Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
((4-OXO-3,4-DIHYDROQUINAZOLIN-8-YL)METHYL)AMINE DERIVATIVES AS P13K INHIBITORS FOR THE TREATMENT OF CANCER
Document Type and Number:
WIPO Patent Application WO/2024/064024
Kind Code:
A1
Abstract:
The present invention relates to ((4-oxo-3,4-dihydroquinazolin-8-yl)methyl)amine derivatives of formula (I) as well as to said compounds for use as P13K inhibitors in methods of treatment of cancer.

Inventors:
BLAKE JAMES F (US)
BOYS MARK LAURENCE (US)
MARESKA DAVID A (US)
PAYETTE JOSHUA NATHANIEL (US)
SCHULTE CHRISTIE A (US)
YESTREPSKY BRYAN (US)
ZHAO QIAN (US)
Application Number:
PCT/US2023/032852
Publication Date:
March 28, 2024
Filing Date:
September 15, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ONKURE INC (US)
International Classes:
C07D403/04; A61K31/519; A61P35/00; A61P35/02; A61P35/04; C07D239/88; C07D401/04; C07D401/08; C07D403/08; C07D413/08; C07D417/08; C07D471/04
Domestic Patent References:
WO2015200352A12015-12-30
WO2023060262A12023-04-13
WO2023078401A12023-05-11
WO2023081209A12023-05-11
WO2023159155A12023-08-24
WO2023192416A12023-10-05
WO2023207881A12023-11-02
WO2023081209A12023-05-11
WO2023078401A12023-05-11
WO2023060262A12023-04-13
WO2023056407A12023-04-06
WO2021202964A12021-10-07
WO2023159155A12023-08-24
Foreign References:
CN116715658A2023-09-08
US3817837A1974-06-18
US3850752A1974-11-26
US3939350A1976-02-17
US3996345A1976-12-07
US4277437A1981-07-07
US4275149A1981-06-23
US4366241A1982-12-28
Other References:
POSOR, Y.EICHHORN-GRUNIG, M.HAUCKE, V.: "Phosphoinositides in endocytosis", BIOCHIM BIOPHYS ACTA, vol. 1851, no. 6, 2015, pages 794 - 804
BACKER, J. M.: "The intricate regulation and complex functions of the Class III phosphoinositide 3-kinase Vps34", BIOCHEM J., vol. 473, no. 15, 2016, pages 2251 - 2271
FRUMAN, D. A.CHIU H.HOPKINS B. D.BAGRODIA, S.CANTLEY, L. C.ABRAHAM R. T.: "The PI3K pathway in human disease", CELL, vol. 170, no. 4, 2017, pages 605 - 635, XP085153697, DOI: 10.1016/j.cell.2017.07.029
PORTA, C.PAGLINO, C.MOSCA, A.: "Targeting PI3K/Akt/mTOR signaling in cancer", FRONT ONCOL., vol. 4, no. 64, 2014, pages 1 - 11
VANHAESEBROECK ET AL., J MOL MED (BERL)., vol. 94, 2016, pages 5
HAWKINS, P. T.STEPHENS, L. R.: "PI3K signalling in inflammation", BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1851, no. 6, 2015, pages 882 - 897
OKKENHAUG, K.BILANCIO, A.FARJOT, G.PRIDDLE, H.SANCHO, S.PESKETT, E.PEARCE, W.MEEK, S. E.SALPEKAR, A.WATERFIELD, M. D.: "Impaired Band T cell antigen receptor signaling in p110delta PI 3-kinase mutant mice", SCIENCE, vol. 297, no. 5583, 2002, pages 1031 - 1034
ALI, K.BILANCIO, A.THOMAS, M.PEARCE, W.GILFILLAN, A. M.TKACZYK, C.KUEHN, N.GRAY, A.GIDDINGS, J.PESKETT, E.: "Essential role for the p110delta phosphoinositide 3-kinase in the allergic response", NATURE, vol. 431, no. 7011, 2004, pages 1007 - 1011
SOLER ET AL., J EXP MED., vol. 210, 2013, pages 1937
LIU, P.CHENG, H.ROBERTS, T. M.ZHAO, J. J.: "Targeting the phosphoinositide 3-kinase pathway in cancer", NAT REV DRUG DISCOV., vol. 8, no. 8, 2009, pages 627 - 644, XP055020926, DOI: 10.1038/nrd2926
JACKSON, S. P.SCHOENWAELDER, S. M.GONCALVES, I.NESBITT, W. S.YAP, C. L.WRIGHT, C. E.KENCHE, V.ANDERSON, K. E.DOPHEIDE, S. M.YUAN, : "PI 3-kinase p110beta: a new target for antithrombotic therapy", NAT MED., vol. 11, no. 5, 2005, pages 507 - 514, XP007910884, DOI: 10.1038/nm1232
YANG, J.NIE, J.MA, X.WEI, Y.PENG, Y.WEI, X: "Targeting PI3K in cancer: mechanisms and advances in clinical trials", MOL CANCER, vol. 18, no. 26, 2019, pages 1 - 28
GONCALVES ET AL., N ENG J MED., vol. 379, 2018, pages 2052
THORPE, L. M.YUZUGULLU, H.ZHAO, J. J.: "PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting", NAT REV CANCER, vol. 15, no. 1, 2015, pages 7 - 24
JIANG, N.DAI, Q.SU, X.FU, J.FENG, X.: "Role of PI3K/AKT pathway in cancer: the framework of malignant behavior", MOL BIOL REP., vol. 47, no. 6, 2020, pages 4587 - 4629, XP037170870, DOI: 10.1007/s11033-020-05435-1
HOPKINS, B. D.PAULI, C.DU, X.WANG, D. G.LI, X.WU, D.AMADIUME, S. C.GONCALVES, M. D.HODAKOSKI, C.LUNDQUIST, M. R.: "Suppression of insulin feedback enhances the efficacy of PI3K inhibitors", NATURE, vol. 560, no. 7719, 2018, pages 499 - 503, XP036579402, DOI: 10.1038/s41586-018-0343-4
RUGO, H. S.LACOUTURE, M. E.GONCALVES, M. D.MASHARANI, U.AAPRO, M. S.O'SHAUGHNESSY, J. A.: "A multidisciplinary approach to optimizing care of patients treated with alpelisib", THE BREAST, vol. 61, 2022, pages 156 - 167, XP086949899, DOI: 10.1016/j.breast.2021.12.016
GADKAR, K.FRIEDRICH, C.HUREZ, V.RUIZ, M. L.DICKMANN, L.JOLLY, M. K.SCHUTT, L.JIN, J.WARE, J. A.RAMANUJAN, S.: "Quantitative systems pharmacology model-based investigation of adverse gastrointestinal events associated with prolonged treatment with PI3-kinase inhibitors", CPT PHARMACOMETRICS SYST PHARMACOL., vol. 11, no. 5, 2021, pages 616 - 627
CASTEL, P.TOSKA, E.ENGELMAN, J. A.SCALTRITI, M.: "The present and future of PI3K inhibitors for cancer therapy", NAT CANCER, vol. 2, no. 6, 2021, pages 587 - 597
LEMIEUX, G. A.BERTOZZI, C. R.: "Chemoselective ligation reactions with proteins, oligosaccharides and cells", TRENDS IN BIOTECHNOLOGY, vol. 16, no. 12, 1998, pages 506 - 513, XP004143811, DOI: 10.1016/S0167-7799(98)01230-X
HANG, H.C.BERTOZZI, C. R.: "Chemoselective approaches to glycoproteon assembly", ACCOUNTS OF CHEMICAL RESEARCH, vol. 34, no. 9, 2001, pages 727 - 736, XP001099796, DOI: 10.1021/ar9901570
KIICK ET AL., PROC NATL ACAD SCI US.A., vol. 99, 2002, pages 19
KEPPLER-NOREUIL, K. M.RIOS, J. J.PARKER, V. E.SEMPLE, R. K.LINDHURST, M. J.SAPP, J. C.ALOMARI, A.EZAKI, M.DOBYNS, W.BIESECKER, L. : "PIK3CA-related overgrowth spectrum (PROS): diagnostic and testing eligibility criteria, differential diagnosis, and evaluation", AM J MED GENET A., vol. 167, no. 2, 2015, pages 287 - 295, XP055683886, DOI: 10.1002/ajmg.a.36836
KUREK ET AL., AM. J. HUM. GENET., vol. 90, 2012, pages 1108
ORG. LETT., 2020, pages 1648 - 1654
ORGANIC PROCESS RESEARCH & DEVELOPMENT, 2021, pages 642 - 647
ANGEW. CHEMIE INT. ED., 2017, pages 12774 - 12777
NATURE CHEMISTRY, 2022, pages 1068 - 1077
KLAUS ET AL., ANGEW. CHEM. INT. ED., vol. 45, 2006, pages 154
PRAKASH ET AL., J. AM. CHEM. SOC., vol. 111, 1989, pages 393
ZHAO, Y.HUANG, W.ZHENG, J.HU, J.: "Efficient and direct nucleophilic difluoromethylation of carbonyl compounds and imines with Me SiCF H at ambient or low temperature", ORG. LETT., vol. 13, no. 19, 2011, pages 5342 - 5345, XP055951346, DOI: 10.1021/ol202208b
REICHEL ET AL., ANGEW. CHEM. INT. ED., vol. 59, 2020, pages 12268
TIAN ET AL., MOLECULAR CATALYSIS, 2021, pages 111345
FANG ET AL., ORGANIC & BIOMOLECULAR CHEMISTRY, vol. 10, no. 12, 2012, pages 2389
LI ET AL., J. ORG. CHEM., vol. 80, no. 19, 2015, pages 9392
ZHANG ET AL., TETRAHEDRON LETT., vol. 66, 2021, pages 152835
HIKAWA ET AL., J. ORG. CHEM., vol. 77, no. 16, 2012, pages 7046
GARAD ET AL., J. ORG. CHEM., vol. 82, no. 12, 2017, pages 6366
KIM ET AL., TETRAHEDRON LETT., vol. 55, no. 15, 2014, pages 2340
QIAN ET AL., J. ORG. CHEM., vol. 83, no. 16, 2018, pages 9201
SHALIT, T. ET AL., TETRAHEDRON LETT., vol. 51, 2010, pages 5988 - 5991
IRANPOOR, N. ET AL., TETRAHEDRON, vol. 65, 2009, pages 3893 - 3899
PANDAY, S. K., MINI-REV. ORG. CHEM., vol. 16, no. 2, 2019, pages 127 - 140
FUKUYAMATOHRU ET AL., TETRAHEDRON LETT., vol. 38, no. 33, 1997, pages 5831 - 5834
DATTAELLMAN, J. ORG. CHEM., vol. 75, 2010, pages 6283 - 6285
ELLMAN ET AL., ACC. CHEM. RES., vol. 35, 2002, pages 984 - 995
ELLMAN ET AL., J. ORG. CHEM., vol. 72, 2007, pages 626 - 629
COLYER ET AL., J. ORG. CHEM., vol. 71, no. 18, 2006, pages 6859 - 6862
HUA ET AL., SYNTHESIS, vol. 970, no. 11, 1991, pages 4
ZHU ET AL., J. CHEM. RES., vol. 39, no. 7, 2015, pages 390 - 393
YANG ET AL., ORG. PROCESS RES. & DEV., vol. 26, no. 6, 2022, pages 1690 - 1750
SURRYBUCHWALD, CHEM. SCI., vol. 2, no. 1, 2011, pages 27 - 50
GONCALVES, M. D.HOPKINS, B. D.CANTLEY, L. C.: "Phosphatidylinositol 3-Kinase, growth disorders, and cancer", N ENGL J MED., vol. 379, no. 21, 2018, pages 2052 - 2062
KIICK, K. L.SAXON, E.TIRRELL, D. A.BERTOZZI, C. R.: "Incorporation of azides into recombinant proteins for chemoselective modification by the Staudinger ligation", PROC NATL ACAD SCI USA., vol. 99, no. 1, 2002, pages 19 - 24, XP002904363, DOI: 10.1073/pnas.012583299
KLAUS, S.NEUMANN, H.ZAPF, A.STRUBING, D.HUBNER, S.ALMENA, J.RIERMEIER, T.GROΒ, P.SARICH, M.KRAHNERT, W.: "A general and efficient method for the formylation of aryl and heteroaryl bromides", ANG CHEM INT ED., vol. 45, no. 1, 2006, pages 154 - 158, XP002512982, DOI: 10.1002/ANIE.200502697
KUREK, K. C.LUKS, V. L.AYTURK, U. M.ALOMARI, A. I.FISHMAN, S. J.SPENCER S. A.MULLIKEN, J. S.BOWEN, M. E.YAMAMOTO, G. L.KOZAKEWICH,: "Somatic mosaic activating mutations in PIK3CA cause CLOVES syndrome", AM J HUM GENET., vol. 90, no. 6, 2012, pages 1108 - 1115, XP028522209, DOI: 10.1016/j.ajhg.2012.05.006
PRAKASH, G.KRISHNAMURTI, R.OLAH, G.: "Synthetic methods and reactions. 141. Fluoride-induced trifluoromethylation of carbonyl compounds with trifluormethyltrimethylsilane (TMS-CF ). A trifluoromethide equivalent", JAM CHEM SOC., vol. 111, no. 1, 1989, pages 393 - 395
REICHEL, M.KARAGHIOSOFF, K.: "Reagents for selective fluoromethylation: A challenge in organofluorine chemistry", ANG CHEM INT ED., vol. 59, no. 30, 2020, pages 12268 - 12281, XP072094647, DOI: 10.1002/anie.201913175
SOLER, A.SERRA, H.PEARCE, W.ANGULO, A.GUILLERMET-GUIBERT, J.FRIEDMAN, L. S.VINALS, F.GERHARDT, H.CASANOVAS, O.GRAUPERA, M.: "Inhibition of the p110a isoform of PI 3-kinase stimulates nonfunctional tumor angiogenesis", J EXP MED., vol. 210, no. 10, 2004, pages 1937 - 1945
VANHAESEBROECK, B.WHITEHEAD M. A.PINEIRO, R.: "Molecules in medicine mini-review: isoforms of PI3K in biology and disease", J MOL MED (BERL, vol. 94, no. 1, 2016, pages 5 - 11, XP035801328, DOI: 10.1007/s00109-015-1352-5
Attorney, Agent or Firm:
LOWEN, Gregory T. (US)
Download PDF:
Claims:
IN THE CLAIMS

1. A compound of Formula (1) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein:

Ri is alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted;

R2 is H, Ci-C4 alkyl, C3-C7 cycloalkyl, CF3, CH2F or CF2H, and where R2 is not H, the carbon atom attached to R2 is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer;

R3 is H or C1-C4 alkyl;

Rs is H, C1-C4 alkyl, C3-C7 cycloalkyl, heteroaryl, CF3, CH2F or CF2H;

Rs is H, C1-C4 alkyl, C3-C7 cycloalkyl, halogen, CN, CF3, OCF3, OCH3, CH2F or CF2H; each R7 is independently H, C1-C4 alkyl, C3-C7 cycloalkyl, halogen, CN, CF3, OCF3, OCH3, CH2F or CF2H;

R4 is -LI-L2-L3-L4-LS-RS;

LI is CHRg, (CHR9-O), (CHRg-S), (CHRg-NR10), C=O, C=S or a bond;

L2 is cycloalkyl that is optionally part of a bridged, fused or spiro ring system, C(Rg)=C(Rg), C=C or a bond;

L3 is independently CHRg, O, S, NCRg, N(C=O) or a bond;

L4 is CHRg, C=O, C=S or a bond; L5 is H, cycloalkyl, heterocyclyl, aryl, heteroaryl or a bond, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and the cycloalkyl and/or heterocyclyl is optionally part of a bridged, fused or spiro ring system;

Rs is H, -CR9R10R11, -OR11, -SR11, -NR10R11, C(=O)Rn, C(=O)NRgRn, C(=O)ORi2, C1-C6 alkyl, C1-C6 fluoroalkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the C1-C6 alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and the cycloalkyl and/or heterocyclyl is optionally part of a bridged, fused or spiro ring system; each of Rg and R10 is independently H or C1-C3 alkyl; and each Rn is independently H, C1-C6 alkyl, C1-C6 fluoroalkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the C1-C6 alkyl, C1-C6 fluoroalkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted; or alternatively, R10 and R1 together with the attached nitrogen atom may form a ring; with the proviso that for R4, at least one of Li, L2, L3, L4, L5 and R8 is a carbon-containing moiety and R4 is directly attached to the (benzopyrimidin-4(3H)-one) core structure by a carbon atom.

2. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L3 and L4 is a bond and L2 is not a bond.

3. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L3 and L4 is a bond and L2 is cycloalkyl that is optionally part of a bridged, fused or spiro ring system.

4. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L3 and L4 is a bond and L2 is cycloalkyl that is part of a bridged ring system.

5. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L3 and L4 is a bond and L2 is cycloalkyl that is part of a fused ring system.

6. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L3 and L4 is a bond and L2 is cycloalkyl that is part of a spiro ring system.

7. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L3 and L4 is a bond and L2 is C(Rg)=C(Rg).

8. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L3 and L4 is a bond and L2 is CEC.

9. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L2, L3 and L4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted.

10. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L2, L3 and L4 is a bond and L5 is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and R8 is H.

11. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L2, L3 and L4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and R8 is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted.

12. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L2, L3 and L4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and R8 is -CRgR10Rn.

13. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L2, L3 and L4 is a bond and L5 is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and R8 is -OR11.

14. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L2, L3 and L4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and Rs is -SR11.

15. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein for R4, each of Li, L2, L3 and L4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and Rs is -N R10R11.

16. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein R2 is CH3 or CH2F.

17. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein R3 is H.

18. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein Rs is CH3.

19. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein Rs is CH3.

20. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein each R? is independently H or F.

21. The compound according to claim 1 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein R2, RS and Rs are CH3.

22. The compound according to claim 1, wherein the compound of Formula (1) is a compound of Formula (2)

or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein:

R4 and R7 are as defined in the compound of Formula (1); the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer; and the listing of substituents within brackets indicates individual compounds containing one of each of the substituents.

23. The compound according to claim 1, wherein the compound of Formula (1) is a compound of Formula (3)

or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein: each of Xi, X2 and X3 is independently N, CH or substituted C, where at least one of Xi, X2 and X3 is N;

R4 and R7 are as defined in the compound of Formula (1); the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer; and the listing of substituents within brackets indicates individual compounds containing one of each of the substituents.

24. The compound according to claim 1, wherein the compound of Formula (1) is a compound of Formula (4)

or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein: each of X1, X2 and X3 is independently N, CH or substituted C;

R7 is as defined in the compound of Formula (1); the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer; the listing of substituents within brackets indicates individual compounds containing one of each of the substituents; and

R4 is selected from the group consisting of: [H or CrC4 alkyl] wherein L3, L4, L5 and R8 are as defined in the compound of Formula (1).

25. The compound according to claim 1, wherein the compound of Formula (1) is a compound of Formula (5) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein: the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer, the listing of substituents within brackets indicates individual compounds containing one of each of the substituents, and the heteroaryl or heterocyclyl rings linked to the bicyclo[l.l.l]pentane moiety are unsubstituted or substituted.

26. The compound according to claim 1, wherein the compound of Formula (1) is a compound of Formula (6) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein:

Ri is a substituted or unsubstituted heterocyclyl, aryl or heteroaryl, wherein the heterocyclyl, aryl or heteroaryl ring directly attached to the nitrogen atom linked to the asymmetric center attached to the isoquinoline moiety contains a carboxylic acid substituent at the ortho position to the point of attachment;

R4 and R7 are defined as in the compound of Formula (1); the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer; and the listing of substituents within brackets indicates individual compounds containing one of each of the substituents.

27. A pharmaceutical composition comprising the compound of any one of claims 1 to 26 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.

28. The pharmaceutical composition according to claim 27, further comprising one or more anti-cancer agents.

29. The pharmaceutical composition according to claim 28, wherein the one or more anticancer agents are selected from the group consisting of cyclophosphamide, dacarbazine, cisplatin, methotrexate, mercaptopurine, thioguanine, fluorouracil, cytarabine, vinblastine, paclitaxel, doxorubicin, bleomycin, mitomycin, prednisone, tamoxifen, flutamide, asparaginase, rituximab, trastuzumab, imatinib, retinoic acid, amifostine, camptothecin, topotecan, thalidomide, lenalidomide, a CDK inhibitor and a proteasome inhibitor.

30. A method of treating a disease in which PI3K activity is implicated in a subject in need of such treatment, the method comprising administering to the subject a therapeutically effective amount of the compound of any one of claims 1 to 26 or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof.

31. The method of claim 30, wherein the disease is cancer.

32. The method of claim 30, wherein the disease is congenital lipomatous overgrowth, vascular malformations, epidermal naevi, scoliosis/skeletal and spinal syndrome (CLOVES), mosaic tissue overgrowth syndromes, venous malformations and brain malformations associated with severe epilepsy or PIK3CA-related overgrowth syndrome (PROS).

33. The method of claim 30, wherein the disease is a cancer bearing a PI3Ka H1047R mutation.

Description:
((4-OXO-3,4-DIHYDROQUINAZOLIN-8-YL)METHYL)AMINE DERIVATIVES AS P13K INHIBITORS FOR THE TREATMENT OF CANCER

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims priority to U.S. Provisional Application No. 63/407,873, filed September 19, 2022, the disclosure of which is incorporated in its entirety for all purposes.

BACKGROUND OF THE INVENTION

[0002] Phosphatidylinositol lipids (Pls) and their various phosphorylated subspecies are second messengers involved in a wide array of cellular vesicle trafficking and signal transduction processes. Phosphoinositide 3' kinases (PI3Ks) are a family of enzymes responsible for phosphorylation of the 3' hydroxyl position of the inositol ring of Pls. PI3Ks are subdivided into 3 classes according to their structure and substrates. Class II PI3Ks (PI3K-C2a, PI3K-C2P, PI3K- C2y) and Class III PI3Ks (vps34) are monomeric enzymes primarily associated with endocytosis and autophagy (Posor et al., Biochim Biophys Acta 2015, 1851, 794; Backer, Biochem J. 2016, 473, 2251). The Class I PI3Ks are heterodimeric, consisting of a catalytic kinase subunit (pllOa, P, y, 6) and one of several regulatory subunits that determine binding partners and subcellular localization. Class I PI3Ks are activated upon interaction with receptor tyrosine kinases (RTKs), Ras-related GTPases, G-protein coupled receptors, and/or related adaptor proteins, and in their active form convert phosphatidylinositol 4,5-diphosphate (PIP2) to phosphatidyl 3,4,5- triphosphate (PIP3) (Fruman et al., Cell 2017, 170, 605).

[0003] High local concentrations of PIP3 promote the recruitment and activation of downstream signaling partners, including AKT and mTOR. Activation of the AKT/mTOR pathways are implicated in several growth-related roles and pathologies including glucose regulation, cell survival, angiogenesis, and proliferation (Porta et al., Front Oncol. 2014, 4, 1), indicating a role for Class I PI3Ks as a critical upstream regulator of these functions.

[0004] Class I PI3Ks are further subdivided into 4 isoforms (a, p, y, and 6) based on the identity of their catalytic (pllOa, pliop, pllOy, or p1108) and regulatory (p85a or its various splice variants, p85P, p55y, or plOl) subunits, giving rise to distinct roles in cellular physiology (Vanhaesebroeck et al., J Mol Med (Berl). 2016, 94, 5). PI3Ky and PI3K6 are mostly expressed in leukocytes and play an important role in pro-inflammatory pathways (Hawkins et. al., Biochimica et Biophysica Acta 2015, 1851, 882; Okkenhaug et al., Science 2002, 297, 1031; Ali et al., Nature 2004, 431, 1007). PI3Ka and p are more ubiquitously expressed and share similar but not identical roles. For example, PI3Ka has a nonredundant role in angiogenesis (Soler et al., J Exp Med. 2013, 210, 1937), while PI3KfJ is known to serve a specific function in platelet aggregation (Liu et. al., Nat Rev Drug Discov. 2009, 8, 627; Jackson et al., Nat Med. 2005, 11, 507).

[0005] Elevation or constitutive activation of the PI3K pathway is one of the most frequent events in human cancers. The PI3K pathway is overactivated through a variety of mechanisms, including activating mutation of PI3K isoforms, up-regulation of PI3K isoforms, loss or inactivation of the tumor suppressor PTEN, or hyperactivation of tyrosine kinase growth factor receptors or other upstream signaling partners (Yang et al., Mol Cancer 2019, 18, 1). Mutations in the gene coding for PI3Ka or mutations which lead to upregulation of PI3Ka have been found to occur in many human cancers such as lung, stomach, endometrial, ovarian, bladder, breast, colon, brain, prostate, and skin cancers (Goncalves et al., N Eng J Med. 2018, 379,2052). In particular, PIK3CA, the gene encoding the pllOa subunit of PI3Ka, is frequently mutated or amplified in a variety of tumor types. Missense mutations occur in all domains of pllOa, but cluster in two 'hot spots', the most common being E542K and E545K in the helical domain, and H1047R in the kinase domain. Helical domain mutations reduce inhibition of pllOa by p85 or facilitate direct interaction of pllOa with insulin receptor substrate 1 (IRS1)37, whereas kinase domain mutations increase interaction of pllOa with lipid membranes, concomitantly upregulating signaling events. (Thorpe et al., Nat Rev Cancer 2015, 15, 7).

[0006] The development of inhibitors for the PI3K pathway has been challenging due to the inability to achieve dosing sufficient for tumor suppression without adverse events. To date PI3K inhibitors in the clinic (alpelisib, buparlisib, copanlisib, duvelisib, idelal isib, pictilisib, taselisib, and others) have caused dose-dependent adverse events such as hyperglycemia, rash, fatigue, diarrhea, etc. (Jiang et al., Mol Biol Rep. 2020, 47, 4587) which are known on- target toxicities. Hyperglycemia is a result of the body not producing enough insulin or aberrant utilization. The pancreas regulates insulin release in response to changes in blood glucose levels, resulting in either glucose uptake by muscle and fat cells when insulin levels are high or gluconeogenesis by the liver when insulin levels are low. Tissue cellular response to insulin requires PI3K signaling through the ubiquitously expressed pllOa sub-unit. As a result, pan-PI3K inhibition of the target disrupts glucose metabolism in tissues, leading to insulin resistance (Hopkins et al., Nature 2018, 560, 499). To mitigate adverse events, selective PI3K isoform inhibitors were developed. The severity of the adverse event is dependent on the select isoform, for example PI3Ka inhibitors are associated with hyperglycemia and rash due to the pllOa sub-unit role in insulin response (Rugo et al., The Breast 2022, 61, 156). Similarly, use of a selective PI3K6 inhibitor (idelalisib), where the pll06 sub-unit is highly expressed in immune cells, causes severe diarrhea and colitis. Inhibition with a dual inhibitor (taselisib), a potent PI3K6 inhibitor possessing modest PI3Ka inhibition led to gastrointestinal (Gl) side effects, but a highly selective and potent PI3K6 inhibitor (umbralisib) reported no Gl related adverse events (Gadkar et al., CPT Pharmacometrics Syst Pharmacol. 2021, 11, 616). Such amelioration of adverse events with highly isoform selective and potent inhibitors demonstrates that a strategy to mitigate toxicity by developing mutant selective isoform inhibitors is promising for decreasing the severity of toxicity. Furthermore, selective inhibition of the mutant PI3Ka isoform over wild type may suppress cancer signaling while having minimal effect on PI3K signaling in healthy cells bearing just wild type PI3Ka, leading to a reduction in the toxicities associated with nonselective PI3K inhibition (Castel et al., Nat Cancer 2021 2, 587).

[0007] There is currently an interest in developing PI3K inhibitors for cancer therapy (WO 2023/081209, WO 2023/078401, WO 2023/060262, WO 2023/056407, WO 2021/202964, WO 2023/159155). However, there is a continued need for novel potent and selective PI3K inhibitors, either as single agents or as combination therapies, in the treatment of cancer.

SUMMARY OF THE INVENTION

[0008] An aspect of the invention is a compound of Formula (1) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound thereof, or a pharmaceutically acceptable salt thereof, wherein: Ri is alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted;

R2 is H, C1-C4 alkyl, C3-C7 cycloalkyl, CF 3 , CH 2 F or CF2H, and where R2 is not H, the carbon atom attached to R2 is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer;

R3 is H or C1-C4 alkyl;

Rs is H, C1-C4 alkyl, C3-C7 cycloalkyl, heteroaryl, CF 3 , CH 2 F or CF2H;

Rs is H, C1-C4 alkyl, C3-C7 cycloalkyl, halogen, CN, CF 3 , OCF 3 , OCH3, CH 2 F or CF2H; each R7 is independently H, C1-C4 alkyl, C3-C7 cycloalkyl, halogen, CN, CF 3 , OCF 3 , OCH3, CH 2 F or CF 2 H;

R4 is L1-L 2 -L3-L4-L5-R8;

Li is CHR 9 , (CHR 9 -O), (CHRg-S), (CHRg-NR 10 ), C=O, C=S or a bond;

L 2 is cycloalkyl that is optionally part of a bridged, fused or spiro ring system, C(R 9 )=C(R 9 ), C=C or a bond;

L 3 is independently CHR 9 , O, S, NCR 9 , N(C=O) or a bond;

L4 is CHRg, C=O, C=S or a bond;

Ls is H, cycloalkyl, heterocyclyl, aryl, heteroaryl or a bond, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and the cycloalkyl and/or heterocyclyl is optionally part of a bridged, fused or spiro ring system;

Rs is H, -CRgR 10 Rn, -ORn, -SRn, -NR10R11, C(=O)Rn, C(=O)NRgRn, C(=O)ORi2, Ci-Cs alkyl, Ci-Cs fluoroalkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the Ci-Cs alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and the cycloalkyl and/or heterocyclyl is optionally part of a bridged, fused or spiro ring system; each of R 9 and R 10 is independently H or C1-C3 alkyl; and each Rn is independently H, C 1 -C 6 alkyl, C 1 -C 6 fluoroalkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the Ci-C 6 alkyl, Ci-C 6 fluoroalkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted; or alternatively, R i0 and Rn together with the attached nitrogen atom may form a ring. In an exemplary embodiment, the ring is 4- to 7- membered substituted or unsubstituted non-aromatic heterocyclic ring containing (in addition to the nitrogen atom) 0, 1 or 2 heteroatoms which may be N, O, S or Si, with the proviso that if the ring size is 4 or 5, the number of additional heteroatoms will be 0 or 1 and if the ring size is from 6 to 7, the number of additional heteroatoms will be 0, 1 or 2, where if the ring is substituted, the substituents include, but are not limited to, one or more of CH3, F, Cl, CF 3 , CF2H, CH 2 F, OCH3, cyclopropyl, CH 2 CF 3 , an oxetane ring, or COR a where R a is C1-C4 alkyl, O-C1-C4 alkyl, or NRbRc where Rb and R c are independently H or Ci-C4alkyl; with the proviso that for R4, at least one of Li, L 2 , L3, L4, Ls and Rs is a carbon-containing moiety and R4 is directly attached to the (benzopyrimidin-4(3H)-one) core structure by a carbon atom.

[0009] In an exemplary embodiment of R4, each of Li, L3 and L4 is a bond and L 2 is not a bond.

[00010] In an exemplary embodiment of R4, each of Li, L3 and L4 is a bond and L 2 is cycloalkyl that is optionally part of a bridged, fused or spiro ring system.

[00011] In an exemplary embodiment of R4, each of Li, L3 and L4 is a bond and L 2 is cycloalkyl that is part of a bridged ring system.

[00012] In an exemplary embodiment of R 4 , each of Li, L 3 and L 4 is a bond and L 2 is cycloalkyl that is part of a fused ring system.

[00013] In an exemplary embodiment of R4, each of Li, L3 and L4 is a bond and L 2 is cycloalkyl that is part of a spiro ring system.

[00014] In an exemplary embodiment of R4, each of Li, L3 and L4 is a bond and L 2 is C(Rg)=C(Rg).

[00015] In an exemplary embodiment of R4, each of Li, L3 and L4 is a bond and L 2 is CEC.

[00016] In an exemplary embodiment of R4, each of Li, L 2 , L3 and L4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted.

[00017] In an exemplary embodiment of R4, each of Li, L 2 , L3 and L4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and Rs is H.

[00018] In an exemplary embodiment of R 4 , each of Li, L 2 , L 3 and L 4 is a bond and L 5 is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and R 8 is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the Ci-Cs alkyl, cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted. [00019] In an exemplary embodiment of R 4 , each of Li, L 2 , L 3 and L 4 is a bond and L 5 is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and Rs is -CRgR 10 Ru.

[00020] In an exemplary embodiment of R 4 , each of Li, L 2 , L 3 and L 4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and Rs is -OR 10 or -ORn.

[00021] In an exemplary embodiment of R 4 , each of Li, L 2 , L 3 and L 4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and Rs is -SRn.

[00022] In an exemplary embodiment of R 4 , each of Li, L 2 , L 3 and L 4 is a bond and Ls is cycloalkyl, heterocyclyl, aryl or heteroaryl, where each of the cycloalkyl, heterocyclyl, aryl or heteroaryl is unsubstituted or substituted, and Rs is -NR 10 Rn.

[00023] In an exemplary embodiment of the compound of Formula (1), R 2 is CH 3 or CH 2 F.

[00024] In an exemplary embodiment of the compound of Formula (1), R 3 is H.

[00025] In an exemplary embodiment of the compound of Formula (1), R 5 is CH 3 .

[00026] In an exemplary embodiment of the compound of Formula (1), Rs is CH 3 .

[00027] In an exemplary embodiment of the compound of Formula (1), R? is H.

[00028] In an exemplary embodiment of the compound of Formula (1), R 2 , Rs and Rs are CH 3 .

[00029] In an exemplary embodiment of the compound of Formula (1), Ri is heterocyclyl, aryl or heteroaryl, where each of the heterocyclyl, aryl or heteroaryl is unsubstituted or substituted.

[00030] In an exemplary embodiment of the compound of Formula (1), Ri is heterocyclyl, where the heterocyclyl is unsubstituted or substituted.

[00031] In an exemplary embodiment of the compound of Formula (1), Ri is aryl, where the aryl is unsubstituted or substituted.

[00032] In an exemplary embodiment of the compound of Formula (1), Ri is heteroaryl, where the heteroaryl is unsubstituted or substituted.

[00033] In an exemplary embodiment of the compound of Formula (1), Ri is heterocyclyl, where the heterocyclyl is unsubstituted or substituted, R 2 is CH 3 and R 3 is H. [00034] In an exemplary embodiment of the compound of Formula (1), Ri is aryl, where the aryl is unsubstituted or substituted, R 2 is CH 3 and R 3 is H.

[00035] In an exemplary embodiment of the compound of Formula (1), Ri is heteroaryl, where the heteroaryl is unsubstituted or substituted, R 2 is CH 3 and R 3 is H.

[00036] In an exemplary embodiment of the compound of Formula (1), Ri is heterocyclyl, where the heterocyclyl is unsubstituted or substituted, R 2 is CH 3 , R 3 is H and R 7 is H.

[00037] In an exemplary embodiment of the compound of Formula (1), Ri is aryl, where the aryl is unsubstituted or substituted, R 2 is CH 3 , R 3 is H and R 7 is H.

[00038] In an exemplary embodiment of the compound of Formula (1), Ri is heteroaryl, where the heteroaryl is unsubstituted or substituted, R 2 is CH 3 , R 3 is H and R 7 is H.

[00039] In an exemplary embodiment of the compound of Formula (1), Ri is heterocyclyl, where the heterocyclyl is unsubstituted or substituted, R 2 is CH 3 , R 3 is H, R 7 is H and Rs is CH 3 .

[00040] In an exemplary embodiment of the compound of Formula (1), Ri is aryl, where the aryl is unsubstituted or substituted, R 2 is CH 3 , R 3 is H, R 7 is H and R 5 is CH 3 .

[00041] In an exemplary embodiment of the compound of Formula (1), Ri is heteroaryl, where the heteroaryl is unsubstituted or substituted, R 2 is CH 3 , R 3 is H, R 7 is H and Rs is CH 3 .

[00042] In an exemplary embodiment of the compounds of Formula (1) where R 2 is not H, the carbon atom attached to R 2 exists as the (R)-enantiomer.

[00043] In an exemplary embodiment, the compound of Formula (1) is a compound of Formula (2) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein R 2 and R 4 are defined as in the compound of Formula (1) and the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer. The listing of substituents within brackets indicates individual compounds containing one of each of the substituents.

[00044] In an exemplary embodiment, the compound of Formula (1) is a compound of Formula (3) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein: each of Xi, X 2 and X 3 is independently N, CH or substituted C, where at least one of Xi, X 2 and X 3 is N;

R 4 and R 7 are defined as in the compound of Formula (1), the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer, and the listing of substituents within brackets indicates individual compounds containing one of each of the substituents.

[00045] In an exemplary embodiment of the compound of Formula (3), Xi is N, X 2 is CH or substituted C, and X 3 is CH or substituted C.

[00046] In an exemplary embodiment of the compound of Formula (3), X 2 is N, Xi is CH or substituted C, and X 3 is CH or substituted C. [00047] In an exemplary embodiment of the compound of Formula (3), X 3 is N, Xi is CH or substituted C, and X 2 is CH or substituted C.

[00048] In an exemplary embodiment of the compound of Formula (3), Xi and X 3 are N, and X 2 is CH or substituted C.

[00049] In an exemplary embodiment of the compound of Formula (3), Xi, X 2 and X 3 are N.

[00050] In an exemplary embodiment, the compound of Formula (1) is a compound of Formula (4) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein: each of Xi, X 2 and X 3 is independently N, CH or substituted C;

R 7 is defined as in the compound of Formula (1), the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer, the listing of substituents within brackets indicates individual compounds containing one of each of the substituents, and

R4 is selected from the group consisting of: wherein L 3 , L 4 , Ls and Rs is defined as in the compound of Formula (1).

[00051] In an exemplary embodiment of the compound of Formula (4), Xi is N, X2 is CH or substituted C, and X 3 is CH or substituted C.

[00052] In an exemplary embodiment of the compound of Formula (4), X2 is N, Xi is CH or substituted C, and X 3 is CH or substituted C.

[00053] In an exemplary embodiment of the compound of Formula (4), X 3 is N, Xi is CH or substituted C, and X 2 is CH or substituted C.

[00054] In an exemplary embodiment of the compound of Formula (4), Xi and X 3 are N, and X 2 is CH or substituted C.

[00055] In an exemplary embodiment of the compound of Formula (4), Xi, X 2 and X 3 are N.

[00056] In an exemplary embodiment of the compound of Formula (4), Xi, X2 and X 3 are CH or substituted C.

[00057] In an exemplary embodiment, the compound of Formula (1) is a compound of Formula (5) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein: the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer, the listing of substituents within brackets indicates individual compounds containing one of each of the substituents, and the heteroaryl or heterocyclyl rings linked to the bicyclo[l.l.l]pentane moiety are unsubstituted or substituted.

[00058] In an exemplary embodiment of Formula (5), the fluoroalkyl linked to the bicyclo[l.l.l]pentane moiety is CF 3 , CHF 2 , CH 2 F, CF 2 CH 3 , CFHCH 3 , CH 2 CF 3 , OCF 3 , OCHF 2 , OCH 2 F, OCF 2 CH 3 , OCFHCH 3 or OCH 2 CF 3 .

[00059] In an exemplary embodiment of Formula (5), the heteroaryl linked to the bicyclo[l.l.l]pentane moiety is furan, benzofuran, thiophene, benzothiophene, pyrrole, indole, isoindole, 7-azaindole, 4-azaindole, 5-azaindole, 6-azaindole, 7-azaindazole, pyridine, quinoline, isoquinoline, oxazole, isoxazole, benzoxazole, pyrazole, imidazole, benzimidazole, thiazole, benzothiazole, isothiazole, 1,2,4-triazole, 1,2,3-triazole, tetrazole, 1,2,5-oxadiazole, 1,2,3-oxadiazole, 1,3,4-thiadiazole, pyridazine, pyrimidine, pyrazine, 1,2,4-triazine or 1,3,5- triazine, where the heteroaryl is either unsubstituted or substituted with one or more fluorine atoms and/or fluoroalkyl groups.

[00060] In an exemplary embodiment of Formula (5), the heterocyclyl linked to the bicyclo[l.l.l]pentane moiety is azirine, aziridine, azetidine, 2,3-dihydroazete, 1,3-diazetidine, 2H-oxete, thietane, 2H-thiete, azetidin-2-one, morpholine, thiomorpholine, pyrrolidinone, pyrrolidinine, 2-pyrroline, 3-pyrroline, pyrazolidine, 2-pyrazoline, 2-imidazoline, imidazolidine, piperidine, piperazine, pyridin-2-ones, oxirane, thiirane, oxetane, propylene oxide, 1,3- dioxolane, 1,2-oxathiolane, 1,3-oxathiolane, sulfolane, 2,4-thiazolidinedione, succinimide, 2- oxazolidone, dioxane, hydantoin, valerolactam, tetrahydrofuran, tetrahydropyran, 2H-pyran, 4H-pyran, thiane, 2H-thiopyran, 1,3-dithiane, 1,4-dithiane, 1,3,5-trithiane or pyrrolizidine, where heterocyclyl is either unsubstituted or substituted with one or more fluorine atoms and/or fluoroalkyl groups. [00061] In an exemplary embodiment, the compound of Formula (1) is a compound of Formula (6) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein:

Ri is a substituted or unsubstituted heterocyclyl, aryl or heteroaryl, wherein the heterocyclyl, aryl or heteroaryl ring directly attached to the nitrogen atom linked to the asymmetric center attached to the isoquinoline moiety contains a carboxylic acid substituent at the ortho position to the point of attachment,

R4 and R7 are defined as in the compound of Formula (1), the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer, and the listing of substituents within brackets indicates individual compounds containing one of each of the substituents.

[00062] In an exemplary embodiment of the compound of Formula (6), Ri is heteroaryl.

[00063] In an exemplary embodiment of the compound of Formula (6), Ri is aryl.

[00064] An aspect of the invention is a pharmaceutical composition comprising any compound of the invention as described herein (such as any one of Formula (1), (2), (3), (4), (5) or (6)) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound thereof, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier. [00065] In an exemplary embodiment, the pharmaceutical composition comprising any compound of the invention as described herein or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound thereof, or a pharmaceutically acceptable salt thereof further comprises one or more anti-cancer agents.

[00066] Another aspect of the invention is a method of treating a disease in which PI3K activity is implicated in a subject in need of such treatment, the method comprising administering to the subject a therapeutically effective amount of any compound of the invention as described herein (such as any one of Formula (1), (2), (3), (4), (5) or (6)) or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound thereof, or a pharmaceutically acceptable salt thereof.

[00067] In an exemplary embodiment, the disease to be treated is cancer. In a particular embodiment, the disease is a cancer bearing a PI3Ka H1047 mutation (such as H1047R).

DETAILED DESCRIPTION OF THE INVENTION

[00068] The term "at risk for" as used herein, refers to a medical condition or set of medical conditions exhibited by a patient which may predispose the patient to a particular disease or affliction. For example, these conditions may result from influences that include, but are not limited to, behavioral, emotional, chemical, biochemical, or environmental influences.

[00069] The term "effective amount" as used herein, refers to a particular amount of a pharmaceutical composition comprising a therapeutic agent that achieves a clinically beneficial result (/.e., for example, a reduction of symptoms). Toxicity and therapeutic efficacy of such compositions can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, which can be expressed as the ratio LD50/ ED50- Compounds that exhibit large therapeutic indices are preferred. The data obtained from these cell culture assays and additional animal studies can be used in formulating a range of dosages for human use. The dosages of such compounds lie preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, the sensitivity of the patient, and the route of administration. [00070] The term "symptom" as used herein, refers to any subjective or objective evidence of disease or physical disturbance observed by the patient. For example, subjective evidence is usually based upon patient self-reporting and may include, but is not limited to, pain, headache, visual disturbances, nausea and/or vomiting. Alternatively, objective evidence is usually a result of medical testing including, but not limited to, body temperature, complete blood count, lipid panels, thyroid panels, blood pressure, heart rate, electrocardiogram, tissue body imaging scans and other medical testing results.

[00071] The term "disease" as used herein, refers to any impairment of the normal state of the living animal or one of its parts that interrupts or modifies the performance of the vital functions. Typically manifested by distinguishing signs and symptoms, a disease is usually a response to i) environmental factors (such as malnutrition, industrial hazards, or climate); ii) specific infective agents (such as worms, bacteria, or viruses); iii) inherent defects of the organism (such as genetic anomalies); and/or iv) combinations of these factors.

[00072] The terms "reduce", "inhibit", "diminish", "suppress", "decrease", "prevent" and grammatical equivalents thereof (including "lower", "smaller", etc.) when used in reference to the expression of any symptom in an untreated subject relative to a treated subject, indicate that the quantity and/or magnitude of the symptoms in the treated subject is lower than in the untreated subject by any amount that is recognized as clinically relevant by any medically trained personnel. In one embodiment, the quantity and/or magnitude of the symptoms in the treated subject is at least 10% lower than, at least 25% lower than, at least 50% lower than, at least 75% lower than, and/or at least 90% lower than the quantity and/or magnitude of the symptoms in the untreated subject.

[00073] The term "inhibitory compound" as used herein, refers to any compound capable of interacting with (/.e., for example, attaching, binding, etc.) to a binding partner under conditions such that the binding partner becomes unresponsive to its natural ligands. Inhibitory compounds may include, but are not limited to, small organic molecules, antibodies, and proteins/peptides.

[00074] The term "attached" as used herein, refers to any interaction between a medium (or carrier) and a drug. Attachment may be reversible or irreversible. Such attachment includes, but is not limited to, covalent bonding, ionic bonding, Van der Waals forces or friction, and the like. A drug is attached to a medium (or carrier) if it is impregnated, incorporated, coated, in suspension with, in solution with, mixed with, etc. [00075] The term "drug" or "compound" as used herein, refers to any pharmacologically active substance capable of being administered which achieves a desired effect. Drugs or compounds can be synthetic or naturally occurring, non-peptide, proteins or peptides, oligonucleotides or nucleotides, polysaccharides, or sugars.

[00076] The term "administered" or "administering" as used herein, refers to any method of providing a composition to a patient such that the composition has its intended effect on the patient. An exemplary method of administering is by a direct mechanism such as, local tissue administration (/.e., for example, extravascular administration, such as subcutaneous, intramuscular, or intraperitoneal), intravenous, oral ingestion, transdermal patch, topical, inhalation, suppository, etc.

[00077] The term "patient" as used herein, is a human or animal and needs not be hospitalized. For example, out-patients and persons in nursing homes are "patients." A patient may be a human or non-human animal of any age and therefore includes both adults and juveniles (/.e., children). It is not intended that the term "patient" connote a need for medical treatment. Therefore, a patient may voluntarily be subject to experimentation, whether clinical or in support of basic science studies.

[00078] The term "subject" as used herein, refers to, but is not limited to, humans (e.g., a male or female of any age group, e.g., a pediatric subject (e.g., infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult or senior adult)) and/or other primates (e.g., monkeys); non-human mammals, such as cows, pigs, horses, sheep, mice, goats, cats, dogs; and/or birds, such as chickens, ducks and/or geese.

[00079] The term "affinity" as used herein, refers to any attractive force between substances or particles that causes them to enter into and remain in chemical combination. For example, an inhibitor compound that has a high affinity for a receptor will provide greater efficacy in preventing the receptor from interacting with its natural ligands, than an inhibitor with a low affinity.

[00080] The term "derived from" as used herein, refers to the source of a compound or sequence. In one respect, a compound or sequence may be derived from an organism or particular species. In another respect, a compound or sequence may be derived from a larger complex or sequence. [00081] The term "test compound" as used herein, refers to any compound or molecule considered a candidate as an inhibitory compound.

[00082] The term "combination therapy" as used herein refers to refers to a dosing regimen of two or more different therapeutically active agents during a period of time, wherein the therapeutically active agents are administered together or separately. In one embodiment the combination therapy is a non-fixed combination.

[00083] The term "non-fixed combination" as used herein refers to two or more different therapeutic agents that are formulated as separate compositions or dosages such that they may be administered separately to a subject in need thereof either simultaneously or sequentially with variable intervening time limits.

[00084] The term "synergy" or "synergistic" as used herein refers to the phenomenon where the combination of two therapeutic agents of a combination therapy is greater in terms of measured results than the sum of the effect of each agent when administered alone.

[00085] The term "in vivo" as used herein refers to an event that takes place in a subject's body.

[00086] The term "in vitro" as used herein refers to an event that takes places outside of a subject's body.

[00087] The term "protein" as used herein, refers to any of numerous naturally occurring extremely complex substances (such as an enzyme or antibody) that contain amino acid residues joined by peptide bonds, and which include carbon, hydrogen, nitrogen, oxygen, and typically sulfur. In general, a protein comprises amino acids having an order of magnitude within the hundreds.

[00088] The term "peptide" as used herein, refers to any of various amides that are derived from two or more amino acids by combination of the amino group of one acid with the carboxyl group of another and are usually obtained by partial hydrolysis of proteins. In general, a peptide comprises amino acids having an order of magnitude with the tens.

[00089] The term "pharmaceutically acceptable" or "pharmacologically acceptable" as used herein, refers to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human.

[00090] The term, "pharmaceutically acceptable carrier" as used herein, includes any and all solvents, or a dispersion medium including, but not limited to, water, ethanol, a polyol (such as, for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, vegetable oils, coatings, isotonic and absorption delaying agents, liposome, commercially available cleansers, and the like. Supplementary bioactive ingredients also can be incorporated into such carriers.

[00091] The term "pharmaceutically acceptable salt" as used herein, refers to a salt that does not adversely impact the biological activity and properties of the compound and is suitable for use in contact with the tissues of subjects without undue toxicity, irritation and/or allergic response and the like. Pharmaceutically acceptable salts include those derived from suitable inorganic acids, organic acids and bases, and include hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, nitric acid, acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, malonic acid, ascorbic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, benzoic acid, naphthalene sulfonic acid, lactic acid, succinic acid, oxalic acid, stearic acid, and the like. In some instances, pharmaceutically acceptable salts are obtained by reacting a compound having acidic group described herein with a base to form a salt such as an ammonium salt, an alkali metal salt (e.g., a sodium or a potassium salt), an alkaline earth metal salt (e.g., a calcium or a magnesium salt), a salt formed from an organic base, and an amino acid salt. Pharmaceutically acceptable salts derived from appropriate bases include alkali metals, alkaline earth metals, and ammonium and quaternary ammonium compounds. Specific metals include, but are not limited to, sodium, lithium, potassium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.

Organic bases from which salts may be prepared include, for example, primary, secondary, and tertiary amines.

[00092] The term "prodrug" as used herein, refers to a compound that is transformed in vivo to yield a disclosed compound or a pharmaceutically acceptable form of the compound. A prodrug may be inactive when administered to a subject, but is converted in vivo to an active compound. In various instances, a prodrug has improved physicochemical properties (such as bioavailability) and/or delivery properties over the parent compound. Prodrugs are typically designed to enhance pharmaceutically and/or pharmacokinetically based properties associated with the parent compound. The prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in subject. Prodrugs include compounds wherein a hydroxy, amino, or mercapto group is bonded to any group that, when the prodrug is administered to a subject, cleaves to form a free hydroxy, free amino, or free mercapto group, respectively. Prodrugs are well known to be prepared from carboxylic acids in the form of, for example, carboxylate esters or thioesters.

[00093] The term, "purified" or "isolated" as used herein, may refer to a composition (such as, for example, a peptide composition) that has been subjected to treatment (e.g., fractionation) to remove various other components, and which composition substantially retains its expressed biological activity.

[00094] The term "sample" as used herein, includes, for example, environmental and biological samples. Environmental samples include material from the environment such as soil and water. Biological samples include animal (e.g., human), fluids (e.g., blood, plasma, and serum), solids (e.g., stool), tissue, liquid foods (e.g., milk), and solid foods (e.g., vegetables). For example, a pulmonary sample may be collected by bronchoalveolar lavage (BAL) which comprises fluid and cells derived from lung tissues. A biological sample may comprise a cell, tissue extract, body fluid, chromosomes or extrachromosomal elements isolated from a cell, genomic DNA (in solution or bound to a solid support such as for Southern blot analysis), RNA (in solution or bound to a solid support such as for Northern blot analysis), cDNA (in solution or bound to a solid support) and the like.

[00095] The term "biologically active" as used herein, refers to any molecule having structural, regulatory or biochemical functions. For example, biological activity may be determined, for example, by restoration of wild-type growth in cells lacking protein activity. Cells lacking protein activity may be produced by many methods (i.e., for example, point mutation and frame-shift mutation). Complementation is achieved by transfecting cells which lack protein activity with an expression vector which expresses the protein, a derivative thereof, or a portion thereof.

[00096] The term "label" or "detectable label" as used herein, refers to any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Such labels include biotin for staining with labeled streptavidin conjugate, magnetic beads (e.g., Dynabeads’), fluorescent dyes (e.g., fluorescein, Texas Red’, rhodamine, green fluorescent protein, and the like), radiolabels (e.g., 3 H, 125 l, 35 S, 14 C, or 32 P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and calorimetric labels such as colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads. Patents teaching the use of such labels include, but are not limited to, U.S. Patent Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241 (all herein incorporated by reference in their entireties). The labels contemplated in the present invention may be detected by conventional methods. For example, radiolabels may be detected using photographic film or scintillation counters, fluorescent markers may be detected using a photodetector to detect emitted light. Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting, the reaction product produced by the action of the enzyme on the substrate, and calorimetric labels are detected by simply visualizing the colored label.

[00097] The term "conjugate" as used herein, refers to any compound that has been formed by the joining of two or more moieties.

[00098] A "moiety" or "group" as used herein, is any type of molecular arrangement designated by formula, chemical name, or structure. Within the context of certain embodiments, a conjugate comprises one or more moieties or chemical groups. This means that the formula of the moiety is substituted at some position in order to be joined and be a part of the molecular arrangement of the conjugate. Although moieties may be directly covalently joined, it is not intended that the joining of two or more moieties must be directly to each other. A linking group, a crosslinking group, or a joining group refers to any molecular arrangement that will connect moieties by covalent bonds such as, but not limited to, one or more amide group(s). Additionally, although the conjugate may be unsubstituted, the conjugate may have a variety of additional substituents connected to the linking groups and/or connected to the moieties.

[00099] A "polymer" or "polymer group" as used herein, refers to a chemical species or group composed of repeatedly linked moieties. Within certain embodiments, it is preferred that the number of repeating moieties is 3 or more or greater than 10. The linked moieties may be identical in structure or may vary in their moiety structures. A "monomeric polymer" or "homopolymer" is a polymer that contains the same repeating, asymmetric subunit. A "copolymer" is a polymer derived from two or more types of monomeric species (/.e., two or more different chemical asymmetric subunits). "Block copolymers" are polymers comprised of two or more species of polymer subunits linked by covalent bonds.

[000100] The term "substituted" as used herein, refers to at least one hydrogen atom of a molecular arrangement that is replaced with a non-hydrogen substituent. The number of substituents present depends on the number of hydrogen atoms available for replacement and includes replacement of more than one hydrogen atom bound to a single atom (such as in the case of a carbon atom or a silicon atom which may be available for mono-, di- or tri- substitution or in the case of a nitrogen atom which may be available for mono-, di- or trisubstitution or in the case of an oxygen atom or a sulfur atom which may be available for mono-substitution). In the case of an oxo substituent ("=O"), two hydrogen atoms are replaced (which provides, for example, -(CH 2 )-C(=O)-CH3 as a substituent when the two hydrogen atoms of the middle carbon atom of -CH 2 -CH 2 -CH3 are replaced). When substituted, one or more of the groups below are "substituents." Substituents include, but are not limited to, halogen (e.g., F, Cl, Br, I), hydroxy (OH), oxo, cyano (CN), nitro (NO2), amino, alkylamino, dialkylamino, branched or unbranched alkyl (e.g., methyl, ethyl, propyl, isopropyl, sec-butyl, etc.), cycloalkyl (e.g., cyclopropyl), fluoroalkyl (e.g., CF 3 , CF 2 H, CH 2 F, CH 2 CF 3 , CH 2 CF2H, CHFCHF2, CF2CH 2 F, CF2CF 3 , CF2CH3, CF(CH 3 ) 2 , CH 2 CH 2 CF 3 , CF2CH 2 CF 3 , CF2CF2CF 3 , etc.) or more generally, haloalkyl (e.g., CH 2 CI, CH(CH 3 )Br, etc.), O-alkyl (alkoxy) (e.g., OCH 3 , OCH 2 CH3, OCH(CH 3 )2, etc.), O- cycloalkyl (e.g., O-cyclopropyl), O-haloalkyl (e.g., OCF 2 H, OCH 2 F, OCF 3 , OCH 2 CF 3 , OCH 2 CF2H, OCHFCHF2, OCF2CH 2 F, OCF2CF 3 , OCF2CH3, OCF(CH 3 ) 2 , OCH 2 CH 2 CF 3 , OCF2CH 2 CF 3 , OCF2CF2CF 3 or OCH 2 CI), O-aryl (e.g., O-phenyl), O-heteroaryl, O-heterocyclyl, thioalkyl (e.g., S-CH 3 ), hydroxyalkyl (e.g., CH 2 OH), alkyl ether (e.g., CH 2 OCH3), alkynyl (e.g., -CsCRf), alkenyl (e.g., -CRf=CRfR g ), aryl (e.g., phenyl), arylalkyl (e.g., CH 2 Ph), heteroaryl (e.g., pyridyl or any 5- or 6- membered heteroaryl ring), heteroarylalkyl (e.g., CH 2 -pyridine), heterocyclyl, heterocycloalkyl and as well as, -NR f R g , -NR f C(=O)R g , -NR f C(=O)NR f NR g , -NR f -C(=O)ORfSO 2 R g , -C(=O)R f , -C(=O)OR f , -OR f , -C(=O)NR f R g , -OC(=O)NRfR g , -SR f , -SOR f , -S(=O) 2 R f , -OS(=O) 2 R f and -S(=O)OR f , where each R f and R g may be the same or different and are independently, hydrogen, alkyl (e.g., CH3), substituted alkyl, haloalkyl, aryl, substituted aryl, arylalkyl, substituted arylalkyl, heterocyclyl, substituted heterocyclyl, heterocycloalkyl, substituted heterocycloalkyl, heteroaryl or substituted heteroaryl. In addition, the above substituents may be further substituted with one or more substituents as defined above, such that a substituent may constitute, for example, a substituted alkyl, a substituted aryl, a substituted arylalkyl, a substituted heterocyclyl, or a substituted heterocycloalkyl.

[000101] The term "unsubstituted" as used herein, refers to any compound that does not contain extra substituents attached to the compound. For example, an unsubstituted compound refers to the chemical makeup of the compound without added substituents (e.g., no non-hydrogen substituents). For example, unsubstituted proline is a proline amino acid even though the amino group of proline may be considered as disubstituted with alkyl groups. [000102] The term "bond" as used herein in describing a substituent with atoms on both sides, refers to the absence of that substituent. For example, in the 4-atom sequence A-B-C-D, when B and C are both listed as being bonds, the result is the 2-atom sequence A-D. If only B is listed as being a bond, the result is the 3-atom sequence A-C-D.

[000103] The term "alkyl" as used herein, refers to any straight chain or branched, non-cyclic or cyclic, unsaturated or saturated aliphatic hydrocarbon containing from 1 to 10 carbon atoms, while the term "lower alkyl" has the same meaning as alkyl but contains from 1 to 3 carbon atoms. The term "higher alkyl" has the same meaning as alkyl but contains from 4 to 10 carbon atoms. Representative saturated straight chain alkyls include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, n-nonyl, and the like, while saturated branched alkyls include, but are not limited to, isopropyl, sec-butyl, isobutyl, tertbutyl, isopentyl, and the like. As used herein, a methyl substituent may be depicted as "CH3" or "Me" or as a terminal bond with no indication of specific atoms.

[000104] The term "cycloalkyl" as used herein, refers to saturated and unsaturated cyclic alkyls. Representative saturated cyclic alkyls include, but are not limited to, C 3 -C 14 (such as C 3 - C7) cycloalkyls, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclododecyl, and the like; while unsaturated cyclic alkyls include, but are not limited to, cyclobutenyl, cyclopentenyl and cyclohexenyl, cyclohexadiene, and the like. Cyclic alkyls are also referred to herein as "homocycles" or "homocyclic rings".

[000105] The term "bicyclic compounds" as used herein, encompasses "bridged" compounds, "fused" compounds and "spiro" compounds as described.

[000106] The term "spiro" or "spirocyclic" as used herein, refers to chemical structures having at least two rings sharing one common atom. The rings may be cycloalkyl, heterocyclyl or a combination thereof, and may include one or more aryl or heteroaryl rings. Examples include, but are not limited to, spirocyclic cyclopropanes, spirocyclic aziridines, spirocyclic cyclobutanes, spirocyclic azetidines, spirocyclic oxetanes, spirocyclic cyclopentanes, spirocyclic pyrrolidines, spirocyclic 1,3-dioxolanes, spirocyclic dioxanes, spirocyclic oxathiolanes, spirocyclic thiazolidines, spirocyclic cyclohexanes, spirocyclic piperidines and spirocyclic piperizines, where the other ring is cycloalkyl (e.g., cyclobutane, cyclopentane or cyclohexane) or heterocyclyl (e.g., piperidine, tetrahydropyran, tetrahydrofuran, azetidine or pyrrolidine). Exemplary embodiments include, but are not limited to, l,4-dioxaspiro[4.5]decane, 1,4-dioxa- 8-azaspiro[4.5]decane, 2-azaspiro[4.4]nonane, 2-azaspiro[4.4]nonane, 2,7- diazaspiro[4.4]nonane, 3-azaspiro[5.5]undecane, 3,9-diazaspiro[5.5]undecane, 6- azaspiro[3.4]octane, 6-azaspiro[2.5]octane, l,3-dihydrospiro[indene-2,3'-pyrrolidine] and 3,4- dihydro-2H-spiro[naphthalene-l,4'-piperidine],

[000107] The term "bridged" as used herein, refers to a compound containing two nonadjacent atoms common to two rings. Exemplary embodiments include, but are not limited to, norbornane, bicyclo[l.l.l]pentane, bicyclo[2.2.1]heptane, l,4-diazabicyclo[2.2.2]octane, 3,8-diazabicyclo[3.2.1]octane, 3-azabicyclo[3.2.1]octane, bicyclo[3.2.1]octane, 3,6- diazabicyclo[3.1.1]heptane, 3,6-diazabicyclo[2.2.1]heptane, and other bridged piperazines and bridged piperidines.

[000108] The term "fused" as used herein, refers to polycyclic ring systems in which any two adjacent rings have two, and only two, adjacent atoms in common (ortho-fused) and polycyclic ring systems in which a ring contains two, and only two, adjacent atoms in common with each of two or more rings of a contiguous series of ortho-fused rings (ortho- and peri-fused). An exemplary embodiment is pentalene and dibenzoxepine (ortho-fused) and pyrene (ortho- and peri-fused). Ortho-fused systems have "n" common sides and "2n" common atoms while perifused systems have "n" common sides and less than "2n" atoms in common. Other exemplary fused systems include, but are not limited to, fused cyclopropyl rings, fused aziridine rings, fused cyclobutane rings, fused azetidine rings, fused cyclopentane rings, fused pyrrolidine rings, fused cyclohexane rings, fused piperidine rings, fused tetrahydropyran rings and fused piperazine rings, where each of these rings may be fused to an identical or different ring, such a pyrrolidine ring fused to another pyrrolidine ring (e.g., octahydropyrrolo[3,4-c]pyrrole) or to a cyclohexane ring (e.g., octahydro-lH-indole or octahydro-lH-isoindole). Other examples include fused aryl or heteroaryl rings, such as a pyridine ring fused with a cycloalkyl ring (e.g., cyclopentane or cyclohexane) or with a heterocyclyl ring (e.g., tetrahydrofuran or tetrahydropyran).

[000109] The term "aromatic" or "aryl" as used herein, refers to any aromatic carbocyclic (i.e., all of the ring atoms are carbon) substituent such as, but not limited to, phenyl (from benzene), tolyl (from toluene), xylyl (from xylene) or multi-ring systems (e.g., naphthyl (from naphthalene) and anthracenyl (from anthracene).

[000110] The term "arylalkyl" or "aralkyl" as used herein, refers to any alkyl having at least one alkyl hydrogen atom replaced with an aryl moiety such as, but not limited to, benzyl, - (CH 2 ) 2 phenyl, -(CH 2 ) 3 phenyl, -CH(phenyl)2, and the like. [000111] The term "halogen" as used herein, refers to any fluoro, chloro, bromo, or iodo moiety.

[000112] The term "haloalkyl" as used herein, refers to any alkyl where at least one hydrogen atom (and including all hydrogen atoms) has been replaced with a halogen atom, such as, for example, trifluoromethyl, dichloromethyl, difluoromethyl, monofluoromethyl, monobromomethyl, 1,1,1-trifluoroethyl and the like.

[000113] The term "heteroaromatic" or "heteroaryl" as used herein, refers to any aromatic heterocyclic ring of 5 to 10 or more members and having at least one heteroatom selected from nitrogen, oxygen or sulfur, and containing at least 1 carbon atom, including, but not limited to, both mono- and bicyclic- ring systems. The heteroaryl ring may be attached as a substituent via a ring heteroatom or a carbon atom. Representative heteroaromatics include, but are not limited to, furan, benzofuran, thiophene, benzothiophene, pyrrole, indole, isoindole, 7-azaindole, 4-azaindole, 5-azaindole, 6-azaindole, 7-azaindazole, pyridine, quinoline, isoquinoline, oxazole, isoxazole, benzoxazole, pyrazole, imidazole, benzimidazole, thiazole, benzothiazole, isothiazole, 1,2,4-triazole, 1,2,3-triazole, tetrazole, 1,2,5-oxadiazole, 1,2,3-oxadiazole, 1,3,4-thiadiazole, pyridazine, pyrimidine, pyrazine, 1,2,4-triazine, 1,3,5- triazine, cinnoline, phthalazine, quinazoline, 1,8-naphthylpyridine, pyrido[3,2-d]pyrimidine, pyrido[4,3-d]pyrimidine, pyrido[3,4-b]pyrazine, pyrido[2,3-b]pyrazine, pteridine, triazolo- pyridines and the like.

[000114] The term "heteroarylalkyl" as used herein, means any alkyl having at least one alkyl hydrogen atom replaced with a heteroaryl moiety, such as -CH 2 bpyridinyl, -CH 2 hpyrim idinyl, and the like.

[000115] The term "heterocycle" or "heterocyclyl" or "heterocyclic ring" as used herein, refers to a nonaromatic ring which is either saturated or unsaturated and which contains 1 or more heteroatoms independently selected from nitrogen, oxygen, sulfur and silicon, wherein each of the nitrogen and sulfur heteroatoms may be in an oxidized state, and each of the nitrogen and silicon heteroatoms is substituted or unsubstituted and the nitrogen heteroatoms may be optionally quaternized, and includes bicyclic rings in which any of the above heterocycles are fused to an aryl or heteroaryl ring. The heterocyclic ring may be attached as a substituent via a ring heteroatom or a carbon atom. In various embodiments, heterocycles may contain 3 to 14 or more ring atoms (such as 3- to 7-membered monocyclic rings or 7- to 10- membered bicyclic rings) and include, but are not limited to, 2H-azirine, azetidine, 2,3- dihydroazete, 1,3-diazetidine, 2H-oxete, thietane, 2H-thiete, azetidin-2-one, morpholine, thiomorpholine, pyrrolidinone, pyrrolidinine, 2-pyrroline, 3-pyrroline, pyrazolidine, 2- pyrazoline, 2-imidazoline, imidazolidine, piperidine, piperazine, pyridin-2-ones (such as 2- pyridone and l-methyl-2-pyridone), ethylene oxide (oxirane), ethylene imine (aziridine), ethylene sulfide (thiirane), oxetane, propylene oxide, 1,3-dioxolane, 1,2-oxathiolane, 1,3- oxathiolane, sulfolane, 2,4-thiazolidinedione, succinimide, 2-oxazolidone, dioxane, hydantoin, valerolactam, tetrahydrofuran, tetrahydropyran, 2H-pyran, 4H-pyran, thiane, 2H-thiopyran, 1,3-dithiane, 1,4-dithiane, 1,3,5-trithiane, pyrrolizidine, 1, 4,5,6- tetrahydrocyclopenta[b]pyrrole, tetrahydropyridine, tetrahydropyrimidine, tetrahydrothiophene, tetrahydrothiopyran, indoline, isoindoline, decahydroisoquinoline, decahydroquinoline, 1,2,3,4-tetrahydroquinoline, 1,2-dihydroquinoline, 2H- benzo[e][l,3]oxazine, 2H-benzo[b][l,4]oxazine, quinolin-2(lH)-one, isoquinolin-l(2H)-one, quinuclidine, 1-azaadamantane, 2-azaadamantane, 2,3-dihydroazepine, 2,5-dihydroazepine, oxepane, azonane, spiro[cyclobutane-l,3'-indole], l-oxaspiro[4,5]decane, 1,6- dioxaspiro[3,4]octane, 2-oxa-7-azaspiro[3,5]nonane, l,4-dioxa-7-azaspiro[4,4]nonane, 1,3- diazaspiro[4,4]non-2-en-4-one, 2,9-diazaspiro[5,5]undecan-l-one, 8-azaspiro[4,5]decane-7,9- dione, l,4-dithia-7-azaspiro[4,4]nonane, and the like.

[000116] The term "heterocycloalkyl" as used herein, refers to any alkyl having at least one alkyl hydrogen atom replaced with a heterocycle, such as -Cl- morpholinyl, and the like.

[000117] The term "alkylamino" as used herein, means at least one alkyl moiety attached through a nitrogen bridge (/.e., -N-(alkyl) n ), where n = 1 or 2, such as alkylamino or dialkylamino) including, but not limited to, methylamino, ethylamino, dimethylamino, diethylamino, and the like.

[000118] The term "alkyloxy" or "alkoxy", as used herein, means any alkyl moiety attached through an oxygen bridge (/.e., -O-alkyl) such as, but not limited to, methoxy, ethoxy, and the like.

[000119] The term "thioalkyl" as used herein, means any alkyl moiety attached through a sulfur bridge (/.e., -S-alkyl) such as, but not limited to, methylthio, ethylthio, and the like.

[000120] The term "alkenyl" as used herein, refers to an unbranched or branched hydrocarbon chain having one or more carbon-carbon double bonds therein and may also be referred to as an "unsaturated alkyl". The double bond of an alkenyl group can be unconjugated or conjugated to another unsaturated group. Suitable alkenyl groups include, but are not limited to vinyl, allyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, 2-ethylhexenyl, 2-propyl-2-butenyl, 4-(2-methyl-3-butene)-pentenyl. An alkenyl group can be unsubstituted or substituted with one or two suitable substituents.

[000121] The term "alkynyl" as used herein, refers to unbranched or branched hydrocarbon chain having one or more carbon-carbon triple bonds therein and may also be referred to as an "unsaturated alkyl". The triple bond of an alkynyl group can be unconjugated or conjugated to another unsaturated group. Suitable alkynyl groups include, but are not limited to ethynyl, propynyl, butynyl, pentynyl, hexynyl, methylpropynyl, 4-methyl-l-butynyl, 4-propyl-2- pentynyl-, and 4-butyl-2-hexynyl. An alkynyl group can be unsubstituted or substituted with one or two suitable substituents.

[000122] As used herein, "reactive groups" refer to nucleophiles, electrophiles, or radically active groups, i.e., groups that react in the presence of radicals. A nucleophile is a moiety that forms a chemical bond to its reaction partner (the electrophile) by donating both bonding electrons. Electrophiles accept these electrons. Nucleophiles may take part in nucleophilic substitution, whereby a nucleophile becomes attracted to a full or partial positive charge on an element and displaces the group it is bonded to. Alternatively, nucleophiles may take part in substitution of carbonyl group. Carboxylic acids are often made electrophilic by creating succinyl esters and reacting these esters with aminoalkyls to form amides. Other common nucleophilic groups are thiolalkyls, hydroxylalkyls, primary and secondary amines, and carbon nucleophiles such as enols and alkyl metal complexes. Other preferred methods of ligating proteins, oligosaccharides and cells using reactive groups are disclosed (Lemieux et al., Trends in Biotechnology 1998, 16, 506, incorporated herein by reference in its entirety). In yet another preferred method, one provides reactive groups for the Staudinger ligation, i.e., "click chemistry" with an azide comprising moiety and alkynyl reactive groups to form triazoles. Michael additions of a carbon nucleophile enolate with an electrophilic carbonyl, or the Schiff base formation of a nucleophilic primary or secondary amine with an aldehyde or ketone may also be utilized. Other methods of bioconjugation are provided (Hang et al. Accounts of Chemical Research 2001, 34, 7 27 and Kiick et al. Proc Natl Acad Sci US. A. 2002, 99, 19, both of which are incorporated by reference in its entirety).

[000123] The term "biocompatible" as used herein, refers to any material that does not illicit a substantial detrimental response in the host. There is always concern when a foreign object is introduced into a living body that the object will induce an immune reaction, such as an inflammatory response that will have negative effects on the host. In the context of this invention, biocompatibility is evaluated according to the application for which it was designed: for example, a bandage is regarded as biocompatible with the skin, whereas an implanted medical device is regarded as biocompatible with the internal tissues of the body. Preferably, biocompatible materials include, but are not limited to, biodegradable and biostable materials. A substantial detrimental response has not occurred if an implant comprising the material is in close association to its implant site within the host animal and the response is better than a tissue response recognized and established as suitable from materials provided in an ASTM. ASTM subcommittee F04.16 on Biocompatibility Test Methods has developed biocompatibility standards for medical and surgical materials and devices which includes E1262-88, F612-20, F719-20el, F720-17, F748-16, F749-20, F750-20, F756-17; F763-04, F813-20, F895-11, F981-04, F1027-86, F1408-20a, F1439-03, F1877-16, F1903-18, F1904-14, F1983-14, F1984-99, F2147- 01, F2148-18, F2382-18, F2808-17, F1288-19 and F2909-19, each of which is incorporated herein by reference. For example, materials that are to be used in contact with the blood stream must be composed of materials that meet hemocompatibility standards. One of these tests is for damage to red blood cells, which can result in hemolysis that is, rupturing of the cells, as described in F756-17 Standard Practice for Assessment of Hemolytic Properties of Materials.

[000124] As used herein, a "bioactive substance" refers to any of a variety of chemical moieties and that binds with a biomolecule such as, but not limited to, peptides, proteins, enzymes, receptors, substrates, lipids, antibodies, antigens, and nucleic acids. In certain preferred embodiments, the bioactive substance is a biomolecule but it is not intended that the bioactive substance be limited to biomolecules. In other preferred embodiments, the bioactive substances provide hydrophobic, hydrophilic, or electrostatic interactions, such as polycarboxylic acids that are anionic at physiological pH. In other preferred embodiment, the alkaline growth factors (with isoelectric point above 7) are retained via favorable electrostatic interactions by the polycarboxylates, and subsequently released in a controlled and sustained manner.

[000125] "Cancer" is a term used for a physiological condition in mammals that is typically characterized by unregulated cell growth. Examples of cancer include, but are not limited to, carcinoma, lymphoma, leukemia, blastoma, and sarcoma. More particular examples of such cancers include squamous cell carcinoma, small cell lung cancer, non-small cell lung cancer (NSCLC), glioma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, acute myeloid leukemia (AML), multiple myeloma, gastrointestinal cancer, renal cell carcinoma, renal cancer (e.g., advanced renal cell carcinoma), ovarian cancer, liver cancer, lymphoblastic leukemia, lymphocytic leukemia, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, melanoma, chondrosarcoma, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, brain cancer, stomach cancer, urothelial carcinoma (including local advanced or metastatic urothelial carcinoma), bladder cancer, hepatoma, breast cancer and head and neck cancer.

[000126] The term "stereoisomer" refers to compounds that have the same atomic connectivity but different atomic arrangement in space. Stereoisomers include cis-trans isomers, E and Z isomers, enantiomers, diastereomers and atropisomers. In the context of the present invention, the term "enantiomerically pure" is understood to mean that the compound in question with respect to the absolute configuration of the chiral center is present in an enantiomeric excess of more than 95%, preferably more than 97%.

[000127] The present disclosure contemplates all such compounds, including cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereomers isomers, (D)- isomers, (L)-isomers, atropisomers, tautomers and racemic and other mixtures thereof, such as enantiomers or diastereomeric enriched mixtures, all of which are within the scope of the present disclosure. Insofar as compounds of the invention as defined herein may exist in optically active or racemic forms by virtue of one or more asymmetric carbon atoms, the invention includes in its definition any such optically active or racemic form. The synthesis of optically active compounds may be carried out by standard techniques of organic chemistry well known in the art such as, for example, by synthesis from optically active starting materials or by resolution of a racemic compound. Similarly, the enantiomeric or diastereomeric purity of a compound may be evaluated using standard laboratory techniques.

[000128] The pharmaceutical compositions of the invention can take any suitable form for the desired route of administration. Where the composition is to be administered orally, any suitable orally deliverable dosage form can be used, including without limitation water, glycols, oils, alcohols, and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs, emulsions, and solutions; or solid carriers such as starches, sugars, kaolin, diluents, lubricants, binders, disintegrating agents, and the like in the case of powders, pills, capsules, and tablets. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit forms. Injectable compositions or intravenous infusions are also provided in the form of solutions, suspensions, and emulsions. For parenteral compositions, the carrier usually comprises sterile water and possibly other ingredients to aid solubility. Injectable solutions may be prepared in which the carrier comprises a saline solution, a glucose solution, or a mixture of a saline and a glucose solution. Suitable oils include, for example, peanut oil, sesame oil, cottonseed oil, corn oil, soybean oil, synthetic glycerol esters of long chain fatty acids, and mixtures of these and other oils. In compositions suitable for percutaneous administration, the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives as needed, where the additives may facilitate administration of the composition to the skin and/or may facilitate preparation of the compositions to be delivered. These compositions may be administered in various ways, e.g., as a transdermal patch or as an ointment. Acid or base addition salts of the compounds of the invention are typically more suitable in the preparation of aqueous compositions due to their increased water solubility over the corresponding neutral form of the compounds.

[000129] The pharmaceutical compositions of the invention may comprise one or more of a filler, diluent, adjuvant, vehicle, or other excipient to facilitate storage and/or administration of the active ingredients contained therein.

[000130] In an exemplary embodiment, a pharmaceutical composition according to the present invention may contain one or more additional therapeutic agents, for example, to increase efficacy or to decrease undesired side effects. In a particular embodiment, the pharmaceutical composition further contains one or more additional therapeutic agents useful to treat or inhibit a disease mediated directly or indirectly by PI3K. Examples of such agents include, without limitation, agents to treat or inhibit cancer, Huntington's disease, cystic fibrosis, liver fibrosis, renal fibrosis, pulmonary fibrosis, skin fibrosis, rheumatoid arthritis, diabetes, or heart failure.

[000131] In a specific embodiment, the additional therapeutic agent to be included is an anti-cancer agent. Examples of an anti-cancer agent include, but are not limited to, DNA- damaging cytotoxic drugs, alkylating agents such as cyclophosphamide, dacarbazine, and cisplatin; anti-metabolites such as methotrexate, mercaptopurine, thioguanine, fluorouracil, and cytarabine; plant alkaloids such as vinblastine and paclitaxel; antitumor antibiotics such as doxorubicin, bleomycin and mitomycin; hormones/antihormones such as prednisone, tamoxifen, and flutamide; other types of anticancer agents such as asparaginase, rituximab, trastuzumab, imatinib, retinoic acid, and derivatives, colony stimulating factors, amifostine, camptothecin, topotecan, thalidomide analogs such as lenalidomide, and proteasome inhibitors such as Velcade.

[000132] In another embodiment, the present invention provides a method of inhibiting or treating diseases arising from abnormal cell proliferation and/or differentiation in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of one or more compounds according to the present invention. In one embodiment, the method of inhibiting or treating disease comprises administering to a subject in need thereof, a composition comprising an effective amount of one or more compounds of the invention and a pharmaceutically acceptable carrier. The composition to be administered may further contain a therapeutic agent such as an anti-cancer agent.

[000133] The compounds of the invention are defined herein by their chemical structures and/or chemical names and are generally listed according to the IUPAC or CAS nomenclature system. Abbreviations that are well known to one of ordinary skill in the art may be used. When a compound is referred to by both a chemical structure and a chemical name, and the chemical structure and chemical name conflict, the chemical structure is intended to be determinative of the compound's identity.

[000134] The present invention includes compounds labeled with various radioactive or nonradioactive isotopes. Examples of atomic isotopes may include, but are not limited to, deuterium ( 2 H), tritium ( 3 H), iodine-125 ( 125 l), carbon-14 ( 14 C), nitrogen-15 ( 15 N), sulfur-35 ( 35 S) and chlorine-36 ( 36 CI). In an exemplary embodiment, one or more hydrogen atoms in a compound of the invention can be replaced by deuterium. In various embodiments, a compound of the invention includes at least one deuterium atom, or two or more deuterium atoms, or three or more deuterium atoms, etc. As described herein, compounds of the invention may also be radiolabeled with a radioactive isotope such as tritium ( 3 H), iodine-125 ( 125 l), and carbon-14 ( 14 C). A radiolabeled compound is useful as a therapeutic or prophylactic agent, provides a reagent for research such as for an assay, and/or provides a diagnostic agent for techniques such as in vivo imaging. Synthetic methods for incorporating isotopes into organic compounds are well known in the art.

[000135] In an embodiment of the invention, a compound of the invention as defined herein (such as a compound of any one of Formula (1), (2), (3), (4), (5) or (6)) or a pharmaceutically- acceptable salt thereof, exists as a single enantiomer being in an enantiomeric excess (% ee) of > 95%, such as > 98%, such as > 99%.

[000136] In an embodiment of the invention, a pharmaceutical composition comprises a compound of the invention as defined herein (such as a compound of any one of Formula (1),

(2), (3), (4), (5) or (6)) or a pharmaceutically-acceptable salt thereof, where the compound exists as a single enantiomer being in an enantiomeric excess (% ee) of > 95%, such as > 98%, such as > 99%.

[000137] In an exemplary embodiment of the invention, the disease or disorder to be treated by the compounds of the invention is selected from congenital lipomatous overgrowth, vascular malformations, epidermal naevi, scoliosis/skeletal and spinal syndrome (CLOVES), mosaic tissue overgrowth syndromes, venous malformations and brain malformations associated with severe epilepsy or PIK3CA-related overgrowth syndrome (PROS) (Keppler- Noreuil et al., Am J Med Genet A. 201S, 167A, 287; Kurek et al. Am. J. Hum. Genet. 2012, 90, 1108).

[000138] In an exemplary embodiment of the invention, the cancer to be treated is a cancer bearing a PI3K H1047 mutation (such as H1047R) (Thorpe et al., Nat Rev Cancer 2015, 15, 7).

[000139] The compounds of the invention (such as defined by any one of Formula (1), (2),

(3), (4), (5) or (6)) are typically PI3Ka H1047R mutant-selective inhibitors that exhibit greater selectivity for the H1047R mutation over the wild-type. As such, the compounds may decrease the amount of phosphorylated AKT (pAKT) and decrease proliferation selectively in PI3Ka H1047R mutant cell lines, preferably across several tumor types.

[000140] A PI3K H1047R mutant selective inhibitor of the invention (such as defined by a compound of any one of Formula (1), (2), (3), (4), (5) or (6)) dosed in combination with a selective estrogen receptor degrader (SERD) such as, but not limited to, fulvestrant, elacestrant, camizestrant or vepdegestrant may exhibit a combination benefit leading to tumor regression in ER+ / PI3K H1047R mutant tumors such as, but not limited to, the breast cancer xenograft model T47D, at doses where little or no regression would be observed with either single agent.

[000141] A PI3K H1047R mutant selective inhibitor of the invention (such as defined by a compound of any one of Formula (1), (2), (3), (4), (5) or (6)) dosed in combination with a HER2 inhibitor such as, but not limited to, tucatinib or trastuzumab may exhibit a combination benefit leading to tumor regression in ER- / HER2+ / PI3K H1047R mutant tumors such as, but not limited to, the breast cancer xenograft model HCC1954, at doses where little or no regression would be observed with either single agent.

[000142] Compounds of Formula (1) of the present invention were generally prepared according to the synthetic routes identified in Schemes 1 to 3:

Scheme 1

[000143] Beginning from aminobenzamides 1 that are generally known, intermediate bromoquinazolin-4-ones 4 can be generated by various cyclization methods. In an exemplary embodiment with R4-CO2H 2 (where the scope of R4 is defined in Formula (1)), cyclization of 1 may be effected by treatment with T 3 P and Hunig's base or pyridine, followed by K 2 CO 3 (Scheme 1). For inputs 2, where R 4 is a substituted [l,l,l]-bicyclopentane, a number of methods are known for their preparation (Org. Lett. 2020, 1648-1654; Organic Process Research & Development 2021, 642-647; Angew. Chemie Int. Ed. 2017, 12774-12777; Nature Chemistry 2022, 1068-1077). In an exemplary embodiment with R4-CHO 3 (where the scope of R4 is defined in Formula (1)), cyclization with 1 can be achieved by condensation mediated by MgSO 4 and PTSA followed by oxidation with DDQ. Acetylquinazolinones 5 may be created by carbonylations of 4. In some exemplary embodiments, the bromine substituent of 4 may be replaced with an acetyl group by treatment with tributyl(l-ethoxyvinyl)tin and a catalytic palladium species (e.g., but not limited to, Pd(PPh 3 )4 or PdCI 2 (PPh 3 ) 2 ) at elevated temperature, followed by hydrolysis with aqueous HCI, to yield ketones 5. Intermediates 5 when R 3 = H may also be generated by formylation of 4 through a variety of known methods (e.g., but not limited to, palladium-catalyzed carbonylation in the presence of H 2 (Klaus, et al., Angew. Chem. Int. Ed. 2006, 45, 154), or cyanation followed by DIBAL reduction). Alternatively, intermediates 5 can serve as a platform for further expansion into diverse R 3 substitutions (e.g., trifluoromethyl, difluoromethyl, fluoromethyl, alkyls, etc.) through a variety of known techniques (including, but not limited, to Prakash, et al., J. Am. Chem. Soc. 1989, 111, 393; Zhao, et al., Org. Lett. 2011, 13, 5342; Reichel, et al., Angew. Chem. Int. Ed. 2020, 59, 12268; etc.). Alcohol-substituted quinazolinones of general structure 6 may be prepared by the reduction of 5. In an exemplary embodiment, the reduction may be achieved by treatment of 5 with NaBFU in MeOH to yield 6. Alternatively, the bromine substituent of aminobenzamides 1 may be replaced with an acetyl group to give acetylbenzamides 7, of which the ketone may be reduced and protected (such as with protecting groups like tert-butyldimethylsilyl) to give intermediates 8. Cyclization in manners similar to that from 1 to 4 may be effected to provide quinazolinones 9 from aminobenzamides 8. Finally, deprotections of the secondary alcohol groups within intermediates 9 can of course yield compounds 6. It should be understood that apart from the methodologies described in Scheme 1, there are other reported methods that can be utilized to prepare quinazolin-4-ones such as 4 or 9 and their derivatives (for examples: Tian et al., Molecular Catalysis 2021, 111345; Fang et al., Organic & Biomolecular Chemistry 2012, 10(12), 2389; Li et al., J. Org. Chem. 2015, 80(19), 9392; Zhang et al., Tetrahedron Lett. 2021, 66, 152835; Hikawa et al., J. Org. Chem. 2012, 77(16), 7046; Garad et al., J. Org. Chem.

2017, 82(12), 6366; Kim et al., Tetrahedron Lett. 2014, 55(15), 2340; Qian et al., J. Org. Chem.

2018, 83(16), 9201).

[000144] In Scheme 2, alcohols 6 can be converted to anthranilic acid derivatives 11 via a number of different methodologies. The alcohol functionality of 6 may first be converted to a leaving group such as a bromide or a mesylate utilizing commonly known methods.

Nucleophilic displacement of the benzylic reactive groups of 10 by an anthranilic acid ester (methyl, t-butyl or other commonly employed esters may be used) can give compounds 11. An alternate approach to the preparations of compounds 11 is to use a Mitsunobu type of reaction of the anthranilic acid ester with alcohols 6 to give 11 directly. In some cases, the transient use of an activating group (such as a 2,4-dinitrobenzenesulfonyl group) on the anthranilic acid amine functionality can facilitate the Mitsunobu reaction. The use of 2,3- dichloro-5,6-dicyanobenzoquinone (DDQ) and triphenylphosphine may also be employed in the direct reaction of the alcohol 6 with the anthranilic acid ester (Shalit, T.; et al., Tetrahedron Lett. 2010, 51, 5988-5991; Iranpoor, N.; et al., Tetrahedron 2009, 65, 3893-3899; Panday, S. K., Mini-Rev. Org. Chem. 2019, 16(2), 127-140; Fukuyama, Tohru; et al., Tetrahedron Lett. 1997, 38(33), 5831-5834). Ester intermediates 11 can be converted to the corresponding carboxylic acids. In some exemplary embodiments when R = Me, de-esterification of intermediates 11 can be achieved via treatment with metal hydroxides (e.g., but not limited to, LiOH or NaOH). In other exemplary embodiments, intermediates 11 when R = tert-butyl may be de-esterified by their treatment with an appropriate acid (e.g., but not limited to, TFA, or HCI in 1,4-dioxane or water). Separation of racemic acids via known chiral HPLC chromatographic techniques (e.g., but not limited to, use of DAICEL Chiralpak columns) may yield enantiomerically enriched compounds of general structures 12 and 13.

Scheme 2

[000145] An alternate mode of synthesis leading to single enantiomer intermediates 18 is depicted in Scheme 3. This reaction sequence utilizes the formation of a chiral sulfinyl imine to set the stereocenter of the subsequent synthetic products. Ketones 5 may be converted to a chiral sulfinyl-imines 14 via known procedures, which may then in turn be reduced to sulfinylamines 15 in a stereo-controlled fashion using suitable reducing agents (Datta and Ellman, J. Org. Chem. 2010, 75, 6283-6285; Ellman et al., Acc. Chem. Res. 2002, 35, 984-995; Ellman et al., J. Org. Chem. 2007, 72, 626-629; Colyer et al., J. Org. Chem. 2006, 71(18), 6859-6862). The use of the R isomer of the sulfinyl group 14, generally, results predominantly in the R,R-isomer of 15 when (for example) the reducing agent used is a mixture of sodium borohydride and cerium chloride-heptahydrate. The use of this particular reducing system has been shown to be effective at reducing imines and may often give enhanced stereo-control in similar reductions (Hua et al., Synthesis 1991, (11), 970-4; Zhu et al., J. Chem. Res. 2015, 39(7), 390-393). The major isomer may be separated from the other minor isomer via standard chromatographic means. As has been demonstrated in the preceding literature references, a judicious choice of the antipode of the sulfinyl-imine and the reducing agent may give access to either antipode of the sulfinyl-amine. Sulfinyl-amines 15 can be cleaved to the single enantiomer of chiral amines 16 using standard conditions (such as hydrogen chloride on dioxane). Standard coupling reactions of amines 16 with aryl iodides 17 (such as an Ullman coupling or Buchwald-Hartwig coupling) may then give the resulting anthranilic acid derivatives 18 (Yang et al., Org. Process Res. & Dev. 2022, 26(6), 1690-1750; Surry and Buchwald, Chem. Sci. 2011, 2(1), 27-50). Ester hydrolysis may then provide carboxylic acids 19.

[000146] While Schemes 1-3 describe the general approaches used to prepare the compounds of Formulae (l)-(6) of the present invention, the specific order of the reactions performed in each of the Schemes may be modified to optimize the preparation of particular compounds.

[000147] The following compounds represent various embodiments of the present invention where only R4 has been varied in a structure where all of the other "R" variables have been set. The listing of substituents within brackets for a given compound indicates individual compounds containing one of each of the substituents. or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer, wherein the listing of substituents within brackets indicates individual compounds containing one of each of the substituents, and wherein R 4 is selected from wherein X = CH or N, with the proviso that in a given ring the number of nitrogen atoms is no more than 4, or wherein R 4 is selected from wherein X = CH or N, with the proviso that in a given ring the number of nitrogen atoms is no more than 4.

[000148] The following compounds represent various embodiments of the present invention where each R 1 and R 4 have been varied in a structure where all other "R" variables have been set. The listing of substituents within brackets for a given compound indicates individual compounds containing one of each of the substituents. or a solvate, enantiomer, diastereomer, tautomer, polymorph or isotope-labeled compound, or a pharmaceutically acceptable salt thereof, wherein the carbon marked with * is a chiral center and exists as a (R)- and (S)-racemic mixture or as either the (R)- or (S)- enantiomer, wherein the listing of substituents within brackets indicates individual compounds containing one of each of the substituents, and wherein R4 is selected from wherein X = CH or N, with the proviso that in a given ring the number of nitrogen atoms is no more than 4, or wherein R4 is selected from wherein X = CH or N, with the proviso that in a given ring the number of nitrogen atoms is no more than 4; and wherein R/ is selected from

wherein X is CH or N.

Experimental

[000149] All commercially available solvents and reagents were used as received. All

NMR spectra were recorded using a Bruker Avance III HD 300 MHz or Bruker Avance III HD 400 MHz. MS samples were analyzed on a Shimadzu LCMS-2020 mass spectrometer with electrospray ionization operating in positive and negative ion mode. Samples were introduced into the mass spectrometer using chromatography. All final products had a purity of > 90 %, unless specified otherwise in the experimental details. HPLC purity was measured on a Shimadzu Acquity HPLC system.

[000150] The following represents acronyms used in the experimental section for well- known chemical solvents, reagents, parameters and techniques: 1H NMR: proton nuclear magnetic resonance spectroscopy

ACN: acetonitrile

AcOH: acetic acid c-alkyl: cycloalkyl c-Bu: cyclobutyl c-Pr: cyclopropyl

CeC3: cerium (III) chloride CH 2 CI 2 : dichloromethane

CHCI 3 : chloroform

CS2CO3: cesium carbonate

DBAD: di-tert-butyl azodicarboxylate

DCM: dichloromethane

DIBAL: diisobutylaluminum hydride

DIEA: N ,/\/-diisopropylethylamine

DMF: N ' N -dimethylformamide

DMSO: dimethyl sulfoxide

DTAD: di-tert-butyl azodicarboxylate

EA: ethyl acetate ee: enantiomeric excess

Et 2 O: diethyl ether

Et 3 N: triethylamine

EtOAc: ethyl acetate

EtOH: ethanol

FA: formic acid H 2 O: water h: hours

HATU: l-[Bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5-b]pyri dinium 3-oxide hexafluorophosphate

HCI: hydrochloric acid

Hex: hexanes

HPLC: high-performance liquid chromatography

IPA: isopropanol

K 2 CO 3 : potassium carbonate KOAc: potassium acetate

LiOH: lithium hydroxide mCPBA: meto-chloroperoxybenzoic acid

Me: methyl

MeCN: acetonitrile

MeOH: methanol mg: milligram min: minutes mL: milliliter

MS2O: methanesulfonic anhydride

MsCI: methanesulfonyl chloride

NaBFU: sodium borohydride

N 2 : nitrogen

Na 2 CO 3 : sodium carbonate

Na 2 SO 4 : sodium sulfate

NaCI: sodium chloride

NaH: sodium hydrideNaOH: sodium hydroxide

NaHCO 3 : sodium bicarbonate

NaH 2 PO4: monosodium phosphate

NH 3 : ammonia

NH4HCO 3 : ammonium bicarbonate

NMP: N -methylpyrrolidone

Oxetane: 4-membered ring containing 3 carbon ring atoms and 1 oxygen ring atom.

PBr 3 : phosphorous tribromide

PCI5: phosphorous pentachloride Pd-PEPPSI-IHeptCI 3-chloropyridine: dichloro[l,3-bis(2,6-di-4-heptylphenyl)imidazol-2- ylidene](3-chloropyridyl)palladium(ll)

Pd(dppf)Ck: (l,l'-bis(diphenylphosphino)ferrocene)palladium(ll) dichloride

Pd(PPhs)4: tetrakis(triphenylphosphine)palladium(0)

Pd2(dba)s: tris(dibenzylideneacetone)dipalladium(0)

PdCLfPPhsh: bis(triphenylphosphine)palladium(ll) dichloridePE: petroleum ether

POCh: phosphorus oxychloride

PPhs: triphenylphosphine

Prep: preparative

RuPhos: 2-dicyclohexylphosphino-2',6'-diisopropoxybiphenyl

TEA: triethylamine

TFA: trifluoroacetic acid

THF: tetrahydrofuran

Ti(Oi-Pr) 4 : Titanium(IV) isopropoxide

TLC: thin-layer chromatography

Xantphos: 4,5-bis(diphenylphosphino)-9,9-dimethylxanthene

EXAMPLES

[000151] Example 1: 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000152] Example 2: 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 2)

Step 1: Preparation of 8-bromo-2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-2,3-dihydroq uinazolin- 4(lH)-one.

[000153] A solution of 2-amino-3-bromo-N,5-dimethylbenzamide (600 mg, 2.47 mmol) in THF (6 mL) was treated with 5-fluoro-lH-indole-2-carbaldehyde (0.48 g, 2.96 mmol) and 4- methylbenzene-l-sulfonic acid (0.85 g, 4.94 mmol) for 16 h at 25 °C. The reaction was quenched with H 2 O (20 mL) and the resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 5:1) to afford 8-bromo-2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-2,3- dihydroquinazolin-4(lH)-one (400 mg, 41%) as an off-white solid. MS: (ES + ) m/z = 388.0 [M+H] + . Step 2: Preparation of 8-bromo-2-(5-fluoro-lH-indol-2-yl)-3,6-dimethylquinazolin-4( 3H)-one.

[000154] A solution of 8-bromo-2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-2,3- dihydroquinazolin-4(lH)-one (500 mg, 1.29 mmol) in MeOH (5 mL) was treated with DDQ (0.58 g, 2.58 mmol) for 2 h at room temperature. The reaction was quenched with H 2 O (15 mL) and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by reverse phase flash chromatography with the following conditions: column, C18 silica gel; mobile phase, MeCN in Water (0.1% FA), 10% to 50% gradient in 10 min; detector, UV 254 nm. This resulted in the isolation of 8-bromo-2-(5-fluoro- lH-indol-2-yl)-3,6-dimethylquinazolin-4(3H)-one (480 mg, 96%) as an off-white solid. MS: (ES + ) m/z = 385.9 [M+H] + .

Step 3: Preparation of 8-acetyl-2-(5-fluoro-lH-indol-2-yl)-3,6-dimethylquinazolin-4 (3H)-one.

[000155] A solution of 8-bromo-2-(5-fluoro-lH-indol-2-yl)-3,6-dimethylquinazolin-4( 3H)- one (600 mg, 1.55 mmol) in dioxane (6 mL) was treated with tributyl (1- ethoxyethenyl)stannane (1.12 g, 3.11 mmol) for 16 h at 110 °C. The mixture was cooled to room temperature, aqueous 1 M HCI (10 mL) was added, and the mixture was then stirred for 30 minutes. The resulting mixture was extracted with EtOAc (3 x 150 mL). The combined organic layers were washed with brine (3 x 100 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EA = 2:1) to afford 8-acetyl-2-(5-fluoro-lH-indol-2-yl)-3,6- dimethylquinazolin-4(3H)-one (400 mg, 73%) as an off-white solid. MS: (ES + ) m/z = 350.2 [M+H] + .

Step 4: Preparation of 2-(5-fluoro-lH-indol-2-yl)-8-(l-hydroxyethyl)-3,6-dimethylqu inazolin-

4(3H)-one.

[000156] Sodium borohydride (82 mg, 2.18 mmol) was added in portions to a solution of 8- acetyl-2-(5-fluoro-lH-indol-2-yl)-3,6-dimethylquinazolin-4(3 H)-one (380 mg, 1.09 mmol) in MeOH (5 mL) under a nitrogen atmosphere at 0 °C. The resulting mixture was stirred for 1 h at room temperature and then was quenched by the addition of H 2 O (2 mL). The resulting mixture was concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 1:2) to afford 2-(5-fluoro-lH-indol-2-yl)-8-(l-hydroxyethyl)-3,6-dimethylqu inazolin- 4(3H)-one (200 mg, 52%) as an off-white solid. MS: (ES + ) m/z = 352.2 [M+H] + .

Step 5: Preparation of tert-butyl 2-((N-(l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)-2-nitrophenyl)sulfonamido)benz oate.

[000157] A solution of 2-(5-fluoro-lH-indol-2-yl)-8-(l-hydroxyethyl)-3,6- dimethylquinazolin-4(3H)-one (200 mg, 0.57 mmol) in THF (10 mL) was treated with PPhs (298 mg, 1.14 mmol) for 5 min at room temperature followed by the addition of DTAD (327 mg, 1.42 mmol) in portions at room temperature. A mixture was stirred for 1 h at room temperature. The reaction was quenched with water (30 mL) at room temperature and the resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 1:1) to afford tert-butyl 2- ((N-(l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4-di hydroquinazolin-8-yl)ethyl)-2- nitrophenyl)sulfonamido)benzoate (250 mg, 61%) as an off-white solid. MS: (ES + ) m/z = 712.2 [M+H] + .

Step 6: Preparation of tert-butyl 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate.

[000158] A solution of tert-butyl 2-((N-(l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo- 3,4-dihydroquinazolin-8-yl)ethyl)-2-nitrophenyl)sulfonamido) benzoate (250 mg, 0.35 mmol) in DMF (10 mL) was treated with K 2 CO 3 (0.19 g, 1.44 mmol) and (phenylsulfanyl)potassium (0.21 g, 1.40 mmol) in portions at room temperature. The mixture was stirred for 1 h at 60 °C. The reaction was quenched with H 2 O (10 mL) at room temperature and the resulting mixture was extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 3:1) to afford tert-butyl 2-((l-(2-(5-fluoro-lH- indol-2-yl)-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl)et hyl)amino)benzoate (100 mg, 54%) as an off-white solid. MS: (ES + ) m/z = 527.2 [M+H] + .

Step 7: Preparation of 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid. [000159] A solution of tert-butyl 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate (150 mg, 0.32 mmol) and TFA (2 mL) in DCM (2 mL) was stirred for 3 h at 30 °C . The resulting mixture was concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 1:1) to afford 2-((l-(2-(5-fluoro-lH- indol-2-yl)-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl)et hyl)amino)benzoic acid (90 mg, 60%) as an off-white solid. MS: (ES + ) m/z = 471.0 [M+H] + .

Step 8: Preparation of 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomers 1 and 2).

[000160] Racemic 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (50 mg) was separated by chiral prep-HPLC with the following conditions: Column: CHIRALPAK IG, 2*25 cm, 5 pm; Mobile Phase A: Hex(0.1% FA)-HPLC, Mobile Phase B: EtOH-HPLC; Flow rate: 20 mL/min; Gradient: 10% B to 10% B in 26 min; Wavelengths: 220/254 nm; RTl(min): 19.695; RT2(min): 22.84; Sample Solvent: EtOH-HPLC; Injection Volume: 0.5 mL. This resulted in the isolation of each enantiomer of 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4-dih ydroquinazolin-8- yl)ethyl)amino)benzoic acid as a white solid.

[000161] 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4-dih ydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 1 (17.6 mg, 34%, 99.9% ee). MS: (ES + ) m/z = 471.0 [M+H] + . 1 H NMR: (400 MHz, CD 3 OD) 6 7.95 - 7.89 (m, 2H), 7.68 - 7.57 (m, 2H), 7.48 - 7.40 (m, 1H), 7.21 (s, 1H), 7.15 - 7.00 (m, 2H), 6.57 (d, J = 8.5 Hz, 1H), 6.55 - 6.45 (m, 1H), 5.75 - 5.66 (m, 1H), 3.96 (s, 3H), 2.42 (s, 3H), 1.66 (d, J = 6.6 Hz, 3H).

[000162] 2-((l-(2-(5-fluoro-lH-indol-2-yl)-3,6-dimethyl-4-oxo-3,4-dih ydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 2 (18.8 mg, 37%, 98.6% ee). MS: (ES + ) m/z = 471.0 [M+H] + . 1 H NMR: (300 MHz, CD3OD) 6 7.95 - 7.83 (m, 2H), 7.68 - 7.55 (m, 2H), 7.48 - 7.40 (m, 1H) , 7.20 (s, 1H), 7.17 - 6.99 (m, 2H), 6.58 (d, J = 8.5 Hz, 1H), 6.55 - 6.45 (m, 1H), 5.75 - 5.66 (m, 1H), 3.95 (s, 3H), 2.42 (s, 3H), 1.66 (d, J = 6.7 Hz, 3H).

[000163] Example 3: 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin- 8- yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000164] Example 4: 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin- 8- yl)ethyl)amino)benzoic acid (Enantiomer 2)

Step 1: Preparation of methyl 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin- 8- yl)ethyl)amino)benzoate.

[000165] To a stirred solution of 8-(l-hydroxyethyl)-2-isopentyl-3,6-dimethylquinazolin- 4(3H)-one (440 mg, 1.52 mmol, prepared in a manner similar to Examples 1 and 2 using 4- methylpentanal in place of 5-fluoro-lH-indole-2-carbaldehyde), methyl 2-(2,4- dinitrobenzenesulfonamido)benzoate (640 mg, 1.67 mmol), and PPh 3 (1.00 g, 3.81 mmol) in THF (15 mL) at 0 °C was slowly added dropwise a solution of DTAD (1.05 g, 4.57 mmol) in THF (5 mL). The resulting solution was allowed to react overnight at room temperature. The mixture was concentrated under reduced pressure and the product was purified by prep TLC (PE/EA = 2:1) to afford methyl 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin- 8- yl)ethyl)amino)benzoate (500 mg, 77%) as a yellow semi-solid. MS: (ES + ) m/z = 422.2 (M+H) + .

Step 2: Preparation of 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin- 8- yl)ethyl)amino)benzoic acid (Enantiomers 1 and 2).

[000166] A solution of methyl 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin- 8-yl)ethyl)amino)benzoate (400 mg, 0.94 mmol) and NaOH (189 mg, 4.74 mmol) in MeOH (6 mL) and H 2 O (1.2 mL) was stirred for two days at 40 °C. The mixture was cooled to room temperature, adjusted to pH 5-6 with aqueous 2 M HCI, and extracted with ethyl acetate (3 x 60 mL). The combined organic layers were washed with water (2 x 30 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by prep TLC (CH 2 CI 2 /MeOH = 15:1) to afford 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo- 3,4-dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (250 mg, 64%) as a yellow semi-solid. MS: (ES + ) m/z = 408.1 (M+H) + .

[000167] Racemic 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin- 8- yl)ethyl)amino)benzoic acid (200 mg) was separated by chiral prep-HPLC with the following conditions: Column: (R, R)-WHELK-Ol-Kromasil, 2.11*25 cm, 5 pm; Mobile Phase A: EtOH- HPLC, Mobile Phase B: Hex(0.1% FA)-HPLC; Flow rate: 20 mL/min; Gradient: 20% B to 20% B in 37 min; Wavelengths: 220/254 nm; RTl(min): 23.848; RT2(min): 30.603; Sample Solvent: EtOH- HPLC; Injection Volume: 0.9 mL. This resulted in the isolation of both enantiomers of 2-((l-(2- isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl)ethy l)amino)benzoic acid.

[000168] 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin- 8- yl)ethyl)amino)benzoic acid Enantiomer 1 (42 mg, 21%, white solid, 99.7% ee). MS: (ES + ) m/z = 408.1 (M+H) + . 1 H NMR (400 MHz, DMSO-d6) 6 12.67 (s, 1H), 8.48 (s, 1H), 7.81 - 7.74 (m, 2H),

7.52 (d, J = 2.1 Hz, 1H), 7.21 - 7.03 (m, 1H), 6.55 - 6.39 (m, 1H), 6.42 (d, J = 8.5 Hz, 1H), 5.54 (s, 1H), 3.57 (s, 3H), 3.00 - 2.90 (m, 2H), 2.35 (s, 3H), 1.80 - 1.72 (m, 3H), 1.55 (d, J = 6.6 Hz, 3H), 1.02 - 0.94 (m, 6H).

[000169] 2-((l-(2-isopentyl-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin- 8- yl)ethyl)amino)benzoic acid Enantiomer 2 (40 mg, 19%, yellow solid, 99.3% ee). MS: (ES + ) m/z): 408.1 [M+H] + . 1 H NMR: (300 MHz, DMSO-d6) 6 12.66 (s, 1H), 8.47 (s, 1H), 7.83 - 7.74 (m, 2H),

7.53 (d, J = 2.1 Hz, 1H), 7.21 - 7.03 (m, 1H), 6.55 - 6.39 (m, 2H), 5.55 (s, 1H), 3.58 (s, 3H), 3.00 - 2.90 (m, 2H), 2.36 (s, 3H), 1.86 - 1.70 (m, 3H), 1.56 (d, J = 6.7 Hz, 3H), 1.02 - 0.94 (m, 6H).

[000170] Example 5: 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000171] Example 6: 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid (Enantiomer 2)

Step 1: Preparation of 8-(l-bromoethyl)-2-(4-chlorophenyl)-3,6-dimethylquinazolin-4 (3H)-one.

[000172] A solution of 2-(4-chlorophenyl)-8-(l-hydroxyethyl)-3,6-dimethylquinazolin -4(3H)- one (prepared in a manner similar to Examples 1 and 2 using 4-chlorobenzaldehyde in place of 5-fluoro-lH-indole-2-carbaldehyde, 560 mg, 1.70 mmol) in DCM was treated with PBr 3 (0.5 mL, 5.26 mmol) at 0 °C. The resulting mixture was stirred for overnight at room temperature and then was adjusted to pH 8 with saturated aqueous Na 2 CO 3 . The resulting mixture was extracted with CH 2 CI 2 (3 x 30 mL). The combined organic layers were washed with brine (2 x 20 mL) and then dried over anhydrous Na 2 SC>4. After filtration, the filtrate was concentrated under reduced pressure to afford 8-(l-bromoethyl)-2-(4-chlorophenyl)-3,6-dimethylquinazolin-4 (3H)- one (660 mg, 98%) as a light brown solid. MS: (ES + ) m/z = 391.0 [M+H] + .

Step 2: Preparation of methyl 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate.

[000173] A solution of 8-(l-bromoethyl)-2-(4-chlorophenyl)-3,6-dimethylquinazolin-4 (3H)- one (660 mg, 2.01 mmol) in acetone was treated with methyl anthranilate (0.44 mL, 2.90 mmol) at room temperature. The resulting mixture was stirred overnight at 90 °C and then was concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 5:1) to afford methyl 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoate (540 mg, 58%) as an off-white solid. MS: (ES + ) m/z = 462.1 [M+H] + .

Step 3: Preparation of 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid (Enantiomers 1 and 2). [000174] A solution of methyl 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate (540 mg, 1.17 mmol) and NaOH (233 mg, 5.83 mmol) in MeOH (10 mL) and H 2 O (2 mL) was stirred overnight at 50 °C. The mixture was adjusted to pH 6 with aqueous 1 M HCI. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (3 x 10 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by prep TLC (CH 2 CL 2 MeOH = 15:1) to afford 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (220 mg, 42%) as an off-white solid. MS: (ES + ) m/z = 448.1[M+H] + .

[000175] Racemic 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid (150 mg) was separated by chiral prep HPLC with the following conditions: Column: CHIRALPAK ID, 2*25 cm, 5 pm; Mobile Phase A: Hex(0.1% FA)-HPLC, Mobile Phase B: IPA-HPLC; Flow rate: 20 mL/min; Gradient: 7% B to 7% B in 23 min; Wavelengths: 220/254 nm; RTl(min): 13.609; RT2(min): 17.646; Sample Solvent: EtOH-HPLC; Injection Volume: 0.5 mL. This resulted in the isolation of each enantiomer of 2-((l-(2-(4- chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl) ethyl)amino)benzoic acid as a white solid.

[000176] 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid Enantiomer 1 (41.1 mg, 27%, 99.8% ee). MS: (ES + ) m/z = 448.1 [M+H] + . 1 H NMR: (400 MHz, DMSO-d6) 6 12.70 (s, 1H), 8.41 (d, J = 6.7 Hz, 1H), 7.90 - 7.85 (m, 1H), 7.86 - 7.75 (m, 3H), 7.69 - 7.61 (m, 2H), 7.57 (s, 1H), 7.20 - 7.12 (m, 1H), 6.55 - 6.45 (m, 1H), 6.40 (d, J = 8.5 Hz, 1H), 5.49 - 5.43 (m, 1H), 3.41 (s, 3H), 2.39 (s, 3H), 1.53 (d, J = 6.6 Hz, 3H).

[000177] 2-((l-(2-(4-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid Enantiomer 2 (38 mg, 25%, ~98.4% ee). MS: (ES + ) m/z = 448.1 [M+H] + . 1 H NMR: (400 MHz, DMSO-d6) 6 12.70 (s, 1H), 8.42 (d, J = 6.6 Hz, 1H), 7.90 - 7.85 (m, 1H), 7.86 - 7.75 (m, 3H), 7.69 - 7.61 (m, 2H), 7.58 (s, 1H), 7.20 - 7.12 (m, 1H), 6.54 - 6.45 (m, 1H), 6.40 (d, J = 8.5 Hz, 1H), 5.50 - 5.42 (m, 1H), 3.41 (s, 3H), 2.39 (s, 3H), 1.53 (d, J = 6.6 Hz, 3H).

[000178] Example 7: 2-((l-(2-(3-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid (Enantiomer 1) [000179] Example 8: 2-((l-(2-(3-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid (Enantiomer 2)

[000180] Prepared in a manner similar to Examples 5 and 6 using 3-chlorobenzaldehyde in place of 4-chlorobenzaldehyde.

[000181] 2-((l-(2-(3-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid Enantiomer 1. MS: (ES + ) m/z = 448.1 [M+H] + . NMR: (DMSO-d6, 400 MHz): 6 12.68 (s, 1H), 8.41 (d, J = 6.4 Hz, 1H), 7.85 - 7.78 (m, 2H), 7.82 - 7.70 (m, 2H), 7.70

- 7.55 (m, 3H), 7.21 - 7.11 (m, 1H), 6.55 - 6.34 (m, 2H), 5.51 - 5.43 (m, 1H), 3.40 (s, 3H), 2.39 (s, 3H), 1.53 (d, 7 = 6.6 Hz, 3H).

[000182] 2-((l-(2-(3-chlorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquin azolin-8- yl)ethyl)amino)benzoic acid Enantiomer 2. MS: (ES + ) m/z = 448.1 [M+H] + . 1 H NMR: (DMSO-d6, 400 MHz): 6 12.70 (s, 1H), 8.42 (d, J = 6.3 Hz, 1H), 7.85 - 7.78 (m, 2H), 7.82 - 7.72 (m, 2H), 7.70

- 7.55 (m, 3H), 7.20 - 7.12 (m, 1H), 6.54 - 6.34 (m, 2H), 5.51 - 5.43 (m, 1H), 3.40 (s, 3H), 2.39 (s, 3H), 1.53 (d, 7 = 6.7 Hz, 3H).

[000183] Example 9: 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000184] Example 10: 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 2)

Step 1: Preparation of N-(2-bromo-4-methyl-6-(methylcarbamoyl)phenyl)-2,3-dihydro-l H- indene-2-carboxamide.

[000185] To a stirred solution of 2-amino-3-bromo-N,5-dimethylbenzamide (1.5 g, 6.17 mmol) and 2,3-dihydro-lH-indene-2-carboxylic acid (1.00 g, 6.17 mmol) in ACN (100 mL) was added pyridine (2.44 g, 30.85 mmol) and T3P (7.85 g, 24.68 mmol) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred overnight at 50 °C under nitrogen atmosphere. The resulting mixture was quenched with H 2 O (50 mL) and extracted with EtOAc (2 x 50 mL). The combined organic layers were washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (1% to 5% MeOH in CH 2 CI 2 over 15 min) to afford N-(2-bromo-4-methyl-6-(methylcarbamoyl)phenyl)-2,3-dihydro-l H-indene-2-carboxamide (1.9 g, 79%) as a white solid. MS: (ES + ) m/z = 387.1 [M+H] + .

Step 2: Preparation of 8-bromo-2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethylquinazolin -4(3H)- one.

[000186] To a stirred solution of N-[2-bromo-4-methyl-6-(methylcarbamoyl)phenyl]-2,3- dihydro-lH-indene-2-carboxamide (1.9 g, 4.91 mmol) in DMF (20 mL) was added K 2 COs (1.36 g, 9.81 mmol) in portions at room temperature under nitrogen atmosphere. The resulting mixture was stirred for 3 h at 90 °C under nitrogen atmosphere. The reaction was quenched with H 2 O (50 mL) and extracted with EtOAc (2 x 80 mL). The combined organic layers were washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 , and concentrated under reduced pressure. This resulted in 8-bromo-2-(2,3-dihydro-lH-inden-2-yl)-3,6- dimethylquinazolin-4(3H)-one (1.7 g, 93%) as an off-white solid. MS: (ES + ) m/z = 369.1 [M+H] + .

Step 3: Preparation of 8-acetyl-2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethylquinazoli n-4(3H)- one.

[000187] A mixture of 8-bromo-2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethylquinazolin - 4(3H)-one (1.7 g, 4.60 mmol), tributyl(l-ethoxyethenyl)stannane (2.00 g, 5.53 mmol) and Pd(PPh 3 ) 4 (0.53 g, 0.46 mmol) in anhydrous 1,4-dioxane (20 mL) was stirred for overnight at 100 °C under a nitrogen atmosphere. The reaction mixture was cooled to room temperature and diluted with aqueous 1 M HCI (12 mL). After stirring for 20 min at 50 °C, the reaction mixture was cooled to room temperature and then diluted with water (40 mL) and extracted with ethyl acetate (3 x 80 mL). The combined organic layers were washed with water (3 x 40 mL) and then brine (20 mL). The organics were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to result in the isolation of 8- acetyl-2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethylquinazolin- 4(3H)-one (900 mg, 58%) as an off-white solid. MS: (ES + ) m/z = 333.2 [M+H] + .

Step 4: Preparation of 2-(2,3-dihydro-lH-inden-2-yl)-8-(l-hydroxyethyl)-3,6- dimethylquinazolin-4(3H)-one.

[000188] To a stirred 0 °C solution of 8-acetyl-2-(2,3-dihydro-lH-inden-2-yl)-3,6- dimethylquinazolin-4(3H)-one (900 mg, 2.71 mmol) in MeOH (15 mL) was slowly added NaBH 4 (204 mg, 5.4 mmol) in portions. The resulting mixture was stirred for 2 h at rt and was then quenched with H 2 O (20 mL) and extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (2 x 30 mL), dried over anhydrous Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography, (PE/EA = 3:1) to afford 2-(2,3-dihydro-lH-inden-2-yl)-8-(l-hydroxyethyl)-3,6- dimethylquinazolin-4(3H)-one (700 mg, 77%) as an off-white solid. MS: (ES + ) m/z = 335.1 [M+H] + .

Step 5: Preparation of tert-butyl 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate. [000189] To a solution of 2-(2,3-dihydro-lH-inden-2-yl)-8-(l-hydroxyethyl)-3,6- dimethylquinazolin-4(3H)-one (500 mg, 1.50 mmol), tert-butyl 2-(2,4- dinitrobenzenesulfonamido)benzoate (633 mg, 1.50 mmol) and PPhs (980 mg, 3.74 mmol) in THF (12 mL) was added a solution of (E)-N-[[(tert-butoxy)carbonyl]imino](tert- butoxy)formamide (1.03 g, 4.49 mmol) in tetrahydrofuran (2 mL) at 0 °C under a nitrogen atmosphere. The resulting solution was stirred overnight at room temperature. The resulting solution was concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 2:1) to afford tert-butyl 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate (450 mg, 59%) as a light orange viscous oil. MS: (ES+) m/z = 510.4 [M+H] + .

Step 6: Preparation of 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid.

[000190] A solution of tert-butyl 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo- 3,4-dihydroquinazolin-8-yl)ethyl)amino)benzoate (450 mg, 0.88 mmol) in 4 M HCI in dioxane (12 mL, 48 mmol) was stirred at room temperature overnight. The resulting solution was concentrated under reduced pressure and the product was purified by prep TLC (CI-bCk/MeOH = 25:1) to afford 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (160 mg, 40%) as a light yellow solid. MS: (ES + ) m/z = 454.2 [M+H] + .

Step 7: Preparation of 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomers 1 and 2).

[000191] A racemic mixture of 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (160 mg) was separated by chiral HPLC with the following conditions: Column: CHIRALPAK AD-H, 2*25 cm, 5 pm; Mobile Phase A: Hex(0.1% FA)-HPLC, Mobile Phase B: IPA-HPLC; Flow rate: 20 mL/min; Gradient: 5% B to 5% B in 12 min; Wavelengths: 220/254 nm; RTl(min): 17.051; RT2(min): 20.946; Sample Solvent: EtOH-HPLC; Injection Volume: 0.4 mL. This resulted in the isolation of both enantiomers of 2- ((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4-di hydroquinazolin-8- yl)ethyl)amino)benzoic acid as off-white solids.

[000192] 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 1 (48.7 mg, 30%, >99% ee). MS: (ES + ) m/z = 454.2 [M+H] + . 1 H NMR: (400 MHz, DMSO-d6) 6 12.70 (s, 1H), 8.39 (s, 1H), 7.81 - 7.72 (m, 2H), 7.47 (d, J = 2.2 Hz, 1H), 7.32 - 7.22 (m, 2H), 7.20 - 7.07 (m, 2H), 7.02 - 7.01 (m, 1H), 6.49 - 6.41 (m, 1H), 6.24 (d, J = 8.5 Hz, 1H), 5.44 - 5.36 (m, 1H), 4.21 - 4.09 (m, 1H), 3.68 (s, 3H), 3.45 -

4.42 (m, 4H), 2.33 (s, 3H), 1.35 (d, J = 6.6 Hz, 3H).

[000193] 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 2 (60.1 mg, 37%, >99% ee). MS: (ES + ) m/z = 454.2 [M+H] + . 1 H NMR: (400 MHz, DMSO-d6) 6 12.72 (s, 1H), 8.42 (s, 1H), 7.81 - 7.72 (m, 2H), 7.47 (d, J = 2.1 Hz, 1H), 7.30 - 7.22 (m, 2H), 7.19 - 7.07 (m, 2H), 7.02 - 7.01 (m, 1H), 6.49 - 6.41 (m, 1H), 6.23 (d, J = 8.5 Hz, 1H), 5.44 - 5.36 (m, 1H), 4.21 - 4.09 (m, 1H), 3.68 (s, 3H), 3.45 -

3.42 (m, 4H), 2.33 (s, 3H), 1.35 (d, J = 6.7 Hz, 3H).

[000194] Example 11: 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000195] Example 12: 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 2)

Isomer 1 Isomer 2

[000196] Prepared in a manner similar to Examples 9 and 10 using l-methyl-2- oxopiperidine-4-carboxylic acid in place of 2,3-dihydro-lH-indene-2-carboxylic acid.

[000197] 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 1, 99.9% ee. MS: (ES + ) m/z = 449.2 [M+H] + . 1 H NMR: (300 MHz, CD 3 OD) 6 7.86-7.83 (m, 2H), 7.57 (s, 1H), 7.09 - 7.07 (m, 1H), 6.48 - 6.38 (m, 2H), 5.60 - 5.54 (m, 1H), 3.80 - 3.78 (m, 1H), 3.78 (s, 3H), 3.46 - 3.44 (m, 1H), 3.24-3.22 (m, 1H), 3.06 - 2.93 (m, 4H), 2.74 - 2.66 (m, 1H), 2.34 (s, 3H), 2.24 - 2.20 (m, 2H), 1.52 (d, J = 6.6 Hz, 3H).

[000198] 2-((l-(2-(2,3-dihydro-lH-inden-2-yl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 2, 99.9% ee. MS: (ES + ) m/z = 449.2 [M+H] + . 1 H NMR: (300 MHz, CD3OD) 6 7.86 - 7.83 (m, 2H), 7.57 (s, 1H), 7.09 - 7.07 (m, 1H), 6.48 - 6.38 (m, 2H), 5.60 - 5.54 (m, 1H), 3.80 - 3.78 (m, 1H), 3.78 (s, 3H), 3.46 - 3.44 (m, 1H), 3.24 - 3.22 (m, 1H), 3.06 - 2.93 (m, 4H), 2.74 - 2.66 (m, 1H), 2.34 (s, 3H), 2.24 - 2.20 (m, 2H), 1.52 (d, J = 6.6 Hz, 3H).

[000199] Example 13: 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquina zolin-8- yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000200] Example 14: 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquina zolin-8- yl)ethyl)amino)benzoic acid (Enantiomer 2)

Step 1: Preparation of 3-acetyl-2-amino-N,5-dimethylbenzamide.

[000201] To a stirred solution of 2-amino-3-bromo-N,5-dimethylbenzamide (1.00 g, 4.11 mmol) in dioxane (10 mL) was added tributyl(l-ethoxyethenyl)stannane (1.78 g, 4.94 mmol) and Pd(PPh 3 ) 4 (0.48 g, 0.41 mmol) in portions at room temperature. The resulting mixture was stirred overnight at 100 °C under a N 2 atmosphere and then was cooled to room temperature. Aqueous 1 M HCI (5 mL) was added in portions and the resulting mixture was stirred for 20 min at room temperature. The pH of the mixture was adjusted to 8-9 with 15 wt% aqueous NaHCO 3 solution. The resulting mixture was extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EA = 1:1) to afford 3-acetyl-2-amino-N,5-dimethylbenzamide (0.42 g, 50%) as a yellow oil. MS: (ES + ) m/z = 207.2 [M+H] + .

Step 2: Preparation of 2-amino-3-(l-hydroxyethyl)-N,5-dimethylbenzamide.

[000202] To a stirred solution of 3-acetyl-2-amino-N,5-dimethylbenzamide (0.6 g, 2.91 mmol) in MeOH (20 mL) was added NaBH 4 (0.44 g, 11.6 mmol) in portions slowly at 0 °C. The resulting mixture was stirred for 4 h at room temperature and then was quenched with H 2 O (20 mL). The resulting mixture was extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (DCM/MeOH = 15:1) to afford 2-amino-3-(l-hydroxyethyl)-N,5-dimethylbenzamide (400 mg, 66%) as a light yellow oil. MS: (ES + ) m/z = 209.1 [M+H] + .

Step 3: Preparation of 2-amino-3-(l-((tert-butyldimethylsilyl)oxy)ethyl)-N,5- dimethylbenzamide. 5.0 eq TBSCI, 15 eq 1 H-lmidazole, DMF, 0 °C~rt, overnight

[000203] To a stirred solution of 2-amino-3-(l-hydroxyethyl)-N,5-dimethylbenzamide (400 mg, 1.92 mmol) in DMF (20 mL) was added imidazole (1.31 g, 19.21 mmol) in portions at 0 °C.

This was followed addition of TBSCI (1.45 g, 9.61 mmol) in portions at 0 °C. The resulting mixture was stirred overnight at room temperature and then was quenched with H 2 O (20 mL). The resulting mixture was extracted with EA (3 x 30 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EA = 2:1) to afford 2-amino-3-{l-[(tert-butyldimethylsilyl)oxy]ethyl}-N,5-dimeth ylbenzamide (500 mg, 80%) as an off-white solid. MS: (ES + ) m/z = 323.1 [M+H] + .

Step 4: Preparation of 4-(8-(l-((tert-butyldimethylsilyl)oxy)ethyl)-3,6-dimethyl-4- oxo-l,2,3,4- tetrahydroquinazolin-2-yl)benzonitrile.

[000204] A mixture of 2-amino-3-{l-[(tert-butyldimethylsilyl)oxy]ethyl}-N,5- dimethylbenzamide (1 g, 3.10 mmol), MgSO 4 (1.12 g, 9.30 mmol), p-toluenesulfonic acid (0.16 g, 0.93 mmol) and 4-formylbenzonitrile (0.49 g, 3.72 mmol,) in MeOH (12 mL) was stirred overnight at room temperature. The resulting mixture was quenched with H 2 O (50 mL) and extracted with EtOAc (2 x 100 mL). The combined organic layers were washed with brine (2 x 80 mL), dried over anhydrous Na2SO 4 , and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EA = 1:1) to afford 4-(8-(l- ((tert-butyldimethylsilyl)oxy)ethyl)-3,6-dimethyl-4-oxo-l,2, 3,4-tetrahydroquinazolin-2- yl)benzonitrile (1.1 g, 81%) as an off-white solid. MS: (ES + ) m/z = 436.2 [M+H] + .

Step 5: Preparation of 4-(8-(l-((tert-butyldimethylsilyl)oxy)ethyl)-3,6-dimethyl-4- oxo-3,4- dihydroquinazolin-2-yl)benzonitrile.

[000205] To a stirred solution of 4-(8-(l-((tert-butyldimethylsilyl)oxy)ethyl)-3,6-dimethyl-4- oxo-l,2,3,4-tetrahydroquinazolin-2-yl)benzonitrile (1.1 g, 2.53 mmol) and MnO 2 (3.29 g, 37.88 mmol) in CHCl (15 mL). The resulting mixture was stirred for 3 h at 60 °C, then filtered and the filter cake was washed with ethyl acetate (2 x 20 mL) and DCM (2 x 20 mL). The filtrate was concentrated under reduced pressure to afford 4-(8-(l-((tert- butyldimethylsilyl)oxy)ethyl)-3,6-dimethyl-4-oxo-3,4-dihydro quinazolin-2-yl)benzonitrile (950 mg, 87%) as an off-white solid (used in the next step without further purification). MS: (ES + ) m/z = 434.1 [M+H] + .

Step 6: Preparation of 4-(8-(l-hydroxyethyl)-3,6-dimethyl-4-oxo-3,4-dihydroquinazol in-2- yl)benzonitrile.

[000206] To a stirred solution of 4-(8-(l-((tert-butyldimethylsilyl)oxy)ethyl)-3,6-dimethyl-4- oxo-3, 4-dihydroquinazolin-2-yl)benzonitrile (950 mg, 2.19 mmol) in THF (20 mL) was slowly added TBAF (1 M in THF, 2.15 mL, 2.15 mmol) dropwise at 0 °C. The resulting solution was stirred overnight at room temperature and then poured into ice cold water (50 mL). The resulting solids were collected by filtration and washed with water (5 mL). The filtrate was extracted with EtOAc (3 x 50 mL). The combined organic layers were washed with brine (3 x 50 mL) and washed with saturated aqueous NaHCOs. The organics were dried over Na 2 SO 4 and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EA = 7:3) to afford 4-(8-(l-hydroxyethyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-2-yl)benzonitrile (600 mg, 85%) as an off-white solid. MS: (ES + ) m/z = 320.2 [M+H] + .

Step 7: Preparation of tert-butyl 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate.

[000207] To a solution of 4-(8-(l-hydroxyethyl)-3,6-dimethyl-4-oxo-3,4-dihydroquinazol in- 2-yl)benzonitrile (600 mg, 1.88 mmol), tert-butyl 2-(2,4-dinitrobenzenesulfonamido)benzoate (795 mg, 1.88 mmol) and PPhs (1.23 g, 4.70 mmol) in THF (12 mL) was added a solution of (E)- N-[[(tert-butoxy)carbonyl]imino](tert-butoxy)formamide (1.30 g, 5.64 mmol) in THF (1 mL) at 0 °C under nitrogen atmosphere. The resulting solution was stirred overnight at room temperature under nitrogen atmosphere. The resulting solution was concentrated under reduced pressure and purified by prep TLC (PE/EA = 2:1) to afford tert-butyl 2-((l-(2-(4- cyanophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl)e thyl)amino)benzoate (500 mg, 53%) as a light yellow solid. MS: (ES + ) m/z = 495.4 [M+H] + .

Step 8: Preparation of 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquina zolin-8- yl)ethyl)amino)benzoic acid.

[000208] To a stirred solution of tert-butyl 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo- 3,4-dihydroquinazolin-8-yl)ethyl)amino)benzoate (500 mg, 1.01 mmol) in 4 M HCI in dioxane (12 mL) at room temperature. The resulting solution was stirred overnight at room temperature and then concentrated under reduced pressure. The residue was purified by prep TLC (CH 2 Cl 2 MeOH = 25:1) to afford 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (180 mg, 40%) as a light yellow solid. MS: (ES + ) m/z = 439.1 [M+H] + . Step 9: Preparation of 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquina zolin-8- yl)ethyl)amino)benzoic acid (Enantiomers 1 and 2).

[000209] A racemic mixture of 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (150 mg, 0.34 mmol) was separated by chiral HPLC under the following conditions: Column: CHIRALPAK ID, 2*25 cm, 5 pm; Mobile Phase A: Hex(0.1% TFA)-HPLC, Mobile Phase B: IPA-HPLC; Flow rate: 20 mL/min; Gradient: 25% B to 25% B in 16 min; Wavelengths: 220/254 nm; RTl(min): 10.134; RT2(min): 13.489; Sample Solvent: EtOH-HPLC; Injection Volume: 0.3 mL. This resulted in the isolation of each enantiomer of 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquina zolin-8- yl)ethyl)amino)benzoic acid as a light yellow solid.

[000210] 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquina zolin-8- yl)ethyl)amino)benzoic acid Enantiomer 1 (66.3 mg, 43.9%, 99.9% ee). MS: (ES + ) m/z = 439.2 [M+H] + . 1 H NMR: (400 MHz, DMSO-d6) 6 13.10 (s, 1H), 8.42 (s, 1H), 8.10 - 8.04 (m, 2H), 8.03 - 7.96 (m, 2H), 7.92 - 7.87 (m, 1H), 7.82 - 7.78 (m, 1H), 7.59 (d, J = 2.1 Hz, 1H), 7.16 - 7.15 (m, 1H), 6.50 - 6.49 (m, 1H), 6.44 - 6.38 (m, 1H), 5.45 (m, 1H), 3.39 (s, 3H), 2.40 (s, 3H), 1.54 (d, J = 6.7 Hz, 3H).

[000211] 2-((l-(2-(4-cyanophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroquina zolin-8- yl)ethyl)amino)benzoic acid Enantiomer 2 (55.6 mg, 39.6%, 99.1% ee). MS: (ES + ) m/z = 439.2 [M+H] + . NMR: (400 MHz, DMSO-d6) 6 12.81 (s, 1H), 8.43 (s, 1H), 8.10 - 8.04 (m, 2H), 8.03 - 7.96 (m, 2H), 7.92 - 7.87 (m, 1H), 7.82 - 7.78 (m, 1H), 7.59 (d, J = 2.1 Hz, 1H), 7.16 - 7.15 (m, 1H), 6.54 - 6.46 (m, 1H), 6.44 - 6.38 (m, 1H), 5.45 (m, 1H), 3.39 (s, 3H), 2.40 (s, 3H), 1.53 (d, J = 6.6 Hz, 3H).

[000212] Example 15: 2-((l-(2-(4-chloro-2-fluorophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid

Step 1: Preparation of 8-(l-((tert-butyldimethylsilyl)oxy)ethyl)-2-(4-chloro-2-fluo rophenyl)-3,6- dimethyl-2,3-dihydroquinazolin-4(lH)-one.

[000213] A solution of 2-amino-3-(l-((tert-butyldimethylsilyl)oxy)ethyl)-N,5- dimethylbenzamide (600 mg, 1.86 mmol) in THF (20 mL) was treated with MgSO 4 (660 mg, 5.48 mmol) and p-toluenesulfonic acid (96 mg, 0.56 mmol) at room temperature followed by the addition of 4-chloro-2-fluorobenzaldehyde (380 mg, 2.40 mmol) at room temperature. The resulting mixture was stirred overnight at room temperature. The resulting mixture was quenched with H 2 O (20 mL) and extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 20 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 1:1) to afford 8-(l-((tert-butyldimethylsilyl)oxy)ethyl)-2-(4-chloro-2-fluo rophenyl)-3,6-dimethyl-2,3- dihydroquinazolin-4(lH)-one (765 mg, 88%) as a yellow oil. MS: (ES + ) m/z = 463.2 [M+H] + .

Step 2: Preparation of 2-(4-chloro-2-fluorophenyl)-8-(l-hydroxyethyl)-3,6-dimethylq uinazolin- 4(3H)-one.

[000214] A solution of 8-(l-((tert-butyldimethylsilyl)oxy)ethyl)-2-(4-chloro-2-fluo rophenyl)- 3,6-dimethyl-2,3-dihydroquinazolin-4(lH)-one (650 mg, 1.40 mmol) in MeOH (10 mL) was treated with DDQ (550 mg, 2.42 mmol) at room temperature. The resulting mixture was stirred for 2 h at room temperature. The mixture was then quenched with H 2 O (20 mL) and extracted with EtOAc (3 x 30 mL). The combined organic layers were washed with brine (3 x 30 mL), dried over anhydrous Na 2 SO4, and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EA = 1:1) to afford 2-(4-chloro-2- fluorophenyl)-8-(l-hydroxyethyl)-3,6-dimethylquinazolin-4(3H )-one (240 mg, 49%) as a white solid. MS: (ES + ) m/z = 347.1 [M+H] + .

Step 3: Preparation of 8-(l-bromoethyl)-2-(4-chloro-2-fluorophenyl)-3,6-dimethylqui nazolin-

[000215] A solution of 2-(4-chloro-2-fluorophenyl)-8-(l-hydroxyethyl)-3,6- dimethylquinazolin-4(3H)-one (240 mg, 0.69 mmol,) in CH 2 CI 2 was treated with PBrs (375 mg, 1.38 mmol) at 0 °C. The resulting mixture was stirred overnight at room temperature. The mixture was neutralized to pH 8 with saturated aqueous Na 2 COs. The resulting mixture was extracted with CH 2 CI 2 (3 x 10 mL). The combined organic layers were washed with brine (2 x 10 mL), dried over anhydrous Na 2 SC>4, and then concentrated under reduced pressure to afford 8-(l-bromoethyl)-2-(4-chloro-2-fluorophenyl)-3,6-dimethylqui nazolin-4(3H)-one (253 mg, 89%) as a light brown solid (used in the next step without purification). MS: (ES + ) m/z = 409.0 [M+H] + . Step 4: Preparation of methyl 2-((l-(2-(4-chloro-2-fluorophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate.

[000216] A solution of 8-(l-bromoethyl)-2-(4-chloro-2-fluorophenyl)-3,6- dimethylquinazolin-4(3H)-one (253 mg, 0.62 mmol) in ACN (10 mL) was treated with methyl anthranilate (225 mg, 1.49 mmol) at room temperature. The resulting mixture was stirred overnight at 90 °C and then was cooled to room temperature and concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 5:1) to afford methyl 2-((l- (2-(4-chloro-2-fluorophenyl)-3,6-dimethyl-4-oxo-3,4-dihydroq uinazolin-8- yl)ethyl)amino)benzoate (146 mg, 49%) as an off-white solid. MS: (ES + ) m/z = 480.2 [M+H] + .

Step 5: Preparation of 2-((l-(2-(4-chloro-2-fluorophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid.

[000217] A solution of methyl 2-((l-(2-(4-chloro-2-fluorophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate (130 mg, 0.27 mmol) and NaOH (650 mg, 16.25 mmol) in 10 mL MeOH-H 2 O (10:1) was stirred overnight at 50 °C. The mixture was acidified to pH 6 with aqueous 1 M HCI. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were washed with brine (2 x 10 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by prep TLC (CH 2 CL/ 2 MeOH = 15:1) then purified further by prep HPLC (Column: XBridge Shield RP18 OBD Column, 30*150 mm, 5pm; Mobile Phase A: Water (10 mmol/L NH4HCO3+0.1%NH3.H 2 O), Mobile Phase B: ACN; Flow rate: 60 mL/min; Gradient: 23% B to 53% B in 9 min, 53% B; Wavelength: 254 nm) to afford 2-((l-(2-(4-chloro-2-fluorophenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (12.3 mg, 9.8%) as an off-white solid. MS: (ES + ) m/z = 466.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) 6 8.80 - 8.60 (m, 1H), 7.90 - 7.86 (m, 1H), 7.84 - 7.78 (m, 3H), 7.73 - 7.68 (m, 2H), 7.18 - 7.05 (m, 1H), 6.52 - 6.45 (m, 1H), 6.35 (d, J = 8.5 Hz, 1H), 5.41 (s, 1H), 3.36 (d, J = 1.2 Hz, 3H), 2.40 (s, 3H), 1.51 (d, J = 6.6 Hz, 3H).

[000218] Example 16: 2-((l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000219] Example 17: 2-((l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 2)

Step 1: Preparation of 3-(l-((tert-butyldimethylsilyl)oxy)ethyl)-2-(4,4-dimethylcyc lohexane-l- carboxamido)-N,5-dimethylbenzamide.

[000220] To a stirred solution of 2-amino-3-(l-((tert-butyldimethylsilyl)oxy)ethyl)-N,5- dimethylbenzamide (1 g, 3.10 mmol) and 4,4-dimethylcyclohexane-l-carboxylic acid (483 mg, 3.09 mmol) in ACN (20 mL) was added pyridine (1.23 g, 15.56 mmol) and T3P (7.9 g, 24.83 mmol) dropwise at room temperature under nitrogen atmosphere. The resulting mixture was stirred for overnight at 50 °C under a nitrogen atmosphere. The mixture was quenched with H 2 0 (20 mL) and extracted with EtOAc (2 x 50 mL). The combined organic layers were washed with brine (2 x 20 L), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (CH 2 CI 2 /MeOH = 15:1) to afford 3-(l-((tert-butyldimethylsilyl)oxy)ethyl)-2-(4,4-dimethylcyc lohexane-l- carboxamido)-N,5-dimethylbenzamide (960 mg, 67%) as a white solid. MS: (ES + ) m/z = 461.4 [M+H] + .

Step 2: Preparation of 8-(l-((tert-butyldimethylsilyl)oxy)ethyl)-2-(4,4-dimethylcyc lohexyl)-3,6- dimethylquinazolin-4(3H)-one.

[000221] To a stirred solution of 3-(l-((tert-butyldimethylsilyl)oxy)ethyl)-2-(4,4- dimethylcyclohexane-l-carboxamido)-N,5-dimethylbenzamide (1.02 g, 2.21 mmol) in DMF (15 mL) was added K 2 COs (612 mg, 4.43 mmol) dropwise at room temperature under a nitrogen atmosphere. The resulting mixture was stirred for 4 h at 90 °C. The reaction was quenched with H 2 O (50 mL) and the mixture was extracted with EtOAc (2 x 80 mL). The combined organic layers were washed with brine (2 x 50 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure to yield 8-(l-((tert- butyldimethylsilyl)oxy)ethyl)-2-(4,4-dimethylcyclohexyl)-3,6 -dimethylquinazolin-4(3H)-one (884 mg, 90%) as a light yellow solid. MS: (ES + ) m/z = 443.3 [M+H] + .

Step 3: Preparation of tert-butyl 2-((N-(l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)-2,4-dinitrophenyl)sulfonamido) benzoate.

[000222] Prepared in a manner similar to Steps 6 and 7 in Example 13 to afford tert-butyl 2- ((N-(l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4-oxo-3,4-di hydroquinazolin-8-yl)ethyl)-2,4- dinitrophenyl)sulfonamido)benzoate as a yellow solid. MS: (ES + ) m/z = 734.4 [M+H] + .

Step 4: Preparation of tert-butyl 2-((l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate.

[000223] To a stirred solution of tert-butyl 2-((N-(l-(2-(4,4-dimethylcyclohexyl)-3,6- dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl)ethyl)-2,4-dinitro phenyl)sulfonamido)benzoate (680 mg, 0.91 mmol) in DCM (10 mL) was slowly added Et 3 N (0.39 mL, 2.78 mmol) and 2- sulfanylacetic acid (0.13 mL, 1.44 mmol) by dropwise addition at 0 °C. The resulting solution was stirred for 3 h at rt. The reaction was quenched with H 2 O (20 mL) and extracted with DCM (3 x 60 mL). The combined organic layers were washed with brine (2 x 40 mL), dried over anhydrous Na 2 SO 4 , and then concentrated under reduced pressure. The residue was purified by prep TLC (PE/EA = 1:1) to afford tert-butyl 2-((l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4- oxo-3, 4-dihydroquinazolin-8-yl)ethyl)amino)benzoate (420 mg, 91) as a yellow solid. MS: (ES + ) m/z = 504.4 [M+H] + .

Step 5: Preparation of 2-((l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomers 1 and 2).

[000224] Prepared in a manner similar to Steps 8 and 9 in Example 13 to afford each enantiomer of 2-((l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4-oxo-3,4-dih ydroquinazolin-8- yl)ethyl)amino)benzoic acid as a white solid.

[000225] 2-((l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4-oxo-3,4-dih ydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 1, (99.9% ee). MS: (ES + ) m/z = 448.2 [M+H] + . NMR: (400 MHz, DMSO-d6) 6 12.63 (s, 1H), 8.42 (s, 1H), 7.81 - 7.74 (m, 2H), 7.54 (d, J = 2.2 Hz, 1H), 7.19 - 7.10 (m, 1H), 6.55 - 6.45 (m, 2H), 5.57 (s, 1H), 3.60 (d, J = 3.5 Hz, 3H), 2.94 (d, J = 9.6 Hz, 1H), 2.35 (d, J = 3.9 Hz, 3H), 1.94 - 1.85 (m, 4H), 1.84 - 1.70 (m, 3H), 1.57 - 1.50 (m, 2H), 1.45 - 1.32 (m, 2H), 1.00 - 0.96 (m, 6H).

[000226] 2-((l-(2-(4,4-dimethylcyclohexyl)-3,6-dimethyl-4-oxo-3,4-dih ydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 2, (96.1% ee). MS: (ES + ) m/z = 448.2 [M+H] + . 1 H NMR (400 MHz, DMSO-d6) 6 12.63 (s, 1H), 8.42 (s, 1H), 7.81 - 7.74 (m, 2H), 7.54 (d, J = 2.2 Hz, 1H), 7.19 - 7.10 (m, 1H), 6.55 - 6.45 (m, 2H), 5.57 (s, 1H), 3.60 (d, J = 3.5 Hz, 3H), 2.94 (d, J = 9.6 Hz, 1H), 2.35 (d, J = 3.9 Hz, 3H), 1.94 - 1.85 (m, 4H), 1.84 - 1.70 (m, 3H), 1.57 - 1.50 (m, 2H), 1.45 - 1.32 (m, 2H), 1.00 - 0.96 (m, 6H). [000227] Example 18: 2-((l-(2-(3-(4-fluorophenyl)bicyclo[l.l.l]pentan-l-yl)-3,6-d imethyl-4- oxo-3, 4-dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000228] Example 19: 2-((l-(2-(3-(4-fluorophenyl)bicyclo[l.l.l]pentan-l-yl)-3,6-d imethyl-4- oxo-3, 4-dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 2)

[000229] Prepared in a manner similar to Examples 16 and 17 using 3-(4- fluorophenyl)bicyclo[l.l.l]pentane-l-carboxylic acid in place of 4,4-dimethylcyclohexane-l- carboxylic acid to afford each enantiomer of 2-((l-(2-(3-(4-fluorophenyl)bicyclo[l.l.l]pentan- l-yl)-3,6-dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl)ethyl)am ino)benzoic acid as a white solid.

[000230] 2-((l-(2-(3-(4-fluorophenyl)bicyclo[l.l.l]pentan-l-yl)-3,6-d imethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid Enantiomer 1, 99.9% ee. MS: (ES + ) m/z = 498.1 [M+H] + . 1 H NMR: (300 MHz, CD 3 OD) 6 7.94 - 7.85 (m, 2H), 7.61 (d, J = 2.0 Hz, 1H), 7.40 - 7.31 (m, 2H), 7.20 - 7.01 (m, 3H), 6.56 - 6.45 (m, 2H), 5.70 - 5.55 (m, 1H), 3.84 - 3.77 (m, 3H), 2.75 - 2.61 (m, 6H), 2.41 (s, 3H), 1.67 (d, J = 6.7 Hz, 3H).

[000231] 2-((l-(2-(3-(4-fluorophenyl)bicyclo[l.l.l]pentan-l-yl)-3,6-d imethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid Enantiomer 2, 99.8% ee. MS: (ES + ) m/z = 498.1 [M+H] + . 1H NMR: (300 MHz, CD3OD) 6 7.94 - 7.85 (m, 2H), 7.61 (d, J = 2.0 Hz, 1H), 7.43 - 7.32 (m, 2H), 7.20 - 7.02 (m, 3H), 6.56 - 6.45 (m, 2H), 5.70 - 5.55 (m, 1H), 3.84 - 3.77 (m, 3H), 2.75 - 2.62 (m, 6H), 2.41 (s, 3H), 1.67 (d, J = 6.7 Hz, 3H).

[000232] Example 20: 2-((l-(2-(4-chloro-3-methoxyphenethyl)-3,6-dimethyl-4-oxo-3, 4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000233] Example 21: 2-((l-(2-(4-chloro-3-methoxyphenethyl)-3,6-dimethyl-4-oxo-3, 4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 2)

[000234] Prepared in a manner similar to Examples 16 and 17 using 3-(4-chloro-3- methoxyphenyl)propanoic acid in place of 4,4-dimethylcyclohexane-l-carboxylic acid to afford each enantiomer of 2-((l-(2-(4-chloro-3-methoxyphenethyl)-3,6-dimethyl-4-oxo-3, 4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid as a white solid.

[000235] 2-((l-(2-(4-chloro-3-methoxyphenethyl)-3,6-dimethyl-4-oxo-3, 4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid Enantiomer 1 (>99.9% ee). MS: (ES + ) m/z = 506.0 [M+H] + . 1 H NMR: (400 MHz, DMSO-d6) 6 12.73 (s, 1H), 8.48 (s, 1H), 7.80 - 7.75 (m, 2H), 7.51 (d, J = 2.1 Hz, 1H), 7.29 (d, J = 8.0 Hz, 1H), 7.17 - 7.08 (m, 2H), 6.95 - 6.70 (m, 1H), 6.52 - 6.46 (m, 1H), 6.34 (d, J = 8.5 Hz, 1H), 5.50 - 5.46 (m, 1H), 3.81 (s, 3H), 3.58 (s, 3H), 3.32 - 3.20 (m, 4H), 2.35 (s, 3H), 1.48 (d, J = 6.6 Hz, 3H).

[000236] 2-((l-(2-(4-chloro-3-methoxyphenethyl)-3,6-dimethyl-4-oxo-3, 4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid Enantiomer 2 (96% ee). MS: (ES + ) m/z = 506.0 [M+H] + . 1 H NMR: (400 MHz, DMSO-d6) 6 12.73 (s, 1H), 8.49 (s, 1H), 7.80 - 7.75 (m, 2H), 7.51 (d, J = 2.1 Hz, 1H), 7.29 (d, J = 8.1 Hz, 1H), 7.19 - 7.08 (m, 2H), 6.95 - 6.70 (m, 1H), 6.52 - 6.46 (m, 1H), 6.34 (d, J = 8.5 Hz, 1H), 5.50 - 5.46 (m, 1H), 3.81 (s, 3H), 3.58 (s, 3H), 3.32 - 3.20 (m, 4H), 2.35 (s, 3H), 1.49 (d, J = 6.6 Hz, 3H).

[000237] Example 22: 2-((l-(2-(3-chloro-4-ethoxyphenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 1)

[000238] Example 23: 2-((l-(2-(3-chloro-4-ethoxyphenyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoic acid (Enantiomer 2)

[000239] Prepared in a manner similar to Examples 16 and 17 using 3-chloro-4- ethoxybenzoic acid in place of 4,4-dimethylcyclohexane-l-carboxylic acid to afford each enantiomer of 2-((l-(2-(3-chloro-4-ethoxyphenyl)-3,6-dimethyl-4-oxo-3,4-di hydroquinazolin- 8-yl)ethyl)amino)benzoic acid as a white solid.

[000240] 2-((l-(2-(3-chloro-4-ethoxyphenyl)-3,6-dimethyl-4-oxo-3,4-di hydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 1 (99.9% ee). MS: (ES + ) m/z = 492.2 [M+H] + . NMR: (300 MHz, CD 3 OD) 6 7.95 - 7.82 (m, 2H), 7.77 (d, J = 2.2 Hz, 1H), 7.68 - 7.57 (m, 2H), 7.22 (d, J = 8.6 Hz, 1H), 7.15 - 7.10 (m, 1H), 6.53 - 6.37 (m, 2H), 5.65 - 5.55 (m, 1H), 4.22 (q, J = 7.0 Hz, 2H), 3.55 (s, 3H), 2.40 (s, 3H), 1.59 (d, J = 6.7 Hz, 3H), 1.49 (t, J = 7.0 Hz, 3H).

[000241] 2-((l-(2-(3-chloro-4-ethoxyphenyl)-3,6-dimethyl-4-oxo-3,4-di hydroquinazolin-8- yl)ethyl)amino)benzoic acid Enantiomer 2 (99.9% ee). MS: (ES + ) m/z = 492.2 [M+H] + . 1 H NMR: (300 MHz, CD3OD) 6 7.95 - 7.82 (m, 2H), 7.77 (d, J = 2.2 Hz, 1H), 7.68 - 7.57 (m, 2H), 7.22 (d, J = 8.6 Hz, 1H), 7.15 - 7.10 (m, 1H), 6.53 - 6.37 (m, 2H), 5.65 - 5.55 (m, 1H), 4.22 (q, J = 7.0 Hz, 2H), 3.55 (s, 3H), 2.40 (s, 3H), 1.59 (d, J = 6.7 Hz, 3H), 1.49 (t, J = 7.0 Hz, 3H).

[000242] Example 24: 2-((l-(3,6-dimethyl-4-oxo-2-((lR,5S,6r)-3-(2,2,2-trifluoroet hyl)-3- azabicyclo[3.1.0]hexan-6-yl)-3,4-dihydroquinazolin-8-yl)ethy l)amino)benzoic acid (Enantiomer 1)

[000243] Example 25: 2-((l-(3,6-dimethyl-4-oxo-2-((lR,5S,6r)-3-(2,2,2-trifluoroet hyl)-3- azabicyclo[3.1.0]hexan-6-yl)-3,4-dihydroquinazolin-8-yl)ethy l)amino)benzoic acid (Enantiomer 2)

Step 1: Preparation of tert-butyl (lR,5S,6r)-6-(8-(l-((2-(methoxycarbonyl)phenyl)amino)ethyl)- 3,6-dimethyl-4-oxo-3,4-dihydroquinazolin-2-yl)-3-azabicyclo[ 3.1.0]hexane-3-carboxylate.

[000244] To a stirred solution of tert-butyl (lR,5S,6r)-6-(8-(l-hydroxyethyl)-3,6-dimethyl-4- oxo-3, 4-dihydroquinazolin-2-yl)-3-azabicyclo[3.1.0]hexane-3-carbox ylate (235 mg, 0.59 mmol, prepared in manners similar to Steps 6 and 7 of Example 13 and Steps 1 and 2 of Example 16 using (lR,5S,6r)-3-(tert-butoxycarbonyl)-3-azabicyclo[3.1.0]hexane -6-carboxylic acid in place of 4,4-dimethylcyclohexane-l-carboxylic acid), methyl 2-(2,4- dinitrobenzenesulfonamido)benzoate (269 mg, 0.71 mmol) and PPh 3 (1.23 g, 4.70 mmol) in THF (10 mL) was added a solution of DTAD (1.23 g, 5.88 mmol) in THF (2 mL). The resulting mixture was stirred overnight at room temperature under a nitrogen atmosphere and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (PE/EA = 1:1) to afford tert-butyl (lR,5S,6r)-6-(8-(l-((2- (methoxycarbonyl)phenyl)amino)ethyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-2-yl)-3- azabicyclo[3.1.0]hexane-3-carboxylate (200 mg, 63%) as a white solid. MS: (ES + ) m/z = 533.3 [M+H] + . Step 2: Preparation of methyl 2-((l-(2-((lR,5S,6r)-3-azabicyclo[3.1.0]hexan-6-yl)-3,6-dime thyl- 4-oxo-3,4-dihydroquinazolin-8-yl)ethyl)amino)benzoate.

[000245] A solution of tert-butyl (lR,5S,6r)-6-(8-(l-((2- (methoxycarbonyl)phenyl)amino)ethyl)-3,6-dimethyl-4-oxo-3,4- dihydroquinazolin-2-yl)-3- azabicyclo[3.1.0]hexane-3-carboxylate (190 mg, 0.36 mmol) in DCM (10 mL) and TFA (3 mL) was stirred overnight at room temperature. The reaction was then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (CH 2 Cl 2 MeOH = 10:1) to afford methyl 2-((l-(2-((lR,5S,6r)-3-azabicyclo[3.1.0]hexan-6-yl)-3,6- dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl)ethyl)amino)benzoa te (125 mg, 81%) as a white solid. MS: (ES + ) m/z = 433.3 [M+H] + .

Step 3: Preparation of methyl 2-((l-(3,6-dimethyl-4-oxo-2-((lR,5S,6r)-3-(2,2,2-trifluoroet hyl)-3- azabicyclo[3.1.0]hexan-6-yl)-3,4-dihydroquinazolin-8-yl)ethy l)amino)benzoate.

[000246] To a stirred solution of methyl 2-((l-(2-((lR,5S,6r)-3-azabicyclo[3.1.0]hexan-6-yl)- 3,6-dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl)ethyl)amino)be nzoate (125 mg, 0.28 mmol) in ACN (5 mL) was added K 2 CO 3 (230 mg, 1.66 mmol) and 2,2,2-trifluoroethyl trifluoromethanesulfonate (77 mg, 0.33 mmol). The resulting mixture was stirred overnight at room temperature under a nitrogen atmosphere. The resulting mixture was quenched with H 2 O (10 mL) and extracted with EA (3 x 50 mL). The combined organic layers were washed with brine (50 mL), dried over anhydrous Na 2 SO 4 , and then was concentrated under reduced pressure. The residue was purified by silica gel column chromatography

(PE/EA = 1:1) to afford methyl 2-((l-(3,6-dimethyl-4-oxo-2-((lR,5S,6r)-3-(2,2,2-trifluoroet hyl)-3- azabicyclo[3.1.0]hexan-6-yl)-3,4-dihydroquinazolin-8-yl)ethy l)amino)benzoate (120 mg, 80%) as a white solid. MS: (ES + ) m/z = 515.2 [M+H] + .

Step 4: Preparation of 2-((l-(3,6-dimethyl-4-oxo-2-((lR,5S,6r)-3-(2,2,2-trifluoroet hyl)-3- azabicyclo[3.1.0]hexan-6-yl)-3,4-dihydroquinazolin-8-yl)ethy l)amino)benzoic acid (Enantiomers 1 and 2).

[000247] Prepared in a manner similar to step 2 of Examples 3 and 4 using methyl 2-((l-(3,6- dimethyl-4-oxo-2-((lR,5S,6r)-3-(2,2,2-trifluoroethyl)-3-azab icyclo[3.1.0]hexan-6-yl)-3,4- dihydroquinazolin-8-yl)ethyl)amino)benzoate in place of methyl 2-((l-(2-isopentyl-3,6- dimethyl-4-oxo-3,4-dihydroquinazolin-8-yl)ethyl)amino)benzoa te to yield each enantiomer of 2-((l-(3,6-dimethyl-4-oxo-2-((lR,5S,6r)-3-(2,2,2-trifluoroet hyl)-3-azabicyclo[3.1.0]hexan-6-yl)- 3,4-dihydroquinazolin-8-yl)ethyl)amino)benzoic acid as white solids

[000248] 2-((l-(3,6-dimethyl-4-oxo-2-((lR,5S,6r)-3-(2,2,2-trifluoroet hyl)-3- azabicyclo[3.1.0]hexan-6-yl)-3,4-dihydroquinazolin-8-yl)ethy l)amino)benzoic acid Enantiomer 1 (99.9% ee). MS: (ES + ) m/z = 501.0 [M+H] + . 1 H NMR: (300 MHz, CD 3 OD) 6 7.88 - 7.82 (m, 2H), 7.53 (s, 1H), 7.11 - 7.05 (m, 1H), 6.50 - 6.45 (m, 1H), 6.37 (d, J = 8.4 Hz, 1H), 5.55 - 5.49 (m, 1H), 3.76 (s, 3H), 3.37 - 3.30 (m, 2H), 3.29 - 3.21 (m, 2H), 2.83 (d, J = 7.2 Hz, 2H), 2.55 (s, 1H), 2.35 (s, 5H), 1.57 (d, J = 6.6 Hz, 3H).

[000249] 2-((l-(3,6-dimethyl-4-oxo-2-((lR,5S,6r)-3-(2,2,2-trifluoroet hyl)-3- azabicyclo[3.1.0]hexan-6-yl)-3,4-dihydroquinazolin-8-yl)ethy l)amino)benzoic acid Enantiomer 2 (99.9% ee). MS: (ES + ) m/z = 501.0 [M+H] + . 1 H NMR: (300 MHz, CD 3 OD) 6 7.88-7.82 (m, 2H), 7.53 (s, 1H), 7.11 - 7.05 (m, 1H), 6.50 - 6.45 (m, 1H), 6.36 (d, J = 8.4 Hz, 1H), 5.55 - 5.49 (m, 1H), 3.76 (s, 3H), 3.37 - 3.30 (m, 2H), 3.29 - 3.21 (m, 2H), 2.83 (d, J = 7.2 Hz, 2H), 2.55 (s, 1H), 2.35 (s, 5H), 1.57 (d, J= 6.6 Hz, 3H).

[000250] Examples 26 to 377 were prepared in a manner similar to that described in Schemes 1 to 3 or Examples 1 to 25 and are identified and characterized in Table 1 below. If not specified otherwise, all depicted chiral centers exist as a (R)- and (S)-racemic mixture or as either (R)- or (S)- enantiomer. Table 1. Examples 26 to 377

Assays and Compound Testing

[000251] In vitro cell proliferation: determination of EC50 values for inhibition of proliferation in T47D cells expressing mutant PI3Ka (H1047R) mutation and SKBR3 cells express WT PI3Ka.

[000252] T47D or SKBR3 cells were trypsinized, resuspended in culture media and seeded onto assay ready plates. T47D culture media consisted of RPMI, 10% FBS and insulin (0.2 units/mL). SKBR3 culture media consisted of McCoys 5a and 10% FBS. Cells were seeded at a density of 1,500 cells/well and dispensed in 50 pL onto 384 well assay ready plates (Corning, 89089-790). Assay ready plates had previously been stamped with 10-point dilutions of compounds of interest, as well as controls. The Echo655 is used to stamp plates at 40 nL of compound or DMSO. Cells were grown for 72 hours at 37 ° Celsius and 5% CO 2 . After 72 hours, cells were equilibrated at room temperature for 15 minutes. 30 uL of CellTiter-Glo reagent is added to the plate, which is then shaken for 30 minutes at temperature at 300-500 rpm. Cells are then read on an Envision plate reader. The percentage of inhibition of proliferation was calculated using the following formula: %ln hibition = 100 x (LurmiD - Lumsampie) / (LurriD -Luminh), where D is obtained from cells treated with 0.1% DMSO only; Inh is obtained from cells treated with lOuM Alpelisib. The effective concentration achieving 50% inhibition of proliferation (EC50) is calculated by fitting the Curve using Xlfit (v5.3.1.3), equation 201: Y = Bottom + (Top - Bottom)/(1 + 10 A ((LogEC50 - X)*HillSlope)).

Reagent table:

[000253] In vitro cell pAKT: determination of IC50 values for inhibition of phosphorylation of AKT (pAKT) in T47D cells expressing mutant PI3Ka (H1047R) mutation and SKBR3 cells express WT PI3Ka.

[000254] T47D or SKBR3 cells were trypsinized, resuspended in culture media and seeded onto assay ready plates. T47D culture media consisted of RPMI, 10% FBS and insulin (0.2 units/mL). SKBR3 culture media consisted of McCoy's 5a and 10% FBS. Cells were seeded at a density of 5000 cells/well and dispensed in 12.5 pL onto 384 well assay ready plates (Perkin Elmer, 6008238)). Assay ready plates had previously been stamped with 10-point dilutions of compounds of interest, as well as controls. The Echo655 is used to stamp plates at 12.5 nL of compound or DMSO. Cells were grown for 6 hour at 37 ° Celsius and 5% CO2. After 6 hour, 4 pL of Lysis buffer reagent was added to the plate, which was then centrifuged for 1 minute at 1000 rpm. Then the plate was incubated at room temperature for 30 minutes. After 30 minutes, 4 pL of antibody mix containing Eu cryptate, d2 cryptate, and detection buffer, was added to the plate. The plate was centrifuged for 1 minute at 1000 rpm and then incubated overnight at room temperature. The plate was read on an Envision plate reader using the HTRF protocol. The percentage of inhibition of AKT phosphorylation was calculated using the following formula: %l nhibition = 100 x (pAKTHC - pAKTSample) / (pAKTHC -pAKTLC)), where pAKTHC is obtained from cells treated with 0.1% DMSO only; pAKTLC is obtained from cells treated with lOuM Alpelisib. The IC50 (concentration achieving 50% inhibition of pAKT) is calculated by fitting the Curve using Xlfit (v5.3.1.3), equation 201: Y = Bottom + (Top - Bottom)/(1 + 10 A ((LoglC50 - X)*HillSlope)).

Reagent table: [000255] For EC50 values shown in Table 2, "A" refers to 1 nM < EC50 < 500 nM; "B" refers to 500 nM < EC50 < 2 pM; "C" refers to 2 pM < EC50 < 15 pM; and "D" refers to an EC50 > 15 pM.

Table 2. Cellular proliferation data

[000256] For IC50 values shown in Table 3 "A" refers to 1 nM < IC50 < 500 nM; "B" refers to

500 nM< IC50 < 2 pM; "C" refers to 2 pM < IC50 < 15 pM; and "D" refers to an IC50 > 15 pM.

Table 3. Cellular pAKT data

[000257] CD1 mice were dosed with a single IV or PO dose, followed by serial sampling of plasma at 0.0833 (IV only), 0.25, 0.5, 1, 2, 4, 8, 24 hours post dose. Desired serial concentrations of working solutions were achieved by diluting stock solution of analyte with 50% acetonitrile in water solution. Ten microliters of working solutions (0.5, 1, 2, 5, 10, 50, 100, 500, 1000 ng/mL) were added to 10 pL of the blank Female CD1 Mouse plasma to achieve calibration standards of 0.5 to approximately 1000 ng/mL (0.5, 1, 2, 5, 10, 50, 100, 500, 1000 ng/mL) in a total volume of 20 pL. Five quality control samples at 1 ng/mL, 2 ng/mL, 5 ng/mL, 50 ng/mL and 800 ng/mL for plasma were prepared independently of those used for the calibration curves. These QC samples were prepared on the day of analysis in the same way as calibration standards. 20 pL standards, 20 pL QC samples and 20 pL unknown samples (10 pL plasma with 10 pL blank solution) were added to 200 pL of acetonitrile containing IS mixture for precipitating protein respectively. Then the samples were vortexed for 30 s. after centrifugation at 4 ° Celsius, 4000 rpm for 15 min. The supernatant was diluted with water at a ratio of 1:2 (V/V, 1:2), Then 5 pL of diluted supernatant was injected into the LC/MS/MS system for quantitative analysis. The results are shown in Table 4.

Table 4: mouse pharmacokinetic data

References

[000258] Ali, K., Bilancio, A., Thomas, M., Pearce, W., Gilfillan, A. M., Tkaczyk, C., Kuehn, N., Gray, A., Giddings, J., Peskett, E., Fox, R., Bruce, I., Walker, C., Sawyer, C., Okkenhaug, K., Finan, P., and Vanhaesebroeck, B. 2004. Essential role for the pllOdelta phosphoinositide 3-kinase in the allergic response. Nature 431(7011): 1007-1011.

[000259] Backer, J. M. 2016. The intricate regulation and complex functions of the Class III phosphoinositide 3-kinase Vps34. Biochem J. 473(15): 2251-2271.

[000260] Castel, P., Toska, E., Engelman, J. A., and Scaltriti, M. 2021. The present and future of PI3K inhibitors for cancer therapy. Nat Cancer 2(6): 587-597.

[000261] Fruman, D. A., Chiu H., Hopkins B. D., Bagrodia, S., Cantley, L. C., and Abraham R. T. 2017. The PI3K pathway in human disease. Cell 170(4): 605-635.

[000262] Gadkar, K., Friedrich, C., Hurez, V., Ruiz, M. L., Dickmann, L., Jolly, M. K., Schutt, L., Jin, J., Ware, J. A., and Ramanujan, S. 2021. Quantitative systems pharmacology model-based investigation of adverse gastrointestinal events associated with prolonged treatment with PI3- kinase inhibitors. CPT Pharmacometrics Syst Pharmacol. 11(5): 616-627.

[000263] Goncalves, M. D., Hopkins, B. D., and Cantley, L. C. 2018. Phosphatidylinositol 3- Kinase, growth disorders, and cancer. N Engl J Med. 379(21): 2052-2062. [000264] Hang, H.C. and Bertozzi, C. R. 2001. Chemoselective approaches to glycoproteon assembly. Accounts of Chemical Research 34(9): 727-736.

[000265] Hawkins, P. T. and Stephens, L. R. 2015. PI3K signalling in inflammation. 2015. Biochimica et Biophysica Acta 1851(6): 882-897.

[000266] Hopkins, B. D., Pauli, C., Du, X., Wang, D. G., Li, X., Wu, D., Amadiume, S. C., Goncalves, M. D., Hodakoski, C., Lundquist, M. R., Bareja, R., Ma, Y., Harris, E. M., Sboner, A., Beltran, H., Rubin, M. A., Mukherjee, S., and Cantley, L. C. 2018. Suppression of insulin feedback enhances the efficacy of PI3K inhibitors. Nature 560(7719): 499-503.

[000267] Jackson, S. P., Schoenwaelder, S. M., Goncalves, L, Nesbitt, W. S., Yap, C. L., Wright, C. E., Kenche, V., Anderson, K. E., Dopheide, S. M., Yuan, Y., Sturgeon, S. A., Prabaharan, H., Thompson, P. E., Smith, G. D., Shepherd, P. R., Daniele, N., Kulkarni, S., Abbott, B., Saylik, D., Jones, C., Lu, L., Giuliano, S., Hughan, S. C., Angus, J. A., Robertson, A. D., and Salem, H. H.

2005. PI 3-kinase pllObeta: a new target for antithrombotic therapy. Nat Med. 11(5): 507-514.

[000268] Jiang, N., Dai, Q.., Su, X., Fu, J., and Feng, X. 2020. Role of PI3K/AKT pathway in cancer: the framework of malignant behavior. Mol Biol Rep. 47(6): 4587-4629.

[000269] Keppler-Noreuil, K. M., Rios, J. J., Parker, V. E., Semple, R. K., Lindhurst, M. J.; Sapp, J. C., Alomari, A., Ezaki, M., Dobyns, W., and Biesecker, L. G. 2015. PIK3CA-related overgrowth spectrum (PROS): diagnostic and testing eligibility criteria, differential diagnosis, and evaluation. Am J Med Genet A. 167A(2): 287-295.

[000270] Kiick, K. L., Saxon, E., Tirrell, D. A., and Bertozzi, C. R. 2002. Incorporation of azides into recombinant proteins for chemoselective modification by the Staudinger ligation. Proc Natl Acad Sci USA. 99(1): 19-24.

[000271] Klaus, S., Neumann, H., Zapf, A., Strubing, D., Hubner, S., Almena, J., Riermeier, T., GroR, P., Sarich, M., Krahnert, W., Rossen, K., and Beller, M. 2006. A general and efficient method for the formylation of aryl and heteroaryl bromides. Ang Chem Int Ed. 45(1): 154-158.

[000272] Kurek, K. C„ Luks, V. L, Ayturk, U. M., Alomari, A. I., Fishman, S. J., Spencer S. A., Mulliken, J. B., Bowen, M. E., Yamamoto, G. L., Kozakewich, H. P., and Warman, M. L. 2012.

Somatic mosaic activating mutations in PIK3CA cause CLOVES syndrome. Am J Hum Genet. 90(6): 1108-1115. [000273] Lemieux, G. A. and Bertozzi, C. R. 1998. Chemoselective ligation reactions with proteins, oligosaccharides and cells. Trends in Biotechnology 16(12): 506-513.

[000274] Liu, P., Cheng, H., Roberts, T. M., and Zhao, J. J. 2009. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 8(8): 627-644.

[000275] Okkenhaug, K., Bilancio, A., Farjot, G., Priddle, H., Sancho, S., Peskett, E., Pearce, W., Meek, S. E., Salpekar, A., Waterfield, M. D., Smith, A. J., and Vanhaesebroeck, B. 2002. Impaired B and T cell antigen receptor signaling in pllOdelta PI 3-kinase mutant mice. Science 297(5583): 1031-1034.

[000276] Porta, C., Paglino, C., and Mosca, A. 2014. Targeting PI3K/Akt/mTOR signaling in cancer. Front Oncol. 4(64): 1-11.

[000277] Posor, Y., Eichhorn-Grunig, M., and Haucke, V. 2015. Phosphoinositides in endocytosis. Biochim Biophys Acta 1851(6): 794-804.

[000278] Prakash, G., Krishnamurti, R., and Olah, G. 1989. Synthetic methods and reactions. 141. Fluoride-induced trifluoromethylation of carbonyl compounds with trifluormethyltrimethylsilane (TMS-CF 3 ). A trifluoromethide equivalent. J Am Chem Soc. 111(1): 393-395.

[000279] Reichel, M. and Karaghiosoff, K. 2020. Reagents for selective fluoromethylation: A challenge in organofluorine chemistry. Ang Chem Int Ed. 59(30): 12268-12281.

[000280] Rugo, H. S., Lacouture, M. E., Goncalves, M. D., Masharani, U., Aapro, M. S., and O'Shaughnessy, J. A. 2022. A multidisciplinary approach to optimizing care of patients treated with alpelisib. The Breast 61: 156-167.

[000281] Soler, A., Serra, FL, Pearce, W., Angulo, A., Guillermet-Guibert, J., Friedman, L. S., Vinals, F., Gerhardt, FL, Casanovas, O., Graupera, M., and Vanhaesebroeck, B. 2004. Inhibition of the pllOa isoform of PI 3-kinase stimulates nonfunctional tumor angiogenesis. J Exp Med. 210(10): 1937-1945.

[000282] Thorpe, L. M., Yuzugullu, FL, and Zhao, J. J. 2015. PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting. Nat Rev Cancer 15(1): 7-24.

[000283] Vanhaesebroeck, B., Whitehead M. A., and Pineiro, R. 2016. Molecules in medicine mini-review: isoforms of PI3K in biology and disease. J Mol Med (Berl). 94(1): 5-11. [000284] Yang, J., Nie, J., Ma, X., Wei, Y., Peng, Y., and Wei, X. 2019. Targeting PI3K in cancer: mechanisms and advances in clinical trials. Mol Cancer 18(26): 1-28.

[000285] Zhao, Y., Huang, W., Zheng, J., and Hu, J. 2011. Efficient and direct nucleophilic difluoromethylation of carbonyl compounds and imines with Me 3 SiCF 2 H at ambient or low temperature. Org. Lett. 13(19): 5342-5345.