AU2013204026B2 - Methods of using MEK inhibitors - Google Patents

Methods of using MEK inhibitors Download PDF

Info

Publication number
AU2013204026B2
AU2013204026B2 AU2013204026A AU2013204026A AU2013204026B2 AU 2013204026 B2 AU2013204026 B2 AU 2013204026B2 AU 2013204026 A AU2013204026 A AU 2013204026A AU 2013204026 A AU2013204026 A AU 2013204026A AU 2013204026 B2 AU2013204026 B2 AU 2013204026B2
Authority
AU
Australia
Prior art keywords
alkyl
optionally substituted
hydrogen
another embodiment
compound
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.)
Active
Application number
AU2013204026A
Other versions
AU2013204026A1 (en
Inventor
Peter Lamb
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.)
Exelixis Inc
Original Assignee
Exelixis Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2007334402A external-priority patent/AU2007334402B2/en
Application filed by Exelixis Inc filed Critical Exelixis Inc
Priority to AU2013204026A priority Critical patent/AU2013204026B2/en
Publication of AU2013204026A1 publication Critical patent/AU2013204026A1/en
Application granted granted Critical
Publication of AU2013204026B2 publication Critical patent/AU2013204026B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Abstract

This invention relates to methods of treating cancer with a compound that inhibits protein kinase enzymatic activity and the resultant modulation of cellular activities (such as proliferation, differentiation, programmed cell death, migration, chemoinvasion and metabolism) in combination with anticancer agents.

Description

METHODS OF USING MEK INHIBITORS BACKGROUND OF THE INVENTION
This is a divisional of Australian Patent Application No, 2007334402, the entire contents of which are incorporated herein by reference.
Field of the Invention [0001] This invention relates to methods of treating cancer with a compound that inhibits protein kinase enzymatic activity and the resultant modulation of cellular activities (such as proliferation, differentiation, programmed cell death, migration, chemoinvasion and metabolism) In combination with anticancer agents.
State of the Art [0002] Improvements in the specificity of agents used to treat various disease states such as cancer, metabolic, and inflammatory diseases is of considerable interest because of the therapeutic benefits which would be realized if the side effects associated with the administration of these agents could be reduced. Traditionally, dramatic improvements in the treatment of cancer are associated with identification of therapeutic agents acting through novel mechanisms.
[0003] Protein kinases are enzymes that catalyze the phosphorylation of proteins at the hydroxy groups of tyrosine, serine and threonine residues of proteins. The kinase complement of the human genome contains 518 putative protein kinase genes (Manning et al, Science, (2002), 298, 1912). The consequences of this activity include effects on cell differentiation, proliferation, transcription, translation, metabolism, cell cycle progression, apoptosis, metabolism, cytoskeletal rearrangement and movement; i. e., protein kinases mediate the majority of signal transduction in eukaryotic ceils. Furthermore, abnormal protein kinase activity' has been related to a host of disorders, ranging from relatively non-life threatening diseases such as psoriasis to cancer. Chromosomal mapping has revealed that over 200 kinases map to disease loci, including cancer, inflammatory and metabolic disease.
[0004] Tyrosine kinases can be categorized as receptor type or non-receptor type. Receptor-type tyrosine kinases have an extracellular, a transmernbrane, and an intracellular portion, while non-receptor type tyrosine kinases are wholly intracellular.
[0005] Receptor-type tyrosine kinases are comprised of a large number of transmembrane receptors with diverse biological activity, in fact, about 20 different subfamilies of receptor-type tyrosine kinases have been identified. One tyrosine kinase subfamily, designated the HER subfamily, is comprised of EGFR (HER1), HER2, HER3, and HER4. Ligands of this subfamily of receptors identified so far include epithelial growth factor, TGF-alpha, amphiregulin, HB-EGF, betacellulin and heregulin. Another subfamily of these receptor-type tyrosine kinases is the insulin subfamily, which includes INS-R, 1GF-IR, and IR-R. The PDGF subfamily includes the PDGF-alpha and -beta receptors, CSFIR, c-kit and FLK-II. Then there is the FLK family, which is comprised of the kinase insert domain receptor (KDR), fetal liver kinase-1 (FLK-1), fetal liver kinase-4 (FLK-4) and the fms-like tyrosine kinase-1 (Fit-1). The PDGF and FLK families are usually considered together due to the similarities of the two groups. For a detailed discussion of the receptor-type tyrosine kinases, see Plowman et al. (1994) DN&P 7(6): 334-339, which is hereby incorporated by reference.
[0006] The non-receptor type of tyrosine kinases is also comprised of numerous subfamilies, including Src, Frk, Btk, Csk, Abl, Syk/Zap70, Fes/Fps, Fak, Jak, and Ack. Each of these subfamilies is further sub-divided into varying receptors. For example, the Src subfamily is one of the largest and includes Src, Yes, Fyn, Lyn, Lck, Blk, Hck, Fgr, and Yrk. The Src subfamily of enzymes has been linked to oncogenesis. For a more detailed discussion of the non-receptor type of tyrosine kinases, see Bolen (1993) Oncogene, 8:2025-2031, which is hereby incorporated by reference.
[0007] Serine-threonine kinases play critical roles in intracellular signal transduction and include multiple families, such as.STE, CKI, AGO, CAMK, and CMGC. Important subfamilies include, the MAP kinases, p38, JNK and ERK, which modulate signal transduction resulting from such diverse stimuli as mitogenic, stress, proinfiammatory and antiapoptotic pathways. Members of the MAP kinase subfamily have been targeted for therapeutic intervention, including p38a, JNK isozymes and Raf.
[0008] Since protein kinases and their ligands play critical roles in various cellular activities, deregulation of protein kinase enzymatic activity can lead to altered cellular properties, such as uncontrolled cell growth associated with cancer. In addition to oncological indications, altered kinase signaling is implicated in numerous other pathological diseases, such as immunological disorders, metabolic and cardiovascular diseases, inflammatory diseases, and degenerative diseases. Therefore, both receptor and non-receptor protein kinases are attractive targets for small molecule drug discovery.
[0009] One particularly attractive goal for therapeutic use of kinase modulation relates to oncological indications. For example, modulation of protein kinase activity for the treatment of cancer has been demonstrated successfully with the FDA approval of Gleevec® (imatinib mesylate, produced by Novartis Pharmaceutical Corporation of East Hanover, NJ) for the treatment of Chronic Myeloid Leukemia (CML) and gastrointestinal stroma cancers. Gleevec is a selective Ab! kinase inhibitor.
[0010] Modulation (particularly inhibition) of cell proliferation and angiogenesis, two key cellular processes needed for tumor growth and survival (Matter A. Drug Disc Technol 2001 6, 1005-1024), is an attractive goal for development of small-molecule drugs. Anti-angiogenic therapy represents a potentially important approach for the treatment of solid tumors and other diseases associated with dysregulated vascularization, including ischemic coronary artery disease, diabetic retinopathy, psoriasis and rheumatoid arthritis. As well, cell antiproliferative agents are desirable to slow or stop the growth of tumors.
[0011] One particularly attractive target for small-molecule modulation, with respect to antiangiogenic and antiproliferative activity is MEK. Inhibition of MEK1 (MAPK/ERK Kinase) is a promising strategy to control the growth of tumors that are dependent on aberrant ERK/MAPK pathway signaling (Solit et al., 2006; Wellbrock et al., 2004), The MEK-ERK signal transduction cascade is a conserved pathway which regulates cell growth, proliferation, differentiation, and apoptosis in response to growth factors, cytokines, and hormones. This pathway operates downstream of Ras which is often upregulated or mutated in human tumors. It has been demonstrated that MEK is a critical effector of Ras function. The ERK/MAPK pathway is upregulated in 30% of all tumors and oncogenic activating mutations in K-Ras and B-Raf have been identified in 22% and 18% of all cancers respectively (Allen et al., 2003; Bamford S, 2004; Davies et al., 2002; Malumbres and Barbacid, 2003). A large portion of human cancers, including 66% (B-Raf) of malignant melanomas, 60% (K-Ras) and 4% (B-Raf) of pancreatic cancers, 50% of colorectal cancers (colon, in particular, K-Ras: 30%, B-Raf: 15%), 20% (K-Ras) of lung cancers, 27% (B-Raf) papillary and anaplastic thyroid cancer, and 10-20% (B-Raf) of endometriod ovarian cancers, harbor activating Ras and Raf mutations. Other cancers that may be treatable by inhibiting the ERK/MAPK pathway include kidney cancer (Rika Hoshino, et. al. Oncogene 21 January 1999, Volume 18, Number 3, Pages 813-822), breast cancer (Santen RJ, et. al. Steroid Biochem Mol Biol 2002, 80239), multiple myeloma Hu L et. al. Blood 2003,101, 3126), ovarian cancer Nicosia SV et. al. Hematol Oncol Clin North Am 2003, 17 927), and AML (Milella M et, al. Curr Pharm Des 2005, 11, 2779).
[0012] It has been shown that inhibition of the ERK pathway, and in particular inhibition of MEK. kinase activity, results in anti-metastatic and anti-angiogenic effects largely due to a reduction of cell-cell contact and motility as well as downregulation of vascular endothelial growth factor (VEGF) expression. Furthermore, expression of dominant negative MEK, or ERK. reduced the transforming ability of mutant Ras as seen in cell culture and in primary and metastatic growth of human tumor xenografts in vivo. Therefore, the MEK-ERK signal transduction pathway is an appropriate pathway to target for therapeutic intervention.
[0013] It is well established that combining treatments with different mechanisms of action often leads to enhanced anti-tumor activity as compared to single treatments administered alone. This is true for combinations of chemotherapies (e.g. Kyrgiou M. et. al. J Natl Cancer Inst 2006, 98, 1655) and combinations of antibodies and chemotherapy (e.g. Pasetto LM et. al. Anticancer Res 2006, 26, 3973.
Summary of the Invention [0014] The compositions of the invention are used to treat diseases associated with abnormal and or unregulated cellular activities. Disease states which can be treated by the methods and compositions provided herein include cancer. The invention is directed to methods of treating these diseases by administering a Compound of Formula I in combination with one or more treatment(s).
[0015] One aspect of the Invention is directed to a method of treating cancer which method comprises administering to a patient a therapeutically effective amount of a compound of Formula I:
I or a pharmaceutically acceptable salt or solvate, thereof; or administering a pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I and a pharmaceutically acceptable carrier, excipient, or diluent in combination with one or more treatment(s) selected from surgery, one or more chemotherapeutic agent(s). one or more of the hormone therapy(s), one or more of the antibody(s), hypothermia therapy, radioactive iodine therapy, and radiation wherein the Compound of Formula I is that where A, X, R1, R2, R3, R4, R5, R6, and R7 are as defined in Group A, Group B, Group C, or Group D:
Group A: A is arylene optionally substituted with one, two, three or four groups selected from R10, R12, R14, and R16 where Ri0, R12, R14 and R56 are independently hydrogen, alkyl, alkenyl, alkynyl, halo, haloalkoxy, hydroxy, alkoxy, amino, alkylamino, dialkylamino, haloalkyl, -NHS(0)2R8, -CN, -C(0)R8, -C(0)0R8, -C(0)NR*R8 and -NR8C(0)R8; X is alkyl, halo, haloalkyl, or haloalkoxy; R’, R2, R3, R4, R5 and R6 are independently hydrogen, halo, rsitro, -NR8R8, -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8Rr, -C(0)R8, -C(0)0R8, -C(0)NRsR8’, -NR8C(0)0R8’, -NR8C(0)NRs'R8”, -NR8C(0)0R8', -NR8C(0)R8,,-CH2N(R25)(NR25aR25b), -CH2NR25C(-NH)(NR25aR25b), -CH2NR25C(=NH)(N(R258)(N02), -CH2NR25C(=rNH)(N(R2Sa)(CN), -CH2NR25C(=NH)(R25), -CH2NR25C(NR25aR25b)-CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroarvl, or heterocycloalkyi, where the alkyl, alkenyl, alkynyl, cycloalkyi, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryi, -OR8, -NR8R8’, -NR8S(0)2R9, -CN, -S(0)mR9, -C(Q)R8, -C(0)0R8, -C(0)NRsR8’, -NR8C(0)NR8’R8”, ~NR8C(0)0R8’ and -NRsC(0)R8’; or one of R1 and R2 together with the carbon to wliich they are attached, R3 and R4 together with the carbon to which they are attached, and R5 and R6 together with the carbon to which they are attached form C(O) or C(=NOH); m is 0, 1, or 2; R7 is hydrogen, halo or alkyl; R8, Rs and Rs” are independently selected from hydrogen, hydroxy, optionally substituted alkoxy, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl; where the alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two three, four, or five groups independently selected from alkyl, halo, hydroxy, hydroxyalkyl, optionally substituted alkoxy, alkoxyalkyl, haloalkyl, carboxy, alkoxy carbonyl, alkenyloxycarbonyl, optionally substituted cycloalkyl, optionally substituted cycloalkyloxycarbonyl, optionally substituted aryl, optionally substituted aryloxy, optionally substituted aryloxycarbonyl, optionally substituted arylalkyl, optionally substituted arylalkyloxy, optionally substituted arylalkyloxycarbonyl, nitro, cyano, optionally substituted heterocycloalkyl, optionally substituted heteroaryl, -S(0)nR31 (where n is 0, 1, or 2 and R31 is optionally substituted alkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, or optionally substituted heteroaryl), -NR^SC^R348 (where R34 is hydrogen or alkyl and R34a is alkyl, alkenyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl), -S02NR35Rj5a (where R35 is hydrogen or alkyl and R35d is alkyl, alkenyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl), -NR32C(0)R32a (where R32 is hydrogen or alkyl and R32a is alkyl, alkenyl, alkoxy, or cycloalkyl), -NR30R30 (where R30 and R30 are independently hydrogen, alkyl, or hydroxyalkyl), and -C(0)NR33R',3a (where R33 is hydrogen or alkyl and R33a is alkyl, alkenyl, alkynyl, or cycioalkyl); R9 is alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl; where the alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, or five groups selected from halo, hydroxy, alkyl, haloalkyl, haioalkoxy, amino, alkylamino, and dialkylamino; R23 and R25b are independently hydrogen, alkyl, alkenyl, optionally sbustituted cycioalkyl, or optionally substituted aryl; and R25a is hydrogen, alkyl, or alkenyl;
Group B: A is heteroarylene optionally substituted with one, two, three, or four groups selected from R10, R12, RH,R16 and Ri9 where Ri0, R52, R,4and Ri6 are independently hydrogen, alkyl, alkenyl, alkynyl, halo, haioalkoxy, hydroxy, alkoxy, cyano, amino, alkylamino, dialkylamino, haloalkvl, alkylsulfonylamino, aikylcarbonyl, alkenyl carbonyl, alkoxvcarbonyl, aikenyioxycarbonyl, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, or alkylcarbonylamino; where R19 is hydrogen, alkyl, or alkenyl; and where each alkyl and alkenyl, either alone or as part of another group within R10, R12, R14, Rl<s, and R19 is independently optionally substituted with halo, hydroxy, or alkoxy; X is alkyl, halo, haloalkyl, or haloalkoxy; R1, R2, R3, R4, R5 and R6 are independently hydrogen, halo, nitro, -NR8R8, -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(0)0R8, -C(0)NRsR8’, -NR8C(0)0Rs', -NR8C(0)NR8’R8”, -NR8C(0)0R8’, -NR8C(0)R8’,-CH2N(R25)(NR25aR25b),-CH2NR25C(=NH)(NR25aR25b), -CH2NR25C(=NH)(N(R25a)(N02), -CH2NR25C(=NH)(N(R25a)(CN), -CH2NR25C(==NH)(R25), -CH2NR25C(NR258R25b)=CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkyl, where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycioalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, g optionally substituted arylalkyl, optionally substituted heteroaryl, -OR , -NR8R8’, -NRsS(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(G)NR8’R8” -NR8C(0)0R8’ and -NR8C(0)R8'; or one of R1 and R2 together with the carbon to which they are attached, R’ and R4 together with the carbon to which they are attached, and R5 and R6 together with the carbon to which they are attached form C(O) or C(=NOH); m is l or 2; R7 is hydrogen, halo or alkyl; and R8, R8 and R8 axe independently selected from hydrogen, hydroxy, optionally substituted alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl, where the alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two three, four, or five groups independently selected from alkyl, halo, hydroxy, hydroxyalkyl, optionally substituted alkoxy, alkoxyalkyl, haloalkyl, carboxy, carboxy ester, nitro, cyano, -S(0)„R31 (where n is 0,1, or 2 and R31 is optionally substituted alkyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, or optionally substituted heteroaryl), -NR36S(0)2R36a (where R36 is hydrogen, alkyl, or alkenyl and R36a is alkyl, alkenyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, or optionally substituted heteroaryl), -S(0)2NR3/R37a (where R'’7 is hydrogen, alkyl, or alkenyl and RJ?a is alkyl, alkenyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, or optionally substituted heteroaryl), optionally substituted cycloalkyl, optionally substituted heterocvcloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted aryloxy, optionally substituted arylaikyioxy, optionally substituted heteroaryl, -NHC(0)R32 (where R32 is alkyl, alkenyl, alkoxy, or cycloalkyl) and ~NR30R30’ (where R30 and R30’ are independently hydrogen, alkyl, or hydroxyalkyl), and -C(0)NHRJJ (where R33 is alkyl, alkenyl, alkynyl, or cycloalkyl);
GroupC: A is
(a) where R!0 is hydrogen, alkyl, alkenyl, alkynyl, halo, haloalkoxy, hydroxy, alkoxy, amino, alkylamino, dialkyl amino, haloalkyl, -NHS(0)2R8, -CN, -C(0)R8, -C(0)0R8, -C(0)NR®R8’ and -NR8C(0)R8’; R10a is hydrogen, alkyl, or alkenyl; Y1 is =CH- or =N-; X is alkyl, halo, haloalkyl, or haloalkoxy; R1, R2, R3, R4, R5 and R6 are independently hydrogen, halo, nitro, -NR8R8 , -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(0)0R8, -C(0)NR8R8', -NR8C(0)0Rg’, -NR8C(0)NR8’R8”, -NR8C(0)0R8’, -NR8C(0)R8, -CH2N(R25)(NR25aR25b), -CH2NR25C(=NH)(NR25aR25b), -CH2NR25C(=NH)(N(R25aXN02), -CH2NR25C(=NH)(N(R25a)(CN), -CH2NR25C(-NH)(R25), -CH2NR25C(NR25aR25b)=CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkvl, where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloaikyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloaikyl, optionally substituted aryl,
Q optionally substituted arylalkyl, optionally substituted heteroaryl, -OR , -NR8R8, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)NR8’Rs” -NR8C(0)0R8> and ~NR8C(0)Rr; or one of R! and R2 together with the carbon to which they are attached, R3 and R4 together with the carbon to which they axe attached, and R5 and R6 together with the carbon to which they are attached form C(O) or Cf^NOH); m is 1 or 2; R7 is hydrogen, halo or alkyl; and R8, R8 and R8 are independently selected from hydrogen, hydroxy, optionally substituted alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloaikyl, where the alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloaikyl are independently optionally substituted with one, two three, four, or five groups independently selected from alkyl, halo, hydroxy, hydroxyalkyl, optionally substituted alkoxy, alkoxyalkyl, haloalkyl, caxboxy, carboxy ester, nitro, cyano, -S(0)nRji (where n is 0,1, or 2 and R3! is optionally substituted alkyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heterocycloaikyl, or optionally substituted heteroaryl), -NR36S(0>2R36a (where R36 is hydrogen, alkyl, or alkenyl and R36a is alkyl, alkenyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heterocycloaikyl, or optionally substituted heteroaryl), -S(0)2NR37R37a (where R37 is hydrogen, alkyl, or alkenyl and R:?a is alkyl, alkenyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heterocycloaikyl, or optionally substituted heteroaryl), optionally substituted cycloalkyl, optionally substituted heterocycloaikyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted aryloxy, optionally substituted arylalkyloxy, optionally substituted heteroaryl, -NHC(0)R32 (where R32 is alkyl, alkenyl, alkoxy, or cycloalkyl) and ~NR30R30 (where R30 and R30 are independently hydrogen, alkvl, or hydroxyalkyl), and -C(G)NHR33 (where R33 is alkyl, alkenyl, alkynyl, or cycloalkyl); or
Group D: A is
(b)or
(c) R40 and R40a are independently hydrogen or alkyl; X is alkyl, halo, haloalkyl, or haloalkoxy; R1, R2, R3, R4, R5 and R6 are independently hydrogen, halo, nitro, -NRSR8, -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)0R8’, -NR8C(0)NRrR8”, -NRsC(0)0R8’, -NR8C(0)R8’,-CH2N(R25)(NR2SaR25b),-CH2NR25C(=NH)(NR25aR25b), -CH2NR25C(=NH)(N(R25a)(N02), -CH2NR25C<=NH)(N(R25a)(CN), -CH2NR25C(=NH)(R25), -CH2NR25C(NR25aR25b)~CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkyl, where the alkvl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, © optionally substituted arylalkyl, optionally substituted heteroaryl, -OR , -NR8R8’, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)NR8 R8”, ~NR8C(0)0R8’ and -NR8C(0)R8’; or one of R! and R2 together with the carbon to which they are attached, RJ and R4 together with the carbon to which they are attached, and R5 and R6 together with the carbon to which they are attached form C(O) or C(-NOH); m is 1 or 2; R7 is hydrogen, halo or alkyl; and 8 8' 8" R , R and R° are independently selected from hydrogen, hydroxy, optionally substituted alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl, where the alkyl, alkenyl, alkynyl, aryl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two three, four, or five groups independently selected from alkyl, halo, hydroxy, hydroxyalkyl, optionally substituted alkoxy, alkoxyalkyl, haloalkyl, carboxy, carboxy ester, nitro, cyano, -S(0)nR (where n is 0, 1, or 2 and R is optionally substituted alkyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, or optionally substituted heteroaryl), -NR36S(0)2R36a (where R36 is hydrogen, alkyl, or alkenyl and R36a is alkyl, alkenyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, or optionally substituted heteroaryl), -S(0)2NR37R37a (where R37 is hydrogen, alkyl, or alkenyl and R37a is alkyl, alkenyl, optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, or optionally substituted heteroaryl), optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted aryloxy, optionally substituted arylalkyloxy, optionally substituted heteroaryl, -NHC(0)R (where R is alkyl, alkenyl, alkoxy, or cycloalkyl) and -NR30R30 (where R30 and R30 are independently hydrogen, alkyl, or hydroxyalkyl), and -C(0)NHR33 (where R33 is alkyl, alkenyl, alkynyl, or cycloalkyl).
[0015a] Another aspect of the invention provides a method of treating breast cancer which method comprises administering to a patient in need thereof a therapeutically effective amount of the compound l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(2S)-piperidin-2-yl]azetidin-3-ol
or a pharmaceutically acceptable salt thereof in combination with a taxane.
[0015b] A further aspect of the invention provides a use of a therapeutically effective amount of the compound 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(2S)-piperidin-2-yl]azetidin-3-ol or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of breast cancer, wherein the compound is administered in combination with a taxane. DETAILED DESCRIPTION OF THE INVENTION Definitions for the Mek Compound [0016] The following terms have the indicated meanings throughout: [0017] The symbolmeans a single bond, "=" means a double bond, "=" means a triple bond, and means a single bond and optionally a double bond. When chemical structures are depicted or described, unless explicitly stated otherwise, all carbons are assumed to have hydrogen substitution to conform to a valence of four.
[0018] When chemical structures are depicted or described, unless explicitly stated otherwise, all carbons are assumed to have hydrogen substitution to conform to a valence of four. For example, in the structure on the left-hand side of the schematic below there are nine hydrogens implied. The nine hydrogens are depicted in the right-hand structure. Sometimes a particular atom in a structure is described in textual formula as having a hydrogen or hydrogens as substitution (expressly defined hydrogen), for example, -CH2CH2-. It is understood by one of ordinary skill in the art that the aforementioned descriptive techniques are common in the chemical arts to provide brevity and simplicity to description of otherwise complex structures.
[0019] If a group “R” is depicted as “floating” on a ring system, as for example in the formula:
then, unless otherwise defined, a substituent “R” may reside on any atom of the ring system, assuming replacement of a depicted, implied, or expressly defined hydrogen from one of the ring atoms, so long as a stable structure is formed.
[0020] If a group “R” is depicted as floating on a fused ring system, as for example in the formulae:
then, unless otherwise defined, a substituent “R” may reside on any atom of the fused ring system, assuming replacement of a depicted hydrogen (for example the -NH- in the formula above), implied hydrogen (for example as in the formula above, where the hydrogens are not shown but understood to be present), or expressly defined hydrogen (for example where in the formula above, “X” equals =CH-) from one of the ring atoms, so long as a stable structure is formed. In the example depicted, the “R” group may reside on either the 5-membered or the 6-membered ring of the fused ring system. In the formula depicted above, when y is 2 for example, then the two “R’s” may reside on any two atoms of the ring system, again assuming each replaces a depicted, implied, or expressly defined hydrogen on the ring.
[0021] When a group “R” is depicted as existing on a ring system containing saturated carbons, as for example in the formula:
where, in this example, “y” can be more than one, assuming each replaces a currently depicted, implied, or expressly defined hydrogen on the ring; then, unless otherwise defined, where the resulting structure is stable, two “R’s” may reside on the same carbon. A simple example is when R is a methyl group; there can exist a geminal dimethyl on a carbon of the depicted ring (an “annular” carbon). In another example, two R’s on the same carbon, including that carbon, may form a ring, thus creating a spirocyclic ring (a “spirocyclyl” group) structure with the depicted ring as for example in the formula:
[0Θ22] “Acyl” means a -C(0)R radical where R is optionally substituted alkyl, optionally substituted alkenyl, haloalkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heteroaryl, heteroaralkyl, heterocycloalkyl, or heterocycloalkylalkyl, as defined herein, e.g., acetyl, benzoyl, trifluoromethylcarbonyl, or 2-methoxyethylcarbonyl, and the like.
[0023] “Acylamino” means a -NRR’ group where R is acyl, as defined herein, and R’ is hydrogen or alkyl.
[0024] “Administration” and variants thereof (e.g., “administering” a compound) in reference to a compound of the invention means introducing the compound or a prodrug of the compound into the system of the animal in need of treatment. When a compound of the invention or prodrug thereof is provided in combination with one or more other active agents (e.g,, surgery, radiation, and chemotherapy, etc.), “administration” and its variants are each understood to include concurrent and sequential introduction of the compound or prodrug thereof and other agents.
[0025] “Alkenyl” means a means a linear monovalent hydrocarbon radical of one to six carbon atoms or a branched monovalent hydrocarbon radical of three to 6 carbon atoms which radical contains at least one double bond, e.g., ethenyl, propenyl, l-but-3-enyl, l-pent-3-enyl, l-hex-5-enyl and the like.
[0026] “Alkenylcarbonyl” means a -C(0)R group where R is alkenyl, as defined herein.
[0027] “Alkenyloxycarbonyl” means a -C(0)0R group where R is alkenyl, as defined herein.
[0028] “Alkoxy” means an -OR group where R is alky! group as defined herein. Examples include methoxy, ethoxy, propoxy, isopropoxy, and the like. Lower-alkoxy refers to groups containing one to six carbons.
[0029] “Alkoxyalkyl” means an alkyl group, as defined herein, substituted with at least one, preferably one, two, or three, alkoxy groups as defined herein. Representative examples include methoxymethyl and the like.
[0030] “Alkoxycarbonyl” means a ~C(0)OR group where R is alkyl as defined herein, [0031] “Alkoxycarbonylamino” means a -NR’R” group where R’ is hydrogen, alkyl, hydroxy, or alkoxy and R” is alkoxycarbonyl, as defined herein.
[0032] “Alkyl” means a linear saturated monovalent hydrocarbon radical of one to eight carbon atoms or a branched saturated monovalent hydrocarbon radical of three to eight carbon atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl (including all isomeric forms), or pentyl (including all isomeric forms), and the like.
[0033] “Alkylamino” means a -NHR radical where R is alkyl as defined herein, or an N-oxide derivative, or a protected derivative thereof, e.g., methylamino, ethylamino, «-propylamino. iso-propyl amino, «-butylamino, Ao-butylarnino, tert-butylamino, or methylamino-N-oxide, and the like.
[0034] “Alkylaminoalkyl” means an alkyl group substituted with one or two alkylamino groups, as defined herein.
[0035] “Alkylaminocarbonyl” means a ~C(0)R group where R is alkylamino, as defined herien.
[0036] “AlkyJcarbonyl” means a -C(0)R group where R is alkyl, as defined herein.
[0037] “Alkylcarbonylamino” means a -NRR’ group where R is hydrogen or alkyl as defined herein and R’ is alkylcarbonyl, as defined herein.
[0038] “Alkylcarbonyloxy” means an -GC(0)R group where R is alkyl, as defined herein.
[0039] “Alkylsulfonylamino” means a -NRS(0)2R’ group where R is hydrogen or alkyl as defined herein, and R’ is alkyl, as defined herein.
[0040] “Alkynyl” means a straight or branched hydrocarbon radical having from 2 to 8 carbon atoms and at least one triple bond and includes ethynyl, propynyl, butynyl, pentyn-2-yl and the like.
[0041] “Aminoalkyl” means an alkyl group substiuted with at least one amino group and in another embodiment, one, two or three amino groups.
[0042] “Aminocarbonyl” means a -C(0)NH2 group.
[0043] “Aryl” means a monovalent six- to fourteen-membered, mono- or bi-carbocyclic ring, wherein the monocyclic ring is aromatic and at least one of the rings in the bicyclic ring is aromatic. Unless stated otherwise, the valency of the group may be located on any atom of any ring within the radical, valency rules permitting. Representative examples include phenyl, naphthyl, and indanyl, and the like.
[0044] “Aryiene” means a divalent six- to fourteen-membered, mono- or bi-carbocyclic ring, wherein the monocyclic ring is aromatic and at least one of the rings in the bicyclic ring is aromatic. Representative examples include phenylene, naphthylene, and indanylene, and the like.
[0045] “ArylalkyP means an alkyl group, as defined herein, substituted with one or two aryl groups, as defined herein. Examples include benzyl, phenethyl, and the like.
[0046] “Carboxy ester” means a -C(0)0R group where R is lower alkyl, lower alkenyl, lower alkynyl, cycloalkyl, aryl or arylalkyl, each of which is defined herein. Representative examples include methoxycarbonyl, ethoxycarbonyl, and benzyloxycarbonyl, and the like.
[0047] “Cycloalkyl” means a monocyclic or fused bicyclic, saturated or partially unsaturated (but not aromatic), monovalent hydrocarbon radical of three to ten carbon ring atoms. Fused bicyclic hydrocarbon radical includes bridged ring systems. Unless stated otherwise, the valency of the group may be located on any atom of any ring within the radical, valency rules permitting. One or two ring carbon atoms may be replaced by a -C(O)-, -C(S)-, or -C(=NH)~ group. The term cycloalkyl includes, but is not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexyl, or cyclohex-3-enyl, and the like.
[0048] “Dialkylamino” means a -NRR’ radical where R and R’ are alkyl as defined herein, or an N-oxide derivative, or a protected derivative thereof, e.g., dimethyiamino, diethylamino, iV,A~methyIpropylamino or AOV-methylethylamino, and the like.
[0049] “Dialkylaminoalkyl” means an alkyl group substituted with one or two diaikylamino groups, as defined herein, [0050] “Dialkylaminocarbonyl” means a -C(0)R group where R is diaikylamino, as defined herien.
[0051] “Fused-polycyclic” or “fused ring system” means a polycyclic ring system that contains fused rings and, unless otherwise indicated, can contain bridged rings; that is, where two rings have more than one shared atom in their ring structures. In this application, fused-polycyclics and fused ring systems are not necessarily all aromatic ring systems. Typically, but not necessarily, fused-polycyclics share a vicinal set of atoms, for example naphthalene or 1,2,3,4-tetrahydro-naphthalene. A spiro ring system is not a fused-polycyclic by this definition, but fused polycyclic ring systems of the invention may themselves have spiro rings attached thereto via a single ring atom of the fused-polycyclic. In some examples, as appreciated by one of ordinary skill in the art, two adjacent groups on an aromatic system may be fused together to form a ring structure. The fused ring structure may contain heteroatoms and may be optionally substituted with one or more groups. It should additionally be noted that saturated carbons of such fused groups (i.e. saturated ring structures) can contain two substitution groups.
[0052] “Haloalkoxy” means an -OR’ group where R’ is haloalkyl as defined herein, e.g., trifluoromethoxy or 2,2,2-trifluoroethoxy, and the like.
[0053] “Halogen” or “halo” means fluoro, chloro, bromo and iodo.
[0054] “Haloalkyl” means an alkyl group, as defined herein, that is substituted with one or more halogens, preferably one to five halo atoms. Representative examples include trifiuoromethyl, difluoromethyl, l-chloro-2-fluoro-ethyl, and the like.
[0055] “Heteroaryl” means a monocyclic, fused bicyclic, or fused tricyclic, monovalent radical of 5 to 14 ring atoms containing one or more, preferably one, two, three, or four ring heteroatoms independently selected from -Ο-, -S(0)n- (n is 0, 1, or 2), -N-, -N(RX)-, and the remaining ring atoms being carbon, wherein the ring comprising a monocyclic radical is aromatic and wherein at least one of the fused rings comprising a bicyclic or tricyclic radical is aromatic. One or two ring carbon atoms of any nonaromatic rings comprising a bicyclic or tricyclic radical may be replaced by a -C(0)~, -C(S)-, or -C(=NH)- group. Rx is hydrogen, alkyl, hydroxy, alkoxy, acyl, or alkylsulfonyl. Unless stated otherwise, the valency may be located on any atom of any ring of the heteroaryi group, valency rules permitting. In particular, when the point of valency is located on the nitrogen, R* is absent. The term heteroaryl includes, but is not limited to, 1,2,4-triazolyl, 1,3,5-triazolyl, phthalimidyl, pyridinyl, pyrrolyl, imidazoiyl, thienyl, furanyl, indolyl, 2,3-dihydro-1 /f-indolyl (including, for example, 2,3-dihydro-1 #-indol-2-yl or 2,3-dihydro- lff-indol-S-yl, and the like), isoindolyl, indolinyl, isoindolinyl, benzimidazolyl, benzodioxol-4-yl, benzofuranyl, cinnolinyl, indolizinyl, naphthyridin-3-yl, phthalazin-3-yl, phthalazin-4-yl, pteridinyl, purinyl, quinazolinyl, quinoxalinyl, tetrazoyl, pyrazolyl, pyrazinyl, pyrimidinyl, pyridazinyl, oxazolyl, isooxazolyl, oxadiazolyl, benzoxazolyl, quinolinyl, isoquinolinyl, tetrahydroisoquinolinyl (including, for example, tetrahydroisoquinolin-4-yl or tetrahydroisoquinolin-6-yl, and the like), pyrrolo[3,2-cjpyridinyl (including, for example, pyrrolo[3,2-c]pyridin-2-yl or pyrrolo[3,2-c]pyridin-7-yl, and the like), benzopyranyl, thiazolyl, isothiazolyl, thiadiazolyl, benzothiazolyl, benzothienyl, and the derivatives thereof, or N-oxide or a protected derivative thereof.
[0056] “Heteroarylene” means a monocyclic, fused bicyclic, or fused tricyclic, divalent radical of 5 to 14 ring atoms containing one or more, preferably one, two, three, or four ring heteroatoms independently selected from -0-, -S(0)n- (n is 0, 1, or 2), -N-, -N(R!9)~, and the remaining ring atoms being carbon, wherein the ring comprising a monocyclic radical is aromatic and wherein at least one of the fused rings comprising a bicyclic or tricyclic radical is aromatic. One or two ring carbon atoms of any nonaromatic rings comprising a bicyclic or tricyclic radical may be replaced by a -C(O)-, -C(S)-, or -C(=NH> group. Ri9 is hydrogen, alkyl, or alkenyl. Unless stated otherwise, the valencies may be located on any atom of any ring of the heteroarylene group, valency rules permitting. In particular, when the point of valency is located on the nitrogen, Rx is absent. The term heteroaryi includes, but is not limited to, thien-diyl, benzo[if]isoxazol-diyL benzo[<i]isothiazo 1 -diyl, l/T-indazol-diyl (optionally substituted at the N1 position with R19), benzo[i/]oxazol-diyi, benzo[i/]thiazol-diyl, l//-benzo[iflimidazol-diyl (optionally substituted at the N1 position with R19), l//~benzo[d][l,2,3]triazol-diyl (optionally substituted at the N1 position with R19), imidazo[ 1,2-a]pyridin-diyl, cinnolin-diyl, quinolin-diyl, pyridin-diyl, 1 -oxido-pyridin-diyl, [ 1,2,4]triazolo[4,3-a]pyridin-diyl, and 2,3-dihydroimidazo[ 1,2-a]pyridin-diyl, and the like.
[0057] “Heterocycloalkyl” means a saturated or partially unsaturated (but not aromatic) monovalent monocyclic group of 3 to 8 ring atoms or a saturated or partially unsaturated (but not aromatic) monovalent fused bicyclic group of 5 to 12 ring atoms in which one or more (and in another embodiment, one, two, three, or four) ring heteroatoms independently selected from O, S(0)n (n is 0, 1, or 2), N, N(Ry) (where Ry is hydrogen, alkyl, hydroxy, alkoxy, acyl, or alkylsulfonyl), the remaining ring atoms being carbon. One or two ring carbon atoms may be replaced by a -C(O)-, -C(S)-, or -C(=NH)- group. Fused bicyclic radical includes bridged ring systems. Unless otherwise stated, the valency of the group may be located on any atom of any ring within the radical, valency rules permitting. When the point of valency is located on a nitrogen atom, Ry is absent. The term heterocycloalkyl includes, but is not limited to, azetidinyl, pyrrolidinyl, 2-oxopyrrolidiny 1, 2.5-dihydro-l//-pyrrolyl, piperidinyl, 4-piperidonyl, morpholinyl, piperazinyl, 2-oxopiperazinyl, tetrahydropyranyl, 2-oxopiperidinyl, thiomorpholinyl, thiamorpholinyl, perhydroazepinyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, dihydropyridinyl, tetrahydropyridinyl, oxazolinyl, oxazolidinyl, isoxazolidinyl, thiazolinyl, thiazolidinyl, quinuclidinyl, isothiazolidinyl, octahydroindolyl, octahydroisoindolyl, decahydroisoquinolyl, tetrahydrofuryl, and tetrahydropyranyl, and the derivatives thereof and N-oxide or a protected derivative thereof.
[0058] “Hydroxyalkyl” means an alkyl, as defined herein, substituted with at least one, preferably one, two, or three, hydroxy group(s), provided that if two hydroxy groups are present they are not both on the same carbon atom. Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, 3-hydroxypropyI, l-(hydroxymethyl)-2-methylpropyl, 2-hydroxybutyl, 3-hydroxybutyl, 4-hydroxy butyl, 2,3 -dihy droxypropy 1, 1 -(hydroxymethyl)-2 -hydroxyethyl, 2,3 -dihydroxybutyl, 3,4-dihydroxybutyl and 2-(hydroxymethyl)-3-hydroxypropyl, preferably 2-hydroxyethyl, 2,3-dihydroxypropy 1, and l-(hydroxymethyl)-2-hydroxyethyl, and the like.
[0059] “Hydroxy-amino*’ means a -NH(OH) group.
[0060] “Optional” or “optionally” means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances in which it does not. One of ordinary skill in the art would understand that with respect to any molecule described as containing one or more optional substituents, only sterically practical and/or synthetically feasible compounds are meant to be included. “Optionally substituted ” refers to all subsequent modifiers in a term. So, for example, in the term “optionally substituted arylCj.g alkyl,” both the “Cq.g alkyl” portion and the “aryl” portion of the molecule may or may not be substituted. A list of exemplary optional substitutions is presented below in the definition of “substituted.” [0061j “Optionally substituted alkoxy” means an -OR radical where R is optionally substituted alkyl as defined herein. Representative examples include -OCH2CH2OCH3, -OCH2CH2OH, -OCH2CH(NH2)CH3, and the like.
[0062] “Optionally substituted alkyl” means an alkyl radical, as defined herein, optionally substituted with one or more group(s) (and in another embodiment one, two, three, four, or five groups) independently selected from alkylcarbonyl, alkenylcarbonyl, cycloalkyIcarbonyl, alkylcarbonyloxy, alkenylcarbonyloxy, amino, alkylamino, dialkylamino, aminocarbonyl, alkylaminocarbonyl, dialkylaminocarbonyl, cyano, cyanoalkylaminocarbonyl, alkoxy, alkenyloxy, halo, hydroxy, hydroxyalkoxv, carboxy, alkylcarbonylamino, alkylcarbonyloxy, -S(0)o-2-alkyl, -S(0)o-2-alkenyl, aminosulfonyl, alkylaminosulfonyl, dialkvlaminosulfonyl, -NRcS(0)2-aikyl (where Rc is hydrogen, alkyl, optionally substituted alkenyl, optionally substituted alkynyl, hydroxy, alkoxy, alkenyloxy, or cyanoalkyl), alky laminocarbonyloxy, dialkylaminocarbonyloxy, alkylaminoalkyloxy, dialkylaminoalkyloxy, alkoxycarbonyl, alkenyloxycarbonyl, alkoxycarbonylamino, alkylaminocarbony lamino, dialkylaminocarbonylamino, alkoxyalkyloxy, and -C(0)NRaRb (where Ra and Rb are independently hydrogen, alkyl, optionally substituted alkenyl, optionally substituted alkynyl, hydroxy, alkoxy, alkenyloxy, or cyanoalkyl).
[0063] “Optionally substituted aryl” means an aryl group, as defined herein, which is optionally substituted with one, two, three, four, of five groups selected from halo, haloalkyl, haloalkoxy, hydroxy, alkyl, alkenyl, alkynyl, alkoxy, carboxy, carboxy ester, amino, alkylamino, dialkylamino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted heteroaryl, -C(0)NR’R” (where R’ is hydrogen or alkyl and R” is hydrogen, alkyl, aryl, heteroaryS, or heterocycloalkyl), -NR’C(0)R” (where R’ is hydrogen or alkyl and R” is alkyl, aryl, heteroaryl, or heterocycloalkyl), and -NHS(0)2R’ (where R’ is alkyl, aryl, or heteroaryl).
[0064] “Optionally substituted arylalkyl means an alkyl group substituted with one or two optionally substituted aryl group(s) as defined herein.
[0065] “Optionally substituted arylalkyloxy” means an -OR group where R is optionally substituted arylalkyl, as defined herein.
[0066] “Optionally substituted arylalkyloxycarbonyl” means a -C(0)R group where R is optionally substituted arylalkyloxy, as defined herein.
[0067] “Optionally substituted aryloxy” means an -OR group where R is optionally substituted aryl, as defined herein.
[0068] “Optionally substituted aryloxycarbonyl” means a -C(G)R group where R is optionally substituted aryloxy as defined herein.
[0069] “Optionally substituted cycloalkyl” means a cycloalkyl radical, as defined herein, that is optionally substituted with one, two, three, or four groups independently selected from alkyl, alkenyl, alkvnyl, alkoxy, halo, haloalkyl, haloalkoxy, oxo, hydroxy, eyano, nitro, amino, mono(CrC6)alkyiamino, dialkylamino, haloalkyl, haloalkoxy, aminoalkyl, alkylaminoalky! dialkylaminoalkyl, carboxy, carboxy ester, cycloalkyl, hydroxyalkyl, -C(0)NRsR” (where R’ is hydrogen, alkyl, hydroxy, or alkoxy and R” is hydrogen, alkyl, aryl, heteroaryl, or heterocycloalkyl), optionally substituted heterocycloalkyl, optionally substituted heteroaryl, ~NR’C(0)R” (where R’ is hydrogen or alkyl and R” is alkyl, aryl, heteroaryl, or heterocycloalkyl), and -NHS(0)2R’ (where R’ is alkyl, aryl, or hetercyclyi).
[0070] “Optionally substituted cycloalkyloxycarbonyl” means a -C(0)OR group where R is optionally substituted cycloalkyl as defined herein.
[0071] “Optionally substituted heteroaryl” means a heteroaryl group, as defined herein, optionally substituted with one, two, three, four, or five groups selected from halo, haloalkyl, haloalkoxy, alkyl, alkenyl, alkynyl, alkoxy, hydroxy, oxo (valency rules permitting), carboxy, carboxy ester, amino, alkylamino, dialkylamino, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, heteroaryl, optionally substituted aryl, -C(Q)NR’R” (where Rs is hydrogen or alkyl and R” Is hydrogen, alkyl, aryl, heteroaryl, or heterocycloalkyl), -NR’C(0)R” (where R5 is hydrogen or alkyl and R” is alkyl, aryl, heteroaryl, or heterocycloalkyl), and -NHS(0)2R’ (where R’ is alkyl, aryl, or heteroaryl).
[0072] “Optionally substituted heterocycloalkyl” means a heterocycloalkyl ring, as defined herein, optionally substituted with one, two, three, four, or five groups selected from halo, haloalkyl, haloalkoxy, hydroxy, oxo, alkyl, alkenyl, alkynyi, alkoxy, optionally substituted cycloalkyl, heterocycloalkyl, optionally substituted aryl, optionally substituted heteroaryl, alkylaminoalkyl, dialkylaminoalkyl, carboxy, alkoxycarbonyl, aryloxycarbonyl, arylalky loxycarbonyl, cycloalkyloxycarbonyl, cycloalkylalkyloxycarbonyl, -C(0)NR’R” (where R’. is hydrogen or alkyl and R” is hydrogen, alkyl, aryl, heteroaryl, or heterocycloalkyl), -NR’C(0)R” (where R’ is hydrogen or alkyl and R” is alkyl, aryl, heteroaryl, or heterocycloalkyl), amino, alkylamino, dialkylamino, and ~NHS(0)?R’ (where R’ is alkyl, aryl, or heteroaryl).
[0073] “Saturated bridged ring system” refers to a bicyclic or polycyclic ring system that is not aromatic. Such a system may contain isolated or conjugated unsaturation, but not aromatic or heteroaromatic rings in its core structure (but may have aromatic substitution thereon). For example, hexahydro-furo[3,2-b]furan, 2,3,3a,4,7,7a-hexahydro-1 H-indene, 7-aza-bicyclo[2.2.1 ]heptane, and l,2,3,4,4a,5,S,8a-octahydro-naphthalene are all included in the class “saturated bridged ring system.” [0074] “Spiro”, “Spirocyclyl” or “spiro ring” refers to a ring originating from a particular annular carbon of another ring. For example, as depicted below, a ring atom of a saturated bridged ring system (rings B and B’), but not a bridgehead atom, can be a shared atom between the saturated bridged ring system and a spirocyclyl (ring A) attached thereto.
[0075] “Yield” for each of the reactions described herein is expressed as a percentage of the theoretical yield.
Definitions for the Compound of formula 100 [0076] The terms used to describe the scope of formula 100 are defined in WO 2004/006846 (US Nat’l Stage Application Serial No. 10/522,004) which is herein incorporated by reference. For example “optionally substituted alkyl” for formula 100 has the meaning given in WO 2004/006846 (US Nat’l Stage Application Serial No. 10/522,004). Whenever a compound of formula 100 is described in this application, whether by structure or by use of the term “formula 100,” the terms used to describe that compound are defined by WO 2004/006846 (US Nat’l Stage Application Serial No. 10/522,004).
Definitions for the Compound of formula 101 [0077] The terms used to describe the scope of formula 101 are defined in WO 2005/112932 (US Nat’l Stage Application Serial No. 11/568,789) which is herein incorporated by reference. For example “optionally substituted heterocyclyl” for formula 101 has the meaning given in WO 2005/112932 (US Nat’l Stage Application Serial No. 11/568,789). Whenever a compound of formula 101 is described in this application, whether by structure or by use of the term “formula 101,” the terms used to describe that compound are defined by WO 2005/112932 (US Nat’l Stage Application Serial No. 11/568,789).
Definitions for the Compound of formula A-B-C
[0078] The terms used to describe the scope of formula A-B-C are defined in WO 2005/030140 (US Nat’l Stage Application Serial No. 10/573,336) which is herein incorporated by reference. For example “optionally substituted heterocyclyl” for formula A-B-C has the meaning given in WO 2005/030140 (US Nat’l Stage Application Serial No. 10/573,336). Whenever a compound of formula A-B-C is described in this application, whether by structure or by use of the term “formula A-B-C,” the terms used to describe that compound are defined by WO 2005/030140 (US Nat’l Stage Application Serial No. 10/573,336),
Definitions for the Compound of formula 103 [ΘΘ79] The terms used to describe the scope of formula 103 are defined in WO 2006/014325 (US Nat’l Stage Application Serial No. 11/571,140) which is herein incorporated by reference. For example “optionally substituted heterocyclyl” for formula 103 has the meaning given in WO 2006/014325 (US Nat’l Stage Application Serial No. 11/571,140). Whenever a compound of formula 103 is described in this application, whether by structure or by use of the term “formula 103,” the terms used to describe that compound are defined by WO 2006/014325 (US Nat’l Stage Application Serial No. 11/571.140).
Definitions for the Compound of formula 105 [0080] The terms used to describe the scope of formula 105 are defined in WO 2006/074057 (US Nat’l Stage Application Serial No. 11/722,719) which is herein incorporated by reference. For example “optionally substituted heterocyclyl” for formula 105 has the meaning given in WO 2006/074057 (US Nat’l Stage Application
Serial No. 11/722,719), Whenever a compound of formula 105 is described in this application, whether by structure or by use of the term “formula 105,” the terms used to describe that compound are defined by WO 2006/074057 (US Nat’l Stage Application Serial No. 11/722,719).
Definitions for the Compound of formula 107 [0081] The terms used to describe the scope of formula 107 are defined in WO 2004/050681 (US Nat’l Stage Application Serial No. 10/533,555) which is herein incorporated by reference. For example “optionally substituted aryl” for formula 107 has the meaning given in WO 2004/050681 (US Nat’l Stage Application Serial No. 10/533,555).Whenever a compound of formula 107 is described in this application, whether by structure or by use of the term “formula 107,” the terms used to describe that compound are defined by WO 2004/050681 (US Nat’l Stage Application Serial No. 10/533,555).
Definitions for the Compound of formula 108 [0082] The terms used to describe the scope of formula 108 are defined in WO 2005/117909 (US Nat’l Stage Application Serial No. 11/568,173) which is herein incorporated by reference. For example “optionally substituted aryl” for formula 108 has the meaning given in WO 2005/117909 (US Nat’l Stage Application Serial No. 11/568,173). Whenever a compound of formula 108 is described in this application, whether by structure or by use of the term “formula 108,” the terms used to describe that compound are defined by WO 2005/117909 (US Nat’l Stage Application Serial No. 11/568,173).
Definitions for the Compound of formula 109 [0083] The terms used to describe the scope of formula 109 are defined in WO 2006/071819 (US Nat’l Stage Application Serial No. 11/722,291 which is herein incorporated by reference. For example “optionally substituted aryl” for formula 109 has the meaning given in WO 2006/071819 (US Nat 1 Stage Application Serial No. 11/722,291. Whenever a compound of formula 109 is described in this application, whether by structure or by use of the term “formula 109,” the terms used to describe that compound are defined by WO 2006/071819 (US Nat’l Stage Application Serial No. 11/722,291.
Other Definitions [0084] “AKT inhibitor” includes, for example, LY294002, PKC412, and compounds described in WO 2006/071819 and WO05/117909.
[0085] “Alkylating agent(s)” includes, for example, one or more of the following: Chlorambucil, Chlormethine, Cyclophosphamide, Ifosfamide, Melphalan, Carmustine, Streptozocin, Fotemustine, Lomustine, Streptozocin, Carboplatin, Cisplatin, Oxaliplatin, BBR3464, Busulfan, Dacarbazine, Mechlorethamine, Procarbazine, Temozolomide, ThioTEPA, and Uramustine.
[0086] “Antibody(s)” includes, for example, one or more of the following: IGF1R. antibody (including, for example, “IGF-IR A12 MoAb, 19D12, h7C10 and CP-751871), Alemtuzumab, Bevacizumab (Avastin®), Cetuximab (Erbitux®), Gemtuzumab, Gemtuzumab ozogamicin, Ibritumomab (tiuxetan), Panitumumab, Rituximab, Tositumomab, and Trastuzumab (Herceptin®).
[0087] “Antimetabolite(s)” include, for example, methotrexate, Pemetrexed, Raltitrexed, Cladribine, Clofarabine, Fludarabine, Mercaptopurine, Thioguanine, Capecitabine, Cytarabine, fluorouracil (administered with or without leucovorin or folinic acid), and Gemcitabine.
[0088] “Antimicrotubule agent(s)” includes, for example, Vincristine,Vinblastine, Vinorelbine, Vinflunine, and Vindesine.
[0089] “Aromatase inhibitor(s)” includes, for example, one or more of the following: Aminoglutethimide, Anastrozole (Arimidex®), Letrozole (Femara®), Exemestane (Aromasin®), and Formestane (Lentaron®).
[0090] “Cancer” refers to cellular-proliferative disease states, including but not limited to: Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hanlartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma); Genitourinary tract: kidney (adenocarcinoma, Wiim's tumor [nephroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis defomians), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma], fallopian tubes (carcinoma); Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin’s lymphoma [malignant lymphoma]; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis; Adrenal Glands: neuroblastoma; and breast cancer. Thus, the term “cancerous cell” as provided herein, includes a cell afflicted by any one of the above-identified conditions.
[0091] “cMET inhibitor” includes, for example, compounds described in WO06/108059, WO 2006/014325, and WO 2005/030140. 10092] “EGFR inhibitor”includes, for example, one or more of the following:
Lapatinib (Tykerb®), gefitinib (Iressa®), erlotinib (Tarceva®), Zactima (ZD6474), AEE788 and HKI-272, EKB-569, CI1033, and compounds described in WO 2004/006846 and WO 2004/050681. {0093] “ErbB2 inhibitor” includes, for example, Lapatinib (GW572016), and PKI-166. |0094] “Hormone therapy” or “hormonal therapy” includes, for example, treatment with one or more of the following: steroids (e.g. dexamethasone), finasteride, tamoxifen, and an aromatase inhibitor. {0095] “HSP90 inhibitors)” includes, for example, 17-AAG, 17-DMAG, Geldanamycin, 5-(2,4-dihydroxy-5-isopropylphenyl)-iV-ethyi-4-(4-(morpholinomethyl)phenvl)isoxazole-3-carboxamide [NVP-AUY922 (VER 52296)], 6-chlorO“9”((4-methoxy-3.5-dimethylpyridm-2-yl)methyl)-9//-purin~2-amine (CNF2024, also named BIIB021), compounds disclosed in W02004072051 (which is herein incorporated by reference), compounds disclosed in W02005028434 (which is herein incorporated by reference), compounds disclosed in W02007035620 (which is herein incorporated by reference) and compounds disclosed in W02006091963 (which is herein incorporated by reference).
[0096] “Hypothermia therapy” is a type of treatment in which body tissue is exposed to high temperatures to damage and kill cancer cells or to make cancer ceils more sensitive to the effects of radiation and certain anticancer drugs. {0097] “IGF1R inhibitor(s)” include, for example, Tyrphostin AG 1024 and compounds described in W006/074057. {0098] “Kinase-dependent diseases or conditions" refer to pathologic conditions that depend on the activity of one or more protein kinases. Kinases either directly or indirectly participate in the signal transduction pathways of a variety of cellular activities including proliferation, adhesion, migration, differentiation and invasion. Diseases associated with kinase activities include tumor growth, the pathologic neovascularization that supports solid tumor growth, and associated with other diseases where excessive local vascularization is involved such as ocular diseases (diabetic retinopathy, age-related macular degeneration, and the like) and inflammation (psoriasis, rheumatoid arthritis, and the like). {0099] While not wishing to be bound to theory, phosphatases can also play a role in “kinase-dependent diseases or conditions” as cognates of kinases; that is, kinases phosphorylate and phosphatases dephosphoryiate, for example protein substrates. Therefore compounds of the invention, while modulating kinase activity as described herein, may also modulate, either directly or indirectly, phosphatase activity. This additional modulation, if present, may be synergistic (or not) to activity of compounds of the invention toward a related or otherwise interdependent kinase or kinase family. In any case, as stated previously, the compounds of the invention are useful for treating diseases characterized in part by abnormal levels of cell proliferation (i.e. tumor growth), programmed cell death (apoptosis), cell migration and invasion and angiogenesis associated with tumor growth.
[00100] “Metabolite” refers to the break-down or end product of a compound or its salt produced by metabolism or biotransformation in the animal or human body; for example, biotransformation to a more polar molecule such as by oxidation, reduction, or hydrolysis, or to a conjugate (see Goodman and Gilman, "The Pharmacological Basis of Therapeutics" B.sup.th Ed., Pergamon Press, Gilman et al. (eds), 1990 for a discussion of biotransformation). As used herein, the metabolite of a compound of the invention or its salt may be the biologically active form of the compound in the body. In one example, a prodrug may be used such that the biologically active form, a metabolite, is released in vivo. In another example, a biologically active metabolite is discovered serendipitously, that is, no prodrug design per se was undertaken. An assay for activity of a metabolite of a compound of the present invention is known to one of skill in the art in light of the present disclosure.
[00101] “Patient” for the purposes of the present invention includes humans and other animals, particularly mammals, and other organisms. Thus the methods are applicable to both human therapy and veterinary applications. In an embodiment the patient is a mammal, and in another embodiment the patient is human.
[00102] A “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in Remington’s Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985, which is incorporated herein by reference or S. M.
Berge, et al., “Pharmaceutical Salts,” J. Pharm. Sci., 1977;66:1-19 both of which are incorporated herein by reference.
[00103] Examples of pharmaceutically acceptable acid addition salts include those formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; as well as organic acids such as acetic acid, trifluoroacetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, 3-(4-hydroxybenzoyl)benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, glucoheptonic acid, 4,4’-methylenebis-(3-hydroxy-2-ene- 1-carboxylic acid), 3-phenylpropionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, p-toluenesulfonic acid, and salicylic acid and the like.
[00104] Examples of a pharmaceutically acceptable base addition salts include those formed when an acidic proton present in the parent compound is replaced by a metal ion, such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferable salts are the ammonium, potassium, sodium, calcium, and magnesium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins. Examples of organic bases include isopropylamine, trimethylamine, dieihylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, tromethamine, jV-methylglucamine, polyamine resins, and the like. Exemplary organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
[00105] “Platin(s),5’ and “platin-containing agent(s)” include, for example, cisplatin, carboplatin, and oxaliplatin.
[00106] “Prodrug” refers to compounds that are transformed (typically rapidly) in vivo to yield the parent compound of the above formulae, for example, by hydrolysis in blood. Common examples include, but are not limited to, ester and amide forms ot a compound having an active form bearing a carboxylic add moiety. Examples of pharmaceutically acceptable esters of the compounds of this invention include, but are not limited to, alkyl esters (for example with between about one and about six carbons) the alkyl group is a straight or branched chain. Acceptable esters also include cycloalkyl esters and arylalkyl esters such as, but not limited to benzyl. Examples of pharmaceutically acceptable amides of the compounds of this invention include, but are not limited to, primary amides, and secondary and tertiary alkyl amides (for example with between about one and about six carbons). Amides and esters of the compounds of the present invention may be prepared according to conventional methods. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, “Pro-drugs as Novel Delivery Systems,” Vol 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference for all purposes.
[001071 “Raf inhibitors)” include, for example, sorafenib and compounds described in WO 2005/112932.
[00108] “Rapamycin analogue(s)” include for example, CCI-779, AP23573, RAD001, TAFA93, and compounds described in WO 2004/101583 and US 7,160,867 which are each incorporated herein by reference in their entireties.
[00109] “Receptor Tyrosine Kinase inhibitor(s)” includes, for example, inhibitors of AKT, EGFR, ErbB2, IGF1R, Met, Raf, and VEGFR2. Examples of receptor tyrosine kinase inhibitors can be found in WO 2006/108059 (US Nat l Stage Application Serial No. 11/910,720), WO 2006/074057 (US Nat’i Stage Application Serial No. 11/722,719), WO 2006/071819 (US Nat’i Stage Application Serial No. 11/722,291), WO 2006/014325 (US Nat’S Stage Application Serial No. 11/571,140), WO 2005/117909 (US Nat’i Stage Application Serial No. 11/568,173), WO 2005/030140 (US Nat’i Stage Application Serial No. 10/573,336), WO 2004/050681 US Nat’i Stage Application Serial No. 10/533,555), WO 2005/112932 (US Nat’i Stage Application Serial No, 11/568,789), and WO 2004/006846 (US Nat’i Stage Application Serial No. 10/522,004), each of which is incorporated herein by reference for all purposes. In particular, the applications cited in this paragraph are incorporated for the purpose of providing specific examples and generic embodiments (and the definitions associated with the terms used in the embodiments) of compounds that are useful in the practice of the invention. These references also describe in vitro assays useful in the practice of this invention.
[00110] “Taxane(s)” includes, for example, one or more of the following: Paclitaxel (Taxol®) and Docetaxel (Taxotere®).
[00111] “Therapeutically effective amount” is an amount of a compound of the invention, that when administered to a patient, ameliorates a symptom of the disease. The amount of a compound of the invention which constitutes a “therapeutically effective amount” will vary depending on the compound, the disease state and its severity, the age of the patient to be treated, and the like. The therapeutically effective amount can be determined routinely by one of ordinary' skill in the art having regard to their knowledge and to this disclosure.
[00112] “Topoisomerase inhibitor” includes, for example, one or more of the following: amsacrine, camptothecin, etoposide, etoposide phosphate, exatecan, irinotecan, lurtotecan, and teniposide, and topotecan.
[00113] “Treating” or “treatment” of a disease, disorder, or syndrome, as used herein, includes (i) preventing the disease, disorder, or syndrome from occurring in a human, i.e. causing the clinical symptoms of the disease, disorder, or syndrome not to develop in an animal that may be exposed to or predisposed to the disease, disorder, or syndrome but does not yet experience or display symptoms of the disease, disorder, or syndrome; (ii) inhibiting the disease, disorder, or syndrome, i.e., arresting its development; and (iii) relieving the disease, disorder, or syndrome, i.e., causing regression of the disease, disorder, or syndrome. As is known in the art, adjustments for systemic versus localized delivery, age, body weight, general health, sex, diet, time of administration, drug interaction and the severity of the condition may be necessary, and will be ascertainable with routine experimentation by one of ordinary' skill in the art.
[00114] “SRC and/or ABL kinase inhibitor(s)” includes, for example, dasatinib, imatinib (Gleevec®), and compounds described in WO 2006/074057.
[00115] “VEGFR inhibitor” includes, for example, one or more of the following: ZD6474 (Zactima), sorafenib, Angiozyme, AZD2171, SU5416, PTK787, AEE788, sunitinib (SUTENT), and compounds described in WO 2004/050681 and WO 2004/006846.
Embodiments of the Invention [00116] In one embodiment, the cancer is mediated, at least in part, by inhibiting MEK.
[00117] In another embodiment, the cancer is selected from melanoma, colon cancer, rectal cancer, pancreatic cancer, breast cancer, non-small cell lung cancer, small cell lung cancer, papillary thyroid cancer, anaplastic thyroid cancer, endometrial cancer, and ovarian cancer.
[00118] In another embodiment, one or more of the treatments) is one or more chemotherapeutic agent(s), [00119] In another embodiment, one or more of the chemotherapeutic agent(s) is selected from a taxane(s), a platin(s), a topoisomerase inhibitors), an alkylating agent(s), an antimetabolite(s), an antimicrotubule agent(s), and a bcr-abi inhibitor(s). in another embodiment, one or more of the chemotherapeutic agent(s) is an antimicrotubule agent(s) selected from Vincristine,Vinblastine, Vinorelbine, and Vindesine.
[00120] In another embodiment, one or more of the chemotherapeutic agent(s) is selected from rapamycin, carboplatin, cisplatin, oxaliplatin, gemcitabine, dacarbazine, topotecan, and irinotecan.
[00121] In another embodiment, one or more of the chemotherapeutic agent(s) is an AKT inhibitor. In another embodiment, the AKT inhibitor is selected from a compound in Table 2a and Table 2b.
[00122] In another embodiment, one or more of the chemotherapeutic agent(s) is selected from a compound in Table 2a and Table 2b, [00123] In another embodiment, one or more of the chemotherapeutic agent(s) is a cMET inhibitor. In another embodiment, the cMET inhibitor is selected from a compound in Table 3a, Table 3b, and Table 3c.
[00124] In another embodiment, one or more of the chemotherapeutic agent(s) is selected from a compound in Table 3a, Table 3b, and Table 3c.
[00125] In another embodiment, one or more of the chemotherapeutic agent(s) is an EGFR inhibitor. In another embodiment, the EGFR inhibitor is selected from Lapatinib (Tykerb®), gefitinib (Iressa®), erlotinib (Tarceva®), Zactima (ZD6474), AEE7S8. HKI-272, EKB-569, Cl 1033, and a compound selected from Table 4 and
Table 7. In another embodiment, the EGFR inhibitor is selected from Table 4 and Table 7.
[00126] In another embodiment, one or more of the chemotherapeutic agent(s) is a compound selected from Table 4 and Table 7.
[00127] In another embodiment, one or more of the chemotherapeutic agent(s) is an ErbB2 inhibitor. In another embodiment, the chemotherapeutic agent(s) is selected from lapatinib, EKB-569, HKI272, and CI1033.
[00128] In another embodiment, one or more of the chemotherapeutic agent(s) is an HSP90 inhibitor. In another embodiment, the HSP90 inhibitor is 17-AAG. 17-DMAG, Geldanamycin, and CNF2024.
[00.129] In another embodiment, one or more of the chemotherapeutic agent(s) is an IGF1R inhibitor. In another embodiment, the IGF1R inhibitor is selected from a compound in Table 5a and Table 5b.
[00130] In another embodiment, one or more of the chemotherapeutic agent(s) is selected from a compound in Table 5a and Table 5b.
[00131] In another embodiment, one or more of the chemotherapeutic agent(s) is an Raf inhibitor. In another embodiment, the Raf inhibitor is selected from sorafenib and a compound in Table 6.
[00132] In another embodiment, one or more of the chemotherapeutic agent(s) is a VEGFR inhibitor. In another embodiment, the VEGFR inhibitor is selected from a compound in Table 4 and Table 7.
[00133] In another embodiment, one or more of the chemotherapeutic agent(s) is selected from rapamycin, a rapamycin analogue, ΡΪ103, SF1126, and BEZ235. In another embodiment, one or more of the chemotherapeutic agent(s) is selected from rapamycin, CCI-779, AP23573, RAD001, TAFA93, PI103, SF1126, and BEZ235. In another embodiment, one or more of the chemotherapeutic agent(s) is selected from rapamycin, CCI-779, AP23573, RAD001, PI 103, and SF1126. In another embodiment, one or more of the chemotherapeutic agent(s) is rapamycin. In another embodiment, one or more of the chemotherapeutic agent(s) is a rapamycin analogue.
[00134] In another embodiment, one or more of the chemotherapeutic agent(s) is 2-methyl-2-(4-(3-methyl-2-oxo-8-(quinolin-3-yl)-2,3-dihydro-li7-imidazo[4,5-cjquinoli η-1 -yl)phenyl)propanenitrile.
[00135] In another embodiment, one or more of the treatment(s) is selected from radiation and hypothermia therapy. In another embodiment, one or more of the treatment(s) is radiation.
[00136] In another embodiment, one or more of the treatment(s) is one or more antibody(s). In another embodiment, one or more of the antibody(s) is selected from IGF1R antibody (including, for example, “IGF-1R A12 MoAb, 19D12, h7C10 and CP-751871). Alemtuzumab, Bevacizumab (Avastin®), Cetuximab (Erbitux®), Gemtuzumab, Gemtuzumab ozogamicin, Ibritumomab (tiuxetan), Panitumumab, Rituximab, Tositumomab, and Trastuzumab (Herceptin®).
[00137] In another embodiment, one or more of the treatment(s) is surgery.
[00138] In another embodiment, one or more of the treatment(s) is one or more hormone therapy(s). In another embodiment, one or more of the hormone therapy(s) is selected from tamoxifen and an aromatase inhibitor.
[00139] In another embodiment, one or more of the chemotherapeutic agent(s) is gemcitabine, [00140] In another embodiment, one or more of the chemotherapeutic agent(s) is Imatinib (i.e. Gleevec®).
[00141] In another embodiment, the cancer is primary or relapsed CML and/or acute myelogenous leukemia (AML) and one or more of the treatment(s) is selected from one or more of the chemotherapeutic agent(s) and one or more antibody(s). In another embodiment one or more of the chemotherapeutic agent(s) is selected from Imatinib (i.e. Gleevec®) and PKC412; in another embodiment, one or more of the chemotherapeutic agent(s) is Imatinib (i.e. Gleevec®). In another embodiment one or more of the antibody(s) is selected from “IGF-1R A12 MoAb and trastuzumab.
[00142] In another embodiment, the cancer is prostate cancer and one or more of the treatment(s) is selected from one or more antibody(s). In another embodiment one or more of the antibody(s) is “IGF-1R A12 MoAb.
[00143] In another embodiment, the cancer is malignant melanoma and one or more of the treatment(s) is selected from surgery and one or more chemotherapeutic agent(s). In another embodiment, one or more of the chemotherapeutic agent(s) is selected from an alkylating agent(s), a taxane(s), a platin(s), and a Raf inhibitors). In another embodiment, one or more chemotherapeutic agent(s) is selected from sorafenib, Paclitaxel (Taxol®), Docetaxel (Taxotere®), dacarbazine. rapamycin, imatinib mesylate (Gleevec®), sorafenib, and carboplatin.
[00144] In another embodiment, the cancer is colon or rectal cancer and one or more of the treatment(s) is selected from surgery, radiation, one or more chemotherapeutic agent(s), and one or more antibody(s). In another embodiment, one or more of the chemotherapeutic agent(s) is selected from cisplatin, oxaliplatin, carboplatin, 5-fluorouracil, Capecitabine (Xeloda), Irinotecan (Camptosar), FOLFOX (Folinic acid, 5-FU, Oxaliplatin), and leucovorin. In another embodiment one or more of the antibody(s) is selected from bevacizumab and cetuximab.
[00145] In another embodiment, the cancer is pancreatic cancer and one or more of the treatment(s) is selected from surgery, radiation, and one or more chemotherapeutic agent(s). In another embodiment, one or more of the chemotherapeutic agent(s) is selected from erlotinib (Tarceva®), gemcitabine, 5~fluorouracil, leucovorin, cisplatin, oxaliplatin, carboplatin, gemcitabine, irinotecan, paclitaxel, capecitabine, and streptozocin.
[00146] In another embodiment, the cancer is breast cancer and one or more of the treatment(s) is selected from surgery, radiation, one or more chemotherapeutic agent(s), one or more hormone therapy(s), and one or more antibody(s). In another embodiment one or more of the chemotherapeutic agent(s) is selected from lapatinib (Tykerb®), Paclitaxel (Taxol®), docetaxel, capecitabine, Cyclophosphamide (Cytoxan), methotrexate, fluorouracil, doxorubicin, epirubicin, gemcitabine, carboplatin (Paraplatin), cisplatin (Platinol), vinorelbine (Navelbine), capecitabine (Xeloda), pegylated liposomal doxorubicin (Doxil), and albumin-bound paclitaxel (Abraxane), In another embodiment one or more of the antibody(s) is selected from ** IGF-1R A12 MoAb, bevacizumab (Avastin), and trastuzumab. In another embodiment, one or more of the hormone therapy(s) is selected from tamoxifen, Toremifene (Fareston), Fulvestrant (Faslodex), Megestrol acetate (Megace), ovarian ablation, and an aromatase inhibitor^); in another embodiment, one or more of the aromatase inhibitor(s) is selected from etrozole (Femara), anastrozole (Aximidex), and exemestane (Aromasin). 100147] In another embodiment, the cancer is non-small cell lung cancer and one or more of the treatment(s) is selected from surgery, radiation, one or more antibody(s), and one or more chemotherapeutic agent(s). In another embodiment, the chemotherapeutic agent(s) is selected from cisplatin, oxaliplatin, carboplatin, Zactima (ZD6474), Paclitaxel, Docetaxel (Taxotere®), Gemcitabine (Gemzar®), Vinorelbine,
Irinotecan, Etoposide, Vinblastine, Erlotinib (Tarceva®), and Pemetrexed. In another embodiment, one or more of the antibody(s) is Bevacizumab.
[00148] In another embodiment, the cancer is small cell lung cancer and one or more of the treatment(s) is selected from surgery, radiation, and one or more chemotherapy agent(s). In another embodiment, one or more of the chemotherapy agent(s) is selected from cispiatin, oxaliplatin, carboplatin, etoposide, irinotecan, fosfamide, paclitaxel, docetaxel, gemcitabine, Topotecan, cyclophosphamide/doxorabicin/vincrisdne (CAV), methotrexate, and vinorelbine.
[00149] In another embodiment, the cancer is papillary or anaplastic thyroid cancer, and one or more of the treatment(s) is selected from surgery, radiation, radioactive iodine therapy, one or more hormone therapy(s), and one or more chemotherapeutic agent(s). In another embodiment, one or more of the chemotherapeuti c agent(s) is selected from thyroid hormone pills, Doxorubucin and a platin(s).
[00150] In another embodiment, the cancer is endometrial cancer and one or more of the treatment(s) is selected from surgery, radiation, hormone therapy, and one or more chemotherapeutic agent(s). In another embodiment, one or more of the chemotherapeutic agent(s) is selected from paclitaxel, doxorubicin, and cispiatin. In another embodiment, one or more of the hormone therapy is selected from medroxyprogesterone acetate, megestrol acetate, and Tamoxifen, [09151] In another embodiment, the cancer is ovarian cancer and one or more of the treatments) is selected from surgery, radiation, and one or more chemotherapeutic agent(s). In another embodiment, one or more of the chemotherapeutic agent(s) is selected from a platin(s) compound (such as cispiatin, oxaliplatin and carboplatin), a taxane (such as paclitaxel or docetaxel), topotecan, anthracyclines (such as doxorubicin (Adriamycin) and liposomal doxorubicin (Doxil)), gemcitabine, cyclophosphamide, vinorelbine (Navelbine), hexamethyimelamine, ifosfamide, and etoposide.
[00152] In another embodiment, one or more of the treatment(s) is selected from one or more chemotherapeutic agent(s), radiation, hypothermia therapy, one or more antibody(s), and surgery. In another embodiment, one or more of the chemotherapeutic agent(s) is selected from an EGFR inhibitor, isotretinoin, a platin (e.g,, cispiatin, oxaliplatin, and carboplatin), epirubicin, bleomycin, doxorubicin, cyclophosphamide, a taxane (e.g. docetaxel (Taxotere®)), and fluorouracil [5-FU], In another embodiment, one or more of the chemotherapeutic agent(s) is selected from cisplatin, carbopiatin, and docetaxel. In another embodiment, one or more of the antibody(s) is cetuximab (Erbitux®), [00153] In another embodiment one or more of the treatment(s) is selected from radiation and surgery.
[00154] In another embodiment of the invention, one or more of the treatments is selected from rapamycin, CC1-779, AP23573, RADQ01, carbopiatin, cisplatin, oxaliplatin, gemcitabine, dacarbazine, topotecan, irinotecan, sorafenib, paclitaxel, docetaxel, Lapatinib (Tykerb®), gefitinib (iressa®), erlotinib (Tarceva®), Zactima (ZD6474), 5-fluorouracil, Capecitabine (Xeloda), FOLFOX (Folinic acid, 5-FU, Oxaliplatin), streptozocin, Cyclophosphamide (Cytoxan), methotrexate, doxorubicin, epirubicin, vinorelbine (Navelbine), pegylated liposomal doxorubicin (Doxil), and albumin-bound paclitaxel (Abraxane), Etoposide, Vinblastine, Pemetrexed, leucovorin, fosfamide, cyclophosphamide/doxorubicin/vincristine (CAV), thyroid hormone pills, hexamethylmelamine, ifosfamide, Imatinib (i.e. Gleevec®), “IGF-1R A12 MoAb, IGF-1R 19D12, IGF-1R h7C10, IGF-1R CP-751871, Alemtuzumab, Bevacizumab (Avastin®), Cetuximab (Erbitux®), Gemtuzumab, Gemtuzumab ozogamicin, Ibritumomab (tiuxetan), Panitumumab, Rituximab, Tositumomab, Trastuzumab (Herceptin®), tamoxifen, Toremifene (Fareston), Fulvestrant (Faslodex), Megestrol acetate (Megace), ovarian ablation, medroxyprogesterone acetate, megestrol acetate, and an aromatase inhibitor.
[00155] In another embodiment, one or more of the chemotherapeutic agent(s) is of formula 100:
100 where q is 1, 2, or 3; E is -NR9-, -O, or absent and Y is -CH2CH2-, -CII2-, or absent provided that when E is -NR9- or -Ο-, then Y is -CH2CH2-; R2 is selected from halogen, trihalomethyl, -CN, -NO2, -OR3, and optionally substituted lower alkyl;, R8 is selected from -H, optionally substituted lower alkyl, -CG2R3, -C(0)N(R3)R4, -SO2R4, and -C(0)R3; or a single geometric isomer, stereoisomer, racemate, enantiomer, or diastereomer5 thereof and optionally as a pharmaceutically acceptable salt or hydrate thereof. The terms used to describe the scope of formula 100 are defined in WO 2004/006846 (US Nat’l Stage Application Serial No. 10/522,004) which is herein incorporated by reference. For example “optionally substituted alkyl” for formula 100 has the meaning given in WO 2004/006846 (US NatT Stage Application Serial No. 10/522,004). Whenever a compound of formula 100 is described in this application, whether by structure or by use of the term “formula 100,” the terms used to describe that compound are defined by WO 2004/006846 (US Nat!l Stage Application Serial No. 10/522,004).
[00156] In another embodiment, one or more of the chemotherapeutic agent(s) is of formula 101:
101 or a pharmaceutically acceptable salt, hydrate or solvate thereof, where A is a three- to seven-membered alicyclic, a five- to six-membered ortho-arylene or a five- to six-membered ortho-heteroarylene containing between one and three heteroatoms, either of the aforementioned optionally substituted with up to four R; each R is independently selected from -H, halogen, -CN, -NO2, -OR3, -N(R3)R3, -S(0)o-2R3, -S02N(R3)R3, -C02R3, -C(0)N(R3)R3, -N(R3)S02R3, -N(R3)C(0)R3, -N(R3)C02R3, -C(0)Rj, -OC(0)R3, optionally substituted Ci^alkyl, optionally substituted aryl, optionally substituted aryl Ci^alkyl, optionally substituted heterocyclyl, and optionally substituted heterocyclyl Ci^alkyl; optionally two of R, together with the atoms to which they are attached, form a first ring system fused with A, said first ring system substituted with zero to three of R!;
Xi, X2 and X3 are independently selected from ~CRI= or -N=; each R1 is independently selected from -II, halogen, -CN, -NO2, -OR3, -N(R3)R3, -S(0)o-2R3, -S02N(R3)R3, -CO2R3, -C(0)N(R3)R3, -N(R3)S02R3, -N(R3)C(0)R3, -N(R3)C02R3, -C(0)R3, -OC(0)R3, optionally substituted Ci .«alkyl, optionally substituted aryl, optionally substituted aryl C(-«alkyl, optionally substituted heterocyclyl, and optionally substituted heterocyclyl Chalky!; Z and X are each independently selected from ~C(Ic)=, -N=, -N(R )-, -S(0)o-2~, and -0-; E and Y are each independently selected from absent, -C(R2)(R2)-, -C(:::G)-, -C(R2)~ and ~N=, but E and Y are not both absent, and E and Y are not both -N= when both Z and X are —N=; each R2 is independently selected from R3, -N(R3)(R3), -C(0)N(R3)R3, ~N(R3)C02R3, -N(R3)C(0)N(R3)R3, and -N(R3)C(0)R3; each R3 is independently selected from -H, optionally substituted Ci^alkyl, optionally substituted C3-7alicyclic, optionally substituted aryl, optionally substituted aryl C stalky 1, optionally substituted heterocyclyl, and optionally substituted heterocyclyl Ci^alkyl; optionally two of R3, when taken together with a common nitrogen to which they are attached, form an optionally substituted five- to seven-membered heterocyclyl, said optionally substituted five- to seven-membered heterocyclyl optionally containing at least one additional heteroatom selected from N, O, S, and P; and G is selected from-CQ2R3, -C(0)R3, -C(0)N(R3)R3, -C(0)(NR3), -C(0)NR3[C(R3)2]o-.R3, -C(0)NR30[C(R3)2]o-tR3, -N(R3)C02R3, -N(R3)C(0)N(R3)R3, -N(R3)C(0)R3, -N(R3)R3, -S(0)o-2R3, -S02N(R3)R3, optionally substituted aryl Co-salkyl, and optionally substituted heterocyclyl Co-3alkyl; with the proviso, however, that the compound is not 2-[(3,4-dihydro-3-oxo-2/f-l,4-benzoxazin-6-yl)carbonyl]-iV-(2-furanylmethyl)-benzamide, A-cyclopropyl-2-[(3,4-dihydro-3-oxo-2H-1,4-benzoxazin-6-yl)carbonyl]-benzamide, or 2-[(3,4-dihydro-3-oxo-2/-/~l,4~benzoxazin-6-yl)carbonyl]-A'-(phenylmethyl)-benzamide. The terms used to describe the scope of formula 101 are defined in WO 2005/112932 (US Nat’l Stage Application Serial No. 11/568,789) which is herein incorporated by reference. For example “optionally substituted heterocyclyl55 for formula 101 has the meaning given in WO 2005/112932 (US Nat’l Stage Application Serial No. 11/568,789). Whenever a compound of formula 101 is described in this application, whether by structure or by use of the term “formula 101,” the terms used to describe that compound are defined by WO 2005/112932 (US Nat51 Stage Application Serial No. 11/568,789). 100157] In another embodiment, one or more of the chemotherapeutic agent(s) is of formula A-B-C or a pharmaceutically acceptable salt or hydrate thereof, wherein, A is selected from:
B is selected from: __
and, C is selected from:
wherein R.^ is selected from -H, halogen, trihalomethyl, -C1N, -NH2, NO2, OR", -NR3R3. ~S(0)o-2R35 -S02NR3R3, -CO2R·5, -C(0)NR3R35 -N(R3)S02R, ~N(R3)C(0)R3, -N(R3)C02R3, -C(G)R3, and optionally substituted lower alkyl; q is 0 to 2; each R3 is independently selected from -H, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted arylalkyl, and optionally substituted heteroarylalkyl; two R3, together with the nitrogen to which they are attached, form a four- to seven-membered heteroalicyclic, said four- to seven-membered heteroalicyclic optionally containing one additional heteroatom; when one said additional heteroatom is a nitrogen, then said nitrogen is optionally substituted with a group selected from -H, trihalomethyl, -SG2R5, -S02NR5R5, -CG2R\ -C(0)NR5R5, -C(0)R5, and optionally substituted lower alkyl; each RJ5 is independently selected from -H, -C(=0)R3, -C(=0)0RJ, -C(=G)SR3, -SO2R3, -C(~0)N(R3)R3, and optionally substituted lower alkyl; two R35, together with the nitrogen to which they are attached, can combine to form a heteroalicyclic optionally substituted with between one and four of R60, said heteroalicyclic may have an additional annular heteroatom, and said heteroalicyclic may have an aryl fused thereto, said aryl optionally substituted with an additional one to four of R60; A* is selected from =N~, =C(H)-, and -C(CN)-; A2 is either =N- or =C(H)-; R5 is -H or optionally substituted lower alkyl;
Rs is selected from R3, -S02NR3R3, -C02R3, -C(G)NR3R3, -SG2R3, and -C(G)R3; R9, Ri0, and Ru are each independently selected from ~H, and -QRi2; or R9 is selected from -H, and -OR12, and Ri0 and R11, when taken together, are either an optionally substituted alkylidene or an oxo; and R12 is selected from -H, ~C(0)R3, optionally substituted lower alkylidyne, optionally substituted lower arylalkylidyne, optionally substituted lower heterocyciylalkylidyne, optionally substituted lower alkylidene, optionally substituted lower alkylidenearyl, optionally substituted lower alkylideneheterocyclyl, optionally substituted lower alkyl, optionally substituted lower alkylaryl, optionally substituted aryl, optionally substituted lower heterocyclylalkyl, and optionally substituted heterocyclyl; or two R12,s, when taken together, form 1) a corresponding spirocyclic ketal when said two R12,s stem from Ri0 and Rn, or 2) a corresponding cyclic ketal when said two Ri2’s stem from R9 and one of Ri0 and Ru; E1 is selected from -O-, -CH2-, -N(R5)-, and -S(0)o-2-; Q is a five- to ten-membered ring system, optionally substituted with between zero and four of R20; R20 is selected from -H, halogen, trihalomethyl, -GN, -NO2, -NH2, -OR3, -NR3R3, -S(O)0.2R3, -S02NR3R3, -CO2R3, -C(0)NR3R3, -N(R3)S02R3, -N(R3)C(Q)R3, -N(R3)C02R3, -C(0)R3, and optionally substituted lower alkyl; R60 is selected from -H, halogen, trihalomethyl, -CN, -NOj, -NH2, -OR3, -NR3R3, -S(0)o-2R3, -S02NR3R3, -C02R3, -C(0)NR3R3, -N(R3)S02R3, -N(R3)C(0)R3, -N(R3)C02R3, -C(0)R3, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted heteroarylalkyl, and optionally substituted aryl alkyl; two of R60, when attached to a non-aromatic carbon, can be oxo; each methylene in any of the above formulae is independently optionally substituted with R25; each R25 is independently selected from halogen, trihalomethyl, -CN, -N02, -NHa, -OR3, -NR3R3, -S(0)o-2R-\ -S02NR3R3, -C02R3, -C(0)NR3R3, -N(R3)S02R3, -N(R3)C(0)R3, -N(R3)C02R3, -C(0)R3, optionally substituted aryl, optionally substituted arylalkyl, heteroarylalkyl, and optionally substituted lower alkyl; two of R25, together with the carbon or carbons to which they are attached, can combine to form a three- to seven-membered alicyclic or heteroalicyclic, two of R25 on a single carbon can be oxo; with the proviso that when B is selected from:
and C contains
and the remaining portion of C contains one of:
directly attached to
then A must be one of:
and with the proviso that when C contains
and B is selected from.
then theportion of C directly attached to cannot contain
when R70 is selected from -H, Chalky 1, and CMalkoxyl.
The terms used to describe the scope of formula A-B-C are defined in WO 2005/030140 (US Nat91 Stage Application Serial No. 10/573,336) which is herein incorporated by reference. For example “optionally substituted heterocyclyl” for formula A-B-C has the meaning given in WO 2005/030140 (US Nat’l Stage Application Serial No. 10/573,336). Whenever a compound of formula A-B-C is described in this application, whether by structure or by use of the term “formula A-B-C,” the terms used to describe that compound are defined by WO 2005/030140 (US Nat’l Stage Application Serial No. 10/573,336).
[00158] In another embodiment, one or more of the chemotherapeutic agent(s) is of formula 103:
103 or a pharmaceutically acceptable salt or hydrate hereof, wherein, each of J1, J2, and J3 is independently selected from =N-, =C(R!)-, -N(R*)-, -O- and -S(O)0-2-; each R1 is independently selected from -H, halogen, trihalomethyl, -CN, ~N02, -OR20, -N(R20)R20, -S(0)o.2R20, -SO2N(R20)R20, -CO2R20, -C(O)N(R20)R20, -N(R20)SO2R20, -N(R20)C(O)R20, -NCO2R20, -C(0)R20, optionally substituted C|. ealkyl, optionally substituted aryl, optionally substituted aryl Cj^alkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl Cj^alkyl and -D-R ; R2 is selected from -H, halogen, -OR20, -S(0)o-2R2°, -N02, -N(R20)R20, and optionally substituted Ci^alkyl; J4 is selected from =N-, =C(H>, and =C(CN>;
Ar is either a five- or six-membered arylene or a five- or six-memhered heteroarylene containing between one and three heteroatoms; each R3 is independently selected from -H, halogen, trihalomethyl, -CN, -NO2, -OR20, -N(R20)R20, -S(0)o.2R2°, -SO2N(R20)R20, -CO2R20, -C(O)N(R20)R20, -N(R20)SO2R20, -N(R20)C(O)R20, -NCO2R20, -C(0)R20, optionally substituted Cj. 6alkyl, optionally substituted aryl, optionally substituted aryl Chalky}, optionally substituted heterocyclyl, optionally substituted heterocyclyl Chalky! and a group -B-L-T, wherein B is selected from absent, -N(R13)-, -N(S02R13)-, -0-, -S(Q)0-2-, and -C(K))-; L is selected from absent, -C(=S)N(R13)-, ~C(==NRi4)N(Ri3)-, -SG2N(R13)~, -S02-, -C(~0)N(Ri3)-, -N(R13)-, -C(==0)C,.2alkylN(R13) -, -N(RI3)C,-2alkylC(=0)-, -C(=0)Co.,alkylC(-0)N(Ri3)-, -Co^alkylene-, -C(=0)C0.ialkylC(0)OR3-, -C(=NRi4)Co-(alkyl C(=0)-, -C(O)-, -C(==O)C0-!alkylC(-Q>, and an optionally substituted four to six-membered heterocyclyl containing between one and three annular heteroatoms including at least one nitrogen; and T is selected from -H, -Ri3, -Co^alkyl, -Co^alkylQ, -OCo^alkylQ, -Co-4alkylOQ, -N(R13)C0^alkylQ, -SO2C0-4alkylQ, -0(=0)0^ alky IQ, -C0^alkylN(Ri3)Q, and -C(=0)N (R13 )C0-4alky IQ, wherein each of the aforementioned alkyls and alkylenes of -B-L-T is optionally substituted with one or two of R60; Z is selected from -S(0)o~2-5 -0-, and -NR4-; R4 is either -H or optionally substituted Chalky!; each D is independently selected from -0-, -S(0)o-2-, and -NR5-; each R5 is independently -H or optionally substituted Ci^alkyl; each R13 is independently selected from -H, -C(=0)R20, -C(=0)0R2°, -C(~0)SR20, -S02R2°, -C(=O)N(R20)R20, and optionally substituted Chalky!; two of R13, together with the atom or atoms to which they are attached, can combine to form a heteroalicyclic optionally substituted with between one and four of R60, said heteroalicyclic can comprise up to four annular heteroatoms, and said heteroalicyclic can comprise an aryl or heteroaryl fused thereto, in which case said aryl or heteroaryl is optionally substituted with an additional one to four of R60; each R14 is independently selected from -H, -N02, -N(R20)R20, -CN, -OR20, optionally substituted Ci^alkyl, optionally substituted aryl, optionally substituted aryl Cj. 6alkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl C\. 6alkyl; Q is a five- to ten-membered annular system, optionally substituted with between zero and four of R20; each R3° is independently either R20, or according to formula 120;
120 wherein X1, X2, and optionally XJ, represent the atoms of a saturated bridged ring system, said saturated bridged ring system comprising up to four annular heteroatoms represented by any of X1, X2, and X3; wherein, each X1 is independently selected from -C(R6)R7-, -Ο-, -S(0)o_2-, and -NR8-; each X2 is independently an optionally substituted bridgehead methine or a bridgehead nitrogen; each X3 is independently selected from -C(R6)R7-, -0-, -S(0)o-2-, and -NR8-; Y is either: an optionally substituted lower alkylene linker, between D and either 1) any annular atom of the saturated bridged ring system, except X2 when X2 is a bridgehead nitrogen, or 2) any heteroatom, represented by any of R6 or R'; provided there are at least two carbon atoms between D and any annular heteroatom of the saturated bridged ring system or any heteroatom represented by any of R or R ; or Y is absent, when Y is absent, said saturated bridged ring system, is directly attached to D via an annular carbon of said saturated bridged ring system, unless D is -SO2-, in which case said saturated bridged ring system, is directly attached to D via an any annular atom of said saturated bridged ring system; m and p are each independently one to four; n is zero to two, when n equals zero there is a single bond between the two bridgehead X2’s; R6 and R7 are each independently selected from -H, halogen, trihalomethyl, -CN, -N02, -OR20, -N(R20)R20, -S(0)o-2R2°, -S02N(R20)R20, -C02R.20, -C(G)N(R20)R20, -N(R20)SO2R20, -N(R20)C(0)R20, -NC02R2°, -C(Q)R20, optionally substituted
Chalky!, optionally substituted aryl, optionally substituted aryl C^alkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl Chalky!, and a bond to either Y or D; or R and R , when taken together are oxo; or R6 and R7, when taken together with a common carbon to which they are attached, form a optionally substituted three- to seven-membered spirocyclyl, said optionally substituted three- to seven-membered spirocyclyl optionally containing at least one additional annular heteroatom selected from N, O, S, and P; each Rs is independently selected from -R20, Y, -SO2N(R20)R20, ~CQ2R205 -C(O)N(R20)R20, -S02R2°, and -C(0)R20: each R20 is independently selected from -H, optionally substituted Cj^alkyl, optionally substituted aryl, optionally substituted aryl Chalky!, optionally substituted heterocyclyl, and optionally substituted heterocyclyl Chalky!; or two of R20, when taken together with a common nitrogen to which they are attached, can form an optionally substituted five- to seven-membered heterocyclyl, said optionally substituted five- to seven-membered heterocyclyl optionally containing at least one additional annular heteroatom selected from N, 0, S, and P; each R60 is independently selected from -H, halogen, trihalomethyl, -CN, -N02, -OR20, -N(R20)R20, -S(0)o-2R2°, -SO2N(R20)R20, -C02R2°, -C(O)N(R20)R20, -N(R20)SO2R20, -N(R20)C(O)R20, -N(R20)CO2R20, -C(0)R20, optionally substituted Cj^alkvl, optionally substituted aryl, optionally substituted aryl Ci. galkyl, optionally substituted heterocyclyl, optionally substituted heterocyclyl Ci-6alkyl; two of R60, when taken together with a common carbon to which they are attached, can form an optionally substituted three- to seven-membered alicyclic or heteroalicyclic; and two of R60, when taken together can be oxo.
The terms used to describe the scope of formula 103 are defined in WO 2006/014325 (US Nat’! Stage Application Serial No. 11/571,140) which is herein incorporated by reference. For example “optionally substituted heterocyclyl” for formula 103 has the meaning given in WO 2006/014325 (US Nat’I Stage Application Serial No. 11/571,140). Whenever a compound of formula 103 is described in this application, whether by structure or by use of the term “formula 103 ” the terms used to describe that compound are defined by WO 2006/014325 (US NatT Stage Application Serial No. 11/571,140). (00159] In another embodiment, one or more of the chemotherapeutic agent(s) is ;V-[3-fluoro-4-( {6-(methyloxy)-7-[(3-morpholin-4-ylpropyl)oxyjquinolin-4- yl}oxy)phenyi]-jV'-[2-(4-fluorophenyl)ethyi]ethanediamide or pharmaceutically acceptable salt or hydrate thereof. (00160] In another embodiment, the cMet inhibitor is Ar-[3-fluoro-4-({6-(methvloxy)-7-[(3-morpholin-4-ylpropyl)oxy]quinolin-4-yl}oxy)phenyl]-iV,-[2"(4-iluoropheRyljethyl jethanediamide or a pharmaceutically acceptable salt or hydrate thereof. (00161] In another embodiment, one or more of the chemotherapeutic agent(s) is of formula 105:
105 or a pharmaceutically acceptable salt or hydrate thereof, wherein, V is NRiRia, or O-Rj, wherein
Rj is H, CN, halo, -NR13R14, C(0)NR!3Rm5 C,-C6 alkyl, -C(0)-CrC6 alkyl, -Co-Cg alkyl-R2o, wherein R20 is aryl, heteroaryl, heterocyclyl, or a 5-12 membered fused bicyclical or tricyclic saturated, partially saturated, or unsaturated ring system containing 0-4 ring atoms selected from N, O, and S, wherein aryl, heteroaryl, C3-C7 heterocyclyl, or the 5-12 membered ring system are optionally substituted with one, two, or three groups independently selected from Ci-Q alkyl, and -C0-C6 a!kyl-R2i;
Ria is H or CpCe alkyl; or when V is NRiRia, Ri and Ria together with the nitrogen to which they are attached form a 4-7 membered heterocyclyl or heteroaryl group containing, in addition to the nitrogen, up to two additional heteroatoms independently selected from Ο, N, and S, and wherein each heterocyclyl or heteroaryl group is optionally substituted with one or two of Ci-Cs alkyl, -NR13R14 or C3-C7 cycloalkyl; X is H, halo, CpCg alkyl, NO2, mono-, di-, or tri-halo substituted methyl, NR13R14, C(0)0-Ci-C6 alkyl, or N(Ri3)-C(0)-CpC6 alkyl; Y is Η, halo, OH, Ci-Cg alkyl, Co-Cg alkyl-NRisR^, NR15R16, Ci-Cg alkoxy, -N(Ri3)-(CH2)n-NRi5Ri6, -C(0)0-C,-C6 alkyl, -0-(CH2)n-NRiSRi6, -C(G)~CrCg alkyl, -C0-C6-alkyl-R2i, -0-R2I, -C(0)-R2i, -0-(CH2)n-R2t, -C(0)-NR13Rt4, -C(O)-N(R13)-aryl, -C(0)-N(R!3)-(CH2)„-NR15R16, -C(0)-N(R,3)-(CH2)n-aryl, -C(Q)~ N(Ri3)-(CH2)n-heterocyclyl; or X and Y together with the atoms to which they are attached form a 4-7 membered heterocyclyl or heteroaryl group containing one or two heteroatoms independently selected from Ο, N, and S, wherein the heterocyclyl or heteroaryl group is optionally substituted with one or two moieties independently selected from halo, CrQ alkyl, aryl-Ci-Cg alkyl-, aryl-(CH2)n-0-(CH2)n-aryl-, ary 10H, C^-Cy cycloalky!, heterocyclyl, -aryl-N(R!3)C(0)-C3-C-7 cycloalkyl-C(0)-N(Ri4)-aryl, or a group of the formula -L-M-Q, wherein L is a bond or Cs-C? cycloalkyl, M is Cj-Q alkyl, C2-Cg alkenyl, or C2-C6 alkynyl, Q is NR!3Ri4, N(Ri3)C(0)-Ci-C6 alkyl, heterocyclyl, or a saturated fused bicyclic ring containing one or two heteroatoms independently selected from Ο, N, and S, wherein each aryl, heteroaryl, or heterocyclyl substituent on the group formed by X and Y is optionally further substituted with one or two moieties independently selected from halo, C(0)0-(CH2)„-phenyl, and C(0)-Cj~ C6 alkyl; Z is H, NR2R3, -S-R2a, or -0-R2a, wherein R2 is -Ci-Ce alkyl, -Cj-Ce alkyl-NRi3Ri4, -C(0)-aryl, -Co~C6-alkyl-aryl, -Co-CVaikyl-heteroaryl, -Co-C6-alkyl-(C3-C7-cycloalkyl), -Co-Cfi-alkyl-heterocyclyl, or -Co-Q alkyl-5-12 membered fused bicyclic or tricyclic saturated, partially saturated, or unsaturated ring system containing 0-4 ring atoms selected from N, 0, and S, wherein each alkyl is optionally substituted with phenyl, and each aryl, heteroaryl, C^-Cy cycloalkyl, heterocyclyl, or 5-12 membered ring system is optionally substituted with one, two, or three groups independently selected from halo, mono-, di-, or tri-halo substituted methyl or methoxy, CN, N02, NRi3Ri4, C(0)G~CrC6 alkyl, N(Ri3)C(0)-C1-C6 alkyl, -S02NRi3Rm, ~G-C(0)-NR13Ri4, -Co-C6 alkyl - C(0 )N R15 R16, Ci-C6 alkoxy, Cs-Cg thioalkoxy, -0-(CH2)n-NRi5Ri6> -Ci-Cg alkyl-NRi3Ri4, -N(R,3)-C(0)-Ci-C6 alkyl, -N(Ri3)-C(0)-aryl, -C0-C6 alkyl-C(0)-N(R13)-(CH2)n-NR15Ri6, -C0~C6 alkyl-C(0)-N(Ri3)~ (CH2)n-aryl, -0-(CH2)„-C(0)-N(Rt3)-(CH2)[rNRt5Ri6, -O-(CH2)n-C(0)~NR,5R16, -Co-C6alkyl-C(0)-N(R,3HCH2)„-0-Ci-C6 alkyl, -C0-C6 alkyl-N(Ri3)-C(0)0-CrC6 alkyl, -C0-Qalkyl-C(0)-heterocyclyl, -C0-C6alkyl-C(O)-heteroaryl, -Co-Cealkyl-C(0)-aryl, -Co-C6~alkyi-R2), aryloxy, -0-(CH2)n-R2t, -SO2-heterocyclyl, N(Rt3)-C(0)-C3-C7-cycloalkyl, -Co-Cealkyl C(0)0-R2i ,C3-C?-cycloalkyl, -C0-C6alkylR2!, -SCi-C6alkyl or C1-C6 alkyl optionally substituted with halo or cyano, wherein each aryl, heteroaryl, cycloalkyl, or heterocyclyl substituent is further optionally substituted with 1 -3 groups independently selected from halo, CF3, CpCg alkyl, Cj-Cg haloalkoxy, NR13R14 and Cj-Ce alkoxy; R3 is H or Ci-Ce alkyl: or R2 and R3 together with the nitrogen to which they are attached form a 4-7 membered heterocyclyl or heteroaryl group containing up to three heteroatoms independently selected from Ο, N, and S, and wherein the heterocyclyl or heteroaryl group is optionally substituted with one or two of halo or CpCg alkyl; R2a is aryl or C0-C6 alkyl-heteroaryi, wherein the aryl and heteroaryl are optionally substituted with aryl, -N(R13)-C(0)-C3-C7 cycloalkyl or -C(0)NRi3Ri4;
Rn and R14 are independently H or Cj-C6 alkyl;
Rj5 and R|6 are independently H, C1-C6 alkyl, heteroaryl, or heterocyclyl, or Ri5 and Ri6 together with the nitrogen to which they are attached form a 4-7 membered heterocyclyl or heteroaryl group wherein one or two ring carbons are each optionally replaced with a heteroatom independently selected from 0, N, and S, and wherein each heterocyclyl or heteroaryl group is optionally substituted with one or two moieties independently selected from halo, Ci-Ce alkyl, or -C(0)0-Cj-C6 alkyl; R2i is heterocyclyl, aryl, heteroaryl, or Q-C7 cycloalkyl, and wherein alkyl, aryl, heteroaryl, C5-C7 cycloalkyl, and heterocyclyl are optionally substituted with one or two moieties independently selected from halo, -S(0)2-Co~Ci alkyl, -C(O)-C0-Ci alkyl, -C(0)-H, -C0-C, alkyl-aryl, Cj-Q alkyl, NR!3Ri4, and heterocyclyl; n is 0-6; provided that when V is NH2, X, Y and Z are not simultaneously H.
The terms used to describe the scope of formula 105 are defined in WO 2006/074057 (US Nat’l Stage Application Serial No. 11/722,719) which is herein incorporated by reference. For example “optionally substituted heterocyclyl” for formula 105 has the meaning given in WO 2006/074057 (US Nat’l Stage Application Serial No. 11/722,719). Whenever a compound of formula 105 is described in this application, whether by structure or by use of the term “formula 105,” the terms used to describe that compound are defined by WO 2006/074057 (US Nat’l Stage Application Serial No. 11/722,719).
[00162] In another embodiment, one or more of the chemotherapeutic agent(s) is of formula 107:
107 or a pharmaceutically acceptable salt or hydrate thereof, wherein, each W is independently N or CR1; each R1 is independently selected from -H, halogen, trihaloalkyl, -CN, -NHi, -NO2, -OR6, -N=CNR6R7, -N(R6)C(==NRs)NR6R7, -SR6, -S(Q)i.2R6, ~S02NR6R7, -CO2R6, -C(0)NR6R7, -C(0)N(0R6)R7, -C(=NRs)NR6R7, -N(R6)S02R7, -NC(0)R6, -NC02R6, -C(0)R7, -R7, and -A-R7; provided at least one of R1 is -A-R7, wherein, only for said at least one -A-R7, R' must be an optionally substituted heteroalicyclic ring, and any nitrogen of said optionally substituted heteroalicyclic ring cannot be directly bound to A; A is O, S(0)o-2, and NR6: L is O, S(0)o-2, or NR3; Q is C or N, when Q is N, then R4 does not exist; R" and R3 are each independently -H or -R7; R4 and R5 are each independently selected from -H, -OR6, ~NR6R7, -S(0)o-2R6, -S02NR6R7, -C02R6, -C(0)NR6R7, -N(R6)S02R6, -NC(0)R6, -NC02R6, -C(0)R7, -CN, -N02, -NH2, halogen, trihalomethyl, and -R7; or R4 and R5, when taken together, form a five or six-membered aromatic ring system containing between zero and two nitrogens, said five or six-membered aromatic ring system optionally substituted with between zero and four of R15; R6 is selected from -H, optionally substituted Ci^alkyl, optionally substituted arylC|.galkyl, optionally substituted heterocyclylCj-galkyl, optionally substituted aryl, and optionally substituted heterocyclyl; R7 is selected from -H, optionally substituted Ci^alkyl, optionally substituted arylCi-galkyl, optionally substituted heterocycly 1C j.galkyi, optionally substituted aryl, and optionally substituted heterocyclyl; provided that there are at least two carbons between any heteroatom of R and A or either nitrogen to which R or R are attached; or R6 and R7, when taken together with a common nitrogen to which they are attached, form an optionally substituted five- to seven-membered heterocyclic ring, said optionally substituted five- to seven-membered heterocyclic ring optionally containing at least one additional heteroatom selected from nitrogen, oxygen, sulfur, and phosphorus; R8 is -H, -N02, -CN, -OR6, and optionally substituted Ci-galkyl; X is selected from one of the following six formulae:
wherein m is zero to five, n is zero to three, and Z is N or CR10; R!0 is selected from -H, halogen, trihalomethyl, -NH2, -N02, -OR6, -N=CNR6R7, -NR6R7, -N(R6)C(=NR8)NR6R7, -SR6, ~S(0)i-2R6, -so2nr6r7, -co2r6, -C(0)NR6R7, -C(0)N(0R6)R7, -C(=NR8)NR6R7, -N(R6)S02R6, -NC(0)R6, -NCO2R6, -C(0)R7, and R7;
Kis 0, S, or NR11; R11 is selected from cyano, -NO2, -OR6, -S(0)i-2R6, -S02NR6R7, -CO2R6, -C(0)NR6R7, -C(0)N(OR6)R7, -C(0)R7, and R6; and 6 6 7 each R15 is independently selected from -H, halogen, -NH2, -NO2, -OR , -N=CNR R , -NR6R7, -N(R6)C(-NR8)NR6R7, -SR6, ~S(0)i-2R6, -so2nr6r7, -co2r6, -C(0)NR6R7, -C(0)N(0R6)R7, ~C(=NR8)NR6R7, -N(R6)S02R6, -NC(0)R6, -NCO2R6, -C(0)R7, and R7.
The terms used to describe the scope of formula 107 are defined in WO 2004/050681 (US Nat’l Stage Application Serial No, 10/533,555) which is herein incorporated by-reference. For example “optionally substituted aryl5’ for formula 107 has the meaning given in WO 2004/050681 (US Nat’l Stage Application Serial No. 10/533,555).Whenever a compound of formula 107 is described in this application, whether by structure or by use of the term “formula 107,” the terms used to describe that compound are defined by WO 2004/050681 (US Nat’l Stage Application Serial No. 10/533,555).
[00163] In another embodiment, one or more of the chemotherapeutic agent(s) is of formula 108:
108 or a pharmaceutically acceptable salt or hydrate thereof, wherein, X21 is N or CR22; X22 is N or CR23; X23 is N or CR24, but when X22 is N then X23 is CR24; each of R21, R.22, R23, R24, R25, R26, R27, Ras, R29 and R30, and each R3n ^32 ^ ^33 *s independently selected from -H, halogen, trihalomethyl, -ΟΝ, -N02> -NRasRssa, -S(0)o-2R.35, -S02NR35R35a, -CO2R355 *C(0)NR35R35a, -N(R3s)S02R35» -N(R35)C(0)R35, -N(R35)C02R35, -OR35, "€(0)Κ35, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted heteroeyclyl, optionally substituted heterocyclylaikyl, and optionally substituted arylalkyl; R is selected from -H, halogen, trihalomethyl, -S(0)o-2R35, -S02NR35R35a, -CO2R35, -C(0)NR35R35a, -OR35, -C(0)R35, optionally substituted lower alkyl, optionally substituted aryl, optionally substituted heteroeyclyl, optionally substituted heterocyclylaikyl, and optionally substituted arylalkyl; or two of R25, R26, R27, R285 R29, Rso, R31 or R32, together with the atom or respective atoms to which they are attached, combine to form an optionally substituted spirocyclic ring system, optionally substituted fused ring system, and optionally substituted saturated bridged ring system; each of R35 and R3sa is independently selected from -H, optionally substituted lower alkyl, optionally substituted lower alkoxy, optionally substituted aryl, optionally substituted lower arylalkyl, optionally substituted lower aryl alkoxy, optionally substituted heteroeyclyl, and optionally substituted lower heterocyclylaikyl; or R35 and R35a, together with the atom or respective atoms to which they are attached, combine to form an optionally substituted five- to seven-membered heteroeyclyl; and m is an integer from 0 to 5; n is an integer from 1 to 2; and with the provisos that when X22 is CR23 and X23 is N then R is not optionally substituted aryl, aralkyl or heteroaryl, and that when X23 is N and X23 is CR24 then R is not optional substituted aryl or heteroaryl and R21 is not -NRssRjsa, and that when X22 is CR23 and X23 is CR24 then R21 is not optionally substituted aryl; and that compounds 4-(4-(2-fluorophenyl)piperazin-l-yl)~l//-pyrazolo[3,4-djpyrimidine, 4-(4-(3 -chloropheny l)piperazin-1 -yl)-17/-pyrazolo[3,4-djpyrimidine, 6-(4-(2-nitro-4-(trifluoromeihyl)phenyl)piperazin-1 -yi)-7//-purine, 6-(4-(4-fluorophenyl)piperazin-1 -yl)-7/f-purine, 6-(4-(2,5-dimethylphenyl)piperazin-l-yl)-7//-purine, 6-(4-(3,4-dichlorophenyl)piperazin-1 -yl)-7H-purine, 6-(4-(2-fluorophenyl)piperazin-1 -yl)-7//-purine, 6-(4-(3-ch!oropheny!)piperazin~ 1 -yl)-7/f-purine, 6-(4-(4-methoxyphenyl)piperazin~ 1 -yl)-7//-purine, 6-(4-(4-nitrophenyl)piperazin-1 -yl)-7//-purine, 6-(4-phenvlpiperazin~ 1 -yl)-7//-purine, 4-(4-phenylpiperazin-1 -yl)-7//-pyrrolo[2,3-d]pyrimidine, 4-phenyl~l“(7//”pyrrolo[253"d]pyrimidin-4-yl)piperidin-4-ols 6-(4-(2-methoxyphenyI)piperazin-1 -yl)-7//-purine, 6-(4-(2-cbIorophenyl)piperazin-1 -yl)-7//-purine, 6-(4-o-tolylpiperazin-1 -yl)-7//-purine, 6-(4-(3-(trifluoromethyl)phenyl)piperazin-1 -yl)-7//~purine, 6-(4-(2-methoxyphenyl)piperazin-1 -yl)-7//-purine are not included in Formula I.
The terms used to describe the scope of formula 108 are defined in WO 2005/117909 (US Nat5! Stage Application Serial No. 11/568,173) which is herein incorporated by reference. For example “optionally substituted aryl55 for formula 108 has the meaning given in WO 2005/117909 (US Nat5! Stage Application Serial No. 11/568,173). Whenever a compound of formula 108 is described in this application, whether by structure or by use of the term “formula 108,” the terms used to describe that compound are defined by WO 2005/117909 (US Nat51 Stage Application Serial No. 11/568,173).
[00164] In another embodiment, one or more of the chemotherapeutic agent(s) is of formula 109:
109 or a pharmaceutically acceptable salt or hydrate thereof, wherein:
Rs is H, halo, cyano, aryl, heteroaryl, Cm alkyl, Cj-Ce alkenyl, or C2-C6 alkynyl, wherein the aryl, heteroaryl, alkyl, alkenyl and alkynyl are optionally substituted with one or two groups independently selected from CO2R10, CONR!0Ru, OR10, and NRjoRnj R2 is Η, NH2, SH, OH, or C1-C2 alkyl; R3, R4, Rs, and R* are each independently H, oxo, C02Rio, CONRioRi j, Cm alkyl, Cj-Cg alkoxy, or Ci-Cg aIkoxy-CpC4 alkyl, wherein the CfC4 alkyl, C1-C.6 alkoxy and Ci-Ce alkoxy-Ci-C4 alkyl in each group are independently optionally substituted with 1 or 2 substituents independently selected from C02Rio, CONRjoRn, QRto, and NRjoRu, or
Rs and R5 together with the carbons to which they are attached form a C3-C7 carbocyclic ring, wherein the ring is optionally substituted with H, halo, cyano, nitro, or amino, R4 and Rs together with the carbons to which they are attached form a C3-C7 carbocyclic ring, wherein the ring is optionally substituted with H, halo, cyano, nitro, or amino R3 and Rg together with the carbons to which they are attached form a bridged C5-C7 carbocyclic ring, wherein the ring is optionally substituted with H, halo, cyano, nitro, or amino, or R4 and R5 together with the carbons to which they are attached form a bridged CS-C7 carbocyclic ring, wherein the ring is optionally substituted with H, halo, cyano, nitro, or amino; L is C0-4 alkyl, C2-C6 alkenyl, -N(R12)-, -C(0)N(R12)-, -N(R12)C(0)-, -C(O)-, -0-{CH2)n-, or -(CH2)n-0-, wherein n is 1-4; Q, is N or CRj3, wherein RI3 is H or C(0)NR12(CH2)nNRioRi 1; Q2 is a bond, CR;^ O or N, wherein R14 is H, OH, C1-4 alkyl, C1-4 alkoxy, NR15R15, wherein Rjs is H or C1-4 alkyl, or Q2 and V together form C(=0); when Q2 is a bond, V is absent and Ro is not H; when Qi is CR13 and Q2 is CH, V is H, OH, NH2, Ci-Cg alkoxy, NR10R11, 0(CH2)nNRioRn, 0(CH2)n attached to a C or N of a 4-7 membered heterocyclyl, NRi2(CH2)nNRioRu, NR12C(0)NR12(CH2)„NR,oRii,NR,2C(0)(CH2)„NR,oRii, (CH2)mO(CH2)nNRH)Rii,(CH2)mNR12(CH2)nNRi0Ri„ (CH2)mCHRi2(CH2)nNRjoRn, Cm alkyl optionally substituted with OH or NRjoRu, or V is a 4-7 membered unsaturated cyclic containing 1-3 atom of 0 or N, or V is a bicyclic solublizing group; when Qj is N and Q2 is CH, or when Qi is CRi3 and Q2 is 0 or N, V is H, (CH2)mO(CH2)nNRf0Ru, (CH2)mNRi2(CH2)nNRi0Rn, (CH2)mCHRi2(CH2)nNR10Rn, C(O)NRI2(CH2)„NR!0Rn, C(0) (CH2)nNRi0Rii, C(O)O(CH2)nNR10Ru, C(O)C(O)NRI2(CH2)nNRI0Rii, S02(CH2)„NRtoRns C(0)-C2~C6 alkenyl, or Cm alkyl optionally substituted with OH or NRioRn, or V is a 4-7 membered saturated or unsaturated cyclic or heterocyclic containing 1-3 atoms of O or N, optionally substituted with 1 or 2 Cj-Cs alkoxy groups or V is a “bicyclic solublizing group”; m is 1-3, n is 1-4, W is CrC6 alkyl, NRioRn, or W is aryl, C3-C7 cycloalkyl, heterocyclyl, heteroaryl, or 5-12 membered fused bicylic or tricyclic saturated, partially saturated, or usaturated ring system containing 0-4 ring atoms selected from N, O, and S, wherein each aryl, cycloalkyl, heterocyclyl, heteroaryl, and fused bicyclic or tricyclic ring system is optionally substituted with 1,2, or 3 substituents independently selected from halo, CN, N02, CF3, OH, NRioRn, C1-C6 alkoxy, Cj-Ci alkyl, NO>, C(0)OCi-C6 alkyl, C(0)NRs2-Ci-C<s alkoxy, C(0)NRi2-heterocyclyl, aryl, O-aryl, O-CHj-aryl, N-aryl, wherein each aryl substituent is optionally further substituted with halo, or V, Q2, L, and W together form an aryl ring, heteroaryl ring, C3-C7 cycloalkyl ring, heterocyclyl ring, or a 5-12 membered fused bicylic or tricyclic saturated, partially saturated or usaturated ring system containing 0-4 ring atoms selected from N, O, and S, wherein each ring or ring system is optionally substituted with 1,2, or 3 groups independently selected from halo, CN, N02, CF3, OH, NRioRn, Ci-C6 alkoxy, Ci-C6 alkyl, N02, C(0)0Ci-C6 alkyl, C(0)NRi2-Ci-C(, alkoxy, C(0)NRi2-heterocycly 1, aryl, O-aryl, NH-aryl, wherein each aryl substituent is optionally further substituted with halo; and Rio, Rn and R12 are each independently H or Ci~g alkyl which is optionally substituted with aryl or heteroaryl, provided the compound is not a compound selected from: 4-(4-(2-fluorophenyl)piperazin-l-yl)-l//-pyrazolo[3,4-r/Jpyrimidine; 4-(4~(3-chlorophenyl)piperazin-1 -yl)- l//~pyrazolo[3,4~J]pyrimidine; ethyl 4-(1 //-pyrazolo[3,4-d]pyrimidin-4-yl)piperazine-1 -carboxylate; tert-butyl 4-(1 //-pyrazolo[3,4-J]pyrimidin-4-yl)piperazine-1 -carboxylate; and N-(4-phenoxyphenyl)-4-(l//-pyrazolo[3,4-d]pyrimidin-4-yl)piperazine-l-carboxamide.
The terms used to describe the scope of formula 109 are defined in WO 2006/071819 (US Nat’l Stage Application Serial No. 11/722,291 which is herein incorporated by reference. For example “optionally substituted aryl” for formula 109 has the meaning given in WO 2006/071819 (US Nat’l Stage Application Serial No. 11/722,291. Whenever a compound of formula 109 is described in this application, whether by structure or by use of the term “formula 109,” the terms used to describe that compound are defined by WO 2006/071819 (US Nat’l Stage Application Serial No. 11/722,291.
[00165] For each of the foregoing embodiments, the Compound of Formula I can be selected from any of the following embodiments, including from the Representative Compounds in Table 1.
[00166] In another embodiment of the Invention, the Compound of Formula I is that where R7 is halo and all other groups are as defined in the Summary of the Invention for Group A, Group B, Group C, or Group D. In another embodiment, R7 is iodo or bromo. In another embodiment, R7 is iodo. In another embodiment, the compound is that where R7 is iodo or bromo and all other groups are as defined in the Summary of the Invention for Group A.
[00167] In another embodiment of the Invention, the Compound of Formula I is that where X is halo and all other groups are as defined in the Summary of the Invention for Group A, Group B, Group C, or Group D. In another embodiment, X is fluoro or chloro. In another embodiment, X is fluoro. In another embodiment, the compound is that where X is fluoro or chloro and all other groups are as defined in the Summary of the Invention for Group A.
[00168] In another embodiment of the Invention, the Compound of Formula I is that where R7 and X are halo and all other groups are as defined in the Summary of the invention for Group A, Group B, Group C, or Group D. More specifcally, R7 is iodo and X is fluoro. In another embodiment, the compound is that where R7 is iodo and X is fluoro and all other groups are as defined in the Summary of the Invention for Group A.
[00169] In another embodiment of the Invention, the Compound of Formula I is that where R , R , R , and R are hydrogen and all other groups are as defined in the Summary of the Invention for Group A, Group B, Group C, or Group D. In another embodiment, R!, R2, R5, and R6 are hydrogen and all other groups are as defined in the Summary of the Invention for Group A.
[00170] In another embodiment of the Invention, the compound of Formula I is selected from Group A where all groups are as defined in the Summary of the
Invention.
[00171] In another embodiment of the invention (Al), the Compound of Formula I is that where X and R7 are halo and all other groups are as defined in the Summary of the Invention for a compound of Group A.
[00172] In another embodiment (A2), the compound of Formula I is selected from Group A where R10 and R12 are independently hydrogen or halo. In another embodiment, Rt0 and Ri2 are independently hydrogen or fluoro. In another embodiment, R10 is 3-fluoro and R12 is hydrogen. In another embodiment, R10 and R12 are fluoro, in another embodiment, 3-fluoro and 4-fluoro, 4-fluoro and 5-fSuoro, or 4-fluoro and 6-fluoro.
[00173] In another embodiment of the invention (A3), the compound of Formula I is that where Rl, R2, R5 and R6 are hydrogen and all other groups are as defined in the
Summary of the Invention for Group A.
[00174] in another embodiment (A4), the compound of Formula I is selected from Group A where X, R7, and A are as defined in the Summary of the Invention; and one of R1, R2, R3, R4, R5, and R6 is halo, nitro, -NR8R8’, -OR8, -NHS(0)2R8, -CN, -S(Q)mR8, -S(0)2NR8Rs’, -C(0)R8, -C(0)OR8, -C(0)NR8R8’, -NR8C(0)0R8’, -NR8C(0)NR8R8”, -NR8C(0)0R8’, -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), -CH2NR25C(-NH)(NR25aR25b), -CH2NR25C(=NH)(N(R25a)(N02), -CH2NR25C(-NH)(N(R25a)(CN), -CH2NR25C(=NH)(R25), -CH2NR2SC(NR25aR25b)=CH(NQ2), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NR8R8’, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)OR8, -C(0)NR8R8’, -NR8C(0)NR8 R8”, -NR8C(0)0R8’ and -NR8C(0)R8’; and the others of R!, R2, R3, R4, R5, and R6 are as defined in the Summary of the Invention; or one of R and R2 together with the carbon to which they are attached, R3 and R4 together with the carbon to which they are attached, and R5 and R6 together with the carbon to which they are attached forms C(O) or C(=NOH); and the others of R1, R2, R3, R4, R5, and R6 are as defined in the Summary of the
Invention.
[00175] In another embodiment of the Invention (A5), the compound of Formula I is selected from Group A where X, R7, and A are as defined in the Summary of the Invention; and R3 is halo, nitro, -NR3R8’, -OR8, -NHS(Q)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)0R8’, -NR8C(0)NR8R8” -NR8C(0)0R8’, -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), , -CH2NR25C(=NH)(NR25aR25b)s -CH2NR2sC(=NH)(N(R25a)(N02), -CH2NR25C(~NH)(N(R25a)(CN), -CH2NR25C(=NH)(R25), -CH2NR25C(NR25aR25b)=CH(N02), alkyl, alkenyl, alkynyi, cycloalkyl, heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyi, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NR8R8’, -NRsS(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)OR8, -C(0)NR8R8’, -NR8C(0)NR8’r8”, -NR8C(0)0R8' and ~NR8C(0)R8; and R4 is as defined in the Summary of the Invention; or R3 and R4 together with the carbon to which they are attached form C(O) or C(=NOH); and R1, R2, R5 and R6 are as defined in the Summary of the Invention.
[00176] In another embodiment of embodiment A5, the Compound of Formula I is that where R1, R2, R5 and R6 are hydrogen.
[00177] In another embodiment of the Invention (A6), the compound of Formula I is selected from Group A where X, R7, and A are as defined in the Summary of the
Invention; and R3 and R4 are independently halo, nitro, -NR8R8’, -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(0)ORs, -C(0)NR8R8’, -NR8C(0)0R8’, -NR8C(0)NR8R8’', -NR8C(0)0R8', -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), -CH2NR25C(=NH)(NR25aR25b), -CH2NR25C(=NH)(N(R25a)(N02), -CH2NR25C(=NH)(N(R25a)(CN), -CH2NR25C(=NH)(R25), -CH2NR25C(NR25aR25b)=CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocyeloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocyeloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -ORs, ~NRSR8', -NR8S(Q)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, -C(0)NR8Rr -NRsC(0)NR8R8” -NR8C(0)0R8’ and -NR8C(0)R8’; or R3 and R4 together with the carbon to which they are attached form C(O) or C(=NOH); R!. R2, R5 and R6 are are as defined in the Summary of the Invention.
[00178] In another embodiment of embodiment A, the Compound of Formula I is that where Rs, R2, R5 and R6 are hydrogen.
[00179] In another embodiment of the Invention (A7), the compound of Formula I is selected from Group A where X and R7 are halo; A is phenylene optionally substituted with Ri0 and R12 where Ri0 and R12 are independently hydrogen or halo; R!, R2, R5 and R6 are hydrogen; R3 is hydrogen and R4 is -NR8R8’ (where R8 is hydrogen, hydroxy, alkyl, alkoxy, aryl, cycloalkyl, heteroaryl, or heterocyeloalkyl and R8 is hydroxy, alkoxy, aryl, cycloalkyl, heteroaryl, or heterocyeloalkyl), -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(Q)ORs, -C(0)NR8R8 , -NR8C(0)0R8’, -NR8C(0)NRs,Rs”, -NR8C(0)0R8’, -NRsC(0)R8’, alkenyl, and alkynyl; where the alkenyl and alkynyl are optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocyeloalkyl, optionally substituted aryl, optionally substituted arylalkyl, g optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR , -NR8R8’, -NR8S(0)2R9, -CN, ~S(0)mR9, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)NR8R8”, -NR8C(0)0R8’ and -NR8C(0)R8’; or R3 and R4 together with the carbon to which they are attached form C(O) or C(=NOH); m, R8 , and R9 are as defined in the Summary of the Invention for a compound of
Group A: and unless otherwise specified in this embodiment, R and R are as defined in the Summary of the Invention for a compound of Group A.
[00180] In another embodiment of the Invention (A8), the compound of Formula I is selected from Group A where R3 is hydrogen, halo, hydroxy, alkoxy, or amino. In another embodiment, R3 is hydrogen, fluoro, hydroxy, methoxy, or amino. In another embodiment, R3 is hydrogen or hydroxy, In another embodiment, R3 is hydroxy.
[00181] In another embodiment of embodiment AS, the Compound of Formula I is that where X and R7 are halo; A is phenylene optionally substituted with R10 and R12 where R10 and Ri2 are independently hydrogen or halo; R!, R2, R5 and R6 are hydrogen; and R4, is as defined in the Summary of the invention for a compound of Group A.
[00182] Another embodiment of the Invention (A9) is that where the compound of
Formula I is selected from Group A where R1, R2, R5 and R6 are hydrogen; R3 is hydrogen, halo, hydroxy, alkoxy, or amino; and R4 is heterocycloalkyl, heteroaryl, or alkyl substituted with -NR8R8 where R8 and R8 and all other groups are as defined in the Summary of the Invention for a compound of Group A.
[00183] In another embodiment of embodiment A9, the Compound of Formula I is that where R4 is alkyl substituted with -NR8R8’ where R8 and R8 and all other groups are as defined in the Summary of the Invention for a compound of Group A. In another embodiment, the compound is of Formula 1(a) or 1(b):
1(a);
1(b) where R3 is as defined in A9; X, R7, R8, R8, R10, R12, Ri4, and R16 are as defined in the Summary of the Invention for a compound of Group A.
[00184] In another embodiment of embodiment A9, the Compound of Formula I is that where R4 is heterocycloalkyl.
[00185] In another embodiment of embodiment AS), the compound of Formula I is that where X and R7 are halo; A is phenyiene optionally substituted with R10 and R12 where R10 and R12 are independently hydrogen or halo; R3 is hydroxy; and R4 is alkyl substituted with -NR8R8 or R4 is heterocycloalkyl optionally substituted with one, two, or three groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyi, optionally substituted heteroaryl, -OR8, -NR8R8’, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)NR8'R8”, -NR8C(0)0R8’ and -NR8C(0)R8.’; and where m, R3, R8, R8’, R8”, and R9 are as defined in the Summary of the Invention for a compound of Group A.
[00186] In another embodiment of the Invention (A 10), the compound of Formula I is selected from Group A where R4 is a) hydrogen; b) -CH2N(R25)(NR25aR25b); c) -CH2NR2SC(=NH)(NR25aR25b); d) -CH2NR25C(=NH)(N(R25a)(N02); e) -CH2NR25C(=NH)(N(R2S,,)(CN); f) -CH2NR25C(=NH)(R25); g) -CH2NR25C(NR25aR25b)=CH(N02); h) alkyl; i) alkyl substituted with one or two -OR8 where R8 is hydrogen, aryl, or alkyl where the alkyl is substituted with one or two hydroxy; j) alkyl substituted with one, two, or three halo; k) alkyl substituted with nitro; l) alkyl substituted with -S(0)mR9 (where m is 0 and R9 is aryl); m) alkyl substituted with optionally substituted heterocycloalkyl; n) alkenyl; o) -NR8R8 (where R8 and R8 are independently hydrogen; alkyl; alkenyl; alkyl substituted with one or two hydroxy; alkyl substituted with one or two -NR30R30’ where R30 and R30’ are independently hydrogen, alkyl, or hydroxyalkyl; alkyl substituted with optionally substituted heteroaryl; or alkyl substituted with optionally substituted cycloalkyl); p) -C(0)NR8R8 (where R8 is hydrogen, alkyl, or alkenyl; and R8 is hydrogen; hydroxy; alkyl; alkenyl; alkyl substituted with one or two hydroxy; alkyl substituted with optionally substituted heterocycloalkyl; alkyl substituted with -NR30R30’ where R30 and R30’ are independently hydrogen, alkyl, or hydroxyalkyl; or optionally substituted alkoxy); q) -NR*C(0)0R8 (where R8 and R8 are independently hydrogen, alkyl, or alkenyl); r) alkyl substituted with -NRSR8 (where R8 is hydrogen, alkyl, alkenyl, alkynyl, «» or alkyl substituted with one or two hydroxy; and R is hydrogen; hydroxy; alkoxy; alkyl; alkenyl; alkynyl; optionally substituted alkoxy; alkyl substituted with one or two hydroxy; alkyl substituted with one or two alkoxy; alkyl substituted with -NRj0R30 where R30 and R30 are independently hydrogen, alkyl, or hydroxyalkyl; alkyl substituted with one or two hydroxy and one or two -NR30Rj0 where R30 and R30 are independently hydrogen, alkyl, or hydroxyalkyl; alkyl substituted with one, two, three, four, or five halo; alkyl substituted with optionally substituted cycloalkyl; alkyl substituted with optionally substituted aryl; alkyl substituted with one or two hydroxy and one optionally substituted aryl; alkyl substituted with optionally substituted heterocycloalkyl; alkyl substituted with optionally substituted heteroaryl; heteroaryl; aryl; aryl substituted with one or two hydroxy; aryl substituted with one or two alkoxy; aryl substituted with one or two halo; aryl substituted with one or two -NRj2C(0)R32a where R32 is hydrogen or alkyl and R32a is alkyl, alkenyl, alkoxy, or cycloalkyl; aryl substituted with -NR34S02R34a where R34 is hydrogen or alkyl and R34a is alkyl, alkenyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl; cycloalkyl; eyeloalkyl substituted with one or two hydroxy; cycloalkyl substituted with one or two hydroxy and one or two hydroxyalkyl; cycloalkyl substituted with one or two alkoxy; cycloalkyl substituted with carboxy; cycloalkyl substituted with -C(0)NRj3R33a where R33 is hydrogen or alkyl and R33a is alkyl, alkenyl, alkynyl, or cycloalkyl; alkyl substituted with -C(0)NR33R33a where R33 is hydrogen or alkyl and R3',a is alkyl, alkenyl, alkynyl, or cycloalkyl; cycloalkyl substituted with optionally substituted cycloalkyl; heterocycloalkyl; heterocycloalkyl substituted with alkyl; heterocycloalkyl substituted with alkoxycarbonyl; heterocycloalkyl substituted with optionally substituted arylalkyl; heterocycloalkyl substituted with one or two hydroxy; heterocycloalkyl substituted with one or two alkoxy; heterocycloalkyl substituted with one or two hydroxyalkyl; heterocycloalkyl substituted with one or two hydroxy, one or two alkoxy, and one or two hydroxyalkyl; alkyl substituted with optionally substituted aryloxy; alkyl substituted with -S(Q)nR31 where n is 0 and R3i is alkyl; alkyl substituted with carboxy; alkyl substituted with alkoxycarbonyl; or alkyl substituted with -NRj2C(0)R32“ where Rj2 is hydrogen or alkyl and R32a is alkyl, alkenyl, alkoxy, or cycloalkyl); s) -NRsC(0)R8 (where R8 is hydrogen, alkyl, or alkenyl; and R8 is hydrogen; alkyl; alkyl substituted with one or two hydroxy; alkyl substituted with optionally substituted heterocycloalkyl; alkyl substituted with -NR30R30 where Rj0 and R30 are independently hydrogen, alkyl, hydroxyalkyl, or alkenyl); t) cycloalkyl; u) cycloalkyl substituted with -NR R where R and R are independently hydrogen, alkyl, or alkenyl; v) heterocycloaikyl; w) heterocycloaikyl substituted with -NR R where R and R are independently hydrogen, alkyl, or alkenyl; x) heterocycloaikyl substituted with one or two alkyl; a o y) heterocylcloalkyi substituted with -C(0)0R where R is alkyl or alkenyl; z) alkyl substituted with -NR8C(0)R8 (where Rs is hydrogen, alkyl, or alkenyl and R8 is alkyl; alkenyl; or alkyl substituted with alkoxy, aryl, and one, two, or three halo); aa) heteroaryl; bb) heteroaryl substituted with -NR8R8 where R8 and R8 are independently hydrogen, alkyl, or alkenyl; alkyl substituted with optionally substituted heteroaryl; cc) alkyl substituted with ~NR8S(0)2R9 where R8 is hydrogen, alkyl, or alkenyl and R9 is alkyl or alkenyl; dd) alkyl substituted with -NR8C(0)0Rs where R8 and R8’ are independently hydrogen, alkyl, or alkenyl; ee) alkyl substituted with one aryl and one -NR8R8’ where R8 and R8 are independently hydrogen, alkyl, or alkenyl; or if) alkyl substituted with one or two -OR (where R is hydrogen) and one or two -NR8R8 where R8 and Rs are independently hydrogen, alkyl, or alkenyl. [00187} In another embodiment, R4 is hydrogen, -CH2N(H)(NHCH3), -CH2NHC(-NH)(NH2), -CH2NHC(=NHXNHN02),-CH2NHC(=NHXNHCN), -CH2NHC(:=NH)(phenyl), -CH2NHC(NH2)=CH(N02), methyl, ethyl, hydroxymethyl, 2,3-dihydroxypropyl, 3-hydroxy-2-methyl-prop-2-yl, N-( 1 -methoxy-prop-2-y 1)-aminomethyl, iV-(ethoxypropyr)-aminomethyl, iV-(ethoxyethyl)-aminomethyl, N~( 2,2-di methoxyethy l)-aminomethy 1, iV-(methoxyethyl)-aminomethyl, Ar-(isopropxyethyl)-aminomethyl, trifluoromethyl, 1 -nitro-ethyl, 1 -methyl-1 -nitro-ethyl, 1-nitro-propyl, 3-methyl-1-nitro-butyl, phenylthiomethyl, allyl, ethenyl, 2-methylthio-ethylaminomethyl, 3-methylthio-propylaminomethyl, N-(iert-butoxycarbonylaminopropyl)-aminomethyl, ;V-(1 -carboxyethyl)-aniinomethyl, N-(\R-carboxyethyl)-aminomethyl, N~( 15-carboxyethyl)-aminomethyl, N~( 1 -methoxycarbonylethyl)-aminomethyl, -NH2, -NH(CH2)3CH3, -NHCH3, -NH(CH2CH3), -NHCH2CH(CH3)2, -nhch2ch2oh, -nhch2ch2ch2nh2, -N(CH3)CH2CH2(heteroaryl), -NHCH2(cyeloalkyl), -C(0)NH2, -C(0)NH0H, -C(0)NH(0CH2CH(0H)CH20H), -C(0)NH(CH2)3CH3, -C(0)NHCH2CH-CH2, -C(0)NHCH2CH3, -C(0)NHCH2CH20H, -C(0)NHCH2CH(0H)CH20H, -C(0)NHCH2CH2CH(0H)CH20H, -C(0)NHCH2CH2(piperidin-1 -yl), -C(0)NH(phenyl), -C(Q)NHCH2CH2N(CH2CH3)2, -NHC(0)0C(CH3)3, ”NHC(0)0CH3, azetidinylmethyl, pyrrolidinylmethyl, 3-hydroxy-pyrrolidinylmethyl, 2-(methoxymethyl)-pyrrolidinylmethyl, 25-(methoxymethyl)-pyrrolidinylrnethyl, 2R-(methoxymethyl)-pyrrolidinylmethyl, morpholinylmethyl, hydroxypiperidinylmethyl, 4-alkyl-piperazinylmethyl, 4-alkyl-homopiperazinylmethyl, 4-(heterocycloalkyl)-piperidinylmethyl, 4-(dialkylaminoalkyl)-piperazinylmethyl, A'-hydroxyaminomethyl, iV-methoxyaminomethyl, Af-ethoxyaminomethyl, Af-ethylaminomethyl, 1 -(iV-ethyl-amino)-ethyl, N,iV-diethylaminomethyl, A(iV-dimethy4aminomethyl, aminomethyl, 1-amino-ethyl, 1 ^-amino-ethyl, lS-amino-ethyl, 1 -(methylamino)-ethyl, 1 ~(N, iV-dimethylamino)-ethyl, 1 -amino-1 -methyl-ethyl, 1 -aminopropyl, IS-aminopropyl, l/?-aminopropyl, AA(n-propyl)-aminomethyl, iV-(isopropyl)-aminomethyl, 2-(AAisopropylamino)-ethyl, 3-(Ar-isopropylamino)-2-methyl-prop-2-yl, 1 -(A'-ethyl-amino)-propyl, 1 -(Ar,A?-dieihyl-amino)-propyl, 1-aminobutyl, 1-amino-isobutyl, Ar-(2-aminoethyl)-aminomethyl, AA(n-buty 1 )-aminomethy 1, AZ-isobutylaminomethyl, tert-butylaminomethyl, 1 -(rert-butylamino)-ethyl, sec-butylaminomethyl, Ar-(2-methyl-but-3-yS)-aminomethyl, A-(3,3-dimethyl-butyI)-aminomethyl, iV-(3-methylbut-3-yl)-aminomethyl, Ar-(2-methylbutyi)-aminomethyI, Ar-(pent-3-yl)-aminomethyl, n-pentylaminomethyl, isopentylaminomethyl, 5ec-peniylaminomethyl, neopentylaminomethyl, A-(2,2,4~trimethyl-pent-4-yl)-aminomethyl, A-(2-ethyl~butyl)-aminomethyl, A-allyl-aminomethyl, 3-methyl-but-l-yn-3-ylaminomethyl, AA(2,3-dihydroxypropyioxy)-aminomethyi, iV-cyclopropylaminomethyl, A-cyclobutylaminomethyl, A-cyclopentylaminomethyl, A-cyclopenten-4-y laminomethyl, Λ-( I (R, S)-hydroxy-cyclopent-2-yl)-aminomethyi, A-(15~hydroxy-cyclopent-2-yI)-aminomethyl, N-( 1 /?-hydroxv-cyclopent-2-yl)-aminomethyl, N-( 1 (R, S)-hydroxy- l-methyl-cyclopent-2-yl)-aminomethyl, A-( 15-hydroxy- 1 -methyl-cyclopent-2~yl)-aminomethyi, N-( 1 Λ-hydroxy-1 -methyl-cyclopent- 2-yl)-aminomethyl, A-(3s4-dihydroxy-cyclopentyl)-aminomethyl, N-( 1 -hydroxymethyl-cyclopent-1 -yl)-aminomethy 1, A-(2,3-dihydroxy~4-hydroxymethyl-cyclopentyl)-aminomethyl, N~( 1 (^,^-methoxy-cyclopeni-2-yl)-aminomethyl, N-(\S~ methoxy-cyclopent-2-yl)-aminomethyl, N-(l /?-methoxy-eyclopent-2-yl)-aminomethyl, Λ-( 1 -carboxy-cyclopentyl)-aminomethyl, A-cyclohexylaminomethyl, iV-(l(^,5')-hydroxy-cycIohex-2-yl)-aminomethyls A-(c?>4-hydroxy-cyc!ohexyl)-aminomethyl, A-(/ram-4-hydroxy-cyclohexyl)-aminomethyl, 1 - [A-(c w-4-hydroxy-cyclohexyl)-amino]-ethyl, 1 -[A-(/ra«s-4-hydroxy-cyclohexyl)-amino]"eihyl, N-( 1 (R)~ hydroxy-cyclohex-2-yl)-aminomethyl, A-{l(5)-hydroxy-cyclohex-2-yl)-aminomethyl, A-(l-hydroxymethyi-cycIohexyl)-aminomethyl, iV-(2-cyclohexyl-cyclohexyl)-aminomethyl, A-{(2^,35,4^,6^)-2-{hydroxymethyl)-3,4-dihydroxy-6-niethoxy-tetrahydro-2//-pyran-5-yl}-aminomeihyl, A-(cycloheptyl)-aminomethyl, A-(cyclooctyl)-aminomeihyl, [(1 rs3r,5^>7^)-tricycIo[3.3.1.13,']dec-2-ylamino]methyl, A-[ 1 -(cyclopropylaminocarbonyl)-cy clopentylj-aminomelhyl, -CH2NHC(CH3)2C(0)NH(cyclohexyl), -CH2NHC(CH3)2C(0)NH(CH2CH3), Ar-(1 -benzyloxy-cyclopent-2-yl)-aminomethyl, Ar-(cyclopropy!methyl)-aminomethyl, A-(cyclohexylmethyl)-aminomethyl, A^l-cyc!ohexylethyl)-aminornethyl, iV-{imidazolyl)-aininomethyl, N-(l ,3,5-tria?inyl)-aminomethyl, A-(5-hydroxy-pyrazol-3-yl)-aminomethyl, A-(5-methyl-pyrazol-3-yl)-aminomeihyl, N-(benzimidazolyl)-aminomethyl, A-(pyrimidin-2-y l)-aminomethyl, A-(pyridin-2-yl)-aminomethyl, iV-(pyridin-3-yl)-aminomethyls A-(pyridin-4-yl)-aminomethyl, Ar-indan-1 -yl-aminomethyl, A-indan-2-yl-aminomethyl, phenylaminomethyl, A-(2-hydroxyphenyl)-aminomethyh A-(3-hydroxyphenyl)-aminomethy3, A-(4- hydroxyphenyl)-aminomethyl, jV-(2-methoxyphenyl)-aminomethy 1, N~(3 -methoxyphenyl)-aminomethyl, iV-(4-methoxyphenyl)-aminomethyl, N-(2~ fluorophenyl)-aminomethyl, AA(3-fluorophenyl)-aminomethyl, iV-(4-fluorophenyl)-aminomethvl, A^-iS-chlorophenyli-aminomethyL ./V-(3-chlorophcnyl)-aininoniethyl, N~ (4-chloropheny!)-aminomethyl, iV-(3-methylcarbonylamino-phenyl)-aminomethyl, N-(4-methylcarbonylamino-phenyl)-aminomethyl, AA(2-am inopheny l)-aniinoiTS ethyl, N-(3-aminophenyS)-aminomethyS, iV-(4-aminopheny!)-aminomethyl, N~(2-methylsulfonylaminophenyl)-aminomethyl, AA(3-methylsulfonylaminophenyl)-aminomethyl, iV-(4-incthylsulfonylaininophcnyl)-aininonicthyl, iV-(2-fluoro-4-hydroxy-phenyl)-aminomethyl, iV-(3-fluoro-4-hydroxy-phenyl)-aininomethyl, N-(benzyl)-aminomethyl, iV-(2-hydroxyphenylmethyl)-aminomethyl, N~(3-hydroxyphenylmethyl)-aminomethyl, iV-(4-hydroxyphenylmethyl)-aminomethyl, N-(2-(/V-me ih y ί p i p e r azin-1 -y l)-phenylmethyl)-aminomethyl, iV-(4-alkyl-phenethyl)- aminomethyS, Ar-(l-hydroxy-3-phenyl-prop-2~yl)-aininomeihy!,A;-(pynOSidin-2” ylmethylj-andnorneihyL jV-(AAalkyl-pyrrc>lidinylmethy!)-aininoinethyl, i¥-(iV-alkyI-pyrrolidinylethyl)-aminomethyl, A-(pyrro! i diny!propy! )-aminomeihyI, N-( 1,1-dimethyl-2-pyiTolidin-1 -yl-ethyl)-aminomethyl, /V*(tetrahydroftirany!methyl)-aminomethyl, AA(tetrahydro-2/f-pyran-4-ylmethyl)-aminomethyl, Ar-(tetrahydro-2#-pyranylethyl)-aminomethyl, A?-(piperidin-4-y!methy!)~aminomethyl, N-(N-methylpiperidin-4-ylmethyl)-aminomethy!, AA(iV-ieri-butoxycarbonylpiperidin-4-ylmethyl)-aminomethyl, AA(iV-methylimidazol-4-ylmethyl)-aminomethyl, N-(N-methylimidazol-5-ylmethyl)-aminomethyl, jV-[2-(imidazol-4-yl)-ethyl]-aminomethyl, N-[3-(imidazolyl)-propyl]-aminomethyl, N-(pyridin-3-ylethyl)-aminomethyl, N-(pyridin-4-ylethyl)-aminomethyl, A-(thien-2-ylethyl)-aminomethyl, iV-(furan-2-y!ethyl)-aminomethyl, JV-(5-niethyl-l,3,4-oxadiazol-2--ylmeihyl)-aminomethyl, A-(2-indolin-3-ylethyl)-aminomethyl, 2-(Ai,/V~dimethylamino)-ethylaminomethyl, 2~(N, AT-dimethylamino)-1 -methyl-ethyl aminomethyl,' 3 -aminopropyl aminomethyl, 3-(A^iV-dimethylamino)-propylaminomethyl, 3-(A’,Ai-diethylamino)-propylaminomethyl, N~(N, A^-diisopropylamino6thyl)-aminoniethyl, Λ ~(N, N dimethylaminobutyl)-aminomethyl, A-(3-hydroxypropyl)-aminomethyl, A-(2-hydroxypropyl)-aminomethyS, N-{\,2-dihydroxypropyl)-aminomethyl, AA(l-amino-2-hydroxy-prop-3-yl)-aminomethyl, Ar-(A^-ethoxycarbonyl~piperidin-4-yS)-aminomethyl, jy_(A/„benzylpiperidin-4-yl)-aminomethyl, Af-(homopiperidin-3-yl)-aminomethyl, A-(j¥-benzvlpyrrolidin-3-yl)-aminomethyl, A?-(¥-ethylpiperidin-'3”yl}aminoTnethy1, 2,2,2-trifluoroethylaminomethyl, 3,3,3-trifluoropropyiaminomethyl, 2,2,3,3,3-pentafluoropropylaminomethy 1, -CH2N(CH2CH20H)2, -CH2N(CH3)(CH2CH20H), -CH2NH(CH2CH2OH), -CH2NH(CH2CH2CH2CH20H), -CH2N(CH3)CV-methyl-pyrrolidin-3-yl), -CH2NH(C(CH3)2CH2QH), -NHC(0)CH(CH3)2, -NHC(0)CH2N(CH2CH3)2, ~NHC(G)CH2NH(CH3), ~NHC(0)H, -NHC(0)CH2CH(0H)CH20H, -NHC(0)CH2NH2, -NHC(0)CH2N(CH2CH20H)2, -NHC(0)CH2CH2N(CH2CH20H)2, -NHC(0)CH2(4-alkyl-piperazinyl), -NHC(0)CH2(piperidiny l), jV-(phenyloxyethyl)-aniinomethyl, cyclopentyl, 1-amino-cyclopentyl, (cis, trarts')-!-amino-cyclopentyl, (cis, /ram)-2-amino-cyclopentyl, cis-2-amino-cyclopentyl, tram-2-amino-cyclopentyl, (cis,tram)-2-hydroxy-cyclohexyl, cis- 2-hydroxy-cyclohexyl, tram-2-hydroxy-cyclohexyl, (cis, lram)-2-amino-cyclohexyl, c/5-2-amino-cyclohexyl, tram-2-amino-cyclohexyl, azetidin-3-yl, pyrrolidinyl, N-alkyl-pyrrolidinyl, 3-(dialkylamino)-pyrrolidinyl, piperidinyl, 2-methyl-piperidin-6-yl, iV-iert-butoxycarbonylpiperidin-2-yl, piperazinyl, -CH2NHC(0)CH3, -CH(CH3)NHC(0)CH3, -CH(CH3)NHC(0)C(0CH3)(CF3)phenyl, pyrrol-l-yl, pyrrol- 2-yl, pyrrol-3-yl, imidazol-l-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, N-methyl-imidazol-2-yl, 5-methyl-imidazol-2-yl, l,2,4-triazol-3-yl, thiazol-2-yl, 2-aminopyrimidin-3-yl, pyridinyl, benzimidazolyl, imidazol-l-ylmethyl, imidazol-2-ylmethyl, triazolylmethyl, (5-amino-3-methylpyrazol-l-yl)-methyl, phenoxymethyl, methylsulfonylaminomethyl, 1 -(methoxycarbonylamino)-ethyl, 1 -amino-1 -phenyl-methyl, or l-amino-3-hydroxy-propyl.
[00188] In another embodiment of embodiment A10, the Compound of Formula I is that where X and R7 are halo; A is phenylene optionally substituted with R10 and Ri2 where R10 and R12 are independently hydrogen or halo; R1, R2, Rs and R6 are hydrogen; and R3 is hydrogen, halo, hydroxy, alkoxy, or amino.
[00189] In another embodiment of embodiment A10, the Compound of Formula I is that where R3 is hydrogen and R4 is a) hydrogen; b) ~NR8R8 (where Rs and R8 are independently hydrogen; alkyl; alkenyl; alkyl substituted with one or two hydroxy; alkyl substituted with one or two -NR30R30 where R-50 and R30 are independently hydrogen, alkyl, or hvdroxyalkyl; alkyl substituted with optionally substituted heteroaryl; or alkyl substituted with optionally substituted cycloalkyl); c) ~C(0)NR8R8 (where R8 is hydrogen, alkyl, or alkenyl; and R8 is hydrogen; hydroxy; alkyl; alkenyl; alkyl substituted with one or two hydroxy; alkyl substituted with heterocycloalkyl; alkyl substituted with -NR30Rj0 where R30 and R30 are independently hydrogen, alkyl, or hydroxyalkyl; or optionally substituted alkoxy); d) -NR8C(0)0R8 (where R8 and R8 are independently hydrogen, alkyl, or alkenyl); e) -NR8C(0)R8 (where R8 is hydrogen, alkyl, or alkenyl; and R8 is hydrogen; alkyl; alkyl substituted with one or two hydroxy; alkyl substituted with optionally substituted heterocycloalkyl; alkyl substituted with -NR30R'>0 where R30 and R30 are independently hydrogen, alkyl, hydroxyalkyl, or alkenyl); f) alkyl; g) alkyl substituted with one or two -OR (where R is hydrogen); h) alkyl substituted with -NR8R8 (where K8 is hydrogen, alkyl, alkenyl, alkynyl, or alkyl substituted with one or two hydroxy; and R8 is hydrogen; alkyl; alkenyl; alkynyl; alkyl substituted with one or two hydroxy; heterocycloalkyl substituted with alkyl; or alkyl substituted with -NR30R30 where R30 and R30 are independently hydrogen, alkyl, or hydroxyalkyl); i) heterocycloalkyl; or j) heterocycloalkyl substituted with -NRSR8 (where R8 and R8 are independently hydrogen, alkyl, or alkenyl).
[00190] In another embodiment, R3 is hydrogen and R4 is hydrogen, hydroxymethyl, -NH2, -NH(CH2)3CH3, -NHCH3, -NH(CH2CH3), -NHCH2CH(CH3)2, -NHCH2CH2OH, -NHCH2CH2CH2NH2, -N(CH3)CH2CH2(pyridin-2-yl), -NHCH2(cyclopropyl), -NHCH2(cyclopentyl), -NHCH2(cyclohexyl), -C(0)NH0H, -C(0)NH(0CH2CH(0H)CH20H), -C(0)NH(CH2)3CH3, -C(0)NHCH2CH=CH2, -C(0)NHCH2CH3, -C(0)NHCH2CH20H, -C(0)NHCH2CH(0H)CH20H, -C(0)NHCH2CH2CH(0H)CH20H, -C(0)NHCH2CH2(piperidin-l -yl), -C(0)NH (phenyl), -C(0)NHCH2CH2N(CH2CH3)2, ^-(isopropyl)-aminomethyl, jV,Ar-dimethylaminomethyl, A/-(2-aminoethy 1 )-aminomethyl, -NHC(0)0C(CH3)3, -NHC(0)0CH3, -NHC(0)CH(CH3)2, -NHC(0)CH2NH2, -NHC(0)CH2N(CH2CH3)2, -NHC(0)CH2NH(CH3), -NHC(0)H, -NHC(0)CH2CH(0H)CH20H, -NHC(0)CH2N(CH2CH2OH)2,-NHC(0)CH2CH2N(CH2CH2OH)2, -NHC(0)CH2(4-alkyl-piperazinyl), -NHC(0)CH2(piperidinyl), pyrrolidinyl, 3-(dialky!amino)-pyrrolidinyI, piperidinyl, 2-methyl-piperidin-6-yl, iV-methylpiperidin-2~yl, or piperazin-2-yl.
[00191] In another embodiment of embodiment A10, the Compound of Formula I is that where R3 is alkoxy and R4 is alkyl substituted with -NR8R8 (where R8 and R8 are independently hydrogen, alkyl, or alkenyl). In another embodiment, R3 is methoxy and R4 is alkyl substituted with -NRSR8’ (where R8 and R8 are independently hydrogen, alkyl, or alkenyl).
[00192] In another embodiment of embodiment A10, the Compound of Formula I is that where R3 is halo and R4 is alkyl substituted with -NR8R8 (where R8 and R8 are independently hydrogen, alkyl, or alkenyl). In another embodiment, R3 is fluoro and R4 is alkyl substituted with -NR8R8 (where R8 and R8 are independently hydrogen, alkyl, or alkenyl).
[00193] In another embodiment of embodiment A10, the Compound of Formula I is that where R3 is amino and R4 is alkyl substituted with -NR8R8 (where R8 and R8 are independently hydrogen, alkyl, or alkenyl).
[00194] In another embodiment of embodiment A10, the Compound of Formula I is that where R3 is hydroxy and R4 is a) hydrogen; b) -CH2N(R25)(NR25aR25b); c) -CH2NR25C(-NH)(NR25aR25b); d) -CH2NR25C(-NH)(N(R25a)(NQ2); e) -CH2NR25C(-NH)(N(R25aXCN); f) -CH2NR25C(=NH)(R25); g) -CH2NR25C(NR25aR25b)=CH(NG2); h) alkyl; i) alkenyl; j) alkyl substituted with one or two -OR8 where R8 is hydrogen, aryl, or alkyl where the alkyl is substituted with one or two hydroxy; k) alkyl substituted with one, two, or three halo; l) alkyl substituted with nitro; m) alkyl substituted with -S(0)mR9 (where m is 0 and R9 is aryl); n) alkyl substituted with optionally substituted heterocycloalkyl; o) alkyl substituted with -NR8R8 (where R8 is hydrogen, alkyl, alkenyl, alkynyl, or alkyl substituted with one or two hydroxy; and R8 is hydrogen; hydroxy; alkoxy; alkyl; alkenyl; alkynyl; optionally substituted alkoxy; alkyl substituted with one or two hydroxy; alkyl substituted with ~NR30R30 where R30 and R30 are independently hydrogen, alkyl, or hydroxyalkyl; alkyl substituted with one or two hydroxy and one or two -NR30R30 where R30 and R30 are independently hydrogen, alkyl, or hydroxyalkyl; heterocycloalkyl substituted with alkyl, aikoxycarbonyl, or optionally substituted arylalkyl; alkyl substituted with one, two, three, four, or five halo; alkyl substituted with optionally substituted cycloalkyl; alkyl substituted with optionally substituted aryl; alkyl substituted with one or two hydroxy and one optionally substituted aryl; alkyl substituted with optionally substituted heterocycloalkyl; alkyl substituted with optionally substituted heteroaryl; heteroaryi: aryl; aryl substituted with one or two hydroxy; aryl substituted with one or two alkoxy; aryl substituted with one or two halo; aryl substituted with one or two -NR32C(0)R3za where R32 is hydrogen or alkyl and Rj2a is alkyl, alkenyl, alkoxy, or cycloalkyl; aryl substituted with ~NR34S(>2R34a where R34 is hydrogen or alkyl and R34a is alkyl, alkenyl, cycloalkyl, aryl, heteroaryi, or heterocycloalkyl; cycloalkyl; cycloalkyl substituted with one or two hydroxy; cycloalkyl substituted with one or two hydroxy and one or two hydroxyalkyl; cycloalkyl substituted with one or two alkoxy; cycloalkyl substituted with carboxy; cycloalkyl substituted with -C(0)NR33R33a where R33 is hydrogen or alkyl and R3ja is alkyl, alkenyl, alkynyl, or cycloalkyl; cycloalkyl substituted with optionally substituted cycloalkyl; heterocycloalkyl; heterocycloalkyl substituted with one or two hydroxy; heterocycloalkyl substituted with one or two alkoxy; heterocycloalkyl substituted with one or two hydroxyalkyl; heterocycloalkyl substituted with one or two hydroxy, one or two alkoxy, and one or two hydroxyalkyl; alkyl substituted with -C(0)NR33R33a where Rj3 is hydrogen or alkyl and R3ja is alkyl, alkenyl, alkynyl, or cycloalkyl; alkyl substituted with optionally substituted aryloxy; alkyl substituted with ~S(Q)nR3i where n is 0 and R31 is alkyl; alkyl substituted with carboxy; alkyl substituted with aikoxycarbonyl; or alkyl substituted with -NR32C(0)R32a where R32 is hydrogen or alkyl and R32a is alkyl, alkenyl, alkoxy, or cycloalkyl); p) heterocycloalkyl; q) -C(0)NR8R8 (where R8 is hydrogen, alkyl, or alkenyl; and R8 is hydrogen; alkyl; alkyi; alkenyl; or substituted with one or two hydroxy;); r) alkyl substituted with -NRSC(G)R8 (where R8 is hydrogen, alkyl, or alkenyl and R8 is alkyl; alkenyl; or alkyl substituted with alkoxy, aryl, and one, two, or three halo); s) cycloalkyl; t) cycloalkyl substituted with -NR8R8 where R8 and R8 are independently hydrogen, alkyl, or alkenyl; u) cycloalkyl substituted with -C(G)NR33R33a where R33 is hydrogen or alkyl and Rj3a is alkyl, alkenyl, alkynyl, or cycloalkyl; v) heterocycloalkyl; w) heterocycloalkyl substituted with one or two alkyl; x) heterocylcloalky! substituted with -C(0)OR8 where R8 is alkyl or alkenyl; y) heteroaryl; z) heteroaryl optionally substituted with -NR8R8 where R8 and R8 are independently hydrogen, alkyl, or alkenyl; aa) alkyl substituted with optionally substituted heieroaryl; bb) alkyl substituted with -NR8S(0)2R9 where R8 is hydrogen, alkyl, or alkenyl and R9 is alkyl or alkenyl; cc) alkyl substituted with -NR8C(G)OR8 where R8 and R8 are independently hydrogen, alkyl, or alkenyl; dd) alkyl substituted with one aryl and one -NR8R8 where R8 and R8 are independently hydrogen, alkyl, or alkenyl; or ee) alkyl substituted with one or two -OR8 (where R8 is hydrogen) and one or two -NR8R8 where R8 and R8 are independently hydrogen, alkyl, or alkenyl. {00195] In another embodiment, RJ is hydroxy and R4 is hydrogen, -CH2N(H)(NHCH3), -CH2NHC(=NH)(NH2), -CH2NHC(=NH)(NHN02), -CH2NHC(=NH)(NHCN), ~CH2NHC(=NH)(phenyl), -CH2NHC(NH2)=CH(NG2), methyl, ethyl, hydroxymethyl, 2,3-dihydroxypropy 1, 3-hydroxy-2-methyl-prop-2-yl, jV-(l-methoxy-prop-2-yl)-aminomethyl, iV-(ethoxypropyl)-aminomethyl, ^-(ethoxy ethyl)-aminomethyl, //-(2,2-dimethoxyethyl)-aminomethyl, N-(methoxyethyl)-aminomethyl, //-(isopropxyethyl)-aminomethyl, trifluoromethyl, 1-nitro-ethyl, 1-methyl-1-nitro-ethyl, 1-nitro-propyl, 3-methyl-l -nitro-butyl, phenylthiomethyl, allyl, ethenyl, 2-methylthio-ethylaminomethyl, 3-methyithio-propylaminomethyl, iV-(/er/-butoxycarbonylaminopropyl)-aminomethyl, N-(\-carboxyethyl)-aminomethyl, N-( 1 /Acarboxyethyl)-ammomethyl, ¥-(15-carboxyethyl)-aminomethyl, ¥-(l-methoxycarbonylethyl)-aminomethyl, azetidinylmethyl, pyrrolidinylmethyl, 3-hydroxy-pyrroiidinylmethyl, 2-(methoxymethyl)-pyrrolidinylmethy!, 25-(methoxymethyl)-pyrrolidinylmethyl, 2R-(methoxymethy I)-pyrrol idinylmethyl, morpholinylmethyl, 4-hydroxypiperidinylmethyi, 4-methyl-piperazinylmethyl, 4-methyl-homopiperaziny lmethyl, 4-(piperidinyl)-piperidinylmethyl, 4-[2-(Ar,.¥-diethylamino)-ethyl]-piperazinylmethyi, ¥-hydroxyaminomethyi, ¥-methoxyaminomethyl, ¥-efhoxy aminomethyl, ¥-ethylami nomethyl,1 -(¥-ethyl-amino)-ethyl, ¥,¥-diethylaminomethyl, N, ¥-dimethylaminomethy 1, aminomethyl, 1-amino-ethyl, 1 ^-amino-ethyl, 15-amino-ethyl, 1 -(methylamino)-ethyl, 1 -(¥, ¥-dimethy 1 amino)-ethyl, 1 -amino-1 -methyl-ethyl, 1-aminopropyl, 15-aminopropyl, 1 /?-aminopropyl, ¥-(n-propyl)-aminomethyl, ¥-(isopropyl)-aminomethyl, 2-{¥-isopropyiamino)-ethyl, 3-(¥-isopropylamino)-2-methyl-prop~2-yl, 1 -(TV-ethyl-amino)-propyl, 1 -(¥,¥-diethyl-amino)-propyl, 1-aminobutyl, 1-amino-isobutyl, ¥-(n-butyl)~aminomethyl, A-isobutylaminomethyl, ieri-butyl aminomethyl, 1 -(ter/-butylamino)-ethyl, sec-butylaminomethyl, Ar-(2-methyl-but-3-yl)-aminomethyl, ¥-(3,3-dimethyl-butyl)-aminomethyl, ¥-(3-methylbut-3-yl)-aminomethyl, ¥-(2-methylbutyl)-aminomeihyl, ¥-(pent-3-yl)-aminomethyl, n-pentylaminomethyl. isopentylaminomethyl, seopentylaminomethyl, neopentylaminomethyl, ¥-(2,2,4-trimethyi-pent-4-yl)-aminomethyl, ¥-(2-ethyl-butyl)-aminomethyl, ¥-al!yi-aminomethyl, 3-methyl-but-l-yn-3-ylaminomethyl,¥-(2,3-dihydroxypropy!oxy)-aminomethyl, iV-cyclopropylaminomethyl, ¥-cyclopentyl aminomethyl, ¥-cyclopenten~4-ylaminomethyl, ¥-(l(i?,5)-hydroxy-cyclopeni-2-yi)-aminomethyl, ¥-(15-hydroxy-cyclopent-2-yl)-aminomethyl, ¥-( 1 i?-hydroxy-cyclopent-2-yl)-aminomethy 1, ¥-(1 (R, S)-hydroxy-1 ~methyl-cyelopent-2-yl)-aminomeihyl, ¥-(15-hydroxy-1 -methy 1-cyclopent-2-yl)-aminomethyl, ¥-( 1 ^-hydroxy-1 -methyl-cyclopent-2-yl)-aminomethyl, ¥-(3,4-dihydroxy-cyclopentyl)~aminomethyl, ¥-( 1 -hydroxymethyl -cyclopent- l-yl)-aminomethyl, ¥-(2,3-dihydroxy-4-hydroxymethyI-cvclopentyi)-aminomethyl, ¥-(l(i?J5)-methoxy-cyciopent-2-yl)-aminomethyl, ¥-(15-methoxy-cyclopent-2-yl)-aminomethyl, ¥-(l i?-methoxy-cyclopent-2-yl)-aminomethyl, ¥-(l -carboxy-cyclopentyl)-aminomethyl, ¥-cyc lohexy 1 aminomethyl, ¥-( 1 (R, 5)-hydroxy-cyclohex-2-yl)-aminomethyl, ¥-(l(i?)-hydroxy-cyclohex-2-yi)-aminomethyi, //-( 1 (S)~hydroxy-cycloheX“2-yl)-aminomethyl, iV-(c/5-4-hydroxy-cyclohexyl)-aminomethyl, A-(/raMs-4-hydroxy-cycIohexyl)-aminomethyI, 1 -[AA(cw-4-hydroxy-cyclohexyl)-amino]-ethyl, l-[//-(iraHs-4-hydrQxy~cyclohexyl)-amino]-ethyl, N~( 1 -hydroxymethyi~cyclohexyl)"aminomethyl, //-(2-cyclohexyl-cyclohexyl)-aminomethyl, A^(2/^3S,4J?,6i?)-2-(hydiOxymethyl)-3,4-dihydroxy~6-methoxy-tetrahydro-2//-pyran-5-yl} -aminomethyl, //-(cycloheptyl)-aminomethyl, Ar-(cyclooctyl)-aminomethyI, [(1 r,3r,5/?,7/7)-tricyclo[3,3.1.13'7]dec-2-yiaminojmethyL, //-(1-benzyIoxy-cyclopent-2-yl)-aminomethyl, N- [ 1 -(cyclopropylaminocarbonyl)-cycl opentylj-aminomethyl, -CH2NHC(CH3)2C(0)NH(cyclohexyl), -CH2NHC(CH3)2C(0)NH(CH2CH3), iV-(cyclopropylmethyl)-aminomethyl, //-(cyclohexySmethyl)-aminomethyl, //-(l-cyclohexylethyl)-aminomethyl, A^imidazolyl)-aminomethyl, //-( 1,3,5-triazinyl)-aininomethyl, //-(5-hydroxy-pyrazol-3-yl)-aminomeihyl, Ar-(5-meihyl-pyrazoI-3-yl)-aminomethyl, A^-(benzimidazolyl)-aminomethyl, //-(pynmidi n-2-y l)-aminomeihyl, N-(pyridin-2-y 1)-aminomethyl, A^Cpyridin-S-yO-aminomethyl, //-(pyridin-4-yl)-aminomethy 1, //-indan- 1-yl-aminomethyl, //~indan-2-yl-aminomeihyi, phenyl aminomethyl, //-(2-hydroxyphenyl)-aminomethyl, Af-(3-hydroxyphenyl)-aminomethyi, Ar-(4-hydroxyphenyl)-aminomethyl, Ar-(2-methoxyphenyl)-aminomethyl, //-(3 -methoxyphenyl)-aminomethyl, //-(4-methoxyphenyl)-aminomethyl, //-(2-fluorophenyl)-aminomethyl, //-(3-fluoropheny!)-aminornethyl, jV-(4-fluorophenyl)-aminomethyl, //-(2-chlorophenyl)-aminomethyl, //-(3-chlorophenyl)-aminomethyl, N-(4-chlorophenyl)-aminomethyI. Af-(3-methylcarbonylammo-phenyl)-aminomethyl, N-(4-methylcarbonylamino-phenyl)-aminomethyl, //-(2-aminophenyl)-aminomethyl, //-(3-aminophenyl)-aminomethyl, A-(4-aminophenyl)-aminomethyl, Ai-(2-methylsulfonylaminophenyl)-aminomethyl, //-(3-methylsulfonylaminophenyl)-aminomethyl, //-(4-methyisuifonylaminophenyl)-aminomethyl, //-(2-fluoro-4-hydroxy-phenyl)-aminomethyl, Ar-(3-fluoro-4-hydroxy-phenyl)-aminomethyl, //-(benzyl)-aminornethyl, //-(2-hydroxyphenylmethyl)-aminomethyl, //-(3-hydroxyphenyImethyl)-aminomethyl. //-(4-hydroxyphenylmethyl)-aminomethyl, N-(2-(//-methylpiperazin-l-yl)-phenylmethyl)-aminomethyl, //-(4-methyl-phenethyl)-aminomethyl, //-(1 -hydroxy-3 -phenyl-prop-2-yl)-aminomethy 1, A7-(pyrrolidin-2-ylmethyl)-aminomethyl, //-(A/-ethyl-pyrrolidinylmethyl)-aminomethyl, //-(//-methyl-pyrrolidin-2-ylethyl)-aminomethyl, //-(pyrrolidinylpropyl)-aminomethyl, //-(1,1 -dimethyl-2-pyrro!idin-1 -yl-ethy!)-aminomethyl, //-(tetrahydrofuranylmethyl)-aminomethyl, //-(tetrahydro-2i/-pyran-4-ylmethyl)-aminomethyl, A^(tetrahydro-2//-pyranvlethyl)-aminomethyl, JV-(piperidin-4-y lmethyl)-aminomethyl, N~(N~ methylpiperidin-4~ylmethyl)-aminomethyl, Af~(A'-ierf-butoxycarbonylpiperidm- 4-yImethyl)-aminomethyi, Af-(AAmethy!imidazol~5-ylniethyl)-aminomethyl, N-(N-methylimidazol-4-ylmethyl)-aminomethyl, Af-[2-(imidazol-4-yl)~ethyl]-aminomethyl, iV-[3-(imidazolyl)-propyi]-aminomethyL iV-(pyridin-3-yiethyl)-aminomethy 1, N-(pyridin-4-yiethyl)-aminomethyl, //-(thien-2-ylethyl)-aminomethyl, //-(furan-2-ylethyl)-aminomeihyi, Ar-(5-methyl-1,3,4-oxadiazol-2-yhnethyl)-aminomethyl, N~( 2-indolin-3-ylethyi)-aminomethyl, 2-(Αζ Amimethylamino)-ethylaminomethyl, 2- (JV, Af-dimethylamino)-l-methyl-eihylaminomethyl, 3-aminopropylaminomethyi, 3- (Λζ AAdimethylamino)-propylaminomethyl, S-CA^/Z-diethylamino)-propyl aminomethyl, N-(N, A'-diisopropylaminoethyl)-aminomethyl, N-(N, N-dimethylaminobutyl)-aminomethyl, 3-hydroxypropylaminomethyl, //-(1,2-dihydroxypropyl)-aminomethyl, N-(l -amino-2-hydroxy~prop-3-yl)-aminomethyl, N-(Ar-ethoxycarbonyl-piperidin-4-yl)-aminomethyl, //~(//-benzylpiperidin~4~yl)-aminomethyl, //-(homopiperidin-3-yl)-aminomethy 1, //-(//-benzylpyrrolidin-3-yl)-aminomethyl, //-(//-ethylpiperidin-3-yl)aminomethyl, 2,2,2-trifluoroethylaminomethyl, 3,3,3 -trifluoropropylaminomethy 1, 2,2,3,3,3-pentafluoropropylaminomethyl, -CH2N(CH2CH2OH)2, -CH2N(CH3)(CH2CH20H), -CH2NH(CH2CH2OH), -CH2NH(CH2CH2CH2CH2OH), -CH2NH(C(CH3)2CH2OH), -CH2N(CH3)(//-methyl-pyrrolidin-3-yl), -C(0)NH2, -C(0)NHCH2CH=CH2, -C(0)NHCH2CH(OH)CH2OH,;V~(phenyloxyethyl)-ammomethyl, -CH2NHC(0)CH3, -CH(CH3)NHC(0)CH3, -CH(CH3)NHC(0)C(0CH3)(CF3)phenvl, cyclopentyl, 1-amino-cyclopentyl, (cis, iram)-2-amino-cyclopentyl, {cis, trans)-2-amino-cyclopentyl, cw-2-amino-cyclopentyl, /rcr«i-2-amino-cyclopentyl, (cis, trans)-2-hydroxy-cyclohexyl, cis-2-hydroxy-cydohexyi, /nm?-2-hydroxy-cyclohexyl, (cis,trans)-2-amino-cyclohexyl, cis-2-amino-cyclohexyl, irans~2-amino-cyclohexyl, azetidin-3-yl, pyrrolidinyl, iV-methyl-pyrrolidin-2-yl, //-ethyl-pyrrolidin-2-yi, 3 -(dimethylamino)-pyrrolidinyl, piperidinyl, 2-methyl-piperidin-6-yl, A/-methylpiperidin-2-yl, N-tert-butoxycarbonylpiperidin-2-yl, piperazin-2-yl, pyrrol- 1-yl, pyrrol-2-yl, pyrrol~3~yl, imidazol-l-yl, imidazol-2-yl, imidazol-4-yl, imidazol-5-yl, A^-methyl-imidazol-2-yl, 5-methyl-imidazol-2-yl, l,2,4-triazol-3-yl, thiazol-2-yl, 2-aminopyrimidin-3-yl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, benzimidazolyl, imidazol-1 -ylmethyl, imidazol-2-ylmethyi, triazol-1 -ylmethyl, (5-amino-3-methyl-pyrazol-3-yl)-methyl, phenoxymethyl, 2-hydroxyethyloxymethyl, methylsulfonylaminomethyl, 1- (methoxycarbonylamino)-ethyl, 1-amino-1-phenyl-methyl, or 1 -amino-3-hydroxy-propyl.
[00196] Another embodiment of the Invention (A11) is that where the compound of Formula I is selected from Group A where R3 and R4 together with the carbon to •j which they are attached form C(O) or C(=NOH). In another embodiment, X and R are halo; A is phenylene optionally substituted with R10 and Rf2 where R10 and Ru are independently hydrogen or halo; R1, R2, R5 and R6 are hydrogen; and R3 and R4 together with the carbon to which they are attached form C(O) or C(=NOH).
[00197] Another embodiment of the Invention (A12) is that where the compound of Formula I is selected from Group A where X and R7 are halo; A is phenylene optionally substituted with R10 and R12 where R10 and R12 are independently hydrogen or halo; and R1, R2, R4, R5 and R6 are hydrogen.
[00198] Another embodiment of the Invention (A 13) is that where the compound of Formula I is selected from Group A where A is phenylene.
[00199] Another embodiment of the Invention (A 14) is that where the compound of Formula I is selected from Group A where R1 is hydrogen and R2 is alkyl substituted with -NR8R8 where R8 and R8 and all other groups are as defined in the Summary of the Invention for a compound of Group A.
[00200] Another embodiment of the Invention (A15) is that where the compound of Formula I is selected from Group A where A is phenylene; R7 is iodo or bromo; X is fluoro or chloro; and R!, R2, R5, and R6 are hydrogen; and R10, Ri2, R14, and Ri6 are independently hydrogen or fluoro. In another embodiment, Ri0 is 3-fluoro and R!2, R14, and R16 are hydrogen or halo; R10 is 3-fluoro, Ri2 is 4-fIuoro, and R14 and R16 are hydrogen; Ri0 is 4-fluoro, R12 is 5-fluoro, and R14 and Rs6 are hydrogen; R10 is 4-fluoro, R12 is 6-fluoro, and R14 and Ri6 are hydrogen; or Ri2 is 4-fluoro and Ri0, R14, and R!6 are hydrogen.
[00201] In another embodiment of the invention is a compound of Formula selected form Group A where RJ is hydroxy and R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloaikyl where the cycloalkyl is optionally substituted with groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, RJ is hydroxy and R4 is heterocycloalkyl or alkyl, where the alkyl is optionally substituted with -NR R (where R is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkvl where the cycloalkyl is optionally substituted with groups independently selected from hydroxy and alkyl), [00202] In another embodiment of the Invention (Bl) the compound of Formula 1 is selected from Group B where all groups are as defined in the Summary of the Invention.
[00203] In another embodiment of the invention (B2), the Compound of Formula 1 is that where X and R7 are halo; and all other groups are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, X is fluoro or chioro and R7 is iodo or bromo.
[00204] In another embodiment of the invention (B3), the compound of Formula I is selected from Group B where R3 is halo, nitro, -NR8R8 , -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(G)GR8, -C(0)NRsR8 , -NRsC(0)OR8, -NR8C(0)NR8R8” -NR8C(0)0R8’, -NR8C(0)R8’,-CH2N(R25)(NR25aR25b), -CH2NR25C(=NH)(NR25aR25b), -CH2NR25C(-NH)(N(R25a)(N02), -CFl2NR25C(“NH)(N(R25a)(CN), -CH2NR25C(=NH)(R25), -CFf2NR25C(NR25aR25b)==CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkvl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NR8R8, ~NR8S(Q)2R.5, -CN, ~S(0)mR;:', -C(Q)RS, -C(Q)OR8, -C(0)NR8R8', -NR8C(0)NR8 R8 \ -NR8C(0)0R8 and -NR8C(0)R8 and R4 is as defined in the Summary of the Invention; or R3 and R4 together with the carbon to which they are attached form C(O) or C(-NOH); and all other groups are as defined in the Summary' of the Invention for a compound of Group B. In another embodiment, Rl, R2, R3 and R6 are hydrogen; and X and R7 are halo.
[00205] In another embodiment of the invention (B4), the compound of Formula I is selected from Group B where R3 and R4 are independently halo, nitro, -NRSR8 , -OR8, -NHS(Q)2R8, -CN, -S(0)mR8i -S(0)2NR8R8', -C(0)R8, -C(Q)OR8, -C(0)NR8R8', -NR8C(0)0R8’, -NR8C(0)NR8 R8’’ -NR8C(0)0R8’, -NR8C(0)R8, -CH2N(R25)(NR25aR25b),-CH2NR25C(==NH)(NR25aR25b), -CH2NR25C(-NH)(N(R25a)(N02), -CH2NR25C(-NH)(N(R25a)(CN), -CH2NR2SC(=NH)(R2S), -CH2NR25C(NR25aR25b)-CH(N02)i alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryi, optionally substituted heteroarylalkyl, -OR8, -NR8R8’, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)NR8 R8”, -NR8C(0)0R8’ and -NR8C(0)R8’; or R3 and R4 together with the carbon to which they are attached form C(O) or C(=NOH); and all other groups are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, R1, R2, R5 and R6 are hydrogen; and X and R7 are halo.
[00206] In another embodiment of the in vention (B5), the Compound of Formula I is that where A is heteroarylene selected from thien-diyl, benzo[</]isoxazol-diyI, benzo[c/]isothiazol-diyl, l/f-indazol-diyl (optionally substituted attheNl position with R19 where Ri9 is as defined in the Summary of the Invention for a compound of Group B), benzo\d\oxazol-diy 1, benzo[i/jthiazol-diyl, l//-benzo[i/]imidazol-diyl (optionally substituted at the N1 position with Ri9 where R19 is as defined in the Summary of the Invention for a compound of Group B), I//-benzo[ifj[l,2,3]triazol-diyl (optionally substituted at the N1 position with R19 where R19 is as defined in the Summary of the Invention for a compound of Group B), imidazo[l,2-a]pyridin~diyl, cinnolin-diyl, quinolin-diyl, pyridin-diyl, 1 -oxido-pyridin-diyl, [l,2,4]triazolo[4,3-a]pyridin-diyl, and 2,3-dihydroimidazo[l,2-a]pyridin-diyl; and A is further optionally substituted with one, two, three, or four groups selected from R10, R12, Ri4, and R16 where R10, Ri2, R.14, and R16 and all other groups are as defined in the Summary of the Invention for a compound of Group B. In another embodiment A is selected from thien-3,4-diyl, benzo[i/]isoxazol-5,6-diyl, benzo[if]isothiazol-5,6-diyl, lfT-indazol- 5.6- diyl (optionally substituted at the N1 position with R!9 where R19 is alkyl or alkenyl), benzo[i/]oxazol-5,6-diyl, benzo [ifjthiazol-5,6-diyl, 1 //-benzo[d]imidazoi- 5.6- diyl (optionally substituted at the N1 position with R19 where R.19 is alkyl or alkenyl), l/f-benzo[i/][l,2,3]triazol-5,6-diyl (optionally substituted at the N1 position with R19 where Ri9 is alkyl or alkenyl), imidazo[l,2-a]pyridin~5,6-diyl, cinnolin-6,7-diyl, quinolin-6,7-diyl, pyridin-3,4-diyl, 1 -oxido-pyridin-3,4-diyl, [ 1,2,4]triazolo[4,3-a]pyridin-6,7-diyl, and 2,3-dihydroimidazo[l,2-a]pyridin-6,7-diyl. |00207] In another embodiment of the Invention (B6), the compound of Formula I is selected from Group B where A is thien-diyl and X, R\ R", R3, R4, R5, R6, R7, Ri0, and R12 are as defined in the Summary of the Invention for a compound of Group B. In another embodiment A is thien-3,4-diyl; R10 and R12 are hydrogen; X and R7 are halo; and R1, R2, R5, and R6 are hydrogen. In another embodiment, X is fluoro or ehloro; R7 is iodo or bromo; R3 is hydrogen or hydroxy; and R4 is -NR8R8 (where R8 and R8 are independently hydrogen or alkyl), heterocycloalkyl, heteroaryl (optionally substituted with alkyl), or alkyl where the alkyl is optionally substituted with -NR R (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl).
[00208] In another embodiment (B7), the compound of Formula 1 is more specifically according to Formula 1(c) or 1(d)
1(c);
1(d); where X, R1, R2, R3, R4, R5, R6, R7, R10, R12 and R14 are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, R1, R2, R5, and R6 are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; and R50, R12, and R14 are independently hydrogen, halo, or alkyl. In another embodiment, X is fluoro or ehloro and R7 is iodo or bromo; R10 is hydrogen or halo, in another embodiment hydrogen or fluoro; R12 is hydrogen; R14 is hydrogen or alkyl; and R3 is hydroxy. In another embodiment, R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR R (where R is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloaikyi where the cycloalkyl is optionally substituted with groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pvrrolidinyl, 1 (R,S)-amino-ethy 1,1 (R)-amino-ethyl, 1(^-amino-ethyl, 1 (R, S)-(methylamino)-ethyl, 1 (i?)-(methylamino)-ethyl, 1 (S)-(methylamino)-ethyl, 1 (R, S)-(dimethylamino)-ethyl, 1 (i?)-(dimethylamino)-ethyl, 1 (S)-(dimethylamino)-ethyl, 1 (R, S)-(3,4-ci.?-dihydroxy-cyclopentylamino)-ethyl, 1 (J?)-(3 ,4-cB-dihydroxy· cyclopentylamino)-ethyl, or 1 (5)-(3,4-c/s-dihydroxy-cyclopentylamino)-ethyl.
[00209] In another embodiment of the Invention (B8), the compound of Formula I is more specifically according to Formula 1(e) or 1(f):
1(e);
1(f); where X, R1, R2, R3, R4, R5, R6, R7, Ri0, R12 and R14 are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, R1, R2, R5, and R6 are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; and R10, R12, and R54 are independently hydrogen, halo, or alkyl. In another embodiment, X is fluoro or chloro and R7 is iodo or bromo; Ri0 is hydrogen or halo, in another embodiment hydrogen or fluoro; R12 and R14 are hydrogen; R3 is hydroxy; and R1 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NRSR8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl.
[00210] In another embodiment of the Invention (B9), the compound of Formula I is in another embodiment according to Formula 1(g) or 1(h):
i(g);
i(h); where X, R1, R2, R3, R4, R5, R6, R7, R10, R12, R14, and R19 are as defined in the Summary of the Invention for a compound of Group B, 100211] In another embodiment of embodiment B9, the compound of Formula I is more specifically according to Formula 1(g) or 1(h) where R3 is halo, nitro, -NR8R8’, -OR8, -NHS(0)2Rs, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)Rs, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)GR8’, -NR8C(0)NR8'R8” -NR8C(0)0R8’, -NR8C(G)R8’, -CH2N(R25)(NR25aR25b), -CH2NR25C(~NH)(NR25aR25b), -CH2NR25C(-NH)(N(R2Sa)(N02), -CH2NR25C(=NH)(N(R25a)(CN), -CH2NR25C(=NH)(R25), -CH2NR25C(NR25aR25b)~CH(N02), cycloalkvl, heteroaryl, or heterocycloalkyl; where the cycloalkyl, heteroaryl, and heterocycloalkyl are optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NR8R8’, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)OR8, -C(0)NR8R8’, -NR8C(0)NR8’R8”, -NR8C(0)0R8’ and -NR8C(0)R8; and R4 is as defined in the Summary of the Invention; or R3 and R4 together with the carbon to which they are attached form C(Q) or C(=NOH); and ail other groups are as defined in the Summary' of the Invention for a compound of Group B.
[ΘΘ212] In another embodiment of embodiment B9, the compound of Formula I is more specifically according to Formula 1(g) or 1(h) where R3 is hydroxy and all other groups are as defined in the Summary of the Invention for a compound of Group B. {00213] In another embodiment of embodiment B9, the compound of Formula I is more specifically according to Formula 1(g) or 1(h) where R , R , R , and R are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; Ri0, R*2, and Ri4 are independently hydrogen, halo, or alkyl; and R19 is hydrogen or methyl. In another embodiment, X is fluoro or chloro and R7 is iodo or bromo; R!0 is hydrogen or halo, in another embodiment hydrogen or fluoro; R12 and R14 are hydrogen; R3 is hydroxy; and R4 is heterocycloalkyl, alkyl, or heteroaryi, where the alkyl is optionally substituted with -NR8R8’ (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl.
[00214] In another embodiment of the Invention (BIO), the compound of Formula I is more specifically according to Formula I(i) or I(j):
10);
i(l); where X, R3, R2, R3, R4, R5, R6, R7, R10, Ri2 and Ri4 are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, R , R , R , and R are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; and R!0, R32, and Ri4 are independently hydrogen, halo, or alkyl. In another embodiment, X is fluoro or chloro and R is iodo or bromo; R is hydrogen or halo, in another embodiment hydrogen or fluoro; R32 and R34 are hydrogen; R3 is hydroxy; and R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl.
[00215] In another embodiment of the Invention (Bll), the compound of Formula I is more specifically according to Formula I(k) or I(m):
I(k);
I(m); where X, R1, R2, R3, R4, R5, R6, R7, Ri0, R32 and R14 are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, R3, R2, R5, and R6 are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; and R10, Ri2, and R14 are independently hydrogen, halo, or alkyl. In another embodiment, X is fluoro or chloro and R7 is iodo or bromo; Ri0 is hydrogen or halo, in another embodiment hydrogen or fluoro; R12 and R34 are hydrogen; R3 is hydroxy; and R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl.
[00216] In another embodiment of the Invention (B12), the compound of Formula I is more specifically according to Formula I(n) or I(o):
I(n),
I(o); where X, R1, R2, R3, R4, R5, R6, R7, R10, R12, R14, and R!9 are as defined in the
Summary of the Invention for a compound of Group B.
[00217] In another embodiment of embodiment B12, the compound of Formula I is more specifically according to Formula I(n) or I(o) where R' is halo or alkyl; and all other groups are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, R7 is iodo or bromo. |00218] In another embodiment of embodiment B12, the compound of Formula I is more specifically according to Formula I(n) or I(o) where X is halo, haioalkyl, or haloalkoxy; and all other groups are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, X is halo. In another embodiment X is fluoro or chloro.
[00219] In another embodiment of embodiment B12, the compound of Formula I is more specifically according to Formula I(n) or I(o) where R3 is halo, nitro, -NR8R8’, -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(0)OR8, -C(0)NR8R8', -NR8C(0)0R8', -NR8C(0)NR8 R8”> -NR8C(0)0R8’, -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), -CH2NR25C(~NH)(NR25aR25b), -CH2NR2sC(=NH)(N(R25a)(N02), -CH2NR25C(=NH)(N(R25a)(CN), -CH2NR25C(-NB)(R25), -CH2NR2sC(NR25aR25b)=CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroaryl alkyl, -OR8, -NRSR8’, -NR8S(0)2R9, -CN, ~S(0)mR9, -C(0)R8, ~C(0)0R8, -C(0)NR8R8’,-NR8C(0)NR8'R8”!-NR8C(0)0R8’ and -NR8C(0)R8 ; and R4 is as defined in the Summary of the Invention; or R3 and R4 together with the carbon to which they are attached form C(O) or C(=NOH); and unless otherwise indicated, R8 and R8 are as defined in the Summary of the Invention; and all other groups are as defined in the Summary of the Invention for a compound of Group B, [00220} In another embodiment of embodiment B12, the compound of Formula I is more specifically according to Formula I(n) or I(o) where R19 is alkyl; R1, R2, R5, and R6 are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; and Ri0, R12, and R14 are independently hydrogen or halo. In another embodiment, R19 is methyl; X is fluoro or chloro and R7 is iodo or bromo; Ri0 is hydrogen or fluoro; R12 and R14 are hydrogen; and RJ is hydroxy. In another embodiment, R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R8’ (where R8 is hydrogen or alkyl and R8’ is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pyrrolidinyl, 1 (R, 5)-amino-ethyl, 1 (R)-amino-eihyl, l(S)-amino-ethyl, 1 (R, S)-(methylamino)-ethyl, 1 (R)-(methylamino)-ethyl, 1 (S)-(methylamino)-ethyl, l(i?,5)-(dimethylamino)-ethyl, 1 (R)-(dimethylamino)-ethyl, 1 (S)-(dimethylamino)-ethyl, 1 (R, S)-(3,4-cis-dihvdroxy-cyclopentylamino)-ethyl, 1 (R)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, or l(S)-(3,4-cis-dihydroxy-cyclopentyIamino)-ethyl. {00221] In another embodiment of the Invention (B13), the compound of Formula I is more specifically according to Formula I(p):
I(P) where X, Rf, R2, R3, R4, Rs, R6, R7, R10, R12, and R59 are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, R5, R‘, Rs, and R6 are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; and Ri0 and R12 are independently hydrogen, halo, or alkyl. In another embodiment, X is fluoro or chloro; R7 is iodo or bromo; R10 is hydrogen or halo, in another embodiment hydrogen or fluoro; R12 is hydrogen; R19 is hydrogen or alkyl, in another embodiment hydrogen or methyl; R3 is hydroxy. In another embodiment, R4 is heterocyeloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NRSR8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pyrrolidinyl, l(f?,S)-amino-ethyl, l(R)-amino-ethyl, I (5)-amino-ethyl, 1 (R, 5)-(methylamino)-ethyi, 1 (R)-(methylamino)-ethyl, 1 (S)-(methylamino)-ethyl, 1 (R,S)-(dimethylamino)-ethyl, 1 (R)~(dimethylamino)-ethyl, 1 (S)-(dimethylamino)-ethyl, 1 (R, 5)-(3,4-cis-di hydroxy -cyclopentylamino)-ethvl, 1 (R)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, or 1 (5)-(3,4-cis~dihydroxy-cyclopentylamino)-ethyl.
[00222] In another embodiment of the Invention (B14), the compound of Formula I is more specifically according to Formula I(q):
I(q> where X, R5, R2, R:\ R4, R5, R6, R7, R10, Ri2 R14, and R16 are as defined in the Summary of the Invention for a compound of Group R.
[00223] In another embodiment of embodiment B14, the compound of Formula I is more specifically according to Formula I(q) where R3 is halo, nitro, ~NR8R8’, -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(Q)OR8’, -NRsC(G)NR8’R8” -NR8C(0)0R8’, -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), -CH2NR25C(=NH)(NR25aR25b), -CH2NR25C(=NH)(N(R25a)(NG2), -CH2NR25C(~NH)(N(R25a)(CN), -CH2NR2SC(=NH)(R25), -CH2NR25C(NR25aR2:,b)=:CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloaikyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloaikyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloaikyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NR8R8’, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, -C(0)NR8R8',-NR8C(0)NR8’R8',,-NR8C(0)0R8’ and -NR8C(0)R8 ; and R4 is as defined in the Summary of the Invention; or R3 and R4 together with the carbon to which they are attached form C(O) or C(~NOH); and all other groups are as defined in the Summary of the invention for a compound of Group B.
[00224] In another embodiment of embodiment B14,the compound of Formula I is more specifically according to Formula I(q) where R*, R2, R5, and R6 are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; and R10, Ri2, R14, and R16 are independently hydrogen or halo. In another embodiment, R10 is halo and R12, R14, and R16 are hydrogen. In another embodiment, X is fluoro or chloro; R7 is iodo or bromo; R!0 is chloro; and R3 is hydroxy. In another embodiment, R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R8’ (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pyrrolidinyl, benzimidazolyl, 1 (/?,5)-amino-ethyl, l(i?)-amino-ethyl, 1 (^-amino-ethyl, 1 (R,5)~(methylamino)~ethyl, l(i?)~(methylamino)-ethyl, 1 (S)-(methylamino)-ethyl, 1 (R, 5)-(3,4-cis-dihydroxy-cy clopentylamino)rethyl, 1 (R)-( 3,4-cis-dihydroxy-cyclopentylamino)-ethyl, or 1 (5)-(3,4-cis~dihydroxy-cyclopentylamino)-ethyl.
[00225] In another embodiment of the Invention (B15), the compound of Formula I is more specifically according to Formula I(r):
I(r) where X, Rf, R2, R3, R4, R5, R6, R7, Ri0, Ri2 and R14 are as defined in the Summary of the Invention for a compound of Group B. In another embodiment, R1, R2, R5, and R6 are hydrogen; X and R7 are halo; R3 and R'1 are as defined in the Summary of the Invention for Group B; R50 and Ri2 are independently hydrogen, halo, or alkyl; and R14 is hydrogen, halo, alkyl, or amino. In another embodiment, X is fluoro or chloro; R7 is iodo or bromo; R!0 is hydrogen or halo, in another embodiment hydrogen or fluoro; R12 is hydrogen; R14 is hydrogen, alkyl, or amino, in another embodiment hydrogen, methyl, or amino; R3 is hydroxy. In another embodiment, R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R* (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pyrrolidinyl, 1 (R,5)-amino-ethy 1, 1 (/?)-amino-ethyl, 1 (5)-amino-ethyi, 1 (J?,5)-(methylamino)-ethyl, 1 (R)-(methyiamino)-ethyl, 1 (iS)-(methylamino)-ethyl, 1 (R, 5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, 1 (R)-(3,4-cis-dihydroxy-eyclopentylammo)~ethyl, or 1 (^-(3,4-cis-dihydroxy-cyclopentyiamino)-ethyl.
[00226] In another embodiment of the Invention (B16), the compound of Formula I is more specifically according to Formula I(s):
I(s) where X, R1, R2, R3, R4, R5, R6, R7, R10, R12 and R14 are as defined in the Summary of < λ i £ the Invention for a compound of Group B. In another embodiment, R‘, R^, R , and R are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; and Ri0 and R!2 are independently hydrogen, halo, or alkyl; and R14 is hydrogen, halo, alkyl, or amino. In another embodiment, X is fiuoro or chloro and R7 is iodo or bromo; Ri0 is hydrogen or halo, in another embodiment hydrogen or fiuoro; Ru is hydrogen; Ri4 is hydrogen, methyl, or amino; R3 is hydroxy; and R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8RS (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl.
[00227] In another embodiment of the Invention (BIB), the compound of Formula I is more specifically according to Formula I(u), I(v), I(w), or I(x):
where X, R1, R2, R3, R4, R5, R6, R7, Ri0, R12 and R14 are as defined in the Summary of the Invention for a compound of Group B.
[00228] In another embodiment of embodiment B18, the compound of Formula I is more specifically according to Formula I(u), I(v), I(w), or I(x) where R3 is halo, nitro, ~NR8Rr, -OR8, -NHS(0)2R8, -CN, -S(0)mR8, ~S(0)2NR8R8’, -C(0)R8, -C(0)QR8, -C(0)NR8R8’, -NRsC(0)0R8’, -NR8C(0)NR8R8” -NR8C(0)OR8’, -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), -CH2NR2SC(===NH)(NR25aR25b), -CH2NR25C(-NH)(N(R2Sa)(N02)5 -CH2NR25C(=NH)(N(R25a)(CN), -CH2NR2SC(=NH)(R25), -CH2NR25C(NR25aR25b)-CH(N02), alkyl, alkenyl, alkynyl, cycloaikyl, heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyi, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted aryialkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NR8Rr, -NR8S(Q)2R9, -CN, -S(0)mR9, -C(G)R8, -C(0)0R8, -C(0)NRsRr -NRsC(0)NR8’R8”, -NR8C(0)0R8’ and -NR8C(0)R8'; and R4 is as defined in the Summary of the Invention for a compound of Group B; or R3 and R4 together with the carbon to which they are attached form C(Q) or C(-NOH); and all other groups are as defined in the Summary of the Invention for a compound of Group B.
[00229] In another embodiment of embodiment BIB, the compound of Formula I is more specifically according to Formula I(t), I(u), I(v), or I(w) where R3 and R4 are independently halo, nitre, -NR8R8’, -OR8, -NHS(G)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)0R8’, -NR8C(Q)NR8’R8”, -NR8C(0)0R8’, -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), -CH2NR25C("NH)(NR25aR25b), -CH2NR25C(=NH)(N(R25a)(N02), -CH2NR25C(=NH)(N(R25aXCN), -CH2NR25C(-NH)(R25), -CH2NR25C(NR25aR25b)=CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloaikyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroaryl alkyl, -OR8, -NR8R8, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, ~C(0)NRsR8'i-NR8C(0)NR8’R8”,-NRSC(0)0R8' and -NRsC(0)Rs; or R3 and R4 together with the carbon to which they are attached form C(O) or C^NOH); and all other groups are as defined in the Summary of the Invention for a compound of Group B.
[00230] In another embodiment of embodiment B18, the compound of Formula I is more specifically according to Formula I(u), I(v), I(w), or I(x) where R4 is heterocycloalkyl, heteroaryl (optionally substituted with alkyl), or alkyl where the alkyl is optionally substituted with -NRSR8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl). In another embodiment, R4 is piperidinyl, pyrrolidinyl, 1 (i?,S)-amino-propyl, i(i?)-amino-propyl. 1 (S)-amino-propyl, 1 (R, S)~(methy lamino)-propy i, 1 (R)-(methy lami no)-propy 1, 1 (S)-(methylamino)-propyl, \(R,S)-(3,4-cis-dihydroxy-cyclopentylamino)-propyl, 1 (Λ)-(3,4-c/s-dihydroxy-cyclopentylamino)-propyi, or 1 (5)-(3,4-cis-dihydroxy-cyclopentylamino)-propyl.
[00231] In another embodiment of embodiment B18, the compound of Formula I is more specifically according to Formula I(u), I(v), I(w), or I(x) where R!, R2, Rs, and R6 are hydrogen; X and R7 are halo; R3 and R4 are as defined in the Summary of the Invention for Group B; and Ri0, R12, and R14 are independently hydrogen, halo, or alkyl. In another embodiment, X is fluoro or chloro; R? is iodo or bromo; Rt0 is hydrogen or halo, in another embodiment hydrogen or fluoro; R12 and R14 are hydrogen; and R3 is hydroxy. In another embodiment R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8RS (where R8 is ot hydrogen or alkyl and R is hydrogen, alkyl, or cycloalkyS where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl.
[00232] In another embodiment of the Invention (B19), the compound of Formula I is more specifically according to Formula I(cc)
I(ec) where X, R1, R2, R3, R4, R5, R6, and R7 are as defined in the Summary' of the Invention for a compound of Group B. In another embodiment, R!, R2, R5, and R6 are hydrogen; and X and R7 are halo. In another embodiment, X is fluoro or chloro; and R3 is hydrogen or hydroxy; R7 is iodo or bromo. In another embodiment, R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NRSR8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently-selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pyrrolidinyl, benzimidazolyl, N-methyi-benzimidazolyl, methylaminomethyl, l(i?,5)-amino-ethyl, 1 (R)-amino-ethyl, 1 (5)-amino-ethyl, 1 (R, 5)-(methylamino)-ethyl, 1 (R)-(methylamino)-ethyl, 1 (S)-(methy lamino)-ethyl, l(R,5)-(dimethylamino)-ethyl, 1 (i?)-(dimethylamino)-ethyl, 1 (5)-(dimeihylamino)-ethyl, 1 (R5)-amino-propyl, l(i?)-amino-propyl, l(5)-amino-propyl, 1 (R,5)-(methylamino)-propyl, 1 (i?)-(methylamino)-propyl, 1 (S)-(methylamino)-propyl, 1 (R,5)-(dimethylamino)-propyl, 1 (R)-(dimethylamino)-propyl, 1 (5)-(dimethylamino)-propyl, 1 (R, 5)-(3,4-cis-dihydroxy-cvclopentylamino)-ethyl, 1 (J?)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, or 1 (5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl.
[00233] In an embodiment (B19a) of embodiment B19, the compound of Formula I is that where R4 is heterocycloalkyl or alkyl where the alkyl is optionally substituted with -NR8Rs (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl). In another embodiment, R4 is piperidinyl, pyrrolidinyl, methylaminomethyl, l(R,5)-amino~ethyl, l(i?)-amino-ethyL l(S)~amino-ethyl, 1 (R, 5)-(methylamino)-ethyl, 1 (i?)-(methylamino)-ethyl, 1 (5)-(methylamino)-ethyl, 1 (R5)-(dimethylamino)~ethyl, 1 (R)-(dimethylamino)-ethyl, 1 (5)-(dimethylamino)-ethyl, 1 (R5)-amino-propyl, 1 (R)-amino-propyl, l(5)-amino-propyl, l(i?,5)-(methylamino)-propyl, 1 (i?)~(methylamino)~propylJ 1 (5)-(methylamino)-propyl, l(i?,5)-(dimethylamino)-propyl, 1 (R)-(dimethylamino)-propyl, l(5)-(dimethylamino)-propyl, 1(R,5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, 1 (R)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, or 1(5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl.
[00234] In another embodiment of the Invention (B20), the compound of Formula I is more specifically according to Formula I(dd)
I(dd) where X, R1, R2, R3, R4, R5, R6, and R7 are as defined in the Summary of tlie Invention for a compound of Group B. In another embodiment, Rs, R2, R5, and R6 are hydrogen; and X and R7 are halo. In another embodiment, X is fluoro or chloro; and R3 is hydrogen or hydroxy; R7 is iodo or bromo. In another embodiment, R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pyrrolidinyl, benzimidazolyl, N-methyl-benzimidazolyl, methylaminomethyl, 1 (R, 5')-amino-ethyl, 1 (R)-amino-ethyl, 1 (5)-amino-ethyl, 1 (R, 5)-(methylamino)-ethyl, 1 (R)-(methylamino)-ethyl, 1 (S)-(methylamino)-ethy 1,1 {R, 5)-(dimethy lamino)-ethy 1, 1 (J?)-(dimethylammo)-ethyl, l(5)-(dimethylamino)-ethyl, 1 (i?,S)-amino~propyl, 1 (i?)-amino-propyl, l(5)-amino-propvl, 1 (R,5)-(methylamino)-propyl, 1 (jR)-(methylamino)-propyl, 1 (5)-(methylamino)-propy 1, l(JR,^-(dimethylamino)-propyi} 1 (/?)-(dimethylamino)-propyl, 1 (5)-(dimethyIamino)-propyl, 1 (i?,S)-(3.4-cis-dihydroxy-cyc!opentylamino)-ethyl, l(/?)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, or 1 (5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl.
[00235] In an embodiment (B20a) of embodiment B20, the compound of Formula I is that where R4 is heterocycloalkyl or alkyl where the alkyl is optionally substituted with -NR8R8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl). In another embodiment, R4 is piperidinyl, pyrrolidinyl, methylaminomethyl, 1 (R, 5)-amino-ethyl, 1 (i?)-amino-ethyl, 1 (S)-amino-ethyl, 1 (i?,5)-{methylamino)-ethyl, 1 (/?)-(methylamino)-ethyl, 1 (5)-(methylamino)-ethyl, 1 (R, 5)-(dimethylamino)-ethyl, 1 (i?)-(dimethylamino)-ethyl, 1 (5)-(dimethylamino)-ethyl, 1 (R,5)-amino-propyl, l(i?)-arnino-propyl, 1 (S)-amino-propyl, 1 (j?,5)-(methylamino)-propyl, 1 (i?)-(methylamino)-propyl, 1 (5)-(methylamino)-propyl, 1 (i?,5)-(dimethylamino)-propyI, 1 (/?)-(dimethyIamino)~ propyl, 1 (5)-(dimethylamino)-propyl, 1 (R, 5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, 1 (R)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, or 1 (5)-(3,4-ci s-dihydroxy-cyclopentylamino)-ethyl.
[00236] In one embodiment of the Invention (Cl), the compound of Formula I is selected from Group C where all groups are as defined in the Summary of the Invention.
[00237] In another embodiment of the invention (C2), the compound of Formula I is that where X and R' are halo; and all other groups are as defined for a compound selected from Group C.
[00238] In another embodiment of the invention (C3), the compound of Formula I is selected from Group C where R3 is halo, nitro, -NR8R8 , -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8’, -C(0)R8, -C(0)OR8, -C(0)NR8R8’, -NR8C(0)0R8’, -NR8C(0)NR8R8”, -NR8C(0)0R8', -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), -CH2NR25C(=NH)(NR25aR25b), -CH2NR2SC(=NH)(N(R2S8)(N02)5 -CH2NR25C(=NH)(N(R25a)(CN), -CH2NR25C(=NH)(R25), -CH2NR25C(NR25aR25b)=CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryi, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NRSR8, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, -C(0)NR8R8>NR8C(0)NR8’R8”, -NR8C(0)0R8' and -NR8C(0)R8; and R4 is as defined in the Summary of the Invention; or R3 and R4 together with the carbon to which they are attached form C(O) or C(-NOH); and all other groups are as defined in the Summary of the Invention for a compound of Group C. In another embodiment, R1, R2, R5 and R6 are hydrogen; and X and R7 are halo,
[00239] In another embodiment of the invention (C4), the compound of Formula I is selected from Group C where R3 and R4 are independently halo, nitro, -NR8R8, -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8R8', -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)0R8’, -NR8C(0)NR8’Rs"; -NR8C(0)0R8’, ~NR8C(0)R8’, -CH2N(R25)(NR25aR2Sb), -CH2NR25C(=NH)(NR25aR25b), -CH2NR25C(=NHXN(R25a)(N02), -CH2NR25C(==NH)(N(R25a)(CN), -CH2NR2SC(=NH)(R25), -CH2NR25C(NR2SaR25b)-CHCN02), alkyl, alkenyl, alkynyl, cycloalkyl. heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NR8R8’, -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)0R8, -C(Q)NRsRr -NR8C(0)NR8’R8”, -NR8C(0)0R8’ and -NR8C(0)R8’; or R3 and R4 together with the carbon to which they are attached form C(O) or C(=NOH); and all other groups are as defined in the Summary of the invention for a compound of Group C. In another embodiment, R1, R2, R5 and R6 are hydrogen; and X and R7 are halo.
[00240] In another embodiment of the invention (C5), the compound of Formula I is that where A is
and X, R1, R2, R3, R4, R5, R6, R7, Ri0, and Ri0a are as defined in the Summary of the invention for a compound of Group C. In another embodiment, R ?Λ R5, and R6 are hydrogen; X and R7 are halo; R10 is hydrogen or halo; and RIOa is alkyl. In another embodiment, X is fluoro or chloro; R3 is hydroxy; R7 is iodo or bromo; Ri0 is hydrogen or fluoro; and Ri0a is methyl. In another embodiment, R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR*R8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pyrrolidinyl, benzimidazoiyl, N-methyl-benzimidazolyl, methylaminomethyl, l(i?,5)-amino-ethyl, 1 (R)-amino-ethyl, l(S)-amino-ethyl, 1 (R5)-(methylamino)-ethyl, 1 (R)-(methylamino)-ethyl, 1 (S)-(methylamino)-ethyl, 1 (R,S)-(dimethylamino)-ethyl, 1 (/?)-(dimethylamino)-ethyl, 1 (£)-(dimethylamino)-ethyl, l(R,S)-amino-propyl, l(R)~amino-propyl, l(S)-amino-propyl, 1 (i?,5)-(methy!amino)-propyl, 1 (i?)-(methyl amino) -propyl, 1 (S)-(methylamino)-propyl, 1 (i?, 5)-(dimethylamino)-propy 1, 1 (R)-(dimethylamino)-propyl, 1 (S)-(dimethylamino)-propyl, 1 (R, 5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, l(R)'(3,4-cis-dihydroxy-cyclopentylamino)-ethyi, or 1 (S)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl.
[00241] In another embodiment of the invention (C6), the compound of Formula I is that where A is
and X, R‘, R2, R3, R4, R5, R6, R7, R50, and Ri0a are as defined in the Summary of the invention for a compound of Group C. In another embodiment, R1, R2, R5, and R6 are hydrogen; X and R7 are halo; R!0 is hydrogen or halo; and Ri0a is alkyl. In another embodiment, X is fluoro or chloro; R3 is hydroxy; R7 is iodo or bromo; R10 is hydrogen or fluoro; and RIOa is methyl. In another embodiment, R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalky! is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pyrrolidinyl, benzimidazolyl, N-methylbenzimidazolyl, 1 (R,5)-amino-ethy 1, l(/?)-amino-ethyl, I(5)-amino-ethyl, l(i?,S)-amino-propyl, l(i?)-amino-propyl, l(5)-amino-propyl, 1 (/?,5)-(methylamino)-propyl, l(/?)-(methylamino)-propyl, 1 (5)-(methylamino)-propyl, 1 (R, 5)-(3,4-cis-dihydroxy-cyclopentylamino)-propyl, l(R)-(3,4-cis-dihydroxy-cyclopentylamino)" propyl, 1 (5)-( 3,4-cis-dihydroxy-cycl opentylamino)-propyl, 1 (K, S)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, 1 (R)~(3 34-cis-dihydroxy-cyclopentylamino)-ethyl, or 1(5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl.
[00242] In another embodiment of the Invention (C7), the compound of Formula I is more specifically of Formula l(y) or I(z):
i(y);
I(z) where R1, R2, R5, and R6 are hydrogen; X and R7 are halo; R3, R4, Ri0, R10a, and Y1 are as defined in the Summary of the Invention for a compound of Group C. In another embodiment, X is fluoro or chloro; R7 is iodo or bromo; Ri0 is hydrogen, halo, or alkyl, in another embodiment hydrogen or halo; and Ri0a is alkyl, in another embodiment methyl. In another embodiment R10 is hydrogen or fluoro; R3 is hydroxy; and R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl.
[00243] In one embodiment of the Invention (D), the compound of Formula I is selected from Group D where ail groups are as defined in the Summary of the Invention.
[00244] In another embodiment of the invention (Dl), the compound of Formula I is that where X and R7 are halo; and all other groups are as defined for a compound selected from Group D.
[00245] In another embodiment of the invention (D2), the compound of Formula I is selected from Group D where R3 is halo, nitro, -NR8R8, -OR8, -NHS(0)2R8, -CN, -SfO^R8, -S(0)2NR8R8’, -C(0)R8, -C(Q)QR8, -C(0)NR8R8’, -NR8C(0)0R8’, -NR8C(0)NRs'R8" -NR8C(0)QR8’, -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), -CH2NR25C(=NH)(NR25aR25b)s -CH2NR25C(=NH)(N(R25a)(N02), -CH2NR25C(::=NH)(N(R25a)(CN)5 -CH2NR25C(=NH)(R25), . -CH2NR2:>C(NR25aR";5b)=CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyl are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloaikyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyl, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NR8R8 , -NR8S(0)2R9, -CN, -S(0)mR9, -C(0)R8, -C(0)OR8, -C(0)NR8R8, -NR8C(Q)NR8’R8” -NRsC(0)0R8 and -NRsC(0)R8; and R4 is as defined in the Summary of the Invention; or R3 and R4 together with the carbon to which they are attached form C(O) or C(=NOH); and all other groups are as defined in the Summary of the Invention for a compound of Group C. In another embodiment, R1, R2, R5 and R6 are hydrogen; and X and R7 are halo.
[00246] In another embodiment of the invention (D3), the compound of Formula I is selected from Group D where R and R are independently halo, nitro, -NR R , -OR8, -NHS(0)2R8, -CN, -S(0)mR8, -S(0)2NR8Rr, -C(0)R8, -C(0)OR8, -C(0)NRsR8’, -NRsC(0)OR8 , -NR8C(0)NR8’r8”> -NR8C(0)0R8’, -NR8C(0)R8’, -CH2N(R25)(NR25aR25b), -CH2NR25C(=NH)(NR25aR25b), -CH2NR25C(~NH)(N(R25a)(N02)s -CH2NR25C(=NH)(N(R25a)(CN), -CH2NR25C(=NH)(R25), -CH2NR25C(NR25aR25b)=CH(N02), alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, or heterocycloalkyl; where the alkyl, alkenyl, alkynyl, cycloalkyl, heteroaryl, and heterocycloalkyi are independently optionally substituted with one, two, three, four, five, six or seven groups independently selected from halo, alkyl, haloalkyl, nitro, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted aryl, optionally substituted arylalkyi, optionally substituted heteroaryl, optionally substituted heteroarylalkyl, -OR8, -NR8R8, -NR8S(0)2R9, -CN, -S(0)roR9, -C(0)R8, -C(0)0R8, -C(0)NR8R8’, -NR8C(0)NR8 R8” -NR8C(0)0R8’ and -NR8C(0)R8’; or R3 and R4 together with the carbon to which they are attached form C(O) or Ci^NOH); and all other groups are as defined in the Summary of the Invention for a compound of Group C. In another embodiment, R1, R2, R5 and R6 are hydrogen; and X and R7 are halo, [00247] In another embodiment of the invention (D4), the compound of Formula I is that where A is
I(aa) or
I(bfo) where R40 is hydrogen or methyl (in another embodiment, R40 is hydrogen) and all other groups are as defined in the Summary of the Invention. In another embodiment, R1, Rz, R3, and R6 are hydrogen; X and R7 are halo; and R40 is hydrogen or methyl. In another embodiment, X is fluoro or chloro; and R3 is hydrogen or hydroxy: R7 is iodo or bromo. In another embodiment, R4 is heterocycloalkyl, alkyl, or heteroaryl, where the alkyl is optionally substituted with -NR8R8 (where R8 is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyi is optionally substituted with one or two groups independently selected from hydroxy and alkyl) and the heteroaryl is optionally substituted with alkyl. In another embodiment, R4 is piperidinyl, pyrrolidinyi, benzimidazolyl, //-methyl-benzimidazolyl, methylaminomethyl, 1 (R5)~amino-ethyl, 1 (R)-amino-ethyl, l(S)-amino-ethyl, 1 (R,5)-(methylamino)-ethyl, 1 (R)~(methylamino)-ethyl, 1 (S)-(methylamino)-ethyl, 1 (f?,S)-(dimethylamino)-ethyl, 1 (R)-(dimethylamino)-ethyl, l(S)-(dimethylamino)-ethyl, 1 (/?,S)-amino-propyl, 1 (R)-amino-propyl, 1 (5)~amino-propyl, 1 (R, 5)-(methy lamino)-propy 1, 1 (i?)-(methylamino)-propyl, 1 (5)-(methylamino)-propyl, l(R,S)-(dimethylamino)-propyl, 1 (i?)-(dimethylamino)-propyl, 1 (5)-(dimethylamino)-propyl, !(/?,5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, 1 (R)~( 3,4-cis-dihydroxy-cyclopentylamino)-ethyl, or 1 (5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl.
[00248} In an embodiment (D4a) of the invention of D4, the compound of Formula I is that where R4 is heterocycloalkyl or alkyl where the alkyl is optionally substituted with -NR8R8 (where Rs is hydrogen or alkyl and R8 is hydrogen, alkyl, or cycloalkyl where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl). In another embodiment, R4 is piperidinyl, pyrrolidinyl, methylaminomethyl, 1 (J?,S)-mmno-ethyl, l(J?)-amino-ethyl, l(5)-amino-ethyl, 1 (/?,S)-(methylamino)-ethy 1, 1 (i?)-(methylamino)-ethyl, 1 (S)-(methylarnino)-ethyh 1 (R, 5)-(dimethylamino)-ethyl, 1 (R)-(dimethylamino)-ethyl, 1 (S)-(dimethylamino)-ethyl, 1 (R,S)-amino-propyi, l(j?)-amino-propyl, 1 (S)-amino-propyl, 1 (/?,S)-(methylamino)-propyl, 1 (/?)-(methylamino)-propyl, 1 (5)-(methyIamino)-propyl, 1 (R5>(dimethylamino)~propyl, l(R)-(dimethylamino)-propyl, l(5)-(dimethyfamino)-propyl, 1(R,5)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, 1 (R)-(3,4-cis-dihydroxy-cyclopentylamino)-ethyl, or 1 (5)-(3,4-cis-dihydroxy-cyclopentylamino) -ethyl.
[00249} Another embodiment of the Invention (E) is directed to a Compound ot Formula 1 selected from Group A, Group B, and Group C where Group A A is phenylene optionally substituted with one or two groups selected from Ri0, R!", R14, and R16 where Ri0, Rn, Ri4 and R16 are independently hydrogen or halo; X is halo; R1, R2, R5 and R6 are hydrogen; R3 is hydrogen, halo, hydroxy, alkoxy, or amino; R4 is hydrogen, ~NR8R8’, -C(0)NRsR8’, -NR8C(0)0R8’, -NR8C(0)R8, -CH2N(R25)(NR25aR25b), -CH2NR25C(=NH)(NR25aR2Sb), -CH2NR2SC(=NII)(N(R25a)(N02), -CH2NR25C(-NH)(N(R25a)(CN), -CH2NR25C(=NH)(R2S), -CH2NR25C(NR25aR25b)-CH(N02), alkyl, alkenyl, cycloalkyl, heterocycloalkyl, or heteroaryl; where the R4 alkyl is optionally substituted with one, two, or three groups independently selected from -OR , halo, nitro, -S(0)mR9, optionally substituted heterocycloalkyl, -NR8R8, -NR8C(0)R8, -NR8S(0)2R9, ~NR8C(0)0R8, and aryl; where the R4 cycloalkyl is optionally substituted with one or two groups selected from -OR8 and -NRSRS; where the R4 heterocycloalkyl is optionally substituted with one or two groups independently selected from alkyl and ~C(0)GR8; and where the R4 heteroaryl is optionally substituted with -NR8RS; or R3 and R4 together with the carbon to which they are attached form C(O) or C(=NOH); m is 0; R7 is halo; R8 and R8 are independently selected from hydrogen, hydroxy, alkyl, alkenyl, alkynyl, aryl, heterocycloalkyl, heteroaryl, and cycloalkyl; where the R8 and R8 alkyl are independently optionally substituted with one, two, or three groups indendently selected from hydroxy, -NR30R30 (where R30 and R30 are independently hydrogen, alkyl, or hydroxyalkyl), optionally substituted heteroaryl, optionally substituted cycloalkyl), optionally substituted alkoxy, optionally substituted cycloalkyl, optionally substituted aryl, optionally substituted heterocycloalkyl, optionally substituted heteroaryl, -C(0)NR33R33a (where R33 is hydrogen or alkyl and R33a is alkyl, alkenyl, alkynyl, or cycloalkyl), optionally substituted aryloxy, -S(0)„R31 (where n is 0 and R3' is alkyl), carboxy, alkoxycarbonyl, and -NR3,‘C(0)R32e (where R32 is hydrogen or alkyl and R32a is alkyl, alkenyl, alkoxy, or cycloalkyl); or where the alkyl is optionally substituted with one, two, three, four, or five halo; where the R8 and R8 heteroaryl are independently optionally substituted with one or two groups indendently selected from amino and alkyl; where the R8 and R8 heterocycloalkyl are independently optionally substituted with one, two, or three groups indendently selected from alkyl, alkoxycarbonyl, optionally substituted arylalkyl, hydroxy, alkoxy, and hydroxyalkyl: where the R8 and R8 aryl are independently optionally substituted with one or two groups indendently selected from hydroxy, alkoxy, halo, -NR32C(0)R32a (where R32 is hydrogen or alkyl and Rj2a is alkyl, alkenyl, alkoxy, or cycloalkyl), and -NR34S02R34a (where R34 is hydrogen or alkyl and R34a is alkyl, alkenyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl); and where the R8 and R8 cycloalkyl are independently optionally substituted with one, two, or three groups indendently selected from hydroxy, hydroxyalkyl, alkoxy, carboxy, -C(0)NR33R33a (where R33 is hydrogen or alkyl and R33a is alkyl, alkenyl, alkynyl, or cycloalkyl), and optionally substituted cycloalkyl; and R9 is alkyl or aryl;
Group B A is thien-3,4-diyl, benzo[if]isoxazol-5.6-diyl, l/f-indazol-5,6-diyl (optionally substituted at the N1 position with R19 where R19 is alkyl or alkenyl), benzo[<floxazol-5,6-diyl, benzo[<flthiazol~5,6-diyl, l//-benzo[d]imidazol-5,6-diyl (optionally substituted at the N1 position with R19 where R19 is alkyl or alkenyl), li/-benzo[i/j[l,2,3]triazol-5,6-diyl (optionally substituted at the N1 position with R19 where R19 is alkyl or alkenyl), imidazo[l,2-a]pyridin-6,7-diyl, cinnolin-6,7“diyl5 quinolin-6,7-diyl, pyridin-3,4-diyl, or 1-oxido-pyridin- 3,4-diyl; where A is optionally substituted with one, two, or three groups independently selected from Ri0, R12, R14, Ri6 and R19 where R10, Riz, R14 and R16 are independently hydrogen, alkyl, halo, or amino; and Ri9 is hydrogen or alkyl; X is halo; R;, R2, R5 and R6 are hydrogen; R3 is hydrogen or hydroxy; R4 is -NR8R8, heterocycioalkyl, heteroaryl, or alkyl; where the alkyl is optionally substituted with -NR8R8 and where the heteroaryl is optionaly substituted with alkyl; R·' is halo; R8 is hydrogen or alkyl; and R* is hydrogen, alkyl, or cycloalkyl; where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl;
Group C A is
(a) where Ri0 is hydrogen or halo;
Ri0a is hydrogen or alkyl; Ύ1 is =CH- or =N-; X is halo; R!, R2, R5 and R6 are hydrogen; R3 is hydrogen or hydroxy; R4 is -NR8R8, heterocycloalkyl, heteroaryl, or alkyl; where the alkyl is optionally 8 8’ substituted with -NR R and where the heteroaryl is optionaly substituted with alkyl; R7 is halo; R8 is hydrogen or alkyl; and R8 is hydrogen, alkyl, or cycloalkyl; where the cycloalkyl is optionally substituted with one or two groups independently selected from hydroxy and alkyl.
Representative MEK Compounds [00250] Representative compounds of Formula I are depicted below. The examples are merely illustrative and do not limit the scope of the invention in any way. Compounds of the invention are named according to systematic application of the nomenclature rules agreed upon by the International Union of Pure and Applied Chemistry (IUPAC), International Union of Biochemistry and Molecular Biology (1UBMB), and the Chemical Abstracts Service (CAS). Names were generated using ACD/Labs naming software 8.00 release, product version 8.08.
Table I. Representative MEK Inhibitors
«_______i-----------------------------------
Other Representative Compounds
I
I
. j
Table 6. Representative Raf Inhibitors
Table 7. Representative EGFR and/or VEGFR Inhibitors
General Administration [00251] In one aspect, the invention provides pharmaceutical compositions comprising an inhibitor of MEK according to the invention and a pharmaceutically acceptable carrier, excipient, or diluent. In certain other embodiments, administration may preferably be by the oral route. Administration of the compounds of the invention, or their pharmaceutically acceptable salts, in pure form or in an appropriate pharmaceutical composition, can be carried out via any of the accepted modes of administration or agents for serving similar utilities. Thus, administration can be, for example, orally, nasally, parenterally (intravenous, intramuscular, or subcutaneous), topically, transdermally, intravaginally, intravesically, intraeistemally, or rectally, in the form of solid, semi-solid, lyophilized powder, or liquid dosage forms, such as for example, tablets, suppositories, pills, soft elastic and hard gelatin capsules, powders, solutions, suspensions, or aerosols, or the like, preferably in unit dosage forms suitable for simple administration of precise dosages.
[00252] The compositions can include a conventional pharmaceutical carrier, excipient, and/or adjuvants and a compound of Formula I, and, in addition, may include other medicinal agents and pharmaceutical agents that are generally administered to a patient being treated for cancer.
[00253] Adjuvants include preserving, wetting, suspending, sweetening, flavoring, perfuming, emulsifying, and dispensing agents. Prevention of the action of microorganisms can be ensured by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, and the like. It may also be desirable to include isotonic agents, for example sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form can be brought about by the use of agents delaying absorption, for example, aluminum monostearate and gelatin.
[00254] If desired, a pharmaceutical composition of the invention may also contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, antioxidants, and the like, such as, for example, citric acid, sorbitan monolaurate, triethanolamine oleate, butylalted hydroxytoluene, etc.
[00255] The choice of formulation depends on various factors such as the mode of drug administration (e.g., for oral administration, formulations in the form of tablets, pills or capsules are preferred) and the bioavailability of the drug substance. Recently, pharmaceutical formulations have been developed especially for drugs that show poor bioavailability based upon the principle that bioavailability can be increased by increasing the surface area i.e., decreasing particle size. For example, U.S. Pat. No. 4,107,288 describes a pharmaceutical formulation having particles in the size range from 10 to 1,000 run in which the active material is supported on a crosslinked matrix of macromolecules. U.S. Pat. No. 5,145,684 describes the production of a pharmaceutical formulation in which the drug substance is pulverized to nanoparticles (average particle size of 400 nm) in the presence of a surface modifier and then dispersed in a liquid medium to give a pharmaceutical formulation that exhibits remarkably high bioavailability.
[00256] Compositions suitable for parenteral injection may comprise physiologically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (propyleneglycol, polyethyleneglycol, glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
[00257] One specific route of administration is oral, using a convenient daily dosage regimen that can be adjusted according to the degree of severity of the disease- state to be treated, [00258] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is admixed with at least one inert customary excipient (or carrier) such as sodium citrate or dicalcium phosphate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol, and silicic acid, (b) binders, as for example, cellulose derivatives, starch, alignates, gelatin, polyvinylpyrrolidone, sucrose, and gum acacia, (c) humectants, as for example, glycerol, (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, croscarmellose sodium, complex silicates, and sodium carbonate, (e) solution retarders, as for example paraffin, (f) absorption accelerators, as for example, quaternary ammonium compounds, (g) wetting agents, as for example, cetyl alcohol, and glycerol monostearate, magnesium stearate and the like (h) adsorbents, as for example, kaolin and bentonite, and (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In the case of capsules, tablets, and pills, the dosage forms may also comprise buffering agents.
[00259] Solid dosage forms as described above can be prepared with coatings and shells, such as enteric coatings and others well known in the art. They may contain pacifying agents, and can also be of such composition that they release the active compound or compounds in a certain part of the intestinal tract in a delayed manner. Examples of embedded compositions that can be used are polymeric substances and waxes. The active compounds can also be in microencapsulated form, if appropriate, with one or more of the above-mentioned excipients.
[00260] Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and elixirs. Such dosage forms are prepared, for example, by dissolving, dispersing, etc., a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, and optional pharmaceutical adjuvants in a carrier, such as, for example, water, saline, aqueous dextrose, glycerol, ethanol and the like; solubilizing agents and emulsifiers, as for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propyleneglycol, 1,3-butyleneglycol. dimethylformamide; oils, in particular, cottonseed oil, groundnut oil, com germ oil, olive oil, castor oil and sesame oil, glycerol, tetrahydrofurfuryl alcohol, polyethyleneglycols and fatty acid esters of sorbitan; or mixtures of these substances, and the like, to thereby form a solution or suspension.
[00261] Suspensions, in addition to the active compounds, may contain suspending agents, as for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, or mixtures of these substances, and the like.
[00262] Compositions for rectal administrations are, for example, suppositories that can be prepared by mixing the compounds of the present invention with for example suitable non-irritating excipients or carriers such as cocoa butter, polyethyleneglyco 1 or a suppository wax, which are solid at ordinary temperatures but liquid at body temperature and therefore, melt while in a suitable body cavity and release the active component therein.
[00263] Dosage forms for topical administration of a compound of this invention include ointments, powders, sprays, and inhalants. The active component is admixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants as may be required. Ophthalmic formulations, eye ointments, powders, and solutions are also contemplated as being within the scope of this invention.
[00264] Compressed gases may be used to disperse a compound of this invention in aerosol form. Inert gases suitable for this purpose are nitrogen, carbon dioxide, etc.
[00265] Generally, depending on the intended mode of administration, the pharmaceutically acceptable compositions will contain about 1% to about 99% by weight of a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, and 99% to 1% by weight of a suitable pharmaceutical excipient. In one example, the composition will be between about 5% and about 75% by weight of a compound(s) of the invention, or a pharmaceutically acceptable salt thereof, with the rest being suitable pharmaceutical excipients.
[00266] Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, 18th Ed., (Mack Publishing Company, Easton, Pa., 1990). The composition to be administered will, in any event, contain a therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt thereof, for treatment of a disease-state in accordance with the teachings of this invention.
[00267] The compounds of the invention, or their pharmaceutically acceptable salts or hydrates, are administered in a therapeutically effective amount which will vary depending upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of the compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular disease-states, and the host undergoing therapy. The compounds of the present invention can be administered to a patient at dosage levels in the range of about 0.1 to about 1,000 mg per day. For a normal human adult having a body weight of about 70 kilograms, a dosage in the range of about 0.01 to about 100 mg per kilogram of body weight per day is an example. The specific dosage used, however, can vary. For example, the dosage can depend on a number of factors including the requirements of the patient, the severity of the condition being treated, and the pharmacological activity of the compound being used. The determination of optimum dosages for a particular patient is well known to one of ordinary skill in the art.
[002681 If formulated as a fixed dose, such combination products employ the compounds of this invention within the dosage range described above and the other pharmaceutically active agent(s) within its approved dosage range. Compounds of the instant invention may alternatively be used sequentially with known pharmaceutically acceptable agent(s) when a combination formulation is inappropriate. {00269] Representative pharmaceutical formulations containing a compound of Formula I are described below in the Pharmaceutical Composition Examples.
Utility [00270] Certain compounds of Formula I have been tested using the assay described in Biological Example 1 and have been determined to be MEK inhibitors. As such, compounds of Formula I are useful for treating diseases, particularly cancer in which MEK activity contributes to the pathology and/or symptomatology of the disease. For example, cancer in which MEK activity contributes to its pathology and/or symptomatology include malignant melanomas, colon cancer, rectal cancer, pancreatic cancer, Sung cancer, papillary and anaplastic thyroid cancer, and endometrial cancer, ovarian cancer, and the like.
[00271] Suitable in vitro assays for measuring MEK activity and the inhibition thereof by compounds are known in the art. For example, see WO 2006/061712 for measuring MEK1 and MEK2 in vitro. For further details of an in vitro assay for measuring MEK activity see Biological Examples, Example 1 infra. Following the examples disclosed herein, as well as those disclosed in the art, a person of ordinary skill in the art can determine the inhibitory activity of a compound of this invention.
[00272] Assays for measurement of in vitro efficacy in treatment of cancer are known in the art. For example, see WO 2006/061712, which is herein incorporated by reference, for cell-based assays for colon cancer. In addition, cell-based tumor models are described in Biological Examples, Example 2 and 3 infra.
[00273] Suitable in vivo models for cancer are known to those of ordinary skill in the art (including WO 2006/061712). For further details of in vivo models for colorectal cancer, melanoma, breast adenocarcinoma, and lung anaplastic carcinoma, see Biological Examples 4 and 5, infra. Biological Example 5 describes a particular combination of treatments. In conjunction with what is known in the art, one of ordinary skill in the art would know how to follow these examples to test other combinations of treatments.
General Synthesis {00274] Compounds of this invention can be made by the synthetic procedures described below. The starting materials and reagents used in preparing these compounds are either available from commercial suppliers such as Aldrich Chemical Co. (Milwaukee, Wis.), or Bachem (Torrance, Calif.), or are prepared by methods known to those skilled in the art following procedures set forth in references such as Fieser and Fieser’s Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd’s Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), March’s Advanced Organic Chemistry, (John Wiley and Sons, 4th Edition) and Larock’s Comprehensive Organic Transformations (VCH Publishers Inc., 1989). These schemes are merely illustrative of some methods by which the compounds of this invention can be synthesized, and various modifications to these schemes can be made and will be suggested to one skilled in the art having referred to this disclosure. The starting materials and the intermediates of the reaction may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallization, chromatography and the like. Such materials may be characterized using conventional means, including physical constants and spectral data.
[00275] Unless specified to the contrary, the reactions described herein take place at atmospheric pressure and over a temperature range from about -78 °C to about 150 °C, more preferably from about 0 °C to about 125 °C and most preferably at about room (or ambient) temperature, e.g., about 20 °C. Unless otherwise stated (as in the case of an hydrogenation), all reactions are performed under an atmosphere of nitrogen.
[00276] Prodrugs can be prepared by techniques known to one skilled in the art. These techniques generally modify appropriate functional groups in a given compound. These modified functional groups regenerate original functional groups by routine manipulation or in vivo. Amides and esters of the compounds of the present invention may be prepared according to conventional methods. A thorough discussion of prodrugs is provided in T. Higuchi and V. Stella, “Pro-drugs as Novel Delivery Systems,” Vol 14 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference for all purposes, [00277] The compounds of the invention, or their pharmaceutically acceptable salts, may have asymmetric carbon atoms or quatemized nitrogen atoms in their structure. Compounds of Formula I that may be prepared through the syntheses described herein may exist as single stereoisomers, racemates, and as mixtures of enantiomers and diastereomers. The compounds may also exist as geometric isomers. All such single stereoisomers, racemates and mixtures thereof, and geometric isomers are intended to be within the scope of this invention. Some of the compounds of the invention may exist as tautomers. For example, where a ketone or aldehyde is present, the molecule may exist in the enol form; where an amide is present, the molecule may exist as the imidic acid; and where an enamine is present, the molecule may exist as an inline. All such tautomers are within the scope of the invention.
[00278] The present invention also includes N-oxide derivatives and protected derivatives of compounds of Formula I. For example, when compounds of Formula I contain an oxidizabie nitrogen atom, the nitrogen atom can be converted to an N~ oxide by methods well known in the art. When compounds of Fonnula I contain groups such as hydroxy, carboxy, thiol or any group containing a nitrogen atom(s), these groups can be protected with a suitable “protecting group” or “protective group”. A comprehensive list of suitable protective groups can be found in T.W. Greene, Protective Groups in Organic Synthesis, John Wiley &amp; Sons, Inc. '1991, the disclosure of which is incorporated herein by reference in its entirety. The protected derivatives of compounds of Formula I can be prepared by methods well known in the art. ]00279] Methods for the preparation and/or separation and isolation of single stereoisomers from racemic mixtures or non-racemic mixtures of stereoisomers are well known in the art. For example, optically active (R)- and (S)~ isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. Enantiomers (R- and S-isomers) may be resolved by methods known to one of ordinary skill in the art, for example by: formation of diastereoisomeric salts or complexes which may be separated, for example, by crystallization; via formation of diastereoisomeric derivatives which may be separated, for example, by crystallization, selective reaction of one enantiomer with an enantiomer-specific reagent, for example enzymatic oxidation or reduction, followed by separation of the modified and unmodified enantiomers; or gas-liquid or liquid chromatography in a chiral environment, for example on a chiral support, such as silica with a bound chiral ligand or in the presence of a chiral solvent. It will be appreciated that where a desired enantiomer is converted into another chemical entity by one of the separation procedures described above, a further step may be required to liberate the desired enantiomeric form. Alternatively, specific enantiomer may be synthesized by asymmetric synthesis using optically active reagents, substrates, catalysts or solvents or by converting on enantiomer to the other by asymmetric transformation. For a mixture of enantiomers, enriched in a particular enantiomer, the major component enantiomer may be further enriched (with concomitant loss in yield) by recrystallization.
[00280] In addition, the compounds of the present invention can exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like. In general, the solvated forms are considered equivalent to the unsolvated forms for the purposes of the present invention.
[00281] The chemistry for the preparation of the compounds of this invention is known to those skilled in the art.
[00282] An intermediate of Formula II:
II where R7, X, R10, R12, R14, and R16 are as defined in the Summary of the Invention for Group A can be prepared using procedures known to one of ordinary skill in the art.
In particular, see (for example) US 7,019,033, WO 2002006213, WO 2003062191, WO 2003062189, WO 2002018319, WO2001005392, WO 2000064856, WO 2001005392, WO 9901421, WO 2004056789, Davis, E. M. et al. Org. Process Res. &amp; Dev. 2005, 9, 843-6, and Shapiro, N. et al. Synthetic Commim. 2005, 35, 2265-9 which are incorporated by reference herein. The following intermediates were prepared using similar procedures as described in the above references: 3,4-difluoro-2-[(2-fluoro-4-iodophenyS)amino]benzoic acid; 2-[(2-chloro-4-iodophenyl)amino]- 3,4-difIuorobenzoic acid; 4-fluoro-2~[(2-fluoro-4-iodophenyl)amino]benzoic acid; 4s5”difluoro-2-[(2-fluoro~4-iodophenyl)amino]benzoic acid; and 2-[(4-bromo-2-fluoropheny l)amino] -3,4-difluorobenzoic acid.
[00283] An intermediate of Formula 111(a) or 111(b):
111(a);
111(b) where R7, X, R30, R32, and Ri4 are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art.
In particular for formula 111(a), where R14 is amino or alkyl (particularly methyl); R10 is halo (particularly fluoro); R7 is hydrogen or halo (particularly bromo or chloro); X is halo (particularly chloro); and R32 is hydrogen see for example W02006030610, US2005049419, and US2005/0054701 which are incorporated by reference herein. 6-[(4"bromo-2-chlorophenyl)amino]-7-fluoro~3-methyl-l,2-benzisoxazole-5-carboxylic acid was prepared using methods similar to those disclosed in W02006030610, US2005049419, and US2005/0054701.
[00284] An intermediate of Formula IV(a) or IV{b):
IV(a);
IV(b) where R , X, R , R ", and R are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art.
[00285] An intermediate of Formula V(a) or V(b):
where R7, X, R10, R12, R14, and R19 are as defined in the Summary of the Invention for Group B can be prepared using procedures kno wn to one of ordinary skill in the art.
In particular the halo precursor of V(a) can be prepared using, for example. W02003101968 and W02002083648 which are incorporated by reference herein. In particular the halo precursor of V(b) can be prepared using, for example, US2004192653, US2004180896, US2004I76325 which are incorporated by reference herein. The halo precursors are then reacted with an appropriate aniline to yield the intermediates of Formula V(a) and V(b).
[00286] An intermediate of Formula VI(a) or Vl(b):
where R7, X, R10, R12, and R14 are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art.
In particular, for VI(b) see for example W02000042022 and WO2001005390 which are incorporated by reference herein.
[00287] An intermediate of Formula VII(a) or VII(b):
VH(a);
VII(b) where R7, X, Ri0, R12, and R14 are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art.
For intermediate VII(b) see, for example, W02001005390 and W02000042022 which are incorporated by reference herein.
[00288] An intermediate of Formula VIII(a) or VIII(b):
VIII(a);
VIII(b) where R7, X, R10, R32, Ri4, and R19 are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art.
In particular for formula VIII(b) where Ri0 is halo (particularly fluoro), R12 is hydrogen, R34 is hydrogen, and R19 is hydrogen or alkyl (particularly methyl) or alkenyl (particularly allyl), see WO 05/023251, W02005009975, and WO2001005390 which are incorporated by reference herein. In particular for VIII(a) where X is halo (particularly chloro or fluoro) or alkyl (particularly methyl), R is halo (particularly iodo, bromo, or chloro) or haloalkoxy (particularly trifluormethoxy), Rf0 is halo (particularly fluoro or chloro), Ri4 is hydrogen or alkyl (particularly methyl), and R39 is hydrogen or alkyl (particularly methyl), see for example US 2004/0116710, WO 03/077914, WO 03/077855, WO 00/42022, W02005009975, and WO2001005390 which are incorporated by reference herein. The following intermediates were prepared using similar procedures described in US 2004/0116710, WO 03/077914, WO 03/077855, WO 00/42022, W02005009975, and WO2001005390: 5-[(4-bromo-2-chlorophenyl)amino]-4-fluoro-l-methyi-li:/-benzimidazole-6-carboxylic acid and 4-fluoro-5-[(2-fluoro-4-iodophenyl)amino]-1 -methyl-1 //-benzimidazole-6-carboxylic acid.
[00289] An intermediate of Formula IX:
IX where R7, X, R10, R12, R14, and R56 are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art.
In particular, where Ri0 is hydrogen or halo (particularly chi or o or fluoro); R12 is hydrogen; R14 is hydrogen, amino, alkylamino, or dialkylamino; Ri6 is hydrogen; X is halo (particularly chloro); and R7 is halo (particularly bromo) see for example WO 05/023759, US 2005/0054701, US 2006030610, US 2005049419, and US 2005049276 which are incorporated by reference herein. The following intermediates were prepared using similar procedures as those described in WO 05/023759, as well as US 2006030610 and US 2005/0054701: 7-[(4-bromo-2-chlorophenyl)amino]-8-ch!oroimidazo[ 1,2-a]pyridine-6-carboxylie acid and 8~chloro-7-[(2-fluoro~4-iodophenyl)amino]imidazo[ 1,2-a]pyridine-6-carboxylic acid. The following intermediates can be prepared using similar procedures described in the references given above: 8-FhiorQ~7-[(2-fluoro~4-iodophenyl)aniino]imidazo[l,2-«]pyridine-6-carboxylic acid and 7-[(4-Bromo-2"fluorophenyl)amino]-8-fluoroimidazo[l,2~ o]pyridine-6-carboxylic acid.
[00290] An intermediate of Formula X(a) and X(b):
X(a);
X(b) where R7, X, R10, R12, and R14 are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art.
In particular, where R10 is hydrogen, halo (specifically chloro), or alkyl (specifically methyl), R.12 is hydrogen, and R14 is hydrogen, halo (specifically bromo), see for example WO 06/045514 which is incorporated by reference herein. To prepare the intermediate of Formula X(b), the nitrogen in the pyridine ring of X(a) can then be oxidized with an agent such as MCPBA or H2O2. The following X(a) and X(b) intermediates were prepared using similar methods as disclosed in WO 06/045514: 3-[(2-Fluoro-4-iodophenyl)amino]pyridine-4-carboxylic acid and 3-[(2-Fluoro-4-iodophenyl)amino]pyridine-4-carboxylic acid 1-oxide. The following X(a) intermediates can be prepared using similar methods as disclosed in WO 06/045514: 2-Fluoro-3-[(2-iluoro-4-iodophenyl)amino]pyridine-4-carboxylic acid and 3-[(4-Bromo-2-fluorophenyl)amino]pyridine-4-carboxylic acid. 100291] An intermediate of Formula XI(a):
XI(a);
XI(b) where R7, X, R10, R12, and R14 are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art.
In particular, where R10 is hydrogen, R12 is hydrogen or halo (particularly chloro or fluoro), R14 is amino or halo (particularly chloro), X is halo (particularly chloro), and R7 is halo (particularly bromo) see for example US 2005/0054701, US 200549419, and US 2006030610 which are incorporated by reference herein. The intermediate of Formula XI(b) can be prepared by oxidizing the nitrogen in the pyridine ring of XI(a) with an agent such as MCPBA or H2O2.
[00292] An intermediate of Formula XII:
XII where R7, X, R10, R12, Ri4, and R16 are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art.
In particular, see for example WO 05/051302 which is incorporated by reference herein. The following intermediates can be prepared using similar methods as disclosed in WO 05/051302: 8-Fluoro-7-[(2-fluoro-4-iodophenyl)amino]~4-methylcinno!ine~6-carboxylic acid; 7-[(4~Bromo-2-chlorophenyl)amino]-8-fluoro-4-methylcinnoline~6-carboxylic acid; 7-[(4-Bromo-2-fluorophenyl)amino]-8-fluoro-4-methyicinnoline-6-carboxylic acid; and 7-[(4-BroniO"2-fluorophenyl)amino]cinnoline-6-carboxyiic acid.
[00293] An intermediate of Formula XIII:
XIII where R7, X, R!0, Ri0a, and Y! are as defined in the Summary of the Invention for Group C can be prepared using procedures known to one of ordinary skill in the art, including for example the procedures in US 05/0256123, Wallace, E. M. et al. J. Med, Chem. 2006, 49, 441-4, WO 2005000818, and WO 2005051301 (where Y1 is carbon) which are incorporated by reference herein. 4-[(4~Bromo-2-fluorophenyl)amino]-5~ fluoro-1 -methyI-6-oxo-l,6-dihydropyridine-3-carboxylic acid was prepared using similar procedures to those disclosed in US 05/0256123 and WO 2005051301. 4-Chloro-1 -methyl-6-oxo-1,6~dihydropyridazine-3~carboxylic acid was prepared using similar procedures to those disclosed in US 2005256123.
The following intermediates can be prepared using the methods disclosed in the above references: 4-[(2-Fluoro-4-iodophenyl)amino]-1 -methyl-6-oxo-1,6-dihydropyridine-3 -carboxylic acid; 4-[(4-Bromo-2-chlorophenyl)arnino]-1 -methyl-6-oxo-1,6-dihydropyridine-3-carboxylic acid; 4-[(4-Bromo-2-fluorophenyl)arnino]-1-methyl~6-oxo-l,6-dihydropyridine-3- carboxylic acid; 4- [(4-Bromo-2-chlorophenyl)amino] -1 -methyl-6-oxo-1,6~dihydropyridazine-3-carboxylic acid; 4-[(4-Bromo-2-chlorophenyl)amino]-5-fluoro~ 1 -methyl-6-oxo-1,6-dihydropyridazine- 3- carboxylic acid; and 4- [(4-Bromo-2-fluorophenyl)amino]-1 -methyl-6-oxo-1,6-dihydropyridazine-3 -carboxylic acid.
[00294] An intermediate of Formula XIV:
XIV where R7, X, R10, and Ri4 are as defined in the Summary of the Invention for Group B can be prepared using procedures known to one of ordinary skill in the art. In particular, see for example WO 05/051302 which is incorporated hv reference herein.
[00295] An intermediate of Formula XVI
XVI where X and R7 are as defined in the Summary of the Invention for a Compound of
Group B can be prepared using procedures known to one of ordinary skill in the art. In particular, see for example WO 2001005390 and WO 2000042022 for procedures that can be used to prepare the following: 5-[(2-Fluoro-4-iodophenyl)amino]-l//-benzotriazole-6-carboxylic acid; 5-[(2-Fluoro-4-iodopheny!)amino]-1 -methyl-1H-benzotriazole-6-carboxylic acid; and 4-Fluoro-5~[(2-fiuoro-4-iodophenyl)amino]-1H-benzotriazole-6-carboxylic acid.
[00296] An intermediate of Formula XVII
XVII where X and R7 are as defined in the Summary of the Invention for a Compound of Group B can be prepared using procedures known to one of ordinary skill in the art. In particular, see Example 29, [00297] An intermediate of Formula XVIII(a) or XVIII(b)
XVIII(a) XVIII(b) where X R7 R40 and R40a are as defined in the Summary of the Invention for a Compound of Group D can be prepared using procedures known to one of ordinary skill in the art. In particular, the halo precursors to XVIII(a) and XVIii(b) and respectively
can be prepared using procedures similar to those described in Machon and Dlugosz Acta Poloniae Pharmaceutica 1983,40(1), 1-6 and von Angerer, Science of Synthesis 2004,16, 379-572 (General Review written in English). The halo precursors are then reacted with
using procedures known to one of ordinary skill in the ait and the synthetic methods disclosed herein. The following intermediates can be prepared as described above: 6-[(2-fluoro-4~iodophenyl)amino]-2~oxO“l ,2-dihydropyrimidine-5-carboxylic acid and 4-[(2-f!uoro-4-iodophenyl)amino]-2-oxo-1,2-dihydropyrimidine-5-carboxylic acid.
[00298] An intermediate of Formula XIX
XIX
__. _ *7 where X and K are as defined in the Summary of the Invention for a Compound of Group C can he prepared using methods known to one of ordinary skill in the art. In particular see US 2005049276.
[00299] An intermediate of Formula XX
XX where X and R/ are as defined in the Summary of the Invention for a Compound of Group C can be prepared using methods known to one of ordinary skill in the art. In particular see US 2005049276.
[00300] The synthesis of azetidines substituted at the 3-position can be conveniently carried out according to Scheme 1:
Scheme 1
starting from the iV-diphenylmethyl protected azetidin-3-ol (1), readily prepared by reaction of epichlorohydrin and diphenylmethylamine (Chatterjee, Shym 5,; Toggle, D. J Chemical Communications (London) 1968, 2, 93). Protecting group exchange, from Boc to CBz, on the azetidine is carried out according to literature protocols (Greene, T.W., Wuts, P.G. Protective Groups in Organic Synthesis, Wiley-
Interscience) and subsequent oxidation to the azetidinone (2) where P is CBz provides a useful intermediate for the preparation of compounds of the invention.
[00301] For example, the ketone intermediates of formula 2 can be broadly functionalized at the 3-position according to Scheme 2.
Scheme 2
An intermediate of formula (3), where R4 is as defined in the Summary of the Invention for a compound of Group A, Group B, Group C, or Group D can be prepared by reacting the intermediate 2 with Grignard reagents or other organometallic species of formula 17, such as organolithiums. Alternatively, the intermediate 2 can be reacted with nitroalkane anions of formula 18 prepared in-situ as in the Henry reaction (The Henry reaction, recent examples: Luzzio, F. A. Tetrahedron 2001, 57(6), 915-945) to give (4) where R4’ is hydrogen or alkyl optionally substituted as described for R4 in the Summary of the Invention for a compound of Group A, Group B, Group C, or Group D. Alternatively, the intermediate 2 can be reacted with ketone or aldehyde anions of formula 19 in a Ciaisen-type condensation to give (5) where R4’ is alkyl optionally substituted as described for R4 in the Summary of the Invention for a compound of Group A, Group B, Group C, or Group D and R4” is hydrogen or R4’. In addition, 2 can be reacted with Wittig reagents of formula 20 (where R’ and R” are independently hydrogen, alkyl, alkenyl, aryl, or heteroaryl and the alkyl, alkenyl, aryl, and heteroaryl are optionally substituted as described for R4 in the Sumnmary of the Invention for a compound of Group A, Group B, Group C, or Group D) to prepare intermediates of formula 6, which are also useful as precursors for compounds of the invention, [00302] According to Scheme 3, intermediates of formula (6) where where (R’ and R” are hydrogen and P is a nitrogen-protecting group such as CBz or Boc)
Scheme 3
can be further converted to the corresponding epoxide (7) and subsequent reaction with a suitable nitrogen base or other nucleophiles may be carried out to give access to a broad range of azetidin-3-ol derivatives such as (8), where R8 and R8 are as defined in the Summary of the invention.
[00303] In some cases the preparation of optically pure compounds is desired where the azeti dine contains one or more stereocenters. Numerous techniques for the preparation of optically pure compounds through both resolution techniques and asymmetric synthesis are well known in the art. In one such case, an asymmetric synthesis methodology can be employed where an azetidine precursor of formula (2) is reacted with an intermediate of formula 21 where R’ is not hydrogen, as depicted in Scheme 4.
Scheme 4
One such useful approach makes use of Evans oxazolidinone methodology (Diastereoselective aldoi condensation using a chiral oxazolidinone auxiliary. Gage, James R.; Evans, David A. Organic Syntheses 1990, 68, 83-91). The condensation of an azetidinone (2) with the a chiral oxazolidinone in the presence of a base such as LDA affords an intermediate oxazolidinone (9), where P is a nitrogen-protecting group such as CBz or Boe, with diastereoselectivity. Treatment with lithium hydroxide in aqueous hydrogen peroxide gives carboxylic acid (10) which can be subject to Curtius rearrangement to provide the chiral oxazolidinone (11) then carried forward as required to a useful intermediate (12). Further protecting group manipulation and derivatization as required can be employed to prepare compounds of Formula I.
[00304] Alternatively, a racemic mixture of an intermediate of formula (13), useful to prepare a compound of Formula I where R3 is hydroxy and R4 is heterocycloalkyl (in particular, where R4 is a N-protected piperidine), can be prepared according to Scheme 5,
Scheme 5
In the reaction schemes P and P" are orthogonal nitrogen-protecting groups. For example, P! is Boc and P2 is CBz or P1 is CBz and P2 is Boc. The reaction is carried out in-situ by treating 22 to generate the lithated amine and by subsequently treating it with a ketone such as (2) according to the method of Peter Beak (Beak, Peter; Lee, Won Koo α-Lithioamine synthetic equivalents: syntheses of diastereoisomers from the Boc-piperidines. Journal of Organic Chemistry 1990, 55(9), 2578-80). The racemate (13) thus prepared can be resolved by functionalization, as depicted in Scheme 6, with a chiral acid such as the readily-available Mosher acid (14).
Scheme 6
The resulting diastereomeric esters (15) can be separated by chromatographic means and then carried forward individually as the enantiomerically pure intermediates (R)~ (16) and (8)-(16).
[00305J Compounds of the Invention can be prepared by reacting an intermediate of Formula II, 111(a), 111(b), IV(a), IV(b), V(a), V(b), VI(a), VI(b), VII(a), VII(b), VIII(a), VIII(b), IX, X(a), X(b), XI(a), XI(b), XII, XIII, XIV, XVI, XVII, XVIII(a), XVIII(b), XIX, or XX with intermediate 17 according to Scheme 7:
Scheme 7
The reaction is carried out in a solvent such as DMF, THF, or DCM in the presence of a base such as DIPEA, V-methylmorpholine, DMAP, or triethylamine and optionally in the presence of a coupling agent such as PyBOP, HBTU, or EDCI.
[00306] Alternatively an intermediate of Formula II, 111(a), 111(b), IV(a), IV(b), V(a), V(b), VI(a), VI(b), VII(a), Vll(b), VIII(a), Vlll(b), IX, X(a), X(b), XI(a), XI(b), XII, XIII, XIV, XVI, XVII, XVIII(a), XVIII(b), XIX, or XX can be converted into an acid halide according to Scheme 8
Scheme 8
where Xz is halo, such as chloro or fluoro, and all oilier groups are as defined in the Summary of the Invention for a compound of Group A, Group B, Group €, or Group D. The reaction is carried out in a solvent such as dioxane, THF, or DCM in the presence of a base such as DIPEA, sodium bicarbonate. The acid halide of formula 18 can then be reacted with an azetidine intermediate of formula 17 to prepare a compound of Formula I.
Synthetic Examples [00307] Generally, the compounds listed below were identified by LC-MS, and/or isolated, and characterized by "H-NMR (most typically 400 MHz). Liquid chromatography-mass spectral (LC-MS) analyses were performed using at least one of: a Hewlett-Packard Series 1100 MSD, an Agilent 1100 Series LC/MSD (available from Agilent Technologies Deutschland GmbH of Waldbronn Germany), or a Waters 8-Channel MUX System (available from Waters Corporation of Milford, Massachusetts). Compounds were identified according to either their observed mass [MH4] or [MNaT] ion (positive mode) or [MH'j ion (negative mode). 'H-NMR data for compounds was taken with a Varian AS400 Spectrometer (40GMHz, available from Varian GmbH, Darmstadt, Germany). Starting materials and intermediates used to prepare a compound of the invention are either commercially available or can be prepared by one of ordinary skill in the art.
Reference 1 3,4-difluoro-2-[(2-fluoro-4-iodopheny!)amino] benzoyl fluoride
[00308] To a stirred mixture of 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino] benzoic acid (12 g, 30,5 mmol), prepared using procedures similar to those described in US 7,019,033, in dichloromethane (70 mL) at 0 °C was added pyridine (2.5 mL, 30.8 mmol) followed by dropwise addition of cyanuric fluoride (2.8 mL, 33.6 mmol). The reaction mixture was stirred at 0 °C for 10 minutes and then warmed to room temperature and stirred for 2 hours. The reaction mixture was diluted with water and extracted with dichloromethane (100 mL). The aqueous layer was extracted once with dichloromethane (50 mL). The combined organic layers were washed with saturated aqueous sodium bicarbonate solution, brine, dried over anhydrous sodium sulfate and concentrated in vacuo to give crude product as a brownish solid. Crude product was purified by flash chromatography (plug, 25% ethyl acetate in hexanes) to afford 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino] benzoyl fluoride as a beige solid (11.8 g, 97% yield). 'H NMR (400MHz, CD3OD): 8.41 (s, 1H), 7.80-7.81 (m, 1H), 7.52 (dd, 1H), 7.43-7.47 (m, 1H), 6.96-7.03 (m, 1H), 6.85-6.92 (m, 1H).
Reference 2 2-}(4-bromo-2-chlorophenyl)amino]-3,4-difluorobenzoic acid
[00309] To a solution of 2,3,4-trifluorobenzoic acid (1 g, 5.68 mmol) and 4-bromo-2-chloroaniline (1.2 g, 5.68 mmol) in acetonitrile (10 mL) was added lithium amide (0.39 g, 17.04 mmol) and the reaction stirred at 60 °C for 1.5 hours. The mixture was cooled to room temperature and then to 0 °C and acidified with aq. hydrochloric acid. The obtained precipitate was collected by filtration and washed with cold water and dried in vacuo to afford 2-[(4-bromo-2-chlorophenyl)amino]-3,4-difluorobenzoic acid (1,92 g, 94% yield) as a beige solid. MS (El) for CisI UBrC^NCh;: 363 (MH*).
[00310] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, 2-[(4-iodo-2-fluorophenyl)amino]-3-fluorobenzoic acid was prepared. MS (El) for C13H8F2INO2: 376 (ΜϊΓ).
Reference 3
Phenylmethyl l-oxa-5-azaspiro[2.3]hexane-5-carboxyIatc
[00311] To a solution of azetidin-3-ol hydrochloride in tetrahydrofuran (90 mL) and water (10 mL) was added triethylamine (15 mL, 0.106 mol) followed by slow addition of benzyl chloroformate (8.0 mL, 0.056 mol) at room temperature. The reaction mixture was stirred at room temperature for 16 hours then partitioned with water and ethyl acetate. The organic layer was washed with brine, dried over anhydrous magnesium sulfate and concentrated. The residue was purified by flash chromatography (Si02, 25-50% ethyl acetate in hexanes) to afford phenylmethyl 3-hydroxyazetidine-1 -carboxylate (3.56 g, 33% yield) as a clear and colorless oil. 'H NMR (400 MHz, CDC13): 7.36-7.31 (m, 5H), 5.09 (s, 2H), 4.64-4.57 (m, 1H), 4.22 (dd, 2H), 3.88 (dd, 2H), 2.61 (d, 1H, ./=4.0 Hz). MS (El) for CnH13N03: 208 (MH+).
[00312] To a solution of phenylmethyl 3-hvdroxyazetidine-1 -carboxylate (3.5 g, 0.0168 mol) in dichloromethane (100 mL) was added Dess-Martin periodinane (10.7 g, 0.0.25 mol) at room temperature and stirred for 5 h. The reaction mixture was quenched with 1:1 ratio of saturated aqueous sodium bicarbonate and 1M sodium thiosulfate (200 mL) and then partitioned with dichloromethane. The organic layer was dried over anhydrous magnesium sulfate and concentrated in vacuo to afford phenylmethyl 3-oxoazetidine-1 -carboxylate (3.43 g, 99% yield) as a clear and colorless oil without further purification. 'H NMR (400 MHz, CDC13): 7.39-7.31 (m, 5H), 5.17 (s, 2H), 4.77 (s, 4H). MS (El) for C,iHnN03: 205 (M+).
[00313] A suspension of methyltriphenylphosphonium bromide (23.0 g, 0.0649 mol) and potassium /eri-butoxide (7,3 g, 0.0649 mol) in diethyl ether (140 mL) was stirred at room temperature for 20 min, and then heated to 35 °C for 1 h. To this bright yellow reaction mixture was slowly added a dilute solution of phenylmethyl 3-oxoazetidine-1 -carboxylate (3.33 g, 0.0162 mol) in diethyl ether (50 mL). The reaction mixture was stirred at 35 °C for 12 hours then filtered through a bed of celite and rinsed with ethyl ether. The filtrate was washed with water and brine, dried over anhydrous magnesium sulfate and concentrated. The residue was purified by flash chromatography (Si02, 5-10% ethyl acetate in hexanes) to afford phenylmethyl 3-methylideneazetidine-l -carboxylate (2.46 g, 75% yield) as a clear and colorless oil). *H NMR (400 MHz, CDC13): 7.27-7.22 (m, 5H), 5.02 (s, 2H), 4.93-4.90 (m, 2H), 4.48-4.47 (m, 4H). MS (El) for CJ2Hi3N02: 203 (M4).
[00314] To a solution of phenylmethyl 3-methylideneazetidine-l -carboxylate (2.46 g, 0.0121 mol) in chloroform (100 mL) was added 3-chloroperoxybenzoic acid (12.5 g, 0.0726 mol) at 0 °C, The reaction mixture was allowed to warm up to room temperature over a period of 12 hours then quenched with 1 M sodium thiosulfate / saturated aqueous sodium bicarbonate (1:1). The layers were separated and the organic layer was dried over anhydrous magnesium sulfate then concentrated. The residue was purified by flash chromatography (5-15% ethyl acetate in hexanes) to afford phenylmethyl l-oxa-5-azaspiro[2.3]hexane-5-carboxylate (2.2 g, 83% yield) as clear and colorless oil. !H NMR (400 MHz, CDC13): 7.37-7.29 (m, 5H), 5.12 (s, 2H), 4.35-4.26 (m, 4H), 2.85 (s, 2H). MS (El) for Ci2Hi3N03: 220 (MH+).
Reference 4 4-(2-fluoro-4-iodophenylamino)-l-methyI-6-oxo-l,6-dihydropyridazine“3- carboxylic add
[00315] 4-chloro-1 -methyl-6-oxo-1,6-dihydropyridazine-3 -carboxylic acid was prepared using procedures similar to those disclosed in US 2005256123.
[00316] To a solution of 4-chloro-l-methyl-6-oxo-l,6-dihydropyridazine-3-carboxylic acid (350 mg, 1.855 mmol) and 2-fluoro-4-iodoaniline (1.06 g, 4.453 mmol) in tetrahydrofuran (13.3 mL) was sparged with nitrogen for 5 minutes followed by the slow addition of lithium bis(trimethylsilyl)amide, 1.0 M in THF (7.4 mL). The reaction mixture stirred for an additional 4 hours at room temperature. The mixture was quenched with 1 N HC1 and concentrated in vacuo. The residue was partitioned between ethyl acetate and 1 N aqueous HC1. The aqueous layer was extracted (3x) with ethyl acetate. The combined organic layer was washed with brine, dried over anhydrous sodium sulfate, filtered and evaporated to afford 4-(2-fluoro-4~ iodophenylamino)-l-methyl-6-oxo-l,6-dihydropyridazine-3-carboxylic acid (939 mg, 100% yield). !H NMR (CDCI3): 7.27 (dd, 1H), 7.21 (d, 1H), 6.54 (t, 1H), 4.84 (broad s, 2H), 2.09 (s, 1H), 1.26 (t, 3H); MS (El) for C12H9N3O3FI: 389 (MH+).
[00317] A solution of 4-(2-fluoro-4-iodophenylamino)-1 -methyl-6-oxo- 1,6-dihydropyridazine-3-carboxylic acid (939 mg, 2.413 mmol) in dichloromethane (60 mL) in the presence of dimethylformamide (8.0 mL) was cooled to 0 °C.
Malonyl chloride (1.26 mL, 14.48 mmol) was added and stirred at room temperature for 1 hour. The reaction mixture was evaporated and partitioned between ethyl acetate and 1M aqueous ammonium chloride. The aqueous layer was extracted lx with ethyl acetate. The combined organic layers were washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 4-(2-fluoro-4-iodophenylamino)-l -methyl-6-oxo-1,6-dihydropyridazine-3-carbonyl chloride. This crude material was taken into the next step without further purification. MS (El) for C12H8N3O2CIFI: 408 (MH+).
[00318] To a solution of 4~(2-fluoro-4-iodophenylarnino)~ 1 -methyl-6-oxo- 1.6- dihydropyridazine-3-carbonyl chloride in methanol (15 mL) and benzene (12 mL) was added drop wise trimethylsilyl diazomethane (1 mL) and stirred at room temperature for 15 minutes. The reaction mixture was quenched with acetic acid and evaporated. The residue was partitioned between ethyl acetate and brine. The organic layer was separated, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified on silica gel chromatography column (7:3 hexanes/ethyl acetate) to afford methyl 4-(2-f!uoro-4-iodophenylamino)-1 -methyl-6-oxo~l,6-dihydropyridazine-3"Carboxylate (84.9 mg, 8.7% yield). NMR (CDCI3): 7.49-7.56 (m, 3H), 7.12 (t, 1H), 6.13 (d, 1H), 4.00 (s, 3H), 3.83 (s, 3H); MS (El) for C33HHN3O3FI: 404 (MH+).
[00319] Methyl 4-(2~fluoro-4-iodophenylainino)-1 -methyl-6-oxo-1,6-dihydropyridazine-3-carboxylate (84.9 mg, 0.211 mmol) was dissolved in tetrahydrofuran (5 mL), methanol (2.5 mL) and water (2.5 mL). Aqueous 2 M lithium hydroxide (200 pL) was added at room temperature. After 10 minutes, the reaction mixture was heated to 50 °C for 30 minutes and continued to stir at room temperature for 16 hours at which time the solvents were evaporated. The residue was made acidic with 2 M aqueous hydrochloric acid to pH 2 and extracted with ethyl acetate. The organic layer separated, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to provide 4-(2-fluoro-4-iodophenylamino)-1 -methy 1-6-oxo- 1.6- dihydropyridazine-3-carboxylic acid (54.0 mg, 66% yield). MS (El) for C12H9N3O3FI: 390 (MH+).
Reference 5 1,1-dimethylethyl 2-(3-hydroxy-l-{[(phenyImethyl)oxy]carbonyl}azetidin-3- yl)piperidine-l -carboxylate
[00320] To a solution of 1,1-dimethylethyl piperidine-1 -carboxylate (0.50 g, 2.7 mmol) in anhydrous diethyl ether (9.0 mL) under anhydrous nitrogen gas was added Ar^Ai’y/V-tetramethylethane-1,2-diamine (0.41 mL, 2.7 mmol), and the solution was cooled to -78°C. To this solution was added (2-methvipropyl)lithium (2.1 mL, 1.4 M in cyclohexane, 3.0 mmol) in small portions. To this anion solution was added phenylmethyl 3-oxoazetidine-1 -carboxylate (1.0 g, 5.4 mmol), prepared using procedures as described in Reference 3, in anhydrous ether (2.0 mL), while maintaining the internal temperature at less than -60°C. The solution was allowed to warm to room temperature and stirred overnight. The reaction was quenched with water, and partitioned between water and diethyl ether. The layers were separated and the aqueous layer was extracted with diethyl ether twice. The combined organic layers were dried (magnesium sulfate), filtered and concentrated in vacuo. Chromatography (silica gel, 3:1 hexanes/ethyl acetate) gave 0.13 g (13%) of 1,1-dimethylethyl 2-(3 -hydroxy-1 - {[ (phenvlmethyl)oxy] carbonyl} azetidin-3 - yl)piperidine-1 -carboxylate. !H NMR (400 MHz, CDG3): 7.31 (m, 5H), 5.08 (s, 2H), 4.05 (d, 1H), 4.00 (d, 1H), 3.84 (d, 2H), 3.80 (broad s, 1H), 3.55 (broad s, 1H), 3.10 (broad s, 1H), 1.92 (m, 1H), 1.45-1.62 (m, 6H), 1.43 (s, 9H). MS (El) for C21H30N2O5: 335 (M-tBu), 315 (M-OtBu), EXAMPLE 1 H{3,4-difluoro-2-[(2-flu0ro-4~iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol
[00321] 3,4-Difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoic acid (2.1 g, 5.3 mmol), prepared using procedures similar to those in US 7,019,033, was taken into DMF (10 mL) followed by addition of PyBOP (2.6 g, 5.3 mmol) and the mixture was allowed to stir at room temperature over 15 minutes. Az.etidin-3-ol hydrochloride (870 mg, 8.0 mmol) and DIPEA (1.85 mL, 11.2 mmol) was then added and the mixture was allowed to stir an additional hour at room temperature. The mixture was then partitioned with ethyl acetate and 0.5 M aqueous sodium hydroxide solution.
The organic layer was then washed with water (3x) then brine and dried over anhydrous sodium sulfate. Filtration and concentration followed by silica gel flash chromatography using ethyl acetate: hexanes (5:1) eluent afforded l-({3,4-difhioro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol (2.09 g, 87% yield) as a colorless amorphous solid. !H NMR (400 MHz, CDCI3): 8.47 (s, 1H), 7.39 (dd, 1H), 7.32 (d, 1H), 7.13-7.09 (m, 1H), 6.84-6.78 (m, 1H), 6.63-6.57 (m, 1H), 4.74-4.67 (m, 1H), 4.43-4.39 (m, 2H), 4.20-3.96 (br d, 2H), 2.50 (d, 1H).
[00322] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the compounds in Examples l(a)-(e) were prepared. EXAMPLE 1(a). l-[1-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}-carbonyl)azetidin-3-yl]-A(/V-dimethylpyrrolidin-3-amine. The title compound was prepared by reacting 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoic acid with ;V-methyl-Ar-(2-(pyridin~2-yl)ethyl)azetidin~3-amine. The azetidine intermediate was prepared using procedures similar to those described in Abdel-Magid, et.al, Tetrahedron Letters 1990,31(39), 5595 starting with ter/-butyl 3-oxoazetidine-l-carboxylate, which itself was prepared as described in Example 3. The title compound: *H NMR (400 MHz, d6-DMSO): 8.56 (s, 1H), 7.58 (m, 1H), 7.38 (d, 1H), 7.31 (m, 1H), 7.16 (m, 1H), 6.67 (m, 1H), 4.16 (m, 1H), 3.97 (m, 2H), 3.77 (m, 1H), 3.26 (br s, 4H), 2.63 (m, 1H), 2.42 (br s, 6H), 1.99 (br s, 1H), 1.74 (br s, 1H). MS (El) for C22H24F3IN4O: 545 (MH4), EXAMPLE 1(b). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-iV-methyl-iV-(2-pyridin-2-ylethyl)azetidin-3-amine. The title compound was prepared by reacting 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoic acid with l-(azetidin-3-yl)~N,N-dimeihylpyrrolidin-3-amine. The azetidine intermediate was preparedusing procedures similar to those described in Abdel-Magid, et.al., Tetrahedron Letters 1990, 31(39), 5595 starting with /erf-butyl 3-oxoazetidine-1 -carboxylate, which itself was prepared as described in Example 3. The title compound: lH NMR(400 MHz, CD3OD): 8.50 (d, 1H), 7.94 (t, 1H), 7.50-7.30 (m, 5H), 7.07 (q, 1H), 6.66- 6.61 (m, 1H), 4.52-4.48 (m, 2H), 4.3 l(s, 2H), 4.23-4.18 (m, 1H), 3.48- 3.46 (m, 2H), 3.17-3.13 (m, 2H), 2.88 (s, 3H); MS(EI) for C24H22F3IN4O: 567 (MH*). EXAMPLE 1 (c). 6-(Azetidin-1 -ylcarbonyl)-2,3-difluoro-.¥-(2-fluoro-4-iodophenyl)aniline: 'HNMR (400 MHz, CDCI3): 8.57 (s, 1H), 7.41-7.38 (dd, 1H), 7.34-7.31 (dt, 1H), 7.13-7.09 (m, 1H), 6.83-6.77 (m, 1H), 6.64-6.58 (m, 1H), 4.27 (b, 2H), 4.18 (b, 2H), 2.38-2.30 (p, 2H); MS (El) for C16H12F3IN3O: 433 (MH+). EXAMPLE 1(d). [l-({3,4-Difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]methanol: *H NMR (400 MHz, CDCI3): 8.52 (s, 1H), 7.41-7.38 (dd, 1H), 7.34-7.31 (dt, 1H), 7.15-7.11 (m, 1H), 6.83-6.77 (m, 1H), 6.64-6.58 (m, 1H), 4.29-4.20 (m, 2H), 4.09 (b, 1H), 3.93 (b, 1H), 3.82-3.81 (d, 2H), 2.89-2.75 (m, 1H); MS (El) for C17H14F3IN2O2: 463 (MH‘). EXAMPLE 1(e). 1 -({3,4-Difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidine-3-carboxylic acid: 'H NMR (400 MHz, CDCI3): 7.79 (b, 2H), 7.42-7.38 (dd, 1H), 7.34-7.32 (dt, 1H), 7.15-7.11 (m, 1H), 6.89-6.83 (m, !H), 6.65-6.60 (m, 1H), 4.46-4.29 (m, 4H), 3.55-3.47 (m, 1H); MS (El) for Ci?Hi2F3IN203: 477 (MH+). EXAMPLE 2 iV-[l-({3,4-Din«©r©”2”[(2-fluoro~4-iodophenyl)amino!pheny!}carbonyl)az;etidin-3~ yl] ~N2,N2-d iethy Igly cinamide
[00323] A solution of 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoic acid (200 mg, 0.51 mmol), prepared using procedures similar to those in US 7,019,033, PyBOP (256 mg, 0.51 mmol), commercially available /eri-butyl azetidin-3- ylcarbamate (131 mg, 0.77 mmol) and N, A-diisopropylethylamine (180 pL, 1.02 mmol) in dimethylformamide (3 mL) was stirred at room temperature for 15 hours. The reaction mixture was partitioned between 5% aqueous lithium chloride and ethyl acetate. The organic portion was washed with 20% aqueous citric acid, saturated aqueous sodium bicarbonate, brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a brown residue which was purified by silica gel column chromatography eluting with 30% ethyl acetate in hexanes to afford 1,1-dimethylethyl [ 1 -({3,4-difluoro-2- [(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]carbamate (225 mg, 80% yield) as a colorless oil. 'H NMR (400 MHz, DMSO): 8.56 (s, 1H), 7.60-7.55 (m, 2H), 7.38 (d, 1H), 7.30-7.26 (m, 1H), 7.20-7.13 (m, 1H), 6.71-6.66 (m, 1H), 4.37-4.20 (m, 2H), 4.18-4.06 (m, 1H), 3.98-3.93 (m, 1H), 3.82-3.75 (m, 1H), 1.37 (s, 9H). MS (El) CatH^OsFal: 548 (Mlf).
[00324] A solution of 1,1-dimethylethyl [ 1 -({ 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]carbamate (113 mg, 0.20 mmol) and trifluoroacetic acid (500 pL) in dichloromethane (2 mL) was added stirred at room temperature for one hour then was partitioned between saturated aqueous sodium bicarbonate, and dichloromethane. The organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated to afford a colorless residue which was purified by column chromatography eluting with 10% methanol in dichloromethane to afford 1 -({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-amine (85 mg, 95% yield) as a white foam. SH NMR (400 MHz, CDC13): 8.53 (s, 1H), 7.39 (d, 1H), 7.32 (d, 1H), 7.13-7.09 (m, 1H), 6.84-6.77 (m, 1H), 6.63-6.57 (m, 1H), 4.46-4.39 (m, 2H), 3.98-3.75(br m, 4H); MS (El) for C,6Hi3F3lN30: 448 (MH*).
[00325] A solution of 1-((3,4-difluoro~2-[(2-fluoro-4- iodophenyl)amino]phenyi}carbonyl)azetidin-3-amine (100 mg, 0.22 mmol), PyBOP (131 mg, 0.25 mmol), N,iV-diisopropylethylamine (80 pL, 0.44 mol) and bromoacetic acid (35 mg, 0.25 mmol) in dimethylformamide (1 mL) was stirred at room temperature for 15 hours. The reaction mixture was concentrated in vacuo and the resultant residue was purified by column chromatography eluting with 80% ethyl acetate in hexanes to afford 2-bromo~Ar-[ 1 -({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)azetidin-3 -yljacetamide (102 mg, 82% yield) as a white foam. MS (El) for CigH]4BrF3iN302: 568.
[00326] A solution of 2-bromo-Ar-[l-({3,4~difluoro~2-[(2-f!uoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]acetamide (30 mg, 0.05 mmol) and N, A-diethylamine (100 pL, excess) in dichloromethane (2 mL) was stirred at room temperature for 15 hours. The reaction mixture was concentrated in vacuo and purified by preparative reverse phase HPLC (CH3CN/H2O with 0.1% TFA). Isolated product was concentrated in vacuo to afford N-{\-( {3,4-difluoro-2-[(2-fluoro~4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]-N2.iV2-diethylglydnainide trifluoroacetate salt (13.0 mg, 38% yield) as a white solid. 'H NMR (400 MHz, CDCI3): 9.36 (br s, 1H), 9.25 (d, 1H), 8.60 (s, IH), 7.60 (d, 1H), 7.40 (d, 1H), 7.33- 7.27 (m, IH), 7.22-7.15 (m, 1H), 6.73-6.66 (m, 1H), 4.54-4.40 (m, 2H), 4.25-4.20 (m, 1H), 4.04-3.82 (m, 4H), 3.17-3.12 (m, 4H), 1.18-1.15 (m, 6H); MS (El) C22H24F3IN4O2: 561 (MH+).
[00327] Using the same or analogous synthetic techniques and/or substituting with alternative reagents, the compounds in Examples 2(a)-(n) were prepared. EXAMPLE 2(a). 1,1-Dimethylethyl [1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]carbamate: 'H NMR (400 MHz, CDCI3): 8.52 (br s, IH), 7.40 (dd, 1H), 7.33 (dt, IH), 7.13-7.07 (m, 1H), 6.80 (ddd, 1H), 6.61 (ddd, IH), 5.01-4.88 (br, IH), 4.55-4.37 (br, 4H), 4.05 (br d, IH), 1.43 (s, 9H); MS (El) for C21H21F3IN3O3S: 548 (MH+). EXAMPLE 2(b). !-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-amine trifluoroacetate salt: ]H NMR (400 MHz, CDCI3): 8.53 (s, IH), 7.39 (d, IH), 7.32 (d, IH), 7.13-7.09 (m, IH), 6.84-6.77 (m, IH), 6.63-6.57 (m, IH), 4.46-4.39 (m, 2H), 3.98-3.75(br m, 4H); MS (El) for C16H13F3IN3O: 448 (MH+). EXAMPLE 2(c). iV-[l-({3,4-difluoro-2-[(2-fluoro-4- iodopheny l)amino] phenv 1} carbony l)azetidin-3 -y 1] -2-methylpropanamide: 1H NMR (400 MHz, DMSO): 8.60 (s, IH), 8.38 (d, IH), 7.59 (d, IH), 7.38 (d, IH), 7.32-7.28 (m, IH), 7.18-7.13 (m, IH), 6.72-6.66 (m, IH), 4.45-4.35 (m, IH), 4.18-3.77 (m, 4H), 2.36-2.28 (m, IH), 0.99 (d, 6H); MS (El) (^oH^AS^: 518 (MH+). EXAMPLE 2(d). A-[l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]formamide: lH NMR (400 MHz, DMSO): 8.69 (d, IH), 8.58 (s, IH), 8.02 (s, IH), 7.59 (d, IH), 7.39 (d, IH), 7.31-7.27 (m, IH), 7.19-7.13 (m, IH), 6.70-6.66 (m, IH), 4.55-4.46 (m, IH), 4.42-4.36 (m, IH), 4.20-4.16 (m, IH), 4.01-3.97 (m, IH), 3.82-3.79 (m, IH); MS (El) C17H13F3IN3O2: 476 (MHT). EXAMPLE 2(e). Ar-[l-((3,4-difiuoro-2-[(2-fluoro-4-
iodophenyl)amino]phenyl}carbonyi)azetidin-3-yl]-3,4-dihydroxybutanamide: !H NMR (400 MHz, DMSO): 8.60 (s, 1H), 8.47 (d, 1H), 7.59 (d, 1H), 7.39 (d, 1H), 7.31- 7.28 (m, 1H), 7.20-7.14 (m, 1H), 6.72-6.66 (m, 1H), 4.45-4.35 (m, 2H), 4.18-4.14 (m, 1H), 4.00-3.92 (m, 1H), 3.84-3.78 (m, 2H), 3.31-3.18 (m, 2H), 2.38-2.18 (m, 1H), 2.09-2.03 (m, 1H); MS (El) C20H19F3IN3O4: 550 (MH+). EXAMPLE 2(f). methyl [1 -((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]carbamate: *H NMR (400 MHz, DMSO): 8.58 (s, 1H), 7.84 (d, 1H), 7.59 (d, 1H), 7.39 (d, 1H), 7.35-7.27 (m, 1H), 7.20-7.13 (m, 1H), 6.71-6.66 (m, 1H), 4.38-4.25 (m, 2H), 4.17-4.12 (m, 1H), 4.00-3.97 (m, 1H), 3.83-3.78 (m, 1H), 3.53 (s, 3H); MS (El) CigH,5F3IN303: 506 (MH+). EXAMPLE 2(g). iV-[ 1 -({3,4-difluoro-2- [(2-fluoro-4- iodophenyl)amino]phenyl}carbonyI)azetidin-3-yl]-2-(4-methylpiperazin-l- yl)acetamide trifluoroacetate salt: ]H NMR (400 MHz, DMSO): 8.64 (s, 1H), 8.54 (d, 1H), 7.60 (d, 1H), 7.39 (d, 1H), 7.32-7.29 (m, 1H), 7.21-7.15 (m, 1H), 6.72-6.66 (m, 1H), 4.54-4.28 (m, 2H), 4.19-4.15 (m, 1H), 4.06-4.00 (m, 1H), 3.91-3.84 (m, 1H), 3.44-3.24 (m, 2H), 3.16-2.92 (m, 6H), 2.78 (s, 3H), 2.62-2.50 (m, 2H); MS (El) C23H25F3IN5Q2: 588 (MH+). EXAMPLE 2(h). A^[l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]-7V,A-bis(2-hydroxyethyi)glycinamide trifluoroacetate salt: !H NMR (400 MHz, DMSO): 9.19 (d, 1H), 7.60 (d, 1H), 7.41 (d, 1H), 7.31-7.27 (m, 1H), 7.21-7.15 (m, 1H), 6.73-6.66 (m, 1H), 4.51-4.40 (m, 2H), 4.23-4.18 (m, 1H), 4.05-3.98 (m, 3H), 3.86-3.82 (m, 1H), 3.75-3.69 (m, 3H), 3.32 (br s, 4H) C22H24F3IN4O4: 593 (MH*). EXAMPLE 2(i). N-[ 1-((3,4-difluoro-2-[(2-fluoro-4- i odophenyl)amino]phenyl} carbonyl)azetidin-3 -yl] -2-piperidin-1 -ylacetami de trifluoroacetate salt: !H NMR (400 MHz, DMSO): 9.20 (d, 1H), 7.60 (d, 1H), 7.41 (d, 1H), 7.31-7.27 (m, 1H), 7.21-7.15 (m, 1H), 6.73-6.66 (m, 1H), 4.52-4.40 (m, 2H), 4.24-4.18 (m, 1H), 4.05-4.00 (m, 1H), 3.87-3.80 (m, 3H), 3.40-3.32 (m, 2H), 3.00- 2.91 (m, 2H), 1.82-1.66 (m, 6H); MS (El) C23H24F3IN4O2: 573 (MH+). EXAMPLE 2(|). A-[l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]-N3-(2-hydroxyethyl)-N3-methyl-beta-alaninamide hydrochloride: SH NMR (400 MHz, DMSO): 9.36 (br s, 1H), 8.86 (d, 1H), 8.60 (s, 1H), 7.59 (d, 1H), 7.39 (d, 1H), 7.32-7.26 (m, 1H), 7.21-7.14 (m, ΙΗ), ο. /z-o.66 Cm, 1Η), 5.35-5.33 (m, 1H), 4.48-4.37 (m, 2H), 4.20-4.15 (m, 1H), 4.02-3.96 (m, 1H), 3.84-3.79 (m, 1H), 3.74-3.68 (m, 2H), 3.42-3.06 (m, 4H), 2.75 (s, 3H), 2.65-2.60 (m, 2H); MS (ΕΪ) C22H24F3IN4O3: 577 (MH*). EXAMPLE 2(k). Ar-[l-({3,4~difluoro-2-[(2-fluoro-4- iodophenyl)amino]pheny!}carbonyl)azetidin-3-yl]-N3,N3-bis(2-hydroxyethyl)-beta-alaninamide hydrochloride: !H NMR (400 MHz, DMSO): 9.39 (br s, 1H), 8.91 (d, 1H), 8.61 (s, 1H), 7.59 (d, 1H), 7.39 (d, 1H), 7.31-7.27 (m, 1H), 7.21-7.14 (m, 1H), 6.72- 6.66 (m, 1H), 5.31 (br s, 2H), 4.46-4.36 (m, 2H), 4.20-4.15 (m, 1H), 4.02-3.97 (m, 1H), 3.85-3.72 (m, 5H), 3.30-3.17 (m, 4H), 2.68-2.63 (m, 2H); MS (El) C23H26F3IN4O4: 607 (MH‘). EXAMPLE 2(m). iV-[1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]-N2-methylglycinamide trifluoroacetaie salt: *H NMR (400 MHz, DMSO): 9.09 (d, 1H), 8.69 (br s, 2H), 8.60 (s, 1H), 7.60 (d, 1H), 7.39 (d, 1H), 7.31-7.27 (m, 1H), 7.22-7.15 (m, 1H), 6.73-6.66 (m, 1H), 4.54-4.41 (m, 2H), 4.25-4.19 (m, 1H), 3.99-3.96 (m, 1H), 3.84-3.78 (m, 1H), 3.72- 3.67 (m, 2H), 2.58-2.54 (m, 3H); MS (El) C^HigFsIN^: 519 (MET). EXAMPLE 2(n). ?v-[l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyi)azetidin-3-yl]glycinamide trifluoroacetate salt: 'H NMR (400 MHz, DMSO): 8.59 (s, 1H), 8.46 (br s, 1H), 7.59 (d, 1H), 7.39 (d, 1H), 7.32-7.28 (m, 1H), 7.20-7.13 (m, 1H), 6.72-6.66 (m, 1H), 4.49 (br s, 1H), 4.40-4.35 (m, 1H), 4.18-4.13 (m, 1H), 4.05-4.01 (m, 1H), 3.86-3.81 (m, 1H), 3.07 (s, 2H); MS (El) Ci8Hi6F3IN402: 505 (MH*). EXAMPLE 3 l-({3)4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl)carbonyl)-3- (morpholin-4-ylmethyl)azetidin-3-ol
[00328| A mixture of 3-azetidinol hydrochloride (10 g, 91 mmol), di-teri-butyl dicarbonate (18.8 g, 86.3 mmol) and sodium bicarbonate (15.3 g, 182 mmol) in dioxane:water (400 mL, 1:1) was stirred at room temperature for 15 hours. The organic portion was removed in vacuo and the aqueous portion was extracted with ethyl acetate three times. The combined organic portion was washed with 5% aqueous HC1, water, brine, dried over sodium sulfate, filtered and concentrated invacua to afford 12.8 g, 74 mmol (81%) of 1,1 -dimethylethyl 3-hydroxyazetidine-1-carboxylate as a colorless oil without further purification. SH NMR (400 MHz, DMSO): 5.62 (d, 1H), 4.40-4.33 (m, 1H), 4.02-3.95 (m, 2H), 3.62-3.54 (m, 2H), 1.37 (s, 9H). GC/MSforCgH15N03: 173.
[00329] A solution of oxalyl chloride (545 pL, 6.36 mmol) in dichloromethane (25 mL) was cooled to -78 °C. While maintaining an internal temperature of-78 °C, the dropwise addition of DMSO (903 pL, 12.7 mmol) followed by 1,1 -dimethylethyl 3-hydroxyazetidine-1 -carboxylate (1 g, 5.78 mmol in 30 mL of dichloromethane) and finally triethylamine (3.25 mL, 23.1 mmol in 20 mL of dichloromethane) was performed. The mixture was allowed to warm to room temperature and was stirred for 15 hours. The reaction mixture was diluted with water and partitioned and the organic portion was washed twice with water. The combined aqueous portion was extracted once with dichloromethane. The combined organic portion was washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a yellow oil which was purified by column chromatography. Eluting with 30% ethyl acetate in hexanes, isolated product was concentrated in vacuo to afford 893 mg, 5.20 mmol (90%) of 1,1 -dimethylethyl 3-oxoazetidine-1 -carboxylate as a colorless oil, which solidified upon standing. 'H NMR (400 MHz, DMSO): 4.67 (s, 4H), 1.42 (s, 9H). GC/MS for QH0NO3: 171.
[00330] A mixture of potassium fer/-butoxide (15.5 g, 137 mmol) and methyltriphenylphosphine bromide (49 g, 137 mmol) in diethyl ether (300 mL) was stirred at room temperature for 1 hour, followed by the addition of 1,1 -dimethylethyl 3-oxoazetidine-1-carboxylate (10 g, 58 mmol in 100 mL diethyl ether). The mixture was stirred at 35 °C for 2 hours and then allowed to cool to room temperature. The mixture was filtered through a pad of celite, washing with diethyl ether. The filtrate was partitioned with water and washed twice with water, brine, dried over sodium sulfate, filtered and concentrated in vacuo to give an orange oil which was purified by column chromatography. Eluting with 10% ethyl acetate in hexanes, isolated product was concentrated in vacuo to afford 9.80 g, 58 mmol (100%) of 1,1-dimethylethyl 3-methylideneazetidine-l -carboxylate as a colorless oil. *H NMR (400 MHz, DMSO): 5.05-4.85 (m, 2H), 4.95-4.63 (m, 4H), 1.45 (s, 9H). GC-MS for C9H,5N02: 169.
[00331] To a solution of 1,1-dimethylethyl 3-methylideneazetidine-1 -carboxylate (2.96 g, 17.5 mmol) in chloroform (180 mL) was added 3-chloroperoxybenzoic acid (77%, 13.9 g, 62.0 mmol), and the resulting mixture was stirred at room temperature for 2 days. The reaction mixture was quenched with a 1:1 mixture (150 mL) of 10% sodium thiosulfate and saturated sodium bicarbonate solutions. The organic portion was isolated, dried over sodium sulfate, filtered and concentrated to give an oily residue which was then purified by flash chromatography (15-50% ethyl acetate-hexanes) to give 1,1-dimethylethyl l-oxa-5-azaspiro[2.3]hexane~5-carboxvlate (1.65g, 51%), GC-MS forC9Hi5N03: 185.
[00332] 1,1-Dimethylethyl l-oxa-5-azaspiro[2.3]hexane-5-carboxylate (51 mg, 0.28 mmol) was taken into THF (1 mL) followed by addition of morpholine (123 pL, 1.4 mmol) and the mixture was stirred for one hour at room temperature. The solution was then concentrated and the residue partitioned with ethyl acetate and water. The organic layer was washed once with water then brine and the organic layer dried over anhydrous sodium sulfate. Filtration and concentration gave a colorless oil that was purified by silica gel flash chromatography using ethyl acetate to 10% methanol in dichloromethane as eluents. The combined pure fractions were concentrated and the residue treated with neat TFA (1 mL) for 5 minutes then concentrated. The residue was taken into methanol (2 mL) and basified to pH > 10 by addition of Biorad AG-IX hydroxide form resin. Filtration and concentration afforded 3-(morpholin-4-ylmethyl)azetidin~3~ol (11.6 mg, 24% yield) as a colorless oil. 'H NMR (400 MHz, CD3OD): 3.69-3.66 (m, 4H), 3.55 (d, 2H), 3.49 (d, 2H), 2.66 (s, 2H), 2.57-2.55 (m, 4H).
[00333] 3-(Morpholin-4-ylmethyl)azetidin-3-ol (11.6 mg, 0.07 mmol) was taken into DMF (1 mL) followed by addition of DIPEA (35 pL, 0.21 mmol) and 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoyl fluoride (28 mg, 0.07 mmol), prepared using procedures similar to those described in Reference 1, and the mixture was stirred for 30 minutes at room temperature. The solution was then concentrated in vacuo and the residue purified by preparative reverse phase HPLC. Lyophillization of the combined fractions gave l-({3,4-difluoro-2-[(2-f!uoro-4-iodophenyl)amino]phenyI} carbonyl)-3-(morpholin-4-ylmethyl)azeti din-3 -ol trifluoroaceiate salt (6.3 mg) as a colorless amorphous solid. ]H NMR (400 MHz, CD3OD): 7.48 (d, 1H), 7.36 (d, 1H), 7.33-7.29 (m, 1H), 7.08-7.02 (m, 1H), 6.65-6.60 (m, 1H), 4.39 (br d, 1H), 4.24-4.18 (br, 2H), 4.08-3.96 (br m, 3H), 3.80 (br s, 2H), 3.51 (d, 2H), 3.40 (br s, 2H), 3.24 (br s, 2H).
[00334] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds were prepared. EXAMPLE 3(a). 1-((3,4-dif1uoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-(pyrrolidin-1 -ylmethyl)azetidin-3-ol: MS (El) for C21H21F3IN3O2: 532 (MH+). EXAMPLE 3(b). 1- {[ 1 -((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbonyl)-3 -hydroxy azeti din-3 -yl] methyl} piperidin-4-ol: MS (El) for C22H23F3IN3O3: 562 (MH4). EXAMPLE 3(c). 3-{[bis(2-hydroxyethyi)amino]methyl}-1-((3,4-difluoro-2-[(2- fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: MS (El) for C2iH23F3lN30.1: 566 (MH4). EXAMPLE 3(d). I-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbonyl)-3 - [(4-methylpiperazin-1 -y !)methyl]azetidin-3-ol: MS (El) for C22H24F3IN4O2: 561 (MH4). EXAMPLE 3(e). 1 -((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbonyl)-3 - [(4-methyl-1,4-diazepan-l -yl)methyl]azetidin~ 3-ol: MS (El) for €23Η2δΡ3ΪΝ4θ2: 575 (ΜΗ4). EXAMPLE 3(f). 1 -((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-{imethyl(l-methylpyrrolidin-3-y l)amino] methyl} azeti din-3 -ol: MS (El) for C23H26F3IN4O2: 575 (MH4). EXAMPLE 3(g). 3-(1,4’-bipiperidin-1 '-ylmethyl)-1 -((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: MS (El) for C27H32F3IN3O2: 629 (MH4). EXAMPLE 3(h). 3-({4-[2-(diethylamino)ethyl]piperazin-1 -yl}methyl)-1 -((3,4-difluoro-2~[(2-iluorO“4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol·. MS (El) for C27H35F3IN3O2: 647 (ΜΉ+). EXAMPLE 3(i). 1-((3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino]pheny 1} carbonyl)- 3-(((2-hydroxyethylXmethyl)amino]methyl}azetidin-3-ol: MS (El) for C20H21F3IN3O3: 536 (MH+). EXAMPLE 30). 3-(azetidin~ 1 -ylmethyl)-1 -((3,4-difluoro-2- [(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: MS (El) for C2oHi9F3lN3G2: 518 (MH+). EXAMPLE 3(k). 1-((3,4-difluoro~2-[(2-fluoro-4- iodophenyl)amino]phenyl)carbonyl)-3-{[(l-methylethyl)amino]methyl}azetidin-3-ol: MS (El) for C2oH2iF3lN302: 520 (MH+). EXAMPLE 3(m). 3-(aminomethyl)-l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: MS (El) for C^HisFsINsQi: 478 (m:h+). EXAMPLE 3(a), A-{[l-({3,4~difluoro-2-[(2-fluoro-4-
iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]methyl}acetamide: MS (El) for C19H17F3IN3O3: 520 (MH+). EXAMPLE 3(o). 1-((3,4-difluoro-2- [(2-fluoro-4~ iodopheny l)amino] pheny 1} carbonyi)-3 - {[(1,1 -dimethylethyl)amino]methyl} azetidin- 3-ol: MS (El) for C2jH23F3lN304: 534 (MH+). EXAMPLE 3(q). l-({3,4-diftuoro~2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(hydroxyamino)methyi]azetidin-3-ol: NMR (400 MHz, d,-MeOH): 7.45 (2d, 1H), 7.35 (m, 1H), 7.28 (m, 1H), 7.03 (m, 1H), 6.63 (m, 1H), 4.32 (d, 1H), 4.05 (dd, 2H), 3.85 (d, 1H), 3.00 (s, 2H); MS (El) for C37H15F3IN3O3: 494 (MH4). EXAMPLE 3(r). 1-((3,4-difluoro-2-[(2-fluoro-4- iadophenyl)amino]pheny !}carbony 1)-3- {[(methyloxy)amino]methyl} azetidin-3-ol:1H NMR (400 MHz, dj-MeOH): 7.45 (2d, 1H), 7.35 (m, 1H), 7.27 (m, 1H), 7.04 (m, 1H), 6.62 (m, 1H), 4.26 (d, 1H), 4.08 (d, 1H), 4.00 (d, 1H), 3.84 (d, 1H), 3.30 (s, 3H), 3.00 (d, 2H); MS (El) for CS8H17F3IN3O3: 508 (MH4). EXAMPLE 3(s). 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbonyl)-3 - {[(ethyloxy)aminojmethyl} azetidin-3 -ol:! H NMR (400 MHz, dt-MeOH): 7.45 (2d, 1H), 7.34 (m, 1H), 7.26 (m, 1H), 7.03 (m, 1H), 6.63 (m, 1H), 4.26 (d, 1H), 4.12 (d, 1H), 4.00 (d, 1H), 3.84 (d, 1H), 3.61 (dd, 2H), 3.00 (s, 2H), 1.06 (t, 3H); MS (El) for Cl9Hi9F3lN303: 522 (MH4). EXAMPLE 3(f). 1-((1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]methyl}guanidine acetate salt: 3H NMR (400 MHz, (LrMeOH): 7.46 (2d, 1H), 7.36 (m, 1H), 7.30 (m, 1H), 7.04 (m, 1H), 6.62 (m, 1H), 4.18 (d, 1H), 4.08 (d, 1H), 4.02 (d, 1H), 3.88 (1//), 3.40 (s, 2H); MS (El) for CfsH^INjC^: 520 (MH*). EXAMPLE 3(u). A?-{[l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3- yl]methyl}benzenecarboximidamide hydrochloride: *H NMR (400 MHz, cU-MeOH): 7.70 (d, 3H), 7.58 (m, 2H), 7.46 (dd, 1H), 7.36 (m, 1H), 7.31 (m, 1H), 7.04 (m, 1H), 6.62 (m, 1H), 4.28 (m, 1H), 4.15 (m, 2H), 3.96 (m, 1H), 3.78 (s, 2H); MS (El) for C24H20F3IN4O2: 581 (MH+). EXAMPLE 3(v). l-({3,4-difluoro-2-[(2~fluoro-4~ iodophenyl)amino]phenyl}carbonyl)-3-[(pyrimidin-2-ylaxnino)methyl]azetidin-3-ol hydrochloride: 5H NMR (400 MHz, d4-MeOH): 8.48 (s, 2H), 7.46 (2d, 1H), 7.36 (m, 1H), 7.28 (m, IH), 7.04 (m, 1H), 6.85 (t, 1H), 6.61 (m, 1H), 4.24 (d, IH), 4.06 (t, 2H), 3.87 (d, 1H), 3.75 (d, 2H); MS (El) for C2iHi7F3IN5G2: 556 (ΜΗ4). EXAMPLE 3(w). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(pyri din-2-vlamino)methyl]azetidin-3-ol hydrochloride: *11 NMR (400 MHz, d4-MeOH): 7.87 (dd, IH), 7.85 (dd, IH), 7.46 (2d, IH), 7.36 (m, 2H), 7.06 (m, 2H), 6.89 (m, IH), 6.61 (m, IH), 4.53 (d, 2H), 4.46 (m, IH), 4.28 (m, IH), 4.16 (m, IH), 3.96 (m, IH); MS (El) for C22H18F3IN4O?: 555 (MH+). EXAMPLE 3{x). l-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)araino]pheny 1}carbonyl)-3-[(ethylamino)methyS]azetidin-3-ol: !H NMR (400 MHz, d6-DMSO): 8.61 (s, 2H), 7.59 (d, IH), 7.40 (d, IH), 7.36-7.33 (m, IH), 7.23-7.18 (m, IH), 6.71 (s, 2H), 4.31-4.26 (m, IH), 4.13-4.05 (m, 2H), 3.88-3.84 (m, IH), 3.21 (br m, 2H), 2.97-2.90 (m, 2H), 1.19 (t, 3H). MS (El) for C19H19F3IN3O2: 506 (MH+). EXAMPLE 3(y). 3-[(cyclopropylamino)methyl]-l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]pheny 1}carbonyl)azetidin-3-ol: !H NMR (400 MHz, d^-DMSO): 8.99 (br s, 2H), 8.60 (s, IH), 7.58 (d, IH), 7.39 (d, IH), 7.36-7.33 (m, IH), 7.23-7.16 (m, IH), 6.72 (s, 2H), 4.34-4.29 (m, IH), 4.14-4.04 (m, 2H), 3.88-3.84 (m, IH), 2.70- 2.64 (m, IH), 0.89 (br s, 2H), 0.74-0.69 (br s, 2H). MS (El) for C20H19F3IN3O2: 518 (ΜΙ-Γ). EXAMPLE 3(z). 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-{[(2,2,2-trifluoroethyl)amino]methyl}azetidin-3-ol: !H NMR (400 MHz, d6-DMSO): 8.60 (s, 1H), 7.58 (d, 1H), 7.38 (d, 1H), 7.35- 7.30 (m, 1H), 7.22-7.17 (m, 1H), 6.72-6.67 (m, IH), 4.25-4.19 (m, 1H), 4.07-3.98 (m, 2H), 3.86-3.77 (m, 2H), 3.19-3.09 (m, 2H). MS (El) for CioHieFeH^Cfc: 560 (MB'*). EXAMPLE 3(aa). 1 -({3,4-difluoro-2-[(2-fluoro-4- i odopheny l)aniino] pheny S} carbonyl)-3-( 1 Η-1,2,3-triazol-1 -ylmethyl)azetidin-3-ol: *H NMR (400 MHz, d6-DMSO): 8.55 (s, 1H), 8.04 (s, 1H), 7.66 (s, IH), 7.58 (d, IH), 7.39 (d, 1H), 7.34-7.29 (m, 1H), 7.22-7.15 (m, 1H), 6.72-6.66 (m, 1H), 6.29 (s, 1H), 4.64 (s, 2H), 4.29-4.25 (m, 1H), 4.13-4.09 (m, 1H), 4.00-3.96 (m, 1H), 3.77-3.73 (m, 1H), 3.16 (d, 1H). MS (El) for C19Hi5F3iN5Q2: 530 (MH+). EXAMPLE 3(bb). l-({3,4-difluoro-2-[(2-fluoro~4-iodophenyl)amino]pheny 1} carbony 1)-3 - {[(2,2- dimethy!propyl)ammo]methyl}azetidin-3-ol: lU NMR (400 MHz, d^-DMSO): 8.61 (s, 1H), 8.30 (s, 2H), 7.59 (d, 1H), 7.39 (d, 1H), 7.36-7.17 (m, 4H), 6.77-6.66 (m, 4H), 4.35-4.30 (m, 1H), 4.16-4.08 (m, 2H), 3.92-3.87 (m, 1H), 3.31-3.27 (m, 2H), 2.78- 2.74 (m, 2H), 1.76 (s, 4H), 0.99 (s, 9H). MS (El) for C22H25F3IN3O2: 548 (MH+). EXAMPLE 3(cc). !-({3,4-difluoro-2-[(2-fluoro-4-iodopheny i)aiTiino]pheny 1} carbony 1)-3-( {[2-(4- methylphenyl)ethyl]amino}methyl)azetidin-3-ol acetate salt: *H NMR (400 MHz, CDCI3): 8.48 (s, 1H), 7.39 (dd, 1H), 7.31-7.34 (m, III), 7.08 (dd, 5H), 6.77-6.83 (m, 1H), 6.58-6.63 (m, 1H), 4.20 (br s, 1H), 4.01 (d, 1H), 2.87 (t, 4H), 2.75 (t, 4H), 2.5 (br s, 2H), 2.33 (s, 3H), 2.08 (s, 2H). MS (El) for C26H25F3IN3O2: 594 (M-H). EXAMPLE 3(dd). 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]pheny S}carbonyl)-3-[(2,3-dihydro-l/-/-inden-2-ylamino)methyl]azetidin-3-ol acetate salt: *H NMR (400 MHz, CDCI3): 8.48 (s, 1H), 7.40 (dd, 1H), 7.32-7.34 (m, 1H), 7.15-7.22 (m,4H), 7.10-7.14 (m, 1H), 6.77-6.83 (m, 1H), 6.58-6.64 (m, 1H), 4.22 (br s, 1H), 4.04 (d, 1H), 3.57-3.63 (m, 1H), 3.17 (dd, 2H), 2.94 (s, 2H), 2.75 (dd, 2H), 2.48 (br s, 4H), 2.08 (s, 2H). MS (El) for C26H23F3IN3O2: 592 (M-H). EXAMPLE 3(ee). 1 -((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbony 1)-3 -(([(1 S,2S)-2- hydroxycyclopentyl]amino}methyl)azetidin-3-ol acetate salt: !H NMR (400 MHz, CD3OD): 7.46 (dd, 1H), 7.33-7.37 (m, 1H), 7.26-7.31 (m, 1H), 7.00-7.08 (m, IH), 6.58-6.65 (m, IH), 4.2 (t, 1H), 3.86-4.06 (m, 4H), 2.92-3.10 (m, 3H), 2.00-2.10 (m, 1H), 1,91-1,97 (m. 3H), 1.66-1.78 (m, 2H), 1.52-1.61 (m, 1H), 1.32-1.44 (m, 1H). MS (El) for C22H23F3IN3O3: 560 (M-H). EXAMPLE 3(ff). l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)ami no]phenyl} carbonyl)-3-{[(1,2- dimethylpropyl)amino]methyl}azetidin-3-ol acetate salt: !H NMR (400 MHz, CD3OD): 7.45 (dd, 1H), 7.33-7.37 (m, 1H), 7.26-7.31 (m, 1H), 7.01-7.08 (m, 1H), 6.59- 6.64 (m, 1H), 4.14-4.22 (m, 1H), 3.98-4,06 (m, 2H), 3.84-3.90 (m, 1H), 2.86-3.20 (m, 2H), 2.65 (br s, 1H), 1.92 (s, 2H), 1.76-1.86 (m, 1H), 1.06 (d, 3H), 0.91 (dd, 6H). MS (El) for C22H25F3IN302: 546 (M-H). EXAMPLE 3(gg). 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]pheny 1} carbony 1)-3 -({[ 1 -methy 1-2- (methyloxy)ethyl]amino}methyl)azetidin-3~ol acetate salt: NMR (400 MHz, CD3OD): 7.55 (dd, 1H), 7.33-7.36 (m, 1H), 7.26-7.31 (m, 1H), 7.01-7.09 (m, 1H), 6.59- 6.65 (m, 1H), 4.14-4.22 (ra, 1H), 3.96-4.06 (m, 2H), 3.85-3.92 (m, 1H), 3.40- 3.48 (m, 1H), 3.34 (s, 3H), 2.90-3.15 (m, 3H), 1.94 (s, 3H), 1.11 (d, 3H). MS (El) for C21H23F3IN3O3: 548 (M-H). EXAMPLE 3(hh). 1 -({3,4-difluoro-2-[(2-fluoro-4- iodopheny 1 )am ino] phenyl} carbony 1)-3 - {[(1 -ethylpropyl )amino] methyl} azetidin-3 -ol acetate salt: !H NMR (400 MHz, CD3OD): 7.45 (dd, 1H), 7.33-7.36 (m, 1H), 7.26- 7.31 (m, 1H), 7.01-7.09 (m, 1H), 6.58-6.65 (m, 1H), 4.15-4.20 (m, 1H), 3.99-4.06 (m, 2H), 3.86-3.91 (m, 1H), 2.94 (s, 2H), 2.55-2.63 (m, 1H), 1.92 (s, 2H), 1.48-1.58 (m, 4H), 0.92 (t, 6H). MS (El) for C22H25F3IN3O2: 546 (M-H). EXAMPLE 3(u). l-({3,4-difluoro-2-[(2-fluoro-4-
iodopheny l)amino] phenyl} carbonyl)-3 -(1 //-imidazol-1 -ylmethyl)azetidin-3-ol: 1H NMR (400 MHz, CD3OD): 7.67 (br s, 1H), 7.48 (m, 1H), 7.36 (m, 1H), 6.91 (br s, 1H), 6.63 (m, 1H), 4.25 (s, 2H), 4.22 (m, 1H), 4.02 (m, 2H), 3.82 (m, 1H). MS (El) for C2oHs6F3rN4G2: 529 (MH+), EXAMPLE 3(jj). 3- {[(cyclopropylmethyl)amino]methyl )-1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: !H NMR (400 MHz, CD3OD): 7.47 (m, 1H), 7.36 (m, 1H), 7.31 (m, 1H), 7.05 (m, 1H), 6.62 (m, 1H), 4.30 (m, 1H), 4.24 (m, 2H), 3.99 (m, 1H), 3.66 (m, 2H), 2.91 (d, 2H), 1.08 (m, 1H), 0.71 (m, 2H), 0.40 (m, 2H). MS (El) for C21H21F3IN3O2: 532 (MH+), EXAMPLE 3(kk). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino] phenyl} carbonyl)-3 - {[(phenylmethyl)amino]methyl} azetidin-3-ol hydrochloride: !H NMR (400 MHz, CD3OD): 7.47 (m, 5H), 7.43 (m, 1H), 7.35 (m, 1H), 7.27 (m, 1H), 7.04 (m, 1H), 6.61 (m, 1H), 4.24 (m, 3H), 4.08 (m, 2H), 3.96 (m, 1H). MS (El) for Ca^iFsINsCb: 568 (MH+). EXAMPLE 3(mm). 3-[(butylamino)methyl]-1 -( { 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: !H NMR (400MHz, de-DMSO): 8.56 (s, 1H), 7.57 (dd, 1H), 7.36 (d, 1H), 7.31 (t, 1H), 7.17 (q, 1H), 6.67 (dt, 1H), 4.04 (d, 1H), 3.88 (q, 2H), 3.69 (d, 1H), 2.59 (s, 2H), 1.90 (s, 2H), 1.22-1.33 (m, 4H), 0.84 (t, 3H); MS (El) for €21^3^3()2: 534 (MH+). EXAMPLE 3(nn). 1-((3,4-difluoro-2-[(2-fiuoro-4- iodophenyl)amino]phenyl}carbonyl)-3-({ [(1 -ethylpyrrolidin-2- yl)methyl] amino} methyl)azeti din-3 -ο 1: !H NMR (400MHz, ds-DMSO): 8.59 (s, 1H), 7.57 (dd, 1H), 7.36 (d, 1H), 7.31 (t, 1H), 7.17 (q, 1H), 6.68 (dt, 1H), 4.02 (t, 1H), 3.89 (q, 2H), 3.69 (d, 1H), 2.98 (s, 1H), 2.67-2.76 (m, 1H), 2.62 (s, 1H), 2.39-2.45 (m, 1H), 2.29 (s, 1H), 1.97-2.13 (m, 2H), 1.69 (s, 1H), 1.54 (s, 3H), 0.97 (t, 3H); MS (El) for C24H28F3IN4O2: 589 (MH*). EXAMPLE 3(oo). 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)aniino]phenyl}carbonyl)-3-{[(2-hydroxyethyl)amino]methyl}azetidin-3- ol: !H NMR (400MHz, d6-DMSO): 8.57 (s, 1H), 7.57 (dd, 1H), 7.37 (d, 1H), 7.32 (t, 1H), 7.18 (q, 1H), 6.68 (dt, 1H), 4.06 (d, 1H), 3.87 (d, 2H), 3.70 (d, 1H), 3.42 (t, 2H), 2.65 (s, 2H), 2.56 (dt, 2H). 1.91 (s, 2H); MS (El) for CioH^INsCb: 522 (MH+). EXAMPLE 3(pp). 1-((3,4-difluoro-2-[(2-f!uoro-4-iodophenyl)amino]phenyl}carbonyl)-3-(([2- (dimethylamino)ethyl]amino}methyl)azetidm-3-ol: !H NMR (400MHz, d^-DMSO): 8.58 (s, 1H), 7.57 (dd, 1H), 7.36 (d, 1H), 7.31 (t, 1H), 7.17 (q, IK), 6.68 (dt, 1H), 4.02 (d, 1H), 3.87 (t, 2H), 3.70 (d, 1H), 2.62 (s, 1H), 2.54 (t, 1H), 2.23 (t, 1H), 2.09 (s, 4H), 7.85 (s, 6H); MS (El) for Cs^FsIN^Ch: 549 (MIT). EXAMPLE 3(qq). l-({3,4-difluoro-2-[(2-fluoro~4-iodopheny l)ami nojpheny 1} carbony 1)-3 -( {[2-(1 -methylpyrrolidin-2-yl)ethyl]amino}methyl)azetidin-3-ol: *H NMR (400MHz, d6-DMSO): 8.58 (s, 1H), 7.57 (dt, 1H), 7.36 (d, 1H), 7.31 (t, 1H), 7.17 (q, 1H), 6.68 (dt, 1H), 4.04 (d, !H), 3.89 (d, 2H), 3.79 (d, 1H), 2.88-2.92 (m, 1H), 2.61 (s, 2H), 2.15 (s, 3H), 1.93-2.04 (m, 2H), 1.75-1.83 (m, 3H), 1.54-1.70 (m, 3H), 1.20-1.37 (m, 2H); MS (El) for C24H28F3iN402: 589 (MH+). EXAMPLE 3(rr). 1 -((S^-difluoro^-iP-fluoro^-iodophenyOaminojphenylJcarbonyl^S-fKtetrahydrofuran^-ylmethyl)amino]methyl}azetidin-3-ol: Ή NMR (400MHz5 d6-DMSO): 8.58 (s, 1H), 7.57 (dd, 1H), 7.37 (d, 1H), 7.31 (t, IH), 7.14 (q, 1H), 6.68 (dt, 1H), 5.75 (s, 1H), 4.03 (t, 1H), 3.87 (t, 2H), 3.76 (q5 1H), 3.68 (q, 2H), 3.54-3.58 (m, 1H), 2.63 (s, 2H), 1.91 (s, 2H), 1.71-1.87 (m, 3H), 1.40-1.48 (m, 1H); MS (El) for C22H23F3IN3O3: 562 (MH+). EXAMPLE 3(ss). 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbonyl)-3 - {[(3 -pyrrolidin-1 - ylpropyl)amino]methy 1} azetidin-3-ol: !H NMR (400MHz, d6-DMSO): 8.58 (s, 1H), 7.57 (dd, 1H), 7.36 (d, 1H), 7.31 (t, 1H), 7.17 (q, 1H), 6.68 (dt, !H), 4.04 (d, 1H), 3.89 (d, 2H), 3.69 (d, 1H), 2.60 (s, 1H), 2.34-2.37 (m, 4H), 1.86 (s, 8H), 1.64 (s, 2H), 1.46-1.53 (m, 1H); MS (El) for C24H28F3IN4O2: 589 (MH4). EXAMPLE 3(tt). 1 -((3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino]pheny 1} carbony 1)-3 -( {[2- (methyloxy)ethyl]amino}methyl)azetidin-3-ol: !H NMR (4Q0MHz, ds-DMSO): *H NMR (400MHz, cU-DMSO): 8.57 (s, 1H), 7.57 (dd, 1H), 7.37 (d, 1H), 7.31 (t, 1H), 7.17 (q, 1H), 6.68 (dt, 1H), 4.03 (d, 1H), 3.86 (d, 2H), 3.70 (d, 1H), 3.21 (s, 3H), 2.63 (s, 4H), 1.88 (s, 2H); MS (El) for CaoHstFsINaCb: 536 (MH+). EXAMPLE 3(uu). 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino] phenyl} carbony 1)-3 -(([(1 -methylpiperidin-4- yl)methy 1]amino}methyl)azetidin-3-o!: 'H NMR (400MHz, dg-DMSO): 8.58 (s, 1H), 7.57 (d, 1H), 7.37 (d, 1H), 7.31 (t, 1H), 7.17 (q, 1H), 6.68 (t, 1H), 4.03 (d, 1H), 3.89 (t, 2H), 3.69 (d, 1H), 2.68 (d, 2H), 2.57 (s, 1H), 2.34 (d, 2H), 1.88 (s, 4H), 1.73 (t, 2H), 1.57 (d, 2H), 1.23 (s, 1H), 1.05 (q, 2H); MS (El) for C24H28F3IN4O3: 589 (MH*). EXAMPLE 3(w). 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]pheny 1} carbonyl)-3-(([4- (dimethylamino)butyl]amino}methyl)azetidin-3-ol: 'HNMR (400MHz, d^-OMSO): 7.57 (dd, 1H), 7.36 (d, 1H), 7.31 (t, 1H), 7.18 (q, 1H), 6.68 (dt, 1H), 4.03 (t, 2H), 3.88 (t, 2H), 3.70 (d, 1H), 3.08 (s, 1H), 2.60 (s, 1H), 2.44-2.47 (m, 2H), 2,28-2.33 (m, 1H), 2.07-2.16 (m, 6H), 1.29-1.35 (m, 4H); MS (El) for C23H28F3IN4O2: 577 (Mtf). EXAMPLE 3(ww). 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenvl)amino]phenyl} carbonyi)-3 - ([(2-fur an-2-ylethyl)amino]methyl}azetidin-3-ol: 5H NMR (400MHz, d6-DMSO): 8.58 (s, 1H), 7.57 (d, 1H), 7.49 (s, 1H), 7.36 (d, 1H), 7.31 (t, 1H), 7.17 (q, 1H), 6.68 (t, 1H), 6.33 (s, 1H), 6.08 (s, 1H), 5.72 (s, 1H), 4.04 (d, 1H), 3.87 (d, 2H), 3.70 (d51H), 2.74 (d, 2H), 2.69 (d, 2H), 2.64 (s, 2H); MS (El) for C23H2JF3EN3O3: 572 (MH4). EXAMPLE 3(xx). 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenv l)amino] pheny 1} carbonyl)-3 - {[(2-ethylbutyl)amino]methy 1} azetidin-3 -ol: lHNMR (400MHz, d6-DMSO): 8.58 (s, 1H), 7.56 (dd, 1H), 7.36 (d, 1H), 7.31 (t, 1H), 7.17 (q, 1H), 6.67 (dt, 1H), 4.03 (d, 1H), 3.90 (d, 2H), 3.69 (d, 1H), 2.58 (s, 2H), 2.37 (d, 2H). 1.17-1.27 (m, 5H), 0.78 (t, 6H); MS (El) for C23H27F3IN3O2: 562 (MH4). EXAMPLE 3(yy). 1,1-dimethylethyl [3-(([1-((3,-4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3- yl]methyl}amino)propyI]carbamate: *H NMR (400MHz, de-DMSO): 8.58 (s, 1H), 7.57 (d, 1H), 7.30-7.38 (m, 3H), 7.17 (q, 1H), 6.82 (t, 1H), 6.68 (dt, 1H), 4.07 (d, 1H), 3.89 (d, 2H), 3.70 (d, 1H), 3.36 (s, 2H), 2.93 (q, 2H), 2.61 (s, 2H), 1.46 (t, 2H), 1.36 (s, 9H); MS (El) for C25H30F3IN4O4: 635 (ΜΗ*). EXAMPLE 3(zz). 1-((3,4-difluoro-2-[(2-fluoro-4-iodopheny i)amino]pheny 1} carbony 1)-3 - ([(pyrrolidin-2- ylmethyl)amino]methyl}azetidin-3-ol: 'H NMR (400MHz, ds-DMSO): 8.53 (s, 1H), 7.58 (dd, 1H), 7.37 (d, 1H), 7.33 (d, 1H), 7.18 (q, 1H), 6.67 (dt, 1H), 6.25 (s, 1H), 4.07 (d, 1H), 3.96 (q, 2H), 3.78 (s, 3H), 3.34 (s, 6H), 1.73 (s, 1H), 1.35-1.39 (m, 1H); MS (El) for C22H24F3TN4O2: 561 (MH4). EXAMPLE 3(aaa). 1,1-dimethylethyl 4-[( ([ 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]pheny 1} carbony 1)-3-hydroxyazetidin-3 - yl]methyl}amino)methyl]piperidine-l-carboxylate: !H NMR (400MHz, de-DMSO): 8.56 (s, 1H), 7.56 (dd, 1H), 7.36 (d, 1H), 7.30 (t, 1H), 7.17 (q, 1H), 6.68 (dt, 1H), 4.03 (d, 1H), 3.88 (t, 4H), 3.69 (d, 1H), 2.58 (s, 2H), 2.35 (d, 2H), 1.60 (d, 2H), 1.47 (s, 1H), 1.39 (s, 10H), 0.90 (q, 2H): MS (El) for C28H34F3IN4O4: 675 (MH4). EXAMPLE 3(bbb). l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyr)-3-(([(2- hy droxyphenyl)methyl] amino} methyl)azei ί din-3 -ο 1: !H NMR (400MHz, dg-DMSO): 8.56 (s, 1H), 7.54 (dd, 1H), 7.35 (d, 1H), 7.30 (t, 1H), 7.17 (q, 1H), 7.05 (t, 2H), 6.64-6.72 (m, 3H), 4.07 (d, 1H), 3.90 (t, 2H), 3.78 (s, 2H), 3.72 (d, 1H), 2.65 (s, 2H); MS (El) for C24H21F3IN3O3: 584 (MH4). EXAMPLE 3(ccc). l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-({[(3- hydroxyphenyl)methyl]amino}methyl)azetidin-3-ol: 'H NMR (400MHz, de-DMSO): 8.58 (s, 1H), 7.56 (d, 1H), 7.35 (d, 1H), 7.29 (t, 1H), 7.16 (q, 1H), 7.06 (t, 1H), 6.64-6.72 (m, 3H), 6.60 (dd5 1H), 4.07 (d, 1H), 3.88 (t, 2H), 3.69 (d5 1H), 3.60 (s, 2H), 2.58 (d, 2H); MS (El) for C24H21F3IN3O3: 584 (MH+). EXAMPLE 3(ddd). 1-((3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino]phenyl} carbony 1)-3 ~({[(4- hydroxyphenyl)methyl]amino}methyl)azetidin-3-ol: ’H NMR (400MHz, dg-OMSO): 8.57 (s, 1H), 7.55 (dd, 1H), 7.35 (d, 1H), 7.27 (t, 1H), 7.16 (q, 1H), 7.06 (d, 2H), 6.64-6.70 (m, 3H), 4.04 (d, 1H)S 3.85 (ί, 2H), 3.68 (d, 1H), 3.55 (s, 2H), 2.56 (d, 2H); MS (El) for C24H21F3IN3O3: 584 (MH^. EXAMPLE 3(eee). 3-({[l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]methyl}amino)-5-(hydroxymethyl)cyclopentane-1,2-diol: *H NMR. (400MHz, de-DMSO): 8.60 (broad s, 1H), 7.57 (dd, 1H), 7.37 (d, 1H), 7.32 (t, 1H), 7.16 (q, 1H), 6.68 (ί, 1H), 4.06 (q, 2H), 3.86 (t, 3H), 3.72 (dd, 1H), 3.60 (t, 1H), 3.36-3.43 (m, 2H), 3.30 (dd, 1H), 2.80 (q, 1H), 2.62-2.72 (m, 2H), 1.88-1.95 (m, 1H), 0.82-0.90 (m, 1H); MS (El) for C23H25F3IN3O5: 608 (MH^. EXAMPLE 3(fff). l-({3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino]pheny 1} carbonyl)-3 - {[(piperidin-4- ylmethy!)amino]methyl}azetidin-3-ol: 5H NMR (400MHz, d$-DMSO): 8,59 (broad s, 1H), 7.57 (dd, 1H), 7.37 (d, 1H), 7.30 (t, 1H), 7.17 (q, 1H), 6.68 (dt, 1H), 4.03 (d, 1H), 3.87 (d, 2H), 3.69 (d, 1H), 3.01 (d, 2H), 2.59 (s, 2H), 2.43-2.56 (m, 1H), 2.35 (d, 2H), 1.65 (d, 2FI), 1.47 (s, 1H), 1.07 (q, 2H); MS (El) for C23H26F3IN4O2: 575 (MET). EXAMPLE 3(ggg). 3-{[(3-aminopropyl)amino]methyl)-1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: !H NMR (400MHz, dg-DMSO): 7.57 (dd, 1H), 7.37 (d, 1H), 7.31 (t, 1H), 7.17 (q, 1H)„ 6.68 (dt, 1H), 4.05 (d, 1H), 3.88 (d, 2H), 3.69 (d, 1H), 2.61 (t, 3H), 2.53-2.56 (m, 1H), 1.49 (t, 1.49); MS (El) for C23H26F3IN4O2: 535 (MH+). EXAMPLE 3(hhh). 1-(( 3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino]pheny 1} carbony 1)-3 - [( ([2-(4-methy lpiperazin-1 -yl)phenyl]methyl}amino)meihy!]azetidin-3-ol; !HNMR (400MHz, dg-DMSO): 8.59 (broad s, 1H), 7.55 (dd, 1H), 7.34 (t, 2H), 7.28 (d, 1H), 7.13-7.20 (m, !H), 7.05 (d, 1H), 6.99 (t, 1H), 6.66 (dt, 1H), 4.03 (d, 1H), 3.90 (t, 2H), 3.71 (d, 3H), 2.83 (s, 5H), 2.60 (s, 2H), 2.42 (s, 3H), 2.20 (s, 3H); MS (El) for C29H31F3IN5O2: 666 (MH+). EXAMPLE 3(iii). 3~[(1 //-benzimidazol-2-ylamino)methy I] -1 -({3,4-difluoro-2- [(2-fluoro-4-iodophenyl)amino]phenyI}carbonyl)azeiidin-3-ol: *H NMR (400MHz, CDC13): 8.04 (s, 2H), 7.28-7.35 (m, 2H), 7.23-7.26 (m, 2H), 7.09-7.12 (m, 2H), 6.80 (q, 1H), 6.57-6.63 (m, 1H), 5.28 (broad s, 2H), 4.38 (s, 3H), 4.25 (s, 1H), 4.21 (d, 2H); MS (El) for C24H19F3IN5O2: 594 (MH+). EXAMPLE 3(Jjj). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(l/i-imidazol-2-ylamino)methyl]azetidin-3-ol: !H NMR (400MHz, d6-DMSO): 12.12 (s, 1H), 8.68 (s, 1H), 7.57-7.61 (m, 3H), 7.36-7.41 (m, 2H), 7.19 (q, 1H), 6.99 (s, 1H), 6.91 (s, 1H), 6.71 (dt, 1H), 6.45 (s, 1H), 4.28 (d, 1H), 4.06 (d, 1H), 4.03 (d, 1H), 3.82 (d, 2H); MS (El) for C24H17F3IN5O2: 544 (MH+). EXAMPLE 3(kkk). l-({3,4-difluoro~2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-{2-[(2,2,3,3,3- pentafluoropropyl)amino]ethyl}azetidin-3-ol: *H NMR (400 MHz, dg-DMSO): 8.58 (br s, 1H), 7.56 (dd, 1H), 7.37 (dd, 1H), 7.34-7.28 (m, 1H), 7.22-7.13 (m, 1H), 6.68 (ddd, 1H), 5.82 (br s, 1H), 4.06 (d, 1H), 3.91 (t, 2H), 3.70 (d, 1H), 3.40-3.25 (m, 2H), 2.76 (d, 2H), 2.40-2.31 (m, 1H); MS (El) for CaoH^FsINaOa: 610 (MH+), EXAMPLE 3(mmm). l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbony 1)-3 - {2- [(3,3,3 - trifluoropropyl)amino]ethyl}azetidin-3-ol: 5H NMR (400 MHz, de-DMSO): 8.58 (br s, 1H), 7.57 (dd, 1H), 7.37 (dd, 1H), 7.34-7.28 (m, 1H), 7.22-7.13 (m, 1H), 6.68 (ddd, 1H), 5.76 (br s, 1H), 4.05 (d, 1H), 3.88 (d, 2H), 3.70 (d, 1H), 2.71 (t, 2H), 2.63 (s, 2H), 2.41-2.26 (m, 2H); MS (El) for QwHieFelNaCfe: 574 (MH+). EXAMPLE 3(nnn). l-({3,4-difluoro~2-[(2-fluoro~4-iodophenyi)amino] phenyl} carbony i)-3 -[(2,3-dihvdro -1 //-inden-1 -y!amino)rnethyl]azetidm~3~ol acetate salt: !H NMR (400 MHz, DMSO): 8.61-8.56 (m, 1H), 7.55 (d, 1H), 7.37-7.07 (m, 8H), 6.71-6.64 (m, 1H), 4.16-4.05 (m, 2H), 3.98-3.85 (m, 2H), 3.72-3.68 (m, 1H), 2.90-2.82 (m, 1H), 2.74-2.64 (m, 2H), 1.91 (s, 3H), 1.73-1.63 (m, 1H); MS (El) for C26H23F3IN3O2: 594 (MFT). EXAMPLE 3(ooo). 3-[(cyclooctylamino)methyl]-l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt: *H NMR (400 MHz, DMSO): 8.56 (s, 1H), 7.55 (d, 1H), 7.20-7.14 (m, 2H), 6.70-6.66 (m, 1H), 4.03-3.98 (m, 1H), 3.92-3.86 (m, 2H), 3.72-3.67 (m, 1H), 2.60 (s, 2H), 1.90 (s, 3H), 1.64-1.22 (m, 15H); MS (El) for C25H29F3IN3O2: 588 (MH+). EXAMPLE 3(ppp). 3 - [(eye lohepty larnino)methy 1]-1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt: *H NMR (400 MHz, DMSO): 8.55 (s, 1H), 7.55 (d, 1H), 7.36-7.28 (m, 2H), 7.21-7.14 (m, 1H), 6.70-6.66 (m, 1H), 4.04-4.00 (m, 1H), 3.92-3.85 (m, 2H), 3.71-3.66 (m, 1H), 2.60 (s, 2H), 1.90 (s, 3H), 1.70-1.13 (m, 13H); MS (El) for C^FsINaOa: 574 (MH+). EXAMPLE 3(qqq). 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-{[(2-pyridin-3-ylethyi)amino]methyl}azetidin~ 3-ol acetate salt: !H NMR (400 MHz, DMSO): 8.58 (s, 1H), 8.42-8.37 (m, 2H), 7.62- 7.54 (m, 2H), 7.38-7.27 (m, 3H), 7.21-7.14 (m, 1H), 6.71-6.66 (m, 1H), 4.06-4.02 (m, 1H), 3.90-3.86 (m, 2H), 3.72-3.68 (m, 1H), 2.80-2.64 (m, 6H), 1.90 (s, 3H); MS (El) for C24H22F3IN4O2: 583 (MH+). EXAMPLE 3(rrr). iV-cyclohexyl-N2- {[1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyi)-3~hydroxyazetidin-3-yl]methyi}-2-methylalaninamide acetate salt: rH NMR (400 MHz, DMSO): 8.66 (br s 1H), 8.55 (s, 1H), 7.93-7.90 (m, 1H), 7.58 (d, 1H), 7.40-7.31 (m, 2H), 7.24-7.17 (m, 1H), 6.71-6.66 (m, 1H), 6.60 (br s, 1H), 4.28-4.23 (m, 1H), 4.14-4.02 (m, 2H), 3.89-3.83 (m, 1H), 3.12 (br s, 2H), 1.90 (s, 3H), 1.74-1.42 (m, 11H), 1.31-1.02 (m, 6H); MS (El) for C27H32F3IN4O3: 645 (MH+). EXAMPLE 3(sss). 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-([(tetrahydro-2H-pyran-4-ylmethyl)amino]methy 1}azetidin-3-ol acetate salt: *H NMR (400 MHz, DMSO): 8.56 (s, 1H), 7.56 (d, 1H), 7.38-7.27 (m, 2H), 7.20-7.14 (m, 1H), 6.71-6.66 (m, 1H), 4.05- 4.01 (m, 1H), 3.91-3.78 (m, 4H), 3.71-3.67 (m, 1H), 3.25-3.18 (m, 2H), 2.60 (s, 2H), 2.36 (d, 2H), 1.90 (s, 3H), 1.57-1.50 (m, 3H), 1.13-1.02 (m, 2H); MS (El) for C23H25F3IN3O3: 576 (MH+). EXAMPLE 3(ttt). 1 -((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)-3-(([2-(dimethylamino)-1 -methylethyl]amino}methyl)azetidin-3-ol trifluoroacetate salt: NMR (400 MHz, DMSO): 8.59-8.54 (m, 1H), 7.56 (d, 1H), 7.38-7.28 (m, 2H), 7.21-7.13 (m, 1H), 6.71- 6.63 (m, 1H), 4.04-3.95 (m, 1H), 3.88-3.78 (m, 2H), 3.73-3.68 (m, 1H), 2.70-2.50 (m, 3H), 2.08 (s, 6H), 1.88 (s, 2H), 0.85-0.82 (m, 3H); MS (El) for C22H26F3IN4O2: 563 (MH^. EXAMPLE 3(uuu). A-cyclopropyl-1 -({[ 1 -((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbonyi)-3 -hydroxyazetidin-3- yljmethyl} amino)cyclopentanecarboxamide trifluoroacetate salt: *H NMR (400 MHz, DMSO): 8.80 (br s, 1H), 8.58 (s, 1H), 8.04 (s, 1H), 7.59 (d, 1H), 7.40-7.31 (m, 2H), 7.25-7.16 (m, 1H), 6.74-6.58 (m, 2H), 4.26-3.82 (m, 4H), 3.10 (br s, 2H), 2.69-2.64 (m, 1H), 2.11-1.88 (m, 4H), 1.82-1.61 (m, 4H), 0.67-0.62 (m, 2H), 0.52-0.48 (m, 2H); MS (El) for C26H28F3rN403: 629 (MH*). EXAMPLE 3(vw). N2-{[l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]methyl}-iV-ethyl-2-methylalaninamide acetate salt: 'H NMR (400 MHz, DMSO): 8.60 (s, 1H), 7.60-7.72 (m, 1H), 7.56 (d, 1H), 7.38-7.30 (m, 2H), 7.22-7.14 (m, 1H), 6.69-6.63 (m, 1H), 4.07- 4.04 (m, 1H), 3.95-3.90 (m, 2H), 3.72-3.68 (m, 1H), 3.05-3.01 (m, 2H), 2.47 (br s, 2H), 1.90 (s, 3H), 1.09 (s, 6H), 0.94 (t, 3H); MS (El) for C23H26F3lN403: 591 (MH+). EXAMPLE 3(www). 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(2-methylhydrazino)methyl]azetidin-3-ol acetate salt: ‘H NMR (400 MHz, DMSO): 8.54 (s, 1H), 7.57 (d, 1H), 7.38-7.30 (m, 2H), 7.19-7.12 (m, 1H), 6.69-6.63 (m, HI), 4.04-4.01 (m, 1H), 3.92-3.84 (m, 2H), 3.68-3.63 (m, 1H), 2.55 (s, 2H), 2.39 (s, 3H), 1.90 (s, 3H); MS (El) for C38H18F3IN402: 507 (MH+), EXAMPLE 3(xxx). 3 - [(azetidin-3 -ylamino)methy 1 ] -1 -({3,4-difluoro-2- [(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-oi acetate salt: 'H NMR (400 MHz, DMSO): 7.57 (d, 1H), 7.39-7.30 (m, 2H), 7.20-7.13 (m, 1H), 6.70-6.65 (m, 1H), 4.10- 4.04 (m, 1H), 3.90-3.83 (m, 2H), 3.78-3.67 (m, 3H), 3.61-3.53 (m, 1H), 3.48-3.42 (m, 2H), 2.61-2.54 (m, 2H), 1.90 (s, 3H); MS (El) for C20H20F3IN4O2: 533 (MFf). EXAMPLE 3(yyy). 1 -((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(l,3-thiazol-2-ylamino)methyl]azetidin-3-ol acetate salt: !H NMR (400 MHz, DMSO): 8.60 (s, 1H), 7.57 (d, 1H), 7.38-7.28 (m, 2H), 7.20-7.13 (m, 1H), 6.75 (d, 1H), 6.70-6.64 (m, 1H), 5.93 (d, 1H), 4.26-4.22 (m, 1H), 4.11-4.08 (m, 1H), 4.00-3.88 (m, 3H), 3.74-3.70 (m, 1H), 1.90 (s, 3H); MS (El) for C2oHii,F3iN4Q2S: 561 (MH1). EXAMPLE 3(zzz). 1-((3,4-difluoro-2-[(2-fluoro-4-iodopheny l)ammo]pheny 1} carbon yl)-3-( ([3- (methyloxy)pheny ijamino}methyl)azetidin-3-ol: NMR (400 MHz, DMSO): 8.57 (s, 1H), 7.56 (d, 1H), 7.38-7.30 (m, 2H), 7.20-7.12 (m, 1H), 6.95-6.91 (m, 1H), 6.70- 6.66 (m, 1H), 6.21-6.17 (m, 2H), 6.14-6.10 (m, 1H), 5.94 (s, 1H), 5.49-5.44 (m, 1H), 4.14-4.10 (m, !H), 3.98-3.93 (m, 2H), 3.78-3.75 (m, 1H), 3.65 (s, 3H), 3.21 (d, 2H); MS (El) for C24H21F3IN3O3: 584 (MH4). EXAMPLE 3(ab). 1-((3,4-difiuoro~2-[(2-iluoro-4-iodopheny l)amino]pheny 1} carbonyl)-3 -(([4- (methyloxy)phenyl]amino}methyl)azetidin-3-ol: ‘H NMR (400 MHz, DMSO): 8.56 (s, 1H), 7.58 (d, 1H), 7.39-7.30 (d, 2H), 7.20-7.13 (m, 1H), 6.71-6.66 (m, 3H), 6.55 (d, 2H), 5.93 (s, 1H), 5.00-4.95 (m, 1H), 4.14-4.08 (m, 1H), 3.98-3.92 (m, 2H)S 3.79- 3.74 (m, 1H), 3.63 (s, 3H), 3.13 (d, 2H); MS (El) for C24H2iF3IN303: 584 (MH4), EXAMPLE 3(ac). 1-((3,4-difluoro~2~[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-({[2-(ethyloxy)ethyl]arnino}methyl)azetidin-3-ol: 5H NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.36-7.33 (d, 1H), 7.31-7.26 (m, 1H), 7.08-7.00 (q, 1H), 6.65-6.58 (t, 1H), 4.24-4.16 (d, 1H), 4.08-3.98 (t, 2H), 3.92-3.85 (d, 1H), 3.60-3.55 (t, 2H), 3.54-3.47 (q, 2H), 3.01-2.96 (s, 2H), 2.94-2.89 (t, 2H), 1.20-1.15 (t, 3H); MS (El) for Ca^FsINaOa: 550 (MH4). EXAMPLE 3(ad). 3-(( [2,2-bis(methyloxy)ethyl] amino} methyl)-1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbony!)azeiidin-3-ol acetate salt: JH NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.37-7.32 (d, 1H), 7.30-7.24 (m, 1H), 7.08- 7.00 (q, 1H), 6.65-6.57 (t, 1H), 4.48-4.42 (t, 1H), 4.20-4.11 (d, 1H), 4.02-3.93 (t, 2H), 3.86-3.80 (d, 1H), 3.38-3.34 (s, 6H), 2.84-2.80 (s, 2H), 2.75-2.70 (d, 2H),1.93-1.87 (s, 3H); MS (El) for C21H23F3IN3O4: 566 (MH4). EXAMPLE 3(ae). 1-((3,4-dif!uoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbony 1)-3 - ([(3-hydroxypropyl)amino] methyl} azetidin-3-ol acetate salt: SH NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.38-7.33 (d, 1H), 7.32-7.26 (m, 1H), 7.09-7.00 (q, 1H), 6.66-6.58 (t, 1H), 4.31-4.23 (d, 1H), 4.16-4.05 (t, 2H), 3.99-3.89 (d, 1H), 3.70-3.64 (t, 2H), 3.26-3.22 (s, 2H), 3.11-3.04 (t, 2H), 1.93- 1.89 (s, 3H), 1.89-1.82 (t, 3H); MS (El) for C20H21F3IN3O3: 536 (MH4). EXAMPLE 3(af). 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)aniino]phenyl}carbonyl)-3-([(2-pyridin-4-ylethyl)amino]methyl}azeti din- 3-01 acetate salt: !H NMR (400 MHz, CD3OD): 8.36-8.32 (d, 2H), 7.38-7.33 (d, 1H), 7.26-7.14 (m, 3H), 7.00-6.91 (q, 1H), 4.12-4.04 (d, 1H), 3.96-3.88 (t, 2H), 3.80-3.73 (d, 2H), 2.92-2.74 (m, 6H), 1.87-1.84 (s, 3H); MS (El) for Q^FalN^i 583 (MH4). EXAMPLE 3(ag). l-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)-3-(( [1 -(phenylmethyl)pyrrolidin-3-yl]aniino}methyl)azetidin-3-ol acetate salt: !H NMR (400 MHzs CD3OD): 7.47-7.24 (m, 8H), 7.08-7.00 (q, 1H), 6.64-6.57 (t, 1H), 4.19-4.11 (d, IK), 4.05-3.81 (m, 5H), 3.52-3.44 (m, 1H), 3.09-2.99 (m, 2H), 2.91-2.76 (m, 3H), 1.93-1.91 (s, 3H), 1.82-1.71 (m, 1H); MS (El) for C28H28F3IN4O2: 637 (MH‘). EXAMPLE 3(ah). 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbonyl)-3-({[2-(2-thienyl)ethyl]amino} methyl)azetidin-3-ol acetate salt: 'H NMR (400 MHz, CD3OD): 7.47-7.42 (d, 1H), 7.36-7.31 (d, 1H), 7.30-7.24 (m, 1H), 7.21-7.17 (d, 1H), 7.08-7.00 (q, 1H), 6.93-6.89 (t, 1H), 6.86-6.83 (d, IK), 6.64-6.57 (t, 1H), 4.18-4.11 (d, 1H), 4.01-3.93 (t, 2H), 3.85-3.78 (d, 1H), 3.04-2.97 (t, 2H), 2.92-2.87 (t, 2H), 2.82-2.78 (s, 2H), 1.92-1.87 (s, 3H); MS (El) for C23H2tF3lN302S: 588 (MH+). EXAMPLE 3(ai), 3-[({2-[bis(l-methylethyl)amino]ethyl}amino)methy 1] -1 -({ 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt: 'H NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.36-7.33 (d, 1H), 7.31-7.26 (m, IK), 7.08-7.00 (q, 1H), 6.65-6.58 (t, 1H), 4.18-4.13 (d, 1H), 4.06-3.98 (t, 2H), 3.88-3.82 (d, 2H), 3.57-3.47 (q, 2H), 3.05-2.99 (t, 2H), 2.92-2.85 (t, 4H), 1.92-1.88 (s, 3H), 1.28-1.22 (d, 12H); MS (El) for C25H32F3iN402: 605 (MH^. EXAMPLE 3(aj). l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-({ [2- (phenyloxy)ethyl]amino}methyl)azetidin-3-ol acetate salt: *H NMR (400 MHz, CD3OD): 7.36-7.31 (d, 1H), 7.26-7.22 (d, 1H), 7.20-7.13 (m, 3H), 6.97-6.89 (t, 1H), 6.86-6.80 (m, 3H), 6.54-6.47 (t, 1H), 4.13-4.07 (d, 1H), 4.01-3.96 (t, 2H), 3.79-3.74 (d, 1H), 2.97-2.91 (t, 2H), 2.84-2.79 (s, 2H), 1.84-1.81 (s, 3H); MS (El) for C25H23F3IN3O3: 598 (MH+). EXAMPLE 3(ak). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-{[(2-hydroxypropyl)amino]methyl}azetidin-3-ol acetate salt: 5H NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.36-7.33 (d, 1H), 7.31-7.26 (m, 1H), 7.08-7.00 (q, 1H), 6.65-6.58 (t, 1H), 4.27-4.19 (d, 1H), 4.10-4.00 (m, 2H), 3.15-3.00 (t, 2H), 3.57-3.47 (q, 2H), 3.15-3.00 (t, 2H), 2.87-2.81 (d, 1H), 2.72-2.64 (t, 1H), 1.94-1.91 (s, 3H), 1.19-1.15 (d, 3H); MS (El) for C2oH2^3^303: 536 (MH+). EXAMPLE 3(am). l-({3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino] phenyl) carbonyl)-3-[({2~[(l- methylethyl)oxy]ethyl}amino)methyl]azetidin-3-ol acetate salt: ]H NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.36-7.33 (d, 1H), 7.31-7.26 (m, 1H), 7.08-7.00 (q, 1H), 6.65-6.58 (i, 1H), 4.21-4.13 (d, 1H), 4.04-3.95 (t, 2H), 3.88-3.82 (d, 1H), 3.64-3.51 (m, 3H), 2.89-2.84 (s, 2H), 2.83-2.77 (t, 2H), 1.91-1.89 (s, 3H), 1.15-1.12 (d, 6H); MS (El) for C22H25F3IN3O3: 564 (MH+). EXAMPLE 3(an). 1-((3,4- di fluoro-2- [(2-fluoro-4- iodophenyl)amino]phenyl}earbonyl)-3-{[(l-ethylpiperidin-3- yl)amino]methyi}azeti din-3 -oi acetate salt: *HNMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.36-7.33 (d, 1H), 7.31-7.26 (m, 1H), 7.08-7.00 (q, 1H), 6.65-6.58 (t, 1H), 4.17-4.10 (d, 1H), 4.04-3.95 (t, 2H), 3.88-3.82 (d, 1H), 3.24-3.06 (m, 2H), 2.95-2.75 (in, 6H), 2.76-2.46 (m, 2H), 1.93-1.90 (s, 3H), 1.74-1.62 (m, 1H), 1.44-1.31 (m, 1H), 1.28-1.20 (t, 3H); MS (El) for C24H28F3IN4O2: 589 (MH^). EXAMPLE 3(ao). 1-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-({[(5-methyl-l,3>4-oxadiazol-2-yl)methyl]amino}methy 1 )azetidin-3-ol acetate salt: *H NMR (400 MHz. CD3OD): 7.48- 7.43 (d, 1H), 7.36-7.33 (d, 1H), 7.31-7.26 (m, 1H), 7.08-7.00 (q, 1H), 6.65-6.58 (t, 1H), 4.20-4.13 (d, 1H), 4.00-3.90 (t, 2H), 3.83-3.75 (d, 1H), 2.84-2.78 (s, 2H), 2.53-2.48 (s, 2H), 1.93-1.87 (s, 3H); MS (El) for C2iH,9F3IN503: 574 (MH+). EXAMPLE 3(ap). l-({3,4-difluoro-2-[(2-fluoro-4- iodopheny l)amino] pheny 1} carbony 1)-3 - {[(1 -methy Ibuty l)amino]methy 1} azetidin-3-ol acetate salt: !H NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.38-7.33 (d, 1H), 7.32- 7.27 (m, 1H), 7.09-7.01 (q, 1H), 6.65-6.58 (t, 1H), 4.25-4.19 (d, 1H), 4.12-4.02 (t, 2H), 3.96-3.90 (d, 1H), 3.16-2.96 (m, 3H), 1.91-1.89 (s, 3H), 1.68-1.57 (m, 1H), 1.49- 1.29 (m, 3H), 1.23-1.18 (d, 3H), 0.99-0.92 (t, 3H); MS (El) for C22H25F3IN3O2: 548 (MH+). EXAMPLE 3(aq). 1 -({3,4-di fluoro-2- [(2-fluoro-4 - iodopheny l)amlno] pheny 1} carbonyl)-3 - {[(1 -methy lpropyl)amino] methyl} azeti din- 3 -ol acetate salt: lH NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.32- 7.26 (m, 1H), 7.09-7.01 (q, 1H), 6.65-6.58 (t, 1H), 4.27-4.20 (d, 1H), 4.14-4.03 (t, 2H), 3.98-3.92 (d, 1H), 3.20-3.16 (s, 2H), 3.07-2.97 (m, 1H), 1.91-1.89 (s, 3H), 1.80-1.70 (m, 1H), 1.54-1.41 (m, 1H), 1.26-1.22 (d, 3H), 1.00-0.94 (t, 3H); MS (El) for C21H23F3IN3O2: 534 (MH+). EXAMPLE 3(ar). l-({3,4~difluoro-2-[(2-fluoro-4- iodopheny l)amino] pheny 1} carbony 1)-3 - {[(2 -methy lbutvl)amino]methyl} azetidin-3 -ο I acetate salt: 'HNMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.32-7.26 (m, 1H), 7.09-7.01 (q, 1H), 6.65-6.58 (t, 1H), 4.26-4.19 (d, 1H), 4.10-4.01 (t, 2H), 3.94-3.87 (d, 1H), 3.05-2.99 (s, 2H), 2.77-2.70 (m, 1H), 2.61-2.54 (m, 1H), 1.91-1.89 (s, 3H), 1.73-1.61 (m, 1H), 1.49-1.39 (m, 1H), 1.24-1.12 (m, 1H), 0.94-0.84 (m, 6H); MS (El) for C22H25F3IN3O2: 548 (MH+). EXAMPLE 3(as). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(pentylamino)methyl]azetidin-3-ol acetate salt: 'Ή NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.32-7.26 (m, 1H), 7.09-7.01 (q, 1H), 6.65-6.58 (t, 1H), 4.29-4.23 (d, 1H), 4.15-4.05 (t, 2H), 3.98-3.90 (d, 1H), 3.21-3.18 (s, 2H), 2.93-2.86 (m, 2H), 1.91-1.89 (s, 3H), 1.70-1.60 (m, 2H), 1.42-1.29 (m, 4H), 0.97-0.90 (t, 3H); MS (El) for C22H25F3lN302: 548 (MH+), EXAMPLE 3(at). 3-[(cyclohexylamino)methyl]-l-({3,4~difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt: !H NMR. (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.38-7.34 (d, 1H), 7.33-7.27 (m, 1H), 7.09-7.01 (q, 1H), 6.65- 6.58 (t, 1H), 4.25-4.19 (d, 1H), 4.14-4.03 (t, 2H), 3.98-3.90 (d, 1H), 3.21-3.18 (s, 2H), 2.93-2.86 (m, 1H), 2.07-2.00 (d, 2H), 1.92-1.90 (s, 3H), 1.89-1.82 (d, 2H), 1.73- 1.66 (d, 1H), 1.42-1.14 (m, 5H); MS (El) for C23H25F3IN3O2: 560 (MU*). EXAMPLE 3(au). 3-[(azepan-3-ylamino)methy!]-1 -({3,4-difhioro-2- [(2-fluoro~4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt: NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.32-7.26 (m, 1H), 7.09-7.01 (q, 1H), 6.65- 6.58 (t, 1H), 4.19-4.13 (d, 1H), 4.05-3.95 (t, 2H), 3.90-3.81 (d, 1H), 3.37-3.34 (s, 2H), 3.22-3.03 (m, 2H), 2.91-2.64 (m, 3H), 1.93-1.89 (s, 3H), 1.88-1.52 (m, 6H); MS (El) for C23H26F3IN4O2: 575 (MH+). EXAMPLE 3(av). 1 -({3,4-difluoro-2- [(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-({[2-(2,3-dihydro-l//-indol-3-yl)ethyl]amino}methyl)azetidin-3-oi acetate salt: 'H NMR (400 MHz, CD3OD): 7.58-7.54 (d, 1H), 7.48-7.43 (d, 1H), 7.36-7.33 (d, 1H), 7.31-7.26 (m, 1H), 7.14-6.99 (m, 4H), 6.65-6.58 (t, 1H), 4.25-4.19 (d, 1H), 4.10-4.02 (t, 2H), 3.95-3.88 (d, 1H), 3.23-3.03 (m, 9H), 1.94-1.92 (s, 3H); MS (El) for C27H26F3IN4O2: 623 (MH+). EXAMPLE 3(aw). 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(l,3,5-triazin-2-ylamino)methyl]azetidin-3-ol acetate salt: Ή NMR (400 MHz, CD3OD): 8.48-8.46 (s, 1H), 8.36-8.34 (s, 1H), 7.48-7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.28-7.22 (m, 1H), 7.06-6.98 (q, 1H), 6.65-6.58 (t, 1H), 4.24-4.18 (d, 1H), 4.10-3.96 (t, 2H), 3.84-3.78 (d, 1H), 3.69-3.67 (s, 2H), 1.99-1.97 (s, 3H); MS (El) for C2oH!6F3rN602: 557 (MH+). EXAMPLE 3(ax). l-({3,4-difluoro-2-[(2-f!uoro-4- iodophenyl)amino] phenyl} carbony 1)-3 - {[(4- hydroxycyclohexyl)amino]methyl}azetidin-3-ol acetate salt: 'H NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.32-7.26 (m, 1H), 7.09-7.01 (q, 1H), 6.65-6.58 (t, 1H), 4.22-4.15 (d, 1H), 4.08-3.99 (t, 2H), 3.93-3.87 (d, 1H), 3.56-3.47 (m, 1H), 3.05-3.02 (s, 2H), 2.76-2.68 (m, 1H), 2.03-1.96 (m, 4H), 1.93-1.89 (s, 3H), 1.35-1.23 (m, 4H); MS (El) for C23H25F3IN3O3: 576 (MH+). EXAMPLE 3(ay). 3-[(cyclopent-3-en-1 -ylamino)methyl]-1-((3,4-diflnoro-2~[(2-f]uoro-4-iodophenyl)artuno]phenyl}carbonyl)azetidin-3-ol acetate salt: ]H NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.32-7.26 (m, 1H), 7.09-7.01 (q, 1H), 6.65-6.58 (t, 1H), 5.70-5.65 (s, 2H), 4.20-4.14 (d, 1H), 4.03-3.95 (t, 2H), 3.90-3.81 (d, 1H), 3.58-3.50 (m, 1H), 2.90-2.86 (s, 2H), 2.68-2.58 (m, 2H), 2.26-2.16 (m, 2H), 1.93-1.89 (s, 3H); MS (El) for C22H21F3lN302: 544 (MH+). EXAMPLE 3(az). iV-[4-({[3-({3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino]phenyl} carbonyl)-3 -hydro xyazetidin-3 - yl]methyl}amino)phenvl]acetamide acetate salt: JH NMR (400 MHz, CD3OD): 7.48- 7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.27-7.20 (m, 3H), 7.09-7.01 (q, 1H), 6.65-6.55 (m, 3H), 4.22-4.16 (d, 1H), 4.08-3.98 (t, 2H), 3.88-3.82 (d, 1H), 3.28-3.24 (s, 2H), 2.08- 2.05 (s, 3H), 2.91-2.64 (m, 3H), 1.93-1.89 (s, 3H); MS (El) for C25H22F3IN4O3: 611 (MET). EXAMPLE 3(ba). ;V-[3-({ [1 ~({3,4-difluoro-2-[(2-fluoro-4- i odopheny llamino] phenyl} carbony 1)-3 -hydroxyazetidin-3 - yl]methyl}amino)phenyl]acetamide acetate salt: *H NMR (400 MHz, CD3OD): 7.48- 7.43 (d, 1H), 7.36-7.33 (d, 1H), 7.27-7.20 (m, 1H), 7.04-6.96 (m, 3H), 6.72-6.68 (d, 1H), 6.65-6.58 (t, 1H), 6.40-6.35 (d, 1H), 4.24-4.18 (d, 1H), 4.08-3.98 (t, 2H), 3.87-3.81 (d, 1H), 3.28-3.25 (s, 2H), 2.10-2.07 (s, 3H), 1.97-1.95 (s, 3H); MS (El) for C25H22F3IN4G3: 611 (MHT). EXAMPLE 3(bc). (lR,2S)-4-({[l-({3,4-difluoro-2-[(2-fluoro-4- iodopheny l)amino]pheny!} carbonyl)-3 -hydroxyazetidin-3 -yljmethyl}amino)cyclopentane-1,2-diol acetate salt: !H NMR (400 MHz, DMSO): 8.58-8.54 (s, 1H), 7.61-7.53 (d, 1H), 7.39-7.28 (m, 2H), 7.21-7.13 (m, 1H), 6.71-6.63 (t, 1H), 5.58-5.64 (s, 1H), 5.63-5.58 (s, 1H), 4.06-4.01 (d, 1H), 3.90-3.84 (t, 2H), 3.72-3.66 (d5 1H), 3.31-3.26 (m, 3H), 2.61-2.57 (s, 2H), 2.46-2.36 (m, 2H), 2.02-1.93 (dd, 2H), 1.91-1.88 (s, 3H); MS (El) for C22H23F3IN3O4: 578 (MH+). EXAMPLE 3(bd). l-({3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino]pheny 1} carbonyl)-3-({[ 1 - (hydroxymethyl)cyclohexyl]amino}methyl)azetidin~3-ol acetate salt: JH NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.32-7.26 (m, 1H), 7.09-7.01 (q, 1H), 6.65-6.58 (t, 1H), 4.22-4.15 (d, 1H), 4.08-3.99 (t, 2H), 3.89-3.83 (d, 1H), 3.49-3.45 ($, 2H), 2.86-2.80 (s, 2H), 1.91-1.89 (s, 3H), 1.67-1.34 (m, 10H); MS (El) for C24H27F3IN3O3: 590 (MH+). EXAMPLE 3(be). 3-{[(3-chlorophenyl)amino]methyl)-1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbonyi)azetidin-3 -ol acetate salt: *H NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.37-7.33 (d, 1H), 7.32-7.26 (m, !H), 7.08-6.98 (m, 2H), 6.65-6.55 (m, 3H), 6.53-6.44 (d, 1H), 4.22-4.15 (d, 1H), 4.06-3.98 (t, 2H), 3.88-3.82 (d, IE), 3.27-3.24 (s, 2H), 1.91-1.89 (s, 3H); MS (El) for CisH^ClFjINaOa: 588 (MH*). EXAMPLE 3(bf). 3 - {[(4-chlorophenyl)araino]methyl )-1-((3,4-difluoro-2-[(2-fluoro- 4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt: *H NMR (400 MHz, CD3OD): 7.45-7.40 (d, 1H), 7.35-7.30 (d, 1H), 7.28-7.22 (m, 1H), 7.06-6.97 (m, 3H), 6.62-6.54 (m, 3H), 6.53-6.44 (d, 1H), 4.22-4.15 (d, 1H), 4.06-3.98 (t, 2H), 3.88-3.82 (d, 1H), 3.26-3.22 (s, 2H), 1.96-1.94 (s, 3H); MS (El) for C23H18CIF3IN3O2: 588 (MH+). EXAMPLE 3(bg). 3-[(5-amino-3-methyl-l//-pyrazol-l-yl)methyl]-l-({3,4-diflaoro- 2-[(2~fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt: 'H NMR (400 MHz, CD3OD): 7.38-7.33 (d, 1H), 7.28-7.24 (d, 1H), 7.21-7.15 (m, 1H), 6.98-6.90 (q, 1H), 6.56-6.49 (t, 1H), 5.16-5.14 (s, 1H), 4.36-4.30 (d, 1H), 4.22-4.16 (d, 1H), 3.99-3.97 (s, 1H), 3.95-3.90 (d, 1H), 3.77-3.71 (d, 1H), 1.96-1.92 (s, 3H), 1.85-1.82 (s, 3H); MS (El) for 02^^3^5()2: 558 (MH+). EXAMPLE 3(bh). l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)aniino]phenyl}carbonyl)-3-{[(5-methyl-l/f-pyrazoI-3-yl)amino]methy 1}azetidin-3-ol acetate salt: 'H NMR (400 MHz, CD3OD): 7.38-7.33 (d, 1H), 7.28-7.24 (d, 1H), 7.21-7.15 (m, 1H), 6.98-6.90 (q, 1H), 6.56-6.49 (t, 1H), 5.22-5.19 (s, 1H), 4.15-4.08 (d, 1H), 4.02-3.88 (m, 2H), 3.75-3.68 (d, 1H), 3.20-3.18 (s, 2H), 2.07-2.05 (s, 3H), 1.85-1.82 (s, 3H); MS (El) for CiiH^lNsC^: 558 (ΜΗΓ). EXAMPLE 3(bi). 3-[(diethylamino)methyl]-l-({3,4-difiuoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: 'H NMR (400 MHz, d6-DMSO): 8.54 (s, 1H), 7.58-7.55 (dd, 1H), 7.38-7.35 (dt, 1H), 7.33-7.31 (m, 1H), 7.22-7.15 (m, 1H), 6.69-6.64 (m, IK), 5.56 (b, IK), 4.06-4.04 (d, IK), 3.90-3.88 (m, 2H), 3.72-3.69 (d 1H), 2.51-2.49 (m, 6H), 0.86-0.83 (t, 6H); MS (El) for C21H23F3IN3O2: 534 (UK). EXAMPLE 3(bj). l-({3,4-difluoro-2-[(2~fluoro~4- iodophenyl)amino]pheny 1} carbonyl)-3 - [(dimethylamino)methy l]azetidin-3 -ol: 1H NMR (400 MHz, d6-DMSO): 8.56 (s, 1H), 7.59-7.56 (dd, 1H), 7.38-7.36 (dt, 1H), 7.34-7.33 (m, 1H), 7.21-7.14 (m, 1H), 6.71-6.65 (m, IK), 5.55 (b, 1H), 4.07-4.05 (d, 1H), 3.89-3.84 (t, 2H), 3.74-3.719 (d, 1H), 2.46 (m, 2H), 2.19 (br s, 6H); MS (El) for C^H^FsiNsCV. 506 (MH+). EXAMPLE 3(bk). 1 -({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbony 1)-3-( [(2-hydroxy-1,1-di methy lethy l)amino] me thy 1} azetidin- 3 -ol: 'H NMR (400MHz, CDCI3): 8.40 (s, 1H), 7.38 (dd, 1H), 7.33-7.30 (m, 1H), 7.12 (m, 1H), 6.85-6.79 (m, 1H), 6.63-6.57 (m, 1H), 4.22-4.ll(br m, 4H), 3.55 (s, 2H), 3.15 (s, 2H), 1.32 (s, 6H); MS (El) for C2JH23F3IN3O3: 550 (MH*). EXAMPLE 3(bm). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(prop-2-en-l-ylamino)methyl]azetidin-3-ol): !H NMR (400MHz, CDCI3): 8.47 (s, IK), 7.40 (dd, 1H), 7.34-7.31 (m, 1H), 7.12 (m, 1H), 6.83-6.77 (m, 1H), 6.64-6.59 (m, 1H), 6.64-6.59 (m, IK), 5.88-5.78 (m, 1H), 5.00-5.12 (m, 2H), 4.13 (br m, 4H), 3.26 (d, 2H), 2.88 (d, 2H), 2.02 (s, 1H); MS (El) for C2iH,9F3IN302: 518 (UK*). EXAMPLE 3(bn). 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-(([2-(tetrahydro-2H-pyran-4-yl)ethyl]amino}methyl)azetidin-3-ol): 'H NMR (400MHz, CDCI3): 8.45 (s, 1H), 7.39 (dd, 1H), 7.34-7.31 (m, 1H), 7.14-7.10 (m, 1H), 6.84-6.77 (m, 1H), 6.63-6.58 (m, IK), 4.26-4.04 (m, 4H), 3.95 (dd, 2H), 3.35 (t, 2H), 2.92 (d, 2H), 2.67 (m, 2H), 1.40- 1.25 (m, 8H); MS (El) for C24H27F3IN3O3: 590 (ΜΗ*). EXAMPLE 3(bo). l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl }carbony 1)-3- {[(1,1 -dimethylprop-2-yn-1 -yl)amino]methyl}azetidin-3-ol): *HNMR (400MHz, CDCls): 8.46 (s, 1H), 7.39 (dd, 1H), 7.33-7.30 (m, 1H), 7.15-7.11 (m, 1H), 6.84-6.77 (m, IK), 6.64-6.58 (m, IK), 4.20 (br, 1H), 4.07 (br, 1H), 2.92 (s, 2H), 1.58 (m, 4H), 0.92 (dd, 6h); MS (El) for C22H21F3IN3O2: 572 (MH+). EXAMPLE 3 (bp). 1 -({3,4-difluoro-2-[(2-fluoro-4-iodopheriyl)amino]phenyl} carbonyl)-3-( ([2-( 1 //-imidazol-4-yl)ethyl]amino}methyl)azetidin-3-ol): !H NMR (400MHz, CDCI3): 8.44 (s, 1H), 7.33-7.14 (m, 3H), 7.00 (m, 1H), 6.67 (dd, 1H), 6.59 (s, 1H), 6.44 (m, 1H), 3.93 (d, 2H), 2.75 (m5 2H); 2.60 (m, 1H), 2.42 (m, 1H) 2.02 (AcOH; s, 3H), 1.86 (m, 4H); MS (El) for C22H21F3IN5O2: 572 (MET). EXAMPLE 3(bq). l-({3,4-difluoro-2-[(2-fluoro-4- iodopheny l)aniino]pheny 1} carbony 1)-3-({[3-(eihy 1 oxy)propyl] amino} methy l)azetidin- 3-ol: 5H NMR (400MHz, CDCI3): 8.49 (s, 1H), 7.39 (dd, 1H), 7.34-7.31 (m, 1H), 7.14-7.10 (m, 1H), 6.83-6.76 (m, 1H), 6.64-6.58 (m, 1H), 4.26-4.03 (br m, 4H), 3.53-3.44 (m, 4H), 2.92-2.73 (m, 4H), 1.72 (m, 2H) 1.18 (t, 3H); MS (El) for C22H23F3IN3O3: 564 (MH+). EXAMPLE 3(br). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-{[(3,3-dimethylbuty!)amino]methy!}azetidin-3-ol: 'H NMR (400MHz, CDC13): 8.46 (s, 1H), 7.39 (dd, 1H), 7.34-7.31 (m, 1H), 7.14-7.10 (m, 1H), 6.84-6.77 (m, 1H), 6.63- 6.58 (m, 1H), 4.18 (br, 3H), 3.15 (s, 2H), 2.71 (m, 2H) 2.05 (AcOH; s, 3H), 1.43 (m, 2H), 0.90 (s, 9H); MS (El) for C23H27F3IN3O2: 562 (MH+). EXAMPLE 3(bs). !-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-{[(3-methylbiiiyl)amino]methyl}azetidin-3-ol: !H NMR (400MHz, CDC13): 8.46 (s, 1H), 7.39 (dd, 1H), 7.34-7.30 (m, 1H), 7.14-7.11 (m, 1H), 6.84-6.77 (m, 1H), 6.63- 6.59 (m, 1H), 4.27-3.61 (br m, 6H), 2.98 (m, 2H), 2.72 (t, 2H) 2.05 (AcOH; s, 3H), 1.61 (m,lH), 1.43 (m, 2H), 0.90 (d, 6H); MS (El) for C22H25F3IN3O2: 547 (MH+). EXAMPLE 3(bt). l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)-3 -({[3 - (dimethylamino)propyi]amino}methyl)azetidin-3-ol: MS (El) for C22H26F3IN4O2: 563 (MH4). EXAMPLE 3(bu). l-({3,4-diiluoro-2-[(2-fluoro-4-iodopheny!)amino]phenyl}carbonyl)-3-({[3-(l//-imidazol-l- yl)propyl]amino}methyl)azetidin-3-ol: !H NMR (400MHz, CDCI3): 8.46 (s, 1H), 7.53 (s, 1H), 7.40 (dd, 1H), 7.34-7.30 (m, 1H), 7.14-7.09 (m, 1H), 7.05 (s, 1H), 6.89 (s, 1H), 6.84-6.77 (m, 1H), 6.63-6.59 (m, 1H), 4.24-4.00 (br m, 6H), 2.84 (m, 2H), 2.61 (m5 2H), 1.94 (m, 2H); MS (El) for C23H21F3IN502: 586 (MH4). EXAMPLE 3(bv). l-({3,4-difluoro-2~[(2-fluoro-4~ iodophenyl)amino]phenyl}carbonyl)-3-({[2- (methylthio)ethyl]amino}methyl)azetidin-3-ol: 3H NMR (400MHz, CDCI3): 8.49 (s, 1H), 7.39 (dd, 1H), 7.34-7.31 (m, 1H), 7.14-7.11 (m, 1H), 6.83-6.77 (m, 1H), 6.63-6.59 (m, 1H), 4.26-4.03 (br m, 4H), 2.88 (s, 2H), 2.82 (t, 2H), 2.62 (t, 2H), 2.08 (s, 3H); MS (El) for C23H21F3IN3O2S: 552 (MB'5). EXAMPLE 3(bw). l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-{ [(1,1,3,3- tetramethylbutyl)amino]methyl}azetidin-3-ol: ‘H NMR (400MHz, CDC13): 8.49 (s, 1H), 7.38 (dd, 1H), 7.34-7.30 (m, 1H), 7.14-7.11 (m, 1H), 6.83-6.77 (m, 1H), 6.64- 6.59 (m, 1H), 4.25-4.01 (br m, 4H), 2.82 (s, 2H), 1.45 (s, 2H), 1.15 (s, 6H), 0.90 (s, 9H); MS (El) for C25H3fF3lN302: 590 (MH+). EXAMPLE 3(bx). l-({3,4-difiuoro-2-[(2-fluoro-4-iodopheny l)amino jphenyl} carbony 1)-3 - {[(1,1- dimethylpropyl)amino]methyl}azetidin-3-ol: !H NMR (400MHz, CDC13): 8.50 (s, 1H), 7.39 (dd, 1H), 7.35-7.30 (m, 1H), 7.15-7.11 (m, 1H), 6.83-6.77 (m, 1H), 6.65- 6.59 (m, 1H), 4.27-4.01 (br m, 4H), 2.82 (s, 2H), 1.46 (s, 2H), 1.08 (s, 6H), 0.89 (s, 3H); MS (El) for C22H21F3IN4O3: 548 (MH4). EXAMPLE 3(by). 3-{[(3-amino-2-hydroxypropyl)amino]methyl}-1-((3,4-difluoro- 2- [(2-iluoro-4-iodophenyl)amino]phenyl} carbonyl)azetidin-3 -ol; MS (El) for C23H22F3IN4O3: 551 (MR4). EXAMPLE 3(bz). 1 - {[ 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbonyl)-3-hydroxyazetidin-3-yl] methyl} pyrrolidin-3 -ol: MS (El) for C21H21F3IN3O3: 548 (MH4). EXAMPLE 3(ca). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-({(2S)-2-[(methyloxy)ineihyl]pyrrolidin-l-y 1}methyl)azetidin-3-ο 1: MS (El) for C23H25F3IN3Q3: 576 (MH4). EXAMPLE 3(cb). l-({3,4-difluoro-2-[(2-fluoro-4~ iodopheny l)am inojpheny 1} carbonyl)-3 - {[(2-hydroxyphenyl)amino]meihyl} azetidin- 3- ol: {H NMR (400MHz, CDC13): 8.46 (s, 1 Η), 7.41 (dd, 1H), 7.35-7.30 (m, 1H), 7.15-7.11 (m, 1H), 6.89-5.98 (m, 6H), 4.92 (s, 1H), 4.28-4.05 (br m, 4H), 3.44 (s, 2H); MS (El) for C23H19F3IN3O3: 570 (ΜΗ*). EXAMPLE 3(cd). 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-([(4-hydroxyphenyl)amino]methyl}azetidin-3-ol: ‘H NMR (400MHz, CDCI3): 8.46 (s, 1H), 7.78 (s, 1H), 7.40-7.05 (m, 4H), 6.72 (m, 1H), 6.62 (d, 1H), 6.50 (m, 1H), 6.42 (d, 1H) 4.04-3.98 (m, 4H), 3.18 (s, 2H); MS (El) for C23H19F3IN3Q3: 570 (MH+). EXAMPLE 3(ce). 1 -({3,4~difluoro-2-[(2-fluoro-4- iodophenyl)amino]pheny l}carbonyl)-3- {[(3-hydroxyphenyl)amino]meihyl) azetidin-3-ol: * H NMR (400MHz, CDCI3): 8.52 (s, 1H), 8.22 (s, 1H), 7.39 (dd, 1H), 7.34-7.31 (m, 1H), 7.14-7.11 (m, 1H), 6.85 (dd, 1H), 6.84-6.77 (m, 1H), 6.63-6.59 (m, 1H), 6.15 (d, 1H) 6.09-6.01 (m, 3H), 4.16-3.95 (br m, 4H), 3.22 (d, 2H) 2.15 (AcOH; s, 3H); MS (El) for C2iU]9F3mOy. 570 (MH*). EXAMPLE 3(cf). 1-((3,4-difl uoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3~[(phenyloxy)methyl]azetidin-3-o!: MS (El) for C23H18F3IN2O3: 555 (MH+). EXAMPLE 3(cg). 3-({[1-((3, 4-difluoro-2-[(2-fluoro-4- iodophenyl)ami nojpheny 1} carbonyl)-3-hydroxyazetidin-3 -y IJmeihy 1} amino)propane- 1,2-diol: MS (El) for C20H21F3IN3O4: 552 (MH+). EXAMPLE 3(ch). 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)aminojpheny 1}carbony 1)-3-[(phenylthio)methyl]azetidin-3-ol: !H NMR (400MHz, CDCb): 8.46 (s, 1H), 7.45-7.23 (m, 5H), 7.14-7.05 (m, 1H), 6.78 (dd, 1H), 6.60 (m, 1H), 4.14- 3.92 (br m, 4H), 3.33 (s, 2H); MS (El) for CisHigFsWOj: 571 (ΜΗ1-). EXAMPLE 3(d). 1 -((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-([(4-hydroxybutyl)amino]methyl}azetidin-3-ol): 'H NMR (400MHz, CDC13): 8.43 (s, 1H), 7.38 (dd, 1H), 7.34-7.30 (m, 1H), 7.14-7.10 (m, 1H), 6.84-6.77 (m, 1H), 6.63-6,58 (m, 1H), 4.26-4.04 (m, 4H), 3.61 (m, 2H), 2.96 (s, 2H), 2.73 (s, 2H); MS (El) for C21H23F3IN3O3: 550 (MH*). EXAMPLE 3(cj). 1 -((3,4-difluoro-2~[(2-fliK>ro-4- iodophenyl)amino]pheny 1} carbonyl)-3 - {[(2-hydro xyethyl)oxy]methyi} azetidin-3 -ol: 'HNMR (400MHz, CDC13): 8.51 (s, 1H), 7.39 (dd, 1H), 7.35-7.31 (m, 1H), 7.14-7.11 (m, 1H), 6.84-6.77 (m, 1H), 6.63- 6.59 (m, 1H), 4.21-4.05 (br m, 4H), 3.77 (m, 2H), 3.66 (m, 2H); MS (El) for C19H18F3IN2O4: 523 (MH+). EXAMPLE 3(ck). 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-(([(lS,2S)-2- hydroxycyclohexyl3amino}methyl)azeiidin-3-ol): MS (El) for C23H25F3IN3O3: 576 (MH+). EXAMPLE 3(cm). 1-((3,4-difluoro-2- [(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-{ [(1,1 -dimethyl-2-pyrrolidin-l -ylethyl)amino]methyl}azetidin-3-ol: 'H NMR (400MHz, CDCI3): 8.49 (s, 1H), 7.39 (dd, 1H), 7.34-7.29 (m, 1H), 7.14-7.11 (m, 1H), 6.83-6.77 (m, 1H), 6.64- 6.59 (m, 1H), 4.25-4.07 (br m, 4H), 2.88 (d, 2H), 2.62 (m, 4H), 2.58 (m, 2H), 1.78 (m, 4H), 2.05 (AcOH; s, 3H); MS (El) for C25H30F3IN4O2: 603 (MH+). EXAMPLE 3(cn). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino] phenyl} carbony 1)-3 -({[(1 -methyl -1 //-imidazol-4-yl)methyl]amino}methyl)azetidin-3-ol: *H NMR (400MHz, CDCI3): 8.50 (s, 1H), 7.41-7.11 (m, 3H), 7.12 (m, 1H), 6.85-6.79 (m, 2H), 4.12-3.98 (br m, 4H), 3.78 (s, 2H), 3.66 (s, 3H), 2.95 (s, 2H), 2.08 (AcOH; s, 4H) ,2.05 (AcOH; s, 3H); MS (El) for C22H21F3IN5O2: 572 (MFT). EXAMPLE 3(co). 1 -({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-({[(l -methyl-1 /f-imidazol-5-yl)methyl]amino}methyl)azetidin-3-ol: 'H NMR (400MHz, CDCI3): 8.45 (s, 1H), 7.47 (s, 1H), 7.39 (dd, 1H), 7.33-7.30 (m, 1H), 7.15-7.10 (m, 1H), 6.91 (s, 1H), 6.87-6.77 (m, 1H), 6.63-6.58 (m, 1H), 4.18-4.02 (m, 4H), 3.3.80 (s, 2H), 3.62 (s, 3H), 2.90 (s, 1H), 2.05 (AcOH; s, 3H); MS (El) for C22H21F3IN5O2: 572 (MH+). EXAMPLE 3(cp). 1-((3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino]pheny 1} carbonyl)-3 -(([(2S)-2- (methyloxy)cyclopentyl]amino}methyl)azetidin-3-ol): MS (El) for C23H25F3IN3O3: 576 (ΜΗ*). EXAMPLE 3(cq). 1-((3,4-difluoro-2-[(2-fluoro-4- iodopheny])amino]pheny 1} carbonyl)-3 -(([(1 R)-2-hydroxycyciohexyl]amino}methyi)azetidin~3-o!): MS (El) for C23H25F3IN3O3: 576 (MH+). EXAMPLE 3(cr). A-[3-({[l-({3,4-difluoro-2-[(2-fluoro-4-iodopheny l)amino]pheny 1} carbony 1)-3 -hydroxyazetidin-3 -yl]methyl}amino)phenyl]methanesulfonaxnide: 1H NMR (400MHz, CDCh): 7.33 (dd, 1H), 7.22 (m, 1H), 7.08 (dd, 1H), 6.83-6.77 (m, 1H), 6.03-5.98 (m, 2H), 6.64- 6.59 (m, 1H), 4.08-3.77 (br m, 5H), 2.88 (s, 3H); MS (El) for C24H22F3IN4O4S: 647 (MH^. EXAMPLE 3(cs). 3-{[(4-aminophenyi)amino]methyi}-l-({354-difluoro-2-[(2-fluoro- 4-iodophenyl)amino]phenyl}carbonyi)azetidin-3-ol: !H NMR (400MHz, CDCI3): 8.44 (s, 1H), 7.39 (dd, 1H), 7.34-7.30 (m, 1H), 7.14-7.10 (m, 1H), 6.84-6.77 (m, 1H), 6.64-6.53 (m, 5H), 4.22-4.04 (br m, 4H), 3.34 (s, 2H); MS (El) for C23H20F3IN4O2: 569 (MH+). EXAMPLE 3(ct). l-({3,4-dif!uoro-2-[(2-f!uoro-4-iodophenyl)amino]phenyl} carbony 1)-3 - {[(2-hydroxy-2- methylcyclopentyl)amino]methyl}azetidin-3-ol: MS (El) for C23H25F3IN3O3: 576 (MH+). EXAMPLE 3(cu). 3- [(cyclopentylamino)methyl]-1 -({3,4-difiuoro-2-[(2-fluoro-4- iodophenyl)amino]pheny 1} carbonyl)azetidin-3 -ol: 3H NMR (400 MHz, CD3OD): 7.44 (dd, 1H), 7.36-7.31 (m, 1H), 7.30-7.24 (m, 1H), 7.09-6.99 (m, 1H), 6.64-6.57 (m, 1H), 4.17-4.10 (m,lH), 4.01-3.91 (m, 2H), 3.87-3.79 (m, 1H), 3.07-2.97 (m, 1H), 2.75 (s, 2H), 1.92-1.79 (m, 2H), 1.75-1.62 (m, 2H), 1.61-1.47 (m, 2H), 1.37-1.22 (m, 2H). MS (El) for C22H23F3IN3O2: 546 (MH+) EXAMPLE 3(cv). 3-{[(cyclohexylmethyl)amino]methyl}-l-({3,4-difluoro-2-[(2-
fluoro-4-iodopheny])amino]pheny!}carbonyl)azetidin-3-ol acetate (salt): !H NMR (400 MHz, CD3OD): 7.46 (dd, 1H), 7.39-7.32 (m, 1H), 7.31-7.25 (m, 1H), 7.11-6.99 (m, 1H), 6.67-6.57 (m, 1H), 4.27-4.15 (m, 1H), 4.12-3.97 (m, 2H), 3.96-3.85 (m, 1H), 3 (s,2H), 2.62 (d, 2H), 1.90 (s, 3H), 1.82-1.45 (m, 6H), 1.40-1.07 (m, 3H), 1.04-0.80 (m, 2H). MS (El) for C24H27F3IN3O2: 574 (Mrf). EXAMPLE 3(cw). l-({3,4-difluoro-2-[(2-fluoro-4-
iodophenyl)amino]phenyl}carbonyl)-3-[(propylamino)methyl]azetidin-3-ol: NMR (400 MHz, de-DMSO): δ 8.56 (s, 1H), 7.57 (dd, 1H), 7.37 (dd, 1H), 7.32 (m, 1H), 7.18 (m, IU), 6.67 (m, 1H), 4.03 (d, 1H), 3.89 (m, 2H), 3.69 (d, !H), 2.59 (s, 2H), 2.42 (ΐ, 2Η), 1.90 (s, 3Η), 1.32 (m, 2H), 0.81 (t, 3H); MS (El) for C20H2iF3IN302: 520 (MH+). EXAMPLE 3(cx). 1-((3,4-difl uoro-2- [(2-fIuoro-4- iodophenyl)amino]phenyl}carbonyl)~3-([(2-methylpropyl)aminQ]methyl}azetidin-3-ol: 'HNMR (400 MHz, d6-DMSO): δ 8.56 (s, 1H), 7.56 (dd, 1H), 7.36 (dd, 1H), 7.31 (m, 1H), 7.18 (m, 1H), 6.67 (m, 1H), 4.02 (d, 1H), 3.89 (m, 2H), 3.70 (d, 1H), 2.57 (s, 2H), 2.27 (d, 2H), 1.91 (s, 3H), 1.55 (m, 1H), 0.79 (d, 6H); MS (El) for C2iH23F3lN302: 534 (MH+). EXAMPLE 3(cy). methyl (2xi)-2-deoxy-2-({[l-({3,4-dif!uoro-2-[(2-fluoro-4-iodophenyl)ami nojpheny 1} carbony 1)-3 -hydroxy azetidin-3 -yljmethyl} amino)-beta-D-arabino-hexopyranoside: !H NMR (400 MHz, dj-methanol, ~3:1 mixture of anomers): δ 7.46 (d, 1H), 7.34 (d, 1H), 7.28 (m, 1H), 7.04 (q, 1H), 6.62 (m, 1H), 4.19-5.92 (m, 4H), 3.87-3.78 (m, 2H), 3.68 (m, 1H), 3.56-3.18 (m, 5H), 2.99-2.82 (m, 3H), 2.56 (m, 0.25H), 2.29 (m, 0.75H) MS (El) for C24H27F3IN307: 652 (M-H).
EXAMPLE 3(cz). 3 -({[3 -(diethy lamino)propy l]amino} methyl)-1 -({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt: NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.38-7.33 (d, 1H), 7.32-7.26 (m, 1H), 7.09- 7.00 (q, 1H), 6.66-6.58 (t, IB), 4.24-4.16 (d, 1H), 4.11-3.99 (t, 2H), 3.92-3.85 (d, 1H), 3.10-3.02 (m, 8H), 2.99-2.96 (s, 2H), 2.92-2.87 (t, 2H), 1.93-1.87 (s, 3H), 1.27-1.20 (t, 6H); MS (El) for C^^FalN^: 591 (MH+). EXAMPLE 4 1-((3,4-difluoro-2~[(2-fluoro-4-iodophenyI)amino]phenyl}carbonyl)-iV-(2- hydroxyethyl)azetidme-3-carboxamide
(00335] To a solution of 1 -((3,4-difluoro~2~[(2-fluoiO-4- iodophenyl)amino]pheny 1}carbonyl)azetidine-3-carboxylic acid (15 mg, 0.03 mmol), prepared using procedures similar to those in Example 1, in N, A-dimethylformamide (2.00 mL) was added HBTU (38 mg, 0.10 mmol). The mixture was stirred for 15 minutes at room temperature followed by the addition of 2-aminoethanol (3.6 pL, 0.06 mmol) and jV-methylmorpholine (110 pL, 1.00 mmol). The mixture was allowed to stir at room temperature for 3 d, then diluted the mixture with chloroform (20 mL), and washed with water (30 mL). The aqueous phase was back extracted with chloroform (10 mL). The combined organic phases were dried over sodium sulfate, filtered and the filtrate concentrated in vacuo, The residue was purified by high pressure liquid chromatography to afford the title compound (9.20 mg, 58%) as the trifluoroacetic acid salt: !H NMR (400MHz, CDCI3): 8.54 (s, 1H), 7.41-7.37 (m, 1H), 7.34-7.31 (m, 1H), 7.18-7.14 (m, 1H), 6.85-6.77 (m, 1H), 6.64-6.58 (m, 1H), 4.66 (br, 1H), 4.40-4.24 (br, 3H), 3.83-3.23 (br m, 7H), 1.18 (t, 3H); MS (El) for C19H17F3IN3O3: 542 (MNa+). |00336] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: EXAMPLE 4(a): l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-./V-(3,4-dihydroxybutyl)azetidine-3-carboxamide: !H NMR (400 MHz, CDC13): 8.55 (s, 1H), 7.40 (dd, 1H), 7.31-7.35 (m, 1H), 7.14-7.18 (m, 1H), 6.78-6.84 (m, 1H), 6.59-6.65 (m, 1H), 6.14 (br s, 1H), 4.50-4.60 (m, 1H), 4.20-4.40 (m, 3H), 3.60-3.80 (m, 3H), 3.40-3.52 (m, 2H), 3.20- 3.32 (m, 2H), 1.96 (br s, 1H), 1.18-1.28 (m, 2H). MS (El) for C21H21F3IN3O4: 562 (M-H). EXAMPLE 4(b): iV-butyl-1 -({ 3,4-difluoro-2-[(2-fiuoro-4- iodophenyl)amino]phenyl}carbonyl)azetidine-3-carboxamide: Ή NMR (400MHz, CDCI3): 8.53 (s, 1H), 7.39 (dd, 1H), 7.33-7.31 (m, 1H), 7.17-7.13 (m, 1H), 6.83-6.77 (m, 1H), 6.64-6.58 (m, 1H), 5.50 (m, 1H), 4.57 (br, 1H), 4.29 (br m, 3H), 3.27 (m, 3H), 1.49 (m, 1H), 1.33 (m, 2H), 0.92 (t, 3H); MS (El) for C21H2iF3rN302: 532 (MH+), 554 (MNa+). l~({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbony!)-iV-prop-2-en-l-ylazetidine-3-carboxamide: 1H NMR (400MHz, CDC13): 8.54 (s, 1H), 7.39 (dd, 1H), 7.34-7.31 (m, 1H), 7.17.7.12 (m, 1H), 6.83-6.77 (m, 1H), 6.64-6.58 (m, 1H), 5.88-5.77 (m, 1H), 5.57 (br, 1H), 5.21-5.16 (m, 2H), 4.59 (br, 1H), 4.30 (br m, 3H), 3.9 (tt, 2H), 3.32-3.25 (m, 1H)); MS (El) for C2oH|7F3IN302: 516 (MH4), 538 (MNa4). EXAMPLE 4(c): 1 -({3,4-difluoro-2-[(2-fiuoro-4-
iodophenyl)amino]phenyl}carbonyl)-iV-ethylazetidine-3-carboxamide: !H NMR (400MHz, CDCI3): 8.54 (s, 1H), 7.38 (dd, 1H), 7.33-7.30 (m, 1H),7.17-7.12 (m, 1H), 6.83-6.77 (m, 1H), 6.63-6.57 (m, 1H), 5.55 (br s, 1H), 4.57 (br s, 1H), 4.28 (br m, IH), 3..36-3.29 (m, 2H), 3.27- 3..20 (m, IH), 1.15 (t, 3H); MS (El) for Ci9H,7F3rN302: 504 (MH+), 526 (MNa4). EXAMPLE 4(d): 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-jV-(2-hydroxyethyl)azetidine-3-carboxamide: Ή NMR (400MHz, CDCI3): 8.50 (s, 1H), 7.39 (dd, 1H), 7.33-7.30 (m, 1H),7.16-7.12 (m, 1H), 6.84-6.77 (m, IH), 6.63-6.57 (m, IH), 4.57 (br, IH), 4.28 (br, 3H), 3.73 (t, 2H), 3.49-3.44 (m, 2H), 3.33-3.27 (m, IH), 2.18 (br, IH); MS (El) for C,9H ^3^3()3: 542 (MNa*). EXAMPLE 4(e): 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-Ar-(2-piperidin-l-ylethyl)azetidine~3-carboxamide: fH NMR (400MHz, CDC13): 11.28 (s, IH), 8.55 (s, IH), 7.38 (dd, IH), 7.33-7.30 (m, IH), 7.15-7.10 (m, IH), 6.82-6.76 (m, IH), 6.63-6.58 (m, IH), 4.42 (b, IH), 4.26 (br m, 3H), 3.68 (br s, 2H), 3.58 (br d, 2H), 3.36 (br m, 1H)3.17 (br s, IH), 2.63 (m, 4H), 1.92 (m, 5 H); MS (El) for 587 (MH+). EXAMPLE 4(f); 1-((3,4-difiuoro-2-[(2-fluoro-4- iodopheny!)amino]phenyl}carbonyl)-A-phenylazetidine-3-carboxamide: !H NMR (400MHz, CDCI3): 8.52 (s, IH), 7.50 (d, IH), 7.41-7.27 (m, 4H), 7.16 (m, 2H), 6.85-6.78 (m, IH), 6.65-6.59 (m, IH), 4.37 (br, 3H), 3.43 (m, IH); MS (El) for C23H17F3IN3O2: 574 (MNa+). EXAMPLE 4(g): jV-[2-(diethy lamino)ethyl]-1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidine-3-carboxamide: 'Η NMR (400MHz, CDQ3): 11.43 (s, IH), 8.90 (s, IH), 8.55 (s, IH), 7.39 (dd, IH), 7,33-7.30 (m, IH), 7.15-7.10 (m, IH), 6.87-6.77 (m, IH), 6.63-6.58 (m, IH), 4.44-4.22 (m, 4H), 3.65 (m, 2H), 3.38 (m, IH), 3.19-3.13 (m, 5H), l,33(t, 6H); MS (El) for C2,H2iF3lN302: 575 (MH+). EXAMPLE 4(h): 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-//-[(2,3-dihydroxypropyl)oxy]azetidine-3-carboxamide: MS (El) for C20H19F3IN3O5: 566 (MH1). EXAMPLE 4(5): l-({3,4-Difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-Ai-(2,3-dihydroxypropyi)azetidine-3-carboxamide: 3H NMR (400 MHz, CDCI3): 8.40 (br s, IH), 7.35 (dd, IH), 7.30 (br d, IH), 7.16-7.09 (m, IH), 6.89-6.76 (m, 2H), 6.58 (ddd, IH), 4.58-4.40 (br, IH), 4.27 (brt, 2H), 4.22-4.14 (br, 1H), 4.08-3.12 (m, 5H), 2.18-1.82 (br, 2H); MS (El) for C20H19F3IN3O4: 550 (MH4). EXAMPLE 4{j): 1 -({3,4-Difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-iV-hydroxyazetidine-3-carboxamide: *H NMR (400 MHz, CDCI3): 8.23-8.10 (b, 1H), 7.35-7.28 (m, 2H), 7.14-7.07 (m, 1H), 6.86-6.80 (m, 1H), 6.60-6.54 (m, 1H), 4.52-4.38 (b, 1H), 4.32-4.08 (m, 3H), 3.30-3.21 (m, 1H); MS (El) for C^HoFsINaCV. 492 (MH+). EXAMPLE 5 6~({3“(dimethyiammo)meihyllazeiidiK~l~yl}carl>oo[yl)-2,3-difluoro-A-(2-lliiorO"4- iodophenyI)amline
[00337] A mixture of l-({3.4-difluoro-2-[(2-fluoro~ 4-iodophenyl)amino]phenyl}carbonyl)azetidine-3-carboxylic acid (196 mg, 0.41 mmol), prepared using procedures similar to those in Example 1, triethylamine (58 pL, 0.41 mmol), PyBOP (213 mg, 0.41 mmol) and sodium borohydride (48 mg, 1.24 mmol) in tetrahydrofuran (2 mL) was stirred at room temperature for 15 hours. The reaction mixture was concentrated in vacuo and the resultant residue was partitioned between 20% aqueous citric acid and ethyl acetate. The organic portion was washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a colorless residue that was purified by column chromatography. Eluting with 60% ethyl acetate in hexanes, isolated product was concentrated in vacuo to afford 48 mg, 0.11 mmol (25%) of [ 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-yl]methanol as a white solid. !H NMR (400 MHz, CDC13): 7.44 (d, 1H), 7.34 (d, 1H), 7.28-7.23 (m, 1H), 7.04-6.97 (m, III), 4.26-4.18 (m, 1H), 4.02-3.94 (m, 2H), 3.78-3.72 (m, 1H), 3.03 (d, 2H), 3.34 (s, 1H), 2.80-2.71 (m, 1H). MS (El) for Ci7Hi4F3IN20: 463 (MH4).
[00338] A solution of l-({.3,4-diiluoro-2-[(2-fluoro- 4-iodophenyl)amino]phenyl}carbonyi)azetidin-3-yl]methanol (48 mg, 0.11 mmol), 1,4~diazabicyclo[2.2.2]octane (18 mg, 0.16 mmol) and melhanesulfonyl chloride (10 pL, 0.13 mmol) in tetrahydrofuran (2 mL) was stirred at room temperature for 15 minutes. The mixture was then partitioned between water and ethyl acetate. The organic portion was washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a colorless residue which was purified by column chromatography. Eluting with 70% ethyl acetate in hexanes, isolated product was concentrated in vacuo to afford 28 mg, 0.05 mmol (47%) of [ 1 -({3,4-difluoro-2- [(2-fluoro~4-iodophenyl)amino]phenyi}carbonyl)azetidin-3~yl]methyl methanesulfonate as a colorless residue which was immediately dissolved in ethylene glycol dimethyl ether (2 mL). To the solution was added dimethylamine (excess) and the solution was stirred in a seal tube at 50 °C for 15 hours. The reaction mixture was concentrated in vacuo, and the resultant residue was purified by preparative reverse phase HPLC. Isolated product was concentrated in vacuo to afford 12 mg, 0.02 mmol (40%) of 6-({3-[dimethylamino)methyl]azetidin-l-yl}carbonyl)-2,3-difluoro-jV-(2~fluoro-4-iodophenyl)aniline acetate salt as a white solid. *H NMR (400 MHz, DMSO): 8.54 (br s, 1H), 7.58 (d, 1H), 7.37 (d, 1H), 7.33-7.28 (m, 1H), 7.18-7.12 (m, 1H), 6.70-6.64 (m, 1H), 4.18-4.12 (m, 1H), 3.99-3.76 (m, 1H), 3.52-3.47 (m, 1H), 2.52-2.48 (m, 1H), 2.39 (d, 2H), 1.85 (s, 6H); MS (El) for C19H19F3IN3O: 490 (MH+).
[00339} Using the same or analogous synthetic techniques and/or substituting with alternative reagents, the following compounds of the invention were prepared: EXAMPLE 5(a): 2,3-difluoro-jV-(2-fluoro-4-iodophenyl)-6-[(3-{[(l-methy!ethyl)amino]methyl}azetidin-l-yl)carbony!]aniline: !H NMR (400 MHz, CDCI3): 8.54 (s, 1H), 7.40 (dd, 1H), 7.31-7.33 (m, 1H), 7.11-7.15 (m, 1H), 6.76-6.82 (m, 1H), 6.58-6.64 (m, 1H), 4.23-4.30 (m, 2H), 3.90-4.00 (m, 1H), 3.76-3.84 (m, 1H), 2.69-2.85 (m, 4H), 1.05 (d, 6H). MS (El) for C20H21F3IN3O: 502 (M-H). EXAMPLE 5(b): 2-({[ 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbony!)azetidin~2-yl3methyl}amino)ethanol: MS (El) for C19H19F3IN3O2: 506 (MH+). EXAMPLE 5(c): jV-([l-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl }carbonyl)azetidin-2-yl]methyl} ethane-1,2-diamine: MS (El) for C19H20F3IN4O: 505 (MH4). EXAMPLE 5(d): 6-((3-[dimethylamino)methyl]azetidin-1 -yl}carbonyl)-2,3-difluoro-iV-(2-fluoro-4-iodophenyl)aniline acetate salt: fH NMR (400 MHz, DMSO): 8.54 (br s3 1H), 7.58 (d, 1H), 7.37 (d, 1H), 7.33-7.28 (m, 1H), 7.18-7.12 (m, 1H), 6.70-6.64 (m, 1H), 4.18-4.12 (m, 1H), 3.99-3.76 (m, 1H), 3.52-3.47 (m, 1H), 2.52- 2.48 (m, 1H), 2.39 (d, 2H), 1.85 (s, 6H); MS (El) for C^Hi^INjO: 490 (ΜΗ4). EXAMPLE 6 1-((3,4-Difluoro-2-[(2-fIuoro-4-iodophenyl)amino]phenyl}carbonyl)azetidm-3- one
[00340] 1-((3,4-Difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol (132 mg, 0.295 mmol)ures similar to those in Example 1, was dissolved in dichloromethane (8 mL) and cooled to 0 °C. Dess-Martin periodinane (187 mg, 0.441 mmol) was added and the mixture was stirred at ambient for 2 h. The mixture was quenched with saturated sodium bicarbonate solution: 10% sodium thiosulfate solution (1:1; 6 mL) and diluted with ethyl acetate. The organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 40-50% ethyl acetate in hexanes) gave 1-((3,4-difluoro-2-[(2~fluoro-4-iodophenyl)amino]phenyI}carbonyi)azetidin-3~one (122 mg, 0.273 mmol, 93% yield): NMR (400 MHz, CDC13): 8.43 (br s, 1H), 7.44-7.38 (m, IE), 7.36-7.32 (m, 1H), 7.27-7.20 (m, 1H), 6.86 (ddd, 1H), 6.64 (ddd, 1H), 4.94-4.93 (m, 4H); MS (El) for C16H10F3IN2O2: 447 (MH+). EXAMPLE 7 1-((3,4~Difluoro-2-[(2-fluoro~4~iodopherayl)ammo]phenyl}earboayl)~3-(hydroxymethyl)azetidin-3-ol
[00341] Methyl triphenylphosphonium bromide (508 mg, 1.42 mmol) was treated with potassium rm-butoxide (159 mg, 1.42mmol) in tetrahydrofdran (5 mL) at 0 °C for 10 minutes. l-({3,4-Difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-one (270 mg, 0.605 mmol), prepared using procedures similar to those described in Example 6, was dissolved in tetrahydrofuran (2 mL) and was added to the mixture. The mixture was stirred at ambient for 15 h and then the mixture was filtered and the filtrate was partitioned between ethyl acetate and water. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 20% ethyl acetate in hexanes) gave 2,3-difluoro-Ar-(2-fluoro-4“iodophenyl)-6-[(3-methylideneazetidin-l~yl)carbonyl]aniline (57 mg, 0.128 mmol, 21% yield): 'H NMR (400 MHz, CDC13): 8.56 (br s, 1H), 7.39 (dd, 1H), 7.35-7.30 (m, 1H), 7.18- 7.12 (m, 1H), 6.86-6.76 (m, 1H), 6.62 (ddd, 1H), 5.14-5.00 (br, 2H), 4.74 (br d, 4H); MS (El) for C1?Hi 2F3IN20: 445 (MR").
[00342] 2,3-Difluoro-iV-(2-fluoro-4-iodophenyl)-6-[(3~methylideneazetidin-l-yl)carbonyl]ani!ine (56 mg, 0.126 mmol) and 4-methylmorpholine iV-oxide (44 mg, 0.376 mmol) were dissolved in acetone / water (4:1; 10 mL) and osmium tetroxide (4 wt.% in water; 0.7 mL) was added. The solution was stirred at ambient for 4 h, then was quenched with saturated sodium bisulfite (2 mL) and concentrated in vacuo. The residue was partitioned between ethyl acetate and water. The organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 80% ethyl acetate in hexanes) and then reverse phase HPLC gave l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-(hydroxymethyl)azetidin-3-ol (17 mg, 0.036 mmol, 28% yield): Ή NMR (400 MHz, CDC13): 8,43 (br s, 1H), 7.40 (dd, 1H), 7.35-7.31 (m, 1H), 7.16-7.10 (m, 1H), 6.81 (ddd, 1H), 6.61 (ddd, 1H), 4.25-4.00 (m, 4H), 3.78 (s, 2H); MS (El) for C17HMF3IN2O3: 479 (MH+). EXAMPLE 8 3-(2-ammopyrimidin-4~yI)-l-({3,4-difluoro-2-[(2-fluoro-4- iodopheeyl)amino]phenyl}carbonyl)azetidin-3-ol
[00343] To a solution of 4-iodo-2-(methylthio)pyrimidine (2.00 g, 7.92 mmol) in tetrahydrofuran (4.00 ml) was added isopropylmagnesium chloride (815 mg, 7.92 mmol). The mixture was allowed to stir for 1 h at 0 °C, followed by the addition of 1,1-dimethylethyl 3-oxoazetidiene-1 -carboxylate (1.64 g, 9.60 mmol), prepared using procedures similar to those described in Example 3. The reaction mixture was then allowed to warm to room temperature and stirred for 6h. The mixture was quenched with 1 N hydrochloric acid (10 mL) and extracted with ethyl acetate. The organic layer was separated, dried over anhydrous sodium sulfate, filtered and the filtrate concentrated in vacuo. The residue was purified by column chromatography (SiOj, hexanes/ethyl acetate) to afford 1,1-dimethylethyl 3-hydroxy-3-[2-(methylthio)pyrimidin-4-y 1]azetidine-1 -carboxylate (380 mg, 16%) as a yellow oil. 'HNMR (400 MHz, CDCI3): 8.62-8.59 (d, 1H), 7.36-7.33 (d, 1H), 5.14-5.11 (s, 1H), 4.29-4.24 (d, 2H), 4.13-4.08 (d, 2H), 2.61-2.58 (s, 3H), 1.50-1.47 (s, 9H); MS (El) for Cj3H,9N303S: 298 (Mff).
[00344] A solution of 1,1-dimethylethyl 3-hydroxy-3-[2-(methylthio)pyrimidin-4-yljazetidine-1 -carboxylate (480 mg, 1.62 mmol), and 3-chloroperoxybenzoic (558 mg, 3.23 mmol) acid in dichloromethane (25 mL) was stirred at room temperature for 22 h. The reaction mixture was quenched with a saturated solution of sodium thiosulfate and the pH adjusted to 7 with sodium carbonate. The organic layer was separated, dried over anhydrous sodium sulfate, filtered and the filtrate concentrated in vacuo. The resulting crude 1,1 -d imethy lethyl 3-hydroxy-3-[2- (methylsulfonyl)pyrimidin-4-yl]azetidine-1 -carboxylate (524 mg, 98%) was used without further purification. ]E NMR (400 MHz, CDC13): 9.01-8.97 (d, 1H), 7.96-7.93 (d, 1H), 4.57-4.53 (s, 1H), 4.31-4.27 (d, 2H), 4.23-4.18 (d, 2H), 3.42-3.39 (s, 3H), 1.50-1.47 (s, 9H); MS (El) for C13H19N3O5S: 330 (MH+).
[00345] A solution of 1,1-dimethy lethyl 3-hydroxy-3-[2- (methylsulfonyl)pyrimidin-4-yl]azetidine-l-carboxylate (215 mg, 0.652 mmol), and aqueous ammonia (7 mL, 28% solution) in dioxane (15 mL) within a sealed steel bomb cylinder was heated at 80°C for 4h. The mixture was cooled to room temperature and the solvent was evaporated. The residue was dissolved in dichloromethane and a solution of saturated sodium carbonate. The organic layer was separated, dried over anhydrous sodium sulfate, filtered and the filtrate concentrated in vacuo. The resulting crude 1,1-dimethylethy! 3-(2-aminopyrimidin-4-yl)-3-hydroxyazetidine-1 -carboxylate (140 mg, 100%) was used without further purification. !H NMR (400 MHz, CDC13): 8.38-8.35 (d, 1H), 6.97-6.94 (d, 1H), 5.30-5.28 (s, 2H), 4.23-4.18 (d, 2H), 4.08-4.04 (d, 2H), 1.48-1.45 (s, 9H).
[00346] To a solution of 1,1-dimethy lethyl 3-(2-aininopyrimidin-4-yl)-3-hydroxyazetidine-1 -carboxylate (140 mg, 0.524 mmol) in dichloromethane (10 ml) was added trifluoroacetic acid (3 ml). The reaction mixture was stirred for 2h at room temperature. The mixture was concentrated in vacuo. The resulting crude 3-(2-aminopyrimidin-4-y l)azetidin-3 -ol (87 mg, 100%) was used without further purification.
[00347] A solution of 3,4-difluoro-2-[(2~fluoro~4-iodophenyl)amino]benzoic acid (201 mg, 0.512 mmol), prepared using procedures similar to those described in US 7,019,033, 3-(2-aminopyrimidin-4-yi)azetidin-3-ol (87 mg, 0.52 mmol), benzotriazol-1 -yl-oxy-tris(pyrrolidino)phosphonium hexafluorophosphate (293 mg, 0.563 mmol) and N,A-diisopropylethy lamine (270 uL, 2.82 mmol) in N,/v-dimethylformamide (2 mL) was stirred at room temperature for 20h. The mixture was partitioned between ethyl acetate and saturated sodium bicarbonate. The organic layer was separated and washed with brine, dried over sodium sulfate, filtered and the filtrate concentrated in vacuo. The residue was purified by reverse phase HPLC to afford the title compound 3-(2-aminopyrimidin-4-yl)-l-({3,4~difluoro-2-[(2-fluoro~4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol (22 mg, 7%). 5H NMR (400 MHz, CD3OD): 8.23-8.20 (d, 1H), 7.48-7.43 (d, 1H), 7.35-7.32 (m, 2H), 7.09-7.00 (m, 1H), 6.88-6.84 (d, 1H), 6.70-6.63 (t, 1H), 4.59-4.54 (d, 1H), 4.45-4.40 (d, IH), 4.23-4.18 (d, IH), 3.04-3.99 (t, 1H); MS (El) for C20H15F3IN5O2: 542 (MH4).
[00348] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: EXAMPLE 8(a): 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3~pyridin-2-ylazetidin~3-ol: 'H NMR (400 MHz, CD3OD): 8.47 (m, 1H), 7.80 (m, IH), 7.65 (d, IH), 7.44 (m, 1H), 7.33 (m, 3H), 7.04 (m, 1H), 6.65 (m, 1H), 4.61 (d, 1H), 4.44 (d, 1H), 4.29 (d, 1H), 4.12 (d, IH). MS (El) for CjiH^INjC^: 526 (MH4). EXAMPLE 8(b): 1-((3,4-difluoro-2-[(2~fluoro-4- iodophenyr)amino]phenyl}carbonyl)-3-(l//-imidazol-2~yl)azetidin-3-ol: Ή NMR (400 MHz, CD3OD): 7.42 (m, 1H), 7.37 (m, 1H), 7.32 (m, 1H), 7.02 (m, 3H), 6.63 (m, IH), 4.65 (d, IB), 4.42 (d, 1H), 4.33 (d, 1H), 4.16 (d, 1H). MS (El) for Ci9Hi4F3IN402: 515 (MH4), EXAMPLE 8(e): 3-(1 //-benzimidazol-2-yl)-1-((3,4-difluoro-2~[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: 'H NMR (400 MHz, CD3OD): 7.55 (br s, 2H), 7.42 (m, 2H), 7.33 (m, IH), 7.23 (m, 2H), 7.04 (m, 1H), 6.65 (m, 1H), 4.76 (d, IH), 4.57 (d, IH), 4.43 (d, IH), 4.25 (d, IH). MS (El) for 023^3^4()2: 565 (MH+). EXAMPLE 8(d): 1-((3,4-difluoro-2-[(2-fluoro-4-
iodophenyl)amino]phenyl}carbonyl)-3-(5-methyl-l//-imidazol-2~yl)azetidin-3-ol: SH NMR (400 MHz, CD3OD): 7.41 (m, IH), 7.36 (m, IH), 7.31 (m, IH), 7.02 (m, IH), 6.67 (br s, IH), 6.63 (m, IH), 4.63 (d, IH), 4.39 (d, IH), 4.30 (d, IH), 4.13 (d, IH), 2.18 (s, 3H). MS (El) for C20H16F3IN4O2: 529 (MH4). EXAMPLE 8(e): 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-prop-2-en-l-ylazetidin-3-ol: 'H NMR (400 MHz, CDCI3): 8.47 (br s, IH), 7.40 (dd, IH), 7.35-7.31 (m, IH), 7.15-7.10 (m, IH), 6.81 (ddd, IH), 6.62 (ddd, IH), 5.84-5.72 (m, IH), 5.27-5.20 (m, 2H), 4.22-3.94 (m, 4H), 2.52 (d, 2H), 2.25 (s, IH); MS (El) for C19H16F3IN2O2: 489 (MH4). EXAMPLE 8(f): 3-[l-((3,4-difluoro-2~[(2-fluoro-4- iodophenyl)amino]pheny 1}carbonyl)-3-hydroxyazetidin-3-yljpropane-1,2-diol: 'H NMR (400 MHz, CDC13): 8.43 (br s, IH), 7.39 (dd, IH), 7.35-7.30 (m, IH), 7.16-7,10 (m, 1H), 6,82 (ddd, 1H), 6.61 (ddd, 1H), 4.31-3.91 (m, 5H), 3.68 (br d, 1H), 3.54.3.49 (m> 1H), 2.01-1.80 (m, 2H); MS (El) for C19H18F3IN2O4: 523 (MH+). EXAMPLE 8(g): l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-ethenylazetidin-3-ol: NMR (400 MHz, CDCI3): 8.48 (br s, III), 7.40 (dd, 1H), 7.35-7.31 (m, 1H), 7.17-7.11 (m, 1H), 6.81 (ddd, 1H), 6.62 (ddd, 1H), 6.15 (dd, 1H), 5.39 (d, 1H), 5.28 (d, 1H), 4.30-4.10 (m, 4H); MS (El) for C18H14F3IN2O2: 475 (MH+). EXAMPLE 8(h): 1 -[ 1 -({3,4~dirluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]ethane-l,2-diol hydrochloride: 5H NMR (400 MHz, d6-DMSG): 8.66 (d, 1H), 7,58 (dd, 1H), 7.38 (d, 1H), 7.33-7.27 (m, 1H), 7.17 (q, 1H), 6.74-6.65 (m, 1H), 4.50-3.58 (br, 3H), 4.29 (dd, 1H), 4.14 (dd, 1H), 3.87 (ΐ, 1H), 3.66 (t, 1H), 3.56-3.32 (m, 3H); MS (El) for C18H36F3IN2O4: 509 (MH+). EXAMPLE 8(i): l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-ethylazetidin-3-ol: lH NMR (400 MHz, CDCI3): 8.23 (br s, 1H), 7.40 (d, 1H), 7.33 (d, 1H), 7.15-7.10 (m, 1H), 6.85-6.79 (m, 1H), 6.64-6.58 (m, 1H), 4.14-3.94 (m, 4H), 1.78 (q, 2H), 0.96 (t, 3H): MS (El) for C18H16F3IN2O2: 477 (MH+). EXAMPLE 80): l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-methylazetidin-3-ol: SH NMR (400 MHz, CDCI3): 8.31 (br s, 1H), 7,40 (d, 1H), 7.33 (d, 1H), 7.15-7.11 (m, 1H), 6.85-6.78 (m, 1H), 6.65-6.59 (m, 1H), 4.24-4.04 (m, 4H), 1.55 (s, 3H); MS (El) for C17H14F3IN2O2: 463 (MH+). , EXAMPLE 8(k): 3-(2-aminopyrimidin~4-yl)-l-({3,4-difluoro~2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt: H NMR (400 MHz, CD3OD): 8.22-8.20 (d, 1H), 7.48-7.43 (d, 1H), 7.38-7.30 (m, 1H), 7.09-7.01 (q, 1H), 6.88-6.84 (d, 1H), 6.70-6.61 (t, 1H), 4.59-4.54 (d, 1H), 4.44-4.39 (d, III), 4.23-4.19 (d, 1H), 4.05-3.99 (d, 1H), 3.90-3.81 (d, 1H), 1.99-1.97 (s, 3H); MS (El) for C20H15F3I N5O2: 542 (MH+). EXAMPLE 8(m): l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-(l//-pyrrol-2-yl)azetidin-3-ol: H NMR (400 MHz, CD3OD): 7.37 (dd, 1H), 7.31-7,23 (m, 2H), 7.07-6.97 (m, 1H), 6.73-6.68 (m, 1H), 6.65-6.56 (m, 1H), 6.06-5.98 (m, 2H), 4.49-4.40 (m,lH), 4.32-4,18 (m, 2H), 4.15-88-4.07 (m, 1H). MS (El) for C20H15F3IN3O2: 514 (MH4) EXAMPLE 8(n): 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)araino]phenyl} carbonyl)-3-( 1 -methyl-1 //-imidazol-2-yl)azetidin-3-ol: 1H NMR (400 MHz, CD3OD): 7.34 (dd, 1H), 7.31-7.25 (m, 1H), 7.23-7.18 (m, 1H), 7.11-7.09 (m51H), 7.06-6.97 (m, 1H), 6.89-6.86 (m, 1H), 6.62-6.55 (m, 1H), 4.88-4.80 (m, 1H), 4.52-4.44 (m,lH), 4.38-4.30 (m, 1H), 4.21-4.12 (m, 1H), 3.68 (s, 3H). MS (El) for C20H16F3IN4O2: 529 (MH+). EXAMPLE 9 l~({3,4“difluoro-2”[(2-fluoro-4-iodophenyI)amino]phenyl}carbonyI)-3- (trifluoromethyl)azetidin-3-ol
[00349] 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino] phenyl }carbonyl)aze!idin-3-one (25 mg, 0.056 mmol), prepared using procedures described in Example 6, was taken into DMF (0.5 mL) followed by addition of (trifluoromethyl)trimethylsilane (40 pL, 0.28 mmol) and cesium carbonate (22 mg, 0.067mmol) and the mixture was stirred for one hour at room temperature. The mixture was partitioned with ethyl ether and water and the organic phase washed three times with additional water then brine and dried over anhydrous sodium sulfate. Filtration and concentration followed by silica gel flash chromatography of the residue using hexanes:ethyl acetate 3:2 as eluent afforded 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]pheny! }carbonyl)-3-(trifluoromethyl)azetidin-3-ol (19.8 mg, 69% yield) as a colorless crystalline solid. 'H-NMR (400 MHz, CDCI3): 8.31- 8.26 (br, 1H), 7.40 (d, 1H), 7.33 (d, 1H), 7.13-7.10 (m, 1H), 6.86-6.80 (m, 1H), 6.65- 6.60 (m, 1H), 4.42 (br s, 2H), 4.18 (br s, 2H). MS (El) for Ci7UuFdW2: 517 (ΜΗ*). EXAMPLE 10 1- ({3,4-difluQr0“2-[(2~fluoro-4-i0dophenyl)ammo]phenyl}carbonyl)a£etidin~3-one oxime
[00350] To a solution of 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]pheny 1} carbonyl)azetidin-3-one (100 mg, 0.22 mmol), prepared using procedures similar to those described in Example 6, in dioxane (1.0 mL) was added hydroxylamine (0.10 mL, 50% solution in water, 1.5 mmol), and the resulting solution was heated at 60 °C for 18 h. The mixture was cooled to room temperature and the crude product was purified by reverse phase HPLC to aiford l-({3,4-difluoro~ 2- [(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-one oxime (56 mg, 54% yield): {HNMR (400MHz, CDC13), 8.43 (br s), 7.43-7.39 (m, 2H), 7.35-7.32 (dd,lH), 7.19-7.15 (m, 1H), 6.87-6.81 (m,lH), 6.65-6.59 (m, 1H), 4.89 (br s, 2H), 4.85 (br s, 2H); MS (El) for C16HUF3IN3O2: 462 (ΜΗΓ).
Example 11 i¥-butyl-l-({3,4~difluoro-2-[(2~fluorO"4-iodopheny l)amino] phenyl) earbonyl)azetidm~3~amine
[00351] To a solution of l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl)carbonyl)azetidin-3-amine (0.09 M in acetonitrile, 500 pL, 0.045 mmol), prepared using procedures similar to those described in Example 2, was added triethyl amine (20 pL, 0.135 mmol) and n-butylhromide (6.14 pL, 0.054 mmol) followed by additional acetonitrile (1.0 mL). The reaction mixture was stirred at room temperature for 16 h, at which time it was purified directly by reverse phase HPLC to afford the title compound (8.4 mg). SH NMR (400 MHz, CDCI3): 8.50 (s, 1H), 7.39 (dd, 1H), 7.32 (dd, 1H), 7.13-7.09 (m, 1H), 6.84-6.77 (m, 1H), 6.63-6.57 (m, 1H), 4.35 (br s, 2H), 4.00 (br s, 1H), 3.87 (br s, 1H), 3.74-3.68 (m, 1H), 3.20 (br s, 3.5H), 2.56 (t, 2H), 2.03 (s, 2H), 1.50-1.42 (m, 2H), 1.39-1.29 (m5 2H), 0.91 (t, 3H). MS (El) for C20H21F3IN3O: 504 (MH+). EXAMPLE 12 l-({3,4-difluorO“2-[(2-fluoro-4-iodophe0yI)amino]phenyl}carbonyI)-iV- methylazetidin-3-amine
100352] To a solution of l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}earbonyl)azetidin-3-amine (0.10 M in acetonitrile, 1.0 mL, 0.09 mmol), prepared using procedures similar to those described in Example 2, in 1:1 ratio of methanol and tetrahydrofuran (2.0 mL) was added formaldehyde (37%wt, 6.7 μϋ, 0.09 mmol) followed by sodium cyanoborohydride (11.0 mg, 0.18 mmol). The reaction mixture was stirred at room temperature for 16 h, at which time it was quenched with saturated aqueous ammonium chloride. The solution was then purified directly by reverse phase HPLC to afford the title compound (14.9 mg). !H NMR (400 MHz, CDCI3): 8.13 (br s, 1H), 7.35 (d, 1H), 7.30 (d, 1H), 7.09-7.04 (m, 1H), 6.84-6.78 (m, 1H), 6.60-6.54 (m, 1H), 4.46-4.33 (br m, 4H), 3.93 (br m, 1H), 2.64 (s, 3H). MS (El) for Ci7Hi5F3IN30: 462 (MH+).
[00353] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: EXAMPLE 12(a). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-A-methylazetidin-3-amine: *H NMR (400 MHz, CDCI3): 8.13 (br s, 1H), 7.35 (d, 1H), 7.30 (d, 1H), 7.09-7.04 (m, 1H), 6.84-6.78 (m, 1H), 6.60-6.54 (m, 1H), 4.46-4.33 (br m, 4H), 3.93 (br m, 1H), 2.64 (s, 3H). MS (El) for C17H15F3IN3O: 462 (MHf). EXAMPLE 12(b). 2-{[l-({3,4-difluGro-2-[(2-fluoro-4- iodophenyr)amino]phenyl}carbonyl)azetidin-3-yI]amino}ethanol: !H NMR (400 MHz, CDC13): 8.20 (s, IH), 7.36 (d, 1H), 7.30 (d, 1H), 7.13-7.09 (m, IH), 6.85-6.79 (m, IH), 6.61-6.55 (m, 1H), 4.43 (br m, 3H), 3.98 (br m, 1H), 3.87 (br m, 1H), 3.02 (br m, 1H), 1.24-1.20 (m, 1H). MS (El) for €ι8Η17Ρ31Ν3θ2: 492 (ΜΗ*). EXAMPLE 12(c).,¥-[!-({ 3,4-difluoro-2-[(2-fluoro-4- iodopheny1)amino]phenyl}carbonyl)azetidin-3-yl]propane-l,3-diamine: !H NMR (400 MHz, CDCI3): 8.51 (s, 1H), 7.39 (d, 1H), 7.32 (d, 1H), 7.14-7.10 (m, 1H), 6.84- 6.77 (m, 1H), 6.63-6.57 (m, IH), 4.33 (br s, 2H), 3.99 (br s, 1H), 3.84 (br s, IH), 3.71-
3.64 (m, 1H), 2.91 (t, 2H), 2.70-2.66 (m, 2H), 2.01 (s, 4H), 1.76-1.69 (m, 2H). MS (El) for C19H20F3IN4O: 505 (MH+). EXAMPLE 12(d). l-({3,4-difluoro-2-[(2-fluoro~4~ iodophenyi)ammo]phenyl}earbony1)-A-ethyiazetidin-3~amine: NMR (400 MHz, CDCI3): 8.47 (s, 1H), 7.38 (d, 1H), 7.31 (d, IH), 7.13-7.09 (m, 1H), 6.83-6.77 (m, 1H), 6.62-6.57 (m, IH), 4.49 (br s, 3H), 4.36 (br s, 2H), 4.08 (br s, 1H), 3.94 (br s, 1H), 3.77-3.72 (m, IH), 2.69-2.63 (m, 2H), 1.99 (s, 2H), 1.14 (t, 3H). MS (El) for C18H17F3IN3O: 476 (MH+). EXAMPLE 12(e). l-({3,4-difluoro~2-[(2-fluoro-4-
iodophenyi)amino]phenyl}carbonyl)-iV-(2-meihylpropyl)azeiidin-3-aniine: lH NMR (400 MHz, CDC13): 8.50 (s, 1H), 7.38 (d, 1H), 7.31 (d, 1H), 7.14-7.09 (m, 1H), 6.83-6.76 (m, 1H), 6.63-6.57 (m, 1H), 4.34 (br s, 2H), 4.00 (br s, IH), 3.86 (br s, 1H), 3.71-3.66 (m, 1H), 3.42 (br s, 2H), 2.36 (d, 2H), 2.00 (s, 1H), 1.75-1.65 (m, 1H), 0.91 (d, 6H). MS (El) for C20H21F3IN3O: 504 (MH*). EXAMPLE 12(f). Ar-(cyclopropylmethyi)-l-({3,4-diiluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-amine: 3H NMR (400 MHz, CDCI3): 8.48 (s, 1H), 7.39 (d, 1H), 7.32 (d, 1H), 7.13-7.09 (m, IH), 6.84-6.77 (m, 1H), 6.63- 6.57 (m, 1H), 5.78 (s, 3H), 4.36 (br s, 2H), 4.10 (br s, 1H), 3.94 (br s, 1H), 3.81-3.75 (m, 1H), 2.49 (d, 2H), 2.01 (s, 4H), 0.94-0.86 (m, IH), 0.53 (d, 2H), 0.13 (d, 2H). MS (El) for C2oH19F3IN30: 502 (MH+). EXAMPLE 12(g). iV-(cyclohexylmethyl)-l~({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-amine: ;H NMR (400 MHz, CDCI3): 8.48 (s, IH), 7.38 (dd, IH), 7.31 (d, IH), 7.13-7.08 (m, IH), 6.83-6.77 (m, IH), 6.63- 6.57 (m, IH), 4.55 (br s, 2H), 4.33 (br m, 2H), 4.02 (br s, IH) 3.87 (br s, IH), 3.71- 3.65 (m, 1H), 2.38 (d, 2H), 1,74-1.68 (m, 4H), 1.46-1.36 (m, 1H), 1.27-1.12 (m, 3H), 0.94-0.84 (m, 2H). MS (El) for C23H25F3IN3O: 544 (MH*). EXAMPLE 12(h). A^cyclopentylmethyl)-1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-amine: !H NMR (400 MHz, CDCI3): 8.32 (s, 1H), 7.37 (d, 1H), 7.31 (d, 1H), 7.11-7.07 (m, 1H), 6.84-6.77 (m, 1H), 6.63- 6.57 (m, 1H), 4.44-4.37 (m, 3H), 4.02-3.96 (m, 1H), 2.84 (d, 2H), 2.54 (br s, 5H), 2.20-2.12 (m, 1H), 1.88-1.81 (m, 2H), 1.68-1.54 (m, 4H), 1.24-1.15 (m, 2H). MS (El) for C22H23F3rN30: 530 (MH+). EXAMPLE 13 l-({2,4-difluoro-6-[(2-fluoro-4-iodophenyI)amino]phenyl}carbonyl)azetidin-3- amine
[00354] 2,4,6-Trifluorobenzoic acid (643 mg, 3.65 mmol) and 2-lluoro-4-iodoaniline (1.0 g, 4.22 mmol) were taken into acetonitrile (30 mL) followed by addition of lithium amide (290 mg, 12.7 mmol) and the mixture was heated to 60 °C under a nitrogen atmosphere for one hour. On cooling to room temperature the mixture was added to 1 N aqueous hydrochloric acid (100 mL) and the precipitate formed was collected by filtration and washed once with water then hexanes and dried in vacuo to give 2,4-difluoro-6-[(2-fluoro-4-iodophenyI)amino]benzoic acid (849 mg, 59% yield) as a tan solid. ^-NMR (400 MHz, D6-DMSO): 13.72 (br s, 1H), 9.46 (s, 1H), 7.75 (d, 1H), 7.56 (d, 1H) 7.28 (tr, 1H), 6.73-6.67 (m, 1H), 6.53 (d, 1H).
[00355] 2,4-Difluoro~6-[(2-fluoro~4-iodophenyl)amino]benzoic acid (100 mg, 0.25 mmol) was taken into DMF (1 mL) followed by addition of PyBOP (137 mg, 0.26 mmol) and the mixture was stirred for 15 minutes then NMM (60 pL, 0.5 mmol) and commercially available 1,1-dimethylethyl azetidin-3-ylcarbamate (43 mg, 0.25 mmol) were subsequently added. The mixture was allowed to stir for 12 hours at room temperature then partitioned with ethyl acetate and water. The organic phase was washed three times with additional water then brine and dried over anhydrous sodium sulfate. Filtration and concentration followed by silica gel flash chromatography of the residue using hexanesrethyl acetate 3:1 as eluent afforded 1,1-dimethylethyl [1-({2,4-difluoro-6-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3- yl]carbamate (125 mg) as a colorless oil.
[00356] The oil was taken into trifluoroacetic acid (1 mL) and allowed to stand at room temperature for 5 minutes then concentrated in vacuo. The residue was portioned with ethyl acetate and saturated aqueous sodium bicarbonate and the organic phase washed with brine then dried over anhydrous sodium sulfate. The organic solution was filtered and concentrated then the residue taken into methanol (1 mL) followed by addition of 4 N HC1 in dioxane until the solution was acidic. The solution was concentrated and the residue triturated with ethyl ether to give a thick precipitate. The solid was collected by filtration and dried in vacuo to give 1-((2,4-difluoro-6-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-amine hydrochloride (58 mg, 48% overall yield). ’H-NMR (400 MHz, De-DMSO): 8.67 (br s, 3H), 8.45 (s, 1H), 7.71 (d, 1H), 7.54 (d, 1H), 7.25 (tr, 1H), 6.77 (tr, 1H), 6.48 (d, 1H), 4.28-4.23 (m, 2H), 4.13-4.06 (m, 3H). MS (El) for C^HoIWaO: 448 (MH+). EXAMPLE 14 1-((4,5-difluoro-2~[(2-fluoro-4-iodopheny!)ammo]phenyl}carbonyi)azetidm-3- amine
[00357] 2,4,5-Trifluorobenzoic acid (643 mg, 3.65 mmol) and 2-fluoro~4~ iodoaniline (1.0 g, 4.22 mmol) were taken into acetonitrile (30 mL) followed by addition of lithium amide (290 mg, 12.7 mmol) and the mixture was heated to 60 °C under a nitrogen atmosphere for one hour. On cooling to room temperature the mixture was added to 1 N aqueous hydrochloric acid (100 mL) and the precipitate formed was collected by filtration and washed once with water then hexanes and dried in vacuo to give 4,5-difluoro-2-[(2~fluoro-4~iodophenyl)amino]benzoic acid (624 mg, 43% yield) as a tan solid. 5H-NMR (400 MHz, D6-DMSO): 13.65 (br s, 1H), 9.63 (s, 1H), 7.84 (tr, 1H), 7.71 (d, 1H), 7.52 (d, 1H), 7.32 (tr, 1H), 7.03-6.98 (dd, 1H).
[00358] 4,5-difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoic acid (100 mg, 0.25 mmol) was taken into DMF (1 mL) followed by addition of PyBOP (137 mg, 0.26 mmol) and the mixture was stirred for 15 minutes then NMM (60 pL, 0.5 mmol) and commercially available 1,1-dimethylethyl azetidin-3-ylcarbamate (43 mg, 0.25 mmol) were subsequently added. The mixture was allowed to stir for 12 hours at room temperature then partitioned with ethyl acetate and water. The organic phase was washed three times with additional water then brine and dried over anhydrous sodium sulfate. Filtration and concentration followed by silica gel flash chromatography of the residue using hexanes:ethyl acetate 3:1 as eluent afforded 1,1-dimethylethyl [1-({4,5-difIuoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-yljcarbamate (131 mg) as a colorless oil. The oil was taken into trifluoroacetic acid (1 mL) and allowed to stand at room temperature for 5 minutes then concentrated in vacuo. The residue was portioned with ethyl acetate and saturated aqueous sodium bicarbonate and the organic phase washed with brine then dried over anhydrous sodium sulfate. The organic solution was filtered and concentrated then the residue taken into methanol (1 mL) followed by addition of 4 N HC1 in dioxane until the solution was acidic. The solution was concentrated and the residue triturated with ethyl ether to give a thick precipitate. The solid was collected by filtration and dried in vacuo to give 1 -({4,5 -difluoro-2- [(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-amine hydrochloride (67 mg, 55% overall yield). 'H-NMR (400 MHz, D6-DMSO): 9.02 (s, 1H), 8.54 (br s, 3H), 7.68 (dd, 1H), 7.53-7.47 (m, 2H), 7.22 (tr, 1H), 7.16 (dd, 1H), 4.60 (br s, 1H), 4.23 (br s, 2H), 4.03 (br m, 2H). MS (El) for C!6Ht3F3lN30: 448 (MH+). EXAMPLE 15 l-(P,4-Difluoro-2~((2-fluoro-4-iodophenyI)ammo]phenyl}carbonyl)-iY-(2,3- dihydroxypropyI)-3-hydroxyazetidme~3-carboxamide
[00359j l-(Diphenvlmethyl)azetidin-3-ol hydrochloride (2.75 g, 9.98 mmol), prepared using procedures similar to those described for Scheme 1 of the General Synthetic Section, 3A molecular sieves and 4-methylmorpholine (1.1 mL, 10.0 mmol) were suspended in dichloromethane (20 mL) at 0 °C. 4-Methylmorpholine N-oxide (2.93 g, 25.0 mmol) and tetrapropylammonium perruthenate (140 mg, 0.399 mmol) were added and the mixture was stirred at ambient for 24 h. The mixture was filtered through a plug of silica using 5% triethylamine in ethyl acetate as eluent. The filtrate was concentrated in vacuo and the residue was partitioned between ethyl acetate and saturated sodium bicarbonate solution. The organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 8:1 hexanes: ethyl acetate) gave 1-(dipheny!methyl)azetidin-3-one (871 mg, 3.68 mmol, 37% yield): 'H NMR (400 MHz, CDCI3): 7.50-7.46 (m, 4H), 7.33-7.27 (m, 4H), 7.27-7.19 (m, 2H), 4.59 (s, 1H), 4.01 (s, 4H); MS (El) for C16HiSNO: 238 (MH*). (00360] l-(Diphenylmethyl)azetidin-3-one (600 mg, 2.53 mmol), was dissolved in dichloromethane (1 mL) and treated with triethylamine (0.5 mL, 3.59 mmol) and trimethylsilylcyanide (0.8 mL, 6.01 mmol) at ambient for 2 h and then the mixture was concentrated in vacuo to afford l-(diphenylmethyl)-3~ [(trimethylsilyl)oxy]azetidine-3~carbonitrile (774 mg, 2.30 mmol, 91% yield) as a yellow solid. l-(dipheny!methyl)-3-[(trimethylsily!)oxy]azetidine-3-carbomtrile (250 mg, 0.744 mmol) was dissolved in dichloromethane (2 mL) at 0 °C and concentrated sulfuric acid (0.2 mL) was added dropwise. The mixture was stirred at ambient for 2 h and then was cooled to 0 °C and 25% ammonium hydroxide solution was added carefully dropwise to pH -10-11. The mixture was extracted twice with dichloromethane. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford a residue which was triturated with hexanes/ether to afford 1-(diphenylmethy 1)-3-hydroxyazetidine-3~carboxamide (160 mg, 0.567 mmol, 76% yield) as an off-white solid: !H NMR (400 MHz, CDC13): 7.92 (br s, 1H), 7.39-7.34 (m, 4H), 7.33-7.27 (m, 4H), 7.27-7.19 (m, 2H), 5.61 (br s, 1H), 4.45 (s, 1H), 4.34 (s, 1H), 3.50 (dd, 2H), 3.20 (dd, 2H); MS (El) for Ci7Hi8N202: 283 (MH+).
[00361] l-(Diphenylmethyl)-3-hydroxyazetidine-3-carboxamide (1.1 g, 3.90 mmol) was treated with 10% sodium hydroxide in ethanol (15 mL) and water (2 mL) at reflux for 2 h and then was concentrated in vacuo. The resiude was neutralized with 1 N hydrochloric acid (pH --7) and the precipitate was collected by filtration and lyophilized to afford l-(diphenylmethyl)-3-hydroxyazetidine~3-carboxylic acid (assume 3.90 mmol) which was used without further purification: ’H NMR (400 MHz, de-DMSO): 7.45-7.40 (m, 4H), 7.31-7.25 (m, 4H), 7.21-7.15 (in, 2H), 4.52 (s, 1H), 3.46 (dd, 2H), 3.02 (dd, 2H); MS (El) for Ci7H|7N03: 284 (MH+).
[00362] l-(Diphenylmethyl)-3~hydroxyazetidine~3~carboxylic acid (assume 3.90 mmol) was suspended in methanol (40 mL) and 4 N hydrochloric acid in dioxane (1 mL, 4 mmol) was added. 20 wt% Palladium hydroxide on carbon (100 mg) was added to the solution and the mixture was treated with hydrogen at 40 psi for 2 h. The mixture was filtered and the filtrate was concentrated in vacuo to afford 3-hydroxy azetidine-3 -carboxylic acid hydrochloride which was dissolved in tetrahydrofuran (5 mL) and water (5 mL) and treated with potassium carbonate (1.615 g, 11.7 mmol) and di-/er/-buty! dicarbonate (935 mg, 4.29 mmol) were added. The mixture was stirred at ambient for 17 h and then the mixture was partitioned between ethyl acetate and water. The aqueous portion was extracted with ethyl acetate and then was acidified to pH ~3-4 and extracted twice more with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 1-{[(1,1- dimethylethyl)oxy]carbony 1} -3 -hydroxyazetidine-3-carboxylic acid which was dissolved in DMF (3 mL). Benzotriazol-1 -yloxytris(pyrrolidino)phosphonium hexafluorophosphate (2.028 g, 3.90 mmol) and iV^V-diisopropylethylamine (0.7 mL, 4.03 mmol) were added. The mixture was stirred at ambient for 5 minutes and then allylamine (0.6 mL, 8.03 mmol) was added and the mixture was stirred for 17 h. The mixture was partitioned between ethyl acetate and 5% lithium chloride. The organic portion was washed with 20% citric acid, saturated sodium bicarbonate and brine, then was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, ethyl acetate) gave 1,1-dimethylethyl 3-hydroxy- 3- [(prop-2-en-1 -ylamino)carbonyl]azetidine-1 -earboxylate (782 mg, 3.05 mmol, 78% yield from 1 ~(diphenylmethyl)-3 -hydroxyazetidine-S-carboxamide). 1,1 -Dimethyl ethyl 3 -hydroxy-3-[(prop-2-en~ 1 -vlamino)carbony IJazetidine-1 - earboxylate (782 mg, 3.05 mmol) was dissolved in methanol (10 mL) and 4 N hydrochloric acid in dioxane (2 mL, 8 mmol) was added. The mixture was refluxed for 15 minutes and then was concentrated in vacuo to afford 3-hydroxy-jV-prop~2~en~ 1 -ylazetidine-3 -carboxamide hydrochloride (3.05 mmol).
[00363] 3,4-Difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoic acid (1.20 g, 3.05 mmol), prepared using procedures similar to those described in US 7,019,033, 4- (dimethylamino)pyridine (1.20 g, 9.86 mmol) and 1 -(3-dimethylaminopropyl)- 3- ethylcarbodiimide hydrochloride (701 mg, 3.66 mmol) were dissolved in DMF (10 mL). The mixture was stirred at ambient for 5 minutes and then 3-hydroxy-iV-prop-2-en-1 -ylazetidine-3-carboxamide hydrochloride (3.05 mmol) in DMF (5 mL) was added and the mixture was stirred for 15 h. The mixture was partitioned between ethyl acetate and 5% lithium chloride. The organic portion was washed with 20% citric acid, saturated sodium bicarbonate and brine, then was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 60-85% ethyl acetate in hexanes) and then reverse phase HPLC gave 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyi)amino]phenyl}carbony!)-3-hydroxy-JV-prop-2-en- 1 -ylazetidine-3-carboxamide (150 mg, 0.282 mmol, 9% yield): *H NMR (400 MHz, dg-DMSO): 8.64 (br s, 1H), 8.13 (t, 1H), 7.58 (dd, 1H), 7.38 (dd, 111), 7.34-7.28 (m, 1H), 7.21-7.12 (m, 1H), 6.84 (br s, 1H), 6.72 (ddd, 1H), 5.83-5.72 (m, 1H), 5.10-4.99 (m, 2H), 4.38 (d, 1H), 4.20 (d, 1H), 4.02 (d, 1H), 3.86 (d, 1H), 3.73-3.68 (m, 2H); MS (El) for C20H17F3IN3O3: 532 (MH+), [00364] 1-((3,4-Difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)-3 -hydroxy-iV-prop-2-en-1 -y lazetidine-3-carboxamide (88 mg, 0.166 mmol) and 4- methylmorpholine /v~oxide (58 mg, 0.496 mmol) were dissolved in acetone / water (4:1; 10 mL) and osmium tetroxide (2.5 wt.% in water; 0.1 mL) was added. The solution was stirred at ambient for 15 h, then was quenched with saturated sodium bisulfite (2 mL) and concentrated in vacuo. The residue was partitioned between ethyl acetate and brine. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Purification by reverse phase HPLC gave I-({3,4-difluoro-2~[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-.iV-(2,3- dihydroxypropyl)-3-hydroxyazetidine-3-carboxamide (68 mg, 0.120 mmol. 72% yield): ’H NMR (400 MHz, d6-DMSO): 8.65 (br s, 1H), 7.72 (t, 1H), 7.58 (dd, 1H), 7.41-7.36 (m, 1H), 7.34-7.28 (m, 1H), 7.21-7.12 (m, 1H), 6.92 (br s, 1H), 6.72 (ddd, 1H), 5.00-4.10 (br, 2H), 5.10-4.99 (m, 2H), 4.39 (d, 1H), 4.20 (d, 1H), 4.02 (d, 1H), 3.54-3.45 (m, 1H), 3.34-3.21 (m, 2H), 3.06-2.96 (m, 1H); MS (El) for C20H19F3IN3O5: 566 (MH+).
[00365] EXAMPLE 15(a). Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: 1-((3,4-Difluoro-2-[(2-fluoro-4-
iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidine-3~carboxamide: SH NMR (400 MHz, dg-DMSO): 8.63 (br s, 1H), 7.58 (dd, 1H), 7.42-7.36 (m, 3H), 7.34-7.28 (m, 1H), 7.22-7.12 (m, 1H), 6.76-6.68 (m, 2H), 4.39 (d, 1H), 4.19 (d, 1H), 4.00 (d, 1H), 3.83 (d, 1H); MS (El) for C17H13F3IN3O3:492 (MH4). EXAMPLE 16 6-{[3-(anainomethyl)-3-(methyloxy)azetidiii~l-yl]carbonyI}~2,3-difluoro-iV-(2- fluoro-4-iodophenyl)aniIine
[00366] Phenylmethyl l-oxa-5-azaspiro[2.3]hexane-5-carboxylate (165 mg, 0.75 mmol), prepared using procedures similar to those described in Reference 3, in THF (1 mL) was added to anhydrous ammonia saturated in THF (10 mL) and the mixture was allowed to stir in a sealed vessel at room temperature over 24 hours. The solution was then concentrated and taken back into THF (1 mL) followed by addition of di-Uzri-butyldicarbonate (164 mg, 0.75 mmol) and stirred for one hour at room temperature. The mixture was then concentrated and the residue purified by silica gel flash chromatography using hexanes:ethyl acetate (1:1) as eluent to give phenylmethyl 3 - [({[(1,1 -dimethylethyl)oxy] carbonyl} amino)methyl]-3 -hydroxyazetidine-1 -carboxylate (16.5 mg, 7% yield) and unreacted epoxide (120 mg, 73% recovery). ^-NMR (400 MHz, CDCb): 7.34 (m, 5H), 5.10 (br, 1H), 5.09 (s, 2H), 4.68 (s, 1H), 3.90 (dd AB, 4H), 3.41 (d, 2H), 1.44 (s, 9H).
[00367] Phenylmethyl 3-[({ [(1, l-dimethylethyl)oxy]carbonyl}ammo)methyl]-3-hydroxyazetidine-1 -carboxylate (16.5 mg, 0.05 mmol) and 10% Pd/C (8 mg) were taken into methanol (2 mL) and hydrogenated at ambient pressure over 12 hours. The catalyst was removed by filtration and the filtrate concentrated and dried in vacuo.
The residue was taken into THF (1 mL) followed by addition of DIPEA (10 pL, 0.06 mmol) and 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoyl fluoride (19.8 mg, 0.05 mmol), prepared using procedures similar to those described in Reference 1, and the solution was stirred at room temperature for 30 minutes. Concentration and purification of the residue by silica gel flash chromatography using hexanes.ethyl acetate (1:1.5) afforded 1,1-dimethylethyl {[1-((3,4-difluoro-2[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidine-3-yljmethy 1}carbamate (19 mg, 66% yield).
[00368] 1,1-Dimethylethyl {[1-((3,4-difluoro-2[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidine-3-yl]methyl}carbamate (8.0 mg, 0.014 mmol) and silver (I) oxide (12 mg, 0.05 mmol) were taken into methyl iodide (0.5 mL) and the mixture was brought to reflux for 4 hours. The suspension was then cooled to room temperature and diluted with an excess of ethyl ether then filtered. The filtrate was concentrated and purified by silica gel flash chromatography using hexanes:ethyl acetate (1:1) as eluent to give 1,1-dimethylethyl {[1-((3,4-difluoro-2 [(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)-3 -(methyloxy)azetidine-3 -y 1] me thy 1} carbamate (2 mg). The material was taken into TEA (0.5 mL) and allowed to stand for 5 minutes then concentrated in vacuo. The residue was azetroped twice from methanol (2 mL) and the residue dried in vacuo to afford 6-{[3-(aminomethyl)-3-(methyloxy)azetidin-1 -yl] carbonyl} -2,3 -difluoro-A-(2-f!uoro-4-iodophenyl)aniline trifluoroacetate salt (2.3 mg, 27% yield) as an amorphous solid. MS (El) for CigHnFalNaO: 492 (MFT). EXAMPLE 17 l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyI)araino]phenyl}carbonyl)-3-{2-[(l- methylethyl)amino]ethyI}azetidin-3-ol
[00369] A solution of /erf-butyl acetate (566 pL, 4.2 mmol) in THF (10 mL) was cooled to -78 °C. To the solution was added LHMDS (5.25 mL of a 1.0 M solution in hexanes, 5.25 mmol), and the resulting mixture was stirred for 20 min at -78 °C. To the solution was added 1 -(diphenylmethyl)azetidin-3-one (500 mg, 2.1 mmol), prepared using procedures similar to those described in Example 15. After stirring for 1 h, saturated aqueous ammonium chloride was added, and the mixture was warmed to rt. Water and ether were added, and the resulting biphasic mixture was partitioned. The aqueous phase was extracted once with ether. The combined organic extracts were dried over magnesium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash chromatography (80% hexanes: 20% ethyl acetate) to provide 1,1-dimethylethyl [ 1 -(diphenyImethy 1)-3 -hydroxyazetidin-3 -yl] acetate as a pale yellow solid (644 mg, 1.8 mmol, 87% yield). *H NMR (400 MHz, CDCI3): δ 7.40 (m, 4H), 7.26 (m, 4H), 7.19 (m, 2H), 4.40 (s, 1H), 4.02 (s, 1H), 3.15 (m, 2H), 3.05 (m, 2H), 2.83 (s, 2H), 1.45 (s, 9H).
[00370] To a solution of 1,1-dimethylethyl [l-(diphenylmethyl)-3-hydroxyazetidin-3-yl]acetate (333 mg, 0.94 mmol) in THF (3 mL) at 0 °C was added lithium aluminum hydride (940 pL of a 1.0 M solution in THF, 0.94 mmol). The mixture was stirred for 3 h 20 min while warming to rt. Water (36 pL) was added carefully to the solution, followed by 15% sodium hydroxide (36 pL) and more water (108 pL). The resulting precipitate was removed by filtration through celite, and the filtrate was concentrated to dryness yielding l-(diphenylmethyl)-3-(2-hydroxyethyl)azetidin-3-ol (228 mg, 0.80 mmol, 85% yield) as a colorless syrup. 'H NMR (400 MHz, CDCI3): δ 7.38 (m, 4H), 7.26 (m, 4H), 7.19 (m, 2H), 4.37 (s, 1H), 3.92 (m, 2H), 3.32 (m, 2H), 2.96 (m, 2H), 2.07 (m, 2H).
[00371] Palladium hydroxide (100 mg) was suspended in a solution of l-(diphenylmethyl)-3-(2-hydroxyethy!)azetidin-3-ol (228 mg, 0.80 mmol) in methanol (15 mL), and the mixture was subjected to an atmosphere of hydrogen at 50 psi for 4 h. The catalyst was then removed by filtration through celite, and the filtrate was concentrated in vacuo to provide 3-(2-hydroxyethyi)azetidin-3-ol. This material was used in the subsequent reaction without purification. To a solution of 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoic acid (314 mg, 0.80 mmol), prepared using procedures similar to those described in US 7,019,033, in DMF (4 mL) was added PyBOP (416 mg, 0.80 mmol) and triethylamine (223 pL, 1.6 mmol). Finally, the unpurified 3-(2-hydroxyethyl)azetidin~3~oi was added, and the resulting mixture was stirred at rt for 16 h. Water and ethyl acetate were added, and the layers were separated. The aqueous phase was extracted with once more with ethyl acetate. The combined organic extracts were washed with brine, dried over magnesium sulfate, filtered, and concentrated in vacuo. The residue was purified by flash chromatography, eluting with ethyl acetate, to provide l-({3,4-difluoro~2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyi)-3-(2-hydroxyethyl)azetidin-3-ol as a colorless oil (303 mg, 0.62 mmol, 78% yield). *H NMR (400 MHz, CDC13): 6 8.46 (s, 1H), 7.39 (dd, 1H), 7.32 (m, 1H), 7.13 (m, 1H), 6.81 (m, 1H), 6.60 (m, 1H), 4.37 (br s, 1H), 4.28 (br m, 4H), 3.94 (br s, 2H), 2.19 (br s, 1H), 2.02 (m, 2H); MS (El) for €ΐ8Η,6Ρ3ΪΝ203: 491 (ΜΗ’).
[00372] A solution of oxalyl chloride (13 pL, 0.15 mmol) in dichloromethane (1 mL) was cooled to -78 °C, and DMSO (22 pL, 0.31 mmol) was then added. To this mixture was added l-({3,4-difluoro-2-[(2~fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3~(2-hydroxyethy!)azetidin-3-ol (67.8 mg, 0.14 mmol) as a suspension in dichloromethane (1 mL). After stirring at -78 °C for 10 min, triethylamine (78 pL, 0.56 mmol) was added and the mixture was allowed to warm to rt. The solution was diluted with dichloromethane, and washed with 0.5 N HC1. The aqueous phase wash then extracted with dichloromethane. The organic extracts were combined, dried over magnesium sulfate, filtered, and concentrated. The residue was purified by flash chromatography to provide [1-((3,4-difluoro-2 - [(2 -fluoro-4-iodophenyl)amino]phenyl} carbonyl)-3-hydroxyazetidin-3 -yljacetaldehyde as a white solid (22.1 mg, 0.045 mmol, 32% yield). *H NMR (400 MHz, CDC13): δ 9.82 (s, 1H), 8.46 (s, 1H), 7.39 (m, 1H), 7.33 (m, 1H), 7.11 (m, 1H), 6.81 (m, 1H), 6.61 (m, 1H), 4.32-3.96 (br m, 4H), 3.41 (t, 2H), 3.07 (s, 1H); MS (El) for C18H14F3IN2O3: 491 (MH+).
[00373] To a solution of [ 1 -({3,4-difluoro~2-[(2~fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]acetaldehyde (38.0 mg, 0.078 mmol) in 1,2-dichloroethane (1 mL) was added isopropylamine (27 pL, 0.31 mmol) followed by sodium iriaceioxyborohydride (26 mg, 0.12 mmol). The mixture was stirred for 3 h before quenching with 1 drop of concentrated HC1. The quenched mixture was concentrated to dryness, and then purified by preparative HPLC to provide l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-{2- [(1 -methylsthyl)amino]ethyl}azetidin-3-ο 1 (21.5 mg) as a pale yellow solid. *Η NMR (400 MHz, d6-DMSG): δ 8.54 (s, 1H), 7.57 (dd, 1H), 7.38 (dd, 1H), 7.31 (m, 1H), 7.17 (m, 1H), 6.67 (m, 1H), 4.02 (m, 1H), 3.89 (m, 2H), 3.71 (m, 1H), 2.70 (m, 1H), 2.63 (m, 2H), 1.86 (s, 3H), 1.75 (m, 2H), 0.97 (d, 6H); MS (El) for C21H23F3IN3O2: 534 (MET). EXAMPLE 18 l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]pheny!}carbonyl)-3-{l,l- dimethyl“2-[(l-methylethyI)amino]ethyl}azetidin-3-ol
[00374] To a solution of l-({3,4~difluoro-2-[(2~fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-one (500 mg, 1.12 mmol), prepared using procedures similar to those described in Example 6, in dichloromethane (5 mL) cooled to 0 °C was added titanium tetrachloride (125 pL, 1.12 mmol). The dark brown solution was stirred at 0 °C for 45 minutes, followed by the addition of methyltrimethylsilyl dimethylketene acetal (550 pL, 2.24 mmol) at 0 °C. Upon addition the solution was allowed to warm to room temperature, and was stirred for 1 hour. The reaction mixture was then partitioned between saturated aqueous sodium bicarbonate and ethyl acetate. The aqueous portion was extracted twice using ethyl acetate. The combined organic portion was washed with water, brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a brown oil which was purified by column chromatography. Eluting with 10% diethyl ether in dichloromethane, the isolated product was concentrated in vacuo to afford 520 mg, 0.95 mmol (85%) of methyl 2-[ 1 -({3,4-difIuoro-2~ [(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-y!]-2-methylpropanoate as a white foam. 'H NMR (400 MHz, CDC13): 8.34 (s, 1H), 7.38 (d, 1H), 7.31 (d, 1H), 7.13- 7.08 (m, 1H), 6.85-6.77 (m, 1H), 6.63-6.56 (m, 1H), 4.26-4.20 (m, 2H), 4.13- 4.09 (m, 1H), 4.00-3.93 (m, 1H), 3.70 (s, 3H), 1.23 (s, 6H). MS (El) for C21H20F3IN2O4: 547 (MH').
[00375] A solution of methyl 2-[ 1 -((3,4-difluoro-2-[(2-fluoro-4~iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yS]-2-methylpropanoate (520 mg, 0.95 mmol) in 4N aqueous potassium hydroxide (5 mL) was stirred at 50°C for 1 hour. Using concentrated aqueous hydrochloric acid, the reaction mixture was acidified to pH 5, and then partitioned with ethyl acetate. The aqueous portion was extracted twice using ethyl acetate, and the combined organic portion was washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford 300 mg, 0.56 mmol (59%) of 2-[ 1 -({3,4-difluoro-2-[(2-fluoro- 4-iodophenyl)amino]phenyl}earbonyl)-3-hydroxyazetidin-3-yl]-2-methylpropanoic acid as a white solid. 'H NMR (400 MHz, DMSO): 8.49 (s, 1H), 7.57-7.52 (m, 1H), 7.37-7.25 (m, 2H), 7.17-7.13 (m, 1H), 6.68-6.58 (m, 1H), 3.98-3.94 (m, 2H), 3.80- 3.77 (m, 1H), 3.55-3.52 (m,TH), 0.88 (s, 6H). MS (El) for C2oHisF3IN204: 535 (MH*).
[00376] To solution of 2-[l-({3,4-difluoro-2-[(2-fluoro- 4-iodophenyl)amino]phenyl} carbony 1)-3 -hydroxyazetidin-3 -yl] -2-methylpropanoic acid (300 mg, 0.56 mmol) in tetrahydrofuran (5 mL) was added triethvlamine (80 pL, 0.56 mmol), followed by PyBOP (295 mg, 0.56 mmol) and finally sodium borohydride (64 mg, 1.68 mmol). The mixture was stirred at room temperature for 1 hour. The reaction mixture was quenched by adding 20% aqueous citric acid, and then partitioned with ethyl acetate. The organic portion was washed with saturated aqueous sodium bicarbonate, brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a white solid which was purified by column chromatography. Eluting with 60% ethyl acetate in hexanes, the isolated product was concentrated in vacuo to afford 238 mg, 0.46 mmol (82%) of l-({3,4-difluoro-2-[(2- fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-(2-hydroxy-1,1-dimethyleihyl)azetidin-3-ol as a white solid. NMR (400 MHz, DMSO): 8.53 (s, 1H), 7.57 (d, III), 7.38-7.28 (m, 2H), 7.22-7.15 (m, 1H), 6.70-6.64 (m, 1H), 5.61 (s, 1H), 4.57 (br s5 1H), 4.30-4.27 (m, 1H), 4.18-4.15 (m, 1H), 3.80-3.77 (m, 1H), 3.68- 3.64 (m, 1H), 3.25 (s, 2H), 0.76 (d, 6H); MS (El) for C20H20F3IN2O3: 521 (MH+).
[00377] A mixture of l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-(2-hydroxy-1,1 -dimethyleihyl)azetidin-3-ol (200 mg. 0.38 mmol) and Dess-Martin periodinane (240 mg, 0.57 mmol) in dichloromethane (2 mL) was stirred at room temperature for 2 hours. 10% aqueous sodium thiosulfate (2 mL), and saturated aqueous sodium bicarbonate (2 mL) was added and the mixture was stirred at room temperature for 15 minute. The mixture was partitioned and the aqueous layer was extracted twice using dichloromethane.
The combined organic portion was washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo, to afford a white solid which was purified by column chromatography. Eluting with 30% ethyl acetate in hexanes, the isolated product was concentrated in vacuo to afford 100 mg, 0.20 mmol (53%) of 2-[l-({3,4~ difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]- 2-methylpropanal as a white solid, which was immediately dissolved in tetrahydrofuran (2 mL). To the solution was added isopropylamine (34 μΙ_, 0.40 mmol), followed by triacetoxyborohydride (212 mg, 1.0 mmol). The solution was stirred at room temperature for 15 hours. The reaction mixture was concentrated in vacuo and partitioned between 20% aqueous citric acid and ethyl acetate. The aqueous portion was extracted twice using ethyl acetate, and the combined organic portion was washed with saturated aqueous sodium bicarbonate, brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a yellow oil which was purified by preparative reverse phase HPLC. The isolated product was concentrated in vacuo to afford 50 mg, 0.07 mmol (36%) of l -({3,4-difluoro-2-[(2-fluoro-4-iodopheny!)amino]phenyl}carbonyl)-3-{l,l-dimethyl-2-[(l-methylethyl)amino]ethyl}azetidin-3-ol acetate salt as a white solid. *H NMR (400 MHz, DMSO): 8.47 (br s, 1H), 7.55 (d, 1H), 7.36-7.29 (m, 2H), 7.22-7.15 (m, 1H), 6.68-6.63 (m, 1H), 4.17-4.08 (m, 2H), 3.76-3.73 (m, 1H), 3.56-3.52 (m, 1H), 2.58- 2.51 (m, 1H), 2.45-2.37 (m, 2H), 0.92 (t, 6H), 0.78 (d, 6H); MS (El) for C23H27F3IN3O2: 562 (MH+). EXAMPLE 19 1-((3,4-difIuoro-2-[(2-fluoro-4-iodopheny{)amino]pheiiyl}carbonyl)~3-{[(l- methylethyl)ami0o]methyl}azefidisi"3~amme
[00378] To a solution of the 1 -(diphenylmethyl)-3-[(phenylmethyl)amino]azetidine-3-carhonitrile (0.80 g, 2.2 mmol), prepared using procedures similar to those described in Kozikowski and Fauq Synlett 1991, 11, 783-4, in ethanol (30 mL) was added solid sodium hydroxide (7.5 mmol), and the resulting mixture was stirred at room temperature for 3 days. Water (6 mL) was added to the reaction mixture and stirring was continued at 90 °C for 2 h. The pH of the reaction mixture was adjusted to 5 with concentrated hydrochloric acid and a white solid precipitated. The mixture was cooled , diluted with water (50 mL) and the solid was collected, washed with water then dried in vacuo to give the 1 -(diphenylmethy 1)-3-[(phenylmethyl)amino]azetidine-3-carboxylie acid (0.75g, 88% yield), MS (HI) for C24H24N2O2: 373 (MH+).
[00379] To a mixture of l-(diphenylmethyl)-3-[(phenylmethyl)amino]azetidine-3-carboxylic acid (0.50 g, 1.34 mmol), Α'.Λ-diisopropyIethylamine (0.47 mL, 2.68 mmol) in DMF (3 mL) was added 1-benzotriazolyloxytripyrrolidinylphosphonium hexafluorophosphate (1.34g, 2.68 mol) and the resulting mixture was stirred at room temperature for 10 minutes. To this mixture was added 2-propylamine (0.22 mL, 2.68 mmol) and stirring was continued for 18 h. The reaction mixture was diluted with ethyl acetate (100 mL) and washed with 2% aqueous citric acid, 5% lithium chloride, and brine solutions (50 mL each), dried over sodium sulfate, filtered and concentrated to give an oily residue which was purified by flash chromatography (silica gel, eluting with 15-25% ethyl acetate-hexane) to give l-(diphenylmethyl)-iV-(l-methylethyl)- 3-[(phenylmethyl)amino]azetidine-3-carboxamide (0.51 g, 92% yield), MS (El) for C27H31N3O: 414 (MH*).
[00380] To a solution of the l-(diphenylmethyl)-iV-{ 1 -methylethyl)-3-[(phenylmethyl)amino]azetidine-3-carboxamide (0.40 g, 0.97 mmol) in tetrahydrofuran (10 ruL) at room was added a solution of lithium aluminum hydride in tetrahvdrofuran (1M, 2.90 mL, 2.90 mmol), and the resulting mixture was stirred at 50 °C for 3h. The reaction mixture was cooled to room temperature, quenched with 20% aqueous hydroxide solution (1 mL), diluted with ether (50 mL) and filtered. The filtrate was washed with brine solution (20 mL each), dried over sodium sulfate, filtered and concentrated to give an oily residue which was purified by flash chromatography (silica gel, eluting with 5% methanol-dichloromethane) to give 1 -(diphenylmethyl)-3- {[(1 -methylethyl)amino]methyl} -iV-(phenylmethyl)azetidin-3-amine (0.35g, 90% yield), !H NMR (400 MHz, CDC13): 7.42-7.14 (m, 15H), 4.34 (s, 1H), 3.66 (s, 2H), 3.22-3.18 (d, 2H), 2.97 (s, 2H), 2.90-2.86(d, 2H), 2.68-2.62 (p, 1H), 1.09-1.07 (d, 6H); MS (El) for C27H33N3:400 (MH+).
[00381] To a solution of the l-(diphenylmethyl)-3-{[(l-methylethyl)amino]methyl}-jV-(phenylmethyl)azetidin-3-amine (0.35 g , 0.88 mmol) in methanol was added a solution of hydrogen chloride in dioxane (4 molar solution, 0.96 mL, 4.40 mmol) and the resulting mixture was concentrated to give a white solid which was taken back into methanol. To this solution were added palladium hydroxide (20% on carbon, 0.50 g, 0.19 mmol) and the resulting mixture shaken at 50 psi in a Parr apparatus for 3h. The reaction mixture was filtered and concentrated to give a solid, which was washed with ether and dried in vacuo to give 3 - {[(1 -methy lethy l)amino] methyl} azetidin- 3 - amine hydrochloride as a white solid (0.18 g, 81% yield). MS (El) for C7H!7N3: 144 (MH+), [00382] To a mixture of the 3-{[(l~methylethyl)amino]methyl}azeiidin-3-amine hydrochloride (20 mg, 0.079 mmol) in saturated sodium bicarbonate solution (1.0 mL) and dioxane (1.0 mL) was added 3,4-difluoro-2-[(2~fluoro-4-iodophenyi)amino]benzoy! fluoride (31 mg, 0.079 mmol), prepared using procedures similar to those described in Reference 1, and the resulting mixture was stirred at room temperature for 18 h. The reaction mixture was diluted with water (5 mL) and extracted with ethyl acetate (3x5 mL). The combined extract was washed with water then brine solution (5 mL each), dried over sodium sulfate, filtered and concentrated to give an oily residue which was purified by reverse phase HPLC to afford 1-((3,4-difluoro-2-[(2-fluoro-4~iodophenyl)amino]phenyl}carbonyl)-3-{[(l-methylethyl)amino]methyl}azetidin-3-amine (15 mg, 37% yield). 'H NMR (400 MHz, dU-Methanol): 7.46-7.43 (dd, 1H), 7.35-7.33 (dd, 1H), 7.31-7.27 (m, 1H), 7.08- 7.01 (dd, 1H), 6.63, 6.58 (td, 1H), 4.09-4.07 (d, 1H), 3.91-3.85 (dd, 2H), 3.76-3.73 (d, 1H). 2.80-2.74 (m, 1H), 2.73 (s, 2H), 1.07-1.05 (d, 6H); MS (El) for C20H22F3IN4O: 519 (MH4). EXAMPLE 20 3-(l-amino-2-methylpropyl)-l-({3,4-difluoro-2-[(2-f1uoro-4- iodophenyl)ammo]phenyl}carbonyi)azetidin-3-ol
[00383] 1,1-Dimethylethyl 3-oxoazetidine-1 -carboxylate (677.2 mg, 3.96 mmol), prepared using procedures similar to those described in Example 3, was taken into 2-methyl-1 -ntropropane (5 mL) then cooled to 0 °C followed by addition of potassium iert-butoxide (444 mg, 3.96 mmol) and the resulting mixture was allowed to warm to room temperature over 30 minutes. The mixture was partitioned with ethyl acetate and 0.5 N aqueous hydrochloric acid then once with water and brine then dried over anhydrous magnesium sulfate. Filtration and concentration afforded a residue (1.5 g) that was further purified by silica gel flash chromatography using 3:1 hexanes:ethyl acetate as eluent to give 1,1 -dimethylethyl 3 -hydroxy-3-(2-methyl-1 -nitropropyl)azetidine-1 -carboxylate (730 mg, 67% yield) as a colorless crystalline solid. 'H-NMR (400 MHz, CDC13): 4.50 (d, 1H), 3.93 (dd AB, 2H), 3.85 (s, 2H), 3.58 (s, 1H), 2.54-2.48 (m, 1H), 1.44 (s, 9H), 1.04 (d, 6H).
[00384] 1,1 -Dimethylethyl 3-hydroxy-3-(2-methyl-1 -nitropropyl)azetidine-1 - carboxylate (105 mg, 0.38 mmol) was taken into methanol (1 ml.) followed by addition of 4 N anhydrous hydrogen chloride in dioxane (1 mL) and the acidic solution was allowed to stand for 15 minutes at room temperature then concentrated and dried in vacuo to an amorphous residue. 3,4-Difluoro-2-[(2-fluoro~4-iodophenyl)amino]benzoic acid (150 mg, 0.38 mmol), prepared using procedures similar to those described in US 7,019,033, was taken into DMF (0.7 mL) followed by addition of PyBOP (198 mg, 0.38 mmol) and the solution was allowed to stir for 10 minutes at room temperature. The above amine hydrochloride salt and DIPEA (190 μΤ, 1.1 mmol) in DMF solution (0.7 mL) was added and the mixture was allowed to stir for one hour at room temperature. The mixture was partitioned with ethyl acetate and 0.5 N aqueous hydrochloric acid and the organic phase washed three times with water then brine and dried over anhydrous magnesium sulfate. Filtration and concentration afforded a residue that was further purified by silica gel flash chromatography using 1,5:1 hexanesrethyl acetate as eluent to give l-({3,4~difiuoro- 2-[(2~fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-(2-methyl-l-· nitropropy!)azetidin-3-ol (189 mg, 90% yield) as an amorphous solid. ^-NMR (400 MHz, CDC13): 8.41 (br s, 1H), 7.41 (dd, 1H), 7.34 (d, 1H), 7.09 (br m, 1H), 6.81 (q, 1H), 6.65-6.60 (m, 1H), 4.49 (d, 1H), 4.15-4.09 (m, 4H), 3.66 (s, 1H), 2.56-2.46 (m, 1H) 1.03 (d, 6H).
[00385] l-({3,4-Difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-(2-methyl-1 -nitropropyl)azetidin-3-ol (189 mg, 0.34 mmol) was taken into 4:1 THF.water (5 mL) followed by addition of iron powder (192 mg, 3.4 mmol) and ammonium formate (429 mg, 6.8 mmol) and the mixture was heated to reflux. After four hours additional aliquots of iron powder (192 mg, 3.4 mmol) and ammonium formate (429 mg, 6.8 mmol) were added and the mixture was allowed to reflux an additional 12 hours. The mixture was cooled to room temperature and diluted with ethyl acetate then filtered. The filtrate was partitioned with ethyl acetate and saturated aqueous sodium bicarbonate then the organic layer washed with brine and dried over anhydrous sodium sulfate. Filtration and concentration afforded a residue that was further purified by silica gel flash chromatography using ethyl acetate to 10% methanol in dichloromethane as eluents to give a residue (36.5 mg) that was further purified by preparative reverse phase HPLC to give 3-(1 -amino-2-methylpropyl)-1 -({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol trifluoroacetate salt (7.9 mg) as a colorless amorphous solid after lyophillization of the combined pure fractions. 'H-NMR (400 MHz, D<$-DMSO): 8.63 (s, 1H), 7.58 (dd, 1H), 7.37 (d, 1H), 7.35-7.31 (m, 1H), 7.17 (q, 1H), 6.71-6.66 (m, 1H), 4.23 (dd, 1H), 4.03 (dd, 1H), 3.80 (dd, 1H), 3.66 (dd, 1H), 2.34 (dd, 1H), 1.79-1.70 (m, 1H), 0.84-0.77 (m, 6H). MS (El) for C20H21F3IN3O2: 520 (MH+).
[00386] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: EXAMPLE 20(a). 3-(1 -aminoethy 1)-1-({3,4~difluoro-2-[(2~fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: !H NMR (400 MHz, de-DMSQ): 8.56 (s, 1H), 7.91 (br s, 2H), 7.58 (d, 1H), 7.39 (d, 1H), 7.36-7.32 (m, 1H), 7.24-7.17 (m, 1H), 6.72-6.65 (m, 2H), 4.33-4.29 (m, 1H), 4.23-4.19 (m, 1H), 4.16-4.14 (m, 1H), 4.07-3.94 (m, 1H), 3.82-3.77 (m, 1H), 3.51-3.45 (m, 1H), 1.15-1.12 (m, III), 1.ΙΟΙ .08 (m, 1H). MS (El) for C18H17F3IN3O2: 492 (Μ1Γ). EXAMPLE 20(b). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyi}carbonyl)-3-[l-(ethylamino)ethyl]azetidin-3-oS: 'H NMR (400 MHz, dg-DMSO): 8.61 (d, 1H), 8.50 (s, 1H), 8.20 (s, 1H), 7.59 (d, 1H), 7.39 (d, 1H), 7.36-7.32 (m, 1H), 7.24-7.17 (m, 1H), 6.82 (s, 1H), 6.74-6.67 (m, 1H), 4.38 (d, 1H), 4.27 (d, 1H), 4.18 (d, 1H), 4.06 (d, 2H), 3.99 (d, 1H), 3.89 (d, 1H), 3.82 (d, 1H), 3.49-3.43 (m, 1H), 3.04-2.80 (m, 4H), 1.21-1.12 (m, 6H). MS (El) for C20H21F3IN3O2: 520 (MH+). EXAMPLE 20(c), 1-((3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-(1 -nitroethyl)azetidin-3-ol: !H NMR (400 MHz, d6-DMSO): 8.57 (d, 1H), 7.58 (d, 1H), 7.38 (d, 1H), 7.37-7.33 (m, 1H), 7.22- 7.17 (m, 1H), 6.73-6.66 (m, 1H), 6.57 (s, 1H), 5.06-4.97 (m, 1H), 4.54 (d, 0.5H), 4.37 (d, 0.5 H), 4.29 (d, 0.5H), 4.14 (d, 0.5 H), 4.05 (d, 0.5 H), 3.95 (d, 0.5H), 3.86 (d, 0.5H), 3.80 (d, 0.5H), 1.44-1.38 (m, 3H). MS (El) for 523 (MH+). EXAMPLE 20(d). 1-((3,4-difluoro-2-[(2-fluoro~4- iodophenyl)amino]phenyl}carbonyl)-3-[l-(methylamino)ethyi]azetidin-3-ol; !H NMR (400 MHz, d6-DMSO): 8.63-8.55 (m, 1H), 8.44-8.23 (m, 1H), 7.79 (br s, 1H), 7.60 (d, 1H), 7.39 (d, 1H), 7.36-7.31 (m, 1H), 7.24-7.17 (m, 1H), 6.82 (br s, 0.5H), 6.73-6.65 (m, 1H), 4.38-3.77 (m, 4H), 1.18-1.07 (m, 3H). MS (El) for C19H19F3IN3O2: 505 (M*). EXAMPLE 20(e). methyl (1-(1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-y!]ethyl}carbamate: 'H NMR (400 MHz, d6-DMSO): 8.59 (d, 1H), 7.58 (d, 1H), 7.41-7.05 (m, 4H), 6.72- 6.64 (m, 1H), 5.84 (d, 1H), 4.20 (d, 0.5H), 4.08-4.04 (m, 1H), 3.92-3.85 (m, 1.5H), 3.76-3.71 (m, 1H), 3.69-3.63 (m, 1H), 3.46 (d, 2H), 0.99-0.95 (m, 3H). MS (El) for C20H19F3IN3O4: 550 (MH*). EXAMPLE 20(1). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[l-(dimethylamino)cthyl]azetidin-3-ol: !H NMR (400 MHz, d6-DMSO): 9.45 (s, 1H), 8.61 (d, 1H), 7.60 (d, 1H), 7.39 (d, 1H), 7.38-7.33 (m, 1H), 7.24-7.18 (m, 1H), 7.05 (s, 1H), 6.73-6.66 (m, 1H), 4.48 (d, 0.5H), 4.36 (d, 0.5 H), 4.26 (d, 0.5H), 4.16-4.11 (m, 1H), 4.00-3.94 (m, 1H), 3.86 (d, 0.5H), 3.60-3.54 (m, 1H), 2.75-2.70 (m, 3H), 2.66-2.62 (br s, 3H), 1.22 (dd, 3H). MS (El) tor C20H21F3IN3O2: 520 (MH+). EXAMPLE 20(g). l-({354-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}earbonyl)-3-(l -nitropropyl)az.eiidin-3~ol: *H NMR (400 MHz, CD3OD): 7.46 (m, 1H), 7.35 (m, 1H), 7.28 (m, 1H), 7.07 (m, 1H), 6.61 (m, 1H), 4.65 (m, 1H), 4.44 (m, 1H), 4.25 (m, 1H), 4.02 (m, 1H), 3.86 (m, 1H), 2.04 (m, 1H), 1.76 (m, 1H), 0.94 (m, 3H). MS (El) for CisHiiFsI^CV. 536 (MH"). EXAMPLE 20(h). 3-( 1-aminopropyl)-1-((3,-4-diiluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3~ol: *H NMR (400 MHz, CD3OD): 7.45 (m, 1H), 7.34 (m, 1H), 7.28 (m, 1H), 7.05 (m, 1H), 6.61 (m, IH), 4.21 (m, 1H), 4.09-3.86 (m, 2H), 3.78 (m, 1H), 2.63 (m, 1H), 1.50 (m, 1H), 1.24 (m, 1H), 0.98 (m, 3H). MS (El) for Ci9H19F3lN302: 506 (ΜϊΓ). EXAMPLE 20(i). l-({3,4-difluoro~2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[l-(ethylamino)propyl]azetidin-3-ol: !H NMR (400 MHz, CD3OD): 7.45 (m, 1H), 7.34 (m, 1H), 7.28 (m, 1H), 7.05 (m, 1H), 6.61 (m, 1H), 4.23 (m, 1H), 4.02 (m, 1H), 3.90 (m, 1H), 3.79 (m, 1H), 2.70 (m, 1H), 2.54 (m, 1H), 1.53 (m, 1H), 1.40 (ra, IH), 1.05 (m, 3H), 0.95 (m, 3H). MS (El) for C21H23F3IN3O2: 534 (MH+). EXAMPLE 20(1). 3-[l-(diethylamino)propyl]-l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3~ol: !H NMR (400 MHz, CD3OD): 7.44 (m, IH), 7.33 (in, IH), 7.27 (m, 1H), 7.07 (m, IH), 6.60 (m, 1H), 4.21 (m, IH), 4.10 (m, IH), 4.03-3.70 (m, 2H), 2.71-2.45 (m, 5H), 1.67 (m, IH), 1.49 (m, IH), 0.94 (m, 9H). MS (El) for C23H27F3IN3O2: 562 (MH*). EXAMPLE 20(k). 3-[amino(phenyl)methyl]~l-({3,4-difluoro-2-[(2-iluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol): MS (El) for C23H19F3IN3O2: 554 (MH+). EXAMPLE 20(m). 1-((3,4-di fluoro-2- [(2-fluoro-4- iodophenyl)araino]phenyl}carbonvl)~3-(3-methyl-1 -nitrobutyl)azetidin-3-ol): SH NMR(400MHz, CDC13): 8.38 (s, IH), 7.39 (dd, IH), 7.34-7.31 (m, IH), 7.14-7.10 (τη, IH), 6,84-6,77 (m, IH), 6,63-6,58 (m, IH), 4.68 (dd, IH), 4.23-4.04 (br m, 4H), 2,13 (t, 2H), 1.64-1.44 (br m, 3H), 0.93 (d, 6H); MS (El) for C2!H2!F3IN304: 564 (Mrf), EXAMPLE 20(n). 3-(1 -aminobutyl)-1-((3,4-difIuoro-2- [(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol acetate salt; !H NMR (400 MHz, CD3OD): 7.48-7.43 (d, 1H), 7.38-7.33 (d, 1H), 7.32-7.26 (m, IE), 7.09-7.00 (q, IH), 6.66-6.58 (t, IE), 4.33-4.22 (d, IH), 4.13-3.81 (τη, 3H), 3,17-3,09 (t, IE), 1.93-1.89 (s, 3H), 1.89-1.82 (t, 3H), 1.56-1,24 (m, 4H), 0.97-0.88 (t, 3H); MS (El) for C20H21F3IN3O2: 520 (MH+). EXAMPLE 20(o). 3-(l-aininocyclopentyl)-l-({3s4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3~ol acetate salt: !H NMR (400 MHz, CDCI3): 8.27-8.21 (s, IH), 7,42-7.36 (d, IH), 7.34-7.29 (d, IH), 7.15-7.09 (t, IE), 7.09-7.01 (q, IH), 6.88-6.79 (q, IH), 6.63-6.53 (ra, IH), 4.18-3.92 (m, 4H), 2.12-2.08 (s, 3H), 2.06-1.70 (m, 7H), 0,92-0.68 (m, 4H); MS (El) for C21H21F3IN3O2: 532 (ΜΪ·Γ). EXAMPLE 20(p). JV-{l-[l-({3,4-difluoro-2-[(2-fluoro-4-
iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]ethyl}acetamide: H NMR (400 MHz, CDC13); 8.42 (s, IH), 7.41-7,38 (dd, IH), 7.34-7,32 (dt, IH), 7.12 7.09 (m, IH), 6.85-6.78 (m, IH), 6.63-6.57 (m, IH), 5.76 (b, IH), 4.28-3.98 (m, 5H), 2.00 (s, 3H), 1.20-1.19 (d, 3H); MS (El) for C20H19F3IN3O3: 534 (MH+). EXAMPLE 20(q). (2R)-/V-{1 ~[1 -((3;4-difluoro-2-[(2-fluoro~4-iodophenyl)amino]phenyl} carbonyS)-3 -hydroxyazetidin-3 -y IJethyl} -.>,3,3 -tr ifluoro-2-(methyloxy)-2-phenylpropanamide: SH NMR (400 MHz, CDC13): 8.47 (s, IH), 7.45 7.40 (m, 5H), 7.33-7.31 (m, IH), 7.21-7.19 (m, IH), 7.12-7.05 (m, IH), 6.85-6.76 (m, IH), 6.63-6.58 (m, IH), 4.20-3.99 (m, 5H), 3.36 (s, 1.5H), 3.34 (s,l,5H), 1.27-1.25 (d, 1.5H), 1.24-1.22 (d, 1.5H); MS (El) for CwKfcMFeD^O^ 708 (MH+). EXAMPLE 20(r). (2K)-N~ {(1R)-1 - [ 1 -({3,4-difluoro~2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]ethyl}-3,3,3-trifluoro-2- (methyloxy)-2-phenylpropanamide: SH NMR (400 MHz, CDCS3): 8.49 (s, IH), 7.467.391 (m, 5H), 7.33-7.31 (m, IH), 7.21-7.16 (m, IH), 7.14-7.10 (m, IH), 6.85-6.79 (m, IH), 6.64-6.58 (m, IH), 4.24-4.00 (m, 5H), 3.35 (s, 3H), 1.25-1.23 (d, 3H); MS (El) for C28H24F6IN304: 708 (ΜΗΓ). EXAMPLE 20(s). !-({3,4-difSuoro-2-[(2-fluoro-4-
iodophenyl)amino]pheny I} carbonvl)-3 -(1 -methyl-1 ~niiroethyl)azetidin-3 -ol: H NMR (400 MHz, CDC13): 8.28 (s, 1H), 7.41-7.38 (dd, 1H), 7.34-7.32 (dt, IH), 7.14- 7.10 (m, IK), 6.87-6.81 (m, 1H), 6.64-6.59 (m, IK), 4.33-4.15 (m, 4H), 1.64 (s, 6H); MS (El) for C,9Hi7F3lN3as: 536 (MH+). EXAMPLE 20(t). 3-(1-amino-1 -methylethyl)-1 -({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol: SH NMR (400 MHz, CDC13): 8.30 (s, 1H), 7.39-7.36 (dd, 1H), 7.32-7.30 (dt, 1H), 7.13-7.09 (m, 1H), 6.85-6.79 (m, 1H), 6.62-6.56 (m, IH), 4.25-3.97 (m, 4H), 1.14 (s, 6H); MS (El) for C,9H19F3IN302: 506 (MH+). EXAMPLE 21 1-((3,4-Difluoro-2-[(2-fluoro-4-iodophenyI)amino]phenyl}carbonyI)-3-{l-[(iraws-4-hydroxycyclohexyI)amino]ethyl}azetidin-3-ol hydrochloride
[00387] Potassium ferf-butoxide (1.672 g, 14.9 mmol) and ethyltriphenylphosphonium bromide (5.538 g, 14.9 mmol) were stirred in ether (30 mL) at amibient for 1 h. 1,1-Dimethylethyl 3-oxoazetidine-1 -caxboxylate (954 mg, 6.0 mmol), prepared using procedures similar to those described in Example 3, was added and the mixture was 35 °C for 4.5 h. Mixture was filtered through celite and the solid was washed with ether. The filtrate was washed with water, brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 20% ether in hexanes) gave 1,1 -dimethylethyl 3-ethylideneazetidine-l-carboxylate (506 mg, 2.76 mmol, 49% yield): 1H NMR (400 MHz, CDC13): 5.37-5.28 (m, 1H), 4.47-4.39 (m, 4H), 1.56-1.51 (m, 3H), 1.45 (s, 9H).
[00388] 1,1-Dimethylethyl 3-ethylideneazetidine-1 -carboxylate (506 mg, 2.76 mmol), and 4-methyfmorpholine AZ-oxide (1.04 g, 8.89 mmol) were dissolved in acetone i water (4:1; 30 mL) and osmium tetroxide (2.5 wt.% in i-buianol; 0.2 mL) was added. The solution was stirred at ambient for 5 days, then was quenched with saturated sodium bisulfite (2 mL) and concentrated in vacuo. The residue was partitioned between ethyl acetate and brine. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, ethyl acetate) gave 1,1-dimethylethyl 3-hydroxy~3-(l-hydroxyethyl)azetidine-l~carboxylate (375 mg, 1.73 mmol, 63% yield): !H NMR (400 MHz, CDC13): 4.00-3.77 (m, 5H), 2.65 (br s, 1H), 1.86, (br s, 1H), 1.44 (s, 9H), 1.25 (d, 3H).
[00389] 1,1 -Dimethylethyl 3-hydroxy-3-( 1 -hydroxyethyl)azetidine-1 -carboxylate (200 mg, 0.922 mmol) was dissolved in methanol (5 mL) and 4 N hydrochloric acid in dioxane (1 mL, 4 mmol) was added. The mixture was refluxed for 15 minutes and then was concentrated in vacuo to afford 3-(1 -hydroxyethyl)azetidin-3-ol hydrochloride (0.922 mmol).
[00390] 3,4-Difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoic acid (362 mg, 0.921 mmol), prepared using procedures similar to those described in US 7,019,033, 4-(dimethylamino)pyridine (337 mg, 2.76 mmol) and 1 -(3-dimethylaminopropyl)- 3- ethylcarbodiimide hydrochloride (212 mg, 1.11 mmol) were dissolved in DMF (3 mL). The mixture was stirred at ambient for 5 minutes and then 3-(1-hydroxyethyl)azetidin-3-ol hydrochloride (0.922 mmol) in DMF (2 mL) was added and the mixture was stirred for 15 h. The mixture was partitioned between ethyl acetate and 5% lithium chloride. The organic portion was washed with 20% citric acid, saturated sodium bicarbonate and brine, then was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 80% ethyl acetate in hexanes) gave 1-((3,4-difluoro-2- [(2-fluoro- 4- iodophenyl)amino] phenyl} carbonyl)-3 -(1 -hydroxyethyl)azetidin-3-ol (296 mg, 0.602 mmol, 65% yield): MS (El) for C18H[6F3IN203: 493 (MH+).
[00391] l-({3,4-Difluoro~2-[(2-fluoro-4~iodophenyl)amino]phenyl}carbonyl)- 3-(1 -hydroxyethyl)azetidin-3-ol (267 mg, 0.543 mmol), was dissolved in dichloromethane (10 mL) and treated with 4-(dimethylamino)pyridine (80 mg, 0.661 mmol) and 2,4,6-triisopropylbenzenesulfonyl chloride (183 mg, 0.604 mmol) at ambient for 15 h. Triethylamine (0.076 mL, 0.545 mmol) was added and the mixture was stirred at ambient for 3 h and then at 35 °C for 4 h and then at ambient for a furhter 15 h. 2,4,6-Triisopropylbenzenesulfonyi chloride (110 mg, 0.363 mmol) was added and the mixture was stirred at 35 °C for 3 h and then 4-(dimethylamino)pyridine (SO mg, 0.661 mmol) was added and the mixture was stirred at 35 °C for 2 h. 2,4,6-Triisopropylbenzenesulfonyl chloride (303 mg, 1.0 mmol) was added and the mixture was stirred at 35 °C for a furhter 18 h. The mixture was adsorbed on to silica and purified by column chromatography (silica gel, 30-50% ethyl acetate in hexanes) to give l-[l"({3,4-difluoro-2-[(2-fluoro-4~ iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]ethyl 2,4,6-tris(l-methylethyl)benzenesulfonate (201 mg, 0.265 mmol, 49% yield): MS (El) for C33H38F3IN2O5S: 759 (MFf).
[00392] l-[l-({3,4-Diiluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]ethyl 2,4,6-tris(l-methylethyl)benzenesulfonate (194 mg, 0.256 mmol) was dissolved in tetrahydrofuran (2 mL) and was cooled to 0 °C. Sodium hydride (60 wt% dispersion in oil: 31 mg, 0.775 mmol) was added and the mixture was stirred at 0 °C for 15 minutes. The mixture was quenched with saturated sodium bicarbonate solution and partitioned with ethyl acetate. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 50% ethyl acetate in hexanes) gave 2,3-difluoro-A;-(2~ fluoro-4-iodophenyl)-6~[(2~methyl-l-oxa~5~azaspiro[2.3]hex-5-yl)carbonyl]aniline (120 mg, 0.253 mmol, 99% yield): MS (El) for C18H14F3IN2O2: 475 (MFT).
[00393] 2,3-Difluoro-iV-(2-fluoro-4-iodophenyl)-6-[(2-methyl-l-oxa-5-azaspiro[2.3]hex-5-yl)carbonyl]ani!ine (50 mg, 0.105 mmol) was dissolved in dimethylsulfoxide (0.8 mL) and treated with rrans-4-cyclohexanolamine (70 mg, 0.609 mmol) with 100 W microwave power at 100 °C for 45 minutes. The mixture was purified by reverse phase HPLC and the clean fractions were combined, neutralized with saturated sodium bicarbonate solution and the organic solvent was removed in vacuo. The remaining aqueous residue was extracted twice with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give a residue which was treated with aqueous hydrochloric acid and then was iyophilized to afford l-({3,4-difluoro-2-[(2-fluoro-4~iodophenyl)amino]phenyl }carbonyl)-3~ {l~[(trans-4-hydroxycyclohexyl)amino]ethyl}azetidin-3-ol hydrochloride (36 mg, 0.058 mmol, 55% yield): !H NMR (400 MHz, d6-DMSG): 8.61 (hr s, 0.5H), 8.55 (hr s, G.5H), 8.49-8.33 (m, 1H), 8.08-7.90 (m, 1H), 7.59 (dd, 1H), 7.39 (br d, 1H), 7.37-7.30 (m, 1H), 7.21 (br q, 1H), 6.81 (br d, 1H), 6.77-6.65 (m, 1H), 4.20 (br d, 1H), 4.09-4.02 (m, 1H), 3.97 (br d, 1H), 3.93-3.80 (m, 1H), 3.62-3.47 (m, 1H), 3.03-2.90 (m, 1H), 2.07-1.93 (m, 2H), 1.93-1.77 (m, 2H), 1.54-1.06 (m, 8H); MS (El) lor C24H27F3IN3Q3: 590 (MH+).
[00394] EXAMPLE 21(a), Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compound of the invention was prepared: 1-((3,4-Difl uoro-2-[(2-fluoro-4- iodophenyl)amino]pheny 1} carbonyl)-3 - {1 -[(1,1 -dimethylethyl)amino]ethyl}azetidin- 3-ol: lH NMR (400 MHz, d6-DMSO): 8.63 (br s, 0.4H), 8.53 (br s, 0.6H), 7.56 (dt, 1H), 7.40-7.34 (m, 1H), 7.32-7.26 (m, 1H), 7.25-7.13 (m, 1H), 6.72-6.62 (m, 1H), 5.43 (br s, 1H), 4.14-3.56 (m, 4H), 2.69-2.53 (m, 1H), 1.00-0.85 (br, 12H); MS (El) for C22H25F3IN3O2: 548 (ΜΪ-Γ), EXAMPLE 22(a) and 22(b) 1-((3,4-difluoro-2“[(2"fluoro-4-iodophenyI)ammo]phenyl}earbonyl)-3"[(2R)" piperidin-2-yl] azetidin-3-oi
and 1-((3,4-difluoro-2~[(2-fluoro-4-iodophenyl)amino]pheny!}carbonyl)~3-[(2S)- piperidin-2-yI] azetidin-3-ol
[00395] To a solution of 1,1-dimethylethyl 2-(3-hydroxy-1 -([(phenylmethyl)oxy]carbonyl}azetidin-3-yl)piperidine-l-carboxylate (368 mg, 0.94 mmol), prepared using procedures similar to those described in Reference 5, in dichloromethane (5 mL) was added DMAP (115 mg, 0.94 mmol) and the resulting solution was cooled to 0°C. (i?)-(-)-a-Methoxy-a-trifluoromethylphenylacetyl chloride (105 pL, 0.56 mmol) was added to the solution by syringe and the mixture was allowed to warm to room temperature then stirred an additional 12 hours. The solution was then partitioned with saturated aqueous soldium bicarbonate and the organic phase dried over anhydrous magnesium sulfate then filtered and concentrated to an oily residue. Silica gel flash chromatography using hexanes:ethyl acetate 3:1 as eluent afforded the less polar 1,1-dimethylethyl (2Λ)-2-(1-{[(phenylmethyl)oxy] carbonyl} -3 - {[(2 j?)~3,3,3-trifluoro-2-(methyloxy)-2-phenylpropanoyl]oxy}azetidin-3-yl)piperidine-l-carboxylate (27.5 mg. 5% yield), the more polar 1,1-dimethylethyl (25)-2-( 1 - {[(phenylmethyl)oxy] carbonyl )-3-( [(27?)- 3.3.3- trifluoro-2-(methyloxy)-2-phenylpropanoyl]oxy}azetidin-3-yl)piperidine-l-carboxylate (105 mg, 19% yield) and starting material (253 mg, 69% recovery), [00396] The starting material thus recovered was taken into dichloromethane (3 mL) followed by addition of DMAP (115 mg, 0.94 mmol) and (J?)-(-)-a-methoxy-a-trifluoromethylphenylacetyl chloride (105 pL, 0.56 mmol) and the mixture was allowed to stir at room temperature over 12 hours. Proceeding as before afforded combined 1,1 -dimethylethyl (2R)-2-( 1 - {[(phenylmethyl)oxy] carbonyl }-3-{ [(2 j?)- 3.3.3- trifluoro-2-(methyloxy)-2-phenylpropanoyl]oxy}azetidin-3-yl)piperidine-l-carboxylate (46.6 mg, 8% yield), the more polar 1,1 -dimethylethyl (25)-2-(1-{[(phenylmethyl)oxy]carbonyl}-3-{[(2i?)-3,3,3-trifluoro-2-(methyloxy)-2-phenylpropanoyl]oxy}azetidin-3-yl)piperidine-1-carboxylate (228 mg, 41% yield) and starting material (100.8 mg, 27% recovery).
[00397] The starting material thus recovered was taken into tetrahydrofuramdichloromethane (1:1,2 mL) followed by addition of DMAP (47 mg, 0.39 mmol) and (R)-(-)-a-methoxy-a-trifluoromethylphenylaceiyl chloride (80 pL, 0.43 mmol) and the mixture was heated to 60 °C over 12 hours. Proceeding as before afforded combined less polar 1,1-dimethylethyl (2i?)-2-(l-{[(phenylmethyl)oxy]carbonyl}-3-{[(2i?)-3,3,3-trifluoro-2-(methyloxy)-2-phenylpropanoyl]oxy}azetidin-3-yl)piperidine-l-carboxylate (144 mg, 26 % yield). The chiral ester derivatives thus obtained were again subject to silica gel flash chromatography using hexanes:ethyl acetate 3:1 as eluent to give the pure less polar 1,1-dimethylethyl (2/?)-2-(l-{[(pheny!methyl)oxy]carbonyl}-3-{[(2i?)-3,3,3-trifIuoro- 2-(methyloxy)-2-phenylpropanoyl]oxy}azetidin-3-yl)piperidine-1 -carboxylate (122.8 mg, 22% yield) and the more polar 1,1-dimethylethyl (25)-2-(1-{[(phenylmethyl)oxy]carbonyl}-3-{[(2i?)-3,3,3-trifluoro-2-(methyloxy)-2- phenylpropanoyl]oxy}azetidin-3-yl)piperidine- i -carboxylate (177.6 mg, 32% yield) both as colorless amorphous residues.
[00398] 1,1 -Dimethylethyl (2J?)-2-( 1 - {[(phenylmethyl)oxy]carbonyl }-3 - {[(2i?)~ 3,3,3-trif!uoro-2-(methyloxy)-2-pheny Ipropanoyl] oxy } azetidin-3-yl)piperidine-1-carboxylate (122.8 mg, 0.21 mmol) was taken into methanol (4 mL) followed by addition of 1M aqueous sodium hydroxide (1 mL) and the resulting solution was stirred for one hour at room temperature. The solution was then partitioned with ethyl acetate and IN aqueous hydrochloric acid. The organic layer was washed with brine, dried over anhydrous magnesium sulfate then filtered and concentrated. The residue was purified by silica gel flash chromatography using hexanes:ethyl acetate 2:1 to give 1,1-dimethylethyl (2i?)-2~(3-hydroxy-l-{[(phenylmethyl)oxy]carbonyl}azetidin- 3-yl)piperidine-1 -carboxylate (60.8 mg, 81% yield) a colorless amorphous solid. 1,1-dimethylethyl (28)-2-(3-hydroxy-1 -{[(phenylmethyl)oxy]carbonyl}azetidin-3-yl)piperidine-1 -carboxylate (87.4 mg, 75% yield) was prepared analogously.
[00399] 1,1-Dimethylethyl (2i?)-2-(3-hydroxy-1- {[(phenylmethyi)oxy]carbonyl}azetidin-3-yl)piperidine-l-carboxylate (60.8 mg, 0.16 mmol) and 10% Pd/C (30 mg) were taken into methanol (2 mL) and the mixture hydrogenated at ambient pressure for one hour. The suspension was then filtered through a celite pad and concentrated then dried in vacuo to a colorless solid. The solid amine was taken into THF (1 mL) followed by addition of DIPEA (42 pL, 0.24 mmol) and 3,4-difluoro-2-[(2-fluoro-4-iodophenyS)amino]benzoyl fluoride (63 mg, 0.16 mmol), prepared using procedures similar to those described in Reference 1, and the mixture stirred at room temperature for 30 minutes. The reaction mixture was partitioned with ethyl acetate and 1 N aqueous hydrochloric acid and the organic layer washed with brine, dried over anhydrous magnesium sulfate then filtered and concentrated. Purification of the residue by silica gel flash chromatography using hexanes:ethyl acetate 3:2 as eluent afforded 1,1-dimethylethyl (2/?)-2-[l-({3,4-difluoro-2-[(2-fluoro-4~iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yljpiperidine-1 -carboxylate (74.9 mg, 74% yield) as an amorphous solid. 1,1-Dimethylethy! (2/?)-2-[l-({3,4-difluoro-2-[(2-fluoro~4- iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]piperidine-l-carboxylate lH NMR (400 MHz, CDC13): 8.53 (br s, 0.5H), 8.40 (br s, 0.5H), 7.41-7.38 (dd, 1H), 7.34-7.3l(dt, 1H), 7.17-7.14 (m, 1H), 6.86-6.79 (m, 1H), 6.63-6.587 (m, 1H), 4.24- 3.90 (m, 4H), 3.37-3.23 (m, 1H), 2.90-2.80 (m, 1H), 1.85-1.54 (m, 7H), 1.43 (s, 9H); MS (El) for C26H29F3IN3O4: 576 (M-C4H9+).
[00400] 1,1-dimethylethyl (2j?)-2-[l-({354-difluoiO~2~[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]piperidine-l-carboxylate (74.9 mg, 0.12 mmol) was taken into methanol (1 mL) followed by addition of 4 N HC1 in dioxane (1 mL) and the solution was stirred at room temperature for one hour. The solution was then concentrated and the residue partitioned with chloroform and saturated aqueous sodium bicarbonate, The organic layer was washed with brine, dried over anhydrous sodium sulfate then filtered and concentrated. Purification of the residue by silica gel flash chromatography using ethyl acetate then concentrated aqueous ammonia in chloroform and methanol (0.1:10:1) as eluents afforded 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(2/i)-piperidin-2-yIjazetidin-3-ol (57.3 mg) as a colorless amorphous solid. The free base was taken into methanol (1 mL) then brought to about pH 1 by addition of 4 N HC1 in dioxane and the solution concentrated. The residue was triturated with ethyl ether to afford a suspension. The solid was collected by filtration to afford 1 -((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(2/?)-piperidin-2-yl]azetidin-3-ol hydrochloride salt (49 mg, 72% yield) as a colorless solid. !H NMR (400 MHz, CDCI3): 8.43-8.39 (d, 1H), 7.41-7.38 (dd, 1H), 7.33-7.3l(dt, 1H), 7.14-7.10 (m, 1H), 6.84-6.80 (m, 1H), 6.63-6.57 (m, 1H), 4.12-3.99 (m, 4H), 3.10-3.08 (d, 1H), 2.72-2.69 (d, 1H), 2.64-2.62 (m, 1H), 1.61-1.58 (m, 2H), 1.36-1.16 (m, 4H); MS (El) for C2!H2iF3lN302: 532 (MH+).
[00401] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: EXAMPLE 22(c). 1,1-dimethylethyl (2S)-2-[l-((3,4-difluoro-2-[(2-f!uoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]piperidine-l-carboxylate: 'H NMR (400 MHz, CDC13): 8.52 (br s, 0.5H), 8.39 (br s, 0.5H), 7.41-7.38 (dd, 1H), 7.34-7.3l(dt, 1H), 7.17-7.12 (m, 1H), 6.85-6.79 (m, 1H), 6.63-6.57 (m, 1H), 4.25-3.88 (m, 4H), 3.34-3.26 (m, 1H), 2.80-2.90 (m, 1H), 1.85-1.54 (m, 7H), 1.43 (s, 9H); MS (El) for C26H29F3IN3O4: 576 (M-C4H9+). EXAMPLE 22(d). 1-((3,4-difluoro~2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(2S)-piperidin-2-yl]azetidin-3-ol hydrochloride: SH NMR (400 MHz, d^-Methanol): 7.49-7.46 (dd, 1H), 7.37-7.35(dt, IH), 7.35-7.30 (m, IH), 7.10-7.04 (m, 1H), 6.64-6.59 (m, IH), 4.39-4.32 (dd, IH), 4.21-4.18 (dd, 1H), 4.13-4.07 (m, 1H), 3.97-3.88 (dd, 1H), 3.57-3.32 (m, 1H), 3.02-2.96 (dd,lH), 1.90-1.50 (m, 7H); MS (El) for C21H21F3IN3O2: 532 (Mff). EXAMPLE 22(e). l-({2-[(4-bromo-2-chlorophenyl)amino]-3,4-difluorophenyl}carbonyl)-3-piperidin-2-ylazetidin-3-ol acetate salt: *H NMR (400 MHz, CD3OD): 7.56 (d, 1H), 7.29-7.38 (m, 2H), 7.08-7.16 (m, 1H), 6.64-6.70 (m, IH), 4.30-4.40 (m, 1H), 4.18-4.26 (m, 1H), 4.04-4.14 (m, 1H), 3.90-4.00 (m, 1H), 3.16-3.26 (m, 2H), 2.86-2.96 (m, IH), 1.91 (s, 3H), 1.76-1.88 (m, 3H), 1.44-1.64 (m, 3H). MS (El) for C2!H2,BrClF2N302: 500 (M-H). EXAMPLE 22(f). 1 -({2-[(4-bromo-2-fluorophenyl)amino]-3,4-difluorophenyl}carbonyl)-3~piperidin-2-ylazetidin-3-ol acetate salt: 'H NMR (400 MHz, DMSO): 8.52 (br s, IH), 7.50 (d, IH), 7.35-7.15 (m, 3H), 6.88-6.79 (m, IH), 4.15-3.96 (m, IH), 3.84-3.78 (m, IH), 3.68-3.63 (m, IH), 2.95-2.88 (m, IH), 2.48- 2.40 (m, 2H), 1.71-1.42 (m, 3H), 1.25-1.14 (m, 2H), 1.03-0.90 (m, IH); MS (El) for C2iH2iBrF3N302: 485 (MH4). EXAMPLE 22(g). 1 -({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-pyrrolidin-2-ylazetidin-3-ol: 'H NMR (400 MHz, CD3OD): 7.45 (dd, IH), 7.37-7.31 (m, IH), 7.30-7.25 (m, IH), 7.13-6.99 (m, IH), 6.67-6.54 (m, IH), 4.20-4.09 (m,lH), 4.08-3.91 (m, 2H), 3.88-3.79 (m, IH), 3.27 (t, IH), 2.99-2.89 (m, IH), 2.88-2.81 (m, IH), 1.93-1.67 (m, 3H), 1.55-1.42 (m, IH). MS (El) for C2oHi9F3IN302: 518 (MFT) EXAMPLE 22(h). l-({3,4-difluoro-2~[(2-fiuoro-4- iodophenyl)amino]phenyi}carbony 1)-3-(l -m.ethylpyrrolidin-2~yl)azeiidin-3~oi acetate (salt): ‘H NMR (400 MHz, CD3OD): 7.46 (dd, IH), 7.38-7.26 (m, 2H), 7.12-6.99 (m, IH), 6.66-6.56 (m, IH), 4.37-3.87 (m,4H), 2.94-2.82 (m, IH), 2.75-2.63 (m, 3H), 2.20-2.06 (m, IH), 2.00-1.67 (m, 8H). MS (El) for C2iH21F3IN302: 532 (MH+). EXAMPLE 22(i). l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)ammo]pheny 1} carbonyl)-3-( 1 -ethylpyrrolidin-2-yl)azetidin-3 -ol acetate (salt): ‘HNMR (400 MHz, CD3OD): 7.46 (d, IH), 7.38-7.33 (m, IH), 7.32-7.27 (m, IH), 7.12-7.01 (m, IH), 6.66-6.57 (m, IH), 4.34-3.89 (m,4H), 3.57 (t, IH), 3.51-3.40 (m, IH), 3.28-2.81(m, 3H), 2.25-1.72 (m, 8H), 1.31-1.18 (m, 3H). MS (El) for C22H23F3IN3O2: 546 (Mrf).
EXAMPLE 22<J)- 1 -({4-fluoro-5-[(2-fluoro-4-iodophenyl)amino]-1 -methyl-1H-benzimidazol-6-yl}carbonyl)-3-((2S)-piperidin-2-yl]azetidin-3-ol acetate salt: lH NMR (400 MHz, dj-MeOH): 8.30 (s, IH), 7.56 (s, 1H), 7.42 (d, 1H), 7.24 (d, 1H), 6.34 (m, IH), 4.20 (d, 2H), 3.92 (s, 3H), 3.38-3.24 (m, 3H), 3.08 (bs, 1H), 2.88 (bs (1/7), 1.90-1.70 (m, 3H), 1.66-1.32 (m, 3H); MS (El) for CaH^INsCfe: 568 (MH+). EXAMPLE 22(k). 1 -({7-fluoro-6-[(2-fluoro-4-iodophenyl)amino]-1 -methyl-1//-benzimidazol-5-yl}carbonyl)~3-[(2S)-piperidin-2-yl]azetidin-3-ol acetate salt: 'H NMR (400 MHz, d4-Me()H): 8.22 (s, IH), 7.60 (s, IH), 7.42 (d, IH), 7.26 (d, IH), 6.46 (m, IH), 4.21 (d, 2H), 4.06 (s, 3H), 3.88 (m, IH), 3.38-3.24 (m, 3H), 3.10 (bs, IH), 2.88 (bs (1//), 1.88-1.70 (m, 3H), 1.64-1.28 (m, 3H); MS (El) for C23H24F2IN5O2: 568 (MH+). EXAMPLE 22(m). 4-[(4-bromo-2-fluorophenyl)amino]-3-fluoro-5-({3-hydroxy-3-[(2S)-piperidin-2-yl]azetidin-1 -yl}carbonyl)-1 -methylpyridin-2( 1 //)-one: MS (El) for C2i H23BrF2N403: 498 (MlF). EXAMPLE 22(n). l-({8-chloro-7-[(2-fluoro-4-iodophenyl)amino]imidazo[l,2-a]pyridin-6-yl}carbonyl)-3-[(2S)-piperidin-2-yl]azetidin-3~ol: *H NMR (400MHz, de-DMSO): 8.79 (s, IH), 8.04 (d, IH), 7.91 (d, IH), 7.64 (dd, IH), 7.55 (d, IH), 6.95- 7.02 (m, IH), 4.38 (d, IH), 4.15 (dd, IH), 3.99 (dd, IH), 3.72 (q, IH), 3.32-3.39 (m, IH), 3.00-3.12 (m, IH), 1.93 (t, 3H), 1.51-1.70 (m, 3H); MS (El) for C22H22CIFIN5O2: 532 (MH+). EXAMPLE 22(o). l-({7-[(4-bromo-2-chlorophenyl)amino]-8-chloroimidazo[l,2~ a]pyridin-6~yl}carbonyl)~3~[(2S)-piperidin-2~yl]azetidin-3~ol: 'H NMR (400MHz, drMeOH): 8.85 (s, IH), 8.06 (d, IH), 7.91 (d, IH), 7.71 (d, IH), 7.45 (d, IH), 7.01 (d, IH), 4.48 (d, IH), 4.10-4.27 (m, 2H), 3.87 (q, IH), 3.37 (d, 2H), 3.02 (s, IH), 1.88-1.94 (m, 3H), 1.58-1.69 (m, 3H); CazH^BrChNjOa: 540 (MH1). EXAMPLE 22(p). l-({6-[(4-bromo-2-chlorophenyl)amino]-7-fluoro-3-methyl-l,2-benzisoxazol-5-yl}carbonyS)~3-[(2S)-piperidin-2-yl]azetidin-3-ol: ’H NMR (400MHz, CDCU): 8.50 (m, IH), 7.51 (d, IH), 7.42 (s, IH), 7.26 (dd, IH), 6.79 (dd, IH), 4.20-3.98 (br m, 4H), 3.11 (d , IH), 2.77-2.50 (br m, 5H), 1.80-1.15 (br m, 6H); MS (El) for C23H23BrClFN4Ο3: 537 (MH+). EXAMPLE 22(q). 1 -({3-fluoro-2- [(2-fluoro-4-iodophenyl)amino]phenyi} carbonyl)-3-[(2S)-piperidin-2-yl]azetidin-3-ol: ‘H NMR (400 MHz, di-MeOH): 7.53 (2d, IH), 7.46 (m, 2H), 7.16 (t, IH), 6.86 (m, IH), 6.63 (m, IH), 4.36 (m, IH), 4.22 (m, IH), 4.02 (m, IH), 3.88 (m, IH), 3.08 (d, IH), 2.66 (dd, IH), 2.56 (m, IH), 1.82 (bs, IH), 1.66 (d, IH), 1.58 (d, IH), 1.38 (m, 2H), 1.22 (m, IH); MS (El) for QuH^D^: 514 (MH+). EXAMPLE 22(r). l-({4-fluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3~[(2S)-piperidin-2-yl]azetidin-3-oi: 'HNMR (400 MHz, cL-MeOH): 7.42 (2d, 1H), 7.34-7.18 (m, 4H), 6.46 (m, 1H), 4.10 (m, 2H), 3.84 (m, 2H), 3.04 (d, 1H), 2.52 (dd, 2H), 1.76 (bs, 0.5H), 1.58 (m, 2.5H), 1.32 (m, 2H), 1.18 (m, 0.5H), 1.04 (m, 0.5H); MS (El) for C21H22F2IN3O2: 514 (MH+). EXAMPLE 22(s). 5-[(2-fluoro-4-iodophenyl)amino]-6-({3~hydroxy-3-[(2S)-piperidin-2-yl]azetidin-l-yl}carbonyl)-2-methylpyridazin-3(2F/)-one: !H NMR (400MHz, d6-DMSO): 10.19 (s, 1H), 7.78 (dd, 1H), 7.59 (d, !H), 7.32 (t, 1H), 5.95 (s, 1H), 4.59 (q, 1H), 4.13-4.27 (m, 2H), 3.77 (d, 1H), 3.62 (s, 3H), 3.02 (d, 2H), 2.71 (d, 1H), 1.78 (s, 1H), 1.68 (d, 1H), 1.53 (d, 1H), 1.32 (s, 2H), 1.17 (t, 1H); MS (El) for C20H23FIN5O3: 528 (MH+).
Example 23 l-{[l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3- hydroxyazetidie~3“yl]methyl}-3”Eitroguaiiicline hydrochloride
[00402] To a mixture of 2,3-difluoro-iV-(2-fluoro-4-iodophenyl)-6-( 1 -oxa-5-azaspiro[2,3]hex-5-ylcarbonyl)aniline (0.15g, 0.33 mmol), prepared using procedures similar to those described in Example 21, and nitroguanidine (0.1 g, 1.00 mmol) in tetrahydrofuran (3,00 mL) an aqueous solution of sodium hydroxide (1.0 mL, 2.0 mmol) was added and the reaction mixture was stirred at 70 °C for 16 hours. The reaction mixture was concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC. The fractions were collected, and the solvent was concentrated. The residue was partitioned with ethyl acetate. The organic layer was washed with saturated aqueous sodium bicarbonate, brine and dried over anhydrous sodium sulfate. Filtration and concentration resulted in an amorphous residue, which was dissolved in methanol, and 4 N HCS in dioxane (80 μΕ, 0.33 mmol) was added to the solution. A white precipitate formed and was collected by filtration. The solid was washed with hexane, and dried to afford 76 mg (38%) l-{[l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)-3-hydroxyazetidin-3-yl]methyl} -3-nitroguanidine hydrochloride. NMR (400 MHz, dj-MeOH): 7.46 (2d, 1H), 7.36 (m, 1H), 7.29 (m, 1H), 7.02 (m, 1H), 6.63 (m, 111), 4.22 (m, 1H), 4.01 (m, 2H), 3.86 (m, 1H), 3.51 (d, 2H); MS (El) for CigHi^INeO* 565 (MH+).
[00403] EXAMPLE 23(a). Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: l-cyano-3-([1 ~({3,·4~difluoro-2~[(2-fluoro-4~ iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yl]methyl}guanidine hydrochloride. 'HNMR (400 MHz, drMeOH): 7.47 (2d, 1H), 7.36 (m, 1H), 7.27 (m, 1H), 7.03 (m, 1H), 6.63 (m, 1H), 4.18 (m, 1H), 3.98 (m, 2H), 3.80 (m, 1H), 3.43 (s, 2H); MS (El) for C!9Hi6F3IN602: 545 (MH1). EXAMPLE 24 6-({3-[(ethylamino)methyl]-3-fluoroazetidin-l-yl}carbonyl)-23-difluoro-A-(2- fluoro-4-iodophenyl)aniline
[00404] To 1,1-dimethyiethyl [{1-((3,4-difuoro-2-[(2-fluoro-4-iodopheny!)amino]phenyl}carbonyl)-3-hydroxyazetidin-3- yl]methyl}ethy!carbamate (27 mg, 0.044 mmol), prepared using procedures similar to those in Example 3 and followed by Boc-protection, in chloroform (2.5 mL) added DAST (11.8 pL, 0.089 mmol) and stirred for 3.5 hr at room temperature. Quenched with water (15 mL), partitioned phases and extracted aqueous phase with chloroform (2 X 15mL). The combined chloroform extracts were dried over sodium sulfate, filtered and the filtrate concentrated in vacuo. The residue was purified on a silica gel column to afford 1,1-dimethylethyl [{1-((3,4-difluoro-2- [(2-fluoro- 4-iodophenyl)amino]phenyl}carbonyl)-3-fluoroazetidin-3~yl]methyl}ethylcarbamate (19.0 mg, 70%).
[00405] To the 1,1-dimethylethyl [{1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)~3-fluoroazetidin-3~yl]methyl}ethyicarbamate (19.0 mg, 0.031 mmol) in acetonitrile (1,0 mL) added a solution 4.0N hydrogen chloride in dioxane (1.0 mL). After 1.5hr the solution was concentrated in vacuo.
The residue was purified by preparative reverse phase HPLC to afford the title compound (4.30 mg, 27%). *H NMR (400MHz, CDQ3): 8.25 (s, 1H), 7.33 (dd, 1H), 7.33-7.25 (m, 1H), 7.18-7.14 (m, 1H), 6.84-6.77 (m, 1H), 6.63-6.58 (m, 1H), 4.33- 4.05 (br m, 4H), 3.07-2.95 (br m, 2H), 2.65 (q, 2H), 1.08 (t, 3H); MS (El) for Cs9HigF4lN30: 508 (MH+). EXAMPLE 25 3~(2-ammoeyclohexyl)~l-({3,4-difluoro-2-[(2“fluoro-4- iodophenyl)amino]phenyl}earbony![)azetidm-3-ol
[00406] A solution of l-(trimethylsiloxy)cyclohexene (200 mg, 1.17 mmol) and benzyl 3-oxoazetidine~l-carboxylate (289 mg, 1.41 mmol), prepared using procedures similar to those described in Reference 3, in tetrahydrofuran (3.90 mL) was cooled to -78 °C for 10 minutes followed by the addition of titanium tetrachloride (0.13 mL, 1.17 mmol). The reaction mixture stirred for an additional 5 hours at -78 °C. The mixture was quenched with aqueous sodium bicarbonate and the aqueous layer was extracted with ether (2x). The organic layer was separated, dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated in vacuo. The residue was purified on silica gel chromatography column (3:2 hexanes/ethvl acetate) to afford benzyl 3-hydroxy-3-(2-oxocyclohexyl)azetidine~l-carboxylate (328 mg, 37%) . *H NMR (CDC13): 7.28-7.34 (m, 5H), 5.08 (s, 2H), 4.02 (d, 1H), 3.89 (d, 1H), 3.87 (s, 1H), 3.55 (s, 1H), 2.71 (q, 1H), 2.29-2.43 (m, 2H), 2.11 (s, 2H), 1.95 (s, 1H), 1.66 (d, 3H); MS (El) for Ci7H2iN04: 303 (MH+).
[00407] A solution of benzyl 3-hydroxy-3-(2~oxocyclohexyl)azetidine-l-carboxylate (100 mg, 330 mmol) in methanol (1.60 mL) in the presence of ammonium acetate (191 mg, 2.48 mmol was cooled to 0 °C for 1 hour. Sodium cyanoborohydride (81.5 mg, 1.30 mmol) was added and the mixture was stirred at room temperature for 16 hours. To the reaction mixture was added 6 N hydrogen chloride (800 pL) and extracted with ethyl acetate. The aqueous layer was basified with aqueous sodium bicarbonate (pH 9) and extracted with dichloromethane. The combined organic portion was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford benzyl-3-(2-aminocyclohexyl)-3-hydroxyazetidine-l-carboxylate (73.7 mg, 73%). MS (El) for C17H24N2O3: 305 (MH+).
[00408] To a solution of benzyl-3-(2-aminocyclohexyl)-3-hydroxyazetidine-1- carboxylate (202 mg, 0.663 mmol) in dioxane-water (1:1, 2.5 mL) was added di-tert-butyl dicarbonate (138 mg, 0.630 mmol) and solid sodium bicarbonate (112 mg, 1.33 mmol). The reaction mixture was stirred at room temperature for 2 hours and evaporated. The residue was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford benzyl 3-(2 ~tert~ butoxycarbonylamino)cyclohexyl)“3-hydroxyazetidine-1 -carboxylate (237 mg, 100%). 'H NMR (CH3OH): 7.15-7.21 (m, 5H), 5.45 (s, 0.5H), 5.20 (d, 0.5H), 4.95 (s, 2H), 4.81 (s, 1H), 3.81 (d, 2H), 1.43-1.74 (m, 5H), 1.39 (s, 1H), 1.31 (s, 11H), 1.20 (s, 1H). MS (El) for C22H32N2O5:405 (MH+).
[00409] A solution of benzyl 3-(2-tert-butoxycarbonylamino)cyclohexyl)-3-hydroxyazetidine-1-carboxylate (237 mg, 0.586 mmol) in ethyl acetate (2 mL) was hydrogenated over 10% palladium-carbon (200 mg, 0.586 mmol) at 40 psi for 16 hours. The reaction mixture was filtered and concentrated in vacuo to provide tert-butyl 2-(3-hydroxyazetidin-3-yl)cyclohexylcarbamate (181 mg, 100%). *H NMR (CDCI3): 5.10 (s, 1H), 4.80 ((s, 1H), 3.78-3.86 (m, 1H), 3.61 (d, 1H), 3.57 (s, 1H), 3.36 (d, 1H), 1.77 (s, 2H). 1.40-1.53 (m, 1H), 1.36 (d, 9H), 1.25 (s, 2H). MS (El) for CHH26N2O3: 271 (MH+).
[00410] To a solution of tert-butyl 2-(3-hydroxyazetidin-3-yl)cyclohexylcarbamate (181 mg, 0.669 mmol) and 3,4-difluoro-2-(2-fluorO“4-iodophenylamino)benzoyl fluoride (265 mg, 0.669 mmol), prepared using procedures similar to those described in Reference 1, in tetrahydrofuran (2.2 mL) was added N, N~diisopropylelhylamine (110 pL) at room temperature. After an hour, the reaction mixture was heated to 50 °C and stirred for 45 minutes, at which time it was cooled to room temperature and evaporated. The residue was partitioned between ethyl acetate and 10% citric acid. The organic layer was washed with aqueous sodium chloride, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford tert-butyl-2-(1 -(3,4-difluOFQ~2~(2-fluoro-4-iodophenylamino)benzoy!)-3-hydroxyazetidin-3~ yi)cyclohexylcarbamate. This crude material was taken into the next step without further purification, [00411] 7er/-butyl-2-(l-(3,4-difluoro-2-(2-fluoro-4-iodophenylamino)benzoyl)-3-hydroxyazetidin-3-yl)cyclohexylcarbamate was dissolved in a mixture of methanol (4 mL) and hydrogen chloride (4 M in dioxane) (3 mL). The solution was heated to reflux then cooled to room temperature and stirred for 16 hours. The reaction mixture was concentrated and purified by reverse phase HPLC. The purified fractions were evaporated to dryness and partitioned between ethyl acetate and aqueous sodium bicarbonate. The organic layer was dried over sodium sulfate, filtered and concentrated in vacuo to afford an oil. The residue was taken up in methanol (2 mL) and was added hydrogen chloride (4M in dioxane) (700 pL) and evaporated to dryness to afford the title compound 3~(2~aminocyclohexyl)-l-({3,4-difSuoro-2~[(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)azetidin-3 -ol hydrochloride (44.7 mg, 12%). ‘HNMR (400MHz, d6~DMSO): 8.58 (d, 1H), 7.59 (dd, 1H), 7.54 (s, 2H), 7.38 (d, 1H), 7.33 (t, 1H), 7.16-7.25 (m, 1H), 6.69 (dt, 1H), 6.41 (s, 1H), 4.26 (d, 0.5H), 4.17 (d, 0.5H), 4.04 (t, 1H), 3.90 (t, 1H), 3.79 (d, 0.5H), 3.65-3.73 (m, 0.5H), 3.45-3.51 (m, 1H), 1.88 (s, 1H), 1.65-1.88 (m, 2H), 1.47 (s,4H), 1.16-1.37 (m, 2H); MS (El) for C22H23F3IN3Q2: 546 (MH+).
[00412] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: EXAMPLE 25(c).3-(2-aminocyclopentyl)-l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol; SH NMR (40QMHz, d^-DMSO): 8.56 (d, 1H), 7.82 (d, 1H), 7.59 (td, 1H), 7.45 (s, 1H), 7.38 (d, 1H), 7.30-7.35 (m, 1H), 7.18-7.24 (m, 1H), 6.68-6.72 (m, 1H0,6.41 (s, 0.5H), 6.17 (s, 0.5H), 3.91-4.27 (m, 2.5H), 3.78-3.86 (m, 1H), 3.65-3.73 (m, 1H), 3.44-3.52 (m, 0.5H), 2.19-2.26 (m, 1H), 1.54-1.94 (m, 5H), 1.30-1.39 (m, 1H); MS (El) for C21H21F3IN3O2: 532 (MU'"). EXAMPLE 25(a) and EXAMPLE 25(b) (i-.)-l-({3J4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(trans)-2- hydroxy cyclohexyl]azetidin-3 -ol and (±)-l-({3,4-difluQro~2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(cis)-2- hv droxycyclohexy l]azetidin-3 -ol [00413] The compounds of examples 25a and 25b were synthesized starting from benzyl 3-hydroxy-3-(2-oxycyclohenyl)azetidine-l-earboxylate prepared according to the procedure given in example 25. The ketone was reduced to give benzyl 3-hydroxy-3-(2-hydroxycyclohexyl)azetidine-1 -carboxylate as a mixture of racemic diastereomers which were subjected to hydrogenation to afford 3-(2-hydroxyeyclohexyl)azetidin-3-ol. 3-(2-hydroxycycSohexyl)azetidin-3-ol was then carried forward in a coupling step with 3,4-difluoro-2-(2-fluoro-4~ iodophenylamino)benzoyl fluoride in the usual manner. The coupled material thus obtained was purified by preparative reverse phase HPLC where fraction 1 was tentatively assigned as (±)-l-({334-difluoro-2~[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)-3-[(trans)-2-hydroxycyclohexyl]azetidin-3-ol (Example 25a) and fraction 2 was tentatively assigned as (±)~ 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(cis)-2-hydroxycyclohexyl]azetidin- 3-ol. EXAMPLE 25(a). First eluting fraction: !H NMR (400 MHz, d4-MeOH): 7.44 (2d, 1H), 7.34 (t, 1H), 7.25 (m, 1H), 7.03 (m, 1H), 6.60 (m, 1H), 4.46 (d, 0.5H), 4.28 (d, 0.5H), 4.22 (d, 0.5H), 3.98 (dd, 1H), 3.89 (d, 0.5H), 3.85 (s, 0.5H), 3.77 (d, 0.5H), 3.56 (m, 1H)S 1.90 (m, 1H), 1.46-1.74 (m, 4H), 0.98-1.32 (m, 4H); MS (El) for C22H22F3IN2O3: 547 (MH+). EXAMPLE 25(b). Second eluting fraction: 5H NMR (400 MHz, d^-MeOH): 7.44 (2d, 1H), 7.33 (d, 1H), 7.26 (m, 1H), 7.04 (m, 1H), 6.59 (dd3 1H), 4.20 (m, 1.5H), 4.19 (s, 0.5H), 4.00 (m, 1.5H), 3.86 (dd, 1H), 3.74 (d, 0.5H), 1.76 (m, 2H), 1.50-1.68 (m, 5H), 1.18-1.46 (m, 4H); MS (El) for CzaH^INzC)3: 547 (MH+).
Example 26 3"({!(E)-l~ammo~2~mtroetheay!]ammo}methyi)~l~({3,4-difIuer0~2-[(2-flii©r©-4- iodopheayl)amino]phenyl}carbonyl)azetidin-3-ol
[00414] A solution of 3-(aminomethy!)-l-({3,4-difluoro-2-[(2~iluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol (0.24 g, 0.5 mmol), prepared using procedures similar to those described in Example 3, and commercially available l,l-bis(methylthio)-2-nitroethylene (0.083 g, 0.5 mmol) in ethanol (5 mL) was stirred at 70 °C for 16 hours. The reaction mixture was concentrated in vacuo, The residue was partitioned between ethyl acetate and water. The organic layer was washed with brine, dried over sodium sulfate, filtered and concentrated to afford 0.10 g, (39%) 1-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-({[(Z)-l-(methylthio)~2-nitroethenyl]amino} mcthyl)azetidin-3 -ol. MS (El) for C2oH18F3rN404S: 595 (MH+).
[00415] To a solution of (0.05 g 0.08 mmol) 1 -({3,4-difluoro~2-[(2-fluoro-4-iodopheny l)amino]pheny!) car bony 1)-3-( {[(Z)-l-(methylthio)-2-nitroethenyl]amino}methyl)azetidin-3-ol in ethanol (2 mL) was added ammonium hydroxide (0.1 mL, 0.8 mmol) and the reaction mixture was stirred at 70 °C for 16 hours. The reaction mixture was concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC. The fractions were collected and the solvent was concentrated. The residue was partitioned with ethyl acetate. The organic layer was washed with saturated aqueous sodium bicarbonate, brine and dried over anhydrous sodium sulfate. Filtration and concentration resulted in an amorphous residue, which was dissolved in methanol, and 4 N HC1 in dioxane (40 pL, 0.16 mmol) was added to the solution. A white precipitate formed and was collected by vacuum filtration. The solid was washed with hexane, and dried to afford 42 mg (87%) 3 -( {[(E)-1 -amino-2-nitroethenyl]amino} methyl)-1 -({3,4-difluoro-2-[(2-fluoro- 4-iodophenyl)amino]phenyl} carbonyl)azetidin-3-ol hydrochloride. 5H NMR (400 MHz, ds-MeOH): 7.58 (t, 0.5H), 7.44 (ί, 0.5H), 7.36 (m, 1H), 7.31 (m, 1H), 7.04 (m, 1H), 6.63 (m, 1H), 3.90-4.30 (m, 4H) 3.72 (s, 2H); MS (El) for CieHnFsINsO* 564 (MH+). EXAMPLE 27 l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)ainino]phenyl}carbonyI)-3-(l//-
imidazol-2~yImethyl)azetidin-3-oI
[00416] A solution of 2-methyl-1 -({[2-(trimethylsilyl)ethyl]oxy} methyl)-1//-imidazole (0.5 g, 2.3 mmol) (prepared using procedures similar to those described in Clader ei. al. J. of Med. Chem. 1995, 38(10), 1600-7) in tetrahydrofuran (5 mL) was cooled to -78 °C, and n-butyllithium was added (2.5 M in hexanes, 0.990 mL, 2.5 mmol). After 2 hours, 1,1-dimethyleihyl 3-oxoazetidine-1 -carboxylate (0.60 g, 3.5 mmol), prepared using procedures similar to those described in Example 3, in 2.0 mL tetrahydrofuran was added and the solution was allowed to warm to room temperature and stirred overnight. The reaction mixture was quenched with an excess of saturated aqueous ammonium chloride solution and partitioned between water and ethyl acetate. The layers were separated and the aqueous layer was extracted with ethyl acetate (2 x 10 mL). The combined organic layers were dried over sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 3:1 hexanes/ethyl acetate) gave 0.37 g (41%) of 3~{[l-({[2-(trimethylsilyl)ethyl]oxy}methyl)-li/-imidazol-2-yl]methyl}azetidin-3-ol: 5H NMR (400 MHz, CDC13): 6.96-6.92 (m, 1H), 5.23 (s, 2H), 3.98 (d, 2H), 3.79 (d, 2H), 3.52- 3.47 (m, 2H), 3.13 (s, 2H), 1.43 (s, 9H), 0.94-0.88 (m, 2H), 0.00 (s, 9H).
[00417] 3- {[1 -({[2-(trimethylsi!yl)ethyl]oxy } methyl)-1 //-imidazol-2-yljmethyl}azetidin-3 -ol (0.19 g, 0.49 mmol) was dissolved in dichloromethane (1.5 mL) and trifluroacetic acid (1.5 mL) was added. The reaction mixture was stirred at room temperature overnight and the solvent was removed under vacuum to give 0.16 g of 3-(li/-imidazol-2-ylmethyl)azetidin-3-ol trifluoroacetate salt (87%). The crude residue was used without further purification for the next step.
[00418] To a solution of 3-(1 //-imidazol-2-ylmethyl)azetidin-3-ol trifluoroacetate salt (0.16 g, 0.42 mmol) and N,JV-diisopropylethylamine (0.370 mL, 2.13 mmol) in tetrahydrofuran (2.0 mL) 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoyl fluoride (0.17 g, 0.42 mmol), prepared using procedures similar to those described in Reference 1, was added and the reaction mixture was stirred for 3 hours at room temperature. The solution was partitioned between ethyl acetate and saturated aqueous sodium bicarbonate and the organic layer was dried over sodium sulfate and concentrated in vacuo. Purification by reverse-phase HPLC followed by lyophilization of the pure fractions gave 0.032 g (13%) of 1 -({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-(li/-imidazol-2-ylmethyl)azetidin-3-ol acetate salt: *H NMR (400 MHz, CD3OD): 7.45 (dd, 1H), 7.38-7.33 (m, 1H), 7.25- 7.18 (m, 1H), 7.08-6.96 (m, 1H), 6.89 (s, 2H), 6.65-6.56 (m, !H), 4.33-4.22 (m, 1H), 4.17-4.00 (m, 2H), 3.91-3.80 (m, 1H), 3.08 (s, 2H), 1.96 (s, 3H). MS (El) for C20H16F3IN4O2: 529 (MH+). EXAMPLE 28 3-((1 R)-1 -aminoethyl]-1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)azetidin-3-ol
[00419] To a solution of diisopropylamine (6.5 mL, 46.3 mmol) in THF (200 mL) at -78 °C was added butyllithium (17 mL of a 2.5 M solution in hexanes, 42.5 mmol) over 5 min. The solution of lithium diisopropylamide was stirred for 15 min at -78 °C. A solution of (5)-4-benzyl-3-propionyl-2-oxazolidinone (9.0 g, 38.6 mmol) in THF (100 mL) was added to the lithium diisopropylamide by addition funnel over 26 min. The reaction temperature was kept below' -70 °C during the course of the addition. After the addition, the mixture was stirred for a further 30 min at -78 °C. Then phenylmethyl 3-oxoazetidine-l -carboxylate (9.5 g, 46.3 mmol) was added by addition funnel over 25 minutes as a solution in THF (100 mL). Again, the reaction mixture was kept below -70 °C during the reagent addition. After stirring for an additional 1 hour at -78 °C, the reaction mixture was quenched with saturated ammonium chloride solution and was then allowed to warm to rt. Water was added to dissolve any precipitated ammonium chloride, and ethyl acetate was added. The layers were partitioned, and the aqueous phase was extracted twice with ethyl acetate. The combined organic extracts were washed with 5% aqueous sodium bicarbonate, dried over sodium sulfate, filtered, and concentrated. The residue was purified by column chromatography (50% ethyl acetate: 50% hexanes) to provide phenylmethyl 3-hydroxy-3 ~ { (1R)-1 -methyl-2-oxo-2-[(4S)-2-oxo-4-(phenylmethyl)-1,3-oxazolidin-3-yl]ethyl}azetidine-l-carboxyiate as a white crystalline solid (6.03 g, 13.8 mmol, 36% yield). !H NMR (400 MHz, CDCI3) δ 7.37 (m, 8H), 7.20 (d, 2H), 5.12 (s, 2H), 4.66 (m, 1H), 4.27-4.20 (m, 2H), 4.10 (q, 1H), 4.03-3.93 (m, 3H), 3.28 (dd, 1H), 2.77 (dd, 1H), 1.29 (d, 3H).
[00420] A solution of lithium hydroxide monohydrate (1.16 g, 27.6 mmol) in 30% hydrogen peroxide (13.2 mL, 138 mmol) was prepared and was subsequently added slowly to a solution of phenylmethyl 3-hydroxy-3- { (1 f?)-1 -methyl-2-oxo-2-[(4S)-2-oxo-4-(phenylmethyl)-l,3-oxazolidin-3-yl]ethyl}azetidine-l-carboxylate (6.03 g, 13.8 mmol) in THF (80 mL) and water (20 mL) at 0 °C. After the mixture was stirred for 1 h at rt, the hydrogen peroxide was quenched carefully with 1 M sodium sulfite (150 mL, 150 mmol). The THF was removed in vacuo, and the mixture was then acidified to pH=2 with concentrated hydrochloric acid. The aqueous mixture was extracted twice with ethyl acetate. The combined organic extracts were dried over magnesium sulfate, filtered, and concentrated in vacuo. The resulting residue was purified by column chromatography (gradient, 5% methanol: 95% dichloromethane to 10% methanol: 90% dichloromethane) to provide (2/?)-2-(3-hydroxy-1 -{[(phenylmethyl)oxy]carbonyl}azetidin-3-yl)propanoic acid as a colorless oil (2.77 g, 9.9 mmol, 72% yield). 'H NMR (400 MHz, CDC13) δ 7.37-7.31 (m, 5H), 5.10 (s, 2H), 3.99 (s, 2H), 3.93 (s, 2H), 2.88 (q, 1H), 1.28 (d, 3H); MS (El) for ε34Ηί7Ν05: 280 (MH-).
[00421] To a solution of (2/?)-2-(3-hydroxy-l- {[(phenylmethy!)oxy]carbonyl}azetidin-3-yT)propanoic acid (2.77 g, 9.9 mmol) in toluene (100 mL) was added triethylamine (1.52 mL, 10.9 mmol) followed by diphenyl phosphoryl azide (2.24 mL, 10.4 mmol). The mixture was heated to 80 °C for 2 h and was then cooled to rt. The volatile materials were removed in vacuo, and the residue was purified by column chromatography (gradient: 50% hexanes: 50% ethyl acetate up to 100% ethyl acetate). The desired product, (8i?)-8-methyl-6-oxo-5-oxa-2,7-diazaspiro[3,4]octane-2-carboxylic acid phenylmethyl ester, was isolated as a viscous, colorless syrup (1.84 g, 6.6 mmol, 67% yield). *H NMR (400 MHz, CDC13) δ 7.39-7.32 (m, 5H), 5.66 (hr s, 1H), 5.12 (s, 2H), 4.34 (dd, 1H), 4.30 (dd, 1H), 4.17 (dd, 1H), 4.05 (dd, 1H), 3.98 (q, 1H), 1.34 (d, 3H).
[00422] To a solution of (8J?)-S~methyl-0-oxo-5-oxa~2,7-diazaspiro[3.4]octane~2-carboxylic acid phenylmethyl ester (1.84 g, 6.6 mmol) in methanol (66 mL) was added wet 10% palladium on carbon (50% by mass, 500 mg). The resulting suspension was stirred under 1 atm of hydrogen for 1 h. The catalyst was then removed by filtration through celite. The filtrate was concentrated in vacuo to provide (8i?)-8-methyl~5-oxa-2,7-diazaspiro[3,4]octan-6-one as a white solid (0.99 g, quantitative yield). *H NMR (400 MHz, CDC13) δ 5.23 (br s, 1H), 4.07 (d, 1H), 4.02 (d, 1H), 3.92 (d, 1H), 3.79 (d, 1H), 3.58 (d, 1H), 1.38 (d, 3H); MS (ΕΪ) for C6H10N2O2: 143 (ΜΙ-Γ).
[00423] A solution of (8/f)-8-methy!~5-oxa-2,7-diazaspiro[3.4]octan-6-one (937 mg, 6.6 mmol), acetic acid (0.756 mL, 13.2 mmol), and benzaldehyde (1.0 mL, 9.9 mmol) in methanol (65 mL) was treated with sodium cyanoborohydride (829 mg, 13.2 mmol) at rt for 30 min. The mixture was then cooled to 0 °C, and 3 N hydrochloric acid (100 mL) was added. The methanol was then removed in vacuo. The resulting aqueous solution was washed with ethyl acetate. The ethyl acetate wash was back extracted with 1 N hydrochloric acid, and the aqueous acidic phases were combined and basified with potassium carbonate. The organic phase was discarded. The aqueous mixture was then extracted three times with ethyl acetate. The combined organic extracts were dried over magnesium sulfate, filtered, and concentrated in vacuo. The desired (8^)-8-methyl-2-(phenylmethyl)-5-oxa-2,7-diazaspiro[3.4]octan-6-one was obtained in 93% purity as a milky colorless liquid (1.33 g, 5.73 mmol, 87% yield). MS (El) for C13H16N2Q2: 233 (ΜΗ*).
[00424] To a solution of (8/?)-8~methyl~2-(phenylmethvl)-5~oxa-2,7~ diazaspiro[3.4]octan-6-one (1.33 g, 5.7 mmol) in dioxane (40 mL) and water (20 mL) was added barium hydroxide octahydrate (9.0 g, 28.5 mmol), and the mixture was heated to reflux for 2 h. After cooling to rt, the mixture was acidified with 3 N hydrochloric acid (10 mL) and dichloromethane (50 mL) was added. The biphasic mixture was treated with potassium carbonate (1.6 g, 11.4 mmol) and di-teri-butyl dicarbonate (2.11 g, 9.7 mmol). After stirring vigorously at rt for 17 h. solids were removed by filtration, and the layers were partitioned. The aqueous phase was extracted with dichloromethane, and the organic extracts were combined and dried over magnesium sulfate, filtered, and concentrated. The residue was taken up in methanol (60 inL) and was treated with potassium carbonate (3.0 g, 22 mmol) added in two portions over 4 h at reflux. After cooling, the methanol was removed in vacuo, and the residual solids were loaded directly on to a silica column. After purification (5% methanol: 95% dichloromethane), 1,1-dimethylethyl {(17?)-1 -[3-hydroxy-1 -(phenylmethyl)azetidin-3-yl]ethyl}carbamate was obtained as a colorless syrup (1.07 g, 3.5 mmol, 62% yield). MS (El) for C17H26N2O3: 307 (MFF).
[00425] To a solution of 1,1-dimethylethyl {(17?)-1 -[3 -hydroxy-1 -(phenylmethyi)azetidin-3-yl]ethyl}carbamate (1.07 g, 3.5 mmol) in methanol was added wet 10% palladium on carbon (50% by mass, 250 mg). The resulting suspension was subjected to 1 atmosphere of hydrogen for 7 h, and an additional 250 mg of catalyst was added over the course of the reaction. The catalyst was then removed by filtration through celite. The filtrate was then concentrated in vacuo to provide 1,1-dimethylethyl [(17?)-1 -(3 -hydroxyazetidin-3 -yl)ethyl]carbamate as a colorless syrup (800 mg, quantitative yield). MS (ΕΪ) for C10H20N2O3: 161 (M - tert-butyl + H).
[00426] To a solution of 1,1 -dimethyiethyl [(1 f?)-1 -(3 -hydroxyazetidin-3 -y!)ethyl]carbamate (200 mg, 0.92 mmol) in dichloromethane (5 mL) was added diisopropylethylamine (228 pL, 1.38 mmol) and 3,4-difluoro-2-[(2-fluoro-4-iodopheny!)amino]benzoyl fluoride (prepared according to the procedures described in Reference 1) (363 mg, 0.92 mmol). The mixture was stirred at rt for 16 h, after which the volatile materials were removed in vacuo. The residue was purified by column chromatography (50% hexanes : 50% ethyl acetate) to provide 1,1-dimethylethyl {(1 R)~ 1 -[ 1 -({3,4-difSuoro-2-[(2-fiuoro-4- iodophenyl)amino]phenyI}carbonyl)-3-hydroxyazetidin-3-yl]ethyl}carbamate as a colorless film (333 mg, 0.56 mmol, 61% yield). *H NMR (400 MHz, CDCI3) δ 8.47 (br s, 1H), 7.40 (dd, 1H), 7.32 (d, 1H), 7.12 (m, 1H), 6.81 (m, 1H), 6.61 (m, 1H), 4.74 (br d, 1H), 4.22 (d, 1H), 4.15-4.07 (m, 2H), 3.96 (br s, 1H), 3.77 (m, 1H), 1.43 (s, 9H), 1.18 (d, 3H); MS (El) for C23H25F3IN3O4: 536 (M - iert-butyl + H).
[00427] A solution of 1,1-dimethylethyl {(lR)-l~[l-({354-difluoro-2-[(2-fluoro-4-iodopheny l)amino]phenyl} carbony 1)-3 -hydroxyazetidin-3 -yljethyl} carbamate (333 mg, 0.56 mmol) in methanol (10 mL) was treated with hydrochloric acid (4 N in dioxane, 1.4 mL, 5.6 mmol) at 60 °C for 30 min. After cooling, the volatile materials were removed in vacuo to provide 3 - [(1 i?)~ 1 -aminoethyl] -1-((3,4-dif!uoro-2- [(2-fluoro-4-iodophenyl)amino]phenyl} carbonyl)azetidin-3 -ol hydrochloride as a white solid (285 mg, 0.54 mmol, 97% yield). Ή NMR (400 MHz, DMSO-d*) δ 8.56 (s, 1H), 7.83 (br s, 3H), 7.59 (dd, 1H), 7.39 (d, 1H), 7.34 (m, 1H), 7.21 (q, 1H), 6.69 (m, 1H), 6.65 (s, 1H), 4.25 (dd, 1H), 4.10 (dd, 1H), 3.98 (dd, 1H), 3.80 (m, 1H), 3.48 (m, 1H), 1.11 (dd, 3H); MS (El) for C18H17F3IN3O2: 492 (MH+) [00428] To establish the enantiomeric excess (ee) of this material, 3-[(l J?)-l-aminoethyl]-1 -({3,4-difluoro-2- [(2-fluoro-4~ iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol hydrochloride (21 mg, 0.040 mmol) was dissolved in dichloromethane (400 pL) and was treated with diisopropylethylamine (20 pL, 0.12 mmol) and (i?)-(-)-a-methoxy-a-(triiluoromethyl)phenylacetyl chloride at rt for 15 min. An aliquot was removed and was analyzed by chiral HPLC. The diastereomeric excess of (2S)-N- {(1R)-1 -[ 1 -({3,4-diiluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-hydroxyazetidin-3-yljethyl}-3,3,3-trifluoro-2-(methyloxy)-2-phenylpropanamide was found to be 91%, and by extrapolation the ee of3-[(l/?)-l-aminoethyl]-l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl)carbonyl)azetidin-3-ol was also assigned to be 91%.
[00429] Example 28a. Using the sequence described above, beginning with (R)-4~ benzyl-3-propionyl-2-oxazolidinone, 3 -[(IS)-1 -aminoethyl]-1 -((3,4-difluoro-2 - [(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-oI was prepared using similar procedures except that the phenylmethyl 3-hydroxy-3-{(lS)-l-methyl-2-oxo-2-[(4i?)~ 2-oxo-4-(phenylmethyl)-l,3-oxazolidin-3-yl]ethyl}azetidine-l-carboxylate required additional recrystallizations from isopropanol. Using the same method described above in Example 28, 3-[(lS)-l-aminoethy!]-l-({3,4-difluoro-2~[(2-fluoro-4~ iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol was determined to have 98.4% ee. !H NMR (400 MHz, DMSO-d6) δ 8.56 (s, 1H), 7.84 (br s, 3H), 7.59 (dd, 1H), 7.39 (d, 1H), 7.34 (m, 1H), 7.21 (q, 1H), 6.69 (m, 1H), 6.65 (s, 1H), 4.25 (dd, 1H), 4.10 (dd, 1H), 3.98 (dd, 1H), 3.80 (m, 1H), 3.48 (m, 1H), 1.11 (dd, 3H); MS (El) for CjgHnFslNaOa: 492 (MH+).
[00430] Example 28b. To 3 - [(15)-1 -aminoethy 1]-1-((3,4-difluoro-2- [(2-fluoro-4-iodophenyl)amino]phenyI}carbonyl)azetidin-3-ol (87,4 mg, 0.18 mmol), prepared using procedures similar to those described in Example 28, was added formaldehyde (37% aqueous, 14 mg, 0.18 mmol) in methanol (2 mL) and sodium borohydride (7 mg, 0.18 mmol). The mixture was stirred for 3 h at rt, after which sodium borohydride (16 mg, 0.42 mmol) was added. Upon stirring an additional 1.25 h, more formaldehyde (37% aqueous, 1 drop) was added, and the mixture was stirred 3 days at rt. A further small spatula (~50 mg) of sodium borohydride was then added, and the mixture was stirred at rt for 30 min. After quenching with 1 N HC1, the reaction mixture was purified directly by preparative HPLC. The clean material was converted to its hydrochloride salt to provide l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbony 1)-3-[(15)-1 -(methylamino)ethyl]azetidin-3-ol as a yellow solid (21.7 mg, 0.040 mmol, 22% yield). 'H NMR (400 MHz, CD3OD) δ 7.47 (dd, 1H), 7.36 (d, 1H), 7.31 (m, 1H), 7.06 (q, 1H), 6.62 (dt, 1H), 4.36 (dd, 1H), 4.21-3,91 (in, 3H), 3.44 (q, 1H), 2.66 (s, 3H), 1.29 (br m, 3H); MS (El) for C,9H,9F3iN302: 506 (MH+). EXAMPLE 29 3~{[(l,l“DirosthylethyI)ainino]inethyl}“l"({4-[(2~iluoro-4”iodophenyl)amino]-3“ th ienyl} carbony l)azetid in-3-ol
[00431] To a mixture of methyl 4-oxotetrahydrothiophene-3-carboxylate (1.75 g, 11 mmol) (commercially available or prepared using procedures similar to those described in Rossy et. al. J. Org. Chem. 1980, 45(4), 617-2) in 15 mL of ethanol was added 2-fluoro-4-iodoaniline (2.6 g, 11 mmol) followed by addition of several drops of acetic acid . The mixture was refluxed for 3 hrs. The mixture was cooled to room temperature and the product precipitated. This product was filtered off, washed with ethyl acetate, ether, dried in vacuo to afford the methyl 4-[(2-fluoro-4-iodophenyl)amino]-2,5-dihydrothiophene-3-carboxylate (1.7 g, 42%). !HNMR(d6-DMSQ):9.80 (s,lH), 7.71 (d, 1H), 7.49 (dd, 1H), 7.24 (t, 1H), 4.10 (t, 2H), 3.79 (t, 2H), 3.69 (s, 3H); MS(EI) for C^HuFINC^S: 380 (MH+).
[00432] To a mixture of methyl 4-[(2-fluoro-4-iodophenyl)amino]-2,5-dihydrothiophene-3-carboxylate (1.2 g, 3.16 mmol) in 10 ml of anhydrous toluene was added 2,3,5,6-tetrachlorocyclohexa-2,5-diene-l,4-dione (0.78 g, 3.16 mmol). The mixture was refluxed for 2 h. The mixture was cooled to 50 °C and concentrated in vacuo to dryness and cooled to room temperature. To the residue was added ethanol and the mixture was refluxed for several minutes, cooled to room temperature and light blue crystalline product was filtered off and dried in vacuo to afford methyl 4-[(2-fluoro~4~iodophenyl)amino]thiophene-3-carboxylate (0.74 g, 62%). ’HNMR(d6-DMSO): 8.78 (s, 1H), 8.42(d, ΪΗ), 7.64 (d, 1H), 7.46 (d, 1H), 7.37(t, 1H), 7.14 (s, 1H), 3.85(s, 3H); MS(EI) for Ci2H9FIN02S: 378 (MfT).
[00433] A mixture of methyl 4-[(2-fluoro-4~iodophenyl)amino]thiophene-3-carboxylate (0.74g, 1.96 mmol) in the solution of potassium hydroxide (0.3g) in ethanol / water (4ml/4ml) was heated up to 60 °C and stirred at this temperature for 30 min. The mixture was cooled to room temperature, diluted with 4 ml of water and extracted with ether. The water layer was acidified with 1 N HC1 to pH 2, the product precipitated and was filtered off, washed several times with water and dried in vacuo to afford 4-[(2-fluoro-4-iodophenyl)amino]thiophene-3-carboxylic acid (0.59 g, 83%). 'H NMR (dft-DMSO): 13.20(s, 1H), 9.13 (s, 1H), 8.35 (d, 1H), 7.62 (dd, 1H), 7.48-7.38 (m, 2H), 7.11 (s, 1H); MS(EI) for CuH7FTN02S: 362 (MH‘).
[00434] 4-[(2-fluoro-4~iodophenyl)amino]thiophene-3-carboxylic acid (200 mg, 0.551 mmol), 4-(dimethylamino)pyridine (202 mg, 1.65 mmol) and 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (127 mg, 0.662 mmol) were dissolved in DMF (3 mL). The mixture was stirred at ambient for 5 minutes and then 3-(hydroxymethyl)azetidin-3-ο 1 hydrochloride (72 mg, 0.516 mmol) was added and the mixture was stirred for 15 h. The mixture was partitioned between ethyl acetate and 20% citric acid. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with 5% lithium chloride, saturated sodium bicarbonate and brine, then was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was crystallized from dichloromethane to afford !-({4-[(2-fluoro-4-iodophenyl)amino]-3-thienyl}carbonyl)-3-(hydroxymethyl)azetidin-3-ol (247 mg, 0.551 mmol, quantitative yield) as off-white crystals: MS (El) for CisH^FINaOsS: 449 (MH*).
[00435] 1 -({4-[(2-Fluoro-4-iodophenyi)ammo]-3-thienyl}carbony!)- 3-(hydroxymethyl)azetidin-3-ol (247 mg, 0.551 mmol), was suspended in dichloromethane (10 mL) and treated with 4-(dimethylamino)pyridine (80 mg, 0.661 mmol), and 2,4,6-triisopropyIbenzenesulfonyl chloride (183 mg, 0.604 mmol) at ambient for 15 h. The mixture was adsorbed on to silica and purified by column chromatography (silica gel, 30% ethyl acetate in hexanes) to give [l-({4-[(2~fiuoro-4~ xodophenyl)amino]-3-thienyl}carbonyl)-3-hydroxyazetidin-3-yl]methyl 2,4,6-tris(l-methylethyl)benzenesulfonate (101 mg, 0.141 mmol, 26% yield): MS (El) for C30H36FIN2O5S2: 715 (MH+).
[00436] [ 1 -( {4-[(2-Fluoro-4-iodophenyi)amino]-3 -thienyl} carbonyl)-3 -hydroxyazetidin-3-yljmethyl 2,4,6-tris(l-methylethyl)benzenesulfonate (101 mg, 0.141 mmol) was dissolved in tetrahydrofuran (2 mL) and was treated with sodium hydride (60 wt% dispersion in oil; 17 mg, 0.425 mmol) at ambient for 20 minutes. Tetrahydrofuran (2 mL) and ieri-butylamine (0.1 mL) were added and the mixture was stirred at ambient for 16 h. The mixture was concentrated in vacuo and partitioned between ethyl acetate and water. The organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by reverse phase HPLC and the clean fractions were combined, neutralized with saturated sodium bicarbonate solution and the organic solvent was removed in vacuo. The remaining aqueous residue was extracted twice with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 3-{[(l,l-dimethylethyl)amino]methyl}-l-({4-[(2-fluoro-4-iodophenyl)amino]-3-thienyl}carbonyl)azetidin-3-ol (8 mg, 0.016 mmol, 11% yield): !H NMR (400 MHz, d6~DMSO): 9.64 (br, 1H), 8.08 (d, 1H), 7.59 (dd, 1H), 7.44 (dd, 1H), 7.36 (t, 1H), 7.12 (d, 1H), 4.39 (d, 1H), 4.22 (d, 1H), 4.03 (d, 1H), 3.80 (d, 1H), 2.68 (br, 2H) 1.04 (s, 9H); MS (El) for C19H23FIN3O2S: 504 (MH+).
[00437] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: EXAMPLE 29(a). 3-[(dimethylamino)methyl]-l-({4-[(2~fluoro-4~ iodophenyl)amino]-3-thienyl}carbonyl)azetidin-3-ol: *H NMR (400 MHz, CD3OD): 7.91 (d, 1H), 7.46- 7.41 (m, 2H), 7.33 (t, 1H), 7.00 (d, 1H), 4.66 (s, 1H), 4.49 (s, 1H), 4.30 (s, 1H), 4.15 (s, 1H), 3.54 (s, 1H), 3.17-3.13 (m, 3H), 2.90 (s, 2H), 1.87- 1.83 (m, 3H); MS(EI) for CnH^FIN^S: 476 (MH+). EXAMPLE 29(b). l-({4-[(2-fluoro-4-iodophenyl)amino]-3~ thienyl }carbonyl)azetidin-3-amine: 'H NMR (400 MHz, CD3OD): 7.90 (d, 1H), 7.46- 7.41 (m, 2H), 7.31 (t, 1H), 6.99 (d, 1H), 4.47 (br.s, 2H), 4.22-4.16 (m, 2H); MS(EI) for C,4Hi3FIN3OS: 418 (Mtf). EXAMPLE 30 3-(l-ammoeihyl)-l-({8-chioro-7-[(2-fluoro-4-iodopheny!)aKiinojimidazo|l,2- a]pyridin~6-yl}carbonyl)azetidin-3-ol
[00438] To a suspension of sodium hydride (72 mg, 1.75 mmol, 60% wt) In tetrahydrofuran (1 mL) cooled to 0 °C was added nitroethane (125 pL, 1.75 mmol). The suspension was allowed to warm to room temperature and was stirred for 15 minutes, then cooled back to 0 °C. To the suspension was added dropwise a solution of 1,1-dimethylethyl 3-oxoazetidine-1 -carboxylate (300 mg, 1.75 mmol, in 2 mL of tetrahydrofuran), prepared using procedures similar to those described in Reference 3. The suspension was stirred at room temperature for 1 hour. The reaction mixture was quenched by adding 20% aqueous citric acid, and then was partitioned with ethyl acetate. The aqueous portion was extracted twice using ethyl acetate and the combined organic portion was washed with saturated sodium bicarbonate, brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a colorless oil that was purified by column chromatography. Eluting with 30% ethyl acetate in hexanes, the isolated product was concentrated in vacuo to afford 250 mg, 1.02 mmol (58%) of 1,1-dimethylethyl 3-hydroxy-3 -(1 -nitroethyl)azeiidine-1 -carboxylate as a colorless oil. !H NMR (400 MHz, DMSO): 6.46 (s, 1H), 5.01 (q, 1H), 4.24-3.97 (m, 2H), 3.77- 3.60 (m, 2H), 1.41 (d, 3H), 1.39 (s, 9H).
[00439] 1,1-DimethyIethyl 3-hydroxy-3-( 1 -nitroethyl)azetidine-l -carboxylate was dissolved in methanol (5 mL) and treated with 4 N HC1 in dioxane. The solution was briefly heated to reflux and then was concentrated in vacuo to afford 178 mg, 0.98 mmol (96%) of 3-(l-nitxoethyl)azetidin-3-ol hydrochloride as a white solid. *H NMR (400 MHz, DMSO): 9.30 (br s, 1H), 8.96 (br s, 1H), 5.12 (q, 1H), 4.44-4.38 (m, 1H), 4.22-4.17 (m, 1H), 3.94-3.87 (m, ill), 3.85-3.77 (m, 1H), 1.44 (d, 3H).
[00440] A solution of 8~chloro-7-[(2-fluoro-4~iodophenyl)amino]imidazo[l,2-a]pyridine-6-carboxylic acid (150 mg, 0.35 mmol) (prepared using procedures similar to those described in US 2006030610 and US 2005054701), N,N-diisopropylethylamine (300 pL, 1.74 mmol), PyBOP (180 mg, 0.35 mmol) and 3-(1-nitroethyl)azetidin-3-ol hydrochloride (76 mg, 0.42 mmol) in dimethylformamide (3 mL) was stirred at room temperature for 15 hours. The reaction mixture was then partitioned between 5% aqueous lithium chloride, and ethyl acetate. The aqueous portion was extracted twice using ethyl acetate. The combined organic portion was washed with 20% aqueous citric acid, brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a brown residue which was purified by column chromatography. Eluting with 5% methanol in dichloromethane, the isolated product was concentrated in vacuo to afford 195 mg, 0.35 mmol (100%) of l-({8-chloro-7-[(2-fluoro-4-iodophenyl)amino]imidazo[ 1,2-<z]pyridin-6-yl} carbony!)-3 -(1 -nitroethyl)azetidin-3-ol as a yellow foam. !H NMR (400 MHz, CDCb): 8.28 (s, 1H), 7.68 (s, 1H), 7.59 (s, 1H), 7.43 (d, 1H), 7.31 (d, 1H), 7.23 (br s, 1H), 6.55-6.51 (m, 1H), 6.02 (br s, 1H), 4.79 (q, 1H), 4.45-3.96 (4H), 1.56 (d, 3H). MS (El) for C2oHi9ClFIN604: 560 (MH+).
[00441] To a solution of l-({8-chloro-7-[(2-fluoro-4- iodophenyl)amino]imidazo [ 1,2-a]pyridin-6-yl} carbony 1)-3 -(1 -nitroethy l)azetidin-3 -ol (195 mg 0.35 mmol) in tetrahydrofuran/water (5 mL, 4:1) was added iron powder (193 mg, 3.5 mmol) and ammonium formate (438 mg, 7.0 mmol). The mixture was stirred at 80°C for 1 hour, then cooled to room temperature and filtered through a pad of celite. The celite was washed three times with boiling ethanol (20 mL). The filtrate was concentrated in vacuo and the residue was diluted with ethyl acetate. The precipitate which formed was filtered through a pad a celite and the filtrate was partitioned with water. The aqueous portion was extracted twice with ethyl acetate. The combined organic portion was washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a yellow residue which was purified by preparative reverse phase HPLC. The isolated product was concentrated in vacuo to afford 35 mg, 0.05 mmol (15%) of 3-(l-aminoethyl)-l-({8-chloro-7-[(2-fluoro-4-iodophenyl)amino]imidazo[l,2~a]pyridin-6-yl}carbony!)azetidin-3-ol acetate salt as a white solid. !H NMR (400 MHz, DMSO): 8.79 (s, 1H), 8.00 (s, 1H), 7.61 (s, 1H), 7.54 (d, 1H), 7.32 (d, 1H), 6.54-6.48 (m, 1H), 4.24-4.13 (m, 1H), 3.98-3.84 (m, 2H), 3.61-3.56 (m, 1H), 2.83 (q, 1H), 0.92-0.88 (m, 3H); MS (El) for C^H^ClFlNsOa: 530 (MH+). EXAMPLE 31 i-({8-chIoro-7-i(2-iliioro-4~iodophe0yI)amlno|imidazo[l,2-a]pyridm-6- yI}carbonyI)-3-piperidin-2-y!azetidin-3-ol
[00442] To a solution of 1,1-dimethylethyl 2-(3-hydroxy-1-{[(phenylmethyl)oxy]carbonyl}azetidin-3-yl)piperidine-l-carboxylate (595 mg, 1.52 mmol), prepared using procedures similar to those described in Reference 5, in methanol (5 mL) was added catalytic palladium on carbon (5% wt). The heterogeneous mixture was stirred under a hydrogen gas atmosphere for 15 hours at ambient pressure and then was filtered. The filtrate was concentrated in vacuo to afford 385 mg, 1.50 mmol (98%) of 1,1-dimethylethyl 2-(3-hydroxyazetidin-3-yl)piperidine-1 -carboxy late as a colorless film without further purification.
[00443] A solution of 8-chloro-7-[(2-fluoro-4-iodophenyl)amino]imidazo[l,2-a]pyridine-6-carboxylic acid (78 mg, 0.18 mmol) (prepared using procedures similar to those described in US 2006030610 and US 2005054701), 1,1 -dimeihylethyl 2-(3-hydroxyazetidin-3-yl)piperidine-1 -carboxylate (46.7 mg, 0.18 mmol), 4-(dimethylamino)pyridine (66 mg, 0.55 mmol), and finally 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (42 mg, 0.21 mmol) in dimethylformamide (2 mL) was stirred at room temperature for 15 hours. The reaction mixture was partition between 5% aqueous lithium chloride and ethyl acetate and the aqueous portion was extracted twice using ethyl acetate. The combined organic portion was washed with 1 N HC1, brine, dried over sodium sulfate, filtered and concentrated in vacuo to afford a brown residue which was purified by column chromatography. Eluting with ethyl acetate, the isolated product was concentrated in vacuo to afford 101 mg, 0.15 mmol (83%) of 1,1-dimethylethyl 2-[ 1 -({8-chloro-7-[(2-fluoro-4-iodophenyl)amino]imidazo[l,2-a]pyridin-6-yl}carbonyl)-3-hydroxyazetidin-3 -yl]piperidine-1 -carboxyiate as a white solid. The solid was immediately dissolved in methanol (5 mL) and 4 N HC1 in dioxane was added. The solution was briefly heated to reflux and then was concentrated in vacuo. The resultant residue was purified by preparative reverse phase HPLC. Isolated product was concentrated in vacuo to afford 36 mg5 0,06 mmol (40%) of l-({8-chloro-7-[(2-fluoro-4-iodophenyl)amino]imidazo[l,2-a]pyridin-6-yl}carbonyl)~3-piperidin-2-ylazetidm-3-ol acetate as a white solid. 'H NMR (400 MHz, DMSO): 8.78 (s, 1H), 8.19 (s, 0.5H), 8.15 (s, 0.5H), 8.00 (s, 1H), 7.62 (s, 1H), 7.55 (d, 1H), 7.31 (d, 1H), 6.54-6.49 (m, 1H), 4.24-4.12 (m, 1H), 3.97-3.86 (m, 2H), 3.63-3.56 (m, 1H), 2.98-2.90 (m, 1H), 2.50-2.40 (m, 1H), 1.72-1.61 (m, 1H), 1.56-1.43 (m, 2H), 1.32-1.14 (m, 2H), 1.07-0.94 (m, 1H); MS (El) for C22H22CIFIN5O2: 570 (MH+). (00444] Using the same or analogous synthetic techniques and/or substituting with alternative reagents, the following compounds of the invention were prepared : EXAMPLE 31(a). 1 ·({4-fluoro-5-[(2-fl uoro-4-iodophenyl)amino]-1 -methv\~\H-benzimidazol-6-yl}carbonyl)-3-piperidin-2-ylazetidin-3-ol acetate salt: NMR (400 MHz, DMSO): 8.35 (s, 1H), 7.84-7.77 (m, 1H), 7.54-7.49 (m, 2H), 7.25 (d, 1H), 6.31-6.25 (m, 1H), 4.04-3.92 (m, 2H), 3.90 (s, 3H), 3.86-3.78 (m, 1H), 3.70-3.62 (m, 1H), 2.94-2.85 (m, 1H), 2.45-2.32 (m, 2H), 1.66-1.36 (m, 3H), 1.26-1.08 (m, 2H), 1.01-0.80 (m, 1H); MS (El) for C23H24F2IN5O2: 568 (MH4). EXAMPLE31(a). l-({7-[(4-bromo-2-chlorophenyl)amino]-8-ch!oroimidazo[l,2-a]pyridin-6-yl}carbonyl)-3-piperidin-2~ylazetidin-3-ol acetate salt: !H NMR (400 MHz, DMSO): 8.87 (s, 1H), 8.29 (s, 0.5H), 8.21 (s, G.5H), 8.04 (s, 1H), 7.67-7.63 (m, 2H), 7.32 (d, 1H), 6.59 (d, 1H), 4.35-4.22 (m, 1H), 4.08-3.98 (m, 2H), 3.72-3.67 (m, 1H), 2.96-2.88 (m, 1H), 2.50-2.44 (m, 2H), 1.66-1.42 (m, 3H), 1.26-1.17 (m, 2H), 1.04-0.94 (m, 1H); MS (El) for CaH^BrCy^: 540 (MH4). EXAMPLE 32 3~{l-AisiisiO“3-hydroxypropyl}-l~({3,4"difliiorO“2-[(2"iliioro~4“ iodophenyl)amino]phenyl}carbonyi)azeridin-3-ol trifluoroacetate salt
[00445] Potassium /erf-hutoxide (1.393 g, 12.4 mmol) and [2-(l,3-dioxolan-2-yl)ethyl]-triphenylphosphonium bromide (5.51 g, 12.4 mmol) were stirred in ether (30 mL) at amibient for 1 h. Phenylmethyl 3-oxoazetidine-1 -carboxylate (1.025 g, 5.0 mmol), prepared using procedures similar to those described in Reference 3, was added and the mixture was stirred at 35 °C for 6 h and then at ambient for 4 days. Mixture was filtered through celite and the solid was washed with ether. The filtrate was washed with water, brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 20% ether in hexanes) gave phenylmethyl 3-(2-(1,3-dioxolan-2~vl)ethylidene]azetidine-l-carboxylate (220 mg, 0.761 mmol, 15% yield): !H NMR (400 MHz, CDQ3): 7.39-7.28 (m, 5H), 5.43-5.35 (m, 1H), 5.11 (s, 2H), 4.89 (t, 1H), 4.56 (br d, 4H), 4.00-3.92 (m, 2H), 3.91-3.83 (m, 2H), 2.27 (brt, 2H).
[00446] Phenylmethyl 3-[2-( 1,3-dioxolan-2~yl)ethvlidene]azetidine-1 -carboxylate (220 mg, 0.761 mmol), and 4-methylmorpholine N-oxide (287 mg, 2.45 mmol) were dissolved in acetone / water (4:1; 10 mL) and osmium tetroxide (4 wt.% in water; 0.05 mL) was added. The solution was stirred at ambient for 20 h, then was quenched with saturated sodium bisulfite (2 mL) and concentrated in vacuo. The residue was partitioned between ethyl acetate and brine. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, ethyl acetate) gave phenylmethyl 3-(2-(1,3-dioxolan-2-yl)-1 -hydroxyethyl]-3-hydroxyazetidine-1 -carboxylate (244 mg, 0.755 mmol, 99% yield): 5H NMR (400 MHz, CDC13): 7.38-7.28 (m, 5H), 5.11-5.07 (m, 3H), 4.14- 4.01 (m, 4H), 3.96-3.86 (m, 5H), 3.47 (d, 1H), 2.97-2.94 (m, 1H), 1.98-1.84 (m, 2H).
[00447] Phenyimethyl 3 ~[2-( 1,3"dioxolan-2-yl)-1 -hydroxyethyl] -3 - hydroxyazetidine-1 -carboxylate (235 mg, 0,728 mmol) was dissolved in methanol (5 mL) and treated with 5 wt% palladium on carbon (50 mg) under hydrogen at ambient for 1,5 h. The mixture was filtered and the filtrate was concentrated in vacuo to afford 3"[2"(l,3~dioxolan"2-yl)~l-hydroxyethyl]azetidin~3-ol (0.729 mmol): MS (El) for C8H!5N04: 190(MHf).
[00448] 3,4-Difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoic acid (287 mg, 0.730 mmol), prepared using procedures similar to those described in US 7,019,033, 4~(dimethylamino)pyridine (178 mg, 1.46 mmol) and 1 ~(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (168 mg, 0.88 mmol) were dissolved in DMF (3 mL). The mixture was stirred at ambient for 10 minutes and then 3-[2-(l ,3-dioxolan- 2-yl)-1 -hydroxyethyl]azetidin-3-ol (0.729 mmol) in DMF (2 mL) was added and the mixture was stirred for 15 h. The mixture was partitioned between ethyl acetate and 5% lithium chloride. The organic portion was washed with 20% citric acid, saturated sodium bicarbonate and brine, then was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, gradient 90% ethyl acetate in hexanes to 100% ethyl acetate) gave 1 -({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[2-(l,3-dioxolan~2-yl)~l-hydroxyethyl]azetidin-3-ol (148 mg, 0.262 mmol, 36% yield): MS (El) for C21H20F3IN2O5: 565 (MH+).
[00449] 1 -({3,4-Difluoro-2-[(2-fluoro-4-iQdophenyl)arnino]phenyl}carbonyl)-3-[2-(1,3-dioxolan-2-yl)-1 -hydroxyethyl]azetidin-3-ol (148 mg, 0.262 mmol), was dissolved in dichloromethane (10 mL) and treated with 4-(dimethylamino)pyridine (38 mg, 0.31 mmol), triethylamine (0.036 mL, 0.262 mmol) and 2,4,6-triisopropylbenzenesulfonyl chloride (303 mg, 1.0 mmol) at 35 °C for 15 h. 2,4,6-
Triisopropvlbenzenesulfony 1 chloride (100 mg, 0.33 mmol) was added and the mixture was stirred at 35 °C for 3.5 h. The mixture was adsorbed on to silica and purified by column chromatography (silica gel, 40-50% ethyl acetate in hexanes and then 100% ethyl acetate) to give l-[l-({3,4-difluoro~2-[(2-fiuoro-4-iodophenyl)amino]phenyl} carbonyl)-3 -hydroxyazeiidin-3 -y l]-2-( 1,3 -dioxolan-2-yl)ethyl 2,4,6-tris( 1 -methylethyl)benzenesulfonate (30 mg, 0.0361 mmol, 14% yield): MS (El) for C36H42F3IN2O7S: 831 (MH+).
[00450] l“[l“({3,4-Difluoro-2-[(2-fluorQ"4~iodophenyl)amino]phenyl}carbonyl)~3-hydroxyazetidin-3-yl]-2-(l,3-dioxolan-2-yl)ethyl 2,4,6-tris(l- methylethyl)benzenesulfonate (50 mg, 0.060 mmol) was dissolved in tetrahydrofuran (1 mL) and was cooled to 0 °C. Sodium hydride (60 wt% dispersion in oil; 7 mg, 0.18 mmol) was added and the mixture was stirred at 0 °C for 45 minutes. The mixture was quenched with saturated sodium bicarbonate solution and partitioned with ethyl acetate. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 50% ethyl acetate in hexanes) gave 6-{[2-(l,3-dioxolan-2-ylmethyl)-l-oxa-5-azaspiro[2.3]hex-5-y!]carbonyl}~2,3-difluoro-Ar“(2“fluoro-4-iodophenyl)aniiine (31 mg, 0.057 mmol, 94% yield): MS (El) for C21H18F3IN2O4: 547 (MH*).
[00451] 6- {[2-(1,3-Dioxolan-2-ylmethyl)-1 -oxa-5-azaspiro[2.3]hex-5-yl]carbonyl}-2,3-difluoro~A?-(2-fluoro-4-iodophenyl)aniline (31 mg, 0.057 mmol) was dissolved in dimethylformamide (0.5 mL) and sodium azide (20 mg, 0.308 mmol) was added. The mixture was stirred at ambient for 22 h. The mixture was partitioned between ethyl acetate and 5% lithium chloride. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with water, brine, then was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 50% ethyl acetate in hexanes) gave 3-[I-azido-2-(l,3-dioxoian-2-yl)ethylj~l-({3,4-difluoro-2~[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3~ol (25 mg, 0.042 mmol, 74% yield): MS (El) for C2!H19F3IN504: 590 (MH+).
[00452] 3-[ 1 -Azido-2-( 1,3-dioxolan-2-yl)ethyl]-1 -({3,4-difluoro-2- [(2-fluoro-4 -iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol (24 mg, 0.041 mmol) was dissolved in tetrahydrofuran (0.5 mL) and treated with 5% aqueous hydrochloric acid (0.5 mL) at ambient for 15 h. The misture was neutralised with saturated sodium bicarbonate solution and was extracted twice with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to afford 3-azido-3-[l~({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)aminojphenyl}carbonyl)~3-hydroxyazetidin-3-yljpropanal (21 mg, 0.0385 mmol) which was suspended in ethanol (2 mL) and treated with sodium borohydride (5 mg, 0.132 mmol) at ambient for 2 h. The mixture was quenched with acetic acid (4 drops) and concentrated in vacuo. The residue was partitioned between saturated sodium bicarbonate solution and ethyl acetate. The aqueous portion was extracted with ethyl acetate. The combined organic portion was washed with brine, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Column chromatography (silica gel, 70-80% ethyl acetate in hexanes) gave 3-(l-azido-3- hydroxypropyl)-l-({3,4-difluoro~2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyl)azetidin-3-ol (14 mg, 0.0255 mmol, 62% yield from 3 - [ 1 -azido-2-( 1,3 -dioxol an-2-yl)ethyl ]-1 -({3,4-difluoro-2-[(2-fluoro-4 -iodophenyi)amino]phenyl}carbonyl)azetidin-3-ol): *H NMR (400 MHz, CDCI3): 8.33 (br s, 1H), 7.40 (dd, 1H), 7.32 (br d, 1H), 7.13 (br t, 1H), 6.83 (br q, 1H), 6.61 (ddd, 1H), 4.32-3.94 (m, 4H), 3.92-3.84 (m, 1H), 3.82-3.71 (m, 2H), 2.56 (br, 1H), 1.94 (br, 2H), 1.26 (br, 1H); MS (ΕΪ) for C!9Hi7F3IN503: 548 (MH+).
[00453] 3-(l-Azido-3-hydroxypropyl)-l-({3,4-difluoro-2-[(2-f!uoro-4-iodopheny])amino]phenyl}carbonyl)azetidin-3-ol (14 mg, 0.0255 mmol) was dissolved in tetrahydrofuran and water (1:1, 0.5 mL) and polymer supported triphenyiphosphine (~3 mmol/g; 20 mg, 0.06 mmol) was added. The mixture was stirred at 55 °C for 1 h. TriphenyIphosphine (10 mg, 0.038 mmol) was added and the mixture was stirred at 55 °C for 1.5 h. The mixture was filtered and the filtrate was purified by reverse phase HPLC to afford 3-(1 -amino-3-hydroxypropyl)-1 -((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)azetidin-3-oi trifluoroacetate salt (1.7 mg, 0.003 mmol, 10% yield): *H NMR (400 MHz, CD3OD): 7.47 (dd, 1H), 7.36 (br d, 1H), 7.33-7.28 (m, 1H), 7.05 (br q, 1H), 6.62 (ddd, 1H), 4.38-4.26 (m, 1H), 4.18-4.00 (m, 2H), 3.98-3.88 (m, 1H), 3.78-3.67 (m, 2H), 3.61- 3.56 (m, 1H), 1.87-1.70 (m, 2H); MS (El) for C19Hs9F3IN3Q3: 522 (MH+). EXAMPLE 33 l-({3,4-difluoro-2~[(2-fltioro-4-iodophenyl)amino]plienyl}carbonyl)-3-(6- metfaylpiperidin-2~yl)azetidin-3-oi
[00454] To a solution of ΛζΑ-diisopropyIamine (1.6 mL, 11.2 mmol) cooled to -78 °C in THF (15 mL) was added a 2.5 M solution of n-BuLi in hexane (4.5 mL, 11.2 mmol) dropwise over 5 minutes and the mixture w'as stirred at this temperature for an addition 15 minutes. 6-methyl-1 -(phenylmethyl)piperidine-2-carbonitrile (2.4 g, 11.2 mmol) (prepared using procedures similar to those in Bonin et. al. Tet. Lett. 1982, 23(33), 3369-72) in THF (10 mL) was then added dropwise over 20 minutes and the reaction mixture was stirred for a further 30 minutes. Next a solution of 1,1-dimethylethyl 3-oxoazetidine-l-carboxylate (1.3 g, 7.5 mmoL), prepared using procedures similar to those in Example 3, in THF (10 mL) was added dropwise over 30 minutes. The reaction mixture was gradually warmed to room temperature and allowed to stir overnight. The reaction mixture was quenched with 10% citric acid and extracted with ethyl acetate (3 x 50 mL). The combined organic layers were washed with water, brine, dried over anhydrous sodium sulfate then filtered and concentrated in vacuo to give crude product as yellow oil. Further purification by flash chromatography (30% ethyl acetate in hexanes) afforded 1,1-dimethylethyl 3-[2-cyano-6-methyl-1 -(pheiiylmethyl)piperidin-2~yl]-3-hydroxyazetidine-1 -carboxylate as a pale yellow oil (0.2 g, 7% yield). ]H NMR (400 MHz, CDCL): 7.17-7.40 (m, 5H), 4.42 (d, 1H), 4.04-4.18 (m, 1H), 3.83-4.00 (m, 1H), 3.70-3.75 (m, 2H), 1.70-1.87 (m, 4H), 1.45 (s, 3H), 1.41 (s, 9H), 1.22-1.26 (m,lH), 1.13-1.18 (in, 2H); MS (El) for C22H31N3O3: 386 (ΜΗ").
[00455] To a stirred solution of 1,1-dimethylethyl 3-[2-cyano-6-methyl-l- (phenylmethyl)piperidin-2-yl]-3-hydroxyazetidine-1 -carboxylate (180 mg, 0.47 mmol) in ethanol (1 mL) was added acetic acid (53.5 pL, 0.94 mmol) followed by sodium cyanoborohydride (58.7 mg, 0.94 mmol) and the reaction mixture stirred at 70 °C overnight. After cooling to room temperature the suspension was filtered through celite and the solid washed with additional ethanol. The filtrate was concentrated in vacuo and taken up in ethyl acetate (30 mL). The organic layer was washed with 2 M sodium hydroxide solution. The sodium hydroxide layer was separated and washed with ethyl acetate (10 mL). The combined organic layers were washed with brine, dried over anhydrous magnesium sulfate and concentrated in vacuo to give crude 1,1-dimethylethyl 3 -hvdroxy-3 -[6-methyl-1 -(phenylmethyl)piperidin-2-yl]azetidine- 1-carboxylate as yellow oil (60 mg, 36% yield). Crude product was used further without purification. !H NMR (400 MHz, CDC13): 7.22-7.35 (m, 5H), 4.08 (d, 1H), 3.85-3.96 (m, 3H), 3.57 (d, 1H), 3.33-3.36 (m, 1H), 2.91-3.06 (m, 2H), 1.63-1.70 (m, 4H), 1.44 (s, 9H), 1.23 (d,3H), 1.05 (d, 2H); MS (El) for C2|H32N203: 361 (MIT*).
[00456] To a solution of 1,1-dimethylethyl 3-hydroxy-3-[6-methyl-l-(phenylmethyl)piperidin-2-yl]azetidine-l -carboxylate (60 mg, 0.16 mmol) in methanol (0.5 mL) was added hydrogen chloride (4N in dioxane, 0.5 mL) and the reaction mixture stirred at 60 °C for one hour. The reaction mixture was cooled to room temperature and concentrated in vacuo and aezotroped 3 times from methanol and diethyl ether. On drying the hydrochloride salt of 3-[6-methyl-l-(phenyimethyl)piperidin-2-yl]azetidin-3-ol was obtained as a dark brown residue (40 mg, 81 % yield), which was used further without purification. 'H NMR (400MHz, CD3OD): 7.58-7.63 (m, 2H), 7.47-7.49 (m, 3H), 4.78 (d, 1H), 4.44-4.62 (m, 2H), 4.29 (s, 2H), 4.22-4.26 (m, 1H), 4.12-4.18 (m, 1H), 4.08 (s, 1H), 1.60-2.00 (m, 8H), 1.48 (d, 3H); MS (El) for C!6H25C1N20: 261 (MH4).
[00457] To a solution of 3-[6-methyl-l-(phenylmethyl)piperidin-2-yl]azetidin-3-ol hydrochloride (40 mg, 0.13 mmol) in ethyl acetate (3 mL) was added acetic acid (0.5 mL) and Pd/C (50 mg) and the mixture was hydrogenated at 35 psi for 3 hours. The reaction mixture was filtered through celite. The filtrate was concentrated in vacuo. The obtained residue was dissolved in a small amount of ethyl acetate and concentrated hydrochloric acid was added and the mixture was concentrated in vacuo to give the crude dihydrochloride salt of 3 - [6-methylpiperidin-2-yl] azetidin-3 -ol (20 mg, 54%). The crude product was used further without purification. 'H NMR (400MHz, CD3OD): 4.20-4.40 (m, 1H), 4.00-4.10 (m, 1H), 3.60-3.90 (m, 2H), 1.50-2.00 (m, 6H), 1.45 (d, 3H), 1.26-1.30 (m, 1H); MS (El) for C9H20CI2N2O: 171 (MH+).
[00458] To a 0 °C solution of 3 - [6-methylpiperidin-2~yl] azetidin-3 -ol dihydrochloride (20 mg, 0.08 mmol) in DMF (1 mL) was added N,N-diisopropylethylamine (42 pL, 0.26 mmol) followed by 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino] benzoyl fluoride (32 mg, 0.08 mmol), prepared using procedures similar to those described in Reference 1, and the reaction mixture stirred at 0 °C for 30 min. The mixture was diluted with acetonitrile and purified by preparative reverse phase HPLC (CH3CN/H2O with 0.1% TFA). Fractions were collected and lyophilized to give l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl} carbonyl)-3 -(6-methyipiperidin-2-y!)azetidin-3 -ol acetate salt (7 mg, 16% yield) as a white solid. !H NMR (400MHz, CD3OD): 7.44-7.50 (m, 1H), 7.34-7.37 (m, 1H), 7.28-7.32 (m, 1H), 7.02-7.12 (m, 1H), 6.60-6.63 (m, 1H), 4.10-4.30 (m, 2H), 3.95-4.09 (m, 2H), 3.80-3.95 (m, 1H), 3.55-3.65 (m, 1H), 3.34- 3.36 (m, 1H), 1.90 (s, 3H), 1.62-1.84 (m, 6H), 1.40-1.52 (m, 1H), 1.33 (d, 3H); MS (El) for C22H23F3IN3O2: 546 (MH+). EXAMPLE 34 l~({3,4-difiuoro-2-[(2-ftuoro-4-iodophenyi)amino]phenyl}carbonyi)-3-piperazin- 2-ylazetidin~3~o!
[00459] To a solution of commercially available l,4-bis(phenylmethyl)piperazine- 2,5-dione (2.0 g, 6.8 mmol) in dry THF (50 mL) at -78 °C was added lithium diisopropylamide (2.0 M solution in heptane/THF/ethylbenzene, 3.4 mL, 6.8 mmol). The resulting reddish brown suspension was stirred for 23 min at -78 °C, and then a solution of 1,1-dimethylethyl 3-oxoazetidine-1 -carboxylate (770 mg, 4.5 mmol) in THF (10 mL) was added over 30 min by syringe pump. The mixture became a bright yellow solution as it was allowed to warm to room temperature over 3 hours. The mixture was quenched with saturated aqueous ammonium chloride. Water was added to dissolve precipitated salts, and the resulting mixture was extracted twice with ethyl acetate. The combined organic extracts were dried over magnesium sulfate, filtered, and concentrated. The residue was purified by column chromatography (60% ethyl acetate: 40% hexanes) to provide 1,1-dimethylethyl 3-[3,6-dioxo-l,4- bis(phenylmethy!)piperazin~2-yl]-3-hydroxyazeiidine-l-carboxylate as a colorless foam (1.04 g, 2.23 mmol, 50% yield). 'H NMR (400 MHz, CDC13): 7.39-7.29 (m, 7H), 7.23-7.19 (m, 3H), 5.34 (d, 1H), 4.82 (d, 1H), 4.58 (d, 1H), 4.37 (d, 1H), 4.37 (d, 1H), 4.22 (d, 1H), 4.15 (s, 1H), 4.08 (d, 1H), 3.97 (d, 1H), 3.75 (d, 1H), 3.74 (d, 1H), 3.67 (d, 1H), 3.64 (br s, 1H), 1.43 (s, 9H).
[00460] A solution of 1,1-dimethylethyl 3-[3,6-dioxo-l,4-bis(phenylmethyl)piperazin-2-y!]-3-hydroxyazetidine-1 -carboxylate (1.04 g, 2.2 mmol) in methanol (10 mL) was treated with hydrogen chloride in dioxane (4 N, 5.5 mL, 22 mmol) at 60 °C for 25 min. After cooling to room temperature the solution was concentrated. Ethyl acetate and 2 N hydrochloric acid were added to the residue and the phases were separated. The organic phase was discarded. The aqueous phase was basified with 5 M sodium hydroxide and the resulting solution was extracted 4 times with ethyl acetate. The combined organic extracts were dried over magnesium sulfate, filtered, and concentrated. The residue was purified by column chromatography (85% dichloromethane: 14% methanol: 1% aqueous ammonium hydroxide) to provide 3-(3-hydroxyazetidin~3-yl)-1,4-bis(phenylmethyl)piperazine- 2.5- dione as a colorless film (493 mg, 1.35 mmol, 61% yield). 'H NMR (400 MHz, CDC13): 7.39-7.28 (m, 6H), 7.25-7.20 (m, 4H), 5.39 (d, 1H), 4.80 (d, 1H), 4.44 (d, 1H), 4.36 (d, 1H), 4.26 (d, 1H), 4.11 (s, 1H), 3.97 (d, 1H), 3.83 (d, 1H), 3.71 (d, 1H), 3.27 (m, 2H); MS (El) for C21H23N3O3: 366 (Mif).
[00461] A solution 3-(3 -hydroxyazetidin-3 -yl)-1,4-bi s(phenylmethy!)piperazine- 2.5- dione (493 mg, 1.35 mmol) in ethyleneglycol dimethylether (12 mL) was treated with sodium borohydride (511 mg, 13.5 mmol) followed by slow addition of boron trifluoride-diethyl etherate. The reaction mixture was then heated to reflux for 3 hours. After cooling to 0 °C, methanol (17 mL) was added followed by careful addition of concentrated hydrochloric acid (7 mL). The resulting mixture was heated to reflux for 70 minutes. After cooling to room temperature, insoluble residue was removed by filtration. The filtrate was concentrated to an aqueous mixture of about 10 mL in volume. This mixture was cooled to 0 °C and was then basified to pH 10 with 5 M sodium hydroxide (approximately 17 mL). Dichloromethane (10 mL) was then added followed by di-feri-butyl dicarbonate (442 mg, 2.03 mmol). The mixture was warmed to room temperature and stirred for 15 minutes. The layers were separated and the aqueous phase was extracted twice with dichloromethane. The organic extracts were combined, dried over magnesium sulfate, filtered, and concentrated. The residue was purified by column chromatography (70% hexanes: 30% ethyl acetate) to provide 1,1-dimethylethyl 3-[ 1,4-bis(phenylmethyl)piperazin-2-y 1]-3-hydroxyazetidine-1-carboxylate as a white foam (408 mg, 0.93 mmol, 69% yield). 'H NMR (400 MHz, CDC13): 7.35-7.24 (m, 10H), 4.12 (br s, 1H), 3.88 (d, 1H), 3.78-3.65 (m, 4H), 3.53 (d, 1H), 3.43 (d, 1H), 3.21 (m, 1H), 2.80 (br s, 1H), 2.66 (m, 1H), 2.57-2.37 (m, 4H), 1.41 (s, 9H); MS (El) for ¢^35^()3: 438 (ΜΙ-Γ).
[00462] To a solution of 1,1-dimethylethyl 3-[ 1,4-bis(phenylmethyl)piperazin-2-yl]-3-hydroxyazetidine-1 -carboxylate (408 mg, 0.93 mmol) in methanol (15 mL) was added 10% palladium on carbon (wet), and the resulting suspension was subjected to an atmosphere of hydrogen for 21 hours. The catalyst was removed by filtration through celite, and the filter cake was rinsed with methanol. The combined filtrate was concentrated to provide 1,1-dimethylethyl 3 -hydroxy-3 -piperazin-2-ylazetidine-1-carboxylate as a brown syrup (227 mg, 0.88 mmol, 95% yield). *H NMR. (400 MHz, CDC13): 3.94-3.76 (m, 5H), 3.12 (m, 1H), 3.01 (m, 1H), 2.94-2.81 (m, 3H), 2.78-2.70 (m, 2H); MS (El) for C12H23N3O3: 258 (MH*).
[00463] To a solution of 1,1-dimethylethyl 3-hydroxy-3-piperazin-2-ylazetidine-1 -carboxylate (227 mg, 0.88 mmol) and N,IV-diisopropy lethylamine (436 pL, 2.64 mmol) in THF (5 mL) was added 2-nitrobenzenesulfonyl chloride (195 mg, 0.88 mmol). The mixture was stirred at room temperature for 2 hours. The solution was concentrated and the residue was purified by column chromatography (95% dichloromethane: 5% methanol) to provide 1,1-dimethylethyl 3 -hydroxy-3 -{4-[(2-nitrophenyl)sulfonyl]piperazin-2-yl} azetidine-1 -carboxylate as a white foam (308 mg, 0.70 mmol, 79% yield). 'H NMR (400 MHz, CDC13): 7.98 (m, 1H), 7.72 (m, 2H), 7.64 (m, 1H), 3.96 (d, 1H), 3.94 (d, 1H), 3.85 (d, 1H), 3.79 (d, 1H), 3.79-3.73 (m, 2H), 3.11 (m, 1H), 3.05 (dd, 1H), 3.00 (br s, 1H), 2.94 (dt, 1H), 2.78 (dt, 1H), 2.68 (dd, 1H), 1.45 (s, 9H).
[00464] To a solution of 1,1 -dimethylethyl 3-hydroxy-3- {4-[(2-nitrophenyl)sulfonyl]piperazin-2-yl}azetidine-1 -carboxylate (308 mg, 0.70 mmol) in methanol (10 mL) was added HC1 in dioxane (4 N, 1.75 mL, 7.0 mmol), and the mixture was heated to 60 °C for 30 minutes. The solution was concentrated to provide 3-{4-[(2-nitrophenyl)su!fonyI]piperazin-2-yl}azetidin-3-ol as a sticky white solid. This material was dissolved in dichloromethane (7 mL). To the solution was added N,N-di\sopropylethylamine (1.16 mL, 7.0 mmol) followed by 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]benzoyl fluoride (277 mg, 0.7 mmol), prepared using procedures similar to those described in Reference 1, and the resulting mixture was stirred at room temperature for 16 hours. The solution was concentrated and the residue was purified by column chromatography (95% dichloromethane: 5% methanol) to provide l-({3,4-difluoro-2-[(2-fluoro-4- iodophenyl)amino]phenyl}carbonyi)-3-{4-[(2-nitrophenyl)sulfonyl]piperazin-2~ yl}azetidin-3-ol as a pale yellow foam (453 mg, 0.63 mmol, 90% yield). 'H NMR . (400 MHz, CDCI3): 8.49 (s, 1H), 7.96 (dd, 1H), 7.71 (m, 2H), 7.53 (dd, 1H), 7.39 (dd, 1H), 7.33 (d, 1H), 7.15 (m, 1H), 6.84 (br s, 1H), 6.62 (m, 1H), 4.29-3.97 (br m, 4H), 3.79-3.62 (m, 3H), 3.26-2.99 (br m, 3H), 2.92-2.62 (br m, 3H); MS (El) for C26H23F3IN5O6S: 718 (MH+).
[00465] To a solution of 1-((3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-(4-[(2-nitrophenyl)sulfonyi]piperazin-2-yl}azeiidin-3-ol (139.4 mg, 0.19 mmol) in DMF (1 mL) was added potassium carbonate (79 mg, 0.57 mmol) and thiophenol (21 pL, 0.21 mmol). The mixture was stirred for 45 min at room temperature then quenched with water. The aqueous mixture was extracted twice with ethyl acetate, and the combined organic extracts were dried over magnesium sulfate, filtered, and concentrated. The residue was purified by preparative reverse phase HPLC to provide 1 -({3,4-diffuoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-piperazin-2-ylazetidin-3-ol as a white solid (26.8 mg, 0.05 mmol). !H NMR (400 MHz, CD3OD): 7.45 (dd, 1H), 7.36 (m, 1H), 7.32 (m, 1H), 7.03 (m, 1H), 6.62 (ddd, 1H), 4.51 (br dd, 1H), 4.31 (br dd, 1H), 4.17- 3.92 (m, 4H), 3.73-3.56 (m, 3H), 3.46 (br m, 1H), 3.26 (m, 1H); MS (El) for C20H20F3IN4O2: 533 (MH*). EXAMPLE 36 1,1-Dimethylethyl {(15)-l-[l-({4~[(2-fluoro~4-iodophenyI)amino}-l-methyl-6-oxo- l,6-dihydropyridazin-3-yl}carbonyI)-3-hydroxyazetidin-3-yI]ethyi}carbamate
[00466] To a suspension of 4-[(2-fluoro-4-iodophenyI)amino]~ 1 -methyl-6-oxo-1,6-dihydropyridazine-3-carboxylic acid (50 mg, 0.13 mmol) in DMF (2 mL), prepared using similar procedures to those described in Reference 4, at room temperature was added 1 -hydroxybenzotriazole (36.3 mg, 0.27 mmol) and 1 -(3-dimethylarninopropyl)- 3-ethylcarbodiimide hydrochloride (52 mg, 0.27 mmol) and the reaction was stirred for 2 hours, 1,1-Dimethylethyl [(15)-1 ~(3-hydroxyazetidin-3-yl)ethyl]carbamate (30 mg, 0.13 mmol), prepared using procedures similar to those in Example 28, and triethylamine (0.04 mL) were added and the mixture was stirred for 15 hours. The reaction mixture was partitioned between saturated sodium chloride and ethyl acetate. The organic layer was washed with 5% lithium chloride solution, saturated sodium bicarbonate, dried over anhydrous sodium sulfate, filtered and concentrated in vacuo to give crude product as yellow oil. The oil was purified by column chromatography (silica gel, ethyl acetate) to afford 1,1-dimethylethyl ((lS)-l-[l-({4~[(2-fluoro-4-iodophenyl)amino]-1 -methyl-6-oxo-l ,6-dihydropyridazin-3-yl} carbonyl)-3-hydroxyazetidin-3-yl]ethyl}carbamate as a yellow oil (55 mg, 73% yield): 'Η NMR (400 MHz, CDCls): 10.24-10.23 (m, 1H), 7.52-7.50 (m, 2H), 7.12-7.07 (m, 1H), 6.10- 6.09 (m, 1H), 5.13-5.09 (m, 1H), 4.91-4.82 (m, 1H), 4.60-4.39 (m, 2H), 4.10-4.08 (m, 1H), 4.00-3.87 (m, 2H), 3.70 (d, 3H), 1.43 (s, 9H), 1.24-1.20 (m, 3H); MS (El) for C22H27FIN5O5: 588 (MH+). (00467] Using the same or analogous synthetic techniques and substituting, as necessary, with alternative reagents, the following compounds of the invention were prepared: EXAMPLE 36(a). 1,1-Dimethylethyl {(lS)-l-[l-((5-[(4-bromo-2-chlorophenyl)amino] -4-fluoro-1 -methyl-1 if-benzimidazol-6-yl} carbonyl)-3-hydroxyazetidin-3-yljethyl}carbamate: JHNMR (400 MHz, CDCI3): 7.95 (s, 1H), 7.45-7.44 (m, 1H), 7.33-7.27 (m, 2H), 7.15-7.12 (m, 1H), 6.50-6.47 (m, 1H), 4.82- 4.74 (m, 1H), 4.17-3.92 (m, 4H), 3.86 (s, 3H), 3.74-3.60 (m, 1H), 1.40 (s, 9H), 1.11- 1.06 (m, 3H). MS (El) for CisHsgBrClFNsO-}: 598 (MH4) with a chloro, bromo isotope pattern. EXAMPLE 36(b). 1,1-Dimethylethyl (2S)-2-[l-({5-[(4-bromo-2-chlorophenyl)amino] -4-fl uoro-1-methyl-1 //-benzimidazol-6-yl} carbonyl)~3 -hydroxyazetidin-3-yl]piperidine-l-carboxvlate: MS (El) for C28H32BFCIFN5Q4: 638 (MH4) with a chloro, bromo isotope pattern.
Example 37 6-({3-[(lS)-l-aminoethyl]-3-hydroxyazetidin-l-yl}carbonyi)-5-[(2-fluoro-4-iodophenyl)ammo]-2-methylpyridazm~3(2/f)~oiie acetate salt
[00468] 1,1 -Dimethylethyl ((15)-1-(1-(( 4-[(2-fluoro~4-iodophenyl)amino]-1 - methyl-6-oxo-1,6-dihydropyridazin-3-yl} carbonyl)~3 -hydroxyazetidin-3 - yl]ethyl}carbamate (55 mg, 0.09 mmol), prepared using procedures similar to those described in Example 36, was taken up in methanol (2 mL) and hydrochloric acid (4N in dioxane, 1 mL, 4 mmol) was added and the reaction was stirred at 60 °C for 2 hours. The reaction mixture was concentrated in vacuo and was purified by reverse-phase HPLC followed by lyophilization of the pure fractions to afford 6-({3-[(lS)-l-aminoethyl]-3-hydroxyazetidin-1 -yl} carbonyl)-5-[(2-fluoro-4-iodophenyl)amino]-2-methylpyridazin-3(2//)-one acetate as yellow solid (40 mg, 87%). *H NMR (400 MHz, CDC13): 10.17 (d, 1H), 7.52-7.46 (m, 2H), 7.09 (t, 1H), 6.13-6.12 (m, 1H), 4.51-4.48 (m, 2H), 4.18-4.03 (m, 2H), 3.73 (d, 3H), 3.35-3.28 (m, 1H), 3.22-2.80 (br, 3H), 1.21-1.19 (m, 3H); MS (El) for Ci7Hi9FIN5Q3: 488 (MH+). (00469] Using the same or analogous synthetic techniques and/or substituting with alternative reagents, the following compounds of the invention were prepared: Example 37(a). 3-[(15)-1 -Aminoethyl]-1-((5-[(4-bromo-2-chlorophenyl)amino]-4-fluoro-1 -methyl-li/-benzimidazol~6-yl}carbonyl)azetidin-3-ol hydrochloride. MS (El) for C2oH2oBrClFN502: 498 (MH+) with a chloro, bromo isotope pattern Example 37(b). l-({5-[(4-Bromo~2-chlorophenyl)amino]-4-fSuoro-l-methyl-IH-benzimidazol-6-yl}carbonyl)-3~[(2S)-piperidin-2-yl]azetidin-3-ol hydrochloride. rH NMR (400 MHz, CD3OD): 9.42 (s, 1H), 7.97-7.96 (m, 1H), 7.57 (s, 1H), 7.30-7.27 (m, 1H), 6.70-6.66 (m, 1H), 4.60-4.55 (m, 1H), 4.28 (t, 1H), 4.19 (s, 3H), 4.13-3.98 (m, 2H), 3.38-3.32 (m, 2H), 3.00 (t, 1H), 1.86-1.30 (m, 6H). MS (El) for C23H24BrClFN502. HC1: 538 (MH+) with a chloro, bromo isotope pattern EXAMPLE 38 l-({3-[(2-fluoro-4-iodophenyl)ammo]pyridin-4~yl}carbonyl)-3-((2/5)-piperidm~2- yljazetidin-3-ol
[00470] 3-[(2-Fluoro-4-iodophenyl)amino]pyridine-4-carboxylic acid (200 mg, 0.559 mmol), prepared using procedures similar to those described in WO 2006/045514, was suspended in DMF (7 mL) and 1-hydroxybenzotriazole (151 mg, 1.12 mmol) and 1 -(3 -dimethylaminopropy 1)-3 -ethylcarbodiimide hydrochloride (214 mg, 1.12 mmol) were added. The mixture was stirred at ambient for 10 minutes and then triethylamine (0.078 mL, 0.559 mmol) was added. After a further 20 minutes, 1.1- dimethylethyl (25)-2-(3-hydroxyazetidin~3-yl)piperidine-1 -carboxylate (143 mg, 0.559 mmol), prepared using similar procedures to those described in Example 22(a) and 22(b), and triethylamine (0.16 mL, 1.15 mmol) were added and the mixture was stirred for 15 hours. The mixture was partitioned between ethyl acetate and saturated ammonium chloride. The organic portion was washed with 5% lithium chloride and twice with saturated sodium bicarbonate, then was dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. The residue was purified by column chromatography (silica gel, 60-80% ethyl acetate in hexanes) to give 1,1-dimethylethyl (2S)-2-[I-({3-[(2-fluoro-4-iodophenyl)amino]pyridin-4-yl}carbonyl)-3-hydroxyazetidin-3-yl]piperidine-1 -carboxylate (368 mg, 0.587 mmol, 74% yield): 'H NMR (400 MHz, CDC13): 8.73 (br m, 1H), 8.62 (br s, 1H), 8.14 (d, 1H), 7.47 (dd, 1H), 7.43-7.39 (in, 1H), 7.20-7.12 (m, 2H), 4.38-4.21 (m, 2H), 4.16-4.01 (m, 2H), 4.01- 3.88 (m, 1H), 3.44-3.30 (m, 1H), 2.98-2.83 (m, 1H), 2.00-1.88 (m, 1H), 1.71-1.50 (m, 6H), 1.44 (s, 9H); MS (El) for C25H30FIN4O4: 597 (ΜΗΓ).
[00471] 1,1-Dimethylethyl (25)-2- [l-({3-[(2~f!uoro-4-iodophenyl)amino]pyridin-4-yl }carbonyl)-3-hydroxyazeti din-3-yljpiperi dine-1-carboxylate (24 mg, 0.040 mmol) was dissolved in methanol (2 mL) and treated with 4 N hydrochloric acid in dioxane (0.25 mL, 1 mmol) at reflux for 20 minutes. The mixture was concentrated in vacuo and was purified by reverse-phase HPLC followed by lyophilization of the pure fractions to afford l-({3-[(2-fluoro-4-iodophenyl)amino]pyridin~4-yl}carbonyl)~3-[(2S)-piperidin-2-yl]azetidin-3-ol acetate (14 mg, 0.025 mmol, 63% yield): !H NMR (400 MHz, dg-DMSO): 8.62 (br s, 1H), 8.46 (s, 1H), 8.18 (dd, 1H), 7.65 (dd, 1H), 7.45 (d, 1H), 7.37 (t, 1H), 7.16-7.08 (m, 1H), 4.25 (dd, 1H), 4.04 (dd, 1H), 3.90 (t, 1H), 3.70 (d, 1H), 2.95 (br d, 1H), 2.52-2.42 (m, 2H), 1.78-1.68 (m, 1H), 1.57 (br t, 1H), 1.47 (br d, 1H), 1.35-1.13 (m, 2H), 1.10-0.96 (m, 1H); MS (El) for C20H22FIN4O2: 497 (MH+). EXAMPLE 39 l-{{3-[{2-fluoro-4-iodophenyl)aminoj-l-oxidopyridin-4~yl}carb0Byl)”3"[{25)- piperidi n-2-y I] azetidin -3-ol
[00472] 1,1 -Dimethylethyl (25)-2~[l-({3-[(2-fluoro-4-iodophenyl)amino]pyridin-4-yf }carbonyl)-3-hydroxyazetidin-3-yl]piperidine-l-carboxylate (80 mg, 0.134 mmol), prepared using procedures similar to those described in Example 38, was dissolved in dichloromethane (3 mL) and treated with 3-chloroperoxybenzoic acid (73% pure; 32 mg, 0.135 mmol) at ambient for 7 hours. 3-chloroperoxybenzoic acid (73% pure; 32 mg, 0.135 mmol) was added and the mixture was stirred for 15 hours. The mixture was purified by column chromatography (silica gel, 0-10% ethanol in ethyl acetate) to give 1,1 -dimethylethyl (2S)-2-[ 1 -({3-[(2-fluoro-4-iodophenyl)amino]-l-oxidopyridin- 4-yl} carbonyl)-3-hydroxyazetidin-3-yl]piperidine-1 -carboxylate (57 mg, 0.093 mmol, 69% yield): !H NMR (400 MHz, CDC13): 9.38 (s, 1H), 8.00 (s, 1H), 7.68 (dd, 1H), 7.51 (dd, 1H), 7.46 (d, 1H), 7.19 (br d, 1H), 7.09 (t, 1H), 5.78 (br, 1H), 4.44-3.98 (m, 3H), 3.98-3.87 (m, 1H), 3.49-3.39 (m, 1H), 3.07-2.88 (m, 1H), 2.01-1.91 (m, 1H), 1.70-1.47 (m, 6H), 1.45 (s, 9H); MS (El) for C25H30FIN4O5: 613 (MH*).
[00473] 1,1-Dimethylethyl (2S)-2-[1 -({3-[(2-fluoro-4-iodophenyl)amino]-l-oxidopyridin-4-yl}carbonyl)-3-hydroxyazetidin-3-yl]piperidine-l-carboxylate (57 mg, 0.093 mmol) was dissolved in methanol (2 mL) and treated with 4N hydrochloric acid in dioxane (0.25 mL, 1 mmol) at 50 °C for 2.25 hours. The mixture was concentrated in vacuo and was purified by reverse-phase HPLC followed by lyophilization of the pure fractions to afford l-({3-[(2-fluoro-4-iodophenyl)amino]-l-oxidopyridin~4-yl}carbonyl)-3-[(2S)-piperidin-2~yrjazetidin-3~ol acetate (35 mg, 0.061 mmol, 66% yield): *H NMR (400 MHz, d6-DMSO): 7.83 (s, 1H), 7.72 (dt, 2H), 7.55-7.51 (m, 1H), 7.47-7.41 (m, 1H), 7.24 (t, 1H), 4.45-4.32 (m, 1H), 4.14-3.95 (m, 2H), 3.72 (d, 1H), 2.97 (d, 1H), 2.58-2.43 (m, 2H), 1.80-1.73 (m, 1H), 1.67-1.55 (m, 1H), 1.49 (br d, 1H), 1.38-1.16 (m, 2H), 1.16-1.01 (m, 1H); MS (El) for C20H22FIN4O3: 513 (MH+). EXAMPLE 40 l”({3,4-difluoro-2-[(2-fluoro-4-iodophenyI)amma]phenyl}carbonyI)~3~[(l$)-l- (methylamino)ethyI]azetidin-3-ol
[00474] To 3-[( 1S)-1 -aminoethyl]-1 -((3,4-difluoro-2~[(2-iluoro~4-iodophenyl)amino]phenyl}carbonyl)azetidin-3~o! (87.4 mg, 0.18 mmol), prepared using similar procedures to those described in Example 28, was added formaldehyde (37% aqueous, 14 mg, 0.18 mmol) in methanol (2 mL) and sodium borohydride (7 mg, 0.18 mmol). The mixture was stirred for 3 h at rt, after which sodium borohydride (16 mg, 0.42 mmol) was added. Upon stirring an additional 1.25 h, more formaldehyde (37% aqueous, 1 drop) was added, and the mixture was stirred 3 days at rt. A further small spatula (~50 mg) of sodium borohydride was then added, and the mixture was stirred at rt for 30 min. After quenching with 1 N HC1, the reaction mixture was purified directly by preparative HPLC. The clean material was converted to its hydrochloride salt to provide 1-((3,4-difluoro-2-[(2-fluoro~4-iodophenyl)amino]phenyl} carbony!)-3 - [(1S)-1 -(methylamino)ethy 1 ]azetidin-3 -ol as a yellow solid (21.7 mg, 0.040 mmol, 22% yield). SH NMR (400 MHz, CD3OD) δ 7.47 (dd, 1H), 7.36 (d, 1H), 7.31 (m, 1H), 7.06 (q, 1H), 6.62 (dt, 1H), 4.36 (dd, 1H), 4.21-3.91 (m, 3H), 3.44 (q, 1H), 2.66 (s, 3H), 1.29 (br m, 3H); MS (El) for Ci9H.!9F3lN302: 506 (MIT1).
Biological Example 1
Biochemical Assay [00475] For a biochemical measurement of MEK1 inhibitory activity, compounds of the invention were screened in a triple coupled cRaf-MEK-ERJC2 assay using ALPHASCREEN (Registered Trademark of Perkin Elmer) technology (Perkin Elmer). The compound of the invention, 0.5 pL of 100% DMSO stock solution, is diluted into an assay buffer composed of 20 mM Tris (pH ::: 7.5), 10 mM magnesium chloride, 0.03% CHAPS and 1 mM DTT. Subsequently, 10 pL of substrate mixture is added composed of unactive MEK1 (3 nM), ATP (50 pM), unactive ERK2 (4 nM),
biotinylated MBP peptide (b-FFKNIVTPRTPPPSQGK, 1 μΜ) and antiphospho MBP peptide (0.5 nM). The mixture is then gently shaken for 30 minutes at room temperature followed by addition of active cRaf (5 pL at 0.5 nM) to initiate reaction. The mixture is then shaken for 100 minutes at room temperature then quenched by addition of 10 pL of a mixture of 5pg/mL streptavidin donor beads and 5 pg/mL protein A acceptor beads in detection buffer (75 mM Hepes pH = 7.5, 300 mM sodium chloride, 120 mM EDTA, 0.3% BSA and 0.03% Tween), followed by incubation overnight and signal detection on an ALPHAQuest® (Registered Trademark of Perkin Elmer) plate reader (Perkin Elmer).
[00476] Compounds of the invention are inhibitors of MEK. The extent to which these compounds are MEK inhibitors can be determined by one of ordinary skill in the art. In particular, the compounds can be tested in the assay described in Biological Example 1. When tested in that assay, compounds of the invention demonstrated the ability to bind to MEK. In one embodiment of the invention, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 4 μΜ or less. In another embodiment, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 3 μΜ or less. In another embodiment, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 2 μΜ or less. In another embodiment, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 1.6 μΜ or less. In another embodiment, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 1 μΜ or less. In another embodiment, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 0.7 μΜ or less. In another embodiment, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 0.3 μΜ or less. In another embodiment, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 0.2 μΜ or less. In another embodiment, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 0.1 μΜ or less. In another embodiment, the MEK inhibitor is selected from the compounds in Table 1 having a MEK-binding affinity of about 0.05 μΜ or less.
Biological Example 2
Endogenous ERK Phosphorylation ELISA Assay [00477] MDA-MB-231T (ATCC), Calu-6 (ATCC), HCT 116 (ATCC), A2058 (ATCC), and A375 (ATCC) cells were seeded at 20000, 30000, 30000, 20000, and 30000 cells/well, respectively, onto black 96-well microtiter plates (Costar 3904), in DMEM (Cellgro) containing 10% FBS (Heat-Inactivated, Cellgro), 1% NEAA (Cellgro), and 1% Pen/Strep (Cellgro). SK-MEL-28 (ATCC) cells were seeded at 20000 cells/well in MEM (ATCC) containing 10% FBS (Heat-Inactivated, Cellgro), and 1% Pen/Strep (Cellgro). The cells were then incubated at 37°C, 5% CO2 for 24 h. Serum starvation was performed by replacing the medium with serum-free DMEM or MEM for an additional 24 h. Serial dilutions of test compounds in fresh serum-free medium in a final concentration of 0.3% DMSO (vehicle) were added to the cells and incubated for 1 h. Negative control wells were in serum-free medium + 0.3% DMSO only. After treatment, the medium was removed and cells were fixed with 4% formaldehyde, followed by quenching of endogenous peroxidases with 0.6% H2O2. Plates were then blocked (10% FBS, Cellgro) and incubated with mouse monoclonal anti-phospho-p44/42 ΜΑΡΚ, E10 (1:2000, Cell Signaling), followed by secondary antibody (HRP-conjugated, goat anti-mouse IgG, 1:3000 from Jackson ImmunoResearch Laboratories, Inc). Washing of the plates was performed with PBS-T (0.1% Triton X-100) in between all incubation steps. A luminol-based substrate solution was then added and plates read using the Victor Wallac machine. IC50 values were determined based on total ERK phosphorylation with compound treatment versus total ERK phosphorylation with 0.3% DMSO treatment alone.
Biological Example 3 BrdU Cell Proliferation Assay [00478] MDA-MB-231T (ATCC), Calu-6 (ATCC), HCT 116 (ATCC), A2058 (ATCC), A375 (ATCC), and Colo-205 (ATCC) cells were plated at densities of 2500, 3500, 3500, 2500, 3500, and 15000 cells/well onto 96-well microtiter plates (Cat# 3904, Costar), in DMEM (Cellgro) containing 10% FBS (Heat Inactivated, Cellgro), 1% Pen/Strep (Cellgro), and 1% NEAA (Cellgro). SK MEL-28 (ATCC) and WM-266-4 (ATCC) were plated at densities of 2000 and 6000 cells/well in MEM (ATCC) containing 10% FBS (Heat-Inactivated, Cellgro), and 1% Pen/Strep (Cellgro). The cells were incubated overnight at 37°C, 5% CO2 for 18 h. The next day, ceils were treated with a serial dilution of compound in medium (containing a final concentration of 0.3% DMSO). Triplicate wells were used for each compound concentration. The control wells received 0.3% DMSO media. The cultures were incubated at 37°C, 5% CO2 for an additional 48 h. The cells were assayed for proliferation according to the “Cell Proliferation ELISA, Bromo Deoxyuridine (BrdU) (chemiluminescence) kit” from Roche. The cells were treated with the BrdU labeling solution and then fixed with FixDenat solution. Anti-BrdU-POD (PerOxiDase) conjugate was added to the cells, after which the plates were washed 3x with IX PBS. Substrate solution was added, and the plates were read for luminescence using the Victor Wallac machine. IC50 values were calculated based on the cell proliferation with compound treatment compared to the vehicle control.
Biological Example 4 In vivo mouse models [00479] The ability of a MEK inhibitor, administered as monotherapy (i.e. not in combination with another cancer treatment), to inhibit the growth of the following tumors in mice was examined. The models can also be used by one of ordinary skill in the art to determine the desirability of a particular combination of a MEK inhibitor with another cancer treatment.
[00480] Female athymic nude mice (NCr) 5-8 weeks of age and weighing approximately 20g were purchased from Taconic (Germantown, NY). Prior to initiation of a study, the animals were allowed to acclimate for a minimum of 48 h. During these studies, animals were provided food and water ad libitum and housed in a room conditioned at 70-75°F and 60% relative humidity. A 12 h light and 12 h dark cycle was maintained with automatic timers.
[00481] Colo-205 human colorectal carcinoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), Penicillin-Streptomycin and non-essential amino acids at 37 °C in a humidified, 5% CO2 atmosphere. On day 0, cells were harvested by trypsinization, and 3xl06 cells (passage #3, 92% viability) in 0.1 ml ice-cold Hank’s balanced salt solution were implanted intradermally in the hind-flank of 5-8 week old female athymic nude mice.
[00482] A375 human melanoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), Penici 11 in-Streptomycin and non-essential amino acids at 37 °C in a humidified, 5% CO2 atmosphere. On day 0, cells were harvested by trypsinization, and 5xl06 cells (passage #8, >99% viability) in 0.1 mL ice-cold Hank’s balanced salt solution were implanted intradermally in the hind-flank of 5-8 week old female athymic nude mice.
[00483] A2058 human melanoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), Penicillin-Streptomycin and non-essential amino acids at 37 °C in a humidified, 5% CO2 atmosphere. On day 0, cells were harvested by trypsinization, and 3x 106 cells (passage #5, 80% viability) in 0.1 mL ice-cold Hank’s balanced salt solution were implanted intradermally in the hind-flank of 5-8 week old female athymic nude mice. [00484J MDA-MB-231 human breast adenocarcinoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), Penicillin-Streptomycin and non-essential amino acids at 37 °C in a humidified, 5% CO2 atmosphere. On day 0, cells were harvested by trypsinization, and 1x106 cells (passage #6, >99% viability) in 0.1 mL ice-cold Hank’s balanced salt solution were implanted subcutaneously into the mammary fat pad of 5-8 week old female athymic nude mice.
[00485] Calu-6 human lung anaplastic carcinoma cells were cultured in vitro in DMEM (Mediatech) supplemented with 10% Fetal Bovine Serum (Hyclone), Penicillin-Streptomycin and non-essential amino acids at 37 °C in a humidified, 5% CO2 atmosphere. On day 0, celis were harvested by trypsinization, and 5x106 cells (passage #8, 96% viability) in 0.1 mL ice-cold Hank’s balanced salt solution were implanted intradermally in the hind-flank of 5-8 week old female athymic nude mice.
[00486] For subcutaneous or intradermal tumors, the mean tumor weight of each animal in the respective control and treatment groups was determined twice weekly during the study. Tumor weight (TW) was determined by measuring perpendicular diameters with a caliper, using the following formula: tumor weight (mg) = [tumor volume = length (mm) x width2 (mm2)]/2.
[00487] Percent inhibition of tumor growth (TGI) is determined with the following formula:
where Xq = average TW of all tumors on group day; Xf = TW of treated group on Day f; Yf = TW of vehicle control group on Day f [00488] If tumors regress below their starting sizes, then the percent tumor regression is determined with the following formula:
TGI is calculated individually for each tumor to obtain a mean ± SEM value for each experimental group. Statistical significance is determined using the 2-tailed Student’s t-test (significance defined as P<0.05).
Biological Example S WM-266-4 Human Melanoma Xenograft Model [00489] The WM-266-4 human melanoma cell line is PTEN-deficient, and harbors a heterozygous activating mutation in the gene encoding B-Raf. Therefore, the ability of a MEK inhibitor administered as monotherapy, and in combination with the mTOR inhibitor rapamycin, was examined to inhibit the growth of WM-266-4 xenograft tumors in nude mice.
[00490] Tumors were established in female nude mice and staged when the tumors reached 112 ±6 mg. The MEK compound was dosed orally (in the morning) at 10 mg/kg qd and rapamycin dosed intraperitoneally (in the afternoon, ~7 h after the morning dose) at 5 mg/kg qd were administered as single agents or in combination. The MEK compound administered as monotherapy caused significant tumor growth inhibition Coadministration of rapamycin and the MEK compound resulted in efficacy significantly superior (p < 0.001) to that achieved with either agent given alone (95% TGI, compared with TGI of 60% and 82%).
Summary of Growth Inhibition of WM-266-4 Tumors by a MEK
Inhibitor Administered Alone or in Combination with Rapamycin
“TGI, tumor growth inhibition: bIP, administered intraperitoneally.
Pharmaceutical Composition Examples [00491] The following are representative pharmaceutical formulations containing a compound of Formula I.
Tablet Formulation
The following ingredients are mixed intimately and pressed into single scored tablets.
Ingredient Quantity per tablet, mg compound of this invention 400
Cornstarch 50 croscarmellose sodium 25
Lactose 120 magnesium stearate _ 5_____
Capsule Formulation
The following ingredients are mixed intimately and loaded into a hard-shell gelatin capsule.
Ingredient Quantity per tablet, mg compound of this invention 200 lactose, spray-dried 148 magnesium stearate__2____
Suspension Formulation
The following ingredients are mixed to form a suspension for oral administration. Ingredient Amount compound of this invention_1,0 g _
Ingredient Amount fumaric acid 0.5 g sodium chloride 2.0 g methyl paraben 0.15 g propyl paraben 0.05 g granulated sugar 25.5 g sorbitol (70% solution) 12.85 g
Veegum K (Vanderbilt Co.) 1,0 g
Flavoring 0.035 mL
Colorings 0.5 mg distilled water_q.s. to 100 mL_
Injectable Formulation
The following ingredients are mixed to form an injectable formulation.
Ingredient Amount compound of this invention 1.2 g
sodium acetate buffer solution 0.4 M 2.0 mL
HC1 (1 N) or NaOH (1 M) q.s. to suitable pH water (distilled, sterile) q.s.to 20 mL_______ [00492] All of the above ingredients, except water, are combined and heated to 60-70.degree. C. with stirring. A sufficient quantity of water at 60.degree. C. is then added with vigorous stirring to emulsify the ingredients, and water then added q.s. to 100 g.
Suppository Formulation [00493] A suppository' of total weight 2.5 g is prepared by mixing the compound of the invention with Witepsol.RTM. H-15 (triglycerides of saturated vegetable fatty acid; Riches-Nelson, Inc., New York), and has the following composition:
Ingredient Quantity per tablet, mg compound of this invention 500
Witepsol® H-l_5_ balance______ [00494] The foregoing invention has been described in some detail by way of illustration and example, for purposes of clarity and understanding. The invention has been described with reference to various embodiments and techniques. However, it should be understood that many variations and modifications may be made while remaining within the spirit and scope of the invention. It will be obvious to one of skill in the art that changes and modifications may be practiced within the scope of the appended claims. Therefore, it is to be understood that the above description is intended to be illustrative and not restrictive. All patents, patent applications and publications cited in this application are hereby incorporated by reference in their entirety for all purposes to the same extent as if each individual patent, patent application or publication were so individually denoted.
[00495] The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.
[00496] Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but not the exclusion of any other integer or step or group of integers or steps.

Claims (4)

  1. THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:
    1. A method of treating breast cancer which method comprises administering to a patient in need thereof a therapeutically effective amount of the compound l-({ 3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(2S)-piperidin-2-yl]azetidin-3-ol
    or a pharmaceutically acceptable salt thereof in combination with a taxane.
  2. 2. The method according to claim 1 wherein the taxane is paclitaxel.
  3. 3. Use of a therapeutically effective amount of the compound l-({3,4-difluoro-2-[(2-fluoro-4-iodophenyl)amino]phenyl}carbonyl)-3-[(2S)-piperidin-2-yl]azetidin-3-ol or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of breast cancer, wherein the compound is administered in combination with a taxane.
  4. 4. Use according to claim 3, wherein the taxane is paclitaxel.
AU2013204026A 2006-12-14 2013-04-11 Methods of using MEK inhibitors Active AU2013204026B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013204026A AU2013204026B2 (en) 2006-12-14 2013-04-11 Methods of using MEK inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60/875,412 2006-12-14
AU2007334402A AU2007334402B2 (en) 2006-12-14 2007-12-14 Methods of using MEK inhibitors
AU2013204026A AU2013204026B2 (en) 2006-12-14 2013-04-11 Methods of using MEK inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2007334402A Division AU2007334402B2 (en) 2006-12-14 2007-12-14 Methods of using MEK inhibitors

Publications (2)

Publication Number Publication Date
AU2013204026A1 AU2013204026A1 (en) 2013-05-16
AU2013204026B2 true AU2013204026B2 (en) 2017-01-19

Family

ID=48450637

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013204026A Active AU2013204026B2 (en) 2006-12-14 2013-04-11 Methods of using MEK inhibitors

Country Status (1)

Country Link
AU (1) AU2013204026B2 (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050153962A1 (en) * 2003-12-08 2005-07-14 Nettekoven Matthias H. Thiazole derivatives

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050153962A1 (en) * 2003-12-08 2005-07-14 Nettekoven Matthias H. Thiazole derivatives

Also Published As

Publication number Publication date
AU2013204026A1 (en) 2013-05-16

Similar Documents

Publication Publication Date Title
JP5311673B2 (en) Method of using MEK inhibitor
EP1934174B1 (en) Azetidines as mek inhibitors for the treatment of proliferative diseases
JP2010513288A5 (en)
WO2008124085A2 (en) Methods of using combinations of mek and jak-2 inhibitors
AU2013204026B2 (en) Methods of using MEK inhibitors
AU2015255183C1 (en) Azetidines as MEK inhibitors for the treatment of proliferative diseases
AU2012261703B2 (en) Azetidines as MEK inhibitors for the treatment of proliferative diseases
AU2013203939B2 (en) Azetidines as MEK inhibitors for the treatment of proliferative diseases

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)