.09 (m, 2H), 1 .42 (s, 9H), 1 .40 (m, 2H).
Step 2: tert-butyl 1-(6-(5^^-tetramethyl-5^ ^-tetrahydronaphthalen-2-yl)pyrazin-2- yl)piperidin -4 -ylcarbamate
Following the procedure for tert-butyl 4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperazine-1 -carboxylate, tert-butyl 1 -(6- chloropyrazin-2-yl)piperidin-4-ylcarbamate (0.15g, 0.48mmol) was reacted to give the title compound (0.15 g, 71 %).
1H NMR (500 MHz, CDCI3): 8.20 (s, 1 H), 7.96 (s, 1 H), 7.80 (s, 1 H), 7.62 (d, 1 H), 7.31 (d, 1 H), 4.66 (brs, 1 H), 4.25 (m, 1 H), 4.15 (m, 1 H), 3.65 (m, 1 H), 2.98 (m, 2H), 1 .97 (m, 2H), 1 .42 (s, 9H), 1 .40 (m, 2H).
Step 3 : 1-(6-(5^^-tetramethyl-5^ ^-tetrahydronaphthalen-2-yl)pyrazin-2- yl)piperidin-4-amine dihydrochloride:
Following the procedure for 2-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyrazine dihydrochloride tert-butyl 1 -(6-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperidin-4-ylcarbamate (0.120 g, 0.258mmol) was reacted to give the title compound (0.092 g, 81 %).
LCMS (Method F): RT = 1 .03 min, M+H+ = 365; 1H NMR (500 MHz, DMSO-d6): 8.52 (s, 1 H), 8.47 (s, 2H), 8.47 (s, 1 H), 8.38 (s, 1 H), 8.01 (s, 1 H), 7.84 (d, 1 H), 7.45 (d, 1 H), 4.51 (m, 2H), 3.36 (m, 1 H), 3.15 (m, 2H), 2.09 (m, 2H), 1 .62 (s, 4H), 1 .60-1 .65 (m, 2H), 1 .30 (s, 6H), 1 .26 (s, 6H).
Example 40: 1 -(2-methoxyethyl)-4-(6-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen- 2-yl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000073_0001
To a solution of 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazine (200 mg, 0.57 mmol) in ethanol (5 ml) and triethylamine (0.24 ml, 1 .72 mmol) in a sealable microwave vial was added 1 -chloro-2-methoxyethane (0.081 g, 0.86 mmol). The resulting mixture was heated for 2 hours at 160 °C, cooled to room temperature, concentrated in vacuo and poured into water (100 ml). The product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo. The resultant residue was subjected to flash chromatography (KP-NH, Si02, gradient 0 to 50 % ethyl acetate in cyclohexane) to afford the title compound as a yellow oil. The oil was dissolved in dioxane and 4M HCI in dioxane was added. The solution was stirred at room temperature for 5 min. before being concentrated in vacuo. The residue was triturated with diethyl ether to give the target compound as a yellow solid (121 mg, 52%).
LCMS (Method F): RT = 1 .21 min, M+H+ = 408; 1H NMR (500 MHz, MeOH-d4): 7.91 -7.92 (2H, m), 7.70 (1 H, dd), 7.49 (1 H, d), 7.32 (1 H, d), 7.1 1 (1 H, d), 4.54-4.56 (2H, m), 3.77- 3.84 (4H, m), 3.55-3.59 (2H, m), 3.47 (3H, s), 3.31 -3.35 (4H, m), 1 .78 (4H, s), 1 .35 (6H, s), 1 .33 (6H, s). Example 41 : 3-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)propan-1 -ol
Figure imgf000074_0001
3-(4-(6-(5,5,8,8-tetramethyl-5fi ,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)pfa yl)propan-1-ol:
To a solution of 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazine (200 mg, 0.57 mmol) in ethanol (5 ml) and triethylamine (0.24 ml, 1 .72 mmol) in a sealable microwave vial was added 3-bromopropan-1 -ol (0.1 19 g, 0.858 mmol). The resulting mixture was heated for 2 hours at 160 °C, cooled to room temperature, concentrated in vacuo and poured into water (100 ml). The product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo. The resultant residue was subjected to flash chromatography (KP-NH, Si02, gradient 0 to 50 % ethyl acetate in cyclohexane) to afford the title compound as a yellow solid (1 16 mg, 50%).
LCMS (Method F): RT = 1 .13 min, M+H+ 408; 1 H NMR (500 MHz, CDCI3): 7.92 (1 H, d), 7.72 (1 H, d), 7.53 (1 H, dd), 7.36 (1 H, d), 7.08 (1 H, d), 6.57 (1 H, d), 5.1 1 (1 H, bs) 3.84 (2H, dd), 3.64-3.66 (4H, m), 2.67-2.69 (6H, m), 1 .78 (2H, dd), 1 .71 (4H, s), 1 .34 (6H, s), 1 .30 (6H, s).
Example 42: A/,A/-dimethyl-2-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2- yl)pyridin-2-yl)piperazin-1 -yl)acetamide
Figure imgf000074_0002
N, N-dimethyl-2-(4-(6-(5, 5, 8, 8-tetramethyl-5, 6, 7, 8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin- 1 -yl)acetamide:
To a solution of 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazine (200 mg, 0.57 mmol) in ethanol (5 ml) and triethylamine (0.24 ml, 1 .72 mmol) in a sealable microwave vial was added 2-chloro-/V,/V-dimethylacetamide (0.104 g, 0.858 mmol). The resulting mixture was heated for 2 hours at 160 'Ό, cooled to room temperature, concentrated in vacuo and poured into water (100 ml). The product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo. The resultant residue was subjected to flash chromatography (KP-NH, Si02, gradient 0 to 50 % ethyl acetate in cyclohexane) to afford the title compound as an off white solid (135 mg, 54%).
LCMS (Method F): RT = 1 .17 min, M+H+ = 435; 1H NMR (500 MHz, CDCI3): 7.93 (1 H, d), 7.72 (1 H, dd), 7.53 (1 H, dd), 7.36 (1 H, d), 7.06 (1 H, d), 6.57 (1 H, d), 3.67 (4H, dd), 3.23 (2H, s), 3.10 (3H, s), 2.97 (3H, s), 2.67 (4H, dd), 1 .71 (4H, s), 1 .34 (6H, s), 1 .30 (6H, s).
Example 43: (rac)-1 -methoxy-3-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen- 2-yl)pyridin-2-yl)piperazin-1 -yl)propan-2-ol dihvdrochloride
Figure imgf000075_0001
To a solution of 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazine (100 mg, 0.29 mmol) in ethanol (3 ml) in a sealable microwave vial was added (rac)-2-(methoxymethyl)oxirane (0.039 ml, 0.429 mmol). The resulting mixture was heated for 1 hours at 120 'Ό, cooled to room temperature, concentrated in vacuo and poured into water (100 ml). The product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and
concentrated in vacuo. The resultant residue was subjected to flash chromatography (KP-NH, Si02, gradient 0 to 50 % ethyl acetate in cyclohexane) to afford the title compound as a yellow oil. The oil was dissolved in dioxane and 4M HCI in dioxane was added. The solution was stirred at room temperature for 5 min. before being
concentrated in vacuo. The residue was triturated with diethyl ether to give the target compound as an off white solid (72 mg, 58%).
LCMS (Method F): RT = 1 .15 min, M+H+ = 438; 1H NMR (500 MHz, MeOH-d4): 7.92 (1 H, d), 7.91 (1 H, d), 7.70 (1 H, dd), 7.49 (1 H, d), 7.32 (1 H, d), 7.1 1 (1 H, d), 4.52 (2H, dd), 4.25-4.31 (1 H, m), 3.81 -3.94 (2H, m), 3.66-3.70 (4H, m), 3.42 (3H, s), 3.31 -3.35 (4H, m), 1 .78 (4H, s), 1 .37 (6H, s), 1 .34 (6H, s).
Example 44: A/.A/-dimethyl-2-(4-(6-(5.5.8.8-tetramethyl-5.6.7.8-tetrahvdronaphthalen-2- yl)pyridin-2-yl)piperazin-1 -vDethanamine trihydrochloride
Figure imgf000076_0001
To a solution of /V,/V-dimethyl-2-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen- 2-yl)pyridin-2-yl)piperazin-1 -yl)acetamide (63 mg, 0.15 mmol) in THF (1 ml) was added lithium aluminiumhydride (0.435 ml, 0.435 mmol). The resulting mixture was heated for 2 hours at 80 °C, cooled to room temperature and quenched by dropwise addition of 40% aq. KOH. The resulting suspension was heated at 80 °C for 30 min. and poured into water (100 ml). The product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo. The resultant residue was subjected to flash chromatography (KP-NH, Si02, gradient 0 to 50 % ethyl acetate in cyclohexane) to afford the title compound as a clear oil. The oil was dissolved in dioxane and 4M HCI in dioxane was added. The solution was stirred at room temperature for 5 min. before being concentrated in vacuo. The residue was triturated with diethyl ether to give the target compound as a yellow solid (24 mg, 34%).
LCMS (Method F): RT = 1 .01 min, M+H+ = 421 ; 1H NMR (500 MHz, MeOH-d4): 8.03 (1 H, dd), 7.85 (1 H, d), 7.65 (1 H, dd), 7.55 (1 H, d), 7.32 (1 H, d), 7.25 (1 H, d), 3.69-3.75 (4H, m), 3.60-3.66 (2H, m), 3.48-3.54 (2H, m), 3.31 -3.35 (4H, m), 3.04 (6H, s), 1 .78 (4H, s), 1 .38 (6H, s), 1 .35 (6H, s).
Example 45: 3-hvdroxy-2-(hvdroxymethyl)-2-methyl-1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahvdronaphthalen-2-yl)pyridin-2-yl)piperazin-1 -yl)propan-1 -one hydrochloride
Figure imgf000077_0001
To a solution of 3-hydroxy-2-(hydroxymethyl)-2-methylpropanoic acid (0.032 g, 0.238 mmol) in DMF (2 ml) was added COMU (0.102 g, 0.238 mmol) and Oxyma (0.034 g, 0.238 mmol). The resulting solution was stirred at room temperature for 10 minutes. 1 - (6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperazine (63 mg, 0.15 mmol) and DIPEA (0.083 ml, 0.477 mmol) were added, the resulting solution was stirred at room temperature for 16 hours and poured into water (100 ml). The product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 50 % ethyl acetate in
cyclohexane) to afford the title compound as a clear oil. The oil was dissolved in dioxane and 4M HCI in dioxane was added. The solution was stirred at room temperature for 5 min. before being concentrated in vacuo. The residue was triturated with diethyl ether to give the target compound as a yellow solid (43 mg, 38%).
LCMS (Method F): RT = 1 .62 min, M+H+ = 466; 1H NMR (500 MHz, MeOH-d4): 7.99 (1 H, dd), 7.76 (1 H, dd), 7.62 (1 H, dd), 7.40 (1 H, d), 7.18 (1 H, d), 6.75 (1 H, d), 3.84-3.86 (4H, m), 3.76 (4H, s), 3.21 -3.23 (4H, m), 1 .74 (4H, s), 1 .35 (6H, s), 1 .31 (6H, s) 1 .25 (3H, s).
Example 46: 4-(piperidin-4-yloxy)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDpyrimidine di(trifluoroacetate)
Figure imgf000077_0002
Step 1: tert-butyl 4-((2-chloropyrimidin-4-yl)oxy)piperidine- 1-carboxylate:
To a solution of te/t-butyl 4-hydroxypiperidine-1 -carboxylate (0.675 g, 3.36 mmol) in THF (15 ml) was added potassium te/t-butoxide (0.414 g, 3.69 mmol). The resulting solution was stirred at room temperature for 10 min., cooled to 0 'Ό and 2,4-dichloropyrimidine (0.5 g, 3.36 mmol) in THF (15 ml) was added. The resulting solution was stirred at room temperature for 16 hours and poured into water (100 ml). The product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo to give a 1 :1 mixture of te/t-butyl 4-((4- chloropyrimidin-2-yl)oxy)piperidine-1 -carboxylate and te/t-butyl 4-((2-chloropyrimidin-4- yl)oxy)piperidine-1 -carboxylate as a yellow oil which was used without further purification (884 mg, 84%).
Step 2: tert-butyl 4-((2-(5!5A8-tetramethyl-5^ !8-tetrahydronaphthalen-2-yl)pyrim 4-yl)oxy)piperidine- 1 -carboxylate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, a mixture of te/t-butyl 4-((2-chloropyrimidin-4-yl)oxy)piperidine-1 - carboxylate and te/t-butyl 4-((4-chloropyrimidin-2-yl)oxy)piperidine-1 -carboxylate (0.44 g, 1 .40 mmol) and 4,4,5, 5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen- 2-yl)-1 ,3,2-dioxaborolane (0.44 g, 1 .40 mmol) were reacted to afford the title compound as a clear oil (0.22 g, 34%).
LCMS (Method F): RT = 2.07 min, M+H+ = 466.
Step 3: 4-(piperidin-4-yloxy)-2-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidine di(trifluoroacetate):
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine, te/t-butyl 4-((2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidin-4-yl)oxy)piperidine-1 -carboxylate (100 mg , 0.23 mmol) was reacted to afford the title compound as a white solid (67 mg, 78%).
LCMS (Method F): RT = 1 .01 min, M+H+ = 366; 1H NMR (500 MHz, MeOH-d4): 8.57 (1 H, d), 8.37 (1 H, d), 8.1 1 (1 H, dd), 7.45 (1 H, d), 6.80 (1 H, d), 5.62-5.66 (1 H, m), 3.45-3.52 (2H, m), 3.45-3.52 (2H, m), 2.35-2.37 (2H, m), 2.21 -2.23 (2H, m), 1 .78 (4H, s), 1 .37 (6H, s), 1 .34 (6H, s). Example 47: 2-(piperidin-4-yloxy)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDpyrimidine di(trifluoroacetate)
Figure imgf000079_0001
Step 1: tert-butyl 4-((4-chloropyrimidin-2-yl)oxy)piperidine- 1 -carboxylate:
To a solution of tert-butyl 4-hydroxypiperidine-1 -carboxylate (0.675 g, 3.36 mmol) in THF (15 ml) was added potassium tert-butoxide (0.414 g, 3.69 mmol). The resulting solution was stirred at room temperature for 10 minutes then cooled to 0 °C and 2,4- dichloropyrimidine (0.5 g, 3.36 mmol) in THF (15 ml) added. The resulting solution was stirred at room temperature for 16 hours and poured into water (100 ml). The product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo to give a 1 :1 mixture of tert-butyl 4-((4-chloropyrimidin-2-yl)oxy)piperidine-1 -carboxylate and tert-butyl 4-((2- chloropyrimidin-4-yl)oxy)piperidine-1 -carboxylate as a yellow oil which was used without further purification (884 mg, 84%). Step 2: tert-butyl 4-((4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin- 2-yl)oxy)piperidine- 1 -carboxylate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, a mixture of tert-butyl 4-((2-chloropyrimidin-4-yl)oxy)piperidine-1 - carboxylate and tert-butyl 4-((4-chloropyrimidin-2-yl)oxy)piperidine-1 -carboxylate (0.44 g, 1 .40 mmol) and 4,4,5, 5-tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen- 2-yl)-1 ,3,2-dioxaborolane (0.44 g, 1 .40 mmol) were reacted to afford the title compound as a clear oil (0.27 g, 44%).
LCMS (Method F): RT = 1 .99 min, M+H+ = 466. Step 3: 2-(piperidin-4-yloxy)-4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidine di(trifluoroacetate):
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), tert-butyl 4-((4-(5,5, 8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidin-2-yl)oxy)piperidine-1 -carboxylate (100 mg , 0.23 mmol) was reacted to afford the title compound as a white solid (67 mg, 69%). LCMS (Method F): RT = 1 .01 min, M+H+ = 366; 1H NMR (500 MHz, MeOH-d4): 8.48 (1 H, d), 8.02 (1 H, d), 7.77 (1 H, d), 7.50 (1 H, d), 7.39 (1 H, d), 5.36-5.40 (1 H, m), 3.34-3.40 (2H, m), 3.18-3.21 (2H, m), 2.18-2.22 (2H, m), 2.09-2.12 (2H, m), 1 .66 (4H, s), 1 .26 (6H, s), 1 .22 (6H, s).
Example 48: 2-methyl-4-(piperazin-1 -yl)-6-(5,5, 8,8-tetramethyl-5, 6,7,8- tetrahvdronaphthalen-2-yl)pyrimidine di(trifluoroacetate)
Figure imgf000080_0001
Step 1: tert-butyl 4-(6-chloro-2-methylpyrimidin-4-yl)piperazine-1-carboxylate:
To a solution of 4,6-dichloro-2-methylpyrimidine (1 g, 6.13 mmol) in THF (50ml) was added DBU (0.93 ml, 6.13 mmol) and te/t-butyl piperazine-1 -carboxylate (1 .257 g, 6.75 mmol). The resulting solution was stirred at 70 °C for 16 hours and poured into water (100 ml). The product was extracted with ethyl acetate, the combined organic phases were dried over magnesium sulphate, filtered and concentrated in vacuo to give the title compound as a brown oil which was used without further purification (1 .7 g, 80%).
LCMS (Method F): RT = 1 .30 min, M+H+ = 313.
Step 2: tert-butyl 4-(2-methyl-6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidin-4-yl)piperazine- 1 -carboxylate:
Following the procedure for 6-(3-bromophenyl)-1 ,1 ,4,4-tetramethyl-1 ,2,3,4- tetrahydronaphthalene, tert-butyl 4-(6-chloro-2-methylpyrimidin-4-yl)piperazine-1 - carboxylate (0.15 g, 0.48 mmol) and 4,4, 5,5-tetramethyl-2-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)-1 ,3,2-dioxaborolane (0.15 g, 0.48 mmol) were reacted to afford the title compound as a clear oil (0.12 g, 54%).
LCMS (Method F): RT = 1 .33 min, M+H+ = 465.
Step 3: 2-(piperidin-4-yloxy)-4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidine di(trifluoroacetate): Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate), te/t-butyl 4-(2-methyl-6-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperazine-1 -carboxylate (120 mg , 0.26 mmol) was reacted to afford the title compound as a white solid (82 mg, 87%).
LCMS (Method F): RT = 0.76 min, M+H+ = 365; 1H NMR (500 MHz, MeOH-d4): 7.75 (1 H, d), 7.48-7.53 (2H, m), 7.13 (1 H, s), 4.17-4.20 (4H, m), 3.33 (4H, dd), 2.61 (3H, s), 1 .67 (4H, s), 1 .27 (6H, s), 1 .23 (6H, s).
Example 49: 1 -(6-(4-isobutoxy-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000081_0001
Step 1: 4-bromo-2-isopropylphenol:
To a solution of 2-isopropylphenol (5 ml, 37.2 mmol) in DCM (30 ml) was added bromine (1 .914 ml, 37.2 mmol) slowly during 15 min. The solution was stirred for 1 h and then concentrated in vacuo. The product was obtained as a red-brown liquid (8.29 g) and was used without further purification.
1H NMR (500 MHz, CDCI3): 7.31 (1 H, d), 7.19-7.17 (1 H, dd), 6.66-6.64 (1 H, d), 4.72 (1 H, br s), 3.23-3.15 (1 H, sept), 1 .27-1 .26 (6H, d).
Step 2: tert-butyl 4-(6-(4-hydroxy-3-isopropylphenyl)pyridin-2-yl)piperazine-1- carboxylate:
Following the procedure for tert-butyl 4-(6-(4-(2-hydroxypropan-2-yl)-3- isopropoxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate, 4-bromo-2-isopropylphenol (1 .5 g, 6.97 mmol) and tert-butyl 4-(6-bromopyridin-2-yl)piperazine-1 -carboxylate (1 .592 g, 4.65 mmol) were reacted to afford the title compound as a yellow solid (983 mg, 53 %). LCMS (Method F): RT = 1 .56 min, M+H+ = 398.
Step 3: tert-butyl 4-(6-(4-isobutoxy-3-isopropylphenyl)pyridin-2-yl)piperazine-1- carboxylate: Following the procedure for Methyl 4-bromo-2-isopropoxybenzoate, tert-butyl 4-(6-(4- hydroxy-3-isopropylphenyl)pyridin-2-yl)piperazine-1 -carboxylate (150 mg, 0.377 mmol) and 1 -bromo-2-methylpropane (0.062 ml, 0.566 mmol) were reacted to afford the title compound as a yellow solid (37.7 mg, 22%).
1H NMR (500 MHz, CDCI3): 7.86-7.85 (1 H, d), 7.80-7.78 (1 H, dd), 7.54-7.51 (1 H, dd), 7.08-7.07 (1 H, d), 6.88-6.86 (1 H, d), 6.54-6.53 (1 H, d), 3.79-3.78 (2H, d), 3.65-3.55 (8H, m), 3.41 -3.36 (1 H, sept), 2.17-2.10 (1 H, sept), 1 .49 (9H, s), 1 .29-1 .28 (6H, d), 1 .08-1 .06
(6H, d).
Step 4: 1-(6-(4-isobutoxy-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride: Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl 4-(6-(4-isobutoxy-3- isopropylphenyl)pyridin-2-yl)piperazine-1 -carboxylate (37.7 mg, 0.083 mmol) was reacted to afford the title compound as a yellow solid (20.9 mg, 59%).
LCMS (Method F): RT = 1 .08 min, M+H+ = 354; 1H NMR (500 MHz, MeOH-d4): 8.03-7.99 (1 H, dd), 7.76-7.72 (2H, m), 7.34-7.32 (1 H, d), 7.17-7.15 (1 H, d), 7.10-7.08 (1 H, d), 4.01 - 3.99 (4H, m), 3.89-3.88 (2H, d), 3.48-3.46 (4H, m), 3.46-3.40 (1 H, sept), 2.20-2.13 (1 H, sept), 1 .33-1 .31 (6H, d), 1 .13-1 .12 (6H, d).
Figure imgf000082_0001
Step 1: 4-bromo-2-(tert-butyl)phenol:
To a solution of 2-(tert-butyl)phenol (5 ml, 32.6 mmol) in DCM (30 ml) was added bromine (1 .67 ml; 32.6 mmol) slowly during 15 min. The solution was stirred for 1 h and then concentrated in vacuo. The product was obtained as a red-brown liquid (8.40 g) which was used without further purification. 1H NMR (500 MHz, CDCI3): 7.35 (1 H, d), 7.17-7.15 (1 H, dd), 6.55-6.54 (1 H, d), 4.77 (1 H, br s), 1 .38 (9H, s).
Step 2: tert-butyl 4-(6-(3-(tert-butyl)-4-hydroxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
Following the procedure for tert-butyl 4-(6-(4-(2-hydroxypropan-2-yl)-3- isopropoxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate, 4-bromo-2-(tert-butyl)phenol (1 .5 g, 6.55 mmol) and tert-butyl 4-(6-bromopyridin-2-yl)piperazine-1 -carboxylate (1 .494 g, 4.37 mmol) were reacted to afford the title compound as a yellow solid (658 mg, 37%). LCMS (Method F): RT = 1 .66 min, M+H+ = 412.
Step 3: tert-butyl 4-(6-(3-(tert-butyl)-4-isopropoxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
Following the procedure for Methyl 4-bromo-2-isopropoxybenzoate, tert-butyl 4-(6-(3- (tert-butyl)-4-hydroxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate (150 mg, 0.364 mmol) and 2-iodopropane (0.055 ml, 0.547 mmol) were reacted to afford the title compound as a yellow oil (60.0 mg, 36%).
1H NMR (500 MHz, CDCI3): 7.98-7.97 (1 H, d), 7.79-7.77 (1 H, dd), 7.54-7.51 (1 H, dd), 7.07-7.05 (1 H, d), 6.91 -6.89 (1 H, d), 6.54-6.52 (1 H, d), 4.73-4.69 (1 H, sept), 3.65-3.55 (8H, m), 1 .49 (9H, s), 1 .44 (9H, s), 1 .41 -1 .39 (6H, d).
Step 4: 1-(6-(3-(tert-butyl)-4-isopropoxyphenyl)pyridin-2-yl)piperazine dihydrochloride: Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl 4-(6-(3-(tert-butyl)-4- isopropoxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate (60 mg, 0.132 mmol) was reacted to afford the title compound as a yellow solid (35.6 mg, 63%).
LCMS (Method F): RT = 1 .10 min, M+H+ = 354; 1H NMR (500 MHz, MeOH-d4D): 8.03- 8.00 91 H, dd), 7.81 (1 H, d), 7.75-7.72 (1 H, dd), 7.33-7.32 (1 H, d), 7.17-7.16 (1 H, d), 7.14-7.13 (1 H, d), 4.00-3.98 (4H, m), 3.48-3.45 (4H, m), 1 .47 (9H, s), 1 .45-1 .44 (6H, d).
Example 51 ): 1 -(6-(3-(tert-butyl)-4-isobutoxyphenyl)pyridin-2-yl)piperazine
dihydrochloride
Figure imgf000084_0001
Step 1: tert-butyl 4-(6-(3-(tert-butyl)-4-isobutoxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
Following the procedure for Methyl 4-bromo-2-isopropoxybenzoate, tert-butyl 4-(6-(3- (tert-butyl)-4-hydroxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate (120 mg, 0.292 mmol) and 1 -bromo-2-methylpropane (0.048 ml, 0.437 mmol) were reacted to afford the title compound as a colourless oil (43.6 mg, 32%).
1H NMR (500 MHz, CDCI3): 7.99 (1 H, d), 7.79-7.77 (1 H, dd), 7.54-7.51 (1 H, dd), 7.07- 7.06 (1 H, d), 6.91 -6.90 (1 H, d), 6.54-6.53 (1 H, d), 3.82-3.81 (2H, d), 3.65-3.55 (8H, m), 2.22-2.16 (1 H, sept), 1 .49 (9H, s), 1 .45 (9H, s), 1 .1 1 -1 .10 (6H, d).
Step 2: 1-(6-(3-(tert-butyl)-4-isobutoxyphenyl)pyridin-2-yl)piperazine dihydrochloride: Following the procedure for 8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)-2,8-diazaspiro[4.5]decane dihydrochloride, tert-butyl 4-(6-(3-(tert-butyl)-4- isobutoxyphenyl)pyridin-2-yl)piperazine-1 -carboxylate (43.6 mg, 0.093 mmol) was reacted to afford the title compound as a yellow solid (25.0 mg, 61 %).
LCMS (Method F): RT = 1 .13 min, M+H+ = 368; 1H NMR (500 MHz, MeOH-d4): 8.1 1 -8.07 (1 H, dd), 7.78 (1 H, d), 7.75-7.72 (1 H, dd), 7.35-7.34 (1 H, d), 7.26-7.24 (1 H, d), 7.17-7.16 (1 H, d), 4.03-4.01 (4H, m), 3.94-3.92 (2H, d), 3.50-3.48 (4H, m), 2.25-2.20 (1 H, sept), 1 .50 (9H, s), 1 .17-1 .15 (6H, d).
Example 52): 1 -(6-(2-(benzyloxy)-1 .1 ,3,3-tetramethyl-2,3-dihvdro-1 H-inden-5-yl)pyridin-
2-yl)piperidin-4-amine dihydrochloride
Figure imgf000084_0002
Step 1: 1 , 1 ,3,3-tetramethyl- 1H-inden-2(3H)-one:
Into a 3-necked round-bottomed flask equipped with a reflux condenser, mechanical stirrer, thermometer and dropping funnel was added DMSO (210 ml) and potassium hydroxide (1 12 g, 2 moles). The resulting suspension was heated at 50-55 ° . A solution of inden-2(3H)-one (13.2 g, 0.1 mol) and methyl iodide (50 ml, 0.8 mol) was added by portions so as to keep the temperature between 50 and 60 °C (the heating was stopped after the first portion and cooling was applied when the temperature exceeded 60 °C. Once the addition was completed, the resulting suspension was stirred for another 1 h. The excess methyl iodide was then distilled off. The suspension was then poured onto ice-water (500 ml) and extracted with hexane (2 times 150 ml). The combined hexane extracts were concentrated in vacuo to afford the title compound as a brown oil (8.5 g, 45.1 %).
1H NMR (500 MHz, CDCI3): 7.14-7.08 (m, 4H), 1 .17 (s, 12H). Step 2: 5-bromo- 1, 1 ,3,3-tetramethyl-1 H-inden-2(3H)-one:
To a solution of aluminium chloride (23.90 g, 179 mmol) in dichloroethane (100 ml) was added a solution of 1 ,1 ,3,3-tetramethyl-1 H-inden-2(3H)-one (15 g, 71 .7 mmol) in dichloroethane (50 ml). A solution of bromine (3.69 ml, 71 .7 mmol) in dichloroethane (30 ml) was added over 30 min. The resulting solution was stirred at room temperature overnight. It was then poured onto ice and extracted with DCM. The DCM layer was washed with a 20% aqueous sodium thiosulfate solution then filtered through a phase separator. The filtrate was concentrated in vacuo and the residue was purified by flash chromatography to afford the title compound (17g, 89%), contaminated with appr. 10% of 4,5-dibromo-1 ,1 ,3,3-tetramethyl-1 H-inden-2(3H)-one. This material was used without further purification.
H NMR (500 MHz, CDCI3): 7.33 (d, 1 H), 7.31 (s, 1 H), 7.06 (s, dH), 1 .16 (s, 12H).
Step 3: 5-bromo- 1, 1,3,3-tetramethyl-2,3-dihydro-1H-inden-2-ol:
To a cold stirred solution of 5-bromo-1 ,1 ,3,3-tetramethyl-1 H-inden-2(3H)-one (7 g, 26.2 mmol) in THF/Methanol (7/3, 50 ml) was added sodium borohydride in small portions (2.97 g, 79 mmol). The solution was stirred at 0 ^ for 1 h then room temperature over 72h. It was quenched with a 1 N aqueous NaOH solution then extracted with ethyl acetate, dried over magnesium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography to give the title compound (4.9 g, 69.5%). H NMR (500 MHz, CDCI3): 7.32 (d, 1 H), 7.26 (s, 1 H), 7.01 (d, 1 H), 3.80 (d, 1 H), 2.08 (d, 1 H), 1 .33 (s, 6H), 1 .17 (s, 3H), 1 .16 (s, 3H).
Step 4: 2-(benzyloxy)-5-bromo- 1 , 1 ,3,3-tetramethyl-2,3-dihydro- 1 H-indene:
To a solution of sodium hydride (60% in mineral oil, 0.30 g, 7.58 mmol) in dry THF (15ml) at 0 °C was added a solution of 5-bromo-1 ,1 ,3,3-tetramethyl-2,3-dihydro-1 H-inden-2-ol (1 .2 g, 4.46 mmol) in dry THF (5 ml). The resulting solution was stirred for 30 min at 0 'Ό. Benzyl bromide (0.91 g, 5.35 mmol) was added and the solution was stirred at room temperature overnight. It was quenched with ice water, diluted with DCM and filtered through a phase separator cartridge. The filtrate was concentrated in vacuo and the residue was purified by flash chromatography using a Biotage SP4 instrument to afford the title compound (0.97 g, 61 %).
1H NMR (500 MHz, CDCI3): 7.4-7.2 (m, 7H), 6.99 (d, 1 H), 4.77 (s, 2H), 3.66 (s, 1 H), 1 .36 (s, 3H), 1 .35 (s, 3H), 1 .24 (s, 3H), 1 .22 (s, 3H).
Step 5: 2-(2-(benzyloxy)- 1, 1 ,3,3-tetramethyl-2,3-dihydro-1 H-inden-5-yl)-4,4,5,5- tetramethyl- 1,3,2-dioxaborolane:
Nitrogen was bubbled through a suspension of 2-(benzyloxy)-5-bromo-1 ,1 ,3,3- tetramethyl-2,3-dihydro-1 H-indene (1 .1 g, 3.06 mmol), PdCI2(dppf) CH2CI2 (0.250 g, 0.306 mmol), potassium acetate (0.9 g, 9.18 mmol) and 4,4,4',4', 5,5,5', 5'-octamethyl- 2,2'-bi(1 ,3,2-dioxaborolane) (1 .166 g, 4.59 mmol) in DMF (10 ml) for 10 minutes. The mixture was heated at 100 'Ό for 15h. After cooling to room temperature the solution was diluted with water (3 ml) and DCM (15 ml). The mixture was filtered through a phase separator cartridge and the filtrate was concentrated in vacuo. The residue was purified by flash chromatography using a Biotage SP4 instrument to afford the title compound (0.43 g, 35%).
1H NMR (500 MHz, CDCI3): 7-7.7.0 (m, 8H), 4.78 (d, 2H), 3.67 (d, 1 H), 1 .4-1 .2 (m, 24H).
Step 6: tert-butyl 1-(6-(2-(benzyloxy)-1 , 1,3,3-tetramethyl-2,3-dihydro- 1H-inden-5- yl)pyridin-2-yl)piperidin-4-ylcarbamate:
Nitrogen was bubbled through a solution of 2-(2-(benzyloxy)-1 ,1 ,3,3-tetramethyl-2,3- dihydro-1 H-inden-5-yl)-4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (0.148 g, 0.365 mmol) , tert-butyl 1 -(6-bromopyridin-2-yl)piperidin-4-ylcarbamate (0.100 g, 0.281 mmol)
PdCI2(dppf) CH2CI2 (0.018 g, 0.022 mmol) and 2N aqueous sodium carbonate (0.291 mL, 0.561 mmol) in dioxane (1 .2 ml) for 10 min and subsequently heated at 90 °C for 15h. After cooling to room temperature, the reaction mixture was diluted with DCM/water and filtered through a phase separator cartridge. The filtrate was concentrated in vacuo and the residue purified by flash chromatography using a Biotage SP4 instrument to afford the title compound (0.039 g, 25%).
LCMS (Method F): RT = 2.06 min, M+H+ = 350; 1H NMR (500 MHz, CDCI3): 7.84 (d, 1 H), 7.70 (s, 1 H), 7.50 (t, 1 H), 7.40 (2, 2H), 7.38 (d, 2H), 7.30 (t, 1 H), 7.19 (d, 1 H), 7.05 (d, 1 H), 6.58 (d, 1 H), 4.80 (s, 2H), 4.49 (brs, 1 H), 4.31 (m, 2H), 3.71 (m, 2H), 3.02 (m, 2H), 2.02 (m, 2H), 1 .70 (m, 1 H), 1 .45 (s, 9H), 1 .39 (s, 3H), 1 .29 (s, 3H), 1 .27 (s, 3H).
Step 7 : 1-(6-(2-(benzyloxy)-1, 1 ,3 etramethyl-2,3-dihydro-1H-inden-5-yl)pyridin-2- yl)piperidin-4-amine
Figure imgf000087_0001
1 -(6-(2-(benzyloxy)-1 ,1 ,3,3-tetramethyl-2,3- dihydro-1 H-inden-5-yl)pyridin-2-yl)piperidin-4-ylcarbamate (50 mg, 0.09 mmol) was added to a 4N HCI solution in dioxane (2ml_, 8mmol). The resulting solution was stirrred at 50 °C for 15 min then concentrated in vacuo to afford the title compound (48 mg, 100%).
LCMS (Method F): RT = 1 .28 min, M+H+ = 456; 1H NMR (500 MHz, CDCI3): 8.77 (brs, 3H), 7-7.8 (m, 1 1 H), 4.75 (s, 2H), 4.4 (m, 2H), 3.4 (m, 2H), 2.5 (m, 2H), 2.15 (m, 2H), 1 .41 (s, 3H), 1 .35 (s, 3H), 1 .30 (s, 3H), 1 .25 (s, 3H).
Figure imgf000087_0002
Step 1 : 4-bromo-2-(tert-butyl)- 1-isopropoxybenzene:
A solution of 4-bromo-2-(tert-butyl)phenol (10 g, 43.6 mmol) in DMF (50 ml) was cooled to 0 °C. NaH (60 % in mineral oil, 3.49 g, 87 mmol) was added slowly. After 30 min, 2- iodopropane (13.07 ml, 131 mmol) was added. Further DMF (40 ml) was added to mobilise the mixture, and it was stirred at RT for 18h. The mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo to afford the title compound as a yellow liquid (12.35 g, >100%). Used without further purification.
1H NMR (500 MHz, CDCI3): 7.35-7.34 (1 H, d), 7.24-7.22 (1 H, dd), 6.71 -6.69 (1 H, d), 4.63-4.58 (1 H, sept), 1 .38-1 .37 (6H, d), 1 .36 (9H, s).
Step 2: 2-(3-(tert-butyl)-4-isopropoxyphenyl)-4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolane: To a solution of 4-bromo-2-(tert-butyl)-1 -isopropoxybenzene (1 1 .8 g, 43.5 mmol) in DMF (150 ml) was added 4,4,4',4', 5,5, 5', 5'-octamethyl-2,2'-bi(1 ,3,2-dioxaborolane) (16.57 g, 65.3 mmol) and potassium acetate (12.81 g, 131 mmol). Nitrogen was bubbled through the mixture, and PdCI2(dppf)CH2CI2 (1 .777 g, 2.176 mmol)) was added. The mixture was heated at 80 °C for 18 h under nitrogen. After cooling to RT, the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 20 % EtOAc in cyclohexane) to afford the title compound as a yellow solid (7.60 g, 55%). 1H NMR (500 MHz, CDCI3): 7.72 (1 H, d), 7.64-7.62 (1 H, dd), 6.85-6.83 (1 H, d), 4.72-4.68 (1 H, sept), 1 .40 (9H, s), 1 .37-1 .36 (6H, d), 1 .32 (12H, s). Step 3: 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine:
To a solution of 2-(3-(tert-butyl)-4-isopropoxyphenyl)-4,4,5,5-tetramethyl-1 ,3,2- dioxaborolane (7.604 g, 23.89 mmol) and 2,4-dichloropyrimidine (2.97 g, 19.91 mmol) in dioxane (80 ml), was added 2M aq Na2C03 (39.8 ml, 80 mmol). Nitrogen was bubbled through the mixture, and PdCI2(dppf)CH2CI2 (0.813 g, 0.996 mmol) was added. Nitrogen was bubbled through the mixture for a further 5 min. The mixture was heated at 90 °C for 18 h under nitrogen. After cooling to RT, the mixture was concentrated in vacuo. The residue was partitioned between DCM and water, filtered through a phase separator cartridge and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 20 % EtOAc in cyclohexane) to afford the title compound as a yellow solid (2.19 g, 36 %).
LCMS (Method F): RT = 1 .80 min, M+H+ = 305.
Step 4: 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin- 1 -yl)ethanol: To a solution of 2-(piperazin-1 -yl)ethanol (42.7mg, 0.328 mmol) in THF (0.5 ml), in a sealable vial, was added DIPEA (0.086 ml, 0.492 mmol). A solution of 4-(3-(tert-butyl)-4- isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) in THF (0.500 ml) was added and the vial was sealed. After heating to 70 'Ό o/n the mixture was partitioned between DCM and water, filtered through a phase separator cartridge and loaded onto a 2 g SCX cartridge. The cartridge was washed with MeOH, and then the desired compound was eluted with 7N NH3 in MeOH. The product containing fractions were concentrated in vacuo to afford the title compound as an orange gum (62 mg, 95%).
LCMS (Method F): RT = 1 .13 min, M+H+ = 399; 1H NMR (500 MHz, MeOH-d4): 8.31 -8.30 (1 H, d), 8.03 (1 H, d), 7.86-7.84 (1 H, dd), 6.92-6.90 (1 H, d), 6.90-6.89 (1 H, d), 4.75-4.70 (1 H, sept), 3.96-3.93 (4H, m), 3.69-3.66 (2H, t), 2.62-2.60 (6H, m), 1 .43 (9H, s), 1 .42- 1 .40 (6H, d).
Example 54: cis-A/l-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)cvclohexane-1 ,4- diamine
Figure imgf000089_0001
Following the procedure for 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)piperazin-1 -yl)ethanol, cis-cyclohexane-1 ,4-diamine (37.5 mg, 0.328 mmol) and 4-(3- (tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) were reacted to afford the title compound as an orange gum (50 mg, 81 %).
LCMS (Method F): RT = 1 .06 min, M+H+ = 383; 1H NMR (500 MHz, MeOH-d4): 8.26-8.25 (1 H, d), 8.04-8.01 (1 H, m), 7.81 -7.80 (1 H, d), 6.92-6.90 (1 H, d), 6.89-6.88 (1 H, d), 5.19 (1 H, br s), 4.74-4.71 (1 H, sept), 4.1 1 (1 H, br s), 2.91 (1 H, br s), 1 .90-1 .85 (2H, m), 1 .79- 1 .73 (4H, m), 1 .43 (9H, s), 1 .41 -1 .40 (6H, d).
Example 55: 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-2-one
Figure imgf000090_0001
Following the procedure for 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)piperazin-1 -yl)ethanol, piperazin-2-one trifluoroacetate (70.3 mg, 0.328 mmol) and 4- (3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) were reacted to afford the title compound as an orange gum (55 mg, 92 %).
LCMS (Method F): RT = 1 .51 min, M+H+ = 369; 1H NMR (500 MHz, MeOH-d4): 8.34-8.33 (1 H, d), 8.02-8.01 (1 H, d), 7.91 -7.89 (1 H, dd), 6.98-6.97 (1 H, d), 6.93-6.91 (1 H, d), 6.14 (1 H, br s), 4.76-4.71 (1 H, sept), 4.56 (2H, s), 4.16-4.14 (2H, t), 3.53-3.50 (2H, m), 1 .43 (9H, s), 1 .42-1 .41 (6H, d).
Example 56: (fi)-4-(3-(tert-butvn-4-isopropoxyphenyl)-2-(3-methylpiperazin-1 - vDpyrimidine
Figure imgf000090_0002
To a solution of (R)-tert-butyl 2-methylpiperazine-1 -carboxylate (82 mg, 0.328 mmol) in THF (0.5 ml), in a sealable vial, was added DIPEA (0.086 ml, 0.492 mmol). A solution of 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) in THF (0.5 ml) was added and the vial was sealed. After heating to 70 'Ό o/n the mixture was partitioned between DCM and water and filtered through a phase separator cartridge. HCI in dioxane (4M, 1 .0 ml, 4 mmol) was added and the mixture stirred for 1 h. The reaction was neutralized with sat. aq. NaHC03, filtered through a phase separator cartridge and loaded onto a 2 g SCX cartridge. The cartridge was washed with MeOH, and then the desired compound was eluted with 7N NH3 in MeOH. The product containing fractions were concentrated in vacuo to afford the title compound as an orange gum (47 mg, 78 %).
LCMS (Method F): RT = 1 .1 1 min, M+H+ = 369; 1H NMR (500 MHz, MeOH-d4): 8.33-8.32 (1 H, d), 8.06 (1 H, d), 7.89-7.87 (1 H, dd), 6.95-6.93 (1 H, d), 6.90-6.89 (1 H, d), 6.14 (1 H, br s), 4.80-4.72 (3H, m), 3.15-3.13 (1 H, br d), 3.05-2.90 (3H, m), 2.66-2.62 (1 H, br t), 1 .46 (9H, s), 1 .44-1 .43 (6H, d), 1 .19-1 .18 (3H, d).
Example 57: 7-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-1 ,7- diazaspiro[3.51nonane
Figure imgf000091_0001
Following the procedure for (R)-4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(3- methylpiperazin-1 -yl)pyrimidine, tert-butyl 1 ,7-diazaspiro[3.5]nonane-1 -carboxylate (93 mg, 0.328 mmol) and 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) were reacted to afford the title compound as an orange gum (42 mg, 64%).
LCMS (Method F): RT = 1 .10 min, M+H+ = 395; 1H NMR (500 MHz, MeOH-d4): 8.34-8.33 (1 H, d), 8.07 (1 H, d), 7.90-7.88 (1 H, dd), 6.94-6.93 (1 H, d), 6.90-6.89 (1 H, d), 5.62 (1 H, br s), 4.77-4.72 (1 H, sept), 4.32 (2H, br s), 4.07-4.04 (2H, t), 2.87-2.84 (2H, t), 2.27-2.19 (5H, m), 1 .46 (9H, s), 1 .44-1 .43 (6H, d).
Example 58: 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-4-methyl-1 A- diazepane
Figure imgf000091_0002
Following the procedure for 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)piperazin-1 -yl)ethanol, 1 -methyl-1 ,4-diazepane (46.8 mg, 0.410 mmol) and 4-(3-(tert- butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) were reacted to afford the title compound as an orange gum (55 mg, 88%).
LCMS (Method F): RT = 1 .13 min, M+H+ = 383; 1H NMR (500 MHz, MeOH-d4): 8.33-8.32 (1 H, d), 8.10 (1 H, d), 7.89-7.87 (1 H, dd), 6.94-6.92 (1 H, d), 6.88-6.87 (1 H, d), 4.77-4.72 (1 H, sept), 4.05 (2H, br s), 3.94 (2H, br s), 2.77-2.75 (2H, m), 2.63-2.60 (2H, m), 2.41 (3H, s), 2.09-2.04 (2H, m), 1 .46 (9H, s), 1 .44-1 .43 (6H, d). Example 59: (1 -(4-(3-(tert-butvn-4-isopropoxyphenyl)pyrimidin-2-vnazetidin-3- vDmethan amine
Figure imgf000092_0001
Following the procedure for (R)-4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(3-methyl- piperazin-1 -yl)pyrimidine, tert-butyl (azetidin-3-ylmethyl)carbamate (92 mg, 0.492 mmol) and 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) were reacted to afford the title compound as an orange gum ( 39.7 mg, 68% ).
LCMS (Method F): RT = 0.98 min, M+H+ = 355; 1H NMR (500 MHz, MeOH-d4): 8.33-8.32 (1 H, d), 8.08 (1 H, d), 7.88-7.86 (1 H, dd), 6.94-6.92 (2H, m), 4.77-4.72 (1 H, sept), 4.33- 4.29 (2H, t), 3.93-3.90 (2H, m), 3.05-3.03 (2H, d), 2.82-2.74 (1 H, m), 1 .45 (9H, s), 1 .44- 1 .43 (6H, d). Example 60: 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-1 - vDethanamine
Figure imgf000093_0001
Following the procedure for 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)piperazin-1 -yl)ethanol, 2-(piperazin-1 -yl)ethanamine (63.6 mg, 0.492 mmol) and 4-(3- (tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) were reacted to afford the title compound as an orange gum (52.4 mg, 80%).
LCMS (Method F): RT = 1 .02 min, M+H+ = 398; 1H NMR (500 MHz, MeOH-d4): 8.33-8.32 (1 H, d), 8.05 (1 H, d), 7.88-7.86 (1 H, dd), 6.94-6.92 (1 H, d), 6.90-6.89 (1 H, d), 4.76-4.72 (1 H, sept), 3.96-3.94 (4H, m), 2.88-2.86 (2H, t), 2.60-2.48 (6H, m), 1 .45 (9H, s), 1 .44- 1 .42 (6H, s).
Example 61 : 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-1 -yl)-A/.A/- dimethylethan amine
Figure imgf000093_0002
Following the procedure for 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)piperazin-1 -yl)ethanol, /V,/V-dimethyl-2-(piperazin-1 -yl)ethanamine
(77 mg, 0.492 mmol) and 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) were reacted to afford the title compound as an orange gum (57.9 mg, 83%).
LCMS (Method F): RT = 1 .1 1 min, M+H+ = 426; 1H NMR (500 MHz, MeOH-d4): 8.33-8.32 (1 H, d), 8.05 (1 H, d), 7.88-7.86 (1 H, dd), 6.94-6.92 (1 H, d), 6.90-6.89 (1 H, d), 4.77-4.72 (1 H, sept), 3.97-3.95 (4H, m), 2.61 -2.51 (8H, m), 2.30 (6H, m), 1 .45 (9H, s), 1 .44-1 .43 (6H, s). Example 62: (rac)-1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)pyrrolidin-3- amine
Figure imgf000094_0001
Following the procedure for 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)piperazin-1 -yl)ethanol, (rac)-pyrrolidin-3-amine (42.4 mg, 0.492 mmol) and 4-(3-(tert- butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) were reacted to afford the title compound as an orange gum (50.4 mg, 86%).
LCMS (Method F): RT = 1 .02 min, M+H+ = 355; 1H NMR (500 MHz, MeOH-d4): 8.34-8.32 (1 H, d), 8.1 1 (1 H, d), 7.90-7.87 (1 H, dd), 6.94-6.92 (1 H, d), 6.90-6.89 (1 H, d), 4.77-4.72 (1 H, sept), 3.95-3.85 (2H, br d), 3.77-3.73 (2H, m), 3.45-3.40 (1 H, br s), 2.28-2.21 (1 H, m), 1 .88-1 .82 (1 H, m), 1 .46 (9H, s), 1 .44-1 .43 (6H, d).
Example 63: (4-amino-1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4- vDmethanol
Figure imgf000094_0002
Following the procedure for 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)piperazin-1 -yl)ethanol, ethyl 4-((tert-butoxycarbonyl)amino)piperidine-4-carboxylate (179 mg, 0.656 mmol) and 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (100 mg, 0.328 mmol) were reacted to afford crude ethyl 4-((tert-butoxycarbonyl)amino)-1 -(4- (3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidine-4-carboxylate (200 mg, >100%). This material was dissolved in THF (2 ml) and LiAIH4 (35.1 mg, 0.925 mmol) was added. After stirring at RT for 18 h, 40% aq KOH (0.3 ml) was added slowly, and the mixture stirred at RT for 15 min. The resultant suspension was filtered through celite, washed with EtOAc, and the combined filtrates concentrated in vacuo. The resultant residue was dissolved in dioxane (1 ml) and HCI (4M in dioxane, 2 ml, 8.00 mmol) was added. After stirring at RT for 2 h DCM (2 ml) was added, and the reaction was neutralized with sat. aq. NaHC03, filtered through phase separator cartridge and loaded onto a 2 g SCX cartridge. The cartridge was washed with MeOH, and the desired compound was eluted with 7N NH3 in MeOH. The product containing frations were concentrated in vacuo. Further purification was carried out, using preparative HPLC (Methods C & D) to afford the title compound as a white solid (0.9 mg, 0.6%).
LCMS (Method F): RT = 1 .09 min, M+H+ = 399; 1H NMR (500 MHz, MeOH-d4): 8.35-8.34 (1 H, d), 8.08-8.07 (1 H, d), 7.91 -7.89 (1 H, dd), 6.96-6.95 (1 H, d), 6.91 -6.90 (1 H, d), 4.79- 4.74 (1 H, sept), 4.32-4.29 (2H, m), 3.75-3.69 (4H, m), 1 .75-1 .71 (4H, m), 1 .47 (9H, s), 1 .46-1 .44 (6H, d). Example 64: 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 -vDpyrimidine
dihvdrochloride
Figure imgf000095_0001
Step 1 2-(4-isopropoxy-3-isopropylphenyl)-4,4,5,5-tetramethyl-1,3,2-dioxaborolane: Following the procedure for 2-(3-(tert-butyl)-4-isopropoxyphenyl)-4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolane, 4-bromo-1 -isopropoxy-2-isopropylbenzene (1 .67 g, 6.49 mmol) was reacted to afford the title compound (686 mg, 35%).
1H NMR (500 MHz, CDCI3): 7.64 (1 H, s), 7.62-7.60 (1 H, d), 6.84-6.82 (1 H, d), 4.62-4.58 (1 H, sept), 3.32-3.28 (1 H, sept), 1 .35-1 .33 (6H, d), 1 .33 (12H, s), 1 .23-1 .22 (6H, d).
Step 2: 2-chloro-4-(4-isopropoxy-3-isopropylphenyl)pyrimidine:
Following the procedure for 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine, 2-
(4-isopropoxy-3-isopropylphenyl)-4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (300 mg, 0.986 mmol) and 2,4-dichloropyrimidine (122 mg, 0.822 mmol) were reacted to afford the title compound as a yellow oil (59 mg, 25%).
1H NMR (500 MHz, CDCI3): 8.54-8.52 (1 H, d), 7.93-7.91 (2H, m), 7.57-7.56 (1 H, d), 6.94- 6.92 (1 H, dd), 4.69-4.66 (1 H, sept), 3.36-3.33 (1 H, sept), 1 .38-1 .37 (6H, d), 1 .29-1 .28 (6H, d).
Step 3: tert-butyl 4-(4-(4-isopropoxy-3-isopropylphenyl)pyrimidin-2-yl)piperazine-1- carboxylate:
To a solution of tert-butyl piperazine-1 -carboxylate (37.7 mg, 0.203 mmol) and 2-chloro- 4-(4-isopropoxy-3-isopropylphenyl)pyrimidine (58.9 mg, 0.203 mmol) in dioxane, was added Xantphos (1 1 .72 mg, 0.020 mmol) and potassium phosphate (95 mg, 0.446 mmol). Nitrogen was bubbled through the mixture, and Pd2dba3 (1 1 .13 mg, 0.012 mmol) was added. The mixture was heated at 80 °C for 18 h under nitrogen. The mixture was cooled to RT and filtered through celite (washing with EtOAc). The combined filtrates were concentrated in vacuo and the resultant residue was subjected to flash
chromatography (Si02, gradient 0 to 15 % EtOAc in cyclohexane) to afford the title compound as an orange oil (56 mg, 63%).
1H NMR (500 MHz, CDCI3): 8.32-8.31 (1 H, d), 7.89-7.85 (2H, m), 6.92-6.90 (2H, m), 4.66-4.62 (1 H, sept), 3.91 -3.89 (4H, m), 3.54-3.52 (4H, m), 3.37-3.33 (1 H, sept), 1 .52 (9H, s), 1 .37-1 .36 (6H, d), 1 .24-1 .23 (6H, d).
Step 4: 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin- 1-yl)pyrimidine dihydrochloride: To a solution of tert-butyl 4-(4-(4-isopropoxy-3-isopropylphenyl)pyrimidin-2-yl)piperazine- 1 -carboxylate (56.3 mg, 0.128 mmol) in dioxane (0.5 ml) was added HCI (4M in dioxane, 1 ml, 4.00 mmol). The mixture was stirred at room temperature for 1 h, and then further HCI (4M in dioxane, 0.5 ml, 2.00 mmol) was added. After 1 h at room temperature the mixture was concentrated in vacuo .The resultant solid was slurried in Et20, then lyophilized o/n to afford the title compound as a white solid (19 mg, 36%).
LCMS (Method F): RT = 1 .02 min, M+H+ = 341 ; 1H NMR (500 MHz, MeOH-d4): 8.26-8.24 (1 H, d), 7.98-7.96 (2H, m), 7.31 -7.30 (1 H, d), 6.99-6.97 (1 H, d), 4.70-4.65 (1 H, sept), 4.12-4.10 (4H, t), 3.32-3.30 (4H, t), 3.29-3.22 (1 H, sept), 1 .29-1 .28 (6H, d), 1 .18-1 .17 (6H, d).
Figure imgf000097_0001
Step 1: tert-butyl 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine- 1- carboxylate:
Following the procedure for tert-butyl 4-(4-(4-isopropoxy-3-isopropylphenyl)pyrimidin-2- yl)piperazine-1 -carboxylate, tert-butyl piperazine-1 -carboxylate (29.1 mg, 0.156 mmol) and 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (47.7 mg, 0.156 mmol) were reacted to afford the title compound as a yellow oil (39 mg, 55%).
1H NMR (500 MHz, CDCI3): 8.32-8.31 (1 H, d), 8.02 (1 H, d), 7.87-7.85 (1 H, dd), 6.92-6.90 (2H, m), 4.65-4.60 (1 H, sept), 3.90-3.88 (4H, m), 3.54-3.52 (4H, m), 1 .50 (9H, s), 1 .43 (9H, s), 1 .42-1 .41 (6H, d). Step 2: 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(piperazin- 1 -yl)pyrimidine dihydrochloride: Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin- 2-yl)piperazine-1 -carboxylate (39.4 mg, 0.087 mmol) was reacted to afford the title compound as a white solid (16 mg, 44%).
LCMS (Method F): RT = 1 .09 min, M+H+ = 355; 1H NMR (500 MHz, MeOH-d4): 8.25-8.24 (1 H, d), 8.07-8.06 (1 H, d), 7.98-7.96 (1 H, dd), 7.30-7.28 (1 H, d), 7.01 -6.99 (1 H, d), 4.78- 4.73 (1 H, sept), 4.1 1 -4.09 (4H, t), 3.32-3.30 (4H, t), 1 .35 (9H, s), 1 .33-1 .32 (6H, s).
Example 66: 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-amine dihydrochloride
Figure imgf000097_0002
Step 1: tert-butyl 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine- 1- carboxylate:
Following the procedure for tert-butyl 4-(4-(4-isopropoxy-3-isopropylphenyl)pyrimidin-2- yl)piperazine-1 -carboxylate, tert-butyl piperidin-4-ylcarbamate (31 .3 mg, 0.156 mmol) and 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (47.7 mg, 0.156 mmol) were reacted to afford the title compound as an orange oil (38 mg, 55%).
1H NMR (500 MHz, CDCI3): 8.30-8.29 (1 H, d), 8.02 (1 H, d), 7.86-7.84 (1 H, dd), 6.92-6.90 (1 H, d), 6.87-6.86 (1 H, d), 4.76-4.72 93H, m), 4.48 (1 H, br s), 3.72 (1 H, br s), 3.15-3.10 (2H, m), 2.04-2.02 (2H, m), 1 .56-1 .52 (2H, m), 1 .46 (9H, s), 1 .41 -1 .40 (9H, s), 1 .39-1 .38 (6H, d).
Step 2: 1-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin
dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)piperidin-4-amine (38.3 mg, 0.104 mmol) was reacted to afford the title compound as a white solid (19 mg, 42%).
LCMS (Method F): RT = 1 .21 min, M+H+ = 369; 1H NMR (500 MHz, MeOH-d4): 8.15-8.14 (2H, m), 8.07-8.05 (1 H, dd), 7.42-7.41 (1 H, d), 7.07-7.05 (1 H, d), 4.82-4.76 (1 H, sept), 4.76-4.65 (2H, br m), 3.51 -3.46 (1 H, m), 3.33-3.28 (2H, dt), 2.20-2.14 (2H, m), 1 .73-1 .65 (2H, ddd), 1 .36 (9H, s), 1 .35-1 .34 (6H, d).
Example 67: 1 -(4-(3-(tert-butyl)-4-(cvclopentyloxy)phenyl)pyrimidin-2-yl)piperidin-4-amine dihydrochloride
Figure imgf000098_0001
Step 1: 4-bromo-2-(tert-butyl)- 1-(cyclopentyloxy)benzene:
Following the procedure for 4-bromo-2-(tert-butyl)-1 -isopropoxybenzene, 4-bromo-2- (tert-butyl)phenol (10 g, 43.6 mmol) and bromocyclopentane (14.04 ml, 131 mmol) were reacted to afford to afford the title compound as a yellow liquid (16.06 g). Used without further purification.
1H NMR (500 MHz, CDCI3): 7.34-7.33 (1 H, d), 7.24-7.23 (1 H, dd), 6.71 -6.69 (1 H, d), 4.43-4.40 (1 H, m), 2.09-2.07 (2H, m), 1 .91 -1 .89 (2H, m), 1 .83-1 .80 (2H, m), 1 .67-1 .64 (2H, m), 1 .34 (9H, s).
Step 2: 2-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)-4,4,5,5-tetramethyl- 1,3,2- dioxaborolane:
Following the procedure for 2-(3-(tert-butyl)-4-isopropoxyphenyl)-4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolane, 4-bromo-2-(tert-butyl)-1 -(cyclopentyloxy)benzene (12.9 g, 43.4 mmol) was reacted to afford the title compound as a yellow oil (3.96 g, 26%).
Used without further purification.
Step 3: 4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)-2-chloropyrimidine:
Following the procedure for 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine, 2- (3-(tert-butyl)-4-(cyclopentyloxy)phenyl)-4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (3.8 g, 1 1 .06 mmol) and 2,4-dichloropyrimidine (1 .373 g, 9.21 mmol) were reacted to afford the title compound as an orange semi-solid (750 mg, 25%).
H NMR (500 MHz, CDCI3): 8.53-8.52 (1 H, d), 8.01 -8.01 (1 H, d), 7.55-7.52 (2H, m), 6.95- 6.93 (1 H, d), 4.93-4.91 (1 H, m), 1 .98-1 .95 (4H, m), 1 .86-1 .82 (2H, m), 1 .71 -1 .68 (2H, m), 1 .42 (9H, s).
Step 4: 1-(4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)pyrimidin-2-yl)piperidin-4-am dihydrochloride:
To a solution of tert-butyl piperidin-4-ylcarbamate (76 mg, 0.378 mmol) in THF (0.5 ml), in a sealable vial, was added DIPEA (0.086 ml, 0.492 mmol). A solution of 4-(3-(tert- butyl)-4-(cyclopentyloxy)phenyl)-2-chloropyrimidine (50 mg, 0.151 mmol) in THF (0.500 ml) was added, and the vial was sealed. After heating to 70 °C o/n the mixture was partitioned between DCM and water, filtered through phase separator cartridge and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 20 % EtOAc in cyclohexane). The isolated material was dissolved in dioxane (0.5 ml), and HCI (4M in dioxane, 1 ml) was added. After stirring for 2h at room temperature, the mixture was concentrated in vacuo .The resultant solid was slurried in Et20 and then lyophilized o/n to afford the title compound as a pale yellow solid (47 mg, 67 %).
LCMS (Method F): RT = 1 .32 min, M+H+ = 395; 1H NMR (500 MHz, MeOH-d4): 8.15-8.13 (1 H, d), 8.13-8.12 (1 H, d), 8.06-8.04 (1 H, dd), 7.40-7.39 (1 H, d), 7.05-7.03 (1 H, d), 4.96- 4.94 (1 H, sept), 4.71 -4.65 (2H, br m), 3.50-3.44 (1 H, m), 3.32-3.27 (2H, dt), 2.18-2.13 (2H, m), 1 .97-1 .92 (2H, m), 1 .87-1 .82 (2H, m), 1 .77-1 .63 (6H, m), 1 .34 (9H, s).
Example 68: 1 -(2-(3-isopropoxy-4-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine dihydrochloride
Figure imgf000100_0001
Step 1: tert-butyl (1 -(2-chloropyrimidin-4-yl)piperidin-4-yl)carbamate:
To a solution of 2,4-dichloropyrimidine (250 mg, 1 .678 mmol) and tert-butyl piperidin-4- ylcarbamate (370 mg, 1 .846 mmol) in DMF (2.5 ml) was added triethylamine (0.351 ml, 2.52 mmol). After stirring at RT for 18h the reaction mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant solid was slurried in Et20, filtered and dried in vacuo to afford the title compound as a white solid (272 mg, 52%)
1H NMR (500 MHz, CDCI3): 8.06-8.04 (1 H, d), 6.43-6.42 (1 H, d), 4.70-4.20 (3H, br m), 3.75 (1 H, br s), 3.1 1 -3.06 (2H, dt), 2.10-2.07 (2H, m), 1 .41 (9H, s), 1 .41 -1 .38 (2H, m).
Step 2: tert-butyl (1-(2-(3-isopropoxy-4-isopropylphenyl)pyrimidin-4-yl)piperidin-4- yl) carbarn ate:
Following the procedure for 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine, 2- (3-isopropoxy-4-isopropylphenyl)-4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (60 mg, 0.197 mmol) and tert-butyl (1 -(2-chloropyrimidin-4-yl)piperidin-4-yl)carbamate (51 .4 mg, 0.164 mmol) were reacted to afford to afford the title compound as a yellow oil (39.3 mg, 52%). 1H NMR (500 MHz, CDCI3): 8.24-8.23 (1 H, d), 7.88-.784 (2H, m), 7.22-7.21 (1 H, d), 6.35- 6.37 (1 H, m), 4.73 (1 H, br s), 4.56-4.58 (1 H, sept), 4.50-4.70 (3H, br m), 3.72-3.69 (1 H, br s), 3.31 -3.28 (2H, m), 3.10-3.00 (2H, m), 2.05-2.00 (2H, m), 1 .39 (9H, s), 1 .34-1 .32 (6H, d), 1 .27 (9H, s), 1 .20-1 .19 (6H, d).
Step 3: 1 -(2-(3-isopropoxy-4-isopropylphenyl)pyrimidin-4-yl)piperidin
dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl (1 -(2-(3-isopropoxy-4-isopropylphenyl)pyrimidin- 4-yl)piperidin-4-yl)carbamate (39.3 mg, 0.086 mmol) was reacted to afford the title compound as a tan solid (15.5 mg, 41 %).
LCMS (Method F): RT = 0.70 min, M+H+ = 355; 1H NMR (500 MHz, MeOH-d4): 8.25-8.24 (1 H, d), 7.78-7.76 (1 H, dd), 7.72 (1 H, d), 7.49-7.48 (1 H, d), 7.1 19-7.18 (1 H, d), 4.86-4.80 (1 H, sept), 4.85-4.80 (2H, br m), 3.66-3.60 (2H, m), 3.55-3.35 (2H, br m), 3.45-3.40 (1 H, sept), 2.31 -2.29 (2H, br d), 1 .80-1 .70 (2H, br m), 1 .43-1 .42 (6H, d), 1 .28-1 .27 (6H, d).
Figure imgf000101_0001
Step 1: tert-butyl 4-(6-(3-hydroxy-4-isopropylphenyl)pyridin-2-yl)piperazine-1- carboxylate:
To a solution of 5-bromo-2-isopropylphenol (1 .5 g, 6.97 mmol) in DMF (20 ml) was added 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1 ,3,2-dioxaborolane) (2.66 g, 10.46 mmol) and potassium acetate (2.053 g, 20.92 mmol). Nitrogen was bubbled through the mixture, and PdCI2(dppf)CH2CI2 (0.570 g, 0.697 mmol) was added. The mixture was heated at 80 °C for 18 h under nitrogen. After cooling to RT, tert-butyl 4-(6-bromopyridin- 2-yl)piperazine-1 -carboxylate (1 .592 g, 4.65 mmol), PdCI2(dppf)CH2CI2 (0.570 g, 0.697 mmol) and Na2C03 (2M aq) (13.95 ml, 27.9 mmol) were added. The mixture was heated at 80 °C for 5 h under nitrogen. After cooling to RT, the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 30 % EtOAc in cyclohexane) to afford the title compound as a brown oil (1 .0 g, 54%).
LCMS (Method F): RT = 1 .66 min, M+H+ = 398.
Step 2: tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2-yl)piperazine-1- carboxylate:
To a solution of tert-butyl 4-(6-(3-hydroxy-4-isopropylphenyl)pyridin-2-yl)piperazine-1 - carboxylate (120 mg, 0.302 mmol) in DMF (1 .5 ml) was added K2C03 (50.1 mg, 0.362 mmol) and 1 -bromo-2-methylpropane (0.049 ml, 0.453 mmol). The resultant suspension was stirred at 70 °C for 18 h. Further K2C03 (50.1 mg, 0.362 mmol) and 1 -bromo-2- methylpropane (0.049 ml, 0.453 mmol) were added, and heating continued for 72 h. After cooling to RT, the mixture was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc, and the combined organic extracts were washed with water, dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 20 % EtOAc in cyclohexane) to afford the title compound as a yellow oil (58.7 mg, 43%).
1H NMR (500 MHz, CDCI3): 7.50-7.49 (1 H, t), 7.43-7.42 (1 H, dd), 7.40-7.39 (1 H, d), 7.20- 7.18 (1 H, m), 7.04-7.02 (1 H, d), 6.54-6.52 (1 H, d), 3.78-3.77 (2H, d), 3.58-3.52 (8H, m), 3.35-3.30 (1 H, sept), 2.10-2.05 (1 H, sept), 1 .43 (9H, s), 1 .19-1 .17 (6H, d), 1 .03-1 .02 (6H, d).
Step 3: 1-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2-yl)piperazinedihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (58.7 mg, 0.129 mmol) was reacted to afford the title compound as a yellow solid (39.7 mg, 72%).
LCMS (Method F): RT = 1 .16 min, M+H+ = 354; 1H NMR (500 MHz, MeOH-d4): 8.01 -7.97 (1 H, dd), 7.47-7.43 (2H, m), 7.39-7.36 (2H, dd), 7.19-7.17 (1 H, d), 4.01 -3.99 (4H, t), 3.92-3.90 (2H, d), 3.47-3.40 (5H, m), 2.21 -2.15 (1 H, sept), 1 .29-1 .28 (6H, d), 1 .14-1 .12 (6H, d).
Example 70: 1 -(6-(4-isopropyl-3-propoxyphenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000103_0001
Step 1: tert-butyl 4-(6-(4-isopropyl-3-propoxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
Following the procedure for tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate, tert-butyl 4-(6-(3-hydroxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.302 mmol) and 1 -bromopropane (0.041 ml, 0.453 mmol) were reacted to afford the title compound as a yellow oil (64.4 mg, 48%).
LCMS (Method F): RT = 2.03 min, M+H+ = 440.
Step 2: 1-(6-(4-isopropyl-3-propoxyphenyl)pyridin-2-yl)piperazine dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride tert-butyl 4-(6-(4-isopropyl-3-propoxyphenyl)pyridin-2- yl)piperazine-1 -carboxylate (64.4 mg, 0.147 mmol) was reacted to afford the title compound as a yellow solid (43.6 mg, 72%).
LCMS (Method F): RT = 1 .1 1 min, M+H+ = 340; 1H NMR (500 MHz, MeOH-d4): 7.90-7.87 (1 H, dd), 7.50-7.49 (2H, m), 7.35-7.33 (2H, d), 7.08-7.06 (1 H, d), 4.10-4.07 (2H, t), 3.98- 3.96 (4H, t), 3.44-3.37 (5H, m), 1 .94-1 .87 (2H, m), 1 .28-1 .27 (6H, d), 1 .15-1 .12 (3H, t).
Example 71 : 1 -(6-(3-(cvclopentyloxy)-4-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000103_0002
Step 1: tert-butyl 4-(6-(3-(cyclopentyloxy)-4-isopropylphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
Following the procedure for tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate, tert-butyl 4-(6-(3-hydroxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.302 mmol) and bromocyclopentane (0.049 ml, 0.453 mmol) were reacted to afford the title compound as a yellow oil (62.8 mg, 45%). LCMS (Method F): RT = 2.10 min, M+H+ = 466.
Step 2: 1-(6-(3-(cyclopentyloxy)-4-isopropylphenyl)pyridin-2-yl)piperazine
dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(6-(3-(cyclopentyloxy)-4- isopropylphenyl)pyridin-2-yl)piperazine-1 -carboxylate (62.8 mg, 0.135 mmol) was reacted to afford the title compound as a yellow solid (35.7 mg, 60%).
LCMS (Method F): RT = 1 .14 min, M+H+ = 366; 1H NMR (500 MHz, MeOH-d4): 8.08-8.02 (1 H, br m), 7.42-7.35 (4H, m), 7.27-7.22 (1 H, br m), 5.04-5.02 (1 H, br m), 4.03-4.01 (4H, t), 3.48-3.46 (4H, t), 3.37-3.32 (1 H, sept), 2.05-1 .70 (8H, m), 1 .26-1 .25 (6H, d).
Example 72: 1 -(6-(4-isopropyl-3-(prop-1 -en-2-yl)phenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000104_0001
Step 1: tert-butyl 4-(6-(4-isopropyl-3-(((trifluoromethyl)sulfonyl)oxy)phenyl)pyridin-2- yl)piperazine- 1 -carboxylate:
To a solution of tert-butyl 4-(6-(3-hydroxy-4-isopropylphenyl)pyridin-2-yl)piperazine-1 - carboxylate (220 mg, 0.553 mmol) in DCM (3 ml) at 0 °C, was added triflic anhydride (0.1 12 ml, 0.664 mmol) and pyridine (0.063 ml, 0.775 mmol). The reaction mixture was stirred for 30 min and quenched with aq NaHC03. The mixture was passed through a phase separator cartridge and the organic fraction was concentrated in vacuo to afford the title compound as a brown gum (302 mg). This material was used without further purification.
LCMS (Method F): RT = 1 .98 min, M+H+ = 530. Step 2: tert-butyl 4-(6-(4-isopropyl-3-(prop-1-en-2-yl)phenyl)pyridin-2-yl)piperazine- 1- carboxylate:
To a solution of tert-butyl 4-(6-(4-isopropyl-3-(((trifluoromethyl)sulfonyl)oxy)phenyl)- pyridin-2-yl)piperazine-1 -carboxylate (290 mg, 0.548 mmol) in toluene (2.5 ml) and water (1 ml) was added potassium isopropenyltrifluoroborate (105 mg, 0.712 mmol) and Cs2C03 (357 mg, 1 .095 mmol)). Nitrogen was bubbled through the mixture and
Pd(PPh3)4 (31 .6 mg, 0.027 mmol) was added. The mixture was heated at 100 °C for 15 h under nitrogen. After cooling to RT the organic layer was decanted, filtered through a phase separator cartridge and the organic fraction was concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 20 % EtOAc in cyclohexane) to afford the title compound as a yellow oil (1 14.9 mg, 50 %). LCMS (Method F): RT = 2.00 min, M+H+ = 422.
Step 3: 1-(6-(4-isopropyl-3-(prop-1-en-2-yl)phenyl)pyridin-2-yl)piperazine
dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(6-(4-isopropyl-3-(prop-1 -en-2- yl)phenyl)pyridin-2-yl)piperazine-1 -carboxylate (30 mg, 0.071 mmol) was reacted to afford the title compound as a yellow solid (23.4 mg, 83%).
LCMS (Method F): RT = 1 .07 min, M+H+ = 322; 1H NMR (500 MHz, MeOH-d4): 7.87-7.84 (2H, m), 7.67-7.66 (1 H, d), 7.47-7.46 (1 H, d), 7.33-7.32 (1 H, d), 7.06-7.05 (1 H, d), 5.29- 5.28 (1 H, m), 4.89-4.87 (1 H, m), 3.98-3.96 (4H, t), 3.43-3.41 (4H, t), 3.28-3.22 (1 H, sept), 2.1 1 (3H, s), 1 .28-1 .27 (6H, d).
Example 73: 1 -(6-(3,4-diisopropylphenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000105_0001
Step 1: tert-butyl 4-(6-(3,4-diisopropylphenyl)pyridin-2-yl)piperazine- 1 -carboxylate:
A solution of tert-butyl 4-(6-(4-isopropyl-3-(prop-1 -en-2-yl)phenyl)pyridin-2-yl)piperazine- 1 -carboxylate (80 mg, 0.190 mmol) in Ethanol (4 ml) was hydrogenated using an H-cube (Pd/C, full H2, RT, 1 ml/min). The resultant solution was passed through the H-cube twice more (full H2, 50°C, 1 ml/min), and then concentrated in vacuo Xo afford the title compound as a yellow oil (61 .1 mg, 76%).
LCMS (Method F): RT = 2.01 min, M+H+ = 424. Step 2: 1-(6-(3,4-diisopropylphenyl)pyridin-2-yl)piperazine dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(6-(3,4-diisopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (60 mg, 0.142 mmol) was reacted to afford the title compound as a yellow solid (42.0 mg, 75%).
LCMS (Method F): RT = 1 .06 min, M+H+ = 324; 1H NMR (500 MHz, MeOH-d4): 8.17-8.13 (1 H, dd), 7.74 (1 H, d), 7.68-7.66 (1 H, dd), 7.53-7.51 (1 H, d), 7.37-7.34 (2H, m), 4.06- 4.04 (4H, t), 3.51 -3.49 (4H, t), 3.45-3.39 (2H, 2 x sept), 1 .35-1 .34 (6H, d), 1 .32-1 .31 (6H, d). Example 74: 1 -(6-(3-isopropyl-4-propoxyphenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000106_0001
Step 1: tert-butyl 4-(6-(3-isopropyl-4-propoxyphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
Following the procedure for tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate, tert-butyl 4-(6-(4-hydroxy-3-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.302 mmol) and 1 -bromopropane (0.041 ml, 0.453 mmol) were reacted to afford the title compound as a yellow oil (62.6 mg, 47%).
LCMS (Method F): RT = 2.00 min, M+H+ = 440. Step 2: 1-(6-(3-isopropyl-4-propoxyphenyl)pyridin-2-yl)piperazine dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(6-(4-hydroxy-3-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (62 mg, 0.141 mmol) was reacted to afford the title compound as a yellow solid (50.9 mg, 88%).
LCMS (Method F): RT = 1 .07 min, M+H+ = 340; 1H NMR (500 MHz, MeOH-d4): 8.07-8.04 (1 H, dd), 7.73-7.71 (2H, m), 7.35-7.33 (1 H, d), 7.22-7.20 (1 H, d), 7.12-7.10 (1 H, d), 4.09- 4.07 (2H, t), 4.02-4.00 (4H, t), 3.49-3.47 (4H, t), 3.45-3.39 (1 H, sept), 1 .94-1 .87 (2H, m), 1 .32-1 .31 (6H, d), 1 .15-1 .12 (3H, t).
Example 75: 1 -(6-(4-(cvclopentyloxy)-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000107_0001
Step 1: tert-butyl 4-(6-(4-(cyclopentyloxy)-3-isopropylphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
Following the procedure for tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate, tert-butyl 4-(6-(4-hydroxy-3-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.302 mmol) and bromocyclopentane (0.049 ml, 0.453 mmol) were reacted to afford the title compound as a yellow oil (56.0 mg, 40%). LCMS (Method F): RT = 2.07 min, M+H+ = 466.
Step 2: 1-(6-(4-(cyclopentyloxy)-3-isopropylphenyl)pyridin-2-yl)piperazine
dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(6-(4-(cyclopentyloxy)-3- isopropylphenyl)pyridin-2-yl)piperazine-1 -carboxylate (56 mg, 0.120 mmol) was reacted to afford the title compound as a yellow solid (47.1 mg, 89%). LCMS (Method F): RT = 1 .17 min, M+H+ = 366; 1H NMR (500 MHz, MeOH-d4): 8.10-8.07 (1 H, dd), 7.71 -7.68 (2H, m), 7.35-7.34 (1 H, d), 7.25-7.23 (1 H, d), 7.13-7.1 1 (1 H, d), 5.02- 4.97 (1 H, br m), 4.03-4.01 (4H, t), 3.49-3.47 (4H, t), 3.36-3.33 (1 H, sept), 2.05-1 .72 (8H, m), 1 .30-1 .29 (6H, d).
Example 76: (rac)-1 -(6-(3-isopropyl-4-((1 .1 ,1 -trifluoropropan-2-yl)oxy)phenyl)pyridin-2- vDpiperazine dihydrochloride
Figure imgf000108_0001
Step 1: (rac)-tert-butyl 4-(6-(3-isopropyl-4-((1 , 1, 1-trifluoropropan-2-yl)oxy)phenyl)pyridin- 2-yl)piperazine- 1 -carboxylate:
Following the procedure for tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate, tert-butyl 4-(6-(4-hydroxy-3-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.302 mmol) and (rac)-1 ,1 , 1 -trif luoropropan-2-yl trifluoromethanesulfonate (360 mg, 0.453 mmol) were reacted to afford the title compound as a yellow oil (28.7 mg, 19%).
LCMS (Method F): RT = 1 .93 min, M+H+ = 494.
Step 2 (rac)-1 -(6-(3-isopropyl-4-((1 , 1, 1-trifluoropropan-2-yl)oxy)phenyl)pyridin-2- yl)piperazine dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, (rac)-tert-butyl 4-(6-(3-isopropyl-4-((1 ,1 ,1 -trifluoropropan-2- yl)oxy)phenyl)pyridin-2-yl)piperazine-1 -carboxylate (28.7 mg, 0.058 mmol) was reacted to afford the title compound as a yellow solid (18.3 mg, 68%).
LCMS (Method F): RT = 1 .09 min, M+H+ = 394; 1H NMR (500 MHz, MeOH-d4): 8.05-8.02 (1 H, dd), 7.81 -7.80 (1 H, d), 7.77-7.75 (1 H, dd), 7.35-7.33 (1 H, d), 7.25-7.23 (1 H, d), 7.22-7.21 (1 H, d), 5.19-5.14 (1 H, sept), 4.03-4.01 (4H, t), 3.48-3.46 (4H, t), 3.43-3.38 (1 H, sept), 1 .57-1 .55 (3H, d), 1 .32-1 .31 (6H, d). Example 77: (rac)-1 -(6-(4-(sec-butoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000109_0001
Step 1: (rac)-tert-butyl 4-(6-(4-(sec-butoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine- 1- carboxylate:
Following the procedure for tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate, (rac)-tert-butyl 4-(6-(4-hydroxy-3-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.302 mmol) and 2-iodobutane (0.052 ml, 0.453 mmol) were reacted to afford the title compound as a yellow oil (39.9 mg, 29%).
LCMS (Method F): RT = 2.03 min, M+H+ = 454.
Step 2: (rac)-1-(6-(4-(sec-butoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine
dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, (rac)-tert-butyl 4-(6-(4-(sec-butoxy)-3- isopropylphenyl)pyridin-2-yl)piperazine-1 -carboxylate (39.9 mg, 0.088 mmol) was reacted to afford the title compound as a yellow solid (23.1 mg, 62%).
LCMS (Method F): RT = 1 .12 min, M+H+ = 354; 1H NMR (500 MHz, MeOH-d4): 7.88-7.84 (1 H, dd), 7.64 (1 H, d), 7.62-7.60 (1 H, dd), 7.21 -7.19 (1 H, d), 7.02-7.00 (1 H, d), 6.98-6.96 (1 H, d), 4.46-4.40 (1 H, m), 3.89-3.86 (4H, t), 3.35-3.33 (4H, t), 3.33-3.25 (1 H, sept), 1 .74-1 .58 (2H, m), 1 .23-1 .22 (3H, d), 1 .19-1 .17 (6H, dd), 0.95-0.92 (3H, t). Example 78: 4-(3-(tert-butyl)-4-(cvclopentyloxy)phenyl)-2-(piperazin-1 -yl) pyrimidine dihydrochloride
Figure imgf000110_0001
Following the procedure for 1 -(4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)pyrimidin-2- yl)piperidin-4-amine dihydrochloride, 4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)-2- chloropyrimidine (50 mg, 0.151 mmol) and piperazine (32.5mg, 0.378 mmol) were reacted to afford the title compound as a pale yellow solid (36 mg, 53%).
LCMS (Method F): RT = 1 .22 min, M+H+ = 381 ; 1H NMR (500 MHz, MeOH-d4): 8.35-8.34 (1 H, d), 8.25-8.24 (1 H, d), 8.21 -8.19 (1 H, dd), 7.61 -7.60 (1 H, d), 7.17-7.15 (1 H, d), 5.09- 5.05 (1 H, m), 4.29-4.27 (4H, t), 3.52-3.50 (4H, t), 2.12-1 .73 (8H, m), 1 .46 (9H, s).
Example 79: (1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4- vDmethan amine
Figure imgf000110_0002
Following the procedure for (R)-4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(3- methylpiperazin-1 -yl)pyrimidine, 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) and tert-butyl (piperidin-4-ylmethyl)carbamate (70.3 mg, 0.328 mmol) were reacted to afford the title compound as a yellow gum (63 mg, 95%).
LCMS (Method F): RT = 0.97 min, M+H+ = 383; 1H NMR (500 MHz, CDCI3): 8.30-8.29 (1 H, d), 8.04-8.03 (1 H, d), 7.86-7.84 (1 H, dd), 6.92-6.90 (1 H, d), 6.84-6.83 (1 H, d), 6.94- 6.91 (2H, br d), 4.74-4.70 (1 H, sept), 2.95-2.88 (2H, m), 2.63-2.62 (2H, d), 1 .88-1 .82 (2H, m), 1 .66-1 .56 (1 H, m), 1 .48-1 .47 (2H, d), 1 .43 (9H, s), 1 .41 -1 .40 (6H, d), 1 .27-1 .19 (2H, m). Example 80: 2-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-2,8- diazaspiro[4.51decane
Figure imgf000111_0001
Following the procedure for (fi)-4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(3- methylpiperazin-1 -yl)pyrimidine, 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) and tert-butyl 2,8-diazaspiro[4.5]decane-8-carboxylate (79 mg, 0.328 mmol) were reacted to afford the title compound as a yellow gum (3.2 mg, 5%). LCMS (Method F): RT = 1 .03 min, M+H+ = 409; 1H NMR (500 MHz, CDCI3): 8.23-8.22 (1 H, d), 7.99-7.96 (1 H, m), 7.83-7.75 (1 H, m), 6.86-6.78 (2H, m), 4.67-4.63 (1 H, m), 3.97-3.52 (6H, m), 3.1 1 -3.07 (2H, m), 3.01 -2.96 (2H, m), 2.73-2.57 (1 H, m), 2.04-2.02 (1 H, m), 1 .88-1 .85 (1 H, t), 1 .78-1 .76 (2H, t), 1 .36 (9H, s), 1 .34-1 .33 (6H, d). Example 81 : 1 -(6-(3-isopropyl-4-(2.2.2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000111_0002
Step 1: tert-butyl 4-(6-(3-isopropyl-4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazin 1-carboxylate:
Following the procedure for tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate, tert-butyl 4-(6-(4-hydroxy-3-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.302 mmol) and 1 ,1 ,1 -trifluoro-2-iodoethane (0.045 ml, 0.453 mmol) were reacted to afford the title compound as a yellow oil (44.9 mg, 31 %). - I l l -
LCMS (Method F): RT = 1 .89 min, M+H+ = 480.
Step 2: 1-(6-(3-isopropyl-4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazin
dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(6-(3-isopropyl-4-(2,2,2- trifluoroethoxy)phenyl)pyridin-2-yl)piperazine-1 -carboxylate (44.9 mg, 0.094 mmol) was reacted to afford the title compound as a yellow solid (28.7 mg, 68%).
LCMS (Method F): RT = 1 .01 min, M+H+ = 380; 1H NMR (500 MHz, MeOH-d4): 8.08-8.04 (1 H, dd), 7.81 -7.76 (2H, m), 7.35-7.34 (1 H, d), 7.25-7.23 (1 H, d), 7.19-7.18 (1 H, d), 4.71 - 4.58 (2H, q), 4.04-4.00 (4H, m), 3.49-3.45 (4H, m), 3.45-3.40 (1 H, sept), 1 .33-1 .32 (6H, d).
Example 82: 1 -(6-(3-(tert-butyl)-4-(2.2.2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000112_0001
Step 1: tert-butyl 4-(6-(3-(ten-butyl)-4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2- yl)piperazine- 1 -carboxylate:
Following the procedure for tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate, tert-butyl 4-(6-(3-(tert-butyl)-4-hydroxyphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.292 mmol) and 1 ,1 ,1 -trifluoro-2-iodoethane (0.043 ml, 0.437 mmol) were reacted to afford the title compound as a yellow oil (51 .2 mg, 36%).
LCMS (Method F): RT = 1 .93 min, M+H+ = 494.
Step 2: 1 -(6-(3-(ten-butyl)-4-(2,2,2-trifluoroethoxy)phenyl)pyrid^
dihydrochloride: Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(6-(3-(tert-butyl)-4-(2,2,2- trifluoroethoxy)phenyl)pyridin-2-yl)piperazine-1 -carboxylate (51 .2 mg, 0.104 mmol) was reacted to afford the title compound as a yellow solid (37.1 mg, 77%).
LCMS (Method F): RT = 1 .07 min, M+H+ = 394; 1H NMR (500 MHz, MeOH-d4): 8.03-8.01 (1 H, dd), 7.89 (1 H, d), 7.89-7.81 (1 H, dd), 7.34-7.32 (1 H, d), 7.20-7.15 (2H, m), 4.71 - 4.67 (2H, q), 4.03-3.99 (4H, m), 3.48-3.45 (4H, m), 1 .48 (9H, s).
Example 83: 1 -(2-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine dihydrochloride
Figure imgf000113_0001
Step 1: tert-butyl (1-(2-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4- yl) carbarn ate:
To a solution of 2-(4-isopropoxy-3-isopropylphenyl)-4,4,5,5-tetramethyl-1 ,3,2- dioxaborolane (60 mg, 0.197 mmol) in dioxane (1 ml) was added tert-butyl (1 -(2- chloropyrimidin-4-yl)piperidin-4-yl)carbamate (51 .4 mg, 0.164 mmol) and 2M aq Na2C03 (0.329 ml, 0.657 mmol). Nitrogen was bubbled through the mixture, and
PdCI2(dppf) CH2CI2 (13.42 mg, 0.016 mmol) was added. The mixture was heated to 150 ° for 30 min. under microwave conditions. After cooling to RT, the reaction mixture was partitioned between DCM and water. The organic phase was dried, using a phase separator cartridge, and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 40 % EtOAc in cyclohexane) to afford the title compound as a yellow oil (28.9 mg, 39%).
LCMS (Method F): RT = 1 .23 min, M+H+ = 455.
Step 2: 1 -(2-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4-ami
dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl (1 -(2-(4-isopropoxy-3-isopropylphenyl)py 4-yl)piperidin-4-yl)carbamate (28.9 mg, 0.064 mmol) was reacted to afford the title compound as an off-white solid (1 1 .3 mg, 77%).
LCMS (Method F): RT = 0.69 min, M+H+ = 355; 1H NMR (500 MHz, MeOH-d4): 8.08-8.06 (1 H, d), 7.99-7.97 (1 H, dd), 7.94 (1 H, d), 7.08-7.06 (1 H, d), 7.00-6.98 (1 H, d), 4.72-4.70 (1 H, m), 3.53-3.48 (1 H, m), 3.30-3.20 (5H, m), 2.18-2.16 (2H, br d), 1 .64-1 .61 (2H, m), 1 .31 -1 .30 (6H, d), 1 .19-1 .18 (6H, d).
Example 84: 1 -(6-(4-(cvclopropylmethoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride
Figure imgf000114_0001
Step 1: tert-butyl 4-(6-(4-(cyclopropylmethoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine- 1-carboxylate:
Following the procedure for tert-butyl 4-(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate, tert-butyl 4-(6-(4-hydroxy-3-isopropylphenyl)pyridin-2- yl)piperazine-1 -carboxylate (120 mg, 0.302 mmol) and (bromomethyl)cyclopropane (0.059 ml, 0.604 mmol) were reacted to afford the title compound as a pink oil (61 .4 mg, 45%).
LCMS (Method F): RT = 1 .97 min, M+H+ = 452.
Step 2: 1-(6-(4-(cyclopropylmethoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine dihydrochloride:
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(6-(4-(cyclopropylmethoxy)-3- isopropylphenyl)pyridin-2-yl)piperazine-1 -carboxylate (61 .4 mg, 0.136 mmol) was reacted to afford the title compound as a yellow solid (38.3 mg, 66%).
LCMS (Method F): RT = 1 .03 min, M+H+ = 352; 1H NMR (500 MHz, MeOH-d4): 7.80-7.77 (1 H, dd), 7.67 (1 H, d), 7.64-7.62 (1 H, dd), 7.18-7.17 (1 H, d), 6.94-6.90 (2H, m), 3.86- 3.83 (6H, m), 3.33-3.29 (5H, m), 1 .26-1 .22 (1 H, m), 1 .20-1 .19 (6H, d), 0.57-0.53 (2H, m), 0.31 -0.29 (2H, m). Example 85: 1 -(6-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine dihydrochloride
Figure imgf000115_0001
Step 1: tert-butyl 1-(6-chloropyrimidin-4-yl)piperidin-4-ylcarbamate:
To a solution of 4,6-dichloropyrimidine (0.5 g, 3.36 mmol) in acetonitrile (10 ml) was added tert-butyl piperidin-4-ylcarbamate (0.74 g, 3.69 mmol) and triethylamine (0.47 ml, 3.36 mmol). The mixture was heated at 120 °C for 2 h under microwave conditions, and then concentrated in vacuo. The residue was partitioned between EtOAc and water. The aqueous layer was extracted with EtOAc and the combined organic extracts were washed with aq. ammonium chloride, dried (MgS04) and concentrated in vacuo to afford the title compound as a white solid (0.98 g, 93%).
1H NMR (500 MHz, CDCI3): 8.00 (1 H, s), 7.80 (1 H, s), 4.53 (1 H, br s), 4.27-4.24 (2H, d), 3.75 (1 H, br s), 3.10-3.04 (2H, m), 2.09-2.07 (2H, m), 1 .47 (9H, s), 1 .44-1 .39 (2H, m).
Step 2: tert-butyl (1-(6-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4- yl) carbarn ate:
Following the procedure for tert-butyl (1 -(2-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4- yl)piperidin-4-yl)carbamate, 2-(4-isopropoxy-3-isopropylphenyl)-4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolane (75 mg, 0.247 mmol) and tert-butyl (1 -(6-chloropyrimidin-4- yl)piperidin-4-yl)carbamate (64.3 mg, 0.205 mmol) were reacted to afford the title compound as a yellow oil (81 .0 mg, 87%).
LCMS (Method F): RT = 1 .22 min, M+H+ = 455.
Step 3: 1-(6-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine dihydrochloride
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl (1 -(6-(4-isopropoxy-3-isopropylphenyl)pyrimidin- 4-yl)piperidin-4-yl)carbamate (81 .0 mg, 0.178 mmol) was reacted to afford the title compound as a yellow solid (53.8 mg, 70%).
LCMS (Method F): RT = 0.70 min, M+H+ = 355; 1H NMR (500 MHz, MeOH-d4): 8.56 (1 H, s), 7.67-7.64 (2H, m), 7.23 (1 H, s), 7.08-7.06 (1 H, d), 4.72-4.68 (1 H, m), 3.50-3.47 (1 H, m), 3.29-3.20 (5H, m), 2.16-2.14 (2H, br d), 1 .63-1 .60 (2H, m), 1 .30-1 .29 (6H, d), 1 .18- 1 .17 (6H, d).
Example 86: 2-(4-isopropoxy-3-isopropylphenyl)-4-(piperazin-1 -yl)-1 ,3,5-triazine dihydrochloride
Figure imgf000116_0001
Step 1: tert-butyl 4-(4-chloro- 1 ,3,5-triazin-2-yl)piperazine- 1 -carboxylate:
To a solution of 2,4-dichloro-1 ,3,5-triazine (200 mg, 1 .334 mmol) in acetonitrile (2 ml) at 0 °C was added Et3N (0.279 ml, 2.000 mmol) and tert-butyl piperazine-1 -carboxylate (248 mg, 1 .334 mmol). The mixture was warmed to RT and stirred for 2 h. The mixture was partitioned between EtOAc and brine. The aqueous layer was extracted with EtOAc and the combined organic extracts were dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 100% EtOAc in cyclohexane) to afford the title compound as a white solid (227 mg, 37%).
1H NMR (500 MHz, CDCI3): 8.39 (1 H, s), 3.90-3.86 (4H, m), 3.54-3.52 (4H, m), 1 .51 (9H, s).
Step 2: tert-butyl 4-(4-(4-isopropoxy-3-isopropylphenyl)-1 ,3,5-triazin-2-yl)piperazine-1 - carboxylate:
Following the procedure for tert-butyl (1 -(2-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4- yl)piperidin-4-yl)carbamate, 2-(4-isopropoxy-3-isopropylphenyl)-4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolane (50 mg, 0.164 mmol) and tert-butyl 4-(4-chloro-1 ,3,5-triazin-2- yl)piperazine-1 -carboxylate (41 .1 mg, 0.137 mmol) were reacted to afford the title compound as an off-white solid (5.4 mg, 9%).
LCMS (Method F): RT = 1 .91 min, M+H+ = 442. Step 3: 2-(4-isopropoxy-3-isopropylphenyl)-4-(piperazin- 1-yl)- 1 ,3,5-triazine
dihydrochloride
Following the procedure for 4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 - yl)pyrimidine dihydrochloride, tert-butyl 4-(4-(4-isopropoxy-3-isopropylphenyl)-1 ,3,5- triazin-2-yl)piperazine-1 -carboxylate (6 mg, 0.014 mmol) was reacted to afford the title compound as a white solid (5.8 mg, >100%).
LCMS (Method F): RT = 1 .03 min, M+H+ = 342; 1H NMR (500 MHz, MeOH-d4): 8.80 (1 H, s), 8.24-8.22 (1 H, dd), 8.18-8.17 (1 H, d), 7.18-7.16 (1 H, d), 4.84-4.81 (1 H, sept), 4.45- 4.35 (4H, m), 3.50-3.40 (4H, m), 3.45-3.39 (1 H, sept), 1 .41 -1 .40 (6H, d), 1 .25-1 .24 (6H, d).
Example 87: (rac)-(1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)pyrrolidin-3- vDmethan amine
Figure imgf000117_0001
Following the procedure for (/:?)-4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(3- methylpiperazin-1 -yl)pyrimidine, 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) and (rac)-tert-butyl (pyrrolidin-3-ylmethyl)carbamate (82 mg, 0.410 mmol) were reacted to afford the crude title compound. Further purification was carried out, using preparative HPLC (Methods C & D) to afford the title compound as a white solid (3.4 mg, 5.7%).
LCMS (Method F): RT = 0.93 min, M+H+ = 369; 1H NMR (500 MHz, CDCI3): 8.33-8.32 (1 H, d), 8.08 (1 H, br s), 7.91 -7.89 (1 H,dd), 6.94-6.92 (1 H, d), 6.89-6.88 (1 H, d), 4.76- 4.74 (1 H, sept), 3.95-3.80 (2H, br m), 3.70-3.60 (1 H, br m), 3.45-3.35 (1 H, br m), 2.88- 2.85 (2H, m), 2.50-2.40 (1 H, m), 2.25-2.15 (1 H, m), 1 .80-1 .70 (1 H, m), 1 .46 (9H, s), 1 .44- 1 .43 (6H, d). Example 88: 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-4-phenylpiperidin-4- amine
Figure imgf000118_0001
Step 1: 1-(4-(3-(tei1-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-ol:
Following the procedure for 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)piperazin-1 -yl)ethanol, 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (500 mg, 1 .640 mmol) and piperidin-4-ol (332 mg, 3.28 mmol) were reacted to afford the title compound as a yellow oil (714 mg, >100%) which was used without further purification. LCMS (Method F): RT = 1 .51 min, M+H+ = 370.
Step 2: 1-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperid
To a solution of 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-ol (606 mg, 1 .640 mmol) and Et3N (0.571 ml, 4.10 mmol) in DMSO (10 ml), was added sulfur trioxide pyridine complex (1 .149 g, 7.22 mmol). The reaction mixture was stirred at RT for 18 h. Further Et3N (0.571 ml, 4.10 mmol) and sulfur trioxide pyridine complex (1 149 mg, 7.22 mmol) were added and the reaction was stirred at RT for a further 18 h. The reaction mixture was partitioned between EtOAc and water. The organic extracts were washed with water (6 times), dried (MgS04), and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 50% EtOAc in cyclohexane) to afford the title compound as a yellow oil (100 mg, 17 %).
1H NMR (500 MHz, CDCI3): 8.39-8.38 (1 H, d), 8.07 (1 H, s), 7.92-7.90 (1 H, dd), 7.01 -7.00 (1 H, d), 6.96-6.95 (1 H, d), 4.80-4.70 (1 H, sept), 4.27-4.24 (4H, m), 2.58-2.56 (4H, m), 1 .46 (9H, s), 1 .45-1 .44 (6H, d).
Step 3: N-( 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin
methylpropane-2-sulfinamide:
To a solution of 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-one (100 mg, 0.272 mmol) and 2-methylpropane-2-sulfinamide (36.3 mg, 0.299 mmol) in DCE (2 ml), was added Ti(OEt)4 (0.068 ml, 0.299 mmol). The reaction mixture was heated to 85 °C for 18 h. The reaction mixture was diluted with DCM, and sat. aq.
NaHC03 solution was added. This mixture was stirred at room temperature for 5 min. and the resulting mixture was filtered through celite, the layers separated using a phase separator and the organic filtrates concentrated in vacuo to afford the title compound as a yellow oil (1 18 mg, 92%). Used without further purification.
LCMS (Method F): RT = 1 .76 min, M+H+ = 471 .
Step 4: 1-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin^
To a solution of Phenylmagnesiumbromide, 1 M in THF (0.351 ml, 0.351 mmol) in THF (1 ml) at 0 °C, was slowly added a solution of N-(1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)- pyrimidin-2-yl)piperidin-4-ylidene)-2-methylpropane-2-sulfinamide (55 mg, 0.1 17 mmol) in THF (1 .0 ml). The reaction mixture was warmed to RT, and stirred at RT for 18 h. The mixture was partitioned between DCM and sat. aq. NaHC03, and filtered through a phase separator cartridge. 4M HCI in dioxane (~1 .5ml) was added to the organic filtrate, and the solution stirred for 1 .5 h. The mixture was neutralized with sat. aq. NaHC03, extracted into DCM, and filtered through a phase separator cartridge. The organic filtrate was concentrated in vacuo. Purification was carried out, using preparative HPLC
(Methods C & D), to afford the title compound as a white solid (3.5 mg, 6.7%).
LCMS (Method F): RT = 1 .82 min, M+H+ = 446; 1H NMR (500 MHz, CDCI3): 8.28-8.27 (1 H, d), 8.01 -8.00 (1 H, d), 7.84-7.82 (1 H, dd), 7.49-7.47 (2H, dd), 7.34-7.31 (1 H, t), 7.20- 7.19 (1 H, m), 6.88-6.86 (1 H, d), 6.83-6.82 (1 H, d), 4.70-4.67 (1 H, sept), 4.51 -4.48 (2H, m), 3.64-3.59 (2H, dt), 2.18-2.12 (2H, dt), 1 .77-1 .74 (2H, m), 1 .39 (9H, s), 1 .37-1 .36 (6H, d). Example 89: 4-(piperidin-4-yloxy)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- vDpyrimidine di(trifluoroacetate)
Figure imgf000119_0001
Step 1: 4-chloro-6-(piperidin-4-yloxy)pyrimidine: Following the procedure for te/t-butyl 4- ((4-chloropyrimidin-2-yl)oxy)piperidine-1 -carboxylate, 4,6-dichloropyrimidine (1 .0 g , 6.71 mmol) and tert-butyl 4-hydroxypiperidine-1 -carboxylate (1 .35 g, 6.71 mmol) were reacted to afford the title compound as a white solid (1 .67 g, 80%).
1H NMR (500 MHz, CDCI3): 8.55 (1 H, s), 7.75 (1 H, s), 5.29-5.33 (1 H, m), 3.73-3.77 (2H, m), 1 .94-2.00 (2H, m), 1 .70-1 .76 (2H, m), 1 .47 (9H, s).
Step 2: tert-butyl 4-((6-(5,5,8,8-tetramethyl-5^ ,8-tetrahydronaphthalen-2-yl)pyrimidin- 4-yl)oxy)piperidine-1-carboxylate: Following the procedure for tert-butyl 4-((4-(5,5,8,8- tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)oxy)piperidine-1 - carboxylate, 4-chloro-6-(piperidin-4-yloxy)pyrimidine (0.1 g, 0.39 mmol) and 4,4,5,5- tetramethyl-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ,3,2- dioxaborolane (0.1 g, 0.39 mmol) were reacted to afford the title compound as a clear oil (0.6 g, 40%).
LCMS (Method F): RT = 2.06 min, M+H+ = 466.
Step 3: 4-(piperidin-4-yloxy)-6-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidine di(trifluoroacetate):
Following the procedure for 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)phenyl)piperazine di(trifluoroacetate , tert-butyl 4-((6-(5,5, 8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidin-4-yl)oxy)piperidine-1 -carboxylate (60 mg , 0.13 mmol) was reacted to afford the title compound as a white solid (23 mg, 48%).
LCMS (Method F): RT = 1 .1 1 min, M+H+ = 366; 1H NMR (500 MHz, MeOH-d4): 8.66 (1 H, s), 7.95 (1 H, d), 7.67 (1 H, d), 7.38 (1 H, d), 7.19 (1 H, s), 5.40-5.43 (1 H, m), 3.33-3.38 (2H, m), 3.16-3.19 (2H, m), 2.14-2.16 (2H, m), 2.01 -2.04 (2H, m), 1 .65 (4H, s), 1 .25 (6H, s), 1 .22 (6H, s).
Example 90: 4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyrimidin-2- vDbenzimidamide
Figure imgf000120_0001
Step 1: 4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)benzonitrile:
Following the procedure for tert-butyl 1 -(4-(6-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)phenyl)cyclobutylcarbamate, (4- cyanophenyl)boronic acid (0.49 g, 3.32 mmol) and 2-chloro-4-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine (1 .0 g, 3.32 mmol) were reacted to afford the title compound (766 mg, 63%).
LCMS (Method F): RT = 1 .97 min, M+H+ = 368. Step 2: 4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)benzimidamide:
A solution of LHMDS (1 .09 ml, 1 .09 mmol) in diethyl ether (3 ml) under a nitrogen atmosphere was cooled to 0 'Ό and 4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro- naphthalen-2-yl)pyrimidin-2-yl)benzonitrile (0.2 g, 0.54 mmol) in diethyl ether (3 ml) added dropwise. The solution was warmed to RT over 2 hours and left to stir for 16 hours. The solution was added to 100 ml 1 M HCI (aq) then washed with diethylether (2x 100 ml). The aqueous layer was basified to pH 12 with Na2C03 (aq) and extracted with EtOAc (2x 100 ml), dried (MgS04) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (KP-NH Si02, gradient 0 to 10% MeOH in EtOAc) to afford the title compound as an off white solid (125 mg, 65%).
LCMS (Method F): RT = 1 .31 min, M+H+ = 385; 1H NMR (500 MHz, MeOH-d4): 8.87 (1 H, d), 8.62-8.64 (2H, d), 8.31 (1 H, d), 8.04-8.06 (1 H, dd), 7.91 -7.92 (2H, d), 7.85-7.83 (1 H, d), 7.57-7.55 (1 H, d), 1 .80 (4H, s), 1 .42 (6H, s), 1 .37 (6H, s). Example 91 : 1 -(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyrimidin-2- yl)piperazin-1 -vDethanimine dihydrochloride
Figure imgf000121_0001
A solution of 2-(piperazin-1 -yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidine (0.1 g, 0.285 mmol) and ethyl acetimidate hydrochloride (0.035 g, 0.285 mmol) in DMF (3 ml) was stirred at room temperature for 3 days. The solution was then concentrated in vacuo and the residue purified by flash chromatography. The resulting product was treated with HCI (4N in dioxane, 1 ml). The resulting precipitate was filtered and dried at ambient temperature to afford the title compound as a white solid (53 mg, 43%).
LCMS (Method F): RT = 1 .32 min, M+H+ = 392; 1H NMR (500 MHz, DMSO-d6): 9.59 (s, 1 H), 8.95 (s, 1 H), 8.48 (d, 1 H), 8.09 (s, 1 H), 7.89 (d, 1 H), 7.48 (d, 1 H), 7.36 (d, 1 H), 4.00 (m, 4H), 3.78 (m, 4H), 2.37 (s, 3H), 1 .77 (s, 4H), 1 .32 (s, 6H), 1 .28 (s, 6H).
Example 92 : 4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyrimidin-2- yl)piperazine-1 -carboximidamide dihydrochloride
Figure imgf000122_0001
Step 1 : tert-butyl (4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin- 2-yl)piperazin- 1 -yl)methanediylidenedicarbamate:
A solution of 2-(piperazin-1 -yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidine (0.60 g, 1 .79 mmol), 1 ,3-bis(te/?-butoxycarbonyl)-2-methyl-2- thiopseudourea (0.54 g, 1 .88 mmol), mercury chloride (0.51 g, 1 .97 mmol) and triethylamine (0.596 ml, 4.28 mmol) in DMF (5 ml) was stirred at room temperature overnight. The solution was diluted with ethylacetate (30 ml) and the mercury sulfide precipitate was filtered through a phase separator cartridge. The filtrate was washed with water, dried over magnesium sulfate and concentrated in vacuo. The residue was purified by flash chromatography to afford the title compound as a colourless oil (0.55 g, 51 %).
LCMS (Method F): RT = 1 .99 min, M+H+ = 593; 1H NMR (500 MHz, CDCI3): 10.23 (s, 1 H), 8.35 (d, 1 H), 7.96 (s, 1 H), 7.76 (d, 1 H), 7.40 (d, 1 H), 6.94 (d, 1 H), 4.00 (m, 4H), 3.71 (m, 4H), 2.37 (s, 3H), 1 .72 (s, 4H), 1 .51 , (s, 9H), 1 .49 (s, 9H), 1 .31 (s, 6H), 1 .26 (s, 6H).
Step 2 : 4-(4-(5!5A8-tetramethyl-5^ ,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)piperazine-1 -carboximidamide dihydrochloride: A solution of tert-butyl (4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidin-2-yl)piperazin-1 -yl)methanediylidenedicarbamate (80 mg, 0.135 mmol) and HCI (4N in dioxane 1 ml) was stirred at 65 °C for 30 min then concentrated in vacuo. The resulting residue was triturated in diethyl ether. The solvent was decanted and the resulting solid was dried at ambient temperature to afford the title compound (50 mg, 80%).
LCMS (Method F): RT = 1 .08 min, M+H+ = 393; 1H NMR (500 MHz, DMSO-d6): 8.46 (d, 1 H), 8.09 (s, 1 H), 7.90 (m, 1 H), 7.78 (s, 4H) 7.49 (d, 1 H), 7.40 (d, 1 H), 4.02 (m, 4H), 3.71 (m, 4H), 1 .68 (s, 4H),1 .31 (s, 6H), 1 .27 (s, 6H).
Example 93 : 1 -(1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaDhthalen-2-yl)Dyridin-2- yl)piperidin-4-yl)quanidine dihydrochloride
Figure imgf000123_0001
Step 1 : tert-butyl (tert-butoxycarbonylamino)(1-(6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4-ylam
Following the procedure for tert-butyl (4-(4-(5,5, 8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazin-1 -yl)methanediylidenedicarbamate, 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4-amine dihydrochloride (1 g, 2.29 mmol) and 1 ,3-bis(te/?-butoxycarbonyl)-2-methyl-2- thiopseudourea (0.73 g, 2.52 mmol) were reacted to afford the title compound as an off- white solid (1 .15 g, 83%).
LCMS (Method F): RT = 2.23 min, M+H+ = 606; 1H NMR (500 MHz, CDCI3): 1 1 .56 (s, 1 H), 8.41 (d, 1 H), 7.96 (s, 1 H), 7.75 (d, 1 H), 7.54 (m, 1 H), 7.40 (d, 1 H), 7.06 (d, 1 H), 6,62 (d, 1 H), 4.31 (m, 1 H), 4.29 (m, 2H), 3.22 (m, 2H), 2.1 1 (m, 2H), 1 .72 (s, 4H), 1 ,60 (m, 2H), 1 .51 , (s, 9H), 1 .49 (s, 9H), 1 .31 (s, 6H), 1 .26 (s, 6H).
Step 2 : 1-(1-(6-(5 -tetramethyl-5fi -tetrahydronaphthalen-2-yl)pyridin-2- yl)piperidin-4-yl)guanidine dihydrochloride: Following the procedure for 4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidin-2-yl)piperazine-1 -carboximidamide dihydrochloride, tert-butyl (tert- butoxycarbonylamino)(1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyridin-2-yl)piperidin-4-ylamino)methylenecarbamate (0.2 g, 0.33 mmol) was reacted to afford the title compound as a white solid (0.13 g, 82%).
LCMS (Method F): RT = 1 .30 min, M+H+ = 406; 1H NMR (500 MHz, DMSO-d6): 8.22 (brs, 2H), 7.85 (s, 1 H), 7.76 (brs, 1 H), 7.63 (d, 1 1-1) 7.45 (d, 1 H), 7.16 (d, 1 H), 7.04 (brs, 1 H), 6.3-5.5 (brs, 2H), 4.26 (m, 2H), 3.80 (m, 1 H), 3.21 (m, 2H), 1 .96 (m, 2H), 1 .68 (s, 4H), 1 .51 (m, 2H) 1 .31 (s, 6H), 1 .27 (s, 6H).
Example 94 : 1 -(1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2-yl)pyrimidin-2- yl)piperidin-4-yl)quanidine dihydrochloride
Figure imgf000124_0001
Step 1: tert-butyl (tert-butoxycarbonylamino)(1-(4-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperidin-4-ylam
Following the procedure for tert-butyl (4-(4-(5,5, 8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazin-1 -yl)methanediylidenedicarbamate 1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperidin-4- amine dihydrochloride (0.6 g, 1 .37 mmol) and 1 ,3-bis(te/?-butoxycarbonyl)-2-methyl-2- thiopseudourea (0.44 g, 1 .51 mmol) were reacted to afford the title compound as an off- white solid (0.68 g, 82%).
LCMS (Method F): RT = 2.42 min, M+H+ = 607; 1H NMR (500 MHz, CDCI3): 1 1 .56 (s, 1 H), 8.42 (d, 1 H), 8.35 (d, 1 H), 7.98 (s, 1 H), 7.78 (d, 1 H), 7.42 (d, 1 H), 6,91 (d, 1 H), 4.71 (m, 2H), 4.40 (m, 1 H), 3.31 (m, 2H), 2.1 1 (m, 2H), 1 .74 (s, 4H), 1 ,55 (s, 9H), 1 .50, (s, 9H), 1 .50 (m, 2H), 1 .37 (s, 6H), 1 .33 (s, 6H).
Step 2:_ 1-(1-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)piperidin-4-yl)guanidine dihydrochloride:
Following the procedure for 4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidin-2-yl)piperazine-1 -carboximidamide dihydrochloride, tert-butyl (tert- butoxycarbonylamino)(1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidin-2-yl)piperidin-4-ylamino)methylenecarbamate (0.16 g, 0.26 mmol) was reacted to afford the title compound as a white solid (0.13 g, 98%).
LCMS (Method F): RT = 1 .33 min, M+H+ = 407; 1H NMR (500 MHz, DMSO-d6): 8.41 (d, 1 H), 8.1 1 (d, 1 H), 8.07 (s, 1 H), 7.90 (d, 1 H) 7.50 (d, 1 H), 7.36 (d, 1 H), 7.04 (brs, 1 H), 6.0-5.5 (brs, 2H), 4.60 (m, 2H), 3.80 (m, 1 H), 3.21 (m, 2H), 1 .96 (m, 2H), 1 .46 (s, 4H), 1 .48 (m, 2H) 1 .31 (s, 6H), 1 .28 (s, 6H).
Example 95 : N'-cvano-4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahvdronaphthalen-2- yl)pyrimidin-2-yl)piperazine-1 -carboximidamide
Figure imgf000125_0001
Step 1 : phenyl N-cyano-4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidin-2-yl)piperazine- 1 -carbimidate:
A solution of 2-(piperazin-1 -yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidine (0.24 g, 0.68 mmol), diphenyl cyanocarbonimidate (0.16 g, 0.68 mmol) and triethylamine (0.095 ml, 0.68 mmol) in acetonitrile (5 ml) was heated at 50 °C overnight then concentrated in vacuo. The crude product was used in the next step without further purification.
LCMS (Method F): RT = 1 .87 min, M+H+ = 495.
Step 2 : N'-cyano-4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2-yl)pyrimidin- 2-yl)piperazine- 1 -carboximidamide:
A solution of phenyl A/-cyano-4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidin-2-yl)piperazine-1 -carbimidate (0.25 g, 0.5 mmol) and ammonia (7N in methanol, 10 ml) was heated in a sealed flask at 1 10 °C for 5h. The resulting precipitate was filtered and dried at ambient temperature.
LCMS (Method F): RT = 2.23 min, M+H+ = 418 ; 1H NMR (500 MHz, DMSO-d6): 8.42 (d, 1 H), 8.02 (s, 1 H), 7.85 (d, 1 H), 7.45 (d, 1 H), 7.22 (m, 3H), 4.00 (m, 4H), 3.85 (m, 4H), 3.59 (m, 4H), 1 .68 (s, 4H), 1 .31 , (s, 6H), 1 .27 (s, 6H). Example 96 : 4-(4-(3-(tert-butyl)-4-(prop-1 -en-2-yl)phenyl)pyrimidin-2-yl)piperazine-1 - carboximidamide dihydrochloride
Figure imgf000126_0001
Step 1 : 2-(tert-butyl)-4-(2-chloropyrimidin-4-yl)phenol:
A solution of 4-bromo-2-(tert-butyl)phenol (6.5 g, 28 mmol), potassium acetate (8.35 g, 85 mmol), 4,4,4',4', 5,5,5', 5'-octamethyl-2,2'-bi(1 ,3,2-dioxaborolane (7.2 g, 28.4 mmol) and PdCI2(dppf)-CH2CI2adduct (1 .57 g, 2.84 mmol) in dioxane (100 ml) was heated at 100 °C overnight. An aqueous solution of sodium carbonate (2N, 42.6 ml, 42.5 mmol), PdCI2(dppf)-CH2CI2adduct (1 .57g, 2.84 mmol) and 2,4-dichloropyrimidine (4.2 g, 28.4 mmol) were then added. The resulting solution was heated at 100 °C for 4h. The organic phase was then decanted, dried over MgS04 and concentrated in vacuo. The residue was purified by flash chromatography to afford the title compound (2.38 g, 32%).
1H NMR (500 MHz, DMSO-d6): 10.3 (s, 1 H), 8.65 (d, 1 H), 8.01 (s, 1 H), 7.97 (d, 1 H), 7.90 (d, 1 H), 6.95 (d, 1 H), 1 .40 (s, 9H).
Step 2 : tert-butyl 4-(4-(3-(tert-butyl)-4-hydroxyphenyl)pyrimidin-2-yl)piperazine- 1- carboxylate:
A solution of 2-(tert-butyl)-4-(2-chloropyrimidin-4-yl)phenol (1 g, 3.81 mmol), tert-butyl piperazine-1 -carboxylate (1 .06 g, 5.71 mmol) and triethylamine (0.796 ml, 5.71 mmol) in
THF (6 ml) was heated at 140 °C for 25 min under microwave irradiation. The solution was diluted with ethyl acetate and water and the organic phase was decanted, dried over
MgS04, filtered and concentrated in vacuo. The residue was purified by flash
chromatography to afford the title compound (1 .35 g, 86%).
LCMS (Method F): RT = 1 .65 min, M+H+ = 413; 1H NMR (500 MHz, DMSO-d6): 9.90 (s,
1 H), 8.33 (d, 1 H), 7.98 (s, 1 H), 7.82 (d, 1 H), 7.08 (d, 1 H), 6.98 (d, 1 H), 1 .44 (s, 9H), 1 .40
(s, 9H).
Step 3 : tert-butyl 4-(4-(3-(tert-butyl)-4-(((trifluoromethyl)sulfonyl)oxy)phenyl)pyrimidi yl)piperazine- 1 -carboxylate: A solution of tert-butyl 4-(4-(3-(tert-butyl)-4-hydroxyphenyl)pyrimidin-2-yl)piperazine-1 - carboxylate (1 .35 g, 3.27 mmol), triflic anhydride (1 .016 g, 3.60 mmol) and triethylamine (0.68 ml, 3.91 mmol) in DCM (20 ml) and THF (20 ml) was stirred at RT overnight. The solution was concentrated in vacuo and the residue was purified by flash
chromatography to afford the title compound (1 .26 g, 71 %).
LCMS (Method F): RT = 1 .65 min, M+ = 489 (544 -tBu); 1H NMR (500 MHz, CDCI3): 8.40 (d, 1 H), 8.18 (s, 1 H), 7.92 (d, 1 H), 7.47 (d, 1 H), 6.93 (d, 1 H), 3.91 (m, 4H), 3.54 (m, 4H), 1 .50 (s, 9H), 1 .49 (s, 9H). Step 4 : tert-butyl 4-(4-(3-(tert-butyl)-4-(prop- 1 -en-2-yl)phenyl)pyrimidin-2-yl)piperazine-1 - carboxylate:
A solution of tert-butyl 4-(4-(3-(tert-butyl)-4-(((trifluoromethyl)sulfonyl)oxy)phenyl) pyrimidin-2-yl)piperazine-1 -carboxylate (1 g, 183 mmol), isopropenyl trifluoroborate (0. 4g, 2.75 mmol), tetrakis(triphenylphosphine)palladium(0) (0.02 g, 0.018 mmol) and cesium carbonate (1 .1 9g, 3.67 mmol) in water (5 ml) and toluene (20 ml) was heated at 95 °C overnight. The mixture was diluted with ethyl acetate and water and the organic phase was decanted, dried over MgS04, filtered and concentrated in vacuo. The residue was purified by flash chromatography to afford the title compound (0.5 g, 62.4 %).
LCMS (Method F): RT = 1 .65 min, M+H+ = 437 ; 1H NMR (500 MHz, CDCI3): 8.36 (d, 1 H), 8.14 (s, 1 H), 7.77 (d, 1 H), 7.09 (d, 1 H), 6.93 (d, 1 H), 5.23 (m, 1 H), 4.90 (m, 1 H), 3.91 (m, 4H), 3.54 (m, 4H), 2.04 (s, 3H), 1 .50 (s, 9H), 1 .49 (s, 9H).
Step 5 : 4-(3-(tert-butyl)-4-(prop-1-en-2-yl)phenyl)-2-(piperazin- 1-yl)pyrimidine
dihydrochloride:
A solution of tert-butyl 4-(4-(3-(tert-butyl)-4-(prop-1 -en-2-yl)phenyl)pyrimidin-2- yl)piperazine-1 -carboxylate (0.3 g, 0.68 mmol) and HCI (4N in dioxane, 3 ml) was heated at 50 °C for 30 min then concentrated in vacuo to afford the title compound (0.3 g, 100%).
LCMS (Method F): RT = 1 .07 min, M+H+ = 337.
Step 6 : tert-butyl (((tert-butoxycarbonyl)amino)(4-(4-(3-(tert-butyl)-4-(prop-1-en-2- yl)phenyl)pyrimidin-2-yl)piperazin- 1-yl)methylene)carbamate:
Following the procedure for tert-butyl (4-(4-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazin-1 yl)methanediylidenedicarbamate, 4- (3-(tert-butyl)-4-(prop-1 -en-2-yl)phenyl)-2-(piperazin-1 -yl)pyrimidine dihydrochloride (0.28 g, 0.068 mmol) was reacted to afford the title compound (0.2 g, 50%).
LCMS (Method F): RT = 2.01 min, M+H+ = 580 ; 1H NMR (500 MHz, CDCI3): 10.21 (s, 1 H), 8.36 (d, 1 H), 8.14 (s, 1 H), 7.77 (d, 1 H), 7.08 (d, 1 H), 6.95 (d, 1 H), 5.23 (m, 1 H), 4.90 (m, 1 H), 4.01 (m, 4H), 3.71 (m, 4H), 2.10 (s, 3H), 1 .50 (s, 9H), 1 .49 (s, 9H).
Step 7 : 4-(4-(3-(tert-butyl)-4-(prop-1-en-2-yl)phenyl)pyrimidin-2-yl)piperazine-1- carboximidamide dihydrochloride:
A solution of tert-butyl (((tert-butoxycarbonyl)amino)(4-(4-(3-(tert-butyl)-4-(prop-1 -en-2- yl)phenyl)pyrimidin-2-yl)piperazin-1 -yl)methylene)carbamate (0.2 g, 0.68 mmol) and HCI (4N in dioxane, 3 ml) was heated at 60 °C for 30 min then concentrated in vacuo to afford the title compound (0.15 g, 100%).
LCMS (Method F): RT = 1 .14 min, M+H+ = 379. 1H NMR (500 MHz, DMSO-d6): 8.50 (d, 1 H), 8.20 (s, 1 H), 7.90 (d, 1 H), 7.70 (s, 3H), 7.20 (d, 1 H), 7.10 (d, 1 H), 5.23 (m, 1 H), 4.85 (m, 1 H), 3.95 (m, 4H), 3.62 (m, 4H), 2.10 (s, 3H), 1 .40 (s, 9H).
Example 97: 2-(fer/-butyl)-A/-cvclopentyl-4-(4-(piperazin-1 -yl)pyrimidin-2-yl)aniline trihydrochloride
Figure imgf000128_0001
Step 1: 2-(tert-butyl)-N-cyclopentylaniline:
To a solution of 2-(tert-butyl)aniline (1 .567 ml, 10.05 mmol) in 1 ,2-Dichloroethane (15 ml) was added cyclopentanone (1 .78 ml, 20.1 mmol), sodium triacetoxyborohydride (5.33 g, 25.1 mmol) and acetic acid (1 .726 ml, 30.2 mmol). The reaction mixture was heated in a microwave at 140 'Ό for 10 min. After cooling to RT the mixture was redissolved in CH2CI2. The mixture was washed with water and brine. The organic layer was dried over Na2S04, filtered and concentrated in vacuo. The resultant crude was subjected to flash chromatography (Si02, gradient 0 to 30 % EtOAc in cyclohexane) to afford the title compound (1 .89 g, 87 %). LCMS (Method F): RT = 1 .84 min, M+H+ = 218. Step 2: 4-bromo-2-(tert-butyl)-N-cyclopentylaniline:
To a solution of 2-(te/t-butyl)-/V-cyclopentylaniline (3.78 g, 17.39 mmol) in THF (30 ml) was added tetra- /V-butylammonium tribromide (8.39 g, 17.39 mmol) at O 'C. The reaction mixture was then stirred at RT for 10 min. Water was added to the reaction and the mixture was extracted with EtOAc. The organic layer was washed with brine, dried over Na2S04, filtered and concentrated in vacuo to afford the title compound (4.9 g, 95 %) which was used without further purification.
LCMS (Method F): RT = 1 .94 min, M+H+ = 296/298.
Step 3: 2-(tert-butyl)-N-cyclopentyl-4-(4^,5,5-tetramethyl- 1,3,2-dioxab
To a solution of 4-bromo-2-(te/?-butyl)-/V-cyclopentylaniline (0.9 g, 3.04 mmol) in DMF (7 ml) was added 4,4,4',4', 5,5,5', 5'-ocfamethyl-2,2'-bi(1 ,3,2-dioxaborolane) (0.771 g, 3.04 mmol) and KOAc (0.894 g, 9.1 1 mmol). Nitrogen was bubbled through the mixture for 10 min and then PdCI2(dppf)-CH2CI2 (0.248 g, 0.304 mmol) was added. The mixture was heated for 16 h at 80 °C under nitrogen. After cooling to RT the reaction mixture was diluted with EtOAc. The mixture was washed with brine. The organic layer was dried over Na2S04, filtered and concentrated in vacuo. The resultant crude was subjected to flash chromatography (Si02, gradient 0 to 20 % EtOAc in cyclohexane) to afford the title compound (389 mg, 37 %).
LCMS (Method F): RT = 1 .96 min, M+H+ = 344.
Step 4: tert-butyl 4-(4-(3-(tert-butyl)-4-(cyclopentylamino)phenyl)pyrimidin-2- yl)piperazine- 1 -carboxylate:
To a solution of 2-(ie -butyl)-/V-cyclopentyl-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)aniline (390 mg, 1 .136 mmol) in DMF (2 ml) was added a mixture of terf-butyl 4-(2- chloropyrimidin-4-yl)piperazine-1 -carboxylate and terf-butyl 4-(4-chloropyrimidin-2- yl)piperazine-1 -carboxylate (339 mg, 1 .136 mmol) and 2M aq. Na2C03 (1 .136 ml, 2.272 mmol). The reaction mixture was degassed by bubbling nitrogen through the mixture for 10 min and then PdCI2(dppf)-CH2CI2 (93 mg, 0.1 14 mmol) was added. The mixture was heated for 16 h at 80 °C under nitrogen. After cooling to RT the reaction mixture was diluted with EtOAc. The mixture was washed with brine. The organic layer was dried with Na2S04, filtered and concentrated in vacuo. The resultant crude was subjected to flash chromatography (Si02, gradient 0 to 30 % EtOAc in cyclohexane) to afford the title compound (89 mg, 16%).
LCMS (Method F): RT = 2.02 min, M+H+ = 480; H-NMR (500 MHz, CDCI3): 8.26 (1 H, d), 8.03 (1 H, d), 7.82 (1 H, dd), 6.88 (1 H, d), 6.73 (1 H, d), 4.25 (1 H, d), 3.96-3.93 (1 H, m), 3.90-3.88 (4H, m), 3.54-3.52 (4H, m), 2.10-2.04 (2H, m), 1 .77-1 .67 (4H, m), 1 .57-1 .55 (2H, m), 1 .50 (9H, s), 1 .45 (9H, s).
Step 5: 2-(tert-butyl)-N-cyclopentyl-4-(4-(piperazin- 1-yl)pyrimidin-2-yl)ani
trihydrochloride:
To a solution of te/t-butyl 4-(2-(3-(ie -butyl)-4-(cyclopentylamino)phenyl)pyrimidin-4- yl)piperazine-1 -carboxylate (50mg, 0.104 mmol) in CH2CI2 (1 ml) was added HCI (4M in dioxane, 1 ml, 4.00 mmol). The reaction mixture was then stirred at RT for 4h. The solvents were removed and the resulting solid was dried in vacuo to afford the title compound (41 mg, 95%).
LCMS (Method F): RT = 1 .17 min, M+H+ = 380; 1H NMR (500Mhz, MeOH-d4): 8.20 (1 H, d), 8.17 (1 H, d), 8.13 (1 H, dd), 7.52 (1 H, d), 6.91 (1 H, d), 4.36-4.34 (1 H, m), 4.28-4.26 (4H, m), 4.09-4.05 (1 H, m), 3.52-3.50 (4H, m), 2.17-2.10 (2H, m), 1 .82-1 .72 (4H, m), 1 .70-1 .63 (2H, m), 1 .47 (9H, s).
Example 98: 2-(fer/-butyl)-A/-cvclopentyl-4-(2-(piperazin-1 -yl)pyrimidin-4-yl)aniline trihydrochloride
Figure imgf000130_0001
Step 1: tert-butyl 4-(2-(3-(tert-butyl)-4-(cyclopentylamino)phenyl)pyrimidin-4- yl)piperazine- 1 -carboxylate:
To a solution of 2-(ie -butyl)-/V-cyclopentyl-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)aniline (390 mg, 1 .136 mmol) in DMF (2 ml) was added a mixture of terf-butyl 4-(2- chloropyrimidin-4-yl)piperazine-1 -carboxylate and terf-butyl 4-(4-chloropyrimidin-2- yl)piperazine-1 -carboxylate (339 mg, 1 .136 mmol) and 2M aq. Na2C03 (1 .136 ml, 2.272 mmol). Nitrogen was bubbled through the mixture for 10 min and then PdCI2(dppf)- CH2CI2 (93 mg, 0.1 14 mmol) was added. The mixture was heated for 16 h at 80 °C under nitrogen. After cooling to RT the reaction mixture was diluted with EtOAc. The mixture was washed with brine. The organic layer was dried with Na2S04, filtered and concentrated in vacuo. The resultant crude was subjected to flash chromatography (Si02, gradient 0 to 30 % EtOAc in cyclohexane) to afford the title compound (59 mg, 1 1 %).
LCMS (Method F): RT = 1 .62 min, M+H+ = 480; H-NMR (500 MHz, CDCI3): 8.32 (1 H, d), 8.28 (1 H, d), 8.14 (1 H, dd), 6.72 (1 H, d), 6.29 (1 H, d), 4.19 (1 H, d), 3.99-3.93 (1 H, m), 3.74-3.70 (4H, m), 3.58-3.53 (4H, m), 2.09-2.01 (2H, m), 1 .77-1 .64 (4H, m), 1 .59-1 .54 (2H, m), 1 .49 (9H, s), 1 .46 (9H, s).
Step 2: 2-(tert-butyl)-N-cyclopentyl-4-(2-(piperazin- 1-yl)pyrimidi
trihydrochloride:
To a solution of te/t-butyl 4-(2-(3-(ie -butyl)-4-(cyclopentylamino)phenyl)pyrimidin-4- yl)piperazine-1 -carboxylate (50mg, 0.104 mmol) in CH2CI2 (1 ml) was added HCI (4M in dioxane, 1 ml, 4.00 mmol). The reaction mixture was then stirred at RT for 4h. The solvents were removed and the resulting solid was dried in vacuo to afford the title compound (36 mg, 91 %).
LCMS (Method F): RT = 0.94 min, M+H+ = 380; 1H NMR (500Mhz, MeOH-d4): 8.19 (1 H, d), 8.08 (1 H, d), 8.05 (1 H, d), 7.05 (1 H, d), 6.88 (1 H, d), 4.49 (2H, bs), 4.13 (2H, bs), 4.07-4.02 (1 H, m), 3.47 (4H, bs), 2.15-2.10 (2H, m), 1 .83-1 .71 (4H, m), 1 .65-1 .60 (2H, m), 1 .47 (9H, s).
Example 99: 5-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)picolinimidamide dihvdrochloride
Figure imgf000131_0001
Step 1: 5-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)picolinonitrile:
To a solution of 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)picolinonitrile (91 mg, 0.394 mmol) and 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) in dioxane (2 ml) was charged Na2C03 (1 M aq), (1 ml, 1 mmol). Nitrogen was bubbled through the mixture, and Pd(PPh3)4 (19 mg, 0.016 mmol) was added. The mixture was heated to 120 'Ό for 20 min under microwave conditions. After cooling to RT the reaction mixture was partitioned between DCM and water, filtered through a phase separator cartridge and concentrated in vacuo. The remaining residue was purified by flash chromatography (Si02, 5-30%, EtOAc in cyclohexane) to give the title compound (64 mg, 52%) as an off white solid.
LCMS (Method F): RT = 1 .87 min, M+H+ = 373.
Step 2: 5-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)picolini
dihydrochloride:
To a solution of 5-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)picolinonitrile (30 mg, 0.081 mmol) in methanol (1 ml) was charged sodium methoxide (0.870 mg, 0.016 mmol) the mixture was heated under microwave conditions for 10 min at 100 °C.
Ammonium chloride (4.31 mg, 0.081 mmol) was then charged to the reaction and the mixture heated under microwave conditions for a further 12 min at 80 °C. After cooling to RT the mixture was partitioned between EtOAc and water. The layers were separated and the aqueous layer was extracted with EtOAc. The combined organic extracts were washed with brine, dried (MgS04) filtered and concentrated in vacuo. The resultant residue was loaded onto a 2 g SCX cartridge. The cartridge was washed with MeOH, and then the desired compound was eluted with 7N NH3 in MeOH. The product containing fractions were concentrated in vacuo and the hydrochloride salt of the product was prepared by dissolving this material in dioxane (2 ml) and adding to this a solution of HCI in dioxane (4M, 0.1 ml). The solvent was removed in vacuo and the material was dissolved in water (1 ml) and lyophilized to afford the desired product (15 mg, 40 %) as a white solid.
LCMS (Method F): RT = 1 .22 min, M+H+ = 390; 1H NMR (500 MHz, MeOH-d4): 9.79 (1 H, d), 9.01 (1 H, dd), 8.77 (1 H, d), 8.22-8.24 (2H, m), 8.05 (1 H, dd), 7.80 (1 H, d), 7.04 (1 H, d), 4.75 (1 H, m), 1 .40 (9H, s), 1 .34 (6H, d). Example 100: (5-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)pyridin-2- vDmethanamine dihydrochloride
Figure imgf000133_0001
Using the H-Cube and Raney Nickel as catalyst, a solution of 2.0M ammonia in methanol was flushed through the system at 1 ml/min for 5 min at 80 'Ό and 50 bar with a constant flow of hydrogen. The flow was stopped and a solution of 5-(4-(3-(tert-butyl)- 4-isopropoxyphenyl)pyrimidin-2-yl)picolinonitrile (50 mg, 0.134 mmol) in 100 ml of 2.0M ammonia in methanol was then pumped through the system at a flow rate of 1 .0 ml/min. The solvent was removed in vacuo and the residue purified by flash chromatography (Si02, 5-30%, EtOAc in cyclohexane). The hydrochloride salt of the product was prepared by dissolving this material in dioxane (1 ml) and adding to this a solution of HCI in dioxane (4M, 0.1 ml). The solvent was then removed in vacuo and the material was dissolved in water (1 ml) and lyophilized to afford the desired product (13 mg, 21 %) as a green solid.
LCMS (Method F): RT = 1 .19 min, M+H+ = 377; 1H NMR (500 MHz, MeOH-d4): 9.60
(1 H,d), 8.78 (1 H, dd), 8.72 (1 H, d), 8.23 (1 H, d), 8.04 (1 H, dd), 7.77 (1 H, d), 7.53 (1 H, d),
7.03 (1 H, d), 4.77 (1 H, m), 4.30 (2H, s), 1 .39 (9H, s), 1 .34 (6H, d).
Example 101 : 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine-1 - carboximidamide hydrochloride
Figure imgf000133_0002
Step 1: tert-butyl (((tert-butoxycarbonyl)imino)(4-(4-(3-(tert-butyl)-4- isopropoxyphenyl)pyrimidin-2-yl)piperazin- 1-yl)methyl)carbamate:
To 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(piperazin-1 -yl)pyrimidine hydrochloride (188 mg, 0.440 mmol), 1 ,3-bis(te/t-butoxycarbonyl)-2-methyl-2-thiopseudourea (128 mg, 0.440 mmol) and triethylamine (0.245 ml, 1 .759 mmol) in DMF (1 .5 ml) was charged mercury(ll) chloride (1 19 mg, 0.440 mmol). The reaction was then stirred at RT under a nitrogen atmosphere overnight. The reaction was diluted with EtOAc and water and the layers separated. The organic layer was washed three times with water, dried (MgS04), filtered and concentrated in vacuo. The crude material was purified by flash
chromatography (S1O2, 0-20% ethyl acetate and cyclohexane) to afford the title compound (100 mg, 38%) as a colourless gum.
LCMS (Method F): RT = 1 .98 min M+H+ = 597.
Step 2: 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine- 1 - carboximidamide hydrochloride:
To a solution of tert-butyl (((tert-butoxycarbonyl)imino)(4-(4-(3-(tert-butyl)-4- isopropoxyphenyl)pyrimidin-2-yl)piperazin-1 -yl)methyl)carbamate (50 mg, 0.084 mmol) in dioxane (1 ml) was charged HCI (4M in dioxane, 1 ml). The reaction was then stirred at RT overnight. The solvent was removed in vacuo, and the material slurried in Et20, dissolved in water (1 ml) and lyophilized to afford the title compound (20 mg, 60 %) as a white solid.
LCMS (Method F): RT = 1 .19 min, M+H+ = 397; 1H NMR (500 MHz, MeOH-d4): 8.3
(1 H,d), 8.26 (1 H, d), 8.17 (1 H, dd), 7.53 (1 H, d), 7.46 (1 H, s), 7.17 (1 H, d), 4.91 (1 H, m),
4.15 (4H, m), 3.83 (4H, m), 1 .47 (9H, s), 1 .46 (6H, d).
Example 102: 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine-1 - carboximidamide hydrochloride
Figure imgf000134_0001
To a solution of tert-butyl 5,6-dihydro-[1 ,2,4]triazolo[4,3-a]pyrazine-7(8H)-carboxylate (100 mg, 0.446 mmol) in dichloromethane (5 ml) was charged trifluoroacetic acid (1 .5 ml, 19.47 mmol) and the reaction was stirred at RT for 2 h. The reaction was concentrated in vacuo, the resulting oil was dissolved in DMF (1 .5 ml) and 4-(3-(tert-butyl)-4- isopropoxyphenyl)-2-chloropyrimidine (100 mg, 0.328 mmol) and DIPEA (0.229 ml, 1 .312 mmol) were added. The mixture was heated at 120°C under microwave conditions for 80 min. After cooling the reaction mixture was diluted with water and ethyl acetate and the layers were separated. The aqueous layer was extracted once more with ethyl acetate and the organic extracts were combined, washed four times with water/brine
1 :1 , dried (MgS04), filtered and concentrated in vacuo. The residue was purified by flash chromatography (Si02, 0-10%, MeOH in DCM) to afford the title compound (8 mg, 6%) as a colourless gum.
LCMS (Method F): RT = 1 .53 min, M+H+ = 393; 1H NMR (500 MHz, MeOH-d4): 8.50 (1 H,s), 8.41 (1 H, d), 8.16 (1 H, d), 7.98 (1 H, dd), 7.19 (1 H, d), 7.06 (1 H, d), 5.27 (2H, s), 4.83 (1 H, m), 4.42 (2H, t), 4.29 (2H, t), 1 .48 (9H, s), 1 .43 (6H, d).
Example 103: 2-(4-(1 H-imidazol-2-yl)DiDeridin-1 -yl)-4-(3-(tert-butyl)-4- isopropoxyphenvDpyrimidine
Figure imgf000135_0001
To a solution of 4-(1 H-imidazol-2-yl)piperidine (49.6 mg, 0.328 mmol) and 4-(3-(tert- butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (50 mg, 0.164 mmol) in DMF (2 ml) was charged DIPEA (0.086 ml, 0.492 mmol). The reaction was heated at 120 °C under microwave conditions for 40 min. The reaction mixture was diluted with water and ethyl acetate and the layers were separated. The aqueous layer was extracted once more with ethyl acetate and the organic extracts were combined, washed four times with water/brine 1 :1 , dried (MgS04), filtered and concentrated in vacuo. The residue was purified by flash chromatography (Si02, 0-10%, MeOH in DCM) to afford the title compound (50 mg, 73%) as a white solid.
LCMS (Method F): RT = 1 .17 min, M+H+ = 420; 1H NMR (500 MHz, MeOH-d4): 8.29 (1 H, d), 8.13 (1 H, d), 7.94 (1 H, dd), 7.03-7.06 (2H, m), 6.93 (2H, s), 4.95 (2H, m), 4.82 (1 H, m), 3.07-3.14 (3H, m), 2.03-2.08 (2H, m), 1 .78-1 .87 (2H, m), 1 .46 (9H, s), 1 .43 (6H, d).
Example 104: 2-(tert-butyl)-N-isopropyl-4-(2-(piperazin-1 -yl)pyrimidin-4-yl)aniline
Figure imgf000136_0001
Step 1 : 2-(tert-butyl)-N-isopropylaniline:
To a solution of 2-(tert-butyl)aniline (1 .045 ml, 6.70 mmol) in 1 ,2-Dichloroethane (10 ml) was added propan-2-one ( 0.541 ml, 7.37 mmol), followed by sodium
triacetoxyborohydride ( 2.27 g, 10.72 mmol) and acetic acid ( 0.422 ml, 7.37 mmol). This mixture was heated to 60 °C overnight under a nitrogen atmosphere. After cooling to RT the reaction mixture was partitioned between DCM and water. The organic phase was dried (phase separator cartridge) and concentrated in vacuo to afford the desired product (1 .53 g, 100%) as an orange oil.
1 H NMR (500 MHz, CDCI3): 7.26 (1 H, dd), 7.14 (1 H, m), 6.68-6.72 (2H, m), 3.76 (1 H, m), 1 .46 (9H, s), 1 .29 (6H, d).
Step 2: 4-bromo-2-(tert-butyl)-N-isopropylaniline:
To a solution of 2-(te/?-butyl)-/V-isopropylaniline (1 .282 g, 6.70 mmol) in THF (20 ml) was added tetra-n-butylammonium tribromide (3.23 g, 6.70 mmol) at 0 °C. The reaction mixture was stirred at room temperature for 10 min, quenched with water and extracted with EtOAc. The organic layer was washed with brine, dried (MgS04) and concentrated in vacuo to afford a brown solid.
LC-MS (Method F) RT =1 .82min, M+H+=270/272.
Step 3: 2-(tert-butyl)-N-isopropyl-4-(4^,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)aniline: To a solution of 4-bromo-2-(te/t-butyl)-/V-isopropylaniline (1 .69 g, 6.25 mmol) in DMF (12 ml) was added 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1 ,3,2-dioxaborolane) (1 .588 g, 6.25 mmol) and potassium acetate (1 .841 g, 18.76 mmol). The reaction mixture was degassed by bubbling nitrogen through for 10 min, PdCI2(dppf)-CH2CI2 adduct (0.51 1 g, 0.625 mmol) was added. The mixture was heated to 80 °C for 16 h. After cooling the reaction mixture was diluted with water and extracted with EtOAc. The organic layer was washed twice with water, followed by brine, dried (MgS04), filtered and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 8 % EtOAc in cyclohexane) to afford the title compound as an orange gum (1 .00 g, 50 %).
LC-MS (Method F) RT =1 .87min, M+H+=318.
Step 4: 2-(tert-butyl)-4-(2-chloropyrimidin-4-yl)-N-isopropylaniline:
To a solution of 2,4-dichloropyrimidine (0.939 g, 6.30 mmol) and 2-(tert-butyl)-N- isopropyl-4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)aniline (1 g, 3.15 mmol) in dioxane (30 ml) was charged a solution of sodium carbonate (1 .002 g, 9.46 mmol) in water (10 ml). The reaction was then degassed by bubbling nitrogen through for 10 min, Palladium-tetrakis(triphenylphosphine) (0.182 g, 0.158 mmol) was added and the reaction heated at 90 'Ό under a nitrogen atmosphere overnight. The reaction was diluted with water and EtOAc and the layers separated. The aqueous was extracted twice more with ethyl acetate and the organic layers were combined, dried (MgS04) filtered and concentrated in vacuo. The crude material was purified by flash (S1O2, 0-
15% ethylacetate in cyclohexane) to afford the desired product (557 mg, 58%) as an orange solid.
LC-MS (Method F) RT =1 .71 min, M+H+=304.
Step 5: tert-butyl 4-(4-(3-(tert-butyl)-4-(isopropylamino)phenyl)pyrimidin-2-yl)piperazine- 1-carboxylate:
To a solution of tert-butyl piperazine-1 -carboxylate (245 mg, 1 .317 mmol) and 2-(tert- butyl)-4-(2-chloropyrimidin-4-yl)-N-isopropylaniline (200 mg, 0.658 mmol) in DMF (2 ml) was charged DIPEA (0.345 ml, 1 .975 mmol). The mixture was heated to 120 °C for 20 min under microwave conditions. After cooling to RT the reaction mixture was partitioned between DCM and water. The organic phase was dried (phase separator cartridge) and concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 14 % EtOAc in cyclohexane) to afford the title compound as a yellow oil (212 mg, 71 %).
LCMS (Method F): RT = 1 .88 min, M+H+ = 454. Step 6: 2-(tert-butyl)-N-isopropyl-4-(2-(piperazin- 1 -yl)pyrimidin-4-yl)aniline:
To a solution of tert-butyl 4-(4-(3-(tert-butyl)-4-(isopropylamino)phenyl)pyrimidin-2- yl)piperazine-1 -carboxylate (200 mg, 0.441 mmol) in DCM (2 ml) was charged 4M HCI in dioxane (6 ml, 24.00 mmol). The reaction was stirred at RT overnight. The solvent was removed in vacuo and the residue was partitioned between aq. NaHC03 solution and DCM. The organic phase was dried (phase separator cartridge) and concentrated in vacuo. The resultant residue was purified using preparative HPLC (Method E) to afford the title compound (72 mg, 46%) as a colourless gum.
LCMS (Method F): RT = 1 .06 min, M+H+ = 354; 1H NMR (400 MHz, MeOH-d4): 8.22 (1 H, d), 8.10 (1 H, d), 7.86 (1 H, dd), 7.00 (1 H, d), 6.78 (1 H, d), 3.82-3.88 (5H, m), 2.92 (4H, t), 1 .48 (9H, s), 1 .32 (6H, d).
Example 105: (rac)-2-amino-2-(4-(4-(5,5,8,8-tetramethyl-5,6 ,7,8-tetrahvdronaphthalen-2- yl)pyrimidin-2-yl)phenyl)ethanol ditrifluoroacetate
Figure imgf000138_0001
Step 1: (rac)-tert-butyl (1 -(4-chlorophenyl)-2-hydroxyethyl)carbamate:
To a solution of (rac)-2-amino-2-(4-chlorophenyl)ethanol (2 g, 1 1 .65 mmol) (prepared as described WO2009/47563 A1 ) in THF (20 ml) was added Boc anhydride (2.84 ml, 12.24 mmol) followed by triethylamine (2.436 ml, 17.48 mmol) . The reaction mixture was then stirred at RT and maintained at this temperature overnight. The reaction mixture was concentrated in vacuo, diluted with EtOAc and 1 N HCI and extracted. The organic extracts were then washed with brine, dried (Na2S04) and concentrated in vacuo affording the title compound as a sticky solid (2.8 g, 88%). 1H NMR (500 MHz, CDCI3): 7.34 (2H, m), 7.27 (1 H, d), 5.35 (1 H, m), 4.75 (1 H, m), 3.75- 3.90 (2H, m), 1 .44 (9H, s).
Step 2: (rac)-tert-butyl (2-hydroxy-1-(4-(4,4,5,5-tetramethyl- 1,3,2-dioxaborolan-2- yl)phenyl)ethyl)carbamate:
To a solution of (rac)-tert-butyl (1 -(4-chlorophenyl)-2-hydroxyethyl)carbamate (1 .7 g, 6.26 mmol) in THF (25 ml) was added bis(pinacolato)diboron (1 .747 g, 6.88 mmol), 1 ,3-di- isopropylimidazoliumchloride (0.160 g, 0.375 mmol) and potassium acetate (1 .535 g, 15.64 mmol). Nitrogen was bubbled through the reaction mixture for 10 min before the addition of palladium(ll)acetate (0.042 g, 0.188 mmol). The reaction mixture was then heated at 80 °C for 2 h. The reaction mixture was cooled to room temperature, concentrated, diluted with EtOAc and water and extracted with EtOAc. The organic extracts were then washed with brine, dried (Na2S04), filtered and concentrated in vacuo. The resultant residue was purified by flash chromatography (0 to 50% EtOAc in cyclohexane), affording the title compound as a gummy solid (1 .13 g, 50%).
LCMS (Method F): RT = 1 .32 min, (M-Boc) + H+ = 264.
Step 3: (rac)- tert-butyl (2-hydroxy- 1-(4-(4-(5,5,8,8-tetramethyl-5,6, 7,8- tetrahydronaphthalen-2-yl)pyrimidin-2-yl)phenyl)ethyl)carbamate:
To a solution of (rac)-tert-butyl (2-hydroxy-1 -(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl)ethyl)carbamate (145 mg, 0.399 mmol) and 2-chloro-4-(5,5,8,8-tetramethyl- 5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine (100 mg, 0.332 mmol) in dioxane (1 ml) was charged 1 M aq. sodium carbonate (1 ml, 1 .00 mmol). Nitrogen was bubbled through the reaction mixture for 10 min before the addition of
tetrakis(triphenylphosphine)palladium(0) (19.21 mg, 0.017 mmol). The reaction was sealed and heated under microwave conditions (Biotage initiator) at 120 'Ό for 40 min. The reaction was diluted with water and DCM, the layers separated using a phase separator cartridge and the organic layer concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 45% EtOAc in cyclohexane) to afford the title compound as an orange gum (82 mg, 49%).
LC-MS (Method F) RT = 1 .82 min, M+H+ = 502.
Step 4: (rac)-2-amino-2-(4-(4-(5,5,8,8-tetramethyl-5,6, 7,8-tetrahydronaphthalen-2- yl)pyrimidin-2-yl)phenyl)ethanol ditrifluoroacetate: To a solution of (rac)-tert-butyl (2-hydroxy-1 -(4-(4-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyrimidin-2-yl)phenyl)ethyl)carbamate (40 mg, 0.080 mmol) in DCM (2 ml) was charged TFA (1 ml, 12.98 mmol) the reaction was then stirred at RT for 10 min. The reaction was concentrated in vacuo to afford the desired product (41 mg, 100%) as an off-white solid.
LC-MS (Method F) RT =1 .15 min, M+H+=402; 1H NMR (500 MHz, MeOH-d4): 8.86 (1 H, d), 8.63 (2H, d), 8.30 (1 H, d), 8.05 (1 H, dd), 7.87 (1 H, d), 7.64 (2H, d), 7.55 (1 H, d), 4.48 (1 H, m), 3.88-4.01 (2H, m), 1 .80 (4H, s), 1 .42 (6H, s), 1 .37 (6H, s). Example 106: (rac)-2-amino-2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- vDphenvDethanol ditrifluoroacetate
Figure imgf000140_0001
Step 1: (rac)-tert-butyl ( 1-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)phenyl)- 2-hydroxyethyl)carbamate:
To a solution of (rac)-tert-butyl (2-hydroxy-1 -(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl)ethyl)carbamate (143 mg, 0.394 mmol) and 4-(3-(tert-butyl)-4- isopropoxyphenyl)-2-chloropyrimidine (100 mg, 0.328 mmol) in dioxane (1 ml) was charged 1 M sodium carbonate (1 ml, 1 .000 mmol). Nitrogen was bubbled through the reaction mixture for 10 min before the addition of
tetrakis(triphenylphosphine)palladium(0) (18.96 mg, 0.016 mmol). The reaction was sealed and heated under microwave conditions (CEM) at 120 'C for 40 min. The reaction was diluted with water and DCM, the layers were separated using a phase separator cartridge and the organic layer was then concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 45% EtOAc in cyclohexane) to afford the title compound as a colourless gum (40 mg, 24%).
LC-MS (Method F) RT = 1 .80 min, M+H+ = 506. Step 2: (rac)-2-amino-2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)phenyl)ethanol ditrifluoroacetate:
To a solution of (rac)-tert-butyl (1 -(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2- yl)phenyl)-2-hydroxyethyl)carbamate (40 mg, 0.079 mmol) in DCM was charged TFA (1 ml, 12.98 mmol) the reaction was then stirred at RT for 10 min. The reaction was concentrated in vacuo to afford the desired product as a yellow solid (40 mg, 97%).
LC-MS (Method F) RT = 1 .12 min, M+H+ = 406; 1H NMR (500 MHz, MeOH-d4): 8.79 (1 H, d), 8.61 (2H, d), 8.34 (1 H, d), 8.13 (1 H, dd), 7.81 (1 H, d), 7.63 (2H, d), 7.13 (1 H, d), 4.90 (1 H, m) 4.47-4.49 (1 H, m), 3.88-4.01 (2H, m), 1 .51 (9H, s), 1 .45 (6H, d).
Example 107: (rac)-2-amino-2-(4-(4-(3-(tert-butyl)-4-(isopropylamino)phenyl)pyrimidin-2- vDphenvDethanol:
Figure imgf000141_0001
Step 1: (rac)-tert-butyl (1-(4-(4-(3-(tert-butyl)-4-(isopropylamino)phenyl)pyrimidin-2- yl) phenyl) -2-hydroxyethyl) carbamate:
To a solution of 2-(tert-butyl)-4-(2-chloropyrimidin-4-yl)-/V-isopropylaniline (100 mg, 0.329 mmol) and (rac)-tert-butyl (2-hydroxy-1 -(4-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)phenyl)ethyl)carbamate (143 mg, 0.395 mmol) in dioxane (1 ml) was charged 1 M aq. sodium carbonate (1 ml, 1 .000 mmol). Nitrogen was bubbled through the reaction mixture for 10 min before the addition of tetrakis(triphenylphosphine)palladium(0) (19.02 mg, 0.016 mmol). The reaction was sealed and heated under microwave conditions (CEM) at 120 °C for 60 min. The reaction was diluted with water and DCM and the layers separated using a phase separator cartridge, the organic layer was then concentrated in vacuo. The resultant residue was subjected to flash chromatography (Si02, gradient 0 to 45 % EtOAc in cyclohexane) to afford the title compound as an orange gum (65 mg, 39%).
LC-MS (Method F) RT = 1 .72 min, M+H+ = 505. Step 2: (rac)-2-amino-2-(4-(4-(3-(tert-butyl)-4-(isopropylamino)phenyl)pyrimidin-2- yl)phenyl)ethanol:
To a solution of (rac)-tert-butyl (1 -(4-(4-(3-(tert-butyl)-4-(isopropylamino)phenyl)pyrimidin- 2-yl)phenyl)-2-hydroxyethyl)carbamate (65 mg, 0.129 mmol) in DCM (2 ml) was charged TFA (1 ml, 12.98 mmol) the reaction was then stirred at RT for 10 min. The reaction was concentrated in vacuo and the residue basified using aq. NaHC03 and extracted with
DCM. The layers were separated (phase separator cartridge) and the organic layer concentrated in vacuo. The crude material was purified by flash chromatography (KP-NH 50-100% EtOAc in cyclohexane) followed by preparative HPLC (Method H) to afford the desired product (5 mg, 10%) as a colourless gum.
LC-MS (Method F) RT = 1 .10 min, M+H+ = 405; 1H NMR (500 MHz, MeOH-d4): 8.67 (1 H, d), 8.54 (2H, d), 8.24 (1 H, d), 7.98 (1 H, dd), 7.46-7.47 (3H, m), 6.77 (1 H, d), 4.06-4.24 (2H, m) 4.76-4.87 (2H, m), 3.59-3.63 (1 H, m), 1 .51 (9H, s), 1 .32 (6H, d).
Example 108: 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidine-4- carboximidamide:
Figure imgf000142_0001
Step 1: 1-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidine-4-carbonitri A solution of 4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-chloropyrimidine (200 mg, 0.656 mmol), piperidine-4-carbonitrile (0.148 ml, 1 .312 mmol) and DIPEA (0.344 ml, 1 .968 mmol) in DMF (3 ml) was heated under microwave conditions (CEM) at 120 °C for 20 min. The reaction mixture was diluted with water and EtOAc and the layers were separated. The aqueous layer was extracted once more with EtOAc and the organic extracts were combined, washed four times with water/brine 1 :1 , dried (MgS04), filtered and concentrated in vacuo. The resultant residue was subjected to flash chromatography
(Si02, gradient 0 to 10% EtOAc in cyclohexane) to afford the title compound (166 mg, LC-MS RT = 1 .85 min, M+H+ = 379.
Step 2: 1-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidine-4- carboximidamide:
To a suspension of ammonium chloride (72.1 mg, 1 .347 mmol) in toluene (1 ml) at 0 'Ό was charged a solution of trimethylaluminum (0.674 ml, 1 .347 mmol) 2.0 M in toluene over 5 min. The mixture was then stirred at RT for 25 min. A solution of 1 -(4-(3-(tert- butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidine-4-carbonitrile (170 mg, 0.449 mmol) in toluene (1 ml) was then charged to the reaction and this heated at 80 °C for 18 h under an atmosphere of nitrogen. The reaction was then cooled to 0 'Ό and quenched by the careful addition of dry methanol. After stirring for 20 min at 0 °C the white precipitate was filtered off through celite and the filter cake washed with methanol. The filtrate was then concentrated in vacuo to yield 220 mg of a waxy solid. 70 mg of this crude material was loaded onto a 2 g SCX-2 cartridge. The cartridge was washed with MeOH, and then the desired compound was eluted with 4M NH3 in MeOH to afford the title compound (14 mg, 25%) as an off white solid.
LC-MS (Method F) RT = 1 .14 min, M+H+ = 396; 1H NMR (500 MHz, DMSO-d6): 8.34 (1 H, d), 8.03 (1 H, d), 7.92 (1 H, dd), 7.07-7.09 (2H, m), 6.71 (2H, brs), 4.78-4.87 (3H, m), 2.90 (2H, m) 2.35-2.40 (1 H, m), 1 .81 -1 .83 (2H, m), 1 .51 -1 .59 (2H, m), 1 .41 (9H, s), 1 .35-1 .36 (6H, d).

Claims

What we claims is:
1 . A compound according to formula I
Figure imgf000144_0001
Formula I
wherein:
Figure imgf000144_0002
where RrR8 are independently hydrogen, lower alkyl, phenyl or benzyl and where X is O, CRR', or NR' where R and R' are independently hydrogen or lower alkyl and where for structures c and d Ri and R2 may together form an optionally lower alkyl substituted 5 or 6 membered ring and where R3 is optionally OR" where R" is alkyl or aryl;
X1 , X2, X3, X4, X5, and X6 are independently selected from nitrogen atom or carbon atom optionally substituted with halogen, oxo, cyano, amino, aryl, heteroaryl, optionally substituted lower alkoxyl and optionally substituted lower alkyl,
where if one of X1 , X2, X3, X4, X5, and X6 is an oxo-substituted carbon atom at least one of the others is a nitrogen atom; wherein not more than three of X1, X2, X3, X4, X5, and X6are nitrogen atoms; and wherein Q is a heteroatom containing alkyl, substituted alkyl, optionally substituted cycloalkyi, optionally substituted aryl, or optionally substituted heterocyclyl containing side chain bound through a heteroatom or directly through a carbon atom; and pharmaceutically acceptable salts, stereoisomers and tautomers thereof.
2. The compound of claim 1 wherein the compound is not morpholine-2-carboxylic acid [6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydro-naphthalen-2-yl)-pyridin-2-yl]-amide or a compound selected from compounds of formula (II)
Figure imgf000145_0001
in which, in each case independently of one another:
R1, R2, R3, R4, R5, R6, R7, R8, R9, R 0, R11, R 2, R 3, R 4, R 5, R 6, R 7 represent H, D
(deuterium), A, OR18, CN, F, CI and NR 8R18 ;
wherein R and R2, R3 and R4, R5 and R6, R 0 and R11, R 2 and R 3, R 4 and R 5, R 6 and R 7 may also in each case together form =0 (carbonyl oxygen);
wherein R9 and R1, R and R2, R2 and R3, R3 and R4, R4 and R5, R5 and R6, R6 and R7, R7 and R8, R 0 and R11, R and R 2, R 2 and R 3, R 4 and R 5, R 5 and R 6, R 6 and R 7 may also in each case together form a cycloalkyi having 3, 4, 5, 6 or 7 C atoms or Het having 3, 4, 5, 6 or 7 ring atoms,
wherein R 0 and R 9 if = CR 9, R and R 2, R 3 and R 9 if Y2 = CR 9, R 4 and R 9 if Yi = CR19, R 5 and R 6, R 7 and R 9 if Y2 = CR19 may also in each case together form a C=C double bond with the single bond and the C atoms to which they are attached;
R 8, R 8' represent H, Dor A;
R 9, R 9' represent H, D, A, OR18, NR 8R18, F, CI, Br, CN or Het; Mi, M2, M3, M4 represent CR 9 or N;
Yi, Y2 represent CR19 or N;
V represents C(R 9)(R19'), NR 9 or is absent;
W represents [C(R 9)(R 9')]PZ, CO-[C(R 9)(R 9')]pZ, [C(R 9)(R 9')]PN(R 9)-Z, CO-N(R19) [C(R 9)(R 9')]pZ, N(R 9)-CO-[C(R 9)(R 9')]pZ, CO-0-[C(R 9)(R 9')]pZ, C(0)OR18, OR18, H or D;
wherein V, W and Y2 may also in each case together form a cycloalkyl having 3, 4, 5, 6 or 7 C atoms, in which preferably 1 , 2, 3, 4, 5, 6 or 7 H atoms can be replaced with F, CI, Br, CN and/or OH, OR19, OC(0)R19, NR 9C(0)OZ, C(0)OR19, C(0)N(R 9)(R19') or N(R 9)(R19'), or form Het having 3, 4, 5, 6 or 7 ring atoms, wherein Het is preferably a saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms which can be unsubstituted or singly, doubly or triply substituted with Hal, F, CI, Br, CN, A, OR18, W, SR 8, N02, N(R 9)(R19'), NR 8COOZ, OCONHZ, NR 8S02Z, S02N(R 8)Z, S(0)mZ, COZ, CHO, COZ, =S, =NH, =NA, Oxy (-0 ) and/or =0 (carbonyl oxygen);
Z represents Het, Ar or A;
A represents unbranched or branched alkyl having 1 , 2, 3, 4, 5, 6, 7, 8, 9 or 10 C atoms, in which 1 , 2, 3, 4, 5, 6 or 7 H atoms can be replaced with F, CI, Br, CN and/or OH, OR19, OC(0)R19, NR 9C(0)OZ, C(0)OR19, C(0)N(R 9)(R19') or N(R 9)(R19');
and/or in which one or two CH2 groups can be replaced having O, S, SO, S02,
CO, COO, NR18, NR 8CO, CONR 8, a cycloalkyl having 3, 4, 5, 6 or 7 C atoms,
CH=CH groups and/or CH≡CH groups;
or a cycloalkyl having 3, 4, 5, 6 or 7 C atoms, in which preferably 1 , 2, 3, 4, 5, 6 or 7 H atoms can be replaced with F, CI, Br, CN and/or OH, OR19, OC(0)R19, NR 9C(0)OZ, C(0)OR19, C(0)N(R 9)(R19') or N(R 9)(R19');
Ar represents phenyl, naphthyl or biphenyl substituted singly, doubly or triply with Hal, F, CI, Br, CN, A, OR18, W, SR18, N02, N(R 9)(R19'), NR 8COOZ, OCONHZ, NR 8S02Z, S02N(R 8)Z, S(0)mZ, COZ, CHO, COZ;
Het represents, in each case independently of one another, a mononuclear, binuclear or trinuclear saturated, unsaturated or aromatic heterocycle having 1 to 4 N, O and/or S atoms which can be unsubstituted or singly, doubly or triply substituted with Hal, F, CI, Br, CN, A, OR18, W, SR18, N02, N(R 9)(R19'), NR 8COOZ, OCONHZ, NR 8S02Z, S02N(R 8)Z, S(0)mZ, COZ, CHO, COZ, =S, =NH, =NA, Oxy (-0 ) and/or =0 (carbonyl oxygen); m is1 , 2 or 3,
n, o are 0, 1 or 2,
p is 0, 1 , 2, 3 or 4
with the proviso compounds of the formula (I) in which
(a) V is absent, and
(b) is W = C(0)-CH2-Het
are excluded;
The compound of claim 1 wherein
Figure imgf000147_0001
is selected from substructures 'c' and 'd' wherein Ri , R2, R3, and R4 are defined as above.
4 The compound of any preceding claims wherein R 0 together with a nitrogen atom to which it is bound, forms a guanidino group. 5 The compound of any one of claims 1 to 3 wherein R10 is a double bonded
nitrogen atom and any other Rx moiety bound to the same atom is absent.
6. The compound of any one of claims 1 to 3 having a guanidine or amidino group forming part of the side-chain 'Q' moiety
7. The compound of any preceding claim wherein one or more of X1 , X2, X3, X4, X5, and X6 is an oxo-substituted carbon atom, methyl substituted carbon atom, or fluorine substituted carbon atom. 8. The compound of any preceding claim wherein one or more of X1 , X2, X3, X4, X5, and X6 is a carbon atom or nitrogen atom.
9. The compound of any one of claims 1 to 6 wherein X1 , X2, X3, X4, X5, and X6 are carbon atoms or nitrogen atoms.
10 The compound of any preceding claim wherein the ring formed by X1 , X2, X3, X4, X5, and X6 is aromatic or doubly unsaturated.
1 1 . The compound of claim 10 wherein the ring is aromatic.
12 The compound of any one of claims 1 to 6 wherein the groups X1 , X2, X3, X4, X5, and X6 are selected so as to form, together with the atoms to which they are bonded, the following ring structures:
Figure imgf000148_0001
where Q is as herein defined and wherein the resulting rings may be additionally substituted in accordance with the selections for groups X1 , X2, X3, X4, X5, and X6.
13. The compound of claim 12 wherein the compounds have the rings shown with no additional substitution.
14 The compound of any preceding claim wherein at least one of the heteroatoms in Q is a nitrogen atom. The compound of claim 14 wherein Q contains at least two heteroatoms,. The compound of claim 15 wherein Q contains at least two nitrogen atoms. The compound of claim 14 wherein Q is selected from:
Figure imgf000149_0001
where R10- 14 are selected from hydrogen or optionally substituted lower alkyl, or may combine with other R groups in Q to form optionally substituted cycloalkyi or optionally substituted heterocycloalkyl rings.
18. The compound of any one of claims 1 to 14 wherein the moiety Q is selected from the following structures:
Figure imgf000150_0001
5
Figure imgf000150_0002
Figure imgf000150_0003
10
Figure imgf000150_0004
15
Figure imgf000151_0001
or structural isomers thereof, optionally additionally substituted with, for example, hydroxyl, amino, halogen, lower alkyl, lower alkoxy and cyano.
19. The compound according to claim 18 wherein the compounds have the Q groups shown with no additional substitution.
20. The compound of claim 1 being:
1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)phenyl)piperazine
1 -(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-4-yl)piperidin-4-amine
1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)phenyl)piperazine
8-(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-4-yl)-2,8- diazaspiro[4.5]decane
1 -(4-(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-4- yl)phenyl)cyclobutanamine
2'-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ,2,3,6-tetrahydro-4,4'- bipyridine
5-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ',2',3',6'-tetrahydro-3,4'- bipyridine
(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine
(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine
1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4-amine 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) piperazine 6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ',2',3',6'-tetrahydro-2,4'- bipyridine
1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)cyclobutanamine
8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8- diazaspiro[4.5]decane
1 -(4-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4-amine 1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) piperazine 4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ',2',3',6'-tetrahydro-2,4'- bipyridine
1 -(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)cyclobutanamine
(7S,8aS)-2-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)octahydropyrrolo[1 ,2-a]pyrazin-7-ol
4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-2,4'-bipyridine
1 - (4-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperidin-4- amine
2- (piperazin-1 -yl)-4-(5,5,8!8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl) pyrimidine 2-(pyridin-4-yl)-4-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyrimidine 2-(1 -methyl-1 ,2,3, 6-tetrahydropyridin-4-yl)-4-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidine
1 - (4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)phenyl)cyclobutanamine
8-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)-2,8- diazaspiro[4.5]decane
2- (4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazin yl)ethanol
N-(pyridin-4-ylmethyl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimi 2-amine
2-(pyridin-3-yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 1 -(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperidin amine
4-(piperazin-1 -yl)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 1 -(6-(3-isopropoxy-4-isopropylphenyl)pyridin-2-yl)piperazine 1 - (6-(4-isopropoxy-3-isopropylphenyl)pyridin-2-yl)piperazine
2- (piperazin-1 -yl)-6-(5,5,8!8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrazine 1 -(5-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-3-yl)piperazine 1 -(5-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyridin-3-yl)piperidin-4-amine 4-(piperazin-1 -yl)-6-(5,5 !8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 1 -(6-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperidin-4- amine
1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperidin-4- amine
1 -(2-methoxyethyl)-4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazine
3- (4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperazin-1 - yl)propan-1 -ol
N,N-dimethyl-2-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)acetamide
1 - methoxy-3-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)propan-2-ol
N,N-dimethyl-2-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)ethanamine
3- hydroxy-2-(hydroxymethyl)-2-methyl-1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)piperazin-1 -yl)propan-1 -one
4- (piperidin-4-yloxy)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine
2- (piperidin-4-yloxy)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 2-methyl-4-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidine
1 -(6-(4-isobutoxy-3-isopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(3-(tert-butyl)-4-isopropoxyphenyl)pyridin-2-yl)piperazine
1 -(6-(3-(tert-butyl)-4-isobutoxyphenyl)pyridin-2-yl)piperazine; or
1 -(6-(2-(benzyloxy)-1 ,1 ,3,3-tetramethyl-2,3-dihydro-1 H-inden-5-yl)pyridin-2-yl)piperidin-4- amine;
and pharmaceutically acceptable salts, stereoisomers and tautomers thereof.
21 The compound of claim 1 being: 1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)phenyl)piperazine
1 -(2-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyridin-4-yl)piperidin-4-amine
1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)phenyl)piperazine
8-(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-4-yl)-2,8- diazaspiro[4.5]decane
1 -(4-(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-4- yl)phenyl)cyclobutanamine
2'-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ,2,3,6-tetrahydro-4,4'- bipyridine
5- (5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ',2',3',6'-tetrahydro-3,4'- bipyridine
(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine
(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine
1 -(6-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4-amine 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) piperazine
6- (5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ',2',3',6'-tetrahydro-2,4'- bipyridine
1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)cyclobutanamine
8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8- diazaspiro[4.5]decane
1 -(4-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4-amine 1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl) piperazine 4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ',2',3',6'-tetrahydro-2,4'- bipyridine
1 -(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)cyclobutanamine
(7S,8aS)-2-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)octahydropyrrolo[1 ,2-a]pyrazin-7-ol
4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-2,4'-bipyridine
1 -(4-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperidin amine 2-(piperazin-1 -yl)-4-(5,5,8!8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl) pyrimidine 2-(pyridin-4-yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 2-(1 -methyl-1 ,2,3, 6-tetrahydropyridin-4-yl)-4-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidine
1 - (4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)phenyl)cyclobutanamine
8-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)-2,8- diazaspiro[4.5]decane
2- (4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazin-1 - yl)ethanol
N-(pyridin-4-ylmethyl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin- 2-amine
2-(pyridin-3-yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine
1 -(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperidin-4- amine
4-(piperazin-1 -yl)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 1 -(6-(3-isopropoxy-4-isopropylphenyl)pyridin-2-yl)piperazine
1 - (6-(4-isopropoxy-3-isopropylphenyl)pyridin-2-yl)piperazine
2- (piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrazine 1 -(5-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-3-yl)piperazine 1 -(5-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-3-yl)piperidin-4-amine 4-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperidin-4- amine
1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperidin-4- amine
1 -(2-methoxyethyl)-4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazine
3- (4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperazin-1 - yl)propan-1 -ol
N,N-dimethyl-2-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)acetamide
1 -methoxy-3-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)propan-2-ol N,N-dimethyl-2-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)ethanamine
3- hydroxy-2-(hydroxymethyl)-2-methyl-1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)piperazin-1 -yl)propan-1 -one
4- (piperidin-4-yloxy)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 2-(piperidin-4-yloxy)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 2-methyl-4-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidine
1 -(6-(4-isobutoxy-3-isopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(3-(tert-butyl)-4-isopropoxyphenyl)pyridin-2-yl)piperazine
1 -(6-(3-(tert-butyl)-4-isobutoxyphenyl)pyridin-2-yl)piperazine; or
1 - (6-(2-(benzyloxy)-1 ,1 ,3,3-tetramethyl-2,3-dihydro-1 H-inden-5-yl)pyridin-2-yl)piperidin-4- amine;
2- (4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-1 -yl)ethanol cis-N1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)cyclohexane-1 ,4-diamine 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-2-one
(R)-4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(3-methylpiperazin-1 -yl)pyrimidine
7-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[3.5]nonane
1 - (4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-4-methyl-1 ,4-diazepane
(1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)azetidin-3-yl)methanamine
2- (4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-1 -yl)ethanamine 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-1 -yl)-N,N- dimethylethan amine
(rac)-1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)pyrrolidin-3-amine
(4-amino-1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-yl)metha
4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 -yl)pyrimidine
4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(piperazin-1 -yl)pyrimidine
1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-amine
1 -(4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)pyrimidin-2-yl)piperidin-4-amine
1 -(2-(3-isopropoxy-4-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine
1 -(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(4-isopropyl-3-propoxyphenyl)pyridin-2-yl)piperazine
1 -(6-(3-(cyclopentyloxy)-4-isopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(4-isopropyl-3-(prop-1 -en-2-yl)phenyl)pyridin-2-yl)piperazine 1 -(6-(3,4-diisopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(3-isopropyl-4-propoxyphenyl)pyridin-2-yl)piperazine
1 - (6-(4-(cyclopentyloxy)-3-isopropylphenyl)pyridin-2-yl)piperazine
(rac)-1 -(6-(3-isopropyl-4-(( 1 ,1 ,1 -trif luoropropan-2-yl)oxy)phenyl)pyridin-2-yl)piperazine (rac)-1 -(6-(4-(sec-butoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine
4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)-2-(piperazin-1 -yl) pyrimidine
(1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-yl)methanamine
2- (4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-2,8-diazaspiro[4.5]decane 1 -(6-(3-isopropyl-4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazine
1 -(6-(3-(tert-butyl)-4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazine
1 -(2-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine
1 -(6-(4-(cyclopropylmethoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine
1 - (6-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine
2- (4-isopropoxy-3-isopropylphenyl)-4-(piperazin-1 -yl)-1 ,3,5-triazine
(rac)-(1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)pyrrolidin-3-yl)methanamine 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-4-phenylpiperidin-4-amine 4-(piperidin-4-yloxy)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)benzimidamide 1 -(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazin-1 - yl)ethanimine
4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazine-1 - carboximidamide
1 -(1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4- yl)guanidine
1 - (1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperidin-4- yl)guanidine
N'-cyano-4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)piperazine-1 -carboximidamide
4- (4-(3-(tert-butyl)-4-(prop-1 -en-2-yl)phenyl)pyrimidin-2-yl)piperazine-1 -carboximidamide
2- (tert-butyl)-N-cyclopentyl-4-(4-(piperazin-1 -yl)pyrimidin-2-yl)aniline
2-(tert-butyl)-N-cyclopentyl-4-(2-(piperazin-1 -yl)pyrimidin-4-yl)aniline
5- (4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)picolinimidamide
(5-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)pyridin-2-yl)methanamine
4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine-1 -carboximidamide 4- (4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine-1 -carboximidamide 2-(4-(1 H-imidazol-2-yl)piperidin-1 -yl)-4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidine 2-(tert-butyl)-N-isopropyl-4-(2-(piperazin-1 -yl)pyrimidin-4-yl)aniline
(rac)-2-amino-2-(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)phenyl)ethanol
(rac)-2-amino-2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)phenyl)ethanol
(rac)-2-amino-2-(4-(4-(3-(tert-butyl)-4-(isopropylamino)phenyl)pyrimidin-2- yl)phenyl)ethanol
1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidine-4-carboximidamide and pharmaceutically acceptable salts, stereoisomers and tautomers thereof.
22. The compound of claim 1 or 2 being:
1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)phenyl)piperazine
1 -(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-4-yl)piperidin-4-amine 8-(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-4-yl)-2,8- diazaspiro[4.5]decane
1 -(4-(2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-4- yl)phenyl)cyclobutanamine
2'-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ,2,3,6-tetrahydro-4,4'- bipyridine
5- (5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ',2',3',6'-tetrahydro-3,4'- bipyridine
(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine
(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)methanamine
6- (5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ',2',3',6'-tetrahydro-2,4'- bipyridine
1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)cyclobutanamine
8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8- diazaspiro[4.5]decane
1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4-amine 4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-1 ',2',3',6'-tetrahydro-2,4'- bipyridine
1 - (4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)phenyl)cyclobutanamine
(7S,8aS)-2-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)octahydropyrrolo[1 ,2-a]pyrazin-7-ol
4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)-2,4'-bipyridine
2- (pyridin-4-yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 2-(1 -methyl-1 ,2,3, 6-tetrahydropyridin-4-yl)-4-(5,5,8,8-tetramethyl-5, 6,7,8- tetrahydronaphthalen-2-yl)pyrimidine
1 - (4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)phenyl)cyclobutanamine
8-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)-2,8- diazaspiro[4.5]decane
2- (4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazin-1 - yl)ethanol
N-(pyridin-4-ylmethyl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin- 2-amine
2-(pyridin-3-yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine
1 -(6-(3-isopropoxy-4-isopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(4-isopropoxy-3-isopropylphenyl)pyridin-2-yl)piperazine
4-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-4-yl)piperidin-4- amine
1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperidin-4- amine
1 -(2-methoxyethyl)-4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazine
N,N-dimethyl-2-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)acetamide
1 -methoxy-3-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)propan-2-ol
N,N-dimethyl-2-(4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2- yl)piperazin-1 -yl)ethanamine 3- hydroxy-2-(hydroxymethyl)-2-methyl-1 -(4-(6-(5,5,8,8-tetramethyl-5,6,7,8- tetrahydronaphthalen-2-yl)pyridin-2-yl)piperazin-1 -yl)propan-1 -one
4- (piperidin-4-yloxy)-2-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 2-(piperidin-4-yloxy)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 2-methyl-4-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2- yl)pyrimidine
1 -(6-(4-isobutoxy-3-isopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(3-(tert-butyl)-4-isopropoxyphenyl)pyridin-2-yl)piperazine
1 -(6-(3-(tert-butyl)-4-isobutoxyphenyl)pyridin-2-yl)piperazine; or
1 - (6-(2-(benzyloxy)-1 ,1 ,3,3-tetramethyl-2,3-dihydro-1 H-inden-5-yl)pyridin-2-yl)piperidin-4- amine;
2- (4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-1 -yl)ethanol cis-N1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)cyclohexane-1 ,4-diamine 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-2-one
(R)-4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(3-methylpiperazin-1 -yl)pyrimidine
7-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-1 ,7-diazaspiro[3.5]nonane
1 - (4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-4-methyl-1 ,4-diazepane
(1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)azetidin-3-yl)methanamine
2- (4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-1 -yl)ethanamine 2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazin-1 -yl)-N,N- dimethylethan amine
(rac)-1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)pyrrolidin-3-amine
(4-amino-1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-yl)metha
4-(4-isopropoxy-3-isopropylphenyl)-2-(piperazin-1 -yl)pyrimidine
4-(3-(tert-butyl)-4-isopropoxyphenyl)-2-(piperazin-1 -yl)pyrimidine
1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-amine
1 -(4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)pyrimidin-2-yl)piperidin-4-amine
1 -(2-(3-isopropoxy-4-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine
1 -(6-(3-isobutoxy-4-isopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(4-isopropyl-3-propoxyphenyl)pyridin-2-yl)piperazine
1 -(6-(3-(cyclopentyloxy)-4-isopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(4-isopropyl-3-(prop-1 -en-2-yl)phenyl)pyridin-2-yl)piperazine
1 -(6-(3,4-diisopropylphenyl)pyridin-2-yl)piperazine
1 -(6-(3-isopropyl-4-propoxyphenyl)pyridin-2-yl)piperazine 1 - (6-(4-(cyclopentyloxy)-3-isopropylphenyl)pyridin-2-yl)piperazine
(rac)-1 -(6-(3-isopropyl-4-(( 1 ,1 ,1 -trif luoropropan-2-yl)oxy)phenyl)pyridin-2-yl)piperazine (rac)-1 -(6-(4-(sec-butoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine
4-(3-(tert-butyl)-4-(cyclopentyloxy)phenyl)-2-(piperazin-1 -yl) pyrimidine
(1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-yl)methanamine
2- (4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-2,8-diazaspiro[4.5]decane 1 -(6-(3-isopropyl-4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazine
1 -(6-(3-(tert-butyl)-4-(2,2,2-trifluoroethoxy)phenyl)pyridin-2-yl)piperazine
1 -(2-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine
1 -(6-(4-(cyclopropylmethoxy)-3-isopropylphenyl)pyridin-2-yl)piperazine
1 - (6-(4-isopropoxy-3-isopropylphenyl)pyrimidin-4-yl)piperidin-4-amine
2- (4-isopropoxy-3-isopropylphenyl)-4-(piperazin-1 -yl)-1 ,3,5-triazine
(rac)-(1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)pyrrolidin-3-yl)methanamine 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)-4-phenylpiperidin-4-amine 4-(piperidin-4-yloxy)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)benzimidamide 1 -(4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazin-1 - yl)ethanimine
4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazine-1 - carboximidamide
1 -(1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperidin-4- yl)guanidine
1 - (1 -(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperidin-4- yl)guanidine
N'-cyano-4-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2- yl)piperazine-1 -carboximidamide
4- (4-(3-(tert-butyl)-4-(prop-1 -en-2-yl)phenyl)pyrimidin-2-yl)piperazine-1 -carboximidamide
2- (tert-butyl)-N-cyclopentyl-4-(4-(piperazin-1 -yl)pyrimidin-2-yl)aniline
2-(tert-butyl)-N-cyclopentyl-4-(2-(piperazin-1 -yl)pyrimidin-4-yl)aniline
5- (4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)picolinimidamide
(5-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)pyridin-2-yl)methanamine
4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine-1 -carboximidamide 4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperazine-1 -carboximidamide 2-(4-(1 H-imidazol-2-yl)piperidin-1 -yl)-4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidine 2-(tert-butyl)-N-isopropyl-4-(2-(piperazin-1 -yl)pyrimidin-4-yl)aniline
(rac)-2-amino-2-(4-(4-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyrimidin yl)phenyl)ethanol
(rac)-2-amino-2-(4-(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)phenyl)ethanol (rac)-2-amino-2-(4-(4-(3-(tert-butyl)-4-(isopropylamino)phenyl)pyrimidin-2- yl)phenyl)ethanol
1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidine-4-carboximidamide and pharmaceutically acceptable salts, stereoisomers and tautomers thereof. 23. The compound of any preceding claim for use in therapy.
24. A pharmaceutical composition comprising a compound of any one of claims 1 to 22 and one or more pharmaceutically acceptable excipients. 25. A pharmaceutical composition comprising a compound of any one of claims 1 to 22, one or more other therapeutically active agents and one or more pharmacuetically acceptable excipients
26 The compound of any one of claims 1 to 22 having selectivity for Sphkl over Sphk2 for the treatment of a disease or disorder involving or associated with SphK1 activity and S1 P homeostasis or having selectivity for Sphk2 over Sphkl for the treatment of a disease or disorder involving or associated with SphK2 activity and S1 P homeostasis. 27 The compound of claim 26 being selected from the group:
1 -(3-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)phenyl)piperazine
8-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)-2,8- diazaspiro[4.5]decane
1 - (4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperidin-4- amine
2- (piperazin-1 -yl)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl) pyrimidine 8-(4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)-2,8- diazaspiro[4.5]decane
4-(piperazin-1 -yl)-6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine 1 -(6-(5,5,8,8-tetramethyl-5,67,8-tetrahydronap
amine
4-(piperidin-4-yloxy)-2-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyrimidine 1 -(4-(3-(tert-butyl)-4-isopropoxyphenyl)pyrimidin-2-yl)piperidin-4-amine
4-(4-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)benzimidami 4-(4-(5,5,8,8-tetramethyl-5,67,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)piperazine-1 - carboximidamide
4-(4-(3-(tert-butyl)-4-(prop-1 -en-2-yl)phenyl)pyrimidin-2-yl)piperazine-1 -carboximidamide and pharmaceutically acceptable salts, stereoisomers and tautomers thereof.
28. The compound of any one of claims 1 to 22 having an inhibitory activity on Sphkl and Sphk2 for the treatment of a disease or disorder involving or associated with SphK1 and/or Sphk2 activity and S1 P homeostasis. 29. The compound of claim 28 being selected from the group:
1 -(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrazin-2-yl)piperidin-4- amine
3- (4-(6-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyridin-2-yl)piperazin-1 - yl)propan-1 -ol
2-(piperidin-4-yloxy)-4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidine
4- (4-(5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)pyrimidin-2-yl)benzimidamide 4-(4-(3-(tert-butyl)-4-(prop-1 -en-2-yl)phenyl)pyrimidin-2-yl)piperazine-1 -carboximidamide and pharmaceutically acceptable salts, stereoisomers and tautomers thereof. 30. The compound of any one of claims 1 to 22, and 26 to 29 for the treatment and/or prevention of conditions selected from cancers, hyperproliferative, inflammatory, angiogenic, immune and viral infectious diseases.
31 . A method of treating or preventing a condition selected from cancers,
hyperproliferative, inflammatory, angiogenic, immune and viral infectious diseases comprising administering a compound of any one of claims 1 to 22, and 26 to 29 to a patient in need thereof.
PCT/GB2011/052334 2010-11-26 2011-11-25 Pharmaceutical compounds as inhibitors of sphingosine kinase WO2012069852A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB1020161.4A GB201020161D0 (en) 2010-11-26 2010-11-26 Pharmaceutical compounds
GB1020161.4 2010-11-26

Publications (1)

Publication Number Publication Date
WO2012069852A1 true WO2012069852A1 (en) 2012-05-31

Family

ID=43500763

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/052334 WO2012069852A1 (en) 2010-11-26 2011-11-25 Pharmaceutical compounds as inhibitors of sphingosine kinase

Country Status (2)

Country Link
GB (1) GB201020161D0 (en)
WO (1) WO2012069852A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013513628A (en) * 2009-12-14 2013-04-22 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Inhibitors of sphingosine kinase
JP2016540824A (en) * 2013-12-20 2016-12-28 インスティテュート オブ ファーマコロジー アンド トキシコロジー アカデミー オブ ミリタリー メディカル サイエンシズ ピー.エル.エー.チャイナ Novel piperidine carboxamide compounds, their preparation and use
WO2017129769A1 (en) 2016-01-28 2017-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for enhancing the potency of the immune checkpoint inhibitors
WO2019162325A1 (en) 2018-02-21 2019-08-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of sk1 as biomarker for predicting response to immunecheckpoint inhibitors
US10858359B2 (en) 2016-06-07 2020-12-08 Jacobio Pharmaceuticals Co., Ltd. Heterocyclic ring derivatives useful as SHP2 inhibitors
US10988466B2 (en) 2017-03-23 2021-04-27 Jacobio Pharmaceuticals Co., Ltd. Heterocyclic derivatives useful as SHP2 inhibitors
US11046658B2 (en) 2018-07-02 2021-06-29 Incyte Corporation Aminopyrazine derivatives as PI3K-γ inhibitors
US11926616B2 (en) 2018-03-08 2024-03-12 Incyte Corporation Aminopyrazine diol compounds as PI3K-γ inhibitors

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005058876A1 (en) * 2003-12-16 2005-06-30 Gpc Biotech Ag Pyrazine derivatives as effective compounds against infectious diseases
WO2007100610A2 (en) * 2006-02-23 2007-09-07 Merck & Co., Inc. Pyridine, pyrimidine and pyrazine derivatives as cxcr3 receptor modulators
WO2009047563A1 (en) 2007-10-11 2009-04-16 Astrazeneca Ab Pyrrolo [2, 3 -d] pyrimidin derivatives as protein kinase b inhibitors
DE102008029734A1 (en) 2008-06-23 2009-12-24 Merck Patent Gmbh Thiazolyl-piperidine derivatives
WO2011072791A1 (en) * 2009-12-14 2011-06-23 Merck Patent Gmbh Sphingosine kinase inhibitors

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005058876A1 (en) * 2003-12-16 2005-06-30 Gpc Biotech Ag Pyrazine derivatives as effective compounds against infectious diseases
WO2007100610A2 (en) * 2006-02-23 2007-09-07 Merck & Co., Inc. Pyridine, pyrimidine and pyrazine derivatives as cxcr3 receptor modulators
WO2009047563A1 (en) 2007-10-11 2009-04-16 Astrazeneca Ab Pyrrolo [2, 3 -d] pyrimidin derivatives as protein kinase b inhibitors
DE102008029734A1 (en) 2008-06-23 2009-12-24 Merck Patent Gmbh Thiazolyl-piperidine derivatives
WO2009156041A2 (en) 2008-06-23 2009-12-30 Merck Patent Gmbh Thiazolyl piperdine derivatives
WO2011072791A1 (en) * 2009-12-14 2011-06-23 Merck Patent Gmbh Sphingosine kinase inhibitors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HENKE B R ET AL: "2-Amino-4,6-diarylpyridines as novel ligands for the estrogen receptor", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, PERGAMON, ELSEVIER SCIENCE, GB, vol. 11, no. 14, 23 July 2001 (2001-07-23), pages 1939 - 1942, XP002575776, ISSN: 0960-894X, DOI: 10.1016/S0960-894X(01)00321-3 *
ZOOROB H H ET AL: "Reactivity of 2-cinnamoyl-5,6,7,8-tetrahydronaphthalene towards ethyl cyanoacetate, cyanoacetamide and some reactions with the adducts formed", INDIAN JOURNAL OF CHEMISTRY. SECTION B: ORGANIC AND MEDICINAL CHEMISTRY, COUNCIL OF SCIENTIFIC AND INDUSTRIAL RESEARCH (C S I R), IN, vol. 16, no. 1, 1 January 1978 (1978-01-01), pages 53 - 56, XP009143770, ISSN: 0376-4699 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2013513628A (en) * 2009-12-14 2013-04-22 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツング Inhibitors of sphingosine kinase
JP2016540824A (en) * 2013-12-20 2016-12-28 インスティテュート オブ ファーマコロジー アンド トキシコロジー アカデミー オブ ミリタリー メディカル サイエンシズ ピー.エル.エー.チャイナ Novel piperidine carboxamide compounds, their preparation and use
WO2017129769A1 (en) 2016-01-28 2017-08-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for enhancing the potency of the immune checkpoint inhibitors
US10858359B2 (en) 2016-06-07 2020-12-08 Jacobio Pharmaceuticals Co., Ltd. Heterocyclic ring derivatives useful as SHP2 inhibitors
US10988466B2 (en) 2017-03-23 2021-04-27 Jacobio Pharmaceuticals Co., Ltd. Heterocyclic derivatives useful as SHP2 inhibitors
WO2019162325A1 (en) 2018-02-21 2019-08-29 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of sk1 as biomarker for predicting response to immunecheckpoint inhibitors
US11926616B2 (en) 2018-03-08 2024-03-12 Incyte Corporation Aminopyrazine diol compounds as PI3K-γ inhibitors
US11046658B2 (en) 2018-07-02 2021-06-29 Incyte Corporation Aminopyrazine derivatives as PI3K-γ inhibitors

Also Published As

Publication number Publication date
GB201020161D0 (en) 2011-01-12

Similar Documents

Publication Publication Date Title
EP3152210B1 (en) Tank-binding kinase inhibitor compounds
US9980952B2 (en) Modulators of methyl modifying enzymes, compositions and uses thereof
WO2012069852A1 (en) Pharmaceutical compounds as inhibitors of sphingosine kinase
KR102327917B1 (en) NOVEL 4-AMINOPYRAZOLO[3,4-d]PYRIMIDINYLAZABICYCLO DERIVATIVES AND PHARMACEUTICAL COMPOSITION COMPRISING THE SAME
US10487075B2 (en) Substituted mono- and polyazanaphthalene derivatives and their use
EP3647311A1 (en) Rho-associated protein kinase inhibitor, pharmaceutical composition comprising same, and preparation method and use thereof
DK2953952T3 (en) MACROCYCLIC PYRIDAZININE DERIVATIVES
KR102420322B1 (en) Indolizine derivatives as phosphoinositide 3-kinases inhibitors
WO2016147659A1 (en) Bicyclic imidazolo derivative
EP2940017A2 (en) Processes for making compounds useful as inhibitors of atr kinase
AU2006297948A1 (en) Novel imidazo [4,5 -b] pyridine derivatives as inhibitors of glycogen synthase kinase 3 for use in the treatment of dementia and neurodegenerative disorders
KR20160100407A (en) Novel inhibitors of glutaminase
EP3784671B1 (en) Dual atm and dna-pk inhibitors for use in anti-tumor therapy
KR20120007540A (en) Inhibitors of pi3 kinase and/or mtor
US10239873B2 (en) 7-azaindole or 4,7-diazaindole derivatives as IKKϵ epsilon and TBK1 inhibitor and pharmaceutical composition comprising same
AU2004313928A1 (en) Heterocyclic protein kinase inhibitors and uses thereof
KR20170044204A (en) Therapeutic compounds as inhibitors of the orexin-1 receptor
US10752609B2 (en) GSK-3 inhibitors
CN109641909B (en) Mechanism targets for rapamycin signaling pathway inhibitors and therapeutic applications thereof
JP2023513794A (en) Inhibitors of ULK1/2 and methods of use thereof
EP4182322B1 (en) Pyrido oxazine derivatives as alk5 inhibitors
EP4206196A1 (en) Pyrimidine substituted derivatives as tyk2 inhibitors
CN117715898A (en) Six-membered aryl or heteroaryl amide compound, composition and application thereof
US20230090406A1 (en) 6-membered heteroaryl-containing aminopyridine compounds as egfr inhibitors
KR20220073680A (en) Heterocyclic compound as diacylglycerol kinases inhibitor and use thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11794218

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11794218

Country of ref document: EP

Kind code of ref document: A1