WO2014033630A1 - Novel aminothiazole carboxamides as kinase inhibitors - Google Patents

Novel aminothiazole carboxamides as kinase inhibitors Download PDF

Info

Publication number
WO2014033630A1
WO2014033630A1 PCT/IB2013/058033 IB2013058033W WO2014033630A1 WO 2014033630 A1 WO2014033630 A1 WO 2014033630A1 IB 2013058033 W IB2013058033 W IB 2013058033W WO 2014033630 A1 WO2014033630 A1 WO 2014033630A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
equiv
tert
compounds
difluoro
Prior art date
Application number
PCT/IB2013/058033
Other languages
French (fr)
Inventor
Matthew Burger
Yu Ding
Wooseok Han
Gisele Nishiguchi
Alice Rico
Robert Lowell Simmons
Huw Tanner
Lifeng Wan
Original Assignee
Novartis Ag
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 Novartis Ag filed Critical Novartis Ag
Publication of WO2014033630A1 publication Critical patent/WO2014033630A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/427Thiazoles not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • the present invention relates to new compounds and compositions of the new compounds together with pharmaceutically acceptable carriers, and uses of the new compounds, either alone or in combination with at least one additional therapeutic agent, in the prophylaxis or treatment of cancer and other cellular proliferation disorders.
  • PIM-Kinase Provirus Integration of Moloney Kinase (PIM-Kinase) was identified as one of the frequent proto-oncogenes capable of being transcriptionally activated by this retrovirus integration event (Cuypers HT et al, "Murine leukemia virus-induced T-cell lymphomagenesis: integration of pro viruses in a distinct chromosomal region," Cell 37(1): 141-50 (1984); Selten G, et al, "Proviral activation of the putative oncogene Pim-1 in MuLV induced T-cell lymphomas” EMBO J 4(7): 1793-8 (1985)), thus establishing a correlation between over-expression of this kinase and its oncogenic potential.
  • Piml being the proto-oncogene originally identified by retrovirus integration.
  • transgenic mice over- expressing Piml or Pim2 show increased incidence of T-cell lymphomas (Breuer M et al., "Very high frequency of lymphoma induction by a chemical carcinogen in pim-1 transgenic mice” Nature 340(6228):61-3 (1989)), while over-expression in conjunction with c-myc is associated with incidence of B-cell lymphomas (Verbeek S et al., "Mice bearing the E mu-myc and E mu-pim-1 transgenes develop pre-B-cell leukemia prenatally" Mol Cell Biol 11(2): 1176-9 (1991)).
  • Piml, 2 & 3 are Serine/Threonine kinases that normally function in survival and proliferation of hematopoietic cells in response to growth factors and cytokines.
  • Substrates for Pim kinases include regulators of apoptosis such as the Bcl-2 family member BAD. The effects of Pim(s) in these regulators are consistent with a role in protection from apoptosis and promotion of cell proliferation and growth.
  • Bcl-2 family member BAD the Bcl-2 family member BAD.
  • the effects of Pim(s) in these regulators are consistent with a role in protection from apoptosis and promotion of cell proliferation and growth.
  • over- expression of Pim(s) in cancer is thought to play a role in promoting survival and proliferation of cancer cells and, therefore, their inhibitions should be an effective way of treating cancers in which they are over-expressed.
  • Pim kinase inhibitors show activity in animal models of inflammation and autoimmune diseases. See JE Robinson “Targeting the Pim Kinase Pathway for Treatment of Autoimmune and Inflammatory Diseases," for the Second Annual Conference on Anti-Inflammatories: Small Molecule Approaches,” San Diego, CA (Conf. April 2011; Abstract published earlier on-line).
  • the present invention addresses such needs.
  • the present invention provides novel compounds that inhibit activity of one or more Pirns, preferably two or more Pirns, more preferably Piml, Pim2 and Pim3, at nanomolar levels (e.g., IC-50 under 50 nM) and exhibit distinctive characteristics that may provide improved therapeutic effects and pharmacokinetic properties, such as reduced drug-drug interactions associated with inhibition of cytochrome oxidases, relative to compounds previously disclosed.
  • Compounds of the invention contain novel substitution combinations on one or more rings that provide these distinctive properties and are suitable for treating Pim-related conditions such as those described herein.
  • the invention provides compounds of Formula (I) that inhibit one or more Pirn kinases:
  • W is H or NH 2 ;
  • Z is CH or N
  • n 1 or 2;
  • ft 1 is H, OH or OMe
  • R 1 is H
  • R 2 is also H
  • R 3 is H or Ci-C 4 alkyl
  • R 4 is selected from R*, -OR*, -OCH 2 CH 2 OR*, -CH 2 OR*, and a 4-6 membered cyclic ether optionally substituted with OH, OMe, or F, wherein each R* is independently C 1-C4 alkyl,
  • R 4 is not -OMe when Z is N;
  • PIM inhibitor is used herein to refer to a compound that exhibits an IC 50 with respect to PIM Kinase activity of no more than about 100 ⁇ and more typically not more than about 5 ⁇ , as measured in the PIM depletion assays described herein below for at least one of Piml, Pim2 and Pim3.
  • Preferred compounds have on IC 50 below about 1 micromolar on at least one Pirn, and generally have an IC 50 below 100 nM on each of Piml, Pim2 and Pim3.
  • alkyl refers to hydrocarbon groups that do not contain heteroatoms, i.e., they consist of carbon atoms and hydrogen atoms.
  • the phrase includes straight chain alkyl groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like.
  • the phrase also includes branched chain isomers of straight chain alkyl groups, including but not limited to, the following which are provided by way of example: -
  • alkyl' includes primary alkyl groups, secondary alkyl groups, and tertiary alkyl groups. Alkyl groups are described herein according to the number of carbon atoms they contain, e.g., an alkyl group containing up to six carbon atoms is described as a CI -6 or C 1-6 , or C1-C6 alkyl.
  • Typical alkyl groups include straight and branched chain alkyl groups having 1 to 12 carbon atoms, preferably 1-6 carbon atoms.
  • the term 'lower alkyl' or “loweralkyl” and similar terms refer to alkyl groups containing up to 6 carbon atoms.
  • alkenyl refers to alkyl groups as defined above, wherein there is at least one carbon-carbon double bond, i.e., wherein two adjacent carbon atoms are attached by a double bond.
  • alkynyl refers to alkyl groups wherein two adjacent carbon atoms are attached by a triple bond.
  • Typical alkenyl and alkynyl groups contain 2-12 carbon atoms, preferably 2-6 carbon atoms.
  • Lower alkenyl or lower alkynyl refers to groups having up to 6 carbon atoms.
  • An alkenyl or alkynyl group may contain more than one unsaturated bond, and may include both double and triple bonds, but of course their bonding is consistent with well-known valence limitations.
  • alkoxy refers to -OR, wherein R is alkyl.
  • halogen refers to chloro, bromo, fluoro and iodo groups. Typical halo substituents are F and/or CI.
  • Haloalkyl refers to an alkyl radical substituted with one or more halogen atoms, typically 1-3 halogen atoms. The term “haloalkyl” thus includes monohalo alkyl, dihalo alkyl, trihalo alkyl, perhaloalkyl, and the like.
  • Amino refers herein to the group -NH 2 .
  • alkylamino refers herein to the group -NRR where R and R are each independently selected from hydrogen or a lower alkyl, provided -NRR' is not -NH 2 .
  • arylamino refers herein to the group -NRR where R is aryl and R' is hydrogen, a lower alkyl, or an aryl.
  • aralkylamino refers herein to the group -NRR where R is a lower aralkyl and R is hydrogen, a loweralkyl, an aryl, or a loweraralkyl.
  • alkoxyalkyl refers to the group -alki-0-alk 2 where alki is an alkyl linking group, and alk 2 is alkyl, e.g., a group such as -0-(CH 2 ) 2 -0-CH 3 .
  • loweralkoxyalkyl refers to an alkoxyalkyl where alki is loweralkyl and alk 2 is loweralkyl.
  • aryloxyalkyl refers to the group -alkyl-O-aryl, where -alkyl- is a Ci_i2 straight or branched chain alkyl linking group, preferably C 1-6 .
  • aralkoxyalkyl refers to the group -alkyl-O-aralkyl, where aralkyl is preferably a loweraralkyl.
  • aminocarbonyl refers herein to the group -C(0)-NH 2 .
  • substituted aminocarbonyl refers herein to the group -C(0)-NRR' where R is loweralkyl and R' is hydrogen or a loweralkyl. In some embodiments, R and R, together with the N atom attached to them may be taken together to form a "heterocycloalkylcarbonyl” group.
  • arylaminocarbonyl refers herein to the group -C(0)-NRR where R is an aryl and R is hydrogen, loweralkyl or aryl.
  • aralkylaminocarbonyl refers herein to the group - C(0)-NRR' where R is loweraralkyl and R is hydrogen, loweralkyl, aryl, or loweraralkyl.
  • aminosulfonyl refers herein to the group -S(0) 2 -NH 2 .
  • Substituted aminosulfonyl refers herein to the group -S(0)2-NRR' where R is loweralkyl and R' is hydrogen or a loweralkyl.
  • aralkylaminosulfonylaryl refers herein to the group -aryl-S(0) 2 -NH-aralkyl, where the aralkyl is loweraralkyl.
  • Carbonyl refers to the divalent group -C(O)-.
  • Cycloalkyl refers to a mono- di- or poly-cyclic, carbocyclic alkyl substituent in which all ring atoms are carbon. Typical cycloalkyl groups have from 3 to 8 backbone (i.e., ring) atoms.
  • polycyclic refers herein to fused and non-fused alkyl cyclic structures, including spirocyclic ring systems.
  • partially unsaturated cycloalkyl all refer to a cycloalkyl group wherein there is at least one unsaturated carbon-carbon bond in the ring, i.e., wherein two adjacent ring atoms are connected by a double bond or a triple bond.
  • Such rings typically contain 1 or 2 double bonds for 5-6 membered rings, and 1-2 double bonds or one triple bond for 7-8 membered rings.
  • Illustrative examples include cyclohexenyl, cyclooctynyl, cyclopropenyl, cyclobutenyl, cyclohexadienyl, and the like.
  • heterocycloalkyl refers herein to cycloalkyl substituents that have from 1 to 5, and more typically from 1 to 3 heteroatoms as ring members in place of carbon atoms.
  • heterocycloalkyl or “heterocyclyl” groups contain one or two heteroatoms as ring members, typically only one heteroatom for 3-5 membered rings and 1-2 heteroatoms for 6-8 membered rings.
  • Suitable heteroatoms employed in heterocyclic groups of the present invention are nitrogen, oxygen, and sulfur.
  • heterocycloalkyl moieties include, for example, pyrrolidinyl, tetrahydrofuranyl, oxirane, oxetane, oxepane, thiirane, thietane, azetidine, morpholino, piperazinyl, piperidinyl and the like.
  • substituted heterocycle refers to any 3- or 4-membered ring containing a heteroatom selected from nitrogen, oxygen, and sulfur or a 5- or 6-membered ring containing from one to three heteroatoms, preferably 1-2 heteroatoms, selected from the group consisting of nitrogen, oxygen, or sulfur; wherein the 5 -membered ring has 0-2 double bonds and the 6-membered ring has 0-3 double bonds; wherein the nitrogen and sulfur atom maybe optionally oxidized; wherein the nitrogen and sulfur heteroatoms may be optionally quaternized; and including any bicyclic group in which any of the above heterocyclic rings is fused to a benzene ring or another 5- or 6-membered heterocyclic ring or heteroaryl as described herein.
  • Preferred heterocycles include, for example: diazapinyl, pyrrolinyl, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, N-methyl piperazinyl, azetidinyl, N-methylazetidinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, and oxiranyl.
  • the heterocyclic groups may be attached at various positions as will be apparent to those having skill in the organic and medicinal chemistry arts in conjunction with the disclosure herein.
  • substituted heterocyclic groups will have up to four substituent groups.
  • cyclic ether refers to a 3-7 membered ring, unless otherwise specified, containing one oxygen atom (O) as a ring member.
  • the cyclic ether is "optionally substituted” it can be substituted at any carbon atom with a group suitable as a substituent for a heterocyclic group, typically up to three substituents selected from lower alkyl, lower alkoxy, halo, hydroxy, amino, -C(0)-lower alkyl, and - C(0)-lower alkoxy.
  • halo, hydroxy and lower alkoxy are not attached to the carbon atoms of the ring that are bonded directly to the oxygen atom in the cyclic ether ring.
  • oxirane e.g., 3-oxetane
  • tetrahydrofuran including 2-tetrahydrofuranyl and 3-tetrahydrofuranyl
  • tetrahydropyran e.g., 4-tetrahydropyranyl
  • oxepane e.g., oxirane, oxetane (e.g., 3-oxetane), tetrahydrofuran (including 2-tetrahydrofuranyl and 3-tetrahydrofuranyl), tetrahydropyran (e.g., 4-tetrahydropyranyl), and oxepane.
  • Aryl refers to monocyclic and polycyclic aromatic groups having from 5 to 14 backbone carbon or hetero atoms, and includes both carbocyclic aryl groups and heteroaromatic aryl groups.
  • Carbocyclic aryl groups are aryl groups in which all ring atoms in the aromatic ring are carbon, typically including phenyl and naphthyl.
  • Exemplary aryl moieties employed as substituents in compounds of the present invention include phenyl, pyridyl, pyrimidinyl, thiazolyl, indolyl, imidazolyl, oxadiazolyl, tetrazolyl, pyrazinyl, triazolyl, thiophenyl, furanyl, quinolinyl, purinyl, naphthyl, benzothiazolyl, benzopyridyl, and benzimidazolyl, and the like.
  • polycyclic aryl refers herein to fused and non-fused cyclic structures in which at least one cyclic structure is aromatic, such as, for example, benzodioxozolo (which has a heterocyclic structure fused to a phenyl group, naphthyl, and the like.
  • aryl is used, the group is preferably a carbocyclic group; the term “heteroaryl” is used for aryl groups when ones containing one or more heteroatoms are preferred.
  • heteroaryl refers herein to aryl groups having from 1 to 4 heteroatoms as ring atoms in an aromatic ring with the remainder of the ring atoms being carbon atoms, in a 5-14 atom aromatic ring system that can be monocyclic or polycyclic.
  • Monocyclic heteroaryl rings are typically 5-6 atoms in size.
  • heteroaryl moieties employed as substituents in compounds of the present invention include pyridyl, pyrimidinyl, thiazolyl, indolyl, imidazolyl, oxadiazolyl, tetrazolyl, pyrazinyl, triazolyl, thiophenyl, furanyl, quinolinyl, purinyl, benzothiazolyl, benzopyridyl, and benzimidazolyl, and the like.
  • Alkyl or “arylalkyl” refers to an aryl group connected to a structure through an alkylene linking group, e.g., a structure such as -(CH 2 )i_4-Ar, where Ar represents an aryl group.
  • “Lower aralkyl” or similar terms indicate that the alkyl linking group has up to 6 carbon atoms.
  • Optionally substituted or “substituted” refers to the replacement of one or more hydrogen atoms with a non-hydrogen group.
  • Alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl groups described herein may be substituted or unsubstituted.
  • Suitable substitution groups include, for example, hydroxy, nitro, amino, imino, cyano, halo, thio, sulfonyl, thioamido, amidino, imidino, oxo, oxamidino, methoxamidino, imidino, guanidino, sulfonamido, carboxyl, formyl, loweralkyl, haloloweralkyl, loweralkylamino, haloloweralkylamino, lower alkoxy, lower haloalkoxy, lower alkoxyalkyl, alkylcarbonyl, aminocarbonyl, arylcarbonyl, aralkylcarbonyl, heteroarylcarbonyl, heteroaralkylcarbonyl, alkylthio, aminoalkyl, cyanoalkyl, aryl and the like, provided that oxo, imidino or other divalent substitution groups are not placed on aryl or heteroaryl rings due
  • optional substituents for alkyl, alkenyl, alkynyl, cycloalkyl, and heterocycloalkyl groups are 1-3 groups selected from halo, hydroxy, amino, cyano, lower alkoxy, lower alkylsulfonyl, oxy, carboxy, and lower alkoxy carbonyl.
  • optional substituents for aryl and heteroaryl groups are 1-3 groups selected from halo, hydroxy, amino, cyano, lower alkyl, lower alkoxy, lower alkylsulfonyl, carboxy, and lower alkoxy carbonyl.
  • the substitution group can itself be substituted where valence permits, i.e., where the substitution group contains at least one CH, NH or OH having a hydrogen atom that can be replaced.
  • the group substituted onto the substitution group can be carboxyl, halo (on carbon only); nitro, amino, cyano, hydroxy, loweralkyl, loweralkoxy, C(0)R, - OC(0)R, -OC(0)OR, -NRCOR, -CONR 2 , -NRCOOR, -C(S)NR 2 , -NRC(S)R, - OC(0)NR 2 , , -SR, -SO 3 H, -S0 2 R or C3-8 cycloalkyl or 3-8 membered heterocycloalkyl, where each R is independently selected from hydrogen, lower haloalkyl, lower alkoxyalkyl, and loweralkyl, and where two R on the same atom or on directly connected atoms can be linked together to form a 5-6
  • a substituted substituent when a substituted substituent includes a straight chain group, the substitution can occur either within the chain (e.g., 2-hydroxypropyl, 2-aminobutyl, and the like) or at the chain terminus (e.g., 2-hydroxyethyl, 3-cyanopropyl, and the like).
  • Substituted substituents can be straight chain, branched or cyclic arrangements of covalently bonded carbon or heteroatoms.
  • impermissible substitution patterns e.g., methyl substituted with five fluoro groups or a halogen atom substituted with another halogen atom. Such impermissible substitution patterns are well known to the skilled artisan.
  • “Syn” as used herein has its ordinary meaning, and is used in connection with Formula I to indicate that the specified groups are attached to sp 3 hybridized (tetrahedral) carbon centers and extend out from one face of the cyclohexyl or piperidinyl ring, i.e., those groups all project toward the 'alpha' face of the ring, or they all project toward the 'beta' face of the ring.
  • This is thus used as a convenient way to define the relative orientations of two or more groups on a ring, without limiting the compounds to a specific absolute chiral configuration. This reflects the fact that the compounds of the invention have such groups in a specific relative orientation, but are not limited to either enantiomer of that specific relative orientation.
  • such compounds may be racemic, but also include each of the two enantiomers having the specified relative stereochemistry.
  • the compounds of the invention are optically active form as further described herein, and in preferred embodiments of the invention, the compounds are obtained and used in optically active form.
  • the enantiomer having greater potency as an inhibitor of at least two of Piml, Pim2 and Pim3 is selected.
  • the compounds of the invention may be subject to tautomerization and may therefore exist in various tautomeric forms wherein a proton of one atom of a molecule shifts to another atom and the chemical bonds between the atoms of the molecules are consequently rearranged.
  • tautomer refers to the compounds produced by the proton shift, and it should be understood that all tautomeric forms, insofar as they may exist, are included within the invention.
  • the compounds of the invention comprise one or more asymmetrically substituted carbon atoms.
  • asymmetrically substituted carbon atoms can result in the compounds of the invention existing in enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, such as in (R)- or (S)- forms.
  • the compounds of the invention are sometimes depicted herein as single enantiomers, and are intended to encompass the specific configuration depicted and the enantiomer of that specific configuration (the mirror image isomer of the depicted configuration), unless otherwise specified— e.g., where a structure is labeled 'chiral', it represents the specified absolute stereochemistry as a single substantially pure (i.e., at least about 95% pure) enantiomer.
  • the depicted structures herein describe the relative stereochemistry of the compounds where two or more chiral centers, but the invention is not limited to the depicted enantiomer's absolute stereochemistry unless otherwise stated.
  • the invention includes both enantiomers, each of which will exhibit Pim inhibition, even though one enantiomer will be more potent than the other.
  • compounds of the invention have been synthesized in racemic form and separated into individual isomers by chiral chromatography or similar conventional methods, and the analytical data about the two enantiomers do not provide definitive information about absolute stereochemical configuration.
  • the absolute stereochemistry of the most active enantiomer has been identified based on correlation with similar compounds of known absolute stereochemistry, rather than by a definitive physical method such as X- ray crystallography.
  • the preferred enantiomer of a compound described herein is the specific isomer depicted or its opposite enantiomer, whichever has the lower IC-50 for Pim kinase inhibition using the assay methods described herein, i.e., the enantiomer that is more potent as a Pim inhibitor for at least two of Pim 1, Pim2, and Pim3.
  • the term "pharmaceutically acceptable salts” refers to the nontoxic acid or base addition salts of the compounds of Formulas I, II, etc., wherein the compound acquires a positive or negative charge as a result of adding or removing a proton; the salt then includes a counterion of opposite charge from the compound itself, and the counterion is preferably one suitable for pharmaceutical administration under the conditions where the compound would be used.
  • These salts can be prepared in situ during the final isolation and purification of the compounds of Formula I or II, or by separately reacting the base or acid functions with a suitable organic or inorganic acid or base, respectively.
  • Representative salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylproionate, picrate, pivalate, propionate, succinate, sulfate,
  • a basic nitrogen-containing group in compounds of the invention can be quaternized with such agents as loweralkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby obtained.
  • These quaternized ammonium salts when paired with a pharmaceutically acceptable anion can also serve as pharmaceutically acceptable salts.
  • acids which may be employed to form pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, sulfuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, methanesulfonic acid, succinic acid and citric acid.
  • Basic addition salts can be prepared in situ during the final isolation and purification of the compounds of formula (I), or separately by reacting carboxylic acid moieties with a suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia, or an organic primary, secondary or tertiary amine.
  • Counterions for pharmaceutically acceptable salts include, but are not limited to, cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, aluminum salts and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.
  • Other representative organic amines useful for the formation of base addition salts include diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like.
  • ester refers to esters, which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof.
  • Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms.
  • examples of particular pharmaceutically acceptable esters include formates, acetates, propionates, maleates, lactates, hydroxyacetates, butyrates, acrylates and ethylsuccinates.
  • prodrugs refers to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention.
  • prodrug refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, PRO-DRUGS AS NOVEL DELIVERY SYSTEMS, Vol. 14 of the A.C.S. Symposium Series, and in Edward B. Roche, ed., BIOREVERSIBLE CARRIERS IN DRUG DESIGN, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference.
  • any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds, lsotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen,
  • the invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3 H and 14 C, or those into which non-radioactive isotopes, such as 2 H and 13 C are present.
  • Such isotopically labeled compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single- photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single- photon emission computed tomography
  • an 18F or labeled compound may be particularly desirable for PET or SPECT studies.
  • Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90%) deuterium incorporation), at least 6333.3 (95%> deuterium incorporation), at least 6466.7 (97%) deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5%> deuterium incorporation).
  • solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D 2 0, d 6 - acetone, d 6 -DMSO.
  • Compounds of the invention i.e. compounds of formula (I) that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of forming co-crystals with suitable co-crystal formers.
  • These co-crystals may be prepared from compounds of formula (I) by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co-melting, or contacting in solution compounds of formula (I) with the co-crystal former under crystallization conditions and isolating co-crystals thereby formed.
  • Suitable co-crystal formers include those described in WO 2004/078163.
  • the invention further provides co-crystals comprising a compound of formula (I).
  • W is H or NH 2 ;
  • Z is CH or N
  • n 1 or 2;
  • R 1 is H, OH or OMe
  • R 2 is H or Me
  • R 1 is H
  • R 2 is also H
  • R 3 is H or Ci-C 4 alkyl
  • R 4 is selected from R*, -OR*, -OCH 2 CH 2 OR*, -CH 2 OR*, and a 4-6 membered cyclic ether optionally substituted with OH, OMe, or F, wherein each R* is independently C 1 -C4 alkyl,
  • R 4 is not -OMe when Z is N; or a pharmaceutically acceptable salt thereof.
  • W is
  • R T is H, OH, OMe, or F. In certain embodiments, it is selected from H, OH and F.
  • R is H, OH, OMe, or F. In certain embodiments, it is selected from H, OH and F.
  • a pharmaceutical composition comprising a compound of any of the preceding embodiments and at least one pharmaceutically acceptable excipient.
  • composition of embodiment 23, wherein the additional therapeutic agent is selected from irinotecan, topotecan, gemcitabine, 5-fluorouracil, cytarabine, daunorubicin, PI3 Kinase inhibitors, mTOR inhibitors, DNA synthesis inhibitors, leucovorin, carboplatin, cisplatin, taxanes, tezacitabine, cyclophosphamide, vinca alkaloids, imatinib, anthracyclines, rituximab, and trastuzumab.
  • the additional therapeutic agent is selected from irinotecan, topotecan, gemcitabine, 5-fluorouracil, cytarabine, daunorubicin, PI3 Kinase inhibitors, mTOR inhibitors, DNA synthesis inhibitors, leucovorin, carboplatin, cisplatin, taxanes, tezacitabine, cyclophosphamide, vinca alkaloids
  • a method to treat a condition caused or exacerbated by excessive Pirn kinase activity comprises administering to a subject in need thereof an effective amount of a compound of any of embodiments 1-21.
  • the subject has been diagnosed with a condition caused by Pirn kinase.
  • the medicament may be to treat a cancer, such as a cancer selected from carcinoma of the lungs, pancreas, thyroid, ovaries, bladder, breast, prostate or colon, melanoma, myeloid leukemia, multiple myeloma, erythro leukemia, villous colon adenoma, and osteosarcoma. 31.
  • a cancer such as a cancer selected from carcinoma of the lungs, pancreas, thyroid, ovaries, bladder, breast, prostate or colon, melanoma, myeloid leukemia, multiple myeloma, erythro leukemia, villous colon adenoma, and osteosarcoma.
  • the condition is an autoimmune disorder.
  • a method of treating a disease or condition mediated by PIM kinase comprising administering to a subject in need thereof a therapeutically effective amount of a compound according to any one of embodiments 1-25, or a pharmaceutically acceptable salt thereof.
  • the disease is selected from carcinoma of the lungs, pancreas, thyroid, ovaries, bladder, breast, prostate or colon, melanoma, myeloid leukemia, multiple myeloma, erythro leukemia, villous colon adenoma, and osteosarcoma; or the disease is an autoimmune disorder.
  • the disease is an autoimmune disorder.
  • the autoimmune disorder is selected from Crohn's disease, inflammatory bowel disease, rheumatoid arthritis, and chronic inflammatory diseases.
  • Ring A can be pyridyl or certain N- alkylpyrazoles; preferred embodiments of Ring A include:
  • R is methyl, ethyl or isopropyl, especially Me
  • Z can be CH or N; when Z is N, preferred embodiments of the ring containing Z include:
  • R 1 is H or OH
  • R 2 is H or Me, provided that when R 1 is H, R 2 is also H
  • R 3 is H, Me, Et or iPr.
  • preferred embodiments of the ring containing Z include:
  • R 1 is H or OH
  • R 2 is H or Me, provided that when R 1 is H, R 2 is also H
  • R 3 is Me, Et or iPr.
  • n is preferably 1, while n is often 2 when Z is N.
  • the substituent R 4 can influence in vivo properties such as metabolism and clearance rates as well as drug interactions, particularly in combination with the above embodiments of the ring containing Z.
  • Some of the preferred embodiments of R 4 in compounds of the invention include: Me, OMe, iPr, -OiPr, -CH 2 OEt, -CH 2 OiPr, - OCH 2 CH 2 OMe, and groups comprising optionally substituted cyclic ethers, including these:
  • R 1 is preferably H or OH, and R 2 is preferably H.
  • R 3 is typically Me; or R 3 can be H, especially when R 1 is H and/or n is 2.
  • Each enantiomer can be used, and preferably the compound to be used is the enantiomer that has greater activity as a Pirn inhibitor.
  • a therapeutically effective dose will generally be a total daily dose administered to a host in single or divided doses may be in amounts, for example, of from 0.001 to 1000 mg/kg body weight daily, typically 0.01 to 100 mg/kg per day, and more preferred from 0.1 to 30 mg/kg body weight daily. Generally, daily dosage amounts of 1 to 4000 mg, or from 5 to 3000, or from 10 to 2000 mg, or from 100 to 2000 mg are anticipated for human subjects. Dosage unit compositions may contain such amounts of submultiples thereof to make up the daily dose.
  • the compounds of the present invention may be administered orally, parenterally, sublingually, by aerosolization or inhalation spray, rectally, or topically in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. Topical administration may also involve the use of transdermal administration such as transdermal patches or ionophoresis devices.
  • parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection, or infusion techniques. In preferred embodiments, the compound or composition of the invention is administered orally.
  • sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-propanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable nonirritating excipient such as cocoa butter and polyethylene glycols, which are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • a suitable nonirritating excipient such as cocoa butter and polyethylene glycols, which are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules.
  • the active compound may be admixed with at least one inert diluent such as sucrose lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate.
  • the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water.
  • Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, cyclodextrins, and sweetening, flavoring, and perfuming agents.
  • the compounds of the present invention can also be administered in the form of liposomes.
  • liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used.
  • the present compositions in liposome form can contain, in addition to a compound of the present invention, stabilizers, preservatives, excipients, and the like.
  • the preferred lipids are the phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes are known in the art. See, for example, Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.W., p. 33 et seq. (1976).
  • the compounds of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more other agents used in the treatment of cancer.
  • the compounds of the present invention are also useful in combination with known therapeutic agents and anti-cancer agents, and combinations of the presently disclosed compounds with other anti-cancer or chemotherapeutic agents are within the scope of the invention. Examples of such agents can be found in Cancer Principles and Practice of Oncology, V. T. Devita and S. Hellman (editors), 6 th edition (Feb. 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved.
  • Such anti-cancer agents include, but are not limited to, the following: MEK inhibitors, estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic/cytostatic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors and other angiogenesis inhibitors, inhibitors of cell proliferation and survival signaling, apoptosis inducing agents and agents that interfere with cell cycle checkpoints.
  • the compounds of the invention are also useful when co- administered with radiation therapy.
  • the compounds of the invention are also used in combination with known therapeutic or anticancer agents including, for example, estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors.
  • known therapeutic or anticancer agents including, for example, estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors.
  • representative therapeutic agents useful in combination with the compounds of the invention for the treatment of cancer include, for example, MEK inhibitors, irinotecan, topotecan, gemcitabine, 5-fluorouracil, cytarabine, daunorubicin, PI3 Kinase inhibitors, mTOR inhibitors, DNA synthesis inhibitors, leucovorin carboplatin, cisplatin, taxanes, tezacitabine, cyclophosphamide, vinca alkaloids, imatinib (Gleevec), anthracyclines, rituximab, trastuzumab, Revlimid, Velcade, dexamethasone, daunorubicin, cytaribine, clofarabine, Mylotarg, lenalidomide, bortezomib, as well as other cancer
  • chemotherapeutic agents including targeted therapeutics.
  • the compounds of the invention and the other anticancer agents can be administered at the recommended maximum clinical dosage or at lower doses. Dosage levels of the active compounds in the compositions of the invention may be varied so as to obtain a desired therapeutic response depending on the route of administration, severity of the disease and the response of the patient.
  • the combination can be administered as separate compositions or as a single dosage form containing both agents.
  • the therapeutic agents can be formulated as separate compositions, which are given at the same time or different times, or the therapeutic agents, can be given as a single composition.
  • the invention provides a method of inhibiting Piml, Pim2 or Pim3 in a human or animal subject.
  • the method includes administering an effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any of the embodiments of compounds of Formula I or II to a subject in need thereof.
  • 5-alkyl, 4-hydroxy, 3-aminopiperidines can be prepared and modified to yield 5-alkyl, 4-substituted, 3-aminopiperidinyl pyridine amides VI as follows. Reaction of Garner's aldehyde with (R)-4-benzyl-3- propionyloxazolidin-2-one followed by TBS protection of the resulting alcohol affords compound I. Reduction of the oxazolidinone followed by introduction of the azide group yields intermediate II. Deprotection under acidic conditions reveals the corresponding amino alcohol, which upon protection with the Boc group followed by mesylation of the primary alcohol yields intermediate III.
  • Methyl cyclohexanedione can be converted via the monotriflate to the corresponding cyclohexenoneboronate ester which can undergo palladium mediated carbon bond formation with 4-chloro, 3-nitro pyridine to yield nitropyridine substituted cyclohexenone VII.
  • Ketone reduction followed by dehydration yields a cyclohexadiene which upon epoxidation (via bromohydrin formation and HBr elimination), azide epoxide opening, azide reduction and amine Boc protection yields cyclohexenyl Boc amino alcohol nitro pyridyl compound VIII.
  • Nitro pyridyl VIII can be converted to the trans protected amino hydroxy aniline IX by alcohol protection and alkene and nitro reduction.
  • substituted piperidine compounds of the invention upon amide coupling of the cyclohexyl pyridyl anilines IX to heterocyclic acids and subsequent hydroxyl and amine deprotection, substituted cyclohexyl compounds of the invention X can be prepared.
  • cyclohexanediones can be converted via monotriflates to the corresponding cyclohexenoneboronate esters which can undergo palladium mediated carbon bond formation with 4-chloro, 3-nitro pyridine to yield nitropyridine substituted cyclohexenones XI.
  • Reduction of the enone functionality can yield a cyclohexenol XII, which can undergo Mitsunobu reaction with phthalimide to yield a protected aminocyclohexene XIII.
  • Boc protected aminocyclohexane pyridyl aniline XV can also be prepared from cyclohexenol XII in the following manner: alcohol protection, alkene and nitro reduction, pyridyl amine Cbz protection, silyl ether deprotection, Dess-Martin oxidation to the cyclohexanone, reductive amination with benzylamine, Cbz and Bn deprotection and primary aliphatic amine Boc protection.
  • Penultimate amide products which contain a heteroaromatic bromo functionality can be further modified by standard modifications to introduce substituted aryls, alkyls and heteroaryls on place of !3 ⁇ 4.
  • R2 is Br
  • R2 replacements are possible.
  • the compounds and/or intermediates were characterized by high performance liquid chromatography (HPLC) using a Waters Millenium chromatography system with a 2695 Separation Module (Milford, MA).
  • HPLC high performance liquid chromatography
  • the analytical columns were reversed phase Phenomenex Luna CI 8 -5 ⁇ , 4.6 x 50 mm, from Alltech (Deerfield, IL).
  • a gradient elution was used (flow 2.5 mL/min), typically starting with 5% acetonitrile/95% water and progressing to 100% acetonitrile over a period of 10 minutes. All solvents contained 0.1%) trif uoroacetic acid (TFA).
  • UV ultraviolet light
  • HPLC solvents were from Burdick and Jackson (Muskegan, MI), or Fisher Scientific (Pittsburgh, PA).
  • TLC thin layer chromatography
  • glass or plastic backed silica gel plates such as, for example, Baker-Flex Silica Gel 1B2-F flexible sheets.
  • TLC results were readily detected visually under ultraviolet light, or by employing well-known iodine vapor and other various staining techniques.
  • Mass spectrometric analysis was performed on one of three LCMS instruments: a Waters System (Alliance HT HPLC and a Micromass ZQ mass spectrometer; Column: Eclipse XDB-C18, 2.1 x 50 mm; gradient: 5-95% (or 35-95%, or 65-95% or 95-95%) acetonitrile in water with 0.05% TFA over a 4 min period; flow rate 0.8 mL/min; molecular weight range 200-1500; cone Voltage 20 V; column temperature 40°C), another Waters System (ACQUITY UPLC system and a ZQ 2000 system; Column: ACQUITY UPLC HSS-C18, 1.8um, 2.1 x 50mm; gradient: 5-95% (or 35-95%, or 65-95% or 95-95%) acetonitrile in water with 0.05% TFA over a 1.3 min period; flow rate 1.2 mL/min; molecular weight range 150-850; cone Voltage 20 V; column temperature 50°C) or
  • NMR Nuclear magnetic resonance
  • Preparative separations are carried out using a Flash 40 chromatography system and KP-Sil, 60A (Biotage, Charlottesville, VA), or by flash column chromatography using silica gel (230-400 mesh) packing material on ISCO or Analogix purification systems, or by HPLC using a Waters 2767 Sample Manager, C-18 reversed phase column, 30X50 mm, flow 75 mL/min.
  • Typical solvents employed for the Flash 40 Biotage, ISCO or Analogixsystem for silica gel column chromatography are dichloromethane, methanol, ethyl acetate, hexane, n-heptanes, acetone, aqueous ammonia (or ammonium hydroxide), and triethyl amine.
  • Typical solvents employed for the reverse phase HPLC are varying concentrations of acetonitrile and water with 0.1% trifluoroacetic acid.
  • the separation used a Chiralpak AD (AS, OD, OJ, IC or IA) 4.6x100mm column at 40C temperature at a flow rate of 5 mL/min using an isocratic method.
  • the mobile phase was 15% MeOH (or EtOH or IPA or with 0.1% Diethyl amine): 85% C02.
  • the detection wavelength was 220 nm (or 250nm or Diode Array).
  • Chiral SFC-Purification Method Chiral compounds were separated on a Waters Supercritical Fluid Chromatography (SFC). The separation used a Chiralpak AD (AS, OD, OJ, IC or IA) 21x250mm column at 40C temperature at a flow rate of 100 mL/min using an isocratic method. The mobile phase was 15% MeOH (or EtOH or IPA or with 0.1% Diethyl amine): 85% C02. The detection wavelength was 220 nm (or 250nm or Diode Array).
  • SFC Waters Supercritical Fluid Chromatography
  • Chiral HPLC-Analytical Method Chiral compounds were separated on a Waters 2695 HPLC system. The separation used a Chiralpak AD (AS, OD, OJ, IC or IA) 4.6x100mm column at room temperature at a flow rate of 1 mL/min using an isocratic method. The mobile phase was 15% EtOH (or IPA or with 0.1% Diethyl amine): 85% Heptane. The detection wavelength was 220 nm (or 250nm or Diode Array).
  • Chiralpak AD AS, OD, OJ, IC or IA
  • the mobile phase was 15% EtOH (or IPA or with 0.1% Diethyl amine): 85% Heptane.
  • the detection wavelength was 220 nm (or 250nm or Diode Array).
  • Chiral HPLC-Purification Method Chiral compounds were separated on a Waters 2767 HPLC system. The separation used a Chiralpak AD (AS, OD, OJ, IC or IA) 21x250mm column at room temperature at a flow rate of 20 (or 10 -15) mL/min using an isocratic method. The mobile phase was 15% EtOH (or IPA or with 0.1% Diethyl amine): 85% Heptane. The detection wavelength was 220 nm (or 250nm or Diode Array).
  • Chiralpak AD AS, OD, OJ, IC or IA 21x250mm column at room temperature at a flow rate of 20 (or 10 -15) mL/min using an isocratic method.
  • the mobile phase was 15% EtOH (or IPA or with 0.1% Diethyl amine): 85% Heptane.
  • the detection wavelength was 220 nm (or 250nm or Diode Array).
  • organic compounds according to the preferred embodiments may exhibit the phenomenon of tautomerism.
  • chemical structures within this specification can only represent one of the possible tautomeric forms, it should be understood that the preferred embodiments encompasses any tautomeric form of the drawn structure.
  • the residue was partitioned between brine and ethyl acetate, and the layers were separated, the aqueous phase was further extracted with ethyl acetate (4x), the organics were combined, dried over sodium sulfate, filtered, and concentrated.
  • the crude was purified via silica gel chromatography loading in DCM and eluting with 2-50% ethyl acetate and hexanes. The pure fractions were concentrated in vacuo to yield an orange oil.
  • (+/-)-2-azido-6-methyl-4-(3-nitropyridin-4- yl)cyclohex-3-enol 1.0 equiv.
  • ammonium hydroxide 8:1, 0.08 M
  • trimethylphosphine 3.0 equiv.
  • EtOH was added and the solution was concentrated in vacuo. More ethanol was added and the reaction was concentrated again.
  • Dioxane and sat. NaHC0 3 (1 : 1, 0.08 M) were added to the crude, followed by Boc 2 0 (1.0 equiv.).
  • the reaction was cooled to room temperature then quenched with water and diluted with EtOAc. The aqueous layer was separated then extracted with EtOAc. The combined organics were dried over MgSC ⁇ and concentrated in vacuo to yield a brown oil.
  • the crude product was purified by ISCO flash chromatography eluting with ethyl acetate and hexanes (0% to 30% ethyl acetate) to provide ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- methoxyphenyl)thiazole-4-carboxylate as the desired product as a white solid in 73% yield.
  • Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.2 equiv.), butyllithium (1.2 equiv.) and l,3-difluoro-5-isopropoxybenzene (1.0 equiv.) to give 2-(2,6-difluoro-4-isopropoxyphenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane in 99% yield.
  • Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (1.5 equiv.), butyllithium (1.3 equiv.) and 4-(3,5-difluorophenoxy)tetrahydro-2H-pyran (1.0 equiv.) to give 2-(2,6-difluoro-4-((tetrahydro-2H-pyran-4-yl)oxy)phenyl)-4,4,5,5- tetramethyl-l,3,2-dioxaborolane in 33% yield.
  • Method 1 was followed using methyl ethyl 2-bromo-5-((tert- butoxycarbonyl)amino)thiazole-4-carboxylate (0.8 equiv.) and 2-(2,6-difluoro-4- ((tetrahydro-2H-pyran-4-yl)oxy)phenyl)-4,4,5,5-tetramethyl-l ,3,2-dioxaborolane (1.0 equiv.) at 100 °C under microwave irradiation for 20 min to give ethyl 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-((tetrahydro-2H-pyran-4- yl)oxy)phenyl)thiazole-4-carboxylate in 67% yield.
  • Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.5 equiv.), butyllithium (2.4 equiv.) and 3-(3,5-difluorophenyl)oxetan-3-ol (1.0 equiv.) to give 3-(3,5-difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)oxetan-3-ol in 79% yield.
  • Method 1 was followed using ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4- carboxylate (1.0 equiv.) and 3-(3,5-difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl)oxetan-3-ol (2.0 equiv.) at 100 °C under microwave irradiation for 20 min to give ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3-hydroxyoxetan-3- yl)phenyl)thiazole-4-carboxylate in 50% yield.
  • Method 1 was followed using ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4- carboxylate (1.0 equiv.) and 2-(2,6-difluoro-4-(tetrahydro-2H-pyran-4-yl)phenyl)-4,4,5,5- tetramethyl-l,3,2-dioxaborolane (2.0 equiv.) at 100 0 C for 20 min under microwave irradiation to give ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(tetrahydro-2H- pyran-4-yl)phenyl)thiazole-4-carboxylate in 84% yield.
  • reaction solution was quenched by addition of NH 4 Cl (sat) and the solution was extracted with EtOAc, washed with NaCl (sa ) , dried over MgS0 4 , filtered, concentrated and purified by ISCO Si0 2 chromatography (0-100% EtOAc/n-heptanes gradient) to yield 4-(3,5-difluorophenyl)tetrahydro-2H-pyran-4-ol in 71% yield.
  • Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.5 equiv.), butyllithium (2.4 equiv.) and 4-(3,5-difluorophenyl)tetrahydro-2H-pyran-4-ol (1.0 equiv.) to give 4-(3,5-difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl)tetrahydro-2H-pyran-4-ol in 97% yield.
  • Method 1 was followed using methyl ethyl 2-bromo-5-((tert- butoxycarbonyl)amino)thiazole-4-carboxylate (1.0 equiv.) and 4-(3,5-difluoro-4-(4,4,5,5- tetramethyl-l ,3,2-dioxaborolan-2-yl)phenyl)tetrahydro-2H-pyran-4-ol (2.0 equiv.) at 100 °C for 20 min under microwave irradiation to give ethyl 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4-hydroxytetrahydro-2H-pyran-4- yl)phenyl)thiazole-4-carboxylate in 70% yield.
  • Method 1 was followed using ethyl 2-bromo-5-(tert-butoxycarbonylamino)thiazole-4- carboxylate (1.0 equiv.) and 2-(2,6-difluoro-4-(tetrahydro-2H-pyran-4-yl)phenyl)- 4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.0 equiv.) at 100 °C for 20 min in microwave to give ethyl 5-(tert-butoxycarbonylamino)-2-(2,6-difluoro-4-(tetrahydro-2H-pyran-4- yl)phenyl)thiazole-4-carboxylate in 84% yield.
  • reaction solution was quenched by addition of NH 4 Cl( sa t) and the solution was extracted with EtOAc, washed with NaCl(sat), dried over MgS04, filtered, concentrated and purified by ISCO Si0 2 chromatography (0-100% EtOAc/n-heptanes gradient) to yield l-(3,5-difluorophenyl)cyclobutanol in 54% yield.
  • Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2- dioxaborolane (2.5 equiv.), butyllithium (2.4 equiv.) and l-(3,5- difluorophenyl)cyclobutanol (1.0 equiv.) to give l-(3,5-difluoro-4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)phenyl)cyclobutanol in 100% yield.
  • N-Boc protected amine was present, it was removed by treating with excess 4M HCl/ dioxane for 14 hours or by treating with 25% TFA/CH 2 C1 2 for 2 hours. Upon removal of the volatiles in vacuo, the material was purified by RP HPLC yielding after lyophilization the amide product as the TFA salt. Alternatively, the HPLC fractions could be added to EtOAc and solid Na 2 C0 3 , separated and washed with NaCl (sa ) . Upon drying over MgS0 4 , filtering and removing the volatiles in vacuo the free base was obtained. Upon dissolving in MeCN/H 2 0, adding 1 eq. of 1 N HCl and lyophilizing, the HCl salt of the amide product was obtained.
  • the acetate group could be cleaved by treating with K 2 C0 3 (2.0 equiv.) in ethanol at a concentration of 0.1 M for 24 hours.
  • a TBDMS ether was present, it was deprotected prior to Boc removal by treating with 6N HCl, THF, methanol (1 :2: 1) at room temperature for 12 h. After removal of volatiles in vacuo, the Boc amino group was deprotected as described above.
  • the TBDMS ether and Boc group could be both deprotected with 6N HCl, THF, methanol (1 :2: 1) if left at rt for 24 hours, or heated at 60 °C for 3 hours.
  • Pim 1, Pim 2 & Pim 3 AlphaScreen assays using high ATP (11 - 125X ATP Km) were used to determine the biochemical activity of the inhibitors.
  • the activity of Pim 1, Pim 2, & Pim 3 is measured using a homogeneous bead based system quantifying the amount of phosphorylated peptide substrate resulting from kinase-catalyzed phosphoryl transfer to a peptide substrate.
  • Compounds to be tested are dissolved in 100% DMSO and directly distributed to a white 384-well plate at 0.25 ⁇ per well.
  • IC50 the half maximal inhibitory concentration
  • KMS11 human myeloma cell line
  • IMDM IMDM supplemented with 10%> FBS, sodium pyruvate and antibiotics.
  • Cells were plated in the same medium at a density of 2000 cells per well into 96 well tissue culture plates, with outside wells vacant, on the day of assay.
  • Test compounds supplied in DMSO were diluted into DMSO at 500 times the desired final concentrations before dilution into culture media to 2 times final concentrations. Equal volumes of 2x compounds were added to the cells in 96 well plates and incubated at 37 °C for 3 days.

Abstract

The present invention provides a compound of formula (I), as described herein, and pharmaceutically acceptable salts, enantiomers, rotamers, tautomers, or racemates thereof. Also provided are methods of treating a disease or condition mediated by PIM kinase using the compounds of Formula I, and pharmaceutical compositions comprising such compounds.

Description

NOVEL AMINOTHIAZOLE CARBOXAMIDES AS KINASE INHIBITORS
FIELD OF THE INVENTION
The present invention relates to new compounds and compositions of the new compounds together with pharmaceutically acceptable carriers, and uses of the new compounds, either alone or in combination with at least one additional therapeutic agent, in the prophylaxis or treatment of cancer and other cellular proliferation disorders.
BACKGROUND
Infection with the Moloney retrovirus and genome integration in the host cell genome results in development of lymphomas in mice. Provirus Integration of Moloney Kinase (PIM-Kinase) was identified as one of the frequent proto-oncogenes capable of being transcriptionally activated by this retrovirus integration event (Cuypers HT et al, "Murine leukemia virus-induced T-cell lymphomagenesis: integration of pro viruses in a distinct chromosomal region," Cell 37(1): 141-50 (1984); Selten G, et al, "Proviral activation of the putative oncogene Pim-1 in MuLV induced T-cell lymphomas" EMBO J 4(7): 1793-8 (1985)), thus establishing a correlation between over-expression of this kinase and its oncogenic potential. Sequence homology analysis demonstrated that there are three highly homologous Pim-Kinases (Piml, 2 & 3), Piml being the proto-oncogene originally identified by retrovirus integration. Furthermore, transgenic mice over- expressing Piml or Pim2 show increased incidence of T-cell lymphomas (Breuer M et al., "Very high frequency of lymphoma induction by a chemical carcinogen in pim-1 transgenic mice" Nature 340(6228):61-3 (1989)), while over-expression in conjunction with c-myc is associated with incidence of B-cell lymphomas (Verbeek S et al., "Mice bearing the E mu-myc and E mu-pim-1 transgenes develop pre-B-cell leukemia prenatally" Mol Cell Biol 11(2): 1176-9 (1991)). Thus, these animal models establish a strong correlation between Pirn over-expression and oncogenesis in hematopoietic malignancies. In addition to these animal models, Pirn over-expression has been reported in many human malignancies, particularly in hematopoietic and in prostate cancer. Furthermore, mutational activation of several well known oncogenes in hematopoietic malignancies is thought to exert its effects at least in part through Pim(s). For example, targeted down-regulation of Pirn expression impairs survival of hematopoietic cells transformed by Flt3 and BCR/ABL (Adam et al. 2006). Piml, 2 & 3 are Serine/Threonine kinases that normally function in survival and proliferation of hematopoietic cells in response to growth factors and cytokines. Substrates for Pim kinases include regulators of apoptosis such as the Bcl-2 family member BAD. The effects of Pim(s) in these regulators are consistent with a role in protection from apoptosis and promotion of cell proliferation and growth. Thus, over- expression of Pim(s) in cancer is thought to play a role in promoting survival and proliferation of cancer cells and, therefore, their inhibitions should be an effective way of treating cancers in which they are over-expressed. In fact several reports indicate that knocking down expression of Pim(s) with siRNA results in inhibition of proliferation and cell death (Dai JM, et al., "Antisense oligodeoxynucleotides targeting the serine/threonine kinase Pim-2 inhibited proliferation of DU-145 cells," Acta Pharmacol Sin 26(3):364-8 (2005); Fujii et al. 2005; Li et al. 2006). Thus, inhibitors to Piml, 2 and 3 would be useful in the treatment of these malignancies.
In addition to a potential role in cancer treatment and myeloproliferative diseases, such inhibitors could be useful to control expansion of immune cells in other pathologic condition such as autoimmune diseases, allergic reactions and in organ transplantation rejection syndromes. Recent reports demonstrate that Pim kinase inhibitors show activity in animal models of inflammation and autoimmune diseases. See JE Robinson "Targeting the Pim Kinase Pathway for Treatment of Autoimmune and Inflammatory Diseases," for the Second Annual Conference on Anti-Inflammatories: Small Molecule Approaches," San Diego, CA (Conf. April 2011; Abstract published earlier on-line).
A continuing need exists for compounds that inhibit the proliferation of capillaries, inhibit the growth of tumors, treat cancer, modulate cell cycle arrest, and/or inhibit molecules such as Piml, Pim2 and Pim3, and pharmaceutical formulations and medicaments that contain such compounds. A need also exists for methods of administering such compounds, pharmaceutical formulations, and medicaments to patients or subjects in need thereof. The present invention addresses such needs.
Earlier patent applications have described compounds that inhibit Pirns and function as anticancer therapeutics, see, e.g., WO2012/004217, WO2010/026124, WO 2008/106692 and WO2011/124580, and as treatment for inflammatory conditions such as Crohn's disease, inflammatory bowel disease, rheumatoid arthritis, and chronic inflammatory diseases, see e.g., WO 2008/022164. The present invention provides novel compounds that inhibit activity of one or more Pirns, preferably two or more Pirns, more preferably Piml, Pim2 and Pim3, at nanomolar levels (e.g., IC-50 under 50 nM) and exhibit distinctive characteristics that may provide improved therapeutic effects and pharmacokinetic properties, such as reduced drug-drug interactions associated with inhibition of cytochrome oxidases, relative to compounds previously disclosed. Compounds of the invention contain novel substitution combinations on one or more rings that provide these distinctive properties and are suitable for treating Pim-related conditions such as those described herein.
SUMMARY OF THE INVENTION
The invention provides compounds of Formula (I) that inhibit one or more Pirn kinases:
Figure imgf000004_0001
(I)
wherein:
Figure imgf000004_0002
represents a pyridine ring or N-(Ci-C4 alkyl)-pyrazole ring;
W is H or NH2;
Z is CH or N;
n is 1 or 2;
ft1 is H, OH or OMe;
Figure imgf000004_0003
provided that when R1 is H, R2 is also H;
R3 is H or Ci-C4 alkyl; R4 is selected from R*, -OR*, -OCH2CH2OR*, -CH2OR*, and a 4-6 membered cyclic ether optionally substituted with OH, OMe, or F, wherein each R* is independently C 1-C4 alkyl,
provided that R4 is not -OMe when Z is N;
or a pharmaceutically acceptable salt thereof.
Additional embodiments of these compounds and pharmaceutical compositions and uses for these compounds and compositions are described below.
These compounds are inhibitors of Pirn kinases as further discussed herein. These compounds and their pharmaceutically acceptable salts, and pharmaceutical compositions containing these compounds and salts, are useful for therapeutic methods such as treatment of cancers and autoimmune disorders that are caused by or exacerbated by excessive levels of Pirn kinase activity. DETAILED DESCRIPTION OF EMBODIMENTS OF THE INVENTION
"PIM inhibitor" is used herein to refer to a compound that exhibits an IC50 with respect to PIM Kinase activity of no more than about 100 μΜ and more typically not more than about 5 μΜ, as measured in the PIM depletion assays described herein below for at least one of Piml, Pim2 and Pim3. Preferred compounds have on IC50 below about 1 micromolar on at least one Pirn, and generally have an IC50 below 100 nM on each of Piml, Pim2 and Pim3.
The phrase "alkyl" refers to hydrocarbon groups that do not contain heteroatoms, i.e., they consist of carbon atoms and hydrogen atoms. Thus the phrase includes straight chain alkyl groups such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl, undecyl, dodecyl and the like. The phrase also includes branched chain isomers of straight chain alkyl groups, including but not limited to, the following which are provided by way of example: -
CH(CH3)2, -CH(CH3)(CH2CH3), -CH(CH2CH3)2, -C(CH3)3, -C(CH2CH3)3, -CH2CH(CH3) 2, -CH2CH(CH3)(CH2CH3), -CH2CH(CH2CH3)2, -CH2C(CH3)3, -CH2C(CH2CH3)3, -CH(C ¾)-
CH(CH3)(CH2CH3), -CH2CH2CH(CH3)2, -CH2CH2CH(CH3)(CH2CH3), -CH2CH2CH(CH 2CH3)2, -CH2CH2C(CH3)3, -CH2CH2C(CH2CH3)3, -CH(CH3)CH2_
CH(CH3)2, -CH(CH3)CH(CH3)CH(CH3)2, -CH(CH2CH3)CH(CH3)CH(CH3)(CH2CH3), and others. Thus the term 'alkyl' includes primary alkyl groups, secondary alkyl groups, and tertiary alkyl groups. Alkyl groups are described herein according to the number of carbon atoms they contain, e.g., an alkyl group containing up to six carbon atoms is described as a CI -6 or C1-6, or C1-C6 alkyl. Typical alkyl groups include straight and branched chain alkyl groups having 1 to 12 carbon atoms, preferably 1-6 carbon atoms. The term 'lower alkyl' or "loweralkyl" and similar terms refer to alkyl groups containing up to 6 carbon atoms.
The term "alkenyl" refers to alkyl groups as defined above, wherein there is at least one carbon-carbon double bond, i.e., wherein two adjacent carbon atoms are attached by a double bond. The term "alkynyl" refers to alkyl groups wherein two adjacent carbon atoms are attached by a triple bond. Typical alkenyl and alkynyl groups contain 2-12 carbon atoms, preferably 2-6 carbon atoms. Lower alkenyl or lower alkynyl refers to groups having up to 6 carbon atoms. An alkenyl or alkynyl group may contain more than one unsaturated bond, and may include both double and triple bonds, but of course their bonding is consistent with well-known valence limitations.
The term 'alkoxy" refers to -OR, wherein R is alkyl.
As used herein, the term "halogen" or "halo" refers to chloro, bromo, fluoro and iodo groups. Typical halo substituents are F and/or CI. "Haloalkyl" refers to an alkyl radical substituted with one or more halogen atoms, typically 1-3 halogen atoms. The term "haloalkyl" thus includes monohalo alkyl, dihalo alkyl, trihalo alkyl, perhaloalkyl, and the like.
"Amino" refers herein to the group -NH2. The term "alkylamino" refers herein to the group -NRR where R and R are each independently selected from hydrogen or a lower alkyl, provided -NRR' is not -NH2. The term "arylamino" refers herein to the group -NRR where R is aryl and R' is hydrogen, a lower alkyl, or an aryl. The term "aralkylamino" refers herein to the group -NRR where R is a lower aralkyl and R is hydrogen, a loweralkyl, an aryl, or a loweraralkyl.
The term "alkoxyalkyl" refers to the group -alki-0-alk2 where alki is an alkyl linking group, and alk2 is alkyl, e.g., a group such as -0-(CH2)2-0-CH3. The term "loweralkoxyalkyl" refers to an alkoxyalkyl where alki is loweralkyl and alk2 is loweralkyl. The term "aryloxyalkyl" refers to the group -alkyl-O-aryl, where -alkyl- is a Ci_i2 straight or branched chain alkyl linking group, preferably C1-6. The term "aralkoxyalkyl" refers to the group -alkyl-O-aralkyl, where aralkyl is preferably a loweraralkyl.
The term "aminocarbonyl" refers herein to the group -C(0)-NH2 . "Substituted aminocarbonyl" refers herein to the group -C(0)-NRR' where R is loweralkyl and R' is hydrogen or a loweralkyl. In some embodiments, R and R, together with the N atom attached to them may be taken together to form a "heterocycloalkylcarbonyl" group. The term "arylaminocarbonyl" refers herein to the group -C(0)-NRR where R is an aryl and R is hydrogen, loweralkyl or aryl. "aralkylaminocarbonyl" refers herein to the group - C(0)-NRR' where R is loweraralkyl and R is hydrogen, loweralkyl, aryl, or loweraralkyl.
"Aminosulfonyl" refers herein to the group -S(0)2-NH2. "Substituted aminosulfonyl" refers herein to the group -S(0)2-NRR' where R is loweralkyl and R' is hydrogen or a loweralkyl. The term "aralkylaminosulfonylaryl" refers herein to the group -aryl-S(0)2-NH-aralkyl, where the aralkyl is loweraralkyl.
"Carbonyl" refers to the divalent group -C(O)-. "Carboxy" refers to-C(=0)-OH.
"Alkoxycarbonyl" refers to ester -C(=0)-OR wherein R is optionally substituted lower alkyl. "Loweralkoxycarbonyl" refers to ester -C(=0)-OR wherein R is optionally substituted lower loweralkyl. "Cycloalkyloxycarbonyl" refers to -C(=0)-OR wherein R is optionally substituted C3-C8 cycloalkyl.
"Cycloalkyl" refers to a mono- di- or poly-cyclic, carbocyclic alkyl substituent in which all ring atoms are carbon. Typical cycloalkyl groups have from 3 to 8 backbone (i.e., ring) atoms. When used in connection with cycloalkyl substituents, the term "polycyclic" refers herein to fused and non-fused alkyl cyclic structures, including spirocyclic ring systems. The term "partially unsaturated cycloalkyl", "partially saturated cycloalkyl", and "cycloalkenyl" all refer to a cycloalkyl group wherein there is at least one unsaturated carbon-carbon bond in the ring, i.e., wherein two adjacent ring atoms are connected by a double bond or a triple bond. Such rings typically contain 1 or 2 double bonds for 5-6 membered rings, and 1-2 double bonds or one triple bond for 7-8 membered rings. Illustrative examples include cyclohexenyl, cyclooctynyl, cyclopropenyl, cyclobutenyl, cyclohexadienyl, and the like.
The term "heterocycloalkyl" refers herein to cycloalkyl substituents that have from 1 to 5, and more typically from 1 to 3 heteroatoms as ring members in place of carbon atoms. Preferably, heterocycloalkyl or "heterocyclyl" groups contain one or two heteroatoms as ring members, typically only one heteroatom for 3-5 membered rings and 1-2 heteroatoms for 6-8 membered rings. Suitable heteroatoms employed in heterocyclic groups of the present invention are nitrogen, oxygen, and sulfur. Representative heterocycloalkyl moieties include, for example, pyrrolidinyl, tetrahydrofuranyl, oxirane, oxetane, oxepane, thiirane, thietane, azetidine, morpholino, piperazinyl, piperidinyl and the like.
The terms "substituted heterocycle", "heterocyclic group" or "heterocycle" as used herein refers to any 3- or 4-membered ring containing a heteroatom selected from nitrogen, oxygen, and sulfur or a 5- or 6-membered ring containing from one to three heteroatoms, preferably 1-2 heteroatoms, selected from the group consisting of nitrogen, oxygen, or sulfur; wherein the 5 -membered ring has 0-2 double bonds and the 6-membered ring has 0-3 double bonds; wherein the nitrogen and sulfur atom maybe optionally oxidized; wherein the nitrogen and sulfur heteroatoms may be optionally quaternized; and including any bicyclic group in which any of the above heterocyclic rings is fused to a benzene ring or another 5- or 6-membered heterocyclic ring or heteroaryl as described herein. Preferred heterocycles include, for example: diazapinyl, pyrrolinyl, pyrrolidinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, piperidinyl, piperazinyl, N-methyl piperazinyl, azetidinyl, N-methylazetidinyl, oxazolidinyl, isoxazolidinyl, morpholinyl, thiazolidinyl, isothiazolidinyl, and oxiranyl. The heterocyclic groups may be attached at various positions as will be apparent to those having skill in the organic and medicinal chemistry arts in conjunction with the disclosure herein.
Heterocyclic moieties can be unsubstituted or they can be substituted with one or more substituents independently selected from hydroxy, halo, oxo (C=0), alkylimino (RN=, wherein R is a loweralkyl or loweralkoxy group), amino, alkylamino, dialkylamino, acylaminoalkyl, alkoxy, thioalkoxy, lower alkoxyalkoxy, loweralkyl, cycloalkyl or haloalkyl. Typically, substituted heterocyclic groups will have up to four substituent groups.
The term "cyclic ether" as used herein refers to a 3-7 membered ring, unless otherwise specified, containing one oxygen atom (O) as a ring member. Where the cyclic ether is "optionally substituted" it can be substituted at any carbon atom with a group suitable as a substituent for a heterocyclic group, typically up to three substituents selected from lower alkyl, lower alkoxy, halo, hydroxy, amino, -C(0)-lower alkyl, and - C(0)-lower alkoxy. In preferred embodiments, halo, hydroxy and lower alkoxy are not attached to the carbon atoms of the ring that are bonded directly to the oxygen atom in the cyclic ether ring. Specific examples include oxirane, oxetane (e.g., 3-oxetane), tetrahydrofuran (including 2-tetrahydrofuranyl and 3-tetrahydrofuranyl), tetrahydropyran (e.g., 4-tetrahydropyranyl), and oxepane.
"Aryl" refers to monocyclic and polycyclic aromatic groups having from 5 to 14 backbone carbon or hetero atoms, and includes both carbocyclic aryl groups and heteroaromatic aryl groups. Carbocyclic aryl groups are aryl groups in which all ring atoms in the aromatic ring are carbon, typically including phenyl and naphthyl. Exemplary aryl moieties employed as substituents in compounds of the present invention include phenyl, pyridyl, pyrimidinyl, thiazolyl, indolyl, imidazolyl, oxadiazolyl, tetrazolyl, pyrazinyl, triazolyl, thiophenyl, furanyl, quinolinyl, purinyl, naphthyl, benzothiazolyl, benzopyridyl, and benzimidazolyl, and the like. When used in connection with aryl substituents, the term "polycyclic aryl" refers herein to fused and non-fused cyclic structures in which at least one cyclic structure is aromatic, such as, for example, benzodioxozolo (which has a heterocyclic structure fused to a phenyl group, naphthyl, and the like. Where "aryl" is used, the group is preferably a carbocyclic group; the term "heteroaryl" is used for aryl groups when ones containing one or more heteroatoms are preferred.
The term "heteroaryl" refers herein to aryl groups having from 1 to 4 heteroatoms as ring atoms in an aromatic ring with the remainder of the ring atoms being carbon atoms, in a 5-14 atom aromatic ring system that can be monocyclic or polycyclic. Monocyclic heteroaryl rings are typically 5-6 atoms in size. Exemplary heteroaryl moieties employed as substituents in compounds of the present invention include pyridyl, pyrimidinyl, thiazolyl, indolyl, imidazolyl, oxadiazolyl, tetrazolyl, pyrazinyl, triazolyl, thiophenyl, furanyl, quinolinyl, purinyl, benzothiazolyl, benzopyridyl, and benzimidazolyl, and the like.
"Aralkyl" or "arylalkyl" refers to an aryl group connected to a structure through an alkylene linking group, e.g., a structure such as -(CH2)i_4-Ar, where Ar represents an aryl group. "Lower aralkyl" or similar terms indicate that the alkyl linking group has up to 6 carbon atoms.
"Optionally substituted" or "substituted" refers to the replacement of one or more hydrogen atoms with a non-hydrogen group. Alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, and heteroaryl groups described herein may be substituted or unsubstituted. Suitable substitution groups include, for example, hydroxy, nitro, amino, imino, cyano, halo, thio, sulfonyl, thioamido, amidino, imidino, oxo, oxamidino, methoxamidino, imidino, guanidino, sulfonamido, carboxyl, formyl, loweralkyl, haloloweralkyl, loweralkylamino, haloloweralkylamino, lower alkoxy, lower haloalkoxy, lower alkoxyalkyl, alkylcarbonyl, aminocarbonyl, arylcarbonyl, aralkylcarbonyl, heteroarylcarbonyl, heteroaralkylcarbonyl, alkylthio, aminoalkyl, cyanoalkyl, aryl and the like, provided that oxo, imidino or other divalent substitution groups are not placed on aryl or heteroaryl rings due to the well known valence limitations of such rings. In preferred embodiments, unless otherwise specified, optional substituents for alkyl, alkenyl, alkynyl, cycloalkyl, and heterocycloalkyl groups are 1-3 groups selected from halo, hydroxy, amino, cyano, lower alkoxy, lower alkylsulfonyl, oxy, carboxy, and lower alkoxy carbonyl. In preferred embodiments, unless otherwise specified, optional substituents for aryl and heteroaryl groups are 1-3 groups selected from halo, hydroxy, amino, cyano, lower alkyl, lower alkoxy, lower alkylsulfonyl, carboxy, and lower alkoxy carbonyl.
The substitution group can itself be substituted where valence permits, i.e., where the substitution group contains at least one CH, NH or OH having a hydrogen atom that can be replaced. The group substituted onto the substitution group can be carboxyl, halo (on carbon only); nitro, amino, cyano, hydroxy, loweralkyl, loweralkoxy, C(0)R, - OC(0)R, -OC(0)OR, -NRCOR, -CONR2, -NRCOOR, -C(S)NR2, -NRC(S)R, - OC(0)NR2, , -SR, -SO3H, -S02R or C3-8 cycloalkyl or 3-8 membered heterocycloalkyl, where each R is independently selected from hydrogen, lower haloalkyl, lower alkoxyalkyl, and loweralkyl, and where two R on the same atom or on directly connected atoms can be linked together to form a 5-6 membered heterocyclic ring. Unless indicated as optionally substituted, these substitution groups are typically unsubstituted.
When a substituted substituent includes a straight chain group, the substitution can occur either within the chain (e.g., 2-hydroxypropyl, 2-aminobutyl, and the like) or at the chain terminus (e.g., 2-hydroxyethyl, 3-cyanopropyl, and the like). Substituted substituents can be straight chain, branched or cyclic arrangements of covalently bonded carbon or heteroatoms.
It is understood that the above definitions are not intended to include impermissible substitution patterns (e.g., methyl substituted with five fluoro groups or a halogen atom substituted with another halogen atom). Such impermissible substitution patterns are well known to the skilled artisan.
"Syn" as used herein has its ordinary meaning, and is used in connection with Formula I to indicate that the specified groups are attached to sp3 hybridized (tetrahedral) carbon centers and extend out from one face of the cyclohexyl or piperidinyl ring, i.e., those groups all project toward the 'alpha' face of the ring, or they all project toward the 'beta' face of the ring. This is thus used as a convenient way to define the relative orientations of two or more groups on a ring, without limiting the compounds to a specific absolute chiral configuration. This reflects the fact that the compounds of the invention have such groups in a specific relative orientation, but are not limited to either enantiomer of that specific relative orientation. Accordingly, unless described as optically active, such compounds may be racemic, but also include each of the two enantiomers having the specified relative stereochemistry. In some embodiments, the compounds of the invention are optically active form as further described herein, and in preferred embodiments of the invention, the compounds are obtained and used in optically active form. Preferably, the enantiomer having greater potency as an inhibitor of at least two of Piml, Pim2 and Pim3 is selected.
It will also be apparent to those skilled in the art that the compounds of the invention, as well as the pharmaceutically acceptable salts, esters, metabolites and prodrugs of any of them, may be subject to tautomerization and may therefore exist in various tautomeric forms wherein a proton of one atom of a molecule shifts to another atom and the chemical bonds between the atoms of the molecules are consequently rearranged. See, e.g., March, Advanced Organic Chemistry: Reactions, Mechanisms and Structures, Fourth Edition, John Wiley & Sons, pages 69-74 (1992). As used herein, the term "tautomer" refers to the compounds produced by the proton shift, and it should be understood that all tautomeric forms, insofar as they may exist, are included within the invention.
The compounds of the invention comprise one or more asymmetrically substituted carbon atoms. Such asymmetrically substituted carbon atoms can result in the compounds of the invention existing in enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, such as in (R)- or (S)- forms. The compounds of the invention are sometimes depicted herein as single enantiomers, and are intended to encompass the specific configuration depicted and the enantiomer of that specific configuration (the mirror image isomer of the depicted configuration), unless otherwise specified— e.g., where a structure is labeled 'chiral', it represents the specified absolute stereochemistry as a single substantially pure (i.e., at least about 95% pure) enantiomer. The depicted structures herein describe the relative stereochemistry of the compounds where two or more chiral centers, but the invention is not limited to the depicted enantiomer's absolute stereochemistry unless otherwise stated. The invention includes both enantiomers, each of which will exhibit Pim inhibition, even though one enantiomer will be more potent than the other. In some instances, compounds of the invention have been synthesized in racemic form and separated into individual isomers by chiral chromatography or similar conventional methods, and the analytical data about the two enantiomers do not provide definitive information about absolute stereochemical configuration. In such cases, the absolute stereochemistry of the most active enantiomer has been identified based on correlation with similar compounds of known absolute stereochemistry, rather than by a definitive physical method such as X- ray crystallography. Therefore, in certain embodiments, the preferred enantiomer of a compound described herein is the specific isomer depicted or its opposite enantiomer, whichever has the lower IC-50 for Pim kinase inhibition using the assay methods described herein, i.e., the enantiomer that is more potent as a Pim inhibitor for at least two of Pim 1, Pim2, and Pim3.
The terms "S" and "R" configuration, as used herein, are as defined by the IUPAC
1974 RECOMMENDATIONS FOR SECTION E, FUNDAMENTAL STEREOCHEMISTRY, Pure Appl. Chem. 45: 13-30 (1976). The terms a and β are employed for ring positions of cyclic compounds. The a-side of the reference plane is that side on which the preferred substituent lies at the lower numbered position. Those substituents lying on the opposite side of the reference plane are assigned β descriptor. It should be noted that this usage differs from that for cyclic stereoparents, in which "a" means "below the plane" and denotes absolute configuration. The terms a and β configuration, as used herein, are as defined by the CHEMICAL ABSTRACTS INDEX GUIDE -APPENDIX IV (1987) paragraph 203.
As used herein, the term "pharmaceutically acceptable salts" refers to the nontoxic acid or base addition salts of the compounds of Formulas I, II, etc., wherein the compound acquires a positive or negative charge as a result of adding or removing a proton; the salt then includes a counterion of opposite charge from the compound itself, and the counterion is preferably one suitable for pharmaceutical administration under the conditions where the compound would be used. These salts can be prepared in situ during the final isolation and purification of the compounds of Formula I or II, or by separately reacting the base or acid functions with a suitable organic or inorganic acid or base, respectively. Representative salts include but are not limited to the following: acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, cyclopentanepropionate, dodecylsulfate, ethanesulfonate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectinate, persulfate, 3-phenylproionate, picrate, pivalate, propionate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate and undecanoate.
Also, a basic nitrogen-containing group in compounds of the invention can be quaternized with such agents as loweralkyl halides, such as methyl, ethyl, propyl, and butyl chloride, bromides, and iodides; dialkyl sulfates like dimethyl, diethyl, dibutyl, and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides, and others. Water or oil-soluble or dispersible products are thereby obtained. These quaternized ammonium salts when paired with a pharmaceutically acceptable anion can also serve as pharmaceutically acceptable salts.
Examples of acids which may be employed to form pharmaceutically acceptable acid addition salts include such inorganic acids as hydrochloric acid, sulfuric acid and phosphoric acid and such organic acids as oxalic acid, maleic acid, methanesulfonic acid, succinic acid and citric acid. Basic addition salts can be prepared in situ during the final isolation and purification of the compounds of formula (I), or separately by reacting carboxylic acid moieties with a suitable base such as the hydroxide, carbonate or bicarbonate of a pharmaceutically acceptable metal cation or with ammonia, or an organic primary, secondary or tertiary amine. Counterions for pharmaceutically acceptable salts include, but are not limited to, cations based on the alkali and alkaline earth metals, such as sodium, lithium, potassium, calcium, magnesium, aluminum salts and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. Other representative organic amines useful for the formation of base addition salts include diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine and the like.
As used herein, the term "pharmaceutically acceptable ester" refers to esters, which hydrolyze in vivo and include those that break down readily in the human body to leave the parent compound or a salt thereof. Suitable ester groups include, for example, those derived from pharmaceutically acceptable aliphatic carboxylic acids, particularly alkanoic, alkenoic, cycloalkanoic and alkanedioic acids, in which each alkyl or alkenyl moiety advantageously has not more than 6 carbon atoms. Examples of particular pharmaceutically acceptable esters include formates, acetates, propionates, maleates, lactates, hydroxyacetates, butyrates, acrylates and ethylsuccinates.
The term "pharmaceutically acceptable prodrugs" as used herein refers to those prodrugs of the compounds of the present invention which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the zwitterionic forms, where possible, of the compounds of the invention. The term "prodrug" refers to compounds that are rapidly transformed in vivo to yield the parent compound of the above formula, for example by hydrolysis in blood. A thorough discussion is provided in T. Higuchi and V. Stella, PRO-DRUGS AS NOVEL DELIVERY SYSTEMS, Vol. 14 of the A.C.S. Symposium Series, and in Edward B. Roche, ed., BIOREVERSIBLE CARRIERS IN DRUG DESIGN, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference.
Any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds, lsotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen,
2 3H 11 13 carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as H, , C, C, 14C, 15N, 18F 31P, 32P, 35S, 36C1, 125I respectively. The invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3H and 14C, or those into which non-radioactive isotopes, such as 2H and 13C are present. Such isotopically labeled compounds are useful in metabolic studies (with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques, such as positron emission tomography (PET) or single- photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18F or labeled compound may be particularly desirable for PET or SPECT studies. Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.
Further, substitution with heavier isotopes, particularly deuterium (i.e., 2H or D) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements or an improvement in therapeutic index. It is understood that deuterium in this context is regarded as a substituent of a compound of the formula (I). The concentration of such a heavier isotope, specifically deuterium, may be defined by the isotopic enrichment factor. The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope. If a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90%) deuterium incorporation), at least 6333.3 (95%> deuterium incorporation), at least 6466.7 (97%) deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5%> deuterium incorporation).
Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D20, d6- acetone, d6-DMSO.
Compounds of the invention, i.e. compounds of formula (I) that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of forming co-crystals with suitable co-crystal formers. These co-crystals may be prepared from compounds of formula (I) by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co-melting, or contacting in solution compounds of formula (I) with the co-crystal former under crystallization conditions and isolating co-crystals thereby formed. Suitable co-crystal formers include those described in WO 2004/078163. Hence the invention further provides co-crystals comprising a compound of formula (I).
The following enumerated embodiments are representative of certain aspects of the invention:
1. A compound of Formula (I) :
Figure imgf000016_0001
4 alkyl)-pyrazole ring;
W is H or NH2;
Z is CH or N;
n is 1 or 2;
R1 is H, OH or OMe;
R2 is H or Me,
provided that when R1 is H, R2 is also H;
R3 is H or Ci-C4 alkyl;
R4 is selected from R*, -OR*, -OCH2CH2OR*, -CH2OR*, and a 4-6 membered cyclic ether optionally substituted with OH, OMe, or F, wherein each R* is independently C1-C4 alkyl,
provided that R4 is not -OMe when Z is N; or a pharmaceutically acceptable salt thereof. In preferred embodiments, W is
NH2.
2. The compound of embodiment 1, wherein R3 is H.
3. The compound of embodiment 1, wherein R3 is Me.
4. The compound of any of embodiments 1-3, wherein R2 is H.
5. The compound of any of embodiments 1-3, wherein R2 is Me.
6. The compound of any of embodiments 1-3, wherein R1 is H.
7. The compound of any of embodiments 1-5, wherein R1 is OH.
8. The compound of any of the preceding embodiments, wherein Z is CH.
9. The compound of any of the preceding embodiments, which is of the formula IA:
Figure imgf000017_0001
The compound of any one of embodiments 1-7, wherein Z is N.
The compound of embodiment 10, which is of the formula IB:
Figure imgf000018_0001
12. The compound of any of the preceding embodiments, wherein n is 1.
The compound of embodiment 10 or 11, wherein
Figure imgf000018_0002
The compound of embodiment 9 or 11 , wherein represents
Figure imgf000018_0003
pyrazole of the formula where R is methyl, ethyl or isopropyl.
15. The compound of embodiment 14, wherein RN is methyl.
Figure imgf000019_0001
The compound of any one of embodiments 1-12, wherein represents
Figure imgf000019_0002
a pyridine of the formula
17. The compound of any of the preceding embodiments, wherein R is a
Figure imgf000019_0003
tetrahydropyranyl group of the formula:
wherein RT is H, OH, OMe, or F. In certain embodiments, it is selected from H, OH and F.
18. The compound of any of embodiments 1-16, wherein R is an oxetanyl group of
Figure imgf000019_0004
wherein R is H, OH, OMe, or F. In certain embodiments, it is selected from H, OH and F.
19. The compound of any of embodiments 1-16, wherein R is OMe, OEt, or OPr.
20. The compound of any of embodiments 1-16, wherein R4 is -CH2OMe or -CH2OEt 21. The compound of embodiment 1, which is selected from the compounds in Table 1. Each compound and any subset of the compounds in Table 1 represent preferred embodiments of the invention. 22. A pharmaceutical composition comprising a compound of any of the preceding embodiments and at least one pharmaceutically acceptable excipient.
23. The pharmaceutical composition of embodiment 22, further comprising an additional therapeutic agent.
24. The pharmaceutical composition of embodiment 23, wherein the additional therapeutic agent is selected from irinotecan, topotecan, gemcitabine, 5-fluorouracil, cytarabine, daunorubicin, PI3 Kinase inhibitors, mTOR inhibitors, DNA synthesis inhibitors, leucovorin, carboplatin, cisplatin, taxanes, tezacitabine, cyclophosphamide, vinca alkaloids, imatinib, anthracyclines, rituximab, and trastuzumab.
25. A method to treat a condition caused or exacerbated by excessive Pirn kinase activity, wherein the method comprises administering to a subject in need thereof an effective amount of a compound of any of embodiments 1-21. In some embodiments, the subject has been diagnosed with a condition caused by Pirn kinase.
26. The method of embodiment 25, wherein the condition is a cancer.
27. The method of embodiment 26, wherein the cancer is selected from carcinoma of the lungs, pancreas, thyroid, ovary, bladder, breast, prostate, or colon, melanoma, myeloid leukemia, multiple myeloma, erythroleukemia, villous colon adenoma, and osteosarcoma; or the autoimmune disorder is selected from Crohn's disease, inflammatory bowel disease, rheumatoid arthritis, and chronic inflammatory diseases. 28. A compound according to any of embodiments 1-21 for use in therapy.
29. Use of a compound according to any one of embodiments 1-21 for the preparation of a medicament. The medicament may be to treat a cancer, such as a cancer selected from carcinoma of the lungs, pancreas, thyroid, ovaries, bladder, breast, prostate or colon, melanoma, myeloid leukemia, multiple myeloma, erythro leukemia, villous colon adenoma, and osteosarcoma. 31. The compound according to embodiment 30, wherein the cancer is selected from carcinoma of the lungs, pancreas, thyroid, ovaries, bladder, breast, prostate or colon, melanoma, myeloid leukemia, multiple myeloma, erythro leukemia, villous colon adenoma, and osteosarcoma. 32. The compound of embodiment 31 , wherein the condition is an autoimmune disorder.
33. A method of treating a disease or condition mediated by PIM kinase, comprising administering to a subject in need thereof a therapeutically effective amount of a compound according to any one of embodiments 1-25, or a pharmaceutically acceptable salt thereof.
34. The method of embodiment 33, wherein the disease is selected from carcinoma of the lungs, pancreas, thyroid, ovaries, bladder, breast, prostate or colon, melanoma, myeloid leukemia, multiple myeloma, erythro leukemia, villous colon adenoma, and osteosarcoma; or the disease is an autoimmune disorder.
35. The method of embodiment 34, wherein the disease is an autoimmune disorder. 36. The method of embodiment 35, wherein the autoimmune disorder is selected from Crohn's disease, inflammatory bowel disease, rheumatoid arthritis, and chronic inflammatory diseases.
In the compounds of the invention, Ring A can be pyridyl or certain N- alkylpyrazoles; preferred embodiments of Ring A include:
Figure imgf000022_0001
where R is methyl, ethyl or isopropyl, especially Me;
Figure imgf000022_0002
where (Z) indicates the position of Ring A that is attached to the ring containing Z.
In the compounds of Formula I, Z can be CH or N; when Z is N, preferred embodiments of the ring containing Z include:
Figure imgf000022_0003
where R1 is H or OH; R2 is H or Me, provided that when R1 is H, R2 is also H; and R3 is H, Me, Et or iPr.
Alternatively, when Z is CH, preferred embodiments of the ring containing Z include:
Figure imgf000022_0004
where R1 is H or OH; R2 is H or Me, provided that when R1 is H, R2 is also H; and R3 is Me, Et or iPr. In the compounds where Z is CH, n is preferably 1, while n is often 2 when Z is N.
The substituent R4 can influence in vivo properties such as metabolism and clearance rates as well as drug interactions, particularly in combination with the above embodiments of the ring containing Z. Some of the preferred embodiments of R4 in compounds of the invention include: Me, OMe, iPr, -OiPr, -CH2OEt, -CH2OiPr, - OCH2CH2OMe, and groups comprising optionally substituted cyclic ethers, including these:
Figure imgf000023_0001
In compounds of the invention, R1 is preferably H or OH, and R2 is preferably H. R3 is typically Me; or R3 can be H, especially when R1 is H and/or n is 2.
These compounds may be used in racemic form, or the individual enantiomers may be used, or mixtures of the enantiomers may be used. Each enantiomer can be used, and preferably the compound to be used is the enantiomer that has greater activity as a Pirn inhibitor.
For purposes of the present invention, a therapeutically effective dose will generally be a total daily dose administered to a host in single or divided doses may be in amounts, for example, of from 0.001 to 1000 mg/kg body weight daily, typically 0.01 to 100 mg/kg per day, and more preferred from 0.1 to 30 mg/kg body weight daily. Generally, daily dosage amounts of 1 to 4000 mg, or from 5 to 3000, or from 10 to 2000 mg, or from 100 to 2000 mg are anticipated for human subjects. Dosage unit compositions may contain such amounts of submultiples thereof to make up the daily dose. The compounds of the present invention may be administered orally, parenterally, sublingually, by aerosolization or inhalation spray, rectally, or topically in dosage unit formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles as desired. Topical administration may also involve the use of transdermal administration such as transdermal patches or ionophoresis devices. The term parenteral as used herein includes subcutaneous injections, intravenous, intramuscular, intrasternal injection, or infusion techniques. In preferred embodiments, the compound or composition of the invention is administered orally.
Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-propanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or di-glycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
Suppositories for rectal administration of the drug can be prepared by mixing the drug with a suitable nonirritating excipient such as cocoa butter and polyethylene glycols, which are solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum and release the drug.
Solid dosage forms for oral administration may include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound may be admixed with at least one inert diluent such as sucrose lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., lubricating agents such as magnesium stearate. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, cyclodextrins, and sweetening, flavoring, and perfuming agents.
The compounds of the present invention can also be administered in the form of liposomes. As is known in the art, liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used. The present compositions in liposome form can contain, in addition to a compound of the present invention, stabilizers, preservatives, excipients, and the like. The preferred lipids are the phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes are known in the art. See, for example, Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.W., p. 33 et seq. (1976).
While the compounds of the invention can be administered as the sole active pharmaceutical agent, they can also be used in combination with one or more other agents used in the treatment of cancer. The compounds of the present invention are also useful in combination with known therapeutic agents and anti-cancer agents, and combinations of the presently disclosed compounds with other anti-cancer or chemotherapeutic agents are within the scope of the invention. Examples of such agents can be found in Cancer Principles and Practice of Oncology, V. T. Devita and S. Hellman (editors), 6th edition (Feb. 15, 2001), Lippincott Williams & Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved. Such anti-cancer agents include, but are not limited to, the following: MEK inhibitors, estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic/cytostatic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors and other angiogenesis inhibitors, inhibitors of cell proliferation and survival signaling, apoptosis inducing agents and agents that interfere with cell cycle checkpoints. The compounds of the invention are also useful when co- administered with radiation therapy.
Therefore, in one embodiment of the invention, the compounds of the invention are also used in combination with known therapeutic or anticancer agents including, for example, estrogen receptor modulators, androgen receptor modulators, retinoid receptor modulators, cytotoxic agents, antiproliferative agents, prenyl-protein transferase inhibitors, HMG-CoA reductase inhibitors, HIV protease inhibitors, reverse transcriptase inhibitors, and other angiogenesis inhibitors.
In certain presently preferred embodiments of the invention, representative therapeutic agents useful in combination with the compounds of the invention for the treatment of cancer include, for example, MEK inhibitors, irinotecan, topotecan, gemcitabine, 5-fluorouracil, cytarabine, daunorubicin, PI3 Kinase inhibitors, mTOR inhibitors, DNA synthesis inhibitors, leucovorin carboplatin, cisplatin, taxanes, tezacitabine, cyclophosphamide, vinca alkaloids, imatinib (Gleevec), anthracyclines, rituximab, trastuzumab, Revlimid, Velcade, dexamethasone, daunorubicin, cytaribine, clofarabine, Mylotarg, lenalidomide, bortezomib, as well as other cancer
chemotherapeutic agents including targeted therapeutics.
The above compounds to be employed in combination with the compounds of the invention will be used in therapeutic amounts as indicated in the Physicians' Desk Reference (PDR) 47th Edition (1993), which is incorporated herein by reference, or such therapeutically useful amounts as would be known to one of ordinary skill in the art, or provided in prescribing materials such as a drug label for the additional therapeutic agent.
The compounds of the invention and the other anticancer agents can be administered at the recommended maximum clinical dosage or at lower doses. Dosage levels of the active compounds in the compositions of the invention may be varied so as to obtain a desired therapeutic response depending on the route of administration, severity of the disease and the response of the patient. The combination can be administered as separate compositions or as a single dosage form containing both agents. When administered as a combination, the therapeutic agents can be formulated as separate compositions, which are given at the same time or different times, or the therapeutic agents, can be given as a single composition.
In one embodiment, the invention provides a method of inhibiting Piml, Pim2 or Pim3 in a human or animal subject. The method includes administering an effective amount of a compound, or a pharmaceutically acceptable salt thereof, of any of the embodiments of compounds of Formula I or II to a subject in need thereof.
The present invention will be understood more readily by reference to the following examples, which are provided by way of illustration and are not intended to be limiting of the present invention. Synthetic Methods
The compounds of the invention can be obtained through procedures known to those skilled in the art. As shown in Scheme 1, 5-alkyl, 4-hydroxy, 3-aminopiperidines can be prepared and modified to yield 5-alkyl, 4-substituted, 3-aminopiperidinyl pyridine amides VI as follows. Reaction of Garner's aldehyde with (R)-4-benzyl-3- propionyloxazolidin-2-one followed by TBS protection of the resulting alcohol affords compound I. Reduction of the oxazolidinone followed by introduction of the azide group yields intermediate II. Deprotection under acidic conditions reveals the corresponding amino alcohol, which upon protection with the Boc group followed by mesylation of the primary alcohol yields intermediate III. Reduction of the azide affords formation of the piperidine which is subsequently reacted with 4-chloro-3-nitropyridine and following nitro reduction pyridyl aniline IV is obtained. Aniline IV can be coupled with heterocyclic acids, which after silyl group and N-Boc deprotection can afford target amides VI.
Scheme 1
Figure imgf000028_0001
Figure imgf000028_0002
In Scheme 2, synthetic methods to prepare certain aminocyclohexylpyridyl amides
X are depicted. Methyl cyclohexanedione can be converted via the monotriflate to the corresponding cyclohexenoneboronate ester which can undergo palladium mediated carbon bond formation with 4-chloro, 3-nitro pyridine to yield nitropyridine substituted cyclohexenone VII. Ketone reduction followed by dehydration yields a cyclohexadiene which upon epoxidation (via bromohydrin formation and HBr elimination), azide epoxide opening, azide reduction and amine Boc protection yields cyclohexenyl Boc amino alcohol nitro pyridyl compound VIII. Nitro pyridyl VIII can be converted to the trans protected amino hydroxy aniline IX by alcohol protection and alkene and nitro reduction. As described above in Scheme 1 for the preparation of substituted piperidine compounds of the invention, upon amide coupling of the cyclohexyl pyridyl anilines IX to heterocyclic acids and subsequent hydroxyl and amine deprotection, substituted cyclohexyl compounds of the invention X can be prepared. Scheme 2
Figure imgf000029_0001
Alternatively, as shown in Scheme 3, cyclohexanediones can be converted via monotriflates to the corresponding cyclohexenoneboronate esters which can undergo palladium mediated carbon bond formation with 4-chloro, 3-nitro pyridine to yield nitropyridine substituted cyclohexenones XI. Reduction of the enone functionality can yield a cyclohexenol XII, which can undergo Mitsunobu reaction with phthalimide to yield a protected aminocyclohexene XIII. Following nitro and alkene reduction, phthalimide protected aminocyclohexyl pyridyl aniline Va can undergo amide coupling and deprotection, to yield aminocyclohexane amides XIV. The corresponding Boc protected aminocyclohexane pyridyl aniline XV can also be prepared from cyclohexenol XII in the following manner: alcohol protection, alkene and nitro reduction, pyridyl amine Cbz protection, silyl ether deprotection, Dess-Martin oxidation to the cyclohexanone, reductive amination with benzylamine, Cbz and Bn deprotection and primary aliphatic amine Boc protection. Penultimate amide products which contain a heteroaromatic bromo functionality can be further modified by standard modifications to introduce substituted aryls, alkyls and heteroaryls on place of !¾. For example, if R2 is Br, by reaction with boronic acids or organometallic reagents, or conversion to the corresponding boronate ester and reaction with aryl/heteroaryl halides or triflates, a variety of R2 replacements are possible. Scheme 3
Figure imgf000030_0001
XV N HCI/dioxane XVI
Referring to the examples that follow, compounds of the preferred embodiments were synthesized using the methods described herein, or other methods, which are known in the art.
The compounds and/or intermediates were characterized by high performance liquid chromatography (HPLC) using a Waters Millenium chromatography system with a 2695 Separation Module (Milford, MA). The analytical columns were reversed phase Phenomenex Luna CI 8 -5 μ, 4.6 x 50 mm, from Alltech (Deerfield, IL). A gradient elution was used (flow 2.5 mL/min), typically starting with 5% acetonitrile/95% water and progressing to 100% acetonitrile over a period of 10 minutes. All solvents contained 0.1%) trif uoroacetic acid (TFA). Compounds were detected by ultraviolet light (UV) absorption at either 220 or 254 nm. HPLC solvents were from Burdick and Jackson (Muskegan, MI), or Fisher Scientific (Pittsburgh, PA).
In some instances, purity was assessed by thin layer chromatography (TLC) using glass or plastic backed silica gel plates, such as, for example, Baker-Flex Silica Gel 1B2-F flexible sheets. TLC results were readily detected visually under ultraviolet light, or by employing well-known iodine vapor and other various staining techniques. Mass spectrometric analysis was performed on one of three LCMS instruments: a Waters System (Alliance HT HPLC and a Micromass ZQ mass spectrometer; Column: Eclipse XDB-C18, 2.1 x 50 mm; gradient: 5-95% (or 35-95%, or 65-95% or 95-95%) acetonitrile in water with 0.05% TFA over a 4 min period; flow rate 0.8 mL/min; molecular weight range 200-1500; cone Voltage 20 V; column temperature 40°C), another Waters System (ACQUITY UPLC system and a ZQ 2000 system; Column: ACQUITY UPLC HSS-C18, 1.8um, 2.1 x 50mm; gradient: 5-95% (or 35-95%, or 65-95% or 95-95%) acetonitrile in water with 0.05% TFA over a 1.3 min period; flow rate 1.2 mL/min; molecular weight range 150-850; cone Voltage 20 V; column temperature 50°C) or a Hewlett Packard System (Series 1100 HPLC; Column: Eclipse XDB-C18, 2.1 x 50 mm; gradient: 5-95% acetonitrile in water with 0.05% TFA over a 4 min period; flow rate 0.8 mL/min; molecular weight range 150-850; cone Voltage 50 V; column temperature 30°C). All masses were reported as those of the protonated parent ions.
Nuclear magnetic resonance (NMR) analysis was performed on some of the compounds with a Varian 400 MHz NMR (Palo Alto, CA). The spectral reference was either TMS or the known chemical shift of the solvent.
Preparative separations are carried out using a Flash 40 chromatography system and KP-Sil, 60A (Biotage, Charlottesville, VA), or by flash column chromatography using silica gel (230-400 mesh) packing material on ISCO or Analogix purification systems, or by HPLC using a Waters 2767 Sample Manager, C-18 reversed phase column, 30X50 mm, flow 75 mL/min. Typical solvents employed for the Flash 40 Biotage, ISCO or Analogixsystem for silica gel column chromatography are dichloromethane, methanol, ethyl acetate, hexane, n-heptanes, acetone, aqueous ammonia (or ammonium hydroxide), and triethyl amine. Typical solvents employed for the reverse phase HPLC are varying concentrations of acetonitrile and water with 0.1% trifluoroacetic acid.
Chiral separation of enantiomeric mixtures was performed by the following analytical and preparative general methods:
Chiral SFC-Analytical Method: Chiral compounds were separated on a Waters
Supercritical Fluid Chromatography (SFC). The separation used a Chiralpak AD (AS, OD, OJ, IC or IA) 4.6x100mm column at 40C temperature at a flow rate of 5 mL/min using an isocratic method. The mobile phase was 15% MeOH (or EtOH or IPA or with 0.1% Diethyl amine): 85% C02. The detection wavelength was 220 nm (or 250nm or Diode Array).
Chiral SFC-Purification Method: Chiral compounds were separated on a Waters Supercritical Fluid Chromatography (SFC). The separation used a Chiralpak AD (AS, OD, OJ, IC or IA) 21x250mm column at 40C temperature at a flow rate of 100 mL/min using an isocratic method. The mobile phase was 15% MeOH (or EtOH or IPA or with 0.1% Diethyl amine): 85% C02. The detection wavelength was 220 nm (or 250nm or Diode Array).
Chiral HPLC-Analytical Method: Chiral compounds were separated on a Waters 2695 HPLC system. The separation used a Chiralpak AD (AS, OD, OJ, IC or IA) 4.6x100mm column at room temperature at a flow rate of 1 mL/min using an isocratic method. The mobile phase was 15% EtOH (or IPA or with 0.1% Diethyl amine): 85% Heptane. The detection wavelength was 220 nm (or 250nm or Diode Array).
Chiral HPLC-Purification Method: Chiral compounds were separated on a Waters 2767 HPLC system. The separation used a Chiralpak AD (AS, OD, OJ, IC or IA) 21x250mm column at room temperature at a flow rate of 20 (or 10 -15) mL/min using an isocratic method. The mobile phase was 15% EtOH (or IPA or with 0.1% Diethyl amine): 85% Heptane. The detection wavelength was 220 nm (or 250nm or Diode Array).
It should be understood that the organic compounds according to the preferred embodiments may exhibit the phenomenon of tautomerism. As the chemical structures within this specification can only represent one of the possible tautomeric forms, it should be understood that the preferred embodiments encompasses any tautomeric form of the drawn structure.
It is understood that the invention is not limited to the embodiments set forth herein for illustration, but embraces all such forms thereof as come within the scope of the above disclosure.
The examples below as well as throughout the application, the following abbreviations have the following meanings. If not defined, the terms have their generally accepted meanings. ABBREVIATIONS
DAST (diethylamino)sulfurtrifluoride
DCM Dichloromethane
DIAD diisopropylazodicarboxylate
DIEA diisopropylethylamine
DMA Dimethylacetamide
DMAP 4-dimethylaminopyridine
DME 1 ,2-dimethoxyethane
DMF N,N-dimethylformamide
DPPF 1 , 1 '-bis(diphenylphosphino)ferrocene
EDC 1 -(3 -Dimethylaminopropyl)-3 -ethylcarbodiimide hydrochloride
EtOAc ethyl acetate
EtOH Ethanol
HOAT Hydroxyazabenzotriazole
K2C03 Potassium carbonate
MeCN Acetonitrile
MgS04 Magnesium sulfate
MeOH Methanol
Na2C03 sodium carbonate
NaCl Sodium chloride
NaHC03 sodium bicarbonate
NBS N-bromosuccinimide
NMP N-methyl-2-pyrrolidone
Pd2(dba)3 Tris(dibenzylideneacetone)dipalladium(0)
Pd(PPh3)4 Tetrakis(triphenylphospine)palladium(0)
Pd(dppf)Cl2- Dichloro-( 1 ,2-bis(diphenylphosphino)ethan)- DCM Palladium(II) - dichloromothethane adduct
RT or rt room temperature
TBDMSC1 tert-butyldimethylsilylchloride ABBREVIATIONS
TEA Triethylamine
THF tetrahydrofuran
EXAMPLES
Synthesis of (R)-tert-butyl 4-((lR,2R)-3-((R)-4-benzyl-2-oxooxazolidin-3-yl)-l-hydroxy- 2-methyl-3-oxopropyl)-2,2-dimethyloxazolidine-3-carboxylate
Figure imgf000034_0001
To a solution of (R)-4-benzyl-3-propionyloxazolidin-2-one (1.0 equiv.) in DCM (0.13 M) was added TiCl4 (1.0 equiv.) at -40 °C. The mixture was stirred at -40 °C for 10 min (yellow suspension), then DIPEA (2.5 equiv.) was added (dark red solution) and stirred at 0 °C for 20 min. (R)-tert-butyl 4-formyl-2,2-dimethyloxazolidine-3-carboxylate (1.0 equiv.) in DCM (0.5 M) was then added dropwise and the resulting mixture was stirred for 1.5 hours. The reaction was quenched by the addition of aqueous ammonium chloride and the mixture was extracted with ethyl acetate. The organic phase was separated, washed with brine, dried with magnesium sulfate, filtered, and concentrated. The residue was purified via column chromatography eluting with ethyl acetate and hexanes (1 :4) to give (R)-tert-butyl 4-((lR,2R)-3-((R)-4-benzyl-2-oxooxazolidin-3-yl)-l- hydroxy-2-methyl-3-oxopropyl)-2,2-dimethyloxazolidine-3-carboxylate as the major product (5:2) in 58% yield. LC/MS = 363.3 (M+H-Boc), Rt = 1.09 min. Synthesis of flO-tert-butyl 4-(Y lR,2R -3-((R -4-benzyl-2-oxooxazolidin-3-vn-l-(tert- butyldimethylsilyloxy)-2-methyl-3-oxopropyl)-2,2-dimethyloxazolidine-3-carboxylate
Figure imgf000035_0001
To a solution of (R)-tert-butyl 4-((lR,2R)-3-((R)-4-benzyl-2-oxooxazolidin-3-yl)- l-hydroxy-2-methyl-3-oxopropyl)-2,2-dimethyloxazolidine-3-carboxylate (1.0 equiv.) and lutidine (1.8 equiv.) in DCM (0AM) was added TBSOTf (1.4 equiv.) at -40 °C. The reaction mixture was stirred at -40 °C for 2 hours. The solution was diluted with ethyl acetate and washed with sat. NaHC03, sat. NaCl, dried with magnesium sulfate, filtered, and concentrated. The residue was purified by silica gel column chromatography eluting with ethyl acetate and hexanes (1 :4) to give (R)-tert-butyl 4-((lR,2R)-3-((R)-4-benzyl-2- oxooxazolidin-3-yl)-l-(tert-butyldimethylsilyloxy)-2-methyl-3-oxopropyl)-2,2- dimethyloxazolidine-3-carboxylate as the major product (5 :2) in 83% yield. LC/MS = 577.3 (M+H), Rt = 1.33 min (Frac 65%-95% method).
Synthesis of (RV tert-butyl 4-(( 1 R.2SV 1 -(tert-butyldimethylsiryloxyV3 -hydroxy-2- methylpropyl)-2,2-dimethyloxazolidine-3-carboxylate
Figure imgf000035_0002
To a solution of (R)-tert-butyl 4-((lR,2R)-3-((R)-4-benzyl-2-oxooxazolidin-3-yl)- l-(tert-butyldimethylsilyloxy)-2-methyl-3-oxopropyl)-2,2-dimethyloxazolidine-3- carboxylate (1.0 equiv.) and ethanol (3.0 equiv.) in THF (0.09 M) was added LiBH4 (3.0 equiv.) at -30 °C. The reaction mixture was allowed to warm up to 0 °C and stirred at that temperature for 3 hours. The solution was then diluted with diethyl ether and IN NaOH was added. The resulting mixture was extracted with ethyl acetate, the organic layer was separated, washed with sat. NaCl, dried over magnesium sulfate, filtered, and concentrated. The residue was purified via silica gel column chromatography eluting with ethyl acetate and hexanes (1 :4) to give (R)-tert-butyl 4-((lR,2S)-l-(tert- butyldimethylsilyloxy)-3-hydroxy-2-methylpropyl)-2,2-dimethyloxazolidine-3- carboxylate as the major product (5:2 ratio) in 71% yield. LC/MS = 304.3 (M+H-Boc), Rt = 0.95 min (Frac 65%-95% method). Synthesis of (RVtert-butyl 4-(Y lR,2S -3-azido-l-(tert-butyldimethylsilyloxy -
2-methylpropyl)-2,2-dimethyloxazolidine-3-carboxylate
Figure imgf000036_0001
To a solution of (R)-tert-butyl 4-((lR,2S)-l-(tert-butyldimethylsilyloxy)-3- hydroxy-2-methylpropyl)-2,2-dimethyloxazolidine-3-carboxylate (1.0 equiv.), DIAD (2.0 equiv.), and PPh3 (2.0 equiv.) in THF (0.18 M) was added DPPA (2.0 equiv., 1M solution in THF). The reaction mixture was stirred at room temperature overnight. Upon removal of the volatiles under vacuo, the residue was purified by silica gel column chromatography eluting with ethyl acetate and hexanes (1 :6) to give (R)-tert-butyl 4- ((lR,2S)-3-azido-l-(tert-butyldimethylsilyloxy)-2-methylpropyl)-2,2-dimethyloxazol- idine-3-carboxylate as the major product (5:2) in 86% yield. LC/MS = 329.3 (M+H- Boc), Rt = 1.40 min (Frac 65%-95% method).
Synthesis of tert-butyl (2R,3R,4S)-5-azido-3-(tert-butyldimethylsilyloxy)- l-hydroxy-4-methylpentan-2-ylcarbamate
Figure imgf000036_0002
To a solution of (R)-tert-butyl 4-((lR,2S)-3-azido-l-(tert-butyldimethylsilyloxy)- 2-methylpropyl)-2,2-dimethyloxazolidine-3-carboxylate (1.0 equiv.) in EtOH (0.1 M) was added PPTS (1.3 equiv.) and the mixture was refluxed for 2 days. The volatiles were removed under vacuo, the residue was dissolved in DCM (0.1 M) and DIEA (1.5 equiv.) and Boc20 (1.0 equiv.) were added to the reaction mixture. The solution was stirred for 3 hours at room temperature. The solvents were removed under reduced pressure and the residue was diluted with ethyl acetate, washed with water, aqueous NaHS04, aqueous NaHC03, sat. NaCl, the organic phase was dried with magnesium sulfate, filtered, and concentrated. The residue was purified via silica gel column chromatography eluting with ethyl acetate and hexanes (1 :3) to give tert-butyl (2R,3R,4S)-5-azido-3-(tert- butyldimethylsilyloxy)-l-hydroxy-4-methylpentan-2-ylcarbamate as the major isomer (5 :2) in 70% yield. LC/MS = 289.3 (M+H-Boc), Rt = 0.76 min (Frac 65%-95% method).
Synthesis of (2R,3R,4S)-5-azido-2-(tert-butoxycarbonylamino)-3-(tert- butyldimethylsilyloxy)-4-methylpentyl methanesulfonate
Figure imgf000037_0001
To a solution of tert-butyl (2R,3R,4S)-5-azido-3-(tert-butyldimethylsilyloxy)-l- hydroxy-4-methylpentan-2-ylcarbamate (1.0 equiv.) in pyridine (0.2 M) was added MsCl (1.3 equiv.) followed by DMAP (catalytic amount) at 0 °C. The mixture was stirred at that temperature for 1 hour. The solution was diluted with ether and ethyl acetate (4:1), washed with aq. NaHSC"4, sat. NaHC03, brine, dried over magnesium sulfate, filtered, and concentrated. The residue was purified by silica gel column chromatography eluting with ethyl acetate and hexanes (1 :3) to give (2R,3R,4S)-5-azido-2-(tert- butoxycarbonylamino)-3-(tert-butyldimethylsilyloxy)-4-methylpentyl methanesulfonate as the major isomer (5:2) in 90% yield. LC/MS = 367.3 (M+H-Boc), Rt = 0.81 min (Frac 65%-95% method).
Synthesis of tert-butyl (3R.4R.5SV4-(tert-butyldimethylsilyloxyV
5 -methylpiperidin-3 -ylcarbamate
Figure imgf000037_0002
A solution of (2R,3R,4S)-5-azido-2-(tert-butoxycarbonylamino)-3-(tert- butyldimethylsilyloxy)-4-methylpentyl methanesulfonate in MeOH (0.09 M) was degassed with nitrogen for 20 min. DIEA (2.5 equiv.) was added, followed by 10%> Pd/C (0.1 equiv.). The reaction mixture was stirred under a hydrogen balloon for 2 hours. The solution was filtered and the filtrate was concentrated under vacuo to afford tert-butyl (3R,4R,5S)-4-(tert-butyldimethylsilyloxy)-5-methylpiperidin-3-ylcarbamate as the major isomer (5:2) in >99% yield. LC/MS = 345.2 (M+H-Boc), Rt = 0.95 and 0.99 min.
Synthesis of tert-butyl (3R.4R.5 S -4-(tert-butyldimethylsilyloxy -5-methyl- 1 -(3- nitropyridin-4-yl)piperidin-3-ylcarbamate
BocHN
Figure imgf000038_0001
To a solution of tert-butyl (3R,4R,5S)-4-(tert-butyldimethylsilyloxy)-5- methylpiperidin-3-ylcarbamate (1.0 equiv.) in i-PrOH (0.09 M) was added DIEA (2.5 equiv.) and 4-chloro-3-nitropyridine (1.5 equiv.). The reaction mixture was stirred at 60 °C for 2 hours. The volatiles were removed under vacuo, the residue was diluted with ethyl acetate and washed with sat. NaCl. The organic phase was dried with magnesium sulfate, filtered, and concentrated. The crude material was purified by silica gel column chromatography eluting with ethyl acetate and hexanes (1 :2) to give tert-butyl (3R,4R,5S)-4-(tert-butyldimethylsilyloxy)-5-methyl-l-(3-nitropyridin-4-yl)piperidin-3- ylcarbamate in 76% yield. LC/MS = 467.3 (M+H), Rt = 1.09 min.
Synthesis of tert-butyl (3R,4R,5S)-l-(3-aminopyridin-4-yl)-4-(tert- butyldimethylsilyloxy)-5 -methylpiperidin-3 -ylcarbamate
OTBS
BocHN
Figure imgf000038_0002
A solution of tert-butyl (3R,4R,5S)-4-(tert-butyldimethylsilyloxy)-5-methyl-l-(3- nitropyridin-4-yl)piperidin-3-ylcarbamate (1.0 equiv.) in MeOH (0.05 M) was degassed with nitrogen for 20 min. 10% Pd/C (0.2 equiv.) was added to the mixture and the solution was stirred under a hydrogen balloon for 3 hours. The reaction was filtered and the filtrate was concentrated under reduced pressure to give tert-butyl (3R,4R,5S)-l-(3- aminopyridin-4-yl)-4-(tert-butyldimethylsilyloxy)-5 -methylpiperidin-3 -ylcarbamate as the desired product in 94% yield. LC/MS = 437.4 (M+H), Rt = 1.08 min. 1H-NMR (300
MHz, CDCls): δ 8.01 (s, 1H), 7.95 (d, J = 6.0 Hz, 1H), 6.76 (d, J = 6.0 Hz, 1H), 4.44 (b s, 1H), 3.74 (br s, 2H), 3.59-3.55 (m, 1H), 3.25-3.13 (m, 2H), 2.47-2.35 (m, 2H), 1.89 (b s, 2H), 1.44 (s, 9H), 1.04 (d, J = 6.0 , 3H), 0.92 (s, 9H), 0.13 (d, J = 9.0, 6H).
Synthesis of 5-methyl-3-oxocyclohex- 1 -enyltrifluoromethanesulfonate
Figure imgf000039_0001
To a solution of 5-methylcyclohexane-l,3-dione (1.0 equiv.) in DCM (0.5M) was added Na2C03 (1.1 equiv.) and cooled to 0 °C. Added Tf20 (1.0 equiv.) in DCM (5.0 M) dropwise over 1 hr at 0°C under a nitrogen atmosphere. Upon addition, the reaction was stirred for 1 hr at room temperature (dark red solution). The solution was filtered and the filtrate was quenched by careful addition of saturated NaHC03 with vigorous stirring until pH=7. The solution was transferred to a separatory funnel and the layers were separated. The organic layer was washed with brine, dried with Na2S04, filtered, concentrated under vacuo and dried under high vacuum for 15 min to yield 5-methyl-3- oxocyclohex-l-enyl trifluoromethanesulfonate as light yellow oil in 78% yield. The triflate decomposes upon storage and should be used immediately for the next reaction.
LC/MS=259.1/300.1 (M+H and M+CH3CN); Rt = 0.86 min, LC = 3.84 min. 1H-NMR (400 MHz, CDC13) δ ppm: 6.05 (s, 1H), 2.70 (dd, J=17.2, 4.3, 1H), 2.53 (dd, J=16.6, 3.7, 1H), 2.48-2.31 (m, 2H), 2.16 (dd, J=16.4, 11.7, 1H), 1.16 (d, J=5.9, 3H). Synthesis of 5-methyl-3-(4 A5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)cyclohex-2-enone
Figure imgf000040_0001
To a solution of 5-methyl-3-oxocyclohex-l-enyl trifluoromethanesulfonate (1.0 equiv.) in degassed dioxane (0.7 M) was added bis(pinacolato)diboron (2.0 equiv.), KOAc (3.0 equiv.), and Pd(dppf)Cl2-DCM (0.03 equiv.). The reaction was heated to 80 °C for 10 h (initial heating at large scale results in exothermic formation of an orange foam on top of the solution, the heating bath should be removed until the foam retracts, reheating to 80 °C at this point appears to be fine), then cooled to room temperature and filtered through a coarse frit glass funnel. The cake was rinsed with more dioxane and the filtrate solution was used for the next step without further purification. LC/MS = 155.1 (M+H of boronic acid); Rt = 0.41 min, LC = 1.37 min.
Synthesis of 5-methyl-3-(3-nitropyridin-4-yl)cyclohex-2-enone
Figure imgf000040_0002
To a solution of 5-methyl-3-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)cyclohex-2-enone (1.0 equiv.) in degassed dioxane (0.5 M) and 2M Na2C03 (2 equiv.) was added 4-chloro-3-nitropyridine (1.3 equiv.) and Pd(dppf)Cl2-DCM (0.05 equiv.). The reaction was placed under a reflux condenser and heated in an oil bath to 110°C for 1 h. Cooled to room temperature, filtered through a pad of Celite, washed the pad with ethyl acetate and concentrated the filtrate under vacuo. The residue was further pumped at 80 °C on a rotary evaporator for one hour to remove boronate by-products (M+H = 101) via sublimation. The residue was partitioned between brine and ethyl acetate, and the layers were separated, the aqueous phase was further extracted with ethyl acetate (4x), the organics were combined, dried over sodium sulfate, filtered, and concentrated. The crude was purified via silica gel chromatography loading in DCM and eluting with 2-50% ethyl acetate and hexanes. The pure fractions were concentrated in vacuo to yield an orange oil. The oil was placed under high vacuum (-500 mtorr) with seed crystals overnight to yield an orange solid. The solid was further purified via trituration in hexanes to yield 5- methyl-3-(3-nitropyridin-4-yl) cyclohex-2-enone (48% 2 steps). LC/MS = 233.2 (M+H); Rt = 0.69 min, LC = 2.70 min. 1H-NMR (400 MHz, CdCl3) δ ppm: 9.31 (s, IH), 8.88 (d, J=5.1, IH), 7.30 (d, J=5.1, IH), 6.00 (d, J=2.4, IH), 2.62 (dd, J=16.4, 3.5, IH), 2.53-2.34 (m, 3H), 2.23 (dd, J=16.1, 11.7, IH), 1.16 (d, J=6.3, 3H).
Synthesis of cis-(+/-)-5-methyl-3-(3-nitropyridin-4-yl)cvclohex-2-enol
Figure imgf000041_0001
To a solution of 5-methyl-3-(3-nitropyridin-4-yl)cyclohex-2-enone (1.0 equiv.) in EtOH (0.3 M) was added CeCl3-7H20 (1.2 equiv.). The reaction was cooled to 0°C, then NaBH4 (1.2 equiv.) was added in portions. Stirred for 1 h at 0°C, then quenched by adding water, concentrated to remove the EtOH, added EtOAc, extracted the organics, washed with brine, then dried with Na2S04, filtered and concentrated to yield cis-(+/-)-5- methyl-3-(3-nitropyridin-4-yl)cyclohex-2-enol (94%). LC/MS = 235.2 (M+H), LC = 2.62 min.
Synthesis of (+/-)-4-(5-methylcyclohexa- 1 ,3-dienyl)-3-nitropyridine
Figure imgf000041_0002
To a solution of (+/-)-5-methyl-3-(3-nitropyridin-4-yl)cyclohex-2-enol (1.0 equiv.) in dioxane (0.1M) was added p-TSA (1.0 equiv.), and the reaction was stirred at 100 °C for 3 h. The solution was cooled to room temperature, then passed through a pad of neutral alumina eluting with EtOAc to yield (+/-)-4-(5-methylcyclohexa-l,3-dienyl)-3- nitropyridine as a yellow oil in 68% yield. LC/MS = 217.1 (M+H), LC = 3.908 min.
Synthesis of (+/-)-6-bromo-5-methyl-3-(3-nitropyridin-4-yl)cyclohex-2-enol
Figure imgf000042_0001
To a solution of 4-(5-methylcyclohexa-l,3-dienyl)-3-nitropyridine (1.0 equiv.) in THF and water (1 : 1, 0.13 M) was added NBS (1.5 equiv.) and the reaction was stirred at room temperature for 30 min. Upon completion, ethyl acetate and water were added to the reaction, the organic phase was dried with brine, then sodium sulfate, filtered, and concentrated. The crude material was purified via silica gel column chromatography eluting with ethyl acetate and hexanes (1 : 1) to give (+/-)-6-bromo-5-methyl-3-(3- nitropyridin-4-yl)cyclohex-2-enol as a yellow oil in 80% yield. LC/MS = 315.0/313.0 (M+H), LC = 2.966 min.
Synthesis of (+/-)-2-azido-6-methyl-4-(3-nitropyridin-4-yl)cyclohex-3-enol
Figure imgf000043_0001
To a solution of (+/-)-6-bromo-5-methyl-3-(3-nitropyridin-4-yl)cyclohex-2-enol (1.0 equiv.) in THF (0.1 M) was added potassium tert-butoxide (1.5 equiv.). The reaction turned from orange to black almost immediately. By TLC, the formation of product is clean in 30 min. Quenched by adding saturated ammonium chloride and ethyl acetate. The organic phase was dried with brine, then sodium sulfate, filtered, and concentrated. The crude product was dissolved in ethanol and water (3: 1, 0.1 M), and ammonium chloride (2.0 equiv) and sodium azide (2.0 equiv.) were added. The dark orange reaction was stirred at room temperature overnight. The conversion to product is clean as indicated by LC/MS. The reaction was concentrated to remove the ethanol, ethyl acetate and water were added, and the organic phase was dried with sodium sulfate, filtered, and concentrated. The crude material was purified via silica gel column chromatography eluting with ethyl acetate and hexanes (1 : 1) to give (+/-)-2-azido-6-methyl-4-(3- nitropyridin-4-yl)cyclohex-3-enol in 55% yield. LC/MS = 276.0 (M+H), LC = 2.803 min.
Synthesis of (+/-)-tert-butyl 6-hvdroxy-5-methyl- 3-(3-nitropyridin-4-yl)cyclohex-2-enylcarbamate
Figure imgf000044_0001
To a solution of (+/-)-2-azido-6-methyl-4-(3-nitropyridin-4- yl)cyclohex-3-enol (1.0 equiv.) in pyridine and ammonium hydroxide (8:1, 0.08 M) was added trimethylphosphine (3.0 equiv.) and the brown solution was stirred at room temperature for 2 h. Upon completion, EtOH was added and the solution was concentrated in vacuo. More ethanol was added and the reaction was concentrated again. Dioxane and sat. NaHC03 (1 : 1, 0.08 M) were added to the crude, followed by Boc20 (1.0 equiv.). Stirred the reaction mixture at room temperature for 2h, then added water and ethyl acetate. The organic phase was dried with MgSC^, and concentrated. The crude product was purified via silica gel column chromatography eluting with ethyl acetate and hexanes (1 : 1) to afford (+/-)-tert-butyl 6-hydroxy-5-methyl-3-(3-nitropyridin- 4-yl)cyclohex-2-enylcarbamate (59%). LC/MS = 350.1 (M+H), Rt: 0.76 min.
Synthesis of (+/-)-2-(tert-butoxycarbonylamino)-6-methyl- 4-(3-nitropyridin-4-yl)cyclohex-3-enyl acetate
Figure imgf000044_0002
To a solution of (+/-)-tert-butyl 6-hydroxy-5-methyl-3-(3-nitropyridin- 4-yl)cyclohex-2-enylcarbamate (1.0 equiv.) in pyridine (0.1 M) was added Ac20 (2.0 equiv.) and the reaction was stirred at room temperature overnight. Upon completion, the reaction was concentrated to dryness, then worked-up with ethyl acetate and water. The organic phase was dried with brine, then sodium sulfate, filtered, and concentrated to give (+/ -)-2-(tert-butoxycarbonylamino)-6-methyl-4-(3 -nitropyridin-4-yl)cyclohex-3 -enyl acetate in 94% yield. LC/MS = 392.2 (M+H), Rt = 0.94 min.
Synthesis of (+/-)-4-(3-aminopyridin-4-yl)-2-(tert-butoxycarbonylamino)-6- methylcyclohexyl acetate
Figure imgf000045_0001
To a degassed solution of (+/-)-2-(tert-butoxycarbonylamino)-6-methyl- 4-(3-nitropyridin-4-yl)cyclohex-3-enyl acetate (1.0 equiv.) in MeOH and EtOAc (1 : 1, 0.1 M) was added 10%> Pd/C (0.1 equiv.) and the reaction was stirred at room temperature under a hydrogen balloon for 3 days. Upon completion, the solution was filtered through a pad of Celite, the pad was washed with ethyl acetate and the filtrate was concentrated. The crude material contained about 10% of the undesired isomer. The crude was dissolved in ethyl acetate (-20%) and hexanes and heated until all dissolved. The solution was allowed to sit at room temperature for 2 days. The precipitate was then collected to give (+/-)-4-(3-aminopyridin-4-yl)-2-(tert-butoxycarbonylamino)-6- methylcyclohexyl acetate as the pure product in 59% yield. LC/MS = 364.3 (M+H), Rt = 0.63 min. Synthesis of 4-(3-(tert-butyldimethylsilyloxy)-5-methylcvclohex- 1 -enyl)-3-nitropyridine
Figure imgf000046_0001
To a solution of 5-methyl-3-(3-nitropyridin-4-yl)cyclohex-2-enol (1.0 equiv.) in DMF (0.5 M) was added imidazole (4.0 equiv.) and TBDMSC1 (2.5 equiv.). After stirring for 18 hours the solution was portioned between EtOAc and H20 and separated. After washing further with H20 (3x) and NaCl (sat.), drying over MgSC^, filtering and removal of solvents, 4-(3-(tert-butyldimethylsilyloxy)-5-methylcyclohex-l- enyl)-3-nitropyridine was obtained (85%). LC/MS = 349.2 (M+H), LC = 5.99 min.
Synthesis of 4-(3-(tert-butyldimethylsilyloxy)-5-methylcyclohex- 1 -enyl)pyridin-3 -amine
Figure imgf000046_0002
A heterogeneous solution of 4-(3-(tert-butyldimethylsilyloxy)-5- methylcyclohex-l-enyl)-3-nitropyridine (1.0 eq.) and iron (6.0 eq) in acetic acid, at a concentration of 0.4 M, was stirred vigorously for 2 hours. The mixture was then passed through a celite pad, eluting with MeOH. Upon removal of the volatiles in vacuo, the residue was dissolved in EtOAc, washed with Na2C03 (sa ), NaCl(sa ), was dried over MgS04, was filtered and the volatiles were removed in vacuo yielding 4-(3-(tert- butyldimethylsilyloxy)-5-methylcyclohex-l-enyl)pyridin-3 -amine (78%). LCMS (m/z): 319.3 (MH+); LC R, = 3.77 min. Synthesis of 4-(3 -(tert-butyldimethylsilyloxy)-5 -methylcvclohexyl)pyridin-3 -amine
Figure imgf000047_0001
To a solution of 4-(3-(tert-butyldimethylsilyloxy)-5-methylcyclohex-l- enyl)-3-nitropyridine (1.0 equiv.) in methanol, at a concentration of 0.1 M, was added 10% palladium on carbon (0.1 eq.). The resultant heterogeneous solution was put under an atmosphere of hydrogen and was stirred for 15 hours. At this time the mixture was filtered through a pad of celite eluting with methanol. The volatiles were removed in vacuo yielding 4-(3 -(tert-butyldimethylsilyloxy)-5 -methylcyclohexyl)pyridin-3 -amine (90%). LCMS (m/z): 321.3 (MH+); LC R, = 3.85 min.
Synthesis of cis (+/-) benzyl 4-3-(tert-butyldimethylsilyloxy)-5- methylcvclohexyDpyridin-3-ylcarbamate
Figure imgf000047_0002
To a solution of cis-(+/-)-4-(3-(tert-butyldimethylsilyloxy)-5- methylcyclohexyl)pyridin-3 -amine in dichloromethane at a concentration of 0.5 M was added benzyl 2,5-dioxopyrrolidin-l-yl carbonate (1.1 equiv.) and DMAP (0.05 equiv.). After stirring for 16 hours at rt, additional benzyl 2,5-dioxopyrrolidin-l-yl carbonate (0.55 equiv.) and DMAP (0.03 equiv.) were added. After stirring for an additional 24 hours at rt, additional benzyl 2,5-dioxopyrrolidin-l-yl carbonate (0.1 equiv.) and DMAP (0.03 equiv.) were added. After stirring for 18 more hours the solution was partitioned between EtOAc and Na2C03(sat.) and separated. Upon further washing with Na2C03(sat.) (2x) and NaCl(sat.), drying over MgS04, filtering and removal of solvents, cis (+/-) benzyl 4-3 -(tert-butyldimethylsilyloxy)-5 -methylcyclohexyl)pyridin-3 -ylcarbamate was obtained. The crude material was used as is. LC/MS = 455.3 (M+H), LC = 4.39 min.
Synthesis of cis-(+/-)benzyl 4-(3-hydroxy-
5 -methylcvclohexyl)pyridin-3 -ylcarbamate
Figure imgf000048_0001
A solution of cis (+/-) benzyl 4-3-(tert-butyldimethylsilyloxy)-5- methylcyclohexyl)pyridin-3-ylcarbamate in 1 :2: 1 6N HCl/THF/MeOH at a concentration of 0.1 M was stirred at rt for 6 hours. The pH was than adjusted to pH=7 by addition of 6N NaOH and the volatiles were removed in vacuo. The aqueous layer was extracted with EtOAc and the organic was washed with NaCl(sat), dried over MgS04, filtered and upon removal of the volatiles in vacuo, cis-(+/-)benzyl 4-(3-hydroxy-5- methylcyclohexyl)pyridin-3 -ylcarbamate was obtained. The crude material was used as is. LC/MS = 341.2 (M+H), LC = 2.38 min. Synthesis of cis (+/-)-benzyl 4-(3-methyl-5-oxocvclohexyl)pyridin-3 -ylcarbamate
Figure imgf000048_0002
To a 0 °C solution of cis-(+/-)-benzyl 4-(3-hydroxy-5-methyl- cyclohexyl)pyridin-3 -ylcarbamate in wet CH2CI2 at a concentration of 0.16 M was added Dess-Martin Periodinane (1.5 equiv.) and the solution was stirred for 18 hours as it warmed to rt. The solution was partitioned between EtOAc and 1 : 1 10% Na2S203/NaHC03(sat.) and separated. Upon further washing with 1 : 1 10% Na2S203/NaHC03(sat.) (2x) and NaCl(sat), drying over MgS04, filtering, removal of solvents and purification by silica gel chromatography (75-100% EtOAc/hexanes), cis- (+/-)-benzyl-4-(3-methyl-5-oxocyclohexyl)pyridin-3-ylcarbamate was obtained as a white solid (53%, 5 steps). LC/MS = 339.2 (M+H). Synthesis of cis-(+/-)- benzyl 4-(-3-(benzylamino)-
5 -methylcyclohexy l)pyridin-3 -ylcarbamate
Figure imgf000049_0001
A solution of cis-(+/-)-benzyl-4-(3-methyl-5-oxocyclohexyl)pyridin-3- ylcarbamate (1.0 equiv) and benzylamine (3.0 equiv) in MeOH, at a concentration of 0.25 M, was stirred at rt for 2 hours. Upon cooling in a -78 °C bath, LiBH4 (1.1 equiv, 2.0 M in THF) was added and the solution was allowed to warm to rt with stirring over 16 hours. The solution was partitioned between EtOAc and NaHC03(sat ), separated, washed further with NaHC03(sat.) and NaCl(sat), dried over MgS04, filtered and after removal of volatiles in vacuo, cis-(+/-)- benzyl 4-(-3-(benzylamino)-5-methylcyclohexyl)pyridin-3- ylcarbamate was obtained as a 4: 1 mixture of isomers, with the all cis as predominant LC/MS = 430.3 (M+H), LC = 0.62 min.
Synthesis of cis (+/-)-tert-butyl (-3-(3-aminopyridin-4-yl)-5-methylcvclohexylcarbamate
Figure imgf000049_0002
To a solution of cis-(+/-)- benzyl 4-(-3-(benzylamino)-5- methylcyclohexyl)pyridin-3-ylcarbamate was (1.0 equiv.) in methanol, at a concentration of 0.07 M, was added 20% palladium hydroxide on carbon (0.2 eq.). The resultant heterogeneous solution was put under an atmosphere of hydrogen and was stirred for 14 hours. At this time the reaction was purged with Ar, Boc20 (1.0 equiv.) was added and the solution was stirred for 8 hours. Additional Boc20 (1.0 equiv.) was added and the solution was stirred for 16 more hours. At this time the mixture was filtered through a pad of celite eluting with methanol. Upon removal of volatiles in vacuo, purification by silica gel chromatography (2.5-2.5 MeOH/CH2Cl2 with 0.1% DIEA) and recrystallization from 10% EtOAc/hexanes yielded cis (+/-)-tert-butyl (-3-(3-aminopyridin-4-yl)-5- methylcyclohexylcarbamate (49%). LCMS (m/z): 306.3 (MH+), LC R, = 2.59 min . Pure enantiomers could be obtained by chiral chromatography.
Synthesis of ethyl 2-amino-2-cyanoacetate
Figure imgf000050_0001
To a solution of ethyl 2-cyano-2-(hydroxyimino)acetate (leq) in 70 mL of water and 56 mL of aq. sat. sodium bicarbonate was added portionwise throughout 10 minutes Na2S204 (2.8 eq) The reaction mixture was stirred at room temperature for 1 hour. The solution was saturated with sodium chloride, extracted with methylene chloride (300mL x 3) and then the combined organic layers were dried over anhydrous Na2S04, filtered, and concentrated in vacuo to give ethyl 2-amino-2-cyanoacetate, which was used to next step without further (55%). LC/MS (m/z): 129.0 (MH+), Rt: 0.25 min.
Synthesis of ethyl 2-cyano-2-formamidoacetate
Figure imgf000050_0002
A mixture of acetic anhydride (1.80 eq.) and formic acid (1.85 eq.) was heated to 55 °C for 3 h. The reaction mixture was then cooled to 0°C and a solution of ethyl 2-amino-2- cyanoacetate (1.00 eq.) in THF (0.2 M) was added dropwise. The mixture was allowed to warm to RT for 18 h and then concentrated in vacuo. The crude residue was purified by flash column chromatography eluting with (EtOAc : hexanes= 1 : 4) to give ethyl 2- cyano-2-formamidoacetate in 70% yield. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.38 (t, J=7.24 Hz, 3 H) 4.39 (q, J=7.04 Hz, 2 H) 5.48 - 5.64 (m, 1 H) 6.38 - 6.67 (m, 1 H) 7.26 (s, 1 H) 8.32 (s, 1 H).
Synthesis of ethyl 5-aminothiazole-4-carboxylate
Figure imgf000051_0001
To a solution of 2-cyano-2-formamidoacetate (1.00 eq.) in pyridine (0.1 M) was added Lawesson's reagent (1.20 eq.) at RT. The resulting mixture was heated to 120 °C for 16 h. The reaction mixture was then concentrated in vacuo to yield a black oil. The oil was further purified by flash column chromatography eluting with (EtOAc : heptanes= 1 : 1) to afford ethyl 5-aminothiazole-4-carboxylate in 44% yield. LC/MS (m/z): 173.0 (MH+), R,: 0.40 min. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.35 - 1.46 (m, 3 H) 1.52 - 1.72 (m, 1 H) 4.34 - 4.45 (m, 2 H) 5.83 - 6.18 (m, 2 H) 7.22 - 7.30 (m, 1 H) 7.88 (s, l H)
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)thiazole-4-carboxylate
Figure imgf000051_0002
To a solution of ethyl 5-aminothiazole-4-carboxylate (1.00 eq.) in CH3CN (0.1 M) at RT was added Boc-anhydride (1.1 eq.) at RT. The mixture was stirred at RT for 68 h. The reaction mixture was then concentrated in vacuo and purified by flash column chromatography eluting with (EtOAc : heptanes= 1 : 5) to afford ethyl 5-((tert- butoxycarbonyl)amino)thiazole-4-carboxylate in 75% yield. LC/MS (m/z): 273.1 (MH+), Rt = 0.90 min). 1H NMR (400 MHz, <cdcl3>) δ ppm 1.45 (t, J=7.04 Hz, 3 H) 1.50 - 1.61 (m, 10 H) 4.45 (q, J=7.17 Hz, 2 H) 8.28 (s, 1 H) 9.92 (br. s., 1 H)
Synthesis of ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4-carboxylate
Figure imgf000052_0001
To a solution of ethyl 5-(tert-butoxycarbonylamino)thiazole-4-carboxylate (1.0 equiv.) in DCM (0.20 M) was added NBS (1.6 equiv) at RT. The resulting mixture was stirred at RT for 2 hrs. The reaction mixture was then concentrated in vacuo to give ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4-carboxylate in 100% yield and utilized in the next reaction without further purification. LC/MS = 352.9 (MH+), Rt = 1.12 min. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.33 - 1.46 (m, 3 H) 1.48 - 1.61 (m, 9 H) 4.34 - 4.52 (m, 2 H) 9.92 (br. s., 1 H)
Method 1
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- methoxyphenyl)thiazole-4-carboxylate
Figure imgf000052_0002
To a solution of ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4-carboxylate (1.0 equiv.) in THF and water (10: 1, 0.1 M) was added 2,6-difluoro-4-methoxyphenyl)boronic acid (2.0 equiv.) and potassium fluoride (3.00 equiv.). The reaction was degassed with nitrogen, then Pd2(dba)3 (0.20 equiv.) and tri-tert-butylphosphine (0.4 equiv.) were added and the reaction was heated to 100 °C under microwave irradiation for 20 min. LC/MS analysis indicated complete conversion of the starting material to product. The reaction was cooled to room temperature then quenched with water and diluted with EtOAc. The aqueous layer was separated then extracted with EtOAc. The combined organics were dried over MgSC^ and concentrated in vacuo to yield a brown oil. The crude product was purified by ISCO flash chromatography eluting with ethyl acetate and hexanes (0% to 30% ethyl acetate) to provide ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- methoxyphenyl)thiazole-4-carboxylate as the desired product as a white solid in 73% yield. LC/MS = 415.0 (M+H), Rt = 1.26 min; 1H NMR (400 MHz, <cdcl3>) δ ppm 1.45 (t, J=7.24 Hz, 3 H) 1.48 - 1.63 (m, 11 H) 3.74 - 3.89 (m, 3 H) 4.40 - 4.56 (m, 2 H) 6.43 - 6.65 (m, 2 H) 9.95 - 10.07 (m, 1 H).
Method 2
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-methoxyphenyl)thiazole-4- carboxylic acid
Figure imgf000053_0001
To a solution of methyl ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- methoxyphenyl)thiazole-4-carboxylate (1.0 equiv.) in THF/MeOH (2: 1, 0.3 M) was added LiOH (10.0 equiv.) and the reaction was stirred at 55 °C for 36 hour. The solution was quenched with 2N HC1, extracted with ethyl acetate, washed with brine, dried with sodium sulfate, filtered and concentrated to give 5-((tert-butoxycarbonyl)amino)-2-(2,6- difluoro-4-methoxyphenyl)thiazole-4-carboxylic acid in 99% yield. LC/MS = 386.9 (M+H-lBu), Rt = 1.00 min. 1H NMR (400 MHz, <dmso>) d ppm 1.48 (s, 9 H) 3.29 (br. s., 9 H) 3.76 - 3.90 (m, 3 H) 6.84 - 6.98 (m, 2 H) 10.06 (br. s., 1 H)
Method 3 Synthesis of 2-(2,6-difluoro-4-methylphenyl)-4,4,5,5-tetramethyl-l ,3,2- dioxaboroane
Figure imgf000054_0001
To a solution of l,3-difluoro-5-methylbenzene (1.0 eq) in dry THF (0.2M) under an atmosphere of N2 at -78°C was added n-butyllithium (leq, 1.6M in hexanes) slowly keeping the internal temperature below -65°C. The reaction was stirred for 2 hrs at - 78°C, followed by the addition of 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (1.15eq). The reaction was allowed to warm to room temperature. Upon completion, the reaction was quenched with NaHC03 (sat.) and extracted with EtOAc. The organics were washed with brine, dried over Na2S04, filtered and concentrated to yield a 2-(2,6- difluoro-4-methylphenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaboroane as a white solid in 92%. 1H NMR (400 MHz, <cdcl3>) δ ppm 6.67 (dd, J=9.39, 0.78 Hz, 2 H), 2.34 (s, 3 H), 1.38 (s, 12 H).
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- methylphenyl)thiazole-4-carboxylate
Figure imgf000054_0002
Method 1 was followed using ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4- carboxylate (1.0 equiv.) and 2-(2,6-difluoro-4-methylphenyl)-4,4,5,5-tetramethyl-l,3,2- dioxaboroane (2.00 equiv.) to give ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro- 4-methylphenyl)thiazole-4-carboxylate as a solid in 70% yield. LC/MS =399.2 (M+H), Rt = 1.19 min.
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- methylphenyl)thiazole-4-carboxylic acid
Figure imgf000055_0001
To a solution of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- methylphenyl)thiazole-4-carboxylate (l .Oeq) in THF/EtOH (2: 1, 0.3 M) was added LiOH (10.0 equiv.) and the reaction was stirred at 55 °C for 28 hour. The solution was quenched with 2N HCl, extracted with ethyl acetate, washed with brine, dried with sodium sulfate, filtered and concentrated to give 5-((tert-butoxycarbonyl)amino)-2-(2,6- difluoro-4-methylphenyl)thiazole-4-carboxylic acid in 64% yield. LC/MS = 371.2 (M+H), Rt = 0.98 min.
Synthesis of l,3-difluoro-5-isopropoxybenzene
Figure imgf000056_0001
To a solution of 3,5-difluorophenol (1.0 equiv.) in DMF (0.26 M) was added potassium carbonate (2.2 equiv.) followed by 2-iodopropane (1.1 equiv.) and the reaction was stirred overnight at room temperature. The reaction was poured into a separatory funnel and diluted with a 3: 1 (v/v) solution of EtOAc:heptanes. The organic phase was washed with water, then sat'd NaHC03. The remaining organic phase was dried over MgS04, filtered and concentrated in vacuo to provide l,3-difluoro-5-isopropoxybenzene in 88% yield. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.33 (d, J=6.26 Hz, 6 H), 4.48 (dt, J=11.93, 6.16 Hz, 1 H), 6.31 - 6.47 (m, 3 H).
Synthesis of 2-(2,6-difluoro-4-isopropoxyphenyl)-4,4,5,5-tetramethyl-l ,3,2- dioxaborolane
Figure imgf000056_0002
Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.2 equiv.), butyllithium (1.2 equiv.) and l,3-difluoro-5-isopropoxybenzene (1.0 equiv.) to give 2-(2,6-difluoro-4-isopropoxyphenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane in 99% yield. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.24 (s, 12 H), 1.31 - 1.33 (m, 6 H), 4.43 - 4.56 (m, 1 H), 6.31 - 6.44 (m, 2 H). Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- isopropoxy henyl)thiazole-4-carboxylate
Figure imgf000057_0001
Method 1 was followed using methyl ethyl 2-bromo-5-((tert- butoxycarbonyl)amino)thiazole-4-carboxylate (1.0 equiv.) and 2-(2,6-difluoro-4- isopropoxyphenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.0 equiv.) at 90 °C under microwave irradiation for 15 min to give ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6- difluoro-4-isopropoxyphenyl)thiazole-4-carboxylate in 62% yield. LC/MS =443.2 (MH+), Rt = 1.29 min.
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- isopropoxyphenyl)thiazole-4-carboxylic acid
Figure imgf000057_0002
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- isopropoxyphenyl)thiazole-4-carboxylate to give 5-((tert-butoxycarbonyl)amino)-2-(2,6- difluoro-4-isopropoxyphenyl)thiazole-4-carboxylic acid in 76% yield. LC/MS = 415.2 (MH+), Rt = 1.11 min.
Synthesis of l-(ethoxymethyl)-3,5-difluorobenzene
Figure imgf000058_0001
To a solution of (3,5-difluorophenyl)methanol (1.0 equiv.) in DMF (0.225 M) at 0°C was added sodium hydride (1.5 eq). the reaction mixture was stirred for 15 min at 0°C before the dropwise addition of ethyl iodide ( 3.0 equiv.). The ice bath was removed and stirring was continued for 18 hours. The reaction mixture was quenched by the addition of water and diluted with 3: 1 EtOAc: heptanes. The combined organics were washed with water, dried over MgSC^, filtered, concentrated and purified by Si02 chromatography to yield l-(ethoxymethyl)-3,5-difluorobenzene in 99%.
Synthesis of 2-(4-(ethoxymethyl)-2,6-difluorophenyl)-4,4,5,5-tetramethyl-l ,3,2- dioxaborolane
Figure imgf000058_0002
To a solution of l-(ethoxymethyl)-3,5-difluorobenzene (l .Oeq) in dry THF (0.2 M) under an atmosphere of N2 at -78°C was added n-butyllithium (leq, 1.6M in hexanes) slowly keeping the internal temperature below -65°C. The reaction was stirred for 1 hr at -78°C, followed by the addition of 2-(4-(ethoxymethyl)-2,6-difluorophenyl)-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane (2.1 eq). The reaction was allowed to warm to room temperature. Upon completion, the reaction was quenched with NaHC03 (sat.) and extracted with EtOAc. The organics were washed with brine, dried over Na2S04, filtered and concentrated to yield 2-(4-(ethoxymethyl)-2,6-difluorophenyl)-4,4,5,5-tetramethyl- 1,3,2-dioxaborolane in 71%.
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(4-(ethoxymethyl)-2,6- difluorophenyl)thiazole-4-carboxylate
Figure imgf000059_0001
Method 1 was followed using ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4- carboxylate (1.0 equiv.) and 2-(4-(ethoxymethyl)-2,6-difluorophenyl)-4,4,5,5- tetramethyl-l,3,2-dioxaborolane (2.0 equiv.) at 100 °C under microwave irradiation for 20 min to give ethyl 5-((tert-butoxycarbonyl)amino)-2-(4-(ethoxymethyl)-2,6- difhiorophenyl)thiazole-4-carboxylate in 73% yield.. LC/MS =443.2 (MH+), Rt = 1.29 mm.
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(4-(ethoxymethyl)-2,6- difluorophenyl)thiazole-4-carboxylic acid
Figure imgf000060_0001
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(4-(ethoxymethyl)- 2,6-difluorophenyl)thiazole-4-carboxylate (1.0 eq) to give 5-((tert- butoxycarbonyl)amino)-2-(4-(ethoxymethyl)-2,6-difluorophenyl)thiazole-4-carboxylic acid in 76% yield. LC/MS = 415.2 (MH+), Rt = 1.06 min.
Synthesis of 4-(3,5-difluorophenoxy)tetrahydro-2H-pyran
Figure imgf000060_0002
To a solution of 3,5-difluorophenol (1.0 equiv.), tetrahydro-2H-pyran-4-ol (1.2 equiv.), and triphenylphosphine (2.0 equiv.) in THF (0.33 M) at 0 °C was added DIAD (2.0 equiv.) dropwise. The reaction mixture was stirred at rt overnight. The mixture was concentrated and purified by flash chromatography over silica gel (heptanes: ethyl acetate gradient) to give 4-(3,5-difluorophenoxy)tetrahydro-2H-pyran in 90 % yield. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.72 - 1.84 (m, 2 H), 1.96 - 2.09 (m, 2 H), 3.59 (ddd, J=1 1.64, 8.31 , 3.52 Hz, 2 H), 3.90 - 4.04 (m, 2 H), 4.44 (tt, J=7.78, 3.77 Hz, 1 H), 6.32 - 6.53 (m, 3 H). Synthesis of 2-(2,6-difluoro-4-((tetrahvdro-2H-pyran-4-yl)oxy)phenyl)-4,4,5,5- tetramethyl- 1 ,3 ,2-dioxaborolane
Figure imgf000061_0001
Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (1.5 equiv.), butyllithium (1.3 equiv.) and 4-(3,5-difluorophenoxy)tetrahydro-2H-pyran (1.0 equiv.) to give 2-(2,6-difluoro-4-((tetrahydro-2H-pyran-4-yl)oxy)phenyl)-4,4,5,5- tetramethyl-l,3,2-dioxaborolane in 33% yield. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.21 - 1.34 (m, 12 H), 1.78 (dtd, J=12.72, 8.31, 8.31, 3.91 Hz, 2 H), 1.93 - 2.09 (m, 2 H), 3.59 (ddd, J=11.64, 8.31, 3.13 Hz, 2 H), 3.89 - 4.01 (m, 2 H), 4.48 (tt, J=7.78, 3.77 Hz, 1 H), 6.40 (d, J=9.39 Hz, 2 H).
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-((tetrahvdro-2H- pyran-4-yl)oxy)phenyl)thiazole-4-carboxylate
Figure imgf000061_0002
Method 1 was followed using methyl ethyl 2-bromo-5-((tert- butoxycarbonyl)amino)thiazole-4-carboxylate (0.8 equiv.) and 2-(2,6-difluoro-4- ((tetrahydro-2H-pyran-4-yl)oxy)phenyl)-4,4,5,5-tetramethyl-l ,3,2-dioxaborolane (1.0 equiv.) at 100 °C under microwave irradiation for 20 min to give ethyl 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-((tetrahydro-2H-pyran-4- yl)oxy)phenyl)thiazole-4-carboxylate in 67% yield. LC/MS =485.0 (MH+), Rt = 1.23 min. 1H NMR (400 MHz, <cdcl3>) δ ppm 0.88 (t, J=6.85 Hz, 1 H) 1.24 (s, 9 H) 1.26 - 1.29 (m, 4 H) 1.73 - 1.87 (m, 2 H) 1.94 - 2.09 (m, 3 H) 3.52 - 3.66 (m, 2 H) 3.91 - 4.03 (m, 2 H) 4.41 - 4.54 (m, 2 H) 6.45 - 6.62 (m, 2 H) 10.02 (s, 1 H)
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-((tetrahvdro-2H-pyran-4- yl)oxy)phenyl)thiazole-4-carboxylic acid
Figure imgf000062_0001
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- ((tetrahydro-2H-pyran-4-yl)oxy)phenyl)thiazole-4-carboxylate to give 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-((tetrahydro-2H-pyran-4- yl)oxy)phenyl)thiazole-4-carboxylic acid in 70% yield. LC/MS = 457.0 (MH+), Rt = 1.03 min. 1H NMR (400 MHz, <dmso>) δ ppm 1.48 (s, 8 H) 1.51 - 1.64 (m, 2 H) 1.91 - 2.04 (m, 2 H) 3.37 - 3.60 (m, 5 H) 3.82 (d, J=11.74 Hz, 2 H) 4.63 -4.77 (m, 1 H) 6.98 (d, J=10.96 Hz, 2 H) 9.95 - 10.13 (m, 1 H)
Synthesis of l,3-difluoro-5-(2-methoxyethoxy)benzene
Figure imgf000062_0002
To a solution of 3,5-difluorophenol (1.0 equiv.), 2-methoxyethanol (3.0 equiv.) and triphenylphosphine (3.0 equiv) in THF (0.1 M) was added DIAD (3.0 equiv.). After stirring at rt for 18 hours, the volatiles were removed in vacuo and the residue was purified by Si02 chromatography to yield l,3-difluoro-5-(2-methoxyethoxy)benzene in 95%. !H NMR (400 MHz, <cdcl3>) δ ppm 6.41-6.47 m (3 H), 4.08 (t, 2H), 3.74 (t, 2H), 3.45 (s, 3 H).
Synthesis of 2-(2,6-difluoro-4-(2-methoxyethoxy)phenyl)-4,4,5,5-tetramethyl- 1,3.2- dioxaborolane
Figure imgf000063_0001
To a solution of l,3-difluoro-5-(2-methoxyethoxy)benzene (l .Oeq) in dry THF (0.2 M) under an atmosphere of N2 at -78°C was added n-butyllithium (leq, 1.6 M in hexanes) slowly keeping the internal temperature below -65°C. The reaction was stirred for 1 hr at -78°C, followed by the addition of 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.1 eq). The reaction was allowed to warm to room temperature. Upon completion, the reaction was quenched with NaHC03 (sat.) and extracted with EtOAc. The organics were washed with brine, dried over Na2S04, filtered and concentrated to yield 2-(2,6-difluoro-
4-(2-methoxyethoxy)phenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane. 1H NMR (400 MHz, <cdcl3>) δ ppm 6.42 (d, 2 H), 4.10 (m, 2H), 3.74 (m, 2H), 3.44 (s, 3 H), 1.37 (s, 12 H). Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(2- methoxyethoxy)phenyl)thiazole-4-carboxylate
Figure imgf000064_0001
Method 1 was followed using ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4- carboxylate (1.0 equiv.) and 2-(2,6-difluoro-4-((tetrahydro-2H-pyran-4-yl)oxy)phenyl)- 4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.0 equiv.) at 90 °C under microwave irradiation for 15 min to give ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(2- methoxyethoxy)phenyl)thiazole-4-carboxylatein 56% yield. LC/MS =459.2 (MH+), Rt = 1.17 min.
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(2- methoxyethoxy)phenyl)thiazole-4-carboxylic acid
Figure imgf000064_0002
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- ((tetrahydro-2H-pyran-4-yl)oxy)phenyl)thiazole-4-carboxylate to give 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-(2-methoxyethoxy)phenyl)thiazole-4- carboxylic acid in 88% yield. LC/MS = 431.2 (MH+), Rt = 0.99 min. Synthesis of 3-(3,5-difluorophenyl)oxetan-3-ol
Figure imgf000065_0001
To a solution of l-bromo-3,5-difluorobenzene in THF (0.27 M) under Ar was added Mg turnings (1.6 M). A reflux condenser was attached and the solution was submerged in a 90 °C oil bath and refluxed for two hours. The oxetan-3-one (1.0 equiv.) was added in THF via syringe. The solution was left stirring at rt under Ar overnight. The reaction solution was quenched by addition of NH4Cl(sat) and the solution was extracted with EtOAc, washed with NaCl(sat.), dried over MgS04, filtered, concentrated and purified by ISCO Si02 chromatography (0-100% EtOAc/n-heptanes gradient) to yield 3-(3,5- difhiorophenyl)oxetan-3-ol in 56% yield. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 4.82 (d, J=7.63 Hz, 2 H), 4.91 (d, J=7.63 Hz, 2 H), 7.16 - 7.23 (m, 2 H).
Synthesis of 3-(3,5-difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- vDphenvDoxetan-3 -ol
Figure imgf000065_0002
Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.5 equiv.), butyllithium (2.4 equiv.) and 3-(3,5-difluorophenyl)oxetan-3-ol (1.0 equiv.) to give 3-(3,5-difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)oxetan-3-ol in 79% yield. 1H NMR (400 MHz, <cdcl3>) δ ppml .34 - 1.42 (m, 12 H), 4.79 (d, J=7.63 Hz, 2 H), 4.90 (d, J=7.34 Hz, 2 H), 7.17 (d, J=8.22 Hz, 2 H).
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3-hvdroxyoxetan- 3-yl)phenyl)thiazole-4-carboxylate
Figure imgf000066_0001
Method 1 was followed using ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4- carboxylate (1.0 equiv.) and 3-(3,5-difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl)oxetan-3-ol (2.0 equiv.) at 100 °C under microwave irradiation for 20 min to give ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3-hydroxyoxetan-3- yl)phenyl)thiazole-4-carboxylate in 50% yield. LC/MS =457.2 (MH+), Rt = 1.04 min. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.42 - 1.52 (m, 4 H) 1.53 - 1.64 (m, 13 H) 3.25 (br. s., 1 H) 4.43 - 4.55 (m, 2 H) 4.79 (d, J=7.04 Hz, 2 H) 4.95 (d, J=7.04 Hz, 2 H) 7.33 (d, J=9.00 Hz, 2 H) 10.04 (s, 1 H). Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3-hvdroxyoxetan-3- yl)phenyl)thiazole-4-carboxylic acid
Figure imgf000067_0001
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3- hydroxyoxetan-3-yl)phenyl)thiazole-4-carboxylate to give 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3-hydroxyoxetan-3-yl)phenyl)thiazole-4- carboxylic acid in 76% yield. LC/MS = 429.2 (MH+), Rt = 0.85 min.
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3-fluorooxetan-3- yl)phenyl)thiazole-4-carboxylate
Figure imgf000067_0002
To a solution of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3- hydroxyoxetan-3-yl)phenyl)thiazole-4-carboxylate (1.0 equiv.) in CH2CI2 (0.04 M) at - 78 °C under Ar was added methylDAST (1.0 equiv.). After addition, the solution was stirred under Ar at -78 °C for 10 minutes and then the bath was removed. The reaction was allowed to warm up to rt and quenched by addition of NaHC03(Sat.). The solution was diluted with EtOAc, washed with NaHC03(sa ), NaCl(sa ), dried over MgS04, filtered, concentrated to yield ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3- fluorooxetan-3-yl)phenyl)thiazole-4-carboxylate in 99% yield. LC/MS = 459.2 (MH+), R, = 1.17 min.
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3-fluorooxetan-3- yl)phenyl)thiazole-4-carboxylic acid
Figure imgf000068_0001
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(3- fluorooxetan-3-yl)phenyl)thiazole-4-carboxylate to give 5-((tert-butoxycarbonyl)amino)- 2-(2,6-difluoro-4-(3-fluorooxetan-3-yl)phenyl)thiazole-4-carboxylic acid 73% yield. LC/MS = 430.0 (MH+), R, = 0.98 min.
Synthesis of 4-(3,5-difluorophenyl)tetrahvdro-2H-pyran-4-ol
Figure imgf000068_0002
To a solution of l-bromo-3,5-difluorobenzene in THF (0.16 M) under N2 was added Mg turnings (1.6 equiv.). A reflux condenser was attached and the solution was submerged in a 90 °C oil bath and refluxed for 2 hours at which time the heat was removed and the solution cooled to 0°C. Dihydro-2H-pyran-4(3H)-one (1.0 equiv.) in THF was added and the solution was stirred under N2 allowing to warm to rt for 16 hrs. The reaction was quenched by addition of sat. NH4C1 and the solution was extracted with EtOAc, washed with brine, dried over sodium sulfate, filtered, concentrated. The crude material was purified by ISCO Si02 chromatography eluting with 0-100% EtOAc/n-heptanes to yield 4-(3,5-difluorophenyl)tetrahydro-2H-pyran-4-ol in 37% yield. 1H NMR (400 MHz, <cdcl3>) 5 ppm 1.63 (d, J=12.13 Hz, 2 H), 2.11 (ddd, J=13.50, 11.15, 6.65 Hz, 2 H), 3.84 - 3.90 (m, 4 H), 6.72 (tt, J=8.75, 2.20 Hz, 1 H), 6.97 - 7.05 (m, 2 H).
Synthesis of 4-(3,5-difluorophenyl)-3,6-dihvdro-2H-pyran
Figure imgf000069_0001
4-(3,5-difluorophenyl)tetrahydro-2H-pyran-4-ol (1.0 equiv.) was dissolved in DCM (0.2 M) and cooled to 0 °C. TEA (2.8 equiv.) was added to the solution, followed by MsCl (1.3 equiv.). The reaction was stirred at rt for 2hrs. The solution was cooled to 0°C and DBU (3.0 equiv.) was added. The reaction was stirred at rt for 18hrs. The solution was concentrated and the residue was purified by Si02 chromatography (0-100%) EtOAc in Heptanes) to afford 4-(3,5-difluorophenyl)-3,6-dihydro-2H-pyran in 38% yield. 1H NMR (400 MHz, <cdcl3>) δ ppm 2.42 - 2.49 (m, 2 H), 3.93 (t, J=5.48 Hz, 2 H), 4.32 (q, J=2.74 Hz, 2 H), 6.16 - 6.22 (m, 1 H), 6.70 (tt, J=8.80, 2.35 Hz, 1 H), 6.85 - 6.94 (m, 2 H).
Synthesis of 4-(3,5-difluorophenyl)tetrahvdro-2H-pyran
Figure imgf000069_0002
a solution of 4-(3,5-difluorophenyl)-3,6-dihydro-2H-pyran (1.0 equiv.) in methanol 2 M) was added 10%> Pd/C (0.05 equiv.). The reaction was placed under an atmosphere of hydrogen and stirred for 18 hours. Upon completion, the solution was filtered over a pad of Celite, the pad was washed with DCM, the filtrate was concentrated in vacuo to give 4-(3,5-difluorophenyl)tetrahydro-2H-pyran in 71% yield. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.76 (br. s., 4 H), 2.75 (br. s., 1 H), 3.50 (br. s., 2 H), 4.08 (d, J=9.78 Hz, 2 H), 6.56 - 6.94 (m, 3 H).
Synthesis of 2-(2,6-difluoro-4-(tetrahydro-2H-pyran-4-yl)phenyl)-4,4,5,5- tetramethyl- 1 ,3 ,2-dioxaborolane
Figure imgf000070_0001
Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.2 equiv.), butyllithium (1.1 equiv.) and 4-(3,5-difluorophenyl)tetrahydro-2H-pyran (1.0 equiv.) to give 2-(2,6-difluoro-4-(tetrahydro-2H-pyran-4-yl)phenyl)-4,4,5,5-tetramethyl-
1,3,2-dioxaborolane in 100% yield. !H NMR (400 MHz, <cdcl3>) δ ppm 1.16 - 1.19 (m, 12 H), 1.65 - 1.74 (m, 4 H), 2.60 - 2.75 (m, 1 H), 3.37 - 3.51 (m, 2 H), 4.01 (dt, J=11.54, 3.42 Hz, 2 H), 6.67 (d, J=8.22 Hz, 2 H)
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(tetrahydro-2H- pyran-4-yl)phenyl)thiazole-4-carboxylate
Figure imgf000071_0001
Method 1 was followed using ethyl 2-bromo-5-((tert-butoxycarbonyl)amino)thiazole-4- carboxylate (1.0 equiv.) and 2-(2,6-difluoro-4-(tetrahydro-2H-pyran-4-yl)phenyl)-4,4,5,5- tetramethyl-l,3,2-dioxaborolane (2.0 equiv.) at 100 0 C for 20 min under microwave irradiation to give ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(tetrahydro-2H- pyran-4-yl)phenyl)thiazole-4-carboxylate in 84% yield. LC/MS = 469.2 (MH+), R, = 1.20 min. 1H NMR (400 MHz, <cdcl3>) δ ppm 0.83 - 0.92 (m, 1 H) 1.21 - 1.25 (m, 3 H) 1.25 - 1.32 (m, 4 H) 1.45 (t, J=7.04 Hz, 3 H) 1.50 - 1.60 (m, 9 H) 1.65 - 1.88 (m, 5 H) 2.69 - 2.87 (m, 1 H) 3.41 - 3.62 (m, 2 H) 4.02 - 4.17 (m, 2 H) 4.40 - 4.55 (m, 2 H) 6.79 - 6.94 (m, 2 H) 9.96 - 10.08 (m, 1 H)
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(tetrahvdro-2H-pyran-4- yl)phenyl)thiazole-4-carboxylic acid
Figure imgf000072_0001
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4- (tetrahydro-2H-pyran-4-yl)phenyl)thiazole-4-carboxylate to give 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-(tetrahydro-2H-pyran-4-yl)phenyl)thiazole-4- carboxylic acid in 72% yield. LC/MS = 441.1 (MH+), R, = 1.02 min. 1H NMR (400 MHz, <dmso>) δ ppm 1.01 - 1.06 (m, 4 H) 1.39 - 1.54 (m, 9 H) 1.59 - 1.76 (m, 5 H) 2.78 - 2.93 (m, 1 H) 3.34 - 3.57 (m, 5 H) 3.86 - 3.96 (m, 2 H) 7.21 (d, J=9.78 Hz, 2 H) 10.06 (s, 1 H)
Synthesis of 4-(3,5-difluorophenyl)tetrahydro-2H-pyran-4-ol
Figure imgf000072_0002
To a solution of l-bromo-3,5-difluorobenzene (1.6 equiv.) in THF (0.26 M) under Ar was added Mg turnings (1.6 equiv.). A reflux condenser was attached and the solution was submerged in a 90 °C oil bath and refluxed for two hours. The dihydro-2H-pyran-4(3H)- one (1.0 equiv.) was added in THF via syringe. The solution was left stirring at rt under Ar for 5 hrs. The reaction solution was quenched by addition of NH4Cl(sat) and the solution was extracted with EtOAc, washed with NaCl(sa ), dried over MgS04, filtered, concentrated and purified by ISCO Si02 chromatography (0-100% EtOAc/n-heptanes gradient) to yield 4-(3,5-difluorophenyl)tetrahydro-2H-pyran-4-ol in 71% yield. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.59 - 1.68 (m, 3 H), 2.07 - 2.19 (m, 2 H), 3.87 - 3.93 (m, 4 H), 6.72 (tt, J=8.75, 2.20 Hz, 1 H), 6.97 - 7.06 (m, 2 H).
Synthesis of 4-(3,5-difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl)tetrahydro-2H-pyran-4-ol
Figure imgf000073_0001
Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.5 equiv.), butyllithium (2.4 equiv.) and 4-(3,5-difluorophenyl)tetrahydro-2H-pyran-4-ol (1.0 equiv.) to give 4-(3,5-difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenyl)tetrahydro-2H-pyran-4-ol in 97% yield. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.32 - 1.42 (m, 12 H), 1.56 - 1.65 (m, 2 H), 2.11 (d, J=3.13 Hz, 2 H), 3.86 - 3.92 (m, 4 H), 6.99 (d, J=9.00 Hz, 2 H).
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4- hydroxytetrahydro-2H-pyran-4-yl)phenyl)thiazole-4-carboxylate
Figure imgf000074_0001
Method 1 was followed using methyl ethyl 2-bromo-5-((tert- butoxycarbonyl)amino)thiazole-4-carboxylate (1.0 equiv.) and 4-(3,5-difluoro-4-(4,4,5,5- tetramethyl-l ,3,2-dioxaborolan-2-yl)phenyl)tetrahydro-2H-pyran-4-ol (2.0 equiv.) at 100 °C for 20 min under microwave irradiation to give ethyl 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4-hydroxytetrahydro-2H-pyran-4- yl)phenyl)thiazole-4-carboxylate in 70% yield. LC/MS = 485.1 (MH+), R, = 1.07 min. 1H NMR (400 MHz, <cdcl3>) δ ppm 0.88 (t, J=6.85 Hz, 2 H) 1.20 - 1.25 (m, 2 H) 1.38 - 1.49 (m, 3 H) 1.51 - 1.58 (m, 9 H) 1.65 (d, J=13.30 Hz, 3H) 2.07 - 2.20 (m, 2 H) 3.83 - 3.96 (m, 4 H) 4.39 - 4.54 (m, 2 H) 7.16 (d, J=9.78 Hz, 2 H) 10.03 (s, 1 H)
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4-hvdroxytetrahydro-2H- pyran-4-yl)phenyl)thiazole-4-carboxylic acid
Figure imgf000074_0002
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4- hydroxytetrahydro-2H-pyran-4-yl)phenyl)thiazole-4-carboxylate to give 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4-hydroxytetrahydro-2H-pyran-4- yl)phenyl)thiazole-4-carboxylic acid in 86% yield. LC/MS = 457.0 (MH+), R, = 0.86 min. 1H NMR (400 MHz, <dmso>) δ ppm 1.42 - 1.55 (m, 13 H) 1.90 - 2.07 (m, 3 H) 3.60 - 3.81 (m, 6 H) 7.36 (d, J=10.56 Hz, 2 H) 10.07 (s, 1 H)
Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4- fluorotetrahvdro-2H-pyran-4-yl)phenyl)thiazole-4-carboxylate
Figure imgf000075_0001
To a solution of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4- hydroxytetrahydro-2H-pyran-4-yl)phenyl)thiazole-4-carboxylate (1.0 equiv.) in CH2CI2 (0.01 M) at -78 °C was added DASTF (1.0 equiv.) dropwise. The resulting mixture was allowed to warm to RT and stirred at this temperature for a further 2 hrs. The reaction mixture was then quenched with NaHC03 and diluted with EtOAc. The aqueous layer was separated then extracted with EtOAc. The combined organics were dried over MgS04 and concentrated in vacuo to yield ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6- difluoro-4-(4-fluorotetrahydro-2H-pyran-4-yl)phenyl)thiazole-4-carboxylate in 100% yield. LC/MS = 487.1 (MH+), Rt = 1.21 min. The product was utilized in the subsequent reaction without further purification. Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4-fluorotetrahvdro-2H- pyran-4-yl)phenyl)thiazole-4-carboxylic acid
Figure imgf000076_0001
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4- fluorotetrahydro-2H-pyran-4-yl)phenyl)thiazole-4-carboxylate to give 5-((tert- butoxycarbonyl)amino)-2-(2,6-difluoro-4-(4-fluorotetrahydro-2H-pyran-4- yl)phenyl)thiazole-4-carboxylic acid in 62% yield. LC/MS = 459.0 (MH+), R, = 1.01 min. Synthesis of ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(tetrahvdro-2H- pyran-4-yl)phenyl)thiazole-4-carboxylate
Figure imgf000076_0002
Method 1 was followed using ethyl 2-bromo-5-(tert-butoxycarbonylamino)thiazole-4- carboxylate (1.0 equiv.) and 2-(2,6-difluoro-4-(tetrahydro-2H-pyran-4-yl)phenyl)- 4,4,5,5-tetramethyl-l,3,2-dioxaborolane (2.0 equiv.) at 100 °C for 20 min in microwave to give ethyl 5-(tert-butoxycarbonylamino)-2-(2,6-difluoro-4-(tetrahydro-2H-pyran-4- yl)phenyl)thiazole-4-carboxylate in 84% yield. LC/MS = 469.2 (MH+ ), Rt = 1.21 min. 1H NMR (400 MHz, <cdcl3>) δ ppm 0.83 - 0.92 (m, 1 H) 1.21 - 1.25 (m, 3 H) 1.25 - 1.32 (m, 4 H) 1.45 (t, J=7.04 Hz, 3 H) 1.50 - 1.60 (m, 9 H) 1.65 - 1.88 (m, 5 H) 2.69 - 2.87 (m, 1 H) 3.41 - 3.62 (m, 2 H) 4.02 - 4.17 (m, 2 H) 4.40 - 4.55 (m, 2 H) 6.79 - 6.94 (m, 2 H) 9.96 - 10.08 (m, 1 H)
Synthesis of 5-((tert-butoxycarbonyl)amino)-2-(2,6-difluoro-4-(tetrahvdro-2H-pyran-4- yl)phenyl)thiazole-4-carboxylic acid
Figure imgf000077_0001
Method 2 was followed using ethyl 5-((tert-butoxycarbonyl)amino)-2-(2,6-difiuoro-4- (tetrahydro-2H-pyran-4-yl)phenyl)thiazole-4-carboxylate to give 5-((tert- butoxycarbonyl)amino)-2-(2,6-difiuoro-4-(tetrahydro-2H-pyran-4-yl)phenyl)thiazole-4- carboxylic acid in 72% yield. LC/MS = 441.1 (MH+), Rt = 1.02 min. 1H NMR (400 MHz, <dmso>) δ ppm 1.01 - 1.06 (m, 4 H) 1.39 - 1.54 (m, 9 H) 1.59 - 1.76 (m, 5 H) 2.78 - 2.93 (m, 1 H) 3.34 - 3.57 (m, 5 H) 3.86 -3.96 (m, 2 H) 7.21 (d, J=9.78 Hz, 2 H) 10.06 (s, 1 H)
Synthesis of l-(3,5-difluorophenyl)cyclobutanol
Figure imgf000077_0002
To a solution of l-bromo-3,5-difiuorobenzene (1.0 equiv.) in THF (0.26 M) under added Mg turnings (1.6 equiv.). A reflux condenser was attached and the solution was submerged in a 90 °C oil bath and refluxed for two hours. The oxetan-3-one (1.0 equiv.) was added in THF via syringe. The solution was left stirring at rt under Ar for 5 hrs. The reaction solution was quenched by addition of NH4Cl(sat) and the solution was extracted with EtOAc, washed with NaCl(sat), dried over MgS04, filtered, concentrated and purified by ISCO Si02 chromatography (0-100% EtOAc/n-heptanes gradient) to yield l-(3,5-difluorophenyl)cyclobutanol in 54% yield. 1H NMR (400 MHz, CHLOROFORM- d) δ ppm 1.69 - 1.83 (m, 1 H), 2.03 - 2.13 (m, 1 H), 2.31 - 2.43 (m, 2 H), 2.45 - 2.56 (m, 2 H), 6.71 (tt, J=8.80, 2.35 Hz, 1 H), 6.98 - 7.07 (m, 2 H). Synthesis of l-(3,5-difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- vDphenvDcyclobutanol
Figure imgf000078_0001
Method 3 was followed using 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2- dioxaborolane (2.5 equiv.), butyllithium (2.4 equiv.) and l-(3,5- difluorophenyl)cyclobutanol (1.0 equiv.) to give l-(3,5-difluoro-4-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)phenyl)cyclobutanol in 100% yield. 1H NMR (400 MHz, <cdcl3>) δ ppm 1.23 - 1.25 (m, 12 H), 1.69 - 1.82 (m, 1 H), 2.05 - 2.12 (m, 1 H), 2.37 (br. s., 2 H), 2.47 (br. s., 2 H), 7.00 (d, J=8.80 Hz, 2 H). Synthesis of tert-butyl ((l S,3R,5S)-3-(3-(2-bromothiazole-4-carboxamido)pyridin-4-yl)-
5 -methylcyclohexyDcarbamate
Figure imgf000079_0001
A solution of tert-butyl ((l S,3R,5S)-3-(3-aminopyridin-4-yl)-5- methylcyclohexyl)carbamate (1.0 eq.), 2-bromothiazole-4-carboxylic acid (1.2 eq.), HO AT (1.2 eq.) and EDC (1.20) in DMF (0.2 M) was stirred at RT for 17 h. The solution was then diluted with EtOAc and washed subsequenetly with water, IN NaOH then saturated brine solution. The organic phase was dried with magnesium sulfate and concentrated in vaccuo to yield the product tert-butyl ((l S,3R,5S)-3-(3-(2-bromothiazole- 4-carboxamido)pyridin-4-yl)-5-methylcyclohexyl)carbamate as a pale yellow oil which was utilised without further purification. LC/MS = 495.0/497.0 (MH+), Rt = 0.90 min.
Synthesis of N-(4-((lR,3S,5S -3-amino-5-methylcvclohexynpyridin-3-vn-2-(2,6- difluoro-4-(4-hydroxytetrahydro-2H-pyran-4-yl)phenyl)thiazole-4-carboxamide
Figure imgf000079_0002
Method 1 was followed using tert-butyl ((l S,3R,5S)-3-(3-(2-bromothiazole-4- carboxamido)pyridin-4-yl)-5-methylcyclohexyl)carbamate (1.0 equiv.) and 2-(2,6- difluoro-4-(tetrahydro-2H-pyran-4-yl)phenyl)-4,4,5,5-tetramethyl-l ,3,2-dioxaborolane (2.0 equiv.) at 100 °C for 20 min in microwave to give N-(4-((lR,3S,5S)-3-amino-5- methylcyclohexyl)pyridin-3-yl)-2-(2,6-difluoro-4-(4-hydroxytetrahydro-2H-pyran-4- yl)phenyl)thiazole-4-carboxamide in 3% yield. LC/MS = 529.1 (MH+ ), Rt = 0.57 min. Synthesis of N-(4-((lR,3S,5S -3-amino-5-methylcvclohexynpyridin-3-vn-2-(2,6- difluoro-4-(3 -hydroxyoxetan-3 -yl)phenyl)thiazole-4-carboxamide
Figure imgf000080_0001
Method 1 was followed using tert-butyl ((lS,3R,5S)-3-(3-(2-bromothiazole-4- carboxamido)pyridin-4-yl)-5-methylcyclohexyl)carbamate (1.0 equiv.) and 3-(3,5- difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)oxetan-3-ol (2.0 equiv.) at 100 °C for 20 min in microwave to give N-(4-((lR,3S,5S)-3-amino-5- methylcyclohexyl)pyridin-3-yl)-2-(2,6-difluoro-4-(3-hydroxyoxetan-3-yl)phenyl)thiazole- 4-carboxamide in 5% yield. LC/MS = 501.1 (MH+ ), Rt = 0.55 min.
Synthesis of N-(4-((lR,3S,5S -3-amino-5-methylcvclohexynpyridin-3-vn-2-(2,6- difiuoro-4-( 1 -hvdroxycvclobutyl)phenyl)thiazole-4-carboxamide
Figure imgf000080_0002
Method 1 was followed using tert-butyl ((lS,3R,5S)-3-(3-(2-bromothiazole-4- carboxamido)pyridin-4-yl)-5-methylcyclohexyl)carbamate (1.0 equiv.) and 1 -(3,5- difluoro-4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl)cyclobutanol (2.0 equiv.) at 100 °C for 20 min in microwave to give N-(4-((lR,3S,5S)-3-amino-5- methylcyclohexyl)pyridin-3-yl)-2-(2,6-difluoro-4-(l-hydroxycyclobutyl)phenyl)thiazole- 4-carboxamide in 5% yield. LC/MS = 499.1 (MH+ ), Rt = 0.62 min.
Method 4
A homogeneous solution of 1 eq each of amine, carboxylic acid, HO AT and EDC in DMF, at a concentration of 0.5 M, was left standing for 24 hours at which time water and ethyl acetate were added. The organic phase was dried with sodium sulfate and purified via silica gel column chromatography eluting with ethyl acetate and hexanes to give the desired protected amide product. Alternatively the crude reaction mixture was directly purified by HPLC. Upon lyophilization, the TFA salt of the protected amide product was obtained. Alternatively, the HPLC fractions could be added to EtOAc and solid Na2C03, separated and washed with NaCl(sat). Upon drying over MgSC^, filtering and removing the volatiles in vacuo, the protected amide product was obtained as a free base. Alternatively, the crude reaction mixture was used for the deprotection step without further purification.
If an N-Boc protected amine was present, it was removed by treating with excess 4M HCl/ dioxane for 14 hours or by treating with 25% TFA/CH2C12 for 2 hours. Upon removal of the volatiles in vacuo, the material was purified by RP HPLC yielding after lyophilization the amide product as the TFA salt. Alternatively, the HPLC fractions could be added to EtOAc and solid Na2C03, separated and washed with NaCl(sa ). Upon drying over MgS04, filtering and removing the volatiles in vacuo the free base was obtained. Upon dissolving in MeCN/H20, adding 1 eq. of 1 N HCl and lyophilizing, the HCl salt of the amide product was obtained.
If an N-Boc, OAc group were present, prior to Boc deprotection, the acetate group could be cleaved by treating with K2C03 (2.0 equiv.) in ethanol at a concentration of 0.1 M for 24 hours. If a TBDMS ether was present, it was deprotected prior to Boc removal by treating with 6N HCl, THF, methanol (1 :2: 1) at room temperature for 12 h. After removal of volatiles in vacuo, the Boc amino group was deprotected as described above. Alternatively, the TBDMS ether and Boc group could be both deprotected with 6N HCl, THF, methanol (1 :2: 1) if left at rt for 24 hours, or heated at 60 °C for 3 hours.
Following the procedures of Method 4, the following compounds were prepared:
TABLE 1
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
In addition to LC/MS and LC characterization, representative compounds were analyzed by !H-NMR. The following data in Table 2 are typical spectra for representative compounds of the invention.
Table 2
Figure imgf000089_0002
1 H NMR (400 MHz, <cd3od>) d ppm 1.06 (d, J=6.26 Hz, 3 H) 1 .09 -
1.31 (m, 2 H) 1.54 (q, J=1 1.87 Hz, 1 H) 1.78 (br. s, 0 H) 1.95 (d, J=12.13 Hz, 1 H) 2.09 (d, J=12.13 Hz, 1 H) 2.21 (d, J=1 1.74 Hz, 1 H) 3.15 (t, J=12.13 Hz, 1 H) 3.71 - 3.77 (m, 2 H) 4.17 (dd, J=5.09, 3.52 Hz, 2 H) 6.77 (d,J=10.56 Hz, 2 H) 7.68 (d, J=5.87 Hz, 1 H) 7.68 (d, J=5.87 Hz, 1 H) 8.44 (d, J=5.87 Hz, 1 H) 9.22 (s, 1 H)
1 H NMR (400 MHz, <cd3od>) d ppm 1 .12 (d, J=6.26 Hz, 3 H) 1.47 (q, J=12.39 Hz, 1 H) 1 .73 (d, J=12.13 Hz, 2 H) 1.98 (dd, J=13.30, 2.74 Hz, 1 H) 2.24 (d, J=9.78 Hz, 1 H) 3.07 - 3.24 (m, 4 H) 3.41 (s, 3 H) 3.74 (dd,
J=5.48, 3.52 Hz, 2 H) 4.17 (m, J=9.00 Hz, 2 H) 6.79 (d, J=10.96 Hz, 2 H) 7.67 (d, J=5.48 Hz, 1 H) 8.43 (d, J=5.48 Hz, 1 H) 9.24 (s, 1 H)
1 H NMR (400 MHz, <dmso>) d ppm 0.84 (d, J=6.65 Hz, 3 H) 1.75 (d, J=5.09 Hz, 1 H) 2.65 (t, J=12.33 Hz, 1 H) 2.88 - 3.20 (m, 3 H) 3.35 - 3.64 (m, 3 H) 3.78 (d, J=9.78 Hz, 1 H) 3.83 (s, 3 H) 6.89 (d, J=10.56 Hz, 2 H) 7.31 (d, J=6.65 Hz, 1 H) 7.61 (br. s., 2 H) 8.05 (br. s., 3 H) 8.33 (d,J=6.26 Hz, 1 H) 9.03 (s, 1 H) 9.29 (s, 1 H)
1 H NMR (400 MHz, <cd3od>) d ppm 1.03 - 1 .1 1 (m, 3 H) 1 .12 - 1.33 (m, 2 H) 1.52 - 1 .66 (m, 1 H) 1.77 - 1 .92 (m, 1 H) 1.93 - 2.03 (m, 1 H) 2.08 - 2.17 (m, 1 H) 2.19 - 2.29 (m, 1 H) 3.1 1 - 3.25 (m, 1 H) 3.35 - 3.43 (m, 1 H) 4.71 - 4.78 (m, 2 H) 4.91 - 4.95 (m, 2 H) 7.42 - 7.52 (m, 2 H) 7.68 - 7.77 (m, 1 H) 8.44 - 8.51 (m, 1 H) 9.24 - 9.32 (m, 1 H)
1 H NMR (400 MHz, <cd3od>) d ppm 1 .00 (d, J=6.65 Hz, 3 H) 1.35 (d, J=5.87 Hz, 6 H) 1.69 - 1.84 (m, 2 H) 1.91 - 2.08 (m, 1 H) 2.71 - 2.87 (m, 1 H) 3.04 - 3.17 (m, 1 H) 3.22 - 3.27 (m, 1 H) 3.66 - 3.83 (m, 1 H) 3.92 - 4.07 (m, 1 H) 4.59 - 4.72 (m, 1 H) 6.63 - 6.79 (m, 2 H) 7.42 - 7.56 (m, 1 H) 8.24 - 8.41 (m, 1 H) 9.27 - 9.37 (m, 1 H)
H NMR (400 MHz, <cd3od>) d ppm 1.06 (d, J=6.65 Hz, 3 H) 1 .29 - 1.37 (m, 6 H) 1.45 - 1.60 (m, 2 H) 1.73 - 2.00 (m, 2 H) 2.10 (d, J=12.52 Hz, 1 H) 2.21 (d, J=1 1 .74 Hz, 1 H) 2.98 - 3.22 (m, 1 H) 4.67 (dt, J=12.03, 5.92 Hz, 1 H) 6.51 - 6.81 (m, 2 H) 7.69 (d, J=5.87 Hz, 1 H)
8.44 (d, J=5.87 Hz, 1 H) 9.24 (s, 1 H) 1 H NMR (400 MHz, <cd3od>) d ppm 1 .25 (d, J=6.26 Hz, 6 H) 1 .65 - 2.1 1 (m, 6 H) 3.10 - 3.19 (m, 2 H) 3.24 - 3.29 (m, 2 H) 3.30 - 3.40 (m, 1
27
H) 3.64 (s, 3 H) 4.53 - 4.62 (m, 1 H) 6.55 - 6.65 (m, 2 H) 7.43 - 7.48
(m, 1 H)
1 H NMR (400 MHz, <cd3od>) d ppm 1 .66 - 2.12 (m, 6 H) 3.14 - 3.17 (m, 1 H) 3.25 - 3.27 (m, 2 H) 3.31 - 3.42 (m, 2 H) 3.65 (s, 3 H) 4.63 -
28
4.69 (m, 2 H) 4.81 - 4.84 (m, 2 H) 7.32 - 7.39 (m, 2 H) 7.45 - 7.50 (m, 1
H)
1 H NMR (400 MHz, <cd3od>) d ppm 1 .66 - 2.12 (m, 6 H) 3.14 - 3.17 (m, 1 H) 3.25 - 3.27 (m, 2 H) 3.31 - 3.42 (m, 2 H) 3.65 (s, 3 H) 4.63 -
31
4.69 (m, 2 H) 4.81 - 4.84 (m, 2 H) 7.32 - 7.39 (m, 2 H) 7.45 - 7.50 (m, 1
H)
1 H NMR (400 MHz, <dmso>) d ppm 1 .53 - 1 .90 (m, 4 H) 1 .91 - 2.08
32 (m, 2 H) 3.01 - 3.25 (m, 4 H) 3.65 (s, 7 H) 7.26 (t, J=8.80 Hz, 2 H) 7.44
(s, 1 H) 7.45 - 7.57 (m, 2 H) 7.78 (br. s. , 3 H) 8.67 (s, 1 H)
Piml, Pim2, Pim3 AlphaScreen Assays
Pim 1, Pim 2 & Pim 3 AlphaScreen assays using high ATP (11 - 125X ATP Km) were used to determine the biochemical activity of the inhibitors. The activity of Pim 1, Pim 2, & Pim 3 is measured using a homogeneous bead based system quantifying the amount of phosphorylated peptide substrate resulting from kinase-catalyzed phosphoryl transfer to a peptide substrate. Compounds to be tested are dissolved in 100% DMSO and directly distributed to a white 384-well plate at 0.25 μΐ per well. To start the reaction, 5 μΐ of 100 nM Bad peptide (Biotin-AGAGRSRHSSYPAGT-OH (SEQ ID NO: l)) and ATP (concentrations described below) in assay buffer (50 mM Hepes, pH=7.5, 5 mM MgCl2, 0.05% BSA, 0.01% Tween-20, 1 mM DTT) is added to each well. This is followed by the addition of 5 μΐ/well of Pim 1 , Pim 2 or Pim 3 kinase in assay buffer (concentrations described below). Final assay concentrations (described below) are in 2.5% DMSO. The reactions are performed for ~2 hours, then stopped by the addition of 10 μΐ of 0.75 μg/ml anti-phospho Ser/Thr antibody (Cell Signaling), 10 μg/ml Protein A AlphaScreen beads (Perkin Elmer), and 10 μg/ml streptavidin coated AlphaScreen beads in stop/detection buffer (50 mM EDTA, 95 mM Tris, pH=7.5, 0.01% Tween-20). The stopped reactions are incubated overnight in the dark. The phosphorylated peptide is detected via an oxygen anion initiated chemiluminescence/fluorescence cascade using the Envision plate reader (Perkin Elmer).
Figure imgf000092_0001
Compounds of the foregoing examples were tested by the Pirn 1, Pirn 2 & Pirn 3 AlphaScreen assays and found to exhibit an IC50 values as shown in Table 3 below.
IC50, the half maximal inhibitory concentration, represents the concentration of test compound that is required for 50% inhibition of its target in vitro.
Cell Proliferation Assay
KMS11 (human myeloma cell line), were cultured in IMDM supplemented with 10%> FBS, sodium pyruvate and antibiotics. Cells were plated in the same medium at a density of 2000 cells per well into 96 well tissue culture plates, with outside wells vacant, on the day of assay.
Test compounds supplied in DMSO were diluted into DMSO at 500 times the desired final concentrations before dilution into culture media to 2 times final concentrations. Equal volumes of 2x compounds were added to the cells in 96 well plates and incubated at 37 °C for 3 days.
After 3 days plates were equilibrated to room temperature and equal volume of CellTiter-Glow Reagent (Promega) was added to the culture wells. The plates were agitated briefly and luminescent signal was measured with luminometer. The percent inhibition of the signal seen in cells treated with DMSO alone vs. cells treated with control compound was calculated and used to determine EC50 values (i.e., the concentration of a test compound that is required to obtain 50% of the maximum effect in the cells) for tested compounds, as shown in Table 3.
Using the procedures of the Piml, Pim2, Pim3 AlphaScreen Assays the IC50 concentrations of compounds of the previous examples were determined as shown in the Table 3.
Using the procedures of Cell Proliferation Assay, the EC50 concentrations of compounds of the examples were determined in KMS11 cells as shown in Table 3.
TABLE 3
Figure imgf000093_0001
0.00232 0.850
0.00107 0.729
0.00281 0.480
0.00440 2.338
0.00125 0.055
0.00157 0.239
0.00090 0.061
0.00189 0.436
0.00154 0.202
0.00207 5.550
8.72E-06 0.00039 0.000187 0.035
2.87E-05 0.00127 0.000995 1.945 0.000104 0.00597 0.00288 >10.000
1.87E-05 0.00059 0.000407 0.053
9.93E-06 0.00081 0.000278 0.169
1.79E-05 0.00064 0.000446 0.381
1.07E-05 0.00042 0.000322 0.031
1.25E-05 0.00188 0.000201 0.201
1.43E-05 0.00120 0.000251 3.840
3.51 E-05 0.01026 0.001 133 0.892
2.43E-05 0.00472 0.000721 >10.000
1.38E-05 0.00428 0.000199
1.22E-05 0.00350 0.000216
0.02007
0.786413 1
0.02157
1.308977 3 0.03185
35 0.618792
7
Compound structures in the tables marked as "Chiral" were prepared and tested in optically active form, having the absolute stereochemistry as shown; other compounds were prepared and tested in racemic form, and the depicted structure represents the relative stereochemistry at each chiral center.

Claims

1. A compound of Formula (I) :
Figure imgf000097_0001
4 alkyl)-pyrazole ring;
W is H or NH2;
Z is CH or N;
n is 1 or 2;
R1 is H, OH or OMe;
R2 is H or Me,
provided that when R1 is H, R2 is also H;
R3 is H or Ci-C4 alkyl;
R4 is selected from R*, -OR*, -OCH2CH2OR*, -CH2OR*, and a 4-6 membered cyclic ether optionally substituted with OH, OMe, or F, wherein each R* is independently C 1-C4 alkyl,
provided that R4 is not -OMe when Z is N;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1, wherein R3 is H.
3. The compound of claim 1, wherein R3 is Me. The compound of any of claims 1-3, wherein R2 is H. The compound of any of claims 1-3, wherein R2 is Me. The compound of any of claim 4, wherein R1 is H. The compound of any of claim 5, wherein R1 is OH.
8. The compound of claim 1, wherein Z is CH.
9. The compound of claim 1, which is of the formula IA:
Figure imgf000098_0001
11. The compound of claim 10, which is of the formula IB:
Figure imgf000099_0001
The compound of claim 9 or 11 , wherein
The compound of claim 10 or 11, wherein
Figure imgf000099_0002
The compound of claim 9 or 11, wherein represents a pyrazole of the
Figure imgf000099_0003
formula where R is methyl, ethyl or isopropyl.
15. The compound of claim 14, wherein RN is methyl.
Figure imgf000100_0001
The compound of claim 1 , wherein represents a pyridine of the
Figure imgf000100_0002
formula
The compound of claim 1 , wherein R4 is a tetrahydropyranyl group of the
Figure imgf000100_0003
formula:
wherein RT is H, OH, OMe, or F.
18. The compound of claim 1, wherein R4 is an oxetanyl group of the formula:
Figure imgf000100_0004
wherein R0 is H, OH, OMe, or F.
19. The compound of claim 1, wherein R4 is OMe, OEt, or OPr.
20. The compound of claim 1, wherein R4 is -CH2OMe or -CH2OEt.
21. The compound of claim 1 , which is selected from the compounds in Table 1.
22. A pharmaceutical composition comprising a compound of claim 1 and at least one pharmaceutically acceptable excipient.
23. The pharmaceutical composition of claim 22, further comprising an additional therapeutic agent.
24. The pharmaceutical composition of claim 23, wherein the additional therapeutic agent is selected from irinotecan, topotecan, gemcitabine, 5-fluorouracil, cytarabine, daunorubicin, PI3 Kinase inhibitors, mTOR inhibitors, DNA synthesis inhibitors, leucovorin, carboplatin, cisplatin, taxanes, tezacitabine, cyclophosphamide, vinca alkaloids, imatinib, anthracyclines, rituximab, and trastuzumab.
25. A method to treat a condition caused or exacerbated by excessive Pirn kinase activity, which comprises administering to a subject in need thereof an effective amount of a compound of claim 1.
26. The method of claim 25, wherein the condition is a cancer.
27. The method of claim 26, wherein the cancer is selected from carcinoma of the lungs, pancreas, thyroid, ovary, bladder, breast, prostate, or colon, melanoma, myeloid leukemia, multiple myeloma, erythroleukemia, villous colon adenoma, and osteosarcoma; or the autoimmune disorder is selected from Crohn's disease, inflammatory bowel disease, rheumatoid arthritis, and chronic inflammatory diseases. 28. A compound according to claim 1 for use in therapy.
29. Use of a compound according to claim 1 for the preparation of a medicament.
31. The compound according to claim 30, wherein the cancer is selected from carcinoma of the lungs, pancreas, thyroid, ovaries, bladder, breast, prostate or colon, melanoma, myeloid leukemia, multiple myeloma, erythro leukemia, villous colon adenoma, and osteosarcoma.
32. The compound of claim 31 , wherein the condition is an autoimmune disorder.
33. A method of treating a disease or condition mediated by PIM kinase, comprising administering to a subject in need thereof a therapeutically effective amount of a compound according to any one of Claims 1-25, or a pharmaceutically acceptable salt thereof.
34. The method of claim 33, wherein the disease is selected from carcinoma of the lungs, pancreas, thyroid, ovaries, bladder, breast, prostate or colon, melanoma, myeloid leukemia, multiple myeloma, erythro leukemia, villous colon adenoma, and
osteosarcoma; or the disease is an autoimmune disorder.
35. The method of claim 34, wherein the disease is an autoimmune disorder.
36. The method of claim 35, wherein the autoimmune disorder is selected from Crohn's disease, inflammatory bowel disease, rheumatoid arthritis, and chronic inflammatory diseases.
PCT/IB2013/058033 2012-08-31 2013-08-27 Novel aminothiazole carboxamides as kinase inhibitors WO2014033630A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261695618P 2012-08-31 2012-08-31
US61/695,618 2012-08-31

Publications (1)

Publication Number Publication Date
WO2014033630A1 true WO2014033630A1 (en) 2014-03-06

Family

ID=49515425

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2013/058033 WO2014033630A1 (en) 2012-08-31 2013-08-27 Novel aminothiazole carboxamides as kinase inhibitors

Country Status (1)

Country Link
WO (1) WO2014033630A1 (en)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9200004B2 (en) 2013-01-15 2015-12-01 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9278950B2 (en) 2013-01-14 2016-03-08 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9556197B2 (en) 2013-08-23 2017-01-31 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US9580418B2 (en) 2014-07-14 2017-02-28 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
CN106668022A (en) * 2015-11-05 2017-05-17 武汉应内药业有限公司 Application of aminothiazole MyD88 specific inhibitor TJM2010-5
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
WO2018177993A1 (en) 2017-03-31 2018-10-04 Bayer Cropscience Aktiengesellschaft Pyrazoles for controlling arthropods
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004078163A2 (en) 2003-02-28 2004-09-16 Transform Pharmaceuticals, Inc. Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
WO2008022164A2 (en) 2006-08-16 2008-02-21 Boehringer Ingelheim International Gmbh Pyrazine compounds, their use and methods of preparation
WO2008106692A1 (en) 2007-03-01 2008-09-04 Novartis Vaccines And Diagnostics, Inc. Pim kinase inhibitors and methods of their use
WO2009109576A1 (en) * 2008-03-03 2009-09-11 Novartis Ag Pim kinase inhibitors and methods of their use
WO2010026124A1 (en) 2008-09-02 2010-03-11 Novartis Ag Picolinamide derivatives as kinase inhibitors
WO2011029802A1 (en) * 2009-09-08 2011-03-17 F. Hoffmann-La Roche Ag 4-substituted pyridin-3-yl-carboxamide compounds and methods of use
WO2011124580A1 (en) 2010-04-07 2011-10-13 F. Hoffmann-La Roche Ag Pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
WO2012004217A1 (en) 2010-07-06 2012-01-12 Novartis Ag Cyclic ether compounds useful as kinase inhibitors
WO2013045461A1 (en) * 2011-09-27 2013-04-04 F. Hoffmann-La Roche Ag Pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004078163A2 (en) 2003-02-28 2004-09-16 Transform Pharmaceuticals, Inc. Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
WO2008022164A2 (en) 2006-08-16 2008-02-21 Boehringer Ingelheim International Gmbh Pyrazine compounds, their use and methods of preparation
WO2008106692A1 (en) 2007-03-01 2008-09-04 Novartis Vaccines And Diagnostics, Inc. Pim kinase inhibitors and methods of their use
WO2009109576A1 (en) * 2008-03-03 2009-09-11 Novartis Ag Pim kinase inhibitors and methods of their use
WO2010026124A1 (en) 2008-09-02 2010-03-11 Novartis Ag Picolinamide derivatives as kinase inhibitors
WO2011029802A1 (en) * 2009-09-08 2011-03-17 F. Hoffmann-La Roche Ag 4-substituted pyridin-3-yl-carboxamide compounds and methods of use
WO2011124580A1 (en) 2010-04-07 2011-10-13 F. Hoffmann-La Roche Ag Pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use
WO2012004217A1 (en) 2010-07-06 2012-01-12 Novartis Ag Cyclic ether compounds useful as kinase inhibitors
WO2013045461A1 (en) * 2011-09-27 2013-04-04 F. Hoffmann-La Roche Ag Pyrazol-4-yl-heterocyclyl-carboxamide compounds and methods of use

Non-Patent Citations (13)

* Cited by examiner, † Cited by third party
Title
"BIOREVERSIBLE CARRIERS IN DRUG DESIGN", 1987, AMERICAN PHARMACEUTICAL ASSOCIATION AND PERGAMON PRESS
"Cancer Principles and Practice of Oncology", 15 February 2001, LIPPINCOTT WILLIAMS & WILKINS PUBLISHERS
"IUPAC 1974 RECOMMENDATIONS FOR SECTION E, FUNDAMENTAL STEREOCHEMISTRY", PURE APPL. CHEM., vol. 45, 1976, pages 13 - 30
"Methods in Cell Biology", 1976, ACADEMIC PRESS, pages: 33
"Physicians' Desk Reference (PDR", 1993
BREUER M ET AL.: "Very high frequency of lymphoma induction by a chemical carcinogen in pim-1 transgenic mice", NATURE, vol. 340, no. 6228, 1989, pages 61 - 3
CHEMICAL ABSTRACTS INDEX GUIDE-APPENDIX IV, 1987
CUYPERS HT ET AL.: "Murine leukemia virus-induced T-cell lymphomagenesis: integration of proviruses in a distinct chromosomal region", CELL, vol. 37, no. 1, 1984, pages 141 - 50
DAI JM ET AL.: "Antisense oligodeoxynucleotides targeting the serine/threonine kinase Pim-2 inhibited proliferation of DU-145 cells", ACTA PHARMACOL SIN, vol. 26, no. 3, 2005, pages 364 - 8
MARCH: "Advanced Organic Chemistry: Reactions, Mechanisms and Structures", 1992, JOHN WILEY & SONS, pages: 69 - 74
SELTEN G ET AL.: "Proviral activation of the putative oncogene Pim-1 in MuLV induced T-cell lymphomas", EMBO J, vol. 4, no. 7, 1985, pages 1793 - 8
T. HIGUCHI; V. STELLA: "PRO-DRUGS AS NOVEL DELIVERY SYSTEMS", vol. 14, A.C.S. SYMPOSIUM SERIES
VERBEEK S ET AL.: "Mice bearing the E mu-myc and E mu-pim-1 transgenes develop pre-B-cell leukemia prenatally", MOL CELL BIOL, vol. 11, no. 2, 1991, pages 1176 - 9

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9278950B2 (en) 2013-01-14 2016-03-08 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
US9676750B2 (en) 2013-01-14 2017-06-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US11229631B2 (en) 2013-01-15 2022-01-25 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10517858B2 (en) 2013-01-15 2019-12-31 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as PIM kinase inhibitors
US9550765B2 (en) 2013-01-15 2017-01-24 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10828290B2 (en) 2013-01-15 2020-11-10 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as pim kinase inhibitors
US9200004B2 (en) 2013-01-15 2015-12-01 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US10265307B2 (en) 2013-01-15 2019-04-23 Incyte Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9849120B2 (en) 2013-01-15 2017-12-26 Incyte Holdings Corporation Thiazolecarboxamides and pyridinecarboxamide compounds useful as Pim kinase inhibitors
US9556197B2 (en) 2013-08-23 2017-01-31 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US10000507B2 (en) 2013-08-23 2018-06-19 Incyte Corporation Furo- and thieno-pyridine carboxamide compounds useful as pim kinase inhibitors
US9580418B2 (en) 2014-07-14 2017-02-28 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as Pim kinase inhibitors
US9890162B2 (en) 2014-07-14 2018-02-13 Incyte Corporation Bicyclic aromatic carboxamide compounds useful as pim kinase inhibitors
US9822124B2 (en) 2014-07-14 2017-11-21 Incyte Corporation Bicyclic heteroaromatic carboxamide compounds useful as Pim kinase inhibitors
US9802918B2 (en) 2015-05-29 2017-10-31 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9540347B2 (en) 2015-05-29 2017-01-10 Incyte Corporation Pyridineamine compounds useful as Pim kinase inhibitors
US9862705B2 (en) 2015-09-09 2018-01-09 Incyte Corporation Salts of a pim kinase inhibitor
US11066387B2 (en) 2015-09-09 2021-07-20 Incyte Corporation Salts of a Pim kinase inhibitor
US10336728B2 (en) 2015-09-09 2019-07-02 Incyte Corporation Salts of a Pim kinase inhibitor
US11505540B2 (en) 2015-09-09 2022-11-22 Incyte Corporation Salts of a Pim kinase inhibitor
US10450296B2 (en) 2015-10-02 2019-10-22 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US11053215B2 (en) 2015-10-02 2021-07-06 Incyte Corporation Heterocyclic compounds useful as Pim kinase inhibitors
US9920032B2 (en) 2015-10-02 2018-03-20 Incyte Corporation Heterocyclic compounds useful as pim kinase inhibitors
CN106668022A (en) * 2015-11-05 2017-05-17 武汉应内药业有限公司 Application of aminothiazole MyD88 specific inhibitor TJM2010-5
WO2018177993A1 (en) 2017-03-31 2018-10-04 Bayer Cropscience Aktiengesellschaft Pyrazoles for controlling arthropods
US10596161B2 (en) 2017-12-08 2020-03-24 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms
US11278541B2 (en) 2017-12-08 2022-03-22 Incyte Corporation Low dose combination therapy for treatment of myeloproliferative neoplasms

Similar Documents

Publication Publication Date Title
KR101345920B1 (en) Picolinamide derivatives as kinase inhibitors
EP2861585B1 (en) Novel ring-substituted n-pyridinyl amides as kinase inhibitors
WO2014033630A1 (en) Novel aminothiazole carboxamides as kinase inhibitors
WO2014033631A1 (en) N-(3-pyridyl) biarylamides as kinase inhibitors
WO2012120415A1 (en) Tetrasubstituted cyclohexyl compounds as kinase inhibitors
US20120225062A1 (en) Novel kinase inhibitors
EP2935270A1 (en) Aryl-substituted fused bicyclic pyridazine compounds
AU2011265439B2 (en) Picolinamide derivatives as kinase inhibitors

Legal Events

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

Ref document number: 13785628

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: 13785628

Country of ref document: EP

Kind code of ref document: A1