WO2013177668A1 - Protein kinase inhibitors - Google Patents

Protein kinase inhibitors Download PDF

Info

Publication number
WO2013177668A1
WO2013177668A1 PCT/CA2013/000513 CA2013000513W WO2013177668A1 WO 2013177668 A1 WO2013177668 A1 WO 2013177668A1 CA 2013000513 W CA2013000513 W CA 2013000513W WO 2013177668 A1 WO2013177668 A1 WO 2013177668A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
added
compound
solution
reduced pressure
Prior art date
Application number
PCT/CA2013/000513
Other languages
French (fr)
Inventor
Alain Laurent
Yannick Rose
James B. Jaquith
Original Assignee
Pharmascience, 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
Priority claimed from CA2779184A external-priority patent/CA2779184A1/en
Priority claimed from CA2813299A external-priority patent/CA2813299A1/en
Priority to CN201380028248.2A priority Critical patent/CN104379586B/en
Priority to US14/404,497 priority patent/US9796716B2/en
Priority to EP13796590.1A priority patent/EP2855484A4/en
Priority to RU2014145285A priority patent/RU2678767C2/en
Application filed by Pharmascience, Inc. filed Critical Pharmascience, Inc.
Priority to KR1020147036372A priority patent/KR101972990B1/en
Priority to IN2338MUN2014 priority patent/IN2014MN02338A/en
Priority to BR112014029718A priority patent/BR112014029718A2/en
Priority to JP2015514295A priority patent/JP6175495B2/en
Priority to CA2874211A priority patent/CA2874211A1/en
Priority to KR1020197011438A priority patent/KR20190043648A/en
Publication of WO2013177668A1 publication Critical patent/WO2013177668A1/en
Priority to HK15109229.7A priority patent/HK1208460A1/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled

Definitions

  • the present invention relates to a novel family of inhibitors of protein kinases.
  • the present invention relates to inhibitors of the members of the Tec and Src protein kinase families.
  • Protein kinases are a large group of intracellular and transmembrane signaling proteins in eukaryotic cells. These enzymes are responsible for transfer of the terminal (gamma) phosphate from ATP to specific amino acid residues of target proteins. Phosphorylation of specific tyrosine, serine or threonine amino acid residues in target proteins can modulate their activity leading to profound changes in cellular signaling and metabolism. Protein kinases can be found in the cell membrane, cytosol and organelles such as the nucleus and are responsible for mediating multiple cellular functions including metabolism, cellular growth and division, cellular signaling, modulation of immune responses, and apoptosis.
  • the receptor tyrosine kinases are a large family of cell surface receptors with protein tyrosine kinase activity that respond to extracellular cues and activate intracellular signaling cascades (Plowman et al. (1994) DN&P, 7(6) : 334-339).
  • inhibitors of select kinases or kinase families are expected to be useful in the treatment of cancer, autoimmune diseases, and
  • inflammatory conditions including, but not limited to: solid tumors, hematological malignancies, arthritis, graft versus host disease, lupus erythematosus, psoriasis, colitis, illeitis, multiple sclerosis, uveitis, coronary artery vasculopathy, systemic sclerosis, atherosclerosis, asthma, transplant rejection, allergy, dermatomyositis, pemphigus and the like.
  • PDGFR platelet-derived growth factor receptor
  • VEGFR vascular endothelial growth factor receptor
  • intracellular proteins such as members of the Syk, SRC, and Tec families of kinases.
  • Tec kinases are non-receptor tyrosine kinases predominantly, but not exclusively, expressed in cells of hematopoietic origin (Bradshaw JM. Cell Signal. 2010,22 : 1175-84).
  • the Tec family includes Tec, Bruton's tyrosine kinase (Btk), inducible T-cell kinase (Itk), resting lymphocyte kinase
  • Btk is a Tec family kinase which is important in B-cell receptor signaling. Btk is activated by Src-family kinases and phosphorylates PLC gamma leading to effects on B-cell function and survival. Additionally, Btk is important in signal transduction in response to immune complex recognition by macrophage, mast cells and neutrophils. Btk inhibition is also important in survival of lymphoma cells (Herman, SEM. Blood 2011, 117:6287-6289) suggesting that inhibition of Btk may be useful in the treatment of lymphomas.
  • cSRC is the prototypical member of the SRC family of tyrosine kinases which includes Lyn, Fyn, Lck, Hck, Fgr, Blk, Syk, Yrk, and Yes.
  • cSRC is critically involved in signaling pathways involved in cancer and is often over-expressed in human malignancies (Kim LC, Song L, Haura EB. Nat Rev Clin Oncol. 2009 6(10) : 587-9).
  • the role of cSRC in cell adhesion, migration and bone remodeling strongly implicate this kinase in the development and progression of bone metastases.
  • cSRC is also involved in signaling downstream of growth factor receptor tyrosine kinases and regulates cell cycle progression suggesting that cSRC inhibition would impact cancer cell proliferation.
  • SRC family members may be useful in treatments designed to modulate immune function.
  • SRC family members including Lck, regulate T-cell receptor signal transduction which leads to gene regulation events resulting in cytokine release, survival and proliferation.
  • inhibitors of Lck have been keenly sought as immunosuppressive agents with potential application in graft rejection and T-cell mediated autoimmune disease (Martin et al. Expert Opin Ther Pat. 2010, 20 : 1573-93).
  • pyrazolopyrimidines as therapeutic agents.
  • Btk is included in a long list of biologically un-related kinases. No evidence of kinase inhibition or cellular activity was disclosed in WO02/080926 and exemplification centers on amide and sulfonamide derivatives with a limited subset of unsubstituted 4- phenoxyphenyl derivatives.
  • US Patent No. 7,514,444 discloses inhibitors of Btk.
  • Compound 13 (PCI- 32765) of this patent has been reported to show ATP competitive binding to a wide range of kinases including Btk, Lck, Lyn, cSRC, Jak, EGFR, KDR and others (Honigberg, L.A, et al, The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy, PNAS vol. 107 no. 29, 13075-13080).
  • the acrylamide functionality of compound 13 is reported to covalently bind the thiol moiety of Cys481, which is situated adjacent to the ATP binding pocket of Btk, thus inducing "sustained" inhibition of Btk.
  • compound 13 also inhibits various kinases which also feature a Cys adjacent to the ATP binding pocket, such as Bmx, Tec, Txk, Itk, EGFR, ErbB2, ErbB4, Jak3 and Blk. Covalent binding to any of these kinases may diminish the selective nature of this approach.
  • GDC-0834 belongs to a structurally unrelated family of compounds which were recently reported to demonstrate significant Btk selectivity (Liu L., et al, Antiarthritis effect of a novel Bruton's tyrosine kinase (BTK) inhibitor in rat collagen-induced arthritis and mechanism-based
  • GDC-0834 was active in several animal models of autoimmune disease. However, this compound failed in Phase 1 clinical trials as a result of human specific metabolism (Liu L, et al, Significant species difference in amide hydrolysis of GDC-0834, a novel potent and selective Bruton's tyrosine kinase inhibitor, Drug Metab Dispos. 2011
  • regimens such as cancer, inflammatory and autoimmune diseases.
  • the present invention relates to a family of potent and selective, non- covalent Btk inhibitors which demonstrate cellular activity, oral exposure and activity in animal models of inflammation and autoimmune disease.
  • Kinase selectivity and cellular potency are related to specific substitution patterns on the compounds. Synthetic methods are disclosed which provide compounds on multi-gram scale.
  • the present invention relates to a novel family of kinase inhibitors.
  • One aspect of the present invention is directed to a compound of Formula 1 :
  • R 1 is selected from the group consisting of:
  • Y is selected from :
  • Z is selected from :
  • Y-Z-W is selected from :
  • X 1 and X 2 are independently selected from hydrogen, halogen n is an integer from 0 to 2;
  • n is an integer from 0 to 2;
  • n' is an integer from 0 to 2; W is independently selected from :
  • R 2 is selected from hydrogen or alkyl
  • R 3 is selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted carbocyclyl, substituted or unsubstitutded heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl or substituted or unsubstituted heteroaralkyl;
  • R 4 is selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted carbocyclyl, substituted or unsubstitutded heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl or substituted or unsubstituted heteroaraikyl ;
  • R 5 and R 6 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl or R 5 and R 6 can be fused to form a 3 to 8 membered heterocyclyl ring system.
  • Preferred embodiment includes compounds of Formula 1 where W is selected from -OR 3 and R 3 is selected from substituted or unsubstituted aralkyl, or substituted or unsubstituted heteroaraikyl.
  • Preferred embodiment includes compounds of Formula 1 where W is selected from the group consisting of:
  • Preferred embodiment includes compounds of Formula 1 where R x is selected from the group consisting of:
  • Preferred embodiment includes compounds of Formula 1 where Y is selected from the group consisting of:
  • Preferred embodiment includes compounds of Formula 1 where Z is selected from the group consisting of:
  • More preferred embodiment includes compounds of Formula 1 where W is selected from the group consisting of:
  • More preferred embodiment includes compounds of Formula 1 where R 1 is selected from the group consisting of:
  • More preferred embodiment includes compounds of Formula 1 where Z is selected from the group consisting of:
  • More preferred embodiment includes compounds of Formula 1 where Y-2-W is selected from the group consisting of:
  • Another aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound of Formula 1 and a pharmaceutically acceptable carrier, diluent or excipient.
  • Another aspect of the present invention provides a method of modulating kinase function, the method comprising contacting a cell with a compound of the present invention in an amount sufficient to modulate the enzymatic activity of a given kinase or kinases, such as Btk, thereby modulating the kinase function.
  • Another aspect of the present invention provides a method of modulating the target kinase function, the method comprising a) contacting a cell with a compound of the present invention in an amount sufficient to modulate the target kinase function, thereby b) modulating the target kinase activity and signaling.
  • probe comprising a compound of Formula 1 labeled with a detectable label or an affinity tag.
  • the probe comprises a residue of a compound of Formula 1 covalently conjugated to a detectable label.
  • detectable labels include, but are not limited to, a fluorescent moiety, a
  • chemiluminescent moiety a paramagnetic contrast agent, a metal chelate, a radioactive isotope-containing moiety, or biotin.
  • the present invention relates to novel kinase inhibitors. These compounds are found to have activity as inhibitors of protein kinases: including members of the tyrosine kinases Aurora, SRC (more specifically Lck) and Tec (more specifically Btk) kinase families.
  • the pharmaceutical compositions which comprises an effective amount of a compound of Formula 1 with a pharmaceutically acceptable diluent or carrier.
  • the pharmaceutical compositions may be in a conventional pharmaceutical form suitable for oral administration (e.g., tablets, capsules, granules, powders and syrups), parenteral administration (e.g., injections (intravenous, intramuscular, or subcutaneous)), drop infusion preparations, inhalation, eye lotion, topical administration (e.g., ointment), or suppositories.
  • parenteral administration e.g., injections (intravenous, intramuscular, or subcutaneous)
  • drop infusion preparations e.g., inhalation, eye lotion
  • topical administration e.g., ointment
  • suppositories e.g., ointment
  • the compounds may be formulated into pharmaceutically acceptable dosage forms by conventional methods known to those skilled in the art.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those ligands, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material.
  • Each carrier must be acceptable in the sense of being compatible with the other ingredients of the formulation, including the active ingredient, and not injurious or harmful to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch, potato starch, and substituted or unsubstituted ⁇ -cyclodextrin; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, manni
  • pharmaceutically acceptable salt refers to the relatively non-toxic, inorganic and organic acid addition salts of the compound(s). These salts can be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting a purified compound(s) in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, laurylsulphonate salts, and amino acid salts, and the like (See, for example, Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66 : 1-19).
  • the compounds of the present invention may contain one or more acidic functional groups and, thus, are capable of forming
  • pharmaceutically acceptable salts with pharmaceutically acceptable bases.
  • pharmaceutically acceptable salts refers to the relatively non-toxic inorganic and organic base addition salts of a
  • salts can likewise be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting the purified compound(s) in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine.
  • a suitable base such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine,
  • affinity tag means a ligand or group, linked either to a compound of the present invention or to a protein kinase domain, that allows the conjugate to be extracted from a solution.
  • alkyl refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups, including haloalkyl groups such as trifluoromethyl and 2,2,2-trifluoroethyl, etc.
  • Representative alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, (cyclohexyl)methyl, cyclopropylmethyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • alkenyl and alkynyl refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
  • alkenyl groups include vinyl, propen-2-yl, crotyl, isopenten-2- yl, l,3-butadien-2-yl), 2,4-pentadienyl, and l,4-pentadien-3-yl.
  • alkynyl groups include ethynyl, 1- and 3-propynyl, and 3- butynyl.
  • alkyl substituents are lower alkyl groups, e.g., having from 1 to 6 carbon atoms.
  • alkenyl and alkynyl preferably refer to lower alkenyl and alkynyl groups, e.g., having from 2 to 6 carbon atoms.
  • alkylene refers to an alkyl group with two open valencies (rather than a single valency), such as -(CH 2 )i-io- and substituted variants thereof.
  • alkoxy refers to an alkyl group having an oxygen attached thereto. Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like.
  • An "ether” is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxy.
  • alkoxyalkyl refers to an alkyl group substituted with an alkoxy group, thereby forming an ether.
  • amide and “amido” are art-recognized as an amino-substituted carbonyl and includes a moiety that can be represented by the general formula :
  • R 9 , R 10 are as defined above.
  • Preferred embodiments of the amide will not include imides, which may be unstable.
  • amine and “amino” are art-recognized and refer to both unsubstituted and substituted amines and salts thereof, e.g., a moiety that can be represented by the general formulae:
  • R 9 , R 10 and R 10' each independently represent a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -R 8 , or R 9 and R 10 taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure;
  • R 8 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocyclyl or a polycyclyl; and m is zero or an integer from 1 to 8.
  • only one of R 9 or R 10 can be a carbonyl, e.g., R 9 , R 10 , and the nitrogen together do not form an imide.
  • R 9 and R 10 each independently represent a hydrogen, an alkyl, an alkenyl, or -(CH 2 ) m -R 8 .
  • the amino group is basic, meaning the protonated form has a pK a >. 7.00.
  • aralkyl refers to an alkyl group substituted with an aryl group, for example -(CH 2 ) n -Ar.
  • heteroaryl refers to an alkyl group substituted with a heteroaryl group, for example -(CH 2 ) n -Het.
  • aryl as used herein includes 5-, 6-, and 7-membered substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon.
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, anthracene, and phenanthrene.
  • carrier refers to a non- aromatic substituted or unsubstituted ring in which each atom of the ring is carbon.
  • carrier also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is
  • carbocyclic e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Representative carbocyclic groups include cyclopentyl, cyclohexyl, 1-cyclohexenyl, and 3- cyclohexen-l-yl, cycloheptyl.
  • carbonyl is art-recognized and includes such moieties as can be represented by the general formula : wherein X is a bond or represents an oxygen or a sulfur, and R 11 represents a hydrogen, an alkyl, an alkenyl, -(CH 2 ) m -R 8 or a pharmaceutically acceptable salt. Where X is an oxygen and R 11 is not hydrogen, the formula represents an "ester”. Where X is an oxygen, and R 11 is a hydrogen, the formula represents a "carboxylic acid".
  • heteroaryl includes substituted or unsubstituted aromatic 5- to 7-membered ring structures, more preferably 5- to 6-membered rings, whose ring structures include one to four heteroatoms.
  • heteroaryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, isoxazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • heteroatom as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, and sulfur.
  • heterocyclyl or “heterocyclic group” refer to substituted or unsubstituted non-aromatic 3- to 10-membered ring structures, more preferably 3- to 7-membered rings, whose ring structures include one to four heteroatoms.
  • heterocyclyl or “heterocyclic group” also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Heterocyclyl groups include, for example, tetrahydrofuran, tetrahydropyran, piperidine, piperazine, pyrrolidine, morpholine, lactones, and lactams.
  • Hydrocarbyl groups include, but are not limited to aryl, heteroaryl, carbocycle, heterocycle, alkyl, alkenyl, alkynyl, and combinations thereof.
  • polycyclyl or “polycyclic” refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or
  • heterocyclyls in which two or more carbons are common to two adjoining rings, e.g., the rings are "fused rings".
  • Each of the rings of the polycycle can be substituted or unsubstituted.
  • the term "probe” means a compound of the invention which is labeled with either a detectable label or an affinity tag, and which is capable of binding, either covalently or non-covalently, to a protein kinase domain.
  • the probe When, for example, the probe is non-covalently bound, it may be displaced by a test compound.
  • the probe When, for example, the probe is bound covalently, it may be used to form cross-linked adducts, which may be quantified and inhibited by a test compound.
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • Substituents can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety.
  • Compounds of the invention also include all isotopes of atoms present in the intermediates and/or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include deuterium and tritium.
  • Benzoyl chlorides of formula 2-i were condensed with malononitrile to provide intermediate 2-ii.
  • Methylation of intermediate 2-ii with TMS- diazomethane provided intermediate 2-iii.
  • Condensation of intermediate 2-iii with hydrazine provided intermediate 2-iv.
  • Further condensation of intermediate 2-iv with formamidine provided intermediate 2-v.
  • Intermediate 2-v was treated with alcohol F ⁇ OH, under Mitsunobu conditions, to provide intermediate 2-vi.
  • Ullmann condensation of intermediate 2-vi with phenolic intermediates 2-vii provided the desired compounds or intermediates of general formula 2-viii.
  • intermediate 2-iv was treated with an alcohol of formula RiOH, under Mitsunobu conditions to provide intermediate 2-ix.
  • Condensation of intermediate 2-ix with formamidine provided intermediate 2-vi.
  • trimethyl orthoformate and ammonia can be used in place of formamidine, for example, in the conversion of intermediate 2-x to compounds of formula 2-viii.
  • intermediate 3-b (17.5 g, 50.2 mmol) in THF (200 ml) and MeOH (100 ml) was added 2N sodium hydroxide (100 ml, 200 mmol) and the reaction was stirred at room temperature overnight. Volatiles were removed under reduced pressure. 10% aqueous HCI and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure to provide intermediate 3-c as beige solid.
  • intermediate 9-b (4.49 g, 17.07 mmol) in ethanol (8.5 ml) was added a solution of hydrazine monohydrate (2.23 ml, 46.1 mmol) and the reaction was stirred at 100 °C for 1 hour and then cooled to room temperature. Volatiles were removed under reduced pressure to provide intermediate 9-c as a yellow solid.
  • Step 1 Intermediate 11-a To a solution of resorcinol (15.0 g, 136 mmol) in DMF (100 ml), cooled to 0°C, were added imidazole (19.48 g, 286 mmol) and tert- butylchlorodimethylsilane (21.56 g, 143 mmol). The reaction was then stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added; the organic layer was separated, washed 3 times with a saturated aqueous solution of ammonium chloride and brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 11-a as a colorless oil.
  • Tetrabutylammonium fluoride trihydrate (3.93 g, 12.47 mmol) was added to a solution of intermediate 11-b (2.9 g, 8.31 mmol) in THF (15 ml.) and the reaction was stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 , filtered and concentrated under reduced pressure. Purification by silica gel
  • Step 4 Compound 9 A solution of intermediate 10-a (200 mg, 0.56 mmol), intermediate 11-c (229 mg, 0.977 mmol), quinolin-8-ol (16.21 mg, 0.112 mmol), copper (I) chloride (11.05 mg, 0.11 mmol) and cesium carbonate (546 mg, 1.67 mmol), in dimethylacetamide ( 1 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature.
  • dichloromethane 100 mL were sequentially added triethylamine (25.08 ml, 181 mmol), tert-butylchlorodimethylsilane (20.0 g, 133 mmol) portion wise, and the reaction was stirred at room temperature overnight. 10% aqueous citric acid was added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure.
  • Step 2 Intermediate 14-b To a solution of intermediate 14-a (16.0 g, 67.7 mmol) in methanol (100 ml) cooled to 0°C was added portion wise sodium borohydride (1.28 g, 33.8 mmol). After the addition was completed the reaction was stirred at room temperature for 2 hours. Volatiles were removed under reduced pressure. Water and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure to provide intermediate 14-b as a yellow oil.
  • triphenylphosphine (1.32 g, 5.03 mmol) and DIAD (991 ⁇ , 5.03 mmol) drop wise at room temperature; the reaction was stirred at reflux for 2 hours and then cooled to room temperature.
  • Step 2 Compound 17 A solution of intermediate 12-a (200 mg, 0.6 mmol), intermediate 14-d (259 mg, 1.15 mmol), quinolin-8-ol (19.0 mg, 0.13 mmol), copper (I) chloride (13.0 mg, 0.13 mmol) and cesium carbonate (643 mg, 1.97 mmol) in dimethylacetamide (3.0 ml) was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight.
  • triphenylphosphine (10.35 g, 39.5 mmol) and DIAD (7.68 ml, 39.5 mmol) drop wise at room temperature. The reaction was then stirred for 18 hours. Volatiles were removed in vacuo. Purification by silica gel chromatography provided intermediate 15-b as a colorless oil.
  • intermediate 19-a (13.9 g, 36.3 mmol) in THF (182.0 ml) was added a l.OM solution of tetrabutylammonium fluoride in THF (36.3 ml, 36.3 mmol) and the reaction was stirred at room temperature for 1 hour. Saturated aqueous ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 19-b as colorless oil.
  • Tetrabutylammonium fluoride trihydrate (1.81 g, 5.75 mmol) was added to a solution of intermediate 22-a (2.2 g, 5.75 mmol) in THF (23 ml_) and the reaction was stirred at room temperature for 1 hour. Saturated aqueous ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 22-b as a colorless oil.
  • Tetrabutylammonium fluoride trihydrate (2.88 g, 9.14 mmol) was added to a solution of intermediate 28-a (2.1 g, 6.10 mmol) in THF (10 mL) and the reaction was stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel
  • intermediate 29-f 10.25 g, 80.0 mmol
  • DMF 50 ml
  • imidazole 5.99 g, 88.0 mmol
  • tert- butylchlorodimethylsilane 13.27 g, 88.0 mmol
  • the reaction was then stirred at room temperature overnight.
  • a saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed 3 times with a saturated aqueous solution of ammonium chloride and brine, dried over MgS0 4 , filtered and concentrated under reduced pressure.
  • Purification by silica gel chromatography provided intermediate 29-g as a yellow oil.
  • triphenylphosphine (12.15 g, 46.3 mmol) and DIAD (9.0 ml, 46.3 mmol) at room temperature and the reaction was then stirred at room temperature overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 29-h as a yellow oil.
  • intermediate 30-a (12.5 g, 35.9 mmol) in THF (72 ml) was added a 1.0 M solution of TBAF in THF (35.9 ml, 35.9 mmol) and the reaction was stirred at room temperature for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 30-b as a white solid.
  • intermediate 31-c 7.6 g, 21.87 mmol
  • THF 44 ml
  • tetrabutylammonium fluoride trihydrate 5.72 g, 21.87 mmol
  • Saturated aqueous ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over gS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 31-d as white solid.
  • intermediate 32-e (2.2 g, 4.91 mmol) in THF (9.8 ml) was added a 1.0 M solution of TBAF in THF (4.91 ml, 4.91 mmol) and the reaction was stirred at room temperature for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 32-f as a white solid.
  • intermediate 34-c (3.2 g, 12.02 mmol) in MeOH (25.0 ml) was added 4N HCI in dioxane (10.95 ml, 361.0 mmol) and the reaction was stirred overnight at room temperature. Volatiles were removed in vacuo. Diethyl ether was added to the residue; a precipitate formed and was collected by filtration to provide intermediate 34-d- HCI as a white solid.
  • intermediate 35-b (1.50 g, 11.89 mmol) and intermediate 34-b (2.07 g, 11.89 mmol) in DMPU (11.89 mL) and toluene (11.89 mL) was added sodium 2-methylpropan-2-olate (3.43 g, 35.7 mmol) at room temperature.
  • the reaction was stirred overnight at 80 °C and then cooled to room temperature.
  • a saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 35-c as a yellow oil.
  • Step 3 Intermediate 35-d To a solution of intermediate 35-c (3.30 g, 11.77 mmol) in MeOH (36.2 mL) was added 4N HCI in dioxane (10.7 mL, 353 mmol) and the reaction was stirred at room temperature overnight. Volatiles were removed under reduced pressure. Diethyl ether was added to the residue; a precipitate formed and was collected by filtration to provide intermediate 35-d-HCI as a white solid.
  • intermediate 36-c (1.5 g, 5.67 mmol) and H 2 W0 4 (14 mg, 0.057 mmol) in MeOH (22.7 mL) at 40°C was added H 2 0 2 (1.85 mL, 18.16 mmol) drop wise. The mixture was stirred at reflux overnight and then cooled to room temperature. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 36- d as a colorless oil.
  • Step 1 Intermediates 38-b and 38-b' To a solution of lH-imidazole-5-carbaldehyde (5.0 g, 45.4 mmol) in DMF (20 mL) was added a 60% dispersion of NaH mineral oil (1.81 g, 45.4 mmol) portion wise. After stirring for 30 minutes at room temperature, (2- (chloromethoxy)ethyl)trimethylsilane (9.08 g, 54.5 mmol) was added and the reaction was then stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediates 38-b and 38-b' as an inseparable mixture.
  • intermediate 38-b and 38-b' 7.0 g, 29.1 mmol
  • THF 116.0 ml
  • IMaBH 4 1.10 g, 29.1 mmol
  • the reaction was stirred overnight at room temperature and then cooled to 0°C.
  • a saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate.
  • the combined organic extracts were washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure to provide intermediates 38-c and 38-c' as an inseparable mixture.
  • intermediate 40-a (3.40 g, 10.2 mmol) in THF (20 mL) was added a 1.0 M solution of TBAF in THF (10.2 ml, 10.2 mmol) and the reaction was stirred at room temperature for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 40-b as a white solid.
  • intermediate 41-a (1.40 g, 4.21 mmol) in THF (10 ml) was added a 1.0 M solution of TBAF in THF (4.63 ml, 4.63 mmol) and the reaction was stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 41-b as a colorless oil.
  • Step 1 Intermediate 44-b tert-Butyl hydrazinecarboxylate (7.60 g, 57.5 mmol) was added to acetone (50 mL) and the reaction was stirred overnight at room temperature.
  • intermediate 9-b (12.61 g, 47.9 mmol) and TEA (14.70 mL, 105.0 mmol) in EtOH (96.0 ml) was added intermediate 44-c.HCI (6.36 g, 57.5 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel
  • intermediate 49-b-HCI 650 mg, 5.13 mmol
  • EtOH EtOH
  • intermediate 49-b-HCI 650 mg, 5.13 mmol
  • a saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure to provide intermediate 49-c as a white solid.
  • intermediate 50-a (10.0 g, 49.0 mmol) in THF (49.0 mL) and MeOH (49.0 mL) was added sodium cyanoborohydride (3.69 g, 58.8 mmol) portion wise.
  • the reaction was refluxed under nitrogen for 10 minutes, and then cooled to room temperature.
  • 6I ⁇ I HCI 40 mL was added, the mixture was refluxed for 3 hours, cooled to room temperature and stirred overnight.
  • the reaction was filtered to remove inorganic insoluble material and the filtrate was concentrated under reduced pressure and azeotroped three times with toluene to provide intermediate 50-b-HCI as a white solid.
  • N,N-dimethylglycine 102 mg, 0.99 mmol
  • copper(I) iodide 63 mg, 0.33 mmol
  • cesium carbonate 431 mg, 1.32 mmol
  • the reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite.
  • a saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate.
  • the combined organic extracts were washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 77-a and 77-a' as an inseparable mixture.
  • intermediate 82-c (2.0 g, 7.12 mmol) and TEA (1.98 ml, 14.23 mmol) in EtOH (3.50 ml) was added intermediate 43-d-HCI (1.30 g, 8.54 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure to provide intermediate 83-a as a yellow solid.
  • intermediate 82-c (1.00 g, 3.56 mmol) and TEA (1.09 ml, 7.83 mmol) in EtOH (3.50 ml) was added intermediate 42-c (583 mg, 4.27 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of
  • intermediate 82-c (2.0 g, 7.12 mmol) and TEA (2.18 ml, 15.65 mmol) in EtOH (7.12 ml) was added intermediate isopropylhydrazine hydrochloride (944 mg, 8.54 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS0 4 , filtered and concentrated under reduced pressure.
  • Fluorescence polarization-based kinase assays were performed in 384 well- plate format using histidine tagged recombinant human full-length Bruton Agammaglobulinemia Tyrosine Kinase (Btk) and a modified protocol of the KinEASETM FP Fluorescein Green Assay supplied from Millipore. Kinase reaction were performed at room temperature for 60 minutes in presence of 250 ⁇ substrate, 10 ⁇ ATP and variable test article concentrations. The reaction was stopped with EDTA/kinase detection reagents and the
  • Splenocytes were obtained from 6 week old male CD1 mice (Charles River Laboratories Inc.). Mouse spleens were manually disrupted in PBS and filtered using a 70um cell strainer followed by ammonium chloride red blood cell lysis. Cells were washed, resuspended in Splenocyte Medium (HyClone RPMI supplemented with 10% heat-inactivated FBS, 0.5X non-essential amino acids, lOmM HEPES, 50uM beta mercaptoethanol) and incubated at 37 °C, 5% C0 2 for 2h to remove adherent cells.
  • Splenocyte Medium HyClone RPMI supplemented with 10% heat-inactivated FBS, 0.5X non-essential amino acids, lOmM HEPES, 50uM beta mercaptoethanol
  • Suspension cells were seeded in 96 well plates at 50,000 cells per well and incubated at 37°C, 5% C0 2 for lh.
  • Splenocytes were pre-treated in triplicate with 10,000 nM curves of Formula 1 compounds for lh, followed by stimulation of B cell proliferation with 2.5ug/ml anti-IgM F(ab') 2 (Jackson ImmunoResearch) for 72h.
  • Cell proliferation was measured by Cell Titer-Glo Luminescent Assay (Promega).
  • EC 50 values (50% proliferation in the presence of compound as compared to vehicle treated controls) were calculated from dose response compound curves using GraphPad Prism Software.
  • the animals were then returned to their home cage and skin punches (8 mm) were collected from each injection site four hours later.
  • the samples were placed in 1 mL formamide overnight at 80 degrees C (1 skin biopsy per 1 mL formamide in a glass tube). The amount of Evan's blue in the
  • formamide solution was then assessed by spectrophotometry (630 nm) as a measure of serum extravasation into the dermis.
  • mice Male BlORlll mice (7-9 wks on arrival) were habituated to the animal facility for at least 4 days. On experimental day 0 mice were anaesthetized with isoflurane and the dorsal surface was shaved. Collagen, emulsified in Freund's complete adjuvant (CFA) supplemented with additional mycobacterium tuberculosis (TB) H37Ra, was injected intradermal ⁇ at the base of the tail (0.15 mL / animal; 2 mg/mL collagen and 2.5 mg/mL TB in CFA). This CFA treatment was repeated on day 15.
  • CFA complete adjuvant
  • TB mycobacterium tuberculosis
  • RA Day 1 From day 15 to the end of the study animals were scored daily for signs of arthritis.
  • RA Day 1 On the first day of disease (RA Day 1) animals were recruited to the study and grouped using a balanced design based on arthritis score. Once recruited, animals were weighed and dosed twice daily by gavage (PO, BID). recruited animals were then scored twice a week on RA days 1, 5, 8 and 12. At the end of the study (RA day 12) animals were weighed and scored.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Rheumatology (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Transplantation (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to a novel family of inhibitors of protein kinases. In particular, the present invention relates to inhibitors of the members of the Tec and Src protein kinase families.

Description

PROTEIN KINASE INHIBITORS
FIELD OF INVENTION
The present invention relates to a novel family of inhibitors of protein kinases. In particular, the present invention relates to inhibitors of the members of the Tec and Src protein kinase families.
BACKGROUND OF THE INVENTION
Protein kinases are a large group of intracellular and transmembrane signaling proteins in eukaryotic cells. These enzymes are responsible for transfer of the terminal (gamma) phosphate from ATP to specific amino acid residues of target proteins. Phosphorylation of specific tyrosine, serine or threonine amino acid residues in target proteins can modulate their activity leading to profound changes in cellular signaling and metabolism. Protein kinases can be found in the cell membrane, cytosol and organelles such as the nucleus and are responsible for mediating multiple cellular functions including metabolism, cellular growth and division, cellular signaling, modulation of immune responses, and apoptosis. The receptor tyrosine kinases are a large family of cell surface receptors with protein tyrosine kinase activity that respond to extracellular cues and activate intracellular signaling cascades (Plowman et al. (1994) DN&P, 7(6) : 334-339).
Aberrant activation or excessive expression of various protein kinases are implicated in the mechanism of multiple diseases and disorders
characterized by benign and malignant proliferation, excess angiogenesis, as well as diseases resulting from inappropriate activation of the immune system. Thus, inhibitors of select kinases or kinase families are expected to be useful in the treatment of cancer, autoimmune diseases, and
inflammatory conditions including, but not limited to: solid tumors, hematological malignancies, arthritis, graft versus host disease, lupus erythematosus, psoriasis, colitis, illeitis, multiple sclerosis, uveitis, coronary artery vasculopathy, systemic sclerosis, atherosclerosis, asthma, transplant rejection, allergy, dermatomyositis, pemphigus and the like. Examples of kinases that can be targeted to modulate disease include receptor tyrosine kinases such as members of the platelet-derived growth factor receptor (PDGFR), vascular endothelial growth factor receptor (VEGFR) families and intracellular proteins such as members of the Syk, SRC, and Tec families of kinases.
Tec kinases are non-receptor tyrosine kinases predominantly, but not exclusively, expressed in cells of hematopoietic origin (Bradshaw JM. Cell Signal. 2010,22 : 1175-84). The Tec family includes Tec, Bruton's tyrosine kinase (Btk), inducible T-cell kinase (Itk), resting lymphocyte kinase
(Rlk/Txk), and bone marrow-expressed kinase (Bmx/Etk). Btk is a Tec family kinase which is important in B-cell receptor signaling. Btk is activated by Src-family kinases and phosphorylates PLC gamma leading to effects on B-cell function and survival. Additionally, Btk is important in signal transduction in response to immune complex recognition by macrophage, mast cells and neutrophils. Btk inhibition is also important in survival of lymphoma cells (Herman, SEM. Blood 2011, 117:6287-6289) suggesting that inhibition of Btk may be useful in the treatment of lymphomas.
cSRC is the prototypical member of the SRC family of tyrosine kinases which includes Lyn, Fyn, Lck, Hck, Fgr, Blk, Syk, Yrk, and Yes. cSRC is critically involved in signaling pathways involved in cancer and is often over-expressed in human malignancies (Kim LC, Song L, Haura EB. Nat Rev Clin Oncol. 2009 6(10) : 587-9). The role of cSRC in cell adhesion, migration and bone remodeling strongly implicate this kinase in the development and progression of bone metastases. cSRC is also involved in signaling downstream of growth factor receptor tyrosine kinases and regulates cell cycle progression suggesting that cSRC inhibition would impact cancer cell proliferation.
Additionally, inhibition of SRC family members may be useful in treatments designed to modulate immune function. SRC family members, including Lck, regulate T-cell receptor signal transduction which leads to gene regulation events resulting in cytokine release, survival and proliferation. Thus, inhibitors of Lck have been keenly sought as immunosuppressive agents with potential application in graft rejection and T-cell mediated autoimmune disease (Martin et al. Expert Opin Ther Pat. 2010, 20 : 1573-93).
Inhibition of kinases using small molecule inhibitors has successfully led to several approved therapeutic agents used in the treatment of human conditions. Herein, we disclose a novel family of kinase inhibitors. Further, we demonstrate that modifications in compound substitution can influence kinase selectivity and therefore the biological function of that agent.
PCT Publication Nos. WO02/080926 and WO02/76986 disclose
pyrazolopyrimidines as therapeutic agents. Btk is included in a long list of biologically un-related kinases. No evidence of kinase inhibition or cellular activity was disclosed in WO02/080926 and exemplification centers on amide and sulfonamide derivatives with a limited subset of unsubstituted 4- phenoxyphenyl derivatives.
US Patent No. 7,514,444 discloses inhibitors of Btk. Compound 13 (PCI- 32765) of this patent has been reported to show ATP competitive binding to a wide range of kinases including Btk, Lck, Lyn, cSRC, Jak, EGFR, KDR and others (Honigberg, L.A, et al, The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy, PNAS vol. 107 no. 29, 13075-13080). Specifically for Btk, the acrylamide functionality of compound 13 is reported to covalently bind the thiol moiety of Cys481, which is situated adjacent to the ATP binding pocket of Btk, thus inducing "sustained" inhibition of Btk. However, compound 13 also inhibits various kinases which also feature a Cys adjacent to the ATP binding pocket, such as Bmx, Tec, Txk, Itk, EGFR, ErbB2, ErbB4, Jak3 and Blk. Covalent binding to any of these kinases may diminish the selective nature of this approach.
GDC-0834 belongs to a structurally unrelated family of compounds which were recently reported to demonstrate significant Btk selectivity (Liu L., et al, Antiarthritis effect of a novel Bruton's tyrosine kinase (BTK) inhibitor in rat collagen-induced arthritis and mechanism-based
pharmacokinetic/pharmacodynamic modeling : relationships between inhibition of BTK phosphorylation and efficacy. J Pharmacol Exp Ther. 2011 Jul; 338(l) : 154-63). GDC-0834 was active in several animal models of autoimmune disease. However, this compound failed in Phase 1 clinical trials as a result of human specific metabolism (Liu L, et al, Significant species difference in amide hydrolysis of GDC-0834, a novel potent and selective Bruton's tyrosine kinase inhibitor, Drug Metab Dispos. 2011
Oct; 39( 10) : 1840-9).
Inhibition of EGFR has been related to the induction of severe rash with multiple clinical compounds (Tan AR, et al, Markers in the epidermal growth factor receptor pathway and skin toxicity during erlotinib treatment. Ann Oncol. 2008 Jan; 19(l) : 185-90). Similarly, inhibition of KDR (VEGFR2) has been clinically related to hypertension (Howard R. Mellor, et al.,Cardiotoxicity Associated with Targeting Kinase Pathways in Cancer, Toxicological Sciences 120( 1), 14-32 (2011). Therefore, the development of Btk inhibitors which demonstrated greater kinase selectivity could potentially be useful in various B-cell related indications which require acute and/or chronic dosing
regimens, such as cancer, inflammatory and autoimmune diseases.
The present invention relates to a family of potent and selective, non- covalent Btk inhibitors which demonstrate cellular activity, oral exposure and activity in animal models of inflammation and autoimmune disease. Kinase selectivity and cellular potency are related to specific substitution patterns on the compounds. Synthetic methods are disclosed which provide compounds on multi-gram scale. SUMMARY OF THE INVENTION
The present invention relates to a novel family of kinase inhibitors.
Compounds of this class have been found to have inhibitory activity against members of the Tec and Scr protein kinase families.
One aspect of the present invention is directed to a compound of Formula 1 :
Figure imgf000006_0001
Formula 1
wherein
R1 is selected from the group consisting of:
1) hydrogen,
2) alkyl,
3) heteroalkyi,
4) carbocyclyl,
5) heterocyclyl,
6) -C(0)R4, wherein the alkyl, heteroalkyi, carbocyclyl and heterocyclyl may be further substituted by the groups consisting of:
1) hydroxy,
2) alkoxy,
3) alkyl,
4) -OC(0)R4, 5) -OC(0)NR5R6,
6) -C(0)R\
7) -C(0)NR5R6,
8) -NR5R6,
9) -NR2C(0)R4,
10) -NR2S(0)nR4,
11) -NR2C(0)NR5R6;
Y is selected from :
Figure imgf000007_0001
Z is selected from :
Figure imgf000007_0002
Wherein Y-Z-W is selected from :
Figure imgf000007_0003
X1 and X2 are independently selected from hydrogen, halogen n is an integer from 0 to 2;
m is an integer from 0 to 2;
m' is an integer from 0 to 2; W is independently selected from :
1) alkyl,
2) aralkyl,
3) heteroaralkyl,
4) -OR3,
5) -OC(0)R4,
6) -OC(0)NR5R6,
7) -CH20-R4,
8) -NR5R6,
9) -NR2C(0)R4,
10) -l\IR2S(0)nR4,
11) -NR2C(0)NR5R6; wherein the alkyl, aralkyl and heteroaralkyl may be further substituted; R2 is selected from hydrogen or alkyl;
R3 is selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted carbocyclyl, substituted or unsubstitutded heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl or substituted or unsubstituted heteroaralkyl;
R4 is selected from substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted carbocyclyl, substituted or unsubstitutded heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyl or substituted or unsubstituted heteroaraikyl ;
R5 and R6 are independently selected from hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, carbocyclyl, heterocyclyl, aryl, heteroaryl or R5 and R6 can be fused to form a 3 to 8 membered heterocyclyl ring system.
Preferred embodiment includes compounds of Formula 1 where W is selected from -OR3 and R3 is selected from substituted or unsubstituted aralkyl, or substituted or unsubstituted heteroaraikyl.
Preferred embodiment includes compounds of Formula 1 where W is selected from the group consisting of:
Figure imgf000009_0001
Figure imgf000010_0001
Preferred embodiment includes compounds of Formula 1 where Rxis selected from the group consisting of:
Figure imgf000010_0002
Figure imgf000011_0001
Preferred embodiment includes compounds of Formula 1 where Y is selected from the group consisting of:
Figure imgf000011_0002
Preferred embodiment includes compounds of Formula 1 where Z is selected from the group consisting of:
Figure imgf000011_0003
More preferred embodiment includes compounds of Formula 1 where W is selected from the group consisting of:
Figure imgf000011_0004
More preferred embodiment includes compounds of Formula 1 where R1 is selected from the group consisting of:
Figure imgf000012_0001
More preferred embodiment includes compounds of Formula 1 where Z is selected from the group consisting of:
Figure imgf000012_0002
More preferred embodiment includes compounds of Formula 1 where Y-2-W is selected from the group consisting of:
Figure imgf000012_0003
Another aspect of the present invention provides a pharmaceutical composition comprising an effective amount of a compound of Formula 1 and a pharmaceutically acceptable carrier, diluent or excipient.
In another aspect of the present invention, there is provided a use of the compound of Formula 1 as an inhibitor of protein kinase, more particularly, as an inhibitor of Btk. Another aspect of the present invention provides a method of modulating kinase function, the method comprising contacting a cell with a compound of the present invention in an amount sufficient to modulate the enzymatic activity of a given kinase or kinases, such as Btk, thereby modulating the kinase function.
Another aspect of the present invention provides a method of modulating the target kinase function, the method comprising a) contacting a cell with a compound of the present invention in an amount sufficient to modulate the target kinase function, thereby b) modulating the target kinase activity and signaling.
Another aspect of the present invention provides a probe, the probe comprising a compound of Formula 1 labeled with a detectable label or an affinity tag. In other words, the probe comprises a residue of a compound of Formula 1 covalently conjugated to a detectable label. Such detectable labels include, but are not limited to, a fluorescent moiety, a
chemiluminescent moiety, a paramagnetic contrast agent, a metal chelate, a radioactive isotope-containing moiety, or biotin.
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
The present invention relates to novel kinase inhibitors. These compounds are found to have activity as inhibitors of protein kinases: including members of the tyrosine kinases Aurora, SRC (more specifically Lck) and Tec (more specifically Btk) kinase families.
Compounds of the present invention may be formulated into a
pharmaceutical composition which comprises an effective amount of a compound of Formula 1 with a pharmaceutically acceptable diluent or carrier. For example, the pharmaceutical compositions may be in a conventional pharmaceutical form suitable for oral administration (e.g., tablets, capsules, granules, powders and syrups), parenteral administration (e.g., injections (intravenous, intramuscular, or subcutaneous)), drop infusion preparations, inhalation, eye lotion, topical administration (e.g., ointment), or suppositories. Regardless of the route of administration selected the compounds may be formulated into pharmaceutically acceptable dosage forms by conventional methods known to those skilled in the art.
The phrase "pharmaceutically acceptable" is employed herein to refer to those ligands, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
The phrase "pharmaceutically acceptable carrier" as used herein means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material.
Each carrier must be acceptable in the sense of being compatible with the other ingredients of the formulation, including the active ingredient, and not injurious or harmful to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch, potato starch, and substituted or unsubstituted β-cyclodextrin; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; ( 19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic
compatible substances employed in pharmaceutical formulations.
The term "pharmaceutically acceptable salt" refers to the relatively non-toxic, inorganic and organic acid addition salts of the compound(s). These salts can be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting a purified compound(s) in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, laurylsulphonate salts, and amino acid salts, and the like (See, for example, Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66 : 1-19).
In other cases, the compounds of the present invention may contain one or more acidic functional groups and, thus, are capable of forming
pharmaceutically acceptable salts with pharmaceutically acceptable bases. The term "pharmaceutically acceptable salts" in these instances refers to the relatively non-toxic inorganic and organic base addition salts of a
compound(s). These salts can likewise be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting the purified compound(s) in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine,
ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, for example, Berge et al., supra).
As used herein, the term "affinity tag" means a ligand or group, linked either to a compound of the present invention or to a protein kinase domain, that allows the conjugate to be extracted from a solution.
The term "alkyl" refers to substituted or unsubstituted saturated hydrocarbon groups, including straight-chain alkyl and branched-chain alkyl groups, including haloalkyl groups such as trifluoromethyl and 2,2,2-trifluoroethyl, etc. Representative alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, (cyclohexyl)methyl, cyclopropylmethyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like. The terms "alkenyl" and "alkynyl" refer to substituted or unsubstituted unsaturated aliphatic groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond respectively.
Representative alkenyl groups include vinyl, propen-2-yl, crotyl, isopenten-2- yl, l,3-butadien-2-yl), 2,4-pentadienyl, and l,4-pentadien-3-yl.
Representative alkynyl groups include ethynyl, 1- and 3-propynyl, and 3- butynyl. In certain preferred embodiments, alkyl substituents are lower alkyl groups, e.g., having from 1 to 6 carbon atoms. Similarly, alkenyl and alkynyl preferably refer to lower alkenyl and alkynyl groups, e.g., having from 2 to 6 carbon atoms. As used herein, "alkylene" refers to an alkyl group with two open valencies (rather than a single valency), such as -(CH2)i-io- and substituted variants thereof.
The term "alkoxy" refers to an alkyl group having an oxygen attached thereto. Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like. An "ether" is two hydrocarbons covalently linked by an oxygen. Accordingly, the substituent of an alkyl that renders that alkyl an ether is or resembles an alkoxy.
The term "alkoxyalkyl" refers to an alkyl group substituted with an alkoxy group, thereby forming an ether.
The terms "amide" and "amido" are art-recognized as an amino-substituted carbonyl and includes a moiety that can be represented by the general formula :
Figure imgf000016_0001
wherein R9, R10 are as defined above. Preferred embodiments of the amide will not include imides, which may be unstable.
The terms "amine" and "amino" are art-recognized and refer to both unsubstituted and substituted amines and salts thereof, e.g., a moiety that can be represented by the general formulae:
Figure imgf000017_0001
wherein R9, R10 and R10' each independently represent a hydrogen, an alkyl, an alkenyl, -(CH2)m-R8, or R9 and R10 taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; R8 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocyclyl or a polycyclyl; and m is zero or an integer from 1 to 8. In preferred embodiments, only one of R9 or R10 can be a carbonyl, e.g., R9, R10, and the nitrogen together do not form an imide. In even more preferred embodiments, R9 and R10 (and optionally R10') each independently represent a hydrogen, an alkyl, an alkenyl, or -(CH2)m-R8. In certain embodiments, the amino group is basic, meaning the protonated form has a pKa >. 7.00.
The term "aralkyl", as used herein, refers to an alkyl group substituted with an aryl group, for example -(CH2)n-Ar.
The term "heteroaralkyl", as used herein, refers to an alkyl group substituted with a heteroaryl group, for example -(CH2)n-Het.
The term "aryl" as used herein includes 5-, 6-, and 7-membered substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon. The term "aryl" also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, anthracene, and phenanthrene.
The terms "carbocycle" and "carbocyclyl", as used herein, refer to a non- aromatic substituted or unsubstituted ring in which each atom of the ring is carbon. The terms "carbocycle" and "carbocyclyl" also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is
carbocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Representative carbocyclic groups include cyclopentyl, cyclohexyl, 1-cyclohexenyl, and 3- cyclohexen-l-yl, cycloheptyl.
The term "carbonyl" is art-recognized and includes such moieties as can be represented by the general formula :
Figure imgf000018_0001
wherein X is a bond or represents an oxygen or a sulfur, and R11 represents a hydrogen, an alkyl, an alkenyl, -(CH2)m-R8 or a pharmaceutically acceptable salt. Where X is an oxygen and R11 is not hydrogen, the formula represents an "ester". Where X is an oxygen, and R11 is a hydrogen, the formula represents a "carboxylic acid".
The terms "heteroaryl" includes substituted or unsubstituted aromatic 5- to 7-membered ring structures, more preferably 5- to 6-membered rings, whose ring structures include one to four heteroatoms. The term
"heteroaryl" also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, isoxazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. The term "heteroatom" as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen, and sulfur.
The terms "heterocyclyl" or "heterocyclic group" refer to substituted or unsubstituted non-aromatic 3- to 10-membered ring structures, more preferably 3- to 7-membered rings, whose ring structures include one to four heteroatoms. The term terms "heterocyclyl" or "heterocyclic group" also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
Heterocyclyl groups include, for example, tetrahydrofuran, tetrahydropyran, piperidine, piperazine, pyrrolidine, morpholine, lactones, and lactams.
The term "hydrocarbon", as used herein, refers to a group that is bonded through a carbon atom that does not have a =0 or =S substituent, and typically has at least one carbon-hydrogen bond and a primarily carbon backbone, but may optionally include heteroatoms. Thus, groups like methyl, ethoxyethyl, 2-pyridyl, and trifluoromethyl are considered to be hydrocarbyl for the purposes of this application, but substituents such as acetyl (which has a =0 substituent on the linking carbon) and ethoxy (which is linked through oxygen, not carbon) are not. Hydrocarbyl groups include, but are not limited to aryl, heteroaryl, carbocycle, heterocycle, alkyl, alkenyl, alkynyl, and combinations thereof.
The terms "polycyclyl" or "polycyclic" refer to two or more rings (e.g., cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or
heterocyclyls) in which two or more carbons are common to two adjoining rings, e.g., the rings are "fused rings". Each of the rings of the polycycle can be substituted or unsubstituted.
As used herein, the term "probe" means a compound of the invention which is labeled with either a detectable label or an affinity tag, and which is capable of binding, either covalently or non-covalently, to a protein kinase domain. When, for example, the probe is non-covalently bound, it may be displaced by a test compound. When, for example, the probe is bound covalently, it may be used to form cross-linked adducts, which may be quantified and inhibited by a test compound.
The term "substituted" refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that "substitution" or "substituted with" includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term "substituted" is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Substituents can include, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that the moieties substituted on the hydrocarbon chain can themselves be substituted, if appropriate. Compounds of the invention also include all isotopes of atoms present in the intermediates and/or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include deuterium and tritium.
General Synthetic Methods
General Synthetic Method A:
Ulmann condensation of phenol 1-i with ester 1-ii provided intermediate 1-iii. Saponification of intermediate 1-iii yielded intermediate 1-iv. Conversion of intermediate 1-iv to its acid chloride, using for example oxalyl chloride and DMF, provided intermediate 1-v. Condensation of intermediate 1-v with malononitrile yielded intermediate 1-vi. Methylation of intermediate 1-vi with TMS-diazomethane provided intermediate 1-vii. Condensation of 1-vii with hydrazine yielded intermediate 1-viii. Condensation of intermediate 1-viii with formamidine yielded intermediate 1-ix. Intermediate 1-ix was treated with alcohol F^OH, under Mitsunobu conditions, to provide the desired compounds or intermediates of general formula 1-x.
Figure imgf000022_0001
Figure imgf000022_0002
Figure imgf000022_0003
Figure imgf000022_0004
Scheme 1
General Synthetic Method B:
Benzoyl chlorides of formula 2-i were condensed with malononitrile to provide intermediate 2-ii. Methylation of intermediate 2-ii with TMS- diazomethane provided intermediate 2-iii. Condensation of intermediate 2-iii with hydrazine provided intermediate 2-iv. Further condensation of intermediate 2-iv with formamidine provided intermediate 2-v. Intermediate 2-v was treated with alcohol F^OH, under Mitsunobu conditions, to provide intermediate 2-vi. Ullmann condensation of intermediate 2-vi with phenolic intermediates 2-vii provided the desired compounds or intermediates of general formula 2-viii.
Figure imgf000023_0001
2-i; X=l, Br 2-ii; X=l, Br X=l, Br 2-iv; X=l, Br
Figure imgf000023_0002
2-v; X=l, Br 2-vi; X=l, Br
Figure imgf000023_0003
2-vii 2-viii
Scheme 2a Alternatively, intermediate 2-iv was treated with an alcohol of formula RiOH, under Mitsunobu conditions to provide intermediate 2-ix. Condensation of intermediate 2-ix with formamidine provided intermediate 2-vi.
Figure imgf000024_0001
2-iv; X=l, Br 2-ix; X=l, Br 2-vi; X=l, Br
Scheme 2b
In a similar manner, condensation of intermediate 2-iii with a hydrazine of formula RiNHNH2 provided intermediate 2-ix. Ullmann condensation of intermediate 2-ix with phenolic intermediates 2-vii provided intermediates 2- x. Condensation of intermediate 2-x with formamidine provided the desired compounds or intermediates of general formula 2-viii.
Figure imgf000025_0001
Figure imgf000025_0002
Alternatively, trimethyl orthoformate and ammonia can be used in place of formamidine, for example, in the conversion of intermediate 2-x to compounds of formula 2-viii.
Exemplification
The following synthetic methods are intended to be representative of the chemistry used to prepare compounds of Formula 1 and are not intended to be limiting.
Synthesis of Compound 1:
Figure imgf000026_0001
Figure imgf000026_0002
3-g Compound 1
Scheme 3
Step 1: Intermediate 3-a
Benzyl bromide (27.0 ml, 227 mmol) was added drop wise to a stirred suspension of resorcinol (25.0 g, 227 mmol) and potassium carbonate (31.4 g, 227 mmol) in acetone (150 ml) and the reaction was heated under reflux overnight. Volatiles were removed under reduced pressure. Water and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 3-a as a beige oil.
Step 2: Intermediate 3-b
To a solution of compound 3-a (15.0 g, 74.9 mmol) in 1,4-dioxane (200 ml) were sequentially added ethyl 4-bromobenzoate (20.59 g, 90 mmol), N,N- dimethylglycine (4.25 g, 41.2 mmol), copper(I) chloride(3.71 g, 37.5 mmol) and cesium carbonate (61.0 g, 187 mmol). The reaction mixture was stirred at reflux overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, washed with saturated aqueous NaHC03, brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 3-b as a colorless oil.
Step 3: Intermediate 3-c
To a solution of intermediate 3-b (17.5 g, 50.2 mmol) in THF (200 ml) and MeOH (100 ml) was added 2N sodium hydroxide (100 ml, 200 mmol) and the reaction was stirred at room temperature overnight. Volatiles were removed under reduced pressure. 10% aqueous HCI and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 3-c as beige solid.
Step 4: Intermediate 3-d
To a suspension of intermediate 3-c (16.1 g, 50.3 mmol) in dichloromethane ( 100 ml) were added DMF (0.1 ml, 1.29 mmol) and oxalyl chloride (4.4 ml, 50.3 mmol). The solution was stirred at room temperature for 2 hours.
Volatiles were removed under reduced pressure to provide intermediate 3-d as beige solid. Step 5: Intermediate 3-e
To a solution of intermediate 3-d (16.5 g, 48.9 mmol) in toluene (50 ml) and THF (7 ml), cooled to -10 °C, were added malononitrile (3.19 ml, 50.2 mmol) and DIPEA (17.5 ml, 100 mmol) in toluene (50 ml_), drop wise, over a period of 30 minutes. After the addition was completed, the reaction was stirred for 1 hour at 0° C and room temperature overnight. Volatiles were removed under reduced pressure. 1M aqueous HCI and ethyl acetate were added, the organic layer was separated, washed with 1M HCI and brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 3-e as beige solid.
Step 6: Intermediate 3-f
To a solution of intermediate 3-e ( 18.1 g, 49.1 mmol) in acetonitrile (177 ml) and methanol (19.0 ml), cooled to 0°C, were added DIPEA ( 10.3 ml, 59.0 mmol) and a 2M solution of (diazomethyl)trimethylsilane in hexanes (27.0 ml, 54.0 mmol). After the addition was completed, the reaction was stirred at room temperature overnight. Acetic acid (0.56 ml, 9.83 mmol) was added, the reaction was then stirred for 30 minutes and volatiles were removed under reduced pressure. A saturated aqueous solution of NaHC03 and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure.
Purification by silica gel chromatography provided intermediate 3-f as yellow solid.
Step 7: Intermediate 3-g
To a suspension of intermediate 3-f (8.05 g, 21.1 mmol) in ethanol (10.5 ml) was added a solution of hydrazine monohydrate (2.76 ml, 56.8 mmol). The reaction was stirred at 100 °C for 1 hour and then cooled to room
temperature. Water was added; a precipitate formed and was collected by filtration, washed with diethyl ether and dried in vacuo to provide
intermediate 3-g as an off-white solid.
Step 8: Compound 1
Intermediate 3-g (8.0 g, 20.92 mmol) was added to a solution of
formamidine (58.4 ml, 1464 mmol) and the reaction was stirred at 180 °C for 2 hours and then cooled to room temperature. Water and ethyl acetate were added; the organic layer was separated, washed with brine, dried over MgS0 , filtered and concentrated under reduced pressure to provide compound 1 as beige solid. MS (m/z) M + H=410.2
Synthesis of Compound 2:
Figure imgf000029_0001
Scheme 4
To a solution of cyclopentanol (316 mg, 3.66 mmol) in THF was added triphenylphosphine (961 mg, 3.66 mmol) and DIAD (712 μΙ, 3.66 mmol). The yellow solution was stirred for 5 minutes, compound 1 (1.0 g, 2.44 mmol) was added and the reaction was then stirred at room temperature overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided compound 2 as an off-white solid. MS (m/z) M + H=478.2
Synthesis of Compound 3:
Figure imgf000030_0001
Compound 3
Scheme 5
To a solution of (S)-tert-butyl 3-hydroxypiperidine-l-carboxylate (5.65 g, 28.1 mmol) in THF was added triphenylphosphine (7.37 g, 28.1 mmol) and DIAD (5.46 ml, 28.1 mmol). The yellow solution was stirred for 5 minutes, compound 1 ( 10.0 g, 24.42 mmol) was added and the reaction was then stirred at room temperature overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided compound 3 as a white foam. MS (m/z) M + H= 593.1
Synthesis of Compound 4:
Figure imgf000030_0002
Compound 4
Compound 3
Scheme 6 To a solution of compound 3 (1.88 g, 3.17 mmol) in dichloromethane was added 4N HQ in 1,4-dioxane (19.82 ml, 79.0 mmol) and the reaction was stirred at room temperature for 2 hours. Volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 4-2HCI as a white solid. MS (m/z) M + H= 493.1
Synthesis of compound 5
Figure imgf000031_0001
Compound 4 Compound 5
Scheme 7
To a solution of compound 4 2HCI (100 mg, 0.17 mmol) in dichloromethane (2 ml) cooled to 0°C were sequentially added TEA (99 μΙ, 0.70 mmol) and acryloyl chloride (17.6 mg, 0.19 mmol). The reaction was stirred at 0°C for 1 hour. A saturated aqueous solution of ammonium chloride was added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided compound 5 as a white solid. MS (m/z) M + H = 547.1
Synthesis of compound 6
Figure imgf000032_0001
Compound 4 Compound 6
Scheme 8
To a solution of compound 4 2HCI (1.8 g, 3.18 mmol) in dichloromethane (32 ml) cooled to 0°C were sequentially added TEA (1.77 ml, 12.73 mmol) and acetyl chloride (249 μΙ, 3.50 mmol). The reaction was stirred at 0°C for 1 hour and room temperature overnight. A saturated aqueous solution of ammonium chloride was added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with 1% aqueous HCI/methanol gradient provided compound 6-HCI as beige solid. MS (m/z) M + H= 535.1
Synthesis of intermediate 9-d
Figure imgf000033_0001
Figure imgf000033_0002
9-c 9-d
Scheme 9
Step 1: Intermediate 9-a
To a solution of 4-bromobenzoyl chloride (25.0 g, 114 mmol) in toluene (200 ml) and THF (30 ml), cooled to -10 °C, were sequentially added malononitrile (7.60 ml, 120.0 mmol) and DIPEA (39.8 ml, 228 mmol) in toluene (50 ml_) drop wise over a period of 1 hour. After the addition was completed, the reaction was stirred for 1 hour at 0°C and room temperature overnight.
Volatiles were removed under reduced pressure. IN HCI and ethyl acetate were added to the residue, the organic layer was separated, washed twice with IN HCI and brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 9-a as yellow solid.
Step 2: Intermediate 9-b
To a solution of intermediate 9-a (26.4 g, 106 mmol) in acetonitrile (300 ml) and methanol (35.0 ml), cooled to 0°C, was added DIPEA (22.2 ml, 127 mmol) and a 2M solution of diazomethyhtrimethylsilane in hexanes (58.3 ml, 117 mmol). After the addition was completed, the reaction was stirred at room temperature overnight. Acetic acid (1.21 ml, 21.2 mmol) was added, the reaction was stirred for 30 minutes and volatiles were removed under reduced pressure. A saturated aqueous solution of NaHC03 and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 9-b as a yellow solid.
Step 3: Intermediate 9-c
To a suspension of intermediate 9-b (4.49 g, 17.07 mmol) in ethanol (8.5 ml) was added a solution of hydrazine monohydrate (2.23 ml, 46.1 mmol) and the reaction was stirred at 100 °C for 1 hour and then cooled to room temperature. Volatiles were removed under reduced pressure to provide intermediate 9-c as a yellow solid.
Step 4: Intermediate 9-d
Intermediate 9-c (4.49 g, 17.07 mmol) was added to a solution of
formamidine (40.8 ml, 1024 mmol) and the reaction was stirred at 180 °C for 3 hours and then cooled to room temperature. Ethanol was added; a precipitate formed and was collected by filtration, dried in vacuo to provide intermediate 9-d as a beige solid.
Synthesis of intermediate 10-a
Figure imgf000034_0001
Scheme 10
To a solution of intermediate 9-d (1.0 g, 3.45 mmol) in THF was added triphenylphosphine (1.35 g, 5.17 mmol), cyclopentanol (0.47 ml, 5.17 mmol) and DIAD ( 1.0 ml, 5.17 mmol) and the reaction was then stirred at room temperature overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 10-a as white solid. MS (m/z) M + H= 359.6
Synthesis of Compound 9
Figure imgf000035_0001
Scheme 11
Step 1: Intermediate 11-a To a solution of resorcinol (15.0 g, 136 mmol) in DMF (100 ml), cooled to 0°C, were added imidazole (19.48 g, 286 mmol) and tert- butylchlorodimethylsilane (21.56 g, 143 mmol). The reaction was then stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added; the organic layer was separated, washed 3 times with a saturated aqueous solution of ammonium chloride and brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 11-a as a colorless oil.
Step 2: Intermediate 11-b
To a solution of (4-chlorophenyl) methanol ( 1.52 g, 10.70 mmol) in THF (20 ml_) were sequentially added intermediate 11-a (2.88 g, 12.84 mmol), triphenylphosphine (3.37 g, 12.84 mmol) and DIAD (2.53 ml, 12.84 mmol) drop wise at room temperature and the reaction was then stirred for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 11-b as a colorless oil.
Step 3: Intermediate 11-c
Tetrabutylammonium fluoride trihydrate (3.93 g, 12.47 mmol) was added to a solution of intermediate 11-b (2.9 g, 8.31 mmol) in THF (15 ml.) and the reaction was stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 , filtered and concentrated under reduced pressure. Purification by silica gel
chromatography provided intermediate 11-c as a colorless oil.
Step 4: Compound 9 A solution of intermediate 10-a (200 mg, 0.56 mmol), intermediate 11-c (229 mg, 0.977 mmol), quinolin-8-ol (16.21 mg, 0.112 mmol), copper (I) chloride (11.05 mg, 0.11 mmol) and cesium carbonate (546 mg, 1.67 mmol), in dimethylacetamide ( 1 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 9-HCI as a yellow solid. MS (m/z) M + H= 512.2
Synthesis of intermediate -a
Figure imgf000037_0001
12-a
Scheme 12
To a solution of intermediate 9-d (500 mg, 1.72 mmol) in THF (8.6 mL), were sequentially added methanol (105 μΙ, 2.59 mmol), triphenylphosphine (678 mg, 2.59 mmol) and DIAD (503 μΙ, 2.59 mmol) drop wise at room temperature. The solution was then stirred at room temperature overnight. A precipitate formed and was collected by filtration, dried in vacuo to provide intermediate 12-a as a white solid.
Synthesis of compound 16
Figure imgf000038_0001
Scheme 13
A solution of intermediate 12-a (235 mg, 0.77 mmol), intermediate 3-a (271 mg, 1.35 mmol), quinolin-8-ol (22.4 mg, 0.15 mmol), copper (I) chloride (15.3 mg, 0.15 mmol) and cesium carbonate (755 mg, 2.31 mmol) in dimethylacetamide (1 ml) was degassed with nitrogen for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with 1% HCI/methanol gradient provided compound 16-HCI as a beige solid. MS (m/z) M+H= 424.2
Synthesis of compound 17
Figure imgf000039_0001
Figure imgf000039_0002
Scheme 14
Step 1: Intermediate 14-a
To a solution of 3-hydroxybenzaldehyde (14.73 g, 121 mmol) in
dichloromethane ( 100 mL) were sequentially added triethylamine (25.08 ml, 181 mmol), tert-butylchlorodimethylsilane (20.0 g, 133 mmol) portion wise, and the reaction was stirred at room temperature overnight. 10% aqueous citric acid was added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure.
Purification by silica gel chromatography provided intermediate 14-a as a yellow oil.
Step 2: Intermediate 14-b To a solution of intermediate 14-a (16.0 g, 67.7 mmol) in methanol (100 ml) cooled to 0°C was added portion wise sodium borohydride (1.28 g, 33.8 mmol). After the addition was completed the reaction was stirred at room temperature for 2 hours. Volatiles were removed under reduced pressure. Water and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 14-b as a yellow oil.
Step 3: Intermediate 14-c
To a solution of intermediate 14-b ( 1.0 g, 2.09 mmol) in THF (42 ml_) were sequentially added 2-hydroxybenzonitrile (600 mg, 5.03 mmol),
triphenylphosphine (1.32 g, 5.03 mmol) and DIAD (991 μΙ, 5.03 mmol) drop wise at room temperature; the reaction was stirred at reflux for 2 hours and then cooled to room temperature. A saturated aqueous solution of
ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 14-c as a colorless oil.
Step 2: Intermediate 14-d
To a solution of intermediate 14-c (1.22 g, 3.62 mmol) in THF (36.0 ml) was added tetrabutylammonium fluoride trihydrate (946 mg, 3.62 mmol) and the reaction was stirred at room temperature for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel
chromatography provided intermediate 14-d as a white solid.
Step 2: Compound 17 A solution of intermediate 12-a (200 mg, 0.6 mmol), intermediate 14-d (259 mg, 1.15 mmol), quinolin-8-ol (19.0 mg, 0.13 mmol), copper (I) chloride (13.0 mg, 0.13 mmol) and cesium carbonate (643 mg, 1.97 mmol) in dimethylacetamide (3.0 ml) was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight. After cooling to room temperature, water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided compound 17 as a white solid. MS (m/z) M + H= 449.3
Synthes of compound 18:
Figure imgf000041_0001
Scheme 15
Step 1: Intermediate 15-a
To a solution of ethyl 2-methylthiazole-5-carboxylate (5.82 g, 34.0 mmol) in THF (170 ml), cooled to 0 °C, was added a 1.0M solution of LiAIH4 in THF (34.0 ml, 34.0 mmol) and the reaction was slowly warmed to room
temperature and stirred overnight. Water (1.3 ml) was slowly added, followed by 15% NaOH (1.3 ml_). The solution was stirred for 2 hours at room temperature then filtered over celite. The filtrate was concentrated under reduced pressure to provide intermediate 15-a as a yellow oil.
Step 2: Intermediate 15-b
To a solution of intermediate 15-a (7.75 g, 34.5 mmol) and intermediate 11- a (4.25 g, 32.9 mmol), in THF (33 ml_), were sequentially added
triphenylphosphine (10.35 g, 39.5 mmol) and DIAD (7.68 ml, 39.5 mmol) drop wise at room temperature. The reaction was then stirred for 18 hours. Volatiles were removed in vacuo. Purification by silica gel chromatography provided intermediate 15-b as a colorless oil.
Step 3: Intermediate 15-c
To a solution of intermediate 15-b (5.5 g, 16.39 mmol), in THF (82.0 ml), was added a l.OM solution of tetrabutylammonium fluoride in THF (16.4 ml, 16.4 mmol) and the reaction was stirred at room temperature for 30 minutes. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure.
Purification by silica gel chromatography provided intermediate 15-c as beige solid.
Step 4: Compound 18
A solution of intermediate 12-a (200 mg, 0.65 mmol), intermediate 15-c (146 mg, 0.65 mmol), quinolin-8-ol (19.0 mg, 0.13 mmol), copper (I) chloride ( 13.0 mg, 0.13 mmol) and cesium carbonate (643 mg, 1.97 mmol) in dimethylacetamide (6.5 ml) was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C for 2 hours and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS0 , filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with 1% HCI/methanol gradient provided compound 18-2HCI as a beige solid. MS (m/z) M + H= 445.1
Synthesis of compound 15:
Figure imgf000043_0001
Scheme 16
A solution of intermediate 10-a (200 mg, 0.56 mmol), intermediate 14-d (156 mg, 0.68 mmol), quinolin-8-ol (16.2 mg, 0.11 mmol), copper (I) chloride ( 11.0 mg, 0.11 mmol) and cesium carbonate (546 mg, 1.67 mmol) in dimethylacetamide (5.5 ml) was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with 1% HCI/methanol gradient provided compound 15-HCI as beige solid. MS (m/z) M + H= 503.3
Synthesis of intermediate 17-a
Figure imgf000044_0001
Scheme 17
To a solution of intermediate 9-d (650 mg, 2.24 mmol), in THF (22.0 mL), were sequentially added tetrahydro-2H-pyran-4-ol (320 μΙ, 3.36 mmol), triphenylphosphine (881 mg, 3.36 mmol) and DIAD (653 μΙ, 3.36 mmol) drop wise at room temperature. The solution was then stirred at 50°C overnight. Volatiles were removed in vacuo. Purification by silica gel chromatography provided intermediate 17-a as a white solid.
Synthesis of compound 22:
Figure imgf000044_0002
Scheme 18
A solution of intermediate 17-a (200 mg, 0.53mmol), intermediate 14-d (181 mg, 0.80 mmol), quinolin-8-ol (15.5 mg, 0.11 mmol), copper (I) chloride ( 11.5 mg, 0.11 mmol) and cesium carbonate (348 mg, 1.07 mmol) in dimethylacetamide (5.3 ml) was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with 1% HCI/methanol gradient provided compound 22-HCI as a beige solid. MS (m/z) M + H= 519.2
Synthesis of compound 31:
Figure imgf000045_0001
Scheme 19
Step 1: Intermediate 19-a
To a solution of intermediate 14-b ( 10.0 g, 41.9 mmol) in THF (210 ml_) were sequentially added 2-(trifluoromethyl)phenol (6.80 g, 41.9 mmol), triphenylphosphine (13.2 g, 50.33 mmol) and DIAD (9.79 ml, 50.3 mmol) drop wise at room temperature. The reaction was then stirred at room temperature overnight. Saturated aqueous ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 19-a as a colorless oil.
Step 2: Intermediate 19-b
To a solution of intermediate 19-a (13.9 g, 36.3 mmol) in THF (182.0 ml) was added a l.OM solution of tetrabutylammonium fluoride in THF (36.3 ml, 36.3 mmol) and the reaction was stirred at room temperature for 1 hour. Saturated aqueous ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 19-b as colorless oil.
Step 3: Compound 31
A solution of intermediate 12-a (200 mg, 0.66 mmol), intermediate 19-b (265 mg, 0.98 mmol), quinolin-8-ol (19.0 mg, 0.13 mmol), copper (I) chloride (25.5 mg, 0.13 mmol) and cesium carbonate (429 mg, 1.31 mmol) in dimethylacetamide (6.5 ml) was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with 1% HCI/methanol gradient provided compound 31-HCI as white solid. MS (m/z) M+H= 492.1
Synthesis of compound 32:
Figure imgf000047_0001
Scheme 20
A solution of intermediate 10-a (200 mg, 0.55 mmol), intermediate 19-b (225 mg, 0.83 mmol), quinolin-8-ol (16.2 mg, 0.11 mmol), copper (I) iodide (22.0 mg, 0.11 mmol) and cesium carbonate (364 mg, 1.17 mmol) in dimethylacetamide (5.5 ml) was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with 1% HCI/methanol gradient provided compound 32-HCI as beige solid. MS (m/z) M + H= 546.1
Synthesis of compound 36:
Figure imgf000047_0002
Scheme 21 A solution of intermediate 17-a (200 mg, 0.53 mmol), intermediate 19-b (215 mg, 0.80 mmol), quinolin-8-ol (15.5 mg, 0.11 mmol), copper (I) iodide (20.3 mg, 0.11 mmol) and cesium carbonate (348 mg, 1.06 mmol) in dimethylacetamide (5.3 ml) was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with 1% HCI/methanol gradient provided compound 36-HCI as beige solid. MS (m/z) M+H= 562.2
Synthesis of compound 20:
Figure imgf000048_0001
Scheme 22
Step 1: Intermediate 22-a
To a solution of 2-(trifluoromethyl)phenylmethanol (1.43 g, 8.10 mmol) in THF (8.10 ml.) were sequentially added intermediate 11-a (2.0 g, 8.91 mmol), triphenylphosphine (2.55 g, 9.72 mmol) and DIAD ( 1.89 ml, 9.72 mmol) drop wise at room temperature. The reaction was then stirred overnight at room temperature. Saturated aqueous ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS0 / filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 22-a as a colorless oil.
Step 2: Intermediate 22-b
Tetrabutylammonium fluoride trihydrate (1.81 g, 5.75 mmol) was added to a solution of intermediate 22-a (2.2 g, 5.75 mmol) in THF (23 ml_) and the reaction was stirred at room temperature for 1 hour. Saturated aqueous ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 22-b as a colorless oil.
Step 3: Compound 20
A solution of intermediate 12-a (200 mg, 0.65 mmol), intermediate 22-b (309 mg, 1.15 mmol), quinolin-8-ol (19.1 mg, 0.13 mmol), copper (I) chloride ( 13.0 mg, 0.13 mmol) and cesium carbonate (429 mg, 1.31 mmol), in dimethylacetamide (6.5 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 20-HCI as beige solid. MS (m/z) M + H= 492.1
Synthesis of compound
Figure imgf000050_0001
Scheme 23
A solution of intermediate 10-a (200 mg, 0.55 mmol), intermediate 22-b (225 mg, 0.83 mmol), quinolin-8-ol ( 16.2 mg, 0.11 mmol), copper (I) iodide (21.2 mg, 0.11 mmol) and cesium carbonate (364 mg, 1.11 mmol), in dimethylacetamide (5.5 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 29-HCI as beige solid. MS (m/z) M + H= 546.2
Synthesis of compound 23:
Figure imgf000050_0002
Scheme 24 A solution of intermediate 17-a (200 mg, 0.53 mmol), intermediate 22-b (215 mg, 0.80 mmol), quinolin-8-ol (15.5 mg, 0.11 mmol), copper (I) iodide (20.4 mg, 0.11 mmol) and cesium carbonate (348 mg, 1.07 mmol), in dimethylacetamide (5.3 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 23-HCI as beige solid. MS (m/z) M + H= 562.1
Synthesis of compound 30:
Figure imgf000051_0001
Figure imgf000051_0002
Scheme 25
Step 1: Intermediate 25-a
To a solution of 2-(bromomethyl)benzonitrile (1.0 g, 5.10 mmol) and resorcinol (2.81 g, 25.5 mmol) in acetone (51.0 mL) was added cesium carbonate (3.32 g, 10.20 mmol) and the reaction was then stirred at reflux for 2 hours. Volatiles were removed under reduced pressure. Saturated aqueous ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and
concentrated under reduced pressure. Purification by silica gel
chromatography provided intermediate 25-a as white solid.
Step 2: Compound 30
A solution of intermediate 12-a (200 mg, 0.65 mmol), intermediate 25-a (222 mg, 0.98 mmol), quinolin-8-ol (19.1 mg, 0.13 mmol), copper (I) iodide (25.0 mg, 0.13 mmol) and cesium carbonate (429 mg, 1.31 mmol), in dimethylacetamide (6.5 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 30-HCI as beige solid. MS (m/z) M + H= 449.4
Synthesis of compound 12:
Figure imgf000052_0001
Scheme 26 A solution of intermediate 10-a (200 mg, 0.55 mmol), intermediate 25-a (220 mg, 0.98 mmol), quinolin-8-ol (16.2 mg, 0.11 mmol), copper (I) chloride (11.0 mg, 0.11 mmol) and cesium carbonate (546 mg, 1.67 mmol), in dimethylacetamide (5.5 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 12-HCI as a beige solid. MS (m/z) M + H= 503.2
Synthesis of compound 35:
Figure imgf000053_0001
Scheme 27
A solution of intermediate 17-a (200 mg, 0.55 mmol), intermediate 25-a (181.0 mg, 0.80 mmol), quinolin-8-ol ( 15.5 mg, 0.11 mmol), copper (I) iodide (20.3 mg, 0.11 mmol) and cesium carbonate (348 mg, 1.07 mmol), in dimethylacetamide (5.3 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS0 , filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 35-HCI as beige solid. MS (m/z) M + H= 519.2
Synthesis of compound 10:
Figure imgf000054_0001
Scheme 28
Step 1: Intermediate 28-a
To a solution of (3-methoxyphenyl)methanol (1.38 g, 10.0 mmol) in THF (20.0 mL) were sequentially added intermediate 11-a (2.69 g, 12.0 mmol), triphenylphosphine (3.15 g, 12.0 mmol) and DIAD (2.36 ml, 12.0 mmol) drop wise at room temperature and the reaction was then stirred overnight at room temperature. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 28-a as a colorless oil. Step 2: Intermediate 28-b
Tetrabutylammonium fluoride trihydrate (2.88 g, 9.14 mmol) was added to a solution of intermediate 28-a (2.1 g, 6.10 mmol) in THF (10 mL) and the reaction was stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel
chromatography provided intermediate 28-b as a colorless oil.
Step 3: Compound 10
A solution of intermediate 10-a (200 mg, 0.65 mmol), intermediate 28-b (225 mg, 0.97 mmol), quinolin-8-ol (16.2 mg, 0.11 mmol), copper (I) chloride (11.0 mg, 0.1 mmol) and cesium carbonate (546 mg, 1.67 mmol), in dimethylacetamide (5.5 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 10-HCI as a yellow solid. MS (m/z) M + H= 508.1
Synthesis of intermediate 29-i
Figure imgf000056_0001
29-d 29-h 29-i
Scheme 29
Step 1: Intermediate 29-b
Ethyl chloroacetate, 29-a (50.0 g, 0.41 mol), and ethyl formate (30.2 g, 0.41 mol) were taken in anhydrous toluene (500 ml_) and cooled to 0°C. Sodium ethoxide (35.1 g, 0.49 mol) was added portion wise. The reaction mixture was stirred at 0°C for 5 hours and then at room temperature overnight. The reaction mixture was quenched with water (250 mL) and washed twice with diethyl ether. The aqueous layer was cooled to 0 °C and acidified to pH 4-5 using 1 N HCI. The aqueous layer was extracted twice with diethyl ether; the combined organic layers were dried over MgS04 filtered and concentrated under reduced pressure to provide intermediate 29-b as beige oil. Step 2: Intermediate 29-c
To a solution of ethyl 2-chloro-3-oxopropanoate, 29-b (34.7 g, 230 mmol), in toluene (250 ml) was added thioacetamide (26.0 g, 346.0 mmol), the reaction was stirred at 90°C overnight and then cooled to room temperature, diluted with water (300 mL) and then neutralized to pH 7 with a saturated aqueous solution of NaHC03. Ethyl acetate was added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 29-c as beige oil.
Step 4: Intermediate 29-d
To a solution of intermediate 29-c (22.2 g, 130.0 mmol) in THF (430 ml) cooled to 0°C was added a 1.0 M solution of LiAIH4 in THF (91.0 ml, 91.0 mmol) and the solution was slowly warmed to room temperature and stirred for 2 hours. Water (3.5 ml) was slowly added, followed by 3.5 ml 15% NaOH (3.5 ml) and water (10.5 ml) and the mixture was stirred for 1 hour. The reaction was filtered over celite and volatiles were removed in vacuo to provide intermediate 29-d as yellow oil.
Step 5: Intermediate 29-f
To a solution of l-fluoro-3,5-dimethoxybenzene (12.5 g, 80 mmol) in dichloromethane (80 ml), cooled to 0°C, was added 1.0 M solution of boron tribromide in dichloromethane (200 ml, 200 mmol), drop wise over a period of 30 minutes. The reaction was stirred for 1 hour at 0°C and then slowly warmed to room temperature and stirred for 18 hours. The reaction was cooled to 0°C and quenched by the slow addition of MeOH and water. After stirring at room temperature for 1 hour the mixture was filtered and volatiles were removed in vacuo. Ethyl acetate was added to the residue; a precipitate formed and was collected by filtration to provide intermediate 29-f as an orange solid. Step 6: Intermediate 29-g
To a solution of intermediate 29-f (10.25 g, 80.0 mmol) in DMF (50 ml), cooled to 0°C, was added imidazole (5.99 g, 88.0 mmol) and tert- butylchlorodimethylsilane (13.27 g, 88.0 mmol). The reaction was then stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed 3 times with a saturated aqueous solution of ammonium chloride and brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 29-g as a yellow oil.
Step 7: Intermediate 29-h
To a solution of intermediate 29-g (8.0 g, 33.1 mmol) and intermediate 29-d (4.70 g, 36.4 mmol) in THF (20 ml) were sequentially added
triphenylphosphine (12.15 g, 46.3 mmol) and DIAD (9.0 ml, 46.3 mmol) at room temperature and the reaction was then stirred at room temperature overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 29-h as a yellow oil.
Step 8: Intermediate 29-i
To a solution of intermediate 29-h (6.0 g, 16.97 mmol) in THF (85 ml) was added a 1.0 M solution of TBAF in THF (16.97 ml, 16.97 mmol) and the reaction was stirred at room temperature for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Diethyl ether was added to the residue; a precipitate formed and was collected by filtration to provide intermediate 29-i as white solid. Synthesis of intermediate 30-b
Figure imgf000059_0001
29-g 30-a 30-b
Scheme 30
Step 1: Intermediate 30-a
To a solution of intermediate 29-g (9.0 g, 37.1 mmol) and 2- (methylpyrimidin-5-yl)methanol (4.61 g, 37.1 mmol) in THF (37 ml) were sequentially added triphenylphosphine (11.69 g, 44.6 mmol) and DIAD (9.39 ml, 48.3 mmol) at room temperature and the reaction was then stirred at room temperature for 4 days. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 30- a as a yellow solid.
Step 2: Intermediate 30-b
To a solution of intermediate 30-a (12.5 g, 35.9 mmol) in THF (72 ml) was added a 1.0 M solution of TBAF in THF (35.9 ml, 35.9 mmol) and the reaction was stirred at room temperature for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 30-b as a white solid.
Synthesis of intermediate 31-d
Figure imgf000060_0001
Figure imgf000060_0002
29-g 31 -c 31-d
Scheme 31
Step 1: Intermediate 31-b
To a solution of methyl 6-methylnicotinate 31-a (20.10 g, 133 mmol) in THF (90 ml) cooled to 0°C was added drop wise a 1.0 M solution of LiAIH4 in THF ( 100 ml, 100 mmol) and the reaction was then stirred at 0°C for 1 hour. Water (3.8 ml) was slowly added, followed by 15% NaOH (3.5 ml) and water (11.4 ml) and the mixture was stirred at room temperature for 1 hour. The reaction was filtered over celite and volatiles were removed in vacuo to provide intermediate 31-b as a yellow oil.
Step 2: Intermediate 31-c
To a solution of intermediate 29-g (13.2 g, 54.5 mmol) and intermediate 31- b (7.38 g, 59.9 mmol) in THF (50 ml) were sequentially added
triphenylphosphine (21.43 g, 82.0 mmol) and DIAD (17.10 ml, 87.0 mmol) at room temperature and the reaction was then stirred at room temperature for 1 hour. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 31-c as a colorless oil.
Step 3: Intermediate 31-d
To a solution of intermediate 31-c (7.6 g, 21.87 mmol) in THF (44 ml) was added tetrabutylammonium fluoride trihydrate (5.72 g, 21.87 mmol) and the reaction was stirred at room temperature for 1 hour. Saturated aqueous ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over gS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 31-d as white solid.
Synthesis of intermediate 32-f
Figure imgf000061_0001
Scheme 32
Step 1: Intermediate 32-b
To a solution of dimethyl pyridine-2,5-dicarboxylate (13.0 g, 66.6 mmol) in a mixture of THF (110 mL) and ethanol (110 mL) was added calcium chloride (29.6 g, 266 mmol). After stirring at room temperature for 30 minutes the reaction was cooled to 0°C and sodium borohydride (3.78 g, 100 mmol) was added portion wise. After the addition was completed the reaction was stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and dichloromethane were added, the organic layer was separated and the aqueous phase was extracted twice with dichloromethane. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 32- b as a yellow solid.
Step 2: Intermediate 32-c
To a solution of intermediate 32-b (1.70 g, 10.17 mmol) in dichloromethane (203 ml_) was added 3,4-dihydro-2H-pyran (4.28 g, 50.8 mmol) and PPTS (2.56 g, 10.17 mmol) and the reaction was stirred at room temperature overnight. Water was added and the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 32-c as a white solid.
Step 3: Intermediate 32-d
To a solution of intermediate 32-c (2.56 g, 10.17 mmol) in THF (51 ml) cooled to 0°C was added drop wise a 1.0 M solution of DIBALH in hexane (23.39 ml, 23.39 mmol) and the reaction was then stirred at 0°C for 1.5 hour and room temperature overnight. Water (1.0 ml) was slowly added, followed 15% NaOH (3.5 ml) and water (2.3 ml) and the mixture was stirred at room temperature for 30 minutes. The reaction was filtered over celite and volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 32-d as a yellow oil.
Step 4: Intermediate 32-e
To a solution of intermediate 29-g (1.57 g, 6.51 mmol) and intermediate 32- d (2.56 g, 7.17 mmol) in THF (7 ml) were sequentially added
triphenylphosphine (2.56 g, 9.77 mmol) and DIAD (2.04 ml, 10.42 mmol) at room temperature and the reaction was then stirred at room temperature overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 32-e as a yellow solid. Step 5: Intermediate 32-f
To a solution of intermediate 32-e (2.2 g, 4.91 mmol) in THF (9.8 ml) was added a 1.0 M solution of TBAF in THF (4.91 ml, 4.91 mmol) and the reaction was stirred at room temperature for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 32-f as a white solid.
Synthesis of intermediate 33-a
Figure imgf000063_0001
31 -C 33-a
Scheme 33
To a solution of intermediate 31-c (424 mg, 1.82 mmol) in dichloromethane (9.0 ml) was added m-CPBA (538 mg, 2.18 mmol) and the reaction was stirred at room temperature for 4 hours. A saturated aqueous solution of NaHC03 and dichloromethane were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 33-a as a white solid.
Synthesis of intermediate 34-d
Figure imgf000063_0002
Scheme 34
Step 1: Intermediate 34-b
To a solution of 3,5-difluorophenol (15.0 g, 115 mmol) in acetone (200 ml) was added K2C03 (23.90 g, 173 mmol) and bromomethyl methyl ether (15.85 g, 127 mmol). The reaction was then stirred at room temperature overnight and filtered. The filtrate was concentrated under reduced pressure to provide intermediate 34-b as a colorless oil.
Step 2: Intermediate 34-c
To a solution of (l-methyl-lH-imidazol-5-yl) methanol (3.1 g, 27.6 mmol) and intermediate 34-b (4.01 g, 23.04 mmol) in toluene (25.0 ml) and DMPU (25.0 ml) was added sodium 2-methylpropan-2-olate (4.43 g, 46.1 mmol). The reaction was stirred overnight at 80 °C and then cooled to room temperature. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed twice with a saturated aqueous solution of ammonium chloride and brine, dried over MgS04, filtered and concentrated in vacuo. Purification by silica gel chromatography provided intermediate 34-c as beige oil.
Step 3: Intermediate 34-d
To a solution of intermediate 34-c (3.2 g, 12.02 mmol) in MeOH (25.0 ml) was added 4N HCI in dioxane (10.95 ml, 361.0 mmol) and the reaction was stirred overnight at room temperature. Volatiles were removed in vacuo. Diethyl ether was added to the residue; a precipitate formed and was collected by filtration to provide intermediate 34-d- HCI as a white solid.
Synthesis of intermediate 35-d
Figure imgf000065_0001
Scheme 35
Step 1: Intermediate 35-b
To a solution of l,2-dimethyl-lH-imidazole-5-carbaldehyde (1.0 g, 8.06 mmol) in THF (40.3 mL) cooled to 0°C was added drop wise a 1.0 M solution of L1AI H4 in THF (6.04 ml, 6.04 mmol) and the reaction was then stirred at room temperature for 1 hour. Water (250 uL) was slowly added, followed by 15% NaOH (250 uL) and water (750 uL) and the mixture was stirred at room temperature for 1 hour. The reaction was filtered over celite and volatiles were removed in vacuo to provide intermediate 35-b as a white solid.
Step 2: Intermediate 35-c
To a solution of intermediate 35-b (1.50 g, 11.89 mmol) and intermediate 34-b (2.07 g, 11.89 mmol) in DMPU (11.89 mL) and toluene (11.89 mL) was added sodium 2-methylpropan-2-olate (3.43 g, 35.7 mmol) at room temperature. The reaction was stirred overnight at 80 °C and then cooled to room temperature. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 35-c as a yellow oil.
Step 3: Intermediate 35-d To a solution of intermediate 35-c (3.30 g, 11.77 mmol) in MeOH (36.2 mL) was added 4N HCI in dioxane (10.7 mL, 353 mmol) and the reaction was stirred at room temperature overnight. Volatiles were removed under reduced pressure. Diethyl ether was added to the residue; a precipitate formed and was collected by filtration to provide intermediate 35-d-HCI as a white solid.
Synthesis of intermediate 36-f
Figure imgf000066_0001
Scheme 36
Step 1: Intermediate 36-c
To a suspension of 2-(benzyloxy)ethanamine HCI, 36-b (2.08 g, 11.10 mmol), and 2,5-bis(hydroxymethyl)-l,4-dioxane-2,5-diol 36-a (2.00 g, 11.10 mmol) in iPrOH (8 mL), were sequentially added potassium
thiocyanate (1.62 g, 16.7 mmol) and acetic acid (2.03 mL, 35.5 mmol) drop wise. The mixture was stirred at room temperature overnight. Water was added; a precipitate formed and was collected by filtration to provide intermediate 36-c as a white solid.
Step 2: Intermediate 36-d
To a solution of intermediate 36-c (1.5 g, 5.67 mmol) and H2W04 (14 mg, 0.057 mmol) in MeOH (22.7 mL) at 40°C was added H202 (1.85 mL, 18.16 mmol) drop wise. The mixture was stirred at reflux overnight and then cooled to room temperature. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 36- d as a colorless oil.
Step 3: Intermediate 36-e
To a solution of intermediate 36-d (1.46 g, 6.32 mmol) and intermediate 34- b ( 1.0 g, 5.74 mmol) in DMPU (11.48 ml) and toluene (11.48 ml) was added sodium 2-methylpropan-2-olate (1.10 g, 11.48 mmol) at room temperature. The reaction was stirred overnight at 80 °C and then cooled to room temperature. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure.
Purification by silica gel chromatography provided intermediate 36-e as a colorless oil.
Step 4: Intermediate 36-f
To a solution of intermediate 36-e (400 mg, 1.03 mmol) in MeOH (10.4 mL) was added 4N HCI in dioxane (2.50 mL, 10.0 mmol) the reaction was stirred at room temperature overnight. Volatiles were removed under reduced pressure to provide intermediate 36-f-HCI as a white solid.
Synthesis of intermediates 37-f and 37-f
Figure imgf000068_0001
Step 1: Intermediates 37-b and 37-b'
To a solution of lH-imidazole-5-carbaldehyde, 37-a (3.0 g, 31.2 mmol), in DMF (20 ml) was added a 60% dispersion of NaH in mineral oil (1.25 g, 31.2 mmol) portion wise. After stirring for 30 minutes at room temperature, (2- (chloromethoxy)ethyl)trimethylsilane (5.73 g, 34.3 mmol) was added and the reaction was then was stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediates 37-b and 37-b' as an inseparable mixture.
Step 2: Intermediates 37-c and 37-c' 3 000513
To a solution of intermediates 37-b and 37-b' (3.2 g, 14.14 mmol) in THF (56.6 ml) was added NaBH4 (535 mg, 14.14 mmol) at room temperature. The reaction was stirred overnight at room temperature and then cooled to 0°C. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS0 , filtered and concentrated under reduced pressure to provide intermediate 37-c and 37-c' as an inseparable mixture.
Step 3: Intermediate 37-d
To a solution of 3,5-difluorophenol (8.0 g, 61.4 mmol) in acetone (100 ml) was added potassium carbonate (17.0 g, 123.0 mmol) and potassium iodide (1.021 g, 6.15 mmol). The reaction was heated to 65 °C and benzyl bromide (8.03 g, 67.6 mmol) was added. The reaction was then stirred overnight at 65°C, cooled to room temperature and filtered. Volatiles were removed in vacuo. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated in vacuo. Purification by silica gel chromatography provided intermediate 37-d as a colorless oil.
Step 4: Intermediate 37-e and 37-e'
To a solution of intermediates 37-c and 37-c' (1.0 g, 4.38 mmol) and intermediate 37-d (877 mg, 3.98 mmol) in DMPU (7.96 ml) and toluene (7.96 ml) was added sodium 2-methylpropan-2-olate (765 mg, 3.98 mmol) at room temperature. The reaction was stirred overnight at 80 °C and then cooled to room temperature. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediates 37-e and 37-e' as an inseparable mixture. Step 5: Intermediates 37-f and 37-f
A methanol solution of intermediate 37-e and 37-e' (140 mg, 0.32 mmol) was treated with 10% palladium on carbon (70 mg, 0.045 mmol) and purged with H2. The solution was stirred under H2 (1 atm) for 2 hours before being filtered through celite. The filtrate was concentrated in vacuo to provide intermediate 37-f and 37-f as an inseparable mixture.
Synthesis of intermediates 38-e and 38-e'
Figure imgf000070_0001
Step 1: Intermediates 38-b and 38-b' To a solution of lH-imidazole-5-carbaldehyde (5.0 g, 45.4 mmol) in DMF (20 mL) was added a 60% dispersion of NaH mineral oil (1.81 g, 45.4 mmol) portion wise. After stirring for 30 minutes at room temperature, (2- (chloromethoxy)ethyl)trimethylsilane (9.08 g, 54.5 mmol) was added and the reaction was then stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediates 38-b and 38-b' as an inseparable mixture.
Step 2: Intermediates 38-c and 38-c'
To a solution of intermediate 38-b and 38-b' (7.0 g, 29.1 mmol) in THF (116.0 ml) was added IMaBH4 (1.10 g, 29.1 mmol) at room temperature. The reaction was stirred overnight at room temperature and then cooled to 0°C. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediates 38-c and 38-c' as an inseparable mixture.
Step 3: Intermediate 38-d and 38-d'
To a solution of intermediate 38-c and 38-c' (1.0 g, 4.13 mmol) and intermediate 37-d (826 mg, 3.75 mmol) in DMPU (7.50 ml) and toluene (7.50 ml) was added sodium 2-methylpropan-2-olate (721 mg, 7.50 mmol) at room temperature. The reaction was stirred overnight at 80 °C and then cooled to room temperature. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediates 38-d and 38-d' as an inseparable mixture. Step 4: Intermediates 38-e and 38-e'
A methanol solution of intermediates 38-d and 38-d' (200 mg, 0.45 mmol) was treated with 10% palladium on carbon (96 mg, 0.045 mmol) and purged with H2. The solution was stirred under H2 (1 atm) for 2 hours before being filtered through celite. The filtrate was concentrated in vacuo to provide intermediate 38-e and 38-e' as an inseparable mixture.
Synthesis of intermediate 39-b
Figure imgf000072_0001
29-g 39-a 39-b
Scheme 39 Step 1: Intermediate 39-a
To a solution of intermediate 29-g (4.20 g, 17.3 mmol) and pyrimidin-5- ylmethanol (1.90 g, 17.3 mmol) in THF (35 ml_) were sequentially added triphenylphosphine (5.91 g, 22.5 mmol) and DIAD (4.38 ml_, 22.5 mmol) at room temperature and the reaction was then stirred at room temperature for 3 hours. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 39-a as a white solid.
Step 2: Intermediate 39-b
To a solution of intermediate 39-a (5.80 g, 17.3 mmol) in THF (35 mL) was added a 1.0 M solution of TBAF in THF (17.3 ml, 17.3 mmol) and the reaction was stirred at room temperature for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 39-b as a white solid. Synthesis of intermediate 40-b
Figure imgf000073_0001
29-g 40-a 40-b
Scheme 40
Step 1: Intermediate 40-a
To a solution of intermediate 29-g (4.62 g, 19.1 mmol) and pyrazin-2- ylmethanol (2.10 g, 19.1 mmol) in THF (38 mL) were sequentially added triphenylphosphine (7.50 g, 28.6 mmol) and DIAD (5.19 ml, 26.7 mmol) at room temperature and the reaction was then stirred at room temperature overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 40-a as a colorless oil.
Step 2: Intermediate 40-b
To a solution of intermediate 40-a (3.40 g, 10.2 mmol) in THF (20 mL) was added a 1.0 M solution of TBAF in THF (10.2 ml, 10.2 mmol) and the reaction was stirred at room temperature for 1 hour. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 40-b as a white solid.
Synthesis of intermediate 41-b
Figure imgf000073_0002
29-g 41-a 41-b Scheme 41
Step 1: Intermediate 41-a
To a solution of intermediate 29-g (2.60 g, 10.7 mmol) and benzyl alcohol (1.39 g, 12.9 mmol) in THF (20 mL) were sequentially added
triphenylphosphine (3.94 g, 15.02 mmol) and DIAD (2.92 mL, 15.0 mmol) at room temperature and the reaction was then stirred at room temperature overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 41-a as colorless oil.
Step 2: Intermediate 41-b
To a solution of intermediate 41-a (1.40 g, 4.21 mmol) in THF (10 ml) was added a 1.0 M solution of TBAF in THF (4.63 ml, 4.63 mmol) and the reaction was stirred at room temperature overnight. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 41-b as a colorless oil.
Synthesis of intermediate 42-d
Figure imgf000075_0001
Scheme 42
Step 1: Intermediate 42-b
To a solution of cyclopentanone (10.00 g, 119.0 mmol) in MeOH (594 ml_), were added tert-butyl hydrazinecarboxylate (16.50 g, 125.0 mmol) and the reaction was stirred overnight at room temperature. Volatiles were removed under reduced pressure to provide intermediate 42-b as a white solid.
Step 2: Intermediate 42-c
To a solution of intermediate 42-b (10.00 g, 50.40 mmol) in THF (50.4 mL) and MeOH (50.4 mL) was added sodium cyanoborohydride (3.80 g, 60.5 mmol) portion wise. The reaction was refluxed under argon for 10 minutes, and then cooled to room temperature. 6N HCI (25 mL) was added, the mixture was refluxed for 3 hours, cooled to room temperature and stirred overnight. The reaction was filtered to remove inorganic insoluble material and the filtrate was concentrated under reduced pressure and azeotroped three times with toluene. The residue was dissolved in hot isopropanol, cooled to room temperature, diluted with ether and then cooled to 0°C. A precipitate formed and was collected by filtration to provide intermediate 42- HCI as a white solid. Step 3: Intermediate 42-d
To a solution of intermediate 9-b (3.00 g, 11.4 mmol) and TEA (3.50 mL, 25.1 mmol) in EtOH (11.4 mL) was added intermediate 42-c-HCI (1.86 g, 13.7 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel
chromatography provided intermediate 42-d as a white solid.
Synthesis of intermediate 43-e
Figure imgf000076_0001
Scheme 43 Step 1: Intermediate 43-b
To a solution of dihydro-2H-pyran-4-(3H)-one (15.0 g, 150.0 mmol) in MeOH (749 mL), were added tert-butyl hydrazinecarboxylate (20.79 g, 157.0 mmol) and the reaction was stirred overnight at room temperature. Volatiles were removed under reduced pressure to provide intermediate 43-b as a white solid.
Step 2: Intermediate 43-c
A methanol solution of intermediate 43-b (32.1 g, 150.0 mmol) was treated with 10% palladium on carbon (6.39 g, 3.00 mmol), acetic acid (100 μΙ_) and purged with H2. The solution was stirred under H2 ( 1 atm) overnight before being filtered through celite. The filtrate was concentrated in vacuo to provide intermediate 43-c as a white solid.
Step 3: Intermediate 43-d
To a solution of intermediate 43-c (32.4 g, 150 mmol) in MeOH (300 mL) was added 4N HCI in 1,4-dioxane (300 ml, 1200 mmol) and the reaction was stirred at room temperature for 5 hours. Diethyl ether was added and a precipitate formed which was collected by filtration to provide intermediate 43-d-HCI as a white solid.
Step 4: Intermediate 43-e
To a solution of intermediate 9-b (5.00 g, 19.0 mmol) and TEA (5.30 mL, 38.0 mmol) in EtOH (19.0 mL) was added intermediate 43-c-HCI (3.48 g, 22.81 mmol) and the reaction was then stirred for 2 hours at 100 °C.
Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 43- e as a yellow solid.
Synthesis of intermediate 44-d
Figure imgf000078_0001
Figure imgf000078_0002
Step 1: Intermediate 44-b tert-Butyl hydrazinecarboxylate (7.60 g, 57.5 mmol) was added to acetone (50 mL) and the reaction was stirred overnight at room temperature.
Volatiles were removed under reduced pressure to provide intermediate 44-b as a white solid.
Step 2: Intermediate 44-c
To a solution of intermediate 44-b (9.90 g, 57.5 mmol) in THF (57.5 mL) and MeOH (57.5 mL) was added sodium cyanoborohydride (4.34 g, 69.0 mmol) portion wise. The reaction was refluxed under nitrogen for 10 minutes, and then cooled to room temperature. 6N HCI (30 mL) was added, the mixture was refluxed for 3 hours, cooled to room temperature and stirred overnight. The reaction was filtered to remove inorganic insoluble material and the filtrate was concentrated under reduced pressure and azeotroped three times with toluene for complete water removal. The residue was dissolved in hot isopropanol, cooled to room temperature, diluted with ether and then cooled to 0°C. A precipitate formed and was collected by filtration to provide intermediate 44-c-HCI as a white solid.
Step 3: Intermediate 44-d
To a solution of intermediate 9-b (12.61 g, 47.9 mmol) and TEA (14.70 mL, 105.0 mmol) in EtOH (96.0 ml) was added intermediate 44-c.HCI (6.36 g, 57.5 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel
chromatography provided intermediate 44-d as a white solid.
Synthesis of intermediate 45-a
Figure imgf000079_0001
Scheme 45
To a solution of intermediate 9-b (2.0 g, 7.60 mmol) and TEA (2.12 ml, 15.2 mmol) in EtOH (7.60 mL) was added tert-butylhydrazine hydrochloride (1.13 g, 9.12 mmol) and the reaction was then stirred for 2 hours at 100 °C.
Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 45- a as a yellow solid. Synthesis of intermediate 46-a
Figure imgf000080_0001
Scheme 46
To a solution of intermediate 9-b (1.45 g, 5.53 mmol) and TEA (1.54 mL, 11.1 mmol) in EtOH (15.0 mL) was added cyclohexylhydrazine hydrochloride (1.00 g, 6.64 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 46- a as a yellow solid.
Synthesis of intermediate 47-a
Figure imgf000080_0002
Scheme 47
To a solution of intermediate 9-b (2.00 g, 7.60 mmol) and TEA (1.27 mL, 9.12 mmol) in EtOH (7.60 mL) was added 2-hydroxyethylhydrazine (618 pL, 9.12 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 47-a as a white solid.
Synthesis of intermediate 48-c
Figure imgf000081_0001
Scheme 48
Step 1: Intermediate 48-b
To a mixture of sodium hydroxide (7.37 g, 184.0 mmol) and hydrazine monohydrate (46.10 g, 921.0 mmol) heated to 95°C, was added l-chloro-2- methylpropan-2-ol (20.00 g, 184.0 mmol). The reaction was stirred overnight at 95 °C and then cooled to room temperature. Volatiles were removed under reduced pressure. THF (40 mL) and diethyl ether (40 mL) were added to the residue; a precipitate formed which was removed by filtration. The filtrate was concentrated under reduced pressure to provide intermediate 48-b as a colorless oil. Step 2: Intermediate 48-c
To a solution of intermediate 9-b (4.45 g, 16.9 mmol) and TEA (4.71 mL, 33.8 mmol) in EtOH (15.0 mL) was added intermediate 48-b (1.76 g, 16.9 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of
ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 48-c as a white solid.
Synthesis of intermediate 49-c
Figure imgf000082_0001
Scheme 49
Step 1: Intermediate 49-b
To a mixture of sodium hydroxide (1.15 g, 28.8 mmol) and hydrazine monohydrate (7.20 g, 144.0 mmol) heated to 95°C, was added 3-bromo-l propanol (4.00 g, 28.8 mmol) and the reaction was stirred overnight at 95 and then cooled to room temperature. Volatiles were removed under reduced pressure. Ethanol was added to the residue; a precipitate formed and was removed by filtration. The filtrate was concentrated under reduced pressure and 1M HCI in diethyl was added to the residue. After stirring for 15 minutes a precipitate formed and was collected by filtration to provide intermediate 49-b-HCI as a white solid.
Step 2: Intermediate 49-c
To a solution of intermediate 9-b (1.12 g, 4.28 mmol) and TEA (1.19 mL, 8.56 mmol) in EtOH (10.0 mL) was added intermediate 49-b-HCI (650 mg, 5.13 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 49-c as a white solid.
Synthesis of intermediate 50-c
Figure imgf000083_0001
36-a 50-a 50-b
Figure imgf000083_0002
Scheme 50
Step 1: Intermediate 50-a
Dihydroxyacetone dimer (15.0 g) and tert-butyl hydrazinecarboxylate (22.01 g) were dissolved in ethanol (500 mL) and this solution was stirred at room temperature for 2 days. After the reaction mixture was concentrated under reduced pressure, the resulting residue was recrystallized from ethyl acetate to provide intermediate 50-a as a white solid.
Step 2: Intermediate 50-b
To a solution of intermediate 50-a (10.0 g, 49.0 mmol) in THF (49.0 mL) and MeOH (49.0 mL) was added sodium cyanoborohydride (3.69 g, 58.8 mmol) portion wise. The reaction was refluxed under nitrogen for 10 minutes, and then cooled to room temperature. 6I\I HCI (40 mL) was added, the mixture was refluxed for 3 hours, cooled to room temperature and stirred overnight. The reaction was filtered to remove inorganic insoluble material and the filtrate was concentrated under reduced pressure and azeotroped three times with toluene to provide intermediate 50-b-HCI as a white solid.
Step 3: Intermediate 50-c
To a solution of intermediate 9-b (10.70 g, 40.9 mmol) and TEA (12.5 mL,
90.0 mmol) in EtOH (40.9 mL) was added intermediate 50-b-HCI (7.00 g,
49.1 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel
chromatography provided intermediate 50-c as a beige solid. Synthesis of intermediate 51-a
Figure imgf000085_0001
Scheme 51
To a solution of intermediate 9-c (1.40 g, 5.32 mmol) and 3-methyloxetan-3- yl)methanol (1.08 g, 10.64 mmol) in THF (5.3 ml_) were sequentially added triphenylphosphine (1.67 g, 6.39 mmol) and DIAD (1.13 mL, 5.85 mmol) at room temperature and the reaction was then stirred at room temperature for 4 days. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 51-a as a white solid.
Synthesis of intermediate 52-a
Figure imgf000085_0002
Scheme 52
To a solution of intermediate 9-c (500 mg, 1.90 mmol) and 3- morpholinopropan-l-ol (263 μΙ, 1.90 mmol) in THF (19.0 ml) cooled to 0°C were sequentially added triphenylphosphine (498 mg, 1.90 mmol) and DIAD (370 μΙ, 1.90 mmol). The reaction was stirred at 0°C for 1 hour and room temperature overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 52-a as a white foam.
Synthesis of intermediate 53-a
Figure imgf000086_0001
Scheme 53
To a solution of intermediate 9-d (3.00 g, 10.3 mmol) and N-(2- hydroxyethyl)morpholine (1.88 ml, 15.1 mmol) in THF ( 103 ml) cooled to 0°C were sequentially added triphenylphosphine (4.07 mg, 15.1 mmol) and DIAD (3.02 ml, 15.5 mmol). The reaction was stirred at 50°C overnight. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 53-a as a white solid.
Synthesis of intermediate 54-a
Figure imgf000086_0002
Scheme 54
To a solution of intermediate 9-c (500 mg, 1.90 mmol) and 2-(pyrrolidin-l- yl)ethanol (219 mg, 1.90 mmol) in THF (9.5 ml) cooled to 0°C were sequentially added triphenylphosphine (498 mg, 1.90 mmol) and DIAD (370 pi, 1.90 mmol). The reaction was stirred at 0°C for 1 hour and room
temperature for 30 minutes. Volatiles were removed under reduced pressure. Purification by silica gel chromatography provided intermediate 54-a as yellow solid.
Synthesis of Compound 65:
Figure imgf000087_0001
17-a Compound 65
Scheme 55
To a solution of intermediate 17-a (321 mg, 0.85 mmol) and intermediate 31-d (200 mg, 0.85 mmol) in 1,4-dioxane (4.30 ml) were sequentially added N,N-dimethylglycine (265 mg, 2.57 mmol), copper(I) iodide (163 mg, 0.85 mmol) and cesium carbonate (1.12 g, 3.43 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 65-2HCI as a white solid.
Synthesis of Compound 85
Figure imgf000088_0001
Scheme 56
Step 1: Intermediate 56-a
To a solution of intermediate 44-d (5.00 g, 16.4 mmol) and intermediate 31- d (4.20 g, 18.0 mmol) in 1,4-dioxane (54.6 ml) were sequentially added N,N-dimethylglycine (3.80 g, 36.9 mmol), copper(I) iodide (2.34 g, 12.29 mmol) and cesium carbonate (21.35 g, 65.5 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 56-a-HCI as a white solid.
Step 2: Compound 85
Intermediate 56-a-HCI (3.13 g, 6.84 mmol) and trimethyl orthoformate (48.7 ml, 445.0 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with ammonia (7.0 N in MeOH) (48.9 ml, 342.0 mmol). The mixture was stirred at room temperature for 3 days and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 85-2HCI as a white solid. MS (m/z) M + H= 485.2
Synthesis of Compound 91:
Figure imgf000089_0001
Scheme 57
Step 1: Intermediate 57-a
To a solution of intermediate 43-e (192 mg, 0.55 mmol) and intermediate 33-a (190 mg, 0.66 mmol) in 1,4-dioxane (2.8 ml) were sequentially added N,N-dimethylglycine (129 mg, 1.24 mmol), copper(I) iodide (79 mg, 0.42 mmol) and cesium carbonate (722 mg, 2.21 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was
separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 57-a as a yellow foam.
Step 2: Compound 91
Intermediate 57-a (286 mg, 0.55 mmol) and trimethyl orthoformate (3.94 ml, 36.0 mmol) were heated at 110 °C for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 IN ammonia in MeOH (3.96 ml, 27.7 mmol). The mixture was stirred at room temperature for 3 days and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 91-HCI as white solid. MS (m/z) M + H= 543.1
Synthesis of Compound 101:
Figure imgf000090_0001
Scheme 58
Step 1: Intermediate 58-a
To a solution of intermediate 43-e (400 mg, 1.15 mmol) and intermediate 32-f (384 mg, 1.15 mmol) in 1,4-dioxane (2.8 ml) were sequentially added N,N-dimethylglycine (267 mg, 2.59 mmol), copper(I) iodide (165 mg, 0.86 mmol) and cesium carbonate (1.50 g, 4.61 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 58-a as a beige foam. Step 2: Compound 101
Intermediate 58-a (690 mg, 1.15 mmol) and trimethyl orthoformate (8.18 ml, 74.8 mmol) were heated at 110 °C for 4 days. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (8.21 ml, 57.5 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 101-2HCI as a white solid. MS (m/z) M + H= 543.1
Synthesis of Compound 128:
Figure imgf000091_0001
Scheme 59
Step 1: Intermediate 59-a
To a solution of intermediate 44-d (261 mg, 0.85 mmol) and intermediate 32-f (285 mg, 0.85 mmol) in 1,4-dioxane (2.8 ml) were sequentially added N,N-dimethylglycine (198 mg, 1.92 mmol), copper(I) iodide (122 mg, 0.64 mmol) and cesium carbonate (1.11 g, 3.42 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide Purification by reverse phase
chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 59-a-HCI as a beige foam.
Step 2: Compound 128
Intermediate 59-a (405 mg, 0.85 mmol) and trimethyl orthoformate (6.08 ml, 55.6 mmol) were heated at 110 °C for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (6.11 ml, 42.8 mmol). The mixture was stirred at room temperature for 3 days and volatiles were removed under reduced pressure Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 128-2HCI as a white solid. MS (m/z) M + H= 501.1
Synthesis of Compound 78:
Figure imgf000092_0001
Scheme 60
Step 1: Intermediate 60-a
To a solution of intermediate 44-d (300 mg, 0.98 mmol) and intermediate 29-i (259 mg, 1.08 mmol) in 1,4-dioxane (3.9 ml) were sequentially added N,N-dimethylglycine (304 mg, 2.95 mmol), copper(I) iodide ( 187 mg, 0.98 mmol) and cesium carbonate (961 mg, 2.95 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of - ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 60-a-HCI as a beige foam.
Step 2: Compound 78
Intermediate 60-a-HCI (265 mg, 0.57 mmol) and trimethyl orthoformate (1.87 ml, 17.16 mmol) were heated at 110 °C for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (5.7 ml, 11.44 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 78-2HCI as beige solid. MS (m/z) M + H= 491.2
Synthesis of Compound 58:
Figure imgf000093_0001
10-a Compound 58
Scheme 61
To a solution of intermediate 10-a (3.96 g, 11.1 mmol) and intermediate 29-i (2.91 g, 12.2 mmol) in 1,4-dioxane (55.3 ml) were sequentially added N,N- dimethylglycine (3.42 g, 33.2 mmol), copper(I) iodide (2.10 g, 11.07 mmol) and cesium carbonate ( 10.82 g, 33.2 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 58-2HCI as a beige solid. MS (m/z) M + H= 517.2
Synthesis of Compound 117:
Figure imgf000094_0001
Scheme 62
Step 1: Intermediate 62-a
To a solution of intermediate 48-c (200 mg, 0.59 mmol) and intermediate 29-i ( 157 mg, 0.65 mmol) in 1,4-dioxane (1.50 ml) were sequentially added N,N-dimethylglycine (92 mg, 0.89 mmol), copper(I) iodide (57 mg, 0.29 mmol) and cesium carbonate (583 mg, 1.79 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 62-a-HCI as a beige solid.
Step 2: Compound 117
Intermediate 62-a-HCI (322 mg, 0.65 mmol) and trimethyl orthoformate (3.0 ml, 19.57 mmol) were heated at 110 °C for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with ammonia (7.0 N in MeOH) (1.85 ml, 13.0 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 117-2HCI as a white solid. MS (m/z) M + H= 521.1
Synthesis of Compound 100:
Figure imgf000095_0001
50-c 63-a Compound 100
Scheme 63
Step 1: Intermediate 63-a
To a solution of intermediate 50-c (1.42 g, 4.21 mmol) and intermediate 29-i (1.10 g, 4.63 mmol) in 1,4-dioxane (16.8 ml) were sequentially added N,N- dimethylglycine (1.30 g, 12.6 mmol), copper(I) iodide (802 mg, 4.21 mmol) and cesium carbonate (4.12 g, 12.63 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 63- a-HCI as a white solid.
Step 2: Compound 100
Intermediate 63-a-HCI (577 mg, 1.16 mmol) and trimethyl orthoformate (3.82 ml, 35.0 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (3.30 ml, 23.32 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 100-2HCI as white solid. MS (m/z) M + H= 523.2
Synthesis of Compound 113:
Figure imgf000096_0001
Scheme 64
Step 1: Intermediate 64-a
To a solution of compound 100 (198.0 mg, 0.38 mmol) in THF cooled to - 10°C was slowly added a 2.5 M solution of n-butyllithium in THF (304 μΙ, 0.76 mmol). After stirring for 30 minutes, para-toluenesulfonyl chloride (72.0 mg, 0.38 mmol) in THF (2 ml) was added. The reaction was stirred at 60°C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 64-a as beige foam.
Step 2: Compound 113
To a solution of intermediate 64-a (85.0 mg, 0.12 mmol) in THF cooled to - 10°C was slowly added a 2.5 M solution of n-butyllithium in THF (110.0 μΙ, 0.27 mmol). The reaction was stirred at 60°C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and
concentrated under reduced pressure. Purification by silica gel
chromatography provided compound 113 as pale yellow foam. MS (m/z) M + H= 505.2
Synthesis of Compound 125:
Figure imgf000097_0001
51 -a 65-a Compound 125
Scheme 65
Step 1: Intermediate 65-a
To a solution of intermediate 51-a (300 mg, 0.86 mmol) and intermediate 29-i (262 mg, 0.95 mmol) in 1,4-dioxane (1.50 ml) were sequentially added N,N-dimethylglycine (267 mg, 2.59 mmol), copper(I) iodide (165 mg, 0.86 mmol) and cesium carbonate (845 mg, 2.59 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 65-a as a beige foam.
Step 2: Compound 125
Intermediate 65-a (120 mg, 0.23 mmol), trimethyl orthoformate (260 μΙ, 2.3 mmol) and PTSA (catalytic) were stirred at room temperature for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (652 μΙ, 4.6 mmol). The mixture was stirred at room temperature for 3 days and volatiles were removed under reduced pressure. Purification by silica gel chromatography provided compound 125 as a white solid. MS (m/z) M + H= 533.2
Synthesis of Compound 54:
Figure imgf000098_0001
Compound 54
Scheme 66
To a solution of intermediate 17-a ( 1.20 g, 3.21 mmol) and intermediate 29-i (844 mg, 3.53 mmol) in DMF (16.0 ml) were sequentially added N,N- dimethylglycine (992 mg, 9.62 mmol), copper(I) iodide (611 mg, 3.21 mmol) and cesium carbonate (4.18 g, 12.83 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 54-2HCI as a yellow solid. MS (m/z) M+H= 533.1
Synthesis of Compound 55:
Figure imgf000099_0001
Scheme 67
Step 1: Intermediate 67-a
To a solution of intermediate 52-a (397.0 mg, 1.00 mmol) and intermediate 29-i (268 mg, 1.12 mmol) in 1,4-dioxane (5.0 ml) were sequentially added N,N-dimethylglycine (157 mg, 1.53 mmol), copper(I) iodide (97.0 mg, 050 mmol) and cesium carbonate (995 mg, 3.05 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 67- a as beige foam. Step 2: Compound 55
Formamide (2.84 ml, 71.3 mmol) was added to intermediate 67-a (559 mg, 1.0 mmol) and the reaction was stirred at 180 °C for 2 hours. Water and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 55-3HCI as beige solid. MS (m/z) M + H= 576.2
Synthesis of Compound 52:
Figure imgf000100_0001
Scheme 68
To a solution of intermediate 53-a (1.0 g, 2.48 mmol) and intermediate 29-i (771 mg, 3.22 mmol) in DMAC (12.4 ml) were sequentially added quinolin-8- ol (36.0 mg, 0.24 mmol), copper(I) iodide (47.0 mg, 0.24 mmol) and cesium carbonate (808 mg, 2.48 mmol) and the reaction was heated at 140°C for 2 hours. A saturated aqueous solution of ammonium chloride and ethyl acetate were added, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 52-3HCI as white solid. MS (m/z) M + H= 562.1
Synthesis of Compound 57:
Figure imgf000101_0001
Scheme 69
Step 1: Intermediate 69-a
To a solution of intermediate 54-a (425.0 mg, 1.18 mmol) and intermediate 29-i (282 mg, 1.18 mmol) in 1,4-dioxane (5.9 ml) were sequentially added N,N-dimethylglycine ( 182 mg, 1.77 mmol), copper(I) iodide ( 112 mg, 0.59 mmol) and cesium carbonate (1.15 g, 3.54 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 69-a as a beige foam.
Step 2: Compound 57
Formamide (5.53 ml, 139.0 mmol) was added to intermediate 69-a (600 mg, 1.15 mmol) and the reaction was stirred at 180 °C for 4 hours. Water and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 57-3HCI as beige solid. MS (m/z) M + H= 546.2
Synthesis of Compound 102:
Figure imgf000102_0001
70-a Compound 102
Scheme 70
Step 1: Intermediate 70-a
To a solution of intermediate 44-d (4.00 g, 13.1 mmol) and intermediate 30- b (3.38 g, 14.4 mmol) in 1,4-dioxane (43.7 ml) were sequentially added N,N-dimethylglycine (3.04 g, 29.5 mmol), copper(I) iodide (1.87 g, 9.83 mmol) and cesium carbonate (17.08 g, 52.4 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 70-a-HCI as beige foam.
Step 2: Compound 102
Intermediate 70-a-HCI (3.80 g, 7.68 mmol) and trimethyl orthoformate (54.6 ml, 499.0 mmol) were heated at 110 °C for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 l\l ammonia in MeOH (54.8 ml, 384.0 mmol). The mixture was stirred at room temperature for 2 days and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 102-2HCI as white solid. MS (m/z) M+H= 486.2
Synthesis of Compound 129:
Figure imgf000103_0001
45-a 71-a Compound 129
Scheme 71
Step 1: Intermediate 71-a
To a solution of intermediate 45-a (350 mg, 1.10 mmol) and intermediate 30-b (283 mg, 1.20 mmol) in 1,4-dioxane (4.3 ml) were sequentially added N,N-dimethylglycine (1.07 g, 3.29 mmol), copper(I) iodide (209 mg, 1.10 mmol) and cesium carbonate (985 mg, 3.02 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 71-a-HCI as a beige solid. Step 2: Compound 129
Intermediate 71-a-HCI (403.0 mg, 0.85 mmol) and trimethyl orthoformate (6.07 ml, 55.5 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (6.07 ml, 42.7 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 129-2HCI as beige solid. MS (m/z) M + H= 500.1
Synthesis of Compound 106:
Figure imgf000104_0001
43-e 72-a Compound 106
Scheme 72
Step 1: Intermediate 72-a
To a solution of intermediate 43-e (350 mg, 1.00 mmol) and intermediate 30-b (260 mg, 1.10 mmol) in 1,4-dioxane (4.0 ml) were sequentially added N,N-dimethylglycine (312 mg, 3.02 mmol), copper(I) iodide (192 mg, 1.00 mmol) and cesium carbonate (985 mg, 3.02 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 72-a-HCI as a beige solid.
Step 2: Compound 106
Intermediate 72-a-HCI (263 mg, 0.52 mmol) and trimethyl orthoformate ( 1.72 ml, 15.8 mmol) were heated at 110 °C for 2 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (46.2 ml, 324 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 106-2HCI as a beige solid. MS (m/z) M + H= 528.1
Synthesis of Compound 114:
Figure imgf000105_0001
49-c 73-a Compound 114
Scheme 73
Step 1: Intermediate 73-a
To a solution of intermediate 49-c (250 mg, 0.78 mmol) and intermediate 30-b (201 mg, 0.85 mmol) in 1,4-dioxane (3.1 ml) were sequentially added N,N-dimethylglycine (241 mg, 2.33 mmol), copper(I) iodide (148 mg, 0.78 mmol) and cesium carbonate (761 mg, 2.33 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 73-a-HCI as a beige solid.
Step 2: Compound 114
Intermediate 73-a-HCI (106 mg, 0.22 mmol) and trimethyl orthoformate (737 μΙ, 6.74 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (2.24 ml, 4.49 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 114-2HCI as a beige solid. MS (m/z) M + H= 502.2
Synthesis of Compound 130:
Figure imgf000106_0001
50-c 74-a Compound 130
Scheme 74
Step 1: Intermediate 74-a
To a solution of intermediate 50-c (300 mg, 0.89 mmol) and intermediate 30-b (229 mg, 0.97 mmol) in 1,4-dioxane (890 μΙ) were sequentially added N,N-dimethylglycine (275 mg, 2.67 mmol), copper(I) iodide (169 mg, 0.89 mmol) and cesium carbonate (870 mg, 2.67 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 74-a-HCI as beige solid.
Step 2: Compound 130
A solution of intermediate 74-a (125 mg, 0.25 mmol), trimethyl orthoformate (836 μΙ, 7.65 mmol) and PTSA (catalytic) was stirred at room temperature for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with ammonia (7.0 N in MeOH) (2.55 ml, 5.10 mmol). The mixture was stirred at room temperature for 2 days and volatiies were removed under reduced pressure. Purification by reverse phase
chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 130-2HCI as a white solid. MS (m/z) M + H= 518.1
Synthesis of Compound 97:
Figure imgf000107_0001
Scheme 75 Step 1: Intermediate 75-a
To a solution of intermediate 43-e (101 mg, 0.29 mmol) and intermediate 36-f (110 mg, 0.32 mmol) in 1,4-dioxane (1.4 ml) were sequentially added N,N-dimethylglycine (90 mg, 0.87 mmol), copper(I) iodide (56 mg, 0.29 mmol) and cesium carbonate (381 mg, 1.17 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 75-a as a beige solid.
Step 2: Compound 97
Intermediate 75-a (160 mg, 0.26 mmol), trimethyl orthoformate ( 1.86 ml, 17.09 mmol) and TFA (catalytic) were heated at 110 °C for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (1.87 ml, 13.14 mmol). The mixture was stirred at 50°C overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 97-2HCI as a beige solid. MS (m/z) M + H= 636.1
Synthesis of Compound 99:
Figure imgf000109_0001
Compound 97 Compound 99
Scheme 76
A solution of compound 97-2HCI (100 mg, 0.15 mmol) in ethyl acetate was treated with 10% palladium on carbon (32 mg, 0.015 mmol) and purged with H2. The solution was stirred under H2 (1 atm) for 1 hour before being filtered through celite. The filtrate was concentrated in vacuo. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 99-2HCI as white solid. MS (m/z) M + H= 546.2
Synthesis of Compound 93:
Figure imgf000110_0001
Compound 93
Scheme 77
Step 1: Intermediates 77-a and 77-a'
To a solution of intermediate 43-e (115 mg, 0.33 mmol) and intermediates 37-f and 37-f (123 mg, 0.36 mmol) in 1,4-dioxane (1.6 ml) were
sequentially added N,N-dimethylglycine (102 mg, 0.99 mmol), copper(I) iodide (63 mg, 0.33 mmol) and cesium carbonate (431 mg, 1.32 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 77-a and 77-a' as an inseparable mixture.
Step 2: Intermediates 77-b and 77-b'
A solution of intermediates 77-a and 77-a' (100 mg, 0.16 mmol), trimethyl orthoformate (2.0 ml, 18.28 mmol) and PTSA (catalytic) was stirred at room temperature for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (2.0 ml, 14.1 mmol). The mixture was stirred at room temperature for 3 days and volatiles were removed under reduced pressure. Purification by silica gel
chromatography provided intermediates 77-b and 77-b' as an inseparable mixture.
Step 3: Compound 93
4N HCI in 1,4-dioxane (2 mL) was added to intermediates 77-b and 77-b' (60 mg, 0.09 mmol) band the mixture was stirred for 1 hour at room
temperature. Volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 93-2HCI as a white solid. MS (m/z) M + H= 502.2
Synthesis of Compound 118:
Figure imgf000112_0001
Scheme 78
Step 1: Intermediates 78-a and 78-a'
To a solution of intermediate 43-e (276.0 mg, 0.79 mmol) and intermediates 38-e and 38-e' (340 mg, 0.87 mmol) in 1,4-dioxane (795 μΙ) were
sequentially added ΙΜ,Ν-dimethylglycine (246 mg, 2.38 mmol), copper(I) iodide (151 mg, 0.79 mmol) and cesium carbonate (1.03 g, 3.18 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 78-a and 78-a' as an inseparable mixture.
Step 2: Intermediates 78-b and 78-b'
A solution of intermediates 78-a and 78-a' (90 mg, 0.14 mmol), trimethyl orthoformate (1.03 ml, 9.45 mmol) and TFA (catalytic) was stirred at room temperature for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (1.03 ml, 7.27 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure to provide intermediates 78-b and 78- b' as an inseparable mixture.
Step 3: Compound 118
4N HCI in 1,4-dioxane (2.82 mL) was added to intermediates 78-b and 78-b' (60 mg, 0.09 mmol) and the mixture was stirred overnight at room
temperature. Volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 118-2HCI as a white solid. MS (m/z) M + H = 516.2
Synthesis of Compound 90:
Figure imgf000113_0001
47-a 79-a Compound 90
Scheme 79 Step 1: Intermediate 79-a
To a solution of intermediate 79-a (310 mg, 1.0 mmol) and intermediate 34- d (313 mg, 1.21 mmol) in 1,4-dioxane (2.5 ml) were sequentially added N,N- dimethylglycine (156 mg, 1.51 mmol), copper(I) iodide (96.0 mg, 0.50 mmol) and cesium carbonate ( 1.31 g, 4.04 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 79- a as a beige solid.
Step 2: Compound 90
Intermediate 79-a (280 mg, 0.62 mmol) and trimethyl orthoformate (2.05 ml, 18.7 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (1.78 ml, 12.5 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 90-2HCI as a beige solid. MS (m/z) M + H= 476.2
Synthesis of Compound 103:
Figure imgf000115_0001
46-a 80-a Compound 103
Scheme 80
Step 1: Intermediate 80-a
To a solution of intermediate 46-a (300 mg, 0.87 mmol) and intermediate 34-d (247 mg, 0.95 mmol) in 1,4-dioxane (2.1 ml) were sequentially added N,N-dimethylglycine ( 134 mg, 1.30 mmol), copper(I) iodide (83 mg, 0.43 mmol) and cesium carbonate (1.13 g, 3.48 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 80- a-HCI as a beige solid.
Step 2: Compound 103
Intermediate 80-a-HCI (65.0 mg, 0.13 mmol) and trimethyl orthoformate(3.0 ml, 4.01 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (378 μΙ, 2.65 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 103-2HCI as a white solid. MS (m/z) M + H= 514.2
Synthesis of Compound 120:
Figure imgf000116_0001
42-d 81-a Compound 120
Scheme 81
Step 1: Intermediate 81-a
To a solution of intermediate 42-d (176 mg, 0.53 mmol) and intermediate 35-d (145 mg, 0.53 mmol) in 1,4-dioxane (3.5 ml) were sequentially added N,N-dimethylglycine ( 123 mg, 1.19 mmol), copper(I) iodide (76 mg, 0.40 mmol) and cesium carbonate (693 mg, 2.13 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 81-a-HCI as a yellow foam.
Step 2: Compound 120
Intermediate 81-a-HCI (259 mg, 0.53 mmol) and trimethyl orthoformate (3.78 ml, 34.6 mmol) were heated at 110 °C for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (3.80 ml, 26.6 mmol). The mixture was heated at 60°C for 5 hours and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 120-2HCI as a white solid. MS (m/z) M + H = 514.2
Synthesis of intermediate 82-c:
Figure imgf000117_0001
82-b
Scheme 82
Step 1: Intermediate 82-b
To a solution of 4-bromo-2-fluorobenzoyl chloride (16.27 g, 68.5 mmol) in toluene (85 ml) and THF (8.5 ml), cooled to -10 °C, were sequentially added malononitrile (4.75 g, 71.9 mmol) and DIPEA (23.93 ml, 137.0 mmol) in toluene (25 mL) drop wise over a period of 1 hour. After the addition was completed, the reaction was stirred for 2 hours at 0°C and room temperature for an additional 2 hours. Volatiles were removed under reduced pressure. IN HCI and ethyl acetate were added to the residue; the organic layer was separated, washed twice with IN HCI and brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 82-b as a yellow solid.
Step 2: Intermediate 82-c
To a solution of intermediate 82-b (18.29 g, 68.5 mmol) in acetonitrile (247 ml) and methanol (27.4 ml), cooled to 0°C, was added DIPEA (14.36 ml, 82.0 mmol) and a 2M solution of diazomethyl)trimethylsilane in hexanes (37.7 ml, 75.0 mmol). After the addition was completed, the reaction was stirred at room temperature overnight. Acetic acid ( 1.17 ml, 20.5 mmol) was added, the reaction was stirred for 30 minutes and volatiles were removed under reduced pressure. A saturated aqueous solution of NaHC03 and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure.
Purification by silica gel chromatography provided intermediate 82-c as a yellow solid.
Synthesis of Intermediate 83-a:
Figure imgf000118_0001
Scheme 83
To a solution of intermediate 82-c (2.0 g, 7.12 mmol) and TEA (1.98 ml, 14.23 mmol) in EtOH (3.50 ml) was added intermediate 43-d-HCI (1.30 g, 8.54 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure to provide intermediate 83-a as a yellow solid.
Synthesis of Compound 60:
Figure imgf000119_0001
83-a 84-a Compound 60
Scheme 84
Step 1: Intermediate 84-a
To a solution of intermediate 83-a (2.60 g, 7.12 mmol) and intermediate 29-i ( 1.00 g, 4.18 mmol) in 1,4-dioxane (20.9 ml) were sequentially added N,N- dimethylglycine (646 mg, 6.27 mmol), copper(I) iodide (398 mg, 2.09 mmol) and cesium carbonate (4.09 g, 12.5 mmol) . The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 84-a as a beige solid.
Step 2: Compound 60
Formamide ( 11.7 ml, 293 mmol) was added to intermediate 84-a (2.18 g, 4.18 mmol) and the reaction was stirred at 180 °C overnight. Water and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 60-2HCI as white solid. MS (m/z) M + H= 551.2 Synthesis of Compound 73:
Figure imgf000120_0001
83-a 85-a Compound 73
Scheme 85
Step 1: Intermediate 85-a
To a solution of intermediate 83-a (2.60 g, 7.12 mmol) and intermediate 34- d (1.0 g, 4.18 mmol) in 1,4-dioxane (20.9 ml) were sequentially added N,N- dimethylglycine (646 mg, 6.27 mmol), copper(I) iodide (398 mg, 2.09 mmol) and cesium carbonate (4.09 g, 12.5 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04/ filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 85-a as a beige solid.
Step 2: Compound 73
Intermediate 85-a (195.0 mg, 0.38 mmol) and trimethyl orthoformate ( 1.26 ml, 11.59 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (3.86 ml, 7.72 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous 13 000513
HCI/methanol gradient provided compound 73-HCI as white solid. MS (m/z) M + H= 534.1
Synthesis of Compound 95:
Figure imgf000121_0001
83-a 86-a Compound 95
Scheme 86
Step 1: Intermediate 86-a
To a solution of intermediate 83-a (200 mg, 0.54 mmol) and intermediate 31-d (177 mg, 0.65 mmol) in 1,4-dioxane (3.65 ml) were sequentially added N,N-dimethylglycine (127 mg, 1.23 mmol), copper(I) iodide (7.8 mg, 0.41 mmol) and cesium carbonate (714 mg, 2.19 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was
separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 86-a-HCI as a beige solid.
Step 2: Compound 95
Intermediate 86-a-HCI (284 mg, 0.54 mmol) and trimethyl orthoformate (3.90 ml, 35.6 mmol) were heated at 110 °C for 1 hour. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (3.91 ml, 27.4 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 95-2HCI as white solid. MS (m/z) M + H= 545.2
Synthesis of Intermediate 87-a:
Figure imgf000122_0001
Scheme 87
To a solution of intermediate 82-c (1.00 g, 3.56 mmol) and TEA (1.09 ml, 7.83 mmol) in EtOH (3.50 ml) was added intermediate 42-c (583 mg, 4.27 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of
ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel
chromatography provided intermediate 87-a as beige solid.
Synthesis of Compound 92:
Figure imgf000123_0001
87-a 88-a Compound 92
Scheme 88
Step 1: Intermediate 88-a
To a solution of intermediate 87-a (452 mg, 1.29 mmol) and intermediate 34-d (368 mg, 1.42 mmol) in 1,4-dioxane (5.20 ml) were sequentially added N,N-dimethylg!ycine (400 mg, 3.88 mmol), copper(I) iodide (247 mg, 1.29 mmol) and cesium carbonate (1.26 g, 3.88 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 88- a-HCI as a beige solid.
Step 2: Compound 92
Intermediate 88-a-HCI (124.0 mg, 0.25 mmol) and trimethyl orthoformate (935 pi, 7.63 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (2.54 ml, 5.09 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 92-2HCI as a beige solid. MS
(m/z) M + H= 518.2 Synthesis of intermediate 89-a:
Figure imgf000124_0001
82-c 89-a
Scheme 89
To a solution of intermediate 82-c (2.0 g, 7.12 mmol) and TEA (2.18 ml, 15.65 mmol) in EtOH (7.12 ml) was added intermediate isopropylhydrazine hydrochloride (944 mg, 8.54 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure.
Purification by silica gel chromatography provided intermediate 89-a as a beige solid.
Synthesis of Compound 98:
Figure imgf000124_0002
89-a 90-a Compound 98
Scheme 90 Step 1: Intermediate 90-a
To a solution of intermediate 89-a (400 mg, 1.24 mmol) and intermediate 34-d (352 mg, 1.36 mmol) in 1,4-dioxane (4.9 ml) were sequentially added N,N-dimethylglycine (383 mg, 3.71 mmol), copper(I) iodide (236 mg, 1.24 mmol) and cesium carbonate (1.21 g, 3.71 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 90- a-HCI as beige solid.
Step 2: Compound 98
Intermediate 90-a-HCI (208 mg, 0.45 mmol) and trimethyl orthoformate (1.47 ml, 13.4 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (4.48 ml, 8.96 mmol). The mixture was stirred at room temperature for 3 days and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 98-2HCI as a beige solid. MS (m/z) M + H= 492.2
Synthesis of Intermediate 91-d:
Figure imgf000125_0001
91-a 91-b 91-c 91-d
Scheme 91 Step 1: Intermediate 91-b
To a suspension of 4-bromo-3-fluorobenzoic acid (15.0 g, 68.5 mmol) in dichloromethane (342.0 ml) was added DMF ( 106.0 μΙ, 1.37 mmol) and oxalylyl chloride (8.99 ml, 103.0 mmol). The solution was then stirred at room temperature for 3 hours. Volatiles were removed under reduced pressure to provide intermediate 91-b as a yellow solid.
Step 2: Intermediate 91-c
To a solution of intermediate 91-b (16.27 g, 68.5 mmol) in toluene (85.0 ml) and THF (8.5 ml) cooled to -10 °C were sequentially added malononitrile (4.75 g, 71.9 mmol) and DIPEA (23.93 ml, 137 mmol) drop wise over a period of 15 minutes. The reaction was stirred at 0°C for 2 hours and room temperature for an additional 2 hours. Volatiles were removed under reduced pressure. Ethyl acetate and IN HCI were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated in vacuo to provide intermediate 91-c as a yellow solid.
Step 3: Intermediate 91-d
To a solution of intermediate 91-c (18.29 g, 68.5 mmol) in acetonitrile
(247.0 ml) and MeOH (27.4 ml) cooled to 0 °C were sequentially added DIPEA (14.36 ml, 82.0 mmol) and a 2M solution of TMS-Diazomethane in hexanes (37.7 ml, 75.0 mmol). The reaction was then stirred for 5 hours at room temperature. Acetic acid (1.17 ml, 20.55 mmol) was then added and the reaction was stirred for an additional 30 minutes. Volatiles were removed under reduced pressure. A saturated aqueous solution of NaHC03 and ethyl acetate was added, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated in vacuo. Purification by silica gel chromatography provided intermediate 91-d as a beige solid.
Synthesis of Intermediate 92-a:
Figure imgf000127_0001
Scheme 92
To a solution of intermediate 91-d (2.0 g, 7.12 mmol) and TEA (1.98 ml, 14.23 mmol) in EtOH (3.50 ml) was added intermediate 43-d HCI (1.30 g, 8.54 mmol) and the reaction was then stirred for 2 hours at 100 °C. Volatiles were removed under reduced pressure. A saturated aqueous solution of ammonium chloride and ethyl acetate were added to the residue, the organic layer was separated, washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel
chromatography provided intermediate 92-a as a beige solid.
Figure imgf000127_0002
Scheme 93
Step 1: Intermediate 93-a
To a solution of intermediate 92-a (2.60 g, 7.12 mmol) and intermediate 29-i (1.0 g, 4.18 mmol) in 1,4-dioxane (20.90 ml) were sequentially added N,N- dimethylglycine (646 mg, 6.27 mmol), copper(I) iodide (398 mg, 2.09 mmol) and cesium carbonate (4.09 g, 12.54 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 93-a as a beige solid.
Step 2: Compound 59
Formamide (11.67 ml, 293.0 mmol) was added to intermediate 93-a (2.18 g, 4.18 mmol) and the reaction was stirred at 180 °C overnight then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, washed with brine, dried over MgS04, filtered and
concentrated under reduced pressure. Purification by reverse phase
chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 59-2HCI as a yellow solid. MS (m/z) M + H= 551.1
Synthesis of Compound 72:
Figure imgf000128_0001
92-a 94-a Compound 72
Scheme 94 Step 1: Intermediate 94-a
To a solution of intermediate 92-a (291.0 mg, 0.79 mmol) and intermediate 34-d (227.0 mg, 0.87 mmol) in 1,4-dioxane (3.2 ml) were sequentially added Ν,Ν-dimethylglycine (247.0 mg, 2.39 mmol), copper(I) iodide (152.0 mg, 0.79 mmol) and cesium carbonate (780 mg, 2.39 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided intermediate 94-a-HCI as a beige solid.
Step 2: Compound 72:
Intermediate 94-a-HCI (56.0 mg, 0.11 mmol) and trimethyl orthoformate (363 μΙ, 3.32 mmol) were heated at 110 °C for 3 hours. Excess trimethyl orthoformate was removed in vacuo and the residue was treated with 7.0 N ammonia in MeOH (1.10 ml, 2.21 mmol). The mixture was stirred at room temperature overnight and volatiles were removed under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 72-2HCI as a white solid. MS (m/z) M + H= 534.2
Synthesis of Compound 68:
Figure imgf000130_0001
17-a
Scheme 95
To a solution of intermediate 17-a (300.0 mg, 0.80 mmol) and intermediate 39-b ( 177.0 mg, 0.80 mmol) in 1,4-dioxane (3.6 ml) were sequentially added N,N-dimethylglycine (372.0 mg, 3.61 mmol), copper(I) iodide (229.0 mg, 1.20 mmol) and cesium carbonate ( 1.04 g, 3.21 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 68-2HCI as a beige solid. MS (m/z) M + H= 514.2
Synthesis of Compound 69:
Figure imgf000131_0001
17-a
Scheme 96
To a solution of intermediate 17-a (300.0 mg, 0.80 mmol) and intermediate 40-b (177.0 mg, 0.80 mmol) in 1,4-dioxane (3.6 ml) were sequentially added N,N-dimethylglycine (372.0 mg, 3.61 mmol), copper(I) iodide (229.0 mg, 1.20 mmol) and cesium carbonate (1.04 g, 3.21 mmol). The reaction mixture was stirred at reflux overnight, cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04/ filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% aqueous HCI/methanol gradient provided compound 69-2HCI as a beige solid. MS (m/z) M + H= 514.2
Synthesis of Compound 63:
Figure imgf000132_0001
17-a
Scheme 97
A solution of intermediate 17-a (437 mg, 1.17 mmol), intermediate 11-c (255 mg, 1.17 mmol), quinolin-8-ol (34 mg, 0.23 mmol), copper (I) iodide (44.0 mg, 0.23 mmol) and cesium carbonate (761 mg, 2.33 mmol), in dimethylacetamide (1.2 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 140 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 63-HCI as a yellow solid. MS (m/z) M + H= 512.2
Synthesis of Compound 67:
Figure imgf000133_0001
Figure imgf000133_0002
Scheme 98
Step 1: Intermediate 98-b
To a solution of 3-bromo-5-fluorophenol, 98-a (25.00 g, 131.0 mmol), in acetone (654 ml) were sequentially added K2C03 (27.10 g, 196.0 mmol) and chloro(methoxy)methane (11.59 g, 144.0 mmol). The reaction was stirred at room temperature for 2 hours and then filtered. The filtrate was concentrated under reduced pressure to provide intermediate 98-b as a yellow oil.
Step 2: Intermediate 98-d
To a solution of intermediate 98-b (2.00 g, 8.51 mmol) in DMF (17.02 ml), were sequentially added diethylamine (975 μΙ, 9.36 mmol), copper(I)iodide (65 mg, 0.34 mmol) and 5-ethynyl-l-methyl-lH-imidazole 98-c (948 mg, 8.93 mmol). After copper(I)iodide has completely dissolved, Dichlorobis(triphenylphosphine)palladium(II) (119 mg, 0.17 mmol) was added and the reaction was then stirred at 100 °C overnight and then cooled to room temperature. Water and ethyl acetate were added, the organic layer was separated, the aqueous layer was extracted twice with ethyl acetate, the combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by silica gel chromatography provided intermediate 98-d as a beige solid
Step 3: Intermediate 98-e
A solution of intermediate 98-d (600.0 mg, 2.3 mmol) in methanol was treated with 10% palladium on carbon (245.0 mg, 0.01 mmol) and purged with H2. The solution was stirred under H2 ( l atm) overnight before being filtered through celite. The filtrate was concentrated in vacuo to provide intermediate 98-e as a yellow oil.
Step 4: Intermediate 98-f
To a solution of intermediate 98-e (2.4 g, 9.08 mmol) in MeOH (17 ml) was added 4N HCI in dioxane (2.76 ml, 91 mmol). The reaction mixture was stirred at room temperature for 2 hours. Volatiles were removed under reduced pressure, diethyl ether was added to the residue; a precipitate formed and was collected by filtration to provide intermediate 98-f-HCI as a white solid.
Step 5: Compound 67
A solution of intermediate 17-a (292 mg, 0.78 mmol), intermediate 98-f-HCI (200 mg, 0.78 mmol), tetramethylheptane-3,5-dione (287 mg, 1.56 mmol), copper (I) iodide (148 mg, 0.78 mmol) and cesium carbonate (762 mg, 2.33 mmol), in NMP (3.9 ml), was degassed with argon for 10 minutes, heated in a sealed tube at 120 °C overnight and then cooled to room temperature, diluted with ethyl acetate and filtered over celite. A saturated aqueous solution of ammonium chloride was added to the filtrate, the organic layer was separated, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with brine, dried over MgS04, filtered and concentrated under reduced pressure. Purification by reverse phase chromatography eluting with a 1% HCI/methanol gradient provided compound 67-2HCI as a yellow solid. MS (m/z) M + H= 514.3
Compounds 7, 8, 11, 13, 14, 19, 21, 24 to 28, 33, 34, 37 to 51, 53, 56, 61, 62, 64, 66, 70, 71, 74-77, 79 to 84, 86 to 89, 94, 96, 104, 105, 107 to 112, 115, 116, 119, 121, 122, 123, 124, 126, 127, 131, 132, and 133 were prepared using similar methods to those described above.
Table 1 summarizes representative compound of Formula 1.
Table 1: Example Compounds of Formula 1
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
Figure imgf000138_0001
Figure imgf000139_0001
Figure imgf000140_0001
Figure imgf000141_0001
Figure imgf000142_0001
Figure imgf000143_0001
Figure imgf000144_0001
Figure imgf000145_0001
Figure imgf000146_0001
Figure imgf000147_0001
Figure imgf000148_0001
Figure imgf000149_0001
Figure imgf000150_0001
Figure imgf000151_0001
Figure imgf000152_0001
Figure imgf000153_0001
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
ı55
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
ı64
Figure imgf000166_0001
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000169_0001
Figure imgf000170_0001
Figure imgf000171_0001
Figure imgf000172_0001
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
ı77 Kinase Binding
Btk Kinase Inhibition Assay
Fluorescence polarization-based kinase assays were performed in 384 well- plate format using histidine tagged recombinant human full-length Bruton Agammaglobulinemia Tyrosine Kinase (Btk) and a modified protocol of the KinEASE™ FP Fluorescein Green Assay supplied from Millipore. Kinase reaction were performed at room temperature for 60 minutes in presence of 250 μΜ substrate, 10 μΜ ATP and variable test article concentrations. The reaction was stopped with EDTA/kinase detection reagents and the
polarization measured on a Tecan 500 instrument. From the dose-response curve obtained, the IC50 was calculated using Graph Pad Prisms® using a non linear fit curve. The Km for ATP on each enzyme was experimentally determined and the Ki values calculated using the Cheng-Prusoff equation (see: Cheng Y, Prusoff WH. (1973) Relationship between the inhibition constant (Kl) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction". Biochem Pharmacol 22 (23) : 3099- 108). k| values are reported in Tables 2 :
Table 2: Inhibition of Btk
Figure imgf000180_0001
a - Ki<100 nM; b - 100 nM<Ki<1000 nM, c - Ki>1000 nM Splenic Cell Proliferation Assay
Splenocytes were obtained from 6 week old male CD1 mice (Charles River Laboratories Inc.). Mouse spleens were manually disrupted in PBS and filtered using a 70um cell strainer followed by ammonium chloride red blood cell lysis. Cells were washed, resuspended in Splenocyte Medium (HyClone RPMI supplemented with 10% heat-inactivated FBS, 0.5X non-essential amino acids, lOmM HEPES, 50uM beta mercaptoethanol) and incubated at 37 °C, 5% C02 for 2h to remove adherent cells. Suspension cells were seeded in 96 well plates at 50,000 cells per well and incubated at 37°C, 5% C02 for lh. Splenocytes were pre-treated in triplicate with 10,000 nM curves of Formula 1 compounds for lh, followed by stimulation of B cell proliferation with 2.5ug/ml anti-IgM F(ab')2 (Jackson ImmunoResearch) for 72h. Cell proliferation was measured by Cell Titer-Glo Luminescent Assay (Promega). EC50 values (50% proliferation in the presence of compound as compared to vehicle treated controls) were calculated from dose response compound curves using GraphPad Prism Software.
EC50 values are reported in Table 3 :
Table 3: Inhibition of splenic cell proliferation
Figure imgf000182_0001
a - EC50< 100 nM; b -100 nM<EC50<1000 nM, c - EC50> 1000 nM Methods: Mouse Arthus
Mouse Arthus studies were conducted as reported in Braselmann S, Taylor V, Zhao H, Wang S, Sylvain C, Baluom M, Qu K, Herlaar E, Lau A, Young C, Wong BR, Lovell S, Sun T, Park G, Argade A, Jurcevic S, Pine P, Singh R, Grossbard EB, Payan DG, Masuda ES: R406 an orally available spleen tyrosine kinase inhibitor blocks fc receptor signaling and reduces immune- complex mediated inflammation. J Pharmacol Exp Ther, 2006, 319 :998- 1008.
In summary, female Balb/c mice (6-7 weeks on arrival) were habituated to the animal facility for at least 4 days. On the day of the experiment, animals were pre-treated (t= minus 1 h) with compound or vehicle alone by gavage (PO). At t=0, animals were injected intravenously (IV; 0.1 mL/mouse) with saline containing chicken ovalbumin and Evan's blue (10 mg/mL of each). Ten minutes later (t= 10 min), animals were anesthesized with isoflurane, the dorsal surface was shaved and rabbit anti-chicken ovalbumin antibody was then injected intradermally at one site on the right side of the animal (25 μ< in 30 μί). The same amount of isotype control antibody was then injected on the left side.
The animals were then returned to their home cage and skin punches (8 mm) were collected from each injection site four hours later. The samples were placed in 1 mL formamide overnight at 80 degrees C (1 skin biopsy per 1 mL formamide in a glass tube). The amount of Evan's blue in the
formamide solution was then assessed by spectrophotometry (630 nm) as a measure of serum extravasation into the dermis.
Compounds 14, 15, 24, 46, 50, 54, 58, 59, 62, 71, 78, 79, 82, 85, 90, 100, 102, 103, 106, 107, 108, 117, and 125 demonstrated efficacy when
administered by oral gavages at 30 mg/kg.
Mouse CIA model was performed using the methods described by Trentham DE, Townes AS, Kang AH. Autoimmunity to Type II Collagen : An
Experimental Model of Arthritis. J Exp Med 1977; 857-868, and Bendele AM. Animal Models of Rheumatoid Arthritis. J Musculoskel Interact 2001; 377- 385.
In summary, male BlORlll mice (7-9 wks on arrival) were habituated to the animal facility for at least 4 days. On experimental day 0 mice were anaesthetized with isoflurane and the dorsal surface was shaved. Collagen, emulsified in Freund's complete adjuvant (CFA) supplemented with additional mycobacterium tuberculosis (TB) H37Ra, was injected intradermal^ at the base of the tail (0.15 mL / animal; 2 mg/mL collagen and 2.5 mg/mL TB in CFA). This CFA treatment was repeated on day 15.
From day 15 to the end of the study animals were scored daily for signs of arthritis. On the first day of disease (RA Day 1) animals were recruited to the study and grouped using a balanced design based on arthritis score. Once recruited, animals were weighed and dosed twice daily by gavage (PO, BID). Recruited animals were then scored twice a week on RA days 1, 5, 8 and 12. At the end of the study (RA day 12) animals were weighed and scored.
Compounds 14, 58, 78, 85 and 102 demonstrated efficacy when
administered by oral gavages at 30 mg/kg (BID).

Claims

1. A compound of Formula 1 :
Figure imgf000185_0001
wherein R1 is selected from the group consisting of:
1) hydrogen,
2) alkyl,
3) heteroalkyl,
4) carbocyclyl,
5) heterocyclyl, and
6) -C(0)R4; wherein the alkyl, heteroalkyl, carbocyclyl and heterocyclyl may be further substituted by the groups consisting of:
1) hydroxy,
2) alkoxy,
3) alkyl,
4) -OC(0)R4,
5) -OC(0)NR5R6,
6) -C(0)R4, 7) -C(0)NR5R6,
8) -NR5R6,
9) -NR2C(0)R4,
10) -NR2S(0)nR4 and
11) -NR2C(0)NR5R6;
wherein Y is
wherein Z is
wherein Y-Z-
Figure imgf000186_0001
X1 and X2 are independently selected from the group consisting of hydrogen, halogen and cyano; n is an integer from 0 to 2; m is an integer from 0 to 2; m' is an integer from 0 to 2;
W is selected from the group consisting of:
1) alkyl,
2) aralkyl,
3) heteroaralkyl,
4) -OR3, 5) -0C(0)R4,
6) -OC(0)NR5R6,
7) -CH2O-R4,
8) -NR5R6,
9) -NR2C(0)R4,
10) -NR2S(0)nR4 and
11) -l\IR2C(0)NR5R6; wherein the alkyl, aralkyi and heteroaralkyl may be further substituted; R2 is hydrogen or alkyl;
R3 is selected from the group consisting of: substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heteroalkyl, substituted or
unsubstituted carbocyclyl, heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyi, and substituted or unsubstituted heteroaralkyl;
R4 is selected from the group consisting of: substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted heteroalkyl, substituted or
unsubstituted carbocyclyl, heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted aralkyi, and substituted or unsubstituted heteroaralkyl;
R5 and R6 are independently selected from the group consisting of: hydrogen, alkyl, alkenyl, alkynyl, heteroalkyl, carbocyclyl, heterocyclyl, aryl and heteroaryl; alternatively R5 and R6 are fused to form a 3 to 8 membered heterocyclyl ring system. The compound of claim 1, wherein W is -OR3, and wherein R3 is selected from the group consisting of: substituted or unsubstituted aralkyi and substituted or unsubstituted heteroaralkyl.
The compound of claim 1, wherein W is selected from the group consisting of:
Figure imgf000188_0001
Figure imgf000189_0001
4. The compound according to claim 1, wherein W is selected from the group consisting of:
Figure imgf000189_0002
5. The compound according to any one of claims 1 to 4, wherein R1 is selected from the group consisting of:
Figure imgf000189_0003
Figure imgf000190_0001
and
6. The compound according to claim 5, wherein Rl is selected from the group consisting of:
Figure imgf000190_0002
7. The compound according to any one of claims 1 to 6, wherein Y is selected from the group consisting of:
Figure imgf000190_0003
8. The compound according to any one of claims 1 to 5, wherein Z is selected from the group consisting of:
Figure imgf000190_0004
9. The compound according to claim 8, wherein Z is :
Figure imgf000191_0001
10. The compound according to any one of claims 1 to 9, wherein Y-Z-W is selected from the group consisting of:
Figure imgf000191_0002
11. A compound selected from the group consisting of:
Figure imgf000191_0003
Figure imgf000192_0001
Figure imgf000193_0001
Figure imgf000194_0001
Figure imgf000195_0001
Figure imgf000196_0001
Figure imgf000197_0001
Figure imgf000198_0001
Figure imgf000199_0001
Figure imgf000200_0001
Figure imgf000201_0001
Figure imgf000202_0001
Figure imgf000203_0001
Figure imgf000204_0001
Figure imgf000205_0001
Figure imgf000206_0001
Figure imgf000207_0001
206
Figure imgf000208_0001
Figure imgf000209_0001
Figure imgf000210_0001
Figure imgf000211_0001
Figure imgf000212_0001
211
Figure imgf000213_0001
Figure imgf000214_0001
Figure imgf000215_0001
Figure imgf000216_0001
Figure imgf000217_0001
Figure imgf000218_0001
Figure imgf000219_0001
Figure imgf000220_0001
Figure imgf000221_0001
Figure imgf000222_0001
Figure imgf000223_0001
Figure imgf000224_0001
Figure imgf000225_0001
Figure imgf000226_0001
225
Figure imgf000227_0001
Figure imgf000228_0001
Figure imgf000229_0001
Figure imgf000230_0001
Figure imgf000231_0001
Figure imgf000232_0001
Figure imgf000233_0001
Figure imgf000234_0001
PCT/CA2013/000513 2012-05-31 2013-05-28 Protein kinase inhibitors WO2013177668A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
KR1020197011438A KR20190043648A (en) 2012-05-31 2013-05-28 Protein kinase inhibitors
CA2874211A CA2874211A1 (en) 2012-05-31 2013-05-28 Protein kinase inhibitors
US14/404,497 US9796716B2 (en) 2012-05-31 2013-05-28 Selective inhibitors of Tec and Src protein kinase families
EP13796590.1A EP2855484A4 (en) 2012-05-31 2013-05-28 Protein kinase inhibitors
RU2014145285A RU2678767C2 (en) 2012-05-31 2013-05-28 Protein kinase inhibitors
CN201380028248.2A CN104379586B (en) 2012-05-31 2013-05-28 Kinases inhibitor
KR1020147036372A KR101972990B1 (en) 2012-05-31 2013-05-28 Protein kinase inhibitors
IN2338MUN2014 IN2014MN02338A (en) 2012-05-31 2013-05-28
BR112014029718A BR112014029718A2 (en) 2012-05-31 2013-05-28 protein kinase inhibitors
JP2015514295A JP6175495B2 (en) 2012-05-31 2013-05-28 Protein kinase inhibitor
HK15109229.7A HK1208460A1 (en) 2012-05-31 2015-09-18 Protein kinase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CA2779184A CA2779184A1 (en) 2012-05-31 2012-05-31 Protein kinase inhibitors
CA2,779,184 2012-05-31
CA2813299A CA2813299A1 (en) 2013-04-17 2013-04-17 Protein kinase inhibitors
CA2,813,299 2013-04-17

Publications (1)

Publication Number Publication Date
WO2013177668A1 true WO2013177668A1 (en) 2013-12-05

Family

ID=49672221

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CA2013/000513 WO2013177668A1 (en) 2012-05-31 2013-05-28 Protein kinase inhibitors

Country Status (11)

Country Link
US (1) US9796716B2 (en)
EP (1) EP2855484A4 (en)
JP (1) JP6175495B2 (en)
KR (2) KR101972990B1 (en)
CN (1) CN104379586B (en)
BR (1) BR112014029718A2 (en)
CA (1) CA2874211A1 (en)
HK (1) HK1208460A1 (en)
IN (1) IN2014MN02338A (en)
RU (1) RU2678767C2 (en)
WO (1) WO2013177668A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105764906A (en) * 2013-11-26 2016-07-13 制药科学公司 Protein kinase inhibitors
WO2016185160A1 (en) * 2015-05-21 2016-11-24 The University Court Of The University Of Edinburgh Compounds
KR20170023156A (en) * 2014-07-07 2017-03-02 이터니티 바이오사이언스 인코퍼레이티드 Aminopyridazinone compounds as protein kinase inhibitors
EP3071573A4 (en) * 2013-11-21 2017-06-14 Pharmascience Inc. Protein kinase inhibitors
WO2017134685A3 (en) * 2016-02-02 2018-06-28 Sun Pharma Advanced Research Company Limited Novel hydrazino compounds as btk inhibitors
CN109369654A (en) * 2018-11-20 2019-02-22 山东大学 1,3- bis- substituted-4-amino Pyrazolopyrimidines and its preparation method and application

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3025813A1 (en) * 2015-05-27 2016-12-01 Gb005, Inc. Inhibitors of the tec kinase enzyme family
US10927087B2 (en) 2015-12-07 2021-02-23 Dic Corporation Method for producing polymerizable compound
CN111454268B (en) * 2019-01-18 2023-09-08 明慧医药(上海)有限公司 Cyclic molecules as inhibitors of bruton's tyrosine kinase
WO2023083373A1 (en) * 2021-11-15 2023-05-19 微境生物医药科技(上海)有限公司 Compound used as src inhibitor

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2385747A1 (en) * 1999-09-17 2001-03-22 Gavin C. Hirst Pyrazolopyrimidines as therapeutic agents
CA2440724A1 (en) * 2001-03-22 2002-10-17 Abbott Gmbh & Co. Kg Pyrazolopyrimidines as therapeutic agents

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20020063854A (en) 1999-09-17 2002-08-05 애보트 게엠베하 운트 콤파니 카게 Kinase inhibitors as therapeutic agents
SK13752001A3 (en) * 1999-12-27 2002-07-02 Japan Tobacco, Inc. Fused-ring compounds and use thereof as drugs
AUPR392301A0 (en) 2001-03-23 2001-04-26 University Of Newcastle Research Associates Limited, The Protein phosphatase inhibitors
CA2450777C (en) * 2001-08-10 2013-04-09 Novartis Ag Use of c-src inhibitors alone or in combination with sti571 for the treatment of leukaemia
LT2530083T (en) * 2006-09-22 2016-09-26 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
EP2561875A3 (en) * 2007-03-28 2013-06-12 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase
US7718662B1 (en) * 2009-10-12 2010-05-18 Pharmacyclics, Inc. Pyrazolo-pyrimidine inhibitors of bruton's tyrosine kinase
AU2010307006B2 (en) * 2009-10-13 2016-08-11 Ligand Pharmaceuticals Inc. Hematopoietic growth factor mimetic small molecule compounds and their uses
JP6139782B2 (en) * 2013-05-21 2017-05-31 チャンスー メドリューション リミテッド Substituted pyrazolopyrimidine compounds, and pharmaceutically acceptable salts thereof, and solvates, stereoisomers, and tautomers thereof, and pharmaceutical compositions containing them
CN103787839B (en) 2014-01-21 2015-12-02 苏州昊帆生物科技有限公司 The method of synthesis 2,3,4,5,6-Pentafluorophenol

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2385747A1 (en) * 1999-09-17 2001-03-22 Gavin C. Hirst Pyrazolopyrimidines as therapeutic agents
CA2440724A1 (en) * 2001-03-22 2002-10-17 Abbott Gmbh & Co. Kg Pyrazolopyrimidines as therapeutic agents

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 330786-85-1 *
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 330789-30-5 *
DATABASE CAPLUS *
DATABASE CAPLUS Database accession no. 30786-85-1 *
DATABASE CAPLUS Database accession no. 330786-86-2 *
DATABASE CAPLUS Database accession no. 330786-88-4 *
DATABASE CAPLUS Database accession no. 330786-89-5 *
DATABASE CAPLUS Database accession no. 330786-91-9 *
DATABASE CAPLUS Database accession no. 330787-66-1 *
DATABASE CAPLUS Database accession no. 330787-69-4 *
DATABASE CAPLUS Database accession no. 330789-24-7 *
DATABASE CAPLUS Database accession no. 330789-30-5 *
DATABASE CAPLUS Database accession no. 330789-31-6 *
DATABASE CAPLUS Database accession no. 330789-86-1 *
DATABASE CAPLUS Database accession no. 330789-88-3 *
DATABASE CAPLUS Database accession no. 330789-90-7 *
DATABASE CAPLUS Database accession no. 330792-99-9 *
DATABASE CAPLUS Database accession no. 330793-00-5 *
See also references of EP2855484A4 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3071573A4 (en) * 2013-11-21 2017-06-14 Pharmascience Inc. Protein kinase inhibitors
EP3074401A4 (en) * 2013-11-26 2017-05-10 Pharmascience Inc. Protein kinase inhibitors
EP3074401A1 (en) * 2013-11-26 2016-10-05 Pharmascience Inc. Protein kinase inhibitors
CN105764906A (en) * 2013-11-26 2016-07-13 制药科学公司 Protein kinase inhibitors
JP2017522302A (en) * 2014-07-07 2017-08-10 エタニティ バイオサイエンス インコーポレーテッド Aminopyridazinone compounds as protein kinase inhibitors
KR20170023156A (en) * 2014-07-07 2017-03-02 이터니티 바이오사이언스 인코퍼레이티드 Aminopyridazinone compounds as protein kinase inhibitors
KR102073797B1 (en) 2014-07-07 2020-02-05 지앙수 헨그루이 메디슨 컴퍼니 리미티드 Aminopyridazinone compounds as protein kinase inhibitors
WO2016185160A1 (en) * 2015-05-21 2016-11-24 The University Court Of The University Of Edinburgh Compounds
CN107849050A (en) * 2015-05-21 2018-03-27 爱丁堡大学董事会 Compound
JP2018515563A (en) * 2015-05-21 2018-06-14 ザ ユニバーシティー コート オブ ザ ユニバーシティー オブ エジンバラThe University Court Of The University Of Edinburgh Compound
US10294227B2 (en) 2015-05-21 2019-05-21 University Court Of The University Of Edinburgh Compounds
CN107849050B (en) * 2015-05-21 2020-10-13 爱丁堡大学董事会 Compound (I)
WO2017134685A3 (en) * 2016-02-02 2018-06-28 Sun Pharma Advanced Research Company Limited Novel hydrazino compounds as btk inhibitors
CN109369654A (en) * 2018-11-20 2019-02-22 山东大学 1,3- bis- substituted-4-amino Pyrazolopyrimidines and its preparation method and application

Also Published As

Publication number Publication date
EP2855484A4 (en) 2015-12-16
JP6175495B2 (en) 2017-08-02
CN104379586B (en) 2018-01-16
US9796716B2 (en) 2017-10-24
KR20150015527A (en) 2015-02-10
KR101972990B1 (en) 2019-04-26
HK1208460A1 (en) 2016-03-04
US20150191473A1 (en) 2015-07-09
JP2015518009A (en) 2015-06-25
EP2855484A1 (en) 2015-04-08
CA2874211A1 (en) 2013-12-05
IN2014MN02338A (en) 2015-08-14
CN104379586A (en) 2015-02-25
KR20190043648A (en) 2019-04-26
BR112014029718A2 (en) 2017-06-27
RU2014145285A (en) 2016-07-20
RU2678767C2 (en) 2019-02-01

Similar Documents

Publication Publication Date Title
JP6175495B2 (en) Protein kinase inhibitor
JP6001053B2 (en) Protein kinase inhibitor
US9795608B2 (en) Protein kinase inhibitors
CA2667487C (en) Imidazo[1,2-b]pyridazine and pyrazolo[1,5-a]pyrimidine derivatives and their use as protein kinase inhibitors
EP2694515B1 (en) Protein kinase inhibitors
CA2833701A1 (en) Protein kinase inhibitors
CA2779184A1 (en) Protein kinase inhibitors
CA2813299A1 (en) Protein kinase inhibitors
CA2833867A1 (en) Protein kinase inhibitors
WO2014029007A1 (en) Protein 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: 13796590

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2013796590

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2874211

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2015514295

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 14404497

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20147036372

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2014145285

Country of ref document: RU

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014029718

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112014029718

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20141127