WO2005096784A2 - Benzotriazine inhibitors of kinases - Google Patents

Benzotriazine inhibitors of kinases Download PDF

Info

Publication number
WO2005096784A2
WO2005096784A2 PCT/US2005/012057 US2005012057W WO2005096784A2 WO 2005096784 A2 WO2005096784 A2 WO 2005096784A2 US 2005012057 W US2005012057 W US 2005012057W WO 2005096784 A2 WO2005096784 A2 WO 2005096784A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mmol
subject
disorder
methyl
Prior art date
Application number
PCT/US2005/012057
Other languages
French (fr)
Other versions
WO2005096784A3 (en
Inventor
Glenn Noronha
Kathy Barrett
Jianguo Cao
Colleen Gritzen
Xianchang Gong
John Hood
Chi Ching Mak
Andrew Mcpherson
Ved Prakash Pathak
Joel Renick
Richard Soll
Ute Splittgerber
Wolfgang Wrasidlo
Binqi Zeng
Ningning Zhao
Elena Dneprovskaia
Original Assignee
Targegen, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Targegen, Inc. filed Critical Targegen, Inc.
Priority to AU2005231507A priority Critical patent/AU2005231507B2/en
Priority to NZ551027A priority patent/NZ551027A/en
Priority to CN200580018660.1A priority patent/CN101426772B/en
Priority to CA2567574A priority patent/CA2567574C/en
Priority to EP05762774.7A priority patent/EP1809614B1/en
Publication of WO2005096784A2 publication Critical patent/WO2005096784A2/en
Priority to IL178908A priority patent/IL178908A/en
Priority to HK08100766.4A priority patent/HK1110578A1/en
Publication of WO2005096784A3 publication Critical patent/WO2005096784A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D253/00Heterocyclic compounds containing six-membered rings having three nitrogen atoms as the only ring hetero atoms, not provided for by group C07D251/00
    • C07D253/08Heterocyclic compounds containing six-membered rings having three nitrogen atoms as the only ring hetero atoms, not provided for by group C07D251/00 condensed with carbocyclic rings or ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond

Definitions

  • the present invention relates generally to the use of compounds to treat a variety of disorders, diseases and pathologic conditions and more specifically to the use of benzotriazine compounds to treat disorders.
  • c-Src plays a major role in the growth, progression and metastasis of a large number of cancers.
  • c-Src can be the transforming element of the oncogenic Rous sarcoma retrovirus. Subsequently, it has been demonstrated that c-Src kinase can have the oncogenic potential. Gene knockout experiments suggest that inhibition of some members of the Src family might have potential therapeutic benefit.
  • Tyrosine kinases phosphorylate tyrosine residues in peptides and proteins. These enzymes are key elements in the regulation of cell signaling including cell proliferation and cell differentiation.
  • Protein TKs comprise the receptor TKs, including the epidermal growth family members (HERl and HER2 for example), platelet derived growth factor (PDGF) and kinases that play a role in angiogenesis (Tie-2 and KDR for example), and the cellular or non-receptor kinases, which include members of the Src family.
  • c-Src TK is one of three members of the Src family expressed ubiquitously.
  • c-Src is expressed at low levels in most cell types and, in the absence of the appropriate extracellular stimuli, maintained in an inactive conformation through phosphorylation of a regulatory tyrosine domain at Tyr530.
  • Activation of c-Src occurs through dephosphorylation of the Tyr530 site and phosphorylation of a second tyrosine, Tyr419, present in the kinase domain of the enzyme.
  • Src kinase modulates signal transduction through multiple oncogenic pathways, including EGFR, HER2, PDGFR, FGFR and NEGFR.
  • blocking signaling through the inhibition of the kinase activity of Src can be an effective means of modulating aberrant pathways that drive the oncogenic transformation of cells.
  • c-Src TK activity has been associated with adhesion and cytoskeletal changes both in tumor cells and otherwise, ultimately resulting in an invasive phenotype that may be motile.
  • c-Src TK activity has been shown to be an important component in the epithelial to mesenchymal transition that occurs in the early stages of invasion of carcinoma cells.
  • c-Src activity is also known to be essential in the turnover of local adhesions, a critical cell-motility component.
  • c-Src inhibition markedly reduces the rate of lymph and liver metastases.
  • Clinical data supports the link between misregulated Src activity and the increased invasive potential of tumor cells, hi colon tumors, increased c-Src TK activity has been shown to correlate to tumor progression, with the highest activity found in metastatie tissue. Increased Src activity in colon tumors might be an indicator of poor prognosis.
  • enhancement of Src kinase activity has been reported, and in transitional cell carcinoma of the bladder, c-Src activity peaked as superficial tumors became muscle invasive.
  • Increased c-Src TK activity results in breakdown of the E-cadherin-mediated epithelial cell-cell adhesion, which can be restored by Src inhibition.
  • Intimate connections between increased NEGF activity, Src activity, and cellular barrier function related to vascular leak have been also demonstrated.
  • Inhibition of Src results in decrease in vascular leak when exogenous NEGF is administered in in vivo studies. Examples where excessive vascular permeability leads to particularly deleterious effects include pulmonary edema, cerebral edema, and cardiac edema.
  • NEGF-mediated edema has been shown to involve intracellular signaling by Src family kinases, protein kinase C, and Akt kinase.
  • Rho-associated kinases have been linked to thrombin-mediated vascular leakage, and protein kinase C to T ⁇ F-induced leakage.
  • MLCK myosin light chain kinase
  • MLC myosin light chain
  • a general approach to the inhibition of vascular leakage can be to interfere with any of the underlying mechanistic pathways, whether by inhibition of kinase signaling or the intercellular contractile apparatus or other cellular processes. This can then lead to potential treatments for edema and its associated pathologies. For example, inhibiting edema formation should be beneficial to overall patient outcome in situations such as inflammation, allergic diseases, cancer, cerebral stroke, myocardial infarction, pulmonary and cardiac insufficiency, renal failure, and retinopathies, to name a few. Furthermore, as edema is a general consequence of tissue hypoxia, it can also be concluded that inhibition of vascular leakage represents a potential approach to the treatment of tissue hypoxia.
  • interruption of blood flow by pathologic conditions such as thrombus formation
  • medical intervention such as cardioplegia, organ transplantation, and angioplasty
  • inhibitors of vascular leakage especially as in the case of Src inhibitors.
  • a small molecule inhibitor of c-Src can be beneficial for the treatment of several disease states.
  • the present invention provides methods of use for certain chemical compounds such as kinase inhibitors for treatment of various diseases, disorders, and pathologies, for example, cancer, and vascular disorders, such as myocardial infarction (MI), stroke, or ischemia.
  • chemical compounds such as kinase inhibitors for treatment of various diseases, disorders, and pathologies, for example, cancer, and vascular disorders, such as myocardial infarction (MI), stroke, or ischemia.
  • the benzotriazine compounds described in this invention may block the enzymatic activity of some or many of the members of the Src family, in addition to blocking the activity of other receptor and non-receptor kinases.
  • Such compounds may be beneficial for treatment of the diseases where disorders affect cell motility, adhesion, and cell cycle progression, and in addition, diseases with related hypoxic conditions, osteoporosis and conditions, which result from or are related to increases in vascular permeability, inflammation or respiratory distress, tumor growth, invasion, angiogenesis, metastases and apoptosis.
  • kinase inhibitors that can be used to bring about beneficial therapeutic results include inhibitors of Src kinase.
  • each of A can be (CH)o- ⁇ . N, N ⁇ , O, S, or a part of a ring fusion to form a second ring, where the second ring can be an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic;
  • each of B can be (CH)o- ⁇ , N, NH, O, S, ora part of a ring fusion to form a second ring, where the second ring can be an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic, with the further proviso that if each B is (CH)o, R 3 can be any substitutent described below other than hydrogen bonded directly to the position 7 of the adjacent ring;
  • R 0 can be H or lower alkyl
  • L can be a bond, or a substituted or unsubstituted alkyl, alkenyl, or alkynyl linking moiety
  • Ri can be C(R*) 3 , OR, N(R') 2 . NR'C(O)R, NR'C(O)O(R), NR'C(O)N(R') 2 , SR', C(O)(O)R, C(O), C(O)N(R') 2 , SO 3 R, OSO 2 R', SO 2 R, SOR, S(O)N(R) 2 , OS(O)(O)N(R') 2 , S(O)(O)N(R * ) 2 , S(O)N(R) 2 , PO 4 R', OPO 2 R, PO 3 R', PO 2 R', or a 3-6 membered heterocycle with one or more heterocyclic atoms, with each heteroatom being capable of carrying any R' group on it, wherein R' can be hydrogen, lower alkyl, alkyl- hydroxyl, thiol-alkyl, alkyl-thiol, aminoal
  • R 2 is a substitutent situated at position 5,6 or 8 of the ring, wherein R 2 can be methyl, ethyl, n-propyl, wopropyl, n-butyl, z ' so-butyl, tert-butyl, tso-pentyl, phenyl, substituted phenyl, halogen, branched or unbranched alkylamino, branched or unbranched aminoalkyl, branched or unbranched alkyloxo, branched or unbranched oxyalkyl, branched or unbranched thioalkyl, branched or unbranched alkylthiol, CF 3 , sulfonamido, substituted sulfonamido, sulfonate, sulfonate ester, phosphate, phosphate ester, phosphonate, phosphonate ester, carboxo, amido, ureido
  • R 3 can be hydrogen, alkyl, alkoxy, halogen, CF 3 , cyano, substituted alkyl, or hydroxyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, C(R") 3 , OR", N(R") 2 , NR"C(O)R", NR"C(O)NR", R", C(O)(O)R", OC(O)R", C(O)N(R") 2 , C(O), OC(O)N(R") 2 , SO 3 R", OSO 2 R", SO 2 R", SOR", PO 4 R", OPO 2 R", PO 3 R", PO 2 R", wherein R" can be hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, lower alkyl, branched lower alkyl, alkyl-hydroxyl, branched alkyl-hydroxyl, amino-alkyl, branched amino-alkyl, alkyl-a
  • articles of manufacture including packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material includes a label which indicates that the pharmaceutical composition can be used for treatment of disorders associated with compromised vasculostasis, and wherein the pharmaceutical composition includes at least one compound of structure (I).
  • articles of manufacture including packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material includes a label which indicates that the pharmaceutical composition can be used for treatment of disorders associated with vascular permeability leakage or compromised vasculostasis, such as myocardial infarction, stroke, congestive heart failure, an ischemia or reperfusion injury, cancer, arthritis or other arthropathy, retinopathy or vitreoretinal disease, macular degeneration, autoimmune disease, vascular leakage syndrome, inflammatory disease, edema, transplant rejection, burn, or acute or adult respiratory distress syndrome (ARDS) and wherein the pharmaceutical composition includes at least one compound of structure (I).
  • disorders associated with vascular permeability leakage or compromised vasculostasis such as myocardial infarction, stroke, congestive heart failure, an ischemia or reperfusion injury, cancer, arthritis or other arthropathy, retinopathy or vitreoretinal disease, macular degeneration, autoimmune disease, vascular leakage syndrome, inflammatory
  • a disorder associated with compromised vasculostasis including the administration of a therapeutically effective amount of at least one compound of structure (I) or pharmaceutically acceptable salts, hydrates, solvates, crystal forms and individual diastereomers thereof, to a subject in need of such treatment.
  • methods of treating a disorder associated with compromised vasculostasis including the administration of a therapeutically effective amount of at least one compound of structure (I), or pharmaceutically acceptable salts, hydrates, solvates, crystal forms and individual diastereomers thereof, in combination with an anti-inflammatory, chemotherapeutic agent, immunomodulatory agent, therapeutic antibody or a protein kinase inhibitor, to a subject in need of such treatment.
  • methods of treating a subject having or at risk of having a disorder selected from myocardial infarction, vascular leakage syndrome (NLS), cancer, stroke, ARDS, burns, arthritis, edema, retinopathy or vitreoretinal disease, ischemic or reperfusion related tissue injury or damage, autoimmune disease, transplant rejection, inflammatory disease including administering to the subject a therapeutically effective amount of at least one compound of structure (I), thereby treating the subject.
  • a disorder selected from myocardial infarction, vascular leakage syndrome (NLS), cancer, stroke, ARDS, burns, arthritis, edema, retinopathy or vitreoretinal disease, ischemic or reperfusion related tissue injury or damage, autoimmune disease, transplant rejection, inflammatory disease
  • processes for making a pharmaceutical composition including combining a combination of at least one compound of structure (I) or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof and a pharmaceutically acceptable carrier.
  • heterocyclic when used to describe an aromatic ring, refer to the aromatic ring containing at least one heteroatom.
  • heteroatom refers to any atom other than carbon, for example, N, O, or S.
  • aromatic refers to a cyclically conjugated molecular entity with a stability, due to delocalization, significantly greater than that of a hypothetical localized structure, such as the Kekule structure.
  • heterocyclic when not used to describe an aromatic ring, refers to cyclic (i.e., ring-containing) groups other than aromatic groups, the cyclic group being formed by between 3 and about 14 carbon atoms and at least one heteroatom described above.
  • substituted heterocyclic refers, for both aromatic and non-aromatic structures, to heterocyclic groups further bearing one or more substituents described above.
  • alkyl refers to a monovalent straight or branched chain hydrocarbon group having from one to about 12 carbon atoms, for example, methyl, ethyl, n-propyl, wopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl (also known as n- amyl), n-hexyl, and the like.
  • substituted alkyl refers to alkyl groups further bearing one or more substituents such as hydroxy, alkoxy, mercapto, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryloxy, substituted aryloxy, halogen, cyano, nitro, amino, amido, aldehyde, acyl, oxyacyl, carboxyl, sulfonyl, sulfonamide, sulfuryl, and the like.
  • substituents such as hydroxy, alkoxy, mercapto, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryloxy, substituted aryloxy, halogen, cyano, nitro, amino, amido, aldehyde, acyl,
  • lower alkyl refers to alkyl groups having from 1 to about 6 carbon atoms.
  • alkenyl refers to straight-chained or branched hydrocarbyl groups having at least one carbon-carbon double bond, and having between about 2 and about 12 carbon atoms
  • substituted alkenyl refers to alkenyl groups further bearing one or more substituents described above.
  • alkynyl refers to straight-chained or branched hydrocarbyl groups having at least one carbon-carbon triple bond, and having between about 2 and about 12 carbon atoms
  • substituted alkynyl refers to alkynyl groups further bearing one or more substituents described above.
  • aryl refers to aromatic groups having between about 5 and about 14 carbon atoms and the term “substituted aryl” refers to aryl groups further bearing one or more substituents described above.
  • heteroaryl refers to aromatic rings, where the ring structure is formed by between 3 and about 14 carbon atoms and by at least one heteroatom described above, and the term “substituted heteroaryl” refers to heteroaryl groups further bearing one or more substituents described above.
  • alkoxy refers to the moiety — O— alkyl, wherein alkyl is as defined above, and the term “substituted alkoxy” refers to alkoxy groups further bearing one or more substituents described above.
  • cycloalkyl refers to alkyl groups having between 3 and about 8 carbon atoms arranged as a ring, and the term “substituted cycloalkyl” refers to cycloalkyl groups further bearing one or more substituents described above.
  • alkylaryl refers to alkyl-substituted aryl groups and the term “substituted alkylaryl” refers to alkylaryl groups further bearing one or more substituents described above.
  • arylalkyl refers to aryl-substituted alkyl groups and the term “substituted arylalkyl” refers to arylalkyl groups further bearing one or more substituents described above.
  • arylalkenyl refers to aryl-substituted alkenyl groups and the term “substituted arylalkenyl” refers to arylalkenyl groups further bearing one or more substituents described above.
  • arylalkynyl refers to aryl-substituted alkynyl groups and the term “substituted arylalkynyl” refers to arylalkynyl groups further bearing one or more substituents described above.
  • arylene refers to divalent aromatic groups having between 5 and about 14 carbon atoms and the term “substituted arylene” refers to arylene groups further bearing one or more substituents described above.
  • kinase refers to any enzyme that catalyzes the addition of phosphate groups to a protein residue; for example, serine and threonine kinases catalyze the addition of phosphate groups to serine and threonine residues.
  • Src kinase refers to the related homologs or analogs belonging to the mammalian family of Src kinases, including, for example, c-Src, Fyn, Yes and Lyn kinases and the hematopoietic-restricted kinases Hck, Fgr, Lck and Blk.
  • Src kinase signaling pathway and “Src cascade” refer to both the upstream and downstream components of the Src signaling cascade.
  • terapéuticaally effective amount refers to the amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician, e.g., restoration or maintenance of vasculostasis or prevention of the compromise or loss or vasculostasis; reduction of tumor burden; reduction of morbidity and/or mortality.
  • composition refers to the fact that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • administration of a compound or “administering a compound” refer to the act of providing a compound of the invention or pharmaceutical composition to the subject in need of treatment.
  • antibody refers to intact molecules of polyclonal or monoclonal antibodies, as well as fragments thereof, such as Fab and F(ab') 2 , Fv and SCA fragments which are capable of binding an epitopic determinant.
  • vasculostasis refers to the maintenance of the homeostatic vascular functioning leading to the normal physiologic functioning.
  • vasculostatic agents refers to agents that seek to address conditions in which vasculostasis is compromised by preventing the loss of or restoring or maintaining vasculostasis.
  • compounds having the structure (I) are provided for treatment of various diseases, disorders, and pathologies, as well as pharmaceutically acceptable salts, hydrates, solvates, crystal forms, N-oxides, and individuals diastereoners of compounds having the structure (I):
  • each of A can be (CH) 0 _ ⁇ , N, NH, O, S, or a part of a ring fusion to form a second ring, where the second ring can be an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic;
  • each of B can be (CH) 0 - ⁇ , N, MI, O, S, ora part of a ring fusion to form a second ring, where the second ring can be an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic, with the further proviso that if each B is (CH) 0 , R 3 can be any substitutent described below, other than hydrogen, bonded directly to the position 7 of the adjacent ring;
  • R 0 can be H or lower alkyl
  • L can be a bond, or a substituted or unsubstituted alkyl, alkenyl, or alkynyl linking moiety
  • Ri can be C(R') 3 , OR', N(R) 2 , NR'C(O)R, NR'C(O)O(R')- NR'C(O)N(R')2, SR, C(O)(O)R', C(O), C(O)N(R) 2 , SO 3 R, OSO 2 R, SO 2 R', SOR, S(O)N(R') 2 , OS(O)(O)N(R') 2 , S(O)(O)N(R') 2 .
  • R can be hydrogen, lower alkyl, alkyl- hydroxyl, thiol-alkyl, alkyl-thiol, aminoalkyl, alkylamino, branched alkyl, branched alkyl hydroxyl, branched thio-alkyl, branched alkyl-thiol, branched aminoalkyl, branched alkylamino, or a closed 3-6 membered carbocycle or heterocycle, with each heteroatom in the 3-6 membered heterocycle being capable of carrying any R' group on it, and wherein each R can be independent in case there is more than one R';
  • R 2 is a substitutent situated at position 5,6 or 8 of the ring, wherein R 2 can be methyl, ethyl, n-propyl, z ' sopropyl, n-butyl, iso-butyl, tert-butyl, tsopentyl, phenyl, substituted phenyl, halogen, branched or unbranched alkylamino, branched or unbranched aminoalkyl, branched or unbranched alkyloxo, branched or unbranched oxyalkyl, branched or unbranched thioalkyl, branched or unbranched alkylthiol, CF 3 , sulfonamido, substituted sulfonamido, sulfonate, sulfonate ester, phosphate, phosphate ester, phosphonate, phosphonate ester, carboxo, amido, ureido,
  • R 3 can be hydrogen, alkyl, alkoxy, halogen, CF 3 , cyano, substituted alkyl, or hydroxyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, C(R") , OR", N(R") 2 , NR"C(O)R", NR"C(O)NR", R", C(O)(O)R", OC(O)R", C(O)N(R") 2 , C(O), OC(O)N(R") 2 , SO 3 R", OSO 2 R", SO 2 R", SOR", PO 4 R", OPO 2 R", PO 3 R", PO 2 R", wherein R" can be hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, lower alkyl, branched lower alkyl, alkyl-hydroxyl, branched alkyl-hydroxyl, amino-alkyl, branched amino-alkyl, alkyl-amin
  • n is an integer that can have value between 1 and 5, with the further proviso that if n > 2, then each group R is independent of the other groups R 3 ,
  • the compounds and methods of the present invention are useful in treating a variety of disorders associated with compromised vasculostasis and other disorders, including but not limited to, for example: stroke, cardiovascular disease, myocardial infarction, congestive heart failure, cardiomyopathy, myocarditis, ischemic heart disease, coronary artery disease, cardiogenic shock, vascular shock, pulmonary hypertension, pulmonary edema (including cardiogenic pulmonary edema), cancer, pleural effusions, rheumatoid arthritis, diabetic retinopathy, retinitis pigmentosa, and retinopathies, including diabetic retinopathy and retinopathy of prematurity, inflammatory diseases, restenosis, edema (including edema associated with pathologic situations such as cancers and edema induced by medical interventions such as chemotherapy), asthma, acute or adult respiratory distress syndrome
  • agents e.g., chemotherapeutic agents or protein therapeutic agents described below
  • T-cell mediated hypersensitivity diseases including contact hypersensitivity, delayed- type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); Type 1 diabetes; psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' disease; Addison's disease (autoimmune disease of the adrenal glands); autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; cancers, including those where kinases such as Src-family kinases are activated or overexpressed, such as colon carcinoma and thymoma, or cancers where kinase activity facilitates tumor growth or survival; glomerulonephritis, serum sickness; uticaria; allergic diseases such as respiratory allergies
  • Src-family tyrosine kinases other than Lck are important in the Fc gamma receptor induced respiratory burst of neutrophils as well as the Fc gamma receptor responses of monocytes and macrophages.
  • the compositions and methods of the present invention may be useful in inhibiting the Fc gamma induced respiratory burst response in neutrophils, and may also be useful in inhibiting the Fc gamma dependent production of TNF alpha.
  • the ability to inhibit Fc gamma receptor dependent neutrophil, monocyte and macrophage responses would result in additional anti-inflammatory activity for the compounds employed in invention methods.
  • compositions and methods of the present invention may also be useful in the treatment of autoimmune glomerulonephritis and other instances of glomerulonephritis induced by deposition of immune complexes in the kidney that trigger Fc gamma receptor responses and which can lead to kidney damage.
  • Fc epsilon receptor induced degranulation of mast cells and basophils that plays an important role in asthma, allergic rhinitis, and other allergic disease.
  • Fc epsilon receptors are stimulated by IgE-antigen complexes.
  • Compounds employed in the methods of the present invention may inhibit the Fc epsilon induced degranulation responses.
  • the ability to inhibit Fc epsilon receptor dependent mast cell and basopbil responses may result in additional anti-inflammatory activity for the present compounds beyond their effect on T cells.
  • the present invention also provides articles of manufacture comprising packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material comprises a label which indicates that the pharmaceutical composition can be used for treatment of disorders and wherein the pharmaceutical composition comprises a compound according to the present invention.
  • the invention provides a pharmaceutical composition including a therapeutic agent and a compound of the invention, wherein the compound is present in a concentration effective to reduce vascular leakage associated with indications or therapeutic agents which have vascular leak as a side effect.
  • administration of a compound of the invention can be in conjunction with E -2, immunotoxins, antibodies or chemotherapeutics.
  • IL-2, immunotoxin, antibody or chemotherapeutic concentration can be determined by one having ordinary skill in the art according to standard treatment regimen or, for example, as determined by an in vivo animal assay.
  • the present invention also provides pharmaceutical compositions comprising IL-2, immunotoxin, antibody or chemotherapeutic and at least one invention compound in an amount effective for inhibiting vascular permeability, and a pharmaceutically acceptable vehicle or diluent.
  • the compositions of the present invention may contain other therapeutic agents, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques known in the art of pharmaceutical formulation.
  • the compounds of the invention may be formulated into therapeutic compositions as natural or salt forms.
  • Pharmaceutically acceptable non-toxic salts include the base addition salts (formed with free carboxyl or other anionic groups) which may be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino-ethanol, histidine, procaine, and the like.
  • Such salts may also be formed as acid addition salts with any free cationic groups and will generally be formed with inorganic acids such as, for example, hydrochloric, sulfuric, or phosphoric acids, or organic acids such as acetic, citric, p-toluenesulfonic, methanesulfonic acid, oxalic, tartaric, mandelic, and the like.
  • Salts of the invention include amine salts formed by the protonation of an amino group with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like.
  • Salts of the invention also include amine salts formed by the protonation of an amino group with suitable organic acids, such as p-toluenesulfonic acid, acetic acid, and the like.
  • suitable organic acids such as p-toluenesulfonic acid, acetic acid, and the like.
  • Additional excipients which are contemplated for use in the practice of the present invention are those available to those of ordinary skill in the art, for example, those found in the United States Pharmacopeia Vol. XXII and National Formulary Vol. XVII, U.S. Pharmacopeia Convention, Inc., Rockville, MD (1989), the relevant contents of which is incorporated herein by reference, i addition, polymorphs of the invention compounds are included in the present invention.
  • compositions of the invention may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, intrathecal, or intracisternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents.
  • suitable means for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, intrathecal, or intracisternal injection or infusion techniques (e.g., as
  • the present compounds may, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps.
  • the present compounds may also be administered liposomally.
  • mammals including, but not limited to, cows, sheep, goats, horses, dogs, cats, guinea pigs, rats or other bovine, ovine, equine, canine, feline, rodent or murine species can be treated.
  • the method can also be practiced in other species, such as avian species (e.g., chickens).
  • compositions for the administration of the compounds of this embodiment either alone or in combination with IL-2, immunotoxin, antibody or chemotherapeutic may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients.
  • the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation.
  • the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases.
  • compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated to form osmotic therapeutic tablets for control release.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxy- propylmethylcellulose, sodium alginate, polyvinyl-pyrrolidorie, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbit
  • the aqueous suspensions may also contain one or more preservatives, for example ethyl, or n- propyl, ?-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
  • Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives.
  • a dispersing or wetting agent e.g., sodium EDTA
  • suspending agent e.g., sodium EDTA
  • preservatives e.g., sodium EDTA, sodium bicarbonate, sodium bicarbonate
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • sweetening agents for example glycerol, propylene glycol, sorbitol or sucrose.
  • Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
  • compositions may be in the form of a sterile injectable aqueous or oleagenous suspension.
  • This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a parenterally-acceptable diluent or solvent or cosolvent or complexing agent or dispersing agent or excipient or combination thereof, for example 1,3-butanediol, polyethylene glycols, polypropylene glycols, ethanol or other alcohols, povidones, various brands of TWEEN surfactant, sodium dodecyl sulfate, sodium deoxycholate, dimethylacetamide, polysorbates, poloxamers, cyclodextrins, lipids, and excipients such as inorganic salts (e.g., sodium chloride), buffering agents (e.g., sodium citrate, sodium phosphate), and sugars (e.g., saccharose and dextrose).
  • a parenterally-acceptable diluent or solvent or cosolvent or complexing agent or dispersing agent or excipient or combination thereof for example 1,3-butan
  • Suitable vehicles and solvents that may be employed are water, dextrose solutions, Ringer's solutions and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables.
  • these pharmaceutical compositions may be formulated and administered systemically or locally. Techniques for formulation and administration may be found in the latest edition of "Remington's Pharmaceutical Sciences” (Mack Publishing Co, Easton Pa.). Suitable routes may, for example, include oral or transmucosal administration; as well as parenteral delivery, including intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, or intranasal administration.
  • the pharmaceutical compositions of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiologically buffered saline.
  • penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds maybe prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow, for the preparation of highly concentrated solutions.
  • the compounds of the present invention may also be administered in the form of suppositories for rectal administration of the drug.
  • These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and polyethylene glycols.
  • creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention are employed.
  • topical application shall include mouthwashes and gargles).
  • the invention compounds are administered in combination with an anti-inflammatory agent, antihistamines, chemotherapeutic agent, immunomodulator , therapeutic antibody or a protein kinase inhibitor, e.g., a tyrosine kinase inhibitor, to a subject in need of such treatment.
  • chemotherapeutic agents include antimetabolites, such as methotrexate, DNA cross-linking agents, such as cisplatin/carboplatin; alkylating agents, such as canbusil; topoisomerase I inhibitors such as dactinomicin; microtubule inhibitors such as taxol (paclitaxol), and the like.
  • chemotherapeutic agents include, for example, a vinca alkaloid, mitomycin-type antibiotic, bleomycin-type antibiotic, antifolate, colchicine, demecoline, etoposide, taxane, anthracycline antibiotic, doxorubicin, daunorubicin, carminomycin, epirubicin, idarubicin, mithoxanthrone, 4-dimethoxy-daunomycin, 11-deoxydaunorabicin, 13- deoxydaunorubicin, adriamycin-14-benzoate, adriamycin-14-octanoate, adriamycin-14- naphthaleneacetate, amsacrine, carmustine, cyclophosphamide, cytarabine, etoposide, lovastatin, melphalan, topetecan, oxalaplatin, chlorambucil, methtrex
  • therapeutic antibodies include antibodies directed against the HER2 protein, such as trastuzumab; antibodies directed against growth factors or growth factor receptors, such as bevacizumab, which targets vascular endothelial growth factor, and OSI-774, which targets epidermal growth factor; antibodies targeting integrin receptors, such as Nitaxin (also known as MEDI-522), and the like.
  • Classes of anticancer agents suitable for use in compositions and methods of the present invention include, but are not limited to: 1) alkaloids, including, microtubule inhibitors (e.g., Nincristine, Vinblastine, and Vindesine, etc.), microtubule stabilizers (e.g., Paclitaxel [Taxol], and Docetaxel, Taxotere, etc.), and chromatin function inhibitors, including, topoisomerase inhibitors, such as, epipodophyllotoxins (e.g., Etoposide [VP-16], and Teniposide [VM- 26], etc.), and agents that target topoisomerase I (e.g., Camptothecin and Isirinotecan [CPT-11], etc.); 2) covalent D ⁇ A-binding agents [alkylating agents], including, nitrogen mustards (e.g., Mechlorethamine, Chlorambucil, Cyclophosphamide, Ifosphamide, and Busulfan [
  • the pharmaceutical composition and method of the present invention may further comprise other therapeutically active compounds as noted herein which are usually applied in the treatment of the above mentioned pathological conditions.
  • other therapeutic agents include the following: cyclosporins (e.g., cyclosporin A), CTLA4-Ig, antibodies such as ICAM-3, anti-IL-2 receptor (Anti-Tac), anti-CD45RB, anti-CD2, anti-CD3 (OKT-3), anti-CD4, anti-CD80, anti-CD86, agents blocking the interaction between CD40 and gp39, such as antibodies specific for CD40 and/or gp39 (i.e., CD154), fusion proteins constructed from CD40 and gp39 (CD40Ig and CD8gp39), inhibitors, such as nuclear translocation inhibitors, of NF-kappa B function, such as deoxyspergualin (DSG), cholesterol biosynthesis inhibitors such as HMG CoA reductase inhibitors (lovastatin and simvastatin), non-steroidal anti
  • cytokine encompasses chemokines, interleukins, lymphokines, monokines, colony stimulating factors, and receptor associated prqteins, and functional fragments thereof.
  • functional fragment refers to a polypeptide or peptide which possesses biological function or activity that is identified through a defined functional assay.
  • the cytokines include endothelial monocyte activating polypeptide II (EMAP- II), granulocyte-macrophage-CSF (GM-CSF), granulocyte-CSF (G-CSF), macrophage- CSF (M-CSF), IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-12, and IL-13, interferons, and the like and which is associated with a particular biologic, morphologic, or phenotypic alteration in a cell or cell mechanism.
  • EMP- II endothelial monocyte activating polypeptide II
  • GM-CSF granulocyte-macrophage-CSF
  • G-CSF granulocyte-CSF
  • M-CSF macrophage- CSF
  • IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-12, and IL-13 interferons
  • an appropriate dosage level can generally be between about 0.01 and about 500 mg per 1 kg of patient body weight per day which can be administered in single or multiple doses.
  • the dosage level can be between about 0.01 and about 250 mg/kg per day; more narrowly, between about 0.5 and about 100 mg/kg per day.
  • a suitable dosage level can be between about 0.01 and about 250 mg/kg per day, between about 0.05 and about 100 mg/kg per day, or between about 0.1 and about 50 mg/kg per day, or about 1.0 mg/kg per day.
  • the dosage can be between about 0.05 and about 0.5 mg/kg per day, or between about 0.5 and about 5 mg/kg per day, or between about 5 and about 50 mg/kg per day.
  • the compositions can be provided in the form of tablets containing between about 1.0 and about 1,000 mg of the active ingredient, for example, about 1.0, about 5.0, about 10.0, about 15.0, about'20.0, about 25.0, about 50.0, about 75.0, about lOO.O, about 150.0, about 200.0, about 250.0, about 300.0, about 400.0, about 500.0, about 600.0, about 750.0, about 800.0, about 900.0, and about 1,000.0 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the compounds can be administered on a regimen of 1 to 4 times per day, such as once or twice per day. There may be a period of no administration followed by another regimen of administration.
  • administration of the compound is closely associated with the schedule of IL- 2 administration.
  • administration can be prior to, simultaneously with or immediately following IL-2 administration.
  • Compounds of the present invention can be used, alone or in combination with an effective amount of a therapeutic antibody (or therapeutic fragment thereof), a chemotherapeutic or an immunotoxic agent, for treatment of tumors. While doxorubicin, docetaxel, or taxol are described in the present application as illustrative examples of chemotherapeutic agents, it should be understood that the invention includes combination therapy including a compound of the invention, including but not limited to vasculostatic agents, such as tyrosine, serine or threonine kinase inhibitors, for example, Src-family inhibitors, and any chemotherapeutic agent or therapeutic antibody.
  • vasculostatic agents such as tyrosine, serine or threonine kinase inhibitors, for example, Src-family inhibitors, and any chemotherapeutic agent or therapeutic antibody.
  • Reverse-phase HPLC chromatography was carried out on Gilson 215 liquid handler equipped with Waters SymmetryShieldTM RP18 7 ⁇ m (40 x 100mm) Prep-Pak cartridge.
  • Mobile phase consisted of standard acetonitrile (ACN) and DI Water, each with 0.1% TFA added. Purification was carried out at a flow rate of 40mL/ min, and a gradient such that the peak of interest was eluted between 12-15 min in a 30 min run.
  • one of three methods A, B, or C can be used for synthesizing some of the compounds of the present invention.
  • Those having ordinary skill in the art can determine, depending on variety of factors, including the particular compound that is sought to be made, whether to selected the method A, B or C.
  • the synthetic method A is shown by the reaction scheme (II). About 1 equivalent of compound 1 was mixed with 2 equivalent of cynamide in a vial. The mixture was heated to about 100°C until the mixture was completely melted. The mixture was cooled down to room temperature and concentrated HCI was added. The mixture was then again heated at about 100°C for about 40 minutes and cooled down in ice water. About 14 moles of NaOH were carefully added to the above reaction mixture followed by heating the mixture at about 100°C for about 2 hours, and by cooling down to room temperature.
  • Compound 3 was dissolved in N, N-dimethylacetamide in a vial with a septum. Catalytic amount of 10% palladium on carbon was added to the mixture. A balloon filled with hydrogen was placed on the top of the vial. The mixture was stirred at room temperature for about 2 hours. Celite was used to remove the palladium and carbon. Solvent was removed under vacuum and compound 4, shown by the reaction scheme (II), was isolated.
  • the synthetic method B is shown by the reaction scheme (III).
  • Compounds 1, 2, 3, and 4 were consecutively prepared and isolated as described in Method A. About 1 equivalent of compound 4 was dissolved in an aniline followed by adding about 2 equivalents of sulfamic acid. The mixture was heated at about 200°C overnight. Compound 5 was isolated by HPLC.
  • the synthetic method C is shown by the reaction scheme (IN).
  • Compounds 1, 2, 3, and 4 were consecutively prepared and isolated as described in Method A. About 1 equivalent of compound 4 was dissolved in substituted phenylarnine followed by adding about 2 equivalent of sulfamic acid. The mixture was heated at about 200°C overnight.
  • Compound 5 was isolated by HPLC and was dissolved in dry CH 2 CI 2 . The mixture was cooled to about -78°C using a dry ice-acetone bath. About 2 equivalents of BBr 3 (IM solution in CH 2 C1 2 ) was added dropwise to the mixture at about -78°C under nitrogen atmosphere.
  • a balloon filled with hydrogen was placed on the top of the vial.
  • the mixture was stirred at room temperature for about 2 hours.
  • Celite was used to remove the palladium and carbon.
  • the crude product and about 200 mg (0.74 mmol) of l-[2-(4- bromo-phenoxy)-ethyl]-pyrrolidine were dissolved in 10 ml of toluene.
  • About 17 mg (0.018 mmol) of Pd(dba) 3 , about 34 mg (0.054 mmol) of BINAP, and about 50 mg (0.226 mmol) of KOt-Bu were added to the solution.
  • the mixture was kept at about 100°C for about 24 hours under argon.
  • the crude product was purified by preparative HPLC.
  • the reactants were flushed with argon and diluted with ethylene glycol dimethyl ether (6 mL), ethanol (1 mL) and DI water (1 mL).
  • the reaction vessel was outfitted with condenser and refluxed for 18 hours.
  • the reaction was cooled to ambient temperature and the crude product was filtered, diluted with ethyl acetate, and washed with brine.
  • the brine layer was back extracted once with fresh ethyl acetate.
  • the organic phases were combined and dried over sodium sulfate (Na 2 SO 4 ). Filtration was followed by evaporation and silica gel chromatography (2:3 EtOAc/hexanes) provided the desired product as a yellow powder (0.08 g, 74% yield).
  • the reactants were flushed with argon, diluted with dioxane (6 mL) and outfitted with reflux condenser. The reaction was heated to reflux for 18 hours, filtered hot, diluted with ethyl acetate and washed with brine. The brine layer was back extracted once with fresh ethyl acetate. The organic phases were combined and dried over sodium sulfate (Na 2 SO 4 ). Filtration followed by evaporation provided crude product, whichwas dissolved in minimum amount of DCM and precipitated out with excess hexanes. Solids were filtered off and dried to yield a yellow powder (0.06 g, 91% yield).
  • the reactants were flushed with argon and diluted with ethylene glycol dimethyl ether (20 mL), ethanol (5 mL) and DI water (5 mL).
  • the reaction vessel was outfitted with condenser and refluxed for 4 hours.
  • the reaction was filtered hot and diluted with ethyl acetate.
  • the Organic layer was isolated and concentrated to a dark residue. This was dissolved in DMF (6 mL) and slowly diluted with water so as to precipitate out the product. Solids were filtered off and dried to yield an orange solid (1 g, 82% yield).

Abstract

The invention provides benzotriazine compounds having formula (I). The benzotriazine compounds of the invention are capable of inhibiting kinases, such members of the Src kinase family, and various other specific receptor and non-receptor kinases.

Description

BENZOTRIAZINE INHIBITORS OF KINASES
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the benefit of priority under 35 U.S.C. § 119(e) of patent applications U.S. Serial Number 60/561,237 filed April 8, 2004, and 60/643,439 filed January 12, 2005, the entire content of which is incorporated herein by reference.
FIELD OF THE INVENTION
[0002] The present invention relates generally to the use of compounds to treat a variety of disorders, diseases and pathologic conditions and more specifically to the use of benzotriazine compounds to treat disorders.
BACKGROUND
[0003] c-Src plays a major role in the growth, progression and metastasis of a large number of cancers. c-Src can be the transforming element of the oncogenic Rous sarcoma retrovirus. Subsequently, it has been demonstrated that c-Src kinase can have the oncogenic potential. Gene knockout experiments suggest that inhibition of some members of the Src family might have potential therapeutic benefit.
[0004] Tyrosine kinases (TKs) phosphorylate tyrosine residues in peptides and proteins. These enzymes are key elements in the regulation of cell signaling including cell proliferation and cell differentiation. Protein TKs comprise the receptor TKs, including the epidermal growth family members (HERl and HER2 for example), platelet derived growth factor (PDGF) and kinases that play a role in angiogenesis (Tie-2 and KDR for example), and the cellular or non-receptor kinases, which include members of the Src family. [0005] c-Src TK is one of three members of the Src family expressed ubiquitously. c-Src is expressed at low levels in most cell types and, in the absence of the appropriate extracellular stimuli, maintained in an inactive conformation through phosphorylation of a regulatory tyrosine domain at Tyr530. Activation of c-Src occurs through dephosphorylation of the Tyr530 site and phosphorylation of a second tyrosine, Tyr419, present in the kinase domain of the enzyme.
[0006] Src kinase modulates signal transduction through multiple oncogenic pathways, including EGFR, HER2, PDGFR, FGFR and NEGFR. Thus, blocking signaling through the inhibition of the kinase activity of Src can be an effective means of modulating aberrant pathways that drive the oncogenic transformation of cells.
[0007] There exists a body of evidence of misregulated increased kinase activity of c- Src in several human tumor types, most notably colon and breast tumors. Misregulated c- Src TK activity has also been associated with adhesion and cytoskeletal changes both in tumor cells and otherwise, ultimately resulting in an invasive phenotype that may be motile. c-Src TK activity has been shown to be an important component in the epithelial to mesenchymal transition that occurs in the early stages of invasion of carcinoma cells. c-Src activity is also known to be essential in the turnover of local adhesions, a critical cell-motility component. In in vivo models of metastases, c-Src inhibition markedly reduces the rate of lymph and liver metastases. Clinical data supports the link between misregulated Src activity and the increased invasive potential of tumor cells, hi colon tumors, increased c-Src TK activity has been shown to correlate to tumor progression, with the highest activity found in metastatie tissue. Increased Src activity in colon tumors might be an indicator of poor prognosis. In breast and ovarian cancers, enhancement of Src kinase activity has been reported, and in transitional cell carcinoma of the bladder, c-Src activity peaked as superficial tumors became muscle invasive.
[0008] Biochemically, cellular stimuli that lead to Src activation result in increased association between Src and the cytoskeleton. As a result, Src mediates the phosphorylation of many intracellular substrates such as EGFR, FAK, PYK2, paxillin, Stat3, and cyclin D. The biological effects of these interactions affect cell motility, adhesion, cell cycle progression, and apoptosis and might have some connection to the disease related effects stated above. Thus, Src plays a role in responses to regional hypoxia, limited nutrients, and internal cellular effects to self-destruct.
[0009] Increased c-Src TK activity results in breakdown of the E-cadherin-mediated epithelial cell-cell adhesion, which can be restored by Src inhibition. Intimate connections between increased NEGF activity, Src activity, and cellular barrier function related to vascular leak have been also demonstrated. Inhibition of Src results in decrease in vascular leak when exogenous NEGF is administered in in vivo studies. Examples where excessive vascular permeability leads to particularly deleterious effects include pulmonary edema, cerebral edema, and cardiac edema.
[0010] The cascade of events leading to loss of endothelial barrier function is complex and incompletely understood. Data support some role for kinases in this process. For example, NEGF-mediated edema has been shown to involve intracellular signaling by Src family kinases, protein kinase C, and Akt kinase. Rho-associated kinases have been linked to thrombin-mediated vascular leakage, and protein kinase C to TΝF-induced leakage. Kinases are believed to mediate the phosphorylation of junctional proteins such as beta-catenin and vascular endothelial NE-cadherin, leading to the dissolution of adherens junctions and the dissociation of cadherin-catenin complexes from their cytoskeletal anchors. Proteins which regulate the intercellular contractile machinery such as myosin light chain kinase (MLCK) and myosin light chain (MLC) are also activated, resulting in cellular contraction, and therefore an opening of intercellular junctions.
[0011] A general approach to the inhibition of vascular leakage can be to interfere with any of the underlying mechanistic pathways, whether by inhibition of kinase signaling or the intercellular contractile apparatus or other cellular processes. This can then lead to potential treatments for edema and its associated pathologies. For example, inhibiting edema formation should be beneficial to overall patient outcome in situations such as inflammation, allergic diseases, cancer, cerebral stroke, myocardial infarction, pulmonary and cardiac insufficiency, renal failure, and retinopathies, to name a few. Furthermore, as edema is a general consequence of tissue hypoxia, it can also be concluded that inhibition of vascular leakage represents a potential approach to the treatment of tissue hypoxia. For example, interruption of blood flow by pathologic conditions (such as thrombus formation) or medical intervention (such as cardioplegia, organ transplantation, and angioplasty) could be treated both acutely and prophylactically using inhibitors of vascular leakage, especially as in the case of Src inhibitors.
[0012] Accordingly, a small molecule inhibitor of c-Src can be beneficial for the treatment of several disease states.
SUMMARY
[0013] The present invention provides methods of use for certain chemical compounds such as kinase inhibitors for treatment of various diseases, disorders, and pathologies, for example, cancer, and vascular disorders, such as myocardial infarction (MI), stroke, or ischemia.
[0014] The benzotriazine compounds described in this invention may block the enzymatic activity of some or many of the members of the Src family, in addition to blocking the activity of other receptor and non-receptor kinases. Such compounds may be beneficial for treatment of the diseases where disorders affect cell motility, adhesion, and cell cycle progression, and in addition, diseases with related hypoxic conditions, osteoporosis and conditions, which result from or are related to increases in vascular permeability, inflammation or respiratory distress, tumor growth, invasion, angiogenesis, metastases and apoptosis.
[0015] According to the embodiments of the invention, some examples of kinase inhibitors that can be used to bring about beneficial therapeutic results include inhibitors of Src kinase. [0016] According to one embodiment of the invention, there are provided compounds having the structure (I), and pharmaceutically acceptable salts, hydrates, solvates, crystal forms, N-oxides, and individuals diastereoners thereof:
Figure imgf000006_0001
(D
[0017] wherein each of A can be (CH)o-ι. N, NΗ, O, S, or a part of a ring fusion to form a second ring, where the second ring can be an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic;
[0018] each of B can be (CH)o-ι, N, NH, O, S, ora part of a ring fusion to form a second ring, where the second ring can be an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic, with the further proviso that if each B is (CH)o, R3 can be any substitutent described below other than hydrogen bonded directly to the position 7 of the adjacent ring;
[0019] R0 can be H or lower alkyl;
[0020] L can be a bond, or a substituted or unsubstituted alkyl, alkenyl, or alkynyl linking moiety;
[0021] Ri can be C(R*)3, OR, N(R')2. NR'C(O)R, NR'C(O)O(R), NR'C(O)N(R')2, SR', C(O)(O)R, C(O), C(O)N(R')2, SO3R, OSO2R', SO2R, SOR, S(O)N(R)2, OS(O)(O)N(R')2, S(O)(O)N(R*)2, S(O)N(R)2, PO4R', OPO2R, PO3R', PO2R', or a 3-6 membered heterocycle with one or more heterocyclic atoms, with each heteroatom being capable of carrying any R' group on it, wherein R' can be hydrogen, lower alkyl, alkyl- hydroxyl, thiol-alkyl, alkyl-thiol, aminoalkyl, alkylamino, branched alkyl, branched alkyl hydroxyl, branched thio-alkyl, branched alkyl-thiol, branched aminoalkyl, branched alkylamino, or a closed 3-6 membered carbocycle or heterocycle, with each heteroatom in the 3-6 membered heterocycle being capable of carrying any R group on it, and wherein each R1 can be independent in case there is more than one R';
[0022] R2 is a substitutent situated at position 5,6 or 8 of the ring, wherein R2 can be methyl, ethyl, n-propyl, wopropyl, n-butyl, z'so-butyl, tert-butyl, tso-pentyl, phenyl, substituted phenyl, halogen, branched or unbranched alkylamino, branched or unbranched aminoalkyl, branched or unbranched alkyloxo, branched or unbranched oxyalkyl, branched or unbranched thioalkyl, branched or unbranched alkylthiol, CF3, sulfonamido, substituted sulfonamido, sulfonate, sulfonate ester, phosphate, phosphate ester, phosphonate, phosphonate ester, carboxo, amido, ureido, substituted carboxo, substituted amido, substituted ureido, or a 3-6 membered carbocycle or heterocycle attached to positions 5, 6 or 8 directly or through group L, each heteroatom being capable of carrying any group R2, with the further proviso that either one, two or three substituents R2 can be present in the ring, and each of the substituents R2 can be the same or different;
[0023] R3 can be hydrogen, alkyl, alkoxy, halogen, CF3, cyano, substituted alkyl, or hydroxyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, C(R")3, OR", N(R")2, NR"C(O)R", NR"C(O)NR", R", C(O)(O)R", OC(O)R", C(O)N(R")2, C(O), OC(O)N(R")2, SO3R", OSO2R", SO2R", SOR", PO4R", OPO2R", PO3R", PO2R", wherein R" can be hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, lower alkyl, branched lower alkyl, alkyl-hydroxyl, branched alkyl-hydroxyl, amino-alkyl, branched amino-alkyl, alkyl-amino, branched alkyl-amino, thiol-alkyl, branched thiol-alkyl, alkyl- thiol, branched thiol-alkyl, or may form a closed 3-6 membered heterocycle with one or more heterocyclic atoms, branched alkyl, branched alkyl hydroxyl, where each R" can be independent in case there is more than one R"; [0024] n is an integer that can have value between 1 and 5, with the further proviso that if n > 2, then each group R3 is independent of the other groups R3,
[0025] with the further proviso that if each A is (CH)0, L is a bond.
[0026] In yet another embodiment, there are provided articles of manufacture including packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material includes a label which indicates that the pharmaceutical composition can be used for treatment of disorders associated with compromised vasculostasis, and wherein the pharmaceutical composition includes at least one compound of structure (I).
[0027] In another embodiment, there are provided articles of manufacture including packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material includes a label which indicates that the pharmaceutical composition can be used for treatment of disorders associated with vascular permeability leakage or compromised vasculostasis, such as myocardial infarction, stroke, congestive heart failure, an ischemia or reperfusion injury, cancer, arthritis or other arthropathy, retinopathy or vitreoretinal disease, macular degeneration, autoimmune disease, vascular leakage syndrome, inflammatory disease, edema, transplant rejection, burn, or acute or adult respiratory distress syndrome (ARDS) and wherein the pharmaceutical composition includes at least one compound of structure (I).
[0028] In another embodiment, there are provided methods of treating a disorder associated with compromised vasculostasis, including the administration of a therapeutically effective amount of at least one compound of structure (I) or pharmaceutically acceptable salts, hydrates, solvates, crystal forms and individual diastereomers thereof, to a subject in need of such treatment.
[0029] In yet another embodiment, there are provided methods of treating a disorder associated with compromised vasculostasis including the administration of a therapeutically effective amount of at least one compound of structure (I), or pharmaceutically acceptable salts, hydrates, solvates, crystal forms and individual diastereomers thereof, in combination with an anti-inflammatory, chemotherapeutic agent, immunomodulatory agent, therapeutic antibody or a protein kinase inhibitor, to a subject in need of such treatment.
[0030] In others embodiment, there are provided methods of treating a subject having or at risk of having a disorder selected from myocardial infarction, vascular leakage syndrome (NLS), cancer, stroke, ARDS, burns, arthritis, edema, retinopathy or vitreoretinal disease, ischemic or reperfusion related tissue injury or damage, autoimmune disease, transplant rejection, inflammatory disease, including administering to the subject a therapeutically effective amount of at least one compound of structure (I), thereby treating the subject.
[0031] hi another embodiment, there are provided processes for making a pharmaceutical composition including combining a combination of at least one compound of structure (I) or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof and a pharmaceutically acceptable carrier.
DETAILED DESCRIPTION
A. Terms and Definitions.
[0032] The following terminology and definitions apply as used in the present application, generally in conformity with the terminology recommended by the International Union of Pure and Applied Chemistry (IUPAC):
[0033] The term "heterocyclic," when used to describe an aromatic ring, refer to the aromatic ring containing at least one heteroatom.
[0034] The term "heteroatom" refers to any atom other than carbon, for example, N, O, or S. [0035] The term "aromatic" refers to a cyclically conjugated molecular entity with a stability, due to delocalization, significantly greater than that of a hypothetical localized structure, such as the Kekule structure.
[0036] The term "heterocyclic," when not used to describe an aromatic ring, refers to cyclic (i.e., ring-containing) groups other than aromatic groups, the cyclic group being formed by between 3 and about 14 carbon atoms and at least one heteroatom described above.
[0037] The term "substituted heterocyclic" refers, for both aromatic and non-aromatic structures, to heterocyclic groups further bearing one or more substituents described above.
[0038] The term "alkyl" refers to a monovalent straight or branched chain hydrocarbon group having from one to about 12 carbon atoms, for example, methyl, ethyl, n-propyl, wopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl (also known as n- amyl), n-hexyl, and the like.
[0039] The term "substituted alkyl" refers to alkyl groups further bearing one or more substituents such as hydroxy, alkoxy, mercapto, cycloalkyl, substituted cycloalkyl, heterocyclic, substituted heterocyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, aryloxy, substituted aryloxy, halogen, cyano, nitro, amino, amido, aldehyde, acyl, oxyacyl, carboxyl, sulfonyl, sulfonamide, sulfuryl, and the like.
[0040] The term "lower alkyl" refers to alkyl groups having from 1 to about 6 carbon atoms.
[0041] The term "alkenyl" refers to straight-chained or branched hydrocarbyl groups having at least one carbon-carbon double bond, and having between about 2 and about 12 carbon atoms, and the term "substituted alkenyl" refers to alkenyl groups further bearing one or more substituents described above. [0042] The term "alkynyl" refers to straight-chained or branched hydrocarbyl groups having at least one carbon-carbon triple bond, and having between about 2 and about 12 carbon atoms, and the term "substituted alkynyl" refers to alkynyl groups further bearing one or more substituents described above.
[0043] The term "aryl" refers to aromatic groups having between about 5 and about 14 carbon atoms and the term "substituted aryl" refers to aryl groups further bearing one or more substituents described above.
[0044] The term "heteroaryl" refers to aromatic rings, where the ring structure is formed by between 3 and about 14 carbon atoms and by at least one heteroatom described above, and the term "substituted heteroaryl" refers to heteroaryl groups further bearing one or more substituents described above.
[0045] The term "alkoxy" refers to the moiety — O— alkyl, wherein alkyl is as defined above, and the term "substituted alkoxy" refers to alkoxy groups further bearing one or more substituents described above.
[0046] The term "cycloalkyl" refers to alkyl groups having between 3 and about 8 carbon atoms arranged as a ring, and the term "substituted cycloalkyl" refers to cycloalkyl groups further bearing one or more substituents described above.
[0047] The term "alkylaryl" refers to alkyl-substituted aryl groups and the term "substituted alkylaryl" refers to alkylaryl groups further bearing one or more substituents described above.
[0048] The term "arylalkyl" refers to aryl-substituted alkyl groups and the term "substituted arylalkyl" refers to arylalkyl groups further bearing one or more substituents described above.
[0049] The term "arylalkenyl" refers to aryl-substituted alkenyl groups and the term "substituted arylalkenyl" refers to arylalkenyl groups further bearing one or more substituents described above. [0050] The term "arylalkynyl" refers to aryl-substituted alkynyl groups and the term "substituted arylalkynyl" refers to arylalkynyl groups further bearing one or more substituents described above.
[0051] The term "arylene" refers to divalent aromatic groups having between 5 and about 14 carbon atoms and the term "substituted arylene" refers to arylene groups further bearing one or more substituents described above.
[0052] The term "kinase" refers to any enzyme that catalyzes the addition of phosphate groups to a protein residue; for example, serine and threonine kinases catalyze the addition of phosphate groups to serine and threonine residues.
[0053] The terms "Src kinase," "Src kinase family," and "Src family" refer to the related homologs or analogs belonging to the mammalian family of Src kinases, including, for example, c-Src, Fyn, Yes and Lyn kinases and the hematopoietic-restricted kinases Hck, Fgr, Lck and Blk.
[0054] The terms "Src kinase signaling pathway," and "Src cascade" refer to both the upstream and downstream components of the Src signaling cascade.
[0055] The term "therapeutically effective amount" refers to the amount of the compound or pharmaceutical composition that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by the researcher, veterinarian, medical doctor or other clinician, e.g., restoration or maintenance of vasculostasis or prevention of the compromise or loss or vasculostasis; reduction of tumor burden; reduction of morbidity and/or mortality.
[0056] The term "pharmaceutically acceptable" refers to the fact that the carrier, diluent or excipient must be compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. [0057] The terms "administration of a compound" or "administering a compound" refer to the act of providing a compound of the invention or pharmaceutical composition to the subject in need of treatment.
[0058] The term "antibody" refers to intact molecules of polyclonal or monoclonal antibodies, as well as fragments thereof, such as Fab and F(ab')2, Fv and SCA fragments which are capable of binding an epitopic determinant.
[0059] The term "vasculostasis" refers to the maintenance of the homeostatic vascular functioning leading to the normal physiologic functioning. The term "vasculostatic agents" refers to agents that seek to address conditions in which vasculostasis is compromised by preventing the loss of or restoring or maintaining vasculostasis.
B. Embodiments of the Invention.
[0060] According to an embodiment of the invention, compounds having the structure (I) are provided for treatment of various diseases, disorders, and pathologies, as well as pharmaceutically acceptable salts, hydrates, solvates, crystal forms, N-oxides, and individuals diastereoners of compounds having the structure (I):
Figure imgf000013_0001
(I)
[0061] wherein each of A can be (CH)0_ι, N, NH, O, S, or a part of a ring fusion to form a second ring, where the second ring can be an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic;
[0062] each of B can be (CH)0-ι, N, MI, O, S, ora part of a ring fusion to form a second ring, where the second ring can be an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic, with the further proviso that if each B is (CH)0, R3 can be any substitutent described below, other than hydrogen, bonded directly to the position 7 of the adjacent ring;
[0063] R0 can be H or lower alkyl;
[0064] L can be a bond, or a substituted or unsubstituted alkyl, alkenyl, or alkynyl linking moiety;
[0065] Ri can be C(R')3, OR', N(R)2, NR'C(O)R, NR'C(O)O(R')- NR'C(O)N(R')2, SR, C(O)(O)R', C(O), C(O)N(R)2, SO3R, OSO2R, SO2R', SOR, S(O)N(R')2, OS(O)(O)N(R')2, S(O)(O)N(R')2. S(O)N(R')2, PO4R\ OPO2R', PO3R, PO2R', or a 3-6 membered heterocycle with one or more heterocyclic atoms, with each heteroatom being capable of carrying any R' group on it, wherein R can be hydrogen, lower alkyl, alkyl- hydroxyl, thiol-alkyl, alkyl-thiol, aminoalkyl, alkylamino, branched alkyl, branched alkyl hydroxyl, branched thio-alkyl, branched alkyl-thiol, branched aminoalkyl, branched alkylamino, or a closed 3-6 membered carbocycle or heterocycle, with each heteroatom in the 3-6 membered heterocycle being capable of carrying any R' group on it, and wherein each R can be independent in case there is more than one R';
[0066] R2 is a substitutent situated at position 5,6 or 8 of the ring, wherein R2 can be methyl, ethyl, n-propyl, z'sopropyl, n-butyl, iso-butyl, tert-butyl, tsopentyl, phenyl, substituted phenyl, halogen, branched or unbranched alkylamino, branched or unbranched aminoalkyl, branched or unbranched alkyloxo, branched or unbranched oxyalkyl, branched or unbranched thioalkyl, branched or unbranched alkylthiol, CF3, sulfonamido, substituted sulfonamido, sulfonate, sulfonate ester, phosphate, phosphate ester, phosphonate, phosphonate ester, carboxo, amido, ureido, substituted carboxo, substituted amido, substituted ureido, or a 3-6 membered carbocycle or heterocycle attached to positions 5, 6 or 8 directly or through group L, each heteroatom being capable of carrying any group R2, with the further proviso that either one, two or three substituents R can be present in the ring, and each of the substituents R2 can be the same or different;
[0067] R3 can be hydrogen, alkyl, alkoxy, halogen, CF3, cyano, substituted alkyl, or hydroxyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, C(R") , OR", N(R")2, NR"C(O)R", NR"C(O)NR", R", C(O)(O)R", OC(O)R", C(O)N(R")2, C(O), OC(O)N(R")2, SO3R", OSO2R", SO2R", SOR", PO4R", OPO2R", PO3R", PO2R", wherein R" can be hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, lower alkyl, branched lower alkyl, alkyl-hydroxyl, branched alkyl-hydroxyl, amino-alkyl, branched amino-alkyl, alkyl-amino, branched alkyl-amino, thiol-alkyl, branched thiol-alkyl, alkyl- thiol, branched thiol-alkyl, or may form a closed 3-6 membered heterocycle with one or more heterocyclic atoms, branched alkyl, branched alkyl hydroxyl, where each R" can be independent in case there is more than one R";
[0068] n is an integer that can have value between 1 and 5, with the further proviso that if n > 2, then each group R is independent of the other groups R3,
[0069] with the further proviso that if each A is (CH)0, L is a bond.
[0070] Some exemplary compounds described by formula (I) that can be used include the following compounds:
Figure imgf000015_0001
(IX)
Figure imgf000016_0001
(XI)
Figure imgf000016_0002
(XLNIII)
Figure imgf000016_0003
(XLIX)
Figure imgf000016_0004
(L)
Figure imgf000016_0005
(LII)
Figure imgf000017_0001
(LΠI)
Figure imgf000017_0002
(LIN)
Figure imgf000017_0003
(CXXXII)
Figure imgf000017_0004
(CXXXIV)
Figure imgf000017_0005
(cxxxNiπ)
Figure imgf000018_0001
(CCXLIΠ)
Figure imgf000018_0002
(CCLN)
Figure imgf000018_0003
(CCLNIII)
Figure imgf000018_0004
(CCLX)
Figure imgf000018_0005
(CCLXI)
Figure imgf000019_0001
(CCLXII)
Figure imgf000019_0002
(CCLXIII)
Figure imgf000019_0003
(CCLXN)
Figure imgf000019_0004
(CCLXNI)
Figure imgf000019_0005
(CCLXXII)
Figure imgf000020_0001
(CCLXXNII)
Figure imgf000020_0002
(LI)
Figure imgf000020_0003
(LXX)
Figure imgf000020_0004
(LXXI)
Figure imgf000020_0005
(CXD
Figure imgf000021_0001
(CXIV)
Figure imgf000021_0002
(CXV)
Figure imgf000021_0003
(CCLXXIX)
Figure imgf000021_0004
(LVIII)
Figure imgf000021_0005
(LX)
Figure imgf000022_0001
(LXΠ)
Figure imgf000022_0002
(LXNI)
Figure imgf000022_0003
(LXXIII)
Figure imgf000022_0004
(LXXNI)
Figure imgf000022_0005
(LXXNIII)
Figure imgf000023_0001
(xcπ)
Figure imgf000023_0002
(XCNII)
Figure imgf000023_0003
(XCIX)
Figure imgf000023_0004
(CXNII)
Figure imgf000024_0001
(CXXI)
Figure imgf000024_0002
(CXXIN)
Figure imgf000024_0003
(CXXN)
Figure imgf000024_0004
(CXLNI)
Figure imgf000024_0005
(CL)
Figure imgf000025_0001
(CLI)
Figure imgf000025_0002
(CLNII)
Figure imgf000025_0003
(CLXIN)
Figure imgf000025_0004
(CLXNI)
Figure imgf000025_0005
(CLXXI)
Figure imgf000026_0001
(CLXXII)
Figure imgf000026_0002
(CLXXIII)
Figure imgf000026_0003
(CLXXIN)
Figure imgf000026_0004
(CLXXN)
Figure imgf000026_0005
(CLXXXIN)
Figure imgf000027_0001
(CLXXXNI)
Figure imgf000027_0002
(CXC)
Figure imgf000027_0003
(ccxπ)
Figure imgf000027_0004
(CCXXNI)
Figure imgf000028_0001
(CCXXXI)
Figure imgf000028_0002
(CCXLNII)
Figure imgf000028_0003
(CCXLNIII)
Figure imgf000028_0004
(CCL)
Figure imgf000028_0005
(CCLII)
Figure imgf000029_0001
(CCLXNII)
Figure imgf000029_0002
(CCLXXIII)
Figure imgf000029_0003
(CCLXXN)
Figure imgf000029_0004
(CCLXXX)
Figure imgf000029_0005
(X)
Figure imgf000030_0001
(XII)
Figure imgf000030_0002
(XIII)
Figure imgf000030_0003
(XIN)
Figure imgf000030_0004
(XN)
Figure imgf000030_0005
(XNI)
Figure imgf000031_0001
(XNII)
Figure imgf000031_0002
(XNπi)
Figure imgf000031_0003
(XIX)
Figure imgf000031_0004
(XX)
Figure imgf000031_0005
(XXI)
Figure imgf000031_0006
(XXIV)
Figure imgf000032_0001
(XXV)
Figure imgf000032_0002
(XXVI)
Figure imgf000032_0003
(XXVIII)
Figure imgf000032_0004
(XXIX)
Figure imgf000032_0005
(XXX)
Figure imgf000033_0001
(XLΠ)
Figure imgf000033_0002
(CXXX)
Figure imgf000033_0003
(XXII)
Figure imgf000033_0004
(XLfll)
Figure imgf000033_0005
(XLV)
Figure imgf000034_0001
(XLVI)
Figure imgf000034_0002
(xcrv)
Figure imgf000034_0003
(CLIΠ)
Figure imgf000035_0001
(CLV)
Figure imgf000035_0002
(CCLXXVIII)
Figure imgf000035_0003
(LXXXVII)
Figure imgf000035_0004
(LXXXVΠI)
Figure imgf000035_0005
(LXXXIX)
Figure imgf000036_0001
(XC)
Figure imgf000036_0002
(CXL)
Figure imgf000036_0003
(CXLII)
Figure imgf000036_0004
(CXLIII)
Figure imgf000037_0001
(CXLIV)
Figure imgf000037_0002
(CXCVIII)
Figure imgf000037_0003
(CCV)
Figure imgf000037_0004
(CCVII)
Figure imgf000037_0005
(CCXVII)
Figure imgf000038_0001
(CCLXVIII)
Figure imgf000038_0002
(CCLXX)
Figure imgf000038_0003
(LXXXVI)
Figure imgf000038_0004
(CXXXVI)
Figure imgf000038_0005
(CLXXX)
Figure imgf000039_0001
(CCXLTV)
Figure imgf000039_0002
(CLXIX)
Figure imgf000039_0003
(CCXX)
Figure imgf000039_0004
(CLIX)
Figure imgf000039_0005
(CI)
Figure imgf000040_0001
(CLXX)
Figure imgf000040_0002
(CLXXXII)
Figure imgf000040_0003
(CCXXIII)
Figure imgf000040_0004
(LXNII)
Figure imgf000040_0005
(LXVΠI)
Figure imgf000041_0001
(LXIX)
Figure imgf000041_0002
(CIV)
Figure imgf000041_0003
(CV)
Figure imgf000041_0004
(cv-o
Figure imgf000041_0005
(CVII)
Figure imgf000042_0001
(CVIII)
Figure imgf000042_0002
(CIX)
Figure imgf000042_0003
(CXXVII)
Figure imgf000042_0004
(CXXVIII)
Figure imgf000042_0005
(CLXΠI)
Figure imgf000043_0001
(CLXXVIII)
Figure imgf000043_0002
(CLXXXVIII)
Figure imgf000043_0003
(CXCV)
Figure imgf000043_0004
(CCXL)
Figure imgf000043_0005
(CCXLI)
Figure imgf000044_0001
(CCXLII)
Figure imgf000044_0002
(CCLIII)
Figure imgf000044_0003
(CCLVI)
Figure imgf000044_0004
(CLXI)
Figure imgf000044_0005
(CCLXXI)
Figure imgf000045_0001
(XXXI)
Figure imgf000045_0002
(XXXII)
Figure imgf000045_0003
(XXXIII)
Figure imgf000045_0004
(XXXIV)
Figure imgf000046_0001
(XXXV)
Figure imgf000046_0002
(XXXVI)
Figure imgf000046_0003
(XXXVII)
Figure imgf000046_0004
(XXXVIII)
Figure imgf000047_0001
(XXXIX)
Figure imgf000047_0002
(XL)
Figure imgf000047_0003
(XLI)
Figure imgf000047_0004
(LV)
Figure imgf000048_0001
(LXIII)
Figure imgf000048_0002
(CXX)
Figure imgf000048_0003
(CCXLV)
Figure imgf000048_0004
(CCLI)
Figure imgf000048_0005
(LVΠ)
Figure imgf000049_0001
(LIX)
Figure imgf000049_0002
(LXI)
Figure imgf000049_0003
(LXV)
Figure imgf000049_0004
(LXXII)
Figure imgf000049_0005
(LXXV)
Figure imgf000050_0001
(LXXVII)
Figure imgf000050_0002
(XCI)
Figure imgf000050_0003
(XCVI)
Figure imgf000050_0004
(XCVIII)
Figure imgf000050_0005
(C)
Figure imgf000051_0001
(CII)
Figure imgf000051_0002
(CXVI)
Figure imgf000051_0003
(CXXIII)
Figure imgf000051_0004
(CXLVIII)
Figure imgf000051_0005
(CCXLVI)
Figure imgf000052_0001
(CCXLIX)
Figure imgf000052_0002
(CCLIV)
Figure imgf000052_0003
(CCLIX)
Figure imgf000052_0004
(CCLXIV)
Figure imgf000052_0005
(CLXVΠI)
Figure imgf000053_0001
(cxcπ)
Figure imgf000053_0002
(CCXXXVII)
Figure imgf000053_0003
(CCXXXVIII)
Figure imgf000053_0004
(CCXXXIX)
Figure imgf000053_0005
(CCLXXIV)
Figure imgf000054_0001
(LXXIX)
Figure imgf000054_0002
(LXXX)
Figure imgf000054_0003
(LXXXI)
Figure imgf000054_0004
(LXXXII)
Figure imgf000054_0005
(LXXXIII) and
Figure imgf000055_0001
(CLXXVI)
[0071] The compounds and methods of the present invention, either when administered alone or in combination with other agents (e.g., chemotherapeutic agents or protein therapeutic agents described below) are useful in treating a variety of disorders associated with compromised vasculostasis and other disorders, including but not limited to, for example: stroke, cardiovascular disease, myocardial infarction, congestive heart failure, cardiomyopathy, myocarditis, ischemic heart disease, coronary artery disease, cardiogenic shock, vascular shock, pulmonary hypertension, pulmonary edema (including cardiogenic pulmonary edema), cancer, pleural effusions, rheumatoid arthritis, diabetic retinopathy, retinitis pigmentosa, and retinopathies, including diabetic retinopathy and retinopathy of prematurity, inflammatory diseases, restenosis, edema (including edema associated with pathologic situations such as cancers and edema induced by medical interventions such as chemotherapy), asthma, acute or adult respiratory distress syndrome (ARDS), lupus, vascular leakage, transplant (such as organ transplant, acute transplant or hetero graft or homo graft (such as is employed in burn treatment)) rejection; protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, transplantation tolerance induction; ischemic or reperfusion injury following angioplasty; arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis; inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic lupus crythematosis); graft vs. host diseases; T-cell mediated hypersensitivity diseases, including contact hypersensitivity, delayed- type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); Type 1 diabetes; psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' disease; Addison's disease (autoimmune disease of the adrenal glands); autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; cancers, including those where kinases such as Src-family kinases are activated or overexpressed, such as colon carcinoma and thymoma, or cancers where kinase activity facilitates tumor growth or survival; glomerulonephritis, serum sickness; uticaria; allergic diseases such as respiratory allergies (asthma, hayfever, allergic rhinitis) or skin allergies; mycosis fungoides; acute inflammatory responses (such as acute or adult respiratory distress syndrome and ischemia/reperfusion injury); dermatomyositis; alopecia areata; chronic actinic dermatitis; eczema; Behcet's disease; Pustulosis palmoplanteris; Pyoderma gangrenum; Sezary's syndrome; atopic dermatitis; systemic schlerosis; morphea; peripheral limb ischemia and ischemic limb disease; bone disease such as osteoporosis, osteomalacia, hyperparathyroidism, Paget's disease, and renal osteodystrophy; vascular leak syndromes, including vascular leak syndromes induced by chemotherapies or immunomodulators such as IL-2; spinal cord and brain injury or trauma; glaucoma; retinal diseases, including macular degeneration; vitreoretinal disease; pancreatitis; vasculatides, including vasculitis, Kawasaki disease, thromboangiitis obliterans, Wegener's granulomatosis, and Behcet's disease; scleroderma; preeclampsia; thalassemia; Kaposi's sarcoma; von Hippel Lindau disease; and the like.
[0072] Src-family tyrosine kinases other than Lck, such as Hck and Fgr, are important in the Fc gamma receptor induced respiratory burst of neutrophils as well as the Fc gamma receptor responses of monocytes and macrophages. The compositions and methods of the present invention may be useful in inhibiting the Fc gamma induced respiratory burst response in neutrophils, and may also be useful in inhibiting the Fc gamma dependent production of TNF alpha. The ability to inhibit Fc gamma receptor dependent neutrophil, monocyte and macrophage responses would result in additional anti-inflammatory activity for the compounds employed in invention methods. This activity would be especially of value, for example, in the treatment of inflammatory diseases, such as arthritis or inflammatory bowel disease. The compositions and methods of the present invention may also be useful in the treatment of autoimmune glomerulonephritis and other instances of glomerulonephritis induced by deposition of immune complexes in the kidney that trigger Fc gamma receptor responses and which can lead to kidney damage.
[0073] In addition, certain Src-family tyrosine kinases, such as Lyn and Src, may be important in the Fc epsilon receptor induced degranulation of mast cells and basophils that plays an important role in asthma, allergic rhinitis, and other allergic disease. Fc epsilon receptors are stimulated by IgE-antigen complexes. Compounds employed in the methods of the present invention may inhibit the Fc epsilon induced degranulation responses. The ability to inhibit Fc epsilon receptor dependent mast cell and basopbil responses may result in additional anti-inflammatory activity for the present compounds beyond their effect on T cells.
[0074] The present invention also provides articles of manufacture comprising packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material comprises a label which indicates that the pharmaceutical composition can be used for treatment of disorders and wherein the pharmaceutical composition comprises a compound according to the present invention. Thus, in one aspect, the invention provides a pharmaceutical composition including a therapeutic agent and a compound of the invention, wherein the compound is present in a concentration effective to reduce vascular leakage associated with indications or therapeutic agents which have vascular leak as a side effect. For example, administration of a compound of the invention can be in conjunction with E -2, immunotoxins, antibodies or chemotherapeutics. In these cases, IL-2, immunotoxin, antibody or chemotherapeutic concentration can be determined by one having ordinary skill in the art according to standard treatment regimen or, for example, as determined by an in vivo animal assay.
[0075] The present invention also provides pharmaceutical compositions comprising IL-2, immunotoxin, antibody or chemotherapeutic and at least one invention compound in an amount effective for inhibiting vascular permeability, and a pharmaceutically acceptable vehicle or diluent. The compositions of the present invention may contain other therapeutic agents, and may be formulated, for example, by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (for example, excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques known in the art of pharmaceutical formulation.
[0076] The compounds of the invention may be formulated into therapeutic compositions as natural or salt forms. Pharmaceutically acceptable non-toxic salts include the base addition salts (formed with free carboxyl or other anionic groups) which may be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino-ethanol, histidine, procaine, and the like. Such salts may also be formed as acid addition salts with any free cationic groups and will generally be formed with inorganic acids such as, for example, hydrochloric, sulfuric, or phosphoric acids, or organic acids such as acetic, citric, p-toluenesulfonic, methanesulfonic acid, oxalic, tartaric, mandelic, and the like. Salts of the invention include amine salts formed by the protonation of an amino group with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like. Salts of the invention also include amine salts formed by the protonation of an amino group with suitable organic acids, such as p-toluenesulfonic acid, acetic acid, and the like. Additional excipients which are contemplated for use in the practice of the present invention are those available to those of ordinary skill in the art, for example, those found in the United States Pharmacopeia Vol. XXII and National Formulary Vol. XVII, U.S. Pharmacopeia Convention, Inc., Rockville, MD (1989), the relevant contents of which is incorporated herein by reference, i addition, polymorphs of the invention compounds are included in the present invention.
[0077] Pharmaceutical compositions of the invention may be administered by any suitable means, for example, orally, such as in the form of tablets, capsules, granules or powders; sublingually; buccally; parenterally, such as by subcutaneous, intravenous, intramuscular, intrathecal, or intracisternal injection or infusion techniques (e.g., as sterile injectable aqueous or non-aqueous solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; or rectally such as in the form of suppositories; in dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents. The present compounds may, for example, be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved by the use of suitable pharmaceutical compositions comprising the present compounds, or, particularly in the case of extended release, by the use of devices such as subcutaneous implants or osmotic pumps. The present compounds may also be administered liposomally.
[0078] In addition to primates, such as humans, a variety of other mammals can be treated according to the method of the present invention. For instance, mammals including, but not limited to, cows, sheep, goats, horses, dogs, cats, guinea pigs, rats or other bovine, ovine, equine, canine, feline, rodent or murine species can be treated. However, the method can also be practiced in other species, such as avian species (e.g., chickens).
[0079] The pharmaceutical compositions for the administration of the compounds of this embodiment either alone or in combination with IL-2, immunotoxin, antibody or chemotherapeutic may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which constitutes one or more accessory ingredients. In general, the pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredient into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired formulation. In the pharmaceutical composition the active object compound is included in an amount sufficient to produce the desired effect upon the process or condition of diseases. The pharmaceutical compositions containing the active ingredient may be in a form suitable for oral use, for example, as tablets, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. [0080] Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. They may also be coated to form osmotic therapeutic tablets for control release.
[0081] Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.
[0082] Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example sodium carboxymethylcellulose, methylcellulose, hydroxy- propylmethylcellulose, sodium alginate, polyvinyl-pyrrolidorie, gum tragacanth and gum acacia; dispersing or wetting agents may be a naturally-occurring phosphatide, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides, for example polyethylene sorbitan monooleate. Also useful as a solubilizer is polyethylene glycol, for example. The aqueous suspensions may also contain one or more preservatives, for example ethyl, or n- propyl, ?-hydroxybenzoate, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents, such as sucrose or saccharin.
[0083] Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an anti-oxidant such as ascorbic acid.
[0084] Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.
[0085] Syrups and elixirs may be formulated with sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative and flavoring and coloring agents.
[0086] The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a parenterally-acceptable diluent or solvent or cosolvent or complexing agent or dispersing agent or excipient or combination thereof, for example 1,3-butanediol, polyethylene glycols, polypropylene glycols, ethanol or other alcohols, povidones, various brands of TWEEN surfactant, sodium dodecyl sulfate, sodium deoxycholate, dimethylacetamide, polysorbates, poloxamers, cyclodextrins, lipids, and excipients such as inorganic salts (e.g., sodium chloride), buffering agents (e.g., sodium citrate, sodium phosphate), and sugars (e.g., saccharose and dextrose). Among the acceptable vehicles and solvents that may be employed are water, dextrose solutions, Ringer's solutions and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables.
[0087] Depending on the condition being treated, these pharmaceutical compositions may be formulated and administered systemically or locally. Techniques for formulation and administration may be found in the latest edition of "Remington's Pharmaceutical Sciences" (Mack Publishing Co, Easton Pa.). Suitable routes may, for example, include oral or transmucosal administration; as well as parenteral delivery, including intramuscular, subcutaneous, intramedullary, intrathecal, intraventricular, intravenous, intraperitoneal, or intranasal administration. For injection, the pharmaceutical compositions of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiologically buffered saline. For tissue or cellular administration, penetrants appropriate to the particular barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds maybe prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow, for the preparation of highly concentrated solutions.
[0088] The compounds of the present invention may also be administered in the form of suppositories for rectal administration of the drug. These compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are cocoa butter and polyethylene glycols.
[0089] For topical use, creams, ointments, jellies, solutions or suspensions, etc., containing the compounds of the present invention are employed. (For purposes of this application, topical application shall include mouthwashes and gargles).
[0090] In one aspect, the invention compounds are administered in combination with an anti-inflammatory agent, antihistamines, chemotherapeutic agent, immunomodulator , therapeutic antibody or a protein kinase inhibitor, e.g., a tyrosine kinase inhibitor, to a subject in need of such treatment. While not wanting to be limiting, chemotherapeutic agents include antimetabolites, such as methotrexate, DNA cross-linking agents, such as cisplatin/carboplatin; alkylating agents, such as canbusil; topoisomerase I inhibitors such as dactinomicin; microtubule inhibitors such as taxol (paclitaxol), and the like. Other chemotherapeutic agents include, for example, a vinca alkaloid, mitomycin-type antibiotic, bleomycin-type antibiotic, antifolate, colchicine, demecoline, etoposide, taxane, anthracycline antibiotic, doxorubicin, daunorubicin, carminomycin, epirubicin, idarubicin, mithoxanthrone, 4-dimethoxy-daunomycin, 11-deoxydaunorabicin, 13- deoxydaunorubicin, adriamycin-14-benzoate, adriamycin-14-octanoate, adriamycin-14- naphthaleneacetate, amsacrine, carmustine, cyclophosphamide, cytarabine, etoposide, lovastatin, melphalan, topetecan, oxalaplatin, chlorambucil, methtrexate, lomustine, thioguanine, asparaginase, vinblastine, vindesine, tamoxifen, or mechlorethamine. While not wanting to be limiting, therapeutic antibodies include antibodies directed against the HER2 protein, such as trastuzumab; antibodies directed against growth factors or growth factor receptors, such as bevacizumab, which targets vascular endothelial growth factor, and OSI-774, which targets epidermal growth factor; antibodies targeting integrin receptors, such as Nitaxin (also known as MEDI-522), and the like. Classes of anticancer agents suitable for use in compositions and methods of the present invention include, but are not limited to: 1) alkaloids, including, microtubule inhibitors (e.g., Nincristine, Vinblastine, and Vindesine, etc.), microtubule stabilizers (e.g., Paclitaxel [Taxol], and Docetaxel, Taxotere, etc.), and chromatin function inhibitors, including, topoisomerase inhibitors, such as, epipodophyllotoxins (e.g., Etoposide [VP-16], and Teniposide [VM- 26], etc.), and agents that target topoisomerase I (e.g., Camptothecin and Isirinotecan [CPT-11], etc.); 2) covalent DΝA-binding agents [alkylating agents], including, nitrogen mustards (e.g., Mechlorethamine, Chlorambucil, Cyclophosphamide, Ifosphamide, and Busulfan [Myleran], etc.), nitrosoureas (e.g., Carmustine, Lomustine, and Semustine, etc.), and other alkylating agents (e.g., Dacarbazine, Hydroxymethylmelamine, Thiotepa, and Mitocycin, etc.); 3) noncovalent DΝA-binding agents [antitumor antibiotics], including, nucleic acid inhibitors (e.g., Dactinomycin [Actinomycin D], etc.), anthracyclines (e.g., Daunorubicin [Daunomycin, and Cerubidine], Doxorubicin [Adriamycin], and Idarubicin [Idamycin], etc.), anthracenediones (e.g., anthracycline analogues, such as, [Mitoxantrone], etc.), bleomycins (Blenoxane), etc., and plicamycin (Mithramycin), etc.; 4) antimetabolites, including, antifolates (e.g., Methotrexate, Folex, and Mexate, etc.), purine antimetabolites (e.g., 6-Mercaptopurine [6-MP, Purinethol], 6- Thioguanine [6-TG], Azathioprine, Acyclovir, Ganciclovir, Chlorodeoxyadenosine, 2- chlorodeoxyadenosine [CdA], and 2'-Deoxycoformycin [Pentostatin], etc.), pyrimidine antagonists (e.g., fluoropyrimidines [e.g., 5-fluorouracil (Adrucil), 5-fluorodeoxyuridine (FdUrd) (Floxύridine)] etc.), and cytosine arabinosides (e.g., Cytosar [ara-C] and Fludarabine, etc.); 5) enzymes, including, L-asparaginase, and hydroxyurea, etc.; 6) hormones, including, glucocorticoids, such as, antiestrogens (e.g., Tamoxifen, etc.), nonsteroidal antiandrogens (e.g., Flutamide, etc.), and aromatase inhibitors (e.g., anastrozole [Arimidex], etc.); 7) platinum compounds (e.g., Cisplatin and Carboplatin, etc.); 8) monoclonal antibodies conjugated with anticancer drugs, toxins, and/or radionuclides, etc.; 9) biological response modifiers (e.g., interferons [e.g., IFΝ-.alpha., etc.] and interleukins [e.g., IL-2, etc.], etc.); 10) adoptive immunotherapy; 11) hematopoietic growth factors; 12) agents that induce tumor cell differentiation (e.g., all- trans-retinoic acid, etc.); 13) gene therapy techniques; 14) antisense therapy techniques; 15) tumor vaccines; 16) therapies directed against tumor metastases (e.g., Batimistat, etc.); and 17) inhibitors of angiogenesis.
[0091] The pharmaceutical composition and method of the present invention may further comprise other therapeutically active compounds as noted herein which are usually applied in the treatment of the above mentioned pathological conditions. Examples of other therapeutic agents include the following: cyclosporins (e.g., cyclosporin A), CTLA4-Ig, antibodies such as ICAM-3, anti-IL-2 receptor (Anti-Tac), anti-CD45RB, anti-CD2, anti-CD3 (OKT-3), anti-CD4, anti-CD80, anti-CD86, agents blocking the interaction between CD40 and gp39, such as antibodies specific for CD40 and/or gp39 (i.e., CD154), fusion proteins constructed from CD40 and gp39 (CD40Ig and CD8gp39), inhibitors, such as nuclear translocation inhibitors, of NF-kappa B function, such as deoxyspergualin (DSG), cholesterol biosynthesis inhibitors such as HMG CoA reductase inhibitors (lovastatin and simvastatin), non-steroidal antiinflammatory drugs (NSAJODs) such as ibuprofen and cyclooxygenase inhibitors such as rofecoxib, steroids such as prednisone or dexamethasone, gold coriipounds, antiproliferative agents such as methotrexate, FK506 (tacrolimus, Prograf), mycophenolate mofetil, cytotoxic drugs such as azathioprine and cyclophosphari ide, TNF-a inhibitors such as tenidap, anti-TNF antibodies or soluble TNF receptor, and rapamycin (sirolimus or Rapamune) or derivatives thereof.
[0092] Other agents that may be administered in combination with invention compounds include protein therapeutic agents such as cytokines, immunomodulatory agents and antibodies. As used herein the term "cytokine" encompasses chemokines, interleukins, lymphokines, monokines, colony stimulating factors, and receptor associated prqteins, and functional fragments thereof. As used herein, the term "functional fragment" refers to a polypeptide or peptide which possesses biological function or activity that is identified through a defined functional assay. [0093] The cytokines include endothelial monocyte activating polypeptide II (EMAP- II), granulocyte-macrophage-CSF (GM-CSF), granulocyte-CSF (G-CSF), macrophage- CSF (M-CSF), IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-12, and IL-13, interferons, and the like and which is associated with a particular biologic, morphologic, or phenotypic alteration in a cell or cell mechanism.
[0094] When other therapeutic agents are employed in combination with the compounds of the present invention they may be used for example in amounts as noted in the Physician Desk Reference (PDR) or as otherwise determined by one having ordinary skill in the art.
[0095] In the treatment or prevention of conditions which involve compromised vasculostasis an appropriate dosage level can generally be between about 0.01 and about 500 mg per 1 kg of patient body weight per day which can be administered in single or multiple doses. For example, the dosage level can be between about 0.01 and about 250 mg/kg per day; more narrowly, between about 0.5 and about 100 mg/kg per day. A suitable dosage level can be between about 0.01 and about 250 mg/kg per day, between about 0.05 and about 100 mg/kg per day, or between about 0.1 and about 50 mg/kg per day, or about 1.0 mg/kg per day. For example, within this range the dosage can be between about 0.05 and about 0.5 mg/kg per day, or between about 0.5 and about 5 mg/kg per day, or between about 5 and about 50 mg/kg per day. For oral administration, the compositions can be provided in the form of tablets containing between about 1.0 and about 1,000 mg of the active ingredient, for example, about 1.0, about 5.0, about 10.0, about 15.0, about'20.0, about 25.0, about 50.0, about 75.0, about lOO.O, about 150.0, about 200.0, about 250.0, about 300.0, about 400.0, about 500.0, about 600.0, about 750.0, about 800.0, about 900.0, and about 1,000.0 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. The compounds can be administered on a regimen of 1 to 4 times per day, such as once or twice per day. There may be a period of no administration followed by another regimen of administration. Preferably, administration of the compound is closely associated with the schedule of IL- 2 administration. For example, administration can be prior to, simultaneously with or immediately following IL-2 administration.
[0096] It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
[0097] Compounds of the present invention can be used, alone or in combination with an effective amount of a therapeutic antibody (or therapeutic fragment thereof), a chemotherapeutic or an immunotoxic agent, for treatment of tumors. While doxorubicin, docetaxel, or taxol are described in the present application as illustrative examples of chemotherapeutic agents, it should be understood that the invention includes combination therapy including a compound of the invention, including but not limited to vasculostatic agents, such as tyrosine, serine or threonine kinase inhibitors, for example, Src-family inhibitors, and any chemotherapeutic agent or therapeutic antibody.
C. Examples
[0098] The following examples are provided to further illustrate the advantages and features of the present invention, but are not intended to limit the scope of the invention.
1. General Methodology
[0099] All experiments were performed under anhydrous conditions (i.e. dry solvents) in an atmosphere of argon, except where stated, using oven-dried apparatus and employing standard techniques in handling air-sensitive materials. Aqueous solutions of sodium bicarbonate (NaHCO3) and sodium chloride (brine) were saturated. [00100] Analytical thin layer chromatography (TLC) was carried out on Merck
Kieselgel 60 F254 plates with visualization by ultraviolet and/or anisaldehyde, potassium permanganate or phosphomolybdic acid dips.
[0100] Reverse-phase HPLC chromatography was carried out on Gilson 215 liquid handler equipped with Waters SymmetryShield™ RP18 7μm (40 x 100mm) Prep-Pak cartridge. Mobile phase consisted of standard acetonitrile (ACN) and DI Water, each with 0.1% TFA added. Purification was carried out at a flow rate of 40mL/ min, and a gradient such that the peak of interest was eluted between 12-15 min in a 30 min run.
[0101] NMR spectra: IH Nuclear magnetic resonance spectra were recorded at 500 MHz. Data are presented as follows: chemical shift, multiplicity (s = singlet, d = doublet, t = triplet, q = quartet, qn = quintet, dd = doublet of doublets, m = multiplet, bs = broad singlet), coupling constant (J/Hz) and integration. Coupling constants were taken directly from the spectra and are uncorrected.
[0102] Low resolution mass spectra: Electrospray (ES+) ionization was used. The protonated parent ion (M+H) or fragment of highest mass is quoted. Analytical gradient consisted of 10% ACN in water ramping up to 100% ACN over 5 minutes unless otherwise stated.
2. General Synthetic Procedures.
[0103] Whereever Suzuki coupling was used, the following general procedure was employed. To a mixture of boronic acid, aryl bromide, and palladium tetrakis(triphenylphosphine)palladium(0) (Pd(Ph3)4) in 4:1 DME/EtOH was added a 2 M aqueous solution of sodium carbonate. The mixture was flushed with argon for 5 min and a condenser was added under argon flow. The reaction was heated to reflux (ca. 100 °C) for 2-18 h. The crude mixture was filtered and the solid filter cake was rinsed thoroughly with MeOH and DCM. The filtrate was concentrated in vacuo and purified by column chromatography. [0104] Whereever Buchwald coupling was used, the following general procedure was employed. A mixture of amine, bromide, Cs2CO3, Xantphos, and Pd2(dba)3 in dioxane was purged with argon for 5 min after which a condenser was added under argon flow. The reaction was heated to reflux (ca 110 °C) for 4-18h. The crude mixture was filtered and the solid filter cake was rinsed thoroughly with MeOH and DCM. The filtrate was concentrated in vacuo and purified by column chromatography.
[0105] Whereever deprotection of aryl methoxy was necessary, the following general procedure was employed. To a solution of the aryl methoxy compound in DCM was added BBr3 (either 1 M in DCM or neat). The reaction was checked for completion and additional BBr3 was added if needed. The reaction was quenched with NaHCO3 and basified to ca. pH 10-11. The biphasic mixture was filtered and the collected solid was rinsed with water. Trace solvents were removed in vacuo.
[0106] In general, one of three methods A, B, or C, can be used for synthesizing some of the compounds of the present invention. Those having ordinary skill in the art can determine, depending on variety of factors, including the particular compound that is sought to be made, whether to selected the method A, B or C.
Method A:
Figure imgf000069_0001
H2 10% Pd/C
NH2
Figure imgf000069_0002
(II) [0107] The synthetic method A is shown by the reaction scheme (II). About 1 equivalent of compound 1 was mixed with 2 equivalent of cynamide in a vial. The mixture was heated to about 100°C until the mixture was completely melted. The mixture was cooled down to room temperature and concentrated HCI was added. The mixture was then again heated at about 100°C for about 40 minutes and cooled down in ice water. About 14 moles of NaOH were carefully added to the above reaction mixture followed by heating the mixture at about 100°C for about 2 hours, and by cooling down to room temperature.
[0108] Compound 2, shown by the reaction scheme (II), was isolated by filtration and washed several times with water, methanol and diethyl ether. About 1 equivalent of compound 2 was dissolved in N, N-dimethylacetamide, and ethanol solution containing about 4 equivalent of boronic acid and the aqueous solution containing about 0.15 equivalent of potassium carbonate were added. About 0.1 equivalent of triphenylphosphine and about 0.0246 equivalent of tris(dibenzyllideneatone) dipalladium (0) were added to the mixture. The mixture was reflux overnight. The crude product was poured into saturated NaHCO3 solution, and CH2CI2 was used to extract the product.
[0109] Compound 3, shown by the reaction scheme (II), was isolated by removing solvent in the organic phase. Compound 3 was dissolved in N, N-dimethylacetamide in a vial with a septum. Catalytic amount of 10% palladium on carbon was added to the mixture. A balloon filled with hydrogen was placed on the top of the vial. The mixture was stirred at room temperature for about 2 hours. Celite was used to remove the palladium and carbon. Solvent was removed under vacuum and compound 4, shown by the reaction scheme (II), was isolated. About 1 equivalent of compound 4 was dissolved in anhydrous toluene solution containing about 1.2 equivalent of bromide R3Br, about 0.025 equivalent of Pd(dba)3, about 0.07 equivalent of BINAP, and about 0.6 equivalent of KOt-Bu.. The mixture was kept at about 100°C for 24 hours under argon. Compound 5, shown by the reaction scheme (II), was then isolated by high pressure liquid chromatography (HPLC). Method B:
Figure imgf000071_0001
(πi)
[0110] The synthetic method B is shown by the reaction scheme (III). Compounds 1, 2, 3, and 4 were consecutively prepared and isolated as described in Method A. About 1 equivalent of compound 4 was dissolved in an aniline followed by adding about 2 equivalents of sulfamic acid. The mixture was heated at about 200°C overnight. Compound 5 was isolated by HPLC.
Method C:
Figure imgf000071_0002
(IN) [0111] The synthetic method C is shown by the reaction scheme (IN). Compounds 1, 2, 3, and 4 were consecutively prepared and isolated as described in Method A. About 1 equivalent of compound 4 was dissolved in substituted phenylarnine followed by adding about 2 equivalent of sulfamic acid. The mixture was heated at about 200°C overnight. Compound 5 was isolated by HPLC and was dissolved in dry CH2CI2. The mixture was cooled to about -78°C using a dry ice-acetone bath. About 2 equivalents of BBr3 (IM solution in CH2C12) was added dropwise to the mixture at about -78°C under nitrogen atmosphere. The mixture was then allowed to warm to about 0°C and stirred overnight at about 0°C, followed by adding saturated aqueous solution of ΝaHCO3 at about 0°C and by separating the mixture in a separator funnel. The water layer was extracted twice with CH2C1 , and the combined organic layer was washed with brine and dried over Na2SO4. Compound 6 was isolated by removing solvent under vacuum. Compound 6 was dissolved in acetone, and about 6 equivalent of K2CO3 and about 2 equivalent of R3C1 were added to the mixture. The mixture was heated to reflux and stirred overnight, cooled and water was added. Compound 7 was isolated by prep HPLC.
Example 1 Synthesis of 7-bromo-benzo[l,2,41triazin-3-ylamine-l-oxide
Figure imgf000072_0001
(V)
[0112] About 2.48 g (11.4 mmol) of 4-bromo-2-nitro-phenylamine was mixed with about 1.51 g (about 36 mmol) of cynamide in a 20 ml vial. The mixture was heated to about 100°C till the mixture was totally melted. The mixture was cooled down to room temperature and about 6.5 ml of concentrated HCI was added. The mixture was heated at about 100°C for about 40 minutes and cooled down in ice water. About 6.5 ml of 14M NaOH was carefully added to the above reaction mixture. The resulted mixture was heated at about 100°C for about 2 hours, then cooled down to room temperature, and filtrated. After filtration, the precipitate was washed several times with water, methanol and diethylether to remove the starting material. About 0.739 g of the product having " formula (N) was obtained. Yield: about 27%. ESI-MS: [M+H]+, 241, 243; 1H ΝMR (DMSO-d6): δ 7.48 (d, J= 9.0 Hz, 1 H), 7.89 (dd, J7 = 9.0 Hz, J2 = 2.1 Hz, 1 H), 8.26 (d, .7= 2.1 Hz, I H).
Example 2 Synthesis of 7-bromo-5-methyl-benzo[l,2,41triazin-3-ylamine-l-oxide
Figure imgf000073_0001
(VI)
[0113] About 1 g (4.33 mmol) of 4-bromo-2-methyl-6-nitro-phenylamine was mixed with about 0.5 g (12 mmol) of cynamide and about 5 g pyridine hydrochloride in a 20 ml vial. The mixture was heated to reflux overnight. The mixture was cooled down to room temperature and 10% ΝaOH was carefully added. The resulted mixture was heated at about 100°C for about 2 hours, then cooled down to room temperature, and filtrated. After filtration, the precipitate was washed several times with water, acetone and diethyl ether to remove the starting material. About 0.4 g of the product having the formula (NI) was obtained. Yield: about 36%. ESI-MS: [M+H]+, 255, 257; 1H ΝMR (DMSO-d6): δ 2.45 (s, 3 H), 7.81 (s, 1 H), 8.26 (s, 1 H).
Example 3 Synthesis of 7-bromo-5-methyl-benzo[l,2,4]triazin-3-ylamine
Figure imgf000073_0002
(VII) [0114] About 4.26g of 7-bromo-5-methyl-benzo[l,2,4]triazin-3-ylamine-l-oxide (NI) prepared as described in Example 2, was dissolved in about 100 ml of acetic acid, and about 1 g of iron powder was added to the solution. The mixture was refluxing for about 10 minutes. The iron was removed by filtration. The solvent in the filtrate was removed under vacuum. Water was added to remove the salt from the product. About 3.9 g of the pure product having formula (Nil) was obtained.
Example 4 Synthesis of l-(3-chloropropanyl)pyrrolidine
Figure imgf000074_0001
(VIII)
[0115] To the solution of about 186 g (1.18 mol) of l-bromo-3-chloropropane in about 200 ml of ether was added about 2 equivalent of pyrrolidine at about 0°C. After addition, the mixture was allowed to warm to room temperature and stirred overnight. The resulting white solid residue was removed and to the clear solution was added ice- cold 10% hydrochloric acid. The ether layer was discarded and the acid layer was basified with ice-cold 20% ΝaOH, extracted with ether and dried over a2SO4. Ether was removed and the residue was distilled under vacuum (95 °C /30 mmHg). About 91 g of product having formula (VIII) was obtained in a form of a pale yellow liquid. The yield was about 53 %.
Example 5 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin-3-yI1- [4-(2-pyrroIidin-l-yl-ethoxy)-phenyll-amine
Figure imgf000074_0002
[0116] About 1 g (3.78 mmol) of 7-bromo-5-methyl-benzo[l,2,4]triazin-3-ylamine and about 2.04 g (7.56 mmol) of l-[2-(4-bromo-ρhenoxy)-ethyl]-pyrrolidine were dissolved in about 500 ml of toluene. About 174 mg (0.19 mmol) of Pd(dba)3, about 340 mg (0.54 mmol) of BINAP, and about 500 mg (4.46 mmol) KO'-Bu were added to the solution. The mixture was kept at about 100°C for about 24 hours under argon. The crude product was purified by preparative HPLC. About 90 mg of [7-(2,6-dimethyl-phenyl)-5- me yl-benzo[l,2,4]1riazm-3-yl]-[4-(2-pyrrolidin-l-yl-ethoxy)-phenyl]-amine (product having formula (IX)) was isolated. Yield: about 5.2%. ESI-MS: [M+H]+, 454.6; 1H NMR (DMSO-d6): δ 1.90-2.05 (m, 10 H), 2.64(s, 3H), 3.15 (m, 2 H), 3.59-3.64 (m, 4 H), 4.31 (t, J= 5.2 Hz, 2 H), 7.09(d, J= 9.2 Hz, 2 H), 7.18 (d, J= 7.4 Hz, 2 H), 7.23 (m, 1 H), 7.60 (s, 1 H), 7.91 (s, 1 H), 7.98 (d, J= 9.2 Hz, 2 H).
Example 6 Synthesis of l-{4-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin- 3-ylamino1-phenyl}-cvcIopropanecarbonitrile
Figure imgf000075_0001
(X)
[0117] About 50 mg (0.2 mmol) of 7-bromo-5-methyl-benzo[l,2,4]triazin-3-ylamine and about 84 mg (0.38 mmol) of l-(4-bromo-phenyl)-cyclopropanecarbonitrile were dissolved in about 10 ml of toluene. About 8 mg (0.009 mmol) of Pd(dba)3, about 17 mg (0.027 mmol) of BINAP, and about 50 mg (0.226 mmol) of KO'-Bu were added to the solution. The mixture was kept at about 100°C for about 24 hours under argon. The crude product was purified by preparative HPLC. About 5 mg of l-{4-[7-(2,6-dimethyl- phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]-phenyl}-cyclopropanecarbonitrile (product having formula (X)) was isolated. ESI-MS: [M+H]+, 406.5; 1H NMR (DMSO- de): δ 1.49 (q, 2 H, J = 5.3 Hz), 1.72 (q, 2 H, J = 5.3 Hz), 2.05 (s, 6 H), 2.67(s, 3H), 7.18 (d, J= 7.4 Hz, 2 H), 7.23 (m, 1 H), 7.38 (d, J= 6.85 Hz, 2 H), 7.63 (s, 1 H), 7.91 (s, 1 H), 8.06 (d, J= 6.85 Hz, 2 H).
Example 7 Synthesis of r7-(2.6-dimethyl-phenyl)-benzo[l,2,41triazin-3-ylH4-f2- pyrrolidin-l-yl-ethoxy)-phenyl]-amine
Figure imgf000076_0001
(XI)
[0118] To a solution of about 100 mg (0.42 mmol) of 7-bromo-benzo[l,2,4]triazin-3- ylamine-1 -oxide dissolved in about 6 ml of N, N-dimethylacetamide in a 20 ml vial, were added about 240 mg (1.6 mmol) of 2,6-dimethylphenylboronic acid dissolved in about 1 ml of ethanol and about 64 mg (0.6 mmol) of potassium carbonate dissolved in about 1 ml of water. About 9 mg (0.034 mmol) of triphenylphosphine and about 9 mg (9.83 mmol) of tris(dibenzylideneacetone) dipalladium (0) were added to the mixture. The mixture was reflux overnight. The crude product was poured into about 50 ml of saturated aqueous solution of NaHCO3, and CH2C12 was used to extract the product. Solvent in the organic phase was removed under vacuum. The residue was dissolved in a mixture of about 2 ml of N, N-dimethylacetamide and about 1 ml of ethyl alcohol in a 20 ml vial with a septum. Catalytic amount of 10% palladium on carbon was added to the mixture. A balloon filled with hydrogen was placed on the top of the vial. The mixture was stirred at room temperature for about 2 hours. Celite was used to remove the palladium and carbon. The crude product and about 200 mg (0.74 mmol) of l-[2-(4- bromo-phenoxy)-ethyl]-pyrrolidine were dissolved in 10 ml of toluene. About 17 mg (0.018 mmol) of Pd(dba)3, about 34 mg (0.054 mmol) of BINAP, and about 50 mg (0.226 mmol) of KOt-Bu were added to the solution. The mixture was kept at about 100°C for about 24 hours under argon. The crude product was purified by preparative HPLC. About 2 mg of [7-(2,6-dimethyl-phenyl)-benzo[l,2,4]triazin-3-yl]-[4-(2- pyrrolidin-l-yl-ethoxy)-phenyl] -amine (product having formula (XI)) was isolated. Yield: about 1.6%; ESI-MS: [M+H]+, 440.6; 1H MR (DMSO-d6): δ 1.90-2.05 (m, 10 H), 3.15 (m, 2 H), 3.59-3.64 (m, 4 H), 4.31 (t, J= 5.15 Hz, 2 H), 7.09(d, J= 9.06 Hz, 2 H), 7.18 (d, J= 7.4 Hz, 2 H), 7.23 (m, 1 H), 7.70 (d, J= 8.7 Hz, 1 H), 7.78 (d, J= 8.7 Hz, 1 H), 7.90 (d, J= 9.06 Hz, 2 H), 8.08 (s, 1 H).
Example 8 Synthesis of f7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin-3-yll- quinolin-8-vI-amine by Method A
Figure imgf000077_0001
(XH)
[0119] About 100 mg (0.4 mmol) of 7-bromo-5-methyl-benzo[l,2,4]triazin-3-ylamine and about 158 mg (0.8 mmol) of 8-bromo-quinoline were dissolved in about 10 ml toluene. About 17 mg (0.018 mmol) of Pd(dba)3, about 17 mg (0.027 mmol) of BINAP, and about 50 mg (0.226 mmol) of KOt-Bu were added to the solution. The mixture was kept at about 100°C for about 24 hours under argon. The crude product was purified by preparative HPLC. About 50 mg [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin- 3-yl]-quinolin-8-yl-amine (product having formula (XII)) was isolated. Yield: about 33.8%; ESI-MS: [M+H]+, 392; 1H NMR (DMSO-d6): δ 2.07 (s, 6 H), 2.79(s, 3H), 7.19 (d, J= 7.4 Hz, 2 H), 7.25 (m, 1 H), 7.71-7.79 (m, 4 H), 8.04 (s, 1 H), 8.50 (d, J= 8.3 Hz, 1 H), 9.05 (m, 2 H).
Example 9 Synthesis of [7-(2,6-dimethyl-phenyπ-5-methyI-benzo[l,2,41triazin-3-yl]- pyridin-2-yI-amine by Method A
Figure imgf000078_0001
[0120] Title product having formula (XIII) was synthesized using Method A described above. ESI-MS: [M+H]+, 342;1H MR (500 MHz, DMSO-d6): δ 2.05 (s, 6H), 2.71 (s, 3H), 7.18-7.26 (m, 4 H), 7.71 (s, 1 H), 8.00 (m, 2 H), 8.44 (m, 2 H), 11.42 (s, IH, NH).
Example 10 Synthesis of [7-(2-6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin-3- yll-quinoIin-3-yl-amine by Method A
Figure imgf000078_0002
(XIV)
[0121] Title product having formula (XIII) was synthesized using Method A described above. ESI-MS: [M+H]+, 392; 1H NMR (500 MHz, DMSO-d6): δ 2.07 (s, 6H), 2.77 (s, 3H), 7.18 (d, J= 7.4 Hz, 2 H), 7.23 (m, 1 H), 7.62 (m, 2H), 7.70 (s, IH), 7.94 (dd, Jx = 8.14 Hz, J2 = 1.48 Hz, IH), 8.01 (m, 2H), 9.16 (s, IH), 9.30 (s, IH), 11.44 (s, IH, NH).
Example 11 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzofl,2,4]triazin-3- yn-m-tolyl-amine by Method B
Figure imgf000079_0001
(XV)
[0122] Title product having formula (XV) was synthesized using Method B described above. ESI-MS: [M+H]+, 355; 1H NMR (CDC13): δ 2.10 (s, 6 H), 2.44 (s, 3 H), 2.74 (s, 3 H), 6.97 (d, J = 7.6 Hz, 1 H), 7.17 (d, J = 7.6 Hz, 2 H), 7.22 (m, 1 H), 7.33 (m, 1 H), 7.49 (s, 1 H), 7.74 (d, J= 1.6 Hz, 1 H), 7.96 (s, 1 H), 7.99 (s, 1 H).
Example 12 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin-3- yl]-p-tolyl-amine by Method B
Figure imgf000079_0002
(XNI)
[0123] Title product having formula (XVI) was synthesized using Method B described above. ESI-MS: [M+H]+, 355; 1H NMR (CDC13): δ 2.09 (s, 6 H), 2.38 (s, 3 H), 2.72 (s, 3 H), 7.16 (d, J = 7.6 Hz, 2 H), 7.22 (m, 2 H), 7.47 (s, 1 H), 7.80 (d, J= 8.4 Hz, 2 H), 7.96 (s, 1 H). Example 13 Synthesis of (2,3-dimethyl-phenylVr7-(2,6-dimethyl-phenyl)-5-methyl- benzo[l, 2,41 triazin-3-yI] -amine by Method B
Figure imgf000080_0001
(xNπ)
[0124] Title product having formula (XNII) was synthesized using Method B described above. ESI-MS: [M+H]+, 369; 1H ΝMR (CDC13): δ 2.09 (s, 6 H), 2.35 (s, 3 H), 2.40 (s, 3 H), 2.72 (s, 3 H), 7.03 (d, J = 7.6 Hz, 1 H), 7.16 (d, J = 7.2 Hz, 2 H), 7.20-7.24 (m, 2 H), 7.45 (s, 1 H), 7.95 (s, 1 H), 8.20 (d, J= 7.6 Hz, 1 H).
Example 14 Synthesis of (2,4-dimethyl-phenyl)-[7-(2,6-dimethyl-phenyl)-5-methyI- benzo[l ,2,4] triazin-3-yI] -amine by Method B
Figure imgf000080_0002
(XVIII)
[0125] Title product having formula (XVIII) was synthesized using Method B described above. ESI-MS: [M+H]+, 369; 1H NMR (CDC13): δ 2.09 (s, 6 H), 2.36 (s, 3 H), 2.44 (s, 3 H), 2.66 (s, 3 H), 7.11-7.20 (m, 5 H), 7.45 (s, 1 H), 7.95 (s, 1 H), 8.20 (d, J= 8.4 Hz, I H). Example 15 Synthesis of (2,5-dimethyl-phenyl)-[7-(2,6-dimethyl-phenyl)-5-methyl- benzo[l,2,4]triazin-3-yI]-amine by Method B
Figure imgf000081_0001
(XIX)
[0126] Title product having formula (XIX) was synthesized using Method B described above. ESI-MS: [M+H]+, 369; 1H NMR (CDC13): δ 2.09 (s, 6 H), 2.42 (s, 3 H), 2.43 (s, 3 H), 2.69 (s, 3 H), 6.90 (d, J = 7.6 Hz, 1 H), 7.16 (d, J = 7.6 Hz, 2 H), 7.26 (m, 2 H), 7.47 (s, 1 H), 7.95 (s, 1 H), 8.40 (s, 1 H).
Example 16 Synthesis of (3,4-dimethyl-phenylH7-(2,6-dimethyl-phenyl)-5-methyl- benzo[l,2,4]triazin-3-yl]-amine by Method B
Figure imgf000081_0002
(XX)
[0127] Title product having formula (XX) was synthesized using Method B described above. ESI-MS: [M+H]+, 369; 1H NMR (CDCI3): δ 2.09 (s, 6 H), 2.29 (s, 3 H), 2.34 (s, 3 H), 2.72 (s, 3 H), 7.17-7.23 (m, 4 H), 7.47 (s, 1 H), 7.68 (d, J = 8.0 Hz, 1 H), 7.92 (s, 1 H), 7.95 (s, 1 H). Example 17 Synthesis of (3-chloro-4-fluoro-phenylV[7-(2,6-dimethyl-phenyl)-5- methyl-benzo[1.2.41triazin-3-yll-amine by Method B
Figure imgf000082_0001
(XXI)
[0128] Title product having formula (XXI) was synthesized using Method B described above. ESI-MS: [M+H]+, 393; 1H NMR (CDC13): δ 2.09 (s, 6 H), 2.74 (s, 3 H), 7.17 (d, J = 7.6 Hz, 2 H), 7.24 (m, 1 H), 7.54 (s, 1 H), 7.65 (m, 1 H), 8.06 (s, 1 H), 8.30 (s, 1 H), 8.32 (dd, J7 = 6.8 Hz, J2 = 3.2 Hz, 1 H).
Example 18 Synthesis of [7-(2-chloro-phenyl -5-methyl-benzo[l,2,41triazin-3-yl1- phenyl-amine by Method B
Figure imgf000082_0002
(XXII)
[0129] Title product having formula (XXII) was synthesized using Method B described above. ESI-MS: [M+H]+, 347; 1H NMR (CDC13): δ 2.75 (s, 3 H), 7.15 (t, J = 7.6 Hz, 1 H), 7.37-7.39 (m, 2 H), 7.44-7.48 (m, 3 H), 7.53 (d, J = 7.6 Hz, 1 H), 7.81 (s, 1 H), 7.92 (d, J = 7.6 Hz, 2 H), 8.24 (s, 1 H). Example 19 Synthesis of (4-methoxy-phenyl)-[7-(2-methoxy-phenylV5-methyl- benzofl,2,4]triazin-3-yI]-amine by Method B
Figure imgf000083_0001
(XXIII)
[0130] Title product having formula (XXIII) was synthesized using Method B described above. ESI-MS: [M+H]+, 373; 1H NMR (CDC13): δ 2.70 (s, 3 H), 3.83 (s, 3 H), 3.87 (s, 3 H), 6.99 (d, J= 8.8 Hz, 2 H), 7.05, (d, J= 8 Hz, 1 H), 7.09 (m, 1 H), 7.38 (m, 1 H), 7.45 (d, J= 8 Hz, 1 H), 7.81 (d, J = 8.8 Hz, 2 H), 7.88 (s, 1 H), 8.29 (s, 1 H).
Example 20 Synthesis of (4-butyI-phenyl)-[7-(2,6-dimethyI-phenyl)-5-methyl- benzo[l,2,41triazin-3-yl]-amine by Method B
Figure imgf000083_0002
(XXIV)
[0131] Title product having formula (XXIV) was synthesized using Method B described above. ESI-MS: [M+H]+, 397.3; 1H NMR (CDC13): δ 0.95 (t, J = 7.6 Hz, 3 H), 1.32 (m, 2 H), 1.64 (m, 2 H), 2.10 (s, 6 H), 2.64 (t, J = 7.6 Hz, 2 H), 2.66 (s, 3 H), 7.16 (d, J = 8.4 Hz, 2 H), 7.16 (d, J= 1.6 Hz, 2 H), 7.22 (m, 1 H), 7.47 (s, 1 H), 7.82 (d, j = 8.4 Hz, 2 H), 7.95 (s, 1 H). Example 21 Synthesis of [7-(2.6-dimethyl-phenylV5-methvI-benzo[l,2,4]triazin-3- yl]-(2-methoxy-phenyI)-amine by Method B
Figure imgf000084_0001
(XXV)
[0132] Title product having formula (XXV) was synthesized using Method B described above. ESI-MS: [M+H]+, 371.2; 1H NMR (CDC13): δ 2.10 (s, 6 H), 2.75 (s, 3 H), 4.01 (s, 3 H), 6.98 (dd, Jj = 1.6 Hz, J2 = 2 Hz, 1 H), 7.10 (m, 2 H), 7.16 (d, J= 7.6 Hz, 2 H), 7.22 (m, 1 H), 7.49 (s, 1 H), 7.97 (s, 1 H), 8.84 (dd, J7 = 7.6 Hz, J2 = 2.4 Hz, 1 H).
Example 22 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,41triazin-3- vH-(3-methoxy-phenvD-amine by Method B
Figure imgf000084_0002
(XXVI)
[0133] Title product having formula (XXVI) was synthesized using Method B described above. ESI-MS: [M+H]+, 371.2; 1H NMR (CDCI3): δ 2.10 (s, 6 H), 2.75 (s, 3 H), 3.92 (s, 3 H), 6.70 (d, J= 7.6 Hz, 1 H), 7.16 (d, J= 7.6 Hz, 2 H), 7.22 (m, 1 H), 7.26- 7.35 (m, 2 H), 7.49 (s, 1 H), 7.88 (s, 1 H), 7.98 (s, 1 H). Example 23 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo [1,2,4] triazin-3- vn-(4-methoxy-phenyl)-amine by Method B (product TGI 00-412-D
Figure imgf000085_0001
(XXVII)
[0134] Title product having formula (XXNII) was synthesized using Method B described above. ESI-MS: [M+H]+, 371.2; 1H MR (CDC13): δ 2.10 (s, 6 H), 2.70 (s, 3 H), 3.86 (s, 3 H), 7.0 (d, J= 9.2 Hz, 2 H), 7.16 (d, J= 7.6 Hz, 2 H), 7.22 (m, 1 H), 7.46 (s, 1 H), 7.80 (d, J= 9.2 Hz, 2 H), 7.94 (s, 1 H).
Example 24 Synthesis of f7-(2,6-dimethyl-phenyI)-5-methyI-benzo [1,2,4] triazin-3- yl1-(4-ethoxy-phenvD-amine by Method B
Figure imgf000085_0002
(XXVIII)
[0135] Title product having formula (XXVIII) was synthesized using Method B described above. ESI-MS: [M+H]+, 385.3; 1H ΝMR (CDC13): δ 1.44 (t, J= 7.2 Hz, 3 H), 2.10 (s, 6 H), 2.70 (s, 3 H), 4.08 (q, J= 7.2 Hz, 2 H), 6.98 (d, J= 9.2 Hz, 2 H), 7.16 (d, J = 7.2 Hz, 2 H), 7.22 (m, 1 H), 7.46 (s, 1 H), 7.80 (d, J= 9.2 Hz, 2 H), 7.94 (s, 1 H). Example 25 Synthesis of (3-chIoro-phenyl)-[7-(2,6-dimethyl-phenyl -5-methyl- benzo[l,2,4]triazin-3-yl]-amine by Method B
Figure imgf000086_0001
(XXIX)
[0136] Title product having formula (XXIX) was synthesized using Method B described above. ESI-MS: [M+H]+, 375.3; 1H MR (CDC13): δ 2.10 (s, 6 H), 2.76 (s, 3 H), 7.10 (d, J= 9.2 Hz, 1 H), 7.17 (d, J= 7.6 Hz, 2 H), 7.22 (m, 1 H), 7.37 (m, 1 H), 7.49 (s, 1 H), 7.64 (d, J= 9.2 Hz, 1 H), 8.00 (s, 1 H), 8.25 (s, 1 H).
Example 26 Synthesis of (3,4-dichloro-phenyl)-[7-(2,6-dimethyl-phenyl)-5-methyl- benzo[l,2,4]triazin-3-vU-amine by Method B
Figure imgf000086_0002
(XXX)
[0137] Title product having formula (XXX) was synthesized using Method B described above. ESI-MS: [M+H]+, 409.3; 1H NMR (CDC13): δ 2.09 (s, 6 H), 2.75 (s, 3 H), 7.17 (d, J= 7.6 Hz, 2 H), 7.24 (m, 1 H), 7.47 (d, J= 8.8 Hz, 1 H), 7.54(s, 1 H), 7.62 (d, J= 8.8 Hz, 1 H), 8.01 (s, 1 H), 8.38 (s, 1 H). Example 27 Synthesis of [7-(3,5-dimethoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3- yll-m-tolyl-amine by Method B
Figure imgf000087_0001
(XXXI)
[0138] Title product having formula (XXXI) was synthesized using Method B described above. ESI-MS: [M+H]+, 387.4; 1H NMR (CDC13): δ 2.43 (s, 3 H), 2.76 (s, 3 H), 3.89 (s, 6 H), 6.53 (s, 1 H), 6.85 (s, 2 H), 6.96 (d, J= 7.2 Hz, 1 H), 7.33(m, 1 H), 7.72 (m, 2 H), 7.94 (s, 1 H), 8.36 (s, 1 H).
Example 28 Synthesis of [7-(3,5-dimethoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3- vH-p-tolyl-amine by Method B
Figure imgf000087_0002
(XXXII)
[0139] Title product having formula (XXXII) was synthesized using Method B described above. ESI-MS: [M+H]+, 387.4; 1H NMR (CDCI3): δ 2.38 (s, 3 H), 2.74 (s, 3 H), 3.89 (s, 6 H), 6.53 (s, 1 H), 6.85 (s, 2 H), 7.23 (d, J= 8.0 Hz, 2 H), 7.78(d, J= 8.0 Hz, 2 H), 7.92 (s, 1 H), 8.35 (s, 1 H). Example 29 Synthesis of [7-(3.5-dimethoxy-phenyl)-5-methyl-benzo[l,2,41triazin-3- yl]-(2,4-dimethyl-phenyl)-amine by Method B
Figure imgf000088_0001
(XXXIII)
[0140] Title product having formula (XXXIII) was synthesized using Method B described above. ESI-MS: [M+H]+, 401.4; 1H NMR (CDC13): δ 2.36 (s, 3 H), 2.43 (s, 3 H), 2.69 (s, 3 H), 3.89 (s, 6 H), 6.53 (s, 1 H), 6.85 (s, 2 H), 7.11 (s, 1 H), 7.14 (d, J- 8.8 Hz, 1 H), 7.90 (s, 1 H), 8.30 (d, J= 8.8 Hz, 1 H), 8.35 (s, 1 H).
Example 30 Synthesis of [7-(3,5-dimethoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3- yl]-(2,5-dimethyl-phenyl)-amine by Method B
Figure imgf000088_0002
(XXXIV)
[0141] Title product having formula (XXXIV) was synthesized using Method B described above. ESI-MS: [M+H]+, 401.4; 1H NMR (CDC13): δ 2.43 (s, 6 H), 2.72 (s, 3 H), 3.89 (s, 6 H), 6.53 (s, 1 H), 6.86 (s, 2 H), 6.91 (d, J= 7.2 Hz, 1 H), 7.16 (d, J= 7.2 Hz, 1 H), 7.93 (s, 1 H), 8.36 (s, 1 H), 8.40 (s, 1 H). Example 31 Synthesis of [7-(3,5-dimethoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3- yn-(3,4-dimethyl-phenyl)-amine by Method B
Figure imgf000089_0001
(XXXV)
[0142] Title product having formula (XXXV) was synthesized using Method B described above. ESI-MS: [M+H]+, 401.4; 1H NMR (CDC13): δ 2.29 (s, 3 H), 2.34 (s, 3 H), 2.75 (s, 3 H), 3.89 (s, 6 H), 6.53 (s, 1 H), 6.86 (s, 2 H), 7.20 (d, J= 7.6 Hz, 1 H), 7.65 (d, J= 7.6 Hz, 1 H), 7.68 (s, 1 H), 7.92 (s, 1 H), 8.36 (s, 1 H).
Example 32 Synthesis of (4-butyl-phenyl)-[7-(3,5-dimethoxy-phenvI)-5-methyl- benzo[l,2,4]triazin-3-yIl-amine by Method B
Figure imgf000089_0002
(XXXVI)
[0143] Title product having formula (XXXVI) was synthesized using Method B described above. ESI-MS: [M+H]+, 429.4; 1H NMR (CDC13): δ 0.95 (t, J= 7.2 Hz, 3 H), 1.40 (m, 2 H), 1.64 (m, 2 H), 2.64 (t, J- 8 Hz, 2 H), 2.75 (s, 3 H), 3.89 (s, 6 H), 6.53 (s, 1 H), 6.86 (s, 2 H), 7.24 (d, J= 8.4 Hz, 2 H), 7.80 (d, J= 8.4 Hz, 2 H), 7.92 (s, 1 H), 8.35 (s, 1 H). Example 33 Synthesis of [7-(3,5-dimethoxy-phenvI)-5-methyl-benzo[l,2,41triazin-3- yl]-(4-ethoxy-phenyl)-amine by Method B
Figure imgf000090_0001
(XXXVII)
[0144] Title product having formula (XXXVII) was synthesized using Method B described above. ESI-MS: [M+H]+, 417.3; 1H NMR (CDC13): δ 1.44 (t, J= 6.8 Hz, 3 H), 2.73 (s, 3 H), 3.89 (s, 6 H), 4.07 (q, J= 6.8 Hz, 2 H), 6.53 (s, 1 H), 6.86 (s, 2 H), 6.98 (d, J= 8.8 Hz, 2 H), 7.80 (d, J= 8.8 Hz, 2 H), 7.91 (s, 1 H), 8.34 (s, 1 H).
Example 34 Synthesis of [7-(3,5-dimethoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3- vπ-pyridin-4-vI-amine by Method B
Figure imgf000090_0002
(XXXVIII)
[0145] Title product having formula (XXXVIII) was synthesized using Method B described above. ESI-MS: [M+H]+, 374.4; 1H NMR (CDCI3): δ 2.82 (s, 3 H), 3.89 (s, 6 H), 6.53 (s, 1 H), 6.87 (s, 2 H), 7.87 (d, J= 5.2 Hz, 2 H), 8.02 (s, 1 H), 8.43 (s, 1 H), 8.60 (d, J= 5.2 Hz, 2 H). Example 35 Synthesis of r7-(3,5-dimethoxy-phenylV5-methyl-benzo[l,2,41triazin-3- yl]-(2,3-dimethvI-phenylVamine by Method B
Figure imgf000091_0001
(XXXIX)
[0146] Title product having formula (XXXIX) was synthesized using Method B described above. ESI-MS: [M+H]+, 401.4; 1H NMR (CDC13): δ 2.36 (s, 3 H), 2.39 (s, 3 H), 2.68 (s, 3 H), 3.89 (s, 6 H), 6.53 (s, 1 H), 6.86 (s, 2 H), 7.04 (d, J= 7.6 Hz, 1 H), 7.22 (m, 1 H), 7.90 (s, 1 H), 8.18 (d, /= 7.6 Hz, 1 H), 8.35 (s, 1 H).
Example 36 Synthesis of [7-(3,5-dimethoxy-phenyl)-5-methyl-benzofl,2,41triazin-3- yl]-(2-methoxy-phenyl -amine by Method B
Figure imgf000091_0002
(XL)
[0147] Title product having formula (XL) was synthesized using Method B described above. ESI-MS: [M+H]+, 403.4; 1H NMR (CDCI3): δ 2.77 (s, 3 H), 3.89 (s, 6 H), 3.99 (s, 3 H), 6.53 (s, 1 H), 6.86 (s, 2 H), 7.00 (d, J= 7.6 Hz, 1 H), 7.08 (m, 2 H), 7.93 (s, 1 H), 8.36 (s, 1 H), 8.80 (d, J= 7.6 Hz, 1 H). Example 37 Synthesis of f7-(3,5-dimethoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3- yl]-(4-methoxy-phenyl)-amine by Method B
Figure imgf000092_0001
(XLI)
[0148] Title product having formula (XLI) was synthesized using Method B described above. ESI-MS: [M+H]+, 403.4; 1H NMR (CDC13): δ 2.73 (s, 3 H), 3.86 (s, 3 H), 3.89 (s, 6 H), 6.53 (s, 1 H), 6.86 (s, 2 H), 7.00 (d, J= 9.2 Hz, 2 H), 7.82 (d, J= 9.2 Hz, 2 H), 7.91 (s, 1 H), 8.35 (s, 1 H).
Example 38 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,41triazin-3- yll-pyridin-4-yl-amine by Method A
Figure imgf000092_0002
(XLII)
[0149] Title product having formula (XLII) was synthesized using Method A described above. ESI-MS: [M+H]+, 442.4; 1H NMR (DMSO-d6): δ 2.04 (s, 6 H), 2.75 (s, 3 H), 7.18 (d, 2 H), 7.25 (m, 1 H), 7.84 (s, 1 H), 8.15 (s, 1 H), 8.40 (d, 2 H), 8.75 (d, 2H). Example 39 Synthesis of [7-(2,6-dichloro-phenyI)-5-methyl-benzo[l,2,4]triazin-3- yl]-(3-methoxy-phenyf. -amine by Method B
Figure imgf000093_0001
(XLIII)
[0150] Title product having formula (XXLTII) was synthesized using Method B described above. ESI-MS: [M+H]+, 411.3; 1H NMR (DMSO-d6): δ 2.68 (s, 3 H), 3.83 (s, 3 H), 6.67 (d, J= 1.1 Hz, 1 H), 7.30 (t, J = 8.2 Hz, 1 H), 7.46 (d, J = 7.7 Hz, 1 H), 7.52 (m, 1 H), 7.66 (d, J= 8.2 Hz, 2 H), 7.73 (s, 1 H), 7.96 (s, 1 H), 8.12 (s, 1 H).
Example 40 Synthesis of [7-(2,6-dichloro-phenyl)-5-methyl-benzo[l,2,41triazin-3-yl]-
(4-methoxy-phenyI)-amine by Method B
Figure imgf000093_0002
(XD-V)
[0151] Title product having formula (XLIN) was synthesized using Method B described above. ESI-MS: [M+H]+, 411.3; 1H ΝMR (CDC13): δ 2.72 (s, 3 H), 3.86 (s, 3 H), 7.0 (d, J= 8.8 Hz, 2 H), 7.30 (m, 1 H), 7.46 (d, J = 8.4 Hz, 2 H), 7.54 (s, 1 H), 7.81 (d, J= 8.8 Hz, 2 H), 8.09 (s, 1 H). Example 41 Synthesis of [7-(2,6-dichloro-phenyl)-5-methyl-benzo[l,2,41triazin-3- vH-pyridin-4-yl-amine by Method A
Figure imgf000094_0001
(XLV)
[0152] Title product having formula (XLV) was synthesized using Method A described above. ESI-MS: [M+H]+, 382.4; 1HNMR (DMSO-d6): δ 2.77 (s, 3 H), 7.5 (t, 1 H), 7.70 (d, 2 H), 7.90 (s, 1 H), 8.30 (s, 1 H), 8.40 (d, 2 H), 8.70 (d, 2 H).
Example 42 Synthesis of [7-(2,6-dichloro-phenyl)-5-methyI-benzo[l,2,41triazin-3- yll-pyridin-3-yl-amine by Method A
Figure imgf000094_0002
(XLVI)
[0153] Title product having formula (XLVI) was synthesized using Method A described above. ESI-MS: [M+H]+, 382.4; 1H NMR (DMSO-d6): δ 2.68 (s, 3 H), 7.5 (t, 1 H), 7.70 (m, 3 H), 7.80 (s, 1 H), 8.20 (s, 1 H), 8.40 (m, 1 H), 8.60 (d, 1 H), 9.35 (s, 1 H).
Example 43 Synthesis of [4-(2-diethylamino-ethoxy)-phenyl1-[7-(2,6-dimethyl- phenyl)-5-methyl-benzofl,2,4]triazin-3-yl]-amine by Method C
Figure imgf000095_0001
(XLNπi)
[0154] Title product having formula (XLNIII) was synthesized using Method C described above. ESI-MS: [M+H]+, 456.3; 1H ΝMR (DMSO-d6): δ 1.25 (t, J= 7.2 Hz, 6 H), 2.05 (s, 6 H), 2.63 (s, 3H), 3.23 (m, 4 H), 3.59 (t, J= 5.0 Hz, 2 H), 4.44 (t, J= 5.0 Hz, 2 H), 7.10 (d, J= 9.2 Hz, 2 H), 7.16 (d, J= 7.4 Hz, 2 H), 7.21 (m, 1 H), 7.60 (s, 1 H), 7.91 (s, 1 H), 8.0 (d, J= 9.2 Hz, 2 H).
Example 44 Synthesis of f7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin-3- vIH4-(2-morpholin-4-yl-ethoxy)-phenyl]-amine by Method C
Figure imgf000095_0002
(XLIX)
[0155] Title product having formula (XLIX) was synthesized using Method C described above. ESI-MS: [M+H]+, 470.3; 1H ΝMR (DMSO-d6): δ 2.06 (s, 6 H), 2.64 (s, 3H), 3.20 (br, 2 H), 3.52-3.58 (b, 4 H), 3.75 (b, t, 2 H), 3.99 (b, 2 H), 4.38 (b, 2 H), 7.06 (d, J= 8.4 Hz, 2 H), 7.16 (d, J= 7.6 Hz, 2 H), 7.21 (m, 1 H), 7.58 (s, 1 H), 7.89 (s, 1 H), 7.96 (d, J= 8.4 Hz, 2 H). Example 45 Synthesis of r7-f2,6-dimethyl-phenyI)-5-methyl-benzo[l,2,4]triazin-3- vn-[4-(3-pyrrolidin-l-yl-propoxy)-phenyl]-amine by Method C
Figure imgf000096_0001
(L)
[0156] Title product having formula (L) was synthesized using Method C described above. ESI-MS: [M+H]+, 468.3; 1H NMR (CDC13): δ 2.09-2.21 (m, 10 H), 2.32 (m, 2 H), 2.70 (s, 3H), 2.84 (m, 2 H), 3.33 (b, 2 H), 3.91 (m, 2 H), 4.10 (t, = 5.6 Hz, 2 H), 6.95 (d, J= 9.2 Hz, 2 H), 7.16 (d, J= 7.6 Hz, 2 H), 7.21 (m, 1 H), 7.48 (s, 1 H), 7.82 (d, J = 9.2 Hz, 2 H), 7.94 (s, 1 H), 8.04 (s, 1 H, NH).
Example 46 Synthesis of [7-(2,6-dichloro-phenyl)-5-methyl-benzofl,2,41triazin-3- yl]-[4-(2-pyrroIidin-l-yl-ethoxy)-phenvI1-amine by Method C
Figure imgf000096_0002
(LI)
[0157] Title product having formula (LI) was synthesized using Method C described above. ESI-MS: [M+H]+, 494.3; 1H NMR (DMSO-d6): δ 1.90 (m, 2 H), 2.04 (m, 2 H), 2.63 (s, 3H), 3.15 (m, 2 H), 3.60 (m, 4 H), 3.98 (m, b, 2 H), 4.32 (t, J= 4.8 Hz, 2 H), 7.10 (d, J= 9.2 Hz, 2 H), 7.51 (t, j = 8.2 Hz, 1 H), 7.67 (d, J= 8.2 Hz, 2 H), 7.69 (s, 1 H), 7.98 (d, J= 9.2 Hz, 2 H), 8.08 (s, 1 H). Example 47. Synthesis of [7-(2.6-dimethyl-phenylV5-methyl-benzo[l,2,41triazin-3- yll-{4-[2-(4-methyl-piperazin-l-yl)-ethoxyl-phenvU-amine by Method C
Figure imgf000097_0001
(LII)
[0158] Title product having formula (LII) was synthesized using Method C described above. ESI-MS: [M+H]+, 483.3; 1H NMR (CDC13): δ 2.10 (s, 6 H), 2.69 (s, 3 H), 2.81 (s, 3 H), 3.10-3.50 (b, 10 H), 4.29 (b, 2 H), 6.98 (d, J= 8.8 Hz, 2 H), 7.16 (d, J= 7.6 Hz, 2 H), 7.21 (m, 1 H), 7.47 (s, 1 H), 7.84 (d, J= 8.8 Hz, 2 H), 7.94 (s, 1 H), 8.05 (s, 1 H, NH).
Example 48 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin-3- yl1-[3-(2-pyrrolidin-l-yl-ethoxy)-phenyl]-amine by Method C
Figure imgf000097_0002
(LIII)
[0159] Title product having formula (LIII) was synthesized using Method C described above. ESI-MS: [M+H]+, 454.3; 1H NMR (DMSO-d6): δ 1.85-2.06 (br, m, 10 H), 2.68 (s, 3H), 3.15 (br, 2 H), 3.60 (br, 4 H), 4.37 (br, 2 H), 6.73 (d, J= 8.2 Hz, 1 H), 7.18 (d, J = 7.4 Hz, 2 H), 7.22 (m, 1 H), 7.34 (m, 1 H), 7.55 (d, J= 8.2 Hz, 1 H), 7.65 (s, 1 H), 7.95 (s, 1 H), 7.98 (s, 1 H) Example 49 Synthesis of [3-(2-diethylamino-ethoxy -phenyll-[7-(2,6-dimethyl- phenvD-5-methyl-benzo[l,2,41 triazin-3-yll-amine by Method C
Figure imgf000098_0001
(LIN)
[0160] Title product having formula (LIV) was synthesized using Method C described above. ESI-MS: [M+H]+, 456.3; 1H ΝMR (DMSO-d6): δ 1.27 (t, J = 7.2 Hz, 6 H), 2.05 (s, 6 H), 2.68 (s, 3 H), 3.25 (br, 4 H), 3.58 (br, 2 H), 4.38 (br, 2 H), 6.72 (d, J = 8.8 Hz, 1 H), 7.19 (d, J= 7.3 Hz, 2 H), 7.24 (m, 1 H), 7.33 (m, 1 H), 7.51 (d, J= 8.8 Hz, 1 H), 7.66 (s, 1 H), 7.96 (s, 1 H), 8.02 (s, 1 H).
Example 50 Synthesis of 7-(2-chloro-5-methoxy-phenyl -5-methyl- benzo[l,2,41triazin-3-ylamine
Figure imgf000098_0002
(LV)
[0161] 7-bromo-5-methyl-benzo[l,2,4]triazin-3-ylamine (33.47 mmol, 1.0 equiv), 1- chloro-4-methoxy-2-boronic acid (50.21 mmol, 1.5 equiv), Pd(PPH3) (3.347 mmol, 0.1 equiv), and Νa2CO3 (133.9 mmol, 4.0 equiv) dissolved in DME/EtOH/water 6:1:1 and refluxed at 100 °C under an argon blanket for 4 h. The reaction was cooled to room temperature and diluted with 100 mL DCM and filtered. Precipitate recovered was suspended in water, filtered and rinsed with ether. Precipitate afforded product: 7-(2- chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine, a green solid (8.37 g, 84% yield). Rf = 0.85 (9:1 DCM/MeOH). 1H NMR (DMSO-d6): δ 3.35 (s, 6H), 7.01 (dd, J = 8.8 Hz, J = 3.0, IH), 7.09 (d, J = 3.0 Hz, IH), 7.48 (d, J = 8.8 Hz, IH), 7.75 (bm, 2H). MS (ES+) m/z = 303. LC retention time 3.03 min.
Example 51 Synthesis of 3-bromo-N-(2-dimethylamino-ethyl)-benzenesulfonamide
Figure imgf000099_0001
(LV-0
[0162] 3-bromobenzoyl chloride (4.56 mmol, 1.0 equiv) and N,N- dimethylethylenediamine (9.11 mmol, 2.0equiv) dissolved in 20 mL of anhydrous DCM and placed under an argon atmosphere. Et3N (22.8 mmol, 5.0 equiv) added to solution mixture via syringe and stirred at room temperature for 18 h. EtOAc added to mixture and the organics were washed with saturated NaHCO3 and brine, dried ( Na2SO4) and concentrated under reduced pressure to give 3-bromo-N-(2-dimethylamino-ethyl)- benzenesulfonamide, a pale yellow oil (1.20g, 86% yield). Material used as was in the next reaction. 1H NMR (DMSO-d6): δ 2.17 (s, 6H), 2.40 (t, J= 6.8 Hz, 2H), 3.35 (q, J= 6.7 Hz, 2H), 7.42 (t, J= 7.9 Hz, IH), 7.70 (d, J= 7.9 Hz, IH), 7.84 (d, J= 7.8 Hz, IH), 8.01 (s, IH), 8.53 (t, J= 2.3 Hz, IH). MS (ES+): m/z = 227 (m-Br) LC retention time 1.54 min.
Example 52 Synthesis of 3-[7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo [1 ,2,4] triazin-3-ylamino] -N-(2-dimethylamino-ethyl)-benzenesulfonamide
Figure imgf000099_0002
(LVII) [0163] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (0.333 mmol, 1.0 equiv), 3-bromo-N-(2-dimethylamino-ethyl)-benzenesulfonamide (0.499 mmol, 1.5 equiv), Cs2CO3 (0.999 mmol, 3.0 equiv), Pd2(dba)3 (0.0333 mmol, 0.1 equiv), and Xantphos (0.0666 mmol, 0.2 equiv) were dissolved in 7 mL dioxane. The reaction was placed under an argon atmosphere and refluxed at 100°C for 18 h. The organics were filtered to remove excess inorganics, dissolved in EtOAc, washed with saturated NaHCO3 and brine, dried over Na2SO4 and concentrated under reduced pressure to give 3 - [7-(2-chloro-5 -methoxy-phenyl)-5 -methyl-benzo [ 1 ,2,4]triazin-3 -ylamino] -N-(2- dimethylamino-ethyl)-benzenesulfonamide, which was precipitated using EtOAc/hexanes (1:5 v/v) to give a dark orange solid (168.4mg, 96% yield). Rf= 0.50 MS (ES+) m/z = 527.
Example 53 Synthesis of 3-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-ylaminol-N-(2-dimethylamino-ethyl)-benzenesuIfonamide
Figure imgf000100_0001
(LVIII)
[0164] 3-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]- N-(2-dimethylamino-ethyl)-benzenesulfonamide (0.32 mmol, 1.0 equiv) was dissolved in 10 mL of anhydrous DCM and placed under an argon atmosphere. 4 mL of BBr3 (0.1 M) added via syringe and the reaction stirred at room temperature for 2 h. BBr3 was quenched with saturated NaHCO3 until pH = 7. The mixture was filtered and washed with water and Et2O to give 3-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamino]-N-(2-dimethylamino -ethyl)-benzenesulfonamide, as an orange solid (132.2 mg, 81% yield). 1H NMR (DMSO-d6): δ 2.71 (s, 2H), 3.01 (bs, 2H), 6.87 (dd, J= 8.8 Hz, J= 2.9 Hz, IH), 6.94 (d, J= 3.0 Hz, IH), 7.41 (d, J= 8.8 Hz, IH), 7.51 (d, J= 7.9 Hz, IH), 7.64 (t, J= 8.1 Hz, 2H), 7.90 (s, IH), 8.06 (d, J= 6.7 Hz, IH), 8.20 (s, IH), 8.89 (s, IH), 9.93 (s, IH), 11.34 (s, IH). MS (ES+) m/z = 513. LC retention time 2.24 min.
Example 54 Synthesis of 4-[7-(2-chIoro-5-methoxy-phenyl)-5-methyl- benzofl,2,41triazin-3-ylamino]-N-methyl-N-(2-pyrrolidin-l-yl-ethyl)- benzenesulfonamide
Figure imgf000101_0001
(LIX)
[0165] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (0.33 mmol, 1.0 equiv), 4-bromo-N-methyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.5 mmol, 1.5 equiv), Cs2CO3 (0.99 mmol, 3.0 equiv), Pd2(dba)3 (0.033 mmol, 0.10 equiv), and Xantphos (0.066 mmol, 0.2 equiv) were dissolved in 10 mL dioxane and refluxed at 100°C under an argon atmosphere for 18 h. The reaction was cooled to room temperature and filtered. The filtrate was dissolved in EtOAc and washed with saturated NaHCO3 and brine, and the organic phase was dried (Na2SO4). The organics were concentrated under reduced pressure to afford residue that was purified by reverse phase HPLC using a 10-50-75 gradient over 30 minutes, and again concentrated under reduced pressure to afford 4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3- ylamino]-N-methyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide, as a tan residue (168 mg, 90% yield). Rf = 0.50 (9:1 DCM/MeOH) MS (ES+) m/z = 567. LC retention time 2.82 min.
Example 55 Synthesis of 4-[7-(2-chIoro-5-hydroxy-phenyl)-5-methyl- benzofl,2,4]triazin-3-ylamino]-N-methyl-N-(2-pyrrolidin-l-yl-ethyl)- benzenesulfonamide
Figure imgf000102_0001
(LX)
[0166] 4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]- N-methyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.297 mmol, 1.0 equiv) was dissolved in 10 mL anhydrous DCM and placed under an argon atmosphere. 4 mL of BBr3 (0.1 M) was added via syringe and the reaction was stirred at room temperature for 2 h. BBr3 was quenched with saturated NaHCO3 until pH = 7. The mixture was filtered and washed with water and Et2O to give 4-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl- benzo[ 1 ,2,4]triazin-3-ylamino]-N-methyl-N-(2-pyrrolidin- 1 -yl-ethyl)- benzenesulfonamide, as a dark green solid (120.5 mg, 74% yield). Rf = 0.61 1H NMR (DMSO-d6): δ 1.90(m, 2H), 2.01 (m, 2H), 2.72 (s, 6H), 3.06 (m, 2H), 3.30 (t, J= 5.6 Hz, 2H), 3.38 (t, J= 6.0 Hz, 2H), 3.59 (m, 2H), 6.89 (dd, J= 8.8 Hz, J= 2.9 Hz, IH), 6.96 (d, J= 2.9 Hz, IH), 7.40 (d, J= 8.6 Hz, IH), 7.87 (d, J= 8.9Hz, 2H), 7.92 (d, J= 0.9 Hz, IH), 8.21 (d, J= 1.7Hz, IH), 8.30 (d, J= 9.0 Hz, H), 11.50 (s, IH). MS (ES+) m/z = 553 LC retention time 2.41 min. Elemental for C27H30C12N6O3S 5H2O Calculated: C 47.72, H 5.93, N 11.37. Found: C 46.59, H 5.67, N 11.42.
Example 56 Synthesis of [7-(2-chloro-5-methoxy-phenyl)-6-methyl- benzo[l,2,41triazin-3-yl]-[4-(2-pyrroIidin-l-yl-ethoχy -phenyn-amine
Figure imgf000103_0001
(LXI)
[0167] 7-(2-chloro-5-methoxy-phenyl)-6-methyl-benzo[l,2,4]triazin-3-ylamine (0.67 mmol, 1.0 equiv), l-[2-(4-bromophenoxy)-ethyl] pyrrolidine (0.005 mmol, 1.5 equiv), Cs2CO3 (2.01 mmol, 3.0 equiv), Pd2(dba)3 (0.067 mmol, 0.1 equiv), and Xantphos (0.134 mmol, 0.2 equiv) were dissolved in 20 mL dioxane, placed under an argon atmosphere and refluxed at 100°C for 18 h. The reaction was cooled to room temperature and filtered. The filtrate was dissolved in EtOAc, washed with saturated NaHCO3 and brine and organic phase was dried (Na2SO4). The organics were concentrated under reduced pressure to afford residue that was precipitated out using EtOAc/hexanes (1:5 v/v) to give [7-(2-chloro-5-methoxy-phenyl)-6-methyl-benzo[l,2,4]triazin-3-yl]-[4-(2-pyrrolidin-l- yl-ethoxy)-phenyl]-amine, as an orange solid (271.2 mg, 83% yield). Rf = 0.56 (9:1 DCM/MeOH). MS (ES+) m/z = 491. LC retention time 2.72 min.
Example 57 Synthesis of 4-chloro-3-{6-methyl-3-[4-(2-pyrrolidin-l-yl-ethoxy)- phenylamino1-benzo[l,2,41triazin-7-yll-phenol
Figure imgf000103_0002
(LXII) [0168] [7-(2-chloro-5-methoxy-ρhenyl)-6-methyl-benzo[l,2,4]triazin-3-yl]-[4-(2- pyrrolidin-l-yl-ethoxy)-phenyl] -amine (0.553 mmol, 1.0 equiv) was dissolved in 10 mL anhydrous DCM and stirred under an argon atmosphere. 6 mL of BBr3 (0.1 M) was added via syringe and the reaction was stirred at room temperature for 2 h. BBr3 was quenched with saturated NaHCO3 until pH = 7. The mixture was filtered and washed with water and Et2θ to give 4-chloro-3-{6-methyl-3-[4-(2-pyrrolidin-l-yl-ethoxy)- phenylamino]-benzo[l,2,4]triazin-7-yl}-phenol, as a dark green solid (245.2 mg 93% yield). Rf = 0.65 (9:1 DCM/MeOH) 1H NMR (DMSO-d6): δ 1.69 (m, 4H), 2.24 (s, 3H), 2.54 (m, 4H), 2.79 (t, J= 5.9 Hz, 2H), 4.07 (t, J= 5.9 Hz, 2H), 6.78 (d, J= 2.9 Hz, IH), 6.87 (dd, J= 8.8 Hz, J= 2.9 Hz, IH), 6.95 (d, J= 11.9 Hz, 2H), 7.38 (dd, J- 8.8 Hz, J= 3.6 Hz, IH), 7.65 (s, IH), 7.85 (s, J= 9.1Hz, 2H), 7.98 (s, IH), 9.93 (s, IH), 10.67 (s, IH). MS (ES+): m/z = 476. LC retention time 2.32 min.
Example 58 Synthesis of 7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo [1 ,2,4] triazin-3-ylamine
Figure imgf000104_0001
(LV)
[0169] 7-bromo-5-methyl-benzo[l,2,4]triazin-3-ylamine (33.47 mmol, 1.0 equiv), 1- chloro-4-methoxy-2-boronic acid (50.21 mmol, 1.5 equiv), Pd(PPH3)4 (3.347 mmol, 0.1 equiv), andNa2CO3 (133.9 mmol, 4.0 equiv) were dissolved in DME/EtOH/water 6:1:1 and refluxed at 100°C under an argon blanket for 4 h. The reaction was cooled to room temperature, diluted with 100 mL DCM and filtered. The precipitate that was recovered was suspended in water, filtered and rinsed with ether. The precipitate afforded 7-(2- chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine, as a green solid (8.37 g, 84% yield). Rf = 0.85 (9:1 DCM/MeOH). 1H MR (DMSO-d6): δ 3.35 (s, 6H), 7.01 (dd, J = 8.8 Hz, J = 3.0, IH), 7.09 (d, J = 3.0 Hz, IH), 7.48 (d, J - 8.8 Hz, IH), 7.75 (bm, 2H). MS (ES+) m/z = 303. LC retention time 3.03 min.
Example 59 Synthesis of l-(3-bromo-benzenesulfonyl)-4-methyl-piperazine
(LXΓV)
[0170] 3-bromo-benzenesulfonyl chloride (3.914 mmol, 1.0 equiv) and 1-methyl- piperizine (7.83 mmol, 2.0 equiv) were dissolved in 15 mL DCM. Et3N (15.66 mmol, 4.0 equiv) was added via syringe to the reaction and stirred at room temperature for 2 h. The reaction was diluted with DCM, the organic phase was extracted with saturated NaHCO3 and brine and dried (Na2SO4). The organics were removed under reduced pressure to give l-(3-bromo-benzenesulfonyl)-4-methyl-piperazine, as a white solid (1.1 g. 88% yield). 1H NMR (DMSO-de): δ 2.14 (s, 3H), 2.35 (m, 4H), 2.92 (m, 4H), 7.62 (t, J= 7.9 Hz, 2H0, 7.55 (dd, J= 7.9 Hz, J= 0.8Hz, IH), 7.85 (t, J= 1.8 Hz, IH), 7.95 (dd, J= 9.1 Hz, J = 0.9 Hz, IH) MS (ES +) m/z = 321 LC retention time 1.74 min.
Example 60 Synthesis of [7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzofl,2,41triazin-3-yl1-[3-(4-methvI-piperazine-l-sulfonyI)-phenyl]-amine
Figure imgf000105_0002
(LXV)
[0171] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1 ,2,4]triazin-3-ylamine (0.315 mmol, 1.0 equiv), l-(3-bromo-benzenesulfonyl)-4-methyl-piperazine (0.472 mmol, 1.5 equiv), Pd2(dba)3 (0.0315 mmol, 0.1 equiv), Cs2CO3 (0.945 mmol, 3.0 equiv), and Xantphos (0.063 mmol, 0.2 equiv) were dissolved in 10 mL of dioxane , purged of air and placed under an argon atmosphere to reflux at 100°C for 18 h. The reaction was cooled to room temperature and filtered to remove excess Cs2CO3. The filtrate was extracted with EtOAc and washed with saturated NaHCO3 and brine, and the organics were dried (Na2SO4). The organics were removed under reduced pressure to afford brown residue which was precipitated using DCM/Et2O (1:5 v/v) to give [7-(2-chloro-5- methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-yl]-[3-(4-methyl-piperazine-l- sulfonyl)-phenyl]-amine, as a yellow solid (45 mg, 27% yield). 1HNMR (DMSO-d6): δ 2.12 ( s, 3H), 2.37 (m, 4H), 2.71 (s, 3H), 2.95 (m, 4H), 3.83 (s, 3H), 7.06 (dd, = 8.9 Hz, J= 3.1 Hz,lH), 7.14 (d, J= 3.1 Hz, IH), 7.41 (dd, J= 6.3 Hz, J= 1.1 Hz, IH), 7.54 (d, J = 8.7 Hz, IH), 7.67 (t, J= 8.1 Hz, 2H), 8.06 (dd, J= 7.6 Hz, J= 1.6 Hz, IH), 8.27 (d, J= 1.9 Hz, IH), 8.9 (bs, IH). MS (ES+): m/z = 540 LC retention time 2.70 min.
Example 61 Synthesis of 4-chloro-3-{5-methyl-3-[3-(4-methyl-piperazine-l- sulf onvD-phenylaminol -b enzo [1 ,2,4] triazin-7-y -phenol
Figure imgf000106_0001
(LXNI)
[0172] [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[r,2,4]triazin-3-yl]-[3-(4- methyl-piperazine-l-sulfonyl)-phenyl] -amine (0.0742 mmol, 1.0 equiv) was dissolved in 5 mL anhydrous DCM and placed under an argon atmosphere. 200 mL of BBr3 (0.1 M) was added via syringe to the amine solution and stirred at room temperature for 2 h. BBr3 was quenched with saturated ΝaHCO3 until pH was adjusted to 7. The mixture was filtered and rinsed with water and Et2O to give 4-chloro-3-{5-methyl-3-[3-(4-methyl- piperazine-l-sulfonyl)-phenylamino]-benzo[l,2,4]triazin-7-yl}-phenol, as a tan solid (24.3 mg, 62%yield). 1H NMR (DMSO-d6): δ 2.71 (s, 6H), 2.72 (bs, 2H), 3.16 (m, 2H), 3.46 (m, 2H), 3.78 (m, 2H), 6.88 (dd, J= 8.8Hz, J= 2.9 Hz, IH), 6.95 (d, J= 2.9 Hz, IH), 7.41 (d, J= S.l Hz, IH), 7.45 (dd, J= 7.9 Hz, J- 1.7 Hz, IH), 7.72 (t, J= 8.1 Hz, 2H), 7.91 (d, J= 1.7 Hz, IH), 8.12 (dd, J= 8.3 Hz, J= 1.7 Hz, IH), 8.21 (d, J= 1.9 Hz, IH), 8.93 (d, J= 1.9 Hz, IH), 9.99 (s, IH), 11,46 (s, IH). MS(ES+): m/z = 526. Elemental for C25H36Cl2N6O8S 5H2O Calculated: C 46.08, H 5.57, N 12.90. Found: C 46.84, H 5.04, N 12.66.
Example 62 Synthesis of 7-(2-chloro-5-nitro-phenyl)-5-methyl-benzo[l,2,4]triazin-3- ylamine
Figure imgf000107_0001
(LXVII)
[0173] 7-bromo-5-methyl-benzo[l,2,4]triazin-3-ylamine (3.35 mmol, 1.0 equiv), 2- chloro-5-nitrophenyl-boronic acid (5.02 mmol, 1.5 equiv), Pd(PPh3)4 (0.335 mmol, 0.1 equiv), and Na CO3 (13.4 mmol, 4.0 equiv) were dissolved in a mixture of DME/EtOH/H2O (4:1:1 v/v/v) and refluxed at 100°C for 18 h. The reaction was diluted with 50 mL DCM and filtered. The precipitate was suspended in water to remove excess Na2CO3 and filtered again with a final Et O wash to remove water. The crude precipitate gave 7-(2-chloro-5-nitro-phenyl)-5-methyl-benzo[l,2,43triazin-3-ylamine, as a dark green solid ( l.Og, 94% yield). Rf = 0.55 MS (ES+) m/z = 316 LC retention time 2.96 min.
Example 63 Synthesis of 4-[7-(2-chloro-5-nitro-phenyl)-5-methyl- benzori.2,4]triazin-3-ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesuIfonamide
Figure imgf000108_0001
(LXVIII)
[0174] 7-(2-chloro-5-nitro-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (0.65 mmol, 1.0 equiv), 4-bromo-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.95 mmol, 1.5 equiv), Xantphos (0.065 mmol, 0.10 equiv), Pd(OAc)2 (0.033 mmol, 0.05 equiv), and potassium-t-butoxide (1.3 mmol, 2.0 equiv) were dissolved in 45 mL of dioxane and placed under an argon atmosphere. The reaction mixture was refluxed at 100°C for 18 h and then cooled to room temperature. Inorganic salts were filtered out of solution and the organics were concentrated under reduced pressure. The residue was precipitated using EtOAC/hexanes (1:5 v/v) to afford 4-[7-(2-chloro-5-nitro-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide, as a red solid (232 mg, 63% yield). Rf = 0.42 MS (ES+) m/z = 570. LC retention time 2.56 min
Example 64 Synthesis of 4-[7-(5-amino-2-chloro-phenyl)-5-methyl- b enzo [1 ,2,41 triazin-3-ylamino] -N-(2-pyr rolidin-1 -yl-ethvQ-b enzenesulfonamide
Figure imgf000108_0002
(LXIX)
[0175] 4-[7-(2-chloro-5-nitiO-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]-N-(2- pyrrolidin-l-yl-ethyl)-b enzenesulfonamide (0.41 mmol, 1.0 equiv) was dissolved in 20 mL of MeOH, and the reaction was evacuated of air by vacuum and placed under a blanket of argon. Pd/C (10% by wt) was added to the reaction, followed by another evacuation of argon, and by blanketing with hydrogen. The reaction was stirred under hydrogen blanket for 4 h, filtered through Celite, and concentrated by reduced pressure to give a crude residue, which was precipitated using EtOAc/hexanes (1:5 v/v) to give 4-[7- (5 -amino-2-chloro-phenyl)-5 -methyl-benzo [ 1 ,2,4] triazin-3 -ylamino] -N-(2-pyrrolidin- 1 - yl-ethyl)-benzenesulfonamide, as a dark green solid (89.4 mg, 41% yield). 1H NMR (DMSO-d6): δ 1.61 (m,4H), 2.34 (m, 4H), 2.42 (t, J= 6.9 Hz, 2H), 2.71 (s, 3H), 2.85 (t, J = 7.1 Hz, 2H), 5.43 (s, IH), 6.65 (dd, J= 8.7 Hz, J= 2.8 Hz, IH), 6.73 (d, J= 2.7 Hz,lH), 7.21 (d, J= 8.7 Hz, IH), 7.83 (d, J= 8.9 Hz, 2H), 7.89 (s, IH), 8.15 (s, IH), 8.21 (d, J= 8.8 Hz, 2H). MS (ES+) m/z = 538, LC retention time 2.13 min.
Example 65 Synthesis of {4-[7-(2,6-dichloro-phenyl)-5-methyl-benzofl,2,41triazin-3- ylamino]-phenyl}-piperazin-l-yl-methanone
Figure imgf000109_0001
(LXX)
[0176] 4-(4-bromo-benzoyl)-piperazine-l -carboxylic acid tert-butyl ester (1.5 equiv, 0.49 mmol), 7-(2,6-dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (1.0 equiv, 0.33 mmol), Xantphos (0.1 equiv, 0.123 mmol), palladium acetate (0.05 equiv, 0.0165 mmol), and Potassium-t-butoxide (2.0 equiv, 0.66 mmol) were dissolved in 10 mL of dioxane, purged of air and placed under an argon blanket before refluxing at 100°C for 18 h. The reaction was cooled to room temperature and filtered before extracting with EtOAc and washing with saturated bicarbonate and brine, followed by drying over Na SO4. The organics were condensed under reduced pressure to give brown oil that was precipitated out using EtOAc/hexanes (1:5 v/v) to afford brown/tan precipitate. BOC protecting group was removed with 30% TFA DCM (v/v) stirring at room temperature for 1 h.
[0177] The product was concentrated, then suspended in DCM, extracted with saturated bicarbonate and brine, dried over Na2SO4, and condensed again to afford {4-[7- (2,6-dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]-phenyl}-piperazin-l-yl- methanone, as an orange solid (26.7 mg, 34% yield). 1H NMR (DMSO-d6): δ 2.69 (s, 3H), 3.19 (bt, J= 4.85 Hz, 4H), 3.74 (bs, 4H), 7.52 (d, J= 8.05 Hz, 2H), 7.56 (d, J= 8.75 Hz, IH), 7.67 (d, J= 8.2 Hz, 2H), 7.76 (s, IH), 8.13 (d, J= 14.4 Hz, 2H), 8.15 (s, IH), 11.25 (s, IH). MS (ES+) m/z = 492, LC retention time 2.45 minutes.
Example 66 Synthesis of 4-[7-(2,6-dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3- ylamino1-N-methyl-N-r2-pyrrolidin-l-yI-ethyl)-benzenesttlfonamide
Figure imgf000110_0001
(LXXI)
[0178] 4-bromo-N-methyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (1.0 equiv, 0.82 mmol), 7-(2,6-dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (1.5 equiv, 1.23 mmol), Cs2CO3 (3.0 equiv, 2.46 mmol), Pd2(dba)3 (0.1 equiv, 0.082 mmol), and Xantphos (0.2 equiv, 0.1634 mmol) were dissolved in 15 mL dioxane and purged of air, then placed under an argon blanket and refluxed for 18 h at 100°C. The reaction was cooled to room temperature and filtered and condensed under reduced pressure. The residue that was recovered was dissolved in EtOAc and extracted with saturated bicarbonate and brine; the organics were then dried (Na2SO4) and concentrated to afford brown oil. Using EtOAc/hexanes (1 :5 v/v) the residue was precipitated out to afford 4-[7- (2,6-dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]-N-methyl-N-(2- pyrrolidin-l-yl-ethyl)-benzenesulfonamide, as a tan solid, (297.2 mg, 76% yield). 1H NMR (DMSO-de): δ 1.95 (dm, 4H), 2.72 (s, 3H), 2.73 (s, 3H), 3.04 (q, J= 7.5 Hz, 2H), 3.35 (dt, J= 5.3 Hz, J= 5.4 Hz, 4H), 3.58 (q, J= 5.2 Hz, 2H), 7.52 (t, J= 7.6 Hz, IH), 7.67 (d, J= 3.2 Hz, 2H), 7.79 (s, IH), 7.87 (d, J= 3.2 Hz, 2H),8.18 (s, IH), 8.31 (s, J= 9.0 Hz, 2H), 10.51 (bs, IH), 11.55 (s, IH). MS (ES+) m/z = 51 A, LC retention time 2.80 minutes.
Example 67 Synthesis of [7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-yl1-[4-(4-methyl-piperazine-l-sulfonyl)-phenyl]-amme
Figure imgf000111_0001
(LXXII)
[0179] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (1.0 equiv., 0.75 mmol), l-(4-bromo-benzenesulfonyl)-4-methyl-piperazine (1.5 equiv., 1.125 mmol), Xantphos (0.1 equiv., 0.074 mmol), palladium acetate (0.05 equiv., 0.0375 mmol), and potassium-tert-butoxide (2.0 equiv, 1.5 mmol) were dissolved in 20 mL of dioxane and bubbled with argon. The reaction was stirred in an oil bath at 100°C for 18 h under an argon blanket. The reaction was cooled to room temperature and filtered to remove inorganics, concentrated under reduced pressure, then brought up in EtOAc and extracted using saturated NaHCO3 and brine. The organics were dried over Na2SO4 and concentrated under reduced pressure then precipitated using EtOAc/ Hexanes (1:5 v/v) to afford brown solid (182.3 mg, 45% yield). MS (ES+) : m/z = 540 LC retention time: 2.67 min.
Example 68 Synthesis of 4-chloro-3-{5-methyl-3-[4-(4-methyl-piperazine-l- sulfonyl)-phenylamino]-benzo [1 ,2,4] triazin-7-yl}-phenol
Figure imgf000112_0001
(LXXΠI)
[0180] [7-(2-chloro-5-methoxy-ρhenyl)-5-methyl-benzo[l,2,4]triazin-3-yl]-[4-(4- methyl-piperazine-l-sulfonyl)-phenyl] -amine (1.0 equiv., 0.339 mmol) was dissolved in 10 mL anhydrous DCM, purged of air via house vacuum, then blanketed with argon. Neat BBr3 (4.0 equiv., 1.355 mmol) was added via syringe dropwise and allowed to stir at room temperature for 2 h. The reaction was quenched with saturated NaHCO ; the precipitate was filtered and rinsed with Et2θ to afford crude orange solid. The compound was purified by prep. HPLC using 10-50-75 gradient, collecting peaks for 9-14 minutes. A yellow solid was recovered (70.5 mg, 40% yield). 1H NMR (DMSO-d6): δ 1.98 (s, 3H), 2.37 (bs, 4H), 2.72 (s, 3H), 2.90 (bs, 4H), 6.88 (dd, J= 2.94 Hz, 8.81 Hz, IH), 6.95 (d, J= 2.92 Hz, IH), 7.41 (d, J= 8.79 Hz, IH), 7.78 (d, J= 8.89 Hz, 2H), 7.92 (m, IH), 8.21 (d, J= 1.35 Hz, IH), 8.28 (d, J= 8.88 Hz, 2H), 11.47 (s, IH). MS (ES+): m/z = 526 LC retention time: 2.30 minutes.
Example 69 Synthesis of l~[2-(3-bromo-phenylsuIfanyl)-ethyll-pyrrolidine
Figure imgf000112_0002
(LXXIV)
[0181] l-(2-chloroethyl)pyrrolidine hydrochloride (1.0 equiv., 8.82 mmol) and 3- bromothiophenol (1.5 equiv., 13.23 mmol) were dissolved in acetonitrile (100 mL). Potassium carbonate (10.0 equiv., 88.2 mmol) was added to the reaction while stirring. The reaction was kept in oil bath at 80°C to reflux for 18 h. The reaction was then cooled to room temperature and filtered to remove excess inorganics, and the organics were extracted with saturated NaHCO3 and brine. The organics were dried over Na2SO4 then concentrated under reduced pressure. The resulting crude oil was purified by flash chromatography using 4:6 EtOAc/hexanes. The fractions were concentrated to afford product as a yellow oil (792.3 mg, 32% yield). Rf = 0.75 MS (ES+): m/z = 287 LC retention time: 1.81 min.
Example 70 Synthesis of [7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-vn-[3-(2-pyrrolidin-l-yl-ethvIsulfanyl)-phenyl]-amine
Figure imgf000113_0001
(LXXV)
[0182] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (1.0 equiv., 083 mmol), l-[2-(3-bromo-phenylsulfanyl)-ethyl] -pyrrolidine ( 1.5 equiv., 1.25 mmol), Xantphos (0.1 equiv., 0.084 mmol), palladium acetate (0.05 equiv., 0.042 mmol), and potassium-tert-butoxide (2.0 equiv., 1.66 mmol) were dissolved in 10 mL of dioxane, and argon was bubbled through the solution. The reaction was run under an argon blanket while refluxing at 100°C for 18 h. The reaction was then cooled to room temperature, filtered to remove inorganics, extracted using EtOAc with saturated NaHCO3 and brine, dried over Na2SO4, and concentrated under reduced pressure. A crude material was precipitated using 1 : 5 (v/v) EtOAc/ hexanes to afford a bright orange solid (311.8 mg, 74% yield). Rf = 0.48. Example 71 Synthesis of 4-chIoro-3-{5-methyl-3-[3-(2-pyrrolidin-l-yl- ethylsulfanylVphenylamino1-benzofl,2,41triazin-7-yl}-phenol
Figure imgf000114_0001
(LXXVI)
[0183] [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-yl]-[3-(2- pyrrolidin-l-yl-ethylsulfanyl)-phenyl] -amine (1.0 equiv., 0.62 mmol) was dissolved in anhydrous DCM, purged of air using house vacuum, and placed under an argon blanket. Neat BBr (4.0 equiv., 2.5 mmol) was then added via syringe and allowed to stir at room temperature for 2 h. The reaction was quenched using saturated NaHCO3; the precipitate was filtered and washed with water and Et2O to afford a dark orange/brown solid. The compound was purified by flash chromatography using 90: 10 DCM/MeOH to recover a burnt orange solid (64.6 mg, 22% yield). Rf = 0.32 1H NMR (DMSO-de): δ 1.68 (bs, 4H), 2.68 (s, 3H), 2.73 (bs, 2H), 3.14 (t, J= 6.87 Hz, 2H), 6.87 (d, J= 8.10 Hz, IH), 6.93 (s, IH), 7.03 (d, J= 7.42 Hz, IH), 7.34 (t, J= 7.58 Hz, IH), 7.40 (d, J= 8.65 Hz, IH), 7.77 (d, J= 7.95 Hz, IH), 8.16 (s, 2H), 9.93 (s, IH), 11.01 (s, IH). MS (ES+): m/z = 493
Example 72 Synthesis of 4-f7-(2-chlo-■o-5-methoxy- henv^)-6-metl^yl- benzo[l,2,41triazm-3-yl mino]-N-methyl-N-(2-pyrroIidin-l-yl-ethylV benzenesulfonamide
Figure imgf000114_0002
(LXXVII) [0184] 7-(2-chloro-5-methoxy-phenyl)-6-methyl-benzo[l,2,4]triazin-3-ylamine (1.0 equiv., 0.28 mmol), 4-bromo-N-methyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (1.5 equiv., 0.42 mmol), Cs2CO3 (3.0 equiv., 0.84 mmol), Pd2(dba)3 (0.1 equiv., 0.028 mmol), and Xantphos (0.2 equiv., 0.056 mmol) were dissolved in 20 mL of dioxane and argon was bubbled through the reaction mixture. The reaction was kept in oil bath to reflux at 100°C for 18 -h. The reaction was then cooled to room temperature, filtered to remove inorganics, extracted with EtOAc, saturated NaHCO3 and brine, and the organics were dried over Na2SO4, then concentrated under reduced pressure to give an orange solid (147.2 mg, 93% yield). The resulting crude material was used as is to prepare 4-[7- (2-chloro-5-hydroxy-phenyl)-6-methyl-benzo[l,2,4]triazin-3-ylamino]-N-methyl-N-(2- pyrrolidin-l-yl-ethyl)-benzenesulfonamide as described in the next example.
Example 73 Synthesis of 4-[7-(2-chloro-5-hydroxy-phenyI)-6-methyl- benzo[l,2,41triazin-3-ylamino]-N-methyl-N-(2-pyrrolidin-l-yl-ethyl)- benzenesulfonamide
Figure imgf000115_0001
(LXXVIII)
[0185] 4-[7-(2-chloro-5-methoxy-phenyl)-6-methyl-benzo[l,2,4]triazin-3-ylamino]- N-methyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (1.0 equiv., 0.26 mmol) obtained as described in Example 72, was dissolved in anhydrous DCM, purged of air using house vacuum, and placed under an argon blanket. Neat BBr3 (4.0 equiv., 1.04 mmol) was then added via syringe and allowed to stir at room temperature for 2 h. The reaction was quenched using saturated NaHCO3, and the resulting precipitate was filtered and washed with water and Et2O to afford an olive green solid. The compound was purified by flash chromatography using 90: 10 DCM/MeOH to recover a yellow/orange solid (88.2 mg, 62% yield). Rf = 0.19 1H NMR (DMSO-d6): δ 1.19 (t, J= 7.32 Hz, 2H), 1.89 (m, 2H), 2.01 (m, 2H), 2.29 (s, 3H), 2.73 (s, 3H), 3.06 (m, 4H), 6.82 (d, J= 2.85 Hz, IH), 6.91 (dd, J= 2.91 Hz, 8.61 Hz, IH), 7.40 (d, J= 8.81 Hz, IH), 7.79 (s, IH), 7.83 (d, J= 8.95 Hz, 2H), 8.11 (s, IH), 8.26 (d, J- 8.90 Hz, 2H). MS (ES+): m/z = 555 LC retention time: 2.28 min.
Example 74 Synthesis of Acetic acid 4-chloro-3-{5-methyl-3-[4-(2-pyrrolidin-l-yl- ethoxyVphenylamino] -b enzo f 1 ,2,4] triazin-7-yll-phenyl ester
Figure imgf000116_0001
(LXXIX)
[0186] To a solution of 4-chloro-3- {5-methyl-3-[4-(2-pyrrolidin-l-yl-ethoxy)- phenylamino]-benzo[l,2,4]triazin-7-yl}-phenol hydrochloride (1.0 equiv., 0.978 mmol) in DCM (30 mL) was added neat Et3N (10 equiv., 4.89 mmol) under an argon rich atmosphere and stirred for 10 minutes. Neat Acetyl chloride (1.2 equiv., 1.17 mmol) was added dropwise via syringe to the reaction mixture and allowed to stir at room temperature for 24 h. The reaction was quenched using saturated NaHCO3; the organics were dried over Na2SO4 and then concentrated down under reduced pressure. The resulting crude solid was then brought up in DCM and purified by flash chromatography using a solvent system of 85:10:5 (v/v/v) DCM/MeOH/Et3N. The fractions were concentrated under reduced pressure and precipitated out using 1:5 (v/v) DCM/hexanes to afford an orange solid (409.1 mg, 75% yield). 1H NMR (DMSO-d6) : δ 1.68 (m, 4H), 2.29 (s, 3H), 2.51 (m, 4H), 2.63 (s, 3H), 2.78 (t, J= 5.94 Hz, 2H), 4.07 (t, J= 5.99 Hz, 2H), 6.99 (dd, J= 7.05 Hz, 12.55 Hz, 2H), 7.27 (dd, J= 5.90 Hz, 8.75 Hz, IH), 7.41 (d, J - 2.80 Hz, IH), 7.67 (d, J= 8.75 Hz, IH), 7.85 (m, IH), 7.93 (d, J= 9.05 Hz, 2H), 8.17 (m, IH), 10.83 (s, IH). Rf = 0.29 MS (ES+) m/z = 520, LC retention time 2.57 minutes. Example 75 Synthesis of Acetic acid 4-chloro-3-{5-methyl-3-[3-(2-pyrrolidin-l-vI- ethoxy)-phenylamino1-benzo[l,2,4]triazin-7-yl}-phenyl ester
Figure imgf000117_0001
(LXXX)
[0187] The title product was synthesized and had the following characteristics. 1H NMR (DMSO-d6) : δ 1.73 (m, 4H), 2.30 (s, 3H), 2.65 (m, 4H), 2.68 (s, 3H), 2.92 (m, 2H), 4.16 (t, J= 5.85 Hz, 2H), 6.67 (dd, J- 2.49 Hz, 8.24 Hz, IH), 7.29 (t, J= 8.45 Hz, 2H), 7.43 (d, J= 2.74 Hz, IH), 7.48 (d, J= 9.58 Hz, IH), 7.68 (d, J= 8.69 Hz, IH), 7.92 (d, J= 11.72 Hz, 2H), 8.23 (s, IH). MS (ES+) m/z =519, LC retention time: 2.61 minutes.
Example 76 Synthesis of 4-methyl-benzoic acid 4-chloro-3- 5-methyl-3-[4-(2- pyrrolidin-l-yl-ethoxy)-phenylamino]-benzo[l,2,4]triazin-7-yI}-phenyl ester
Figure imgf000117_0002
(LXXXI)
[0188] The title product was synthesized and had the following characteristics. 1H NMR (DMSO-de) : δ 1.69 (m, 4H), 2.42 (s, 3H), 2.52 (m, 4H), 2.64 (s, 3H), 2.78 (t, J= 5.96 Hz, 2H), 4.06 (t, J= 5.97 Hz, 2H), 6.99 (d, J= 5.0 Hz, 2H), 7.42 (d, J= 7.5 Hz, 2H), 7.44 (d, J= 2.8 Hz, IH), 7.58 (d, J= 2.8 Hz, IH), 7.72 (d, J= 8.7 Hz, IH), 7.88 (d, J= 0.85 Hz, IH), 7.92 (d, J= 9.15 Hz, 2H), 8.05 (d, J= 6.55 Hz, 2H), 8.21 (d, J= 1.75 Hz, IH), 10.83 (s, IH). MS (ES+) m/z =594.5, LC retention time: 3.08 minutes. Example 77 Synthesis of 2,2-dimethyl-propionic acid 4-chloro-3-{5-methyl-3-[4-(2- pyrrolidin-l-yI-ethoxy)-phenylamino]-benzo[l,2,41triazin-7-yl}-phenyl ester
Figure imgf000118_0001
(LXXXII)
[0189] The title product was synthesized and had the following characteristics. 1H NMR (DMSO-de) : δ 1.31 (s, 9H), 1.69 (m, 4H), 2.54 (m, 4H), 2.64 (s, 3H), 2.80 (t, J= 5.67 Hz, 2H), 4.07 (t, J= 5.95 Hz, 2H), 7.0 (d, J= 2.2 Hz, 2H), 7.25 (dd, J= 2.55 Hz, 8.45 Hz, IH), 7.39 (d, J= 2.7 Hz, IH), 7.67 (d, J= 8.75 Hz, IH), 7.85 (d, J 0.9 Hz, IH), 7.93 (d, J= 9.15 Hz, 2H), 8.19 (d, J= 1.65 Hz, IH), 10.84 (s, IH). MS (ES+) m/z =560.4, LC retention time: 2.98 minutes.
Example 78 Synthesis of Isobutyric acid 4-chloro-3-{5-methyl-3-f4-(2-pyrrolidin-l- yl-ethoxy)-phenylamino]-benzo[l,2,4]triazin-7-yI}-phenyl ester
Figure imgf000118_0002
(LXXXIII)
[0190] The title product was synthesized and had the following characteristics. 1H NMR (DMSO-d6) : δ 1.23 (s, 3H), 1.25 (s, 3H), 1.69 (m, 4H), 2.53 (m, 4H), 2.64 (s, 3H), 2.79 (t, J= 5.88 Hz, 2H), 4.07 (t, J= 5.94 Hz, 2H), 6.99 (d, J= 7.05 Hz, 2H), 7.25 (dd, J = 2.85 Hz, 8.8 Hz, IH), 7.41 (d, J= 2.85 Hz, IH), 7.67 (d, J= 8.8Hz, IH), 7.85 (d, J= 1.1 Hz, IH), 7.93 (d, J= 9.05 Hz, 2H), 8.18 (d, J= 1.85 Hz, IH), 10.84 (s, IH). MS (ES+) m/z =548.7, LC retention time: 2.80 minutes. Example 79 Synthesis of 7-bromo-l-oxy-benzo[1.2,4]triazin-3-ylamine
Figure imgf000119_0001
(LXXXIV)
[0191] 4-bromo-2-nitro-phenylamine (11.0 g) and cyanamide (11.0 g) were weighed out into a 1000 mL round bottom flask and heated to 100°C until melted. Heating was removed and concentrated HCI (35 mL) was added slowly in multiple small portions as this addition was extremely exothermic. Upon complete addition of HCI, reaction heating was resumed at 110°C for 4 hours. Reaction heating was then removed and 30% NaOH was added slowly in sufficient quantity so as to bring the reaction pH to about 13- 14. This resulted in copious yellow precipitate. Reaction heating was resumed for 1 hour. The reaction mixture was cooled, diluted with water (400 mL) and filtered. Isolated solids were dried, taken up in DCM and filtered again to provide the pure material. Yellow solids (9.56 g, 80% yield). MS (ESI+): m/z = 243.0, !H NMR (DMSO- de): δ 7.46(d, J=9.1 Hz, IH), 7.88 (dd, J= 9.0 Hz, J= 2.4 Hz, IH), 8.25 (d, J= 2.3 Hz, IH).
Example 80 Synthesis of 7-bromo-5-nitro-l-oxy-benzo[l,2,4]triazin-3-ylamine
Figure imgf000119_0002
(LXXXV)
[0192] In a 1000 mL round bottom flask, 7-bromo-l-oxy-benzo[l,2,4]triazin-3- ylamine (9.5 g, 39.4 mmol, 1 equiv) was dissolved completely in sulfuric acid (24 mL) and chilled to 0°C using an ice bath. Fuming nitric acid (24 mL) was added in 3 portions over several minutes while the solution was stirred. The reaction was then allowed to come to ambient temperature and the stirring was continued for 16 hours. The contents were then poured carefully onto ice and the resulting solids yellow were collected and dried (10.75 g, 95% yield). MS (ESI+): m/z = 288.0, LC retention time: 2.39 min. Rf = 0.65, 30% EtOAc/ hexanes. 1H NMR (DMSO-d6): δ 7.99 (bs, 2H), 8.49 (d, J= 2.4 Hz, IH), 8.59 (d, J= 2.4 Hz, IH).
Example 81 Synthesis of 7-(2,6-dimethvI-phenyl)-5-nitro-l-oxy-benzo[l,2,41triazin-
3-yIamine
Figure imgf000120_0001
(LXXXVI)
[0193] An oven dried 25 mL round bottom flask was charged with 7-bromo-5-nitro-l- oxy-benzo[l,2,4]triazin-3-ylamine (0.1 g, 0.35 mmol, 1 equiv), 2,6-dimethyl phenyl boronic acid (0.105 g, 0.69 mmol, 2 equiv), sodium carbonate (0.15 g, 1.39 mmol, 4 equiv) and tetrakis(triphenylphosphine) palladium(O) (0.04 g, 0.035 mmol, 0.1 equiv). The reactants were flushed with argon and diluted with ethylene glycol dimethyl ether (6 mL), ethanol (1 mL) and DI water (1 mL). The reaction vessel was outfitted with condenser and refluxed for 18 hours. The reaction was cooled to ambient temperature and the crude product was filtered, diluted with ethyl acetate, and washed with brine. The brine layer was back extracted once with fresh ethyl acetate. The organic phases were combined and dried over sodium sulfate (Na2SO4). Filtration was followed by evaporation and silica gel chromatography (2:3 EtOAc/hexanes) provided the desired product as a yellow powder (0.08 g, 74% yield). MS (ESI+): m/z = 312.4, LC retention time: 2.96 min. Rf = 0.42, 30% EtOAc/hexanes. Example 82 Synthesis of 4-[7-(2,6-dimethyl-phenyl)-5-nitro-l-oχy- benzofl,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesnlfonamide
Figure imgf000121_0001
(LXXXVII)
[0194] In a dry 25 mL round bottom flask, 7-(2,6-dimethyl-phenyl)-5-nitro- 1 -oxy- benzo[l,2,4]triazin-3-ylamine (0.069 g, 0.22 mmol, 1 equiv), 4-bromo-N-(2-pyrrolidin-l- yl-ethyl)-benzenesulfonamide (O.llg, 0.33 mmol, 1.5 equiv), cesium carbonate (0.22g, 0.67 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.026 g, 0.044 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.021 g, 0.022mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (8 L) and outfitted with reflux condenser. The reaction was heated to reflux for 6 hours, then filtered hot, and the solvents were diluted with ethyl acetate and washed with brine. The brine layer was back extracted once with fresh ethyl acetate. The organic phases were combined and dried over sodium sulfate (Na2SO4). Filtration followed by evaporation and silica gel chromatography (6:1 DCM/ MeOH) provided desired product as a yellow powder (0.079 g, 63% yield). MS (ESI+):m/z = 564.4, LC retention time: 2.73 min.
Example 83 Synthesis ϋf 4-[5-amino-7-(2,6-dimethyl-phenyl)-benzofl,2,4]triazin-3- ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000121_0002
(LXXXVIII) [0195] 4-[7-(2,6-dimethyl-ρhenyl)-5-nitro-l-oxy-benzo[l,2,4]triazin-3-ylamino]-N- (2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.079 g, 0.14 mmol, 1 equiv) was combined with 10% palladium on carbon (0.029 g) and flushed with argon. The reactants were then diluted with methanol (5 mL) and the reaction atmosphere was evacuated and replaced with hydrogen. A hydrogen balloon was affixed and the reaction was allowed to stir for 3 hours. Argon was then bubbled through the reaction mixture and contents were filtered though a pad of Celite. Solvents were evaporated to provide a green solid product (0.045 g, 62% yield). MS (ESI+): 518.4 (M+H), r.t .= 2.56 min.
Example 84 Synthesis of N-{7-(2,6-dimethyl-phenyl)-3-r4-(2-pyrrolidin-l-yl- ethylsulfamoyl -phenylamino]-benzo[l,2,4]triazin-5-yl}-acetamide TFA salt
Figure imgf000122_0001
(LXXXIX)
[0196] A 0°C solution of 4-[5-amino-7-(2,6-dimethyl-phenyl)-benzo[l,2,4]triazin-3- ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.045 g, 0.087 mmol, 1 equiv) in DCM (3 L) was treated with acetyl chloride (0.011 g, 0.14 mmol, 1.6 equiv) and allowed to warm to ambient temperature. After 18 hours, the reaction solvents were evaporated to provide crude residue. HPLC purification provided the TFA salt of desired product as a yellow powder (0.011 g, 23% yield). MS (ESI+): m/z = 560.4, LC retention time: 2.67 min. 1H MR (DMSO-d6): δ 1.85-1.88 (m, 2H), 1.99-2.01 (m, 2H), 2.07 (s, 6H), 2.28 (s, 3H), 3.02-3.08 (m, 4H), 3.23-3.27 (m, 2H), 3.54-3.57 (m, 2H), 7.20 (d, J=7.6 Hz, 2H), 7.24-7.27 (m, IH), 7.87 (d, J=8.9 Hz, 2H), 7.89 (t, J=6.1 Hz, IH), 7.94 (d, J=1.8 Hz, IH), 8.26 (d, J=8.9 Hz, 3H), 9.56 (bs, IH), 9.84 (s, IH) 11.48 (s, IH). Example 85 Synthesis of N-r3-(4-racetyl-f2-pyrrolidin-l-yl-ethyl)-sttlfamoyll- phenylamino|-7-(2,6-dimethyl-phenyl)-benzofl,2,4]triazin-5-yl]-acetamide
Figure imgf000123_0001
(XC)
[0197] A 0°C degree solution of 4-[5-amino-7-(2,6-dimethyl-phenyl)- benzo[l,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.045 g, 0.087 mmol, 1 equiv) in DCM (3 mL) was treated with acetyl chloride (0.011 g, 0.14 mmol, 1.6 equiv) and allowed to warm to ambient temperature. After 18 hours, the reaction solvents were evaporated to provide crude residue. HPLC purification provided the TFA salt of the desired product as a yellow powder (0.004 g, 8% yield). MS (ESI+): m/z = 602.4, LC retention time: 2.83 min. 1H NMR (DMSO-d6): δ 1.87-1.89 (m, 2H), 2.04-2.08 (m, 8H), 2.29 (s, 3H), 2.36 (s, 3H), 3.09-3.11 (m, 2H), 3.64-3.66 (m, 2H), 4.08 (t, J=7.0 Hz, 2H), 7.20 (d, J=7.6 Hz, 2H), 7.24-7.27 ( , IH), 7.96 (d, J=1.6 Hz, IH), 8.04 (d, J=9.0 Hz, 2H), 8.26-8.30 (m, 3H), 9.73 (bs, IH), 9.85 (s, IH) 11.60 (s, IH).
Example 86 Synthesis of 4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-l-yI-ethyl -benzenesnlfonamide
Figure imgf000123_0002
(XCI)
[0198] hi a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.157 g, 0.523 mol, 1 equiv), 4-bromo-N-(2-pyrrolidin- l-yl-ethyl)-benzenesulfonamide (0.261 g, 0.785 mmol, 1.5 equiv), cesium carbonate (0.512 g, 1.57 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.061 g, 0.10 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.048 g, 0.052mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (5 mL) and outfitted with reflux condenser. The reaction was heated to reflux for 18 hours, then filtered hot, and the solvents were diluted with ethyl acetate and washed with brine. The brine layer was back extracted once with fresh ethyl acetate. The organic phases were combined and dried over sodium sulfate (Na2SO4). Filtration followed by evaporation provided slightly impure product. Solids were diluted in minimum amount of DCM and precipitated out using excess hexanes. The resulting solids were filtered off and dried to yield a yellow powder (0.276 g, 95% yield). MS (ESI+): m/z = 553.3, LC retention time: 2.77 min.
Example 87 Synthesis of 4-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl- benzo[l,2,4]triazm-3-ylammo]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide TFA salt
Figure imgf000124_0001
(XCII)
[0199] 4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1 ,2,4]triazin-3-ylamino]- N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.055 g, 0.099 mmol, 1 equiv) was diluted with 15 mL DCM and chilled to 0°C using an ice bath. A 1.0 M solution of BBr3 in DCM (0.25 mL, 0.25 mmol, 2.5 equiv) was then added in one portion resulting in a dark reaction mixture. The reaction was allowed to come to ambient temperature and stirred for 15 hours, then quenched by carefully pouring onto a saturated solution of sodium bicarbonate followed by sonication for 3-5 minutes. Additional DCM was added and this mixture was washed with sodium bicarbonate followed by brine. The organic phase was separated, dried over sodium sulfate (Na2SO4), filtered and evaporated to dryness. HPLC purification provided the TFA salt of desired product as a yellow solid (0.013 g, 25% yield). MS (ESI+): m/z = 539.3 , LC retention time: 2.50 min. 1H NMR (DMSO-d6): δ 1.86 (bs, 2H), 1.99(bs, 2H), 2.71 (s, 3H), 2.28 (s, 3H), 3.04-3.07 (m, 4H), 3.23(bs, 2H), 3.55(s, 2H), 6.53 (s, IH), 6.89 (dd, J=8.7 Hz, J=2.9 Hz, IH), 6.94 (d, J=2.9 Hz, IH), 7.41 (d, J=8.7 Hz, IH), 7.83 (bs, IH), 7.87 (d, J=8.8 Hz, 2H), 7.92 (s, IH), 8.22 (d, J=1.7 Hz, IH), 8.26 (d, J=8.8 Hz, 2H), 9.55 (bs, IH), 9.96 (s, IH) 11.44 (s, IH).
Example 88 Synthesis of 3-(3-amino-5-methyl-benzo[l,2,41triazin-7-yl)-4-chloro- phenol
Figure imgf000125_0001
(XCIII)
[0200] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (0.158
1 g, 0.527 mmol, 1 equiv) was diluted with 10 mL DCM and chilled to 0°C using an ice bath. A 1.0 M solution of BBr3 in DCM (3.16 mL, 3.16 mmol, 6.0 equiv) was then added in one portion resulting in a dark reaction mixture. The reaction was allowed to come to ambient temperature and stirred for 3.5 hours, then quenched by carefully pouring onto a saturated solution of sodium bicarbonate followed by sonication for 3-5 minutes. Additional DCM was added and this mixture was washed with sodium bicarbonate followed by brine. The organic phase was separated, dried over sodium sulfate (Na2SO4), filtered and evaporated to dryness. Solids were then diluted with minimum amount of ethyl acetate and precipitated out with hexanes. Product was filtered off and dried to yield a yellow solid (0.110 g, 73 % yield). MS (ESI+): m/z = 287.1, LC retention time: 2.46 min. Example 89 Synthesis of N-{5-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-ylaminol-pyridin-2-yl}-acetamide TFA salt
Figure imgf000126_0001
(XCTV)
[0201] In a dry 25 mL round bottom flask, 3-(3-amino-5-methyl-benzo[l,2,4]triazin- 7-yl)-4-chloro-phenol (0.0641 g, 0.224 mmol, 1 equiv), N-(5-bromo-pyridin-2-yl)- acetamide (0.0723. g, 0.336 mmol, 1.5 equiv), cesium carbonate (0.220 g, 0.672 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.026 g, 0.0448 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.0205 g, 0.0224 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (8 mL) and outfitted with reflux condenser. The reaction was heated to reflux for 18 hours, then filtered hot and solvents were diluted with ethyl acetate and washed with brine. The brine layer was back extracted once with fresh ethyl acetate. The organic phases were combined and dried over sodium sulfate (Na2SO4). Filtration followed by evaporation and HPLC purification provided the TFA salt of desired product as a yellow powder (0.004 g, 4% yield). MS (ESI+): m/z = 421.1, LC retention time: 2.44 min. 1H NMR (DMSO-d6): δ 2.09 (s, 3H), 2.65 (s, 3H), 6.87 (dd, J=8.7 Hz, J=2.9 Hz, IH), 6.93 (d, J=2.9 Hz, IH), 7.40 (d, J=8.7 Hz, IH), 7.86 (s, IH), 8.11 (d, J=8.9 Hz, IH), 8.16 (d, J=1.7 Hz, IH), 8.39 (dd, J=9.0 Hz, J=2.7 Hz, IH), 8.93 (d, J=2.5 Hz, IH), 9.92 (bs, IH), 10.47 (s, IH), 11.05 (s, IH). Example 90 Synthesis of l-[2-(3-bromo-phenoxy)-etI--yl1-pyrrolidine
Figure imgf000127_0001
(XCV)
[0202] 3-bromo-ρhenol (5.34 g, 30.9 mmol, 1 equiv), l-(2-chloro-ethyl)-pyrrolidine (5.24 g, 30.9 mmol, 1 equiv) and potassium carbonate (K2CO3) (34 g, 24.7 mmol, 8 equiv) were weighed out into a 500 mL round bottom flask, were diluted with 150 mL DMF and stirred for 2 days. The contents were poured onto ethyl acetate water mixture and extracted once. The organic layer was separated, washed with brine and dried over sodium sulfate (Na2SO4). Filtration followed by concentration provided crude product as a pale yellow oil. Column chromatography (1 :9 ethyl acetate/ hexanes) provided desired product as a yellow oil (3.6 g, 43% yield). 1H NMR (DMSO-d6): δ 1.65-1.69 (m, 4H), 2.72 (s, IH), 2.75 (t, J= 5.8 Hz, 2H), 2.88 (s, IH), 4.06 (t, J= 5.8 Hz, 2H), 6.95 (dd, J= 8.35 Hz, IH), 7.11 (d, J=7.7, IH), 7.13-7.14 (m, IH), 7.22 (t, J= 8.1Hz, IH).
Example 91 Synthesis of [7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-yI]-[3-(2-pyrrolidin-l-yl-ethoxy -phenyl1-amine
Figure imgf000127_0002
(XCVI)
[0203] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.113 g, 0.377 mmol, 1 equiv), l-[2-(3-bromo-phenoxy)- ethyl]-pyrrolidine (0.153 g, 0.565 mmol, 1.5 equiv), cesium carbonate (0.368 g, 1.13 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.044 g, 0.0753 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.034 g, 0.0376 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (8 mL) and outfitted with reflux condenser. The reaction was heated to reflux for 18 hours, . . then filtered hot and the solvents were diluted with ethyl acetate and washed with brine. The brine layer was back extracted once with fresh ethyl acetate. The organic phases were combined and dried over sodium sulfate (Na2SO4). Filtration followed by evaporation and column chromatography (1 :5 MeOH/ DCM) provided desired product as a yellow powder (0.12 g, 65% yield). MS (ESI+): m/z = 491.1, LC retention time: 2.94 min.
Example 92 Synthesis of 4-chloro-3-{5-methyl-3-f3-(2-pyrroIidin-l-yl-ethoxy)- phenylamino1-benzo[l,2,41triazin-7-yl}-phenol TFA salt
Figure imgf000128_0001
(XCNII)
[0204] [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-yl]-[3-(2- pyrrolidin-l-yl-ethoxy)-phenyl] -amine (0.120 g, 0.245 mmol, 1 equiv) was diluted with 8 mL DCM and chilled to 0°C using an ice bath. A 1.0 M solution of BBr3 in DCM (2 mL, 1.96 mmol, 8.0 equiv) was then added in one portion resulting in a dark reaction mixture. The reaction was allowed to come to ambient temperature and stirred for 3.5 hours, then quenched by carefully pouring onto a saturated solution of sodium bicarbonate, followed by sonication for 5 minutes. Additional DCM was added and this mixture was washed with sodium bicarbonate followed by brine. The organic phase was separated, dried over sodium sulfate (Νa SO4), filtered and evaporated to dryness. Solids were then diluted with minimum amount of ethyl acetate and precipitated out with hexanes. Product was filtered off and dried. HPLC purification provided TFA salt of desired product as a yellow solid (0.008 g, 7 % yield). MS (ESI+): m/z = 476.1, LC retention time: 2.64 min. 1H NMR (DMSO-d6): δ 1.88-1.91 (m, 2H), 2.04-2.07 (m, 2H), 2.68 (s, 3H), 3.17-3.20 (m, 2H), 3.64-3.66 (m, 4H), 4.36 (t, J=4.7 Hz, 2H), 6.74 (dd, J=8.1 Hz, J=2.3 Hz, IH), 6.88 (dd, J=8.5 Hz, J=2.8 Hz, IH), 6.93 (d, J=2.9 Hz, IH), 7.35 (t, J=8.2 Hz, IH), 7.41 (d, J=8.7 Hz, IH), 7.55 (d, J=8.7 Hz, IH), 7.88 (s, IH), 7.95-7.96 (m, IH), 8.17 (d, J=1.7 Hz, IH), 9.79 (bs, IH), 9.95 (s, IH), 11.04 (s, IH).
Example 93 Synthesis of {4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzofl,2,4]triazin-3-yIaminol-phenyll-(4-methyl-piperazin-l-yl)-methanone
Figure imgf000129_0001
(xcviπ)
[0205] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.282 g, 0.94 mmol, 1 equiv), (4-bromo-phenyl)-(4- methyl-piperazin-l-yl)-methanone (0.399 g, 1.41 mmol, 1.5 equiv), cesium carbonate (0.919 g, 2.82 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.109 g, 0.188 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.086 g, 0.094 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (12 mL) and outfitted with reflux condenser. The reaction was heated to reflux for 18 hours, then filtered hot and solvents were evaporated. The residue was diluted with toluene and water (approx. 2: 1), and the mixture was heated to 70°C and stirred for 20 min. The contents were then poured into 250 mL separatory funnel and shaken. The resulting precipitate was filtered off, washed with water and dried to yield a yellow powder (0.275 g, 58% yield). MS (ESI+): m/z = 504.1, LC retention time: 1.75 min.
Example 94 Synthesis of {4-[7-(2-chloro-5-hydroxy-phenyI)-5-methyl- benzo[l,2,4]triazin-3-ylamino]-phenyll-(4-methyl-piperazin-l-yl)-methanone TFA salt
Figure imgf000130_0001
(XCIX)
[0206] {4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]- phenyl}-(4-methyl-piperazin-l-yl)-methanone (0.139 g, 0.276 mmol, 1 equiv) was diluted with 10 mL DCM and chilled to 0°C using an ice bath. A 1.0 M solution of BBr3 in DCM (1.66 mL, 1.66 mmol, 6.0 equiv) was then added in one portion resulting in a dark reaction mixture. The reaction was allowed to come to ambient temperature and stirred for 1 hour, then quenched by carefully pouring onto a saturated solution of sodium bicarbonate followed by sonication for 5 minutes. The resulting solids were filtered off and dried. HPLC purification provided desired product as TFA salt to yield a yellow solid (0.024 g, 18 % yield). MS (ESI+): m/z = 489.1, LC retention time: 2.11 min. 1H NMR (DMSO-d6): δ 2.69 (s, 3H), 2.84 (s, 3H), 3.22 (bs, 2H), 6.88 (dd, J=8.8 Hz, J=2.9 Hz, IH), 6.93 (d, J=2.9 Hz, IH), 7.41 (d, J=8.7 Hz, IH), 7.55 (d, J=8.7 Hz, 2H), 7.89 (s, IH), 8.14 (d, J=8.7 Hz, 2H), 8.19 (d, J=1.3 Hz, IH), 9.88 (bs, IH), 9.95 (bs, IH), 11.24 (s, IH).
Example 95 Synthesis of 5-[7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,41triazm-3-ylammo]-pyridine-2-carboxylic acid (2-pyrrolidin-l-yl-ethylV amide
Figure imgf000131_0001
(C)
[0207] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.090 g, 0.299 mmol, 1 equiv), 5-bromo-pyridine-2- carboxylic acid (2-pyrrolidin-l-yl-ethyl)-amide (0.134 g, 0.449mmol, 1.5 equiv), cesium carbonate (0.293 g, 0.899 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.035 g, 0.059 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.027 g, 0.0299 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (8 mL) and outfitted with reflux condenser. The reaction was heated to reflux for 24 hours, then sonicated and slowly diluted with water. The resulting orange precipitate was filtered and dried. Column chromatography (1:5 MeOH/ DCM) provided desired product as a yellow solid (0.085 g, 55% yield).
Example 96 Synthesis of 5-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-ylamino1-pyridine-2-carboxylic acid (2-pyrrolidin-l-yl-ethyl)- amide HCI salt
Figure imgf000131_0002
(CD [0208] 5-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo [ 1 ,2,4]triazin-3-ylamino] - ■ pyridine-2-carboxylic acid (2-pyrrolidin-l-yl-ethyl)-amide (0.085 g, 0.164 mmol, 1 equiv) was diluted with 20 mL DCM and chilled to 0°C using an ice bath. A 1.0 M solution of BBr3 in DCM (1 mL, 0.984 mmol, 6.0 equiv) was then added in one portion resulting in a dark reaction mixture. The reaction was allowed to come to ambient temperature and stirred for 6 hours, then quenched by carefully pouring onto a saturated solution of sodium bicarbonate followed by sonication for 5 minutes. The resulting solids were filtered off, dried, and dissolved in DCM (10 mL) and MeOH (20 mL) mixture and treated with 2.0 M solution of HCI in diethyl ether (0.16 mL, 0.317 mmol, 2 equiv). The solvents were evaporated leaving desired product as HCI salt in a form of a yellow solid (0.084 g, 95 % yield). MS (ESI+): m/z = 504.1, LC retention time: 2.23 min. 1H NMR (DMSO-d6): δ 1.86-1.88 (m, 2H), 1.99-2.02 (m, 2H), 2.70 (s, 3H), 3.01- 3.05 (m, 2H), 3.33-3.37 (m, 2H), 6.90 (dd, J=8.7 Hz, J=2.9 Hz, IH), 6.97 (d, J=2.9 Hz, IH), 7.41 (d, J=8.8 Hz, IH), 7.92-7.93 (m, IH), 8.12 (d, J=8.7 Hz, IH), 8.22 (d, J=1.5 Hz, IH), 8.51 (dd, J=8.6 Hz, J=2.5 Hz, IH), 9.06 (t, J=5.9 Hz, IH), 9.43 (d, J=2.4 Hz, IH), 10.17 (bs, IH), 11.50 (s, IH).
Example 97 Synthesis of [7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-yn-f4-(3-pyrrolidin-l-yl-propane-l-sulfonyI)-phenyl]-amine
Figure imgf000132_0001
(CH)
[0209] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.0361 g, 0.122 mmol, 1 equiv), l-[3-(4-bromo- benzenesulfonyl)-propyl]-pyrrolidine (0.061 g, 0.184 mmol, 1.5 equiv), cesium carbonate (0.120 g, 0.367 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.014 g, 0.0244 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.012 g, 0.0122 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (6 mL) and outfitted with reflux condenser. The reaction was heated to reflux for 18 hours, filtered hot, diluted with ethyl acetate and washed with brine. The brine layer was back extracted once with fresh ethyl acetate. The organic phases were combined and dried over sodium sulfate (Na2SO4). Filtration followed by evaporation provided crude product, whichwas dissolved in minimum amount of DCM and precipitated out with excess hexanes. Solids were filtered off and dried to yield a yellow powder (0.06 g, 91% yield).
Example 98 Synthesis of 4-chloro-3-{5-methyl-3-[4-(3-pyrrolidin-l-yl-propane-l- sulfonyl)-phenylamino]-benzo[l,2,4]triazin-7-yl}-phenol TFA salt
Figure imgf000133_0001
(CIII)
[0210] [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-yl]-[4-(3- pyrrolidin-l-yl-propane-l-sulfonyl)-phenyl]-amine (0.080 g, 0.145 mmol, 1 equiv) was diluted with 10 mL DCM and chilled to 0°C using an ice bath. A 1.0 M solution of BBr3 in DCM (0.87 mL, 0.87 mmol, 6.0 equiv) was then added in one portion resulting in a dark reaction mixture. The reaction was allowed to come to ambient temperature and stirred for 6 hours, then quenched by carefully pouring onto a saturated solution of sodium bicarbonate followed by sonication for 3-5 minutes. The resulting solids were filtered off and dried. HPLC purification provided desired product as TFA salt in a form of a yellow solid (0.013 g, 17 % yield). MS (ESI+): m/z = 538.1, LC retention time: 2.35 min. 1H NMR (DMSO-de): δ 1.81-1.84 (m, 2H), 1.93-2.00 (m, 4H), 2.72 (s, 3H), 2.95- 2.99 (m, 2H), 3.20-3.24 (m, 2H), 3.53-3.54 (m, 2H), 6.89 (dd, J=8.7 Hz, J=2.9 Hz, IH), 6.94 (d, J=2.8 Hz, IH), 7.41 (d, J=8.7 Hz, IH), 7.95 (d, J=9.0 Hz, IH), 8.23 (d, J=1.6 Hz, IH), 8.31 (d, J=8.9 Hz, 2H), 9.61 (bs, IH), 9.98 (s, IH), 11.52 (s, IH). Example 99 Synthesis of N-[3-(3-amino-5-methyl-benzofl,2,41triazin-7-ylVphenyll- methanesulfonamide
Figure imgf000134_0001
(CIV)
[0211] An oven dried 25 mL round bottom flask was charged with 7-bromo-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.889 g, 3.72 mmol, 1 equiv), 3-(methylsulfonylamino) phenyl boronic acid (1.12 g, 5.21 mmol, 1.4 equiv), sodium carbonate (1.57 g, 14.9 mmol, 4 equiv) and tetrakis(triphenylphosphine) palladium(O) (0.43 g, 0.372 mmol, 0.1 equiv). The reactants were flushed with argon and diluted with ethylene glycol dimethyl ether (20 mL), ethanol (5 mL) and DI water (5 mL). The reaction vessel was outfitted with condenser and refluxed for 4 hours. The reaction was filtered hot and diluted with ethyl acetate. The Organic layer was isolated and concentrated to a dark residue. This was dissolved in DMF (6 mL) and slowly diluted with water so as to precipitate out the product. Solids were filtered off and dried to yield an orange solid (1 g, 82% yield).
Example 100 Synthesis of 4-[7-(3-methanesulfonylamino-phenyl)-5-methyl- benzo[l,2,41triazin-3-ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide TFA salt
Figure imgf000134_0002
(CV)
[0212] In a dry 25 mL round bottom flask, N-[3-(3-amino-5-methyl- benzo[l,2,4]triazin-7-yl)-phenyl]-methanesulfonamide (0.12 g, 0.365 mmol, 1 equiv), 4- bromo-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.182 g, 0.55 mmol, 1.5 equiv), cesium carbonate (0.357 g, 1.09 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9- dimethyl xanthene (0.0422 g, 0.073 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.033 g, 0.036 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (10 mL) and outfitted with reflux condenser. The reaction was heated to reflux for 2 hours, then filtered hot and the solvents were evaporated. HPLC purification provided desired product as TFA salt, in a form of a yellow powder (0.02 g, 9% yield). MS (ESI+): m/z = 582.1, LC retention time: 2.21 min. 1H NMR (DMSO-d6): δ 1.85-1.87 (m, 2H), 1.99-2.01 (m, 2H), 2.74 (s, 3H), 3.04-3.08 (m, 3H), 3.04-3.07 (m, 7H), 3.23-3.25 (m, 2H), 3.56(bs, 2H), 7.29 (dd, J=8.0 Hz, J=1.6 Hz, IH), 7.52 (t, J=7.9 Hz, IH), 7.63 (d, J=8.0 Hz, 2H), 7.66 (bs, IH), 7.83-7.88 (m, 3H), 8.18 (bs, IH), 8.25 (d, J=8.9 Hz, 2H), 8.43 (d, J=1.7 Hz, IH), 9.57 (bs, IH), 9.92 (s, IH) 11.43 (s, lH).
Example 101 Synthesis of 5-methyl-7-(3-nitro-phenyl)-benzofl,2,41triazin-3- ylamine
Figure imgf000135_0001
(CVI)
[0213] An oven dried 25 mL round bottom flask was charged with 7-bromo-5-methyl- benzo[l,2,4]triazin-3-ylamine (1.05 g, 4.49 mmol, 1 equiv), 3-Nitro phenyl boronic acid (1.05 g, 6.29 mmol, 1.4 equiv), sodium carbonate (1.9 g, 18 mmol, 4 equiv) and tetrakis(triphenylphosphine) palladium(O) (0.519 g, 0.449 mmol, 0.1 equiv). The reactants were flushed with argon and diluted with ethylene glycol dimethyl ether (6 mL), ethanol (1 mL) and DI water (1 mL). The reaction vessel was outfitted with condenser and refluxed for 2.5 hours. The reaction was cooled to ambient temperature and diluted with water. The resulting precipitate was filtered off, washed thoroughly with water and dried . Brown solids (0.9 g, 73% yield). MS (ESI+): m/z = 282.4, LC retention time: 2.77 min.
Example 102 Synthesis of 4-[5-methyl-7-(3-nitro-phenyl)-benzo[l,2,4]triazin-3- ylamino1-N-f2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000136_0001
(cvn)
[0214] hi a dry 25 mL round bottom flask, 5-methyl-7-(3-nitro-phenyl)- benzo[l,2,4]triazin-3-ylamine (0.223 g, 0.7935 mmol, 1 equiv), 4-bromo-N-(2- pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.396 g, 1.19 mmol, 1.5 equiv), cesium carbonate (0.776 g, 2.38 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.092 g, 0.159 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.073 g, 0.079 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (10 mL) and outfitted with a reflux condenser. The reaction was heated to reflux for 5 hours, then filtered hot and the solvents were evaporated. Silica gel column purification provided desired product as a yellow powder (0.16 g, 39% yield). MS (ESI+): m/z = 582.1
Example 103 Synthesis of 4-[5-methyl-7-(3-methylamino-phenyl - benzo[l,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-l-yl-ethylVbenzenesulfonamide TFA salt
Figure imgf000136_0002
(CVIII) [0215] 4-[7-(3-amino-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]-N-(2- pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.031 g, 0.062 mmol, 1 equiv) and cesium carbonate (0.012 g, 0.037 mmol, 0.6 equiv) were combined and diluted with dioxane (5 mL). The mixture was then treated with methyl iodide (0.013 g, 0.092 mmol, 1.5 equiv) and stirred at ambient temperature. After initial 5-10 minutes of stirring, DMF (1 mL) was added to improve solubility, followed by stirring for 16 hours, after which the solvents were evaporated and the resulting residue purified by HPLC chromatography to yield a yellow solid, the TFA salt (0.07g, 22% yield). MS (ESI+): m/z = 518.7, LC retention time: 1.86 min. 1H NMR (DMSO-d6): δ 2.08 (bs, 4H), 2.74 (s, 3H), 3.02 (s, 3H), 3.24 (m, 2H), 3.46-3.49 (m, 5H), 3.52-3.56(m, 3H), 6.8 (d, J=8.2 Hz, IH), 7.19 (m, 2H), 7.26-7.29 (m, IH), 7.88 (d, J=8.8 Hz, IH), 7.94 (t, J=7.7 Hz, IH), 8.16 (d, J=1.7 Hz, IH), 8.25 (d, J=8.8 Hz, 2H), 8.36 (d, J=1.7 Hz, IH), 11.44 (s, IH).
Example 104 Synthesis of 4-[7-(3-amino-phenyl)-5-methyl-benzo[l,2,4]triazin-3- ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000137_0001
(CDC)
[0216] 4-[5-methyl-7-(3-nitro-ρhenyl)-benzo[l,2,4]triazin-3-ylamino]-N-(2- pyrrolidin-l-yl-ethyl)-berizenesulfonamide (0.16 g, 0.3 mmol, 1 equiv) was combined with 10% palladium on carbon (0.135 g) and flushed with argon. The reactants were then diluted with methanol (15 mL), and rthe eaction atmosphere was evacuated and replaced with hydrogen. A hydrogen balloon was affixed and the reaction was allowed to stir for 3 hours. Argon was then bubbled through the reaction mixture and the contents were filtered though a pad of Celite. The solvents were evaporated to provide the product as an orange solid (0.15 g, 98% yield). MS (ESI+): m/z = 504.6, LC retention time: 1.84 min. 1H NMR (DMSO-d6): δ 1.64 (bs, 4H), 2.72 (s, 3H), 2.87 (bs, 2H), 5.26 (s, 2H), 6.65 (d, J=7.8 Hz, IH), 7.00 (d, J=8.1 Hz, IH), 7.05 (m, IH), 7.18 (t, J=7.8 Hz, IH), 7.83 (d, J=8.8 Hz, 2H), 8.12 (s, IH), 8.21 (d, J=8.8 Hz, 2H), 8.27 (d, J=1.7 Hz, IH), 11.44 (s, IH).
Example 105 Synthesis of 4-(4-bromo-benzenesulfonyl -piperazine-l-carboχylic acid tert-butyl ester
Figure imgf000138_0001
(CX)
[0217] To a stirring, 0°C solution of piperazine-1 -carboxylic acid tert-butyl ester (1.17 g, 6.29 mmol, 1.05 equiv) and TEA (1.67 mL, 11.98 mmol, 2.0 equiv) in DCM (10 mL) was added 4-bromo-benzenesulfonyl chloride (1.53 g, 5.99 mmol, 1 equiv). The reaction was allowed to warm to ambient temperature and stir for 3 h. The reaction solvents were evaporated and the resulting solids were dried, washed with water and filtered, yielding a white powder (2.24 g, 92% yield). MS (ESI+): m/z = 306.9, LC retention time: 3.17 min. Rf = 0.56 (30% EtOAc/hexanes).
Example 106 Synthesis of 4-{4-[7-(2,6-dichloro-phenyl)-5-methyl- benzo[l,2,41triazin-3-yIamino]-benzenesulfonyl}-piperazine-l-carboxylic acid tert- butyl ester
Figure imgf000138_0002
(CXI) [0218] In a dry 25 mL round bottom flask, 7-(2,6-dichloro-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.205 g, 0.672 mmol, 1 equiv), 4-(4-bromo- benzenesulfonyl)-piperazine-l -carboxylic acid tert-butyl ester (0.408 g, 1.01 mmol, 1.5 equiv), cesium carbonate (0.657 g, 2.02 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9- dimethyl xanthene (0.078 g, 0.134 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.0615 g, 0.067 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (20 mL) and outfitted with a reflux condenser. The reaction was heated to reflux for 2 hours, then filtered hot, and the solvents were evaporated. Purification on silica gel flash column provided desired product as a yellow solid (0.185 g, 44% yield). MS (ESI+): m/z = 529.0 (M -100), LC retention time: 3.51min. 1H NMR (DMSO-d6): δ 1.33 (s, 9H), 2.72 (s, 3H), 2.87 (m, 4 H), 3.4 (m, 4H), 7.5 (m, IH), 7.67 (d, J=8.7 Hz, 2H), 7.78 (m, 3H), 8.18 (d, J=1.7 Hz, IH), 8.30 (d, J=8.8 Hz, 2H), 11.53 (s, lH).
Example 107 Synthesis of [7-(2,6-dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3- yl1-[4-(piperazine-l-sttlfonylVphenyll-amine TFA salt
Figure imgf000139_0001
(CXII)
[0219] 4-{4-[7-(2,6-dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]- benzenesulfonyl}-piperazine-l -carboxylic acid tert-butyl ester (0.0696 g, 0.111 mmol, 1 equiv) was diluted with DCM (5 mL) and treated with TFA (0.051 mL, 0.664 mmol, 6 equiv). The resulting red solution was stirred at ambient temperature for 6 hours. Additional 6 equivalents of TFA were then added and the reaction was stirred for 12 hours. The solvents were then evaporated and the resulting solids collected and dried to yield a yellow powder (0.062 g, 99% yield). MS (ESI+): m/z = 528.9 , LC retention time: 2.70 min. 1H NMR (DMSO-d6): δ 2.72 (s, 3H), 3.13 (m, 4 H), 3.22 (m, 4H), 7.53 (m, IH), 7.68 (d, J=8.2 Hz, 2H), 7.81 (d, J=1.6 Hz, IH), 7.84 (d, J=8.7 Hz, 2 H), 8.20 (d, J=1.7 Hz, IH), 8.32 (d, J=8.8 Hz, 2H), 8.62 (bs, 2H), 11.53 (s, IH).
Example 108 Synthesis of 4-bromo-N-isopropyl-N-(2-pyrrolidin-l-yl-ethyl)- benzenesulfonamide
Figure imgf000140_0001
(CXIII)
[0220] 4-bromo-N-isopropyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (1.03 g, 3.09 mmol, 1 equiv), 2-bromo-propane (3.8 g, 31 mmol, 10 equiv) and cesium carbonate (2 g, 6.2 mmol, 2.0 equiv) were refluxed in acetone for 24h. The solvents were then removed and the resulting residue was dissolved in ethyl acetate and washed with water. The organic phase was then washed again with brine, dried over sodium sulfate, filtered and evaporated to yield the oily residue (0.86 g, 74% yield). MS (ESI+): m/z = 377.5, LC retention time: 2.16 min. R = 0.48 (10% MeOH:DCM).
Example 109 Synthesis of [7-(2,6-dichloro-phenyl -5-methyl-benzofl,2,4]triazin-3- yl]-[4-(piperazine-l-sulfonyl)-phenyl] -amine HCI salt
Figure imgf000140_0002
(CXIV)
[0221] In a dry 25 mL round bottom flask, 7-(2,6-dichloro-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.162 g, 0.533 mmol, 1 equiv), 4-bromo-N-isopropyl-N- (2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.3 g, 0.8 mmol, 1.5 equiv), cesium carbonate (0.522 g, 1.59 mmol, 3 equiv), 4,5-bis(diρhenylphosρhino)-9,9-dimethyl xanthene (0.062 g, 0.106 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.049 g, 0.0533 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (20 mL), and outfitted with a reflux condenser. The reaction was heated to reflux for 2 hours, then filtered hot and the solvents were evaporated. Purification on silica gel flash column provided desired product, 0.144 g yellow solids, which were taken up in 5 mL DCM and treated with 2 mL 2M HCI in ether solution. The solvents were then evaporated and the resulting solids were collected and dried to yield a yellow solid (0.16 g, 50% yield). MS (ESI+): m/z = 599.2, LC retention time: 2.99 min. 1H MR (DMSO-d6): δ 0.98 (d, 6H), 1.88 (m, 2 H), 2.01 (m, 2H), 2.72 (s, 3H), 3.02-3.08 (m, 2H), 3.37 (m, 2H), 3.46 (m, 2H), 3.62 (m, 2H), 4.02 (m, IH), 7.53 (m, IH), 7.67 (d, J=8.1 Hz, 2H), 7.79 (s, IH), 7.92 (d, J=8.7 Hz, 2H), 8.18 (d, J=1.6 Hz, IH), 8.27 (d, J=8.7 Hz, 2H), 10.7 (m, IH), 11.51 (s, IH).
Example 110 Synthesis of 4-[7-(2,6-dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3- ylamino]-N-(2-pyrroIidin-l-yl-ethyl)-benzenesulfonamide HCI salt
Figure imgf000141_0001
(CXV)
[0222] In a dry 25 mL round bottom flask, 7-(2,6-dichloro-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.193 g, 0.633 mmol, 1 equiv), 4-bromo-N-(2-pyrrolidin- l-yl-ethyl)-benzenesulfonamide (0.316 g, 0.95 mmol, 1.5 equiv), cesium carbonate (0.62 g, 1.89 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.073 g, 0.127 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.058 g, 0.0533 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (20 mL), and outfitted with a reflux condenser. The reaction was heated to reflux for 16 hours, then filtered hot and the solvents were evaporated. HPLC purification provided desired product. Pure HPLC fractions were combined, diluted with ethyl acetate and washed with saturated sodium bicarbonate solution. The organic phase was evaporated to dryness, diluted with DCM (10 mL) and treated with 2 mL 2M HCI in ether solution. The solvents were then evaporated and the resulting solids were collected and dried to yield a yellow solid (0.091 g, 26% yield). MS (ESI+): m/z = 559.6, LC retention time: 2.71 min. 1H NMR (DMSO-d6): δ 1.86 (bs, 2H), 1.98 (bs, 2H), 2.71 (s, 3H), 2.98-3.01 (m, 2H), 3.09-3.13 (m, 2H), 3.21-3.25 (m, 2H), 3.51-3.53 (m, 2H), 7.52 (m, IH), 7.67 (d, J=8.7 Hz, 2H), 7.79 (m, IH), 7.89 (dd, J=8.7 Hz, J=2.9 Hz, IH), 7.95 (t, J=6.1 Hz, IH), 8.18 (d, J=1.7 Hz, IH), 8.26 (d, J=8.8 Hz, 2H), 10.37 (bs, IH), 11.44 (s, IH).
Example 111 Synthesis of 7-(2-chloro-5-methoxy-phenyl)-5-methyl- b enzo [1 ,2,4] triazin-3-ylamine
Figure imgf000142_0001
(CXVI)
[0223] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.214 g, 0.713 mmol, 1 equiv), 4-bromo-N-isopropyl-N- (2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.321g, 0.856 mmol, 1.2 equiv), Cesium carbonate (0.698 g, 2.14 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.083 g, 0.143 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.065 g, 0.071 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (20 mL), and outfitted with a reflux condenser. The reaction was heated to reflux for 18 hours, then cooled to room temperature and the solvents were removed. The residue was diluted with DCM and purified via ISCO silica gel column chromatography (0% to 6% methanol in chloroform as solvent system), yielding a yellow powder (0.265 g, 62% yield). MS (ESI+): m/z = 597.7, LC retention time: 2.94 min. Rf = 0.45 (10% MeOH: DCM). Example 112 Synthesis of 4-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-ylamino1-N-isopropyl-N-(2-pyrroIidin-l-yl-ethyl - benzenesulfonamide HCI salt
Figure imgf000143_0001
(CXVII)
[0224] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (0.158 g, 0.527 mmol, 1 equiv) was diluted with 10 mL DCM and chilled to 0°C using an ice bath. A 1.0 M solution of BBr3 in DCM (3.16 mL, 3.16 mmol, 6.0 equiv) was then added in one portion resulting in a dark reaction mixture. The reaction was allowed to come to ambient temperature and stirred for 3.5 hours, then quenched by carefully pouring onto a saturated solution of sodium bicarbonate, followed by sonication for 3-5 minutes. Additional DCM was added and this mixture was washed with sodium bicarbonate followed by brine. The organic phase was separated, dried over sodium sulfate (Na2SO4), filtered and evaporated to dryness. The resulting solids were then diluted with minimum amount of ethyl acetate and precipitated out with hexanes. The product was filtered off and dried yielding a yellow solid (0.110 g, 73 % yield). MS (ESI+): m/z = 582.0, LC retention time: 2.57 min. 1H NMR (DMSO-d6): δ 0.97 (d, J=6.7 Hz, 6H), 1.87 (m, 2H), 2.02 (m, 2H), 2.71 (s, 3H), 3.04 (m, 2H), 3.29-3.37 (m, 2H), 3.49(m, 2H), 3.61 (m, 2H), 4.00 (m, IH), 6.89 (dd, J=8.7 Hz, J=2.8 Hz, IH), 6.96 (d, J=2.8 Hz, IH), 7.40 (d, J=8.7 Hz, IH), 7.91-7.94 (m,' 3H), 8.21 (d, J=1.5 Hz, IH), 8.26 (d, J=8.9 Hz, 2H), 10.0 (s, IH), 11.17 (bs, IH) 11.47 (s, IH). Example 113 Synthesis of 4-(4-bromo-benzenesulfonyl -piperidine-l-carboxylic acid benzyl ester
Figure imgf000144_0001
(cxvm)
[0225] 4-(4-bromo-phenylsulfanyl)-piperidine-l -carboxylic acid benzyl ester (1.2 g, 2.96 mmol, 1 equiv) and sodium perborate tetrahydrate (NaBO -4H2θ) (1.36 g, 8.87 mmol, 3 equiv) were heated to 55°C in HO Ac and stirred for 18h. The reaction was cooled to room temperature and poured onto water. The aqueous phase was extracted with EtOAc (3 x 100 mL). The organic phases were combined and washed carefully with saturated sodium bicarbonate solution (CAUTION: copious gas evolution), dried over sodium sulfate, filtered and evaporated to yield a white solid (1.2 g, 93% yield). Rf = 0.16 (20% EtOAc/hexanes).
Example 114 Synthesis of 4-(4-bromo-phenylsnlfanyl)-piperidine-l-carboxylic acid benzyl ester
Figure imgf000144_0002
(CXIX)
[0226] 4-bromo-benzenethiol (0.634 g, 3.36 mmol, 1 equiv), 4-bromo-piperidine-l- carboxylic acid benzyl ester (lg, 3.36 mmol, 1 equiv) and cesium carbonate (2.18 g, 6.7 mmol, 2 equiv) were refluxed in acetone for 2 h. The reaction solvents were then removed and the resulting white solids were taken up in EtOAc and washed with water, then with brine. The organic phase was then dried over sodium sulfate, filtered and evaporated to yield clear oil (1.2 g, 88 % yield). Rf = 0.45 (20% EtOAc: Hexanes).
Example 115 Synthesis of 4-{4-[7-(2-chloro-5-methoxy-phenyl -5-methyl- benzori,2,41triazin-3-ylamino]-benzenesulfonyl|-piperidine-l-carboxylic acid benzyl ester
Figure imgf000145_0001
(CXX)
[0227] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.132 g, 0.44 mmol, 1 equiv), 4-(4-bromo- benzenesulfonyl)-piperidine-l-carboxylic acid benzyl ester (0.232 g, 0.529 mmol, 1.2 equiv), Cesium carbonate (0.432 g, 1.33 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9- dimethyl xanthene (0.051 g, 0.088 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.040 g, 0.044 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (12 mL) and outfitted with a reflux condenser. The reaction was heated to reflux for 18 hours, then cooled to room temperature and slowly diluted with water. The resulting precipitate was collected and dried yielding a yellow powder (0.271 g, 93% yield). MS (ESI+): m/z = 658.2, LC retention time: 3.41 min.
Example 116 Synthesis of 4-chloro-3-(5-methyl-3-[4-(piperidine-4-sulfonylV phenylamino]-benzo[l,2,41triazin-7-yl|-phenol HCI salt
Figure imgf000146_0001
(CXXI)
[0228] 4-{4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3- ylamino]-benzenesulfonyl}-piperidine-l-carboxylic acid benzyl ester (0.165 g, 0.251 mmol, 1 equiv) was diluted with 15 mL DCM and chilled to 0°C using an ice bath. A 1.0 M solution of BBr3 in DCM (2.5 mL, 2.5 mmol, 10 equiv) was then added in one portion resulting in a dark reaction mixture. The reaction was allowed to come to ambient temperature and stirred for 3.5 hours, then quenched by carefully pouring onto a saturated solution of sodium bicarbonate followed by sonication for 3-5 minutes. The resulting solids were filtered off and dried. HPLC purification provided desired product as TFA salt in a form of a yellow solid (0.028 g, 22 % yield). MS (ESI+): m/z = 510.0, LC retention time: 2.27 min. 1H NMR (DMSO-d6): δ 1.68-1.77 (m, 2H), 2.02-2.07 (m, 2H), 2.72 (s, 3H), 2.82-2.88 (m, 2H), 3.51-3.58 (m, IH), 6.89 (dd, J=8.8 Hz, J=2.9 Hz, IH), 6.95 (d, J=2.9 Hz, IH), 7.41 (d, J=8.7 Hz, IH), 7.88 (d, J=8.8 Hz, 2H), 7.93 (s, IH), 8.22 (d, J=1.7 Hz, IH), 8.33 (d, J=8.8 Hz, 2H), 10.01 (s, IH), 11.56 (s, IH).
Example 117 Synthesis of l-[2-(4-bromo-phenylsulfanyl)-ethyl]-pyrrolidine
Figure imgf000146_0002
(CXXII)
[0229] 4-bromo-benzenethiol (7.05 g, 37.3 mmol, 1 equiv), l-(2-chloro-ethyl)- pyrrolidine hydrochloride (8.8 g, 52.2 mmol, 1.4 equiv) and cesium carbonate (26.8 g, 82.06 mmol, 2.2 equiv) were refluxed in acetone for 18 h. The reaction solvents were then evaporated, and the resulting residue was diluted with EtOAc and washed with water. The organic phase was then dried over sodium sulfate, filtered and evaporated to yield a pale brown oil (8 g, 75 % yield). Rf = 0.1 (40% EtOAc: Hexanes).
Example 118 Synthesis of [7-(2-chIoro-5-methoxy-phenyI)-5-methyl- benzo[l,2,41triazin-3-yl1-[4-(2-pyrrolidin-l-yl-ethylsulfanyl)-phenvI1-amine
Figure imgf000147_0001
(CXXIII)
[0230] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine (0.625 g, 2.08 mmol, 1 equiv), l-[2-(4-bromo- phenylsulfanyl)-ethyl]-pyrrolidine (0.894 g, 3.12 mmol, 1.5 equiv), cesium carbonate (2.04 g, 6.25 mmol, 3 equiv), 4,5-bis(diphenylphosphino)-9,9-dimethyl xanthene (0.24 g, 0.417 mmol, 0.2 equiv) and tris(dibenzylideneacetone) dipalladium (0.19 g, 0.208 mmol, 0.1 equiv) were combined. The reactants were flushed with argon, diluted with dioxane (12 mL), outfitted with reflux condenser and heated to reflux for 16 hours. The reaction was then filtered and the solvents were removed. The residue was taken up in minimum amount of DCM (ca. 5 mL) and precipitated out with excess hexanes. The resulting precipitate was collected and dried. 0.5 g of this crude product was taken on and purified by ISCO silica gel chromatography to afford product as an orange solid (0.224 g, 22% yield). MS (ESI+): m/z = 506.1, LC retention time: 2.91 min. xample 119 Synthesis of 4-chloro-3-{5-methyl-3-[4-(2-pyrroIidin-l-yl-ethylsulfanyl)- phenylamino]-benzo[l,2,4]triazin-7-yl}-phenol TFA salt
Figure imgf000148_0001
(CXXIN)
[0231] [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-yl]-[4-(2- pyrrolidin-l-yl-ethylsulfanyl)-phenyl]-amine (Q.22 g, 0.435 mmol, 1 equiv) was diluted with 20 mL DCM and chilled to 0°C using an ice bath. A 1.0 M solution of BBr3 in DCM (3.4 mL, 3.5 mmol, 10 equiv) was then added in one portion resulting in a dark reaction mixture. The reaction was allowed to come to ambient temperature and stirred for 3.5 hours, then quenched by carefully pouring onto a saturated solution of sodium bicarbonate followed by sonication for 5 minutes. The resulting solids were filtered off and dried. HPLC purification provided desired product as TFA salt in a form of an orange solid (0.088 g, 41 % yield). MS (ESI+): m/z = 492.1, LC retention time: 2.52 min. 1H ΝMR (DMSO-de): δ 1.83-1.86 (m, 2H), 1.98-2.01 (m, 2H), 2.67 (s, 3H), 3.01-3.05 (m, 2H), 3.23-3.26 (m, 2H), 3.31-3.35 (m, 2H), 3.57-3.58 (m, 2H), 6.88 (dd, J=8.8 Hz, J=2.9 Hz, IH), 6.93 (d, J=2.9 Hz, IH), 7.41 (d, J=8.7 Hz, IH), 7.51 (d, J=8.8 Hz, 2H), 7.87 (s, IH), 8.07 (d, J=8.8 Hz, 2H), 8.17 (d, J=1.7 Hz, IH), 9.64 (bs, IH), 9.95 (bs, IH), 11.11 (s, IH).
Example 120 Synthesis of 4-(3-(4-(2-(pyrroIidin-l-yl ethoxy)phenylamino -5- methylbenzo[e]fl,2,41triazin-7-yI)-3,5-dimethylphenol
Figure imgf000149_0001
(CXXN)
[0232] The title compound was synthesized as shown by the reaction scheme (CXXNI).
Figure imgf000149_0002
Figure imgf000149_0003
Figure imgf000149_0004
Pd2(dba)3/BINAP/KOBu'
Figure imgf000149_0005
(CXXVI)
[0233] To a solution of the 7-bromo-5-methylbenzo[l,2,4]triazin-3-yl-amine-l-oxide 1 shown on scheme (CCXXVI) (100 mg, 0.39 mmol) in N,N-dimethylacetamide (6 mL) was added a solution of 4-methoxy-2,6-dimethylphenylboronic acid 2 (210 mg, 1.17 mmol) in EtOH (1 mL), a solution of K2CO3 (90 mg, 0.65 mmol) in H2O (1 mL), tris(dibenzylidenacetone)dipalladium [0] (Pd2 (dba)3, 9 mg, 0.01 mmol), and PPh3 (9 mg, 0.034 mmol). The suspension was heated under reflux for 18 h. The cold reaction mixture was poured into saturated NaHCO3 and CH2CI2 was used to extract the product. The crude product was purified by chromatography (SiO2/Hexane : EtOAc = 1 : 1) to afford 3 as yellow solid. The 3 was reduced to 4 by hydrogenation, as shown by scheme (CCXXVI). To a solution of 4 in CH2C12 was added 1.0 M BBr3 in CH2C12 (1 L, 1 mmol). The mixture was stirred overnight at room temperature. The saturated NaHCO3 was added and the organic layer was separated. The aqueous layer was extracted with CH2C12 (3 x 10 mL). The combined organic solution was dried (MgSO ) to afford 5.
[0234] As further shown by scheme (CCXXVI), to a suspension of 5 in dry PhMe (1 mL) was added l-(2-(4-bromophenoxy)ethyl)pyrrolidine 6 (44.4 mg, 0.16 mmol), rac- 2,2*-bis(diρhenyl-phosphino)-l,r-binaphthyl (BINAP, 8 mg, 0.012 mmol), Pd2 (dba) 3 (4 mg, 0.004 mmol), and KOBut (38 mg, 0.33 mmol). The mixture was heated under reflux for 48 h under argon. The solid was filtered off and washed with PhMe. The product was purified by preparative HPLC (Symmetry Shield-C18, 200 x 40 mm, 7 μm, starting from 30% solvent A (0.1% TFA in H2O) to 100% solvent B (0.1% TFA in MeCN), flow rate 40 ml/min, λ = 282 nm). Fractions containing the products were combined to yield the title product (CXXV) as its TFA salt (1.3 mg, 3%). 1H NMR (CDC13): δ 2.02-2.04 (m, 2H); 2.12-2.17 (m, 2H); 2.93 (s, 3H); 3.01 (s, 6H); 3.59-3.61 (m, 2H); 3.94-4.00 ( , 4H); 4.40 (t, J= 4.9 Hz, 2H); 6.65 (s, IH); 7.30 (s, IH); 7.48 (d, J= 7.9 Hz, 2H); 7.61 (s, IH); 8.06 (s, IH); 8.09 (d, J= 7.9 Hz, IH); 8.92 (br, IH). MS (ESI+): m/z = 470.3.
Example 121 Synthesis of [7-(3,5-dimethyl-isoxazol-4-yl)-5-methyl- benzo[l,2,41triazm-3-yl]-(4-methoxy-phenyl -amine
Figure imgf000151_0001
(CXXVII)
[0235] The reaction scheme (CXXVIII) includes an illustration of the synthesis of the title compound.
Figure imgf000151_0002
Figure imgf000151_0003
Cs2C03/Dioxane/Reflux
Figure imgf000151_0005
Figure imgf000151_0004
(CXXVIII)
[0236] The mixture of 5-methyl-7-(3,5-dimethylisoxazol-4-yl)benzo[e][l,2,4]triazin- 3-amine (compound 7 on scheme (CXXVIII)) (107 mg, 0.42 mmol), sulfamic acid (81.4 mg, 0.84 mmol), and 4-methoxybenzenamine 8 (774 mg, 6.3 mmol) was heated at 180°C overnight. The product was purified by chromatography (SiO / CH2C12) and the title product (48.7 mg, 32%) was obtained as a yellow solid. 1H NMR (CD2C12): δ 2.34 (s, 3H); 2.49 (s, 3H); 2.70 (s, 3H); 3.84 (s, 3H); 6.99 (d, J= 8.9 Hz, 2H); 7.57 (s, IH); 7.83 (d, J= 8.9 Hz, 2H); 8.03 (s, IH); 8.10 (br, IH). MS (ESI+):rn/z = 362.1. Example 122 Synthesis of [7-(3,5-dimethyl-isoxazol-4-yl -5-methyl- benzo[l,2,41triazin-3-yll-{4-f2-(4-methyl-piperazin-l-yl)-ethoxy1-phenyll-amine
Figure imgf000152_0001
(CXXIX)
[0237] The synthesis of the title compound is also illustrated by the reaction scheme (CXXVIII). Compound (CXXVII) obtained as described in Example 121 (48.7 mmg, 0.13 mmol) was demethylated by using BBr3/ CH2CI2 to obtain compound 9 shown by the scheme (CXXVIII) (46 mg, 99%). To a solution of 9 (18.8 mg, 0.054 mmol) in anhydrous 1,4-dioxane (10 mL) was added Cs2CO3 (176 mg, 0.54 mmol), and l-(2- bromoethyl)-4-methylpiperazine 10 shown by the scheme (CXXVIII) (40 mg, 0.11 mmol). The mixture was heated under reflux overnight. The product was purified by preparative HPLC (Symmetry Shield-C18, 200 x 40 mm, 7 μm, starting from 30% solvent A (0.1% TFA in H2O) to 100% solvent B (0.1% TFA in MeCN), flow rate 40 ml/min, λ = 288 nm). Fractions containing the products were combined. The title compound was obtained (0.8 mg, 3%) as its TFA salt. 1H NMR (CD2Cl2):δ 2.24 (s, 3H); 2.39 (s, 3H); 2.61 (s, 3H); 2.73 (s, 3H); 3.17 (t, J= 4.5 Hz, 2H); 3.25-3.40 (m, 8H); 4.20 (t, J= 4.5 Hz, 2H); 6.91 (d, J= 9.0 Hz, 2H); 7.48 (s, IH); 7.75 (d, J= 9.0 Hz, 2H); 7.94 (s, IH). MS (ESI+): m/z = 474.2.
Example 123 Synthesis of 3-(5-methyl-7-(2,6-dimethylphenyl)benzo[eiri.2.41triazin-
3-ylamino)b enzene-sulfonamide
Figure imgf000153_0001
(CXXX)
[0238] The title compound was synthesized as shown by the reaction scheme (CXXXI).
Figure imgf000153_0002
Pd2(dba)3/BINAP/KOBuf
Figure imgf000153_0003
Figure imgf000153_0004
(CXXXI)
[0239] To a suspension of compound 11 shown on scheme (CXXXI) (200 mg, 0.75 mmol) in dry PhMe (5 mL) was added 3-bromobenzenesulfonamide 12 (268 mg, 1.13 mmol) , rac-2,2'-bis(diphenylρhosphino)-l,r-binaphthyl (BINAP, 141 mg, 0.23 mmol), Pd2 (dba)3 (69 mg, 0.075 mmol), and KOBut (170 mg, 1.51 mmol). The mixture was heated under reflux for 20 h. The solid was filtered off and washed with PhMe. The product was purified by preparative HPLC (Symmetry Shield-C18, 200 x 40 mm, 7 μm, starting from 30% solvent A (0.1% TFA in H2O) to 100% solvent B (0.1% TFA in MeCN), flow rate 40 ml/min, λ = 288 nm). Fractions containing the products were combined. The title compound was obtained (10.4 mg, 3%) as its TFA salt. 1H NMR (DMSO- ): δ 2.06 (s, 6H); 2.71 (s, 3H); 7.19 (d, J= 7.3 Hz, 2H); 7.24 (m, IH); 7.35 (br, 2H); 7.53 (d, J= 7.9 Hz, IH); 7.60 (t, J= 7.9 Hz, IH); 7.65 (s, IH); 7.97 (s, IH); 8.05 (d, J= 7.9 Hz, IH); 8.85 (s, IH); 11.24 (s, IH). MS (ESI+): m/z = 420.1. Example 124 Synthesis of 3-(5-methyl-7-(2,6-dimethylphenyl benzo[e][l,2,4]triazin- 3-ylamino -N-(2-pyrrolidin-l-yl)ethyl)benzenesulfonamide hydrochloride
Figure imgf000154_0001
(CXXXII)
[0240] The title compound was synthesized as shown by the reaction scheme (CXXXIII).
Figure imgf000154_0003
Figure imgf000154_0002
Pd2(dba)3/BINAP/KOBuf
Figure imgf000154_0004
(CXXXIII)
[0241] To a solution of 3-bromobenzenesulfonyl chloride 13 shown on scheme (CXXXIII) (930 mg, 3.7 mmol) in anhydrous CH2C12 (10 mL) was added 2-(pyrrolidin- l-yl)ethanamine 14 (840 mg, 7.32 mmol) and Et3N (1.85 g, 18.3 mmol). The mixture was stirred 24 h at room temperature. Saturated NaHCO3 (50 mL) was added and organic layer was separated and washed with brine (3 x 20 mL), dried (Na2SO4). The solvent was removed and product 15 shown on scheme (CXXXIII) was obtained as a colorless oil. [0242] The title compound was prepared by a method that was analogous to that used to synthesize compound (CXXX) (Example 123) using compounds 15 (780 mg, 2.54 mmol) and 11 (447 mg, 1.69 mmol) to give the title compound (260 mg, 31%) as a norange solid. 1H NMR (CD2C12): δ 1.57-163 (m, 4H); 2.10 (s, 6H); 2.29 (br, 4H); 2.50 (t, J= 5.9 Hz, 2H); 2.80 (s, 3H); 3.04 (t, J= 5.6 Hz, 2H); 7.17 (d, J= 7.7 Hz, 2H); 7.21- 7.24 ( , IH); 7.57-7.60 (m, 3H); 8.03 (s, 2H); 8.87 (br, IH); 8.97 (s, IH). MS (ESI+): m/z = 517.4.
Example 125 Synthesis of 3-(5-methyl-7-(2,6-dimethylphenyl)benzo[elfl,2,4]triazin- 3-ylamino)-N-(2-(dimethylamino)ethyl)benzenesnlfonamide hydrochloride
Figure imgf000155_0001
(CXXXIV)
[0243] The title compound was synthesized as shown by the reaction scheme (CXXXV).
Figure imgf000155_0003
Figure imgf000155_0002
1) Pd2(dba)3/BINAP/KOBu'
2) HCI
Figure imgf000155_0004
(CXXXV) [0244] Compoimd 17 shown on scheme (CXXXV) was prepared by a method that was analogous to that used for preparation of compound 15 shown on scheme (CXXXIII), except compounds 16 (700 mg, 7.9 mmol) and 13 (1.2 g, 3.9 mmol) were used to yield the compound 17 (1.2 g, 99%) as a colorless oil. The title compoimd (CXXXrV) was prepared by a method that was analogous to that used to synthesize compound (CXXX) (Example 123), except compounds 17 (840 mg, 2.53 mmol) and 11 (450 mg, 1.69 mmol) were used, as shown by scheme (CXXXV), to give title compound (CXXXIV) (260 mg, 31%) as a orange solid. 1H NMR (CD2C12): δ 2.11 (s, 6H); 2.84 (s, 9H); 3.30 (br, 2H); 3.59 (br, 2H); 7.15-7.18 (m, 3H); 7.21-7.24 (m, IH); 7.45 (d, J= 7.6 Hz, IH); 7.56 (s, IH); 7.84-7.86 (m, 2H); 7.93 (s, IH); 9.06 (s, IH); 9.37 (s, IH); 11.87 (br, IH). MS (ESI+): m/z = 491.3.
Example 126 Synthesis of 2-(5-methvI-7-(2,6-dimethylphenvI)benzofe]fl,2,4]triazin- 3-ylamino)-N-(2-(pyrrolidin-l-yl)ethyl)thiazole-4-carboxamide
Figure imgf000156_0001
(CXXXVI)
[0245] The title compound was synthesized as shown by the reaction scheme (CXXXVII).
Figure imgf000157_0001
(CXXXVII)
[0246] To a solution of 11 shown by scheme (CXXXVII) (646 mg, 2.44 mmol) in dry DMF (20 mL) was added ethyl 2-bromothiazole-4-carboxylate 18 (750 mg, 3.17 mmol) , rac-2,2'-bis(diphenylphosphino)-l,l'-binaρhthyl (BINAP, 304 mg, 0.48 mmol), Pd2 (dba)3 (223 mg, 0.24 mmol), and Cs2CO3 (1.6 g, 4.88 mmol). The mixture was heated under reflux for 20 h. The solid was filtered off and washed with DMF. The filtrate was concentrated. The residue was dissolved in CH2C12 and washed with brine (3 x 100 mL). The crude product ethyl 2-(5-methyl-7-(2,6-dimethylphenyl)benzo[e][l,2,4Jtriazin-3- ylamino)thiazole-4-carboxylate 19 was dissolved in MeOH (10 mL) and THF ( 10 mL), followed by adding solution of LiOH (586 mg, 24.4 mmol) in H2O (10 mL). The mixture was heated at 60°C for 2 h. The solvent was removed in vacuo. The product was purified using chromatograpy (SiO2/CH2Cl2: MeOH: NH3.H2O = 100 : 10: 2.5 and then 100: 100 : 50). 2-(5-methyl-7-(2,6-dimethylphenyl)benzo[e] [1,2,4] triazin-3- ylamino)thiazole-4-carboxylic acid (750 mg, 78%) (compound 20 shown by scheme (CXXXVII)) was obtained as an orange solid.
[0247] The synthesis was continued by dissolving compound 20 (400 mg, 1.02 mmol) in anhydrous CH2C12 (20 mL), followed by adding 2-chloro-4,6-dimethoxy-l,3,5-trizine (CDMT, 198 mg, 1.12 mmol) and 4-methylmophline (NMM, 0.23 mL, 2.04 mmol). The mixture was stirred at room temperature for 0.5 h, followed by adding 2-(pyrrolidin-l- yl)ethanamine 21 (198 mg, 1.12 mmol). The reaction was stirred at room temperature overnight. The solid was filtered off and washed with CH2C12. The filtrate was washed with saturated NaHCO3 (1 x 50 mL) and then brine (2 x 50 mL). The organic solution was dried (Na2SO4).
[0248] The product was purified by preparative HPLC (Symmetry Shield-Cl 8, 200 x 40 mm, 7 μm, starting from 30% solvent A (0.1% TFA in H2O) to 100% solvent B (0.1% TFA in MeCN), flow rate 40 ml/min, λ = 282 nm). Fractions containing the products were combined. The title product (CXXXVI) was obtained (110 mg, 20%) as its TFA salt. 1H NMR (DMSO- ): δ 1.86-1.89 (m, 2H); 2.02-2.05 (m, 2H); 2.05 (s, 6H); 2.80 (s, 3H); 3.05-3.09 (m, 2H); 3.35-3.38 (m, 2H); 3.64-3.67 (m, 4H); 7.20 (d, J= 7.7 Hz, 2H); 7.24-7.27 (m, IH); 7.78 (s, IH); 7.97 (s, IH); 8.10 (s, IH); 8.25 (t, J= 6.1 Hz, IH); 9.42 (br, IH); 12.56 (s, IH). MS (ESI+): m/z = 488.2.
Example 127 Synthesis of 3-(5-methyl-7-(2,6-dimethylphenyl)benzo[e][l,2,41triazin- 3-ylaminoVN-methyl-N-(2-(pyrrolidin-l-yl)ethyl)benzenesuIfonamide hydrochloride
Figure imgf000158_0001
(CXXXVIII)
[0249] The title compound was synthesized as shown by the reaction scheme (CXXXIX).
Figure imgf000159_0001
(CXXXIX)
[0250] To a solution of ethyl formate (10 mL) in anhydrous Et2O was added compound 21 as shown on scheme (CXXXIX) (2 mL). The solution was stirred at room temperature for 2 h. The solvent was removed in vacuo, followed by adding 1.0 M LiAlIL in THF (5 mL). The mixture was heated under reflux overnight and cooled. Water was then added slowly. The solid was filtered off and washed with THF. The filtrate was concentrated and remained aqueous, and was extracted with Et2O (3 x 20 mL) and dried (Na2SO4). The solvent was removed and the residue was dissolved in CH2C12 (10 mL), followed by adding 4-bromobenzenesulfonyl chloride 24 (1.2 g, 4.72 mmol) and Et3N (3 mL). The mixture was stirred at room temperature for 3 hours, then saturated NaHCO3 (100 mL) was added. The organic layer was separated and the aqueous was extracted with CH2CI2 (2 x 10 mL). Combined organic solution was dried (Na2SO4). The solvent was removed in vacuo and afforded compound 25 shown on scheme (CXXXIX) as a yellow oil.
[0251] The title compound (CXXXNIII) was prepared by using a method that was analogous to that used to synthesize compound (CXXX) (Example 123) except compounds 25 (600 mg, 1.72 mmol) and 11 (351 mg, 1.33 mmol) were used to give title compound (CXXXNIII) (276 mg, 37%) as its HCI salt. 1H ΝMR (DMSO- ): δ 1.88-1.91 (m, 2H); 2.02-2.05 (m, 2H); 2.06 (s, 6H); 2.72 (s, 3H); 2.74 (s, 3H); 3.05-3.09 (m, 2H); 3.30-3.38 (m, 4H); 3.58-3.61 (m, 2H); 7.19 (d, J= 7.3 Hz, 2H); 7.23-7.26 (m, IH); 7.70 (s, IH); 7.86 (d, J= 8.9 Hz, 2H); 8.01 (s, IH); 8.31 (d, J= 8.9 Hz, 2H); 10.18 (br, IH); 11.47 (s, IH). MS (ESI+): m/z = 531.3.
Example 128 Synthesis of [7-f2,6-dimethyI-phenyl)-5-nitro-l-oxy- benzofl,2,4]triazm-3-ylH4-(2-pyrrolidin-l-yl-ethoxy)-phenyIl-amme
Figure imgf000160_0001
(CXL)
[0252] The title compound was synthesized as shown by the reaction scheme (CXLI). The intermediate compounds 27 and 28 shown on scheme (CXLI) had the following spectral characteristics:
[0253] Compound 27: 1H NMR (DMSO- ): δ 7.30 (d, J= 8.8 Hz, IH); 7.97 (dd, J= 8.8 Hz, J= 2.0 Hz, IH); 8.25 (d, J= 2.0 Hz, IH). MS (ESI+): m/z = 241. Compoimd 28: 1H NMR (DMSO- ):δ 8.22 (d, J= 2.3 Hz, IH); 8.27 (d, J= 2.3 Hz, IH). MS (ESI+): m/z = 286.
Figure imgf000161_0001
26 27
Figure imgf000161_0002
(CXLI)
[0254] To a solution of compound 28 (500 mg, 1.75 mmol) in 1,2-dimethoxyethane (DME, 20 mL) was added solution of compound 29 (526 mg, 3.5 mmol) in EtOH (2 mL), solution of Na2CO3 (750 mg, 7.0 mmol) in H2O (2mL), and Pd(PPh3)4 (202 mg, 0.2 mmol), as shown by scheme (CXLI). The mixture was heated under reflux overnight. The solid was filtered off and washed with EtOAc. The filtrate was washed with brine (1 x 100 mL). The organic solution was separated and dried (Na2SO4). The product was purified by chromatograph ((SiO2/Hexane : EtOAc = 1 : 1) to yield compound 30 (450 mg, 82%>) as a yellow solid. [0255] To a solution of 30 (450 mg, 1.45 mmol) in 1,4-dioxane (20 mL) were added compound 6 (586 mg, 2.17 mmol), Cs2CO3 (1.90 g, 5.8 mmol), Pd2(dba)3 (133 mg, 0.14 mmol), and 4,5-bis(diphenylphosphino)-9,9-dimethyxanthene (Xant Phos, 251 mg, 0.43 mmol). The mixture was heated under reflux for 4 h under Ar. The solid was filtered off and washed with brine (1 x 100 mL). The organic solution was separated and dried (Na2SO4). The solvent was removed until 5 mL and hexane (50 mL) was added, the solid was collected by filtration. The crude product was purified by chromatograph (SiO2/ CH2C12; then CH2C12 : MeOH : NH3.H2O = 100 : 10 : 2.5) and afforded the title compound (CXL) (124 mg, 17%). 1H NMR (CD2C12): δ 1.82 (br, 4H); 2.08 (s, 6H); 2.69 (br, 4H); 2.95 (br, 2H); 4.16 (t, J= 5.5 Hz, 2H); 6.99 (d, J= 7.1 Hz, 2H); 7.17 (d, J= 7.5 Hz, 2H); 7.24-7.27 (m, IH); 7.52 (br, IH); 7.72 (d, J= 7.1 Hz, 2H); 8.13 (d, J= 1.9 Hz, IH); 8.35 (d, J= 1.9 Hz, IH). MS (ESI+): m/z = 501.3.
Example 129 Synthesis of 7-(2,6-dimethyI-phenyI)-N3-[4-(2-pyrrolidin-l-yl-ethoxy)- phenvH-benzo[l,2,4]triazine-3,5-diamine
Figure imgf000162_0001
o
(CXLII)
[0256] Compound (CXL) synthesized as described in Example 128 was hydrogenated (scheme (CXLI)) to afford the title compound (CXLII) (yield, 73%). 1H NMR (CD2C12): δ 1.84 (br, 4H); 2.10 (s, 6H); 2.75 (br, 4H); 2.99 (br, 2H); 4.16 (t, J= 4.25 Hz, 2H); 4.78 (br, 2H); 6.80 (d, J= 1.6 Hz, IH); 6.99 (d, J= 6.9 Hz, 2H); 7.13 (d, J= 7.6 Hz, 2H); 7.17-7.20 (m, IH); 7.42 (d, J= 1.6 Hz, IH); 7.71 (d, J= 6.9 Hz, 2H); 7.96 (br, IH). MS (ESI+): m/z = 455.4. Example 130 Synthesis of N-(3-(4-(2-(pyrrolidm-l-yl)ethoxy)phenylaminoV7-(2.6- dimethylphenyl)benzo[e1-[l,2,4]triazin-5-yl)acetamide hydrochloride
Figure imgf000163_0001
(CXLIII)
[0257] To a solution of compound (CXLII) obtained as described in Example 129 (122 mg, 0.27 mmol) in anhydrous CH2CI2 (5 mL) was added acetyl chloride (25 mg, 0.32 mmol). The solution was stirred at RT for 0.5 h and solvent was removed in vacuo. The residue was dissolved in CH2CI2 (2 mL) and Et2O was added to precipate product. The product was collected by filtration and dried. The title compound (CXLIII) (92 mg, 64%) was obtained as its HCI salt. 1H NMR (CD2C12): δ 2.06-2.12 (m, 2H); 2.10 (s, 6H); 2.18-2.21 (m, 2H); 2.28 (s, 3H); 2.97-3.02 (m, 2H); 3.48-3.51 (m, 2H); 3.83-3.85 (m, 2H); 4.57 (t, J= 4.8 Hz, 2H); 7.06 (d, J= 6.8 Hz, 2H); 7.14 (d, J= 7.4 Hz, 2H); 7.19-7.22 (m, IH); 7.73 (d, J= 6.8 Hz, 2H); 7.77 (d, J= 1.8 Hz, IH); 8.48 (s, IH); 8.57 (br, IH); 8.94 (s, IH); 12.4 (br, IH). MS (ESI+): m/z = 497.4.
Example 131 Synthesis of N-(3-(4-(2-(pyrrolidin-l-yl)ethoxytohenylaminoV7-(2,6- dimethylphenyl)benzofe]-[l,2,41triazin-5-yl)methylsulfonyl amide hydrochloride
Figure imgf000163_0002
(CXLIV) [0258] The title compound was synthesized as shown by the reaction scheme (CXLV).
Figure imgf000164_0001
(CXLV)
[0259] As shown by scheme (CXLV), to a solution of compound (CXLII) obtained as described in Example 129 (40 mg, 0.09 mmol) in anhydrous PhMe (5 mL) was added methylsulfonyl chloride (13 mg, 0.11 mmol). The solution was heated under reflux for 18 h and solvent was removed in vacuo. The residue was dissolved in CH2C1 (2 mL) and Et2O was added to precipate product. The product was purified by HPLC. The title compound (CXLIV) (30 mg, 60%) was obtained as its HCI salt. 1H NMR (CD2C12): δ 2.00-2.02 (m, 2H); 2.12 (s, 6H); 2.08-2.12 (m, 2H); 3.14 (s, 3H); 3.18-3.21 (m, 2H); 3.61-3.64 (m, 2H); 3.73-3.76 (m, 2H); 4.62 (t, J= 5.0 Hz, 2H); 7.13 (d, J= 9.1 Hz, 2H); 7.19 (d, J= 7.1 Hz, 2H); 7.20-7.25 (m, IH); 7.69 (d, J= 1.8 Hz, IH); 7.88 (d, J= 1.8 Hz, IH); 7.95 (d, J= 9.1 Hz, IH); 8.49 (s, IH); 9.92 (s, IH); 12.4 (br, IH). MS (ESI+): m/z = 533.4.
Example 132 Synthesis of j4-[7-(2-chloro-5-hvdroxy-phenyl)-6-methyl- benzofl,2,4]triazin-3-ylamino]-phenyl}-(4-methyl-piperazin-l-yl)-methanone HCl salt
Figure imgf000164_0002
(CXLVI) [0260] The title compound was synthesized as shown by the reaction scheme (CXLVH).
Figure imgf000165_0001
(CXLVII)
[0261] To a solution of a methoxylated product in MeSO3H (50 mg, 0.083 mmol) in anhydrous CH2C12 (2 mL) was added 1.0 M BBr3 (0.2 mL, 0.2 mmol) in CH2C12, as shown by scheme (CXL VII). The mixture was stirred at RT overnight. Sat. NaHCO (10 mL) was added. The solid was collected and washed with H2O and CHC13. The crude product was further purified by HPLC and transferred its HCI salt. The title product (CXLVI) (30 mg, 69%) was obtained as orange solid. 1H NMR (DMSO- ): δ 2.28 (s, 3H); 2.72 (s, 3H); 2.79 (s, 3H); 2.75-2.80 (m, 4H); 3.06-3.11 (m, 4H); 6.81 (d, J= 2.9 Hz, IH); 6.81 (dd, J= 8.8 Hz, J= 2.9 Hz, IH); 7.40 (d, J= 8.9 Hz, IH); 7.52 (d, J= 6.9 Hz, IH); 7.76 (s, IH); 8.07 (s, IH); 8.09 (d, J= 6.9 Hz, 2H); 9.98 (br, IH); 10.78 (br, IH); 11.13 (s, IH). MS (ESI+): m/z = 489.2.
Example 133 Synthesis of 7-(2-chloro-5-methoxy-phenyl -5-methyl- benzo [1 ,2,41 triazin-3-yl] - [4-(2-pyrrolidin-l- yl-ethoxy)-phenyl] -amine
Figure imgf000165_0002
(CXLVIII)
[0262] The title compound was synthesized as shown by the reaction scheme (CXLLX).
Figure imgf000166_0001
(CXLIX)
[0263] Compound 33 shown by scheme (CXLIX) was prepared by a method that was analogous to that used to make compound 30 as shown on scheme (CXLI), except 2- chloro-5-methoxyphenylboronic acid (compound 31) (510 mg, 2.73 mmol) and 7-bromo- 5-methylbenzo[e][l,2,4]triazin-3-amine (compound 32) (503 mg, 2.10 mmol) were used to produce compound 33 (500 mg, 80%) as a yellow solid. The title compound (CXLVIII) was then prepared by a method that was analogous to that used to make compound (CXL) as described in Example 128, except compounds 33 (500 mg, 1.66 mmol) and 6 (674 mg, 2.49 mmol) were used as shown by scheme (CXLIX) to afford the title compound (CXLVIII) (440 mg, 54%) as a yellow solid.
Example 134 Synthesis of 3-(3-(4-(2-(pyrrolidin-l-vDethoxy)phenylamino)-5- methylbenzo[e][l,2,4]triazin-7-yl)-4-chlorophenol hydrochloride
Figure imgf000166_0002
(CL) [0264] The title compound (CL) was prepared by a method that was analogous to that used to make compound (CXLVI) as described in Example 132, except compound (CXLVIII) as described in Example 133 (440 mg, 0.90 mmol) was use to produce the title compound (CL) (387 mg, 84%) as HCI salt and orange solid. 1H NMR (DMSO-/): δ 1.88-1.92 (m, 2H); 1.97-2.05 (m, 2H); 2.64 (s, 3H); 3.09-3.14 (m, 2H); 3.57-3.61 (m, 4H); 4.36 (t, J= 4.9 Hz, 2H); 6.87 (dd, J= 8.8 Hz, J= 2.9 Hz, IH); 6.94 (d, J= 2.9 Hz, IH); 7.09 (d, J= 9.0 Hz, 2H); 7.39 (d, J= 8.8 Hz, IH); 7.83 (d, J= 1.6 Hz, IH); 7.98 (d, J= 9.0 Hz, 2H); 8.12 (d, J= 1.6 FIz, IH); 9.97 (s, IH); 10.55 (br, IH); 10.87 (s, IH). MS (ESI+): m/z = 476.3.
Example 135 Synthesis of 3-(7-(2-chloro-5-hydroxyphenyl)-5- methylbenzo[e1[l,2,4]triazin-3-ylamino)-N-(2-(pyrrolidin-l-yl)ethyl)benzamide hydrochloride
Figure imgf000167_0001
(CLI)
[0265] The title compound was synthesized as shown by the reaction scheme (CLII).
Figure imgf000168_0001
Figure imgf000168_0002
(CLII)
[0266] Compoimd 36 shown on scheme (CLII) was prepared by a method that was analogous to that used to make compound 20 shown on scheme (CXXXVII), except using compounds 34 (ethyl 3-bromobenzoate) (84 mg, 0.37 mmol) and 33 (74 mg, 0.24 mmol) were used to afford compound 36 (72 mg, 71%) as a yellow solid. The title compound (CLI) was prepared by a method that was analogous to that used to make compound (CXXXVI) as described in Example 126, except using compounds 36 (35 mg, 0.083 mmol) and 37 (N^N^dimethylethane-l^-diamine) (15 mg, 0.17 mmol) were used to afford the title compound (CLI) (6.3 mg, 15%) as its HCI solid and yellow solid. 1H NMR (DMSO-/): δ 2.71 (s, 3H); 2.85 (s, 6H); 3.28 (t, J= 5.3 Hz, 2H); 3.55-3.76 (m, 2H); 6.88 (dd, J= 8.7 Hz, J= 2.9 Hz, IH); 6.95 (d, J= 2.9 Hz, IH); 7.40 (d, J= 8.8 Hz, IH); 7.52 (t, J= 7.9 Hz, IH); 7.61 (d, J= 7.9 Hz, IH); 7.87 (d, J= 1.7 Hz, IH); 8.10 (dd, J= 8.1 Hz, J= 1.8 Hz, IH); 8.17 (d, J= 1.8 Hz, IH); 8.74 (s, IH); 8.77 (t, J= 5.6 Hz, 1H);9.30 (br, IH); 9.97 (s, IH); 11.14 (s, IH). MS (ESI+): m/z = 477.1. Example 136 Synthesis of 3-(3-(3-(dimethylamino)phenyIaminoV5- methylbenzo[e1[l,2,41triazin-7-vl -4-chlorophenol hydrochloride
Figure imgf000169_0001
(CLIII)
[0267] The title compoimd was synthesized as shown by the reaction scheme (CLIN).
Figure imgf000169_0002
Figure imgf000169_0003
(CLIN)
[0268] As shown by scheme (CLIV), to a solution of compound 33 (520 mg, 1.73 mmol) in anhydrous dioxane (20 mL) were added 3-bromo-Ν,Ν-dimethylbenzenamine (compound 38) (692 mg, 3.46 mmol), Cs2CO3 (2.3 g, 6.92 mmol), Pd2 (dba) 3 (158 mg, 0.17 mmol), and xantphos (300 mg, 0.52 mmol). The resulted mixture was heated under reflux for 2 h. The solid was filtered off and washed with EtOAc. The filtrate was washed with brine (1 x 100 mL). The organic layer was separated and dried (Na2SO4). The crude product was purified by chromatography (SiO2/ CH Cl2j then 5% MeOH in CH2C12) and afforded compound 39 as shown by scheme (CLIV). Compound 39 was then demethylated by using BBr3/CH2Cl2 to afford the title product (CLIII) (80 mg, 10%) as its HCI salt and yellow solid. 1H NMR (DMSO-/): δ 2.70 (s, 3H); 3.07 (s, 6H); 6.88 (dd, J= 8.7 Hz, J= 2.9 Hz, IH); 6.94 (d, J= 2.9 Hz, IH); 7.36 (br, 2H); 7.40 (d, J= 8.8 Hz, IH); 7.48 (br, IH); 7.86 (s, IH); 8.10 (br, IH); 8.16 (d, J= 1.7 Hz, IH); 9.98 (br, IH); 11.02 (s, IH). MS (ESI+): m/z = 406.1.
Example 137 Synthesis of 3-(3-(6-methoxypyridin-3-ylamino -5- methylb enzo [el [1 ,2,4] triazin-7-yr, -4-chlorophenol hydrochloride
Figure imgf000170_0001
(CLV)
[0269] The title compound was synthesized as shown by the reaction scheme (CLVI).
Figure imgf000170_0002
Figure imgf000170_0003
(CLVI)
[0270] The title compoimd was prepared by a method that was analogous to that used to make compound (CLIII), as described in Example 136, except using compounds 33 (37 mg, 0.13 mmol) and 40 (5-bromo-2-methoxypyridine) (36.4 mg, 0.20 mmol) shown on scheme (CLVI) were used to afford the title compound (CLV) (13 mg, 23%) as its HCI salt in a form of a red solid. 1H NMR (DMSO-/): δ 64 (s, 3H); 3.87 (s, 3H); 6.88 (dd, J= 8.7 Hz, J= 2.9 Hz, IH); 6.92 (d, J= 8.9 Hz, IH); 6.95 (d, J= 2.9 Hz, IH); 7.39 (d, J= 8.8 Hz, IH); 7.84 (dd, J= 1.9 Hz, J= 1.0 Hz, IH); 8.14 (d, J= 1.7 Hz, IH); 8.24 (dd, J= 8.9 Hz, J= 2.9 Hz, IH); 8.88 (d, J= 2.6 Hz, IH); 10.00 (br, IH); 10.92 (s, IH). MS (ESI+): m/z = 394.0.
Example 138 Synthesis of 3-(3-(2-(pyrrolidin-l-yl)ethylamino -5- methyIbenzo[e]fl,2,4]triazin-7-yl)-4-chlorophenol hydrochloride
Figure imgf000171_0001
(CLVII)
[0271] The title compound was synthesized as shown by the reaction scheme (CLVπi).
Figure imgf000171_0002
(CLVIII)
[0272] As shown by scheme (CLVIII), to a solution of compound 33 (320 mg, 1.06 mmol) in anhydrous DMF (20 mL) were added K2CO3 (1.2 g, 8.51 mmol), and l-(2- chloroethyl)pyrrolidine 42 (217 mg, 1.27 mmol). The mixture was heated at 150 °C for 72 h. The solid was filtered off and washed with CH2C12. The filtrate was concentrated in vacuo. The residue was dissolved in CH C12 and washed with brine (2 x 50 mL). The organic layer was separated and dried (Na2SO4). The solvent was removed and the residue was demethylated by using BBr3/CH2Cl2. The product was purified by preparative HPLC. The title compound (CLVII) (90 mg, 21%) was obtained as HCI salt in a form of a yellow solid. 1H NMR (DMSO-/): δ 1.88-1.90 (m, 2H); 1.97-2.02 (m, 2H); 2.58 (s, 3H); 3.05-3.10 (m, 2H); 3.44-3.48 (m, 4H); 6.86 (dd, J= 8.7 Hz, J= 2.9 Hz, IH); 6.94 (d, J= 2.9 Hz, IH); 7.37 (d, J= 8.6 Hz, IH); 7.76 (dd, J= 1.8 Hz, J= 1.0 Hz, IH); 8.05 (d, J= 1.8 Hz, IH); 10.05 (br, IH); 10.78 (br, IH). MS (ESI+): m/z = 384.1.
Example 139 Synthesis of 3-(5-(2-(pyrrolidin-l-yl)ethoχy)-3-(pyridin-3- yIamino)benzo [el [1 ,2,41 triazin-7-yl)-4-chlorophenol dihydrochloride
Figure imgf000172_0001
(CLIX)
[0273] The title compound was synthesized as shown by the reaction scheme (CLX).
o
-N02 ' N°2 Br2/Dioxane Br NH 2 1) NH2CN/HCI/110°C Br γ
OH * OH 22)) 3: 0% NaOH/110°C <!,H
44 45 46
Raney Ni/H2
Figure imgf000173_0001
Figure imgf000173_0002
(CLX)
[0274] As shown by scheme (CLX), to a solution of 2-amino-3-nitrophenol 44 (10 g, 65 mmol) in anhydrous 1,4-dioxane (1 L) was added bromine (12.5 g, 78 mmol). The mixture was stirred at room temperature overnight. The solid was collected by filtration, washed with dioxane and dried. 2-amino-5-bromo-3-nitrophenol 45 (18 g, 90%) was obtained as its HBr salt in a form of a yellow solid. 1H NMR (DMSO-/): δ 6.28 (br, 2H); 6.99 (d, J= 2.2 Hz, IH); 7.57 (d, J= 2.1 Hz, IH). MS (ESI+): m/z = 234. Compoimd 45 (6 g, 19.1 mmol) was converted into compound 46 by using a method that was analogous to that used to synthesize compound 27, as shown on scheme (CXLI). Hydrogenation of compound 46 affords compound 47 (3.1 g, 67%) as a red solid. [0275] Compound 48 was then prepared by using a method that was analogous to that used to synthesize compound 33, as described in Example 133 (scheme (CXLIX)) except compounds 47 (870 mg, 3.6 m ol) and 31 (740 mg, 4.0 mmol) were used to afford compound 48 (108 mg, 10%) as yellow solid. 1H NMR (DMSO-/): δ 7.02 (dd, J= 8.9 Hz, J= 3.1 Hz, IH); 7.07 (d, J= 3.1 Hz, IH); 7.16 (d, J= 1.9 Hz, IH); 7.49 (d, J= 8.9 Hz, IH); 7.60 (br, 2H); 7.69 (d, J= 1.9 Hz, IH). MS (ESI+): m/z = 303.7.
[0276] To a solution of compound 48 (47 mg, 0.15) in Me2CO (20 mL) were then added Cs2CO3 (202 mg, 0.62 mmol), l-(2-chloroethyl)pyrrolidine 43 (26.4 mg, 0.15 mmol), and Nai (15 mg, 0.1 mmol). The mixture was heated under reflux overnight. The solid was filtered off and washed with Me2CO. The filtrate was concentrated in vacuo. The residue was dissolved in CH C12 and washed with brine (2 x 50 mL). The organic layer was separated and dried (Na2SO4). The solvent was removed in vacuo and compound 49 was obtained as a yellow solid. Compound 51 was then prepared by using a method that was analogous to that used to synthesize compound 39 (scheme (CLIV), Example 136), except compound s 49 (62 mg, 0.15 mmol) and 50 (3-bromopyridine) (36.7 mg, 0.23 mmol) were used to afford compound 51 as a yellow solid.
[0277] Compound 51 was demethylated as shown by scheme (CLX), by using BBr3/CH2Ci2 to afford the title compound (CLIX) (60 mg, 56%) as its HCI salt in a form of a yellow solid. 1H NMR (DMSO-/): δ 1.82-1.84 (m, 2H); 1.99-2.01 (m, 2H); 3.36- 3.40 (m, 2H); 3.69-3.70 (m, 2H); 3.90-3.92 (m, 2H); 4.68(t, J= 4.2 Hz, 2H); 6.92 (dd, J= 8.6 Hz, J= 2.9 Hz, IH); 7.00 (d, J= 2.9 Hz, IH); 7.43 (d, J= 8.8 Hz, 2H); 7.58 (d, J= 1.5 Hz, IH); 8.01 (br, IH); 8.06 (d, J= 1.5 Hz, IH); 8.62 (d, J= 4.9 Hz, IH); 8.69 (d, J= 8.8 Hz, IH); 9.77 (s, IH); 10.06 (br, IH); 10.79 (br, IH); 11.90 (s, IH). MS (ESI+): m/z = 463.1. Example 140 Synthesis of 7-(lH-indol-4-yl)-5-methyl-benzo[l,2,4]triazin-3-yl-amine
Figure imgf000175_0001
(CLXI)
[0278] The title compound was synthesized as shown by the reaction scheme (CLXII).
Figure imgf000175_0002
Figure imgf000175_0003
(CLXII)
[0279] As shown by scheme (CLXII), the solution of 4-bromo-lH-indole 52 (1.56 g, 7.96 mmol) in anhydrous Et2O (20 mL) was cooled in ice-H O, followed by adding NaH (0.22 g, 9.6 mmol). The suspension was stirred at OoC for 30 min. The flask was further cooled in dry ice-acetone and 2.5 M BuLi in hexane (8 mL, 20 mmol) was dropped. After stirring for 30 min, the neat B(OMe)3 was dropped and the mixture was stirred at room temperature overnight. The mixture was put into 1 M H3PO4 in portions and stirred for 30 min. The organic layer was separated and the aqueous layer was extracted with Et2O (2 x 20 mL). The combined organic layer was washed with 1 M NaOH (3 x 50 mL). The combined aqueous was actified with 1 M H3PO4 (pH < 2) and extracted with Et2O (3 x 20 mL). The combined organic solution was dried (Na2SO4). The solvent was removed and compound 53 (1.16 g, 91%) was obtained, as demonstrated by scheme (CLXII), as a wax solid.
[0280] The title compound (CLXI) was prepared by using a method that was analogous to that used to synthesize compound 33, as described in Example 133 (scheme (CXLIX)), except compounds 53 (1.16 g, 7.2 mmol) and 31 (also described in Example 133 (scheme (CXLIX)) (1.15 g, 4.8 mmol) were used to afford the title compoimd (CLXI) (220 mg, 17%) as a yellow solid. 1H NMR (DMSO-/): δ 2.59 (s, 3H); 6.64 (t, J = 1.1 Hz, IH); 7.22-7.25 (m, 2H); 7.46-7.48 (m, 2H); 7.64 (br, 2H); 8.02 (d, J= 1.9 Hz, IH); 8.24 (d, J= 1.9 Hz, IH); 11.35 (s, IH). MS (ESI+): m/z = 276.2.
Example 141 Synthesis of 4-(7-(lH-indol-4-yl)-5-methyIbenzo[e][l,2,4]triazin-3- ylamino)-N-(2-(pyrrolidin-l-vDethyl) benzenesttlfonamide hydrochloride
Figure imgf000176_0001
(CLXIII)
[0281] The title compound was prepared was prepared by using a method that was analogous to that used to synthesize compound (CXL), as described in Example 128, except compound (CLXI) (described in Example 140) (37 mg, 0.13 mmol) and compound 54 (shown on scheme (CLXII)) (53 mg, 0.16 mmol) were used to afford the title compound (CLXIII) (5.7 mg, 8%) as its HCI salt and orange solid. 1H NMR (DMSO-/): δ 1.85-1.88 (m, 2H); 1.98-2.01 (m, 2H); 2.78 (s, 3H); 2.98-3.04 (m, 2H); 3.08-3.12 (m, 2H); 3.22-3.26 (m, 2H); 3.53-3.56 (m, 2H); 6.69 (t, J= 1.9 Hz, IH); 7.25- 7.31 (m, 2H); 7.51-7.52 (m, 2H); 7.88 (d, J= 9.0 Hz, 2H); 8.23 (s, IH); 8.27 (d, J= 9.0 Hz, IH); 8.43 (d, J= 1.8 Hz, IH); 10.04 (br, IH); 11.40 (s, IH); 11.42 (s, IH). MS (ESI+): m/z = 528.4.
Example 142 Synthesis of 4-(7-(2-bromo-5-hydroxyphenyl)-5- methylbenzo[e][l,2,41triazin-3-ylaminoVN-(2-(pyrrolidin-l-yl)ethyl) benzenesnlfonamide hydrochloride
Figure imgf000177_0001
(CLXIV)
[0282] The title compoimd was synthesized as shown by the reaction scheme (CLXV).
Figure imgf000177_0002
Figure imgf000177_0003
(CLXV) [0283] The intermediate compounds 57 and 58 shown on scheme (CLXV) were prepared as follows. Compoimd 57 was prepared by using a method that was analogous to that used to synthesize compound 30 shown on scheme (CXLI) and described in Example 128, except 2-bromo-5-methoxyphenylboronic acid (compound 56) (233 mg, 1.1 mmol) and 7-bromo-5-methylbenzo[e][l,2,4]triazin-3-amine (compound 32) (240 mg, 1.0 mmol) were used to afford compound 57 (300 mg, 87%) as a yellow solid. Compound 58 was prepared by using a method that was analogous to that used to synthesize compound (CXL), as described in Example 128, except compounds 57 (150 mg, 0.43 mmol) and 54 (174 mg, 0.52 mmol) were used, as shown on scheme (CLXV) to afford compound 58 as a yellow solid.
[0284] The title compound was prepared by using a method that was analogous to that used to synthesize compound (CXL VI), as described in Example 132, except compoimd 58 (0.90 mmol) was used to afford the title compound (CLXIN) (51 mg, 20% in two steps) as HCI salt and orange solid. 1H ΝMR (DMSO-/): δ 1.83-1.86 (m, 2H); 1.95-2.05 (m, 2H); 2.71 (s, 3H); 2.95-3.01 (m, 2H); 3.12-3.16 (m, 2H); 3.21-3.24 (m, 2H); 3.48- 3.53 (m, 2H); 6.84 (dd, J= 8.8 Hz, J= 2.9 Hz, IH); 6.97 (d, J= 2.9 Hz, IH); 7.55 (d, J= 8.8 Hz, IH); 7.88 (d, J= 8.8 Hz, 2H); 7.87 (d, J= 1.8 Hz, IH); 8.04 (t, J= 6.0 Hz, IH); 8.17 (d, J= 1.8 Hz, IH); 8.25 (d, J= 8.8 Hz, 2H); 10.12 (s, IH); 10.85 (br, IH); 11.44 (s, IH). MS (ESI+): m/z = 583.0.
Example 143 Synthesis of 4-(7-(2-chloro-6-hvdroxyphenyl)-5- methylbenzo[e][l,2,41triazin-3-ylamino)-Ν-(2-(pyrrolidin-l- vDethvDbenzensuIfonamide hydrochloride
Figure imgf000178_0001
(CLXNI) [0285] The title compound was synthesized as shown by the reaction scheme (CLXNII).
Figure imgf000179_0001
Figure imgf000179_0002
Dioxane/Pd2(dba)3/Xant Phos Cs2C03/Reflux/4 h
Figure imgf000179_0003
Figure imgf000179_0004
(CLXNII)
[0286] As shown by scheme (CLXNII), compound 60 was prepared by using a method that was analogous to that used to synthesize compound 30 shown on scheme (CXLI) and described in Example 128, except 2-chloro-6-methoxyphenylboronic acid 59 (1 g, 4.18 mmol) and 7-bromo-5-methylbenzo[e][l,2,4]triazin-3 -amine 32 (1.2 g, 6.28 mmol) were used to give compound 60 (7-(2-chloro-6-methoxyphenyl)-5- methylbenzo[e][l,2,4]triazin-3-amine) (1.12 g, 90%) as a yellow solid. Compound 61 was prepared was prepared by using a method that was analogous to that used to synthesize compound (CXL), as described in Example 128, except compounds 60 (390 mg, 1.30 mmol) and 54 (518 mg, 1.56 mmol) were used to afford compound 61 (4-(7-(2- chloro-6-methoxyphenyl)-5-methylbenzo[e] [ 1 ,2,4] triazin-3-ylamino)-Ν-(2-(pyrrolidin-l - yl)ethyl)benzensulfonamide) (680 mg, 95%) as a yellow solid. [0287] The title compound was prepared by using a method that was analogous to that used to synthesize compound (CXLVI), as described in Example 132, except compound 61 (680 mg, 1.23 mmol) was used to afford the title compound 4-(7-(2-chloro-6- hychoxyphenyl)-5-methylbenzo[e][l,2,4]triazin-3-ylamino)-N-(2-(pyrrolidin-l- yl)ethyl)benzensulfonamide (CLXVI) (529 mg, 75%) as HCI salt and yellow solid. 1H NMR (DMSO-/): δ 1.83-1.87 (m, 2H); 1.95-2.01 (m, 2H); 2.70 (s, 3H); 2.97-3.04 (m, 2H); 3.09-3.13 (m, 2H); 3.21-3.25 (m, 2H); 3.51-3.56 (m, 2H); 7.01 (d, J= 8.3 Hz IH); 7.06 (d, J= 7.9 Hz, IH); 7.27 (t, J= 8.0 Hz, IH); 7.77 (s, IH); 7.88 (d, J= 8.9 Hz, 2H); 7.92 (t, J= 6.0 Hz, IH); 8.09 (s, IH); 8.26 (d, J= 8.9 Hz, 2H); 10.09 (s, IH); 10.22 (br, IH); 11.40 (s, IH). MS (ESI+): m/z = 540.5.
Example 144 Synthesis of 3-{6-methyl-3-[4-(2-pyrrolidin-l-yl-ethylsuIfamoyI)- phenylamino]-benzo[l,2,41triazin-7-yl}-benzamide
Figure imgf000180_0001
(CLXVIII)
[0288] Compound 52 (scheme (CLXII), Example 140) (0.11 g, 0.22 mmol), 3- carbamoylphenylboronic acid (45 mg, 0.27 mmol), Pd(PPh3)4 (25 mg, 0.022 mmol), and 2 M Na2CO3 (0.5 mL, 1.0 mmol) were suspended in a mixture of DME/ethanol (4:1, 10 mL). The resulting mixture was heated at reflux for 6 h. The mixture was allowed to cool to room temperature, filtered and washed with DCM. The filtrate was concentrated and the residue was purified by HPLC. The corrected fractions were poured into saturated NaHCO3 and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO , and filtered. The filtrate was concentrated to afford the free base compoimd, which was converted to HCI salt according to method C and furnished the title compound as a yellow solid (40 mg, 32% overall). 1H NMR (DMSO- d6): δ 1.78-1.90 (m, 2H), 1.90-2.03 (m, 2H), 2.45 (s, 3H), 2.95-3.10 (m, 2H), 3.12 (q, J = 6.2 Hz, 2H), 3.23 (q, J= 6.1 Hz, 2H), 3.48-3.55 (m, 2H), 7.47 (s, IH), 7.61 (t, J= 7.8 Hz, IH), 7.68 (d, J= 7.8 Hz, IH), 7.83 (s, IH), 7.86 (d, J= 8.9 Hz, IH), 7.95-8.05 (m, 3H), 8.12 (s, IH), 8.20-8.24 (m, 3H), 10.55 (br s, IH), 11.33 (s, IH). MS (ESI+): m/z 533.
Example 145 Synthesis of 2-(4-{6-[7-(2,6-dichloro-phenylV5-methyl- benzo l,2,41triazin-3-ylamino1-2-methyl-pyrimidin-4-yl}-piperazin-l-yl)-ethanol
Figure imgf000181_0001
(CLXIX)
[0289] To synthesize the title compound (CLXIX), two intermediate compounds 62 (2-[4-(6-chloro-2-methyl-pyrimidin-4-yl)-piperazin-l-yl]-ethanol) and 63 (7-(2,6- dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine) shown below were used.
Figure imgf000181_0002
62
Figure imgf000181_0003
63 [0290] To synthesize compound 62, to a solution of 4,6-dichloro-2-methyl- pyrimidine (5.0 g, 31 mmol) and 2-piperazin-l-yl-ethanol (2.7 g, 21 mmol) in dioxane (25 mL) was added DEPEA (3.0 L, 17 mmol). The mixture was heated at reflux for 16 h. The mixture was allowed to cool to room temperature and poured into water. The reaulting aqueous layer was extracted with EtOAc and the combined organic layers washed with brine, dried over Na2SO4 and filtered. The filtrate was concentrated and the residue purified by flash chromatography on silica gel (5-10% MeOH/DCM) to afford compound 62 as a brown liquid (2.1 g, 39%). MS (ESI+): m/z 257.
[0291] To synthesize the title compound (CLXIX), a suspension of compound 62 (0.15 g, 0.49 mmol), compound (LXIII) (0.16 g, 0.62 mmol), Pd(OAc)2 (7 mg, 0.031 mmol), Xantphos (36 mg, 0.062 mmol) and potassium tert-butoxide (0.11 g, 0.98 mmol) in dioxane/DMF (4 mL, 3/1 v/v) was sealed in a microwave reaction tube and irradiated with microwave at 160 °C for 20 min. After cooling down to room temperature, the cap was removed and the resulting mixture filtered and the filtered solid washed with DCM. The filtrate was concentrated and the residue purified by flash chromatography on silica gel (5-10% MeOH/DCM) to afford the free base compound. The free base compound was converted to HCI salt according to method C and furnished the title compound as a yellow solid (0.18 g, 65% overall). 1H NMR (DMSO-d6): δ 2.54 (s, 3H), 2.75 (s, 3H), 3.15-3.25 (m, 4H), 3.59-3.72 (m, 6H), 3.82 (t, J= 4.6 Hz, 2H), 4.56 (br s, IH), 7.55 (t, J = 7.8 Hz, IH), 7.67 (d, J= 8.2 Hz, 2H), 7.91-7.93 (m, IH), 8.31 (d, J= 1.5 Hz, IH), 10.93 (br s, IH), 11.83 (br s, IH). MS (ESI+): m/z 525.
Example 146 Synthesis of 4-chloro-3-(3-{6-[4-(2-hydroxy-ethyl)-piperazin-l-yll-2- methyl-pyrimidin-4-ylamino}-5-methyl-benzo[l,2,4]triazin-7-yl)-phenol
Figure imgf000182_0001
(CLXX) [0292] To synthesize the title compound (CLXX), an intermediate compound 64 (2- (4- {6-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1 ,2,4]triazin-3-ylamino]-2- methyl-pyrimidin-4-yl}-piperazin-l-yl)-ethanol) shown below was used.
Figure imgf000183_0001
64
[0293] To synthesize compound 64, a suspension of compound 16 (scheme (CXXXV), Example 124) (0.10 g, 0.33 mmol), compound 53 (Example 145) (0.10 g, 0.39 mmol), Pd(OAc)2 (5 mg, 0.022 mmol), Xantphos (26 mg, 0.045 mmol) and potassium tert-butoxide (80 mg, 0.71 mmol) in dioxane/DMF (2.5 mL, 4/1 v/v) was sealed in a microwave reaction tube and irradiated with microwave at 160°C for 20 min. After cooling down to room temperature, the cap was removed and the resulting mixture filtered and the filtered solid washed with DCM. The filtrate was concentrated and the residue purified by flash chromatography on silica gel (5-10% MeOH/DCM) to yield compound 64 as a yellow solid (43 mg, 25%). MS (ESI+): m/z 521.
[0294] The title compoimd (CLXX) in the free base form was prepared from compound 64 according to method B and without further purification. The free base compound was then converted to HCI salt according to method C as a yellow solid (25 mg, 60% overall). 1H NMR (DMSO-d6): δ 2.74 (s, 3H), 3.15-3.25 (m, 4H), 3.55-3.75 (m, 6H), 3.82 (t, J= 5.1 Hz, 2H), 4.54 (br s, IH), 6.91 (dd, J= 8.8 Hz, J= 2.9 Hz, IH), 6.98 (d, J= 2.9 Hz, IH), 7.42 (d, J= 8.8 Hz, IH), 7.75 (s, IH), 8.03 (dd, J= 1.8 Hz, J= 0.9 Hz, IH), 8.30 (d, J= 1.8 Hz, IH), 10.05 (br s, IH), 10.78 (br s, IH), 11.73 (br s, IH). MS (ESI+): m/z 507 . Example 147 Synthesis of 4-[7-(2-chloro-5-hydroχy-phenyl)-5-methyl- benzo[l,2,41triazin-3-ylamino]-N-ethyl-N-(2-pyrrolidin-l-yl-ethyl)- benzenesulfonamide
Figure imgf000184_0001
(CLXXI)
[0295] To synthesize the title compound (CLXXI), intermediate compounds 65 (4- bromo-N-ethyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide) and 66 (4-[7-(2-chloro- 5 -methoxy-phenyl)-5 -methyl-benzo [ 1 ,2,4]triazin-3 -ylamino] -N-ethyl-N-(2-pyrrolidin- 1 - yl-ethyl)-benzenesulfonamide) shown below were used.
Figure imgf000184_0002
65
Figure imgf000184_0003
66
[0296] To synthesize compoimd 65, a solution of compound 38 (scheme (CLIV), Example 135) (1.0 g, 3.0 mmol), iodoethane (0.25 mL, 3.1 mmol) and cesium carbonate (1.5 g, 4.6 mmol) in acetonitrile (25 L) was stirred at room temperature for 16 h. The mixture was poured into water and extracted with EtOAc. The combined organic layers were washed with brine, dried over Νa2SO4 and filtered. The filtrate was concentrated and the residue purified by flash chromatography on silica gel (8% MeOH/DCM) to afford compound 65 as a white solid (0.80 g, 74%). 1H NMR (DMSO-d6): δ 1.03 (t, J= 7.1 Hz, 3H), 1.55-1.75 (m, 4H), 2.35-2.75 (m, 4H), 3.15-3.25 (m, 4H), 3.32 (br s, 2H), 7.76 (d, J= 8.8 Hz, 2H), 7.81 (d, J= 8.6 Hz, 2H). MS (ESI+): m/z 363 .
[0297] To synthesize compound 66, a suspension of compound 16 (scheme (CXXXV), Example 124) (0.10 g, 0.33 mmol), compound 65 (0.15 g, 0.42 mmol), Pd(OAc)2 (5 mg, 0.022 mmol), Xantphos (26 mg, 0.045 mmol) and potassium tert- butoxide (80 mg, 0.71 mmol) in dioxane/DMF (3 mL, 2/1 v/v) was sealed in a microwave reaction tube and irradiated with microwave at 160°C for 20 min. After cooling down to room temperature, the cap was removed, the resulting mixture was filtered and the filtered solid was washed with DCM. The filtrate was concentrated and the residue titrated in MeOH. Compound 66 was obtained as a yellow solid (0.17 g, 89%) after filtration and washed with MeOH. MS (ESI+): m/z 581.
[0298] The title compound (CLXXI) in the free base form was prepared from compound 66 according to method B and the crude product purified by flash chromatography on silica gel (10% MeOH/DCM). The free base compound was then converted to HCI salt according to method C to yield a yellow solid (0.13 g, 77% overall). 1H NMR (DMSO-d6): δ 1.04 (t, J= 7.1 Hz, 3H), 1.80-1.92 (m, 2H), 1.95-2.10 (m, 2H), 2.72 (s, 3H), 3.00-3.15 (m, 2H), 3.23 (q, J= 7.1 Hz, 2H), 3.28-3.38 (m, 2H), 3.40-(m, 2H), 3.53-3.63 (m, 2H), 6.89 (dd, J= 8.7 Hz, J= 2.8 Hz, IH), 6.95 (d, J= 2.9 Hz, IH), 7.42 (d, J= 8.8 Hz, IH), 7.90 (d, J= 8.8 Hz, 2H), 7.93 (dd, J= 1.8 Hz, J= 0.9 Hz, IH), 8.23 (d, J= 1.8 Hz, IH), 8.28 (d, J= 8.8 Hz, 2H), 9.97 (s, IH), 10.01 (br s, IH), 11.48 (s, IH). MS (ESI+): m/z 567.
Example 148 Synthesis of 4-[7-(2-chIoro-5-hvdroxy-phenyI)-5-methyl- benzo[l,2,41triazin-3-yIaminol-N-methyl-N-(2-morpholin-4-yl-ethyl - benzenesulfonamide
Figure imgf000186_0001
(CLXXII)
[0299] To synthesize the title compound (CLXXII), intermediate compounds 67 (4- bromo-N-methyl-benzenesulfonamide), 68 (4-bromo-N-methyl-N-(2-morpholin-4-yl- ethyl)-benzenesulfonamide) and 69 (4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamino]-N-methyl-N-(2-morpholin-4-yl-ethyl)- benzenesulfonamide) shown below were used.
Figure imgf000186_0002
67
Figure imgf000186_0003
68
Figure imgf000186_0004
69 [0300] To synthesize compound 67, to a solution of 4-bromosulfonyl chloride (5.0 g, 20 mmol) in DCM (30 mL) was added methylamine (2 M in THF, 20 mL, 40 mmol). The mixture was stirred at room temperature for 1 h and then the solvent was removed. A saturated NaHCO3 solution was added to the residue and then extracted with EtOAc. The combined organic layers were washed with water, brine, dried over anhydrous Na2SO4 and filtered. The filtrate was concentrated and the residue triturated in hexane. Compound 67 was obtained as a white solid after filtration (4.0 g, 82%). 1H NMR (DMSO-d6): δ 2.41 (d, J= 5.0 Hz, 3H), 7.56 (q, J= 4.9 Hz, IH), 7.70 (d, J= 8.7 Hz, 2H), 7.83 (d, J= 8.7 Hz, 2H).
[0301] To synthesize compound 68, a solution of compound 67 (1.0 g, 4.0 mmol), 4- (2-chloro-ethyl)-morpholine (0.85 g, 4.6 mmol) and cesium carbonate (3.3 g, 10 mmol) in acetonitrile (30 mL) was heated at reflux for 15 h. The mixture was cooled and poured into water. The aqueous layer was extracted with EtOAc and the combined organic layers washed with brine, dried over Na2SO4 and filtered. The filtrate was concentrated and the residue triturated in Et2O/hexane (1/1) to afford compoimd 68 as an off-white solid (1.2 g, 83%) after filtration. MS (ESI+): m/z 363.
[0302] To synthesize compound 69 a suspension of compound 16 (scheme (CXXXV), Example 124) (0.10 g, 0.33 mmol), compound 68 (0.15 g, 0.41 mmol), Pd(OAc)2 (5 mg, 0.022 mmol), Xantphos (26 mg, 0.045 mmol) and potassium tert-butoxide (80 mg, 0.71 mmol) in dioxane/DMF (3 mL, 2/1 v/v) was sealed in a microwave reaction tube and irradiated with microwave at 160 °C for 20 min. After cooling down to room temperature, the cap was removed and the resulting mixture filtered and the filtered solid washed with DCM. The filtrate was concentrated and the residue triturated in DCM/hexane (1/5). Compound 69 was obtained as a green solid (0.18 g, 93%) after filtration and washed with hexane. 1H NMR (DMSO-d6): δ 2.35-2.42 (m, 4H), 2.46 (t, J = 6.7 Hz, 2H), 2.72 (s, 3H), 2.73 (s, 3H), 3.10 (t, J= 6.7 Hz, 2H), 3.55 (t, J= 4.5 Hz, 2H), 3.84 (s, 3H), 7.07 (dd, J= 8.9 Hz, J= 3.0 Hz, IH), 7.16 (d, J= 3.1 Hz, IH), 7.54 (d, J= 8.9 Hz, IH), 7.84 (d, J= 8.9 Hz, 2H), 7.92 (s, IH), 8.26 (d, J= 8.9 Hz, 2H), 8.28 (d, J = 1.8 Hz, IH), 11.45 (s, IH). MS (ESI+): m/z 583. [0303] The title compound (CLXXII) in the free base form was prepared from compound 69 according to method B and the crude product purified by flash chromatography on silica gel (10% MeOH/DCM). The free base compound was then converted to HCI salt according to method C as a brown solid (0.10 g, 57% overall). 1H NMR (DMSO-d6): δ 3.10-3.20 (m, 2H), 3.30-3.40 (m, 4H), 3.50 (br d, J= 11.9 Hz, 2H), 3.78 (br t, J= 11.8 Hz, 2H), 3.99 (br d, J= 11.9 Hz, 2H), 6.89 (dd, J= 8.8 Hz, J- 2.9 Hz, IH), 6.96 (d, J= 2.9 Hz, IH), 7.41 (d, J= 8.8 Hz, IH), 7.88 (d, J= 8.9 Hz, 2H), 7.91- 7.93 (m, IH), 8.22 (d, J= 1.8 Hz, IH), 8.31 (d, J= 8.9 Hz, 2H), 10.00 (s, IH), 10.56 (br s, IH), 11.50 (s, IH). MS (ESI+): m/z 569 (M+H)+.
Example 149 Synthesis of 4-chloro-3-[3-(4-(f(2-diethyIamino-ethyl)-methyl-amino]- methyl}-phenylamino -5-methyl-benzo[l,2,4]triazin-7-yl]-phenol
Figure imgf000188_0001
(CLXXIII)
[0304] To synthesize the title compound (CLXXIII), intermediate compounds 70 (N- (4-bromo-benzyl)-N',N'-diethyl-N-methyl-ethane-l,2-diamine) and 71 (N-{4-[7-(2- chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1 ,2,4]triazin-3-ylamino]-benzyl} -N',N - diethyl-N-methyl-ethane-l,2-diamine) shown below were used.
Figure imgf000188_0002
70
Figure imgf000189_0001
71
[0305] To synthesize compound 70, a solution of N,N-diethyl-N -methyl-ethane- 1,2- diamine (3.4 g, 26 mmol), 4-bromobenzyl bromide (5.0 g, 20 mmol) and cesium carbonate (13 g, 40 mmol) in acetonitrile (60 mL) was heated at reflux for 18 h. The mixture was cooled and poured into water. The aqueous layer was extracted with EtOAc and the combined organic layers washed with brine, dried over Νa2SO4, and filtered. The filtrate was concentrated and the crude compound 70 was used in the next step without purification.
[0306] To synthesize compound 71 a suspension of compound 16 (scheme (CXXXV), Example 124) (0.10 g, 0.33 mmol), compound 70 (0.15 g, 0.50 mmol), Pd(OAc)2 (5 mg, 0.022 mmol), Xantphos (26 mg, 0.045 mmol) and potassium tert-butoxide (80 mg, 0.71 mmol) in dioxane/DMF (3 mL, 2/1 v/v) was sealed in a microwave reaction tube and irradiated with microwave at 160°C for 20 min. After cooling down to room temperature, the cap was removed, the resulting mixture was filtered, and the filtered solid was washed with DCM. The filtrate was concentrated and the residue was purified by flash chromatography on silica gel (10% MeOH/DCM to 20% MeOH, 2% TEA in DCM) to afford compound 71 as an orange solid (0.10 g, 59%). 1H NMR (DMSO-d6): δ 0.93 (t, J= 7.1 Hz, 6H), 2.15 (s, 3H), 2.35-2.46 (m, 8H), 2.66 (s, 3H), 3.47 (s, 2H), 3.83 (s, 3H), 7.05 (dd, J= 8.9 Hz, J= 3.1 Hz, IH), 7.14 (d, J= 3.2 Hz, IH), 7.32 (d, J= 8.5 Hz, IH), 7.53 (d, J= 8.9 Hz, 2H), 7.87-7.89 (m, IH), 7.98 (d, J= 8.6 Hz, 2H), 8.21 (d, J= 1.8 Hz, IH), 10.95 (s, IH). MS (ESI+): m/z 519 .
[0307] The title compound (CLXXIII) in the free base form was prepared from compound 71 according to method B and the crude product purified by HPLC. The corrected fractions were poured into saturated NaHCO3 and extracted with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, and filtered. The filtrate was concentrated to afford the free base compound, which was converted to HCI salt according to method C and furnished the title compound as an orange solid (40 mg, 19% overall). 1H NMR (DMSO-d6): δ 1.26 (t, J= 7.2 Hz, 6H), 2.69 (s, 3H), 2.71 (br s, 3H), 3.10-3.25 (m, 4H), 3.45-3.62 (m, 4H), 4.20-4.30 (m, IH), 4.51 (br d, J= 11.3 Hz, IH), 6.89 (dd, J= 8.7 Hz, J= 2.9 Hz, IH), 6.95 (d, J= 2.9 Hz, IH), 7.41 (d, J= 8.7 Hz, IH), 7.67 (d, J= 8.0 Hz, 2H), 7.89 (s, IH), 8.13 (d, J= 8.4 Hz, 2H), 8.18 (d, J= 1.7 Hz, IH), 10.00 (s, IH), 10.81 (br s, IH), 11.19 (s, IH), 11.23 (br s, IH). MS (ESI+): m/z 505.
Example 150 Synthesis of 4-[7-(2-fluoro-3-hydroxy-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000190_0001
(CLXXIN)
[0308] To synthesize the title compound (CLXXIN), intermediate compounds 72 (7- (2-fluoro-3-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine) and 73 (4-[7-(2- fluoro-3-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-l-yl- ethyl)-benzenesulfonamide) shown below were used.
Figure imgf000190_0002
72
Figure imgf000191_0001
73
[0309] To synthesize compound 72, compound 2 (scheme (CXXVI), Example 120) (1.0 g, 4.2 mmol), 2-fluoro,3-methoxybenzene boronic acid (0.87 g, 5.1 mmol), Pd(PPh3)4 (0.40 g, 0.35 mmol), 2 M Na2CO3 (5 mL, 10 mmol) were suspended in a mixture of DME/ethanol (4:1, 25 mL). The resulting mixture was heated at reflux for 1 h. The mixture was allowed to cool to room temperature, filtered and washed with DCM. The filtrate was concentrated and the residue triturated in MeOH. The resulting green solid was filtered and washed with MeOH. The resulting crude compound 72 was used in the next step without further purification. MS (ESI+): m/z 285.
[0310] To synthesize compound 73, a suspension of compound 72 (0.10 g, 0.35 mmol), compound 38 (scheme (CLIV), Example 135) (0.14 g, 0.42 mmol), Pd(OAc)2 (5 mg, 0.022 mmol), Xantphos (26 mg, 0.045 mmol) and potassium tert-butoxide (80 mg, 0.71 mmol) in dioxane/DMF (3 mL, 2/1 v/v) was sealed in a microwave reaction tube and irradiated with microwave at 160 °C for 20 min. After cooling down to RT, the cap was removed and the resulting mixture filtered and the filtered solid washed with DCM. The filtrate was concentrated and the residue purified by flash chromatography on silica gel (10-20% MeOH/DCM) to afford compound 73 as a yellow solid (80 mg, 43%). 1H NMR (DMSO-d6): δ 1.55-1.65 (m, 4H), 2.30-2.40 (m, 4H), 2.44 (t, J= 7.0 Hz, 2H), 2.72 (s, 3H), 2.80-2.90 (m, 2H), 3.92 (s, 3H), 7.20-7.35 (m, 3H), 7.80 (d, J= 8.5 Hz, IH), 7.84 (d, J= 8.9 Hz, 2H), 8.04 (s, IH), 8.21 (d, J= 8.9 Hz, 2H), 8.35 (s, IH), 11.40 (s, IH). MS (ESI+): m/z 537.
[0311] The title compound (CLXXIV) in the free base form was prepared from compound 73 (71 mg, 0.13 mmol) according to method B. The free base compound was then converted to HCI salt according to method C as an orange solid (48 mg, 66% overall). 1H NMR (DMSO-d6): δ 1.80-1.90 (m, 2H), 1.95-2.03 (m, 2H), 2.73 (s, 3H), 2.95-3.05 (m, 2H), 3.10 (q, J= 6.2 Hz, 2H), 3.24 (q, J= 6.1 Hz, 2H), 3.48-3.58 (m, 2H), 7.00-7.20 (m, 3H), 7.88 (d, J= 9.0 Hz, 2H), 7.91 (t, J= 6.1 Hz, IH), 8.05 (s, IH), 8.25 (d, J= 9.0 Hz, 2H), 8.34 (s, IH), 10.05 (br s, IH), 10.10 (s, IH), 11.45 (s, IH). MS (ESI+): m/z 523.
Example 151 Synthesis of 4-f7-(2-fl^loro- -hvdro y-phenv^)-5-methyl- benzofl,2,41triazin-3-vIamino^-N-(2- yrroIidin-l-yl-ethyl)-benzenesulfonam
Figure imgf000192_0001
(CLXXN)
[0312] To synthesize the title compound (CLXXN), intermediate compounds 74 (7- (2-fluoro-6-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine) and 75 (4-[7-(2- fluoro-6-methoxy-ρhenyl)-5-memyl-benzo[l,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-l-yl- ethyl)-benzenesulfonamide) shown below were used.
Figure imgf000192_0002
74
Figure imgf000192_0003
75 [0313] To synthesize compound 74, compound 2 (scheme (CXXVI), Example 120) (1.0 g, 4.2 mmol), 2-fluoro,6-methoxybenzene boronic acid (0.87 g, 5.1 mmol), Pd(PPh3)4 (0.40 g, 0.35 mmol), 2 M Νa2CO3 (5 mL, 10 mmol) were suspended in a mixture of DME/ethanol (4:1, 25 mL). The resulting mixture was heated at reflux for 1 h. The mixture was allowed to cool to room temperature, filtered and washed with DCM. The filtrate was concentrated and the residue triturated in MeOH. The resulting green solid was filtered and washed with MeOH. The resulting crude compound 74 was used in the next step without further purification. MS (ESI+): m/z 285.
[0314] To synthesize compound 75, a suspension of compound 74 (0.10 g, 0.35 mmol), compound 38 (scheme (CLIV), Example 135) (0.14 g, 0.42 mmol), Pd(OAc)2 (5 mg, 0.022 mmol), Xantphos (26 mg, 0.045 mmol) and potassium tert-butoxide (80 mg, 0.71 mmol) in dioxane/DMF (3 mL, 2/1 v/v) was sealed in a microwave reaction tube and irradiated with microwave at 160 °C for 20 min. After cooling down to room temperature, the cap was removed, the resulting mixture was filtered, and the filtered solid was washed with DCM. The filtrate was concentrated and the residue purified by flash chromatography on silica gel (10-20% MeOH/DCM) to afford compound 75 as a yellow solid (0.14 g, 74%). 1H NMR (DMSO-d6): δ 1.60-1.72 (m, 4H), 2.69 (s, 3H), 2.40-2.60 (m, 2H), 2.85-2.95 (m, 2H), 3.27-3.40 (m, 4H), 3.81 (s, 3H), 7.00 (t, J= 8.8 Hz, IH), 7.06 (d, J= 8.5 Hz, IH), 7.47 (dd, J= 15.3 Hz, J= 8.4 Hz, IH), 7.83 (s, IH), 7.84 (d, J= 9.0 Hz, 2H), 8.18 (s, IH), 8.22 (d, J= 8.9 Hz, 2H), 11.37 (s, IH). MS (ESI+): m/z 537.
[0315] The title compound (CLXXV) in the free base form was prepared from compound 75 (0.14 g, 0.26 mmol) according to method B. The free base compound was then converted to HCI salt according to method C as an orange solid (0.10 g, 69% overall). 1H NMR (DMSO-d6): δ 1.80-1.90 (m, 2H), 1.95-2.03 (m, 2H), 2.70 (s, 3H), 2.97-3.05 (m, 2H), 3.10 (q, J= 6.3 Hz, 2H), 3.24 (q, J= 6.1 Hz, 2H), 3.50-3.60 (m, 2H), 6.81 (t, J= 9.1 Hz, IH), 6.89 (d, J= 8.3 Hz, 2H), 7.28 (dd, J= 15.2 Hz, J= 8.3 Hz, IH), 7.88 (dd, J= 8.8 Hz, 2H), 7.89 (s, IH), 7.90 (t, J= 6.1 Hz, IH), 8.21 (s, IH), 8.26 (d, J= 9.0 Hz, 2H), 10.04 (br s, IH), 10.20 (s, IH), 11.40 (s, IH). MS (ESI+): m/z 523 . Example 152 Synthesis of benzoic acid 4-chIoro-3-f5-methvI-3-[4-(2-pyrrolidin-l-yl- ethoxy)-phenylaminol-benzo[l,2,41triazin-7-yI}-phenyl ester
Figure imgf000194_0001
(CLXXNI)
[0316] The title compound was synthesizrd and had the following characteristics. 1H ΝMR (DMSO-d6): δ 1.65-1.72 (m, 4H), 2.50-2.56 (m, 4H), 2.63 (s, 3H), 2.79 (t, J= 5.9 Hz, 2H), 4.07 (t, J= 6.0 Hz, 2H), 7.00 (d, J= 9.1 Hz, 2H), 7.46 (dd, J= 8.7 Hz, J= 2.8 Hz, IH), 7.60-7.65 (m, 3H), 7.75 (d, J= 8.6 Hz, IH), 7.76 (t, J= 7.4 Hz, IH), 7.90 (dd, J = 1.8 Hz, J= 1.0 Hz, IH), 7.93 (d, J= 9.1 Hz, 2H), 8.16 (d, J= 8.4 Hz, 2H), 8.22 (d, J= 1.8 Hz, IH), 10.84 (br s, IH). MS (ESI+): m/z 581.
[0317] For all the Examples 153-159 that follow, the synthetic procedures that were used corresponded to the general synthetic scheme (CLXXNII).
Figure imgf000194_0002
(CLXXNII) Example 153 Synthesis of N-(4-(2-(pyrrolidin-l-yl)ethoxy)phenyl)-6-methvI-7-(4- methylpyridin-3-yl)benzo[el[l,2,4]triazin-3-amine
Figure imgf000195_0001
(CLXXVIII)
[0318] The synthesis of the title compound (CLXXVIII) can be generally described by the reaction scheme (CLXXIX).
Figure imgf000195_0002
76 77 Title Compound
(CLXXIX)
[0319] To synthesize the title compound, the general Buchwald procedure was employed using compounds 76 (124 mg, 0.5 mmol), 77 (171 mg, 0.6 mmol), Cs2CO3 (660 mg, 2.0 mmol), Xantphos (58 mg, 0.1 mmol), Pd2(dba)3 (47 mg, 0.05 mmol), 3A mol sieves (ca. 500 mg), and 10 mL dioxane for 18 h. The reaction mixture was concentrated in vacuo and purified using column chromatography (1:9 MeOH/CHCl3) to afford the title compound as a red-orange solid (128 mg, 49%). Rf = 0.24 (1 :9 MeOH/CHCl3); 1H NMR DMSO-d61.87-1.94 (m, 2H), 2.02-2.11 (m, 2H), 2.12 (s, 3H), 2.19 (s, 3H), 2.30 (s, 1.7H), 3.12-3.20 (m, 2H), 3.60-3.66 (m, 4H), 4.31 (t, J= 5.0 Hz, 2H), 7.07 (d, J= 9.1 Hz, 2H), 7.45 (d, J= 5.0 Hz, IH), 7.69 (s, IH), 7.91 (d, J= 9.0 Hz, 2H), 8.07 (s, IH), 8.41 (s, IH), 8.54 (d, J= 5.1 Hz, IH), 9.72 (br s, IH), 10.75 (s, IH). Example 154 Synthesis of 5-(5-methyl-7-(2,6-dimethylphenyI)benzofe][l,2,41triazin- 3-ylamino)-l-(3-(pyrrolidin-l-yl)propyI)pyridin-2(lH)-one
Figure imgf000196_0001
(CLXXX)
[0320] The synthesis of the title compound (CLXXX) can be generally described by the reaction scheme (CLXXXI).
CH3CN Δ
Figure imgf000196_0002
Figure imgf000196_0003
78 79 80 81
Figure imgf000196_0004
82 83 84
Figure imgf000196_0005
reverse addition
85 86
THF/dioxane/DMF/H2θ
Figure imgf000196_0007
Figure imgf000196_0006
84 Title Compound
(CLXXXI) [0321] First, 2-(3-bromopropoxy)-5-nitropyridine (compound 80 on scheme (CLXXXI)) was synthesized. To synthesize, to a mixture of compounds 78 (5.0 g, 32 mmol) and 79 (4.1 mL, 47 mmol) in toluene (160 mL) was added 50% aqueous NaOH (1.9 mL, 49 mmol) followed by Adogen 464 (0.6 g, 1.6 mmol). The biphasic reaction mixture was stirred for 5 h. at 60 °C, cooled, and concentrated in vacuo. The crude mixture was purified by column chromatography (3:7-4:7 EtoAc/hexanes) to afford compound 80 as an impure pale-yellow oil (3.7 g, 45%). Rf = 0.5 (3:7 EtOAc/hexanes 1H NMR CDC13: δ 2.35 (quintet, J= 6.4 Hz, 2H), 3.56 (t, J= 6.7 Hz, 2H), 4.57 (t, J= 5.9 Hz, 2H), 6.82 (d, J= 9.6 Hz, IH), 8.36 (dd, J= 9.5, 3.0 Hz, IH), 9.07 (d, J= 2.6 Hz, IH).
[0322] Next, compound 81 (l-(3-bromopropyl)-5-nitropyridin-2(lH)-one) was synthesized by refluxing bromide 80 (3.7 g, 14 mmol) in acetonitrile (70 mL) for 5 h., cooling, and concentrating in vacuo. The crude mixture was purified by column chromatography (1:1 EtOAc/hexanes) to afford compound 81 as a pale-yellow oil (2.5 g, 68%, 90% yield corrected for starting impurity). Rf = 0.2(3:7 EtOAc/hexanes); 1H NMR CDC13: δ 2.38 (quintet , J= 6.4 Hz, 2H), 3.43 (d, J= 6.1 Hz, 2H), 4.20 (t, J= 6.9 Hz, 2H), 6.57 (d, J= 10.1 FIz, IH), 8.11 (dd, J= 10.1, 3.1 Hz, IH), 8.67 (d, J= 3.0 Hz, IH).
[0323] Next, compound 83 (5-nitro-l-(3-(pyrrolidin-l-yl)propyl)pyridin-2(lH)-one) was synthesized by adding pyrrolidine (14 mL, 169 mmol) to compound 81 (2.5 g, 9.5 mmol) in acetonitrile (50 mL); solution immediately became yellow. The reaction sat for 14 h. to ensure completion, was concentrated in vacuo, and purified by column chromatography (1:10:189 NH OH/MeOH/CH3Cl) to afford compound 83 as a pale- yellow oil (2.3 g, 95%). LC retention time: 1.09 min. MS (ESI+) m/z= 252.1 1H NMR CDC13 :δ 1.73-1.75 (m, 4H), 1.92 (quintet , J= 6.5 Hz, 2H), 2.38-2.46 (m, 6H), 4.10 (t , J = 6.5 Hz, 2H), 6.48 (d, J= 10.0 Hz, IH), 8.03 (dd, J= 10.1, 3.1 Hz, IH), 8.84 (d, J= 3.0 Hz, IH); 13C NMR CDCl3:δ 23.7, 26.7, 49.2, 51.8, 53.8, 119.3, 130.2, 133.1, 141.2, 161.7.
[0324] To compound 83 (2.1 g, 8.3 mmol) in MeθH (65 mL) was added palladium acetate (178.4 mg, 0.8 mmol), and the suspension was bubbled with H until the palladium turned black (ca. 10 min). The suspension was stirred under H2 for 3h., purged with argon, and filtered through celite using methanol to thoroughly wash the filter cake. The organic solution was concentrated in vacuo to afford compound 84 (5-amino-l-(3- (pyrrolidin-l-yl)propyl)pyridin-2(lH)-one) as a black oil (1.8 g, quant.). LC retention time = 0.5 min MS (ESI+) m/z = 222.2 1H NMR CDCl3:δ 1.91-2.00 (m, 4H), 2.15 (quintet, J= 7.3 Hz, 2H), 2.82 (t, J= 7.5 Hz, 2H), 2.88-2.95 (m, 4H), 3.63 (br s, 2H), 3.99 (t, J= 7.0 Hz, 2H), 6.49 (d, J= 9.5 Hz, IH) 6.91 (d, J= 3.0 Hz, IH), 7.04 (dd, J= 9.5, 3.0 Hz, IH).
[0325] To copper (I) bromide (581 mg, 4.1 mmol) in CH3CN (25 mL) was added 90 % isoamyl nitrite (0.5 mL, 3.4 mmol) and the solution warmed to 65 °C. A suspension of amine 85 in CH3CN (10 mL) was added and the reaction was refluxed for 90 min. The black solution was filtered, diluted with 20% HCL (80 mL) and extracted with ether (2x80 mL). The combined organic layers were washed with 20% HCI (2x40 mL), dried (MgSO4), filtered, and concentrated in vacuo to afford compound 86 (3-bromo-5-methyl- 7-(2,6-dimethylphenyl)benzo[e][l,2,4]triazine) as an oily solid which was used immediately in the next reaction (717 mg, 54%). Rf = 0.58 (3:7 EtOAc/hexanes).
[0326] To compound 86 (167 mg, 0.5 mmol) in THF (2 mL) was added a solution of 0.1 M (1:1:2:6 THF/dioxane/DMF/water) compoimd 84 (5.0 mL, 0.5 mmol) obtained as described above. After 22 h. under reflux with only minimal forward reaction, TEA (140 μL, 1.0 mmol) was added and continued reflux for 42 h. The reaction was diluted with water (10 mL) and extracted with EtOAc (3x15 mL). The organic layers were combined, dried (MgSO4), filtered, and concentrated in vacuo. The crude mixture was purified by HPLC to afford the title product as an orange solid (3.7 mg, 1.3%). LC retention time = 2.54 min,MS (ESI+) m/z = 469.4 1H NMR DMSO-d6: δ 1.81-1.88 (m, 2H), 1.97-2.02 (m, 2H), 2.05 (s, 6H), 2.08-2.16 (m, 2H), 2.65 (s, 3H), 2.95-3.05 (m, 2H), 3.15-3.24 (m, 2H), 4.05 (t, J= 6.8 Hz, 2H), 6.58 (d, J= 9.7 Hz, IH), 7.15-7.28 (m, 3H), 7.63 (s, IH), 7.79 (dd, J= 9.8, 2.9 Hz, IH), 7.94 (d, J= 1.6 Hz, IH), 8.62 (br s, IH), 9.51 (br s, IH), 10.71 (br s, IH). Example 155 Synthesis of 2-f7-(2-chloro-5-hydroxyphenyI)-5- methyIbenzo[e][l,2,4]triazin-3-ylamino)-N-(2-(pyrrolidin-l-yl)ethyl)thiazole-4- carboxamide
Figure imgf000199_0001
(CLXXXII)
[0327] The synthesis of the title compound (CLXXXII) can be generally described by the reaction scheme (CLXXXIII).
Figure imgf000199_0002
85 86 87
Figure imgf000199_0003
Title Compound
(CLXXXπi)
[0328] The general Buchwald procedure was employed using compounds 85 (254 mg, 0.84 mmol), 86 (214 mg, 0.70 mmol), Cs2CO3 (937 mg, 2.88 mmol), Xantphos (83 mg, 0,14 mmol), Pd2(dba)3 (66 mg, 0.07 mmol), and 15 mL dioxane for 22 h. The crude reaction mixture was diluted with 50 mL water and extracted with DCM (4x75 mL). The combined organic layers were concentrated in vacuo and purified using column chromatography (1:5:94 NH4OH/MeOH/DCM) to afford compound 87 as a yellow solid (188 mg, 51%). Rf = 0.06 (1:20:179 % NH4OH/MeOH/CHCl3); LC retention time: 2.67 min, MS (ESI+) m/z = 524.2/526.4.
[0329] The general aryl methoxy deprotection procedure was then employed using aryl methoxy compound 87 (188 mg, 0.36 mmol) and IM BBr3 in DCM (2.1 mL, 2.1 mmol), in 12 mL DCM for 1 h. The reaction was quenched and filtered to afford the title compound (CLXXXII) as a brown solid (132 mg, 72%). LC retention time: 2.20 min, MS (ESI+) m/z = 510.5/512.3 1H NMR DMSO-d6: δ 1.72(s, 4H), 2.52(shoulder under DMSO), 2.62 (t, J= 6.4 Hz, 2H), 2.76 (s, 3H), 3.43 (q, J= 6.1 Hz, 2H), 6.88 (dd, J= 8.8, 2.9 Hz, IH), 6.95 (d, J= 2.9 Hz, IH), 7.41 (d, J= 8.7 Hz, IH), 7.83 (s, IH), 7.86 (t, J= 5.8 Hz, IH), 7.94 (s, IH), 8.23 (s, IH), 9.94 (br s, IH). Calcd for C24H24ClN7O2S-2.5H2O.0.1Br: C 51.14, H 5.36, N 17.39. Found C 50.74, H 4.79, N 17.03.
Example 156 Synthesis of 3-(3-(4-(2-(pyrrolidin-l-yl)ethoxy)phenylamino)-6- methyIbenzo[e1[l,2,41triazm-7-yl)-4-fluorophenol
Figure imgf000200_0001
(CLXXXIV)
[0330] The synthesis of the title compound (CLXXXIV) can be generally described by the reaction scheme (CLXXXV).
Figure imgf000200_0002
88 77 89
Figure imgf000201_0001
Title Compound
(CLXXXV)
[0331] The general Buchwald procedure was employed using compounds 88 (2-(7-(2- chloro-5-methoxyphenyl)-5-methylbenzo[e] [ 1 ,2,4]triazin-3 -ylamino)-N-(2-(pyrrolidin- 1 - yl)ethyl)thiazole-4-carboxamide) (254 mg, 0.94 mmol), 77 (described above) (177 μL, 0.85 mmol), Cs2CO3 (1.1 g, 3.42 mmol), Xantphos (102 mg, 0.18 mmol), Pd2(dba)3 (78 mg, 0.09 mmol), and 17 mL dioxane for 18 h, to obtain compound 89 (N-(4-(2- (pyrrolidin-l-yl)ethoxy)phenyl)-7-(2-fluoro-5-methoxyphenyl)-6- methylbenzo[e] [ 1 ,2,4]triazin-3-amine). Chromatography was run at 1 : 10: 189 NH4OH/MeOH/CHCl3 to afford the title compound as a red solid (181 mg, 45%). Rf = 0.06 (1:20:179 % NH4OH/MeOH/CHCl3); LC retention time: 2.65 min, MS (ESI+) m/z = 474.2.
[0332] The general aryl methoxy deprotection procedure was then employed using aryl methoxy compound 89 (181 mg, 0.38 mmol) and IM BBr3 in DCM (2.3 mL, 2.3 mmol), in 10 mL DCM for 90 min. The reaction was quenched and filtered. The water soluble product was recovered by extraction with EtOAc (50 mL). The organic layer was combined with the filtered solid, concentrated in vacuo, and purified using HPLC to afford the title compoimd (CLXXXV) as a red solid (30 mg, 14%). LC retention time: 2.26 min, MS (ESI+) m/z = 460.2 1H NMR DMSO-d6: δ 1.86-1.94 (m, 2H), 2.02-2.08 (m, 2H), 2.32 (s, 3H), 3.11-3.20 (m, 2H), 3.58-3.66 (m, 4H), 4.30 (t, J= 5.0 Hz, 2H), 6.76 (dd, J= 6.2, 3.1 Hz, IH), 6.86 (dt, J= 8.9, 3.6 Hz, IH), 7.07 (d, J= 9.1 Hz, 2H), 7.16 (t, J= 9.2 Hz, IH), 7.64 (s, IH), 7.91 (d, J= 9.1 Hz, 2H), 8.09 (s, IH), 9.63 (s, IH), 9.71 (br s, IH), 10.75 (s, IH). Example 157 Synthesis of 4-r7-(2-fluoro-5-hvdroxy-phenvIV6-methyl- benzo[l,2,41triazin-3-ylamino]-N-(2-pyrroIidin-l-yl-ethyl)-benzenesnlfonamide
Figure imgf000202_0001
(CLXXXVI)
[0333] The synthesis of the title compound (CLXXXVI) can be generally described by the reaction scheme (CLXXXVII).
Figure imgf000202_0002
90 91
Figure imgf000202_0003
92 Title Compound
(CLXXXVII)
[0334] The general Suzuki procedure was employed using compounds 90 (79 mg, 0.47 mmol), 91 (157 mg, 0.32 mmol), Pd(Ph3)4 (39 mg, 0.03 mmol), and 2M aqueous Na2CO3 (0.6 mL, 1.2 mmol) for 2.5 h to afford, after chromatography (1:10:189 NH4OH/MeOH/CHCl3), compound 92 (4-[7-(2-fluoro-5-methoxy-phenyl)-6-methyl- benzo[ 1 ,2,4]triazin-3-ylamino]-N-(2-pyrrolidin- 1 -yl-ethyl)-benzenesulfonamide), as an orange solid (89 mg, 52%). LC retention time: 2.66 min, MS (ESI+) m/z = 537.2.
[0335] The general aryl methoxy deprotection procedure was then employed using aryl methoxy compound 92 (89 mg, 0.17 mmol) and IM BBr3 in DCM (1 mL, 1.0 mmol), in 20 mL DCM for 90 min. The reaction was quenched, filtered, and purified using HPLC to afford the title compound (CLXXXVI), as an orange solid (19 mg, 22%). LC retention time: 2.24 min, MS (ESI+) m/z = 523.2 1H NMR DMSO-d6: δ 1.83-1.91 (m, 2H), 1.96-2.04 (m, 2H), 2.36 (s, 3H), 2.99-3.08 (m, 4H), 3.21-3.27 (m, 2H), 3.52- 3.60 (m, 2H), 6.78 (dd, J= 6.2, 3.1 Hz, IH), 6.88 (dt, J= 9.2 Hz, IH), 7.79 (s, IH), 7.84 (d, J= 8.9 Hz, 2H), 8.86 (t, J= 6.2 Hz, IH), 8.19 (s, IH), 8.22 (d, J= 8.9 Hz, 2H), 9.52 (br s, IH), 9.66 (s, IH).
Example 158 Synthesis of 4-[6-methyl-7-(lH-pyrazol-4-yI)-benzo[l,2,4]triazin-3- ylamino1-N-(2-pyrrolidin-l-yl-ethyl)-benzenesuIfonamide
Figure imgf000203_0001
(CLXXXVIII)
[0336] The synthesis of the title compound (CLXXXVIII) can be generally described by the reaction scheme (CLXXXIX).
Figure imgf000203_0002
93 91 Title Compound
(CLXXXIX) [0337] The general Suzuki procedure was employed using compounds 93 (117 mg, 2.2 mmol), 91 (206 mg, 0.42 mmol), Pd(Ph3)4 (53 mg, 0.05 mmol), and CsF (128 mg, 0.84 mmol) in 4:1 DME/MEOH (4 mL) and 2M aqueous Na2CO3 (0.8 mL, 1.2 mmol) for 16 h. Additional amounts of compound 93 (308 mg, 1.6 mmol), Pd(Ph3)4 (53 mg, 0.05 mmol), and CsF (178 mg, 1.2 mmol) were added and the reaction was refluxed for 7 h. to afford, after HPLC purification, the title compound (CLXXXVIII), as a yellow solid (62 mg, 31%). LC retention time: 1.94 min, MS (ESI+) m/z = 479.2 1H NMR DMSO-d6: δ 1.82-1.91 (m, 2H), 7.97-2.04 (m, 2H), 2.64 (s, 3H), 2.99-3.08 (m, 4H), 3.22-3.27 (m, 2H), 3.53-3.59 (m, 2H), 7.76 (s, IH), 7.84 (d, J= 7.1 Hz, 2H), 7.86 (t, J= 6.2 Hz, IH), 8.11 (br s, IH), 8.20 (d, J= 7.1 Hz, 2H), 8.35 (s, IH), 9.57 (br s, IH), 11.24 (s, IH).
Example 159 Synthesis of 4-[7-(2-chloro-5-hvdroxy-phenyl)-6-methyl- benzo[l,2,41triazin-3-ylamino1-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000204_0001
(CXC)
[0338] The synthesis of the title compound (CXC) can be generally described by the reaction scheme (CXCI).
Figure imgf000204_0002
94 91 95
Figure imgf000205_0001
Title Compound
(CXCI)
[0339] The general Suzuki procedure was employed using compounds 94 (194 mg, 1.0 mmol), 91 (252 mg, 0.5 mmol), Pd(Ph3)4 (59 mg, 0.05 mmol), and 2M aqueous Na2CO3 (1.0 mL, 2.0 mmol) for 15 h. The reaction was diluted with DCM (10 mL) and water (10 mL) and the biphasic mixture was filtered to afford compound 95 (4-[7-(2- chloro-5 -methoxy-phenyl)-6-methyl-benzo [ 1 ,2,4] triazin-3 -ylamino] ~N-(2-pyrrolidin- 1 - yl-ethyl)-benzenesulfonamide), as an orange solid (85 mg, 15%). LC retention time: 2.75 min, MS (ESI+) m/z = 553.2/555.4.
[0340] The general aryl methoxy deprotection procedure was then employed using aryl methoxy compound 95 (86 mg, 0.16 mmol) and IM BBr3 in DCM (0.93 mL, 0.93 mmol), in 5 mL DCM for 3 h. The reaction was quenched, filtered, and purified using HPLC to afford the title compound (CXC), as an orange solid (19 mg, 23%). LC retention time: 2.42 min, MS (ESI+) m/z = 539.1/541.4 JH NMR DMSO-d6: δ 1.83-1.89 (m, 2H), 1.94-2.03 (m, 2H), 2.29(s, 3H), 2.98-3.05 (m, 2H), 3.09 (q, J=6.2 Hz, 2H), 3.22- 3.25 (m, 2H), 3.50-3.57 (m, 2H), 6.81 (d, J= 2.9 Hz, IH), 6.91 (dd, J= 8.8, 2.9 Hz, IH), 7.41 (d, J= 8.8Hz, IH), 7.80 (s, IH), 7.85 (d, J= 8.9 Hz, 2H), 7.92 (t, J= 4.5 Hz, IH), 8.11 (s, IH), 8.21 (d, J= 8.9 Hz, 2H), 9.98 (s, 2H), 11.33 (s, IH).
Example 160 Synthesis of 4-[7-(3-hvdroxymethyl-phenyl)-6-methyl- benzo[l,2,4]triazin-3-ylamino1-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000206_0001
(CXCII)
[0341] The synthesis of the title compound (CXCII) can be generally described by the reaction schemes (CXCIII) and (CXCIV).
Figure imgf000206_0002
(CXCIII)
[0342] The mixture of 7-bromo-6-methylbenzo[e][l,2,4]triazin-3-amine (240 mg, 1.0 mmol), 3-(hydroxymethyl)phenylboronic acid (310 mg, 2.0 mmol), ethylene glycol dimethyl ether (10 mL), EtOH (1 mL), H2O (1 mL), Pd(PPh3)4 (120 mg, 0.1 mmol) and Na2CO (1.1 g, 10 mmol) was degassed with argon for 5 minutes and refluxed for overnight under argon. The reaction mixture was brought to room temperature, and the solid was removed by filtration. After evaporation of the volatiles, the crude product was purified by silica gel column (3.5 x 16 cm) chromatography with 10% CH3OH/CHCl3 as an eluent to give a yellow solid product (200 mg, 75%).
[0343] The product of reaction shown by scheme (CXCIII) was further reacted as shown by scheme (CXCIV).
Figure imgf000207_0001
(CXCIV)
[0344] A mixture of (3-(3-amino-6-methylbenzo[e] [ 1 ,2,4]triazin-7- yl)phenyl)methanol (200 mg, 0.75 mmol), 4-bromo-N-(2-pyrrolidin-l-yl-ethyl)- benzenesulfonamide (380 mg, 1.13 mmol), cesium carbonate (730 mg, 2.25 mmol), Xantphos (90 mg, 0.15 mmol), Pd2(dba)3 (70 mg, 0.075 mmmol) in 15 mL of anhydrous dioxane was degassed with argon for 5 minutes and was refluxed for 4 h. The reaction mixture was brought to room temperature and the solvent was removed under reduced pressure. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography using 30% CH3OH in CHC13 as an eluent. The solid was washed with acetone to afford the title compoimd (CXCII), as a bright yellow solid (200 mg, 52%). 1H NMR DMSO-d6: δ 1.67 (br s, 4H), 2.44 (s, 3H), 2.50 (br s, 4H), 2.88 (br s, 2H), 4.60 (d, J = 3.6 Hz, 2H), 5.28 (br s, IH), 7.43 (m, 5H), 7.80 (m, 3H), 8.12 (s, IH), 8.18 (d, J= 8.7 Hz, 2H), 11.25 (s, IH). MS (ESI+) m/z = 519.
Example 161 Synthesis of 4-f7-(5-chloro-thiophen-2-yl)-6-methyl- benzo[l,2,4]triazin-3-ylamino1-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000207_0002
(CXCV)
[0345] The synthesis of the title compound (CXCV) can be generally described by the reaction schemes (CXCVI) and (CXCVII).
Figure imgf000208_0001
(CXCVI)
[0346] The mixture of 7-bromo-6-methylbenzo[e] [ 1 ,2,4]triazin-3-amine (310 mg, 1.3 mmol), 5-chloro-2-thiopheneboronic acid (320 mg, 2.0 mmol), ethylene glycol dimethyl ether (10 mL), EtOH (1 mL), H2O (1 mL), Pd(PPh3)4 (150 mg, 0.13 mmol) and Na2CO3 (1.38 g, 13 mmol) was degassed with argon for 5 minutes and refluxed for 16 h under argon. The reaction mixture was brought to room temperature, and the solid was removed by filtration. After evaporation of the volatiles, the crude product was purified by silica gel column (3.5 x 16 cm) chromatography using 10% CH3OH/CHCl3 as an eluent to give a yellow solid (220 mg, 61%).
[0347] The product of reaction shown by scheme (CXCVI) was further reacted as shown by scheme (CXC VII).
Figure imgf000208_0002
(cxcvπ)
[0348] A mixture of 7-(5-chlorothiophen-2-yl)-6-methylbenzo[e][l,2,4]triazin-3- amine (110 mg, 0.4 mmol), 4-bromo-N-(2-pyrrolidin-l-yl-ethyl)benzenesulfonamide (260 mg, 0.78 mmol), cesium carbonate (390 mg, 1.2 mmol), Xantphos(46 mg, 0.08 mmol), Pd2(dba)3(36 mg, 0.04 mmmol) in anhydrous dioxane (15 mL) was degassed with argon for 5 minutes and the reaction mixture was refluxed for 16 h. The solvent was removed under reduced pressure and the crude product was purified by silica gel column (3.5 x 16 cm) chromatography using 30% CH3OH in CHC13 as an eluent, to afford the title product (CXCV), as an orange solid (110 mg, 52%).1H NMR DMSO-d6: δ 1.65 (br s, 4H), 2.43 (br s, 4H), 2.60 (s, 3H), 2.86 (t, J= 6.5 Hz, 2H), 7.26 (d, J= 3.9 Hz, IH), 7.31 (d, J= 3.9 Hz, IH), 7.45 (br s, IH), 7.81 (m, 3H), 8.16 (d, J= 9.3 Hz, 2), 8.32 (s, IH), 11.33 (s, IH). MS (ESI+) m/z = 529.
Example 162 Synthesis of 4-(6-methoxy-7-o-tolyl-benzo[l,2,41triazin-3-ylamino)-N- (2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000209_0001
(CXCVIII)
[0349] The synthesis of the title compound (CXCVIII) can be generally described by the sequence of reaction schemes (CXCIX), (CC), (CCI), (CCII), (CCIII), and (CCIV).
CH3OH
Figure imgf000209_0002
(CXCIX)
[0350] To freshly prepared sodium methoxide using sodium (900 mg, 39 mmol) and anhydrous methanol (20 mL) was added 3-chloro-6-nitroaniline (2.55 g, 15 mmol). The reaction mixture was heated in a sealed tube at 120°C for 4 hours. The solvent was removed under reduced pressure and the crude solid was dissolved in water (50 mL) and extracted with ethyl acetate (3 x 50 mL). The combined ethyl acetate layer was dried over sodium sulfate. The sodium sulfate was removed by filtration and the solvent was removed. The crude was dried to yield a yellow solid (1.75 g, 69%).
[0351] The product of reaction shown by scheme (CXCIX) was further reacted as shown by scheme (CC).
Figure imgf000210_0001
(CC)
[0352] A mixture of 3-methoxy-6-nitroaniline (940 mg, 5.6 mmol) and NBS (1.1 g, 6.2 mmol) in glacial acetic acid (25 mL) was heated to reflux under argon for 3 h. The reaction mixture was brought to room temperature and the mixture was diluted with water (100 mL). The yellow precipitate was collected by filtration, washed with water to yield a yellow solid (1 g, 72%).
[0353] The product of reaction shown by scheme (CC) was further reacted as shown by scheme (CCI).
Figure imgf000210_0002
(CCI)
[0354] 4-bromo-3-methoxy-6-r-itroaniline (lg, 4.05 mmol) was mixed with cyanamide (1.7 g, 40 mmol) and heated to melt at 90°C. Concentrated HCI (10 mL) was cautiously added drop wise within 20 minutes. The reaction mixture was heated to reflux until all starting material reacted ( about 2 h). Another batch of cyanamide (1.7 g, 40 mmol) and hydrochloric acid (10 mL) was added and the reaction continued for another hour. The ice cooled reaction mixture was brought to pH 13 with 30% sodium hydroxide. The mixture was heated to reflux for 3 h. The precipitate was collected by filtration and washed with water to give a yellow solid (900 mg, 82%).
[0355] The product of reaction shown by scheme (CCI) was further reacted as shown by scheme (CCII).
Figure imgf000211_0001
(ccπ)
[0356] A mixture of 7-bromo-6-methoxybenzo[e][l,2,4]triazin-3-amine-l-N-oxide (350 mg, 1.3 mmol), 2-methyl phenylboronic acid (350 mg, 2.6 mmol), ethylene glycol dimethyl ether (30 mL), EtOH (3 mL), H2O (3 mL), Pd(PPh3)4 (150 mg, 0.13 mmol) and Na2CO3 (1.38 g, 13 mmol) was degassed with argon for 5 minutes and refluxed for 16 h under argon. The reaction mixture was cooled to room temperature, and the solid was removed by filtration. After evaporation of the volatiles, the crude product was purified by silica gel column (3.5 x 16 cm) chromatography using 5% CH3OH/CHCl3 as an eluent to give a solid. The solid was washed with dichloromethane/hexanes (1 : 1) to give a yellow solid (220 mg, 61%).
[0357] The product of reaction shown by scheme (CCII) was further reacted as shown by scheme (CCIII).
Figure imgf000211_0002
(ccπi)
[0358] A mixture of 6-methoxy-7-o-tolylbenzo[e][l,2,4]triazin-3-amine-l-N-oxide (230 mg, 0.82 mmol), 4-bromo-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (540 mg, 1.64 mmol), cesium carbonate (800 mg, 2.45 mmol), Xantphos (94 mg, 0.16 mmol), Pd2(dba)3(75 mg, 0.08 mmmol) in 15 mL of anhydrous dioxane was degassed with argon for 5 minutes and was refluxed for 4 h. The reaction mixture was cooled to room temperature and the solvent was removed under reduced pressure. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography using 20% CH3OH in CHC13 as an eluent. The solid was washed with minimum amount of methanol ( about 5 mL) to give a orange solid (340 mg, 66%).
[0359] The product of reaction shown by scheme (CCffl) was further reacted as shown by scheme (CCIV).
H2, Raney Ni
Figure imgf000212_0001
(CCIV)
[0360] 4-(6-methoxy-l-oxy-7-o-tolyl-benzo[l,2,4]triazin-3-ylamino)-N-(2-pyrrolidin- l-yl-ethyl)-benzenesulfonamide (140 mg, 0.26 mmol) was reduced in methanol (25 mL) and ethyl acetate (25 mL) mixture with catalytic amount of Raney Ni and hydrogen for 4 h. The catalyst was removed by filtration and the solvent was removed under reduced pressure to afford the title product (CXCVIII), as a yellow solid (60 mg, 44%).1H NMR DMSO-d6: δ 1.63 (br s, 4H), 2.11 (s, 3H), 2.37 (br s, 4H), 2.44 (br s, 2H), 2.85 (t, J= 6.8 Hz, 2H), 3.97 (s, 3H), 7.27 (m, 5H),7.42 (br s, IH), 7.79 (d, J= 8.8 Hz, 2H), 8.02 (s, IH), 8.18 (d, J= 8.9 Hz, 2H), 11.12 (s, IH). MS (ESI+) m/z = 519.
Example 163 Synthesis of 4-(6-hydroxy-7-o-tolyl-benzo[l,2,4]triazin-3-ylamino)-N- (2-pyrrolidin-l-yI-ethyl)-benzenesuIfonamide
Figure imgf000212_0002
(CCV) [0361] The synthesis of the title compound (CCV) can be generally described by the of reaction scheme (CCVI).
pyridine HCI
160°C in a sealed tube
Figure imgf000213_0001
(CCNI)
[0362] 4-(6-methoxy-7-o-tolyl-benzo[ 1 ,2,4]triazin-3-ylamino)-Ν-(2-pyrrolidin- 1 -yl- ethyl)-benzenesulfonamide (70 mg, 0.14 mmol) and pyridine hydrochloride (1.0 g, 8.6 mmol) was heated at 160°C in a seal tube for 4 h. The crude was suspended in diluted HCI (30 mL), and the crude product was purified by silica gel column with 20% CH3OH in CHC13 as an eluent to afford the title compoimd (CCVI), as a yellow solid (15 mg, 22%). 1H NMR DMSO-d6: δ 1.65 (br s, 4H), 2.17 (s, 3H), 2.43 (br s, 4H), 2.86 (br s, 2H), 3.97 (s, 3H), 7.27 (m, 5H),7.42 (br s, IH), 7.79 (d, J= 8.8 Hz, 2H), 8.02 (s, IH), 8.18 (d, J= 8.9 Hz, 2H), 11.12 (s, IH). MS (ESI+) m/z = 519.
Example 164 Synthesis of 4-(6-ethoxy-7-o-tolyl-benzo[l,2,41triazin-3-ylamino)-N-(2- pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000213_0002
(CCVII)
[0363] The synthesis of the title compound (CCVII) can be generally described by the sequence of reaction schemes (CCVIII), (CCIX), (CCX), and (CCXI). pyridine HCI, 175°C in a sealed tube
Figure imgf000214_0001
Figure imgf000214_0002
(CCNIII)
[0364] The mixture of 7-bromo-6-methoxybenzo[e][l,2,4]triazin-3-amine-l-Ν-oxide (170 mg, 0.6 mmol) and pyridine hydrochloride (1.0 g, 8.6 mmol) was heated at 170°C in a seal tube for 2 h. The crude was triturated with dilute HCI (30 mL). The resulting yellow precipitate (100 mg, 62%) was isolated by filtration.
[0365] The product of reaction shown by scheme (CCNIII) was further reacted as shown by scheme (CCIX).
Figure imgf000214_0003
(CCIX)
[0366] 3-amino-7-o-tolylbenzo[e][l,2,4]triazin-6-ol-l-N-oxide (60 mg, 0.2mmol) was reduced in methanol (10 mL) and ethyl acetate (10 mL) with catalytic amount of Raney Ni and hydrogen at room temperature for 4 hours. The catalyst was removed by filtration and the solvent was removed. The solid was washed with acetone to give a yellow solid (50 mg, 89%).
[0367] The product of reaction shown by scheme (CCIX) was further reacted as shown by scheme (CCX).
Figure imgf000215_0001
(CCX)
[0368] A mixture of 3-amino-7-o-tolylbenzo[e][l,2,4]triazin-6-ol (0.06 g, 0.22 mmol), ethyl bromide (0.24 g, 2.2 mmol), cesium, carbonate (0.3 g, 0.92mmol) and KI (0.1 g, 0.6 mmol) in acetone/methanol (5:1, 20 mL) was refluxed for 3 hours. After removing the solvent, the crude product was purified by silica gel column with 5% methanol/ chloroform as an eluent as an orange solid (0.046 g, 73%).
[0369] The product of reaction shown by scheme (CCX) was further reacted as shown by scheme (CCXI).
Figure imgf000215_0002
(CCXI)
[0370] A mixture of 6-ethoxy-7-o-tolylbenzo[e] [ 1 ,2,4]triazin-3-amine (0.046 g, 0.21 mmol), 4-bromo-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.14 g, 0.43 mmol), cesium carbonate (0.21 g, 0.63 mmol), Xantphos (25 mg, 0.15 mmol), Pd2(dba)3(20 mg, 0.021 mmol) in 10 mL of anliydrous dioxane was degassed with argon for 5 minutes and was refluxed for 16 h. The solvent was removed and the crude product was purified by silica gel column (3.5 x 16 cm) chromatography using 20% CH3OH in CHC13 as an eluent to afford the title compound (CCVII), as a yellow solid (61 mg, 52%). 1H NMR DMSO-d6: δ 1.27 (t, J= 7Hz, 3 H), 1.62 (m, 4H), 2.13 (s, 3H), 2.35 (br s, 4H), 2.43 (t, J = 7.3 Hz, 2H), 2.85 (t, J= 6.9 Hz, 2H), 4.29 (m, 2H), 7.30 (m, 5H), 7.42 (br s, IH), 7.79 (d, J= 8.9 Hz), 8.02 (s, IH), 8.18 (d, J= 8.9 Hz, 2H). 11.09 (s, IH). MS (ESI+) m/z = 533. Example 165 Synthesis of 4- r7-(3-hydroxy-phenvIV6-methoxy-benzo [1,2,41 triazin-3- ylamino1-N-(2-pyrroIidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000216_0001
(CCXII)
[0371] The synthesis of the title compound (CCXII) can be generally described by the sequence of reaction schemes (CCXIII), (CCXIV), (CCXV), and (CCXVI).
Figure imgf000216_0002
(CCXIII)
[0372] The mixture of 7-bromo-6-methoxybenzo[e][l,2,4]triazin-3-amine-l-N-oxide (0.14 g, 0.52 mmol), 3-benzyloxyphenylboronic acid (0.24 g, O.lOmmol), ethylene glycol dimethyl ether (15 mL), EtOH (1.5 mL), H2O (1.5 mL), Pd(PPh3)4 (60 mg, 0.05 mmol) and Na2CO3 (0.55 g, 5.2 mmol) was degassed with argon for 5 minutes and refluxed for 16 h under argon. The solid was filtered off, and the volatiles were evaporated. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography using 10% CH3OH/CHCl3 as an eluent to give a yellow solid (0. 14 g, 72%).
[0373] The product of reaction shown by scheme (CCXIII) was further reacted as shown by scheme (CCXIN).
Figure imgf000217_0001
(CCXIV)
[0374] A mixture of 7-(3-(benzyloxy)phenyl)-6-methoxybenzo[e] [ 1 ,2,4]triazin-3- amine-1-N-oxide (0.11 g, 0.29 mmol), 4-bromo-N-(2-pyrrolidin-l-yl-ethyl)- benzenesulfonamide (0.20 g, 0.6 mmol), cesium carbonate (0.29 g, 0.89 mmol), Xantphos (34 mg, 0.06 mmol), Pd2(dba)3(27 mg, 0.029 mmol) in 15 mL of anliydrous dioxane was degassed with argon for 5 minutes and was refluxed overnight. The reaction mixture was brought to room, temperature and the solvent was removed under reduced pressure. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography using 30% CH3OH in CHC13 as an eluent. The solid was washed with acetone to give a yellow solid (0.1 g, 54%).
[0375] The product of reaction shown by scheme (CCXIV) was further reacted as shown by scheme (CCXV).
H2. Raney Ni
(CCXV)
[0376] 4-[7-(3-benzyloxy-phenyl)-6-methoxy-l-oxy-benzo[l,2,4]triazin-3-ylamino]- N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.1 g, 0.16mmol) was dissolved in methanol (10 mL) and ethyl acetate (10 mL) with catalytic amount of Raney Ni and atmosphere hydrogen at room temperature for 4 hours. The catalyst was removed by 217
filtration. The solvent was removed by reduced pressure. The solid was used for the next reaction without further purification.
[0377] The product of reaction shown by scheme (CCXV) was further reacted as shown by scheme (CCX VI).
Figure imgf000218_0001
(CCXVI)
[0378] The above 4-[7-(3-benzyloxy-phenyl)-6-methoxy-benzo[ 1 ,2,4]triazin-3- ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide in chloroform (10 mL) and ethyl acetate (10 mL) was stirred with IM BBr3 in dichloromethane (10 mL, 10 mmol) at room temperature for 5 h. The reaction was quenched by methanol (1 mL). After removing the solvent, the crude product was purified by silica gel column chromatography with 20% % CH3OH/CHCl3 as an eluent to give a solid. The solid was washed with acetone to afford the title compound (CCXII), as a yellow solid (0.03g, 36% in 2-steps).1H NMR DMSO-d6: δ 1.62 (br s, 4H), 2.36 (br s, 4H), 2.43 (t, J= 7 Hz, 2H), 2.85 (t, J= 6.9 Hz, 2H), 4.02 (s, 3H),6.83 (d, J= 8 Hz, IH), 6.99 (m, 2H), 7.27 (t, J= 8.7 Hz), 7.42 (br s, IH), 7.80 (d, J= 8.8 Hz, 2H), 8.12 (s, IH), 8.17 (d, J= 8.8 Hz, 2H), 9.55 (s, IH), 11.10 (s, IH). MS (ESI+) m/z = 521.
Example 166 Synthesis of 4-[6-(2-hvdroxy-ethoχy)-7-o-toIyl-benzofl,2,41triazin-3- ylamino1-N-(2-pyrrolidin-l-yl-ethyl)-benzenesuIfonamide
Figure imgf000219_0001
(CCXNII)
[0379] The synthesis of the title compound (CCXNII) can be generally described by the sequence of reaction schemes (CCXNffl) and (CCXIX).
Figure imgf000219_0002
(ccxviπ)
[0380] 3-amino-7-o-tolylbenzo[e][l,2,4]triazin-6-ol (0.1 g, 0.4 mmol), 2- bromoethanol (0.5 g, 4 mmol), cesium carbonate (0.3 g, 0.92mmol) and KI (0.1 g, 0.6 mmol) was stiired in acetone/methanol (5:1, 20 mL) at room temperature for overnight. After cooling down, the solvent was removed by reduced pressure. The crude was purified by silica gel column with 20% methanol/chloroform as an eluent to give an orange solid (0.04g, 34%).
[0381] The product of reaction shown by scheme (CCXVIII) was further reacted as shown by scheme (CCXIX).
Figure imgf000220_0001
(CCXIX)
[0382] A mixture of 2-(3-amino-7-o-tolylbenzo[e] [ 1 ,2,4]triazin-6-yloxy)ethanol (0.04g, 0.13 mmol), 4-bromo-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.09 g, 0.27 mmol), cesium carbonate (0.13 g, 0.4 mmol), Xantphos (20 mg, 0.03 mmol), Pd2(dba)3(10 mg, 0.01 mmol) in 10 mL of anhydrous dioxane was degassed with argon for 5 minutes and was refluxed for overnight. The reaction mixture was brought to room temperature and the solvent was removed under reduced pressure. The crude product was purified by silica gel column (3.5 x 10 cm) cliromatography using 30% CH3OH in CHC13 as an eluent to afford the title compound (CCXVII), as an orange solid (0.01 g, 14%).1H NMRDMSO-d6: δ 1.65 (br s, 4H), 2.10 (s, 3H), 2.41 (br s, 4H), 2.87 (t, J= 6.9Hz, 2H), 3.87 (t, J= 5.0Hz, 2H), 4.42 (t, J= 4.9 Hz, 2H), 5.92 (s, IH), 7.10 (d, J= 7.3 Hz, IH), 7.24 (m, 4H), 7.48 (br s, IH), 7.76 (m, 4H). MS (ESI+) m/z = 549.
Example 167 Synthesis of f7-(2,6-dichloro-phenyl)-5-methyl-benzo[l,2,4]triazin-3- yI]-[2-methyl-6-(2-pyrrolidin-l-yl-ethoxy)-pyrimidin-4-yl]-amine
Figure imgf000220_0002
(CCXX)
[0383] The synthesis of the title compound (CCXX) can be generally described by the sequence of reaction schemes (CCXXI) and (CCXXII). + N^
.N .1. N THF, reflux ^
NaO' CI^^^CI excess
(CCXXI)
[0384] 4,6-dichloro-2-methylpyrimidine (1 g, 6.1 mmol) was added into a freshly prepared sodium 2-(pyrrolidin-l-yl)ethoxide from 2-(pyrrolidin-l-yl)ethanol (0.35g, 3.1mmol) and sodium (0.08g, 3.5mmol) in anhydrous THF (10 mL) with stirring under argon. The mixture was refluxed under argon for 2h. The reaction mixture was cooled to room temperature and the solvent was removed under reduced pressure. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography with CHC13 as an eluent to give a pale yellow oil (540 mg, 73%). 1H NMR CDC13: δ 1.78 (m, 4H), 2.56 (s, 3H), 2.58 (m, 4H), 2.84 (t, J= 5.8 Hz, 2H), 4.48 (t, J= 5.8 Hz, 2H), 6.59 (s, IH).
[0385] The product of reaction shown by scheme (CCXXI) was further reacted as shown by scheme (CCXXII).
Figure imgf000221_0001
(CCXXII)
[0386] A mixture of 7-(2,6-dichlorophenyl)-5-methylbenzo[e] [ 1 ,2,4]triazin-3-amine (170 mg, 0.56 mmol), 4-(2-(pyrrolidin-l-yl)ethoxy)-6-chloro-2-methylpyrimidine (270 mg, 1.12 mmol), cesium carbonate (550 mg, 1.7 mmol), Xantphos (65 mg, 0.11 mmol), Pd2(dba) (51 mg, 0.056 mmol) in anhydrous dioxane (25 mL) was degassed with argon for 5 minutes and was refluxed for 1 h. The reaction mixture was cooled to room temperature and the solvent was removed under reduced pressure. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography with 10% CH3OH in CHC13 as an eluent to give the title compound (CCXX), as a yellow solid. (100 mg, 35%). 1H NMR DMSO-d6: δ 1.72 (br s, 4H), 2.6 (m, 7H), 2.92 (br s, 2H), 4.49(br s, 2H), 7.53 (t, J = 8.2 Hz, IH), 7.67 (s, IH), 7.69 (s, IH), 7.79 (s, IH), 7.83 (d, J= 1.9 Hz, IH), 8.23 8.23 (d, J= 1.8 Hz, IH), 11.59 (s, IH). MS(ESI+) m/z = 510.
Example 168 Synthesis of 4-chIoro-3-(5-methvI-3-r2-methyl-6-(2-pyrrolidin-l-yl- ethoxy)-pyrimidin-4-ylaminol-benzo[l,2,4]triazin-7-yl}-phenol
Figure imgf000222_0001
(CCXXIII)
[0387] The synthesis of the title compound (CCXXIII) can be generally described by the sequence of reaction schemes (CCXXIV) and (CCXXN).
Figure imgf000222_0002
(CCXXIV)
[0388] A mixture of 7-(2-chloro-5-methoxyphenyl)-5-methylbenzo[e] [ 1 ,2,4]triazin-3- amine (340 mg, 1.3 mmol), 4-(2-(pyrrolidin-l-yl)ethoxy)-6-chloro-2-methylpyrimidine (270 mg, 1.12 mmol), cesium carbonate (l.lg, 3.37 mmol), Xantphos (130 mg, 0.22 mmol), Pd2(dba)3 (100 mg, 0.11 mmol) in anhydrous dioxane (25mL) was degassed with argon for 5 minutes and was refluxed for 1 h. The reaction mixture was cooled to room temperature and the solvent was removed under reduced pressure. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography with 10% CH3OH in CHC13. The solid was washed with acetone/hexanes (1:5) to yield a yellow solid (300 mg, 53%). [0389] The product of reaction shown by scheme (CCXXIV) was further reacted as shown by scheme (CCXXV).
Figure imgf000223_0001
(CCXXV)
[0390] The N-(6-(2-(pyrrolidin- 1 -yl)ethoxy)-2-methylpyrimidin-4-yl)-7-(2-chloro-5- methoxyphenyl)-5-methylbenzo[e][l,2,4]triazin-3-amine (270 mg, 0.53 mmol) in chloroform (10 mL) was stirred with IM BBr3 in dichloromethane (5 mL, 5 mmol) at room temperature for 3 h. The reaction was quenched by methanol (1 mL). After removing the solvent, the crude product was neutralized with saturated sodium bicarbonate (1 x 25 mL). The crude product was extracted by ethyl acetate (2 x 50 mL). The combined ethyl acetate was dried over sodium sulfate. The salt was removed by filtration and the solvent was removed by vacuum. The crude product was purified by silica gel column chromatography with 20% CH3OH/CHCl3 as an eluent to afford the title compound (CCXXIII), as a yellow solid (100 mg, 38%). 1H NMR DMSO-d6: δ 1.68 (m, 4H), 2.48 (s, 3H), 2.53 (m, 4H), 2.65 (s, 3H), 2.81 (t, J= 6.0 Hz, 2H), 4.44 (t, J= 5.8 Hz, 2H), 6.88 (dd, J= 8.8 Hz, J= 2.9 Hz, IH), 6.94 (d, J= 2.9 Hz, IH), 7.40 (d, J= 8.8 Hz , IH), 7.73 (s, IH), 7.93 (s, IH), 8.24 (d, J= 1.6 Hz, IH), 9.95 (s, IH), 11.55 (s, IH). MS(ESI+) m/z = 492.
Example 169 Synthesis of 4-f7-(6-chloro-2-fluoro-3-hydroxy-phenyI)-5-methyl- benzo[l,2,41triazin-3-ylaminol-N-(2-pyrroIidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000224_0001
(CCXXVI)
[0391] The synthesis of the title compound (CCXXVI) can be generally described by the sequence of reaction schemes (CCXXVII), (CCXXVIII), (CCXXIX), and (CCXXX).
Figure imgf000224_0002
(CCXXVII)
[0392] 2.5M nBuLi (14.9mL, 37.4 mmol) was added dropwise over 5 minutes into a mixture of 4-chloro-2-fluoro-l-methoxybenzene (5g, 31.1 mmol) in anhydrous THF (30 mL) at -78°C. After the mixture was stirred at -78°C for 20 minutes, anhydrous trimethyl borate (4.9g, 46.6 mmol) was added into the solution at -78°C. The reaction mixture was brought to room temperature for over period of 2 h. The reaction was quenched by 2N HCI (1 mL). THF was removed by vacuum. The crude product was diluted with 2N HCI (100 mL). The acidic solution was extracted with ethyl acetate (2 x 50 mL). The combined ethyl acetate fraction was dried over sodium sulfate. The sodium sulfate was removed by filtration and the solvent was removed by vacuum. The oil was titrated with hexanes/chloroform (1:1) to yield a solid. The precipitate was collected by filtration and washed with hexanes to yield a white solid (2.5g, 39%). 1H NMR DMSO-d6: δ 3.81 (s, 3H), 7.12 (m, 2H), 8.66 (s, 2H). [0393] The product of reaction shown by scheme (CCXXVII) was further reacted as shown by scheme (CCXXNIII).
DME / EtOH / H20
Figure imgf000225_0001
(CCXXNIII)
[0394] A mixture of 7-bromo-5-methylbenzo[e][l,2,4]triazin-3-amine (420 mg, 1.8 mmol), 6-chloro-2-fluoro-3-methoxyphenylboronic acid (350 mg, 5.4 mmol), ethylene glycol dimethyl ether (20 mL), EtOH (2 mL), H2O (2 mL), Pd(PPh3)4 (200 mg, 0.17 mmol) and Νa CO3 (1.87 g, 18 mmol) was degassed with argon for 5 minutes and refluxed for 16 h under argon. The reaction mixture was cooled to room temperature, and the solid was removed by filtration. After evaporation of the volatiles, the crude product was purified by silica gel column (3.5 x 16 cm) chromatography with 5% CH3OH/CHCl3 as an eluent to give a solid. The solid was washed with chloroform to give a yellow solid (220 mg, 39%). 1H NMR DMSO-d6: δ 2.52 (s, 3H), 3.90 (s, 3H), 7.29 (t, J= 9 Hz, IH), 7.43 (dd, J= 9 Hz, J= 1.7 Hz, IH), 7.62 (s, IH), 7.78 (br s, 2H), 8.02 (d, J= 1.6 Hz, IH). MS(ESI+) m/z = 319.
[0395] The product of reaction shown by scheme (CCXXNIII) was further reacted as shown by scheme (CCXXIX).
Figure imgf000225_0002
(CCXXIX)
[0396] A mixture of 7-(6-chloro-2-fluoro-3-methoxyphenyl)-5- methylbenzo[e][l,2,4]triazin-3-amine (240 mg, 0.75 mmol), 4-bromo-Ν-(2-pyrrolidin-l- yl-ethyl)benzenesulfonamide (330 mg, 0.99 mmol), cesium carbonate (740 mg, 2.27 mmol), Xantphos (90 mg, 0.16 mmol), Pd2(dba)3 (70 mg, 0.08 mmol) in anhydrous dioxane (30mL) was degassed with argon for 5 minutes and was refluxed for 2 h. The reaction mixture was cooled to room temperature and the solvent was removed under reduced pressure. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography with 10% CH3OH in CHC13. The solid was washed with acetone/hexanes (2:1) to yield a yellow solid. (250 mg, 58%). 1H NMR DMSO-d6: δ 1.62 (m, 4H), 2.36 (br s, 4H), 2.43 (t, J= 7 Hz, 3H), 2.70 (s, 3H), 2.86 (t, J= 7 Hz, 2H), 3.92 (s, 3H), 7.32 (t, J= 9 Hz, IH), 7.42 (br s, IH), 7.46 (dd, J= 9 Hz, J= 1.8 Hz, IH), 7.84 (m, 3H), 8.21 (m, 3H), 11.43 (s, IH). MS(ESI+) m/z = 571.
[0397] The product of reaction shown by scheme (CCXXIX) was further reacted as shown by scheme (CCXXX).
Figure imgf000226_0001
(CCXXX)
[0398] A mixture of 4-[7-(6-chloro-2-fluoro-3-methoxy-phenyl)-5-methyl- benzo [ 1 ,2,4]triazin-3-ylamino] -N-(2-pyrrolidin- 1 -yl-ethyl)-benzenesulfonamide (170 mg, 0.53 mmol) in chloroform (5 mL) was stirred with IM BBr3 in dichloromethane (5 mL, 5 mmol) at room temperature for overnight. The reaction was quenched by methanol (1 mL). After removing the solvent, the crude product was neutralized with saturated sodium bicarbonate (1 x 25 mL). The crude product was extracted by ethyl acetate (2 x 50 mL). The combined ethyl acetate fraction was dried over sodium sulfate. The salt was removed by filtration and the solvent was removed by vacuum. The crude product was purified by silica gel column chromatography with 20% CH3OH/CHCl3 as an eluent to afford the title compound ((CCXXVI), as a yellow solid (lOmg, 6%). 1H NMR DMSO- de: δ 1.62 (m, 4H), 2.36 (br s, 4H), 2.44 (t, J= 7 Hz, 3H), 2.70 (s, 3H), 2.86 (t, J= 7 Hz, 2H), 7.08 (t, J= 9 Hz, 1H), 7.29 (dd, J= 9 Hz, J= 1.6 Hz, IH), 7.42 (br s, IH), 7.85 (m, 3H), 8.22 (m, 3H), 10.4 (br s, IH), 11.42 (s, IH). MS(ESI+) m/z = 557.
Example 170 Synthesis of 4-[7-(5-hydroxy-2-methyl-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamino1-N-(2-pyrrolidin-l-yl-ethyl)-benzenesuIfonamide
Figure imgf000227_0001
(CCXXXI)
[0399] The synthesis of the title compoimd (CCXXXI) can be generally described by the sequence of reaction schemes (CCXXXII), (CCXXXIII), (CCXXXIV), (CCXXXV), and (CCXXXVI).
Figure imgf000227_0002
(CCXXXII)
[0400] A solution of 5-methoxy-2-methylbenzenamine (1.3g, 9.48 mmol) in 48% HBr (13 mL) and EtOH (10 mL) was cooled to 0°C and NaNO2 (0.78g, 11.30 mmol) in water (5 mL) was added dropwise over 15 minutes while keeping the temperature of the reaction between 0-5°C. After the clear pale brown solution was kept at 0°C with stirring for lh, this solution was transferred into the boiling solution of CuBr (6.8g, 47.4 mmol) in 48% HBr (25 mL). The solution was refluxed for overnight. The reaction mixture was cooled to room temperature and the reaction was diluted with water (50 mL). The acidic solution was extracted with ethyl acetate (2 x 50 mL). The combined ethyl acetate was washed by saturated NaCI (1 x 50 mL) and was dried over sodium sulfate. The sodium sulfate was removed by filtration and the solvent was removed by vacuum. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography with hexanes as an eluent to yield colorless oil (750 mg, 39%).
[0401] The product of reaction shown by scheme (CCXXXII) was further reacted as shown by scheme (CCXXXIII).
Figure imgf000228_0001
(CCXXXIII)
[0402] 2.5M nBuLi (1.8 mL, 4.5 mmol) was added dropwise over 5 minutes into a mixture of 2-bromo-4-methoxy-l-methylbenzene (750mg, 3.73 mmol) in anhydrous THF (30 mL) at -78°C. After the mixture was stirred at -78°C for 20 minutes, anhydrous trimethyl borate (0.62g, 6 mmol) was added into the solution at -78°C. The reaction mixture was brought to room temperature for over period of 2 h. The reaction was quenched by 2N HCI (1 mL). The THF was removed by vacuum. The crude product was diluted with 2N HCI (100 mL). The acidic solution was extracted with ethyl acetate (2 x 50 mL). The combined ethyl acetate fraction was dried over sodium sulfate. The sodium sulfate was removed by filtration and the solvent was removed by vacuum to yield a pale yellow solid (0.43g, 69%).
[0403] The product of reaction shown by scheme (CCXXXIII) was further reacted as shown by scheme (CCXXXIN).
Figure imgf000228_0002
(CCXXXIN) [0404] A mixture of 7-bromo-5-methylbenzo[e] [ 1 ,2,4]triazin-3-amine (500 mg, 2.1 mmol), 5-methoxy-2-methylphenylboronic acid (430 mg, 2.6 mmol), ethylene glycol dimethyl ether (10 mL), EtOH (1 mL), H2O (1 mL), Pd(PPh3)4 (240 mg, 0.21 mmol) and Na2CO3 (2.2 g, 21 mmol) was degassed with argon for 5 minutes and refluxed for 16 h under argon. The reaction mixture was cooled to room temperature, and the solid was removed by filtration. After evaporation of the volatiles, the crude product was purified by silica gel column (3.5 x 16 cm) chromatography with 5% CH OH/CHCl3 as an eluent to give a solid. The solid was washed with chloroform to give a dark yellow solid (220 mg, 38%). 1H NMR DMSO-d6: δ 2.22 (s, 3H), 2.54 (s, 3H), 3.77 (s, 3H), 6.89 (s, IH), 6.90 (d, J= 3 Hz, IH), 7.24 (d, J= 8 Hz, IH), 7.66 (br s, IH), 7.68 (s, IH), 7.95 (d, J= 1.6Hz, IH). MS(ESI+) m/z = 281.
[0405] The product of reaction shown by scheme (CCXXIV) was further reacted as shown by scheme (CCXXXV).
Pd2(dba)3
Figure imgf000229_0001
(CCXXXV)
[0406] A mixture of 7-(5-methoxy-2-methylphenyl)-5-methylbenzo[e] [ 1 ,2,4]triazin-3- amine (220 mg, 0.79 mmol), 4-bromo-N-(2-pyriOlidin-l-yl-ethyl)benzenesulfonamide (340 mg, 1.0 mmol), cesium carbonate (770 mg, 2.36 mmol), Xantphos (90 mg, 0.16 mmol), Pd2(dba)3 (70 mg, 0.08 mmol) in anhydrous dioxane (30mL) was degassed with argon for 5 minutes and was refluxed for 2 h. The reaction mixture was cooled to room temperature and the solvent was removed under reduced pressure. The crude product was purified by silica gel column (3.5 x 16 cm) chromatography with 10%) CH3OH in CHC13. The solid was washed with acetone/hexanes (2:1) to yield a yellow solid. (150 mg, 36%).1HNMRDMSO-d6: δ 1.62 (m, 4H), 2.25(s, 3H), 2.36 (br s, 4H), 2.43 (t, J= 7 Hz, 2H), 2.71 (s, 3H), 2.86 (t, J= 7 Hz, 2H), 3.79 (s, 3H), 6.93 (m, 2H), 7.28 (d, J= 9 Hz, IH), 7.40 (br s, IH), 7.83 (d, J= 9Hz, 2H), 7.89 (d, J= 1.6Hz, IH), 8.15 (d, J= 9Hz, 2H), 11.35 (br s, IH).
[0407] The product of reaction shown by scheme (CCXXV) was further reacted as shown by scheme (CCXXXVI).
Figure imgf000230_0001
(CCXXXNI)
[0408] A mixture of 4-[7-(5-methoxy-2-methyl-phenyl)-5-methyl-benzo[l,2,4]triazin- 3-ylamino]-Ν-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (80 mg, 0.15 mmol) in chloroform (10 mL) was stirred with IM BBr3 in dichloromethane (10 mL, 10 mmol) at room temperature for overnight. The reaction was quenched by methanol (1 mL). After removing the solvent, the crude product was neutralized with saturated sodium bicarbonate (1 x 25 mL). The crude product was extracted by ethyl acetate (2 x 50 mL). The combined ethyl acetate was dried over sodium sulfate. The salt was removed by filtration and the solvent was removed by vacuum. The crude product was purified by silica gel column chromatography with 20% CH3OH/CHCl3 as an eluent to afford the title product (CCXXXI), as a yellow solid (20mg, 26%). 1H NMR DMSO-d6: δ 1.62 (m, 4H), 2.20 (s, 3H), 2.35 (br s, 4H), 2.43 (t, J= 7 Hz, 2H), 2.71 (s, 3H), 2.85 (t, J= 6.6 Hz, 2H), 6.75 (m, 2H), 7.15 (d, J= 9 Hz, J= 1.6 Hz, IH), 7.40 (br s, IH), 7.84 (m, 3H), 8.09 (d, J= 1.4Hz, IH), 8.21 (d, J= 9Hz, 2H), 9.38 (br s, IH), 11.34 (br s, IH). MS (ESI+) m/z = 519. Example 171 Synthesis of f7-(2'-hvdroxyethyl-isopropylamino)-6-methyl- benzori,2,41triazin-3-ylamino]-N-f2-pyrrolidin-l-yl-ethylVbenzene-4-sulfonamide
Figure imgf000231_0001
(CCXXXVII)
[0409] To a solution of 2-(isopropylamino)ethanol 48 μL, 0.416 mmol), 4-(7-bromo- 6-methyl-benzo[ 1 ,2,4]triazin-3-ylamino)-N-(2-pyrrolidin- 1 -yl-ethyl)- benzenesulfonamide (245 mg, 0.5 mmol) in dioxane was added xantphos (13 mg, 0.02 mmol) and cesium carbonate (202 mg, 0.62 mmol). The solution was degassed using argon, and Pd2(dba)3 a(l 1 mg, 0.012 mmol) was added. The reaction was refluxed overnight under argon, and quenched with 2 mL of water. The crude reaction mixture was evaporated on a rotoevaporator, diluted with water (15 mL) and extracted with chloroform (2X50 mL). The chloroform extract was dried over sodium sulfate, and the chloroform was evaporated. The residue was purified using prep HPLC products were isolated to afford the title product (CCXXXVII) as a yellow solid (3 mg, % yield). 1H NMR DMSO-d6: δ 1.05 (d, J = 6.2 Hz, 6H), 1.6-1.7 (m, 4H), 2.3-2.4 (m, 4H), 2.42 (s, 3H), 2.4-2.5 (m, 2H), 2.8-2.95 (m, 3H), 3.0-3.1 (m, 2H), 4.23-4.35 (m, 2H), 7.35-7.45 (br s, IH), 7.67 (s, IH), 7.7 (d, J = 0.9 Hz), 7.76 (d, J =8.95 Hz, 2H), 8.1 (d, J = 7 Hz, 2H), 10.94 (s, IH). MS (ESI+) m/z = 514.2.
Example 172 Synthesis of 4-(7-{[2-(2-hydroxy-ethoxy)-ethyl]-isopropyl-amino}-6-methyl- benzo[l,2,4]triazin-3-ylamino)-N-(2-pyrroIidin-l-yl-ethyl)-benzenesuIfonamide
Figure imgf000231_0002
(CCXXXVIII) [0410] The title product was synthesized and had the following characteristics. 1H NMR DMSO-d6: δ 1.0 (d, J = 6.6 Hz, 6H), 1.53-1.65 (m, 4H), 2.33-2.39 (m, 4 H), 2.35 (s, 3H), 2.42-2.47 (m, 2H), 2.58 (t, J = 6.7 Hz, 2H), 2.83 (t, J = 6.7 Hz, 2H), 2.90 (t, J = 5.6 Hz, 2H), 2.95-3.05 (m, IH), 3.42 (t, J = 6.6 Hz, 2H), 4.03 (br s, 2H), 4.19 (t, J = 5.6 Hz, 2H), 7.35-7.45 (br s, IH), 7.69 (s, IH), 7.7 (d, J = 0.9 Hz), 7.76 (d, J =8.95 Hz, 2H), 8.1 (d, J = 7.05 Hz, 2H), 10.93 (s, IH). MS (ESI+) m/z = 558.3.
Example 173 Synthesis of 4-(7-bromo-6-methyl-benzo[l,2,41triazin-3-ylamino)-N-(2- pyrrolidin-l-yl-ethvD-benzenesulfonamide
Figure imgf000232_0001
(CCXXXIX)
[0411] A mixture of amine 1 (2.5 g, 10.45 mmol), bromide 2 (4.18 g, 12.54 mmol), Cs2CO3 (5.1 g, 15.67 mmol), Xantphos (363 mg, 0.627 mmol), Pd2(dba)3 (286 mg, 0.31 mmol), and 3 A mol sieves in dioxane (70 mL) was purged with argon for 5 min, and was heated to reflux for 16 h under argon. Dioxane was removed in vacuo and the resulting mixture was partitioned between CHC13 and water (200 mL each). The layers were separated and the aqueous layer was extracted twice more with CHC13 (200 mL). The organic layers were combined and concentrated in vacuo. The crude product was purified by flash column chromatography (0.5 % NH4OH /10 % MeOH/89.5% dichloromethane).Orange-yellow solid (1.5 g, 30%). 1H NMR DMSO-d6: δ 1.82-1.92 (m, 2H), 1.95-2.05 (m, 2H), 2.59 (s, 3H), 2.96-3.08 (m, 4H), 3.2-3.28 (m, 2H), 3.53-3.61 (m, 2H), 7.83 (d, J = 8.8 Hz, 2H), 7.85 (s, IH), 7.87 (t, I = 6.1 Hz, IH), 8.18 (d, J = 8.8 Hz, IH), 8.68 (s, IH), 9.46-9.57 (br s, IH), 11.4 (s, IH). MS (ESI+) m/z = 493. Example 174 Synthesis of r6-methyl-7-(3-methyl-thiophen-2-ylVbenzo[1.2,41triazin- 3-ylH4-(2-pyrrolidin-l-vI-ethoxyVphenyl1-aιnine
Figure imgf000233_0001
(CCXL)
[0412] To a mixture of 3-methylthiophen-2-yl-2-boronic acid (252 mg, 2 mmol), 3- ammo-7-bromo-6-methyl benzo-l,2,3-triazine (239 mg, 1 mmol) and Pd(Ph3)4) (58 mg, 0.05 mmol) in 4:1 DME/EtOH was added a 2M aqueous solution of sodium carbonate (4 mL). The mixture was flushed with argon for 5 min and was heated under reflux (ca. 100 °C) for 16 h. The volatiles were evaporated and the residue was triturated with chloroform- water (1:1, 200 mL). The chloroform layer was separated and filtered through a small silica plug. The silica plug was washed with 200 mL of 10% methanol in chloroform. On evaporation the crude product was obtained as a brown solid (255 mg).
[0413] A mixture of the crude above amine product (125 mg, 0.5 mmol), l-(2-(4- bromophenoxy)ethyl)pyrrolidine (121 μL, 0.58 mmol), Cs2CO3 (638 mg, 1.96 mmol), Xantphos (28 mg, 0.049 mmol), Pd2(dba)3 (22 mg, 0.02 mmol), and 3 A mol sieves in dioxane (5 mL) was purged with argon for 5 min, and was heated to reflux for 16 h under argon. The volatiles were evaporated and the residue was triturated with chloroform-water (1:1, 200 mL). The chloroform layer was separated and filtered through a small silica plug. The silica plug was washed with 200 mL of 10% methanol in chloroform. The solvent was evaporated and the crude product was purified using HPLC to afford the title product (CCXL), as a red solid (29 mg, 13 %). !H NMR DMSO-d6: δ 1.67-1.76 (m, 4H), 2.06 (s, 3H), 2.31 (s, 3H), 2.54-2.6 (m, 4H), 2.81 (t, J = 5.57 Hz, 2H), 4.07 (t, J = 5.9 Hz, 2H), 6.95 (d, J = 6.95 Hz, 2H), 7.07 (d, J = 5.1 Hz, IH), 7.58 (d, J = 5.2 Hz, IH), 7.66 (s, IH), 7.85 (d, J = 6.95 Hz, 2H), 8.09 (s, IH), 10.71 (s, IH). MS (ESΪ+) m/z = 446.1. O 2005/096784
233
Example 175 Synthesis of {4-[6-methyl-7-(4-methyI-pyridin-3-vD benzo[l,2,4]triazin-3-ylamino]-phenyll-(4-methyl-p piippeerraazziinn--ll--yyll))--mmeetthhanone mesylate
Figure imgf000234_0001
(CCXLI)
[0414] To a mixture of 4-methylpyridin-3-yl-3-boronic acid (272 mg, 2 mmol), 3- amino-7-bromo-6-methyl benzo-l,2,3-triazine (239 mg, 1 mmol) and Pd(Ph3)4) (58 mg, 0.05 mmol) in 4:1 DME EtOH was added a 2M aqueous solution of sodium carbonate (4 mL). The mixture was flushed with argon for 5 min and was heated under reflux (ca. 100 °C) for 16 h. The volatiles were evaporated and the residue was triturated with chloroform-water (1:1, 200 mL). The chloroform layer was separated and filtered through a small silica plug. The silica plug was washed with 200 mL of 10% methanol in chloroform. On evaporation the crude product was obtained as a brown solid (260 mg).
[0415] A mixture of the above crude amine product (125 mg, 0.5 mmol), l-(2-(4- bromophenoxy)ethyl)pyrrolidine (170 mg, 0.6 mmol), Cs2CO3 (651 mg, 2.0 mmol), Xantphos (58 mg, 0.1 mmol), Pd2(dba)3 (45 mg, 0.05 mmol), and 3 A mol sieves in dioxane (5 mL) was purged with argon for 5 min, and was heated to reflux for 16 h under argon. The volatiles were evaporated and the residue was triturated with chloroform-water (1:1, 200 mL). The chloroform layer was separated and filtered through a small silica plug. The silica plug was washed with 200 mL of 10% methanol in chloroform. The solvent was evaporated and the crude product was purified using HPLC, to afford the title product (CCXXLI), as an orange solid (91 mg, 40.2 %). 1H NMR DMSO-d6: δ 2.16,2.22, 2.31, 2.84 (4s, 3H each), 3.05-3.18 (m, 4H), 3.45-3.55 (m, 4H), 7.53 (d, J = 8.6 Hz, 2H), 7.58 (d, J = 4.95 Hz, IH), 7.81 (s, 3H), 8.10 (d, J = 8.6 Hz, 2H), 8.17 (s, IH), 8.5 (s, IH), 8.62 (d, J = 4.5 Hz, IH), 9.67-9.76 (br s, IH), 11.15 (s, 3H). MS (ESI+) m/z = 454.3.
Example 176 Synthesis of {4-[7-(4-chloro-pyridin-3-yl)-6-methyl- benzo[l,2,4]triazin-3-ylamino]-phenyli-(4-methyl-piperazin-l-yl)-methanone mesylate
Figure imgf000235_0001
(CCXLII)
[0416] To a mixture of 4-chloropyridin-3-yl-3-boronic acid (314 mg, 2 mmol), 3- v amino-7-bromo-6-methyl benzo-l,2,3-triazine (239 mg, 1 mmol) and Pd(Ph3)4) (58 mg, 0.05 mmol) in 4:1 DME EtOH was added a 2M aqueous solution of sodium carbonate (4 mL). The mixture was flushed with argon for 5 min and was heated under reflux (ca. 100 °C) for 16 h. The volatiles were evaporated and the residue was triturated with chloroform-water (1:1, 200 mL). The chloroform layer was separated and filtered through a small silica plug. The silica plug was washed with 200 mL of 10% methanol in chloroform. On evaporation the crude product was obtained as a brown solid (260 mg).
[0417] A mixture of above crude amine product (125 mg, 0.46 mmol), l-(2-(4- bromophenoxy)ethyl) pyrrolidine (156 mg? 0.55 mmol), Cs2CO3 (600 mg, 1.84 mmol), Xantphos (53 mg, 0.1 mmol), Pd2(dba)3 (45 mg, 0.05 mmol), and 3 A mol sieves in dioxane (10 mL) was purged with argon for 5 min, and was heated to reflux for 16 h under argon. The volatiles were evaporated and the residue was triturated with chloroform-water (1:1, 200 mL). The chloroform layer was separated and filtered through a small silica plug. The silica plug was washed with 200 mL of 10% methanol in chloroform. The solvent was evaporated and the crude product was purified using HPLC. The purified product was converted to mesylate salt using methane sulfonic acid, in a form of a yellow solid (51 mg, 18 % yield). 1H NMR DMSO-d6: δ 2.30,2.44, 2.80 (3s, 3H each), 3.05-3.17 (m, 2H), 3.3-3.52 (m, 4H), 7.53 (d, J = 6.9 Hz, 2H), 7.68 (d, J = 8.5 Hz, IH), 7.78 (s, 3H), 8.07 (d, J = 2.65 Hz, 2H), 8.09 (d, J = 8.65, IH), 8.27 (s, IH), 8.59 (d, J = 2.4 Hz, IH), 9.67-9.76 (br s, IH), 11.17 (s, IH). MS (ESI+) m/z = 474.2.
Example 177 Synthesis of {4-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin- 3-ylaminol-phenv -piperazin-l-yI-methanone
Figure imgf000236_0001
(CCXLIΠ)
[0418] A mixture of 7-(2,6-dimethylphenyl)-3-amino-5-methyl- 1,2,3 -triazine (1.66, 6.29 mmol), 4-(4-bromo-benzoyl)-piperazine-l -carboxylic acid tert-butyl ester (2.79 g, 7.55 mmol), potassium t-butoxide (1.05 g, 9.43 mmol), BINAP (235 mg, 0.377 mmol), Pd2(dba)3 (172 mg, 0.18 mmol), and 3 A mol sieves in toluene (120 mL) was purged with argon for 5 min, and was heated to reflux for 16 h under argon. The volatiles were evaporated and the residue was triturated with chloroform- water (1:1, 500 mL). The chloroform layer was separated and filtered through a small silica plug. The silica plug was washed with 400 mL of 10% methanol in chloroform. The solvent was evaporated and the crude product was treated with trifluoroacetic acid (25 mL) in dichloromethane (25 mL) for 1.5 h. The solvent was evaporated and the crude product was purified by column chromatography using chloroform, methanol, ammonia (85:10:5). Some amout was converted to mesylate salt, in a form of a rusty red solid (1.51 g, 54 % yield). H NMR DMSO-d6: δ 2.05 (s, 6H), 2.69 (s, 3H), 2.69-2.75 (m, 4H), 3.37-3.57 (m, 4H), 7.19 (d, J = 7.6 Hz, 2H), 7.25 (dd, J = 6.2 and 8.6 Hz, IH), 7.45 (dd, J = 1.75 and 6.85 Hz, 2H), 7.64 (dd, J = 1 and 1.85 Hz, IH), 7.95 (d, J = 1.85 Hz, IH), 8.09 (dd, J = 1.7 and 6.85 Hz, 2H), 11.12 (s, IH). MS (ESI+) m/z = 453.3. Example 178 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methvI-benzo[l,2,4]triazin-3- yl] - [2-(2-p yrrolidin-1 -yl-ethoxy)-p yridin-4-yll -amine mesylate
Figure imgf000237_0001
(CCXLIV)
[0419] The title product wa synthesized, having the following characteristics. 1H NMR DMSO-d6: δ 2.85-2.95 (m, 2H), 1.99-2.08 (m, 2H), 2.05 (s, 3H), 2.29 (s, 3H), 2.71 (s, 3H), 3.07-3.2 (m, 2H), 3.56-3.7 (m, 4H), 4.6 (t, J = 5.2 Hz, 2H), 7.19 (d, J = 7.45 Hz, 2H), 7.25 (dd, J = 6.3 and 8.6 Hz, 2H), 7.61 (dd, J = 1.75 and 5.8 Hz, IH), 7.66 (d, J = 1.75 Hz, IH), 7.29 (d, J = 1.0 Hz, IH), 8.08 (d, J = 1.9 Hz, IH), 8.12 (d, J = 5.8 Hz, IH), 11.39 (s, IH). MS (ESI+) m/z = 455.3.
Example 179 Synthesis of 7-(2,6-dichloro-3-methoxyphenvD-5- methylb enzo [e] [1 ,2,4] triazin-3-amine
Figure imgf000237_0002
(CCXLV)
[0420] To prepare the title compound (CCXLV), an intermediate compoimd 96 (2,6- dichloro-3-methoxyphenylboronic acid) was synthesized first.
XOY xxcB(
CI ιOH)2
96 [0421] To synthesize compound 96, to 2,4-dichloroanisole (5 mL, 36 mmol) in THF (22 mL) at -78 °C was added 2.5 M nBuLi (17 mL, 43 mmol) keeping the temperature below -65 °C. After 30 min, the reaction was poured into frozen (-78 °C) trimethylborate (14 mL, 215 mmol) followed by another portion of trimethylborate (10 mL, 90 mmol) and allowed to warm to room temperature over 16 h. The solid suspension was poured into a 1 N HCI aqueous solution and stirred for lh. The suspension was filtered to give 3.6 g (46%) of a white solid contaminated most likely with B(OH)4. A second batch of crystals formed which were collected as an off-white solid that was compound 96 (5.8 g, 74%). 1H NMR DMSO-d6: δ 8.53 (s, 2H), 7.29 (d, J= 8.8 Hz, IH), 7.06 (d, J= 8.8 Hz, IH), 3.83 (s, 3H).
[0422] The general Suzuki procedure was then employed using compound 32 (Example 140) (794 mg, 3.32 mmol), compound 96 (816 mg, 3.70 mmol), Pd(Ph3)4 (386 mg, 0.33 mmol), and 2M aqueous Na2CO3 (7 mL, 14.0 mmol) in 4:1 DME/EtOH (34 mL) for 16 h to afford, after filtration, the title compound (CCXLV), as a cmde pale- orange solid (922 mg, 83%) which was used as is with no further purification. LC retention time: 2.98 min, MS (ESI+) m/z = 334.9.
Example 180 Synthesis of 4-[7-(2,6-dichloro-3-methoxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-yIamino1-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000238_0001
(CCXLVI)
[0423] The general Buchwald procedure was employed using crude compound (CCXLV) (Example 179) (305 mg, 0.91 mmol), compound 54 (Example 140) (304 mg, 0.91 mmol), Cs2CO3 (1.2 g, 3.62 mmol), Xantphos (104 mg, 0.18 mmol), Pd2(dba)3 (82 mg, 0.09 mmol), and 18 mL dioxane for 3 h. Chromatography was run at 0-10% gradient MeOH in CHCl3(with 0.1% NH4OH) to afford the title compound (CCXLVI), as a yellow-brown solid (373 mg, 63%). LC retention time: 2.69 min, MS (ESI+) m/z = 587.1.
Example 181 Synthesis of 4-[7-(2,6-dichloro-3-hydroxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-ylamino]-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide
Figure imgf000239_0001
(CCXLVΠ)
[0424] The general aryl methoxy deprotection procedure was employed using aryl methoxy compound (CCXLVI) (Example 180) (337 mg, 0.57 mmol) and BBr3 (271 μL, 2.9 mmol), in 15 mL DCM for lh. The reaction was quenched and the solids filtered and purified using HPLC. The aqueous HPLC fractions (100 mL) were neutralized with sat. aq. NaHCO3 and extracted with EtOAc (2x 150 mL). The combined organic layers were dried, filtered through celite and concentrated in vacuo. The HCI salt of the product was formed using MeOH and 4M HCI in dioxane, crashed out using Et2O, and collected by filtration as a yellow solid (204 mg, 58%). LC retention time: 2.34 min, MS (ESI+) m/z = 573.1 1H NMR DMSO-d6 :δ 1.80-1.87 (m, 2H), 1.92-1.99 (m, 2H), 2.71 (s, 3H), 2.93- 3.02 (m, 2H), 3.14 (q, J= 6.3 Hz, 2H), 3.18-3.24 (m, 2H), 3.47-3.55 (m, 2H), 7.17 (d, J= 8.9, Hz, IH), 7.43 (d, J= 8.9, Hz, IH), 7.75 (dd, J= 1.8, 1.0 Hz, IH), 7.89 (d, J= 8.9 Hz, 2H), 8.02 (t, J= 5.8 Hz IH), 8.11 (d, J= 1.7 Hz, IH), 8.26 (d, J= 8.9 Hz, 2H), 10.78 (br s, IH), 10.85 (s, IH), 11.47 (s, IH).
Example 182 Synthesis of 4-chIoro-3-[3-f4-f[(2-hydroxy-ethyl)-isopropyI-aminol- methvI}-phenylamino)-5-methyl-benzo[l,2.41triazin-7-vn-phenoI
Figure imgf000240_0001
(CCXLVΠI)
[0425] To synthesize the title product, intermediate compounds 97 and 98 shown below were used.
Figure imgf000240_0002
97
Figure imgf000240_0003
98
[0426] The general Buchwald procedure was employed using compounds 97 (142 mg, 0.50 mmol), 98 (104 mg, 0.38 mmol), Cs2CO3 (473 mg, 1.45 mmol), Xantphos (44 mg, 0.08 mmol), Pd2(dba)3 (34 mg, 0.04 mmol), and 7 mL dioxane for 2 h. The product was purified using HPLC to afford the TFA salt of the title compoimd (CCXLVIII), as a yellow-brown solid (45 mg, 21%). LC retention time: 2.32 min, MS (ESI+) m/z = 478.0 1H NMR DMSO-d6: δ 1.31 (d, J= 6.6, Hz, 3H), 1.37 (d, J= 6.6, Hz, IH), 2.69 (s, 3H), 3.01-3.11 (m, IH), 3.22-3.28 (m, IH), 3.47-3.54 (m, IH), 3.62-3.72 (m, 2H), 4.29 (dd, J = 13.2, 6.4, Hz, IH), 4.36 (dd, J= 13.2, 4.1, Hz, IH), 5.31 (br s, IH), 6.88 (dd, J= 8.7, 2.9, Hz, IH), 6.93 (d, J= 2.9 Hz, IH), 7.41 (d, J= 8.7, Hz, IH), 7.61 (d, J= 8.6 Hz, IH), 7.89 (dd, J= 1.8, 1.0 Hz, IH), 8.13 (d, J= 8.7 Hz, IH), 8.19 (d, J= 1.7 Hz, IH), 8.94 (br s, IH), 9.95 (s, IH), 11.17 (s, IH).
Example 183 Synthesis of N-(4-f2-(pyrrolidin-l-ynethoxy phenyl)-7-(2,6-dichloro-3- methoχyphenyl)-5-methylbenzo lei f 1.2,4] triazin-3-amine
Figure imgf000241_0001
(CCXLIX)
[0427] The general Buchwald procedure was employed using crude compound (CCXLV) (Example 179) (159 mg, 0.47 mmol), compound 6 (Example 120) (134 mg, 0.50 mmol), Cs2CO3 (628 mg, 1.93 mmol), Xantphos (56 mg, 0.10 mmol), Pd2(dba)3 (46 mg, 0.05 mmol), and 10 mL dioxane for 16 h. Chromatography was run at 0-10% gradient MeOH in CHCl3(with 0.1% NH4OH) followed by HPLC and NaHCO3 extraction to afford the title compound (CCXLIX), as a red solid (110 mg, 44%). LC retention time: 2.74 min, MS (ESI+) m/z = 524.1.
Example 184 Synthesis of 3-(3-(4-(2-(pyrrolidin-l-yl)ethoxy)phenylamino -5- methylbenzo[e][l,2,41triazin-7-yl)-2,4-dichlorophenol
Figure imgf000241_0002
(CCL)
[0428] The general aryl methoxy deprotection procedure was employed using aryl methoxy compound (CCXLIX) (Example 183) (110 mg, 0.21 mmol) and BBr3 (120 μL, 1.27 mmol), in 4 mL DCM for 20 min. The reaction was quenched and the solids filtered and purified using HPLC. The aqueous HPLC fractions (100 mL) were neutralized with sat. aq. NaHCO and extracted with EtOAc (2x 150 mL). The combined organic layers were dried, filtered through celite and concentrated in vacuo. The HCI salt of the product was formed using MeOH and 4M HCI in dioxane, crashed out using Et2O, and collected the title compound (CCL) by filtration, as an orange solid (28 mg, 24%). LC retention time: 2.34 min, MS(ESI+) m/z = 510.0 !H NMR DMSO-d6: δ 1.90 (br s, 2H), 2.03 (br s, 2H), 2.64 (s, 3H), 3.12-3.16 (m, 2H), 3.59 (br s, 4H), 4.34 (t, J= 5.0, Hz, 2H), 7.10 (d, J = 9.1, Hz, 2H), 7.10 (d, J= 8.9, Hz, IH), 7.42 (d, J= 8.9, Hz, IH), 7.65 (dd, J= 1.8, 1.0 Hz, IH), 7.99 (d, J= 9.1 Hz, 2H), 8.02 (d, J= 1.7 Hz, IH), 10.18 (br s, IH), 10.71 (s, IH), 10.90 (s, IH).
Example 185 Synthesis of benzyl 4-(4-(7-(2,6-dichloro-3-methoxyphenyl)-5- methylbenzo[e][l,2,4]triazin-3-ylamino)phenylsulfonyl)piperidine-l-carboxylate
Figure imgf000242_0001
(CCLI)
[0429] To synthesize the title product, intermediate compound 99 shown below was used.
Figure imgf000242_0002
99
[0430] The general Buchwald procedure was employed using crude compound (CCLV) (example 179) (114 mg, 0.34 mmol), intermediate compound 99 (100 mg, 0.23 mmol), Cs2CO3 (312 mg, 0.96 mmol), Xantphos (28 mg, 0.05 mmol), Pd2(dba)3 (21 mg, 0.02 mmol), and 5 mL dioxane for 3 h. Cliromatography was run at 0-50% gradient EtOAc in hexanes to afford the title compound (CCLI), as a crude yellow solid (104 mg, 65%). LC retention time: 3.37 min, MS(ESI+) m/z = 692.2.
Example 186 Synthesis of 3-(3-(4-(piperidin-4-ylsulfonyl)phenylamino)-5- methylbenzo[el[l,2,41triazin-7-yl)-2,4-dichlorophenol
Figure imgf000243_0001
(CCLII)
[0431] The general aryl methoxy deprotection procedure was employed using aryl methoxy compound (CCLI) (Example 185) (104 mg, 0.15 mmol) and BBr3 (85 μL, 0.90 mmol), in 3 mL DCM for 5 min. The reaction was quenched and the solids filtered and purified using HPLC. The aqueous HPLC fractions (100 mL) were neutralized with sat. aq. NaHCO3 and extracted with EtOAc (2x 150 mL). The combined organic layers were dried, filtered through celite and concentrated in vacuo. The HCI salt of the title product was formed using MeOH and 4M HCI in dioxane, crashed out using Et2O, and collected by filtration as a yellow solid (34 mg, 41%). LC retention time: 2.23 min, MS (ESI+) m/z = 544.0 1H NMR DMSO-d6: δ 1.67-1.78 (m, 2H), 2.03-2.09 (m, 2H), 2.72 (s, 3H), 2.83- 2.91 (m, 2H), 3.36 (m against H2O peak), 3.55 (tt, J= 11.9, 0.8, Hz, IH), 7.13 (d, J= 8.9, Hz, IH), 7.44 (d, J= 8.8, Hz, IH), 7.77 (s, IH), 7.89 (d, J= 9.0 Hz, 2H), 8.14 (d, J= 1.3 Hz, IH), 8.34 (d, J= 8.9 Hz, 2H), 8.43 (br s, IH), 8.96 (br s, IH), 10.75 (s, IH), 11.59 (s, IH). Example 187 Synthesis of (4 7-f3.5-dimethyl-isoxazol-4-ylV5-methyl- benzo[l,2,41triazin-3-ylamino1-phenyl}-(4-methyl-piperazin-l-yl)-methanone
Figure imgf000244_0001
(CCLIII)
[0432] A mixture of above amine (compound (CCLII), Example 186) (125 mg, 0.46 mmol), l-(2-(4-bromophenoxy)ethyl) pyrrolidine (156 mg, 0.55 mmol), Cs2CO3 (600 mg, 1.84 mmol), Xantphos (53 mg, 0.1 mmol), Pd2(dba)3 (45 mg, 0.05 mmol), and 3 A mol sieves in dioxane (10 mL) was purged with argon for 5 min, and was heated to reflux for 16 h under argon. The volatiles were evaporated and the residue was triturated with chloroform- water (1:1, 200 mL). The chloroform layer was separated and filtered through a small silica plug. The silica plug was washed with 200 mL of 10% methanol in chloroform. The solvent was evaporated and the crude product was purified using HPLC. The purified product was converted to mesylate salt using methane sulfonic acid, to afford the title product (CCLIII), as an orange solid (54 mg, % yield). 1H NMR DMSO-d6: δ 2.16,2.22, 2.31, 2.84 (4s, 3H each), 3.05-3.18, 3.24-3.36, 3.43-3.55 (3m, 8 H), 7.55 (d, J = 7 Hz, 2H), 7.89 (d, J = 1.8 Hz, IH), 8.13 (d, J = 7.0 Hz, 2H), 8.22 (d, J = 1.7 Hz, IH), 9.86-9.97 (br s, IH), 11.25 (s, IH). MS (ESI+) m/z = 458.2
[0433] The following Examples 188-192 describe the synthesis of some of the intermediate compounds 100 (7-bromo-5-methyl-benzo[l,2,4]triazin-3-ylamine), 101 (7- bromo-6-methyl-benzo[ 1 ,2,4]triazin-3-ylamine), 102 ((4-bromo-phenyl)-(4-methyl- piperazin-l-yl)-methanone), 103 (4-bromo-N-(2-pyrrolidin-l-yl-ethyl)-benzamide), 104 (4-bromo-N-(2-dimethylamino-ethyl)-benzamide), 105 (3-bromo-N-(2-pyrrolidin-l-yl- ethyl)-benzamide), 106 (3R-(3-bromo-benzoylamino)-pyrrolidine-l-carboxylic acid tert- butyl ester), 107 (l-[3-(3-bromo-benzenesulfonyl)-propyl]-pyrrolidine), and 108 (7-(2,6- dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine) that were used to synthesize the title compounds of Examples 188-190, 193-205, 208, 209, and 213 that follow.
Figure imgf000245_0001
100
Figure imgf000245_0002
104
Figure imgf000246_0001
105
Figure imgf000246_0002
106
Figure imgf000246_0003
107
Figure imgf000246_0004
108
Example 188 Synthesis of (4-bromo-phenyl)-(4-methyl-piperazin-l-yl)-mefhanone
(intermediate compound 102
[0434] To a solution of 4-bromo-benzoic acid (1.0 g, 5.0 mmol) in 50 mL acetonitrile and 5 mL DMF were added N-methylpiperazine (566 μl, 5.1 mmol) and EDC (980 mg, 5.1 mmol). The reaction mixture was stirred at room temperature for 16 h. The solvent was removed and the residue was dissolved in 60 mL CH2C12 and successively washed with water (50 mL) and aqueous saturated NaHCO3 solution. The organic phase was dried (MgSO4) and the solvent was removed. The crude product was purified by flash column chromatography (Rf = 0.54, CH2Cl /MeOH, 90:10) to yield the intermediate compoimd 102. 1HNMR CDC13: δ 2.32 (s, 3H), 2.33 (m, 2H), 2.49 (m, 2H), 3.41 (m, 2H), 3.78 (m, 2H), 7.27 (d, J= 8.3 Hz, 2H), 7.53 (d, J= 8.3 Hz, 2H). MS (ESI+) m/z = 283/285.
Example 189 Synthesis of 4-bromo-N-(2-dimethylamino-ethyl)-benzamide
(intermediate compound 104)
[0435] To a solution of 4-bromo-benzoic acid (5.02 g, 25 mmol) in 200 mL acetonitrile were added N'.N1 -dimethylethylenediamine (2.8 mL, 25 mmol) and EDC (4.9 g, 25.5 mmol). The mixture was stirred at room temperature for 2 h. The white precipitate was removed by filtration and the solvent was evaporated. The residue was dissolved in 150 mL CH2C1 , and was washed successively with water (150 mL) and aqueous saturated NaHCO3 (150 mL). The organic phase was dried (MgSO ) and the solvent was evaporated. The crude product was purified by flash column chromatography Rf = 0.2, (CH2Cl2/MeOH, 90:10) to afford the title intermediate compound 104 (3.68 g, 54%). MS (ESI+) m/z = 271/273.
Example 190 Synthesis of 3-bromo-N-(2-pyrrolidin-l-yl-ethyl)-benzamide
(intermediate compound 105)
[0436] The title compound was prepared according to the procedure described for the intermediate compound 104 (Example 189). The crude product was used without further purification (3.7 g, 50%). MS (ESI+) m/z = 297/299.
Example 191 Synthesis of 3R-(3-bromo-benzoyIamino)-pyrrolidine-l-carboxyIic acid tert-butyl ester (intermediate compound 106)
[0437] A solution of 3-bromobenzoic acid (4.0 g, 20 mmol) and EDC (3.83 g, 20 mmol) in 50 mL acetonitrile was added dropwise to a solution of (3R)-(+)-3- aminopyrrolidine (1.72 g, 20 mmol) in 100 mL acetonitrile. The reaction mixture was stirred at room temperature for 2 h and the solvent was removed. The residue was dissolved in 50 mL CH2C12 and washed with brine (50 mL). The organic phase was dried (MgSO ) and the solvent was removed. The crude product was purified by silica gel column chromatography (Rf = 0.22, CH2Cl2/MeOH, 75:15) to give 3-bromo-N- pyrrolidin-3-yl-benzamide as a colorless solid (2.0 g, 37 %). To a solution of the above precursor (1.14 g, 4.24 mmol) in 30 mL CH2C1 were added di-tert-butyldicarbonate (925 mg, 4.24 mmol) and triethylamine (596 μl, 4.24 mmol). The reaction mixture was stirred at room temperature for lh. The solvent was removed and the crude product was purified by silica gel column chromatography (Rf = 0.66, CH2Cl2/MeOH, 90:10) to afford the title intermediate compound 106, as a viscous, colorless oil (1.1 g, 70%). MS (ESI+) m/z = 369/371.
Example 192 Synthesis of l-[3-(3-bromo-benzenesulfonyl)-propyl]-pyrrolidine
(intermediate compound 107)
[0438] To a solution of 3-bromothiophenol (4.0 g, 21.2 mmol) in 50 mL methanol was added NaOMe (2.28 g, 42 mmol). The mixture was stirred at room temperature for lh and was added dropwise to 22 mL of 1,3-dibromopropane (42.5 g, 210 mmol) at room temperature. The reaction mixture was stirred at room temperature for 16 h and quenched with water (50 mL). The crude product was extracted with CH C12 (100 mL) and the combined organic phase was dried (MgSO4). The volatiles were removed under reduced pressure. The crude product in 150 mL CH2C12 was treated with 3-chloroperoxybenzoic acid (4.9 g, 20 mmol) at 0 °C for 1 h. Another batch of mCPBA (4.9 g, 20 mmol) was added and the stirring was continued for another 30 min at 0 °C before the mixture was allowed to warm to room temperature. The reaction mixture was diluted with CH2C12 and EtOAc (20 mL each) and washed twice with saturated aqueous NaHCO3 solution. The organic phase was dried (MgSO4) and the product was purified by silica gel column chromatography (R = 0.53, EtOAc/hexanes 50:50) to give l-bromo-3-(3-bromo-propane- l-sulfonyl)benzene as a colorless solid (5.47 g, 76 %).
[0439] A mixture of l-bromo-3-(3-bromo-propane-l-sulfonyl)benzene (5.47 g, 16.4 mmol Cs2CO3 (10.7 g, 32.8 mmol) and pyrrolidine (2.71 mL, 32.8 mmol) ) in 100 mL anhydrous 1.4-dioxane was stirred at room temperature for 16 h. The reaction mixture was quenched with saturated aqueous sodium bicarbonate (100 mL) and was extracted with CH2C12 (140 mL). The combined organic phase was dried (MgSO4), and the solvent was removed. The product was dried in vacuo to afford the title intermediate compound 107, as a brown oil (4.95 g, 91 %).
Example 193 Synthesis of [7-(5-chloro-2-methoxy-phenyI)-5-methyl- benzo[l,2,41triazin-3-yn-[4-(2-pyrrolidin-l-yl-ethoχy)-phenyl]-amine trifluoroacetate
Figure imgf000249_0001
(CCLIV)
[0440] To a solution of inteπnediate compound 100 (24 mg, 0.1 mmol) in 2 mL DMA were added 5-chloro-2-methoxyphenylboronic acid (75 mg, 0.4 mmol) in ethanol (500 μl), K2CO3 (2.8 mg, 0.02 mmol) in water (200 μl), PPh3 (3 mg, 0.01 mmol) and Pd (dba)3 (9 mg, 0.01 mmol). The reaction mixture was stirred at 80 °C for 16 h under argon, cooled to room temperature and poured into aqueous saturated NaHCO3 (50 mL). The product was extracted with CH2C12 (100 mL) and purified by reverse phase preparative HPLC using a gradient of acetonitrile/water containing 0.1 % TFA as the eluent. The combined product fractions were washed with aqueous saturated NaHCO solution. The product was extracted with CH2C12, the combined organic phase was dried (MgSO4) and the solvent was evaporated to give a precursor 7-(5-chloro-2-methoxy- phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (25.1 mg, 83 %). To a solution of the purified precursor in 4 mL anhydrous toluene were added l-[2-(4-bromo-phenoxy)- ethyl]-pyrrolidine (41 μl, 0.2 mmol), BINAP (3.7 mg, 6 μmol), Pd2(dba)3 (2.3 mg, 2.5 μmol) and KO-'Bu (9.3 mg, 0.083 mmol). Under argon, the reaction mixture was stirred at 100°C for 18 h. It was cooled to room temperature and poured into aqueous saturated NaCI solution. The product was extracted with CH2C12 and purified by reverse phase preparative HPLC to afford the title copound(CCLIV) as trifluoroacetate salt, in a form of a red solid (5 mg, 10 %). 1H NMR DMSO-d6: δ 1.90 (m, 2H), 2.05 (m, 2H), 2.64 (s, 3H), 3.15 (m, 2H), 3.61 (t, J= 5.0 Hz, 2H), 3.63 (m, 2H), 4.31 (t, J= 5.0 Hz, 2H), 7.09 (d, J= 9.0 Hz, 2H), 7.22 (d, J= 8.9 Hz, 2H), 7.47 (dd, J= 8.9 Hz, J= 2.6 Hz, IH), 7.52 (d, J= 2.6 Hz,lH), 7.93 (br s, IH), 7.99 (d, J= 9.0 Hz, 2H), 8.25 (br s, IH), 11.x (s, IH). MS (ESI+) m/z = 490.
Example 194 Synthesis of (4-[7-(2,6-dimethyl-phenyl)-5-methyI-benzo[l,2,4]triazin- 3-ylamino1-phenylH4-methyl-piperazin-l-yl)-methanone trifluoroacetate
Figure imgf000250_0001
(CCLV)
[0441] The compound was prepared from intermediate compounds 108 and 102, following the coupling procedure described above. The product was purified by reverse phase preparative HPLC to afford the title product in a form of trifluoroacetate, salt as a yellow-orange solid (yield, 56 %). 1H NMR methanol-d4: δ 2.09 (s, 6H), 2.76 (s, 3H), 2.98 (s, 3H), 3.30-3.50 (m, 8H), 7.16 (br d, J= 6.9 Hz, 2H), 7.21 (dd, J= 6.2 Hz, J= 8.7, IH), 7.59 (d, J= 8.7 Hz, 2H), 7.60 (br s, IH), 7.91 (br s, IH), 8.18 (d, J= 8.7 Hz), 11.12 (s, IH). MS (ESI+) m/z = 467.
Example 195 Synthesis of [7-(3,5-dimethyl-isoxazoI-4-yl)-5-methyl- benzo[l,2,41triazin-3-yll-[4-(2-pyrrolidin-l-yl-ethoxy)-phenyll-amine
Figure imgf000250_0002
(CCLNI) [0442] The synthesis of the title product followed the reaction scheme (CCLVπ).
Figure imgf000251_0001
(CCLVII)
[0443] 7-(3,5-dimethyl-isoxazol-4-yl)-5-methyl-benzo[l,2,4]triazin-3-ylamine (designated as compound 10 on scheme (CCLVII)) was prepared from the above described intermediate compound 100 and 3,5-dimethylisoxazole-4-boronic acid following the coupling procedure outlined above. A solution of the intermediate (160 mg, 0.627 mmol) and sulfamic acid (122 mg, 1.25 mmol) in methoxyaniline (1.2 g, 9.4 mmol) was stirred at 135 °C for 6 h to form [7-(3,5-dimethyl-isoxazol-4-yl)-5-methyl- benzo[l,2,4]triazin-3-yl]-(4-methoxy-phenyl)-amine (designated as compound 11 on scheme (CCLVII)), which was purified by silica gel column chromatography (Rf = 0.24, hexanes/EtOAc, 70:30) (24.2 mg, 11%). The purified precursor, dissolved in 4 mL CH C12, was treated with BBr3 (32 μl, 0.34 mmol) at room temperature for 2 h. The reaction mixture was diluted with CH2C12 (20 mL) and washed with aqueous saturated sodium thiosulphate.
[0444] The organic phase was dried (Na2SO4) and the solvent evaporated to yield 4- [7-(3,5-dimethyl-isoxazol-4-yl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]-phenol (designated as compound 12 on scheme (CCLVII)) (15 mg, 65%), to a solution of which (4.5 mg, 0.013 mmol) in 3 mL 1,4-dioxane were added Cs CO3 (21 mg, 0.065 mmol) and l-(2-chloro-ethyl)-pyrrolidine (4.4 mg, 0.026 mmol). The mixture was heated under reflux for 2h and the solvent was removed. The crude product was purified by silica gel column chromatography using CH2Cl2/methanol/NEt3 (90:10:0.1) as the mobile phase to afford the title product (CCLVI), as a red solid (2.6 mg, 45%).1H NMR methanol-d4: δ 1.89 (m, 4H), 2.34 (s, 3H), 2.50 (s, IH), 2.70 (s, 3H), 2.79 (m, 4H), 3.02 (t, J= 5.6 Hz, 2H), 4.18 (t, J= 5.6 Hz, 2H), 7.01 (d, J= 9.0 Hz, 2H), 7.69 (s, IH), 7.89 (d, J= 9.0 Hz, 2H), 8.02 (br s, IH). MS (ESI+) m/z = 445.
Example 196 Synthesis of 4-[7-(2,6-dimethyl-phenyl)-5-methyl-benzofl,2,4]triazin- 3-ylaminol-N-(2-pyrrolidm-l-yl-ethyl)-benzamide
Figure imgf000252_0001
(CCLVIII)
[0445] The compound was prepared from the intermediate compounds 108 and 103 following the above-described coupling procedure. The crude product was purified by reverse phase preparative HPLC to afford the title product (CCLVIII), as a yellow-orange solid. 1H NMR methanol-d4: δ 2.06 (m, 2H), 2.09 (s, 6H), 2.20 (m, 2H), 2.77 (s, 3H), 3.18 (m, 2H), 3.46 (t, J= 5.8 Hz, 2H), 3.78 (t, J= 5.8 Hz, 2H), 3.82 (m, 2H), 7.16 (br d, J = 6.9 Hz, 2H), 7.21 (dd, J= 6.2 Hz, J= 8.7, IH), 7.60 (br s, IH), 7.91 (br s, IH), 7.96 (d, J= 8.9 Hz), 8.16 (d, J= 8.9 Hz, 2H). MS (ESI+) m/z = 481.
Example 197 Synthesis of [7-(2-chloro-5-methoχy-phenyl)-5-methyl- benzo[l,2,41triazin-3-yll-[4-f2-pyrrolidin-l-yl-ethoxy)-phenyl]-amine trifluoroacetate
Figure imgf000253_0001
(CCLIX)
[0446] The product was prepared from the intermediate compound 100, 2-chloro-5- methoxyphenylboronic acid, and l-[2-(4-bromo-phenoxy)-ethyl]-pyrrolidine following the above-described coupling procedures. The crude product was purified by reverse phase preparative HPLC to afford the title product (CCLIX) in a form of trifluoroacetate salt, as an orange solid (100 mg, 10 %).1H NMR methanol-d4: δ 2.08 (m, 2H), 2.22 (m, 2H), 2.70 (s, 3H), 3.27 (m, 2H), 3.68 (t, J= 5.0 Hz, 2H), 3.75 (m, 2H), 4.37 (t, J= 5.0 Hz, 2H), 7.00 (dd, J= 8.8 Hz, J= 3.0, IH), 7.05 (d, J= 3.0, IH), 7.10 (d, J= 9.0 Hz, 2H), 7.46 (d, J= 8.8 Hz, IH), 7.81 (br s,lH), 7.96 (d, J= 9.0 Hz, IH), 8.11 (br s, IH). MS (ESI+) m/z = 490.
Example 198 Synthesis of N-(2-dimethylamino-ethyl)-4-[7-(2,6-dimethyl-phenyl)-5- methyl-benzo[l,2,41triazin-3-yIamino]-benzamide dihvdrate mesylate
Figure imgf000253_0002
(CCLX)
[0447] The compound was prepared from the intermediate compounds 108 and 104 following the above-described coupling procedure. The crude product was purified by silica gel column chromatography using methanol/CFI2Cl2/NEt3 (50:50:0.1) as the mobile phase and was re-crystallized from CH2C1 to give N-(2-dimethylamino-ethyl)-4-[7-(2,6- dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]-beι z--mide, to a solution of 80 mg of which in 1.5 mL methanol was added methanesulfonic acid (9.5 μl, 0.146 mmol) and the mixture was stirred for 5 min until clear. The clear solution was added dropwise to 20 mL Et O. The yellow precipitate was isolated by filtration and dried in vacuo to afford the title product (CCLX), as a yellow solid (71 mg, 80%)^ NMR Methanol-d : δ 2.09 (s, 6H), 2.70 (s, 3H), 2.77 (s, 3H), 3.00 (s, 6H), 3.39 (t, J= 5.9 Hz, 2H), 3.78 (t, J= 5.9 Hz, 2H), 7.16 (br d, J= 7.9 Hz, 2H), 7.21 (dd, J= 6.2 Hz, J= 8.8, IH), 7.60 (br s, IH), 7.91 (br s, IH), 7.97 (d, J= 8.8 Hz), 8.17 (d, J= 8.8 Hz, 2H). Analyzed: (C28H38N6O6S), calculated: C, 57.32, H, 6.53, N, 14.32, found C, 57.00, H, 6.55, N, 14.30. MS (ESI+) m/z = 455.
Example 199 Synthesis of 3-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,41triazin- 3-ylamino1-N-(2-pyrroIidin-l-yI-ethyI)-benzamide trifluoroacetate
Figure imgf000254_0001
(CCLXI)
[0448] The compound was prepared from the intermediate compounds 108 and 104 following the above-described coupling procedure. The crude product was purified by reverse phase preparative HPLC to afford the title product (CCLXI), as a yellow solid (140 mg, 31 %). 1H NMRmethanol-d4: δ 2.05 (m, 2H), 2.09 (s, 6H), 2.21 (m, 2H), 2.78 (s, 3H), 3.18 (m, 2H), 3.47 (t, J= 5.9 Hz, 2H), 3.80 (t, J= 5.9 Hz, 2H), 3.83 (m, 2H), 7.16 (br d, J= 7.5 Hz, 2H), 7.21 (dd, J= 6.2 Hz, J= 8.7, IH), 7.53 (t, J= 7.8 Hz, IH), 7.56 (d, J= 7.8, IH), 7.58 (br s, IH), 7.89 (br s, IH), 8.15 (d, J= 7.8 Hz, IH), 8.70 (br s, IH). MS (ESI+) m/z = 481. Example 200 Synthesis of 3-[7-(2,6-dimethyI-phenyl)-5-methyl-benzofl,2,4]triazin- 3-ylamino]-N-pyrrolidin-3-yI-benzamide trifluoroacetate
Figure imgf000255_0001
(CCLXII)
[0449] To a solution of intermediate compound 108 (66 mg, 0.25 mmol) in 4 mL anhydrous toluene were added 7 (92 mg, 0.25 mmol), BINAP (16 mg, 25 μmol), Pd (dba)3 (12 mg, 12.5 μmol) and Cs2CO3 (122 mg, 0.375 mmol). The reaction mixture was stirred at 90 °C for 18 h under argon. The mixture was heated in the microwave at 170 °C for 10 min and poured into aqueous saturated NaCI solution. The product was extracted with CH2C12 and purified by reverse phase preparative HPLC to afford the title product (CCLXII), as a yellow solid (45 mg, 32%).1H NMR DMSO-d6: δ 2.01 (m, IH), 2.06 (s, 6H), 2.24 (m, IH), 2.67 (s, 3H), 3.53-3.95 (m, 5H), 7.19 (br d, J= 7.6 Hz, 2H), 7.24 (dd, J= 6.2 Hz, J= 8.7, IH), 7.50 (br t, J= 7.8 Hz, IH), 7.65 (br s, IH), 7.96 (br s, IH), 8.07 (br s, IH), 8.17 (d, J= 8.7 Hz, 2H), 11.07 (s, IH). MS (ESI+) m/z = 453.
Example 201 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[l,2,4]triazin-3- vn-f3-(3-pyrrolidin-l-yl-propane-l-sulfonyl)-phenvn-amine monohydrate mesylate
Figure imgf000255_0002
(CCLXIII)
[0450] The compound was prepared from the intermediate compounds 108 and 107 following the above-described coupling procedures. The crude product was purified by reverse phase preparative HPLC to give [7-(2,6-dimethyl-phenyl)-5-methyl- benzo[l,2,4]triazin-3-yl]-[3-(3-pyrrolidin-l-yl-propane-l-sulfonyl)-phenyl]-amine trifluoroacetate salt, which was converted to the free amine by washing with saturated aqueous NaHCO2 solution and extraction with CH2C12. The combined organic phases were dried (MgSO ) and the solvent was removed to yield [7-(2,6-dimethyl-phenyl)-5- methyl-benzo[ 1 ,2,4]triazin-3-yl] -[3 -(3-pyrrolidin- 1 -yl-propane- 1 -sulfonyl)-phenyl] - amine as a yellow solid, which (132 mg, 0.256 mmol) was combined with methanesulfonic acid (15.78 ul, 0.24 mmol) in 3 mL CH2C12 and with Et2O (80 mL). The yellow precipitate was isolated by filtration and dried in vacuo to afford the title product (CCLXIII), as a yellow solid (145 mg, 93%).1H NMR DMSO-d6: δ 1.82 (m, 2H), 1.98 (m, 4H), 2.06 (s, 6H), 2.30 (s, 3H), 2.72 (s, 3H), 2.97 (m, 2H), 3.23 (m, 2H), 3.45 (t, J= 7.8 Hz, 2H), 3.51 (m, 2H), 7.19 (d, J= 7.0 Hz, 2H), 7.24 (dd, J= 6.3 Hz, J= 8.6, IH), 7.60 (d, J= 8.0 Hz, IH), 7.68 (s, IH), 7.72 (t, J= 8.0 Hz, IH), 8.00 (s, IH), 8.13 (d, 8.0 Hz, IH), 9.04 (s, IH), 9.38 (br s, IH), 11.40 (s, IH). Analyzed: (C30H39N5O6S2), calculated C, 57.21, H, 6.24, N, 11.12, found C, 56.56, H, 5.79, N, 11.04 MS (ESI+) m/z = 516.
Example 202 Synthesis of {4-[7-(2-chloro-5-methoxy-phenyl)-6-methyI- benzori,2,41triazin-3-ylamino]-phenyll-(4-methyl-piperazin-l-yl)-methanone monohydrate mesylate
Figure imgf000256_0001
(CCLXIV)
[0451] The title product was synthesized from the inteπnediate compounds 101, 102, and 2-chloro-5-methoxyphenylboronic acid, following the above-described coupling procedures. The crude product was purified by reverse phase preparative HPLC and the resulting trifluoroacetate salt was converted to the free amine by washing with saturated aqueous NaHCO3 solution and extraction with CH2C12 to give {4-[7-(2-chloro-5- methoxy-phenyl)-6-methyl-benzo[l,2,4]triazin-3-ylamino]-ρhenyl}-(4-methyl-piperazin- l-yl)-methanone as a yellow solid (49.5%). The free base was converted to the title product (CCLXIV) (189 mg, 96 %). 1H NMR DMSO-d6: δ 2.29 (s, 3H), 2.31 (s, 3H, mesylate), 2.85 (br s, 3H), 3.11 (m, 2H), 3.30-3.50 (m, 6H), 3.85 (s, 3H), 7.03 (d, J= 3.0 Hz, IH), 7.09 (dd, J= 8.9, J= 3.0 Hz, IH), 7.52-7.55 (m, 3H), 7.76 (s, IH), 8.09 (d, J= 8.9 Hz, 2H), 8.10 (s, IH), 9.72 (br s, IH), 11.13 (s, IH). Anal. (C28H33ClN6O6S), Calcd C, 54.50, H, 5.39, N, 13.62, found 54.31, H, 5.64, N, 13.38 MS(ESI+) m/z = 503.
Example 203 Synthesis of {4-[7-(5-chloro-2-methyl-phenyl)-6-methyl- benzofl,2,4]triazin-3-yIamino]-phenyl}-(4-methyl-piperazin-l-yl)-methanone monohydrate mesylate
Figure imgf000257_0001
(CCLXV)
[0452] The title product was synthesized and purified as described in Example 202 for compound (CCLXIV), except 5-chloro-2-methylphenylboronic acid was used instead of 2-chloro-5-methoxyphenylboronic acid. 1H NMR DMSO-d6: δ 2.04 (s, 3H), 2.21 (s, 3H), 2.32 (s, 3H, mesylate), 2.84 (br s, 3H), 3.11 (m, 2H), 3.30-3.50 (m, 6H), 7.31 (d, J= 2.0 Hz, IH), 7.41-7.45 (m, 2H), 7.52 (d, J= 8.6 Hz, 2H), 7.78 (s, IH), 8.08 (s, IH), 8.09 (d, J = 8.6 Hz, 2H), 9.75 (br s, IH), 11.12 (s, IH). Analyzed; (C28H33ClN6O5S), calculated: C, 55.95, H, 5.53, N, 13.98, found: C, 55.59, H, 5.57, N, 13.78. MS (ESI+) m/z = 487. Example 204 Synthesis of [7-(5-fluoro-2-methyl-phenyl)-6-methyl- benzo[l,2,4]triazin-3-yll-r4-(2-pyrrolidin-l-yI-ethoxy)-phenyn-amine trifluoroacetate
Figure imgf000258_0001
(CCLXVI)
[0453] A precursor, 7-(5-fluoro-2-methyl-phenyl)-6-methyl-benzo[l,2,4]triazin-3- ylamine, was prepared using the above-described coupling procedure. A mixture of the precursor (113 mg, 0.42 mmol), l-[2-(4-bromo-phenoxy)-ethyl] -pyrrolidine (174 μl 0.84 mmol), Xantphos (49 mg, 0.084 mmol), Pd2(dba)3 (39 mg, 0.042 mmol) and Cs2CO3 (274 mg, 0.84 mmol) in anhydrous dioxane (4 mL) was heated under reflux for 2 h under argon. The reaction mixture was cooled to room temperature and quenched with brine. The product was extracted with CH C12 and purified by reverse phase preparative HPLC to afford the title product (CCXVI), as an orange solid (10.3 mg, 4.3%). 1H NMR DMSO-d6: δ 1.90 (m, 2H), 2.02 (s, 3H), 2.03 (m, 2H), 2.19 (s, 3H), 3.15 (m, 2H), 3.61 (m, 4H), 4.31 (t, J= 5.0, 2H), 7.07 (d, J= 9.0 Hz, IH), 7.10 (dd, J= 9 Hz, J= 2.8 Hz, IH), 7.21 (ddd, J= 8.7 Hz, J= 8.7 Hz, J= 2.8 Hz, IH), 7.41 (dd, J= 6 Hz, J= 8.7, IH), 7.67 (s, IH), 7.91 (d, J= 9.0 Hz, 2H), 8.00 (s, IH), 9.77 (br s, IH), 10.73 (s, IH). MS (ESI+) m/z = 458.
Example 205 Synthesis of 3-{6-methyl-3-[4-(2-pyrrolidin-l-yl-ethoxy)- phenylamino]-benzo[l,2,4]triazin-7-yl}-phenol hydrochloride
Figure imgf000258_0002
(CCLXVII) [0454] A precursor, 3-(3-amino-6-methyl-benzo[l,2,4]triazin-7-yl)-phenol, was prepared from the intermediate compound 101 and 3-hydroxyphenylboronic acid using the above-described coupling. The final product was synthesized from the above precursor (0.73 mmol, 185 mg) and l-[2-(4-bromo-phenoxy)-ethyl]-pyrrolidine (303 μl, 1.46 mmol) according to the procedure described above. The product was purified by reverse phase preparative HPLC and was converted to the free amine by washing with saturated aqueous NaHCO3 solution and extraction with CH2C12 (94.6 mg, 29 %). The free amine was converted to the title copound (CCLXNII), a red-orange solid (81.3 mg, 80 %). 1H ΝMR DMSO-d6: δ 1.90 (m, 2H); 2.04 (m, 2H), 2.40 (s, 3H), 3.13 (m, 2H), 3.59 (m, 4H), 4.34 (t, J= 5.0 Hz, 2H), 6.84 (s, IH), 6.84-6.88 (m, 2H), 7.06 (d, J= 9.0 Hz, 2H), 7.30 (dd, J= 7.8 Hz, J= 7.8 Hz, IH), 7.63 (s, IH), 7.90 (d, J= 9.0 Hz, 2H), 9.64 (s, OH), 10.39 (br s, IH), 10.70 (s, IH). MS(ESI+) m/z = 442.
Example 206 Synthesis of Ν-{3-[4-(2-pyrrolidin-l-yl-ethylsulfamoyl)-phenyIamino]- 7-o-tolyl-benzo[l,2,4]triazin-6-yl}-acetaιnide trifluoroacetate
(CCLXVIII)
[0455] As a preliminary step, the synthesis of the title product included a process shown by the reaction scheme (CCLXIX).
Figure imgf000260_0001
13 14
Figure imgf000260_0002
(CCLXIX)
[0456] The reaction scheme (CCLXIX) illustrates a synthetic process rthat included dropwise adding of 6.4 g bromine (40 mmol) to a solution of 4-nitrobenzene-l,3-diamine (5.56 g, 36.3 mmol), in 50 mL 1,4-dioxane, followed by stirring the solution was room temperature for 16 h. The yellow precipitate was isolated by filtration, washed with 1,4- dioxane (30 mL) and dried in vacuo to give 4-bromo-6-nitro-benzene-l,3-diamine dihydrobromide salt designated on the reaction scheme (CCLXIX) as compound 13 (8 g, 95%).
[0457] A mixture of compound 13 (6.0 g, 15.3 mmol) and cyanamide (3.9 g, 92 mmol) was heated to a melt at 90 °C. To the melt was cautiously added concentrated HCI (45 mL) and the reaction mixture was stirred at 95 °C for 2h. It was cooled to room temperature and the reaction mixture was brought to pH 12 with 30% sodium hydroxide (about 65 mL). The mixture was heated to 90 °C for 2h, cooled to room temperature and poured into water (1L). The yellow precipitate was isolated by filtration, washed with water (100 mL) and acetone (20 mL) and was dried to give 7-bromo-l-oxy- benzo[l,2,4]triazine-3,6-diamine designated on the reaction scheme (CCLXIX) as compound 14 (1.3 g, 33 %). To a suspension of the crude product in 140 mL MeOH were added Raney Ni and the mixture was hydrogenated for 2h. Solids were removed by filtration through a short silica gel column. The solvent was evaporated to give 7-bromo- benzo[l,2,4]triazine-3,6-diamine designated on the reaction scheme (CCLXIX) as compound 15 (1.0 g, 82%). To a solution of compoimd 15 (1.0 g, 4 mmol) in 60 mL DMA were added 2-methylphenylboronic acid (884 mg, 6.5 mmol) in 16 mL ethanol, 2CO3 (332 mg, 2.4 mmol) in 6 mL water, PPh3 (262 mg, 1 mmol) and Pd2(dba)3 (184 mg, 0.2 mmol). The reaction mixture was stirred for 4 h at 100 °C under argon, cooled to room temperature and poured into aqueous saturated NaHCO3 solution (100 mL). The product was extracted with CH2C12 (150 mL) and purified by reverse phase preparative HPLC. The product was washed with saturated aqueous NaHCO3 solution and extracted with EtOAc (100 mL). The combined organic phases were dried (Na2SO4) and the solvent removed. The crude product was re-crystallized from MeOH to give 7-o-tolyl- benzo[l,2,4]triazine-3,6-diamine designated on the reaction scheme (CCLXIX) as compound 16 (133.7 mg, 13 %).
[0458] To a solution of compound 16 (80 mg, 0.32 mmol) in 10 mL 1,4-dioxane was added potassium bis(trimethylsilyl)amide (127 mg, 0.64 mmol) and the mixture was stirred at room temperature under argon for lh. To the dark red solution was added acetylchloride (45 μl, 0.64 mmol) and stirring was continued for 30 min at room temperature. The product was poured into saturated aqueous NaHCO3 solution (40 mL) and was extracted with EtOAc (100 mL). The combined organic phases were dried (Na2SO4) and the solvent was evaporated. The crude product was purified by reverse phase preparative HPLC to give a precursor, N-(3-amino-7-o-tolyl-benzo[l,2,4]triazin-6- yl)-acetamide, as trifluoroacetate salt, which was converted to the free base (53.1 mg, 57 %) by washing with saturated aqueous NaHCO3 solution. The final product was synthesized from the purified precursor and 4-bromo-N-(2-pyrrolidin-l-yl-ethyl)- benzenesulfonamide following the above-described coupling procedure. The purification using reverse phase HPLC afforded the title product (CCLXVIII), as a yellow solid (81.7 mg, 68 %). 1H NMR DMSO-d6: δ 1.87 (m, 2H); 2.01 (m, 2H), 2.02 (s, 3H), 2.11 (s, 3H), 3.04 (m, 2H), 3.07 (td, J= 6.1 Hz, J= 6.1 Hz, 2H), 3.25 (td, J= 6.1 Hz, J= 6.1 Hz, 2H), 3.57 (m, 2H), 7.30 (d, J= 7.8 Hz, IH), 7.36-7.44 (m, 3H), 7.85 (m, IH), 7.86 (d, J= 9.0 Hz, 2H), 8.06 (s, IH), 8.20 (d, J= 9.0 Hz, 2H), 8.41 (s, IH), 8.91 (s, IH), 9.59 (br s, IH), 11.27 (s, IH). MS (ESI+) m/z = 546. Example 207 Synthesis of 4-[6-(3 ert-Butyl-ureido)-7-o olyl-ben;- o[l,2τ4]triazin-3- ylamino -N-(2-pyrrolidιn-l-yl-ethyI)-benzenesulfonamide trifluoroacetate
Figure imgf000262_0001
(CCLXX)
[0459] To a solution of 7-o-tølyl-benzo[l,2,4]triazine-3,6-diamme (compound 16 on scheme (CCLXIX)) (0.12 mmol, 30 mg) in 5 mL anhydrous 1,4-dioxane was added potassium bis(trimethylsilyl)amide (53 mg, 0.26 mmol) and the mixture was stirred at room temperature under argon for 1.5 h. To the red solution was added tert- butylisocyanate (28.5 μl, 0.36 mmol) and stirring was continued for 2 h at room temperature. The product was poured into saturated aqueous NaHCO3 solution (40 mL) and was extracted with EtOAc (100 mL). The combined organic phases were dried (Na2SO ) and the solvent was evaporated to give l-(3-amino-7~o-tolyl-benzo[l,2,4] triazin-6-yl)-3-tert-butyl-urea (28 mg, 67%). The title product was prepared as a red solid (27.8 mg, 48 %) from the above intermediate and 4-bromo-N-(2-pyrrolidin-l-yl- ethyl)-benzenesulfonamide following the above-described coupling procedure and was purified by reverse phase preparative HPLC. 1HNMRDMSO-d6: δ 1.37 (s, 9H), 1.87 (m, 2H); 2.01 ( , 2H), 2.16 (s, 3H), 3.04 (m, 4H), 3.24 (td, J= 6.1 Hz, J= 6.1 Hz, 2H), 3.56 (ms 2H), 7.21 (s, IH), 7.33-7.40 (m, 4H), 7.50 (d, J= 9.0 Hz, 2H), 7.75 (d, J= 9.0 Hz, 2H), 7.88 (t, J= 6.1 Hz, IH), 7.99 (s, IH), 8.34 (s, IH), 8.83 (s, IH), 9.53 (br s, IH), 10.21 (s, IH). MS (ESI+) m/z = 603. Example 208 Synthesis of 4-[7-(5-hvdroχymethyl-thiophen-2-yl)-6-methyl- benzo[l,2,41triazin-3-ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide trifluoroacetate
Figure imgf000263_0001
(CCLXXI)
[0460] A precursor, [5-(3-amino-6-methyl-benzo[l,2,4]triazin-7-yl)-thiophen-2-yl]- methanol, was prepared from the intermediate compound 101 and 5- hydroxymethylthiophene-2-boronic acid according to the above-described coupling procedure. The final compound was synthesized from the precursor and 4-bromo-N-(2- pyrrolidin-l-yl-ethyl)-benzenesulfonamide according according to the above-described coupling procedure. The product was purified by reverse phase preparative HPLC to afford the title product (CCLXXI), as an orange solid. 1H NMR DMSO-d6: δ 1.86 (m, 2H); 2.00 (m, 2H), 2.63 (s, 3H), 3.00-3.07 (m, 4H), 3.24 (m, 2H), 3.56 (m, 2H), 4.71 (br s, 2H), 5.60 (m, IH), 7.07 (d, J= 3.5 Hz, IH), 7.27 (d, 3.5 Hz, IH), 7.80 (s, IH), 7.84 (d, J= 9.0 Hz, 2H), 7.88 (t, 6.1 Hz, IH), 8.20 (d, J= 9.0 Hz, 2H), 8.30 (s, IH), 9.60 (br s, IH), 11.35 (s, IH). MS (ESI+) m/z = 525.
Example 209 Synthesis of 4-[7-(5-cvano-2-methyl-phenyl)-6-methyl- benzofl,2,4]triazin-3-ylamino1-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide trifluoroacetate
Figure imgf000263_0002
(CCLXXII) [0461] A precursor, 3-(3-amino-6-methyl-benzo[l,2,4]triazin-7-yl)-4-methyl- benzonitrile, was prepared from the intermediate compound 101 and 2-methyl-5- cyanoboronic acid according to the above-described coupling procedure. The crude product was purified by silica gel column chromatography (Rf = 0.2, EtOAc/hexanes/NEt3, 60:40:0.5). The product was synthesized from the precursor and 4- bromo-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide according to the above-described coupling procedure. The product was purified by reverse phase preparative HPLC to afford the title product (CCLXXII), as an orange solid (42 mg, 50 %). 1H NMR DMSO- d6: δ 1.87 (m, 2H); 2.01 (m, 2H), 2.15 (s, 3H), 2.21 (s, 3H), 3.00-3.08 (m, 4H), 3.25 (td, J = 6.1 Hz, J= 6.1 Hz, 2H), 3.57 (m, 2H), 7.62 (d, J= 8.0 Hz, IH), 7.75 (s, IH), 7.82-7.88 (m, 4H), 8.15 (s, IH), 8.21 (d, J= 9.0 Hz, 2H), 9.54 (br s, IH), 11.34 (s, IH). MS (ESI+) m/z = 528
Example 210 Synthesis of 4-chloro-3-[5-methyl-3-(4-pyrrolidin-l-yl-butylamino)- benzo[l,2,4]triazin-7-yl]-phenol trifluoroacetate salt
Figure imgf000264_0001
(CCLXXIII)
[0462] A suspension of 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[l,2,4]triazin- 3-ylamine (80 mg, 0.27 mmol) and sulfamic acid (66 mg, 0.68 mmol) in 3 mL 4- pyrrolidinobutyl amine was stirred at 200 °C for 16 h. The mixture was diluted with EtOAc (5 mL) and purified by silica gel column chromatography (Rf = 0.22, CH2Cl2/MeOH/NEt3, 90:10:0.1) (40 mg, 35%). The purified intermediate (40 mg, 0.094 mmol) was treated with BBr3 (80.3 μl, 0.47 mmol) at room temperature for 2h and was poured into saturated aqueous sodium thiosulphate. The product was extracted with 20 mL CH2Cl2/methanol (90:10). The combined organic phases were dried (Na2SO4), and the solvent was evaporated. The crude product was purified by reverse phase preparative HPLC to afford the title product (CCLXXIII), as a red solid (43 mg, 87%). 1H NMR DMSO-d6: δ 1.68-1.79 (m, 4H), 1.87 (m, 2H); 2.00 (m, IH), 2.09 (m, IH), 2.56 (s, 3H), 2.97-3.03 (m, 2H), 3.18 (m, 2H), 3.40-3.55 (m, 4H), 6.85 (dd, J= 8.6 Hz, J= 2.9 Hz, IH), 6.89 (d, J= 2.9 Hz, IH), 7.38 (d, J= 8.6 Hz, IH), 7.73 (s, IH), 8.02 (s, IH), 8.60 (br s, IH), 9.50 (br s, IH), 9.92 (br s, IH). MS(ESI+) m/z = 412.
Example 211 Synthesis of 4-[7-(3-hydroxy-piperidin-l-yl)-6-methyl- benzo[l,2,4]triazin-3-ylaminol-N-(2-pyrroIidin-l-yl-ethyl)-benzenesulfonamide trifluoroacetate
Figure imgf000265_0001
(CCLXXIV)
[0463] To a solution of 4-(7-bromo-6-methyl-benzo[ 1 ,2,4]triazin-3-ylamino)-N-(2- pyrrohdin-l-yl-ethyl)-benzenesulfonamide (71 mg, 0.145 mmol) in 2 mL anhydrous xylene were added piperidin-3-ol hydrochloride (80 mg, 0.58 mmol), xantphos (34 mg, 0.058 mmol), Pd(OAc)2 (7 mg, 0.029 mmol) and KOtBu (98 mg, 0.87 mmol). The reaction mixture was heated in a microwave at 150°C for 20 min and poured into aqueous saturated NaHCO3 solution. The product was extracted with CH2C12 (50 mL) and purified by reverse phase preparative HPLC to afford the title product (CCLXIV), as a brown solid (7.5 mg, 8.3%). 1H NMR DMSO-d6: δ 1.86 (m, 4H); 2.00 (m, 4H), 2.50 (s, 3H), 3.00-3.08 (m, 4H), 3.20-3.30 (m, 4H), 3.55 (m, 4H), 4.90 (m, IH), 7.67 (s, IH), 7.73 (s, IH), 7.80 (d, J= 8.9 Hz, 2H), 8.15 (d, J= 8.9 Hz, 2H), 9.50 (br s, 2H), 11.06 (s, IH). MS(ESI+) m z = 512. Example 212 Synthesis of 4-chloro-3-{6-chloro-3-[4-(2-pyrrolidin-l-yl-ethoxy)- phenylamino1-benzo[l,2,4]triazin-7-vU-phenol
Figure imgf000266_0001
(CCLXXV)
[0464] As a preliminary step, the synthesis of the title product included a process shown by the reaction scheme (CCLXXVI).
Figure imgf000266_0002
17
Figure imgf000266_0003
(CCLXXVI)
[0465] To a melt of 5-chloro-2-nitroaniline (5 g, 29 mmol) and cyanamide (9.8 g, 231 mmol) at 100°C was cautiously added concentrated HCI (50 mL) and the mixture was stirred at 100°C for 2.5 h. The reaction mixture was cooled to room temperature and the pH was adjusted to pH 11 using 30 % aqueous NaOH. The reaction mixture was stirred at 100 °C for 2 h, cooled to room temperature and poured into water (200 mL). The yellow precipitate was isolated by filtration, washed with CH C12 (20 mL) and dried to give 6- chloro-l-oxy-benzo[l,2,4]triazin-3-ylamine, designated on the reaction scheme (CCLXXVI) as compound 17 (3.42 g, 60%). To a solution of compound 17 (983 mg, 5 mmol) in 30 mL glacial acetic acid was added NBS (1.78 g, 10 mmol) and the mixture was heated to reflux for 4 h. It was cooled to room temperature and poured into water (700 mL). The yellow precipitate was isolated by filtration and was hydrogenated in MeOH (100 mL) for 2 h using Raney Ni (1.0 g). Solids were removed by filtration and the solvent was evaporated. The crude product was purified by silica gel column chromatography using EtOAc as the mobile phase to give 7-bromo-6-chloro- benzo[l,2,4]triazin-3-ylamine, designated on the reaction scheme (CCLXXVI) as compound 18 (95.6 mg, 7.4%).
[0466] 6-chloro-7-(2-chloro-5-methoxy-phenyl)-benzo[l,2,4]triazin-3-ylamine designated on the reaction scheme (CCLXXVI) as compound 19 (54 mg, 68%) was prepared from compound 18 and 2-chloro-5-methoxyphenylboronic acid according to the above-described coupling procedure. The crude product was purified by silica gel column chromatography (Rf = 0.41, EtOAc/hexanes 50:50). The title compound (CCLXXV) was prepared from compound 19 and l-[2-(4-bromo-phenoxy)-ethyl]-pyrrolidine following the above-described coupling procedure, followed by demethylation. The coupling product was purified by reverse phase preparative HPLC to give [6-chloro-7-(2-chloro-5- methoxy-phenyl)-benzo[ 1 ,2,4]triazin-3-yl]-[4-(2-pyrrolidin- 1 -yl-ethoxy)-phenyl] -amine (28.7 mg, 27%). Deprotection of the methoxy group with BBr3 (22 μl, 0.23 mmol) in 2 mL CH C12 at room temperature yielded 4-chloro-3-{6-chloro-3-[4-(2-pyrrolidin-l-yl- ethoxy)-phenylamino]-benzo[l,2,4]triazin-7-yl} -phenol, which was purified by reverse phase preparative HPLC and converted to the free amine to give a brown solid (13 mg, 57%). 1H NMR DMSO-d6: δ 1.68 (m, 4H), 2.50-2.55 (m, 4H), 2.79 (t, J= 5.9 Hz, 2H), 4.07 (t, J= 5.9, 2H), 7.07 (d, J= 9.0 Hz, IH), 6.77 (d, J= 2.8, IH), 6.84 (dd, J= 8.8 Hz, J= 2.8 Hz, IH), 6.98 (d, J= 9.0 Hz, 2H), 7.32 (d, J= 8.8, IH), 7.85 (d, J= 9.0 Hz, 2H), 7.90 (s, IH), 8.22 (s, IH), 10.90 (br s, IH). MS(ESI+) m/z = 496.
Example 213 Synthesis of 4-[7-(5-cyano-2-methyl-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide trifluoroacetate
Figure imgf000268_0001
(CCLXXNII)
[0467] 3-(3-amino-5-methyl-benzo[l,2,4]triazin-7-yl)-4-methyl-benzonitrile was prepared from the intermediate compound 100 and 2-methyl-3-cyanophenylboronic acid according to the above-described coupling procedure. The crude product was purified by silica gel column chromatography (Rf = 0.35, EtOAc/hexanes, 50:50) (72.4 mg, 77%). The product was purified and reacted with 4-bromo-Ν-(2-pyrrolidin-l-yl-ethyl)- benzenesulfonamide following the above-described coupling procedure. The crude product was purified by reverse phase preparative HPLC to afford the title product (CCLXXNII), as a yellow solid (30 mg, 18%). 1H ΝMR DMSO-d6: δ 1.87 (m, 2H); 2.01 (m, 2H), 2.41 (s, 3H), 2.72 (s, 3H), 3.04 (m, 2H), 3.07 (td, J= 6.2 Hz, J= 6.2 Hz, 2H), 3.25 (td, J= 6.2 Hz, J= 6.2 Hz, 2H), 3.57 (m, 2H), 7.61 (d, J= 8.2 Hz, IH), 7.81-7.87 (m, 2H), 7.88 (d, J= 8.9 Hz, 2H), 7.94 (br s, IH), 8.26 (d, J= 8.9 Hz, 2H), 8.27 (br s, IH), 9.63 (br s, IH), 11.46 (s, IH). MS (ESI+) m/z = 528.
Example 214 Synthesis of {4-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl- benzo[l,2,41triazin-3-ylaminol-phenyl}-(l,l-dioxo-116-thiomorpholin-4-yl)- methanone
Figure imgf000268_0002
(ccLxxviπ) [0468] To prepare the title compound, the intermediate compound 109 ((4-bromo- phenyl)-(l,l-dioxo-116-thiomorpholin-4-yl)-methanone) shown below was synthesized first.
Figure imgf000269_0001
109
[0469] To prepare the intermediate compound 109, a solution of 4-bromo-benzoic acid (2.0 g, 10.0 mmol) in 20 mL acetonitrile were added thiomorpholine (950 μl, 10 mmol) and EDC (1.92 g, 10 mmol). The reaction mixture was stirred at room temperature for 16 h. The solvent was removed and the residue was dissolved in 60 mL CH2C12 and washed with aqueous saturated NaHCO3 solution (100 mL). The organic phase was dried (Na2SO4) and the solvent was removed. The crude product was purified by flash column chromatography (Rf = 0.45, EtOAc/hexanes, 50:50). To a solution of the above intermediate (100 mg, 0.35 mmol) in CH2C12 (30 mL) were added mCPBA (79 mg, 0.35 mmol) at 0° C. The mixture was stirred for 2 h before another batch of mCPBA (79 mg, 0.35 mmol) was added. The mixture was stirred at RT for 16 h and was then quenched with aqueous saturated NaHCO3 solution (100 mL). The inteπnediate compound 109 was extracted with CH2C1 (100 mL) and used without further purification (105 mg, 94 %). 1H NMR DMSO-d6: δ 3.24 (m, 4H), 3.67 (m, 2H), 3.99 (m, 2H), 7.46 (d, J= 8.2 Hz, 2H), 7.68 (d, J= 8.2 Hz, 2H). MS(ESI+) m/z = 182/184.
[0470] A mixture of 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo [ 1 ,2,4]triazin-3- ylamine (50 mg, 0.17 mmol), intermediate compound 109 (80 mg, 0.25 mmol), Xantphos (20 mg, 0.0334 mmol), Pd2(dba)3 (16 mg, 0.0167 mmol) and Cs2CO3 (109 mg, 0.334 mmol) in anhydrous dioxane (4 mL) was refluxed for 4 h under argon. The reaction mixture was cooled to room temperature, diluted with CH C12 (20 mL) and filtered. The crude product was purified by flash column chromatography to give {4-[7-(2-chloro-5- methoxy-phenyl)-5-methyl-benzo[ 1 ,2,4]triazin-3-ylamino]-phenyl} -(1 , 1 -dioxo- 116- thiomorpholin-4-yl)-methanone, an amount of which (54 mg, 0.1 mmol) in CH2C12 (8 mL) was treated with a 1.0 M solution of BBr3 in CH2C12 (500 μl, 0.5 mmol) at 0° C to give {4-[7-(2-chloro-5-hydroxy-ρhenyl)-5-methyl-benzo[l,2,4]triazin-3-ylamino]- phenyl}-(l,l-dioxo-116-thiomorpholin-4-yl)-methanone. The crude product was purified by reverse phase preparative HPLC to afford the title compound (CCLXXVIII), as a yellow powder (15.7 mg, 30 %). 1H NMR DMSO-d6: δ 2.70 (s, 3H), 3.26-3.30 (m, 4H), 3.90 (m, 4H), 6.88 (dd, J= 8.7 Hz, J= 2.9 Hz, IH), 6.93 (d, J= 2.9 Hz, IH), 7.41 (d, J= 8.7 Hz, IH), 7.58 (d, J= 8.7 Hz, 2H), 7.89 (br s, IH), 8.12 (d, J= 8.7 Hz, 2H), 8.19 (br s, IH), 9.93 (s, IH), 11.22 (s, IH). MS(ESI+) m/z = 524.
Example 215 Synthesis of [7-(2,6-dichloro-phenyI)-5-methyI-benzofl,2,4]triazin-3- ylH4-(3-pyrrolidin-l-yl-propane-l-sulfonyl)-phenyl]-amme
Figure imgf000270_0001
(CCLXXIX)
[0471] The title compound was prepared from 7-(2,6-dichloro-phenyl)-5-methyl- benzo[l,2,4]triazin-3-ylamine and the intermediate compound 109 following the above- described coupling procedure, followed by purification by reverse phase preparative HPLC, to afford a yellow solid (62 mg, 31 %). 1H NMR DMSO-d6: δ 1.84 (m, 2H), 1.99 (m, 4H), 2.72 (s, 3H), 2.95 (m, 2H), 3.20 (m, 2H), 3.40-3.53 (m, 4H), 7.53 (t, J= 8.2 Hz, IH), 7.68 (d, J= 8.2 Hz, IH), 7.80 (br s, IH), 7.95 (d, J= 8.9 Hz, 2H), 8.20 (br s, IH), 8.31 (d, J= 8.9 Hz, 2H), 10.25 (br s, IH), 11.57 (s, IH). MS(ESI+) m/z = 556. Example 216 Synthesis of 4-f7-(2-chloro-6-fluoro-5-hvdroxyphenyl)-5-methyl- benzo[l,2,41triazin-3-ylamino]-N-methyl-N-(2-pyrroIidine-l-yl- ethyDbenzenesulfonamide
Figure imgf000271_0001
(CCLXXX)
[0472] Standard Buchwald conditions were used, followed by demethylation using BBr3. Yellow solid (17 mg, 18 %). 1H NMR (DMSO-d6): δ 1.85-1.95 (m, 2H), 1.97-2.09 (m, 2H), 2.72, 2.73 (2s, 3H each), 3.05-3.15 (m, 2H), 3.25-3.3, 3.37-3.45, 3.6-3.68 (3m, 2H each), 7.08 (t, J= 8.97 Hz, IH), 7.29 (dd, J= 1.5 and 9.0 Hz, IH), 7.85 (s, IH), 7.88 (d, J = 9.0 Hz, 2H), 8.24 (d, J= 1.5 Hz, IH), 8.31 (d, J= 9.0 Hz, 2H), 9.55 (br s, IH), 10.40 (s, IH), 11.5 (s, IH). MS (ESI+): m/z = 446.1.
Example 217 Testing of Inhibition of Kinases In Vitro
[0473] The ability of compounds of the present invention to inhibit the activity of three groups of kinases was tested. Kinases tested included the src family (primarily src and yes), some angio genie growth factor receptors (Fgfrl, Vegfr, and Pdgfrβ) and the ephrin, EphB4. All lcinase reactions were conducted in 96-well plates with a final reaction volume of 50 ul.
Src Family
[0474] Recombinant human c-Src or Yes (28 ng/well, Panvera/ϊnvitrogen, Madison WI), ATP (3 μM), a tyrosine kinase substrate (PTK2, 250 μM, Promega Corp., Madison WI), and test agents (at concentrations ranging from about 1 nM/1 to about 100 μM/1), in the presence of Src kinase reaction buffer (Upstate USA, Lake Placid NY). After reacting for about 90 minutes at room temperature, residual ATP was determined using a luciferase-based assay (KinaseGlo, Promega Corp.) as a measure of kinase activity. Data from four wells were then averaged and used to determine IC50 values for the test compounds (Prism software package, GraphPad Software, San Diego CA).
Growth Factor Receptors
[0475] Pdgfrβ (0.16ug/well, Panvera/hivitrogen) 500 nM ATP and the PTK2 peptide (700uM) were combined with compoimd and reaction buffer as noted above for src. The reaction was incubated for 60 minutes at 37C, and the residual ATP concentration was determined using the luciferase-based technique also noted above.
[0476] FGFR1 and NEGFR2 kinase assays were similarly performed. FGFR1 (76ng/well, Panvera/frivitrogen) was combined with 12.5mg/ml poly(glu4tyr) (Sigma) and 2.5uM ATP. NEGFR2 (14.1U/well, Cell Signaling/ProQinase) was used with 0.3mg/ml poly(glu4tyr) and 1.5uM ATP. Both were incubated for 60 minutes at 37C, and the residual ATP was measured via luminescence, per the procedure described above.
EphB4
[0477] EphB4 kinase activity was similarly measured, using the luciferase-based technique described above. 28.9mU/well EphB4 (Upstate) was reacted with lmg/ml poly(glu4tyr), 6uM ATP and test reagents. The reaction was incubated for 60 minutes at 37C and the residual ATP concentration was measured.
[0478] The test results for inhibition of Src kinase are presented in Table 1, and the test results for inhibition of some other kinases (i.e., Yes, EphB4, and Pdgfrβ) are presented in Table 2. The abbreviation "IC50" means that a particular compound of the invention, when present at the specified concentration, inhibited the kinase by 50%.
Table 1. Tests Results of Inhibition of Src Kinase by Some Compounds of the Invention
Figure imgf000273_0001
Figure imgf000274_0001
Figure imgf000275_0001
Figure imgf000276_0001
Figure imgf000277_0001
Figure imgf000278_0001
Figure imgf000279_0001
Figure imgf000280_0001
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
Figure imgf000287_0001
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000291_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0001
4-[7-(2-fluoro-6-hydroxy- 123 phenyl)-5-methyl- benzo[ 1 ,2,4]triazin-3-ylamino]-
Figure imgf000312_0001
N-(2-pyrrolidin- 1 -yl-ethyl)- benzenesulfonamide
4-[7-(2-chloro-5-hydroxy- 3.7 phenyl)-6-methyl- benzo[ 1 ,2,4]triazin-3-ylaniino]-
Figure imgf000312_0002
N-methyl-N-(2-pyrrolidin-l -yl- ethyl)-benzenesulfonamide
4-[7-(2-chloro-5-hydroxy- 1.6 phenyl)-5 -methyl- benzo[ 1 ,2,4]triazin-3-ylamino]-
Figure imgf000312_0003
N-methyl-N-(2-pyrrolidin- 1 -yl- ethyl)-benzamide
2,4-dichloro-3-{5-methyl-3-[4- 1.1
(2-pyrrolidin- 1 -yl-ethoxy)- phenylamino]-
Figure imgf000312_0004
benzo[ 1 ,2,4]triazin-7-yl} -phenol
4-chloro-2-fluoro-3-{5-methyl- 1.0 3-[4-(3-pyrrolidin-l-yl-propane- 1 -sulfonyl)-phenylamino]-
Figure imgf000312_0005
benzo[l,2,4]triazin-7-yl}-phenol
4- [7-(2 ,6-difluoro-3-hydroxy- 3.1 phenyl)-5 -methyl- benzo[l ,2,4]triazin-3-ylamino]-
Figure imgf000312_0006
N-(2-pyrrolidin- 1 -yl-ethyl)- benzenesulfonamide 2,4-dichloro-3-{5-methyl-3-[4- 2.4
(piperidine-4-sulfonyl)- phenylamino]-
Figure imgf000313_0001
benzo[l ,2,4]triazin-7-yl} -phenol
Acetic acid 4-chloro-3-{5- 53.2 methyl-3-[4-(2-pyrrolidin- 1 -yl- ethoxy)-phenylamino] -
Figure imgf000313_0002
benzo[ 1 ,2,4]triazin-7-yl} -phenyl ester
4-[7-(5-hydroxy-2-methyl- 0.955 phenyl)-5-methyl- benzo[ 1 ,2,4]triazin-3-ylamino]-
Figure imgf000313_0003
N-(2-pyrrolidin- 1 -yl-ethyl)- benzenesulfonamide
Benzoic acid 4-chloro-3-{5- 79.2 methyl-3-[4-(2-pyπOlidin- 1 -yl- ethylsulfamoyl)-phenylamino]-
Figure imgf000313_0004
benzo[ 1 ,2,4]triazin-7-yl} -phenyl ester
Benzoic acid 4-chloro-3-{5- > 10000 methyl-3-[4-(2-pyrrolidin- 1 -yl- ethoxy)-phenylamino]-
Figure imgf000313_0005
benzo[ 1 ,2,4]triazin-7-yl} -phenyl ester
2,4-dichloro-3-{5-methyl-3-[4- 2.1 (3 -pyrrolidin- 1 -yl-propane- 1 - sulfonyl)-phenylamino]-
Figure imgf000313_0006
benzo[ 1 ,2,4]triazin-7-yl} -phenol 2,4-difluoro-3- {5-methyl-3-[4- 3.8 (3-pyrrolidin- 1 -yl-propane- 1 - sulfonyl)-phenylamino]-
Figure imgf000314_0001
benzo[l ,2,4]triazin-7-yl} -phenol
4-[7-(6-chloro-2-fluoro-3- 1.2 hydroxy-phenyl)-5 -methyl- benzo[ 1 ,2,4]triazin-3-ylamino]-
Figure imgf000314_0002
N-methyl-N-(2-pyrrolidin- 1 -yl- ethyl)-benzenesulfonamide
4- [7-(6-chloro-2 -fluoro-3- 0.542 hydroxy-phenyl)-5 -methyl- benzo[ 1 ,2,4]triazin-3-ylamino]-
Figure imgf000314_0003
N-methyl-N-(2-pyrrolidin- 1 -yl- ethyl)-benzamide
[7-(2,6-dichloro-phenyl)-5- 90.3 methyl-1-oxy- benzofl ,2,4]triazin-3-yl]-[4-(2-
Figure imgf000314_0004
pyrrolidin- 1 -yl-ethoxy)-phenyl] - amine
Acetic acid 4-chloro-3-{5- 133 methyl-3-[3-(2-pyrrolidin- 1 -yl- ethoxy)-phenylamino] -
Figure imgf000314_0005
benzo[l,2,4]triazin-7-yl}-phenyl ester
4-methyl-benzoic acid 4-chloro- 267 3-{5-methyl-3-[4-(2-pyrrolidin- 1 -yl-ethoxy)-phenylamino]-
Figure imgf000314_0006
benzo[l ,2,4]triazin-7-yl} -phenyl ester 2,2-dimethyl-propionic acid 4- > 10000 chloro-3- {5-methyl-3-[4-(2- pyrrolidin- 1 -yl-ethoxy)-
Figure imgf000315_0001
phenylamino]- benzo[ 1 ,2,4]triazin-7-yl} -phenyl ester
Isobutyric acid 4-chloro-3-{5- 10000 methyl-3-[4-(2-pyrrolidin- 1 -yl- ethoxy)-phenylamino]-
Figure imgf000315_0002
benzo[ 1 ,2,4]triazin-7-yl} -phenyl ester
4-[7-(2,6-dichloro-3-hydroxy- 3.6 phenyl)-5-methyl- benzo[ 1 ,2,4]triazin-3-ylamino]-
Figure imgf000315_0003
N-methyl-N-(2-pyrrolidin- 1 -yl- ethyl)-benzenesulfonamide
Table 2. Tests Results of Inhibition of Selected Kinases by Some Compounds of the
Invention All data represent IC50 in nM.
Figure imgf000315_0004
Figure imgf000316_0001
Figure imgf000317_0001
Figure imgf000318_0001
Figure imgf000319_0001
Figure imgf000320_0001
Figure imgf000321_0001
Figure imgf000322_0001
Figure imgf000323_0001
Figure imgf000324_0001
Figure imgf000325_0001
Figure imgf000326_0001
[0479] Although the invention has been described with reference to the above examples, it will be understood that modifications and variations are encompassed witliin the spirit and scope of the invention. Accordingly, the invention is limited only by the following claims.

Claims

WHAT IS CLAIMED IS:
1. A compound of structure (I):
Figure imgf000327_0001
(I)
wherein each of A is independently selected from a group consisting of (CH)o-ι, N, NH, O, S, and a part of a ring fusion to form a second ring, where the second ring is an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic;
each of B is independently selected from a group consisting of (CH)o-ι, N, NH, O, S, and a part of a ring fusion to form a second ring, where the second ring is an aromatic, a heteroaromatic, a bicyclic aromatic, a bicyclic aromatic heterocyclic ring, or a bicyclic with only the first ring being aromatic or heteroaromatic;
Ro is selected from a group consisting of H and lower alkyl;
L is selected from a group consisting of a bond, and a substituted or unsubstituted alkyl, alkenyl, or alkynyl linking moiety;
R. is selected from a group consisting of C(R')3, OR', N(R')2, NR'C(O)R', NR'C(O)O(R*), NR*C(O)N(R')2, SR\ C(O)(O)R', C(O), C(O)N(R')2. SO3R', OSO2R*, SO2R", SOR', S(O)N(R')2, OS(O)(O)N(R')2, S(O)(O)N(R')2, S(O)N(R')2, PO-tR', OPO2R', PO3R', PO2R', and a 3-6 membered heterocycle with one or more heterocyclic atoms with each heteroatom independently being capable of carrying any R' group on it, wherein R' is selected from a group consisting of hydrogen, lower alkyl, alkyl-hydroxyl, thiol-alkyl, alkyl-thiol, aminoalkyl, alkylamino, branched alkyl, branched alkyl hydroxyl, branched thio-alkyl, branched alkyl-thiol, branched aminoalkyl, brnached alkylamino, and a closed 3-6 membered carbocycle or heterocycle, with each heteroatom in the 3-6 membered heterocycle being capable of carrying any R* group on it, and wherein each R' is independent in case there is more than one R';
R2 is a substitutent situated at position 5,6 or 8 of the ring, wherein R2 is selected from a group consisting of methyl, ethyl, 7?-propyl, iso-propyl, 7J-butyl, iso-butyl, te7-t-butyl, zso-pentyl, phenyl, substituted phenyl, halogen, branched or unbranched alkylamino, branched or unbranched aminoalkyl, branched or unbranched alkyloxo, branched or unbranched oxyalkyl, branched or unbranched thioalkyl, branched or unbranched alkylthiol, CF3, sulfonamido, substituted sulfonamido, sulfonate, sulfonate ester, phosphate, phosphate ester, phosphonate, phosphonate ester, carboxo, amido, ureido, substituted carboxo, substituted amido, substituted ureido, or a 3-6 membered carbocycle or heterocycle attached to positions 5, 6 or 8 directly or through group L, each heteroatom independently being capable of carrying any group R2, with the further proviso that either one, two or three substituents R2 are present in the ring, each of the substituents R2 being the same or different;
R3 selected from a group consisting of hydrogen, alkyl, alkoxy, halogen, CF3, cyano, substituted alkyl, or hydroxyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, heterocycle, C(R")3, OR", N(R")2, NR"C(O)R", NR"C(O)NR", R", C(O)(O)R", OC(O)R", C(O)N(R")2, C(O), OC(O)N(R")2, SO3R", OSO2R", SO2R", SOR", PO4R", OPO2R", PO3R", PO2R", wherein R" is hydrogen, aryl, substituted aryl, heteroaryl, substituted heteroaryl, lower alkyl, branched lower alkyl, alkyl-hydroxyl, branched alkyl-hydroxyl, amino-alkyl, branched amino-alkyl, alkyl-amino, branched alkyl-amino, thiol-alkyl, branched thiol-alkyl, alkyl-thiol, branched thiol-alkyl, or may form a closed 3-6 membered heterocycle with one or more heterocyclic atoms, branched alkyl, branched alkyl hydroxyl, where each R" is independent in case there is more than one R"; n is an integer having the value between 1 and 5, with the further proviso that if n > 2, then each group R3 is independent of the other groups R3,
with the further proviso that if each A is (CH)0, L is a bond,
with the further proviso that if each B is (CH)0, R3 is any substitutent described above other than hydrogen and is bonded directly to the position 7 of the adjacent ring; and pharmaceutically acceptable salts, hydrates, solvates, crystal forms, N- oxides, and individuals diastereoners thereof.
2. The compound of claim 1 selected from the group consisting of:
Figure imgf000329_0001
(IX)
Figure imgf000329_0002
(XI)
Figure imgf000330_0001
(XLNIII)
Figure imgf000330_0002
(XLIX)
Figure imgf000330_0003
(L)
Figure imgf000330_0004
(LII)
Figure imgf000330_0005
(LIII)
Figure imgf000331_0001
(LIV)
Figure imgf000331_0002
(CXXXII)
Figure imgf000331_0003
(CXXXIV)
Figure imgf000331_0004
(CXXXVIII)
Figure imgf000331_0005
(CCXLIII)
Figure imgf000332_0001
(CCLV)
Figure imgf000332_0002
(CCLVIII)
Figure imgf000332_0003
(CCLX)
Figure imgf000332_0004
(CCLXI)
Figure imgf000332_0005
(CCLXII)
Figure imgf000333_0001
(CCLXIII)
Figure imgf000333_0002
(CCLXV)
Figure imgf000333_0003
(CCLXVI)
Figure imgf000333_0004
(CCLXXII) and
Figure imgf000333_0005
(CCLXXVII)
3. The compound of claim 1 selected from the group consisting of:
Figure imgf000334_0001
(LI)
Figure imgf000334_0002
(LXX)
Figure imgf000334_0003
(LXXI)
Figure imgf000334_0004
(CXII)
Figure imgf000334_0005
(CXIV)
Figure imgf000335_0001
(CXV)
and
Figure imgf000335_0002
(CCLXXIX) 4. The compound of claim 1 selected from the group consisting of:
Figure imgf000335_0003
(LVIII)
Figure imgf000335_0004
(LX)
Figure imgf000336_0001
(LXII)
Figure imgf000336_0002
(LXVI)
Figure imgf000336_0003
(LXXIII)
Figure imgf000336_0004
(LXXVI)
Figure imgf000336_0005
(LXXVIII)
Figure imgf000337_0001
(XCII)
Figure imgf000337_0002
(XCVII)
Figure imgf000337_0003
(XCIX)
Figure imgf000337_0004
(CIII)
Figure imgf000337_0005
(CXVII)
Figure imgf000338_0001
(CXXI)
Figure imgf000338_0002
(CXXIV)
Figure imgf000338_0003
(CXXV)
Figure imgf000338_0004
(CXLVI)
Figure imgf000338_0005
(CL)
Figure imgf000339_0001
(CLI)
Figure imgf000339_0002
(CLVII)
Figure imgf000339_0003
(CLXIV)
Figure imgf000339_0004
(CLXVI)
Figure imgf000339_0005
(CLXXI)
Figure imgf000340_0001
(CLXXII)
Figure imgf000340_0002
(CLXXIII)
Figure imgf000340_0003
(CLXXIV)
Figure imgf000340_0004
(CLXXV)
Figure imgf000340_0005
(CLXXXIV)
Figure imgf000341_0001
(CLXXXVI)
Figure imgf000341_0002
(CXC)
Figure imgf000341_0003
(CCXII)
Figure imgf000341_0004
(CCXXVI)
Figure imgf000341_0005
(CCXXXI)
Figure imgf000342_0001
(CCXLVII)
Figure imgf000342_0002
(CCXLVIII)
Figure imgf000342_0003
(CCL)
Figure imgf000342_0004
(CCLII)
Figure imgf000342_0005
(CCLXVII)
Figure imgf000343_0001
(CCLXXIII)
(CCLXXV) and
Figure imgf000343_0003
(CCLXXX)
5. The compound of claim 1 selected from the group consisting of:
Figure imgf000343_0004
(X)
Figure imgf000343_0005
Figure imgf000344_0001
(XIII)
Figure imgf000344_0002
(XIV)
Figure imgf000344_0003
(XV)
Figure imgf000344_0004
(XVII)
Figure imgf000344_0005
(XVIII)
Figure imgf000345_0001
(XIX)
Figure imgf000345_0002
(XX)
Figure imgf000345_0003
(XXI)
Figure imgf000345_0004
(XXIV)
Figure imgf000345_0005
(XXV)
Figure imgf000346_0001
(XXVI)
Figure imgf000346_0002
(XXVIII)
Figure imgf000346_0003
(XXIX)
Figure imgf000346_0004
(XXX)
Figure imgf000346_0005
(XLII) and
Figure imgf000347_0001
(CXXX)
The compound of claim 1 selected from the group consisting of:
Figure imgf000347_0002
(XXII)
Figure imgf000347_0003
(XLIII)
Figure imgf000347_0004
(XLV)
Figure imgf000347_0005
(XLVI) and
Figure imgf000348_0001
(CXI)
The compound of claim 1 selected from the group consisting of:
Figure imgf000348_0002
(XCIV)
Figure imgf000348_0003
(CLIII)
Figure imgf000349_0001
(CLV) and
Figure imgf000349_0002
(CCLXXVIII)
8. The compound of claim 1 selected from the group consisting of:
Figure imgf000349_0003
(LXXXVII)
Figure imgf000349_0004
(LXXXVIII)
Figure imgf000350_0001
(LXXXIX)
Figure imgf000350_0002
(XC)
Figure imgf000350_0003
(CXL)
Figure imgf000350_0004
(CXLII)
Figure imgf000351_0001
(CXLIII)
Figure imgf000351_0002
(CXLIV)
Figure imgf000351_0003
(CXCVIII)
Figure imgf000351_0004
(CCV)
Figure imgf000351_0005
(CCVII)
Figure imgf000352_0001
(CCXVII)
Figure imgf000352_0002
(CCLXVIII) and
Figure imgf000352_0003
(CCLXX)
9. The compound of claim 1 having the formula:
Figure imgf000352_0004
(LXXXVI)
10. The compound of claim 1 selected from the group consisting of:
Figure imgf000353_0001
(CXXXVI)
Figure imgf000353_0002
(CLXXX) and
Figure imgf000353_0003
(CCXLIV)
11. The compound of claim 1 selected from the group consisting of:
Figure imgf000353_0004
(CLXIX) and
Figure imgf000354_0001
(CCXX)
12. The compound of claim 1 having the formula:
Figure imgf000354_0002
(CLIX)
13. The compound of claim 1 selected from the group consisting of:
Figure imgf000354_0003
(CI)
Figure imgf000354_0004
(CLXX)
Figure imgf000355_0001
(CLXXXII) and
Figure imgf000355_0002
(CCXXIII)
14. The compound of claim 1 selected from the group consisting of:
Figure imgf000355_0003
(LXVII)
Figure imgf000355_0004
(LXVIII)
Figure imgf000356_0001
(LXIX)
Figure imgf000356_0002
(CIV)
Figure imgf000356_0003
(CV)
Figure imgf000356_0004
(CVI)
Figure imgf000356_0005
(CVII)
Figure imgf000357_0001
(CVIII) and
Figure imgf000357_0002
(CIX)
15. The compound of claim 1 selected from the group consisting of:
Figure imgf000357_0003
(CXXVII)
Figure imgf000357_0004
(CXXVIII)
Figure imgf000358_0001
(CLXIII)
Figure imgf000358_0002
(CLXXVIII)
Figure imgf000358_0003
(CLXXXVIII)
Figure imgf000358_0004
(CXCV)
Figure imgf000358_0005
(CCXL)
Figure imgf000359_0001
(CCXLI)
Figure imgf000359_0002
(CCXLII)
Figure imgf000359_0003
(CCLIII)
Figure imgf000359_0004
(CCLVI)
Figure imgf000359_0005
(CLXI) and
Figure imgf000360_0001
(CCLXXI)
16. The compound of claim 1 selected from the group consisting of:
Figure imgf000360_0002
(XXXI)
Figure imgf000360_0003
(XXXII)
Figure imgf000360_0004
(XXXIII)
Figure imgf000361_0001
(XXXIV)
Figure imgf000361_0002
(XXXV)
Figure imgf000361_0003
(XXXVI)
Figure imgf000361_0004
(XXXVII)
Figure imgf000362_0001
(XXXVIII)
Figure imgf000362_0002
(XXXIX)
Figure imgf000362_0003
(XL)
Figure imgf000362_0004
(XLI)
Figure imgf000363_0001
(LV)
Figure imgf000363_0002
(LXIII)
Figure imgf000363_0003
(CXX)
Figure imgf000363_0004
(CCXLV) and
Figure imgf000363_0005
(CCLI)
7. The compound of claim 1 selected from the group consisting of:
Figure imgf000364_0001
(LVII)
Figure imgf000364_0002
(LIX)
Figure imgf000364_0003
(LXI)
Figure imgf000364_0004
(LXV)
Figure imgf000364_0005
(LXXII)
Figure imgf000365_0001
(LXXV)
Figure imgf000365_0002
(LXXVII)
Figure imgf000365_0003
(XCI)
Figure imgf000365_0004
(XCVI)
Figure imgf000365_0005
(XCVIII)
Figure imgf000366_0001
(C)
Figure imgf000366_0002
(CH)
Figure imgf000366_0003
(CXVI)
Figure imgf000366_0004
(CXXIII)
Figure imgf000366_0005
(CXLVIII)
Figure imgf000367_0001
(CCXLVI)
Figure imgf000367_0002
(CCXLIX)
Figure imgf000367_0003
(CCLIV)
Figure imgf000367_0004
(CCLIX) and
Figure imgf000367_0005
(CCLXIV)
18. The compound of claim 1 selected from the group consisting of:
Figure imgf000368_0001
(CLXVIII) and
Figure imgf000368_0002
(CXCII)
19. The compound of claim 1 selected from the group consisting of:
Figure imgf000368_0003
(CCXXXVII)
Figure imgf000368_0004
(CCXXXVIII)
Figure imgf000369_0001
(CCXXXIX) and
Figure imgf000369_0002
(CCLXXIV)
20. The compound of claim 1 selected from the group consisting of:
Figure imgf000369_0003
(LXXIX)
Figure imgf000369_0004
(LXXX)
Figure imgf000370_0001
(LXXXI)
Figure imgf000370_0002
(LXXXII)
Figure imgf000370_0003
(LXXXIII)
and
Figure imgf000370_0004
(CLXXVI)
21. A method for treating a disorder associated with compromised vasculostasis, comprising administering to a subject in need thereof a therapeutically effective amount of at least one compound of claim 1.
22. The method of claim 21, wherein the disorder is myocardial infarction, stroke, congestive heart failure, an ischemia or reperfusion injury, cancer, arthritis or other arthropathy, retinopathy or vitreoretinal disease, macular degeneration, autoimmune disease, vascular leakage syndrome, inflammatory disease, edema, transplant rejection, burn, or acute or adult respiratory distress syndrome (ARDS).
23. The method of claim 21, wherein the disorder is vascular leakage syndrome (VLS).
24. The method of claim 21 , wherein the disorder is cancer.
25. The method of claim 2 wherein the disorder is a vitreoretinal disease.
26. The method of claim 2 , wherein the disorder is ARDS.
27. The method of claim 2 wherein the disorder is autoimmune disease.
28. The method of claim 2 , wherein the disorder is burn.
29. The method of claim 2 wherein the disorder is stroke.
30. The method of claim 2 wherein the disorder is myocardial infarction.
31. The method of claim 2 , wherein the disorder is ischemia or reperfusion injury.
32. The method of claim 2 , wherein the disorder is arthritis.
33. The method of claim 2 , wherein the disorder is edema.
34. The method of claim 2 , wherein the disorder is transplant rejection.
35. The method of cl aim 2 , wherein the disorder is inflammatory disease.
36. The method of claim 2 , wherein the disorder is congestive heart failure.
37. The method of claim 2 wherein the disorder is associated with a kinase.
38. The method of claim 37, wherein the kinase is a tyrosine kinase.
39. The method of claim 37, wherein the kinase is a serine kinase or a threonine kinase.
40. The method of claim 37, wherein the kinase is a Src family kinase.
41. A pharmaceutical composition comprising at least one compound of claim 1 and a pharmaceutically acceptable carrier therefore.
42. An article of manufacture comprising packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material comprises a label which indicates that the pharmaceutical composition can be used for treatment of disorders associated with compromised vasculostasis, and wherein the pharmaceutical composition comprises at least one compound of claim 1.
43. An article of manufacture comprising packaging material and a pharmaceutical composition contained within the packaging material, wherein the packaging material comprises a label which indicates that the pharmaceutical composition can be used for treatment of disorders associated with vascular permeability leakage or compromised vasculostasis selected from myocardial infarction, stroke, congestive heart failure, an ischemia or reperfusion injury, cancer, arthritis or other arthropathy, retinopathy or vitreoretinal disease, macular degeneration, autoimmune disease, vascular leakage syndrome, inflammatory disease, edema, transplant rejection, burn, or acute or adult respiratory distress syndrome (ARDS), and wherein the pharmaceutical composition comprises at least one compound of claim 1.
44. The article of manufacture of claim 43, wherein the disorder is cancer.
45. A method of treating a disorder associated with compromised vasculostasis, comprising the administration of a therapeutically effective amount of at least one compound of claim 1 or pharmaceutically acceptable salts, hydrates, solvates, crystal forms and individual diastereomers thereof, to a subject in need of such treatment.
46. The method of claim 45, wherein the disorder is vascular leakage syndrome (VLS).
47. The method of claim 45, wherein the disorder is cancer.
48. The method of claim 45, wherein the disorder is a vitreoretinal disease.
49. The method of claim 45, wherein the disorder is ARDS.
50. The method of claim 45, wherein the disorder is an autoimmune disease.
51. The method of claim 45, wherein the disorder is burn.
52. The method of claim 45, wherein the disorder is stroke.
53. The method of claim 45, wherein the disorder is myocardial infarction.
54. The method of claim 45, wherein the disorder is ischemia or reperfusion injury.
55. The method of claim 45, wherein the disorder is arthritis.
56. The method of claim 45, wherein the disorder is edema.
57. The method of claim 45, wherein the disorder is transplant rejection.
58. The method of claim 45, wherein the disorder is inflammatory disease.
59. A method of treating a disorder associated with compromised vasculostasis comprising the administration of a therapeutically effective amount of at least one compound of claim 1, or pharmaceutically acceptable salts, hydrates, solvates, crystal forms and individual diastereomers thereof, in combination with an anti-inflammatory agent, chemotherapeutic agent, immunomodulatory agent, therapeutic antibody, or a protein kinase inhibitor, to a subject in need of such treatment.
60. A method of treating a subject having or at risk of having myocardial infarction comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
61. A method of treating a subject having or at risk of having vascular leakage syndrome (VLS) comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
62. A method of treating a subject having or at risk of having cancer comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
63. A method of treating a subject having or at risk of having stroke comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
64. A method of treating a subject having or at risk of having ARDS comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
65. A method of treating a subject having or at risk of having burns comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
66. A method of treating a subject having or at risk of having arthritis comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1 , thereby treating the subject.
67. A method of treating a subject having or at risk of having edema comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
68. A method of treating a subject having or at risk of having vascular leakage syndrome (VLS) comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
69. A method of treating a subject having or at risk of having retinopathy or vitreoretinal disease comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
70. A method of treating a subject having or at risk of having ischemic or reperfusion related tissue injury or damage, comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
71. A method of treating a subject having or at risk of having an autoimmune disease, comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
72. A method of treating a subject having or at risk of having transplant rejection, comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
73. A method of treating a subject having or at risk of having inflammatory disease, comprising administering to the subject a therapeutically effective amount of at least one compound of claim 1, thereby treating the subject.
74. A process for making a pharmaceutical composition comprising combining a combination of at least one compound of claim 1 or its pharmaceutically acceptable salts, hydrates, solvates, crystal forms salts and individual diastereomers thereof, and a pharmaceutically acceptable carrier.
PCT/US2005/012057 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases WO2005096784A2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
AU2005231507A AU2005231507B2 (en) 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases
NZ551027A NZ551027A (en) 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases
CN200580018660.1A CN101426772B (en) 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases
CA2567574A CA2567574C (en) 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases
EP05762774.7A EP1809614B1 (en) 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases
IL178908A IL178908A (en) 2004-04-08 2006-10-26 Benzotriazine inhibitors of kinases
HK08100766.4A HK1110578A1 (en) 2004-04-08 2008-01-21 Benzotriazine inhibitors of kinases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US56123704P 2004-04-08 2004-04-08
US60/561,237 2004-04-08
US64343905P 2005-01-12 2005-01-12
US60/643,439 2005-01-12

Publications (2)

Publication Number Publication Date
WO2005096784A2 true WO2005096784A2 (en) 2005-10-20
WO2005096784A3 WO2005096784A3 (en) 2009-02-19

Family

ID=35125563

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/012057 WO2005096784A2 (en) 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases

Country Status (10)

Country Link
US (3) US7456176B2 (en)
EP (2) EP2543376A1 (en)
KR (1) KR20070011458A (en)
AU (1) AU2005231507B2 (en)
CA (1) CA2567574C (en)
HK (1) HK1110578A1 (en)
IL (1) IL178908A (en)
NZ (2) NZ588139A (en)
RU (1) RU2006139258A (en)
WO (1) WO2005096784A2 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006131835A2 (en) * 2005-02-01 2006-12-14 Sentinel Oncology Limited Heterocyclic triazines as hypoxic selective protein kinase inhibitors
EP1799656A1 (en) * 2004-08-25 2007-06-27 Targegen, Inc. Heterocyclic compounds and methods of use
EP1893216A1 (en) * 2005-06-08 2008-03-05 Targegen, Inc. Methods and compositions for the treatment of ocular disorders
WO2010076238A1 (en) 2008-12-29 2010-07-08 Fovea Pharmaceuticals Sa Substituted quinazoline compounds
WO2010092041A1 (en) 2009-02-13 2010-08-19 Fovea Pharmaceuticals Sa [1, 2, 4] triazolo [1, 5 -a] pyridines as kinase inhibitors
US7858782B2 (en) 2006-12-15 2010-12-28 Abraxis Bioscience, Llc Triazine derivatives and their therapeutical applications
WO2011161159A1 (en) 2010-06-22 2011-12-29 Fovea Pharmaceuticals Heterocyclic compounds, their preparation and their therapeutic application
JP2012528136A (en) * 2009-05-26 2012-11-12 センティネル・オンコロジー・リミテッド Substituted benzotriazines and quinoxalines as inhibitors of p70S6 kinase
EP2543376A1 (en) * 2004-04-08 2013-01-09 Targegen, Inc. Benzotriazine inhibitors of kinases
US8877924B2 (en) 2009-06-09 2014-11-04 NantBio Inc. Benzyl substituted triazine derivatives and their therapeutical applications
US8969347B2 (en) 2008-06-03 2015-03-03 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
AU2009322346B2 (en) * 2008-12-03 2015-07-02 The Scripps Research Institute Stem cell cultures
US9078902B2 (en) 2009-06-09 2015-07-14 Nantbioscience, Inc. Triazine derivatives and their therapeutical applications
US9359379B2 (en) 2012-10-02 2016-06-07 Intermune, Inc. Anti-fibrotic pyridinones
US9527816B2 (en) 2005-05-10 2016-12-27 Intermune, Inc. Method of modulating stress-activated protein kinase system
US10233195B2 (en) 2014-04-02 2019-03-19 Intermune, Inc. Anti-fibrotic pyridinones
US10246393B2 (en) 2016-10-31 2019-04-02 Tosoh Corporation Method for producing aromatic compound

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050282814A1 (en) * 2002-10-03 2005-12-22 Targegen, Inc. Vasculostatic agents and methods of use thereof
KR20110050745A (en) * 2002-10-03 2011-05-16 탈자진 인코포레이티드 Vasculostatic agents and methods of use thereof
US8133900B2 (en) 2005-11-01 2012-03-13 Targegen, Inc. Use of bi-aryl meta-pyrimidine inhibitors of kinases
US8604042B2 (en) * 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
KR101467723B1 (en) 2005-11-01 2014-12-03 탈자진 인코포레이티드 Bi-aryl meta-pyrimidine inhibitors of kinases
ZA200804679B (en) * 2005-12-08 2010-02-24 Millenium Pharmaceuticals Inc Bicyclic compounds with kinase inhibitory activity
US7691858B2 (en) * 2006-04-25 2010-04-06 Targegen, Inc. Kinase inhibitors and methods of use thereof
WO2009062112A2 (en) * 2007-11-09 2009-05-14 The Salk Institute For Biological Studies Use of tam receptor inhibitors as antimicrobials
CA2723358A1 (en) * 2008-05-05 2009-11-12 Allison B. Reiss Method for improving cardiovascular risk profile of cox inhibitors
EP2447256A1 (en) * 2010-10-21 2012-05-02 Laboratorios Lesvi, S.L. Process for obtaining dronedarone
AU2010363329A1 (en) 2010-11-07 2013-05-09 Targegen, Inc. Compositions and methods for treating myelofibrosis
WO2016089208A2 (en) 2014-12-04 2016-06-09 Stichting Maastricht Radiation Oncology "Maastro-Clinic" Sulfonamide, sulfamate and sulfamide derivatives of anti-cancer agents
CN116410159B (en) * 2023-06-09 2023-08-22 济南国鼎医药科技有限公司 Preparation method and application of En Qu Ti-Ni intermediate

Family Cites Families (161)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2003199A (en) * 1930-05-31 1935-05-28 Johnson Frank James Automatic coal stoker
US2003149A (en) * 1931-05-22 1935-05-28 Autographic Register Co Manifolding
US2003065A (en) 1931-06-20 1935-05-28 John R Ditmars Composition for coating sheets, fibrous stocks, and the like
US2004102A (en) * 1932-02-24 1935-06-11 Daniel A Dickey Hollow steel propeller construction
US2004138A (en) * 1932-11-30 1935-06-11 Byers A M Co Method of making wrought iron pipe
US2002165A (en) * 1933-07-08 1935-05-21 Charles A Winslow Air cleaner
US2003187A (en) * 1933-10-02 1935-05-28 Frederick H Shaw Automobile radio device
US2003166A (en) * 1933-10-26 1935-05-28 Zancan Ottavio Front drive for motor cars
US2001051A (en) * 1933-12-01 1935-05-14 Angelina Mariani Tamperproof meter box with cutout control and fuse drawer for electric meters
US2004092A (en) * 1933-12-15 1935-06-11 John L Chaney Device for indicating the temperature of liquids
US2003060A (en) * 1934-04-02 1935-05-28 Ernest L Heckert Thermostatic controlling device
US2667486A (en) * 1951-05-24 1954-01-26 Research Corp 2,4-diamino pteridine and derivatives
IL26578A (en) 1965-10-04 1970-11-30 Merck & Co Inc Pteridine compounds and their preparation
DE2255947A1 (en) 1972-11-15 1974-05-22 Bayer Ag SUBSTITUTED 3-AMINO-BENZO-1,2,4TRIAZINE-DI-N-OXIDES (1,4), METHOD FOR MANUFACTURING AND USING THEM AS ANTIMICROBIAL AGENTS
IE38778B1 (en) * 1973-02-02 1978-05-24 Ciba Geigy New 1,2,4-benzotriazine derivatives
FR2275461A1 (en) 1974-06-18 1976-01-16 Labaz NEW STABILIZERS FOR POLYMERS AND COPOLYMERS OF VINYL CHLORIDE
AU535258B2 (en) * 1979-08-31 1984-03-08 Ici Australia Limited Benzotriazines
EP0059524A1 (en) 1981-02-09 1982-09-08 Smith and Nephew Associated Companies p.l.c. Pharmaceutical composition containing aminopteridines or aminopyrimido(4,5-d)pyrimidines
DE3205638A1 (en) 1982-02-17 1983-08-25 Hoechst Ag, 6230 Frankfurt Trisubstituted pyrimidine-5-carboxylic acids and their derivatives, processes for their preparation, and their use as pesticides
US4490289A (en) * 1982-09-16 1984-12-25 Hoffmann-La Roche Inc. Homogeneous human interleukin 2
US5616584A (en) * 1986-09-25 1997-04-01 Sri International 1,2,4-benzotriazine oxides as radiosensitizers and selective cytotoxic agents
JP2598100B2 (en) 1988-08-31 1997-04-09 キヤノン株式会社 Electrophotographic photoreceptor
US5214059A (en) * 1989-07-03 1993-05-25 Hoechst-Roussel Pharmaceuticals Incorporated 2-(aminoaryl) indoles and indolines as topical antiinflammatory agents for the treatment of skin disorders
US5665543A (en) * 1989-07-18 1997-09-09 Oncogene Science, Inc. Method of discovering chemicals capable of functioning as gene expression modulators
US5776502A (en) * 1989-07-18 1998-07-07 Oncogene Science, Inc. Methods of transcriptionally modulating gene expression
US5062685A (en) 1989-10-11 1991-11-05 Corning Incorporated Coated optical fibers and cables and method
JPH03127790A (en) 1989-10-11 1991-05-30 Morishita Pharmaceut Co Ltd N-(1h-tetrazol-5-yl)-2-anilino-5-pyrimidinecarboxamides and synthetic intermediate therefor
GB9003553D0 (en) 1990-02-16 1990-04-11 Ici Plc Herbicidal compositions
JP2839106B2 (en) 1990-02-19 1998-12-16 キヤノン株式会社 Electrophotographic photoreceptor
MY107955A (en) * 1990-07-27 1996-07-15 Ici Plc Fungicides.
GB9016800D0 (en) * 1990-07-31 1990-09-12 Shell Int Research Tetrahydropyrimidine derivatives
DE4025891A1 (en) * 1990-08-16 1992-02-20 Bayer Ag PYRIMIDYL-SUBSTITUTED ACRYLIC ACID ESTERS
JPH05345780A (en) * 1991-12-24 1993-12-27 Kumiai Chem Ind Co Ltd Pyrimidine or triazine derivative and herbicide
HUT63941A (en) 1992-05-15 1993-11-29 Hoechst Ag Process for producing 4-alkyl-substituted pyrimidine-5-carboxanilide derivatives, and fungicidal compositions comprising same
US5482951A (en) 1992-05-29 1996-01-09 Kumiai Chemical Industry Co., Ltd. Triazole derivatives as well as insecticide and acaricide
US5763441A (en) 1992-11-13 1998-06-09 Sugen, Inc. Compounds for the treatment of disorders related to vasculogenesis and/or angiogenesis
JPH0741461A (en) 1993-05-27 1995-02-10 Eisai Co Ltd Sulfonic acid ester derivative
JPH0782183A (en) 1993-09-09 1995-03-28 Canon Inc Liquid crystal instermediate compound, liquid crystal compound, high molecular liquid crystal compound, liquid crystal copolymer, composition thereof, liquid crystal element and recorder
DE4338704A1 (en) * 1993-11-12 1995-05-18 Hoechst Ag Stabilized oligonucleotides and their use
US5530000A (en) * 1993-12-22 1996-06-25 Ortho Pharmaceutical Corporation Substituted pyrimidinylaminothiazole derivatives useful as platelet aggreggation inhibitors
GB9506466D0 (en) * 1994-08-26 1995-05-17 Prolifix Ltd Cell cycle regulated repressor and dna element
US5597826A (en) * 1994-09-14 1997-01-28 Pfizer Inc. Compositions containing sertraline and a 5-HT1D receptor agonist or antagonist
DE19502912A1 (en) * 1995-01-31 1996-08-01 Hoechst Ag G-Cap Stabilized Oligonucleotides
US6326487B1 (en) * 1995-06-05 2001-12-04 Aventis Pharma Deutschland Gmbh 3 modified oligonucleotide derivatives
US5827850A (en) 1995-09-25 1998-10-27 Sanofi Pharmaceuticals, Inc. 1,2,4-benzotriazine oxides formulations
CA2232989C (en) 1995-09-25 2008-01-29 Sanofi Pharmaceuticals, Inc. 1,2,4-benzotriazine oxides formulations
JPH09274290A (en) 1996-02-07 1997-10-21 Fuji Photo Film Co Ltd Developing solution and processing method for silver halide photographic material
DE69734321T2 (en) * 1996-02-23 2006-08-10 Eli Lilly And Co., Indianapolis NON-PEPTIDIC VASOPRESSIN VIA ANTAGONISTS
DE59707681D1 (en) * 1996-10-28 2002-08-14 Rolic Ag Zug Crosslinkable, photoactive silane derivatives
ATE348155T1 (en) * 1996-11-20 2007-01-15 Introgen Therapeutics Inc AN IMPROVED METHOD FOR PRODUCTION AND PURIFICATION OF ADENOVIRAL VECTORS
JP3734903B2 (en) * 1996-11-21 2006-01-11 富士写真フイルム株式会社 Development processing method
JPH10153838A (en) 1996-11-22 1998-06-09 Fuji Photo Film Co Ltd Method for processing silver halide photographic sensitive material
JP3720931B2 (en) * 1996-11-26 2005-11-30 富士写真フイルム株式会社 Processing method of silver halide photographic light-sensitive material
US5935383A (en) * 1996-12-04 1999-08-10 Kimberly-Clark Worldwide, Inc. Method for improved wet strength paper
JPH10207019A (en) 1997-01-22 1998-08-07 Fuji Photo Film Co Ltd Method for processing silver halide photographic sensitive material
JPH10213820A (en) 1997-01-31 1998-08-11 Canon Inc Liquid crystal element and liquid crystal device
DE59807348D1 (en) * 1997-02-05 2003-04-10 Rolic Ag Zug Photocrosslinkable silane derivatives
JPH10260512A (en) 1997-03-19 1998-09-29 Fuji Photo Film Co Ltd Method for processing silver halide photosensitive material
US6070126A (en) * 1997-06-13 2000-05-30 William J. Kokolus Immunobiologically-active linear peptides and method of identification
EP0994100B1 (en) 1997-06-24 2006-08-30 Nikken Chemicals Company, Limited 3-anilino-2-cycloalkenone derivates
US6635626B1 (en) * 1997-08-25 2003-10-21 Bristol-Myers Squibb Co. Imidazoquinoxaline protein tyrosine kinase inhibitors
US6685938B1 (en) * 1998-05-29 2004-02-03 The Scripps Research Institute Methods and compositions useful for modulation of angiogenesis and vascular permeability using SRC or Yes tyrosine kinases
US6136971A (en) * 1998-07-17 2000-10-24 Roche Colorado Corporation Preparation of sulfonamides
US6378526B1 (en) * 1998-08-03 2002-04-30 Insite Vision, Incorporated Methods of ophthalmic administration
US6288082B1 (en) * 1998-09-29 2001-09-11 American Cyanamid Company Substituted 3-cyanoquinolines
US6297258B1 (en) * 1998-09-29 2001-10-02 American Cyanamid Company Substituted 3-cyanoquinolines
WO2000025780A1 (en) 1998-10-29 2000-05-11 Bristol-Myers Squibb Company Compounds derived from an amine nucleus that are inhibitors of impdh enzyme
FR2792314B1 (en) 1999-04-15 2001-06-01 Adir NOVEL AMINOTRIAZOLE COMPOUNDS, PROCESS FOR THEIR PREPARATION AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THE SAME
AU5636900A (en) * 1999-06-30 2001-01-31 Merck & Co., Inc. Src kinase inhibitor compounds
AU5570600A (en) * 1999-07-01 2001-01-22 Ajinomoto Co., Inc. Heterocyclic compounds and medicinal use thereof
TWI262914B (en) 1999-07-02 2006-10-01 Agouron Pharma Compounds and pharmaceutical compositions for inhibiting protein kinases
AU763976B2 (en) 1999-07-23 2003-08-07 Shionogi & Co., Ltd. Th2 differentiation inhibitors
US6093838A (en) * 1999-08-16 2000-07-25 Allergan Sales, Inc. Amines substituted with a dihydro-benzofuranyl or with a dihydro-isobenzofuranyl group, an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
US6127382A (en) * 1999-08-16 2000-10-03 Allergan Sales, Inc. Amines substituted with a tetrahydroquinolinyl group an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
JP2001089412A (en) 1999-09-22 2001-04-03 Otsuka Pharmaceut Co Ltd Benzene derivative or its pharmaceutically acceptable salt
US6506769B2 (en) * 1999-10-06 2003-01-14 Boehringer Ingelheim Pharmaceuticals, Inc. Heterocyclic compounds useful as inhibitors of tyrosine kinases
DE60025243T2 (en) 1999-10-12 2006-06-22 Takeda Pharmaceutical Co. Ltd. PYRIMIDIN-5-CARBOXIMIDE COMPOUNDS, METHOD FOR THE PRODUCTION THEREOF AND THEIR USE
US6638929B2 (en) * 1999-12-29 2003-10-28 Wyeth Tricyclic protein kinase inhibitors
US6153752A (en) * 2000-01-28 2000-11-28 Creanova, Inc. Process for preparing heterocycles
US20020165244A1 (en) * 2000-01-31 2002-11-07 Yuhong Zhou Mucin synthesis inhibitors
GB0004887D0 (en) * 2000-03-01 2000-04-19 Astrazeneca Uk Ltd Chemical compounds
US6605615B2 (en) * 2000-03-01 2003-08-12 Tularik Inc. Hydrazones and analogs as cholesterol lowering agents
JP2001247411A (en) 2000-03-09 2001-09-11 Tomono Agrica Co Ltd Pest-controlling agent
US6608048B2 (en) * 2000-03-28 2003-08-19 Wyeth Holdings Tricyclic protein kinase inhibitors
MXPA02009763A (en) * 2000-04-04 2003-03-27 Shionogi & Co Oily compositions containing highly fat soluble drugs.
US6471968B1 (en) * 2000-05-12 2002-10-29 Regents Of The University Of Michigan Multifunctional nanodevice platform
DE10024622A1 (en) 2000-05-18 2001-11-22 Piesteritz Stickstoff New N-(2-pyrimidinyl)-(thio)phosphoric acid triamide compounds are urease inhibitors useful for preventing hydrolysis of urea fertilizers or for reducing release of ammonia in animal stalls
EP1170353B1 (en) 2000-07-06 2005-11-02 Fuji Photo Film Co., Ltd. Liquid crystal composition comprising liquid crystal molecules and aligment promoter
JP5000068B2 (en) * 2000-08-11 2012-08-15 ベーリンガー インゲルハイム ファーマシューティカルズ インコーポレイテッド Heterocyclic compounds useful as inhibitors of tyrosine kinases
US20020137755A1 (en) 2000-12-04 2002-09-26 Bilodeau Mark T. Tyrosine kinase inhibitors
JP2002221770A (en) 2001-01-24 2002-08-09 Fuji Photo Film Co Ltd Silver halide photographic sensitive material and method of processing the same
WO2002096903A2 (en) * 2001-05-28 2002-12-05 Aventis Pharma S.A. Chemical derivatives and the use thereof as an anti-telomerase agent
ES2320204T3 (en) * 2001-05-29 2009-05-20 Bayer Schering Pharma Aktiengesellschaft CDK INHIBITING PYRIMIDINES, ITS PREPARATION AND ITS USE AS MEDICATIONS.
US6689778B2 (en) * 2001-07-03 2004-02-10 Vertex Pharmaceuticals Incorporated Inhibitors of Src and Lck protein kinases
UY27487A1 (en) * 2001-10-17 2003-05-30 Boehringer Ingelheim Pharma PIRIMIDINE DERIVATIVES, MEDICATIONS CONTAINING THESE COMPOUNDS, THEIR EMPLOYMENT AND PROCEDURE FOR PREPARATION
EP1453516A2 (en) * 2001-10-17 2004-09-08 Boehringer Ingelheim Pharma GmbH & Co.KG Novel tri-substituted pyrimidines, method for production and use thereof as medicament
US20060292206A1 (en) * 2001-11-26 2006-12-28 Kim Steven W Devices and methods for treatment of vascular aneurysms
US20030187026A1 (en) * 2001-12-13 2003-10-02 Qun Li Kinase inhibitors
US20030166932A1 (en) * 2002-01-04 2003-09-04 Beard Richard L. Amines substituted with a dihydronaphthalenyl, chromenyl, or thiochromenyl group, an aryl or heteroaryl group and an alkyl group, having retinoid-like biological activity
WO2003074515A1 (en) * 2002-03-01 2003-09-12 Smithkline Beecham Corporation Diamino-pyrimidines and their use as angiogenesis inhibitors
MY141867A (en) 2002-06-20 2010-07-16 Vertex Pharma Substituted pyrimidines useful as protein kinase inhibitors
WO2004005283A1 (en) 2002-07-09 2004-01-15 Vertex Pharmaceuticals Incorporated Imidazoles, oxazoles and thiazoles with protein kinase inhibiting activities
ES2337782T3 (en) 2002-07-29 2010-04-29 Rigel Pharmaceuticals, Inc. METHODS TO TREAT OR PREVENT AUTOIMMUNITY DISEASES WITH 2,4-PYRIMIDINDIAMINE COMPOUNDS.
DE60316810T2 (en) * 2002-08-02 2008-07-17 Ab Science 2- (3-AMINOARYL) AMINO-4-ARYL-THIAZOLE AND THEIR USE AS C-KIT INHIBITORS
JP4741948B2 (en) * 2002-08-14 2011-08-10 バーテックス ファーマシューティカルズ インコーポレイテッド Protein kinase inhibitors and their use
UY27939A1 (en) 2002-08-21 2004-03-31 Glaxo Group Ltd COMPOUNDS
US7230101B1 (en) 2002-08-28 2007-06-12 Gpc Biotech, Inc. Synthesis of methotrexate-containing heterodimeric molecules
DE10240261A1 (en) 2002-08-31 2004-03-11 Clariant Gmbh Production of aryllithium-electrophile reaction products of interest for the pharmaceutical and agrochemical industries comprises using an organolithium compound prepared by reacting an aryl halide with lithium
DE10240262A1 (en) * 2002-08-31 2004-03-11 Clariant Gmbh Production of aryllithium-electrophile reaction products of interest for the pharmaceutical and agrochemical industries comprises using an organolithium compound prepared by reacting an aryl halide with lithium
KR20110050745A (en) * 2002-10-03 2011-05-16 탈자진 인코포레이티드 Vasculostatic agents and methods of use thereof
US20050282814A1 (en) * 2002-10-03 2005-12-22 Targegen, Inc. Vasculostatic agents and methods of use thereof
WO2004032709A2 (en) 2002-10-04 2004-04-22 Caritas St.Elisabeth's Medical Center Of Boston, Inc. Inhibition of src for treatment of reperfusion injury related to revascularization
US7189712B2 (en) 2002-10-10 2007-03-13 Smithkline Beecham Corporation 1,3-Oxazole compounds for the treatment of cancer
AU2003301662A1 (en) 2002-10-21 2004-05-13 Bristol-Myers Squibb Company Quinazolinones and derivatives thereof as factor xa inhibitors
AU2003286711A1 (en) 2002-10-25 2004-05-13 Vertex Pharmaceuticals Incorporated Indazolinone compositions useful as kinase inhibitors
DE10250708A1 (en) 2002-10-31 2004-05-19 Boehringer Ingelheim Pharma Gmbh & Co. Kg New alkyne compounds having MCH antagonist activity and medicaments containing these compounds
CN100513397C (en) 2002-11-19 2009-07-15 记忆药物公司 Pyridine n-oxide compounds as phosphodiesterase 4 inhibitors
MXPA05005585A (en) 2002-12-06 2005-07-27 Warner Lambert Co Benzoxazin-3-ones and derivatives thereof as inhibitors of pi3k.
JP3837670B2 (en) 2002-12-12 2006-10-25 富士通株式会社 Data relay apparatus, associative memory device, and associative memory device utilization information retrieval method
AU2003299807A1 (en) 2002-12-20 2004-07-22 Pharmacia Corporation Heteroarylalkanoic acids as integrin receptor antagonists
UA80767C2 (en) 2002-12-20 2007-10-25 Pfizer Prod Inc Pyrimidine derivatives for the treatment of abnormal cell growth
EP1572210A1 (en) 2002-12-20 2005-09-14 Pharmacia Corporation The r-isomer of beta amino acid compounds as integrin receptor antagonists derivatives
JP2006512387A (en) 2002-12-24 2006-04-13 アストラゼネカ アクチボラグ Compound
US7144911B2 (en) 2002-12-31 2006-12-05 Deciphera Pharmaceuticals Llc Anti-inflammatory medicaments
PL232814B1 (en) 2003-02-07 2019-07-31 Janssen Pharmaceutica Nv Hiv inhibiting 1,2,4-triazines
EP1597237B1 (en) 2003-02-07 2016-07-27 Janssen Pharmaceutica NV Pyrimidine derivatives for the prevention of HIV infection
EP1594512A4 (en) 2003-02-11 2007-07-11 Kemia Inc Compounds for the treatment of viral infection
CL2004000303A1 (en) 2003-02-20 2005-04-08 Tibotec Pharm Ltd COMPOUNDS DERIVED FROM PYRIMIDINS AND TRIAZINES; PREPARATION PROCESS; PHARMACEUTICAL COMPOSITION; AND ITS USE TO INHIBIT HIV REPLICATION.
ES2634840T5 (en) 2003-04-24 2022-12-22 Coopervision Int Ltd Hydrogel contact lenses and packaging systems and production methods thereof
DK1660458T3 (en) 2003-08-15 2012-05-07 Irm Llc 2,4-Pyrimidine diamines useful in the treatment of neoplastic diseases, inflammatory disorders and disorders of the immune system.
JP2007505858A (en) 2003-09-18 2007-03-15 ノバルティス アクチエンゲゼルシャフト 2,4-Di (phenylamino) pyrimidine useful for the treatment of proliferative disorders
EP2409704B1 (en) 2003-09-26 2017-09-20 Exelixis, Inc. c-Met modulators and methods of use
WO2005035541A1 (en) 2003-10-07 2005-04-21 Amedis Pharmaceuticals Ltd. Silicon compounds and their use
DE10356579A1 (en) 2003-12-04 2005-07-07 Merck Patent Gmbh amine derivatives
KR20070026390A (en) 2004-01-23 2007-03-08 암젠 인코포레이션 Compounds and methods of use
CA2567574C (en) * 2004-04-08 2013-01-08 Targegen, Inc. Benzotriazine inhibitors of kinases
WO2005108370A1 (en) 2004-04-16 2005-11-17 Ajinomoto Co., Inc. Benzene compounds
EP1799656A4 (en) 2004-08-25 2009-09-02 Targegen Inc Heterocyclic compounds and methods of use
US7210697B2 (en) * 2004-12-16 2007-05-01 Tricam International, Inc. Convertible handle
CA2593803A1 (en) 2005-01-26 2006-08-03 Irm Llc Thiazole-amide compounds and compsitions as protein kinase inhibitors
GB0501999D0 (en) 2005-02-01 2005-03-09 Sentinel Oncology Ltd Pharmaceutical compounds
JP2008533166A (en) 2005-03-16 2008-08-21 ターゲジェン インコーポレーティッド Pyrimidine compounds and methods of use
US20070032493A1 (en) * 2005-05-26 2007-02-08 Synta Pharmaceuticals Corp. Method for treating B cell regulated autoimmune disorders
WO2006128129A2 (en) 2005-05-26 2006-11-30 Synta Pharmaceuticals Corp. Method for treating cancer
NZ563984A (en) 2005-06-08 2011-11-25 Targegen Inc Methods and compositions for the treatment of ocular disorders
WO2007008541A2 (en) 2005-07-08 2007-01-18 Kalypsys, Inc. Cellular cholesterol absorption modifiers
EP1940843A4 (en) 2005-08-11 2010-09-15 Ariad Pharma Inc Unsaturated heterocyclic derivatives
TW200745066A (en) 2005-09-16 2007-12-16 Torrent Pharmaceuticals Ltd Novel PTP1B inhibitors
US20070072682A1 (en) * 2005-09-29 2007-03-29 Crawford James T Iii Head to head electronic poker game assembly and method of operation
KR101467723B1 (en) 2005-11-01 2014-12-03 탈자진 인코포레이티드 Bi-aryl meta-pyrimidine inhibitors of kinases
US8133900B2 (en) 2005-11-01 2012-03-13 Targegen, Inc. Use of bi-aryl meta-pyrimidine inhibitors of kinases
US8604042B2 (en) 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
US20070161645A1 (en) * 2005-11-02 2007-07-12 Targegen, Inc. Thiazole inhibitors targeting resistant kinase mutations
US7691858B2 (en) * 2006-04-25 2010-04-06 Targegen, Inc. Kinase inhibitors and methods of use thereof
US8030487B2 (en) 2006-07-07 2011-10-04 Targegen, Inc. 2-amino—5-substituted pyrimidine inhibitors
WO2009026346A1 (en) 2007-08-20 2009-02-26 Targegen Inc. Thiazolidine compounds, and methods of making and using same
WO2009046416A1 (en) 2007-10-05 2009-04-09 Targegen Inc. Anilinopyrimidines as jak kinase inhibitors
WO2009049028A1 (en) 2007-10-09 2009-04-16 Targegen Inc. Pyrrolopyrimidine compounds and their use as janus kinase modulators
WO2009055674A1 (en) 2007-10-26 2009-04-30 Targegen Inc. Pyrrolopyrimidine alkynyl compounds and methods of making and using same
MX2010008744A (en) 2008-02-08 2012-09-28 Targegen Inc Pteridine derivatives for treating respiratory disease.
WO2010017122A2 (en) 2008-08-05 2010-02-11 Targegen Inc. Methods of treating thalassemia

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
None
See also references of EP1809614A4

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2543376A1 (en) * 2004-04-08 2013-01-09 Targegen, Inc. Benzotriazine inhibitors of kinases
US8481536B2 (en) 2004-04-08 2013-07-09 Targegen, Inc. Benzotriazine inhibitors of kinases
US8372971B2 (en) 2004-08-25 2013-02-12 Targegen, Inc. Heterocyclic compounds and methods of use
EP1799656A1 (en) * 2004-08-25 2007-06-27 Targegen, Inc. Heterocyclic compounds and methods of use
EP1799656A4 (en) * 2004-08-25 2009-09-02 Targegen Inc Heterocyclic compounds and methods of use
AU2005276974B2 (en) * 2004-08-25 2012-08-02 Targegen, Inc. Heterocyclic compounds and methods of use
EP2532653A1 (en) * 2004-08-25 2012-12-12 Targegen, Inc. Benzo[1,2,4]triazines as protein kinase modulators
US8084618B2 (en) 2004-08-25 2011-12-27 Targegen, Inc. Heterocyclic compounds and methods of use
WO2006131835A3 (en) * 2005-02-01 2007-05-10 Sentinel Oncology Ltd Heterocyclic triazines as hypoxic selective protein kinase inhibitors
WO2006131835A2 (en) * 2005-02-01 2006-12-14 Sentinel Oncology Limited Heterocyclic triazines as hypoxic selective protein kinase inhibitors
US10010536B2 (en) 2005-05-10 2018-07-03 Intermune, Inc. Method of modulating stress-activated protein kinase system
US9527816B2 (en) 2005-05-10 2016-12-27 Intermune, Inc. Method of modulating stress-activated protein kinase system
EP1893216A4 (en) * 2005-06-08 2012-08-08 Targegen Inc Methods and compositions for the treatment of ocular disorders
EP1893216A1 (en) * 2005-06-08 2008-03-05 Targegen, Inc. Methods and compositions for the treatment of ocular disorders
US7858782B2 (en) 2006-12-15 2010-12-28 Abraxis Bioscience, Llc Triazine derivatives and their therapeutical applications
EP2923703A1 (en) 2006-12-15 2015-09-30 Abraxis BioScience, Inc. Triazine derivatives and their therapeutical applications
EP2425840A1 (en) 2006-12-15 2012-03-07 Abraxis BioScience, Inc. Triazine derivatives and their therapeutical applications
US8580786B2 (en) 2006-12-15 2013-11-12 Nant Holdings Ip, Llc Triazine derivatives and their therapeutical applications
US8969347B2 (en) 2008-06-03 2015-03-03 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
US9290450B2 (en) 2008-06-03 2016-03-22 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
USRE47142E1 (en) 2008-06-03 2018-11-27 Intermune, Inc. Compounds and methods for treating inflammatory and fibrotic disorders
US10975352B2 (en) 2008-12-03 2021-04-13 The Scripps Research Institute Methods of enhancing cell survival of stem cells
US9896655B2 (en) 2008-12-03 2018-02-20 The Scripps Research Institute Methods of enhancing cell survival of stem cells
AU2009322346B2 (en) * 2008-12-03 2015-07-02 The Scripps Research Institute Stem cell cultures
US10351822B2 (en) 2008-12-03 2019-07-16 The Scripps Research Institute Methods of enhancing cell survival of stem cells
US9340525B2 (en) 2008-12-03 2016-05-17 The Scripps Research Institute Stem cell cultures
WO2010076238A1 (en) 2008-12-29 2010-07-08 Fovea Pharmaceuticals Sa Substituted quinazoline compounds
WO2010092041A1 (en) 2009-02-13 2010-08-19 Fovea Pharmaceuticals Sa [1, 2, 4] triazolo [1, 5 -a] pyridines as kinase inhibitors
US8716473B2 (en) 2009-05-26 2014-05-06 Sentinel Oncology Limited Substituted benzotriazines and quinoxalines as inhibitors of P7OS6 kinase
JP2012528136A (en) * 2009-05-26 2012-11-12 センティネル・オンコロジー・リミテッド Substituted benzotriazines and quinoxalines as inhibitors of p70S6 kinase
US8877924B2 (en) 2009-06-09 2014-11-04 NantBio Inc. Benzyl substituted triazine derivatives and their therapeutical applications
US9409903B2 (en) 2009-06-09 2016-08-09 Nantbioscience, Inc. Benzyl substituted triazine derivatives and their therapeutical applications
US9078902B2 (en) 2009-06-09 2015-07-14 Nantbioscience, Inc. Triazine derivatives and their therapeutical applications
WO2011161159A1 (en) 2010-06-22 2011-12-29 Fovea Pharmaceuticals Heterocyclic compounds, their preparation and their therapeutic application
US9675593B2 (en) 2012-10-02 2017-06-13 Intermune, Inc. Anti-fibrotic pyridinones
US9359379B2 (en) 2012-10-02 2016-06-07 Intermune, Inc. Anti-fibrotic pyridinones
US10376497B2 (en) 2012-10-02 2019-08-13 Intermune, Inc. Anti-fibrotic pyridinones
US10898474B2 (en) 2012-10-02 2021-01-26 Intermune, Inc. Anti-fibrotic pyridinones
US10233195B2 (en) 2014-04-02 2019-03-19 Intermune, Inc. Anti-fibrotic pyridinones
US10544161B2 (en) 2014-04-02 2020-01-28 Intermune, Inc. Anti-fibrotic pyridinones
US10246393B2 (en) 2016-10-31 2019-04-02 Tosoh Corporation Method for producing aromatic compound

Also Published As

Publication number Publication date
IL178908A0 (en) 2011-08-01
WO2005096784A3 (en) 2009-02-19
US8481536B2 (en) 2013-07-09
EP2543376A1 (en) 2013-01-09
US7456176B2 (en) 2008-11-25
HK1110578A1 (en) 2008-07-18
AU2005231507B2 (en) 2012-03-01
EP1809614A2 (en) 2007-07-25
AU2005231507A1 (en) 2005-10-20
CA2567574A1 (en) 2005-10-20
EP1809614B1 (en) 2014-05-07
RU2006139258A (en) 2008-05-20
US20110294796A1 (en) 2011-12-01
KR20070011458A (en) 2007-01-24
NZ551027A (en) 2011-01-28
NZ588139A (en) 2012-02-24
CA2567574C (en) 2013-01-08
US20090275569A1 (en) 2009-11-05
US20050245524A1 (en) 2005-11-03
EP1809614A4 (en) 2010-02-17
IL178908A (en) 2015-09-24

Similar Documents

Publication Publication Date Title
AU2005231507B2 (en) Benzotriazine inhibitors of kinases
DK1951684T3 (en) BIARYLMETAPYRIMIDIN kinase inhibitors
AU2003282726B2 (en) Vasculostatic agents and methods of use thereof
AU2006227628A1 (en) Pyrimidine compounds and methods of use
WO2007056075A2 (en) Six membered heteroaromatic inhibitors targeting resistant kinase mutations
WO2008008234A1 (en) 2-amino-5-substituted pyrimidine inhibitors
JP6175139B2 (en) New triazine compounds
CN111566100B (en) Pyrimidine compound, preparation method and medical application thereof
EP1877398B1 (en) Quinazoline derivatives as egf and/or erbb2 tyrosine kinase inhibitors
CN101426772B (en) Benzotriazine inhibitors of kinases
AU2012202928A1 (en) Benzotriazine inhibitors of kinases

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KM KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 178908

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2005231507

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 551027

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 12006502199

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 200609267

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2006139258

Country of ref document: RU

Ref document number: 1020067023392

Country of ref document: KR

Ref document number: 6617/DELNP/2006

Country of ref document: IN

Ref document number: 2005762774

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2567574

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2005231507

Country of ref document: AU

Date of ref document: 20050407

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005231507

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 200580018660.1

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 1020067023392

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2005762774

Country of ref document: EP