CA2567574C - Benzotriazine inhibitors of kinases - Google Patents

Benzotriazine inhibitors of kinases Download PDF

Info

Publication number
CA2567574C
CA2567574C CA2567574A CA2567574A CA2567574C CA 2567574 C CA2567574 C CA 2567574C CA 2567574 A CA2567574 A CA 2567574A CA 2567574 A CA2567574 A CA 2567574A CA 2567574 C CA2567574 C CA 2567574C
Authority
CA
Canada
Prior art keywords
compound
mmol
disorder
methyl
treat
Prior art date
Legal status (The legal status 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 status listed.)
Expired - Fee Related
Application number
CA2567574A
Other languages
French (fr)
Other versions
CA2567574A1 (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
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
TargeGen Inc
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
Publication of CA2567574A1 publication Critical patent/CA2567574A1/en
Application granted granted Critical
Publication of CA2567574C publication Critical patent/CA2567574C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

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

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

DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PLUS D'UN TOME.
CECI EST LE TOME DE _2 NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des Brevets.

JUMBO APPLICATIONS / PATENTS

THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE: For additional volumes please contact the Canadian Patent Office.

BENZOTRIAZINE INHIBITORS OF KINASES
CROSS REFERENCE TO RELATED APPLICATIONS
[0001]

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 (HER1 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 VEGFR. 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.
In colon tumors, increased c-Src TK activity has been shown to correlate to tumor progression, with the highest activity found in metastatic 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.

[00091 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 VEGF 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 VEGF is administered in in vivo studies.
Examples where excessive vascular permeability leads to particularly deleterious effects include pulmonary edema, cerebral edema, and cardiac edema.

[00101 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, VEGF-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 TNF-induced leakage. Kinases are believed to mediate the phosphorylation of junctional proteins such as beta-catenin and vascular endothelial VE-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.
[00111 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 5"
6"
B`B,B
(R3) n 11 B;B,B1 7 NN2 A~I A`A 4' 2" / ,A~. ~A L
N \
1,A R1 6 4 N' L Ro (I) [0017] wherein each of A can be (CH)0_1, N, NH, 0, 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)0_1, N, NH, 0, 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] Ro can be H or lower alkyl;

[0020] L can be a bond, or a substituted or unsubstituted alkyl, alkenyl, or alkynyl linking moiety;

[0021] R1 can be C(R')3, OR', N(R')2a 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')2a PO4R', OPO2R', PO3R', PO2R', or a 3-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!;

[0022] R2 is a substitutent situated at position 5,6 or 8 of the ring, wherein R2 can be methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tent-butyl, iso-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", POOR", 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)o, 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, bum, 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.

[00291 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 (VLS), 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] In 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.
[00351 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.

[00381 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, iso-propyl, n-butyl, iso-butyl, tent-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, ainido, aldehyde, acyl, oxyacyl, carboxyl, sulfonyl, sulfonamide, sulfuryl, and the like.

[00401 The term "lower alkyl" refers to alkyl groups having from 1 to about 6 carbon atoms.

10041] 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 -0-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 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):

5"

B B,B
(R3)n BB,B, 7 N N
2 A:' A"A 41 211 ,A ,A L\ :~~3 6 I N N 11 '4:, RI
L Ro (I) [0061] wherein each of A can be (CH)0_1, N, NH, 0, 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)o_t, N, NH, 0, 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;

[0063] Ro can be H or lower alkyl;

[0064] L can be a bond, or a substituted or unsubstituted alkyl, alkenyl, or alkynyl linking moiety;

[0065] Rl 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-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 amino alkyl, 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, iso-propyl, n-butyl, iso-butyl, tent-butyl, iso-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;

[0067] 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)NK", 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 R3 is independent of the other groups R3, [0069] with the further proviso that if each A is (CH)o, L is a bond.

[0070] Some exemplary compounds described by formula (I) that can be used include the following compounds:

N: / I O
CH3 \
N N
H
(IX) N'N
I / O\~ N~
CH3 \
N N "a (XI) \ I \ N; IN /~I O~/~ IN

N N
H
(XLVIII) \ I \ NN
N N
H
(XLIX) \ I \ N. N / Obi N
N N
H

(L) \ I \ N; N O
N N
H

(LII) N,N

N N
H
(LIII) N,N

H

(LIV) Me N,N
H
Me N~N ,S~NNC) H p' O
Me (CXXXII) N, N
N
N H O~0 (CXXXIV) N, / ~S`.O
~I /N~-N
N H

(CXXXVIII) \ I \ NN N
N~N~ NH
H

(CCXLIII) \ I \ N;N OA. N N

H
(CCLV) O
N; N N
N N eH
H

(CCLVIII) O
\ I \ N: / I H
NN \
H
(CCLX) N N

NN \ I N'-~ N
j:y H V
(CCLXI) N 'N H
NN \ I N,.
H
O ONH
(CCLXII) \ I \ N,N

N H oSO No (CCLXIII) O
CI \ I \ N'N N-^') NN \IN1-1 H
(CCLXV) F \ I \ N / I ON
V
N N
H
(CCLXVI) NC alo~ NOSN'~NH
N N
H
(CCLXXII) 0. ,0 NC \ I \ N N / I S,N'- ~N
N N
H
(CCLXXVII) CI

\ I \ N N N
/ I
CI N N
\/
H

(LI) __CI
O
/ \ N\N / N

I \ I NH
CI N N
H
(LXX) CI

N, N NN
\ I 0 CI
N Na H

(LXXI) \ CI

N
CI I / \ I NH
N N
H
(CXII) CI

N
/

CI N N
H
(CXIV) CI

N. N / OS N ~\i N
CI \ I H
N N
H
(CXV) CI

N ~S0 N
"N / I ~
CI N~N \
H
(CCLXXIX) CI

HO I / \ N,N
/ J. \ .
N H N
OS N
O

(LVIII) CI

HO I / \ N, N SN
N N
H
(LX) CI

HO I / \ N'N
~N
\I
N Na H

(LXII) CI

HO I / \ N; N rNi IJ. \I NJ
N H OS.\
O
(LXVI) CI

HO I\ / N' N N

N N
H
(LXXIII) CI

HO \ N'N az~-~ -~ N
N N S
H
(LXXVI) \ CI

HON ; N / O N ~i N
N N
H
(LXXVIII) CI

HO \ N'N / OS N~~N/
I H
N NH
(XCII) CI

HO N'N
a~~, ,,~ N
N N O
H
(XCVII) CI O

HO I ~ N' N N~
N
NN~
H
(XCIX) CI
N 0S0 ~
HO 'N / I N
N~N \
H
(Cm) CI
ON ,O
HO N'N

N N
H
(CXVII) CI

H O N N;N SO

I OH
N N
H
(CXXI) CI

HO N NI S~\N
N N
H
(CXXIV) HO

N, N / O-~ N
N N a (CXXV) HO I / OW, NN I H

(CXLVI) CI

HO N' N / N
N N
H
(CL) CI
HO I / \ N, N / N
NON \ I N,_) --r H
H O
(CLI) CI
HO \ N'N
N~N -\ ND
H

(CLVII) Br o\ .~O
HO I / \ N / S
\ ~ HN~
N H

(CLXIV) CI

N sN
OH I NN NCI
H

(CLXVI) II
HO \ I / N; N / S.NiN

N N \~
H
(CLXXI) Cl 0 rO
J
HO \ N' N 1 NN

\
N N
H
(CLXXII) ~ CI
HO \ I N'N N
N N
H
(CLXXIII) OH

F O
n N,. SN
N N
~O H
NN I
H
(CLXXIV) \ I \ N N / S,N' N
OH I / NN \ I H
H
(CLXXV) F

HO I N'N - 0 N NH
(CLXXXIV) F

HO N' IN HN--~
N N
H
(CLXXXVI) CI
HO I / N, N OS.O
/ HN
NN
H rN
(CXC) 'N' OS
HO N
HN_ O N N ~N J
H

(CCXII) CI

HO \ I ~ N' N OO
F I / HN_~
N H NJ
(CCXXVI) HO \ I \ N; N 0S"O
HN
N H

(CCXXXI) CI

HO \ I / N; N OS~O
HN
CI NN \ I ~N~
H
(CCXLVII) CI

HO \ I / N; N N~\iOH
N N
H
(CCXLVIII) CI

HO N N / O~\NV
CI \
N N
H
(CCL) CI
O\
\ I / / ,O
HO N'N S
CI I \ I ~NH
N N
H
(CCLII) HO NN

N N
H
(CCLXVII) Cl HO \ \ N,N
NN
H

(CCLXXIII) CI

o HO \ N, NI ja N
CI N N
H
(CCLXXV) Cl 0 HO N' N / "'NN
F \ O
N N a (CCLXXX) CH3 N~N \ N
H
(X) \ I \ N NN

H N\
(XII) \ IN N N
H
(XIII) ~N
Iq N, N~N
H
(XIV) N.N
/
N Na H

(XV) I/ \I
N N
H
(XVI) N'N
N NJ?"
H
(XVII) N;N
N \
H
(XVIII) cN

NN
H
(XIX) N, N

N N
H
I

(XX) \ I \ N F
`N

N N CI
H

(XXI) N `N

NN
H
(XXIV) ;N O /

N N :o (XXV) N; IN 9~4 N N
H
(XXVI) N;N
N N
H

(XXVIII) N'N acl N N H

(XXIX) N / CI
N N CI
H

(XXX) N'N N

N N
H
(XLII) N, N

N H O~So (CXXX) CI

N'N
N N
H
(XXII) CI

N;N
N N ao H

(XLIII) CI

N. N / N
CI Ili" N N
H
(XLV) CI

N` N /
CI N
N
H

(XLVI) CI
O,S ~
N
N I I N~ 4-CI ON O
N H O
(CXI) Cl HO I / N,N

(XCIII) CI
H
HO N,N / I N
N N O
H
(_XCIV) CI
HO N. N /

N N N
H

(CLIII) CI

HO N,N N O1 N N
H
(CLV) CI N ' N p HO / N~
O
N~N 0 H
(CCLXXVIII) I Oo O0 N
( ~GN,N / S.H~' N N

(LXXXVII) ON ,O
N, IN S,NN
N N

(LXXXVIII) o, ,o N N / SN
H
N N
HN O H
(LXXXIX) 0~ sO
N, N s' N N
N N
HN O H

(XC) I \ N, N / ~ O\/\ N
N N

(CXL) N, N N
N

(CXLII) NN / I O~~No N N
HN ( H

(CXLIII) /
N, N O
N N
HN, - H
ps 0 (CXLIV) NN HN
O N H N

(CXCVIII) N, N S`~O
HN
HO NN NC]
H
(CCV) N,N S\

_0 NN NC]
H

(CCVII) N, N / OS.=O
HN
HO~\O I / N~N \ I -N~]
H
(CCXVII) Oõ
\ I \ N, N / NN
/ \ I H
HN N N
H
(CCLXVIII) N 'N / S N- N
aN \ H
HN N
H
O NH

(CCLXX) N 'N

(LXXXVI) N; N S HN-/-NC) NN~
H O
(CXXXVI) \ I / N,N O

N,,,~ N
N N
H
(CLXXX) N' N N
N N ND
(CCXLIV) cl N.N N N
cI NN N
H
N ~~OH
(CLXIX) cl \ ~ \ N Jam. N N N~
cl N N \ O-~
H
(CCXX) cl HO I / "N N N

H
CN \

(CLIX) cl O
N; / N~~NC
HO
H
NN & N
H
(CI) CI

HO N'N N N
NHN

N'-'OH
(CLXX) CI O
NH
HO N N N \-\N
NNS
H
(CLXXXII) CI

HO N'N NJ N
N
NN"~O~
H
(CCXXIII) CI

N, (LXVII) CI
0, 0 02N N' N / , N N
H
N N
H
(LXVIII) CI
O O
H2N I / \ N; N N-~~N
N N
H
(LXIX) ~S

N~NH2 (CIV) O O O
jS. I / \ N; N SNNo N N J
H
(CV) 02N N" N
N-5~ NH2 (CVI) O. 0 O2N I / \ N' N / N
N~N \ I H
H
(CVII) O~ O
N N: N / SNN
H I
N N H
J
H

(CVIII) O\ O
H2N 'N / N
N~N I H
H

(CIX) O
N N N OMe ~
N~NH /
Me (CXXVII) N

O N, N / O~~N
N N a (CXXVIII) OõO
N " N SNi--'-'N D
HN_ H
N

(CLXIII) N N' N / N

aN N
H
(CLXXVIII) HN
N \\ 0 ,N HN
N:,~ N
H ~N
p /\d~
(CLXXXVIII) CI
S 0~
i I N:N / I
~HN~

(cxcv) g N; N
/~
\/
N Na H

(CCXL) O
N~ N N N
NN N
H
(CCXLI) CI O

N~ I ~ N;N / N
NN ~ I N
H

(CCXLII) O p N\ I \ N,N \ N") / N~N / ~N
H

(CCLIII) O
NX N" N N
N N
H
(CCLVI) / NN
HN
L / N~NH2 (CLXI) HO
S N' N ON N "~'~ N
H
N N
H
(CCLXXI) O

O \ I N'N
N N
H

(XXXI) O 'N
N
N N
H
(XXXII) O
O \ I ~ N /
NN ~
H
(XXXIII) O~
O \ I N'N
NN
H
(XXXIV) O~

O N 'N /
N N
H
(XXXV) O~
OI N,N /
NN \
H
(XXXVI) O~
O I N;N / I O~
N N
H
(XXXVII) O~

O N"N / N
N N
H
(XXXVIII) O

N
O ,N /
N N
H
(SIX) O
O \ I \ NIN
/ NN
H O
(XL) O
O N;N /
N NH

(XLI) CI
MeO N

(LV) CI
MeO I / \ N; N

N~NH2 (LXIII) CI
~ \ I 0S0 O N I / \ N
N H y O
(CXX) N. ~ N

(CCXLV) CI
OO
O \ I / N; N / S
CI \ u O I /
N H II
O
(CCLI) CI
MeO I / \ N' N

(LVII) CI

MeO N, N S.NN -15 N

H
(LIX) CI

MeO N' N O N
/
N N
H
(LXI) CI

MeO N' N N
/ ~ ~ I NJ
N H N /S.-\
O
(LXV) CI

O N;N / N
~~ ONE
N N
H
(LXXII) CI

O N;N
N N S ,~ N
H

(LXXV) CI

O I/ N. N O S N
N N
H
(LXXVII) / CI
O I N. N / ON /0 S N
H
I
N N~
H
(XCI) CI

N; N

N N O
H

(XCVI) CI O
e I

NN ~
H
(XCVIII) CI O
No N
N; N X~N
NN H
H

(C) 0 ,N N
N N
H
(CII) CI

0 \ I \ N N / O S N NO
N N
H
(CXVI) / CI

O \ I \ N'N / SN
.5~ -1-, 0 N N
H
(CXXIII) CI

~0 I / \ N,,N 0N
/
N N
H
(CXLVIII) CI
0 \ I / N N OS "0 HN
CI N~N \ I --\'-N3 H
(CCXLVI) CI

O \ I / N: N / ON
CI \ I v N N
H
(CCXLIX) OMe CI \ \ N' N / O~\N~
N NH

(CCLIV) CI

MeO \ I \ N'N / O~\N
/ \ I v N N
H
(CCLIX) MeO N / I N
NN \
415:::~ O
H

(CCLXIV) O
HEN \ I \ N; N / S ' N N
~O H
0 NN \
H
(CLXVIII) HO N, N / OS., N H

(CXCII) Y
O H
` , N
HO.,~~ N N' N S N

N N
H
(CCXXXVII) Y H
O N N. N / OS N ~\ N
~I
H NH
OH

(CCXXXVIII) H
Br N. N \S, N N

:)::::~N N
H
(CCXXXIX) OõO
HO N I 'N I 'N
H
N N
H
(CCLXXIV) p cI

AO / I \ N, N p'/--'N
N N
H
(LXXIX) O CI

A0 ( / \ N"N

N N O
H

(LXXX) O CI

\ O I / \ N; N O~~ N
N N /
H
(LXXXI) O CI

N N
H
(LXXXII) O CI

O I / \ N' N N
N N
H
(LXXXIII) and O~~ N
N N/ I
H
(CLXXVI) [00711 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 (ARKS), lupus, vascular leakage, transplant (such as organ transplant, acute transplant or heterograft or homograft (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 Fe 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 basophil 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 IL-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.

[00761 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).
In 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, p-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 T
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.
[00871 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 m -the art. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form.
Additionally, suspensions of the active compounds may be 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 cisplatinlcarboplatin; alkylating agents, such as canbusil; topoisomerase I
inhibitors such ,M
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 -deoxydaunorubicin, 13-deoxydaunorubicin, adriamycin-14-benzoate, adriamycin-14-octanoate, adriamycin-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 HER-2 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 Vitaxin (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., Vincristine, 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 DNA-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 DNA-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-chlorodeoxyadeno sine [CdA], and 2'-Deoxycoformycin [Pentostatin], etc.), pyrimidine antagonists (e.g., fluoropyriinidines [e.g., 5-fluorouracil (Adrucil), 5-fluorodeoxyuridine (FdUrd) (Floxiuridine)] 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., IFN-.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 gp,39 (i.e., CD 154), fusion proteins constructed from CD40 and gp39 (CD401g 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 antiinflannnatory drugs (NSAIDs) such as ibuprofen and cyclooxygenase inhibitors such as rofecoxib, steroids such as prednisone or dexamethasone, gold compounds, antiproliferative agents such as methotrexate, FK506 (tacrolimus, Prograf), mycophenolate mofetil, cytotoxic drugs such as azathioprine and cyclophosphainide, 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 proteins, 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 maybe 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 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 100.0, 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 maybe 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 SymmetryShieldTM RP 18 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.
[0101] NMR spectra: 1H 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, in =
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:

o- o-Br I \ NOZ N 1) Cone HCI Br N'.,N G3-B(OH)2 G3 I \ N
t N
NHZ NHZ 2) NaOH NNH Pd(dba)3 " NJINH2 2 P(Ph)3, K2CO3 G
10% PdIC
G3 N,'N Gj-Br G3 N N

N),N'G1 Pd(dba)3 N NH2 GZ H BINAP, KOt-Bu G2 4 (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 HCl 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 CH2C12 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 T.
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:

B r NO2 I N"N
+ NI 1) Conc HCI Br N N G3-B(OH)2 G3 +
NH2 NH2 2) NaOH Nlk Pd(dba)3 N NH2 G2 G2 2 P(Ph)3, K2CO3 G2 10% Pd/C

1:: N: N H2NSO3H G3 N;
N)N'G1 E G1-NH2 (III) [01101 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 equivalents of sulfamic acid. The mixture was heated at about 200 C overnight.
Compound 5 was isolated by HPLC.
Method C:

o_ 0.
Br N02 N 1) Conc HCI Br N N R2-B(OH)2 03 N N
NH2 NH2 2) NaOH N~NH Pd(dba)3 NNH2 z P(Ph)3, K2CO3 G2 10% PdIC
OMe G3 N N H2N G3 N'N
OMe BBr3 G3 Nz N G{-CI G3 N`NI / GI, ~ OH - I / \ Ad N H
N N
H 0z 7 (IV) [0111] The synthetic method C is shown by the reaction scheme (IV). 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 phenylamine 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 CH2C12. The mixture was cooled to about -78 C using a dry ice-acetone bath. About 2 equivalents of BBr3 (1M
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 NaHCO3 at about 0 C and by separating the mixture in a separator funnel. The water layer was extracted twice with CH2C12, 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 K2C03 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-benzof1,2,41triazin-3-ylamine-l-oxide O' Br N

N
aN.5~ Nhl2 (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 HC1 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 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 (V) was obtained. Yield: about 27%. ESI-MS: [M+H]+, 241, 243; 1H NMR
(DMSO-d6): 8 7.48 (d, J= 9.0 Hz, 1 H), 7.89 (dd, J1= 9.0 Hz, J2 = 2.1 Hz, 1 H), 8.26 (d, J= 2.1 Hz, 1 H).

Example 2 Synthesis of 7-bromo-5-methyl-benzo[1,2,41triazin-3-ylamine-l-oxide O' Br N N

(VI) [01131 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% NaOH 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 (VI) was obtained. Yield: about 36%. ESI-MS: [M+H]+, 255, 257; 'H NMR (DMSO-d6): 8 2.45 (s, 3 H), 7.81 (s, 1 H), 8.26 (s, 1 H).

Example 3 Synthesis of 7-bromo-5-methyl-benzo[1,2,4]triazin-3-ylamine Br N,N

(VII) [0114] About 4.26g of 7-bromo-5-methyl-benzo[1,2,4]triazin-3-ylamine-1-oxide (VI) 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 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 (VII) was obtained.

Example 4 Synthesis of 1-(3-chloropropanyl)pyrrolidine (VIII) [0115] To the solution of about 186 g (1.18 mol) of 1-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% NaOH, extracted with ether and dried over Na2SO4.
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-benzofl,2,4]triazin-3-yll-[4-(2-pyrrolidin-l-yl-ethoxy)-phenyll-amine N; N N

N N
H
(IX) [0116] About 1 g (3.78 mmol) of 7-bromo-5-methyl-benzo[1,2,4]triazin-3-ylamine and about 2.04 g (7.56 mmol) of 1-[2-(4-bromo-phenoxy)-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) E'-Ot-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-methyl-benzo[1,2,4]triazin-3-yl]-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyl]-amine (product having formula (IX)) was isolated. Yield: about 5.2%. ESI-MS: [M+H]+, 454.6;

NMR (DMSO-d6): 6 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 1-{4-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylaminol-phenyl}-cyclopropanecarbonitrile / CHs H
(X) [0117] About 50 mg (0.2 mmol) of 7-bromo-5-methyl-benzo[1,2,4]triazin-3-ylamine and about 84 mg (0.38 mmol) of 1-(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 1 - {4-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamino]-phenyl}-cyclopropanecarbonitrile (product having formula (X)) was isolated. ESI-MS: [M+H]+, 406.5; 1H NMR (DMSO-d6): 8 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 [7-(2,6-dimethyl-phenyl)-benzo[1,2,4]triazin-3-yll-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyll-amine I \ N; N / ( O~\ NV

N N
H
(XI) [0118] To a solution of about 100 mg (0.42 mmol) of 7-bromo-benzo[1,2,4]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 mrol) 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 CH2CI2 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 1-[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[1,2,4]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 NMR (DMSO-d6): 8 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 [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yll-guinolin-8-yl-amine by Method A

\ I \ N
NN
H N

(XII) [0119] About 100 mg (0.4 mmol) of 7-bromo-5-methyl-benzo[1,2,4]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 nunol) 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[1,2,4]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): 8 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-phenyl)-5-methyl-benzof1,2,4]triazin-3-yll-pyridin-2-yl-amine by Method A
N
N N N
H
(XIII) [0120] Title product having formula (XIII) was synthesized using Method A
described above. ESI-MS: [M+H]+, 342;1H NMR (500 MHz, DMSO-d6): S 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, 1H, NH).

Example 10 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-Quinolin-3-yl-amine by Method A

N,N
N~N N
H

(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): 5 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, 1H), 7.94 (dd, J1=
8.14 Hz, J2 =1.48 Hz, 1H), 8.01 (m, 2H), 9.16 (s, 1H), 9.30 (s, 1H), 11.44 (s, 1H, NH).

Example 11 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-m-tolyl-amine by Method B
Iq N/
NN \
H
(XV) [0122] Title product having formula (XV) was synthesized using Method B
described above. ESI-MS: [M+H]+, 355; 1H NMR (CDC13): 6 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= 7.6 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[1,2,4]triazin-yll-p-tolyl-amine by Method B

\ I \ N,N
N N
H

(XVI) [0123] Title product having formula (XVI) was synthesized using Method B
described above. ESI-MS: [M+H]+, 355; 1H NMR (CDC13): 8 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-phenyl)-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4ltriazin-3-yll-amine by Method B
N;N
NN
H\
(XVII) [0124] Title product having formula (XVII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 369; 1H NMR (CDC13): 8 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-methyl-benzo[1,2,4]triazin-3-yll-amine by Method B

N;N
N~N
H\
(XVIII) [0125] Title product having formula (XVIII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 369; 1H NMR (CDC13): 8 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, 1 H).

Example 15 Synthesis of (2,5-dimethyl-phenyl)-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,41triazin-3-yll-amine by Method B
N; N

N15:~ N
H
(XIX) [0126] Title product having formula (XIX) was synthesized using Method B
described above. ESI-MS: [M+H]+, 369; 1H NMR (CDC13): 8 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-phenyl)-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,41triazin-3-yll-amine by Method B

N`N
N N
H
(XX) [0127] Title product having formula (XX) was synthesized using Method B
described above. ESI-MS: [M+H]+, 369; 1H NMR (CDC13): 8 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-phenyl)-f7-(2,6-dimethyl-phenyl)-5-methyl-benzo11,2,41triazin-3-yll-amine by Method B

N,N / F

N N CI
H

(XXI) [0128] Title product having formula (XXI) was synthesized using Method B
described above. ESI-MS: [M+H]+, 393; 1H NMR (CDC13): 8 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, J1= 6.8 Hz, J2 = 3.2 Hz, 1 H).

Example 18 Synthesis of f7-(2-chloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yll-phenyl-amine by Method B

CI

N'N /
N N 'O
(XXII) [0129] Title product having formula (XXII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 347; 'H NMR (CDC13): 8 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-methoxv-phenyl)-5-methyl-benzo[1,2,41triazin-3-yll-amine by Method B
U
N; N O
N N
H
(XXIII) [0130] Title product having formula (XXIII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 373; 1H NMR (CDC13): S 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. O5, (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-butyl-phenyl)-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,41triazin-3-yll-amine by Method B

N
NN
H

(XXIV) [0131] Title product having formula (XXIV) was synthesized using Method B
described above. ESI-MS: [M+H]+, 397.3; 1H NMR (CDC13): 6 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= 7.6 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-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-(2-methoxy-phenyl)-amine by Method B
N ')0 N N
H
(XXV) [0132] Title product having formula (XXV) was synthesized using Method B
described above. ESI-MS: [M+H]+, 371.2;1H NMR (CDC13): 6 2.10 (s, 6 H), 2.75 (s, 3 H), 4.01 (s, 3 H), 6.98 (dd, J1= 7.6 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, J1= 7.6 Hz, J2 = 2.4 Hz, 1 H).

Example 22 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-(3-methoxy-phenyl)-amine by Method B

N,N 9~4 N N O
H
(XXVI) [0133] Title product having formula (XXVI) was synthesized using Method B
described above. ESI-MS: [M+H]+, 371.2; 1H NMR (CDC13): 6 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 17-(2,6-dimethyl-phenyl)-5-methyl-b enzof1,2,41triazin-yll-(4-methoxy-phenyl)-amine by Method B (product TG100-412-1) N; N , O

N N
H
(XXVII) [0134] Title product having formula (XXVII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 371.2; 1H NMR (CDCl3): 6 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 17-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,41triazin-yll-(4-ethoxy-phenyl)-amine by Method B

i N;N 0 N N
H
(XXVIII) [0135] Title product having formula (XXVIII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 385.3; 1H NMR (CDC13): S 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-chloro-phenyl)-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yll-amine by Method B
N acl 9~4 N N (XXIX) [01361 Title product having formula (XXIX) was synthesized using Method B
described above. ESI-MS: [M+H]+, 375.3;1H NMR (CDC13): 8 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[1,2,4ltriazin-3-yll-amine by Method B

N,N CI
N N CI
H

(XXX) [01371 Title product having formula (XXX) was synthesized using Method B
described above. ESI-MS: [M+H]+, 409.3; 'H NMR (CDC13): 8 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[1,2,4]triazin-yll-m-tolyl-amine by Method B
O

N
N N
H
(XXXI) [0138] Title product having formula (XXXI) was synthesized using Method B
described above. ESI-MS: [M+H]+, 387.4; 1H NMR (CDC13): 8 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[1,2,4]triazin-_yll-p-tolyl-amine by Method B

O

N N
H
(XXXII) [0139] Title product having formula (XXXII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 387.4; 1H NMR (CDC13): 6 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[1,2,4]triazin-yll-(2,4-dimethyl-phenyl)-amine by Method B
O

H
(XXXIII) [0140] Title product having formula (XXXIII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 401.4; 1H NMR (CDC13): 8 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[1,2,4]triazin-yll-(2,5-dimethyl-phenyl)-amine by Method B

0~
0 N ' N

H
(XXXIV) [0141] Title product having formula (XXXIV) was synthesized using Method B
described above. ESI-MS: [M+H]+, 401.4; 1H NMR (CDC13): 8 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-dimethoxv-phenyl)-5-methyl-benzo[1,2,4]triazin-yl1-(3,4-dimethyl-phenyl)-amine by Method B
O

O NN

H
(XXXV) [0142] Title product having formula (XXXV) was synthesized using Method B
described above. ESI-MS: [M+H]+, 401.4; 1H NMR (CDC13): 6 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-dimethoxv-phenyl)-5-methyl-benzo[1,2,41triazin-3-yll-amine by Method B

O
ibN-5~ N JOY
H

(XXXVI) [0143] Title product having formula (XXXVI) was synthesized using Method B
described above. ESI-MS: [M+H]+, 429.4; 1H NMR (CDC13): 6 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-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-(4-ethoxy-phenyl)-amine by Method B
O

N

N N
H
(XXXVII) [0144] Title product having formula (XXXVII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 417.3;1H NMR (CDC13): 8 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[1,2,4]triazin-yll-pyridin-4-yl-amine by Method B

O
OI ~ N
N N
H
(XXXVIII) [0145] Title product having formula (XXXVIII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 374.4; 1H NMR (CDC13): 8 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 [7-(3,5-dimethoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-(2,3-dimethyl-phenyl)-amine by Method B
O

1 'N
N
O

NN
H
(XXXIX) [0146] Title product having formula (XXXIX) was synthesized using Method B
described above. ESI-MS: [M+H]+, 401.4; 1H NMR (CDC13): 8 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, J= 7.6 Hz, 1 H), 8.35 (s, 1 H).

Example 36 Synthesis of [7-(3,5-dimethoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-(2-methoxy-phenyl)-amine by Method B

O
O NN
N~N
H
(XL) [0147] Title product having formula (XL) was synthesized using Method B
described above. ESI-MS: [M+H]+, 403.4; 1H NMR (CDC13): 8 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 17-(3,5-dimethoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-(4-methoxy-phenyl)-amine by Method B
O

O ba N'N /.I
N N O~
H

(XLI) [0148] Title product having formula (XLI) was synthesized using Method B
described above. ESI-MS: [M+H]+, 403.4; 1H NMR (CDC13): 6 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-benzoEl,2,4]triazin-yll-pyridin-4-yl-amine by Method A

N' N N
N N
H
(XLII) [0149] Title product having formula (XLII) was synthesized using Method A
described above. ESI-MS: [M+H]+, 442.4; 1H NMR (DMSO-d6): 6 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-phenyl)-5-methyl-benzof1,2,4]triazin-yll-(3-methoxy-phenyl)-amine by Method B
CI

CI I
N N O
H

(XLIII) [0150] Title product having formula (XXLIII) was synthesized using Method B
described above. ESI-MS: [M+H]+, 411.3; 1H NMR (DMSO-d6): 8 2.68 (s, 3 H), 3.83 (s, 3 H), 6.67 (d, J= 7.7 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[1,2,4]triazin-3-yll-(4-methoxy-phenyl)-amine by Method B

CI

N;NI O1-1 N N
H
(XLIV) [0151] Title product having formula (XLIV) was synthesized using Method B
described above. ESI-MS: [M+H]+, 411.3; 'H NMR (CDC13): 8 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[1,2,4]triazin-yll-pyridin-4-yl-amine by Method A
CI

N / N
CI \
N N
H
(XLV) [01521 Title product having formula (XLV) was synthesized using Method A
described above. ESI-MS: [M+H]+, 382.4; 1H NMR (DMSO-d6): 6 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-methyl-benzo[1,2,4]triazin-yll-pyridin-3-yl-amine by Method A

CI

N, N

CI I / \ N
N N
H
(XLVI) [01531 Title product having formula (XLVI) was synthesized using Method A
described above. ESI-MS: [M+H]+, 382.4; 1H NMR (DMSO-d6): 6 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)-phenyll-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yll-amine by Method C
9~q N \ I 0 N N
H
(XLVIII) [0154] Title product having formula (XLVIII) was synthesized using Method C
described above. ESI-MS: [M+H]+, 456.3; 1H NMR (DMSO-d6): 8 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 [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-[4-(2-morpholin-4-yl-ethoxy)-phenyll-amine by Method C

\ ( \ N, N O'-"-~'N
N N \I ~,O
H

(XLIX) [0155] Title product having formula (XLIX) was synthesized using Method C
described above. ESI-MS: [M+H]+, 470.3; 'H NMR (DMSO-d6): 8 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 [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-[4-(3-pyrrolidin-1-yl-propoxy)-phenyll-amine by Method C
/
\ I I \ 'D
N, N / I O,/\iN

N N
H

(L) [01561 Title product having formula (L) was synthesized using Method C
described above. ESI-MS: [M+H]+, 468.3; 1H NMR (CDC13): 8 2.09-2.21 (m, 10 H), 2.32 (m, H), 2.70 (s, 3H), 2.84 (m, 2 H), 3.33 (b, 2 H), 3.91 (m, 2 H), 4.10 (t, J= 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-benzo[1,2,4]triazin-yll-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyll-amine by Method C

CI
I N'N O'-"-"N
N N
H
(LI) [01571 Title product having formula (LI) was synthesized using Method C
described above. ESI-MS: [M+H]+, 494.3; 1H NMR (DMSO-d6): 8 1.90 (m, 2 H), 2.04 (m, 2 H), 2.63 (s, 3H), 3.15 (m, 2 H), 3.60 (in, 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-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-{4-[2-(4-methyl-piperazin-l-yl)-ethoxyl-phenyl}-amine by Method C
O~\N
N N
H N
(LII) [0158] Title product having formula (LII) was synthesized using Method C
described above. ESI-MS: [M+H]+, 483.3; 1H NMR (CDC13): 6 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[1,2,4]triazin-yll-[3-(2-pyrrolidin-1-yl-ethoxy)-phenyll-amine by:Method C

N;N
N N
H
(LIII) [0159] Title product having formula (LIII) was synthesized using Method C
described above. ESI-MS: [M+H]+, 454.3; 1H NMR (DMSO-d6): 8 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-phenyl)-5-methyl-benzo[1,2,41 triazin-3-yll-amine by Method C
NN N O
919( H
(LIV) [0160] Title product having formula (LIV) was synthesized using Method C
described above. ESI-MS: [M+H]+, 456.3; 1H NMR (DMSO-d6): 8 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-b enzo [1,2,41 triazin-3-ylamine CI
MeO N
N

(LV) [0161] 7-bromo-5-methyl-benzo[1,2,4]triazin-3-ylamine (33.47 mmol, 1.0 equiv), chloro-4-methoxy-2-boronic acid (50.21 mmol, 1.5 equiv), Pd(PPH3)4 (3.347 mmol, 0.1 equiv), and Na2CO3 (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[1,2,4]triazin-3-ylamine, a green solid (8.37 g, 84% yield). Rf = 0.85 (9:1 DCM/MeOH). 1H NMR (DMSO-d6): 8 3.35 (s, 6H), 7.01 (dd, J = 8.8 Hz, J = 3.0, 1H), 7.09 (d, J = 3.0 Hz, 1H), 7.48 (d, J = 8.8 Hz, 1H), 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 H
Br ,N~~
DSO
(LVI) [01621 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): 8 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, 1H), 7.70 (d, J= 7.9 Hz, 1H), 7.84 (d, J=
7.8 Hz, 1H), 8.01 (s, 1H), 8.53 (t, J= 2.3 Hz, 1H). 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-benzof 1,2,4]triazin-3-ylaminol-N-(2-dimethylamino-ethyl)-benzenesulfonamide CI

MeO N' N

N

(LVII) [0163] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]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-f7-(2-chloro-5-hydroxy-phenyl)-5-methyl-b enzo [1,2,41 triazin-3-ylaminol-N-(2-dimethylamino-ethyl)-b enzenesulfon amide CI

HO N,N

N H 0o i (LVM) [0164] 3-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamino]-N-(2-dimethylamino-ethyl)-benzenesulfonamide (0.32 mmol, 1.0 equiv) was dissolved in 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 Et20 to give 3-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamino]-N-(2-dimethylamino -ethyl)-benzenesulfonamide, as an orange solid (132.2 mg, 81% yield). 1H NMR (DMSO-d6): 8 2.71 (s, 211), 3.01 (bs, 2H), 6.87 (dd, J= 8.8 Hz, J= 2.9 Hz, 1H), 6.94 (d, J= 3.0 Hz, 1H), 7.41 (d, J= 8.8 Hz, 1H), 7.51 (d, J= 7.9 Hz, 1H), 7.64 (t, J= 8.1 Hz, 2H), 7.90 (s, 1H), 8.06 (d, J=
6.7 Hz, 1H), 8.20 (s, 1H), 8.89 (s, 1H), 9.93 (s, 1H), 11.34 (s, 1H). MS (ES+) m/z = 513.
LC retention time 2.24 min.

Example 54 Synthesis of 4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo [1,2,4]triazin-3-ylaminol-N-methyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide CI

O\ ~O
MeO N, N N
N N
H
(LIX) .

[0165] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine (0.33 mmol, 1.0 equiv), 4-bromo-N-methyl-N-(2-pyrrolidin-1-yl-ethyl)-ben.zenesulfonamide (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[1,2,4]triazin-3-ylamino]-N-methyl-N-(2-pyrrolidin-1-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-chloro-5-hydroxy-phenyl)-5-methyl-benzo [1,2,4]triazin-3-ylaminol-N-methyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide ci HO I N. N O S-N - ~N y NN
H

(LX) [01661 4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamino]-N-methyl-N-(2-pyrrolidin-1-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 Et20 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): 6 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, 1H), 6.96 (d, J= 2.9 Hz, 1H), 7.40 (d, J= 8.6 Hz, 1H), 7.87 (d, J= 8.9Hz, 2H), 7.92 (d, J=
0.9 Hz, 1H), 8.21 (d, J=1.7Hz, 1H), 8.30 (d, J= 9.0 Hz, H), 11.50 (s, 1H). MS (ES+) na/z = 553 LC retention time 2.41 min. Elemental for C27H30C12N603S 5H20 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 [1,2,41 triazin-3-yll-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyll-amine CI

MeO N, N 0 N
N N
H
(LXI) [01671 7-(2-chloro-5-methoxy-phenyl)-6-methyl-benzo[1,2,4]triazin-3-ylamine (0.67 mmol, 1.0 equiv), 1-[2-(4-broinophenoxy)-ethyl] pyrrolidine (0.005 minol, 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 [ 1,2,4]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-1-yl-ethoxy)-phenylaminol-benzo [1,2,4]triazin-7-yl}-phenol CI
HO N,N / O~~N
N N "a (LXII) [0168] [7-(2-chloro-5-methoxy-phenyl)-6-methyl-benzo[1,2,4]triazin-3-yl]-[4-(2-pyrrolidin- 1-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 Et20 to give 4-chloro-3-{6-methyl-3-[4-(2-pyrrolidin-1-yl-ethoxy)-phenylamino]-benzo[1,2,4]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): 5 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, 1H), 6.87 (dd, J= 8.8 Hz, J= 2.9 Hz, 1H), 6.95 (d, J= 11.9 Hz, 2H), 7.38 (dd, J=
8.8 Hz, J=
3.6 Hz, 1H), 7.65 (s, 1H), 7.85 (s, J= 9.1Hz, 2H), 7.98 (s, 1H), 9.93 (s, 1H), 10.67 (s, 1H). MS (ES+): m/z = 476. LC retention time 2.32 min.

Example 58 Synthesis of 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,41triazin-3-ylamine CI

MeO N' N
N~NH2 (LV) [0169] ' 7-bromo-5-methyl-benzo[1,2,4]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), and Na2CO3 (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[1,2,4]triazin-3-ylamine, as a green solid (8.37 g, 84% yield). Rf = 0.85 (9:1 DCM/MeOH). 1H NMR (DMSO-d6): 8 3.35 (s, 6H), 7.01 (dd, J = 8.8 Hz, J = 3.0, 1H), 7.09 (d, J = 3.0 Hz, 1H), 7.48 (d, J = 8.8 Hz, 1H), 7.75 (bm, 2H). MS (ES+) m/z = 303. LC retention time 3.03 min.

Example 59 Synthesis of 1-(3-bromo-benzenesulfonyl)-4-methyl-piperazine NN
N
Br (LXIV) [0170] 3-bromo-benzenesulfonyl chloride (3.914 mmol, 1.0 equiv) and 1-methyl-piperizine (7.83 minol, 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 1-(3-bromo-benzenesulfonyl)-4-methyl-piperazine, as a white solid (1.1 g. 88%
yield).

1H NMR (DMSO-d6): 8 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, 1H), 7.85 (t, J= 1.8 Hz, 1H), 7.95 (dd, J=
9.1 Hz, J
= 0.9 Hz, 1H) MS (ES +) m/z = 321 LC retention time 1.74 min.

Example 60 Synthesis of [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo [1,2,4]triazin-3-yll- [3-(4-methyl-piperazine-l-sulfonyl)-phenyll-amine CI

MeO N' N N
N H OS\0 (LXV) [0171] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamine (0.315 mmol, 1.0 equiv), 1-(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 [ 1,2,4]triazin-3-yl]-[3-(4-methyl-piperazine-l -sulfonyl)-phenyl]-amine, as a yellow solid (45 mg, 27% yield). 1H NMR (DMSO-d6): 8 2.12 (s, 3H), 2.37 (m, 4H), 2.71 (s, 3H), 2.95 (m, 4H), 3.83 (s, 3H), 7.06 (dd, J= 8.9 Hz, J= 3.1 Hz,1H), 7.14 (d, J= 3.1 Hz, 1H), 7.41 (dd, J= 6.3 Hz, J= 1.1 Hz, 1H), 7.54 (d, J
= 8.7 Hz, 1H), 7.67 (t, J= 8.1 Hz, 2H), 8.06 (dd, J= 7.6 Hz, J= 1.6 Hz, 1H), 8.27 (d, J=
1.9 Hz, 1H), 8.9 (bs, 1H). MS (ES+): rya/z = 540 LC retention time 2.70 min.

Example 61 Synthesis of 4-chloro-3-d5-methyl-3-[3-(4-methyl-piperazine-l-sulfonyl)-phenylaminol -lb enzo [1,2,4]triazin-7-yl}-phenol CI
HO I I N / I N
NN" v slN,_) H

(LXVI) [0172] [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yl]-[3-(4-methyl-piperazine-l-sulfonyl)-phenyl]-amine (0.0742 mmol, 1.0 equiv) was dissolved in 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 NaHCO3 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-1-sulfonyl)-phenylamino]-benzo[1,2,4]triazin-7-yl}-phenol, as a tan solid (24.3 mg, 62%yield). 1H NMR (DMSO-d6): 8 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, 1H), 6.95 (d, J=
2.9 Hz, 1H), 7.41 (d, J= ~.7 Hz, 1H), 7.45 (dd, J= 7.9 Hz, J= 1.7 Hz, 1H), 7.72 (t, J=
8.1 Hz, 2H), 7.91 (d, J= 1.7 Hz, 1H), 8.12 (dd, J= 8.3 Hz, J= 1.7 Hz, 1H), 8.21 (d, J=
1.9 Hz, 1H), 8.93 (d, J= 1.9 Hz, 1H), 9.99 (s, 1H), 11.46 (s, 1H). MS(ES+): m/z = 526.
Elemental for C25H36C12N608S 5H20 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[1,2,4ltriazin-3-lyamine CI

O2N N ' N
N-5~ NH2 (LXVII) [0173] 7-bromo-5-methyl-benzo[1,2,4]triazin-3-ylainine (3.35 mmol, 1.0 equiv), chloro-5-nitrophenyl-boronic acid (5.02 mmol, 1.5 equiv), Pd(PPh3)4 (0.335 mmol, 0.1 equiv), and Na2CO3 (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 Et2O wash to remove water. The crude precipitate gave 7-(2-chloro-5-nitro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine, as a dark green solid ( 1.0g, 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-b enzo [1,2,4]triazin-3-ylaminol -N-(2-pyrrolidin-1-yl-ethyl)-b enzenesulfonamide CI
O~ O

I H
N N
H
(LXVIII) [0174] 7-(2-chloro-5-nitro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine (0.65 mmol, 1.0 equiv), 4-bromo-N-(2-pyrrolidin-1-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[1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonainide, 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-benzo [1,2,4]triazin-3-ylaminol -N-(2-pyrrolidin-1-yl-ethyl)-b enzenesulfonamide CI
O~ 0 H2N I / N'N S,NN
H
N N
H
(LXIX) [0175] 4-[7-(2-chloro-5-nitro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide (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-b enzo [ 1, 2,4] triazin-3 -ylamino] -N-(2-pyrrolidin- l -yl-ethyl)-benzenesulfonamide, as a dark green solid (89.4 mg, 41% yield). 1H
NMR
(DMSO-d6): S 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, 1H), 6.65 (dd, J= 8.7 Hz, J= 2.8 Hz, 1H), 6.73 (d, J=
2.7 Hz,1H), 7.21 (d, J= 8.7 Hz, 1H), 7.83 (d, J= 8.9 Hz, 2H), 7.89 (s, 1H), 8.15 (s, 1H), 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-benzof1,2,4]triazin-3-ylaminol-phenyll-pip erazin-1-yl-methanone \ N`N / H
?IN3H

(LXX) [01761 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[1,2,4]triazin-3-ylamine (1.0 equiv, 0.33 mmol), Xantphos (0.1 equiv, 0.123 mmol), palladium acetate (0.05 equiv, 0.0165 minol), 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 Na2SO4. 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 Na2SO4a and condensed again to afford {4-[7-(2, 6-dichloro-phenyl)-5 -methyl-b enzo [ 1,2,4]triazin-3 -ylamino] -phenyl } -piperazin- l -yl-methanone, as an orange solid (26.7 mg, 34% yield). 1H NMR (DMSO-d6): 8 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, 1H), 7.67 (d, J= 8.2 Hz, 2H), 7.76 (s, 1H), 8.13 (d, J=14.4 Hz, 2H), 8.15 (s, 1H), 11.25 (s, 1H). MS (ES+) m/z = 492, LC retention time 2.45 minutes.

Example 66 Synthesis of 4-[7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylaminol-N-methyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide CI O

N N S'N'-'N
?INNooI
W
(LXXI) [0178] 4-bromo-N-methyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide (1.0 equiv, 0.82 mmol), 7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine (1.5 equiv, 1.23 mmol), Cs2CO3 (3.0 equiv, 2.46 minol), 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[ 1,2,4]triazin-3-ylamino]-N-methyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide, as a tan solid, (297.2 mg, 76%
yield). 1H
NMR (DMSO-d6): 5 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, 1H), 7.67 (d, J= 3.2 Hz, 2H), 7.79 (s, 1H), 7.87 (d, J= 3.2 Hz, 2H),8.18 (s, 1H), 8.31 (s, J=
9.0 Hz, 2H), 10.51 (bs, 1H), 11.55 (s, 1H). MS (ES+) m/z = 574, LC retention time 2.80 minutes.

Example 67 Synthesis of f 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzof 1,2,41triazin-3-yll-f4-(4-methyl-piperazine-l-sulfonyl)-phenyll-amine 0 s0 0 N; N S,N
N N
H
(LXXII) [0179] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine (1.0 equiv., 0.75 mmol), 1-(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)-phenylaminol-benzo [1,2,41 triazin-7-yll-phenol CI
~0 HO N, N /

N H
(L
XXIII) [01801 [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yl]-[4-(4-methyl-piperazine-l-sulfonyl)-phenyl]-amine (1.0 equiv., 0.339 mmol) was dissolved in 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 NaHCO3i the precipitate was filtered and rinsed with Et20 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): 8 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, 1H), 6.95 (d, J= 2.92 Hz, 1H), 7.41 (d, J= 8.79 Hz, ,1H), 7.78 (d, J= 8.89 Hz, 2H), 7.92 (m, 1H), 8.21 (d, J=1.35 Hz, 111), 8.28 (d, J= 8.88 Hz, 2H), 11.47 (s, 1H). MS (ES+):
m/z = 526 LC retention time: 2.30 minutes.

Example 69 Synthesis of 1-[2-(3-bromo-phenylsulfanyl)-ethyll-pyrrolidine Br Si~N

(LXXIV) [01811 1-(2-chloroethyl)pyrrolidine hydrochloride (1.0 equiv., 8.82 mmol) and 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 [1,2,4]triazin-3-yll-[3-(2-pyrrolidin-1-vl-ethylsulfanyl)-phenyll-amine CI
O N,N

N N S No H

(LXXV) [01821 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamine (1.0 equiv., 083 mmol), 1-[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-chloro-3-{5-methyl-3-f3-(2-pyrrolidin-1-yl-ethylsulfanyl)-phenylaminol-benzo [1,2,4]triazin-7-yll-phenol CI

HO NI N ~/
N N S
H

(LXXVI) [01831 [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yl]-[3-(2-pyrrolidin-1-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 BBr3 (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 Et20 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= 0.321H NMR (DMSO-d6): 8 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, 1H), 6.93 (s, 1H), 7.03 (d, J= 7.42 Hz, 1H), 7.34 (t, J= 7.58 Hz, 1H), 7.40 (d, J= 8.65 Hz, 1H), 7.77 (d, J= 7.95 Hz, 1H), 8.16 (s, 2H), 9.93 (s, 1H), 11.01 (s, 1H). MS (ES+): in/z = 493 Example 72 Synthesis of 4-f7-(2-chloro-5-methoxy-phenyl)-6-methyl-benzo [1,2,4]triazin-3-ylaminol-N-methyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide CI

O N. N al O NN /
N N
H
(LXXVII) [01841 7-(2-chloro-5-methoxy-phenyl)-6-methyl-benzo[ 1,2,4]triazin-3-ylamine (1.0 equiv., 0.28 mmol), 4-bromo-N-methyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide (1.5 equiv., 0.42 minol), 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 [ 1,2,4]triazin-3-ylamino]-N-methyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide as described in the next example.

Example 73 Synthesis of 4-[7-(2-chloro-5-hydroxy-phenyl)-6-methyl-benzo [1,2,41triazin-3-ylaminol-N-methyl-N-(2-pyrrolidin-1-vl-ethyl)-benzenesulfonamide CI
O\ ,O
HO NcN / N
N N
H
(LXXVIII) [01851 4-[7-(2-chloro-5-methoxy-phenyl)-6-methyl-benzo[ 1,2,4]triazin-3-ylamino]-N-methyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulforamide (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 NaHCO3a 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= 0.191H NMR (DMSO-d6): 5 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, 1H), 6.91 (dd, J= 2.91 Hz, 8.61 Hz, 1H), 7.40 (d, J= 8.81 Hz, 1H), 7.79 (s, 1H), 7.83 (d, J= 8.95 Hz, 2H), 8.11 (s, 1H), 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-ethoxy)-phenylaminol-benzo[1,2,4]triazin-7-y1}-phenyl ester IOI CI
O N / N
I N.

N N "a (LXXIX) [01861 To a solution of 4-chloro-3-{5-methyl-3-[4-(2-pyrrolidin-1-yl-ethoxy)-phenylamino]-benzo[1,2,4]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 NaHCO3i '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) : 6 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, 1H), 7.41 (d, J
= 2.80 Hz, 1H), 7.67 (d, J= 8.75 Hz, 1H), 7.85 (m, 1H), 7.93 (d, J= 9.05 Hz, 2H), 8.17 (m, 1H), 10.83 (s, 1H). 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-1-yl-ethoxy)-phenylaminol-benzo[1,2,4ltriazin-7-yl}-phenyl ester 0 N: N

N N
H
(LXXX) [0187) The title product was synthesized and had the following characteristics. 1H NMR

(DMSO-d6) : 8 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, 1H), 7.29 (t, J= 8.45 Hz, 2H), 7.43 (d, J= 2.74 Hz, 1H), 7.48 (d, J= 9.58 Hz, 1H), 7.68 (d, J= 8.69 Hz, 1H), 7.92 (d, J=
11.72 Hz, 2H), 8.23 (s, 1H). 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-1-yl-ethoxy)-phenylaminol-benzo[1,2,4]triazin-7-yl}-phenyl ester N N/ 0~~ N
\
H
(LXXXI) [0188] The title product was synthesized and had the following characteristics. 1H
NMR (DMSO-d6) : 8 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, 1H), 7.58 (d, J= 2.8 Hz, 1H), 7.72 (d, J= 8.7 Hz, 1H), 7.88 (d, J=
0.85 Hz, 1H), 7.92 (d, J= 9.15 Hz, 2H), 8.05 (d, J= 6.55 Hz, 2H), 8.21 (d, J=
1.75 Hz, 1H), 10.83 (s, 1H). MS (ES+) in/z =594.5, LC retention time: 3.08 minutes.

Example 77 Synthesis of 2,2-dimethyl-propionic acid 4-chloro-3-{5-methyl-3-f4-(2-pyrrolidin-1-yl-ethoxy)-phenylaminol-benzo[1,2,4ltriazin-7-yll-phenyl ester O CI

N N
H
(LXXXII) [0189] The title product was synthesized and had the following characteristics. 1H

NMR (DMSO-d6) : 8 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, 1H), 7.39 (d, J= 2.7 Hz, 1H), 7.67 (d, J= 8.75 Hz, 1H), 7.85 (d, J=
0.9 Hz, 1H), 7.93 (d, J= 9.15 Hz, 2H),'8.19 (d, J=1.65 Hz, 1H), 10.84 (s, 1H). MS
(ES+) m/z =560.4, LC retention time: 2.98 minutes.

Example 78 Synthesis of Isobutyric acid 4-chloro-3-{5-methyl-3-[4-(2-pyrrolidin-l-yl-ethoxy)-phenylaminol-benzo[1,2,4ltriazin-7-yl}-phenyl ester CI
O
O I ~= N; N / O N
'ji N N
H
(LXXXIII) [0190] The title product was synthesized and had the following characteristics. 1H
NMR (DMSO-d6) : 8 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, 14), 7.41 (d, J= 2.85 Hz, 1H), 7.67 (d, J= 8.8Hz, 1H), 7.85 (d, J=
1.1 Hz, 1H), 7.93 (d, J= 9.05 Hz, 2H), 8.18 (d, J=1.85 Hz, 1H), 10.84 (s, 1H).
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 Br eN
N

(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 HC1(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, 1H NMR
(DMSO-d6): 6 7.46(d, J=9.1 Hz, 1H), 7.88 (dd, J= 9.0 Hz, J= 2.4 Hz, 1H), 8.25 (d, J=
2.3 Hz, 1H).
Example 80 Synthesis of 7-bromo-5-nitro-l-oxy-benzo[1,2,4]triazin-3-ylamine OE) Br-. NO, N
N-5:~ NH2 (LXXXV) [0192] In a 1000 mL round bottom flask, 7-bromo-l-oxy-benzo[1,2,4]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+): in/z = 288.0, LC retention time: 2.39 min. Rf=
0.65, 30% EtOAc/ hexanes. 1H NMR (DMSO-d6): 8 7.99 (bs, 2H), 8.49 (d, J= 2.4 Hz, 1H), 8.59 (d, J= 2.4 Hz, 1H).

Example 81 Synthesis of 7-(2,6-dimethyl-phenyl)-5-nitro-l-oxy-benzo[1,2,4]triazin-3-ylamine '?~q O

(LXX(VI) [0193] An oven dried 25 mL round bottom flask was charged with 7-bromo -5 -nitro- 1 -oxy-benzo[1,2,4]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(0) (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+): ;n/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-oxy-benzo[1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide 0 O` 0 IqN,N 1SNN
H
N N

(LXXXVII) [01941 In a dry 25 mL round bottom flask, 7-(2,6-dimethyl-phenyl)-5-nitro-l-oxy-benzo[ 1,2,4]triazin-3-ylamine (0.069 g, 0.22 mmol, 1 equiv), 4-bromo-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide (0.11 g, 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 mL) 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 of 4-[5-amino-7-(2,6-dimethyl-phenyl)-benzo[1,2,4]triazin-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide OõO
N " N Ni~N
H
N N

(LXXXVIII) [01951 4- [ 7-(2, 6-dimethyl-phenyl)-5 -nitro- 1 -oxy-benzo [ 1,2,4] triazin-3 -ylamino] -N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonarnide (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-[4-(2-pyrrolidin-1-vl-ethylsulfamoyl)-phenylaminol-benzo [1,2,4]triazin-5-yl}-acetamide TFA salt 0"0 N, N S~N'-~N
N N H
Iq HN '~O H
(LXXXIX) [01961 A 0 C solution of 4-[5-amino-7-(2,6-dimethyl-phenyl)-benzo[1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide (0.045 g, 0.087 mmol, I
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 desired product as a yellow powder (0.011 g, 23% yield). MS (ESI+): sn/z = 560.4, LC
retention time: 2.67 min.1H NMR (DMSO-d6): S 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, 1H), 7.87 (d, J=8.9 Hz, 2H), 7.89 (t, J=6.1 Hz, 1H), 7.94 (d, J=1.8 Hz, 1H), 8.26 (d, J=8.9 Hz, 3H), 9.56 (bs, 1H), 9.84 (s, 1H) 11.48 (s, 1H).

Example 85 Synthesis of N-[3-{4-[acetyl-(2-pyrrolidin-1-yl-ethyl)-sulfamoyll-phenylamino}-7-(2,6-dimethyl-phenyl)-b enzo [ 1,2,41 triazin-5-yll -acetamide N.N S N,- N~

N N
HN O H
(XC) [01971 A 0 C degree solution of 4-[5-amino-7-(2,6-dimethyl-phenyl)-benzo[1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-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): S 1.87-1.89 (m, 2H), 2.04-2.08 (m, 8H), 2.29 (s, 3H), 2.36 (s, 3H), 3.09-3.11 (in, 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 (m, 1H), 7.96 (d, J=1.6 Hz, 1H), 8.04 (d, J=9.0 Hz, 2H), 8.26-8.30 (m, 3H), 9.73 (bs, 1H), 9.85 (s, 1H) 11.60 (s, 1H).

Example 86 Synthesis of 4-[7-(2-chloro-5-metho)y-phenyl)-5-methyl-benzo [1,2,41 triazin-3-ylaminol -N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide OO
\ , O I N " N S,N ,, N
~, ~I H
N N
H
(XCI) [01981 In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine (0.157 g, 0.523 minol, 1 equiv), 4-bromo-N-(2-pyrrolidin-1-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+): fn/z = 553.3, LC retention time: 2.77 min.

Example 87 Synthesis of 4-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl-benzof1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide TFA
salt CI
0"0 HO I N; N / SN
H Ili, N N \
H
(XCIl) [0199] 4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-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): 6 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, 1H), 6.89 (dd, J=8.7 Hz, J=2.9 Hz, 1H), 6.94 (d, J=2.9 Hz, 1H), 7.41 (d, J=8.7 Hz, 1H), 7.83 (bs, 1H), 7.87 (d, J=8.8 Hz, 2H), 7.92 (s, 1H), 8.22 (d, J=1.7 Hz, 1H), 8.26 (d, J=8.8 Hz, 2H), 9.55 (bs, 1H), 9.96 (s, 1H) 11.44 (s, 1H).

Example 88 Synthesis of 3-(3-amino-5-methyl-benzo[1,2,4]triazin-7-yl)-4-chloro-henol CI

HO N. N
N&NH2 (XCIII) [0200] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]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. 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-15-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl-benzo(1,2,4ltriazin-3-ylaminol-pyridin-2-ylfl-acetamide TFA salt CI

HO N N N
/
N N
H
(XCIV) [02011 In a dry 25 mL round bottom flask, 3-(3-amino-5-methyl-benzo[1,2,4]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): 6 2.09 (s, 3H), 2.65 (s, 3H), 6.87 (dd, J=8.7 Hz, J=2.9 Hz, 1H), 6.93 (d, J=2.9 Hz, 1H), 7.40 (d, J=8.7 Hz, 1H), 7.86 (s, 1H), 8.11 (d, J=8.9 Hz, IH), 8.16 (d, J=1.7 Hz, 1H), 8.39 (dd, J=9.0 Hz, J=2.7 Hz, 1H), 8.93 (d, J=2.5 Hz, 1H), 9.92 (bs, 1H), 10.47 (s, 1H), 11.05 (s, 1H).

Example 90 Synthesis of 1-[2-(3-bromo-phenoxy)-ethyll-pyrrolidine i~NJ
Br O

(XCV) [0202] 3-bromo-phenol (5.34 g, 30.9 mmol, 1 equiv), 1-(2-chloro-ethyl)-pyrrolidine (5.24 g, 30.9 mmol, 1 equiv) and potassium carbonate (K2CO3) (34 g, 24.7 mmol, 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): S 1.65-1.69 (m, 4H), 2.72 (s, 1H), 2.75 it, J= 5.8 Hz, 2H), 2.88 (s, 1H), 4.06 (t, J= 5.8 Hz, 2H), 6.95 (dd, J=
8.35 Hz, 1H), 7.11 (d, J=7.7, 1H), 7.13-7.14 (m, 1H), 7.22 (t, J= 8.1Hz, IH).

Example 91 Synthesis of [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo 11,2,4ltriazin-3-yll- [3-(2-pyrrolidin-1-yl-ethoxy)-phenyll-amine CI

O N; N

N N
H
(XCVI) [0203] In a dry 25 mL round bottom flask, 7-(2-ch.loro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine (0.113 g, 0.377 mmol, 1 equiv), 1-[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-[3-(2-pyrrolidin-1-yl-ethoxy)-phenylaminol-benzo[1,2,4]triazin-7-yl}-phenol TFA salt CI
HO \ N\
N~
N N O
.H
(XCVII) [0204] [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]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 (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. HPLC purification provided TFA salt of desired product as a yellow solid (0.008 g, 7 % yield). MS (ESI+): 'n/z = 476.1, LC retention time:
2.64 min.
1H NMR (DMSO-d6): 5 1.88-1.91 (in, 2H), 2.04-2.07 (in, 2H), 2.68 (s, 3H), 3.17-3.20 (m, 2H), 3.64-3.66 (m, 4H), 4.36 (t, .1=4.7 Hz, 2H), 6.74 (dd, J=8.1 Hz, J=2.3 Hz, 1H), 6.88 (dd, J=8.5 Hz, J=2.8 Hz, I H), 6.93 (d, J=2.9 Hz, 1H), 7.35 (t, J=8.2 Hz, 1H), 7.41 (d, J=8.7 Hz, 1H), 7.55 (d, J=8.7 Hz, 1H), 7.88 (s, 1H), 7.95-7.96 (m, 1H), 8.17 (d, J=1.7 Hz, 1H), 9.79 (bs, 1H), 9.95 (s, 1H), 11.04 (s, 1H).

Example 93 Synthesis of {4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo [1,2,4]triazin-3-ylaminol-phenyl}-(4-methyl-piperazin-1-yl)-methanone CI O
/ I

NN N
H

(XCVIII) [02051 In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamine (0.282 g, 0.94 mmol, 1 equiv), (4-bromo-phenyl)-(4-methyl-piperazin-1-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-phenyl)-5-methyl-benzo [1,2,4]triazin-3-ylaminol-phenyl}-(4-methyl-piperazin-1-y1)-methanone TFA
salt CI
d HO NN / N
N N e (XCIX) [02061 {4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamino]-phenyl}-(4-methyl-piperazin-1-yl)-methanone (0.139 g, 0.276 mmol, 1 equiv) was diluted with 10 rnL 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. 'H NMR (DMSO-d6): 8 2.69 (s, 3H), 2.84 (s, 3H), 3.22 (bs, 2H), 6.88 (dd, J=8.8 Hz, J=2.9 Hz, 1H), 6.93 (d, J=2.9 Hz, 1H), 7.41 (d, J=8.7 Hz, 1H), 7.55 (d, .1=8.7 Hz, 2H), 7.89 (s, 1H), 8.14 (d, J=8.7 Hz, 2H), 8.19 (d, J=1.3 Hz, 1H), 9.88 (bs, 1H), 9.95 (bs, 1H), 11.24 (s, 1H).

Example 95 Synthesis of 5-[7-(2-chloro-5-methoxy-phenyl)-5-methvl-benzof1,2,4]triazin-3-ylaminol-pyridine-2-carboxylic acid (2-pyrrolidin-l-vl-ethyl)-amide CI

0 N.,N NNo NN ", N H
H

(C) [0207] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamine (0.090 g, 0.299 mmol, 1 equiv), 5-bromo-pyridine-2-carboxylic acid (2-pyrrolidin-1-yl-ethyl)-amide (0.134 g, 0.449mmo1, 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-benzof1,2,4]triazin-3-ylaminol-pyridine-2-carboxylic acid (2-pyrrolidin-1-yl-ethyl) amide HC1 salt N
HO NcN / N/-i H
NN N
H

(cn [02081 5-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-b enzo [ 1,2,4]triazin-3-ylamino] -pyridine-2-carboxylic acid (2-pyrrolidin-1-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 HC1 in diethyl ether (0.16 mL, 0.317 mmol, 2 equiv). The solvents were evaporated leaving desired product as HC1 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. 'H NMR (DMSO-d6): 6 1.86-1.88 (m, 2H), 1.99-2.02 (in, 2H), 2.70 (s, 3H), 3.01-3.05 (m, 2H), 3.33-3.37 (in, 2H), 6.90 (dd, J=8.7 Hz, J=2.9 Hz, IH), 6.97 (d, J=2.9 Hz, 1H), 7.41 (d, J=8.8 Hz, 1H), 7.92-7.93 (m, 1H), 8.12 (d, J=8.7 Hz, 1H), 8.22 (d, J=1.5 Hz, 1H), 8.51 (dd, J=8.6 Hz, J=2.5 Hz, 1H), 9.06 (t, J=5.9 Hz, 1H), 9.43 (d, J=2.4 Hz, 1H), 10.17 (bs, 1H), 11.50 (s, 1H).

Example 97 Synthesis of 17-(2-chloro-5-methoxy-phenyl)-5-methyl-benzof 1,2,41triazin-3-yll-[4-(3-pyrrolidin-1-vl-propane-l-sulfonyl)-phenyll-amine CI
0 I / \ NN O S~ND
N N
H
(CII) [0209] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamine (0.0361 g, 0.122 mmol, 1 equiv), 1-[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-1-yl-propane-l-sulfonyl)-phenylaminol-benzo[1,2,4]triazin-7-yl}-phenol TFA salt CI

N ~S
HO ~ N
'N
N~N'I/
H
(CIII) [02101 [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-yl]-[4-(3-pyrrolidin-1-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+): na/z = 538.1, LC retention time: 2.35 min. 'H NMR (DMSO-d6): S 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, 1H), 6.94 (d, J=2.8 Hz, 1H), 7.41 (d, J8.7 Hz, 1H), 7.95 (d, J=9.0 Hz, 1H), 8.23 (d, J=1.6 Hz, 1H), 8.31 (d, J=8.9 Hz, 2H), 9.61 (bs, 1H), 9.98 (s, 1H), 11.52 (s, 1H).

Example 99 Synthesis of N-.[3-(3-amino-5-methyl-benzo[1,2,41triazin-7-vl)-phenyll-methanesulfonamide ,-IS,N I / N' N
H I/
N~NH2 (CIV) [02111 An oven dried 25 mL round bottom flask was charged with 7-bromo-5-methyl-benzo[1,2,4]triazin-3-ylamine (0.889 g, 3.72 mmol, 1 equiv), 3-(methylsulfonylarnino) 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(0) (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(1,2,41triazin-3-Ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide TFA
salt O O O\ O
%S\ H N, ~S. N
N I H
I /
N N
H
(CV) [02121 In a dry 25 mL round bottom flask, N-[3-(3-amino-5-methyl-benzo[1,2,4]triazin-7-yl)-phenyl]-iethanesulfonamide (0.12 g, 0.365 mmol, 1 equiv), 4-bromo-N-(2-pyrrolidin-1-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): S 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 (in, 2H), 3.56(bs, 2H), 7.29 (dd, J=8.0 Hz, J=1.6 Hz, 1H), 7.52 (t, J=7.9 Hz, 1H), 7.63 (d, J=8.0 Hz, 2H), 7.66 (bs, 1H), 7.83-7.88 (m, 3H), 8.18 (bs, 1H), 8.25 (d, J=8.9 Hz, 2H), 8.43 (d, J=1.7 Hz, 1H), 9.57 (bs, 1H), 9.92 (s, 1H) 11.43 (s, iH).

Example 101 Synthesis of 5-methyl-7-(3-nitro-phenyl)-benzo[1,2,4]triazin-3-ly amine O2N N' N

(CVI) [0213] An oven dried 25 mL round bottom flask was charged with 7-bromo-5-methyl-benzo[1,2,4]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(0) (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[1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide 02N N. N / S,NN
I H
N N
H
(CVII) [0214] In a dry 25 mL round bottom flask, 5-methyl-7-(3-nitro-phenyl)-benzo[1,2,4]triazin-3-ylamine (0.223 g, 0.7935 mmol, 1 equiv), 4-bromo-N-(2-pyrrolidin-1-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[1,2,41triazin-3-ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide TFA
salt N N, N j S,NN
~ I H
H N N
H
(CVIII) [0215] 4-[7-(3-amino-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-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): 8 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, 1H), 7.19 (m, 2H), 7.26-7.29 (m, 1H), 7.88 (d, J=8.8 Hz, 1H), 7.94 (t, J=7.7 Hz, 1H), 8.16 (d, J=1.7 Hz, 1H), 8.25 (d, J=8.8 Hz, 2H), 8.36 (d, J=1.7 Hz, 1H), 11.44 (s, 1H).

Example 104 Synthesis of 4-[7-(3-amino-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide O
H2N N;N / S N
H
N N
H
(CIX) [0216] ' 4-[5-methyl-7-(3-nitro-phenyl)-benzo[1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin- 1-yl-ethyl)-benzenesulfonamide (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): 8 1.64 (bs, 4H), 2.72 (s, 3H), 2.87 (bs, 2H), 5.26 (s, 2H), 6.65 (d, J=7.8 Hz, 1H), 7.00 (d, J=8.1 Hz, 1H), 7.05 (m, 1H), 7.18 (t, J=7.8 Hz, 1H), 7.83 (d, J=8.8 Hz, 2H), 8.12 (s, 1H), 8.21 (d, J=8.8 Hz, 2H), 8.27 (d, J=1.7 Hz, 1H), 11.44 (s, 1H).

Example 105 Synthesis of 4-(4-bromo-benzenesulfonyl)-piperazine-l-carboxylic acid tert-butyl ester O'S O
Z
'~ N~
LN O
Br O
(CX) [0217] To a stirring, 0 C solution of piperazine-l-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[1,2,4]triazin-3-ylaminol-benzenesulfonyl}-piperazine-l-carboxylic acid tert-butyl ester CI
S
N a, ,N N

CI ~N O
N H Y
O
(CXI) [0218] In a dry 25 mL round bottom flask, 7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,4]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+): nz/z = 529.0 (M -100), LC retention time:
3.51min. 1H NMR (DMSO-d6): 5 1.33 (s, 9H), 2.72 (s, 3H), 2.87 (m, 4 H), 3.4 (m, 4H), 7.5 (m, 1H), 7.67 (d, .I=8.7 Hz, 2H), 7.78 (m, 3H), 8.18 (d, J=1.7 Hz, 1H), 8.30 (d, J=8.8 Hz, 2H), 11.53 (s, 1H).

Example 107, Synthesis of [7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yll-[4-(piperazine-l-sulfonyl)-phenyll-amine TFA salt CI
O~ ~O
N'zN S'N--') CI I / \ I ~NH
N N
H
(CXII) [0219] 4- {4-[7-(2,6-dichloro-phenyl)-5-methyl-benzo[ 1,2,4]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, 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+): zn/z = 528.9, LC
retention time:
2.70 min. 1H NMR (DMSO-d6): 5 2.72 (s, 3H), 3.13 (m, 4 H), 3.22 (m, 4H), 7.53 (m, 1H), 7.68 (d, J=8.2 Hz, 2H), 7.81 (d, J=1.6 Hz, 1H), 7.84 (d, J=8.7 Hz, 2 H), 8.20 (d, J=1.7 Hz, 1H), 8.32 (d, J=8.8 Hz, 2H), 8.62 (bs, 2H), 11.53 (s, 1H).

Example 108 Synthesis of 4-bromo-N-isopropyl-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide O S N/

Br (CXIII) [0220] 4-bromo-N-isopropyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide (1.03 g, 3.09 mmol, 1 equiv), 2-bromo-propane (3.8 g, 31 rnmol, 10 equiv) and cesium carbonate (2 g, 6.2 rnmol, 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. Rf = 0.48 (10% MeOH:DCM).

Example 109 Synthesis of [7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,41triazin-yll-[4-(piperazine-l-sulfonyl)-phenyll-amine HCl salt CI

I/ 'D
N; N SN

CI NN
H
(CXIV) [0221] In a dry 25 mL round bottom flask, 7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine (0.162 g, 0.533 mmol, 1 equiv), 4-bromo-N-isopropyl-N-(2-pyrrolidin-1-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(diphenylphosphino)-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 HCl 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 inin.
1H NMR (DMSO-d6): 6 0.98 (d, 6H), 1.88 (in, 2 H), 2.01 (m, 2H), 2.72 (s, 3H), 3.02-3.08 (m, 2H), 3.37 (m, 2H), 3.46 (in, 2H), 3.62 (m, 2H), 4.02 (m, 1H), 7.53 (m, 1H), 7.67 (d, J=8.1 Hz, 2H), 7.79 (s, 1H), 7.92 (d, J=8.7 Hz, 2H), 8.18 (d, J=1.6 Hz, 1H), 8.27 (d, J=8.7 Hz, 2H), 10.7 (in, 1H), 11.51 (s, 1H).

Example 110 Synthesis of 4-[7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide HCl salt CI
N, N S N N /
CI H
NN
H
(CXV) [0222] In a dry 25 mL round bottom flask, 7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine (0.193 g, 0.633 mmol, 1 equiv), 4-bromo-N-(2-pyrrolidin-1-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
HCl 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): 8 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, 1H), 7.67 (d, J=8.7 Hz, 2H), 7.79 (m, 1H), 7.89 (dd, J=8.7 Hz, J=2.9 Hz, 1H), 7.95 (t, J=6.1 Hz, 1H), 8.18 (d, J=1.7 Hz, 1H), 8.26 (d, J=8.8 Hz, 2H), 10.37 (bs, 1H), 11.44 (s, 1H).

Example 111 Synthesis of 7-(2-chloro-5-methoxy-phenyl)-5-methyl-b enzo [1,2,41 triazin-3-ylamine CI

O N, N 'NN
N N
H
(CXVI) [0223] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamine (0.214 g, 0.713 mmol, 1 equiv), 4-bromo-N-isopropyl-N-(2-pyrrolidin-,1-yl-ethyl)-benzenesulfonamide (0.321 g, 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 [1,2,4]triazin-3-ylaminol-N-isopropyl-N-(2-pyrrolidin-1-vl-ethyl) benzenesulfonamide HCl salt CI

HO N N / I S,N N
N N
H
(CXVII) [0224] 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]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): S 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, 1H), 6.89 (dd, J=8.7 Hz, J=2.8 Hz, 1H), 6.96 (d, J=2.8 Hz, 1H), 7.40 (d, J=8.7 Hz, 1H), 7.91-7.94 (m, 3H), 8.21 (d, J=1.5 Hz, 1H), 8.26 (d, J=8.9 Hz, 2H), 10.0 (s, 1H), 11.17 (bs, 1H) 11.47 (s, 1H).

Example 113 Synthesis of 4-(4-bromo-benzenesulfonyl)-piperidine-l-carboxylic acid benzyl ester OõO
S
B
O
(CXVIII) [0225] 4-(4-bromo-phenylsulfanyl)-piperidine-1-carboxylic acid benzyl ester (1.2 g, 2.96 mmol, 1 equiv) and sodium perborate tetrahydrate (NaBO3.4112O) (1.36 g, 8.87 mmol, 3 equiv) were heated to 55 C in HOAc 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-phenylsulfanyl)-piperidine-l-carboxylic acid benzyl ester S
\ I Nu O \
Br II
O
(CXIX) [0226] 4-bromo-benzenethiol (0.634 g, 3.36 mmol, 1 equiv), 4-bromo-piperidine-l-carboxylic acid benzyl ester (1g, 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-{4-17-(2-chloro-5-methoxy-phenyll-5-methyl-benzo(1,2,41triazin-3-ylaminol-benzenesulfonyl}-piperidine-l-carboxylic acid benzyl ester CI
O\ s0 O \ ~ \ N\ IN S 0-N H
O
(CXX) [02271 In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]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-sulfonyl)-phenylaminol-benzo[1,2,41triazin-7-yl}-phenol HCl salt / CI
HO I N, N OSO
OH
N N
H
(CXXI) [0228] 4- {4-[7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]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): 6 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, 1H), 6.89 (dd, J=8.8 Hz, J=2.9 Hz, 1H), 6.95 (d, J=2.9 Hz, 1H), 7.41 (d, J=8.7 Hz, 1H), 7.88 (d, J=8.8 Hz, 2H), 7.93 (s, 1H), 8.22 (d, J=1.7 Hz, 1H), 8.33 (d, J=8.8 Hz, 2H), 10.01 (s, 1H), 11.56 (s, 1H).

Example 117 Synthesis of 1-[2-(4-bromo-phenylsulfanyl)-ethyll-pyrrolidine / I S~\N
Br (CXXII) [0229] 4-bromo-benzenethiol (7.05 g, 37.3 mmol, 1 equiv), 1-(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-chloro-5-methoxy-phenyl)-5-methyl-benzo f 1,2,4]triazin-3-yll-[4-(2-pyrrolidin-1-yl-ethylsulfanyl)-phenyll-amine CI

0 N' N SN
N N
H
(CXXIII) [0230] In a dry 25 mL round bottom flask, 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine (0.625 g, 2.08 mmol, 1 equiv), 1-[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-pyrrolidin-1-vl-ethylsulfanyl)-phenylaminol-benzo[1,2,41triazin-7-yl}-phenol TFA salt CI

HO N' N S'--~ N
N N
H
(CXXIV) [0231] [7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-yl]-[4-(2-pyrrolidin- 1-yl-ethylsulfanyl)-phenyl]-amine (0.22 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
NMR (DMSO-d6): S 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, 1H), 6.93 (d, J=2.9 Hz, 1H), 7.41 (d, J=8.7 Hz, 1H), 7.51 (d, J=8.8 Hz, 2H), 7.87 (s, 1H), 8.07 (d, J=8.8 Hz, 2H), 8.17 (d, J=1.7 Hz, 1H), 9.64 (bs, 1H), 9.95 (bs, 1H), 11.11 (s, 1H).

Example 120 Synthesis of 4-(3-(4-(2-(pyrrolidin-1-yl)ethoxv)phenylamino)-5-methylbenzo f el f 1,2,4ltriazin-7-yl)-3,5-dimethylphenol HO

N N
N N
H
(CXXV) [0232] The title compound was synthesized as shown by the reaction scheme (CXXVI).

MeO Me Br Me O
~N, N B(OH)2 MeO
/ .,~NH2 + Me 2 Me Pd2(dba)3/Ph3P Me 1NNH2 1 K2C03/DMA 3 Me H2/10% Pd-C MeO Me HO Me N; N BBr3/CH2CI2 I / l \ NN
THF-MeOH
Me N5~NH2 Me N"~NH2 4 Me 5 Me N
~ O _j / 6 HO Me-ND
Br N,N I O

Pd2(dba)3/BINAP/KOBut Me H /
Me (CXXVI) [0233] To a solution of the 7-bromo-5-methylbenzo[ 1,2,4]triazin-3-yl-amine- 1 -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 K2C03 (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 CH2C12 was used to extract the product.
The crude product was purified by chromatography (Si02/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 mL, 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 (MgSO4) to afford 5.
[0234] As further shown by scheme (CCXXVI), to a suspension of 5 in dry PhMe (1 mL) was added 1-(2-(4-bromophenoxy)ethyl)pyrrolidine 6 (44.4 mg, 0.16 mmol), rac-2,2'-bis(diphenyl-phosphino)-1,1'-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-C 18, 200 x 40 nun, 7 ,um, starting from 30% solvent A (0.1% TFA in H20) to 100% solvent B (0.1% TFA in McCN), flow rate 40 ml/min, A = 282 nm). Fractions containing the products were combined to yield the title product (CXXV) as its TFA salt (1.3 mg, 3%). 'H NMR (CDC13): 8 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 (m, 4H); 4.40 (t, J= 4.9 Hz, 2H); 6.65 (s, 1H); 7.30 (s, 1H); 7.48 (d, J= 7.9 Hz, 2H); 7.61 (s, 1H); 8.06 (s, 1H); 8.09 (d, J= 7.9 Hz, 1H); 8.92 (br, IH). MS (ESI+): to/z =
470.3.

Example 121 Synthesis of [7-(3,5-dimethyl-isoxazol-4-yl)-5-methyl-benzof 1,2,41 triazin-3-yll-(4-methoxy_phenyl)-amine O
N N; OMe N NH
Me (CXXVII) [0235] The reaction scheme (CXXVIII) includes an illustration of the synthesis of the title compound.

OMe O /
N N H2N 8 N N, OMe BBr3/CH2CI2 N N

Me Me NCO
,,Me J
N N N l OH Br~N 10 Me N
N N
~NH CSZCO3/Dioxane/Reflux HN Me _ O\^N

9 CF3CO2HN-Me (CXXVIII) [0236] The mixture of 5-methyl-7-(3,5-dimethylisoxazol-4-yl)benzo[e][1,2,4]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 overnight. The product was purified by chromatography (Si02/ CH2C12) and the title product (48.7 mg, 32%) was obtained as a yellow solid. 1H NMR (CD2C12): 6 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, 1H); 7.83 (d, J= 8.9 Hz, 2H); 8.03 (s, 1H); 8.10 (br, 1H). MS (ESI+):m/z = 362.1.

Example 122 Synthesis of 17-(3,5-dimethyl-isoxazol-4-yl)-5-methyl-benzo f 1,2,4ltriazin-3-yll-f4- f 2-(4-methyl-piperazin-1-yl)-ethoxyl -phenyl}-amine '1O
N~ I

\ N. r--'\N-Me Me (CXXIX) [02371 The synthesis of the title compound is also illustrated by the reaction scheme (CXXVIII). Compound (CXXVII) obtained as described in Example 121 (48.7 ming, 0.13 mmol) was demethylated by using BBr3/ CH2C12 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 1-(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 ,Um, starting from 30%
solvent A (0.1 % TFA in H2O) to 100% solvent B (0.1 % TFA in MeCN), flow rate ml/min, A = 288 nm). Fractions containing the products were combined. The title compound was obtained (0.8 mg, 3%) as its TFA salt. 1H NMR (CD2Cl2):S 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, 1H); 7.75 (d, J= 9.0 Hz, 2H); 7.94 (s, 1H). MS (ESI+): m/z = 474.2.

Example 123 Synthesis of 3-(S-methyl-7-(2,6-dimethylphenyl)benzo[e][1,2,4]triazin-3-ylamino)b enzene-sulfonamide N'N
N H /

SAO O

(CXXX) [02381 The title compound was synthesized as shown by the reaction scheme (CXXXI).

Me \
N: N Br I S02NH2 Me N
12 ,N
Me N NH2 Pd2(dba)3/BINAP/KOBuf Me NH S02NH2 Me Me (CXXXI) 102391 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(diphenylphosphino)-1,1'-binaphthyl (BINAP, 141 mg, 0.23 mmol), Pd2 (dba)3 (69 mg, 0.075 mmol), and KOBut (170 mg, 1.51 rnmol). 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,um, starting from 30% solvent A (0.1 % TFA in H2O) to 100% solvent B (0.1 % TFA in MeCN), flow rate 40 ml/min, A = 288 nm). Fractions containing the products were combined. The title compound was obtained (10.4 mg, 3%) as its TFA salt. 1H
NMR
(DMSO-d6): 8 2.06 (s, 6H); 2.71 (s, 3H); 7.19 (d, J= 7.3 Hz, 2H); 7.24 (m, 1H); 7.35 (br, 2H); 7.53 (d, J= 7.9 Hz, 1H); 7.60 (t, J= 7.9 Hz, 1H); 7.65 (s, 1H); 7.97 (s, 1H);
8.05 (d, J= 7.9 Hz, 1H); 8.85 (s, 1H); 11.24 (s, 1H). MS (ESI+): in/z = 420.1.

Example 124 Synthesis of 3-(5-methyl-7-(2,6-dimethylphenyl)benzoleif1,2,4ltriazin-3-ylamino)-N-(2-pyrrolidin-1-yl)ethyl)benzenesulfonamide hydrochloride Me Me NN 6,0 SN~\N~
Me (CXXXII) [0240] The title compound was synthesized as shown by the reaction scheme (CXXXIII).

/ Me N,N
~iN~ I ~\ H Me N~NH2 H2N 14 Br' A- N--'N Me 11 Br ' v 'SO CI \O
2 Et3N/CH2CI2 O
13 15 Pd2(dba)3/BINAP/KOBut Me N N H
Me N!~J- H S cN'-'--'N
Me O O
(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-1-yl)ethanamine 14 (840 mg, 7.32 rnmol) 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): b 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 (m, 1H); 7.57-7.60 (m, 3H); 8.03 (s, 2H); 8.87 (br, 1H); 8.97 (s, 1H). MS
(ESI+):
in/z = 517.4.

Example 125 Synthesis of 3-(5-methyl-7-(2,6-dimethylphenyl)benzo[e][1,2,4]triazin-3-ylamino)-N-(2-(dimethylamino)ethyl)benzenesulfonamide hydrochloride N,N
-,ill N
N N S, (CXXXIV) [0243] The title compound was synthesized as shown by the reaction scheme (CXXXV).

Me N,N
H NN~ H Me I)-NH
2 16 Br S-NN Me 11 Br' SO CI ~O
2 Et3N/CH2CI2 O
13 17 1) Pd2(db8)3/BINAP/KOBut 2) HCI

/ Me ~c: )-NC ,S\N"~N/
Me O O HCI
(CXXXV) [0244] Compound 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 compound (CXXXIV) 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): 8 2.11 (s, 6H);
2.84 (s, 9H); 3.30 (br, 2H); 3.59 (br, 211); 7.15-7.18 (m, 3H); 7.21-7.24 (m, 1H); 7.45 (d, J= 7.6 Hz, 1H); 7.56 (s, 1H); 7.84-7.86 (m, 2H); 7.93 (s, 1H); 9.06 (s, 111); 9.37 (s, 1H); 11.87 (br, 1H). MS (ESI+): m/z = 491.3.

Example 126 Synthesis of 2-(5-methyl-7-(2,6-dimethylphenyl)benzo[e][1,2,4]triazin-3-ylamino)-N-(2-(pyrrolidin-1-yl)ethyl)thiazole-4-carboxamide N N S ~HH- -N J

H
(CXXXVI) [0245] The title compound was synthesized as shown by the reaction scheme (CXXXVII).

/ I Me CO2Et Me N CO2Et N I
,N N N7\~N
N~ N ;
I UGH
Me !~NH Br S 18 Me N N S

Me 11 DMF/Pd2(dba)3/BINAP/KOBut Me 19 THE/MeOH/H20 Me C02H Nom/ / Me ONO

Me NI-N Me N-N S CF3CO2H
H CDMT/NMM/CH2Ci2 H
Me 20 Me (CXXXVII) [0246] To a solution of 11 shown by scheme (CXXXVII) (646 ing, 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)-1,1'-binaphthyl (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][1,2,4]triazin-3-ylamino)thiazole-4-carboxylate 19 was dissolved in MeOH (10 inL) and THE (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 (Si02/CH2Ci2: MeOH: NH3.H20 = 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-1,3,5-trizine (CDMT, 198 mg, 1.12 mmol) and 4-methylmophline (NMIVI, 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-C18, 200 x 40 mm, 7 ,um, starting from 30% solvent A (0.1 % TFA in H2O) to 100% solvent B
(0.1 %
TFA in MeCN), flow rate 40 ml/min, A = 282 nm). Fractions containing the products were combined. The title product (CXXXVI) was obtained (110 mg, 20%) as its TFA
salt. 1H NMR (DMSO-d6): 6 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, 1H); 7.78 (s, 1H); 7.97 (s, 1H); 8.10 (s, 1H); 8.25 (t, J= 6.1 Hz, 1H); 9.42 (br, 1H); 12.56 (s, 1H). MS (ESI+): na/z = 488.2.

Example 127 Synthesis of 3-(5-methyl-7-(2,6-dimethylphenyl)benzo[el [1,2,41triazin-3-ylamino)-N-methyl-N-(2-(pyrrolidin-1-yl)ethyl)benzenesulfonamide hydrochloride N H

(CXXXVIII) [0249] The title compound was synthesized as shown by the reaction scheme (CXXXIX).

H N ~/ 21 O ~sN LiAIH4/THF N Br / 24 HCOZEt 2 ~H 22 Reflux/18 h H 23 Et N/CH CI /RT/1 8 h Et20/RT/2 h 3 2 z Me N N
n i Me 0, O
oS 0 Me N~NH2 N,N I SNi~N
Nei N Me 11 Me N~N I HCI
Br 25 1)PhMe/ Pd2(dba)3/BINAP/KOBuf Me H
2) HCI

(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
LiAlH4. in THE (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 (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 CH2C12 (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 (CXXXVIII) 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 minol) were used to give title compound (CXXXVIII) (276 mg, 37%) as its HC1 salt. 1H NMR (DMSO-d6): 6 1.88-1.91 (rn, 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, 1H); 7.70 (s, 1H); 7.86 (d, J= 8.9 Hz, 2H); 8.01 (s, 1H); 8.31 (d, J= 8.9 Hz, 2H); 10.18 (br, 1H);
11.47 (s, IH). MS (ESI+): m/z = 531.3.

Example 128 Synthesis of f7-(2,6-dimethyl-phenyl)-5-nitro-l-oxy-benzof 1,2,41triazin-3-yll-f4-(2-pyrrolidin-1-yl-ethoxy)-phenyll-amine / N N / I N
N N

(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-d): 8 7.30 (d, J= 8.8 Hz, 1H); 7.97 (dd, J=
8.8 Hz, J= 2.0 Hz, 1H); 8.25 (d, J= 2.0 Hz, 1H). MS (ESI+): m/z = 241.
Compound 28:
1H NMR (DMSO-d6):8 8.22 (d, J= 2.3 Hz, 1H); 8.27 (d, J= 2.3 Hz, 1H). MS
(ESI+):
m/z = 286.

Br NO2 1) NH2CN/HCI/110 C/2 h Br 5 N

NH2 2) 30%NaOH/110 C/0.5 h NNH2 Me O I (OH)2 HNO3/H2SO4 Br / N,N B
Me 29 \ N"J-NH2 DME/EtOH/H20 NO2 28 Pd(Ph3P)4/Na2CO3 Reflux/18 h Me Me O Br O_ Me FC O
N'N 6 No N
N
Me N~NH2 Dioxane/Pd2(dba)3/Xant Phos 02N N HN
NO2 Cs2CO3/Reflux/4 h0 N-~ Me Me H2/Pd C Me N=N CH3COCI O 'N
/-NH 1) CH2CI2 ONH N={N O
00-0- N 2) HCI Me H N
HCI
Me NH2 (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 ((Si02/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 (Si02/
CH2C12, then CH2C12 : MeOH : NH3.H20 =100: 10 : 2.5) and afforded the title compound (CXL) (124 mg, 17%). 'H NMR (CD2C12): 8 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, 1H); 7.52 (br, 1H); 7.72 (d, J= 7.1 Hz, 2H); 8.13 (d, J= 1.9 Hz, 1H); 8.35 (d, J= 1.9 Hz, 1H). MS (ESI+): m/z = 501.3.

Example 129 Synthesis of 7-(2,6-dimethvl-phenyl)-N3-f4-(2-pyrrolidin-1-yl-ethoxy)-phenyll-benzo f 1,2,41triazine-3,5-diamine ONC) N /

N N

(CXLII) [0256] Compound (CXL) synthesized as described in Example 128 was hydrogenated (scheme (CXLI)) to afford the title compound (CXLII) (yield, 73%). 'H NMR
(CD2CI2):
8 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, 1H); 6.99 (d, J= 6.9 Hz, 2H); 7.13 (d, J= 7.6 Hz, 2H);
7.17-7.20 (in, 1H); 7.42 (d, J= 1.6 Hz, 1H); 7.71 (d, J= 6.9 Hz, 2H); 7.96 (br, 1H). MS
(ESI+): m/z = 455.4.

Example 130 Synthesis of N-(3-(4-(2-(pyrrolidin-l-yl)ethoxy)phenylamino)-7-(2,6-dimethylphenyl)benzofel-f1,2,4ltriazin-5-yl)acetamide hydrochloride Iq N'N 0, N N

r H

(CXLIII) [02571 To a solution of compound (CXLII) obtained as described in Example 129 (122 mg, 0.27 mmol) in anhydrous CH2Cl2 (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 CH2Cl2 (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 HCl salt. 1H NMR (CD2Cl2): 8 2.06-2.12 (m, 2H); 2.10 (s, 6H);
2.18-2.21 (rn, 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, 1H); 7.73 (d, J= 6.8 Hz, 2H); 7.77 (d, J= 1.8 Hz, 1H); 8.48 (s, 1H); 8.57 (br, 1H);
8.94 (s, 1H); 12.4 (br, 1H). MS (ESI+): Tn/z = 497.4.

Example 131 Synthesis of N-(3-(4-(2-(pyrrolidin-l-yl)ethoxy)phenylamino)-7-(2,6-dimethylphenyl)benzofel-11,2,4ltriazin-5-yl)methylsulfonyl amide hydrochloride N N
HN. OS 1-1 H
"0 (CXLN) [02581 The title compound was synthesized as shown by the reaction scheme (CXLV).

Me Me N~ N; O~\N~
N CH3S02CI Me NN ( HCI
Me H 1) PhMe/Reflux O~ NH "
NH2 2) HCI OAS
Me (CXLV) [02591 As shown by scheme (CXLV), to a solution of compound (CXLII) obtained as described in Example 129 (40 mg, 0.09 nunol) in anhydrous PhMe (5 mL) was added methylsulfonyl chloride (13 mg, 0.11 rmnol). The solution was heated under reflux for 18 h and solvent was removed in vacuo. The residue was dissolved in CH2C12 (2 mL) and Et20 was added to precipate product. The product was purified by HPLC. The title compound (CXLIV) (30 mg, 60%) was obtained as its HCl salt. 1H NMR (CD2C12): S
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, 1H); 7.69 (d, J= 1.8 Hz, 1H); 7.88 (d, J= 1.8 Hz, 1H); 7.95 (d, J= 9.1 Hz, 1H); 8.49 (s, 1H); 9.92 (s, 1H); 12.4 (br, 1H). MS
(ESI+): m/z = 533.4.

Example 132 Synthesis of (4-[7-(2-chloro-5-hydroxy-phenyl)-6-methyl-benzo[1,2,4]triazin-3-ylaminol-phenyll-(4-methyl-piperazin-l-yl)-methanone HCl salt CI O
HO ~ N' N / OW, N N e H

(CXLVI) [02601 The title compound was synthesized as shown by the reaction scheme (CXLVII).

0 N \-~ N- O
CI N=N \ BBr3/CH2C12 CI :zN
0 NON" N //--NH eHCI
N
MeO HO

(CXLVII) [02611 To a solution of a methoxylated product in McSO3H (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 (CXLVII). The mixture was stirred at RT overnight. Sat. NaHCO3 (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-d6): 6 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, 1H); 6.81 (dd, J= 8.8 Hz, J= 2.9 Hz, 1H); 7.40 (d, J= 8.9 Hz, 1H); 7.52 (d, J= 6.9 Hz, 1H); 7.76 (s, 1H); 8.07 (s, 1H); 8.09 (d, J= 6.9 Hz, 2H); 9.98 (br, 1H);
10.78 (br, 1H); 11.13 (s, 1H). MS (ESI+): m/z = 489.2.

Example 133 Synthesis of 7-(2-chloro-5-methoxy-phenyl)-5-methyl-benzof1,2,4ltriazin-3-yll-f4-(2-pyrrolidin-l- yl-ethoxy)-phenyll-amine CI

O N; N O
N N
H
(CXLVIII) [02621 The title compound was synthesized as shown by the reaction scheme (CXLIX).

Br N; N
~NH CI O-/-N CI N 2 N, Br 6 Me 32 Me0 MeO \ B(OH)2 DME/EtOH/H20 N~NH2 Dioxane/Pd2(dba)3/Xant Phos 31 Pd(Ph3P)4/Na2CO3 33 Me Cs2CO3/Reflux/4 h Reflux/18 h n -N ~/ f-D
CI
-ZZ O N
N~
-N
1) BBr3/CH2CI2 HCI
MeO N H 2) HCI CI N`N
` NCH
Me HO
Me (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 rmnol) and 7-bromo-5-methylbenzo[e][1,2,4]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-1-yl)ethoxy)phenylamino)-5-methylbenzo[e] [1,2,41triazin-7-yl)-4-chlorophenol hydrochloride CI
HO N N O\~N
N
H
(CL) [02641 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 HC1 salt and orange solid. 1H NMR (DMSO-d6):
S 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, 1H); 6.94 (d, J=
2.9 Hz, 1H); 7.09 (d, J= 9.0 Hz, 2H); 7.39 (d, J= 8.8 Hz, 1H); 7.83 (d, J= 1.6 Hz, 1H); 7.98 (d, J= 9.0 Hz, 2H); 8.12 (d, J= 1.6 Hz, 1H); 9.97 (s, 1H); 10.55 (br, 1H); 10.87 (s, 1H). MS
(ESI+): m/z = 476.3.

Example 135 Synthesis of 3-(7-(2-chloro-5-hydroxyphenyl)-5-methylbenzo[e] [1,2,4]triazin-3-ylamino)-N-(2-(pyrrolidin-1-yl)ethyl)benzamide hydrochloride CI

HO N' N
H ~N~
NN J::IY N J
H O
(CLI) [02651 The title compound was synthesized as shown by the reaction scheme (CLII).

CI CI
MeO Br CO Et MeO \

\ ~ N 34 33 N Dioxane/Pd2(dba)3/Xant Phos N N
Me N( Cs2CO3/Reflux/4 h Me H --NH2 35 CO2Et CI
UGH
THF/MeOH/H20/RT 0/RT MeO ~

36 Me Me CI
H2N Me N, 37 HO N H HCI Me 1) CDMT/NMM/CH2CI2 N~N NN..
2) BBr3/CH2CI2 H 0 2) HCI Me (CLII) [02661 Compound 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 (N1,N1-dimethylethane-1,2-diamine) (15 mg, 0.17 mmol) were used to afford the title compound (CLI) (6.3 mg, 15%) as its HCl solid and yellow solid.
1H NMR (DMSO-d6): S 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, 1H); 6.95 (d, J= 2.9 Hz, IH); 7.40 (d, J=
8.8 Hz, 1H); 7.52 (t, J= 7.9 Hz, 1H); 7.61 (d, J= 7.9 Hz, 1H); 7.87 (d, J= 1.7 Hz, 1H); 8.10 (dd, J= 8.1 Hz, J= 1.8 Hz, 1H); 8.17 (d, J= 1.8 Hz, 1H); 8.74 (s, 1H); 8.77 (t, J=
5.6 Hz, 1H);9.30 (br, 1H); 9.97 (s, 1H); 11.14 (s, 1H). MS (ESI+): fn/z = 477.1.

Example 136 Synthesis of 3-(3-(3-(dimethylamino)phenylamino)-5-methylbenzolel [1,2,4]triazin-7-yl)-4-chlorophenol hydrochloride CI

HO N;N
N N N
H

(CLIII) [0267] The title compound was synthesized as shown by the reaction scheme (CLIV).
CI
CI N Br aNMe2 Me0 \ I ~ N' N I ~
MeO I IN 38 NJ~NH2 Dioxane/Pd2(dba)3/Xant Phos N~H / NMe2 33 Me Cs2CO3/Reflux/4 h Me CI

1) BBr3/CH2CI2 HO \ I / I N' N I HCI
2) HCI N--L-H NMe2 Me (CLIV) [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-N,N-dimethylbenzenamine (compound 38) (692 mg, 3.46 rnmol), 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 (Na2S04).
The crude product was purified by chromatography (Si02/ CH2C12, then 5% MeOH
in CH2C12) and afforded compound 39 as shown by, scheme (CLIV). Compound 39 was then demethylated by using BBr3/CH2C12 to afford the title product (CLIII) (80 mg, 10%) as its HC1 salt and yellow solid. 1H NMR (DMSO-d6): 5 2.70 (s, 3H); 3.07 (s, 6H); 6.88 (dd, J= 8.7 Hz, J= 2.9 Hz, 1H); 6.94 (d, J= 2.9 Hz, 1H); 7.36 (br, 2H); 7.40 (d, J= 8.8 Hz, 1H); 7.48 (br, 1H); 7.86 (s, 1H); 8.10 (br, 1H); 8.16 (d, J= 1.7 Hz, 1H);
9.98 (br, 1H); 11.02 (s, 1H). MS (ESI+): in/z = 406.1.

Example 137 Synthesis of 3-(3-(6-methoxypyridin-3-ylamino)-5-methylbenzo[el [1,2,4]triazin-7-yD-4-chlorophenol hydrochloride CI

N ~
Zz~
N N
H
(CLV) [0269] The title compound was synthesized as shown by the reaction scheme (CLVI).
N- OMe CI

CI N Br I NMeO NH2 Phos 33 Me Cs2CO3/Reflux/4 h Me 41 CI
N N OMe 1) BBr31CH2CI2 HO
2) HCI N )-N, HCI
Me (CLVI) [0270] The title compound was prepared by a method that was analogous to that used to make compound (CLIII), as described in Example 136, except using compounds (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 HC1 salt in a form of a red solid. 1H NMR (DMSO-d6): 8 64 (s, 3H); 3.87 (s, 3H); 6.88 (dd, J= 8.7 Hz, J= 2.9 Hz, 1H); 6.92 (d, J= 8.9 Hz, 1H); 6.95 (d, J= 2.9 Hz, 1H); 7.39 (d, J= 8.8 Hz, 1H); 7.84 (dd, J= 1.9 Hz, J= 1.0 Hz, 1H); 8.14 (d, J= 1.7 Hz, 1H); 8.24 (dd, J= 8.9 Hz, J= 2.9 Hz, 1H); 8.88 (d, J= 2.6 Hz, 1H); 10.00 (br, 1H); 10.92 (s, 1H).
MS (ESI+): m/z = 394Ø

Example 138 Synthesis of 3-(3-(2-(pyrrolidin-1-yl)ethylamino)-5-methylbenzo[e] [1,2,4]triazin-7-vll-4-chlorophenol hydrochloride CI

HO NN
NN
H

(CLVII) [02711 The title compound was synthesized'as shown by the reaction scheme (CLVIII).

CI NO CI

N'N 1) DMF/K2CO3/80 C/72 h HO N HC!
Me0 N
N~NHZ 2) BBr31CH2CI2 3) HCI N H/~

Me Me (CLVIII) [02721 As shown by scheme (CLVIII), to a solution of compound 33 (320 mg, 1.06 mmol) in anhydrous DMF (20 mL) were added K2C03 (1.2 g, 8.51 mmol), and 1-(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 CH2C12 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/CH2C12. The product was purified by preparative HPLC. The title compound (CLVII) (90 mg, 21%) was obtained as HCl salt in a form of a yellow solid. 1H NMR (DMSO-d6): 8 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, 1H); 6.94 (d, J= 2.9 Hz, 1H); 7.37 (d, J= 8.6 Hz, 1H); 7.76 (dd, J= 1.8 Hz, J=
1.0 Hz, 1H); 8.05 (d, J= 1.8 Hz, 1H); 10.05 (br, 1H); 10.78 (br, 1H). MS (ESI+): m/z =
384.1.

Example 139 Synthesis of 3-(5-(2-(pyrrolidin-1-yl)ethoxy)-3-(pyridin-3-ylamino)benzo [e] 11,2,41 triazin-7-yl)-4-chlorophenol dihydrochloride CI

HO N- N N.
N N
CN O H
(CLIX) [0273] The title compound was synthesized as shown by the reaction scheme (CLX).

N02 Br2/Dioxane Br / 1) NH2CN/HCI/110 C Br N' NI

OH OH 2) 30% NaOH/110 C OH

/ CI

Raney Ni/H2 Br N MeO I B(OH)2 CI
N'5~NH2 31 N' N
OH DME/EtOH/H20 Me0 ' ~NH
47 Pd(Ph3P)4/Na2CO3 48 N z Reflux/18 h OH

CI N
CIl"~43 MeO N' N Br 50 Cs2CO3/Me2CO/Reflux 49 N~NH2 Dioxane/Pd2(dba)3/Xant Phos Cs2CO3/Reflux/4 h O',,--\N

CI CI
N N\ N. N
MeO I N / 1) BBr31CH2CI2 HO / I I ~) /) 51 H 2) HCI N H 2HCI
~\ Nv O~\ NV

(CLX) [0274] As shown by scheme (CLX), to a solution of 2-ainino-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-d6): S 6.28 (br, 2H); 6.99 (d, J= 2.2 Hz, 1H); 7.57 (d, J= 2.1 Hz, 1H). MS (ESI+): m/z = 234.
Compound 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 minol) and 31 (740 mg, 4.0 mmol) were used to afford compound 48 (108 mg, 10%) as yellow solid. 1H NMR (DMSO-d6): 8 7.02 (dd, J=
8.9 Hz, J= 3.1 Hz, 1H); 7.07 (d, J= 3.1 Hz, 1H); 7.16 (d, J= 1.9 Hz, 1H); 7.49 (d, J= 8.9 Hz, 1H); 7.60 (br, 2H); 7.69 (d, J= 1.9 Hz, 111). MS (ESI+): fn/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), 1-(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 CH2C12 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/CH2C12 to afford the title compound (CLIX) (60 mg, 56%) as its HCl salt in a form of a yellow solid. 1H NMR (DMSO-d6): 8 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, 1H); 7.00 (d, J= 2.9 Hz, 1H); 7.43 (d, J= 8.8 Hz, 2H); 7.58 (d, J=
1.5 Hz, 1H); 8.01 (br, 1H); 8.06 (d, J= 1.5 Hz, 1H); 8.62 (d, J= 4.9 Hz, 1H);
8.69 (d,,J=
8.8 Hz, 1H); 9.77 (s, 1H); 10.06 (br, 1H); 10.79 (br, 1H); 11.90 (s, 1H). MS
(ESI+): fn/z = 463.1.

Example 140 Synthesis of 7-(1H-indol-4-yl)-5-methyl-benzo[1,2,4]triazin-3-vl-amine N=N
/~-NH2 ~ / N

HN /

(CLXI) [0278] The title compound was synthesized as shown by the reaction scheme (CLXII).
Br/ N,,N
N=N
Br 1) NaH B(OH)2 N~NH2 />-NH2 \ 2) B(OMe)3 I \ Me 32 / \ N
\ I N 3) Hai'0 H DME/EtOH/H20 HN /
H
Pd(Ph3P)4/Na2CO3 52 53 Reflux/1 8 h it, N=N H - 00 n ~N \ / NNHCI
Br __\ / S,NN / \ / 55 -Dioxane/Pd2(dba)3/Xant Phos HN /
Cs2CO3/Reflux/4 h N=N N 3;0 NIO
/ \ N~ N CI
HN

(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-H20, 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 compound (CLXI) (220 mg, 17%) as a yellow solid. 1H NMR (DMSO-d6): 8 2.59 (s, 3H); 6.64 (t, J
= 1.1 Hz, 1H); 7.22-7.25 (m, 2H); 7.46-7.48 (m, 2H); 7.64 (br, 2H); 8.02 (d, J= 1.9 Hz, 1H); 8.24 (d, J= 1.9 Hz, 1H); 11.35 (s, 1H). MS (ESI+): m/z = 276.2.

Example 141 Synthesis of 4-(7-(1H-indol-4-yl)-5-methylbenzo[e][1,2,4]triazin-3-ylamino)-N-(2-(pyrrolidin-1-yl)ethyl) benzenesulfonamide hydrochloride O
~=N,~N / N
HN
NON H
H

(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 HC1 salt and orange solid. 1H NMR
(DMSO-d6): S 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, 1H); 7.25-7.31 (m, 2H); 7.51-7.52 (m, 2H); 7.88 (d, J= 9.0 Hz, 2H); 8.23 (s, 1H); 8.27 (d, J= 9.0 Hz, 1H); 8.43 (d, J= 1.8 Hz, 1H); 10.04 (br, 1H); 11.40 (s, 1H); 11.42 (s, 1H). MS
(ESI+): m/z = 528.4.

Example 142 Synthesis of 4-(7-(2-bromo-5-hydroxyphenyl)-5-methylbenzo[el [1,2,4]triazin-3-ylamino)-N-(2-(pyrrolidin-l-yl)ethyl) benzenesulfonamide hydrochloride Br HO N N HN

N H N~]
(CLXN) [02821 The title compound was synthesized as shown by the reaction scheme (CLXV).

Br / I N:N
N),NH2 Br Br Me 32 N, N
MeO
MeO B(OH)2 DME/EtOH/H20 NNH
Pd(Ph3P)4/Na2CO3 2 56 Reflux/18 h 57 Me O
Br/ S1;N ,~N Br O O N
H 54 N, =S,N
MeO N I H
Dioxane/Pd2(dba)3/Xant Phos N'N
Cs2CO3/Reflux/4 h H
Me 58 Br 1) BBr3/CH2CI2 HO N; N / S-HN
2) HCI N~N HCI
Me H

(CLXV) [0283] The intermediate compounds 57 and 58 shown on scheme (CLXV) were prepared as follows. Compound 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][1,2,4]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 (CXLVI), as described in Example 132, except compound 58 (0.90 mmol) was used to afford the title compound (CLXIV) (51 mg, 20% in two steps) as HCI salt and orange solid. 1H NMR (DMSO-d6): 8 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, 1H); 6.97 (d, J= 2.9 Hz, 1H);
7.55 (d, J=
8.8 Hz, 1H); 7.88 (d, J= 8.8 Hz, 2H); 7.87 (d, J= 1.8 Hz, 1H); 8.04 (t, J= 6.0 Hz, 1H);
8.17 (d, J= 1.8 Hz, 1H); 8.25 (d, J= 8.8 Hz, 2H); 10.12 (s, 1H); 10.85 (br, 1H); 11.44 (s, 1H). MS (ESI+): m/z = 583Ø

Example 143 Synthesis of 4-(7-(2-chloro-6-hydroxyphenyl)-5-methylbenzof el [1,2,4ltriazin-3-ylamino)-N-(2-(pyrrolidin-l-yl)ethyl)benzensulfonamide hydrochloride CI
N ~S`O
N , OH I / ~ ~ ~ HN--\-N
N N

(CLXVI) [0285] The title compound was synthesized as shown by the reaction scheme (CLXVII).

VB(0 CI
N , H)2 N
Br N OMe 59 \ / I 'N
N-,J-NHz DME/EtOH/HZO OMe N)-NH2 32 Pd(Ph3P)4/Na2CO3/Reflux/18 h 60 90%
R, 11P
CI O O
SN'~N Q1NN
/ H
Br0 54 H
Dioxane/Pd2(dba)3/Rant Phos OMe \ I N~N
Cs2CO3/Reflux/4 h H 61 L95%

CI
OSO^/
1) BBr3/CH2Cl2 POOH N; \ N N
N H HCI
2) HCI XN' N
H
(CLXVII) [0286] As shown by scheme (CLXVII), 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][1,2,4]triazin-3-amine 32 (1.2 g, 6.28 mmol) were used to give compound 60 (7-(2-chloro-6-methoxyphenyl)-5-methylbenzo[e][1,2,4]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)-N-(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-hydroxyphenyl)-5-methylbenzo[e] [ 1,2,4]triazin-3-ylamino)-N-(2-(pyrrolidin-l -yl)ethyl)benzensulfonamide (CLXVI) (529 mg, 75%) as HCl salt and yellow solid.

NMR (DMSO-d6): 8 1.83-1.87 (m, 2H); 1.95-2.01 (in, 2H); 2.70 (s, 3H); 2.97-3.04 (m, 2H); 3.09-3.13 (m, 2H); 3.21-3.25 (in, 2H); 3.51-3.56 (m, 2H); 7.01 (d, J= 8.3 Hz 1H);
7.06 (d, J= 7.9 Hz, I H); 7.27 (t, J= 8.0 Hz, 1H); 7.77 (s, 1H); 7.88 (d, J=
8.9 Hz, 2H);
7.92 (t, J= 6.0 Hz, 1H); 8.09 (s, 1H); 8.26 (d, J= 8.9 Hz, 2H); 10.09 (s, 1H);
10.22 (br, 1H); 11.40 (s, 1H). MS (ESI+): ma/z = 540.5.

Example 144 Synthesis of 3-{6-methyl-3-14-(2-pyrrolidin-1-yl-ethylsulfamoyl)-phenylamin of -b enzo [1,2,4]triazin-7-yl}-b enzamide Q
H2N N,N S,NN
O I / \ N N O H
H
(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, 1.01nmol) were suspended in a mixture of DME/ethanol (4:1, 1nL). 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 Na2SO4a and filtered. The filtrate was concentrated to afford the free base compound, which was converted to HCl salt according to method C and furnished the title compound as a yellow solid (40 mg, 32% overall). 1H NMR
(DMSO-d6): 6 1.78-1.90 (m, 2H), 1.90-2.03 (m, 2H), 2.45 (s, 3H), 2.95-3.10 (in, 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, 1H), 7.61 (t, J= 7.8 Hz, 1H), 7.68 (d, J= 7.8 Hz, 1H), 7.83 (s, 1H), 7.86 (d, J= 8.9 Hz, 1H), 7.95-8.05 (m, 3H), 8.12 (s, 1H), 8.20-8.24 (m, 3H), 10.55 (br s, 1H), 11.33 (s, 1H). MS
(ESI+): fn/z 533.

Example 145 Synthesis of 2-(4-{6-[7-(2,6-dichloro-phenyl)-5-methyl-b enzo [1,2,4]triazin-3-ylaminol -2-methyl-pyrimidin-4-yll-piperazin-l-yl)-ethanol CI

N, 'N N N
CI I

NN ON H "-'~OH

(CLXIX) [0289] To synthesize the title compound (CLXIX), two intermediate compounds 62 (2-[4-(6-chloro-2-methyl-pyrimidin-4-yl)-piperazin-1-yl]-ethanol) and 63 (7-(2,6-dichloro-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamine) shown below were used.

Ni N
CI \ N
N'--'OH

CI
\ I / N.N
CI

[0290] To synthesize compound 62, to a solution of 4,6-dichloro-2-methyl-pyrimidine (5.0 g, 31 mmol) and 2-piperazin-1-yl-ethanol (2.7 g, 21 mmol) in dioxane (25 mL) was added DIPEA (3.0 mL, 17 inmol). 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% McOH/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 HCl salt according to method C and furnished the title compound as a yellow solid (0.18 g, 65% overall). 1 H NMR (DMSO-d6): S 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, 1H), 7.55 (t, J
= 7.8 Hz, 1H), 7.67 (d, J= 8.2 Hz, 2H), 7.91-7.93 (m, 1H), 8.31 (d, J= 1.5 Hz, 1H), 10.93 (br s, 1H), 11.83 (br s, 1H). MS (ESI+): m/z 525.

Example 146 Synthesis of 4-chloro-3-(3-{6-[4-(2-hydroxy-ethyl)-piperazin-1-v11-methyl-pyrimidin-4-ylamin o}-5-methyl-b enzo [1,2,4]triazin-7-yl)-phenol CI
HO N'.N N N

NN ON H ~--'OH

(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]-methyl-pyrimidin-4-yl} -piperazin- 1 -yl)-ethanol) shown below was used.

CI

N
Me0 \ \ I N'N

N H ON
'-~OH

[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 compound (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 HCl salt according to method C as a yellow solid (25 mg, 60% overall). 1H NMR (DMSO-d6): S 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, 1H), 6.91 (dd, J= 8.8 Hz, J= 2.9 Hz, 1H), 6.98 (d, J= 2.9 Hz, 1H), 7.42 (d, J= 8.8 Hz, 1H), 7.75 (s, 1H), 8.03 (dd, J=
1.8 Hz, J=
0.9 Hz, 1H), 8.30 (d, J= 1.8 Hz, 1H), 10.05 (br s, 1H), 10.78 (br s, 1H), 11.73 (br s, 1H).
MS (ESI+): m/z 507.

Example 147 Synthesis of 4-[7-(2-chloro-5-hydroxy-phenyl)-5-methyl-b enzo [1,2,4]triazin-3-ylaminol-N-ethyl-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide CI O
N'N / S.Ni~N
HO

N N
H
(CLXXI) [0295] To synthesize the title compound (CLXXI), intermediate compounds 65 (4-bromo-N-ethyl-N-(2-pyrrolidin-1-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- l -yl-ethyl)-benzenesulfonamide) shown below were used.

O
Ii ~~ O IN ~i N
i\%
Br CI O
MeO N' N S,NN

N N
H

[0296] To synthesize compound 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 mL) 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 Na2SO4 and filtered. The filtrate was concentrated and the residue purified by flash chromatography on silica gel (8% McOH/DCM) to afford compound 65 as a white solid (0.80 g, 74%). 1H NMR (DMSO-d6): 8 1.03 (t, J=
7.1 Hz, 3H), 1.55-1.75 (in, 4H), 2.35--2.75 (m, 4H), 3.15-3.25 (in, 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 mrnol), compound 65 (0.15 g, 0.42 mrnol), 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 farm 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 HCl salt according to method C to yield a yellow solid (0.13 g, 77%
overall). 1H NMR (DMSO-d6): 8 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 (in, 2H), 6.89 (dd, J= 8.7 Hz, J= 2.8 Hz, 1H), 6.95 (d, J= 2.9 Hz, 1H), 7.42 (d, J= 8.8 Hz, 1H), 7.90 (d, J= 8.8 Hz, 2H), 7.93 (dd, J=1.8 Hz, J= 0.9 Hz, 1H), 8.23 (d, J= 1.8 Hz, 1H), 8.28 (d, J= 8.8 Hz, 2H), 9.97 (s, 1H), 10.01 (br s, 1H), 11.48 (s, 1H). MS (ESI+): m/z 567.

Example 148 Synthesis of 4-f7-(2-chloro-5-hydroxy-phenyl)-5-methyl-benzo f 1,2,41 triazin-3-ylaminol-N-methyl-N-(2-morpholin-4-yl-ethyl)-benzenesulfonamide CI 0 r 'O
HO \ I / N'N 1'NN

N N
H
(CLXXII) [02991 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[ 1,2,4]triazin-3-ylamino]-N-methyl-N-(2-morpholin-4-yl-ethyl)-benzenesulfonamide) shown below were used.

O
n \ I ~ NH
Br O rO
/ S, i, N J
\ I O
Br CI 0 (0 n J
MeO N 1 N~~N
N N
H

[0300] To synthesize compound 67, to a solution of 4-bromosulfonyl chloride (5.0 g, 20 mmol) in DCM (30 mL) was added methylarnine (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): 8 2.41 (d, J= 5.0 Hz, 3H), 7.56 (q, J= 4.9 Hz, 1H), 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), (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 Et20/hexane (1/1) to afford compound 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 nunol), 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): 8 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, 1H), 7.16 (d, J= 3.1 Hz, 1H), 7.54 (d, J= 8.9 Hz, 1H), 7.84 (d, J= 8.9 Hz, 2H), 7.92 (s, 1H), 8.26 (d, J= 8.9 Hz, 2H), 8.28 (d, J
=1.8 Hz, 1H), 11.45 (s, 1H). 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 HCl salt according to method C as a brown solid (0.10 g, 57%
overall). 1H
NMR (DMSO-d6): 5 3.10-3.20 (in, 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, 1H), 6.96 (d, J= 2.9 Hz, 1H), 7.41 (d, J= 8.8 Hz, 1H), 7.88 (d, J= 8.9 Hz, 2H), 7.91-7.93 (m, 1H), 8.22 (d, J= 1.8 Hz, 1H), 8.31 (d, J= 8.9 Hz, 2H), 10.00 (s, 1H), 10.56 (br s, 1H), 11.50 (s, 1H). MS (ESI+): na/z 569 (M+H)+.

Example 149 Synthesis of 4-chloro-3-[3-(4-{[(2-diethylamino-ethyl)-methyl-aminol-methyl}-phenylamino)-5-methyl-benzo [1,2,41 triazin-7-YI] -phenol HO \ \ I N,N \ NN
N
H
(CLXXIII) [0304] To synthesize the title compound (CLXXIII), intermediate compounds 70 (N-(4-bromo-benzyl)-N',N'-diethyl-N-methyl-ethane-1,2-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-1,2-diamine) shown below were used.

\ I N N
Br cl MeO \ \ I N N N
N N \
H

[0305] To synthesize compound 70, a solution ofN,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 Na2SO4, 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):

6 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, 1H), 7.14 (d, J= 3.2 Hz, 1H), 7.32 (d, J=
8.5 Hz, 1H), 7.53 (d, J= 8.9 Hz, 2H), 7.87-7.89 (m, 1H), 7.98 (d, J= 8.6 Hz, 2H), 8.21 (d, J= 1.8 Hz, 1H), 10.95 (s, 1H). 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 HCl salt according to method C and furnished the title compound as an orange solid (40 mg, 19% overall). 1H NMR (DMSO-d6): 8 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, 1H), 4.51 (br d, J=11.3 Hz, 1H), 6.89 (dd, J= 8.7 Hz, J= 2.9 Hz, 1H), 6.95 (d, J= 2.9 Hz, 1H), 7.41 (d, J=
8.7 Hz, 1H), 7.67 (d, J= 8.0 Hz, 2H), 7.89 (s, 1H), 8.13 (d, J= 8.4 Hz, 2H), 8.18 (d, J= 1.7 Hz, 1H), 10.00 (s, 1H), 10.81 (br s, 1H), 11.19 (s, 1H), 11.23 (br s, 1H). MS
(ESI+): m/z 505.

Example 150 Synthesis of 4-[7-(2-fluoro-3-hydroxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylaminol-N (2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide OH

,N
N S N

N N
H
(CLXXIV) [0308] To synthesize the title compound (CLXXIV), intermediate compounds 72 (7-(2-fluoro-3-methoxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine) and 73 (4-[7-(2-fluoro-3-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide) shown below were used.

OMe F

N,N
N:~ NH2 OMe F
O
~x>o1rN
H

[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 minol), 2 M Na2C03 (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% McOH/DCM) to afford compound 73 as a yellow solid (80 mg, 43%). 1H
NMR (DMSO-d6): S 1.55-1.65 (m, 4H), 2.30-2.40 (in, 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, 1H), 7.84 (d, J= 8.9 Hz, 2H), 8.04 (s, 1H), 8.21 (d, J= 8.9 Hz, 2H), 8.35 (s, 1H), 11.40 (s, 1H). 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 HCl salt according to method C as an orange solid (48 mg, 66%
overall). 1H NMR (DMSO-d6): S 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, 1H), 8.05 (s, 1H), 8.25 (d, J= 9.0 Hz, 2H), 8.34 (s, 1H), 10.05 (br s, 1H), 10.10 (s, 1H), 11.45 (s, 1H). MS
(ESI+): m/z 523.

Example 151 Synthesis of 4-[7-(2-fluoro-6-hydroxy-phenyl)-5-methyl-b enzo [1,2,4]triazin-3-ylaminol -N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide F
O
N N N
\ I \
OH I / \ I H
N N
H
(CLXXV) [0312] To synthesize the title compound (CLXXV), intermediate compounds 74 (7-(2-fluoro-6-methoxy-phenyl)-5-methyl-benzo[ 1,2,4]triazin-3-ylamine) and 75 (4-[7-(2-fluoro-6-methoxy-phenyl)-5-methyl-benzo [ 1,2,4]triazin-3 -ylamino] -N-(2-pyrrolidin- l -yl-ethyl)-benzenesulfonamide) shown below were used.

F
\ I \ N,N
OMe i N'NH2 F

N
N, / oS N - ' I H
OMe N N\
H

[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 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 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): S 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, 1H), 7.06 (d, J= 8.5 Hz, 1H), 7.47 (dd, J=15.3 Hz, J= 8.4 Hz, 1H), 7.83 (s, 1H), 7.84 (d, J= 9.0 Hz, 2H), 8.18 (s, 1H), 8.22 (d, J= 8.9 Hz, 2H), 11.37 (s, 1H).
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 HCl salt according to method C as an orange solid (0.10 g, 69%
overall). 1H NMR (DMSO-d6): 8 1.80-1.90 (m, 2H), 1.95-2.03 (in, 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, 111), 6.89 (d, J= 8.3 Hz, 2H), 7.28 (dd, J= 15.2 Hz, J=
8.3 Hz, 1H), 7.88 (dd, J= 8.8 Hz, 2H), 7.89 (s, 1H), 7.90 (t, J= 6.1 Hz, 1H), 8.21 (s, 1H), 8.26 (d, J=
9.0 Hz, 2H), 10.04 (br s, 1H), 10.20 (s, 1H), 11.40 (s, 1H). MS (ESI+): m/z 523.

Example 152 Synthesis of benzoic acid 4-chloro-3-{5-methyl-3-[4-(2-pyrrolidin-l-yl-ethoxy)-phenylaminol-benzo[1,2,4]triazin-7-yl}-phenyl ester \ O \ \ N'~N / O~~N
N N
H
(CLXXVI) [0316] The title compound was synthesizrd and had the following characteristics. 1H
NMR (DMSO-d6): 6 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, 1H), 7.60-7.65 (m, 3H), 7.75 (d, J= 8.6 Hz, 1H), 7.76 (t, J= 7.4 Hz, 1H), 7.90 (ad, J
= 1.8 Hz, J= 1.0 Hz, 1H), 7.93 (d, J= 9.1 Hz, 2H), 8.16 (d, J= 8.4 Hz, 2H), 8.22 (d, J=
1.8 Hz, 1H), 10.84 (br s, 1H). 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 (CLXXVII).

R1 Br N N
DME/EtOH (4:1) +
Rq ~ N 1), NH2 Pd(PPhs)q R3 N; N
3 ~=
R 2 B(OH)2 RS 2M Na2CO3, 100 C RZR4 N NH2 Rs RI
R6 Pd2(dba)3, Xantphos, 6 / R
+ Cs2CO3, dioxane R3 N" N
Br Ar, 100 C RR4 N N

(CLXXVII) Example 153 Synthesis of N-(4-(2-(pyrrolidin-1-yl)ethoxy)phenyl)-6-methyl-7-(4-methylpyridin-3-yl)benzolel [1,2,4ltriazin-3-amine /
N N; N
N N
H
(CLXXVIII) [0318] The synthesis of the title compound (CLXXVIII) can be generally described by the reaction scheme (CLXXIX).

~ ~\N1 ~ -----= \ aN ' N / N
I \ N' N + / ~ O
Br v V

76 77 Title Compound (CLXXLX) [03191 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), 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/CHC13) to afford the title compound as a red-orange solid (128 mg, 49%). Rf = 0.24 (1:9 McOHICHC13); 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 (in, 4H), 4.31 (t, J= 5.0 Hz, 2H), 7.07 (d, J= 9.1 Hz, 2H), 7.45 (d, J= 5.0 Hz, 1H), 7.69 (s, 1H), 7.91 (d, J= 9.0 Hz, 2H), 8.07 (s, 1H), 8.41 (s, 1H), 8.54 (d, J= 5.1 Hz, 1H), 9.72 (br s, 1H), 10.75 (s, 1H).

Example 154 Synthesis of 5-(5-methyl-7-(2,6-dimethylphenyl)benzofelf1,2,41triazin-3-ylamino)-1-(3-(pyrrolidin-1-yl)propy1)uyridin-2(1H)-one IuIIIIIiii.N,N O
NN ~N, N
H

(CLXXX) [0320] The synthesis of the title compound (CLXXX) can be generally described by the reaction scheme (CLXXXI).

CI 50% NaOH O 0 + HO~~Br Adogen 464 CH3CN
OzN N 02N N O 02N C N~/\iBr toluene, 60 C Br + HN3 CH3CN
02N \ N~iN MeOH H2N N,,,, N'D

N
O

/ I '~ N IN \ (1.5 eq), CuBr (2 eq) I / N,,N

N NH2 CH3CN N Br reverse addition / 0 THE/dioxane/DMF/H20 \ I , NN / O
HzN N,iN A N N\ f:
H

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, 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: 8 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, 1H), 8.36 (dd, J= 9.5, 3.0 Hz, 1H), 9.07 (d, J=
2.6 Hz, 1H).
[0322] Next, compound 81 (1-(3-bromopropyl)-5-nitropyridin-2(1H)-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);

CDC13: 8 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 Hz, 1H), 8.11 (dd, J= 10.1, 3.1 Hz, 1H), 8.67 (d, J= 3.0 Hz, 1H).
[0323] Next, compound 83 (5-nitro-1-(3-(pyrrolidin-l-yl)propyl)pyridin-2(1H)-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 NH4OH/MeOH/CH3Cl) to afford compound 83 as a pale-yellow oil (2.3 g, 95%). LC retention time: 1.09 min. MS (ESI+) in/z= 252.1 1H
NMR
CDC13 :8 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, 1H), 8.03 (dd, J= 10.1, 3.1 Hz, 1H), 8.84 (d, J= 3.0 Hz, 1H); 13C NMR CDC13:5 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 MCOH (65 mL) was added palladium acetate (178.4 mg, 0.8 mmol), and the suspension was bubbled with H2 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-1-yl)propyl)pyridin-2(1H)-one) as a black oil (1.8 g, quant.). LC
retention time = 0.5 min MS (ESI+) m/z = 222.21H NMR CDC13:6 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, 1H) 6.91 (d, J= 3.0 Hz, 1H), 7.04 (dd, J=
9.5, 3.0 Hz, 1H).

[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 (801nL) and extracted with ether (2x80 mL). The combined organic layers were washed with 20% HC1(2x40 mL), dried (MgSO4), filtered, and concentrated in vacuo to afford compound 86 (3-bromo-5-methyl-7-(2,6-dimethylphenyl)benzo[e][1,2,4]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 THE (2 mL) was added a solution of 0.1 M (1:1:2:6 THE/dioxane/DMF/water) compound 84 (5.0 mL, 0.5 mmol) obtained as described above. After 22 h. under reflux with only minimal forward reaction, TEA (140 L, 1.0 mmol) was added and continued reflux for 42 h. The reaction was diluted with water (10 mL) and extracted with EtOAc (3x 15 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 = 469.41H NMR DMSO-d6: S 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, 1H), 7.15-7.28 (m, 3H), 7.63 (s, 1H), 7.79 (dd, J= 9.8, 2.9 Hz, 1H), 7.94 (d, J= 1.6 Hz, 1H), 8.62 (br s, 1H), 9.51 (br s, 1H), 10.71 (br s, 1H).

Example 155 Synthesis of 2-(7-(2-chloro-5-hydroxyphenyl)-5-methylbenzo[e] [1,2,4]triazin-3-ylamino)-N-(2-(Pyrrolidin-1-yl)ethyl)thiazole-carboxamide NH
HO N, N N \ N
N;~H~S

(CLXXXII) [03271 The synthesis of the title compound (CLXXXII) can be generally described by the reaction scheme (CLXXXIII).

CI CI
NH -NH
O \ I\ N N
N + N\ N O \ I I N N N
N~NH2 BrIS / NNS 0 H

CI o BBr3 NH
D CM HO \ \ N, N INII N

Title Compound (CLXXXIII) [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/CHC13); 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 1M 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 (CLXXXI) 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: b 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, 1H), 6.95 (d, J= 2.9 Hz, 1H), 7.41 (d, J= 8.7 Hz, 1H), 7.83 (s, 1H), 7.86 (t, J=
5.8 Hz, 1H), 7.94 (s, 1H), 8.23 (s, 1H), 9.94 (br s, 1H). Calcd for C24H24C1N702S=2.5H20Ø1Br: 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-methylbenzo[e] [1,2,4]triazin-7-yl)-4-fluorophenol O N' N / ON
4)s H
N N
H
(CLXXXIV) [03301 The synthesis of the title compound (CLXXXIV) can be generally described by the reaction scheme (CLXXXV).

F
O N;N + OWN p N'N 0-N
-1j, 'ja NNHZ Br \ I I/ N
N
H

F
HO N, N ON
N'J'N
H

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-l-yl)ethyl)thiazole-4-carboxamide) (254 mg, 0.94 mmol), 77 (described above) (177 L, 0.85 mmol), Cs2CO3 (1.1 g, 3.42 mmol), Xantphos (102 mg, 0.18 mmol), Pd2(dba)3 (78 mg, 0.09 rnmol), and 17 mL dioxane for 18 h, to obtain compound 89 (N-(4-(2-(pyrrolidin-1-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/CHC13 to afford the title compound as a red solid (181 mg, 45%). Rf 0.06 (1:20:179 % NH4OH/MeOH/CHC13); 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 1M 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 compound (CLXXXV) as a red solid (30 mg, 14%). LC retention time:
2.26 min, MS (ESI+) m/z = 460.2 1H NMR DMSO-d6: 8 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, 1H), 6.86 (dt, J= 8.9, 3.6 Hz, 1H), 7.07 (d, J= 9.1 Hz, 2H), 7.16 (t, J= 9.2 Hz, 1H), 7.64 (s, 1H), '7.91 (d, J= 9.1 Hz, 2H), 8.09 (s, 1H), 9.63 (s, 1H), 9.71 (br s, 1H), 10.75 (s, 1H).

Example 157 Synthesis of 4-f7-(2-fluoro-5-hydroxy-phenyl)-6-methyl-benzo f 1,2,44 triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide F
HO N HSO, N
N N

H C) (CLXXXVI) [0333] The synthesis of the title compound (CLXXXVI) can be generally described by the reaction scheme (CLXXXVII).

F
\ Br ~ N, / ~ N~-N3 O / B.OH + N
/ N~N \
OH H

F / F

0 'N HO \ I \ N / I S
~HN~ HN
NN \ --' / NN \
H r-N H 0 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/CHC13), 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 1M BBr3 in DCM (1 mL, 1.0 mmol), in 20 mL DCM for 90 min. The reaction was quenched, filtered, and purified using BPLC 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 'H NMR DMSO-d6: 8 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, 1H), 6.88 (dt, J= 9.2 Hz, 1H), 7.79 (s, 1H), 7.84 (d, J= 8.9 Hz, 2H), 8.86 (t, J= 6.2 Hz, 1H), 8.19 (s, 1H), 8.22 (d, J= 8.9 Hz, 2H), 9.52 (br s, 1H), 9.66 (s, 1H).

Example 158 Synthesis of 4-[6-methyl-7-(1H-pyrazol-4-yl)-benzo[1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide HN
N~ \ N N / S
~HN
NN ~

H O
(CLXXXVIII) [0336] The synthesis of the title compound (CLXXXVIII) can be generally described by the reaction scheme (CLXXXIX).

HN Br N S'o " Z N ,S.,O
N ~ \ N / ~~N HN~ xcNcx O H N H N
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, 3 1%). LC retention time: 1.94 min, MS (ESI+) m/z = 479.21H NMR DMSO-d6: S
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, 1H), 7.84 (d; J= 7.1 Hz, 2H), 7.86 (t, J= 6.2 Hz, 1H), 8.11 (br s, 111), 8.20 (d, J= 7.1 Hz, 2H), 8.35 (s, 1H), 9.57 (br s, 1H), 11.24 (s, 1H).

Example 159 Synthesis of 4-[7-(2-chloro-5-hydroxy-phenyl)-6-methyl-benzo [1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide CI

N OS
HO N HN--~
NN a H- N
(CXC) [0338] The synthesis of the title compound (CXC) can be generally described by the reaction scheme (CXCI).

ci CI Br N' N OS O N OS'00 + /HN O N / HN Ili, O B(OH)2 N H\ I I N~H

CI
HO I N' N I HN
NN
H N
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 rnmol), 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-l -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 1M 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 = 539.1/541.41H NMR DMSO-d6: S 1.83-1.89 (in, 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, 1H), 6.91 (dd, J= 8.8, 2.9 Hz, 1H), 7.41 (d, J= 8.8Hz, 1H), 7.80 (s, 1H), 7.85 (d, J= 8.9 Hz, 2H), 7.92 (t, J= 4.5 Hz, 1H), 8.11 (s, 1H), 8.21 (d, J= 8.9 Hz, 2H), 9.98 (s, 2H), 11.33 (s, 1H).

Example 160 Synthesis of 4-[7-(3-hydroxymethyl-phenyl)-6-methyl-benzo [1,2,4] triazin-3-ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide S~'O
HOB M Na aN I (CXCII) [0341] The synthesis of the title compound (CXCII) can be generally described by the reaction schemes (CXCIII) and (CXCIV).

DME / EtOH / H2O
Br N I Na2CO3 / N + H Pd (PPh3)4, reflux (CXCIII) [0342] The mixture of 7-bromo-6-methylbenzo[e][1,2,4]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 minol) and Na2CO3 (1.1 g, 10 mmol) was degassedwith 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/CHC13 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).

HO

Pd2 (dba)3 Xantphos N"N - 0 H Cs2CO3 + Br S_1 Dioxane, reflux (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-1-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 1.6 cm) chromatography using 30% CH3OH in CHC13 as an eluent. The solid was washed with acetone to afford the title compound (CXCII), as a bright yellow solid (200 mg, 52%). 1H
NMR DMSO-d6: 8 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, 1H), 7.43 (m, 5H), 7.80 (m, 3H), 8.12 (s, 1H), 8.18 (d, J= 8.7 Hz, 2H), 11.25 (s, 1H). MS (ESI+) ml- 7= 519.

Example 161 Synthesis of 4,-[7-(5-chloro-thiophen-2-vl)-6-methyl-benzo [1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-vl-ethyl)-benzenesulfonamide CI

S
N~ IN / S
HN-\-N~]
N H

(CXCV) [0345] The synthesis of the title compound (CXCV) can be generally described by the reaction schemes (CXCVI) and (CXCVII).

OH DME / EtOH / H2O
Na2CO3 Br N, N + I \\ B=OH Pd (PPh3)4, reflux )C(N NH2 CI

(CXCVI) [0346] The mixture of 7-bromo-6-methylbenzo[e][1,2,4]triazin-3-amine (310 mg, 1.3 minol), 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/CHC13 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 (CXCVII).

Pd2 (dba)3 Xantphos N a H Cs2CO3 CI S I N + Br S-NN Dioxane, reflux (CXCVII) [0348] A mixture of 7-(5-chlorothiophen-2-yl)-6-methylbenzo[e][1,2,4]triazin-3-amine (110 mg, 0.4 mmol), 4-bromo-N-(2-pyrrolidin-1-yl-ethyl)benzenesulfonamide (260 mg, 0.78 mmol), cesium carbonate (390 mg, 1.2 mmol), Xantphos(46 mg, 0.08 minol), 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: 5 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, 1H), 7.31 (d, J= 3.9 Hz, 1H), 7.45 (br s, 1H), 7.81 (m, 3H), 8.16 (d, J= 9.3 Hz, 2), 8.32 (s, 1H), 11.33 (s, 1H). MS (ESI+) fn/z = 529.

Example 162 Synthesis of 4-(6-methoxy-7-o-tolyl-benzo[1,2,4]triazin-3-ylamino)-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide N
\ ,N / HN
JI
N
C]
p NN
H
(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).

Na (2.6 eq.) N'0 CH30Na / CH3OH N
reflux in a sealed tube NH2 I ' Cl p NH2 (CXCIX) [0350] To freshly prepared sodium methoxide using sodium (900 mg, 39 inmol) 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).

O
N"O NBS (1 eq.) / AcOH 'O' reflux Br N"0 (CC) [0352] A mixture of 3-methoxy-6-nitroaniline (940 mg, 5.6 rmol) 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).

0 1) HCI

Br I j N,O + CN 2) NaOH Br I N N
NH2 NH2 " \%~
O

(CCI) [0354] 4-bromo-3-methoxy-6-nitroaniline (lg, 4.05 mmol) was mixed with cyanamide (1.7 g, 40 mmol) and heated to melt at 90 C. Concentrated HC1(10 mL) was cautiously added dropwise 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).

O OH DME / EtOH / H2O
Br \ N N B, Na2C03 \( /
I + I~.. OH Pd (PPh3)4, reflux / I O-O N NH \%\ \ N+
2 ;N

(CCII) [0356] A mixture of 7-broino-6-methoxybenzo[e][1,2,4]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/CHC13 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).

Pd dba 3 II + Xantphos aN N+ + O H Cs2CO3 Br S-NDioxane, reflux (CCIII) [0358] A mixture of 6-methoxy-7-o-tolylbenzo[e][1,2,4]triazin-3-amine-l-N-oxide (230 mg, 0.82 mmol), 4-bromo-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide (540 mg, 1.64 nnnol), 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 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 (CCIII) was further reacted as shown by scheme (CCIV).

O' N*N
O TM
i -_O aN N S-N,_H2, Raney N
H " N
---~a O
(CCIV) [0360] ' 4-(6-methoxy-l-oxy-7-o-tolyl-benzo[1,2,4]triazin-3-ylamino)-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide (140 mg, 0.26 mmol) was reduced in methanol (25 mL) TM
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%).'H NMR
DMSO-d6 8 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, 511),7.42 (br s, 1H), 7.79 (d, J= 8.8 Hz, 2H), 8.02 (s, 111), 8.18 (d, J= 8.9 Hz, 2H), 11.12 (s, 1H). MS (ESI+) ink = 519.

Example 163 Synthesis of 4-(6-hydroxy-7-o-tolyl-benzo[1,2,41triazin-3-ylamino)-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamiide N
.~O
HN~ //~~,, HO N H Nja NJ

(CCV) [0361] The synthesis of the title compound (CCV) can be generally described by the of reaction scheme (CCVI).

N pyridine HCI
a 0 H 160 C in a sealed tube \O NH S-N
O LD
(CCVI) [0362] 4-(6-methoxy-7-o-tolyl-benzo[1,2,4]triazin-3-ylamino)-N-(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 HCl (30 mL), and the crude product was purified by silica gel column with 20%

in CHC13 as an eluent to afford the title compound (CCVI), as a yellow solid (15 mg, 22%). 1H NMR DMSO-d6: 6 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, 1H), 7.79 (d, J= 8.8 Hz, 2H),'8.02 (s, 1H), 8.18 (d, J= 8.9 Hz, 2H), 11.12 (s, 1H). MS (ESI+) m/z = 519.

Example 164 Synthesis of 4-(6-ethoxy-7-o-tolyl-benzo[1,2,4]triazin-3-ylamino)-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide I N,N. / I S
HN
H NC]
\O / NN \

(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 HCl, 1750C / 0-IIIIIN. N a HO NNH2 a (CCVIII) [0364] The mixture of 7-bromo-6-methoxybenzo[e][1,2,4]triazin-3-amine-l-N-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 HCl (30 mL). The resulting yellow precipitate (100 mg, 62%) was isolated by filtration.

[0365] The product of reaction shown by scheme (CCVIII) was further reacted as shown by scheme (CCIX).

N+ H2, Raney Ni 'N IIiLIIINN

(CCIX) [0366] 3-amino-7-o-tolylbenzo[e][1,2,4]triazin-6-of-1-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).

Cs2CO3 / KI
N Acetone / CH3OH
N + ~Br reflux \ \ N

(CCX) [0368] A mixture of 3-amino-7-o-tolylbenzo[e][1,2,4]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).

Pd2 (dba)3 +
\ N'N H Xantphos Br ~. / S-NN Cs2CO3 N NH2 O Dioxane, reflux (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-1-yl-ethyl)-benzenesulfonamide (0.14 g, 0.43 mmol), cesium carbonate (0.21 g, 0.63 mmol), Xantphos (25 mg, 0.15 inmol), Pd2(dba)3(20 mg, 0.021 mmol) in 10 mL of anhydrous 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%).

DMSO-d6: S 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, 1H), 7.79 (d, J= 8.9 Hz), 8.02 (s, 1H), 8.18 (d, J= 8.9 Hz, 2H). 11.09 (s, 1H). MS
(ESI+) na/z =
533.

Example 165 Synthesis of 4-f7-(3-h_ydroxy-phenyl)-6-methoxy-benzof1,2,4 triazin-3-ylaminol-N-(2-pyrrolidin-i-yl-ethyl)-benzenesulfonamide HO N~N /
HN
O N~N \ I ~-\\
H

(CCXII) [0371] The synthesis of the title compound (CCXII) can be generally described by the sequence of reaction schemes (CCXIII), (CCXIV), (CCXV), and (CCXVI).

0- OBn' DME/EtOH/H2O
+ Na2CO3 Br \ N " N + Pd (PPh3)4, reflux ~O N NH2 B(OH)2 (CCXIII) [0372] The mixture of 7-bromo-6-methoxybenzo[e][1,2,4]triazin-3-amine-l-N-oxide (0.14 g, 0.52 mmol), 3-benzyloxyphenylboronic acid (0.24 g, 0.l0mmol), 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/CHC13 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 (CCXIV).

OBn Pd2 (dba)3 _ _ O H Xantphos \ + Br 0_U Cs CO
I2LJjN:N v N
DioxanrefluxO NNH2 (CCXIV) [0374] A mixture of 7-(3-(benzyloxy)phenyl)-6-methoxybenzo[e] [ 1,2,4]triazin-amine- 1 -N-oxide (0.11 g, 0.29 mmol), 4-bromo-N-(2-pyrrolidin-1-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 anhydrous 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).

OBn O-\ N
O H2, Raney Ni --0 u H
N H g-NN
O
(CCXV) [0376] 4-[7-(3-benzyloxy-phenyl)-6-methoxy-l-oxy-benzo[1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-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 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 (CCXVI).

OBn BBr3 N CHCI3 / EtOAc N _ room temp.
-_O H
N N S-_NN
O
(CCXVI) [0378] The above 4-[7-(3-benzyloxy-phenyl)-6-methoxy-benzo[1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonainide in chloroform (10 mL.) and ethyl acetate (10 mL) was stirred with 1M BBr3 in dichloromethane (10 mL, 10 mmol) at room temperature for 5 h. The reaction was quenched by methanol (1 niL). After removing the solvent, the crude product was purified by silica gel column chromatography with 20% % CH3OH/CHC13 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: 8 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, 1H), 6.99 (m, 2H), 7.27 (t, J= 8.7 Hz), 7.42 (br s, 1H), 7.80 (d, J= 8.8 Hz, 2H), 8.12 (s, 1H), 8.17 (d, J= 8.8 Hz, 2H), 9.55 (s, 1H), 11.10 (s, 1H). MS (ESI+) m/z = 521.

Example 166 Synthesis of 4-[6-(2-hydroxy-ethoxy)-7-o-tolyl-benzo[1,2,4]triazin ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide 'N I HN
HO~~O . ~~N~N N3 H
(CCXVII) [0379] The synthesis of the title compound (CCXVII) can be generally described by the sequence of reaction schemes (CCXVIII) and (CCXIX).

i I Cs2CO3 / KI

N Acetone / CH3OH 5,0 + HOreflux NN

(CCXVIII) [0380] 3-amino-7-o-tolylbenzo[e][1,2,4]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 stirred 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).

Pd2 (dba)3 N. O Xantphos H Cs2CO3 -N~~N Dioxane, reflux HOI / NNH + Br S~~

(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-1-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) chromatography using 30% CH3OH in as an eluent to afford the title compound (CCXVII), as an orange solid (0.01 g, 14%).1H
NMR DMSO-d6: b 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, 211), 4.42 (t, J= 4.9 Hz, 2H), 5.92 (s, 1H), 7.10 (d, J=
7.3 Hz, 1H), 7.24 (m, 4H), 7.48 (br s, 1H), 7.76 (m, 4H). MS (ESI+) m/z = 549.

Example 167 Synthesis ,of [7-(2,6-dichloro-phenyl)-5-methyl-benzo[1,2,41triazin-3-yll-[2-methyl-6-(2-pyrrolidin-l -yl-ethoxy)-pyrimidin-4-yll-amine cIXIN.N CI
Nill N
CI NN 1 O' N
H
(CCXX) [0383] The synthesis of the title compound (CCXX) can be generally described by the sequence of reaction schemes (CCXXI) and (CCXXII).

+ , N^N THF, reflux ~~ N
NaOCI ~ CI
excess (CCXXI) [0384] 4,6-dichloro-2-methylpyrimidine (1 g, 6.1 mmol) was added into a freshly prepared sodium 2-(pyrrolidin-1-yl)ethoxide from 2-(pyrrolidin-1-yl)ethanol (0.35g, 3.lrnmol) 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: 6 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, 1H).
[0385] The product of reaction shown by scheme (CCXXI) was further reacted as shown by scheme (CCXXII).

Pd2 (dba)3 CI Xantphos Cs2CO3 N: N + N INj Dioxane, reflux CIN'D

(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-1-yl)ethoxy)-6-chloro-2-inethylpyrimidine (270 mg, 1.12 mmol), cesium carbonate (550 mg, 1.7 mmol), Xantphos (65 mg, 0.11 mmol), Pd2(dba)3 (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 as an eluent to give the title compound (CCXX), as a yellow solid. (100 mg, 35%). 1H
NMR DMSO-d6: 6 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, 1H), 7.67 (s, 1H), 7.69 (s, 1H), 7.79 (s, 1H), 7.83 (d, J= 1.9 Hz, 1H), 8.23 8.23 (d, J= 1.8 Hz, 1H), 11.59 (s, 1H). MS(ESI+) m/z = 510.

Example 168 Synthesis of 4-chloro-3-{5-methyl-3-[2-methyl-6-(2-pyrrolidin-1-yl-ethoxy)-pyrimidin-4-ylaminol-benzo [1,2,4]triazin-7-yl}-phenol / CI

HO \ I N~ N IN_~ N
N-;~ N)O~iNC>
H

(CCXXIII) [0387] The synthesis of the title compound (CCXXIII) can be generally described by the sequence of reaction schemes (CCXXIV) and (CCXXV).

CI Pd2 (dba)3 Xantphos N' N + Cs2CO3 0 N N Dioxane, reflux N NH2 CI' v 'O-,--- N

(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-1-yl)ethoxy)-6-chloro-2-methylpyrimidine (270 mg, 1.12 mmol), cesium carbonate (1.1g, 3.37 mmol), Xantphos (130 mg, 0.22 mmol), Pd2(dba)3 (100 mg, 0.11 mmol) in anhydrous dioxane (25n-1L) 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).

CI
O N,.N Ni _N BBr3 NN N
H

(CCXXV) [0390] The N-(6-(2-(pyrrolidin-1-yl)ethoxy)-2-methylpyrimidin-4-yl)-7-(2-chloro-5-methoxyphenyl)-5-methylbenzo[e][1,2,4]triazin-3-amine (270 mg, 0.53 mmol) in chloroform (10 mL) was stirred with 1M 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/CHC13 as an eluent to afford the title compound (CCXXIII), as 'a yellow solid (100 mg, 38%). 1H NMR DMSO-d6: 8 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, 1H), 6.94 (d, J= 2.9 Hz, 1H), 7.40 (d, J= 8.8 Hz, 1H), 7.73 (s, 1H), 7.93 (s, 1H), 8.24 (d, J= 1.6 Hz, 1H), 9.95 (s, 1H), 11.55 (s, 1H).
MS(ESI+) na/z = 492.

Example 169 Synthesis of 4-[7-(6-chloro-2-fluoro-3-hydroxy-phenyl)-5-methyl-benzo [1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide CI

HO N'N HN
F N H ~N

(CCXXVI) [0391] The synthesis of the title compound (CCXXVI) can be generally described by the sequence of reaction schemes (CCXXVII), (CCXXVIII), (CCXXIX), and (CCXXX).
CI 1) nBuLi 2) B(OMe)3 F

(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 THE
(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
HC1(1 mL). THE was removed by vacuum. The crude product was diluted with 2N
HCl (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: b 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 (CCXXVIII).

DME / EtOH / H2O
Br N, CI Na2CO3 N + I Pd (PPhj)4, reflux N NH2 0 B(OH)2 F
(CCXXVIII) [0394] A mixture of 7-bromo-5-methylbenzo[e][1,2,4]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 Na2CO3 (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%

as an eluent to give a solid. The solid was washed with chloroform to give a yellow solid (220 mg, 39%). 111 NMR DMSO-d6 8 2.52 (s, 3H), 3.90 (s, 3H), 7.29 (t, J= 9 Hz, 1H), 7.43 (dd, J= 9 Hz, J= 1.7 Hz, 1H), 7.62 (s, 1H), 7.78 (br s, 2H), 8.02 (d, J=
1.6 Hz, 1H).
MS(ESI+) m/z = 319.

[0395] The product of reaction shown by scheme (CCXXVIII) was further reacted as shown by scheme (CCXXIX).

Pd dba CI 0\ N Xantphos3 \ N / S, Cs2CO3 0 N + Dioxane, reflux F NNH2 Br (CCXXIX) [0396] A mixture of 7-(6-chloro-2-fluoro-3-methoxyphenyl)-5-methylbenzo[e][1,2,4]triazin-3-amine (240 mg, 0.75 mmol), 4-bromo-N-(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:
S 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, 1H), 7.42 (br s, 1H), 7.46 (dd, J= 9 Hz, J= 1.8 Hz, 1H), 7.84 (m, 3H), 8.21 (m, 3H), 11.43 (s, 1H). MS(ESI+) na/z = 571.

[0397] The product of reaction shown by scheme (CCXXIX) was further reacted as shown by scheme (CCXXX).

CI
H
O N; N S NN BBrs I '~~ I ~1-1 I ~b 0 F
N N
H
(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 ing, 0.53 mmol) in chloroform (5 mL) was stirred with 1M 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/CHC13 as an eluent to afford the title compound ((CCXXVI), as a yellow solid (10mg, 6%). 1H NMR DMSO-d6: 6 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, 1H), 7.42 (br s, 1H), 7.85 (m, 3H), 8.22 (m, 3H), 10.4 (br s, 1H), 11.42 (s, 1H). MS(ESI+) m/z = 557.

Example 170 Synthesis of 4-17-(5-hydroxy-2-methyl-phenyl)-5-methyl-benzo[1,2,41triazin-3-ylaminol-N-(2-pyrrolidin-l-yl-ethyl)-benzenesulfonamide \ I ` 0 Fi0 N HN-~_ N H N~
(CCXXXI) [0399] The synthesis of the title compound (CCXXXI) can be generally described by the sequence of reaction schemes (CCXXXII), (CCXXXIII), (CCXXXN), (CCXXXXV), and (CCXXXVI).

NH2 1) NaNO2 / HBr 2) CuBr (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 inL).
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%).

[04011 The product of reaction shown by scheme (CCXXXII) was further reacted as shown by scheme (CCXXXIII).

1) nBuLi Br 2) B(OMe)3 (CCXXXIII) [04021 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 THE (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 HCl (1 mL). The THE was removed by vacuum. The crude product was diluted with 2N HCl (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%).

[04031 The product of reaction shown by scheme (CCXXXIII) was further reacted as shown by scheme (CCXXXIV).

Pd(PPh3)4, Na2CO3 LB(OH)2 Br N; DME / EtOH / H2O
+ N NH2 O

(CCXXXIV) [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% CH3OH/CHC13 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: S 2.22 (s, 3H), 2.54 (s, 3H), 3.77 (s, 3H), 6.89 (s, 1H), 6.90 (d, J= 3 Hz, 1H), 7.24 (d, J= 8 Hz, 1H), 7.66 (br s, 1H), 7.68 (s, 1H), 7.95 (d, J=
1.6Hz, 1H). MS(ESI+) fn/z = 281.

[0405] The product of reaction shown by scheme (CCXXIV) was further reacted as shown by scheme (CCXXXV).

Pd2(dba)3 H Xantphos N ~/ N ~O Cs2CO3 + Dioxane reflux N;N
~aBr N NHg (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-pyrrolidin-1-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%).1H NMR DMSO-d6: 8 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, 1H), 7.40 (br s, 1H), 7.83 (d, J= 9Hz, 2H), 7.89 (d, J=1.6Hz, 1H), 8.15 (d, J=
9Hz, 2H), 11.35 (br s, 1H).

[0407] The product of reaction shown by scheme (CCXXV) was further reacted as shown by scheme (CCXXXVI).

0 BBr s \\ NH
NN S.
N N
H
(CCXXXVI) [0408] A mixture of 4-[7-(5-methoxy-2-methyl-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamino]-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide (80 mg, 0.15 mmol) in chloroform (10 mL) was stirred with 1M BBr3 in dichloromethane (10 mL, 10 minol) 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/CHC13 as an eluent to afford the title product (CCXXXI), as a yellow solid (20mg, 26%). 1H NMR DMSO-d6: S 1.62 (in, 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, 1H), 7.40 (br s, 1H), 7.84 (m, 3H), 8.09 (d, J= 1.4Hz, 1H), 8.21 (d, J= 9Hz, 2H), 9.38 (br s, 1H), 11.34 (br s, 1H). MS
(ESI+) ma/z = 519.

Example 171 Synthesis of [7-(2'-hydroxyethyl-isopropylamino)-6-methyl-benzo[1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzene-4-sulfonamide Y
O H
k , N
HO' N N'~ N So N I N
H
(CCXXXVII) [04091 To a solution of 2-(isopropylamirio)ethanol 48 L, 0.416 mmol), 4-(7-bromo-6-methyl-benzo[ 1,2,4]triazin-3-ylamino)-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonam.ide (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: 8 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, 1H), 7.67 (s, 1H), 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, 1H). MS (ESI+) m/z = 514.2.

Example 172 Synthesis of 4-(7-f [2-(2-hydroxy-ethoxy)-ethyll-isopropyl-amino)-6-methyl-benzo [1,2,4]triazin-3-ylamino)-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide Y H
phi N N N S`
N H
OH

(CCXXXVIII) [0410] The title product was synthesized and had the following characteristics. 1H NMR
DMSO-d6: 8 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, 1H), 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, 1H), 7.69 (s, 1H), 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, 1H). MS (ESI+) m/z = 558.3.

Example 173 Synthesis of 4-(7-bromo-6-methyl-benzoIi,2,4Itriazin-3-ylamino)-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide H
Br N N OS' N N
~~ O V
N N
H

(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: 6 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, 1H), 7.87 (t, J 6.1 Hz, 1H), 8.18 (d, J = 8.8 Hz, 1H), 8.68 (s, 1H), 9.46-9.57 (br s, 1H), 11.4 (s, 1H). MS (ESI+) m/z =
493.

Example 174 Synthesis of [6-methyl-7-(3-methyl-thiophen-2-yl)-benzo[1,2,4]triazin-3-yll-[4-(2-pyrrolidin-1-yl-ethoxy)-phenyll-amine O~/~
g N N
N N/ I
H

(CCXL) [0412] To a mixture of 3-methylthiophen-2-yl-2-boronic acid (252 mg, 2 mmol), amino-7-bromo-6-methyl benzo-1,2,3-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), 1-(2-(4-bromophenoxy)ethyl)pyrrolidine (121 L, 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 %). 1H NMR DMSO-d6:

8 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, 1H), 7.58 (d, J = 5.2 Hz, 1H), 7.66 (s, 1H), 7.85 (d, J = 6.95 Hz, 2H), 8.09 (s, 1H), 10.71 (s, 1H). MS
(ESI+) m/z = 446.1.

Example 175 Synthesis of 14-[6-methyl-7-(4-methyl-pyridin-3-yl)-b enzo f 1,2,4]triazin-3-ylaminol-phenyll-(4-methyl-piperazin-1-yl)-methanone mesylate O
N~ NN N
N N
H
(CCXLI) [0414] To a mixture of 4-methylpyridin-3-yl-3-boronic acid (272 mg, 2 mmol), 3-amino-7-bromo-6-methyl benzo-1,2,3-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), 1-(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: S 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, 1H), 7.81 (s, 3H), 8.10 (d, J = 8.6 Hz, 2H), 8.17 (s, 1H), 8.5 (s, 1H), 8.62 (d, J = 4.5 Hz, 1H), 9.67-9.76 (br s, 1H), 11.15 (s, 3H). MS (ESI+) m/z = 454.3.

Example 176 Synthesis of {4-[7-(4-chloro-pyridin-3-yl)-6-methyl-b enzo [1,2,4]triazin-3-ylaminol-phenyl}-(4-methyl-piperazin-1-y1)-methanone mesylate CI
O
N~ I N;N , N
N N e (CCXLII) [0416] To a mixture of 4-chloropyridin-3-yl-3-boronic acid (314 mg, 2 mmol), 3-amino-7-bromo-6-methyl benzo-1,2,3-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), 1-(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: 8 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, 1H), 7.78 (s, 3H), 8.07 (d, J = 2.65 Hz, 2H), 8.09 (d, J = 8.65, 1H), 8.27 (s, 1H), 8.59 (d, J = 2.4 Hz, 1H), 9.67-9.76 (br s, 1H), 11.17 (s, 1H). MS (ESI+) m/z =
474.2.
Example 177 Synthesis of {4-[7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-33-ylaminol-phenyl}-piperazin-1-yl-methanone O
NN NH
H
(CCXLIII) [04181 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-1-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). 1H
NMR DMSO-d6: S 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, 1H), 7.45 (dd, J = 1.75 and 6.85 Hz, 2H), 7.64 (dd, J = 1 and 1.85 Hz, 1H), 7.95 (d, J = 1.85 Hz, 1H), 8.09 (dd, J
= 1.7 and 6.85 Hz, 2H), 11.12 (s, 1H). MS (ESI+) m/z = 453.3.

Example 178 Synthesis of [7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-yll-[2-(2-pyrrolidin-1-vl-ethoxy)-pyridin-4-yll-amine mesylate N.
NN O-'iN
H

(CCXLIV) [0419] The title product wa synthesized,, having the following characteristics. 1H
NMR DMSO-d6: 8 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, 1H), 7.66 (d, J =
1.75 Hz, 1H), 7.29 (d, J = 1.0 Hz, 1H), 8.08 (d, J =1.9 Hz, 1H), 8.12 (d, J =
5.8 Hz, 1H), 11.39 (s, 1H). MS (ESI+) m/z = 455.3.

Example 179 Synthesis of 7-(2,6-dichloro-3-methoxyphen yl)-5-methylb enzo [el [1,2,41 triazin-3-amine CI
O N,N
CI NNH

(CCXLV) [0420] To prepare the title compound (CCXLV), an intermediate compound 96 (2,6-dichloro-3-inethoxypheriylboronic acid) was synthesized first.

(CI
O B(OH)2 JC
CI

[0421] To synthesize compound 96, to 2,4-dichloroanisole (5 mL, 36 mmol) in THE
(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 inmol) and allowed to warm to room temperature over 16 h. The solid suspension was poured into a 1 N HCl 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%). 'H NMR DMSO-d6: S 8.53 (s, 2H), 7.29 (d, J= 8.8 Hz, 1H), 7.06 (d, J= 8.8 Hz, 1H), 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 crude pale-orange solid (922 mg, 83%) which was used as is with no further purification.
LC
retention time: 2.98 min, MS (ESI+) ma/z = 334.9.

Example 180 Synthesis of 4-[7-(2,6-dichloro-3-metboxy-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide CI
O
O \ I / N, N / S, HN ~
CI NN, \ I N
H

(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 11111101), 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 CHC13(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 [1,2,4]triazin-3-ylaminol-N-(2-pyrrolidin-1-yl-ethyl)-benzenesulfonamide CI
HO N` N / oS,O
~HfV
CI NN \ I ~N~
H
(CCXLVII) [0424] The general aryl methoxy deprotection procedure was employed using aryl methoxy compound (CCXLVI) (Example 180) (337 mg, 0.57 mmol) and BBr3 (271 L, 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 HCl 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 :8 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, 1H), 7.43 (d, J= 8.9, Hz, 1H), 7.75 (dd, J=1.8, 1.0 Hz, 1H),'7.89 (d, J= 8.9 Hz, 2H), 8.02 (t, J= 5.8 Hz 1H), 8.11 (d, J= 1.7 Hz, 1H), 8.26 (d, J= 8.9 Hz, 2H), 10.78 (br s, 1H), 10.85 (s, 111), 11.47 (s, 1H).

Example 182 Synthesis of 4-chloro-3-[3-(4-{[(2-hydroxy-ethyl)-isopropyl-aminol-methyl}-phenylamino)-5-methyl-benzo [1,2,41 triazin-7-vIl -phenol CI

HO I / N,'N (yy~OH
N N
H
(CCXLVIII) [0425] To synthesize the title product, intermediate compounds 97 and 98 shown below were used.

CI
HO \ / I N, N

N-,,_iOH
Br [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 compound (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: 6 1.31 (d, J= 6.6, Hz, 3H), 1.37 (d, J= 6.6, Hz, 1H), 2.69 (s, 3H), 3.01-3.11 (m, 1H), 3.22-3.28 (m, 1H), 3.47-3.54 (m, 1H), 3.62-3.72 (m, 2H), 4.29 (dd, J
= 13.2, 6.4, Hz, 1H), 4.36 (dd, J=13.2, 4.1, Hz, 1H), 5.31 (br s, 1H), 6.88 (dd, J= 8.7, 2.9, Hz, 1H), 6.93 (d, J= 2.9 Hz, 1H), 7.41 (d, J= 8.7, Hz, 1H), 7.61 (d, J=
8.6 Hz, 1H), 7.89 (dd, J=1.8, 1.0 Hz, 1H), 8.13 (d, J= 8.7 Hz, 1H), 8.19 (d, J= 1.7 Hz, 1H), 8.94 (br s, 1H), 9.95 (s, 1H), 11.17 (s, 1H).

Example 183 Synthesis of N-(4-(2-(pyrrolidin-1-yl)ethoxy)phenyl)-7-(2,6-dichloro-3-methoxyphenyl)-5-methylbenzo [e] 11,2,41 triazin-3-amine CI
~\ I N' 00 N N
CI \ I \ I ~/
N N
H
(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 CHC13(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-1-yl)ethoxy)phenylamino)-5-methylbenzo[el [1,2,41triazin-7-yl)-2,4-dichlorophenol CI
HO N'N O~/~N
N N
H
(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 L, 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. NaHCO3 and extracted with EtOAc (2x 150 mL). The combined organic layers were dried, filtered through celite and concentrated in vacuo. The HC1 salt of the product was formed using MeOH and 4M HCl 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 1H NMR DMSO-d6: 8 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, 1H), 7.42 (d, J= 8.9, Hz, 1H), 7.65 (dd, J= 1.8, 1.0 Hz, 1H), 7.99 (d, J= 9.1 Hz, 2H), 8.02 (d, J= 1.7 Hz, 1H), 10.18 (br s, 1H), 10.71 (s, 1H), 10.90 (s, 1H).

Example 185 Synthesis of benzvl 4-(4-(7-(2,6-dichloro-3-methoxyphenyf-5-methylbenzo[el [1,2,4]triazin-3-ylamino)phenylsulfonyl)piperidine-l-carboxylate CI
O~,O
I \
~O \ / I N; 4)rs-a CI
/
N H II
O
(CCLI) [04291 To synthesize the title product, intermediate compound 99 shown below was used.

S I \
Br \ ON O

O

[04301 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. Chromatography 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[e] [1,2,4]triazin-7-yl)-2,4-dichlorophenol CI

5s' HO NCI \NH
N N
H
(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 L, 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 HCl salt of the title product was formed using MeOH and 4M HCl 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 = 544.01H NMR DMSO-d6: S 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, 1H), 7.13 (d, J= 8.9, Hz, 1H), 7.44 (d, J= 8.8, Hz, 1H), 7.77 (s, 1H), 7.89 (d, J= 9.0 Hz, 2H), 8.14 (d, J= 1.3 Hz, 1H), 8.34 (d, J= 8.9 Hz, 2H), 8.43 (br s, 1H), 8.96 (br s, 1H), 10.75 (s, 1H), 11.59 (s, 1H).

Example 187 Synthesis of {4- f7-(3,5-dimethyl-isoxazol-4-yl)-5-methyl-benzo f 1,2,41 triazin-3-ylaminol-phenyll-(4-methyl-piperazin-l-vl)-methanone O O
N\ N,,N ~ \ N
NN / NI
H

(CCLIII) [0432] A mixture of above amine (compound (CCLII), Example 186) (125 mg, 0.46 mmol), 1-(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
mot 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: 8 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, 1H), 8.13 (d, J = 7.0 Hz, 2H), 8.22 (d, J =
1.7 Hz, 1H), 9.86-9.97 (br s, 1H), 11.25 (s, 1H). MS (ESI+) nz/z = 458.2 [0433] The following Examples 188-192 describe the synthesis of some of the intermediate compounds 100 (7-bromo-5-methyl-benzo[1,2,4]triazin-3-ylamine), 101 (7-bromo-6-methyl-benzo[1,2,4]triazin-3-ylamine),102 ((4-bromo-phenyl)-(4-methyl-piperazin-1-yl)-methanone),103 (4-bromo-N-(2-pyrrolidin-1-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 (1-[3-(3-bromo-benzenesulfonyl)-propyl]-pyrrolidine), and 108 (7-(2,6-dimethyl-phenyl)-5-methyl-benzo[1,2,4]triazin-3-ylamine) that were used to synthesize the title compounds of Examples 188-190, 193-205, 208, 209, and 213 that follow.

Br N

Br \ N,NH2 i \ N
Br ,,e N N
H
Br Br H
Br N,_,,-,,N
O v H
Br N`'.
O N%
Boc ~ I

Br \ 0>S~0 N\

\ N

I~
N~NH2 Example 188 Synthesis of (4-bromo-phenyl)-(4-methyl-piperazin-1-y1)-methanone (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 l, 5.1 mmol) and EDC (980 mg, 5.1 minol). 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, CH2C12/MeOH, 90:10) to yield the intermediate compound 102. 1H NMR CDC13: 8 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) [04351 To a solution of 4-bromo-benzoic acid (5.02 g, 25 mmol) in 200 mL
acetonitrile were added NI,NI-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 CH2C12, and was washed successively with water (150 mL) and aqueous saturated NaHCO3 (150 mL). The organic phase was dried (MgSO4) and the solvent was evaporated. The crude product was purified by flash column chromatography Rf =
0.2, (CH2C12/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) [04361 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-benzoylamino)-pyrrolidine-l-carboxylic acid tert-butyl ester (intermediate compound 106) [04371 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 (MgSO4) and the solvent was removed. The crude product was purified by silica gel column chromatography (Rf= 0.22, CH2C12/1VIeOH, 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 CH2C12 were added di-tert-butyldicarbonate (925 ing, 4.24 mmol) and triethylamine (596 l, 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, CH2C12/MeOH, 90:10) to afford the title intermediate compound 106, as a viscous, colorless oil (1.1 g, 70%). MS (ESI+) na/z =
369/371.

Example 192 Synthesis of 1-13-(3-bromo-benzenesulfonyl)-propyll-pyrrolidine (intermediate compound 107) [04381 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 CH2C12 (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 (MgS04) and the product was purified by silica gel column chromatography (Rf= 0.53, EtOAc/hexanes 50:50) to give 1-bromo-3-(3-bromo-propane-1-sulfonyl)benzene as a colorless solid (5.47 g, 76 %).

[04391 A mixture of 1-bromo-3-(3-bromo-propane-l-sulfonyl)benzene (5.47 g, 16.4 mmol Cs2CO3 (10.7 g, 32.8 mmol) and pyrrolidine (2.712nL, 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 CH2Cl2 (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-phenyl)-5-riethyl-benzo [1,2,4]triazin-3-yll- [4-(2-pyrrolidin-1-yl-ethoxy)-phenyll-amine trifluoroacetate OMe CI N' N
N N /
H
(CCLIV) [04401 To a solution of intermediate compound 100 (24 mg, 0.1 mmol) in 2 mL
DMA
were added 5-chloro-2-methoxyphenylboronic acid (75 mg, 0.4 inmol) in ethanol (500 l), K2C03 (2.8 mg, 0.02 mmol) in water (200 l), PPh3 (3 mg, 0.01 mmol) and Pd2(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 CH2Cl2 (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 NaHCO3 solution. The product was extracted with CH2Cl2, 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[ 1,2,4]triazin-3-ylamine (25.1 mg, 83 %). To a solution of the purified precursor in 4 mL anhydrous toluene were added 1-[2-(4-bromo-phenoxy)-ethyl] -pyrrolidine (41 l, 0.2 rmol), BINAP (3.7 mg, 6 mol), Pd2(dba)3 (2.3 mg, 2.5 mol) and KO tBu (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 NaCl solution. The product was extracted with CH2Cl2 and purified by reverse phase preparative HPLC to afford the title copound(CCLIV) as trifluoroacetate salt, in a form of DEMANDES OU BREVETS VOLUMINEUX
LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVETS
COMPREND PLUS D'UN TOME.

NOTE: Pour les tomes additionels, veillez contacter le Bureau Canadien des Brevets.

JUMBO APPLICATIONS / PATENTS

THIS SECTION OF THE APPLICATION / PATENT CONTAINS MORE
THAN ONE VOLUME.

NOTE: For additional volumes please contact the Canadian Patent Office.

Claims (88)

1. A compound of structure (I):

wherein each of A is independently selected from a group consisting of C, CH, N, 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 C, CH, N, 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 A1' and B1" is C;

R0 is selected from a group consisting of H and lower alkyl;

each L is independently selected from a group consisting of a bond, and a substituted or unsubstituted alkyl, alkenyl, or alkynyl linking moiety;

R1 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, PO4R', OPO2R', PO3R', PO2R', and a 3-6 membered heterocycle with one or more heterocyclic atoms with each heteroatom independently optionally 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, branched alkylamino, and a closed 3-6 membered carbocycle or heterocycle, with each heteroatom in the 3-6 membered heterocycle optionally carrying any R' group on it, and wherein each R' is independent in case there is more than one R';

R2 is a substituent situated at position 5, 6 or 8 of the ring, wherein R2 is selected from a group consisting of methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, tert-butyl, iso-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, and a 3-6 membered carbocycle or heterocycle attached to positions 5, 6 or 8 directly or through group L, each heteroatom independently optionally 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, 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", and 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 each other group R3, or a pharmaceutically acceptable salt, hydrate, solvate, N-oxide, or individual diastereomer thereof.
2. The compound of claim 1 selected from the group consisting of:

3. The compound of claim 1 selected from the group consisting of:

4. The compound of claim 1 selected from the group consisting of:

5. The compound of claim 1 selected from the group consisting of:

and
6. The compound of claim 1 selected from the group consisting of:

and
7. The compound of claim 1 selected from the group consisting of:

8. The compound of claim 1 selected from the group consisting of:

9. The compound of claim 1 selected from the group consisting of,
10. The compound of claim 1 selected from the group consisting of:

and
11. The compound of claim 1 having the formula:

12. The compound of claim 1 selected from the group consisting of:

and
13. The compound of claim I selected from the group consisting of.

14. The compound of claim 1 selected from the group consisting of:

15. The compound of claim 1 selected from the group consisting of:

16. The compound of claim 1 selected from the group consisting of:

17. The compound of claim 1 selected from the group consisting of:

18. The compound of claim 1 selected from the group consisting of:

19. The compound of claim 1 selected from the group consisting of:

20. Use of a therapeutically effective amount of at least one compound as defined in any one of claims 1 to 19 to treat a disorder associated with compromised vasculostasis in a subject in need thereof.
21. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat a disorder associated with compromised vasculostasis.
22. The use of claim 20 or 21, wherein the disorder is myocardial infarction, stroke, congestive heart failure, an ischemia or reperfusion injury, cancer, arthritis, 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 use of claim 20 or 21, wherein the disorder is vascular leakage syndrome (VLS).
24. The use of claim 20 or 21, wherein the disorder is cancer.
25. The use of claim 20 or 21, wherein the disorder is a vitreoretinal disease.
26. The use of claim 20 or 21, wherein the disorder is ARDS.
27. The use of claim 20 or 21, wherein the disorder is autoimmune disease.
28. The use of claim 20 or 21, wherein the disorder is burn.
29. The use of claim 20 or 21, wherein the disorder is stroke.
30. The use of claim 20 or 21, wherein the disorder is myocardial infarction.
31. The use of claim 20 or 21, wherein the disorder is ischemia or reperfusion injury.
32. The use of claim 20 or 21, wherein the disorder is arthritis.
33. The use of claim 20 or 21, wherein the disorder is edema.
34. The use of claim 20 or 21, wherein the disorder is transplant rejection.
35. The use of claim 20 or 21, wherein the disorder is inflammatory disease.
36. The use of claim 20 or 21, wherein the disorder is congestive heart failure.
37. The use of claim 20 or 21, wherein the disorder is associated with a kinase.
38. The use of claim 37, wherein the kinase is a tyrosine kinase.
39. The use of claim 37, wherein the kinase is a serine kinase or a threonine kinase.
40. The use of claim 37, wherein the kinase is a Src family kinase.
41. A pharmaceutical composition comprising at least one compound as defined in any one of claims 1 to 19 and a pharmaceutically acceptable carrier therefor.
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 is for use for treatment of disorders associated with compromised vasculostasis, and wherein the pharmaceutical composition comprises at least one compound as defined in any one of claims 1 to 19 and a pharmaceutically acceptable carrier therefor.
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 is for use 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, 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 as defined in any one of claims 1 to 19 and a pharmaceutically acceptable carrier therefor.
44. The article of manufacture of claim 43, wherein the disorder is cancer.
45. Use of a therapeutically effective amount of at least one compound as defined in any one of claims 1 to 19, or pharmaceutically acceptable salts, hydrates, solvates, and individual diastereomers thereof, to treat a disorder associated with compromised vasculostasis in a subject in need thereof.
46. Use of at least one compound as defined in any one of claims 1 to 19, or pharmaceutically acceptable salts, hydrates, solvates, and individual diastereomers thereof, in the manufacture of a medicament to treat a disorder associated with compromised vasculostasis.
47. The use of claim 45 or 46, wherein the disorder is vascular leakage syndrome (VLS).
48. The use of claim 45 or 46, wherein the disorder is cancer.
49. The use of claim 45 or 46, wherein the disorder is a vitreoretinal disease.
50. The use of claim 45 or 46, wherein the disorder is ARDS.
51. The use of claim 45 or 46, wherein the disorder is an autoimmune disease.
52. The use of claim 45 or 46, wherein the disorder is burn.
53. The use of claim 45 or 46, wherein the disorder is stroke.
54. The use of claim 45 or 46, wherein the disorder is myocardial infarction.
55. The use of claim 45 or 46, wherein the disorder is ischemia or reperfusion injury.
56. The use of claim 45 or 46, wherein the disorder is arthritis.
57. The use of claim 45 or 46, wherein the disorder is edema.
58. The use of claim 45 or 46, wherein the disorder is transplant rejection.
59. The use of claim 45 or 46, wherein the disorder is inflammatory disease.
60. Use of a therapeutically effective amount of at least one compound as defined in any one of claims 1 to 19, or pharmaceutically acceptable salts, hydrates, solvates, and individual diastereomers thereof, in combination with an anti-inflammatory agent, chemotherapeutic agent, immunomodulatory agent, therapeutic antibody, or a protein kinase inhibitor, to treat a disorder associated with compromised vasculostasis to a subject in need thereof.
61. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent myocardial infarction in a subject in need thereof.
62. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent vascular leakage syndrome (VLS) in a subject in need thereof.
63. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent cancer in a subject in need thereof.
64. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent stroke in a subject in need thereof.
65. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent ARDS in a subject in need thereof.
66. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent burns in a subject in need thereof.
67. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent arthritis in a subject in need thereof.
68. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent edema in a subject in need thereof.
69. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent retinopathy or vitreoretinal disease in a subject in need thereof.
70. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent ischemic or reperfusion related tissue injury or damage in a subject in need thereof.
71. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent autoimmune disease in a subject in need thereof.
72. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent transplant rejection in a subject in need thereof.
73. Use of an effective amount of at least one compound as defined in any one of claims 1 to 19 to treat or prevent inflammatory disease in a subject in need thereof.
74. A process for making a pharmaceutical composition comprising combining a combination of at least one compound as defined in any one of claims 1 to 19, or its pharmaceutically acceptable salts, hydrates, solvates, salts and individual diastereomers thereof, and a pharmaceutically acceptable carrier.
75. Use of at least one compound as defined in any one of claims I to 19, or pharmaceutically acceptable salts, hydrates, solvates, and individual diastereomers thereof, in combination with an anti-inflammatory agent, chemotherapeutic agent, immunomodulatory agent, therapeutic antibody, or a protein kinase inhibitor, in the manufacture of a medicament to treat a disorder associated with compromised vasculostasis.
76. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent myocardial infarction.
77. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent vascular leakage syndrome (VLS).
78. Use of at least one compound as defined in any one of claims 1to 19 in the manufacture of a medicament to treat or prevent cancer.
79. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent stroke.
80. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent ARDS.
81. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent burns.
82. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent arthritis.
83, Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent edema.
84. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent retinopathy or vitreoretinal disease.
85. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent ischemic or reperfusion related tissue injury or damage.
86. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent autoimmune disease.
87. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent transplant rejection.
88. Use of at least one compound as defined in any one of claims 1 to 19 in the manufacture of a medicament to treat or prevent inflammatory disease.
CA2567574A 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases Expired - Fee Related CA2567574C (en)

Applications Claiming Priority (5)

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
PCT/US2005/012057 WO2005096784A2 (en) 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases

Publications (2)

Publication Number Publication Date
CA2567574A1 CA2567574A1 (en) 2005-10-20
CA2567574C true CA2567574C (en) 2013-01-08

Family

ID=35125563

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2567574A Expired - Fee Related CA2567574C (en) 2004-04-08 2005-04-07 Benzotriazine inhibitors of kinases

Country Status (10)

Country Link
US (3) US7456176B2 (en)
EP (2) EP1809614B1 (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)

Families Citing this family (30)

* 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
EP1549614A4 (en) * 2002-10-03 2008-04-16 Targegen Inc Vasculostatic agents and methods of use thereof
EP1809614B1 (en) * 2004-04-08 2014-05-07 TargeGen, Inc. Benzotriazine inhibitors of kinases
NZ588896A (en) * 2004-08-25 2012-05-25 Targegen Inc Heterocyclic compounds and methods of use
GB0501999D0 (en) * 2005-02-01 2005-03-09 Sentinel Oncology Ltd Pharmaceutical compounds
AU2006244072B2 (en) 2005-05-10 2012-09-20 Intermune, Inc. Pyridone derivatives for modulating stress-activated protein kinase system
BRPI0606172A2 (en) * 2005-06-08 2009-06-02 Targegen Inc methods and compositions for treating eye disorders
US8604042B2 (en) * 2005-11-01 2013-12-10 Targegen, Inc. Bi-aryl meta-pyrimidine inhibitors of kinases
RU2448959C2 (en) 2005-11-01 2012-04-27 Таргеджен, Инк. Bi-aryl-metha-pyrimidine kinase inhibitors
US8133900B2 (en) 2005-11-01 2012-03-13 Targegen, Inc. Use of bi-aryl meta-pyrimidine inhibitors of kinases
JP2009519908A (en) * 2005-12-08 2009-05-21 ミレニアム・ファーマシューティカルズ・インコーポレイテッド Bicyclic compounds having kinase inhibitory activity
WO2007127366A2 (en) * 2006-04-25 2007-11-08 Targegen, Inc. Kinase inhibitors and methods of use thereof
CA2672893C (en) 2006-12-15 2016-02-23 Abraxis Bioscience, Inc. Triazine derivatives and their therapeutical applications
EP2219671A4 (en) * 2007-11-09 2011-02-09 Salk Inst For Biological Studi Use of tam receptor inhibitors as immunoenhancers and tam activators as immunosuppressors
MX2010012080A (en) * 2008-05-05 2011-04-11 Univ Winthrop Hospital Method for improving cardiovascular risk profile of cox inhibitors.
ES2567283T3 (en) 2008-06-03 2016-04-21 Intermune, Inc. Compounds and methods to treat inflammatory and fibrotic disorders
SG196784A1 (en) 2008-12-03 2014-02-13 Scripps Research Inst Stem cell cultures
EA019110B1 (en) 2008-12-29 2014-01-30 Фовеа Фармасьютикалз Substituted quinazoline compounds
EP2396324A1 (en) 2009-02-13 2011-12-21 Fovea Pharmaceuticals Ý1, 2, 4¨triazolo ý1, 5 -a¨pyridines as kinase inhibitors
GB0908905D0 (en) 2009-05-26 2009-07-01 Sentinel Oncology Ltd Pharmaceutical compounds
WO2010144550A1 (en) 2009-06-09 2010-12-16 Abraxis Bioscience, Llc Triazine derivatives and their therapeutical applications
JP2012529517A (en) 2009-06-09 2012-11-22 アブラクシス バイオサイエンス リミテッド ライアビリティー カンパニー Benzyl-substituted triazine derivatives and their therapeutic applications
TW201204723A (en) * 2010-06-22 2012-02-01 Fovea Pharmaceuticals Heterocyclic compounds, their preparation and their therapeutic application
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
AR092742A1 (en) 2012-10-02 2015-04-29 Intermune Inc ANTIFIBROTIC PYRIDINONES
WO2015153683A1 (en) 2014-04-02 2015-10-08 Intermune, Inc. Anti-fibrotic pyridinones
WO2016089208A2 (en) 2014-12-04 2016-06-09 Stichting Maastricht Radiation Oncology "Maastro-Clinic" Sulfonamide, sulfamate and sulfamide derivatives of anti-cancer agents
JP7023080B2 (en) 2016-10-31 2022-02-21 東ソー株式会社 Method for producing aromatic compounds
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
IL44058A (en) * 1973-02-02 1978-10-31 Ciba Geigy Ag 3amino-1,2,4-benzotriazine 1,4-di-noxide derivatives, their preparation and compositions for the control of microorganisms containing them
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
NZ316340A (en) 1995-09-25 1999-04-29 Sanofi Winthrop Inc 1,2,4-benzotriazine oxides aqueous parenteral formulations
US5827850A (en) 1995-09-25 1998-10-27 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
WO1997030707A1 (en) * 1996-02-23 1997-08-28 Eli Lilly And Company NON-PEPTIDYL VASOPRESSIN V1a ANTAGONISTS
DE59707681D1 (en) * 1996-10-28 2002-08-14 Rolic Ag Zug Crosslinkable, photoactive silane derivatives
CN1244215B (en) * 1996-11-20 2010-11-03 荷兰克鲁塞尔公司 Improved method for the 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
US6235736B1 (en) 1997-06-24 2001-05-22 Nikken Chemicals Co., Ltd. 3-anilino-2-cycloalkenone derivatives
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
US6297258B1 (en) * 1998-09-29 2001-10-02 American Cyanamid Company Substituted 3-cyanoquinolines
US6288082B1 (en) * 1998-09-29 2001-09-11 American Cyanamid Company Substituted 3-cyanoquinolines
EP1126843A4 (en) 1998-10-29 2005-06-15 Bristol Myers Squibb Co 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
BR0012046A (en) * 1999-07-01 2002-05-14 Ajinomoto Kk Heterocyclic compound, pharmaceutical composition, inhibitor of activation of ap-1 or an inhibitor of activation of nf-kappab, inhibitor of the production of inflammatory cytokine, and inhibitor of the production of matrix metalloprotease or inhibitor of the expression of inflammatory cell adhesion factor
TWI262914B (en) 1999-07-02 2006-10-01 Agouron Pharma Compounds and pharmaceutical compositions for inhibiting protein kinases
TR200200099T2 (en) 1999-07-23 2002-06-21 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
EP1223170B1 (en) 1999-10-12 2005-12-28 Takeda Pharmaceutical Company Limited Pyrimidine-5-carboxamide compounds, process for producing the same and use thereof
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
WO2001064646A2 (en) * 2000-03-01 2001-09-07 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
EP1273287A1 (en) * 2000-04-04 2003-01-08 Shionogi & Co., Ltd. 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
US6875483B2 (en) 2000-07-06 2005-04-05 Fuji Photo Film Co., Ltd. Liquid crystal composition comprising liquid crystal molecules and alignment promoter
DE60132702T2 (en) * 2000-08-11 2009-03-19 Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield Heterocyclic compounds 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
EP1401833A2 (en) * 2001-05-28 2004-03-31 Aventis Pharma S.A. Chemical derivatives and the use thereof as an anti-elomerase agent
EP1392662B1 (en) * 2001-05-29 2009-01-07 Bayer Schering Pharma Aktiengesellschaft Cdk inhibiting pyrimidines, production thereof and their use as medicaments
US6689778B2 (en) * 2001-07-03 2004-02-10 Vertex Pharmaceuticals Incorporated Inhibitors of Src and Lck protein kinases
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
DK1438053T3 (en) * 2001-10-17 2008-12-08 Boehringer Ingelheim Pharma Pyrimidine derivatives, drug containing these compounds, their use and methods for their preparation
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
DE60314500T2 (en) * 2002-03-01 2008-02-07 Smithkline Beecham Corp. DIAMINOPYRIMIDINE AND ITS USE AS ANGIOGENESEHEMMER
MY141867A (en) 2002-06-20 2010-07-16 Vertex Pharma Substituted pyrimidines useful as protein kinase inhibitors
JP4570955B2 (en) 2002-07-09 2010-10-27 バーテクス ファーマスーティカルズ インコーポレイテッド Imidazoles with protein kinase inhibitory activity
IL166241A (en) 2002-07-29 2011-12-29 Rigel Pharmaceuticals Inc 2,4-pyrimidinediamine compounds for use in the treatment of autoimmune diseases
MXPA05001277A (en) * 2002-08-02 2005-10-06 Ab Science 2-(3-aminoaryl)amino-4-aryl-thiazoles and their use as c-kit inhibitors.
EP1546117A2 (en) * 2002-08-14 2005-06-29 Vertex Pharmaceuticals Incorporated Protein kinase inhibitors and uses thereof
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
US20050282814A1 (en) 2002-10-03 2005-12-22 Targegen, Inc. Vasculostatic agents and methods of use thereof
EP1549614A4 (en) * 2002-10-03 2008-04-16 Targegen Inc Vasculostatic agents and methods of use thereof
US20060167021A1 (en) 2002-10-04 2006-07-27 Caritas St. Elizabeth's Medical Center Of Boston, Inc. Inhibition of src for treatment of reperfusion injury related to revascularization
AU2003287178A1 (en) 2002-10-10 2004-05-04 Smithkline Beecham Corporation Chemical compounds
AU2003301662A1 (en) 2002-10-21 2004-05-13 Bristol-Myers Squibb Company Quinazolinones and derivatives thereof as factor xa inhibitors
WO2004037814A1 (en) 2002-10-25 2004-05-06 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
AU2003299600A1 (en) 2002-12-20 2004-07-29 Pharmacia Corpration The r-isomer of beta amino acid compounds as integrin receptor antagonists derivatives
EP1592421A1 (en) 2002-12-20 2005-11-09 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
ES2314271T3 (en) 2002-12-24 2009-03-16 Astrazeneca Ab THERAPEUTIC QUINAZOLINE DERIVATIVES.
US7144911B2 (en) 2002-12-31 2006-12-05 Deciphera Pharmaceuticals Llc Anti-inflammatory medicaments
ATE402152T1 (en) 2003-02-07 2008-08-15 Janssen Pharmaceutica Nv HIV-INHIBITING 1,2,4-TRIAZINE
NZ541902A (en) 2003-02-07 2008-12-24 Janssen Pharmaceutica Nv Pyrimidine derivatives for the prevention of HIV infection
WO2004071426A2 (en) 2003-02-11 2004-08-26 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.
HUE035379T2 (en) 2003-04-24 2018-05-02 Coopervision Int Holding Co Lp Hydrogel contact lenses and package systems and production methods for same
TWI378923B (en) 2003-08-15 2012-12-11 Novartis Ag Pyrimidine derivatives
MXPA06003054A (en) 2003-09-18 2006-05-31 Novartis Ag 2,4-di (phenylamino) pyrimidines useful in the treatment of proliferative disorders.
PL2210607T3 (en) 2003-09-26 2012-01-31 Exelixis Inc N-[3-fluoro-4-({6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxy]quinolin-4-yl}oxy)phenyl]-N'-(4-fluorophenyl)cyclopropane-1,1-dicarboxamide for the treatment of cancer
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
JP4932495B2 (en) 2004-01-23 2012-05-16 アムゲン インコーポレイテッド Compound and method of use
EP1809614B1 (en) 2004-04-08 2014-05-07 TargeGen, Inc. Benzotriazine inhibitors of kinases
WO2005108370A1 (en) 2004-04-16 2005-11-17 Ajinomoto Co., Inc. Benzene compounds
NZ588896A (en) 2004-08-25 2012-05-25 Targegen Inc Heterocyclic compounds and methods of use
US7210697B2 (en) * 2004-12-16 2007-05-01 Tricam International, Inc. Convertible handle
AU2006209183B2 (en) 2005-01-26 2009-11-19 Irm Llc Compounds and compositions as protein kinase inhibitors
GB0501999D0 (en) 2005-02-01 2005-03-09 Sentinel Oncology Ltd Pharmaceutical compounds
MX2007011500A (en) * 2005-03-16 2007-11-21 Targegen Inc 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
BRPI0606172A2 (en) 2005-06-08 2009-06-02 Targegen Inc methods and compositions for treating eye 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
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
RU2448959C2 (en) 2005-11-01 2012-04-27 Таргеджен, Инк. Bi-aryl-metha-pyrimidine kinase inhibitors
WO2007056075A2 (en) 2005-11-02 2007-05-18 Targegen, Inc. Six membered heteroaromatic inhibitors targeting resistant kinase mutations
WO2007127366A2 (en) 2006-04-25 2007-11-08 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
WO2009026345A1 (en) 2007-08-20 2009-02-26 Targegen Inc. Thiazolidinone 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

Also Published As

Publication number Publication date
US20050245524A1 (en) 2005-11-03
EP1809614A2 (en) 2007-07-25
RU2006139258A (en) 2008-05-20
HK1110578A1 (en) 2008-07-18
WO2005096784A3 (en) 2009-02-19
EP1809614B1 (en) 2014-05-07
NZ588139A (en) 2012-02-24
KR20070011458A (en) 2007-01-24
NZ551027A (en) 2011-01-28
AU2005231507A1 (en) 2005-10-20
AU2005231507B2 (en) 2012-03-01
IL178908A (en) 2015-09-24
CA2567574A1 (en) 2005-10-20
EP2543376A1 (en) 2013-01-09
US7456176B2 (en) 2008-11-25
US20110294796A1 (en) 2011-12-01
IL178908A0 (en) 2011-08-01
WO2005096784A2 (en) 2005-10-20
EP1809614A4 (en) 2010-02-17
US20090275569A1 (en) 2009-11-05
US8481536B2 (en) 2013-07-09

Similar Documents

Publication Publication Date Title
CA2567574C (en) Benzotriazine inhibitors of kinases
CA2628283C (en) Bi-aryl meta-pyrimidine inhibitors of kinases
US6407103B2 (en) Indeno [1,2-c] pyrazol-4-ones and their uses
CA2600531A1 (en) Pyrimidine compounds and methods of use
JP6175139B2 (en) New triazine compounds
WO2008008234A1 (en) 2-amino-5-substituted pyrimidine inhibitors
KR100891439B1 (en) Substituted benzimidazole compounds, and a pharmaceutical composition for the treatment of cancer comprising them
WO2017071607A1 (en) CRYSTAL FORM OF 4H-PYRAZOLO[1,5-α]BENZOIMIDAZOLE COMPOUND, PREPARATION METHOD THEREFOR AND INTERMEDIATE THEREOF
CN101426772B (en) Benzotriazine inhibitors of kinases
AU2012202928A1 (en) Benzotriazine inhibitors of kinases
CA3149846A1 (en) Quinoline derivatives as protein kinase inhibitors
CN111303135A (en) 4- (4-pyrazolyloxy) quinoline compound, preparation method, pharmaceutical composition and application thereof
Pandya et al. Synthesis and Anti Cervical Cancer Activity of Novel 5H-Thiochromeno [4, 3-d] pyrimidines
MX2008005810A (en) Bi-aryl meta-pyrimidine inhibitors of kinases

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20170407