WO2010132598A1 - Heteroaryl compounds as pikk inhibitors - Google Patents

Heteroaryl compounds as pikk inhibitors Download PDF

Info

Publication number
WO2010132598A1
WO2010132598A1 PCT/US2010/034596 US2010034596W WO2010132598A1 WO 2010132598 A1 WO2010132598 A1 WO 2010132598A1 US 2010034596 W US2010034596 W US 2010034596W WO 2010132598 A1 WO2010132598 A1 WO 2010132598A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
amino
imidazo
pyrazol
pyrimidinyl
Prior art date
Application number
PCT/US2010/034596
Other languages
French (fr)
Inventor
Christiane Bode
Alessandro Boezio
Alan C. Cheng
Deborah Choquette
James R. Coats
Katrina W. Copeland
Hongbing Huang
Daniel La
Richard Lewis
Hongyu Liao
Michele Potashman
John Stellwagen
Shuyan Yi
Mark Norman
Markian Stec
Emily A. Peterson
Russell Graceffa
Original Assignee
Amgen Inc.
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 Amgen Inc. filed Critical Amgen Inc.
Priority to EP10719514.1A priority Critical patent/EP2430013B1/en
Priority to AU2010249040A priority patent/AU2010249040B2/en
Priority to US13/319,683 priority patent/US20120190666A1/en
Priority to JP2012510985A priority patent/JP2012526836A/en
Priority to CA2760778A priority patent/CA2760778A1/en
Priority to MX2011012037A priority patent/MX2011012037A/en
Publication of WO2010132598A1 publication Critical patent/WO2010132598A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention provides compounds that are PIKK (Phosphoinositide-3 -kinase related kinase) inhibitors, more specifically, mTOR and/or PBK ⁇ kinase inhibitors and are therefore useful for the treatment of diseases treatable by inhibition of kinases, specifically PB kinases, more specifically, mTOR and/or PBK ⁇ , such as cancer. Also provided are pharmaceutical compositions containing such compounds and processes for preparing such compounds.
  • PIKK Phosphoinositide-3 -kinase related kinase
  • PB kinases are a family of lipid kinases that have been found to play a key role in the regulation of many cellular processes including proliferation, survival, carbohydrate metabolism, and motility. PBKs are considered to have an important role in intracellular signal transduction. In particular, the PBKs generate and convey signals that have important roles in cancer. PBKs are ubiquitously expressed, are activated by a high proportion of cell surface receptors, especially those linked to tyrosine kinases, and influence a variety of cellular functions and events. Although some PBK activity is likely to be essential for cellular health, PBKs are a diverse group of enzymes for which there is increasing evidence of functional specialization. This opens up the possibility of developing iso form-selective inhibitors that can be used to treat cancer.
  • PBK The primary enzymatic activity of PBK is the phosphorylation of inositol lipids (phosphoinositides) on the 3-position of the inositol headgroup.
  • PB kinases catalyze the addition of phosphate to the 3'-OH position of the inositol ring of inositol lipids generating phosphatidyl inositol monophosphate, phosphatidyl inositol diphosphate and phosphatidyl inositol triphosphate.
  • PBKs There are a total of eight mammalian PBKs, which have been divided into three main classes on the basis of sequence homology, in vitro substrate preference, and method of activation and regulation.
  • Enzymes of a first class (Class I) have a broad substrate specificity and phosphorylate phosphatidylinositiol (Ptdlns), PtdIns(4)P and PtdIns(4,5)P 2 .
  • Class I PB kinases include mammalian pi 10a, pi lO ⁇ , pi lO ⁇ and pi lO ⁇ . Different members of the PB- kinase family generate different lipid products. To date, four 3-phosphorylated inositol lipids have been identified in vivo.
  • lipids are bound by proteins that contain the appropriate lipid recognition module and which either act as effectors or transmit the PBK signal onwards.
  • the most familiar form of PBK is a heterodimeric complex, consisting of a 110 kDa catalytic subunit now known as pi 10a and an 85 kDa regulatory/adapter subunit, p85 ⁇ .
  • Phosphatidylinositol 3-kinase-alpha (PBK ⁇ ), a dual specificity lipid and protein kinase, is composed of an 85 kDa regulatory subunit and a 110 kDa catalytic subunit.
  • the protein includes a catalytic subunit, which uses ATP to phosphorylate Ptdlns, PtdIns(4)P and
  • PtdIns(4,5)P 2 PtdIns(4,5)P 2 .
  • PTEN a tumor suppressor
  • PIP3 phosphatidylinositol (3,4,5)- trisphosphate
  • PIP3 protein kinase B
  • AKTl protein kinase B
  • PBK ⁇ has been implicated in the control of cytoskeletal reorganization, apoptosis, vesicular trafficking and proliferation and differentiation processes.
  • Increased copy number and expression of the pi 10a gene is associated with a number of cancers such as ovarian cancer, cervical cancer, breast cancer, colon cancer, rectal cancer, endometrial cancer, stomach cancer, liver cancer, lung cancer, thyroid cancer, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), and glioblastomas.
  • Mammalian target of rapamycin is a serine/threonine kinase of approximately 289 kDa in size and a member of the evolutionary conserved eukaryotic TOR kinases.
  • the mTOR protein is a member of the PIKK family of proteins due to its C-terminal homology (catalytic domain) with PB-kinase and the other family members, e.g. DNA dependent protein kinase (DNA-PKcs), Ataxia-telangiectasia mutated (ATM).
  • DNA-PKcs DNA dependent protein kinase
  • ATM Ataxia-telangiectasia mutated
  • mTOR kinase is a central regulator of cell growth and survival by mediating multiple important cellular functions including translation, cell cycle regulation, cytoskeleton reorganization, apoptosis and autophagy.
  • mTOR resides in two biochemically and functionally distinct complexes that are conserved from yeast to human.
  • rapamycin sensitive mTOR-Raptor complex regulates translation by activation of p70S6 kinase and inhibition of eIF4E binding protein 4EBP 1 through phosphorylation, which is the best-described physiological function of mTOR signaling.
  • mTORCl activity is regulated by extracellular signals (growth factors and hormones) through the PBK/ AKT pathway, and by nutrient availability, intracellular energy status and oxygen through the regulators like LKBl and AMPK. Rapamycin and its analogues inhibit mTORCl activity by disrupting the interaction between mTOR and raptor.
  • the rapamycin-insensitive complex, mT0RC2 was discovered only recently.
  • the mT0RC2 complex contains other proteins including Rictor and mSinl. mT0RC2 phosphorylates AKT at the hydrophobic Ser473 site, and appears to be essential for AKT activity. Other substrates of mT0RC2 include PKCA: and SGKl. How mT0RC2 activity is regulated is not well understood.
  • the mTORCl pathway can be activated by elevated PBK/ AKT signaling or mutations in the tumor suppressor genes PTEN or TSC2, providing cells with a growth advantage by promoting protein synthesis.
  • Cancer cells treated with the mTORCl inhibitor rapamycin show growth inhibition and, in some cases, apoptosis.
  • Three rapamycin analogues, CCI-779 (Wyeth), RADOOl (Novartis) and AP23573 (Ariad) are in clinical trials for the treatment of cancer. However response rates vary among cancer types from a low of less than 10% in patients with glioblastoma and breast cancer to a high of around 40% in patients with mantle cell lymphoma.
  • rapamycin can actually induce a strong AKT phosphorylation in tumors by attenuating the feedback inhibition on receptor tyrosine kinases mediated by p70S6K, one of the downstream effectors of mTORCl.
  • p70S6K receptor tyrosine kinases mediated by p70S6K
  • mTORCl inhibition-induced phospho-AKT leads to increased cancer cell survival and acquisition of additional lesions, this could counteract the effects of growth inhibition by rapamycin analogues and explain the variable response rate.
  • the present invention provides PIKK inhibitors, particularly PI3K ⁇ and mTOR inhibitors, which are useful for treating PI3K ⁇ and mTOR mediated diseases and conditions.
  • R 1 is hydrogen or alkyl
  • R 2 is methyl or ethyl
  • Z 2 is -N- or -CR 4 - where R 4 is R d , -(alkylene)heterocycloalkyl,
  • Z 3 is -N- or -CR 5 - where R 5 is R d , -(alkylene) n aryl, -(alkylene)-heteroaryl, -(alkylene)heterocycloalkyl, -(alkylene) n O-(alkylene) n aryl, -(alkylene) n N(R a )(alkylene) n aryl, -(alkylene) n N(R a )(alkylene) n heteroaryl, or -(alkylene) n O(alkylene) n heteroaryl provided that only two of Z 1 , Z 2 and Z 3 can simultaneously be -N-; or when Z 2 is -CR 4 - and Z 3 is -CR 5 - then R 4 and R 5 together with the atoms to which they are attached can form ring A which is phenyl or a 5 or 6 membered heteroaryl ring and ring A is substitute
  • Z 4 is -N- or -C-; provided that when Z 5 is -CR 6 - where R 6 together with Z 6 forms phenyl, and Z 7 is -N-, then Z 4 is -C-;
  • Z 5 , Z 6 or Z 7 are each independently selected from -N- or -CR 6 - provided at least one of Z 4 , Z 5 , Z 6 and Z 7 is -N- where R 6 is R d ; or R 6 together with the adjacent ring atom can form phenyl or 5 or six membered heteroaryl ring wherein the phenyl or heteroaryl ring is substituted with R 1 , R J , or R k where R 1 , R J , or R k are independently R d , -(alkylene) n aryl, - (alkylene)heteroaryl, -( alkylene)heterocycloalkyl, -(alkylene) n O(alkylene) n aryl, - (alkylene) n N(R a )(alkylene) n aryl, -(alkylene 2 ) n N(R a )( alkylene) n heteroaryl,
  • each R a is independently hydrogen or R b ; or when two R a are attached to a nitrogen atom, either alone or part of another group, the two R a s together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring with is optionally substituted with one, two or three substitutents independently selected from oxo, halo, alkyl, alkenyl, alkynyl, cyano, nitro, alkylcarbonyl, carboxy, alkoxycarbonyl, hydroxyl, alkoxy, alkonyloxy, alkylthio, alkylsulfonyl, aminosulfonyl, sulfonylamino, amino, alkylamino, or dialkylamino; each R b is independently alkyl, cycloalkyl, phenyl, heteroaryl, or benzyl wherein the
  • this invention is directed to a method of treating PBK mediated diseases and disorders in a patient in need thereof which method comprises administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof.
  • the PBKalpha and/or mTOR mediated disease such as cancer.
  • this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient.
  • the present invention is directed to the use of a compound of
  • the present invention is directed to compound of Formula (I) for use in therapy.
  • the therapy is cancer.
  • Cancers which may be treated with compounds of the present invention include, without limitation, carcinomas such as cancer of the bladder, breast, colon, rectum, kidney, liver, lung (small cell lung cancer, and non-small-cell lung cancer), esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia, acute lymphocitic leukemia, chronic lyelogenous leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T- cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid lineage
  • Alkyl means a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl (including all isomeric forms), pentyl (including all isomeric forms), and the like.
  • Alkenyl means a linear monovalent hydrocarbon radical of two to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbon atoms containing a double bond, e.g., ethenyl, propenyl, 2-propenyl, butenyl (including all isomeric forms), pentyl (including all isomeric forms), and the like.
  • Alkynyl means a linear monovalent hydrocarbon radical of two to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbon atoms containing a triple bond, e.g., ethynyl, propynyl, 2-propynyl, butynyl (including all isomeric forms), pentyl (including all isomeric forms), and the like.
  • Alkylene means a linear saturated divalent hydrocarbon radical of one to six carbon atoms or a branched saturated divalent hydrocarbon radical of three to six carbon atoms unless otherwise stated e.g., methylene, ethylene, propylene, 1-methylpropylene, 2-methylpropylene, butylene, pentylene, and the like.
  • Amino means a -NH 2 .
  • Alkylamino means a -NHR radical where R is alkyl as defined above, e.g., methylamino, ethylamino, propylamino, or 2-propylamino, and the like.
  • Alkoxy means a -OR radical where R is alkyl as defined above, e.g., methoxy, ethoxy, propoxy, or 2-propoxy, n-, iso-, or tert-butoxy, and the like.
  • Alkylthio means a -SR radical where R is alkyl as defined above, e.g., methylthio, and the like.
  • Alkylsulfonyl means a -SO 2 R radical where R is alkyl as defined above, e.g., methylsulfonyl, ethylsulfonyl, and the like.
  • Alkoxycarbonyl means a -C(O)OR radical where R is alkyl as defined above, e.g., methoxycarbonyl, ethoxycarbonyl, and the like.
  • Alkylcarbonylamino means a -NHC(O)R radical where R is alkyl as defined above, e.g., methylcarbonylamino, ethylcarbonylamino, and the like.
  • Alkonyloxy means a -OC(O) R radical where R is alkyl as defined above, e.g., acetyloxy, and the like.
  • Alkoxyalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two alkoxy groups, as defined above, e.g., 2-methoxyethyl, 1-, 2-, or 3-methoxypropyl, 2-ethoxyethyl, and the like.
  • Alkoxyalkyloxy or "alkoxyalkoxy” means a -OR radical where R is alkoxyalkyl as defined above, e.g., methoxyethoxy, 2-ethoxyethoxy, and the like.
  • Aminoalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two, -NRR' where R is hydrogen, alkyl, or -COR a where R a is alkyl, each as defined above, and R' is selected from hydrogen, alkyl, hydroxyalkyl, alkoxyalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or haloalkyl, each as defined herein, e.g., aminomethyl, methylaminoethyl, 2-ethylamino-2-methylethyl, 1,3-diaminopropyl, dimethylaminomethyl, diethylaminoethyl, acetylaminopropyl, and the like.
  • Aminoalkoxy means a -OR radical where R is aminoalkyl as defined above, e.g., 2- aminoethoxy, 2-dimethylaminopropoxy, and the like.
  • Aminosulfonyl means a -SO 2 NRR' radical where R and R' are independently hydrogen or alkyl as defined above, e.g., methylaminosulfonyl, aminosulfonyl, and the like.
  • Acyl means a -COR radical where R is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heterocyclyl, or heterocyclylalkyl, each as defined herein, e.g., acetyl, propionyl, benzoyl, pyridinylcarbonyl, and the like.
  • R is alkyl
  • the radical is also referred to herein as alkylcarbonyl.
  • Aryl means a monovalent monocyclic or bicyclic aromatic hydrocarbon radical of 6 to
  • Alkyl means a -(alkylene)-R radical where R is aryl as defined above.
  • Cycloalkyl means a cyclic saturated monovalent hydrocarbon radical of three to ten carbon atoms, e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, and the like.
  • Cycloalkylalkyl means a -(alkylene)-R radical where R is cycloalkyl as defined above; e.g., cyclopropylmethyl, cyclobutylmethyl, cyclopentylethyl, or cyclohexylmethyl, and the like.
  • Carboxy means -COOH.
  • Dialkylamino means a -NRR' radical where R and R' are independently alkyl as defined herein, e.g., dimethylamino, and the like.
  • Halo means fluoro, chloro, bromo, or iodo, preferably fluoro or chloro.
  • Haloalkyl means alkyl radical as defined above, which is substituted with one or more halogen atoms, preferably one to five halogen atoms, preferably fluorine or chlorine, including those substituted with different halogens, e.g., -CH 2 Cl, -CF 3 , -CHF 2 , -CH 2 CF 3 , -CF 2 CF 3 , - CF(CH 3 ) 2 , and the like.
  • fluoroalkyl When the alkyl is substituted with only fluoro, it is referred to in this Application as fluoroalkyl.
  • Haloalkoxy means a -OR radical where R is haloalkyl as defined above e.g., -OCF 3 , - OCHF 2 , and the like.
  • R is haloalkyl where the alkyl is substituted with only fluoro, it is referred to in this Application as fluoroalkoxy.
  • Hydroalkyl means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom.
  • Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxy ethyl, 2-hydroxypropyl, 3-hydroxypropyl, l-(hydroxymethyl)-2- methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxybutyl, 2,3-dihydroxypropyl, 1- (hydroxymethyl)-2-hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2- (hydroxymethyl)-3-hydroxypropyl, preferably 2-hydroxyethyl, 2,3-dihydroxypropyl, and 1- (hydroxymethyl)-2-hydroxyethyl.
  • Hydroalkoxy or "hydroxyalkyloxy” means a -OR radical where R is hydroxyalkyl as defined above.
  • Heterocyclyl or “heterocycloalkyl” means a saturated or unsaturated monovalent monocyclic group of 4 to 8 ring atoms in which one or two ring atoms are heteroatom selected from N, O, or S(O) n , where n is an integer from 0 to 2, the remaining ring atoms being C.
  • the heterocyclyl ring is optionally fused to a (one) aryl or heteroaryl ring as defined herein provided the aryl and heteroaryl rings are monocyclic.
  • heterocyclyl ring fused to monocyclic aryl or heteroaryl ring is also referred to in this Application as "bicyclic heterocyclyl" ring and is a subset of fused heterocyclyl. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a -CO- group. More specifically the term heterocyclyl includes, but is not limited to, pyrrolidino, piperidino, homopiperidino, 2-oxopyrrolidinyl, 2-oxopiperidinyl, morpholino, piperazino, tetrahydropyranyl, thiomorpholino, and the like.
  • heterocyclyl ring When the heterocyclyl ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic.
  • heterocyclyl group contains at least one nitrogen atom, it is also referred to herein as heterocycloamino and is a subset of the heterocyclyl group.
  • heterocyclyl group When the heterocyclyl group is a saturated ring and is not fused to aryl or heteroaryl ring as stated above, it is also referred to herein as saturated monocyclic heterocyclyl.
  • each R x is hydrogen or R y and R y is independently hydrogen, alkyl, cycloalkyl, phenyl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl group is substituted with one, two or three substitutents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino, or dialkylamino and Y is aryl, heteroaryl, or heterocyloalkyl substituted with 0, 1, or 2 substitutents independently selected from alkyl, halo, haloalkoxy, hydroxyl, haloalkyl -CN
  • Heterocyclylalkyl means a -(alkylene)-R radical where R is heterocyclyl ring as defined above e.g., tetraydrofuranylmethyl, piperazinylmethyl, morpholinylethyl, and the like.
  • Heteroaryl means a monovalent monocyclic or bicyclic aromatic radical of 5 to 10 ring atoms where one or more, preferably one, two, or three, ring atoms are heteroatom selected from N, O, or S, the remaining ring atoms being carbon.
  • Representative examples include, but are not limited to, pyrrolyl, thienyl, thiazolyl, imidazolyl, furanyl, indolyl, isoindolyl, oxazolyl, isoxazolyl, benzothiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl, and the like.
  • each R x is hydrogen or R y and R y is independently hydrogen, alkyl, cycloalkyl, phenyl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl group is substituted with one, two or three substitutents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino, or dialkylamino and Y is aryl, heteroaryl, or heterocyloalkyl substituted with 0, 1, or 2 substitutents independently selected from alkyl, halo, haloalkoxy, hydroxyl,
  • Heteroaralkyl means a -(alkylene)-R radical where R is heteroaryl as defined above.
  • heterocyclyl group optionally substituted with an alkyl group means that the alkyl may but need not be present, and the description includes situations where the heterocyclyl group is substituted with an alkyl group and situations where the heterocyclyl group is not substituted with alkyl.
  • a “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound.
  • Such salts include: acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as formic acid, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4- hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesul
  • pharmaceutically acceptable means that the referenced substance, such as a compound of Formula I or a salt of a compound of Formula lor a formulation containing a compound of Formula I or a particular excipent, are suitable for administration to a patient.
  • excipient means any pharmaceutically acceptable additive, carrier, diluent, adjuvant, or other ingredient, other than the active pharmaceutical ingredient (API), which is typically included for formulation and/or administration to a patient.
  • API active pharmaceutical ingredient
  • Treating" or “treatment” of a disease includes: preventing the disease, i.e. causing the clinical symptoms of the disease not to develop in a mammal that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease; inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms; or relieving the disease, i.e., causing regression of the disease or its clinical symptoms.
  • a “therapeutically effective amount” means the amount of a compound of Formula (I) that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease. The “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated.
  • prodrug means compounds that are transformed in vivo to yield a compound of the present invention. The transformation may occur by various mechanisms, such as through hydrolysis in blood.
  • a discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems,” Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
  • a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the add group with a group such as (Ci-C 8 alkyl, (C 2 -C I 2 )alkanoyloxymethyl, 1- (alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1 -methyl- l-(alkanoyloxy)ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1- (alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1 -methyl- l-(alkoxycarbonyloxy)- ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, l-(N-(alkoxycarbonyl)aminomethyl having from 4 to 10 carbon atom
  • a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as (Ci-C 6 )alkanoyloxymethyl, l-((Ci-C 6 )alkanoyloxy)ethyl, 1-methyl-l- ((Ci-C 6 )alkanoyloxy)ethyl, (Ci-C 6 )alkoxycarbonyloxymethyl, N-(Ci-Ce)alkoxycarbonyl- aminomethyl, succinoyl, (Ci-Ce)alkanoyl, ⁇ -amino(Ci-C 4 )alkanoyl, arylacyl and ⁇ -aminoacyl, or ⁇ -aminoacyl- ⁇ -aminoacyl, where each ⁇ -aminoacyl group is independently selected from the naturally occurring L-amino acids, -P(O)(OH) 2 ,
  • the compounds of the present invention may contain asymmetric or chiral centers, and therefore, exist in different stereoisomeric forms. It is contemplated that all stereoisomeric forms of the compounds as well as mixtures thereof, including racemic mixtures, form part of the present invention. In addition, the present invention contemplates all geometric and positional isomers. For example, if the compound contains a double bond, both the cis and trans forms (designated as Z and E, respectively), as well as mixtures, are contemplated. Mixture of stereoisomers, such as diastereomeric mixtures, can be separated into their individual stereochemical components on the basis of their physical chemical differences by known methods such as chromatography and/or fractional crystallization.
  • Enantiomers can also be separated by converting the enantiomeric mixture into a diasteromeric mixture by reaction with an appropriate optically active compound (e.g., an alcohol), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Also, some compounds may be atropisomers (e.g., substituted biaryls).
  • an appropriate optically active compound e.g., an alcohol
  • separating the diastereomers e.g., converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers.
  • some compounds may be atropisomers (e.g., substituted biaryls).
  • the compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water (hydrate), ethanol, and the like.
  • pharmaceutically acceptable solvents such as water (hydrate), ethanol, and the like.
  • the present invention contemplates and encompasses both the solvated and unsolvated forms.
  • compounds of the present invention may exist in different tautomeric forms. All tautomers of compounds of the present invention are contemplated. For example, all of the tautomeric forms of the imidazole moiety are included in this invention. Also, for example, all keto-enol or imine-enamine forms of the compounds are included in this invention.
  • the present invention also includes isotopically-labelled compounds, which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as 2 H, 3 H, 13 C, 14 C, 15 N, 16 O, 17 0, 31 P, 32 P, 35 S, 18 F, and 36 Cl.
  • isotopically-labelled compounds of the present invention for example those into which radioactive isotopes such as 3 H and 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3 H, and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detection.
  • Isotopically labelled compounds of this invention can generally be prepared by substituting a readily available isotopically labelled reagent for a non- isotopically labelled reagent.
  • LG generally refer to groups that are displaceable by a nucleophile.
  • Such leaving groups are known in the art.
  • Examples of leaving groups include, but are not limited to, halides (e.g., I, Br, F, Cl), sulfonates (e.g., mesylate, tosylate), sulfides (e.g., SCH 3 ), N-hydroxsuccinimide, N-hydroxybenzotriazole, and the like.
  • nucleophiles include, but are not limited to, amines, thiols, alcohols, Grignard reagents, anionic species (e.g., alkoxides, amides, carbanions) and the like.
  • the compounds of Formula (Ia) are those wherein:
  • R 1 is hydrogen and 1S a ring of formula
  • R is methyl and R a is hydrogen, alkyl, or cyanomethyl, preferably hydrogen or methyl.
  • the compounds of Formula (Ia) are those wherein:
  • R 1 is hydrogen and is a ring of formula:
  • the compounds of Formula (Ia) and in embodiments A, B, C above and groups contained therein, the compounds of Formula (Ia) are those wherein R 1 is hydrogen and Ar 1 is heteroaryl substituted as defined in the Summary.
  • Ar 1 is pyrazolyl, preferably pyrazol-3-yl or pyrazol-5-yl, substituted as defined in the Summary.
  • Ar 1 is pyridyl, benzoxazolyl, pyrimidinyl, indazolyl, indolyl, benzthiazolyl, pyrazinyl, pyridazinyl, isobenzoxazolyl, or isobenzthiazolyl, each ring substituted as defined in the Summary.
  • the compounds of Formula (Ia) and in embodiments A, B, C and D, above and groups contained therein, the compounds of Formula (Ia) are those wherein
  • R 6 is hydrogen, alkyl, substituted alkyl, carboxy, OR a , halo, haloalkyl, heterocycloalkyl, -(alkylene)heterocycloalkyl, -( alkylene) n O(alkylene) n aryl, -( alkylene) n N(R a )(alkylene c ) n aryl, -(alkylene) n N(R a )(alkylene) n C 5 _ 8 heteroaryl, alkenyl, -C0NR a R a or -NR a R a .
  • R J is hydrogen or alkyl and R k is heteroaryl or aryl.
  • the compounds of Formula (I) are those wherein:
  • R 1 is hydrogen, 1-piperazinyl, 4-methyl-l-piperazinyl, 4-(l-methylethyl)-l- piperazinyl, 4-(2-methoxy ethyl)- 1-piperazinyl, fluoro, 3-(methylsulfonyl)phenyl, 4-pyridinyl, 2-methoxyethoxy, 4-morpholinyl, 2-(l-pyrrolidinyl)ethoxy, l-tert-butoxycarbonyl-3,6- dihydro-l(2H)-pyridin-4-yl, 3-methoxyphenyl, 3-fluoro-4-methylaminocarbonylphenyl, A- acetylaminomethylphenyl, l-(2-methoxy ethyl)- lH-pyrazol-4-yl, 2-fluorophenyl, 2- methylphenyl, 2-methoxy-3-pyridinyl, 3-furanyl, 3,5-dimethyl
  • R 1 is hydrogen, 6-piperazin-l- yl, 8-fluoro, 6-(2-methoxyethoxy)-, 6-morpholin-4-yl-, 6-(3-methoxyphenyl)-, 6-(3-fluoro-4- methylaminocarbonylphenyl)-, 6-(4-methylcarbonylaminomethylphenyl)-, 6-( 1 -(2- methoxyethylpyrazol-4-yl)-, 6-(2-fluorophenyl)-, 6-(2-methylphenyl)-, 6-(2-methoxypyridin-3- yl)-, 6-furan-3-yl, 6-(3,5-isoxazol-4-yl)-, 6-(4-fluoro-2-methylphenyl)-, 6-(3-methylisoxazol-5- yl)-, 6-(3-acetylaminophenyl)-, 6-(3-cyclopropylcarbonylaminophenyl
  • Ar 1 is aryl, preferably phenyl, substituted as defined above.
  • Ar 1 is heteroaryl, preferably pyrazol-3-yl or pyrazol-5-yl substituted as defined above.
  • in one group of compounds Ar 1 is heterocycloalkyl substituted as defined above.
  • Ar 1 is 5-fluoro-6-methoxy-3-pyridinyl, lH-pyrazol-3-yl, 1 -methyl- lH-pyrazol-3-yl, 4-(3-ethylureido)phenyl, lH-pyrrolo[2,3- b]pyridin-6-yl, 6-(trifluoromethyl)-2-pyridinyl, 2-pyridinyl, lH-pyrrolo[3,2-b]pyridine-5-yl, 4- hydroxypyridin-2-yl, 4-(3-phenylureido)phenyl, 5-(3-ethylureido)pyridine-2-yl, 4-(3- methylureido)phenyl, 3-hydroxyphenyl, 4-acetylaminophenyl, 4-(3-(4-pyridinyl)ureidophenyl, 3-amino-pyrimidin-5-yl, 4-pyridazinyl
  • Ar 1 is 5-fluoro-6- methoxy-3-pyridinyl, 1 H-pyrazol-3 -yl, 1 -methyl- 1 H-pyrazol-3 -yl, 4-(3-ethylureido)phenyl, 4- (3-phenylureido)phenyl, 4-(3-methylureido)phenyl, 3-acetylaminophenyl, 1- (phenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2-methylphenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2-fluorophenyl-aminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(4-cyanophenylaminocarbonyl)- 1 H- pyrazol-3-yl, 1 -(2 -trifluoromethylphenylaminocarbonyl)- 1 H-pyrazol-3 -yl,
  • Z 1 is -N-
  • Z 2 is -N- or -CR 4 - where R 4 is R d , - (alkylene)heterocycloalkyl, -(alkylene) n O(alkylene) n aryl, -(alkylene) n N(R a )(alkylene) n aryl, -( alkylene) n -N(R a )-(alkylene) n heteroaryl, or -( alkylene) n O(alkylene) n heteroaryl;
  • Z 3 is -N- or -CR 5 - where R 5 is R d , -(alkylene) n aryl, -(alkylene)-heteroaryl,
  • Z 1 is -N-
  • Z 2 is -CH-
  • Z 3 is -CR 5 - where R 5 is R d , -(alkylene) n aryl, -(alkylene)-heteroaryl, -(alkylene)heterocycloalkyl, -(alkylene) n O-(alkylene) n aryl, -(alkylene) n N(R a )(alkylene) n aryl, -(alkylene) n N(R a )(alkylene) n heteroaryl, or -(alkylene) n O(alkylene) n heteroaryl.
  • Z 1 is -N-
  • Z 2 is -CH-
  • ethoxy-4-pyrimidinyl is 4-amino-6-methyl-l,3,5-triazin-2-yl,
  • Representative compounds of Formula (I) are:
  • the starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Bachem (Torrance, Calif), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March's Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition) and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989).
  • the reactions described herein take place at atmospheric pressure over a temperature range from about -78 0 C to about 150 0 C, more preferably from about 0 0 C to about 125 0 C and most preferably at about room (or ambient) temperature, e.g., about 20 0 C.
  • Compounds of Formula (I) where R 6 is ethoxycarbonyl can be prepared by reacting the compound of formula 6 with potassium (Z)-2-cyano-l-ethoxycarbonyl-ethenolate in the presence of an inorganic acid such as sulfuric acid, and the like.
  • the compound of Formula (I) can be converted to other compounds of Formula (I).
  • hydrolysis of the ester group in a compound of Formula (I) where R 6 is ethoxycarbonyl provides the corresponding compound of Formula (I) where R 6 is carboxy .
  • the carboxy group can be coverted to - CONR a R a by methods well known in the art.
  • the compounds of the present invention are useful for the treatment of PBK and/or mTOR mediated diseases and disorders such as cancer.
  • Cancers which may be treated with compounds of the present invention include, without limitation, carcinomas such as cancer of the bladder, breast, colon, rectum, kidney, liver, lung (small cell lung cancer, and non-small- cell lung cancer), esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia, acute lymphocitic leukemia, chronic lyelogenous leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non- Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid
  • the compounds of the present invention can also be used to treat hyperproliferative disorders such as thyroid hyperplasia (especially Grave's disease), and cysts (such as hypervascularity of ovarian stroma, characteristic of polycystic ovarian syndrome (Stein- Leventhal syndrome)).
  • the compounds of the present invention can also be used to treat the following diseases or conditions: asthma, chronic obstructive pulmonary disease (COPD), emphysema, psoriasis, contact dermatitis, conjunctivitis, allergic rhinitis, systemic lupus erythematosus (SLE), ulcerative colitis, Crohn's disease, multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, Alzheimer's disease, atheroscleosis and Huntington's diesese.
  • COPD chronic obstructive pulmonary disease
  • emphysema psoriasis
  • contact dermatitis conjunctivitis
  • allergic rhinitis allergic rhinitis
  • systemic lupus erythematosus (SLE) systemic lupus erythematosus
  • the compounds of the present invention can be administered alone, in combination with other compounds of the present invention, or with other pharmaceutically active compounds.
  • the other pharmaceutically active compounds can be intended to treat the same disease or condition as the compounds of the present invention or a different disease or condition. If the patient is to receive or is receiving multiple pharmaceutically active compounds, the compounds can be administered simultaneously, or sequentially.
  • the active compounds may be found in one tablet or in separate tablets, which can be administered at once or sequentially in any order.
  • the compositions may be different forms. For example, one or more compound may be delivered via a tablet, while another is administered via injection or orally as a syrup. All combinations, delivery methods and administration sequences are contemplated.
  • kits comprises two separate pharmaceutical compositions: a compound of the present invention, and a second pharmaceutical compound.
  • the kit comprises a container for containing the separate compositions such as a divided bottle or a divided foil packet. Additional examples of containers include syringes, boxes and bags.
  • the kit comprises directions for the use of the separate components.
  • the kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician or veterinarian.
  • Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of the tablets or capsules to be packed. Next, the tablets or capsules are placed in the recesses and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a result, the tablets or capsules are sealed in the recesses between the plastic foil and the sheet. Preferably the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.
  • a memory aid on the kit, e.g., in the form of numbers next to the tablets or capsules whereby the numbers correspond with the days of the regimen which the tablets or capsules so specified should be ingested.
  • a memory aid is a calendar printed on the card, e.g., as follows "First Week, Monday, Tuesday, . . . etc . . . Second Week, Monday, Tuesday, . . . " etc.
  • a “daily dose” can be a single tablet or capsule or several pills or capsules to be taken on a given day.
  • a daily dose of a compound of the present invention can consist of one tablet or capsule, while a daily dose of the second compound can consist of several tablets or capsules and vice versa.
  • the memory aid should reflect this and aid in correct administration of the active agents.
  • a dispenser designed to dispense the daily doses one at a time in the order of their intended use.
  • the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen.
  • a memory-aid is a mechanical counter which indicates the number of daily doses that has been dispensed.
  • a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
  • the compounds of the present invention and other pharmaceutically active agents can be administered to a patient either orally, rectally, parenterally, (for example, intravenously, intramuscularly, or subcutaneously) intracisternally, intravaginally, intraperitoneally, intravesically, locally (for example, powders, ointments or drops), or as a buccal or nasal spray. All methods that are used by those skilled in the art to administer a pharmaceutically active agent are contemplated.
  • compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions, or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions.
  • suitable aqueous and nonaqueous carriers, diluents, solvents, or vehicles include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents.
  • adjuvants such as preserving, wetting, emulsifying, and dispersing agents.
  • Microorganism contamination can be prevented by adding various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like.
  • Prolonged absorption of injectable pharmaceutical compositions can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Solid dosage forms for oral administration include capsules, tablets, powders, and granules.
  • the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or
  • fillers or extenders as for example, starches, lactose, sucrose, mannitol, and silicic acid;
  • binders as for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia;
  • humectants as for example, glycerol;
  • disintegrating agents as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate;
  • solution retarders as for example, paraffin;
  • absorption accelerators as for example, quaternary ammonium compounds;
  • wetting agents as for example, cetyl alcohol and glycerol monostearate;
  • wetting agents as for example, cetyl alcohol and glycerol monostearate
  • compositions of a similar type may also be used as fillers in soft and hard filled gelatin capsules using such excipients as lactose or milk sugar, as well as high molecular weight polyethylene glycols, and the like.
  • Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may also contain opacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedding compositions that can be used are polymeric substances and waxes. The active compounds can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs.
  • the liquid dosage form may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsif ⁇ ers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, and sesame seed oil, glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, or mixtures of these substances, and the like.
  • inert diluents commonly used in the art, such as water or other solvents, solubil
  • the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions in addition to the active compound, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, or mixtures of these substances, and the like.
  • compositions for rectal administration are preferable suppositories, which can be prepared by mixing the compounds of the present invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax, which are solid at ordinary room temperature, but liquid at body temperature, and therefore, melt in the rectum or vaginal cavity and release the active component.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax, which are solid at ordinary room temperature, but liquid at body temperature, and therefore, melt in the rectum or vaginal cavity and release the active component.
  • Dosage forms for topical administration of a compound of the present invention include ointments, powders, sprays and inhalants.
  • the active compound or fit compounds are admixed under sterile condition with a physiologically acceptable carrier, and any preservatives, buffers, or propellants that may be required.
  • Opthalmic formulations, eye ointments, powders, and solutions are also contemplated as being within the scope of this invention.
  • the compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 to about 3,000 mg per day.
  • dosage levels in the range of about 0.1 to about 3,000 mg per day.
  • a dosage in the range of about 0.01 to about 100 mg per kilogram body weight is typically sufficient.
  • the specific dosage and dosage range that can be used depends on a number of factors, including the requirements of the patient, the severity of the condition or disease being treated, and the pharmacological activity of the compound being administered. The determination of dosage ranges and optimal dosages for a particular patient is within the ordinary skill in the art.
  • the compounds of Formula (I) or a pharmaceutically acceptable salt thereof may also be administered in combination with one or more additional pharmaceutically active compounds/agents.
  • the additional pharmaceutically active agent is an agent that can be used to treat a cancer.
  • an additional pharmaceutically active agent can be selected from antineoplastic agents, anti-angiogenic agents, chemotherapeutic agents and peptidal cancer therapy agents.
  • the antineoplastic agents are selected from antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents, kinase inhibitors, miscellaneous agents and combinations thereof.
  • the additional pharmaceutically active compounds/agents may be a traditional small organic chemical molecules or can be macromolecules such as a proteins, antibodies, peptibodies, DNA, RNA or fragments of such macromolecules.
  • specific pharmaceutically active agents that can be used in the treatment of cancers and that can be used in combination with one or more compound of the present invention include: methotrexate; tamoxifen; fluorouracil; 5-fluorouracil; hydroxyurea; mercaptopurine; cisplatin; carboplatin; daunorubicin; doxorubicin; etoposide; vinblastine; vincristine; paclitaxel; thioguanine; idarubicin; dactinomycin; imatinib; gemcitabine; altretamine; asparaginase; bleomycin; capecitabine; carmustine; cladibrine; cyclophosphamine; cytarabine; de
  • Additional pharmaceutically active agents that can be used in the treatment of cancers and that can be used in combination with one or more compound of the present invention include: epoetin alfa; darbepoetin alfa; panitumumab; pegf ⁇ lgrastim; palifermin; filgrastim; denosumab; ancestim; AMG 102; AMG 386; AMG 479; AMG 655; AMG 745; AMG 951; and AMG 706, or a pharmaceutically acceptable salt thereof.
  • the compounds of the present invention can also be used in combination with pharmaceutically active agents that treat nausea. Examples of agents that can be used to treat nausea include: dronabinol; granisetron; metoclopramide; ondansetron; and prochlorperazine; or a pharmaceutically acceptable salt thereof.
  • the compounds of the present invention can be used in combination with other agents that can be used to treat cancer such as acemannan; aclarubicin; aldesleukin; alitretinoin; amifostine; amrubicin; amsacrine; anagrelide; arglabin; arsenic trioxide; BAM 002
  • the compounds of the present invention may also be used in combination with radiation therapy, hormone therapy, surgery and immunotherapy, which therapies are well know to those skilled in the art.
  • the gradient was followed by a 2 min return to 40% solvent B; or (B) Using a Waters 20 x 50 mm column at 20 mL/min or Phenomenex Gemni 5 ⁇ Cl 8 100 x 30mm (Phenomenex, Torrance, CA).
  • the mobile phase used was a mixture of solvent A (H 2 O/0.1% TFA) and solvent B (ACN/0.1% TFA) with a >10 min gradient from 5% to 100% solvent B.
  • the gradient is followed by a 2 min return to 5% ACN.
  • Mass Spectra Unless otherwise indicated, all mass spectral data for starting materials, intermediates and/or exemplary compounds are reported as mass/charge (m/z), having an (M+H + ) or (M-H " ) molecular ion, depending on the inonization mode (positive or negative). The molecular ion reported was obtained by electrospray detection method. Compounds having an isotopic atom, such as bromine and the like, are reported according to the detected isotopic pattern, as appreciated by those skilled in the art.
  • Percents of solid reagents specified are percent by weight with respect to the total weight, and percents of solvents are specified by percent by volume with respect to the total volume, unless indicated otherwise.
  • Run 2 1.027 g. 2.5 mL ammonium hydroxide.
  • Run 3 1.034 g, 2.5 mL ammonium hydroxide.
  • Run 4 1.118 g, 2.6 mL ammonium hydroxide.
  • Run 5 1.117 g, 2.5 mL ammonium hydroxide.
  • Run 6 1.149 g, 2.7 mL ammonium hydroxide.
  • Run 7 1.264 g, 2.6 mL ammonium hydroxide.
  • Run 8 1.106 g, 2.6 mL ammonium hydroxide.
  • Run 9 1.075 g, 2.7 mL ammonium hydroxide.
  • 6-Chloro-2-methylpyrimidine-4,5-diamine (8.89 g, 56.1 mmol, the material from Step 1) was suspended in ethyl ortho formate (100 mL, 601 mmol) in a flask fitted with a reflux condenser and placed in a preheated oil bath (100 0 C) and stirred for 75 minutes. The reaction mixture was cooled to room temperature, concentrated, treated with hexanes and filtered. The solid washed with hexanes and dried to give 6-chloro-2-methyl-9H-purine. MS (ESI pos. ion) m/z: 169. Calcd: 168. Step 3
  • 6-Chloro-2-methyl-9H-purine (9.45 g, 56 mmol, the material from Step 2) was suspended in DCM (100 mL) and p-toluenesulfonic acid (Acros Organics, Geel, Belgium, 12% in acetic acid, 0.90 mL, 5.6 mmol) and 2,3-dihydropyran (6.6 mL, 73 mmol) were added.
  • the reaction flask was fitted with a reflux condenser and placed in a preheated oil bath (50 0 C) and stirred under nitrogen for 30 minutes. The reaction mixture was cooled to room temperature and stirred overnight. After stirring overnight, the reaction mixture was diluted with DCM and treated with saturated sodium bicarbonate (75 mL).
  • LDA was prepared by dropwise addition of n-butyllithium, 2.5 M solution in hexanes (Aldrich, St. Louis, MO) (14.7 mL, 36.8 mmol) to N,N-diisopropylamine (5.42 mL, 38.4 mmol) in THF (40 mL) cooled in an ice bath.
  • the LDA solution was cooled to -78 0 C and a solution of 4,6-dichloro-2-methylpyrimidine (Aldrich, St. Louis, MO) (5.448 g, 33.4 mmol) in THF (50 mL) was added dropwise over 1 h. A dark solution was obtained.
  • N- methyl-N-(2-pyridyl)formamide (TCI Tokyo Kasei Kogyo Co., Ltd.) (4.80 mL, 40.1 mmol) in THF (20 mL) was added dropwise to the solution at -78 0 C over 20 min. The resulting solution was stirred for 30 min and then quenched with a solution of acetic acid (2.10 mL, 36.8 mmol) in THF (20 mL) added dropwise at -78 0 C over 10 min. The solution was stirred at -78 0 C for 30 min.
  • Methylmagnesium bromide, 3M in ether (Aldrich, St. Louis, MO) (10.0 mL, 30 mmol) was added slowly to a white suspension of 2,4,6-trichloro-l,3,5-triazine (Aldrich, St. Louis, MO) (3.68 g, 20 mmol) in DCM (25.0 mL, 389 mmol) at 0 0 C and the resulting yellow suspension was warmed up to room temperature and stirring was continued until disappearance of starting material (TLC, KMnO 4 stain, 3 h). The reaction mixture was carefully quenched with NH 4 Cl(aq) at 0 0 C and then diluted with water and DCM (25.0 mL).
  • Step 1 n-Butyllithium solution, 1.6 M in hexane (0.184 niL, 2.200 mmol, Aldrich, St. Louis, MO) was added to a solution of diisopropylamine (0.314 mL, 2.200 mmol) in THF (5 mL) at 0 0 C. The reaction mixture was stirred at 0 0 C for 15 min. Tributyltin hydride (0.527 mL, 2.000 mmol, Aldrich, St. Louis, MO) was added dropwisely. The solution was stirred at 0 0 C for 15 min.
  • Cesium carbonate (0.860 mL, 10.74 mmol) was added to a mixture of 4-chloro-6- methyl-l,3,5-triazin-2-amine (1.10 g, 7.61 mmol) and l-(chloromethyl)-4-methoxybenzene (1.10 mL, 8.11 mmol) in DMF (8.0 mL) at rt. After 40 min, more l-(chloromethyl)-4- methoxybenzene (1.10 mL, 8.11 mmol) was added. After another 1 h, more cesium carbonate (0.860 mL, 10.74 mmol) was added.
  • reaction mixture was diluted with EtOAc (30 niL) and filtered through a pad of Celite® (diatomaceous earth). The filtrate was transferred to a separatory funnel, diluted with more EtOAc, and washed with water. The organic layer was dried over Na 2 SO 4 and concentrated. The resulting slurry was filtered and washed with 1 : 1 hexane-EtOAc. The filtrate was purified by chromatography on silica using 5 to 100% DCM in hexane to give the product as a soft white solid (1.8 g).
  • a glass microwave reaction vessel was charged with l-(2-methyl-6-(methylthio)- pyrimidin-4-yl)hydrazine (3 g, 18 mmol), 3-aminocrotononitrile (6 g, 70 mmol, Aldrich, St. Louis, MO) and isopropanol (10 rnL).
  • the reaction mixture was sealed and heated at 100 0 C for 15 h. After cooling to room temperature, the suspension was filtered. The filtrate was concentrated in vacuo and the residue was treated with hexane and filtered to provide a yellow solid.
  • a glass microwave reaction vessel was charged with tris(dibenzylideneacetone) dipalladium (0) (78 mg, 85 ⁇ mol, Aldrich, St. Louis, MO), 2-dicyclohexylphosphino-2,4,6,-tri- i-propyl-l,l-biphenyl (82 mg, 170 ⁇ mol, Strem Chemical, Inc., Newburyport, MA ), t-butanol (3 mL) and H 2 O (0.12 mL). Ar was bubbled in for 1 minute. The reaction mixture was sealed and heated at 110 0 C for 2 minutes and then cooled to room temperature.
  • a glass microwave reaction vessel was charged with 6-methoxy-N-(3 -methyl- 1 -(2- methyl-6-(methylsulfinyl)pyrimidin-4-yl)- 1 H-pyrazol-5 -yl)pyridin-3 -amine (100 mg, 279 ⁇ mol) dissolved in isopropanol (3 mL) and aqueous ammonium hydroxide (28-30%, 5 mL). The solution was stirred and heated at 120 0 C for 40 minutes. After cooling to room temperature, the mixture was concentrated in vacuo.
  • a glass microwave reaction vessel was charged with tris(dibenzylideneacetone) dipalladium(O) (195 mg, 212 ⁇ mol, Aldrich, St. Louis, MO), 2-dicyclohexylphosphino-2,4,6,- tri-i-propyl-l,l-biphenyl (203 mg, 0.425 mmol, Strem Chemical, Inc., Newburyport, MA ), t- butanol (5 mL) and H 2 O (0.2 mL). Ar was bubbled in for 1 minute, and then the reaction mixture was heated at 120 0 C for 10 minutes and then cooled to room temperature.
  • tris(dibenzylideneacetone) dipalladium(O) 195 mg, 212 ⁇ mol, Aldrich, St. Louis, MO
  • 2-dicyclohexylphosphino-2,4,6,- tri-i-propyl-l,l-biphenyl 203 mg, 0.425 mmol, Strem Chemical
  • Step 2 A solution of N-(3 -methyl- 1 -(2-methyl-6-(methylthio)pyrimidin-4-yl)- 1 H-pyrazol-5 - yl)-lH-indazol-4-amine (150 mg, 427 ⁇ mol) in DCM-DMF (5:1, 6 mL) was cooled to 0 0 C and treated with 3-chloroperoxybenzoic acid (147 mg, 77%, 658 ⁇ mol). The reaction mixture was stirred at 0 0 C for 1 h and at room temperature for an additional 30 minutes. The reaction was quenched with aqueous sodium thiosulfate and extracted with CH 2 Cl 2 ..
  • a glass microwave reaction vessel was charged with N-(3 -methyl- 1 -(2 -methyl-6- (methylsulf ⁇ nyl)pyrimidin-4-yl)-l H-pyrazol-5 -yl)-lH-indazol-4-amine (50 mg, 136 ⁇ mol), isopropanol and DMF (4:1, 3 mL), and NH 4 OH (5 mL, 30%).
  • the vessel was sealed and heated 120 0 C for 2 h. After cooling to room temperature, the mixture was concentrated in vacuo.
  • Step 1 A mixture of ethyl (ethoxymethylene)cyanoacetate (2.3 g, 14 mmol, Aldrich, St. Louis,
  • a glass microwave reaction vessel was charged with 5-amino-l-(2-methyl-6- (methylthio)pyrimidin-4-yl)-lH-pyrazole-4-carboxylic acid (1.4 g, 5.4 mmol) and H 2 O (10 mL).
  • the reaction mixture was stirred and heated in a Smith Synthesizer® microwave reactor (Personal Chemistry, Inc., Upssala, Sweden) at 200 0 C for 1 h.
  • the suspension was filtered to give l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-l H-pyrazol-5 -amine (1.0 g, 85% yield) as a white solid, m/z (ESI, +ve ion) 222 (M+H) + .
  • a glass reaction vessel was charged with N-(l-(2-methyl-6-(methylsulfinyl)pyrimidin- 4-yl)-l H-pyrazol-5 -yl)-lH-indazol-4-amine (0.28 g, 792 ⁇ mol) and ammonia (2 N in 2- propanol, 20 mL, 40 mmol).
  • the reaction mixture was sealed and heated at 100 0 C for 5h and then cooled to room temperature.
  • Step 1 Triethylamine (21.1 mL, 151 mmol) was added dropwise to a mixture of chloroacetic acid (13.0 g, 138 mmol, Alfa Aesar, Ward Hill, MA) and water (35 mL). After 10 minutes, 2- aminopyridine (15.5 g, 165 mmol, Fluka, Buchs, Switzerland) was added. The reaction mixture was heated at 90 0 C. After 2 h, the solution was allowed to cool to rt. EtOH (6 mL) was added and the mixture was stirred at 5 0 C for 30 min. The solution was filtered and the filtercake was washed with cold EtOH and then dried under vacuum to give a white solid (19.7 g).
  • the white solid was added to a 500-mL round-bottomed flask and suspended in toluene (140 mL). The mixture was heated to reflux and then phosphorus oxychloride (32.4 mL, 348 mmol) was added slowly over 45 min. The reaction mixture was stirred at reflux for 17 h. It was allowed to cool to rt and slowly poured into ice water (300 mL) over 15 min and then stirred for 15 min. The phases were separated and the aqueous phase was cooled to 0 0 C and neutralized with 1 N NaOH and IO N NaOH.
  • Step 2 A glass microwave reaction vessel was charged with 2-chloroimidazo[l,2-a]pyridine (0.850 g, 5.57 mmol), 1,4-dioxane (8.5 mL), ethanol (4.25 niL), 6-chloro-2-methyl-9- (tetrahydro-2H-pyran-2-yl)-9H-purine (2.11 g, 8.36 mmol), potassium carbonate (1.54 g, 11.14 mmol), triphenylphosphine (0.073 g, 0.279 mmol) and palladium(II) acetate (0.125 g, 0.557 mmol, Strem).
  • MeOH/CH 2 Cl 2 MeOH/CH 2 Cl 2 and loaded onto Si-TsOH (4 grams, Silicycle, SiliCycle Inc., Quebec City, Canda).
  • the Si-TsOH plug was eluted with 10% MeOH/CH 2 Cl 2 and then the cartridge was eluted with 10% MeOH(2 M NH 3 )/CH 2 C1 2 to elute off the product.
  • the 10% MeOH(2 M NH 3 )/CH 2 C1 2 eluent was concentrated to give 6-(2-chloroimidazo[l,2-a]pyridin-3-yl)-2- methyl-9H-purine (0.24 g, 15 %) as an off-white solid, m/z (ESI, +ve ion) 285 (M+H) + .
  • the solution was diluted with MeOH and DMSO to make it homogeneous and was purified by Prep-HPLC (Phenomenex Gemini 5 micron, C 18, 100 x 30 mm, 5 to 50% CH 3 CN(0.1% TFA)/H 2 O(0.1% TFA) over 20 min then 100% CH 3 CN(0.1% TFA) for 3 minutes at 20 mL/min) with the fractions containing product concentrated to give the title compound (0.039 g, 44 %) as an orange solid, m/z (ESI, +ve ion) 358 (M+H) + .
  • 2,7-Cichloroimidazo[l,2-a]pyridine was prepared according to preparation of 2- chloroimidazo[l,2-a]pyridine in Example 15. 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(lH- pyrazol-3-yl)imidazo[l ,2-a]pyridin-2-amine using 4-chloropyridin-2-amine. m/z (ESI, +ve ion) 187.0 (M+H) + Step 2
  • reaction was filtered and directly purified via Gilson reverse phase (0.1% TFA in CH 3 CN/H 2 O, gradient 10% to 70%) and purified by MPLC (ISCO) with DCM:MeOH+NH 4 OH 100:0 to 90:10 to afforded after freebasing with SCX column 3-(4- amino-6-methyl-l,3,5-triazin-2-yl)-7-chloro-N-(lH-pyrazol-3-yl)imidazo[l,2-a]pyridin-2- amine (2.0 mg, 5.85 ⁇ mol, 1.5 % yield), m/z (ESI, +ve ion) 342.0 (M+H) +
  • Step 5 In a 10 mL microwave sealed tube under N 2 were dissolved lH-pyrazol-3 -amine (319 mg, 3.84 mmol), 6-(2-chloroimidazo[l,2-b]pyridazin-3-yl)-2-methylpyrimidin-4-amine (200 mg, 0.767 mmol), Xantphos (89 mg, 0.153 mmol), Pd(dba) 2 (88 mg, 0.153 mmol) and cesium carbonate (1500 mg, 4.60 mmol) in 3 mL of/?-dioxane then stirred and heated at 130 0 C with microwave.
  • reaction mixture was concentrated under reduced pressure with silica and purified by MPLC (ISCO) with DCM:MeOH+NH 4 OH 100:0 to 90: 10 and then reverse phase purified with Gilson to afforded after freebasing with SCX column 3-(6-amino- 2-methylpyrimidin-4-yl)-N-(lH-pyrazol-3-yl)imidazo[l,2-b]pyridazin-2-amine as a yellow solid, m/z (ESI, +ve ion) 308.1 (M+H) + .
  • a glass microwave reaction vessel was charged with tris(dibenzylideneacetone)- dipalladium (0) (125 mg, 0.136 mmol), 2-di-tert-butylphosphino-3,4,5,6-tetramethyl-2',4',6'- triisopropyl-l,l'-biphenyl (131 mg, 0.273 mmol), tert-Butanol (6.82 mL, 1.364 mmol) , water (0.273 mL, 1.364 mmol). The vial was capped, the vacum and Ar refill process was repeated three times and the mixture was heated at 120 0 C for 10 minutes.
  • a glass microwave reaction vessel was charged with ethyl 5 -(I H-pyrazol-3 -ylamino)- l-(2-methyl-6-(methylsulfinyl)pyrimidin-4-yl)-l H-pyrazole-3 -carboxylate (43.5 mg, 0.116 mmol) and ammonia, 2M solution in 2-propanol (3.86 mL, 0.116 mmol).
  • the reaction mixture was stirred and heated in a Initiator microwave reactor (Personal Chemistry, Biotage AB, Inc., Upssala, Sweden) at 100 0 C for 2 h.
  • the syringe was rinsed with another portion of THF (0.2 mL).
  • the reaction mixture turned to a clear orange-red colored solution.
  • the mixture was maintained at rt under N 2 for 18 h.
  • the reaction mixture was poured to a sat. NH4CI solution, and extracted with EtOAc.
  • the organic extract was dried over Na 2 SO 4 .
  • the solution was filtered and concentrated in vacuo to give the crude material as a yellow solid.
  • the residue was diluted with MeOH/DMSO and purified by preparative HPLC (Gilson: 10- 80% (0.1% TFA in CH3CN) in H 2 O over 15 min).
  • Step 1 l-(2-Methyl-6-(methylthio)pyrimidin-4-yl)hydrazine (2.0 g, 12 mmol), methyl 4-(2- cyanoacetyl)benzoate (2 g, 12 mmol) and acetic acid (0.7 ml, 12 mmol) were dissolved in ethanol (23 mL) in a sealed tube and heated at 100 0 C overnight. After cooling to rt, the desired product crashed out from EtOH.
  • a glass microwave reaction vessel was charged with 6-(2-chloroimidazo[l,2-a]pyridin- 3-yl)-2-methyl-9-(tetrahydro-2H-pyran-2-yl)-9H-purine (0.222 g, 0.602 mmol, from Example 13, Step 2) and 2-butanol (6 mL). To this mixture was added 3-aminopyrazole (250 mg, 3.01 5 mmol) followed by p-toluenesulfonic acid (282 mg, 1.806 mmol). The resulting slurry was stirred and heated with microwave irradiation (Biotage initiator) at 130 0 C for 1O h.
  • microwave irradiation Biotage initiator
  • Step 2 To a 0.5-2 mL Personal Chemistry microwave vial was added was added N-(6- methoxypyridin-3-yl)imidazo[l,2-a]pyridin-2-amine (0.075 g, 0.31 mmol), 4-chloro-N,N- bis(4-methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine (0.18 g, 0.47 mmol), potassium carbonate (0.086 g, 0.62 mmol), triphenylphosphine (0.016 g, 0.062 mmol), palladium (II) acetate (7.0 mg, 0.031 mmol) and 1,4-dioxane (1.0 mL).
  • the vial was sealed and carefully evacuated and backfilled with N 2 . This was repeated twice.
  • the reaction mixture was heated at 140 0 C for 3 h in the microwave.
  • the reaction mixture was poured into aq. NaHCO 3 and extracted with EtOAc.
  • the combined extracts were washed with brine and then dried (Na 2 SO 4 ) and concentrated onto silica.
  • Step l To a 10-20 rnL Personal Chemistry microwave vial was added was added 2- chloroimidazo[l,2-a]pyridine (0.50 g, 3.3 mmol), 4-chloro-N,N-bis(4-methoxybenzyl)-6- methyl-l,3,5-triazin-2-amine (1.89 g, 4.92 mmol), potassium carbonate (0.906 g, 6.55 mmol), triphenylphosphine (0.172 g, 0.655 mmol), palladium (II) acetate (0.074 g, 0.33 mmol) and 1,4-dioxane (10 mL). The vial was sealed and carefully evacuated and backfilled with N 2 . This was repeated twice.
  • Step 3 A glass microwave reaction vessel was charged with 2-chloro-6-fluoroimidazo[ 1 ,2- ajpyridine (0.85 g, 4.99 mmol), triphenylphosphine (0.26 g,, 1.0 mmol), palladium diacetate (112 mg, 0.50 mmol), potassium carbonate (0.14 g, 9.99 mmol) and 4-chloro-N,N-bis(4- methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine (2691 mg, 6.99 mmol) in dioxane (15 mL). The vial was sealed and purged with argon for several minutes.
  • the reaction mixture was stirred and heated in an Emrys Optmizer microwave reactor (Personal Chemistry, Biotage AB, Inc., Upssala, Sweden) at 140 0 C for 3 h.
  • the reaction mixture was diluted with satd. NaHCO 3 (10 mL) and extracted with DCM. The combined organic extracts were dried over Na 2 SO 4 . The solution was filtered and concentrated in vacuum to give the crude material.
  • Step 4 To a 10-mL vial was added 4-(2-chloro-6-fluoroimidazo[l,2-a]pyridin-3-yl)-N,N-bis(4- methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine (139 mg, 0.27 mmol), 6-methoxypyridin-3- amine (40 mg, 0.32 mmol) (Aldrich, St Louis, MO), sodium 2-methylpropan-2-olate (64 mg, 0.67 mmol), chloro[2-(dicyclohexylphosphino)-3,6-dimethoxy-2'-4'-6'-tri-i-propyl-l,r- biphenyl][2-(2-aminoethyl)phenyl]palladium(II) (Strem, Newburyport, MA) (2 mg, 2.7 ⁇ mol), and dioxane (1.5 niL).
  • the vial was sealed and purged with argon for several minutes and stirred at 90 0 C for 6 h.
  • the reaction mixture was diluted with satd NaHCO 3 (5 mL) and extracted with EtOAc. The combined organic extracts were dried over Na 2 SO 4 , and the solution was filtered and concentrated in vacuo to give the crude material.
  • Zinc(II) chloride (4.50 g, 33.0 mmol) was flammed dried under vacuum and then dissolved in THF (30.0 mL) to make a 1 M solution.
  • THF 30.0 mL
  • 2,2,6, 6-tetramethylpiperidine 4.24 g, 30 mmol
  • n-Bbutyllithium (12.00 mL, 30.0 mmol) was added dropwise, and allowed the reaction mixture to warm to -10 0 C over 1 hr.
  • Zinc(II) chloride (4.50 g, 33.0 mmol) solution was added dropwise, and stirring was continued for 30 min at -10 0 C, then 30 min at rt. This solution in THF (0.42 M) was used for coupling reactions and was stored under nitrogen. Dissolved 2-chloro-7-methoxyimidazo[ 1 ,2-a]pyridine (210 mg, 1.150 mmol) in THF
  • reaction mixture was stirred for 16 hrs at rt until reaction was complete by LCMS.
  • the reaction mixture was quenched reaction with NH 4 Cl and aqueous layer was extracted with EtOAc, dried organics over Na 2 SO 4 , and concentrated under rotary evaporation.
  • the crude product was purified via MPLC (ISCO) with a gradient hexanes:EtOAc column 0: 100 to 100:0.
  • Step 5 Charged a flask with Pd 2 dba 3 (132 mg, 0.144 mmol) and CyP-Ft-Bu 2 (208 mg, 0.289 mmol) and degassed, backfilling with nitrogen three times. The solids were dissolved in t- BuOH (2094 ⁇ L) and water (130 ⁇ L, 7.22 mmol) and heated to 100 0 C for 15 min.
  • reaction mixture was cooled to rt and lH-pyrazol-3 -amine (120 mg, 1.445 mmol), 4-(2-chloro- 7-methoxyimidazo[l,2-a]pyridin-3-yl)-6-methyl-l,3,5-triazin-2-amine (210 mg, 0.722 mmol) and cesium carbonate (706 mg, 2.167 mmol) were added.
  • the reaction mixture was degassed again with nitrogen and heated at 100 0 C for 4 hrs until the reaction was complete by LCMS.
  • reaction mixture was redissolved in DMSO and purified via HPLC (Gilson) with a gradient water :acetonitrile 85:15 to 30:70 with 0.1% TFA.
  • N-(l-(lH-benzo- [d]imidazol-2-yl)-l H-pyrazol-3 -yl)-3-(6-amino-2-methylpyrimidin-4-yl)imidazo[ 1,2- b]pyridazin-2-amine 2,2,2-trifluoroacetate was collected directly as the TFA salt as a yellow solid, m/z (ESI, +ve ion) 424.5 (M+H) + .
  • reaction mixture was concentrated, then redissolved mixture in DMSO and purified via HPLC (Gilson) with a gradient water :acetonitrile 85:15 to 10:90 with 0.1% TFA to afford 3-(3-(6-amino-2-methylpyrimidin- 4-yl)imidazo[l,2-b]pyridazin-2-ylamino)-N-phenyl-lH-pyrazole-l-carboxamide (25 mg, 0.046 mmol, 12 % yield) as the TFA salt, m/z (ESI, +ve ion) 427.5 (M+H) + .
  • reaction mixture was concentrated, then redissolved in DMSO and purified via HPLC (Gilson) with a gradient water :acetonitrile 90:10 to 50:50 with 0.1% TFA to afford 3-(3-(6-amino-2-methylpyrimidin-4-yl)imidazo[l,2-b]pyridazin-2- ylamino)-N-(2-methoxyethyl)-lH-pyrazole-l-carboxamide (1.05 mg, 2.010 ⁇ mol, 1 % yield ) as the TFA salt, m/z (ESI, +ve ion) 409.5 (M+H) + .
  • Step l To a solution of 2-chloroacetic acid (1.661 mL, 24.08 mmol) and acetonitrile (4.01 mL) was added triethylamine 99% (5.02 mL, 36.1 mmol), dropwise under N 2 .
  • 2-Amino-5- bromopyridine (5.00 g, 28.9 mmol) was added as a solid in a single portion.
  • the reaction mixture was maintained at rt for 3 min until majority of chloropyridine had dissolved and then heated at 90 0 C for 60 min. The reation appeared to undergo an uncontrolled exotherm after 50 minutes at 90 0 C and turned dark brown.
  • 6-Bromo-2-chloroimidazo[l,2-a]pyridine was prepared according to preparation of 2- chloroimidazo[l,2-b]pyridazine in Example 18, Step 2.
  • LCMS trifluoroacetic acid modifier, ESI) m/z: 231.0 (M+H) + Step 3
  • 1,4-Dioxane (2.58 mL) was added to 4-(6-bromo-2-chloroimidazo[l,2-a]pyridin-3-yl)- 6-methyl-l,3,5-triazin-2-amine (0.175 g, 0.515 mmol), [l,l-bis(diphenylphosphino)- ferrocene]palladium(II) dichloride dichloromethane adduct (0.042 g, 0.052 mmol), and 2- fluorobenzeneboronic acid (0.079 g, 0.567 mmol) under N 2 .
  • 6-(2-Chloroimidazo[l ,2-a]pyridin-3-yl)-2-methylpyrimidin-4-amine was prepared according to the preparation of 6-(6-bromo-2-chloroimidazo[l,2-a]pyridin-3-yl)-2- methylpyrimidin-4-amine in Example A, step 4.
  • LCMS trifluoroacetic acid modifier, ESI
  • a microwave vial was charged with 2-chloro-6-(pyrimidin-5-yl)imidazo[l,2- b]pyridazine (0.366 g, 1.580 mmol), 4,6-dichloro-2-methylpyrimidine (0.567 g, 3.48 mmol, Aldrich), cesium carbonate (1.030 g, 3.16 mmol, Strem), triphenylphosphine (0.083 g, 0.316 mmol, Fluka) and palladium(ii) acetate (0.035 g, 0.158 mmol, Strem).
  • the vial was sealed with septa cap, flushed with N 2 , and dioxane was added via syringe (6.32 mL).
  • reaction mixture was sparged with N 2 for 10 minutes, then irradiated in the microwave (Biotage Initiator) at 130 0 C for 2 h.
  • microwave Biotage Initiator
  • the crude reaction mixture was absorbed directly onto a 25 g pre- packed silica loading cartridge for purification by MPLC (Teledine Isco combiFlash
  • a sealable microwave vial was charged with 2-chloro-3-(6-chloro-2-methylpyrimidin- 4-yl)-6-(pyrimidin-5-yl)imidazo[l,2-b]pyridazine (0.380 g, 1.061 mmol) and the vial was sealed using a septa cap.
  • IO N ammonia in MeOH (17.10 mL, 790 mmol) was added via syringe to generate a tan slurry. Gaseous ammonia was bubbled through the slurry for 5 min, then the mixture was irradiated in the microwave (Biotage Initiator) at 100 0 C for 2 h. Upon cooling to rt, a tan solid precipitated.
  • a sealable vial was charged with tris(dibenzylideneacetone)dipalladium(0) (32.4 mg, 0.035 mmol, Strem) and (r)-l-[(s)-2-(dicyclohexylphosphino)ferrocenyl]ethyl-di-tert- butylphosphine (39.3 mg, 0.071 mmol, Strem).
  • the vial was sealed with a septa cap and flushed with N 2 .
  • tert-Butanol (1 mL) was added, followed by water (63.8 ⁇ L, 3.54 mmol) and the resulting mixture was sparged with N 2 for 5 min. The solution was heated at 120 0 C for 10 min, and then cooled to rt.
  • a sealable vial was charged with N-(6-(2-( 1 -acetyl- lH-pyrazol-3 -ylamino)-6- (pyrimidin-5-yl)imidazo[ 1 ,2-a]pyridin-3-yl)-2-methylpyrimidin-4-yl)acetamide (54.0 mg, 0.115 mmol) followed by CH 2 Cl 2 (5 mL) and MeOH (0.5 mL) and MP-Carbonate (159 mg, 3.07 mmol/gram loading, Biotage). The reaction mixture was heated at 60 0 C for 1 hour. The solution was filtered to remove the MP-Carbonate and the filtrate was concentrated for purification by MPLC (Teledine Isco combiFlash Companion).
  • the reaction mixture was diluted with DMF and purified by Prep- HPLC (Phenomenex Gemini 5 micron, C 18, 100 x 30 mm, 5 to 55% CH 3 CN(0.1% TFA)/H 2 O(0.1% TFA) over 20 min then 100% CH 3 CN(0.1% TFA) for 3 minutes at 20 ml/min) with the fractions containing suspected product concentrated to afford the title compound as a tan solid (0.011 g, 10 % yield).
  • a glass reaction vessel was charged with 3-(6-(bis(4-methoxybenzyl)amino)-2- methylpyrimidin-4-yl)-N-(l H-pyrazol-3 -yl)imidazo[l,2-b]pyridazin-2-amine (0.200 g, 0.365 mmol) and DMF (3 mL).
  • the reaction vial was purged with nitrogen and sodium hydride (0.015 g, 0.621 mmol) was added.
  • the resulting slurry was stirred at room temperature for 5 minutes, until gas evolution ceased.
  • 3-(Bromomethyl)pyridine (0.063 g, 0.365 mmol) was added and the reaction was heated to 50 0 C for 15 h.
  • a glass vial was charged with benzene- 1,4-diamine (1.659 g, 15.34 mmol), cesium carbonate (5.00 g, 15.34 mmol), Pd 2 (dba) 3 (0.281 g, 0.307 mmol), JosiPhos (0.340 g, 0.614 mmol) and 6-(2-chloroimidazo[l,2-b]pyridazin-3-yl)-2-methylpyrimidin-4-amine (0.8 g, 3.07 mmol).
  • the vial was sealed, evacuated and backfilled with nitrogen; this was repeated twice.
  • t-BuOH (8.60 mL) and water (0.553 mL, 30.7 mmol) were added via syringe.
  • Nl-(3-(6-amino-2-methylpyrimidin-4- yl)imidazo[l,2-b]pyridazin-2-yl)benzene-l,4-diamine 0.385 g, 1.158 mmol
  • 4-nitrophenyl carbonochloridate 0.233 g, 1.158 mmol
  • N-ethyl-N-isopropylpropan-2-amine 0.202 mL, 1.158 mmol
  • Step 1 In a 10-mL sealed tube under N 2 were dissolved 3,5-dichloropyridazine (1.00 g, 6.71 mmol) and morpholine (1.754 mL, 20.14 mmol) portion wise in 2 mL of MeCN at 0 0 C. After 1 h, the reaction mixture was concentrated under reduced pressure with silica and purified by MPLC (ISCO) with DCM:MeOH+NH 4 OH 100:0 to 90:10 to afford 4-(6-chloropyridazin-4- yl)morpholine (1.100 g, 5.51 mmol, 82 % yield), m/z (ESI, +ve ion) 200.0 (M+H) + Step 2
  • Example 1 In vitro assays The following assays can be used to determine the degree of activity of individual compounds as PI3 kinase and/or mTOR inhibitors. Recombinant expression of PI3K enzymes
  • Full length pi 10 subunits of PI3K ⁇ , ⁇ and ⁇ , N-terminally labeled with polyHis tag, can be co-expressed with p85 with Baculo virus expression vectors in sf9 insect cells.
  • Pl 10/p85 heterodimers can be purified by sequential Ni-NTA, Q-HP, Superdex-100 chromatography.
  • Purified ⁇ , ⁇ and ⁇ isozymes can be stored at -20 0 C in 2OmM Tris, pH 8, 0.2M NaCl, 50% glycerol, 5mM DTT, 2mM Na cholate.
  • Truncated PI3K ⁇ , residues 114-1102, N-terminally labeled with polyHis tag, can be expressed with Baculo virus in Hi5 insect cells.
  • the ⁇ isozyme can be purified by sequential Ni-NTA, Superdex-200, Q-HP chromatography.
  • the ⁇ isozyme can be stored frozen at -80 0 C in NaH 2 PO 4 , pH 8, 0.2M NaCl, 1% ethylene glycol, 2mM ⁇ -mercaptoethanol.
  • PBK ATPLoss In vitro PB kinase enzyme assays
  • PBK enzyme assays (alpha, beta, delta and gamma) can be performed in 25 ⁇ L with the above final concentrations of components in white polyproplyene plates.
  • Phosphatidyl inositol phosphoacceptor, PtdIns(4,5)P2 (e.g,. P4508) can be obtained from Echelon Biosciences, Salt Lake City, UT.
  • the ATPase activity of the alpha and gamma isozymes may not be greatly stimulated by PtdIns(4,5)P2 under these conditions, it can be omitted from the assay of these isozymes.
  • Test compounds can be dissolved in DMSO and diluted with threefold serial dilutions.
  • the compound in DMSO (1 ⁇ L) may be added per test well, and the inhibition relative to reactions containing no compound, with and without enzyme can be determined. After assay incubation at RT, the reaction can be stopped and residual ATP can be determined by addition of an equal volume of a commercial ATP bio luminescence kit (Perkin Elmer EasyLite, Perkin Elmer, Waltham, MA) according to the manufacturer's instructions, and detected using an Analyst GT luminometer.
  • a commercial ATP bio luminescence kit Perkin Elmer EasyLite, Perkin Elmer, Waltham, MA
  • HCT 116 human colon carcinoma cell lines can be grown in McCoy's 5 A growth medium (GIBCO, Carlsbad, CA) containing 10% FBS (GIBCO, Carlsbad, CA) and Xl Penicillin-streptomycin-glutamine (GIBCO, Carlsbad, CA). Prior to the assay, cells can be detached from the culture flask with trypsin, and re-suspended in complete media to give a final concentration of 1.6 x 10 5 cells per mL.
  • HCTl 16 cell suspension Aliquots (100 ⁇ l) of the HCTl 16 cell suspension can be seeded into each well of a 96 well tissue culture plate to give a final density of 16,000 cells per well. Cells can then be incubated overnight at 37 0 C. The following day the cells can be treated with serially diluted test compounds and incubated for 2 hours at 37 0 C.
  • the culture media on the HCT 116 cells can be replaced with 189 ⁇ L McCoys media, supplemented with 0.1% BSA (ICN Biomedicals, Inc., Costa Mesa, CA).
  • Compounds can be prepared as either 10 mM or 0.5 mM stock solutions in DMSO, and serially diluted 3 fold in a 10-point dose-response curve to give final concentrations that are 200-fold greater than the desired final test concentration. Aliquots (l ⁇ L) of serially-diluted compounds can be transferred to 96 well tissue culture plates containing the HCT 116 cells. As a minimum response control, each plate can contain wells having a final concentration of 2.5 ⁇ M of a potent PI3K inhibitor which had previously been shown to completely inhibit Akt phosphorylation at this test concentration. As a maximum response control, wells can contain 0.5% DMSO in place of compound.
  • the plates can be mixed at 700 rpm for 2 min to ensure even distribution of the test compound and incubated for 2 hours at 37 0 C.
  • Cells can then be stimulated with insulin-like growth factor 1 (Sigma, St Louis, MO) at final concentration of 100ng/ml for 15 minutes at 37 0 C.
  • the media can then be removed and the cells treated with 80 ⁇ L cell-lysis buffer (MSD) containing a cocktail of protease and phosphatase inhibitors for one hour at 4 0 C. 25 ⁇ L Cell lysate can then be transferred to pre-blocked MSD assay plates pre-coated with a capture antibody specific for Akt, and the plates can be incubated for 2 hours at room temperature.
  • MSD cell-lysis buffer
  • the cell lysates can then be removed and plates can then be washed four times with 200 ⁇ l per well of Tris wash buffer (500 mM Tris, PH 7.5, 1.5 M NaCl, 0.2% Tween-20). Subsequently cells can be incubated for 1 hour at room temperature with a 25 ⁇ L solution containing the detection antibody, anti-phospho Akt (Ser 473) labeled with an electrochemiluminescent compound (Meso Scale Discovery SULPHO-TAGTM label, MSD, Gaithersburg, MD). The detection antibody can be removed and plates can then be washed four times with 200 ⁇ L per well of Tris wash buffer.
  • Tris wash buffer 500 mM Tris, PH 7.5, 1.5 M NaCl, 0.2% Tween-20.
  • cells can be incubated for 1 hour at room temperature with a 25 ⁇ L solution containing the detection antibody, anti-phospho Akt (Ser 473) labeled with an electrochemiluminescent compound (Meso Scal
  • the pAkt AlphaScreen® assay determines whether there is phosphorylation of Akt at Serine 473 by recruitment of a phosphospecific antibody. This assay was performed using U87 MG cells.
  • the U87 growth media consists of MEM (Gibco, Carlsbad, CA) supplemented with 10% FBS (Gibco,), Ix Non-Essential Amino Acids (Gibco,) and Ix Penicillin/Streptomycin/Glutamine (Gibco).
  • the cells were maintained weekly using 0.05% Trypsin (Gibco) and replated in 150 mm TC- Treated Culture Dishes (Corning, Corning, NY).
  • the first day of the assay the adherent cells were trypsinized, media was added to the loose cells and cells were mixed to a homogenous mixture. 0.5 ml of the homogenous mixture was counted on the Beckman Coulter ® Vi-CELL TM XR (Fullerton, CA). 50 frames of cells were counted and the number of viable cells was determined. The cells were then diluted to 0.25 million cells per ml, and centrifuged at 200 rcf for 5 minutes. The media was removed and the cells were reconstituted in fresh media for plating. The cells were plated at 20 ⁇ l per well on the PerkinElmer ® FlexDrop PLUS in Low Volume 384 Well White Tissue Culture Plates
  • the compound plates were prepared, the cells were treated with compound and the pAkt reaction mix was added to the cell lysate.
  • 384 well compound plates were prepared containing 1 ⁇ l of compound per well starting at 5 mM and diluted 1 :2 across the row, resulting in a 22 well serial dilution. 39 ⁇ l of growth media was added to the compound plate in rows 1-22 using the PerkinElmer ® FlexDrop PLUS resulting in a DMSO concentration of 2.5%.
  • the cell plates and diluted compound plates were put onto the VELOCITY 11 VPREP 384 ST where the compound plate was mixed and 5 ⁇ l of serially diluted compound or controls was added to the cell plate. The final concentration of the compounds was 25 ⁇ M serially diluted to 11.9 pM in 0.5% DMSO.
  • the cell plates were then incubated with compound for two hours at 37 0 C, 5% CO 2 . After two hours, the media in the cell plates was aspirated using the BioTek ® ELx405HT plate washer (Winooski, VT) removing the majority of media and compound without disturbing the adherent U87 cells.
  • the following assay reagents are components of the SureFire ® Akt (Ser 473) Phosphorylation 5OK Point Kit (TGR BioSciences, Sydney, Austalia) and an IgG Detection Kit (PerkinElmer, Waltham, MA). 5 ⁇ l of Ix Lysis Buffer was added to each well using the PerkinElmer ® FlexDrop PLUS.
  • the plates were then incubated at room temperature on a shaker for ten minutes.
  • the AlphaScreen ® reaction was prepared under low light conditions (subdued or green light) including p-Akt (Ser 473) Reaction Buffer, Dilution Buffer, Activation Buffer, Acceptor Beads and Donor Beads at a ratio of 40:20: 10: 1 : 1 respectively.
  • the AlphaScreen ® reaction was added to the cell lysate at 6 ⁇ l per well using the PerkinElmer ® FlexDrop PLUS. The plates were placed in a humid environment to reduce edge effects and incubated overnight at room temperature with restricted air flow in the dark.
  • the PI3K AlphaScreen ® assay measures the activity of a panel of four phosphoinositide 3-kinases: PI3K ⁇ , PI3K ⁇ , PI3K ⁇ , and PI3K ⁇ . Each of these enzymes phosphorylates the 3'-hydroxyl group on phosphatidylinositiol (4,5)-bisphosphate (PIP 2 ) to produce phosphatidylinositol (3,4,5)-trisphosphate (PIP 3 ).
  • This phosphorylation activity is measured using a GST-tagged PIP3 binding protein (Echelon Biosciences, Salt Lake City, UT), an anti-GST-tagged Acceptor bead, and streptavidin-Donor bead.
  • the interaction of biotinylated-PIP3 analog (IP 4 ) and the PIP3 binding protein brings both Acceptor and Donor beads together producing, upon excitation of the Donor beads at 680 nm, a singlet oxygen species leading to the luminescent AlphaScreen ® signal.
  • PIP 3 is produced via phophorylation of PIP2 by a PI3K
  • PIP3 competes with biotinylated-PIP 3 analog (IP 4 ) for binding to the PIP 3 binding protein.
  • the enzyme reaction buffer is made using sterile water (Baxter, Deerfield, IL) and 5OmM Tris HCl pH 7, 14mM MgCl 2 , 2mM sodium cholate, and 10OmM NaCl. 2mM DTT is added fresh the day of the experiment.
  • the AlphaScreen ® reaction buffer is made using sterile water and 1OmM Tris HCl pH 7.5, 15OmM NaCl, 0.10% Tween 20, and 3OmM EDTA. ImM DTT is added fresh the day of the experiment.
  • the source plates for this assay are 384-well Greiner clear polypropylene plates containing test compounds at 5mM and diluted 1 :2 over 22 points. Columns 23 and 24 contain only DMSO as these are designated for positive and negative controls. Source plates are replicated into 384-well Optiplates (PerkinElmer, Waltham, MA), 0.5 ⁇ L/well, to make assay- ready plates.
  • PBK ⁇ is diluted to 1.6nM
  • PBK ⁇ is diluted to 0.8nM
  • PBK ⁇ is diluted to 15nM
  • PBK ⁇ is diluted to 1.6nM.
  • Two different 2X substrate solutions are made in enzyme reaction buffer.
  • PI(4,5)P2 (Echelon Biosciences, Salt Lake City, UT) is diluted to lO ⁇ M and ATP is diluted to 20 ⁇ M. This solution is used in the assays testing PBK ⁇ and PBK ⁇ .
  • PI(4,5)P2 is diluted to lO ⁇ M and ATP is diluted to 8 ⁇ M. This solution is used in the assays testing PBK ⁇ and PBK ⁇ .
  • the AlphaScreen ® reaction solutions are made using beads from the anti-GST
  • AlphaScreen ® kit (PerkinElmer, Waltham, MA). Two solutions are made in Alphascreen reaction buffer to 4X working concentrations. In one solution, biotinylated-IP 4 (Echelon Biosciences, Salt Lake City, UT) is diluted to 4OnM and streptavadin-Donor Beads are diluted to 80 ⁇ g/mL. In the second solution, PIP 3 -binding protein (Echelon Biosciences, Salt Lake City, UT) is diluted to 4OnM and anti-GST- Acceptor Beads are diluted to 80 ⁇ g/mL. lO ⁇ L/well of enzyme reaction buffer is added to Column 24 of the assay ready plates in place of enzyme. This is done for plates in the PBK ⁇ , ⁇ , and ⁇ assays.
  • biotinylated-IP 4 (Echelon Biosciences, Salt Lake City, UT) is diluted to 4OnM and streptavadin-Donor Beads are diluted to 80 ⁇ g/mL
  • lO ⁇ L/well of 2X enzyme (PBK ⁇ , ⁇ , ⁇ ) is added to Columns 1-23 of the appropriate assay ready plates (for PBK ⁇ lO ⁇ L is added to Columns 1-24).
  • lO ⁇ L/well of the appropriate substrate solution (the solution with 20 ⁇ M ATP for PBK ⁇ and ⁇ assays, and the solution with 8 ⁇ M ATP for PBK ⁇ and ⁇ assays) is then added to Columns 1-24 of the plates. Plates are then incubated at room temperature for 20 minutes.
  • the mTOR LanthaScreen is a TR-FRET assay measuring the phosphorylation of mTOR's substrate 4EBP 1.
  • 384 well compound plates were prepared containing 1 ⁇ l of compound per well starting at 5 mM and diluted 1 :2 across the row, resulting in a 22 well serial dilution.
  • 24 ⁇ l of assay buffer (Invitrogen, PV4794) with 2 mM DTT was added to the compound plate in rows 1-24 using the VELOCITYI 1 TM VPREP TM 384 ST resulting in a DMSO concentration of 4%.
  • the compound plate was mixed and 2.5 ⁇ l of serially diluted compound or controls was added to the assay plate (Costar, 3658).
  • the assay was conducted on the PerkinElmer ® FlexDrop PLUS. A 5 ⁇ l mix of 800 nM GFP-4E-BP1 (Invitrogen, PV4759) and 20 ⁇ M ATP (Amgen) was added to rows 1-24. 2.5 ⁇ l of 0.6 ⁇ g/ml of mTOR Enzyme (Amgen) was added to rows 1-23. 2.5 ⁇ l of assay buffer was added to row 24 for the low control. The final concentration of the compounds was 50 ⁇ M serially diluted to 23.84 pM in 1% DMSO. The final high control had 1% DMSO and the low control was a no enzyme control with a concentration of 1% DMSO.
  • the final concentrations in the assay reagents were 400 nM GFP-4E-BP1, 10 ⁇ M of ATP and 0.15 ⁇ g/ml of mTOR enzyme.
  • the compound, enzyme, and substrate were incubated for 90 minutes.
  • 10 ⁇ l of stop solution was added (20 mM Tris, pH 7.5 (Invitrogen, 15567-027), 0.02 % Sodium Azide (Teknova, S0208), 0.01 % NP-40 (Roche, 11754599001), 20 mM EDTA (Invitrogen, 15575-038) and 4 nM of Tb-anti-p4E-BPl (Invitrogen, PV4758)) for a final concentration of 2 nM of Tb-anti-p4E-BP 1.
  • the p4EBP 1 AlphaScreen assay determines whether there is phosphorylation of 4EBP 1 at Thr37/Thr46 by recruitment of a phosphospecific antibody. This assay was performed using U87 MG cells.
  • the U87 growth media consists of MEM (Gibco, 51200-038) supplemented with 10% FBS (Gibco, 16140-071), Ix Non-Essential Amino Acids (Gibco, 11140-050) and Ix Penicillin/Streptomycin/Glutamine (Gibco, 10378-016).
  • the cells were maintained weekly using 0.05% Trypsin (Gibco, 25300-054) and replated in 150 mm TC- Treated Culture Dishes (Corning, 430599).
  • the first day of the assay the adherent cells were trypsinized, media was added to the loose cells and cells were mixed to a homogenous mixture. 0.5 ml of the homogenous mixture was counted on the Beckman Coulter ® Vi-CELL TM XR. 50 frames of cells were counted and the number of viable cells was determined. The cells were then diluted to 0.25 million cells per ml, and centrifuged at 200 rcf for 5 minutes. The media was removed and the cells were reconstituted in fresh media for plating.
  • the cells were plated at 20 ⁇ l per well on the PerkinElmer ® FlexDrop PLUS in Low Volume 384 Well White Tissue Culture Plates (Corning, 3826) with a final cell density of 5K cells per well. The plates were incubated overnight at 37° Celsius, 5% CO 2 .
  • the compound plates were prepared, the cells were treated with compound and the p4EBP 1 reaction mix was added to the cell lysate.
  • 384 well compound plates were prepared by Amgen's Sample Bank containing 1 ⁇ l of compound per well starting at 5 mM and diluted 1 :2 across the row, resulting in a 22 well serial dilution. 39 ⁇ l of growth media was added to the compound plate in rows 1-22 using the PerkinElmer ® FlexDrop PLUS resulting in a DMSO concentration of 2.5%.
  • control columns were added manually; 40 ⁇ l of 2.5% DMSO (Sigma, D4540- 100ml) in growth media was added to the plate for the high control and 40 ⁇ l of 50 ⁇ M of AMG2203766 with 2.5% DMSO was added to the plate as the low control.
  • the cell plates and diluted compound plates were put onto the VELOCITYI 1 TM
  • VPREP TM 384 ST where the compound plate was mixed and 5 ⁇ l of serially diluted compound or controls was added to the cell plate.
  • the final concentration of the compounds was 25 ⁇ M serially diluted to 11.9 pM in 0.5% DMSO.
  • the final high control had 0.5% DMSO and the low control concentration was 10 ⁇ M AMG2203766 in 0.5% DMSO.
  • the cell plates were then incubated with compound for two hours at 37° Celsius, 5% CO 2 . After two hours, the media in the cell plates was aspirated using the BioTek ® ELx405HT plate washer removing the majority of media and compound without disturbing the adherent U87 cells.
  • the following assay reagents are components of the SureFire Phospho-4EBP 1 (Thr37/Thr46) 5OK Point Kit (TGR BioSciences, TGR4ES50K) and an IgG Detection Kit (PerkinElmer, 6760617R). 5 ⁇ l of Ix Lysis Buffer was added to each well using the PerkinElmer ® FlexDrop PLUS . The plates were then incubated at room temperature on a shaker for ten minutes.
  • the AlphaScreen reaction was prepared under low light conditions (subdued or green light) including p-4E-BPl (Thr37/46) Reaction Buffer, Activation Buffer, Acceptor Beads and Donor Beads at a ratio of 60: 10: 1 : 1 respectively.
  • the AlphaScreen reaction was added to the cell lysate at 6 ⁇ l per well using the PerkinElmer R FlexDrop PLUS. The plates were placed in a humid environment to reduce edge effects and incubated overnight at room temperature with restricted air flow in the dark.
  • Tablet Formulation The following ingredients are mixed intimately and pressed into single scored tablets.
  • Ingredient Quantity per tablet Mg compound of this invention 400 cornstarch 50 croscarmellose sodium 25 lactose 120 magnesium stearate 5
  • Capsule Formulation The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule.

Abstract

The present invention provides compounds that are PIKK inhibitors, more specifically, mTOR and/or PI3Kα kinase inhibitors and are therefore useful for the treatment of diseases treatable by inhibition of kinases, specifically PI3 kinases, more specifically, mTOR and/or PI3Kα, such as cancer. Also provided are pharmaceutical compositions containing such compounds and processes for preparing such compounds.

Description

HETEROARYL COMPOUNDS AS PIKK INHIBITORS
Field of the Invention
The present invention provides compounds that are PIKK (Phosphoinositide-3 -kinase related kinase) inhibitors, more specifically, mTOR and/or PBKα kinase inhibitors and are therefore useful for the treatment of diseases treatable by inhibition of kinases, specifically PB kinases, more specifically, mTOR and/or PBKα, such as cancer. Also provided are pharmaceutical compositions containing such compounds and processes for preparing such compounds.
Background
PB kinases are a family of lipid kinases that have been found to play a key role in the regulation of many cellular processes including proliferation, survival, carbohydrate metabolism, and motility. PBKs are considered to have an important role in intracellular signal transduction. In particular, the PBKs generate and convey signals that have important roles in cancer. PBKs are ubiquitously expressed, are activated by a high proportion of cell surface receptors, especially those linked to tyrosine kinases, and influence a variety of cellular functions and events. Although some PBK activity is likely to be essential for cellular health, PBKs are a diverse group of enzymes for which there is increasing evidence of functional specialization. This opens up the possibility of developing iso form-selective inhibitors that can be used to treat cancer.
The primary enzymatic activity of PBK is the phosphorylation of inositol lipids (phosphoinositides) on the 3-position of the inositol headgroup. PB kinases catalyze the addition of phosphate to the 3'-OH position of the inositol ring of inositol lipids generating phosphatidyl inositol monophosphate, phosphatidyl inositol diphosphate and phosphatidyl inositol triphosphate.
There are a total of eight mammalian PBKs, which have been divided into three main classes on the basis of sequence homology, in vitro substrate preference, and method of activation and regulation. Enzymes of a first class (Class I) have a broad substrate specificity and phosphorylate phosphatidylinositiol (Ptdlns), PtdIns(4)P and PtdIns(4,5)P2. Class I PB kinases include mammalian pi 10a, pi lOβ, pi lOδ and pi lOγ. Different members of the PB- kinase family generate different lipid products. To date, four 3-phosphorylated inositol lipids have been identified in vivo. These lipids are bound by proteins that contain the appropriate lipid recognition module and which either act as effectors or transmit the PBK signal onwards. The most familiar form of PBK is a heterodimeric complex, consisting of a 110 kDa catalytic subunit now known as pi 10a and an 85 kDa regulatory/adapter subunit, p85α.
Phosphatidylinositol 3-kinase-alpha (PBKα), a dual specificity lipid and protein kinase, is composed of an 85 kDa regulatory subunit and a 110 kDa catalytic subunit. The protein includes a catalytic subunit, which uses ATP to phosphorylate Ptdlns, PtdIns(4)P and
PtdIns(4,5)P2. PTEN, a tumor suppressor, can dephosphorylate phosphatidylinositol (3,4,5)- trisphosphate (PIP3), the major product of PB kinase Class I. PIP3, in turn, is required for translocation of protein kinase B (AKTl, PKB) to the cell membrane, where it is phosphorylated and activated by upstream kinases. The effect of PTEN on cell death is mediated through the PBKα/AKT 1 pathway.
PBKα has been implicated in the control of cytoskeletal reorganization, apoptosis, vesicular trafficking and proliferation and differentiation processes. Increased copy number and expression of the pi 10a gene (PIK3CA) is associated with a number of cancers such as ovarian cancer, cervical cancer, breast cancer, colon cancer, rectal cancer, endometrial cancer, stomach cancer, liver cancer, lung cancer, thyroid cancer, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), and glioblastomas.
Mammalian target of rapamycin (mTOR) is a serine/threonine kinase of approximately 289 kDa in size and a member of the evolutionary conserved eukaryotic TOR kinases. The mTOR protein is a member of the PIKK family of proteins due to its C-terminal homology (catalytic domain) with PB-kinase and the other family members, e.g. DNA dependent protein kinase (DNA-PKcs), Ataxia-telangiectasia mutated (ATM).
It has been demonstrated that mTOR kinase is a central regulator of cell growth and survival by mediating multiple important cellular functions including translation, cell cycle regulation, cytoskeleton reorganization, apoptosis and autophagy. mTOR resides in two biochemically and functionally distinct complexes that are conserved from yeast to human.
The rapamycin sensitive mTOR-Raptor complex (mTORCl) regulates translation by activation of p70S6 kinase and inhibition of eIF4E binding protein 4EBP 1 through phosphorylation, which is the best-described physiological function of mTOR signaling. mTORCl activity is regulated by extracellular signals (growth factors and hormones) through the PBK/ AKT pathway, and by nutrient availability, intracellular energy status and oxygen through the regulators like LKBl and AMPK. Rapamycin and its analogues inhibit mTORCl activity by disrupting the interaction between mTOR and raptor. The rapamycin-insensitive complex, mT0RC2, was discovered only recently. Unlike mTORCl which contains raptor, the mT0RC2 complex contains other proteins including Rictor and mSinl. mT0RC2 phosphorylates AKT at the hydrophobic Ser473 site, and appears to be essential for AKT activity. Other substrates of mT0RC2 include PKCA: and SGKl. How mT0RC2 activity is regulated is not well understood.
The mTORCl pathway can be activated by elevated PBK/ AKT signaling or mutations in the tumor suppressor genes PTEN or TSC2, providing cells with a growth advantage by promoting protein synthesis. Cancer cells treated with the mTORCl inhibitor rapamycin show growth inhibition and, in some cases, apoptosis. Three rapamycin analogues, CCI-779 (Wyeth), RADOOl (Novartis) and AP23573 (Ariad) are in clinical trials for the treatment of cancer. However response rates vary among cancer types from a low of less than 10% in patients with glioblastoma and breast cancer to a high of around 40% in patients with mantle cell lymphoma.
Recent studies demonstrated that rapamycin can actually induce a strong AKT phosphorylation in tumors by attenuating the feedback inhibition on receptor tyrosine kinases mediated by p70S6K, one of the downstream effectors of mTORCl. For example, in Phase I clinical trials of RADOOl, an increase in pAKT (+22.2 to 63.1% of initial values) was observed after dosing. If mTORCl inhibition-induced phospho-AKT leads to increased cancer cell survival and acquisition of additional lesions, this could counteract the effects of growth inhibition by rapamycin analogues and explain the variable response rate. Therefore, identifying and developing small molecules that target the catalytic activity of mTOR (inhibiting both mTORCl and mT0RC2) will lead to more effective therapeutics to treat cancer patients by preventing the activation of AKT that is caused by mTORCl specific inhibitors like rapamycin and its analogues. Dysregulated mTOR activity has been shown to associate with variety of human cancers such as breast, lung, kidney, brain, ovarian, colon, cervical, endometrial, prostate, liver, thyroid, GI tract, blood and lymphoma and other diseases such as hamartoma syndromes, rheumatoid arthritis, multiple sclerosis. In view of the important role of mTOR in biological processes and disease states, catalytic inhibitors of this protein kinase are desirable.
In view of the important role of PI3Kα and mTOR in biological processes and disease states, inhibitors of these protein kinases are desirable. The present invention provides PIKK inhibitors, particularly PI3Kα and mTOR inhibitors, which are useful for treating PI3Kα and mTOR mediated diseases and conditions. SUMMARY
In one aspect, provided is a compound of Formula (I):
Figure imgf000005_0001
(I) wherein:
Ar1 is an aryl, heteroaryl, cycloalkyl or heterocyclyl ring, wherein each ring is substituted with Rd, Re, or Rf where Rd, Re, or Rf are independently selected from hydrogen, halo, haloalkyl, haloalkoxy, cyano, nitro, alkyl, alkenyl, alkynyl, substituted alkyl, aryl, heteroaryl, heterocycloalkyl, -C(=O)NRaRa, -C(=O)Rb, -C(=O)ORb, -C(=NRa)NRaRa, -ORa, -OC(=O)Rb, -OC(=O)NRaRa, -O-alkylN(Ra)C(=O)ORb, -OC(=O)N(Ra)S(=O)2Rb, -O- alkylNRaRa, -O-alkylORa, -SRa, -S(=O)Rb, -S(=O)2Rb, -S(=O)2NRaRa, -S(=O)2N(Ra)C(=O)Rb, -S(=O)2N(Ra)C(=O)ORb, -S(=O)2N(Ra)C(=O)NRaRa, -NRaRa, -N(Ra)C(=O)Rb, -N(Ra)C(=O)ORb, -N(Ra)C(=O)NRaRa, -N(Ra)C(=S)NRaRa, -N(Ra)C(=NRa)NRaRa, -N(Ra)S(=O)2Rb, -N(Ra)S(=O)2NRaRa, -NRa-alkylene-NRaRa, or -NRa-alkylene-ORa ;
R1 is hydrogen or alkyl;
R2 is methyl or ethyl;
Z1 -N- or -CR3- where R3 is H or alkyl; Z2 is -N- or -CR4- where R4 is Rd, -(alkylene)heterocycloalkyl,
-(alkylene)nO(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -( alkylene)n-N(Ra)- (alkylene)nheteroaryl, or -( alkylene)nO(alkylene)nheteroaryl;
Z3 is -N- or -CR5- where R5 is Rd, -(alkylene)naryl, -(alkylene)-heteroaryl, -(alkylene)heterocycloalkyl, -(alkylene)nO-(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)nheteroaryl, or -(alkylene)nO(alkylene)nheteroaryl provided that only two of Z1, Z2 and Z3 can simultaneously be -N-; or when Z2 is -CR4- and Z3 is -CR5- then R4 and R5 together with the atoms to which they are attached can form ring A which is phenyl or a 5 or 6 membered heteroaryl ring and ring A is substituted with Rg or Rh where Rg or Rh are independently Rd, -(alkylene)n_ heterocycloalkyl, -(alkylene)nO(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -( alkylene)nN(Ra)(alkylene)nheteroaryl, or -(alkylene)nO(alkylene)nheteroaryl;
Z4 is -N- or -C-; provided that when Z5 is -CR6- where R6 together with Z6 forms phenyl, and Z7 is -N-, then Z4 is -C-;
Z5, Z6 or Z7 are each independently selected from -N- or -CR6- provided at least one of Z4, Z5, Z6 and Z7 is -N- where R6 is Rd ; or R6 together with the adjacent ring atom can form phenyl or 5 or six membered heteroaryl ring wherein the phenyl or heteroaryl ring is substituted with R1, RJ, or Rk where R1, RJ, or Rk are independently Rd, -(alkylene)naryl, - (alkylene)heteroaryl, -( alkylene)heterocycloalkyl, -(alkylene)nO(alkylene)naryl, - (alkylene)nN(Ra)(alkylene)naryl, -(alkylene 2)nN(Ra)( alkylene)nheteroaryl, or -( alkylene)nO(alkylene)nheteroaryl; each Ra is independently hydrogen or Rb; or when two Ra are attached to a nitrogen atom, either alone or part of another group, the two Ras together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring with is optionally substituted with one, two or three substitutents independently selected from oxo, halo, alkyl, alkenyl, alkynyl, cyano, nitro, alkylcarbonyl, carboxy, alkoxycarbonyl, hydroxyl, alkoxy, alkonyloxy, alkylthio, alkylsulfonyl, -alkyl-OH, aminosulfonyl, sulfonylamino, amino, alkylamino, or dialkylamino; each Rb is independently alkyl, cycloalkyl, phenyl, heteroaryl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl is substituted with 0, 1, 2 or 3 substituents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, hydroxyl, unsubstituted heterocycloalkyl, phenyl, alkylcarbonylamino, alkylamino or dialkylamino; and each n is independently 0 or 1 ; or a pharmaceutically acceptable salt thereof; provided the compound of Formula (I) is not:
Figure imgf000006_0001
or a salt thereof.
With compound of Formula (I), in one embodiment the invention is directed to compounds of Formula (Ia) where: each Ra is independently hydrogen or Rb; or when two Ra are attached to a nitrogen atom, either alone or part of another group, the two Ras together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring with is optionally substituted with one, two or three substitutents independently selected from oxo, halo, alkyl, alkenyl, alkynyl, cyano, nitro, alkylcarbonyl, carboxy, alkoxycarbonyl, hydroxyl, alkoxy, alkonyloxy, alkylthio, alkylsulfonyl, aminosulfonyl, sulfonylamino, amino, alkylamino, or dialkylamino; each Rb is independently alkyl, cycloalkyl, phenyl, heteroaryl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl is substituted with 0, 1, 2 or 3 substituents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino or dialkylamino; and other groups are as defined in Formula (I) above.
Compounds of Formula (Ia) are a subset of compounds of Formula (I).
In a second aspect, this invention is directed to a method of treating PBK mediated diseases and disorders in a patient in need thereof which method comprises administering to the patient a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof. In one embodiment, the PBKalpha and/or mTOR mediated disease such as cancer.
In a third aspect, this invention is directed to a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient. In a fourth aspect, the present invention is directed to the use of a compound of
Formula I or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a PBKalpha and/or mTOR mediated disease such as cancer.
In a fifth aspect, the present invention is directed to compound of Formula (I) for use in therapy. Preferably the therapy is cancer. Cancers which may be treated with compounds of the present invention include, without limitation, carcinomas such as cancer of the bladder, breast, colon, rectum, kidney, liver, lung (small cell lung cancer, and non-small-cell lung cancer), esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia, acute lymphocitic leukemia, chronic lyelogenous leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T- cell-lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid lineage (including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia); tumors of mesenchymal origin (including fibrosarcoma and rhabdomyosarcoma, and other sarcomas, e.g., soft tissue and bone); tumors of the central and peripheral nervous system (including astrocytoma, neuroblastoma, glioma and schwannomas); and other tumors (including melanoma, seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi's sarcoma). Other cancers that can be treated with a compound of the present invention include endometrial cancer, head and neck cancer, glioblastoma, malignant ascites, and hematopoietic cancers.
Detailed Description
Definitions: Unless otherwise stated, the following terms used in the specification and claims are defined for the purposes of this Application and have the following meaning:
"Alkyl" means a linear saturated monovalent hydrocarbon radical of one to six carbon atoms or a branched saturated monovalent hydrocarbon radical of three to six carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl (including all isomeric forms), pentyl (including all isomeric forms), and the like.
"Alkenyl" means a linear monovalent hydrocarbon radical of two to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbon atoms containing a double bond, e.g., ethenyl, propenyl, 2-propenyl, butenyl (including all isomeric forms), pentyl (including all isomeric forms), and the like. "Alkynyl" means a linear monovalent hydrocarbon radical of two to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbon atoms containing a triple bond, e.g., ethynyl, propynyl, 2-propynyl, butynyl (including all isomeric forms), pentyl (including all isomeric forms), and the like.
"Alkylene" means a linear saturated divalent hydrocarbon radical of one to six carbon atoms or a branched saturated divalent hydrocarbon radical of three to six carbon atoms unless otherwise stated e.g., methylene, ethylene, propylene, 1-methylpropylene, 2-methylpropylene, butylene, pentylene, and the like.
"Amino" means a -NH2.
"Alkylamino" means a -NHR radical where R is alkyl as defined above, e.g., methylamino, ethylamino, propylamino, or 2-propylamino, and the like.
"Alkoxy" means a -OR radical where R is alkyl as defined above, e.g., methoxy, ethoxy, propoxy, or 2-propoxy, n-, iso-, or tert-butoxy, and the like.
"Alkylthio" means a -SR radical where R is alkyl as defined above, e.g., methylthio, and the like. "Alkylsulfonyl" means a -SO2R radical where R is alkyl as defined above, e.g., methylsulfonyl, ethylsulfonyl, and the like.
"Alkoxycarbonyl" means a -C(O)OR radical where R is alkyl as defined above, e.g., methoxycarbonyl, ethoxycarbonyl, and the like. "Alkylcarbonylamino" means a -NHC(O)R radical where R is alkyl as defined above, e.g., methylcarbonylamino, ethylcarbonylamino, and the like.
"Alkonyloxy" means a -OC(O) R radical where R is alkyl as defined above, e.g., acetyloxy, and the like.
"Alkoxyalkyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two alkoxy groups, as defined above, e.g., 2-methoxyethyl, 1-, 2-, or 3-methoxypropyl, 2-ethoxyethyl, and the like.
"Alkoxyalkyloxy" or "alkoxyalkoxy" means a -OR radical where R is alkoxyalkyl as defined above, e.g., methoxyethoxy, 2-ethoxyethoxy, and the like. "Aminoalkyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two, -NRR' where R is hydrogen, alkyl, or -CORa where Ra is alkyl, each as defined above, and R' is selected from hydrogen, alkyl, hydroxyalkyl, alkoxyalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, or haloalkyl, each as defined herein, e.g., aminomethyl, methylaminoethyl, 2-ethylamino-2-methylethyl, 1,3-diaminopropyl, dimethylaminomethyl, diethylaminoethyl, acetylaminopropyl, and the like.
"Aminoalkoxy" means a -OR radical where R is aminoalkyl as defined above, e.g., 2- aminoethoxy, 2-dimethylaminopropoxy, and the like.
"Aminosulfonyl" means a -SO2NRR' radical where R and R' are independently hydrogen or alkyl as defined above, e.g., methylaminosulfonyl, aminosulfonyl, and the like.
"Acyl" means a -COR radical where R is alkyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heterocyclyl, or heterocyclylalkyl, each as defined herein, e.g., acetyl, propionyl, benzoyl, pyridinylcarbonyl, and the like. When R is alkyl, the radical is also referred to herein as alkylcarbonyl. "Aryl" means a monovalent monocyclic or bicyclic aromatic hydrocarbon radical of 6 to
10 ring atoms e.g., phenyl or naphthyl. The aryl ring can optionally be substituted with one, two or three substituents independently selected from alkyl, hydroxy, alkoxy, or halo, and/or one or two substituents independently selected from -C(=O)Ry, -C(=O)ORy, -C(=O)NRXRX, -C(=NRX)NRXRX, -ORX, -OC(=O)RX, -0C(=0)NRxRx, -OC(=O)N(RX)S(=O)2RX, -O- alkylNRxRx, -O-alkylORx, -SRX, -S(=O)Ry, -S(=O)2Ry, -S(=O)2NRXRX, -S(=O)2N(Rx)C(=O)Ry, -S(=O)2N(Rx)C(=O)ORy, -S(=O)2N(RX)C(=O)NRXRX, -NRXRX, -N(Rx)C(=O)Ry, - (CRxRx)mN(Rx)C(=O)Ry, -N(Rx)C(=O)ORy, -N(RX)C(=O)NRXRX, -N(RX)C(=NRX)NRXRX, -N(Rx)S(=O)2Ry, -N(RX)S(=O)2NRXRX, -alkylNRxRx, -NRxC2.6alkylNRxRx, -NRxC2.6alkyl0Rx, -N(Rx)(CRxRx)m-Y, -(CRxRx)mY, or -(CRxRx)mORx where m is 0-3, each Rx is hydrogen or Ry and Ry is independently hydrogen, alkyl, cycloalkyl, phenyl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl group is substituted with one, two or three substitutents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino, or dialkylamino and Y is aryl, heteroaryl, or heterocyloalkyl substituted with 0, 1 , or 2 substitutents independently selected from alkyl, halo, haloalkoxy, hydroxyl, haloalkyl -CN, nitro, acyl, carboxy, alkoxycarbonyl, hydroxyalkyl, alkoxyalkyl, hydroxyalkoxy, aminoalkyl, or aminoalkoxy. In compounds of Formula (Ia), aryl is not substituted with - (CRxRx)mN(Rx)C(=O)Ry, either alone or in combination with other substituents.
"Aralkyl" means a -(alkylene)-R radical where R is aryl as defined above. "Cycloalkyl" means a cyclic saturated monovalent hydrocarbon radical of three to ten carbon atoms, e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, and the like.
Cycloalkylalkyl" means a -(alkylene)-R radical where R is cycloalkyl as defined above; e.g., cyclopropylmethyl, cyclobutylmethyl, cyclopentylethyl, or cyclohexylmethyl, and the like. "Carboxy" means -COOH.
"Dialkylamino" means a -NRR' radical where R and R' are independently alkyl as defined herein, e.g., dimethylamino, and the like.
"Halo" means fluoro, chloro, bromo, or iodo, preferably fluoro or chloro. "Haloalkyl" means alkyl radical as defined above, which is substituted with one or more halogen atoms, preferably one to five halogen atoms, preferably fluorine or chlorine, including those substituted with different halogens, e.g., -CH2Cl, -CF3, -CHF2, -CH2CF3, -CF2CF3, - CF(CH3)2, and the like. When the alkyl is substituted with only fluoro, it is referred to in this Application as fluoroalkyl.
"Haloalkoxy" means a -OR radical where R is haloalkyl as defined above e.g., -OCF3, - OCHF2, and the like. When R is haloalkyl where the alkyl is substituted with only fluoro, it is referred to in this Application as fluoroalkoxy.
"Hydroxyalkyl" means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to six carbons substituted with one or two hydroxy groups, provided that if two hydroxy groups are present they are not both on the same carbon atom. Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxy ethyl, 2-hydroxypropyl, 3-hydroxypropyl, l-(hydroxymethyl)-2- methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxybutyl, 2,3-dihydroxypropyl, 1- (hydroxymethyl)-2-hydroxyethyl, 2,3-dihydroxybutyl, 3,4-dihydroxybutyl and 2- (hydroxymethyl)-3-hydroxypropyl, preferably 2-hydroxyethyl, 2,3-dihydroxypropyl, and 1- (hydroxymethyl)-2-hydroxyethyl.
"Hydroxyalkoxy" or "hydroxyalkyloxy" means a -OR radical where R is hydroxyalkyl as defined above.
"Heterocyclyl" or "heterocycloalkyl" means a saturated or unsaturated monovalent monocyclic group of 4 to 8 ring atoms in which one or two ring atoms are heteroatom selected from N, O, or S(O)n, where n is an integer from 0 to 2, the remaining ring atoms being C. The heterocyclyl ring is optionally fused to a (one) aryl or heteroaryl ring as defined herein provided the aryl and heteroaryl rings are monocyclic. The heterocyclyl ring fused to monocyclic aryl or heteroaryl ring is also referred to in this Application as "bicyclic heterocyclyl" ring and is a subset of fused heterocyclyl. Additionally, one or two ring carbon atoms in the heterocyclyl ring can optionally be replaced by a -CO- group. More specifically the term heterocyclyl includes, but is not limited to, pyrrolidino, piperidino, homopiperidino, 2-oxopyrrolidinyl, 2-oxopiperidinyl, morpholino, piperazino, tetrahydropyranyl, thiomorpholino, and the like. When the heterocyclyl ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic. When the heterocyclyl group contains at least one nitrogen atom, it is also referred to herein as heterocycloamino and is a subset of the heterocyclyl group. When the heterocyclyl group is a saturated ring and is not fused to aryl or heteroaryl ring as stated above, it is also referred to herein as saturated monocyclic heterocyclyl. The heterocyclyl ring can optionally be substituted with one, two or three substituents independently selected from alkyl, hydroxy, alkoxy, or halo, and/or one or two substituents independently selected from -C(=O)Ry, -C(=O)ORy, -C(=O)NRXRX, -C(=NRX)NRXRX, -ORX, -OC(=O)RX, -OC(=O)NRXRX, -OC(=O)N(RX)S(=O)2RX, -O- alkylNRxRx, -O-alkylORx, -SRX, -S(=O)Ry, -S(=O)2Ry, -S(=O)2NRXRX, -S(=O)2N(Rx)C(=O)Ry, -S(=O)2N(Rx)C(=O)ORy, -S(=O)2N(RX)C(=O)NRXRX, -NRXRX, -N(Rx)C(=O)Ry, -N(Rx)C(=O)ORy, -N(RX)C(=O)NRXRX, -N(RX)C(=NRX)NRXRX, -N(Rx)S(=O)2Ry,
-N(RX)S(=O)2NRXRX, -alkylNRxRx, -NRxC2.6alkylNRxRx, -NRxC2.6alkyl0Rx, -N(Rx)(CRxRx)m- Y, -(CRxRx)mY, or -(CRxRx)mORx where m is 0-3, each Rx is hydrogen or Ry and Ry is independently hydrogen, alkyl, cycloalkyl, phenyl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl group is substituted with one, two or three substitutents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino, or dialkylamino and Y is aryl, heteroaryl, or heterocyloalkyl substituted with 0, 1, or 2 substitutents independently selected from alkyl, halo, haloalkoxy, hydroxyl, haloalkyl -CN, nitro, acyl, carboxy, alkoxycarbonyl, hydroxyalkyl, alkoxyalkyl, hydroxyalkoxy, aminoalkyl, or aminoalkoxy. Heterocyclylalkyl" means a -(alkylene)-R radical where R is heterocyclyl ring as defined above e.g., tetraydrofuranylmethyl, piperazinylmethyl, morpholinylethyl, and the like.
"Heteroaryl" means a monovalent monocyclic or bicyclic aromatic radical of 5 to 10 ring atoms where one or more, preferably one, two, or three, ring atoms are heteroatom selected from N, O, or S, the remaining ring atoms being carbon. Representative examples include, but are not limited to, pyrrolyl, thienyl, thiazolyl, imidazolyl, furanyl, indolyl, isoindolyl, oxazolyl, isoxazolyl, benzothiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazolyl, tetrazolyl, and the like. The heteroaryl ring can optionally be substituted with one, two or three substituents independently selected from alkyl, hydroxy, alkoxy, or halo, and/or one or two substituents independently selected from -C(=O)Ry, -C(=O)ORy, -C(=O)NRXRX, -C(=NRX)NRXRX, -ORX, -OC(=O)RX, -OC(=O)NRXRX, -OC(=O)N(RX)S(=O)2RX, -O-alkylNRxRx, -O-alkylORx, -SRX, -S(=O)Ry, -S(=O)2Ry, -S(=O)2NRXRX, -S(=O)2N(Rx)C(=O)Ry, -S(=O)2N(Rx)C(=O)ORy, -S(=O)2N(RX)C(=O)NRXRX, -NRXRX, -N(Rx)C(=O)Ry, -N(Rx)C(=O)ORy, -N(RX)C(=O)NRXRX, -N(RX)C(=NRX)NRXRX, -N(Rx)S(=O)2Ry, -N(RX)S(=O)2NRXRX, -NRxC2.6alkylNRxRx,
-NRxC2.6alkyl0Rx, -N(Rx)(CRxRx)n-Y, -(CRxRx)nY, or -(CRxRx)nORx where n is 0-3, each Rx is hydrogen or Ry and Ry is independently hydrogen, alkyl, cycloalkyl, phenyl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl group is substituted with one, two or three substitutents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino, or dialkylamino and Y is aryl, heteroaryl, or heterocyloalkyl substituted with 0, 1, or 2 substitutents independently selected from alkyl, halo, haloalkoxy, hydroxyl, haloalkyl - CN, nitro, acyl, carboxy, alkoxycarbonyl, hydroxyalkyl, alkoxyalkyl, hydroxyalkoxy, aminoalkyl, or aminoalkoxy.
"Heteroaralkyl" means a -(alkylene)-R radical where R is heteroaryl as defined above. "Substituted alkyl" means a alkyl radical as defined above in which one, two, or three hydrogen atoms are independently replaced by hydroxyl, alkoxy, or halo, and/or one or two hydrogen atoms are independently replaced by -C(=O)Ry, -C(=O)ORy, -C(=O)NRXRX, -C(=NRX)NRXRX, -ORX, -OC(=O)RX, -OC(=O)NRXRX, -OC(=O)N(RX)S(=O)2RX, -O- alkylNRxRx, -O-alkylORx, -SRX, -S(=O)Ry, -S(=O)2Ry, -S(=O)2NRXRX, -S(=O)2N(Rx)C(=O)Ry, -S(=O)2N(Rx)C(=O)ORy, -S(=O)2N(RX)C(=O)NRXRX, -NRXRX, -N(Rx)C(=O)Ry, -N(Rx)C(=O)ORy, -N(RX)C(=O)NRXRX, -N(RX)C(=NRX)NRXRX, -N(Rx)S(=O)2Ry, -N(RX)S(=O)2NRXRX, -NRxC2.6alkylNRxRx, -NRxC2.6alkyl0Rx,-N(Rx)(CRxRx)m-Y, - (CRxRx)mY, or -(CRxRx)mORx where m is 0-3, each Rx is hydrogen or Ry and Ry is independently hydrogen, alkyl, cycloalkyl, phenyl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl group is substituted with one, two or three substitutents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino, or dialkylamino and Y is aryl, heteroaryl, or heterocyloalkyl Rx is hydrogen or Ry and Ry is independently hydrogen, alkyl, cycloalkyl, phenyl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl group is substituted with one, two or three substitutents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino, or dialkylamino and Y is aryl, heteroaryl, or heterocyloalkyl substituted with 0, 1, or 2 substitutents independently selected from alkyl, halo, haloalkoxy, hydroxyl, haloalkyl -CN, nitro, acyl, carboxy, alkoxycarbonyl, hydroxyalkyl, alkoxyalkyl, hydroxyalkoxy, aminoalkyl, or aminoalkoxy. The term "oxo", when used as a substitutent, means the =0 group, which is typically attached to a carbon atom.
"Optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, "heterocyclyl group optionally substituted with an alkyl group" means that the alkyl may but need not be present, and the description includes situations where the heterocyclyl group is substituted with an alkyl group and situations where the heterocyclyl group is not substituted with alkyl. A "pharmaceutically acceptable salt" of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. Such salts include: acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as formic acid, acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4- hydroxybenzoyl)benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 4,4'-methylenebis-(3-hydroxy-2-ene-l-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, tromethamine, JV-methylglucamine, and the like. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, which is incorporated herein by reference.
The term "pharmaceutically acceptable" means that the referenced substance, such as a compound of Formula I or a salt of a compound of Formula lor a formulation containing a compound of Formula I or a particular excipent, are suitable for administration to a patient.
The term "excipient" means any pharmaceutically acceptable additive, carrier, diluent, adjuvant, or other ingredient, other than the active pharmaceutical ingredient (API), which is typically included for formulation and/or administration to a patient. The phrase in the definition of groups Ar1 in the claims and in the specification of this
Application "....each ring substituted with Rd, Re, or Rf where Rd, Re, or Rf are independently selected from hydrogen, halo, " and similar phrases used for others groups in the claims and in the specification with respect to the compound of Formula (I) means that the rings can be unsubstituted, mono-, di-, or trisubstituted unless indicated otherwise. "Sulfonylamino" means -NHSO2R' where R' is alkyl.
"Treating" or "treatment" of a disease includes: preventing the disease, i.e. causing the clinical symptoms of the disease not to develop in a mammal that may be exposed to or predisposed to the disease but does not yet experience or display symptoms of the disease; inhibiting the disease, i.e., arresting or reducing the development of the disease or its clinical symptoms; or relieving the disease, i.e., causing regression of the disease or its clinical symptoms. A "therapeutically effective amount" means the amount of a compound of Formula (I) that, when administered to a mammal for treating a disease, is sufficient to effect such treatment for the disease. The "therapeutically effective amount" will vary depending on the compound, the disease and its severity and the age, weight, etc., of the mammal to be treated.
The term "prodrug" means compounds that are transformed in vivo to yield a compound of the present invention. The transformation may occur by various mechanisms, such as through hydrolysis in blood. A discussion of the use of prodrugs is provided by T. Higuchi and W. Stella, "Pro-drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987.
To illustrate, if the compound of the invention contains a carboxylic add functional group, a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the add group with a group such as (Ci-C8 alkyl, (C2-C I2)alkanoyloxymethyl, 1- (alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1 -methyl- l-(alkanoyloxy)ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1- (alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1 -methyl- l-(alkoxycarbonyloxy)- ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, l-(N-(alkoxycarbonyl)aminomethyl having from 4 to 10 carbon atoms, 3- phthalidyl, 4-crotonolactonyl, gamma-butyrolacton-4-yl, di-N,N-(Ci-C2)alkylamino(C2- C3)alkyl (such as β-dimethylaminoethyl), carbamoyl-(Ci-C2)alkyl, N5N-CIi(C1- C2)alkylcarbamoyl-(Ci-C2)alkyl and piperidino-, pyrrolidino- or morpholino(C2-3)alkyl. Similarly, if a compound of the present invention comprises an alcohol functional group, a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as (Ci-C6)alkanoyloxymethyl, l-((Ci-C6)alkanoyloxy)ethyl, 1-methyl-l- ((Ci-C6)alkanoyloxy)ethyl, (Ci-C6)alkoxycarbonyloxymethyl, N-(Ci-Ce)alkoxycarbonyl- aminomethyl, succinoyl, (Ci-Ce)alkanoyl, α-amino(Ci-C4)alkanoyl, arylacyl and α-aminoacyl, or α-aminoacyl-α-aminoacyl, where each α-aminoacyl group is independently selected from the naturally occurring L-amino acids, -P(O)(OH)2, -P(O)(O(C 1-C6)alkyl)2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate).
The compounds of the present invention may contain asymmetric or chiral centers, and therefore, exist in different stereoisomeric forms. It is contemplated that all stereoisomeric forms of the compounds as well as mixtures thereof, including racemic mixtures, form part of the present invention. In addition, the present invention contemplates all geometric and positional isomers. For example, if the compound contains a double bond, both the cis and trans forms (designated as Z and E, respectively), as well as mixtures, are contemplated. Mixture of stereoisomers, such as diastereomeric mixtures, can be separated into their individual stereochemical components on the basis of their physical chemical differences by known methods such as chromatography and/or fractional crystallization. Enantiomers can can also be separated by converting the enantiomeric mixture into a diasteromeric mixture by reaction with an appropriate optically active compound (e.g., an alcohol), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Also, some compounds may be atropisomers (e.g., substituted biaryls).
The compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water (hydrate), ethanol, and the like. The present invention contemplates and encompasses both the solvated and unsolvated forms.
It is also possible that compounds of the present invention may exist in different tautomeric forms. All tautomers of compounds of the present invention are contemplated. For example, all of the tautomeric forms of the imidazole moiety are included in this invention. Also, for example, all keto-enol or imine-enamine forms of the compounds are included in this invention.
Those skilled in the art will recognize that the compound names and structures contained herein may be based on a particular tautomer of a compound. While the name or structure for only a particular tautomer may be used, it is intended that all tautomers are encompassed by the present invention, unless stated otherwise.
The present invention also includes isotopically-labelled compounds, which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine and chlorine, such as 2H, 3H, 13C, 14C, 15N, 16O, 170, 31P, 32P, 35S, 18F, and 36Cl.
Compounds of the present invention that contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of this invention. Certain isotopically- labelled compounds of the present invention, for example those into which radioactive isotopes such as 3H and 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detection. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labelled compounds of this invention can generally be prepared by substituting a readily available isotopically labelled reagent for a non- isotopically labelled reagent.
In synthesizing compounds of the present invention, it may be desirable to use certain leaving groups. The term "leaving groups" ("LG") generally refer to groups that are displaceable by a nucleophile. Such leaving groups are known in the art. Examples of leaving groups include, but are not limited to, halides (e.g., I, Br, F, Cl), sulfonates (e.g., mesylate, tosylate), sulfides (e.g., SCH3), N-hydroxsuccinimide, N-hydroxybenzotriazole, and the like. Examples of nucleophiles include, but are not limited to, amines, thiols, alcohols, Grignard reagents, anionic species (e.g., alkoxides, amides, carbanions) and the like.
Embodiments:
A. In one embodiment, the compounds of Formula (Ia) are those wherein:
R1 is hydrogen and 1S a ring of formula
Figure imgf000017_0002
Figure imgf000017_0001
where R is methyl and Ra is hydrogen, alkyl, or cyanomethyl, preferably hydrogen or methyl.
B. In another embodiment, the compounds of Formula (Ia) are those wherein:
R1 is hydrogen and
Figure imgf000017_0003
is a ring of formula:
Figure imgf000017_0004
C. In another embodiment, the compounds of Formula (Ia), and in embodiments A, B, C above and groups contained therein, the compounds of Formula (Ia) are those wherein R1 is hydrogen and Ar1 is aryl substituted as defined in the Summary. Within this group, in one group of compounds Ar1 is phenyl substituted at the 3 -position of the pheny ring with hydroxyl or 4-position with -N(Ra)C(=0)NRaRa, or -NHC0Rb. D. In another embodiment the compounds of Formula (Ia) and in embodiments A, B, C above and groups contained therein, the compounds of Formula (Ia) are those wherein R1 is hydrogen and Ar1 is heteroaryl substituted as defined in the Summary. Within this group of compounds in one group, Ar1 is pyrazolyl, preferably pyrazol-3-yl or pyrazol-5-yl, substituted as defined in the Summary. Within this group of compounds in another group, Ar1 is pyridyl, benzoxazolyl, pyrimidinyl, indazolyl, indolyl, benzthiazolyl, pyrazinyl, pyridazinyl, isobenzoxazolyl, or isobenzthiazolyl, each ring substituted as defined in the Summary. Preferably, Ar1 is pyridyl, benzoxazolyl, pyrimidinyl, indazolyl, indolyl, benzthiazolyl, pyrazinyl, pyridazinyl, isobenzoxazolyl, isobenzthiazolyl, each ring substituted with Rd selected from hydrogen, -N(Ra)C(=O)NRaRa, -NHCORb, amino, -ORa (where Ra is hydrogen or alkyl), haloalkyl, or halo.
E. In another embodiment the compounds of Formula (Ia) and in embodiments A, B, C and D, above and groups contained therein, the compounds of Formula (Ia) are those wherein
Figure imgf000018_0001
is a ring of formula:
Figure imgf000018_0002
preferably
Figure imgf000018_0003
Figure imgf000018_0004
(0 where R6, RJ and Rk are as defined in the Summary.
Preferably, R6 is hydrogen, alkyl, substituted alkyl, carboxy, ORa, halo, haloalkyl, heterocycloalkyl, -(alkylene)heterocycloalkyl, -( alkylene)nO(alkylene)naryl, -( alkylene)nN(Ra)(alkylene c)naryl, -(alkylene)nN(Ra)(alkylene)nC5_8heteroaryl, alkenyl, -C0NRaRa or -NRaRa. Preferably, RJ is hydrogen or alkyl and Rk is heteroaryl or aryl. F. In another embodiment, the compounds of Formula (I) are those wherein:
Figure imgf000019_0001
where R1 is as defined in the Summary, preferably R1 is Rd selected from hydrogen, halo, haloalkyl, substituted alkyl, aryl, heteroaryl, heterocycloalkyl, -C(=O)NRaRa, -C(=O)ORb, - CORb, -ORa, -O-alkylNRaRa, -O-alkylORa, -NRaRa, or -NRa-alkylene-ORa ; each Ra is independently hydrogen or Rb or when two Ra are attached to a nitrogen atom, either alone or part of another group, the two Ras together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring, each Rb is independently alkyl, phenyl, or heteroaryl wherein the alkyl is substituted with 0, 1, 2 or 3 substituents independently selected from alkyl or alkoxy.
Preferably, R1 is Rd where Rd is as defined above and wherein the aryl ring can optionally be substituted with one, two or three substituents independently selected from alkyl, alkoxy, or halo, and/or one or two substituents independently selected from, -C(=O)ORy, -C(=O)NRXRX, -ORX, -SRX, -S(=O)2Ry, -S(=O)2NRXRX, -NRXRX, -N(Rx)C(=O)Ry, -(CRxRx)mN(Rx)C(=O)Ry or -(CRxRx)mORx where m is 1-3, each Rx is hydrogen or Ry and Ry is independently hydrogen, alkyl, or cycloalkyl; the heterocycloalkyl ring is optionally be substituted with one, two or three substituents independently selected from alkyl, hydroxy, -C(=O)Ry, -C(=O)ORy, -C(=O)NRXRX, -S(=O)2Ry, or -(CRxRx)mORx where m is 0-3, each Rx is hydrogen or Ry and Ry is independently hydrogen or alkyl; the heteroaryl ring is optionally be substituted with one, two or three substituents independently selected from alkyl, -ORX, -C(=O)NRXRX, or -(CRxRx)mORx where m is 1-3, each Rx is hydrogen or Ry and Ry is independently hydrogen or alkyl; and substituted alkyl is an alkyl radical in which one, two, or three hydrogen atoms are independently replaced by hydroxyl or -NCORy where Ry is alkyl.
Preferably R1 is hydrogen, 1-piperazinyl, 4-methyl-l-piperazinyl, 4-(l-methylethyl)-l- piperazinyl, 4-(2-methoxy ethyl)- 1-piperazinyl, fluoro, 3-(methylsulfonyl)phenyl, 4-pyridinyl, 2-methoxyethoxy, 4-morpholinyl, 2-(l-pyrrolidinyl)ethoxy, l-tert-butoxycarbonyl-3,6- dihydro-l(2H)-pyridin-4-yl, 3-methoxyphenyl, 3-fluoro-4-methylaminocarbonylphenyl, A- acetylaminomethylphenyl, l-(2-methoxy ethyl)- lH-pyrazol-4-yl, 2-fluorophenyl, 2- methylphenyl, 2-methoxy-3-pyridinyl, 3-furanyl, 3,5-dimethyl-4-isoxazolyl, 4-fluoro-2- methylphenyl, 2-(l-methylethoxy)phenyl, 3-(methylsulfanyl)phenyl, l,2,3,6-tetrahydro-4- pyridinyl, 3-methyl-5-isoxazolyl, l-(2-methoxyethyl)-lH-pyrazol-4-yl, 3-acetylaminophenyl, 3-cyclopropylaminophenyl, 3-isopropanoylaminophenyl, 3-pyridazinyl, 1 -methyl- 1,2, 3,6- tetrahydro-4-pyridinyl, 4-acetylaminophenyl, 2-methyl-4-pyridinyl, 2-methyl-3 -pyridinyl, 5- pyrimidinyl, 3-pyridinyl, phenylamino, 4-methoxyphenylamino, methoxy, 3 -fluorophenyl, A- methylaminocarbonylphenyl, pyridine-3-ylamino, 4-acetylaminophenyl, 3-methoxy- phenylamino, pyridine-4-ylamino, pyrimidin-5-ylamino, 4-acetylaminophenylamino, 2- methyl-4-pyridinylamino, 4-methyl-3 -pyridinyl, 2,6-dimethyl-4-pyridinyl, 4-isopropylamino- carbonylphenyl, 3,4-dihydro-l(2H)-isoquinolinone-6-yl, 4-ethylaminocarbonylphenyl, A- carboxy-2-methylphenyl, 4-methyl-5 -pyrimidinyl, 2-methyl-4-methylaminocarbonylphenyl, hydroxyl, 6-methylaminocarbonylpyridin-3-yl, 2-(l-pyrrolidinyl)ethoxy, 4-N,N- dimethylaminocarbonylpiperazin-1-yl, 4-methylsulfonylpiperazin-l-yl, 4-N- methylaminocarbonylpiperazin- 1 -yl, 4-acetylpiperazin- 1 -yl, 3 -hydroxymethyl-4- methoxyphenyl, pyridine-4-yl, 3,6-dihydro-2H-pyran-4-yl, 2-methyl-4-pyridinyl, 1-methyl- lH-imidazol-5-yl, 3,4-dihydroxybutyl, l-(l-methylethyl)-lH-pyrazol-4-yl, aminocarbonyl, quinolin-7-yl, 4-methylsulfonylphenyl, 4-methylaminosulfonyl-phenyl, 3-methyl- sulfonylphenyl, 2-methoxyethylamino, 4-hydroxypiperidin-l-yl, 1 -methyl- lH-pyrazol-4-yl, 2- hydroxyprop-2-yl, acetyl, or trifluoromethyl. More preferably, R1 is hydrogen, 6-piperazin-l- yl, 8-fluoro, 6-(2-methoxyethoxy)-, 6-morpholin-4-yl-, 6-(3-methoxyphenyl)-, 6-(3-fluoro-4- methylaminocarbonylphenyl)-, 6-(4-methylcarbonylaminomethylphenyl)-, 6-( 1 -(2- methoxyethylpyrazol-4-yl)-, 6-(2-fluorophenyl)-, 6-(2-methylphenyl)-, 6-(2-methoxypyridin-3- yl)-, 6-furan-3-yl, 6-(3,5-isoxazol-4-yl)-, 6-(4-fluoro-2-methylphenyl)-, 6-(3-methylisoxazol-5- yl)-, 6-(3-acetylaminophenyl)-, 6-(3-cyclopropylcarbonylaminophenyl)-, 6-(4- isopropylcarbonylaminophenyl)-, 6-(pyridazin-3-yl)-, 6-( 1 -methyl- 1 ,2,3 ,4-tetrahydropyridin-4- yl)-, 6-(4-methylaminocarbonylphenyl)-, 6-pyridin-3-yl-, 6-phenylamino-, 7-methoxy, 6- (pyrimidin-5-yl)-, 6-(pyridine-3-ylamino)-, 6-(4-acetylaminophenyl)-, 6-(3- methoxyphenylamino)-, 6-(pyridine-4-ylamino)-, 6-(pyrimidin-5-ylamino)-, 6-(4- acetylaminophenylamino)-, 6-(2-methylpyridin-3-ylamino)-, 6-(2-methylpyridin-4-ylamino)-, 6-(4-methylpyridin-3-yl)-, 6-(2,6-dimethylpyridin-4-yl)-, 6-(3,4-dihydro-l(2H)-isoquinolon-6- yl)-, 6-(4-ethylaminocarbonylphenyl)-, 6-(4-methylpyrimidin-5-yl)-, 6-(4- methylaminocarbonyl-2-methylphenyl)-, 6-(6-methylaminocarbonylpyridin-3-yl)-, 6-(4- dimethylaminocarbonylpiperazin- 1 -yl)-, 6-(3-hydroxymethyl-4-methoxyphenyl)-, 6-pyridin-4- yl, 6-(3,6-dihydropyran-4-yl)-, 6-(2-methylpyridin-4-yl)-, 6-(l-methylimidazol-5-yl)-, 6-(l- isopropylpyrazol-4-yl)-, 6-(6-methylsulfonylphenyl)-, 6-(methylaminosulfonylphenyl)-, 6-(3- methylsulfonylphenyl)-, 6-(4-methylsulfonylpiperazin-l-yl)-, 6-(2-hydroxypropan-2-yl)-, 6- trifluoromethyl, or 7-trifluoromethyl (i.e., R1 is located at the 6-position of the bicyclic rings above unless indicated otherwise).
G. In another embodiment, within compounds of Formula (I) and embodiment F and groups contained therein, in one group of compounds Ar1 is aryl, preferably phenyl, substituted as defined above. Within this embodiment, in another group of compounds Ar1 is heteroaryl, preferably pyrazol-3-yl or pyrazol-5-yl substituted as defined above. Within this embodiment, in one group of compounds Ar1 is heterocycloalkyl substituted as defined above.
Within this group in yet another group of compounds Ar1 is an aryl, heteroaryl, or heterocyclyl ring, wherein each ring is substituted with Rd, Re, or Rf where Rd, Re, or Rf are independently selected from hydrogen, halo, haloalkyl, alkyl, alkyl substituted with -NRyRy (where Ry is alkyl) or alkoxy, -(CRyRy)nY (where Y is aryl or heteroaryl), -ORX (where Rx is hydrogen or alkyl), heteroaryl, heterocycloalkyl, -C(=O)NRaRa, -C(=O)Rb, -C(=O)ORb, -ORa, -O-alkylORa, -S(=O)2Rb, -NRaRa, -N(Ra)C(=O)Rb, -N(Ra)C(=O)NRaRa, -N(Ra)S(=O)2Rb, or -NRa-alkylene-ORa ; each Ra is independently hydrogen or Rb; or when two Ra are attached to a nitrogen atom, either alone or part of another group, the two Ras together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring with is optionally substituted with one, two or three substitutents independently selected from alkyl, alkylcarbonyl, - alkylOH, or hydroxyl; each Rb is independently alkyl, cycloalkyl, phenyl, heteroaryl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl is substituted with 0, 1, 2 or 3 substituents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino dialkylamino, hydroxyl, unsubstituted heterocycloalkyl, or phenyl. Preferably, Ar1 is 5-fluoro-6-methoxy-3-pyridinyl, lH-pyrazol-3-yl, 1 -methyl- lH-pyrazol-3-yl, 4-(3-ethylureido)phenyl, lH-pyrrolo[2,3- b]pyridin-6-yl, 6-(trifluoromethyl)-2-pyridinyl, 2-pyridinyl, lH-pyrrolo[3,2-b]pyridine-5-yl, 4- hydroxypyridin-2-yl, 4-(3-phenylureido)phenyl, 5-(3-ethylureido)pyridine-2-yl, 4-(3- methylureido)phenyl, 3-hydroxyphenyl, 4-acetylaminophenyl, 4-(3-(4-pyridinyl)ureidophenyl, 3-amino-pyrimidin-5-yl, 4-pyridazinyl, 1 -acetyl- lH-pyrazol-3-yl, 3-(methylsulfonyl)phenyl, 3- pyridinyl, 3-methyl-5-isothiazolyl, 4-(4-methyl-l-piperazinyl)phenyl, lH-pyrazolo[3,4- c]pyridin-3-yl, 4-fluoro-lH-indazol-3-yl)imidazo[l,2-b]pyridazin-2-yl, l-(2-
(dimethylamino)ethyl)- 1 H-pyrazol-3 -yl, 1 H-pyrazolo [3 ,4-b]pyridin-3 -yl, 3 -(4-methyl- 1 - piperazinyl)-phenyl, 3-acetylaminophenyl, pyridine-2-yl, 1 -ethyl- 1 H-pyrazol-3 -yl, quinolin-3- yl, isoquinolin-3-yl, 1-methylaminocarbonyl-l H-pyrazol-3 -yl, 1-methylaminocarbonyl-lH- pyrazol-5 -yl, 1 -(2-hydroxyethylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2- hydroxy ethylaminocarbonyl)- 1 H-pyrazol-5 -yl, 1 -(morpholin-4-ylcarbonyl)- 1 H-pyrazol-3 -yl, 1 -(morpholin-4-ylcarbonyl)- 1 H-pyrazol-5 -yl, 1 -(phenoxycarbonyl)- 1 H-pyrazol-3 -yl, 1 - (phenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(phenylaminocarbonyl)- 1 H-pyrazol-5 -yl, 1 - (methoxycarbonyl)- 1 H-pyrazol-3 -yl, 1 -(methoxycarbonyl)- 1 H-pyrazol-5 -yl, 1 -(phenoxycarbonyl)- 1 H-pyrazol-4-yl, 1 -(methylaminocarbonyl)- 1 H-pyrazol-4-yl, 1 -(pyridine-2-yl)- 1 H- pyrazol-3-yl, l-(methylsulfonyl)-l H-pyrazol-3 -yl, l-(benzoxazol-2-yl)-l H-pyrazol-3 -yl, 1- (benzimidazol-2-yl)-l H-pyrazol-5 -yl, l-(isopropylaminocarbonyl)-l H-pyrazol-3 -yl, 1- (benzylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(ethylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2- methoxyphenyl-aminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2-methylphenylaminocarbonyl)- 1 H- pyrazol-3 -yl, 1 -(4-methoxyphenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2-fluorophenyl- aminocarbonyl)-l H-pyrazol-3 -yl, l-(4-cyanophenylaminocarbonyl)-l H-pyrazol-3 -yl, l-(2- trifluoromethyl-phenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(3 -methylphenylaminocarbonyl)- 1 H-pyrazol-3 -yl, l-(2,5-difluorophenylaminocarbonyl)-lH-pyrazol-3-yl, l-(3,5- difluorophenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(4-methylphenylaminocarbonyl)- IH- pyrazol-3-yl, 1 -(3, 5 -dichlorophenyl-aminocarbonyl)-l H-pyrazol-3 -yl, l-(2-chloro-5- trifluoromethylphenylaminocarbonyl)-l H-pyrazol-3 -yl, l-(2,5-dichlorophenylamino- carbonyl)-l H-pyrazol-3 -yl, l-(2,3-dichloro-phenylaminocarbonyl)-lH-pyrazol-3-yl, l-(2- chloro-4-trifluoromethyl-phenylaminocarbony I)- 1 H-pyrazol-3 -yl, 1 -(benzimidazol-2-yl)- 1 H- pyrazol-3-yl, l-(imidazol-2-yl)-l H-pyrazol-5 -yl, 1-methylsulfonyl-l H-pyrazol-3 -yl, 1- aminocarbonyl- 1 H-pyrazol-3 -yl, 1 -aminocarbonyl- 1 H-pyrazol-5 -yl, 1 -azetidin- 1 -ylcarbonyl- 1 H-pyrazol-3 -yl, 1 -azetidin- 1-ylcarbonyl-l H-pyrazol-5 -yl, l-(2-methylaminoethylamino)- carbonyl- 1 H-pyrazol-3 -yl, 1 -(2-methylaminoethylamino)carbonyl- 1 H-pyrazol-5 -yl, 1 - dimethylaminocarbonyl- 1 H-pyrazol-3 -yl, 1 -dimethylaminocarbony 1- 1 H-pyrazol-5 -yl, 1 - piperazin- 1 -ylcarbonyl- 1 H-pyrazol-3 -yl, 1 -(2-methoxyethylamino)carbonyl- 1 H-pyrazol-3 -yl, 1 -(2-methoxyethylamino)carbonyl- 1 H-pyrazol-5 -yl, 1 -(2-morpholin-4-ylethylamino)- carbonyl- 1 H-pyrazol-3 -yl, 1 -(2-methylpropylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -( 1 - phenethylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(benzyl)- 1 H-pyrazol-3 -yl, 1 -(pyridine-3 - ylmethyl)- 1 H-pyrazol-3 -yl, 1 -( 1 ,4-benzoxazin-3 -(4H)-one-7-yl)- 1 H-pyrazol-3 -yl, 5 -(4- hydroxyazetidin- 1 -yl)pyridine-3 -yl, 5 -(4-(propan-2-ol)azetidin- 1 -yl)pyridine-3 -yl, quinolin-7- yl, quinolin-6-yl, isoquinolin-7-yl, 6-methoxypyridin-3-yl, 6-ethoxypyridin-3-yl, lH-indazol- 3 -yl, 5 -fluoro-6-methoxypyridin-3 -yl, 1 -(6-(2-methoxyethoxy)pyrimidin-3 -yl)- 1 H-pyrazol-3 - yl, 3-cyclopropylureidophenyl, 3-benzylcarbonylaminophenyl, 3-benzoylaminophenyl, 3-(4- chlorophenylsulfonylamino)-phenyl, 3 -methylsulfonylaminophenyl, 3 -(4-fluorophenyl- ureido)phenyl, 3-(benzylureido)-phenyl, 3-ethylaminocarbonylphenyl, 6-ethylamino- carbonylpyridin-3yl, 3-ethylcarbonyl-aminophenyl, 3-methylaminocarbonylphenyl, 3- ethylaminocarbonylphenyl, 3 -isopropylaminocarbonylphenyl, 3 -phenylaminocarbonylphenyl, 6-methylaminocarbonyl-pyridin-3yl, 6-isopropylaminocarbonylpyridin-3yl, 6-phenylamino- carbonylpyridin-3yl, 3-aminocarbonylphenyl, 4-(2-diethylaminoethylaminocarbonyl)phenyl, 4- phenylaminocarbonylphenyl, 1 -(3 -methylbutanoyl)- 1 H-pyrazol-3 -yl, 1 -isopropyloxy-carbonyl- 1 H-pyrazol-3 -yl, l-isopropyloxycarbonyl-lH-pyrazol-5-yl, 2-methoxypyridin-4-yl, 1- isopropylcarbonyl-1 H-pyrazol-3 -yl, l-isopropylcarbonyl-lH-pyrazol-5-yl, l-(3,3- dimethylbutanoyl)- 1 H-pyrazol-3 -yl, 1 -cyclohexyloxycarbonyl- 1 H-pyrazol-5 -yl, 1 - cyclohexyloxycarbonyl-1 H-pyrazol-3 -yl, 2-methoxypyridin-3-yl, 2-oxopyridin-3-yl, l-(3- methylbutyl)-l H-pyrazol-3 -yl, l-(2-methoxyethyl)-l H-pyrazol-3 -yl, lH-indazol-4-yl, 6- chloro-5 -methylsulfonylaminopyridin-3 -y 1, 6-methoxy-5 -methylsulfonylaminopyridin-3 -yl, lH-indazol-6-yl, lH-benzimidazol-6-yl, 4-(3-cyclopropylureido)phenyl, 4-piperidin-l- ylcarbonylaminophenyl, 4-pyrrolidin- 1 -ylcarbonylaminophenyl. 4-azetidin- 1 - ylcarbonylaminophenyl, 4-( 1 -methylpiperazin- 1 -ylcarbonylaminophenyl, 4-(3 -(2- methoxyethyl)ureido)phenyl, 4-morpholin-4-yl-ylcarbonylaminophenyl, 4-( 1 -acetylpiperazin- 1 -ylcarbonylaminophenyl, thiazol-3-yl, 4-(2-methylmorpholin-4-yl-ylcarbonylamino)phenyl, or 4-(3-methylmorpholin-4-yl-ylcarbonylamino)phenyl. More preferably, Ar1 is 5-fluoro-6- methoxy-3-pyridinyl, 1 H-pyrazol-3 -yl, 1 -methyl- 1 H-pyrazol-3 -yl, 4-(3-ethylureido)phenyl, 4- (3-phenylureido)phenyl, 4-(3-methylureido)phenyl, 3-acetylaminophenyl, 1- (phenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2-methylphenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2-fluorophenyl-aminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(4-cyanophenylaminocarbonyl)- 1 H- pyrazol-3-yl, 1 -(2 -trifluoromethylphenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2,5- difluorophenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2,5 -dichlorophenylamino-carbonyl)- 1 H- pyrazol-3-yl, l-(2,3-dichlorophenylaminocarbonyl)-lH-pyrazol-3-yl, l-(imidazol-2-yl)-lH- pyrazol-5-yl, 1-aminocarbonyl-l H-pyrazol-3 -yl, 1-aminocarbonyl-l H-pyrazol-5 -yl, 6- methoxypyridin-3 -yl, 6-chloro-5 -methylsulfonylaminopyridin-3 -yl, 6-methoxy-5 - methylsulfonylaminopyridin-3-yl, lH-indazol-6-yl, 4-(3-cyclopropylureido)phenyl, or 4- morpholin-4-yl-ylcarbonylaminophenyl.
H. In another embodiment, within compounds of Formula (I) and embodiments F and/or G and groups contained therein, in one group of compounds:
Figure imgf000024_0001
where Z1 is -N-, Z2 is -N- or -CR4- where R4 is Rd, - (alkylene)heterocycloalkyl, -(alkylene)nO(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -( alkylene)n-N(Ra)-(alkylene)nheteroaryl, or -( alkylene)nO(alkylene)nheteroaryl; Z3 is -N- or -CR5- where R5 is Rd, -(alkylene)naryl, -(alkylene)-heteroaryl,
-(alkylene)heterocycloalkyl, -(alkylene)nO-(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)nheteroaryl, or -(alkylene)nO(alkylene)nheteroaryl provided that only two of Z1, Z2 and Z3 can simultaneously be -N-; or when Z2 is -CR4- and Z3 is -CR5- then R4 and R5 together with the atoms to which they are attached can form ring A which is phenyl or a 5 or 6 membered heteroaryl ring and ring A is substituted with Rg or Rh where Rg or Rh are independently Rd, -(alkylene)n_ heterocycloalkyl, -(alkylene)nO(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -( alkylene)nN(Ra)(alkylene)nheteroaryl, or -(alkylene)nO(alkylene)nheteroaryl. Preferably, Z1 is -N-, Z2 is -CH-, and Z3 is -CR5- where R5 is Rd, -(alkylene)naryl, -(alkylene)-heteroaryl, -(alkylene)heterocycloalkyl, -(alkylene)nO-(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)nheteroaryl, or -(alkylene)nO(alkylene)nheteroaryl.
More preferably, Z1 is -N-, Z2 is -CH-, and Z3 is -CR5- where R5 is Rd, or - (alkylene)nN(Ra)(alkylene)n_heteroaryl; more preferably where Rdis -O-alkylORa, -NRaRa, -N(Ra)C(=O)Rb, -N(Ra)C(=O)NRaRa, or -N(Ra)S(=O)2Rb; each Ra is independently hydrogen or Rb; or when two Ra are attached to a nitrogen atom, either alone or part of another group, the two Ras together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring with is optionally substituted with one, two or three substitutents independently selected from alkyl; and each Rb is independently alkyl, cycloalkyl, phenyl, heteroaryl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl is substituted with 0, 1, 2 or 3 substituents independently selected from halo, alkyl, alkoxy, amino, cyano, or dialkylamino. Within compounds of Formula (I) and embodiments F and/or G and/or G and groups contained therein, in one group of compounds R1 is hydrogen or alkyl; preferably hydrogen; R2 is methyl.
1. In another embodiment, within compounds of Formula (I) and embodiments F and/or G and groups contained therein, in one group of compounds
Figure imgf000025_0001
4-amino-6-methyl-l,3,5-triazin-2-yl, 2-methyl-6-(phenylamino)-4- pyrimidinyl, 6-((2-fluorophenyl)amino)-2-methyl-4-pyrimidinyl, 6-((4-fluorophenyl)amino)-2- methyl-4-pyrimidinyl, 6-((4-methoxyphenyl)amino)-2-methyl-4-pyrimidinyl, 2-methyl-6-(3 - pyridazinylamino)-4-pyrimidinyl, 6-(4-methyl- 1 -piperazinyl)-3 -pyridazinylamino-4- pyrimidinyl, 6-(5-methoxy-2-pyridinyl)amino-2-methyl-4-pyrimidinyl, 6-((3- fluorophenyl)amino)-2-methyl-4-pyrimidinyl, 6-((4-methoxy-2-pyridinyl)amino)-2-methyl-4- pyrimidinyl, 6-((6-(4-morpholinyl)-3 -pyridazinyl)amino)-4-pyrimidinyl, 6-amino-2-methyl-4- pyrimidinyl, 6-((5 -(2-methoxyethoxy)-3 -pyridazinyl)amino)-2-methyl-4-pyrimidinyl, 2- methyl-6-((6-(4-morpholinyl)-3-pyridazinyl)amino)-4-pyrimidinyl, 6-((5-methoxy-3- pyridazinyl)amino)-2-methyl-4-pyrimidinyl, 2-methyl-6-((5-methyl-3-pyridazinyl)amino)-4- pyrimidinyl, 6-acetylamino-2-methylpyrimidin-4-yl, 2-methyl-6-((5-(2,2,2-trifluoroethoxy)-3- pyridazinyl)amino)-4-pyrimidinyl, 2-methyl-6-(methylamino)-4-pyrimidinyl, 2-methyl-6-(2- pyrazinylamino)-4-pyrimidinyl, 2-methyl-6-(l , 1 -dimethylureido)-4-pyrimidinyl, 2-methyl-6- (1 -ethylureido)-4-pyrimidinyl, 2-methyl-6-(4-methylpiperazin- 1 -carbonylamino)-4- pyrimidinyl, 2-methyl-6-(4-morpholin-4-carbonylamino)-4-pyrimidinyl, 2-methyl-6-( 1 H- pyrazol-3 -ylamino)-4-pyrimidinyl, 6-(dimethylamino)-2-methyl-4-pyrimidinyl, 2-methyl-6- (1 ,3-thiazol-2-ylamino)-4-pyrimidinyl, 2-methyl-6-(2-pyridinylamino)-4-pyrimidinyl, 2- methyl-6-(3-pyridazinylamino)-4-pyrimidinyl, 2-methyl-6-(4-pyrimidinylamino)-4- pyrimidinyl, 2-methyl-6-methylsulfonylamino-4-pyrimidinyl, 6-methyl-2-cyanomethylamino- 4-pyrimidinyl, 2-methyl-6-dimethylaminomethyl-carbonylamino-4-pyrimidinyl, 2-methyl-6- aminomethyl-carbonylamino-4-pyrimidinyl, 2-methyl-6-cyclopropylcarbonylamino-4- pyrimidinyl, 2-methyl-6-methoxymethyl-carbonylamino-4-pyrimidinyl, 2-methyl-6- cyanomethylcarbonylamino-4-pyrimidinyl, 2-methyl-6-(3-methoxybenzyl)carbonyl-amino-4- pyrimidinyl, 2-methyl-6-(3-fluorobenzyl)carbonylamino-4-pyrimidinyl, 2-methyl-6-(2,4- difluorobenzyl)carbonyl-amino-4-pyrimidinyl, 2-methyl-6-(3-cyanophenyl)carbonylamino-4- pyrimidinyl, 2-methyl-6-(3,3,3-trifluoropropanoyl)amino-4-pyrimidinyl, 6-((6-(2- methoxyethoxy)-3-pyridazinyl)amino)-2-methyl-4-pyrimidinyl, 6-((6-(2-(2-hydroxyethyl)- morpholin-4-yl)-3-pyridazinyl)amino)-2-methyl-4-pyrimidinyl, 6-((6-(2-methylmorpholin-4- yl)-3-pyridazinyl)amino)-2-methyl-4-pyrimidinyl, 6-((6-(3-methylmorpholin-4-yl)-3- pyridazinyl)amino)-2-methyl-4-pyrimidinyl, 6-((6-(morpholin-4-yl)-3 -pyridazinyl)-amino)-2- methyl-4-pyrimidinyl, 2-methyl-6-(3,3,3-trifluoropropyl)amino-4-pyrimidinyl, 2-methyl-6- (pyridine-3-yl)amino-4-pyrimidinyl, 2-methyl-6-(l,3-oxazol-5-yl)carbonylamino-4- pyrimidinyl, 2-methyl-6-(isoxazol-3-yl)amino-4-pyrimidinyl, 2-methyl-6-(l -(2-methoxy- ethyl)pyrazol-4-yl)amino-4-pyrimidinyl, 2-methyl-6-((l-((3RS)-tetrahydro-3-furanyl)-lH- pyrazol-4-yl)amino)-4-pyrimidinyl, 6-(imidazo[ 1 ,2-a]pyridin-2-ylamino)-2-methyl-4- pyrimidinyl, 6-(furo[3,2-b]pyridin-6-ylamino)-2-methyl-4-pyrimidinyl, or 6-((6-(2-methoxy-
ethoxy)-4-pyrimidinyl. Preferably,
Figure imgf000026_0001
is 4-amino-6-methyl-l,3,5-triazin-2-yl,
2-methyl-6-(3 -pyridazinylamino)-4-pyrimidinyl, 6-(4-methyl- 1 -piperazinyl)-3 - pyridazinylamino-4-pyrimidinyl, 6-((5 -(2-methoxyethoxy)-3 -pyridazinyl)amino)-2-methyl-4- pyrimidinyl, 2-methyl-6-((6-(4-morpholinyl)-3-pyridazinyl)amino)-4-pyrimidinyl, 6-((5- methoxy-3 -pyridazinyl)amino)-2-methyl-4-pyrimidinyl, 2-methyl-6-((5 -methyl-3 - pyridazinyl)amino)-4-pyrimidinyl, 6-acetylamino-2-methylpyrimidin-4-yl, 2-methyl-6- (methylamino)-4-pyrimidinyl, 2-methyl-6-(2-pyrazinylamino)-4-pyrimidinyl, 2-methyl-6-(3 - pyridazinylamino)-4-pyrimidinyl, 2-methyl-6-(4-pyrimidinylamino)-4-pyrimidinyl, 6-methyl- 2-cyanomethylamino-4-pyrimidinyl, 2-methyl-6-cyanomethylcarbonylamino-4-pyrimidinyl, 2- methyl-6-(isoxazol-3-yl)amino-4-pyrimidinyl, 2-methyl-6-(l-(2-methoxy-ethyl)pyrazol-4- yl)amino-4-pyrimidinyl, 2-methyl-6-((l-((3RS)-tetrahydro-3-furanyl)-lH-pyrazol-4-yl)amino)- 4-pyrimidinyl, or 6-((6-(2-methoxy-ethoxy)-4-pyrimidinyl.
Representative compounds of Formula (I) are:
Figure imgf000026_0002
Figure imgf000027_0001
Figure imgf000028_0001
Figure imgf000029_0001
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
General Synthetic Scheme
Compounds of this invention can be made by the methods depicted in the reaction schemes shown below.
The starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co., (Milwaukee, Wis.), Bachem (Torrance, Calif), or Sigma (St. Louis, Mo.) or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March's Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition) and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989). These schemes are merely illustrative of some methods by which the compounds of this invention can be synthesized, and various modifications to these schemes can be made and will be suggested to one skilled in the art having referred to this disclosure. The starting materials and the intermediates, and the final products of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography and the like. Such materials may be characterized using conventional means, including physical constants and spectral data.
Unless specified to the contrary, the reactions described herein take place at atmospheric pressure over a temperature range from about -78 0C to about 150 0C, more preferably from about 00C to about 1250C and most preferably at about room (or ambient) temperature, e.g., about 200C.
Figure imgf000063_0001
Compounds of Formula (I) where **— — other than pyrazo IyI ring and other groups are as defined in the Summary can be prepared as illustrated and described in Scheme 1 below.
Scheme 1
Figure imgf000063_0002
X = CI, Br, I 2
Treatment of a compound of formula 1 where LG is a leaving group such as halo, with compound of formula 2 under Suzuki coupling reaction conditions provides a compound of formula 3. The reaction is typically carried out in the presence of a transition metal catalyst (e.g., tetrakis(triphenylphosphine) palladium (0), and the like) at elevated temperature in organic solvents (e.g., ethanol, methanol, toluene, tetrahydrofuran, dioxane, and the like) and typically also requires a base (e.g., sodium carbonate, sodium acetate, and the like), and water as a co-solvent. Compounds of formula 1 and 2 are either commercially available or can be made by methods well known in the art. (please provide a list of commercially available compound within the scope and/or lit. references). Alternatively, the trialkylstannae derivative of 1 can be used instead of boronic acid derivative.
Treatment of a compound of formula 3 with an amine of formula 4 where Ar1 is as defined in the Summary, provides a compound of Formula (I) where R1 is hydrogen. Compounds of formula 4 are either commercially available or can be made by methods well known in the art. (please provide a list of commercially available compound within the scope and/or lit. references) Compounds of Formula (I) where
Figure imgf000064_0001
s pyrazol-1-yl ring where R6 is methyl and other groups are as defined in the Summary can be prepared as illustrated and described in Scheme 2 below.
Scheme 2
Figure imgf000064_0002
(i)
Treatment of a compound of formula 5 with sodium thiomethoxide, followed by treatment of resulting methylthio derivative of 5 with hydrazine in the presence of nonnucleophlic amine such as triethylamine, and the like, at high temperature provides the hydrazine compound of formula 6. Treatment of 6 with 3-aminoacrylonitrile in an alcoholic solvent such as isopropanol, and the like at high temperatures provides the 5-aminopyrazole compound of formula 7 which upon treatment with an aryl halide of formula 8 where Ar1 is as defined in the Summary in the presence of a Pd catalyst provides a compound of formula 8. The methylthio group in compound 9 is then converted to methylsulfinyl group by treating 8 with an oxidizing agent such as m-CPBA, and the like, and then treated with ammonia to give the compound of Formula (I).
Compounds of Formula (I) where R6 is ethoxycarbonyl can be prepared by reacting the compound of formula 6 with potassium (Z)-2-cyano-l-ethoxycarbonyl-ethenolate in the presence of an inorganic acid such as sulfuric acid, and the like. The compound of Formula (I) can be converted to other compounds of Formula (I). For example, hydrolysis of the ester group in a compound of Formula (I) where R6 is ethoxycarbonyl provides the corresponding compound of Formula (I) where R6 is carboxy . The carboxy group can be coverted to - CONRaRa by methods well known in the art. Utility
The compounds of the present invention are useful for the treatment of PBK and/or mTOR mediated diseases and disorders such as cancer. Cancers which may be treated with compounds of the present invention include, without limitation, carcinomas such as cancer of the bladder, breast, colon, rectum, kidney, liver, lung (small cell lung cancer, and non-small- cell lung cancer), esophagus, gall-bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage (including leukemia, acute lymphocitic leukemia, chronic lyelogenous leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non- Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma); hematopoietic tumors of myeloid lineage (including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia); tumors of mesenchymal origin (including fibrosarcoma and rhabdomyosarcoma, and other sarcomas, e.g., soft tissue and bone); tumors of the central and peripheral nervous system (including astrocytoma, neuroblastoma, glioma and schwannomas); and other tumors (including melanoma, seminoma, teratocarcinoma, osteosarcoma, xenoderoma pigmentosum, keratoctanthoma, thyroid follicular cancer and Kaposi's sarcoma). Other cancers that can be treated with a compound of the present invention include endometrial cancer, head and neck cancer, glioblastoma, malignant ascites, and hematopoietic cancers. The compounds of the present invention can also be used to treat hyperproliferative disorders such as thyroid hyperplasia (especially Grave's disease), and cysts (such as hypervascularity of ovarian stroma, characteristic of polycystic ovarian syndrome (Stein- Leventhal syndrome)).
The compounds of the present invention can also be used to treat the following diseases or conditions: asthma, chronic obstructive pulmonary disease (COPD), emphysema, psoriasis, contact dermatitis, conjunctivitis, allergic rhinitis, systemic lupus erythematosus (SLE), ulcerative colitis, Crohn's disease, multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, Alzheimer's disease, atheroscleosis and Huntington's diesese.
Administration and Pharmaceutical Composition
In addition, the compounds of the present invention can be administered alone, in combination with other compounds of the present invention, or with other pharmaceutically active compounds. The other pharmaceutically active compounds can be intended to treat the same disease or condition as the compounds of the present invention or a different disease or condition. If the patient is to receive or is receiving multiple pharmaceutically active compounds, the compounds can be administered simultaneously, or sequentially. For example, in the case of tablets, the active compounds may be found in one tablet or in separate tablets, which can be administered at once or sequentially in any order. In addition, it should be recognized that the compositions may be different forms. For example, one or more compound may be delivered via a tablet, while another is administered via injection or orally as a syrup. All combinations, delivery methods and administration sequences are contemplated. Since one aspect of the present invention contemplates the treatment of the disease/conditions with a combination of pharmaceutically active agents that may be administered separately, the invention further relates to combining separate pharmaceutical compositions in kit form. The kit comprises two separate pharmaceutical compositions: a compound of the present invention, and a second pharmaceutical compound. The kit comprises a container for containing the separate compositions such as a divided bottle or a divided foil packet. Additional examples of containers include syringes, boxes and bags. Typically, the kit comprises directions for the use of the separate components. The kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician or veterinarian.
An example of such a kit is a so-called blister pack. Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of the tablets or capsules to be packed. Next, the tablets or capsules are placed in the recesses and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a result, the tablets or capsules are sealed in the recesses between the plastic foil and the sheet. Preferably the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.
It may be desirable to provide a memory aid on the kit, e.g., in the form of numbers next to the tablets or capsules whereby the numbers correspond with the days of the regimen which the tablets or capsules so specified should be ingested. Another example of such a memory aid is a calendar printed on the card, e.g., as follows "First Week, Monday, Tuesday, . . . etc . . . Second Week, Monday, Tuesday, . . . " etc. Other variations of memory aids will be readily apparent. A "daily dose" can be a single tablet or capsule or several pills or capsules to be taken on a given day. Also, a daily dose of a compound of the present invention can consist of one tablet or capsule, while a daily dose of the second compound can consist of several tablets or capsules and vice versa. The memory aid should reflect this and aid in correct administration of the active agents.
In another specific embodiment of the invention, a dispenser designed to dispense the daily doses one at a time in the order of their intended use is provided. Preferably, the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen. An example of such a memory-aid is a mechanical counter which indicates the number of daily doses that has been dispensed. Another example of such a memory-aid is a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.
The compounds of the present invention and other pharmaceutically active agents, if desired, can be administered to a patient either orally, rectally, parenterally, (for example, intravenously, intramuscularly, or subcutaneously) intracisternally, intravaginally, intraperitoneally, intravesically, locally (for example, powders, ointments or drops), or as a buccal or nasal spray. All methods that are used by those skilled in the art to administer a pharmaceutically active agent are contemplated.
Compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions, or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents, or vehicles include water, ethanol, polyols (propylene glycol, polyethylene glycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. Microorganism contamination can be prevented by adding various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like. Prolonged absorption of injectable pharmaceutical compositions can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin. Solid dosage forms for oral administration include capsules, tablets, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, mannitol, and silicic acid; (b) binders, as for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia; (c) humectants, as for example, glycerol; (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (a) solution retarders, as for example, paraffin; (f) absorption accelerators, as for example, quaternary ammonium compounds; (g) wetting agents, as for example, cetyl alcohol and glycerol monostearate; (h) adsorbents, as for example, kaolin and bentonite; and (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, and tablets, the dosage forms may also comprise buffering agents.
Solid compositions of a similar type may also be used as fillers in soft and hard filled gelatin capsules using such excipients as lactose or milk sugar, as well as high molecular weight polyethylene glycols, and the like.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may also contain opacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedding compositions that can be used are polymeric substances and waxes. The active compounds can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. In addition to the active compounds, the liquid dosage form may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifϊers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils, in particular, cottonseed oil, groundnut oil, corn germ oil, olive oil, castor oil, and sesame seed oil, glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, or mixtures of these substances, and the like.
Besides such inert diluents, the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. Suspensions, in addition to the active compound, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, or mixtures of these substances, and the like.
Compositions for rectal administration are preferable suppositories, which can be prepared by mixing the compounds of the present invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax, which are solid at ordinary room temperature, but liquid at body temperature, and therefore, melt in the rectum or vaginal cavity and release the active component.
Dosage forms for topical administration of a compound of the present invention include ointments, powders, sprays and inhalants. The active compound or fit compounds are admixed under sterile condition with a physiologically acceptable carrier, and any preservatives, buffers, or propellants that may be required. Opthalmic formulations, eye ointments, powders, and solutions are also contemplated as being within the scope of this invention.
The compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 to about 3,000 mg per day. For a normal adult human having a body weight of about 70 kg, a dosage in the range of about 0.01 to about 100 mg per kilogram body weight is typically sufficient. The specific dosage and dosage range that can be used depends on a number of factors, including the requirements of the patient, the severity of the condition or disease being treated, and the pharmacological activity of the compound being administered. The determination of dosage ranges and optimal dosages for a particular patient is within the ordinary skill in the art.
The compounds of Formula (I) or a pharmaceutically acceptable salt thereof, may also be administered in combination with one or more additional pharmaceutically active compounds/agents. In a particular embodiment, the additional pharmaceutically active agent is an agent that can be used to treat a cancer. For example, an additional pharmaceutically active agent can be selected from antineoplastic agents, anti-angiogenic agents, chemotherapeutic agents and peptidal cancer therapy agents. In yet another embodiment, the antineoplastic agents are selected from antibiotic-type agents, alkylating agents, antimetabolite agents, hormonal agents, immunological agents, interferon-type agents, kinase inhibitors, miscellaneous agents and combinations thereof. It is noted that the additional pharmaceutically active compounds/agents may be a traditional small organic chemical molecules or can be macromolecules such as a proteins, antibodies, peptibodies, DNA, RNA or fragments of such macromolecules. Examples of specific pharmaceutically active agents that can be used in the treatment of cancers and that can be used in combination with one or more compound of the present invention include: methotrexate; tamoxifen; fluorouracil; 5-fluorouracil; hydroxyurea; mercaptopurine; cisplatin; carboplatin; daunorubicin; doxorubicin; etoposide; vinblastine; vincristine; paclitaxel; thioguanine; idarubicin; dactinomycin; imatinib; gemcitabine; altretamine; asparaginase; bleomycin; capecitabine; carmustine; cladibrine; cyclophosphamine; cytarabine; decarazine; docetaxel; idarubicin; ifosfamide; irinotecan; fludarabine; mitomycin; mitoxane; mitoxantrone; topotecan; vinorelbine; adriamycin; mithram; imiquimod; alemtuzmab; exemestane; bevacizumab; cetuximab; azacitidine; clofarabine; decitabine; desatinib; dexrazoxane; docetaxel; epirubicin; oxaliplatin; erlotinib; raloxifene; fulvestrant; letrozole; gefϊtinib; gemtuzumab; trastuzumab; gefitinib; ixabepilone; lapatinib; lenalidomide; aminolevulinic acid; temozolomide; nelarabine; sorafenib; nilotinib; pegaspargase; pemetrexed; rituximab; dasatinib; thalidomide; bexarotene; temsirolimus; bortezomib; vorinostat; capecitabine; zoledronic acid; anastrozole; sunitinib; aprepitant and nelarabine, or a pharmaceutically acceptable salt thereof. Additional pharmaceutically active agents that can be used in the treatment of cancers and that can be used in combination with one or more compound of the present invention include: epoetin alfa; darbepoetin alfa; panitumumab; pegfϊlgrastim; palifermin; filgrastim; denosumab; ancestim; AMG 102; AMG 386; AMG 479; AMG 655; AMG 745; AMG 951; and AMG 706, or a pharmaceutically acceptable salt thereof. The compounds of the present invention can also be used in combination with pharmaceutically active agents that treat nausea. Examples of agents that can be used to treat nausea include: dronabinol; granisetron; metoclopramide; ondansetron; and prochlorperazine; or a pharmaceutically acceptable salt thereof.
In addition, the compounds of the present invention can be used in combination with other agents that can be used to treat cancer such as acemannan; aclarubicin; aldesleukin; alitretinoin; amifostine; amrubicin; amsacrine; anagrelide; arglabin; arsenic trioxide; BAM 002
(Novelos); bicalutamide; broxuridine; celmoleukin; cetrorelix; cladribine; clotrimazole; DA
3030 (Dong-A); daclizumab; denileukin diftitox; deslorelin; dilazep; docosanol; doxercalciferol; doxifluridine; bromocriptine; cytarabine; HIT diclofenac; interferon alfa; tretinoin; edelfosine; edrecolomab; eflornithine; emitefur; epirubicin; epoetin beta; etoposide phosphate; exisulind; fadrozole; finasteride; fludarabine phosphate; formestane; fotemustine; gallium nitrate; gemtuzumab zogamicin; gimeracil/oteracil/tegafur combination; glycopine; goserelin; heptaplatin; human chorionic gonadotropin; human fetal alpha fetoprotein; ibandronic acid; interferon alfa; interferon alfa natural; interferon alfa-2; interferon alfa-2a; interferon alfa-2b; interferon alfa-Nl; interferon alfa-n3; interferon alfacon-1; interferon alpha natural; interferon beta; interferon beta-la; interferon beta-lb; interferon gamma natural; interferon gamma-la; interferon gamma-lb; interleukin-1 beta; iobenguane; irsogladine; lanreotide; LC 9018 (Yakult); leflunomide; lenograstim; lentinan sulfate; letrozole; leukocyte alpha interferon; leuprorelin; levamisole + fluorouracil; liarozole; lobaplatin; lonidamine; lovastatin; masoprocol; melarsoprol; metoclopramide; mifepristone; miltefosine; mirimostim; mismatched double stranded RNA; mitoguazone; mitolactol; mitoxantrone; molgramostim; nafarelin; naloxone + pentazocine; nartograstim; nedaplatin; nilutamide; noscapine; novel erythropoiesis stimulating protein; NSC 631570 octreotide; oprelvekin; osaterone; paclitaxel; pamidronic acid; peginterferon alfa-2b; pentosan polysulfate sodium; pentostatin; picibanil; pirarubicin; rabbit antithymocyte polyclonal antibody; polyethylene glycol interferon alfa-2a; porfϊmer sodium; raltitrexed; rasburicase; rhenium Re 186 etidronate; RII retinamide; romurtide; samarium (153 Sm) lexidronam; sargramostim; sizofiran; sobuzoxane; sonermin; strontium-89 chloride; suramin; tasonermin; tazarotene; tegafur; temoporfin; teniposide; tetrachlorodecaoxide; thymalfasin; thyrotropin alfa; toremifene; tositumomab-iodine 131; treosulfan; tretinoin; trilostane; trimetrexate; triptorelin; tumor necrosis factor alpha natural; ubenimex; bladder cancer vaccine; Maruyama vaccine; melanoma lysate vaccine; valrubicin; verteporfm; virulizin; zinostatin stimalamer; abarelix; AE 941 (Aeterna); ambamustine; antisense oligonucleotide; bcl-2 (Genta); APC 8015 (Dendreon); dexaminoglutethimide; diaziquone; EL 532 (Elan); EM 800 (Endorecherche); eniluracil; etanidazole; fenretinide; filgrastim SDOl (Amgen); galocitabine; gastrin 17 immunogen; HLA-B7 gene therapy (Vical); granulocyte macrophage colony stimulating factor; histamine dihydrochloride; ibritumomab tiuxetan; ilomastat; IM 862 (Cytran); interleukin-2; iproxifene; LDI 200 (Milkhaus); leridistim; lintuzumab; CA 125 monoclonal antibody(MAb) (Biomira); cancer MAb (Japan Pharmaceutical Development); HER-2 and Fc MAb (Medarex); idiotypic 105AD7 MAb (CRC
Technology); idiotypic CEA MAb (Trilex); LYM-I -iodine 131 MAb (Techniclone); polymorphic epithelial mucin-yttrium 90 MAb (Antisoma); marimastat; menogaril; mitumomab; motexafm gadolinium; MX 6 (Galderma); nolatrexed; P 30 protein; pegvisomant; porfiromycin; prinomastat; RL 0903 (Shire); rubitecan; satraplatin; sodium phenylacetate; sparfosic acid; SRL 172 (SR Pharma); SU 5416 (SUGEN); TA 077 (Tanabe); tetrathiomolybdate; thaliblastine; thrombopoietin; tin ethyl etiopurpurin; tirapazamine; cancer vaccine (Biomira); melanoma vaccine (New York University); melanoma vaccine (Sloan Kettering Institute); melanoma oncolysate vaccine (New York Medical College); viral melanoma cell lysates vaccine (Royal Newcastle Hospital); or valspodar. It is noted that the agents recited above may also be administered as pharmaceutically acceptable salts when appropriate.
The compounds of the present invention may also be used in combination with radiation therapy, hormone therapy, surgery and immunotherapy, which therapies are well know to those skilled in the art.
EXAMPLES
The examples presented below illustrate specific embodiments of the present invention. These examples are meant to be representative and are not intended to limit the scope of the claims in any manner. The starting materials for the specific examples below are generally available from commercial sources, unless otherwise specified. When helpful, commercial sources may be specifically indicated.
Analytical Methods: Unless otherwise indicated, HPLC analyses were run on an Agilent Model 1100 system with an Agilent Technologies Zorbax SB-Cs(5 μ) reverse phase column (4.6 x 150 mm) run at 30 0C with a flow rate of about 1.50 mL/min (Agilent Technologies, Santa Clara, CA). The mobile phase used solvent A (H2(M).1% TFA) and solvent B (ACN/0.1% TFA) with a 11 min gradient from 5% to 100% ACN. The gradient was followed by a 2 min. return to 5% ACN and about a 2.5 min. re-equilibration (flush).
LC-MS Methods:
Unless otherwise indicated, samples were run on an Agilent model- 1100 LC-MSD system with an Agilent Technologies XDB-Cg (3.5μ) reverse phase column (4.6 x 75 mm) at 30 0C. The flow rate was constant and ranged from about 0.75 mL/min to about 1.0 mL/min. The mobile phase used a mixture of solvent A (H2CVO.1% HCO2H or TFA) and solvent B (ACN/0.1% HCO2H or TFA) with a 5 to 9 min time period for a gradient from 10% to 90% solvent B. The gradient was followed by a 0.5 min period to return to 10% solvent B and a 2.5 min 10% solvent B re-equilibration (flush) of the column. Preparative HPLC Methods:
Where indicated, compounds of the present invention were purified via reverse phase HPLC using a Gilson (Gilson, Middleton, WI) or Shimadzu (Columbia, MD) workstation utilizing one of the following two protocols: (A) Using a 50 x 100 mm column (Waters,
Exterra, C 18, 5μ) (Waters, Milford, MA) at 50 mL/min. The mobile phase used was a mixture of solvent A (H2CVlO mM ammonium carbonate at pH about 10, adjusted with cone. NH4OH) and solvent B (85:15 ACN/water, 10 mM ammonium carbonate at pH of about 10 adjusted with cone. NH4OH). Each purification run utilized a >10 min gradient from 40% to 100% solvent B followed by a 5 min flow of 100% solvent B. The gradient was followed by a 2 min return to 40% solvent B; or (B) Using a Waters 20 x 50 mm column at 20 mL/min or Phenomenex Gemni 5μ Cl 8 100 x 30mm (Phenomenex, Torrance, CA). The mobile phase used was a mixture of solvent A (H2O/0.1% TFA) and solvent B (ACN/0.1% TFA) with a >10 min gradient from 5% to 100% solvent B. The gradient is followed by a 2 min return to 5% ACN.
Proton NMR Spectra:
Unless otherwise indicated, all 1H NMR spectra were run on a Varian (Varian, Palo Alto, CA) series Mercury 300 MHz instrument or a Bruker (Bruker, Bilerica, MA) series 400MHz instrument. Where so characterized, all observed protons are reported as parts-per- million (ppm) downfield from tetramethylsilane (TMS) or other internal reference in the appropriate solvent indicated.
Mass Spectra (MS) Unless otherwise indicated, all mass spectral data for starting materials, intermediates and/or exemplary compounds are reported as mass/charge (m/z), having an (M+H+) or (M-H") molecular ion, depending on the inonization mode (positive or negative). The molecular ion reported was obtained by electrospray detection method. Compounds having an isotopic atom, such as bromine and the like, are reported according to the detected isotopic pattern, as appreciated by those skilled in the art.
The following abbreviations may be used herein: Ac2O acetic anhydride ACN acetonitrile
A-phos Bis[(di-tert-butylphenyl phosphine)]palladium dichloride aq aqueous
ATP adenosine 5 '-triphosphate
Calcd or Calc'd calculated
Cone. concentrated
DCM dichloromethane
DMAP dimethyl aminopyridine
DME dimethoxyl ethyl ether
DMF N,N-dimethylformamide
DMSO dimethyl sulfoxide
DTT dithiothreitol
ESI electrospray ionization
Et2O diethyl ether
Et3N triethylamine
EtOAc ethyl acetate
EtOH ethyl alcohol
FBS fetal bovine serum g grams h hour
HCO2H formic acid
Hex hexanes
HOAc acetic acid
HPLC high pressure liquid chromatography
IPA or iPrOH or iPr isopropyl alcohol
IPr2NEt N-ethyl diisopropylamine
KOAc potaisum hydroxyacetate
LCMS, LC-MS or
LC/MS liquid chromatography mass spectroscopy m/z mass divided by charge
MeCN acetonitrile
MeI iodomethane
MeOH methyl alcohol mg milligrams min minutes mL milliliters
MS mass spectra
MsCl mesylchloride
NMP 1 -methyl-2-pyrrolidinone
NMR nuclear magnetic resonance
PG protecting group
PIP2 phosphatidylinositol bisphosphate
Pos. ion positive ion py or pyr pyridine rt or PvT room temperature
Sat. saturated
TFA trifuoroacetic acid
TFAA trifluoroacetic anhydride
THF tetrahydrofuran
TMS tetramethylsilane
Ts or tosyl para-toluene sulfonyl
TsCl para-toluene sulfonyl chloride wt Weight
Percents of solid reagents specified are percent by weight with respect to the total weight, and percents of solvents are specified by percent by volume with respect to the total volume, unless indicated otherwise.
Example 1 Synthesis of 6-chloro-2-methyl-9-(tetrahydro-2H-pyran-2-yl)-9H-purine
Figure imgf000076_0001
Step l
2-Methyl-4,6-dichloro-5-aminopyrimidine (Aldrich, 1.05 g) and ammonium hydroxide
(3.0 mL, J.T. Baker, Phillipsburg, NJ, 28.0% - 30.0 %) were placed in a microwave vial. The vial was sealed and heated in a CEM microwave reactor (CEM Corporation, Matthews, NC) at
120 0C and 40 Watts for 25 minutes. The reaction mixture was cooled to room temperature. This procedure was repeated a total of nine times using the following amounts of 2-methyl-4,6- dichloro-5-aminopyrimidine under the same reaction conditions:
Run 2: 1.027 g. 2.5 mL ammonium hydroxide.
Run 3: 1.034 g, 2.5 mL ammonium hydroxide.
Run 4: 1.118 g, 2.6 mL ammonium hydroxide. Run 5 : 1.117 g, 2.5 mL ammonium hydroxide.
Run 6: 1.149 g, 2.7 mL ammonium hydroxide.
Run 7: 1.264 g, 2.6 mL ammonium hydroxide.
Run 8: 1.106 g, 2.6 mL ammonium hydroxide.
Run 9: 1.075 g, 2.7 mL ammonium hydroxide.
All the runs were combined and concentrated to give 6-chloro-2-methylpyrimidine-4,5-diamin
. MS (ESI pos. ion) m/z: 159. Calcd. MS: 158.
Step 2
6-Chloro-2-methylpyrimidine-4,5-diamine (8.89 g, 56.1 mmol, the material from Step 1) was suspended in ethyl ortho formate (100 mL, 601 mmol) in a flask fitted with a reflux condenser and placed in a preheated oil bath (100 0C) and stirred for 75 minutes. The reaction mixture was cooled to room temperature, concentrated, treated with hexanes and filtered. The solid washed with hexanes and dried to give 6-chloro-2-methyl-9H-purine. MS (ESI pos. ion) m/z: 169. Calcd: 168. Step 3
6-Chloro-2-methyl-9H-purine (9.45 g, 56 mmol, the material from Step 2) was suspended in DCM (100 mL) and p-toluenesulfonic acid (Acros Organics, Geel, Belgium, 12% in acetic acid, 0.90 mL, 5.6 mmol) and 2,3-dihydropyran (6.6 mL, 73 mmol) were added. The reaction flask was fitted with a reflux condenser and placed in a preheated oil bath (50 0C) and stirred under nitrogen for 30 minutes. The reaction mixture was cooled to room temperature and stirred overnight. After stirring overnight, the reaction mixture was diluted with DCM and treated with saturated sodium bicarbonate (75 mL). The layers were separated, and the aqueous phase was extracted with DCM. The organic extracts were combined, dried over sodium sulfate, filtered, concentrated, and dried under high vacuum at 45 0C (in a water bath) and then at room temperature and then at 60 0C and finally at room temperature again to afford 6-chloro-2-methyl-9-(tetrahydro-2H-pyran-2-yl)-9H-purine (13.73 g, 97% over 3 steps). MS (ESI pos. ion) m/z: 253. Calcd. MS: 252. 1H NMR (CDCl3, 400 MHz) δ 8.26 (s, IH), 5.78 (d, J = 10.56 Hz, IH), 4.19 (d, J = 11.93 Hz, IH), 3.84 - 3.76 (m, IH), 2.80 (s, 3H), 2.20 - 1.96 (m, 3H), 1.89 - 1.64 (m, 3H).
Example 2
Synthesis of 4-chloro-6-methyl- 1 -(tetrahydro-2H-pyran-2-yl)- 1 H-pyrazolo [3 ,4-d]pyrimidine
Figure imgf000077_0001
Step l
LDA was prepared by dropwise addition of n-butyllithium, 2.5 M solution in hexanes (Aldrich, St. Louis, MO) (14.7 mL, 36.8 mmol) to N,N-diisopropylamine (5.42 mL, 38.4 mmol) in THF (40 mL) cooled in an ice bath. The LDA solution was cooled to -78 0C and a solution of 4,6-dichloro-2-methylpyrimidine (Aldrich, St. Louis, MO) (5.448 g, 33.4 mmol) in THF (50 mL) was added dropwise over 1 h. A dark solution was obtained. A solution of N- methyl-N-(2-pyridyl)formamide (TCI Tokyo Kasei Kogyo Co., Ltd.) (4.80 mL, 40.1 mmol) in THF (20 mL) was added dropwise to the solution at -78 0C over 20 min. The resulting solution was stirred for 30 min and then quenched with a solution of acetic acid (2.10 mL, 36.8 mmol) in THF (20 mL) added dropwise at -78 0C over 10 min. The solution was stirred at -78 0C for 30 min. The resulting solution of 4,6-dichloro-2-methylpyrimidine-5-carbaldehyde was treated dropwise with a solution of anhydrous hydrazine (1.10 rnL, 35.07 mmol) at -78 0C. The reaction mixture was stirred for 15 min, and then the cooling bath was removed and the reaction mixture stirred at RT for 1 h. The reaction mixture was concentrated and partitioned between water (110 mL) and EtOAc (110 mL). The organic layer was washed with saturated aqueous NaHCO3 (100 mL), separated, dried (MgSO4), treated with activated charcoal and filtered through a plug of silica, washing with EtOAc. The filtrate was concentrated and purified by flash chromatography on silica eluting with 5% acetone / DCM to 25% EtOAc / hexane. The residue was suspended in DCM (3 mL), cooled in a freezer, and filtered to give 4- chloro-6-methyl-lH-pyrazolo[3,4-d]pyrimidine (330 mg, 5.86% yield) as a tan solid. 1H NMR (400 MHz, d6-DMSO) δ 14.25 (bs, 1 H); 8.35 (s, 1 H); 2.68 (s, 3 H). Step 2
A suspension of 4-chloro-6-methyl-lH-pyrazolo[3,4-d]pyrimidine (325 mg, 1.93 mmol) in EtOAc (3 mL) was treated with 3,4-dihydro-2H-pyran (0.53 mL, 5.78 mmol) and MP-TsOH resin (Biotage) (72 mg, 4.3 mmol/g, 0.15 eq) and the resulting suspension was heated at 90 0C for 3 h, after which time LCMS indicated essentially complete conversion. The solution was filtered, washed with EtOAc, and concentrated to give a pale yellow oil. Purification by flash chromatography (EtOAc / hexane; 5% to 20%) gave 4-chloro-6-methyl-l- (tetrahydro-2H-pyran-2-yl)-lH-pyrazolo[3,4-d]pyrimidine (482 mg, 98.9% yield) as a colorless oil. 1H NMR (400 MHz, d6-DMSO) δ 8.44 (s, 1 H); 5.91 - 6.02 (m, 1 H); 3.91 - 3.99 (m, 1 H); 3.67 - 3.77 (m, 1 H); 2.72 (s, 3 H); 2.36 - 2.48 (m, 1 H); 1.96 - 2.08 (m, 1 H); 1.87 - 1.95 (m, 1 H); 1.32 - 1.84 (m, 3 H).
Example 3 Synthesis of 2,4-cichloro-6-methyl-l,3,5-triazine
Figure imgf000078_0001
Methylmagnesium bromide, 3M in ether (Aldrich, St. Louis, MO) (10.0 mL, 30 mmol) was added slowly to a white suspension of 2,4,6-trichloro-l,3,5-triazine (Aldrich, St. Louis, MO) (3.68 g, 20 mmol) in DCM (25.0 mL, 389 mmol) at 0 0C and the resulting yellow suspension was warmed up to room temperature and stirring was continued until disappearance of starting material (TLC, KMnO4 stain, 3 h). The reaction mixture was carefully quenched with NH4Cl(aq) at 0 0C and then diluted with water and DCM (25.0 mL). The separated organic layer was dried, filtered and concentrated to give 2,4-dichloro-6-methyl-l,3,5-triazine as a yellow solid (2.94 g, 90%) which was used for further reaction without purification. 1H- NMR (CDCl3, 400 MHz) δ 2.74 (s, 3 H).
Example 4
Synthesis of 2-chloro-4-methyl-6-(methylthio)-l,3,5-triazine
Cl
^ Η SMe
Sodium methanethiolate (0.49 g, 7.0 mmol) was added portionwise at 0 0C to a stirred cloudy solution of 2,4-dichloro-6-methyl-l,3,5-triazine (1.04 g, 6.3 mmol) in toluene (10 mL, 94 mmol) over 15 min. After addition, the pale yellow mixture was stirred at the same temperature for another 1 h, and water (10 mL) was added. The separated aqueous layer was extracted with EtOAc and the combined organic layers were washed with brine, dried over Na2SO4, and concentrated to give the crude residue which was purified with flash column chromatography (hexanes to 70 % DCM in hexanes) to give 2-chloro-4-methyl-6-(methylthio)- 1,3,5-triazine (0.87 g, 78% yield) as a white solid. MS (API-ES) m/z 176 (M+H)+; 1H NMR (d6-DMSO, 400 MHz) δ 2.55 (s, 3 H) 2.51 (br. s., 3 H).
Example 5
Synthesis of 4-chloro-6-methyl-l ,3,5-triazin-2-amine
Cl
SN "NH,
A solution of ammonia, 2.0M methyl alcohol (Aldrich, St. Louis, MO) (36.0 mL, 72 mmol) was added dropwise at room temperature (slightly exothermic) to a stirred yellow suspension of 2,4-dichloro-6-methyl-l,3,5-triazine (2.9373 g, 18 mmol) in toluene (20.0 mL, 188 mmol) over 1.5 h. The resulting mixture was stirred for an additional 2.5 h, concentrated and purified (ISCO, DCM τo 10 % MeOH in DCM) to give the desired product 4-chloro-6- methyl-l,3,5-triazin-2-amine (1.88 g, 73%) as a yellow solid. MS (API-ES) m/z 145 (M+H)+; 1U NMR (CD3OD, 300 MHz) δ 2.32 (s, 3H). Example 6
Synthesis of 6-chloro-2-methylpyrimidin-4-amine
Figure imgf000080_0001
Ammonia, 2.0 M in methyl alcohol (6.0 mL, 12 mmol) was added to a stirred yellow suspension of 4,6-dichloro-2-methylpyrimidine (Aldrich) (0.487 g, 3 mmol) in 1,4-dioxane (10.0 mL) at room temperature. The resulting mixture was sealed and stirred at 70 0C overnight. After cooling, the reaction mixture was concentrated and the crude residue was dissolved in DCM/MeOH and mixed with SiO2. The solvent was evaporated and the residue was purified by flash column chromatography (pure DCM τo 10% MeOH in DCM) to give 6- chloro-2-methylpyrimidin-4-amine (0.25 g, 58%) as a white solid. MS (API-ES) m/z 144 (M+H)+.
Example 7
Synthesis of 6-chloro-5-fluoro-2-methylpyrimidin-4-amine
Figure imgf000080_0002
A mixture of 4,6-dichloro-5-fluoro-2-methylpyrimidine (1.55 g, 8.6 mmol) in aqueous ammonium hydroxide (10.00 mL, 90 mmol) and MeOH (1.00 mL, 25 mmol) was heated at 70 0C for 2 h (sealed tube). After cooling, 10 mL water was added and stirred for 30 min. The solid was isolated, washed with water, and dried to give the desired product 6-chloro-5-fluoro- 2-methylpyrimidin-4-amine (0.9244 g, 67%) as a white solid. MS (API-ES) m/z 163 (M+H)+.
Example 8
Synthesis of 2-methyl-4-(methylthio)-6-(tributylstannyl)pyrimidine
Figure imgf000080_0003
Step 1 n-Butyllithium solution, 1.6 M in hexane (0.184 niL, 2.200 mmol, Aldrich, St. Louis, MO) was added to a solution of diisopropylamine (0.314 mL, 2.200 mmol) in THF (5 mL) at 0 0C. The reaction mixture was stirred at 0 0C for 15 min. Tributyltin hydride (0.527 mL, 2.000 mmol, Aldrich, St. Louis, MO) was added dropwisely. The solution was stirred at 0 0C for 15 min. The reaction mixture was cooled down to -78 0C, 4,6-dichloro-2-methylpyrimidine (326 mg, 2.000 mmol, Aldrich, St. Louis, MO) in THF (2 mL) was then added and the mixture was stirred at -78 0C for 8 h. The reaction mixture was quenched by saturated aqueous KF (4 mL), and extracted with EtOAc (30 mL). The organic extract was washed with saturated NaCl (5 mL) and dried over Na2SO4. The solution was filtered and concentrated in vacuo to give 4- chloro-2-methyl-6-(tributylstannyl)pyrimidine as a light-yellow oil. The crude material was used directly in the next step without purification. Step 2
Sodium thiomethoxide (140 mg, 2 mmol) was added to 4-chloro-2-methyl-6- (tributylstannyl)pyrimidine (835 mg, 2 mmol) in tetrahydrofuran (10 mL). The reaction mixture was stirred at room temperature for 48 h. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc. The organic extract was washed with saturated NaCl and dried over Na2SO4. The solution was filtered and concentrated in vacuo to give the crude material as an orange oil. The crude product was purified by silica gel chromatography, eluting with 5% EtOAc/hexanes to give 2-methyl-4-(methylthio)-6-(tributylstannyl)-pyrimidine (256 mg, 30 % yield). 1H NMR (300 MHz, CDCl3) δ 7.07 (s, 1 H); 2.62 (s, 3 H); 2.51 (s, 3 H); 1.44 - 1.70 (m, 6 H); 1.21 - 1.42 (m, 6 H); 0.98 - 1.21 (m, 6 H); 0.91 (t, 9 H).
Example 9 Synthesis of 4-chloro-N,N-bis(4-methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine
Cl
-^N N(PMB)2
Cesium carbonate (0.860 mL, 10.74 mmol) was added to a mixture of 4-chloro-6- methyl-l,3,5-triazin-2-amine (1.10 g, 7.61 mmol) and l-(chloromethyl)-4-methoxybenzene (1.10 mL, 8.11 mmol) in DMF (8.0 mL) at rt. After 40 min, more l-(chloromethyl)-4- methoxybenzene (1.10 mL, 8.11 mmol) was added. After another 1 h, more cesium carbonate (0.860 mL, 10.74 mmol) was added. After another 30 min, the reaction mixture was diluted with EtOAc (30 niL) and filtered through a pad of Celite® (diatomaceous earth). The filtrate was transferred to a separatory funnel, diluted with more EtOAc, and washed with water. The organic layer was dried over Na2SO4 and concentrated. The resulting slurry was filtered and washed with 1 : 1 hexane-EtOAc. The filtrate was purified by chromatography on silica using 5 to 100% DCM in hexane to give the product as a soft white solid (1.8 g). LCMS (ES, pos.): cacld for C20H2IClN4O2: 384.1; found: 385.1 (M+H)+. 1H NMR (400 MHz, CDCl3) δ 7.16 (t, J=8.12 Hz, 4 H); 6.81 - 6.95 (m, 4 H); 4.74 (s, 2 H); 4.69 (s, 2 H); 3.81 (s, 6 H); 2.45 (s, 3 H).
Example 10 Synthesis of 6-(5-(6-methoxypyridin-3-ylamino)-3 -methyl- lH-pyrazol-1 -yl)-2- methylpyrimidin-4-amine
Figure imgf000082_0001
Step l
A solution of 4,6-dichloro-2-methylpyrimidine (5 g, 31 mmol, Aldrich, St. Louis, MO) in THF (40 mL) was treated with sodium thiomethoxide (2.6 g, 37 mmol, Aldrich, St. Louis, MO) and the suspension was stirred at room temperature for 2 h. LCMS indicated a mixture of 4-chloro-2-methyl-6-(methylthio)pyrimidine and 2-methyl-4,6-bis(methylthio)pyrimidine. The reaction was carried on as such for the next step without work up. m/z (ESI, +ve ion) 175 (M+H)+. Step 2
The reaction mixture containing 4-chloro-2-methyl-6-(methylthio)pyrimidine from Step 1 was treated with triethylamine (8.5 mL, 61 mmol) and anhydrous hydrazine (1.5 mL, 46 mmol, Aldrich, St. Louis, MO) and the suspension was heated at reflux for 4 h.. Filtration of the cooled mixture gave a white solid which was washed by hexane, methanol and water, and air-dried to give l-(2-methyl-6-(methylthio)pyrimidin-4-ylhydrazine (3.5 g, 67% yield) as a white crystalline solid, m/z (ESI, +ve ion) 171 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 8.14 (s, 1 H); 6.37 (s, 1 H); 4.29 (s, 2 H); 2.41 (s, 3 H); 2.26 (s, 3 H). Step 3
A glass microwave reaction vessel was charged with l-(2-methyl-6-(methylthio)- pyrimidin-4-yl)hydrazine (3 g, 18 mmol), 3-aminocrotononitrile (6 g, 70 mmol, Aldrich, St. Louis, MO) and isopropanol (10 rnL). The reaction mixture was sealed and heated at 100 0C for 15 h. After cooling to room temperature, the suspension was filtered. The filtrate was concentrated in vacuo and the residue was treated with hexane and filtered to provide a yellow solid. The crude product was chromatographed through a RediSep®, Teledyne ISCO, Lincoln, NE, pre-packed silica gel column (40 g) eluting with a gradient of 2-5 % MeOH in CH2Cl2 to give 3 -methyl- l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-lH-pyrazol-5 -amine (2.1 g, 51% yield) as a white solid, m/z (ESI, +ve ion) 236 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 7.35 (s, 1 H); 6.90 (s, 2 H); 5.25 (s, 1 H); 2.07 (s, 3 H). Step 4
A glass microwave reaction vessel was charged with tris(dibenzylideneacetone) dipalladium (0) (78 mg, 85 μmol, Aldrich, St. Louis, MO), 2-dicyclohexylphosphino-2,4,6,-tri- i-propyl-l,l-biphenyl (82 mg, 170 μmol, Strem Chemical, Inc., Newburyport, MA ), t-butanol (3 mL) and H2O (0.12 mL). Ar was bubbled in for 1 minute. The reaction mixture was sealed and heated at 110 0C for 2 minutes and then cooled to room temperature. 3 -Methyl- 1 -(2- methyl-6-(methylthio)pyrimidin-4-yl)-lH-pyrazol-5 -amine (200mg, 850 μmol), 5-bromo-2- methoxypyridine (320 mg, 1.7 mmol, Aldrich, St. Louis, MO) and cesium carbonate (554 mg, 1.7mmol, Aldrich, St. Louis, MO) were added and the reaction mixture was stirred and heated at 120 0C for 2.5 h. After cooling to room temperature, the mixture was filtered through a Celite® (diatomaceous earth) pad and the pad was washed with 10% methanol in DCM. The crude product was concentrated and chromatographed through a RediSep®, Teledyne ISCO, Lincoln, NE, pre-packed silica gel column (40 g) eluting with a gradient of 10-100% EtOAc in hexane to give 6-methoxy-N-(3 -methyl- 1 -(2 -methyl-6-(methylthio)pyrimidin-4-yl)- IH- pyrazol-5-yl)pyridin-3 -amine (160mg, 55% yield) as a brown solid, m/z (ESI, +ve ion) 343 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 10.13 (s, 1 H); 8.15 (d, J=2.74 Hz, 1 H); 7.70 (dd, J=8.80, 2.93 Hz, 1 H); 7.43 (s, 1 H); 6.85 (d, J=8.80 Hz, 1 H); 5.80 (s, 1 H); 3.84 (s, 3 H); 2.64 (s, 3 H); 2.58 (s, 3 H); 2.16 (s, 3 H). Step 5
A solution of 6-methoxy-N-(3 -methyl- 1 -(2 -methyl-6-(methylthio)pyrimidin-4-yl)- IH- pyrazol-5-yl)pyridin-3 -amine (140 mg, 0.4 mmol) in CH2Cl2 (10 mL) was cooled to 0 0C and treated with 3-chloroperoxybenzoic acid (0.2 g, 77%, 0.8 mmol, Aldrich, St. Louis, MO). The reaction mixture was stirred at 0 0C for 1 h and then quenched with aqueous sodium thoisulfate. The aqueous layer was extracted with CH2Cl2 and the combined organic extracts were washed with saturated NaHCO3 and brine, and then dried over Na2SO4, filtered and concentrated in vacuo. T he residue was chromatographed through a RediSep®, Teledyne ISCO, Lincoln, NE, pre-packed silica gel column (40 g) eluting with a gradient of 0-5 % MeOH in CH2Cl2 to give 6-methoxy-N-(3 -methyl- 1 -(2 -methyl-6-(methylsulfinyl)pyrimidin-4- yl)-lH-pyrazol-5-yl)pyridin-3 -amine (0.1 g, 68% yield) as a yellow solid, m/z (ESI, +ve ion) 359 (M+H)+. Step 6
A glass microwave reaction vessel was charged with 6-methoxy-N-(3 -methyl- 1 -(2- methyl-6-(methylsulfinyl)pyrimidin-4-yl)- 1 H-pyrazol-5 -yl)pyridin-3 -amine (100 mg, 279 μmol) dissolved in isopropanol (3 mL) and aqueous ammonium hydroxide (28-30%, 5 mL). The solution was stirred and heated at 120 0C for 40 minutes. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was chromatographed through a RediSep®, Teledyne ISCO, Lincoln, NE, pre-packed silica gel column (40 g) eluting with a gradient of 2-5 % MeOH in DCM to give 6-(5-(6-methoxypyridin-3-ylamino)-3-methyl- lH-pyrazol-l-yl)-2-methylpyrimidin-4-amine (30 mg, 35% yield) as a white solid, m/z (ESI, +ve ion) 312 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 10.69 (s, 1 H); 8.10 (d, J=2.93 Hz, 1 H); 7.65 (dd, J=8.80, 2.93 Hz, 1 H); 7.00 (br s, 2 H); 6.83 (d, J=9.00 Hz, 1 H); 6.64 (s, 1 H); 5.77 (d, J=12.32 Hz, 2 H); 3.83 (s, 3 H); 2.42 (s, 3 H); 2.14 (s, 3 H).
Example 11
Synthesis of N-(I -(6-amino-2-methylpyrimidin-4-yl)-3 -methyl- 1 H-pyrazol-5 -yl)-lH-indazol-
4-amine
Figure imgf000084_0001
Step l
A glass microwave reaction vessel was charged with tris(dibenzylideneacetone) dipalladium(O) (195 mg, 212 μmol, Aldrich, St. Louis, MO), 2-dicyclohexylphosphino-2,4,6,- tri-i-propyl-l,l-biphenyl (203 mg, 0.425 mmol, Strem Chemical, Inc., Newburyport, MA ), t- butanol (5 mL) and H2O (0.2 mL). Ar was bubbled in for 1 minute, and then the reaction mixture was heated at 120 0C for 10 minutes and then cooled to room temperature. 3-Methyl- l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-lH-pyrazol-5 -amine (500 mg, 2.1 mmol), 4-bromo- lH-indazole (461 mg, 2.3 mmol, J&W Phamlab, Levittown, PA), and cesium carbonate (1.4 g, 4.2 mmol) were added to the reaction mixture which was then sealed and heated at 120 0C for 15 h. After cooling to room temperature, the mixture was filtered through a celite pad, washed with 50% MeOH-DCM, and the filtrate was concentrated in vacuo. The residue was chromatographed through a RediSep®, Teledyne ISCO, Lincoln, NE, pre-packed silica gel column (40 g) eluting with a gradient of 0-5 % MeOH in CH2Cl2 to give N-(3 -methyl- 1 -(2- methyl-6-(methylthio)pyrimidin-4-yl)-lH-pyrazol-5-yl)-lH-indazol-4-amine (170 mg, 22.8% yield) as an off-white solid, m/z (ESI, +ve ion) 352 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 13.25 (s, 1 H); 11.18 (s, I H); 8.17 (s, 1 H); 7.52 (s, 1 H); 7.31 (t, J=8.02 Hz, 1 H); 7.15 (d, J=8.41 Hz, 1 H); 7.00 (d, J=7.63 Hz, 1 H); 6.27 (s, 1 H); 2.74 (s, 3 H); 2.60(s, 3 H); 2.25 (s, 3 H). Step 2 A solution of N-(3 -methyl- 1 -(2-methyl-6-(methylthio)pyrimidin-4-yl)- 1 H-pyrazol-5 - yl)-lH-indazol-4-amine (150 mg, 427 μmol) in DCM-DMF (5:1, 6 mL) was cooled to 0 0C and treated with 3-chloroperoxybenzoic acid (147 mg, 77%, 658 μmol). The reaction mixture was stirred at 0 0C for 1 h and at room temperature for an additional 30 minutes. The reaction was quenched with aqueous sodium thiosulfate and extracted with CH2Cl2.. The combined organic extracts were washed with saturated NaHCO3 and brine, dried over Na2SO4, filtered and concentrated. The residue was chromatographed through a RediSep®, Teledyne ISCO, Lincoln, NE, pre-packed silica gel column (40 g) eluting with a gradient of 2-5 % MeOH in CH2Cl2 to give N-(3 -methyl- 1 -(2 -methyl-6-(methylsulfinyl)pyrimidin-4-yl)-l H-pyrazol-5 -yl)- lH-indazol-4-amine as a yellow solid, m/z (ESI, +ve ion) 368 (M+H)+. Step 3
A glass microwave reaction vessel was charged with N-(3 -methyl- 1 -(2 -methyl-6- (methylsulfϊnyl)pyrimidin-4-yl)-l H-pyrazol-5 -yl)-lH-indazol-4-amine (50 mg, 136 μmol), isopropanol and DMF (4:1, 3 mL), and NH4OH (5 mL, 30%). The vessel was sealed and heated 120 0C for 2 h. After cooling to room temperature, the mixture was concentrated in vacuo. The residue was purified by preparative HPLC (Phenomenex Synergi 4 μ MAX-RP 150 x 21.2 mm, 20 to 50 % CH3CN/ H2O, 0.1 % TFA, over 12 min) (Phenomenex, Torrance, CA) to give N-(I -(6-amino-2-methylpyrimidin-4-yl)-3 -methyl- 1 H-pyrazol-5 -y I)-I H-indazol-4- amine (5 mg, 11% yield) as a yellow solid, m/z (ESI, +ve ion) 321 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 13.17 (s, 1 H); 11.58 (s, 1 H); 8.13 (s, 1 H); 7.30 (t, J=7.92 Hz, 1 H); 7.20 (brs, 2 H); 7.11 (d, J=8.22 Hz, 1 H); 6.96 (d, J=7.43 Hz, 1 H); 6.75 (s, 1 H); 6.20 (s, 1 H); 2.55 (s, 3 H); 2.22 (s, 3 H).
Example 12 Synthesis of N-( 1 -(6-amino-2-methylpyrimidin-4-yl)- 1 H-pyrazol-5 -yl)- 1 H-indazol-4-amine
Step 1 A mixture of ethyl (ethoxymethylene)cyanoacetate (2.3 g, 14 mmol, Aldrich, St. Louis,
MO), l-(2-methyl-6-(methylthio)pyrimidin-4-yl)hydrazine (2.1 g, 12 mmol) and ethanol (50 mL) was heated at reflux for 5 h. The suspension was cooled to room temperature, solvent was removed in vacuo, and the residue was treated with water and filtered. The solid was washed with water, methanol, and hexane and air-dried to give ethyl 5-amino-l-(2-methyl-6- (methylthio)pyrimidin-4-yl)-lH-pyrazole-4-carboxylate (3.2 g, 88% yield) as a white powder. m/z (ESI, +ve ion) 294 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 7.84 (s, 1 H); 7.71 (s, 2 H); 7.44 (s, 1 H); 4.22 (q, J=7.17 Hz, 2 H); 2.61 (s, 3 H); 2.57 (s, 3 H); 1.28 (t, J=7.14 Hz, 3 H). Step 2
A mixture of ethyl 5-amino-l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-lH-pyrazole-4- carboxylate (3 g, 10 mmol) and aqueous lithium hydroxide (0.4 mL, 41 mmol, Aldrich, St. Louis, MO) in dioxane-H2O (4:1, 50 mL) was stirred and heated at 85 0C for 16 h. After cooling to room temperature, the white suspension was neutralized to PH<5 with HCl (5 N). Filtration provided 5-amino-l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-lH-pyrazole-4- carboxylic acid (2 g, 74% yield) as a white solid, m/z (ESI, +ve ion) 266 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 7.59 (s, 1 H); 7.47 (s, 2 H); 7.39 (s, 1 H); 2.58 (s, 3 H); 2.55 (s, 3 H). Step 3
A glass microwave reaction vessel was charged with 5-amino-l-(2-methyl-6- (methylthio)pyrimidin-4-yl)-lH-pyrazole-4-carboxylic acid (1.4 g, 5.4 mmol) and H2O (10 mL). The reaction mixture was stirred and heated in a Smith Synthesizer® microwave reactor (Personal Chemistry, Inc., Upssala, Sweden) at 200 0C for 1 h. The suspension was filtered to give l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-l H-pyrazol-5 -amine (1.0 g, 85% yield) as a white solid, m/z (ESI, +ve ion) 222 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 7.44 (s, 2 H); 6.96 (s, 2 H); 5.40 (d, J=I.37 Hz, 1 H); 2.58 (s, 3 H); 2.55 (s, 3 H). Step 4
A glass microwave reaction vessel was charged with 2-dicyclohexylphosphino-2,4,6,- tri-i-propyl-l,l-biphenyl (323 mg, 0.68 mmol, Strem Chemical, Inc., Newburyport, MA),
Pd2(dba)3 (350 mg, 339 mmol, Aldrich, St. Louis, MO ), t-butanol (10 mL) and H2O (0.4 mL). Ar was bubbled in for 1 minute. The tube was sealed and heated at 110 0C for 5 minutes and then cooled to room temperature. 4-Bromo-lH-indazole (490 mg, 2.5 mmol, J&W pharmlab, Levittown, PA), l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-lH-pyrazol-5 -amine (500 mg, 2.3 mmol) and cesium carbonate (1.5 g, 4.6 mmol, Aldrich, St. Louis, MO) were added and the mixture was sealed and heated at 110 0C for 16 h. After cooling to room temperature, the mixture was diluted with 50% methanol-DCM, filtered through a Celite® (diatomaceous earth) pad, and concentrated in vacuo. The residue was chromatographed through a RediSep®, Teledyne ISCO, Lincoln, NE, pre-packed silica gel column (40 g) eluting with a gradient of 2- 10 % MeOH in CH2Cl2 to give N-(l-(2-methyl-6-(methylthio) pyrimidin-4-yl)-l H-pyrazol-5 - yl)-lH-indazol-4-amine (250 mg, 33% yield) as yellow solid, m/z (ESI, +ve ion) 338 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 11.15 (s, 1 H); 8.19 (s, 1 H); 7.79 (d, J=I.76 Hz, 1 H); 7.61 (s, 1 H); 7.33 (t, J=7.92 Hz, 1 H); 7.16 (d, J=8.41 Hz, 1 H); 7.02 (d, J=7.43 Hz, 1 H); 6.40 (d, J= 1.96 Hz, 1 H); 2.76 (s, 3 H); 2.61 (s, 3 H). Step 5
A solution of N-( 1 -(2-methyl-6-(methylthio)pyrimidin-4-yl)- 1 H-pyrazol-5 -yl)- 1 H- indazol-4-amine (250 mg, 741 μmol) in DMF (5 mL) was cooled to 0 0C and treated with 3- chloroperoxybenzoic acid (250mg, 77%, 1.1 mmol). The reaction mixture was stirred at 0 0C for 1 h and at room temperature for additional 1 h, and then the reaction was quenched with sodium thiosulfate and extracted with DCM. The combined organic extracts were washed with saturated NaHCO3 and brine, dried over Na2SO4, and concentrated in vacuo to give N-(l-(2- methyl-6-(methylsulfmyl) pyrimidin-4-yl)-l H-pyrazol-5 -yl)-lH-indazol-4-amine (280 mg, 100% yield) as a brown oil. The crude product was taken on to the next step without further purification, m/z (ESI, +ve ion) 354 (M+H)+. Step 6
A glass reaction vessel was charged with N-(l-(2-methyl-6-(methylsulfinyl)pyrimidin- 4-yl)-l H-pyrazol-5 -yl)-lH-indazol-4-amine (0.28 g, 792 μmol) and ammonia (2 N in 2- propanol, 20 mL, 40 mmol). The reaction mixture was sealed and heated at 100 0C for 5h and then cooled to room temperature. The mixture was concentrated in vacuo, and the residue was chromatographed through a RediSep®, Teledyne ISCO, Lincoln, NE, pre-packed silica gel column (40 g) eluting with a gradient of 5-20 % MeOH in CH2Cl2 to give N-(l-(6-amino-2- methylpyrimidin-4-yl)-lH-pyrazol-5-yl)-lH-indazol-4-amine (60mg, 25% yield) as a yellow solid, m/z (ESI, +ve ion) 303 (M+H)+. 1U NMR (400 MHz, d6-DMSO) δ 13.20 (brs, 1 H); 11.58 (s, 1 H); 8.15 (s, 1 H); 7.69 (d, J=I.56 Hz, 1 H); 7.31 (t, J=7.92 Hz, 1 H); 7.28 (brs, 2 H); 7.12 (d, J=8.41 Hz, 1 H); 6.98 (d, J=7.63 Hz, 1 H); 6.80 (s, 1 H); 6.34 (d, J=I.76 Hz, 1 H); 2.56 (s, 3 H).
Example 13 Synthesis of 3-(3-(2-methyl-9H-purin-6-yl)imidazo[l,2-a]pyridin-2-ylamino)phenol
Figure imgf000088_0001
Step 1 Triethylamine (21.1 mL, 151 mmol) was added dropwise to a mixture of chloroacetic acid (13.0 g, 138 mmol, Alfa Aesar, Ward Hill, MA) and water (35 mL). After 10 minutes, 2- aminopyridine (15.5 g, 165 mmol, Fluka, Buchs, Switzerland) was added. The reaction mixture was heated at 90 0C. After 2 h, the solution was allowed to cool to rt. EtOH (6 mL) was added and the mixture was stirred at 5 0C for 30 min. The solution was filtered and the filtercake was washed with cold EtOH and then dried under vacuum to give a white solid (19.7 g). The white solid was added to a 500-mL round-bottomed flask and suspended in toluene (140 mL). The mixture was heated to reflux and then phosphorus oxychloride (32.4 mL, 348 mmol) was added slowly over 45 min. The reaction mixture was stirred at reflux for 17 h. It was allowed to cool to rt and slowly poured into ice water (300 mL) over 15 min and then stirred for 15 min. The phases were separated and the aqueous phase was cooled to 0 0C and neutralized with 1 N NaOH and IO N NaOH. The precipitate that formed was filtered and the filtercake was washed with water and then dried by passing air through the filter frit to afford the title compound (12.78 g, 61 %) as a pale yellow solid, m/z (ESI, +ve ion) 153 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 8.50 (d, J = 6.8 Hz, 1 H); 8.05 (s, 1 H); 7.53 (d, J = 9.2 Hz, 1 H); 7.30 - 7.35 (m, 1 H); 6.98 (t, J = 6.8 Hz, 1 H). Step 2 A glass microwave reaction vessel was charged with 2-chloroimidazo[l,2-a]pyridine (0.850 g, 5.57 mmol), 1,4-dioxane (8.5 mL), ethanol (4.25 niL), 6-chloro-2-methyl-9- (tetrahydro-2H-pyran-2-yl)-9H-purine (2.11 g, 8.36 mmol), potassium carbonate (1.54 g, 11.14 mmol), triphenylphosphine (0.073 g, 0.279 mmol) and palladium(II) acetate (0.125 g, 0.557 mmol, Strem). The reaction mixture was stirred and heated in an Emrys Optmizer microwave reactor (Personal Chemistry, Biotage AB, Inc., Upssala, Sweden) at 130 0C for 60 min. The mixture was poured into water (200 mL) and extracted with EtOAc (3 x 100 mL). Purification by silica gel chromatography (50 to 100% EtOAc/hexane) followed by elution with 2.0 to 5.0% MeOH (2 M in NH3)ZCH2Cl2 gave 6-(2-chloroimidazo[l,2-a]pyridin-3-yl)-2-methyl-9H- purine with the THP protecting group intact. This material was dissolved in 10%
MeOH/CH2Cl2 and loaded onto Si-TsOH (4 grams, Silicycle, SiliCycle Inc., Quebec City, Canda). The Si-TsOH plug was eluted with 10% MeOH/CH2Cl2 and then the cartridge was eluted with 10% MeOH(2 M NH3)/CH2C12 to elute off the product. The 10% MeOH(2 M NH3)/CH2C12 eluent was concentrated to give 6-(2-chloroimidazo[l,2-a]pyridin-3-yl)-2- methyl-9H-purine (0.24 g, 15 %) as an off-white solid, m/z (ESI, +ve ion) 285 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 13.47 (br s, 1 H); 8.90 (d, J = 6.8 Hz, 1 H); 8.56 (br s, 1 H); 7.72 (d, J = 9.0 Hz, 1 H); 7.49 - 7.58 (m, 1 H); 7.12 (t, J = 6.7 Hz, 1 H); 2.76 (s, 3 H). Step 3
A mixture of 6-(2-chloroimidazo[l,2-a]pyridin-3-yl)-2-methyl-9H-purine (0.0700 g, 0.246 mmol), 3-aminophenol (0.161 g, 1.48 mmol), ethanol (1.3 mL) and one drop of cone. HCl was heated for 4 h at 155 0C in a microwave reactor. The solution was diluted with MeOH and DMSO to make it homogeneous and was purified by Prep-HPLC (Phenomenex Gemini 5 micron, C 18, 100 x 30 mm, 5 to 50% CH3CN(0.1% TFA)/H2O(0.1% TFA) over 20 min then 100% CH3CN(0.1% TFA) for 3 minutes at 20 mL/min) with the fractions containing product concentrated to give the title compound (0.039 g, 44 %) as an orange solid, m/z (ESI, +ve ion) 358 (M+H)+. 1H NMR (400 MHz, d6-DMSO) δ 13.53 (br s, 1 H); 12.78 (s, 1 H); 10.52 (d, J=7.0 Hz, 1 H); 9.28 (s, 1 H); 8.62 (s, 1 H); 7.49 - 7.61 (m, 3 H); 7.21 (d, J=8.8 Hz, 1 H); 7.07 - 7.15 (m, 2 H); 6.37 (dd, J=7.9, 1.9 Hz, 1 H); 2.74 (s, 3 H).
Example 14
Synthesis of N-(3-(2-methyl-9H-purin-6-yl)imidazo[l,2-a]pyridin-2-yl)-lH-indazol-4-amine
Figure imgf000090_0001
A mixture of 6-(2-chloroimidazo[l,2-a]pyridin-3-yl)-2-methyl-9H-purine (0.0730 g, 0.256 mmol), lH-indazol-4-amine (0.205 g, 1.538 mmol), ethanol (1.25 rnL) and cone HCl (1 drop) was heated at 155 0C for 9 h in a microwave reactor. The reaction mixture was poured into aq. NaHCO3 and was extracted with 25% iPrOH/CHCl3. The combined extracts were washed with brine, dried (Na2SO4) and concentrated onto silica. Purification by silica gel chromatography (2.5 to 95% MeOH (2 M in NHs)ZCH2Cl2) gave a dark solid that was mainly product. Further purification by Prep-HPLC (Phenomenex Gemini 5 micron, C18, 100 x 30 mm, 5 to 55% CH3CN(0.1 % TFA)/H2O(0.1 % TFA) over 20 min then 100% CH3CN(0.1 %
TFA) for 3 minutes at 20 niL/min) gave N-(3-(2-methyl-9H-purin-6-yl)imidazo[l,2-a]pyridin- 2-yl)-lH-indazol-4-amine 2,2,2-trifluoroacetate (0.0186 g, 0.049 mmol, 19.02 % yield) as an orange solid, m/z (ESI, +ve ion) 382.0 (M+H)+.
Example 15
Synthesis of 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(lH-pyrazol-3-yl)imidazo[l,2- a]pyridin-2-amine
Figure imgf000090_0002
Step 1
In a 20 mL microwave sealed tube under N2 were dissolved 2,4-dichloro-6-methyl- 1,3,5-triazine (1612 mg, 9.83 mmol), 2-chloroimidazo[l,2-a]pyridine (750 mg, 4.92 mmol), palladium(II) acetate (110 mg, 0.492 mmol), triphenylphosphine (258 mg, 0.983 mmol) and potassium carbonate (1359 mg, 9.83 mmol) in 10 mL ofp-dioxane then stirred and heated at 150 0C with microwave for 5h. After 5h, the reaction mixture based on LC-MS showed the desired compound. The reaction mixture was diluted with DCM then neutralized with H2O. The aqueous phase was extracted with DCM:MeOH (90:10) then the organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The crude mixture was purified by MPLC (ISCO) with Hexanes:AcOEt 100:0 to 0:100 to afforded 2-chloro-3-(4-chloro-6- methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridine (750 mg, 2.68 mmol, 54.5 % yield) as a solid. m/z (ESI, +ve ion) 280.2 (M+H)+. Step 2
In a 25-mL round bottom flask under N2 were dissolved 2-chloro-3-(4-chloro-6-methyl- l,3,5-triazin-2-yl)imidazo[l,2-a]pyridine (750 mg, 2.68 mmol) in ammonia in MeOH (9M) (8.92 mL, 80 mmol) and stirred at rt. After 30 min the reaction mixture based on LC-MS showed conversion to the desired compound. The crude mixture was concentrated under reduced pressure to afforded 4-(2-chloroimidazo[l,2-a]pyridin-3-yl)-6-methyl-l,3,5-triazin-2- amine (750 mg, 2.88 mmol, 107 % yield) as a solid, m/z (ESI, +ve ion) 261.0 (M+H)+. Step 3
In a 10-mL sealed tube under N2 were dissolved 4-(2-chloroimidazo[l,2-a]pyridin-3- yl)-6-methyl-l,3,5-triazin-2-amine (750 mg, 2.88 mmol), lH-pyrazol-3 -amine (478 mg, 5.75 mmol) and TsOHH2O (821 mg, 4.32 mmol) in 5 mL of 2-BuOH then stirred and heated at 150 0C. After 5h the reaction mixture based on LC-MS showed conversion to the desired compound. The reaction mixture was diluted with MeOH and freebased with K2CO3. Silica was added to the mixture and concentrated under reduced pressure. The crude mixture was purified by MPLC (ISCO) with DCM:MeOH+NH4OH 100:0 to 90: 10 to afforded 3-(4-amino- 6-methyl-l,3,5-triazin-2-yl)-N-(lH-pyrazol-3-yl)imidazo[l,2-a]pyridin-2-amine (75 mg, 0.244 mmol, 8.48 % yield) as a off-white solid m/z (ESI, +ve ion) 308.0(M+H)+. An alternative procedure for step 3 is found in step 5 of Example 27.
Example 16
Synthesis of 3-(4-methyl-6-(methylamino)-l,3,5-triazin-2-yl)-N-(lH-pyrazol-3- yl)imidazo[l ,2-a]pyridin-2-amine
Figure imgf000091_0001
Prepared according to preparation of 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(lH- pyrazol-3-yl)imidazo[l,2-a]pyridin-2-amine using MeNH2 (2N in MeOH) in the Step 2. m/z (ESI, +ve ion) 322.2 (M+H)+
Example 17
Synthesis of 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-7-chloro-N-(lH-pyrazol-3- yl)imidazo[l ,2-a]pyridin-2-amine
Figure imgf000092_0001
Step 1
2,7-Cichloroimidazo[l,2-a]pyridine was prepared according to preparation of 2- chloroimidazo[l,2-a]pyridine in Example 15. 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(lH- pyrazol-3-yl)imidazo[l ,2-a]pyridin-2-amine using 4-chloropyridin-2-amine. m/z (ESI, +ve ion) 187.0 (M+H)+ Step 2
2,7-Dichloro-3-(4-chloro-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridine ( prepared according Org. Lett. 2009, 11, 1837-1840), was dissolved 2,7-dichloroimidazo[l,2-a]pyridine (0.6 g, 3.21 mmol) in THF (2.67 mL) and added a solution of (2,2,6,6-tetramethylpiperidin-l- yl)zinc(II) lithium chloride (11.46 mL, 4.81 mmol) dropwise and let stir for 30 min at rt under nitrogen. In a separated flask, dissolved Pd(PPh3)4 (0.371 g, 0.321 mmol) and 2,4-dichloro-6- methyl-l,3,5-triazine (0.789 g, 4.81 mmol) in THF (2.67 mL) under nitrogen and then transferred with cannula dropwise into the zinc reagent and let stir 16 hrs at rt until reaction was completed. The reaction mixture was neutralized with NH4Cl and extracted with EtOAc, dried over Na2SO4 and filtered. The reaction mixture was concentrated under reduced and the crude mixture was purified by MPLC (ISCO) with Hexanes:AcOEt 100:0 to 0:100 to afforded 2,7-dichloro-3-(4-chloro-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridine (120 mg, 0.381 mmol, 11.89 % yield) as a brown solid, m/z (ESI, +ve ion) 314.0 (M+H)+. Step 3
4-(2,7-Dichloroimidazo[l,2-a]pyridin-3-yl)-6-methyl-l,3,5-triazin-2-amine was prepared according to preparation of 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(lH-pyrazol- 3-yl)imidazo[l,2-a]pyridin-2-amine Example 15 in step 3. m/z (ESI, +ve ion) 295.0 (M+H)+ Step 4
In a 2 rnL microwave sealed tube under N2 were dissolved cesium carbonate (397 mg, 1.220 mmol), lH-pyrazol-3 -amine (101 mg, 1.220 mmol), and 4-(2,7-dichloroimidazo[l,2- a]pyridin-3-yl)-6-methyl-l,3,5-triazin-2-amine (120 mg, 0.407 mmol) in DMSO (407 μL) and heated to 100 0C. After Ih the reaction mixture based on LC-MS showed conversion to the desired compound. The reaction was filtered and directly purified via Gilson reverse phase (0.1% TFA in CH3CN/H2O, gradient 10% to 70%) and purified by MPLC (ISCO) with DCM:MeOH+NH4OH 100:0 to 90:10 to afforded after freebasing with SCX column 3-(4- amino-6-methyl-l,3,5-triazin-2-yl)-7-chloro-N-(lH-pyrazol-3-yl)imidazo[l,2-a]pyridin-2- amine (2.0 mg, 5.85 μmol, 1.5 % yield), m/z (ESI, +ve ion) 342.0 (M+H)+
Example 18
Synthesis of 3-(6-amino-2-methylpyrimidin-4-yl)-N-(lH-pyrazol-3-yl)imidazo[l ,2- b]pyridazin-2-amine
Figure imgf000093_0001
Step l
In a 250-mL round bottom flask under N2 were dissolved 2-chloroacetic acid (2.94 g, 31.1 mmol), pyridazin-3 -amine hydrochloride (4.09 g, 31.1 mmol) and triethylamine (12.58 mL, 93 mmol) in 50 mL of MeCN then stirred and heated at 80 0C. After Ih the reaction mixture was concentrated under reduced pressure and the crude 2-(6-iminopyridazin-l(6H)- yl)acetic acid was used without further purification in the next step. Step 2
In a 250-mL round bottom flask under N2 was dissolved the crude 2-(6-iminopyridazin- l(6H)-yl)acetic acid and phosphorus oxychloride (14.49 mL, 155 mmol) in 50 mL of 1,2- dichlorobenze then stirred and heated at 100 0C. After 2 h, the reaction mixture based on LC- MS showed conversion to the desired compound. The reaction mixture was diluted with DCM then neutralized with ice/H2O. The aqueous phase was extracted with DCM then the organic layer was dried over Na2SO4, filtered and concentrated under reduced pressure. The crude mixture was purified by MPLC (ISCO) with Hexanes first to remove the 1 ,2-dichlorobenzene and then Hexanes:AcOEt 100:0 to 0:100 to afforded 2-chloroimidazo[l,2-b]pyridazine (740 mg, 4.82 mmol, 15.50 % yield) as a white solid, m/z (ESI, +ve ion) 150.2 (M+H)+. Step 3
In a 20 mL microwave sealed tube under N2 were dissolved 4,6-dichloro-2- methylpyrimidine (1.974 g, 12.11 mmol), 2-chloroimidazo[l,2-b]pyridazine (620 mg, 4.04 mmol), triphenylphosphine (212 mg, 0.807 mmol), palladium(II) acetate (91 mg, 0.404 mmol) and potassium carbonate (1.674 g, 12.11 mmol) in 8 mL of/?-dioxane then stirred and heated at 130 0C. After 5 h, the reaction mixture was concentrated under reduced pressure with silica and the crude mixture was purified by MPLC (ISCO) with Hexanes:AcOEt 100:0 to 0:100 to afforded 2-chloro-3-(6-chloro-2-methylpyrimidin-4-yl)imidazo[l,2-b]pyridazine (1.1 g, 3.93 mmol, 97 % yield) contaminated with PPhβ as a yellow solid, m/z (ESI, +ve ion) 280.0 (M+H)+. Step 4
In a 20 mL microwave sealed tube under N2 was dissolved 2-chloro-3-(6-chloro-2- methylpyrimidin-4-yl)imidazo[l,2-b]pyridazine (1.1 g, 3.93 mmol) in ammonia (9 M in MeOH) (10.0 mL, 70.0 mmol) then stirred and heated at 130 0C with microwave. After 1 h, the reaction mixture was cooled at 0 0C and then the solid filtered to afforded 6-(2- chloroimidazo[l,2-b]pyridazin-3-yl)-2-methylpyrimidin-4-amine (380 mg, 1.458 mmol, 37.1 % yield) as a white solid, m/z (ESI, +ve ion) 261.2(M+H)+. Step 5 In a 10 mL microwave sealed tube under N2 were dissolved lH-pyrazol-3 -amine (319 mg, 3.84 mmol), 6-(2-chloroimidazo[l,2-b]pyridazin-3-yl)-2-methylpyrimidin-4-amine (200 mg, 0.767 mmol), Xantphos (89 mg, 0.153 mmol), Pd(dba)2(88 mg, 0.153 mmol) and cesium carbonate (1500 mg, 4.60 mmol) in 3 mL of/?-dioxane then stirred and heated at 130 0C with microwave. After 1O h, the reaction mixture was concentrated under reduced pressure with silica and purified by MPLC (ISCO) with DCM:MeOH+NH4OH 100:0 to 90: 10 and then reverse phase purified with Gilson to afforded after freebasing with SCX column 3-(6-amino- 2-methylpyrimidin-4-yl)-N-(lH-pyrazol-3-yl)imidazo[l,2-b]pyridazin-2-amine as a yellow solid, m/z (ESI, +ve ion) 308.1 (M+H)+.
An alternative procedure is described in step 5 of Example 27. Example 19
Synthesis of thyl 5 -amino- 1 -(2-methy l-6-(methy lthio)pyrimidin-4-yl)- 1 H-pyrazole-3 - carboxylate
Figure imgf000095_0001
Step 1.
Sulfuric acid (4.03 mL, 57.6 mmol) was added dropwise to water (192 mL, 57.6 mmol) in a flask using a pipette while cooling with ice water bath. To a 250-mL round-bottomed flask was added l-(2-methyl-6-(methylthio)pyrimidin-4-yl)hydrazine (9800 mg, 57.6 mmol) and potassium (Z)-2-cyano-l-ethoxycarbonyl-ethenolate (10.30O g, 57.5 mmol) and the above sulfuric acid (4.03 mL, 57.6 mmol) solution in water (192 mL, 57.6 mmol) in DCM (192 mL) (pm). The reaction mixture was stirred at room temperature for 20 hours. After the completion of reaction, the organic layer was separated and the aqueous layer was extracted with DCM. The combined organic layers were then washed with NaHCOβ, dried over Na2SOφ The residue was redissolved in 100 mL of isopropanol at 1000C, after cooling to room temperature, the desired product precipitated out of the solution. Filtration provided the desired product ethyl 5 -amino- l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-l H-pyrazole-3 -carboxylate as off- white solid (4.5 g). The mother liquor was concentrated in vacuo, and the residue was redissolved in DCM and was subject to ISCO purification. The crude material was absorbed onto a plug of silica gel and purified by chromatography through a Redi-Sep pre-packed silica gel column (40 g), eluting with a gradient of 0% to 50% EtOAc in hexane, to provide another 1.4 gram of desired product. The combined product ethyl 5-amino-l-(2-methyl-6- (methylthio)pyrimidin-4-yl)-l H-pyrazole-3 -carboxylate (5.9 g, 35.1% yield) were carried over next step, m/z 294.2 (M+H)+. Step 2
A glass microwave reaction vessel was charged with tris(dibenzylideneacetone)- dipalladium (0) (125 mg, 0.136 mmol), 2-di-tert-butylphosphino-3,4,5,6-tetramethyl-2',4',6'- triisopropyl-l,l'-biphenyl (131 mg, 0.273 mmol), tert-Butanol (6.82 mL, 1.364 mmol) , water (0.273 mL, 1.364 mmol). The vial was capped, the vacum and Ar refill process was repeated three times and the mixture was heated at 120 0C for 10 minutes. After cooling to RT, ethyl 5- amino-l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-lH-pyrazole-3-carboxylate (400 mg, 1.364 mmol), 3-bromopyrazole (220 mg, 1.500 mmol), Si-carbonate (2338 mg, 1.636 mmol) were mixed. The vial was capped, the vacum and Ar refill process was repeated three times and plastic ring was put above the reaction mixture. The vial was capped, the vacum and Ar refill process was repeated three times and the mixture was heated at 150 0C for 4 h. After cooling to room temperature, the reaction mixture was diliuted w/ EtOAc and filterred through a pad of silica gel. The residue was washed w/ EtOAc. The solution was concentrated under vacum and the crude material was purified by chromatography through a Redi-Sep pre-packed silica gel column (12 g), eluting with a gradient of 0 % to 10 % MeOH in CH2CL2, to provide ethyl 5 -( 1 H-pyrazol-3 -ylamino)- 1 -(2-methyl-6-(methylthio)pyrimidin-4-yl)- 1 H-pyrazole-3 - carboxylate (284 mg, 0.790 mmol, 57.9 % yield) as off-white solid, m/z 360.2 (M+H)+. Step 3
To a 10 mL round-bottomed flask was added 3-chloroperoxybenzoic acid 77% max. (137 mg, 0.793 mmol) to a solution of ethyl 5 -(I H-pyrazol-3 -ylamino)- 1 -(2 -methyl-6-
(methylthio)pyrimidin-4-yl)-l H-pyrazole-3 -carboxylate (190 mg, 0.529 mmol) in CH2Cl2 (5.28 mL) at 00C. The reaction mixture was slowly warmed to room temperature and stirred for 2 h. To the reaction mixture, was added aq. Na2S2θ3, followed by the addition of sat. NaHCOβ. T he reaction mixture was diluted with and extracted with DCM. The organic extract was dried over Na2SO4. The solution was filtered and concentrated in vacuo to give the crude material as a tan oil. The crude material was absorbed onto a plug of silica gel and purified by chromatography through a Redi-Sep pre-packed silica gel column (12 g), eluting with a gradient of 0 % to 10 % MeOH in CH2CL2, to provide ethyl 5 -(I H-pyrazol-3 -yl-amino)-l -(2- methyl-6-(methylsulfinyl)pyrimidin-4-yl)-l H-pyrazole-3 -carboxylate (150 mg, 0.400 mmol, 76 % yield) as tan solid, m/z (M+H)+. Step 4
A glass microwave reaction vessel was charged with ethyl 5 -(I H-pyrazol-3 -ylamino)- l-(2-methyl-6-(methylsulfinyl)pyrimidin-4-yl)-l H-pyrazole-3 -carboxylate (43.5 mg, 0.116 mmol) and ammonia, 2M solution in 2-propanol (3.86 mL, 0.116 mmol). The reaction mixture was stirred and heated in a Initiator microwave reactor (Personal Chemistry, Biotage AB, Inc., Upssala, Sweden) at 100 0C for 2 h. The crude material was purified by reverse-phase preparative HPLC using 0.1% TFA in CH3CN/H2O, gradient 10 % to 70% over 20 min to provide ethyl 5 -( 1 H-pyrazol-3 -ylamino)- 1 -(6-amino-2-methylpyrimidin-4-yl)- 1 H-pyrazole-3 - carboxylate as a off-white powder, m/z 329.0 (M+H)+. Example 20
Synthesis of 5-(lH-pyrazol-5-ylamino)-l-(6-amino-2-methylpyrimidin-4-yl)-N-cyclopropyl- lH-pyrazole-3-carboxamide
Figure imgf000097_0001
A 10 niL of round bottom flask was charged with CH2Cl2 (0.55 mL), then cyclopropanamine (19.23 μL, 0.274 mmol), the solution was cooled to 0 0C and trimethylaluminum (137 μL, 0.274 mmol) was added. The solution was stirred at the same temperature for 30 min and then at room temperature for 30 min. A suspension of ethyl 5- (1 H-pyrazol-3 -ylamino)- 1 -(6-amino-2-methylpyrimidin-4-yl)- 1 H-pyrazole-3 -carboxylate (18.0 mg, 0.055 mmol) in THF (0.2 mL) was added dropwise. The syringe was rinsed with another portion of THF (0.2 mL). The reaction mixture turned to a clear orange-red colored solution. The mixture was maintained at rt under N2 for 18 h. The reaction mixture was poured to a sat. NH4CI solution, and extracted with EtOAc. The organic extract was dried over Na2SO4. The solution was filtered and concentrated in vacuo to give the crude material as a yellow solid. The residue was diluted with MeOH/DMSO and purified by preparative HPLC (Gilson: 10- 80% (0.1% TFA in CH3CN) in H2O over 15 min). Clean fractions were combined and concentrated to afford pure 5 -(I H-pyrazol-5 -ylamino)- l-(6-amino-2 -methylpyrimidin-4-y I)-N- cyclopropyl-1 H-pyrazole-3 -carboxamide (10.6 mg, 0.031 mmol, 57.0 % yield) as a yellow amorphous solid, m/z 340.2 (M+H)+.
Example 21
Synthesis of 5 -(I H-pyrazol-3 -ylamino)- l-(6-amino-2-methylpyrimidin-4-yl)-l H-pyrazole-3 - carboxylic acid
Figure imgf000097_0002
To a 15-mL round-bottom flask was added methyl 5-(lH-pyrazol-3-ylamino)-l-(6- amino-2-methylpyrimidin-4-yl)-lH-pyrazole-3-carboxylate (30 mg, 0.095 mmol) and lithium hydroxide monohydrate (13.26 μL, 0.477 mmol) in THF (0.57 mL)/methanol (191 μL)/water (0.2 mL) at room temperature. The reaction mixture was stirred over weekend (~60h). The reaction mixture was neutralized by minimum amount of aq. sat. NaHCOβ. The reaction mixture was concentrated in vacuo and then redissolved in methanol. The solution was filtered and concentrated in vacuo to give the crude material, which was was purified by reverse-phase preparative HPLC 0.1% TFA in CH3CN/H2O, gradient 20% to 70% over 15 min to provide 5- (lH-pyrazol-3-ylamino)-l-(6-amino-2-methylpyrimidin-4-yl)-lH-pyrazole-3-carboxylic acid (20 mg, 0.067 mmol, 69.8 % yield) 104415-32-4 as yellow solid, m/z 301.2 (M+H)+.
Example 22
Synthesis of methyl 4-( 1 -(6-amino-2-methylpyrimidin-4-yl)-5 -(3 -hydroxyphenylamino)- 1 H- pyrazol-3-yl)benzoate
Figure imgf000098_0001
Step 1 l-(2-Methyl-6-(methylthio)pyrimidin-4-yl)hydrazine (2.0 g, 12 mmol), methyl 4-(2- cyanoacetyl)benzoate (2 g, 12 mmol) and acetic acid (0.7 ml, 12 mmol) were dissolved in ethanol (23 mL) in a sealed tube and heated at 100 0C overnight. After cooling to rt, the desired product crashed out from EtOH. The suspension was filtered and washed with hexanes giving methyl 4-(5-amino-l-(2-methyl-6-(methylthio)pyrimidin-4-yl)-lH-pyrazol-3-yl)benzoate as a yellow solid (4 g , 96% yield), m/z 356.2 (M+H)+. Step 2
A suspension of methyl 4-(5 -amino- l-(2-methyl-6-(methylthio)pyrimidin-4-yl)- IH- pyrazol-3-yl)benzoate (100 mg, 0.28 mmol), 3-bromophenol (73 mg, 0.42 mmol), palladium diacetate (13 mg, 56 μmol), xantphos (65 mg, 0.11 mmol) and cesium carbonate (275 mg, 0.84 mmol) inl,4-dioxane (1 mLl) in a pressure vial was degassed by a stream of nitrogen and sealed. The reaction was stirred at 150 0C for 12 hours. After cooling to rt, the reaction mixture was diluted with EtOAc, passed through a celite pad and concentrated in vacuo. The crude material was absorbed onto a plug of silica gel and purified by chromatography through a Redi-Sep pre-packed silica gel column (12 g), eluting with a gradient of 0 % to 10 % MeOH in CH2CL2, to provide Methyl 4-(5-(3-hydroxyphenylamino)-l-(2-methyl-6-(methylthio)- pyrimidin-4-yl)-lH-pyrazol-3-yl)benzoate. m/z 448.2(M+H)+. Step 3
A suspension of methyl 4-(5-(3-hydroxyphenylamino)-l-(2-methyl-6-(methylthio)- pyrimidin-4-yl)-lH-pyrazol-3-yl)benzoate (70 mg, 0.15 mmol) in dichloromethane (0.5 mL) was treated with m-chloroperoxybenzoic acid (51 mg, 0.3 mmol) at 0 0C. The reaction mixture was allowed to warm to rt for 3 hours. To the reaction mixture, was added aq. Na2S2θ3, followed by the addition of sat. NaHCOβ. The reaction mixture was extracted with DCM. The organic extract was dried over Na2SO4. The solution was filtered and concentrated in vacuo to give the crude material as a tan oil. The crude material was absorbed onto a plug of silica gel and purified by chromatography through a Redi-Sep pre-packed silica gel column (4 g), eluting with a gradient of 0 % to 10 % MeOH in CH2Cl2, to provide methyl 4-(5-(3- hydroxyphenylamino)- 1 -(2-methyl-6-(methylsulfinyl)pyrimidin-4-yl)- 1 H-pyrazol-3 - yl)benzoate. m/z 462.2. Step 4
Ammonia, 2.0M solution in 2-propanol (3.6 mL, 0.11 mmol) was added to methyl 4-(5-(3- hydroxyphenylamino)- 1 -(2-methyl-6-(methylsulfinyl)pyrimidin-4-yl)- 1 H-pyrazol-3 - yl)benzoate (50 mg, 0.11 mmol) in a microwave vial at rt. The vial was sealed and heated to 100 0C in microwave for 150 min. After cooling to room temperature, the reaction mixture was concentrated in vacuo to give the crude material, which was was purified by reverse- phase preparative HPLC 0.1% TFA in CH3CN/H2O, gradient 20% to 70% over 15 min to provide methyl 4-( 1 -(6-amino-2-methylpyrimidin-4-yl)-5 -(3 -hydroxyphenylamino)- 1 H- pyrazol-3-yl)benzoate. m/z 401.2.
Example 23
Synthesis of 3-(2-methyl-9H-purin-6-yl)-N-(lH-pyrazol-5-yl)imidazo[l,2-a]pyridin-2-amine
Figure imgf000100_0001
A glass microwave reaction vessel was charged with 6-(2-chloroimidazo[l,2-a]pyridin- 3-yl)-2-methyl-9-(tetrahydro-2H-pyran-2-yl)-9H-purine (0.222 g, 0.602 mmol, from Example 13, Step 2) and 2-butanol (6 mL). To this mixture was added 3-aminopyrazole (250 mg, 3.01 5 mmol) followed by p-toluenesulfonic acid (282 mg, 1.806 mmol). The resulting slurry was stirred and heated with microwave irradiation (Biotage initiator) at 130 0C for 1O h. The mixture was poured into saturated aqueous NaHCO3 (20 mL) and extracted with EtOAc (1x10 mL). The layers were separated and the aqueous layer was further extracted with CH2Cl2 (2x20 mL). The combined organic layers were dried (Na2SO4) and concentrated in vacuo. The0 residue was taken up in minimal MeOH/DMSO and purified by preparative HPLC (Gilson: 10- 80% (0.1% TFA in CH3CN) in H2O over 15 min) to yield 3-(2-methyl-9H-purin-6-yl)-N-(lH- pyrazol-5-yl)imidazo[l,2-a]pyridin-2-amine as the 2,2,2-trifluoroacetate salt (8.0 mg, 0.018 mmol, 2.98 % yield), m/z (ESI, +ve ion) 332.0 (M+H)+. 1U NMR (400 MHz, d6-DMSO) δ ppm 2.80 (s, 3 H); 6.75 (d, J=2.15 Hz, 1 H); 7.26 (td, J=6.92, 1.42 Hz, 1 H); 7.57 - 7.83 (m, 45 H); 8.65 (s, 1 H); 10.55 - 10.74 (m, 1 H); 13.33 (br. s., 1 H); 13.66 (br.s., 1 H).
Example 24
Synthesis of 3-(4-amino-6-methyl- 1 ,3,5-triazin-2-yl)-N-(6-methoxypyridin-3-yl)imidazo[ 1 ,2- a]pyridin-2-amine
Step l
To a sealable vial was added 2-chloroimidazo[l,2-a]pyridine (0.30 g, 1.97 mmol), chloro[2-(dicyclohexylphosphino)-3 ,6-dimethoxy-2'-4'-6'-tri-i-propyl- 1 , 1 '-biphenyl] [2-(2- aminoethyl)phenyl]palladium(II) (0.016 g, 0.020 mmol, Strem), and sodium tert-butoxide5 (0.472 g, 4.92 mmol). The mixture was carefully evacuated and backfilled with N2. This was repeated twice. To the mixture was added 1,4-dioxane (4.0 rnL) and 5-amino-2- methoxypyridine (0.25 mL, 2.36 mmol, Aldrich). The mixture was carefully evacuated and backfilled with N2. This was repeated twice. The mixture was heated and stirred at 90 0C for 1 h and then was poured into aq. NaHCO3 and extracted with EtOAc (3 x 100 mL). The combined extracts were washed with brine, dried (Na2SO4) and concentrated onto silica.
Purification by silica gel chromatography (1.0 to 6.0% MeOH (2 M in NH3)/CH2C12) afforded N-(6-methoxypyridin-3-yl)imidazo[l,2-a]pyridin-2-amine as a green solid (0.363 g, 1.51 mmol, 77% yield). MS (ESI positive ion) m/z: 241 (M+ 1). Step 2 To a 0.5-2 mL Personal Chemistry microwave vial was added was added N-(6- methoxypyridin-3-yl)imidazo[l,2-a]pyridin-2-amine (0.075 g, 0.31 mmol), 4-chloro-N,N- bis(4-methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine (0.18 g, 0.47 mmol), potassium carbonate (0.086 g, 0.62 mmol), triphenylphosphine (0.016 g, 0.062 mmol), palladium (II) acetate (7.0 mg, 0.031 mmol) and 1,4-dioxane (1.0 mL). The vial was sealed and carefully evacuated and backfilled with N2. This was repeated twice. The reaction mixture was heated at 140 0C for 3 h in the microwave. The reaction mixture was poured into aq. NaHCO3 and extracted with EtOAc. The combined extracts were washed with brine and then dried (Na2SO4) and concentrated onto silica. Purification by silica gel chromatography (0.0 to 4.5% MeOH (2 M in NH3)/CH2C12) afforded 3-(4-(bis(4-methoxybenzyl)amino)-6-methyl-l,3,5- triazin-2-yl)-N-(6-methoxypyridin-3-yl)imidazo[l,2-a]pyridin-2-amine (0.156 g, 0.265 mmol, 85% yield). MS (ESI positive ion) m/z: 589 (M+l). Step 3
To a sealable vial was added 3-(4-(bis(4-methoxybenzyl)amino)-6-methyl-l,3,5-triazin- 2-yl)-N-(6-methoxypyridin-3-yl)imidazo[l,2-a]pyridin-2-amine (0.075 g, 0.13 mmol), and a solution of trifluoromethanesulfonic acid (0.040 mL, 0.450 mmol) in trifluoroacetic acid (2.00 mL, 26.0 mmol) (~2% solution by volume). The solution was stirred at room temperature for 3 h. The reaction mixture was concentrated to about half the volume by flushing with a stream of nitrogen. The solution was then carefully pippetted into sat NaHCO3 that had been cooled in an ice-water bath. The pH of the solution was confirmed to be near neutral. A precipitate had formed and the solution was centrifuged and the liquid was pipetted off. The precipitate was washed with water and then dried under vacuum. It was then washed with EtOAc and then dried under vacuum to afford 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(6- methoxypyridin-3-yl)imidazo[l,2-a]pyridin-2-amine (0.042 g, 0.121 mmol, 95 % yield) as a light yellow solid. MS (ESI positive ion) m/z: 349 (M+l). 1H NMR (400 MHz, DMSO-J6) δ ppm 2.41 (s, 3 H), 3.85 (s, 3 H), 6.85 (d, J= 9.0 Hz, I H), 7.11 (t, J= 6.6 Hz, 1 H), 7.40 (br s, 1 H), 7.50 (t, J= 7.8 Hz, 1 H), 7.55-7.65 (m, 2 H), 8.36 (d, J= 7.8 Hz, 1 H), 8.71 (s, 1 H), 9.98 (d, J= 6.7 Hz, 1 H), 10.32 (br s, 1 H).
Example 25
Synthesis of 3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-N-(5 -fluoro-6-methoxypyridin-3 - yl)imidazo[l,2-a]pyridin-2-amine.
Figure imgf000102_0001
Step l To a 10-20 rnL Personal Chemistry microwave vial was added was added 2- chloroimidazo[l,2-a]pyridine (0.50 g, 3.3 mmol), 4-chloro-N,N-bis(4-methoxybenzyl)-6- methyl-l,3,5-triazin-2-amine (1.89 g, 4.92 mmol), potassium carbonate (0.906 g, 6.55 mmol), triphenylphosphine (0.172 g, 0.655 mmol), palladium (II) acetate (0.074 g, 0.33 mmol) and 1,4-dioxane (10 mL). The vial was sealed and carefully evacuated and backfilled with N2. This was repeated twice. The reaction mixture was heated at 140 0C for 3 h in the microwave. The reaction mixture was poured into aq. NaHCO3 (100 mL) and was extracted with EtOAc. The combined extracts were washed with brine and then dried (Na2SO4) and concentrated onto silica. Purification by silica gel chromatography (5.0 to 50% EtOAc/hexane) afforded 4-(2- chloroimidazo[l,2-a]pyridin-3-yl)-N,N-bis(4-methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine ( (0.67 g, 1.33 mmol, 41 % yield). MS (ESI positive ion) m/z: 501 (M+ 1). Step 2
To a sealable vial was added 4-(2-chloroimidazo[l,2-a]pyridin-3-yl)-N,N-bis(4- methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine (0.125 g, 0.250 mmol), chloro[2- (dicyclohexylphosphino)-3 ,6-dimethoxy-2'-4'-6'-tri-i-propyl- 1 , 1 '-biphenyl] [2-(2- aminoethyl)phenyl]palladium(II) (1.99 mg, 2.50 μmol, Strem), and sodium tert-butoxide
(0.060 g, 0.624 mmol). The mixture was carefully evacuated and backfilled with N2. This was repeated twice. To the mixture was added 1,4-dioxane (1.0 mL) and 5-fluoro-6- methoxypyridin-3 -amine (0.043 g, 0.30 mmol, Anichem, LLC, North Brunswick, NJ). The mixture was carefully evacuated and backfilled with N2. This was repeated twice. The reaction mixture was heated and stirred at 90 0C for 6 h and then was poured into aq. NaHCO3 and extracted with EtOAc. The combined extracts were washed with brine, dried (Na2SO4) and concentrated onto silica. Purification by silica gel chromatography (5.0 to 30% EtOAc/hexane) afforded 3-(4-(bis(4-methoxybenzyl)amino)-6-methyl-l,3,5-triazin-2-yl)-N-(5- fluoro-6-methoxypyridin-3-yl)imidazo[l,2-a]pyridin-2-amine (0.065 g, 43 % yield) as a yellow solid. MS (ESI positive ion) m/z: 607 (M+ 1). Step 3
To a sealable vial was added 3-(4-(bis(4-methoxybenzyl)amino)-6-methyl-l,3,5- triazin-2-yl)-N-(5-fluoro-6-methoxypyridin-3-yl)imidazo[ 1 ,2-a]pyridin-2-amine (0.060 g, 0.099 mmol) and a solution of trifluoromethanesulfonic acid (0.040 mL, 0.450 mmol) in trifluoroacetic acid (2.00 mL, 26.0 mmol) (~2% TfOH in TFA by volume). The solution was stirred at rt for 3 h. The reaction mixture was carefully pippetted into sat. aq NaHCO3 (30 mL) and the pH of the solution was confirmed to be neutral. The solution was centrifuged and the supernatant liquid was pippetted off. The solid was washed with water and then dried under vacuum. The solid was washed with 1 :1 EtOAc/hexane (2 x) and then dried under vacuum to afford 3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-N-(5 -fluoro-6-methoxypyridin-3 - yl)imidazo[l,2-a]pyridin-2-amine as a white solid (0.031 g, 86% yield). MS (ESI positive ion) m/z: 367 (M+l). 1H NMR (400 MHz, DMSO-J6) δ ppm 2.42 (s, 3 H), 3.94 (s, 3 H), 7.13 (t, J = 6.7 Hz, 1 H), 7.44 (br s, 1 H), 7.52 (t, J= 7.6 Hz, 1 H), 7.60-7.69 (m, 2 H), 8.50 (dd, J = 12.8, 1.9 Hz, 1 H), 8.56 (br s, 1 H), 9.97 (d, J= 6.8 Hz, 1 H), 10.46 (br s, 1 H).
Example 26
Synthesis of 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-fluoro-N-(6-methoxypyridin-3- yl)imidazo[l ,2-a]pyridin-2-amine
Figure imgf000103_0001
Stepl
To a 100-mL round-bottomed flask was added 2-amino-5-fluoropyridine (1.08 g, 9.62 mmol) (Sigma- Aldrich, St. Louis, MO) and ethyl bromoacetate (3.73 mL, 33.7 mmol) (Sigma- Aldrich, St. Louis, MO). The reaction mixture was stirred at room temperature for 16 h. To the thick suspension was added ether (20 mL) and the resulting white precipitate was filtered off, washed with ether, and dried in vacuum. Ethyl 2-(5-fluoro-2-iminopyridin-l(2H)- yl)acetate hydrobromide (1.9 g, 6.81 mmol) was obtained as a white solid. MS (ESI positive ion) m/z: 199.1 (M+ 1). Step 2
To a 25-mL, round-bottomed flask was added ethyl 2-(5-fluoro-2-iminopyridin-l(2H)- yl)acetate hydrobromide (1.92 g, 6.88 mmol) and phosphorus oxychloride (6.30 mL, 68.8 mmol). The reaction mixture was heated at 110 0C for 3 h, allowed to cool to room temperature, and the solvent was eliminated under vacuum. To the greenish syrup obtained was added ice water and NH4OH carefully until the pH of the solution was made basic. The solution was extracted with DCM and the combined organic extracts were dried over Na2SO4. The solution was filtered and concentrated in vacuum to give the crude material that was absorbed onto a plug of silica gel and purified by chromatography through a Redi-Sep prepacked silica gel column (40 g), eluting with a gradient of 30 to 40 % EtOAc in hexane. 2- Chloro-6-fluoroimidazo[l,2-a]pyridine was obtained as a white solid (0.85 g, 4.99 mmol). Step 3 A glass microwave reaction vessel was charged with 2-chloro-6-fluoroimidazo[ 1 ,2- ajpyridine (0.85 g, 4.99 mmol), triphenylphosphine (0.26 g,, 1.0 mmol), palladium diacetate (112 mg, 0.50 mmol), potassium carbonate (0.14 g, 9.99 mmol) and 4-chloro-N,N-bis(4- methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine (2691 mg, 6.99 mmol) in dioxane (15 mL). The vial was sealed and purged with argon for several minutes. The reaction mixture was stirred and heated in an Emrys Optmizer microwave reactor (Personal Chemistry, Biotage AB, Inc., Upssala, Sweden) at 140 0C for 3 h. The reaction mixture was diluted with satd. NaHCO3 (10 mL) and extracted with DCM. The combined organic extracts were dried over Na2SO4. The solution was filtered and concentrated in vacuum to give the crude material. The crude material was absorbed onto a plug of silica gel and purified by chromatography through a Redi-Sep pre-packed silica gel column (40 g), eluting with a gradient of 20 % to 50 % EtOAc in hexane, to provide 4-(2-chloro-6-fluoroimidazo[l,2-a]pyridin-3-yl)-N,N-bis(4- methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine (0.66g, 1.28 mmol) as white solid. MS (ESI positive ion) m/z: 518.8 (M+). Step 4 To a 10-mL vial was added 4-(2-chloro-6-fluoroimidazo[l,2-a]pyridin-3-yl)-N,N-bis(4- methoxybenzyl)-6-methyl-l,3,5-triazin-2-amine (139 mg, 0.27 mmol), 6-methoxypyridin-3- amine (40 mg, 0.32 mmol) (Aldrich, St Louis, MO), sodium 2-methylpropan-2-olate (64 mg, 0.67 mmol), chloro[2-(dicyclohexylphosphino)-3,6-dimethoxy-2'-4'-6'-tri-i-propyl-l,r- biphenyl][2-(2-aminoethyl)phenyl]palladium(II) (Strem, Newburyport, MA) (2 mg, 2.7 μmol), and dioxane (1.5 niL). The vial was sealed and purged with argon for several minutes and stirred at 90 0C for 6 h. The reaction mixture was diluted with satd NaHCO3 (5 mL) and extracted with EtOAc. The combined organic extracts were dried over Na2SO4, and the solution was filtered and concentrated in vacuo to give the crude material. The crude material was absorbed onto a plug of silica gel and purified by chromatography through a Redi-Sep prepacked silica gel column (12 g), eluting with a gradient of 20 % to 40 % EtOAc in hexane, to provide 3-(4-(bis(4-methoxybenzyl)amino)-6-methyl-l,3,5-triazin-2-yl)-6-fluoro-N-(6- methoxypyridin-3-yl)imidazo[l,2-a]pyridin-2-amine (47 mg, 0.08 mmol) as yellow solid. MS (ESI positive ion) m/z: 606.8 (M+). Step 5
To a 25 -mL round-bottomed flask was added 3-(4-(bis(4-methoxybenzyl)amino)-6-methyl- 1 ,3,5-triazin-2-yl)-6-fluoro-N-(6-methoxypyridin-3-yl)imidazo[l ,2-a]pyridin-2-amine (47 mg, 0.08 mmol) and 2% triflic acid in TFA (1 mL). The reaction mixture was stirred at room temperature for 5 h, and the solvents were eliminated under vacuum. The residue was dissolved in DCM and washed with satd NaHCO3 solutionand dried over Na2SO4. The solution was filtered and concentrated in vacuo to give the crude material, which was triturated with ether and filtered off. 3-(4-Amino-6-methyl-l,3,5-triazin-2-yl)-6-fluoro-N-(6-methoxypyridin- 3-yl)imidazo[l,2-a]pyridin-2-amine was obtained as a yellow solid (12 mg, 0.03 mmol). MS (ESI positive ion) m/z: 367.0 (M+ 1). IH NMR (400 MHz, DMSO-J6) δ ppm 2.43 (s, 3 H) 3.84 (s, 3 H) 6.86 (d, J=8.41 Hz, 1 H) 7.49 (s, 1 H) 7.53 - 7.67 (m, 2 H) 7.74 (s, 1 H) 8.32 (d, J = 7.82 Hz, 1 H) 8.68 (br. s., 1 H) 10.04 (br. s., 1 H) 10.24 (br. s., 1 H).
Example 27
Synthesis of 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-7-methoxy-N-(lH-pyrazol-3- yl)imidazo[l ,2-a]pyridin-2-amine
Figure imgf000105_0001
Step 1
Dissolved 2-chloroacetic acid (1.269 g, 13.43 mmol) in acetonitrile (2.238 niL) and then added triethylamine (2.81 mL, 20.14 mmol) at rt. 4-methoxypyridin-2-amine (2.0 g, 16.11 mmol) was added and the reation mixture was and heated to 90 0C for 2 hrs until reaction was complete. After concentration, the crude product 2-(2-imino-4-methoxypyridin- l(2H)-yl)acetic acid (2.40 g, 13.2 mmol, 98% yield) was used in the next step, m/z (ESI, +ve ion) 183.2 (M+H)+. Step 2
In a 50 mL round bottom flask, dissolved 2-(2-imino-4-methoxypyridin-l(2H)-yl)acetic acid (2.44 g, 13.39 mmol) in toluene (13.39 mL) and added phosphoryl trichloride (3.68 mL, 40.2 mmol) and the reaction mixture was stirred and heated to 110 0C for 16 hrs, until the reaction was complete based on LCMS. The reaction mixture was transferred dropwise to a flask with water and ice then stirred for 30 min. The phases were seperated, organic layer discarded, and the aqueous phase was neutralized with 6 N NaOH while in an ice-water bath. The aqueous layer was extracted with DCM, and the organic layer was dried over Na2SO4, filtered then concentrated under rotary evaporation. Purification by MPLC (ISCO) with a gradient DCM:MeOH + NH4OH 100:0 to 90:10 afforded 2-chloro-7-methoxyimidazo[l,2- ajpyridine (250 mg, 1.369 mmol, 10.22 % yield) as a tan solid, m/z (ESI, +ve ion) 183.0 (M+H)+.
Step 3
Zinc(II) chloride (4.50 g, 33.0 mmol) was flammed dried under vacuum and then dissolved in THF (30.0 mL) to make a 1 M solution. In a separate dried 250 mL round bottom flask under nitrogen, dissolved 2,2,6, 6-tetramethylpiperidine (4.24 g, 30 mmol) in dry THF (30.0 mL), and then cooled it to -40 0C. n-Bbutyllithium (12.00 mL, 30.0 mmol) was added dropwise, and allowed the reaction mixture to warm to -10 0C over 1 hr. Zinc(II) chloride (4.50 g, 33.0 mmol) solution was added dropwise, and stirring was continued for 30 min at -10 0C, then 30 min at rt. This solution in THF (0.42 M) was used for coupling reactions and was stored under nitrogen. Dissolved 2-chloro-7-methoxyimidazo[ 1 ,2-a]pyridine (210 mg, 1.150 mmol) in THF
(958 μL) and added the solution of (2,2,6,6-tetramethylpiperidin-l-yl)zinc(II) lithium chloride (4107 μL, 1.725 mmol) (0.42 M) dropwise and let stir for 30 min at rt under nitrogen. In a separate flask, dissolved palladium tetrakis (133 mg, 0.115 mmol) and 2,4-dichloro-6-methyl- 1,3,5-triazine (283 mg, 1.725 mmol) in more THF (958 μL) under nitrogen and cannulated the mixture into the -chloro-7-methoxyimidazo[l,2-a]pyridine solution. The reaction mixture was stirred for 16 hrs at rt until reaction was complete by LCMS. The reaction mixture was quenched reaction with NH4Cl and aqueous layer was extracted with EtOAc, dried organics over Na2SO4, and concentrated under rotary evaporation. The crude product was purified via MPLC (ISCO) with a gradient hexanes:EtOAc column 0: 100 to 100:0. to give 2-chloro-3-(4- chloro-6-methyl-l,3,5-triazin-2-yl)-7-methoxyimidazo[l,2-a]pyridine (150 mg, 0.484 mmol, 42.1 % yield) as a yellow solid, m/z (ESI, +ve ion) 310.0 (M+H)+. Step 4
Dissolved 2-chloro-3-(4-chloro-6-methyl-l,3,5-triazin-2-yl)-7-methoxyimidazo[l,2- ajpyridine (150 mg, 0.484 mmol) in 0.5 M ammonia in dioxane (2902 μL, 1.451 mmol) in a sealed tube and the reaction mixture was stirred for 16 hrs at rt. Concentration of the reaction mixture gave crude 4-(2-chloro-7-methoxyimidazo-[l,2-a]pyridin-3-yl)-6-methyl-l,3,5-triazin- 2-amine that was used in the next step, m/z (ESI, +ve ion) 291.0 (M+H)+. Step 5 Charged a flask with Pd2dba3 (132 mg, 0.144 mmol) and CyP-Ft-Bu2 (208 mg, 0.289 mmol) and degassed, backfilling with nitrogen three times. The solids were dissolved in t- BuOH (2094 μL) and water (130 μL, 7.22 mmol) and heated to 100 0C for 15 min. The reaction mixture was cooled to rt and lH-pyrazol-3 -amine (120 mg, 1.445 mmol), 4-(2-chloro- 7-methoxyimidazo[l,2-a]pyridin-3-yl)-6-methyl-l,3,5-triazin-2-amine (210 mg, 0.722 mmol) and cesium carbonate (706 mg, 2.167 mmol) were added. The reaction mixture was degassed again with nitrogen and heated at 100 0C for 4 hrs until the reaction was complete by LCMS. The reaction mixture was then rotary evaporated onto silica, and purified with MPLC (ISCO) with a gradient DCM:MeOH + NH4OH 100:0 to 90:10 to afford 3-(4-amino-6-methyl-l,3,5- triazin-2-yl)-7-methoxy-N-(lH-pyrazol-3-yl)imidazo[l,2-a]pyridin-2-amine (30.0 mg, 88.9 μmol, 12.3% yield), m/z (ESI, +ve ion) 338.0 (M+H)+. 1H NMR (400 MHz, DMSO-J6) δ ppm 2.33 (s, 3 H) 3.89 (s, 3 H) 6.72 (br. s., 1 H) 6.80 (s, 1 H) 7.02 (br. s., 1 H) 7.32 (br. s., 2 H) 7.61 (s, 1 H) 9.78 (d, J=7.53 Hz, 1 H) 10.41 (br. s., 1 H) 12.14 (br. s., 1 H).
Example 28 Synthesis of N-(l-(lH-benzo[d]imidazol-2-yl)-lH-pyrazol-3-yl)-3-(6-amino-2- methylpyrimidin-4-yl)imidazo[ 1 ,2-b]pyridazin-2-amine
Figure imgf000108_0001
3 -(6- Amino-2-methylpyrimidin-4-yl)-N-( 1 H-pyrazol-3 -yl)imidazo [ 1 ,2-b]pyridazin-2- amine (100 mg, 0.325 mmol), p-toluenesulfonic acid (61.9 mg, 0.325 mmol), and 2-chloro-lH- benzo[d]imidazole (59.6 mg, 0.390 mmol) were dissolved in 2-butanol (651 μL) and heated in the microwave at 100 0C for 30 min until reaction was complete by LCMS. After concentration, the reaction mixture was redissolved in DMSO and purified via HPLC (Gilson) with a gradient water :acetonitrile 85:15 to 30:70 with 0.1% TFA. N-(l-(lH-benzo- [d]imidazol-2-yl)-l H-pyrazol-3 -yl)-3-(6-amino-2-methylpyrimidin-4-yl)imidazo[ 1,2- b]pyridazin-2-amine 2,2,2-trifluoroacetate was collected directly as the TFA salt as a yellow solid, m/z (ESI, +ve ion) 424.5 (M+H)+. 1H NMR (500 MHz, DMSO-J6) δ ppm 2.66 (s, 3 H) 7.07 - 7.22 (m, 2 H) 7.32 (d, J=2.67 Hz, 1 H) 7.44 - 7.59 (m, 3 H) 7.93 (br. s., 1 H) 8.18 (dd, J=8.98, 1.50 Hz, 1 H) 8.56 (d, J=2.67 Hz, 1 H) 8.71 (dd, J=4.65, 1.44 Hz, 1 H) 10.87 (br. s., 1 H). Example 29
Synthesis of 3-(3-(6-amino-2-methylpyrimidin-4-yl)imidazo[ 1 ,2-b]pyridazin-2-ylamino)-N- phenyl- 1 H-pyrazole- 1 -carboxamide
Figure imgf000108_0002
3 -(6- Amino-2-methylpyrimidin-4-yl)-N-( 1 H-pyrazol-3 -yl)imidazo [ 1 ,2-b]pyridazin-2- amine (120 mg, 0.390 mmol) was dissolved in THF (781 μL) and isocyanatobenzene (63.8 μL, 0.586 mmol) was added. The reaction mixture was stirred at rt for 2 hrs until it was complete by LCMS (two isomers on the pyrazole). The reaction mixture was concentrated, then redissolved mixture in DMSO and purified via HPLC (Gilson) with a gradient water :acetonitrile 85:15 to 10:90 with 0.1% TFA to afford 3-(3-(6-amino-2-methylpyrimidin- 4-yl)imidazo[l,2-b]pyridazin-2-ylamino)-N-phenyl-lH-pyrazole-l-carboxamide (25 mg, 0.046 mmol, 12 % yield) as the TFA salt, m/z (ESI, +ve ion) 427.5 (M+H)+. 1H NMR (500 MHz, DMSO-J6) δ ppm 2.63 (s, 3 H) 7.16 (t, J=7.32 Hz, 1 H) 7.26 (d, J=I.91 Hz, 1 H) 7.29 (d, J=2.35 Hz, 1 H) 7.39 (t, J=7.85 Hz, 2 H) 7.44 (d, J=7.27 Hz, 1 H) 7.48 (dd, J=8.82, 4.65 Hz, 1 H) 7.74 (d, J=8.12 Hz, 2 H) 7.87 (br. s., 1 H) 8.09 - 8.23 (m, 1 H) 8.39 (d, J=2.67 Hz, 1 H) 8.63 - 8.77 (m, 1 H) 10.10 (s, 1 H) 10.99 (br. s., 1 H).
Example 30 Synthesis of 3-(3-(6-amino-2-methylpyrimidin-4-yl)imidazo[l,2-b]pyridazin-2-ylamino)-N-(2- methoxy ethyl)- 1 H-pyrazole- 1 -carboxamide
Figure imgf000109_0001
Stepl
In a 50-mL sealed tube under N2 were dissolved 3-(6-amino-2-methylpyrimidin-4-yl)- N-(I H-pyrazol-3 -yl)imidazo[l,2-b]pyridazin-2-amine (1.20 g, 3.90 mmol), and phenyl carbonochloridate (0.539 mL, 4.30 mmol) in THF (24 mL), then added hunig'sbase (0.682 mL, 3.90 mmol). Let stir at rt for 1 hr until reaction was complete by LCMS (two isomers on the pyrazole). The crude solution of phenyl 3-(3-(6-amino-2-methylpyrimidin-4-yl)imidazo[l,2- b]pyridazin-2-ylamino)-l H-pyrazole- 1-carboxylate (1.67 g, 3.9 mmol, 100% yield) was used in the next step, m/z (ESI, +ve ion) 428.4 (M+H)+. Step 2
3 -(3 -(6-Amino-2-methylpyrimidin-4-yl)imidazo [ 1 ,2-b]pyridazin-2-ylamino)- 1 H- pyrazole-1-carboxylate (133 mg, 0.311 mmol) was dissolved in THF (2 niL) (what is the second reagent??) and the reaction mixture wasstirred at rt for 1 hr until reaction was complete by LCMS (2 isomers formed). The reaction mixture was concentrated, then redissolved in DMSO and purified via HPLC (Gilson) with a gradient water :acetonitrile 90:10 to 50:50 with 0.1% TFA to afford 3-(3-(6-amino-2-methylpyrimidin-4-yl)imidazo[l,2-b]pyridazin-2- ylamino)-N-(2-methoxyethyl)-lH-pyrazole-l-carboxamide (1.05 mg, 2.010 μmol, 1 % yield ) as the TFA salt, m/z (ESI, +ve ion) 409.5 (M+H)+. 1H NMR (400 MHz, METHANOL-J4) δ ppm 2.75 (s, 3 H) 3.42 (s, 3 H) 3.56 - 3.64 (m, 4 H) 7.08 (d, J=2.84 Hz, 1 H) 7.49 (dd, J=9.00, 4.69 Hz, 1 H) 8.05 (dd, J=9.05, 1.61 Hz, 1 H) 8.18 (br. s., 1 H) 8.20 (d, J=2.84 Hz, 1 H) 8.63 (dd, J=4.60, 1.57 Hz, I H).
Example 31
Synthesis of 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(2-fluorophenyl)-N-(lH-pyrazol-3- yl)imidazo[l ,2-a]pyridin-2-amine 2,2,2-trifluoroacetate
Figure imgf000110_0001
Step l To a solution of 2-chloroacetic acid (1.661 mL, 24.08 mmol) and acetonitrile (4.01 mL) was added triethylamine 99% (5.02 mL, 36.1 mmol), dropwise under N2. 2-Amino-5- bromopyridine (5.00 g, 28.9 mmol) was added as a solid in a single portion. The reaction mixture was maintained at rt for 3 min until majority of chloropyridine had dissolved and then heated at 90 0C for 60 min. The reation appeared to undergo an uncontrolled exotherm after 50 minutes at 90 0C and turned dark brown. Oil bath was removed and reaction was cooled to rt, yellow/tan solid was collected by vacuum filtration, rinsing with minimal acetonitrile. The solid was dried under reduced pressure to yield 2-(5-bromo-2-iminopyridin-l(2H)-yl)acetic acid (2.657 g, 11.50 mmol, 47.8 % yield) as a light brown solid. LCMS (trifluoroacetic acid modifier, ESI) m/z: 231.0 (M+H)+ Step 2
6-Bromo-2-chloroimidazo[l,2-a]pyridine was prepared according to preparation of 2- chloroimidazo[l,2-b]pyridazine in Example 18, Step 2. LCMS (trifluoroacetic acid modifier, ESI) m/z: 231.0 (M+H)+ Step 3
Added anhydrous THF (35.1 mL) to 6-bromo-2-chloroimidazo[l,2-a]pyridine (8.13 g, 35.1 mmol) under N2 atmosphere, followed by (2,2,6,6-tetramethylpiperidin-l-yl)zinc(II) lithium chloride solution in THF (122 mL, 63.2 mmol) at rt. The reaction mixture was maintained for 30 min at rt. In a separate flask combined 4,6-dichloro-2-methylpyrimidine (17.18 g, 105 mmol) and palladium tetrakis (4.06 g, 3.51 mmol) in THF (20 mL) and the slurry was to the solution containing of 6-bromo-2-chloroimidazo[l,2-a]pyridine and (2,2,6,6- tetramethylpiperidin-l-yl)zinc(II) lithium chloride at rt. After 18 h, the solution was diluted with MeOH and concentrated to generate a brown sludge. This was filtered though a buchner funnel rinsing with CH2Cl2 to afford pure desired product 6-bromo-2-chloro-3-(6-chloro-2- methylpyrimidin-4-yl)imidazo[l,2-a]pyridine (6.87 g, 19.19 mmol, 54.6 % yield) as an orange solid. LCMS (trifluoroacetic acid modifier, ESI) m/z: 359.1 (M+H)+ Step 4 To 6-bromo-2-chloro-3-(6-chloro-2-methylpyrimidin-4-yl)imidazo[l ,2-a]pyridine
(2.105 g, 5.88 mmol) was added ammonia in MeOH (7N) (0.127 mL, 5.88 mmol) in a sealed vessel. Ammonia (gas) (0.127 mL, 5.88 mmol) was bubbled through solution for 1 minute and heated at 100 0C for 2 h with microwave. Solid was collected by vacuum filtration, rinsing with IPA and water, then IPA to give 6-(6-bromo-2-chloroimidazo[l,2-a]pyridin-3-yl)-2- methylpyrimidin-4-amine (1.705 g, 5.04 mmol, 86 % yield) as a light yellow amorphous solid. LCMS (trifluoroacetic acid modifier, ESI) m/z: 340.0 (M+H)+ Step 5
1,4-Dioxane (2.58 mL) was added to 4-(6-bromo-2-chloroimidazo[l,2-a]pyridin-3-yl)- 6-methyl-l,3,5-triazin-2-amine (0.175 g, 0.515 mmol), [l,l-bis(diphenylphosphino)- ferrocene]palladium(II) dichloride dichloromethane adduct (0.042 g, 0.052 mmol), and 2- fluorobenzeneboronic acid (0.079 g, 0.567 mmol) under N2. A solution of sodium carbonate anhydrous granular (0.086 mL, 2.061 mmol) was added as a 2M solution in water (2 mL). The reaction mixture was stirred and heated at 80 0C for 2 hr. The crude material was purified by chromatography through a Redi-Sep pre-packed silica gel column (40 g), eluting with a gradient of 0% to 10% MeOH in CH2CL2, to provide 4-(2-chloro-6-(2- fluorophenyl)imidazo[l,2-a]pyridin-3-yl)-6-methyl-l,3,5-triazin-2-amine (0.193 g, 0.544 mmol, 106 % yield) as a yellow solid. LCMS (trifluoroacetic acid modifier, ESI) m/z: 355.0 (M+H)+ Step 6
3-(4-Amino-6-methyl-l,3,5-triazin-2-yl)-6-(2-fluorophenyl)-N-(lH-pyrazol-3- yl)imidazo[l,2-a]pyridin-2-amine 2,2,2-trifluoroacetate was prepared according to Example 27, step 5. The crude material was purified by reverse-phase preparative HPLC using 0.1% TFA in CH3CN/H2O, gradient 10% to 90% over 20 min to provide 3-(4-amino-6-methyl- l,3,5-triazin-2-yl)-6-(2-fluorophenyl)-N-(lH-pyrazol-3-yl)imidazo[l,2-a]pyridin-2-amine
2,2,2-trifluoroacetate (0.028 g, 0.054 mmol, 10.71 % yield) as an orange solid. LCMS (formic acid modifier, ESI) m/z: 402.0 (M+H)+ 1U NMR (400 MHz, DMSO-J6) δ ppm 2.41 (s, 3 H); 6.79 (d, J=2.05 Hz, 1 H); 7.29 - 7.46 (m, 2 H); 7.46 - 7.62 (m, 2 H); 7.62 - 7.80 (m, 5 H); 7.83 (br. s., 1 H); 10.19 (br. s., 1 H); 10.43 (br. s., 1 H).
Example 32
Synthesis of 3-(6-amino-2-methylpyrimidin-4-yl)-N-(lH-pyrazol-3-yl)imidazo[l ,2-a]pyridin-
2-amine 2,2,2-trifluoroacetate
Figure imgf000112_0001
Step 1
2-Chloro-3-(6-chloro-2-methylpyrimidin-4-yl)imidazo[ 1 ,2-a]pyridine was synthesized according to the preparation of 2-chloro-3-(6-chloro-2-methylpyrimidin-4-yl)imidazo[l,2- b]pyridazine in Example 18 step 3. LCMS (trifluoroacetic acid modifier, ESI) m/z: 279.0 (M+H)+ Step 2
6-(2-Chloroimidazo[l ,2-a]pyridin-3-yl)-2-methylpyrimidin-4-amine was prepared according to the preparation of 6-(6-bromo-2-chloroimidazo[l,2-a]pyridin-3-yl)-2- methylpyrimidin-4-amine in Example A, step 4. LCMS (trifluoroacetic acid modifier, ESI) m/z: 260.2 (M+H)+ Step 3
3 -(6- Amino-2-methylpyrimidin-4-yl)-N-( 1 H-pyrazol-3 -yl)imidazo [ 1 ,2-a]pyridin-2- amine 2,2,2-trifluoroacetate was prepared according to procedure described in Example 27, Step 5. The crude material was purified by reverse-phase preparative HPLC using 0.1% TFA in CH3CN/H2O, gradient 10% to 90% over 20 min to provide 3-(6-amino-2-methyl-pyrimidin- 4-yl)-N-(l H-pyrazol-3 -yl)imidazo[l,2-a]pyridin-2-amine 2,2,2-trifluoroacetate (0.031 g, 0.074 mmol, 12.77 % yield) as yellow solid. LCMS (trifluoroacetic acid modifier, ESI) m/z: 307.5 (M+H)+. 1H NMR (400 MHz, MeOH) δ ppm 2.68 (s, 3 H); 6.62 (br. s., 1 H); 6.89 (s, 1 H); 7.13 - 7.30 (m, 1 H); 7.66 (br. s., 3 H); 8.85 (m, 1 H).
Example 33
Synthesis of 2-(3-methoxyphenyl)-N-(2-methyl-6-(2-(l -methyl- lH-pyrazol-3- ylamino)imidazo[l,2-a]pyridin-3-yl)pyrimidin-4-yl)acetamide 2,2,2-trifluoroacetate
Figure imgf000113_0001
Step l
3 -(6- Amino-2-methylpyrimidin-4-yl)-N-( 1 -methyl- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2- a]pyridin-2-amine was prepared according to the procedure described in Example 18, Step 5 using l-methyl-lh-pyrazol-3-amine. LCMS (trifluoroacetic acid modifier, ESI) m/z: 321.2 (M+H)+
Step 2
3 -(6- Amino-2-methylpyrimidin-4-yl)-N-( 1 -methyl- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2- a]pyridin-2-amine (0.085 g, 0.265 mmol) and 2-(3-methoxyphenyl)acetic acid (0.057 g, 0.345 mmol) were put under N2. Ethyl acetate (1.327 mL), followed by diisopropylethylamine (0.231 mL, 1.327 mmol) and finally 1-propanephosphonic acid cyclic anhydride, 50 wt. % solution in ethyl acetate (1.351 mL, 2.123 mmol) were added. The reaction mixture was stirred and heated to 55 0C for 2 h. The concentrated crude material was purified by chromatography through a Redi-Sep pre-packed silica gel column (40 g), eluting with a gradient of 1% to 10% MeOH in CH2Cl2, to provide unpure product. Purification by reverse- phase preparative HPLC, 0.1% TFA in CH3CN/H2O, gradient 10% to 90% over 15 min providee 2-(3 -methoxyphenyl)-N-(2-methyl-6-(2-( 1 -methyl- 1 H-pyrazol-3 -ylamino)- imidazo[l,2-a]pyridin-3-yl)pyrimidin-4-yl)acetamide 2,2,2-trifluoroacetate (0.047 g, 0.081 mmol, 30.4 % yield) as a yellow solid.
LCMS (trifiuoroacetic acid modifier, ESI) m/z: 469.2 (M+H)+
1H NMR (400 MHz, DMSO-J6) δ ppm 2.62 (s, 3 H); 3.76 (m, J=4.60 Hz, 8 H); 6.67 (d, J=2.25 Hz, 1 H); 6.79 - 6.87 (m, 1 H); 6.88 - 6.97 (m, 2 H); 7.16 - 7.29 (m, 2 H); 7.49 (t,
J=7.73 Hz, 1 H); 7.54 - 7.64 (m, 2 H); 8.30 (s, 1 H); 8.78 (d, J=6.65 Hz, 1 H); 10.40 (br. s., 1 H); 11.10 (s, 1 H).
Example 34 Synthesis of 3 -(6-amino-2-methylpyrimidin-4-yl)-N-( 1 -(2-(dimethylamino)ethyl)- 1 H- pyrazol-3-yl)imidazo[ 1 ,2-b]pyridazin-2-amine 2,2,2-trifluoroacetate
Figure imgf000114_0001
The title compound was prepared according to procedure described in of Example 27, Step 5 using l-(2-(dimethylamino)ethyl)-lH-pyrazol-3-amine.
Example 35
Synthesis of 3 -(6-amino-2-methylpyrimidin-4-yl)-N-( 1 H-pyrazol-3 -yl)-6-(pyrimidin-5 - yl)imidazo[ 1 ,2-b]pyridazin-2-amine
Figure imgf000115_0001
Step l
A sealable vial was charged with 2,6-dichloroimidazo[l,2-b]pyridazine (1.17 g, 6.22 mmol), pyrimidine-5-boronic acid (1.388 g, 11.20 mmol, Maybridge), and dichloro(l,l- bis(diphenylphosphinoferrocene))palladium(ii) dichloromethane adduct (0.356 g, 0.436 mmol, Strem). The vial was sealed with a septa cap and flushed with N2. Dioxane (24.89 mL) was added followed by an aqueous solution of sodium carbonate (6.55 mL, 12.45 mmol). The reaction mixture was sparged with N2 for 10 min, and then heated at 80 0C for 3 h. The reaction mixture was concentrated and purified by MPLC (Teledine Isco combiFlash
Companion) as described: The crude residue was taken up in minimal CH2CVMeOH (20 mL) and absorbed onto a minimal amount of loose silica gel, which was then packed into a 25 g custom loading cartridge and passed through a Redi-Sep® pre-packed silica gel column (80 g) using a gradient of 1% MeOH in CH2Cl2 to 10% MeOH in CH2Cl2 to afford 2-chloro-6- (pyrimidin-5-yl)imidazo[l,2-b]pyridazine (1.32 g, 5.70 mmol, 92 % yield) as a orange amorphous solid. Step 2
A microwave vial was charged with 2-chloro-6-(pyrimidin-5-yl)imidazo[l,2- b]pyridazine (0.366 g, 1.580 mmol), 4,6-dichloro-2-methylpyrimidine (0.567 g, 3.48 mmol, Aldrich), cesium carbonate (1.030 g, 3.16 mmol, Strem), triphenylphosphine (0.083 g, 0.316 mmol, Fluka) and palladium(ii) acetate (0.035 g, 0.158 mmol, Strem). The vial was sealed with septa cap, flushed with N2, and dioxane was added via syringe (6.32 mL). The reaction mixture was sparged with N2 for 10 minutes, then irradiated in the microwave (Biotage Initiator) at 130 0C for 2 h. The crude reaction mixture was absorbed directly onto a 25 g pre- packed silica loading cartridge for purification by MPLC (Teledine Isco combiFlash
Companion). The residue was passed through a Redi-Sep® pre-packed silica gel column (40 g) using first a gradient of 5% EtOAc in CH2Cl2 to 100% EtOAc followed by a gradient of 1% MeOH in CH2Cl2 to 10% MeOH in CH2Cl2 to afford 2-chloro-3-(6-chloro-2-methylpyrimidin- 4-yl)-6-(pyrimidin-5-yl)imidazo[l,2-b]pyridazine (0.380 g, 1.061 mmol, 67.1 % yield) as a tan solid.
Step 3
A sealable microwave vial was charged with 2-chloro-3-(6-chloro-2-methylpyrimidin- 4-yl)-6-(pyrimidin-5-yl)imidazo[l,2-b]pyridazine (0.380 g, 1.061 mmol) and the vial was sealed using a septa cap. IO N ammonia in MeOH (17.10 mL, 790 mmol) was added via syringe to generate a tan slurry. Gaseous ammonia was bubbled through the slurry for 5 min, then the mixture was irradiated in the microwave (Biotage Initiator) at 100 0C for 2 h. Upon cooling to rt, a tan solid precipitated. The solid was collected by vacuum filtration to give 6- (2-chloro-6-(pyrimidin-5-yl)imidazo[ 1 ,2-b]pyridazin-3-yl)-2-methylpyrimidin-4-amine (0.2394 g, 0.707 mmol, 44.7 % yield) as a tan solid. Step 4
A sealable vial was charged with tris(dibenzylideneacetone)dipalladium(0) (32.4 mg, 0.035 mmol, Strem) and (r)-l-[(s)-2-(dicyclohexylphosphino)ferrocenyl]ethyl-di-tert- butylphosphine (39.3 mg, 0.071 mmol, Strem). The vial was sealed with a septa cap and flushed with N2. tert-Butanol (1 mL) was added, followed by water (63.8 μL, 3.54 mmol) and the resulting mixture was sparged with N2 for 5 min. The solution was heated at 120 0C for 10 min, and then cooled to rt.
In a separate vial was charged 6-(2-chloro-6-(pyrimidin-5-yl)imidazo[l,2-b]pyridazin- 3-yl)-2-methylpyrimidin-4-amine (120.0 mg, 0.354 mmol), cesium carbonate (404 mg, 1.240 mmol, Alfa Aesar), and 3-aminopyrazole (147 μL, 1.771 mmol, Alfa Aesar). The vial was sealed with a septa cap and flushed with N2. tert-Butanol (1771 μL) was added, was added and the reaction mixture was sparged with N2 for 5 mins. The catalyst solution was transferred via syringe to the vial that contained the coupling substrates. The resulting mixture was heated at 100 0C for 18 h. The crude reaction mixture was absorbed directly onto a 25 g loading cartridge for purification by MPLC (Teledine Isco combiFlash Companion). The residue was passed through a Redi-Sep® pre-packed silica gel column (40 g) using a gradient of 0.1% NH4OH and 1% MeOH in CH2Cl2 to 1% NH4OH and 10% MeOH in CH2Cl2 to afford the title compound 3 -(6-amino-2-methylpyrimidin-4-yl)-N-( 1 H-pyrazol-3 -yl)-6-(pyrimidin-5 - yl)imidazo[l,2-b]pyridazin-2-amine (100.0 mg, 0.259 mmol, 73.3 % yield) as a orange amorphous solid. IH NMR (500 MHz, DMSO-J6) δ ppm 2.46 (s, 3 H) 6.82 - 6.88 (m, 1 H) 7.12 (br. s, 2 H) 7.62 - 7.65 (m, 1 H) 7.70 (s, 1 H) 7.98 - 8.06 (m, 1 H) 8.12- 8.19 (m, 1 H) 9.36 (s, 1 H) 9.59 (s, 2 H) 11.28 (s, 1 H) 12.17 (s, 1 H); LRMS m/z calcd = 385.2, observed (M +H) = 386.4. Example 35
Synthesis ofN-(6-(2-(lH-pyrazol-3-ylamino)-6-(pyrimidin-5-yl)imidazo[l,2-a]pyridin-3-yl)-2- methylpyrimidin-4-yl)acetamide
Figure imgf000117_0001
Step l
To a vial charged with 3-(6-amino-2-methylpyrimidin-4-yl)-N-(lH-pyrazol-3-yl)-6- (pyrimidin-5-yl)imidazo[l,2-a]pyridin-2-amine (prepared as described in Example 31) was added 1,4-dioxane (2445 μL), followed by pyridine (316 μL, 3.91 mmol, Aldrich) then acetic anhydride (231 μL, 2.445 mmol, Aldrich). The vial was sealed with a septa cap and heated at 80 0C for 2 h. Additional acetic anhydride (231 μL, 2.445 mmol) was added and the solution was heated at 80 0C for an additional 18 h. The solution was concentrated for purification by MPLC (Teledine Isco combiFlash Companion). The crude residue was taken up in minimal CH2Cl2 and absorbed onto a 5 g pre-packed silica loading cartridge and passed through a Redi- Sep® pre-packed silica gel column (40 g) using a gradient of 1% MeOH in CH2Cl2 to 10% MeOH in CH2Cl2 to afford N-(6-(2-(l -acetyl- lH-pyrazol-3 -ylamino)-6-(pyrimidin-5 - yl)imidazo[l,2-a]pyridin-3-yl)-2-methylpyrimidin-4-yl)acetamide (54.0 mg, 0.115 mmol, 23.57 % yield) as a yellow solid. Step 2
A sealable vial was charged with N-(6-(2-( 1 -acetyl- lH-pyrazol-3 -ylamino)-6- (pyrimidin-5-yl)imidazo[ 1 ,2-a]pyridin-3-yl)-2-methylpyrimidin-4-yl)acetamide (54.0 mg, 0.115 mmol) followed by CH2Cl2 (5 mL) and MeOH (0.5 mL) and MP-Carbonate (159 mg, 3.07 mmol/gram loading, Biotage). The reaction mixture was heated at 60 0C for 1 hour. The solution was filtered to remove the MP-Carbonate and the filtrate was concentrated for purification by MPLC (Teledine Isco combiFlash Companion). The crude residue was taken up in minimal 90/10/1 CH2Cl2/MeOH/NH4OH and absorbed onto a 5 g loading cartridge and passed through a Redi-Sep® pre-packed silica gel column (12 g) using a gradient of 0.1% NH4OH and 1% MeOH in CH2Cl2 to 1% NH4OH and 10% MeOH in CH2Cl2 to afford N-(6- (2-( 1 H-pyrazol-3 -ylamino)-6-(pyrimidin-5 -yl)imidazo [ 1 ,2-a]pyridin-3 -yl)-2-methyl-pyrimidin- 4-yl)acetamide (15.0 mg, 0.035 mmol, 7.19 % yield) as a yellow amorphous solid. IH NMR (400 MHz, DMSO-J6) δ ppm 2.19 (s, 3 H) 2.62 (s, 3 H) 6.82 (br. s, 1 H) 7.65 (br. s, 1 H) 7.69 - 7.77 (m, 1 H) 7.88 - 7.97 (m, 1 H) 8.50 (s,l H) 9.13 - 9.18 (m, 1 H) 9.25 (s, 1 H) 9.28 (s, 2 H) 10.47 (s, 1 H) 10.96 (s, 1 H) 12.22 (br. s, 1 H); LRMS m/z calcd = 426.2, observed (M +H) = 427.5.
Example 36
Synthesis of N-(3-(3-(6-amino-2-methylpyrimidin-4-yl)imidazo[ 1 ,2-b]pyridazin-2- ylamino)phenyl)acetamide
Figure imgf000118_0001
Step 1
Nl-(3-(6-amino-2-methylpyrimidin-4-yl)imidazo[l,2-b]pyridazin-2-yl)benzene-l,3- diamine was synthesized utilizing the procedure of Example 27, Step 5. m/z=333.2, cald
Figure imgf000118_0002
Step 2
To a solution of acetyl chloride (0.017 mg, 0.217 μmol) in DMF (0.453 μL) was added Nl-(3-(6-amino-2-methylpyrimidin-4-yl)imidazo[l,2-b]pyridazin-2-yl)benzene-l,3-diamine (0.060 mg, 0.181 μmol) and triethylamine (0.035 μL, 0.253 μmol). The reaction mixture was shaken at rt 15 h, quenched with 100 uL MeOH, then purified directly by prepatory HPLC. 1H NMR (500 MHz, DMSO-J6) δ ppm 2.06 (s, 3 H) 6.98 (br. s., 2 H) 7.12 - 7.29 (m, 2 H) 7.35 (dd, J=8.93, 4.77 Hz, 1 H) 7.58 - 7.79 (m, 2 H) 7.91 (m, 1 H) 8.06 (dd, J=8.93, 1.22 Hz, 1 H) 8.54 - 8.72 (m, 1 H) 9.91 (s, 1 H) 11.47 (s, 1 H). m/z= 375.2, calcd 374.16 for Ci9Hi8N8O.
Example 37
N-(5-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-2-methoxy-
3 -pyridiny l)methanesulfonamide
Figure imgf000119_0001
To a 2-5 niL Personal Chemistry microwave vial was added 4-(2-chloroimidazo[l,2- a]pyridin-3-yl)-6-methyl-l,3,5-triazin-2-amine (0.050 g, 0.192 mmol), N-(5-amino-2- methoxypyridin-3-yl)methanesulfonamide (0.094 g, 0.432 mmol), methanol (1.50 mL) and hydrochloric acid (cone, 5 drops). The reaction mixture was stirred and heated at 155 0C for 80 min in the microwave. The reaction mixture was diluted with DMF and purified by Prep- HPLC (Phenomenex Gemini 5 micron, C 18, 100 x 30 mm, 5 to 55% CH3CN(0.1% TFA)/H2O(0.1% TFA) over 20 min then 100% CH3CN(0.1% TFA) for 3 minutes at 20 ml/min) with the fractions containing suspected product concentrated to afford the title compound as a tan solid (0.011 g, 10 % yield). MS (ESI positive ion) m/z: 442 (M+ 1). 1H NMR (400 MHz, DMSO-J6) δ ppm 10.30 (br s, 1 H), 9.97 (d, J= 6.8 Hz, 1 H), 9.25 (s, 1 H), 8.53 (s, 1 H), 8.37 (s, 1 H), 7.77 (br s, 1 H), 7.48 - 7.60 (m, 3 H), 7.13 (t, J= 6.7 Hz, 1 H), 3.92 (s, 3 H), 3.09 (s, 3 H), 2.43 (s, 3 H).
Example 38
Synthesis of 3 -(6-amino-2-methylpyrimidin-4-yl)-N-( 1 -(pyridin-3 -ylmethyl)- 1 H-pyrazol-3 - yl)imidazo[ 1 ,2-b]pyridazin-2-amine
Figure imgf000119_0002
A glass reaction vessel was charged with 3-(6-(bis(4-methoxybenzyl)amino)-2- methylpyrimidin-4-yl)-N-(l H-pyrazol-3 -yl)imidazo[l,2-b]pyridazin-2-amine (0.200 g, 0.365 mmol) and DMF (3 mL). The reaction vial was purged with nitrogen and sodium hydride (0.015 g, 0.621 mmol) was added. The resulting slurry was stirred at room temperature for 5 minutes, until gas evolution ceased. 3-(Bromomethyl)pyridine (0.063 g, 0.365 mmol) was added and the reaction was heated to 50 0C for 15 h. The mixture was poured into water (20 mL) and extracted with DCM. The combined organic layers were dried (MgSO4) and concentrated in vacuo. The crude reaction was treated with a premixed solution of trifluoromethanesulfonic acid (6.62 mL, 74.5 mmol) in TFA (0.098 mL, 1.278 mmol). The solution was stirred at room temperature for 1 h and concentrated in vacuo. The residue was taken up in minimal MeOH and purified by preparative HPLC (Gilson: 10-80% (0.1% TFA in CH^ CN) in H2O over 15 min). The combined fractions were washed with saturated NaHCO3 and extracted with DCM (3x25 mL). The combined organic layers were dried (MgSO4) and concentrated in vacuo to yield 3-(6-amino-2-methylpyrimidin-4-yl)-N-(l-(pyridin-3-ylmethyl)- lH-pyrazol-3-yl)imidazo[l,2-b]pyridazin-2-amine (37 mg, 0.093 mmol, 25.4 % yield), m/z (ESI, +ve ion) 399.3 (M+H)+. IH NMR (400 MHz, DMSO-J6) δ ppm 2.42 (s, 3 H) 5.32 (s, 2 H) 6.88 (d, J=2.25 Hz, 1 H) 6.97 (br s, 2 H) 7.32 (dd, J=9.00, 4.69 Hz, 2 H) 7.66 (s, 1 H) 7.80 (d, J=2.15 Hz, 2 H) 8.03 (dd, J=8.95, 1.61 Hz, 1 H) 8.49 - 8.56 (m, 2 H) 8.59 (dd, J=4.70, 1.56 Hz, I H) 11.06 (br s, I H)
Example 39
1 -(5 -(3 -(6-amino-2-methylpyrimidin-4-yl)imidazo [ 1 ,2-b]pyridazin-2-ylamino)pyridin-2- yl)azetidin-3-ol
Figure imgf000120_0001
Step l
To 2-chloro-5-nitropyridine (1.30 g, 8.20 mmol) and 3 -hydroxy azetidine hydrochloride (0.923g, 8.43 mmol) was added dry DMF (5ml), and the reaction mixture was stirred in water bath at RT while triethylamine (2.349 mL, 16.85 mmol) was added dropwise. The mixture was stirred RT 3 h, then diluted EtOAc washed water, brine. Aqueous phase was further extracted with EtOAc, and the combined organics evaporated at reduced pressure. The residue was boiled with toluene then allowed to stand at RT. The resulting yellow solid l-(5-nitropyridin-2- yl)azetidin-3-ol was collected by filtration and dried in-vacuuo Step 2
To l-(5-nitropyridin-2-yl)azetidin-3-ol (0.50 g, 2.56 mmol) in ethanol (25 ml) was added ammonium formate (1.615 g, 25.6 mmol). To the reaction mixture was then added a slurry of 5% Pd/C (70 mg) in water ImI under Ar atmosphere and the reaction mixture was stirred under Ar at rt for 45 min in a water bath. The catalyst was removed by filtration through a celite plug and washed with ethanol. Solvent was stripped from the filtrate and residue extracted from minimum of aq NaHCO3 with EtOAc, and solvent stripped to afford a red oil, which was used without further purification in the next step. Step 3 A pressure vessel under nitrogen was charged with Pd2(dba)3 (0.070 g, 0.076 mmol),
JosiPhos CyPF-t-Bu (0.086 g, 0.155 mmol), l-(5-aminopyridin-2-yl)azetidin-3-ol (0.310 g, 1.877 mmol), 6-(2-chloroimidazo[l,2-b]pyridazin-3-yl)-2-methylpyrimidin-4-amine (0.200 g, 0.767 mmol), and cesium carbonate (1.25 g, 3.84 mmol) in t-BuOH (3 mL) and water (0.21 mL, 11.6 mmol). The reaction mixture was degassed with argon, the vessel sealed, then heated at 120 0C for 36 h. Solvent was then stripped at reduced pressure and the residue was chromatographed on silica gel, eluent 90:10:1 DCM MeOH: NH4OH . Fractions containing the product, eluting just above the excess aminopyridine SM were pooled and solvent stripped. The residue was crystallised from hot ethanol as an orange solid and dried in vacuuo to give 70 mg is the title compound m/z 390 MH+
Example 40
Synthesis of 3 -(6-amino-2-methylpyrimidin-4-yl)-N-( 1 -isopentyl- 1 H-pyrazol-3- yl)imidazo[ 1 ,2-b]pyridazin-2-amine
Figure imgf000121_0001
Step l
5-Nitro-lH-pyrazole (0.4 g, 3.54 mmol) was suspended in DMF (7.07 mL) and cooled to 0 0C under N2. To this was added sodium hydride (60% wt in oil) (0.283 g, 7.07 mmol) portionwise and resulting mixture was stirred for 10 mins at 0 0C. l-Bromo-3-methylbutane (1.695 mL, 14.15 mmol) was added via syringe and the mixture was warmed up to room temperature over 1 h and stirred for an additional 2 h. Water (10 mL) was added dropwise and mixture extracted with DCM. Organic layers collected, dried over sodium sulfate, concentrated and dried to completion to afford brown oil. T his was purified by MPLC (30- 50% EtOAc/Hexanes) to provide l-isopentyl-3-nitro-lH-pyrazole (400mg, 2.183 mmol, 61.7 % yield) as a tan solid. MS m/z = 184.2 [M+l]+. Calc'd for C8Hi3N3O2: 183.1. Step 2 l-Isopentyl-3-nitro-lH-pyrazole (0.37 g, 2.020 mmol) and pd/c (10% wt) (0.215 g, 0.202 mmol) under a blanket of N2 were suspended in ethanol (26.9 mL). The reaction mixture was purged with nitrogen and a hydrogen fϊlled-balloon was attached via an adaptor, and stirred at room temperature for 16 h. The reaction mixture was filtered through a celite cake and washed with ethanol and DCM. The filtrate was concentrated to yield 1-isopentyl- lH-pyrazol-3 -amine (309mg, 2.017 mmol, 100 % yield) as a tan oil. MS m/z = 154.2 [M+l]+. Calc'd for C8Hi5N3: 153.1. Step 3
3-(6-Amino-2-methylpyrimidin-4-yl)-N-(l-isopentyl-lH-pyrazol-3-yl)imidazo[l,2- b]pyridazin-2-amine was synthesized utilizing the procedure described in Example 27, Step 5.
Example 41 Synthesis of N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-b]pyridazin-2- yl)amino)phenyl)-4-morpholinecarboxamide
Figure imgf000122_0001
Step l
A glass vial was charged with benzene- 1,4-diamine (1.659 g, 15.34 mmol), cesium carbonate (5.00 g, 15.34 mmol), Pd2(dba)3 (0.281 g, 0.307 mmol), JosiPhos (0.340 g, 0.614 mmol) and 6-(2-chloroimidazo[l,2-b]pyridazin-3-yl)-2-methylpyrimidin-4-amine (0.8 g, 3.07 mmol). The vial was sealed, evacuated and backfilled with nitrogen; this was repeated twice. t-BuOH (8.60 mL) and water (0.553 mL, 30.7 mmol) were added via syringe. The reaction mixture was stirred at 120 0C overnight. The crude material was purified by MPLC (Biotage), eluting with a gradient of 0% to 10% 2M NH3-MeOH in CH2CL2, to provide Nl-(3-(6-amino- 2-methylpyrimidin-4-yl)imidazo[l,2-b]pyridazin-2-yl)benzene-l,4-diamine (0.88 g, 2.65 mmol, 86 % yield) as a brown solid, m/z (ESI, +ve ion) 333.0 (M+H)+ . Step 2
To a 100 rnL round-bottomed flask was added Nl-(3-(6-amino-2-methylpyrimidin-4- yl)imidazo[l,2-b]pyridazin-2-yl)benzene-l,4-diamine (0.385 g, 1.158 mmol), 4-nitrophenyl carbonochloridate (0.233 g, 1.158 mmol) and N-ethyl-N-isopropylpropan-2-amine (0.202 mL, 1.158 mmol) in THF (7.72 mL). The solution was stirred at room temperature overnight. The reaction mixture was concentrated to give a brown solid. This solid and DIPEA (0.588 mL) were dissolved in DMF (7 mL) to provide a stock solution of 4-nitrophenyl 4-(3-(6-amino-2- methylpyrimidin-4-yl)imidazo [ 1 ,2-b]pyridazin-2-ylamino)phenylcarbamate, which was used immediately for the next step. Step 3
To a screw capped vial was added the above stock solution (1 mL) and morpholine (0.021 mL, 0.241 mmol). The reaction mixture was stirred at room temperature overnight. The crude material was purified by preparative HPLC to provide N-(4-(3-(6-amino-2- methylpyrimidin-4-yl)imidazo [ 1 ,2-b]pyridazin-2-ylamino)phenyl)morpholine-4-carboxamide as a yellow solid, m/z (ESI, +ve ion) 446.2 (M+H)+. 1H NMR (500 MHz, DMSO-J6 ) δ 11.22 (s, 1 H), 8.52 - 8.57 (m, 1 H), 8.37 (s, IH), 7.99 - 8.05 (m, IH), 7.62 - 7.71 (m, 3H), 7.33 - 7.49 (m, 2H), 7.29 (dd, J= 8.93, 4.77 Hz, IH), 6.83 - 7.01 (m, 2H), 3.57 (m, 4H), 3.36 - 3.45 (m, 4H), 2.50 (s, 3H).
Example 42
Synthesis ofΛ/-(6-(2-(lH-pyrazol-3-ylamino)imidazo[l,2-b]pyridazin-3-yl)-2- methylpyrimidin-4-yl)acetamide
Figure imgf000123_0001
Step 1
In a 50-mL sealed tube under N2 were dissolved 3-(6-amino-2-methylpyrimidin-4-yl)- Λ/-(lH-pyrazol-3-yl)imidazo[l,2-b]pyridazin-2-amine (1.028 g, 3.35 mmol) (see Example 18) and Boc-anhydride (7.77 mL, 33.5 mmol) in 5 mL of DCM and 5 mL of MeCN and the reaction mixture was stirred and heated at 80 0C. After 5 h, the reaction mixture was dissolved in a minimum of MeOH and then pass thru a conditioned Isolute ® SPE column (SCX-2) and then washed with MeOH. The compound was eluted using a 2 M Ammonia in MeOH and then the solution was concentrated under reduced pressure. The crude mixture was purified by MPLC (ISCO) with DCM:MeOH+NH4OH 100:0 to 90: 10 to afforded tert-butγ\ 3-(3-(6- amino-2-methylpyrimidin-4-yl)imidazo [ 1 ,2-b]pyridazin-2-ylamino)- 1 H-pyrazole- 1 - carboxylate (750 mg, 1.841 mmol, 55.0 % yield), m/z (ESI, +ve ion) 408.2 (M+H)+ Step 2
In a 10-mL sealed tube under N2 were dissolved tert-hvXy\ 3-(3-(6-amino-2- methylpyrimidin-4-yl)imidazo[ 1 ,2-b]pyridazin-2-ylamino)- lH-pyrazole- 1 -carboxylate (200 mg, 0.491 mmol) and acetic anhydride (0.232 mL, 2.454 mmol) in 1 mL of pyridine then stirred and heated at 100 0C. After 1 h, the reaction mixture was concentrated under reduced pressure with silica and purified by MPLC (ISCO) with DCM:MeOH+NH4OH 100:0 to 90:10 to afford tert-hvXy\ 3-(3-(6-acetamido-2-methylpyrimidin-4-yl)imidazo[l,2-b]pyridazin-2- ylamino)-l H-pyrazole- 1 -carboxylate (20 mg, 0.057 mmol, 11.66 % yield). Step 3
In a 25-mL round bottom flask was tert-bvXy\ 3-(3-(6-acetamido-2-methylpyrimidin-4- yl)imidazo[l,2-b]pyridazin-2-ylamino)-lH-pyrazole-l-carboxylate and TFA (0.945 mL, 12.27 mmol) in 1 mL of DCM then stirred at rt. After 1 h, the reaction mixture was concentrated under reduced pressure. The crude mixture was purified by Gilson reverse phase to afforded N-(6-(2-( lH-pyrazol-3 -ylamino)imidazo [ 1 ,2-b]pyridazin-3 -yl)-2-methylpyrimidin-4- yl)acetamide (10 mg, 0.029 mmol, 5.8 % yield) as a yellow solid, m/z (ESI, +ve ion) 350.0 (M+H)+. 1H NMR (400 MHz, DMSO-J6) δ ppm 10.87 (s, 1 H) 10.80 (s, 1 H) 9.30 (s, 1 H) 8.63 (dd, J=4.69, 1.56 Hz, 1 H) 8.06 (dd, J=9.00, 1.56 Hz, 1 H) 7.65 (d, J=2.25 Hz, 1 H) 7.39 (dd, J=9.00, 4.69 Hz, 1 H) 6.84 (d, J=2.25 Hz, 1 H) 2.64 (s, 3 H) 2.15 (s, 3 H).
Example 43 Synthesis of 3-(6-amino-2-methylpyrimidin-4-yl)-6-morpholino-Λ/-(lH-pyrazol-3- yl)imidazo[ 1 ,2-b]pyridazin-2-amine
Figure imgf000125_0001
Step 1
In a 25-mL sealed tube under N2 was dissolved 2,6-dichloroimidazo[l,2-b]pyridazine (1.00 g, 5.32 mmol) in morpholine (2.317 mL, 26.6 mmol) and heated at 60 0C. After 2 h, the reaction mixture was diluted with water and the solid was filtered to afford 4-(2- chloroimidazo[l,2-b]pyridazin-6-yl)morpholine (1.20 g, 5.03 mmol, 95 % yield). The crude 4- (2-chloroimidazo[l,2-b]pyridazin-6-yl)morpholine (1.20 g, 5.03 mmol, 95 % yield) was used without further purification in the next step, m/z (ESI, +ve ion) 239.1 (M+H)+. IH NMR (400 MHz, DMSO-J6) δ ppm 10.49 (br. s., 1 H) 7.90 (d, J=9.78 Hz, 1 H) 7.62 (s, 2 H) 7.36 (d, J=9.49 Hz, 1 H) 6.71 - 6.82 (m, 1 H) 3.78 - 3.87 (m, 4 H) 3.44 - 3.63 (m, 4 H) 2.57 (s, 3 H) Step 2 to 4
The title compound was synthesized according the procedures described in Example 18, Step 3 to 5 using 4-(2-chloroimidazo[l,2-b]pyridazin-6-yl)morpholine. m/z (ESI, +ve ion) 393.2 (M+H)+. NMR
Example 44
Synthesis ofΛ/-(2-methyl-6-(2-(l-methyl-lH-pyrazol-3-ylamino)imidazo[l,2-b]pyridazin-3- yl)pyrimidin-4-yl)acetamide
Figure imgf000125_0002
Step l
The title compound was synthesized utilizing the procedure described in Example 18 Step 5 using l-methyl-lH-pyrazol-3-amine, followed by procedure described in Example 42, Step 2. m/z (ESI, +ve ion) 393.2 (M+H)+. 1H NMR (400 MHz, DMSO-J6) δ ppm 10.81 (s, 1 H) 9.30 (s, 1 H) 8.63 (dd, J=4.69, 1.66 Hz, 1 H) 8.05 (dd, J=9.00, 1.66 Hz, 1 H) 7.60 (d, J=2.05 Hz, 1 H) 7.38 (dd, J=9.00, 4.69 Hz, 1 H) 6.81 (d, J=2.25 Hz, 1 H) 5.75 (s, 1 H) 3.78 (s, 3 H) 2.63 (s, 3 H) 2.15 (s, 3 H)
Example 45
Synthesis of 3-(6-amino-2-methylpyrimidin-4-yl)-7-morpholino-Λ/-(lH-pyrazol-3- yl)imidazo[ 1 ,2-b]pyridazin-2-amine
Figure imgf000126_0001
Step 1 In a 10-mL sealed tube under N2 were dissolved 3,5-dichloropyridazine (1.00 g, 6.71 mmol) and morpholine (1.754 mL, 20.14 mmol) portion wise in 2 mL of MeCN at 0 0C. After 1 h, the reaction mixture was concentrated under reduced pressure with silica and purified by MPLC (ISCO) with DCM:MeOH+NH4OH 100:0 to 90:10 to afford 4-(6-chloropyridazin-4- yl)morpholine (1.100 g, 5.51 mmol, 82 % yield), m/z (ESI, +ve ion) 200.0 (M+H)+ Step 2
In a 10-mL microwave sealed tube under N2 was dissolved 4-(6-chloropyridazin-4- yl)morpholine (1.100 g, 5.51 mmol, 82 % yield) in ammonium hydroxide saturated with NH4Cl (5 mL) then stirred and heated at 150 0C with microwave. After 2 h, the reaction mixture was concentrated under reduced pressure with silica and purified by MPLC (ISCO) with DCM:MeOH+NH4OH 100:0 to 90: 10 to afforded 5-morpholinopyridazin-3-amine (0.50Og, 2.77 mmol, 41.3 % yield), m/z (ESI, +ve ion) 181.2 (M+H)+ Step 3 to 7
The title compound was synthesized according to the procedure described in Example 18, Steps 1 to 5 using 5-morpholinopyridazin-3-amine. m/z (ESI, +ve ion) 393.2 (M+H)+. 1H NMR (400 MHz, DMSO-J6) δ ppm 10.48 (s, 1 H) 8.58 (d, J=2.84 Hz, 1 H) 7.65 (d, J=2.35 Hz, 1 H) 7.54 (s, 1 H) 7.27 (d, J=2.84 Hz, 1 H) 6.78 (d, J=2.25 Hz, 1 H) 3.74 - 3.85 (m, 4 H) 3.39 - 3.54 (m, 4 H) 2.57 (s, 3 H) Example 45
Synthesis of 3-(6-amino-2-methylpyrimidin-4-yl)-8-fluoro-Λ/-(lH-pyrazol-3-yl)imidazo[l,2- a]pyridin-2-amine
Figure imgf000127_0001
Step 1 to 5
The title compound was synthesized synthesized according to the procedure described in Example 18, Step 1 to 5 using 3-fluoropyridin-2-amine. m/z (ESI, +ve ion) 325.0 (M+H)+. 1H NMR (400 MHz, DMSO-J6) δ ppm 10.04 (br. s., 1 H) 8.61 (d, J=7.34 Hz, 1 H) 7.61 (d, J=2.25 Hz, 1 H) 7.38 (dd, J=10.76, 7.83 Hz, 1 H) 7.03 - 7.11 (m, 1 H) 6.81 (s, 1 H) 6.68 (s, 1 H) 2.52 (br. s., 3 H)
Example 46
Synthesis of 3-(2-methyl-6-(6-(4-methylpiperazin- 1 -yl)pyridazin-3-ylamino)pyrimidin-4-yl)- N-(lH-pyrazol-3-yl)imidazo[l,2-b]pyridazin-2-amine
Figure imgf000127_0002
Step 1
3-Amino-6-chloropyridazine (0.500 g, 3.86 mmol) was combined with 1-methyl- piperazine (2.143 mL, 19.30 mmol) and the reaction was heated at 150 0C for 4 h in the microwave. The reaction mixture was concentrated in vacuo and the crude material purified via MPLC (isocratic 90:10:1 DCM:MeOH:NH4OH) to provide 6-(4-methylpiperazin-l- yl)pyridazin-3 -amine as a orange solid. The material was moved forward without further purification, m/z (ESI, +ve ion) 194.2 (M+H)+ Step 2
In a sealed tube under nitrogen were dissolved 2-chloro-3-(6-chloro-2- methylpyrimidin-4-yl)imidazo[l,2-b]pyridazine (0.100 g, 0.357 mmol), JosiPhos CyPF-t-Bu (0.040 g, 0.071 mmol), 6-(4-methylpiperazin-l-yl)pyridazin-3 -amine (0.083 g, 0.428 mmol), Pd2(dba)3 (0.033 g, 0.036 mmol), and cesium carbonate (0.349 g, 1.071 mmol) in t-BuOH
(1.000 mL) and water (0.064 mL, 3.57 mmol). The vessel was purged with nitrogen, and then stirred at 90 0C for 18 h. The crude material was purified via MPLC, eluting with a gradient of 0% to 100% 90:10:1 DCM:MeOH:NH4OH to provide N-(6-(2-chloroimidazo[l,2-b]pyridazin- 3-yl)-2-methylpyrimidin-4-yl)-6-(4-methylpiperazin-l-yl)pyridazin-3-amine (0.086 g, 0.197 mmol, 55.1 % yield), m/z (ESI, +ve ion) 437.1 (M+H)+ Step 3
The title compound was synthesized according to the procedure described in Example 27, Step 5 using N-(6-(2-chloroimidazo [1, 2-b]pyridazin-3 -yl)-2-methylpyrimidin-4-yl)-6-(4- methylpiperazin-l-yl)pyridazin-3 -amine, m/z (ESI, +ve ion) 484.2 (M+H)+. 1H NMR (DMSO-d6) δ: 12.18 (br. s., IH), 10.95 (s, IH), 10.28 (s, IH), 8.52 - 8.67 (m, 2H), 7.94 - 8.10 (m, 2H), 7.65 (s, IH), 7.25 - 7.47 (m, 2H), 6.86 (s, IH), 3.41 - 3.62 (m, 4H), 2.58 (s, 3H), 2.41 - 2.48 (m, 4H), 2.24 (s, 3H)
Example 47 Synthesis of 3 -(6-(5 -(2-methoxyethoxy)pyridazin-3 -ylamino)-2-methylpyrimidin-4-yl)-N-( 1 H- pyrazol-3-yl)imidazo[ 1 ,2-b]pyridazin-2-amine
Figure imgf000128_0001
Step 1 To a solution of 3,5-dichloropyridazine (2.53 g, 16.98 mmol) in THF (16.98 mL) at 0
0C under nitrogen was added sodium methoxyethoxide (8.89 mL, 18.68 mmol) dropwise, after which the flask was warmed to RT and stirred for 1 h. The reaction mixture was concentrated in vacuo, and purified via MPLC (eluting with a gradient 0-100% 90: 10: 1 DCM:MeOH:NH4OH in DCM) to yield 3-chloro-5-(2-methoxyethoxy)pyridazine (2.34 g, 12.41 mmol, 73.1 % yield) as an orange oil. m/z (ESI, +ve ion) 189.3 (M+H)+ Step 2
In a sealed pressure vessel under nitrogen were dissolved 6-(2-chloroimidazo[l,2- b]pyridazin-3-yl)-2-methylpyrimidin-4-amine, JosiPhos CyPF-t-Bu (0.468 g, 0.844 mmol), 3- chloro-5-(2-methoxyethoxy)pyridazine (1.831 g, 9.71 mmol), Pd2(dba)3 (0.386 g, 0.422 mmol), and cesium carbonate (6.87 g, 21.10 mmol) in t-BuOH (11.82 mL) and water (0.760 mL, 42.2 mmol), and then heated at 120 0C for 5 h. The crude reaction mixture was concentrated and then purified via MPLC (eluting with 0-100% 90:10:1 DCM:MeOH:NH4OH in DCM) to yield crude material as a dark brown solid. The solid triturated with IPA, yielding N-(6-(2-chloroimidazo[l,2-b]pyridazin-3-yl)-2-methylpyrimidin-4-yl)-5-(2- methoxyethoxy)pyridazin-3-amine. The material was carried forward without further purification, m/z (ESI, +ve ion) 413.0 (M+H)+ Step 3 The title compound was synthesized according to the procedure described in Example
27, Step 5 using N-(6-(2-chloroimidazo[l,2-b]pyridazin-3-yl)-2-methylpyrimidin-4-yl)-5-(2- methoxyethoxy)pyridazin-3-amine. m/z (ESI, +ve ion) 460.0 (M+H)+. 1H NMR (DMSO-d6) δ: 12.19 (br. s., IH), 10.92 (s, IH), 10.62 (s, IH), 8.78 (s, IH), 8.68 (d, J = 2.5 Hz, IH), 8.61 (d, J = 3.5 Hz, IH), 8.05 (d, J = 8.7 Hz, IH), 7.95 (s, IH), 7.66 (br. s., IH), 7.37 (dd, J = 9.1, 4.6 Hz, IH), 6.86 (br. s., IH), 4.30 (dd, J = 5.2, 3.5 Hz, 2H), 3.66 - 3.81 (m, 2H), 3.34 (s, 3H), 2.65 (s, 3H).
Biological Examples
Example 1 In vitro assays The following assays can be used to determine the degree of activity of individual compounds as PI3 kinase and/or mTOR inhibitors. Recombinant expression of PI3K enzymes
Full length pi 10 subunits of PI3K α, β and δ, N-terminally labeled with polyHis tag, can be co-expressed with p85 with Baculo virus expression vectors in sf9 insect cells. Pl 10/p85 heterodimers can be purified by sequential Ni-NTA, Q-HP, Superdex-100 chromatography. Purified α, β and δ isozymes can be stored at -20 0C in 2OmM Tris, pH 8, 0.2M NaCl, 50% glycerol, 5mM DTT, 2mM Na cholate. Truncated PI3Kγ, residues 114-1102, N-terminally labeled with polyHis tag, can be expressed with Baculo virus in Hi5 insect cells. The γ isozyme can be purified by sequential Ni-NTA, Superdex-200, Q-HP chromatography. The γ isozyme can be stored frozen at -80 0C in NaH2PO4, pH 8, 0.2M NaCl, 1% ethylene glycol, 2mM β-mercaptoethanol.
Figure imgf000130_0001
In vitro PB kinase enzyme assays (PBK ATPLoss)
PBK enzyme assays (alpha, beta, delta and gamma) can be performed in 25 μL with the above final concentrations of components in white polyproplyene plates. Phosphatidyl inositol phosphoacceptor, PtdIns(4,5)P2 (e.g,. P4508) can be obtained from Echelon Biosciences, Salt Lake City, UT. The ATPase activity of the alpha and gamma isozymes may not be greatly stimulated by PtdIns(4,5)P2 under these conditions, it can be omitted from the assay of these isozymes. Test compounds can be dissolved in DMSO and diluted with threefold serial dilutions. The compound in DMSO (1 μL) may be added per test well, and the inhibition relative to reactions containing no compound, with and without enzyme can be determined. After assay incubation at RT, the reaction can be stopped and residual ATP can be determined by addition of an equal volume of a commercial ATP bio luminescence kit (Perkin Elmer EasyLite, Perkin Elmer, Waltham, MA) according to the manufacturer's instructions, and detected using an Analyst GT luminometer.
Cell-based phospho-AKT Ser473 assay (HCTl 16 Cell) This assay determines the ability of a compound to inhibit the phosphorylation of
Serine 473 in Akt using a MSD based sandwich immunoassay (Meso Scale Detection, Meso Scale Discovery (MSD), Gaithersburg, MD). HCT 116 human colon carcinoma cell lines can be grown in McCoy's 5 A growth medium (GIBCO, Carlsbad, CA) containing 10% FBS (GIBCO, Carlsbad, CA) and Xl Penicillin-streptomycin-glutamine (GIBCO, Carlsbad, CA). Prior to the assay, cells can be detached from the culture flask with trypsin, and re-suspended in complete media to give a final concentration of 1.6 x 105 cells per mL. Aliquots (100 μl) of the HCTl 16 cell suspension can be seeded into each well of a 96 well tissue culture plate to give a final density of 16,000 cells per well. Cells can then be incubated overnight at 37 0C. The following day the cells can be treated with serially diluted test compounds and incubated for 2 hours at 37 0C. The culture media on the HCT 116 cells can be replaced with 189 μL McCoys media, supplemented with 0.1% BSA (ICN Biomedicals, Inc., Costa Mesa, CA). Compounds can be prepared as either 10 mM or 0.5 mM stock solutions in DMSO, and serially diluted 3 fold in a 10-point dose-response curve to give final concentrations that are 200-fold greater than the desired final test concentration. Aliquots (lμL) of serially-diluted compounds can be transferred to 96 well tissue culture plates containing the HCT 116 cells. As a minimum response control, each plate can contain wells having a final concentration of 2.5μM of a potent PI3K inhibitor which had previously been shown to completely inhibit Akt phosphorylation at this test concentration. As a maximum response control, wells can contain 0.5% DMSO in place of compound. The plates can be mixed at 700 rpm for 2 min to ensure even distribution of the test compound and incubated for 2 hours at 37 0C. Cells can then be stimulated with insulin-like growth factor 1 (Sigma, St Louis, MO) at final concentration of 100ng/ml for 15 minutes at 37 0C. The media can then be removed and the cells treated with 80 μL cell-lysis buffer (MSD) containing a cocktail of protease and phosphatase inhibitors for one hour at 4 0C. 25 μL Cell lysate can then be transferred to pre-blocked MSD assay plates pre-coated with a capture antibody specific for Akt, and the plates can be incubated for 2 hours at room temperature. The cell lysates can then be removed and plates can then be washed four times with 200 μl per well of Tris wash buffer (500 mM Tris, PH 7.5, 1.5 M NaCl, 0.2% Tween-20). Subsequently cells can be incubated for 1 hour at room temperature with a 25 μL solution containing the detection antibody, anti-phospho Akt (Ser 473) labeled with an electrochemiluminescent compound (Meso Scale Discovery SULPHO-TAG™ label, MSD, Gaithersburg, MD). The detection antibody can be removed and plates can then be washed four times with 200 μL per well of Tris wash buffer. An aliquot of 150 μL of diluted MSD read buffer can then be applied to each well, and the electrochemiluminescent signal can be measured using a MSD SECTOR™ plate reader (Meso Scale Discovery, Gaithersburg, MD). This instrument measures the intensity of emitted light to determine a quantitative measure of phosphorylated Akt in each well. The dose-response data obtained with each compound can be analyzed and the IC50 inhibition of Akt phosphorylation at Ser473 can be calculated. pAkt AlphaScreen OJ87 Cell)
The pAkt AlphaScreen® assay (PerkinElmer, Waltham, MA) determines whether there is phosphorylation of Akt at Serine 473 by recruitment of a phosphospecific antibody. This assay was performed using U87 MG cells. The U87 growth media consists of MEM (Gibco, Carlsbad, CA) supplemented with 10% FBS (Gibco,), Ix Non-Essential Amino Acids (Gibco,) and Ix Penicillin/Streptomycin/Glutamine (Gibco). The cells were maintained weekly using 0.05% Trypsin (Gibco) and replated in 150 mm TC- Treated Culture Dishes (Corning, Corning, NY).
The first day of the assay, the adherent cells were trypsinized, media was added to the loose cells and cells were mixed to a homogenous mixture. 0.5 ml of the homogenous mixture was counted on the Beckman Coulter® Vi-CELL XR (Fullerton, CA). 50 frames of cells were counted and the number of viable cells was determined. The cells were then diluted to 0.25 million cells per ml, and centrifuged at 200 rcf for 5 minutes. The media was removed and the cells were reconstituted in fresh media for plating. The cells were plated at 20 μl per well on the PerkinElmer® FlexDrop PLUS in Low Volume 384 Well White Tissue Culture Plates
(Corning) with a final cell density of 5K cells per well. The plates were incubated overnight at 37° Celsius, 5% CO2.
On the second day, the compound plates were prepared, the cells were treated with compound and the pAkt reaction mix was added to the cell lysate. 384 well compound plates were prepared containing 1 μl of compound per well starting at 5 mM and diluted 1 :2 across the row, resulting in a 22 well serial dilution. 39 μl of growth media was added to the compound plate in rows 1-22 using the PerkinElmer® FlexDrop PLUS resulting in a DMSO concentration of 2.5%. The cell plates and diluted compound plates were put onto the VELOCITY 11 VPREP 384 ST where the compound plate was mixed and 5 μl of serially diluted compound or controls was added to the cell plate. The final concentration of the compounds was 25 μM serially diluted to 11.9 pM in 0.5% DMSO.
The cell plates were then incubated with compound for two hours at 37 0C, 5% CO2. After two hours, the media in the cell plates was aspirated using the BioTek® ELx405HT plate washer (Winooski, VT) removing the majority of media and compound without disturbing the adherent U87 cells. The following assay reagents are components of the SureFire® Akt (Ser 473) Phosphorylation 5OK Point Kit (TGR BioSciences, Adelaide, Austalia) and an IgG Detection Kit (PerkinElmer, Waltham, MA). 5 μl of Ix Lysis Buffer was added to each well using the PerkinElmer® FlexDrop PLUS. The plates were then incubated at room temperature on a shaker for ten minutes. The AlphaScreen® reaction was prepared under low light conditions (subdued or green light) including p-Akt (Ser 473) Reaction Buffer, Dilution Buffer, Activation Buffer, Acceptor Beads and Donor Beads at a ratio of 40:20: 10: 1 : 1 respectively. The AlphaScreen® reaction was added to the cell lysate at 6 μl per well using the PerkinElmer® FlexDrop PLUS. The plates were placed in a humid environment to reduce edge effects and incubated overnight at room temperature with restricted air flow in the dark.
On the final day of the experiment, the plates were read on the PerkinElmer® En Vision 2103 Multilable Reader using the standard AlphaScreen® readout. The POC is calculated and the data is analyzed to report the IC50 IP for pAkt at Serine 473.
Activity data for the compounds tested in the PI3K cell based Akt assay is provided in Table 1 under the column heading U87
In vitro PI3K AlphaScreen® assay
The PI3K AlphaScreen® assay (PerkinElmer, Waltham, MA) measures the activity of a panel of four phosphoinositide 3-kinases: PI3Kα, PI3Kβ, PI3Kγ, and PI3Kδ. Each of these enzymes phosphorylates the 3'-hydroxyl group on phosphatidylinositiol (4,5)-bisphosphate (PIP2) to produce phosphatidylinositol (3,4,5)-trisphosphate (PIP3). This phosphorylation activity is measured using a GST-tagged PIP3 binding protein (Echelon Biosciences, Salt Lake City, UT), an anti-GST-tagged Acceptor bead, and streptavidin-Donor bead. The interaction of biotinylated-PIP3 analog (IP4) and the PIP3 binding protein brings both Acceptor and Donor beads together producing, upon excitation of the Donor beads at 680 nm, a singlet oxygen species leading to the luminescent AlphaScreen® signal. When PIP3 is produced via phophorylation of PIP2 by a PI3K, PIP3 competes with biotinylated-PIP3 analog (IP4) for binding to the PIP3 binding protein. In the absence of this interaction, proximity of the Donor and Acceptor beads is decreased, producing a loss of luminescent signal which is inversely proportional to PI3K activity. An inhibitor reduces activity of the enzyme, resulting in less PIP3 production and greater luminescence.
The enzyme reaction buffer is made using sterile water (Baxter, Deerfield, IL) and 5OmM Tris HCl pH 7, 14mM MgCl2, 2mM sodium cholate, and 10OmM NaCl. 2mM DTT is added fresh the day of the experiment. The AlphaScreen® reaction buffer is made using sterile water and 1OmM Tris HCl pH 7.5, 15OmM NaCl, 0.10% Tween 20, and 3OmM EDTA. ImM DTT is added fresh the day of the experiment.
The source plates for this assay are 384-well Greiner clear polypropylene plates containing test compounds at 5mM and diluted 1 :2 over 22 points. Columns 23 and 24 contain only DMSO as these are designated for positive and negative controls. Source plates are replicated into 384-well Optiplates (PerkinElmer, Waltham, MA), 0.5μL/well, to make assay- ready plates.
The different PBK isoforms are each diluted in enzyme reaction buffer to 2X working solutions. PBKα is diluted to 1.6nM, PBKβ is diluted to 0.8nM, PBKγ is diluted to 15nM, and PBKδ is diluted to 1.6nM. Two different 2X substrate solutions are made in enzyme reaction buffer. In one solution, PI(4,5)P2 (Echelon Biosciences, Salt Lake City, UT) is diluted to lOμM and ATP is diluted to 20μM. This solution is used in the assays testing PBKα and PBKβ. In a second solution, PI(4,5)P2 is diluted to lOμM and ATP is diluted to 8μM. This solution is used in the assays testing PBKγ and PBKδ. The AlphaScreen® reaction solutions are made using beads from the anti-GST
AlphaScreen® kit (PerkinElmer, Waltham, MA). Two solutions are made in Alphascreen reaction buffer to 4X working concentrations. In one solution, biotinylated-IP4 (Echelon Biosciences, Salt Lake City, UT) is diluted to 4OnM and streptavadin-Donor Beads are diluted to 80μg/mL. In the second solution, PIP3-binding protein (Echelon Biosciences, Salt Lake City, UT) is diluted to 4OnM and anti-GST- Acceptor Beads are diluted to 80μg/mL. lOμL/well of enzyme reaction buffer is added to Column 24 of the assay ready plates in place of enzyme. This is done for plates in the PBKα, β, and δ assays.
Using a 384-well dispensing Multidrop (Titertek, Huntsville, AL), lOμL/well of 2X enzyme (PBKα, β, δ) is added to Columns 1-23 of the appropriate assay ready plates (for PBKγ lOμL is added to Columns 1-24). lOμL/well of the appropriate substrate solution (the solution with 20μM ATP for PBKα and β assays, and the solution with 8μM ATP for PBKγ and δ assays) is then added to Columns 1-24 of the plates. Plates are then incubated at room temperature for 20 minutes.
In the dark, lOμL/well of the Donor Bead solution is added to Columns 1-24 of the plates to quench the enzyme reaction. The plates are incubated at room temperature for 30 minutes. Still in the dark, lOμL/well of the Acceptor Bead solution is also added to Columns 1- 24 of the plates. The plates are then incubated in the dark for 1.5 hours. The plates are read on an Envision Multilabel Plate Reader (PerkinElmer, Waltham, MA) with a 680nm excitation filter and a 520-620nm emission filter. Activity data for the compounds tested in the assay is provided in Table 1 under the column heading PBKα AlphaScreen®. niTOR LanthaScreen
The mTOR LanthaScreen is a TR-FRET assay measuring the phosphorylation of mTOR's substrate 4EBP 1. 384 well compound plates were prepared containing 1 μl of compound per well starting at 5 mM and diluted 1 :2 across the row, resulting in a 22 well serial dilution. 24 μl of assay buffer (Invitrogen, PV4794) with 2 mM DTT was added to the compound plate in rows 1-24 using the VELOCITYI 1 VPREP 384 ST resulting in a DMSO concentration of 4%. The compound plate was mixed and 2.5 μl of serially diluted compound or controls was added to the assay plate (Costar, 3658).
The assay was conducted on the PerkinElmer® FlexDrop PLUS. A 5 μl mix of 800 nM GFP-4E-BP1 (Invitrogen, PV4759) and 20 μM ATP (Amgen) was added to rows 1-24. 2.5 μl of 0.6 μg/ml of mTOR Enzyme (Amgen) was added to rows 1-23. 2.5 μl of assay buffer was added to row 24 for the low control. The final concentration of the compounds was 50 μM serially diluted to 23.84 pM in 1% DMSO. The final high control had 1% DMSO and the low control was a no enzyme control with a concentration of 1% DMSO. The final concentrations in the assay reagents were 400 nM GFP-4E-BP1, 10 μM of ATP and 0.15 μg/ml of mTOR enzyme. The compound, enzyme, and substrate were incubated for 90 minutes. At this point, 10 μl of stop solution was added (20 mM Tris, pH 7.5 (Invitrogen, 15567-027), 0.02 % Sodium Azide (Teknova, S0208), 0.01 % NP-40 (Roche, 11754599001), 20 mM EDTA (Invitrogen, 15575-038) and 4 nM of Tb-anti-p4E-BPl (Invitrogen, PV4758)) for a final concentration of 2 nM of Tb-anti-p4E-BP 1.
Sixty minutes later the plates were read on the PerkinElmer® En Vision 2103 Multilable Reader using the Excitation filter 340 nm and the Emission filters 520 nm and 495 nm. The ratio of 520 nm/495 nm was calculated and the POC data was analyzed to report the IC50 IP for the phosphorylation of 4EBP 1. Activity data for the compounds tested in the assay is provided in Table 1 under the column heading mTOR.
p4EBPl AlphaScreen
The p4EBP 1 AlphaScreen assay determines whether there is phosphorylation of 4EBP 1 at Thr37/Thr46 by recruitment of a phosphospecific antibody. This assay was performed using U87 MG cells. The U87 growth media consists of MEM (Gibco, 51200-038) supplemented with 10% FBS (Gibco, 16140-071), Ix Non-Essential Amino Acids (Gibco, 11140-050) and Ix Penicillin/Streptomycin/Glutamine (Gibco, 10378-016). The cells were maintained weekly using 0.05% Trypsin (Gibco, 25300-054) and replated in 150 mm TC- Treated Culture Dishes (Corning, 430599).
The first day of the assay, the adherent cells were trypsinized, media was added to the loose cells and cells were mixed to a homogenous mixture. 0.5 ml of the homogenous mixture was counted on the Beckman Coulter® Vi-CELL XR. 50 frames of cells were counted and the number of viable cells was determined. The cells were then diluted to 0.25 million cells per ml, and centrifuged at 200 rcf for 5 minutes. The media was removed and the cells were reconstituted in fresh media for plating. The cells were plated at 20 μl per well on the PerkinElmer® FlexDrop PLUS in Low Volume 384 Well White Tissue Culture Plates (Corning, 3826) with a final cell density of 5K cells per well. The plates were incubated overnight at 37° Celsius, 5% CO2.
On the second day, the compound plates were prepared, the cells were treated with compound and the p4EBP 1 reaction mix was added to the cell lysate. 384 well compound plates were prepared by Amgen's Sample Bank containing 1 μl of compound per well starting at 5 mM and diluted 1 :2 across the row, resulting in a 22 well serial dilution. 39 μl of growth media was added to the compound plate in rows 1-22 using the PerkinElmer® FlexDrop PLUS resulting in a DMSO concentration of 2.5%. The control columns were added manually; 40 μl of 2.5% DMSO (Sigma, D4540- 100ml) in growth media was added to the plate for the high control and 40 μl of 50 μM of AMG2203766 with 2.5% DMSO was added to the plate as the low control. The cell plates and diluted compound plates were put onto the VELOCITYI 1
VPREP 384 ST where the compound plate was mixed and 5 μl of serially diluted compound or controls was added to the cell plate. The final concentration of the compounds was 25 μM serially diluted to 11.9 pM in 0.5% DMSO. The final high control had 0.5% DMSO and the low control concentration was 10 μM AMG2203766 in 0.5% DMSO. The cell plates were then incubated with compound for two hours at 37° Celsius, 5% CO2. After two hours, the media in the cell plates was aspirated using the BioTek® ELx405HT plate washer removing the majority of media and compound without disturbing the adherent U87 cells. The following assay reagents are components of the SureFire Phospho-4EBP 1 (Thr37/Thr46) 5OK Point Kit (TGR BioSciences, TGR4ES50K) and an IgG Detection Kit (PerkinElmer, 6760617R). 5 μl of Ix Lysis Buffer was added to each well using the PerkinElmer® FlexDrop PLUS . The plates were then incubated at room temperature on a shaker for ten minutes. The AlphaScreen reaction was prepared under low light conditions (subdued or green light) including p-4E-BPl (Thr37/46) Reaction Buffer, Activation Buffer, Acceptor Beads and Donor Beads at a ratio of 60: 10: 1 : 1 respectively. The AlphaScreen reaction was added to the cell lysate at 6 μl per well using the PerkinElmerR FlexDrop PLUS. The plates were placed in a humid environment to reduce edge effects and incubated overnight at room temperature with restricted air flow in the dark.
On the final day of the experiment, the plates were read on the PerkinElmer® En Vision 2103 Multilable Reader using the standard AlphaScreen readout. The POC is calculated and the data is analyzed to report the IC50 IP for p4EBP 1 at Thr37/Thr46.
Table 1
Figure imgf000137_0001
Figure imgf000138_0001
Formulation Examples
The following are representative pharmaceutical formulations containing a compound of Formula (I).
Tablet Formulation The following ingredients are mixed intimately and pressed into single scored tablets.
Ingredient Quantity per tablet Mg compound of this invention 400 cornstarch 50 croscarmellose sodium 25 lactose 120 magnesium stearate 5
Capsule Formulation
The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule.
Ingredient Quantity per capsule me compound of this invention 200 lactose spray dried 148 magnesium stearate 2
Capsule Formulation The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule.
Ingredient Quantity per capsule me compound of this invention 200 lactose spray dried 148 magnesium stearate 2
The foregoing invention has been described in some detail by way of illustration and example, for purposes of clarity and understanding. It will be obvious to one of skill in the art that changes and modifications may be practiced within the scope of the appended claims. Therefore, it is to be understood that the above description is intended to be illustrative and not restrictive. The scope of the invention should, therefore, be determined not with reference to the above description, but should instead be determined with reference to the following appended claims, along with the full scope of equivalents to which such claims are entitled. All patents, patent applications and publications cited in this application are hereby incorporated by reference in their entirety for all purposes to the same extent as if each individual patent, patent application or publication were so individually denoted.

Claims

What is claimed is:
1. A compound of Formula (I):
Figure imgf000140_0001
(I) wherein:
Ar1 is an aryl, heteroaryl, cycloalkyl or heterocyclyl ring, wherein each ring is substituted with Rd, Re, or Rf where Rd, Re, or Rf are independently selected from hydrogen, halo, haloalkyl, haloalkoxy, cyano, nitro, alkyl, alkenyl, alkynyl, substituted alkyl, aryl, heteroaryl, heterocycloalkyl, -C(=O)NRaRa, -C(=O)Rb, -C(=O)ORb, -C(=NRa)NRaRa, -ORa, -OC(=O)Rb, -OC(=O)NRaRa, -O-alkylN(Ra)C(=O)ORb, -OC(=O)N(Ra)S(=O)2Rb, -O- alkylNRaRa, -O-alkylORa, -SRa, -S(=O)Rb, -S(=O)2Rb, -S(=O)2NRaRa, -S(=O)2N(Ra)C(=O)Rb, -S(=O)2N(Ra)C(=O)ORb, -S(=O)2N(Ra)C(=O)NRaRa, -NRaRa, -N(Ra)C(=O)Rb, -N(Ra)C(=O)ORb, -N(Ra)C(=O)NRaRa, -N(Ra)C(=S)NRaRa, -N(Ra)C(=NRa)NRaRa, -N(Ra)S(=O)2Rb, -N(Ra)S(=O)2NRaRa, -NRa-alkylene-NRaRa, or -NRa-alkylene-ORa ;
R1 is hydrogen or alkyl;
R2 is methyl or ethyl;
Z1 -N- or -CR3- where R3 is H or alkyl; Z2 is -N- or -CR4- where R4 is Rd, -(alkylene)heterocycloalkyl,
-(alkylene)nO(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -( alkylene)n-N(Ra)- (alkylene)nheteroaryl, or -( alkylene)nO(alkylene)nheteroaryl;
Z3 is -N- or -CR5- where R5 is Rd, -(alkylene)naryl, -(alkylene)-heteroaryl, -(alkylene)heterocycloalkyl, -(alkylene)nO-(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)nheteroaryl, or -(alkylene)nO(alkylene)nheteroaryl provided that only two of Z1, Z2 and Z3 can simultaneously be -N-; or when Z2 is -CR4- and Z3 is -CR5- then R4 and R5 together with the atoms to which they are attached can form ring A which is phenyl or a 5 or 6 membered heteroaryl ring and ring A is substituted with Rg or Rh where Rg or Rh are independently Rd, -(alkylene)n_ heterocycloalkyl, -(alkylene)nO(alkylene)naryl, -(alkylene)nN(Ra)(alkylene)naryl, -( alkylene)nN(Ra)(alkylene)nheteroaryl, or -(alkylene)nO(alkylene)nheteroaryl;
Z4 is -N- or -C-; provided that when Z5 is -CR6- where R6 together with Z6 forms phenyl, and Z7 is -N-, then Z4 is -C-;
Z5, Z6 or Z7 are each independently selected from -N- or -CR6- provided at least one of Z4, Z5, Z6 and Z7 is -N- where R6 is Rd ; or R6 together with the adjacent ring atom can form phenyl or 5 or six membered heteroaryl ring wherein the phenyl or heteroaryl ring is substituted with R1, RJ, or Rk where R1, RJ, or Rk are independently Rd, -(alkylene)naryl, - (alkylene)heteroaryl, -( alkylene)heterocycloalkyl, -(alkylene)nO(alkylene)naryl, - (alkylene)nN(Ra)(alkylene)naryl, -(alkylene 2)nN(Ra)( alkylene)nheteroaryl, or -( alkylene)nO(alkylene)nheteroaryl; each Ra is independently hydrogen or Rb; or when two Ra are attached to a nitrogen atom, either alone or part of another group, the two Ras together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring with is optionally substituted with one, two or three substitutents independently selected from oxo, halo, alkyl, alkenyl, alkynyl, cyano, nitro, alkylcarbonyl, carboxy, alkoxycarbonyl, hydroxyl, alkoxy, alkonyloxy, alkylthio, alkylsulfonyl, -alkyl-OH, aminosulfonyl, sulfonylamino, amino, alkylamino, or dialkylamino; each Rb is independently alkyl, cycloalkyl, phenyl, heteroaryl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl is substituted with 0, 1, 2 or 3 substituents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, hydroxyl, unsubstituted heterocycloalkyl, phenyl, alkylcarbonylamino, alkylamino or dialkylamino; and each n is independently 0 or 1 ; or a pharmaceutically acceptable salt thereof; provided the compound of Formula (I) is not:
Figure imgf000141_0001
or a salt thereof.
2. The compound of Claim 1 wherein each Ra is independently hydrogen or Rb; or when two Ra are attached to a nitrogen atom, either alone or part of another group, the two Ras together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring with is optionally substituted with one, two or three substitutents independently selected from oxo, halo, alkyl, alkenyl, alkynyl, cyano, nitro, alkylcarbonyl, carboxy, alkoxycarbonyl, hydroxyl, alkoxy, alkonyloxy, alkylthio, alkylsulfonyl, aminosulfonyl, sulfonylamino, amino, alkylamino, or dialkylamino; and each Rb is independently alkyl, cycloalkyl, phenyl, heteroaryl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl is substituted with 0, 1, 2 or 3 substituents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino or dialkylamino.
3. The compound of Claim 1 wherein R1 is hydrogen and in R
Figure imgf000142_0001
N Z1 is -
N-, Z2 is -CH- and Z3 is -CR5- where R5 is Rd or -(alkylene)nN(Ra)(alkylene)nheteroaryl.
4. The compound of Claim 1 or 3 wherein R2 is methyl.
5. The compound of any of the Claims 1, 3 or 4 wherein Ar1 is aryl substituted as defined above.
6. The compound of any of the Claims 1, 3 or 4 wherein Ar1 is heteroaryl substituted as defined above.
7. The compound of any of the Claims 1, 3 or 4 wherein Ar1 is pyrazolyl substituted as defined above.
8. The compound of any of the Claims 1, 3 or 4 wherein Ar1 is an aryl, heteroaryl, or heterocyclyl ring, wherein each ring is substituted with Rd, Re, or Rf where Rd, Re, or Rf are independently selected from hydrogen, halo, haloalkyl, alkyl, alkyl substituted with -NRyRy (where Ry is alkyl) or alkoxy, -(CRyRy)nY (where Y is aryl or heteroaryl), -ORX (where Rx is hydrogen or alkyl), heteroaryl, heterocycloalkyl, -C(=O)NRaRa, -C(=O)Rb, -C(=O)ORb, -ORa, -O-alkylORa, -S(=O)2Rb, -NRaRa, -N(Ra)C(=O)Rb, -N(Ra)C(=O)NRaRa, -N(Ra)S(=O)2Rb, or -NRa-alkylene-ORa ; each Ra is independently hydrogen or Rb; or when two Ra are attached to a nitrogen atom, either alone or part of another group, the two Ras together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring with is optionally substituted with one, two or three substitutents independently selected from alkyl, alkylcarbonyl, - alkylOH, or hydroxyl; and each Rb is independently alkyl, cycloalkyl, phenyl, heteroaryl, or benzyl wherein the alkyl, cycloalkyl, phenyl, or benzyl is substituted with 0, 1, 2 or 3 substituents independently selected from halo, alkyl, haloalkyl, alkoxy, amino, cyano, alkylamino dialkylamino, hydroxyl, unsubstituted heterocycloalkyl, or phenyl.
9. The compound of any of the Claims 1, 3 or 4 wherein Ar1 is 5-fluoro-6-methoxy-3- pyridinyl, lH-pyrazol-3-yl, 1 -methyl- lH-pyrazol-3-yl, 4-(3-ethylureido)phenyl, IH- pyrrolo[2,3-b]pyridin-6-yl, 6-(trifluoromethyl)-2-pyridinyl, 2-pyridinyl, lH-pyrrolo[3,2- b]pyridine-5-yl, 4-hydroxypyridin-2-yl, 4-(3-phenylureido)phenyl, 5-(3-ethylureido)pyridine- 2-yl, 4-(3-methylureido)phenyl, 3-hydroxyphenyl, 4-acetylaminophenyl, 4-(3-(4- pyridinyl)ureidophenyl, 3-amino-pyrimidin-5-yl, 4-pyridazinyl, 1 -acetyl- lH-pyrazol-3-yl, 3- (methylsulfonyl)phenyl, 3-pyridinyl, 3-methyl-5-isothiazolyl, 4-(4-methyl-l- piperazinyl)phenyl, lH-pyrazolo[3,4-c]pyridin-3-yl, 4-fluoro-lH-indazol-3-yl)imidazo[l,2- b]pyridazin-2-yl, l-(2-(dimethylamino)ethyl)-lH-pyrazol-3-yl, lH-pyrazolo[3,4-b]pyridin-3- yl, 3-(4-methyl-l-piperazinyl)-phenyl, 3-acetylaminophenyl, pyridine -2 -yl, 1 -ethyl- IH- pyrazol-3-yl, quinolin-3-yl, isoquinolin-3-yl, l-methylaminocarbonyl-lH-pyrazol-3-yl, 1- methylaminocarbonyl- 1 H-pyrazol-5 -yl, 1 -(2-hydroxyethylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 - (2-hydroxyethylaminocarbonyl)- 1 H-pyrazol-5 -yl, 1 -(morpholin-4-ylcarbonyl)- 1 H-pyrazol-3 - yl, 1 -(morpholin-4-ylcarbonyl)- 1 H-pyrazol-5 -yl, l-(phenoxycarbonyl)-l H-pyrazol-3 -yl, 1- (phenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(phenylaminocarbonyl)- 1 H-pyrazol-5 -yl, 1 - (methoxycarbonyl)- 1 H-pyrazol-3 -yl, 1 -(methoxycarbonyl)- 1 H-pyrazol-5 -yl, 1 -(phenoxy- carbonyl)- 1 H-pyrazol-4-yl, 1 -(methylaminocarbonyl)- 1 H-pyrazol-4-yl, 1 -(pyridine-2-yl)- 1 H- pyrazol-3-yl, l-(methylsulfonyl)-l H-pyrazol-3 -yl, l-(benzoxazol-2-yl)-l H-pyrazol-3 -yl, 1- (benzimidazol-2-yl)-l H-pyrazol-5 -yl, l-(isopropylaminocarbonyl)-l H-pyrazol-3 -yl, 1- (benzylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(ethylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2- methoxyphenyl-aminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2-methylphenylaminocarbonyl)- 1 H- pyrazol-3 -yl, 1 -(4-methoxyphenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(2-fluorophenyl- aminocarbonyl)-l H-pyrazol-3 -yl, l-(4-cyanophenylaminocarbonyl)-l H-pyrazol-3 -yl, l-(2- trifluoromethyl-phenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(3 -methylphenylaminocarbonyl)- 1 H-pyrazol-3 -yl, l-(2,5-difluorophenylaminocarbonyl)-lH-pyrazol-3-yl, l-(3,5- difluorophenylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(4-methylphenylaminocarbonyl)- IH- pyrazol-3-yl, 1 -(3, 5 -dichlorophenyl-aminocarbonyl)-l H-pyrazol-3 -yl, l-(2-chloro-5- trifluoromethy lphenylaminocarbony I)- 1 H-pyrazol-3 -yl, 1 -(2,5 -dichlorophenylamino- carbonyl)-l H-pyrazol-3 -yl, l-(2,3-dichloro-phenylaminocarbonyl)-lH-pyrazol-3-yl, l-(2- chloro-4-trifluoromethyl-phenylaminocarbony I)- 1 H-pyrazol-3 -yl, 1 -(benzimidazol-2-yl)- 1 H- pyrazol-3-yl, l-(imidazol-2-yl)-l H-pyrazol-5 -yl, 1-methylsulfonyl-l H-pyrazol-3 -yl, 1- aminocarbonyl- 1 H-pyrazol-3 -yl, 1 -aminocarbonyl- 1 H-pyrazol-5 -yl, 1 -azetidin- 1 -ylcarbonyl- 1 H-pyrazol-3 -yl, 1 -azetidin- 1 -ylcarbonyl- 1 H-pyrazol-5 -yl, 1 -(2-methylaminoethylamino)- carbonyl- 1 H-pyrazol-3 -yl, 1 -(2-methylaminoethylamino)carbonyl- 1 H-pyrazol-5 -yl, 1 - dimethylaminocarbonyl- 1 H-pyrazol-3 -yl, 1 -dimethylaminocarbony 1- 1 H-pyrazol-5 -yl, 1 - piperazin- 1 -ylcarbonyl- 1 H-pyrazol-3 -yl, 1 -(2-methoxyethylamino)carbonyl- 1 H-pyrazol-3 -yl, 1 -(2-methoxyethylamino)carbonyl- 1 H-pyrazol-5 -yl, 1 -(2-morpholin-4-ylethylamino)- carbonyl- 1 H-pyrazol-3 -yl, 1 -(2-methylpropylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -( 1 - phenethylaminocarbonyl)- 1 H-pyrazol-3 -yl, 1 -(benzyl)- 1 H-pyrazol-3 -yl, 1 -(pyridine-3 - ylmethyl)- 1 H-pyrazol-3 -yl, 1 -( 1 ,4-benzoxazin-3 -(4H)-one-7-yl)- 1 H-pyrazol-3 -yl, 5 -(4- hydroxyazetidin- 1 -yl)pyridine-3 -yl, 5 -(4-(propan-2-ol)azetidin- 1 -yl)pyridine-3 -yl, quinolin-7- yl, quinolin-6-yl, isoquinolin-7-yl, 6-methoxypyridin-3-yl, 6-ethoxypyridin-3-yl, lH-indazol- 3 -yl, 5 -fluoro-6-methoxypyridin-3 -yl, 1 -(6-(2-methoxyethoxy)pyrimidin-3 -yl)- 1 H-pyrazol-3 - yl, 3-cyclopropylureidophenyl, 3-benzylcarbonylaminophenyl, 3-benzoylaminophenyl, 3-(4- chlorophenylsulfonylamino)-phenyl, 3 -methylsulfonylaminophenyl, 3 -(4-fluorophenyl- ureido)phenyl, 3-(benzylureido)-phenyl, 3-ethylaminocarbonylphenyl, 6-ethylamino- carbonylpyridin-3yl, 3-ethylcarbonyl-aminophenyl, 3-methylaminocarbonylphenyl, 3- ethylaminocarbonylphenyl, 3 -isopropylaminocarbonylphenyl, 3 -phenylaminocarbonylphenyl, β-methylaminocarbonyl-pyridin-Syl, β-isopropylaminocarbonylpyridin-Syl, 6-phenylamino- carbonylpyridin-3yl, 3-aminocarbonylphenyl, 4-(2-diethylaminoethylaminocarbonyl)phenyl, 4- phenylaminocarbonylphenyl, 1 -(3 -methylbutanoyl)- 1 H-pyrazol-3 -yl, 1 -isopropyloxy-carbonyl- 1 H-pyrazol-3 -yl, 1-isopropyloxycarbonyl-l H-pyrazol-5 -yl, 2-methoxypyridin-4-yl, 1- isopropylcarbonyl-1 H-pyrazol-3 -yl, 1-isopropylcarbonyl-l H-pyrazol-5 -yl, l-(3,3- dimethylbutanoyl)- 1 H-pyrazol-3 -yl, 1 -cyclohexyloxycarbonyl- 1 H-pyrazol-5 -yl, 1 - cyclohexyloxycarbonyl-1 H-pyrazol-3 -yl, 2-methoxypyridin-3-yl, 2-oxopyridin-3-yl, l-(3- methylbutyl)-l H-pyrazol-3 -yl, l-(2-methoxyethyl)-l H-pyrazol-3 -yl, lH-indazol-4-yl, 6- chloro-5 -methylsulfonylaminopyridin-3 -y 1, 6-methoxy-5 -methylsulfonylaminopyridin-3 -yl, lH-indazol-6-yl, lH-benzimidazol-6-yl, 4-(3-cyclopropylureido)phenyl, 4-piperidin-l- ylcarbonylaminophenyl, 4-pyrrolidin- 1 -ylcarbonylaminophenyl. 4-azetidin- 1 - ylcarbonylaminophenyl, 4-( 1 -methylpiperazin- 1 -ylcarbonylaminophenyl, 4-(3 -(2- methoxyethyl)ureido)phenyl, 4-morpholin-4-yl-ylcarbonylaminophenyl, 4-( 1 -acetylpiperazin- 1 -ylcarbonylaminophenyl, thiazol-3-yl, 4-(2-methylmorpholin-4-yl-ylcarbonylamino)phenyl, or 4-(3-methylmorpholin-4-yl-ylcarbonylamino)phenyl.
10. The compound of any of the Claims 1 and 3-9 wherein
Figure imgf000145_0001
where R1 is defined above.
11. The compound of any of the Claims 1 and 3-9 wherein
Figure imgf000145_0002
wherein R1 is Rd selected from hydrogen, halo, haloalkyl, substituted alkyl, aryl, heteroaryl, heterocycloalkyl, -C(=O)NRaRa, -C(=O)ORb,-CORb, -ORa, -O-alkylNRaRa, -O- alkylORa, -NRaRa, or -NRa-alkylene-ORa ; each Ra is independently hydrogen or Rb or when two Ra are attached to a nitrogen atom, either alone or part of another group, the two Ras together with the nitrogen atom to which they are attached can form a monocyclic heterocyclyl ring, each Rb is independently alkyl, phenyl, or heteroaryl wherein the alkyl, is substituted with 0, 1, 2 or 3 substituents independently selected from alkyl, or alkoxy and wherein: and where the aryl ring can optionally be substituted with one, two or three substituents independently selected from alkyl, alkoxy, or halo, and/or one or two substituents independently selected from, -C(=O)ORy, -C(=O)NRXRX, -ORX, -SRX, -S(=O)2Ry, -S(=O)2NRXRX, -NRXRX, -N(Rx)C(=O)Ry, -(CRxRx)mN(Rx)C(=O)Ry or -(CRxRx)mORx where m is 1-3, each Rx is hydrogen or Ry and Ry is independently hydrogen, alkyl, or cycloalkyl; the heterocycloalkyl ring is optionally be substituted with one, two or three substituents independently selected from alkyl, hydroxy, -C(=O)Ry, -C(=O)ORy, -C(=O)NRXRX, -S(=O)2Ry, or -(CRxRx)mORx where m is 0-3, each Rx is hydrogen or Ry and Ry is independently hydrogen or alkyl;. the heteroaryl ring is optionally be substituted with one, two or three substituents independently selected from alkyl, -ORX, -C(=O)NRXRX, or -(CRxRx)mORx where m is 1-3, each Rx is hydrogen or Ry and Ry is independently hydrogen or alkyl; and substituted alkyl is an alkyl radical in which one, two, or three hydrogen atoms are independently replaced by hydroxyl or -NCORy where Ry is alkyl.
12. The compound of any of the Claims 1 and 3-9 wherein
Figure imgf000146_0001
ΛΛΛΛΛ where R1 is hydrogen, 1-piperazinyl, 4-methyl-l-piperazinyl, 4-(l-methylethyl)-l- piperazinyl, 4-(2-methoxy ethyl)- 1-piperazinyl, fluoro, 3-(methylsulfonyl)phenyl, 4-pyridinyl, 2-methoxyethoxy, 4-morpholinyl, 2-(l-pyrrolidinyl)ethoxy, l-tert-butoxycarbonyl-3,6- dihydro-l(2H)-pyridin-4-yl, 3-methoxyphenyl, 3-fluoro-4-methylaminocarbonylphenyl, A- acetylaminomethylphenyl, l-(2-methoxy ethyl)- lH-pyrazol-4-yl, 2-fluorophenyl, 2- methylphenyl, 2-methoxy-3-pyridinyl, 3-furanyl, 3,5-dimethyl-4-isoxazolyl, 4-fluoro-2- methylphenyl, 2-(l-methylethoxy)phenyl, 3-(methylsulfanyl)phenyl, l,2,3,6-tetrahydro-4- pyridinyl, 3-methyl-5-isoxazolyl, l-(2-methoxyethyl)-lH-pyrazol-4-yl, 3-acetylaminophenyl, 3-cyclopropylaminophenyl, 3-isopropanoylaminophenyl, 3-pyridazinyl, 1 -methyl- 1,2, 3,6- tetrahydro-4-pyridinyl, 4-acetylaminophenyl, 2-methyl-4-pyridinyl, 2-methyl-3-pyridinyl, 5- pyrimidinyl, 3-pyridinyl, phenylamino, 4-methoxyphenylamino, methoxy, 3 -fluorophenyl, A- methylaminocarbonylphenyl, pyridine-3-ylamino, 4-acetylaminophenyl, 3-methoxy- phenylamino, pyridine-4-ylamino, pyrimidin-5-ylamino, 4-acetylaminophenylamino, 2- methyl-4-pyridinylamino, 4-methyl-3-pyridinyl, 2,6-dimethyl-4-pyridinyl, 4-isopropylamino- carbonylphenyl, 3,4-dihydro-l(2H)-isoquinolinone-6-yl, 4-ethylaminocarbonylphenyl, A- carboxy-2-methylphenyl, 4-methyl-5 -pyrimidinyl, 2-methyl-4-methylaminocarbonylphenyl, hydroxyl, 6-methylaminocarbonylpyridin-3-yl, 2-(l-pyrrolidinyl)ethoxy, 4-N,N- dimethylaminocarbonylpiperazin- 1 -yl, 4-methylsulfonylpiperazin- 1 -yl, 4-N- methylaminocarbonylpiperazin- 1 -yl, 4-acetylpiperazin- 1 -yl, 3 -hydroxymethyl-4- methoxyphenyl, pyridine-4-yl, 3,6-dihydro-2H-pyran-4-yl, 2-methyl-4-pyridinyl, 1-methyl- lH-imidazol-5-yl, 3,4-dihydroxybutyl, l-(l-methylethyl)-lH-pyrazol-4-yl, aminocarbonyl, quinolin-7-yl, 4-methylsulfonylphenyl, 4-methylaminosulfonyl-phenyl, 3-methyl- sulfonylphenyl, 2-methoxyethylamino, 4-hydroxypiperidin-l-yl, 1 -methyl- lH-pyrazol-4-yl, 2- hydroxyprop-2-yl, acetyl, or trifluoromethyl.
13. The compound of Claim 1 selected from the group consisting of: 3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-N-( 1 H-pyrazol-3 -yl)imidazo [ 1 ,2-a]pyridin-2-amine
3 -(3 -(2-methyl-9H-purin-4-yl)imidazo [ 1 ,2-a]pyridin-2-ylamino)phenol
3 -(4-methyl-6-(methylamino)- 1 ,3 ,5 -triazin-2-yl)-N-( 1 H-pyrazol-3 -yl)imidazo [ 1 ,2-a]pyridin-2- amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(6-methoxypyridin-3-yl)imidazo[l,2-a]pyridin-2- amine
N-(3-(2-methyl-9H-purin-4-yl)imidazo[l,2-a]pyridin-2-yl)-lH-indazol-4-amine
7-chloro-3-(4-amino-6-methyl)-l,3,5-triazin-2-yl)-N-(lH-pyrazol-3-yl)imidazo[l,2-a]pyridin-2- amine
3 -(6-amino-2-methylpyrimidin-4-yl)-N-(l H-pyrazol-3 -yl)imidazo[l,2-b]pyridazin-2-amine
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-y l)-N-(5 -fluoro-6-methoxypyridin-3 -yl)imidazo [ 1 ,2- a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(6-methoxypyridin-3-yl)-6-fluoroimidazo[l,2- a]pyridin-2-amine
3 -( 1 -(6-amino-2-methylpyrimidin-4-yl)-3 -methyl- 1 H-pyrazol-5 -ylamino)phenol
5 -( 1 H-pyrazol-5 -ylamino)- 1 -(6-amino-2-methy lpyrimidin-4-yl)-N-(pyridin-3 -yl)- 1 H-pyrazole- 3-carboxamide
5 -(I H-pyrazol-5 -ylamino)- l-(6-amino-2-methy lpyrimidin-4-yl)-N-(pyridin-4-yl)-lH-pyrazole- 3-carboxamide
(5 -(I H-pyrazol-5 -ylamino)- l-(6-amino-2-methylpyrimidin-4-yl)-l H-pyrazol-3 - yl)(morpholino)methanone
5 -( 1 H-pyrazol-5 -ylamino)- 1 -(6-amino-2-methylpyrimidin-4-yl)-N-methyl- 1 H-pyrazole-3 - carboxamide ethyl 5 -( 1 H-pyrazol-3 -ylamino)- 1 -(6-amino-2-methylpyrimidin-4-yl)- 1 H-pyrazole-3 - carboxylate
5 -(I H-pyrazol-5 -ylamino)- l-(6-amino-2-methylpyrimidin-4-yl)-N-cyclopropyl-l H-pyrazole-3 - carboxamide methyl 5 -( 1 H-pyrazol-3 -ylamino)- 1 -(6-amino-2-methylpyrimidin-4-yl)- 1 H-pyrazole-3 - carboxylate
6-(5 -( 1 H-pyrazol-3 -ylamino)-3 -methyl- 1 H-pyrazol- 1 -yl)-2-methylpyrimidin-4-amine
6-(5 -( 1 H-pyrazol-3 -ylamino)-3 -methyl- 1 H-pyrazol- 1 -yl)-N,2-dimethylpyrimidin-4-amine 5-(lH-pyrazol-3-ylamino)-l-(6-amino-2-methylpyrimidin-4-yl)-lH-pyrazole-3-carboxylic acid
N-( 1 -(6-amino-2-methylpyrimidin-4-yl)-3 -methyl- 1 H-pyrazol-5 -yl)- 1 H-indazol-5 -amine
N-(l-(6-amino-2-methylpyrimidin-4-yl)-3 -methyl- lH-pyrazol-5-yl)-lH-indazol-4-amine
phenyl 1 -(6-amino-2-methylpyrimidin-4-yl)-5 -(3 -hydroxyphenylamino)- 1 H-pyrazole-3 - carboxylate
6-(5 -(6-methoxypyridin-3 -ylamino)-3 -methyl- 1 H-pyrazol- 1 -yl)-2-methy lpyrimidin-4-amine
N-(I -(6-amino-2-methylpyrimidin-4-yl)- 1 H-pyrazol-5 -yl)- 1 H-indazol-4-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(5-fluoro-6-methoxy-3-pyridinyl)-6-(l- piperazinyl)imidazo[ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(5-fluoro-6-methoxy-3-pyridinyl)-6-(4-methyl-l- piperazinyl)imidazo[ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl- 1,3, 5-triazin-2-yl)-N-(5-fluoro-6-methoxy-3-pyridinyl)-6-(4-(l -methyl- ethyl)- 1 -piperazinyl)-imidazo [ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(5-fluoro-6-methoxy-3-pyridinyl)-6-(4-(2- methoxy ethyl)- 1 -piperazinyl)imidazo [ 1 ,2-a]pyridin-2-amine
3-(2-methyl-6-(phenylamino)-4-pyrimidinyl)-N-lH-pyrazol-3-ylimidazo[l,2-b]pyridazin-2- amine
3 -(6-((2-fluorophenyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2- b]pyridazin-2-amine
3 -(6-((4-fluorophenyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2- b]pyridazin-2-amine
3 -(6-((4-methoxyphenyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2- b]pyridazin-2-amine
N-(I -methyl- 1 H-pyrazol-3 -yl)-3 -(2 -methyl-6-(3 -pyridazinylamino)-4-pyrimidinyl)imidazo [1,2- b]pyridazin-2-amine
3 -(2-methyl-6-((6-(4-methyl- 1 -piperazinyl)-3 -pyridazinyl)amino)-4-pyrimidinyl)-N- 1 H- pyrazol-3 -ylimidazo [ 1 ,2-b]pyridazin-2-amine
3 -(6-((5 -methoxy-2-pyridinyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [1,2- b]pyridazin-2-amine
3 -(6-((3 -fluorophenyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [1,2- b]pyridazin-2-amine
3 -(6-((4-methoxy-2-pyridinyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [1,2- b]pyridazin-2-amine 3 -(2-methyl-6-((6-(4-morpholinyl)-3 -pyridazinyl)amino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 - ylimidazo [ 1 ,2-b]pyridazin-2-amine
1 -(4-((3 -(6-amino-2-methy l-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)phenyl)-3 - ethylurea
3-(6-amino-2-methyl-4-pyrimidinyl)-N-lH-pyrrolo[2,3-b]pyridin-6-ylimidazo[l,2-a]pyridin-2- amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(6-(trifluoromethyl)-2-pyridinyl)imidazo [ 1 ,2-a]pyridin- 2-amine
3-(6-amino-2-methyl-4-pyrimidinyl)-N-2-pyridinylimidazo[l,2-a]pyridin-2-amine
3 -(6-((5 -(2-methoxyethoxy)-3 -pyridazinyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 - ylimidazo [l,2-b]pyridazin-2-amine
N-(3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)-lH-pyrrolo[3,2-b]pyridin- 5 -amine
2-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)-4-pyridinol
l-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-3- phenylurea
1 -(6-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-a]pyridin-2-yl)amino)-3-pyridinyl)-3- ethylurea
3 -(2 -methyl-6-((6-(4-morpho linyl)-3 -pyridazinyl)amino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 - ylimidazo [ 1 ,2-a]pyridin-2-amine l-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-3- methylurea
Structure contains atom(s) in disallowed valence state!
3-((3-(6-((5-(2-methoxyethoxy)-3-pyridazinyl)amino)-2-methyl-4-pyrimidinyl)imidazo[l,2- b]pyridazin-2-yl)amino)phenol
N-(4-((3-(6-((5-(2-methoxyethoxy)-3-pyridazinyl)amino)-2-methyl-4-pyrimidinyl)imidazo[l,2- b]pyridazin-2-yl)amino)phenyl)acetamide
3 -(6-((5 -methoxy-3 -pyridazinyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo- [ 1 ,2-b]pyridazin-2-amine l-ethyl-3-(4-((3-(6-((5-(2-methoxyethoxy)-3-pyridazinyl)amino)-2-methyl-4-pyrimidinyl)- imidazo [ 1 ,2-b]pyridazin-2-yl)amino)phenyl)urea
3 -(2-methyl-6-((5-methyl-3 -pyridazinyl)amino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [1,2- b]pyridazin-2-amine
1 -(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-a]pyridin-2-yl)amino)phenyl)-3-(4- pyridinyl)urea 3 -(6-((5 -(2-methoxyethoxy)-3 -pyridazinyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 - ylimidazo [ 1 ,2-a]pyridin-2-amine
N-(6-(2-((4-((ethylcarbamoyl)amino)phenyl)amino)imidazo[l,2-a]pyridin-3-yl)-2-methyl-4- pyrimidinyl)acetamide
3-(2-methyl-6-((5-(2,2,2-trifluoroethoxy)-3-pyridazinyl)amino)-4-pyrimidinyl)-N-lH-pyrazol- 3 -ylimidazo [ 1 ,2-b]pyridazin-2-amine
N~5 — (3 -(6-((5 -(2-methoxyethoxy)-3 -pyridazinyl)amino)-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2- b]pyridazin-2-yl)-2,5-pyrimidinediamine
8-fluoro-3-(6-((5-(2-methoxyethoxy)-3-pyridazinyl)amino)-2-methyl-4-pyrimidinyl)-N-lH- pyrazol-3 -ylimidazo [ 1 ,2-a]pyridin-2-amine
N-(6-(8-fluoro-2-((l-methyl-lH-pyrazol-3-yl)amino)imidazo[l,2-a]pyridin-3-yl)-2-methyl-4- pyrimidinyl)acetamide
3-(6-amino-2-methyl-4-pyrimidinyl)-N-4-pyridazinylimidazo[l,2-b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(l -methyl- 1 H-pyrazol-3 -yl)-6-(3- (methylsulfonyl)phenyl)imidazo [ 1 ,2-a]pyridin-2-amine
3 -(2-methyl-6-(methylamino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2-b]pyridazin-2- amine
N-(I -acetyl- 1 H-pyrazol-3 -yl)-3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2- amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-l H-pyrazol-3 -yl-6-(4-pyridinyl)imidazo[ 1,2- b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-6-(2-methoxyethoxy)-N-l H-pyrazol-3 -ylimidazo [1,2- b]pyridazin-2-amine
N-(2-methyl-6-(2-(l H-pyrazol-3 -ylamino)imidazo[ l,2-b]pyridazin-3-yl)-4-pyrimidinyl)- acetamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-6-(4-morpholinyl)-N-l H-pyrazol-3 -ylimidazo [1,2- b]pyridazin-2-amine
3 -(2 -methyl-6-(methylamino)-4-pyrimidinyl)-6-(4-morpholinyl)-N-l H-pyrazol-3 - ylimidazo [l,2-b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -yl-6-(2-( 1 -pyrro lidinyl)ethoxy)- imidazo[ 1 ,2-b]pyridazin-2-amine
3 -(2-methyl-6-(2-pyrazinylamino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2-b]pyridazin- 2-amine l,l-dimethyl-3-(2-methyl-6-(2-(lH-pyrazol-3-ylamino)imidazo[l,2-b]pyridazin-3-yl)-4- pyrimidinyl)urea l-ethyl-3 -(2 -methyl-6-(2-(l H-pyrazol-3 -ylamino)imidazo[ l,2-b]pyridazin-3-yl)-4- pyrimidinyl)urea 4-methyl-N-(2-methyl-6-(2-(lH-pyrazol-3-ylamino)imidazo[l,2-b]pyridazin-3-yl)-4- pyrimidinyl)- 1 -piperazinecarboxamide
N-(2-methyl-6-(2-(lH-pyrazol-3-ylamino)imidazo[l,2-b]pyridazin-3-yl)-4-pyrimidinyl)-4- morpholinecarboxamide
3 -(2-methyl-6-( 1 H-pyrazol-3 -ylamino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2- b]pyridazin-2-amine
3 -(6-(dimethylamino)-2-methyl-4-pyrimidinyl)-N-( 1 -methyl- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2- b]pyridazin-2-amine
3 -(2 -methyl-6-(l, 3 -thiazol-2-ylamino)-4-pyrimidinyl)-N-l H-pyrazol-3 -ylimidazo [1,2- b]pyridazin-2-amine
3 -(2-methyl-6-(2-pyridinylamino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2-b]pyridazin- 2-amine
3 -(2-methyl-6-(3 -pyridazinylamino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2- b]pyridazin-2-amine
3 -(2-methyl-6-(4-pyrimidinylamino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2- b]pyridazin-2-amine
3 -(2-methyl-6-(2-pyrimidinylamino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2- b]pyridazin-2-amine
N-(2-methyl-6-(2-((l -methyl- lH-pyrazol-3-yl)amino)imidazo[l,2-b]pyridazin-3-yl)-4- pyrimidinyl)acetamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-7-(4-morpholinyl)-N-l H-pyrazol-3 -ylimidazo [1,2- b]pyridazin-2-amine
3 -(2 -methyl-6-(methylamino)-4-pyrimidinyl)-7-(4-morpholinyl)-N-l H-pyrazol-3 - ylimidazo [l,2-b]pyridazin-2-amine
N-(2-methyl-6-(2-(l H-pyrazol-3 -ylamino)imidazo[ l,2-b]pyridazin-3-yl)-4-pyrimidinyl)- methanesulfonamide
((2 -methyl-6-(2-(l H-pyrazol-3 -ylamino)imidazo[ l,2-b]pyridazin-3-yl)-4-pyrimidinyl)amino)- acetonitrile
3-(6-amino-2-methyl-4-pyrimidinyl)-8-fluoro-N-lH-pyrazol-3-ylimidazo[l,2-a]pyridin-2- amine
N~2~,N~2~-dimethyl-N-(2-methyl-6-(2-((l -methyl- 1 H-pyrazol-3 -yl)amino)imidazo[ 1,2- b]pyridazin-3-yl)-4-pyrimidinyl)glycinamide
N-(2-methyl-6-(2-((l -methyl- lH-pyrazol-3-yl)amino)imidazo[l,2-b]pyridazin-3-yl)-4- pyrimidinyl)glycinamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(3 -(methylsulfonyl)phenyl)imidazo [ 1 ,2-b]pyridazin-2- amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-3 -pyridinylimidazo [ 1 ,2-b]pyridazin-2-amine 3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(3 -methyl-5 -isothiazolyl)imidazo [ 1 ,2-b]pyridazin-2- amine tert-butyl 4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2- a]pyridin-6-yl)-3 ,6-dihydro- 1 (2H)-pyridinecarboxylate
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-y l)-6-(3 -methoxyphenyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2- a]pyridin-2-amine
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-6- yl)-2-fluoro-N-methylbenzamide
N-(4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin- 6-yl)benzyl)acetamide
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-y l)-6-( 1 -(2-methoxy ethyl)- 1 H-pyrazol-4-yl)-N- 1 H- pyrazol-3 -ylimidazo [ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(2-fluorophenyl)-N-lH-pyrazol-3-ylimidazo[l,2- a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(2-methylphenyl)-N-lH-pyrazol-3-ylimidazo[l,2- a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(2-methoxy-3-pyridinyl)-N-lH-pyrazol-3- ylimidazo[l ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(3-furanyl)-N-lH-pyrazol-3-ylimidazo[l,2-a]pyridin- 2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(3,5-dimethyl-4-isoxazolyl)-N-lH-pyrazol-3- ylimidazo[l ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(4-fluoro-2-methylphenyl)-N-lH-pyrazol-3- ylimidazo[l ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(2-(l-methylethoxy)phenyl)-N-lH-pyrazol-3- ylimidazo[l ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(3-(methylsulfanyl)phenyl)-N-lH-pyrazol-3- ylimidazo[l ,2-a]pyridin-2-amine
N-(4-(3-(4-methyl-6-(methylamino)-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2- a]pyridin-6-yl)benzyl)acetamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-lH-pyrazol-3-yl-6-(l,2,3,6-tetrahydro-4-pyridinyl)- imidazo[ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(3-methyl-5-isoxazolyl)-N-lH-pyrazol-3-ylimidazo- [ 1 ,2-a]pyridin-2-amine
6-(l-(2-methoxyethyl)-lH-pyrazol-4-yl)-3-(4-methyl-6-(methylamino)-l,3,5-triazin-2-yl)-N- 1 H-pyrazol-3 -ylimidazo [l,2-a]pyridin-2-amine
N-(3-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin- 6-yl)phenyl)acetamide N-(3-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin- 6-yl)phenyl)cyclopropanecarboxamide
N-(4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin- 6-yl)phenyl)-2-methylpropanamide
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-y I)-N- 1 H-pyrazol-3 -yl-6-(3 -pyridazinyl)imidazo [ 1 ,2- a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(l-methyl-l,2,3,6-tetrahydro-4-pyridinyl)-N-lH- pyrazol-3-ylimidazo[ 1 ,2-a]pyridin-2-amine
N-(4-(3 -(6-amino-2-methyl-4-pyrimidinyl)-2-( 1 H-pyrazol-3 -ylamino)imidazo [ 1 ,2-b]pyridazin- 6-yl)benzyl)acetamide
N-(2-methyl-6-(2-(l H-pyrazol-3 -ylamino)imidazo[l,2-b]pyridazin-3-yl)-4-pyrimidinyl)- cyclopropanecarboxamide
N-(4-(3-(6-amino-2-methyl-4-pyrimidinyl)-2-(l H-pyrazol-3 -ylamino)imidazo[l,2-b]pyridazin- 6-yl)phenyl)acetamide
4-(3-(6-amino-2-methyl-4-pyrimidinyl)-2-(l H-pyrazol-3 -ylamino)imidazo[l,2-b]pyridazin-6- yl)-N-methylbenzamide
2-methoxy-N-(2-methyl-6-(2-((l -methyl- 1 H-pyrazol-3 -yl)amino)imidazo[l,2-b]pyridazin-3- yl)-4-pyrimidinyl)acetamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2-a]pyridin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(l -methyl- 1 H-pyrazol-3 -yl)imidazo[l,2-a]pyridin-2- amine
2-cyano-N-(2-methyl-6-(2-((l -methyl- 1 H-pyrazol-3 -yl)amino)imidazo[l,2-a]pyridin-3-yl)-4- pyrimidinyl)acetamide
2-(3-methoxyphenyl)-N-(2-methyl-6-(2-((l -methyl- lH-pyrazol-3-yl)amino)imidazo[l,2-a]- pyridin-3-yl)-4-pyrimidinyl)acetamide
2-(3-fluorophenyl)-N-(2-methyl-6-(2-((l-methyl-lH-pyrazol-3-yl)amino)imidazo[l,2-a]- pyridin-3-yl)-4-pyrimidinyl)acetamide
2-(2,4-difluorophenyl)-N-(2-methyl-6-(2-((l-methyl-lH-pyrazol-3-yl)amino)imidazo[l,2- b]pyridazin-3-yl)-4-pyrimidinyl)acetamide
3 -cyano-N-(2-methyl-6-(2-((l -methyl- 1 H-pyrazol-3 -yl)amino)imidazo[ l,2-b]pyridazin-3-yl)-4- pyrimidinyl)benzamide
3 ,3 ,3 -trifluoro-N-( 1 -methyl- 1 H-pyrazol-3 -yl)-N-(3 -(2-methyl-6-((3 ,3 ,3 -trifluoropropanoyl)- amino)-4-pyrimidinyl)imidazo[ 1 ,2-a]pyridin-2-yl)propanamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -methyl- 1 H-pyrazol-3 -yl)-6-(2-methyl-4-pyridinyl)- imidazo[ 1 ,2-b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -methyl- 1 H-pyrazol-3 -yl)-6-(2-methyl-3 -pyridinyl)- imidazo[ 1 ,2-b]pyridazin-2-amine 3 ,3 ,3 -trifluoro-N-(2-methyl-6-(2-(( 1 -methyl- 1 H-pyrazol-3 -yl)amino)imidazo [ 1 ,2-b]pyridazin-3 - yl)-4-pyrimidinyl)propanamide
3 ,3 ,3 -trifluoro-N-(2-methyl-6-(2-(( 1 -methyl- 1 H-pyrazol-3 -yl)amino)imidazo [ 1 ,2-a]pyridin-3 - yl)-4-pyrimidinyl)propanamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -methyl- 1 H-pyrazol-3 -yl)-6-(5 -pyrimidiny l)imidazo- [ 1 ,2-b]pyridazin-2-amine
3 -(6-((6-(2-methoxyethoxy)-3 -pyridazinyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 - ylimidazo[l,2-b]pyridazin-2-amine
2-(4-(6-((2-methyl-6-(2-(l H-pyrazol-3 -ylamino)imidazo[ l,2-b]pyridazin-3-yl)-4- pyrimidinyl)amino)-3-pyridazinyl)-2-morpholinyl)ethanol
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(4-(4-methyl- 1 -piperazinyl)phenyl)imidazo [1,2- b]pyridazin-2-amine
3-(2-methyl-6-((6-(2-methyl-4-morpholinyl)-3-pyridazinyl)amino)-4-pyrimidinyl)-N-lH- pyrazol-3-ylimidazo[ 1 ,2-b]pyridazin-2-amine
3-(2-methyl-6-((6-(3-methyl-4-morpholinyl)-3-pyridazinyl)amino)-4-pyrimidinyl)-N-lH- pyrazol-3-ylimidazo[ 1 ,2-b]pyridazin-2-amine
N-(3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)-lH-pyrazolo[3,4- c]pyridin-3 -amine
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(4-fluoro-lH-indazol-3-yl)imidazo[l,2-b]pyridazin-2- amine
N-(3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)-lH-pyrazolo[4,3- c]pyridin-3 -amine
N-(2-methyl-6-(2-(l H-pyrazol-3 -ylamino)imidazo[l,2-a]pyridin-3-yl)-4-pyrimidinyl)acetamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(2-(dimethylamino)ethyl)- 1 H-pyrazol-3 -yl)imidazo- [ 1 ,2-b]pyridazin-2-amine
N-(6-(2-((l-(2-(dimethylamino)ethyl)-l H-pyrazol-3 -yl)amino)imidazo-[ l,2-b]pyridazin-3-yl)- 2-methyl-4-pyrimidinyl)acetamide
N-(2-methyl-6-(2-((l -methyl- 1 H-pyrazol-3 -yl)amino)imidazo[l,2-a]pyridin-3-yl)-4- pyrimidinyl)acetamide
3 -(2-methyl-6-((6-(4-morpholinyl)-3 -pyridazinyl)amino)-4-pyrimidinyl)-N-( 1 -methyl- 1 H- pyrazol-3-yl)imidazo[l,2-b]pyridazin-2-amine
N-(3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)-lH-pyrazolo[3,4- b]pyridin-3 -amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(3 -(4-methyl- 1 -piperazinyl)-phenyl)imidazo [ 1 ,2- b]pyridazin-2-amine
N-(3-((3-(6-(acetylamino)-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)- phenyl)acetamide 3 -(6-amino-2-methyl-4-pyrimidinyl)-6-( 1 -(2-methoxyethyl)- 1 H-pyrazol-4-yl)-N-( 1 -methyl- 1 H- pyrazol-3 -yl)imidazo [ 1 ,2-a]pyridin-2-amine
1 -(4-((3-(4-amino-6-methyl- 1 ,3,5-triazin-2-yl)imidazo[ 1 ,2-a]pyridin-2-yl)amino)phenyl)-3- methylurea
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-y I)-N- 1 H-pyrazol-3 -yl-6-(3 -pyridinyl)imidazo [ 1 ,2- a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N~6 — phenyl-N~2 — 1 H-pyrazol-3 -ylimidazo[ 1,2- a]pyridine-2,6-diamine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N~6 — (4-methoxyphenyl)-N~2 — lH-pyrazol-3- ylimidazo[l,2-a]pyridine-2,6-diamine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-7-methoxy-N-lH-pyrazol-3-ylimidazo[l,2-a]pyridin-2- amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-lH-pyrazol-3-yl-6-(5-pyrimidinyl)imidazo[l,2- a]pyridin-2-amine
3 -(4-methyl-6-(methylamino)- 1 ,3 ,5 -triazin-2-yl)-N- 1 H-pyrazol-3 -yl-6-(5 -pyrimidinyl)- imidazo[ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(3-fluorophenyl)-N-lH-pyrazol-3-ylimidazo[l,2- a]pyridin-2-amine
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-6- yl)-N-methylbenzamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N~2 — 1 H-pyrazol-3 -yl-N~6 — 3-pyridinylimidazo[l,2- a]pyridine-2,6-diamine
N-(4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin- 6-yl)phenyl)acetamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N~6 — (3-methoxyphenyl)-N~2 — lH-pyrazol-3- ylimidazo[l,2-a]pyridine-2,6-diamine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N~2 — 1 H-pyrazol-3 -yl-N~6 — 4-pyridinylimidazo[l,2- a]pyridine-2,6-diamine
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-N~2 — 1 H-pyrazol-3 -yl-N~6 — 5 -pyrimidinylimidazo- [ 1 ,2-a]pyridine-2,6-diamine
N-(4-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2- a]pyridin-6-yl)amino)phenyl)acetamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(2-methyl-3-pyridinyl)-N-lH-pyrazol-3- ylimidazo[l ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N~6— (2-methyl-4-pyridinyl)-N~2~- lH-pyrazol-3- ylimidazo[l,2-a]pyridine-2,6-diamine
N-methyl-4-(3-(4-methyl-6-(methylamino)-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)- imidazo[ 1 ,2-a]pyridin-6-yl)benzamide 3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(4-methyl-3-pyridinyl)-N-lH-pyrazol-3-ylimidazo- [ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(2,6-dimethyl-4-pyridinyl)-N-lH-pyrazol-3-yl- imidazo[ 1 ,2-a]pyridin-2-amine
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-6- yl)-N-( 1 -methylethyl)benzamide
6-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-6- yl)-3 ,4-dihydro- 1 (2H)-isoquinolinone
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-6- yl)-N-ethylbenzamide
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-6- yl)-3-methylbenzoic acid
3 -(2-methyl-6-(methylamino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -yl-6-(5 -pyrimidinyl)imidazo- [ 1 ,2-b]pyridazin-2-amine
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-y l)-6-(4-methyl-5 -pyrimidinyl)-N- 1 H-pyrazol-3 - ylimidazo[l ,2-a]pyridin-2-amine
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-6- yl)-N,3-dimethylbenzamide
5-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-6- yl)-N-methyl-2-pyridinecarboxamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-7-ol
tert-butyl 4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2- a]pyridin-7-yl)-3 ,6-dihydro- 1 (2H)-pyridinecarboxylate
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-lH-pyrazol-3-yl-7-(l,2,3,6-tetrahydro-4-pyridinyl)- imidazo[ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-7-(2-methoxyethoxy)-N-lH-pyrazol-3-ylimidazo[l,2- a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-lH-pyrazol-3-yl-6-(lH-pyrrolo[2,3-b]pyridin-5- yl)imidazo[l,2-a]pyridin-2-amine
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)-imidazo[l,2-a]pyridin-7- yl)-N-methylbenzamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-lH-pyrazol-3-yl-7-(2-(l-pyrrolidinyl)ethoxy)- imidazo[ 1 ,2-a]pyridin-2-amine
4-(3-(6-amino-2-methyl-4-pyrimidinyl)-2-(l H-pyrazol-3 -ylamino)-imidazo[l,2-b]pyridazin-6- yl)-N,N-dimethyl- 1 -piperazinecarboxamide
3-(6-amino-2-methyl-4-pyrimidinyl)-N-2-pyridinylimidazo[l,2-b]pyridazin-2-amine 3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -ethyl- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2-b]pyridazin-2- amine
N-(3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)-3 -quinolinamine
N-(3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)-3 -isoquinolinamine
N-(3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)-2-quinolinamine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -methyl- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2-b]pyridazin-2- amine
3-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-N-methyl-lH- pyrazole- 1 -carboxamide
5-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-N-methyl-lH- pyrazole- 1 -carboxamide
3-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-N-(2- hydroxy ethyl)- 1 H-pyrazole- 1 -carboxamide
5-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-N-(2- hydroxy ethyl)- 1 H-pyrazole- 1 -carboxamide
3-(4-amino-6-methyl- 1 ,3 ,5-triazin-2-yl)-N-( 1 -(4-morpholinylcarbonyl)- 1 H-pyrazol-3-yl)- imidazo[ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl- 1 ,3 ,5-triazin-2-yl)-N-( 1 -(4-morpholinylcarbonyl)- 1 H-pyrazol-5-yl)- imidazo[ 1 ,2-a]pyridin-2-amine
5-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-N-3-pyridinyl- 1 H-pyrazole- 1 -carboxamide
4-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-N-methyl-lH- pyrazole- 1 -carboxamide
3-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-N-phenyl-lH- pyrazole- 1 -carboxamide
5-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-N-phenyl-lH- pyrazole- 1 -carboxamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-lH-pyrazol-4-ylimidazo[l,2-a]pyridin-2-amine
methyl 3-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-lH- pyrazole- 1 -carboxylate methyl 5-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-lH- pyrazole- 1 -carboxylate phenyl 4-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-lH- pyrazole- 1 -carboxylate 3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-phenyl- 1 H- pyrazole- 1 -carboxamide
5 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-phenyl- 1 H- pyrazole- 1 -carboxamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-methyl- 1 H- pyrazole- 1 -carboxamide
5 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-methyl- 1 H- pyrazole- 1 -carboxamide
4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)-N-phenyl-lH- pyrazole- 1 -carboxamide
4-((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-methyl- 1 H- pyrazole- 1 -carboxamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(2-pyridinyl)- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2- a]pyridin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(methylsulfonyl)- 1 H-pyrazol-3 -yl)imidazo [1,2- a]pyridin-2-amine
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(l-(l,3-benzoxazol-2-yl)-lH-pyrazol-5-yl)imidazo[l,2- a]pyridin-2-amine phenyl 3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)- 1 H- pyrazole- 1 -carboxylate
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(l -(I H-benzimidazol-2-yl)-l H-pyrazol-3 -yl)imidazo- [ 1 ,2-a]pyridin-2-amine
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-a]pyridin-2-yl)amino)-N-(l -methylethyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)-N-benzyl-lH- pyrazole- 1 -carboxamide methyl 4-(((3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)-lH- pyrazol- 1 -yl)carbonyl)amino)benzoate
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-ethyl- 1 H- pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)-N-(2- methoxyphenyl)- 1 H-pyrazole- 1 -carboxamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-4-ylimidazo [ 1 ,2-a]pyridin-2-amine
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-(2-methyl- phenyl)- 1 H-pyrazole- 1 -carboxamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-(4-methoxy- phenyl)- 1 H-pyrazole- 1 -carboxamide 3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N-phenyl- 1 H- pyrazole-1-carboxamide, trifluoroacetic acid
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N-methyl- 1 H- pyrazole- 1 -carboxamide
5 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N-methyl- 1 H- pyrazole- 1 -carboxamide methyl 4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-a]pyridin-2-yl)amino)- IH- pyrazole- 1 -carboxylate
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(l-(l,3-benzoxazol-2-yl)-lH-pyrazol-3-yl)imidazo[l,2- b]pyridazin-2-amine methyl 5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-lH- pyrazole- 1 -carboxylate
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2- methylphenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2- fluorophenyl)- 1 H-pyrazole- 1 -carboxamide
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(l-(l,3-benzoxazol-2-yl)-lH-pyrazol-5-yl)imidazo[l,2- b]pyridazin-2-amine
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(4- cyanophenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2- (trifluoromethyl)phenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(3- methylphenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2,5- difluorophenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(3,5- difluorophenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-b]pyridazin-2-yl)amino)-N-(l - methylethyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(4- methylphenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(3,5- dichlorophenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2-chloro-5- (trifluoromethyl)phenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2,5- dichlorophenyl)- 1 H-pyrazole- 1 -carboxamide 3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2,3- dichlorophenyl)- 1 H-pyrazole- 1 -carboxamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N-(2-chloro-4- (trifluoromethyl)phenyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-(acetylamino)-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2- methylphenyl)- 1 H-pyrazole- 1 -carboxamide
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(l-(lH-benzimidazol-2-yl)-lH-pyrazol-3-yl)imidaz- [ 1 ,2-b]pyridazin-2-amine
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(l-(lH-imidazol-2-yl)-lH-pyrazol-3-yl)imidazo[l,2- b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(methylsulfonyl)- 1 H-pyrazol-3 -yl)imidazo [1,2- b]pyridazin-2-amine
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-l H-pyrazole- 1- carboxamide
5 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-l H-pyrazole- 1- carboxamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -( 1 -azetidinylcarbonyl)- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2- b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -( 1 -azetidinylcarbonyl)- 1 H-pyrazol-5 -yl)imidazo [ 1 ,2- b]pyridazin-2-amine
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2-(methyl- amino)-ethyl)- 1 H-pyrazole- 1 -carboxamide
5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2- (methylamino)ethyl)- 1 H-pyrazole- 1 -carboxamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N,N-dimethyl- 1 H-pyrazole- 1 -carboxamide
5 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N,N-dimethyl- 1 H-pyrazole- 1 -carboxamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -( 1 -piperazinylcarbonyl)- 1 H-pyrazol-3 -yl)imidazo- [ 1 ,2-b]pyridazin-2-amine
5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2- hydroxy ethyl)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2-methoxy- ethyl)- 1 H-pyrazole- 1 -carboxamide
5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2-methoxy- ethyl)- 1 H-pyrazole- 1 -carboxamide
5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-N-(2-methyl- propyl)- 1 H-pyrazole- 1 -carboxamide 3 -(6-amino-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -yl-6-(5 -pyrimidinyl)imidazo [ 1 ,2-b] - pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -( 1 -phenylethyl)- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2-b]- pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(l -benzyl- 1 H-pyrazol-3 -yl)imidazo[l,2-b]pyridazin-2- amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(3 -pyridinylmethyl)- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2- b]pyridazin-2-amine
7-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)-2H-l,4-benzoxazin- 3(4H)-one l-(5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-2-pyridinyl)-3- azetidinol
2-(l-(5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-2- pyridinyl)-3-azetidinyl)-2-propanol
N-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)-7-quinolinamine
N-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)-6-quinolinamine
N-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)-7-isoquinolinamine
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(7-quinolinylamino)-imidazo[l,2-a]pyridin-6-yl)- N,N-dimethyl- 1 -piperazinecarboxamide
N-(3-(4-amino-6-methyl- 1 ,3,5-triazin-2-yl)-6-(4-(methylsulfonyl)- 1 -piperazinyl)imidazo[ 1 ,2- a]pyridin-2-yl)-7-quinolinamine
N-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(l-piperazinyl)-imidazo[l,2-a]pyridin-2-yl)-7- quinolinamine
4-(3-(4-(acetylamino)-6-methyl-l,3,5-triazin-2-yl)-2-((6-methoxy-3-pyridinyl)amino)- imidazo[ 1 ,2-a]pyridin-6-yl)-N,N-dimethyl- 1 -piperazinecarboxamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(6-ethoxy-3-pyridinyl)-imidazo[l,2-a]pyridin-2- amine
N-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)-lH-indazol-3-amine
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-((5-fluoro-6-methoxy-3- pyridinyl)amino)imidazo[ 1 ,2-a]pyridin-6-yl)-N,N-dimethyl- 1 -piperazinecarboxamide
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-((6-methoxy-3-pyridinyl)amino)imidazo[l,2- a]pyridin-6-yl)-N-methyl- 1 -piperazinecarboxamide
6-(4-acetyl-l-piperazinyl)-3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(6-methoxy-3- pyridinyl)imidazo[ 1 ,2-a]pyridin-2-amine (5-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-5-ylamino)imidazo[l,2-a]pyridin-6- yl)-2-methoxyphenyl)methanol
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-y I)-N- 1 H-pyrazol-5 -yl-6-(4-pyridinyl)imidazo [ 1 ,2- a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(3,6-dihydro-2H-pyran-4-yl)-N-lH-pyrazol-5- ylimidazo [ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(2-methyl-4-pyridinyl)-N-lH-pyrazol-5- ylimidazo [ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l, 3, 5-triazin-2-yl)-6-(l -methyl- lH-imidazol-5-yl)-N-l H-pyrazol-5 - ylimidazo [ 1 ,2-a]pyridin-2-amine
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-5-ylamino)-midazo[l,2-a]pyridin-6- yl)- 1 ,2-butanediol
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-6-( 1 -(I -methylethyl)- 1 H-pyrazol-4-yl)-N- 1 H-pyrazol- 5 -ylimidazo [ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-5-ylamino)-imidazo[l,2-a]pyridine-6- carboxamide
N-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-5-ylamino)imidazo[l,2-a]pyridin-6- yl)methyl)acetamide
3-(2-methyl-6-((2,2,2-trifluoroethyl)amino)-4-pyrimidinyl)-N-lH-pyrazol-5-ylimidazo[l,2- b]pyridazin-2-amine
3 -(2-methyl-6-(2-pyrazinylamino)-4-pyrimidinyl)-N-( 1 -methyl- 1 H-pyrazol-3 -yl)imidazo [1,2- b]pyridazin-2-amine
3 -(2-methyl-6-(3 -pyridinylamino)-4-pyrimidinyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2-b]pyridazin- 2-amine
2-cyano-N-(2-methyl-6-(2-((l -methyl- 1 H-pyrazol-3 -yl)amino)-imidazo[l,2-b]pyridazin-3-yl)- 4-pyrimidinyl)acetamide
N-(2-methyl-6-(2-((l -methyl- lH-pyrazol-3-yl)amino)imidazo[l,2-b]pyridazin-3-yl)-4- pyrimidinyl)-l,3-oxazole-5-carboxamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -methyl- 1 H-pyrazol-3 -yl)-6-(5 -pyrimidiny l)imidazo- [ 1 ,2-a]pyridin-2-amine
N-(4-(3-(6-amino-2-methyl-4-pyrimidinyl)-2-((l-methyl-lH-pyrazol-3-yl)amino)imidazo[l,2- a]pyridin-6-yl)phenyl)acetamide
3-(4-amino-6-methyl-l, 3, 5 -triazin-2-y I)-N-(I -methyl- 1 H-pyrazol-3 -yl)-6-(7-quinolinyl)- imidazo-[ 1 ,2-a]pyridin-2-amine
N-(4-(3-(6-amino-2-methyl-4-pyrimidinyl)-2-((l -methyl- 1 H-pyrazol-3 -yl)amino)imidazo[ 1,2- b]pyridazin-6-yl)benzyl)acetamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(l-methyl-lH-pyrazol-3-yl)-6-(5-pyrimidinyl)- imidazo[ 1 ,2-a]pyridin-2-amine 4-(3 -(6-amino-2-methyl-4-pyrimidinyl)-2-(( 1 -methyl- 1 H-pyrazol-3 -yl)amino)imidazo [ 1 ,2- b]pyridazin-6-yl)-N-methylbenzamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(6-(2-methoxyethoxy)-4-pyrimidinyl)- 1 H-pyrazol-3 - yl)imidazo[ 1 ,2-b]pyridazin-2-amine
3 -(6-(3-isoxazolylamino)-2-methyl-4-pyrimidinyl)-N-l H-pyrazol-3 -ylimidazo[l,2-b]pyridazin- 2-amine
2-cyano-N-(2-methyl-6-(2-((l -methyl- lH-pyrazo 1-3 -yl)amino)-6-(5-pyrimidinyl)imidazo[ 1,2- a]pyridin-3-yl)-4-pyrimidinyl)acetamide
3 -(6-((I -(2 -methoxy ethyl)- lH-pyrazol-4-yl)amino)-2-methyl-4-pyrimidinyl)-N-l H-pyrazol-3 - ylimidazo[l,2-b]pyridazin-2-amine
3-(2-methyl-6-((l-((3R)-tetrahydro-3-furanyl)-lH-pyrazol-4-yl)amino)-4-pyrimidinyl)-N-lH- pyrazol-3-ylimidazo[l,2-b]pyridazin-2-amine, 3-(2-methyl-6-((l-((3S)-tetrahydro-3-furanyl)- lH-pyrazol-4-yl)amino)-4-pyrimidinyl)-N-l H-pyrazol-3 -ylimidazo[l,2-b]pyridazin-2-amine
3-(6-(imidazo[l,2-a]pyridin-2-ylamino)-2-methyl-4-pyrimidinyl)-N-lH-pyrazol-3- ylimidazo[l,2-b]pyridazin-2-amine
3 -(6-(furo [3, 2-b]pyridin-6-ylamino)-2-methyl-4-pyrimidinyl)-N-l H-pyrazol-3 -ylimidazo[ 1,2- b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -yl-6-(5 -pyrimidinyl)imidazo [1,2- a]pyridin-2-amine
3-(2-methyl-6-((l-((3R)-tetrahydro-3-furanyl)-lH-pyrazol-4-yl)amino)-4-pyrimidinyl)-N-lH- pyrazol-3-ylimidazo[l,2-a]pyridin-2-amine, 3-(2-methyl-6-((l-((3S)-tetrahydro-3-furanyl)-lH- pyrazol-4-yl)amino)-4-pyrimidinyl)-N-l H-pyrazol-3 -ylimidazo[l,2-a]pyridin-2-amine
3 -(6-((6-(2 -methoxy ethoxy)-4-pyrimidinyl)amino)-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 - ylimidazo[l,2-b]pyridazin-2-amine l-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)phenyl)-3- cyclopropylurea
N-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)- acetamide
N-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-2- phenylacetamide
N-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)- benzamide
N-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-4- chlorobenzenesulfonamide
N-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)- methanesulfonamide l-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-3- phenylurea
1 -(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-a]pyridin-2-yl)amino)phenyl)-3-(4- fluorophenyl)urea 1 -(3 -((3 -(6-amino-2-methy l-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)phenyl)-3 - benzylurea
1 -(3 -((3 -(6-amino-2-methy l-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)phenyl)-3 - methylurea
1 -(3 -((3 -(6-amino-2-methy l-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)phenyl)-3 - ethylurea
4-((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-ethylbenzamide
5 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-ethyl-2- pyridinecarboxamide
N-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)phenyl)- acetamide
N-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)phenyl)- propanamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-methyl- benzamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-ethylbenzamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-a]pyridin-2-yl)amino)-N-(l -methylethyl)- benzamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N-methyl- benzamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N-ethyl- benzamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N-phenyl- benzamide
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-N-( 1 -methyl- ethyl)benzamide
5 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-methyl-2- pyridinecarboxamide
5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-a]pyridin-2-yl)amino)-N-(l -methylethyl)- 2-pyridinecarboxamide
5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)-N-phenyl-2- pyridinecarboxamide
4-((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)benzamide
4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)-N-(2- (diethylamino)-ethyl)benzamide 4-((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-phenylbenzamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(4-(methylsulfonyl)phenyl)-N-lH-pyrazol-3- ylimidazo [ 1 ,2-a]pyridin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(3 -methylbutanoyl)- 1 H-pyrazol-3 -yl)imidazo [1,2- b]pyridazin-2-amine
1 -methylethyl 5 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)- 1 H-pyrazole- 1 -carboxylate
1 -methylethyl 3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)- 1 H-pyrazole- 1 -carboxylate
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(2-methoxy-4-pyridinyl)imidazo [ 1 ,2-b]pyridazin-2- amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(2-methylpropanoyl)- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2- b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(2-methylpropanoyl)- 1 H-pyrazol-5 -yl)imidazo [ 1 ,2- b]pyridazin-2-amine
4-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]pyridin-6- yl)-N-methylbenzenesulfonamide
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-y l)-6-(3 -(methylsulfonyl)phenyl)-N- 1 H-pyrazol-3 - ylimidazo [ 1 ,2-a]pyridin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(3 ,3 -dimethylbutanoyl)- 1 H-pyrazol-3 -ylimidazo [ 1 ,2-b]pyridazin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(l-methyl-lH-pyrazol-3-yl)-6-(4-(methylsulfonyl)- phenyl)imidazo[ 1 ,2-a]pyridin-2-amine cyclohexyl 5-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-lH- pyrazole- 1 -carboxylate cyclohexyl 3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)-lH- pyrazole- 1 -carboxylate
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(2-methoxy-3 -pyridinyl)imidazo [ 1 ,2-b]pyridazin-2- amine
3 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)-2( 1 H)-pyridinone
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-( 1 -(3 -methylbutyl)- 1 H-pyrazol-3 -yl)imidazo [ 1 ,2- b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(l -(2 -methoxy ethyl)- 1 H-pyrazol-3 -yl)imidazo[ 1,2- b]pyridazin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-6-(3 -(methylsulfonyl)phenyl)-N- 1 H-pyrazol-3 - ylimidazo [l,2-b]pyridazin-2-amine N-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)-lH-indazol-4-amine
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(6-methoxy-3-pyridinyl)-6-(4-morpholinyl)- imidazo[ 1 ,2-b]pyridazin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(6-methoxy-3-pyridinyl)-6-(4-morpholinyl)- imidazo[ 1 ,2-b]pyridazin-2-amine
3-(6-amino-2-methyl-4-pyrimidinyl)-N~6 — (2-methoxyethyl)-N~2 — (6-methoxy-3- pyridinyl)imidazo[ 1 ,2-b]pyridazine-2,6-diamine
3-(6-amino-2-methyl-4-pyrimidinyl)-N~2 — lH-indazol-4-yl-N~6 — (2-methoxyethyl)imidazo- [ 1 ,2-b]pyridazine-2,6-diamine
1 -(3 -(6-amino-2-methyl-4-pyrimidinyl)-2-((6-methoxy-3 -pyridinyl)amino)imidazo [ 1 ,2- b]pyridazin-6-yl)-4-piperidinol
4-(3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-2-((6-methoxy-3 -pyridinyl)amino)imidazo [ 1 ,2- a]pyridin-7-yl)-N,N-dimethyl- 1 -piperazinecarboxamide
4-(3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-2-((5 -fluoro-6-methoxy-3 -pyridinyl)amino)- imidazo[ 1 ,2-a]pyridin-7-yl)-N,N-dimethyl- 1 -piperazinecarboxamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(5-fluoro-6-methoxy-3-pyridinyl)-7-(4- (methylsulfonyl)- 1 -piperazinyl)imidazo[ 1 ,2-a]pyridin-2-amine
N-(5-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-2-chloro-3- pyridinyl)methanesulfonamide
N-(5-((3-(4-amino-6-methyl-l,3,5-triazin-2-yl)imidazo[l,2-a]pyridin-2-yl)amino)-2-methoxy- 3 -pyridiny l)methanesulfonamide
3-(6-amino-2-methyl-4-pyrimidinyl)-N-lH-indazol-6-ylimidazo[l,2-b]pyridazin-2-amine
3-(6-amino-2-methyl-4-pyrimidinyl)-N-lH-benzimidazol-6-ylimidazo[l,2-b]pyridazin-2-amine
N-(3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)-lH-benzimidazol-6-amine
3-(4-amino-6-methyl-l, 3, 5-triazin-2-yl)-N-(6-methoxy-3-pyridinyl)-7-(l -methyl- lH-pyrazol-4- yl)imidazo[l,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l, 3, 5-triazin-2-yl)-N-(6-methoxy-3-pyridinyl)-6-(l -methyl- lH-pyrazol-4- yl)imidazo[l,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(6-methoxy-3-pyridinyl)-6-(4-morpholinyl)- imidazo[ 1 ,2-a]pyridin-2-amine
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(6-methoxy-3-pyridinyl)-6-(4-morpholinyl)imidazo- [ 1 ,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-(6-methoxy-3-pyridinyl)-6-(3-pyridinyl)- imidazo[ 1 ,2-a]pyridin-2-amine 4-(3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-2-((6-methoxy-3 -pyridinyl)amino)imidazo [ 1 ,2- a]pyridin-6-yl)-N,N-dimethyl- 1 -piperazinecarboxamide
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-N-(6-methoxy-3 -pyridinyl)-6-(4-(methylsulfonyl)- 1 - piperazinyl)imidazo[ 1 ,2-a]pyridin-2-amine
2-(3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-2-((6-methoxy-3 -pyridinyl)-amino)imidazo [ 1 ,2- a]pyridin-6-yl)-2-propanol
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-N-(6-methoxy-3 -pyridinyl)-6-( 1 -piperazinyl)imidazo- [ 1 ,2-a]pyridin-2-amine
1 -(3-(4-amino-6-methyl- 1 ,3,5-triazin-2-yl)-2-(7-quinolinylamino)-imidazo[ 1 ,2-a]pyridin-6- yl)ethanone
2-(3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-2-(7-quinolinylamino)-imidazo[l,2-a]pyridin-6-yl)- 2-propanol
1 -(3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-2-((5 -fluoro-6-methoxy-3 -pyridinyl)amino)- imidazo[ 1 ,2-a]pyridin-6-yl)ethanone
2-(3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-yl)-2-((5 -fluoro-6-methoxy-3 -pyridinyl)amino)- imidazo[ 1 ,2-a]pyridin-6-yl)-2-propanol
4-(3 -(6-amino-2-methyl-4-pyrimidinyl)-2-((5 -fluoro-6-methoxy-3 -pyridinyl)amino)- imidazo [ 1 ,2-b]pyridazin-6-yl)-N,N-dimethyl- 1 -piperazinecarboxamide l-(3-(6-amino-2-methyl-4-pyrimidinyl)-2-((5-fluoro-6-methoxy-3-pyridinyl)amino)imidazo- [ 1 ,2-a]pyridin-6-yl)ethanone
2-(3-(6-amino-2-methyl-4-pyrimidinyl)-2-((5-fluoro-6-methoxy-3-pyridinyl)amino)imidazo- [ 1 ,2-a]pyridin-6-yl)-2-propanol
4-(3-(6-amino-2-methyl-4-pyrimidinyl)-2-((5-fluoro-6-methoxy-3-pyridinyl)amino)- imidazo [ 1 ,2-a]pyridin-6-yl)-N,N-dimethyl- 1 -piperazinecarboxamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-N-lH-pyrazol-3-yl-6-(trifluoromethyl)imidazo[l,2- a]pyridin-2-amine
3 -(4-amino-6-methyl- 1 ,3 ,5 -triazin-2-y I)-N- 1 H-pyrazol-3 -yl-7-(trifluoromethy l)imidazo [ 1 ,2-a] - pyridin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -yl-7-(trifluoromethyl)imidazo [ 1 ,2-a] - pyridin-2-amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -yl-6-(trifluoromethyl)imidazo [ 1 ,2-a] - pyridin-2-amine l-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)phenyl)-3- cyclopropylurea l-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)phenyl)-3- ethylurea
N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-b]pyridazin-2-yl)amino)phenyl)- 1 - piperidinecarboxamide N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-b]pyridazin-2-yl)amino)phenyl)- 1 - pyrrolidinecarboxamide
N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-b]pyridazin-2-yl)amino)phenyl)- 1 - azetidinecarboxamide
N-(4-((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)phenyl)-4- methyl- 1 -piperazinecarboxamide
1 -(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-b]pyridazin-2-yl)amino)phenyl)-3-(2- methoxyethyl)urea
N-(4-((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)phenyl)-4- morpholinecarboxamide
4-acetyl-N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)- phenyl)- 1 -piperazinecarboxamide
3-(6-amino-2-methyl-4-pyrimidinyl)-N- 1 ,3-thiazol-4-ylimidazo[ 1 ,2-b]pyridazin-2-amine
N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-4- morpholinecarboxamide
4-acetyl-N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)- 1 -piperazinecarboxamide
N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-2- methyl-4-morpholinecarboxamide
3-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-l- methyl- 1 -( 1 -methylethyl)urea
N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-3- methyl-4-morpholinecarboxamide l-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-3- cyclopropylurea ethyl 2-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]-pyridazin-2-yl)amino)-lH- imidazole-4-carboxylate
3 -(2-methyl-4-pyrimidinyl)-6-(3 -(methylsulfonyl)phenyl)-N- 1 H-pyrazol-3 -ylimidazo [ 1 ,2- b]pyridazin-2-amine
N-(3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]-pyridazin-2-yl)-3H-imidazo[4,5- b]pyridin-5 -amine
N-(2-methyl-6-(2-(( 1 -methyl- 1 H-pyrazol-3-yl)amino)-6-(3 - (methylsulfonyl)phenyl)imidazo [ 1 ,2-a]pyridin-3 -yl)-4-pyrimidinyl)acetamide
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(3-(ethylsulfonyl)-phenyl)-N-lH-pyrazol-3- ylimidazo[l ,2-a]pyridin-2-amine
3-(6-amino-2-methyl-4-pyrimidinyl)-6-(3,6-dihydro-2H-pyran-4-yl)-N-lH-pyrazol-3- ylimidazo[l,2-b]pyridazin-2-amine N-(5 -((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b] -pyridazin-2-yl)amino)-3 - pyridinyl)acetamide
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(2-methyl-l,3-thiazol-4-yl)imidazo[l,2-b]pyridazin-2- amine
3 -(6-amino-2-methyl-4-pyrimidinyl)-N- 1 ,3 -thiazol-5 -ylimidazo- [ 1 ,2-b]pyridazin-2-amine
1 -(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[ 1 ,2-a]-pyridin-2-yl)amino)phenyl)-3-(2- methylpropyl)urea
N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]-pyridin-2-yl)amino)phenyl)-4,4- difluoro- 1 -piperidinecarboxamide
(2S)-N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-a]pyridin-2-yl)amino)phenyl)-2- methyl- 1 -pyrrolidine-carboxamide
N-(2-methyl-6-(2-((l -methyl- lH-pyrazol-3-yl)amino)-6-(2-methyl-3-pyridinyl)imidazo[l, 2- a]pyridin-3-yl)-4-pyrimidinyl)acetamide
N-(6-(6-( 1 -(2-methoxy ethyl)- 1 H-pyrazol-4-yl)-2-(( 1 -methyl- 1 H-pyrazol-3 - yl)amino)imidazo[l,2-a]pyridin-3-yl)-2-methyl-4-pyrimidinyl)acetamide
N-(5-((3-(6-(acetylamino)-2-methyl-4-pyrimidinyl)imidazo-[l,2-b]pyridazin-2-yl)amino)-3- pyridinyl)acetamide
N-(2-methyl-6-(2-(lH-pyrazol-3-ylamino)-6-(5-pyrimidinyl)-imidazo[l,2-a]pyridin-3-yl)-4- pyrimidinyl)acetamide
8-fluoro-3-(2-methyl-4-pyrimidinyl)-N-lH-pyrazol-3-ylimidazo-[l,2-a]pyridin-2-amine
3-(4-amino-6-methyl-l,3,5-triazin-2-yl)-6-(4-(ethylsulfonyl)-phenyl)-N-lH-pyrazol-3- ylimidazo[l ,2-a]pyridin-2-amine
3-(2,6-dimethyl-4-pyrimidinyl)-6-(3-(methylsulfonyl)phenyl)-N-lH-pyrazol-3-ylimidazo[l,2- b]pyridazin-2-amine
N-(6-(8-fluoro-2-(lH-pyrazol-3-ylamino)imidazo[l,2-a]-pyridin-3-yl)-2-methyl-4- pyrimidinyl)acetamide
N-(3-(6-amino-2-methyl-4-pyrimidinyl)-6-(3-(methylsulfonyl)-phenyl)imidazo[l,2- b]pyridazin-2-yl)- 1 ,3-benzenediamine
3 -((8-fluoro-3 -(2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-( 1 -methylethyl)- 1 H-pyrazole- 1 -carboxamide
5-((3-(6-(acetylamino)-2-methyl-4-pyrimidinyl)-8-fluoro-imidazo[l,2-a]pyridin-2-yl)amino)- 1 H-pyrazole- 1 -carboxamide
3-((3-(6-(acetylamino)-2-methyl-4-pyrimidinyl)-8-fluoroimidazo[l,2-a]pyridin-2-yl)amino)- 1 H-pyrazole- 1 -carboxamide
N-(2-methyl-6-(2-(l H-pyrazol-3 -ylamino)-6-(5-pyrimidinyl)-imidazo[l,2-b]pyridazin-3-yl)-4- pyrimidinyl)acetamide 5 -((8-fluoro-3 -(2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-a]pyridin-2-yl)amino)-N-( 1 -methylethyl)- 1 H-pyrazole- 1 -carboxamide
(2R)-N-(4-((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)phenyl)- 2-methyl-4-morpholine-carboxamide
3 -(4-((3 -(6-amino-2-methy l-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)phenyl)- 1 - methyl- 1 -( 1 -methylethyl)urea
(3R)-N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)phenyl)- 3-methyl-4-morpholine-carboxamide
(2S)-N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]pyridazin-2-yl)amino)phenyl)- 2-methyl- 1 -pyrrolidine-carboxamide
(3 S)-N-(4-((3 -(6-amino-2-methyl-4-pyrimidinyl)imidazo [ 1 ,2-b]pyridazin-2-yl)amino)phenyl)- 3 -methyl- 1 -pyrrolidine-carboxamide
N-(3-((3-(6-amino-2-methyl-4-pyrimidinyl)-6-(3-(methyl-sulfonyl)-phenyl)imidazo[l,2- b]pyridazin-2-yl)amino)phenyl)acetamide
N-(6-(2-((4-((cyclopropylcarbamoyl)amino)phenyl)amino)-imidazo[l,2-b]pyridazin-3-yl)-2- methyl-4-pyrimidinyl)-acetamide
3-(6-amino-2-methyl-4-pyrimidinyl)-6-(5,6-dihydro-2H-pyran-3-yl)-N-lH-pyrazol-3- ylimidazo[l,2-b]pyridazin-2-amine
2-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]-pyridazin-2-yl)amino)phenyl)-N- cyclopropylacetamide
3-((3-(6-amino-2-methyl-4-pyrimidinyl)imidazo[l,2-b]-pyridazin-2-yl)amino)-N-(5-fluoro-2- (trifluoromethyl)phenyl)- 1 H-pyrazole- 1 -carboxamide
6-fluoro-8-methoxy-3-(2-methyl-4-pyrimidinyl)-N-lH-pyrazol-3-ylimidazo[l,2-a]pyridin-2- amine
N-(3-(6-amino-2-methyl-4-pyrimidinyl)-6-(3-(methylsulfonyl)-phenyl)imidazo[l,2- b]pyridazin-2-yl)- 1 ,4-benzenediamine
N-(4-((3-(6-amino-2-methyl-4-pyrimidinyl)-6-(3-(methyl-sulfonyl)-phenyl)imidazo[l,2- b]pyridazin-2-yl)amino)phenyl)acetamide
3 -(6-amino-2-methyl-4-pyrimidinyl)-N-(l -methyl- lH-pyrazol-3 -yl)-6-(3 - (methylsulfonyl)phenyl)imidazo[ 1 ,2-b]pyridazin-2-amine
3-(6-amino-2-methyl-4-pyrimidinyl)-N-(l-(4,5-dihydro-l,3-oxazol-2-yl)-lH-pyrazol-3- yl)imidazo[ 1 ,2-b]pyridazin-2-amine
6-fluoro-3 -(2-methyl-4-pyrimidinyl)-N- 1 H-pyrazol-3 -yl-8-(3 -pyridinyloxy)imidazo [ 1 ,2- a]pyridin-2-amine or a pharmaceutically acceptable salt thereof.
14. A pharmaceutical composition comprising a compound of any of the Claims 1-13 or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable excipient.
15. A method of treating melanoma, ovarian cancer, cervical cancer, breast cancer, colon cancer, rectal cancer, endometrial cancer, pancreatic cancer, lung cancer, stomach cancer, glioblastoma, liver cancer, prostate cancer, acute lyelogeous leukemia, chronic lyelogenous leukemia, or thyroid cancer, the method comprising administering to a patient in need thereof a therapeutically effective amount of a compound of any of the Claims 1-13, or a pharmaceutically acceptable salt thereof.
16. Compound of any of the Claim 1-13 for use in therapy.
17. The compound of Clain 16 wherein the therapy is cancer.
PCT/US2010/034596 2009-05-13 2010-05-12 Heteroaryl compounds as pikk inhibitors WO2010132598A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
EP10719514.1A EP2430013B1 (en) 2009-05-13 2010-05-12 Heteroaryl compounds as pikk inhibitors
AU2010249040A AU2010249040B2 (en) 2009-05-13 2010-05-12 Heteroaryl compounds as PIKK inhibitors
US13/319,683 US20120190666A1 (en) 2009-05-13 2010-05-12 Heteroaryl Compounds as PIKK Inhibitors
JP2012510985A JP2012526836A (en) 2009-05-13 2010-05-12 Heteroaryl compounds as PIKK inhibitors
CA2760778A CA2760778A1 (en) 2009-05-13 2010-05-12 Heteroaryl compounds as pikk inhibitors
MX2011012037A MX2011012037A (en) 2009-05-13 2010-05-12 Heteroaryl compounds as pikk inhibitors.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US17782509P 2009-05-13 2009-05-13
US61/177,825 2009-05-13

Publications (1)

Publication Number Publication Date
WO2010132598A1 true WO2010132598A1 (en) 2010-11-18

Family

ID=42470855

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/034596 WO2010132598A1 (en) 2009-05-13 2010-05-12 Heteroaryl compounds as pikk inhibitors

Country Status (7)

Country Link
US (1) US20120190666A1 (en)
EP (1) EP2430013B1 (en)
JP (1) JP2012526836A (en)
AU (1) AU2010249040B2 (en)
CA (1) CA2760778A1 (en)
MX (1) MX2011012037A (en)
WO (1) WO2010132598A1 (en)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011153509A1 (en) 2010-06-04 2011-12-08 Amgen Inc. Piperidinone derivatives as mdm2 inhibitors for the treatment of cancer
WO2013049250A1 (en) 2011-09-27 2013-04-04 Amgen Inc. Heterocyclic compounds as mdm2 inhibitors for the treatment of cancer
US8415376B2 (en) 2008-05-30 2013-04-09 Amgen Inc. Inhibitors of PI3 kinase
WO2013096630A1 (en) * 2011-12-23 2013-06-27 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
JP2014506601A (en) * 2011-02-23 2014-03-17 インテリカイン, エルエルシー Heterocyclic compounds and uses thereof
US8765746B2 (en) 2010-10-13 2014-07-01 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
US8796271B2 (en) 2010-08-11 2014-08-05 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
US8796314B2 (en) 2009-01-30 2014-08-05 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
WO2014130470A1 (en) 2013-02-19 2014-08-28 Amgen Inc. Cis-morpholinone and other compounds as mdm2 inhibitors for the treatment of cancer
WO2014151863A1 (en) 2013-03-14 2014-09-25 Amgen Inc. Heteroaryl acid morpholinone compounds as mdm2 inhibitors for the treatment of cancer
US8859768B2 (en) 2010-08-11 2014-10-14 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
US9029411B2 (en) 2008-01-25 2015-05-12 Millennium Pharmaceuticals, Inc. Thiophenes and uses thereof
US9062038B2 (en) 2010-08-11 2015-06-23 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
US9090601B2 (en) 2009-01-30 2015-07-28 Millennium Pharmaceuticals, Inc. Thiazole derivatives
JP2015522051A (en) * 2012-07-13 2015-08-03 ユーシービー バイオファルマ エスピーアールエル Imidazopyridine derivatives as modulators of TNF activity
US9139589B2 (en) 2009-01-30 2015-09-22 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
KR101875720B1 (en) * 2011-01-10 2018-07-09 인피니티 파마슈티칼스, 인코포레이티드 Processes for preparing isoquinolinones and solid forms of isoquinolinones
US10214519B2 (en) 2016-09-23 2019-02-26 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10227350B2 (en) 2016-09-23 2019-03-12 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10227342B2 (en) 2014-06-19 2019-03-12 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
US10479770B2 (en) 2016-09-23 2019-11-19 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
WO2020106647A2 (en) 2018-11-19 2020-05-28 Amgen Inc. Combination therapy including a krasg12c inhibitor and one or more additional pharmaceutically active agents for the treatment of cancers
EP3738593A1 (en) 2019-05-14 2020-11-18 Amgen, Inc Dosing of kras inhibitor for treatment of cancers
EP3805232A1 (en) 2013-06-10 2021-04-14 Amgen Inc. Crystalline synthetic intermediate useful in processes for making a mdm2 inhibitor
WO2021076655A1 (en) 2019-10-15 2021-04-22 Amgen Inc. Combination therapy of kras inhibitor and shp2 inhibitor for treatment of cancers
WO2021126816A1 (en) 2019-12-16 2021-06-24 Amgen Inc. Dosing regimen of a kras g12c inhibitor
KR20210126065A (en) * 2019-05-13 2021-10-19 광동 어글레이어 압토일렉트라닉 머티어리얼즈 컴퍼니 리미티드 Imidazo nitrogen heterocyclic compound and its application
CN114174293A (en) * 2019-07-24 2022-03-11 默克专利股份公司 4- (imidazo [1,2-a ] pyridin-3-yl) -pyrimidine derivatives
EP4039256A1 (en) 2013-11-11 2022-08-10 Amgen Inc. Combination therapy including an mdm2 inhibitor and dasatinib or nilotinib for the treatment of chronic myeloid leukemia
WO2022197755A1 (en) * 2021-03-17 2022-09-22 Shire Human Genetic Therapies, Inc. Imidazopyridinyl inhibitors of plasma kallikrein
WO2023009475A1 (en) * 2021-07-26 2023-02-02 President And Fellows Of Harvard College Rock2 inhibitors and uses thereof
US11766436B2 (en) 2018-05-04 2023-09-26 Amgen Inc. KRAS G12C inhibitors and methods of using the same
US11787796B2 (en) 2019-09-18 2023-10-17 Takeda Pharmaceutical Company Limited Plasma Kallikrein inhibitors and uses thereof
US11827635B2 (en) 2019-05-21 2023-11-28 Amgen Inc. Solid state forms
EP4061851A4 (en) * 2019-11-21 2023-12-20 BeiGene, Ltd. Methods of cancer treatment using anti-ox40 antibodies in combination with pi3 kinase delta inhibitors
US11905281B2 (en) 2017-05-22 2024-02-20 Amgen Inc. KRAS G12C inhibitors and methods of using the same

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA114194C2 (en) * 2012-03-15 2017-05-10 Сігнал Фармасьютікалз, Елелсі Treatment of cancer with tor kinase inhibitors
CN105164114B (en) * 2013-05-01 2018-03-23 豪夫迈·罗氏有限公司 The miazines and their purposes of the Heterocyclylalkyl substitution of C connections
US11352328B2 (en) 2016-07-12 2022-06-07 Arisan Therapeutics Inc. Heterocyclic compounds for the treatment of arenavirus
CA3226908A1 (en) * 2021-07-30 2023-02-02 Scott Bryan HOYT Multi-cyclic irak and flt3 inhibiting compounds and uses thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007027842A1 (en) * 2005-08-31 2007-03-08 Bayer Healthcare Llc Anilinopyrazole derivatives useful for the treatment of diabetes
US20070093490A1 (en) * 2005-09-02 2007-04-26 Olaf Prien Substituted imidazo[1,2b]pyridazines as kinase inhibitors, their preparation and use as medicaments
WO2007129052A1 (en) * 2006-05-03 2007-11-15 Astrazeneca Ab Pyrazole derivatives and their use as pi3k inhibitors
WO2008042639A1 (en) * 2006-10-02 2008-04-10 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2008138889A2 (en) * 2007-05-11 2008-11-20 Novartis Ag 3, 6-disubstituted-imidazo [1, 2-b] pyridazines and 3, 5-disubstituted pyrazolo[1, 5-a] pyrimidines as phosphatidylinositol-3-kinase inhibitors
WO2009050183A2 (en) * 2007-10-17 2009-04-23 Novartis Ag Imidazo [1, 2-a] pyridine derivatives useful as alk inhibitors

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6514977B1 (en) * 1997-05-22 2003-02-04 G.D. Searle & Company Substituted pyrazoles as p38 kinase inhibitors
NL1010576C2 (en) * 1998-11-17 2000-05-18 Skf Eng & Res Centre Bv Screw actuator, comprising a multifunctional sleeve and a claw.
DE102005042742A1 (en) * 2005-09-02 2007-03-08 Schering Ag Substituted imidazo [1,2b] pyridazines as kinase inhibitors, their production and use as pharmaceuticals
EP2231661A1 (en) * 2007-12-19 2010-09-29 Amgen, Inc. Inhibitors of pi3 kinase
JP2012518037A (en) * 2009-02-18 2012-08-09 アムジエン・インコーポレーテツド Indole / benzimidazole compounds as mTOR kinase inhibitors

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007027842A1 (en) * 2005-08-31 2007-03-08 Bayer Healthcare Llc Anilinopyrazole derivatives useful for the treatment of diabetes
US20070093490A1 (en) * 2005-09-02 2007-04-26 Olaf Prien Substituted imidazo[1,2b]pyridazines as kinase inhibitors, their preparation and use as medicaments
WO2007129052A1 (en) * 2006-05-03 2007-11-15 Astrazeneca Ab Pyrazole derivatives and their use as pi3k inhibitors
WO2008042639A1 (en) * 2006-10-02 2008-04-10 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2008138889A2 (en) * 2007-05-11 2008-11-20 Novartis Ag 3, 6-disubstituted-imidazo [1, 2-b] pyridazines and 3, 5-disubstituted pyrazolo[1, 5-a] pyrimidines as phosphatidylinositol-3-kinase inhibitors
WO2009050183A2 (en) * 2007-10-17 2009-04-23 Novartis Ag Imidazo [1, 2-a] pyridine derivatives useful as alk inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
ORG. LETT., vol. 11, 2009, pages 1837 - 1840

Cited By (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9029411B2 (en) 2008-01-25 2015-05-12 Millennium Pharmaceuticals, Inc. Thiophenes and uses thereof
US8415376B2 (en) 2008-05-30 2013-04-09 Amgen Inc. Inhibitors of PI3 kinase
US9139589B2 (en) 2009-01-30 2015-09-22 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
US9090601B2 (en) 2009-01-30 2015-07-28 Millennium Pharmaceuticals, Inc. Thiazole derivatives
US8796314B2 (en) 2009-01-30 2014-08-05 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
EP4092012A1 (en) 2010-06-04 2022-11-23 Amgen Inc. Piperidinone derivatives as mdm2 inhibitors for the treatment of cancer
EP3483143A1 (en) 2010-06-04 2019-05-15 Amgen, Inc Piperidinone derivatives as mdm2 inhibitors for the treatment of cancer
WO2011153509A1 (en) 2010-06-04 2011-12-08 Amgen Inc. Piperidinone derivatives as mdm2 inhibitors for the treatment of cancer
EP2927213A1 (en) 2010-06-04 2015-10-07 Amgen Inc. Piperidinone derivatives as MDM2 inhibitors for the treatment of cancer
US8796271B2 (en) 2010-08-11 2014-08-05 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
US8796268B2 (en) 2010-08-11 2014-08-05 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
US8859768B2 (en) 2010-08-11 2014-10-14 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
US9062038B2 (en) 2010-08-11 2015-06-23 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
US8765746B2 (en) 2010-10-13 2014-07-01 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
KR101875720B1 (en) * 2011-01-10 2018-07-09 인피니티 파마슈티칼스, 인코포레이티드 Processes for preparing isoquinolinones and solid forms of isoquinolinones
JP2014506601A (en) * 2011-02-23 2014-03-17 インテリカイン, エルエルシー Heterocyclic compounds and uses thereof
WO2013049250A1 (en) 2011-09-27 2013-04-04 Amgen Inc. Heterocyclic compounds as mdm2 inhibitors for the treatment of cancer
EP2793880A4 (en) * 2011-12-23 2015-06-24 Millennium Pharm Inc Heteroaryls and uses thereof
JP2015503504A (en) * 2011-12-23 2015-02-02 ミレニアム ファーマシューティカルズ, インコーポレイテッドMillennium Pharmaceuticals, Inc. Heteroaryl and uses thereof
WO2013096630A1 (en) * 2011-12-23 2013-06-27 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
JP2015522051A (en) * 2012-07-13 2015-08-03 ユーシービー バイオファルマ エスピーアールエル Imidazopyridine derivatives as modulators of TNF activity
WO2014130470A1 (en) 2013-02-19 2014-08-28 Amgen Inc. Cis-morpholinone and other compounds as mdm2 inhibitors for the treatment of cancer
WO2014151863A1 (en) 2013-03-14 2014-09-25 Amgen Inc. Heteroaryl acid morpholinone compounds as mdm2 inhibitors for the treatment of cancer
EP3805232A1 (en) 2013-06-10 2021-04-14 Amgen Inc. Crystalline synthetic intermediate useful in processes for making a mdm2 inhibitor
EP4039256A1 (en) 2013-11-11 2022-08-10 Amgen Inc. Combination therapy including an mdm2 inhibitor and dasatinib or nilotinib for the treatment of chronic myeloid leukemia
US11958850B2 (en) 2014-06-19 2024-04-16 Takeda Pharmaceutical Company Limited Heteroaryl compounds for kinase inhibition
US10227342B2 (en) 2014-06-19 2019-03-12 Ariad Pharmaceuticals, Inc. Heteroaryl compounds for kinase inhibition
US10227350B2 (en) 2016-09-23 2019-03-12 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10214519B2 (en) 2016-09-23 2019-02-26 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US10479770B2 (en) 2016-09-23 2019-11-19 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
US11905281B2 (en) 2017-05-22 2024-02-20 Amgen Inc. KRAS G12C inhibitors and methods of using the same
US11766436B2 (en) 2018-05-04 2023-09-26 Amgen Inc. KRAS G12C inhibitors and methods of using the same
WO2020106647A2 (en) 2018-11-19 2020-05-28 Amgen Inc. Combination therapy including a krasg12c inhibitor and one or more additional pharmaceutically active agents for the treatment of cancers
US11918584B2 (en) 2018-11-19 2024-03-05 Amgen Inc. Combination therapy including a KRASG12C inhibitor and one or more additional pharmaceutically active agents for the treatment of cancers
KR20210126065A (en) * 2019-05-13 2021-10-19 광동 어글레이어 압토일렉트라닉 머티어리얼즈 컴퍼니 리미티드 Imidazo nitrogen heterocyclic compound and its application
KR102615337B1 (en) 2019-05-13 2023-12-19 광동 어글레이어 압토일렉트라닉 머티어리얼즈 컴퍼니 리미티드 Imidazo nitrogen heterocyclic compounds and their applications
WO2020232130A1 (en) 2019-05-14 2020-11-19 Amgen Inc. Dosing of kras inhibitor for treatment of cancers
EP3738593A1 (en) 2019-05-14 2020-11-18 Amgen, Inc Dosing of kras inhibitor for treatment of cancers
US11827635B2 (en) 2019-05-21 2023-11-28 Amgen Inc. Solid state forms
CN114174293A (en) * 2019-07-24 2022-03-11 默克专利股份公司 4- (imidazo [1,2-a ] pyridin-3-yl) -pyrimidine derivatives
US11787796B2 (en) 2019-09-18 2023-10-17 Takeda Pharmaceutical Company Limited Plasma Kallikrein inhibitors and uses thereof
WO2021076655A1 (en) 2019-10-15 2021-04-22 Amgen Inc. Combination therapy of kras inhibitor and shp2 inhibitor for treatment of cancers
EP4061851A4 (en) * 2019-11-21 2023-12-20 BeiGene, Ltd. Methods of cancer treatment using anti-ox40 antibodies in combination with pi3 kinase delta inhibitors
WO2021126816A1 (en) 2019-12-16 2021-06-24 Amgen Inc. Dosing regimen of a kras g12c inhibitor
WO2022197755A1 (en) * 2021-03-17 2022-09-22 Shire Human Genetic Therapies, Inc. Imidazopyridinyl inhibitors of plasma kallikrein
WO2023009475A1 (en) * 2021-07-26 2023-02-02 President And Fellows Of Harvard College Rock2 inhibitors and uses thereof

Also Published As

Publication number Publication date
AU2010249040A1 (en) 2011-12-01
MX2011012037A (en) 2012-02-28
CA2760778A1 (en) 2010-11-18
JP2012526836A (en) 2012-11-01
AU2010249040B2 (en) 2013-08-22
EP2430013B1 (en) 2014-10-15
US20120190666A1 (en) 2012-07-26
EP2430013A1 (en) 2012-03-21

Similar Documents

Publication Publication Date Title
EP2430013B1 (en) Heteroaryl compounds as pikk inhibitors
AU2010241723B2 (en) Inhibitors of PI3 kinase and / or mTOR
EP2307400B1 (en) Inhibitors of pi3 kinase
JP5520831B2 (en) Inhibitors of PI3 kinase
US8729074B2 (en) Inhibitors of PI3 kinase
ES2656192T3 (en) SKY inhibitors
EA016028B1 (en) Fused heterocyclic derivatives and use thereof
KR20130094710A (en) 5,7-substituted-imidazo[1,2-c]pyrimidines as inhibitors of jak kinases
KR20110016931A (en) Fused nitrogen containing heterocycles and compositions thereof 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: 10719514

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2760778

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2010249040

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2012510985

Country of ref document: JP

Ref document number: MX/A/2011/012037

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2010719514

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2010249040

Country of ref document: AU

Date of ref document: 20100512

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13319683

Country of ref document: US