WO2015073267A1 - Substituted heteroaryl compounds and methods of use - Google Patents

Substituted heteroaryl compounds and methods of use Download PDF

Info

Publication number
WO2015073267A1
WO2015073267A1 PCT/US2014/063971 US2014063971W WO2015073267A1 WO 2015073267 A1 WO2015073267 A1 WO 2015073267A1 US 2014063971 W US2014063971 W US 2014063971W WO 2015073267 A1 WO2015073267 A1 WO 2015073267A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
cycloalkyl
membered heterocyclyl
disease
alkyl
Prior art date
Application number
PCT/US2014/063971
Other languages
French (fr)
Inventor
Ning Xi
Minxiong LI
Xiaobo Li
Original Assignee
Calitor Sciences, Llc
Sunshine Lake Pharma Co., Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Calitor Sciences, Llc, Sunshine Lake Pharma Co., Ltd. filed Critical Calitor Sciences, Llc
Publication of WO2015073267A1 publication Critical patent/WO2015073267A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention provides novel substituted heteroaryl compounds, and salts thereof, which are useful in the treatment of proliferative disease, autoimmune disease, allergic disease, inflammatory disease, transplantation rejection, and other diseases, in mammals.
  • this invention relates to compounds that modulate the activity of Janus kinases, resulting in the modulation of inter- and/or intra-cellular signaling.
  • This invention also relates to a method of using such compounds in the treatment of proliferative disease, autoimmune disease, allergic disease, inflammatory disease, transplantation rejection, and other diseases in mammals, especially humans, and to pharmaceutical compositions containing such compounds.
  • Cytokines are low-molecular weight polypeptides or glycoproteins that stimulate biological responses in virtually all cell types. Cytokines are critical for normal cell differentiation, proliferation, activation and immunoregulation but also contribute to growth of malignant cells and drive immune-mediated disease. A major subset of immunoregulatory cytokines, roughly 60, use the type I and type II cytokine receptors and pharmacological targeting of these cytokines/cytokines receptors is useful in treating immune-related diseases, skin disorders, myeloid proliferative disorders, cancer, and other diseases. (O'Sullivan et al, Mol. Immunol, 2007, 44:2497; Murray J., Immunol, 2007, 178:2623).
  • cytokines controls the growth and differentiation of hematopoietic cells and orchestrates all aspects of immune response.
  • the family of cytokines that bind type I and type II cytokine receptors includes interleukins, interferons, and colony stimulating factor, as well as classic hormones such erythropoietin, prolactin and growth hormone. Binding of a cytokine to its cell surface receptor initiates intracellular signaling cascades that transduce the extracellular signal to the nucleus, ultimately leading to changes in gene expression.
  • Janus kinase family of protein tyrosine kinases (JAKs) and Signal Transducers and Activators of Transcription (STATs) ["Jakinibs: A New Class of Kinase Inhibitors in Cancer and Autoimmune Disease.” Curr Opin Pharmacol. 2012 August; 12(4): 464-470].
  • JAK Janus kinase
  • STATs Signal Transducers and Activators of Transcription
  • Cytokines bind to their receptors, causing receptor dimerization, and this enables JAKs to phosphorylate each other as well as specific tyrosine motifs within the cytokine receptors.
  • STATs that recognize these phosphotyrosine motifs are recruited to the receptor, and are then themselves activated by a JAK-dependent tyrosine phosphorylation event. Upon activation, STATs dissociate from the receptors, dimerize, and translocate to the nucleus to bind to specific DNA sites and alter transcription.
  • JAKl Janus kinase-1
  • JAK2 Janus kinase-2
  • JAK3 Janus kinase, leukocyte; JAKL; L-JAK and Janus kinase-3)
  • TYK2 protein-tyrosine kinase 2
  • JAKl, JAK2 and TYK2 are ubiquitously expressed
  • JAK3 is reported to be preferentially expressed in natural killer (NK) cells and not resting T cells ("Biology and significance of the JAK/STAT signaling pathways.” Growth Factors, April 2012; 30(2): 88).
  • JAKl is essential for signaling for certain type I and type II cytokines. It interacts with the common gamma chain (yc) of type I cytokine receptors to elicit signals from the IL-2 receptor family, the IL-4 receptor family, the gpl30 receptor family. It is also important for transducing a signal by type I (IFN- ⁇ / ⁇ ) and type II (IFN- ⁇ ) interferons, and members of the IL-10 family via type II cytokine receptors.
  • yc common gamma chain
  • JAKl is functionally and physically associated with the type I interferon (e g., IFNalpha), type II interferon (e.g., IFNgamma), IL-2 and IL-6 cytokine receptor complexes. Furthermore, characterization of tissues derived from JAKl knockout mice demonstrated critical roles for this kinase in the IFN, IL-IO, IL- 2/IL-4, and IL-6 pathways.
  • JAKl expression of JAKl in cancer cells enables individual cells to contract, potentially allowing them to escape their tumor and metastasize to other parts of the body. Elevated levels of cytokines which signal through JAKl have been implicated in a number of immune and inflammatory diseases. JAKl or JAK family kinase inhibitors may be useful for modulating or treating in such diseases. (Kisseleva et al, 2002, Gene 285: 1-24; Levy et al, 2005, Nat. Rev. Mol. Cell Biol. 3:651-662).
  • a humanized monoclonal antibody targeting the IL-6 pathway was approved by the European Commission for the treatment of moderate-to-severe rheumatoid arthritis (Scheinecker et al., 2009, Nat. Rev. Drug Discov. 8:273-274).
  • JAK2 is implicated in signaling by members of the type II cytokine receptor family (e.g. interferon receptors), the GM-CSF receptor family, the gpl30 receptor family. JAK2 signaling is implicated in signaling by members of the type II cytokine receptor family (e.g. interferon receptors), the GM-CSF receptor family, the gpl30 receptor family. JAK2 signaling is implicated in signaling by members of the type II cytokine receptor family (e.g. interferon receptors), the GM-CSF receptor family, the gpl30 receptor family. JAK2 signaling is
  • SUBSTITUTE SHEET RULE 26 activated downstream from the prolactin receptor.
  • JAK2V617F an acquired activating JAK2 mutation
  • the mutant JAK2 protein is able to activate downstream signaling in the absence of cytokine stimulation, resulting in autonomous growth and/or hypersensitivity to cytokines and is believed to play a role in driving these diseases. Additional mutations or translocations resulting dysregulated JAK2 function have been described in other malignancies (Ihle J.N. and Gilliland D.G., Curr. Opin. Genet.
  • JAK2 JAK2
  • Inhibitors of JAK2 have been described to be useful in myeloproliferative diseases (Santos et al, Blood, 2010, 115: 1131; Barosi G. and Rosti V., Curr. Opin. Hematol, 2009, 16: 129, Atallah E. and Versotvsek S., Exp. Rev. Anticancer Ther. 2009, 9:663).
  • JAK3 associates exclusively with the gamma common cytokine receptor chain, which is present in the IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 cytokine receptor complexes.
  • JA 3 is predominantly expressed in immune cells and transduces a signal in response to its activation via tyrosine phosphorylation by interleukin receptors. Since JAK3 expression is restricted mostly to hematopoietic cells, its role in cytokine signaling is thought to be more restricted than other JA s. Mutations of JAK3 result in severe combined immunodeficiency (SCID). (O'Shea et al, 2002, Cell, 109 (suppl): S121-S131).
  • SCID severe combined immunodeficiency
  • JA 3 and JAK3 -mediated pathways have been targeted for immunosuppressive indications (e.g., transplantation rejection and rheumatoid arthritis) (Baslund et al., 2005, Arthritis & Rheumatism 52:2686-2692; Changelian et al, 2003, Science 302: 875-878).
  • immunosuppressive indications e.g., transplantation rejection and rheumatoid arthritis
  • TYK2 is implicated in IFN-a, IL-6, IL-10 and IL-12 signaling.
  • Biochemical studies and gene-targeted mice uncovered the crucial role of TYK2 in immunity.
  • Tyk2-deficient mice are viable and fertile but display multiple immunological defects, most prominently high sensitivity to infections and defective tumor surveillance.
  • inhibition of TYK2 results in increased resistance against allergic, autoimmune and inflammatory diseases.
  • targeting Tyk2 appears to be a promising strategy for the treatment of IL-12-, IL-23- or Type 1 IFN- mediated diseases.
  • JAK/STAT signaling pathway is involved in a variety of hyperproliferative and cancer-related processes including cell-cycle progression, apoptosis, angiogenesis, invasion, metastasis and evasion of the immune system (Haura et al., Nature Clinical Practice Oncology, 2005, 2(6), 315-324; Verna et al., Cancer and Metastasis Reviews, 2003, 22, 423-434).
  • the JAK/STAT signaling pathway is important in the genesis and differentiation of hematopoietic cells and regulating both pro- and anti-inflammatory and immune responses (O' Sullivan et al., Molecular Immunology 2007, 44:2497).
  • the JAK STAT pathway and in particular all four members of the JAK family, are believed to play a role in the pathogenesis of the asthmatic response, chronic obstructive pulmonary disease, bronchitis, and other related inflammatory diseases of the lower respiratory tract.
  • the JAK/STAT pathway has also been implicated to play a role in inflammatory diseases/conditions of the eye including, but not limited to, ulceris, uveitis, scleritis, conjunctivitis, as well as chronic allergic responses. Since cytokines utilize different patterns of JAK kinases (O'Sullivan et al, Mol.
  • Rheumatoid arthritis is an autoimmune disease characterized by chronic joint inflammation. Patients with rheumatoid arthritis treated with JAK inhibitor showed that inhibition of JAK1 and JAK3 blocks signalling by multiple cytokines that are important for lymphocyte function, including interleukin-2 (IL-2), IL-4, IL-7, IL-9, IL-15 and IL-21.
  • IL-2 interleukin-2
  • IL-4 interleukin-4
  • IL-7 IL-9
  • IL-15 IL-15
  • IL-21 interleukin-21
  • JAK2 has been shown to be the most important upstream activator mediating STAT3 activation in human tumor cell lines derived from various solid tumors [Mohamad Bassam Sonbol, Belal Firwana, Ahmad Zarzour, Mohammad Morad, Vishal Rana and Ramon V. Tiu "Comprehensive review of JAK inhibitors in myeloproliferative neoplasms.” Therapeutic Advances in Hematology 2013, 4(1), 15-35; Hedvat M, Huszar D, Herrmann A, Gozgit J M, Schroeder A, Sheehy A, et al.
  • JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors. Cancer Cell 2009; 16(6):487-97.]. Therefore, inhibition of JAK kinases may have a beneficial role in the therapeutic treatment of these diseases.
  • JAK inhibitors have gathered attention as a new drug category for both immunosuppresion and antiinflammatory drug, and for cancer drug.
  • new or improved agents which inhibit kinases such as Janus kinases are continually needed that act as immunosuppressive agents for organ transplants, as well as agents for the prevention and treatment of autoimmune diseases (e.g., multiple sclerosis, psoriasis, rheumatoid arthritis, asthma, type I diabetes, inflammatory bowel disease, Crohn's disease, autoimmune thyroid disorders, Alzheimer's disease), diseases involving a hyperactive inflammatory response (e.g., eczema), allergies, chronic obstructive pulmonary disease, bronchitis, cancer (e.g., prostate, leukemia, multiple myeloma), and some immune reactions (e.g., skin rash or contact dermatitis or diarrhea) caused by other therapeutics, to name a few.
  • autoimmune diseases e.g., multiple sclerosis, psoria
  • the invention provides compounds that inhibit, regulate, and/or modulate one or more JAK kinase activities, and are useful for treating proliferative diseases, autoimmune diseases, allergic diseases, inflammatory diseases, transplantation rejections, and their co-morbidities.
  • This invention also provides methods of making the compound, methods of using such compounds in the treatment of said diseases in mammals, especially in humans, and pharmaceutical compositions containing these compounds.
  • Z is C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3, 4 or 5 R 1 groups;
  • Z 1 is H, C1-C12 alkyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z 1 is optionally substituted with 1, 2, 3, 4 or 5 R 2 groups;
  • each R 4 and R 5 is independently H, F, CI, Br, I, N 3 , CN, OH, NH 2 , C1-C12 alkyl, C1-C12 alkoxy, Ci-Ci2 alkylamino, C 2 -Ci 2 alkenyl, C 2 -Ci 2 alkynyl, C 3 -Ci 2 cycloalkyl, 3-12 membered heterocyclyl, C6-Ci 2 aryl or 5-12 membered heteroaryl, or R 4 and R 5 taken together with the carbon atom to which they are attached form a C 3 -Ci 2 cycloalkyl or 3-12 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3, 4 or 5 R 6 groups; each
  • Z is C3-C6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3 or 4 R 1 groups.
  • Z 1 is H, C1-C4 alkyl, C3-C6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z 1 is optionally substituted with 1, 2, 3 or 4 R 2 groups.
  • each R 4 and R 5 is independently H, F, CI, Br, I, N 3 , CN, OH, NH 2 , Ci-C 6 alkyl, Ci-C 6 alkoxy, Ci-C 6 alkylamino, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-7 membered heterocyclyl, phenyl or 5-6 membered heteroaryl, or R 4 and R 5 taken together with the carbon atom to which they are attached form a C3-C6 cycloalkyl or 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 R 6 groups.
  • each R 6 is independently F, CI, CN, N 3 , Ci-C 6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl,
  • SUBSTITUTE SHEET RULE 26 NH 2 , -NH(Ci-C 6 alkyl), -NH(CH 2 )n-(C3-C 6 cycloalkyl), -NH(CH 2 ) n -(3-7 membered heterocyclyl), -NH(CH 2 ) n -phenyl, -NH(CH 2 ) n -(5-6 membered heteroaryl), -N(Ci-C 6 alkyl) 2 , -N[(CH 2 )n-(C3-C 6 cycloalkyl)] 2 , -N[(CH 2 ) n -(3-7 membered heterocyclyl)] 2 , -N[(CH 2 )n-phenyl] 2 , -N[(CH 2 ) n -(5-6 membered heteroaryl)] 2 , OH, -0(Ci-C 6 alkyl), -0(CH 2 )
  • each R a , R b and R c is independently H, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C6 cycloalkyl, -(C1-C2 alkylene)-(C3-C6 cycloalkyl), 3-7 membered heterocyclyl, -(C1-C2 alkylene)-(3-7 membered heterocyclyl), phenyl, -(C1-C2 alkylene)-phenyl, 5- 6 membered heteroaryl or -(C1-C2 alkylene)-(5-6 membered heteroaryl), or R a and R b taken together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2 or 3 substitutents independently selected from F, CI, CN, N 3 , OH,
  • Z is C4-C6 cycloalkyl, or 4-, 5- or 6-membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3 or 4 R 1 groups.
  • Z is :
  • Z 1 is H, methyl, ethyl, propyl, isopropyl or cyclopropyl.
  • each R a , R b and R c is independently H, methyl, ethyl, propyl, isopropyl, cyclopropyl or butyl, wherein each R a , R b and R c is optionally independently substituted with 1 , 2 or 3 substitutents independently selected from F, CI, CN, N 3 , OH, NH 2 , methyl, ethyl, -CF 3 , - OCH 3 and -CH 3 NH 2 .
  • composition comprising the compound disclosed herein, and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof.
  • the pharmaceutical composition disclosed herein further comprising a therapeutic agent selected from the group consisting of chemotherapeutic agents, anti-proliferative agents, phosphodiesterase 4 (PDE4) inhibitors, p 2 -adrenoreceptor agonists, corticosteroids, nonsteroidal GR agonists, anticholinergic agents, antihistamines and combinations thereof.
  • a therapeutic agent selected from the group consisting of chemotherapeutic agents, anti-proliferative agents, phosphodiesterase 4 (PDE4) inhibitors, p 2 -adrenoreceptor agonists, corticosteroids, nonsteroidal GR agonists, anticholinergic agents, antihistamines and combinations thereof.
  • provided herein is a method of preventing, treating or lessening the severity of a JAK-mediated disease in a patient by administering to the patient with the compound disclosed herein or the pharmaceutical composition disclosed herein.
  • the JAK-mediated disease is a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease or a transplantation rejection.
  • the JAK-mediated disease is cancer, polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), chronic obstruction pulmonary disease (COPD), asthma, systemic lupus erythematosis, cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis, type I diabetes mellitus, allergic airway disease, sinusitis, eczema, hives, food allergies, allergies to insect venom, inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection or cell transplant rejection.
  • CML chronic myelogenous leukemia
  • COPD chronic obstruction pulmonary disease
  • asthma systemic lupus erythematosis, cutaneous lupus erythematos
  • SUBSTITUTE SHEET RULE 26 [040] In another aspect, provided herein is the compound or the pharmaceutical composition disclosed herein for use in preventing, treating or lessening the severity of a JAK-mediated disease in a patient.
  • a medicament for treating cancer polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), chronic obstruction pulmonary disease (COPD), asthma, systemic and cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis, type I diabetes mellitus, allergic airway disease, sinusitis, eczema, hives, food allergies, allergies to insect venom, inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection or cell transplant rejection.
  • provided herein is a method of modulating the activity of the Janus kinase with the compound or the pharmaceutical composition disclosed herein.
  • any embodiment disclosed herein can be combined with other embodiments as long as they are not contradictory to one another, even though the embodiments are described under different aspects of the invention.
  • any technical feature in one embodiment can be applied to the corresponding technical feature in other embodiment as long as they are not contradictory to one another, even though the embodiments are described under different aspects of the invention.
  • the term "subject" refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human.
  • primates e.g., humans, male or female
  • the subject is a primate.
  • the subject is a human.
  • patient refers to a human (including adults and children) or other animal. In one embodiment, “patient” refers to a human.
  • Stereoisomers refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. Stereoisomers include enantiomer, diastereomers, conformer (rotamer), geometric (cis/trans) isomer, atropisomer, etc.
  • Chiral refers to molecules which have the property of non-superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • Enantiomers refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • Diastereomer refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties or biological activities. Mixture of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography such as HPLC.
  • any asymmetric atom (e.g., carbon or the like) of the compound(s) disclosed herein can be present in racemic or enantiomerically enriched, for example the (R)-, (S)- or (R,S)- configuration.
  • each asymmetric atom has at least 50 % enantiomeric excess, at least 60
  • the compounds can be present in the form of one of the possible stereoisomers or as mixtures thereof, such as racemates and diastereoisomer mixtures, depending on the number of asymmetric carbon atoms.
  • Optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituent may be E or Z configuration. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis- or trans-configuration.
  • Any resulting mixtures of stereoisomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric isomers, enantiomers, diastereomers, for example, by chromatography and/or fractional crystallization.
  • racemates of final products or intermediates can be resolved into the optical antipodes by methods known to those skilled in the art, e.g., by separation of the diastereomeric salts thereof.
  • Racemic products can also be resolved by chiral chromatography, e.g., high performance liquid chromatography (HPLC) using a chiral adsorbent.
  • HPLC high performance liquid chromatography
  • Preferred enantiomers can also be prepared by asymmetric syntheses. See, for example, Jacques, et al, Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Principles of Asymmetric Synthesis (2 nd Ed.
  • tautomer or "tautomeric form” refers to structural isomers of different energies which are interconvertible via a low energy barrier. Where tautomerization is possible (e.g. in solution), a chemical equilibrium of tautomers can be reached.
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganization of some of the bonding electrons.
  • keto-enol tautomerization is the interconversion of pentane-2,4-dione and 4-hydroxypent-3-en-2-one tautomers.
  • tautomerization is phenol-keto tautomerization.
  • phenol-keto tautomerization is the interconversion of pyridin-4-ol and pyridin-4(lH)-one tautomers.
  • compounds disclosed herein may optionally be substituted with one or more substituents, such as those illustrated below, or as exemplified by particular classes, subclasses, and species of the invention.
  • substituents such as those illustrated below, or as exemplified by particular classes, subclasses, and species of the invention.
  • the phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted”.
  • substituted refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent.
  • an optionally substituted group may have a substituent at each substitutable position of the group. When more than one position in a given structure can be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position.
  • substituents of compounds disclosed herein are disclosed in groups or in ranges. It is specifically intended that the invention include each and every individual subcombination of the members of such groups and ranges.
  • C1-C6 alkyl is specifically intended to individually disclose methyl, ethyl, C 3 alkyl, C 4 alkyl, C 5 alkyl, and C 6 alkyl
  • 4-7 membered heterocyclyl is specifically intended to individually disclose 4- membered heterocyclyl, 5- membered heterocyclyl, 6- membered heterocyclyl and 7- membered heterocyclyl.
  • linking substituents are described. Where the structure clearly requires a linking group, the Markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the Markush group definition for that variable lists “alkyl” or “aryl” then it is understood that the “alkyl” or “aryl” represents a linking alkylene group or arylene group, respectively.
  • alkyl refers to a saturated linear or branched-chain monovalent hydrocarbon radical of 1 to 20 carbon atoms, wherein the alkyl radical may be optionally substituted independently with one or more substituents described below. Unless otherwise specified, the alkyl group contains 1-20 carbon atoms. In one embodiment, the alkyl group contains 1-12 carbon atoms. In another embodiment, the alkyl group contains 1-6 carbon atoms. In still another embodiment, the alkyl group contains 1-4 carbon atoms. In yet another embodiment, the alkyl group contains 1-3 carbon atoms. The alkyl radical may be optionally substituted independently with one or more substituents described herein.
  • alkyl group examples include methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), 1 -propyl (n-Pr, n- propyl, -CH 2 CH 2 CH 3 ), 2-propyl (z ' -Pr, /-propyl, -CH(CH 3 ) 2 ), 1 -butyl (zi-Bu, n-butyl, - CH 2 CH 2 CH 2 CH 3 ), 2-methyl-l-propyl (/-Bu, /-butyl, -CH 2 CH(CH 3 ) 2 ), 2-butyl (s-Bu, s-butyl, -
  • alkylene refers to a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms. Unless otherwise specified, the alkylene group contains 1-12 carbon atoms. In one embodiment, the alkylene group contains 1-6 carbon atoms. In another embodiment, the alkylene group contains 1-4 carbon atoms. In still another embodiment, the alkylene group contains 1-3 carbon atoms. In yet another embodiment, the alkylene group contains 1-2 carbon atoms.
  • the alkylene group is exemplified by methylene (-CH 2 -), ethylidene (-CH 2 CH 2 -), isopropylidene (-CH(CH 3 )CH 2 -), and the like.
  • alkenyl refers to a linear or branched-chain monovalent hydrocarbon radical of 2 to 12 carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp 2 double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having "cz ' s" and “trans” orientations, or alternatively, "E” and "Z” orientations.
  • the alkenyl group contains 2-8 carbon atoms.
  • the alkenyl group contains 2-6 carbon atoms.
  • the alkenyl group contains 2-4 carbon atoms.
  • alkynyl refers to a linear or branched-chain monovalent hydrocarbon radical of 2 to 12 carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein.
  • the alkynyl group contains 2-8 carbon atoms.
  • the alkynyl group contains 2-6 carbon atoms.
  • the alkynyl group contains 2-4 carbon atoms.
  • Some non-limiting examples of the alkynyl group include ethynyl (-C ⁇ CH), propynyl (propargyl, -CH 2 C ⁇ CH), -C ⁇ C-CH 3 , and the like.
  • alkoxy refers to an alkyl group, as previously defined, attached to the principal carbon atom through an oxygen atom. Unless otherwise specified, the alkoxy group contains 1-12
  • the alkoxy group contains 1-6 carbon atoms. In another embodiment, the alkoxy group contains 1-4 carbon atoms. In still another embodiment, the alkoxy group contains 1-3 carbon atoms.
  • the alkoxy radical may be optionally substituted independently with one or more substituents described herein.
  • alkoxy groups include methoxy (MeO, -OCH3), ethoxy (EtO, -OCH2CH3), 1-propoxy (n-PrO, n-propoxy, - OCH2CH2CH3), 2-propoxy (z ' -PrO, z ' -propoxy, -OCH(CH 3 )2), 1-butoxy (n-BuO, n-butoxy, - OCH2CH 2 CH 2 CH 3 ), 2-methyl-l-propoxy (z-BuO, z-butoxy, -OCH 2 CH(CH 3 ) 2 ), 2-butoxy (s-BuO, s- butoxy, -OCH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC(CH 3 ) 3 ), 1-pentoxy (n- pentoxy, -OCH2CH 2 CH2CH 2 CH 3 ), 2-pentoxy (-OCH2CH3 ),
  • haloalkyl refers to alkyl, alkenyl, or alkoxy, as the case may be, substituted with one or more halogen atoms.
  • haloalkyl and haloalkoxy are include trifluoromethyl (-CF 3 ), trifluoromethoxy (-OCF 3 ) and the like.
  • carbocycle refers to a monovalent or multivalent non-aromatic, saturated or partially unsaturated ring having 3 to 12 carbon atoms as a monocyclic, bicyclic or tricyclic ring system.
  • the carbobicyclyl system includes spiro carbobicyclyl and fused carbobicyclyl.
  • carbocyclyl groups include cycloalkyl, cycloalkenyl, and cycloalkynyl.
  • carbocyclyl group examples include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-l-enyl, l-cyclopent-2-enyl, 1-cyclopent- 3-enyl, cyclohexyl, 1-cyclohex-l-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, cyclohexadienyl, and the like.
  • cycloalkyl refers to a monovalent or multivalent saturated ring having 3 to 12 carbon atoms as a monocyclic, bicyclic, or tricyclic ring system. In one embodiment, the cycloalkyl contains 3-12 carbon atoms. In another embodiment, the cycloalkyl contains 3-8 carbon atoms. In still another embodiment, the cycloalkyl contains 4-6 carbon atoms. In yet another embodiment, the cycloalkyl contains 3-6 carbon atoms.
  • the cycloalkyl radical may be optionally substituted independently with one or more substituents described herein.
  • heterocycle refers to a amonovalent or multivalent, saturated or partially unsaturated, non-aromatic monocyclic, bicyclic or tricyclic ring containing 3-12 ring atoms of which at least one ring atom is selected from nitrogen, sulfur and oxygen, and which may, unless otherwise specified, be carbon
  • Ring sulfur atoms may be optionally oxidized to form S-oxides.
  • Ring nitrogen atoms maybe optionally oxidized to form N-oxides.
  • the heterocyclyl contains saturated heterocyclyl and partially unsaturated heterocyclyl. The heterocyclyl has one or more points of attachment to the rest of the molecule.
  • heterocyclyl examples include oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1,3- dioxolanyl, dithiolanyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, thioxanyl, dithianyl, homopiperazinyl, homopiperidinyl,
  • heterocyclyl wherein the ring sulfur atom is oxidized are sulfolanyl, 1,1-dioxo-thiomorpholinyl, and the like.
  • the heterocyclyl group may be optionally substituted with one or more substituents described herein.
  • Ring sulfur atoms may be optionally oxidized to form S-oxides.
  • Ring nitrogen atoms maybe optionally oxidized to form N-oxides.
  • the 4-7 membered heterocyclyl contains 4-7 membered saturated heterocyclyl and partially unsaturated heterocyclyl.
  • the 4-7 membered heterocyclyl has one or more points of attachment to the rest of the molecule.
  • Some non-limiting examples of 4-7 membered heterocyclyl include azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 1-pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, dihydropyrazolyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1,3-dioxolanyl, dithiolanyl, isoxazolidinyl, isothiazolidinyl, 1 ,2- oxazinanyl,
  • SUBSTITUTE SHEET RULE 26 morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, thioxanyl, dithianyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl (1,4-oxazepinyl, 1 ,2-oxazepinyl), diazepinyl (1,4-diazepinyl, 1 ,2-diazepinyl), thiazepinyl (1,4-thiazepinyl, 1 ,2-thiazepinyl), dioxpinyl (1,4- dioxpinyl, 1,2-dioxpinyl), and the like.
  • heterocyclyl wherein the ring sulfur atom is oxidized are sulfolanyl, 1,1-dioxotetrahydrothiophenyl, l,l-dioxotetrahydro-2H- thiopyranyl, 1,1-dioxothiomorpholinyl, and the like.
  • the 4-7 membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
  • Ring sulfur atoms may be optionally oxidized to form S-oxides.
  • Ring nitrogen atoms maybe optionally oxidized to form N-oxides.
  • 4- membered heterocyclyl contains 4-membered saturated heterocyclyl and partially unsaturated heterocyclyl. Some non-limiting examples of 4-membered heterocyclyl include azetidinyl, oxetanyl, thietanyl, and the like. The 4-membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
  • Ring sulfur atoms may be optionally oxidized to form S-oxides.
  • Ring nitrogen atoms maybe optionally oxidized to form N-oxides.
  • 5- membered heterocyclyl contains 5-membered saturated heterocyclyl and partially unsaturated heterocyclyl.
  • 5-membered heterocyclyl include pyrrolidinyl, 1- pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, dihydropyrazolyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1,3- dioxolanyl, dithiolanyl, isoxazolidinyl, isothiazolidinyl, and the like.
  • a non-limiting examples of heterocyclyl wherein the ring sulfur atom is oxidized is 1,1-dioxotetrahydrothiophenyl.
  • the 5-membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
  • Ring sulfur atoms may be optionally oxidized to form S-oxides.
  • Ring nitrogen atoms maybe optionally oxidized to form N-oxides.
  • the 6-membered heterocyclyl contains 6-membered saturated heterocyclyl and partially unsaturated heterocyclyl.
  • 6-membered heterocyclyl include tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, dihydropyridyl, tetrahydropyridyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, thioxanyl, dithianyl, and the like.
  • heterocyclyl wherein the ring sulfur atom is oxidized are 1,1-dioxothiomorpholinyl, l,l-dioxotetrahydro-2H-thiopyranyl, and the like.
  • the 6-membered heterocyclyl group is optionally substituted with one or more substituents described herein.
  • Ring sulfur atoms may be optionally oxidized to form S-oxides.
  • Ring nitrogen atoms maybe optionally oxidized to form N-oxides.
  • the 3-7 membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
  • Ring sulfur atoms may be optionally oxidized to form S-oxides.
  • Ring nitrogen atoms maybe optionally oxidized to form N-oxides.
  • 7-12 membered heterocyclyl include indolinyl, 1,2,3,4-tetrahydroisoquinolinyl, 1,3-benzodioxolyl, 2-oxa-5-azabicyclo[2.2.1]hept-5-yl, and the like.
  • the 7-12 membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
  • fused bicyclic ring refers to a monovalent or multivalent saturated or partially unsaturated bridged ring system, which refers to a bicyclic ring system that is not aromatic. Such a system may contain isolated or conjugated unsaturation, but not aromatic or heteroaromatic rings in its core structure (but may have aromatic substitution thereon).
  • spirocyclyl spirocyclic
  • spiro bicyclyl spiro bicyclic
  • spiro bicyclic a monovalent or multivalent, saturated or partially unsaturated ring system wherein a ring originating from a particular annular carbon of another ring.
  • a saturated bridged ring system ring B and B'
  • ring A and ring B share an atom between the two saturated ring system, which terms as a "spirocyclyl” or "spiro bicyclyl”.
  • Each ring in the fused bicyclyl or the spiro bicyclyl can be either a carbocyclyl or a heterocyclyl, and each ring is optionally substituted independently with one or more substituents described herein.
  • heterocycloalkyl refers to a monovalent or multivalent saturated ring having 3 to 12 ring atoms as a monocyclic, bicyclic, or tricyclic ring system in which at least one ring atom is selected from nitrogen, sulfur and oxygen.
  • n membered where n is an integer typically describes the number of ring- forming atoms in a moiety where the number of ring-forming atoms is n.
  • piperidinyl is an example of a 6 membered heterocycloalkyl
  • 1,2,3,4-tetrahydro-naphthalenyl is an example of a 10 membered carbocyclyl group.
  • heteroatom refers to one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, including any oxidized form of nitrogen, sulfur, or phosphorus; the quaternized form of any basic nitrogen; or a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4- dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR (as in N-substituted pyrrolidinyl).
  • halogen refers to Fluoro (F), Chloro (CI), Bromo (Br), or Iodo (I).
  • azido or "N 3" refers to an azide moiety. This radical may be attached, for
  • SUBSTITUTE SHEET RULE 26 example, to a methyl group to form azidomethane (methyl azide, MeN 3 ); or attached to a phenyl group to form phenyl azide (PhN 3 ).
  • aryl refers to monocyclic, bicyclic, and tricyclic carbocyclic ring systems having a total of 6 to 14 ring members, preferably, 6 to 12 ring members, and more preferably 6 to 10 ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3 to 7 ring members and that has one or more points of attachment to the rest of the molecule.
  • aryl may be used interchangeably with the term “aryl ring” or "aromatic ring”. Some non-limiting examples of the aryl group would include phenyl, naphthyl, and anthracenyl.
  • the aryl radical is optionally substituted independently with one or more substituents described herein.
  • heteroaryl or “heteroaromatic ring” refers to monocyclic, bicyclic, and tricyclic ring systems having a total of 5 to 12 ring members, preferably, 5 to 10 ring members, and more preferably 5 to 6 ring members, wherein at least one ring in the system is aromatic, at least one aromatic ring in the system contains one or more heteroatoms, wherein each ring in the system contains 5 to 7 ring members and that has one or more points of attachment to the rest of the molecule.
  • heteroaryl may be used interchangeably with the term “heteroaryl ring” or the term “heteroaromatic ring”.
  • heteroaryl refers to a 5-12 membered heteroaryl comprises 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In another embodiment, heteroaryl refers to a 5-10 membered heteroaryl comprises 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In another embodiment, heteroaryl refers to a 5-6 membered heteroaryl comprises 1, 2, 3 or 4 heteroatoms independently selected from O, S and N.
  • the heteroaryl radicals are optionally substituted independently with one or more substituents described herein.
  • 5-6 membered heteroaryl group examples include 2-furanyl, 3- furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5- isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3- pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyrimidonyl, pyrimidinedionyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5- tetrazolyl), triazolyl (e.g.,
  • bicycles heteroaryl group examples include benzimidazolyl, benzofuryl, benzothiophenyl, indolyl (e.g., 2-indolyl), purinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4- quinolinyl), and isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl or 4-isoquinolinyl),
  • azolyl refers to a 5- or a 9-membered heteroaryl ring system containing at least one nitrogen atom.
  • 5-membered azolyl rings include pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, diazolyl, triazolyl, and the like.
  • 9-membered azolyl rings include indazolyl, pyrazolopyridinyl, lH-benzo[d]imidazolyl, and the like.
  • Carboxy or “carboxyl”, whether used alone or with other terms, such as “carboxyalkyl”, refers to -CO2H.
  • alkylamino embraces “N-alkylamino” and "N,N-dialkylamino” where amino groups are independently substituted with one alkyl radical or with two alkyl radicals, respectively.
  • alkylamino are "lower alkylamino" radicals having one or two alkyl radicals of one to six carbon atoms, attached to a nitrogen atom.
  • alkylamino are alkylamino radicals having one or two alkyl radicals of one to four carbon atoms, attached to a nitrogen atom.
  • alkylamino include N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino, and the like.
  • aminoalkyl refers to linear or branched alkyl radicals having one to about ten carbon atoms any one of which may be substituted with one or more amino radicals. More preferred aminoalkyl radicals are "lower aminoalkyl” radicals having 1-6 carbon atoms and one or more amino radicals. Examples of such radicals include aminomethyl, aminoethyl, aminopropyl, aminobutyl and aminohexyl.
  • a bond drawn from a substituent to the center of one ring within a ring system represents substitution of the substituent at any substitutable position on the ring system.
  • Structure b represents possible substitution in any of the positions on the ring C and ring D shown in Structure c ⁇ Structure g.
  • SUBSTITUTE SHEET RULE 26 system (as shown in Structure h) represents connection of the connecting bond attached to the rest of the molecule at any substitutable position on the ring system.
  • Structure h represents possible connection attached to the rest of the molecule in any of the position on ring E.
  • protecting group refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound.
  • an "amino-protecting group” is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxy-carbonyl (BOC, Boc), benzyloxycarbonyl (CBZ, Cbz) and 9- fluorenylmethylenoxy-carbonyl (Fmoc).
  • a "hydroxy-protecting group” refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality.
  • Suitable protecting groups include acetyl and silyl.
  • a "carboxy-protecting group” refers to a substituent of the carboxy group that blocks or protects the carboxy functionality.
  • Common carboxy-protecting groups include -CH2CH2SO2PI1, cyanoethyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxy- methyl, 2-(p-toluenesulfonyl)-ethyl, 2-(/?-nitrophenylsulfenyl)-ethyl, 2-(diphenylphosphino)-ethyl, nitroethyl and the like.
  • protecting groups and their use see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991; and P. J. Kocienski, Protecting Groups, Thieme, Stuttgart, 2005.
  • prodrug represents a compound that is transformed in vivo into a compound of Formula (I). Such a transformation can be affected, for example, by hydrolysis in blood or enzymatic transformation of the prodrug form to the parent form in blood or tissue.
  • Prodrugs of the compounds disclosed herein may be, for example, esters. Esters that may be utilized as prodrugs in the present invention are phenyl esters, aliphatic (C1-C24) esters, acyloxymethyl esters, carbonates, carbamates, and amino acid esters. For example, a compound disclosed herein that contains an OH group may be acylated at this position in its prodrug form.
  • prodrug forms include phosphates, such as, for example those phosphates resulting from the phosphonation of an OH group on the parent compound.
  • phosphates such as, for example those phosphates resulting from the phosphonation of an OH group on the parent compound.
  • a "metabolite” refers to a product produced through metabolism in the body of a specified compound or salt thereof.
  • the metabolites of a compound may be identified using routine techniques known in the art and their activities determined using tests such as those described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound.
  • the invention includes metabolites of compounds disclosed herein, including compounds produced by a process comprising contacting a compound disclosed herein with a mammal for a period of time sufficient to yield a metabolic product thereof.
  • a "pharmaceutically acceptable salt” refers to organic or inorganic salts of a compound disclosed herein.
  • the pharmaceutically acceptable salts are well known in the art. For example, Berge et al, describe pharmaceutically acceptable salts in detail in J. Pharm. Sci., 1977, 66, 1-19, which is incorporated herein by reference.
  • Some non-limiting examples of the pharmaceutically acceptable salt include salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid.
  • compositions of the pharmaceutically acceptable salt include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate,
  • compositions derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (Ci-C 4 alkyl) 4 salts.
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • compositions include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, Ci-C 8 sulfonate and aryl sulfonate.
  • counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, Ci-C 8 sulfonate and aryl sulfonate.
  • a "solvate” refers to an association or complex of one or more solvent molecules and a compound disclosed herein.
  • solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine.
  • hydrate refers to the complex where the solvent molecule is water.
  • the term “treat”, “treating” or “treatment” of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof).
  • “treat”, “treating” or “treatment” refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient.
  • “treat”, “treating” or “treatment” refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
  • “treat”, “treating” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder.
  • Inflammatory disorder/disease can refer to any disease, disorder, or syndrome in which an excessive or unregulated inflammatory response leads to excessive inflammatory symptoms, host tissue damage, or loss of tissue function.
  • “Inflammatory disorder/disease” also refers to a pathological state mediated by influx of leukocytes and/or neutrophil chemotaxis.
  • Inflammation refers to a localized, protective response elicited by injury or destruction of tissues, which serves to destroy, dilute, or wall off (sequester) both the injurious agent and the injured tissue. Inflammation is notably associated with influx of leukocytes and/or neutrophil chemotaxis. Inflammation can result from infection with pathogenic organisms and viruses and from noninfectious means such as trauma or reperfusion following myocardial infarction or stroke, immune response to foreign antigen, and autoimmune responses. Accordingly, inflammatory disorders amenable to treatment with the compounds disclosed herein encompass disorders associated with reactions of the specific defense system as well as with reactions of the nonspecific defense system.
  • Specific defense system refers to the component of the immune system that reacts to the presence of specific antigens.
  • inflammation resulting from a response of the specific defense system include the classical response to foreign antigens, autoimmune diseases, and delayed type hypersensitivity response mediated by T-cells.
  • Chronic inflammatory diseases, the rejection of solid transplanted tissue and organs, e.g., kidney and bone marrow transplants, and graft versus host disease (GVHD), are further examples of inflammatory reactions of the specific defense system.
  • Allergic disease refers to any symptoms, tissue damage, or loss of tissue function resulting from allergy.
  • Arthritic disease refers to any disease that is characterized by inflammatory lesions of the joints attributable to a variety of etiologies.
  • Dermatis refers to any of a large family of diseases of the skin that are characterized by inflammation of the skin attributable to a variety of etiologies.
  • Transplant rejection refers to any immune reaction directed against grafted tissue, such as organs or cells (e.g., bone marrow), characterized by a loss of function of the grafted and surrounding tissues, pain, swelling, leukocytosis, and thrombocytopenia.
  • the therapeutic methods of the present invention include methods for the treatment of disorders associated with inflammatory cell activation.
  • cancer and “cancerous” refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth.
  • a “tumor” comprises one or more cancerous cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies.
  • squamous cell cancer e.g., epithelial squamous cell cancer
  • lung cancer including small-cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
  • NSCLC non-small cell lung cancer
  • adenocarcinoma of the lung and squamous carcinoma of the lung cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer
  • novel compounds which are inhibitors of kinase activity are disclosed.
  • Compounds which are JAK kinase inhibitors may be useful in the treatment of diseases associated with inappropriate kinase activity, in particular inappropriate JAK kinase activity, for example in the treatment and prevention of diseases mediated by JAK kinase involved signalling pathways.
  • diseases include proliferative disease, autoimmune disease, allergic disease, inflammatory disease, transplantation rejection, and their comorbidities.
  • a compound of the present invention may be useful in the treatment of diseases such as cancer, polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), chronic obstruction pulmonary disease (COPD), asthma, systemic
  • the compounds disclosed herein may show selectivity for Janus kinases over other kinases.
  • Z is C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3, 4 or 5 R 1 groups;
  • Z 1 is H, C1-C12 alkyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z 1 is optionally substituted with 1, 2, 3, 4 or 5 R 2 groups;
  • each m is independently 0, 1 or 2;
  • each n is independently 0, 1, 2, 3 or 4.
  • Z is C 3 -C 6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3 or 4 R 1 groups.
  • Z 1 is H, C1-C4 alkyl, C 3 -C 6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z 1 is optionally substituted with 1, 2, 3 or 4 R 2 groups.
  • each R 4 and R 5 is independently H, F, CI, Br, I, N 3 , CN, OH, - NH 2 , Ci-C 6 alkyl, Ci-C 6 alkoxy, Ci-C 6 alkylamino, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-7 membered heterocyclyl, phenyl or 5-6 membered heteroaryl, or R 4 and R 5 taken together with the carbon atom to which they are attached form a C3-C6 cycloalkyl or 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 R 6 groups.
  • each R 6 is independently F, CI, CN, N3, Ci-C 6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, - NH 2 , -NH(Ci-C 6 alkyl), -NH(CH 2 )n-(C3-C 6 cycloalkyl), -NH(CH 2 ) n -(3-7 membered heterocyclyl), -NH(CH 2 ) n -phenyl, -NH(CH 2 ) n -(5-6 membered heteroaryl), -N(Ci-C 6 alkyl) 2 , -N[(CH 2 )n-(C3-C 6 cycloalkyl)] 2 , -N[(CH 2 ) n -(3-7 membered heterocycl
  • each R a , R b and R c is independently H, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C6 cycloalkyl, -(C1-C2 alkylene)-(C3-C6 cycloalkyl), 3-7 membered heterocyclyl, -(C1-C2 alkylene)-(3-7 membered heterocyclyl), phenyl, -(C1-C2 alkylene)-phenyl, 5- 6 membered heteroaryl or -(C1-C2 alkylene)-(5-6 membered heteroaryl), or R a and R b are taken together with the nitrogen atom to which they attached form a 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2 or 3 substitutents independently selected from F, CI, CN, N 3 , OH,
  • Z is C4-C6 cycloalkyl, or 4-, 5- or 6-membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3 or 4 R 1 groups.
  • Z is :
  • Z 1 is H, methyl, ethyl, propyl, isopropyl or cyclopropyl.
  • each R a , R b and R c is independently H, methyl, ethyl, propyl, isopropyl, cyclopropyl or butyl, wherein each R a , R b and R c is optionally independently substituted with 1, 2 or 3 substitutents independently selected from F, CI, CN, N 3 , OH, NH 2 , methyl, ethyl, - CF 3 , -OCH 3 and -CH 3 NH 2 .
  • the compounds disclosed herein may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of compounds of Formula (I), including but not limited to, diastereomers, enantiomers, atropisomers, conformers (rotamers) and geometric (cis/trans) isomers as well as mixtures thereof such as racemic mixtures, form part of the present invention.
  • the compounds of Formula (I) can be in the form of salts.
  • the salts are pharmaceutically acceptable salts.
  • pharmaceutically acceptable indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith.
  • the salts are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds of Formula (I) and/or for separating enantiomers of compounds of Formula (I).
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/di
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.
  • compositions can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table.
  • the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like.
  • Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two. Generally, use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable.
  • the appropriate base such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like
  • the compounds disclosed herein, including their salts can also be obtained in the form of their hydrates, or include other solvents such as ethanol, DMSO, and the like, used for their crystallization.
  • the compounds of the present invention may inherently or by design form solvates with pharmaceutically acceptable solvents (including water); therefore, it is intended that the invention embrace both solvated and unsolvated forms.
  • SUBSTITUTE SHEET RULE 26 structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2 H (deuterium, D), 3 H, U C, 13 C, 14 C, 15 N, 17 0, 18 0, 18 F, 31 P, 32 P, 35 S, 36 C1, 125 I, respectively.
  • the compounds of the invention include isotopically enriched compounds as defined herein, for example those into which radioactive isotopes, such as 3 H, 14 C and 18 F, or those into which non-radioactive isotopes, such as 2 H and 13 C are present.
  • isotopically enriched compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F- enriched compound may be particularly desirable for PET or SPECT studies.
  • Isotopically-enriched compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying examples and preparations using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • substitution with heavier isotopes may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements or an improvement in therapeutic index.
  • deuterium in this context is regarded as a substituent of a compound of Formula (I).
  • concentration of such a heavier isotope, specifically deuterium may be defined by the isotopic enrichment factor.
  • isotopic enrichment factor as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5%) deuterium incorporation), at least 6000 (90%> deuterium incorporation), at least 6333.3 (95%) deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99%) deuterium incorporation), or at least 6633.3 (99.5%> deuterium incorporation).
  • Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D 2 0, i-acetone, DMSO- i.
  • composition comprising a therapeutically effective amount of the compound disclosed herein, and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof.
  • the composition is a liquid, solid, semi-solid, gel, or an aerosol form.
  • a method of treating a disease or disorder modulated by one or more Janus kinases comprising administering to a mammal in need of such treatment an effective amount of a compound or a pharmaceutical composition disclosed herein.
  • the disease or disorder is selected from proliferative disease, autoimmune disease, allergic disease, inflammatory disease or transplantation rejection.
  • the compound or the pharmaceutical composition disclosed herein for use in the treatment of disease or disorder selected from proliferative disease, autoimmune disease, allergic disease, inflammatory disease or transplantation rejection.
  • the present invention provides a pharmaceutical composition that include a compound of disclosed herein, or a compound listed in Table 1; and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof.
  • the amount of compound in the pharmaceutical composition disclosed herein is such that is effective to detectably inhibit a protein kinase in a biological sample or in a patient.
  • SUBSTITUTE SHEET RULE 26 for treatment or where appropriate, as a pharmaceutically acceptable derivative thereof.
  • pharmaceutically acceptable derivative include pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
  • compositions disclosed herein may be prepared and packaged in bulk form wherein a safe and effective amount of the compound disclosed herein can be extracted and then given to the patient such as with powders or syrups.
  • the pharmaceutical compositions disclosed herein may be prepared and packaged in unit dosage form wherein each physically discrete unit contains the compound disclosed herein.
  • the pharmaceutical compositions of the invention typically may contain, for example, from 0.5 mg to 1 g, or from 1 mg to 700 mg, or from 5 mg to 100 mg of the compound disclosed herein.
  • pharmaceutically acceptable excipient means a pharmaceutically acceptable material, composition or vehicle involved in giving form or consistency to the pharmaceutical composition.
  • Each excipient must be compatible with the other ingredients of the pharmaceutical composition when commingled such that interactions which would substantially reduce the efficacy of the compound disclosed herein when administered to a patient and interactions which would result in pharmaceutical compositions that are not pharmaceutically acceptable are avoided.
  • each excipient must be pharmaceutically-acceptable, e.g., of sufficiently high purity.
  • Suitable pharmaceutically acceptable excipients will vary depending upon the particular dosage form chosen.
  • suitable pharmaceutically acceptable excipients may be chosen for a particular function that they may serve in the composition.
  • certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of uniform dosage forms.
  • Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of stable dosage forms.
  • Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the carrying or transporting of the compound or compounds disclosed herein once administered to the patient from one organ, or portion of the body, to another organ, or portion of the body.
  • Certain pharmaceutically acceptable excipients may be chosen for their ability to enhance patient compliance.
  • Suitable pharmaceutically acceptable excipients comprise the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweetners, flavoring agents, flavor masking agents, coloring agents, anticaking agents, hemectants, chelating agents,
  • SUBSTITUTE SHEET RULE 26 plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants, and buffering agents.
  • certain pharmaceutically acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the formulation and what other excipients are present in the formulation.
  • Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically-acceptable excipients in appropriate amounts for use in the invention.
  • resources that are available to the skilled artisan which describe pharmaceutically acceptable excipients and may be useful in selecting suitable pharmaceutically acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press).
  • compositions disclosed herein are prepared using techniques and methods known to those skilled in the art. Some of the methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company).
  • the invention is directed to process for the preparation of a pharmaceutical composition
  • a pharmaceutical composition comprising the compound disclosed herein and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof, which comprises mixing the ingredients.
  • a pharmaceutical composition comprising the compound disclosed herein may be prepared by, for example, admixture at ambient temperature and atmospheric pressure.
  • dosage forms include those adapted for (1) oral administration such as tablets, capsules, caplets, pills, troches,
  • SUBSTITUTE SHEET RULE 26 powders, syrups, elixers, suspensions, solutions, emulsions, sachets, and cachets; (2) parenteral administration such as sterile solutions, suspensions, and powders for reconstitution; (3) transdermal administration such as transdermal patches; (4) rectal administration such as suppositories; (5) inhalation such as aerosols, solutions, and dry powders; and (6) topical administration such as creams, ointments, lotions, solutions, pastes, sprays, foams, and gels.
  • the compounds disclosed herein will be formulated for oral administration. In another embodiment, the compounds disclosed herein will be formulated for inhaled administration. In a further embodiment, the compounds disclosed herein will be formulated for intranasal administration. In another embodiment, the compounds disclosed herein will be formulated for transdermal administration. In a further embodiment, the compounds disclosed herein will be formulated for topical administration.
  • compositions provided herein can be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets.
  • Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach.
  • Enteric- coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates.
  • Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation.
  • Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material.
  • Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating.
  • Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.
  • the tablet dosage forms can be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled- release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.
  • the pharmaceutical compositions provided herein can be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate.
  • the hard gelatin capsule also known as the dry-filled capsule (DFC)
  • DFC dry-filled capsule
  • the soft elastic capsule (SEC) is a soft,
  • SUBSTITUTE SHEET RULE 26 globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol.
  • the soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid.
  • the liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545.
  • the capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
  • compositions provided herein can be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups.
  • An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in- water or water-in-oil.
  • Emulsions may include a pharmaceutically acceptable nonaqueous liquid or solvent, emulsifying agent, and preservative.
  • Suspensions may include a pharmaceutically acceptable suspending agent and preservative.
  • Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di(lower alkyl) acetal of a lower alkyl aldehyde, e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol.
  • Elixirs are clear, sweetened, and hydroalcoholic solutions.
  • Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative.
  • a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.
  • liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) provided herein, and a dialkylated mono- or poly-alkylene glycol, including, 1 ,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350- dimethyl ether, polyethylene glycol-550- dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol.
  • a dialkylated mono- or poly-alkylene glycol including, 1 ,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350- dimethyl ether, polyethylene glycol-550- dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol.
  • These formulations can further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
  • antioxidants such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
  • antioxidants such as
  • dosage unit formulations for oral administration can be microencapsulated.
  • the composition can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
  • compositions provided herein for oral administration can be also provided in the forms of liposomes, micelles, microspheres, or nanosystems.
  • Micellar dosage forms can be prepared as described in U.S. Pat. No. 6,350,458.
  • compositions provided herein can be provided as non-effervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form.
  • Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents.
  • Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide.
  • Coloring and flavoring agents can be used in all of the above dosage forms.
  • the compounds disclosed herein may also be coupled with soluble polymers as targetable drug carriers.
  • soluble polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxidepolylysme substituted with palmitoyl residues.
  • the compounds disclosed herein may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
  • compositions provided herein can be formulated as immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed-release forms.
  • compositions provided herein can be co-formulated with other active ingredients which do not impair the desired therapeutic action, or with substances that supplement the desired action.
  • compositions provided herein can be administered parenterally by injection, infusion, or implantation, for local or systemic administration.
  • Parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, intravesical, and subcutaneous administration.
  • compositions provided herein can be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection.
  • dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science (see, Remington: The Science and Practice of Pharmacy, supra).
  • compositions intended for parenteral administration can include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases.
  • aqueous vehicles water-miscible vehicles
  • non-aqueous vehicles non-aqueous vehicles
  • antimicrobial agents or preservatives against the growth of microorganisms stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emul
  • Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection.
  • Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil.
  • Water-miscible vehicles include, but are not limited to, ethanol, 1,3-butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, N-methyl-2-pyrrolidone, N,N-dimethylacetamide, and dimethyl sulfoxide.
  • liquid polyethylene glycol e.g., polyethylene glycol 300 and polyethylene glycol 400
  • propylene glycol e.g., N-methyl-2-pyrrolidone, N,N-dimethylacetamide, and dimethyl sulfoxide.
  • Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoates, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl- and propyl-parabens, and sorbic acid.
  • Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose.
  • Suitable buffering agents include, but are not limited to, phosphate and citrate.
  • Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite.
  • Suitable local anesthetics include, but are not limited to, procaine hydrochloride.
  • Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone.
  • Suitable emulsifying agents include those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate.
  • Suitable sequestering or chelating agents include, but are not limited to EDTA.
  • Suitable pH adjusting the pH adjustment include sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone.
  • Suitable emulsifying agents include those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate.
  • Suitable sequestering or chelating agents include
  • SUBSTITUTE SHEET RULE 26 agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid.
  • Suitable complexing agents include, but are not limited to, cyclodextrins, including a- cyclodextrin, ⁇ -cyclodextrin, hydroxypropyl-P-cyclodextrin, sulfobutylether-P-cyclodextrin, and sulfobutylether 7-P-cyclodextrin (CAPTISOL ® , CyDex, Lenexa, KS).
  • compositions provided herein can be formulated for single or multiple dosage administration.
  • the single dosage formulations are packaged in an ampoule, a vial, or a syringe.
  • the multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art.
  • the pharmaceutical compositions are provided as ready-to-use sterile solutions.
  • the pharmaceutical compositions are provided as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with a vehicle prior to use.
  • the pharmaceutical compositions are provided as ready-to-use sterile suspensions.
  • the pharmaceutical compositions are provided as sterile dry insoluble products to be reconstituted with a vehicle prior to use.
  • the pharmaceutical compositions are provided as ready-to-use sterile emulsions.
  • compositions provided herein can be formulated as immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed-release forms.
  • the pharmaceutical compositions can be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot.
  • the pharmaceutical compositions provided herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions diffuse through.
  • Suitable inner matrixes include polymethylmethacrylate, polybutyl-methacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinyl acetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinyl alcohol, and cross-linked partially hydrolyzed polyvinyl acetate.
  • Suitable outer polymeric membranes include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate
  • SUBSTITUTE SHEET RULE 26 copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinyl chloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer.
  • the pharmaceutical compositions disclosed herein can be formulated in any dosage forms that are adapted for administration to a patient by inhalation, for example as a dry powder, an aerosol, a suspension, or a solution composition.
  • the pharmaceutical compositions disclosed herein can be formulated in a dosage form adapted for administration to a patient by inhalation as a dry powder.
  • the pharmaceutical compositions disclosed herein can be formulated in a dosage form adapted for administration to a patient by inhalation via a nebulizer.
  • Dry powder compositions for delivery to the lung by inhalation typically comprise the compounds disclosed herein as a finely divided powder together with one or more pharmaceutically-acceptable excipients as finely divided powders.
  • compositions particularly suited for use in dry powders are known to those skilled in the art and include lactose, starch, mannitol, and mono-, di-, and polysaccharides.
  • the finely divided powder may be prepared by, for example, micronisation and milling.
  • the size-reduced (eg micronised) compound can be defined by a D50 value of about 1 to about 10 microns (for example as measured using laser diffraction).
  • Aerosols may be formed by suspending or dissolving the compound disclosed herein in a liquified propellant.
  • Suitable propellants include halocarbons, hydrocarbons, and other liquified gases.
  • Representative propellants include: trichlorofluoromethane (propellant 11), dichlorofluoromethane (propellant 12), dichlorotetrafluoroethane (propellant 114), tetrafluoroethane (HFA-134a), 1,1-difluoroethane (HFA-152a), difluoromethane (HFA-32), pentafluoroethane (HFA-12), heptafluoropropane (HFA-227a), perfluoropropane, perfluorobutane, perfluoropentane, butane, isobutane, and pentane. Aerosols comprising the compound disclosed herein will typically be administered to a patient via a metered
  • the aerosol may contain additional pharmaceutically-acceptable excipients typically used with MDIs such as surfactants, lubricants, cosolvents and other excipients to improve the physical stability of the formulation, to improve valve performance, to improve solubility, or to improve taste.
  • additional pharmaceutically-acceptable excipients typically used with MDIs such as surfactants, lubricants, cosolvents and other excipients to improve the physical stability of the formulation, to improve valve performance, to improve solubility, or to improve taste.
  • compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the patient for a
  • the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmaceutical Research, 3(6), 318 (1986).
  • compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.
  • Ointments, creams and gels may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents.
  • bases may thus, for example, include water and/or oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol.
  • Thickening agents and gelling agents which may be used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents.
  • Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents or thickening agents.
  • Powders for external application may be formed with the aid of any suitable powder base, for example, talc, lactose or starch. Drops may be formulated with an aqueous or nonaqueous base also comprising one or more dispersing agents, solubilizing agents, suspending agents or preservatives.
  • Topical preparations may be administered by one or more applications per day to the affected area; over skin areas occlusive dressings may advantageously be used. Continuous or prolonged delivery may be achieved by an adhesive reservoir system.
  • compositions may be applied as a topical ointment or cream.
  • the compound disclosed herein When formulated in an ointment, the compound disclosed herein may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the compound disclosed herein may be formulated in a cream with an oil-in- water cream base or a water-in-oil base.
  • the present invention provides a method of using a compound disclosed herein, or a pharmaceutical composition comprising the compound disclosed herein for the treatment, prevention, or amelioration of a disease or disorder that is mediated or otherwise affected via JAK kinase, including JAKl, JAK2, JAK3 or TYK2 kinase activiy or one or more symptoms of diseases or disorders that are mediated or otherwise affected via JAK kinase, including JAKl,
  • JAK kinase can be wild type and/or mutant form of JA l, JAK2, JAK3 or TYK2 kinase.
  • a method of using a compound disclosed herein or a pharmaceutical composition comprising a compound disclosed herein for the treatment, prevention, or amelioration of a disease or disorder that is mediated or otherwise affected via inappropriate JAKl kinase activity or one or more symptoms of diseases or disorders that are mediated or otherwise affected via inappropriate JAKl kinase activity.
  • a disease, a disorder or one or more symptoms of diseases or disorders is related to the inappropriate activity of JAK2 kinase.
  • a disease, a disorder or one or more symptoms of diseases or disorders is related to the inappropriate activity of JAK3 kinase.
  • Inappropriate JAK kinase activity refers to any JAK kinase activity that deviates from the normal JAK kinase activity expected in a particular patient. Inappropriate JAK kinase may take the form of, for instance, an abnormal increase in activity, or an aberration in the timing and or control of JAK kinase activity. Such inappropriate activity may result then, for example, from overexpression or mutation of the protein kinase leading to inappropriate or uncontrolled activation. Accordingly, in another aspect the invention is directed to methods of treating such diseases and disorders.
  • diseases or disorders include without limitation: myeloproliferative disorders such as polycythemia vera (PCV), essential thrombocythemia and idiopathic myelofibrosis (IMF); leukemia such as myeloid leukemia including chronic myeloid leukemia (CML), imatinib-resistant forms of CML, acute myeloid leukemia (AML), and a subtype of AML, acute megakaryoblastic leukemia (AMKL); lymphoproliferative diseases such as myeloma; cancer including head and neck cancer, prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, brain tumor, pancreatic cancer and renal carcinoma; and allergic or inflammatory diseases or disorders related to immune dysfunction, immunodeficiency, immunomodulation, autoimmune diseases, transplantation rejection, graft- versus-host disease, wound healing, kidney disease, multiple sclerosis, thyroiditis, type 1 diabetes, sarcoidosis
  • PCV polycythemia vera
  • provided herein is the compound or the pharmaceutical composition disclosed herein for preventing and/or treating proliferative disease, autoimmune disease, allergic disease, inflammatory disease or transplantation rejection in mammals including humans.
  • SUBSTITUTE SHEET RULE 26 [209]
  • a method of treating a mammal having, or at risk of having a disease or disclosed herein comprising administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or the compounds disclosed herein.
  • a method of treating a mammal having, or at risk of having proliferative disease, autoimmune disease, allergic disease, inflammatory disease or transplantation rejection comprising administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or the compounds disclosed herein.
  • a method of treatment and/or prophylaxis of a mammal susceptible to or afflicted with a proliferative disease comprising administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or compounds disclosed herein.
  • the proliferative disease is selected from cancer (e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer), polycythemia vera, essential thrombocytosis, myelofibrosis, leukemia (e.g. AML, CML, ALL or CLL), and multiple myeloma.
  • the compound or the pharmaceutical composition disclosed herein for use in the treatment, and/or prophylaxis of a proliferative disease.
  • the proliferative disease is selected from cancer (e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer), polycythemia vera, essential thrombocytosis, myelofibrosis, leukemia (e.g. AML, CML, ALL or CLL), and multiple myeloma.
  • the use of the compound or the pharmaceutical composition disclosed herein for use in the manufacture of a medicament for the treatment, and/or prophylaxis of a proliferative disease is selected from cancer (e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer), polycythemia vera, essential thrombocytosis, myelofibrosis, leukemia (e.g. AML, CML, ALL or CLL), and multiple myeloma.
  • cancer e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer
  • polycythemia vera e.g. uterine leiomyosarcoma or prostate cancer
  • essential thrombocytosis e.g. myelofibrosis
  • myelofibrosis e.g. AML, CML, ALL or CLL
  • multiple myeloma e.g. AML, CML, ALL or CLL
  • a method of treatment and/or prophylaxis of a mammal susceptible to or afflicted with an autoimmune disease comprise administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or compounds disclosed herein.
  • the autoimmune disease is selected from COPD, asthma, systemic and cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis and type I diabetes mellitus.
  • the compound or the pharmaceutical composition disclosed herein for use in the treatment, and/or prophylaxis of an autoimmune disease.
  • the autoimmune disease is selected from COPD, asthma, systemic and
  • SUBSTITUTE SHEET RULE 26 cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis and type I diabetes mellitus.
  • the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for the treatment, and/or prophylaxis of an autoimmune disease is selected from COPD, asthma, systemic and cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis and type I diabetes mellitus.
  • a mammal susceptible to or afflicted with an allergic disease comprising administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or the compounds disclosed herein.
  • the allergic disease is selected from allergic airway disease, sinusitis, eczema and hives, food allergies and allergies to insect venom.
  • the compound or the pharmaceutical composition disclosed herein for use in the treatment, and/or prophylaxis of an allergic disease.
  • the allergic disease is selected from allergic airway disease, sinusitis, eczema and hives, food allergies and allergies to insect venom.
  • the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for the treatment, or prophylaxis of an allergic disease is selected from allergic airway disease, sinusitis, eczema and hives, food allergies and allergies to insect venom.
  • kits for treatment and/or prophylaxis of a mammal susceptible to or afflicted with an inflammatory disease comprise administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or the compounds disclosed herein.
  • the inflammatory disease is selected from inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis and psoriatic arthritis.
  • the compound or the pharmaceutical composition disclosed herein for use in the treatment, and/or prophylaxis of an inflammatory disease.
  • the inflammatory disease is selected from inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis and psoriatic arthritis.
  • the inflammatory disease is selected from inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis and psoriatic arthritis.
  • transplantation rejection is organ transplant rejection, tissue transplant rejection and cell transplant rejection.
  • transplantation rejection is organ transplant rejection, tissue transplant rejection and cell transplant rejection.
  • transplantation rejection is organ transplant rejection, tissue transplant rejection and cell transplant rejection.
  • the present invention provides the compound or the pharmaceutical composition disclosed herein for use as a pharmaceutical especially in the treatment and/or prophylaxis of the aforementioned diseases or disorders. Also provided herein is the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for the treatment and/or prophylaxis of one of the aforementioned diseases or disorders.
  • a particular regimen of the present method comprises the administration to a subject suffering from a disease involving inflammation, of an effective amount of a compound disclosed herein for a period of time sufficient to reduce the level of inflammation in the subject, and preferably terminate the processes responsible for said inflammation.
  • a special embodiment of the method comprises administering of an effective amount of a compound disclosed herein to a subject patient suffering from or susceptible to the development of rheumatoid arthritis, for a period of time sufficient to reduce or prevent, respectively, inflammation in the joints of said patient, and preferably terminate, the processes responsible for said inflammation.
  • a further particular regimen of the present method comprises the administration to a subject suffering from a disease involving proliferative disease, of an effective amount of a compound disclosed herein for a period of time sufficient to reduce the level of proliferative
  • a particular embodiment of the method comprises administering of an effective amount of a compound disclosed herein to a subject patient suffering from or susceptible to the development of cancer, for a period of time sufficient to reduce or prevent, respectively, solid tumor of said patient, and preferably terminate, the processes responsible for said solid.
  • a compound disclosed herein can be administered as the sole active agent or it can be administered in combination with other therapeutic agents, including other compounds that demonstrate the same or a similar therapeutic activity and that are determined to be safe and efficacious for such combined administration.
  • provided herein is a method of treating, preventing, or ameliorating a disease or disorder comprising administering a safe and effective amount of a combination comprising the compound disclosed herein together with one or more therapeutically active agents. In one embodiment, the combinations comprising one or two other therapeutic agents.
  • Example of other therapeutic agents may include without limitation anti-cancer agents, including chemotherapeutic agents and antiproliferative agents; anti-inflammatory agents and immunomodulatory agents or immunosuppressive agents.
  • a product comprising a compound disclosed herein and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy.
  • the therapy is the treatment of a disease or disorder mediated by the activity of the JAK enzymes.
  • Products provided as a combined preparation include a composition comprising the compound disclosed herein and the other therapeutic agent(s) together in the same pharmaceutical composition, or the compound disclosed herein and the other therapeutic agent(s) in separate form, e.g. in the form of a kit.
  • a pharmaceutical composition comprising a compound disclosed herein and another therapeutic agent(s).
  • the pharmaceutical composition may comprise a pharmaceutically acceptable excipient, carrier, adjuvant or veichle as described above.
  • the invention provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound disclosed herein.
  • the kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet.
  • a container, divided bottle, or divided foil packet An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like.
  • the invention also provides the use of a compound disclosed herein for treating a disease or condition mediated by the activity of the JAK enzymes, wherein the patient has previously (e.g. within 24 hours) been treated with another therapeutic agent.
  • the invention also provides the use of another therapeutic agent for treating a disease or condition mediated by the activity of the JAK enzymes, wherein the patient has previously (e.g. within 24 hours) been treated with a compound disclosed herein.
  • the compounds disclosed herein may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to, other drugs e.g. immunosuppressive or immunomodulating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of alio- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders, or a chemotherapeutic agent, e.g a malignant cell anti-proliferative agent.
  • the compounds disclosed herein may be used in combination with a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a mTOR inhibitor, e.g.
  • rapamycin 40-O-(2- hydroxyethyl)-rapamycin, CCI779, ABT578, AP23573, TAFA-93, biolimus-7 or biolimus-9; an ascomycin having immuno-suppressive properties, e.g. ABT-281, ASM981, etc.; corticosteroids; cyclophosphamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenolic acid or salt; mycophenolate mofetil; 15- deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; a PKC inhibitor, e.g.
  • immunosuppressive monoclonal antibodies e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40, CD45, CD52, CD58, CD80, CD86 or their ligands
  • other immunomodulatory compounds e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g.
  • CTLA41g for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA- 1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or antihistamines; or antitussives, or a bronchodilatory agent; or an angiotensin receptor blockers; or an anti-infectious agent.
  • adhesion molecule inhibitors e.g. LFA- 1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists
  • antihistamines or antitussives, or a bronchodilatory agent
  • an angiotensin receptor blockers or an anti-infectious agent.
  • a combination comprising a compound disclosed herein together with a P2-adrenoreceptor agonist.
  • P2-adrenoreceptor agonists include salmeterol, salbutamol, formoterol, salmefamol, fenoterol, carmoterol, etanterol, naminterol, clenbuterol, pirbuterol, flerbuterol, reproterol, bambuterol, indacaterol, terbutaline and salts thereof, for example the xinafoate (l-hydroxy-2-naphthalenecarboxylate) salt of salmeterol, the sulphate salt or free base of salbutamol or the fumarate salt of formoterol.
  • long-acting P2-adrenoreceptor agonists for example, compounds which provide effective bronchodilation for about 12 h or longer
  • the P2-adrenoreceptor agonist may be in the form of a salt formed with a pharmaceutically acceptable acid selected from sulphuric, hydrochloric, fumaric, hydroxynaphthoic (for example 1- or 3-hydroxy-2-naphthoic), cinnamic, substituted cinnamic, triphenylacetic, sulphamic, sulphanilic, naphthaleneacrylic, benzoic, 4-methoxybenzoic, 2- or 4-hydroxybenzoic, 4-chlorobenzoic and 4- phenylbenzoic acid.
  • a pharmaceutically acceptable acid selected from sulphuric, hydrochloric, fumaric, hydroxynaphthoic (for example 1- or 3-hydroxy-2-naphthoic), cinnamic, substituted cinnamic, triphenylacetic, sulphamic, sulphanilic, naphthaleneacrylic, benzoic, 4-meth
  • a combination comprising a compound disclosed herein together with corticosteroids.
  • Suitable corticosteroids refer to those oral and inhaled corticosteroids and their pro-drugs which have anti-inflammatory activity.
  • Examples include methyl prednisolone, prednisolone, dexamethasone, fluticasone propionate, 6a,9a-difluoro-l 1 ⁇ - hydroxy- 16a-methyl- 17a- [(4-methyl- 1 ,3 -thiazole-5 -carbonyl)oxy] -3 -oxo-androsta- 1 ,4-diene- 17 ⁇ - carbothioic acid (Sj-fluoromethyl ester, 6a,9a-difluoro-17a-[(2-furanylcarbonyl)oxy]-l 1 ⁇ - hydroxy-16a-methyl-3 -oxo-androsta- 1 ,4-diene-l 7 ⁇ -carbothioic acid (5 ⁇ -fluoromethyl ester (fluticasone furoate), 6a,9a-difluoro-l ⁇ -hydroxy-16a-methyl-3-oxo-17a-propionyloxy-androsta- acid (S
  • Preferred corticosteroids include fluticasone propionate, 6a,9a-difluoro-l ⁇ -hydroxy-16a-methyl- 17 -[(4-methyl-l,3-thiazole-5-carbonyl)oxy]-3-oxo-androsta-l ,4-diene-17 ⁇ -carbothioic acid (S)- fluoromethyl ester, 6a,9a-difluoro-17a-[(2-furanylcarbonyl)oxy]-l ⁇ -hydroxy-16a-methyl-3-oxo- androsta-l ,4-diene-17 ⁇ -carbothioic acid (5)-fluoromethyl ester, 6a,9a-difluoro-l ⁇ -hydroxy-16a- methyl-3-oxo-17a-(2,2 -tetramethycyclopropylcarbonyl)oxy-androsta-l ,4-diene-17P-
  • SUBSTITUTE SHEET RULE 26 carbothioic acid (S)-cyanomethyl ester and 6a,9a-difluoro-l ip-hydroxy-16a-methyl-17a-(l- methyl cyclopropylcarbonyl)oxy-3-oxo-androsta-l,4-diene-17P-carbothioic acid (5)-fluoromethyl ester.
  • the corticosteroid is 6a, 9a-difluoro-17a-[(2-furanylcarbonyl)oxy]-l ip- hydroxy-16a-methyl-3-oxo-androsta-l,4-diene-17P-carbothioic acid (5)-fluoromethyl ester.
  • Non-steroidal compounds having glucocorticoid agonism that may possess selectivity for transrepression over transactivation and that may be useful in combination therapy include those covered in the following patents: WO 03/082827, WO 98/54159, WO 04/005229, WO 04/009017, WO 04/018429, WO 03/104195, WO 03/082787, WO 03/082280, WO 03/059899, WO 03/101932, WO 02/02565, WO 01/16128, WO 00/66590, WO 03/086294, WO 04/026248, WO 03/061651 and WO 03/08277. Further non-steroidal compounds are covered in: WO 2006/000401, WO 2006/000398 and WO 2006/015870.
  • NSAID's non-steroidal anti-inflammatory drugs
  • NSAID's include sodium cromoglycate, nedocromil sodium, phosphodiesterase (PDE) inhibitors (for example, theophylline, PDE4 inhibitors or mixed PDE3/PDE4 inhibitors), leukotriene antagonists, inhibitors of leukotriene synthesis (for example montelukast), iNOS inhibitors, tryptase and elastase inhibitors, beta-2 integrin antagonists and adenosine receptor agonists or antagonists (e.g.
  • adenosine 2a agonists adenosine 2a agonists
  • cytokine antagonists for example chemokine antagonists, such as a CCR3 antagonist
  • inhibitors of cytokine synthesis or 5 -lipoxygenase inhibitors.
  • An iNOS (inducible nitric oxide synthase inhibitor) is preferably for oral administration.
  • iNOS inhibitors include those disclosed in WO 93/13055, WO 98/30537, WO 02/50021, WO 95/34534 and WO 99/62875.
  • CCR3 inhibitors include those disclosed in WO 02/26722.
  • the invention provides the use of the compounds disclosed herein in combination with a phosphodiesterase 4 (PDE4) inhibitor, especially in the case of a formulation adapted for inhalation.
  • PDE4-specific inhibitor useful in this aspect of the invention may be any compound that is known to inhibit the PDE4 enzyme or which is discovered to act as a PDE4 inhibitor, and which are only PDE4 inhibitors, not compounds which inhibit other members of the PDE family, such as PDE3 and PDE5, as well as PDE4.
  • Compounds include czs-4-cyano-4-(3- cyclopentyloxy-4- methoxyphenyl)cyclohexan-l-carboxylic acid, 2-carbomethoxy-4-cyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan- 1 -one and cis- [4-cyano-4-(3 - cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-l-ol].
  • czs -4-cyano-4-[3- (cyclopentyloxy)-4-methoxyphenyl]cyclohexane-l-carboxylic acid also known as cilomilast
  • salts, esters, pro-drugs or physical forms which is described in U.S. Patent No. 5,552,438
  • a combination comprising a compound disclosed herein together with an anticholinergic agent.
  • anticholinergic agents are those compounds that act as antagonists at the muscarinic receptors, in particular those compounds which are antagonists of the Mi or M 3 receptors, dual antagonists of the Mi/M 3 or ⁇ 2 / ⁇ 3 , receptors or pan-antagonists of the Mi/M 2 /M 3 receptors.
  • Exemplary compounds for administration via inhalation include ipratropium (for example, as the bromide, CAS 22254-24-6, sold under the name ATROVENT ® ), oxitropium (for example, as the bromide, CAS 30286-75-0) and tiotropium (for example, as the bromide, CAS 136310-93-5, sold under the name SPIRIVA ® ). Also of interest are revatropate (for example, as the hydrobromide, CAS 262586-79-8) and LAS-34273 which is disclosed in WO 01/04118.
  • Exemplary compounds for oral administration include pirenzepine (CAS 28797-61-7), darifenacin (CAS 133099-04-4, or CAS 133099-07-7 for the hydrobromide sold under the name ENABLEX ® ), oxybutynin (CAS 5633-20-5, sold under the name DITROPAN ® ), terodiline (CAS 15793-40-5), tolterodine (CAS 124937-51-5, or CAS 124937-52- 6 for the tartrate, sold under the name DETROL ® ), otilonium (for example, as the bromide, CAS 26095-59-0, sold under the name SPASMOMEN ® ), trospium chloride (CAS 10405-02-4) and solifenacin (CAS 242478-37-1, or CAS 242478-38-2 for the succinate also known as YM-905 and sold under the name VESICARE ® ).
  • pirenzepine CAS 28797-61
  • HI antagonists include, without limitation, amelexanox, astemizole, azatadine, azelastine, acrivastine, brompheniramine, cetirizine, levocetirizine, efletirizine, chlorpheniramine, clemastine, cyclizine, carebastine, cyproheptadine, carbinoxamine, descarboethoxyloratadine, doxylamine, dimethindene, ebastine, epinastine, efletirizine, fexofenadine, hydroxyzine, ketotifen, loratadine, levocabastine, mizolastine, mequitazine, mianserin, noberastine, meclizine, norastemizole, olopatadine, picum
  • the invention provides a combination comprising a compound disclosed herein together with an H3 antagonist (and/or inverse agonist).
  • H3 antagonists include, for example, those compounds disclosed in WO 2004/035556 and in WO 2006/045416.
  • Other histamine receptor antagonists which may be used in combination with the compounds disclosed herein include antagonists (and/or inverse agonists) of the H4 receptor, for example, the compounds disclosed in Jablonowski et al, J. Med. Chem. 46:3957-3960 (2003).
  • SUBSTITUTE SHEET RULE 26 [245] In still another aspect, provided herein is a combination comprising a compound disclosed herein together with a PDE4 inhibitor and a P2-adrenoreceptor agonist.
  • a combination comprising a compound disclosed herein together with an anticholinergic and a PDE-4 inhibitor.
  • compositions comprising a combination as defined above together with a pharmaceutically acceptable excipient or carrier represent a further aspect of the invention.
  • the individual compounds of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations.
  • the individual compounds will be administered simultaneously in a combined pharmaceutical formulation.
  • Appropriate doses of known therapeutic agents will readily be appreciated by those skilled in the art.
  • the invention thus provides, in a further aspect, a pharmaceutical composition comprising a combination of a compound disclosed herein together with another therapeutically active agent.
  • the pharmaceutical composition comprises a combination of a compound disclosed herein together with a PDE4 inhibitor.
  • the pharmaceutical composition comprises a combination of a compound disclosed herein together with a P2-adrenoreceptor agonist.
  • the pharmaceutical composition comprises a combination of a compound disclosed herein together with a corticosteroid.
  • the pharmaceutical composition comprises a combination of a compound disclosed herein together with a non-steroidal GR agonist.
  • the pharmaceutical composition comprises a combination of a compound disclosed herein together with an anticholinergic agent.
  • the pharmaceutical composition comprises a combination of a compound disclosed herein together with an antihistamine.
  • compositions disclosed herein may be, for example, surgery, radiotherapy, chemotherapy, signal transduction inhibitors or modulators (e.g. kinase inhibitors or modulators) and/or monoclonoal antibodies.
  • a compound disclosed herein may also be used to advantage in combination with each other or in combination with other therapeutic agents, especially other antiproliferative agents.
  • antiproliferative agents include, but are not limited to, aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active agents; alkylating agents; histone deacetylase inhibitors; compounds that induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies;
  • aromatase inhibitor relates to a compound which inhibits the estrogen production, i.e., the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively.
  • the term includes, but is not limited to, steroids, especially atamestane, exemestane and formestane; and, in particular, nonsteroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketoconazole, vorozole, fadrozole, anastrozole and letrozole.
  • Exemestane can be administered, e.g., in the form as it is marketed, e.g., under the trademark AROMASIN ® .
  • Formestane can be administered, e.g., in the form as it is marketed, e.g., under the trademark LENTARON ® .
  • Fadrozole can be administered, e.g., in the form as it is marketed, e.g., under the trademark AFEMA ® .
  • Anastrozole can be administered, e.g., in the form as it is marketed, e.g., under the trademark ARIMIDEX ® .
  • Letrozole can be administered, e.g., in the form as it is marketed, e.g., under the trademark FEMARA ® or FEMAR ® .
  • Aminoglutethimide can be administered, e.g., in the form as it is marketed, e.g., under the trademark ORIMETEN ® .
  • a combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, e.g., breast tumors.
  • anti-estrogen relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level.
  • the term includes, but is not limited to, tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride.
  • Tamoxifen can be administered, e.g., in the form as it is marketed, e.g., under the trademark NOLVADEX ® .
  • Raloxifene hydrochloride can be administered, e.g., in the form as it is marketed, e.g., under the trademark EVISTA ® .
  • Fulvestrant can be formulated as disclosed in U.S. Patent No. 4,659,516 or it can be
  • SUBSTITUTE SHEET RULE 26 administered, e.g., in the form as it is marketed, e.g., under the trademark FASLODEX ® .
  • a combination of the invention comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, e.g., breast tumors.
  • anti-androgen relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (CASODEX ® ), which can be formulated, e.g., as disclosed in U.S. Patent No. 4,636,505.
  • gonadorelin agonist includes, but is not limited to, abarelix, goserelin and goserelin acetate.
  • Goserelin is disclosed in U.S. Patent No. 4,100,274 and can be administered, e.g., in the form as it is marketed, e.g., under the trademark ZOLADEX ® .
  • Abarelix can be formulated, e.g., as disclosed in U.S. Patent No. 5,843,901.
  • topoisomerase I inhibitor includes, but is not limited to, topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148 (compound Al in WO 99/17804).
  • Irinotecan can be administered, e.g., in the form as it is marketed, e.g., under the trademark CAMPTOSAR ® .
  • Topotecan can be administered, e.g., in the form as it is marketed, e.g., under the trademark HYCAMTIN ® .
  • topoisomerase II inhibitor includes, but is not limited to, the anthracyclines, such as doxorubicin, including liposomal formulation, e.g., CAELYX ® ; daunorubicin; epirubicin; idarubicin; nemorubicin; the anthraquinones mitoxantrone and losoxantrone; and the podophillotoxines etoposide and teniposide.
  • Etoposide can be administered, e.g., in the form as it is marketed, e.g., under the trademark ETOPOPHOS ® .
  • Teniposide can be administered, e.g., in the form as it is marketed, e.g., under the trademark VM 26-BRISTOL ® .
  • Doxorubicin can be administered, e.g., in the form as it is marketed, e.g., under the trademark ADRIBLASTIN ® or ADRIAMYCIN ® .
  • Epirubicin can be administered, e.g., in the form as it is marketed, e.g., under the trademark FARMORUBICIN ® .
  • Idarubicin can be administered, e.g., in the form as it is marketed, e.g., under the trademark ZAVEDOS ® .
  • Mitoxantrone can be administered, e.g., in the form as it is marketed, e.g., under the trademark NOVANTRON ® .
  • microtubule active agent relates to microtubule stabilizing, microtubule destabilizing agents and microtublin polymerization inhibitors including, but not limited to, taxanes, e.g., paclitaxel and docetaxel; vinca alkaloids, e.g., vinblastine, especially vinblastine sulfate; vincristine, especially vincristine sulfate and vinorelbine; discodermolides; cochicine; and epothilones and derivatives thereof, e.g., epothilone B or D or derivatives thereof.
  • Paclitaxel may
  • SUBSTITUTE SHEET RULE 26 be administered, e.g., in the form as it is marketed, e.g., TAXOL ® .
  • Docetaxel can be administered, e.g., in the form as it is marketed, e.g., under the trademark TAXOTERE ® .
  • Vinblastine sulfate can be administered, e.g., in the form as it is marketed, e.g., under the trademark VINBLASTIN R.P ® .
  • Vincristine sulfate can be administered, e.g., in the form as it is marketed, e.g., under the trademark FARMISTIN ® .
  • Discodermolide can be obtained, e.g., as disclosed in U.S. Patent No. 5,010,099. Also included are epothilone derivatives which are disclosed in WO 98/10121, U.S. Patent No. 6,194,181, WO 98/25929, WO 98/08849, WO 99/43653, WO 98/22461 and WO 00/31247. Especially preferred are epothilone A and/or B.
  • alkylating agent includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel).
  • Cyclophosphamide can be administered, e.g., in the form as it is marketed, e.g., under the trademark CYCLOSTIN ® .
  • Ifosfamide can be administered, e.g., in the form as it is marketed, e.g., under the trademark HOLOXAN ® .
  • histone deacetylase inhibitors or "HDAC inhibitors” relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes compounds disclosed in WO 02/22577, especially N-hydroxy-3-[4-[[(2-hydroxyethyl)[2-(lH- indo]-3-y ethy[]-amino]methyl]phenyl]-2E-2-propenamide, A3 ⁇ 4ydroxy-3-[4-[[[2-(2-methyl-l H- indol-3-yl)-ethy[]-amino]methyl]phenyl]-2E-2-propenamide and pharmaceutically acceptable salts thereof. It further especially includes suberoyianilide hydroxamic acid (SAHA).
  • SAHA suberoyianilide hydroxamic acid
  • anti-plastic antimetabolite includes, but is not limited to, 5-fluorouracil or 5- FU; capecitabine; gemcitabine; DNA demethylating agents, such as 5-azacytidine and decitabine; methotrexate and edatrexate; and folic acid antagonists, such as pemetrexed.
  • Capecitabine can be administered, e.g., in the form as it is marketed, e.g., under the trademark XELODA ® .
  • Gemcitabine can be administered, e.g., in the form as it is marketed, e.g., under the trademark GEMZAR ® .
  • the monoclonal antibody trastuzumab which can be administered, e.g., in the form as it is marketed, e.g., under the trademark HERCEPTIN ® .
  • platinum compound includes, but is not limited to, carboplatln, cis-platin, cisplatinum and oxaliplatin.
  • Carboplatin can be administered, e.g., in the form as it is marketed, e.g., under the trademark CARBOPLAT ® .
  • Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g., under the trademark ELOXATIN ® .
  • SUBSTITUTE SHEET RULE 26 is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, e.g., a) compounds targeting, decreasing or inhibiting the activity of the p!ate!et-derived growth factor- receptors (PDGFR), such as compounds which target, decrease or mhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, e.g., a .
  • PDGFR p!ate!et-derived growth factor- receptors
  • -phenyl-2-pyrirnidine-arnine derivative e.g., imatinib, SU101 , SU6668 and GFB-1 1 1
  • IGF-IR insulin-like growth factor receptor I
  • compounds such as compounds which target, decrease or mhibit the activity of IGF-IR, especially compounds which inhibit the IGF-IR receptor, such as those compounds disclosed in WO 02/092599
  • a tyrphostin is preferably a low molecular weight (Mr ⁇ 1500) compound, or a pharmaceutically acceptable salt thereof, especially a compound selected from the benzylidenemalonitrile class or the S-arylbenzenemalonudie or bisubstrate quinoiine class of compounds, more especially any compound selected from the group consisting of Tyrphostin A23/RG-5081 Q, AG 99, Tyrphostin AG 213, Tyrphostin AG 1748, Tyrphostin AG 490, Tyrphostin B44, Tyrphostin B44 (+) enantiomer, Tyrphostin AG 555, AG 494, Tyrphostin AG 556, AG957 and adaphostin (4- ⁇ [(2,5-d.ihyd.roxyphenyl)methyl]amino ⁇ - benzoie acid adamantyl ester, NSC 680410, adaphostin; and
  • compoimds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, e.g., EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, and are in particular those compounds, proteins or monoclonal antibodies generically and specifical ly disclosed in W 97/02266, e.g., the compound of Example 39, or in EP 0564409; WO 99/03854; EP 0520722; EP 0566226; EP 0787722; EP 0837063; U.S.
  • HE CEPTIN trastuzumab
  • cetuximab cetuximab
  • Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g., unrelated to protein or lipid kinase inhibition, e.g., thalidomide (THALOMID ® ) and TNP-470.
  • TAALOMID ® thalidomide
  • TNP-470 TNP-470.
  • Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are, e.g., inhibitors of phosphatase 1, phosphatase 2A, PTEN or CDC25, e.g., okadaic acid or a derivative thereof,
  • SUBSTITUTE SHEET RULE 26 Compounds that induce cell differentiation processes are e.g. retinoic acid, ⁇ -, ⁇ - or ⁇ - tocopherol or ⁇ -, ⁇ - or ⁇ -tocotrienol.
  • cyclooxygenase inhibitor includes, but is not limited to, e.g., Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CELEBREX ® ), rofecoxib (VIOXX ® ), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, e.g., 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid or lumiracoxib.
  • Cox-2 inhibitors such as celecoxib (CELEBREX ® ), rofecoxib (VIOXX ® ), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, e.g., 5-methyl-2-(2'-chloro-6'-fluoroanilino)phen
  • bisphosphonates includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid.
  • Etridonic acid can be administered, e.g., in the form as it is marketed, e.g., under the trademark DIDRONEL ® .
  • Clodronic acid can be administered, e.g., in the form as it is marketed, e.g., under the trademark BONEFOS ® .
  • Titanium acid can be administered, e.g., in the form as it is marketed, e.g., under the trademark SKELID ® .
  • Pamidronic acid can be administered, e.g., in the form as it is marketed, e.g., under the trademark AREDIATM.
  • AREDIATM e.g., AREDIATM
  • Alendronic acid can be administered, e.g., in the form as it is marketed, e.g., under the trademark FOSAMAX ® .
  • Ibandronic acid can be administered, e.g., in the form as it is marketed, e.g., under the trademark BONDRANAT ® .
  • “Risedronic acid” can be administered, e.g., in the form as it is marketed, e.g., under the trademark ACTONEL ® .
  • "Zoledronic acid” can be administered, e.g., in the form as it is marketed, e.g., under the trademark ZOMETA ® .
  • mTOR inhibitors relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity, such as sirolimus (Rapamune ® ), everolimus (CerticanTM), CCI-779 and ABT578.
  • heparanase inhibitor refers to compounds which target, decrease or inhibit heparin sulphate degradation.
  • the term includes, but is not limited to, PI-88.
  • biological response modifier refers to a lymphokine or interferons, e.g., interferon ⁇ .
  • inhibitor of Ras oncogenic isoforms e.g., H-Ras, K-Ras or N-Ras, as used herein, refers to compounds which target, decrease or inhibit the oncogenic activity of Ras, e.g., a "farnesyl transferase inhibitor”, e.g., L-744832, DK8G557 or Rl 15777 (Zarnestra).
  • telomerase inhibitor refers to compounds which target, decrease or inhibit the activity of telomerase.
  • Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, e.g., telomestatin.
  • methionine aminopeptidase inhibitor refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase.
  • Compounds which target, decrease or inhibit the activity of methionine aminopeptidase are, e.g., bengamide or a derivative thereof.
  • proteasome inhibitor refers to compounds which target, decrease or inhibit the activity of the proteasome.
  • Compounds which target, decrease or inhibit the activity of the proteasome include, e.g., PS-341 and MLN 341.
  • matrix metalloproteinase inhibitor or "MMP inhibitor”, as used herein, includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g., hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ996.
  • MMP inhibitor matrix metalloproteinase inhibitor
  • agents used in the treatment of hematologic malignancies includes, but is not limited to, FMS-like tyrosine kinase inhibitors, e.g., compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1- b-D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors, e.g., compounds which target, decrease or inhibit anaplastic lymphoma kinase.
  • FMS-like tyrosine kinase inhibitors e.g., compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1- b-D-arabinofuransylcytosine (ara-c) and bisulfan
  • ALK inhibitors e.g., compounds which target, decrease or inhibit anaplastic lymphoma kina
  • FMS-like tyrosine kinase receptors are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, e.g., PKC412, midostaurin, a staurosporine derivative, SU1 1248 and MLN518.
  • HSP90 inhibitors includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteasome pathway.
  • Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90, e.g., 17- allylamino, 17-demethoxygeldanamycin (17AAG), a geldanamycin derivative, other geldanamycin related compounds, radicicol and HDAC inhibitors.
  • antiproliferative antibodies includes, but is not limited to, trastuzumab (HERCEPTINTM), Trastuzumab-DMl , erlotinib (TARCEVATM), bevacizumab (AVASTINTM), rituximab (RITUXAN ® ), PR064553 (anti-CD40) and 2C4 antibody.
  • HERCEPTINTM trastuzumab
  • Trastuzumab-DMl erlotinib
  • AVASTINTM bevacizumab
  • rituximab rituximab
  • PR064553 anti-CD40
  • SUBSTITUTE SHEET RULE 26 biological activity For the treatment of acute myeloid leukemia (AML), compounds disclosed herein can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML. In particular, compounds disclosed herein can be administered in combination with, e.g., farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
  • drugs useful for the treatment of AML such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
  • a compound disclosed herein may also be used to advantage in combination with each other or in combination with other therapeutic agents, especially other anti-malarial agents.
  • anti-malarial agents include, but are not limited to proguanil, chlorproguanil, trimethoprim, chloroquine, mefloquine, lumefantrine, atovaquone, pyrimethamine-sulfadoxine, pyrimethamine- dapsone, halofantrine, quinine, quinidine, amodiaquine, amopyroquine, sulphonamides, artemisinin, arteflene, artemether, artesunate, primaquine, inhaled NO, L-arginine, Dipropylenetri- amine NONOate (NO donor), Rosiglitzone (PPAR- ⁇ agonist), activated charcoal, Erythropoietin, Levamisole, and pyronaridine.
  • anti-malarial agents include, but are not limited to proguanil, chlor
  • a compound disclosed herein may also be used to advantage in combination with each other or in combination with other therapeutic agents, such as used for the treatment of Leishmaniosis, Trypanosomiasis, Toxoplasmosis and Neurocysticercosis.
  • Such agents include, but are not limited to chloroquine sulfate, atovaquone-proguanil, artemether- lumefantrine, quinine- sulfate, artesunate, quinine, doxycycline, clindamycin, meglumine antimoniate, sodium stibogluconate, miltefosine, ketoconazole, pentamidine, amphotericin B (AmB), liposomal- AmB, paromomycine, eflornithine, nifurtimox, suramin, melarsoprol, prednisolone, benznidazole, sulfadiazine, pyrimethamine, clindamycin, trimetropim, sulfamethoxazole, azitromycin, atovaquone, dexamethasone, praziquantel, albendazole, beta-lactams, fluoroquinolones
  • a compound disclosed herein may also be used to advantage in combination with known therapeutic processes, e.g., the administration of hormones or especially radiation.
  • a compound disclosed herein may also be used to advantage in combination with known therapeutic processes, e.g., the administration of hormones or especially radiation.
  • SUBSTITUTE SHEET RULE 26 disclosed herein may in particular be used as a radiosensitizer, especially for the treatment of tumors which exhibit poor sensitivity to radiotherapy.
  • ком ⁇ онент there is meant either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound disclosed herein and a combination partner may be administered independently at the same time or separately within time intervals that especially allow that the combination partners show a cooperative, e.g., synergistic, effect or any combination thereof.
  • coadministration or “combined administration” or the like as utilized herein are meant to encompass administration of the selected combination partner to a single subject in need thereof (e.g. a patient), and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
  • pharmaceutical combination means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non- fixed combinations of the active ingredients.
  • fixed combination means that the active ingredients, e.g. a compound disclosed herein and a combination partner, are both administered to a patient simultaneously in the form of a single entity or dosage.
  • non-fixed combination means that the active ingredients, e.g. a compound disclosed herein and a combination partner, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient.
  • cocktail therapy e.g. the administration of three or more active ingredients.
  • the methods of treatment disclosed herein comprise administering a safe and effective amount of a compound or a pharmaceutically composition disclosed herein to a patient in need thereof.
  • Individual embodiments disclosed herein include methods of treating any one of the above-mentioned disorders by administering a safe and effective amount of a compound disclosed herein or a pharmaceutical composition containing a compound disclosed herein to a patient in need thereof.
  • the compounds disclosed herein or pharmaceutically compositions containing the compounds disclosed herein may be administered by any suitable route of administration, including both systemic administration and topical administration.
  • Systemic administration includes oral administration, parenteral administration, transdermal administration and rectal administration.
  • Parenteral administration is typically by injection or infusion, including intravenous, intramuscular, and subcutaneous injection or infusion.
  • Topical administration includes application to the skin as well as intraocular, otic, intravaginal, inhaled and intranasal
  • the compounds disclosed herein or pharmaceutical compositions containing the compounds disclosed herein may be administered orally. In another embodiment, the compounds disclosed herein or pharmaceutically compositions containing the compounds disclosed herein may be administered by inhalation. In a further embodiment, the compounds disclosed herein or pharmaceutical compositions containing the compounds disclosed herein may be administered intranasally.
  • the compounds disclosed herein or pharmaceutically compositions containing the compounds disclosed herein may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time.
  • doses may be administered one, two, three, or four times per day.
  • a dose is administered once per day.
  • a dose is administered twice per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect.
  • Suitable dosing regimens for a compound disclosed herein or a pharmaceutical composition containing a compound disclosed herein depend on the pharmacokinetic properties of that compound, such as absorption, distribution, and half-life, which can be determined by the skilled artisan.
  • suitable dosing regimens, including the duration such regimens are administered, for a compound disclosed herein or a pharmaceutical composition containing a compound disclosed herein depend on the disorder being treated, the severity of the disorder being treated, the age and physical condition of the patient being treated, the medical history of the patient to be treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual patient's response to the dosing regimen or over time as individual patient needs change.
  • the compound of the present invention may be administered either simultaneously with, or before or after, one or more other therapeutic agent.
  • the compound of the present invention may be administered separately, by the same or different route of administration, or together in the same pharmaceutical composition as the other agents.
  • the pharmaceutical composition or combination of the present invention can be in unit dosage of about 1-1000 mg of active ingredient(s) for a subject of about 50-70 kg, or about 1-500 mg or about 1-250 mg or about 1-150 mg or about 0.5-100 mg, or about 1- 50 mg of active ingredients.
  • the therapeutically effective dosage of a compound, the pharmaceutical composition, or the combinations thereof, is dependent on the species of the subject, the body weight, age and individual condition, the disorder or disease or the severity thereof being treated. A physician,
  • SUBSTITUTE SHEET RULE 26 clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to prevent, treat or inhibit the progress of the disorder or disease.
  • the above-cited dosage properties are demonstrable in vitro and in vivo tests using advantageously mammals, e.g., mice, rats, dogs, monkeys or isolated organs, tissues and preparations thereof.
  • the compounds of the present invention can be applied in vitro in the form of solutions, e.g., aqueous solutions, and in vivo either enterally, parenterally, advantageously intravenously, e.g., as a suspension or in aqueous solution.
  • the therapeutically effective dose is from about 0.1 mg to about 2,000 mg per day of a compound provided herein.
  • the pharmaceutical compositions therefore should provide a dosage of from about 0.1 mg to about 2000 mg of the compound.
  • pharmaceutical dosage unit forms are prepared to provide from about 1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 20 mg to about 500 mg or from about 25 mg to about 250 mg of the essential active ingredient or a combination of essential ingredients per dosage unit form.
  • the pharmaceutical dosage unit forms are prepared to provide about 10 mg, 20 mg, 25 mg, 50 mg, 100 mg, 250 mg, 500 mg, 1000 mg or 2000 mg of the essential active ingredient.
  • a prodrug of a compound disclosed herein is a functional derivative of the compound which, upon administration to a patient, eventually liberates the compound disclosed herein in vivo.
  • Administration of a compound disclosed herein as a prodrug may enable the skilled artisan to do one or more of the following: (a) modify the onset of the activity of the compound in vivo; (b) modify the duration of action of the compound in vivo; (c) modify the transportation or distribution of the compound in vivo; (d) modify the solubility of the compound in vivo; and (e) overcome a side effect or other difficulty encountered with the compound.
  • Typical functional derivatives used to prepare prodrugs include modifications of the compound that are chemically or enzymatically cleavable in vivo. Such modifications, which include the preparation of phosphates, amides, esters, thioesters, carbonates, and carbamates, are well known to those skilled in the art.
  • the compounds in this invention may be prepared by methods described herein, wherein the substituents are as defined for Formula (I), above, except where further noted.
  • SUBSTITUTE SHEET RULE 26 following non-limiting schemes and examples are presented to further exemplify the invention.
  • Persons skilled in the art will recognize that the chemical reactions described herein may be readily adapted to prepare a number of other compounds of the invention, and alternative methods for preparing the compounds of this invention are deemed to be within the scope of this invention.
  • the synthesis of non-exemplified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions.
  • other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention.
  • H NMR spectra were recorded with a Bruker 400 MHz or 600 MHz spectrometer at ambient temperature. ! H NMR spectra were obtained as CDCI3, DMSO- i, CD3OD or acetone- ⁇ i6 solutions (reported in ppm), using TMS (0 ppm) or chloroform (7.26 ppm) as the reference standard. When peak multiplicities are reported, the following abbreviations are used: s (singlet), d (doublet), t (triplet), m (multiplet), br (broadened), dd (doublet of doublets), dt (doublet of triplets), td (triplet of doublets). Coupling constants, when given, are reported in Hertz (Hz).
  • ester group in compound (17) is then converted to carboxylic acid (18) through basic hydrolysis.
  • Compound (18) is allowed to condense with amine compound (9) to pro vied amide (19).
  • Halogenation of compound (29) by POBr 3 affords azole (30), azole (30) is couple with borate acid (16) in the presence of a suitable Pd catalyst, such as PdCl2(dppf).DCM, affords compound (31). The ester group in compound (31) is then converted to carboxylic acid (32) through basic hydrolysis. Compound (32) is allowed to condense with amine compound (9) to affords the desired kinase inhibitor (33).
  • Some compounds having formula (36) can be prepared by a general method illustrated in Scheme 5 and described in details in the examples. As showing in Scheme 5, Condensation of azole (28) with compound (5) under an acidic condition provides amino azole (34) at elvated temperature. The ester group in compound (34) is then converted to carboxylic acid (35) through basic hydrolysis. Compound (35) is allowed to condense with amine compound (9) to affords the desired kinase inhibitor (36).
  • Step 1) fert-butyl (l-(5-cvanopyridin-2-yl)piperidin-4-yl)carbamate
  • Step 1) fert-butyl (l-(5-cyanopyridin-2-yl)piperidin-3-yl)carbamate
  • Step 2 fert-butyl 4-(((benzyloxy)carbonyl)(methyl)amino)piperidine-l-carboxylate
  • Step 1) fert-butyl 4-(2-((tert-butoxycarbonyl)amino)pyrazolon,5-a1pyrimidine-3- carboxamido)piperidine- 1 -carboxylate
  • Step 1) tert-butyl (3-((3-hydroxycyclohexyl)carbamoyl)pyrazolo[l,5-alpyrimidin-2-yl)carbamate
  • Step 2) tert-butyl (3-((l-(2-cyanoacetyl)pyrrolidin-3-yl)carbamoyl)pyrazolo[l,5-alpyrimidin-2- yPcarbamate
  • Step 1) fert-butyl (l-(5-cvanopyridin-2-yl)azetidin-3-yl)carbamate
  • Step 2) (6 -6-(3-aminopyrrolidin-l-yl)nicotinonitrile
  • Step 4) (6 -2-amino-N-(l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)pyrazolo[l , ⁇ pyrimidine-3- carboxamide
  • Step 1) (i?)-fert-butyl (l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamate
  • Step 2) (i?)-6-(3-aminopyrrolidin-l-yl)nicotinonitrile
  • Step 9) fert-butyl 3 -( 1 -(5 -cvanopyridin-2-yl)piperidin-4-yl carbamoyl) ⁇ - cvclopropyrpyrazolo[ 1 ,5-a]pyrimidin-2-yl carbamate
  • Step 2) ethyl 2-(bis(tert-butoxycarbonyl)amino)-5-chloropyrazolori,5-a1pyrimidine-3-carboxylat
  • the LC/MS/MS system used in the analysis consists of an Agilent 1200 Series vacuum degasser, binary pump, well-plate autosampler, thermostatted column compartment, the Agilent G6430 Triple Quadrupole Mass Spectrometer with an electrosprayionization (ESI) source. Quantitative analysis was carried out using MRM mode. The parameters for MRM transitions are

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention provides novel heterocyclic compounds, pharmaceutical acceptable salts and formulations thereof useful in preventing, treating or lessening the severity of a JAK-mediated disease. The invention also provides pharmaceutically acceptable compositions comprising such compounds and methods of using the compositions in the treatment of JAK-mediated disease.

Description

SUBSTITUTED HETEROARYL COMPOUNDS AND METHODS OF USE
CROSS- REFERENCE TO RELATED APPLICATION
[001] This application claims the benefit of U.S. Provisional Application Serial Number
61/904,460, filed on November 15, 2013, which is hereby incorporated by reference in its entirety.
FIELD OF THE INVENTION
[002] The present invention provides novel substituted heteroaryl compounds, and salts thereof, which are useful in the treatment of proliferative disease, autoimmune disease, allergic disease, inflammatory disease, transplantation rejection, and other diseases, in mammals. In particular, this invention relates to compounds that modulate the activity of Janus kinases, resulting in the modulation of inter- and/or intra-cellular signaling. This invention also relates to a method of using such compounds in the treatment of proliferative disease, autoimmune disease, allergic disease, inflammatory disease, transplantation rejection, and other diseases in mammals, especially humans, and to pharmaceutical compositions containing such compounds.
BACKGROUND OF THE INVENTION
[003] Cytokines are low-molecular weight polypeptides or glycoproteins that stimulate biological responses in virtually all cell types. Cytokines are critical for normal cell differentiation, proliferation, activation and immunoregulation but also contribute to growth of malignant cells and drive immune-mediated disease. A major subset of immunoregulatory cytokines, roughly 60, use the type I and type II cytokine receptors and pharmacological targeting of these cytokines/cytokines receptors is useful in treating immune-related diseases, skin disorders, myeloid proliferative disorders, cancer, and other diseases. (O'Sullivan et al, Mol. Immunol, 2007, 44:2497; Murray J., Immunol, 2007, 178:2623).
[004] Enabled by advances in molecular biology, it is now clear that an array of cytokines controls the growth and differentiation of hematopoietic cells and orchestrates all aspects of immune response. The family of cytokines that bind type I and type II cytokine receptors includes interleukins, interferons, and colony stimulating factor, as well as classic hormones such erythropoietin, prolactin and growth hormone. Binding of a cytokine to its cell surface receptor initiates intracellular signaling cascades that transduce the extracellular signal to the nucleus, ultimately leading to changes in gene expression. The pathway involves the Janus kinase family of protein tyrosine kinases (JAKs) and Signal Transducers and Activators of Transcription (STATs) ["Jakinibs: A New Class of Kinase Inhibitors in Cancer and Autoimmune Disease." Curr Opin Pharmacol. 2012 August; 12(4): 464-470]. [005] Janus kinase (JAK) is a family of intracellular, non-receptor tyrosine kinases that transduce cytokine -mediated signals via the JAK-STAT pathway. The JAK family plays a role in the cytokine-dependent regulation of proliferation and function of cells involved in immune response. Cytokines bind to their receptors, causing receptor dimerization, and this enables JAKs to phosphorylate each other as well as specific tyrosine motifs within the cytokine receptors. STATs that recognize these phosphotyrosine motifs are recruited to the receptor, and are then themselves activated by a JAK-dependent tyrosine phosphorylation event. Upon activation, STATs dissociate from the receptors, dimerize, and translocate to the nucleus to bind to specific DNA sites and alter transcription.
[006] Currently, there are four known mammalian JAK family members: JAKl (Janus kinase-1), JAK2 (Janus kinase-2), JAK3 (Janus kinase, leukocyte; JAKL; L-JAK and Janus kinase-3) and TYK2 (protein-tyrosine kinase 2). While JAKl, JAK2 and TYK2 are ubiquitously expressed, JAK3 is reported to be preferentially expressed in natural killer (NK) cells and not resting T cells ("Biology and significance of the JAK/STAT signaling pathways." Growth Factors, April 2012; 30(2): 88).
[007] JAKl is essential for signaling for certain type I and type II cytokines. It interacts with the common gamma chain (yc) of type I cytokine receptors to elicit signals from the IL-2 receptor family, the IL-4 receptor family, the gpl30 receptor family. It is also important for transducing a signal by type I (IFN-α/β) and type II (IFN-γ) interferons, and members of the IL-10 family via type II cytokine receptors. Genetic and biochemical studies have shown that JAKl is functionally and physically associated with the type I interferon (e g., IFNalpha), type II interferon (e.g., IFNgamma), IL-2 and IL-6 cytokine receptor complexes. Furthermore, characterization of tissues derived from JAKl knockout mice demonstrated critical roles for this kinase in the IFN, IL-IO, IL- 2/IL-4, and IL-6 pathways.
[008] Expression of JAKl in cancer cells enables individual cells to contract, potentially allowing them to escape their tumor and metastasize to other parts of the body. Elevated levels of cytokines which signal through JAKl have been implicated in a number of immune and inflammatory diseases. JAKl or JAK family kinase inhibitors may be useful for modulating or treating in such diseases. (Kisseleva et al, 2002, Gene 285: 1-24; Levy et al, 2005, Nat. Rev. Mol. Cell Biol. 3:651-662). A humanized monoclonal antibody targeting the IL-6 pathway (Tocilizumab) was approved by the European Commission for the treatment of moderate-to-severe rheumatoid arthritis (Scheinecker et al., 2009, Nat. Rev. Drug Discov. 8:273-274).
[009] JAK2 is implicated in signaling by members of the type II cytokine receptor family (e.g. interferon receptors), the GM-CSF receptor family, the gpl30 receptor family. JAK2 signaling is
2
SUBSTITUTE SHEET RULE 26 activated downstream from the prolactin receptor. Studies have identified a high prevalence of an acquired activating JAK2 mutation (JAK2V617F) in myleoproliferative disorders such as polycythemia vera, essential thrombocythemia and idiopathic myelofibrosis, etc. The mutant JAK2 protein is able to activate downstream signaling in the absence of cytokine stimulation, resulting in autonomous growth and/or hypersensitivity to cytokines and is believed to play a role in driving these diseases. Additional mutations or translocations resulting dysregulated JAK2 function have been described in other malignancies (Ihle J.N. and Gilliland D.G., Curr. Opin. Genet. Dev., 2007, 17:8; Sayyah J. and Sayeski P.P., Curr. Oncol. Rep., 2009, 11 : 117). Inhibitors of JAK2 have been described to be useful in myeloproliferative diseases (Santos et al, Blood, 2010, 115: 1131; Barosi G. and Rosti V., Curr. Opin. Hematol, 2009, 16: 129, Atallah E. and Versotvsek S., Exp. Rev. Anticancer Ther. 2009, 9:663).
[010] JAK3 associates exclusively with the gamma common cytokine receptor chain, which is present in the IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 cytokine receptor complexes. JA 3 is predominantly expressed in immune cells and transduces a signal in response to its activation via tyrosine phosphorylation by interleukin receptors. Since JAK3 expression is restricted mostly to hematopoietic cells, its role in cytokine signaling is thought to be more restricted than other JA s. Mutations of JAK3 result in severe combined immunodeficiency (SCID). (O'Shea et al, 2002, Cell, 109 (suppl): S121-S131). Based on its role in regulating lymphocytes, JA 3 and JAK3 -mediated pathways have been targeted for immunosuppressive indications (e.g., transplantation rejection and rheumatoid arthritis) (Baslund et al., 2005, Arthritis & Rheumatism 52:2686-2692; Changelian et al, 2003, Science 302: 875-878).
[011] TYK2 is implicated in IFN-a, IL-6, IL-10 and IL-12 signaling. Biochemical studies and gene-targeted mice uncovered the crucial role of TYK2 in immunity. Tyk2-deficient mice are viable and fertile but display multiple immunological defects, most prominently high sensitivity to infections and defective tumor surveillance. In contrast, inhibition of TYK2 results in increased resistance against allergic, autoimmune and inflammatory diseases. Particularly, targeting Tyk2 appears to be a promising strategy for the treatment of IL-12-, IL-23- or Type 1 IFN- mediated diseases. These include but are not limited to rheumatoid arthritis, multiple sclerosis, lupus, psoriasis, psoriatic arthritis, inflammatory bowel disease, uveitis, sarcoidosis, and tumors (Shaw, M. et al, Proc. Natl. Acad. Sci. USA, 2003, 100, 11594-11599; Ortmann, R.A., and Shevach, E.M. Clin. Immunol, 2001, 98, 109-118; Watford et al, Immunol. Rev., 2004, 202: 139). ["Janus Kinase (JAK) Inhibitors in Rheumatoid Arthritis." Current Rheumatology Reviews, 2011, 7, 306-312].
[012] A fully human monoclonal antibody targeting the shared p40 subunit of the IL-12 and 11- 23 cytokines (Ustekinumab) was recently approved by the European Commission for the treatment
3
SUBSTITUTE SHEET RULE 26 of moderate-to-severe plaque psoriasis (Krueger et al, 2007, N. Engl. J. Med. 356:580-92; Reich et al., 2009, Nat. Rev. Drug Discov. 8:355-356). In addition, an antibody targeting the IL-12 and IL-23 pathways underwent clinical trials for treating Crohn's Disease (Mannon et al., 2004, N.Engl. J. Med. 351 :2069-79).
[013] When dysregulated, JAK-mediated responses can positively or negatively affect cells leading to over-activation and malignancy or immune and hematopoietic deficiencies, respectively, and suggests the utility for use of inhibitors of JAK kinases. The JAK/STAT signaling pathway is involved in a variety of hyperproliferative and cancer-related processes including cell-cycle progression, apoptosis, angiogenesis, invasion, metastasis and evasion of the immune system (Haura et al., Nature Clinical Practice Oncology, 2005, 2(6), 315-324; Verna et al., Cancer and Metastasis Reviews, 2003, 22, 423-434). In addition, the JAK/STAT signaling pathway is important in the genesis and differentiation of hematopoietic cells and regulating both pro- and anti-inflammatory and immune responses (O' Sullivan et al., Molecular Immunology 2007, 44:2497).
[014] Therefore, the JAK STAT pathway, and in particular all four members of the JAK family, are believed to play a role in the pathogenesis of the asthmatic response, chronic obstructive pulmonary disease, bronchitis, and other related inflammatory diseases of the lower respiratory tract. The JAK/STAT pathway has also been implicated to play a role in inflammatory diseases/conditions of the eye including, but not limited to, iritis, uveitis, scleritis, conjunctivitis, as well as chronic allergic responses. Since cytokines utilize different patterns of JAK kinases (O'Sullivan et al, Mol. Immunol, 2007, 44:2497; Murray J., Immunol, 2007, 178:2623), there may be utility for antagonists of JAK kinases with differing intra-family selectivity profiles in diseases associated with particular cytokines or in diseases associated with mutations or polymorphisms in the JAK/STAT pathways.
[015] Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic joint inflammation. Patients with rheumatoid arthritis treated with JAK inhibitor showed that inhibition of JAK1 and JAK3 blocks signalling by multiple cytokines that are important for lymphocyte function, including interleukin-2 (IL-2), IL-4, IL-7, IL-9, IL-15 and IL-21. [Fleischmann, R. et al. "Placebo-controlled trial of tofacitinib monotherapy in rheumatoid arthritis." N. Engl. J. Med. 367, 495-507 (2012)]. It was conjectured that small-molecule inhibitors that directly inactivate specific JAK isoforms would also reduce not only the clinical symptoms of RA, but also suppress the upregulation of many of the proinflammatory cytokines that are critical in driving RA disease progression. ["Inhibitors of JAK for the treatment of rheumatoid arthritis: rationale and clinical data." Clin. Invest. (2012) 2(1), 39-47].
4
SUBSTITUTE SHEET RULE 26 [016] Persistent activation of STAT3 or STAT5 has been demonstrated in a wide spectrum of solid human tumors including breast, pancreatic, prostate, ovarian and hepatic carcinomas, as well as in the majority of hematopoietic tumors including lymphomas and leukemias. In this context, inactivation of JAK/STAT signaling in many hematopoietic tumors resulted in inhibition of cell proliferation and/or induction of apoptosis. Although STAT3 in tumor cells can be activated by various kinases, JAK2 has been shown to be the most important upstream activator mediating STAT3 activation in human tumor cell lines derived from various solid tumors [Mohamad Bassam Sonbol, Belal Firwana, Ahmad Zarzour, Mohammad Morad, Vishal Rana and Ramon V. Tiu "Comprehensive review of JAK inhibitors in myeloproliferative neoplasms." Therapeutic Advances in Hematology 2013, 4(1), 15-35; Hedvat M, Huszar D, Herrmann A, Gozgit J M, Schroeder A, Sheehy A, et al. "The JAK2 inhibitor AZD1480 potently blocks Stat3 signaling and oncogenesis in solid tumors." Cancer Cell 2009; 16(6):487-97.]. Therefore, inhibition of JAK kinases may have a beneficial role in the therapeutic treatment of these diseases.
[017] Clearly, JAK inhibitors have gathered attention as a new drug category for both immunosuppresion and antiinflammatory drug, and for cancer drug. Thus, new or improved agents which inhibit kinases such as Janus kinases are continually needed that act as immunosuppressive agents for organ transplants, as well as agents for the prevention and treatment of autoimmune diseases (e.g., multiple sclerosis, psoriasis, rheumatoid arthritis, asthma, type I diabetes, inflammatory bowel disease, Crohn's disease, autoimmune thyroid disorders, Alzheimer's disease), diseases involving a hyperactive inflammatory response (e.g., eczema), allergies, chronic obstructive pulmonary disease, bronchitis, cancer (e.g., prostate, leukemia, multiple myeloma), and some immune reactions (e.g., skin rash or contact dermatitis or diarrhea) caused by other therapeutics, to name a few. The compounds, compositions and methods described herein are directed toward these needs and other ends.
SUMMARY OF THE INVENTION
[018] The invention provides compounds that inhibit, regulate, and/or modulate one or more JAK kinase activities, and are useful for treating proliferative diseases, autoimmune diseases, allergic diseases, inflammatory diseases, transplantation rejections, and their co-morbidities. This invention also provides methods of making the compound, methods of using such compounds in the treatment of said diseases in mammals, especially in humans, and pharmaceutical compositions containing these compounds.
[019] Specifically, in one aspect, provided herein is a compound having Formula (I):
5
SUBSTITUTE SHEET RULE 26
Figure imgf000007_0001
or a stereoisomer, a tautomer, an N-oxide, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each of X, Z, Z1 and R7 is as defined herein.
[020] In one embodiment, Z is C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3, 4 or 5 R1 groups;
Z1 is H, C1-C12 alkyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z1 is optionally substituted with 1, 2, 3, 4 or 5 R2 groups;
X is H, -NRaRb, -N(Ra)C(=0)Rc, -N(Rc)C(=0)NRaRb, -N(Ra)C(=0)ORc or -N(Ra)S(=0)mRc;
R7 is H, NO2, N3, CN, C3-C12 cycloalkyl, -ORc, -C(=0)Rc, -OC(=0)Rc, -C(=0)ORc or - C(=0)NRaRb, with the proviso that when X is H, R7 is C3-C12 cycloalkyl, or when each of X and R7 is H, Z has formula (Z-34):
Figure imgf000007_0002
(Z-34) wherein R7 is optionally substituted with 1, 2, 3, 4 or 5 R6 groups; each R1 and R2 is independently H, F, N02, N3, CN, C1-C12 alkyl, C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C12 aryl, 5-12 membered heteroaryl, - (CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)mRc, -S(=0)2NRaRb, -C(=0)Rc, -OC(=0)Rc, - N(Ra)C(=0)Rc, -(CR4R5)„C(=0)ORc, -(CR4R5)„C(=0)NRaRb, -C(=NRc)NRaRb, N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, wherein each R1 and R2 is optionally independently substituted with 1, 2, 3, 4 or 5 R3 groups; each R3 is independently H, F, CI, Br, I, N02, N3, CN, C1-C12 alkyl, C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C12 aryl, 5-12 membered heteroaryl, - (CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)mRc, -S(=0)2NRaRb, -C(=0)Rc, -OC(=0)Rc, - N(Ra)C(=0)Rc, -(CR4R5)„C(=0)ORc, -(CR4R5)„C(=0)NRaRb, -C(=NRc)NRaRb, N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, or two adjacent R3 taken together with the atoms to which they are attached form a C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C12
6
SUBSTITUTE SHEET RULE 26 aryl or 5-12 membered heteroaryl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3, 4 or 5 R6 groups; each R4 and R5 is independently H, F, CI, Br, I, N3, CN, OH, NH2, C1-C12 alkyl, C1-C12 alkoxy, Ci-Ci2 alkylamino, C2-Ci2 alkenyl, C2-Ci2 alkynyl, C3-Ci2 cycloalkyl, 3-12 membered heterocyclyl, C6-Ci2 aryl or 5-12 membered heteroaryl, or R4 and R5 taken together with the carbon atom to which they are attached form a C3-Ci2 cycloalkyl or 3-12 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3, 4 or 5 R6 groups; each R6 is independently F, CI, Br, I, CN, N02, N , C1-C12 alkyl, C2-Ci2 alkenyl, C2-Ci2 alkynyl, C3-Ci2 cycloalkyl, 3-12 membered heterocyclyl, C6-Ci2 aryl, 5-12 membered heteroaryl, - NH2, -NH(Ci-Ci2 alkyl), -NH(CH2)„-(C -Ci2 cycloalkyl), -NH(CH2)„-(3-12 membered heterocyclyl), -NH(CH2)„-(C6-Ci2 aryl), -NH(CH2)„-(5-12 membered heteroaryl), -N(Ci-Ci2 alkyl)2, -N[(CH2)„-(C -Ci2 cycloalkyl)] 2, -N[(CH2)„-(3-12 membered heterocyclyl)]2, -N[(CH2)„- (C6-C12 aryl)]2, -N[(CH2)„-(5-12 membered heteroaryl)]2, OH, -0(Ci-Ci2 alkyl), -0(CH2)„-C -Ci2 cycloalkyl), -0(CH2)„-(3-12 membered heterocyclyl), -0(CH2)„-(C6-Ci2 aryl) or -0(CH2)„-(5-12 membered heteroaryl); each Ra, Rb and Rc is independently H, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, -(C1-C4 alkylene)-(C3-C6 cycloalkyl), 3-7 membered heterocyclyl, -(C1-C4 alkylene)- (3-7 membered heterocyclyl), C6-Cio aryl, -(C1-C4 alkylene)-(C6-Cio aryl), 5-10 membered heteroaryl or -(C1-C4 alkylene)-(5-10 membered heteroaryl), or Ra and Rb taken together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 substituents independently selected from F, CI, CN, N3, OH, NH2, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 alkoxy and Ci-C6 alkylamino;
each m is independently 0, 1 or 2; and each n is independently 0, 1, 2, 3 or 4.
[021] In another embodiment, R7 is H, N02, C -C6 cycloalkyl, -ORc, -C(=0)Rc, -OC(=0)Rc, -C(=0)ORc or -C(=0)NRaRb, with the proviso that when X is H, R7 is C -C6 cycloalkyl, or when each of X and R7 is H, Z has formula (Z-34):
Figure imgf000008_0001
(Z-34)
7
SUBSTITUTE SHEET RULE 26 wherein R7 is optionally substituted with 1 , 2 or 3 R6 groups.
[022] In one embodiment, provided herein is a compound having Formula (II):
Figure imgf000009_0001
wherein X is -NRaRb, -N(Ra)C(=0)Rc, -N(Rc)C(=0)NRaRb, -N(Ra)C(=0)ORc or -N(Ra)S(=0)mRc.
[023] In another embodiment, Z is C3-C6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3 or 4 R1 groups.
[024] In one embodiment, Z1 is H, C1-C4 alkyl, C3-C6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z1 is optionally substituted with 1, 2, 3 or 4 R2 groups.
[025] In another embodiment, each R1 and R2 is independently H, F, N3, CN, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, -(CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)mRc, -S(=0)2NRaRb, -C(=0)Rc, - N(Ra)C(=0)Rc, -(CR4R5)„C(=0)NRaRb, -N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, wherein each R1 and R2 is optionally independently substituted with 1, 2, 3 or 4 R3 groups.
[026] In one embodiment, each R3 is independently H, F, CI, CN, N3, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, - (CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)2NRaRb, -C(=0)Rc, -N(Ra)C(=0)Rc, (CR4R5)„C(=0)NRaRb, -N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, or two adjacent R3 taken together with the atoms to which they are attached form a C3-C6 cycloalkyl, 3-7 membered heterocyclyl, phenyl or 5-6 membered heteroaryl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 R6 groups.
[027] In another embodiment, each R4 and R5 is independently H, F, CI, Br, I, N3, CN, OH, NH2, Ci-C6 alkyl, Ci-C6 alkoxy, Ci-C6 alkylamino, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-7 membered heterocyclyl, phenyl or 5-6 membered heteroaryl, or R4 and R5 taken together with the carbon atom to which they are attached form a C3-C6 cycloalkyl or 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 R6 groups.
[028] In one embodiment, each R6 is independently F, CI, CN, N3, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl,
8
SUBSTITUTE SHEET RULE 26 NH2, -NH(Ci-C6 alkyl), -NH(CH2)n-(C3-C6 cycloalkyl), -NH(CH2)n-(3-7 membered heterocyclyl), -NH(CH2)n-phenyl, -NH(CH2)n-(5-6 membered heteroaryl), -N(Ci-C6 alkyl)2, -N[(CH2)n-(C3-C6 cycloalkyl)]2, -N[(CH2)n-(3-7 membered heterocyclyl)]2, -N[(CH2)n-phenyl]2, -N[(CH2)n-(5-6 membered heteroaryl)] 2, OH, -0(Ci-C6 alkyl), -0(CH2)n-(C3-C6 cycloalkyl), -0(CH2)n-(3-7 membered heterocyclyl), -0(CH2)n-phenyl or -0(CH2)n-(5-6 membered heteroaryl).
[029] In another embodiment, each Ra, Rb and Rc is independently H, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C6 cycloalkyl, -(C1-C2 alkylene)-(C3-C6 cycloalkyl), 3-7 membered heterocyclyl, -(C1-C2 alkylene)-(3-7 membered heterocyclyl), phenyl, -(C1-C2 alkylene)-phenyl, 5- 6 membered heteroaryl or -(C1-C2 alkylene)-(5-6 membered heteroaryl), or Ra and Rb taken together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2 or 3 substitutents independently selected from F, CI, CN, N3, OH, NH2, C1-C4 alkyl, C1-C4 haloalkyl, C1-C4 alkoxy and C1-C4 alkylamino.
[030] In one embodiment, Z is C4-C6 cycloalkyl, or 4-, 5- or 6-membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3 or 4 R1 groups.
[031] In another embodiment, Z is :
Figure imgf000010_0001
(Z-17) (Z-18) (Z-19) (Z-20) (Z-21 ) (Z-22) (Z-23) (Z-24)
Figure imgf000010_0002
(Z-25) (Z-26) (Z-27) (Z-28) (Z-29) (Z-30)
9
SUBSTITUTE SHEET RULE 26
Figure imgf000011_0001
(Z-33) or (Z-34)
or a stereoisomer thereof, wherein Z is optionally substituted by 1, 2 or 3 R1 groups.
[032] In one embodiment, Z1 is H, methyl, ethyl, propyl, isopropyl or cyclopropyl.
[033] In another embodiment, X is H, -NH2, -NHMe, -NHC(=0)Me, -NHC(=0)NHMe or - NHC(=0)NMe2.
[034] In one embodiment, each Ra, Rb and Rc is independently H, methyl, ethyl, propyl, isopropyl, cyclopropyl or butyl, wherein each Ra, Rb and Rc is optionally independently substituted with 1 , 2 or 3 substitutents independently selected from F, CI, CN, N3, OH, NH2, methyl, ethyl, -CF3, - OCH3 and -CH3NH2.
[035] In another aspect, provided herein is a pharmaceutical composition comprising the compound disclosed herein, and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof.
[036] In one embodiment, the pharmaceutical composition disclosed herein further comprising a therapeutic agent selected from the group consisting of chemotherapeutic agents, anti-proliferative agents, phosphodiesterase 4 (PDE4) inhibitors, p2-adrenoreceptor agonists, corticosteroids, nonsteroidal GR agonists, anticholinergic agents, antihistamines and combinations thereof.
[037] In another aspect, provided herein is a method of preventing, treating or lessening the severity of a JAK-mediated disease in a patient by administering to the patient with the compound disclosed herein or the pharmaceutical composition disclosed herein.
[038] In one embodiment, the JAK-mediated disease is a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease or a transplantation rejection.
[039] In another embodiment, the JAK-mediated disease is cancer, polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), chronic obstruction pulmonary disease (COPD), asthma, systemic lupus erythematosis, cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis, type I diabetes mellitus, allergic airway disease, sinusitis, eczema, hives, food allergies, allergies to insect venom, inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection or cell transplant rejection.
10
SUBSTITUTE SHEET RULE 26 [040] In another aspect, provided herein is the compound or the pharmaceutical composition disclosed herein for use in preventing, treating or lessening the severity of a JAK-mediated disease in a patient.
[041] In another aspect, provided herein is the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for treating cancer, polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), chronic obstruction pulmonary disease (COPD), asthma, systemic and cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis, type I diabetes mellitus, allergic airway disease, sinusitis, eczema, hives, food allergies, allergies to insect venom, inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection or cell transplant rejection.
[042] In another aspect, provided herein is a method of modulating the activity of the Janus kinase with the compound or the pharmaceutical composition disclosed herein.
[043] In still another aspect, provided herein is use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for modulating the activity of Janus kinase.
[044] In yet another aspect, provided herein are methods for preparation, separation and purification of the compounds represented by Formula (I).
[045] Biological test results indicate that the compounds provided herein can be used as preferable inhibitors of Janus kinases.
[046] Any embodiment disclosed herein can be combined with other embodiments as long as they are not contradictory to one another, even though the embodiments are described under different aspects of the invention. In addition, any technical feature in one embodiment can be applied to the corresponding technical feature in other embodiment as long as they are not contradictory to one another, even though the embodiments are described under different aspects of the invention.
[047] The foregoing merely summarizes certain aspects of the invention and is not intended to be limiting in nature. These aspects and other aspects and embodiments are described more fully below.
DETAILED DESCRIPTION OF THE INVENTION
DEFINITIONS AND GENERAL TERMINOLOGY
[048] Reference will now be made in detail to certain embodiments of the invention, examples of
11
SUBSTITUTE SHEET RULE 26 which are illustrated in the accompanying structures and formulas. The invention is intended to cover all alternatives, modifications, and equivalents which may be included within the scope of the present invention as defined by the claims. One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in the practice of the present invention. The present invention is in no way limited to the methods and materials described herein. In the event that one or more of the incorporated literature, patents, and similar materials differs from or contradicts this application, including but not limited to defined terms, term usage, described techniques, or the like, this application controls.
[049] It is further appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment. Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.
[050] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one skilled in the art to which this invention belongs. All patents and publications referred to herein are incorporated by reference in their entirety.
[051] As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, and the Handbook of Chemistry and Physics, 75th Ed. 1994. Additionally, general principles of organic chemistry are described in Sorrell et al., "Organic Chemistry", University Science Books, Sausalito: 1999, and Smith et al, "March's Advanced Organic Chemistry", John Wiley & Sons, New York: 2007, all of which are incorporated by reference in their entireties.
[052] The grammatical articles "a", "an" and "the", as used herein, are intended to include "at least one" or "one or more" unless otherwise indicated herein or clearly contradicted by the context. Thus, the articles are used herein to refer to one or more than one (i.e. at least one) of the grammatical objects of the article. By way of example, "a component" means one or more components, and thus, possibly, more than one component is contemplated and may be employed or used in an implementation of the described embodiments.
[053] As used herein, the term "subject" refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human.
12
SUBSTITUTE SHEET RULE 26 [054] As used herein, "patient" refers to a human (including adults and children) or other animal. In one embodiment, "patient" refers to a human.
[055] The term "comprising" is meant to be open ended, including the indicated component but not excluding other elements.
[056] "Stereoisomers" refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space. Stereoisomers include enantiomer, diastereomers, conformer (rotamer), geometric (cis/trans) isomer, atropisomer, etc.
[057] "Chiral" refers to molecules which have the property of non-superimposability of the mirror image partner, while the term "achiral" refers to molecules which are superimposable on their mirror image partner.
[058] "Enantiomers" refer to two stereoisomers of a compound which are non-superimposable mirror images of one another.
[059] "Diastereomer" refers to a stereoisomer with two or more centers of chirality and whose molecules are not mirror images of one another. Diastereomers have different physical properties, e.g. melting points, boiling points, spectral properties or biological activities. Mixture of diastereomers may separate under high resolution analytical procedures such as electrophoresis and chromatography such as HPLC.
[060] Stereochemical definitions and conventions used herein generally follow Parker et al., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, New York and Eliel et al, "Stereochemistry of Organic Compounds", John Wiley & Sons, Inc., New York, 1994.
[061] Many organic compounds exist in optically active forms, i.e., they have the ability to rotate the plane of plane-polarized light. In describing an optically active compound, the prefixes D and L, or R and S, are used to denote the absolute configuration of the molecule about its chiral center(s). The prefixes d and / or (+) and (-) are employed to designate the sign of rotation of plane-polarized light by the compound, with (-) or / meaning that the compound is levorotatory. A compound prefixed with (+) or d is dextrorotatory. A specific stereoisomer may be referred to as an enantiomer, and a mixture of such stereoisomers is called an enantiomeric mixture. A 50:50 mixture of enantiomers is referred to as a racemic mixture or a racemate, which may occur where there has been no stereoselection or stereospecificity in a chemical reaction or process.
[062] Any asymmetric atom (e.g., carbon or the like) of the compound(s) disclosed herein can be present in racemic or enantiomerically enriched, for example the (R)-, (S)- or (R,S)- configuration. In certain embodiments, each asymmetric atom has at least 50 % enantiomeric excess, at least 60
13
SUBSTITUTE SHEET RULE 26 % enantiomeric excess, at least 70 % enantiomeric excess, at least 80 % enantiomeric excess, at least 90 % enantiomeric excess, at least 95 % enantiomeric excess, or at least 99 % enantiomeric excess in the (R)- or (S)- configuration.
[063] Depending on the choice of the starting materials and procedures, the compounds can be present in the form of one of the possible stereoisomers or as mixtures thereof, such as racemates and diastereoisomer mixtures, depending on the number of asymmetric carbon atoms. Optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. If the compound contains a double bond, the substituent may be E or Z configuration. If the compound contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis- or trans-configuration.
[064] Any resulting mixtures of stereoisomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric isomers, enantiomers, diastereomers, for example, by chromatography and/or fractional crystallization.
[065] Any resulting racemates of final products or intermediates can be resolved into the optical antipodes by methods known to those skilled in the art, e.g., by separation of the diastereomeric salts thereof. Racemic products can also be resolved by chiral chromatography, e.g., high performance liquid chromatography (HPLC) using a chiral adsorbent. Preferred enantiomers can also be prepared by asymmetric syntheses. See, for example, Jacques, et al, Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Principles of Asymmetric Synthesis (2nd Ed. Robert et al, Elsevier, Oxford, UK, 2012); Eliel et al, Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); and Wilen et al, Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN, 1972). Chiral Separation Techniques: A Practical Approach (Subramanian, G. Ed., Wiley- VCH Verlag GmbH & Co. KGaA, Weinheim, Germany, 2007).
[066] The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. Where tautomerization is possible (e.g. in solution), a chemical equilibrium of tautomers can be reached. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerizations. Valence tautomers include interconversions by reorganization of some of the bonding electrons. A specific example of keto-enol tautomerization is the interconversion of pentane-2,4-dione and 4-hydroxypent-3-en-2-one tautomers. Another example of tautomerization is phenol-keto tautomerization. A specific example of phenol-keto tautomerization is the interconversion of pyridin-4-ol and pyridin-4(lH)-one tautomers. Unless
14
SUBSTITUTE SHEET RULE 26 otherwise stated, all tautomeric forms of the compounds disclosed herein are within the scope of the invention.
[067] As described herein, compounds disclosed herein may optionally be substituted with one or more substituents, such as those illustrated below, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase "optionally substituted" is used interchangeably with the phrase "substituted or unsubstituted". In general, the term "substituted" refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent. Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group. When more than one position in a given structure can be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position.
[068] At various places in the present specification, substituents of compounds disclosed herein are disclosed in groups or in ranges. It is specifically intended that the invention include each and every individual subcombination of the members of such groups and ranges. For example, the term "C1-C6 alkyl" is specifically intended to individually disclose methyl, ethyl, C3 alkyl, C4 alkyl, C5 alkyl, and C6 alkyl; the term "4-7 membered heterocyclyl" is specifically intended to individually disclose 4- membered heterocyclyl, 5- membered heterocyclyl, 6- membered heterocyclyl and 7- membered heterocyclyl.
[069] At various places in the present specification, linking substituents are described. Where the structure clearly requires a linking group, the Markush variables listed for that group are understood to be linking groups. For example, if the structure requires a linking group and the Markush group definition for that variable lists "alkyl" or "aryl" then it is understood that the "alkyl" or "aryl" represents a linking alkylene group or arylene group, respectively.
[070] The term "alkyl" or "alkyl group" refers to a saturated linear or branched-chain monovalent hydrocarbon radical of 1 to 20 carbon atoms, wherein the alkyl radical may be optionally substituted independently with one or more substituents described below. Unless otherwise specified, the alkyl group contains 1-20 carbon atoms. In one embodiment, the alkyl group contains 1-12 carbon atoms. In another embodiment, the alkyl group contains 1-6 carbon atoms. In still another embodiment, the alkyl group contains 1-4 carbon atoms. In yet another embodiment, the alkyl group contains 1-3 carbon atoms. The alkyl radical may be optionally substituted independently with one or more substituents described herein. Some non-limiting examples of the alkyl group include methyl (Me, -CH3), ethyl (Et, -CH2CH3), 1 -propyl (n-Pr, n- propyl, -CH2CH2CH3), 2-propyl (z'-Pr, /-propyl, -CH(CH3)2), 1 -butyl (zi-Bu, n-butyl, - CH2CH2CH2CH3), 2-methyl-l-propyl (/-Bu, /-butyl, -CH2CH(CH3)2), 2-butyl (s-Bu, s-butyl, -
15
SUBSTITUTE SHEET RULE 26 CH(CH3)CH2CH3), 2-methyl-2-propyl (t-Bu, t-butyl, -C(CH3)3), 1-pentyl (n-pentyl, - CH2CH2CH2CH2CH3), 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (-CH(CH2CH3)2), 2-methyl-2- butyl (-C(CH3)2CH2CH3), 3-methyl-2-butyl (-CH(CH3)CH(CH3)2), 3-methyl-l-butyl (- CH2CH2CH(CH3)2), 2-methyl-l-butyl (-CH2CH(CH3)CH2CH3), 1-hexyl (-
CH2CH2CH2CH2CH2CH3), 2-hexyl (-CH(CH3)CH2CH2CH2CH3), 3-hexyl (- CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (-C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (- CH(CH3)CH(CH3)CH2CH3), 4-methyl-2-pentyl (-CH(CH3)CH2CH(CH3)2), 3-methyl-3-pentyl (- C(CH3)(CH2CH3)2), 2-methyl-3-pentyl (-CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (- C(CH3)2CH(CH3)2), 3,3-dimethyl-2-butyl (-CH(CH3)C(CH3)3, 1-heptyl, 1-octyl, and the like.
[071] The term "alkylene" refers to a saturated divalent hydrocarbon group derived from a straight or branched chain saturated hydrocarbon by the removal of two hydrogen atoms. Unless otherwise specified, the alkylene group contains 1-12 carbon atoms. In one embodiment, the alkylene group contains 1-6 carbon atoms. In another embodiment, the alkylene group contains 1-4 carbon atoms. In still another embodiment, the alkylene group contains 1-3 carbon atoms. In yet another embodiment, the alkylene group contains 1-2 carbon atoms. The alkylene group is exemplified by methylene (-CH2-), ethylidene (-CH2CH2-), isopropylidene (-CH(CH3)CH2-), and the like.
[072] The term "alkenyl" refers to a linear or branched-chain monovalent hydrocarbon radical of 2 to 12 carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp2 double bond, wherein the alkenyl radical may be optionally substituted independently with one or more substituents described herein, and includes radicals having "cz's" and "trans" orientations, or alternatively, "E" and "Z" orientations. In one embodiment, the alkenyl group contains 2-8 carbon atoms. In another embodiment, the alkenyl group contains 2-6 carbon atoms. In still another embodiment, the alkenyl group contains 2-4 carbon atoms. Some non-limiting examples of the alkenyl group include ethylenyl or vinyl (-CH=CH2), allyl (-CH2CH=CH2), and the like.
[073] The term "alkynyl" refers to a linear or branched-chain monovalent hydrocarbon radical of 2 to 12 carbon atoms with at least one site of unsaturation, i.e., a carbon-carbon, sp triple bond, wherein the alkynyl radical may be optionally substituted independently with one or more substituents described herein. In one embodiment, the alkynyl group contains 2-8 carbon atoms. In another embodiment, the alkynyl group contains 2-6 carbon atoms. In still another embodiment, the alkynyl group contains 2-4 carbon atoms. Some non-limiting examples of the alkynyl group include ethynyl (-C≡CH), propynyl (propargyl, -CH2C≡CH), -C≡C-CH3, and the like.
[074] The term "alkoxy" refers to an alkyl group, as previously defined, attached to the principal carbon atom through an oxygen atom. Unless otherwise specified, the alkoxy group contains 1-12
16
SUBSTITUTE SHEET RULE 26 carbon atoms. In one embodiment, the alkoxy group contains 1-6 carbon atoms. In another embodiment, the alkoxy group contains 1-4 carbon atoms. In still another embodiment, the alkoxy group contains 1-3 carbon atoms. The alkoxy radical may be optionally substituted independently with one or more substituents described herein. Some non-limiting examples of alkoxy groups include methoxy (MeO, -OCH3), ethoxy (EtO, -OCH2CH3), 1-propoxy (n-PrO, n-propoxy, - OCH2CH2CH3), 2-propoxy (z'-PrO, z'-propoxy, -OCH(CH3)2), 1-butoxy (n-BuO, n-butoxy, - OCH2CH2CH2CH3), 2-methyl-l-propoxy (z-BuO, z-butoxy, -OCH2CH(CH3)2), 2-butoxy (s-BuO, s- butoxy, -OCH(CH3)CH2CH3), 2-methyl-2-propoxy (t-BuO, t-butoxy, -OC(CH3)3), 1-pentoxy (n- pentoxy, -OCH2CH2CH2CH2CH3), 2-pentoxy (-OCH(CH3)CH2CH2CH3), 3-pentoxy (- OCH(CH2CH3)2), 2-methyl-2-butoxy (-OC(CH3)2CH2CH3), 3-methyl-2-butoxy (- OCH(CH3)CH(CH3)2), 3-methyl-l-butoxy (-OCH2CH2CH(CH3)2), 2-methyl-l-butoxy (- OCH2CH(CH3)CH2CH3), and the like.
[075] The term "haloalkyl", "haloalkenyl" or "haloalkoxy" refers to alkyl, alkenyl, or alkoxy, as the case may be, substituted with one or more halogen atoms. Some non-limiting examples of haloalkyl and haloalkoxy are include trifluoromethyl (-CF3), trifluoromethoxy (-OCF3) and the like.
[076] The term "carbocycle", "carbocyclyl" or "carbocyclic ring" refers to a monovalent or multivalent non-aromatic, saturated or partially unsaturated ring having 3 to 12 carbon atoms as a monocyclic, bicyclic or tricyclic ring system. The carbobicyclyl system includes spiro carbobicyclyl and fused carbobicyclyl. Some non-limiting examples of carbocyclyl groups include cycloalkyl, cycloalkenyl, and cycloalkynyl. Further non-limiting examples of carbocyclyl group include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-l-enyl, l-cyclopent-2-enyl, 1-cyclopent- 3-enyl, cyclohexyl, 1-cyclohex-l-enyl, l-cyclohex-2-enyl, l-cyclohex-3-enyl, cyclohexadienyl, and the like.
[077] The term "cycloalkyl" refers to a monovalent or multivalent saturated ring having 3 to 12 carbon atoms as a monocyclic, bicyclic, or tricyclic ring system. In one embodiment, the cycloalkyl contains 3-12 carbon atoms. In another embodiment, the cycloalkyl contains 3-8 carbon atoms. In still another embodiment, the cycloalkyl contains 4-6 carbon atoms. In yet another embodiment, the cycloalkyl contains 3-6 carbon atoms. The cycloalkyl radical may be optionally substituted independently with one or more substituents described herein.
[078] The term "heterocycle", "heterocyclyl", or "heterocyclic ring" as used interchangeably herein refers to a amonovalent or multivalent, saturated or partially unsaturated, non-aromatic monocyclic, bicyclic or tricyclic ring containing 3-12 ring atoms of which at least one ring atom is selected from nitrogen, sulfur and oxygen, and which may, unless otherwise specified, be carbon
17
SUBSTITUTE SHEET RULE 26 or nitrogen linked, and of which a -CH2- group can optionally be replaced by a -C(=0)- group. Ring sulfur atoms may be optionally oxidized to form S-oxides. Ring nitrogen atoms maybe optionally oxidized to form N-oxides. The heterocyclyl contains saturated heterocyclyl and partially unsaturated heterocyclyl. The heterocyclyl has one or more points of attachment to the rest of the molecule.
[079] Some non-limiting examples of heterocyclyl include oxiranyl, azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1,3- dioxolanyl, dithiolanyl, tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, thioxanyl, dithianyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl, diazepinyl, thiazepinyl, indolinyl, 1,2,3,4-tetrahydroisoquinolinyl, 1,3-benzodioxolyl, 2-oxa-5- azabicyclo[2.2.1]hept-5-yl, isoxazolidinyl, isothiazolidinyl, 1 ,2-oxazinanyl, 1 ,2-thiazinanyl, hexahydropyridazinyl, oxazepinyl (1,4-oxazepinyl, 1 ,2-oxazepinyl), diazepinyl (1,4-diazepinyl, 1 ,2-diazepinyl), dioxpinyl (1,4-dioxpinyl, 1,2-dioxpinyl), thiazepinyl (1,4-thiazepinyl, 1,2- thiazepinyl), and the like. Some non-limiting examples of heterocyclyl wherein -CH2- group is replaced by -C(=0)- moiety are 2-oxopyrrolidinyl, oxo-l,3-thiazolidinyl, 2-piperidinonyl, 3,5- dioxopiperidinyl, and the like. Some non-limiting examples of heterocyclyl wherein the ring sulfur atom is oxidized are sulfolanyl, 1,1-dioxo-thiomorpholinyl, and the like. The heterocyclyl group may be optionally substituted with one or more substituents described herein.
[080] In one embodiment, heterocyclyl may be 4-7 membered heterocyclyl, which refers to a monovalent or multivalent, saturated or partially unsaturated, non-aromatic monocyclic ring containing 4-7 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen, and of which may, unless otherwise specified, be carbon or nitrogen linked, and of which a -CH2- group can optionally be replaced by a -C(=0)- group. Ring sulfur atoms may be optionally oxidized to form S-oxides. Ring nitrogen atoms maybe optionally oxidized to form N-oxides. The 4-7 membered heterocyclyl contains 4-7 membered saturated heterocyclyl and partially unsaturated heterocyclyl. The 4-7 membered heterocyclyl has one or more points of attachment to the rest of the molecule. Some non-limiting examples of 4-7 membered heterocyclyl include azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, 1-pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, pyrazolidinyl, dihydropyrazolyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1,3-dioxolanyl, dithiolanyl, isoxazolidinyl, isothiazolidinyl, 1 ,2- oxazinanyl, 1 ,2-thiazinanyl, hexahydropyridazinyl, tetrahydropyranyl, dihydropyranyl, 2H- pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, dihydropyridyl, tetrahydropyridyl, piperidinyl,
18
SUBSTITUTE SHEET RULE 26 morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, thioxanyl, dithianyl, homopiperazinyl, homopiperidinyl, oxepanyl, thiepanyl, oxazepinyl (1,4-oxazepinyl, 1 ,2-oxazepinyl), diazepinyl (1,4-diazepinyl, 1 ,2-diazepinyl), thiazepinyl (1,4-thiazepinyl, 1 ,2-thiazepinyl), dioxpinyl (1,4- dioxpinyl, 1,2-dioxpinyl), and the like. Some non- limiting examples of heterocyclyl wherein -CH2- group is replaced by -C(=0)- moiety are 2-oxopyrrolidinyl, oxo-l,3-thiazolidinyl, 2-piperidinonyl, 3,5-dioxopiperidinyl, and the like. Some non-limiting examples of heterocyclyl wherein the ring sulfur atom is oxidized are sulfolanyl, 1,1-dioxotetrahydrothiophenyl, l,l-dioxotetrahydro-2H- thiopyranyl, 1,1-dioxothiomorpholinyl, and the like. The 4-7 membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
[081] In another embodiment, heterocyclyl may be a 4-membered heterocyclyl, which refers to a monovalent or multivalent, saturated or partially unsaturated, non-aromatic monocyclic ring containing 4 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen, and which may, unless otherwise specified, be carbon or nitrogen linked, and of which a - CH2- group can optionally be replaced by a -C(=0)- group. Ring sulfur atoms may be optionally oxidized to form S-oxides. Ring nitrogen atoms maybe optionally oxidized to form N-oxides. The
4- membered heterocyclyl contains 4-membered saturated heterocyclyl and partially unsaturated heterocyclyl. Some non-limiting examples of 4-membered heterocyclyl include azetidinyl, oxetanyl, thietanyl, and the like. The 4-membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
[082] In another embodiment, heterocyclyl may be a 5-membered heterocyclyl, which refers to a monovalent or multivalent, saturated or partially unsaturated, non-aromatic monocyclic ring containing 5 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen, and which may, unless otherwise specified, be carbon or nitrogen linked, and of which a - CH2- group can optionally be replaced by a -C(=0)- group. Ring sulfur atoms may be optionally oxidized to form S-oxides. Ring nitrogen atoms maybe optionally oxidized to form N-oxides. The
5- membered heterocyclyl contains 5-membered saturated heterocyclyl and partially unsaturated heterocyclyl. Some non-limiting examples of 5-membered heterocyclyl include pyrrolidinyl, 1- pyrrolinyl, 2-pyrrolinyl, 3-pyrrolinyl, pyrazolinyl, dihydropyrazolyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothienyl, dihydrothienyl, 1,3- dioxolanyl, dithiolanyl, isoxazolidinyl, isothiazolidinyl, and the like. Some non-limiting examples of heterocyclyl wherein -CH2- group is replaced by -C(=0)- moiety are 2-oxopyrrolidinyl, oxo- 1,3-thiazolidinyl, and the like. A non-limiting examples of heterocyclyl wherein the ring sulfur atom is oxidized is 1,1-dioxotetrahydrothiophenyl. The 5-membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
19
SUBSTITUTE SHEET RULE 26 [083] In still another embodiment, heterocyclyl may be a 6-membered heterocyclyl, which refers to a monovalent or multivalent, saturated or partially unsaturated, non-aromatic monocyclic ring containing 6 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen, and which may, unless otherwise specified, be carbon or nitrogen linked, and of which a - CH2- group can optionally be replaced by a -C(=0)- group. Ring sulfur atoms may be optionally oxidized to form S-oxides. Ring nitrogen atoms maybe optionally oxidized to form N-oxides. The 6-membered heterocyclyl contains 6-membered saturated heterocyclyl and partially unsaturated heterocyclyl. Some non-limiting examples of 6-membered heterocyclyl include tetrahydropyranyl, dihydropyranyl, 2H-pyranyl, 4H-pyranyl, tetrahydrothiopyranyl, piperidinyl, dihydropyridyl, tetrahydropyridyl, morpholinyl, thiomorpholinyl, piperazinyl, dioxanyl, thioxanyl, dithianyl, and the like. Some non-limiting examples of heterocyclyl wherein -CH2- group is replaced by -C(=0)- moiety include 2-piperidinonyl, 3,5-dioxopiperidinyl, 1 ,2-oxazinanyl, 1 ,2-thiazinanyl, hexahydropyridazinyl, and the like. Some non-limiting examples of heterocyclyl wherein the ring sulfur atom is oxidized are 1,1-dioxothiomorpholinyl, l,l-dioxotetrahydro-2H-thiopyranyl, and the like. The 6-membered heterocyclyl group is optionally substituted with one or more substituents described herein.
[084] In one embodiment, heterocyclyl may be 3-7 membered heterocyclyl, which refers to a monovalent or multivalent, saturated or partially unsaturated, non-aromatic monocyclic ring containing 3-7 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen, and of which may, unless otherwise specified, be carbon or nitrogen linked, and of which a -CH2- group can optionally be replaced by a -C(=0)- group. Ring sulfur atoms may be optionally oxidized to form S-oxides. Ring nitrogen atoms maybe optionally oxidized to form N-oxides. The 3-7 membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
[085] In yet another embodiment, heterocyclyl refers to a 7-12 membered heterocyclyl, which refers to a monovalent or multivalent, saturated or partially unsaturated, non-aromatic spiro or fused bicyclyl ring containing 7-12 ring atoms, of which at least one ring atom is selected from nitrogen, sulfur and oxygen, and which may, unless otherwise specified, be carbon or nitrogen linked, and of which a -CH2- group can optionally be replaced by a -C(=0)- group. Ring sulfur atoms may be optionally oxidized to form S-oxides. Ring nitrogen atoms maybe optionally oxidized to form N-oxides. Some non-limiting examples of 7-12 membered heterocyclyl include indolinyl, 1,2,3,4-tetrahydroisoquinolinyl, 1,3-benzodioxolyl, 2-oxa-5-azabicyclo[2.2.1]hept-5-yl, and the like. The 7-12 membered heterocyclyl group may be optionally substituted with one or more substituents described herein.
20
SUBSTITUTE SHEET RULE 26 [086] The terms "fused bicyclic ring", "fused cyclic", "fused bicyclyl" and "fused cyclyl" are used interchangeably refer to a monovalent or multivalent saturated or partially unsaturated bridged ring system, which refers to a bicyclic ring system that is not aromatic. Such a system may contain isolated or conjugated unsaturation, but not aromatic or heteroaromatic rings in its core structure (but may have aromatic substitution thereon).
[087] The terms "spirocyclyl", "spirocyclic", "spiro bicyclyl" and "spiro bicyclic" are used interchangeably and refer to a monovalent or multivalent, saturated or partially unsaturated ring system wherein a ring originating from a particular annular carbon of another ring. For example, as depicted below in Structure a, a saturated bridged ring system (ring B and B') is termed as "fused bicyclyl", whereas ring A and ring B share an atom between the two saturated ring system, which terms as a "spirocyclyl" or "spiro bicyclyl". Each ring in the fused bicyclyl or the spiro bicyclyl can be either a carbocyclyl or a heterocyclyl, and each ring is optionally substituted independently with one or more substituents described herein.
Figure imgf000022_0001
Structure a
[088] The term "heterocycloalkyl" refers to a monovalent or multivalent saturated ring having 3 to 12 ring atoms as a monocyclic, bicyclic, or tricyclic ring system in which at least one ring atom is selected from nitrogen, sulfur and oxygen.
[089] The term "n membered" where n is an integer typically describes the number of ring- forming atoms in a moiety where the number of ring-forming atoms is n. For example, piperidinyl is an example of a 6 membered heterocycloalkyl and 1,2,3,4-tetrahydro-naphthalenyl is an example of a 10 membered carbocyclyl group.
[090] The term "unsaturated" refers to a moiety having one or more units of unsaturation.
[091] The term "heteroatom" refers to one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, including any oxidized form of nitrogen, sulfur, or phosphorus; the quaternized form of any basic nitrogen; or a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4- dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR (as in N-substituted pyrrolidinyl).
[092] The term "halogen" refers to Fluoro (F), Chloro (CI), Bromo (Br), or Iodo (I).
[093] The term "azido" or "N3" refers to an azide moiety. This radical may be attached, for
21
SUBSTITUTE SHEET RULE 26 example, to a methyl group to form azidomethane (methyl azide, MeN3); or attached to a phenyl group to form phenyl azide (PhN3).
[094] The term "aryl" refers to monocyclic, bicyclic, and tricyclic carbocyclic ring systems having a total of 6 to 14 ring members, preferably, 6 to 12 ring members, and more preferably 6 to 10 ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3 to 7 ring members and that has one or more points of attachment to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aryl ring" or "aromatic ring". Some non-limiting examples of the aryl group would include phenyl, naphthyl, and anthracenyl. The aryl radical is optionally substituted independently with one or more substituents described herein.
[095] The term "heteroaryl" or "heteroaromatic ring" refers to monocyclic, bicyclic, and tricyclic ring systems having a total of 5 to 12 ring members, preferably, 5 to 10 ring members, and more preferably 5 to 6 ring members, wherein at least one ring in the system is aromatic, at least one aromatic ring in the system contains one or more heteroatoms, wherein each ring in the system contains 5 to 7 ring members and that has one or more points of attachment to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the term "heteroaryl ring" or the term "heteroaromatic ring". In one embodiment, heteroaryl refers to a 5-12 membered heteroaryl comprises 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In another embodiment, heteroaryl refers to a 5-10 membered heteroaryl comprises 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. In another embodiment, heteroaryl refers to a 5-6 membered heteroaryl comprises 1, 2, 3 or 4 heteroatoms independently selected from O, S and N. The heteroaryl radicals are optionally substituted independently with one or more substituents described herein.
[096] Some non-limiting examples of the 5-6 membered heteroaryl group include 2-furanyl, 3- furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5- isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3- pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyrimidonyl, pyrimidinedionyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5- tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, pyrazolyl (e.g., 2-pyrazolyl), pyrazolonyl, isothiazolyl, 1,2,3-oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3-triazolyl, 1,2,3-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, pyrazinyl, 1,3,5-triazinyl, and the like. Some non-limiting examples of the bicycles heteroaryl group include benzimidazolyl, benzofuryl, benzothiophenyl, indolyl (e.g., 2-indolyl), purinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4- quinolinyl), and isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl or 4-isoquinolinyl),
22
SUBSTITUTE SHEET RULE 26 imidazo[l,2-a]pyridyl, pyrazolo[l,5-a]pyridyl, pyrazolo[3,4-£]pyridyl, pyrazolo[l,5-a]pyrimidyl, imidazo[ 1 ,2-£]pyridazinyl, [ 1 ,2,4]triazolo[4,3-£]pyridazinyl, [ 1 ,2,4]triazolo[ 1 ,5-a]pyrimidinyl and [l,2,4]triazolo[l,5-a]pyridyl, and the like.
[097] The term "azolyl" refers to a 5- or a 9-membered heteroaryl ring system containing at least one nitrogen atom. Some non-limiting examples of 5-membered azolyl rings include pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, diazolyl, triazolyl, and the like. Some non-limiting examples of 9-membered azolyl rings include indazolyl, pyrazolopyridinyl, lH-benzo[d]imidazolyl, and the like.
[098] The term "carboxy" or "carboxyl", whether used alone or with other terms, such as "carboxyalkyl", refers to -CO2H. The term "carbonyl", whether used alone or with other terms, such as "aminocarbonyl", denotes -(C=0)-.
[099] The term "alkylamino" embraces "N-alkylamino" and "N,N-dialkylamino" where amino groups are independently substituted with one alkyl radical or with two alkyl radicals, respectively. In one embodiment alkylamino are "lower alkylamino" radicals having one or two alkyl radicals of one to six carbon atoms, attached to a nitrogen atom. In another embodiment alkylamino are alkylamino radicals having one or two alkyl radicals of one to four carbon atoms, attached to a nitrogen atom. Some non-limiting examples of alkylamino include N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino, and the like.
[100] The term "aminoalkyl" refers to linear or branched alkyl radicals having one to about ten carbon atoms any one of which may be substituted with one or more amino radicals. More preferred aminoalkyl radicals are "lower aminoalkyl" radicals having 1-6 carbon atoms and one or more amino radicals. Examples of such radicals include aminomethyl, aminoethyl, aminopropyl, aminobutyl and aminohexyl.
[101] As described herein, a bond drawn from a substituent to the center of one ring within a ring system (as shown in the below Structures) represents substitution of the substituent at any substitutable position on the ring system. For example, as depicted below, Structure b represents possible substitution in any of the positions on the ring C and ring D shown in Structure c ~ Structure g.
Figure imgf000024_0001
Structure b Structure C Structure d Structure e Structure f Structure g
[102] As described herein, a connecting bond drawn from the center of one ring within a ring
23
SUBSTITUTE SHEET RULE 26 system (as shown in Structure h) represents connection of the connecting bond attached to the rest of the molecule at any substitutable position on the ring system. For example, Structure h represents possible connection attached to the rest of the molecule in any of the position on ring E.
Figure imgf000025_0001
Structure h
[103] The term "protecting group" or "PG" refers to a substituent that is commonly employed to block or protect a particular functionality while reacting other functional groups on the compound. For example, an "amino-protecting group" is a substituent attached to an amino group that blocks or protects the amino functionality in the compound. Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxy-carbonyl (BOC, Boc), benzyloxycarbonyl (CBZ, Cbz) and 9- fluorenylmethylenoxy-carbonyl (Fmoc). Similarly, a "hydroxy-protecting group" refers to a substituent of a hydroxy group that blocks or protects the hydroxy functionality. Suitable protecting groups include acetyl and silyl. A "carboxy-protecting group" refers to a substituent of the carboxy group that blocks or protects the carboxy functionality. Common carboxy-protecting groups include -CH2CH2SO2PI1, cyanoethyl, 2-(trimethylsilyl)ethyl, 2-(trimethylsilyl)ethoxy- methyl, 2-(p-toluenesulfonyl)-ethyl, 2-(/?-nitrophenylsulfenyl)-ethyl, 2-(diphenylphosphino)-ethyl, nitroethyl and the like. For a general description of protecting groups and their use, see T. W. Greene, Protective Groups in Organic Synthesis, John Wiley & Sons, New York, 1991; and P. J. Kocienski, Protecting Groups, Thieme, Stuttgart, 2005.
[104] The term "prodrug" as used herein, represents a compound that is transformed in vivo into a compound of Formula (I). Such a transformation can be affected, for example, by hydrolysis in blood or enzymatic transformation of the prodrug form to the parent form in blood or tissue. Prodrugs of the compounds disclosed herein may be, for example, esters. Esters that may be utilized as prodrugs in the present invention are phenyl esters, aliphatic (C1-C24) esters, acyloxymethyl esters, carbonates, carbamates, and amino acid esters. For example, a compound disclosed herein that contains an OH group may be acylated at this position in its prodrug form. Other prodrug forms include phosphates, such as, for example those phosphates resulting from the phosphonation of an OH group on the parent compound. A thorough discussion of prodrugs is provided in Higuchi et al, Pro-drugs as Novel Delivery Systems, Vol. 14, A.C.S. Symposium Series; Roche et al., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987; Rautio et al, Prodrugs: Design and Clinical Applications, Nat. Rev. Drug Discovery, 2008, 7, 255-270, and Hecker et al, Prodrugs of Phosphates and Phosphonates, J.
24
SUBSTITUTE SHEET RULE 26 Med. Chem., 2008, 51 , 2328-2345, all of which are incorporated herein by reference.
[105] A "metabolite" refers to a product produced through metabolism in the body of a specified compound or salt thereof. The metabolites of a compound may be identified using routine techniques known in the art and their activities determined using tests such as those described herein. Such products may result for example from the oxidation, reduction, hydrolysis, amidation, deamidation, esterification, deesterification, enzymatic cleavage, and the like, of the administered compound. Accordingly, the invention includes metabolites of compounds disclosed herein, including compounds produced by a process comprising contacting a compound disclosed herein with a mammal for a period of time sufficient to yield a metabolic product thereof.
[106] A "pharmaceutically acceptable salt" refers to organic or inorganic salts of a compound disclosed herein. The pharmaceutically acceptable salts are well known in the art. For example, Berge et al, describe pharmaceutically acceptable salts in detail in J. Pharm. Sci., 1977, 66, 1-19, which is incorporated herein by reference. Some non-limiting examples of the pharmaceutically acceptable salt include salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid.
[107] Other examples of the pharmaceutically acceptable salt include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like.
[108] Pharmaceutically acceptable salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(Ci-C4 alkyl)4 salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further examples of the pharmaceutically acceptable salt include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, Ci-C8 sulfonate and aryl sulfonate.
25
SUBSTITUTE SHEET RULE 26 [109] A "solvate" refers to an association or complex of one or more solvent molecules and a compound disclosed herein. Examples of solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine. The term "hydrate" refers to the complex where the solvent molecule is water.
[110] As used herein, the term "treat", "treating" or "treatment" of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment "treat", "treating" or "treatment" refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient. In yet another embodiment, "treat", "treating" or "treatment" refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. In yet another embodiment, "treat", "treating" or "treatment" refers to preventing or delaying the onset or development or progression of the disease or disorder.
[I l l] "Inflammatory disorder/disease" as used herein can refer to any disease, disorder, or syndrome in which an excessive or unregulated inflammatory response leads to excessive inflammatory symptoms, host tissue damage, or loss of tissue function. "Inflammatory disorder/disease" also refers to a pathological state mediated by influx of leukocytes and/or neutrophil chemotaxis.
[112] "Inflammation" as used herein refers to a localized, protective response elicited by injury or destruction of tissues, which serves to destroy, dilute, or wall off (sequester) both the injurious agent and the injured tissue. Inflammation is notably associated with influx of leukocytes and/or neutrophil chemotaxis. Inflammation can result from infection with pathogenic organisms and viruses and from noninfectious means such as trauma or reperfusion following myocardial infarction or stroke, immune response to foreign antigen, and autoimmune responses. Accordingly, inflammatory disorders amenable to treatment with the compounds disclosed herein encompass disorders associated with reactions of the specific defense system as well as with reactions of the nonspecific defense system.
[113] "Specific defense system" refers to the component of the immune system that reacts to the presence of specific antigens. Examples of inflammation resulting from a response of the specific defense system include the classical response to foreign antigens, autoimmune diseases, and delayed type hypersensitivity response mediated by T-cells. Chronic inflammatory diseases, the rejection of solid transplanted tissue and organs, e.g., kidney and bone marrow transplants, and graft versus host disease (GVHD), are further examples of inflammatory reactions of the specific defense system.
26
SUBSTITUTE SHEET RULE 26 [114] "Autoimmune disease" as used herein refers to any group of disorders in which tissue injury is associated with humoral or cell-mediated responses to the body's own constituents.
[115] "Allergic disease "as used herein refers to any symptoms, tissue damage, or loss of tissue function resulting from allergy. "Arthritic disease" as used herein refers to any disease that is characterized by inflammatory lesions of the joints attributable to a variety of etiologies. "Dermatitis" as used herein refers to any of a large family of diseases of the skin that are characterized by inflammation of the skin attributable to a variety of etiologies. "Transplant rejection" as used herein refers to any immune reaction directed against grafted tissue, such as organs or cells (e.g., bone marrow), characterized by a loss of function of the grafted and surrounding tissues, pain, swelling, leukocytosis, and thrombocytopenia. The therapeutic methods of the present invention include methods for the treatment of disorders associated with inflammatory cell activation.
[116] The terms "cancer" and "cancerous" refer to or describe the physiological condition in mammals that is typically characterized by unregulated cell growth. A "tumor" comprises one or more cancerous cells. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia or lymphoid malignancies. More particular examples of such cancers include squamous cell cancer (e.g., epithelial squamous cell cancer), lung cancer including small-cell lung cancer, non-small cell lung cancer ("NSCLC"), adenocarcinoma of the lung and squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastric or stomach cancer including gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, rectal cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney or renal cancer, prostate cancer, vulval cancer, thyroid cancer, hepatic carcinoma, anal carcinoma, penile carcinoma, as well as head and neck cancer.
DESCRIPTION OF COMPOUNDS OF THE INVENTION
[117] In the present invention, novel compounds which are inhibitors of kinase activity, in particular JAK kinase activity, are disclosed. Compounds which are JAK kinase inhibitors may be useful in the treatment of diseases associated with inappropriate kinase activity, in particular inappropriate JAK kinase activity, for example in the treatment and prevention of diseases mediated by JAK kinase involved signalling pathways. Such diseases include proliferative disease, autoimmune disease, allergic disease, inflammatory disease, transplantation rejection, and their comorbidities. In particular, a compound of the present invention may be useful in the treatment of diseases such as cancer, polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), chronic obstruction pulmonary disease (COPD), asthma, systemic
27
SUBSTITUTE SHEET RULE 26 and cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis, type I diabetes mellitus, allergic airway disease, sinusitis, eczema, hives, food allergies, allergies to insect venom, inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection, cell transplant rejection, to name a few.
[118] In one embodiment, the compounds disclosed herein may show selectivity for Janus kinases over other kinases.
[119] In one aspect, provided herein is a compound having Formula (I):
Figure imgf000029_0001
or a stereoisomer, a tautomer, an N-oxide, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein each of X, Z, Z1 and R7 is as defined herein.
[120] In one embodiment, Z is C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3, 4 or 5 R1 groups;
Z1 is H, C1-C12 alkyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z1 is optionally substituted with 1, 2, 3, 4 or 5 R2 groups;
X is H, -NRaRb, -N(Ra)C(=0)Rc, -N(Rc)C(=0)NRaRb, -N(Ra)C(=0)ORc or -N(Ra)S(=0)mRc;
R7 is H, NO2, N3, CN, C3-C12 cycloalkyl, -ORc, -C(=0)Rc, -OC(=0)Rc, -C(=0)ORc or - C(=0)NRaRb, with the proviso that when X is H, R7 is C3-C12 cycloalkyl, or when each of X and R7 is H, Z has formula (Z-34):
Figure imgf000029_0002
(Z-34) wherein R7 is optionally substituted with 1, 2, 3, 4 or 5 R6 groups; each R1 and R2 is independently H, F, N02, N3, CN, C1-C12 alkyl, C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C12 aryl, 5-12 membered heteroaryl, - (CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)mRc, -S(=0)2NRaRb, -C(=0)Rc, -OC(=0)Rc, - N(Ra)C(=0)Rc, -(CR4R5)„C(=0)ORc, -(CR4R5)„C(=0)NRaRb, -C(=NRc)NRaRb,
28
SUBSTITUTE SHEET RULE 26 N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, wherein each R1 and R2 is optionally independently substituted with 1, 2, 3, 4 or 5 R3 groups; each R3 is independently H, F, CI, Br, I, N02, N3, CN, C1-C12 alkyl, C2-C12 alkenyl, C2-C12 alkynyl, C3-Ci2 cycloalkyl, 3-12 membered heterocyclyl, C6-C12 aryl, 5-12 membered heteroaryl, - (CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)mRc, -S(=0)2NRaRb, -C(=0)Rc, -OC(=0)Rc, - N(Ra)C(=0)Rc, -(CR4R5)„C(=0)ORc, -(CR4R5)„C(=0)NRaRb, -C(=NRc)NRaRb, N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, or two adjacent R3 taken together with the atoms to which they are attached form a C3-Ci2 cycloalkyl, 3-12 membered heterocyclyl, C6-C12 aryl or 5-12 membered heteroaryl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3, 4 or 5 R6 groups; each R4 and R5 is independently H, F, CI, Br, I, N , CN, OH, NH2, C1-C12 alkyl, C1-C12 alkoxy, Ci-Ci2 alkylamino, C2-Ci2 alkenyl, C2-Ci2 alkynyl, C3-Ci2 cycloalkyl, 3-12 membered heterocyclyl, C6-Ci2 aryl or 5-12 membered heteroaryl, or R4 and R5 taken together with the carbon atom to which they are attached form a C3-Ci2 cycloalkyl or 3-12 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3, 4 or 5 R6 groups; each R6 is independently F, CI, Br, I, CN, N02, N , C1-C12 alkyl, C2-Ci2 alkenyl, C2-Ci2 alkynyl, C3-Ci2 cycloalkyl, 3-12 membered heterocyclyl, C6-Ci2 aryl, 5-12 membered heteroaryl, - NH2, -NH(Ci-Ci2 alkyl), -NH(CH2)„-(C -Ci2 cycloalkyl), -NH(CH2)„-(3-12 membered heterocyclyl), -NH(CH2)„-(C6-Ci2 aryl), -NH(CH2)„-(5-12 membered heteroaryl), -N(Ci-Ci2 alkyl)2, -N[(CH2)„-(C -Ci2 cycloalkyl)] 2, -N[(CH2)„-(3-12 membered heterocyclyl)]2, -N[(CH2)„- (C6-C12 aryl)]2, -N[(CH2)„-(5-12 membered heteroaryl)]2, OH, -0(Ci-Ci2 alkyl), -0(CH2)„-C -Ci2 cycloalkyl), -0(CH2)„-(3-12 membered heterocyclyl), -0(CH2)„-(C6-Ci2 aryl) or -0(CH2)„-(5-12 membered heteroaryl); each Ra, Rb and Rc is independently H, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, -(C1-C4 alkylene)-(C3-C6 cycloalkyl), 3-7 membered heterocyclyl, -(C1-C4 alkylene)- (3-7 membered heterocyclyl), C6-Cio aryl, -(C1-C4 alkylene)-(C6-Cio aryl), 5-10 membered heteroaryl or -(C1-C4 alkylene)-(5-10 membered heteroaryl), or Ra and Rb taken together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 substituents independently selected from F, CI, CN, N3, OH, NH2, Ci-C6 alkyl, Ci-C6 haloalkyl, Ci-C6 alkoxy and Ci-C6 alkylamino;
each m is independently 0, 1 or 2; and
29
SUBSTITUTE SHEET RULE 26 each n is independently 0, 1, 2, 3 or 4.
[121] In another embodiment, R7 is H, N02, C3-C6 cycloalkyl, -ORc, -C(=0)Rc, -OC(=0)Rc, -C(=0)ORc or -C(=0)NRaRb, with the proviso that when X is H, R7 is C3-C6 cycloalkyl, or when each of X and R7 is H, Z has formula (Z-34):
Figure imgf000031_0001
(Z-34) wherein R7 is optionally substituted with 1, 2 or 3 R6 groups.
[122] In one embodiment, provided herein is the compound having Formula (II):
Figure imgf000031_0002
Wherein X is -NRaRb, -N(Ra)C(=0)Rc, -N(Rc)C(=0)NRaRb, -N(Ra)C(=0)ORc or -N(Ra)S(=0)mRc.
[123] In another embodiment, Z is C3-C6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3 or 4 R1 groups.
[124] In one embodiment, Z1 is H, C1-C4 alkyl, C3-C6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z1 is optionally substituted with 1, 2, 3 or 4 R2 groups.
[125] In another embodiment, each R1 and R2 is independently H, F, N3, CN, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, -(CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)mRc, -S(=0)2NRaRb, -C(=0)Rc, - N(Ra)C(=0)Rc, -(CR4R5)„C(=0)NRaRb, -N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, wherein each R1 and R2 is optionally independently substituted with 1, 2, 3 or 4 R3 groups.
[126] In one embodiment, each R3 is independently H, F, CI, CN, N3, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, - (CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)2NRaRb, -C(=0)Rc, -N(Ra)C(=0)Rc, (CR4R5)„C(=0)NRaRb, -N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, or two adjacent R3 taken together with the atoms to which they are attached form a C3-C6 cycloalkyl, 3-7 membered heterocyclyl, phenyl or 5-6 membered heteroaryl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 R6 groups.
30
SUBSTITUTE SHEET RULE 26 [127] In another embodiment, each R4 and R5 is independently H, F, CI, Br, I, N3, CN, OH, - NH2, Ci-C6 alkyl, Ci-C6 alkoxy, Ci-C6 alkylamino, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-7 membered heterocyclyl, phenyl or 5-6 membered heteroaryl, or R4 and R5 taken together with the carbon atom to which they are attached form a C3-C6 cycloalkyl or 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 R6 groups.
[128] In one embodiment, each R6 is independently F, CI, CN, N3, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, - NH2, -NH(Ci-C6 alkyl), -NH(CH2)n-(C3-C6 cycloalkyl), -NH(CH2)n-(3-7 membered heterocyclyl), -NH(CH2)n-phenyl, -NH(CH2)n-(5-6 membered heteroaryl), -N(Ci-C6 alkyl)2, -N[(CH2)n-(C3-C6 cycloalkyl)]2, -N[(CH2)n-(3-7 membered heterocyclyl)]2, -N[(CH2)n-phenyl]2, -N[(CH2)n-(5-6 membered heteroaryl)] 2, OH, -0(Ci-C6 alkyl), -0(CH2)n-(C3-C6 cycloalkyl), -0(CH2)n-(3-7 membered heterocyclyl), -0(CH2)n-phenyl or -0(CH2)n-(5-6 membered heteroaryl).
[129] In another embodiment, each Ra, Rb and Rc is independently H, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C6 cycloalkyl, -(C1-C2 alkylene)-(C3-C6 cycloalkyl), 3-7 membered heterocyclyl, -(C1-C2 alkylene)-(3-7 membered heterocyclyl), phenyl, -(C1-C2 alkylene)-phenyl, 5- 6 membered heteroaryl or -(C1-C2 alkylene)-(5-6 membered heteroaryl), or Ra and Rb are taken together with the nitrogen atom to which they attached form a 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2 or 3 substitutents independently selected from F, CI, CN, N3, OH, NH2, C1-C4 alkyl, C1-C4 haloalkyl, C1-C4 alkoxy and C1-C4 alkylamino.
[130] In one embodiment, Z is C4-C6 cycloalkyl, or 4-, 5- or 6-membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3 or 4 R1 groups.
[131] In another embodiment, Z is :
Figure imgf000032_0001
(Z-9) , (Z-10) , (Z-1 1 ) , (Z-12) , (Z-13) , (Z-14) , Z-15 , (Z-16) ,
31
SUBSTITUTE SHEET RULE 26
Figure imgf000033_0001
(Z-17) (Z-18) , (Z-19) , (Z-20) , (Z-21 ) , (Z-22) , (Z-23) ? (Z-24) ?
Figure imgf000033_0002
(Z-25) , (Z-26) , (Z-27) , (Z-28) , (Z-29) , (Z-30) , (Z-31 ) , (Z-32)
Figure imgf000033_0003
(Z-33) or (Z-34)
or a stereoisomer thereof, wherein Z is optionally substituted by 1, 2 or 3 R1 groups.
[132] In one embodiment, Z1 is H, methyl, ethyl, propyl, isopropyl or cyclopropyl.
[133] In another embodiment, X is H, -NH2, -NHMe, -NHC(=0)Me, -NHC(=0)NHMe or - NHC(=0)NMe2.
[134] In one embodiment, each Ra, Rb and Rc is independently H, methyl, ethyl, propyl, isopropyl, cyclopropyl or butyl, wherein each Ra, Rb and Rc is optionally independently substituted with 1, 2 or 3 substitutents independently selected from F, CI, CN, N3, OH, NH2, methyl, ethyl, - CF3, -OCH3 and -CH3NH2.
[135] In yet another embodiment, some non-limiting examples of the compound disclosed herein, and their pharmaceutically acceptable salts and solvates thereof, are shown in the following:
Table 1
Figure imgf000033_0004
(1 ) (2) (3) (4)
32
SUBSTITUTE SHEET RULE 26
Figure imgf000034_0001
Figure imgf000034_0002
SUBSTITUTE SHEET RULE 26 [136] Unless otherwise stated, all stereoisomers, tautomers, solvates, metabolites, salts, and pharmaceutically acceptable prodrugs of the compounds of Formula (I) are within the scope of the invention.
[137] The compounds disclosed herein may contain asymmetric or chiral centers, and therefore exist in different stereoisomeric forms. It is intended that all stereoisomeric forms of compounds of Formula (I), including but not limited to, diastereomers, enantiomers, atropisomers, conformers (rotamers) and geometric (cis/trans) isomers as well as mixtures thereof such as racemic mixtures, form part of the present invention.
[138] In the structures shown herein, where the stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined.
[139] The compounds of Formula (I) may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention, as defined by the claims.
[140] The compounds of Formula (I) can be in the form of salts. In one embodiment, the salts are pharmaceutically acceptable salts. The phrase "pharmaceutically acceptable" indicates that the substance or composition must be compatible chemically and/or toxicologically, with the other ingredients comprising a formulation, and/or the mammal being treated therewith. In another embodiment, the salts are not necessarily pharmaceutically acceptable salts, and which may be useful as intermediates for preparing and/or purifying compounds of Formula (I) and/or for separating enantiomers of compounds of Formula (I).
[141] Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, polygalacturonate, propionate, stearate, succinate, subsalicylate, tartrate, tosylate and trifluoroacetate salts.
[142] Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
34
SUBSTITUTE SHEET RULE 26 [143] Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.
[144] Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
[145] Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
[146] Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
[147] The pharmaceutically acceptable salts of the present invention can be synthesized from a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two. Generally, use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable. Lists of additional suitable salts can be found, e.g., in "Remington's Pharmaceutical Sciences", 20th ed., Mack Publishing Company, Easton, Pa., (1985); and in "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" by Stahl and Wermuth (Wiley- VCH, Weinheim, Germany, 2002).
[148] Furthermore, the compounds disclosed herein, including their salts, can also be obtained in the form of their hydrates, or include other solvents such as ethanol, DMSO, and the like, used for their crystallization. The compounds of the present invention may inherently or by design form solvates with pharmaceutically acceptable solvents (including water); therefore, it is intended that the invention embrace both solvated and unsolvated forms.
[149] Any formula given herein is also intended to represent isotopically unenriched forms as well as isotopically enriched forms of the compounds. Isotopically enriched compounds have
35
SUBSTITUTE SHEET RULE 26 structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2H (deuterium, D), 3H, UC, 13C, 14C, 15N, 170, 180, 18F, 31P, 32P, 35S, 36C1, 125I, respectively.
[150] In another aspect, the compounds of the invention include isotopically enriched compounds as defined herein, for example those into which radioactive isotopes, such as 3H, 14C and 18F, or those into which non-radioactive isotopes, such as 2H and 13C are present. Such isotopically enriched compounds are useful in metabolic studies (with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18F- enriched compound may be particularly desirable for PET or SPECT studies. Isotopically-enriched compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying examples and preparations using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
[151] Further, substitution with heavier isotopes, particularly deuterium (i.e., 2H or D) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements or an improvement in therapeutic index. It is understood that deuterium in this context is regarded as a substituent of a compound of Formula (I). The concentration of such a heavier isotope, specifically deuterium, may be defined by the isotopic enrichment factor. The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope. If a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5%) deuterium incorporation), at least 6000 (90%> deuterium incorporation), at least 6333.3 (95%) deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99%) deuterium incorporation), or at least 6633.3 (99.5%> deuterium incorporation). Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D20, i-acetone, DMSO- i.
36
SUBSTITUTE SHEET RULE 26 [152] In another aspect, provided herein are intermediates for preparing the compounds disclosed herein.
[153] In another aspect, provided herein are methods of preparing, methods of separating, and methods of purifying the compounds disclosed herein.
[154] In another aspect, provided herein is a pharmaceutical composition comprising a therapeutically effective amount of the compound disclosed herein, and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof.
[155] In some embodiments, the composition is a liquid, solid, semi-solid, gel, or an aerosol form.
[156] In another aspect, provided herein is a method of treating a disease or disorder modulated by one or more Janus kinases, comprising administering to a mammal in need of such treatment an effective amount of a compound or a pharmaceutical composition disclosed herein. In one embodiment, the disease or disorder is selected from proliferative disease, autoimmune disease, allergic disease, inflammatory disease or transplantation rejection.
[157] In another aspect, provided herein is the compound or the pharmaceutical composition disclosed herein for use in the treatment of disease or disorder selected from proliferative disease, autoimmune disease, allergic disease, inflammatory disease or transplantation rejection.
[158] In another aspect, provided herein is the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for the treatment of disease or disorder selected from proliferative disease, autoimmune disease, allergic disease, inflammatory disease or transplantation rejection.
[159] In another aspect, provided herein is the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for modulating the activity of Janus kinase.
PHARMACEUTICAL COMPOSITION, FORMULATIONS AND ADMINISTRATION OF THE COMPOUNDS OF THE INVENTION
[160] The present invention provides a pharmaceutical composition that include a compound of disclosed herein, or a compound listed in Table 1; and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof. The amount of compound in the pharmaceutical composition disclosed herein is such that is effective to detectably inhibit a protein kinase in a biological sample or in a patient.
[161] It will also be appreciated that certain compounds disclosed herein can exist in free form
37
SUBSTITUTE SHEET RULE 26 for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof. Some non-limiting examples of pharmaceutically acceptable derivative include pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
[162] The pharmaceutical compositions disclosed herein may be prepared and packaged in bulk form wherein a safe and effective amount of the compound disclosed herein can be extracted and then given to the patient such as with powders or syrups. Alternatively, the pharmaceutical compositions disclosed herein may be prepared and packaged in unit dosage form wherein each physically discrete unit contains the compound disclosed herein. When prepared in unit dosage form, the pharmaceutical compositions of the invention typically may contain, for example, from 0.5 mg to 1 g, or from 1 mg to 700 mg, or from 5 mg to 100 mg of the compound disclosed herein.
[163] As used herein, "pharmaceutically acceptable excipient" means a pharmaceutically acceptable material, composition or vehicle involved in giving form or consistency to the pharmaceutical composition. Each excipient must be compatible with the other ingredients of the pharmaceutical composition when commingled such that interactions which would substantially reduce the efficacy of the compound disclosed herein when administered to a patient and interactions which would result in pharmaceutical compositions that are not pharmaceutically acceptable are avoided. In addition, each excipient must be pharmaceutically-acceptable, e.g., of sufficiently high purity.
[164] Suitable pharmaceutically acceptable excipients will vary depending upon the particular dosage form chosen. In addition, suitable pharmaceutically acceptable excipients may be chosen for a particular function that they may serve in the composition. For example, certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of uniform dosage forms. Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the production of stable dosage forms. Certain pharmaceutically acceptable excipients may be chosen for their ability to facilitate the carrying or transporting of the compound or compounds disclosed herein once administered to the patient from one organ, or portion of the body, to another organ, or portion of the body. Certain pharmaceutically acceptable excipients may be chosen for their ability to enhance patient compliance.
[165] Suitable pharmaceutically acceptable excipients comprise the following types of excipients: diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweetners, flavoring agents, flavor masking agents, coloring agents, anticaking agents, hemectants, chelating agents,
38
SUBSTITUTE SHEET RULE 26 plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants, and buffering agents. The skilled artisan will appreciate that certain pharmaceutically acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the formulation and what other excipients are present in the formulation.
[166] Skilled artisans possess the knowledge and skill in the art to enable them to select suitable pharmaceutically-acceptable excipients in appropriate amounts for use in the invention. In addition, there are a number of resources that are available to the skilled artisan which describe pharmaceutically acceptable excipients and may be useful in selecting suitable pharmaceutically acceptable excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press).
[167] In Remington: The Science and Practice of Pharmacy, 21st edition, 2005, ed. D.B. Troy, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds. J. Swarbrick and J. C. Boylan, 1988-1999, Marcel Dekker, New York, the contents of each of which is incorporated by reference herein, are disclosed various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the compounds disclosed herein, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention.
[168] The pharmaceutical compositions disclosed herein are prepared using techniques and methods known to those skilled in the art. Some of the methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company).
[169] Accordingly, in another aspect the invention is directed to process for the preparation of a pharmaceutical composition comprising the compound disclosed herein and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof, which comprises mixing the ingredients. A pharmaceutical composition comprising the compound disclosed herein may be prepared by, for example, admixture at ambient temperature and atmospheric pressure.
[170] The compounds disclosed herein will typically be formulated into a dosage form adapted for administration to the patient by the desired route of administration. For example, dosage forms include those adapted for (1) oral administration such as tablets, capsules, caplets, pills, troches,
39
SUBSTITUTE SHEET RULE 26 powders, syrups, elixers, suspensions, solutions, emulsions, sachets, and cachets; (2) parenteral administration such as sterile solutions, suspensions, and powders for reconstitution; (3) transdermal administration such as transdermal patches; (4) rectal administration such as suppositories; (5) inhalation such as aerosols, solutions, and dry powders; and (6) topical administration such as creams, ointments, lotions, solutions, pastes, sprays, foams, and gels.
[171] In one embodiment, the compounds disclosed herein will be formulated for oral administration. In another embodiment, the compounds disclosed herein will be formulated for inhaled administration. In a further embodiment, the compounds disclosed herein will be formulated for intranasal administration. In another embodiment, the compounds disclosed herein will be formulated for transdermal administration. In a further embodiment, the compounds disclosed herein will be formulated for topical administration.
[172] The pharmaceutical compositions provided herein can be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets. Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric- coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.
[173] The tablet dosage forms can be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled- release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.
[174] The pharmaceutical compositions provided herein can be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate. The hard gelatin capsule, also known as the dry-filled capsule (DFC), consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft,
40
SUBSTITUTE SHEET RULE 26 globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid. The liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545. The capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
[175] The pharmaceutical compositions provided herein can be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups. An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in- water or water-in-oil. Emulsions may include a pharmaceutically acceptable nonaqueous liquid or solvent, emulsifying agent, and preservative. Suspensions may include a pharmaceutically acceptable suspending agent and preservative. Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di(lower alkyl) acetal of a lower alkyl aldehyde, e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, and hydroalcoholic solutions. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative. For a liquid dosage form, for example, a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.
[176] Other useful liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) provided herein, and a dialkylated mono- or poly-alkylene glycol, including, 1 ,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350- dimethyl ether, polyethylene glycol-550- dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations can further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
41
SUBSTITUTE SHEET RULE 26 [177] Where appropriate, dosage unit formulations for oral administration can be microencapsulated. The composition can also be prepared to prolong or sustain the release as for example by coating or embedding particulate material in polymers, wax or the like.
[178] The pharmaceutical compositions provided herein for oral administration can be also provided in the forms of liposomes, micelles, microspheres, or nanosystems. Micellar dosage forms can be prepared as described in U.S. Pat. No. 6,350,458.
[179] The pharmaceutical compositions provided herein can be provided as non-effervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form. Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents. Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide.
[180] Coloring and flavoring agents can be used in all of the above dosage forms.
[181] The compounds disclosed herein may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamidephenol, or polyethyleneoxidepolylysme substituted with palmitoyl residues. Furthermore, the compounds disclosed herein may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
[182] The pharmaceutical compositions provided herein can be formulated as immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed-release forms.
[183] The pharmaceutical compositions provided herein can be co-formulated with other active ingredients which do not impair the desired therapeutic action, or with substances that supplement the desired action.
[184] The pharmaceutical compositions provided herein can be administered parenterally by injection, infusion, or implantation, for local or systemic administration. Parenteral administration, as used herein, include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, intravesical, and subcutaneous administration.
42
SUBSTITUTE SHEET RULE 26 [185] The pharmaceutical compositions provided herein can be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection. Such dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science (see, Remington: The Science and Practice of Pharmacy, supra).
[186] The pharmaceutical compositions intended for parenteral administration can include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases.
[187] Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection. Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil. Water-miscible vehicles include, but are not limited to, ethanol, 1,3-butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, N-methyl-2-pyrrolidone, N,N-dimethylacetamide, and dimethyl sulfoxide.
[188] Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoates, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl- and propyl-parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose. Suitable buffering agents include, but are not limited to, phosphate and citrate. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable emulsifying agents include those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to EDTA. Suitable pH adjusting
43
SUBSTITUTE SHEET RULE 26 agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including a- cyclodextrin, β-cyclodextrin, hydroxypropyl-P-cyclodextrin, sulfobutylether-P-cyclodextrin, and sulfobutylether 7-P-cyclodextrin (CAPTISOL®, CyDex, Lenexa, KS).
[189] The pharmaceutical compositions provided herein can be formulated for single or multiple dosage administration. The single dosage formulations are packaged in an ampoule, a vial, or a syringe. The multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art.
[190] In one embodiment, the pharmaceutical compositions are provided as ready-to-use sterile solutions. In another embodiment, the pharmaceutical compositions are provided as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with a vehicle prior to use. In yet another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile suspensions. In yet another embodiment, the pharmaceutical compositions are provided as sterile dry insoluble products to be reconstituted with a vehicle prior to use. In still another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile emulsions.
[191] The pharmaceutical compositions provided herein can be formulated as immediate or modified release dosage forms, including delayed-, sustained-, pulsed-, controlled-, targeted-, and programmed-release forms.
[192] The pharmaceutical compositions can be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot. In one embodiment, the pharmaceutical compositions provided herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions diffuse through.
[193] Suitable inner matrixes include polymethylmethacrylate, polybutyl-methacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinyl acetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinyl alcohol, and cross-linked partially hydrolyzed polyvinyl acetate.
[194] Suitable outer polymeric membranes include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate
44
SUBSTITUTE SHEET RULE 26 copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinyl chloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer.
[195] In another aspect, the pharmaceutical compositions disclosed herein can be formulated in any dosage forms that are adapted for administration to a patient by inhalation, for example as a dry powder, an aerosol, a suspension, or a solution composition. In one embodiment, the pharmaceutical compositions disclosed herein can be formulated in a dosage form adapted for administration to a patient by inhalation as a dry powder. In a further embodiment, the pharmaceutical compositions disclosed herein can be formulated in a dosage form adapted for administration to a patient by inhalation via a nebulizer. Dry powder compositions for delivery to the lung by inhalation typically comprise the compounds disclosed herein as a finely divided powder together with one or more pharmaceutically-acceptable excipients as finely divided powders. Pharmaceutically-acceptable excipients particularly suited for use in dry powders are known to those skilled in the art and include lactose, starch, mannitol, and mono-, di-, and polysaccharides. The finely divided powder may be prepared by, for example, micronisation and milling. Generally, the size-reduced (eg micronised) compound can be defined by a D50 value of about 1 to about 10 microns (for example as measured using laser diffraction).
[196] Aerosols may be formed by suspending or dissolving the compound disclosed herein in a liquified propellant. Suitable propellants include halocarbons, hydrocarbons, and other liquified gases. Representative propellants include: trichlorofluoromethane (propellant 11), dichlorofluoromethane (propellant 12), dichlorotetrafluoroethane (propellant 114), tetrafluoroethane (HFA-134a), 1,1-difluoroethane (HFA-152a), difluoromethane (HFA-32), pentafluoroethane (HFA-12), heptafluoropropane (HFA-227a), perfluoropropane, perfluorobutane, perfluoropentane, butane, isobutane, and pentane. Aerosols comprising the compound disclosed herein will typically be administered to a patient via a metered dose inhaler (MDI). Such devices are known to those skilled in the art.
[197] The aerosol may contain additional pharmaceutically-acceptable excipients typically used with MDIs such as surfactants, lubricants, cosolvents and other excipients to improve the physical stability of the formulation, to improve valve performance, to improve solubility, or to improve taste.
[198] Pharmaceutical compositions adapted for transdermal administration may be presented as discrete patches intended to remain in intimate contact with the epidermis of the patient for a
45
SUBSTITUTE SHEET RULE 26 prolonged period of time. For example, the active ingredient may be delivered from the patch by iontophoresis as generally described in Pharmaceutical Research, 3(6), 318 (1986).
[199] Pharmaceutical compositions adapted for topical administration may be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils. Ointments, creams and gels, may, for example, be formulated with an aqueous or oily base with the addition of suitable thickening and/or gelling agent and/or solvents. Such bases may thus, for example, include water and/or oil such as liquid paraffin or a vegetable oil such as arachis oil or castor oil, or a solvent such as polyethylene glycol. Thickening agents and gelling agents which may be used according to the nature of the base include soft paraffin, aluminium stearate, cetostearyl alcohol, polyethylene glycols, woolfat, beeswax, carboxypolymethylene and cellulose derivatives, and/or glyceryl monostearate and/or non-ionic emulsifying agents.
[200] Lotions may be formulated with an aqueous or oily base and will in general also contain one or more emulsifying agents, stabilising agents, dispersing agents, suspending agents or thickening agents.
[201] Powders for external application may be formed with the aid of any suitable powder base, for example, talc, lactose or starch. Drops may be formulated with an aqueous or nonaqueous base also comprising one or more dispersing agents, solubilizing agents, suspending agents or preservatives.
[202] Topical preparations may be administered by one or more applications per day to the affected area; over skin areas occlusive dressings may advantageously be used. Continuous or prolonged delivery may be achieved by an adhesive reservoir system.
[203] For treatments of the eye or other external tissues, for example mouth and skin, the compositions may be applied as a topical ointment or cream. When formulated in an ointment, the compound disclosed herein may be employed with either a paraffinic or a water-miscible ointment base. Alternatively, the compound disclosed herein may be formulated in a cream with an oil-in- water cream base or a water-in-oil base.
USE OF THE COMPOUNDS AND COMPOSITIONS OF THE INVENTION
[204] The present invention provides a method of using a compound disclosed herein, or a pharmaceutical composition comprising the compound disclosed herein for the treatment, prevention, or amelioration of a disease or disorder that is mediated or otherwise affected via JAK kinase, including JAKl, JAK2, JAK3 or TYK2 kinase activiy or one or more symptoms of diseases or disorders that are mediated or otherwise affected via JAK kinase, including JAKl,
46
SUBSTITUTE SHEET RULE 26 JAK2, JAK3 or TYK2 kinase activity. JAK kinase can be wild type and/or mutant form of JA l, JAK2, JAK3 or TYK2 kinase.
[205] In one embodiment, provided herein is a method of using a compound disclosed herein or a pharmaceutical composition comprising a compound disclosed herein for the treatment, prevention, or amelioration of a disease or disorder that is mediated or otherwise affected via inappropriate JAKl kinase activity or one or more symptoms of diseases or disorders that are mediated or otherwise affected via inappropriate JAKl kinase activity. In another embodiment, a disease, a disorder or one or more symptoms of diseases or disorders is related to the inappropriate activity of JAK2 kinase. In yet another embodiment, a disease, a disorder or one or more symptoms of diseases or disorders is related to the inappropriate activity of JAK3 kinase.
[206] "Inappropriate JAK kinase activity" refers to any JAK kinase activity that deviates from the normal JAK kinase activity expected in a particular patient. Inappropriate JAK kinase may take the form of, for instance, an abnormal increase in activity, or an aberration in the timing and or control of JAK kinase activity. Such inappropriate activity may result then, for example, from overexpression or mutation of the protein kinase leading to inappropriate or uncontrolled activation. Accordingly, in another aspect the invention is directed to methods of treating such diseases and disorders.
[207] Consistent with the description above, such diseases or disorders include without limitation: myeloproliferative disorders such as polycythemia vera (PCV), essential thrombocythemia and idiopathic myelofibrosis (IMF); leukemia such as myeloid leukemia including chronic myeloid leukemia (CML), imatinib-resistant forms of CML, acute myeloid leukemia (AML), and a subtype of AML, acute megakaryoblastic leukemia (AMKL); lymphoproliferative diseases such as myeloma; cancer including head and neck cancer, prostate cancer, breast cancer, ovarian cancer, melanoma, lung cancer, brain tumor, pancreatic cancer and renal carcinoma; and allergic or inflammatory diseases or disorders related to immune dysfunction, immunodeficiency, immunomodulation, autoimmune diseases, transplantation rejection, graft- versus-host disease, wound healing, kidney disease, multiple sclerosis, thyroiditis, type 1 diabetes, sarcoidosis, psoriasis, allergic rhinitis, inflammatory bowel disease including Crohn's disease and ulcerative colitis (UC), systemic lupus erythematosis (SLE), arthritis, osteoarthritis, rheumatoid arthritis, osteoporosis, asthma and chronic obstructive pulmonary disease (COPD) and dry eye syndrome (or keratoconjunctivitis sicca (KCS)).
[208] In one aspect, provided herein is the compound or the pharmaceutical composition disclosed herein for preventing and/or treating proliferative disease, autoimmune disease, allergic disease, inflammatory disease or transplantation rejection in mammals including humans.
47
SUBSTITUTE SHEET RULE 26 [209] In yet another aspect, provided herein is a method of treating a mammal having, or at risk of having a disease or disclosed herein, said method comprising administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or the compounds disclosed herein. In a particular aspect, provided here is a method of treating a mammal having, or at risk of having proliferative disease, autoimmune disease, allergic disease, inflammatory disease or transplantation rejection.
[210] In additional method of treatment aspects, provided herein is a method of treatment and/or prophylaxis of a mammal susceptible to or afflicted with a proliferative disease, said methods comprising administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or compounds disclosed herein. In a specific embodiment, the proliferative disease is selected from cancer (e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer), polycythemia vera, essential thrombocytosis, myelofibrosis, leukemia (e.g. AML, CML, ALL or CLL), and multiple myeloma.
[211] In another aspect, provided herein is the compound or the pharmaceutical composition disclosed herein for use in the treatment, and/or prophylaxis of a proliferative disease. In a specific embodiment, the proliferative disease is selected from cancer (e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer), polycythemia vera, essential thrombocytosis, myelofibrosis, leukemia (e.g. AML, CML, ALL or CLL), and multiple myeloma.
[212] In yet another aspect, provided herein is the use of the compound or the pharmaceutical composition disclosed herein for use in the manufacture of a medicament for the treatment, and/or prophylaxis of a proliferative disease. In a specific embodiment, the proliferative disease is selected from cancer (e.g. solid tumors such as uterine leiomyosarcoma or prostate cancer), polycythemia vera, essential thrombocytosis, myelofibrosis, leukemia (e.g. AML, CML, ALL or CLL), and multiple myeloma.
[213] In another aspect, provided herein is a method of treatment and/or prophylaxis of a mammal susceptible to or afflicted with an autoimmune disease. The methods comprise administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or compounds disclosed herein. In a specific embodiment, the autoimmune disease is selected from COPD, asthma, systemic and cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis and type I diabetes mellitus.
[214] In another aspect, provided herein is the compound or the pharmaceutical composition disclosed herein for use in the treatment, and/or prophylaxis of an autoimmune disease. In a specific embodiment, the autoimmune disease is selected from COPD, asthma, systemic and
48
SUBSTITUTE SHEET RULE 26 cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis and type I diabetes mellitus.
[215] In yet another aspect, provided here is the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for the treatment, and/or prophylaxis of an autoimmune disease. In a specific embodiment, the autoimmune disease is selected from COPD, asthma, systemic and cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis and type I diabetes mellitus.
[216] In a method of treatment aspects, provided herein are methods of treatment and/or prophylaxis of a mammal susceptible to or afflicted with an allergic disease. The methods comprising administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or the compounds disclosed herein. In a specific embodiment, the allergic disease is selected from allergic airway disease, sinusitis, eczema and hives, food allergies and allergies to insect venom.
[217] In another aspect, provided herein is the compound or the pharmaceutical composition disclosed herein for use in the treatment, and/or prophylaxis of an allergic disease. In a specific embodiment, the allergic disease is selected from allergic airway disease, sinusitis, eczema and hives, food allergies and allergies to insect venom.
[218] In yet another aspect, provided herein is the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for the treatment, or prophylaxis of an allergic disease. In a specific embodiment, the allergic disease is selected from allergic airway disease, sinusitis, eczema and hives, food allergies and allergies to insect venom.
[219] In another aspect, provided herein are methods of treatment and/or prophylaxis of a mammal susceptible to or afflicted with an inflammatory disease. The methods comprise administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or the compounds disclosed herein. In a specific embodiment, the inflammatory disease is selected from inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis and psoriatic arthritis.
[220] In another aspect, provided herein is the compound or the pharmaceutical composition disclosed herein for use in the treatment, and/or prophylaxis of an inflammatory disease. In a specific embodiment, the inflammatory disease is selected from inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis and psoriatic arthritis.
[221] In yet another aspect, provided herein is the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for the treatment, and/or
49
SUBSTITUTE SHEET RULE 26 prophylaxis of an inflammatory disease. In a specific embodiment, the inflammatory disease is selected from inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis and psoriatic arthritis.
[222] In another aspect, provided herein are methods of treatment and/or prophylaxis of a mammal susceptible to or afflicted with transplantation rejection. The methods comprising administering an effective condition-treating or condition-preventing amount of one or more of the pharmaceutical compositions or the compound of the invention herein described. In a specific embodiment, the transplantation rejection is organ transplant rejection, tissue transplant rejection and cell transplant rejection.
[223] In another aspect, provided herein is the compound or the pharmaceutical composition disclosed herein for use in the treatment, and/or prophylaxis of transplantation rejection. In a specific embodiment, the transplantation rejection is organ transplant rejection, tissue transplant rejection and cell transplant rejection.
[224] In yet another aspect, provided herein is the use of the compound or the pharmaceutical composition disclosed herein for use in the manufacture of a medicament for the treatment and/or prophylaxis of transplantation rejection. In a specific embodiment, the transplantation rejection is organ transplant rejection, tissue transplant rejection and cell transplant rejection.
[225] The present invention provides the compound or the pharmaceutical composition disclosed herein for use as a pharmaceutical especially in the treatment and/or prophylaxis of the aforementioned diseases or disorders. Also provided herein is the use of the compound or the pharmaceutical composition disclosed herein in the manufacture of a medicament for the treatment and/or prophylaxis of one of the aforementioned diseases or disorders.
[226] A particular regimen of the present method comprises the administration to a subject suffering from a disease involving inflammation, of an effective amount of a compound disclosed herein for a period of time sufficient to reduce the level of inflammation in the subject, and preferably terminate the processes responsible for said inflammation. A special embodiment of the method comprises administering of an effective amount of a compound disclosed herein to a subject patient suffering from or susceptible to the development of rheumatoid arthritis, for a period of time sufficient to reduce or prevent, respectively, inflammation in the joints of said patient, and preferably terminate, the processes responsible for said inflammation.
[227] A further particular regimen of the present method comprises the administration to a subject suffering from a disease involving proliferative disease, of an effective amount of a compound disclosed herein for a period of time sufficient to reduce the level of proliferative
50
SUBSTITUTE SHEET RULE 26 disease in the subject, and preferably terminate the processes responsible for said proliferative disease. A particular embodiment of the method comprises administering of an effective amount of a compound disclosed herein to a subject patient suffering from or susceptible to the development of cancer, for a period of time sufficient to reduce or prevent, respectively, solid tumor of said patient, and preferably terminate, the processes responsible for said solid.
COMBINATION THERAPY
[228] A compound disclosed herein can be administered as the sole active agent or it can be administered in combination with other therapeutic agents, including other compounds that demonstrate the same or a similar therapeutic activity and that are determined to be safe and efficacious for such combined administration.
[229] In one aspect, provided herein is a method of treating, preventing, or ameliorating a disease or disorder comprising administering a safe and effective amount of a combination comprising the compound disclosed herein together with one or more therapeutically active agents. In one embodiment, the combinations comprising one or two other therapeutic agents.
[230] Example of other therapeutic agents may include without limitation anti-cancer agents, including chemotherapeutic agents and antiproliferative agents; anti-inflammatory agents and immunomodulatory agents or immunosuppressive agents.
[231] In another aspect, provided herein is a product comprising a compound disclosed herein and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy. In one embodiment, the therapy is the treatment of a disease or disorder mediated by the activity of the JAK enzymes. Products provided as a combined preparation include a composition comprising the compound disclosed herein and the other therapeutic agent(s) together in the same pharmaceutical composition, or the compound disclosed herein and the other therapeutic agent(s) in separate form, e.g. in the form of a kit.
[232] In another aspect, provided herein is a pharmaceutical composition comprising a compound disclosed herein and another therapeutic agent(s). In one embodiment, the pharmaceutical composition may comprise a pharmaceutically acceptable excipient, carrier, adjuvant or veichle as described above.
[233] In another aspect, the invention provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound disclosed herein. In one embodiment, the kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet. An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like.
51
SUBSTITUTE SHEET RULE 26 [234] The invention also provides the use of a compound disclosed herein for treating a disease or condition mediated by the activity of the JAK enzymes, wherein the patient has previously (e.g. within 24 hours) been treated with another therapeutic agent. The invention also provides the use of another therapeutic agent for treating a disease or condition mediated by the activity of the JAK enzymes, wherein the patient has previously (e.g. within 24 hours) been treated with a compound disclosed herein.
[235] The compounds disclosed herein may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to, other drugs e.g. immunosuppressive or immunomodulating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of alio- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders, or a chemotherapeutic agent, e.g a malignant cell anti-proliferative agent. For example, the compounds disclosed herein may be used in combination with a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a mTOR inhibitor, e.g. rapamycin, 40-O-(2- hydroxyethyl)-rapamycin, CCI779, ABT578, AP23573, TAFA-93, biolimus-7 or biolimus-9; an ascomycin having immuno-suppressive properties, e.g. ABT-281, ASM981, etc.; corticosteroids; cyclophosphamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenolic acid or salt; mycophenolate mofetil; 15- deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; a PKC inhibitor, e.g. as disclosed in WO 02/38561 or WO 03/82859, e.g. the compound of Example 56 or 70; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40, CD45, CD52, CD58, CD80, CD86 or their ligands; other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA41g (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA- 1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or antihistamines; or antitussives, or a bronchodilatory agent; or an angiotensin receptor blockers; or an anti-infectious agent.
[236] Where the compounds disclosed herein are administered in conjunction with other immunosuppressive/immunomodulatory, anti-inflammatory, chemotherapeutic or anti- infectious therapy, dosages of the co-administered immunosuppressant, immunomodulatory, antiinflammatory, chemotherapeutic or anti-infectious compound will of course vary depending on the type of co-drug employed, e.g. whether it is a steroid or a calcineurin inhibitor, on the specific drug employed, on the condition being treated and so forth.
52
SUBSTITUTE SHEET RULE 26 [237] In one aspect, provided herein is a combination comprising a compound disclosed herein together with a P2-adrenoreceptor agonist. Examples of P2-adrenoreceptor agonists include salmeterol, salbutamol, formoterol, salmefamol, fenoterol, carmoterol, etanterol, naminterol, clenbuterol, pirbuterol, flerbuterol, reproterol, bambuterol, indacaterol, terbutaline and salts thereof, for example the xinafoate (l-hydroxy-2-naphthalenecarboxylate) salt of salmeterol, the sulphate salt or free base of salbutamol or the fumarate salt of formoterol. In one embodiment, long-acting P2-adrenoreceptor agonists, for example, compounds which provide effective bronchodilation for about 12 h or longer, are preferred.
[238] The P2-adrenoreceptor agonist may be in the form of a salt formed with a pharmaceutically acceptable acid selected from sulphuric, hydrochloric, fumaric, hydroxynaphthoic (for example 1- or 3-hydroxy-2-naphthoic), cinnamic, substituted cinnamic, triphenylacetic, sulphamic, sulphanilic, naphthaleneacrylic, benzoic, 4-methoxybenzoic, 2- or 4-hydroxybenzoic, 4-chlorobenzoic and 4- phenylbenzoic acid.
[239] In another aspect, provided herein is a combination comprising a compound disclosed herein together with corticosteroids. Suitable corticosteroids refer to those oral and inhaled corticosteroids and their pro-drugs which have anti-inflammatory activity. Examples include methyl prednisolone, prednisolone, dexamethasone, fluticasone propionate, 6a,9a-difluoro-l 1 β- hydroxy- 16a-methyl- 17a- [(4-methyl- 1 ,3 -thiazole-5 -carbonyl)oxy] -3 -oxo-androsta- 1 ,4-diene- 17β- carbothioic acid (Sj-fluoromethyl ester, 6a,9a-difluoro-17a-[(2-furanylcarbonyl)oxy]-l 1 β- hydroxy-16a-methyl-3 -oxo-androsta- 1 ,4-diene-l 7 β-carbothioic acid (5^-fluoromethyl ester (fluticasone furoate), 6a,9a-difluoro-l ^-hydroxy-16a-methyl-3-oxo-17a-propionyloxy-androsta-
Figure imgf000054_0001
acid (S -(2-oxo-tetrahydro-furan-3(5)-yl) ester, 6a,9a-difluoro-l 1 β- hydroxy- 16a-methyl-3 -oxo- 17a-(2,2,3 ,3-tetramethycyclopropylcarbonyl)oxy-androsta- 1 ,4-diene-
Figure imgf000054_0002
ethycyclopropylcarbonyl)oxy-3-oxo-androsta-l ,4-diene-17β-carbothioic acid (5)-fluoromethyl ester, beclomethasone esters (for example the 17-propionate ester or the 17,21-dipropionate ester), budesonide, flunisolide, mometasone esters (for example mometasone furoate), triamcinolone acetonide, rofleponide, ciclesonide (16a,17-[[(cz5)-cyclohexylmethylene]bis(oxy)]-l ^,21- dihydroxy-pregna-l ,4-diene-3,20-dione), butixocort propionate, RPR-106541 , and ST-126. Preferred corticosteroids include fluticasone propionate, 6a,9a-difluoro-l ^-hydroxy-16a-methyl- 17 -[(4-methyl-l,3-thiazole-5-carbonyl)oxy]-3-oxo-androsta-l ,4-diene-17β-carbothioic acid (S)- fluoromethyl ester, 6a,9a-difluoro-17a-[(2-furanylcarbonyl)oxy]-l ^-hydroxy-16a-methyl-3-oxo- androsta-l ,4-diene-17β-carbothioic acid (5)-fluoromethyl ester, 6a,9a-difluoro-l ^-hydroxy-16a- methyl-3-oxo-17a-(2,2 -tetramethycyclopropylcarbonyl)oxy-androsta-l ,4-diene-17P-
53
SUBSTITUTE SHEET RULE 26 carbothioic acid (S)-cyanomethyl ester and 6a,9a-difluoro-l ip-hydroxy-16a-methyl-17a-(l- methyl cyclopropylcarbonyl)oxy-3-oxo-androsta-l,4-diene-17P-carbothioic acid (5)-fluoromethyl ester. In one embodiment the corticosteroid is 6a, 9a-difluoro-17a-[(2-furanylcarbonyl)oxy]-l ip- hydroxy-16a-methyl-3-oxo-androsta-l,4-diene-17P-carbothioic acid (5)-fluoromethyl ester.
[240] In another aspect, provided herein is a combination comprising a compound disclosed herein together with non-steroidal GR agonist. Non-steroidal compounds having glucocorticoid agonism that may possess selectivity for transrepression over transactivation and that may be useful in combination therapy include those covered in the following patents: WO 03/082827, WO 98/54159, WO 04/005229, WO 04/009017, WO 04/018429, WO 03/104195, WO 03/082787, WO 03/082280, WO 03/059899, WO 03/101932, WO 02/02565, WO 01/16128, WO 00/66590, WO 03/086294, WO 04/026248, WO 03/061651 and WO 03/08277. Further non-steroidal compounds are covered in: WO 2006/000401, WO 2006/000398 and WO 2006/015870.
[241] In another aspect, provided herein is a combination comprising a compound disclosed herein together with non-steroidal anti-inflammatory drugs (NSAID's). Examples of NSAID's include sodium cromoglycate, nedocromil sodium, phosphodiesterase (PDE) inhibitors (for example, theophylline, PDE4 inhibitors or mixed PDE3/PDE4 inhibitors), leukotriene antagonists, inhibitors of leukotriene synthesis (for example montelukast), iNOS inhibitors, tryptase and elastase inhibitors, beta-2 integrin antagonists and adenosine receptor agonists or antagonists (e.g. adenosine 2a agonists), cytokine antagonists (for example chemokine antagonists, such as a CCR3 antagonist) or inhibitors of cytokine synthesis, or 5 -lipoxygenase inhibitors. An iNOS (inducible nitric oxide synthase inhibitor) is preferably for oral administration. Examples of iNOS inhibitors include those disclosed in WO 93/13055, WO 98/30537, WO 02/50021, WO 95/34534 and WO 99/62875. Examples of CCR3 inhibitors include those disclosed in WO 02/26722.
[242] In one embodiment, the invention provides the use of the compounds disclosed herein in combination with a phosphodiesterase 4 (PDE4) inhibitor, especially in the case of a formulation adapted for inhalation. The PDE4-specific inhibitor useful in this aspect of the invention may be any compound that is known to inhibit the PDE4 enzyme or which is discovered to act as a PDE4 inhibitor, and which are only PDE4 inhibitors, not compounds which inhibit other members of the PDE family, such as PDE3 and PDE5, as well as PDE4. Compounds include czs-4-cyano-4-(3- cyclopentyloxy-4- methoxyphenyl)cyclohexan-l-carboxylic acid, 2-carbomethoxy-4-cyano-4-(3- cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan- 1 -one and cis- [4-cyano-4-(3 - cyclopropylmethoxy-4-difluoromethoxyphenyl)cyclohexan-l-ol]. Also, czs -4-cyano-4-[3- (cyclopentyloxy)-4-methoxyphenyl]cyclohexane-l-carboxylic acid (also known as cilomilast) and its salts, esters, pro-drugs or physical forms, which is described in U.S. Patent No. 5,552,438
54
SUBSTITUTE SHEET RULE 26 issued 03 September, 1996; this patent and the compounds it discloses are incorporated herein in full by reference.
[243] In another aspect, provided herein is a combination comprising a compound disclosed herein together with an anticholinergic agent. Examples of anticholinergic agents are those compounds that act as antagonists at the muscarinic receptors, in particular those compounds which are antagonists of the Mi or M3 receptors, dual antagonists of the Mi/M3 or Μ23, receptors or pan-antagonists of the Mi/M2/M3 receptors. Exemplary compounds for administration via inhalation include ipratropium (for example, as the bromide, CAS 22254-24-6, sold under the name ATROVENT®), oxitropium (for example, as the bromide, CAS 30286-75-0) and tiotropium (for example, as the bromide, CAS 136310-93-5, sold under the name SPIRIVA®). Also of interest are revatropate (for example, as the hydrobromide, CAS 262586-79-8) and LAS-34273 which is disclosed in WO 01/04118. Exemplary compounds for oral administration include pirenzepine (CAS 28797-61-7), darifenacin (CAS 133099-04-4, or CAS 133099-07-7 for the hydrobromide sold under the name ENABLEX®), oxybutynin (CAS 5633-20-5, sold under the name DITROPAN®), terodiline (CAS 15793-40-5), tolterodine (CAS 124937-51-5, or CAS 124937-52- 6 for the tartrate, sold under the name DETROL®), otilonium (for example, as the bromide, CAS 26095-59-0, sold under the name SPASMOMEN®), trospium chloride (CAS 10405-02-4) and solifenacin (CAS 242478-37-1, or CAS 242478-38-2 for the succinate also known as YM-905 and sold under the name VESICARE®).
[244] In another aspect, provided herein is a combination comprising a compound disclosed herein together with an HI antagonist. Examples of HI antagonists include, without limitation, amelexanox, astemizole, azatadine, azelastine, acrivastine, brompheniramine, cetirizine, levocetirizine, efletirizine, chlorpheniramine, clemastine, cyclizine, carebastine, cyproheptadine, carbinoxamine, descarboethoxyloratadine, doxylamine, dimethindene, ebastine, epinastine, efletirizine, fexofenadine, hydroxyzine, ketotifen, loratadine, levocabastine, mizolastine, mequitazine, mianserin, noberastine, meclizine, norastemizole, olopatadine, picumast, pyrilamine, promethazine, terfenadine, tripelennamine, temelastine, trimeprazine and triprolidine, particularly cetirizine, levocetirizine, efletirizine and fexofenadine. In a further embodiment the invention provides a combination comprising a compound disclosed herein together with an H3 antagonist (and/or inverse agonist). Examples of H3 antagonists include, for example, those compounds disclosed in WO 2004/035556 and in WO 2006/045416. Other histamine receptor antagonists which may be used in combination with the compounds disclosed herein include antagonists (and/or inverse agonists) of the H4 receptor, for example, the compounds disclosed in Jablonowski et al, J. Med. Chem. 46:3957-3960 (2003).
55
SUBSTITUTE SHEET RULE 26 [245] In still another aspect, provided herein is a combination comprising a compound disclosed herein together with a PDE4 inhibitor and a P2-adrenoreceptor agonist.
[246] In yet another aspect, provided herein is a combination comprising a compound disclosed herein together with an anticholinergic and a PDE-4 inhibitor.
[247] The combinations referred to above may conveniently be presented for use in the form of a pharmaceutical composition and thus pharmaceutical compositions comprising a combination as defined above together with a pharmaceutically acceptable excipient or carrier represent a further aspect of the invention.
[248] The individual compounds of such combinations may be administered either sequentially or simultaneously in separate or combined pharmaceutical formulations. In one embodiment, the individual compounds will be administered simultaneously in a combined pharmaceutical formulation. Appropriate doses of known therapeutic agents will readily be appreciated by those skilled in the art.
[249] The invention thus provides, in a further aspect, a pharmaceutical composition comprising a combination of a compound disclosed herein together with another therapeutically active agent.
[250] In one embodiment, the pharmaceutical composition comprises a combination of a compound disclosed herein together with a PDE4 inhibitor.
[251] In another embodiment, the pharmaceutical composition comprises a combination of a compound disclosed herein together with a P2-adrenoreceptor agonist.
[252] In another embodiment, the pharmaceutical composition comprises a combination of a compound disclosed herein together with a corticosteroid.
[253] In another embodiment, the pharmaceutical composition comprises a combination of a compound disclosed herein together with a non-steroidal GR agonist.
[254] In another embodiment, the pharmaceutical composition comprises a combination of a compound disclosed herein together with an anticholinergic agent.
[255] In still another embodiment, the pharmaceutical composition comprises a combination of a compound disclosed herein together with an antihistamine.
[256] In the field of medical oncology it is normal practice to use a combination of different forms of treatment to treat each patient with cancer. In medical oncology the other component(s) of such conjoint treatment in addition to compositions disclosed herein may be, for example, surgery, radiotherapy, chemotherapy, signal transduction inhibitors or modulators (e.g. kinase inhibitors or modulators) and/or monoclonoal antibodies.
56
SUBSTITUTE SHEET RULE 26 [257] A compound disclosed herein may also be used to advantage in combination with each other or in combination with other therapeutic agents, especially other antiproliferative agents. Such antiproliferative agents include, but are not limited to, aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active agents; alkylating agents; histone deacetylase inhibitors; compounds that induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; agents used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Flt-3; Hsp90 inhibitors; temozolomide (TEMODAL®); and leucovorin.
[258] The term "aromatase inhibitor", as used herein, relates to a compound which inhibits the estrogen production, i.e., the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to, steroids, especially atamestane, exemestane and formestane; and, in particular, nonsteroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketoconazole, vorozole, fadrozole, anastrozole and letrozole. Exemestane can be administered, e.g., in the form as it is marketed, e.g., under the trademark AROMASIN®. Formestane can be administered, e.g., in the form as it is marketed, e.g., under the trademark LENTARON®. Fadrozole can be administered, e.g., in the form as it is marketed, e.g., under the trademark AFEMA®. Anastrozole can be administered, e.g., in the form as it is marketed, e.g., under the trademark ARIMIDEX®. Letrozole can be administered, e.g., in the form as it is marketed, e.g., under the trademark FEMARA® or FEMAR®. Aminoglutethimide can be administered, e.g., in the form as it is marketed, e.g., under the trademark ORIMETEN®. A combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, e.g., breast tumors.
[259] The term "anti-estrogen", as used herein, relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level. The term includes, but is not limited to, tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride. Tamoxifen can be administered, e.g., in the form as it is marketed, e.g., under the trademark NOLVADEX®. Raloxifene hydrochloride can be administered, e.g., in the form as it is marketed, e.g., under the trademark EVISTA®. Fulvestrant can be formulated as disclosed in U.S. Patent No. 4,659,516 or it can be
57
SUBSTITUTE SHEET RULE 26 administered, e.g., in the form as it is marketed, e.g., under the trademark FASLODEX®. A combination of the invention comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, e.g., breast tumors.
[260] The term "anti-androgen", as used herein, relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (CASODEX®), which can be formulated, e.g., as disclosed in U.S. Patent No. 4,636,505.
[261] The term "gonadorelin agonist", as used herein, includes, but is not limited to, abarelix, goserelin and goserelin acetate. Goserelin is disclosed in U.S. Patent No. 4,100,274 and can be administered, e.g., in the form as it is marketed, e.g., under the trademark ZOLADEX®. Abarelix can be formulated, e.g., as disclosed in U.S. Patent No. 5,843,901. The term "topoisomerase I inhibitor", as used herein, includes, but is not limited to, topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148 (compound Al in WO 99/17804). Irinotecan can be administered, e.g., in the form as it is marketed, e.g., under the trademark CAMPTOSAR®. Topotecan can be administered, e.g., in the form as it is marketed, e.g., under the trademark HYCAMTIN®.
[262] The term "topoisomerase II inhibitor", as used herein, includes, but is not limited to, the anthracyclines, such as doxorubicin, including liposomal formulation, e.g., CAELYX®; daunorubicin; epirubicin; idarubicin; nemorubicin; the anthraquinones mitoxantrone and losoxantrone; and the podophillotoxines etoposide and teniposide. Etoposide can be administered, e.g., in the form as it is marketed, e.g., under the trademark ETOPOPHOS®. Teniposide can be administered, e.g., in the form as it is marketed, e.g., under the trademark VM 26-BRISTOL®. Doxorubicin can be administered, e.g., in the form as it is marketed, e.g., under the trademark ADRIBLASTIN® or ADRIAMYCIN®.
[263] Epirubicin can be administered, e.g., in the form as it is marketed, e.g., under the trademark FARMORUBICIN®. Idarubicin can be administered, e.g., in the form as it is marketed, e.g., under the trademark ZAVEDOS®. Mitoxantrone can be administered, e.g., in the form as it is marketed, e.g., under the trademark NOVANTRON®.
[264] The term "microtubule active agent" relates to microtubule stabilizing, microtubule destabilizing agents and microtublin polymerization inhibitors including, but not limited to, taxanes, e.g., paclitaxel and docetaxel; vinca alkaloids, e.g., vinblastine, especially vinblastine sulfate; vincristine, especially vincristine sulfate and vinorelbine; discodermolides; cochicine; and epothilones and derivatives thereof, e.g., epothilone B or D or derivatives thereof. Paclitaxel may
58
SUBSTITUTE SHEET RULE 26 be administered, e.g., in the form as it is marketed, e.g., TAXOL®. Docetaxel can be administered, e.g., in the form as it is marketed, e.g., under the trademark TAXOTERE®. Vinblastine sulfate can be administered, e.g., in the form as it is marketed, e.g., under the trademark VINBLASTIN R.P®. Vincristine sulfate can be administered, e.g., in the form as it is marketed, e.g., under the trademark FARMISTIN®. Discodermolide can be obtained, e.g., as disclosed in U.S. Patent No. 5,010,099. Also included are epothilone derivatives which are disclosed in WO 98/10121, U.S. Patent No. 6,194,181, WO 98/25929, WO 98/08849, WO 99/43653, WO 98/22461 and WO 00/31247. Especially preferred are epothilone A and/or B.
[265] The term "alkylating agent", as used herein, includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel). Cyclophosphamide can be administered, e.g., in the form as it is marketed, e.g., under the trademark CYCLOSTIN®. Ifosfamide can be administered, e.g., in the form as it is marketed, e.g., under the trademark HOLOXAN®.
[266] The term "histone deacetylase inhibitors" or "HDAC inhibitors" relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes compounds disclosed in WO 02/22577, especially N-hydroxy-3-[4-[[(2-hydroxyethyl)[2-(lH- indo]-3-y ethy[]-amino]methyl]phenyl]-2E-2-propenamide, A¾ydroxy-3-[4-[[[2-(2-methyl-l H- indol-3-yl)-ethy[]-amino]methyl]phenyl]-2E-2-propenamide and pharmaceutically acceptable salts thereof. It further especially includes suberoyianilide hydroxamic acid (SAHA).
[267] The term "antineoplastic antimetabolite" includes, but is not limited to, 5-fluorouracil or 5- FU; capecitabine; gemcitabine; DNA demethylating agents, such as 5-azacytidine and decitabine; methotrexate and edatrexate; and folic acid antagonists, such as pemetrexed. Capecitabine can be administered, e.g., in the form as it is marketed, e.g., under the trademark XELODA®. Gemcitabine can be administered, e.g., in the form as it is marketed, e.g., under the trademark GEMZAR®. Also included is the monoclonal antibody trastuzumab which can be administered, e.g., in the form as it is marketed, e.g., under the trademark HERCEPTIN®.
[268] The term "platin compound", as used herein, includes, but is not limited to, carboplatln, cis-platin, cisplatinum and oxaliplatin. Carboplatin can be administered, e.g., in the form as it is marketed, e.g., under the trademark CARBOPLAT®. Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g., under the trademark ELOXATIN®.
[269] The term "compounds targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or further anti-angiogenic compounds", as used herein, includes, but
59
SUBSTITUTE SHEET RULE 26 is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, e.g., a) compounds targeting, decreasing or inhibiting the activity of the p!ate!et-derived growth factor- receptors (PDGFR), such as compounds which target, decrease or mhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, e.g., a . -phenyl-2-pyrirnidine-arnine derivative, e.g., imatinib, SU101 , SU6668 and GFB-1 1 1; b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor- receptors (FGFR); c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as compounds which target, decrease or mhibit the activity of IGF-IR, especially compounds which inhibit the IGF-IR receptor, such as those compounds disclosed in WO 02/092599; d) compounds targeting, decreasing or inhibiting the activity of the Trk receptor tyrosine kinase family; e) compounds targeting, decreasing or inhibiting the activity of the Axl receptor tyrosine kinase family; f) compounds targeting, decreasing or inhibiting the activi ty of the c- et receptor; g) compounds targeting, decreasing or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase; h) compounds targeting, decreasing or inhibiting the activity of the c-kit receptor tyrosine kinases - (part of the PDGFR family), such as compounds which target, decrease or inhibit the activity of the c-Kit receptor tyrosine kinase family, especially compounds which inhibit the c-Kit receptor, e.g., imatinib; i) compounds targeting, decreasing or inhibiting the activity of members of the e-Abl family and their gene-fusion products, e.g., BCR-Abl kinase, such as compounds which target decrease or inhibit the activity of e-Abl family members and their gene fusion products, e.g., a jV-phenyl-2- p rimidine-amine derivative, e.g., imatinib, PD180970, AG957, NSC 680410 or PDI73955 from ParkeDavis; j) compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK, FAK, PDK and Ras/MAPK family members, or Pl(3) kinase family, or of the Pl(3)-kinase-related kinase family, and/or members of the cyclin-dependent kinase family (CDK) and are especially those
60
SUBSTITUTE SHEET RULE 26 staurosporine derivatives disclosed in U.S. Patent No. 5,093,330, e.g., midostaurin; examples of further compounds include, e.g., UCN-01 ; safmgol; BAY 43-9006; Bryostatin 1 ; Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; Isis 3521; LY333531/LY379196; isoehinolme compounds, such as those disclosed in WO 00/09495; FTis; PD184352; or QAN697 (a PL3K inhibitor); k) compounds targeting, decreasing or inhibiting the activity of protein-tyrosme kinase inhibitors, such as compounds which target, decrease or inhibit the activity of protein-tyrosine kinase inhibitors include imatmib mesylate (GLEEVEC®) or tyrphostin. A tyrphostin is preferably a low molecular weight (Mr < 1500) compound, or a pharmaceutically acceptable salt thereof, especially a compound selected from the benzylidenemalonitrile class or the S-arylbenzenemaloniriie or bisubstrate quinoiine class of compounds, more especially any compound selected from the group consisting of Tyrphostin A23/RG-5081 Q, AG 99, Tyrphostin AG 213, Tyrphostin AG 1748, Tyrphostin AG 490, Tyrphostin B44, Tyrphostin B44 (+) enantiomer, Tyrphostin AG 555, AG 494, Tyrphostin AG 556, AG957 and adaphostin (4-{[(2,5-d.ihyd.roxyphenyl)methyl]amino}- benzoie acid adamantyl ester, NSC 680410, adaphostin; and
1) compoimds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR, ErbB2, ErbB3, ErbB4 as homo- or hetero-dirners), such as compounds which target, decrease or inhibit the activity of the epidermal grow h factor receptor family are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, e.g., EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, and are in particular those compounds, proteins or monoclonal antibodies generically and specifical ly disclosed in W 97/02266, e.g., the compound of Example 39, or in EP 0564409; WO 99/03854; EP 0520722; EP 0566226; EP 0787722; EP 0837063; U.S. Patent No. 5,747,498; WO 98/10767; WO 97/30034; WO 97/49688; WO 97/38983 and, especially, WO 96/30347, e.g., compound known as CP 358774; WO 96/33980, e.g., compound ZD 1839; and WO 95/03283, e.g., compound ZM 105180, e.g., trastuzumab (HE CEPTIN), cetuximab, ressa, Tarceva, OSI-774, CI-1033, EKB-569, GW-2016, El . l , E2.4, E2.5, E6.2, E6.4, E2. l l, E6.3 or E7.6.3; and 7H-pyrrolo-[2,3-dJpyrimidine derivatives which are disclosed in WO 03/013541.
[270] Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g., unrelated to protein or lipid kinase inhibition, e.g., thalidomide (THALOMID®) and TNP-470.
[271] Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are, e.g., inhibitors of phosphatase 1, phosphatase 2A, PTEN or CDC25, e.g., okadaic acid or a derivative thereof,
61
SUBSTITUTE SHEET RULE 26 [272] Compounds that induce cell differentiation processes are e.g. retinoic acid, α-, γ- or δ- tocopherol or α-, γ- or δ-tocotrienol.
[273] The term cyclooxygenase inhibitor, as used herein, includes, but is not limited to, e.g., Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CELEBREX®), rofecoxib (VIOXX®), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, e.g., 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid or lumiracoxib.
[274] The term "bisphosphonates", as used herein, includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid. "Etridonic acid" can be administered, e.g., in the form as it is marketed, e.g., under the trademark DIDRONEL®. "Clodronic acid" can be administered, e.g., in the form as it is marketed, e.g., under the trademark BONEFOS®. "Tiludronic acid" can be administered, e.g., in the form as it is marketed, e.g., under the trademark SKELID®. "Pamidronic acid" can be administered, e.g., in the form as it is marketed, e.g., under the trademark AREDIA™. "Alendronic acid" can be administered, e.g., in the form as it is marketed, e.g., under the trademark FOSAMAX®. "Ibandronic acid" can be administered, e.g., in the form as it is marketed, e.g., under the trademark BONDRANAT®. "Risedronic acid" can be administered, e.g., in the form as it is marketed, e.g., under the trademark ACTONEL®. "Zoledronic acid" can be administered, e.g., in the form as it is marketed, e.g., under the trademark ZOMETA®.
[275] The term "mTOR inhibitors" relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity, such as sirolimus (Rapamune®), everolimus (Certican™), CCI-779 and ABT578.
[276] The term "heparanase inhibitor", as used herein, refers to compounds which target, decrease or inhibit heparin sulphate degradation. The term includes, but is not limited to, PI-88.
[277] The term "biological response modifier", as used herein, refers to a lymphokine or interferons, e.g., interferon γ.
[278] The term "inhibitor of Ras oncogenic isoforms", e.g., H-Ras, K-Ras or N-Ras, as used herein, refers to compounds which target, decrease or inhibit the oncogenic activity of Ras, e.g., a "farnesyl transferase inhibitor", e.g., L-744832, DK8G557 or Rl 15777 (Zarnestra).
[279] The term "telomerase inhibitor", as used herein, refers to compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, e.g., telomestatin.
62
SUBSTITUTE SHEET RULE 26 [280] The term "methionine aminopeptidase inhibitor", as used herein, refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase. Compounds which target, decrease or inhibit the activity of methionine aminopeptidase are, e.g., bengamide or a derivative thereof.
[281] The term "proteasome inhibitor", as used herein, refers to compounds which target, decrease or inhibit the activity of the proteasome. Compounds which target, decrease or inhibit the activity of the proteasome include, e.g., PS-341 and MLN 341.
[282] The term "matrix metalloproteinase inhibitor" or "MMP inhibitor", as used herein, includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g., hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ996.
[283] The term "agents used in the treatment of hematologic malignancies", as used herein, includes, but is not limited to, FMS-like tyrosine kinase inhibitors, e.g., compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1- b-D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors, e.g., compounds which target, decrease or inhibit anaplastic lymphoma kinase.
[284] Compounds which target, decrease or inhibit the activity of FMS-like tyrosine kinase receptors (Flt-3R) are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, e.g., PKC412, midostaurin, a staurosporine derivative, SU1 1248 and MLN518.
[285] The term "HSP90 inhibitors", as used herein, includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteasome pathway. Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90, e.g., 17- allylamino, 17-demethoxygeldanamycin (17AAG), a geldanamycin derivative, other geldanamycin related compounds, radicicol and HDAC inhibitors.
[286] The term "antiproliferative antibodies", as used herein, includes, but is not limited to, trastuzumab (HERCEPTIN™), Trastuzumab-DMl , erlotinib (TARCEVA™), bevacizumab (AVASTIN™), rituximab (RITUXAN®), PR064553 (anti-CD40) and 2C4 antibody. By antibodies is meant, e.g., intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least two intact antibodies, and antibodies fragments so long as they exhibit the desired
63
SUBSTITUTE SHEET RULE 26 biological activity. For the treatment of acute myeloid leukemia (AML), compounds disclosed herein can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML. In particular, compounds disclosed herein can be administered in combination with, e.g., farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
[287] A compound disclosed herein may also be used to advantage in combination with each other or in combination with other therapeutic agents, especially other anti-malarial agents. Such anti-malarial agents include, but are not limited to proguanil, chlorproguanil, trimethoprim, chloroquine, mefloquine, lumefantrine, atovaquone, pyrimethamine-sulfadoxine, pyrimethamine- dapsone, halofantrine, quinine, quinidine, amodiaquine, amopyroquine, sulphonamides, artemisinin, arteflene, artemether, artesunate, primaquine, inhaled NO, L-arginine, Dipropylenetri- amine NONOate (NO donor), Rosiglitzone (PPAR-γ agonist), activated charcoal, Erythropoietin, Levamisole, and pyronaridine.
[288] A compound disclosed herein may also be used to advantage in combination with each other or in combination with other therapeutic agents, such as used for the treatment of Leishmaniosis, Trypanosomiasis, Toxoplasmosis and Neurocysticercosis. Such agents include, but are not limited to chloroquine sulfate, atovaquone-proguanil, artemether- lumefantrine, quinine- sulfate, artesunate, quinine, doxycycline, clindamycin, meglumine antimoniate, sodium stibogluconate, miltefosine, ketoconazole, pentamidine, amphotericin B (AmB), liposomal- AmB, paromomycine, eflornithine, nifurtimox, suramin, melarsoprol, prednisolone, benznidazole, sulfadiazine, pyrimethamine, clindamycin, trimetropim, sulfamethoxazole, azitromycin, atovaquone, dexamethasone, praziquantel, albendazole, beta-lactams, fluoroquinolones, macrolides, aminoglycosides, sulfadiazine and pyrimethamine.
[289] The structure of the active agents identified by code nos., generic or trade names may be taken from the actual edition of the standard compendium "The Merck Index" or from databases, e.g., Patents International, e.g., IMS World Publications.
[290] The above-mentioned compounds, which can be used in combination with a compound disclosed herein, can be prepared and administered as described in the art, such as in the documents cited above.
[291] A compound disclosed herein may also be used to advantage in combination with known therapeutic processes, e.g., the administration of hormones or especially radiation. A compound
64
SUBSTITUTE SHEET RULE 26 disclosed herein may in particular be used as a radiosensitizer, especially for the treatment of tumors which exhibit poor sensitivity to radiotherapy.
[292] By "combination", there is meant either a fixed combination in one dosage unit form, or a kit of parts for the combined administration where a compound disclosed herein and a combination partner may be administered independently at the same time or separately within time intervals that especially allow that the combination partners show a cooperative, e.g., synergistic, effect or any combination thereof. The terms "coadministration" or "combined administration" or the like as utilized herein are meant to encompass administration of the selected combination partner to a single subject in need thereof (e.g. a patient), and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time. The term "pharmaceutical combination" as used herein means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non- fixed combinations of the active ingredients. The term "fixed combination" means that the active ingredients, e.g. a compound disclosed herein and a combination partner, are both administered to a patient simultaneously in the form of a single entity or dosage. The term "non-fixed combination" means that the active ingredients, e.g. a compound disclosed herein and a combination partner, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.
METHODS OF TREATMENT
[293] In one embodiment, the methods of treatment disclosed herein comprise administering a safe and effective amount of a compound or a pharmaceutically composition disclosed herein to a patient in need thereof. Individual embodiments disclosed herein include methods of treating any one of the above-mentioned disorders by administering a safe and effective amount of a compound disclosed herein or a pharmaceutical composition containing a compound disclosed herein to a patient in need thereof.
[294] In one embodiment, the compounds disclosed herein or pharmaceutically compositions containing the compounds disclosed herein may be administered by any suitable route of administration, including both systemic administration and topical administration. Systemic administration includes oral administration, parenteral administration, transdermal administration and rectal administration. Parenteral administration is typically by injection or infusion, including intravenous, intramuscular, and subcutaneous injection or infusion. Topical administration includes application to the skin as well as intraocular, otic, intravaginal, inhaled and intranasal
65
SUBSTITUTE SHEET RULE 26 administration. In one embodiment, the compounds disclosed herein or pharmaceutical compositions containing the compounds disclosed herein may be administered orally. In another embodiment, the compounds disclosed herein or pharmaceutically compositions containing the compounds disclosed herein may be administered by inhalation. In a further embodiment, the compounds disclosed herein or pharmaceutical compositions containing the compounds disclosed herein may be administered intranasally.
[295] In another embodiment, the compounds disclosed herein or pharmaceutically compositions containing the compounds disclosed herein may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. For example, doses may be administered one, two, three, or four times per day. In one embodiment, a dose is administered once per day. In a further embodiment, a dose is administered twice per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect. Suitable dosing regimens for a compound disclosed herein or a pharmaceutical composition containing a compound disclosed herein depend on the pharmacokinetic properties of that compound, such as absorption, distribution, and half-life, which can be determined by the skilled artisan. In addition, suitable dosing regimens, including the duration such regimens are administered, for a compound disclosed herein or a pharmaceutical composition containing a compound disclosed herein depend on the disorder being treated, the severity of the disorder being treated, the age and physical condition of the patient being treated, the medical history of the patient to be treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual patient's response to the dosing regimen or over time as individual patient needs change.
[296] The compound of the present invention may be administered either simultaneously with, or before or after, one or more other therapeutic agent. The compound of the present invention may be administered separately, by the same or different route of administration, or together in the same pharmaceutical composition as the other agents.
[297] The pharmaceutical composition or combination of the present invention can be in unit dosage of about 1-1000 mg of active ingredient(s) for a subject of about 50-70 kg, or about 1-500 mg or about 1-250 mg or about 1-150 mg or about 0.5-100 mg, or about 1- 50 mg of active ingredients. The therapeutically effective dosage of a compound, the pharmaceutical composition, or the combinations thereof, is dependent on the species of the subject, the body weight, age and individual condition, the disorder or disease or the severity thereof being treated. A physician,
66
SUBSTITUTE SHEET RULE 26 clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to prevent, treat or inhibit the progress of the disorder or disease. The above-cited dosage properties are demonstrable in vitro and in vivo tests using advantageously mammals, e.g., mice, rats, dogs, monkeys or isolated organs, tissues and preparations thereof. The compounds of the present invention can be applied in vitro in the form of solutions, e.g., aqueous solutions, and in vivo either enterally, parenterally, advantageously intravenously, e.g., as a suspension or in aqueous solution.
[298] In one embodiment, the therapeutically effective dose is from about 0.1 mg to about 2,000 mg per day of a compound provided herein. The pharmaceutical compositions therefore should provide a dosage of from about 0.1 mg to about 2000 mg of the compound. In certain embodiments, pharmaceutical dosage unit forms are prepared to provide from about 1 mg to about 2000 mg, from about 10 mg to about 1000 mg, from about 20 mg to about 500 mg or from about 25 mg to about 250 mg of the essential active ingredient or a combination of essential ingredients per dosage unit form. In certain embodiments, the pharmaceutical dosage unit forms are prepared to provide about 10 mg, 20 mg, 25 mg, 50 mg, 100 mg, 250 mg, 500 mg, 1000 mg or 2000 mg of the essential active ingredient.
[299] Additionally, the compounds disclosed herein may be administered as prodrugs. As used herein, a "prodrug" of a compound disclosed herein is a functional derivative of the compound which, upon administration to a patient, eventually liberates the compound disclosed herein in vivo. Administration of a compound disclosed herein as a prodrug may enable the skilled artisan to do one or more of the following: (a) modify the onset of the activity of the compound in vivo; (b) modify the duration of action of the compound in vivo; (c) modify the transportation or distribution of the compound in vivo; (d) modify the solubility of the compound in vivo; and (e) overcome a side effect or other difficulty encountered with the compound. Typical functional derivatives used to prepare prodrugs include modifications of the compound that are chemically or enzymatically cleavable in vivo. Such modifications, which include the preparation of phosphates, amides, esters, thioesters, carbonates, and carbamates, are well known to those skilled in the art.
GENERAL SYNTHETIC PROCEDURES
[300] In order to illustrate the invention, the following examples are included. However, it is to be understood that these examples do not limit the invention and are only meant to suggest a method of practicing the invention.
[301] Generally, the compounds in this invention may be prepared by methods described herein, wherein the substituents are as defined for Formula (I), above, except where further noted. The
67
SUBSTITUTE SHEET RULE 26 following non-limiting schemes and examples are presented to further exemplify the invention. Persons skilled in the art will recognize that the chemical reactions described herein may be readily adapted to prepare a number of other compounds of the invention, and alternative methods for preparing the compounds of this invention are deemed to be within the scope of this invention. For example, the synthesis of non-exemplified compounds according to the invention may be successfully performed by modifications apparent to those skilled in the art, e.g., by appropriately protecting interfering groups, by utilizing other suitable reagents known in the art other than those described, and/or by making routine modifications of reaction conditions. Alternatively, other reactions disclosed herein or known in the art will be recognized as having applicability for preparing other compounds of the invention.
[302] In the examples described below, unless otherwise indicated all temperatures are set forth in degrees Celsius. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company, Shanghai Medpep. Co Ltd, Aladdin-Shanghai Jinchun Reagents, Ltd, and were used without further purification unless otherwise indicated. Common solvents were purchased from commercial suppliers such as Shantou XiLong Chemical Factory, Guangdong Guanghua Reagent Chemical Factory Co. Ltd., Guangzhou Reagent Chemical Factory, Tainjin YuYu Fine Chemical Ltd., Qingdao Tenglong Reagent Chemical Ltd., and Qingdao Ocean Chemical Factory.
[303] Anhydrous THF, dioxane, toluene, and ether were obtained by refluxing the solvent with sodium. Anhydrous CH2CI2 and CHCI3 were obtained by refluxing the solvent with CaH2. EtOAc, PE, hexanes, DMA and DMF were treated with anhydrous Na2S04 prior use.
[304] The reactions set forth below were done generally under a positive pressure of nitrogen or argon or with a drying tube (unless otherwise stated) in anhydrous solvents, and the reaction flasks were typically fitted with rubber septa for the introduction of substrates and reagents via syringe. Glassware was oven dried and/or heat dried.
[305] Column chromatography was conducted using a silica gel column. Silica gel (300 - 400 mesh) was purchased from Qingdao Ocean Chemical Factory.
[306] lH NMR spectra were recorded with a Bruker 400 MHz or 600 MHz spectrometer at ambient temperature. !H NMR spectra were obtained as CDCI3, DMSO- i, CD3OD or acetone-<i6 solutions (reported in ppm), using TMS (0 ppm) or chloroform (7.26 ppm) as the reference standard. When peak multiplicities are reported, the following abbreviations are used: s (singlet), d (doublet), t (triplet), m (multiplet), br (broadened), dd (doublet of doublets), dt (doublet of triplets), td (triplet of doublets). Coupling constants, when given, are reported in Hertz (Hz).
68
SUBSTITUTE SHEET RULE 26 [307] Low -resolution mass spectral (MS) data were generally determined on an Agilent 6120 quadrupole HPLC-MS (Zorbax SB-C18, 2.1 x 30 mm, 3.5 micron, 6 minutes run, 0.6 mL/min flow rate, 5% to 95% (0.1% formic acid in C¾CN) in (0.1% formic acid in H20)) with UV detection at 210/254 nm and electrospray ionization (ESI).
[308] Purities of compounds were assessed by Agilent 1260 pre-HPLC or Calesep pump 250 pre-HPLC (column: NOVASEP 50/80 mm DAC) with UV detection at 210 nm and 254 nm.
[309] The following abbreviations are used throughout the specification:
AcOH, HAc, CH3COOH acetic acid
Ac20 acetic anhydride
ACN, MeCN, CH3CN acetonitrile
Boc20 di-tert-butyl dicarbonate
BOC, Boc butyloxycarbonyl
«-BuOH «-butyl alcohol
Cbz-Cl benzyl chloroformate
CH2C12, DCM methylene chloride
CDCI3 chloroform deuterated
DIEA, DIPEA, z'-Pr2NEt N,N-diisopropylethylamine
DMF dimethylformamide
DMAP 4-dimethylaminopyridine
DMSO dimethylsulfoxide
EDC, EDCI l-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
EDTA ethylenediaminetetraacetic acid
EtOAc, EA ethyl acetate
Et20 ether g gram
h hour
HATU 2-(7-aza- 1 H-benzotriazole- 1 -yl)- 1 , 1 ,3 ,3 -tetramethyluronium hexafluorophosphate HC1 hydrochloric acid HO AT 1 -hydroxy-7-azabenzotriazole
HBTU 2-( 1 H-benzotriazole- 1 -yl)- 1 , 1 ,3 ,3 -tetramethyluronium hexafluorophosphate
HATU 2-(7-aza- 1 H-benzotriazole- 1 -yl)- 1 , 1 ,3 ,3-tetramethyluronium hexafluorophosphate 3PO4 potassium phosphate
ΚΟΗ potassium hydroxide
LiOH lithium hydroxide
mL, ml milliliter min minute
MeOH methanol
M mol/L
NaHCC"3 sodium bicarbonate
NH4CI ammonium chloride Na2C03 sodium carbonate Na2SC"4 sodium sulfate
NaOH sodium hydroxide
Pd/C palladium on carbon Pd(OH)2 palladium hydroxide PE petroleum ether (60-90 °C)
POBr3 phosphorus oxybromide
POCI3 phosphorus oxychloride
PdCl2(dppf).DCM, PdCl2(dppf).CH2Cl2 1, l'-bis(diphenylphosphino)ferrocene- palladium(II)dichloride dichloromethane complex
RT, rt, r.t. room temperature
Rt retention time
TFA trifluoroacetic acid
TEA, Et3N triethylamine
[310] Representative synthetic procedures for the preparation of the compounds disclosed herein is outlined below in following Scheme 1~ Scheme 5. Unless otherwise indicated, each of Z and Z carry the definitions set forth above in connection with Formula (I) and p is 1, 2 or 3.
Scheme 1 :
(8) (10) (11)
[311] Some compounds having formula (11) can be prepared by a general method illustrated in Scheme 1 and described in details in the examples. As showing in Scheme 1, ethyl 2-cyanoacetate (I) is reacted with 2,2,2-trichloroacetonitrile (2) with an aid of a base, such as Et3N, to give compound Q). Compound (3) is then treated with hydrazine in water to give azole (4). Condensation of azole (4) with compound (5) under an acidic condition provides amino azole (6) at elvated temperature (ranging from 50 °C to 200 °C). Amine (6) is reacted with (Boc)20 in the presence of a base such as Na2C03, NaHC03 or Et3N to give N-protected compound (7). The ester group in compound (7) is then converted to carboxylic acid (8) through basic hydrolysis. Compound (8) is allowed to condense with amine compound (9) to provied amide (10). Final removal of the Boc-protecting group in compound (10) under acidic conditions, such as trifluoroacetic acid (TFA) in DCM, or HC1 in EtOAc affords the desired kinase inhibitor (11).
Scheme 2:
71
SUBSTITUTE SHEET RULE 26
Figure imgf000073_0001
Figure imgf000073_0002
Figure imgf000073_0003
[312] Some compounds having formula (20) can be prepared by a general method illustrated in Scheme 2 and described in details in the examples. As showing in Scheme 2, substituted azole (4) condense with 1,3-dimethyluracil (12) under an alkaline condition provides compound (13). Halogenation of compound (13) by POBr3 affords amino azole (14), amino azole (14) is reacted with (Boc)20 in the presence of a base such as Na2C03, DMAP or Et3N to give N-protected compound (15). The compound (15) is couple with borate acid (16) in the presence of a suitable Pd catalyst, such as PdCl2(dppf).DCM, affords compound (17). The ester group in compound (17) is then converted to carboxylic acid (18) through basic hydrolysis. Compound (18) is allowed to condense with amine compound (9) to pro vied amide (19). Final removal of the Boc-protecting group in compound (19) under acidic conditions, such as trifluoroacetic acid (TFA) in DCM, or HC1 in EtOAc affords the desired kinase inhibitor (20).
Scheme 3:
72
SUBSTITUTE SHEET RULE 26
Figure imgf000074_0001
Figure imgf000074_0002
Figure imgf000074_0003
(25) (26)
[313] Some compounds having formula (26) can be prepared by a general method illustrated in Scheme 3 and described in details in the examples. As showing in Scheme 3, compound (13) is halogenated by POCI3 affords amino azole (21), amino azole (21) is reacted with (Boc)20 in the presence of a base such as Na2C03, DMAP or Et3N to give N-protected compound (22). The compound (22) is reacted with MeONa in MeOH, affords compound (23). The ester group in compound (23) is then converted to carboxylic acid (24) through basic hydrolysis. Compound (24) is allowed to condense with amine compound (9) to provied amide (25). Final removal of the Boc- protecting group in compound (25) under acidic conditions, such as trifluoroacetic acid (TFA) in DCM, or HC1 in EtOAc affords the desired kinase inhibitor (26).
Scheme 4:
Figure imgf000074_0004
(29) (30)
73
SUBSTITUTE SHEET RULE 26
Figure imgf000075_0001
(21) (321 (33)
[314] Some compounds having formula (33) can be prepared by a general method illustrated in Scheme 4 and described in details in the examples. As showing in Scheme 4, ethyl 2-cyanoacetate (1) is reacted with 1 ,1 ,3,3-tetramethoxypropane (5) at elvated temperature to give (Z)-ethyl 2- cyano-3-ethoxyacrylate (27) is reacted with hydrazine in water and alcohol to give azole (28). Azole (28) condensed with 1 ,3-dimethyluracil (12) under an alkaline condition provides compound (29). Halogenation of compound (29) by POBr3 affords azole (30), azole (30) is couple with borate acid (16) in the presence of a suitable Pd catalyst, such as PdCl2(dppf).DCM, affords compound (31). The ester group in compound (31) is then converted to carboxylic acid (32) through basic hydrolysis. Compound (32) is allowed to condense with amine compound (9) to affords the desired kinase inhibitor (33).
Scheme 5:
Figure imgf000075_0002
(28) (24) (25) 126)
[315] Some compounds having formula (36) can be prepared by a general method illustrated in Scheme 5 and described in details in the examples. As showing in Scheme 5, Condensation of azole (28) with compound (5) under an acidic condition provides amino azole (34) at elvated temperature. The ester group in compound (34) is then converted to carboxylic acid (35) through basic hydrolysis. Compound (35) is allowed to condense with amine compound (9) to affords the desired kinase inhibitor (36).
EXAMPLES
Example 1 2-amino-N-(piperidin-3-yl)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide
74
SUBSTITUTE SHEET RULE 26
Figure imgf000076_0001
Step 1) ( )-ethyl 3-amino-4,4,4-trichloro-2-cyanobut-2-enoate
[316] To a solution of ethyl 2-cyanoacetate (41.22 g, 364.41 mmol) and 2,2,2- trichloroacetonitrile (100.00 g, 692.58 mmol) in EtOH (120 mL) was added Et3N (2.00 g, 19.76 mmol). The reaction mixture was cooled to 0 °C and stirred for 2 h, then allowed to warm to room temperature and concentrated in vacuo. The residue was diluted with DCM (100 mL), and the resulting mixture was filtered through a pad of silica gel, eluting with DCM (1 L). The combined filtrates were concentrated in vacuo to give the title compound as a pale yellow solid (82.34 g, 87.7%).
MS (ESI, pos. ion) m/z: 257.0 [M+H]+;
¾ NMR (400 MHz, CDC1 ): δ (ppm) 10.22 (s, 1H), 6.86 (s, 1H), 4.33 (q, J = 7.12 Hz, 2H), 1.38 (t, J = 7.12 Hz, 3H).
Step 2) ethyl 3,5-diamino-lH-pyrazole-4-carboxylate
[317] To a solution of (Z)-ethyl 3-amino-4,4,4-trichloro-2-cyanobut-2-enoate (82.34 g, 319.77 mmol) in DMF (250 mL) was added a solution of hydrazine in water (50% [w/w], 46.08 g, 718.88 mmol). The reaction mixture was stirred at 100 °C for 2 h and concentrated in vacuo. The residue was diluted with DCM (100 mL), and the resulting mixture was stood overnight. The solid was collected by Alteration, and then washed with DCM (50 mL x 3) to give the title compound as a pale yellow solid (40.40 g, 74.2%).
MS (ESI, pos. ion) m/z: 171.0 [M+H]+; lH NMR (400 MHz, OMSO-ds): δ (ppm) 5.35 (s, 4H), 4.14 (q, J = 7.12 Hz, 2H), 1.24 (t, J =7.12 Hz, 3H).
Step 3) ethyl 2-aminopyrazolo[l ,5-a]pyrimidine-3-carboxylate
[318] To a solution of ethyl 3,5-diamino-lH-pyrazole-4-carboxylate (5.00 g, 29.38 mmol) in DMF (80 mL) were added 1 , 1 ,3,3-tetramethoxypropane (14.50 mL, 88.15 mmol) and AcOH (0.34 mL, 5.88 mmol). The reaction mixture was stirred at 100 °C for 14 h, and then concentrated in vacuo. The residue was partitioned between DCM (50 mL) and water (50 mL). The organic phase was separated and the aqueous phase was extracted with DCM (100 mL x 3). The combined
75
SUBSTITUTE SHEET RULE 26 organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (a solution of NH3 in MeOH (7 M)/DCM (v/v) =1/100) to give the title compound as a pale yellow solid (3.52 g, 58.1%).
MS (ESI, pos. ion) m/z: 207.1 [M+H]+;
!H NMR (400 MHz, CDC1 ): δ (ppm) 8.60 (dd, J= 4.40 Hz, 1.76 Hz, 1H), 8.46 (dd, J = 6.76 Hz, 1.76 Hz, 1H), 6.86 (dd, J = 6.72 Hz, 4.40 Hz, 1H), 4.50 (q, J = 7.08 Hz, 2H), 1.47 (t, J = 7.08 Hz, 3H).
Step 4) ethyl 2-((tert-butoxycarbonyl)amino)pyrazolori,5-a1pyrimidine-3-carboxylate
[319] To a suspension of ethyl 2-aminopyrazolo[l ,5-a]pyrimidine-3-carboxylate (1.60 g, 7.76 mmol), DMAP (0.19 g, 1.55 mmol) and Et3N (3.25 mL, 23.28 mmol) in DCM (15 mL) was added Boc20 (3.30 mL, 15.52 mmol). The reaction mixture was stirred at room temperature for 3 h and then concentrated in vacuo. The residue was purified by a silica gel column chromatography (100% DCM) to give the title compound as a yellow solid (1.24 g, 52.1%).
MS (ESI, pos. ion) m/z: 251.0 [(M-C4H8)+H]+;
!H NMR (400 MHz, CDC1 ): δ (ppm) 8.87 (dd, J= 4.20 Hz, 1.80 Hz, 1H), 8.73 (dd, J = 7.00 Hz, 1.84 Hz, 1H), 7.16 (dd, J = 6.96 Hz, 4.16 Hz, 1H), 4.38 (q, J = 7.04 Hz, 2H), 1.45 (s, 9H), 1.35 (t, J= 7.12 Hz, 3H).
Step 5) 2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-alpyrimidine-3-carboxylic acid
[320] To a suspension of ethyl 2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3- carboxylate (1.24 g, 4.05 mmol) in EtOH (75 mL) was added a solution of LiOH in water (10% [w/w], 20 mL, 83.30 mmol). The reaction mixture was stirred at 80 °C overnight, then cooled to room temperature and quenched with a solution of citric acid in water (10% [w/w], 75 mL). The mixture was concentrated in vacuo, and the resulting residue was partitioned between EtOAc (50 mL) and a saturated aqueous solution of citric acid (50 mL). The organic phase was separated and the aqueous layer was extracted with EtOAc (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo to give the title compound as a pale yellow solid (0.91 g, 80.5%>).
MS (ESI, pos. ion) m/z: 223.2 [M-C4H8+H]+;
!H NMR (400 MHz, CDC1 ): δ (ppm) 8.99 (s, 1H), 8.85 (dd, J= 6.84 Hz, 1.64 Hz, 1H), 8.67 (dd, J = 4.40 Hz, 1.64 Hz, 1H), 7.07 (dd, J= 6.84 Hz, 4.40 Hz, 1H), 1.59 (s, 9H).
76
SUBSTITUTE SHEET RULE 26 Step 6) fert-butyl 3-(2-((tert-butoxycarbonyl)amino)pyrazolori,5-a1pyrimidine-3- carboxamido)piperidine- 1 -carboxylate
[321 ] To a suspension of 2-((tert-butoxycarbonyl)amino)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxylic acid (200 mg, 0.72 mmol) and tert-butyl 4-aminopiperidine-l -carboxylate (144 mg, 0.72 mmol) in DCM (8 mL) was added HATU (334 mg, 0.88 mmol) and Et3N (0.2 mL, 1.44 mmol). The reaction mixture was stirred at room temperature overnight and quenched with water (30 mL). The resulting mixture was extracted with EtOAc (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/PE (v/v) =1/2) to give the title compound as a pale yellow solid (300 mg, 90.5%).
MS (ESI, pos. ion) m/z: 461.0 [M+H]+;
!H NMR (600 MHz, CDC1 ): δ (ppm) 9.89 (s, 1H), 8.77 (dd, J = 6.8, 1.5 Hz, 1H), 8.53 (s, 1H), 7.96 (s, 1H), 6.92 (dd, J = 6.6, 4.4 Hz, 1H), 4.20 (s, 1H), 3.63 (m, 4H), 1.98 (s, 1H), 1.76 (m, 2H), 1.60 (s, 9H), 1.56 (s, 10H).
Step 7) 2-amino-N-(piperidin-3-yl)pyrazolo[ 1 ,5-alpyrimidine-3-carboxamide
[322] To a solution of tert- vXy\ 3-(2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3- carboxamido)piperidine-l -carboxylate (300 mg, 0.65 mmol) in DCM (10 mL) was added a solution of HCl in EtOAc (4 M, 10 mL, 40 mmol). The reaction mixture was stirred at room temperature overnight, and then concentrated in vacuo. The residue was dissolved in water (30 mL), and the resulting mixture was adjusted to pH = 10 with a saturated Na2C03 aqueous solution and then extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (a solution of NH3 in MeOH (7 M)/MeOH/DCM (v/v/v) = 2/100/800) to give the title compound as a beige solid (170 mg, 100%).
MS (ESI, pos. ion) m/z: 261.1 [M+H]+;
!H NMR (600 MHz, DMSO-Λ): δ (ppm) 8.90 (dd, J= 6.7, 1.6 Hz, 1H), 8.50 (dd, J = 4.4, 1.6 Hz, 1H), 7.82 (d, J= 7.4 Hz, 1H), 6.97 (dd, J= 6.7, 4.5 Hz, 1H), 6.51 (s, 2H), 3.93 (m, 1H), 2.99 (dd, J = 11.5, 2.8 Hz, 1H), 2.75 (m, 1H), 2.61 (m, 1H), 2.51 (m, 1H), 1.90 (s, 1H), 1.83 (br s, 1H), 1.64 (m, 1H), 1.48 (m, 2H).
Example 2 2-amino-N-(l-(2-cvanoacetyl)piperidin-3-yl)pyrazolo[l,5-a1pyrimidine-3-carboxamide
77
SUBSTITUTE SHEET RULE 26
Figure imgf000079_0001
[323] To a suspension of 2-amino-N-(piperidin-3-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (0.14 g, 0.55 mmol) and 2-cyanoacetic acid (52 mg, 0.60 mmol) in DCM (3 mL) were added HATU (0.26 g, 0.66 mmol) and Et3N (0.11 g, 1.09 mmol). The reaction mixture was stirred at room temperature overnight, and then diluted with DCM (50 mL). The mixture was washed with water (20 mLx2) followed by brine (30 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica chromatography (DCM/MeOH (v/v) = 30/1) to give the title compound as a light yellow solid (52 mg , 28%).
MS (ESI, pos. ion) m/z: 328.0 [M+H]+;
¾ NMR (600 MHz, DMSO-Λ): δ (ppm) 8.95-8.89 (m, 1H), 8.48 (m, 1H), 7.79-7.73 (t, J= 4.5 Hz, 1H), 7.00 (m, 1H), 6.51 (d, J = 6.7 Hz, 2H), 4.09 (m, 1H), 3.98 (m, 1H), 3.95 (m, 1H), 3.65 (m, 1H), 3.44 (m, 1H), 3.29-3.21 (m, 1H), 3.16 (m, 1H), 1.92 (m, 1H), 1.73 (m, 1H), 1.70-1.63 (m, 1H), 1.50 (m, 1H).
Example 3 2-amino-N-(l-(5-cvanopyridin-2-yl)piperidin-4-yl)pyrazolori,5-a1pyrimidine-3- carboxamide trifluoroacetic acid salt
Figure imgf000079_0002
Step 1) fert-butyl (l-(5-cvanopyridin-2-yl)piperidin-4-yl)carbamate
[324] A solution of tert- vXy\ piperidin-4-ylcarbamate (3.00 g, 15.00 mmol), 6- chloronicotinonitrile (2.08 g, 15.00 mmol) and Na2C03 (3.20 g, 30.19 mmol) in DMF (40 mL) was heated to 90 °C and stirred for 4 h. The reaction mixture was cooled to room temperature, diluted with water (120 mL), and extracted with EtOAc (100 mL x 3). The combined organic phases were washed with brine (300 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo.
78
SUBSTITUTE SHEET RULE 26 The residue was washed with PE/EtOAc (10/1 (v/v), 80 mL) to give the title compound as a white solid (4.50 g, 99 %).
MS ( ESI, pos. ion) m/z: 247.0 [M-C4H8 + H]+;
!H NMR (400 MHz, CDCb): δ (ppm) 8.40 (d, J = 2.36 Hz, 1H), 7.62-7.59 (dd, J = 2.36 Hz, 9.08 Hz, 1H), 6.63 (d, J = 9.08 Hz, 1H), 4.45 (m, 1H), 4.36-4.33 (m, 2H), 3.75 (m, 1H), 3.14-3.07 (m, 2H), 2.08-2.06 (m, 2H), 1.45 (s, 9H), 1.43-1.37 (m, 2H).
Step 2) 6-(4-aminopiperidin-l-yl)nicotinonitrile hydrochloric
[325] To a solution of tert-butyl (l-(5-cyanopyridin-2-yl)piperidin-4-yl)carbamate (4.00 g, 13.23 mmol) in DCM (20 mL) was added a solution of HC1 in EtOAc (3.5 M, 4 mL) at 0 °C dropwise. The reaction mixture was allowed to warm to room temperature and stirred overnight. The mixture was concentrated in vacuo to afford the title compound as a white solid (3.13 g, 99 %).
MS (ESI, pos. ion) m/z: 203.0 [M+H]+.
Step 3) tert-butyl (3-((l-(5-cvanopyridin-2-yl)piperidin-4-yl)carbamoyl)pyrazolori,5-a1pyrimidin- 2-yl)carbamate
[326] To a suspension of 2-((tert-butoxycarbonyl)amino)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxylic acid (0.10 g, 0.36 mmol) in DCM (5.0 mL) was added 6-(4-aminopiperidin-l-yl)nicotinonitrile hydrochloric (99 mg, 0.41 mmol), HATU (0.16 g, 0.43 mmol) and Et3N (0.06 mL, 0.43 mmol). The reaction mixture was stirred at room temperature overnight, and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/PE (v/v) =1/2) to give the title compound as a light yellow solid (0.11 g, 66%).
MS (ESI, pos. ion) m/z: 463.4 [M+H]+;
'H NMR (400 MHz, CDCb): δ (ppm) 9.81 (s, 1H), 8.78 (d, J= 6.7 Hz, 1H), 8.52 (m, 1H), 8.49 (s, 1H), 7.87 (d, J = 6.9 Hz, 1H), 7.72 (d, J= 8.5 Hz, 1H), 6.94 (dd, J = 6.7, 4.3 Hz, 1H), 6.80 (d, J = 8.8 Hz, 1H), 4.47 (m, 2H), 4.36 (m, 1H), 3.41 (m, 2H), 2.25 (m, 2H), 1.74 (m, 2H), 1.57 (s, 9H).
Step 4) 2-amino-N-(l-(5-cvanopyridin-2-yl)piperidin-4-yl)pyrazolo[l,5-a]pyrimidine-3- carboxamide trifluoroacetic acid salt
[327] To a suspension of tert-butyl (3-((l-(5-cyanopyridin-2-yl)piperidin-4- yl)carbamoyl)pyrazolo[l,5-a]pyrimidin-2-yl)carbamate (0.11 g, 0.24 mmol) in DCM (5mL) was added trifluoroacetic acid (3.0 mL). The reaction mixture was stirred at room temperature overnight, and concentrated in vacuo. The residue was diluted with MeOH/DCM (2 mL/3 mL), and the resulting mixture was stirred vigorously for another 15 min. The solid was collected by Alteration, and then dried in vacuo to give the title compound as a grey solid (28 mg, 25%).
79
SUBSTITUTE SHEET RULE 26 MS (ESI, pos. ion) m/z: 363.3 [M+H]+;
!H NMR (400 MHz, OMSO-de): δ (ppm) 8.90 (dd, J = 6.7, 1.5 Hz, 1H), 8.49 (d, J = 2.1 Hz, 1H), 8.47 (dd, J= 4.5, 1.5 Hz, 1H), 7.85 (dd, J= 9.1, 2.3 Hz, 1H),7.70 (d, J = 7.7 Hz, 1H), 7.02-6.94 (m, 2H), 6.5 l(s, 2H), 4.33 (m, 2H), 4.16 (m, 1H), 3.24 (m, 2H),2.04-1.94 (m, 2H), 1.48 (m, 2H).
Example 4 2-amino-N-(l-(5-cvanopyridin-2-yl)piperidin-3-yl)pyrazolori,5-a1pyrimidine-3- carboxamide
Figure imgf000081_0001
Step 1) fert-butyl (l-(5-cyanopyridin-2-yl)piperidin-3-yl)carbamate
[328] To a suspension of tert-butyl piperidin-3-yl carbamate (1.23 g, 6.14 mmol) in DMF (20 mL) were added 6-chloronicotinonitrile (0.85 g, 6.14 mmol) and Na2C03 (1.30 g, 12.28 mmol). The reaction mixture was stirred at room temperature for 10 h, then diluted with water (50 mL) and extracted with EtOAc (50 mL x 4). The combined organic phases were washed with brine (50 mL x 2), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/PE (v/v) = 1/3) to give the title compound as a white solid (1.79 g, 96%).
MS (ESI, pos. ion) m/z: 303.2 [M+H]+;
!H NMR (600 MHz, CDCls): δ (ppm) 8.40 (d, J = 2.2 Hz, 1H), 7.60 (dd, J= 9.0, 2.3 Hz, 1H), 6.70 (d, J = 8.8 Hz, 1H), 4.60 (m, 1H), 3.98 (m, 2H), 3.68 (m, 1H), 3.48 (m, 1H), 3.37 (m, 1H), 2.04- 1.92 (m, 1H), 1.86-1.70 (m, 1H), 1.72-1.63 (m, 1H), 1.51 (s, 9H).
Step 2) 6-(3-aminopiperidin-l-yl)nicotinonitrile
[329] To a suspension of tert-butyl (l-(5-cyanopyridin-2-yl)piperidin-3-yl)carbamate (1.79 g, 5.92 mmol) in DCM (20.0 mL) was added a solution of HC1 in EtOAc (3.0 M, 6.0 mL, 18 mmol). The reaction was stirred at room temperature overnight, then concentrated in vacuo. The residue was diluted with water (20 mL), and the resulting mixture was washed with DCM (30 mL x 2). The aqueous layer was treated with a saturated Na2C03 aqueous solution until pH = 9, and then extracted with DCM/MeOH (10/l(v/v), 30 mL x 3). The combined organic phases were washed
80
SUBSTITUTE SHEET RULE 26 with brine (50 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo to give the title compound as a light yellow solid (1.08 g, 90%).
MS (ESI, pos. ion) m/z: 203.1 [M+H]+;
1H NMR (600 MHz, CDCls): δ (ppm) 8.37 (d, J = 1.7 Hz, 1H), 7.56 (dd, J= 9.1, 2.3 Hz, 1H), 6.61 (d, J = 9.0 Hz, 1H), 4.27 (m, 1H), 4.09 (m, 1H), 3.14-3.00 (m, 1H), 2.91-2.77 (m, 2H), 2.09-1.95 (m, 1H), 1.82 (m, 1H), 1.65-1.47 (m, 1H), 1.43-1.31 (m, 1H).
Step 3) tert-butyl (3-((l-(5-cyanopyridin-2-yl)piperidin-3-yl)carbamoyl)pyrazolo[l,5-alpyrimidin- 2-yl)carbamate
[330] To a suspension of 2-((tert-butoxycarbonyl)amino)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxylic acid (0.20 g, 0.72 mmol) and 6-(3-aminopiperidin-l-yl)nicotinonitrile (0.15 g, 0.72 mmol) in DMF (2 mL) was added HATU (0.33 g, 0.86 mmol) and Et3N (0.20 mL, 1.44 mmol). The reaction mixture was stirred at room temperature overnight, and then diluted with DCM (50 mL). The mixture was washed with water (30 mL x 2) followed by brine (30 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) = 100/1) to give the title compound as a yellow solid (0.18 g, 54%).
MS (ESI, pos. ion) m/z: 463.1 [M+H]+;
!H NMR (600 MHz, CDC13): δ (ppm) 9.78 (s, 1H), 8.76 (dd, J = 6.9, 1.7 Hz, 1H), 8.41-8.39 (m, 1H), 8.36 (dd, J = 4.3, 1.7 Hz, 1H), 7.94 (d, J = 7.6 Hz, 1H), 7.57 (m, 1H), 6.92 (dd, J = 6.9, 4.3 Hz, 1H), 6.72 (t, J = 8.2 Hz, 1H), 4.30-4.19 (m, 1H), 4.15-4.07 (m, 1H), 3.70-3.58 (m, 2H), 2.16- 2.08 (m, 1H), 1.95-1.84 (m, 2H), 1.81-1.66 (m, 2H), 1.63 (s, 9H).
Step 4) 2-amino-N-(l-(5-cyanopyridin-2-yl)piperidin-3-yl)pyrazolo[l,5-alpyrimid carboxamide
[331] To a suspension of tert- vXy\ (3-((l-(5-cyanopyridin-2-yl)piperidin-3- yl)carbamoyl)pyrazolo[l,5-a]pyrimidin-2-yl)carbamate (0.18 g, 0.39 mmol) in DCM (5.0 mL) was added HCl (3.0 M in EtOAc, 5.0 mL, 15 mmol). The reaction mixture was stirred at room temperature for 4 h, then concentrated in vacuo. The residue was partitioned between saturated Na2C03 aqueous solution (10 mL) and DCM (10 mL), and the resulting mixture was stirred at room temperature for 30 min, then extracted with DCM (15 mL x 3). The combined organic phases were washed with brine (30 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) = 50/1) to give the title product as a yellow solid (76 mg, 54%).
81
SUBSTITUTE SHEET RULE 26 MS (ESI, pos. ion) m/z: 363.1 [M+H]+;
^ NMR ^OO MHz, CDCI3): δ (ppm) 8.43 (dd, J = 6.7, 1.4 Hz, 1H), 8.36 (d, J= 1.9 Hz, 1H), 8.21 (dd, J= 4.3, 1.4 Hz, 1H), 7.84 (d, J= 7.3 Hz, 1H), 7.53 (dd, J = 9.1, 2.2 Hz, 1H), 6.76 (dd, J= 6.6, 4.5 Hz, 1H), 6.71 (d, J= 9.1 Hz, 1H), 5.67 (s, 2H), 4.22 (m, 1H), 4.07 (m, 1H), 3.93 (m, 1H), 3.68- 3.62 (m, 1H), 3.57 (m, 1H), 2.14-2.07 (m, 1H), 1.87 (m, 3H).
Example 5 2-amino-N-(l-(2-cyanoacetyl)piperidin-4-yl)-N-methylpyrazolo[l,5-alpyri carboxamide
Figure imgf000083_0001
Step 1) fert-butyl 4-(methylamino)piperidine-l-carboxylate
[332] To a suspension of N-(tert-butoxycarbonyl)-4-piperidone (10.00 g, 50.19 mmol) in EtOH (75 mL) was added 10% Pd/C (1.00 g, 10% wt.), NH3Me (33% in ethanol, 27 mL, 0.20 mol) and HCOOH (46 mg, 1.00 mmol). The reaction mixture was sealed into an autoclave under 4 MPa H2 atmosphere and stirred at 50 °C overnight, then cooled to room temperature and filtered. The filtrate was concentrated in vacuo to give the title compound as light yellow liquid (10.70 g, 99%>).
MS (ESI, pos. ion) m/z: 215.3 [M+H]+;
'H NMR (400 MHz, CDC13): δ (ppm) 4.01 (m, 2H), 2.79 (m, 2H), 2.48 (m, 1H), 2.42 (s, 3H), 1.83 (m, 2H), 1.44 (s, 9H), 1.28-1.15 (m, 2H).
Step 2) fert-butyl 4-(((benzyloxy)carbonyl)(methyl)amino)piperidine-l-carboxylate
[333] To a suspension of tert- vXy\ 4-(methylamino)piperidine-l-carboxylate (5.00 g, 23.33 mmol) in 2 M NaOH (70 mL, 0.14 mmol) was added Cbz-Cl (4.50 mL, 32.66 mmol) slowly at 0 °C. The reaction mixture was allowed to warm to room temperature and stirred overnight. The solid was collected by filteration, then washed with PE (50 mL) and dried in vacuo to give the title compound as a white solid (7.00 g, 86%>).
MS (ESI, pos. ion) m/z: 371.3 [M+Na]+, 249.3 [M-Boc+H]+;
'H NMR (400 MHz, CDC1 ): δ (ppm) 7.46-7.29 (m, 5H), 5.17 (s, 2H), 4.21 (m, 3H), 2.82 (m, 5H), 1.63 (m, 4H), 1.48 (s, 9H).
82
SUBSTITUTE SHEET RULE 26 Step 3) benzyl methyl(piperidin-4-yl)carbamate trifluoroacetic acid salt
[334] To a suspension of tert-butyl 4-(((benzyloxy)carbonyl)(methyl)amino)piperidine-l- carboxylate (7.00 g, 20.09 mmol) in DCM (50 mL) was added TFA (6.00 mL, 80.36 mmol). The reaction mixture was stirred at room temperature for 4 h, and then concentrated in vacuo. The residue was diluted with Et20 (50 mL), and the resulting mixture was stirred at room temperature for another 15 min. The solid was collected by Alteration, and then dried in vacuo to give the title compound as a white solid (6.70 g, 89 %).
MS (ESI, pos. ion) m/z: 249.2 [M-TFA+H]+;
¾ NMR (400 MHz, CDCb): δ (ppm) 7.50-7.26 (m, 5H), 5.10 (s, 2H), 4.22-4.06 (m, 1H), 3.34 (m, 2H), 3.00 (m, 2H), 2.76 (s, 3H), 1.88 (m, 2H), 1.74 (m, 2H).
Step 4) benzyl (l-(2-cyanoacetyl)piperidin-4-yl)(methyl)carbamate
[335] To a suspension of benzyl methyl(piperidin-4-yl)carbamate trifluoroacetic acid salt (6.70 g, 18.49 mmol), cyanoacetic acid (1.73 g, 20.34 mmol), EDCI (4.25 g, 22.19 mmol) and HO AT (3.02 g, 22.19 mmol) in DCM (50 mL) was added Et3N (7.74 mL, 55.47 mmol). The reaction mixture was stirred at room temperature overnight, and then diluted with DCM (50 mL). The mixture was washed with a saturated NaHC03 aqueous solution (80 mL x 2), followed by water (80 mL) and brine (80 mL), then dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/PE (v/v) = 2/1) to give the title compound as a white solid (3.80 g, 65%).
MS (ESI, neg. ion) m/z: 314.1 [M-H] ~;
'H NMR (400 MHz, CDCb): δ (ppm) 7.43-7.30 (m, 5H), 5.16 (s, 2H), 4.76-4.61 (m, 1H), 4.30 (m, 1H), 3.86-3.69 (m, 1H), 3.62-3.42 (m, 2H), 3.24 (m, 1H), 2.82 (s, 3H), 2.68 (m 1H), 1.87-1.56 (m, 4H).
Step 5) 3-(4-(methylamino)piperidin-l-yl)-3-oxopropanenitrile
[336] To a suspension of benzyl (l-(2-cyanoacetyl)piperidin-4-yl)(methyl)carbamate (3.80 g, 12.05 mmol) in MeOH (40 mL) was added 10% Pd/C (0.76 g, 20% wt). The reaction mixture was stirred at 40 °C overnight in a nitrogen atmosphere, then filtered. The filtrate was concentrated in vacuo and the resulting residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) = 10/1) to give the title compound as a white solid (0.28 g, 12%>).
MS (ESI, pos. ion) m/z: 182.3 [M + H]+;
'H NMR (400 MHz, CDCb): δ (ppm) 4.42-4.29 (m, 1H), 3.79-3.66 (m, 1H), 3.51 (s, 2H), 3.23 (m, 1H), 3.01-2.89 (m, 1H), 2.73-2.59 (m, 1H), 2.47 (s, 3H), 1.98 (m, 2H), 1.57-1.40 (m, 2H).
83
SUBSTITUTE SHEET RULE 26 Step 6) fert-butyl (3-((l-(2-cvanoacetyl)piperidin-4-yl)(methyl)carbamoyl)pyrazolori,5- q]pyrimidin-2-yl)carbamate
[337] To a suspension of 2-((tert-butoxycarbonyl)amino)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxylic acid (0.15 g, 0.54 mmol) and 3-(4-(methylamino)piperidin-l-yl)-3-oxopropanenitrile (98 mg, 0.54 mmol) in anhydrous DCM (6 mL) was added HATU (0.25 g, 0.65 mmol) and Et3N (0.15 mL, 1.08 mmol). The reaction mixture was stirred at room temperature for 5 h, and then diluted with DCM (30 mL). The resulting mixture was washed with water (20 mL) followed by saturated NaHC03 aqueous solution (20 mL) and brine (20 mL), then dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) = 20/1) to give the title compound as a light yellow solid (0.23 g, 96%).
MS (ESI, pos. ion) m/z: 442.3 [M + H]+;
!H NMR (400 MHz, CDC1 ): δ (ppm) 8.98 (s, 1H), 8.76 (dd, J= 6.84 Hz, 1.40 Hz, 1H), 8.53 (dd, J = 4.00 Hz, 1.56 Hz, 1H), 6.90 (dd, J = 6.84 Hz, 4.24 Hz, 1H), 4.76 (m, 1H), 3.85 (m, 1H), 3.65- 3.44 (m, 2H), 3.23 (m, 1H), 3.05 (s, 3H), 2.74 (m, 1H), 2.16-1.72 (m, 4H), 1.56 (s, 9H).
Step 7) 2-amino-N-(l -(2-cyanoacetyl)piperidin-4-yl)-N-methylpyrazolo[ 1 ,5-a]pyrimidine-3- carboxamide
[338] To a suspension of tert- vXy\ (3-((l-(2-cyanoacetyl)piperidin-4-yl)(methyl)carbamoyl) pyrazolo[l,5-a]pyrimidin-2-yl)carbamate (0.23 g, 0.52 mmol) in DCM (5 mL) was added TFA (0.20 mL, 2.60 mmol). The reaction mixture was stirred at room temperature for 4 h, then partitioned between DCM (20 mL) and saturated NaHC03 aqueous solution (20 mL). The separated organic phase was washed with brine (20 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica chromatography (DCM/MeOH (v/v) = 25/1) to give the title compound as a light yellow solid (40 mg, 22%).
MS (ESI, pos. ion) m/z: 342.3 [M+H]+;
¾ NMR (400 MHz, CDC1 ): δ (ppm) 8.51-8.31 (m, 2H), 6.76 (dd, J= 6.76 Hz, 4.36 Hz, 1H), 5.32 (s, 2H), 4.75 (m, 1H), 4.56 (m, 1H), 3.84 (m, 1H), 3.62-3.44 (m, 2H), 3.28 (m, 1H), 3.04 (s, 3H), 2.71 (m, 1H), 2.05 (m, 2H), 1.84 (m, 2H).
Example 6 2-amino-N-(piperidin-4-yl)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide
84
SUBSTITUTE SHEET RULE 26
Figure imgf000086_0001
Step 1) fert-butyl 4-(2-((tert-butoxycarbonyl)amino)pyrazolon,5-a1pyrimidine-3- carboxamido)piperidine- 1 -carboxylate
[339] To a suspension of 2-((tert-butoxycarbonyl)amino)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxylic acid (200 mg, 0.72 mmol) and tert-butyl 4-aminopiperidine-l -carboxylate (144 mg, 0.72 mmol) in DCM (8 mL) was added HATU (334 mg, 0.88 mmol) and Et3N (0.2 mL, 1.44 mmol). The reaction mixture was stirred at room temperature for 5 h, then quenched with water (30 mL) and extracted with EtOAc (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/PE (v/v) = 1/2) to give the title compound as a pale yellow solid (300 mg, 90.5%).
MS (ESI, pos. ion) m/z: 461.3 [M+H]+;
¾ NMR (600 MHz, DMSO-Λ): δ (ppm) 9.79 (s, 1H), 9.19 (dd, J= 6.8, 1.5 Hz, 1H), 8.73 (dd, J = 4.3, 1.5 Hz, 1H), 7.79 (d, J= 7.7 Hz, 1H), 7.21 (dd, J= 6.8, 4.4 Hz, 1H), 4.01 (m, 1H), 3.87 (d, J = 10.9 Hz, 2H), 2.97 (br. s, 2H), 1.89 (dd, J = 12.5, 2.8 Hz, 2H), 1.50 (s, 9H), 1.44 (s, 1H), 1.42 (s, 10H).
Step 2) 2-amino-N-(piperidin-4-yl)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide
[340] To a solution of tert- vXy\ 4-(2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3- carboxamido)piperidine-l -carboxylate (300 mg, 0.65 mmol) in DCM (10 mL) was added HC1 (4 M in EtOAc, 10 mL, 40 mmol). The reaction mixture was stirred at room temperature for 8 h, then concentrated in vacuo. The residue was dissolved in water (30 mL) and adjusted to pH = 10 with saturated Na2C03 aqueous solution, then extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (a solution of NH3 in MeOH (7 M)/MeOH/DCM (v/v/v) =2/100/800) to give the title compound as a beige solid (70 mg, 41.4%).
MS (ESI, pos. ion) m/z: 261.1 [M+H]+;
85
SUBSTITUTE SHEET RULE 26 ¾ NMR (600 MHz, OMSO-de): δ (ppm) 8.91 (dd, J= 6.7, 1.5 Hz, 1H), 8.51 (dd, J = 4.5, 1.5 Hz, 1H), 7.65 (d, J = 7.9 Hz, 1H), 6.98 (dd, J = 6.7, 4.5 Hz, 1H), 6.50 (s, 2H), 3.90 (m, 1H), 2.95 (d, J = 12.6 Hz, 2H), 2.58 (m, 4H), 2.00 (m, 1H), 1.84 (d, J= 9.4 Hz, 2H).
Example 7 2-amino-N-(l-(2-cvanoacetyl)piperidin-4-yl)pyrazolori,5-a1pyrimidine-3-carboxam
Figure imgf000087_0001
[341] To a solution of 2-amino-N-(piperidin-4-yl)pyrazolo[l,5-a]pyrimidine-3-carboxamide (40 mg, 0.15 mmol) and 2-cyanoacetic acid (15 mg, 0.18 mmol) in DCM/DMF (2 mL/0.5 mL) was added HATU (68 mg, 0.18 mmol) and Et3N (30 mg, 0.30 mmol). The reaction mixture was stirred at room temperature for 2.5 h, then quenched with water (30 mL) and extracted with DCM (50 mL x 3). The combined organic phases were washed with brine (50 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/PE (v/v) =8/1) to give the title compound as a beige solid (40 mg, 81.6%).
MS (ESI, pos. ion) m/z: 328.0 [M+H]+;
!H NMR (600 MHz, OMSO-de): δ (ppm) 8.91 (dd, J= 6.7, 1.4 Hz, 1H), 8.49 (dd, J = 4.4, 1.4 Hz, 1H), 7.67 (d, J = 7.6 Hz, 1H), 6.98 (dd, J = 6.7, 4.5 Hz, 1H), 6.51 (s, 2H), 4.19 (d, J = 13.7 Hz, 1H), 4.07 (d, J = 3.3 Hz, 2H), 4.02 (d, J = 6.5 Hz, 1H), 3.64 (d, J = 14.1 Hz, 1H), 3.21 (m, 1H), 2.92 (m, 1H), 1.93 (m, 2H), 1.55 (m, 1H), 1.37 (m, 1H).
Example 8 2-amino-N-(3-hvdroxycvclohexyl)pyrazolon,5-a1pyrimidine-3- carboxamide
Figure imgf000087_0002
Step 1) tert-butyl (3-((3-hydroxycyclohexyl)carbamoyl)pyrazolo[l,5-alpyrimidin-2-yl)carbamate
[342] To a suspension of 2-((tert-butoxycarbonyl)amino)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxylic acid (200 mg, 0.72 mmol) and 3-aminocyclohexanol (83 mg, 0.72 mmol) in DCM (8 mL) was
86
SUBSTITUTE SHEET RULE 26 added HATU (334 mg, 0.88 mmol) and Et3N (0.2 mL, 1.44 mmol). The reaction mixture was stirred at room temperature overnight, then quenched with water (30 mL) and extracted with EtOAc (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filteted and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/PE (v/v) = 1/2) to give the title compound as a pale yellow solid (200 mg, 74.0%).
MS (ESI, pos. ion) m/z: 376.1 [M+H]+;
8.1 A (Rf = 0.5): ¾ NMR (600 MHz, CDC13) δ (ppm): 9.92 (s, 1H), 8.78 (dd, J= 6.8, 1.4 Hz, 1H),
8.55 (dd, J = 4.3, 1.4 Hz, 1H), 7.84 (d, J = 7.7 Hz, 1H), 6.92 (dd, J = 6.8, 4.4 Hz, 1H), 4.47 (m, 1H), 4.17 (m, 1H), 1.98 (m, 1H), 1.87 (m, 3H), 1.73 (m, 2H), 1.56 (s, 12H).
8.1 B (Rf = 0.4):1H NMR (600 MHz, CDC1 ) δ (ppm): 9.94 (s, 1H), 8.77 (dd, J= 6.9, 1.7 Hz, 1H), 8.54 (dd, J = 4.3, 1.6 Hz, 1H), 7.88 (d, J = 7.4 Hz, 1H), 6.92 (dd, J = 6.9, 4.3 Hz, 1H), 4.09 (m, 2H), 3.83 (ddd, J = 14.0, 9.9, 3.9 Hz, 1H), 2.35 (d, J = 11.9 Hz, 1H), 2.03 (m, 3H), 1.89 (m, 1H),
1.56 (s, 12H).
Step 2) 2-amino-N-(3-hydroxycyclohexyl)pyrazolo[ 1 ,5-alpyrimidine-3-carboxamide
[343] To a solution of tert- vXy\ (3-((3-hydroxycyclohexyl)carbamoyl)pyrazolo[l,5-a]pyrimidin- 2-yl)carbamate (200 mg, 0.53 mmol) in DCM (10 mL) was added HC1 (4 M in EtOAc, 8 mL, 32 mmol). The reaction mixture was stirred at room temperature overnight, and then concentrated in vacuo. The residue was dissolved in water (30 mL), and the resulting mixture was adjusted to pH = 10 with saturated Na2C03 aqueous solution and extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (MeOH/DCM (v/v) = 1/50) to give the title compound as a beige solid (75 mg, 51.4%).
MS (ESI, pos. ion) m/z: 276.1 [M+H]+;
8.2 A: !H NMR (600 MHz, CDC1 ): δ (ppm) 8.90 (dd, J = 6.7, 1.5 Hz, 1H), 8.50 (dd, J = 4.4, 1.5 Hz, 1H), 7.67 (d, J= 8.4 Hz, 1H), 6.97 (dd, J= 6.7, 4.5 Hz, 1H), 6.50 (s, 2H), 4.56 (d, J= 3.6 Hz, 1H), 4.26 (m, 1H), 3.88 (s, 1H), 2.00 (m, 1H), 1.71 (m, 3H), 1.63 (m, 1H), 1.55 (d, J = 11.3 Hz, 1H), 1.48 (m, 2H).
8.2 B: !H NMR (600 MHz, CDC1 ): δ (ppm) 8.90 (dd, J = 6.4, 1.2 Hz, 1H), 8.49 (m, 1H), 7.66 (d, J= 7.4 Hz, 1H), 6.97 (dd, J= 6.6, 4.5 Hz, 1H), 6.50 (s, 2H), 4.70 (d, J= 4.3 Hz, 1H), 3.84 (m, 1H), 2.10 (d, J = 11.4 Hz, 1H), 1.84 (d, J = 10.0 Hz, 1H), 1.80 (d, J = 11.6 Hz, 1H), 1.71 (m, 1H), 1.35 (d, J= 9.0 Hz, 1H), 1.15 (m, 4H).
87
SUBSTITUTE SHEET RULE 26 Example 9 2-amino-N-(pwolidin-3-yl)pyrazolo[l ,5-a]pyrimidine-3-carboxamide
Figure imgf000089_0001
Step fert-butyl 3-(2-((tert-butoxycarbonyl)amino)pyrazolori,5-a1pyrimidine-3- carboxamido)pyrrolidine- 1 -carboxylate
[344] To a suspension of 2-((tert-butoxycarbonyl)amino)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxylic acid (200 mg, 0.72 mmol) and tert-butyl 3 -aminopyrrolidine-1 -carboxylate (134 mg, 0.72 mmol) in DCM (8 mL) was added HATU (334 mg, 0.88 mmol) and Et3N (0.2 mL, 1.44 mmol). The reaction mixture was stirred at room temperature overnight, then quenched with water (30 mL), and extracted with EtOAc (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/PE (v/v) =1/2) to give the title compound as a white solid (300 mg, 93.3%).
MS (ESI, pos. ion) m/z: 447.0 [M+H]+;
!H NMR (600 MHz, CDC13): δ (ppm) 9.79 (s, 1H), 8.78 (d, J = 6.8 Hz, 1H), 8.54 (s, 1H), 7.96 (s, 1H), 6.94 (s, 1H), 4.68 (m, 1H), 3.76 (dd, J = 11.4, 6.4 Hz, 1H), 3.58 (m, 2H), 3.34 (m, 1H), 2.29 (m, 1H), 2.02 (m, 1H), 1.56 (s, 9H), 1.49 (s, 9H).
Step 2) 2-amino-N-(pyrrolidin-3-yl)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide
[345] To a solution of tert- vXy\ 3-(2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3- carboxamido)pyrrolidine-l -carboxylate (300 mg, 0.67 mmol) in DCM (10 mL) was added a solution of HCl in EtOAc (4 M, 10 mL, 40 mmol). The reaction mixture was stirred at room temperature overnight, and then concentrated in vacuo. The residue was dissolved in water (30 mL), and the resulting mixture was adjusted to pH 10 with saturated Na2C03 and extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (a solution of NH in MeOH (7 M)/MeOH/DCM (v/v/v) =1/50/500) to give the title compound as a yellow solid (166 mg, 100%).
MS (ESI, pos. ion) m/z: 247.1 [M+H]+;
88
SUBSTITUTE SHEET RULE 26 lH NMR (600 MHz, OMSO-ds): δ (ppm) 8.90 (d, J = 5.8 Hz, 1H), 8.50 (d, J = 3.2 Hz, 1H), 7.75 (d, J = 7.1 Hz, 1H), 6.98 (dd, J = 6.5, 4.6 Hz, 1H), 6.51 (s, 2H), 4.36 (m, 1H), 3.09 (dd, J = 10.9, 6.7 Hz, 1H), 2.94 (m, 1H), 2.83 (m, 1H), 2.65 (dd, J= 11.0, 4.4 Hz, 1H), 2.08 (dt, J= 13.9, 7.8 Hz, 1H), 1.99 (m, 1H), 1.59 (m, 1H).
Example 10 2-amino-N-(l -(5-cvanopyridin-2-yl)pyrrolidin-3-yl)pyrazolor 1 ,5-a1pyrimidine-3- carboxamide
Figure imgf000090_0001
Step 1) tert-butyl (l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamate
[346] To a suspension of tert-butyl pyrrolidin-3-ylcarbamate (1.14 g, 6.14 mmol) in DMF (10 mL) was added 6-chloronicotinonitrile (0.85 g, 6.14 mmol) and sodium carbonate (1.31 g, 1.23 mmol). The reaction mixture was stirred at room temperature overnight, diluted with water (40 mL) and then extracted with EtOAc (30 mL x 3). The combined organic phases were washed with brine (50 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EtOAc (v/v) = 4/1) to give the title compound as a white solid (1.32 g, 75%).
MS (ESI, pos. ion) m/z: 289.1 [M+H]+;
'H NMR (600 MHz, CDCls): δ (ppm) 8.38 (d, J= 1.9 Hz, 1H), 7.57 (dd, J= 8.9, 2.1 Hz, 1H), 6.33 (d, J = 8.9 Hz, 1H), 4.79-4.73 (m, 1H), 4.35 (s, 1H), 3.87-3.26 (m, 4H), 2.32-2.24 (m, 1H), 2.03- 1.96 (m, 1H), 1.43 (s, 9H).
Step 2) 6-(3-aminopyrrolidin-l-yl)nicotinonitrile
[347] To a solution of tert-butyl (l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamate (1.00 g, 3.47 mmol) in DCM (10 mL) was added a solution of HC1 in EtOAc (3 M, 10 mL). The reaction mixture was stirred at room temperature overnight, then diluted with saturated Na2C03 aqueous solution (40 mL) and extracted with DCM (30 mL x 3). The combined organic phases were washed with brine (50 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo to give the title compound as yellow oil (0.38 g, 58%).
89
SUBSTITUTE SHEET RULE 26 MS (ESI, pos. ion) m/z: 189.1 [M+H]+.
Step 3) tert-butyl (3-((l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamoyl)pyrazolo[l,5- q]pyrimidin-2-yl)carbamate
[348] To a solution of 2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (562 mg, 2.02 mmol) in DMF (5 mL) were added 6-(3-aminopyrrolidin-l-yl)nicotinonitrile (380 mg, 2.02 mmol), HATU (922 mg, 2.42 mmol) and Et3N (0.6 mL, 4.32 mmol). The reaction mixture was stirred at 45 °C overnight, diluted with water (20 mL) and then extracted with DCM (15 mL x 3). The combined organic phases were washed with brine (30 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) = 200/1) to give the title compound as a white solid (170 mg, 19%).
MS (ESI, pos. ion) m/z: 393.1 [M-C4H8+H)]+;
¾ NMR (600 MHz, CDC1 ): δ (ppm) 9.71 (s, 1H), 8.79-8.72 (m, 1H), 8.52-8.44 (m, 1H), 8.02 (d, J= 6.2 Hz, 1H), 7.62 (d, J = 8.9 Hz, 1H), 6.92 (dd, J = 6.5, 4.5 Hz, 1H), 6.41 (d, J = 8.8 Hz, 1H), 4.85-4.68 (m, 1H), 3.97-3.55 (m, 4H), 2.50-2.45 (m, 1H), 2.23-2.17 (m, 1H), 1.54 (s, 9H).
Step 4) 2-amino-N-(l-(5-cvanopyridin-2-yl)pwolidin-3-yl)pyrazolori,5-a1p imidine-3- carboxamide
[349] To a solution of tert- vXy\ (3-((l-(5-cyanopyridin-2-yl)pyrrolidin-3- yl)carbamoyl)pyrazolo[l,5-a]pyrimidin-2-yl)carbamate (170 mg, 0.38 mmol) in DCM/MeOH (2 mL/1 mL) was added a solution of HC1 in EtOAc (3 M, 1 mL). The reaction mixture was stirred at room temperature overnight, and then concentrated in vacuo. The residue was diluted with saturated Na2C03 (20 mL), and extracted with DCM (15 mL x 3). The combined organic phases were washed with brine (30 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel chromatography (DCM/MeOH (v/v) = 50/1) to give the title compound as a white solid (82 mg, 62%).
MS (ESI, pos. ion) m/z: 349.0 [M+H]+;
!H NMR (600 MHz, CDC1 ): δ (ppm) 8.50-8.42 (m, 1H), 8.37 (d, J = 3.0 Hz, 1H), 7.96 (d, J = 6.2 Hz, 1H), 7.63 (dd, J = 8.9, 2.1 Hz, 1H), 6.80 (dd, J = 6.7, 4.5 Hz, 1H), 6.41 (d, J = 8.9 Hz, 1H), 5.69 (s, 2H), 4.91-4.82 (m, 1H), 3.99-3.51 (m, 4H), 2.54-2.43 (m, 1H), 2.27-2.15 (m, 1H).
Example 11 2-amino-N-(l -(2-cvanoacetyl)p wolidin-3-yl)pyrazolo[ 1 ,5-a]pyrimidine-3- carboxamide
90
SUBSTITUTE SHEET RULE 26
Figure imgf000092_0001
Step 1) fert-butyl (l-(2-cvanoacetyl)pyrrolidin-3-yl)carbamate
[350] To a solution of tert-butyl pyrrolidin-3-ylcarbamate (1.86 g, 10.0 mmol) in DMF (59 mL) were added HATU (7.61 g, 12 mmol), z'-Pr2NEt (5.2 mL, 30 mmol) and cyanoacetic acid (1.02 g, 12.0 mmol). The reaction mixture was stirred at room temperature for 14 h, then concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAc/PE (v/v) = 2/3) to give the title compound as yellow oil (1.10 g, 43.5%).
MS (ESI, pos. ion) m/z: 198.1 [(M -C4H8)+H]+.
Step 2) tert-butyl (3-((l-(2-cyanoacetyl)pyrrolidin-3-yl)carbamoyl)pyrazolo[l,5-alpyrimidin-2- yPcarbamate
[351] To a solution of fert-butyl (l-(2-cyanoacetyl)pyrrolidin-3-yl)carbamate (253 mg, 1.0 mmol) in DCM (5 mL) was added a solution of HC1 in EtOAc (4 M, 5 mL, 20.0 mmol). The reaction mixture was stirred at room temperature for 4 h, and concentrated in vacuo to give the crude product which was used in the next step without further purification.
[352] The crude product was dissolved in DMF (8 mL), then to the solution were added HATU (570 mg, 1.5 mmol), z'-Pr2NEt (0.52 mL, 3.0 mmol) and 2-((tert- butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (333 mg, 1.2 mmol). The resulting mixture was stirred at room temperature for 12 h, concentrated in vacuo and the residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) =15/1) to give the title compound as yellow oil (60 mg, 14.5%).
MS (ESI, pos. ion) m/z: 358.1 [(M -C4H8)+H]+.
Step 3) 2-amino-N-(l -(2-cyanoacetyl)pyrrolidin-3-yl)pyrazolo[ 1 ,5-a]pyrimidine-3-carboxamide
[353] To a solution of tert- vXyl (3-((l-(2-cyanoacetyl)pyrrolidin-3-yl)carbamoyl)pyrazolo[l,5- a]pyrimidin-2-yl)carbamate (60 mg, 0.15 mmol) in DCM (3 mL) was added a solution of HC1 in EtOAc (4 M, 3 mL). The reaction mixture was stirred at room temperature for 3.5 h, then treated with saturated aqueous Na2C03 solution until pH = 10. The resulting mixture was extracted with DCM (50 mL x 2). The combined organic phases were dried over Na2S04, concentrated in vacuo,
91
SUBSTITUTE SHEET RULE 26 and the residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) =40/1) to give the title compound as a white solid (27 mg, 59%).
MS (ESI, pos. ion) m/z: 314.1 [M+H]+;
!H NMR (600 MHz, CDCb): δ (ppm) 8.91-8.92 (m, 1 H), 8.50 (s, 1 H), 7.78-7.83 (m, 1 H), 6.98- 7.01 (m, 1 H), 6.52 (s, 2 H), 4.56-4.59 (m, 1 H), 3.97-4.02 (m, 1 H), 3.67-3.81 (m, 1H), 3.34-3.58 (m, 2 H), 3.28-3.30( m, 1H), 2.18-2.28 (m, 1H), 1.89-2.03 (m,l H), 1.55-1.69 (m, 1H).
Example 12 2-amino-N-(l-(5-cyanopyridin-2-yl)azetidin-3-yl)pyrazolo[l,5-al pyrimidine-3- carboxamide
Figure imgf000093_0001
Step 1) fert-butyl (l-(5-cvanopyridin-2-yl)azetidin-3-yl)carbamate
[354] To a suspension of 6-chloronicotinonitrile (0.40 g, 2.89 mmol) in DMF (15 mL) was added tert-butyl azetidin-3-ylcarbamate (0.50 g, 2.89 mmol) and Na2C03 (0.61 g, 5.77 mmol). The mixture was stirred at room temperature overnight, then diluted with H20 (30 mL), extracted with EA (40 mLx5). The combined organic layer was washed with brine (50 mLx2), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EA/PE (v/v) = 1/4) to afford a light yellow solid (0.72 g, 91%).
MS (ESI, pos. ion) m/z: 275.2 [M+H]+;
'H NMR (400 MHz, CDC13): δ (ppm) 8.40 (d, J= 1.8 Hz, 1H), 7.61 (dd, J= 8.8, 2.2 Hz, 1H), 6.25 (d, J= 8.7 Hz, 1H), 5.03 (br, 1H), 4.66 (br, 1H), 4.44 ( m, 2H), 3.95 (m, 2H), 1.46 (s, 9H).
Step 2) 6-(3-aminoazetidin-l-yl)nicotinonitrile
[355] To a suspension of tert-butyl (l-(5-cyanopyridin-2-yl)azetidin-3-yl)carbamate (0.72 g, 2.62 mmol) in DCM (10 mL) was added a solution of HC1 in EA (10 mL, 3.5 M). The mixture was stirred at room temperature overnight, then concentrated in vacuo. The reidue was diluted with DCM (20 mL) and saturated Na2C03 aqueous solution (20 mL). The organic layer was separated and the aqueous layer was extracted with the mixture of DCM and MeOH (10/1 (v/v), 30 mLx3).
92
SUBSTITUTE SHEET RULE 26 The combined organic layers were washed with brine (50 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) = 40/1 to 20/1) to afford light yellow liquid (0.26 g, 56%).
MS (ESI, pos. ion) m/z: 175.2 [M+H]+;
¾ NMR (400 MHz, CDCls): δ (ppm) 8.39 (d, J= 1.7 Hz, 1H), 7.58 (dd, J= 8.8, 2.2 Hz, 1H), 6.24 (d, J= 8.8 Hz, 1H), 4.42-4.34 (m, 2H), 4.10-4.01 (m, 1H), 3.77 (m, 2H).
Step 3) tert-butyl (3-((l-(5-cyanopyridin-2-yl)azetidin-3-yl)carbamoyl)pyrazolo[l,5-al pyrimidin- 2-yl)carbamate
[356] To a suspension of 2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3- carboxylic acid (0.20 g, 0.72 mmol) in DCM (15 mL) was added 6-(3-aminoazetidin-l- yl)nicotinonitrile (0.14 g, 0.79 mmol), HATU (0.33 g, 0.86 mmol) and Et3N (0.20 mL, 1.44 mmol). The reaction mixture was stirred at room temperature overnight, and then diluted with DCM (100 mL), washed with brine (50 mLx2), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by silica gel column chromatography (DCM/MeOH (v/v) = 100/1 to 80/1) to afford a light yellow solid (0.25 g, 80%).
MS (ESI, pos. ion) m/z: 379.1 [(M-56)+H]+;
!H NMR (400 MHz, CDC13): δ (ppm) 9.66 (s, 1H), 8.80 (dd, J= 6.9, 1.7 Hz, 1H), 8.57 (dd, J= 4.4, 1.7 Hz, 1H), 8.44 (d, J= 1.7 Hz, 1H), 8.33 (d, J= 7.2 Hz, 1H), 7.64 (dd, J= 8.7, 2.2 Hz, 1H), 6.98 (dd, J= 6.9, 4.4 Hz, 1H), 6.31 (d, J= 8.8 Hz, 1H), 5.10 (m, 1H), 4.58 (m, 2H), 4.16 (m, 2H), 1.57 (s, 9H).
Step 4) 2-amino-N-(l-(5-cvanopyridin-2-yl)azetidin-3-yl)pyrazolori,5-a1pyrimidine- 3- carboxamide
[357] To a suspension of tert- vXy\ (3-((l-(5-cyanopyridin-2-yl)azetidin-3-yl)carbamoyl) pyrazolo[l,5-a]pyrimidin-2-yl)carbamate (0.20 g, 0.46 mmol) in DCM (10 mL) was added a solution of HC1 in EA (10 mL, 3.5 M). The reaction mixture was stirred at room temperature overnight, then concentrated in vacuo. The reidue was diluted with DCM (20 mL) and saturated Na2C03 aqueous solution (10 mL). The organic layer was separated and the aqueous layer was extracted with the mixture of DCM and MeOH (10/1 (v/v), 30 mLx3). The combined organic layers was washed with brine (50 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM to DCM/MeOH (v/v) = 50/1) to afford a light yellow solid for (55 mg, 36%>).
MS (ESI, pos. ion) m/z: 335.2 [M+H]+;
93
SUBSTITUTE SHEET RULE 26 Purity:96%;
!H NMR (400 MHz, OMSO-de): δ (ppm) 8.89 (d, J = 6.6 Hz, 1H), 8.51 (d, J = 3.7 Hz, 1H), 8.46 (m, 1H), 8.22 (d, J = 7.3 Hz, 1H), 7.86-7.80 (m, 1H), 7.00 (dd, J = 6.5, 4.6 Hz, 1H), 6.49 (m, 3H), 4.98-4.85 (m, 1H), 4.42 (m, 2H), 4.06 (m, 2H).
Example 13 (6 -2-amino-N-(l-(5-cvanopyridin-2-yl)pyrrolidin-3-yl)pyrazolori,5-a1 pyrimidine-3- carboxamide
Figure imgf000095_0001
[358] To a suspension of 6-chloronicotinonitrile (1.08 g, 7.78 mmol) in DMF (70 mL) were added {S)-tert- vXy\ pyrrolidin-3-ylcarbamate (1.51 g, 8.12 mmol) and Na2C03 (1.72 g, 16.24 mmol). The reaction mixture was stirred at room temperature overnight and concentrated in vacuo. The residue was dissolved in a mixture of EtOAc and water (1/1 (v/v), 100 mL), and extracted with EtOAc (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, concentrated in vacuo to give the title compound as a beige solid (2.24 g, 100%).
MS (ESI, pos. ion) m/z: 289.2 [M+H]+;
!H NMR (400 MHz, CDCls): δ (ppm) 8.42 (d, J= 1.8 Hz, 1H), 8.03 (s, 1H), 7.61 (dd, J = 8.9, 2.2 Hz, 1H), 6.36 (d, J = 8.9 Hz, 1H), 4.74 (br. s, 1H), 3.79 (br. s, 1H), 3.62 (br. s, 2H), 3.41 (br. s, 1H), 2.32 (td, J= 13.5, 7.1 Hz, 1H), 2.02 (dt, J= 17.9, 6.0 Hz, 1H), 1.47 (s, 9H).
Step 2) (6 -6-(3-aminopyrrolidin-l-yl)nicotinonitrile
[359] To a solution of {S)-tert- vXy\ (l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamate (2.24 g, 7.78 mmol) in DCM (20 mL) was added a solution of HC1 in EtOAc (15 mL, 60 mmol). The reaction mixture was stirred at room temperature for 1 h and concentrated in vacuo. The residue was dissolved in water (30 mL) and adjusted to pH = 10 with saturated Na2C03 aqueous solution, then extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, concentrated in vacuo. The residue was purified by a
94
SUBSTITUTE SHEET RULE 26 silica gel column chromatography (MeOH/DCM (v/v) = 1/20) to give the title compound as a beige solid (0.52 g, 35.5%).
MS (ESI, pos. ion) m/z: 188.0 [M+H]+;
¾ NMR (400 MHz, CDCls): δ (ppm) 8.43 (d, J= 1.9 Hz, 1H), 7.60 (dd, J= 8.9, 2.3 Hz, 1H), 6.36 (d, J = 8.9 Hz, 1H), 3.81 (m, 1H), 3.71 (br. s, 2H), 3.56 (br. s, 1H), 3.28 (br. s, 1H), 2.25 (dt, J = 12.7, 7.1 Hz, 1H), 1.88 (m, 1H).
Step 3) (S)-tert- vXy\ (3-((l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamoyl)pyrazolo [1,5- a1pyrimidin-2-yl)carbamate
[360] To a solution of 2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3- carboxylic acid (263.3 mg, 0.946 mmol) and (5)-6-(3-aminopyrrolidin-l-yl) nicotinonitrile (178.1 mg, 0.946 mmol) in DCM (20 mL) were added HATU (428.2 mg, 1.126 mmol) and Et3N (0.19 mg, 1.877 mmol). The reaction mixture was then stirred at room temperature for 2 h, then quenched with water (30 mL), and extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, concentrated in vacuo. The residue was purified by a silica gel column chromatography (MeOH/DCM (v/v) = 1/80) to give the title compound as a beige solid (424.3 mg, 100%).
MS (ESI, pos. ion) m/z: 393.2 [(M-C4H8)+H]+;
!Η ΝΜΚ (400 MHz, CDC13): δ (ppm) 9.74 (s, 1H), 8.78 (dd, J= 6.9, 1.7 Hz, 1H), 8.51 (dd, J= 4.3, 1.6 Hz, 1H), 8.45 (d, J= 2.2 Hz, 1H), 8.03 (d, J= 6.6 Hz, 1H), 7.63 (dd, J= 8.9, 2.3 Hz, 1H), 6.94 (dd, J= 6.9, 4.4 Hz, 1H), 6.41 (d, J = 8.9 Hz, 1H), 4.84 (m, 1H), 3.95 (br. s, 1H), 3.76 (br. s, 2H), 3.59 (br. s, 1H), 2.49 (td, J= 13.4, 6.9 Hz, 1H), 2.22 (td, J= 13.2, 6.3 Hz, 1H), 1.57 (s, 9H).
Step 4) (6 -2-amino-N-(l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)pyrazolo[l ,^ pyrimidine-3- carboxamide
[361] To a solution of {S)-tert- vXy\ (3-((l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamoyl) pyrazolo[l,5-a]pyrimidin-2-yl)carbamate (424.3 mg, 0.946 mmol) in DCM (20 mL) was added a solution of HC1 in EtOAc (10 mL, 40 mmol). The reaction mixture was stirred at room temperature for 1.5 h and concentrated in vacuo. The residue was dissolved in water (30 mL) and adjusted to pH = 10 with a saturated Na2C03 aqueous solution, then extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, concentrated in vacuo. The residue was purified by a silica gel column chromatography (MeOH/DCM (v/v) =1/80) to give the title compound as a beige solid (0.23 g, 69.8%).
MS (ESI, pos. ion) m/z: 349.1 [M+H]+;
95
SUBSTITUTE SHEET RULE 26 ¾ NMR (400 MHz, CDCI3): δ (ppm) 8.46 (dd, J= 5.2, 1.7 Hz, 2H), 8.37 (dd, J= 4.5, 1.7 Hz, 1H), 7.95 (d, J= 7.2 Hz, 1H), 7.63 (dd, J= 8.9, 2.3 Hz, 1H), 6.80 (dd, J = 6.7, 4.4 Hz, 1H), 6.41 (d, J = 9.0 Hz, 1H), 5.68 (s, 2H), 4.87 (m, 1H), 3.95 (br. s, 1H), 3.75 (br. s, 2H), 3.56 (br. s, 1H), 2.49 (td, J= 13.5, 7.1 Hz, 1H), 2.21 (m, 1H).
Example 14 (i?)-2-amino-N-(l-(5-cvanopyridin-2-yl)pwolidin-3-yl)pyrazolori ,5-a1 pyrimidine-3- carboxamide
Figure imgf000097_0001
Step 1) (i?)-fert-butyl (l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamate
[362] To a suspension of 6-chloronicotinonitrile (1.00 g, 7.22 mmol) in DMF (25 mL) were added {R)-tert- vXy\ pyrrolidin-3-ylcarbamate (1.34 g, 7.22 mmol) and Na2C03 (1.53 g, 14.43 mmol). The reaction mixture was stirred at room temperature overnight, quenched with water (50 mL), and extracted with EtOAc (50 mL x 4). The combined organic phases were washed with brine (80 mL x 2), dried over anhydrous Na2S04, concentrated in vacuo. The residue was purified by a silica gel column chromatography (EtOAC/PE (v/v) =1/4) to give the title compound as a white solid (1.39 g, 66.8%).
MS (ESI, pos. ion) m/z: 289.2 [M+H]+;
'H NMR (400 MHz, CDCI3): δ (ppm) 8.43 (d, J= 1.8 Hz, 1H), 7.62 (dd, J= 8.9, 2.3 Hz, 1H), 6.37 (d, J= 8.9 Hz, 1H), 4.69 (m, 1H), 4.36 (m, 1H), 3.80 (m, 1H), 3.63 (m, 2H), 3.41 (m, 1H), 2.33 (m, lH), 2.02 (m, 1H), 1.47 (s, 9H).
Step 2) (i?)-6-(3-aminopyrrolidin-l-yl)nicotinonitrile
[363] To a solution of {R)-tert- vXy\ (l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamate (0.80 g, 2.77 mmol) in DCM (20 mL) was added a solution of HC1 in EtOAc (10 mL, 40 mmol). The reaction mixture was stirred at room temperature for 2 h and concentrated in vacuo. The residue was dissolved in water (30 mL) and adjusted to pH = 10 with a saturated Na2C03 aqueous solution, then extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, concentrated in vacuo. The residue was purified by a
96
SUBSTITUTE SHEET RULE 26 silica gel column chromatography (MeOH/DCM (v/v) = 1/20) to give the title compound as a beige solid (0.48 g, 92.1%).
MS (ESI, pos. ion) m/z: 188.9 [M+H]+;
!H NMR (400 MHz, CDCls): δ (ppm) 8.43 (d, J= 2.0 Hz, 1H), 7.60 (dd, J= 8.9, 2.3 Hz, 1H), 6.36 (d, J = 8.9 Hz, 1H), 3.81 (dt, J = 10.4, 5.3 Hz, 1H), 3.71 (br. s, 2H), 3.57 (br. s, 1H), 3.29 (br. s, 1H), 2.25 (dt, J= 12.7, 6.8 Hz, 1H), 1.88 (dt, J= 18.5, 6.2 Hz, 1H).
Step 3) (i?)-tert-butyl (3-((l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamoyl) pyrazolo[l,5- a1pyrimidin-2-yl)carbamate
[364] To a solution of 2-((tert-butoxycarbonyl)amino)pyrazolo[l,5-a]pyrimidine-3- carboxylic acid (289.4 mg, 1.04 mmol) and (i?)-6-(3-aminopyrrolidin-l-yl) nicotinonitrile (194.7 mg, 1.03 mmol) in DCM (20 mL) were added HATU (471.5 mg, 1.24 mmol) and Et3N (215.4 mg, 2.13 mmol). The reaction mixture was then stirred at room temperature for 2 h, quenched with water (30 mL), and extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, concentrated in vacuo. The residue was purified by a silica gel column chromatography (MeOH/DCM (v/v) = 1/80) to give the title compound as a beige solid (461.9 mg, 100%).
MS (ESI, pos. ion) m/z: 393.2 [(M-C4H8)+H]+;
!H NMR (400 MHz, CDC13): δ (ppm) 9.75 (s, 1H), 8.78 (dd, J= 6.9, 1.6 Hz, 1H), 8.52 (dd, J= 4.3, 1.5 Hz, 1H), 8.45 (d, J= 2.1 Hz, 1H), 8.04 (d, J= 6.9 Hz, 1H), 7.63 (dd, J= 8.9, 2.2 Hz, 1H), 6.94 (dd, J= 6.9, 4.4 Hz, 1H), 6.41 (d, J= 8.9 Hz, 1H), 4.84 (dq, J= 12.1, 6.0 Hz, 1H), 3.96 (br. s, 1H), 3.76 (br. s, 3H), 3.59 (br. s, 1H), 2.50 (dt, J = 20.3, 6.9 Hz, 1H), 2.23 (td, J = 13.1, 6.6 Hz, 1H), 1.57 (s, 9H).
Step 4) (i?)-2-amino-N-(l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)pyrazolo[l ,5-al pyrimidine-3- carboxamide
[365] To a solution of {R)-tert- vXy\ (3-((l-(5-cyanopyridin-2-yl)pyrrolidin-3-yl)carbamoyl) pyrazolo[l,5-a]pyrimidin-2-yl)carbamate (312.2 mg, 0.696 mmol) in DCM (20 mL) was added a solution of HC1 in EtOAc (10 mL, 40 mmol). The reaction mixture was stirred at room temperature for 1.5 h and concentrated in vacuo. The residue was dissolved in water (30 mL) and adjusted to pH = 10 with a saturated Na2C03 aqueous solution, then extracted with DCM (100 mL x 3). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, concentrated in vacuo. The residue was purified by a silica gel column chromatography (MeOH/DCM (v/v) = 1/80) to give the title compound as a beige solid (174.3 mg, 71.9%).
97
SUBSTITUTE SHEET RULE 26 MS (ESI, pos. ion) m/z: 349.1 [M+H]+;
¾ NMR (400 MHz, CDCls): δ (ppm) 8.46 (dd, J= 5.3, 1.6 Hz, 2H), 8.37 (dd, J= 4.5, 1.7 Hz, 1H), 7.95 (d, J= 6.5 Hz, 1H), 7.63 (dd, J= 8.9, 2.3 Hz, 1H), 6.80 (dd, J = 6.8, 4.5 Hz, 1H), 6.41 (d, J = 8.9 Hz, 1H), 5.68 (s, 2H), 4.88 (m, 1H), 3.95 (br. s, 1H), 3.76 (br. s, 2H), 3.58 (br. s, 1H), 2.49 (dt, J= 13.7, 6.3 Hz, 1H), 2.21 (dt, J= 11.7, 5.2 Hz, 1H).
Example 15 2-amino-N-(l -(5-cyanopyridin-2-yl)piperidin-4-yl)-5-cyclopropylpyrazolo[ 1 ,5- g]pyrimidine-3-carboxamide
Figure imgf000099_0001
Step 1) ( )-ethyl 3-amino-4A4-trichloro-2-cvanobut-2-enoate
[366] To a solution of ethyl 2-cyanoacetate (41.22 g, 364.41 mmol) and 2,2,2- trichloroacetonitrile (100.00 g, 692.58 mmol) in EtOH (120 mL) was added Et3N (2.00 g, 19.76 mmol). The reaction mixture was cooled to 0 °C and stirred for 2 h, then allowed to warm to room temperature and concentrated in vacuo. The residue was diluted with DCM (100 mL), and the resulting mixture was filtered through a pad of silica gel, eluting with DCM (1 L). The combined filtrates were concentrated in vacuo to give the title compound as a pale yellow solid (82.34 g, 87.7%).
MS (ESI, pos. ion) m/z: 257.0 [M+H]+;
¾ NMR (400 MHz, CDC13): δ (ppm) 10.22 (s, 1H), 6.86 (s, 1H), 4.33 (q, J= 7.12 Hz, 2H), 1.38 (t, J= 7.12 Hz, 3H).
Step 2) ethyl 3,5-diamino-lH-pyrazole-4-carboxylate
[367] To a solution of (Z)-ethyl 3-amino-4,4,4-trichloro-2-cyanobut-2-enoate (82.34 g, 319.77 mmol) in DMF (250 mL) was added a solution of hydrazine in water (50% [w/w], 46.08 g, 718.88 mmol). The reaction mixture was stirred at 100 °C for 2 h and then concentrated in vacuo. The residue was diluted with DCM (100 mL), and the resulting mixture was stood overnight. The solid
98
SUBSTITUTE SHEET RULE 26 was collected by Alteration, and then washed with DCM (50 mL x 3) to give the title compound as a pale yellow solid (40.40 g, 74.2%).
MS (ESI, pos. ion) m/z: 171.0 [M+H]+;
!H NMR (400 MHz, OMSO-de): δ (ppm) 5.35 (s, 4H), 4.14 (q, J = 7.12 Hz, 2H), 1.24 (t, J =7.12 Hz, 3H).
Step 3) ethyl 2-amino-5-oxo-4,5-dihydropyrazolo[l,5-alpyrimidine-3-carboxylate
[368] To a solution of sodium ethoxide in EtOH (0.49 N, 100 mL) were added ethyl 3,5- diamino-lH-pyrazole-4-carboxylate (2.08g, 0.012 mol), followed by solid 1,3-dimethyluracil (1.7 g, 0.012mol) and then the reaction mixture was stirred overnight at 90 °C. The reaction mixture was cooled to r.t, filtered to give the title compound as a pink solid (2.1 g, 79%).
¾ NMR (300MHz, OMSO-de): δ (ppm) 11.30 (brs, 1H), 8.23 (d, J= 7.86 Hz, 1H), 5.93 (brs, 2H), 5.89 (d, J= 7.83 Hz, 1H), 4.26 (q, J= 7.08 Hz, 2H), 1.28 (t, J= 7.08 Hz, 3H).
Step 4) ethyl 2-amino-5-bromopyrazolori,5-a1pyrimidine-3-carboxylate
[369] To a solution of ethyl 2-amino-5-oxo-4,5-dihydropyrazolo[l,5-a]pyrimidine-3- carboxylate (1.0 g, 4.50 mmol) in ACN (20 mL) was added POBr3 (6.45 g, 22.5 mmol). The reaction mixture was stirred at 40 °C for 18 h and concentrated to 5 mL in vacuo, then added to a mixture of water and ice (100 mL) with vigorous stirring. The resulting mixture was adjusted to PH = 7-8 with saturated NaHC03 aqueous solution at 0 °C, and extracted with EA (50 mL x 6). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 10/1) to give the title compound as a yellowish solid (550 mg, 43%)
MS (ESI, pos. ion) m/z: 282.7 [M+ H] +, 284.7 [M+ H] +.
Step 5) ethyl 2-(bis(tert-butoxycarbonyl)amino)-5-bromopyrazolo[l,5-a]pyrimidine-3-carboxylate
[370] To a solution of ethyl 2-amino-5-bromopyrazolo[l,5-a]pyrimidine-3-carboxylate (210 mg, 0.737 mmol) in DCM (10 mL), were added (Boc)20 (354 mg, 1.62 mmol), TEA (0.28 mL, 2.21 mmol) and DMAP (9 mg, 0.0737 mmol). The reaction mixture was stirred for 24 h at room temperature then diluted with DCM (20 mL), washed with saturated NH4C1 aqueous solution (10 mL), followed by water (10 mL) and brine (10 mL). The organic phase was dried over anhydrous Na2S04, concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 40/1) to afford the title compound as a yellow solid (264 mg, 74%).
MS (ESI, pos. ion) m/z: 506.7 [M+23]+, 508.6[M+23]+.
99
SUBSTITUTE SHEET RULE 26 Step 6) ethyl 2-(bis(tert-butoxycarbonyl)amino)-5-cvclopropylpyrazolori,5-a1pyrimidine-3- carboxylate and
[371] To a solution of ethyl 2-(bis(tert-butoxycarbonyl)amino)-5-bromopyrazolo[l,5-a] pyrimidine-3-carboxylate (200 mg, 0.412 mmol) and cyclopropyl boronic acid (106 mg, 1.24 mmol) in dioxane (2 mL) was added PdCl2(dppf).DCM (17 mg, 0.0206 mmol) under N2. The reaction mixture was stirred at 90 °C for 1.5 h, then K3PO4 (175 mg, 0.824 mmol) was added to the solution, stirred for another 4 h. After that, the resulting mixture was diluted with EA (20 mL), washed with water (20 mL) followed by brine (20 mL), dried over anhydrous Na2S04, filtered, concentrated in vacuo. The residue was purified by a silica gel column chromatography (PE/EA (v/v) = 4/1 to 5/2) to give the title compound as a yellowish solid (154 mg, 84%).
MS (ESI, pos. ion) m/z: 247.0 [M+ H]+;
!H NMR (300 MHz, CDCI3): δ (ppm) 9.24 (s, 1H), 8.50 (d, J = 7.1 Hz, 1H), 6.76 (d, J = 7.1 Hz, 1H), 4.40 (q, J = 7.1 Hz, 2H), 2.17-2.08 (m, 1H), 1.54 (s, 9H), 1.45 (t, J = 7.1 Hz, 3H), 1.29-1.23 (m, 2H), 1.20-1.13 (m, 2H).
Step 7) 2-(ter^butoxycarbonylamino)-5-cyclopropylpyrazolo[l,5-alpyrimidine-3-carboxylic acid
[372] To a solution of ethyl 2-(bis(tert-butoxycarbonyl)amino)-5-cyclopropylpyrazolo[l,5- a]pyrimidine-3-carboxylate (160 mg, 0.358 mmol) in a mixture of methanol and water (10 ml/2 ml), was added lithium hydroxide (86 mg, 3.58 mmol). The reaction mixture was stirred at reflux for 4 h and then concentrated to 2 mL in vacuo. The residue was diluted with water (15 mL), and the resulting mixture was adjusted to pH = 2 with 1M HCl aqueous solution, then white solid was appeared. The solid was collected by filtering to give the title compound as a white solid (64 mg, 57%).
MS (ESI, pos. ion) m/z: 340.8 [M+23]+;
!H NMR (300 MHz, DMSO-t ί): δ (ppm) 12.73 (brs, 1H), 9.46 (s, 1H), 9.06 (d, J = 7.1 Hz, 1H), 7.26 (d, J= 7.1 Hz, 1H), 2.45-2.37 (m, 1H), 1.67 (s, 9H), 1.30-1.25 (m, 4H).
Step 8) 6-(4-aminopiperidin-l-yl)nicotinonitrile
[373] To a solution of tert-butyl (l-(5-cyanopyridin-2-yl)piperidin-4-yl)carbamate (1.79 g, 5.92 mmol) in DCM (20 mL) was added HCl (3.0 M, 6.0 mL, 18 mmol) at 0 °C dropwise. The reaction was allowed warmed to room temperature and stirred overnight, then the mixture was concentrated in vacuo. The residue was diluted with water (20 mL), and the resulting mixture was washed with DCM (30 mL x 2). The aqueous layer was treated with saturated Na2C03 until pH = 9, and then extracted with DCM/MeOH (10/l(v/v), 30 mL x 3). The combined organic phases were washed
100
SUBSTITUTE SHEET RULE 26 with brine (50 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo to give the title compound as as a white solid (1.08 g, 90%).
MS (ESI, pos. ion) m/z: 203.0 [M+ H]+.
Step 9) fert-butyl 3 -( 1 -(5 -cvanopyridin-2-yl)piperidin-4-yl carbamoyl)^ - cvclopropyrpyrazolo[ 1 ,5-a]pyrimidin-2-yl carbamate
[374] To a solution of 2-(tert-butoxycarbonylamino)-5-cyclopropylpyrazolo[l,5-a]pyrimidine-3- carboxylic acid (140 mg, 0.442 mmol) in acetonitrile (15 ml) were added HBTU (201 mg, 0.531 mmol), DIPEA (250 μί, 1.55 mmol), and 6-(4-aminopiperidin-l-yl)nicotinonitrile) (179 mg, 0.884 mmol). The reaction mixture was stirred at room temperature overnight and then concentrated in vacuo. The residue was diluted with EA (50 mL) and washed with water (20 mL x 2) followed by brine (30 mL). The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/ EA (v/v) = 10/1) to give the title compound as a white solid (214 mg, 96%).
MS (ESI, pos. ion) m/z: 502.8 [M+ H]+;
!H NMR (300 MHz, CDCls): δ (ppm) 9.71 (s, 1H), 8.53 (d, J = 7.1 Hz, 1H), 8.42 (d, J = 2.2 Hz, 1H), 7.84 (d, J= 7.5 Hz, 1H), 7.64 (dd, J = 9.0, 2.3 Hz, 1H), 6.77 (d, J= 7.1 Hz, 1H), 6.69 (d, J = 9.1 Hz, 1H), 4.36-4.31 (m, 2H), 4.32-4.21 (m, 1H), 3.36-3.27 (m, 2H), 2.22-2.16 (m, 2H), 2.13- 2.07 (m, 1H), 1.66-1.43 (m, 11H), 1.26-1.12 (m, 4H).
Step 10) 2-amino-N-(l-(5-cyanopyridin-2-yl)piperidin-4-yl)-5-cyclopropylpyrazolo[l,5- alpyrimidine-3-carboxamide
[375] To a solution of tert-butyl-3-(l-(5-cyanopyridin-2-yl)piperidin-4-ylcarbamoyl)-5- cyclopropylpyrazolo[l,5-a]pyrimidin-2-ylcarbamate (100 mg, 0.199 mmoL) in DCM (10 mL) was added a solution of HCl in EA (10 mL, 40 mmol). The reaction mixture was stirred at room temperature for 30 h and then concentrated in vacuo. The residue was dissolved in 1 M HCl aqueous solution (15 mL), and the resulting mixture was adjusted to PH = 8 with 2 M NaOH aqueous solution, then extracted with DCM (10 mL x 6). The combined organic phases were washed with brine (100 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA/MeOH (v/v/v) = 200/10/1) to afford the title compound as a white solid (50 mg, 63 %>).
MS (ESI, pos. ion) m/z:403.0 [M+ H]+;
!H NMR (300 MHz, DMSO-Λ): δ (ppm) 8.59 (d, J = 6.9 Hz, 1H), 8.42 (d, J = 2.1 Hz, 1H), 7.75 (dd, J = 9.1, 2.3 Hz, 1H), 7.71 (d, J = 7.5 Hz, 1H), 6.92 (d, J = 9.1 Hz, 1H), 6.85 (d, J = 7.0 Hz,
101
SUBSTITUTE SHEET RULE 26 1H), 6.28 (brs, 2H), 4.28-4.21 (m, 2H), 4.17-4.04 (m, 1H), 3.48-3.08 (m, 2H), 2.20-2.09 (m, 1H), 2.09-1.95 (m, 2H), 1.52-1.39 (m, 2H), 1.12-1.06 (m, 2H), 1.04-1.00 (m, 2H).
Example 16 2-amino-N-(l-(5-cyanopyridin-2-yO^
alpyrimidine-3-carboxamide
Figure imgf000103_0001
Step 1) ethyl 2-amino-5-chloropyrazolo[l,5-alpyrimidine-3-carboxylate
[376] To a solution of ethyl 2-amino-5-oxo-4,5-dihydropyrazolo[l,5-a]pyrimidine-3-carboxylate (500 mg, 2.25 mmol) in ACN (20 mL) was added POCb (2.0 mL, 22.5 mmol). The reaction mixture was stirred at 40 °C for 20 h, then quenched with a mixture of water and ice (50 mL). The resulting mixture was adjusted to PH = 7~8 with saturated NaHC03 aqueous solution at 0 °C and then extracted with EA (50 mL x 6). The combined organic layers were washed with brine (150 mL), dried over anhydrous Na2S04 and filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 10/1) to afford the title compound as a white solid (210 mg, 39%).
¾ NMR (300 MHz, CDC13): δ (ppm) 8.31 (d, J= 7.0 Hz, 1H), 6.81 (d, J= 6.9 Hz, 1H), 5.53 (brs, 2H), 4.44 (q, J= 7.1 Hz, 2H), 1.44 (t, J= 7.1 Hz, 3H).
Step 2) ethyl 2-(bis(tert-butoxycarbonyl)amino)-5-chloropyrazolori,5-a1pyrimidine-3-carboxylat
[377] To a solution of ethyl 2-amino-5-chloropyrazolo[l,5-a]pyrimidine-3-carboxylate (86 mg, 0.357 mmol) in DCM (4 mL) were added (Boc)20 (93 mg, 0.428 mmol), TEA (0.09 mL, 0.715 mmol) and DMAP (4 mg, 0.0357 mmol). The reaction mixture was stirred at room temperature overnight then diluted with DCM (20 mL) and washed with NH4CI aqueous solution (10 mL), followed by water (10 mL) and brine (10 mL). The organic phase was dried over anhydrous Na2SC"4, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 10/1) to afford the title compound as a white solid (76 mg, 62%).
102
SUBSTITUTE SHEET RULE 26 !H NMR (300 MHz, CDCI3): δ (ppm) 8.56 (d, J= 7.2 Hz, 1H), 7.03 (d, J= 7.2 Hz, 1H), 4.38 (q, J = 7.1 Hz, 2H), 1.42 (s, 18H), 1.38 (t, J = 7.1 Hz, 3H).
Step 3 ) methyl 2-(tert-butoxycarbonylamino)-5-methoxypyrazolori,5-a1pyrimidine-3-carboxylate
[378] Na (30 mg, 1.30 mmol) was dissolved in MeOH (10 mL), and the resulting solution was added to ethyl 2-(bis(tert-butoxycarbonyl)amino)-5-chloropyrazolo[l,5-a]pyrimidine-3- carboxylate (140 mg, 0.318 mmol) dropwise. The reaction mixture was stirred at room temperature overnight. The resulting mixture was concentrated in vacuo, the residue was dissolved in EA (50 mL), washed with water (40 mL) followed by brine (40 mL). The organic phase was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 10/1) to afford the title compound as a white solid (100 mg, 75 %).
!H NMR (300 MHz, CDCI3): δ (ppm) 9.13 (s, 1H), 8.45 (d, J = 7.4 Hz, 1H), 6.41 (d, J = 7.4 Hz, 1H), 4.10 (s, 3H), 3.96 (s, 3H), 1.55 (s, 9H).
Step 4) 2-(tert-butoxycarbonylamino)-5-methoxypyrazolo[l,5-alpyrimidine-3- carboxylic acid
[379] To a solution of methyl-2-(tert-butoxycarbonylamino)-5-methoxypyrazolo[l,5-a]- pyrimidine-3-carboxylate (100 mg, 0.308 mmol) in methanol/water (10 ml/2 ml) was added lithium hydroxide (74 mg, 3.08 mmol). The reaction mixture was stirred at reflux for 5 h and then concentrated to 2 mL in vacuo. The residue was diluted with water (15 mL) and adjusted to pH = 2 with 1M HC1 aqueous solution, then extracted with DCM (10 mL x 5). The combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 10/1 to DCM/EA/MeOH (v/v/v) = 20/2/1) to afford the title compound as a white solid (62 mg, 65 %).
MS (ESI, pos. ion) m/z:330.9 [M+23]+;
!H NMR (300 MHz, DMSO-t ί): δ (ppm) 8.85 (d, J = 7.4 Hz, 1H), 6.64 (d, J = 7.4 Hz, 1H), 4.02 (brs, 1H), 3.98 (s, 3H), 1.48 (s, 9H).
Step 5) tert-butyl 3-(l-(5-cyanopyridin-2-yl)piperidin-4-ylcarbamoyl)-5-methoxypyrazolo[l,5- q]pyrimidin-2-yl carbamate
[380] To a solution of 2-(tert-butoxycarbonylamino)-5-methoxypyrazolo[l,5-a]pyrimidine-3- carboxylic acid (34 mg, 0.110 mmol) in acetonitrile (5 ml) were added HBTU (50 mg, 0.132 mmol), DIPEA (62
Figure imgf000104_0001
0.385 mmol) and 6-(4-aminopiperidin-l-yl)nicotinonitrile (44 mg, 0.219 mmol). The reaction mixture was stirred at room temperature for 2.5 h and then concentrated in vacuo. The residue was diluted with DCM (15 mL) and washed with water (8 mL x 2) followed by
103
SUBSTITUTE SHEET RULE 26 brine (10 mL). The organic layers dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 25/1 to 10/1) to give the title compound as a white solid (51 mg, 94%).
MS (ESI, pos. ion) m/z: 492.7 [M+ H]+;
!H NMR (300 MHz, CDCb): δ (ppm) 9.66 (s, 1H), 8.47 (d, J = 7.4 Hz, 1H), 8.42 (d, J = 2.2 Hz, 1H), 7.63 (dd, J= 9.0, 2.3 Hz, 1H), 7.54 (d, J= 7.6 Hz, 1H), 6.67 (d, J = 9.1 Hz, 1H), 6.37 (d, J = 7.4 Hz, 1H), 4.41-4.34 (m, 2H), 4.32-4.19 (m, 1H), 4.01 (s, 3H), 3.31-3.21 (m, 2H), 2.24-2.19 (m, 2H), 1.63-1.45 (m, 11H).
Step 6) 2-amino-N-(l-(5-cvanopyridin-2-yl)piperidin-4-yl)-5-methoxypyrazolori,5-a1 pyrimidine- 3-carboxamide
[381] To a suspension of tert-butyl-3-(l-(5-cyanopyridin-2-yl)piperidin-4-ylcarbamoyl)-5- methoxypyrazolo[l,5-a]pyrimidin-2-ylcarbamate (40 mg, 0.081 mmoL) in DCM (5 mL) was added a solution of HCl in EA (10 mL, 40mmol). The reaction mixture was stirred at room temperature for 24 h and then concentrated in vacuo. The residue was dissolved in HCl aqueous solution (1 M, 5 mL), and the resulting mixture was adjusted to PH = 8 with 2 M NaOH aqueous solution, and then extracted with DCM (5 mL x 5). The combined DCM layers were washed with brine (10 mL), dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA/MeOH (v/v/v) = 20/2/1) to afford the title compound as a white solid (25 mg, 81 %).
MS (ESI, pos. ion) m/z:393.2 [M+ H]+;
1H NMR (300 MHz, D2O): 5 (ppm) 8.22 (d, J= 7.6 Hz, 1H), 8.14 (d, J= 1.8 Hz, 1H), 7.78 (d, J = 9.6 Hz, 1H), 7.07 (d, J = 9.9 Hz, 1H), 6.35 (d, J = 7.6 Hz, 1H), 4.05-3.95 (m, 1H), 3.96-3.86 (m, 2H), 3.73 (brs, 5H), 3.37-3.27 (m, 2H), 2.09-2.02 (m, 2H), 1.50-1.35 (m, 2H).
Example 17 N-(l-(5-cyanopyridin-2-yl)piperidin-4-yl)-5-cyclopropylpyrazolo[l,5-al pyrimidine- 3-carboxamide
Figure imgf000105_0001
104
SUBSTITUTE SHEET RULE 26 Step 1) (Z ethyl 2-cyano-3-ethoxyacrylate
[382] A solution of ethyl 2-cyanoacetate (11.3 g, 100 mmol), tri ethyl orthoformate (15 g, 101 mmol) and acetic anhydride (50 mL) was stirred at 140 °C overnight. The resulting mixture was concentrated in vacuo and the residue was diluted with a mixture of ether and n-hexane (1/10 (v/v), 60 mL), then white precipitate was appeared. Filtrated and collected the filter cake to give the title compound as a white solid (10.5 g (47%).
LC-MS (ESI, pos. ion) m/z: 170.2 [M+H]+.
Step 2) ethyl 5-amino-lH-pyrazole-4-carboxylate
[383] A solution of (Z)-ethyl 2-cyano-3-ethoxyacrylate (10.5 g, 48.5 mmol) and hydrazine hydrate (5.5 g, 73 mmol) in ethanol (100 mL) was heated to reflux and stirred for 3 h. The resulting mixture was concentrated in vacuo, the residue was diluted with ethyl acetate (100 mL) and washed with water (3 x 20 mL) followed by brine (50 mL). The organic layer was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (EA/PE (v/v) = 1/2) to afford the title compound as a yellow solid (7.2 g, 52%).
LC-MS (ESI, pos. ion) m/z: 156.2 [M+H]+;
!H NMR (400 MHz, CDCls): δ (ppm) 7.73 (s, 1H), 6.58 (br, 3H), 4.28 (q, J= 7.1 Hz, 2H), 1.33 (t, J= 7.1 Hz, 3H).
Step 3) ethyl 5-oxo-4,5-dihydropyrazolo[l,5-a]pyrimidine-3-carboxylate
[384] To a solution of sodium ethoxide (520mg, 7.6mmol) in EtOH (20mL) were added ethyl 3- amino-lH-pyrazole-4-carboxylate (1 g, 6.45 mmol) and solid 1,3-dimethyluracil (1.35g, 9.68mmol), under a N2 atmosphere. The reaction mixture was stirred at 90 °C for 3 h then cooled to 0 °C in ice-water bath. Amber precipitate was appeared. The resulting amber precipitate was filtered off and dissolved in water (25 mL) and then the resulting solution was neutralized with acetic acid. White solid was appeared, filtered and collected the solid, and then azeotroped to dryness by refluxing in toluene (100 mL) to give the title compound as a white solid (900 mg, 67%).
!H NMR (300 MHz, DMSO-Λ): δ (ppm) 11.78 (brs, 1H), 8.58 (d, J = 8.0 Hz, 1H), 8.15 (s, 1H), 6.16 (d, J= 7.9 Hz, 1H), 4.28 (q, J= 7.0 Hz, 2H), 1.29 (t, J= 7.0 Hz, 3H).
Step 4) ethyl 5-bromopyrazolo[l,5-a]pyrimidine-3-carboxylate
[385] To a solution of ethyl 5-bromopyrazolo[l,5-a]pyrimidine-3-carboxylate (850 mg, 4.10 mmol) in CH3CN (30 mL) was added POBr3 (11.66 g, 40.67mmol).The reaction mixture was
105
SUBSTITUTE SHEET RULE 26 stirred at 60 °C for 4 h and then concentrated in vacuo. The residue was dissolved in water (10 mL) and the resulting solution was adjusted to pH = 7 with 2 M NaHC03 aqueous solution, then extracted with DCM (50 mL x 3). The combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/MeOH (v/v) = 260/1) to afford the title compound as a white solid (670mg, 61%).
Ή ΝΜΡ βΟΟ MHz, CDC1 ): δ (ppm) 8.55-8.51 (m, 2H), 7.13 (d, J= 7.2 Hz, 1H), 4.42 (q, J= 7.1 Hz, 2H), 1.42 (t, J= 7.1 Hz, 3H).
Step 5) ethyl 5-cvclopropylpyrazolori,5-a1pyrimidine-3-carboxylate
[386] To a solution of cyclopropylboronic acid (174mg, 2.1mmol), KOAc (330mg, 3.4mmol), ethyl 5-bromopyrazolo[l,5-a]pyrimidine-3-carboxylate (500mg, 1.7mmol) in dioxane (15ml) was added PdCl2(dppf).DCM (138mg, 0.17mmol). The reaction mixture was stirred at 90 °C for 2.5 h under a nitrogen atmosphere and then concentrated in vacuo. The residue was diluted with water and then extracted with DCM (20 x 3). The combined organic layers were washed with brine and dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 6/1) to give the title compound as a brown solid (220 mg, 57%).
!H NMR (300 MHz, CDC1 ): δ (ppm) 8.51 (d, J = 7.2 Hz, 1H), 8.47 (s, 1H), 6.78 (d, J = 7.2 Hz, 1H), 4.39 (q, J= 7.1 Hz, 2H), 2.25-2.16 (m, 1H), 1.41 (t, J= 7.1 Hz, 3H), 1.33-1.25 (m, 2H), 1.25- 1.14 (m, 2H).
Step 6) 5-cvclopropylpyrazolori,5-a1pyrimidine-3-carboxylic acid
[387] To a solution of ethyl 5-cyclopropylpyrazolo[l,5-a]pyrimidine-3-carboxylate (40 mg, 0.17 mmol) in methanol/water (5mL/lmL) was added lithium hydroxide (24 mg, 1.1 mmol). The reaction mixture was stirred at room temperature for 15 h and then concentrated in vacuo. The residue was partitioned between water and dichloromethane, the aqueous phase was separated and adjusted to pH = 5 with 1M acetic acid aqueous solution then extract with DCM (15 mL x 3). The combined DCM layers were washed with brine (20 mL) and concentrated in vacuo to give the crude product as a white solid (35mg, 100%). The crow product was used in the next step without purification .
¾ NMR (300MHz,DMSO-</i): δ (ppm) 12.16 (s, 1H), 9.04 (d, J= 6 Hz, 1H), 8.45 (s, 1H), 7.14 (d, J= 6 Hz, 1H), 2.30 (m, 1H), 1.15 (m, 4H).
106
SUBSTITUTE SHEET RULE 26 Step 7) N-(l-(5-cvanopyridin-2-yl)piperidin-4-yl)-5-cvclopropylpyrazolori,5-a1 pyrimidine-3- carboxamide
[388] To a solution of 5-cyclopropylpyrazolo[l,5-a]pyrimidine-3-carboxylic acid (25 mg, 0.12 mmol) in acetonitrile (5 ml) were added 6-(4-aminopiperidin-l-yl)nicotinonitrile (37 mg, 0.18 mmol), HBTU (56 mg, 0.144 mmol) and DIPEA (56 mg, 0.42 mmol). The reaction mixture was stirred at room temperature for 20 h and then concentrated in vacuo. The residue was partitioned with dichloromethane and water, the organic layer was separated and washed with brine, dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 4/1) to give the title compound as a white solid
(37mg, 77%).
MS (ESI, pos. ion) m/z:388.0 [M+ H]+;
!H NMR (300MHz,CDCl3): δ (ppm) 8.55 (d, J= 6 Hz, 2H), 8.43 (d, J= 3 Hz, 1H), 7.93 (d, J= 6 Hz, 1H), 7.62 (dd, J= 3 Hz, J= 3 Hz, 1H), 6.85 (d, J= 6 Hz, 1H), 6.67 (d, J= 9 Hz, 1H), 4.32 (m, 3H), 3.32 (m, 2H), 1.57 (m, 2H), 2.17 (m, 3H), 1.16 (m, 4H).
Example 18 N-(l-(5-cyanopyridin-2-yl)piperidin-3-yl)pyrazolo[l,5-alpyrimidine- carboxamide
N
Figure imgf000108_0001
Step 1) ethyl pyrazolo[l,5-alpyrimidine-3-carboxylate
[389] To a solution of ethyl 5-amino-lH-pyrazole-4-carboxylate (3.0 g, 19.3 mmol) in acetic acid (40 mL) and ethanol (10 mL) was added 1,1,3,3-tetramethoxypropane (3.48 g, 21.4 mmol). The reaction mixture was stirred at 90 °C overnight and cooled down to room temperature and then concentrated in vacuo. The residue was diluted with ethyl acetate (100 mL) and washed with saturated NaHC03 aqueous solution (20 mL) and then dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography (DCM/EA (v/v) = 6/1) to give the title compound as a light yellow solid (3.2 g, 85%).
LC-MS (ESI, pos. ion) m/z: 192.3 [M+H]+;
!H NMR (600 MHz, CDC1 ): δ (ppm) 8.81 (m, 1H), 8.78 (m, 1H), 8.61 (s, 1H), 7.04 (dd, J = 6.9, 4.1 Hz, 1H), 4.47 (q, J= 7.1 Hz, 2H), 1.44 (t, J = 7.1 Hz, 3H).
107
SUBSTITUTE SHEET RULE 26 Step 2) pyrazolo[l,5-a]pyrimidine-3-carboxyric acid
[390] To a solution of ethyl pyrazolo[l,5-a]pyrimidine-3-carboxylate (3.15 g, 16.4 mmol) in ethanol (50 mL) and water (40 mL) was added potassium hydroxide aqueous solution (2 M, 40 mL). The reaction mixture was stirred at 50 °C for 2 h and cooled down to room temperature and then concentrated in vacuo. The resulting mixture was adjusted to pH = 2 with 1M hydrochloric acid aqueous solution, and white solid was appeared, then filtered to afford the product as a white solid (2.6 g, 97%).
LC-MS (ESI, neg. ion) m/z: 162.1 [M-H] .
!H NMR (600 MHz, DMSO-Λ): δ (ppm) 9.26 (dd, J= 7.0, 1.7 Hz, 1H), 8.80 (dd, J = 4.1, 1.7 Hz, 1H), 8.58 (s, 1H), 7.26 (dd, J= 7.0, 4.1 Hz, 1H).
Step 3) N-(l-(5-cyanopyridin-2-yl)piperidin-3-yl)pyrazolo[l,5-alpyrimidine-3 carboxamide
[391] To a solution of pyrazolo[l,5-a]pyrimidine-3-carboxylic acid (654 mg, 4.0 mmol) in DMF (15 mL) were added 6-(3-aminopiperidin-l-yl) nicotinonitrile (623 mg, 3.08 mmol), HATU (1.52 g, 4.0 mmol) and triethylamine (622 mg, 6.16 mmol). The reaction mixture was stirred at room temperature overnight. The reaction mixture was concentrated in vacuo, and the residue was diluted with EA (50 mL), washed with water (3 x 15 mL), followed by brine (30 mL). The organic phase was dried over anhydrous Na2S04, filtered and concentrated in vacuo. The residue was purified by a silica gel column chromatography on silica gel (MeOH/CH2Cl2 (v/v) = 1/30) to give the title compound as a white solid (898 mg, 84%).
Purity: HPLC 99.6%;
LC-MS (ESI, pos. ion) m/z: 348.2 [M+H] +;
!H NMR (600 MHz, CDCls) : δ (ppm) 8.79 (dd, J = 7.0, 1.7 Hz, 1H), 8.68 (s, 1H), 8.48 (dd, J = 4.1, 1.7 Hz, 1H), 8.37 (d, J= 1.7 Hz, 1H), 8.02 (d, J = 7.1 Hz, 1H), 7.53 (dd, J = 9.1, 2.3 Hz, 1H), 7.00 (dd, J = 7.0, 4.1 Hz, 1H), 6.75 (d, J = 9.1 Hz, 1H), 4.25-4.31 (m, 1H), 4.07 (dd, J = 13.3, 3.1 Hz, 1H), 3.98 -3.92(m, 1H), 3.77-3.69 (m, 1H), 3.66 (dd, J= 13.3, 7.2 Hz, 1H), 2.17-2.11 (m, 1H), 1.96 -1.85 (m, 2H), 1.79-1.72 (m, 1H).
BIOLOGICAL TESTING
[392] The LC/MS/MS system used in the analysis consists of an Agilent 1200 Series vacuum degasser, binary pump, well-plate autosampler, thermostatted column compartment, the Agilent G6430 Triple Quadrupole Mass Spectrometer with an electrosprayionization (ESI) source. Quantitative analysis was carried out using MRM mode. The parameters for MRM transitions are
108
SUBSTITUTE SHEET RULE 26 in the Table A. Table A
Figure imgf000110_0001
[393] An Agilent XDB-C18, 2.1 x 30 mm, 3.5 μΜ column was used for the analysis. 5 of the samples were injected. Analysis condition: The mobile phase was 0.1% formic acid in water (A) and 0.1% formic acid in methanol (B). The flow rate was 0.4 mL/min. And the gradient of Mobile phase was in the Table B.
Table B
Figure imgf000110_0002
[394] Alternatively, an Agilent 6330 series LC/MS/MS spectrometer equipped with G1312A binary pumps, a G1367A autosampler and a G1314C UV detector were used in the analysis. An ESI source was used on the LC/MS/MS spectrometer. The analysis was done in positive ion mode as appropriate and the MRM transition for each analyte was optimized using standard solution. A Capcell MP-C18 100 x 4.6 mm I.D., 5 μΜ column (Phenomenex, Torrance, California, USA) was used during the analysis. The mobile phase was 5 mM ammonia acetate, 0.1% MeOH in water (A): 5 mM ammonia acetate, 0.1% MeOH in acetonitrile (B) (70:30, v/v). The flow rate was 0.6 mL/min. Column was maintained at ambient temperature. 20 μΐ^ of the samples were injected.
Example A: Compound Stability In Human and Rat Liver Microsomes
[395] Human or rat liver microsomes incubations were conducted in duplicate in polypropylene
109
SUBSTITUTE SHEET RULE 26 tubes. The typical incubation mixtures consisted of human or rat liver microsomes (0.5 mg protein/mL), compounds of interest (5 μΜ) and NADPH (1.0 mM) in a total volume of 200 potassium phosphate buffer (PBS, 100 mM, pH 7.4). Compounds were dissolved in DMSO and diluted with PBS such that the final concentration of DMSO was 0.05%. The enzymatic reactions were commenced with the addition of protein after a 3-min preincubation and incubated in a water bath open to the air at 37 °C. Reactions were terminated at various time points (0, 5, 10, 15, 30, 60 min) by adding equal volume of ice-cold acetonitrile. The samples were stored at -80 °C until LC/MS/MS assays.
[396] The concentrations of compounds in the incubation mixtures of human or rat liver microsomes were determined by a LC/MS/MS method. The ranges of the linearity in the concentration range were determined for each tested compounds.
[397] A parallel incubation was performed using denatured microsomes as the negative control, and reactions were terminated at various time points (0, 15, 60 min) after incubation at 37 °C.
[398] Dextromethorphan (70 μΜ) was selected as the positive control, and reactions were terminated at various time points (0, 5, 10, 15, 30, 60 min) after incubation at 37 °C. Both positive and negative control samples were included in each assay to ensure the integrity of the microsomal incubation system.
Data Analysis
[399] The concentrations of compounds in human or rat liver microsome incubations were plotted as a percentage of the relevant zero time point control for each reaction. The in vivo CLint were extrapolated (ref : Naritomi, Y.; Terashita, S.; Kimura, S.; Suzuki, A.; Kagayama, A.; and Sugiyama, Y.; Prediction of human hepatic clearance from in vivo animal experiments and in vitro metabolic studies with liver microsomes from animals and humans. Drug Metab. Dispos., 2001, 29: 1316-1324).
[400] Exemplary results from selected compounds of the invention are listed in Table 2. The compounds disclosed herein exhibited desirable stability when the compounds were incubated in human and rat liver microsomes.
Table 2 Stability of selected compounds of the invention in Human and rat liver microsomes
Figure imgf000111_0001
110
SUBSTITUTE SHEET RULE 26 Ex. 1 oo ND 344.8 7.20
Ex. 2 oo ND 124.0 20.03
Ex. 4 18.41 94.42 2.38 1044.89
Ex. 5 3250 0.53 1529 1.62
Ex. 6 oo ND 145.2 17.11
Ex. 7 oo ND 841.7 2.95
Ex. 8 727.0 2.39 498.6 4.98
Ex. 9 oo ND 155.9 15.93
Ex. 10 466.0 3.73 65.05 38.18
Ex. 11 12276 0.14 984.2 2.52
Ex. 12 oo ND 361.8 6.86
Ex. 13 232.7 7.47 67.13 37.00
Ex. 14 328.4 5.29 67.43 36.83
Ex. 15 15.97 108.85 5.06 490.85
Ex. 18 36.34 47.83 6.44 385.79
ND: not determined
Example B: Evaluation of Pharmacokinetics After Intravenous and Oral Administration of The Compounds Disclosed Herein In Mice, Rats, Dogs And Monkeys
[401] The compounds disclosed herein are assessed in pharmacokinetic studies in mice, rats, dogs or monkeys. The compounds are administered as a water solution, 2% HPMC + 1% TWEEN®80 in water solution, 5% DMSO + 5% solutol in saline, 4% MC suspension or capsule. For the intravenous administration, the animals are generally given at 1 or 2 mg/kg dose. For the oral (p.o.) dosing, mice and rats are generally given 5 or 10 mg/kg dose, and dogs and monkeys are generally given 10 mg/kg dose. The blood samples (0.3 mL) are drawn at 0.25, 0.5, 1.0, 2.0, 3.0, 4.0, 6.0, 8.0, 12 and 24 h time points or 0.083, 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0 and 24 h time points and centrifuged at 3,000 or 4000 rpm for 2 to 10 min. The plasma solutions are collected, and stored at -20 °C or -70 °C until analyzed by LC/MS/MS as described above.
[402] Exemplary study results from examples disclosed herein are listed in Table 3. The compounds disclosed herein exhibited optimized pharmacokinetic properties with desirable oral
111
SUBSTITUTE SHEET RULE 26 bioavailability (F) and half-life (T1/2) when the compounds were administered orally or intravenously.
Table 3 Pharmacokinetic profiles of selected compounds of the invention in rats
Figure imgf000113_0001
Example C: Kinase Activity Assay
[403] The efficacy of the compounds disclosed herein as inhibitors of JAK kinases can be evaluated as follows.
General Description for Kinase Assays
[404] Kinase assays can be performed by measurement of incorporation of γ-33Ρ ATP into immobilized myelin basic protein (MBP). High binding white 384 well plates (Greiner) are coated with MBP (Sigma #M-1891) by incubation of 60 μΕΛνεΙΙ of 20 μg/mL MBP in Tris-buffered saline (TBS; 50 mM Tris pH 8.0, 138 mM NaCl, 2.7 mM KC1) for 24 h at 4°C. Plates are washed 3 x with 100 TBS. Kinase reactions are carried out in a total volume of 34 μΕ in kinase buffer (5 mM Hepes pH 7.6, 15 mM NaCl, 0.01% bovine gamma globulin (Sigma #1-5506), 10 mM MgCb, 1 mM DTT, 0.02% TritonX-100). Compound dilutions are performed in DMSO and added to assay wells to a final DMSO concentration of 1%. Each data point is measured in duplicate, and at least two duplicate assays are performed for each individual compound determination. Enzyme
112
SUBSTITUTE SHEET RULE 26 is added to final concentrations of 10 nM or 20 nM, for example. A mixture of unlabeled ATP and γ-33Ρ ATP is added to start the reaction (2 x 106 cpm of γ-33Ρ ATP per well (3000 Ci/mmole) and 10 μΜ unlabeled ATP, typically. The reactions are carried out for 1 h at room temperature with shaking. Plates are washed 7x with TBS, followed by the addition of 50 μίΛνεΙΙ scintillation fluid (Wallac). Plates are read using a Wallac Trilux counter. This is only one format of such assays; various other formats are possible, as known to one skilled in the art.
[405] The above assay procedure can be used to determine the IC50 for inhibition and/or the inhibition constant, K . The IC50 is defined as the concentration of compound required to reduce the enzyme activity by 50% under the condition of the assay. The IC50 value is estimated by preparing a 10 point curve using a ½ log dilution series (for example, a typical curve may be prepared using the following compound concentrations: 10 μΜ, 3 μΜ, 1 μΜ, 0.3 μΜ, 0.1 μΜ, 0.03 μΜ, 0.01 μΜ, 0.003 μΜ, 0.001 μΜ and 0 μΜ).
JAK KIANSE GENERAL ASSAY PROTOCOL
JAK1 (h)
[406] JAK1 (h) is incubated with 20 mM Tris/HCl pH 7.5, 0.2 mM EDTA, 500 μΜ GEEPLYWSFPAKK , 10 mM MgAcetate and [γ-33Ρ-ΑΤΡ] (specific activity approx. 500 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 μΐ^ of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
JAK2 (h)
[407] JAK2 (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 100 μΜ KTFCGTPEYLAPEVRREPRILSEEEQEMFRDFDYIADWC, 10 mM MgAcetate and [γ-33Ρ-ΑΤΡ] (specific activity approx. 500 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 μΐ, οΐ the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
JAK3 (h)
[408] JAK3 (h) is incubated with 8 mM MOPS pH 7.0, 0.2 mM EDTA, 500 μΜ GGEEEEYFELVK K , 10 mM MgAcetate and [γ-33Ρ-ΑΤΡ] (specific activity approx. 500
113
SUBSTITUTE SHEET RULE 26 cpm/pmol, concentration as required). The reaction is initiated by the addition of the MgATP mix. After incubation for 40 minutes at room temperature, the reaction is stopped by the addition of 3% phosphoric acid solution. 10 μΐ^ of the reaction is then spotted onto a P30 filtermat and washed three times for 5 minutes in 75 mM phosphoric acid and once in methanol prior to drying and scintillation counting.
[409] The kinase assays described herein were performed at Millipore UK Ltd, Dundee Technology Park, Dundee DD2 1 SW, UK.
[410] Alternatively, the kinase activities of the compounds can be measured using KJNOMEscan™, which is based on a competition binding assay that quantitatively measures the ability of a compound to compete with an immobilized, active-site directed ligand. The assay was performed by combining three components: DNA-tagged kinase; immobilized ligand; and a test compound. The ability of the test compound to compete with the immobilized ligand was measured via quantitative PCR of the DNA tag.
[41 1] For most assays, kinase-tagged T7 phage strains were prepared in an E. coli host derived from the BL21 strain. E. coli were grown to log-phase and infected with T7 phage and incubated with shaking at 32 °C until lysis. The lysates were centrifuged and filtered to remove cell debris. The remaining kinases were produced in HEK-293 cells and subsequently tagged with DNA for qPCR detection. Streptavidin-coated magnetic beads were treated with biotinylated small molecule ligands for 30 minutes at room temperature to generate affinity resins for kinase assays. The liganded beads were blocked with excess biotin and washed with blocking buffer (SEABLOCK™ (Pierce), 1% BSA, 0.05% TWEEN®20, 1 mM DTT) to remove unbound ligand and to reduce nonspecific binding. Binding reactions were assembled by combining kinases, liganded affinity beads, and test compounds in lx binding buffer (20% SEABLOCK™, 0.17x PBS, 0.05% TWEEN®20, 6 mM DTT). All reactions were performed in polystyrene 96-well plates in a final volume of 0.135 mL. The assay plates were incubated at room temperature with shaking for 1 hour and the affinity beads were washed with wash buffer (lx PBS, 0.05% TWEEN®20). The beads were then re-suspended in elution buffer (lx PBS, 0.05% TWEEN®20, 0.5 μΜ non-biotinylated affinity ligand) and incubated at room temperature with shaking for 30 minutes. The kinase concentration in the eluates was measured by qPCR.
[412] The kinase activity assays described herein were performed using KINOMEscan™ Profiling Service at DiscoveRx Corporation, 42501 Albrae St. Fremont, CA 94538, USA.
[413] Exemplary study results from examples disclosed herein are listed in Table 4. The compounds disclosed herein exhibited potent activities in the JAK kinase assays. Table 4 lised the
114
SUBSTITUTE SHEET RULE 26 IC50S of some examples described herein in the JAK1 and JAK2 kinase assays. Table 4 JAK1 and JAK2 Kinase inhibition data
Figure imgf000116_0001
NT: not tested
[414] Finally, it should be noted that there are alternative ways of implementing the present invention. Accordingly, the present embodiments are to be considered as illustrative and not restrictive and the invention is not be limited to the details given herein, but may be modified within the scope and equivalents of the appended claims. All publications and patents cited herein are incorporated by reference.
115
SUBSTITUTE SHEET RULE 26

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A compound having Formula (I):
Figure imgf000117_0001
or a stereoisomer, a tautomer, an N-oxide, a solvate, a metabolite, a pharmaceutically acceptable salt or a prodrug thereof, wherein:
Z is C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z is optionally substituted with 1, 2, 3, 4 or 5 R1 groups;
Z1 is H, C1-C12 alkyl, C3-C12 cycloalkyl or 3-12 membered heterocyclyl, wherein Z1 is optionally substituted with 1 , 2, 3, 4 or 5 R2 groups;
X is H, -NRaRb, -N(Ra)C(=0)Rc, -N(Rc)C(=0)NRaRb, -N(Ra)C(=0)ORc or -N(Ra)S(=0)mRc;
R7 is H, NO2, N3, CN, C3-C12 cycloalkyl, -ORc, -C(=0)Rc, -OC(=0)Rc, -C(=0)ORc or - C(=0)NRaRb, with the proviso that when X is H, R7 is C3-C12 cycloalkyl, or when each of X and R7 is H, Z has formula (Z-34):
Figure imgf000117_0002
(Z-34) wherein R7 is optionally substituted with 1 , 2, 3, 4 or 5 R6 groups;
each R1 and R2 is independently H, F, N02, N3, CN, C1-C12 alkyl, C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C12 aryl, 5-12 membered heteroaryl, -(CR4R5)n- ORc, -(CR4R5)„-NRaRb, -S(=0)mRc, -S(=0)2NRaRb, -C(=0)Rc, -OC(=0)Rc, -N(Ra)C(=0)Rc, - (CR4R5)„C(=0)ORc, -(CR4R5)„C(=0)NRaRb, -C(=NRc)NRaRb, -N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, wherein each R1 and R2 is optionally independently substituted with 1, 2, 3, 4 or 5 R3 groups; each R3 is independently H, F, CI, Br, I, N02, N3, CN, C1-C12 alkyl, C2-C12 alkenyl, C2-C12 alkynyl, C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C12 aryl, 5-12 membered heteroaryl, - (CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)mRc, -S(=0)2NRaRb, -C(=0)Rc, -OC(=0)Rc, -N(Ra)C(=0)Rc, -(CR4R5)„C(=0)ORc, -(CR4R5)„C(=0)NRaRb, -C(=NRc)NRaRb, -N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, or two adjacent R3 taken together with the atoms to which they are attached to form a C3-C12 cycloalkyl, 3-12 membered heterocyclyl, C6-C12 aryl or 5-12 membered heteroaryl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3, 4 or 5 R6 groups;
each R4 and R5 is independently H, F, CI, Br, I, N3, CN, OH, NH2, C1-C12 alkyl, C1-C12 alkoxy, Ci-Ci2 alkylamino, C2-Ci2 alkenyl, C2-Ci2 alkynyl, C3-Ci2 cycloalkyl, 3-12 membered heterocyclyl, C6-Ci2 aryl or 5-12 membered heteroaryl, or R4 and R5 taken together with the carbon atom to which they are attached form a C3-Ci2 cycloalkyl or 3-12 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3, 4 or 5 R6 groups;
each R6 is independently F, CI, Br, I, CN, N02, N3, C1-C12 alkyl, C2-Ci2 alkenyl, C2-Ci2 alkynyl, C3-Ci2 cycloalkyl, 3-12 membered heterocyclyl, C6-Ci2 aryl, 5-12 membered heteroaryl, -NH2, - NH(Ci-Ci2 alkyl), -NH(CH2)„-(C3-Ci2 cycloalkyl), -NH(CH2)„-(3-12 membered heterocyclyl), - NH(CH2)„-(C6-Ci2 aryl), -NH(CH2)„-(5-12 membered heteroaryl), -N(Ci-Ci2 alkyl)2, -N[(CH2)„-(C3- C12 cycloalkyl)]2, -N[(CH2)„-(3-12 membered heterocyclyl)]2, -N[(CH2)„-(C6-Ci2 aryl)]2, -N[(CH2)„- (5-12 membered heteroaryl)]2, OH, -0(Ci-Ci2 alkyl), -0(CH2)„-C3-Ci2 cycloalkyl), -0(CH2)„-(3-12 membered heterocyclyl), -0(CH2)n-(C6-Ci2 aryl) or -0(CH2)n-(5-12 membered heteroaryl);
each Ra, Rb and Rc is independently H, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, -(C1-C4 alkylene)-(C3-C6 cycloalkyl), 3-7 membered heterocyclyl, -(C1-C4 alkylene)-(3-7 membered heterocyclyl), C6-Cio aryl, -(C1-C4 alkylene)-(C6-Cio aryl), 5-10 membered heteroaryl or - (C1-C4 alkylene)-(5-10 membered heteroaryl), or Ra and Rb taken together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 substituents independently selected from F, CI, CN, N3, OH, NH2, Ci-Ce alkyl, Ci-Ce haloalkyl, Ci-Ce alkoxy and Ci-Ce alkylamino;
each m is independently 0, 1 or 2; and
each n is independently 0, 1 , 2, 3 or 4.
2. The compound of claim 1, wherein R7 is H, N02, C3-C6 cycloalkyl, -ORc, -C(=0)Rc, - OC(=0)Rc, -C(=0)ORc or -C(=0)NRaRb, with the proviso that when X is H, R7 is C3-C6 cycloalkyl, or when each of X and R7 is H, Z has formula (Z-34):
Figure imgf000119_0001
(Z-34) wherein R7 is optionally substituted with 1 , 2 or 3 R6 groups.
3. The compound of claim 1 having Formula (II):
Figure imgf000119_0002
wherein X is -NRaRb, -N(Ra)C(=0)Rc, -N(Rc)C(=0)NRaRb, -N(Ra)C(=0)ORc or -N(Ra)S(=0)mRc.
4. The compound of claim 1 or 3, wherein Z is C3-C6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z is optionally substituted with 1 , 2, 3 or 4 R1 groups.
5. The compound of claim 1 or 3, wherein Z1 is H, C1-C4 alkyl, C3-C6 cycloalkyl or 4-7 membered heterocyclyl, wherein Z1 is optionally substituted with 1 , 2, 3 or 4 R2 groups.
6. The compound of claim 1 or 3, wherein each R1 and R2 is independently H, F, N3, CN, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Cs cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, -(CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)mRc, -S(=0)2NRaRb, -C(=0)Rc, - N(Ra)C(=0)Rc, -(CR4R5)„C(=0)NRaRb, -N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, wherein each R1 and R2 is optionally independently substituted with 1, 2, 3 or 4 R3 groups.
7. The compound of claim 1 or 3, wherein each R3 is independently H, F, CI, CN, N3, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Cs cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, -(CR4R5)„-ORc, -(CR4R5)„-NRaRb, -S(=0)2NRaRb, -C(=0)Rc, -N(Ra)C(=0)Rc, - (CR4R5)„C(=0)NRaRb, -N(Rc)C(=0)NRaRb, -N(Ra)S(=0)mRc or -C(=0)NRaRb, or two adjacent R3 taken together with the atoms to which they are attached form a C3-C6 cycloalkyl, 3-7 membered heterocyclyl, phenyl or 5-6 membered heteroaryl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 R6 groups.
8. The compound of claim 1 or 3, wherein each R4 and R5 is independently H, F, CI, Br, I, N3, CN, OH, NH2, Ci-Ce alkyl, Ci-Ce alkoxy, Ci-Ce alkylamino, C2-C6 alkenyl, C2-C6 alkynyl, C3-C6 cycloalkyl, 3-7 membered heterocyclyl, phenyl or 5-6 membered heteroaryl, or R4 and R5 taken together with the carbon atom to which they are attached form a C3-C6 cycloalkyl or 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2, 3 or 4 R6 groups.
9. The compound of claim 1 or 3, wherein each R6 is independently F, CI, CN, N3, Ci-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-Cs cycloalkyl, 3-7 membered heterocyclyl, phenyl, 5-6 membered heteroaryl, NH2, -NH(Ci-C6 alkyl), -NH(CH2)„-(C3-C6 cycloalkyl), -NH(CH2)„-(3-7 membered heterocyclyl), -NH(CH2)n-phenyl, -NH(CH2)n-(5-6 membered heteroaryl), -N(Ci-C6 alkyl)2, - N[(CH2)„-(C3-C6 cycloalkyl)]2, -N[(CH2)„-(3-7 membered heterocyclyl)] 2, -N[(CH2)„-phenyl]2, - N[(CH2)„-(5-6 membered heteroaryl)]2, OH, -0(Ci-C6 alkyl), -0(CH2)„-(C3-C6 cycloalkyl), -0(CH2)„- (3-7 membered heterocyclyl), -0(CH2)n-phenyl or -0(CH2)n-(5-6 membered heteroaryl).
10. The compound of claim 1 or 3, wherein each Ra, Rb and Rc is independently H, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, C3-C6 cycloalkyl, -(C1-C2 alkylene)-(C3-C6 cycloalkyl), 3-7 membered heterocyclyl, -(C1-C2 alkylene)-(3-7 membered heterocyclyl), phenyl, -(C1-C2 alkylene) -phenyl, 5-6 membered heteroaryl or -(C1-C2 alkylene)-(5-6 membered heteroaryl), or Ra and Rb taken together with the nitrogen atom to which they are attached form a 3-7 membered heterocyclyl group, wherein each of the above substituents is optionally independently substituted with 1, 2 or 3 substitutents independently selected from F, CI, CN, N3, OH, NH2, C1-C4 alkyl, C1-C4 haloalkyl, C1-C4 alkoxy and C1-C4 alkylamino.
11. The compound of claim 1 or 3, wherein Z is C4-C6 cycloalkyl, or 4-, 5- or 6-membered heterocyclyl, wherein Z is optionally substituted with 1 , 2, 3 or 4 R1 groups.
12. The compound of claim 1 or 3, wherein Z is :
Figure imgf000120_0001
(Z-1 ) (Z-2) (Z-3) (Z-4) (Z-5) (Z-6) (Z-7) (Z-8)
Figure imgf000121_0001
Figure imgf000121_0002
(Z-17) (Z-18) (Z-19) (Z-20) (Z-21 ) (Z-22) (Z-23) (Z-24)
Figure imgf000121_0003
(Z-25) , (Z-26) , (Z-27) , (Z-28) , (Z-29) , (Z-30) , (Z-31 ) , (Z-32)
Figure imgf000121_0004
(Z-33) nr (Z-34)
or a stereoisomer thereof, wherein Z is optionally substituted by 1, 2 or 3 R1 groups.
13. The compound of claim 1 or 3, wherein Z1 is H, methyl, ethyl, propyl, isopropyl or cyclopropyl.
14. The compound of claim 1 or 3, wherein X is H, -NH2, -NHMe, -NHC(=0)Me, - NHC(=0)NHMe or -NHC(=0)NMe2.
15. The compound of claim 1 or 3, wherein each Ra, Rb and Rc is independently H, methyl, ethyl, propyl, isopropyl, cyclopropyl or butyl, wherein each Ra, Rb and Rc is optionally independently substituted with 1, 2 or 3 substitutents independently selected from F, CI, CN, N3, OH, NH2, methyl, ethyl, -CF3, -OCH3 and -CH3NH2.
16. The compound of claim 1 or 3 having one of the following structures:
Figure imgf000122_0001
Figure imgf000122_0002
Figure imgf000122_0003
121
Figure imgf000123_0001
(17) or (18)
17. A pharmaceutical composition comprising the compound of any one of claims 1 to 16, and a pharmaceutically acceptable excipient, carrier, adjuvant, vehicle or a combination thereof.
18. The pharmaceutical composition of claim 17 further comprising a therapeutic agent selected from the group consisting of chemotherapeutic agents, anti-proliferative agents, phosphodiesterase 4 (PDE4) inhibitors, 2-adrenoreceptor agonists, corticosteroids, non-steroidal GR agonists, anticholinergic agents, antihistamines and combinations thereof.
19. A method of preventing, treating or lessening the severity of a JAK-mediated disease in a patient by administering to the patient with the compound of any one of claims 1 to 16 or the pharmaceutical composition of any one of claims 17 to 18.
20. The method of claim 19, wherein the JAK-mediated disease is a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease or a transplantation rejection.
21. The method of claim 19, wherein the JAK-mediated disease is cancer, polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), chronic obstruction pulmonary disease (COPD), asthma, systemic lupus erythematosis, cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis, type I diabetes mellitus, allergic airway disease, sinusitis, eczema, hives, food allergies, allergies to insect venom, inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection or cell transplant rejection.
22. The compound of any one of claims 1 to 16 or the pharmaceutical composition of any one of claims 17 to 18 for use in preventing, treating or lessening the severity of a JAK- mediated disease in a patient.
23. The compound or pharmaceutical composition of claim 22, wherein the JAK-mediated disease is a proliferative disease, an autoimmune disease, an allergic disease, an inflammatory disease or a transplantation rejection.
24. The compound or pharmaceutical composition of claim 22, wherein the JAK-mediated disease is cancer, polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), chronic obstruction pulmonary disease (COPD), asthma, systemic and cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis, type I diabetes mellitus, allergic airway disease, sinusitis, eczema, hives, food allergies, allergies to insect venom, inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection or cell transplant rejection.
25. Use of the compound of any one of claims 1 to 16 or the pharmaceutical composition of any one of claims 17 to 18 in the manufacture of a medicament for preventing or treating cancer, polycythemia vera, essential thrombocytosis, myelofibrosis, chronic myelogenous leukemia (CML), chronic obstruction pulmonary disease (COPD), asthma, systemic and cutaneous lupus erythematosis, lupus nephritis, dermatomyositis, Sjogren's syndrome, psoriasis, type I diabetes mellitus, allergic airway disease, sinusitis, eczema, hives, food allergies, allergies to insect venom, inflammatory bowel syndrome, Chron's disease, rheumatoid arthritis, juvenile arthritis, psoriatic arthritis, organ transplant rejection, tissue transplant rejection or cell transplant rejection.
26. A method of modulating the activity of Janus kinase with the compound of any one of claims 1 to 16, or the pharmaceutical composition of any one of claims 17 to 18.
27. Use of the compound of any one of claims 1 to 16 or the pharmaceutical composition of any one of claims 17 to 18 in the manufacture of a medicament for modulating the activity of Janus kinase.
28. The compound of any one of claims 1 to 16 or the pharmaceutical composition of any one of claims 17 to 18 for use in modulating the activity of Janus kinase.
PCT/US2014/063971 2013-11-15 2014-11-05 Substituted heteroaryl compounds and methods of use WO2015073267A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361904460P 2013-11-15 2013-11-15
US61/904,460 2013-11-15

Publications (1)

Publication Number Publication Date
WO2015073267A1 true WO2015073267A1 (en) 2015-05-21

Family

ID=53057884

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/063971 WO2015073267A1 (en) 2013-11-15 2014-11-05 Substituted heteroaryl compounds and methods of use

Country Status (1)

Country Link
WO (1) WO2015073267A1 (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9346815B2 (en) 2014-05-23 2016-05-24 Genentech, Inc. 5-chloro-2-difluoromethoxyphenyl pyrazolopyrimidine compounds, compositions and methods of use thereof
WO2017161116A1 (en) 2016-03-17 2017-09-21 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as pi3k kinase inhibitors
WO2017176960A1 (en) * 2016-04-06 2017-10-12 Lysosomal Therapeutics Inc. Pyrazolo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US9868742B2 (en) 2016-05-05 2018-01-16 Lysosomal Therapeutics Inc. Substituted imidazo[1,2-b]pyridazines, substituted imidazo[1,5-b] pyridazines, related compounds, and their use in the treatment of medical disorders
US9920061B2 (en) 2016-04-06 2018-03-20 Lysosomal Therapeutics Inc. Imidazo[1,5-A]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US10246466B2 (en) 2014-01-24 2019-04-02 Tp Therapeutics, Inc. Diaryl macrocycles as modulators of protein kinases
US10307426B2 (en) 2017-05-22 2019-06-04 Genentech, Inc. Therapeutic compounds and compositions, and methods of use thereof
US10316044B2 (en) 2015-07-02 2019-06-11 Tp Therapeutics, Inc. Chiral diaryl macrocycles as modulators of protein kinases
CN109952303A (en) * 2016-10-14 2019-06-28 林伯士拉克许米公司 TYK2 inhibitor and application thereof
US10570135B2 (en) 2014-11-06 2020-02-25 Lysosomal Therapeutics Inc. Substituted pyrazolo[1,5-A]pyrimidines and their use in the treatment of medical disorders
US10751341B2 (en) 2014-11-06 2020-08-25 Lysosomal Therapeutics Inc. Substituted pyrrolo[1,2-a]pyrimidines and their use in the treatment of medical disorders
US10786508B2 (en) 2014-11-06 2020-09-29 Lysosomal Therapeutics Inc. Substituted imidazo[1,5-A]-pyrimidines and their use in the treatment of medical disorders
US11124516B2 (en) 2016-04-06 2021-09-21 BIAL-BioTech Investments, Inc. Pyrrolo[1,2-A]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
JP2022502515A (en) * 2018-10-15 2022-01-11 ニンバス ラクシュミ, インコーポレイテッド TYK2 inhibitors and their use
US11345698B2 (en) 2016-05-05 2022-05-31 Bial—R&D Investments, S.A. Substituted imidazo[1,2-a]pyridines, substituted imidazo[1,2-a]pyrazines, related compounds, and their use in the treatment of medical disorders
WO2022169921A1 (en) 2021-02-04 2022-08-11 Bristol-Myers Squibb Company Benzofuran compounds as sting agonists
RU2799332C2 (en) * 2016-04-06 2023-07-04 Биал - Ар Энд Ди Инвестментс, С.А. Pyrazolo[1,5-a]pyrimidinylcarboxamides and their use for the treatment of pathological conditions
US11981684B2 (en) 2017-07-28 2024-05-14 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7528138B2 (en) * 2004-11-04 2009-05-05 Vertex Pharmaceuticals Incorporated Pyrazolo[1,5-a]pyrimidines useful as inhibitors of protein kinases
WO2010051549A1 (en) * 2008-10-31 2010-05-06 Genentech, Inc. Pyrazolopyrimidine jak inhibitor compounds and methods

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7528138B2 (en) * 2004-11-04 2009-05-05 Vertex Pharmaceuticals Incorporated Pyrazolo[1,5-a]pyrimidines useful as inhibitors of protein kinases
WO2010051549A1 (en) * 2008-10-31 2010-05-06 Genentech, Inc. Pyrazolopyrimidine jak inhibitor compounds and methods

Cited By (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10618912B2 (en) 2014-01-24 2020-04-14 Turning Point Therapeutics, Inc. Diaryl macrocycles as modulators of protein kinases
US10246466B2 (en) 2014-01-24 2019-04-02 Tp Therapeutics, Inc. Diaryl macrocycles as modulators of protein kinases
US9346815B2 (en) 2014-05-23 2016-05-24 Genentech, Inc. 5-chloro-2-difluoromethoxyphenyl pyrazolopyrimidine compounds, compositions and methods of use thereof
US9604984B2 (en) 2014-05-23 2017-03-28 Genentech, Inc. 5-chloro-2-difluoromethoxyphenyl pyrazolopyrimidine compounds, compositions and methods of use thereof
US11932645B2 (en) 2014-11-06 2024-03-19 Bial—R & D Investments, S.A. Substituted pyrazolo[1,5-a]pyrimidines and their use in the treatment of medical disorders
US11351173B2 (en) 2014-11-06 2022-06-07 Bial—R&D Investments, S.A. Substituted pyrrolo[1,2-a]pyrimidines and their use in the treatment of medical disorders
US11091492B2 (en) 2014-11-06 2021-08-17 Bial—R&D Investments, S.A. Substituted pyrazolo[1,5-a]pyrimidines and their use in the treatment of medical disorders
US11400095B2 (en) 2014-11-06 2022-08-02 Bial—R&D Investments, S.A. Substituted imidazo[1,5-a]pyrimidines and their use in the treatment of medical disorders
US10786508B2 (en) 2014-11-06 2020-09-29 Lysosomal Therapeutics Inc. Substituted imidazo[1,5-A]-pyrimidines and their use in the treatment of medical disorders
US10570135B2 (en) 2014-11-06 2020-02-25 Lysosomal Therapeutics Inc. Substituted pyrazolo[1,5-A]pyrimidines and their use in the treatment of medical disorders
US10751341B2 (en) 2014-11-06 2020-08-25 Lysosomal Therapeutics Inc. Substituted pyrrolo[1,2-a]pyrimidines and their use in the treatment of medical disorders
US10316044B2 (en) 2015-07-02 2019-06-11 Tp Therapeutics, Inc. Chiral diaryl macrocycles as modulators of protein kinases
WO2017161116A1 (en) 2016-03-17 2017-09-21 Infinity Pharmaceuticals, Inc. Isotopologues of isoquinolinone and quinazolinone compounds and uses thereof as pi3k kinase inhibitors
US10787454B2 (en) 2016-04-06 2020-09-29 BIAL—BioTech Investments, Inc. Imidazo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US11192892B2 (en) 2016-04-06 2021-12-07 Bial—R&D Investments, S.A. Substituted pyrazolo[1,5-a]pyrimidines for the treatment of medical disorders
WO2017176960A1 (en) * 2016-04-06 2017-10-12 Lysosomal Therapeutics Inc. Pyrazolo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
JP2019510790A (en) * 2016-04-06 2019-04-18 リソソーマル・セラピューティクス・インコーポレイテッドLysosomal Therapeutics Inc. Pyrazolo [1,5-a] pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
CN109311887A (en) * 2016-04-06 2019-02-05 溶酶体治疗股份有限公司 Pyrazolo [1,5-a] pyrimidine radicals benzamide compound and they treatment medical conditions in purposes
IL262177A (en) * 2016-04-06 2018-11-29 Lysosomal Therapeutics Inc Pyrazolo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorder
US10934298B2 (en) 2016-04-06 2021-03-02 BIAL—BioTech Investments, Inc. Substituted pyrazolo[1,5-a]pyrimidines for the treatment of medical disorders
US9920061B2 (en) 2016-04-06 2018-03-20 Lysosomal Therapeutics Inc. Imidazo[1,5-A]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US11124516B2 (en) 2016-04-06 2021-09-21 BIAL-BioTech Investments, Inc. Pyrrolo[1,2-A]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
AU2017246455B2 (en) * 2016-04-06 2021-09-30 Bial - R&D Investments, S.A. Pyrazolo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
RU2799332C2 (en) * 2016-04-06 2023-07-04 Биал - Ар Энд Ди Инвестментс, С.А. Pyrazolo[1,5-a]pyrimidinylcarboxamides and their use for the treatment of pathological conditions
US11453675B2 (en) 2016-04-06 2022-09-27 Bial—R&D Investments, S.A. Imidazo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
US9840510B1 (en) 2016-04-06 2017-12-12 Lysosomal Therapeutics Inc. Pyrazolo[1,5-a]pyrimidinyl carboxamide compounds and their use in the treatment of medical disorders
JP7038063B2 (en) 2016-04-06 2022-03-17 リソソーマル・セラピューティクス・インコーポレイテッド Pyrazolo [1,5-a] pyrimidinylcarboxamide compounds and their use in the treatment of medical disorders
US11345698B2 (en) 2016-05-05 2022-05-31 Bial—R&D Investments, S.A. Substituted imidazo[1,2-a]pyridines, substituted imidazo[1,2-a]pyrazines, related compounds, and their use in the treatment of medical disorders
US9868742B2 (en) 2016-05-05 2018-01-16 Lysosomal Therapeutics Inc. Substituted imidazo[1,2-b]pyridazines, substituted imidazo[1,5-b] pyridazines, related compounds, and their use in the treatment of medical disorders
US11168087B2 (en) 2016-05-05 2021-11-09 Bial—R&D Investments, S.A. Substituted imidazo[1,2-b]pyridazines, substituted imidazo[1,5-b]pyridazines, related compounds, and their use in the treatment of medical disorders
US11878979B2 (en) 2016-05-05 2024-01-23 Bial—R&D Investments, S.A. Substituted imidazo[1,2-b]pyridazines, substituted imidazo[1,5-b]pyridazines, related compounds, and their use in the treatment of medical disorders
US11220508B2 (en) 2016-10-14 2022-01-11 Nimbus Lakshmi, Inc. TYK2 inhibitors and uses thereof
CN109952303A (en) * 2016-10-14 2019-06-28 林伯士拉克许米公司 TYK2 inhibitor and application thereof
US10307426B2 (en) 2017-05-22 2019-06-04 Genentech, Inc. Therapeutic compounds and compositions, and methods of use thereof
US11981684B2 (en) 2017-07-28 2024-05-14 Turning Point Therapeutics, Inc. Macrocyclic compounds and uses thereof
JP2022502515A (en) * 2018-10-15 2022-01-11 ニンバス ラクシュミ, インコーポレイテッド TYK2 inhibitors and their use
WO2022169921A1 (en) 2021-02-04 2022-08-11 Bristol-Myers Squibb Company Benzofuran compounds as sting agonists

Similar Documents

Publication Publication Date Title
US9399637B2 (en) Substituted heteroaryl compounds and methods of use
WO2015073267A1 (en) Substituted heteroaryl compounds and methods of use
US10059689B2 (en) Substituted heteroaryl compounds and methods of use
WO2016000615A1 (en) Heteroaryl compounds and pharmaceutical applications thereof
WO2015094803A1 (en) Substituted heteroaryl compounds and methods of use
US10266521B2 (en) Substituted heteroaryl compounds and methods of use
WO2016190847A1 (en) Substituted heteroaryl compounds and methods of use
US10683297B2 (en) Substituted heteroaryl compounds and methods of use
WO2018169700A1 (en) Substituted heteroaryl compounds and methods of use

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: 14862642

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14862642

Country of ref document: EP

Kind code of ref document: A1