Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
DUAL INHIBITORS OF SOLUBLE EPOXIDE HYDROLASE AND 5-LIPOXYGENASE
Document Type and Number:
WIPO Patent Application WO/2021/214048
Kind Code:
A1
Abstract:
The invention pertains to a novel structure (I) that provides an activity as a dual inhibitor of the enzymes soluble epoxide hydrolase (sEH) and 5-lipoxygenase (5-LOX). The invention pertains to multiple derivatives of the new class of dual inhibitors, their application in medicine, pharmaceutical compositions comprising them as well as to methods for synthesizing the new compounds.

Inventors:
PROSCHAK EUGEN (DE)
HIESINGER KERSTIN (DE)
STEINHILBER DIETER (DE)
HOFMANN BETTINA (DE)
Application Number:
PCT/EP2021/060226
Publication Date:
October 28, 2021
Filing Date:
April 20, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
JOHANN WOLFGANG GOETHE UNIV FRANKFURT AM MAIN (DE)
International Classes:
C07C275/20; A61P19/00; A61P29/00
Foreign References:
US5814648A1998-09-29
US5698155A1997-12-16
Other References:
BROOKS C D W ET AL: "(R)-(+)-N-[3-[5-[(4-Fluorophenyl)methyl]-2-thienyl]-1-methyl-2-propynyl]-N-hydroxyurea (ABT-761), a Second Generation 5-Lopxygenase Inhibitor", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 38, 1 January 1995 (1995-01-01), pages 4768 - 4775, XP002351353, ISSN: 0022-2623, DOI: 10.1021/JM00024A004
ULRIKE GARSCHA ET AL: "Pharmacological profile and efficiency in vivo of diflapolin, the first dual inhibitor of 5-lipoxygenase-activating protein and soluble epoxide hydrolase", SCIENTIFIC REPORTS, vol. 7, no. 1, 24 August 2017 (2017-08-24), XP055725888, DOI: 10.1038/s41598-017-09795-w
HIESINGER KERSTIN ET AL: "Development of multitarget agents possessing soluble epoxide hydrolase inhibitory activity", PROSTAGLANDINS AND OTHER LIPID MEDIATORS, vol. 140, 2019 - 26 December 2018 (2018-12-26), pages 31 - 39, XP085576310, ISSN: 1098-8823, DOI: 10.1016/J.PROSTAGLANDINS.2018.12.003
HIESINGER KERSTIN ET AL: "Design, Synthesis, and Structure-Activity Relationship Studies of Dual Inhibitors of Soluble Epoxide Hydrolase and 5-Lipoxygenase", JOURNAL OF MEDICINAL CHEMISTRY, vol. 63, no. 20, 12 October 2020 (2020-10-12), US, pages 11498 - 11521, XP055805047, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.0c00561
SHEN, H. C.HAMMOCK, B. D.: "Discovery of Inhibitors of Soluble Epoxide Hydrolase: A Target with Multiple Potential Therapeutic Indications", J. MED. CHEM., vol. 55, no. 5, 2012, pages 1789 - 1808, XP055190495, DOI: 10.1021/jm201468j
NEWMAN, J. W. ET AL.: "Epoxide Hydrolases: Their Roles and Interactions with Lipid Metabolism", PROG. LIPID RES, vol. 44, no. 1, 2005, pages 1 - 51, XP004769681, DOI: 10.1016/j.plipres.2004.10.001
"Imig, J. D. Epoxides and Soluble Epoxide Hydrolase in Cardiovascular Physiology", PHYSIOL. REV, vol. 92, no. 1, 2012, pages 101 - 130
HUANG, H.WENG, J.WANG, M.-H: "EETs/sEH in Diabetes and Obesity-Induced Cardiovascular Diseases", PROSTAGLANDINS OTHER LIPID MEDIAT, vol. 125, 2016, pages 80 - 89, XP029736012, DOI: 10.1016/j.prostaglandins.2016.05.004
"UnitProt", Database accession no. P09917
BROOKS, C. D.STEWART, A. O.BASHA, A.BHATIA, P.RATAJCZYK, J. D.MARTIN, J. G.CRAIG, R. A.KOLASA, T.BOUSKA, J. B.LANNI, C: "R)-(+)-N-[ -[ -[( -Fluorophenyl)Methyl]-2-Thienyl]-i-Methyl- -Propynyl]-N-Hydroxyurea (ABT- 6 ), a Second-Generation -Lipoxygenase Inhibitor", J. MED. CHEM., vol. 38, no. 24, 1995, pages 4768 - 4775, XP002351353, Retrieved from the Internet DOI: 10.1021/jm00024a004
"Handbook of Pharmaceutical Excipients", 1986, AMERICAN PHARMACEUTICAL ASSOCIATION
STEWART, A.O.BROOKS, D. W.N,O-BIS: "phenoxycarbonyl)hydroxylamine: a new reagent for the direct synthesis of substituted N-hydroxyureas", J. ORG. CHEM., vol. 57, 1992, pages 5020 - 5023, XP055003256, Retrieved from the Internet DOI: 10.1021/jo00044a046
CHEN, L.DENG, H.CUI, H.FANG, J.ZUO, Z.DENG, J.LI, Y.WANG, X.ZHAO, L: "Inflammatory Responses and Inflammation-Associated Diseases in Organs", ONCOTARGET, vol. 9, no. 6, 2018, pages 7204 - 7218, Retrieved from the Internet
RADMARK, O.WERZ, O.STEINHILBER, D.SAMUELSSON, B: "Lipoxygenase, a Key Enzyme for Leukotriene Biosynthesis in Health and Disease", BIOCHIM. BIOPHYS. ACTA, vol. 1851, no. 4, 2015, pages 331 - 339, Retrieved from the Internet
HAEGGSTROM, J. Z: "Leukotriene Biosynthetic Enzymes as Therapeutic Targets", J. CLIN. INVEST, vol. 128, no. 7, 2018, pages 2680 - 2690, Retrieved from the Internet
HIESINGER, KWAGNER, K. M.HAMMOCK, B. D.PROSCHAK, E.HWANG, S. H: "Development of Multitarget Agents Possessing Soluble Epoxide Hydrolase Inhibitory Activity", PROSTAGLANDINS OTHER LIPID MEDIAT, vol. 140, 2019, pages 31 - 39, XP085576310, Retrieved from the Internet DOI: 10.1016/j.prostaglandins.2018.12.003
HOXHA, M.ZAPPACOSTA, B: "CYP-Derived Eicosanoids: Implications for Rheumatoid Arthritis", PROSTAGLANDINS OTHER LIPID MEDIAT, vol. 146, 2020, pages 106405, Retrieved from the Internet
WAGNER, K. M.MCREYNOLDS, C. B.SCHMIDT, W. K.HAMMOCK, B. D: "Soluble Epoxide Hydrolase as a Therapeutic Target for Pain, Inflammatory and Neurodegenerative Diseases", PHARMACOL. THER, vol. 180, 2017, pages 62 - 76, XP085276381, Retrieved from the Internet DOI: 10.1016/j.pharmthera.2017.06.006
MCGETTIGAN, P.HENRY, D: "Use of Non-Steroidal Anti-Inflammatory Drugs That Elevate Cardiovascular Risk: An Examination of Sales and Essential Medicines Lists in Low-, Middle-, and High-Income Countries", PLOS MED, vol. 10, no. 2, 2013, pages eiooi388, Retrieved from the Internet
BECKER, J. C.DOMSCHKE, W.POHLE, T.: "Current Approaches to Prevent NSAID-Induced Gastropathy--COX Selectivity and Beyond", BR. J. CLIN. PHARMACOL, vol. 58, no. 6, 2004, pages 587 - 600, XP002349932, Retrieved from the Internet DOI: 10.1111/j.1365-2125.2004.02198.x
SZCZEKLIK, A: "The Cyclooxygenase Theory of Aspirin-Induced Asthma", EUR. RESPIR. J., vol. 3, no. 5, 1990, pages 588 - 593
JUNG, O.JANSEN, F.MIETH, A.BARBOSA-SICARD, E.PLIQUETT, R. U.BABELOVA, A.MORISSEAU, C.HWANG, S. H.TSAI, C.HAMMOCK, B. D. ET AL.: "Inhibition of the Soluble Epoxide Hydrolase Promotes Albuminuria in Mice with Progressive Renal Disease", PLOS ONE, vol. 5, no. 8, 2010, pages e11979, Retrieved from the Internet
SALA, A.PROSCHAK, ESTEINHILBER, D.ROVATI, G. E: "Two-Pronged Approach to Anti-Inflammatory Therapy through the Modulation of the Arachidonic Acid Cascade", BIOCHEM. PHARMACOL, vol. 158, 2018, pages 161 - 173, Retrieved from the Internet
LIU, J.-Y.YANG, J.INCEOGLU, B.QIU, H.ULU, A.HWANG, S.-H.CHIAMVIMONVAT, N.HAMMOCK, B. D: "Inhibition of Soluble Epoxide Hydrolase Enhances the Anti-Inflammatory Effects of Aspirin and -Lipoxygenase Activation Protein Inhibitor in a Murine Model", BIOCHEM. PHARMACOL, vol. 79, no. 6, 2010, pages 880 - 887, XP026856946, Retrieved from the Internet
GARSCHA, U.ROMP, E.PACE, S.ROSSI, A.TEMML, V.SCHUSTER, D.KONIG, S.GERSTMEIER, J.LIENING, S.WERNER, M. ET AL.: "Pharmacological Profile and Efficiency in Vivo of Diflapolin, the First Dual Inhibitor of -Lipoxygenase-Activating Protein and Soluble Epoxide Hydrolase", SCI. REP., vol. 7, no. 1, 2017, pages 9398, XP055725888, Retrieved from the Internet DOI: 10.1038/s41598-017-09795-w
MEIRER, K.GLATZEL, D.KRETSCHMER, S.WITTMANN, S. K.HARTMANN, M.BLOCHER, R.ANGIONI, C.GEISSLINGER, G.STEINHILBER, D.HOFMANN, B. ET A: "Design, Synthesis and Cellular Characterization of a Dual Inhibitor of -Lipoxygenase and Soluble Epoxide Hydrolase", MOL. BASEL SWITZ, vol. 22, no. 1, 2016, Retrieved from the Internet
HIESINGER, K.SCHOTT, A.KRAMER, J. S.BLOCHER, R.WITT, F.WITTMANN, S. K.STEINHILBER, D.POGORYELOV, D.GERSTMEIER, J.WERZ, O. ET AL.: "Design of Dual Inhibitors of Soluble Epoxide Hydrolase and LTA Hydrolase", ACS MED. CHEM. LETT., 2019, Retrieved from the Internet
HWANG, S. H.WAGNER, K. M.MORISSEAU, C.LIU, J.-Y.DONG, H.WECKSLER, A. T.HAMMOCK, B. D.: "Synthesis and Structure-Activity Relationship Studies of Urea-Containing Pyrazoles as Dual Inhibitors of Cyclooxygenase-2 and Soluble Epoxide Hydrolase", J. MED. CHEM., vol. 54, no. 8, 2011, pages 3037 - 3050, XP055181803, Retrieved from the Internet DOI: 10.1021/jm2001376
ZHANG, G.PANIGRAHY, D.HWANG, S. H.YANG, J.MAHAKIAN, L. MWETTERSTEN, H. I.LIU, J.-Y.WANG, Y.INGHAM, E. STAM, S. ET AL.: "Dual Inhibition of Cyclooxygenase-2 and Soluble Epoxide Hydrolase Synergistically Suppresses Primary Tumor Growth and Metastasis", PROC. NATL. ACAD. SCI. U. S. A., vol. 111, no. 30, 2014, pages 11127 - 11132, XP055578595, Retrieved from the Internet DOI: 10.1073/pnas.1410432111
GARTUNG, A.YANG, J.SUKHATME, V. P.BIELENBERG, D. R.FERNANDES, D.CHANG, J.SCHMIDT, B. A.HWANG, S. H.ZURAKOWSKI, D.HUANG, S. ET AL.: "Suppression of Chemotherapy-Induced Cytokine/Lipid Mediator Surge and Ovarian Cancer by a Dual COX- SEH Inhibitor", PROC. NATL. ACAD. SCI. U. S. A., vol. 116, no. 5, 2019, pages 1698 - 1703, Retrieved from the Internet
ZHANG, C.-Y.DUAN, J.-X.YANG, H.-H.SUN, C.-C.ZHONG, W.-J.TAO, J.-H.GUAN, X.-X.JIANG, H.-L.HAMMOCK, B. D.HWANG, S. H. ET AL.: "COX- SEH Dual Inhibitor PTUPB Alleviates Bleomycin-Induced Pulmonary Fibrosis in Mice via Inhibiting Senescence", FEBSJ, 2019, Retrieved from the Internet
DILEEPAN, M.RASTLE-SIMPSON, S.GREENBERG, Y.WIJESINGHE, D. S.KUMAR, N. G.YANG, J.HWANG, S. H.HAMMOCK, B. D.SRIRAMARAO, P.RAO, S. P: "Effect Of Dual SEH/COX- Inhibition on Allergen-Induced Airway Inflammation", FRONT. PHARMACOL, vol. 10, 2019, pages 1118, Retrieved from the Internet
PODOLIN, P. L.BOLOGNESE, B. J.FOLEY, J. F.LONG, E.PECK, B.UMBRECHT, S.ZHANG, X.ZHU, P.SCHWARTZ, B.XIE, W. ET AL.: "In Vitro and in Vivo Characterization of a Novel Soluble Epoxide Hydrolase Inhibitor", PROSTAGLANDINS OTHER LIPID MEDIAT, vol. 104-105, 2013, pages 25 - 31, Retrieved from the Internet
KLAPARS, A.BUCHWALD, S. L.: "Copper-Catalyzed Halogen Exchange in Aryl Halides: An Aromatic Finkelstein Reaction", J. AM. CHEM. SOC., vol. 124, no. 50, 2002, pages 14844 - 14845, XP002337383, Retrieved from the Internet DOI: 10.1021/ja028865v
ZHI, W.LI, J.ZOU, D.WU, Y.WU, Y.: "Palladium-Catalyzed Diastereoselective Synthesis of β,β-Diarylpropionic Acid Derivatives and Its Application to the Total Synthesis of (R)-Tolterodine and the Enantiomer of a Key Intermediate for MK-8718", TETRAHEDRON LETT., vol. 59, no. 6, 2018, pages 537 - 540, XP085337013, Retrieved from the Internet DOI: 10.1016/j.tetlet.2017.12.082
WOLF, N. M.MORISSEAU, C.JONES, P. D.HOCK, B.HAMMOCK, B. D.: "Development of a High-Throughput Screen for Soluble Epoxide Hydrolase Inhibition", ANAL. BIOCHEM., vol. 3, no. 1, 2006, pages 71 - 80, XP024942103, Retrieved from the Internet DOI: 10.1016/j.ab.2006.04.045
KRETSCHMER, S. B. M.WOLTERSDORF, S.VOGT, D.LILLICH, F. F.RIIHL, M.KARAS, M.MAUCHER, I. V.ROOS, J.HAFNER, A.-K.KAISER, A. ET AL.: "Characterization of the Molecular Mechanism of -Lipoxygenase Inhibition by -Aminothiazoles", BIOCHEM. PHARMACOL, vol. 123, 2017, pages 52 - 62, XP029847648, Retrieved from the Internet DOI: 10.1016/j.bcp.2016.09.021
ELDRUP, A. B.SOLEYMANZADEH, F.TAYLOR, S. J.MUEGGE, I.FARROW, N. A.JOSEPH, D.; MCKELLOP, K.MAN, C. C.KUKULKA, A.DE LOMBAERT, S: "Structure-Based Optimization of Arylamides as Inhibitors of Soluble Epoxide Hydrolase", J. MED. CHEM., vol. 52, no. 19, 2009, pages 5880 - 5895, XP055437663, Retrieved from the Internet DOI: 10.1021/jm9005302
GILBERT, N. C.BARTLETT, S. G.WAIGHT, M. T.NEAU, D. B.BOEGLIN, W. E.BRASH, A. R.NEWCOMER, M. E: "The Structure of Human -Lipoxygenase", SCIENCE, vol. 331, no. 6014, 2011, pages 217 - 219, Retrieved from the Internet
GILBERT, N. C.RUI, Z.NEAU, D. B.WAIGHT, M. T.BARTLETT, S. G.BOEGLIN, W. E.BRASH, A. R.NEWCOMER, M. E: "Conversion of Human -Lipoxygenase to a -Lipoxygenase by a Point Mutation to Mimic Phosphorylation at Serine-66", FASEB J. OFF. PUBL. FED. AM. SOC. EXP. BIOL., vol. 26, no. 8, 2012, pages 3222 - 3229, Retrieved from the Internet
HAFNER, A.-K.CERNESCU, M.HOFMANN, B.ERMISCH, M.HORNIG, M.METZNER, J.SCHNEIDER, G.BRUTSCHY, B.STEINHILBER, D.: "Dimerization of Human -Lipoxygenase", BIOL. CHEM., vol. 392, no. 12, 2011, pages 1097 - 1111, Retrieved from the Internet
STEWART, A. O.BROOKS, D. W.: "N,O-Bis(Phenoxycarbonyl)Hydroxylamine: A New Reagent for the Direct Synthesis of Substituted N-Hydroxyureas", J. ORG. CHEM., vol. 57, no. 18, 1992, pages 5020 - 5023, XP055003256, Retrieved from the Internet DOI: 10.1021/jo00044a046
HAHN, S.ACHENBACH, J.BUSCATO, E.KLINGLER, F.-M.SCHROEDER, M.MEIRER, K.HIEKE, M.HEERING, J.BARBOSA-SICARD, E.LOEHR, F. ET AL.: "Complementary Screening Techniques Yielded Fragments That Inhibit the Phosphatase Activity of Soluble Epoxide Hydrolase", CHEMMEDCHEM, vol. 6, no. 12, 2011, pages 2146 - 2149, Retrieved from the Internet
LUKIN, A.KRAMER, J.HARTMANN, M.WEIZEL, L.HERNANDEZ-OLMOS, V.FALAHATI, K.BURGHARDT, I.KALINCHENKOVA, N.BAGNYUKOVA, D.ZHURILO, N. ET: "Discovery of Polar Spirocyclic Orally Bioavailable Urea Inhibitors of Soluble Epoxide Hydrolase", BIOORGANIC CHEM., vol. 80, 2018, pages 655 - 667, XP085445941, Retrieved from the Internet DOI: 10.1016/j.bioorg.2018.07.014
BRUNGS, M.RADMARK, O.SAMUELSSON, B.STEINHILBER, D.: "Sequential Induction of 5-Lipoxygenase Gene Expression and Activity in Mono Mac 6 Cells by Transforming Growth Factor Beta and 1,25-Dihydroxyvitamin D3.", PROC. NATL. ACAD. SCI., vol. 92, no. 1, 1995, pages 107 - 111, Retrieved from the Internet
STEINHILBER, D.HERRMANN, T.ROTH, H. J: "Separation of Lipoxins and Leukotrienes from Human Granulocytes by High-Performance Liquid Chromatography with a Radial-Pak Cartridge after Extraction with an Octadecyl Reversed-Phase Column", J. CHROMATOGR. B. BIOMED. SCI. APP., vol. 493, 1989, pages 361 - 366, Retrieved from the Internet
WERZ, O.BURKERT, E.SAMUELSSON, B.RADMARK, O.STEINHILBER, D: "Activation of -Lipoxygenase by Cell Stress Is Calcium Independent in Human Polymorphonuclear Leukocytes", BLOOD, vol. 99, no. 3, 2002, pages 1044 - 1052, Retrieved from the Internet
XING, L.MCDONALD, J. J.KOLODZIEJ, S. A.KURUMBAIL, R. G.WILLIAMS, J. M.WARREN, C. J.O'NEAL, J. M.SKEPNER, J. E.ROBERDS, S. L: "Discovery of Potent Inhibitors of Soluble Epoxide Hydrolase by Combinatorial Library Design and Structure-Based Virtual Screening", J. MED. CHEM., vol. 54, no. 5, 2011, pages 1211 - 1222, Retrieved from the Internet
HIESINGER, K.KRAMER, J. S.ACHENBACH, J.MOSER, D.WEBER, J.WITTMANN, S. K.MORISSEAU, C.ANGIONI, C.GEISSLINGER, G.KAHNT, A. S. ET AL.: "Computer-Aided Selective Optimization of Side Activities of Talinolol", ACS MED. CHEM. LETT., vol. 10, no. 6, 2019, pages 899 - 903, Retrieved from the Internet
ADAMS, P. D.AFONINE, P. V.BUNKOCZI, G.CHEN, V. B.DAVIS, I. W.ECHOLS, N.HEADD, J. J.HUNG, L.-W.KAPRAL, G. J.GROSSE-KUNSTLEVE, R. W.: "PHENIX: A Comprehensive Python-Based System for Macromolecular Structure Solution", ACTA CRYSTALLOGR. D BIOL. CRYSTALLOGR, vol. 66, no. 2, 2010, pages 213 - 221
OSTER, L.TAPANI, S.XUE, Y.KACK, H: "Successful Generation of Structural Information for Fragment-Based Drug Discovery", DRUG DISCOV. TODAY, vol. 20, no. 9, 2015, pages 1104 - 1111, Retrieved from the Internet
EMSLEY, P.COWTAN, K.: "Coot: Model-Building Tools for Molecular Graphics", ACTA CRYSTALLOGR. D BIOL. CRYSTALLOGR, vol. 60, no. 12, 2004, pages 2126 - 2132
MORIARTY, N. W.GROSSE-KUNSTLEVE, R. W.ADAMS, P. D.: "lectronic Ligand Builder and Optimization Workbench (ELBOW): A Tool for Ligand Coordinate and Restraint Generation", ACTA CRYSTALLOGR. D BIOL. CRYSTALLOGR., vol. 65, no. 10, 2009, pages 1074 - 1080
SCHIERLE, S.FLAUAUS, C.HEITEL, P.WILLEMS, S.SCHMIDT, J.KAISER, A.WEIZEL, L.GOEBEL, T.KAHNT, A. S.GEISSLINGER, G. ET AL.: "Boosting Anti-Inflammatory Potency of Zafirlukast by Designed Polypharmacology", J. MED. CHEM., vol. 61, no. 13, 2018, pages 5758 - 5764, Retrieved from the Internet
BLOCHER, R.LAMERS, C.WITTMANN, S. K.MERK, D.HARTMANN, M.WEIZEL, L.DIEHL, O.BRUGGERHOFF, A.BOB, M.KAISER, A. ET AL.: "N-Benzylbenzamides: A Novel Merged Scaffold for Orally Available Dual Soluble Epoxide Hydrolase/Peroxisome Proliferator-Activated Receptor γ Modulators", J. MED. CHEM., vol. 59, no. 1, 2016, pages 61 - 81, Retrieved from the Internet
Attorney, Agent or Firm:
KUTTENKEULER, David et al. (DE)
Download PDF:
Claims:
CLAIMS

A compound having the formula I: wherein:

R is H or a C to C 0, straight or branched, alkyl or alkenyl,

A is an aryl or heteroaryl, preferably is substituted or unsubstituted phenyl, tolyl, pyridyl, furanyl, thiophenyl, thiazolyl, pyrimidinyl, n is an integer from l to io,

Y is C or N,

R2 is selected from H, substituted or unsubstituted alkyl, phenyl, substituted phenyl, pyridyl, furanyl, thiophenyl, thiazolyl, pyrimidinyl, and preferably is substituted phenyl.

R3 and R4 are independently selected from H, F, Cl, Br, I, CF3, OMe, OCF3, CHF2, Me, S02Me, SMe, S02NH2, NHS02Me, ethyl; and solvates, salts, stereoisomers, complexes, polymorphs, crystalline forms, racemic mixtures, diastereomers, enantiomers, tautomers, isotopically labelled forms, prodrugs, and combinations thereof.

2. The compound of claim 1, which has not more than one stereogenic centers.

3- The compound of claim 1 or 2, wherein the compound has an activity as dual inhibitor of soluble epoxide hydrolase (sEH) and 5-lipoxygenase (5-LOX), 4- The compound of any one of claims 1 to 3, wherein the compound has an activity as a selective 5-LOX inhibitor, and has no inhibitory effect on 12- and/ or 15-LOX.

5. The compound of any one of claims 1 to 4, which has an activity of inhibiting sEH with an IC5o of less than about 0.05 mM, preferably less than about 0.01 pM, more preferably of less than about 0.005 pM; and(/ or) which has an activity of inhibiting 5-LOX with an

IC5o of less than about 1 pM, more preferably of less than about 0.5 pM, more preferably of less than about 0.3 pM.

6. The compound of any one of claims l to 5, wherein the compound has any one of the following structures:

7. The compound of any one of claims 1 to 6, for use in the treatment of a disease.

8. The compound for use of claim 7, wherein the disease is a disorder associated with an activity of 5-LOX and/ or sEH, and/ or with a metabolism or metabolites of arachidonic acid (AA). 9. The compound for use of claim 8, wherein the disease is a metabolic disorder or an inflammatory disorder, such as acute or chronic inflammation, rheumatoid arthritis, a cardiovascular disease, inflammatory bowel disease, or sepsis.

10. The compound for use of any one of claims 7 to 9, wherein the use in medicine comprises the administration of a therapeutically effective amount of the compound, or of solvates, salts, stereoisomers, complexes, polymorphs, crystalline forms, racemic mixtures, diastereomers, enantiomers, tautomers, isotopically labelled forms, prodrugs, and combinations thereof, to a subject in need of the treatment.

11. A method for producing the compound of any one of claims 1 to 6.

12. A method of inhibiting the enzymatic function of a sEH protein, the method comprising the steps of contacting the sEH protein with the compound recited in any one of claims 1 to 6.

A method of inhibiting the enzymatic function of a 5-LOX protein, the method comprising the steps of contacting the 5-LOX protein with the compound recited in any one of claims 1 to 6.

14. The method according to claim 12 or 13, wherein the enzymatic function of sEH is the catalytic metabolization of epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acids.

15. The method according to any one of claims 12 to 14, wherein the sEH protein is contacted with the compound within a cell.

Description:
DUAL INHIBITORS OF SOLUBLE EPOXIDE HYDROLASE AND 5-LIPOXYGENASE

FIELD OF THE INVENTION

[1] The invention pertains to a novel structure that provides an activity as a dual inhibitor of the enzymes soluble epoxide hydrolase (sEH) and 5-lipoxygenase (5-LOX). The invention pertains to multiple derivatives of the new class of dual inhibitors, their application in medicine, pharmaceutical compositions comprising them as well as to methods for synthesizing the new compounds.

DESCRIPTION

[2] Inflammation is a complex physiological process which is activated by the immune system upon a harmful stimulus. 1 Amongst others, inflammation is mediated by different metabolites of the arachidonic acid (AA). AA is metabolized by a cascade of biochemical reactions, which are subdivided into three major pathways, namely the 5-lipoxygenase (5-LOX), the cyclooxygenase (COX) and cytochrome P450 (CYP450) pathway. Lipids generated by the 5- LOX branch are leukotrienes (LTs) and lipoxins (LXs). The LTs are pro-inflammatory and chemotactic mediators, hence, the inhibition of 5-LOX is an established strategy to counteract asthma, 2 and is under intensive investigation for a diverse inflammatory diseases. 3 The prostanoids produced by the COX pathway are divided into prostaglandins (PGs), and thromboxane (TX), while the metabolites of the CYP450 branch are called epoxyeicosatrienoic acids (EETs). The soluble epoxide hydrolase (sEH), located in the CYP450 branch, converts the anti-inflammatory EETs to the less biological active dihydroxyeicosatrienoic acids (DHETs). 4 Hence, inhibition of sEH leads to accumulation of EETs and pronounced anti-inflammatory effects in the settings of acute and chronic inflammation, such as rheumatoid arthritis, 5 cardiovascular disease, inflammatory bowel disease, and sepsis. 6

[3] Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most popular anti inflammatory drugs on the market. NSAIDs effectively target the COX pathway, however they are associated with a broad range of side effects. 7 Some of these side effects are caused by shunting of AA metabolites within the AA cascade. 8 One prominent example is aspirin induced asthma, caused by accumulation of unmetabolized AA which shunts to the LOX pathway resulting in increased leukotriene E4 (LTE 4 ) levels. 9 Another shunting effect was reported by Jung et al. 10 showing that treatment of mice with a sEH inhibitor promoted proteinuria possibly due to the shift to higher LT levels. Shunting effects might be prevented by inhibition of more than one enzyme in the AA cascade. 11 Especially the combination of sEH inhibition with one of the remaining branches of the AA cascade seems to be a valuable strategy to design efficient and safe anti-inflammatory compounds. 4 First evidence on these findings was reported by Liu et al, who demonstrated synergistic effects of a sEH inhibitor with either 5-lipoxygenase activating protein (FLAP) inhibitor or COX inhibitor aspirin in a lipopolysaccharide (LPS) induced inflammation in mice. 12 Encouraged by these findings, several successful attempts to design multitarget ligands affecting sEH and an additional enzyme or receptor of the AA cascade were made. 4

[4] Garsha et al. developed diflapolin 1, a dual sEH and FLAP inhibitor, which decreased leukotriene formation in vivo in a zymosan-induced mouse peritonitis model. 13 A dual sEH/5- LOX inhibitor 2 (KM55) blocked the LPS induced adhesion of leukocytes to endothelial cells by impairing leukocyte function. 14 The dual SEH/LTA4H inhibitor 3 reduced the leukotriene B 4 (LTB 4 ) and prostaglandin levels in bacteria-activated Mi and M2 macrophages. 15 A dual inhibitor of sEH and COX-24 (PTUBP) 16 exhibits excellent in vivo efficacy in murine models of cancer, 1748 pulmonary fibrosis, 19 and allergen-induced airway inflammation 20 (Scheme 1).

[5] The soluble epoxide hydrolase (sEH) is a downstream enzyme of the CYP pathway of arachidonic acid metabolism and also holds promise in the treatment of NAFLD/NASH and other metabolic diseases such as type 2 diabetes mellitus (Shen, H. C.; Hammock, B. D. Discovery of Inhibitors of Soluble Epoxide Hydrolase: A Target with Multiple Potential Therapeutic Indications. J. Med. Chem. 2012, 55 (5), 1789-1808; Newman, J. W. et al; Epoxide Hydrolases: Their Roles and Interactions with Lipid Metabolism. Prog. Lipid Res. 2005, 44 (1), 1-51; Imig, J. D. Epoxides and Soluble Epoxide Hydrolase in Cardiovascular Physiology. Physiol. Rev. 2012, 92 (1), 101-130; Huang, H.; Weng, J.; Wang, M.-H. EETs/sEH in Diabetes and Obesity-Induced Cardiovascular Diseases. Prostaglandins Other Lipid Mediat. 2016, 125, 80-89.). It converts epoxyeicosatrienoic acids (EETs) formed by CYP enzymes from arachidonic acid to the respective dihydroxyeicosatrienoic acids (DHETs). Since EETs exhibit robust anti inflammatory activities, sEH inhibition constitutes an anti-inflammatory strategy. sEH is expressed throughout the body with especially high levels in heart, liver and kidney.

[6] The 5-lipoxygenase (5-LO or 5-LOX) pathway is the major source of proinflammatory leukotrienes (LTs) produced from the metabolism of arachidonic acid (AA). Cytosolic phospholipase A 2 (cPLA 2 ) liberates arachidonic acid from membrane phospholipids. The arachidonic acid is presented by FLAP to 5-LO. The 5-LO enzyme converts arachidonic acid to an unstable intermediate called 5-hydroperoxyeicosatetraenoic acid (5-HPETE), which is then dehydrated by 5-LO to produce LLA,, a pivotal intermediate in the biosynthesis of inflammatory and anaphylactic mediator. Depending upon cell-type, ULA, is then converted into the chemoattractant LTB 4 (in neutrophils and monocytes), or LTC, (in human eosinophils, mast cells and basophils), which becomes sequentially cleaved to form the extracellular metabolites LTD 4 and LTE 4 . Further, omega-oxidation and subsequent b-oxidation from the methyl terminus of the LTE 4 is a major metabolic route for sulfidopeptide leukotrienes, which are known as cysteinyl leukotrienes (cysLTs) in humans.

[7] Abnormal production of LTs contributes to a variety of diseases and disorders because LTs are very potent molecules that act through receptors at subnanomolar concentrations. LTC 4 and LTD 4 cause hypotension in humans by causing a significant reduction in coronary blood flow. LTC 4 and LTD 4 constrict coronary arteries and distal segments of the pulmonary artery. LTC 4 and LTD 4 can cause plasma exudation and are more than 1000-times more potent than histamine in this respect. In addition to their effects on coronary blood flow, LTC 4 and LTD 4 are potent constrictors of bronchial smooth muscles. Leukotrienes also stimulate bronchial mucus secretion and cause mucosal edema. The sulfidopeptide leukotrienes have potent effects on micro vasculature. Studies of mucosal biopsies from the bronchi of aspirin-intolerant asthmatics demonstrate that LTC4S is amplified, which correlates with an overproduction of cysLTs and bronchial hyperreactivity.

[8] Scheme 1: Dual inhibitors of the AA cascade.

[9] In view of the foregoing it was an object of the present invention to develop a novel class of dual inhibitors of sEH and 5-LOX.

BRIEF DESCRIPTION OF THE INVENTION

[10] Generally, and by way of brief description, the main aspects of the present invention can be described as follows:

[11] In a first aspect, the invention pertains to a compound having the formula I: wherein: R is H or a C to C 0 , straight or branched, alkyl or alkenyl,

A is an aryl or heteroaryl, preferably is substituted or unsubstituted phenyl, tolyl, pyridyl, furanyl, thiophenyl, thiazolyl, pyrimidinyl, n is an integer from l to io,

Y is C or N,

R 2 is selected from H, substituted or unsubstituted alkyl, phenyl, substituted phenyl, pyridyl, furanyl, thiophenyl, thiazolyl, pyrimidinyl, and preferably is substituted phenyl.

R 3 and R 4 are independently selected from H, F, Cl, Br, I, CF 3 , OMe, OCF 3 , CHF 2 , Me, S0 2 Me, SMe, S0 2 NH 2 , NHS0 2 Me, ethyl; and solvates, salts, stereoisomers, complexes, polymorphs, crystalline forms, racemic mixtures, diastereomers, enantiomers, tautomers, isotopically labelled forms, prodrugs, and combinations thereof.

[12] In a second aspect, the invention pertains to a method of producing/ synthesizing the compounds of the invention.

[13] In a third aspect, the invention pertains to a method of inhibiting the enzymatic function of a sEH protein, the method comprising the steps of contacting the sEH protein with the compound of the invention.

[14] In a fourth aspect, the invention pertains to a method of inhibiting the enzymatic function of a 5-LOX protein, the method comprising the steps of contacting the 5-LOX protein with the compound of the invention.

[15] In a fifth aspect, the invention pertains to a method for moderating an inflammatory response in a mammalian subject, the method comprising the administration of a compound of the invention to the mammalian subject.

[16] In a sixth aspect, the invention pertains to a method of treating an inflammatory disorder in a subject in need of the treatment, the method comprising the administration of a compound of the invention to the mammalian subject

DETAILED DESCRIPTION OF THE INVENTION

[17] In the following, the elements of the invention will be described. These elements are listed with specific embodiments, however, it should be understood that they may be combined in any manner and in any number to create additional embodiments. The variously described examples and preferred embodiments should not be construed to limit the present invention to only the explicitly described embodiments. This description should be understood to support and encompass embodiments which combine two or more of the explicitly described embodiments or which combine the one or more of the explicitly described embodiments with any number of the disclosed and/or preferred elements. Furthermore, any permutations and combinations of all described elements in this application should be considered disclosed by the description of the present application unless the context indicates otherwise.

[18] In a first aspect, the invention pertains to a compound having the formula I: wherein:

R , R 2 , R 3 and R 4 are independently selected from H, an unsubstituted, monosubstituted, or polysubstituted C -C 0 alkyl or heteroalkyl, wherein said alkyl is straight, branched or cyclic, a unsubstituted, monosubstituted or polysubstituted C -C 0 alkenyl or heteroalkenyl, wherein said alkenyl is straight, branched or cyclic, an unsubstituted, monosubstituted, or polysubstituted aryl or heteroaryl, an unsubstituted, monosubstituted, or polysubstituted benzyl group, an acyl group, such as formyl, acetyl, trichloroacetyl, fumaryl, maleyl, succinyl, benzoyl, or acyl groups being branched, heteroatom-substituted or aryl-substituted, a sugar or another acetal, halogen, such as F, Cl, Br, or I, or halogen substituted alkyl or alkoxy such CF 3, OCF 3 , or CHF 2 , is a sulfonyl group, or is S0 2 Me, SMe, S0 2 NH 2 , NHS0 2 Me; A is an aryl or heteroaryl, preferably is substituted or unsubstituted phenyl, tolyl, pyridyl, furanyl, thiophenyl, thiazolyl, pyrimidinyl; n is an integer from 1 to 10,

Y is C or a hetero atom, preferably wherein Y is C or N; and solvates, salts, stereoisomers, complexes, polymorphs, crystalline forms, racemic mixtures, diastereomers, enantiomers, tautomers, isotopically labelled forms, prodrugs, and combinations thereof.

[19] The soluble epoxide hydrolase (sEH) shall refer to a protein such as the soluble version of human soluble epoxide hydrolase (sEH). The protein is shown in the UnitProt database under accession number P34913 (www.unitprot.org). The amino acid sequence is shown in SEQ ID NO: 1. The protein has the following enzymatic activity: an epoxide + H2O = an ethanediol.

[20] The protein 5- lipoxygenase, or arachidonate 5-lipoxygenase, shall refer to a protein having similarity to human 5- lipoxygenase (5-LOX). The protein is shown in the UnitProt database under accession number P09917. The amino acid sequence is shown in SEQ ID NO: 2. The protein has the following enzymatic activity: (5Z,8Z,nZ,i4Z)-eicosa -5,8,11,14-tetraenoate + 0 2 = H 2 O + leukotriene

[21] As used herein, the terms “compound of the invention” or “compounds of the invention” refers to any compound disclosed in the present application or a pharmaceutically acceptable salt thereof, such as a compound of formula (I), or any of the compounds depicted herein elsewhere, or a pharmaceutically acceptable salt thereof.

[22] As used herein, the terms “halo” and “halogen” refer to fluoro, chloro, bromo or iodo.

[23] “Aliphatic” refers to straight-chain, branched or cyclic C 1 -C 10 hydrocarbons which are completely saturated or which contains one or more units of unsaturation but which are not aromatic. Examples of aliphatic groups include linear, branched or cyclic alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl, etc. An aliphatic group may be optionally substituted by 1-6 substituents. Suitable substituents on an aliphatic group include: 3-12 member heterocyclyl, C 5 -C 0 aryl, 5-12 member heteroaryl, halide, -N0 2 , NH 2 , NR 2 , -CN, -COR, -COOR, -CONR 2 , -OH, -OR, -OCOR, -SR, -SOR, - S0 2 R, -S0NR 2 , -S0 2 NR 2 , wherein R is H, C 1 -C 10 alkyl, 3-10 member heterocyclyl.

[24] "C 1 -C 10 alkyl" refers to a straight chain or branched saturated hydrocarbon radical having from 1 to 10 carbon atoms. A C 1 -C 10 alkyl group may be optionally substituted by at least one substituent. Suitable substituents on a C 1 -C 10 alkyl group include, but are not limited to, 3-10 member heterocyclyl, C 5 -C 2 aryl, 5-12 member heteroaryl, halide, -N0 2 , -NR 2 , -CN, -COR, - COOR, -CONR 2 , -OH, -OR, -OCOR, -SR, -SOR, -S0 2 R, -S0NR 2 , -S0 2 NR 2 , wherein each R is independently selected from the group consisting of -H, C 1 -C 10 alkyl, 3-12 member heterocyclyl, C 5 -C 10 aryl, and 5-10 member heteroaryl. Examples of C 1 -C 10 alkyl groups include, but are not limited to methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, iso butyl, tert-butyl, pentyl, neo pentyl, sec -pentyl, hexyl, heptyl, octyl, and the like, including substituted forms thereof. Further, the term “alkyl” refers to a straight chain or branched saturated hydrocarbon radical of 1 to 20 carbon atoms (“C -C 20 alkyl”), or 1 to 10 carbon atoms ("C 1 -C 10 alkyl”), or 1 to 8 carbon atoms (“C -Ce alkyl”), or l to 5 carbon atoms (“C 1 -C 5 alkyl"), or 1 to 4 carbon atoms (“C 1 -C 4 alkyl”), or 1 to 3 carbon atoms (“C 1 -C 3 alkyl”).

[25] "Cycloalkyl" refers to a cyclic saturated hydrocarbon radical having from 3 to 20 carbon atoms ("C 3 -C 20 cycloalkyl"), including 3 to 12 carbon atoms ("C 3 -C 12 cycloalkyl"). A cycloalkyl group may be monocyclic and where permissible may be bicyclic or polycyclic. A cycloalkyl group may be optionally substituted by at least one substituent. Suitable substituents on a cycloalkyl group are the same as those described for an alkyl group. Examples of cycloalkyl groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, nobomyl, adamantyl, and the like, including substituted forms thereof.

[26] “C 2 -C 10 alkenyl” refers to a straight chain or branched unsaturated hydrocarbon radical having from 2 to 10 carbon atoms. A C 2 -C 0 alkenyl group may have one or more points of unsaturation (i.e., one or more carbon-carbon double bonds). In the case where C 2 -C 0 alkenyl has more than one carbon-carbon double bond, the carbon-carbon double bonds can be conjugated or unconjugated. A C 2 -C 0 alkenyl group may be optionally substituted by at least one substituent. Suitable substituents on a C 2 -C 0 alkenyl group are the same as those described for a C 2 -C 10 alkyl group. Examples of C 2 -C 0 alkenyl include, but are not limited to, ethenyl, 1- propenyl, 2-propenyl, 1-butenyl, 2-butenyl, iso-butenyl, and the like, including substituted forms thereof. An alkenyl group may have one or more points of unsaturation (i.e., one or more carbon-carbon double bonds). In the case where an alkenyl group has more than one carbon- carbon double bond, the carbon-carbon double bonds can be conjugated or unconjugated. An alkenyl group may be substituted or unsubstituted. Suitable substituents on an alkenyl group are the same as those described for a C 1 -C 10 alkyl group.

[27] As used herein, the term“C 2 -C 0 alkynyl” as used by itself or as part of another group refers to straight chain and branched non-cyclic hydrocarbons having from 2 to 10 carbon atoms and including at least one carbon-carbon triple bond. Straight chain and branched C 2 -C 0 alkynyl groups can be acetylenyl, propynyl, butyn-l-yl, butyn-2- yl, pentyn-l-yl, pentyn-2-yl, 3- methylbutyn-l-yl, pentyn-4-yl, hexyn-l-yl, hexyn- 2-yl, hexyn-5-yl, and the like. C2-6 alkynyl groups include acetylenyl (i.e., ethynyl), propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3 -methyl- 1- butynyl, 4-pentynyl, and the like. In certain embodiments, the C2-6 alkynyl group can be a C 2- alkynyl group. C 2- alkynyl groups can be ethynyl, propynyl, butynyl, and 2-butynyl groups.

[28] “Alkoxy" refers to -OR, wherein R is C 1 -C 10 alkyl, C 2 -C 0 alkenyl, C 2 -C 0 alkynyl, C 3 -C 2 cycloalkyl or (C 1 -C 5 alkylene)-(C 3 -C 2 cycloalkyl). A “C 1 -C 12 alkoxy" refers to an alkoxy group, as defined herein, wherein R has 1 to 12 total carbon atoms.

[29] The term "aryl" refers to an all-carbon monocyclic ring or polycyclic ring of 6 to 20 carbon atoms having a completely conjugated pi-electron system. Examples of aryl include but are not limited to phenyl, naphthyl, and anthracenyl. C 6 -C 0 aryl refers to aryl with 6-10 carbon atoms in the cyclic structure, including phenyl and naphthyl.

[30] "Heteroaryl" refers to a monocyclic or fused ring group containing one, two, three or four ring heteroatoms selected from N, O, and S, the remaining ring atoms being C, and, in addition, having a completely conjugated pi-electron system. Examples, without limitation, of unsubstituted heteroaryl groups are pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrimidine, quinoline, isoquinoline, purine, tetrazole, triazine, and carbazole. The heteroaryl group may be substituted or unsubstituted. Typical substituents include C1-C10 aliphatic, 3-10 membered heterocyclyl, 6-10 membered aryl, halide, -N0 2 , NH 2 , NR 2 , -CN, -COR, -COOR, -C0NR 2 , -OH, -OR, -OCOR, -SR, -SOR, -S0 2 R, -S0NR 2 , -S0 2 NR 2 , wherein R is a C 3 -C 0 aliphatic, 3-10 membered heterocyclyl, C 5 -C 0 aryl, and 5-10 membered heteroaryl.

[31] Examples of typical monocyclic heteroaryl groups include, but are not limited to: pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, isoxazolyl, oxazolyl, isothiazolyl, thiazolyl, 1,2,3-triazolyl, 1,3,4-triazolyl, l-oxa-2,3,-diazolyl, l-oxa-2,4-dizolyl, l-oxa-2,5- diazolyl, l-oxa-3,4- diazolyl, l-thia-3,4-diazolyl, l-thia-2,3-diazolyl, l-thia-2, 4, -diazolyl, 1- thia-2,5-diazolyl, 1-thia- 3,4-diazolyl, tetrazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, and triazinyl.

[32] Examples of bicyclic heteroaryl groups include, but are not limited to: benzofuranyl, benzothiophenyl, indolyl, benzimidazolyl, indazolyl, benzotriazolyl, pyrrolo[2,3-h]pyridinyl, pyrrolo[2,3-c]pyridinyl, pyrrolo[3,2-c]pyridinyl, py r rol 0 [3, 2-b] pyridinyl, imidazo[4,5- b] pyridinyl, imidazo[4,5-c]pyridinyl, pyrazolo[4,3-d]pyridinyl, pyrazolo[4,3-c]pyridinyl, pyrazolo[3,4-c]pyridinyl, pyrazolo[3,4-h]pyridinyl, isoindolyl, indazolyl, purinyl, indolininyl, imidazo[i,2-a]pyridinyl, imidazo[i,5-a]pyridinyl, pyrazolo[i,5-a]pyridinyl, pyrrolo[i,2- hjpyridazinyl, imidazo[i,2-c]pyrimidinyl, thienopyrimidinyl, quinolinyl, isoquinolinyl, cinnolinyl, azaquinazoline, quinoxalinyl, phthalazinyl, 1,6-naphthyridinyl, 1,7- naphthyridinyl, 1,8-naphthyridinyl, 1,5-naphthyridinyl, 2,6-naphthyridinyl, 2,7- naphthyridinyl, pyrido[3,2- d] pyrimidinyl, pyrido[4,3-d]pyrimidinyl, pyrido[3,4-d]pyrimidinyl, pyrido[2,3-d]pyrimidinyl, pyrido[2,3-h]pyrazinyl, pyrido[3,4-h]pyrazinyl, pyrimido[5,4-d]pyrimidinyl, pyrazino[2,3- h] pyrazinyl, and pyrimido[4,5-d]pyrimidinyl.

[33] As used herein, the term “amino” refers to -NH 2 .

[34] As used herein, the term “hydroxy” refers to -OH.

[35] As used herein, the term “cyano” refers to -CN.

[36] As used herein, the term “nitro” refers to -N0 2 .

[37] As used herein, the term “carboxylic acid” refers to -COOH. [38] Optional substituents on optionally substituted groups, when not otherwise indicated, include one or more groups, for example, 1, 2, or 3 groups, independently selected from the group consisting of halo, halo(Ci-6)alkyl, aryl, heterocycle, cycloalkyl, Ci-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, aryl(Ci-6)alkyl, aryl(C2-6)alkenyl, aryl(C2-6)alkynyl, cycloalkyl(Ci-6)alkyl, heterocyclo(Ci-6)alkyl, hydroxy(Ci-6)alkyl, amino(Ci-6)alkyl, carboxy(Ci-6)alkyl, alkoxy(Ci- 6)alkyl, nitro, amino, ureido, cyano, alkylcarbonylamino, hydroxy, thiol, alkylcarbonyloxy, aryloxy (e.g., phenoxy and benzyloxy), aryl(Ci-6)alkyloxy, carboxamido, sulfonamido, azido, Ci- 6 alkoxy, halo(Ci-6)alkoxy, carboxy, aminocarbonyl and mercapto(Ci-6)alkyl groups mentioned above. Preferred optional substituents include halo, halo(Ci-6)alkyl, hydroxy(Ci-6)alkyl, amino(Ci-6)alkyl, hydroxy, nitro, Ci-6 alkyl, Ci-6 alkoxy, halo(Ci-6)alkoxy and amino.

[39] Compounds of the disclosure encompass all the salts of the disclosed compounds of formula (I). The present disclosure includes all non-toxic pharmaceutically acceptable salts thereof of the disclosed compounds. In certain embodiments, pharmaceutically acceptable addition salts can be inorganic and organic acid addition salts and basic salts. In certain embodiments, pharmaceutically acceptable salts can be metal salts such as sodium salt, potassium salt, caesium salt and the like; alkaline earth metals such as calcium salt, magnesium salt and the like; organic amine salts such as triethylamine salt, pyridine salt, picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, Ay V - d i b e n zy 1 et h y 1 e n e d i a m i n e salt and the like; inorganic acid salts such as hydrochloride, hydrobromide, phosphate, sulphate and the like; organic acid salts such as citrate, lactate, tartrate, maleate, fumarate, mandelate, acetate, dichloroacetate, trifluoroacetate, oxalate, formate and the like; sulfonates such as methanesulfonate, benzenesulfonate, p-toluenesulfonate and the like; and amino acid salts such as arginate, asparginate, glutamate and the like.

[40] In certain embodiments, compounds of the disclosure can contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms, such as epimers. The present disclosure is meant to encompass the uses of all such possible forms, as well as their racemic and resolved forms and mixtures thereof. The individual enantiomers may be separated according to methods known to those of ordinary skill in the art in view of the present disclosure. When the compounds described herein contain double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that they include both E and Z geometric isomers. All tautomers are intended to be encompassed by the present disclosure as well.

[41] As used herein, the term “stereoisomers” is a general term for all isomers of individual molecules that differ only in the orientation of their atoms in space. It includes enantiomers and isomers of compounds with more than one chiral center that are not mirror images of one another (diastereomers). [42] The term “chiral center” refers to a carbon atom to which four different groups are attached.

[43] The term “epimer” refers to diastereomers that have opposite configuration at only one of two or more tetrahedral stereogenic centers present in the respective molecular entities.

[44] The term “stereogenic center” is an atom, bearing groups such that an interchanging of any two groups leads to a stereoisomer.

[45] The terms “enantiomer” and “enantiomeric” refer to a molecule that cannot be superimposed on its mirror image and hence is optically active wherein the enantiomer rotates the plane of polarized light in one direction and its mirror image compound rotates the plane of polarized light in the opposite direction.

[46] The term “racemic” refers to a mixture of equal parts of enantiomers and which mixture is optically inactive.

[47] The compound of the invention in some preferred embodiments may include one or more stereogenic centers. However, even more preferred is a compound which has not more than two, preferably not more than one chiral center and/or stereogenic centers.

[48] The compound of the invention is preferred wherein in formula I R 2 is substituted phenyl, and wherein said substitution is identical to R 3 .

[49] In further preferred embodiments R is methyl.

[50] In further preferred embodiments A is phenyl.

[51] In further preferred embodiments n is 2.

[52] In further preferred embodiments R 3 is H and R 4 is F in para position.

[53] In further preferred embodiments R 2 is:

[54] In further preferred embodiments:

R is methyl,

A is phenyl, n is 2, R 3 is H and ¾ is F in para position, and

R 2 is:

[55] In further preferred embodiments the compound of the invention has any one of the following structures:

[56] Preferably the compound of the invention has an activity as dual inhibitor of soluble epoxide hydrolase (sEH) and 5-lipoxygenase (5-LOX), and preferably, wherein the activity as 5- LOX inhibitor is selective, and has no inhibitory effect on 12- and/or 15-LOX. Hence, it is preferred that the compound of the invention has an activity of inhibiting sEH with an IC 50 of less than about 0.05 mM, preferably less than about 0.01 mM, more preferably of less than about 0.005 pM; and(/or) which has an activity of inhibiting 5-LOX with an IC 50 of less than about 1 pM, more preferably of less than about 0.5 pM, more preferably of less than about 0.3 pM.

[57] In preferred aspects and embodiments the compounds of the invention find application in the treatment of animal or human diseases. Most preferably in a disease associated with the activity of 5-LOX and sEH.

[58] As used herein, the terms “treating,” “treat,” or “treatment” refers to both therapeutic treatment and prophylactic or preventative measures, wherein the object is to prevent or slow down (lessen) an undesired physiological condition, disorder or disease, or to obtain beneficial or desired clinical results. For the purposes of this invention, beneficial or desired clinical results include, but are not limited to, alleviation of symptoms; diminishment of the extent of the condition, disorder or disease; stabilization (i.e., not worsening) of the state of the condition, disorder or disease; delay in onset or slowing of the progression of the condition, disorder or disease; amelioration of the condition, disorder or disease state; and remission (whether partial or total), whether detectable or undetectable, or enhancement or improvement of the condition, disorder or disease. Treatment includes eliciting a clinically significant response with or without excessive levels of side effects. Treatment can include inhibiting a pathological metabolic phenotype or inflammatory response in a subject. In certain embodiments, compounds of the disclosure can be used in combination with at least one other therapeutic agent.

[59] Most preferably the compounds of the invention due to their activity are advantageously useful in the treatment of a metabolic disorder, preferably a metabolic disorder, autoimmune disorder or inflammatory disease.

[60] The term “inflammatory disease” refers to a condition in a subject characterized by inflammation, e.g. chronic inflammation. Illustrative, non-limiting examples of inflammatory disorders include, but are not limited to, rheumatoid arthritis (RA), inflammatory bowel disease (IBD), asthma, encephalitis, chronic obstructive pulmonary disease (COPD), inflammatory osteolysis, allergic disorders, septic shock, pulmonary fibrosis (e g , idiopathic pulmonary fibrosis), inflammatory vacuhtides (e g polyarteritis nodosa, Wegner's granulomatosis, Takayasu’s arteritis, temporal arteritis, and lymphomatoid granulomatosus), post-traumatic vascular angioplasty (e.g. , restenosis after angioplasty), undifferentiated spondyloarthropathy, undifferentiated arthropathy, arthritis, inflammatory osteolysis, chronic hepatitis, and chronic inflammation resulting from chronic viral or bacterial infections.

[61] As used herein, the terms “disorder” and “disease” are used interchangeably to refer to a condition in a subject. In particular, the term “autoimmune disease” is used interchangeably with the term “autoimmune disorder” to refer to a condition in a subject characterized by cellular, tissue and/or organ injury caused by an immunologic reaction of the subject to its own cells, tissues and/or organs. The term “inflammatory disease” is used interchangeably with the term “inflammatory disorder” to refer to a condition in a subject characterized by inflammation, preferably chronic inflammation. Autoimmune disorders may or may not be associated with inflammation. Moreover, inflammation may or may not be caused by an autoimmune disorder. Thus, certain disorders may be characterized as both autoimmune and inflammatory disorders. Preferably the disease to be treated or prevented in accordance with the invention is a disease associated with a pathological immune response, such as an inflammatory disease, such as of acute and chronic inflammation, rheumatoid arthritis cardiovascular disease, inflammatory bowel disease, and sepsis.

[62] The term “metabolic disorder” refers to any disorder that involves an alteration in the normal metabolism of carbohydrates, lipids, proteins, nucleic acids or a combination thereof. A metabolic disorder is associated with either a deficiency or excess in a metabolic pathway resulting in an imbalance in metabolism of nucleic acids, proteins, lipids, and/or carbohydrates. Factors affecting metabolism include, but are not limited to, the endocrine (hormonal) control system (e.g., the insulin pathway, the enteroendocrine hormones including GLP-i, GLP-2, oxyntomodulin, PYY or the like), the neural control system (e.g. GLP-i in the brain) or the like. Preferably the disease treated with the compounds of the invention is a disease associated with the metabolism or metabolites of arachidonic acid (AA) metabolic pathway.

[63] In context of the herein disclosed invention the use in medicine comprises the administration of a therapeutically effective amount of the compound, or of solvates, salts, stereoisomers, complexes, polymorphs, crystalline forms, racemic mixtures, diastereomers, enantiomers, tautomers, isotopically labelled forms, prodrugs, and combinations thereof, to a subject in need of the treatment.

[64] When administered to a subject, a compound of the disclosure can be administered as a component of a composition that comprises a pharmaceutically acceptable carrier or excipient. Such compositions also constitute an aspect of the invention. A compound of the disclosure can be administered by any appropriate route, as determined by the medical practitioner. Methods of administration may include intradermal, intramuscular, intraperitoneal, parenteral, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, buccal, intracerebral, intravaginal, transdermal, transmucosal, rectal, by inhalation, or topical (such as to the ears, nose, eyes, or skin). Delivery can be either local or systemic. In certain embodiments, administration can result in the release of a compound of the disclosure into the bloodstream.

[65] Pharmaceutical compositions of the present disclosure can take the form of solutions, suspensions, emulsions, tablets, pills, pellets, powders, multi particulates, capsules, capsules containing liquids, capsules containing powders, capsules containing multi-particulates, lozenges, sustained-release formulations, suppositories, transdermal patches, transmucosal films, sub-lingual tablets or tabs, aerosols, sprays, or any other form suitable for use. In one embodiment, the composition is in the form of a tablet. In certain embodiments, the composition can be in the form of a capsule (see, e.g. U.S. Patent No. 5,698,155). Other examples of suitable pharmaceutical excipients are described in Remington's Pharmaceutical Sciences 1447-1676 (Alfonso R. Gennaro ed., 19th ed. 1995), incorporated herein by reference.

[66] Pharmaceutical compositions of the present disclosure can comprise a suitable amount of a pharmaceutically acceptable excipient so as to provide the form for proper administration to the subject. In certain embodiments, the pharmaceutical excipient can be a diluent, suspending agent, solubilizer, binder, disintegrant, preservative, coloring agent, lubricant, and the like. The pharmaceutical excipient can be a liquid, such as water or an oil, including those of petroleum, animal, vegetable, or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil, and the like. The pharmaceutical excipient can be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea, and the like. Auxiliary, stabilizing, thickening, lubricating, and coloring agents can be used. In certain embodiments, the pharmaceutically acceptable excipient can be sterile when administered to a subject. Water can be an excipient when a compound of the disclosure is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid excipients, such as for injectable solutions. In certain embodiments, the pharmaceutical excipients can include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol, and the like. In certain embodiments, the compositions can contain minor amounts of wetting or emulsifying agents, or pH buffering agents. Specific examples of pharmaceutically acceptable carriers and excipients that can be used to formulate oral dosage forms are described in the Handbook of Pharmaceutical Excipients, American Pharmaceutical Association (1986).

[67] In certain embodiments, the compounds of the disclosure can be formulated for oral administration. A compound of the disclosure to be orally delivered can be in the form of tablets, capsules, gelcaps, caplets, lozenges, aqueous or oily solutions, suspensions, granules, powders, emulsions, syrups, or elixirs, for example. When a compound of the disclosure is incorporated into oral tablets, such tablets can be compressed, tablet triturates, enteric-coated, sugar-coated, film-coated, multiply compressed or multiply layered.

[68] The daily dose of a compound or composition of the invention administered to a subject is generally in the range from 0.3 mg to too mg (typically from 3 mg to 50 mg) per day per kilogram of body weight, for example 3-10 mg/kg/day. An intravenous dose may be, for example, in the range from 0.3 mg to 1.0 mg/kg, which can suitably be administered as infusion of 10 ng to too ng per kilogram of body weight per minute. Suitable infusion solutions for these purposes may contain, for example, 0.1 ng to too mg, typically 1 ng to too mg, per milliliter. Single doses may contain, for example, 1 mg to 10 g of the active ingredient. Thus, ampoules for injections may contain, for example, from 1 mg to too mg, and orally administrable single-dose formulations, for example tablets or capsules, may contain, for example, from 1 .0 to 1000 mg, typically from 10 to 600 mg. For treatment of the abovementioned conditions, the compounds of the formula I themselves may be used as the compound, but they are preferably present with a compatible carrier in the form of a pharmaceutical composition. The carrier must of course be acceptable in the sense that it is compatible with the other constituents of the composition and is not harmful to the patient's health. The carrier may be a solid or a liquid or both and is preferably formulated with the compound as a single dose, for example as a tablet, which may contain from 0.05% to 95% by weight of the active ingredient. Other pharmaceutically active substances may likewise be present, including other compounds of formula I. The inventive pharmaceutical compositions can be produced by one of the known pharmaceutical methods, which essentially involve mixing the ingredients with pharmacologically acceptable carriers and/or excipients.

[69] In a second aspect, the invention pertains to a method of producing/ synthesizing the compounds of the invention.

[70] The method of the second aspect preferably comprises at least a step of amide coupling followed by a Sonogashira coupling.

[71] The term "Sonogashira coupling" is art-recognized and refers to the formation of a carbon-carbon bond between a terminal alkyne and an aryl halide or vinyl halide (or their pseudohalide equivalents), with concomitant elimination of the halide.

[72] The term “amide coupling” refers to a formation of a chemical bond (amide bond) between the -COOH group of a carboxylic acid and the -NH2 group of an amine.

[73] The reaction method of the second aspect may in preferred embodiments comprise at least the following reaction (1): [74] Reaction (1)

Wherein R x is And wherein:

R , R 2 , R 3 and R 4 are independently selected from H, an unsubstituted, monosubstituted, or polysubstituted C 1 -C 10 alkyl or heteroalkyl, wherein said alkyl is straight, branched or cyclic, a unsubstituted, monosubstituted or polysubstituted C 1 -C 10 alkenyl or heteroalkenyl, wherein said alkenyl is straight, branched or cyclic, an unsubstituted, monosubstituted, or polysubstituted aryl or heteroaryl, an unsubstituted, monosubstituted, or polysubstituted benzyl group, an acyl group, such as formyl, acetyl, trichloroacetyl, fumaryl, maleyl, succinyl, benzoyl, or acyl groups being branched, heteroatom-substituted or aryl-substituted, a sugar or another acetal, halogen, such as F, Cl, Br, or I, or halogen substituted alkyl or alkoxy such CF 3, OCF 3 , or CHF 2 , is a sulfonyl group, or is S0 2 Me, SMe, S0 2 NH 2 , NHS0 2 Me; A is an aryl or heteroaryl, preferably is substituted or unsubstituted phenyl, tolyl, pyridyl, furanyl, thiophenyl, thiazolyl, pyrimidinyl; n is an integer from 1 to 10,

Y is C or a hetero atom, preferably wherein Y is C or N.

[75] Preferably, A, R R 2 , R 3 and R 4 are defined as for the compound of the first aspect. [76] The method of the second aspect may in preferred embodiments comprise further

(preparatory) steps of : [77] Reaction (2)

[79] In Reaction (2) and (3) R is as defined above. Accordingly it is preferred that the steps indicated in Reactions (1) to (3) are performed preferably in the order a d.

[80] In particular, the method for preparing the compounds of the invention is according to the following description: phenyl chloroformate and hydroxylamine hydrochloride were coupled to yield the protected X- hydroxy carbamate, which subsequently reacted in a Mitsunobu reaction to derivatives i4a-h, which were deprotected by ammonolysis under high pressure of ammonia in an autoclave. In case of optically pure alkyne derivatives 8c, d, the assumption was made, that use of stereochemically pure reagents 13c, d leads to intermediate 14c, d with defined configuration. Sonogashira coupling was performed using alkyne derivatives 8a-h and amides i5a-p,r-aa to yield the final products 7a-ag.

[81] In a third aspect, the invention pertains to a method of inhibiting the enzymatic function of a sEH protein, the method comprising the steps of contacting the sEH protein with the compound of the invention.

[82] Preferably the enzymatic function of sEH is the catalytic metabolization of epoxyeicosatrienoic acids (EETs) to dihydroxyeicosatrienoic acids. In this context it is preferred that the sEH protein is contacted with the compound within a cell.

[83] In a fourth aspect, the invention pertains to a method of inhibiting the enzymatic function of a 5-LOX protein, the method comprising the steps of contacting the 5-LOX protein with the compound of the invention. [84] Preferably the enzymatic function of 5-LOX is the catalytic metabolization of arachidonic acid to 5-hydroperoxyeicosatetraenoic acid (5-HpETE). In this context it is preferred that the 5- LOX protein is contacted with the compound within a cell.

[85] In a fifth aspect, the invention pertains to a method for moderating an inflammatory response in a mammalian subject, the method comprising the administration of a compound of the invention to the mammalian subject.

[86] In a sixth aspect, the invention pertains to a method of treating an inflammatory disorder in a subject in need of the treatment, the method comprising the administration of a compound of the invention to the mammalian subject

[87] The terms “of the [present] invention”, “in accordance with the invention”, “according to the invention” and the like, as used herein are intended to refer to all aspects and embodiments of the invention described and/ or claimed herein.

[88] As used herein, the term “comprising” is to be construed as encompassing both “including” and “consisting of’, both meanings being specifically intended, and hence individually disclosed embodiments in accordance with the present invention. Where used herein, “and/or” is to be taken as specific disclosure of each of the two specified features or components with or without the other. For example, “A and/or B” is to be taken as specific disclosure of each of (i) A, (ii) B and (iii) A and B, just as if each is set out individually herein. In the context of the present invention, the terms “about” and “approximately” denote an interval of accuracy that the person skilled in the art will understand to still ensure the technical effect of the feature in question. The term typically indicates deviation from the indicated numerical value by ±20%, ±15%, ±10%, and for example ±5%. As will be appreciated by the person of ordinary skill, the specific such deviation for a numerical value for a given technical effect will depend on the nature of the technical effect. For example, a natural or biological technical effect may generally have a larger such deviation than one for a man-made or engineering technical effect. As will be appreciated by the person of ordinary skill, the specific such deviation for a numerical value for a given technical effect will depend on the nature of the technical effect. For example, a natural or biological technical effect may generally have a larger such deviation than one for a man-made or engineering technical effect. Where an indefinite or definite article is used when referring to a singular noun, e.g. “a”, “an” or “the”, this includes a plural of that noun unless something else is specifically stated.

[89] It is to be understood that application of the teachings of the present invention to a specific problem or environment, and the inclusion of variations of the present invention or additional features thereto (such as further aspects and embodiments), will be within the capabilities of one having ordinary skill in the art in light of the teachings contained herein.

[90] Unless context dictates otherwise, the descriptions and definitions of the features set out above are not limited to any particular aspect or embodiment of the invention and apply equally to all aspects and embodiments which are described.

[91] All references, patents, and publications cited herein are hereby incorporated by reference in their entirety.

BRIEF DESCRIPTION OF THE FIGURES AND SEQUENCES

[92] The figures show:

[93] Figure 1: shows design of the merged dual inhibitor 7. Clinical candidates 5 (Atreleuton) and 6 (GSK2256294) served as starting point and were subdivided into pharmacophore features (dotted: iron chelating N- hydroxy urea moiety; boxes: lipophilic central core; horizontal lines: amide as epoxide mimetic; vertical lines: terminal aromatic substituent). The recombination of the essential pharmacophore features led to the design of compound 7, which can be retrosynthetically disconnected into three synthons.

[94] Figure 2: shows the evaluation of 7ad in human PMNL and in mice. (A): 5-LOX activity in human PMNL. (B): Pharmacokinetic profiling of 4zd in male Swiss CD-i mice (3 mg/kg dosage, p.o.). 7ad revealed an acceptable bioavailability and a half-life of 1.3 hours.

[95] Figure 3: shows that compound 7ad exhibits anti-inflammatory and anti-fibrotic activities in the UUO model. (A - D) AZAN (A), Sirius Red (B, C) and F4/80 (D) staining demonstrating fibrotic changes and macrophage infiltration in representative kidney sections of ligated (UUO) and contralateral kidneys of vehicle or 7ad-treated mice after 7 days. (B, C) Sirius Red-stained sections were imaged under bright light (Sirius Red) and under polarized light (Sirius Red (Pol)) to detect birefringence of collagen fibers. (E, F) Graphs show the collagen quantification of Sirius Red (E) or macrophage-positive area of F4/8o-stained kidney sections (F) of vehicle (white bars) or 7ad-treated (black bars) mice using ImageJ software version 1.51k. (G, H) Real-time quantitative RT-PCR analysis normalized to GAPDH of fibrosis- associated genes Coliai and FNi (G) and inflammation-associated genes TNFa and CCL2 (H) of whole kidney homogenates of ligated (UUO) and contralateral (Ctrl) kidneys of vehicle (white bars) or 7ad-treated (black bars) mice at day 7. Data are expressed as means ± SEM (E, F). n = 5-6 (E, F). *p < 0.05 and **p < 0.01 compared to UUO kidneys of vehicle-treated mice using two-way analysis of variance and Sidak post hoc test.

[96] The sequences show:

[97] SEQ ID NO: 1 — sEH protein 10 20 30 40 50

MWLEILLTSV LGFAIYWFIS RDKEETLPLE DGWWGPGTRS AAREDDSIRP 60 70 80 90 100

FKVETSDEEI HDLHQRIDKF RFTPPLEDSC FHYGFNSNYL KKVISYWRNE 110 120 130 140 150

FDWKKQVEIL NRYPHFKTKI EGLDIHFIHV KPPQLPAGHT PKPLLMVHGW 160 170 180 190 200

PGSFYEFYKI IPLLTDPKNH GLSDEHVFEV ICPSIPGYGF SEASSKKGFN 210 220 230 240 250 SVATARIFYK LMLRLGFQEF YIQGGDWGSL ICTNMAQLVP SHVKGLHLNM 260 270 280 290 300

ALVLSNFSTL TLLLGQRFGR FLGLTERDVE LLYPVKEKVF YSLMRESGYM 310 320 330 340 350

HIQCTKPDTV GSALNDSPVG LAAYILEKFS TWTNTEFRYL EDGGLERKFS 360 370 380 390 400

LDDLLTNVML YWTTGTIISS QRFYKENLGQ GWMTQKHERM KVYVPTGFSA 410 420 430 440 450

FPFELLHTPE KWVRFKYPKL ISYSYMVRGG HFAAFEEPEL LAQDIRKFLS

VLERQ

[98] SEQ ID NO: 2 - 5-LOX protein

10 20 30 40 50

MPSYTVTVAT GSQWFAGTDD YIYLSLVGSA GCSEKHLLDK PFYNDFERGA 60 70 80 90 100

VDSYDVTVDE ELGEIQLVRI EKRKYWLNDD WYLKYITLKT PHGDYIEFPC 110 120 130 140 150

YRWITGDVEV VLRDGRAKLA RDDQIHILKQ HRRKELETRQ KQYRWMEWNP 160 170 180 190 200

GFPLSIDAKC HKDLPRDIQF DSEKGVDFVL NYSKAMENLF INRFMHMFQS 210 220 230 240 250

SWNDFADFEK IFVKISNTIS ERVMNHWQED LMFGYQFLNG CNPVLIRRCT 260 270 280 290 300

ELPEKLPVTT EMVECSLERQ LSLEQEVQQG NIFIVDFELL DGIDANKTDP 310 320 330 340 350

CTLQFLAAPI CLLYKNLANK IVPIAIQLNQ IPGDENPIFL PSDAKYDWLL 360 370 380 390 400

AKIWVRSSDF HVHQTITHLL RTHLVSEVFG IAMYRQLPAV HPIFKLLVAH 410 420 430 440 450

VRFTIAINTK AREQLICECG LFDKANATGG GGHVQMVQRA MKDLTYASLC 460 470 480 490 500

FPEAIKARGM ESKEDIPYYF YRDDGLLVWE AIRTFTAEW DIYYEGDQW 510 520 530 540 550

EEDPELQDFV NDVYVYGMRG RKSSGFPKSV KSREQLSEYL TW IFTASAQ 560 570 580 590 600

HAAVNFGQYD WCSWIPNAPP TMRAPPPTAK GW TIEQIVD TLPDRGRSCW 610 620 630 640 650

HLGAVWALSQ FQENELFLGM YPEEHFIEKP VKEAMARFRK NLEAIVSVIA 660 670

ERNKKKQLPY YYLSPDRIPN SVAI

EXAMPLES

[99] Certain aspects and embodiments of the invention will now be illustrated by way of example and with reference to the description, figures and tables set out herein. Such examples of the methods, uses and other aspects of the present invention are representative only, and should not be taken to limit the scope of the present invention to only such representative examples.

[100] The examples show:

[101] Example 1: Synthesis of a Novel Class of Dual sEH and 5-LOX Inhibitors

[102] Encouraged by these findings, the inventors designed an orally available dual inhibitor of 5-LOX and sEH in order to enable the investigation of this designed multi target ligand (DML) in different inflammatory in vivo models. Previous attempts yielded DML 2, which exhibits several drawbacks. First, the synthetic access to 2 employs a linear route with an introduction of the X- hydroxy urea moiety within the last three steps with mediocre yields, which hampers the exploration of structure-activity relationships. Second, the flexible linker and the urea functionality are potential drawbacks in terms of metabolic stability and solubility. Therefore, the inventors employed a novel design strategy, incorporating the typical X- hydroxy urea pharmacophore of 5-LOX inhibitors as in 5 (Atreleuton) 21 and the amide moiety which acts as epoxide mimetic in sEH inhibitors like in 6 (GSK2256294) 22 , both compounds advanced in clinical trials. The aforementioned essential pharmacophore groups were merged at the overlapping lipophilic central core and the terminal aromatic moiety, leading to the dual ligand 1 as a starting point for exploration of the structure activity relationship (SAR) (Figure 1). The retrosynthetic disconnection defined the distinct molecular parts for optimization: the central core, the alkyl substituent between the X-hydroxy urea and the alkyne linker, and the terminal aromatic substituent.

[103] The retrosynthetic analysis of compound 7a suggested a convergent synthesis route consisting of two main steps; an amide coupling followed by a Sonogashira coupling (Figure 1). The alkyne derivatives 8a-h were synthesized in a three-step procedure displayed in Scheme 2. First, phenyl chloroformate 11 and hydroxylamine hydrochloride were coupled to yield the protected X-hydroxy carbamate 12, which subsequently reacted in a Mitsunobu reaction to derivatives i4a-h. Deprotection was accomplished by ammonolysis of i4a-h in acceptable yields under high pressure of ammonia in an autoclave. In case of optically pure alkyne derivatives 8c, d, the assumption was made, that use of stereochemically pure reagents 13c, d leads to intermediate 14c, d with defined configuration. This assumption is justified by a closer look of the mechanism of the Mitsunobu reaction, which suggests the inversion of the stereogenic center directly linked to the hydroxyl moiety. To verify this, the derivatives 8c and 8d were coupled with (S)-(+)-a-methoxyphenylacetic acid and the diastereomeric ratio (dr) was determined (dr 9:1 in both cases). Subsequently, Sonogashira coupling was performed using alkyne derivatives 8a-h and amides isa-p,r-aa to yield the final products 7a-ag.

[104] Scheme 2. Synthetic route to the alkyne derivatives 8a-h and the target structures 7a-

7af. a) i) NaHC0 3 in water, hydroxylamine hydrochloride, o °C, 0.5 h, ii) 11, NaHC0 3 in water, rt,

2h; b) PPh 3 , DIAD, dry THF, o °C rt, 1 h; c) NH 3 , tButanohTHF (3:1), -78 °C rt, 5-7 bar, 16- 20 h; d) Pd(ACN) 2 Cl2, PPh 3 , Cul, DIPA, dry EE, rt, 36-48 h.

[105] Starting from acyl chlorides 16a or 16b, the amide coupling was performed directly under microwave irradiation, except for (2-(trifluoromethoxy)phenyl)methanamine loj, where milder reaction conditions were required. The coupling of acids 17a, 17b, 18, and 20 were performed with standard amide coupling reagents as PyBOP and H0Bt-H 2 0, or HBTU and EDC-HCl. In order to facilitate Sonogashira coupling a halogen exchange from bromine (amide 19 and 21) to iodine was accomplished in a similar way to the published procedure of Klapars et al. 23 (Scheme 3).

[106] Scheme 3. Preparation of amide intermediates. a) DCE, DIPEA, 90 °C pw, 1 h; b) loj, 16b, DCM, DIPEA, 4-DMAP, rt, i8h; c) 10a, k-o PyBOP, DIPEA/DIPA, dry THF, rt, 16-20 h; d) ior-u, PyBOP, H0Bt-H 2 0, dry THF, rt, 16 h /60 °C pw, lh. e) methanesulfonyl chloride, NEt 3 , DCM, o reflux, 17 h f) i) 18, HBTU, dry DMF, o °C, ii) 10a, 4-methylmorpholine, EDC-HC1, rt, 72 h; g) Nal, Cul, trans-i,2-cyclohexandiamine, dry 1,4-dioxane, 155 °C pw, 8 h. h) 10a, PyBOP, DIPEA, dry DMF, rt, 48 h.

[107] Evaluation of the inhibitory potential of inhibitors 7 on both targets showed that 7t is the most promising compound for further optimization. To increase the metabolic stability, halogen atoms as well as sulfonamide substituent were introduced to the benzhydryl moiety (Scheme 4). The substituted cinnamic acids 22a, h were converted into the primary amides 23a, b with ethyl chloroformate and ammonia, while the unsubstituted cinnamamide 23c was commercially available. b-Arylation via Heck reaction with palladium(II) acetate yielded 24a-j in acceptable yields and hydrogenation (25a-j) followed by reduction with lithium aluminium hydride gave the amines ioj,k-s. Furthermore, the inventors changed the tertiary carbon atom against nitrogen (7ae). Therefore, in a reductive amination amine 26 was coupled with 2-chloroacetic acid, which was reduced in situ to the corresponding aldehyde. The yielded tertiary amine 27 reacted via a Gabriel reaction to structure 28. Using hydrazine hydrate the phthalimide derivative 28 was cleaved to yield the free amine lot.

[108] Scheme 4. Synthetic route to benzhydryl derived amines. a) i) NEt 3 , ethyl chloroformate, THF, -15 °C, 1 h, ii) 32 % ammonia in water, -15 °C rt, 18 h; b) iodoaryl derivative, Pd(0Ac) 2 , NEt 3 , 100 °C, 16-48 h; c) 10 wt% Pd/C, hydrogen atmosphere (balloon/autoclave), MeOH, rt, 16-48 h; d) 241I or 24g, Pd(0H) 2 , hydrogen atmosphere (balloon), EtOH, rt, 16-48 h; e) LiAlH 4 in THF, dry THF, o°C reflux, 2-5 h; f) i) chloroacetic acid, NaBH 4 , toluene, rt, 3 h, ii) reflux, 4 h; g) potassium phthalimide, DMF, 140 °C pw, lh; h) N 2 H 4 -H 2 0, EtOH, reflux, 16 h. [109] To examine the influence of the second stereogenic center of the unsymmetrically substituted benzhydryl moiety an Evans auxiliary assisted asymmetric synthesis was performed. Zhi et al. 24 published a synthetic route for the asymmetric synthesis of (i?)-tolterodine, which was adapted for the synthesis of IOU (Scheme 5). First the cinnamic acid 22a was converted into the acyl chloride 29 and coupled with the Evans auxiliary. Secondly, 30 was linked with a boronic acid to compound 31 with palladium catalysis. The auxiliary was cleaved with LiBH 4 and the resulted alcohol 32 was oxidized to the aldehyde 33 with Dess-Martin periodinane. Last step was a reductive amination with NaBH(OAc) 3 and ammonia to the amine iou. Amide coupling and Sonogashira coupling led to the (R,R) compound 7af (Scheme 2 and Scheme 3). The (S,R) diastereomer 7ag was obtained via chiral chromatographic separation of 7ab.

[110] Scheme 5. Synthetic route for the asymmetric synthesis of amine precursor iou. a) oxalyl chloride, dry THF, rt, 17 h; b) i) (S)-4-tert-butyl-2-oxazolidinone, nBuLi in THF, - 78 °C, ii) 25, DCM, -78 °C — o °C, 1.5 h, c) (4-fluorophenyl)boronic acid, Pd(0Ac) 2 , bipy, MeOH:water (1:3), 80 °C, 14 h; d) LiBH 4 , Et 2 0, o °C rt, 4 h; e) Dess-Martin periodinane, DCM, rt, 2.5 h, f) NH 4 OAC in EtOH, NaBH(OAc) 3 , 32% ammonia in water, reflux, 13 h.

[111] Example 2: Inhibition of sEH and 5-LOX

[112] The inhibitory potency on both targets were evaluated using enzyme activity assays with recombinantly expressed proteins. For sEH, the conversion of the fluorogenic substrate PHOME (3-phenyl-cyano(6-methoxy-2-naphthalenyl)methyl ester-2-oxiraneacetic acid) was monitored. 25 The enzymatic activity of 5-LOX was accessed using the endogenous substrate AA and monitoring the product formation by HPLC. 26 The prototype inhibitor 1 yielded promising

IC 5o values of 2.6 mM (sEH) and 1.0 pM (5-LOX) and served as a starting point for further optimization. By changing the substitution pattern of the central core from para to meta (compound 7b) the inhibitory potency towards sEH was almost 30-fold increased, together with a slightly better IC 50 value for 5-LOX. An introduction of a heterocyclic moiety as well as a methyl substituent (compounds 7c-e) resulted in a loss of potency on both targets, suggesting that plain 1,3-substituted phenyl core serves best for further optimization efforts.

[113] Table 1. In vitro inhibitory activity of the dual sEH/5-LOX inhibitors - variation of the central aromatic core.

[114] a All values were measured at least thrice as triplicates (n ³ 3). 95% confidence interval is displayed in brackets. b All values were measured at least thrice (n ³ 3). 95% confidence interval is displayed in brackets, n.d. = not determined.

[115] Next optimization steps concerned the alkyl chain adjacent to the - hydroxyurea moiety. The alkyl chain elongation was favored by both enzymes, while the introduction of a hydrogen {jf) led to loss of potency towards 5-LOX. Unbranched chains (71-j) were slightly better tolerated by 5-LOX, while the opposite was observed for sEH. The influence of the stereochemistry was examined with the methyl derivative. The alcohol precursor 13c and 13d was commercially available in both orientations and coupled as shown in Scheme 2. The R derivative 7I1 inhibited the 5-LOX slightly better than the S derivative 7g. In general, the length of the alkyl chain has only small effects on the inhibitory potency towards 5-LOX, therefore further optimization steps were performed with the P- methyl substituent which yielded satisfactory submicromolar IC 50 values towards both enzymes.

[116] Table 2. Influence of the alkyl chain towards the inhibitory potential.

[117] a All values were measured at least thrice as triplicates (n ³ 3). 95% confidence interval is displayed in brackets. b All values were measured at least thrice (n ³ 3). 95% confidence interval is displayed in brackets, c enantiomeric ratio undetermined.

[118] The following optimization step was applied to the terminal aromatic moiety. The development of sEH inhibitors is facilitated by co-crystal structures and chemically diverse published inhibitors. In contrast, crystallographic data of 5-LOX inhibitors is absent. With these circumstances, structure-based development of a 5-LOX inhibitor is hindered, and the inventors decided to use aromatic substituents frequently used in published sEH inhibitors and evaluated the inhibitory potential regarding both enzymes. By keeping the meta substitution at the central ring the inventors started to synthesize several amides with an alkyl linker ending in an aryl moiety. An ethyl linker as well as a meta substitution of the new introduced aryl group is disfavored by both enzymes (7111-0). An ortho or ortho/para substitution pattern seems to be tolerated by the 5-LOX (7p, 7q), but the 2,5-substitution pattern is less tolerated in case of a trifluoromethyl group (71·, 7s) (Table 3). The best IC 50 values on both targets were achieved with a propyl linker with a benzhydryl group (7t), previously published by Eldrup et al. 27 [119] Table 3. Influence of the left aromatic moiety toward the inhibition of both enzymes.

[120] a All values were measured at least thrice as triplicates (n ³ 3). 95% confidence interval is displayed in brackets. b All values were measured at least thrice (n ³ 3). 95% confidence interval is displayed in brackets. [121] As stated in the publication by Eldrup et al., the unsubstituted benzhydryl group is prone to metabolic oxidation. 27 It also holds true for compound t, of which only 27% remained after 1 h of incubation in rat liver microsomes (RLM) (Table 5). In order to enhance the metabolic stability in RLM the inventors introduced halogen and sulfonamide residue in position 4 of one of the phenyl groups. The sulfonamide derivatives (yy, yz ) were disfavored by 5-LOX, in contrast a 3,4-dichlorophenyl ring increased the inhibitory potential (7aa) (Table 4). The combination of two halogenated phenyl rings gave more metabolic stable inhibitors (4ab-4ag). Regarding the sEH inhibitory activity the differentiation between IC 50 values of these compounds is difficult due to the resolution limit of the assay system, where a concentration of 3 nM enzyme is employed. Nevertheless, the substitution on both phenyl rings did not impair the inhibitory activity regarding the sEH, while the inhibitory activity against 5-LOX was slightly decreased. To examine the influence of the second stereogenic center between the unsymmetrically substituted benzhydryl moiety both diastereomers (R,R 4af and R,S 4ag) were prepared and evaluated. 4af exhibited a slightly higher inhibitory activity regarding 5-LOX. [122] Table 4. Structure activity relationship of the introduced moieties raising the metabolic stability.

[123] a diastereomeric ratio undetermined, b R (stereoisomer near the iV-hydroxyurea moiety), diastereomeric ratio undetermined (stereoisomer next to the benzhydryl moiety), c R configuration, d All values were measured at least thrice as triplicates (n ³ 3). 95% confidence interval is displayed in brackets. e All values were measured at least thrice (n ³ 3). 95% confidence interval is displayed in brackets.

[124] Example s: In Vitro Pharmacological Characterization

[125] The symmetric substitution with two fluorine atoms (7ad) avoided a second chiral center and was tolerated by both enzymes. Another strategy to circumvent the second stereogenic center was the replacement of the carbon atom between both phenyl groups by a nitrogen (compound 7ae). This modification also gained similar IC 50 values as 7ad, but the metabolic stability of 7ae in RLM is very low (Table 5). The substitution pattern of both phenyl rings (7ab, 7ac, 7ad) raised the metabolic stability in RLM in comparison to the unsubstituted (7t) and the amine derivative (7ae) (Table 5). For further selection the solubility limit in DPBS (Dulbecco's phosphate-buffered saline) buffer was determined. Therefore, absorption change of a dilution series of the tested compound in buffer was measured and compared to buffer control. The solubility can be considered as moderate and most modifications did not influence the solubility, just 7ac shows a slightly higher solubility (Table 5). Additionally, the inventors determined the toxicity of selected compounds with a CellTiter-Glo® Luminescent Cell Viability Assay Kit. Luciferase catalyzes with the help of ATP Beetle Luciferin to Oxyluciferin and the luminescence is detected by a luminescent reader. The number of viable cells is determined by quantification of ATP, which is an indicator of active cells. Compound 7ad showed no cell toxicity up to a concentration of 25 mM in contrast 7ab exhibited cell toxicity already at 7 mM. Hence, 7ad was selected for a detailed analysis.

[126] Table 5. Microsomal stability and water solubility of selected inhibitors.

[127] Example 4: Cellular and in vivo characterization

[128] The efficacy concerning the 5-LOX was demonstrated on cellular level. Therefore, 7ad was tested in polymorphonuclear leukocytes (PMNLs), where the formation of LOX products 5- HETE and LTB 4 was monitored by HPLC. 7ad could enter the cell and inhibited selectively 5- LOX (Figure 2A, IC 50 = 0.21 pM), and slightly enhanced the activity of 12- and 15-LOX (data not shown). The inhibitory potency in PMNL (polymorphonuclear neutrophils) was comparable to the reference inhibitor 5 (atreleuton).

[129] Encouraged by the previous results, an in vivo study PK/PD study of 7ad in male Swiss CD-i mice was performed. Thereby, an oral single dose of 3 mg/kg was administered to nine mice and three additional mice got vehicle (1% methylcellulose:Tween8o 99:1) as control. The isolated serum was analyzed with LC-MS and showed that ad has an acceptable pharmacokinetic profile (Figure 2B, C max = 0.7 pM, t max = 1 h, t /2 = 1.3 h). Effective concentrations above the IC 50 values of sEH and 5-LOX could be reached over 3-4 h. Furthermore, we analyzed the EET/DHET ratio to verify the sEH inhibition. If the sEH is inhibited, then the EETs should accumulate and the ratio of EET/DHET is increased. This effect could be observed in the plasma samples of the treated mice. The evaluation of 5-LOX target engagement in vivo was not possible in na ' ive mice that were not treated with an inflammatory stimulus. Hence, yad seems to be suitable for inhibiting sEH and 5-LOX in vivo.

[130] The invention provides an orally available potent dual sEH/5-LOX inhibitor. Compound yad was optimized by subsequent variation of the lipophilic parts of the initially designed prototype inhibitor ya considering in vitro inhibitory potency towards both enzymes and in vitro metabolic stability. The SAR analysis was strengthened by the X-ray structure of sEH in complex with y\v and molecular modelling of 5-LOX. Compound yad can be used as a tool to investigate the therapeutic potential of dual sEH/5-LOX inhibitors in vivo, using rodent models of diseases related to acute and chronic inflammation. Furthermore, physiological and pathophysiological consequences of dual inhibition of the CYP and LOX branch of the AA cascade on the lipidome level can be accessed upon application of yad.

[131] Chemistry materials and general procedures: All purchased solvents and chemicals from different suppliers as Sigma Aldrich Chemie GmbH, Acros Organics, Alfa Aesar, fluorochem, TCI Europe were used without further purification. For thin layer chromatography (TLC) TLC plates F254 from Merck were used and aromatic systems were visualized with ultraviolet light (254 and 365 nm). Purification with column chromatography occurred either with silica gel 60 (230-400 mesh ASTM) from Fluka or with a flash system from in Puriflash XS420 from Interchim with columns from Biotage (particle size 50 pm) or Agela technologies (particle size 60 A). NMR spectra were recorded on a DPX250, AV300, AV400 and AV500. The chemical shifts were reported in ppm relative to tetramethylsilane. Spectra were calibrated with the internal signal of non-deuterated solvent. The reported multiplicities were singlet (s), doublet (d), triplet (t), quartet (q) and multiplet (m) and coupling constant (J) were given in hertz (Hz). The purity of the compounds was determined with HPLC (LCMS 2020 from Shimadzu). The used column Luna top CI8(2) 100A (250 x 4.60 mm) and Luna top CI8(2) (250 x 21.20 mm) from Phenomenex were used for analytical and preparative purpose, respectively. UPLC runs were performed with a MultoHigh UC (50 mm x 2 mm) column from CS Chromatography- Service GmbH. Conditions were as followed: eluent ACN/0.1% aqueous formic acid, flow rate was 0.5 mL/min (UPLC), l mL/min (scout column) or 21 mL/min (semi-preparative column) with an UV monitoring at 254 and 280 nm. The specific conditions were described in the experimental procedures of the compounds yag was isolated with a chiral column Chiral cel OJ- RH (4.6 x 150 mm, particle size 5 pm) from Daicel. The eluent of the used HPLC System Agilent 1290 infinity II was ACN and 0.1% aqueous formic acid with a flow rate of 0.5 mL/min. The runs were isocratic with 36% ACN. All final compounds exhibit a purity over 95% at 254 nm. Mass detection occurred either on a LCMS-2020 from shimadzu or a VG platform II from Fison instruments Ltd. High resolution mass was measured in a MALDI LTQ Orbitrap XL instrument from Thermo Scientific.

[132] If the used equivalents differ from the common procedure, the equivalents are listed in the respective experiment.

[133] Phenyl (phenoxycarbonyl)-oxycarbamate (12) was synthesized according published literature: Stewart, A.O., Brooks, D. W. N,0-Bis(phenoxycarbonyl)hydroxylamine: a new reagent for the direct synthesis of substituted N-hydroxyureas. J. Org. Chem. 1992, 57, 5020- 5023 (https://d0i.0rg/10.1021/i000044a046.

[134] Example 5: Characterization of 7ad in a mouse UUO model

[135] TO investigate the therapeutic potential of 7ad, the compound was applied intraperitoneally into mice subjected to unilateral ureter obstruction to trigger kidney inflammation and fibrosis. Treatment with 7ad strongly diminished UUO-induced tubulointerstitial fibrosis as demonstrated by attenuated blue staining of collagen fibers in AZAN-stained sections (Fig. 3A) and reduced fibrillar collagen in Sirius red-stained sections (Fig. 3 B, C, E). Furthermore, the attenuated fibrotic response upon 7ad application was confirmed by diminished mRNA expression of classic fibrogenic markers, such as collagen I (Coital) and fibronectin-i (FNi) (Fig. 3G). The infiltration of macrophages into tubulointerstitial regions correlates with inflammation and fibrosis severity and was therefore assessed using an F4/80 antibody. 35 7ad significantly ameliorated UUO-induced macrophage infiltration into the obstructed kidneys (Fig. 3D, F) and attenuated the mRNA synthesis of Ccl2 (also known as MCPi), an important chemoattractant for macrophages, and of pro- inflammatory TNFa (Fig. 3G). These data suggest the use of 7ad as promising therapeutic approach to treat inflammatory and fibrotic conditions.

[136] General procedure for the synthesis of protected N-hydroxy carbamates i4a-h (Procedure A)

[137] Under an inert atmosphere 1.1 PPh 3 , 0.9 eq alkyne derivative i3a-h and 1 eq of 12 were dissolved in dry THF and cooled to o °C. To this solution 1.1 eq DIAD were added dropwise, while waiting for the decolorization of DIAD between each drop. The solution stirred for 1 h at room temperature. The solvent was evaporated under reduced pressure and the yellow oil was pre-adsorbed on silica gel and purified with column chromatography. An oil was obtained.

[138] Phenyl but-3-yn-2-yl((phenoxycarbonyl)oxy)carbamate 14a: procedure A; 1.67 g (6.36 mmol) PPh 3 , 1.3 mL (6.36 mmol) DIAD, 0.5 mL (5.20 mmol) 13a (3-butyn-2-ol), 1.58 g (5.78 mmol) 12, 25 mL THF; eluent of column chromatography hexane: Et 2 0 3:1-2:15 yield: yellow oil (1.46 g, 4.49 mmol, 87%); Ή-NMR (250 MHz, DMSO-de) d = 7.54 - 7.20 (m, 10H), 5·39 - 5-29 (m, lH), 3.62 (d, J=i.6 Hz, lH), 1.55 (d, J= 6.9 Hz, 3H) ppm; «H-NMR (75 MHz, DMS0-d 6 ) d = 150.4, 130.0, 129.8, 127.0, 126.6, 121.4, 120.7, H5-2, 30.6 ppm.

[139] Phenyl (phenoxycarbonyl)oxy(prop-2-yn-i-yl)carbamate 14b: procedure A; 0.4 mL (6.65 mmol) 13b (2-propyn-i-ol), 1.8 mL (8.87 mmol) DIAD, 2.33 g (8.87 mmol) PPh 3 , 2.02 g (7.39 mmol) 12, 25 mL THF; eluent of column chromatography hexane: Et 2 0 3:i-2:i; yield: transparent oil (2.24 g, 7.20 mmol, 98%); TH-NMR (250 MHz, DMSO-de) <5= 7.53 - 7.43 (m, 4H), 7-40 - 7-32 (m, 4H), 7 2 4 - 7· 17 (m, 2H), 4-76 (d, J=2.4 Hz, 2H), 3-53 (L =2-4 Hz, lH) ppm; MS (ESI) m/z: 312.00 [M + H + ].

[140] (S)-Phenyl but-3-yn-2-yl((phenoxycarbonyl)oxy)carbamate 14c: procedure A; 0.5 mL (6.65 mmol) 13c (i?)-3-butyn-2-ol, 1.7 mL (8.87 mmol) DIAD, 2.33 g (8.87 mmol) PPh 3 , 2.02 g (7.39 mmol) 12, 20 mL THF; eluent of column chromatography hexane: Et 2 o 3:1; yield: white oil (1.99 g, 6.12 mmol, 83%); Ή-NMR (250 MHz, CDC1 3 ) d = 7.59 - 7.42 (m, 4H), 7.42 - 7.27 (m, 4H), 7.24 - 7.19 (m, 2H), 5.33 (qd, J= 2.3, 6.9 Hz, lH), 3.61 (d, J=i.i Hz, lH), 1.56 (d, J= 6.9 Hz, 3H) ppm; MS (ESI) m/z: 325.95 [M + H + ].

[141] (R)-Phenyl but-3-yn-2-yl((phenoxycarbonyl)oxy)carbamate I4d: procedure A; 0.5 mL (6.65 mmol) 13d (GS>3-Butyn-2-ol), 1.7 mL (8.87 mmol) DIAD, 2.33 g (8.87 mmol) PPh 3 , 2.02 g (7.39 mmol) 12, 20 mL THF; eluent of column chromatography hexane: acetone 1:1; yield: white oil (0.47 g, 1.45 mmol, 20%); Ή-NMR (250 MHz, CDC1 3 ) d = 7.51 - 7.34 (m, 4H), 7.33 - 7.10 (m, 6H), 5.30 - 5.20 (m, lH), 2.46 - 2.44 (m, lH), 1.56 (d, J=7.o Hz, 3H) ppm; MS (ESI) m/z: 325.85 [M + H + ].

[142] Phenyl pent-i-yn-3-yl((phenoxycarbonyl)oxy)carbamate 14e: procedure A, 0.1 mL (1.65 mmol) 13e (i-pentyn-3-ol), 0.4 mL (2.20 mmol) DIAD, 0.58 g (2.20 mmol) PPh 3 , 0.50 g (1.83 mmol) 12, 11 mL THF; eluent of column chromatography hexane: Et 2 0 3:i-2:i; yield: yellow oil (0.43 g, 1.26 mmol, 69%); Ή-NMR (250 MHz, DMSO-de) <5= 7.54 - 7.43 (m, 4H), 7.40

- 7.29 (m, 4H), 7.24 - 7.19 (m, 2H), 5.13 - 5.09 (m, lH), 3.62 (d, J=2.i Hz, lH), 1.97 - 1.85 (m, 2H), 1.05 (t, J=7- 3 Hz, 3H) ppm; MS (ESI) m/z: 339.90 [M + H + ].

[143] Phenyl (4-methylpent-i-yn-3-yl)((phenoxycarbonyl)oxy)carbamate I4g: procedure A; 0.7 mL (6.65 mmol) I3g (4-methyl-i-pentyn-3-ol), 1.8 mL (8.87 mmol) DIAD, 2.33 g (8.87 mmol) PPh 3 , 2.02 g (7.39 mmol) 12, 25 mL THF; eluent of column chromatography hexane: Et 2 03:i-2:i; yield: yellow oil (1.35 g, 3.82 mmol, 52%); Ή-NMR (250 MHz, DMS0-d 6 ) d = 7.57

- 7-41 (m, 5H), 7-41 - 7-25 (m, 3H), 7· 22 - 7.17 (m, 2H), 4-91 - 4-89 (m, lH), 3.65 (d, J= 1.9 Hz, lH), 2.24 - 2.17 (m, lH), 1.08 (d, J=7-0 Hz, 6H) ppm; MS (ESI) m/z: 353.85 [M + H + ].

[144] Phenyl (5-methylhex-i-yn-3-yl)((phenoxycarbonyl)oxy)carbamate I4h: procedure A; 0.9 mL (6.65 mmol) I3h (5-methyl-i-hexyn-3-ol), 1. mL (8.87 mmol) DIAD, 2.33 g (8.87 mmol) PPh 3 , 2.02 g (7.39 mmol) 12, 25 mL THF; eluent of column chromatography hexane: E t2 0 4:1- 2:1; yield: transparent oil (1.62 g, 4.41 mmol, 60%); Ή-NMR (250 MHz, DMS0-d 6 ) d = 7.57 - 7.26 (m, 8H), 7.26 - 7.19 (m, 2H), 5.17 - 5.09 (m, lH), 3.64 (d, J= 2.2 Hz, lH), 1.79 (d, J= 9.7 Hz, 3H), 1.01 - 0.91 (m, 6H) ppm; MS (ESI) m/z: 367.90 [M + H + ].

[145] General procedure for the synthesis of deprotected iV-hydroxy ureas 8a-h (Procedure B)

[146] i4a-h were solved and cooled with liquid nitrogen. Ammonia was condensed to this mixture and the solution was transferred to an autoclave. The reaction stirred under an ammonia atmosphere (5-7 bar) for 16-20 h at room temperature. After the reaction time the autoclave was ventilated and stirred for 1 h at ambient conditions to remove the ammonia. The solvent was removed under reduced pressure and the residue was purified via column chromatography.

[147] i-(But-3-yn-2-yl)-i-hydroxyurea 8a: procedure B; 0.38 g (1.17 mmol) 14a, 3 bar NH 3 atmosphere, 4 mL 'butanol, eluent of column chromatography DCM:MeOH 9:1, dirty white solid (0.91 g, 0.71 mmol, 61%); Ή-NMR (250 MHz, DMSO-de) d = 9.22 (s, lH), 6.47 (s, 2H), 4.85 (qd, J= 2.3, 7.0 Hz, lH), 3.03 (d, J= 2.3 Hz, lH), 1.25 (d, J=7.i Hz, 3H) ppm; MS (ESI) m/z: 151.05 [M + Na + ].

[148] i-Hydroxy-i-(prop-2-yn-i-yl)urea 8b: procedure B; 1.17 g 14b, 5 bar NH 3 atmosphere, modified solvent 12 mL ibutanol: c hexane 1:1, eluent of column chromatography DCM:MeOH 9:1, brown solid (0.41 g, 3.58 mmol, 96%); Ή-NMR (250 MHz, DMSO-d 6 ) <5 = 9.48 (s, lH), 6.48 (s, 2H), 4.05 (d, J=2.4 Hz, 2H), 3.05 (t, J=2.4 Hz, lH) ppm; MS (ESI) m/z: 114.90 [M + H + ].

[149] GS)-i-(But-3-yn-2-yl)-i-hydroxyurea 8c: procedure B; 2.75 g (8.44 mmol) 14c; 4.5 bar NH 3 atmosphere, modified solvent 6 mL 'butanohTHF 1:3, eluent of column chromatography DCM:MeOH 95:5-9:1, yellow solid (0.55 g, 4.32 mmol, 51%); Ή-NMR (250 MHz, DMSO-de) <5 = 9.23 (s, lH), 6.49 (s, 2H), 4.86 (qd, J= 2.3, 7.0 Hz, lH), 3.04 (d, J= 2.3 Hz, lH), 1.25 (d, J=7.i Hz, 3H) ppm; MS (ESI) m/z: 129.00 [M + H + ].

[150] (R)-i-(But-3-yn-2-yl)-i-hydroxyurea 8d: procedure B; 1.71 g (5.26 mmol) 141I; 7 bar NH 3 atmosphere, modified solvent 10 mL 'propanol, eluent of column chromatography DCM:MeOH ammonia 98:1-9:1, dirty white solid (0.56 g, 4.35 mmol, 83%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 9.22 (s, lH), 6.47 (s, 2H), 4.85 (qd, J= 2.3, 7.0 Hz, lH), 3.03 (d, J= 2.3 Hz, lH), 1.25 (d, J=7.o Hz, 3H) ppm; MS (ESI) m/z: 129.05 [M + H + ].

[151] i-Hydroxy-i-(pent-i-yn-3-yl)urea 8e: procedure B; 0.42 g (1.24 mmol) 14e, 6 bar NH 3 atmosphere, modified solvent 6 mL THF:hexane 3:1, eluent of column chromatography DCM:MeOH 98:2-9:1, white solid (0.14 g, 0.96 mmol, 77%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 9·ΐ8 (S, lH), 6.46 (s, 2H), 4.62 (td, J=7- 8, 2.3 Hz, lH), 3.05 (d, J= 2.3 Hz, lH), 1.70 - 1.58 (m, 2H), 0.90 (t, J=7A Hz, 3H) ppm; MS (ESI) m/z: 143.05 [M + H + ]. [152] i-(Hex-i-yn-3-yl)-i-hydroxyurea 8f: procedure B; 0.65 g (1.83 mmol) I4f, 6 bar NH 3 atmosphere, modified solvent 6 mL THF:hexane 3:1, eluent of column chromatography DCM:MeOH 98:2-9:1, white solid (0.15 g, 0.96 mmol, 52%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 9.17 (s, lH), 6.45 (s, 2H), 4.72 (td, J=7-7, 2.3 Hz, lH), 3.03 (d, J= 2.3 Hz, lH), 1.78 - 1.16 (m, 4H), 0.87 (t, J=7.3 Hz, 3H) ppm; MS (ESI) m/z: 156.60 [M + H + ].

[153] i-Hydroxy-i-(4-methylpent-i-yn-3-yl)urea 8g: procedure B; 1.33 g (3.75 mmol) I4g, 7 bar NH 3 atmosphere, modified solvent 12 mL 'butanol: hexane 1:1, eluent of column chromatography DCM:MeOH 100:0-9:1, dirty white solid (0.53 g, 3.37 mmol, 90%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 9.14 (s, lH), 6.44 (s, 2H), 4.36 (dd, J= 2.3, 9.9 Hz, lH), 3.05 (d, J= 2.3 Hz, lH), 1.99 - 1.82 (m, lH), 1.00 (d, J=6.8 Hz, 3H), 0.88 (d, J= 6.6 Hz, 3H) ppm; MS (ESI) m/z: 156.90 [M + H + ].

[154] i-hydroxy-i-(5-methylhex-i-yn-3-yl)urea 8h: procedure B; 1.60 g (3.75 mmol) 14I1, 4 bar NH 3 atmosphere, modified solvent 12 mL 'butanol: hexane 1:1, eluent of column chromatography DCM:MeOH 100:0-9:1, dirty yellow solid (0.53 g, 3.37 mmol, 82%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 9.17 (s, lH), 6.46 (s, 2H), 4.79 (dt, J= 2.3, 7.8 Hz, lH), 3.03 (d, J= 2.3 Hz, lH), 1.87 - 1.35 (m, 3H), 0.87 (dd, J= 3.7, 6.8 Hz, 6H) ppm; MS (ESI) m/z: 170.90 [M + H + ].

[155] General procedure for the synthesis of amide derivatives i5a-j from acyl chlorides (Procedure G)

[156] 1 eq 4-iodobenzoyl chloride 16a or 3-iodobenzoyl chloride 16b was dissolved in dry 1,2- dichloroethan (DCE). At o °C 1 eq of the corresponding amine loa-i and 3 eq DIPEA were added and heated to 90 °C under microwave irradiation in a sealed tube. After cooling to room temperature the solvent was removed, and the residue was resolved in ethyl acetate. The organic phase was washed with water (3x), saturated aqueous NaHC0 3 solution (2x), 2 M aqueous HC1 (2x) and brine (lx). After drying over MgS0 4 and filtration the organic solvent was evaporated under reduced pressure. The solid was purified with column chromatography.

[157] 4-Iodo-V-(2-(trifliioromethyl)benzyl)benzamide 15a: procedure G; 0.63 g (2.36 mmol) 16a, 0.3 mL (2.36 mmol) 10a, 0.8 mL (4.37 mmol) DIPEA, 3 mL DCE; white solid (0.81 g, 2.00 mmol, 84%); Ή-NMR (250 MHz, DMSO-de) d = 9.17 (t, J= 5.7 Hz, lH), 7.92 - 7.87 (m, 2H), 7.75 - 7.62 (m, 4H), 7.53 - 7.45 (m, 2H), 4.65 (d, J=5.8 HZ, 2H) ppm; MS (ESI) m/z: 405.80 [M + H + ].

[158] 3-Iodo-A/-(2-(trifluoromethyl)benzyl)benzamide 15b: procedure G; 0.33 g (1.24 mmol, 0.7 eq) 16b, 0.2 mL (i.8immol) 10a, 0.6 mL (3.72 mmol, 3.0 eq) DIPEA, 5 mL DCE; white solid (0.44 g, 1.10 mmol, 88%), Ή-NMR (250 MHz, DMSO-de) d = g.ig (t, J= 5.7 Hz, lH), 8.28 (t, J=i.6 Hz, lH), 7.93 (dd, J= 1.7, 7.8 Hz, 2H), 7.81 - 7.59 (m, 2H), 7.59 - 7.41 (m, 2H), 7.31 (t, J=7.8 Hz, lH), 4.65 (d, J= 5.6 Hz, 2H) ppm; MS (ESI) m/z: 405.70 [M + H + ]. [159] A/-(2-(iii-Indol-3-yl)ethyl)-3-iodobenzamide 15c: procedure G; 0.29 g (1.09 mmol) 16b, 0.38 g (2.40 mmol, 2.2 eq) 2-(iif-indol-3-yl)ethanamine (10b), 1.1 mL (6.54 mmol) DIPEA; eluent of column chromatography hexane:ethyl acetate 1:3, 5 mL DCE; brownish solid (0.33 g, 0.84 mmol, 77%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 10.82 (s, lH), 8.69 (t, J= 5.6 Hz, lH), 8.18 (t, J= 1.6 Hz, lH), 7.94 - 7.72 (m, 2H), 7.60 - 7-54 (m, lH), 7.40 - 6.92 (m, 5H), 3.59 - 3.47 (m, 2H), 2.94 (t, J=7A Hz, 2H) ppm; MS (ESI) m/z: 389.01 [M - H + ].

[160] A/-(4-Fluorophenethyl)-3-iodobenzamide I5d: procedure G; 0.48 g (1.81 mmol, 1.6 eq) 16b, 0.1 mL (1.13 mmol) 3-fluorophenethylamine (10c), 0.6 mL (3.39 mmol, 3.0 eq) DIPEA, , 5 mL DCE; eluent of column chromatography hexane: acetone 2:1; yellow oil (0.40 g, 1.09 mmol, 96%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 8.62 (t, J=54 Hz, lH), 8.14 (t, J=i.6 Hz, lH), 7.93 - 7-73 (m, 2H), 7-34 - 7-19 (m, 3H), 7-15 - 7·q6 (m, 2H), 3-50 - 3-40 (m, 2H), 2.82 (t, J=7- 3 Hz, 2H) ppm; MS (ESI) m/z: 369.80 [M + H + ].

[161] A/-(3-Fluorobenzyl)-3-iodobenzamide 15e: procedure G; 0.48 g (1.81 mmol, 1.6 eq) 16b, 0.1 mL (1.13 mmol) 3-fluorobenzylamine (lod), 0.6 mL (3.39 mmol, 3.0 eq) DIPEA, 5 mL DCE; eluent of column chromatography hexane:acetone 2:1; yellow oil (0.38 g, 1.08 mmol, 95%); Ή- NMR (250 MHz, DMS0-d 6 ) <5 = 9.15 (t, J= 5.7 Hz, lH), 8.24 (t, J=i.6 Hz, lH), 7.98 - 7.83 (m, 2H), 7.44 - 7.23 (m, 2H), 7.22 - 6.94 (m, 3H), 4.47 (d, J= 5.9 Hz, 2H) ppm; MS (ESI) m/z: 353.80 [M - H + ].

[162] A/-(2,4-Dichlorobenzyl)-3-iodobenzamide I5f; procedure G, 0.53 g (1.99 mmol, 1.6 eq) 16b, 0.2 mL (1.24 mmol) (2,4-dichlorophenyl)methanamine (loe), 0.6 mL (3.72 mmol, 3.0 eq) DIPEA, 5 mL DCE; white solid (0.40 g, 1.20 mmol, 97%); Ή-NMR (250 MHz, DMS0-d 6 ) d = 9.15 (t, =54 Hz, lH), 8.25 (t, 1.6 Hz, lH), 8.06 - 7.82 (m, 2H), 7.62 (d, J=i.8 Hz, lH), 7.58 -

7.25 (m, 3H), 4.50 (d, J= 5.6 Hz, 2H) ppm; MS (ESI) m/z: 405.70 [M + H + ].

[163] 3-Iodo-V-(4-methoxy-2-(trifliioromethyl)benzyl)benzamide I5g: procedure G; 048 g

(1.81 mmol, 1.5 eq) 16b, 0.25 g (1.24 mmol) (4-methoxy-2-

(trifluoromethyl)phenyl)methanamine (lof), 0.6 mL (372 mmol, 3.0 eq) DIPEA, 5 mL DCE; white solid (046 g, 1.05 mmol, 85%); Ή-NMR (250 MHz, DMS0-d 6 ) <5 = 9.10 (t, J= 5.6 Hz, lH),

8.25 (t, J=i.6 Hz, lH), 7.94 - 7.88 (m, 2H), 748 - 742 (m, lH), 7.34 - 7.20 (m, 3H), 4.56 (d, d=5-5 Hz, 2H), 3.81 (s, 3H) ppm; MS (ESI) m/z: 435.80 [M + H + ].

[164] V-(4-Fliioro-2-(trifliioromethyl)benzyl)-3-iodobenzamide ish: procedure G; 048 g (1.81 mmol) 16b, 0.3 mL (i.8immol) (4-fluoro-2-(trifluoromethyl)-phenyl)methanamine (log), 0.7 mL (4.11 mmol, 3.0 eq) DIPEA, 5 mL DCE; eluent of column chromatography hexane:acetone 2:1; yellow oil (0.51 g, 1.21 mmol, 89%); Ή-NMR (250 MHz, DMS0-d 6 ) d =9.20 (t, J=5-3 Hz, lH), 8.28 - 8.24 (m, lH), 7.97 - 7.89 (m, 2H), 7.67 - 7.50 (m, 3H), 7.31 (t, J=7· 8 Hz, lH), 4.61 (d, J= 5.3 Hz, 2H) ppm; MS (ESI) m/z: 423.75 [M + H + ]. [165] A/-(3,3-Diphenylpropyl)-3-iodobenzamide 151: procedure G; 0.33 g (1.24 mmol) 16b, 0.26 g (1.24 mmol) 3,3-diphenylpropan-i-amine (loh), 0.6 mL (3.72 mmol, 3.0 eq) DIPEA, 5 mL DCE; white solid (0.42 g, 0.94 mmol, 76%), Ή-NMR (250 MHz, DMSO-de) <5 = 8.55 (t, J= 5-2 Hz, lH), 8.15 (t, =i.6 Hz, lH), 7.90 - 7.79 (m, 2H), 7.31 - 7.25 (m, 11H), 4.08 - 3.99 (m, lH), 3.22 - 3.10 (m, 2H), 2.30 (q, J=7.5 Hz, 2H) ppm; MS (ESI) m/z: 441.80 [M + H + ].

[166] A/-(3-(4-Fluorophenyl)-3-phenylpropyl)-3-iodobenzamide 15 : procedure G; 0.18 g (0.69 mmol) 16b, 0.16 g (0.69 mmol) 3-(4-fluorophenyl)-3-phenylpropan-i-amine (101), 0.4 mL (2.05 mmol) DIPEA, 3 mL DCE, eluent of column chromatography hexane:ethyl acetate 3:1; white solid (0.13 g, 0.28 mmol, 41%), Ή-NMR (250 MHz, DMSO-d 6 ) d = 8.55 (t, J= 5.2 Hz, lH), 8.15 (t, =i.6 Hz, lH), 7.90 - 7.79 (m, 2H), 7.31 - 7.25 (m, 10H), 4.08 - 3.99 (m, lH), 3.22 - 3.10 (m, 2H), 2.30 (q, J=7- 5 Hz, 2H) ppm; MS (ESI) m/z: 460.00 [M + H + ].

[167] 3-Iodo-V-(2-(trifliioromethoxy)benzyl)benzamide 15k: To a solution of 190 mg (0.71 mmol) 16b in 5 mL DCM 0.4 mL (2.14 mmol, 3 eq) (2- (trifluoromethoxy)phenyl)methanamine (loj) and 2 mg (0.01 mmol, 0.02 eq) 4-DMAP were added. Additionally, 0.4 mL (2.14 mmol, 3 eq) DIPEA was added and the mixture stirred for 18 h at room temperature. The organic phase was washed with water (2x), 2 M aqueous HC1 (2x) and brine (lx). After drying over MgS0 4 and filtration the organic solvent was evaporated under reduced pressure. A brown solid was obtained (0.29 g, 0.68 mmol, 95%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 9.11 (t, J= 5.7 Hz, lH), 8.25 (t, J=i.6 Hz, lH), 7.92 (td, J= 1.5, 7.8 Hz, 2H), 7.69 - 7.19 (m, 5H), 4.53 (d, J=5.8 HZ, 2H) ppm; MS (ESI) m/z: 421.75 [M + H + ].

[168] General procedure for the synthesis of amide derivatives i5l-q (Procedure H)

[169] 1 eq acid derivative 17a or 17b were dissolved in dry THF and 1.8 eq PyBOP were added. 1.1 eq corresponding amine 10 and 2 eq DIPEA or DIPA, dissolved in dry THF, were added. The reaction stirred for 16-20 h at room temperature, afterwards the solvent was removed. The organic phase was washed with water (3x), saturated aqueous NaHC0 3 solution (2x) and brine (lx). After drying over MgS0 4 and filtration the organic solvent was evaporated under reduced pressure. The solid was purified with column chromatography.

[170] 3-Iodo-4-methyl-V-(2-(trifliioromethyl)benzyl)benzamide 15I: procedure H; 0.15 g (0.57 mmol) 10a, 0.15 g (0.57 mmol) 3-iodo-4-methyl-benzoic acid (17b), 0.53 g (1.02 mmol) PyBOP, 0.2 mL (1.14 mmol) DIPA in 15 mL THF; white solid (0.19 g, 0.45 mmol, 79%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 9.13 (t, J= 5.7 Hz, lH), 8.37 (d, J=i.8 Hz, lH), 7.86 (dd, J=i.8, 7.9 Hz, lH), 7.80 - 7.58 (m, 2H), 7.58 - 7.39 (m, 3H), 4.64 (d, J= 5.5 Hz, 2H), 2.42 (s, 3H) ppm; MS (ESI) m/z: 419.75 [M + H + ].

[171] 3-Iodo-V-(3-phenyl-3-(4-(trifliioromethyl)phenyl)propyl)benz amide 15m: procedure H; 0.13 g (0.47 mmol) 3-phenyl-3-(4-(trifluoromethyl)phenyl)propan-i-amine (10k), 0.12 g (0.47 mmol) 17a, 0.36 g (0.70 mmol) PyBOP, 0.1 mL (0.36 mmol) DIPA in 10 mL THF; eluent of column chromatography hexane:ethyl acetate 3:1; transparent oil (0.17 g, 0.33 mmol, 72%); Ή-NMR (250 MHz, acetone-ch) d = 8.20 - 8.i8 (m, lH), 8.04 - 7.72 (m, 3H), 7.72 - 7.53 (m, 4H), 7-43 - 7-14 (m, 6H), 4.26 (t, J=7- 8 Hz, lH), 3.41 - 3.34 (m, 2H), 2.52 - 2.41 (m, 2H) ppm; MS (ESI) m/z: 509.80 [M + H + ].

[172] 3-Iodo-lV-(3-phenyl-3-(4-(trifluoromethoxy)phenyl)propyl)ben zamide 15h: procedure H; 0.15 g (0.52 mmol) 3-phenyl-3-(4-(trifluoromethoxy)phenyl)propan-i-amine (10I), 0.13 g (0.52 mmol) 17a, 0.30 g (0.57 mmol) PyBOP, 0.3 mL (1.55 mmol) DIPA in 13 mL THF; white solid (0.25 g, 0.47 mmol, 90%); Ή-NMR (250 MHz, acetone-d 6 ) d = 8.19 (t, 7=1.7 Hz, lH), 7.90

- 7-83 (m, 3H), 7-52 - 7-45 (m, 2H), 7.40 - 7 1 8 (m, 8H), 4.21 (t, 7=7.8 Hz, lH), 3.42 - 3.32 (m, 2H), 2.49 - 2.38 (m, 2H) ppm; MS (ESI) m/z: 525.85 [M + H + ].

[173] A/-(3-(4-Chlorophenyl)-3-phenylpropyl)-3-iodobenzamide 150: procedure H; 86 mg (350 pmol) 3-(4-chlorophenyl)-3-phenylpropan-i-amine (10m), 86 mg (350 pmol) 17a, 237 mg (525 pmol) PyBOP, 0.1 mL (700 pmol) DIPA, 17 mL THF, 2 mL DMF; eluent of flash chromatography hexane:ethyl acetate 3:1; white solid (112 mg, 235 pmol, 67%); Ή-NMR (250 MHz, acetone-d 6 ) d = 8.19 (t, J=i.8 Hz, lH), 7.89 - 7.83 (m, 3H), 7.41 - 7.15 (m, 10H), 4.15 (t, J=8.i Hz, lH), 3.41 - 3.31 (m, 2H), 2.47 - 2.37 (m, 2H) ppm; MS (ESI) m/z: 475-85 [M + H + ].

[174] A/-(3-(3,4-Dichlorophenyl)-3-phenylpropyl)-3-iodobenzamide I5p: procedure H; 0.50 g (1.78 mmol) 3-(3,4-dichlorophenyl)-3-phenylpropan-i-amine (ion), 0.45 g (1.78 mmol) 17a, 1.02 g (1.96 mmol) PyBOP, 0.9 mL (5.34 mmol) DIPA, 10 mL THF, 1 h 60 °C microwave irradiation; eluent of flash chromatography hexane:acetone 4:1; yellow oil (0.86 g, 1.69 mmol, 95%); Ή-NMR (250 MHz, acetone-d 6 ) d = 8.i8 (t, J= 2.5 Hz, lH), 7.93 - 7.78 (m, 2H), 7.56 (d, J= 2.5 Hz, lH), 7.50 - 7-46 (m, lH), 7.44 - 7.13 (m, 8H), 4.19 (t, J=7-5 Hz, lH), 3.42 - 3.34 (m, 2H), 2.48 - 2.39 (m, 2H) ppm; MS (ESI) m/z: 511.05 [M + H + ].

[175] A/-(3-(4-aminophenyl)-3-phenylpropyl)-3-iodobenzamide I5q: 0.23 g (0.89 mmol, 1 eq) 3-iodobenzoic acid, 0.46 g (0.89 mmol, 1 eq) PyBOP were suspended in dry THF and 0.3 mL (1.78 mmol, 2 eq) DIPEA added. The solution stirred for 13 h at room temperature. The solution was added slowly to 0.20 g (0.89 mmol) 4-(3-amino-i-phenylpropyl)aniline (100), solved in 8 mL dry THF via a syringe. The mixture stirred for 1 h at room temperature. The solvent was evaporated under reduced pressure and the residue was solved in ethyl acetate. The organic phase was washed with saturated aqueous NaHC0 3 solution (2x), with saturated aqueous NH 4 C1 solution (2x) and brine (lx). After drying over MgS0 4 and filtration the organic solvent was removed under reduced pressure. The solid was purified with column chromatography hexane:acetone 4:i-2:i and a transparent oil was obtained (0.10 g, 0.23 mmol, 25%); Ή-NMR (300 MHz, acetone-d 6 ) d = 8.19 (t, J= 1.7 Hz, lH), 7.88 - 7.77 (m, 3H), 7.31 - 7.22 (m, 6H), 7.04 - 6.99 (m, 2H), 6.62 - 6.57 (m, 2H), 4.44 (bs, 2H), 3.94 (t, J=7-8 Hz, lH), 3.39 - 3.31 (m, 2H), 2.37

- 2.29 (m, 2H) ppm; MS (ESI) m/z: 457.03 [M + H + ]. [176] 3-Iodo-A/-(3-(4-(methylsulfonamido)phenyl)-3-phenylpropyl)be nzamide 151·: To a solution of 103 mg (226 pmol, 1 eq) I5q in 10 mL dry DCM 18 pL (234 pmol, 1 eq) methanesulfonyl chloride were added at o °C. 64 pL (456 pmol, 2 eq) NEt 3 were added and the solution stirred for 17 h under reflux conditions. The mixture was allowed to cool to room temperature and 2 M aqueous HC1 was added. The organic phase was separated, and the aqueous phase was extracted with ethyl acetate. The organic phases were combined, dried over MgS0 4 and filtered. After the removal of the solvent the residue was purified with column chromatography (hexane:acetone 2:1). A transparent oil was obtained (108 mg, 202 pmol, 89%); Ή-NMR (250 MHz, acetone-d 6 ) <5 = 8.49 (bs, lH), 8.21 (t, J= 1.7 Hz, lH), 7.87 (dd, 7=1.7, 7.8 Hz, 2H), 7-37 - 7-21 (m, 11H), 4.11 (t, «7=7.8 Hz, lH), 3.45 - 3.33 (m,2H), 2.93 (s, 3H), 2.47 - 2.36 (m, 2H). ppm; MS (ESI) m/z: 533.02 [M - H + ].

[177] General procedure for the synthesis of amide derivatives i5s-y (Procedure I)

[178] 1 eq 17a, 1.1 eq PyBOP, 0.5 eq H0Bt-H 2 0 and 2 eq DIPEA were dissolved in dry THF and 1 eq of the corresponding amine 10, solved in dry THF, was added portionwise. The mixture was either heated 1 h via microwave irradiation at 60 °C or stirred 16-20 h at room temperature. The solvent was evaporated, and the residue was purified with column chromatography.

[179] 3-Iodo-A/-(3-phenyl-3-(4-sulfamoylphenyl)propyl)benzamide 15s: procedure I; 0.80 g (0.31 mmol) 17a, 0.90 g (0.31 mmol) 4-(3-amino-i-phenylpropyl)benzene-sulfonamide (lop), 0.24 g (0.47 mmol) PyBOP, 0.07 g (0.47 mmol) H0Bt-H 2 0, 0.2 mL (0.93 mmol) DIPEA, 2 mL THF, 1 h 60 °C microwave irradiation; eluent of flash chromatography hexane:acetone 3:2; yellow oil (0.15 g, 0.29 mmol, 95%); Ή-NMR (300 MHz, acetone-d 6 ) d = 8.21 - 8.20 (m, lH), 7.90 - 7.80 (m, 5H), 7.56 - 7.52 (m, 2H), 7.40 - 7.17 (m, 6H), 6.47 (bs, 2H), 4.29 (t, «7=7.3 Hz, lH), 3.42 - 3.34 (m, 2H), 2.51 - 2.43 (m, 2H) ppm; MS (ESI) m/z: 543.06 [M + Na + ].

[180] A/-(3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)propyl)- 3-iodobenzamide I5t: procedure I; 0.20 g (0.79 mmol) 17a, 0.24 g (0.79 mmol) 3-(4-fluorophenyl)-3-(4- (trifluoromethyl)phenyl)propan-i-amine (loq), 0.45 g (0.87 mmol) PyBOP, 0.06 g (0.40 mmol) H0Bt-H 2 0, 0.3 mL (1.58 mmol) DIPEA, 35 mL THF, room temperature 16 h; eluent of column chromatography hexane:acetone 2:1; yellow solid (0.31 g, 0.58 mmol, 74%); Ή-NMR (300 MHz, acetone-d 6 ) d = 8.20 (td, J= 1.7, 0.4 Hz, lH), 7.92 - 7.75 (m, 3H), 7.69 - 7.54 (m, 4H), 7.44 - 7.39 (m, 2H), 7.26 (t, J=7.8 HZ, lH), 7.10 - 7.04 (m, 2H), 4.29 (t, J=7-8 Hz, lH), 3.42 - 3.34 (m, 2H), 2.50 - 2.42 (m, 2H) ppm; MS (ESI) m/z: 550.00 [M + Na + ].

[181] A/-(3-(4-Fluorophenyl)-3-(4-(trifluoromethoxy)phenyl)propyl) -3-iodobenzamide 15U: procedure I; 0.27 g (1.06 mmol) 17a, 0.33 g (1.06 mmol) 3-(4-fluorophenyl)-3-(4- (trifluoromethoxy)phenyl)propan-i-amine (lor), 0.61 g (1.16 mmol) PyBOP, 0.08 g (0.53 mmol) H0Bt-H 2 0, 0.4 mL (1.12 mmol) DIPEA, 8 mL THF, 1 h 60 °C microwave irradiation; eluent of column chromatography hexane:ethyl acetate 9:1-3:!; transparent oil (q·55 g, l.Ol mmol, 96%); Ή-NMR (300 MHz, DMSO-de) d = 8.56 (t, J= 5.2 Hz, lH), 8.15 (t, J=i.6 Hz, lH), 7.97 - 7.76 (m, 2H), 7.52 - 7.20 (m, 7H), 7.12 (t, J= 8.9 Hz, 2H), 4.14 (t, =7-4 Hz, lH), 3.24 - 3.12 (m, 2H), 2.33 - 2.23 (m, 2H) ppm; MS (ESI) m/z: 542.10 [M - H + ].

[182] A/-(3,3-Bis(4-fluorophenyl)propyl)-3-iodobenzamide 15V: procedure I; 0.20 g (0.79 mmol) 17a, 0.20 g (0.79 mmol) 3,3-bis(4-fluorophenyl)propan-i-amine (10s), 0.45 g (0.87 mmol) PyBOP, 0.06 g (0.40 mmol) H0Bt-H 2 0, 0.3 mL (1.58 mmol) DIPEA, 35 mL THF, room temperature 18 h; eluent of column chromatography hexane:acetone 2:1; transparent oil (0.32 g, 0.67 mmol, 85%); Ή-NMR (300 MHz, acetone-de) d = 8.19 (t, J=i.6 Hz, lH), 7.89 - 7.85 (m, 3H), 742 - 7-23 (m, 5H), 7-10 - 7-00 (m, 4H), 4-17 (t, J=7- 8 Hz, lH), 3.40 - 3-31 (m, 2H), 2.45 - 2.34 (m, 2H) ppm; MS (ESI) m/z: 500.07 [M + Na + ].

[183] A/-(2-(Diphenylamino)ethyl)-3-iodobenzamide 15x1 procedure I; 0.60 g (0.24 mmol) 17a, 0.50 g (0.24 mmol) N 1 , A^-Diphenylethan-i, 2-diamine lot, 0.14 g (0.26 mmol) PyBOP, 0.02 g (0.12 mmol) H0Bt-H 2 0, 0.1 mL (0.47 mmol) DIPEA, 30 mL THF, room temperature 24 h; eluent of column chromatography hexane:ethyl acetate 3:i-2:i; transparent oil (0.10 g, 0.23 mmol, 96%); lH-NMR (250 MHz, acetone-d 6 ) d = d = 8.17 (t, J=i.6 Hz, lH), 8.01 (bs, lH), 7.90 - 7.83 (m, 2H), 7.31 - 7.23 (m, 5H), 7.11 - 7.06 (m, 4H), 6.97 - 6.91 (m, 2H), 4.02 - 3.96 (m, 2H), 3.72 - 3.62 (m, 2H) ppm; MS (ESI) m/z: 465.10 [M + Na + ].

[184] (R)-A/-(3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)prop yl)-3-iodobenzamide I5y: procedure I; 43 mg (0.17 mmol) 17a, 50 mg (0.17 mmol) (R)-3-(4-fluorophenyl)-3-(4- (trifluoromethyl)phenyl)propan-i-amine (IOU), 131 mg (0.25 mmol) PyBOP, 40 mg (0.25 mmol) H0Bt-H 2 0, 0.1 mL (0.50 mmol) DIPEA, 10 mL THF, room temperature 18 h; eluent of column chromatography hexane:ethyl acetate 9:i-3:i; transparent oil (55 mg, 0.10 mmol, 63%); Ή-NMR (250 MHz, acetone-d 6 ) d = 8.19 (t, J=i.6 Hz, lH), 7.90 - 7.84 (m, 3H), 7-67 - 7-56 (m, 4H), 746 - 7-39 (m, 2H), 7 2 6 (t, J=7- 8 Hz, lH), 7.11 - 7.03 (m,2H), 4-30 (t, J=7.8 Hz, lH), 3.43 - 3.33 (m, 2H), 2.51 - 2.41 (m, 2H) ppm; MS (ESI) m/z: 549.96 [M + Na + ].

[185] 5-Bromo-V-(2-(trifluoromethyl)benzyl)nicotinamide 19: Under an argon atmosphere 0.24 g (1.17 mmol, 1 eq) 18, 0.67 (1.76 mmol, 1.5 eq) HBTU were dissolved in 15 mL dry DMF and cooled to o °C. To the solution 0.23 g (1.29 mmol, 1.1 eq) 10a, 0.7 mL (7.02 mmol, 6.0 eq) 4-methylmorphline and 0.34 g (1.76 mmol, 1.5 eq) EDC-HC1 were added. The approach stirred for 3 d at room temperature. The solvent was evaporated, and the residue was suspended in water. The aqueous phase was extracted with ethyl acetate (4x) and the combined organic phase was washed with brine. After drying over MgS0 4 and filtration the solvent was removed, and the crude product was purified via flash chromatography (hexane:ethyl acetate 2:1). A white solid was obtained (0.27 g, 0.75 mmol, 64%); Ή-NMR (300 MHz, DMS0-d 6 ) d = 9.39 (t, J=5-6 Hz, lH), 9.04 (d, J=i.8 Hz, lH), 8.89 (d, J= 2.2 Hz, lH), 8.50 (t, J= 2.1 Hz, lH), 7.77 - 7.46 (m, 4H), 4.67 (d, J=5-5 Hz, 2H) ppm; MS (ESI) m/z: 358.90 [M + H + ]. [186] 5-Iodo-/V-(2-(trifhioromethyl)benzyl)nicotinamide 15Z: Under argon atmosphere 260 mg (0.72 mmol, 1 eq) 19, 217 mg (1.45 mmol, 2 eq) dry Nal, 7 mg (0.04 mmol, 0.05 eq) Cul were suspended in 3 mL dry 1,4-dioxane. 8 pL (0.08 mmol, 0.1 eq) trans- 1,2- cyclohexandiamine was added and the approach was heated to 155 °C via microwave irradiation for 8 h. The suspension was allowed to cool to room temperature and diluted with ethyl acetate and aqueous ammonia (10%). The aqueous phase was extracted with ethyl acetate (3x), the combined organic phase was dried over MgS0 4 and filtered. The solvent was evaporated, and the crude product was used without further purification.

[187] 5-Bromo-lV-(2-(trifluoromethyl)benzyl)furan-2-carboxamide 21: 0.17 g (0.87 mmol, 1 eq) 20 were dissolved in 10 mL dry DMF and 0.3 mL (2.28 mmol, 2.6 eq) 10a, 0.7 mL (3.90 mmol, 4.4 eq) DIPEA and 0.54 g (1.04 mmol, 1.2 eq) PyBOP were added. The solution stirred for 2 d at room temperature. The approach was concentrated and diluted with DCM. The organic phase was washed with saturated aqueous NaHC0 3 solution (3x), dried over MgS0 4 and filtered. The solvent was evaporated, and the residue was purified with flash chromatography (hexane: ethyl acetate 3:1) yielding a brownish solid (0.17 g, 0.49 mmol, 55%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 9.05 (t, J=4.8 Hz, lH), 7.74 - 7.63 (m, 2H), 7.51 - 7.45 (m, 2H), 7.22 (d, J= 3.6 Hz, lH), 6.79 (d, J= 3.6 Hz, lH), 4.60 (d, J=4.8 Hz, 2H) ppm; MS (ESI) m/z: 349.70 [M + H + ].

[188] 5-Iodo-A/-(2-(trifluoromethyl)benzyl)furan-2-carboxamide I5aa: Under argon atmosphere 150 mg (0.43 mmol, 1 eq) 21, 129 mg (0.86 mmol, 2 eq) dry Nal, 4 mg (0.02 mmol, 0.05 eq) Cul were suspended in 3 mL dry 1,4-dioxane. 6 pL (0.04 mmol, 0.1 eq) trans- 1,2- cyclohexandiamine was added and the approach was heated to 110 °C via microwave irradiation for 21 h. The suspension was allowed to cool to room temperature and diluted with ethyl acetate and aqueous ammonia (10%). The aqueous phase was extracted with ethyl acetate (3x), the combined organic phase was dried over MgS0 4 and filtered. The solvent was evaporated, and the crude product was used without further purification.

[189] General procedure for the synthesis of target structures 7a-7af (Procedure C)

[190] 1 eq iodo derivative, 1.1 eq alkyne derivative, 0.02 eq Bis(acetonitrile)dichloropalladium (II), 0.04 eq Cul and 0.04 eq PPh 3 were suspended in dry ethyl acetate. 1-12 eq DIPA were added and the mixture stirred for 36-48 h at room temperature. The suspension was washed with water (3x), the combined aqueous phase was extracted with ethyl acetate (lx). The combined organic phase was washed with brine (lx) and dried over MgS0 4 . The MgS0 4 was filtered off and the solvent was evaporated under reduced pressure. The residue was purified with column chromatography and most derivative were purified further with semi-preparative HPLC to gain over 95% purity. [191] 4-(3-(i-Hydroxyureido)but-i-yn-i-yl)-V-(2-(trifluoromethyl)b enzyl)benzamide 7a: procedure C; 320 mg (0.79 mmol) 15a, 110 mg (0.78 mmol) 8a, 4 mg (0.02 mmol) Pd(ACN) 2 Cl 2 , 6 mg (0.03 mmol) Cul, 8 mg (0.03 mmol) PPh 3 , 0.1 mL (0.95 mmol, 1.2 eq) DIPA, 18 mL ethyl acetate, 36 h, eluent of flash chromatography hexane:ethyl acetate 1:2, orange solid (280 mg, 0.69 mmol, 88%); Ή-NMR (250 MHz, DMSO-de) <5 = 9.38 (s, lH), 9.17 (t, J= 5.8 Hz, lH), 7.94 - 7.89 (m, 2H), 7.76 - 7.61 (m, 2H), 7.56 - 7.46 (m, 4H), 6.56 (s, 2H), 5.17 (q, J=7. o Hz, lH), 4.66 (d, J=54 Hz, 2H), 1.38 (d, J=7-0 Hz, 3H) ppm; «C-NMR (75 MHz, DMSO-d 6 ) d = 165.8, i6ΐ·4, 1374, 137-3, 1334, 132-7, I3ΐ·4, 129-3, 128.3, 127.6, 127.3, 126.3, 125-9, 125-7 (q), 125.5, -92-2, 81.2, 45.9, 184 ppm; purity (HPLC-MS): 99%, tiu 346 min (method: gradient of 50% ACN to 90% within 10 min); HRMS (MALDI): m/z calculated for G 20 H 18 F 3 N 3 O 3 + H + [M + H + ]: 406.13730; found: 406.13664.

[192] 3-(3-(i-Hydroxyureido)but-i-yn-i-yl)-/V-(2-(trifluoromethyl) benzyl)benzamide 7b: procedure C; 120 mg (0.30 mmol) 15b, 40 mg (0.33 mmol) 8a, 2 mg (0.01 mmol) Pd(ACN) 2 Cl 2 ,

2 mg (0.01 mmol) Cul, 3 mg (0.01 mmol) PPh 3 , 0.1 mL (0.36 mmol, 1.2 eq) DIPA, 8 mL ethyl acetate, 20 h, further purification with preparative HPLC , white solid (96 mg, 0.24 mmol, 79%); Ή-NMR (400 MHz, DMSO-d 6 ) d = 9-36 (s, lH), 9.21 (t, J= 5.6 Hz, lH), 7.97 (s, lH), 7.89 (d, J=7- 8 Hz, lH), 7.80 - 741 (m, 6H), 6.56 - 6.53 (m, 2H), 5.16 (q, J=7.o Hz, lH), 4.67 - 4.63 (m, 2H), 1.38 (d, J=7.o Hz, 3H) ppm; «C-NMR (100.6 MHz, DMSO-de) d = 165.7, i6i-4, 1374, 134-3, 134-2, 132.7, 130.1, 128.8, 128.3, 1274, 127-3, 125-8 (q), 122.7, 90.7, 81.08, 45.8, 18.5 ppm; purity (UPLC-MS): 99%, tiu 3.57 min (method: gradient of 25% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated for C 2O H I8 F 3 N 3 0 3 + H + [M + H + ]: 406.13730; found: 406.13709.

[193] 5-(3-(i-Hydroxyureido)but-i-yn-i-yl)- -(2-(trifluoromethyl)benzyl)nicotinamide 7c: procedure C; 140 mg (0.35 mmol) 15Z, 50 mg (040 mmol) 8a, 2 mg (0.01 mmol) Pd(ACN) 2 Cl 2 ,

3 mg (0.01 mmol) Cul, 4 mg (0.01 mmol) PPh 3 , 0.1 mL (043 mmol, 1.2 eq) DIPA, 10 mL ethyl acetate, 49 h, further purification with flash chromatography (hexane: ethyl acetate i:i);white solid (110 mg, 0.27 mmol, 76%); Ή-NMR (300 MHz, DMSO-de) d = 944 - 9.37 (m, 2H), 9.01 (s, lH), 8.74 (s, lH), 8.30 (t, J= 2.0 Hz, lH), 7.76 - 745 (m, 4H), 6.60 (s, 2H), 5.20 (q, J=7.o Hz, lH), 4.67 (d, J=54 Hz, 2H), 140 (d, J=7-0 Hz, 3H) ppm; «C-NMR (75 MHz, DMSO-d 6 ) d = 164.3, I6I-4, 153-8, 147-7, 137-3, 137-0 (d), 132.8, 129.1, 128.7, 127-5, 1264 (d), 126.0, 125.8 (q), 122.7, 119-2, 94.1, 78.1, 46.0, 184 ppm; purity (UPLC-MS): 97%, tiu 2.98 min (method: gradient of 25% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated for C gH 7 F 3 N 4 0 3 + H + [M + H + ]: 407.13255; found: 407.13251.

[194] 5-(3-(i-Hydroxyureido)but-i-yn-i-yl)- -(2-(trifluoromethyl)benzyl)furan-2- carboxamide 7d: procedure C; 140 mg (0.35 mmol) I5aa, 50 mg (040 mmol) 8a, 2 mg (0.01 mmol) Pd(ACN) 2 Cl 2 , 3 mg (0.01 mmol) Cul, 4 mg (0.01 mmol) PPh 3 , 0.1 mL (043 mmol,

1.2 eq) DIPA, 10 mL ethyl acetate, 48 h, further purification with preparative HPLC (5 min 30% ACN, gradient from 30% ACN to 80% within 15 min), white solid (4 mg, 0.01 mmol, 3%); Ή- NMR (500 MHz, DMS0-d 6 ) <5 = 9.45 (s, lH), 9.17 (t, J= 6.0 Hz, lH), 7.72 (d, J=7-7 Hz, lH), 7.67 - 7.64 (m, lH), 7.50 - 7.45 (m, 2H), 7.18 (d, J= 3.5 Hz, lH), 6.90 (d, J= 3.6 Hz, lH), 6.61 (s, 2H), 5.18 (q, J=7.i Hz, lH), 4.59 (d, J= 5.8 Hz, 2H), 1.38 (d, J=7.i Hz, 3H) ppm; «C-NMR (100.6 MHz, DMSO-d 6 ) d = 161.9, 157.7, 148.2, 137.7, 137-7, 133-2, 129.2, 128.6, 127.8, 126.6, 126.3, 126.2 (q), 126.0, 123.9, 117-5, 1154, 96.2, 72.3, 464, 18.8 ppm; purity (UPLC-MS): 97%, t R : 3.39 min (method: gradient of 20% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated for C 18 H 16 F 3 N 3 O 4 + K + [M + K + ]: 434.07245; found: 434.07242.

[195] 3-(3-(i-Hydroxyureido)but-i-yn-i-yl)-4-methyl- -(2-(trifluoromethyl)benzyl)- benzamide 7e: procedure C; 90 mg (0.22 mmol) 15I, 31 mg (0.24 mmol) 8a, 1 mg (0.004 mmol, 0.03 eq) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol, 0.04 eq) Cul, 5 mg (0.01 mmol, 0.09 eq) PPh 3 , 0.1 mL (0.26 mmol, 1.2 eq) DIPA, 12 mL ethyl acetate, 39 h, further purification with column chromatography (ethyl acetate 100%); yellow solid (42 mg, 0.10 mmol, 47%); Ή-NMR (300 MHz, DMSO-d 6 ) d = 9-36 (s, lH), 9.15 (t, J= 5.7 Hz, lH), 7.94 (d, J=i.8 Hz, lH), 7.85 - 7.33 (m, 6H), 6.59 (s, 2H), 5.18 (q, J=7- o Hz, lH), 4.65 (d, J= 5.4 Hz, 2H), 2.41 (s, 3H), 1.41 (d, J=7- o Hz, 3H) ppm; «C-NMR (75 MHz, DMSO-d 6 ) d = 165.7, i6ΐ·7, 143-3, 137-6 (d), 132.7, 131-6, 130.2, 129.6, 128.3, 127.3, 126.3, 125.9, 125.7 (q), 122.5, 94-6, 80.0, 46.1, 20.2, 18.8 ppm; purity (UPLC-MS): 98% t R : 3.99 min (method: gradient of 20% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated C 2I H 2O F 3 N 3 0 3 + H + [M + H + ]: 420.15295; found: 420.15267.

[196] 3-(3-(i-Hydroxyureido)prop-i-yn-i-yl)- -(2-(trifluoromethyl)benzyl)benzamide 7f: procedure C; 100 mg (0.25 mmol) 15b, 30 mg (0.27 mmol) 8b, 1 mg (0.005 mmol) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol) Cul, 3 mg (0.01 mmol) PPh 3 , 0.1 mL (0.30 mmol, 1.2 eq) DIPA, 15 mL ethyl acetate, 39 h, further purification with column chromatography (hexane:ethyl acetate 4:i-o:i);brown solid (50 mg, 0.13 mmol, 52%); Ή-NMR (300 MHz, DMSO-d 6 ) d = 9.62 (s, lH), 9.23 (t, J= 5.7 Hz, lH), 8.00 (t, J=i.4 Hz, lH), 7.92 (td, J= 1.3, 7-8 Hz, lH), 7.77 - 7.43 (m, 6H), 6.56 (s, 2H), 4-66 (d, J=54 Hz, 2H), 4-35 (s, 2H) ppm; «C-NMR (75 MHz, DMSO-d 6 ) d = 165.7, i6i.6, 137-4, 134-3 (d), 132.7, 132.1, 131.5 (d), 130.2, 128.9, 128.8, 128.7, 128.3, 127-5, 127.3, 126.3, 125.8 (q), 122.6, 87.1, 81.8 ppm; purity (HPLC-MS): 97% t R : 7.62 min (method: gradient of 30% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C gH 6 F 3 N 3 0 3 + H + [M + H + ]: 392.12165; found: 392.12133.

[197] GS)-3-(3-(i-Hydroxyureido)but-i-yn-i-yl)- -(2-(trifluoromethyl)benzyl)benzamide 7g: procedure C; 80 mg (0.20 mmol) 15b, 30 mg (0.22 mmol) 8c, 1 mg (0.004 mmol) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol) Cul, 3 mg (0.01 mmol) PPh 3 , 0.1 mL (0.24 mmol, 1.2 eq) DIPA, 11 mL ethyl acetate, 43 h, further purification with column chromatography (hexane:acetone 2:3), further purification with preparative HPLC (5% ACN to 90% within 10 min, 90% ACN for 6 min);white solid (27 mg, 0.07 mmol, 34%); Ή-NMR (500 MHz, acetone-de) d = 8.69 (s, lH), 8.42 (t, J=5-3 Hz, lH), 8.05 - 7-91 (m, 2H), 7.73 (d, J=7- 9 Hz, lH), 7.70 - 7-54 (m, 3H), 7-53 - 7-43 (m, 2H), 6.13 (s, 2H), 5.27 (q, J=7.o Hz, lH), 4.83 (d, J= 5.7 Hz, 2H), 1.45 (d, J=7-0 Hz, 3H) ppm; «C-NMR (125 MHz, acetone-d 6 ) d = i66.8, 162.0, 135.7, 1334, 131 2 , 129.8, 129.5, 128.2, 128.0, 126.6 (q), 124.4, 90.8, 82.2, 47.3, 40.6 (q), 18.6 ppm; purity (HPLC-MS): 97% tiu 9.66 min (method: gradient of 10% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 20 H 18 F 3 N 3 O 3 + H + [M + H + ]: 406.13730; found: 406.13715.

[198] (R)-3-(3-(i-Hydroxyureido)but-i-yn-i-yl)-A/-(2-(trifluoromet hyl)benzyl)benzamide 7I1: procedure C; 80 mg (0.20 mmol) 15b, 30 mg (0.22 mmol) 8d, 1 mg (0.004 mmol) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol) Cul, 3 mg (0.01 mmol) PPh 3 , 0.1 mL (0.24 mmol, 1.2 eq) DIPA, 15 mL ethyl acetate, 43 h, further purification with column chromatography (hexane:acetone 2:3), further purification with preparative HPLC (linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min);white solid (29 mg, 0.07 mmol, 36%); Ή-NMR (500 MHz, acetone- d 6 ) d = 8.69 (s, lH), 8.42 (t, J= 5.4 Hz, lH), 8.02 - 7.91 (m, 2H), 7.76 - 7.52 (m, 4H), 7.52 - 7.43 (m, 2H), 6.14 (s, 2H), 5.27 (q, J=7.o Hz, lH), 4.83 (d, J= 5.7 Hz, 2H), 1.45 (d, J=7- o Hz, 3H) ppm; «C-NMR (125 MHz, acetone-de) d = 166.9, I62.O, 138.6, 138.6, 135.7, 135.1, 133 , 131.2, 129.8, 129.5, 128.2, 128.0, 126.6 (q), 124.4, 90.8, 82.2, 47.3, 40.6 (q), 18.6 ppm; purity (HPLC- MS): 97% tiu 9.65 min (method: gradient of 10% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 2O H I8 F 3 N 3 0 3 + H + [M + H + ]: 406.13730; found: 406.13723.

[199] 3-(3-(i-Hydroxyureido)pent-i-yn-i-yl)- -(2-(trifluoromethyl)benzyl)benzamide 71: procedure C; 80 mg (0.20 mmol) 15b, 30 mg (0.22 mmol) 8e, 1 mg (0.004 mmol) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol) Cul, 3 mg (0.01 mmol) PPh 3 , 0.1 mL (0.24 mmol, 1.2 eq) DIPA, 15 mL ethyl acetate, 43 h, further purification with column chromatography (hexane:acetone 2:3), further purification with preparative HPLC (linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min);white solid (58 mg, 0.14 mmol, 70%); Ή-NMR (500 MHz, acetone-d 6 ) d = 8.67 (s, lH), 8.43 (t, J= 6.0 Hz, lH), 8.00 (t, 7=1.5 Hz, lH), 7.94 (td, J= 1.5, 7.8 Hz, lH), 7.74 - 7.71 (m, lH), 7.69 - 7-55 (m, 3H), 7-53 - 743 (m, 2H), 6.13 (s, 2H), 5.05 (t, J=7- 8 Hz, lH), 4.83 (d, J= 5.7 Hz, 2H), 1.91 - 1.83 (m, 2H), 1.02 (t, =74 Hz, 3H) ppm; ^C-NMR (125 MHz, acetone-d 6 ) d = I66·9, I62.3, 138.6, 138.6, 135.7, i35 i , 133-3, 131.2, 129.8, 129.5, 128.2, 128.0, 126.7 (q), 126.6, 124.4, 89.9, 83.0, 53.3, 40.6 (q), 26.6, 11.1 ppm; purity (HPLC-MS): 97% tiu 10.03 min (method: gradient of 10% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 2I H 2O F 3 N 3 0 3 + H + [M + H + ]: 420.15295; found: 420.15279.

[200] 3-(3-(i-Hydroxyureido)hex-i-yn-i-yl)- -(2-(trifluoromethyl)benzyl)benzamide j : procedure C; 80 mg (0.20 mmol) 15b, 30 mg (0.22 mmol) 8f, 1 mg (0.004 mmol) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol) Cul, 2 mg (0.01 mmol) PPh 3 , 0.1 mL (0.24 mmol, 1.2 eq) DIPA, 15 mL ethyl acetate, 43 h, further purification with column chromatography (hexane:acetone 2:3), further purification with preparative HPLC (linear gradient from 5% ACN to 90% within 10 min, 90%

ACN for 6 min);white solid (57 mg, 0.13 mmol, 67%); Ή-NMR (500 MHz, acetone-d 6 ) d = 8.65 (s, lH), 8.43 (t, J=5-5 Hz, lH), 8.12 - 7.76 (m, 2H), 7.73 (d, J=7- 9 Hz, lH), 7.69 - 7.55 (m, 3H), 7-52 - 7-34 (m, 2H), 6.12 (s, 2H), 5.16 (t, J=7-7 Hz, lH), 4.83 (d, J= 5.7 Hz, 2H), 1.83 (q, J=7-7 Hz, 2H), 1.53 - 1.45 (m, 2H), 0.95 (t, J=7A Hz, 3H) ppm; «C-NMR (125 MHz, acetone-de) d = I66·9, 102.2, 138.6, 138.6, 135.7, I35·ΐ, 133-3, I3ΐ·2, 129.8, 129.5, 128.2, 128.0, 126.8, 126.6 (q), 124.6, 124.4, 90.0, 82.9, 5ΐ·4, 40.6 (q), 35-3, 20.0, 13.9 ppm; purity (HPLC-MS): 97% t R : 10.51 min (method: gradient of 10% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 22 H 22 F 3 N 3 O 3 + Na + [M + Na + ]: 456.15055; found: 456.15027.

[201] 3-(3-(i-Hydroxyureido)-4-methylpent-i-yn-i-yl)-A/-(2-(triflu oromethyl)benzyl)- benzamide 7k: procedure C; 100 mg (0.25 mmol) 15b, 42 mg (0.27 mmol) 8g, 1 mg (0.005 mmol) Pd(ACN) 2 Cl 2 , 6 mg (0.03 mmol, 0.13 eq) Cul, 11 mg (0.04 mmol, 0.17 eq) PPh 3 , 0.1 mL (0.30 mmol, 1.2 eq) DIPA, 15 mL ethyl acetate, 39 h, further purification with column chromatography (ethyl acetate 100%), further purification with preparative HPLC (30% ACN for 3 min, linear gradient from 30% ACN to 80% within 8 min, 90% ACN for 6 min); white solid (20 mg, 0.05 mmol, 19%); Ή-NMR (300 MHz, DMS0-d 6 ) d = 9.36 - 9.15 (m, 2H), 8.06 - 7.15 (m, 7H), 6.53 (s, 2H), 4.79 - 4.55 (m, 3H), 3.68 - 3.11 (m, lH), 2.12 - 1.97 (m, lH), 1.10 (d, J=6.8 Hz, 3H), 0.95 (d, J= 6.5 Hz, 3H) ppm; «C-NMR (125 MHz, DMSO-de) d = 165.7, i6i.8, 137.5 (d), 134-3, 134-2, 132.8, 130.1, 128.9, 128.3, 127-4 (d), 126.3, 125 8 (q), 122.8, 89.3, 82.5, 56.7, 30.5, 20.0, 19.4 ppm; purity (UPLC-MS): 99% t R : 3.93 min (method: gradient of 20% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated C 22 H 22 F 3 N 3 O 3 + H + [M + H + ]: 434.16860; found: 434.16810.

[202] 3-(3-(i-Hydroxyureido)-5-methylhex-i-yn-i-yl)-N-(2-(trifluor omethyl)benzyl)- benzamide 7I: procedure C; 100 mg (0.25 mmol) 15b, 46 mg (0.27 mmol) 8h, 1 mg (0.005 mmol, 0.03 eq) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol) Cul, 11 mg (0.04 mmol, 0.17 eq) PPh 3 , 0.1 mL (0.30 mmol, 1.2 eq) DIPA, 15 mL ethyl acetate, 39 h, further purification with column chromatography (DCM:MeOH 9:1); yellow solid (62 mg, 0.14 mmol, 56%); Ή-NMR (300 MHz, DMS0-d 6 ) d = 9-33 (s, lH), 9.24 (t, J=5.8 Hz, lH), 7.97 - 7.87 (m, 2H), 7.76 - 7.44 (m, 6H), 6.56 (s, 2H), 5.09 (t, J=7.8 HZ, lH), 4.65 (d, J= 5.4 Hz, 2H), 1.81 - 1.59 (m, 3H), 0.92 (dd, =3-5, 6.5 Hz, 6H) ppm; «C-NMR (125 MHz, DMSO-de) d = 165.7, i6i.6, 137.4, 134-3, 134-2, 132.7, 130.1, 128.9, 128.3,127.4 (d), 126.3, 125-8 (q), 122.7, 89.8, 81.7, 48.5, 40.9, 24.3, 22.2 (d) ppm; purity (HPLC-MS): 96% t R : 10.96 min (method: gradient of 10% ACN to 90% within 10 min, hold 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 23 H 24 F 3 N 3 0 3 + H + [M + H + ]: 448.18425; found: 448.18368.

[203] -(2-(iH-Indol-3-yl)ethyl)-3-(3-(i-hydroxyureido)but-i-yn-i-y l)benzamide 7m: procedure C; 150 mg (0.38 mmol) 15c, 52 mg (0.41 mmol) 8a, 3 mg (0.01 mmol, 0.03 eq) Pd(ACN) 2 Cl 2 , 5 mg (0.03 mmol, 0.07 eq) Cul, 5 mg (0.02 mmol) PPh 3 , 0.1 mL (0.44 mmol, 1.2 eq) DIPA, 16 mL ethyl acetate, 24 h, purification with flash chromatography (DCM:MeOH

100:0-9:1), further purification with preparative HPLC (linear gradient from 20% ACN to 90% within 10 min, 90% ACN for 10 min); white solid (47 mg, 0.12 mmol, 34%); Ή-NMR (300 MHz, DMS0-d 6 ) d = io.8o (s, lH), 9.37 (s, lH), 8.72 (t, J= 5.6 Hz, lH), 7.89 - 7.81 (m, 2H), 7.60 - 7.42 (m, 3H), 7.36 - 7.31 (m, lH), 7.17 (d, J= 2.2 Hz, lH), 7.10 - 6.94 (m, 2H), 6.56 (s, 2H), 5.15 (q, J=7.0 Hz, lH), 3.58 - 3.48 (m, 2H), 2.94 (t, =7-4 Hz, 2H), 1.38 (d, J=7-0 Hz, 3H) ppm; «C- NMR (75 MHz, DMS0-d 6 ) d = 136.2, 135.0, 130.0, 128.7, 127-3, 122.6, 120.9, n8.22, 111.9, ni-4, 45.8, 25.1, 18.6 ppm; purity (UPLC-MS): 99% t R : 3.03 min (method: gradient of 20% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated C 22 H 22 N 4 O 3 + H + [M + H + ]: 391.17647; found: 391.17631.

[204] -(4-Fluorophenethyl)-3-(3-(i-hydroxyureido)but-i-yn-i-yl)ben zamide 7h: procedure C; 80 mg (0.21 mmol) 151I, 29 mg (0.23 mmol) 8a, 2 mg (0.007 mmol, 0.03 eq) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol, 0.06 eq) Cul, 3 mg (0.01 mmol, 0.06 eq) PPh 3 , 0.1 mL (0.25 mmol, 1.2 eq) DIPA, 9 mL ethyl acetate, 20 h, purification with preparative HPLC (30% ACN for 5 min, linear gradient from 30% ACN to 80% within 10 min); white solid (30 mg, 0.08 mmol, 39%); Ή-NMR (300 MHz, DMSO-d 6 ) d = 9-37 (s, lH), 8.65 (t, 7=5.5 Hz, lH), 7.84 - 7.75 (m, 2H), 7-54 - 7-41 (m, 2H), 7-30 - 7-23 (m, 2H), 7-14 - 7-07 (m, 2H), 6.55 (s, 2H), 5.15 (¾ J=7- o Hz, lH), 3.51 - 3.41 (m, 2H), 2.83 (t, « 7=7-2 Hz, 2H), 1.38 (d, 7=7. o Hz, 3H) ppm; «C-NMR (75 MHz, DMSO-d 6 ) d = 165-3, i6i-5, 135-7 (d), 134-8, 133-8, 130.5, 130.4, 129-9, 128.7,127.2, 122.5, H5-i, H4-8, 90.6,

81.1, 45.8, 40.9, 34.1, 18.5 ppm; purity (UPLC-MS): 99% t R : 2.89 min (method: gradient of 20% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated C 20 H 20 FN 3 O 3 + H + [M + H + ]: 370.15615; found: 370.15644.

[205] -(3-Fluorobenzyl)-3-(3-(i-hydroxyureido)but-i-yn-i-yl)benzam ide 70: procedure C; 70 mg (0.21 mmol) 15e, 29 mg (0.23 mmol) 8a, 2 mg (0.007 mmol, 0.03 eq) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol, 0.05 eq) Cul, 3 mg (0.01 mmol, 0.06 eq) PPh 3 , 0.1 mL (0.25 mmol, 1.2 eq) DIPA, 10 mL ethyl acetate, 41 h, purification with preparative HPLC (30% ACN for 5 min, linear gradient from 30% ACN to 80% within 10 min); white solid (16 mg, 0.05 mmol, 22%); Ή-NMR (300 MHz, DMS0-d 6 ) d = 9.38 (s, lH), 9.21 (t, J= 5.9 Hz, lH), 7.99 - 7.79 (m, 2H), 7.61 - 7.31 (m, 3H), 7-23 - 6.98 (m, 3H), 6.59 (s, 2H), 5-15 (q, 7=7- o Hz, lH), 4.47 (d, 7= 5.9 Hz, 2H), 1.37 (d, « 7=7-0 Hz, 3H) ppm; «C-NMR (75 MHz, DMSO-d 6 ) d = 165.5, 142.6 (d), 134.5, 134-0, 130.3 (d),

130.1, 128.9, 127-4, 123-3, 122.6, 114.1, 113.8 (d), 113.4, 90.6, 81.1, 45.8, 42.3, 18.5 ppm; purity (UPLC-MS): 98% t R : 2.93 min (method: gradient of 20% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated C gH 8 FN 3 0 3 + K + [M + K + ]: 394.09638; found: 394.09612.

[206] 3-(3-(i-Hydroxyureido)biit-i-yn-i-yl)-/V-(2-(trifhioromethox y)benzyl)benzamide jp: procedure C; 98 mg (0.23 mmol) 15k, 33 mg (0.26 mmol, 1.2 eq) 8a, 2 mg (0.01 mmol, 0.03 eq) Pd(ACN) 2 Cl 2 , 2 mg (0.02 mmol, 0.07 eq) Cul, 6 mg (0.02 mmol, 0.1 eq) PPh 3 , 0.1 mL (0.30 mmol, 1.2 eq) DIPA, 10 mL ethyl acetate, 5 mL THF, 41 h, purification with column chromatography (hexane: ethyl acetate 4:1), further purification with preparative HPLC (linear gradient from 10% ACN to 90% within 10 min); off-white solid (60 mg, 0.14 mmol, 61%); Ή- NMR (300 MHz, DMSO-d 6 ) d = 9.36 (s, lH), 9.15 (t, J= 5.7 Hz, lH), 7.94 - 7.85 (m, 2H), 7.58 - 7.34 (m, 6H), 6.56 (s, 2H), 5.15 (q, J= . o Hz, lH), 4.53 (d, J= 5.9 Hz, 2H), 1.37 (d, J=7-i Hz, 3H) ppm; «C-NMR (75 MHz, DMSO-d 6 ) d = 165.5, i6ΐ·4, I46.i (d), 134.4, 134-L I3ΐ·7, I30.i, 129.3, 128.8, 128.7, 127-5, 122.6, 120.4 (q), 90.6, 81.1, 45-8, 18.5 ppm; purity (HPLC-MS): 96% t R : 9.86 min (method: gradient of 10% ACN to 90% within 10 min); HRMS (MALDI): m/z calculated C 20 H l8 F 3 N 3 0 4 + H + [M + H + ]: 422.13222; found: 422.13171.

[207] A/-(2,4-Dichlorobenzyl)-3-(3-(i-hydroxyureido)but-i-yn-i-yl) benzamide 7q: procedure C; 75 mg (0.19 mmol) I5f, 27 mg (0.23 mmol, 1.2 eq) 8a, 2 mg (0.01 mmol, 0.04 eq) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol, 0.06 eq) Cul, 3 mg (0.01 mmol, 0.07 eq) PPh 3 , 0.1 mL (0.22 mmol, 1.2 eq) DIPA, 9 mL ethyl acetate, 20 h, purification with preparative HPLC (linear gradient from 50% ACN to 80% within 10 min); white solid (41 mg, 0.10 mmol, 55%); Ή-NMR (500 MHz, DMSO-d 6 ) d = 9.37 (s, lH), 9.17 (t, J= 5.7 Hz, lH), 7.94 (s, lH), 7.87 (dd, J= 1.3, 7.8 Hz, lH), 7.63 - 7-6O (m, lH), 7.59 - 7.54 (m, lH), 7.49 (t, J=7-7 Hz, lH), 7.43 - 7.36 (m, 2H), 6.55 (s, 2H), 5.15 (q, J= . o Hz, lH), 4.50 (d, J= 5.7 Hz, 2H), 1.38 (d, 7=7. o Hz, 3H) ppm; «C-NMR (125 MHz, DMSO-d 6 ) d = 165.6, 161.5, 135.4, 134-2 (d), 132.9, 132.3 (d), 132.1 (d), 131.5, 131.4, 130.2 (d), 128.9 (d), 128.7, 128.6, 127.4 (d), 122.7, 90.7, 81.1, 45.8, 18.6 ppm; purity (UPLC-MS): 99% t R : 3.37 min (method: gradient of 20% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated CigH 17 Cl 2 N 3 0 3 + H + [M + H + ]: 406.07197; found: 406.07188.

[208] 3-(3-(i-Hydroxyureido)but-i-yn-i-yl)- -(4-methoxy-2-(trifluoromethyl)benzyl)- benzamide 71·: procedure C; 74 mg (0.17 mmol) I5g, 29 mg (0.23 mmol, 1.3 eq) 8a, 2 mg (0.007 mmol, 0.04 eq) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol, 0.07 eq) Cul, 3 mg (0.01 mmol, 0.07 eq) PPh 3 , 0.1 mL (0.20 mmol, 1.2 eq) DIPA, 9 mL ethyl acetate, 20 h, purification with preparative HPLC (30% ACN 1 min, linear gradient from 30% ACN to 80% within 6 min, linear gradient to 90% ACN within 4 min); white solid (22 mg, 0.05 mmol, 30%); Ή-NMR (300 MHz, DMS0-d 6 ) d = 9-39 (s, lH), 9.16 (t, J= 5.6 Hz, lH), 7.97 - 7.88 (m, 2H), 760 - 7.44 (m, 3H), 7.26 - 7.22 (m, 2H), 6.58 (s, 2H), 5-17 (¾ J=7- o Hz, lH), 4.59 (d, J=5-3 Hz, 2H), 3 83 (s, 3H), 1.39 (d, J=7- o Hz, 3H) ppm; «C-NMR (75 MHz, DMSO-de) d = 165.6, 161.4, 158.0, 1344, 134-1, 130.5, 130.1, 128.8, 127.4, 127.1, 126.0, 122.6, 122.4, 117-7, in-5 (q), 90.6, 81.1, 55.6, 45.8, i8.6.ppm; purity (UPLC- MS): 99% t R : 3.53 min (method: gradient of 20% ACN to 90% within 5 min); HRMS (MALDI): m/z calculated C 2i H 20 F 3 N 3 0 4 + H + [M + H + ]: 436.14787; found: 436.14737.

[209] -(4-fluoro-2-(trifluoromethyl)benzyl)-3-(3-(i-hydroxyureido) but-i-yn-i-yl)benzamide 7s: procedure C; 100 mg (0.24 mmol) 15I1, 33 mg (0.26 mmol) 8a, 3 mg (0.01 mmol, 0.05 eq) Pd(ACN) 2 Cl 2 , 4 mg (0.02 mmol, 0.09 eq) Cul, 7 mg (0.03 mmol, 0.1 eq) PPh 3 , 0.1 mL (0.28 mmol, 1.2 eq) DIPA, 10 mL ethyl acetate, 5 mL THF, 41 h, purification with column chromatography (hexane: ethyl acetate 1:4), further purification with preparative HPLC (linear gradient from 10% ACN to 90% within 10 min); white solid (65 mg, 0.15 mmol, 65%); Ή-NMR (250 MHz, DMS0-d 6 ) d = 9.36 (s, lH), 9.21 (t, J= 5.6 Hz, lH), 7.97 - 7.86 (m, 2H), 7.66 - 745 (m, 5H), 6.54 (s, 2H), 5.16 (q, J=7- o Hz, lH), 4.61 (d, J= 5.3 Hz, 2H), 1.38 (d, J=7 o Hz, 3H) ppm; «C-NMR (75 MHz, DMSO-d 6 ) d = 165.7, 162.2, 161.5, 158.9, 134.2, 133.6, 131.3 (d), 130.1,

128.9, 127.4, 122.7, 119-6, 119.4, 113-5 (q), H3-2 (q), 90.7, 81.1, 45.8, 18.6 ppm; purity (HPLC- MS): 97% t Ri 9.91 min (method: gradient of 10% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 2O H I7 F 4 N 3 0 3 + H + [M + H + ]: 424.12788; found: 424.12847.

[210] A/-(3,3-Diphenylpropyl)-3-(3-(i-hydroxyureido)but-i-yn-i-yl) benzamide 7t: procedure C; 100 mg (0.23 mmol) 151, 42 mg (0.26 mmol, 1.5 eq) 8a, 3 mg (0.01 mmol, 0.05 eq) Pd(ACN) 2 Cl 2 , 5 mg (0.02 mmol, 0.11 eq) Cul, 4 mg (0.02 mmol, 0.07 eq) PPh 3 , 0.1 mL (0.27 mmol, 1.2 eq) DIPA, 10 mL ethyl acetate, 5 mL THF, 41 h, purification with column chromatography (hexane: ethyl acetate 1:4), further purification with preparative HPLC (30% ACN for 3 min, linear gradient from 30% ACN to 80% within 8 min); off-white solid (48 mg, 0.11 mmol, 48%); Ή-NMR (300 MHz, acetone-de) d = 8.89 (s, lH), 8.11 - 7.76 (m, 3H), 7.75 - 6.79 (m, 12H), 6.25 - 6.10 (m, 2H), 5.28 (q, J=7.o Hz, lH), 4.12 (t, =7-8, 7.8 Hz, lH), 3.42 - 3.32 (m, 2H), 2.48 - 2.37 (m, 2H), 1.45 (d, J=7.o Hz, 3H) ppm; «C-NMR (75 MHz, acetone-d 6 ) d = 166.6, 162.3, 145.9, 136.2, 134.7, I3i-i, 129-3, 128.7, 127-9, 127 0 , 124.2, 90.6, 82.4, 49.8, 47.3, 39-5, 35-9, 18.7 ppm; purity (HPLC-MS): 97% tiu 10.57 min (method: gradient of 10% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 27 H 27 N 3 0 3 + H + [M + H + ]: 442.21252; found: 442.21247.

[211] -(3-(4-Fluorophenyl)-3-phenylpropyl)-3-(3-(i-hydroxyureido)b ut-i-yn-i-yl)benzamide 711: procedure C; 114 mg (0.25 mmol) 15J, 46 mg (0.36 mmol, 1.5 eq) 8a, 3 mg (0.01 mmol, 0.05 eq) Pd(ACN) 2 Cl 2 , 5 mg (0.03 mmol, 0.12 eq) Cul, 4 mg (0.02 mmol, 0.07 eq) PPh 3 , 0.1 mL (0.30 mmol, 1.2 eq) DIPA, 10 mL ethyl acetate, 38 h, purification with column chromatography (ethyl acetate 100%), further purification with preparative HPLC (linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min); white solid (35 mg, 0.08 mmol, 34%); Ή-NMR (300 MHz, acetone-d 6 ) d = 8.68 (s, lH), 7.87 - 7.80 (m, 3H), 7.53 - 7.01 (m, 11H), 6.12 (s, 2H), 5.27 (q, J=7. o Hz, lH), 4.15 (t, J=7.8 Hz, lH), 3.41 - 3.33 (m, 2H), 2.46 - 2.37 (m, 2H), 1.45 (d, J= 6.9 Hz, 3H) ppm; «C-NMR (75 MHz, acetone-de) d = i66.6, 163.8, 162.1, 160.6, 145.7 (d), 142.0, 136.3, 134.7, 131-0, 130.5, 130.3, 129-4, 128.6, 127.9, 127-1, 124-2, 116.0, 115.7, 90.6, 82.4,

48.9, 47.4, 39-4, 36.0, 18.6 ppm; purity (HPLC-MS): 99% tiu 10.62 min (method: gradient of 10% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 27 H 26 FN 3 0 3 + H + [M + H + ]: 460.20310; found: 460.20133.

[212] 3-(3-(i-Hydroxyureido)but-i-yn-i-yl)- -(3-phenyl-3-(4-(trifluoromethyl)phenyl)- propyObenzamide 7v: procedure C; 155 mg (0.30 mmol) 15m, 43 mg (0.34 mmol) 8a, 2 mg (0.006 mmol) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol) Cul, 3 mg (0.01 mmol) PPh 3 , 0.1 mL (0.37 mmol, 1.2 eq) DIPA, 8 mL ethyl acetate, 38 h, purification with column chromatography

(hexane:acetone 2:3), further purification with preparative HPLC (linear gradient from 10% ACN to 90% within 10 min, 90% ACN for 6 min); white solid (64 mg, 0.13 mmol, 41%); Ή-NMR (300 MHz, acetone-d 6 ) d = 8.72 (s, lH), 7.92 - 7.80 (m, 3H), 7.66 - 7.19 (m, 11H), 6.14 (s, 2H), 5.28 (q, J=7- o Hz, lH), 4.27 (t, J=7.8 Hz, lH), 3.43 - 3.35 (m, 2H), 2.53 - 2.44 (m, 2H), 1.45 (d, J=7.o Hz, 3H) ppm; «C-NMR (75 MHz, acetone-de) d = i66.6, 162.1, 150.6, 144.9, 136.2, 134.7, 131.0, 129.5, 129-4, 129-3, 128.7, 127-8, 127.4, 126.2 (q), 124.2, 90.6, 82.3, 47.3, 39.3, 35.6, 18.6 ppm; purity (HPLC-MS): 98% t Ri 11 .40 min (method: gradient of 10% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 28 H 26 F 3 N 3 O 3 + H + [M + H + ]: 510.19990; found: 510.119834.

[213] 3-(3-(i-Hydroxyureido)but-i-yn-i-yl)- -(3-phenyl-3-(4-(trifluoromethoxy)phenyl)- propyObenzamide 7w: procedure C; 119 mg (0.23 mmol) 15h, 32 mg (0.25 mmol) 8a, 1 mg (0.004 mmol) Pd(ACN) 2 Cl 2 , 4 mg (0.02 mmol, 0.09 eq) Cul, 4 mg (0.01 mmol, 0.06 eq) PPh 3 , 0.1 mL (0.27 mmol, 1.2 eq) DIPA, 8 mL ethyl acetate, 38 h, purification with column chromatography (hexane: ethyl acetate 1:4), further purification with preparative HPLC (linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min); white solid (30 mg, 0.06 mmol, 25%); Ή-NMR (500 MHz, acetone-de) d = 8.72 (s,iH), 8.08 - 7.78 (m, 3H), 7.54 - 7.17 (m, 11H), 6.13 (s, 2H), 5-27 (q, J=7- o Hz, lH), 4.21 (t, J=7-8 Hz, lH), 3.40 - 3.35 (m, 2H),

2.48 - 2.42 (m, 2H), 1.45 (d, 7=7. o Hz, 3H) ppm; «C-NMR (125 MHz, acetone-d 6 ) d = i66.6, 162.1, 148.3 (q), 145.4, 145-4, 136.3, 134-8, 131.1, 130.4, 129 6, 129.4, 128.8, 127.9, 124-3, 122.0, 90.7, 82.4, 49.1, 47.4, 39.4, 35.9, 18.7 ppm; purity (HPLC-MS): 97% t R : 8.41 min (method: gradient of 20% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 28 H 26 F 3 N 3 O 4 + Na + [M + Na + ]: 548.17676; found: 548.17552.

[214] -(3-(4-Chlorophenyl)-3-phenylpropyl)-3-(3-(i-hydroxyureido)b ut-i-yn-i-yl)benzamide 7w: procedure C; 108 mg (0.23 mmol) 150, 32 mg (0.25 mmol) 8a, 1 mg (0.01 mmol) Pd(ACN) 2 Cl 2 , 4 mg (0.02 mmol, 0.09 eq) Cul, 4 mg (0.01 mmol, 0.06 eq) PPh 3 , 0.1 mL (0.27 mmol, 1.2 eq) DIPA, 8 mL ethyl acetate, 38 h, purification with column chromatography (hexane: ethyl acetate 1:4), further purification with preparative HPLC (linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min); white solid (19 mg, 0.04 mmol, 18%); Ή- NMR (500 MHz, acetone-d 6 ) d = 8.72 (s, lH), 8.09 - 7.73 (m, 3H), 7.52 (td, 7=1.3, 7.6 Hz, lH),

7.48 - 7.2.2. (m, 9H), 7.21 - 7.16 (m, lH), 6.14 (s, 2H), 5.27 (q, J=7. o Hz, lH), 4.16 (t, J=7.8 Hz, lH), 3-37 (q, 7=6.7 Hz, 2H), 2.42 (q, 7=7.4 Hz, 2H), 1.45 (d, J=7- o Hz, 3H) ppm; «C-NMR (125 MHz, acetone-d 6 ) d = 166.5, I62.O, 145-4, 144-9, 136.2, 134.7, 132.3, 131.0, 130.4, 129-4, 129-3, 129.2, 128.6, 127.8, 127.2, 124.2, 90.6, 82.3, 49.0, 47.3, 39.3, 35.8, 18.6 ppm; purity (HPLC-MS): 98% t R : 11.21 min (method: gradient of 10% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 27 H 26 C1N 3 0 3 + H + [M + H + ]: 476.17355; found: 476.17231.

[215] (R)-3-(3-(i-Hydroxyureido)but-i-yn-i-yl)- -(3-(4-(methylsulfonamido)phenyl)-3- phenylpropyObenzamide 7y: procedure C; 108 mg (0.20 mmol) 15G, 29 mg (0.22 mmol) 8d,

1 mg (0.004 mmol) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol) Cul, 2 mg (0.01 mmol) PPh 3 , 0.3 mL (2.43 mmol, 12 eq) DIPA, 8 mL ethyl acetate, 40 h, purification with column chromatography (ethyl acetate 100%), further purification with preparative HPLC (5% ACN for 2 min, linear gradient from 5% ACN to 90% within 14 min, 90% ACN for 6 min); white solid (40 mg, 0.08 mmol, 37%); Ή-NMR (300 MHz, acetone-d 6 ) d = 8.62 (d, J= 55.1 Hz, lH), 7.84 - 7.80 (m, 3H), 7.53 - 7.17 (m, 12H), 6.16 (bs, 2H), 5.28 (q, J=7- o Hz, lH), 4.12 (t, J=7-8 Hz, lH), 3.43 - 3.35 (m, 2H), 2-94 (s, 3H), 2.47 - 2.38 (m, 2H), 1.46 (d, J=7.o Hz, 3H) ppm; «C-NMR (125 MHz, acetone-d 6 ) d = I66·5, 102.2 (d), 145.9 (d), 142.0, 136.3, 134.7, 131-0 (d), 129.5, 128.6, 127.0, 124.2, 121.5 (d), 90.6, 82.3, 49.2 (d), 47.3, 39.5, 39.3, 35.9, 18.6 ppm; purity (HPLC-MS): 96% t R : 12.59 min (method: 5% ACN for 2 min, linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 28 H 30 N 4 O 5 S+ H + [M + H + ]: 535.20097; found: 535-19989·

[216] (R)-3-(3-(i-Hydroxyureido)but-i-yn-i-yl)- -(3-phenyl-3-(4-sulfamoylphenyl)- propyl)benzamide 7z: procedure C; 73 mg (140 nmol) 15s, 20 mg (154 nmol) 8d, 1 mg (3 nmol) Pd(ACN) 2 Cl 2 , 1 mg (3 pmol, 0.02 eq) Cul, 2 mg (5 pmol) PPh 3 , 0.1 mL (1680 pmol, 12 eq) DIPA, 5 mL ethyl acetate, 36 h, purification with column chromatography (hexane:acetone i:2-i:4), further purification with preparative HPLC (5% ACN for 2 min, linear gradient from 5% ACN to 90% within 14 min, 90% ACN for 6 min); white solid (35 mg, 67 pmol, 48%); Ή-NMR (300 MHz, acetone-d 6 ) d = 8.70 (s, lH), 7.99 - 7.77 (m, 5H), 7.58 - 7.15 (m, 9H), 6.49 (s, 2H), 6.15 (s, 2H), 5-27 (q, J=7- o Hz, lH), 4.25 (t, J=7- 8 Hz, lH), 3.43 - 3.34 (m, 2H), 2.52 - 2.43 (m, 2H), 1.45 (d, J=7- o Hz, 3H) ppm; «C-NMR (125 MHz, acetone-de) d = I6I.2, 149.3, 144-1, 142.1, 135-3, 133-8, 130.1, 128.6, 128.8, 128.3, 1278, 126.9, 126.4, 126.3, 123-3, 89.7, 81.4, 48.5, 46.4, 38.4, 34.6, 17.7 ppm; purity (HPLC-MS): 97% t R : 12.14 min (method: 5% ACN for 2 min, linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 27 H 28 N 4 0 5 S+ H + [M + H + ]: 521.18532; found: 521.18435.

[217] (R)- -(3-(3,4-Dichlorophenyl)-3-phenylpropyl)-3-(3-(i-hydroxyurei do)but-i-yn-i-yl)- benzamide 7aa: procedure C; 0.15 g (0.29 mmol) I5p, 41 mg (0.32 mmol) 8d, 3 mg (0.01 mmol, 0.04 eq) Pd(ACN) 2 Cl 2 , 2 mg (0.01 mmol) Cul, 6 mg (0.02 mmol, 0.08 eq) PPh 3 , 0.1 mL (0.35 mmol, 1.2 eq) DIPA, 8 mL ethyl acetate, 38 h, purification with column chromatography (hexane:acetone i:i-2:3), further purification with preparative HPLC (5% ACN for 2 min, linear gradient from 5% ACN to 90% within 14 min, 90% ACN for 6 min); white solid (80 mg, 0.16 mmol, 53%); Ή-NMR (300 MHz, acetone-de) d = 8.75 (s, lH), 7.92 - 7.79 (m, 3H), 7.56 - 7.19 (m, 10H), 6.16 (s, 2H), 5.28 (q, J=7- o Hz, lH), 4.19 (t, J=7-8 Hz, lH), 3.43 - 3.34 (m, 2H), 2.49 - 2.39 (m, 2H), 1.45 (d, J=7- o Hz, 3H) ppm; «C-NMR (125 MHz, acetone-d 6 ) d = 165.7, I6I.2, 161.2,146.2, 143.9, 135-3, 133-9, 133-7, 131-7, 130-5, 130.1, 129.9, 129-4, 128.7, 128.5, 128.0, 127.8, 127.0, 126.6, 123.3, 89.8, 81.5, 47.9, 46.4, 38.2, 34.6, 17.7 ppm; purity (HPLC-MS): 96% t R : 14.84 min (method: 5% ACN for 2 min, linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 27 H 25 C1 2 N 3 0 3 + H + [M + H + ]: 510.13457; found: 510.13361.

[218] (i?)-A/-(3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)pro pyl)-3-(3-(i-hydroxy- ureido)but-i-yn-i-yl)benzamide 7ab: procedure C; 64 mg (121 nmol) I5t, 17 mg (133 nmol) 8d, 1 mg (2 pmol) Pd(ACN) 2 Cl 2 , 1 mg (5 pmol) Cul, 1 mg (4 pmol) PPh 3 , 0.2 mL (1450 pmol, 12 eq) DIPA, 8 mL ethyl acetate, 65 h, purification with column chromatography (hexane: acetone 2:3), further purification with preparative HPLC (5% ACN for 2 min, linear gradient from 5% ACN to 90% within 14 min, 90% ACN for 6 min); white solid (35 mg, 66 pmol, 55%); Ή-NMR (300 MHz, acetone-d 6 ) d = 8.65 (s, lH), 7.91 - 7.79 (m, 3H), 7.67 - 7.50 (m, 5H), 7.46 - 7.39 (m, 3H), 7.14 - 7.03 (m, 2H), 6.11 (bs, 2H), 5.27 (q, J= . o Hz, lH), 4.31 (t, J=7-8 Hz, lH), 3.42 - 3.35 (m, 2H), 2.51 - 2.43 (m, 2H), 1.45 (d, J=7.o Hz, 3H) ppm; «C-NMR (75 MHz, acetone-d 6 ) d = 166.5, 163.3, 162.0, i6ΐ·4, 150.4, 141.0 (d), 136.2, 134.7, ΐ3ΐ·0, 130.6 (d), 129.4, 127-8, 126.3 (q), 124 2 , 116.2, 116.0, 90.7, 82.3, 48.5, 47.3, 39.1, 35.7, 18.6 ppm; purity (HPLC-MS): 95% t R : 15.03 min (method: 5% ACN for 2 min, linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 28 H 25 F 4 N 3 0 3 + H + [M + H + ]: 528.19048; found: 528.18925.

[219] (R)- -(3-(4-Fluorophenyl)-3-(4-(trifluoromethoxy)phenyl)propyl)-3 -(3-(i-hydroxy- ureido)but-i-yn-i-yl)benzamide 7ac: procedure C; 150 mg (276 pmol) 1511, 39 mg (304 pmol) 8d, 1 mg (5 pmol) Pd(ACN) 2 Cl 2 , 1 mg (5 pmol, 0.02 eq) Cul, 3 mg (11 pmol) PPh 3 , 0.1 mL (0.33 mmol, 1.2 eq) DIPA, 8 mL ethyl acetate, 38 h, purification with column chromatography (hexane: ethyl acetate 1:4), further purification with preparative HPLC (5% ACN for 2 min, linear gradient from 5% ACN to 90% within 14 min, 90% ACN for 6 min); white solid (25 mg, 46 pmol, 17%); Ή-NMR (300 MHz, acetone-de) d = 8.67 (bs, lH), 8.03 - 7.75 (m, 3H), 7.55 - 7.35 (m, 6H), 7.31 - 7.18 (m, 2H), 7.10 - 7.03 (m, 2H), 6.12 (bs, 2H), 5.27 (q, J= . o Hz, lH), 4.24 (t, J=7- 8 Hz, lH), 3.41 - 3.33 (m, 2H), 2.48 - 2.39 (m, 2H), 1.45 (d, J=7- o Hz, 3H) ppm; «C-NMR (125 MHz, acetone-d 6 ) d = i66.6, 163.3, 162.0, 161.3, 148.3 (q), 145.2, 141.3 (d), 136.2, 134.7, 130.5, 130.4, 130.3, 129.4, 127-8, 121.9, 115-9, 90.6, 82.3, 48.1, 47.3, 39.2, 36.0, 18.6 ppm; purity (HPLC-MS): 97% t R : 15.23 min (method: 5% ACN for 2 min, linear gradient from 5% ACN to 90% within in 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 28 H 25 F 4 N 3 0 4 + H + [M + H + ]: 544.18540; found: 544.18424.

[220] (R)- -(3,3-Bis(4-fluorophenyl)propyl)-3-(3-(i-hydroxyureido)but-i -yn-i-yl)benzamide 7ad: procedure C; 177 mg (371 pmol) 15V, 52 mg (408 pmol) 8d, 4 mg (12 pmol, 0.04 eq) Pd(ACN) 2 Cl 2 , 3 mg (15 pmol) Cul, 8 mg (30 pmol, 0.08 eq) PPh 3 , 0.3 mL (0.33 mmol, 5 eq) DIPA, 8 mL ethyl acetate, 32 h, purification with column chromatography (hexane: acetone 1:1), further purification with preparative HPLC (5% ACN for 2 min, linear gradient from 5% ACN to 90% within 14 min, 90% ACN for 6 min); white solid (60 mg, 126 pmol, 43%); Ή-NMR (400 MHz, acetone-d 6 ) d = 8.71 (bs, lH), 7.93 - 7.80 (m, 3H), 7.52 (td, J= 1.4, 7.7 Hz, lH), 7.44 - 7.36 (m, 5H), 7.12 - 7.01 (m, 4H), 6.13 (bs, 2H), 5.29 (q, J=7-0 Hz, lH), 4.19 (t, « 7=7-8 Hz, lH), 3-39 - 3-34 (m, 2H), 2.45 - 2.38 (m, 2H), 1.45 (d, « 7=6-3 Hz, 3H) ppm; ^-NMR (100 MHz, acetone-d 6 ) d = i66.6, 163.5, 162.1, 161.1, 141.8 (d), 136.3, 134.7, 131 0 , 130.4, 130.3, 129.4, 127.8, 124.3, 116.0, 115.8, 90.6, 82.4, 48.0, 47.4, 39.3, 36.2, 18.6 ppm; purity (HPLC-MS): 96% t R : 14.28 min (method: 5% ACN for 2 min, linear gradient from 5% ACN to 90% within in 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 27 H 25 F 2 N 3 0 3 + Na + [M + Na + ]: 500.17457; found: 500.17562.

[221] (i?)- -(2-(Diphenylamino)ethyl)-3-(3-(i-hydroxyureido)but-i-yn-i-y l)benzamide 7ae: procedure C; 90 mg (203 pmol) 15X, 29 mg (223 pmol) 8d, 2 mg (8 pmol, 0.04 eq) Pd(ACN) 2 Cl 2 , 2 mg (8 pmol) Cul, 4 mg (16 pmol, 0.08 eq) PPh 3 , 0.1 mL (1015 pmol, 5 eq) DIPA, 8 mL ethyl acetate, 32 h, purification with column chromatography (hexane:acetone 1:1), further purification with preparative HPLC (5% ACN for 2 min, linear gradient from 5% ACN to 90% within 10 min, 90% ACN for 6 min); white solid (38 mg, 86 pmol, 42%); Ή-NMR (500 MHz, acetone-d 6 ) d = 8.71 (s, lH), 8.03 (t, J= 5.8 Hz, lH), 7.85 - 7.80 (m, 2H), 7.53 (td, J= 1.4, 7.6 Hz, lH), 7.42 (t, J=7- 9 Hz, lH), 7.31 - 7.24 (m, 4H), 7.12 - 7.06 (m, 4H), 6.97 - 6.91 (m, 2H), 6.13 (s, 2H), 5.28 (q, J=7. o Hz, lH), 4.02 - 3.97 (m, 2H), 3.71 - 3 64 (m, 2H), 1.46 (d, 7=7.2 Hz, 3H) ppm; «C-NMR (100 MHz, acetone-de) d = 167.0, I62.I, 148.8, 136.0, 134-8, 131.0, 130.2, 129-4, 127-8, 124-3, 122.2, 121.6, 90-7, 82.3, 51.7, 47.3, 38.4, i8.6 ppm; purity (HPLC-MS): 98% t R : 14.52 min (method: 5% ACN for 2 min, linear gradient from 5% ACN to 90% within in 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 26 H 26 N 4 0 3 + H + [M + H + ]: 443.20777; found: 443.20728.

[222] -((R)-3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)propyl )-3-((R)-3-(i-hydroxy- ureido)but-i-yn-i-yl)benzamide 7af: procedure C; 55 mg (104 pmol) IOU, 15 mg (114 pmol) 8d, 1 mg (4 pmol, 0.04 eq) Pd(ACN) 2 Cl 2 , 1 mg (4 pmol) Cul, 2 mg (8 pmol) PPh 3 , 0.3 mL (0.52 mmol, 5 eq) DIPA, 8 mL ethyl acetate, 32 h, purification with column chromatography (hexane:acetone 1:1), further purification with preparative HPLC (5% ACN for 2 min, linear gradient from 5% ACN to 90% within 14 min, 90% ACN for 6 min); white solid (35 mg, 66 pmol, 64%); Ή-NMR (400 MHz, acetone-d 6 ) d = 8.64 (bs, lH), 7.93 - 7.80 (m, 3H), 7.66 - 7.58 (m, 4H), 7-52 (td, 7=1.3, 7-8 Hz, lH), 7.46 - 738 (m, 3H), 7.11 - 7.03 (m, 2H), 6.09 (bs, 2H), 5 2 7 (q, « 7=7-0 Hz, lH), 4.31 (t, « 7=7-9 Hz, lH), 3.41 - 3.35 (m, 2H), 2.51 - 2.43 (m, 2H), 1.45 (d, « 7=7- o Hz, 3H) ppm; «C-NMR (100 MHz, acetone-de) d = i66.6, 162.0, 161.5, 150.5, 141.0, 134.8, 131.0, 130.6, 130.5, 129.4, 127-8, 126.3 (q), 124-2, 116.2, 116.0, 90.7, 82.3, 48.6, 47.4, 39.2, 35.8, 18.6 ppm; purity (HPLC-MS): 99% t R : 15.05 min (method: 5% ACN for 2 min, linear gradient from 5% ACN to 90% within in 10 min, 90% ACN for 6 min); HRMS (MALDI): m/z calculated C 28 H 25 F 4 N 3 0 3 + H + [M + H + ]: 528.19048; found: 528.18885.

[223] 3-(4-(Trifluoromethyl)phenyl)acrylamide 23a: 2.14 g (9.92 mmol) 3-(4-

(trifluoromethyl)phenyl)acrylic acid (22a) was solved in 15 mL THF and cooled to -15 °C. 2.3 mL (1.40 mmol, 1.7 eq) ethyl chloroformate, diluted in 4 mL THF, were added slowly. The mixture stirred for 1 h at - 15 °C. Additionally, 49 mL (406.27 mmol, 32%, 41 eq) NH 4 OH were added and the reaction stirred for 18 h at -15 °C - rt. The suspension was extracted with DCM (3x) and the combined organic phase was washed with saturated aqueous NaHC0 3 solution (3x) and brine (lx). After drying over MgS0 4 and filtration the solvent was evaporated under reduced pressure. The solid was recrystallized with ethyl acetate yield 1.43 g (6.66 mmol, 67 %); Ή- NMR (300 MHz, acetone-d 6 ) d = 7.92 - 7.70 (m, 4H), 7.60 (d, J=15.8 Hz, lH), 7.08 (s, lH), 6.86 (d, J= 16.7 Hz, lH), 6.59 (s, lH) ppm; MS (ESI) m/z: 214.85 [M + H + ].

[224] General procedure for Heck coupling (compounds 24a-j, Procedure D)

[225] 1 eq cinnamamide derivative 23a-c was suspended in triethylamine. For solubility reasons an organic solvent maybe added. 1.5 eq iodoaryl derivative and 0.05 eq Pd(0Ac) 2 were added and the reaction mixture was heated to 100 °C for 16-48 h. All solvent components were evaporated, and the brown oil was pre-adsorbed on silica gel. Column chromatography yielded a white solid.

[226] 3-(4-Fluorophenyl)-3-phenylacrylamide 24a: procedure D; 0.30 g (2.04 mmol) cinnamamide 23c, 0.68 g (3.06 mmol) i-fluoro-4-iodobenzene, 0.02 g (0.10 mmol) Pd(0Ac) 2 , 5 mL NEt 3 ; eluent of column chromatography hexane:acetone 1:1; white solid (0.32 g, 1.34 mmol, 66%); Ή-NMR (300 MHz, acetone-de) d = 7.48 - 7.17 (m, 7H), 7.15 - 7.09 (m, 2H), 6.91 - 6.17 (m, 3H) ppm; MS (ESI) m/z: 241.95 [M + H + ].

[227] 3-Phenyl-3-(4-(trifluoromethyl)phenyl)acrylamide 24b: procedure D; 0.30 g (2.04 mmol) cinnamamide 23c, 0.85 g (3.06 mmol) i-iodo-4-(trifluoromethyl)benzene, 0.02 g (0.10 mmol) Pd(0Ac) 2 , 5 mL NEt 3 , eluent of column chromatography hexane:acetone 1:1; white solid (0.34 g, 1.17 mmol, 57%); Ή-NMR (250 MHz, acetone-d 6 ) d = 7.75 (d, J= 8.3 Hz, 2H), 7.57 - 7.10 (m, 7H), 6.71 - 6.48 (m, 2H), 6.37 - 6.32 (m, lH) ppm; MS (ESI) m/z: 291.90 [M + H + ].

[228] 3-Phenyl-3-(4-(trifluoromethoxy)phenyl)acrylamide 24c: procedure D; 70 mg (0.48 mmol) cinnamamide 23c, 0.1 mL (0.71 mmol) i-iodo-4-(trifluoromethoxy)benzene, 5 mg (24 pmol) Pd(0Ac) 2 , 1 mL NEt 3 , eluent of column chromatography hexane:acetone 4:1; white solid (0.08 g, 0.25 mmol, 53%); Ή-NMR (250 MHz, DMSO-de) d = 7.59 - 7.08 (m, 10H), 7.00 (bs, lH), 6.45 - 6.44 (m, lH) ppm; MS (ESI) m/z: 307.85 [M + H + ].

[229] 3-(4-Chlorophenyl)-3-phenylacrylamide 241I: procedure D; 0.20 g (1.36 mmol) cinnamamide 23c, 0.50 g (2.08 mmol) i-chloro-4-iodobenzene, 0.03 g (0.17 mmol) Pd(0Ac) 2 , 5 mL NEt 3 ; eluent of column chromatography hexane:acetone 2:1; white solid (0.24 g, 0.92 mmol, 68%); Ή-NMR (300 MHz, acetone-de) d = 7.40 - 7.23 (m, 9H), 6.49 - 6.26 (m, 3H) ppm; MS (ESI) m/z: 257.90 [M + H + ]. [230] 3-(4-Aminophenyl)-3-phenylacrylamide 24e: procedure D; 1.30 g (8.80 mmol) cinnamamide 23c, 2.95 g (13.20 mmol) 4-iodoaniline, 0.10 g (0.44 mmol) Pd(0Ac) 2 , 15 mL NEt 3 ; eluent of column chromatography hexane:acetone 1:3; brown solid (0.98 g, 4.11 mmol, 47%); Ή-NMR (300 MHz, DMS0-d 6 ) <5 = 7.34 - 7.29 (m, 3H), 7.11 - 7.08 (m, 2H), 7.00 (bs, lH), 6.89 - 6.83 (m, 2H), 6.67 (bs, lH), 6.51 - 6.47 (m, 2H), 6.22 (s, 0.8 H), 6.06 (s, 0.2H), 5.38 (bs, 2H) ppm; MS (ESI) m/z: 239.13 [M + H + ].

[231] 3-Phenyl-3-(4-sulfamoylphenyl)acrylamide 24f: procedure D; 0.35 g (2.36 mmol) cinnamamide 23c, 1.00 g (3.54 mmol) 4-iodobenzenesulfonamide, 0.03 g (0.12 mmol) Pd(0Ac) 2 , 5 mL NEt 3 ; eluent of column chromatography hexane:acetone 2:3; white solid (0.35 g, 1.16 mmol, 49%); Ή-NMR (300 MHz, acetone-de) d = 7.75 - 7.71 (m, 2H), 7.34 - 7.23 (m, 5H), 7.15 - 7.09 (m, 2H), 6.83 - 6.30 (m, 5H) ppm; MS (ESI) m/z : 302.85 [M + H + ].

[232] 3-(3,4-Dichlorophenyl)-3-phenylacrylamide 24g: procedure D; 1.50 g (10.20 mmol) cinnamamide 23c, 4.17 g (15.30 mmol) i,2-dichloro-4-iodobenzene, 0.12 g (0.51 mmol) Pd(0Ac) 2 , 20 mL NEt 3 ; eluent of column chromatography hexane:acetone i:i-2:3; white solid (1.32 g, 4.50 mmol, 44%); Ή-NMR (300 MHz, acetone-de) d = .58 - 7.54 (m, lH), 7.46 - 7.36 (m, 4H), 7.31 - 7.21 (m, 3H), 6.56 - 6.54 (m, 2H), 6.31 (bs, lH) ppm; MS (ESI) m/z: 293.45 [M + 2H + ]

[233] 3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)acrylamide 24I1: procedure D, 0.81 g (3.77 mmol) 3-(4-(trifluoromethyl)phenyl)acrylamide 23a; 1.23 g (5.65 mmol) i-fluoro-4- iodobenzene, 0.04 g (0.19 mmol) Pd(0Ac) 2 , 35 mL NEt 3 , 12 mL THF, eluent of column chromatography hexane:acetone 1:1-2:35 white solid (0.79 g, 2.56 mmol, 86%); Ή-NMR (250 MHz, acetone-d 6 ) d = 7.73 - 7.68 (m, 2H), 7.52 - 7.27 (m, 4H), 7.18 - 7.11 (m, 2H), 6.87 - 6.79 (m, lH), 6.58 - 6.57 (m, lH), 6.31 (bs, lH) ppm; MS (ESI) m/z: 309.65 [M + H + ].

[234] 3-(4-Fluorophenyl)-3-(4-(trifluoromethoxy)phenyl)acrylamide 241: procedure D, 0.83 g

(5.03 mmol) 3-(4-fluorophenyl)acrylamide 23b, 1.2 mL (7.54 mmol) i-iodo-4-

(trifluoromethoxy)benzene, 0.92 g (4.02 mmol, 0.08 eq) Pd(0Ac) 2 , 20 mL NEt 3 , 9 mL THF, eluent of column chromatography hexane: acetone 2:1-2:35 white solid (1.30 g, 4.01 mmol, 79%); Ή-NMR (250 MHz, acetone-d 6 ) d = 7.32 - 7.25 (m, 2H), 7.20 - 7.12 (m, 4H), 7.03 - 6.97 (m, 2H), 6.56 (s, lH), 6.37 (s, lH), 6.20 (s, lH) ppm; MS (ESI) m/z: 325.75 [M + H + ].

[235] 3,3-Bis(4-fluorophenyl)acrylamide 24j: procedure D, 0.70 g (4.24 mmol) 3-(4- fluoropheny acrylamide 23b, 0.7 mL (6.36 mmol) i-fluoro-4-iodobenzene, 0.49 g (0.21 mmol) Pd(0Ac) 2 , 20 mL NEt 3 , 9 mL THF, eluent of column chromatography hexane:acetone 2:1-2:35 white solid (0.52 g, 2.00 mmol, 47%); Ή-NMR (250 MHz, acetone-d 6 ) d = 7.37 - 7.2.2. (m, 4H), 7.18 - 7.08 (m, 4H), 6.62 (bs, lH), 6.44 (s, lH), 6.28 (bs, lH) ppm; MS (ESI) m/z: 260.15 [M + H + ].

[236] General procedure for hydrogenation of 24a-j (Procedure E) [237] The acrylamide derivative 4a-j was dissolved in methanol/in ethanol and 10 wt% Pd/C or Pd(0H) 2 were added. The suspension was set under vacuum and flushed with hydrogen (either in an autoclave or with a balloon). The reaction stirred for 16-48 h at room temperature. After filtration over Celite the solvent was evaporated, and the yielded oil was used without further purification.

[238] 3-(4-Fluorophenyl)-3-phenylpropanamide 25a: procedure E; 0.31 g (1.28 mmol) 24a, 0.03 g Pd/C, 20 mL MeOH, 40 h, 3.5 bar hydrogen, transparent oil (0.31 g, 1.27 mmol, 99%); Ή-NMR (300 MHz, acetone-d 6 ) d = 7.47 - 7.13 (m, 7H), 7.05 - 6.98 (m, 2H), 6.76 (s, lH), 6.11 (s, lH), 4.60 (t, J=7- 8 Hz, lH), 2.94 (d, J=7.8 Hz, 2H) ppm; MS (ESI) m/z: 243.95 [M + H + ].

[239] 3-Phenyl-3-(4-(trifluoromethyl)phenyl)propenamide 25b: procedure E; 0.32 g (1.11 mmol) 24b, 0.03 g Pd/C, 20 mL MeOH, 40 h, 3.1 bar hydrogen, transparent oil (0.32 g, 1.09 mmol, 98%); Ή-NMR (300 MHz, acetone-de) d = 7.67 - 7.49 (m, 4H), 7.37 - 7.12 (m, 5H), 6.81 (s, lH), 6.14 (s, lH), 4.70 (t, J=7- 8 Hz, lH), 3.01 (dd, J= 4.5, 7.8 Hz, 2H) ppm; MS (ESI) m/z: 293.28 [M + H + ].

[240] 3-Phenyl-3-(4-(trifluoromethoxy)phenyl)propanamide 25c: procedure E; 0.26 g (0.84 mmol) 24c, 0.03 g Pd/C, 20 mL MeOH, 40 h, 3.2 bar hydrogen, transparent oil (0.24 g, 0.77 mmol, 92%); Ή-NMR (300 MHz, acetone-de) d = 7.45 - 7.19 (m, 9H), 6.79 (bs, lH), 6.11 (bs, lH), 4.65 (t, J=7- 8 Hz, lH), 2.97 (dd, J= 1.7, 7.8 Hz, 2H) ppm; MS (ESI) m/z: 309.90 [M + H + ].

[241] 3-(4-Chlorophenyl)-3-phenylpropanamide 251I: procedure E; 0.20 g (0.78 mmol) 241I, 0.01 g Pd(0H) 2 on charcoal, 15 mL EtOH, 18 h, hydrogen atmosphere via a balloon, transparent oil (0.19 g, 0.71 mmol, 92%); Ή-NMR (300 MHz, CDC1 3 ) d = 7.65 - 7.10 (m, 11H), 4.60 (t, J=7- 9 Hz, lH), 3.13 - 2.78 (m, 2H) ppm; MS (ESI) m/z: 259.65 [M + H + ].

[242] 3-(4-Aminophenyl)-3-phenylpropanamide 25e: procedure E; 0.48 g (2.02 mmol) 24e, 0.05 g Pd/C, 50 mL MeOH, 72 h, 3.8 bar hydrogen, transparent oil (0.48 g, 1.99 mmol, 99%); Ή-NMR (300 MHz, acetone-d 6 ) d = 7.33 - 6.99 (m, 9H), 6.63 - 6.58 (m, 3H), 6.06 (bs, lH), 4.51 - 4.43 (m, lH), 3.00 - 2.96 (m, 2H) ppm; MS (ESI) m/z: 240.95 [M + H + ].

[243] 3-Phenyl-3-(4-sulfamoylphenyl)propenamide 25f: procedure E; 0.35 g (1.12 mmol) 24f; 0.04 g Pd/C, 20 mL MeOH, 43 h hydrogen atmosphere via a balloon, transparent oil (0.26 g, 0.87 mmol, 75%); Ή-NMR (250 MHz, acetone-de) d = 7.83 - 7.74 (m, 2H), 7.52 - 7.46 (m, 2H), 7.32 - 7.28 (m, 5H), 6.83 (bs, lH), 6.49 (s, 2H), 6.16 (bs, lH), 4.68 (t, J=7.8 Hz, lH), 3.01 (dd, J=5- 3, 7-9 Hz, 2H) ppm; MS (ESI) m/z: 346.10 [M + ACN + H + ].

[244] 3-(3,4-Dichlorophenyl)-3-phenylpropanamide 25g: procedure E; 0.10 g (0.39 mmol) 24g, 0.01 g Pd(0H) 2 on charcoal, 6 mL EtOH, 18 h, 3 bar hydrogen, transparent oil (0.10 g, 0.33 mmol, 85%); Ή-NMR (250 MHz, CDC1 3 ) d = 7.55 - 7.40 (m, 2H), 7.39 - 7.12 (m, 6H), 6.79 (bs, lH), 6.12 (bs, lH), 4.61 (t, J=7.5 Hz, lH), 3.11 - 2.85 (m, 2H) ppm; MS (ESI) m/z: 337.75 [M + ACN + H + ].

[245] 3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)propanamide 25I1: procedure E; 0.56 g (1.82 mmol) 24I1, 0.06 g Pd/C, 20 mL MeOH, 18 h; 3.3 bar hydrogen, transparent oil (0.57 g,

1.82 mmol, 100%); Ή-NMR (250 MHz, acetone-de) d = h.hh - 7.31 (m, 6H), 7.09 - 7.01 (m, 2H),

6.82 (bs, lH), 6.14 (bs, lH), 4.71 (t, J=7- 8 Hz, lH), 3.03 - 2.98 (m, 2H) ppm; MS (ESI) m/z: 311.85 [M + H + ].

[246] 3-(4-Fluorophenyl)-3-(4-(trifluoromethoxy)phenyl)propanamide 251: procedure E; 0.40 g (1.23 mmol) 241, 0.40 g Pd/C, 25 mL MeOH, 14 h hydrogen atmosphere via a balloon, transparent oil (0.39 g, 1.19 mmol, 96%); Ή-NMR (250 MHz, acetone-d 6 ) d = 7.56 - 7.18 (m, 6H), 7.09 - 6.99 (m, 2H), 6.79 (bs, lH), 6.13 (bs, lH), 4.66 (t, J=7.8 Hz, lH), 2.99 - 2.94 (m, 2H) ppm; MS (ESI) m/z: 327.75 [M + H + ].

[247] 3,3-Bis(4-fluorophenyl)propenamide 25J: procedure E; 0.50 g (1.93 mmol) 24j, 0.05 g Pd/C, 30 mL MeOH, 43 h hydrogen atmosphere via a balloon, transparent oil (0.37 g, 1.42 mmol, 74%); Ή-NMR (250 MHz, acetone-de) d = 7.42 - 7.20 (m, 4H), 7.07 - 6.97 (m, 4H), 6.78 (bs, lH), 6.15 (bs, lH), 4.61 (t, J=7- 8 Hz, lH), 2.93 (d, J=8.3 Hz, 2H) ppm; MS (ESI) m/z: 261.85 [M + H + ].

[248] General procedure for reduction of primary amides 25a-j to the corresponding amines ioi,k-s (Procedure F)

[249] 1 eq amide derivative 25a-j was dissolved in dry THF and cooled to o °C. 1.5-2.5 eq LiAlH 4 -solution in THF were added slowly. The mixture was heated under reflux conditions for 2-5 h. After cooling to room temperature the hydride was quenched with acetone or ethyl acetate and stirred with saturated aqueous potassium sodium tartrate solution for at least 30 min. The phases were separated, and the organic solvent was removed under reduced pressure. The residue was purified with column chromatography to obtain a colorless oil.

[250] 3-(4-Fluorophenyl)-3-phenylpropan-i-amine 101: procedure F; 0.30 g (1.23 mmol) 25a, 3.1 mL (3.0 mmol, 1 M in THF) LiAlH 4 solution, 5 mL THF, 5 h reflux, eluent of column chromatography hexane:acetone 1:2; 1% NEt 3 ; yellow oil (0.17 g, 0.72 mmol, 59%); Ή-NMR (250 MHz, CDCI 3 ) d = 7-23 (s, 7H), 6.92 - 6.83 (m, 2H), 3.95 (t, J=7- 9 Hz, lH), 3.05 (t, J=7- 2 Hz, 2H), 1.92 (t, J= 1.3 Hz, 2H) ppm; MS (ESI) m/z: 229.95 [M + H + ].

[251] 3-Phenyl-3-(4-(trifluoromethyl)phenyl)propan-i-amine 10k: procedure F; 0.31 g (1.06 mmol) 25b, 2.7 mL (2.65 mmol, 1 M in THF) LiAlH 4 solution, 5 mL THF, 5 h reflux, eluent of column chromatography DCM:MeOH 10:1; yellow oil (0.14 g, 0.49 mmol, 46%); Ή- NMR (250 MHz, acetone-d 6 ) d = 7.65 - 7.55 (m, 4H), 7.38 - 7.17 (m, 5H), 4.32 (t, =7-4 Hz, lH), 3.12 (t, J= 6.6 Hz, 2H), 2.39 (q, J=7.3 Hz, 2H) ppm; MS (ESI) m/z: 279.90 [M + H + ]. [252] 3-Phenyl-3-(4-(trifluoromethoxy)phenyl)propan-i-amine 10I: procedure F; 0.59 g (1.89 mmol) 25c, 8.5 mL (8.51 mmol, 1 M in THF) LiAlH 4 solution, 5 mL THF, 4 h reflux, eluent of column chromatography DCM:MeOH ammoma 10 :0.1-10:1; yellow oil (0.15 g, 0.52 mmol, 28%); Ή-NMR (250 MHz, acetone-d 6 ) d = 7.48 - 7.42 (m, 2H), 7.40 - 7.06 (m, 7H), 4.24 (t, J=7-7 Hz, lH), 3.14 (t, J=7-7 Hz, 2H), 2.35 (q, J=7.3 Hz, 2H) ppm; MS (ESI) m/z: 296.21 [M + H + ].

[253] 3-(4-Chlorophenyl)-3-phenylpropan-i-amine 10m: procedure F; 0.19 g (0.71 mmol) 25d, 1.8 mL (1.78 mmol, 1 M in THF) LiAlH 4 solution, 7 mL THF, 6 h reflux, eluent of column chromatography DCM:MeOH ammoma 95:5; yellow oil (0.09 g, 0.37 mmol, 51%); Ή-NMR (250 MHz, acetone-d 6 ) d = 7.35 - 7.13 (m, 9H), 4.18 (t, J= 8.0 Hz, lH), 3.09 (t, J= 8.2 Hz, 2H), 2.38 - 2.24 (m, 2H) ppm; MS (ESI) m/z: 245.70 [M + H + ].

[254] 3-(3,4-Dichlorophenyl)-3-phenylpropan-i-amine ion: procedure F; 1.26 g (4.29 mmol) 2 5g, 9-4 mL (9.44 mmol, 1 M in THF) LiAlH 4 solution, 10 mL THF, 6 h reflux, eluent of column chromatography DCM:MeOH ammoma 95:5; yellow oil (0.53 g, 1.89 mmol, 44%); Ή-NMR (400 MHz, acetone-d 6 ) d = 7.55 - 7.42 (m, 2H), 7.32 - 7.28 (m, 6H), 4.22 (t, J=7.5 Hz, lH), 3.11 (t, J=7.5 Hz, 2H), 2.42 - 2.25 (m, 2H) ppm; MS (ESI) m/z: 322.25 [M + ACN + H + ].

[255] 4-(3-Amino-i-phenylpropyl)aniline 100: procedure F; 0.48 g (1.99 mmol) 25e, 5.0 mL (4:97 mmol, 1 M in THF) LiAlH 4 solution, 20 mL THF, 16 h reflux, eluent of column chromatography DCM:MeOH ammoma 98:2-95:5; yellow oil (0.23 g, 0.99 mmol, 50%); Ή-NMR (250 MHz, CD 3 CN) d = 7-42 - 6.90 (m, 7H), 6.58 - 6.52 (m, 2H), 4.31 - 3.69 (m, 3H), 2.55 - 2.45 (m, 2H), 2.10 - 2.01 (m, 2H) ppm; MS (ESI) m/z: 227.00 [M + H + ].

[256] 4-(3-Amino-i-phenylpropyl)benzenesulfonamide lop: procedure F; 0.53 g (1.74 mmol) 25f, 7.8 mL (7.81 mmol, 1 M in THF) LiAlH 4 solution, 26 mL THF, 5 h reflux, eluent of column chromatography DCM:MeOH ammoma 98:2; yellow oil (0.20 g, 0.67 mmol, 39%); Ή-NMR (250 MHz, acetone-d 6 ) d = 7.84 - 7.77 (m, 2H), 7.54 - 7.48 (m, 2H), 7.37 - 7.30 (m, 4H), 7.23 - 7.14 (m, lH), 6.47 (s, 2H), 4.29 (t, J=7.9 Hz, lH), 3.11 (t, J= 6.9 Hz, 2H), 2.37 (q, J=7.2 Hz, 2H ) ppm; MS (ESI) m/z: 289.10 [M - H + ].

[257] 3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)propan-i-ami ne loq: procedure F; 0.39 g (1.24 mmol) 25I1, 3.7 mL (3.72 mmol, 1 M in THF) LiAlH 4 solution, 15 mL THF, 4 h reflux, eluent of column chromatography DCM:MeOH ammoma 10 :0.1-10:1; yellow oil (0.23 g, 0.79 mmol, 64%); Ή-NMR (250 MHz, CD 3 CN) d = 7.66 - 7·6i (m, 2H), 7.52 (d, J=8.i Hz, 2H), 7.40 - 7-33 (m, 2H), 7.12 - 7.03 (m, 2H), 4.35 (t, J=7- 9 Hz, lH), 3.43 (2H, bs), 2.84- 2.78 (m, 2H), 2.54 - 2.45 (m, 2H) ppm; MS (ESI) m/z: 339.00 [M +ACN + H + ].

[258] 3-(4-Fluorophenyl)-3-(4-(trifluoromethoxy)phenyl)propan-i-am ine lor: procedure F; 0.67 g (2.07 mmol) 251, 4.4 mL (4.35 mmol, 1 M in THF) LiAlH 4 solution, 5 mL THF, 4 h reflux, eluent of column chromatography DCM:MeOH ammoma 10 :0.1-10:1; yellow oil (0.34 g, 1.09 mmol, 52%); Ή-NMR (250 MHz, DMSO-d 6 ) d = 7.44 - 7.24 (m, 6H), 7.14 - 7.06 (m, 2H), 4.19 (t, J= 8.2 Hz, lH), 2.45 - 2.37 (m, 2H), 2.13 - 2.02 (m, 2H) ppm; MS (ESI) m/z : 313.90 [M + H + ].

[259] 3,3-Bis(4-fluorophenyl)propan-i-amine 10s: procedure F; 0.34 g (1.31 mmol) 25J, 3.9 m L (3-94 mmol, 1 M in THF) LiAlH 4 solution, 5 mL THF, 3.5 h reflux, eluent of column chromatography DCM:MeOH 9:1; yellow oil (0.16 g, 0.66 mmol, 50%); Ή-NMR (250 MHz, CD 3 CN) d = 7-34 - 7-25 (m, 4H), 7.07-7.OO (m, 4H), 4.10 (t, J=7- 5 Hz, lH), 2.50 (t, J=7- O Hz, 2H), 2.13 - 2.04 (m, 2H) ppm; MS (ESI) m/z: 247.90 [M + H + ].

[260] lV-(2-Chloroethyl)-lV-phenylaniline 27: 3.5 mL (58.5 mmol, 6.5 eq) chloroacetic acid were diluted in 100 mL toluene over a time period of 20 min. 1.7 g (45.0 mmol, 5 eq) NaBH 4 were added portionwise. The suspension stirred for 3 h at room temperature. Additionally, 1.3 mL (9.0 mmol, 1 eq) diphenylamine 26 were added and the reaction mixture stirred for 4 h under reflux conditions. The reaction was allowed to cool to room temperature and was quenched with 2 M aqueous NaOH. The phases were separated, and the organic phase was washed with brine. The organic phase was dried over MgS0 4 , filtrated and the solvent was removed under reduced pressure. Column chromatography (hexane:ethyl acetate 15:1) obtained a transparent oil (1.4 g, 6.0 mmol, 67%); Ή-NMR (250 MHz, DMSO-de) d =7-33 - 7.26 (m, 4H), 7.07 - 6.92 (m, 6H), 4.05 (t, J= 6.8 Hz, 2H), 3.75 (t, J= 6.7 Hz, 2H) ppm; MS (ESI) m/z: 231.85 [M + H + ].

[261] 2-(2-(Diphenylamino)ethyl)isoindolin-i,3-dione 28: 1.39 g (6 mmol, 1 eq) 27 were dissolved in 5 ml DMF and 1.12 g (6 mmol, 1 eq) potassium phthalimide were added. The mixture was heated to 140 °C for 1 h under microwave irradiation. The reaction mixture was diluted with ethyl acetate and washed with water (3x). The aqueous phase was extracted with ethyl acetate and the combined phases were dried over MgS0 4 . After filtration and evaporation the residue was purified with column chromatography (hexane:ethyl acetate 9:i-5:i). A yellow wax was isolated (1.32 g, 3.86 mmol, 64%); Ή-NMR (250 MHz, DMS0-d 6 ) d = hhh (s, 4H), 7.24 - 7-i7(m, 4H), 7.00 - 6.84 (m, 6H), 4.04 - 3.99 (m, 2H), 3.86 - 3.81 (m, 2H) ppm; MS (ESI) m/z: 365.05 [M + Na + ].

[262] N 1 , A^-Diphenylethan-i, 2-diamine lot: 1.22 g (3.56 mmol, 1 eq) 28 were dissolved in 10 mL EtOH. 1.8 mL (35.60 mmol, 10 eq) hydrazine hydrate were added and the reaction stirred for 16 h under reflux conditions and 20 h at room temperature. The solvent was removed, and the residue was suspended in hexane. The solid was filtered off and the organic solvent was evaporated under reduced pressure. Column chromatography yielded a yellow oil (0.10 g, 0.49 mmol, 14%); Ή-NMR (250 MHz, DMSO-de) d = 7.24 - 7.22 (m, 6H), 7.01 - 6.87 (m, 4H), 6.68 (bs, 2H), 3.67 (t, J=7.i Hz, 2H), 2.71 (t, J=7-3 Hz, 2H) ppm; MS (ESI) m/z: 212.90 [M + H + ]. [263] (S', )-4-(ieri-Butyl)-3-(3-(4-(trifluoromethyl)phenyl)acryloyl)ox azolidin-2-one 30: To a solution of 0.34 g (2.32 mmol, 1 eq) 0S)-4-tert-butyl-2-oxazolidinone 1.0 mL (2.55 mmol, 2.5 M in THF, 1.1 eq) nBuLi were added dropwise at -78 °C. The mixture was stirred for 15 min and afterwards 0.54 g (2.32 mmol) (E)-3-(4-(trifluoromethyl)phenyl)acryloyl chloride 29, dissolved in 4 mL dry DCM, were added dropwise and stirred for 1 h at -78 °C and 30 min at o °C. (29 was prepared as followed: 0.52 g (2.32 mmol, 1 eq) (E)-3-(4-(trifluoromethyl)-phenyl)acrylic acid 22a were solved in 4 mL dry THF and cooled with an ice bath. 0.3 mL (2.78 mmol, 1.2 eq) oxalyl chloride were added and the reaction mixture stirred for 17 h at room temperature. The suspension was evaporated under reduced pressure). The mixture was quenched with saturated aqueous NH 4 C1 solution and extracted with ethyl acetate (3x). The combined organic phase was dried over MgS0 4 , filtered and the solvent was evaporated. The residue was purified with column chromatography (hexane:ethyl acetate 9:1, 5:1, 3:1); yellow oil (0.53 g, 1.55 mmol, 67%, Ή-NMR (250 MHz, CDCI 3 ) d = 8.01 (d, J= 15.7 Hz, lH), 7.91 - 7.58 (m, 5H), 4.62 - 4.55 (m, lH), 4.38 - 4.22 (m, 2H), 0.98 (s, 9H) ppm; MS (ESI) m/z: 342.03 [M + H + ].

[264] GS)-4-(tert-Butyl)-3-(CR)-3-(4-fluorophenyl)-3-(4-(trifluoro methyl)phenyl)- propanoyl)oxazolidin-2-one 31: 0.32 g (0.94 mmol, 1 eq) 30, 0.26 g (1.88 mmol, 2 eq) (4- fluorophenyl)boronic acid, 0.01 g (0.05 mmol, 0.05 eq) Pd(0Ac) 2 and 0.03 g (0.19 mmol, 0.2 eq) 2,2’-bipyridine were suspended in MeOH:water (1:3) in a microwave vial. The vial was sealed and heated to 80 °C for 14 h. The suspension was diluted with ethyl acetate and washed with 2 M aqueous NaOH (2x), with water (lx) and brine (lx). The organic phase was dried over MgS0 4 , filtered and the solvent was evaporated under reduced pressure. The residue was purified with column chromatography (hexane:ethyl acetate 5:i-3:i); off-white solid (0.25 g, 0.56 mmol, 60%); Ή-NMR (250 MHz, CDC1 3 ) d = 7-55 (d, =8.2 Hz, 2H), 7.40 (d, J=8.3 Hz, 2H), 7.34 - 7.08 (m, 2H), 7.02 - 6.94 (m, 2H), 4.74 (t, J=7- 8 Hz, lH), 4.38 - 4 0 9 (m, 3H), 3-91 (dd, J=7- 8, 16.7 Hz, lH), 3.60 (dd, J=7-9, 16.7 Hz, lH), 0.75 (s, 9H) ppm; MS (ESI) m/z: 436.15 [M - H + ].

[265] (R)-3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)propan-i -ol 32: Under an inert atmosphere 0.41 g (0.94 mmol) 31 were dissolved in 5 mL diy Et 2 0 and cooled with an ice bath. 0.8 mL (1.68 mmol, 1.8 eq) LiBH 4 suspension in dry THF were added and the mixture stirred for 4 h at room temperature. The reaction was quenched with acetone and stirred with saturated aqueous potassium sodium tartrate solution for 30 min. The solution was extracted with ethyl acetate (3x) and the combined organic phases were dried over MgS0 4 . After filtration and removal of the solvent the residue was purified with column chromatography (hexane:acetone 7:i-3:i); white solid (0.25 g, 0.56 mmol, 60%); Ή-NMR (250 MHz, CDC1 3 ) d = 7.56 - 7.52 (m, 2H), 7.36 - 7.33 (m, 2H), 7.24 - 7.16 (m, 2H), 7.02 - 6.96 (m, 2H), 4.24 (t, J=7- 9 Hz, lH), 3.60 (t, J= 6.0 Hz, 2H), 2.33 - 2.26 (m, 2H), 1.55 (bs, lH) ppm; MS (ESI) m/z: 297.15 [M - H + ]. [266] (i?)-3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)propana l 33: Under an inert atmosphere 0.15 g (0.51 mmol) 32, 0.33 g (0.77 mmol, 1.5 eq) Dess-Martin periodinane in 10 mL DCM and 0.01 mL water were stirred for 2.5 h at room temperature. The suspension was diluted with DCM and quenched with saturated aqueous NaHC0 3 solutiomsaturated aqueous NaS 2 0 3 solution (1:1). The phases were separated, and the aqueous phase was extracted with DCM (3x). The combined organic phase was washed with saturated aqueous NaHC0 3 solution (2x), water (2x) and brine (lx). The organic phase was dried over MgS0 4 , filtrated and evaporated. The purification with column chromatography (hexane:ethyl acetate 5:i-2:i) yielded a transparent oil (0.11 g, 0.37 mmol, 72%), Ή-NMR (250 MHz, CDC1 3 ) d = 9.76 (t, 7=1.5 Hz, lH), 7.56 (d, J= 8.4 Hz, 2H), 7.32 (d, J= 8.3 Hz, 2H), 7.22 - 7.13 (m, 2H), 7.06 - 6.95 (m, 2H), 4.69 (t, J=7- 6 Hz, lH), 3.19 (dd, J= 1.5, 7.6 Hz, 2H) ppm; MS (ESI) m/z: 295.25 [M - H + ].

[267] (i?)-3-(4-Fluorophenyl)-3-(4-(trifluoromethyl)phenyl)propan- i-amine IOU: A saturated solution of 0.29 g (3.68 mmol, 10 eq) dry NH 4 OAc in 20 mL Ethanol was prepared. To this solution 0.11 g (0.37 mmol) 33 were added and a suspension was formed. 0.49 g (2.21 mmol, 6 eq) sodium triacetoxyboro hydride and 3 mL (32% in water, 24.1 mmol, 66 eq) ammonia were added and the mixture stirred for 13 h under reflux conditions. The reaction mixture was allowed to cool to room temperature and was diluted with ethyl acetate and saturated aqueous NaHC0 3 solution (1:1). The phases were separated, and the aqueous phase was extracted with ethyl acetate (3x). The combined organic phase was dried over MgS0 4 and filtered. The solvent was removed under reduced pressure and the residue was purified via column chromatography (D CM : MeOH ammonia 9:i). A yellowish oil was obtained (0.05 g, 0.18 mmol, 48%); Ή-NMR (250 MHz, CDCL j ) d = 7.56 - 7.48 (m, 2H), 7.35 - 7.29 (m, 2H), 7.21 - 7.16 (m, 2H), 7.01 - 6.90 (m, 2H), 4.60 (s, 2H), 4.03 - 4.03 (m, lH), 2.88 - 2.74 (m, 2H), 2.56 - 2.41 (m, 2H) ppm; MS (ESI) m/z: 298.10 [M + H + ].

[268] sEH expression and purification

[269] The full length protein (aai-aa555) was isolated as published previously by Hahn et al. and Lukin et al. The protocols were slightly modified. In brief, sEH was expressed in E. coli BL2i-(DE3) cells with the ZYP5052 autoinduction media with kanamycin at 16 °C for 36 h. After lyses the protein was isolated by nickel affinity chromatography and a size exclusion chromatography. Buffer for the size exclusion was 50 mM Tris, 500 mM NaCl, 5% glycerol (HC1) pH 8. If the protein was stored at -80 °C glycerol was added (final concentration 25% v/v). Aliquots of the protein were flash frozen with liquid nitrogen and stored at -80 °C.

[270] sEH-H activity assay with PHOME

[271] To determine the IC 50 values of the potential inhibitors a fluorescence assay was adapted from Hahn et al. and Lukin et al. The non-fluorescent substrate PHOME (3-phenyl-cyano(6- methoxy-2-naphthalenyl)methyl ester-2-oxiraneacetic acid) is hydrolyzed to the fluorescent 6- methoxy-2-naphtaldehyde by the sEH hydrolase activity. To monitor the fluorescence (/ ¾ .„,= 330 nm and A ex = 465 nm) 1 pL of a DMSO dilution series of the compound to test was pipetted into a black flat bottom 96-well plate. 89 pL of a mix of recombinant human full length sEH (final concentration 3 nM in each well) in Bis-Tris buffer (pH 7) with 0.1 mg/ml BSA and Triton-X 100 (0.01% (w/v) final concentration) was added. After an incubation time of 30-45 min 10 pL of substrate in buffer (50 pM final concentration) were added quickly. The fluorescence was monitored for 30-45 mins (one point per minute) by a Tecan Infinite F200 Pro plate reader. As blank 1 pL pure DMSO and buffer without protein and as positive control 1 pL pure DMSO and the protein-buffer mix were used. All experiments were done in triplicates and in three independent measurements. To analyze the inhibitoiy potential of the tested compounds the percent inhibition was calculated by referencing the slope (in the linear phase) of the reaction to the slopes of the positive and negative controls. Further fitting was performed with the software GraphPad Prism 7 with a sigmoidal dose response curve fit (variable slope with 4 parameters).

REFERENCES

[272] The references are:

1. (1) Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L.

Inflammatory Responses and Inflammation-Associated Diseases in Organs. Oncotarget 2018, 9 (6), 7204-7218. https://d0i.0rg/10.18632/0nc0target.23208.

(2) Radmark, O.; Werz, O.; Steinhilber, D.; Samuelsson, B. 5-Lipoxygenase, a Key Enzyme for Leukotriene Biosynthesis in Health and Disease. Biochim. Biophys. Acta 2015, 1851 (4), 331-339· https://d0i.0rg/10.1016/j.bbalip.2014.08.012.

(3) Haeggstrom, J. Z. Leukotriene Biosynthetic Enzymes as Therapeutic Targets. J. Clin. Invest. 2018, 128 (7), 2680-2690. https://doi.org/10.1172/JCI97945.

(4) Hiesinger, K.; Wagner, K. M.; Hammock, B. D.; Proschak, E.; Hwang, S. H. Development of Multitarget Agents Possessing Soluble Epoxide Hydrolase Inhibitory Activity. Prostaglandins Other Lipid Mediat. 2019, 140, 31-39. https://d0i.0rg/10.1016/j.pr0staglandins.2018.12.003.

(5) Hoxha, M.; Zappacosta, B. CYP-Derived Eicosanoids: Implications for Rheumatoid

Arthritis. Prostaglandins Other Lipid Mediat. 2020, 146, 106405. https://d0i.0rg/10.1016/j.pr0staglandins.2019.106405.

(6) Wagner, K. M.; McReynolds, C. B.; Schmidt, W. K.; Hammock, B. D. Soluble Epoxide

Hydrolase as a Therapeutic Target for Pain, Inflammatory and Neurodegenerative Diseases. Pharmacol. Ther. 2017, 180, 62-76. https:/ / doi.org/ 10. ioi6/j.pharmthera.2017.06.006.

(7) McGettigan, P.; Henry, D. Use of Non-Steroidal Anti-Inflammatory Drugs That Elevate Cardiovascular Risk: An Examination of Sales and Essential Medicines Lists in Low-, Middle-, and High-Income Countries. PLoS Med. 2013, 10 (2), eiooi388. https://d0i.0rg/10.1371/j0urnal.pmed.1001388.

(8) Becker, J. C.; Domschke, W.; Pohle, T. Current Approaches to Prevent NSAID-Induced Gastropathy— COX Selectivity and Beyond. Br. J. Clin. Pharmacol. 2004, 58 (6), 587- 600. https://d0i.0rg/10.1111/j.1365-2125.2004.02198.x. (9) Szczeklik, A. The Cyclooxygenase Theory of Aspirin-Induced Asthma. Eur. Respir. J. 1990, 3 (5), 588-593·

(10) Jung, O.; Jansen, F.; Mieth, A.; Barbosa-Sicard, E.; Pliquett, R. U.; Babelova, A.; Morisseau, C.; Hwang, S. H.; Tsai, C.; Hammock, B. D.; et al. Inhibition of the Soluble Epoxide Hydrolase Promotes Albuminuria in Mice with Progressive Renal Disease. PloS One 2010, 5 (8), eii979- https://d0i.0rg/10.1371/j0urnal.p0ne.0011979.

(11) Sala, A.; Proschak, E.; Steinhilber, D.; Rovati, G. E. Two-Pronged Approach to Anti- Inflammatory Therapy through the Modulation of the Arachidonic Acid Cascade. Biochem. Pharmacol. 2018, 158, 161-173. https://d0i.0rg/10.1016/j.bcp.2018.10.007.

(12) Liu, J.-Y.; Yang, J.; Inceoglu, B.; Qiu, H.; Ulu, A.; Hwang, S.-H.; Chiamvimonvat, N.;

Hammock, B. D. Inhibition of Soluble Epoxide Hydrolase Enhances the Anti- Inflammatory Effects of Aspirin and 5-Lipoxygenase Activation Protein Inhibitor in a Murine Model. Biochem. Pharmacol. 2010, 79 (6), 880-887. https:/ / doi.org/ I0.i0i6/j.bcp.2009.i0.025.

(13) Garscha, U.; Romp, E.; Pace, S.; Rossi, A.; Temml, V.; Schuster, D.; Konig, S.; Gerstmeier, J.; Liening, S.; Werner, M.; et al. Pharmacological Profile and Efficiency in Vivo of Diflapolin, the First Dual Inhibitor of 5-Lipoxygenase-Activating Protein and Soluble Epoxide Hydrolase. Sci. Rep. 2017, 7(1), 9398. https://d0i.0rg/10.1038/s41598- 017-09795-w.

(14) Meirer, K.; Glatzel, D.; Kretschmer, S.; Wittmann, S. K.; Hartmann, M.; Blocher, R.;

Angioni, C.; Geisslinger, G.; Steinhilber, D.; Hofmann, B.; et al. Design, Synthesis and Cellular Characterization of a Dual Inhibitor of 5-Lipoxygenase and Soluble Epoxide Hydrolase. Mol. Basel Switz. 2016, 22 (1). https: / / doi.org/ io.3390/molecules220ioo45.

(15) Hiesinger, K.; Schott, A.; Kramer, J. S.; Blocher, R.; Witt, F.; Wittmann, S. K.; Steinhilber, D.; Pogoryelov, D.; Gerstmeier, J.; Werz, O.; et al. Design of Dual Inhibitors of Soluble Epoxide Hydrolase and LTA4 Hydrolase. ACS Med. Chem. Lett. 2019. https:/ / doi.org/ 10.1021/ acsmedchemlett.9b00330.

(16) Hwang, S. H.; Wagner, K. M.; Morisseau, C.; Liu, J.-Y.; Dong, H.; Wecksler, A. T.;

Hammock, B. D. Synthesis and Structure-Activity Relationship Studies of Urea- Containing Pyrazoles as Dual Inhibitors of Cyclooxygenase-2 and Soluble Epoxide Hydrolase. J. Med. Chem. 2011, 54 (8), 3037-3050. https: / / doi.org/ I0.i02i/jm200i376.

(17) Zhang, G.; Panigrahy, D.; Hwang, S. H.; Yang, J.; Mahakian, L. M.; Wettersten, H. L; Liu, J.-Y.; Wang, Y.; Ingham, E. S.; Tam, S.; et al. Dual Inhibition of Cyclooxygenase-2 and Soluble Epoxide Hydrolase Synergistically Suppresses Primary Tumor Growth and Metastasis. Proc. Natl. Acad. Sci. U. S. A. 2014, 111 (30), 11127-11132. https://d0i.0rg/10.1073/pnas.1410432111.

(18) Gartung, A.; Yang, J.; Sukhatme, V. P.; Bielenberg, D. R.; Fernandes, D.; Chang, J.; Schmidt, B. A.; Hwang, S. H.; Zurakowski, D.; Huang, S.; et al. Suppression of Chemotherapy-Induced Cytokine/Lipid Mediator Surge and Ovarian Cancer by a Dual COX-2/SEH Inhibitor. Proc. Natl. Acad. Sci. U. S. A. 2019, 116 (5), 1698-1703. https: / / doi.org/ 10.1073/pnas.1803999116.

(19) Zhang, C.-Y.; Duan, J.-X.; Yang, H.-H.; Sun, C.-C.; Zhong, W.-J.; Tao, J.-H.; Guan, X.-X.; Jiang, H.-L.; Hammock, B. D.; Hwang, S. H.; et al. COX-2/SEH Dual Inhibitor PTUPB Alleviates Bleomycin-Induced Pulmonary Fibrosis in Mice via Inhibiting Senescence. FEBSJ. 2019. https://doi.org/lo.llll/febs.i5io5.

(20) Dileepan, M.; Rastle-Simpson, S.; Greenberg, Y.; Wijesinghe, D. S.; Kumar, N. G.; Yang, J.; Hwang, S. H.; Hammock, B. D.; Sriramarao, P.; Rao, S. P. Effect Of Dual SEH/COX-2 Inhibition on Allergen-Induced Airway Inflammation. Front. Pharmacol. 2019, 10, 1118. https: / / doi.org/ 10.3389Zfphar.2019.01118. (21) Brooks, C. D.; Stewart, A. 0.; Basha, A.; Bhatia, P.; Ratajczyk, J. D.; Martin, J. G.; Craig,

R. A.; Kolasa, T.; Bouska, J. B.; Lanni, C. (R)-(+)-N-[3-[5-[(4-Fluorophenyl)Methyl]-2- Thienyl]-i-Methyl- 2-Propynyl]-N-Hydroxyurea (ABT-761), a Second-Generation 5- Lipoxygenase Inhibitor. J. Med. Chem. 1995, 38 (24), 4768-4775. https: / / doi.org/ io.i02i/jmooo24aoo4.

(22) Podolin, P. L.; Bolognese, B. J.; Foley, J. F.; Long, E.; Peck, B.; Umbrecht, S.; Zhang, X.; Zhu, P.; Schwartz, B.; Xie, W.; et al. In Vitro and in Vivo Characterization of a Novel Soluble Epoxide Hydrolase Inhibitor. Prostaglandins Other Lipid Mediat. 2013, 104- 105, 25-31. https://d0i.0rg/10.1016/j.pr0staglandins.2013.02.001.

(23) Klapars, A.; Buchwald, S. L. Copper-Catalyzed Halogen Exchange in Aryl Halides: An Aromatic Finkelstein Reaction. J. Am. Chem. Soc. 2002, 124 (50), 14844-14845. https://doi.org/ I0.i02i/ja028865v.

(24) Zhi, W.; Li, J.; Zou, D.; Wu, Y.; Wu, Y. Palladium-Catalyzed Diastereoselective Synthesis of b,b-Diarylpropionic Acid Derivatives and Its Application to the Total Synthesis of (R)- Tolterodine and the Enantiomer of a Key Intermediate for MK-8718. Tetrahedron Lett. 2018, 59 (6), 537-540. https://d0i.0rg/10.1016/j.tetlet.2017.12.082.

(25) Wolf, N. M.; Morisseau, C.; Jones, P. D.; Hock, B.; Hammock, B. D. Development of a High-Throughput Screen for Soluble Epoxide Hydrolase Inhibition. Anal. Biochem. 2006, 355 (1), 71-80. https://d0i.0rg/10.1016/j.ab.2006.04.045.

(26) Kretschmer, S. B. M.; Woltersdorf, S.; Vogt, D.; Lillich, F. F.; Riihl, M.; Karas, M.; Maucher, I. V.; Roos, J.; Hafner, A.-K.; Kaiser, A.; et al. Characterization of the Molecular Mechanism of 5-Lipoxygenase Inhibition by 2-Aminothiazoles. Biochem. Pharmacol. 2017, 123, 52-62. https://d0i.0rg/10.1016/j.bcp.2016.09.021.

(27) Eldrup, A. B.; Soleymanzadeh, F.; Taylor, S. J.; Muegge, L; Farrow, N. A.; Joseph, D.; McKellop, K.; Man, C. C.; Kukulka, A.; De Lombaert, S. Structure-Based Optimization of Arylamides as Inhibitors of Soluble Epoxide Hydrolase. J. Med. Chem. 2009, 52 (19), 5880-5895. https://d0i.0rg/10.1021/jm9005302.

(28) Gilbert, N. C.; Bartlett, S. G.; Waight, M. T.; Neau, D. B.; Boeglin, W. E.; Brash, A. R.; Newcomer, M. E. The Structure of Human 5-Lipoxygenase. Science 2011, 331 (6014), 217-219. https:/ / doi.org/ 10.1126/science.1197203.

(29) Gilbert, N. C.; Rui, Z.; Neau, D. B.; Waight, M. T.; Bartlett, S. G.; Boeglin, W. E.; Brash, A. R.; Newcomer, M. E. Conversion of Human 5-Lipoxygenase to a 15-Lipoxygenase by a Point Mutation to Mimic Phosphorylation at Serine-663. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2012, 26 (8), 3222-3229. https://d0i.0rg/10.1096/5.12-205286.

(30) Hafner, A.-K.; Cernescu, M.; Hofmann, B.; Ermisch, M.; Hornig, M.; Metzner, J.; Schneider, G.; Brutschy, B.; Steinhilber, D. Dimerization of Human 5-Lipoxygenase. Biol. Chem. 2011, 392 (12), 1097-1111. https://d0i.0rg/10.1515/BC.2011.200.

(31) Stewart, A. O.; Brooks, D. W. N,0-Bis(Phenoxycarbonyl)Hydroxylamine: A New Reagent for the Direct Synthesis of Substituted N-Hydroxyureas. J. Org. Chem. 1992, 57 (18), 5020-5023. https://d0i.0rg/10.1021/j000044a046.

(32) Hahn, S.; Achenbach, J.; Buscato, E.; Klingler, F.-M.; Schroeder, M.; Meirer, K.; Hieke,

M.; Heering, J.; Barbosa-Sicard, E.; Loehr, F.; et al. Complementary Screening Techniques Yielded Fragments That Inhibit the Phosphatase Activity of Soluble Epoxide Hydrolase. ChemMedChem 2011, 6 (12), 2146-2149. https: / / doi.org/ io.ioo2/cmdc.2onoo433.

(33) Lukin, A.; Kramer, J.; Hartmann, M.; Weizel, L.; Hernandez-Olmos, V.; Falahati, K.; Burghardt, L; Kalinchenkova, N.; Bagnyukova, D.; Zhurilo, N.; et al. Discovery of Polar Spirocyclic Orally Bioavailable Urea Inhibitors of Soluble Epoxide Hydrolase. Bioorganic Chem. 2018, 80, 655-667. https://d0i.0rg/10.1016/j.bi00rg.2018.07.014. (34) Brungs, M.; Radmark, O.; Samuelsson, B.; Steinhilber, D. Sequential Induction of 5- Lipoxygenase Gene Expression and Activity in Mono Mac 6 Cells by Transforming Growth Factor Beta and 1,25-Dihydroxyvitamin D3. Proc. Natl. Acad. Sci. 1995, 92 (1), 107-111. https://d0i.0rg/10.1073/pnas.92.1.107.

(35) Steinhilber, D.; Herrmann, T.; Roth, H. J. Separation of Lipoxins and Leukotrienes from Human Granulocytes by High-Performance Liquid Chromatography with a Radial-Pak Cartridge after Extraction with an Octadecyl Reversed-Phase Column. J. Chromatogr. B. Biomed. Sci.App. 1989, 493, 361-366. https://doi. org/10.1016/80378-4347(00)82742-

(36) Werz, O.; Burkert, E.; Samuelsson, B.; Radmark, O.; Steinhilber, D. Activation of 5- Lipoxygenase by Cell Stress Is Calcium Independent in Human Polymorphonuclear Leukocytes. Blood 2002, 99 (3), 1044-1052. https://d0i.0rg/10.1182/bl00d.V99.3.1044.

(37) Xing, L.; McDonald, J. J.; Kolodziej, S. A.; Kurumbail, R. G.; Williams, J. M.; Warren, C. J.; O’Neal, J. M.; Skepner, J. E.; Roberds, S. L. Discovery of Potent Inhibitors of Soluble Epoxide Hydrolase by Combinatorial Library Design and Structure-Based Virtual Screening. J. Med. Chem. 2011, 54 (5), 1211-1222. https://d0i.0rg/10.1021/jm101382t.

(38) Hiesinger, K.; Kramer, J. S.; Achenbach, J.; Moser, D.; Weber, J.; Wittmann, S. K.; Morisseau, C.; Angioni, C.; Geisslinger, G.; Kahnt, A. S.; et al. Computer-Aided Selective Optimization of Side Activities of Talinolol. ACS Med. Chem. Lett. 2019, 10 (6), 899- 903. https://d0i.0rg/10.1021/acsmedchemlett.9b00075.

(39) Kabsch, W. Xds. Acta Crystallogr. D Biol. Crystallogr. 2010, 66 (2), 125-132.

(40) Adams, P. D.; Afonine, P. V.; Bunkoczi, G.; Chen, V. B.; Davis, I. W.; Echols, N.; Headd, J. J.; Hung, L.-W.; Kapral, G. J.; Grosse-Kunstleve, R. W. PHENIX: A Comprehensive Python-Based System for Macromolecular Structure Solution. Acta Crystallogr. D Biol. Crystallogr. 2010, 66 (2), 213-221.

(41) Oster, L.; Tapani, S.; Xue, Y.; Rack, H. Successful Generation of Structural Information for Fragment-Based Drug Discovery. Drug Discov. Today 2015, 20 (9), 1104-1111. https://d0i.0rg/10.1016/j.drudis.2015.04.005.

(42) Emsley, P.; Cowtan, K. Coot: Model-Building Tools for Molecular Graphics. Acta Crystallogr. D Biol. Crystallogr. 2004, 60 (12), 2126-2132.

(43) Moriarty, N. W.; Grosse-Kunstleve, R. W.; Adams, P. D. Electronic Ligand Builder and Optimization Workbench (ELBOW): A Tool for Ligand Coordinate and Restraint Generation. Acta Crystallogr. D Biol. Crystallogr. 2009, 65 (10), 1074-1080.

(44) Schierle, S.; Flauaus, C.; Heitel, P.; Willems, S.; Schmidt, J.; Kaiser, A.; Weizel, L.; Goebel, T.; Kahnt, A. S.; Geisslinger, G.; et al. Boosting Anti-Inflammatory Potency of Zafirlukast by Designed Polypharmacology. J. Med. Chem. 2018, 61 (13), 5758-5764. https:/ / doi.org/ 10.1021/ acs.jmedchem.8boo458.

(45) Blocher, R.; Lamers, C.; Wittmann, S. K.; Merk, D.; Hartmann, M.; Weizel, L.; Diehl, O.;

Bruggerhoff, A.; BoB, M.; Kaiser, A.; et al. N-Benzylbenzamides: A Novel Merged Scaffold for Orally Available Dual Soluble Epoxide Hydrolase/Peroxisome Proliferator- Activated Receptor g Modulators. J. Med. Chem. 2016, 59 (1), 61-81. https:/ / doi.org/ io.i02i/acs.jmedchem.5boi239.