Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
HETEROCYCLIC P2Y 14 RECEPTOR ANTAGONISTS
Document Type and Number:
WIPO Patent Application WO/2022/155037
Kind Code:
A1
Abstract:
Disclosed are compounds for treating or preventing a disease or disorder responsive to antagonism of a P2Y14R receptor agonist in a mammal in need thereof, for example, compounds of formulas (I) and (II), wherein R1‑R8, X, Y, Z, X', Y', Z', and A are as defined herein, that are useful in treating an inflammatory such as asthma, cystic fibrosis, and sterile inflammation of the kidney.

Inventors:
JACOBSON KENNETH A (US)
JUNG YOUNG-HWAN (US)
WEN ZHIWEI (US)
Application Number:
PCT/US2022/011226
Publication Date:
July 21, 2022
Filing Date:
January 05, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
US HEALTH (US)
JACOBSON KENNETH A (US)
JUNG YOUNG HWAN (US)
WEN ZHIWEI (US)
International Classes:
C07D211/18; A61K31/4192; A61K31/44; A61P3/10; A61P25/00; A61P29/00; C07D205/04; C07D209/02; C07D211/34; C07D211/70; C07D223/04; C07D249/06; C07D255/02; C07D261/08; C07D403/10
Domestic Patent References:
WO2019157417A12019-08-15
WO2009070873A12009-06-11
Other References:
JUNG, Y. H.YU, J.WEN, Z.SALMASO, V.PHUNG, N. B.KARCZ, T.CHEN, Z.DUCA, S.BENNETT, J. M.DUDAS, S.: "Exploration of alternative scaffolds for P2Y 14 receptor antagonists containing a biaryl core", J. MED. CHEM., vol. 63, 2020, pages 9563 - 9589, XP055905973
MUFTI FATMA ET AL: "P2Y 14 Receptor Antagonists Reverse Chronic Neuropathic Pain in a Mouse Model", ACS MEDICINAL CHEMISTRY LETTERS, vol. 11, no. 6, 11 June 2020 (2020-06-11), US, pages 1281 - 1286, XP055905980, ISSN: 1948-5875, Retrieved from the Internet DOI: 10.1021/acsmedchemlett.0c00115
MEISTER JAROSLAWNA ET AL: "The G Protein-coupled Receptor P2Y14 Influences Insulin Release and Smooth Muscle Function in Mice", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 289, no. 34, 1 August 2014 (2014-08-01), US, pages 23353 - 23366, XP055907207, ISSN: 0021-9258, DOI: 10.1074/jbc.M114.580803
"Remington's Pharmaceutical Sciences", 1990, MACK PUBLISHING COMPANY, pages: 1445
BERGE, S. M., JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 1 - 19
"Pharmaceutics and Pharmacy Practice", 1982, J. B. LIPPINCOTT CO., pages: 238 - 250
"ASHP Handbook on Injectable Drugs", 1986, pages: 622 - 630
YU, J.CIANCETTA, A.DUDAS, S.DUCA, S.LOTTERMOSER, J.JACOBSON, K.A.: "Structure-guided modification of heterocyclic antagonists of the P2YI4 receptor", J. MED. CHEM., vol. 61, 2018, pages 4860 - 4882, XP002791818, DOI: 10.1021/acs.jmedchem.8b00168
JUNG, Y. H.YU, J.WEN, Z.SALMASO, V.PHUNG, N. B.KARCZ, T.CHEN, Z.DUCA, S.BENNETT, J. M.DUDAS, S.: "Exploration of alternative scaffolds for P2Y 14 receptor antagonists containing a biaryl core", J. MED. CHEM., vol. 63, 2020, pages 9563 - 9589
BATTISTONE, M. A.MENDELSOHN, A. C.SPALLANZANI, R. G.ALLEGRETTI, A. S.LIBERMAN, R. N.SESMA, J.KALIM, S.WALL, S. M.BONVENTRE, J. V.L: "Pro-inflammatory P2Yi4 receptor inhibition protects against ischemic acute kidney injury in mice", J. CLIN. INVEST., vol. 130, 2020, pages 3734 - 3749
XU, J.MORINAGA, H.OH, D.LI, P.CHEN, A.TALUKDAR, S.LAZAROWSKI, E.;OLEFSKY, J. M.KIM, J. J.: "GPR105 ablation prevents inflammation and improves insulin sensitivity in mice with diet-induced obesity", J. IMMUNOL., vol. 189, no. 4, 2012, pages 1992 - 1999
JAIN, S.JACOBSON, K. A.: "80th Scientific Session", 2020, AMERICAN DIABETES ASSOC., article "Adipocyte specific ablation of P2Y14R improves glucose metabolism in mice with diet-induced obesity"
KARCZ, T. P.WHITEHEAD, G. S.NAKANO, H.WILLIAMS, J. G.DETERDING, L. J.JACOBSON, K. ACOOK, D. N., UDP-GLUCOSE AND P2YI4 RECEPTOR AMPLIFY ALLERGEN-INDUCED AIRWAY EOSINOPHILIA
MUFTI, F.JUNG, Y. H.GIANCOTTI, L. A.YU, J.CHEN, Z..PHUNG, N. B.JACOBSON, K. A.SALVEMINI, D.: "P2Y 14 receptor antagonists reverse chronic neuropathic pain in a mouse model", ACS MED. CHEM. LETT., vol. 11, 2020, pages 1281 - 1286
LIN, JLIU, F.ZHANG, Y-Y.SONG, N.LIU, M-K.FANG, X-Y.LIAO, D-QZHOU, C.WANG, H.SHEN, J-F.: "P2Yi4 receptor is functionally expressed in satellite glial cells and mediates interleukin-1β and chemokine CCL2 secretion", J. CELL. PHYSIOL., vol. 234, 2019, pages 21199 - 21210, XP071324281, DOI: 10.1002/jcp.28726
JACOBSON, K. A.; DELICADO, E. G.; GACHET, C.; KENNEDY, C.; VON KIIGELGEN, I.; LI, B.; MIRAS-PORTUGAL, T.; NOVAK, I.; SCHONEBERG, T: "Update ofP2Y receptor pharmacology: IUPHAR review:''27", BR..J. PHARMACOL., vol. 177, 2020, pages 2413 - 2433, XP071094126, DOI: 10.1111/bph.15005
KINOSHITA, MNASU-TADA, K.FUJISHITA, K.SATO, K.KOIZUMI, S: "Secretion of matrix metalloproteinase-9 from astrocytes by inhibition of tonic P2Yi -receptor-mediated signal(s", CELL. MOL. NEUROBIOL., vol. 33, 2013, pages 47 - 58, XP035154138, DOI: 10.1007/s10571-012-9869-4
ZHANG, Z.HAO, K.LI, H.LU, R.LIU, C.ZHOU, M.LI, B.MENG, Z.HU, Q.JIANG, C: "Design, synthesis and anti-inflammatory evaluation of 3-amide benzoic acid derivatives as novel P2Yi4 receptor antagonists", EUR. J. MED. CHEM., vol. 181, 2019, pages 111564, XP085857433, DOI: 10.1016/j.ejmech.2019.111564
HAMEL, M.HENAULT, M.HYJAZIE, H.MORIN, N.BAYLY, C.SKOREY, K.THERIEN, A.G.MANCINI, J.BRIDEAU, C.KARGMAN, S: "Discovery of novel P2Yi4 agonist and antagonist using conventional and nonconventional methods", J. BIOMOL. SCREENING, vol. 16, no. 9, 2011, pages 1098 - 1105, XP055347326, DOI: 10.1177/1087057111415525
WANG, W.LIU, C.LI, H.TIAN, S.LIU, Y.WANG, N.YAN, D.LI, H.HU, Q: "Discovery of novel and potent P2Y14R antagonists via structure-based virtual screening for the treatment of acute gouty arthritis", J. ADV. RES., vol. 23, 2020, pages 133 - 142, XP086081478, DOI: 10.1016/j.jare.2020.02.007
JUNKER, A.BALASUBRAMANIAN, R.CIANCETTA, AULIASSI, E.KISELEV, E.MARTIRIGGIANO, C.TRUJILLO, KMTCHEDLIDZE, G.BIRDWELL, L.BROWN, K. A.: "Structure-based design of 3-(4-aryl-lH-l,2,3-triazol-l-yl)-biphenyl derivatives as P2YI4 receptor antagonists", J. MED. CHEM., vol. 59, 2016, pages 6149 - 6168
ZHANG, K.; ZHANG, J.; GAO, Z. G.; PAOLETTA, S.; ZHANG, D.; HAN, G. W.; LI, T.; MA, L.; ZHANG, W.; MIILLER, C. E.; YANG, H.; JIANG,: "Agonist-bound structure of the human P2Y12R receptor", NATURE, vol. 509, 2014, pages 119 - 122, XP055200854, DOI: 10.1038/nature13288
KISELEV, E.BARRETT, M.KATRITCH, V.PAOLETTA, S.WEITZER, C. DHAMMES, E.YIN, A. L.ZHAO, Q.STEVENS, R. C.HARDEN, T. K.: "Exploring a 2-naphthoic acid template for the structure-based design of P2Y 14 receptor antagonist molecular probes", ACS CHEM. BIOL., vol. 9, 2014, pages 2833 - 2842, XP055625193, DOI: 10.1021/cb500614p
BARRETT, M. O.SESMA, J. I.BALL, C. B.JAYASEKARA, P. S.JACOBSON, K. ALAZAROWSKI, E. R.HARDEN, T. K.: "A selective high affinity antagonist of the P2Yi4 receptor inhibits UDP-glucose-stimulated chemotaxis of human neutrophils", MOL. PHARMACOL., vol. 84, 2013, pages 41 - 49, XP055347174, DOI: 10.1124/mol.113.085654
ROBICHAUD, J.; FOURNIER, J.-F.; GAGNE, S.; GAUTHIER, J. Y.; HAMEL, M.; HAN, Y.; HENAULT, M.; KARGMAN, S.; LEVESQUE, J. F.; MAMANE,: "Applying the pro-drug approach to afford highly bioavailable antagonists of P2Yi4", BIOORG. MED. CHEM. LETT., vol. 27, no. 14, 2011, pages 4366 - 4368
MAZAK, K.NOSZAL, B: "Physicochemical properties of zwitterionic drugs in therapy", CHEMMEDCHEM, vol. I5, 2020, pages 1102 - 1
SABBADIN, D.CIANCETTA, A.MORO, S: "Bridging molecular docking to membrane molecular dynamics to investigate GPCR-ligand recognition: The human A2A adenosine receptor as a key study", J. CHEM. INF. MODEL., vol. 54, no. 1, 2014, pages 169 - 183
STRUPCZEWSKI, J. T.ALLEN, R. C.GARDNER, B. A.SCHMID, B. L.STACHE, U.GLAMKOWSKI, E. J.JONES, M. C.ELLIS, D. B.HUGER, F. P.DUNN, R. : "Synthesis and neuroleptic activity of 3-(l-substituted-4-piperidinyl)-l,2-benzisoxazoles", J. MED. CHEM., vol. 28, 1985, pages 761 - 769, XP001106671, DOI: 10.1021/jm00383a012
BALLESTEROS, J. A.WEINSTEIN, H: "Integrated methods for the construction of three dimensional models and computational probing of structure function relations in G protein-coupled receptors", METHODS NEUROSCI, vol. 25, 1995, pages 366 - 428, XP009139377, DOI: 10.1016/S1043-9471(05)80049-7
PENNINGTON, L. D.MOUSTAKAS, D. T.: "The necessary nitrogen atom: A versatile high-impact design element for multiparameter optimization", J. MED. CHEM., vol. 60, no. 9, 2017, pages 3552 - 3579, XP055542615, DOI: 10.1021/acs.jmedchem.6b01807
GARDELL, S.J.; HOPF, M.; KHAN, A.; DISPAGNA M, HAMPTON SESSIONS. E.; FALTER, R.; KAPOOR, N.; BROOKS, J.; CULVER, J.; PETUCCI, C; M: "Boosting NAD+ with a small molecule that activates NAMPT", NAT. COMMUN., vol. 10, 2019, pages 3241, XP055712364, DOI: 10.1038/s41467-019-11078-z
GAUTHIER, J. Y.; BELLEY, M.; DESCHENES, D.; FOUMIER, J. F.; GAGNE, S.; GAREAU, Y.; HAMEL, M.; HENAULT, M.; HYJAZIE, H.; KARGMAN, S: "The identification of 4,7-disubstituted naphthoic acid derivatives as UDP-competitive antagonists of P2Y14", BIOORG. MED. CHEM. LETT., vol. 21, 2011, pages 2836 - 2839, XP028208731, DOI: 10.1016/j.bmcl.2011.03.081
VALKÓ, K.: "Application of high-performance liquid chromatography based measurements of lipophilicity to model biological distribution", J. CHROMATOGR. A, vol. 1037, 2004, pages 299 - 310, XP004507002, DOI: 10.1016/j.chroma.2003.10.084
BESNARD, J.; RUDA, G. F.; SETOLA, V.; ABECASSIS, K.; RODRIGUIZ, R. M.; HUANG, X. P.; NORVAL, S.; SASSANO, M. F.; SHIN, A. I.; WEBS: "Automated design of ligands to polypharmacological profiles", NATURE, vol. 492, 2012, pages 215 - 220
WIBLE, B. A.; HAWRYLUK, P.; FICKER, E.; KURYSHEV, Y. A.; KIRSCH, G.; BROWN, A. M.: "HERG-Lite: a novel comprehensive high-throughput screen for drug-induced hERG risk", J. PHARMACOL. TOXICOL. METHODS, vol. 52, no. 1, 2005, pages 136 - 145, XP004985400, DOI: 10.1016/j.vascn.2005.03.008
PIPER, D. R.; DUFF, S. R.; ELIASON, H. C.; FRAZEE, W. J.; FREY, E. A.; FUERSTENAU-SHARP, M.; JACHEC, C.; MARKS, B. D.; POLLOK, B. : "Development of the predictor HERG fluorescence polarization assay using a membrane protein enrichment approach", ASSAY DRUG DEV. TECHNOL, vol. 6, 2008, pages 213 - 223, XP055609997, DOI: 10.1089/adt.2008.137
PEREZ CDANIEL KBCOHEN SM: "Evaluating prodrug strategies for esterase-triggered release of alcohols", CHEMMEDCHEM, vol. 8, 2013, pages 1662 - 1667
BENNETT, G. J.XIE, Y. K.: "A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man", PAIN, vol. 33, 1988, pages 87 - 107, XP024378950, DOI: 10.1016/0304-3959(88)90209-6
WHITEHEAD, G. S.THOMAS, S. Y.SHALABY, K. H.NAKANO, K.MORAN, T. P.WARD, J. M.FLAKE, G. P.NAKANO, H.COOK, D. N.: "TNF is required for TLR ligand-mediated but not protease-mediated allergic airway inflammation", J. CLIN. INVEST., vol. 127, 2017, pages 3313 - 3326
MIGNANI, S.RODRIGUES, J.TOMAS, H.JALAL, RSINGH, P. P.MAJORAL, J.-P.VISHWAKARMA, R. A.: "Present drug-likeness filters in medicinal chemistry during the hit and lead optimization process: how far can they be simplified?", DRUG DISC. TODAY, vol. 23, no. 3, 2018, pages 605 - 615
DALVIE, D. K.KALGUTKAR, A. S.KHOJASTEH-BAKHT, S. C.OBACH, R. S.O'DONNELL, J. P: "Biotransformation reactions of five-membered aromatic heterocyclic rings", CHEM. RES. TOXICOL., vol. 75, no. 3, 2002, pages 269 - 299, XP055239789, Retrieved from the Internet DOI: 10.1021/tx015574b
ZHU, J.MO, J.LIN, H.CHEN, Y.SUN, H.: "The recent progress of isoxazole in medicinal chemistry", BIOORG. MED. CHEM., vol. 26, no. 12, 2018, pages 3065 - 3075, XP085408648, DOI: 10.1016/j.bmc.2018.05.013
SUN, H.SHI, M.ZHANG, W.ZHENG, Y.-M.XU, Y.-Z.SHI, J.-J.LIU, T.;GUNOSEWOYO, H.PANG, T.GAO, Z. B.: "Development of novel alkoxyisoxazoles as sigma-1 receptor antagonists with antinociceptive efficacy", J. MED. CHEM., vol. 59, 2016, pages 6329 - 6343
RATNATILAKA NABHUKET, P.JITHAVECH, P.ONGPIPATTANAKUL, B.ROJSITTHISAK, P: "Interspecies differences in stability kinetics and plasma esterases involved in hydrolytic activation of curcumin diethyl disuccinate, a prodrug of curcumin", RSC ADVANCES, vol. 9, no. 8, 2019, pages 4626 - 4634
"Schrodinger Release 2019-3: Maestro", 2019, SCHRODINGER, LLC
SASTRY, G. M.ADZHIGIREY, MDAY, T.ANNABHIMOJU, R.SHERMAN, W: "Protein and ligand preparation: Parameters, protocols, and influence on virtual screening enrichments", J. COMPUT. AIDEDMOL. DES., vol. 27, no. 3, 2013, pages 221 - 234
HARDER, E.; DAMM, W.; MAPLE, J.; WU, C.; REBOUL, M.; XIANG, J. Y.; WANG, L.; LUPYAN, D.; DAHLGREN, M. K.; KNIGHT, J. L.; KAUS, J. : "OPLS3: A force field providing broad coverage of drug-like small molecules and proteins", J. CHEM. THEORY COMPUT., vol. 12, no. 1, 2016, pages 281 - 296
FRIESNER, R. A.BANKS, J. L.MURPHY, R. B.HALGREN, T. A.KLICIC, J. J.MAINZ, D. T.REPASKY, M. P.KNOLL, E. H.SHELLEY, M.PERRY, J. K.: "Glide: A new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy", J. MED. CHEM., vol. 47, no. 7, 2004, pages 1739 - 1749, XP002592019, DOI: 10.1021/JM0306430
DOERR, S.HARVEY, M. J.NOE, F.DE FABRITIIS: "G. HTMD: High-throughput molecular dynamics for molecular discovery", J. CHEM. THEORY COMPUT., vol. 12, no. 4, 2016, pages 1845 - 1852
HUMPHREY, W.DALKE, A.SCHULTEN, K: "VMD: Visual molecular dynamics", J. MOL. GRAPH., vol. 14, no. 1, 1996, pages 33 - 38, XP055140690, DOI: 10.1016/0263-7855(96)00018-5
LOMIZE, M. APOGOZHEVA, I. D.JOO, H.MOSBERG, H. I.LOMIZE, A. L.: "OPM Database and PPM web server: Resources for positioning of proteins in membranes", NUCLEIC ACIDS RES., vol. 40, 2012, pages D370 - 6
JORGENSEN, W. L.CHANDRASEKHAR, J.MADURA, J. D.IMPEY, R. W.KLEIN, M. L.: "Comparison of simple potential functions for simulating liquid water", J. CHEM. PHYS., vol. 79, no. 2, 1983, pages 926
HARVEY, M. J.GIUPPONI, G.FABRITIIS, G. D: "ACEMD: Accelerating biomolecular dynamics in the microsecond time scale", J. CHEM. THEORY COMPUT., vol. 5, no. 6, 2009, pages 1632 - 1639
BEST, R. B.; ZHU, X.; SHIM, J.; LOPES, P. E. M.; MITTAL, J.; FEIG, M.; MACKERELL, A. D.: "Optimization of the additive CHARMM all-atom protein force field targeting improved sampling of the backbone cp, *( and side-chain Xi and %2 dihedral angles", J. CHEM. THEORY COMPUT., vol. 8, no. 9, 2012, pages 3257 - 3273
KLAUDA, J. B.VENABLE, R. M.FREITES, J. A.O'CONNOR, J. W.TOBIAS, D. J.MONDRAGON-RAMIREZ, C.VOROBYOV, IMACKERELL, A. D.PASTOR, R. W.: "Update of the CHARMM all-atom additive force field for lipids: Validation on six lipid types", J. PHYS. CHEM. B, vol. 114, no. 23, 2010, pages 7830 - 7843, XP055323592, DOI: 10.1021/jp101759q
VANOMMESLAEGHE, K.MACKERELL, A. D.: "Automation of the CHARMM general force field (CGenFF) I: Bond perception and atom typing", J. CHEM. INF. MODEL., vol. 52, no. 12, 2012, pages 3144 - 3154
VANOMMESLAEGHE, K.RAMAN, E. P.MACKERELL, A. D.: "Automation of the CHARMM general force field (CGenFF) II: Assignment of bonded parameters and partial atomic charges", J. CHEM. INF. MODEL., vol. 52, no. 12, 2012, pages 3155 - 3168
KRAUTLER, V.; VAN GUNSTEREN, W. F.; HIINENBERGER, P. H.: "A fast SHAKE algorithm to solve distance constraint equations for small molecules in molecular dynamics simulations", J. COMPUT. CHEM., vol. 22, no. 5, 2001, pages 501 - 508
ESSMANN, U.PERERA, LBERKOWITZ, M. L.DARDEN, T.LEE, H.PEDERSEN, L. G.: "A smooth particle mesh Ewald method", J. CHEM. PHYS., vol. 103, no. 19, 1995, pages 8577, XP055441882, DOI: 10.1063/1.470117
PHILLIPS, J. C.BRAUN, R.WANG, W.GUMBART, J.TAJKHORSHID, E.VILLA, E.CHIPOT, C.SKEEL, R. DKALE, L.SCHULTEN, K: "Scalable molecular dynamics with NAMD", J. COMPUT. CHEM., vol. 26, no. 16, 2005, pages 1781 - 1802, XP055484133, DOI: 10.1002/jcc.20289
WILLIAMS, T.KELLEY, C, GNUPLOT, Retrieved from the Internet
TETKO, I. V.TANCHUK, V. Y.: "Application of associative neural networks for prediction of lipophilicity in ALOGPS 2.1 program", J. CHEM. INF. COMPUT. SCI., vol. 42, 2002, pages 1136 - 1145
Attorney, Agent or Firm:
PILLAI, Xavier et al. (US)
Download PDF:
Claims:
CLAIM(S): 1. A compound of formula (I) or formula (II): wherein X R1 is halo or CF3, R2 is selected from the group consisting of COOH, COOR12, CONHOH, CH2CON(R11)2, CHR7OCOR8, (CH2)oNR9, tetrazolyl, CH2OPO(OH)2, and from the group consisting of NHR4, COOR5, (C≡C)n(CH2)mR6, CONH(CH2)qNH2, COCF

R is H or COR wherein R is C1-C6 alkyl, C6-C10 aryl, o R5 is C1-C6 alkyl, C3-C8 cycloalkyl, or benzyl, R6 is NH2, or CONH2, R7-R9 are independently hydrogen or C1-C6 alkyl, R10 is H, C1-C10 alkylcarbonyl, or C1-C10 alkyloxycarbonyl, R11 is hydrogen or C1-C6 alkyl, R12 is C1-C6 alkyl, C3-C8 cycloalkyl, or benzyl, m, o, and q are independently integers of 1 to about 10, and n is zero or an integer of 1 to about 10; a compound of formula (III)

wherein R1 is CF3, R2 is COOH eterocyclic ring having at least one nitrogen atom, wherein the heterocyclic ring comprises 1, 2, or 3 rings, fused or linked at one or more atoms, and wherein the heterocyclic ring is saturated or unsaturated; and wherein the heterocyclic ring is optionally substituted with one or more of substituents selected from the group consisting of alkyl, alkoxy, and hydroxy; or a compound of formula (IV) or (V): ety can a six membered sugar moiety, particularly an aldohexose including allose, altrose, glucose, mannose, gulose, idose, galactose, and talose, including derivatives thereof, particularly the following derivatives thereof, wherein:

2. The compound, salt, or stereoisomer of claim 1, wherein R1 is CF3. 3. The compound, salt, or stereoisomer of claim 1 or 2, wherein the compound is of formula (I).

4. The compound, salt, or stereoisomer of any one of claims 1-3, wherein R3 is selected from NH2, NHCOCH3, NHCOC6H5, NHCOC(CH3)3 , and The compound, salt, or stereoisomer of any one of claims 1-3, wherein R3 is COOR5. 6. The compound, salt, or stereoisomer of any one of claims 1-3, wherein R3 is (C≡C)n(CH2)mR6 or CONH(CH2)qNH2. 7. The compound, salt, or stereoisomer of claim 6, wherein R3 is selected from CONH(CH2)3NH2, CH2CONH2, (CH2)3NH2, and C≡CCH2NH2. 8. The compound, salt, or stereoisomer of claim 1 or 2, wherein the compound is of formula (II). 9. The compound, salt, or stereoisomer of any one of claims 1-8, wherein R2 is H. 10. The compound, salt, or stereoisomer of any one of claims 1-9, wherein R2 is COOR12, CH2CON(R11)2, CHR7OCOR8, (CH2)oNR9, tetrazolyl, CH2OPO(OH)2, or und, salt, or stereoisomer of claim 1, wherein the compound is: wherein R2 is COOH or C

12. The compound, salt, or stereoisomer of claim 9, wherein R3 is selected from the group consisting of d . 13. The compound, salt, or stereoisomer of claim 12, wherein R3 i and wherein the configuration of the three chiral centers is (S,S,S). 14. The compound, salt, or stereoisomer of claim 9, wherein R3 is selected from the group consisting o selected from the group consisting of

16. The compound, salt, or stereoisomer of any one of claims 12-15, wherein R2 is H. 17. The compound, salt, or stereoisomer of any one of claims 12-15, wherein R2 is COOR12, CH2CON(R11)2, CHR7OCOR8, (CH2)oNR9, tetrazolyl, CH2OPO(OH)2, or mpound, salt, or stereoisomer of claim 1, wherein the compound is: wherein the configurat 19. The compound, salt or stereoisomer of claim 1, wherein the compound is of formula (III). 20. The compound, salt or stereoisomer of claim 1, wherein the compound is of formula (IV) or (V):

21. A pharmaceutical composition comprising a compound, salt, or stereoisomer of any one of claims 1-20 and a pharmaceutically acceptable carrier. 22. A compound, salt, or stereoisomer of any one of claims 1-20, or the pharmaceutical composition of claim 21, for use in antagonizing a P2Y14R receptor in a mammal in need thereof. 23. A compound, salt, or stereoisomer of any one of claims 1-20, or the pharmaceutical composition of claim 21, for use in treating or preventing an inflammatory condition in a mammal in need thereof. 24. The compound, salt, or stereoisomer for use according to claim 23, wherein the inflammatory condition is selected from the group consisting of asthma, cystic fibrosis, and sterile inflammation of the kidney. 25. A compound, salt, or stereoisomer of any one of claims 1-20, or the pharmaceutical composition of claim 21, for use in treating pain in a mammal in need thereof.

Description:
HETEROCYCLIC P2Y14 RECEPTOR ANTAGONISTS CROSS-REFERENCE TO A RELATED APPLICATION [0001] This patent application claims the benefit of United States Provisional Patent Application No.63/138,581, filed January 18, 2021, the disclosure of which is incorporated herein in its entirety for all purposes. STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT [0002] This invention was made with Government support under Grant Number ZIADK003116 awarded by the National Institutes of Health. The Government has certain rights in this invention. BACKGROUND OF THE INVENTION [0003] Extracellular nucleotides released by tissue and organs during stress or injury activate a class of cell-surface receptors (P2Rs) to boost the innate and adaptive immune responses (1-3). This mechanism acts as a time-dependent component of the signaling purinome, along with the anti-inflammatory adenosine receptors (ARs, also termed P1 receptors), to protect the organism in various challenged circumstances. The P2Y 14 receptor (P2Y 14 R) responds to endogenous agonists uridine-5′-diphosphoglucose and uridine-5′-diphosphate to mediate inflammatory activity, in part by activating neutrophil motility (4-6). Structurally, the P2Y14R belongs to the δ-branch of rhodoposin-like G protein-coupled receptors (GPCRs). Three subtypes of the P2YRs are preferentially coupled to inhibition of adenylate cyclase through guanine nucleotide inhibitory (G i ) protein: P2Y 12 R, P2Y 13 R and P2Y 14 R. Selective P2Y 14 R antagonists are sought as potential agents for treating asthma, sterile inflammation of the kidney, diabetes and neurodegeneration (7-12). However, only a few antagonists are known, so there is a clear need for more competitive P2Y 14 R antagonists. Other subtypes of the P2YR family in general, e.g., P2Y 2 R and P2Y 6 R, are also associated with proinflammatory effects, and their antagonists are desired for providing anti-inflammatory activity (13, 14). [0004] Antagonists of the P2Y14R were first reported by Black and colleagues (58), and of the two classes of antagonists reported, naphthoic acids and pyrido[4,3-d]pyrimidines, only the former appeared to be competitive antagonists. Thus, there is an unmet need for diverse competitive P2Y14R antagonists. BRIEF SUMMARY OF THE INVENTION [0005] The invention provides a compound of formula (I) or formula (II): wherein X, Y 1 R is halo or CF3, R 2 is selected from the group consisting of COOH, COOR 12 , CONHOH, CH2CON(R 11 )2, CHR 7 OCOR 8 , (CH2)oNR 9 , tetrazolyl, CH2OPO(OH)2 an R 3 is selected from the group consisting of NHR 4 , COO (CH 2 ) q NH 2 , R is H or COR wherein R is C1-C6 alkyl, C6-C10 aryl, o R 5 is C 1 -C 6 alkyl, C 3 -C 8 cycloalkyl, or benzyl, R 6 is NH2, or CONH2, R 7 -R 9 are independently hydrogen or C1-C6 alkyl, R 10 is H, C 1 -C 10 alkylcarbonyl, or C 1 -C 10 alkyloxycarbonyl, R 11 is hydrogen or C 1 -C 6 alkyl, R 12 is C1-C6 alkyl, C3-C8 cycloalkyl, or benzyl, and m, o, and q are independently integers of 1 to about 10, and n is zero or an integer of 1 to about 10, or a pharmaceutically acceptable salt thereof, as well as stereoisomers thereof. [0006] The present invention further provides a compound of formula (III)

wherein R 1 is CF 3 , R 2 is COO having at least one nitrogen atom. [0007] The present invention further provides a compound of formula (IV) or (V):

mammal in need thereof, comprising administering to the mammal an effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof. [0009] The invention further provides a method of treating or preventing an inflammatory condition in a mammal in need thereof, comprising administering to the mammal an effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof. [0010] The invention additionally provides a method of treating pain in a mammal in need thereof, comprising administering to the mammal an effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof. BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWING(S) [0011] FIG.1A-B show the inhibition of binding of fluorescent antagonists 11, 19, 29, and 32 at hP2Y 14 R (FIG.1A) and at mP2Y 14 R (FIG.2B) exhibited by four P2Y 14 R antagonist determinations, performed in triplicate). [0012] FIG.2 shows reversal of CCI-induced mechano-allodynia in the mouse induced by compound 32 (10 µmol/kg, i.p.). Mean±SD is shown, n=3, ANOVA, *P<0.05 vs. Day 0 and † P<0.05 vs. day 7 post-injury.32 was dissolved in 5% DMSO in water as vehicle, and a volume of 0.2 mL was injected. [0013] FIG.3 shows the effects of P2Y14R antagonists and their prodrugs, namely 32, 37b, 3a, 37a, 1b, and 37c, compared to reference antagonist 1a, during allergen challenge reduces eosinophilic airway inflammation in a protease model of asthma. Effect on BALF leukocytes of i.p. injection of the P2Y14R antagonists 2 d post-challenge of ASP/OVA-sensitized mice. * P <0.05, ** P <0.01, *** P <0.001, **** P <0.0001 vs. vehicle by one-way ANOVA followed by Dunnett’s test. [0014] FIG.4 shows the reversal of established mechano-allodynia in the CCI mouse model by compound 14 (10 µmol/kg, i.p.) at the time of peak pain, i.e. seven days post-injury. Mean±SD is shown, n=3, ANOVA, *P<0.05 vs. Day 0 and † P<0.05. The vehicle consisted of 5% aqueous DMSO (0.2 mL injected). [0015] FIG.5 shows the structures of reference compound 1a, compound 1b, compound 5, and fluorescent inhibitor 2. [0016] FIG.6 shows that compound 114 (10 µmol/kg, i.p.) reversed the established mechano-allodynia in the CCI mouse model at the time of peak pain, i.e., seven days post-injury. Mean ± SD is shown, n=3, ANOVA, *P<0.05 vs. Day 0 and † P<0.05. The vehicle consisted of 5% aqueous DMSO (0.2 mL injected). [0017] FIG.7A-E depict the activities of P2Y14R antagonists 114 and its ester prodrug 141, carbamate prodrug 142, and double prodrug 143 in a mouse model of asthma (dose: 20 µmol/kg (9.8–12.2 mg/kg); concentration: 2 µmol/mL (2 mM) in 10% DMSO, 30% PEG-400, 60% H 2 O; administration: 10 µL/g of body weight).7-Day sensitization with ovalbumin/aspergillus was followed by a single challenge (ovalbumin, aerosol) at day 14. Antagonists were given 30 min prior to challenge. Harvesting of cells and cell counting were on day 16. DETAILED DESCRIPTION OF THE INVENTION [0018] In an aspect, the invention provides a compound of formula (I) or formula (II): wherein X, 1 R is halo or CF 3 , R 2 is selected from the group consisting of COOH, COOR 12 , CONHOH, CH2CON(R 11 )2, CHR 7 OCOR 8 , (CH2)oNR 9 , tetrazolyl, CH2OPO(OH)2, and R 3 is selected from the group consisting of NHR 4 ,

R 4 is H or COR 6 wherein R 6 is C 1 -C 6 alkyl, C 6 -C 10 aryl, o R 5 is C 1 -C 6 alkyl, C 3 -C 8 cycloalkyl, or benzyl, R 6 is NH2, or CONH2, R 7 -R 9 are independently hydrogen or C1-C6 alkyl, R 10 is H, C 1 -C 10 alkylcarbonyl, or C 1 -C 10 alkyloxycarbonyl, R 11 is hydrogen or C1-C6 alkyl, R 12 is C1-C6 alkyl, C3-C8 cycloalkyl, or benzyl, and m, o, and q are independently integers of 1 to about 10, and n is zero or an integer of 1 to about 10, or a pharmaceutically acceptable salt thereof, as well as stereoisomers thereof. [0019] In certain aspects, R 1 is CF 3 . [0020] In certain aspects, the compound is of formula (I). [0021] In certain aspects of the compound of formula (I), R 3 is selected from NH2, NHCOCH3, NHCOC6H5, NHCOC(CH3)3 and [0022] In certain aspects, R 3 is COOR [0023] In certain aspects, R 3 is (C≡C) n(CH2)mR 6 or CONH(CH2)qNH2. [0024] In certain of these aspects, R 3 is selected from CONH(CH2)3NH2, CH2CONH2, (CH 2 ) 3 NH 2 , and C≡CCH 2 NH 2 . [0025] In certain aspects, R 3 is selected from Br, (CH 2 ) 2 -CN, [0027] In certain aspects of formula (II), R 3 is selected from Br, (CH 2 ) 2 -CN, [0028] In certain particular aspects, R 3 i [0029] In certain aspects, R 2 is H. 2 [0030] In certain aspects, R is COOR 12 , CH 2 CON(R 11 ) 2 , CHR 7 OCOR 8 , (CH 2 ) o NR 9 , tetrazolyl, CH 2 OPO(OH) 2 , o . [0031] In a particular asp

wherein R 2 is COOH or COOCH2C [0032] In certain aspects, R 3 is selected from [0033] In certain particular aspects, R 3 is , and wherein the configuration of the three chiral centers is (S,S,S).

[0034] In certain aspects, R 3 is selected from . [0036] In certain aspects, R 2 is COOR 12 , CH2CON(R 11 )2, CHR 7 OCOR 8 , (CH2)oNR 9 , tetrazolyl, CH2OPO(OH)2, o [0037] In a particular asp wherein the configuration about th [0038] The present invention further provides a compound of formula (III)

wherein R 1 is CF3, R 2 is COOH , and R 3 is a 4-8 membered heterocyclic ring having at least one nitrogen atom, wherein the heterocyclic ring comprises 1, 2, or 3 rings, fused or linked at one or more atoms, and wherein the heterocyclic ring is saturated or unsaturated, and wherein the heterocyclic ring is optionally substituted with one or more of substituents selected from the group consisting of alkyl, alkoxy, and hydroxy. Examples of compounds of formula (III) those wherein R 3 is: ; ; ; ; [ ] p p p ( ) ( )

, wherein: an a six membered sugar moiety, particularly an aldohexose, including allose, altrose, glucose, mannose, gulose, idose, galactose, or talose, including derivatives thereof, particularly acetyloxy derivatives thereof; for example, , straight-chain or branched alkyl substituent containing from, for example, 1 to about 6 carbon atoms, preferably from 1 to about 4 carbon atoms, more preferably from 1 to 2 carbon atoms. isobutyl, tert-butyl, pentyl, isoamyl, hexyl, and the like. [0041] The term “cycloalkyl,” as used herein, means a cyclic alkyl substituent containing from, for example, about 3 to about 8 carbon atoms, preferably from about 4 to about 7 carbon atoms, and more preferably from about 4 to about 6 carbon atoms. Examples of such substituents include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like. The cyclic alkyl groups may be unsubstituted or further substituted with alkyl groups such as methyl groups, ethyl groups, and the like. [0042] The term “alkylcarbonyl,” as used herein, refers to an alkyl group linked to a carbonyl group and further linked to a molecule via the carbonyl group, e.g., alkyl-C(=O)-. The term “alkoxycarbonyl,” as used herein, refers to an alkoxy group linked to a carbonyl group and further linked to a molecule via the carbonyl group, e.g., alkyl-O-C(=O)-. [0043] The term “halo” or “halogen,” as used herein, means a substituent selected from Group VIIA, such as, for example, fluorine, bromine, chlorine, and iodine. [0044] The term “tetrazolyl”, as used herein, refers to a group of the formula [0045] Whenever a range of the number of atoms in a structure is indicated ( C1-C8, C1-C6, C1-C4, or C2-C12, C2-C8, C2-C6, C2-C4 alkyl, alkenyl, alkynyl, etc.), it is specifically contemplated that any sub-range or individual number of carbon atoms falling within the indicated range also can be used. Thus, for instance, the recitation of a range of 1-8 carbon atoms (e.g., C1-C8), 1-6 carbon atoms (e.g., C1-C6), 1-4 carbon atoms (e.g., C1-C4), 1-3 carbon atoms (e.g., C1-C3), or 2-8 carbon atoms (e.g., C2-C8) as used with respect to any chemical group (e.g., alkyl, alkylamino, etc.) referenced herein encompasses and specifically describes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and/or 12 carbon atoms, as appropriate, as well as any sub-range thereof (e.g., 1-2 carbon atoms, 1-3 carbon atoms, 1-4 carbon atoms, 1-5 carbon atoms, 1-6 carbon atoms, 1-7 carbon atoms, 1-8 carbon atoms, 1-9 carbon atoms, 1-10 carbon atoms, 1-11 carbon atoms, 1-12 carbon atoms, 2-3 carbon atoms, 2-4 carbon atoms, 2-5 carbon atoms, 2-6 carbon atoms, 2-7 carbon atoms, 2-8 carbon atoms, 2-9 carbon atoms, 2-10 carbon atoms, 2-11 carbon atoms, 2-12 carbon atoms, 3-4 carbon atoms, 3-5 carbon atoms, 3-6 carbon atoms, 3-7 carbon atoms, 3-8 carbon atoms, 3-9 carbon atoms, 3-10 carbon atoms, 3-11 carbon atoms, 3-12 carbon atoms, 4-5 carbon atoms, 4-6 carbon atoms, 4-7 carbon atoms, 4-8 carbon atoms, 4-9 carbon atoms, 4-10 carbon atoms, 4-11 carbon atoms, and/or 4-12 carbon atoms, etc., as appropriate). Similarly, the recitation of a range of 6-10 carbon atoms (e.g., C6-C10) as used with respect to any chemical group (e.g., aryl) referenced herein encompasses and specifically describes 6, 7, 8, 9, and/or 10 carbon atoms, as appropriate, as well as any sub-range thereof (e.g., 6-10 carbon atoms, 6-9 carbon atoms, 6-8 carbon atoms, 6-7 carbon atoms, 7-10 carbon atoms, 7-9 carbon atoms, 7-8 carbon atoms, 8-10 carbon atoms, and/or 8-9 carbon atoms, etc., as appropriate). [0046] In any of the above aspects, the compound or salt of formula (I) or formula (II) can have at least one asymmetric carbon atom. When the compound or salt has at least one asymmetric carbon atom, the compound or salt can exist in the racemic form, in the form of its pure optical isomers, or in the form of a mixture wherein one isomer is enriched relative to the other. In particular, in accordance with the present invention, when the inventive compounds have a single asymmetric carbon atom, the inventive compounds may exist as racemates, i.e., as mixtures of equal amounts of optical isomers, i.e., equal amounts of two enantiomers, or in the form of a single enantiomer. As used herein, "single enantiomer" is intended to include a compound that comprises more than 50% of a single enantiomer (i.e., enantiomeric excess more than 60%, more than 70%, more than 80%, more than 90%, or up to 100% pure enantiomer). [0047] When the compound or salt has more than one chiral center, the compound or salt can therefore exist as a mixture of diastereomers or in the form of a single diastereomer. As used herein, “single diastereomer” is intended to mean a compound that comprises more than 50% of a single diastereomer (i.e., diastereomeric excess more than 60%, more than 70%, more than 80%, more than 90%, or up to 100% pure diastereomer). [0048] The phrase “pharmaceutically acceptable salt” is intended to include nontoxic salts synthesized from the parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two. Generally, nonaqueous media such as ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington’s Pharmaceutical Sciences, 18th ed., Mack Publishing Company, Easton, PA, 1990, p.1445, and Berge, S. M., at al., Journal of Pharmaceutical Science, 66, 1-19 (1977). [0049] Suitable bases include inorganic bases such as alkali and alkaline earth metal bases, e.g., those containing metallic cations such as sodium, potassium, magnesium, calcium and the like. Non-limiting examples of suitable bases include sodium hydroxide, potassium hydroxide, sodium carbonate, and potassium carbonate. Suitable acids include inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulfuric acid, phosphoric acid, and the like, and organic acids such as p-toluenesulfonic, methanesulfonic acid, benzenesulfonic acid, oxalic acid, p-bromophenylsulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, maleic acid, tartaric acid, fatty acids, long chain fatty acids, and the like. Preferred pharmaceutically acceptable salts of inventive compounds having an acidic moiety include sodium and potassium salts. Preferred pharmaceutically acceptable salts of inventive compounds having a basic moiety (e.g., a dimethylaminoalkyl group) include hydrochloride and hydrobromide salts. The compounds of the present invention containing an acidic or basic moiety are useful in the form of the free base or acid or in the form of a pharmaceutically acceptable salt thereof. [0050] It should be recognized that the particular counterion forming a part of any salt of this invention is usually not of a critical nature, so long as the salt as a whole is pharmacologically acceptable and as long as the counterion does not contribute undesired qualities to the salt as a whole. [0051] It is further understood that the above compounds and salts may form solvates, or exist in a substantially uncomplexed form, such as the anhydrous form. As used herein, the term "solvate" refers to a molecular complex wherein the solvent molecule, such as the crystallizing solvent, is incorporated into the crystal lattice. When the solvent incorporated in the solvate is water, the molecular complex is called a hydrate. Pharmaceutically acceptable solvates include hydrates, alcoholates such as methanolates and ethanolates, acetonitrilates and the like. These compounds can also exist in polymorphic forms. [0052] The present invention further provides a pharmaceutical composition comprising a compound as described above and a pharmaceutically acceptable carrier. The present invention provides a pharmaceutical composition comprising a pharmaceutically acceptable carrier and an effective amount, e.g., a therapeutically effective amount, including a prophylactically effective amount, of one or more of the aforesaid compounds, or salts thereof, of the present invention. [0053] The pharmaceutically acceptable carrier can be any of those conventionally used and is limited only by chemico-physical considerations, such as solubility and lack of reactivity with the compound, and by the route of administration. It will be appreciated by one of skill in the art that, in addition to the following described pharmaceutical compositions; the compounds of the present invention can be formulated as inclusion complexes, such as cyclodextrin inclusion complexes, or liposomes. [0054] The pharmaceutically acceptable carriers described herein, for example, vehicles, adjuvants, excipients, or diluents, are well known to those who are skilled in the art and are readily available to the public. It is preferred that the pharmaceutically acceptable carrier be one which is chemically inert to the active compounds and one which has no detrimental side effects or toxicity under the conditions of use. [0055] The choice of carrier will be determined in part by the particular active agent, as well as by the particular method used to administer the composition. Accordingly, there is a wide variety of suitable formulations of the pharmaceutical composition of the present invention. The following formulations for oral, aerosol, parenteral, subcutaneous, intravenous, intraarterial, intramuscular, interperitoneal, intrathecal, rectal, and vaginal administration are merely exemplary and are in no way limiting. [0056] Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice; (b) capsules, sachets, tablets, lozenges, and troches, each containing a predetermined amount of the active ingredient, as solids or granules; (c) powders; (d) suspensions in an appropriate liquid; and (e) suitable emulsions. Liquid formulations may include diluents, such as water and alcohols, for example, ethanol, benzyl alcohol, and the polyethylene alcohols, either with or without the addition of a pharmaceutically acceptable surfactant, suspending agent, or emulsifying agent. Capsule forms can be of the ordinary hard- or soft-shelled gelatin type containing, for example, surfactants, lubricants, and inert fillers, such as lactose, sucrose, calcium phosphate, and cornstarch. Tablet forms can include one or more of lactose, sucrose, mannitol, corn starch, potato starch, alginic acid, microcrystalline cellulose, acacia, gelatin, guar gum, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, calcium stearate, zinc stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, disintegrating agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible carriers. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such carriers as are known in the art. [0057] The compounds of the present invention, alone or in combination with other suitable components, can be made into aerosol formulations to be administered via inhalation. These aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also may be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer. [0058] Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The compound can be administered in a physiologically acceptable diluent in a pharmaceutical carrier, such as a sterile liquid or mixture of liquids, including water, saline, aqueous dextrose and related sugar solutions, an alcohol, such as ethanol, isopropanol, or hexadecyl alcohol, glycols, such as propylene glycol or polyethylene glycol, glycerol ketals, such as 2,2-dimethyl-1,3-dioxolane-4-methanol, ethers, such as poly(ethyleneglycol) 400, an oil, a fatty acid, a fatty acid ester or glyceride, or an acetylated fatty acid glyceride with or without the addition of a pharmaceutically acceptable surfactant, such as a soap or a detergent, suspending agent, such as pectin, carbomers, methylcellulose, hydroxypropylmethylcellulose, or carboxymethylcellulose, or emulsifying agents and other pharmaceutical adjuvants. [0059] Oils, which can be used in parenteral formulations include petroleum, animal, vegetable, or synthetic oils. Specific examples of oils include peanut, soybean, sesame, cottonseed, corn, olive, petrolatum, and mineral. Suitable fatty acids for use in parenteral formulations include oleic acid, stearic acid, and isostearic acid. Ethyl oleate and isopropyl myristate are examples of suitable fatty acid esters. Suitable soaps for use in parenteral formulations include fatty alkali metal, ammonium, and triethanolamine salts, and suitable detergents include (a) cationic detergents such as, for example, dimethyl dialkyl ammonium halides, and alkyl pyridinium halides, (b) anionic detergents such as, for example, alkyl, aryl, and olefin sulfonates, alkyl, olefin, ether, and monoglyceride sulfates, and sulfosuccinates, (c) nonionic detergents such as, for example, fatty amine oxides, fatty acid alkanolamides, and polyoxyethylene-polypropylene copolymers, (d) amphoteric detergents such as, for example, alkyl-beta-aminopropionates, and 2-alkyl-imidazoline quaternary ammonium salts, and (3) mixtures thereof. [0060] The parenteral formulations will typically contain from about 0.5 to about 25% by weight of the active ingredient in solution. Suitable preservatives and buffers can be used in such formulations. In order to minimize or eliminate irritation at the site of injection, such compositions may contain one or more nonionic surfactants having a hydrophile-lipophile balance (HLB) of from about 12 to about 17. The quantity of surfactant in such formulations ranges from about 5 to about 15% by weight. Suitable surfactants include polyethylene sorbitan fatty acid esters, such as sorbitan monooleate and the high molecular weight adducts of ethylene oxide with a hydrophobic base, formed by the condensation of propylene oxide with propylene glycol. The parenteral formulations can be presented in unit-dose or multi-dose sealed containers, such as ampoules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described. [0061] The compounds of the present invention may be made into injectable formulations. The requirements for effective pharmaceutical carriers for injectable compositions are well known to those of ordinary skill in the art. See Pharmaceutics and Pharmacy Practice, J. B. Lippincott Co., Philadelphia, Pa., Banker and Chalmers, eds., pages 238-250 (1982), and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 (1986). [0062] Topical formulations, including those that are useful for transdermal drug release, are well-known to those of skill in the art and are suitable in the context of the invention for application to skin. Topically applied compositions are generally in the form of liquids, creams, pastes, lotions and gels. Topical administration includes application to the oral mucosa, which includes the oral cavity, oral epithelium, palate, gingival, and the nasal mucosa. In some aspects, the composition contains at least one active component and a suitable vehicle or carrier. It may also contain other components, such as an anti-irritant. The carrier can be a liquid, solid or semi- solid. In aspects, the composition is an aqueous solution. Alternatively, the composition can be a dispersion, emulsion, gel, lotion or cream vehicle for the various components. In one aspect, the primary vehicle is water or a biocompatible solvent that is substantially neutral or that has been rendered substantially neutral. The liquid vehicle can include other materials, such as buffers, alcohols, glycerin, and mineral oils with various emulsifiers or dispersing agents as known in the art to obtain the desired pH, consistency and viscosity. It is possible that the compositions can be produced as solids, such as powders or granules. The solids can be applied directly or dissolved in water or a biocompatible solvent prior to use to form a solution that is substantially neutral or that has been rendered substantially neutral and that can then be applied to the target site. In aspects of the invention, the vehicle for topical application to the skin can include water, buffered solutions, various alcohols, glycols such as glycerin, lipid materials such as fatty acids, mineral oils, phosphoglycerides, collagen, gelatin and silicone based materials. [0063] Additionally, the compounds of the present invention may be made into suppositories by mixing with a variety of bases, such as emulsifying bases or water-soluble bases. Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams, or spray formulas containing, in addition to the active ingredient, such carriers as are known in the art to be appropriate. [0064] The dose administered to a mammal, particularly, a human, in accordance with the present invention should be sufficient to effect the desired response. Such responses include reversal or prevention of the adverse effects of the disease for which treatment is desired or to elicit the desired benefit. One skilled in the art will recognize that dosage will depend upon a variety of factors, including the age, condition, and body weight of the human, as well as the source, particular type of the disease, and extent of the disease in the human. The size of the dose will also be determined by the route, timing and frequency of administration as well as the existence, nature, and extent of any adverse side-effects that might accompany the administration of a particular compound and the desired physiological effect. It will be appreciated by one of skill in the art that various conditions or disease states may require prolonged treatment involving multiple administrations. [0065] Suitable doses and dosage regimens can be determined by conventional range-finding techniques known to those of ordinary skill in the art. Generally, treatment is initiated with smaller dosages that are less than the optimum dose of the compound. Thereafter, the dosage is increased by small increments until the optimum effect under the circumstances is reached. The present inventive method typically will involve the administration of about 0.1 to about 300 mg of one or more of the compounds described above per kg body weight of the animal or mammal. [0066] The therapeutically effective amount of the compound or compounds administered can vary depending upon the desired effects and the factors noted above. Typically, dosages will be between 0.01 mg/kg and 250 mg/kg of the subject’s body weight, and more typically between about 0.05 mg/kg and 100 mg/kg, such as from about 0.2 to about 80 mg/kg, from about 5 to about 40 mg/kg or from about 10 to about 30 mg/kg of the subject’s body weight. Thus, unit dosage forms can be formulated based upon the suitable ranges recited above and the subject’s body weight. The term “unit dosage form” as used herein refers to a physically discrete unit of therapeutic agent appropriate for the subject to be treated. [0067] Alternatively, dosages are calculated based on body surface area and from about 1 mg/m 2 to about 200 mg/m 2 , such as from about 5 mg/m 2 to about 100 mg/m 2 will be administered to the subject per day. In particular aspects, administration of the therapeutically effective amount of the compound or compounds involves administering to the subject from about 5 mg/m 2 to about 50 mg/m 2 , such as from about 10 mg/m 2 to about 40 mg/m 2 per day. It is currently believed that a single dosage of the compound or compounds is suitable, however a therapeutically effective dosage can be supplied over an extended period of time or in multiple doses per day. Thus, unit dosage forms also can be calculated using a subject’s body surface area based on the suitable ranges recited above and the desired dosing schedule. [0068] In certain aspects, the invention provides a method of antagonizing a P2Y 14 R receptor in a mammal in need thereof, comprising administering to the mammal an effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof. [0069] In certain aspects, the invention provides a method of treating or preventing an inflammatory condition in a mammal in need thereof, comprising administering to the mammal an effective amount of a compound of the invention or a pharmaceutically acceptable salt thereof. [0070] In these aspects, the inflammatory condition is selected from the group consisting of asthma, cystic fibrosis, and sterile inflammation of the kidney. [0071] In certain aspects, the invention provides a method of treating pain in a mammal in need thereof, comprising administering to the mammal an effective amount a compound of the invention or a pharmaceutically acceptable salt thereof. [0072] In certain aspects, the invention provides a compound of the invention or a pharmaceutically acceptable salt thereof, for use in antagonizing a P2Y 14 R receptor in a mammal in need thereof. [0073] In certain aspects, the invention provides a compound of the invention or a pharmaceutically acceptable salt thereof, for use in treating or preventing an inflammatory condition in a mammal in need thereof. [0074] In these aspects, the inflammatory condition is selected from the group consisting of asthma, cystic fibrosis, and sterile inflammation of the kidney. [0075] In certain aspects, the invention provides a compound of the invention or a pharmaceutically acceptable salt thereof, for use in treating pain in a mammal in need thereof. [0076] Chemistry [0077] Schemes 1-14 depict exemplary syntheses of compounds of the invention. [0078] Schemes 1A and 1B show synthesis of triazole-containing derivatives of the compound of formula (I) with acyl or acylamino substituents for R 3 . Scheme 1A

Scheme 1B:

Reagents and conditions: (a) Pd(PPh3)4, K2CO3, DMF, 85 °C, 5 h, 40-68%; (b) TFA:THF = 2:1, rt, 0.5 h, 57-87%; (c) KOH, MeOH:H 2 O = 2:1, 50 °C, 50-82%; (d) acetic anhydride, DCM, rt, 2 h, 76%; (e) benzoyl chloride, TEA, DCM, rt, 18 h, 63%; (f) Boc-isonipecotic acid (Boc-Inp- OH), HATU, DIPEA, DMF, rt, 2 h, 94%. [0079] Scheme 2 shows synthesis of triazole-containing derivatives of the compound of formula (I) with acyl or acylamino substituents for R 3 . Scheme 2 Reagents and conditions: (a) Pd(PPh3)4, K2CO3, DMF, 85 °C, 5 h, 27-55%; (b) PdCl2(dppf), dimethoxyethane: Na 2 CO 3 (aq, 2M) = 10:1, 60 °C, 4 h, 73%; (c) Pd(PPh 3 ) 4 , K 2 CO 3 , DMF:Water = 1:1, 90 °C, 12 h, 64%; (d) KOH, MeOH:H 2 O = 2:1, 50 °C, 39-88%; (e) TFA:THF = 2:1, rt, 0.5 h, 62-98%. [0080] Scheme 3 shows synthesis of triazole-containing derivatives of the compound of formula (I) with cyclic groups for R 3 . Scheme 3 N N N O N O N N O F C N F C N F C N 3 O 3 OH K 2 CO 3 , DMF, μW, 100 °C, 0.5 h, 80-91%; (c) Pd(PPh 3 ) 4 , K 2 CO 3 , DMF:water = 1:1, 90 °C, 12 h, 30-70%; (d) KOH, MeOH:H2O = 2:1, 50 °C, 43-74%; (e) TFA:THF = 2:1, rt, 0.5 h, 52-90%. [0081] Scheme 4 shows a synthesis of naphthalene-containing isoxazole derivative 32. Scheme 4

MeOH:H2O = 2:1, 50 °C, 98%. [0082] Scheme 5 shows a synthesis of amide-containing isoxazole derivative 33. Scheme 5 , HATU, DIPEA, DMF, rt, 5 h, 70%; (c) B 2 pin 2 , PdCl 2 (dppf), KOAc, dioxane, 70 °C, 6 h, 65%; (d) Pd(PPh3)4, K2CO3, DMF, 85 °C, 5 h, 45%; (e) KOH, MeOH, H2O, 50 o C, 3 h, 62%. [0083] Scheme 6 shows a synthesis of triazole-containing derivatives 34 – 36. Scheme 6

azidotrimethylsilane, acetonitrile, rt, 1 h, 37-65%; (c) 4-ethynyl-α,α,α-trifluorotoluene, CuSO 4 . 5H 2 O, Na ascorbate, TBTA, tert-BuOH:H 2 O=1:1, rt, 45 min, 36-69%; (d) tert-butyl 4-(4- (4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)piperidi ne-1-carboxylate, Pd(PPh3)4, K2CO3, DMF, 85 o C, 4 h, 35-91%; (e) KOH, MeOH:H2O = 2:1, 50 o C, 5 h, 68-91%; (f) TFA:THF = 2:1, rt, 0.5 h, 60-65%. [0084] Scheme 7 shows a synthesis of compounds 37a – 37c. Scheme 7

72%; (b) TFA:THF = 2:1, rt, 0.5 h, 78%; (c) 2-chloro-N,N-dimethylacetamide, Cs 2 CO 3 , DMF, 45 o C, 1 h, 68%. [0085] Scheme 8 shows the preparation of 4-bromophenyl intermediates containing modified piperidine rings for Suzuki coupling. Scheme 8

t, overnight, 60–62%; (c) TMSCF 3 , NaI, THF, 65°C, overnight, 26%; (d) TFA, DCM, rt, 1 h, 75%; (e) (+) C-C (-) (4.5 V, 2 F/mol), MeOH, Et4NOTs, 95%; (f) NH4Cl, 100 o C, 73%; (g) i, dibromobenzene, magnesium, THF, sonication; ii, ketone, THF, 0 o C, 2 h; 42–86%; (h) H 2 SO 4 , 80 o C, 30 min, 15%. (i) K 2 CO 3 , PhCH 2 Br, DMF, 70 o C. [0086] Scheme 9 shows a general scheme for preparation of derivatives of the lead P2Y14R antagonist, piperidine 1, via Suzuki coupling followed by ester hydrolysis. Compound 63 is 3-(4- bromophenyl)pyrrolidine hydrochloride, starting material for 75. Scheme 9

Reagents and conditions: (a) Pd(PPh 3 ) 4 , K 2 CO 3 , DMF, MW, 150 o C, 30 min, 37-76%; (b) for substrate 56 and 57: Pd(PPh3)4, aq. Na2CO3, DME-EtOH, 80 o C, 30 min, 25–40%; (c) MeOH, aq KOH, overnight, 42–76%. [0087] Scheme 10 shows a preparation of bridged piperidine analogues 114 and 116 containing a 2-azanorbornane moiety, 119 and 121 containing a lactam; and ring-opened 134 and 136 containing 1-amino-3-hydroxymethylcyclopentane, and 138 and 140 containing 1-amino-3- carboxylcyclopentane. The synthesis maintained absolute stereochemistry originating from chiral bicyclic precursor 176.

Reagents and conditions: (a) 1-bromo-4-iodobenzene, (PPh3)2PdCl2, formic acid, TEA, THF, 65 o C overnight, quantitative; (b) TEA, DMAP, (Boc) 2 O, DCM, rt, overnight.46% 178 and 36% 179; (c) TFA, DCM, rt, 1 h, 88–95%; (d) H 3 N-BH 3 , B(C 6 F 5 ) 3 , BF 3 Et 2 O, DCE, 75 o C, 24 h, 52% 182 and 48% 183; (e) boronic acid pinacol ester (164), Na2CO3, Pd(PPh3)4, DME-H2O (4:1), 85 °C, overnight, 68–93%; (f) LiOH, THF-MeOH-H 2 O (3:1:1), rt, 3 h, 44–66%; (g) LiOH, THF- MeOH-H 2 O (3:1:1), rt, 1.5 h, 40–62%; (h) LiOH, THF-MeOH-H 2 O (3:1:1), rt, overnight, 73– 77%. (i) 2-chloro-N,N-dimethylacetamide, Cs2CO3, DMF, 45 o C, 2 h, 86%. [0088] Scheme 11 shows a preparation of analogues 115 and 117 with 2-azanorbornane, 120 and 122 with lactam, 135 and 137 with 1-amino-3-hydroxymethylcyclopentane, and 139 and 141 with 1-amino-3-carboxylcyclopentane with absolute stereochemistry from 192. a

Scheme 11 HF, 65 o C overnight, quantitative; (b) TEA, DMAP, (Boc)2O, DCM, rt, overnight.55% 194 and 41% 195; (c) TFA, DCM, rt, 1 h, 78–79%; (d) H3N-BH3, B(C6F5)3, BF3Et2O, DCE, 75 o C, 24 h, 51% 198 and 44% 199; (e) 164, Na 2 CO 3 , Pd(PPh 3 ) 4 , DME-H 2 O (4:1), 85 °C, overnight, 68–93%; (f) LiOH, THF-MeOH-H2O (3:1:1), rt, 3 h, 44–66%; (g) LiOH, THF-MeOH-H2O (3:1:1), rt, 1.5 h, 40–62%; (h) LiOH, THF-MeOH-H2O (3:1:1), rt, overnight, 73–77%. (i) 2-chloro-N,N- dimethylacetamide, Cs 2 CO 3 , DMF, 45 o C, 2 h, 80%. [0089] Scheme 12 shows a preparation of N-acetyl 2-azanorbornane derivatives. Scheme 12 %. [0090] Scheme 13 shows a preparation of nortropane derivatives from hydroxylated intermediate 127s. [0091] Scheme 13 Pd/C, 100 psi, 3 h, overall yield 19% from 127s. [0092] Scheme 14 shows a preparation of isonortropane derivatives, including both hydroxylated 131, 132 and nonhydroxylated 133 and 133s analogues. [0093] Scheme 14 5 psi), overnight, 20%. [0094] Scheme 15 shows the preparation of isoquinuclidine derivatives from hydroxylated intermediate 206. [0095] Scheme 15 H2, Pd/C, 100 psi, 3 h. [0096] Scheme 16 shows a preparation of compounds 1c and 1d.

Scheme 16 d = 2:1, v/v), 0 oC for 3 h → room temperature for 12 h, 71%.1d: (b) Trifluoroacetic anhydride, 0 o C for 1hr to rt for 2 hr. [0097] Scheme 17 shows a preparation of phosphate 3b.

Scheme 17 diethylphosphoramidite, tetrazole, THF, rt, 1 h, then MCPBA, -78 o C, 84%; (c) TFA:THF = 2:1, rt, 2 h, 69%. [0098] Synthesis of compounds of formula (IV) or (V): PPTN analogues with β-D-glucose bridged with triazole were prepared via click reaction (Scheme 18). The azide 523 was obtained from substitution of bromo starting material 522 with sodium azide. PPTN analogues 525-527 modified with alkynes tethered at the N of piperidine were prepared by reacting PPTN with iodo-substituted alkynes. The click reaction between alkynes 525-527 and azide 523 was catalyzed by CuSO4/sodium ascorbate at 90 o C overnight and provided acetyl protected ligands 504, 506, and 507, which were further hydrolyzed to yield PPTN analogues 508, 510, and 511 with β-D-glucose bridged by various-length alkyltriazolyl linker, respectively. [0099] Scheme 18. PPTN analogues with β-D-glucose bridged by triazole (A) OAc OAc A cO O a A O N OH rt, overnight, 43%; (c) 523, CuSO4.5H2O, sodium ascorbate, DMF/H2O (9:1), 90 o C, overnight, quantitative; (d) NaOH (3 M), MeOH, rt, overnight, 79%. Scheme 19. PPTN-glucose analogues bridged with propyltriazolyl linker CF 3 O Reagents and conditions: (a) 5-iodo-1-pentyne, CuSO4.5H2O, sodium ascorbate, DMF/H2O (9:1), 90 o C, overnight; (b) 502, K 2 CO 3 , DMF, rt, overnight; (c) NaOH (3 M), MeOH, rt, overnight. Scheme 20. Preparation of C-α/β-D-glycosyl analogue of PPTN O O OH OH (b) In(OTf)3, acetone, rt, overnight; (c) NaBH(OAc)3, HOAc, DCM, 0-5 o C, 2 h; (d) NaOH (3 M), MeOH, rt, overnight. Scheme 21. Alternative synthetic strategy for the preparation of C-β-D-glycosyl analogue of PPTN MOMO HO MOMO c a MOMO O b MOMO O B Reagents and conditions: (a) i) chloromethyl methyl ether, DIPEA, Bu4NI, DCM, 0 o C to rt, 48 h; ii) dimethyltitanocene, toluene, 70 o C, 24 h; (b) 9-BBN-H, THF, reflux, 5 h; (c) 1-bromo-2- iodo-ethane, Pd(PPh 3 ) 4 , K 2 PO 4 , dioxane, 60 o C, 24 h; (d) 2, K 2 CO 3 , DMF, rt, overnight; (e) HCl, MeOH, H2O, rt, overnight. Scheme 22. Preparation of O-β-D-glycosyl analogue of PPTN F 3 C O F 3 C F C O 3 O H NaOH (3 M), MeOH, rt, overnight; (c) 522, Ag2O, TMSOTf, DMF, rt, 5 min; (d) NaOH (3 M), MeOH, rt, overnight. Scheme 23. Preparation of O-α-D-glycosyl analogue of PPTN

tonitrile, reflux, 5 h; (c) 541, Ag 2 O, TMSOTf, DMF, rt, 5 min; (d) i) H 2 , Pd/C, DMF, 5 h; ii) NaOH (3 M), MeOH, rt, overnight Scheme 24. Preparation of PPTN analogues with O-β-glucuronide O Ac OAc F 3 C F 3 C AcO O O F 3 C O overnight. Scheme 25. Preparation of truncated PPTN-glucose analogue

CF 3 O CF 3 C F O 3 O CF 3 B O Ag2O, TMSOTf, DMF, rt, 5 min; (c) NaOH (3 M), MeOH, rt, overnight. EXAMPLES The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope. Pharmacological Characterization The inhibition by the compounds in accordance with aspects of the invention of the binding of fluorescent tracer 2 was measured in hP2Y14R-expressing CHO (references 1 and 2). Therefore, several of the compounds (11, 19, 29, 32) were compared in affinity at hP2Y 14 R and mP2Y 14 R (Figure 2) using the fluorescent binding procedures. 2 The solubility (using the pION method) 2,25,26 and lipophilicity (based on the HPLC retention time) 2,27 were determined for the 5-(hydroxymethyl)isoxazol-3-yl compounds 29 and 35. Absorption, distribution, metabolism, excretion and toxicology (ADMET) properties were determined for compound 32 (Tables 4 and 5), by the same methods as in Jung et al. 2 The potent 5-(hydroxymethyl)isoxazol-3-yl derivative 32 was first tested in a well- established mouse model of neuropathic pain caused by chronic constriction of the sciatic nerve (CCI) 32 at a dose of 10 µmol/kg (4.9 mg/kg, i.p., Fig. 3). Previously, it was demonstrated that 1a, 1b, 3a and other P2Y 14 R antagonists of this series were effective in reducing chronic pain in this model and reached full reversal in some cases (Table S3, Supporting information). 2,7 A maximal 71.0±17.4% reversal of CCI-induced mechano-allodynia was observed 1 h post-injection of 32, and the degree of protection declined during the following 2 h and was not statistically significant at 3 or 5 h. No side effects were evident at this dose. Selected compounds were examined for their ability to reduce eosinophils in the bronchoalveolar lavage fluid (BALF) in an ovalbumin/Aspergillus mouse asthma model. 2,17,33 Compounds 1a, 1b, 3a and 32 (10 mg/kg) were administered prior to allergen challenge at a dose of 20 µmol/kg, i.p. Airway inflammation was determined two days post-challenge (Fig. 4). Also, the corresponding prodrug derivatives 37a–37c of triazole 3a, 5-(hydroxymethyl)isoxazole 32 and N-acetyl-piperidine 1b derivatives, respectively, were administered at the same dose. Eosinophil counts in the BALF were reduced following administration of reference antagonist 1a or several of the prodrugs. In fact, 37b and 37c significantly reduced eosinophils to a greater extent than the parent antagonists, which showed no significant reduction. Other immune cells showed no significant effect, except the lymphocyte count following administration of prodrug 37a trended higher than in vehicle control. EXAMPLE 1 This example demonstrates synthesis of compounds, in accordance with aspects of the invention. General procedures Deprotection reactions: Method A: A mixture of compound (1 eq) and potassium hydroxide (5 eq) in methanol:water (2:1) was stirred at 50 o C. This mixture was neutralized with 1N HCl until pH was 5-6. The slightly acidic mixture was evaporated under reduced pressure and purified by silica gel column chromatography (dichloromethane:methanol:acetic acid=95:5:0.1) or semipreparative HPLC (10 mM triethylammonium acetate buffer:acetonitrile=80:20 to 20:80 in 40 min) to afford the compound as a white solid. Method B: A solution of compound in trifluoroacetic acid:tetrahydrofuran (2:1) was stirred at room temperature. The solvent was evaporated with toluene under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane:methanol=95:5) or semipreparative HPLC (10 mM triethylammonium acetate buffer:acetonitrile=80:20 to 20:80 in 40 min) to afford the compound as a white solid. General procedure: Suzuki reaction Suzuki coupling: Method C: A mixture of compound 38 (1 eq), Pd(PPh 3 ) 4 (0.06 eq) and potassium carbonate (3 eq) in N,N-dimethylformamide was purged with nitrogen gas for 15 min, and then various aryl halides (1.2 eq) was added to the mixture. The mixture was stirred at 85 o C for 5 h, and then allowed to be cooled at room temperature. This mixture was partitioned ethyl acetate (5 mL) and water (10 mL). The aqueous layer was extracted with ethyl acetate (5 mL x 2), and the combined organic layer was washed with brine (3 mL), dried (MgSO4 or Na2SO4), filtered and evaporated under reduced pressure. The residue was purified by silica gel column chromatography to afford the compound as a white solid. Method D: The mixture of compound 38 (1 eq) and aryl halide (1.2 eq) in dimethoxyethane/2M Na2CO3 aqueous solution (10:1) was purged with nitrogen gas for 30 min, and then PdCl 2 (dppf) (0.1 eq) was added to the mixture. The mixture was stirred at 60 °C for 4 h. After cooling at room temperature, this mixture was partitioned ethyl acetate (5 mL) and water (10 mL). The aqueous layer was extracted with ethyl acetate (5 mL x 2), and then the combined organic layer was washed with brine (3 mL), dried (MgSO 4 or Na 2 SO 4 ), filtered and evaporated under reduced pressure. The residue was purified by silica gel column chromatography to afford the compound as a white solid. Method E: A mixture of compound 38 (1 eq) and aryl bromide (1.2 eq) in DMF/H2O (10:1; 20 mM) was purged with nitrogen gas for 30 min, and then Pd(PPh 3 ) 4 (0.05 eq) and Na2CO3 (3 eq) were added to the mixture, which was stirred for 1 h at 40−120 °C. The aqueous layer was extracted with ethyl acetate twice, and the combined organic layer was washed with brine, dried (MgSO 4 or Na 2 SO 4 ), filtered, and evaporated under reduced pressure. The residue was purified by silica gel column chromatography to afford the compound as white solids. Method F: The mixture of compound 38 (1 eq), Pd(PPh3)4 (0.06 eq), potassium carbonate (3 eq) and various aryl halides (1.2 eq) in N,N-dimethylformamide was purged with nitrogen gas for 15 min. The mixture was stirred at 100 o C for 30–90 min in microwave. After microwave irradiation, and then allowed to be cooled at room temperature. This mixture was partitioned ethyl acetate (5 mL) and water (10 mL). The aqueous layer was extracted with ethyl acetate (5 mL x 2), and the combined organic layer was washed with brine (3 mL), dried (Na2SO4), filtered, and evaporated under reduced pressure. The residue was purified by silica gel column chromatography to afford the compound as a white solid. 4-(4-(1-Formylpiperidin-4-yl)phenyl)-7-(4-(trifluoromethyl)p henyl)-2-naphthoic acid (1c). To a solution of acetic formic anhydride (1 mL from Ac2O and formic acid = 2:1, v/v) was slowly added compound 1a (10 mg, 0.019 mmol) at 0 o C. After complete addition, cooling was continued for 3 h, and the mixture was then stirred at rt for 12 h. The reaction mixture was evaporated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane:methanol = 50:1) to afford compound 1c (7.0 mg, 71%) as a white solid; HPLC purity 98% (R t = 14.62 min); 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.73 (s, 1H), 8.64 (s, 1H), 8.08-7.96 (m, 5H), 7.90-7.88 (m, 3H), 7.50-7.44 (m, 4H), 4.37-4.34 (m, 1H), 3.85- 3.81 (m, 1H), 3.22-3.16 (m, 1H), 2.97-2.90 (m, 1H), 2.77-2.70 (m, 1H), 1.96-1.89 (m, 2H), 1.68- 1.47 (m, 2H); MS (ESI, m/z) 504.2 [M+1] + ; ESI-HRMS calcd. m/z for C 30 H 25 NO 3 F 3 503.1787, found 504.1792 [M+1] + . 4-(4-(1-(2,2,2-trifluoroacetyl)piperidin-4-yl)phenyl)-7-(4-( trifluoromethyl)phenyl)-2- naphthoic acid (1d). To trifluoroacetic anhydride (100 µL) was slowly added compound 1a (10 mg, 0.019 mmol) at 0 o C. After complete addition, cooling was continued for 1 h, and the mixture was then stirred at rt for 2 h. The reaction mixture was evaporated under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane:methanol = 50:1) to afford compound 1d (10.0 mg, 92%) as a white solid; HPLC purity 99% (Rt = 16.92 min); 1 H NMR (400 MHz, , CDCl 3 ) δ 8.80 (s, 1H), 8.27 (s, 1H), 8.12-8.07 (m, 2H), 7.86-7.77 (m, 5H), 7.53 (d, J = 8.00 Hz, 2H), 7.39 (d, J = 8.00 Hz, 2H), 4.81-4.78 (m, 1H), 4.25-4.22 (m, 1H), 3.37-3.30 (m, 1H), 3.01-2.92 (m, 2H), 2.15-2.11 (m, 2H), 1.91-1.81 (m, 2H); MS (ESI, m/z) 570.1 [M-1]-, ESI-HRMS calcd. m/z for C 31 H 22 NO 3 F 6 570.1504, found 570.1493 [M-1]-. (4'-(Piperidin-4-yl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2 ,3-triazol-1-yl)-[1,1'- biphenyl]-3-yl)methyl dihydrogen phosphate (3b). Method B: Yield 69%; HPLC purity 96% (R t = 8.58 min); 1 H NMR (400 MHz, DMSO-d6) δ 9.20-9.19 (m, 1H), 8.05-8.03 (m, 2H), 7.96 (s, 1H), 7.85 (s, 1H), 7.79 (d, J = 8.00 Hz, 2H), 7.74 (s, 1H), 7.67 (d, J = 8.40 Hz, 2H), 7.34 (d, J = 7.60 Hz, 2H), 4.82 - 4.80 (m, 2H), 2.96 −2.92 (m, 2H), 2.66 − 2.58 (m, 3H), 1.71−1.67 (m, 2H), 1.56-1.49 (m, 2H); MS (ESI, m/z) 559.2 [M+1]+; ESI-HRMS calcd. m/z for C27H27N4O4F3P 559.1722, found 559.1716 [M+1]+. 4'-Amino-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-1 -yl)-[1,1'-biphenyl]-3- carboxylic acid (7). Method A: Yield 62%; HPLC purity 97% (Rt = 8.96 min); 1 H NMR (400 MHz, CD3OD) δ 9.25 (s, 1H), 8.48 (s, 1H), 8.37 (s, 2H), 8.16 (d, J = 8.00 Hz, 2H), 7.79 (d, J = 8.00 Hz, 2H), 7.73 (d, J = 8.40 Hz, 2H), 7.62 (d, J = 8.40 Hz, 2H); MS (ESI, m/z) 425.1 [M+1] + ; ESI-HRMS calcd. m/z for C 22 H 16 N 4 O 2 F 3 425.1225, found 425.1221 [M+1] + . 4'-Acetamido-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triaz ol-1-yl)-[1,1'-biphenyl]- 3-carboxylic acid (8). Method A: Yield 76%; HPLC purity 99% (R t = 7.95 min); 1 H NMR (400 MHz, CD3OD) δ 9.21 (s, 1H), 8.46 (s, 1H), 8.34 (d, J =7.20 Hz, 2H), 8.13 (d, J =8.00 Hz, 2H), 7.76 (d, J =8.40 Hz, 2H), 7.72 (s, 4H), 2.15 (s, 3H); MS (ESI, m/z) 467.1 [M+1] + ; ESI-HRMS calcd. m/z for C 24 H 18 N 4 O 3 F 3 467.1331, found 467.1330 [M+1] + . 4'-Benzamido-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triaz ol-1-yl)-[1,1'- biphenyl]-3-carboxylic acid (9). Method A: Yield 73%; HPLC purity 99% (R t = 9.48 min); 1 H NMR (400 MHz, DMSO-d6) δ 9.70 (s, 1H), 8.41 (s, 1H), 8.29 (broad s, 2H), 8.21 (d, J = 7.60 Hz, 2H), 8.00-7.97 (m, 4H), 7.90-7.83 (m, 4H), 7.62-7.54 (m, 3H); MS (ESI, m/z) 529.1 [M+1] + ; ESI-HRMS calcd. m/z for C 29 H 20 N 4 O 3 F 3 529.1488, found 529.1489 [M+1] + . 4'-((tert-Butoxycarbonyl)amino)-5-(4-(4-(trifluoromethyl)phe nyl)-1H-1,2,3-triazol-1- yl)-[1,1'-biphenyl]-3-carboxylic acid (10). Method A: Yield 50%; HPLC purity 99% (R t = 10.36 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.25 (s, 1H), 8.48 (s, 1H), 8.37-8.35 (m, 2H), 8.16 (d, J =8.40 Hz, 2H), 7.78 (d, J =8.00 Hz, 2H), 7.71 (d, J =8.40 Hz, 2H), 7.58 (d, J = 8.40 Hz, 2H), 1.54 (s, 9H); MS (ESI, m/z) 525.2 [M+1] + ; ESI-HRMS calcd. m/z for C 27 H 24 N 4 O 4 F 3 525.1750, found 525.1755 [M+1] + . 4'-(Piperidine-4-carboxamido)-5-(4-(4-(trifluoromethyl)pheny l)-1H-1,2,3-triazol-1- yl)-[1,1'-biphenyl]-3-carboxylic acid (11). Method B: Yield 87%; HPLC purity 99% (R t = 21.20 min); 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.03-8.01 (m, 4H), 7.83 (s, 1H), 7.68 (d, J =8.40 Hz, 2H), 7.30-7.25 (m, 5H), 2.70- 2.68 (m, 2H), 2.23-2.20 (m, 2H), 2.00-1.93 (m, 1H), 1.44-1.27 (m, 4H); MS (ESI, m/z) 536.2 [M+Na] + ; ESI-HRMS calcd. m/z for C28H25N5O3F3536.1909, found 536.1913 [M+1] + . 4'-(1-(tert-Butoxycarbonyl)piperidine-4-carboxamido)-5-(4-(4 - (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1'-biphen yl]-3-carboxylic acid (12). Method A: Yield 82%; HPLC purity 98% (Rt = 24.10 min); 1 H NMR (400 MHz, DMSO-d 6 ) δ 9.73 (s, 1H), 8.43 (s, 1H), 8.38 (s, 1H), 8.27 (s, 1H), 8.20 (d, J =8.00 Hz, 2H), 7.90 (d, J =8.40 Hz, 2H), 7.91-7.77 (m, 4H), 4.05-3.97 (m, 2H), 2.92-2.75 (m, 3H), 1.85-1.76 (m, 2H), 1.56-1.45 (m, 2H), 1.41 (s, 9H); MS (ESI, m/z) 658.2 [M+Na] + ; ESI-HRMS calcd. m/z for C 33 H 32 N 5 O 5 F 3 Na 658.2253, found 658.2256 [M+Na] + . 5-(4-(4-(Trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1 '-biphenyl]-3,4'- dicarboxylic acid (13). Method A: Yield 68%; HPLC purity 99% (R t = 13.46 min); 1 H NMR (400 MHz, DMSO-d 6 ) δ 9.77 (s, 1H), 8.56-8.53 (m, 2H), 8.36 (broad s, 1H), 8.20 (d, J =8.00 Hz, 2H), 8.11 (d, J =8.40 Hz, 2H), 8.00 (d, J =8.40 Hz, 2H), 7.91 (d, J =8.40 Hz, 2H); MS (ESI, m/z) 454.1 [M+1] + ; ESI-HRMS calcd. m/z for C23H15N3O4F3454.1015, found 454.1017 [M+1] + . 4'-((3-Aminopropyl)carbamoyl)-5-(4-(4-(trifluoromethyl)pheny l)-1H-1,2,3-triazol-1- yl)-[1,1'-biphenyl]-3-carboxylic acid (14). Method B: Yield 63%; HPLC purity 97% (Rt = 12.68 min); 1 H NMR (400 MHz, DMSO-d 6 ) δ 9.73 (s, 1H), 8.55 (s, 1H), 8.51 (s, 1H), 8.37 (s, 1H), 8.23-8.20 (m, 4H), 8.00 (d, J =8.40 Hz, 2H), 7.90 (d, J =8.00 Hz, 2H), 3.51-3.46 (m, 2H), 3.00-2.99 (m, 2H), 1.93-1.90 (m, 2H); MS (ESI, m/z) 510.2 [M+1] + ; ESI-HRMS calcd. m/z for C26H23N5O3F3510.1753, found 510.1754 [M+1] + . 4'-Bromo-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-1 -yl)-[1,1'-biphenyl]-3- carboxylic acid (15). Method A: Yield 77%; HPLC purity 99% (R t = 12.93 min); 1 H NMR (400 MHz, CD3OD) δ 9.17 (s, 1H), 8.43 (s, 1H), 8.35 (s, 1H), 8.25 (s, 1H), 8.16 (d, J =8.00 Hz, 2H), 7.79 (d, J =8.00 Hz, 2H), 7.72 (d, J =8.00 Hz, 2H), 7.66 (d, J =8.40 Hz, 2H); MS (ESI, m/z) 488.0, 490.0 [M+1] + ; ESI-HRMS calcd. m/z for C 22 H 14 N 3 O 2 F 3 Br 488.0221, found 488.0218 [M+1] + . 4'-(2-Amino-2-oxoethyl)-5-(4-(4-(trifluoromethyl)phenyl)-1H- 1,2,3-triazol-1-yl)- [1,1'-biphenyl]-3-carboxylic acid (16). Method A: Yield 64%; HPLC purity 99% (R t = 8.55 min); 1 H NMR (400 MHz, CD3OD) δ 9.24 (s, 1H), 8.50 (s, 1H), 8.38 (s, 2H), 8.16 (d, J =8.00 Hz, 2H), 7.79-7.74 (m, 4H), 7.48 (d, J =7.60 Hz, 2H), 3.60 (s, 2H); MS (ESI, m/z) 467.1 [M+1] + ; ESI-HRMS calcd. m/z for C 24 H 18 N 4 O 3 F 3 467.1331, found 467.1332 [M+1] + . 4'-(2-Cyanoethyl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3- triazol-1-yl)-[1,1'- biphenyl]-3-carboxylic acid (17): The mixture of compound 38 (30 mg, 0.063 mmol), Pd(PPh 3 ) 4 (4 mg, 0.003 mmol), and 2 M K 2 CO 3 aqueous solution (90 µL, 0.180 mmol) in N,N-dimethylformamide:water (1:1, 3mL) was purged with nitrogen gas for 15 min, and then 3-(4-bromophenyl)propionitrile (16 mg, 0.076 mmol) was added to the mixture. The mixture was stirred at 90 o C for 12 h, and then allowed to be cooled at room temperature. This mixture was partitioned ethyl acetate (5 mL) and water (10 mL). The aqueous layer was extracted with ethyl acetate (5 mL x 2), and then the combined organic layer was washed with brine (3 mL), dried (MgSO4), filtered and evaporated under reduced pressure. The residue was purified by silica gel column chromatography (chloroform:methanol = 10:1) to afford the compound 17 (18 mg, 61%) as a white solid; HPLC purity 99% (Rt = 7.63 min); 1 H NMR (400 MHz, CD3OD) δ 9.27 (s, 1H), 8.53 (s, 1H), 8.40 (m, 2H), 8.17 (d, J =8.00 Hz, 2H), 7.80-7.77 (m, 4H), 7.48 (d, J =8.00 Hz, 2H), 3.03 (t, J =7.20 Hz, 2H), 2.81 (t, J =7.20 Hz, 2H); MS (ESI, m/z) 463.1 [M+1] + ; ESI-HRMS calcd. m/z for C 25 H 18 N 4 O 2 F 3 463.1382, found 467.1381 [M+1] + . 4'-(3-Aminopropyl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3 -triazol-1-yl)-[1,1'- biphenyl]-3-carboxylic acid (18). Method B: Yield 98%; HPLC purity 98% (Rt = 8.14 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.27 (s, 1H), 8.52 (s, 1H), 8.42 (s, 1H), 8.38 (s, 1H), 8.17 (d, J =7.60 Hz, 2H), 7.80- 7.75 (m, 4H), 7.43 (d, J =8.00 Hz, 2H), 2.99 (t, J =7.60 Hz, 2H), 2.82 (t, J =7.60 Hz, 2H), 2.05- 2.02 (m, 2H); MS (ESI, m/z) 467.2 [M+1] + ; ESI-HRMS calcd. m/z for C25H22N4O2F3467.1695, found 467.1689 [M+1] + . 4'-(3-Aminoprop-1-yn-1-yl)-5-(4-(4-(trifluoromethyl)phenyl)- 1H-1,2,3-triazol-1-yl)- [1,1'-biphenyl]-3-carboxylic acid (19). Method B: Yield 62%; HPLC purity 97% (Rt = 10.49 min); 1 H NMR (400 MHz, DMSO-d 6 ) δ 9.67 (s, 1H), 8.42 (s, 1H), 8.26 (s, 1H), 8.21 (d, J =8.00 Hz, 2H), 8.16 (s, 1H), 7.88 (d, J =8.40 Hz, 2H), 7.79 (d, J =8.00 Hz, 2H), 7.53 (d, J =8.00 Hz, 2H), 3.53 (s, 2H); MS (ESI, m/z) 463.1 [M+1] + ; ESI-HRMS calcd. m/z for C25H18N4O2F3463.1382, found 463.1380 [M+1] + . 4'-(3-((tert-Butoxycarbonyl)amino)propyl)-5-(4-(4-(trifluoro methyl)phenyl)-1H- 1,2,3-triazol-1-yl)-[1,1'-biphenyl]-3-carboxylic acid (20). Method A: Yield 66%; HPLC purity 99% (Rt = 10.56 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.24 (s, 1H), 8.49 (s, 1H), 8.38-8.36 (m, 2H), 8.16 (d, J =8.00 Hz, 2H), 7.78 (d, J =8.00 Hz, 2H), 7.70 (d, J =8.40 Hz, 2H), 7.37 (d, J =8.00 Hz, 2H), 3.10 (t, J =7.20 Hz, 2H), 2.71 (t, J =7.20 Hz, 2H), 1.87-1.78 (m, 2H), 1.45 (s, 9H); MS (ESI, m/z) 589.2 [M+Na] + ; ESI- HRMS calcd. m/z for C 30 H 29 N 4 O 4 F 3 Na 589.2039, found 589.2043 [M+1] + . 4'-(4-Hydroxybutyl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2, 3-triazol-1-yl)-[1,1'- biphenyl]-3-carboxylic acid (21). Method A: Yield 39%; HPLC purity 99% (Rt = 7.40 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.25 (s, 1H), 8.50 (s, 1H), 8.38 (s, 2H), 8.16 (d, J =8.00 Hz, 2H), 7.79 (d, J =8.40 Hz, 2H), 7.70 (d, J =8.00 Hz, 2H), 7.37 (d, J =8.00 Hz, 2H), 3.59 (t, J =6.40 Hz, 2H), 2.72 (t, J =7.60 Hz, 2H), 1.78-1.71 (m, 2H), 1.64-1.57 (m, 2H); MS (ESI, m/z) 482.2 [M+1] + ; ESI-HRMS calcd. m/z for C 26 H 23 N 3 O 3 F 3 482.1692, found 482.1694 [M+1] + . 4'-(1-Aminocyclopropyl)-5-(4-(4-(trifluoromethyl)phenyl)-1H- 1,2,3-triazol-1-yl)- [1,1'-biphenyl]-3-carboxylic acid (22). Method A: Yield 69%; HPLC purity 98% (R t = 6.80 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.27 (s, 1H), 8.55-8.39 (m, 3H), 8.15 (broad s, 2H), 7.88 (broad s, 2H), 7.78 (broad s, 2H), 7.63 (broad s, 2H), 1.43-1.38 (m, 4H); MS (ESI, m/z) 465.2 [M+1] + ; ESI-HRMS calcd. m/z for C25H20N4O2F3 465.1538, found 465.1539 [M+1] + . 4'-(1-(Aminomethyl)cyclopropyl)-5-(4-(4-(trifluoromethyl)phe nyl)-1H-1,2,3-triazol- 1-yl)-[1,1'-biphenyl]-3-carboxylic acid (23). Method B: Yield 52%; HPLC purity 97% (R t = 10.85 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.08 (s, 1H), 8.38 (s, 1H), 8.30 (s, 1H), 8.16 (s, 1H), 8.09 (d, J =8.00 Hz, 2H), 7.77- 7.73 (m, 4H), 7.55 (d, J =8.00 Hz, 2H), 3.22 (s, 2H), 1.09-1.06 (m, 4H); MS (ESI, m/z) 479.2 [M+1] + ; ESI-HRMS calcd. m/z for C26H22N4O2F3479.1695, found 479.1693 [M+1] + . 4'-(1-(((tert-Butoxycarbonyl)amino)methyl)cyclopropyl)-5-(4- (4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1'-biphen yl]-3-carboxylic acid (24). Method A: Yield 51%; HPLC purity 99% (Rt = 21.57 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.26 (s, 1H), 8.51 (s, 1H), 8.40-8.38 (m, 2H), 8.17 (d, J =8.00 Hz, 2H), 7.80 (d, J =8.00 Hz, 2H), 7.73 (d, J =8.00 Hz, 2H), 7.51 (d, J =8.00 Hz, 2H), 3.34 (s, 2H), 1.39-1.28 (m, 9H), 0.94-0.86 (m, 4H); MS (ESI, m/z) 601.2 [M+Na] + ; ESI-HRMS calcd. m/z for C 31 H 29 N 4 O 4 F 3 Na 601.2039, found 601.2040 [M+Na] + . 4'-(1-Aminocyclobutyl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1 ,2,3-triazol-1-yl)-[1,1'- biphenyl]-3-carboxylic acid (25). Method B: Yield 65%; HPLC purity 99% (R t = 8.69 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.27 (s, 1H), 8.55 (s, 1H), 8.45 (s, 1H), 8.41 (s, 1H), 8.16 (d, J =8.00 Hz, 2H), 7.93 (d, J =8.40 Hz, 2H), 7.79 (d, J = 8.40 Hz, 2H), 7.69 (d, J =8.40 Hz, 2H), 2.89-2.82 (m, 2H), 2.70-2.63 (m, 2H), 2.33-2.24 (m, 1H), 2.07-1.97 (m, 1H); MS (ESI, m/z) 479.3 [M+1] + ; ESI- HRMS calcd. m/z for C 26 H 22 N 4 O 2 F 3 479.1695, found 479.1696 [M+1] + . 4'-(1-((tert-Butoxycarbonyl)amino)cyclobutyl)-5-(4-(4-(trifl uoromethyl)phenyl)-1H- 1,2,3-triazol-1-yl)-[1,1'-biphenyl]-3-carboxylic acid (26). Method A: Yield 47%; HPLC purity 99% (R t = 10.76 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.18 (s, 1H), 8.40-8.39 (m, 2H), 8.26 (s, 1H), 8.17 (d, J =8.00 Hz, 2H), 7.80-7.76 (m, 4H), 7.59 (d, J = 8.40 Hz, 2H), 2.60-2.46 (m, 4H), 2.16-2.07 (m, 1H), 1.99-1.88 (m, 1H), 1.40- 1.25 (m, 9H); MS (ESI, m/z) 579.2 [M+1] + ; ESI-HRMS calcd. m/z for C 31 H 30 N 4 O 4 F 3 579.2219, found 579.2219 [M+1] + . 4'-(3-(Hydroxymethyl)oxetan-3-yl)-5-(4-(4-(trifluoromethyl)p henyl)-1H-1,2,3- triazol-1-yl)-[1,1'-biphenyl]-3-carboxylic acid (27). Method A: Yield 47%; HPLC purity 99% (R t = 8.71 min); 1 H NMR (400 MHz, CD3OD) δ 9.25 (s, 1H), 8.51 (s, 1H), 8.41 (s, 1H), 8.39 (s, 1H), 8.15 (d, J =8.40 Hz, 2H), 7.80- 7.77 (m, 4H), 7.32 (d, J =8.00 Hz, 2H), 4.97 (d, J = 5.60 Hz, 2H), 4.89 (d, J = 6.00 Hz, 2H), 3.94 (s, 2H); MS (ESI, m/z) 496.2 [M+1] + ; ESI-HRMS calcd. m/z for C 26 H 21 N 3 O 4 F 3 496.1484, found 496.1488 [M+1] + . 4'-(Isoxazol-3-yl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3 -triazol-1-yl)-[1,1'- biphenyl]-3-carboxylic acid (28). Method A: Yield 74%; HPLC purity 98% (Rt = 11.86 min); 1 H NMR (400 MHz, CD3OD) δ 9.28 (s, 1H), 8.75 (s, 1H), 8.57 (s, 1H), 8.46 (s, 2H), 8.17 (d, J = 7.60 Hz, 2H), 8.05 (d, J =8.00 Hz, 2H), 7.94 (d, J = 8.40 Hz, 2H), 7.79 (d, J =8.00 Hz, 2H), 7.00 (s, 1H); MS (ESI, m/z) 477.1 [M+1] + ; ESI-HRMS calcd. m/z for C 25 H 16 N 4 O 3 F 3 477.1175, found 477.1177 [M+1] + . 4'-(5-(Hydroxymethyl)isoxazol-3-yl)-5-(4-(4-(trifluoromethyl )phenyl)-1H-1,2,3- triazol-1-yl)-[1,1'-biphenyl]-3-carboxylic acid (29). Method A: Yield 43%; HPLC purity 98% (R t = 20.10 min); 1 H NMR (400 MHz, DMSO-d6) δ 9.77 (s, 1H), 8.57 (s, 1H), 8.54 (s, 1H), 8.36 (s, 1H), 8.20 (d, J =8.00 Hz, 2H), 8.08- 8.01 (m, 4H), 7.91 (d, J = 8.00 Hz, 2H), 7.05 (s, 1H), 5.75 (t, J = 6.00 Hz, 1H), 4.64 (d, J = 6.00 Hz, 2H); MS (ESI, m/z) 507.1 [M+1] + ; ESI-HRMS calcd. m/z for C 26 H 18 N 4 O 4 F 3 507.1280, found 507.1271 [M+1] + . 4'-(5-(2-Hydroxyethyl)isoxazol-3-yl)-5-(4-(4-(trifluoromethy l)phenyl)-1H-1,2,3- triazol-1-yl)-[1,1'-biphenyl]-3-carboxylic acid (30). Method A: Yield 55%; HPLC purity 95% (Rt = 10.69 min); 1 H NMR (400 MHz, DMSO-d6) δ 9.74 (s, 1H), 8.51 (s, 1H), 8.46 (s, 1H), 8.35 (s, 1H), 8.20 (d, J =8.00 Hz, 2H), 8.03- 7.98 (m, 4H), 7.90 (d, J = 8.00 Hz, 2H), 6.93 (s, 1H), 3.77 (t, J = 6.40 Hz, 2H), 2.97 (d, J =6.40 Hz, 2H); MS (ESI, m/z) 521.1 [M+1] + ; ESI-HRMS calcd. m/z for C 27 H 20 N 4 O 4 F 3 521.1437, found 521.1435 [M+1] + . 4'-(2H-Tetrazol-5-yl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1, 2,3-triazol-1-yl)-[1,1'- biphenyl]-3-carboxylic acid (31). Method A: Yield 43%; HPLC purity 98% (Rt = 15.95 min); 1 H NMR (400 MHz, CD 3 OD) δ 9.24 (s, 1H), 8.49 (s, 1H), 8.45 (s, 1H), 8.35 (s, 1H), 8.22-8.17 (m, 4H), 7.92 (d, J =8.40 Hz, 2H), 7.80 (d, J = 8.00 Hz, 2H); MS (ESI, m/z) 478.1 [M+1] + ; ESI-HRMS calcd. m/z for C23H15N7O2F3478.1239, found 478.1242 [M+1] + . 4-(4-(5-(Hydroxymethyl)isoxazol-3-yl)phenyl)-7-(4-(trifluoro methyl)phenyl)-2- naphthoic acid (32). Method A: Yield 93%; HPLC purity 98% (Rt = 12.07 min); 1 H NMR (400 MHz, DMSO-d6) δ 8.79 (s, 1H), 8.68 (s, 1H), 8.09-8.05 (m, 5H), 8.01-7.98 (m, 1H), 7.95 (m, 1H), 7.91-7.89 (m, 2H), 7.70 (d, J = 8.40 Hz, 2H), 7.04 (s, 1H), 5.80-5.73 (m, 1H), 4.66-4.63 (m, 2H); MS (ESI, m/z) 490.1 [M+1] + ; ESI-HRMS calcd. m/z for C28H19NO4F3490.1266, found 490.1269 [M+1] + . 4'-(5-(Hydroxymethyl)isoxazol-3-yl)-5-(4-(trifluoromethyl)be nzamido)-[1,1'- biphenyl]-3-carboxylic acid (33). Method A: Yield 62%; HPLC purity 99% (Rt = 10.29 min); 1 H NMR (400 MHz, CD3OD) δ 8.35 (broad s, 2H), 8.17-8.13 (m, 3H), 7.97 (d, J =8.40 Hz, 2H), 7.86-7.81 (m, 4H), 6.82 (s, 1H), 4.74 (s, 2H); MS (ESI, m/z) 483.1 [M+1] + ; ESI-HRMS calcd. m/z for C 25 H 18 N 2 O 5 F 3 483.1168, found 483.1170 [M+1] + . 4-(4-(Piperidin-4-yl)phenyl)-6-(4-(4-(trifluoromethyl)phenyl )-1H-1,2,3-triazol-1- yl)picolinic acid (34). Method B: Yield 60%; HPLC purity 99% (Rt = 7.87 min); 1 H NMR (400 MHz, DMSO-d6) δ 9.55 (s, 1H), 8.31-8.26 (m, 4H), 7.91-7.85 (m, 4H), 7.47 (d, J =8.00 Hz, 2H), 3.68- 3.65 (m, 1H), 3.02-2.88 (m, 4H), 1.95-1.83 (m, 4H); MS (ESI, m/z) 494.2 [M+1] + ; ESI-HRMS calcd. m/z for C26H23N5O2F3494.1804, found 494.1805 [M+1] + . 2-(4-(Piperidin-4-yl)phenyl)-6-(4-(4-(trifluoromethyl)phenyl )-1H-1,2,3-triazol-1- yl)isonicotinic acid (35). Method B: Yield 65%; HPLC purity 95% (R t = 10.72 min); 1 H NMR (400 MHz, DMSO-d6) δ 9.79 (s, 1H), 8.42 (s, 2H), 8.34-8.33 (m, 4H), 7.89 (d, J =8.00 Hz, 2H), 7.44 (d, J =8.00 Hz, 2H), 3.62-3.59 (m, 1H), 3.07-2.92 (m, 4H), 2.03-1.89 (m, 4H); MS (ESI, m/z) 494.2 [M+1] + ; ESI-HRMS calcd. m/z for C 26 H 23 N 5 O 2 F 3 494.1804, found 494.1805 [M+1] + . 6-(4-(Piperidin-4-yl)phenyl)-4-(4-(4-(trifluoromethyl)phenyl )-1H-1,2,3-triazol-1- yl)picolinic acid (36). Method B: Yield 65%; HPLC purity 99% (R t = 11.24 min); 1 H NMR (400 MHz, DMSO-d6) δ 9.94 (s, 1H), 8.71 (s, 1H), 8.56 (s, 1H), 8.28 (d, J =7.20 Hz, 2H), 8.19 (d, J =7.60 Hz, 2H), 7.93 (d, J =7.20 Hz, 2H), 7.46 (d, J =7.20 Hz, 2H), 3.43-3.40 (m, 2H), 3.07-2.92 (m, 3H), 2.03-2.00 (m, 2H), 1.91-1.84 (m, 2H); MS (ESI, m/z) 494.2 [M+1] + ; ESI-HRMS calcd. m/z for C26H23N5O2F3494.1804, found 494.1808 [M+1] + . 2-(Dimethylamino)-2-oxoethyl 4'-(piperidin-4-yl)-5-(4-(4-(trifluoromethyl)phenyl)- 1H-1,2,3-triazol-1-yl)-[1,1'-biphenyl]-3-carboxylate (37a). Method B: Yield 78%; HPLC purity 97% (Rt = 20.45 min); 1 H NMR (400 MHz, CD3OD) δ 9.29 (s, 1H), 8.59 (s, 1H), 8.47 (s, 1H), 8.45 (s, 1H), 8.17 (d, J =8.00 Hz, 2H), 7.82- 7.79 (m, 4H), 7.47 (d, J =8.00 Hz, 2H), 5.18 (s, 2H), 3.56-3.53 (m, 2H), 3.23-3.16 (s, 2H), 3.13 (s, 3H), 3.05-2.99 (m, 1H), 3.02 (s, 3H), 2.18-2.14 (m, 2H), 2.03-1.92 (m, 2H); MS (ESI, m/z) 578.3 [M+1] + ; ESI-HRMS calcd. m/z for C31H31N5O3F3578.2379, found 578.2390 [M+1] + . 2-(Dimethylamino)-2-oxoethyl 4-(4-(5-(hydroxymethyl)isoxazol-3-yl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (37b). To a solution of 32 (3.5 mg, 0.007 mmol) in DMF (1 mL) was added Cs2CO3 (3.0 mg, 0.010 mmol). 2-Chloro-N,N-dimethylacetamide (1.0 mg, 0.007 mmol) was added to the reaction mixture under N 2 atmosphere, and the reaction was stirred at 45 °C for 1 h under N 2 atmosphere. The reaction mixture was partitioned ethyl acetate (5 mL) and water (5 mL), and the aqueous phase was extracted with ethyl acetate (2 x 5 mL). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under pressure. The residue was purified by silica gel chromatography (chloroform:methanol = 30:1) to afford compound 37b (2.7 mg, 68%) as a white solid; HPLC purity 99% (Rt = 17.73 min); 1 H NMR (400 MHz, CD3OD) δ 8.85-8.84 (m, 1H), 8.45-8.44 (m, 1H), 8.08-7.97 (m, 7H), 7.81 (d, J =8.40 Hz, 2H), 7.66 (d, J =8.00 Hz, 2H), 6.88 (s, 1H), 5.14 (s, 2H), 4.76 (s, 2H), 3.12 (s, 3H), 3.00 (s, 3H); MS (ESI, m/z) 575.2 [M+1] + ; ESI-HRMS calcd. m/z for C32H26N2O5F3575.1794, found 575.1786 [M+1] + . 2-(Dimethylamino)-2-oxoethyl 4-(4-(1-acetylpiperidin-4-yl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (37c). Compound 1b (24 mg, 0.046 mmol) was converted to compound 37c (20 mg, 72%) as a white solid, using a similar procedure to that used in the preparation of compound 87; HPLC purity 99% (R t = 16.43 min); 1 H NMR (400 MHz, CD 3 OD) δ 8.80 (s, 1H), 8.42 (s, 1H), 8.03- 7.98 (m, 4H), 7.93-7.91 (m, 1H), 7.80 (d, J = 7.60 Hz, 2H), 7.47-7.43 (m, 4H), 5.14 (s, 2H), 4.75-4.72 (m, 1H), 4.12-4.08 (m, 1H), 3.27-3.24 (m, 1H), 3.12 (s, 3H), 3.00 (s, 3H), 2.95-2.92 (m, 1H), 2.81-2.75 (m, 1H), 2.18 (s, 3H), 2.04-1.97 (m, 2H), 1.84-1.68 (m, 2H); MS (ESI, m/z) 603.2 [M+1] + ; ESI-HRMS calcd. m/z for C35H34N2O4F3603.2471, found 603.2479 [M+1] + . Methyl 4'-((tert-butoxycarbonyl)amino)-5-(4-(4-(trifluoromethyl)phe nyl)-1H-1,2,3- triazol-1-yl)-[1,1'-biphenyl]-3-carboxylate (39). Method C: Yield 40%; 1 H NMR (400 MHz, CDCl3) δ 8.40 (s, 1H), 8.33 (s, 1H), 8.29 (broad s, 2H), 8.06 (d, J =8.00 Hz, 2H), 7.74 (d, J =8.40 Hz, 2H), 7.64 (d, J =8.40 Hz, 2H), 7.52 (d, J =8.40 Hz, 2H), 4.01 (s, 3H), 1.54 (s, 9H); MS (ESI, m/z) 539.2 [M+1] + ; ESI-HRMS calcd. m/z for C 28 H 26 N 4 O 4 F 3 539.1906, found 539.1916 [M+1] + . Methyl 4'-amino-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-1 -yl)-[1,1'- biphenyl]-3-carboxylate (40). Method B: Yield 57%; 1 H NMR (400 MHz, CDCl 3 ) δ 8.38 (s, 1H), 8.32 (s, 1H), 8.26- 8.24 (m, 2H), 8.06 (d, J =8.00 Hz, 2H), 7.74 (d, J =8.00 Hz, 2H), 7.54 (d, J =8.40 Hz, 2H), 6.81 (d, J =8.40 Hz, 2H), 4.00 (s, 3H); MS (ESI, m/z) 439.1 [M+1] + ; ESI-HRMS calcd. m/z for C 23 H 18 N 4 O 2 F 3 439.1382, found 439.1378 [M+1] + . Methyl 4'-acetamido-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triaz ol-1-yl)-[1,1'- biphenyl]-3-carboxylate (41). 40 (12 mg, 0.027 mmol) was dissolved in dichloromethane (1 mL) and degassed for 30 min with nitrogen gas. Acetic anhydride (3.2 μL, 0.033 mmol) was added to the reaction and was stirred for 2 h at room temperature. The reaction mixture was partitioned dichloromethane (10 mL) and saturated aqueous Na 2 CO 3 solution (10 mL), and the aqueous phase was extracted with dichloromethane (10 mL x 2). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under pressure. The residue was purified by silica gel chromatography (hexane:ethyl acetate = 2:1) to afford compound 41 (10 mg, 76%) as a white solid; 1 H NMR (400 MHz, CDCl 3 ) δ 8.40 (s, 1H), 8.34 (s, 1H), 8.32-8.30 (m, 2H), 8.06 (d, J =8.00 Hz, 2H), 7.74 (d, J =8.40 Hz, 2H), 7.67 (s, 4H), 4.01 (s, 3H), 2.23 (s, 3H); MS (ESI, m/z) 481.2 [M+1] + ; ESI- HRMS calcd. m/z for C 25 H 20 N 4 O 3 F 3 481.1488, found 481.1487 [M+1] + . Methyl 4'-benzamido-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triaz ol-1-yl)-[1,1'- biphenyl]-3-carboxylate (42). 40 (12 mg, 0.027 mmol) was dissolved in dichloromethane (1 mL) at 0 o C. Triethylamine (4.2 μL, 0.030 mmol) and benzoyl chloride (3.2 μL, 0.027 mmol) were added, and the mixture was stirred for 18 h at room temperature. The reaction mixture was partitioned dichloromethane (10 mL) and water (10 mL), and the aqueous phase was extracted with dichloromethane (10 mL x 2). The combined organic layers were dried over Na 2 SO 4 , filtered, and concentrated under pressure. The residue was purified by silica gel chromatography (hexane:ethyl acetate = 3:1) to afford compound 42 (9 mg, 63%) as a white solid; 1 H NMR (400 MHz, CDCl 3 ) δ 8.41 (s, 1H), 8.37 (s, 1H), 8.33 (broad s, 2H), 8.10-8.05 (m, 2H), 7.94-7.90 (m, 3H), 7.82 (d, J =8.40 Hz, 2H), 7.75-7.72 (m, 3H), 7.61-7.45 (m, 3H), 4.02 (s, 3H); MS (ESI, m/z) 543.2 [M+1] + ; ESI-HRMS calcd. m/z for C30H21N4O3F3543.1644, found 543.1647 [M+1] + . tert-Butyl 4-((3'-(methoxycarbonyl)-5'-(4-(4-(trifluoromethyl)phenyl)-1 H-1,2,3- triazol-1-yl)-[1,1'-biphenyl]-4-yl)carbamoyl)piperidine-1-ca rboxylate (43). To a solution of compounds 40 (27 mg, 0.061 mmol) in N,N-dimethylformamide (2 mL) were added Boc-Inp-OH (15 mg, 0.067 mmol), HATU (25 mg, 0.067 mmol) and N,N- diisopropylethylamine (13 µL, 0.080 mmol), and then this reaction mixture was stirred at room temperature for 2 h. The reaction mixture was partitioned ethyl acetate (5 mL) and water (5 mL), and the aqueous layer was extracted with ethyl acetate (5 mL x 2). The combined organic layer was washed brine (3 mL), dried over MgSO 4 , filtered and evaporated under reduced pressure. The residue was purified by silica gel column chromatography (hexane:ethyl acetate = 3:1) to afford compound 43 (37 mg, 94%) as a white solid; 1 H NMR (400 MHz, CDCl3) δ 8.40 (s, 1H), 8.34-8.33 (m, 1H), 8.32-8.30 (m, 2H), 8.06 (d, J =8.40 Hz, 2H), 7.74 (d, J =8.00 Hz, 2H), 7.68 (s, 4H), 4.25-4.18 (m, 2H), 4.01 (s, 3H), 2.84-2.78 (m, 2H), 2.46-2.40 (m, 1H), 1.95-1.92 (m, 2H), 1.78-1.76 (m, 2H), 1.47 (s, 9H); MS (ESI, m/z) 672.2 [M+ Na] + ; ESI-HRMS calcd. m/z for C 34 H 34 N 5 O 5 F 3 Na 672.2410, found 672.2413 [M+Na] + . 3'-(Methoxycarbonyl)-5'-(4-(4-(trifluoromethyl)phenyl)-1H-1, 2,3-triazol-1-yl)-[1,1'- biphenyl]-4-carboxylic acid (44). Method C: Yield 45%; 1 H NMR (400 MHz, DMSO-d 6 ) δ 9.78 (s, 1H), 8.59-8.56 (m, 2H), 8.36 (s, 1H), 8.19 (d, J =8.00 Hz, 2H), 8.10 (d, J =8.40 Hz, 2H), 8.00 (d, J =8.40 Hz, 2H), 7.91 (d, J =8.40 Hz, 2H), 3.98 (s, 3H); MS (ESI, m/z) 468.1 [M+1] + ; ESI-HRMS calcd. m/z for C 24 H 17 N 3 O 4 F 3 468.1171, found 468.1169 [M+1] + . Methyl 4'-((3-((tert-butoxycarbonyl)amino)propyl)carbamoyl)-5-(4-(4 - (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1'-biphen yl]-3-carboxylate (45). Method C: Yield 68%; 1 H NMR (400 MHz, CD 3 OD) δ 9.15 (s, 1H), 8.41 (s, 1H), 8.32 (s, 1H), 8.25 (s, 1H), 8.04 (d, J =8.00 Hz, 2H), 7.93 (d, J =8.40 Hz, 2H), 7.78 (d, J =8.40 Hz, 2H), 7.71 (d, J =8.00 Hz, 2H), 3.95 (s, 3H), 3.44 (t, J =6.80 Hz, 2H), 3.17-3.14 (m, 2H), 1.80-1.76 (m, 2H); MS (ESI, m/z) 646.2 [M+Na] + ; ESI-HRMS calcd. m/z for C 32 H 32 N 5 O 5 F 3 Na 646.2253, found 646.2255 [M+Na] + . 4'-((3-((tert-Butoxycarbonyl)amino)propyl)carbamoyl)-5-(4-(4 - (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1'-biphen yl]-3-carboxylic acid (46). Method A: Yield 77%; 1 H NMR (400 MHz, CD 3 OD) δ 9.27 (s, 1H), 8.57 (s, 1H), 8.46 (s, 1H), 8.41 (s, 1H), 8.15 (d, J =8.00 Hz, 2H), 7.99 (d, J =7.60 Hz, 2H), 7.89 (d, J =8.00 Hz, 2H), 7.78 (d, J =8.00 Hz, 2H), 3.47-3.43 (m, 2H), 3.17-3.14 (m, 2H), 1.80-1.77 (m, 2H); MS (ESI, m/z) 610.2 [M+1] + ; ESI-HRMS calcd. m/z for C 31 H 31 N 5 O 5 F 3 610.2277, found 610.2270 [M+1] + . Methyl 4'-bromo-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-1 -yl)-[1,1'- biphenyl]-3-carboxylate (47). Method C: Yield 53%; 1 H NMR (400 MHz, CDCl3) δ 8.40 (s, 1H), 8.35-8.32 (m, 3H), 8.06 (d, J =8.00 Hz, 2H), 7.74 (d, J =8.40 Hz, 2H), 7.65 (d, J =8.40 Hz, 2H), 7.57 (d, J =8.40 Hz, 2H), 4.02 (s, 3H); MS (ESI, m/z) 502.0, 504.0 [M+1] + ; ESI-HRMS calcd. m/z for C 23 H 16 N 3 O 2 F 3 Br 502.0378, found 502.0382 [M+1] + . Methyl 4'-(2-amino-2-oxoethyl)-5-(4-(4-(trifluoromethyl)phenyl)-1H- 1,2,3-triazol-1- yl)-[1,1'-biphenyl]-3-carboxylate (48). Method D: Yield 73%; 1 H NMR (400 MHz, CDCl3) δ 8.41 (s, 1H), 8.36-8.33 (m, 3H), 8.06 (d, J =8.00 Hz, 2H), 7.75-7.69 (m, 2H), 7.50-7.44 (m, 2H), 7.16 (d, J =8.40 Hz, 2H), 4.02 (s, 3H), 3.67 (s, 2H); MS (ESI, m/z) 481.2 [M+1] + ; ESI-HRMS calcd. m/z for C25H20N4O3F3481.1488, found 481.1492 [M+1] + . Methyl 4'-(3-((tert-butoxycarbonyl)amino)propyl)-5-(4-(4-(trifluoro methyl)phenyl)- 1H-1,2,3-triazol-1-yl)-[1,1'-biphenyl]-3-carboxylate (49). Method E: Yield 55%; 1 H NMR (400 MHz, CDCl 3) δ 8.40 (s, 1H), 8.35 (s, 1H), 8.33-8.31 (m, 2H), 8.06 (d, J =8.00 Hz, 2H), 7.74 (d, J =8.40 Hz, 2H), 7.62 (d, J =8.40 Hz, 2H), 7.33 (d, J =8.00 Hz, 2H), 4.01 (s, 3H), 3.20-3.18 (m, 2H), 2.72 (t, J =7.60 Hz, 2H), 1.90-1.82 (m, 2H), 1.45 (s, 9H); MS (ESI, m/z) 525.2 [M+1-tert-butyl] + . Methyl 4'-(3-((tert-butoxycarbonyl)amino)prop-1-yn-1-yl)-5-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1'-biphen yl]-3-carboxylate (50). Method C: Yield 35%; 1 H NMR (400 MHz, CD 3 OD) δ 9.24 (s, 1H), 8.51 (s, 1H), 8.40 (s, 1H), 8.34 (s, 1H), 8.13 (d, J =8.00 Hz, 2H), 7.78-7.74 (m, 4H), 7.56 (d, J =8.40 Hz, 2H), 4.10-4.08 (m, 2H), 4.00 (s, 3H), 1.48 (s, 9H); MS (ESI, m/z) 577.2 [M+1] + ; ESI-HRMS calcd. m/z for C 31 H 28 N 4 O 4 F 3 577.2063, found 577.2057 [M+1] + . Methyl 4'-(4-hydroxybutyl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2, 3-triazol-1-yl)- [1,1'-biphenyl]-3-carboxylate (51). Method E: Yield 27%; 1 H NMR (400 MHz, CDCl 3 ) δ 8.48 (s, 1H), 8.40 (s, 1H), 8.32 (s, 2H), 8.06 (d, J =8.00 Hz, 2H), 7.74 (d, J =8.00 Hz, 2H), 7.62 (d, J =8.00 Hz, 2H), 7.33 (d, J =8.00 Hz, 2H), 4.01 (s, 3H), 3.72-3.68 (m, 2H), 2.73 (t, J =7.60 Hz, 2H), 1.80-1.72 (m, 2H), 1.68-1.58 (m, 2H); MS (ESI, m/z) 496.2 [M+1] + ; ESI-HRMS calcd. m/z for C 27 H 25 N 3 O 3 F 3 496.1848, found 496.1855 [M+1] + . 4'-(3-((tert-Butoxycarbonyl)amino)prop-1-yn-1-yl)-5-(4-(4-(t rifluoromethyl)phenyl)- 1H-1,2,3-triazol-1-yl)-[1,1'-biphenyl]-3-carboxylic acid (52). Method A: Yield 88%; 1 H NMR (400 MHz, DMSO-d 6 ) δ 9.76 (s, 1H), 8.50 (d, J =6.40 Hz, 2H), 8.30 (s, 1H), 8.19 (d, J =8.00 Hz, 2H), 7.91-7.88 (m, 4H), 7.58 (d, J =8.40 Hz, 2H), 4.02 (s, 2H), 1.41 (s, 9H); MS (ESI, m/z) 563.2 [M+1] + ; ESI-HRMS calcd. m/z for C 30 H 26 N 4 O 4 F 3 563.1906, found 563.1897 [M+1] + . tert-Butyl (3-(4-bromobenzamido)propyl)carbamate (54a). 4-Bromobenzoic acid 53a (Scheme S2, Supporting information, 100 mg, 0.497 mmol) was converted to compound 54a (54 mg, 31%) as a white solid, using a similar procedure to that used in the preparation of compound 43; 1 H NMR (400 MHz, CDCl3) δ 7.73 (d, J =8.00 Hz, 2H), 7.57 (d, J =8.40 Hz, 2H), 3.51-3.46 (m, 2H), 3.26-3.22 (m, 2H), 1.71-1.68 (m, 2H), 1.44 (s, 9H). tert-Butyl (3-(4-bromophenyl)prop-2-yn-1-yl)carbamate (54b). To a mixture of 1-bromo-4-iodobenzene 53b (Scheme S2, Supporting information, 300 mg, 1.06 mmol), PdCl 2 (PPh 3 ) 2 (37 mg, 0.053 mmol) and copper iodide (20 mg, 0.106 mmol) in THF were added triethylamine (295 μL, 2.12 mmol) and N-Boc-propargylamine (247 mg, 1.59 mmol) at room temperature under N2 atmosphere, and then this reaction mixture was stirred at room temperature for 12 h under N2 atmosphere. The reaction mixture was partitioned with ethyl acetate (100 mL) and aqueous saturated NH 4 Cl solution (50 mL), and then the aqueous layer was extracted with ethyl acetate (100 mL x 2). The combined organic layer was dried (Na2SO4), filtered and evaporated under reduced pressure. The residue was purified by silica gel column chromatography (hexane:ethyl acetate = 5:1) to afford the compound 54b (310 mg, 94%) as a white solid; 1 H NMR (400 MHz, CDCl 3 ) δ 7.45 (d, J =8.40 Hz, 2H), 7.28 (d, J =8.40 Hz, 2H), 4.16-4.15 (m, 2H), 1.48 (s, 9H). Methyl 4'-(1-((tert-butoxycarbonyl)amino)cyclopropyl)-5-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1'-biphen yl]-3-carboxylate (55). Method E: Yield 30%; 1 H NMR (400 MHz, CDCl3) δ 8.39 (s, 1H), 8.35-8.30 (m, 3H), 8.06 (d, J =8.00 Hz, 2H), 7.74 (d, J =8.00 Hz, 2H), 7.63 (d, J =8.40 Hz, 2H), 7.34 (d, J =8.00 Hz, 2H), 4.01 (s, 3H), 1.46 (s, 9H), 1.35-1.30 (m, 4H); MS (ESI, m/z) 579.2 [M+1] + ; ESI- HRMS calcd. m/z for C 31 H 30 N 4 O 4 F 3 579.2219, found 579.2220 [M+1] + . Methyl 4'-(1-aminocyclopropyl)-5-(4-(4-(trifluoromethyl)phenyl)-1H- 1,2,3-triazol-1- yl)-[1,1'-biphenyl]-3-carboxylate (56). Method B: Yield 90%; 1 H NMR (400 MHz, CD 3 OD) δ 9.29 (s, 1H), 8.56 (s, 1H), 8.49 (s, 1H), 8.41 (s, 1H), 8.17 (d, J =8.00 Hz, 2H), 7.90 (d, J =8.40 Hz, 2H), 7.80 (d, J =8.00 Hz, 2H), 7.64 (d, J =8.00 Hz, 2H), 4.02 (s, 3H), 1.43-1.40 (m, 4H); MS (ESI, m/z) 479.1 [M+1] + ; ESI-HRMS calcd. m/z for C 26 H 22 N 4 O 2 F 3 479.1695, found 479.1699 [M+1] + . Methyl 4'-(1-(((tert-Butoxycarbonyl)amino)methyl)cyclopropyl)-5-(4- (4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1'-biphen yl]-3-carboxylate (57). Method E: Yield 30%; 1 H NMR (400 MHz, CDCl 3 ) δ 8.40 (s, 1H), 8.35-8.30 (m, 3H), 8.06 (d, J =8.40 Hz, 2H), 7.74 (d, J =8.40 Hz, 2H), 7.64 (d, J =8.00 Hz, 2H), 7.44 (d, J =8.00 Hz, 2H), 4.01 (s, 3H), 3.39-3.35 (m, 2H), 1.40 (s, 9H), 0.93-0.79 (m, 4H); MS (ESI, m/z) 615.2 [M+Na] + ; ESI-HRMS calcd. m/z for C32H31N4O4F3Na 615.2195, found 615.2199 [M+Na] + . Methyl 4'-(1-((tert-butoxycarbonyl)amino)cyclobutyl)-5-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1'-biphen yl]-3-carboxylate (58). Method E: Yield 70%; 1 H NMR (400 MHz, CDCl 3 ) δ 8.40 (s, 1H), 8.38 (s, 1H), 8.33 (s, 2H), 8.06 (d, J =8.00 Hz, 2H), 7.74 (d, J =8.00 Hz, 2H), 7.68 (d, J =8.00 Hz, 2H), 7.57 (d, J =7.60 Hz, 2H), 4.01 (s, 3H), 2.63-2.44 (m, 4H), 2.20-2.09 (m, 1H), 1.97-1.85 (m, 1H), 1.47-1.29 (m, 9H); MS (ESI, m/z) 593.3 [M+1] + ; ESI-HRMS calcd. m/z for C 32 H 32 N 4 O 4 F 3 593.2376, found 593.2383 [M+1] + . Methyl 4'-(3-(hydroxymethyl)oxetan-3-yl)-5-(4-(4-(trifluoromethyl)p henyl)-1H- 1,2,3-triazol-1-yl)-[1,1'-biphenyl]-3-carboxylate (59). Method E: Yield 44%; 1 H NMR (400 MHz, CDCl 3 ) δ 8.41 (s, 1H), 8.36 (s, 1H), 8.34 (s, 2H), 8.06 (d, J =8.00 Hz, 2H), 7.75-7.64 (m, 4H), 7.54 (d, J =8.00 Hz, 2H), 5.01 (d, J =5.60 Hz, 2H), 4.83 (d, J =5.60 Hz, 2H), 4.11 (d, J =5.60 Hz, 2H), 4.02 (s, 3H); MS (ESI, m/z) 510.2 [M+1] + ; ESI-HRMS calcd. m/z for C 27 H 23 N 3 O 4 F 3 510.1641, found 510.1640 [M+1] + . Methyl 4'-(isoxazol-3-yl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3 -triazol-1-yl)- [1,1'-biphenyl]-3-carboxylate (60). Method F: Yield 91%; 1 H NMR (400 MHz, acetone-d 6 ) δ 9.48 (s, 1H), 8.88 (d, J =1.60 Hz, 1H), 8.62-8.61 (m, 2H), 8.46-8.45 (m, 1H), 8.26 (d, J =8.00 Hz, 2H), 8.14 (d, J =8.40 Hz, 2H), 8.04 (d, J =8.40 Hz, 2H), 7.88 (d, J =8.00 Hz, 2H), 7.13 (d, J =1.60 Hz, 1H), 4.03 (s, 3H); MS (ESI, m/z) 491.1 [M+1] + ; ESI-HRMS calcd. m/z for C 26 H 18 N 4 O 3 F 3 491.1331, found 491.1328 [M+1] + . Methyl 4'-(5-(hydroxymethyl)isoxazol-3-yl)-5-(4-(4-(trifluoromethyl )phenyl)-1H- 1,2,3-triazol-1-yl)-[1,1'-biphenyl]-3-carboxylate (61). Method E: Yield 46%; 1 H NMR (400 MHz, CDCl 3 ) δ 8.40 (s, 1H), 8.38 (s, 1H), 8.36- 8.35 (m, 2H), 8.04 (d, J =8.40 Hz, 2H), 7.93 (d, J =8.00 Hz, 2H), 7.78 (d, J =8.40 Hz, 2H), 7.72 (d, J =8.00 Hz, 2H), 6.62 (s, 1H), 4.84 (s, 2H), 4.00 (s, 3H); MS (ESI, m/z) 521.2 [M+1] + ; ESI- HRMS calcd. m/z for C 27 H 20 N 4 O 4 F 3 521.1437, found 521.1430 [M+1] + . Methyl 4'-(5-(2-hydroxyethyl)isoxazol-3-yl)-5-(4-(4-(trifluoromethy l)phenyl)-1H- 1,2,3-triazol-1-yl)-[1,1'-biphenyl]-3-carboxylate (62). Method F: Yield 80%; 1 H NMR (400 MHz, DMSO-d 6 ) δ 9.76 (s, 1H), 8.58 (s, 1H), 8.54 (s, 1H), 8.35 (s, 1H), 8.18 (d, J =8.40 Hz, 2H), 8.04-8.00 (m, 4H), 7.90 (d, J =8.40 Hz, 2H), 6.94 (s, 1H), 3.97 (s, 3H), 3.77 (q, J =6.40 Hz, 2H), 2.97 (d, J =6.40 Hz, 2H); MS (ESI, m/z) 535.2 [M+1] + ; ESI-HRMS calcd. m/z for C 28 H 22 N 4 O 4 F 3 535.1593, found 535.1592 [M+1] + . Methyl 4'-(2H-tetrazol-5-yl)-5-(4-(4-(trifluoromethyl)phenyl)-1H-1, 2,3-triazol-1-yl)- [1,1'-biphenyl]-3-carboxylate (63). Method C: Yield 53%; 1 H NMR (400 MHz, CD 3 OD) δ 9.26 (s, 1H), 8.52 (s, 1H), 8.44 (s, 1H), 8.39 (s, 1H), 8.18 (d, J =8.40 Hz, 2H), 8.12 (d, J =8.40 Hz, 2H), 7.90 (d, J =8.40 Hz, 2H), 7.76 (d, J =8.40 Hz, 2H), 4.00 (s, 3H); MS (ESI, m/z) 492.1 [M+1] + ; ESI-HRMS calcd. m/z for C 24 H 17 N 7 O 2 F 3 492.1396, found 492.1399 [M+1] + . Ethyl 4-(4-(5-(hydroxymethyl)isoxazol-3-yl)phenyl)-7-(4-(trifluoro methyl)phenyl)-2- naphthoate (65). Method F: Yield 50%; 1 H NMR (400 MHz, DMSO-d6) δ 8.81 (s, 1H), 8.71 (s, 1H), 8.09-8.05 (m, 5H), 7.99-7.97 (m, 1H), 7.94-7.93 (m, 1H), 7.89 (d, J =8.00 Hz, 2H), 7.68 (d, J =8.00 Hz, 2H), 7.04 (s, 1H), 5.77 (t, J =6.00 Hz, 1H), 4.66 (d, J =6.00 Hz, 2H), 4.41 (q, J =7.20 Hz, 2H), 1.38 (t, J =7.20 Hz, 3H); MS (ESI, m/z) 518.2 [M+1] + ; ESI-HRMS calcd. m/z for C 30 H 23 NO 4 F 3 518.1579, found 518.1582 [M+1] + . Methyl 3-amino-5-bromobenzoate (66). 3-Bromo-5-aminobenzoic acid (65, 1.01 g, 4.62 mmol) was stirred in methanol (15 mL) with ice cooling, and the yellow solution was treated with thionyl chloride (4.00 mL, 55.0 mmol) dropwise over 20 min. The resulting mixture was warm up to room temperature and left stirring for 15 h. The reaction mixture was quenched with aqueous saturated NaHCO3 solution at 0 °C. The solvent was then removed under vacuum, and the residue was suspended in ethyl acetate (200 mL). The organic phase was washed with brine (100 mL), dried (Na 2 SO 4 ) and concentrated in vacuo to afford the title compound 66 (1.08 g, 98%) as a yellow solid; 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.16 (dd, J =1.48, 2.12 Hz, 1H), 7.13 (t, J =1.64 Hz, 1H), 6.96 (t, J =2.00 Hz, 1H), 5.74 (s, 2H), 3.81 (s, 3H); MS (ESI, m/z) 231 [M+1] + ; ESI-HRMS calcd. m/z for C8H8BrNO2229.9817, found 229.9818 [M+1] + . Methyl 3-bromo-5-(4-(trifluoromethyl)benzamido)benzoate (67). To a solution of compounds 66 (70 mg, 0.304 mmol) in N,N-dimethylformamide (6 mL) were added 4-(Trifluoromethyl)benzoic acid (136 mg, 0.717 mmol), HATU (218 mg, 0.573 mmol) and N,N-diisopropylethylamine (250 µL, 1.43 mmol), and then this reaction mixture was stirred at room temperature for 5 h. The reaction mixture was partitioned ethyl acetate (20 mL) and water (10 mL), and the aqueous layer was extracted with ethyl acetate (20 mL x 2). The combined organic layer was washed brine (10 mL), dried over Na2SO4, filtered and evaporated under reduced pressure. The residue was purified by silica gel column chromatography (hexane:ethyl acetate=7:1) to afford compound 67 (86 mg, 70%) as a white solid; 1 H NMR (400 MHz, CDCl3) δ 8.32-8.31 (m, 1H), 8.05-8.04 (m, 1H), 7.99-7.98 (m, 3H), 7.79 (d, J =8.40 Hz, 2H), 3.93 (s, 3H); MS (ESI, m/z) 402.0 [M+1] + ; ESI-HRMS calcd. m/z for C 16 H 12 NO 3 F 3 Br 401.9953, found 401.9948 [M+1] + . Methyl 3-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)-5-(4- (trifluoromethyl)benzamido)benzoate (68). To a solution of compound 67 (40 mg, 0.099 mmol) in 1,4-dioxane (3 mL) were added bis(pinacolato)diboron (63 mg, 0.248 mmol), PdCl2(dppf) (2.0 mg, 0.002 µmol) and potassium acetate (30 mg, 0.298 mmol), and then this reaction mixture was stirred at 70 o C for 6 h. The reaction mixture was partitioned ethyl acetate (20 mL) and water (10 mL), and the aqueous layer was extracted with ethyl acetate (10 mL x 2). The combined organic layer was washed brine (5 mL), dried over Na2SO4, filtered and evaporated under reduced pressure. The residue was purified by silica gel column chromatography (hexane:ethyl acetate=2:1) to afford compound 68 (29.0 mg, 65%) as a white solid; 1 H NMR (400 MHz, CDCl 3 ) δ 8.50 (s, 1H), 8.29 (s, 1H), 8.15 (s, 1H), 7.99 (d, J =8.00 Hz, 2H), 7.78 (d, J =8.40 Hz, 2H), 3.93 (s, 3H), 1.35 (s, 12H); MS (ESI, m/z) 450.2 [M+1] + ; ESI-HRMS calcd. m/z for C 22 H 24 NO 5 F 3 B 450.1700, found 450.1701 [M+1] + . Methyl 4'-(5-(hydroxymethyl)isoxazol-3-yl)-5-(4-(trifluoromethyl)be nzamido)-[1,1'- biphenyl]-3-carboxylate (69). Method C: Yield 45%; 1 H NMR (400 MHz, CD 3 OD) δ 8.44 (m, 1H), 8.36-8.35 (m, 1H), 8.17-8.13 (m, 3H), 7.98 (d, J =8.40 Hz, 2H), 7.87-7.82 (m, 4H), 6.83 (s, 1H), 4.74 (s, 2H), 3.97 (s, 3H); MS (ESI, m/z) 497.1 [M+1] + ; ESI-HRMS calcd. m/z for C26H20N2O5F3497.1324, found 497.1323 [M+1] + . Methyl 2-amino-6-bromoisonicotinate (72). Compound 69 (200 mg, 0.922 mmol) was converted to compound 72 (83 mg, 40%) as a white solid, using a similar procedure to that used in the preparation of compound 66; 1 H NMR (400 MHz, CD 3 OD) δ 7.13 (d, 1H), 7.00 (d, 1H), 3.89 (s, 3H); MS (ESI, m/z) 231.0 [M+1] + ; ESI-HRMS calcd. m/z for C7H8BrN2O2230.9769, found 230.9769 [M+1] + . Methyl 4-amino-6-bromopicolinate (73). Compound 70 (320 mg, 1.47 mmol) was converted to compound 73 (136 mg, 40%) as a white solid, using a similar procedure to that used in the preparation of compound 66; 1 H NMR (400 MHz, DMSO-d 6 ) δ 7.23 (d, 1H), 6.79 (d, 1H), 3.81 (s, 3H); MS (ESI, m/z) 231.0 [M+1] + ; ESI-HRMS calcd. m/z for C 7 H 8 BrN 2 O 2 230.9769, found 230.9765 [M+1] + . Methyl 6-azido-4-bromopicolinate (74). Compound 71 (121 mg, 0.523 mmol) was dissolved in acetonitrile (4 mL) and cooled to 0 °C in an ice bathe. tert-Butyl nitrite (124 µL, 1.047 mmol) was added to the reaction mixture and stirred at 0 °C for 30 min. After that, azidotrimethylsilane (206 µL, 1.57 mmol) was added to the reaction mixture at 0 °C by dropwise. The resulting solution was stirred at room temperature for 12 h. The reaction mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane:ethyl acetate=3:1) to afford compound 74 (50 mg, 37%) as a white solid; 1 H NMR (400 MHz, CDCl3) δ 7.98 (s, 1H), 7.26 (s, 1H), 3.99 (s, 3H). Methyl 2-azido-6-bromoisonicotinate (75). Compound 72 (27 mg, 0.107 mmol) was converted to compound 75 (30 mg, 65%) as a white solid, using a similar procedure to that used in the preparation of compound 74; 1 H NMR (400 MHz, CDCl 3 ) δ 7.76 (d, 1H), 7.29 (d, 1H), 3.94 (s, 3H). Methyl 4-azido-6-bromopicolinate (76). Compound 73 (21 mg, 0.090 mmol) was converted to compound 76 (12 mg, 52%) as a white solid, using a similar procedure to that used in the preparation of compound 74; 1 H NMR (400 MHz, CDCl3) δ 7.75 (broad s, 1H), 7.31 (broad s, 1H), 4.02 (s, 3H). Methyl 4-bromo-6-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-1- yl)picolinate (77). To a solution of compound 74 (50 mg, 0.195 mmol) in tert-BuOH:water (5 mL, 1:1) were added 4-Ethynyl-α,α,α-trifluorotoluene (50 µL, 0.292 mmol) and Tris[(1-benzyl-1H-1,2,3- triazol-4-yl)methyl]amine (TBTA, 7 mg, 0.014 mmol). Subsequently, sodium ascorbate (58 mg, 0.292 mmol, freshly prepared 1 M aqueous solution) and CuSO 4 ·5H 2 O (37 mg, 0.146 mmol, freshly prepared 7.5% aqueous solution) were added into the reaction mixture, and then this reaction mixture was stirred at room temperature for 1.5 h. The solvent of reaction mixture was evaporated under reduced pressure. The residue was purified by silica gel column chromatography (hexane:ethyl acetate=3:1) to afford compound 77 (30 mg, 36%) as a white solid; 1 H NMR (400 MHz, CDCl 3 ) δ 8.98 (s, 1H), 8.65 (d, 1H), 8.31 (d, 1H), 8.08 (d, J =8.00 Hz, 2H), 7.74 (d, J =8.00 Hz, 2H), 4.06 (s, 3H); MS (ESI, m/z) 427.0 [M+1] + ; ESI-HRMS calcd. m/z for C16H11BrN4O2F3427.0017, found 427.0013 [M+1] + . Methyl 2-bromo-6-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-1- yl)isonicotinate (78). Compound 75 (27 mg, 0.107 mmol) was converted to compound 78 (30 mg, 65%) as a white solid, using a similar procedure to that used in the preparation of compound 77; 1 H NMR (400 MHz, CD 3 OD) δ 9.28 (s, 1H), 8.65 (d, 1H), 8.20-8.18 (m, 3H), 7.79 (d, J =8.00 Hz, 2H), 4.03 (s, 3H); MS (ESI, m/z) 427.0 [M+1] + ; ESI-HRMS calcd. m/z for C 16 H 11 BrN 4 O 2 F 3 427.0017, found 427.0018 [M+1] + . Methyl 6-bromo-4-(4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-1- yl)picolinate (79). Compound 76 (7.0 mg, 0.027 mmol) was converted to compound 79 (8.0 mg, 69%) as a white solid, using a similar procedure to that used in the preparation of compound 77; 1 H NMR (400 MHz, CD 3 OD) δ 9.24 (s, 1H), 8.68 (s, 1H), 8.42 (s, 1H), 8.08 (d, J =8.00 Hz, 2H), 7.73 (d, J =8.00 Hz, 2H), 4.04 (s, 3H); MS (ESI, m/z) 427.0 [M+1] + ; ESI-HRMS calcd. m/z for C 16 H 11 BrN 4 O 2 F 3 427.0017, found 427.0022 [M+1] + . Methyl 4-(4-(1-(tert-butoxycarbonyl)piperidin-4-yl)phenyl)-6-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)picolinate (80). Method C: Yield 35%; 1 H NMR (400 MHz, CDCl3) δ 9.05 (s, 1H), 8.67 (d, 1H), 8.41 (d, 1H), 8.10 (d, J =8.00 Hz, 2H), 7.78-7.73 (m, 4H), 7.41 (d, J =8.40 Hz, 2H), 4.35-4.21 (m, 2H), 2.90-2.79 (m, 3H), 1.93-1.89 (m, 2H), 1.75-1.63 (m, 2H), 1.49 (s, 9H); MS (ESI, m/z) 608.3 [M+1] + . Methyl 2-(4-(1-(tert-butoxycarbonyl)piperidin-4-yl)phenyl)-6-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)isonicotinate (81). Method C: Yield 63%; 1 H NMR (400 MHz, CD3OD) δ 9.36 (s, 1H), 8.49 (s, 1H), 8.38 (s, 1H), 8.20-8.16 (m, 4H), 7.78 (d, J =8.40 Hz, 2H), 7.41 (d, J =8.40 Hz, 2H), 4.26-4.16 (m, 2H), 4.04 (s, 3H), 2.95-2.77 (m, 3H), 1.88-1.85 (m, 2H), 1.69-1.59 (m, 2H), 1.49 (s, 9H); MS (ESI, m/z) 608.3 [M+1] + ; ESI-HRMS calcd. m/z for C32H33N5O4F3608.2485, found 608.2492 [M+1] + . Methyl 6-(4-(1-(tert-butoxycarbonyl)piperidin-4-yl)phenyl)-4-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)picolinate (82). Method C: Yield 91%; 1 H NMR (400 MHz, CD3OD) δ 9.44-9.36 (m, 1H), 8.57-8.46 (m, 2H), 8.15-8.10 (m, 4H), 7.81-7.76 (m, 2H), 7.40-7.35 (m, 2H), 4.24-4.21 (m, 2H), 4.04 (s, 3H), 2.88-2.77 (m, 3H), 1.86-1.84 (m, 2H), 1.65-1.62 (m, 2H), 1.49 (s, 9H); MS (ESI, m/z) 608.2 [M+1] + ; ESI-HRMS calcd. m/z for C32H33N5O4F3608.2485, found 608.2474 [M+1] + . 4-(4-(1-(tert-Butoxycarbonyl)piperidin-4-yl)phenyl)-6-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)picolinic acid (83). Method A: Yield 68%; 1 H NMR (400 MHz, DMSO-d6) δ 9.64 (s, 1H), 8.57 (s, 1H), 8.41 (s, 1H), 8.30 (d, J =8.00 Hz, 2H), 7.94 (d, J =8.40 Hz, 2H), 7.87 (d, J =8.00 Hz, 2H), 7.48 (d, J =8.40 Hz, 2H), 4.11-4.05 (m, 2H), 2.90-2.73 (m, 3H), 1.82-1.79 (m, 2H), 1.60-1.50 (m, 2H), 1.42 (s, 9H); MS (ESI, m/z) 594.2 [M+1] + ; ESI-HRMS calcd. m/z for C31H31N5O4F3 594.2328, found 594.2320 [M+1] + . 2-(4-(1-(tert-Butoxycarbonyl)piperidin-4-yl)phenyl)-6-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)isonicotinic acid (84). Method A: Yield 91%; 1 H NMR (400 MHz, CD 3 OD) δ 9.38 (s, 1H), 8.53 (s, 1H), 8.42 (s, 1H), 8.19 (t, J =8.40 Hz, 4H), 7.79 (d, J =8.40 Hz, 2H), 7.42 (d, J =8.00 Hz, 2H), 4.26- 4.15 (m, 2H), 2.96-2.73 (m, 3H), 1.89-1.76 (m, 2H), 1.70-1.55 (m, 2H), 1.49 (s, 9H); MS (ESI, m/z) 616.3 [M+Na] + ; ESI-HRMS calcd. m/z for C31H30N5O4F3Na 616.2148, found 616.2158 [M+Na] + . 6-(4-(1-(tert-Butoxycarbonyl)piperidin-4-yl)phenyl)-4-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)picolinic acid (85). Method A: Yield 77%; 1 H NMR (400 MHz, CD 3 OD) δ 9.27 (s, 1H), 8.58-8.52 (m, 2H), 8.10 (m, 4H), 7.74 (d, J =7.60 Hz, 2H), 7.39 (broad s, 2H), 4.24-4.21 (m, 2H), 2.89-2.73 (m, 3H), 1.89-1.86 (m, 2H), 1.70-1.60 (m, 2H), 1.47 (s, 9H); MS (ESI, m/z) 594.2 [M+1] + ; ESI- HRMS calcd. m/z for C 31 H 31 N 5 O 4 F 3 594.2328, found 594.2318 [M+1] + . tert-Butyl 4-(3'-((2-(dimethylamino)-2-oxoethoxy)carbonyl)-5'-(4-(4- (trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-yl)-[1,1'-biphen yl]-4-yl)piperidine-1-carboxylate (87). To a solution of 86 (166 mg, 0.280 mmol), which was synthesized according to literature procedures reported, 1 in DMF (10 mL) was added K2CO3 (116 mg, 0.840 mmol). 2- Chloro-N,N-dimethylacetamide (68 mg, 0.560 mmol) was added to the reaction mixture under N 2 atmosphere, and the reaction was stirred at 45 °C for 1 h under N 2 atmosphere. The reaction mixture was partitioned ethyl acetate (10 mL) and water (10 mL), and the aqueous phase was extracted with ethyl acetate (2 x 10 mL). The combined organic layers were dried over Na2SO4, filtered, and concentrated under pressure. The residue was purified by silica gel chromatography (hexane:ethyl acetate = 3:1) to afford compound 87 (205 mg, 70%) as a white solid; 1 H NMR (400 MHz, CDCl3) δ 8.43-8.41 (m, 2H), 8.38-8.36 (m, 2H), 8.05 (d, J =8.00 Hz, 2H), 7.73 (d, J =8.40 Hz, 2H), 7.65 (d, J =8.40 Hz, 2H), 7.34 (d, J =8.40 Hz, 2H), 5.05 (s, 2H), 3.76-3.72 (m, 2H), 3.07 (s, 3H), 3.02 (s, 3H), 2.87-2.77 (m, 2H), 2.76-2.67 (m, 1H), 1.89-1.83 (m, 2H), 1.72- 1.62 (m, 2H), 1.49 (s, 9H); MS (ESI, m/z) 622.2 [M+1-tert-butyl] + . tert-Butyl 4-(3'-(hydroxymethyl)-5'-(4-(4-(trifluoromethyl)phenyl)-1H-1 ,2,3-triazol- 1-yl)-[1,1'-biphenyl]-4-yl)piperidine-1-carboxylate (89). To a solution of 88 (35 mg, 0.058 mmol), which was synthesized according to literature procedures reported, 1 in THF at 0 °C, 2 M lithium aluminum hydride solution in THF (2.6 mg, 0.070 mmol) was added dropwise over a period of 0.5 h under N2 atmosphere. After the addition was complete, the reaction mixture was stirred at 0 °C under N2 atmosphere for 1 h. The unreacted LAH was quenched with saturated aqueous NH 4 Cl and the reaction mixture was extracted with dichloromethane 3 times. The combined organic extracts were dried with anhydrous Na2SO4, and the mixture was filtered through celite pad. The filtered mixture was concentrated in vacuum and chromatographed on silica gel with hexanes/ethyl acetate = 3:1 to afford 89 (15 mg, 45%) as a white solid; 1 H NMR (400 MHz, CD 3 OD) δ 9.16 (s, 1H), 8.15 (d, J =8.00 Hz, 2H), 8.05 (s, 1H), 7.90 (s, 1H), 7.80-7.76 (m, 3H), 7.70 (d, J =8.40 Hz, 2H), 7.38 (d, J =8.40 Hz, 2H), 4.80 (s, 2H), 4.25-4.22 (m, 2H), 2.96−2.77 (m, 3H), 1.88-1.85 (m, 2H), 1.68−1.59 (m, 2H), 1.49 (s, 9H); MS (ESI, m/z) 579.2 [M+1]+; ESI-HRMS calcd. m/z for C32H34N4O3F3579.2583, found 579.2588 [M+1]+. tert-Butyl 4-(3'-(((di-tert-butoxyphosphoryl)oxy)methyl)-5'-(4-(4-(trif luoromethyl)phenyl)-1H- 1,2,3-triazol-1-yl)-[1,1'-biphenyl]-4-yl)piperidine-1-carbox ylate (90). A stirred solution of 89 (12 mg, 0.020 mmol) in dry THF (1.0 mL) was treated successively with tetrazole (17 mg, 0.242 mmol) and di-tert-butyl N,N-diethylphosphoramidite (115 µL, 0.414 mmol) at room temperature under N 2 atmosphere. The resulting solution was stirred at room temperature for 1 h under N2 atmosphere, and the reaction mixture was cooled to -78 °C. 3-Chloroperbenzoic acid (MCPBA, contains 50%, 78 mg) was added, and the reaction mixture was warmed to 0 °C and stirred for 0.5 h. The reaction mixture was partitioned ethyl acetate (10 mL) and aqueous saturated NaHCO 3 solution (10 mL), and the aqueous phase was extracted with ethyl acetate (2 x 10 mL). The combined organic layers were dried over Na2SO4, filtered, and concentrated under pressure. The residue was purified by silica gel chromatography (chloroform:methanol = 30:1) to afford compound 90 (13 mg, 84%) as a white solid; 1 H NMR (400 MHz, CD3OD) δ 9.22 (s, 1H), 8.18-8.16 (m, 3H), 8.01 (s, 1H), 7.83-7.79 (m, 3H), 7.73 (d, J =8.40 Hz, 2H), 7.41 (d, J =8.40 Hz, 2H), 5.23-5.22 (m, 2H), 4.27-4.23 (m, 2H), 2.96−2.77 (m, 3H), 1.91-1.87 (m, 2H), 1.71−1.59 (m, 2H), 1.52-1.50 (m, 27H); MS (ESI, m/z) 793.3 [M+Na]+; ESI-HRMS calcd. m/z for C40H50N4O6F3PNa 793.3318, found 793.3303 [M+Na]+. EXAMPLE 2 4-(4-(Pyrrolidin-3-yl)phenyl)-7-(4-(trifluoromethyl)phenyl)- 2-naphthoic acid (105). Procedure A. Into a solution of 175 (15.7 mg, 0.032 mmol) in MeOH (2 mL) was added aq. potassium hydroxide (0.2 M, 1 mL). The resulting mixture was stirred at 50 o C overnight. The reaction was quenched and neutralized by 1 M HCl to pH 6. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, tR = 26 min) to give stereoisomeric mixture 105 as a white powder (8.6 mg, 58%): 1 H NMR (400 MHz, DMSO) δ 8.48 (s, 1H), 8.41 (d, J = 2.0 Hz, 1H), 8.06 (d, J = 8.1 Hz, 2H), 7.98 (d, J = 1.5 Hz, 1H), 7.94 – 7.80 (m, 4H), 7.48 – 7.39 (m, 4H), 3.30 – 3.15 (m, 2H), 3.06 – 2.97 (m, 1H), 2.97 – 2.89 (m, 1H), 2.77 – 2.68 (m, 1H), 2.27 – 2.15 (m, 1H), 1.85 – 1.72 (m, 1H). HRMS m/z [M+H] + for C 28 H 23 NO 2 F 3 calculated 462.1681, found 462.1689. 4-(4-(3-Methylpiperidin-4-yl)phenyl)-7-(4-(trifluoromethyl)p henyl)-2-naphthoic acid (106). Treatment of 165 (13.2 mg, 0.026 mmol) by using Procedure A gave stereoisomeric mixture 106 (5.4 mg, 42%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.65 (s, 1H), 8.34 (d, J = 1.9 Hz, 1H), 8.02 – 7.92 (m, 4H), 7.84 – 7.74 (m, 3H), 7.48 – 7.36 (m, 4H), 3.61 – 3.36 (m, 2H), 3.25 – 2.54 (m, 4H), 2.31 – 2.07 (m, 1H), 1.34 – 1.29 (m, 1H), 1.20 – 1.13 (m, 3H). HRMS m/z [M+H] + for C30H27NO2F3 calculated 490.1994, found 490.1990. 4-(4-(2-Methylpiperidin-4-yl)phenyl)-7-(4-(trifluoromethyl)p henyl)-2-naphthoic acid (107). Treatment of 166 (11 mg, 0.021 mmol) by using Procedure A gave stereoisomeric mixture 7 (8.0 mg, 76%) as a white powder: 1 H NMR (400 MHz, methanol-d4) δ 8.62 (s, 1H), 8.30 (d, J = 2.0 Hz, 1H), 8.03 (s, 1H), 7.96 – 7.91 (m, 2H), 7.77 (t, J = 9.3 Hz, 2H), 7.69 – 7.50 (m, 3H), 7.49 – 7.37 (m, 3H), 3.52 (t, J = 15.3 Hz, 1H), 3.45 – 3.25 (m, 1H), 3.25 – 3.10 (m, 1H), 3.09 – 2.98 (m, 1H), 2.22 – 2.04 (m, 2H), 1.90 – 1.62 (m, 1H), 1.41 (d, J = 6.5 Hz, 2.2H), 1.36 (d, J = 6.5 Hz, 0.8H), 0.97 – 0.84 (m, 1H). HRMS m/z [M+H] + for C30H27NO2F3 calculated 490.1994, found 490.1996. 4-(4-(3-Azabicyclo[4.1.0]heptan-6-yl)phenyl)-7-(4-(trifluoro methyl)phenyl)-2- naphthoic acid (108). Treatment of 168 (8 mg, 0.0156 mmol) by using Procedure A gave diastereomeric mixture 108 (5.6 mg, 74%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.75 – 8.70 (m, 1H), 8.40 (d, J = 1.9 Hz, 1H), 8.02 – 7.87 (m, 5H), 7.80 (d, J = 8.2 Hz, 2H), 7.58 (d, J = 8.1 Hz, 2H), 7.49 (d, J = 8.2 Hz, 2H), 3.85 (dd, J = 13.5, 7.8 Hz, 1H), 3.36 – 3.32 (m, 2H), 2.97 (ddd, J = 13.2, 9.4, 5.9 Hz, 1H), 2.51 – 2.40 (m, 2H), 1.75 – 1.64 (m, 1H), 1.37 (dd, J = 9.1, 5.6 Hz, 1H), 1.18 (t, J = 5.6 Hz, 1H). HRMS m/z [M+H] + for C 30 H 25 NO 2 F 3 calculated 488.1837, found 488.1840. 4-(4-(7,7-Difluoro-3-azabicyclo[4.1.0]heptan-6-yl)phenyl)-7- (4- (trifluoromethyl)phenyl)-2-naphthoic acid (109). Treatment of 169 (5.6 mg, 0.01 mmol) by using Procedure A gave diastereomeric mixture 109 (2.7 mg, 52%) as a white powder: 1 H NMR (400 MHz, methanol-d4) δ 8.63 (s, 1H), 8.37 (s, 1H), 8.02 (s, 1H), 7.96 (d, J = 8.1 Hz, 2H), 7.84 (d, J = 8.9 Hz, 1H), 7.78 (d, J = 8.1 Hz, 2H), 7.72 (d, J = 8.8 Hz, 1H), 7.54 (d, J = 7.7 Hz, 2H), 7.41 (d, J = 7.8 Hz, 2H), 3.82 (t, J = 11.6 Hz, 1H), 3.33 – 3.25 (m, 1H), 3.22 – 3.14 (m, 1H), 2.95 – 2.82 (m, 1H), 2.51 (d, J = 14.9 Hz, 1H), 2.38 (t, J = 11.0 Hz, 1H), 2.27 – 2.15 (m, 1H). HRMS m/z [M+H] + for C 30 H 23 NO 2 F 5 calculated 524.1649, found 524.1655. rac-4-(4-((1S,5R,6S)-2-Azabicyclo[4.1.0]heptan-5-yl)phenyl)- 7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (110). Treatment of 170 (9.8 mg, 0.02 mmol) by using Procedure A gave racemate 110 (2.4 mg, 25%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.56 (s, 1H), 8.35 (d, J = 2.0 Hz, 1H), 8.06 – 8.02 (m, 1H), 7.99 (d, J = 8.9 Hz, 3H), 7.86 – 7.76 (m, 3H), 7.57 (d, J = 8.1 Hz, 2H), 7.52 (d, J = 8.2 Hz, 2H), 3.26 (t, J = 6.6 Hz, 1H), 2.83 – 2.73 (m, 1H), 2.61 – 2.49 (m, 2H), 1.87 – 1.81 (m, 1H), 1.62 – 1.52 (m, 1H), 1.24 – 1.16 (m, 1H), 0.85 (ddd, J = 9.5, 7.3, 5.5 Hz, 1H), 0.67 – 0.59 (m, 1H). HRMS m/z [M+H] + for C 30 H 25 NO 2 F 3 calculated 488.1837, found 488.1831. rac-4-(4-((1R,5R,6R)-2-Azabicyclo[4.1.0]heptan-5-yl)phenyl)- 7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (111). Treatment of 171 (15.4 mg, 0.03 mmol) by using Procedure A gave racemate 111 (0.16 mg, 1%) as a white powder: 1 H NMR (400 MHz, methanol-d4) δ 8.56 (s, 1H), 8.35 (s, 1H), 8.06 – 7.96 (m, 4H), 7.86 – 7.76 (m, 3H), 7.57 (d, J = 8.1 Hz, 1H), 7.52 (d, J = 8.1 Hz, 1H), 3.27 – 3.16 (m, 1H), 2.85 – 2.67 (m, 2H), 2.60 (s, 1H), 1.91 – 1.86 (m, 1H), 1.57 (d, J = 27.1 Hz, 1H), 1.25 (s, 1H), 0.89 (s, 1H), 0.73 – 0.62 (m, 1H). HRMS m/z [M+H] + for C30H25NO2F3 calculated 488.1837, found 488.1833. rac-4-(4-((1S,4S,5S)-2-Azabicyclo[2.2.1]heptan-5-yl)phenyl)- 7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (112). Treatment of 167 (9.3 mg, 0.018 mmol) by using Procedure A gave enantiomeric mixture 112 (4.1 mg, 47%) as a white powder: 1 H NMR (400 MHz, DMSO) δ 8.46 (s, 1H), 8.39 (s, 1H), 8.04 (d, J = 8.0 Hz, 2H), 7.96 (s, 1H), 7.94 – 7.78 (m, 4H), 7.45 – 7.35 (m, 4H), 3.41 – 3.37 (m, 1H), 2.97 (dd, J = 9.1, 5.4 Hz, 1H), 2.79 (dd, J = 9.2, 3.5 Hz, 1H), 2.63 (d, J = 9.4 Hz, 1H), 2.43 – 2.35 (m, 1H), 2.00 (t, J = 10.8 Hz, 1H), 1.75 (dt, J = 12.3, 4.6 Hz, 1H), 1.64 – 1.57 (m, 1H), 1.37 (d, J = 9.6 Hz, 1H). HRMS m/z [M+H] + for C 30 H 25 NO 2 F 3 calculated 488.1837, found 488.1842. rac-4-(4-((1S,4S,5S)-2-Acetyl-2-azabicyclo[2.2.1]heptan-5-yl )phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (113). Into the solution of 112 (0.9 mg, 0.0019 mmol) and TEA (3.2 μL, 2.3 mg, 0.023 mmol) in DMF (0.5 mL) was added acetic anhydride (1.8 μL, 2 mg, 0.019 mmol). The solution was stirred at rt for 90 min. Water (10 μL, 10 mg, 0.56 mmol) was added and the reaction mixture was stirred for another 1 h. The reaction residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, t R = 26 min) to give stereoisomeric mixture 113 as a white powder (0.5 mg, 53%): 1 H NMR (400 MHz, methanol-d4) δ 8.58 (s, 1H), 8.36 (d, J = 1.9 Hz, 1H), 8.05 – 7.96 (m, 4H), 7.83 (dd, J = 8.9, 2.1 Hz, 1H), 7.80 (d, J = 8.2 Hz, 2H), 7.51 (d, J = 8.1 Hz, 2H), 7.46 (d, J = 8.0 Hz, 2H), 4.69 (s, 0.4H), 4.46 (s, 0.6H), 3.60 (dd, J = 9.3, 3.4 Hz, 0.4H), 3.50 – 3.39 (m, 1H), 3.38 – 3.33 (m, 0.6H), 3.25 – 3.14 (m, 1H), 2.85 (d, J = 9.1 Hz, 1H), 2.31 (dt, J = 19.5, 10.3 Hz, 1H), 2.15 (s, 1.8H), 2.10 – 2.06 (m, 0.6H), 2.05 (s, 1.2H), 2.04 – 1.90 (m, 1.4H), 1.81 (d, J = 10.3 Hz, 0.6H), 1.71 (d, J = 10.5 Hz, 0.4H). HRMS m/z [M+H] + for C 32 H 27 NO 2 F 3 calculated 530.1943, found 530.1939. 4-(4-((1S,4S,5S)-2-Azabicyclo[2.2.1]heptan-5-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoic acid (114). Procedure B. LiOH (14.8 mg, 0.62 mmol) was added to a solution of 184 (12.7 mg, 0.025 mmol) in THF-H 2 O-MeOH (3:1:1, 2 mL). The resulting mixture was sonicated for 30 s and stirred at rt for 3 h. The reaction was quenched and neutralized by 1 M HCl. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, t R = 28 min) to give 114 as a white powder (5.3 mg, 44%): 1 H NMR (400 MHz, methanol-d4) δ 8.74 (s, 1H), 8.42 (s, 1H), 8.04 – 7.95 (m, 4H), 7.95 – 7.88 (m, 1H), 7.80 (d, J = 8.0 Hz, 2H), 7.56 – 7.46 (m, 4H), 4.25 (s, 1H), 3.35 – 3.26 (m, 3H), 2.96 (s, 1H), 2.46 – 2.35 (m, 1H), 2.18 (d, J = 15.1 Hz, 1H), 2.11 (d, J = 11.7 Hz, 1H), 1.81 (d, J = 11.8 Hz, 1H). HRMS m/z [M+H] + for C30H25NO2F3 calculated 488.1837, found 488.1846. 4-(4-((1R,4R,5R)-2-Azabicyclo[2.2.1]heptan-5-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoic acid (115). Treatment of 300 (7.56 mg, 0.0147 mmol) by using Procedure B gave 115 (3.9 mg, 54%) as a white powder: 1 H NMR (400 MHz, methanol- d 4 ) δ 8.74 (s, 1H), 8.42 (s, 1H), 8.04 – 7.95 (m, 4H), 7.95 – 7.89 (m, 1H), 7.80 (d, J = 8.1 Hz, 2H), 7.56 – 7.46 (m, 4H), 4.25 (s, 1H), 3.32 – 3.26 (m, 3H), 2.96 (s, 1H), 2.40 (dd, J = 14.6, 9.4 Hz, 1H), 2.22 – 2.14 (m, 1H), 2.11 (d, J = 11.8 Hz, 1H), 1.81 (d, J = 11.7 Hz, 1H). HRMS m/z [M+H] + for C 30 H 25 NO 2 F 3 calculated 488.1837, found 488.1835. 4-(4-((1R,4R,6S)-2-Azabicyclo[2.2.1]heptan-6-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoic acid (116). Treatment of 189 (3.46 mg, 0.0067 mmol) by using Procedure B gave 116 (1 mg, 30%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.77 (d, J = 1.5 Hz, 1H), 8.45 (d, J = 1.9 Hz, 1H), 8.02 (d, J = 7.3 Hz, 3H), 7.99 (d, J = 1.7 Hz, 1H), 7.95 (dd, J = 8.9, 2.0 Hz, 1H), 7.83 (d, J = 8.2 Hz, 2H), 7.57 (d, J = 8.2 Hz, 2H), 7.52 (d, J = 8.2 Hz, 2H), 4.25 (s, 1H), 3.48 (t, J = 7.6 Hz, 1H), 3.28 (d, J = 11.0 Hz, 1H), 3.22 – 3.16 (m, 1H), 2.95 (s, 1H), 2.29 – 2.17 (m, 2H), 2.14 (d, J = 11.8 Hz, 1H), 1.83 (d, J = 11.7 Hz, 1H). HRMS m/z [M+H] + for C30H25NO2F3 calculated 488.1837, found 488.1844. 4-(4-((1S,4S,6R)-2-Azabicyclo[2.2.1]heptan-6-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoic acid (117). Treatment of 105 (23 mg, with 17% TEA;0.037 mmol) by using Procedure B gave 117 (6.5 mg, 36%) as a white powder: 1 H NMR (400 MHz, methanol-d4) δ 8.77 (d, J = 1.6 Hz, 1H), 8.45 (d, J = 2.0 Hz, 1H), 8.04 – 7.92 (m, 5H), 7.82 (d, J = 8.0 Hz, 2H), 7.56 (d, J = 8.0 Hz, 2H), 7.51 (d, J = 8.0 Hz, 2H), 4.24 (s, 1H), 3.47 (t, J = 7.6 Hz, 1H), 3.27 – 3.24 (m, 1H), 3.18 (d, J = 10.9 Hz, 1H), 2.94 (s, 1H), 2.27 – 2.18 (m, 2H), 2.13 (d, J = 12.0 Hz, 1H), 1.83 (d, J = 11.8 Hz, 1H). HRMS m/z [M+H] + for C30H25NO2F3 calculated 488.1837, found 488.1837. 4-(4-((1S,4S,5S)-2-Acetyl-2-azabicyclo[2.2.1]heptan-5-yl)phe nyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (118). Acetic anhydride (4 μL, 4.2 mg, 0.07 mmol) was added to a solution of 114 (3.5 mg, 0.007 mmol) and TEA (11.6 μL, 8.5 mg, 0.084 mmol) in DMF (0.5 mL). The solution was stirred at rt for 90 min. Water (10 μL, 10 mg, 0.56 mmol) was added and the reaction mixture was stirred for another 1 h. The reaction residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, tR = 26 min) to give 118 as a white powder (3 mg, 81%): 1 H NMR (400 MHz, methanol-d 4 ) δ 8.63 (s, 1H), 8.39 (d, J = 2.0 Hz, 1H), 8.06 – 7.98 (m, 4H), 7.87 (dd, J = 8.9, 2.0 Hz, 1H), 7.81 (d, J = 8.2 Hz, 2H), 7.52 (d, J = 8.3 Hz, 2H), 7.48 (d, J = 8.1 Hz, 2H), 4.71 (s, 0.5H), 4.47 (s, 0.5H), 3.62 (dd, J = 9.3, 3.4 Hz, 0.5H), 3.48 – 3.39 (m, 1H), 3.38 – 3.34 (m, 0.5H), 3.29 – 3.23 (m, 1H), 2.87 (d, J = 9.7 Hz, 1H), 2.40 – 2.26 (m, 1H), 2.17 (s, 1.5H), 2.11 – 2.07 (m, 0.5H), 2.06 (s, 1.5H), 2.03 – 1.90 (m, 1.5H), 1.82 (d, J = 10.4 Hz, 0.5H), 1.73 (d, J = 10.5 Hz, 0.5H). HRMS m/z [M+H] + for C 32 H 27 NO 2 F 3 calculated 530.1943, found 530.1945. 4-(4-((1S,4S,5S)-3-Oxo-2-azabicyclo[2.2.1]heptan-5-yl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (119). Treatment of 186 (5.3 mg, 0.01 mmol) by using Procedure B (reaction time, 90 min) gave 119 (3.1 mg, 62%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.58 (s, 1H), 8.35 (d, J = 1.9 Hz, 1H), 8.05 – 7.96 (m, 4H), 7.87 – 7.81 (m, 1H), 7.79 (d, J = 8.1 Hz, 2H), 7.56 – 7.46 (m, 4H), 4.08 (s, 1H), 3.35 – 3.31 (m, 1H), 2.89 (d, J = 2.2 Hz, 1H), 2.32 (t, J = 10.5 Hz, 1H), 2.23 – 2.13 (m, 1H), 2.03 – 1.93 (m, 2H). HRMS m/z [M+H] + for C 30 H 23 NO 3 F 3 calculated 502.1630, found 502.1649. 4-(4-((1R,4R,5R)-3-Oxo-2-azabicyclo[2.2.1]heptan-5-yl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (120). Treatment of 302 (10.6 mg, 0.02 mmol) by using Procedure B (reaction time, 90 min) gave 120 (7.6 mg, 76%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.63 (s, 1H), 8.37 (s, 1H), 8.04 – 7.96 (m, 4H), 7.86 (d, J = 8.9 Hz, 1H), 7.79 (d, J = 8.0 Hz, 2H), 7.51 (s, 4H), 4.08 (s, 1H), 3.34 (s, 1H), 2.89 (s, 1H), 2.33 (t, J = 10.8 Hz, 1H), 2.18 (d, J = 12.9 Hz, 1H), 1.98 (q, J = 9.9, 9.3 Hz, 2H). HRMS m/z [M+H] + for C 30 H 23 NO 3 F 3 calculated 502.1630, found 502.1634. 4-(4-((1R,4R,6S)-3-Oxo-2-azabicyclo[2.2.1]heptan-6-yl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (121). Treatment of 191 (5.3 mg, 0.01 mmol) by using Procedure B (reaction time, 90 min) gave 21 (2.02 mg, 40%) as a white powder: 1 H NMR (400 MHz, methanol-d4) δ 8.57 (s, 1H), 8.35 (d, J = 2.0 Hz, 1H), 8.05 – 7.96 (m, 4H), 7.86 – 7.76 (m, 3H), 7.53 (d, J = 8.3 Hz, 2H), 7.49 (d, J = 8.3 Hz, 2H), 4.00 (s, 1H), 3.38 (dd, J = 9.1, 5.2 Hz, 1H), 2.82 (dd, J = 3.9, 1.8 Hz, 1H), 2.26 (dt, J = 12.9, 4.6 Hz, 1H), 2.19 – 2.09 (m, 1H), 1.98 – 1.88 (m, 2H). HRMS m/z [M+H] + for C30H23NO3F3 calculated 502.1630, found 502.1634. 4-(4-((1S,4S,6R)-3-Oxo-2-azabicyclo[2.2.1]heptan-6-yl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (122). Treatment of 307 (5.4 mg, 0.01 mmol) by using Procedure B (reaction time, 90 min) gave 122 (2.25 mg, 45%) as a white powder: 1 H NMR (400 MHz, methanol-d4) δ 8.59 (s, 1H), 8.36 (d, J = 1.9 Hz, 1H), 8.05 – 7.95 (m, 4H), 7.83 (dd, J = 8.9, 1.9 Hz, 1H), 7.79 (d, J = 8.1 Hz, 2H), 7.52 (d, J = 8.4 Hz, 2H), 7.49 (d, J = 8.3 Hz, 3H), 4.00 (s, 1H), 3.37 (dd, J = 9.1, 5.1 Hz, 1H), 2.82 (d, J = 3.8 Hz, 1H), 2.25 (dt, J = 13.0, 4.6 Hz, 1H), 2.19 – 2.09 (m, 1H), 2.01 – 1.88 (m, 2H). HRMS m/z [M+H] + for C30H23NO3F3 calculated 502.1630, found 502.1628. 4-(4-((1R,3r,5S)-8-Benzyl-3-hydroxy-8-azabicyclo[3.2.1]octan -3-yl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (127s). Treatment of 172 (61.2 mg, 0.096 mmol) by using Procedure A gave 27s (48.3 mg, 83%) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C 38 H 33 NO 3 F 3 calculated 608.2413, found 608.2421. 4-(4-((1R,3r,5S)-3-Hydroxy-8-azabicyclo[3.2.1]octan-3-yl)phe nyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (127). Pd/C (5%, 30 mg) was added to a solution of 127s (42.5 mg, 0.07 mmol) in MeOH. The resulting mixture was stirred under H 2 atmosphere (150 psi) at rt for 7 h. The Pd/C was filtered. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 10% → 100% A in 40 min, flow rate = 5 mL/min, t R = 32 min) to give 27 as a white powder (20.9 mg, 58%): 1 H NMR (400 MHz, MeOD) δ 8.64 (s, 1H), 8.32 (d, J = 1.9 Hz, 1H), 7.94 (d, J = 1.6 Hz, 1H), 7.86 (d, J = 8.1 Hz, 2H), 7.73 (dd, J = 12.1, 8.5 Hz, 3H), 7.62 (d, J = 8.2 Hz, 3H), 7.28 (d, J = 7.9 Hz, 2H), 4.23 – 4.17 (m, 2H), 2.69 (q, J = 9.1, 7.5 Hz, 2H), 2.59 (dd, J = 15.5, 3.5 Hz, 2H), 2.17 – 2.09 (m, 4H). HRMS m/z [M+H] + for C 31 H 27 NO 3 F 3 calculated 518.1943, found 518.1947. 4-(4-(8-Azabicyclo[3.2.1]oct-2-en-3-yl)phenyl)-7-(4-(trifluo romethyl)phenyl)-2- naphthoic acid (128). A solution of 127 (14.9 mg, 0.0288 mmol) in TFA (2 mL) was refluxed in a 90 o C oil bath for 1.5 h. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, tR = 27 min) to give 128 as a white powder (3.9 mg, 27%): 1 H NMR (400 MHz, MeOD) δ 8.69 (s, 1H), 8.35 (d, J = 1.8 Hz, 1H), 7.93 (q, J = 4.1 Hz, 4H), 7.85 (dd, J = 8.9, 1.9 Hz, 1H), 7.74 (d, J = 8.1 Hz, 2H), 7.61 (d, J = 7.9 Hz, 2H), 7.47 (d, J = 7.9 Hz, 2H), 6.51 (d, J = 5.7 Hz, 1H), 4.39 (t, J = 5.7 Hz, 1H), 4.36 – 4.32 (m, 1H), 3.28 – 3.18 (m, 1H), 2.75 (d, J = 18.0 Hz, 1H), 2.41 – 2.25 (m, 2H), 2.25 – 2.12 (m, 1H), 2.08 – 1.96 (m, 1H). HRMS m/z [M+H] + for C 31 H 25 NO 2 F 3 calculated 500.1837, found 500.1841. 4-(4-(3-Azabicyclo[3.2.1]octan-8-yl)phenyl)-7-(4-(trifluorom ethyl)phenyl)-2- naphthoic acid (129). Pd/C (5%, 1 mg) was added to a solution of 128 (1 mg, 0.0019) in MeOH. The resulting mixture was stirred under H 2 atmosphere (100 psi) at rt for 3 h. The Pd/C was filtered. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, t R = 29 min) to give 129 as a white powder (0.88 mg, 92%): 1 H NMR (400 MHz, MeOD) δ 8.75 (s, 1H), 8.44 (s, 1H), 8.05 – 7.96 (m, 4H), 7.96 – 7.90 (m, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.67 (d, J = 8.0 Hz, 2H), 7.55 (d, J = 8.1 Hz, 2H), 4.13 (s, 2H), 3.32 – 3.29 (m, 1H), 2.62 – 2.52 (m, 2H), 2.46 (d, J = 15.4 Hz, 2H), 2.09 – 2.00 (m, 4H). HRMS m/z [M+H] + for C 31 H 27 NO 2 F 3 calculated 502.1994, found 502.1998. 4-(4-((1R,5S,8r)-3-Benzyl-8-hydroxy-3-azabicyclo[3.2.1]octan -8-yl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (130). Treatment of 173 (5 mg, 0.008 mmol) by using Procedure A gave stereoisomeric mixture 130 (1.9 mg, 38%) as a white powder: 1 H NMR (400 MHz, DMSO) δ 8.63 (s, 1H), 8.55 (s, 1H), 8.07 (d, J = 8.1 Hz, 2H), 7.98 – 7.92 (m, 3H), 7.88 (d, J = 8.2 Hz, 2H), 7.66 (d, J = 8.0 Hz, 2H), 7.50 (d, J = 7.9 Hz, 2H), 7.41 – 7.30 (m, 4H), 7.24 (t, J = 7.0 Hz, 1H), 5.13 (s, 1H), 3.58 (s, 2H), 2.93 (d, J = 9.6 Hz, 2H), 2.58 – 2.52 (m, 2H), 2.48 – 2.42 (m, 2H), 1.74 (q, J = 5.2 Hz, 2H), 1.46 – 1.39 (m, 2H). HRMS m/z [M+H] + for C38H33NO3F3 calculated 608.2413, found 608.2421. 4-(4-((1R,5S,8r)-8-Hydroxy-3-azabicyclo[3.2.1]octan-8-yl)phe nyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (131). Pd/C (5%, 30 mg) was added to a solution of 130 (16.5 mg, 0.027 mmol) in DMF (5 mL). The resulting mixture was stirred under H2 atmosphere (70 psi) at rt for 4 h. The Pd/C was filtered. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 35% → 55% A in 40 min, flow rate = 5 mL/min, tR = 26 min) to give 131 as a white powder (12 mg, 85%): 1 H NMR (400 MHz, MeOD) δ 8.77 (d, J = 1.7 Hz, 1H), 8.45 (d, J = 1.9 Hz, 1H), 8.08 – 7.97 (m, 4H), 7.94 (dd, J = 8.9, 1.9 Hz, 1H), 7.82 (d, J = 8.2 Hz, 2H), 7.77 (d, J = 8.2 Hz, 2H), 7.61 (d, J = 8.2 Hz, 2H), 3.93 (d, J = 12.0 Hz, 2H), 3.23 (dd, J = 12.4, 3.0 Hz, 2H), 2.84 (s, 2H), 1.94 – 1.80 (m, 4H). HRMS m/z [M+H] + for C31H27NO3F3 calculated 518.1943, found 518.1942. 4-(4-((1R,5S,8r)-8-Hydroxy-3-methyl-3-azabicyclo[3.2.1]octan -8-yl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (132). Pd/C (5%, 3 mg) was added to a solution of 130 (2.8 mg, 0.0046 mmol) in MeOH (1 mL). The resulting mixture was stirred under H2 atmosphere (125 psi) at rt overnight. The Pd/C was filtered. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, tR = 24 min) to give 132 as a white powder (0.46 mg, 19%): 1 H NMR (400 MHz, MeOD) δ 8.78 (d, J = 1.5 Hz, 1H), 8.46 (d, J = 1.9 Hz, 1H), 8.08 – 7.99 (m, 4H), 7.95 (dd, J = 8.9, 2.0 Hz, 1H), 7.83 (d, J = 8.2 Hz, 2H), 7.78 (d, J = 8.1 Hz, 2H), 7.62 (d, J = 8.1 Hz, 2H), 3.88 (d, J = 11.9 Hz, 2H), 3.50 – 3.42 (m, 2H), 2.98 (s, 3H), 2.86 (s, 2H), 1.96 – 1.82 (m, 4H). HRMS m/z [M+H] + for C 32 H 29 NO 3 F 3 calculated 532.2100, found 532.2110. 4-(4-(3-Benzyl-3-azabicyclo[3.2.1]octan-8-yl)phenyl)-7-(4-(t rifluoromethyl)phenyl)- 2-naphthoic acid (133s). Treatment of 174 (5.3 mg, 0.0085 mmol) by using Procedure A gave 133s (2.3 mg, 45%) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C 38 H 33 NO 2 F 3 calculated 592.2458, found 582.2464. 4-(4-(3-Azabicyclo[3.2.1]octan-8-yl)phenyl)-7-(4-(trifluorom ethyl)phenyl)-2- naphthoic acid (133). Pd/C (5%, 1 mg) was added to a solution of 133s (1.1 mg, 0.0018 mmol) in MeOH/THF (1:1, 1 mL). The resulting mixture was stirred under H 2 atmosphere (100 psi) at rt overnight. The Pd/C was filtered. Volatiles were evaporated, and the residue was purified by RP- HPLC (C18, A: ACN, B: 10 mM TEAA, 35% → 55% A in 40 min, flow rate = 5 mL/min, tR = 26 min) to give 133 as a white powder (0.62 mg, 69%): 1 H NMR (400 MHz, MeOD) δ 8.76 (s, 1H), 8.45 (d, J = 2.0 Hz, 1H), 8.07 (d, J = 8.8 Hz, 1H), 8.03 – 7.98 (m, 3H), 7.94 (dd, J = 8.9, 1.9 Hz, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.65 – 7.58 (m, 4H), 3.42 – 3.36 (m, 2H), 3.36 – 3.32 (m, 2H), 3.09 (d, J = 12.5 Hz, 5H), 2.28 (t, J = 9.7 Hz, 2H), 2.02 (dd, J = 7.1, 1.8 Hz, 2H). HRMS m/z [M+H] + for C 31 H 27 NO 2 F 3 calculated 502.1994, found 502.1995. 4-(4-((1S,2S,4S)-4-Amino-2-(hydroxymethyl)cyclopentyl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (134). Treatment of 185 (16 mg, 0.03 mmol) by using Procedure B gave 134 (10 mg, 66%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.57 (s, 1H), 8.34 (s, 1H), 8.01 (d, J = 2.5 Hz, 1H), 7.95 (t, J = 9.1 Hz, 3H), 7.76 (dd, J = 9.5, 4.5 Hz, 3H), 7.47 – 7.36 (m, 4H), 3.87 (d, J = 8.4 Hz, 1H), 3.64 (d, J = 10.8 Hz, 1H), 3.52 (d, J = 10.6 Hz, 1H), 2.54 (dd, J = 13.9, 7.1 Hz, 1H), 2.43 – 2.18 (m, 3H), 1.74 (q, J = 6.8 Hz, 1H). HRMS m/z [M+H] + for C30H27NO3F3 calculated 506.1943, found 506.1948. 4-(4-((1R,2R,4R)-4-Amino-2-(hydroxymethyl)cyclopentyl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (135). Treatment of 301 (18.9 mg, 0.035 mmol) by using Procedure B gave 135 (12.5 mg, 71%) as a white powder: 1 H NMR (400 MHz, methanol- d4) δ 8.57 (s, 1H), 8.34 (s, 1H), 8.01 (d, J = 2.2 Hz, 1H), 7.94 (t, J = 9.7 Hz, 3H), 7.76 (t, J = 6.9 Hz, 3H), 7.46 – 7.41 (m, 2H), 7.38 (d, J = 8.0 Hz, 2H), 3.87 (d, J = 7.5 Hz, 1H), 3.63 (d, J = 10.8 Hz, 1H), 3.52 (dd, J = 11.0, 5.2 Hz, 1H), 3.23 (q, J = 9.5 Hz, 1H), 2.55 (dt, J = 14.7, 7.8 Hz, 1H), 2.41 – 2.19 (m, 3H), 1.73 (dt, J = 13.8, 7.1 Hz, 1H). HRMS m/z [M+H] + for C30H27NO3F3 calculated 506.1943, found 506.1948. 4-(4-((1S,2R,4R)-2-Amino-4-(hydroxymethyl)cyclopentyl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (136). Treatment of 190 (12 mg, with impurity TEA) by using Procedure B gave 136 (2.5 mg, overall yield% from 188: 45%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.56 (s, 1H), 8.33 (d, J = 2.0 Hz, 1H), 8.00 (d, J = 1.6 Hz, 1H), 7.97 (s, 3H), 7.76 (dd, J = 8.7, 7.0 Hz, 3H), 7.50 (d, J = 8.3 Hz, 2H), 7.47 (d, J = 8.3 Hz, 2H), 3.74 – 3.63 (m, 1H), 3.63 – 3.58 (m, 2H), 3.14 (q, J = 9.4 Hz, 1H), 2.58 – 2.40 (m, 2H), 2.18 – 2.02 (m, 2H), 1.56 (dt, J = 12.4, 8.9 Hz, 1H). HRMS m/z [M+H] + for C 30 H 27 NO 3 F 3 calculated 506.1943, found 506.1949. 4-(4-((1R,2S,4S)-2-Amino-4-(hydroxymethyl)cyclopentyl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (137). Treatment of 306 (19.2 mg, with 20% TEA; 0.03 mmol) by using Procedure B gave 137 (7.7 mg, 50%) as a white powder: 1 H NMR (400 MHz, methanol-d4) δ 8.64 (s, 1H), 8.37 (d, J = 2.0 Hz, 1H), 8.01 – 7.93 (m, 4H), 7.80 (td, J = 6.6, 3.1 Hz, 3H), 7.51 (s, 4H), 3.79 (td, J = 9.4, 6.8 Hz, 1H), 3.66 – 3.57 (m, 2H), 3.29 – 3.23 (m, 1H), 2.63 – 2.45 (m, 2H), 2.26 – 2.05 (m, 2H), 1.70 – 1.58 (m, 1H). HRMS m/z [M+H] + for C30H27NO3F3 calculated 506.1943, found 506.1943. 4-(4-((1S,2S,4S)-4-Amino-2-carboxycyclopentyl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (138). Treatment of 186 (10.3 mg, 0.019 mmol) by using Procedure B (reaction time, overnight; RP-HPLC, A: ACN, B: 10 mM TEAA, 10% → 50% A in 40 min, flow rate = 5 mL/min, tR = 39 min) gave 138 (7.5 mg, 77%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.57 (d, J = 1.6 Hz, 1H), 8.34 (d, J = 1.9 Hz, 1H), 8.01 (d, J = 1.6 Hz, 1H), 8.00 – 7.92 (m, 3H), 7.82 – 7.74 (m, 3H), 7.46 (s, 4H), 3.95 (dq, J = 6.2, 3.2, 2.8 Hz, 1H), 3.79 – 3.68 (m, 1H), 3.05 – 2.96 (m, 1H), 2.55 – 2.36 (m, 2H), 2.35 – 2.23 (m, 1H), 2.12 – 2.04 (m, 1H). HRMS m/z [M+H] + for C 30 H 25 NO 4 F 3 calculated 520.1736, found 520.1733. 4-(4-((1R,2R,4R)-4-Amino-2-carboxycyclopentyl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (139). Treatment of 302 (10.6 mg, 0.02 mmol) by using Procedure B (reaction time, overnight; RP-HPLC, A: ACN, B: 10 mM TEAA, 10% → 50% A in 40 min, flow rate = 5 mL/min, tR = 39 min) gave 139 (8.1 mg, 78%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.58 (s, 1H), 8.35 (s, 1H), 8.04 – 7.95 (m, 4H), 7.85 – 7.76 (m, 3H), 7.51 – 7.46 (m, 4H), 3.95 (s, 1H), 3.79 – 3.70 (m, 1H), 3.02 (s, 1H), 2.55 – 2.37 (m, 2H), 2.36 – 2.24 (m, 1H), 2.09 (d, J = 14.5 Hz, 1H). HRMS m/z [M+H] + for C30H25NO4F3 calculated 520.1736, found 520.1742. 4-(4-((1S,2R,4R)-2-Amino-4-carboxycyclopentyl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (140). Treatment of 191 (5.3 mg, 0.01 mmol) by using Procedure B (reaction time, overnight; RP-HPLC, A: ACN, B: 10 mM TEAA, 10% → 50% A in 40 min, flow rate = 5 mL/min, t R = 39 min) gave 140 (3.8 mg, 73%) as a white powder: 1 H NMR (400 MHz, methanol-d 4 ) δ 8.59 (s, 1H), 8.35 (d, J = 2.0 Hz, 1H), 8.03 – 7.94 (m, 4H), 7.84 – 7.75 (m, 3H), 7.52 (d, J = 8.1 Hz, 2H), 7.48 (d, J = 8.1 Hz, 2H), 3.84 – 3.75 (m, 1H), 3.43 (q, J = 8.2 Hz, 1H), 3.15 – 3.08 (m, 1H), 2.58 (ddd, J = 11.7, 8.2, 3.0 Hz, 1H), 2.45 (dt, J = 15.0, 7.7 Hz, 1H), 2.26 (dt, J = 13.6, 9.1 Hz, 1H), 2.13 (dt, J = 13.9, 4.3 Hz, 1H). HRMS m/z [M+H] + for C 30 H 25 NO 4 F 3 calculated 520.1736, found 520.1733. 4-(4-((1R,2S,4S)-2-Amino-4-carboxycyclopentyl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (141). Treatment of 307 (5.5 mg, 0.01 mmol) by using Procedure B (reaction time, overnight; RP-HPLC, A: ACN, B: 10 mM TEAA, 10% → 50% A in 40 min, flow rate = 5 mL/min, t R = 39 min) gave 141 (2.8 mg, 54%) as a white powder: 1 H NMR (400 MHz, methanol-d4) δ 8.60 (s, 1H), 8.36 (d, J = 2.0 Hz, 1H), 8.03 – 7.94 (m, 4H), 7.85 – 7.75 (m, 3H), 7.53 (d, J = 8.0 Hz, 2H), 7.48 (d, J = 8.1 Hz, 2H), 3.79 (q, J = 7.0 Hz, 1H), 3.42 (q, J = 8.0 Hz, 1H), 3.18 – 3.09 (m, 1H), 2.63 – 2.53 (m, 1H), 2.45 (dt, J = 14.9, 7.8 Hz, 1H), 2.26 (dt, J = 13.6, 9.1 Hz, 1H), 2.18 – 2.10 (m, 1H). HRMS m/z [M+H] + for C30H25NO4F3 calculated 520.1736, found 520.1730. 4-(4-Bromophenyl)-3-methylpiperidine (143). Procedure C. Bromine (0.5 mL) was added into a solution of 3-methyl-4-phenylpiperidine (142, 40 μL, 43.2 mg, 0.24 mmol) in acetic acid (1 mL). The resulting solution was stirred at rt for 10 min and then quenched with 2% aqueous sodium bisulfite solution to remove all the excess bromine added. The residue was basified with 6 N NaOH aqueous solution and extracted with dichloromethane three times . The organic layer was dried over anhydrous Na2SO4. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 0.2% TFA, 30% → 60% A in 40 min, flow rate = 5 mL/min, t R = 25 min) to give 143 (18.6 mg, 30%) as clear solid. The 1 H NMR spectrum with two groups of signals showed that the product is a mixture of diastereomers: 1 H NMR (400 MHz, CDCl3) δ 7.42 (d, J = 7.7 Hz, 2H), 7.06 (d, J = 8.1 Hz, 0.8H), 7.02 (d, J = 8.1 Hz, 1.2H), 3.48 (s, 0.6H), 3.38 (s, 0.4H), 3.27 (s, 0.6H), 3.17 (s, 0.6H), 3.07 – 2.54 (m, 3H), 2.49 (dd, J = 13.7, 9.0 Hz, 1H), 2.20 – 1.93 (m, 2H), 1.07 (d, J = 5.7 Hz, 3H). MS m/z [M+H] + for C12H17N 79 Br calculated 254.0, found 254.1. 4-(4-Bromophenyl)-2-methylpiperidine (145). Treatment of 2-methyl-4- phenylpiperidine (144, 40 μL, 43.2 mg, 0.24 mmol) with bromine by using Procedure C gave 145 (36.3 mg, 62%) as clear solid. The 1 H NMR spectrum with two groups of signals showed that the product is a mixture of diastereomers: 1 H NMR (400 MHz, CDCl 3 ) δ 7.51 – 7.41 (m, 2H), 7.14 – 7.05 (m, 2H), 3.77 – 3.72 (m, 0.3H), 3.51 (d, J = 12.5 Hz, 0.7H), 3.28 – 3.21 (m, 1H), 3.09 – 2.96 (m, 1.3H), 2.83 – 2.71 (m, 0.7H), 2.29 – 2.18 (m, 0.3H), 2.11 – 1.92 (m, 2.7H), 1.91 – 1.76 (m, 1H), 1.48 (d, J = 6.6 Hz, 1H), 1.40 (d, J = 6.3 Hz, 2H). MS m/z [M+H] + for C 12 H 17 N 79 Br calculated 254.0, found 254.1. rac-(1R,4R,5R)-5-(4-Bromophenyl)-2-azabicyclo[2.2.1]heptane (147). Treatment of 5-phenyl-2-azabicyclo[2.2.1]heptane 112a (38.9 mg, 0.22 mmol) with bromine by using Procedure C gave racemate 147 as clear solid (32.6 mg, 59%): 1 H NMR (400 MHz, MeOD) δ 7.48 – 7.38 (m, 2H), 7.20 – 7.14 (m, 2H), 3.61 (s, 1H), 3.00 – 2.89 (m, 2H), 2.78 (dd, J = 9.9, 1.5 Hz, 1H), 2.58 – 2.52 (m, 1H), 2.09 (ddd, J = 12.2, 9.1, 2.6 Hz, 1H), 1.77 (ddd, J = 13.3, 5.4, 3.0 Hz, 1H), 1.73 – 1.66 (m, 1H), 1.56 – 1.47 (m, 1H). MS m/z [M+H] + for C 12 H 15 N 79 Br calculated 252.0, found 252.0. 6-(4-Bromophenyl)-3-azabicyclo[4.1.0]heptane (149). A 25 mL round-bottom flask was flamed dried under argon and charged with 4-(4-bromophenyl)-1,2,3,6-tetrahydropyridine (148, 95.2 mg, 0.4 mmol) and CH 2 Cl 2 (4 mL). Into the solution cooled in ice bath was added diethylzinc (1M in hexane, 1 mL, 1 mmol). The mixture was stirred at 0 o C for 30 min and then diiodomethane (161.1 μL, 535.7 mg, 2 mmol) was added. The resulting mixture was stirred at 0 o C for 1.5 h and then at rt overnight. The reaction was quenched with saturated NH 4 Cl solution and then basified with NaHCO3 solution and extracted with CH2Cl2 three times. The combined organic layer was dried with Na 2 SO 4 . The volatiles were evaporated, and the residue was column chromatographed (CH 2 Cl 2 /MeOH/TEA, 100:0:1→95:5:1) to give 149 as transparent oil (62.8 mg, 62%): 1 H NMR (400 MHz, CDCl3) δ 7.38 (d, J = 8.1 Hz, 2H), 7.20 (d, J = 8.1 Hz, 2H), 5.96 (s, 1H), 3.57 (dd, J = 13.1, 6.5 Hz, 1H), 3.12 (d, J = 13.2 Hz, 1H), 3.07 – 2.92 (m, 1H), 2.77 – 2.66 (m, 1H), 2.26 – 2.05 (m, 2H), 1.33 (q, J = 7.3, 5.7 Hz, 1H), 1.06 (dd, J = 9.3, 5.2 Hz, 1H), 0.92 (t, J = 5.5 Hz, 1H). HRMS m/z [M+H] + for C12H15 79 BrN calculated 252.0388, found 252.0387. N-Boc-7,7-difluoro-6-(4-bromophenyl)-3-azabicyclo[4.1.0]hept ane (151). To a solution of 150 (169 mg, 0.5 mmol) in THF (3 mL) in a pressure tube was added NaI (37.5 mg, 0.25 mmol) and TMSCF3 (369 μl, 710 mg, 2.5 mmol). The reaction vessel was sealed and heated at 65 o C overnight. The volatiles were evaporated, and the residue was column chromatographed (hexane/EtOAc, 90:10→85:15) to give 151 as a white solid (50.5 mg, 26%): 1 H NMR (400 MHz, CDCl3) δ 7.48 (d, J = 8.3 Hz, 2H), 7.15 (d, J = 8.3 Hz, 2H), 4.26-4.09 (m, 1H), 3.99-3.66 (m, 1H), 3.55-3.42 (m, 1H), 3.37-3.12 (m, 2H), 2.29-2.09 (m, 1H), 2.08-1.89 (m, 1H), 1.48 (s, 9H); 19 F NMR (376 MHz, CDCl 3 ) δ -129.24 (dd, J = 153.0, 86.7 Hz, 1F), -145.25 (d, J = 154.0 Hz, 1F). HRMS m/z [M-t-butyl+2H] + for C13H13 79 BrF2NO2 calculated 332.0092, found 332.0103. 6-(4-Bromophenyl)-7,7-difluoro-3-azabicyclo[4.1.0]heptane (152). A TFA solution in THF (67%, 1.5 mL) was added to 151 (12.1 mg, 0.031 mmol) and the resulting mixture was stirred at rt for 0.5 h. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 0.2% TFA, 30% → 60% A in 40 min, flow rate = 5 mL/min, tR = 28 min) to give 152 as clear solid (6.7 mg, 75%): 1 H NMR (400 MHz, CDCl 3 ) δ 7.52 (d, J = 6.8 Hz, 2H), 7.35 (d, J = 7.7 Hz, 2H), 4.08 – 3.72 (m, 1H), 3.42 – 3.12 (m, 2H), 3.04 – 2.80 (m, 1H), 2.53 – 2.12 (m, 3H). HRMS m/z [M+H] + for C12H13 79 BrF2N calculated 288.0199, found 288.0201. N-Boc-4-(4-bromophenyl)-2-methoxypiperidine (154). Into an ElectraSyn vial (20 mL, IKA, Wilmington, NC) equipped with two Pt plate electrodes without diaphragm or reference electrode was added a solution of N-Boc-4-(4-bromophenyl)-piperidine 153 (625 mg, 1.84 mmol) and tetraethylammonium p-toluenesulfonate (200 mg, 0.667 mmol) in MeOH (10 mL). The solution was stirred at 0 o C while a constant voltage of 4.5 V was applied to the electrodes using an ElectraSyn 2.0 Electrochemistry Kit. After 2F/mol of electricity was passed through the solution, MeOH was removed. The residue was partitioned between dichloromethane and aqueous NaHCO 3. The organic layer was dried over anhydrous Na 2 SO 4 . The volatiles were evaporated to give crude 154 as a white powder (646 mg, 95%): 1 H NMR (400 MHz, CDCl3) δ 7.45 – 7.37 (m, 2H), 7.07 (d, J = 8.2 Hz, 2H), 5.56 – 5.51 (m, 0.4H), 5.39 (s, 0.6H), 4.16 – 4.07 (m, 0.6H), 4.00 – 3.91 (m, 0.4H), 3.27 (s, 3H), 3.15 – 2.95 (m, 2H), 2.07 – 1.96 (m, 1H), 1.85 –1.56 (m, 3H), 1.49 (s, 9H). N-Boc-4-(4-bromophenyl)-1,2,3,4-tetrahydropyridine (155). The crude 154 (626.2 mg, 1.69 mmol) was mixed well with ammonium chloride (45.2 mg, 0.84 mmol) in a flask and dried under high vacuum for 2 h. The flask was filled with argon. The resulting mixture was stirred under vacuum in a 110 o C oil bath for 5 h. The residue was column chromatographed (hexane/ethyl acetate = 100:0 → 90:10) to give 155 as a white powder (419.4 mg, 73%): 1 H NMR (400 MHz, CDCl 3 ) δ 7.48 – 7.38 (m, 2H), 7.11 (d, J = 8.1 Hz, 2H), 7.10 – 7.04 (m, 0.4H), 6.94 (d, J = 8.5 Hz, 0.6H), 4.95 – 4.88 (m, 0.4H), 4.84 – 4.76 (m, 0.6H), 3.66 – 3.41 (m, 3H), 2.14 – 2.07 (m, 1H), 1.81 – 1.70 (m, 1H), 1.51 (s, 9H). HRMS m/z [M - t-butyl+2H] + for C 12 H 13 N 81 BrO 2 calculated 284.0109, found 284.0113. rac-(1S,5R,6S)-5-(4-Bromophenyl)-2-azabicyclo[4.1.0]heptane (156) and rac- (1R,5R,6R)-5-(4-bromophenyl)-2-azabicyclo[4.1.0]heptane (157). A 10 mL round-bottom flask was flamed dried under argon and charged with 155 (72 mg, 0.213 mmol) and CH 2 Cl 2 (1.5 mL). Into the solution cooled in ice bath was added diethylzinc (1M in hexane, 0.532 mL, 0.532 mmol). The mixture was stirred at 0 o C for 30 min and then diiodomethane (85.8 μL, 285.2 mg, 1 mmol) was added. The resulting mixture was stirred at 0 o C for 1.5 h and then at rt overnight. The reaction was quenched with saturated NH4Cl solution and then basified with NaHCO3 solution and extracted with CH2Cl2 three times. The combined organic layer was dried with Na 2 SO 4 . The volatiles were evaporated, and the residue was column chromatographed (CH 2 Cl 2 /MeOH/TEA, 100:0:1→90:10:1) to give 156 as transparent solid (16.9 mg, 31%) in addition to 157 (15.8 mg, 29%). 156 has: 1 H NMR (400 MHz, CDCl3) δ 7.49 – 7.44 (m, 2H), 7.29 – 7.24 (m, 2H), 3.17 (t, J = 7.3 Hz, 1H), 2.91 – 2.82 (m, 2H), 2.74 – 2.68 (m, 1H), 1.95 – 1.84 (m, 1H), 1.63 – 1.53 (m, 1H), 1.22 – 1.14 (m, 1H), 1.04 – 0.89 (m, 1H), 0.87 – 0.78 (m, 1H). HRMS m/z [M+H] + for C12H15 79 BrN calculated 252.0388, found 252.0390. 157 has: 1 H NMR (400 MHz, CDCl 3 ) δ 7.46 – 7.42 (m, 2H), 7.29 – 7.25 (m, 2H), 3.15 – 3.06 (m, 1H), 2.75 – 2.69 (m, 1H), 2.65 – 2.60 (m, 1H), 2.55 – 2.47 (m, 1H), 1.77 – 1.68 (m, 1H), 1.46 – 1.37 (m, 1H), 1.05 – 0.97 (m, 1H), 0.79 – 0.72 (m, 1H), 0.48 – 0.42 (m, 1H). HRMS m/z [M+H] + for C12H15 79 BrN calculated 252.0388, found 252.0385. (1R,3r,5S)-8-Benzyl-3-(4-bromophenyl)-8-azabicyclo[3.2.1]oct an-3-ol (159). A mixture of 1,4-dibromobenzene (1.4 g, 6 mmol) and magnesium turnings (109.4 mg, 4.5 mmol) in THF (9 mL) was sonicated for 4 h. The solution was cooled to 0 o C. Into the Grignard reagent was added the solution of N-benzyl nortropinone 158 (646 mg, 3 mmol) in THF (15 mL). The resulting mixture was stirred at o C for 2 h. The residue was partitioned between ethyl acetate and aqueous NaHCO3. The organic layer was dried over anhydrous Na2SO4. Volatiles were evaporated, and the residue was column chromatographed (hexane/ethyl acetate = 100:0 → 40:60) to give 159 as a white powder (1.08 g; 42% of the desired bromo-product in addition to 58% byproduct from debromination of product), which could be used for a Suzuki coupling without further purification. 59 has 1 H NMR (400 MHz, CDCl 3 ) δ 7.54 – 7.21 (m, 9H), 3.61 (s, 2H), 3.31 – 3.25 (m, 2H), 2.34 (dd, J = 14.5, 3.8 Hz, 2H), 2.25 – 2.19 (m, 2H), 2.10 – 2.01 (m, 2H), 1.79 (d, J = 2.1 Hz, 1H), 1.75 (d, J = 2.0 Hz, 1H). HRMS m/z [M+H] + for C20H23 79 BrNO calculated 372.0963, found 372.0960. (1R,5S,8r)-3-Benzyl-8-(4-bromophenyl)-3-azabicyclo[3.2.1]oct an-8-ol (161). A mixture of 1,4-dibromobenzene (1.4 g, 6 mmol) and magnesium turnings (109 mg, 4.5 mmol) in THF (10 mL) was sonicated for 3 h. The solution was cooled to 0 o C. Into the Grignard reagent was added the solution of N-benzyl-3-azabicyclo[3.2.1]octan-8-one 160 (646 mg, 3 mmol) in THF (15 mL). The resulting mixture was stirred at 0 o C for 2 h. The residue was partitioned between ethyl acetate and aqueous NaHCO3. The organic layer was dried over anhydrous Na 2 SO 4 . Volatiles were evaporated, and the residue was column chromatographed (hexane/ethyl acetate = 90:10 → 0:100) to give 161 as a white powder (582.5 mg; 86% of the desired bromo- product in addition to 14% byproduct from debromination of product), which could be used for a Suzuki coupling without further purification. 161 has 1 H NMR (400 MHz, CDCl 3 ) δ 7.58 – 7.28 (m, 9H), 3.72 – 3.67 (m, 2H), 3.00 – 2.89 (m, 2H), 2.77 – 2.65 (m, 2H), 2.49 – 2.35 (m, 2H), 1.93 – 1.86 (m, 2H), 1.55 – 1.41 (m, 2H). HRMS m/z [M+H] + for C20H23 79 BrNO calculated 372.0963, found 372.0959. 3-Benzyl-8-(4-bromophenyl)-3-azabicyclo[3.2.1]octane (162). A solution of 161 (37 mg, 0.1 mmol) in concentrated sulfuric acid (1 mL) was stirred at 80 o C for 30 min. The reaction was cooled to rt and quenched with aqueous NaHCO3 solution. The residue was partitioned between ethyl acetate and aqueous NaHCO 3. The organic layer was dried over anhydrous Na2SO4. Volatiles were evaporated, and the residue was purified with preparative-TLC (hexane/ethyl acetate/TEA = 80:20:1) to give 162 as a white powder (5.3 mg, 15%): 1 H NMR (400 MHz, CD 2 Cl 2 ) δ 7.50 – 7.13 (m, 9H), 3.31 (s, 2H), 2.97 (t, J = 4.4 Hz, 1H), 2.67 – 2.59 (m, 2H), 2.45 (dd, J = 10.8, 3.8 Hz, 2H), 2.35 (d, J = 10.8 Hz, 2H), 2.05 – 1.95 (m, 2H), 1.90 – 1.81 (m, 2H). HRMS m/z [M+H] + for C20H23 79 BrN calculated 356.1014 found 356.1014. Ethyl 4-(4-(3-methylpiperidin-4-yl)phenyl)-7-(4-(trifluoromethyl)p henyl)-2- naphthoate (165). Procedure D. A flame-dried microwave tube (5 mL) was charged with argon, 164 (20 mg, 0.042 mmol), 143 (10.6 mg, 0.042 mmol), tetrakis(triphenylphosphine)palladium(0) (9.6 mg, 0.0084 mmol), potassium carbonate (17.4 mg, 0.126 mmol) and DMF (1 mL). The resulting mixture was degassed with argon for 10 min and stirred at 150 o C for 30 min in microwave. The volatiles were evaporated, and the residue was column chromatographed (CH 2 Cl 2 /MeOH/TEA, 100:0:1→90:10:1) to give 165 (13.2 mg, 61%) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C 32 H 31 NO 2 F 3 calculated 518.2307, found 518.2306. Ethyl 4-(4-(2-methylpiperidin-4-yl)phenyl)-7-(4-(trifluoromethyl)p henyl)-2- naphthoate (166): Treatment of 145 (10.6 mg, 0.042 mmol) by using Procedure D gave 166 (11 mg, 50%) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C 32 H 31 NO 2 F 3 calculated 518.2307, found 518.2307. rac-Ethyl 4-(4-((1S,4S,5S)-2-azabicyclo[2.2.1]heptan-5-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoate (167). Treatment of 147 (8 mg, 0.032 mmol) by using Procedure D gave 167 (9.3 mg, 43%) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C32H29NO2F3 calculated 516.2150, found 516.2156. Ethyl 4-(4-(3-azabicyclo[4.1.0]heptan-6-yl)phenyl)-7-(4-(trifluoro methyl)phenyl)-2- naphthoate (168). Treatment of 149 (10.6 mg, 0.042 mmol) by using Procedure D gave 168 (8 mg, 37%): 1 H NMR (400 MHz, CDCl3) δ 8.69 – 8.64 (m, 1H), 8.22 (d, J = 1.9 Hz, 1H), 8.06 – 8.00 (m, 2H), 7.82 (d, J = 8.1 Hz, 2H), 7.79 – 7.71 (m, 3H), 7.49 (d, J = 8.3 Hz, 2H), 7.45 (d, J = 8.3 Hz, 2H), 4.45 (q, J = 7.1 Hz, 2H), 3.68 – 3.56 (m, 1H), 3.23 – 3.13 (m, 1H), 3.05 – 2.98 (m, 1H), 2.82 – 2.65 (m, 1H), 2.35 (ddd, J = 13.6, 7.9, 5.3 Hz, 1H), 2.24 (dt, J = 14.4, 5.6 Hz, 1H), 1.54 – 1.39 (m, 4H), 1.23 – 1.09 (m, 1H), 1.00 (t, J = 5.4 Hz, 1H). HRMS m/z [M+H] + for C 32 H 29 NO 2 F 3 calculated 516.2150, found 516.2158. Ethyl 4-(4-(7,7-difluoro-3-azabicyclo[4.1.0]heptan-6-yl)phenyl)-7- (4- (trifluoromethyl)phenyl)-2-naphthoate (169). Treatment of 152 (6.7 mg, 0.023 mmol) by using Procedure D. 169 (5.6 mg, 44%) of sufficient purity to be used in next step: 1 H NMR (400 MHz, CDCl 3 ) δ 8.71 (d, J = 1.7 Hz, 1H), 8.25 (d, J = 1.9 Hz, 1H), 8.06 (d, J = 2.0 Hz, 1H), 7.86 – 7.76 (m, 4H), 7.72 – 7.66 (m, 4H), 7.52 – 7.48 (m, 2H), 4.48 (q, J = 7.1 Hz, 2H), 3.51 – 3.41 (m, 1H), 3.37 – 3.29 (m, 1H), 2.93 – 2.75 (m, 2H), 2.34 – 2.22 (m, 1H), 2.02 – 1.97 (m, 2H), 1.47 (t, J = 7.1 Hz, 3H). HRMS m/z [M+H] + for C 32 H 27 NO 2 F 5 calculated 552.1962, found 552.1962. rac-Ethyl 4-(4-((1S,5R,6S)-2-azabicyclo[4.1.0]heptan-5-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoate (170). A flame-dried flask (5 mL) was charged with argon, 164 (37.6 mg, 0.08 mmol), 156 (16.9 mg, 0.067 mmol), Tetrakis(triphenylphosphine)palladium(0) (15.5 mg, 0.0134 mmol), saturated sodium carbonate aqueous solution (169 μL) and DME/EtOH (85:15, 1 mL). The resulting mixture was degassed with argon for 10 min and stirred at 80 o C for 30 min. The volatiles were evaporated, and the residue was column chromatographed (CH2Cl2/MeOH/TEA, 100:0:1→95:5:1) to give 170 (9.8 mg, 25%) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C32H29NO2F3 calculated 516.2150, found 516.2148. rac-Ethyl 4-(4-((1R,5R,6R)-2-azabicyclo[4.1.0]heptan-5-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoate (170). A flame-dried flask (5 mL) was charged with argon, 164 (35.4 mg, 0.075 mmol), 157 (15.8 mg, 0.063 mmol), tetrakis(triphenylphosphine)palladium(0) (14.6 mg, 0.0126 mmol), saturated sodium carbonate aqueous solution (158 μL) and DME/EtOH (85:15, 1 mL). The resulting mixture was degassed with argon for 10 min and stirred at 80 o C for 30 min. The volatiles were evaporated, and the residue was column chromatographed (CH 2 Cl 2 /MeOH/TEA, 100:0:1→95:5:1) to give 71 (15.4 mg, 40%) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C32H29NO2F3 calculated 516.2150, found 516.2147. Ethyl 4-(4-((1R,3r,5S)-8-benzyl-3-hydroxy-8-azabicyclo[3.2.1]octan -3-yl)phenyl)-7- (4-(trifluoromethyl)phenyl)-2-naphthoate (172). Treatment of 159 (47 mg, 0.126 mmol) by using Procedure D gave 172 (61.2 mg, 76%) of sufficient purity to be used in next step: 1 H NMR (400 MHz, CDCl 3 ) δ 8.71 – 8.67 (m, 1H), 8.23 (d, J = 1.9 Hz, 1H), 8.06 (s, 1H), 7.85 – 7.79 (m, 3H), 7.79 – 7.76 (m, 2H), 7.68 (d, J = 8.2 Hz, 2H), 7.53 – 7.46 (m, 4H), 7.34 (d, J = 7.5 Hz, 2H), 7.30 – 7.18 (m, 2H), 4.48 – 4.43 (m, 2H), 3.67 (s, 2H), 3.35 (s, 2H), 2.52 (dd, J = 14.9, 3.5 Hz, 2H), 2.35 – 2.22 (m, 2H), 2.16 – 2.06 (m, 2H), 1.93 (d, J = 2.3 Hz, 1H), 1.90 (s, 1H), 1.48 – 1.44 (m, 3H). HRMS m/z [M+H] + for C 40 H 38 NO 3 F 3 calculated 636.272, found 636.2734. Ethyl 4-(4-((1R,5S,8r)-3-benzyl-8-hydroxy-3-azabicyclo[3.2.1]octan -8-yl)phenyl)-7- (4-(trifluoromethyl)phenyl)-2-naphthoate (173). Treatment of 161 (31.3 mg, 0.084 mmol) by using Procedure D gave 173 (32.8 mg, 61%) as a white powder: 1 H NMR (400 MHz, CDCl 3 ) δ 8.69 (d, J = 1.7 Hz, 1H), 8.23 (d, J = 2.0 Hz, 1H), 8.08 – 8.03 (m, 2H), 7.82 (d, J = 8.2 Hz, 2H), 7.79 – 7.72 (m, 3H), 7.67 (d, J = 8.0 Hz, 2H), 7.53 (d, J = 8.0 Hz, 2H), 7.43 (d, J = 7.5 Hz, 2H), 7.35 (t, J = 7.4 Hz, 2H), 7.31 – 7.21 (m, 1H), 4.47 (q, J = 7.1 Hz, 2H), 3.67 (s, 2H), 2.97 (d, J = 10.4 Hz, 2H), 2.71 (dd, J = 10.7, 3.5 Hz, 2H), 2.53 (s, 2H), 1.91 (q, J = 5.1 Hz, 2H), 1.80 (s, 1H), 1.62 – 1.53 (m, 2H), 1.46 (t, J = 7.1 Hz, 3H). HRMS m/z [M+H] + for C 40 H 37 NO 3 F 3 calculated 636.2726, found 636.2732. Ethyl 4-(4-(3-benzyl-3-azabicyclo[3.2.1]octan-8-yl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (174). Treatment of 162 (5.3 mg, 0.0144 mmol) by using Procedure D (column chromatography; hexane/ethyl acetate/TEA = 100:0:1 → 95:5:1) gave 174 (5.3 mg, 59%) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C40H37NO2F3 calculated 620.2771, found 620.2778. Ethyl 4-(4-(pyrrolidin-3-yl)phenyl)-7-(4-(trifluoromethyl)phenyl)- 2-naphthoate (175). Treatment of 3-(4-bromophenyl)pyrrolidine hydrochloride 163 (11 mg, 0.042 mmol) by using Procedure D gave 75 (15.7 mg, 76%) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C 30 H 27 NO 2 F 3 calculated 490.1994, found 490.1995. Regioisomeric mixture 177 of (1S,4S,5S)-5-(4-bromophenyl)-2- azabicyclo[2.2.1]heptan-3-one (180) and (1R,4R,6S)-6-(4-bromophenyl)-2- azabicyclo[2.2.1]heptan-3-one (181). Into a solution of (1R,4S)-2-azabicyclo[2.2.1]hept-5-en-3- one (176, 54.6 mg, 0.5 mmol), 1-bromo-4-iodobenzene (282.9 mg, 1 mmol), and (PPh 3 ) 2 PdCl 2 (35 mg, 0.05 mmol) in THF (3 mL) was added TEA (209 μL, 151.8 mg, 1.5 mmol) and then formic acid (22.6 μL, 27.6 mg, 0.6 mmol) dropwise. The resulting solution was stirred at 65 o C overnight. The volatiles were evaporated, and the residue was column chromatographed (hexane/ethyl acetate = 100:0 → 70:30) to give 177 as a white solid (158.7mg, quantitative): 1 H NMR (400 MHz, CDCl3) δ 7.45 – 7.39 (m, 2H), 7.15 – 7.05 (m, 2H), 6.72 (s, 0.4H), 6.58 (s, 0.6H), 4.04 – 3.98 (m, 0.6H), 3.88 – 3.83 (m, 0.4H), 3.29 – 3.16 (m, 1H), 2.83 – 2.79 (m, 1H), 2.25 (ddd, J = 12.0, 8.9, 2.5 Hz, 0.6H), 2.15 (ddd, J = 13.0, 8.8, 2.3 Hz, 0.4H), 2.05 – 1.90 (m, 2H), 1.77 – 1.66 (m, 1H). tert-Butyl (1S,4S,5S)-5-(4-bromophenyl)-3-oxo-2-azabicyclo[.2.1]heptane -2- carboxylate (178) and tert-butyl (1R,4R,6S)-6-(4-bromophenyl)-3-oxo-2- azabicyclo[2.2.1]heptane-2-carboxylate (179). To a solution of 177 (138.5 mg, 0.52 mmol), TEA (253.7 μL, 184.2 mg, 1.82 mmol), DMAP (64 mg, 0.52 mmol) in CH 2 Cl 2 (7 mL) was added (Boc) 2 O (340 mg, 1.56 mmol). The resulting solution was stirred at rt overnight. The volatiles were evaporated, and the residue was column chromatographed (hexane/EtOAc, 100:0→80:20) to give 178 (87.6, 46%) and 179 (68.5, 36%) as a white solids. 178 has: 1 H NMR (400 MHz, CDCl 3 ) δ 7.49 – 7.40 (m, 2H), 7.14 – 7.06 (m, 2H), 4.63 (t, J = 2.1 Hz, 1H), 3.37 (dd, J = 9.0, 5.2 Hz, 1H), 2.94 (q, J = 1.7 Hz, 1H), 2.38 (ddd, J = 13.3, 8.9, 2.5 Hz, 1H), 1.99 (ddt, J = 12.5, 7.5, 2.3 Hz, 2H), 1.75 (dt, J = 10.4, 1.5 Hz, 1H), 1.54 (s, 9H). HRMS m/z [M+Na] + for C 17 H 20 79 BrNO 3 Na calculated 388.0524, found 388.0528. 179 has: 1 H NMR (400 MHz, CDCl 3 ) δ 7.49 – 7.44 (m, 2H), 7.16 – 7.12 (m, 2H), 4.51 – 4.44 (m, 1H), 3.37 – 3.30 (m, 1H), 2.95 (dd, J = 4.2, 2.0 Hz, 1H), 2.26 (ddd, J = 13.4, 8.8, 2.2 Hz, 1H), 2.15 (dt, J = 13.3, 4.8 Hz, 1H), 1.90 (dp, J = 10.4, 1.9 Hz, 1H), 1.65 (dt, J = 10.4, 1.5 Hz, 1H), 1.56 (s, 9H). HRMS m/z [M+Na] + for C 17 H 20 79 BrNO 3 Na calculated 388.0524, found 388.0526. (1S,4S,5S)-5-(4-Bromophenyl)-2-azabicyclo[2.2.1]heptan-3-one (180). To a solution of 178 (559 mg, 1.53 mmol) in DCM (8 mL) was added TFA (8 mL). The resulting mixture was stirred at rt for 1 h. Toluene (1 mL) was added and the volatiles were evaporated. The residue was column chromatographed (hexane/EtOAc/TEA, 20:80:1→0:100:1) to give 180 (357 mg, 88%) as a white solid: 1 H NMR (400 MHz, CDCl 3 ) δ 7.47 – 7.39 (m, 2H), 7.16 – 7.07 (m, 2H), 6.42 (s, 1H), 4.01 (q, J = 1.9 Hz, 1H), 3.25 (dd, J = 9.1, 5.0 Hz, 1H), 2.82 (s, 1H), 2.25 (ddd, J = 12.0, 8.9, 2.5 Hz, 1H), 2.05 – 1.92 (m, 2H), 1.74 (dd, J = 9.9, 1.7 Hz, 1H). HRMS m/z [M+H] + for C12H13 79 BrNO calculated 266.0181, found 266.0178. (1R,4R,6S)-6-(4-Bromophenyl)-2-azabicyclo[2.2.1]heptan-3-one (181). To a solution of 179 (289 mg, 0.79 mmol) in DCM (4 mL) was added TFA (4 mL). The resulting mixture was stirred at rt for 1 h. Toluene (1 mL) was added and the volatiles were evaporated. The residue was column chromatographed (hexane/EtOAc/TEA, 20:80:1→0:100:1) to give 181 (200 mg, 95%) as a white solid: 1 H NMR (400 MHz, CDCl 3 ) 1 H NMR (400 MHz, CDCl 3 ) δ 7.47 – 7.38 (m, 2H), 7.18 – 7.07 (m, 2H), 3.89 – 3.83 (m, 1H), 3.24 – 3.15 (m, 1H), 2.81 – 2.75 (m, 1H), 2.19 – 1.97 (m, 2H), 1.97 – 1.87 (m, 1H), 1.75 – 1.67 (m, 1H). HRMS m/z [M+H] + for C 12 H 13 79 BrNO calculated 266.0181, found 266.0185. (1S,4S,5S)-5-(4-Bromophenyl)-2-azabicyclo[2.2.1]heptane (182) and ((1S,2S,4S)-4- amino-2-(4-bromophenyl)cyclopentyl)methanol (183). Procedure E. A dry flask was vacuumed and then then charged with argon, 180 (53.2 mg, 0.2 mmol), tris(perfluorophenyl)borane (102.4 mg, 0.2 mmol), borane ammonia complex (61.7 mg, 2 mmol), boron trifluoride diethyl etherate (24.7 µL, 28.4 mg, 0.2 mmol), 1,2-dichloroethane (4 mL). The resulting solution was stirred at 75 o C for 24 h. DCM and H 2 O were added. The mixture was basified with 6 N NaOH aqueous solution and extracted with dichloromethane three times. The organic layer was dried over anhydrous Na2SO4. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 0.2% TFA, 30% → 60% A in 40 min, flow rate = 5 mL/min) to give 182 (26.1 mg, 52%, tR = 27 min) in addition to 183 (26.2 mg, 48%, tR = 20 min). 182 has: 1 H NMR (400 MHz, methanol-d4) δ 7.46 (d, J = 8.0 Hz, 2H), 7.21 (d, J = 8.0 Hz, 2H), 4.17 (s, 1H), 3.18 – 3.13 (m, 3H), 2.79 (s, 1H), 2.33 (dd, J = 14.6, 9.2 Hz, 1H), 2.03 – 1.89 (m, 2H), 1.74 (d, J = 11.7 Hz, 1H). HRMS m/z [M+H] + for C 12 H 15 N 79 Br calculated 252.0388, found 252.0386. 183 has: 1 H NMR (400 MHz, methanol-d4) δ 7.46 – 7.40 (m, 2H), 7.19 (d, J = 8.1 Hz, 2H), 3.84 (p, J = 6.3 Hz, 1H), 3.54 (dd, J = 10.8, 3.5 Hz, 1H), 3.43 (dd, J = 10.8, 5.3 Hz, 1H), 3.14 – 3.07 (m, 1H), 2.54 – 2.45 (m, 1H), 2.30 – 2.16 (m, 3H), 1.75 – 1.62 (m, 1H). HRMS m/z [M+H] + for C 12 H 17 NO 79 Br calculated 270.0494, found 270.0493. Ethyl 4-(4-((1S,4S,5S)-2-Azabicyclo[2.2.1]heptan-5-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoate (184). Method A. Procedure F. To a vacuum dried mixture of 182 (13 mg, 0.052 mmol), 164 (24.46 mg, 0.052 mmol), Pd(PPh3)4 (6 mg, 0.0052 mmol), and Na2CO3 (8.2 mg, 0.077 mmol) was added sonicated 1,2-dimethoxyethane-water (4:1, 2 mL). The reaction mixture was degassed with argon for 10 min and then stirred at 80 °C overnight. Volatiles were evaporated, and the residue was column chromatographed (CH2Cl2/MeOH/TEA, 100:0:1→90:10:1) to give 184 (18.1 mg, 68%). Method B. A dry flask was vacuumed and then then charged with argon, 186 (18 mg, 0.034 mmol), tris(perfluorophenyl)borane (17.4 mg, 0.034 mmol), borane ammonia complex (10.0 mg, 0.34 mmol), and 1,2-dichloroethane (2 mL). The resulting solution was stirred at 75 o C for 48 h. DCM and H 2 O were added. The mixture was basified with 6 N NaOH aqueous solution and extracted with dichloromethane three times . The organic layer was dried over anhydrous Na 2 SO 4 . Volatiles were evaporated, and the residue was column chromatographed (CH2Cl2/MeOH/TEA, 100:0:1→90:10:1) to give 184 (7.87 mg, 45%) in addition to 185 (3.8, 21%). 184 has: 1 H NMR (400 MHz, CDCl 3 ) δ 8.68 (s, 1H), 8.23 (s, 1H), 8.02 – 7.94 (m, 2H), 7.85 – 7.79 (m, 3H), 7.76 (d, J = 8.3 Hz, 2H), 7.52 (d, J = 7.9 Hz, 2H), 7.31 (d, J = 8.0 Hz, 2H), 4.50 – 4.41 (m, 3H), 3.61 – 3.45 (m, 2H), 3.35 – 3.29 (m, 1H), 3.13 – 3.08 (m, 1H), 2.52 (ddd, J = 15.6, 8.7, 2.3 Hz, 1H), 2.29 – 2.16 (m, 2H), 1.97 (d, J = 12.1 Hz, 1H), 1.46 (t, J = 7.1 Hz, 3H). HRMS m/z [M+H] + for C32H29NO2F3 calculated 516.2150, found 516.2145. Ethyl 4-(4-((1S,2S,4S)-4-amino-2-(hydroxymethyl)cyclopentyl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (185). Treatment of 183 (23.4 mg, 0.087 mmol) by using Procedure F gave 185 (37.7 mg, 81%) as a white powder: 1 H NMR (400 MHz, CDCl3) δ 8.61 (s, 1H), 8.16 (s, 1H), 8.00 (d, J = 7.4 Hz, 2H), 7.83 – 7.66 (m, 5H), 7.40 (q, J = 8.0 Hz, 4H), 4.41 (q, J = 7.2 Hz, 2H), 3.83 (s, 1H), 3.72 – 3.67 (m, 1H), 3.52 (dd, J = 13.7, 6.8 Hz, 1H), 3.29 (dt, J = 24.5, 6.4 Hz, 1H), 2.74 (q, J = 7.3 Hz, 8H), 2.51 – 2.34 (m, 2H), 2.25 (t, J = 10.4 Hz, 1H), 2.12 (dt, J = 17.4, 9.2 Hz, 1H), 1.84 (d, J = 12.6 Hz, 1H), 1.41 (t, J = 7.2 Hz, 3H). HRMS m/z [M+H] + for C 32 H 31 NO 3 F 3 calculated 534.2256, found 534.2248. Ethyl 4-(4-((1S,4S,5S)-3-oxo-2-azabicyclo[2.2.1]heptan-5-yl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (186). Treatment of 180 (160 mg, 0.34 mmol) by using Procedure F (column chromatography; hexane/ethyl acetate = 50:50 → 0:100) gave 186 (168 mg, 93%) as a white powder: 1 H NMR (400 MHz, CDCl 3 ) δ 8.68 (s, 1H), 8.22 (d, J = 1.9 Hz, 1H), 8.07 – 8.00 (m, 2H), 7.81 (d, J = 8.2 Hz, 2H), 7.79 – 7.71 (m, 3H), 7.49 (d, J = 8.2 Hz, 2H), 7.43 (d, J = 8.1 Hz, 2H), 6.33 (s, 1H), 4.46 (q, J = 7.1 Hz, 2H), 4.10 (t, J = 3.5 Hz, 1H), 3.45 (dd, J = 8.9, 5.0 Hz, 1H), 2.99 (s, 1H), 2.35 (ddd, J = 11.8, 8.9, 2.4 Hz, 1H), 2.16 (ddd, J = 12.6, 5.0, 2.3 Hz, 1H), 2.11 – 2.01 (m, 1H), 1.96 – 1.89 (m, 1H), 1.45 (t, J = 7.1 Hz, 3H). HRMS m/z [M+H] + for C32H27NO3F3 calculated 530.1943, found 530.1942. (1R,4R,6S)-6-(4-Bromophenyl)-2-azabicyclo[2.2.1]heptane (187) and ((1R,3R,4S)-3- amino-4-(4-bromophenyl)cyclopentyl)methanol (188). Treatment of 181 (26.6 mg, 0.1 mmol) by using Procedure E gave 187 (6.4 mg, 25%, tR = 27 min) and 188 (3.1 mg, 12%, tR = 20 min). 87 has: 1 H NMR (400 MHz, methanol-d 4 ) δ 7.50 (dt, J = 8.3, 1.9 Hz, 2H), 7.27 – 7.18 (m, 2H), 4.08 (s, 1H), 3.30 – 3.24 (m, 1H), 3.20 (d, J = 10.8 Hz, 1H), 3.10 (d, J = 11.0 Hz, 1H), 2.85 (s, 1H), 2.16 – 2.06 (m, 1H), 2.05 – 1.92 (m, 2H), 1.73 (d, J = 11.8 Hz, 1H). HRMS m/z [M+H] + for C 12 H 15 N 79 Br calculated 252.0388, found 252.0385. 188 has: 1 H NMR (400 MHz, methanol-d 4 ) δ 7.52 (dt, J = 8.5, 2.0 Hz, 2H), 7.26 (dt, J = 8.5, 2.0 Hz, 2H), 3.64 (q, J = 8.2, 7.5 Hz, 1H), 3.56 (q, J = 2.3 Hz, 2H), 3.09 (q, J = 9.3 Hz, 1H), 2.51 – 2.36 (m, 2H), 2.14 – 2.03 (m, 1H), 1.97 (t, J = 10.8 Hz, 1H), 1.56 (td, J = 10.4, 7.3 Hz, 1H). HRMS m/z [M+H] + for C 12 H 17 NO 79 Br calculated 270.0494, found 270.0489. Ethyl 4-(4-((1R,4R,6S)-2-azabicyclo[2.2.1]heptan-6-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoate (189). Treatment of 187 (3.0 mg, 0.012 mmol) by using Procedure F gave 189 (3.5 mg, 56%) of sufficient purity to be used in next step: 1 H NMR (400 MHz, CDCl3) δ 8.67 (s, 1H), 8.21 (d, J = 1.9 Hz, 1H), 8.02 – 7.96 (m, 2H), 7.82 (d, J = 8.2 Hz, 2H), 7.79 – 7.72 (m, 3H), 7.49 – 7.43 (m, 2H), 7.37 (d, J = 8.1 Hz, 2H), 4.45 (q, J = 7.2 Hz, 2H), 4.24 (s, 1H), 3.94 (t, J = 7.4 Hz, 1H), 3.39 – 3.32 (m, 1H), 3.23 – 3.19 (m, 1H), 2.83 (s, 1H), 2.25 – 2.22 (m, 1H), 2.06 – 2.03 (m, 2H), 1.96 – 1.94 (m, 1H), 1.45 – 1.43 (m, 3H). HRMS m/z [M+H] + for C32H29NO2F3 calculated 516.2150, found 516.2141. Ethyl 4-(4-((1S,2R,4R)-2-amino-4-(hydroxymethyl)cyclopentyl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (190). Treatment of 188 (3.1 mg, 0.011 mmol) by using Procedure F gave 190 (12 mg, with TEA) of sufficient purity to be used in next step: 1 H NMR (400 MHz, CDCl 3 ) δ 8.67 (s, 1H), 8.21 (d, J = 1.9 Hz, 1H), 8.02 – 7.97 (m, 2H), 7.81 (d, J = 8.0 Hz, 2H), 7.78 – 7.73 (m, 3H), 7.47 (d, J = 8.0 Hz, 2H), 7.40 (d, J = 8.0 Hz, 2H), 4.45 (q, J = 7.1 Hz, 2H). HRMS m/z [M+H] + for C32H31NO3F3 calculated 534.2256, found 534.2263. Ethyl 4-(4-((1R,4R,6S)-3-oxo-2-azabicyclo[2.2.1]heptan-6-yl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (191). Treatment of 181 (5.3 mg, 0.02 mmol) by using Procedure F (column chromatography; hexane/ethyl acetate = 50:50 → 0:100) gave 191 (10.56 mg, 99.7%) as a white powder: 1 H NMR (400 MHz, CDCl3) δ 8.69 (d, J = 1.7 Hz, 1H), 8.24 (d, J = 2.0 Hz, 1H), 8.07 – 7.99 (m, 2H), 7.82 (d, J = 8.2 Hz, 2H), 7.80 – 7.73 (m, 3H), 7.51 (d, J = 8.3 Hz, 2H), 7.40 (d, J = 8.0 Hz, 2H), 5.91 (s, 1H), 4.47 (q, J = 7.1 Hz, 2H), 4.02 (s, 1H), 3.39 (dd, J = 8.8, 5.3 Hz, 1H), 2.91 (d, J = 3.8 Hz, 1H), 2.32 – 2.17 (m, 2H), 2.08 – 2.03 (m, 1H), 1.93 – 1.88 (m, 1H), 1.45 (t, J = 7.1 Hz, 3H). HRMS m/z [M+H] + for C 32 H 27 NO 3 F 3 calculated 530.1943, found 530.1948. Regioisomeric mixture 193 of (1R,4R,5R)-5-(4-bromophenyl)-2- azabicyclo[2.2.1]heptan-3-one (196) and (1S,4S,6R)-6-(4-bromophenyl)-2- azabicyclo[2.2.1]heptan-3-one (197). Into a solution of (1S,4R)-2-azabicyclo[2.2.1]hept-5-en-3- one (192, 327 mg, 3 mmol), 1-bromo-4-iodobenzene (1.7 g, 6 mmol), and (PPh3)2PdCl2 (210 mg, 0.3 mmol) in THF (12 mL) was added TEA (1.2 mL, 910.7 mg, 9 mmol) and then formic acid (135.7 μL, 165.6 mg, 3.6 mmol) dropwise. The resulting solution was stirred at 65 o C overnight. The volatiles were evaporated, and the residue was column chromatographed (hexane/ethyl acetate = 70:30 → 0:100) to give 193 as a white solid (798 mg, quantitative) of sufficient purity to be used in next step: HRMS m/z [M+H] + for C 12 H 13 NO 79 Br calculated 266.0181, found 266.0176. tert-Butyl (1R,4R,5R)-5-(4-bromophenyl)-3-oxo-2-azabicyclo[2.2.1]heptan e-2- carboxylate (194) and tert-butyl (1S,4S,6R)-6-(4-bromophenyl)-3-oxo-2- azabicyclo[2.2.1]heptane-2-carboxylate (195). To a solution of 193 (798 mg, 3 mmol), TEA (1.46 mL, 1 g, 10.5 mmol), DMAP (366.5 mg, 3 mmol) in CH2Cl2 (25 mL) was added (Boc)2O (1.96 g, 9 mmol). The resulting solution was stirred at rt overnight. The volatiles were evaporated, and the residue was column chromatographed (hexane/EtOAc, 100:0→80:20) to give 194 (600 mg, 55%) and 195 (291 mg, 41%) as a white solids. 94 has: 1 H NMR (400 MHz, CDCl 3 ) δ 7.31 – 7.20 (m, 2H), 6.99 – 6.92 (m, 2H), 4.51 – 4.46 (m, 1H), 3.18 (dd, J = 9.1, 5.1 Hz, 1H), 2.78 – 2.73 (m, 1H), 2.24 (ddd, J = 13.2, 8.9, 2.3 Hz, 1H), 1.83 (ddt, J = 18.8, 10.6, 2.3 Hz, 2H), 1.64 – 1.56 (m, 1H), 1.38 (s, 9H). HRMS m/z [M+Na] + for C17H20 79 BrNO3Na calculated 388.0524, found 388.0526.95 has: 1 H NMR (400 MHz, CDCl3) δ 7.41 – 7.34 (m, 2H), 7.08 (d, J = 8.5 Hz, 2H), 4.39 (d, J = 2.2 Hz, 1H), 3.31 – 3.23 (m, 1H), 2.87 (dd, J = 3.9, 2.0 Hz, 1H), 2.17 (ddd, J = 13.3, 8.7, 2.0 Hz, 1H), 2.13 – 2.01 (m, 1H), 1.86 – 1.78 (m, 1H), 1.63 – 1.55 (m, 1H), 1.49 (s, 9H). HRMS m/z [M+Na] + for C17H20 79 BrNO3Na calculated 388.0524, found 388.0526. MS m/z [M-t-butyl+2H] + for C 13 H 13 81 BrNO 3 calculated 312.0, found 312.0. (1R,4R,5R)-5-(4-Bromophenyl)-2-azabicyclo[2.2.1]heptan-3-one (196). To a solution of 194 (600 mg, 1.638 mmol) in DCM (8 mL) was added TFA (8 mL). The resulting mixture was stirred at rt for 1 h. Toluene (1 mL) was added and the volatiles were evaporated. The residue was column chromatographed (hexane/EtOAc/TEA, 30:70:0.5→0:100:0.5) to give 196 (341.5 mg, 78%) as a white solid: 1 H NMR (400 MHz, CDCl3) δ 7.47 – 7.42 (m, 2H), 7.15 – 7.11 (m, 2H), 5.78 (s, 1H), 4.02 (d, J = 2.8 Hz, 1H), 3.27 (dd, J = 9.1, 5.0 Hz, 1H), 2.84 (s, 1H), 2.26 (ddd, J = 12.0, 8.9, 2.5 Hz, 1H), 2.05 – 1.97 (m, 2H), 1.76 (dd, J = 9.9, 1.7 Hz, 1H). HRMS m/z [M+H] + for C12H13 79 BrNO calculated 266.0181, found 266.0183. (1S,4S,6R)-6-(4-Bromophenyl)-2-azabicyclo[2.2.1]heptan-3-one (197). To a solution of 195 (291 mg, 0.79 mmol) in DCM (4 mL) was added TFA (4 mL). The resulting mixture was stirred at rt for 1 h. Toluene (1 mL) was added and the volatiles were evaporated. The residue was column chromatographed (hexane/EtOAc/TEA, 30:70:0.5→0:100:0.5) to give 197 (167 mg, 79%) as a white solid: 1 H NMR (400 MHz, CDCl 3 ) δ 7.50 – 7.41 (m, 2H), 7.14 – 7.06 (m, 2H), 5.68 (s, 1H), 3.87 (s, 1H), 3.21 (dd, J = 9.0, 5.1 Hz, 1H), 2.84 (d, J = 4.0 Hz, 1H), 2.19 (ddd, J = 12.5, 8.7, 2.3 Hz, 1H), 2.07 – 1.94 (m, 2H), 1.78 – 1.70 (m, 1H). HRMS m/z [M+H] + for C 12 H 13 79 BrNO calculated 266.0181, found 266.0180. (1R,4R,5R)-5-(4-Bromophenyl)-2-azabicyclo[2.2.1]heptane (198) and ((1R,2R,4R)-4- amino-2-(4-bromophenyl)cyclopentyl)methanol (199). Treatment of 196 (53.2 mg, 0.2 mmol) by using Procedure E gave 198 (25.7 mg, 51%, t R = 27 min) and 199 (23.7 mg, 44%, t R = 20 min). 198 has: 1 H NMR (400 MHz, Methanol-d 4 ) δ 7.51 – 7.44 (m, 2H), 7.25 – 7.18 (m, 2H), 4.18 (s, 1H), 3.20 – 3.16 (m, 2H), 3.13 (t, J = 7.2 Hz, 1H), 2.80 (s, 1H), 2.36 – 2.25 (m, 1H), 2.05 – 1.91 (m, 2H), 1.73 (d, J = 11.7 Hz, 1H). HRMS m/z [M+H] + for C 12 H 15 N 79 Br calculated 252.0388, found 252.0390. 199 has 1 H NMR (400 MHz, methanol-d 4 ) δ 7.45 (d, J = 8.4 Hz, 2H), 7.24 – 7.17 (m, 2H), 3.84 (p, J = 6.5 Hz, 1H), 3.54 (dd, J = 10.7, 3.5 Hz, 1H), 3.43 (dd, J = 10.7, 5.3 Hz, 1H), 3.12 (q, J = 9.7 Hz, 1H), 2.50 (dt, J = 13.9, 7.6 Hz, 1H), 2.31 – 2.08 (m, 3H), 1.69 (ddd, J = 14.1, 8.8, 6.0 Hz, 1H). HRMS m/z [M+H] + for C 12 H 17 NO 79 Br calculated 270.0494, found 270.0491. Ethyl 4-(4-((1R,4R,5R)-2-azabicyclo[2.2.1]heptan-5-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoate (300) and ethyl 4-(4-((1R,2R,4R)-4-amino-2- (hydroxymethyl)cyclopentyl)phenyl)-7-(4-(trifluoromethyl)phe nyl)-2-naphthoate (301). Method A was used to prepare 300. Treatment of 198 (7.75 mg, 0.031 mmol) by using Procedure F gave 300 (7.6 mg, 48%) of sufficient purity to be used in next step. Also, method B was used to prepare 300 and 301. A dry flask was vacuumed and then then charged with argon, 102 (185 mg, 0.35 mmol), tris(perfluorophenyl)borane (179.2 mg, 0.35 mmol), borane ammonia complex (108 mg, 3.5 mmol), and 1,2-dichloroethane (20 mL). The resulting solution was stirred at 75 o C for 48 h. DCM and H 2 O were added. The mixture was basified with 6 N NaOH aqueous solution and extracted with dichloromethane three times. The organic layer was dried over anhydrous Na 2 SO 4 . Volatiles were evaporated, and the residue was column chromatographed (CH 2 Cl 2 /MeOH/TEA, 100:0:1→90:10:1) to give 300 (31 mg, 17%) in addition to 301 (46.7, 25%) of sufficient purity to be used in next step. 300 has: 1 H NMR (400 MHz, chloroform-d) δ 8.68 (s, 1H), 8.23 (s, 1H), 8.05 – 7.98 (m, 2H), 7.82 (d, J = 8.2 Hz, 2H), 7.80 – 7.72 (m, 3H), 7.50 (dd, J = 8.1, 2.3 Hz, 2H), 7.36 (d, J = 7.7 Hz, 2H), 4.51 – 4.41 (m, 2H), 4.18 (s, 1H), 3.45 – 3.37 (m, 1H), 3.36 – 3.19 (m, 4H), 2.98 – 2.91 (m, 2H), 2.89 (s, 1H), 2.68 (dd, J = 14.4, 9.3 Hz, 1H), 1.48 – 1.42 (m, 4H). HRMS m/z [M+H] + for C32H29NO2F3 calculated 516.2150, found 516.2142. 301 has HRMS m/z [M+H] + for C 32 H 31 NO 3 F 3 calculated 534.2256, found 534.2255. Ethyl 4-(4-((1R,4R,5R)-3-oxo-2-azabicyclo[2.2.1]heptan-5-yl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (302). Treatment of 196 (109.1 mg, 0.41 mmol) by using Procedure F (column chromatography; hexane/ethyl acetate = 50:50 → 0:100) gave 302 (211.8 mg, 98%) as a white powder: 1 H NMR (400 MHz, CDCl 3 ) δ 8.71 – 8.66 (m, 1H), 8.23 (d, J = 1.9 Hz, 1H), 8.07 – 8.00 (m, 2H), 7.83 (d, J = 8.1 Hz, 2H), 7.81 – 7.74 (m, 3H), 7.52 – 7.48 (m, 2H), 7.43 (d, J = 8.2 Hz, 2H), 5.59 (s, 1H), 4.46 (q, J = 7.1 Hz, 2H), 4.09 (s, 1H), 3.46 (dd, J = 8.7, 5.0 Hz, 1H), 3.00 (s, 1H), 2.35 (ddd, J = 11.7, 8.7, 2.3 Hz, 1H), 2.17 (ddd, J = 12.8, 5.1, 2.3 Hz, 1H), 2.11 – 2.05 (m, 1H), 1.93 (dd, J = 9.8, 1.6 Hz, 1H), 1.45 (t, J = 7.1 Hz, 3H). HRMS m/z [M+H] + for C32H27NO3F3 calculated 530.1943, found 530.1939. (1S,4S,6R)-6-(4-Bromophenyl)-2-azabicyclo[2.2.1]heptane (303) and ((1S,3S,4R)-3- amino-4-(4-bromophenyl)cyclopentyl)methanol (304). Treatment of 97 (53.2 mg, 0.1 mmol) by using Procedure E gave 303 (11.2 mg, 22%, tR = 27 min) and 304 (14.7 mg, 27%, tR = 20 min). 303 has: 1 H NMR (400 MHz, methanol-d 4 ) δ 7.53 – 7.48 (m, 2H), 7.23 – 7.18 (m, 2H), 4.08 (s, 1H), 3.26 (t, J = 7.5 Hz, 1H), 3.23 – 3.16 (m, 1H), 3.10 (d, J = 11.0 Hz, 1H), 2.85 (s, 1H), 2.11 (ddd, J = 11.6, 9.0, 2.4 Hz, 1H), 2.05 – 1.92 (m, 2H), 1.77 – 1.69 (m, 1H). HRMS m/z [M+H] + for C 12 H 15 N 79 Br calculated 252.0388, found 252.0387. 304 has 1 H NMR (400 MHz, methanol- d 4 ) δ 7.54 – 7.49 (m, 2H), 7.26 (d, J = 8.3 Hz, 2H), 3.64 (q, J = 8.7 Hz, 1H), 3.56 (d, J = 5.6 Hz, 2H), 3.09 (q, J = 9.2 Hz, 1H), 2.50 – 2.39 (m, 2H), 2.09 (ddd, J = 13.6, 8.8, 4.6 Hz, 1H), 1.96 (dt, J = 13.7, 9.5 Hz, 1H), 1.62 – 1.50 (m, 1H). MS m/z [M+H] + for C 12 H 17 NO 79 Br calculated 270.0, found 270.0. Ethyl 4-(4-((1S,4S,6R)-2-azabicyclo[2.2.1]heptan-6-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoate (305). Treatment of 303 (12.6 mg, 0.05 mmol) by using Procedure F gave 305 (23 mg, with 17% TEA; yield 74%) of sufficient purity to be used in next step: 1 H NMR (400 MHz, CDCl3) δ 8.65 (s, 1H), 8.20 (d, J = 2.0 Hz, 1H), 8.02 – 7.93 (m, 2H), 7.80 (d, J = 8.2 Hz, 2H), 7.77 – 7.69 (m, 3H), 7.46 (d, J = 7.8 Hz, 2H), 7.36 (d, J = 7.9 Hz, 2H), 4.44 (q, J = 7.3 Hz, 2H), 4.23 (s, 1H), 3.83 (t, J = 7.4 Hz, 1H), 3.34 (dt, J = 10.8, 3.1 Hz, 1H), 3.23 – 3.16 (m, 1H), 2.86 (s, 1H), 2.23 (dd, J = 13.0, 8.9 Hz, 1H), 2.11 – 1.96 (m, 3H), 1.47 – 1.41 (m, 3H). HRMS m/z [M+H] + for C32H29NO2F3 calculated 516.2150, found 516.2145. Ethyl 4-(4-((1R,2S,4S)-2-amino-4-(hydroxymethyl)cyclopentyl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (306). Treatment of 304 (16 mg, 0.06 mmol) by using Procedure F gave 306 (19.2 mg, with 20% TEA; yield 48%) of sufficient purity to be used in next step: 1 H NMR (400 MHz, CDCl 3 ) δ 8.61 (s, 1H), 8.16 (d, J = 2.0 Hz, 1H), 8.02 – 7.89 (m, 2H), 7.84 – 7.66 (m, 5H), 7.41 (s, 4H), 4.49 – 4.36 (m, 2H), 3.75 (dt, J = 17.8, 4.6 Hz, 3H), 2.70 (s, 1H), 2.51 – 2.40 (m, 1H), 2.36 – 2.16 (m, 2H), 1.98 (d, J = 15.5 Hz, 1H), 1.47 – 1.41 (m, 3H). HRMS m/z [M+H] + for C 32 H 31 NO 3 F 3 calculated 534.2256, found 534.2258. Ethyl 4-(4-((1S,4S,6R)-3-oxo-2-azabicyclo[2.2.1]heptan-6-yl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoate (307). Treatment of 197 (5.3 mg, 0.02 mmol) by using Procedure F (column chromatography; hexane/ethyl acetate = 50:50 → 0:100) gave 307 (10.9 mg, quantitative) as a white powder: 1 H NMR (400 MHz, CDCl 3 ) δ 8.69 (s, 1H), 8.24 (d, J = 1.9 Hz, 1H), 8.07 – 7.99 (m, 2H), 7.82 (d, J = 8.2 Hz, 2H), 7.80 – 7.73 (m, 3H), 7.51 (d, J = 8.1 Hz, 2H), 7.40 (d, J = 7.9 Hz, 2H), 5.84 (s, 1H), 4.47 (q, J = 7.1 Hz, 2H), 4.02 (s, 1H), 3.39 (dd, J = 8.5, 5.6 Hz, 1H), 2.94 – 2.88 (m, 1H), 2.31 – 2.17 (m, 2H), 2.08 – 2.03 (m, 1H), 1.93 – 1.88 (m, 1H), 1.46 (t, J = 7.1 Hz, 3H). HRMS m/z [M+H] + for C 32 H 27 NO 3 F 3 calculated 530.1943, found 530.1945. Ethyl 4-(4-((1S,4S,5S)-2-(methylsulfonyl)-2-azabicyclo[2.2.1]hepta n-5-yl)phenyl)-7- (4-(trifluoromethyl)phenyl)-2-naphthoate (202). Methanesulfonyl chloride (0.6 μL, 0.0075 mmol) was added into a solution of 184 (1.1 mg, 0.002 mmol) and N,N-Diisopropylethylamine (1.7 μL, 0.01 mmol) in DCM (0.5 mL). The resulting solution was stirred at rt for 3 h. Volatiles were removed and the residue was column chromatographed (hexane/EtOAc, 70:30→50:50) to give 202 (1.2 mg, quantitative): 1 H NMR (400 MHz, Chloroform-d) δ 8.69 (s, 1H), 8.23 (d, J = 1.9 Hz, 1H), 8.06 – 7.99 (m, 2H), 7.83 (d, J = 8.1 Hz, 2H), 7.80 – 7.73 (m, 3H), 7.51 – 7.46 (m, 2H), 7.38 (d, J = 8.0 Hz, 2H), 4.46 (q, J = 7.1 Hz, 2H), 4.35 (s, 1H), 3.42 (dd, J = 8.7, 3.4 Hz, 1H), 3.31 (d, J = 8.8 Hz, 1H), 3.27 – 3.19 (m, 1H), 2.92 (s, 3H), 2.90 (s, 1H), 2.59 – 2.48 (m, 1H), 2.00 – 1.87 (m, 2H), 1.73 (d, J = 10.7 Hz, 1H), 1.45 (t, J = 7.1 Hz, 3H). HRMS m/z [M+H] + for C 33 H 31 NO 4 F 3 32 S calculated 594.1926, found 594.1927. tert-Butyl (1S,4S,5S)-5-(4-bromophenyl)-2-azabicyclo[2.2.1]heptane-2-ca rboxylate (211). To a solution of 182 (23 mg, 0.09 mmol), TEA (44.5 μL, 32.3 mg, 0.32 mmol), DMAP (11 mg, 0.09 mmol) in CH 2 Cl 2 (1 mL) was added (Boc) 2 O (59 mg, 0.27 mmol). The resulting solution was stirred at rt overnight. The volatiles were evaporated, and the residue was column chromatographed (hexane/EtOAc, 100:0→80:20) to give 211 (19.3, 61%): 1 H NMR (400 MHz, Chloroform-d) δ 7.41 (d, J = 8.4 Hz, 2H), 7.07 (d, J = 8.1 Hz, 2H), 4.36 (s, 0.46H), 4.23 (s, 0.54H), 3.37 – 3.27 (m, 1H), 3.21 – 3.07 (m, 1H), 2.99 – 2.91 (m, 1H), 2.61 (s, 1H), 2.34 – 2.16 (m, 1H), 1.76 – 1.65 (m, 2H), 1.54 – 1.49 (m, 1H), 1.47 (s, 9H). HRMS m/z [M - t-butyl+2H] + for C 13 H 15 79 BrNO 2 calculated 296.0286, found 296.0284. tert-Butyl (1S,4S,5S)-5-(4-(3-(ethoxycarbonyl)-6-(4- (trifluoromethyl)phenyl)naphthalen-1-yl)phenyl)-2-azabicyclo [2.2.1]heptane-2-carboxylate (212). Treatment of 212 (19.3 mg, 0.055 mmol) by using Procedure F gave 212 (34 mg, quantitative): 1 H NMR (400 MHz, Chloroform-d) δ 8.68 (d, J = 1.7 Hz, 1H), 8.23 (d, J = 2.0 Hz, 1H), 8.08 – 8.01 (m, 2H), 7.82 (d, J = 8.1 Hz, 2H), 7.79 – 7.74 (m, 3H), 7.48 (d, J = 7.9 Hz, 2H), 7.37 (d, J = 8.0 Hz, 2H), 4.51 – 4.39 (m, 2H), 4.44 (s, 0.47H), 4.32 (s, 0.53H), 3.45 – 3.35 (m, 1H), 3.24 (dd, J = 29.8, 9.7 Hz, 1H), 3.18 – 3.10 (m, 1H), 2.77 (s, 1H), 2.45 – 2.26 (m, 1H), 1.95 – 1.84 (m, 1H), 1.84 – 1.77 (m, 1H), 1.75 – 1.68 (m, 1H), 1.52 (s, 9H), 1.45 (t, J = 7.1 Hz, 3H). 4-(4-((1S,4S,5S)-2-(tert-Butoxycarbonyl)-2-azabicyclo[2.2.1] heptan-5-yl)phenyl)-7- (4-(trifluoromethyl)phenyl)-2-naphthoic acid (213). Treatment of 212 (34 mg, 0.055 mmol) by using Procedure B gave 213 (22.7 mg, 64%) as a white solid: 1 H NMR (400 MHz, Methanol-d 4 ) δ 8.65 (d, J = 1.8 Hz, 1H), 8.29 (d, J = 1.9 Hz, 1H), 7.96 – 7.86 (m, 4H), 7.81 – 7.71 (m, 3H), 7.40 – 7.30 (m, 4H), 4.30 (d, J = 8.5 Hz, 1H), 3.39 – 3.33 (m, 1H), 3.19 (d, J = 9.6 Hz, 1H), 3.08 (t, J = 7.2 Hz, 1H), 2.69 (s, 1H), 2.31 – 2.18 (m, 1H), 1.91 – 1.82 (m, 1H), 1.82 – 1.76 (m, 1H), 1.70 – 1.60 (m, 1H), 1.50 (s, 9H). tert-Butyl (1S,4S,5S)-5-(4-(3-(hydroxycarbamoyl)-6-(4- (trifluoromethyl)phenyl)naphthalen-1-yl)phenyl)-2-azabicyclo [2.2.1]heptane-2-carboxylate (215). Carbonyldiimidazole (32 mg, 0.2 mmol) was added to a solution of 213 (5.9 mg, 0.01 mmol) in THF (1 mL). The resulting solution was stirred at rt overnight to give intermediate 214 (MS m/z [M+H] + for C 38 H 35 N 3 O 3 F 3 calculated 638.2, found 638.2). Into the reaction residue was added hydroxylamine hydrochloride (14 mg, 0.2 mmol) and the reaction mixture was stirred at rt for 2 h to give 215 (HRMS m/z [M - t-butyl+2H] + for C31H26N2O4F3 calculated 547.1845, found 547.1840) as single product. 215 was acid-labile and decomposed during column chromatography. tert-Butyl (1S,4S,5S)-5-(4-(3-((2-(dimethylamino)-2-oxoethoxy)carbonyl) -6-(4- (trifluoromethyl)phenyl)naphthalen-1-yl)phenyl)-2-azabicyclo [2.2.1]heptane-2-carboxylate (217). To a solution of 213 (164 mg, 0.280 mmol) in DMF (10 mL) was added K 2 CO 3 (116 mg, 0.840 mmol). 2-Chloro-N,N-dimethylacetamide (68 mg, 0.560 mmol) was added to the reaction mixture under N 2 atmosphere, and the reaction was stirred at 45 °C for 1 h under N 2 atmosphere. The reaction mixture was partitioned ethyl acetate (10 mL) and water (10 mL), and the aqueous phase was extracted with ethyl acetate (2 x 10 mL). The combined organic layers were dried over Na2SO4, filtered, and concentrated under pressure. The residue was purified by silica gel chromatography (hexane:ethyl acetate = 3:1) to afford compound 217 (170 mg, 90%) as a white solid; 1 H NMR (400 MHz, methanol-d4) δ 8.74 (s, 1H), 8.42 (s, 1H), 8.04 – 7.95 (m, 4H), 7.95 – 7.88 (m, 1H), 7.80 (d, J = 8.0 Hz, 2H), 7.56 – 7.46 (m, 4H), 5.06 (s, 2H), 4.25 (s, 1H), 3.35 – 3.26 (m, 3H), 2.98 (s, 6H), 2.96 (s, 1H), 2.46 – 2.35 (m, 1H), 2.18 (d, J = 15.1 Hz, 1H), 2.11 (d, J = 11.7 Hz, 1H), 1.81 (d, J = 11.8 Hz, 1H), 1.42 (s, 9H). HRMS m/z [M+H] + for C 39 H 40 N 2 O 5 F 3 calculated 673.1943, found 673.1945. 2-(Dimethylamino)-2-oxoethyl 4-(4-((1S,4S,5S)-2-azabicyclo[2.2.1]heptan-5- yl)phenyl)-7-(4-(trifluoromethyl)phenyl)-2-naphthoate (218). A TFA solution in THF (67%, 1.5 mL) was added to 217 (100.0 mg, 0.148 mmol) and the resulting mixture was stirred at rt for 0.5 h. The solvent was evaporated with toluene under reduced pressure. The residue was purified by silica gel column chromatography (dichloromethane:methanol = 95:5) to afford 218 (75 mg, 88%) the compound as a white solid. 1 H NMR (400 MHz, methanol-d4) δ 8.74 (s, 1H), 8.42 (s, 1H), 8.04 – 7.95 (m, 4H), 7.95 – 7.88 (m, 1H), 7.80 (d, J = 8.0 Hz, 2H), 7.56 – 7.46 (m, 4H), 5.06 (s, 2H), 4.25 (s, 1H), 3.35 – 3.26 (m, 3H), 2.98 (s, 6H), 2.96 (s, 1H), 2.46 – 2.35 (m, 1H), 2.18 (d, J = 15.1 Hz, 1H), 2.11 (d, J = 11.7 Hz, 1H), 1.81 (d, J = 11.8 Hz, 1H). HRMS m/z [M+H] + for C34H32N2O3F3 calculated 573.1943, found 573.1945. (7-(Diethylamino)-2-oxo-2H-chromen-4-yl)methyl 4-(4-((1S,4S,5S)-2- azabicyclo[2.2.1]heptan-5-yl)phenyl)-7-(4-(trifluoromethyl)p henyl)-2-naphthoate (220). N,N'- Dicyclohexylcarbodiimide (4.54 mg, 0.022 mmol) was added to a stirred solution of 213 (12.3 mg, 0.02 mmol), 7-(diethylamino)-4-(hydroxymethyl)coumarin (5.44 mg, 0.022 mmol), and 4- dimethylaminopyridine (2.7 mg, 0.022 mmol) in anhydrous DCM (1 mL) at 0 o C. After 10 min stirring at 0 o C, the reaction mixture was warmed to rt and stirred at rt overnight to give 219 (HRMS m/z [M - t-butyl+2H] + for C 45 H 40 N 2 O 6 F 3 calculated 761.2838, found 761.2847). Into the reaction mixture was added TFA (1 mL). The resulting mixture was stirred at rt for 1 h. Toluene (1 mL) was added to mixture and the volatiles were removed. The residue was column chromatographed (DCM/MeOH/TEA, 100:0:1→80:20:1) to give 220 (14 mg, 98%): 1 H NMR (400 MHz, Chloroform-d) δ 8.76 (s, 1H), 8.28 (d, J = 2.0 Hz, 1H), 8.09 – 8.01 (m, 2H), 7.86 – 7.80 (m, 3H), 7.76 (d, J = 7.7 Hz, 2H), 7.50 (d, J = 7.9 Hz, 2H), 7.41 – 7.36 (m, 3H), 6.61 (dd, J = 9.0, 2.6 Hz, 1H), 6.54 (d, J = 2.6 Hz, 1H), 6.32 (s, 1H), 5.55 (s, 2H), 4.14 (s, 1H), 3.42 (q, J = 7.1 Hz, 4H), 3.30 (q, J = 6.9 Hz, 2H), 3.20 (d, J = 10.3 Hz, 1H), 2.90 (d, J = 3.3 Hz, 1H), 2.68 (dd, J = 14.1, 9.2 Hz, 1H), 1.98 – 1.88 (m, 3H), 1.22 (t, J = 7.1 Hz, 6H). HRMS m/z [M+H] + for C44H40N2O4F3 calculated 717.2940, found 717.2944. EXAMPLE 2 This example demonstrates inhibition of fluorescent antagonist (2) binding in hP2Y14R-expressing CHO cells (X, Y, Z = CH, unless noted). The results are set forth in Tables 1 and 2. Table 1 Compound Structure a = IC 50 , hP2Y 14 R cLogP c 3b R NH 605±69 389 17 R-(CH2)2-CN 17.8±2.9 4.74 R 29 OH 0296±0013 444 a. p-P henyl substituted, unless noted. IC50 values are mean ± SEM for 34 separate determinations, unless noted. b. Reported in Yu et al. 1 , Jung et al. 2 and Mufti et al. 7 c. cLogP calculated using ALOGPS 2.1 program (www.vcclab.org/lab/alogps/). 57 Table 2

Solubility (µg/mL) No. R = IC 50 (nM) Log D 7 4 e cLog P c Le H 112 5.90±0.70 2.73±0.33 0.47±0.06 1.95±0.01 f 5.95 125 239±44 295±007 047±008 194±002 620

EXAMPLE 3 This Example illustrates a method of preparing compounds of formula (III). The synthesis includes a Grignard reaction of ketone starting materials for the generation of bromophenyl intermediates, Suzuki coupling of bromophenyl intermediates with boronic acid pinacol ester to obtain the phenylnaphthalenecarboxylate core structure, hydrolysis of esters to free carboxylic acid, Boc/benzyl deprotection to free amine, and conversion of analogues to their related ligands, as illustrated herein. Compound 405 with azetidine was prepared via Suzuki coupling between commercially available 3-(4-bromophenyl)azetidine 421 and naphthalenyl boronic acid pinacol ester 422 followed by ester hydrolysis. F 3 C O F 3 C O F C O OH

a Reagents and conditions: (a) 422, Na 2 CO 3 , Pd(PPh 3 ) 4 , DME-H 2 O (4:1), 80°C, overnight, 57%; (b) LiOH, THF-MeOH-H2O (3:1:1), rt, 3 h, 47%. EXAMPLE 4 This Example illustrates a method of preparation of PPTN analogues with 2- azaspiro[3.3]heptane, octahydrocyclopenta[c]pyrrole (B), 3-azabicyclo[3.1.1]heptane (C), 1- azacycloheptane (D), and azocane (E) were summarized in Scheme 2. In a one-pot reaction, treatment of ketones 424, 428, 432, 436, and 440 with in-situ generated Grignard reagent 4- bromophenylmagnesium bromide provided bromophenyl intermediates 425, 429, 433, 437, and 441, respectively. Derived from a racemic starting material 428, intermediate 429 is a racemic mixture of left-handed and right-handed helix enantiomers. Stereoselective Grignard reaction of 432 yielded enantiomer 433 with hydroxyl group and one-carbon bridge identified as trans to each other. The Suzuki coupling of naphthalenyl boronic acid pinacol ester 422 with the 4-(4- bromo-phenyl) intermediates 425, 429, 433, 437, and 441 gave their corresponding esters of analogues 426, 430, 434, 438, and 442. The ester hydrolysis of 426, 430, 434, 438, and 442 provided the derivatives 427, 431, 435, 439, and 443. Boc deprotection of 427 with TFA in DCM offered product 408 in addition to unexpected dehydrated product 407. Compound 408 was unstable in the reaction residue in the presence of methanol and partially reacted with methanol and methanol-d4 to give O-methylated ligand 409. Thus, methanol should be avoided if methylated by-product is not needed. Similarly, boc deprotection of 443 gave product 420 and dehydrated compound 419. Nevertheless, boc deprotection of 439 meanwhile dehydrated the hydroxyl group to yield alkene mixture 417. Boc deprotection of compound 435 simply gave ligand 415. Pd-catalyzed debenzylation of 431 with H 2 afforded ligand 411, which was further dehydrated by refluxing in TFA to give 412. Reduction of 407, 412, 417, and 419 by Pd-catalyzed hydrogenation offered PPTN analogues 406 with 2- azaspiro[3.3]heptane, 413 with octahydrocyclopenta[c]pyrrole (B), 416 with 1- azacycloheptane (D), and 418 with azocane (E), respectively. Stereoselective deoxygenation of 415 with sodium borohydride in sulfuric acid afforded 414. Thus, hygroscopic sulfuric acid was employed to readily generate tertiary benzylic carbocation, which was further reduced by sodium borohydride to yield deoxygenated product. Scheme. Preparation of compounds of formula (III) with 2-azaspiro[3.3]heptane (A), octahydrocyclopenta[c]pyrrole (B), 3-azabicyclo[3.1.1]heptane (C), 1-azacycloheptane (D), and azocane (E) (A) CF 3 CF 3 CF O O 3 CF O 3 O H

(C) CF 3 CF O 3 CF 3 O O B H a Reagents and conditions: (a) 4-bromophenylmagnesium bromide (in-situ generated: dibromobenzene, magnesium, THF, sonication, rt, 2 h), THF, 0 o C, 4 h; (b) boronic acid pinacol ester 422, Na 2 CO 3 , Pd(PPh 3 ) 4 , DME-H 2 O (4:1), 85 °C, overnight; (c) LiOH, THF-MeOH-H 2 O (3:1:1), rt, 3 h; (d) TFA, DCM, rt, 1 h; (e) H2, Pd/C, DMF, 3 h; (f) H2, Pd/C, DMF, 5 h; (g) TFA, 90 o C, 2 h; (h) NaBH 4 , THF, H 2 SO 4 , rt, 30 min; Chemical synthesis General Information. Materials and Methods. All chemicals and anhydrous solvents were obtained directly from commercial sources. All reactions were carried out under argon atmosphere using anhydrous solvents unless specified otherwise. Room temperature (rt) refers to 25 ± 5 °C. Silica-gel precoated with F254 on aluminum plates were used for TLC. The spots were examined under ultraviolet light at 254 nm and further visualized by anisaldehyde or cerium ammonium molybdate stain solution. Column chromatography was performed on silica gel (40−63 μm, 60 Å). NMR spectra were recorded on a Bruker 400 MHz spectrometer. Chemical shifts are given in ppm (δ), calibrated to the residual solvent signal peaks of CDCl3 (7.26 ppm), CD3OD (3.31 ppm), or DMSO-d6 (2.50 ppm) for 1 H NMR with coupling constant (J) values reported in Hz. High resolution mass (HRMS) measurements were performed on a proteomics optimized Q-TOF-2 (Micromass-Waters). The RP-HPLC was performed using Phenomenex Luna 5 μm C18(2)100A, AXIA, 21.2 mm × 250 mm column. Antagonist purity was determined as ≥ 95% using Agilent ZORBAX SB-Aq, 5 μm, 4.6 mm × 150 mm column attached to Agilent 1100 HPLC system. The HPLC traces for compounds tested in vivo (xxx) were included in Supporting Information. 4-(4-(Azetidin-3-yl)phenyl)-7-(4-(trifluoromethyl)phenyl)-2- naphthoic acid (405). Procedure A. LiOH (7.4 mg, 0.3 mmol) was added to a solution of 423 (6.25 mg, 0.013 mmol) in THF-H2O-MeOH (3:1:1, 1 mL). The resulting mixture was sonicated for 30 s and stirred at rt for 3 h. The reaction was quenched and neutralized by 1 M HCl. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, tR = 17.2 min) to give 405 as a white powder (2.73 mg, 47%): 1 H NMR (400 MHz, MeOD) δ 8.76 (s, 1H), 8.44 (d, J = 2.0 Hz, 1H), 8.04 – 7.96 (m, 4H), 7.93 (dd, J = 8.8, 1.9 Hz, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.60 (s, 4H), 4.53 – 4.31 (m, 5H). HRMS m/z [M+H] + for C 27 H 21 NO 2 F 3 calculated 448.1524, found 448.1520. 4-(4-(2-Azaspiro[3.3]heptan-6-yl)phenyl)-7-(4-(trifluorometh yl)phenyl)-2- naphthoic acid (406). Procedure B. Pd/C (10%, 3 mg) was added to a solution of 407 (0.0016 mmol) in DMF (0.3 mL). The resulting mixture was bubbled with H 2 at rt for 5 h. The Pd/C was filtered. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, t R = 20 min) to give 406 as a white powder (0.4 mg, 51%): 1 H NMR (400 MHz, MeOD) δ 8.74 (s, 1H), 8.43 (d, J = 2.0 Hz, 1H), 8.01 (dd, J = 8.7, 3.6 Hz, 3H), 7.96 – 7.89 (m, 2H), 7.81 (d, J = 8.1 Hz, 2H), 7.48 (d, J = 8.0 Hz, 2H), 7.42 (d, J = 8.0 Hz, 2H), 4.30 (s, 2H), 4.07 (s, 2H), 3.62 – 3.60 (m, 1H), 2.83 – 2.75 (m, 2H), 2.51 (td, J = 9.7, 2.9 Hz, 2H). HRMS m/z [M+H] + for C 30 H 25 NO 2 F 3 calculated 488.1837, found 488.1833. 4-(4-(2-Azaspiro[3.3]hept-5-en-6-yl)phenyl)-7-(4-(trifluorom ethyl)phenyl)-2- naphthoic acid (407), 4-(4-(6-Hydroxy-2-azaspiro[3.3]heptan-6-yl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (408), and 4-(4-(6-Methoxy-2- azaspiro[3.3]heptan-6-yl)phenyl)-7-(4-(trifluoromethyl)pheny l)-2-naphthoic acid and 4-(4-(6- (methoxy-d3)-2-azaspiro[3.3]heptan-6-yl)phenyl)-7-(4-(triflu oromethyl)phenyl)-2-naphthoic acid (6:4, 409). A TFA solution in THF (67%, 4.5 mL) was added to 427 (all residue from hydrolysis of 426, 0.057 mmol). The resulting mixture was stirred at rt for 1 h. The stir bar was washed with MeOH. Volatiles were evaporated, and the residue was dissolved in MeOD for crude NMR. MeOD was removed and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min) to give 407 (1.8 mg, 7%, tR = 42.4 min), 408 (2.54 mg, 9%, tR = 33.0 min), and 409 (10.5 mg, 36%, tR = 40.6 min).407 has: 1 H NMR (400 MHz, MeOD) δ 8.76 (s, 1H), 8.43 (d, J = 1.9 Hz, 1H), 8.04 – 7.97 (m, 4H), 7.97 – 7.91 (m, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.62 (d, J = 7.9 Hz, 2H), 7.56 (d, J = 8.0 Hz, 2H), 6.55 (s, 1H), 4.42 – 4.31 (m, 4H), 3.20 (s, 2H). HRMS m/z [M+H] + for C 30 H 23 NO 2 F 3 calculated 486.1681, found 486.1682.408 has: 1 H NMR (400 MHz, MeOD) δ 8.72 (s, 1H), 8.41 (d, J = 1.9 Hz, 1H), 7.99 (d, J = 8.7 Hz, 4H), 7.90 (dd, J = 8.9, 1.9 Hz, 1H), 7.80 (d, J = 8.1 Hz, 2H), 7.62 (d, J = 8.0 Hz, 2H), 7.54 (d, J = 7.9 Hz, 2H), 4.30 (s, 2H), 4.09 (s, 2H), 2.94 (d, J = 13.0 Hz, 2H), 2.66 (d, J = 12.5 Hz, 2H). HRMS m/z [M+H] + for C 30 H 25 NO 3 F 3 calculated 504.1787, found 504.1796.409 has: 1 H NMR (600 MHz, MeOD) δ 8.77 – 8.74 (m, 1H), 8.44 (d, J = 1.9 Hz, 1H), 8.05 – 8.00 (m, 4H), 7.94 (dd, J = 8.8, 2.0 Hz, 1H), 7.83 (d, J = 8.2 Hz, 2H), 7.63 – 7.55 (m, 4H), 4.27 (s, 2H), 4.08 (s, 2H), 3.07 (s, 2H), 2.91 – 2.84 (m, 2H), 2.80 – 2.74 (m, 2H). HRMS m/z [M+H] + for C31H27NO3F3 calculated 518.1943, found 518.1939; for C 31 H 24 D 3 NO 3 F 3 calculated 521.2117, found 521.2126. rac-4-(4-((3aR,6aR)-5-hydroxyoctahydrocyclopenta[c]pyrrol-5- yl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (411). Pd/C (10%, 53 mg) was added to a solution of 431 (10.7 mg, 0.0177 mmol) in DMF (5 mL). The resulting mixture was bubbled with H 2 at rt for 5 h. The Pd/C was filtered. The filtrate was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 30% → 50% A in 40 min, flow rate = 5 mL/min, t R = 35 min) to give 411 as a white powder (3.18 mg, 35% overall yield from 430): 1 H NMR (400 MHz, MeOD) δ 8.75 (s, 1H), 8.43 (d, J = 2.0 Hz, 1H), 8.05 – 7.96 (m, 4H), 7.93 (dd, J = 9.0, 2.0 Hz, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.69 (d, J = 8.2 Hz, 2H), 7.54 (d, J = 8.0 Hz, 2H), 3.56 – 3.46 (m, 2H), 2.98 (t, J = 11.2 Hz, 2H), 2.76 – 2.65 (m, 1H), 2.60 – 2.45 (m, 2H), 2.22 (dd, J = 12.6, 5.4 Hz, 1H), 2.06 – 1.95 (m, 2H). HRMS m/z [M+H] + for C 31 H 27 NO 3 F 3 calculated 518.1943, found 518.1944. rac-4-(4-((3aR,6aR)-1,2,3,3a,4,6a-Hexahydrocyclopenta[c]pyrr ol-5-yl)phenyl)-7- (4-(trifluoromethyl)phenyl)-2-naphthoic acid (412). A solution of 411 (0.012 mmol, crude reaction residue without HPLC purification) in TFA (2 mL) was refluxed in a 90 o C oil bath for 2 h. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 45% → 60% A in 40 min, flow rate = 5 mL/min, tR = 21 min) to give 412 as a white powder (1.9 mg, 33% overall yield from 431): 1 H NMR (400 MHz, MeOD) δ 8.75 (s, 1H), 8.43 (d, J = 2.0 Hz, 1H), 8.07 – 7.96 (m, 4H), 7.94 (dd, J = 8.9, 2.0 Hz, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.68 (d, J = 8.1 Hz, 2H), 7.53 (d, J = 8.0 Hz, 2H), 6.61 (s, 1H), 3.62 (d, J = 3.3 Hz, 1H), 3.53 (dd, J = 10.2, 6.5 Hz, 1H), 3.26 – 3.15 (m, 1H), 3.04 (d, J = 7.0 Hz, 2H), 2.99 (dd, J = 14.4, 6.0 Hz, 1H), 2.68 – 2.55 (m, 1H), 2.40 (dq, J = 11.9, 5.9 Hz, 1H). HRMS m/z [M+H] + for C 31 H 25 NO 2 F 3 calculated 500.1837, found 500.1834. rac-4-(4-((3aR,6aR)-Octahydrocyclopenta[c]pyrrol-5-yl)phenyl )-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (413). Treatment of 412 (0.021 mmol, crude reaction residue without HPLC purification) by using Procedure B gave 413 (0.57 mg, 5% overall yield from 431) as a white powder: 1 H NMR (400 MHz, MeOD) δ 8.74 (s, 1H), 8.42 (d, J = 1.9 Hz, 1H), 8.06 – 7.95 (m, 4H), 7.93 (d, J = 8.4 Hz, 1H), 7.81 (d, J = 8.0 Hz, 2H), 7.49 (s, 4H), 4.01 (d, J = 9.2 Hz, 1H), 3.48 (p, J = 6.4 Hz, 2H), 3.03 – 2.86 (m, 2H), 2.59 (dd, J = 12.8, 6.4 Hz, 1H), 2.47 – 2.29 (m, 2H), 2.17 – 1.98 (m, 2H), 1.64 (q, J = 11.0 Hz, 1H). HRMS m/z [M+H] + for C 31 H 27 NO 2 F 3 calculated 502.1994, found 502.1986. 4-(4-((1R,5S,6r)-3-Azabicyclo[3.1.1]heptan-6-yl)phenyl)-7-(4 - (trifluoromethyl)phenyl)-2-naphthoic acid (414). Concentrated sulfuric acid (95-98%, 0.9 mL) was added to a mixture of 415 (9 mg, 0.015 mmol) and NaBH 4 (5.7 mg, 0.15 mmol) in THF (0.1 mL) under argon (caution, running the reaction under argon is required to avoid fire/explosion). The resulting solution was stirred at rt for 30 min. The mixture was basified with cold 6 N NaOH aqueous solution and extracted with ethyl acetate three times. The organic layer was dried over anhydrous Na 2 SO 4 . Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 35% → 55% A in 40 min, flow rate = 5 mL/min, tR = 38.8 min) to give 414 as a white powder (0.8 mg, 11%): 1 H NMR (400 MHz, MeOD) δ 8.76 (s, 1H), 8.44 (d, J = 2.0 Hz, 1H), 8.07 – 7.96 (m, 4H), 7.96 – 7.90 (m, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.56 (d, J = 7.8 Hz, 2H), 7.38 (d, J = 7.7 Hz, 2H), 4.09 (q, J = 9.6 Hz, 1H), 3.62 (t, J = 7.2 Hz, 1H), 3.42 – 3.33 (m, 3H), 3.23 (dd, J = 16.7, 7.1 Hz, 2H), 2.74 (q, J = 9.1 Hz, 1H), 2.33 (q, J = 10.5 Hz, 1H). HRMS m/z [M+H] + for C30H24F3NO2 calculated 488.1837, found 488.1829. 4-(4-((1R,5S,6r)-6-Hydroxy-3-azabicyclo[3.1.1]heptan-6-yl)ph enyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (415). Procedure C. A TFA solution in THF (67%, 2 mL) was added to 435 (0.022 mmol, reaction residue). The resulting mixture was stirred at rt for 1 h. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 35% → 55% A in 40 min, flow rate = 5 mL/min, tR = 27.7 min) to give 415 (4.6 mg, 42%) as a white solid: 1 H NMR (400 MHz, DMSO) δ 8.76 (s, 1H), 8.65 (d, J = 2.0 Hz, 1H), 8.06 (t, J = 8.2 Hz, 3H), 7.98 (d, J = 8.9 Hz, 1H), 7.92 – 7.85 (m, 3H), 7.73 (d, J = 7.9 Hz, 2H), 7.59 (d, J = 7.8 Hz, 2H), 3.69 (d, J = 11.7 Hz, 2H), 3.47 (d, J = 11.9 Hz, 2H), 2.89 (d, J = 4.5 Hz, 2H), 1.71 (s, 2H). HRMS m/z [M+H] + for C30H25NO3F3 calculated 504.1787, found 504.1793. 4-(4-(Azepan-4-yl)phenyl)-7-(4-(trifluoromethyl)phenyl)-2-na phthoic acid (416). Treatment of 417 (0.038 mmol, crude reaction residue without HPLC purification) by using Procedure B gave 416 (4 mg, 22% overall yield from 438) as a white powder: 1 H NMR (400 MHz, MeOD) δ 8.77 – 8.72 (m, 1H), 8.42 (d, J = 1.9 Hz, 1H), 8.05 – 7.95 (m, 4H), 7.92 (dd, J = 8.9, 1.9 Hz, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.48 (q, J = 8.1 Hz, 4H), 3.54 – 3.35 (m, 4H), 3.02 (t, J = 10.9 Hz, 1H), 2.22 (d, J = 15.9 Hz, 4H), 2.09 – 1.86 (m, 2H). HRMS m/z [M+H] + for C30H27NO2F3 calculated 490.1994, found 490.1992. Mixture of 4-(4-(2,3,6,7-Tetrahydro-1H-azepin-4-yl)phenyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid and 4-(4-(2,5,6,7-tetrahydro-1H-azepin-4- yl)phenyl)-7-(4-(trifluoromethyl)phenyl)-2-naphthoic acid (417). Treatment of 439 (all residue from hydrolysis of 438, 0.057 mmol) by using Procedure C gave 417 (5.08 mg, 18%) as a mixture of two constitutional isomers (1:1): 1 H NMR (400 MHz, MeOD) δ 8.76 (d, J = 2.8 Hz, 1H), 8.43 (d, J = 2.4 Hz, 1H), 8.05 – 7.97 (m, 4H), 7.97 – 7.90 (m, 1H), 7.81 (d, J = 8.1 Hz, 2H), 7.66 – 7.49 (m, 4H), 6.35 (t, J = 6.2 Hz, 0.5H), 6.21 (t, J = 6.5 Hz, 0.5H), 3.99 (d, J = 6.5 Hz, 1H), 3.59 – 3.49 (m, 1H), 3.48 – 3.41 (m, 1H), 3.39 – 3.32 (m, 1H), 3.14 – 3.05 (m, 1H), 3.06 – 2.99 (m, 1H), 2.74 (d, J = 5.5 Hz, 1H), 2.14 (d, J = 5.7 Hz, 1H). HRMS m/z [M+H] + for C 30 H 25 NO 2 F 3 calculated 488.1837, found 488.1835. 4-(4-(1,2,3,4,7,8-Hexahydroazocin-5-yl)phenyl)-7-(4-(trifluo romethyl)phenyl)-2- naphthoic acid (419) and 4-(4-(5-hydroxyazocan-5-yl)phenyl)-7-(4-(trifluoromethyl)phe nyl)- 2-naphthoic acid (420). Treatment of 443 (all residue from hydrolysis of 442, 0.049 mmol) by using Procedure C gave 419 (4.52 mg, 18%) in addition to 420 (4.67 mg, 18%).419 has: 1 H NMR (400 MHz, MeOD) δ 8.72 (s, 1H), 8.40 (d, J = 2.0 Hz, 1H), 8.03 – 7.95 (m, 5H), 7.90 (dd, J = 9.0, 2.0 Hz, 1H), 7.78 (d, J = 8.1 Hz, 3H), 7.66 (d, J = 8.0 Hz, 3H), 7.51 (d, J = 8.0 Hz, 2H), 6.28 (t, J = 8.3 Hz, 1H), 3.35 – 3.28 (m, 2H), 3.25 – 3.21 (m, 2H), 2.90 (t, J = 6.3 Hz, 2H), 2.72 (dd, J = 12.0, 7.5 Hz, 2H), 2.08 (s, 2H). HRMS m/z [M+H] + for C31H26NO2F3 calculated 502.1994, found 502.2001.20 has: 1 H NMR (400 MHz, MeOD) δ 8.75 (s, 1H), 8.42 (d, J = 1.9 Hz, 1H), 8.00 – 7.94 (m, 4H), 7.91 (dd, J = 8.9, 1.9 Hz, 1H), 7.78 (d, J = 8.1 Hz, 2H), 7.70 (d, J = 8.4 Hz, 2H), 7.65 (d, J = 8.5 Hz, 2H), 3.85 (dt, J = 13.3, 7.0 Hz, 2H), 3.41 (dt, J = 12.5, 6.7 Hz, 2H), 2.70 (dt, J = 13.5, 6.8 Hz, 2H), 2.47 (dt, J = 13.5, 6.7 Hz, 2H), 2.35 (dp, J = 13.7, 6.8 Hz, 2H), 2.24 (dq, J = 13.5, 6.8 Hz, 2H). HRMS m/z [M – OH-] + for C 31 H 27 NO 2 F 3 calculated 502.1994, found 502.1993.Ethyl 4-(4-(azetidin-3- yl)phenyl)-7-(4-(trifluoromethyl)phenyl)-2-naphthoate (423). Procedure D. To a vacuum dried mixture of 421 (5.8 mg, 0.023 mmol), 422 (11 mg, 0.023 mmol), Pd(PPh 3 ) 4 (2.7 mg, 0.0023 mmol), and Na2CO3 (6 mg, 0.057 mmol) was added sonicated 1,2-dimethoxyethane- water (4:1, 1 mL). The reaction mixture was degassed with argon for 10 min and then stirred at 80°C overnight. Volatiles were evaporated, and the residue was column chromatographed (CH 2 Cl 2 /MeOH/TEA, 100:0:1→90:10:1) to give 423 (6.25 mg, 57%): 1 H NMR (400 MHz, CDCl3) δ 8.68 (s, 1H), 8.22 (d, J = 1.9 Hz, 1H), 8.01 (d, J = 1.7 Hz, 1H), 7.97 (d, J = 8.8 Hz, 1H), 7.86 – 7.77 (m, 3H), 7.75 (d, J = 8.3 Hz, 2H), 7.61 (d, J = 7.9 Hz, 2H), 7.55 (d, J = 8.0 Hz, 2H), 4.57 – 4.41 (m, 5H), 3.15 (q, J = 7.3 Hz, 2H), 1.44 (td, J = 7.2, 4.1 Hz, 3H). HRMS m/z [M+H] + for C 32 H 29 NO 2 F 3 calculated 516.2150, found 516.2145. HRMS m/z [M+H] + for C29H25NO2F3 calculated 476.1837, found 476.1837. tert-Butyl 6-(4-bromophenyl)-6-hydroxy-2-azaspiro[3.3]heptane-2-carboxy late (425). Procedure E. A mixture of 1,4-dibromobenzene (2.9 g, 12.5 mmol) and magnesium turnings (60.8 mg, 2.5 mmol) in THF (5 mL) was sonicated for 2 h. The solution was cooled to 0 o C. Into the Grignard reagent was added the solution of 424 (105.6 mg, 0.5 mmol) in THF (10 mL). The resulting mixture was stirred at 0 o C to rt overnight. The residue was partitioned between ethyl acetate and aqueous NaHCO3. The organic layer was dried over anhydrous Na 2 SO 4 . Volatiles were evaporated, and the residue was column chromatographed (hexane/ethyl acetate = 80:20 → 40:60) to give 425 (72.3 mg; 39%): 1 H NMR (400 MHz, CDCl 3 ) δ 7.50 – 7.44 (m, 2H), 7.29 – 7.24 (m, 2H), 4.05 (s, 2H), 3.78 (s, 2H), 2.72 – 2.66 (m, 2H), 2.52 (d, J = 12.7 Hz, 2H), 1.41 (s, 9H). HRMS m/z [M - t-butyl+2H] + for C 13 H 15 N 79 BrO 3 calculated 312.0235, found 312.0234. tert-Butyl 6-(4-(3-(ethoxycarbonyl)-6-(4-(trifluoromethyl)phenyl)naphth alen-1- yl)phenyl)-6-hydroxy-2-azaspiro[3.3]heptane-2-carboxylate (426). Treatment of 425 (25.6 mg, 0.07 mmol) by using Procedure D gave 426 (36.12 mg, 82%): 1 H NMR (400 MHz, CDCl 3 ) δ 8.68 (s, 1H), 8.22 (d, J = 1.8 Hz, 1H), 8.06 – 7.96 (m, 2H), 7.81 (d, J = 8.1 Hz, 2H), 7.76 (td, J = 6.8, 3.3 Hz, 3H), 7.57 (d, J = 8.0 Hz, 2H), 7.52 (d, J = 8.1 Hz, 2H), 4.46 (q, J = 7.1 Hz, 2H), 4.13 (s, 2H), 3.90 (s, 2H), 2.87 (d, J = 12.8 Hz, 2H), 2.64 (d, J = 12.6 Hz, 2H), 1.44 (d, J = 9.7 Hz, 12H). HRMS m/z [M+Na] + for C37H36NO5F3Na calculated 654.2443, found 654.2446. 4-(4-(2-(tert-Butoxycarbonyl)-6-hydroxy-2-azaspiro[3.3]hepta n-6-yl)phenyl)-7- (4-(trifluoromethyl)phenyl)-2-naphthoic acid (427). Treatment of 426 (36 mg, 0.057 mmol) by using Procedure A (without HPLC purification) gave 427 of sufficient purity to be used in next step: 1 H NMR (400 MHz, MeOD) δ 8.50 (s, 1H), 8.21 (s, 1H), 7.91 – 7.80 (m, 4H), 7.67 (dd, J = 14.7, 9.8 Hz, 3H), 7.52 – 7.45 (m, 2H), 7.37 (t, J = 5.3 Hz, 2H), 3.98 (s, 2H), 3.76 (s, 2H), 2.73 (d, J = 12.1 Hz, 2H), 2.52 – 2.45 (m, 2H), 1.32 (d, J = 3.8 Hz, 9H). HRMS m/z [M+Na] + for C35H32NO5F3Na calculated 626.2130, found 626.2136. rac-(3aR,6aR)-2-Benzyl-5-(4-bromophenyl)octahydrocyclopenta[ c]pyrrol-5-ol (29). Treatment of 428 (125.9 mg, 0.5 mmol) by using Procedure E (column chromatography; DCM/MeOH, 100:0→95:5) gave 429 (101.18 mg, 54%): 1 H NMR (400 MHz, CDCl3) δ 7.48 – 7.40 (m, 2H), 7.40 – 7.20 (m, 7H), 3.94 – 3.78 (m, 2H), 2.88 (dq, J = 9.6, 4.5 Hz, 2H), 2.80 (s, 1H), 2.50 (tdt, J = 16.7, 11.3, 8.9 Hz, 3H), 2.29 (dd, J = 12.7, 7.1 Hz, 1H), 2.18 (dtd, J = 16.1, 11.4, 5.6 Hz, 1H), 1.97 (dd, J = 12.6, 4.7 Hz, 1H), 1.76 – 1.61 (m, 2H). HRMS m/z [M+H] + for C20H23NO 79 Br calculated 372.0963, found 372.0966. rac-Ethyl 4-(4-((3aR,6aR)-2-benzyl-5-hydroxyoctahydrocyclopenta[c]pyrr ol-5- yl)phenyl)-7-(4-(trifluoromethyl)phenyl)-2-naphthoate (30). Treatment of 429 (26.1 mg, 0.07 mmol) by using Procedure D gave 430 (33.7 mg, 76%): 1 H NMR (400 MHz, CDCl3) δ 8.65 (s, 1H), 8.20 (d, J = 1.9 Hz, 1H), 8.04 – 7.95 (m, 2H), 7.79 (d, J = 8.2 Hz, 2H), 7.76 – 7.68 (m, 3H), 7.60 (d, J = 8.1 Hz, 2H), 7.54 (d, J = 7.4 Hz, 2H), 7.48 (d, J = 8.0 Hz, 2H), 7.36 (dt, J = 13.7, 7.2 Hz, 3H), 4.44 (q, J = 7.1 Hz, 2H), 4.14 (s, 2H), 3.32 – 3.17 (m, 2H), 2.99 – 2.76 (m, 3H), 2.57 – 2.42 (m, 2H), 2.20 (dd, J = 12.6, 5.2 Hz, 1H), 1.94 (dt, J = 17.9, 12.1 Hz, 2H), 1.44 (t, J = 7.1 Hz, 3H). HRMS m/z [M+H] + for C 40 H 37 NO 3 F 3 calculated 636.2726, found 636.2724. rac-4-(4-((3aR,6aR)-2-Benzyl-5-hydroxyoctahydrocyclopenta[c] pyrrol-5- yl)phenyl)-7-(4-(trifluoromethyl)phenyl)-2-naphthoic acid (31). Treatment of 430 (33.7 mg, 0.053 mmol) by using Procedure A (without HPLC purification) gave 431 of sufficient purity to be used in next step: 1 H NMR (400 MHz, MeOD) δ 8.59 (s, 1H), 8.30 (s, 1H), 8.00 (s, 1H), 7.92 (dd, J = 8.5, 5.3 Hz, 3H), 7.74 (d, J = 7.5 Hz, 3H), 7.65 (dd, J = 15.8, 6.5 Hz, 4H), 7.51 – 7.39 (m, 5H), 4.62 – 4.50 (m, 2H), 3.54 (d, J = 12.9 Hz, 2H), 3.22 (q, J = 13.9 Hz, 2H), 2.94 – 2.66 (m, 2H), 2.47 (dd, J = 13.2, 7.1 Hz, 1H), 2.18 (dd, J = 12.8, 5.0 Hz, 1H), 1.97 (q, J = 11.3 Hz, 2H). HRMS m/z [M+H] + for C38H33NO3F3 calculated 608.2413, found 608.2408. tert-Butyl (1R,5S,6r)-6-(4-bromophenyl)-6-hydroxy-3-azabicyclo[3.1.1]he ptane- 3-carboxylate (433). Treatment of 432 (105.65 mg, 0.5 mmol) by using Procedure E gave 33 (99.36 mg, 54%): 1 H NMR (400 MHz, CDCl3) δ 7.50 (dd, J = 8.7, 2.3 Hz, 2H), 7.39 (d, J = 8.5 Hz, 2H), 3.82 – 3.66 (m, 4H), 2.82 – 2.76 (m, 1H), 2.71 – 2.63 (m, 1H), 1.55 (dt, J = 11.4, 6.2 Hz, 1H), 1.45 (d, J = 2.5 Hz, 9H), 1.33 (d, J = 10.1 Hz, 1H). HRMS m/z [M - t- butyl+2H] + for C13H15N 79 BrO3 calculated 312.0235, found 312.0234. tert-Butyl (1R,5S,6r)-6-(4-(3-(ethoxycarbonyl)-6-(4- (trifluoromethyl)phenyl)naphthalen-1-yl)phenyl)-6-hydroxy-3- azabicyclo[3.1.1]heptane-3- carboxylate (434). Treatment of 433 (25.8 mg, 0.07 mmol) by using Procedure D gave 434 (41 mg, 93%): 1 H NMR (400 MHz, CDCl3) δ 8.69 (d, J = 1.6 Hz, 1H), 8.23 (d, J = 1.9 Hz, 1H), 8.07 – 8.01 (m, 2H), 7.81 (d, J = 8.2 Hz, 2H), 7.79 – 7.73 (m, 3H), 7.71 (d, J = 8.0 Hz, 2H), 7.56 (d, J = 8.0 Hz, 2H), 4.45 (q, J = 7.1 Hz, 2H), 3.94 – 3.77 (m, 4H), 3.02 – 2.94 (m, 1H), 2.88 (d, J = 6.7 Hz, 1H), 1.74 (dt, J = 11.4, 6.1 Hz, 1H), 1.52 (s, 9H), 1.48 – 1.40 (m, 4H). HRMS m/z [M – Boc – O] + for C32H27NO2F3 calculated 514.1994, found 514.2001. 4-(4-((1R,5S,6r)-3-(tert-Butoxycarbonyl)-6-hydroxy-3-azabicy clo[3.1.1]heptan-6- yl)phenyl)-7-(4-(trifluoromethyl)phenyl)-2-naphthoic acid (435). Treatment of 434 (41 mg, 0.065 mmol) by using Procedure A (without HPLC purification) gave 435 of sufficient purity to be used in next step: 1 H NMR (400 MHz, MeOD) δ 8.58 (s, 1H), 8.31 (d, J = 1.9 Hz, 1H), 8.04 (d, J = 1.6 Hz, 1H), 7.97 (dd, J = 8.6, 3.2 Hz, 3H), 7.81 – 7.74 (m, 3H), 7.72 (d, J = 7.9 Hz, 2H), 7.53 (t, J = 6.6 Hz, 2H), 3.84 – 3.69 (m, 4H), 2.96 – 2.82 (m, 2H), 1.69 (dt, J = 11.1, 6.0 Hz, 1H), 1.36 (d, J = 9.7 Hz, 1H). MS m/z [M – Boc – O] + for C 30 H 23 NO 2 F 3 calculated 486.2, found 486.2. tert-Butyl 4-(4-bromophenyl)-4-hydroxyazepane-1-carboxylate (437). Treatment of 436 (106.65 mg, 0.5 mmol) by using Procedure E gave 37 (135 mg, 73%): 1 H NMR (400 MHz, CDCl 3 ) δ 7.45 (d, J = 8.0 Hz, 2H), 7.32 (d, J = 8.3 Hz, 2H), 3.92 – 3.47 (m, 2H), 3.32 (d, J = 11.3 Hz, 2H), 2.27 – 1.67 (m, 7H), 1.47 (d, J = 4.1 Hz, 9H). tert-Butyl 4-(4-(3-(ethoxycarbonyl)-6-(4-(trifluoromethyl)phenyl)naphth alen-1- yl)phenyl)-4-hydroxyazepane-1-carboxylate (438). Treatment of 437 (38.9 mg, 0.105 mmol) by using Procedure D gave 438 (36.4 mg, 82%): 1 H NMR (400 MHz, CDCl3) δ 8.67 (s, 1H), 8.22 (s, 1H), 8.03 (d, J = 9.4 Hz, 2H), 7.81 (d, J = 8.1 Hz, 2H), 7.74 (d, J = 8.0 Hz, 3H), 7.62 (d, J = 8.1 Hz, 2H), 7.55 – 7.45 (m, 2H), 4.46 (q, J = 7.1 Hz, 2H), 3.93 – 3.53 (m, 2H), 3.45 – 3.34 (m, 2H), 2.35 – 2.05 (m, 3H), 1.99 – 1.68 (m, 4H), 1.51 (s, 9H), 1.45 (t, J = 7.2 Hz, 3H). HRMS m/z [M + Na] + for C37H38NO5F3Na calculated 656.2600, found 656.2589. 4-(4-(1-(tert-Butoxycarbonyl)-4-hydroxyazepan-4-yl)phenyl)-7 -(4- (trifluoromethyl)phenyl)-2-naphthoic acid (439). Treatment of 438 (36.4 mg, 0.057 mmol) by using Procedure A (without HPLC purification) gave 439 of sufficient purity to be used in next step: 1 H NMR (400 MHz, MeOD) δ 8.34 (s, 1H), 8.08 (s, 1H), 7.80 (s, 1H), 7.74 (d, J = 8.1 Hz, 3H), 7.55 (d, J = 8.0 Hz, 3H), 7.39 (d, J = 8.1 Hz, 2H), 7.25 (d, J = 7.2 Hz, 2H), 3.46 (s, 2H), 3.23 – 3.16 (m, 2H), 2.16 – 1.51 (m, 6H), 1.27 (s, 9H). MS m/z [M – Boc – O] + for C30H25NO2F3 calculated 488.1837, found 488.1840. tert-Butyl 5-(4-bromophenyl)-5-hydroxyazocane-1-carboxylate (441). Treatment of 440 (113.65 mg, 0.5 mmol) by using Procedure E gave 441 (155 mg, 81%): 1 H NMR (400 MHz, CDCl3) δ 7.44 – 7.39 (m, 2H), 7.37 – 7.32 (m, 2H), 3.46 – 3.24 (m, 4H), 2.06 – 1.77 (m, 6H), 1.58 – 1.45 (m, 2H), 1.44 (s, 9H). HRMS m/z [M - t-butyl+H - OH-] + for C14H17N 79 BrO2 calculated 310.0443, found 310.0437. tert-Butyl 5-(4-(3-(ethoxycarbonyl)-6-(4-(trifluoromethyl)phenyl)naphth alen-1- yl)phenyl)-5-hydroxyazocane-1-carboxylate (442). Treatment of 441 (26.9 mg, 0.07 mmol) by using Procedure D gave 442 (35.5 mg, 78%): 1 H NMR (400 MHz, CDCl 3 ) δ 8.68 (s, 1H), 8.22 (d, J = 1.9 Hz, 1H), 8.07 – 8.00 (m, 2H), 7.81 (d, J = 8.1 Hz, 2H), 7.78 – 7.71 (m, 3H), 7.66 (d, J = 8.1 Hz, 2H), 7.54 – 7.47 (m, 2H), 4.46 (q, J = 7.1 Hz, 2H), 3.56 – 3.37 (m, 5H), 2.27 – 2.06 (m, 2H), 2.00 (d, J = 14.6 Hz, 4H), 1.71 (s, 2H), 1.50 (s, 9H), 1.45 (t, J = 7.1 Hz, 3H). MS m/z [M – Boc – O] + for C 33 H 31 NO 2 F 3 calculated 530.2, found 530.2. 4-(4-(1-(tert-Butoxycarbonyl)-5-hydroxyazocan-5-yl)phenyl)-7 -(4- (trifluoromethyl)phenyl)-2-naphthoic acid (443). Treatment of 442 (32 mg, 0.049 mmol) by using Procedure A (without HPLC purification) gave 443 of sufficient purity to be used in next step: 1 H NMR (400 MHz, MeOD) δ 8.57 (s, 1H), 8.32 – 8.28 (m, 1H), 8.05 – 8.01 (m, 1H), 7.96 (d, J = 8.3 Hz, 3H), 7.82 – 7.73 (m, 3H), 7.67 (d, J = 7.9 Hz, 2H), 7.46 (d, J = 7.9 Hz, 2H), 3.56 (q, J = 11.3 Hz, 2H), 3.45 – 3.28 (m, 2H), 2.27 – 1.87 (m, 6H), 1.67 – 1.53 (m, 2H), 1.49 (s, 9H). MS m/z [M – Boc – O] + for C31H27NO2F3 calculated 502.1994, found 502.1990. EXAMPLE 5 This Example illustrates the affinity of compounds of formula (III). The affinity was determined using a whole cell fluorescent binding assay, which was shown to underestimate the actual affinity. No. MRS # R = IC50 (nM) 425 MRS4833 5.92±0.55 This example demonstrates the species dependence of binding of various P2Y 14 R antagonists, as determined in a fluorescent antagonist binding assay in whole cells (N = 3-4). As is apparent from the results set forth in Table 2, compound 29 had slightly enhanced affinity (IC50184 nM) at the mP2Y14R vs the human homologue, while 11, 19 and 32 had nearly the same affinity in the two species. Desirably, receptor affinity is maintained across species, facilitating performance in in vivo preclinical testing in small animals, with the mouse being the most commonly used species in initial evaluation. Table 2 Compound mP2Y 14 R, hP2Y 14 R, 5 b 902±344 269±121 Fluorescent binding assays using 2 (20 nM, for a 45 min incubation) as tracer. The hP2Y14R was stably expressed in CHO cells, and the mP2Y14R was stably expressed in HEK293 cells. b Data from Jung et al. 2 and Mufti et al. 7 EXAMPLE 7 This example demonstrates some properties of amidomethyl ester prodrugs 37a– c. a The results are set forth in Table 3. The triazole-containing compound 37a was the most rapidly cleaved, while the naphthalene-containing compounds 37b and 37c did not reach 50% cleavage after 72 h at 37 ˚C. All of the compounds were stable for 24 h upon aqueous incubation in the absence of PLE.

Table 3 Compound MW, TPSA cLogP b Parent t1∕2 (min, mean±SEM) g g p g ( g gp ) , determined. c. At pH 7.3 in aqueous medium in the presence of PLE. d. Percent cleavage at 72 h in parentheses. EXAMPLE 8 This examples demonstrates some in vitro and in vivo ADMET data for compound 32 compared to known P2Y14R antagonist 5. a The compound was stable in simulated gastric and intestinal fluids, in plasma of three species, and in human and mouse liver microsomes. Table 4 Test 5 b 32 i l d i i l flid Plasma stability (3 >240 (h); >240 (r); TBD (h); >240 (r); i ) d 240 ( ) 240 ( ) a Pro b Data determined in Yu et al. 1 c Mean ± SD, pION method. d Species tested for plasma stability were human, rat and mouse; species as indicated for microsomal stability. e Method noted in parentheses. e By i.v. administration in rat: 0.5 mg/kg dose. See Table 5 for complete results. ND, not determined. EXAMPLE 9 This example demonstrates some pharmacokinetic parameters a for compound 32 administered by intravenous and intraperitoneal routes. The results are set forth in Table 5.

Table 5 Dose C max T max AUC0-last AUC 0-∞ T 1/2 MRT last Vd Kel F Cl ( /k ) ) In healthy adult male Wistar rats; with 3 per group. Abbreviations: Cmax, maximal concentration; T max , time at which maximal concentration observed; AUC 0 - last , area under the plasma drug concentration–time curve up to the last quantifiable time-point; AUC0-∞, area under the plasma drug concentration–time curve to infinite time; T 1/2 , terminal half-life; MRTlast, mean residence time; Vd, volume of distribution; Kel, elimination rate constant; F, biovailability; Cl, total body clearance. EXAMPLE 10 This example demonstrates a binding comparison of various P2Y14R antagonists at mP2Y 14 R and hP2Y 14 R. The results are set forth in Table 6. Table 6 mP2Y 14 R, hP2Y 14 R, Compound 120 26.2±10.7 39.5±9.8 ding was determined as in Table . C50 o b t o o b d g was dete ed using flow cytometry of whole mP2Y14R-HEK cells in the presence of a fixed concentration (200 nM) of 3 (mean±SEM, n = 3 – 6). bIC 50 values from literature (Yu, J. et al., J. Med. Chem.2018, 61, 4860−4882, Jung, Y. H. et al., J. Med. Chem.2020, 63, 9563–9589) EXAMPLE 11 This example demonstrates ADMET values for two P2Y 14 R antagonists in this study, compared to reference quinuclidine 4.

Table 7 Test 4 a 114 121 i l d i i l fl id c Species tested for plasma stability were human, rat and mouse; species as indicated for microsomal stability. d By i.v. administration: 0.5 mg/kg dose. e hERG inhibition was measured using a fluorescent method. Aqueous acid stability was determined by NMR. EXAMPLE 12 This example demonstrates IC 50 values of hERG ion channel block by selected P2Y14R antagonists and positive control (cisapride) determined in a patch clamp assay. Value in parentheses indicates EC 50 value for stimulation of hERG channel activity. None of the compounds inhibited hERG, but one antagonist 20 stimulated hERG activity with an EC50 of 18 µM. Table 8 Compound IC 50 /(EC 50 ), µM XAMPLE 13 This example illustrates the preparation of prodrugs and the Plasma Protein Binding (PPB) of selected derivatives in three Species, including three active drugs and two prodrugs. Scheme 6. Preparation of Amidomethyl Esters of 114, 121 and 138, as well as Reported Ester of 1, i.e.147, 13 and the Double Prodrug of 115, i.e.143 a

CF 3 O CF 3 O O H a Reagents a o MF, 45 C, 1 h, 76– 98%; (b) TFA, DCM, rt, 1 h, 99%; (c) ethyl chloroformate, N,N-diisopropylethylamine, DMF, rt, overnight, 71%. The results obtained are set forth in Table 9. Table 9. Plasma Protein Binding (PPB) of selected derivatives Compound Mouse, % Rat, % bound Human, % a For the ester p ining in mouse and rat plasma at 5 h is given in italics. EXAMPLE 14 This example illustrates the evaluation of compound 114 in Chronic Constriction Injury Mouse Model of Neuropathic Pain. Compound 114 was evaluated in a validated model of chronic neuropathic pain in the mouse, i.e. following chronic constriction of the sciatic nerve (CCI). This compound was administered by the i.p. route (10 µmol/kg). The reversal of CCI-induced mechano-allodynia by the compound was nearly complete (89.3±10.1%) at 1 h post-injection, and significant partial protection was maintained through 3 h but was absent at 5 h (Figure 6). No obvious side effects were observed in the mice. EXAMPLE 15 This example illustrates the antagonist potency of prodrugs 142 and 143. The carbamate prodrug 142 and the double prodrug 143 having carbamate and ester moieties were derived from the most potent antagonist 114. Prodrugs 142 and 143 were tested in the mouse asthma model in comparison to the HCl salt form of the parent drug 114. The N,N-dimethylamidomethyl ester prodrug 141 and the carbamate prodrug 142 reduced allergic inflammation more effectively than the parent drug 15. Intriguingly, the double prodrug 143 displayed an even stronger potency, dramatically reducing the recruitment of neutrophils, eosinophils, and lymphocytes into the airway. The results are depicted in Fig.7. These promising results suggest that double prodrug 143 could be a candidate for further drug development and therapeutic treatment of asthma and other inflammatory diseases. EXAMPLE 16 Experimental procedures Chemical synthesis General Information. Materials and Methods. All chemicals and anhydrous solvents were obtained directly from commercial sources. All reactions were carried out under argon atmosphere using anhydrous solvents unless specified otherwise. Room temperature (rt) refers to 25 ± 5 °C. Silica-gel precoated with F254 on aluminum plates were used for TLC. The spots were examined under ultraviolet light at 254 nm and further visualized by anisaldehyde or cerium ammonium molybdate stain solution. Column chromatography was performed on silica gel (40−63 μm, 60 Å). NMR spectra were recorded on a Bruker 400 MHz spectrometer. Chemical shifts are given in ppm (δ), calibrated to the residual solvent signal peaks of CDCl3 (7.26 ppm), CD3OD (3.31 ppm), or DMSO-d6 (2.50 ppm) for 1 H NMR with coupling constant (J) values reported in Hz. High resolution mass (HRMS) measurements were performed on a proteomics optimized Q-TOF-2 (Micromass- Waters). The RP-HPLC was performed using Phenomenex Luna 5 μm C18(2)100A, AXIA, 21.2 mm × 250 mm column. Antagonist purity was determined as ≥95% using Agilent ZORBAX SB-Aq, 5 μm, 4.6 mm × 150 mm column attached to Agilent 1100 HPLC system. The HPLC traces for compounds tested in vivo (xxxx) were included in Supporting Information. (2R,3R,4S,5R,6R)-2-(Acetoxymethyl)-6-azidotetrahydro-2H-pyra n-3,4,5-triyl triacetate (523). A mixture of 522 (205.6 mg, 0.5 mmol) and sodium azide (65 mg, 1 mmol) in DMF (4 mL) was stirred at rt for 2 h. The volatiles were evaporated, and the residue was extracted with DCM three times. The combined organic layer was dried with Na2SO4 to give 23 as a white powder (184.7 mg, 99%): 1 H NMR (400 MHz, CDCl 3 ) δ 5.14 (t, J = 9.5 Hz, 1H), 5.01 (t, J = 9.7 Hz, 1H), 4.86 (t, J = 9.2 Hz, 1H), 4.59 (d, J = 8.9 Hz, 1H), 4.18 (dd, J = 12.5, 4.8 Hz, 1H), 4.07 (dd, J = 12.5, 2.3 Hz, 1H), 3.74 (ddd, J = 10.1, 4.8, 2.3 Hz, 1H), 2.00 (s, 3H), 1.98 (s, 3H), 1.94 (s, 3H), 1.91 (s, 3H). HRMS m/z [M+Na] + for C14H19N3O9Na calculated 396.1019, found 396.1017. 4-(4-(1-(Hex-5-yn-1-yl)piperidin-4-yl)phenyl)-7-(4-(trifluor omethyl)phenyl)-2- naphthoic acid (525). A mixture of 502 (11.9 mg, 0.025 mmol), 6-Iodo-1-hexyne (7.3 µL, 11.4 mg, 0.055 mmol), and K2CO3 (13.8 mg, 0.1 mmol) in DMF (1 mL) was stirred at rt overnight. THF-H2O-MeOH (3:1:1, 1 mL) and LiOH (7.4 mg, 0.31 mmol) were added into the solution. The resulting mixture was sonicated for 30 s and stirred at rt for 2 h to hydrolyze the ester byproduct to give product. The reaction was neutralized by 1 M HCl. The volatiles were evaporated, and the residue was column chromatographed (DCM/MeOH/AcOH, 100:0:0.1→95:5:0.1) to give 525 (6 mg, 43%): 1 H NMR (400 MHz, MeOD) δ 8.69 (d, J = 1.7 Hz, 1H), 8.38 (d, J = 1.9 Hz, 1H), 7.97 – 7.93 (m, 4H), 7.86 (dd, J = 8.8, 1.9 Hz, 1H), 7.76 (d, J = 8.1 Hz, 2H), 7.47 (t, J = 6.8 Hz, 4H), 3.71 (d, J = 12.1 Hz, 2H), 3.25 – 3.11 (m, 4H), 3.09 – 2.98 (m, 1H), 2.33 – 2.28 (m, 2H), 2.22 (d, J = 13.9 Hz, 2H), 2.07 (d, J = 13.0 Hz, 2H), 1.94 (d, J = 10.8 Hz, 3H), 1.62 (t, J = 7.6 Hz, 2H); HRMS m/z [M+H] + for C35H33NO2F3 calculated 556.2463, found 556.2466. 4-(4-(1-(4-(1-((2R,3R,4S,5R,6R)-3,4,5-Triacetoxy-6-(acetoxym ethyl)tetrahydro- 2H-pyran-2-yl)-1H-1,2,3-triazol-4-yl)butyl)piperidin-4-yl)ph enyl)-7-(4- (trifluoromethyl)phenyl)-2-naphthoic acid (504). Sodium ascorbate (5.9 mg, 0.03 mmol) was added into a mixture of 525 (8.3 mg, 0.015 mmol), 523 (11.2 mg, 0.03 mmol), and CuSO4.5H2O (3.7 mg, 0.015 mmol) in DMF/H2O (9:1, 2 mL). the resulting mixture was stirred at 90 o C overnight. The volatiles were evaporated, and the residue was column chromatographed (DCM/MeOH/AcOH, 100:0:0.2→90:10:0.2) to give 504 (13 mg, quantitative): 1 H NMR (400 MHz, MeOD) δ 8.63 (s, 1H), 8.32 (s, 1H), 8.09 (s, 1H), 7.89 (q, J = 14.2 Hz, 4H), 7.72 (d, J = 7.8 Hz, 3H), 7.35 (s, 4H), 6.07 (d, J = 8.1 Hz, 1H), 5.50 (td, J = 16.9, 8.2 Hz, 2H), 5.22 (t, J = 9.3 Hz, 1H), 4.28 (dd, J = 12.4, 4.6 Hz, 1H), 4.20 (d, J = 9.8 Hz, 1H), 4.12 (d, J = 12.4 Hz, 1H), 3.66 (s, 2H), 3.21 – 3.06 (m, 4H), 2.99 – 2.89 (m, 1H), 2.79 (s, 2H), 2.09 (s, 4H), 2.00 (d, J = 5.7 Hz, 6H), 1.95 (d, J = 2.5 Hz, 6H), 1.81 (s, 4H). HRMS m/z [M+H] + for C49H52N4O11F3 calculated 929.3585, found 929.3578. 7-(4-(Trifluoromethyl)phenyl)-4-(4-(1-(4-(1-((2R,3R,4S,5S,6R )-3,4,5-trihydroxy- 6-(hydroxymethyl)tetrahydro-2H-pyran-2-yl)-1H-1,2,3-triazol- 4-yl)butyl)piperidin-4- yl)phenyl)-2-naphthoic acid (508). NaOH (3 M, 1 mL) was added into a solution of 504 (13 mg, 0.015 mmol) in MeOH (2 mL). The solution was stirred at rt overnight. Volatiles were evaporated, and the residue was purified by RP-HPLC (C18, A: ACN, B: 10 mM TEAA, 35% → 60% A in 40 min, flow rate = 5 mL/min, tR = 24.9 min) to give 508 as a white powder (9 mg, 79% overall yield from 525): 1 H NMR (400 MHz, DMSO) δ 8.61 (s, 1H), 8.52 (s, 1H), 8.02 (d, J = 8.4 Hz, 3H), 7.89 (d, J = 15.3 Hz, 3H), 7.83 (d, J = 8.1 Hz, 2H), 7.40 (d, J = 2.8 Hz, 4H), 5.42 (d, J = 9.3 Hz, 1H), 3.75 – 3.61 (m, 2H), 3.43 – 3.13 (m, 4H), 2.99 (d, J = 10.5 Hz, 2H), 2.62 (t, J = 7.4 Hz, 2H), 2.58 – 2.52 (m, 1H), 2.34 (s, 2H), 2.02 (t, J = 11.4 Hz, 2H), 1.84 – 1.78 (m, 2H), 1.70 (d, J = 11.8 Hz, 2H), 1.60 (d, J = 7.3 Hz, 2H), 1.51 (s, 2H). HRMS m/z [M+H] + for C 41 H 44 N 4 O 7 F 3 calculated 761.3162, found 761.3154. The invention provides the following aspects: 1. A compound of formula (I) or formula (II): R 1 R is halo or CF 3 , R 2 is selected from COOH, COOR 12 , CONHOH, CH2CON(R 11 )2, CHR 7 OCOR 8 , O (CH2)oNR 9 , tetrazolyl, CH2OPO(OH)2, an R 3 is selected from NHR 4 , CO ONH(CH 2 ) q NH 2 , COCF 3 , O OH N H N HO OH NH 2 HO 2 NH 2 O , R 4 is H or COR 6 wherein R 6 is C 1 -C 6 alkyl, C 6 -C 10 aryl, or R 5 is C1-C6 alkyl, C3-C8 cycloalkyl, or benzyl, R 6 is NH 2 , or CONH 2 , R 7 -R 9 are independently hydrogen or C1-C6 alkyl, R 10 is H, C 1 -C 10 alkylcarbonyl, or C 1 -C 10 alkyloxycarbonyl, R 11 is hydrogen or C1-C6 alkyl, R 12 is C1-C6 alkyl, C3-C8 cycloalkyl, or benzyl, m, o, and q are independently integers of 1 to about 10, and n is zero or an integer of 1 to about 10, or a pharmaceutically acceptable salt thereof, or stereoisomers thereof. 2. The compound or salt or stereoisomers thereof of aspect 1, wherein R 1 is CF3. 3. The compound or salt or stereoisomers thereof of aspect 1 or 2, wherein the compound is of formula (I). 4. The compound or salt or stereoisomers thereof of any one of aspects 1-3, 3 NH wherein R is selected from NH 2 , NHCOCH 3 , NHCOC 6 H 5 , NHCOC(CH 3 ) 3 NBoc and . ompound or salt or stereoisomers thereof of any one of aspects 1-3, wherein R 3 is COOR 5 . 6. The compound or salt or stereoisomers thereof of any one of aspects 1-3, wherein R 3 is (C≡C)n(CH2)mR 6 or CONH(CH2)qNH2. 7. The compound or salt or stereoisomers thereof of aspect 6, wherein R 3 is selected from CONH(CH2)3NH2, CH2CONH2, (CH2)3NH2, and C≡CCH2NH2. 8. The compound or salt or stereoisomers thereof of any one of aspects 1-3, O N wherein R 3 is selected from Br, (CH 2 ) 2 -CN O O compound is of formula (II). 10. The compound or salt or stereoisomers thereof of aspect 9, wherein R 3 is O O selected from Br, (CH2)2-CN O pect 9, wherein R 3 is OH . he compound or salt or stereoisomers thereof of any one of aspects 1-10, wherein R 2 is H. 12. The compound or salt or stereoisomers thereof of any one of aspects 1-10, wherein R 2 is COOR 12 , CH 2 CON(R 11 ) 2 , CHR 7 OCOR 8 , (CH 2 ) o NR 9 , tetrazolyl, O O CH2OPO(OH)2, o 13. Th isomers thereof of aspect 1, wherein the compound is: F 3 C 2CON(CH 3 ) 2 . 14. The compound or salt or stereoisomers thereof of aspect 9, wherein R 3 is NH NH NH NCOCH NH selected from 3 NH NCOCH NH O NH , and wherein the configuration of the three chiral centers is (S,S,S). 16. The compound or salt or stereoisomers thereof of aspect 9, wher 3 ein R is H 2 N HO NH NH HO 2 2 selected from OH O OH reof of any one of aspects 14-16, wherein R 2 is H. p y p , wherein R 2 is COOR 12 , CH2CON(R 11 )2, CHR 7 OCOR 8 , (CH2)oNR 9 , tetrazolyl, O O O CH 2 OPO(OH) 2 , o 19. Th oisomers thereof of aspect 1, wherein the compound is: F3 C O ree chiral centers is (S,S,S). 20. A pharmaceutical composition comprising a compound or salt or stereoisomers thereof of any one of aspects 1-19 and a pharmaceutically acceptable carrier. 21. A compound or salt or stereoisomers thereof of any one of aspects 1-19, for use in antagonizing a P2Y14R receptor in a mammal in need thereof. 22. A compound or salt or stereoisomers thereof of any one of aspects 1-19, for use in treating or preventing an inflammatory condition in a mammal in need thereof. 23. The compound for use according to aspect 22 wherein the inflammatory condition is selected from the group consisting of asthma, cystic fibrosis, and sterile inflammation of the kidney. 24. A compound or salt or stereoisomers thereof of any one of aspects 1-19, for use in treating pain in a mammal in need thereof. 25. A method of antagonizing a P2Y 14 R receptor in a mammal in need thereof, comprising administering to the mammal an effective amount of a compound or salt or stereoisomers thereof of any one of aspects 1-19. 26. A method of treating or preventing an inflammatory condition in a mammal in need thereof, comprising administering to the mammal an effective amount of a compound or salt or stereoisomers thereof of any one of aspects 1-19. 27. The method according to aspect 26, wherein the inflammatory condition is selected from the group consisting of asthma, cystic fibrosis, and sterile inflammation of the kidney. 28. A method of treating pain in a mammal in need thereof, comprising administering to the mammal an effective amount of a compound or salt or stereoisomers thereof of any one of aspects 1-19. 28. The method according to aspect 27, wherein the inflammatory condition is selected from the group consisting of asthma, cystic fibrosis, and sterile inflammation of the kidney. 29. A method of treating pain in a mammal in need thereof, comprising administering to the mammal an effective amount of a compound or salt or stereoisomers thereof of any one of aspects 1-20.

1. Yu, J.; Ciancetta, A.; Dudas, S.; Duca, S.; Lottermoser, J.; Jacobson, K.A. Structure-guided modification of heterocyclic antagonists of the P2Y 14 receptor. J. Med. Chem.2018, 61, 4860−4882, doi: 10.1021/acs.jmedchem.8b00168. 2. Jung, Y. H.; Yu, J.; Wen, Z.; Salmaso, V.; Phung, N. B.; Karcz, T.; Chen, Z.; Duca, S.; Bennett, J. M.; Dudas, S.; Cook, D. N.; Salvemini, D.; Gao, Z. G.; Jacobson, K. A. Exploration of alternative scaffolds for P2Y14 receptor antagonists containing a biaryl core. J. Med. Chem.2020, 63, 9563–9589. 3. Battistone, M. A.; Mendelsohn, A. C.; Spallanzani, R. G.; Allegretti, A. S.; Liberman, R. N.; Sesma, J.; Kalim, S.; Wall, S. M.; Bonventre, J. V.; Lazarowski, E. R.; Brown, D.; Breton, S. Pro-inflammatory P2Y14 receptor inhibition protects against ischemic acute kidney injury in mice. J. Clin. Invest.2020, 130, 3734–3749. 4. Xu, J.; Morinaga, H.; Oh, D.; Li, P.; Chen, A.; Talukdar, S.; Lazarowski, E.; Olefsky, J. M.; Kim, J. J. GPR105 ablation prevents inflammation and improves insulin sensitivity in mice with diet-induced obesity. J. Immunol.2012, 189(4), 1992–1999. 5. Jain, S.; Jacobson, K. A. Adipocyte specific ablation of P2Y 14 R improves glucose metabolism in mice with diet-induced obesity. American Diabetes Assoc., 80 th Scientific Session, Abstract, 2020. 6. Karcz, T. P.; Whitehead, G. S.; Nakano, H.; Williams, J. G.; Deterding, L. J.; Jacobson, K. A.; Cook, D. N. UDP-glucose and P2Y 14 receptor amplify allergen-induced airway eosinophilia. Submitted. 7. Mufti, F.; Jung, Y. H.; Giancotti, L. A.; Yu, J.; Chen, Z..; Phung, N. B.; Jacobson, K. A.; Salvemini, D. P2Y14 receptor antagonists reverse chronic neuropathic pain in a mouse model. ACS Med. Chem. Lett.2020, 11, 1281−1286. 8. Lin, J.; Liu, F.; Zhang, Y-Y.; Song, N.; Liu, M-K.; Fang, X-y.; Liao, D-Q.; Zhou, C.;Wang, H.; Shen, J-F. P2Y14 receptor is functionally expressed in satellite glial cells and mediates interleukin‐1β and chemokine CCL2 secretion. J. Cell. Physiol. 2019, 234, 21199– 21210. 9. Jacobson, K. A.; Delicado, E. G.; Gachet, C.; Kennedy, C.; von Kügelgen, I.; Li, B.; Miras-Portugal, T.; Novak, I.; Schöneberg, T.; Perez-Sen, R.; Thor, D.; Wu, B.; Yang, Z.; Pharmacol.2020, 177, 2413–2433. 10. Kinoshita, M.; Nasu-Tada, K.; Fujishita, K.; Sato, K.; Koizumi, S. Secretion of matrix metalloproteinase-9 from astrocytes by inhibition of tonic P2Y14-receptor-mediated signal(s). Cell. Mol. Neurobiol.2013, 33, 47–58. DOI: 10.1007/s10571-012-9869-4 11. Zhang, Z.; Hao, K.; Li, H.; Lu, R.; Liu, C.; Zhou, M.; Li, B.; Meng, Z.; Hu, Q.; Jiang, C. Design, synthesis and anti-inflammatory evaluation of 3-amide benzoic acid derivatives as novel P2Y 14 receptor antagonists. Eur. J. Med. Chem.2019, 181, 111564. 12. Hamel, M.; Henault, M.; Hyjazie, H.; Morin, N.; Bayly, C.; Skorey, K.; Therien, A.G.; Mancini, J.; Brideau, C.; Kargman, S. Discovery of novel P2Y 14 agonist and antagonist using conventional and nonconventional methods. J. Biomol. Screening 2011, 16(9), 1098– 1105. 13. Wang, W.; Liu, C.; Li, H.; Tian, S.; Liu, Y.; Wang, N.; Yan, D.; Li, H.; Hu, Q. Discovery of novel and potent P2Y 14 R antagonists via structure-based virtual screening for the treatment of acute gouty arthritis. J. Adv. Res.2020, 23, 133–142. doi: https://doi.org/10.1016/j.jare.2020.02.007 14. Junker, A.; Balasubramanian, R.; Ciancetta, A.; Uliassi, E.; Kiselev, E.; Martiriggiano, C.; Trujillo, K.; Mtchedlidze, G.; Birdwell, L.; Brown, K. A.; Harden, T. K.; Jacobson, K. A. Structure-based design of 3-(4-aryl-1H-1,2,3-triazol-1-yl)-biphenyl derivatives as P2Y 14 receptor antagonists. J. Med. Chem.2016, 59, 6149−6168. 15. Zhang, K.; Zhang, J.; Gao, Z. G.; Paoletta, S.; Zhang, D.; Han, G. W.; Li, T.; Ma, L.; Zhang, W.; Müller, C. E.; Yang, H.; Jiang, H.; Cherezov, V.; Katritch, V.; Jacobson, K. A.; Stevens, R. C.; Wu, B.; Zhao, Q. Agonist-bound structure of the human P2Y12R receptor. Nature 2014, 509, 119−122. 16. Kiselev, E.; Barrett, M.; Katritch, V.; Paoletta, S.; Weitzer, C. D.; Hammes, E.; Yin, A. L.; Zhao, Q.; Stevens, R. C.; Harden, T. K.; Jacobson, K. A. Exploring a 2-naphthoic acid template for the structure-based design of P2Y 14 receptor antagonist molecular probes. ACS Chem. Biol.2014, 9, 2833-2842. 17. Barrett, M. O.; Sesma, J. I.; Ball, C. B.; Jayasekara, P. S.; Jacobson, K. A.; Lazarowski, E. R.; Harden, T. K. A selective high affinity antagonist of the P2Y14 receptor 84, 41–49. 18. Robichaud, J.; Fournier, J.-F.; Gagné, S.; Gauthier, J. Y.; Hamel, M.; Han, Y.; Hénault, M.; Kargman, S.; Levesque, J. F.; Mamane, Y.; Mancini, J.; Morin, N.; Mulrooney, E.; Wu, J.; Black, W. C. Applying the pro-drug approach to afford highly bioavailable antagonists of P2Y14. Bioorg. Med. Chem. Lett.2011, 21(14), 4366–4368. 19. Mazák, K.; Noszál, B. Physicochemical properties of zwitterionic drugs in therapy. ChemMedChem 2020, 15, 1102–1. 20. Sabbadin, D.; Ciancetta, A.; Moro, S. Bridging molecular docking to membrane molecular dynamics to investigate GPCR-ligand recognition: The human A₂ A adenosine receptor as a key study. J. Chem. Inf. Model.2014, 54 (1), 169–183. 21. Strupczewski, J. T.; Allen, R. C.; Gardner, B. A.; Schmid, B. L.; Stache, U.; Glamkowski, E. J.; Jones, M. C.; Ellis, D. B.; Huger, F. P.; Dunn, R. W. Synthesis and neuroleptic activity of 3-(l-substituted-4-piperidinyl)-l,2-benzisoxazoles, J. Med. Chem. 1985, 28, 761–769. 22. Ballesteros, J. A.; Weinstein, H. Integrated methods for the construction of three dimensional models and computational probing of structure function relations in G protein- coupled receptors. Methods Neurosci.1995, 25, 366−428. 23. Pennington, L. D.; Moustakas, D. T. The necessary nitrogen atom: A versatile high-impact design element for multiparameter optimization. J. Med. Chem.2017, 60(9), 3552–3579. 24. Gardell, S.J.; Hopf, M.; Khan, A.; Dispagna M, Hampton Sessions. E.; Falter, R.; Kapoor, N.; Brooks, J.; Culver, J.; Petucci, C; Ma, C. T.; Cohen, S. E.; Tanaka, J.; Burgos, E. S.; Hirschi, J. S.; Smith, S. R.; Sergienko, E.; Pinkerton, A. B. Boosting NAD + with a small molecule that activates NAMPT. Nat. Commun.2019, 10, 3241. 25. Kiselyuk, A. S. HNF4 alpha bioactive ligands discovered by a high- throughput screen for modulators of the human insulin promoter. (Doctoral dissertation, pp.65). University of California, San Diego, 2011. (https://escholarship.org/content/qt64k4q3gr/qt64k4q3gr.pdf) 26. Yan, Z.; Caldwell, G. Optimization in Drug Discovery: In Vitro Methods (pp.58). Humana Press. measurements of lipophilicity to model biological distribution. J. Chromatogr. A 2004, 1037, 299–310. 28. Besnard, J.; Ruda, G. F.; Setola, V.; Abecassis, K.; Rodriguiz, R. M.; Huang, X. P.; Norval, S.; Sassano, M. F.; Shin, A. I.; Webster, L. A.; Simeons, F. R.; Stojanovski, L.; Prat, A.; Seidah, N. G.; Constam, D. B.; Bickerton, G. R.; Read, K. D.; Wetsel, W. C.; Gilbert, I. H.; Roth, B. L.; Hopkins, A. L. Automated design of ligands to polypharmacological profiles. Nature 2012, 492, 215–220. 29. Wible, B. A.; Hawryluk, P.; Ficker, E.; Kuryshev, Y. A.; Kirsch, G.; Brown, A. M. HERG-Lite: a novel comprehensive high-throughput screen for drug-induced hERG risk. J. Pharmacol. Toxicol. Methods 2005, 52(1), 136–145. 30. Piper, D. R.; Duff, S. R.; Eliason, H. C.; Frazee, W. J.; Frey, E. A.; Fuerstenau- Sharp, M.; Jachec, C.; Marks, B. D.; Pollok, B. A.; Shekhani, M. S.; Thompson, D. V.; Whitney, P.; Vogel, K. W.; Hess, S. D. Development of the predictor HERG fluorescence polarization assay using a membrane protein enrichment approach. Assay Drug Dev. Technol.2008, 6, 213–223, DOI: 10.1089/adt.2008.137 31. Perez C, Daniel KB, Cohen SM. Evaluating prodrug strategies for esterase- triggered release of alcohols. ChemMedChem, 2013, 8, 1662–1667. doi: 10.1002/cmdc.201300255. 32. Bennett, G. J.; Xie, Y. K. A peripheral mononeuropathy in rat that produces disorders of pain sensation like those seen in man. Pain 1988, 33, 87–107. 33. Whitehead, G. S.; Thomas, S. Y.; Shalaby, K. H.; Nakano, K.; Moran, T. P.; Ward, J. M.; Flake, G. P.; Nakano, H.; Cook, D. N. TNF is required for TLR ligand– mediated but not protease-mediated allergic airway inflammation. J. Clin. Invest.2017, 127, 3313–3326. 34. Mignani, S.; Rodrigues, J.; Tomas, H.; Jalal, R.; Singh, P. P.; Majoral, J.-P.; Vishwakarma, R. A. Present drug-likeness filters in medicinal chemistry during the hit and lead optimization process: how far can they be simplified? Drug Disc. Today, 2018, 23(3), 605–615. 35. Dalvie, D. K.; Kalgutkar, A. S.; Khojasteh-Bakht, S. C.; Obach, R. S.; O’Donnell, J. P. Biotransformation reactions of five-membered aromatic heterocyclic rings. Chem. Res. Toxicol.2002, 15(3), 269–299. https://doi.org/10.1021/tx015574b 36. Zhu, J.; Mo, J.; Lin, H.; Chen, Y.; Sun, H. The recent progress of isoxazole in medicinal chemistry. Bioorg. Med. Chem.2018, 26(12), 3065–3075. 37. Sun, H.; Shi, M.; Zhang, W.; Zheng, Y.-M.; Xu, Y.-Z.; Shi, J.-J.; Liu, T.; Gunosewoyo, H.; Pang, T.; Gao, Z. B.; Yang, F.; Tang, J.; Yu, L.-F. Development of novel alkoxyisoxazoles as sigma-1 receptor antagonists with antinociceptive efficacy. J. Med. Chem.2016, 59, 6329–6343. 38. Ratnatilaka Na Bhuket, P.; Jithavech, P.; Ongpipattanakul, B.; Rojsitthisak, P. Interspecies differences in stability kinetics and plasma esterases involved in hydrolytic activation of curcumin diethyl disuccinate, a prodrug of curcumin. RSC Advances, 2019, 9(8), 4626–4634. 39. Schrödinger Release 2019-3: Maestro, Schrödinger, LLC, New York, NY, 2019. 40. Sastry, G. M.; Adzhigirey, M.; Day, T.; Annabhimoju, R.; Sherman, W. Protein and ligand preparation: Parameters, protocols, and influence on virtual screening enrichments. J. Comput. Aided Mol. Des.2013, 27 (3), 221–234. 41. Harder, E.; Damm, W.; Maple, J.; Wu, C.; Reboul, M.; Xiang, J. Y.; Wang, L.; Lupyan, D.; Dahlgren, M. K.; Knight, J. L.; Kaus, J. W.; Cerutti, D. S.; Krilov, G.; Jorgensen, W. L.; Abel, R.; Friesner. R. A. OPLS3: A force field providing broad coverage of drug-like small molecules and proteins. J. Chem. Theory Comput.2016, 12 (1), 281–296. 42. Friesner, R. A.; Banks, J. L.; Murphy, R. B.; Halgren, T. A.; Klicic, J. J.; Mainz, D. T.; Repasky, M. P.; Knoll, E. H.; Shelley, M.; Perry, J. K.; Shaw, D. E.; Francis, P.; Shenkin, P. S. Glide: A new approach for rapid, accurate docking and scoring.1. Method and assessment of docking accuracy. J. Med. Chem.2004, 47 (7), 1739–1749. 43. Doerr, S.; Harvey, M. J.; Noé, F.; De Fabritiis, G. HTMD: High-throughput molecular dynamics for molecular discovery. J. Chem. Theory Comput.2016, 12 (4), 1845– 1852. 44. Humphrey, W.; Dalke, A.; Schulten, K. VMD: Visual molecular dynamics. J. Mol. Graph.1996, 14 (1), 33–38, 27. 45. Lomize, M. A.; Pogozheva, I. D.; Joo, H.; Mosberg, H. I.; Lomize, A. L. OPM Database and PPM web server: Resources for positioning of proteins in membranes. Nucleic Acids Res.2012, 40 (Database issue), D370-6. 46. Jorgensen, W. L.; Chandrasekhar, J.; Madura, J. D.; Impey, R. W.; Klein, M. L. Comparison of simple potential functions for simulating liquid water. J. Chem. Phys.1983, 79 (2), 926. 47. Harvey, M. J.; Giupponi, G.; Fabritiis, G. D. ACEMD: Accelerating biomolecular dynamics in the microsecond time scale. J. Chem. Theory Comput.2009, 5 (6), 1632–1639. 48. Best, R. B.; Zhu, X.; Shim, J.; Lopes, P. E. M.; Mittal, J.; Feig, M.; Mackerell, A. D. Optimization of the additive CHARMM all-atom protein force field targeting improved sampling of the backbone φ, ψ and side-chain χ1 and χ2 dihedral angles. J. Chem. Theory Comput.2012, 8 (9), 3257–3273. 49. Klauda, J. B.; Venable, R. M.; Freites, J. A.; O’Connor, J. W.; Tobias, D. J.; Mondragon-Ramirez, C.; Vorobyov, I.; MacKerell, A. D.; Pastor, R. W. Update of the CHARMM all-atom additive force field for lipids: Validation on six lipid types. J. Phys. Chem. B 2010, 114 (23), 7830–7843. 50. Vanommeslaeghe, K.; MacKerell, A. D. Automation of the CHARMM general force field (CGenFF) I: Bond perception and atom typing. J. Chem. Inf. Model.2012, 52 (12), 3144–3154. 51. Vanommeslaeghe, K.; Raman, E. P.; MacKerell, A. D. Automation of the CHARMM general force field (CGenFF) II: Assignment of bonded parameters and partial atomic charges. J. Chem. Inf. Model.2012, 52 (12), 3155–3168. 52. CHARMM General Force Field (CGenFF) program, https://cgenff.umaryland.edu/ 53. Kräutler, V.; van Gunsteren, W. F.; Hünenberger, P. H. A fast SHAKE algorithm to solve distance constraint equations for small molecules in molecular dynamics simulations. J. Comput. Chem.2001, 22 (5), 501–508. 54. Essmann, U.; Perera, L.; Berkowitz, M. L.; Darden, T.; Lee, H.; Pedersen, L. G. A smooth particle mesh Ewald method. J. Chem. Phys.1995, 103 (19), 8577. 55. Phillips, J. C.; Braun, R.; Wang, W.; Gumbart, J.; Tajkhorshid, E.; Villa, E.; Chipot, C.; Skeel, R. D.; Kalé, L.; Schulten, K. Scalable molecular dynamics with NAMD. J. Comput. Chem.2005, 26 (16), 1781–1802. 56. Williams, T.; Kelley, C. Gnuplot 5.0, http://www.gnuplot.info 57. Tetko, I. V.; Tanchuk, V. Y. Application of associative neural networks for prediction of lipophilicity in ALOGPS 2.1 program, J. Chem. Inf. Comput. Sci.2002, 42, 1136–1145. 58. Gauthier, J. Y.; Belley, M.; Deschênes, D.; Fournier, J. F.; Gagné, S.; Gareau, Y.; Hamel, M.; Hénault, M.; Hyjazie, H.; Kargman, S.; Lavallée, G.; Levesque, J. F.; Li, L.; Mamane, Y.; Mancini, J.; Morin, N.; Mulrooney, E.; Robichaud, J.; Thérien, M.; Tranmer, G.; Wang, Z.; Wu, J.; Black, W. C. The identification of 4,7-disubstituted naphthoic acid derivatives as UDP-competitive antagonists of P2Y14. Bioorg. Med. Chem. Lett.2011, 21, 2836–2839. All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein. The use of the terms “a” and “an” and “the” and “at least one” and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. The use of the term “at least one” followed by a list of one or more items (for example, “at least one of A and B”) is to be construed to mean one item selected from the listed items (A or B) or any combination of two or more of the listed items (A and B), unless otherwise indicated herein or clearly contradicted by context. The terms “comprising,” “having,” “including,” and “containing” are to be construed as open-ended terms (i.e., meaning “including, but not limited to,”) unless otherwise noted. Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention. Preferred aspects of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred aspects may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.