Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MODULATORS OF TREX1
Document Type and Number:
WIPO Patent Application WO/2023/137030
Kind Code:
A1
Abstract:
Provided are compounds of Formula (I): and pharmaceutically acceptable salts and compositions thereof, which are useful for treating a variety of conditions associated with TREX1.

Inventors:
COFFIN AARON (US)
Application Number:
PCT/US2023/010541
Publication Date:
July 20, 2023
Filing Date:
January 11, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CONSTELLATION PHARMACEUTICALS INC (US)
International Classes:
C07D239/557; A61K31/506; A61P35/00; C07D403/06; C07D417/14
Domestic Patent References:
WO2020118133A12020-06-11
WO2021222761A12021-11-04
WO2021016317A12021-01-28
Other References:
FRONT. CELL INFECT. MICROBIOL., vol. 2, 2012, pages 168
NATURE, vol. 478, 2011, pages 515 - 518
CELL REP., vol. 3, 2013, pages 1355 - 1361
SCIENCE, vol. 341, 2013, pages 903 - 906
PNAS, vol. 112, 2015, pages 5117 - 5122
J. EXP. MED., vol. 208, 2011, pages 2005 - 2016
EMBO REP., vol. 16, 2015, pages 202 - 212
NAT. REV IMMUNOL., vol. 15, 2015, pages 405 - 414
TRENDS IN CELL BIOL., vol. 27, no. 8, 2017, pages 543 - 4
NATURE COMMUN., vol. 8, 2017, pages 15618
BIOCHIM. BIOPHYS. ACTA, vol. 1833, 2013, pages 1832 - 43
GLICKMAN ET AL.: "Annual Meeting of Society for Immunotherapy of Cancer", 7 November 2018
ANTIMICROB. AGENTS CHEMOTHER., vol. 59, 2015, pages 1273 - 1281
NATURE IMMUNOLOGY, vol. 11, no. 11, 2010, pages 1005
MAZUR DJPERRINO FW: "Identification and expression of the TREX1 and TREX2 cDNA sequences encoding mammalian 3'-->5' exonucleases", J BIOL CHEM., vol. 274, no. 28, August 1999 (1999-08-01), pages 19655 - 60
HOSS MROBINS PNAVEN TJPAPPIN DJSGOUROS JLINDAHL T: "A human DNA editing enzyme homologous to the Escherichia coli DnaQ/MutD protein", EMBO J., vol. 18, no. 13, August 1999 (1999-08-01), pages 3868 - 75
Attorney, Agent or Firm:
DEGRAZIA, Michael, J. et al. (US)
Download PDF:
Claims:
Claims:

1. A compound having the Formula I: or a pharmaceutically acceptable salt thereof, wherein:

R1 is hydrogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, or 3- to 4-membered cycloalkyl;

R2 is hydrogen or (Ci-C4)alkyl optionally substituted with phenyl, wherein said phenyl is optionally substituted with 1 to 3 groups selected from halo, (Ci-C4)alkyl, and halo(Ci-C4)alkyl;

Ring A and Ring B are each independently aryl, heteroaryl, heterocyclyl, or cycloalkyl;

R3, R4’ and R6 are each independently (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, (Ci- C6)alkylORb, (C2-C6)alkenyl, halo(Ci-C6)alkoxy, halo, phenyl, -CN, -NRaC(O)ORb, - NRaC(S)ORb, -C(O)Rb,-NRaC(O)NRbRs, -NRaC(S)NRbRs, -NRaS(O)2NRbRg, -C(S)Rb, - S(O)2RC, -S(O)RC, -C(O)ORd, -C(S)ORd, -C(O)NReRf, -C(S)NRaRe, -NRaC(O)Rd, - NRaC(S)Rd, -ORe, -SRe, -O(Ci-C4)alkylORe, -NReRf, 4- to 6-membered heteroaryl, or 4- to 7- membered heterocyclyl, wherein i) said phenyl for R3 and R4 are each independently optionally substituted with 1 or 2 groups selected from Rg; ii) said (Ci-Ce)alkyl for R3 and R4 are each independently optionally substituted with 1 or 2 groups selected from ORh, -NR'Rk, phenyl, and 5- to 6- membered heteroaryl; and ii) said 4- to 7-membered heterocyclyl and 4- to 6-membered heteroaryl for R3 and R4 are each independently optionally substituted with 1 or 2 groups selected from Rm; and iv) said phenyl and 5- to 6-membered heteroaryl of the optional substituents listed for (Ci-Ce)alkyl in R3 and R4 are each independently optionally substituted with 1 or 2 groups selected from Rg; x is 0, 1, or 2; m and n are each independently an integer from 0 to 3;

R5 is (Ci-C6)alkyl, halo(Ci-C6)alkyl, (C2-C6)alkenyl, -(Ci-C6)alkylORa, -(Ci- Ce)alkylNRaRb, -(Ci-C6)alkyl(C3-C7)cycloalkyl, -(Ci-Ce)alkyl(4- to 7-membered heterocyclyl), -(Ci-Ce)alkyl(5- to 7-membered heteroaryl), phenyl, 5- to 7-membered heteroaryl, (C3-C?)cycloalkyl, (C3-C?)cycloalkenyl, or 4- to 7-membered heterocyclyl, wherein each occurrence of said phenyl, 5- to 7-membered heteroaryl, (C3-C?)cycloalkyl, (C3-C?)cycloalkenyl, or 4- to 7-membered heterocyclyl are optionally and independently substituted with 1 to 3 groups selected from R6, provided R5 is not an optionally substituted isoxazolyl;

Ra, Rb, Rc, Rd, Re, and Rf are each independently hydrogen, halo, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, phenyl, 3- to 4-membered cycloalkyl, 4- to 6-membered heteroaryl, or 4- to 7-membered heterocyclyl, wherein i) said (Ci-Ce)alkyl for Ra, Rb, Rc, Rd, Re and Rf is optionally substituted with 1 or 2 groups selected from phenyl, -ORh, and -NR'Rk; ii) said phenyl, 4- to 6-membered heteroaryl, and 4- to 7-membered heterocyclyl for Ra, Rb, Rc, Rd, Re, and Rf are each optionally and independently substituted with 1 or 2 groups selected from Rg; iii) said 4- to 7-membered heterocyclyl for Ra, Rb, Rc, Rd, Re, and Rf is further optionally substituted with =0; and

Rg, Rh, R1, Rk, and Rm are each independently hydrogen, halo, (Ci-Ce)alkyl, halo(Ci- Ce)alkyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, phenyl, -(Ci-Ce)alkylphenyl, 3- to 4-membered cycloalkyl, 4- to 6-membered heteroaryl, or 4- to 7-membered heterocyclyl, and wherein said 4- to 7-membered heterocyclyl for Rg, Rh, R1 and Rkis further optionally substituted with =0.

2. The compound of Claim 1, or a pharmaceutically acceptable salt thereof, wherein R1 (Ci-C4)alkyl.

3. The compound of Claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein R1 is methyl.

4. The compound of any one of Claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R2 is (Ci-C4)alkyl.

5. The compound of any one of Claims 1 to 4, or a pharmaceutically acceptable salt thereof, wherein R2 is methyl.

6. The compound of any one of Claims 1 to 5, or a pharmaceutically acceptable salt thereof, wherein x is 0.

7. The compound of any one of Claim 1 to 6, or a pharmaceutically acceptable salt thereof, wherein R5 is phenyl or 5- to 7-membered heteroaryl, each optionally substituted with 1 to 3 groups selected from R6.

8. The compound of any one of Claim 1 to 7, or a pharmaceutically acceptable salt thereof, wherein R5 is phenyl or isothiazolyl, each optionally substituted with 1 to 3 groups selected from R6.

9. The compound of any one of Claim 1 to 8, or a pharmaceutically acceptable salt thereof, wherein Ring A is phenyl or 5- to 7-membered heteroaryl.

10. The compound of any one of Claim 1 to 9, or a pharmaceutically acceptable salt thereof, wherein Ring A is phenyl.

11. The compound of any one of Claim 1 to 9, or a pharmaceutically acceptable salt thereof, wherein Ring A is isothiazolyl.

12. The compound of any one of Claim 1 to 11, or a pharmaceutically acceptable salt thereof, wherein Ring B is phenyl or 5- to 7-membered heteroaryl.

13. The compound of any one of Claim 1 to 12, or a pharmaceutically acceptable salt thereof, wherein Ring B is phenyl, pyrazinyl, or pyrazolyl.

14. The compound of any one of Claim 1 to 13, or a pharmaceutically acceptable salt thereof, wherein Ring B is phenyl or pyrazolyl.

15. The compound of any one of Claim 1 to 14, or a pharmaceutically acceptable salt thereof, wherein Ring B is phenyl.

16. The compound of any one of Claim 1 to 14, or a pharmaceutically acceptable salt thereof, wherein Ring B is pyrazolyl.

17. The compound of any one of Claim 1 to 16, or a pharmaceutically acceptable salt thereof, wherein R6 is halo.

18. The compound of any one of Claim 1 to 17, or a pharmaceutically acceptable salt thereof, wherein m is 1 or 2.

19. The compound of any one of Claim 1 to 18, or a pharmaceutically acceptable salt thereof, wherein m is 1.

20. The compound of any one of Claim 1 to 19, or a pharmaceutically acceptable salt thereof, wherein R3 is -CN, -C(O)NReRf, or halo.

21. The compound of any one of Claim 1 to 20, or a pharmaceutically acceptable salt thereof, wherein R3 is -CN, -C(O)N[(Ci-C4)alkyl]2, or halo.

22. The compound of any one of Claim 1 to 21, or a pharmaceutically acceptable salt thereof, wherein R3 is -CN.

23. The compound of any one of Claim 1 to 22, or a pharmaceutically acceptable salt thereof, wherein n is 0, 1, or 2.

24. The compound of any one of Claim 1 to 23, or a pharmaceutically acceptable salt thereof, wherein n is 0 or 1.

25. The compound of any one of Claim 1 to 24, or a pharmaceutically acceptable salt thereof, wherein n is 0.

26. The compound of any one of Claim 1 to 24, or a pharmaceutically acceptable salt thereof, wherein n is 1.

27. The compound of any one of Claim 1 to 26, or a pharmaceutically acceptable salt thereof, wherein R4 is (Ci-C4)alkyl, halo, or (Ci-C4)alkylO(Ci-C4)alkyl.

28. The compound of Claim 1, wherein the compound is selected from

or a pharmaceutically acceptable salt of any of the foregoing.

29. A pharmaceutical composition comprising the compound of any one of Claims 1 to 28, or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier.

30. A method of treating a disease responsive to the inhibition of TREX1 in a subject, comprising administering to the subject, a therapeutically effective amount of a compound of any one of Claims 1 to 28, or a pharmaceutically acceptable salt thereof, or the composition of Claim 29.

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

Description:
MODULATORS OF TREX1

RELATED APPLICATIONS

[0001] This application claims the benefit of priority to U.S. Provisional Application No. 63/298,317, filed January 11, 2022, the entire contents of which are incorporated herein by reference.

BACKGROUND

[0002] A potential immune therapy is needed for cancers related to the innate immune system recognition of non-self, and to detect and protect against potential danger. Cancer cells differ antigenically from their normal counterparts and emit danger signals to alert the immune system similar to viral infection. These signals, which include damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs), further activate the innate immune system resulting in the protection of the host from a variety of threats (Front. Cell Infect. Microbiol. 2012, 2, 168).

[0003] Ectopically expressed single stranded DNA (ssDNA) and double stranded DNA (dsDNA) are known PAMPs and/or DAMPs, which are being recognized by the cyclic GMP- AMP synthase (cGAS), a nucleic acid sensor (Nature 2011, 478, 515-518). Upon sensing of cytosolic DNA, cGAS catalyzes the generation of the cyclic dinucleotide 2’,3’-cGAMP, a potent second messenger and activator of the ER transmembrane adapter protein stimulator of interferon genes (STING) (Cell Rep. 2013, 3, 1355-1361). STING activation triggers phosphorylation of IRF3 via TBK1 which in turn leads to type I interferon production and activation of interferon stimulated genes (ISGs); a pre-requisite to the activation of innate immunity and initiation of adaptive immunity. Production of type I interferons thus constitutes a key bridge between the innate and adaptive immunity (Science 2013, 347, 903- 906).

[0004] Excess type I IFN can be harmful to the host and induce autoimmunity, therefore, negative feedback mechanisms exist that keep type I IFN-mediated immune activation in check. Three prime repair exonuclease I (TREX1) is a 3’-5’ DNA exonuclease responsible for the removal of ectopically expressed ssDNA and dsDNA and is therefore a key repressor of the cGAS/STING pathway (TWAS' 2015, 112, 5117-5122).

[0005] Type I interferons and downstream pro-inflammatory cytokine responses are critical to the development of immune responses and their effectiveness. Type I interferons enhance both the ability of dendritic cells and macrophages to take up, process, present, and cross-present antigens to T cells, and their potency to stimulate T cells by eliciting the upregulation of the co-stimulatory molecules such as CD40, CD80 and CD86 (J. Exp. Med. 2011, 208, 2005-2016). Type I interferons also bind their own receptors and activate interferon responsive genes that contribute to activation of cells involved in adaptive immunity (EMBO Rep. 2015, 16, 202-212).

[0006] From a therapeutic perspective, type I interferons and compounds that can induce type I interferon production have potential for use in the treatment of human cancers (Nat. Rev Immunol. 2015, 15, 405-414). Interferons can inhibit human tumor cell proliferation directly. In addition, type I interferons can enhance anti-tumor immunity by triggering the activation of cells from both the innate and adaptive immune system. Importantly, the antitumor activity of PD-1 blockade requires pre-existing intratumoral T cells. By turning cold tumors into hot and thereby eliciting a spontaneous anti-tumor immunity, type I IFN-inducing therapies have the potential to expand the pool of patients responding to anti -PD-1 therapy as well as enhance the effectiveness of anti-PDl therapy.

[0007] Human and mouse genetic studies suggest that TREX1 inhibition might be amenable to a systemic delivery route and therefore TREX1 inhibitory compounds could play an important role in the anti-tumor therapy landscape. TREX1 is a key determinant for the limited immunogenicity of cancer cells responding to radiation treatment Trends in Cell Biol., 2017, 27 (8), 543-4; Nature Commun., 2017, 8, 15618], TREX1 is induced by genotoxic stress and involved in protection of glioma and melanoma cells to anticancer drugs \Biochim. Biophys. Acta, 2013, 1833, 1832-43], STACT-TREX1 therapy shows robust antitumor efficacy in multiple murine cancer models [Glickman et al, Poster P235, 33 rd Annual Meeting of Society for Immunotherapy of Cancer, Washington DC, Nov. 7-11, 2018], (TREX1) expression correlates with cervical cancer cells growth in vitro and disease progression in vivo [Scientific Reports 1019, 9, 351], Beyond oncology there is also support for agonists of the IFN pathway to be useful in antiviral therapy, for example STING agonists induce an innate antiviral immune response against Hepatitis B Virus via stimulation of the IFN pathway and upregulation of ISG’s [Antimicrob. Agents Chemother. 2015, 59: 1273- 1281] and TREX1 inhibits the innate immune response to HIV type 1 [Nature Immunology, 2010, 11(11), 1005], SUMMARY

[0008] Provided herein are compounds having the Formula I: and pharmaceutically acceptable salts and compositions thereof, wherein R 1 , R 2 , R 3 , R 4 , R 5 , x, m, and n are as described herein. The disclosed compounds and compositions modulate TREX1, and are useful in a variety of therapeutic applications such as, for example, in treating cancer.

DETAILED DESCRIPTION

1. General Description of Compounds

[0009] In a first embodiment, provided herein is a compound of Formula I: or a pharmaceutically acceptable salt thereof, wherein:

R 1 is hydrogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, or 3- to 4-membered cycloalkyl;

R 2 is hydrogen or (Ci-C4)alkyl optionally substituted with phenyl, wherein said phenyl is optionally substituted with 1 to 3 groups selected from halo, (Ci-C4)alkyl, and halo(Ci-C4)alkyl;

Ring A and Ring B are each independently is aryl, heteroaryl, heterocyclyl, or cycloalkyl;

R 3 , R 4 ’ and R 6 are each independently (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, (Ci- C 6 )alkylOR b , (C 2 -C 6 )alkenyl, halo(Ci-C 6 )alkoxy, halo, phenyl, -CN, -NR a C(O)OR b , - NR a C(S)OR b , -C(O)R b ,-NR a C(O)NR b R s , -NR a C(S)NR b R s , -NR a S(O) 2 NR b R s , -C(S)R b , - S(O) 2 R C , -S(O)R C , -C(O)OR d , -C(S)OR d , -C(O)NR e R f , -C(S)NR a R e , -NR a C(O)R d , - NR a C(S)R d , -OR e , -SR e , -O(Ci-C4)alkylOR e , -NR e R f , 4- to 6-membered heteroaryl, or 4- to 7- membered heterocyclyl, wherein i) said phenyl for R 3 and R 4 are each independently optionally substituted with 1 or 2 groups selected from R g ; ii) said (Ci-Ce)alkyl for R 3 and R 4 are each independently optionally substituted with 1 or 2 groups selected from OR h , -NR J R k , phenyl, and 5- to 6- membered heteroaryl; and ii) said 4- to 7-membered heterocyclyl and 4- to 6-membered heteroaryl for R 3 and R 4 are each independently optionally substituted with 1 or 2 groups selected from R m ; and iv) said phenyl and 5- to 6-membered heteroaryl of the optional substituents listed for (Ci-Ce)alkyl in R 3 and R 4 are each independently optionally substituted with 1 or 2 groups selected from R g ; x is 0, 1, or 2; m and n are each independently an integer from 0 to 3;

R 5 is (Ci-C 6 )alkyl, halo(Ci-C 6 )alkyl, (C 2 -C 6 )alkenyl, -(Ci-C 6 )alkylOR a , -(Ci- Ce)alkylNR a R b , -(Ci-C6)alkyl(C3-C7)cycloalkyl, -(Ci-Ce)alkyl(4- to 7-membered heterocyclyl), -(Ci-Ce)alkyl(5- to 7-membered heteroaryl), phenyl, 5- to 7-membered heteroaryl, (C3-C?)cycloalkyl, (C3-C?)cycloalkenyl, or 4- to 7-membered heterocyclyl, wherein each occurrence of said phenyl, 5- to 7-membered heteroaryl, (C3-C?)cycloalkyl, (C3-C?)cycloalkenyl, or 4- to 7-membered heterocyclyl are optionally and independently substituted with 1 to 3 groups selected from R 6 , provided R 5 is not an optionally substituted isoxazolyl;

R a , R b , R c , R d , R e and R f are each independently hydrogen, halo, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, phenyl, 3- to 4-membered cycloalkyl, 4- to 6-membered heteroaryl, or 4- to 7-membered heterocyclyl, wherein i) said (Ci-Ce)alkyl for R a , R b , R c , R d , R e and R f is optionally substituted with 1 or 2 groups selected from phenyl, -OR h , and -NR J R k ; ii) said phenyl, 4- to 6-membered heteroaryl, and 4- to 7-membered heterocyclyl for R a , R b , R c , R d , R e , and R f are each optionally and independently substituted with 1 or 2 groups selected from R g ; and iii) said 4- to 7-membered heterocyclyl for R a , R b , R c , R d , R e , and R f is further optionally substituted with =0; and R g , R h , R 1 , R k , and R m are each independently hydrogen, halo, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, phenyl, -(Ci-Ce)alkylphenyl, 3- to 4-membered cycloalkyl, 4- to 6-membered heteroaryl, or 4- to 7-membered heterocyclyl, and wherein said 4- to 7-membered heterocyclyl for R g , R h , R 1 and R k is further optionally substituted with =0.

2. Definitions

[0010] When used in connection to describe a chemical group that may have multiple points of attachment, a hyphen (-) designates the point of attachment of that group to the variable to which it is defined. For example, -NHC(0)0R a and -NHC(S)OR a mean that the point of attachment for this group occurs on the nitrogen atom.

[0011] The terms “halo” and “halogen” refer to an atom selected from fluorine (fluoro, -F), chlorine (chloro, -Cl), bromine (bromo, -Br), and iodine (iodo, -I).

[0012] The term “alkyl” when used alone or as part of a larger moiety, such as “haloalkyl”, and the like, means saturated straight-chain or branched monovalent hydrocarbon radical.

[0013] The term “alkenyl” when used alone or as part of a larger moiety, means straightchain or branched monovalent hydrocarbon radical having 1 or 2 double bonds.

[0014] “Alkoxy” means an alkyl radical attached through an oxygen linking atom, represented by -O-alkyl. For example, “(Ci-C4)alkoxy” includes methoxy, ethoxy, proproxy, and butoxy.

[0015] The term “haloalkyl” includes mono, poly, and perhaloalkyl groups where the halogens are independently selected from fluorine, chlorine, bromine, and iodine.

[0016] “Haloalkoxy” is a haloalkyl group which is attached to another moiety via an oxygen atom such as, e.g., -OCHF2 or -OCFs.

[0017] The term “aryl” refers to an aromatic carbocyclic ring system having, unless otherwise specified, a total of 6 to 10 ring members. In certain embodiments, “aryl” refers to an aromatic ring system which includes, but is not limited to, phenyl and naphthyl. It will be understood that when specified, optional substituents on an aryl group may be present on any substitutable position and, include, e.g., the position at which the aryl is attached.

[0018] The term “heteroaryl” used alone or as part of a larger moiety refers to a 5- to 12- membered (e.g., a 4- to 6-membered) aromatic radical containing 1-4 heteroatoms selected from N, O, and S. A heteroaryl group may be mono- or bi-cyclic as size permits. Monocyclic heteroaryl includes, for example, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, triazinyl, tetrazinyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, etc. Bi-cyclic heteroaryls include groups in which a monocyclic heteroaryl ring is fused to one or more aryl or heteroaryl rings. Nonlimiting examples include indolyl, imidazopyridinyl, benzooxazolyl, benzooxodiazolyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, quinazolinyl, quinoxalinyl, pyrrolopyridinyl, pyrrolopyrimidinyl, pyrazolopyridinyl, thienopyridinyl, thienopyrimidinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. It will be understood that when specified, optional substituents on a heteroaryl group may be present on any substitutable position and, include, e.g., the position at which the heteroaryl is attached.

[0019] The term “cycloalkyl” refers to a saturated cyclic hydrocarbon having from, unless otherwise specified, 3 to 10 carbon ring atoms. Monocyclic cycloalkyl groups include, without limitation, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl. It will be understood that when specified, optional substituents on a cycloalkyl or may be present on any substitutable position and, include, e.g., the position at which the cycloalkyl is attached.

[0020] The term “cycloalkenyl”, refers to a partially saturated cyclic hydrocarbon having from, unless otherwise specified, from 3 to 10 carbon atoms. Cycloalkenyl groups include, without limitation, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl and cycloheptenyl. It will be understood that when specified, optional substituents on a cycloalkenyl group may be present on any substitutable position and, include, e.g., the position at which the cycloalkenyl group is attached.

[0021] The disclosed compounds exist in various stereoisomeric forms. Stereoisomers are compounds that differ only in their spatial arrangement. Enantiomers are pairs of stereoisomers whose mirror images are not superimposable, most commonly because they contain an asymmetrically substituted carbon atom that acts as a chiral center. “Enantiomer” means one of a pair of molecules that are mirror images of each other and are not superimposable. Diastereomers are stereoisomers that contain two or more asymmetrically substituted carbon atoms. “R” and “S” represent the configuration of substituents around one or more chiral carbon atoms.

[0022] “Racemate” or “racemic mixture” means a compound of equimolar quantities of two enantiomers, wherein such mixtures exhibit no optical activity, z.e., they do not rotate the plane of polarized light.

[0023] When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight pure relative to all of the other stereoisomers. Percent by weight pure relative to all of the other stereoisomers is the ratio of the weight of one stereoisomer over the weight of the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least 60%, 70%, 80%, 90%, 99% or 99.9% by weight optically pure. Percent optical purity by weight is the ratio of the weight of the enantiomer over the weight of the enantiomer plus the weight of its optical isomer.

[0024] When a disclosed compound is named or depicted by structure without indicating the stereochemistry, and the compound has one chiral center, it is to be understood that the name or structure encompasses one enantiomer of compound free from the corresponding optical isomer, a racemic mixture of the compound, or mixtures enriched in one enantiomer relative to its corresponding optical isomer.

[0025] The term “TREX1” refers to three prime repair exonuclease 1 or DNA repair exonuclease 1, which is an enzyme that in humans is encoded by the TREX1 gene. Mazur DJ, Perrino FW (Aug 1999). "Identification and expression of the TREX1 and TREX2 cDNA sequences encoding mammalian 3'— >5' exonucleases". J Biol Chem. 274 (28): 19655-60. doi: 10.1074/jbc.274.28.19655. PMID 10391904; Hoss M, Robins P, Naven TJ, Pappin DJ, Sgouros J, Lindahl T (Aug 1999). "A human DNA editing enzyme homologous to the Escherichia coli DnaQ/MutD protein". EMBO J. 18 (13): 3868-75. doi: 10.1093/emboj/18.13.3868. PMC 1171463. PMID 10393201. This gene encodes the major 3'->5' DNA exonuclease in human cells. The protein is a non-processive exonuclease that may serve a proofreading function for a human DNA polymerase. It is also a component of the SET complex, and acts to rapidly degrade 3' ends of nicked DNA during granzyme A- mediated cell death. Cells lacking functional TREX1 show chronic DNA damage checkpoint activation and extra-nuclear accumulation of an endogenous single-strand DNA substrate. It appears that TREX1 protein normally acts on a single- stranded DNA polynucleotide species generated from processing aberrant replication intermediates. This action of TREX1 attenuates DNA damage checkpoint signaling and prevents pathological immune activation. TREX1 metabolizes reverse-transcribed single-stranded DNA of endogenous retroelements as a function of cell-intrinsic antiviral surveillance, resulting in a potent type I IFN response. TREX1 helps HIV-1 to evade cytosolic sensing by degrading viral cDNA in the cytoplasm. [0026] The term “TREX2” refers to Three prime repair exonuclease 2 is an enzyme that in humans is encoded by the TREX2 gene. This gene encodes a nuclear protein with 3' to 5' exonuclease activity. The encoded protein participates in double-stranded DNA break repair, and may interact with DNA polymerase delta. Enzymes with this activity are involved in DNA replication, repair, and recombination. TREX2 is a 3 '-exonuclease which is predominantly expressed in keratinocytes and contributes to the epidermal response to UVB- induced DNA damage. TREX2 biochemical and structural properties are similar to TREX1, although they are not identical. The two proteins share a dimeric structure and can process ssDNA and dsDNA substrates in vitro with almost identical k ca t values. However, several features related to enzyme kinetics, structural domains, and subcellular distribution distinguish TREX2 from TREX1. TREX2 present a 10-fold lower affinity for DNA substrates in vitro compared with TREX1. In contrast with TREX1, TREX2 lacks a COOH- terminal domain that can mediate protein-protein interactions. TREX2 is localized in both the cytoplasm and nucleus , whereas TREX1 is found in the endoplasmic reticulum, and is mobilized to the nucleus during granzyme A-mediated cell death or after DNA damage. [0027] The terms “subject” and “patient” may be used interchangeably, and means a mammal in need of treatment, e.g., companion animals (e.g., dogs, cats, and the like), farm animals (e.g., cows, pigs, horses, sheep, goats and the like) and laboratory animals (e.g., rats, mice, guinea pigs and the like). Typically, the subject is a human in need of treatment.

[0028] The term “inhibit,” “inhibition” or “inhibiting” includes a decrease in the baseline activity of a biological activity or process.

[0029] As used herein, the terms “treatment,” “treat,” and “treating” refer to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein. In some aspects, treatment may be administered after one or more symptoms have developed, /.< ., therapeutic treatment. In other aspects, treatment may be administered in the absence of symptoms. For example, treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of exposure to a particular organism, or other susceptibility factors), z.e., prophylactic treatment. Treatment may also be continued after symptoms have resolved, for example to delay their recurrence.

[0030] The term “pharmaceutically acceptable carrier” refers to a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions described herein include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, polyethylene glycol and wool fat.

[0031] For use in medicines, the salts of the compounds described herein refer to nontoxic “pharmaceutically acceptable salts.” Pharmaceutically acceptable salt forms include pharmaceutically acceptable acidic/anionic or basic/cationic salts. Suitable pharmaceutically acceptable acid addition salts of the compounds described herein include e.g., salts of inorganic acids (such as hydrochloric acid, hydrobromic, phosphoric, nitric, and sulfuric acids) and of organic acids (such as, acetic acid, benzenesulfonic, benzoic, methanesulfonic, and p-toluenesulfonic acids). Compounds of the present teachings with acidic groups such as carboxylic acids can form pharmaceutically acceptable salts with pharmaceutically acceptable base(s). Suitable pharmaceutically acceptable basic salts include e.g., ammonium salts, alkali metal salts (such as sodium and potassium salts) and alkaline earth metal salts (such as magnesium and calcium salts). Compounds with a quaternary ammonium group also contain a counteranion such as chloride, bromide, iodide, acetate, perchlorate and the like. Other examples of such salts include hydrochlorides, hydrobromides, sulfates, methanesulfonates, nitrates, benzoates and salts with amino acids such as glutamic acid.

[0032] The term “effective amount” or “therapeutically effective amount” refers to an amount of a compound described herein that will elicit a desired or beneficial biological or medical response of a subject e.g., a dosage of between 0.01 - 100 mg/kg body weight/day.

3. Compounds

[0033] In a second embodiment, R 1 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is (Ci-C4)alkyl, wherein the remaining variables are as described above for Formula I. Alternatively, as part of a second embodiment, R 1 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is methyl, wherein the remaining variables are as described above for Formula I.

[0034] In a third embodiment, R 2 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is (Ci-C4)alkyl, wherein the remaining variables are as described above for Formula I or the second embodiment. Alternatively, as part of a second embodiment, R 2 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is methyl, wherein the remaining variables are as described above for Formula I or the second embodiment.

[0035] In a fourth embodiment, x in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is 0, wherein the remaining variables are as described above for Formula I or the second embodiment or third embodiment. [0036] In a fifth embodiment, R 5 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl or 5- to 7-membered heteroaryl, each optionally substituted with 1 to 3 groups selected from R 6 , wherein the remaining variables are as described above for Formula I or any one of the second through fourth embodiments. Alternatively, as part of a fifth embodiment, R 5 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl or isothiazolyl, each optionally substituted with 1 to 3 groups selected from R 6 , wherein the remaining variables are as described above for Formula I or any one of the second through fourth embodiments. In another alternative, as part of a fifth embodiment, R 5 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl optionally substituted with 1 to 3 groups selected from R 6 , wherein the remaining variables are as described above for Formula I or any one of the second through fourth embodiments. In another alternative, as part of a fifth embodiment, R 5 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is isothiazolyl optionally substituted with 1 to 3 groups selected from R 6 , wherein the remaining variables are as described above for Formula I or any one of the second through fourth embodiments.

[0037] In a sixth embodiment, Ring A in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl or 5- to 7-membered heteroaryl, wherein the remaining variables are as described above for Formula I or any one of the second through fifth embodiments. Alternatively, as part of a sixth embodiment, Ring A in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl or isothiazolyl, wherein the remaining variables are as described above for Formula I or any one of the second through fifth embodiments. In another alternative, as part of a sixth embodiment, Ring A in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is isothiazolyl, wherein the remaining variables are as described above for Formula I or any one of the second through fifth embodiments. In another alternative, as part of a sixth embodiment, Ring A in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl, wherein the remaining variables are as described above for Formula I or any one of the second through fifth embodiments.

[0038] In a seventh embodiment, Ring B in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl or 5- to 7-membered heteroaryl, wherein the remaining variables are as described above for Formula I or any one of the second through sixth embodiments. Alternatively, as part of a seventh embodiment, Ring B in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl, pyrazinyl, or pyrazolyl, wherein the remaining variables are as described above for Formula I or any one of the second through sixth embodiments. In another alternative, as part of a seventh embodiment, Ring B in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl or pyrazolyl, wherein the remaining variables are as described above for Formula I or any one of the second through sixth embodiments. In another alternative, as part of a seventh embodiment, Ring B in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is phenyl, wherein the remaining variables are as described above for Formula I or any one of the second through sixth embodiments. In another alternative, as part of a seventh embodiment, Ring B in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is pyrazolyl, wherein the remaining variables are as described above for Formula I or any one of the second through sixth embodiments. In another alternative, as part of a seventh embodiment, Ring B in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is pyrazinyl, wherein the remaining variables are as described above for Formula I or any one of the second through sixth embodiments.

[0039] In an eighth embodiment, R 6 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is halo, wherein the remaining variables are as described above for Formula I or any one of the second through seventh embodiments.

[0040] In an ninth embodiment, m in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is 1 or 2, wherein the remaining variables are as described above for Formula I or any one of the second through eighth embodiments. Alternatively, as part of a ninth embodiment, m in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is 1, wherein the remaining variables are as described above for Formula I or any one of the second through eighth embodiments. In another alternative, as part of a ninth embodiment, m in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is 2, wherein the remaining variables are as described above for Formula I or any one of the second through eighth embodiments.

[0041] In a tenth embodiment, R 3 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is -CN, -C(O)NR e R f , or halo, wherein the remaining variables are as described above for Formula I or any one of the second through ninth embodiments. Alternatively, as part of a tenth embodiment, R 3 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is -CN, -C(O)N[(Ci-C4)alkyl]2, or halo, wherein the remaining variables are as described above for Formula I or any one of the second through ninth embodiments. In another alternative, as part of a tenth embodiment, R 3 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is -CN, wherein the remaining variables are as described above for Formula I or any one of the second through ninth embodiments.

[0042] In an eleventh embodiment, n in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is 0, 1, or 2, wherein the remaining variables are as described above for Formula I or any one of the second through tenth embodiments. Alternatively, as part of an eleventh embodiment, n in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is 0 or 1, wherein the remaining variables are as described above for Formula I or any one of the second through tenth embodiments. In another alternative, as part of an eleventh embodiment, n in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is 0, wherein the remaining variables are as described above for Formula I or any one of the second through tenth embodiments. In another alternative, as part of an eleventh embodiment, n in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is 1, wherein the remaining variables are as described above for Formula I or any one of the second through tenth embodiments.

[0043] In a twelfth embodiment, R 4 in the compound of Formula I, or a pharmaceutically acceptable salt thereof, is (Ci-C4)alkyl, halo, or (Ci-C4)alkylO(Ci-C4)alkyl, wherein the v remaining ariables are as described above for Formula I or any one of the second through eleventh embodiments.

[0044] In one aspect of the compounds described herein, included any one of the disclosed embodiments, R 5 is not an optionally substituted isoxazol-4-yl.

[0045] Compounds having the Formula I are further disclosed in the Exemplification and are included in the present disclosure. Pharmaceutically acceptable salts thereof as well as the neutral forms are included.

4. Uses, Formulation and Administration

[0046] Compounds and compositions described herein are generally useful for modulating the activity of TREX1. In some aspects, the compounds and pharmaceutical compositions described herein inhibit the activity TREX1.

[0047] In some aspects, compounds and pharmaceutical compositions described herein are useful in treating a disorder associated with TREX1 function. Thus, provided herein are methods of treating a disorder associated with TREX1 function, comprising administering to a subject in need thereof, a therapeutically effective amount of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a disclosed compound or pharmaceutically acceptable salt thereof. Also provided is the use of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a disclosed compound or pharmaceutically acceptable salt thereof, for the manufacture of a medicament for treating a disorder associated with TREX1 function. Also provided is a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising a disclosed compound or pharmaceutically acceptable salt thereof, for use in treating a disorder associated with TREX1.

[0048] In some aspects, the compounds and pharmaceutical compositions described herein are useful in treating cancer.

[0049] In some aspects, the cancer treated by the compounds and pharmaceutical compositions described herein is selected from colon cancer, gastric cancer, thyroid cancer, lung cancer, leukemia, pancreatic cancer, melanoma, multiple melanoma, brain cancer, CNS cancer, renal cancer, prostate cancer, ovarian cancer, leukemia, and breast cancer.

[0050] In some aspects, the cancer treated by the compounds and pharmaceutical compositions described herein is selected from lung cancer, breast cancer, pancreatic cancer, colorectal cancer, and melanoma.

[0051] In certain aspects, a pharmaceutical composition described herein is formulated for administration to a patient in need of such composition. Pharmaceutical compositions described herein may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrastemal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. In some embodiments, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the pharmaceutical compositions described herein may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.

[0052] In some aspects, the pharmaceutical compositions are administered orally.

[0053] A specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound described herein in the composition will also depend upon the particular compound in the pharmaceutical composition. EXEMPLIFICATION

Chemical Synthesis

[0054] The representative examples that follow are intended to help illustrate the present disclosure, and are not intended to, nor should they be construed to, limit the scope of the invention.

[0055] General starting materials used were obtained from commercial sources or prepared in other examples, unless otherwise noted.

[0056] The following abbreviations have the indicated meanings: ACN Acetonitrile;

AIBN Azobisisobutyronitrile; BOC /-butyl oxycarbonyl; CS2CO3 Cesium

Carbonate; DEA Diethylamine;DCE 1,2-di chloroethane; DCM or

CH2Q2 Dichloromethane; DIPEA or DIEA N,N-diisoproylethylamine, also known as Hunig’s base; DMA N,N-dimethylacetamide; DMAP 4- (dimethylamino)pyridine; DMF N,N-dimethylformamide; DME 1,2- dimethoxyethane; DMSO Dimethylsulfoxide; EtOAc Ethyl acetate; EtOH Ethanol; FA Formic Acid; HATU(9-(7-Azabenzotriazol-l-yl)-N, N, N, N- tetramethyluroniumhexafluorophosphate; HC1 Hydrochloric acid; IPA Isopropylamine; K2CO3 Potassium Carbonate; LiBr Lithium Bromide; LiCl Lithium Chloride; LiHMDS or LHMDS Lithium hexamethyldisilazane; LiOH Lithium hydroxide; Mel or CH3I Methyl Iodide; MeOH Methanol;Na2CO3 Sodium

Carbonate; Na2SO4 Sodium Sulphate; NaH Sodium hydride; NaOH Sodium Hydroxide; NBS A-Bromosuccinimide; NH4CI Ammonium Chloride; T3P 2,4,6-Tripropyl-l,3,5,2,4,6- trioxatriphosphinane 2,4,6- trioxide; TEA Triethylamine; TFA Trifluoroacetic acid; TFAA Trifluoroacetic anhydride; THF Tetrahydrofuran; TMSC1 Trimethylsilyl chloride.

[0057] The progress of reactions was often monitored by TLC or LC-MS. The LC-MS was recorded using one of the following methods. LCMS METHOD-1:

LCMS Method-2:

LCMS Method-3:

LCMS Method-4:

LCMS Method-5: LCMS Method-6:

[0058] NMR was recorded at room temperature unless noted otherwise on Varian Inova 400 or 500 MHz spectrometers with the solvent peak used as the reference or on Bruker 300 or 400 MHz spectrometers with the TMS peak used as internal reference.

[0059] The compounds described herein may be prepared using the following methods and schemes. Unless specified otherwise, all starting materials used are commercially available.

[0060] General Procedure A: (PH-CON-487SR)

[0061] General Procedure B:

Step 1 Step 2

NBS, AIBN, Blue light

DCE, 80 °C

Step 3

[0062] Step 1: 2-ethyl-5-hvdroxy-6-oxo-l H- yrimidine-4-carboxylate

[0063] To a solution of A-hydroxypropanamidine (100 g, 1.13 mol) in xylene (1.5 L) was added diethyl but-2-ynedioate (182.2 mL, 1.13 mol) slowly. The mixture was stirred at 25°C for 12 hr at which point consumption of starting material was observed by TLC. This mixture was then heated to 160 °C and stirred for 10 hr at which point complete conversion to the desired product was observed by TLC. The mixture was cooled to room temperature and filtered; the resulting solid was washed with pet. ether (300 mL) then collected. The crude product was triturated with MTBE (400 mL) and the product was collected as a white sold (98 g, 425 mmol, 37% yield). 'H NMR (400MHz, CDC1 3 ) 8 = 12.64 (br s, 1H), 10.85 (br s, 1H), 4.52 (q, J= 7.1 Hz, 2H), 2.76 (q, J= 7.6 Hz, 2H), 1.47 (t, J =7.1 Hz, 3H), 1.35 (t, J= 7.6 Hz, 3H)

[0064] Step 2: ethyl 2-ethyl-5-methoxy-l-methyl-6-oxo-pyrimidine-4-carboxylate

[0065] To a solution of ethyl 2-ethyl-5-hydroxy-6-oxo-l//-pyrimidine-4-carboxylate (98 g, 465 mmol) in DMF (1 L) was added CH3I (115 mL, 1.85 mol) and K2CO3 (191g, 1.39 mol). The mixture was heated to 55 °C and stirred for 2 hr then poured into H2O (10 L) and extracted with EtOAc (1.5 L x 4). The combined organic layers were washed with 4% aq. LiCL L (1.5 L x 2), dried over TsfeSCU, filtered and concentrated in vacuo giving the crude product. The residue was triturated with MTBE (50 mL) to give the final product as a white solid (109 g). LCMS (ESI) m/z 241.1 [M+H] +1 H NMR (400MHz, CDCh) 6 = 4.42 (q, J= 7.1 Hz, 2H), 3.97 (s, 3H), 3.57 (s, 3H), 2.79 (q, J= 7.4 Hz, 2H), 1.40 (t, J= 7.2 Hz, 3H), 1.37 - 1.32 (m,3H)

[0066] Step 3: ethyl 2-(l-bromoethyl)-5-methoxy-l-methyl-6-oxopyrimidine-4- carboxylate

[0067] To a stirred solution of ethyl 2-ethyl-5-methoxy-l-methyl-6-oxopyrimidine-4- carboxylate (13.0 g, 54.1 mmol) in DCE (130 mL) was added NBS (19.2 g, 108.2 mmol) followed by 2,2’-Azobis(2-methylpropionitrile) (2.6 g, 16.2 mmol). The resulting mixture was heated to 80 °C and exposed to blue LED light, after 1 h complete conversion to the desired brominated product was observed by LCMS. The reaction was then cooled to rt diluted with 1 N HC1 and the resulting mixture was extracted with CH2CI2 (3 x 100 mL). The organic layer was collected, washed with brine, dried over Na2SO4, filtered and concentrated in vacuo. The crude reaction material was purified by silica gel chromatography (1 : 1 EtOAc/Pet. Ether), fractions containing product were collected and the solvent was removed in vacuo to afford the product as a brown oil (13 g, 69% yield). ESI-MS m/z: Calc’d 318.0 found 319.1 [M+H] + [0068] General Procedure C:

Step 1

[0069] Example 1

[0070] Synthesis of 2-(l-(2-cyanophenyl)-l-phenylpropan-2-yl)-5 -hydroxy- 1 -methyl -6- oxo-N-phenyl-l,6-dihydropyrimidine-4-carboxamide p [0071] Step 1: 2-[bromo(phenyl)methyl]benzonitrile

[0072] A 40-mL sealed tube was charged with 2-benzylbenzonitrile (1.70 g, 8.8 mmol), 1 -bromopyrrolidine-2, 5-dione (2.39 g, 13.2 mmol), 2,2-azobisisobutyronitrile (433.4 mg, 2.6 mmol) and DCE (20 mL). The resulting solution was heated to 80 °C for 1 hour at which point it was cooled to rt and concentrated in vacuo. The resulting crude material was purified by silica gel chromatography (EtOAc/pet. ether 1 :40), fractions containing product were collected and concentrated in vacuo resulting in an orange oil (1.7g, 71% yield)._ESI-MS m/z: Cak’d 271.0 found 272.1 [M+H] + H NMR (300 MHz, DMSO-t/ 6 ) 8 7.89 (dt, J= 7.7, 1.0 Hz, 1H), 7.89 - 7.69 (m, 2H), 7.63 - 7.47 (m, 3H), 7.53 - 7.36 (m, 2H), 7.42 - 7.30 (m, 1H), 6.78 (s, 1H)

[0073] Step 2: bromo[(2-cvanophenyl)(phenyl)methyl]zinc

[0074] A round-bottom flask was charged with zinc dust (0.828 g, 12.6 mmol) followed by DMA (5 mL). The resulting mixture was stirred for 15 min at 60 °C at which point a solution of dibromoethane (0.352 g, 1.9 mmol) and chlorotrimethylsilane (0.102 g, 0.9 mmol) in DMA (2 mL) was added dropwise over 15 min. After stirring for 20 min the reaction was cooled to rt and (2-[bromo(phenyl)methyl]benzonitrile) (1.70 g, 6.25 mmol) in DMA (3 mL) was added dropwise. After stirring for an additional 1 h the resulting solution was used directly in step 3.

[0075] Step 3: ethyl 2-[l-(2-cvanophenyl)-l-phenylpropan-2-yl]-5-methoxy-l- methyl-6-oxopyrimidine-4-carboxylate

[0076] To a stirred solution of ethyl 2-(l-bromoethyl)-5 -methoxy- 1 -methyl -6- oxopyrimidine-4-carboxylate (0.500 g, 1.6 mmol) in DMA (5 mL) was added a solution of bromo[(2-cyanophenyl)(phenyl)methyl]zinc, directly from the previous step, in DMA (10 mL) dropwise at 0 °C. The resulting solution was stirred at rt for 2 h at which point it was quenched by the addition of water. The resulting solution was extracted with EtOAc (3x20 mL), the combined organic layers were washed with NaCl then dried over ISfeSCU and concentrated in vacuo. The resulting crude material was purified by reverse phase Chromatography (1 : 1 MeCN/ftO) giving the product as a white solid (0.460 g, 68% yield) ESI-MS m/z: Cak’d 431.2 found 432.2 [M+H] +

[0077] Step 4: 2-[l-(2-cvanophenyl)-l-phenylpropan-2-yl]-5-methoxy-l-methyl -6- oxopyrimidine-4-carboxylate

[0078] To a solution of ethyl 2-[l-(2-cyanophenyl)-l-phenylpropan-2-yl]-5-methoxy-l- methyl-6-oxopyrimidine-4-carboxylate (0.460 g, 1.07 mmol) dissolved in THF (6 mL) and water (4 mL) was added LiOH H2O (0.089 g, 2.11 mmol) portion wise. The resulting mixture was stirred at rt for 2 h at which point it was concentrated in vacuo giving the desired product as a yellow solid (0.460 g) which was used in subsequent steps with no further purification. ESI-MS m/z: Calc’d 403.2 found 404.2 [M+H] +

[0079] Step 5: 2-(l-(2-cyanoDhenyl)-l-DhenylDroDan-2-yl)-5-methoxy-l-methyl -6- oxo-N-phenyl-l.,6-dihydroDyrimidine-4-carboxamide

[0080] To a stirred solution of 2-[l-(2-cyanophenyl)-l-phenylpropan-2-yl]-5-methoxy-l- methyl-6-oxopyrimidine-4-carboxylic acid (0.550 g, 1.36 mmol) and HATU

(0.778 g, 2.05 mmol) in DMF (5 mL) was added aniline (0.190 g, 2.05 mmol) followed by DIPEA (0.441 g, 3.41 mmol). The resulting mixture was stirred at rt for Ih at which point it was diluted with water (100 mL) and the product was extracted with EtOAc (3 x 50 mL). The organic layers were collected and combined then washed with brine, dried over Na2SO4, and concentrated in vacuo. The resulting crude material was purified by reverse phase C18 chromatography. ESI-MS m/z: Calc’d 478.2 found 479.2 [M+H] +

[0081] Separation of diastereomers was done at this step using reverse phase C18 chromatography: Column Xselect CSH F-Phenyl, 5 um, 19*250 mm; 60% to 70% Acetonitrile/water (0.1% FA) in 10 min; Flow rate: 25 mL/min.

[0082] Peak 1_D1 contained 383 mg of a white solid.

[0083] Peak 2_D2 Contained 103 mg of a white solid.

[0084] Enantiomers of this material were separated by Prep-chiral -HPLC:

[0085] DI: Column: Lux 5um Cellulose-2, 2.12*25cm, 5pm; Mobile Phase A: Hex:MtBE=l : 1(0.5% 2M NH3-MEOH), Mobile Phase B:EtOH— HPLC; Flow rate: 20 mL/min; Gradient: 35% B to 35% B in 39 min

[0086] Peak 1 (Isomer-I DIEI): RT 2.64 min; afforded a white solid (145 mg)

[0087] Peak 2 (Isomer-2_D1E2): RT 26.17 min; afforded a white solid (149 mg)

[0088] D2: Column: Lux 5um Cellulose-2, 2.12*25cm, 5um; Hex:MTBE=l : 1(0.5% 2M

NH3-MEOH), Mobile Phase B:IPA— HPLC; Flow rate:20 mL/min; Gradient: 40% B to 40% B in 22 min).

[0089] Peak 1 (Isomer-3_D2E1): RT 9.04 min; afforded a white solid (39 mg)

[0090] Peak 2 (Isomer-4_D2E2): RT 16.9 min; afforded a white solid (38 mg)

[0091] Step 6: 2-(l-(2-cvanoDhenyl)-l-DhenylDroDan-2-yl)-5-hvdroxy-l-methyl -6- oxo-N-phenyl-l.,6-dihvdroDyrimidine-4-carboxamide:

[0092] To a solution of 2-[l-(2-cyanophenyl)-l-phenylpropan-2-yl]-5-methoxy-l-methyl -

6-oxo-N-phenylpyrimidine-4-carboxamide (0.145 g, 0.33 mmol) dissolved in DMF

(5 ml) was added LiBr (0.395 g, 4.55 mmol). This resulting mixture was heated to 95 °C and stirred for 4h at which point complete conversion to the product was observed by LCMS. The reaction was then cooled to rt and concentrated in vacuo. The resulting crude material was purified by reverse phase Chromatography.

[0093] Isomer-I DIEI : Isolated product as a white solid (0.060g, 42% yield)

[0094] ESI-MS m/z: Cak’d 464.2 found 465.1 [M+H] + ; >98% ee

[0095] 1 H NMR (400 MHz, DMSO-t/ 6 ): 6 11.58 (s, 1H), 9.97 (s, 1H), 7.91 (d, 1H), 7.88 -

7.79 (m, 2H), 7.73 - 7.65 (m, 2H), 7.65 - 7.55 (m, 2H), 7.50 - 7.34 (m, 4H), 7.31 - 7.17 (m, 3H), 5.17 (d, 1H), 4.30 - 4.24 (m, 1H), 3.71 (s, 3H), 1.21 (d, 3H).

[0096] Isomer-2_D1E2: Isolated product as a white solid (0.050g, 34% yield)

[0097] ESI-MS m/z: Cak’d 464.2 found 465.1 [M+H] + ; >98% ee

[0098] 1 H NMR (400 MHz, DMSO-t/ 6 ): 6 11.58 (s, 1H), 9.97 (s, 1H), 7.91 (d, 1H), 7.88 -

7.80 (m, 2H), 7.73 - 7.65 (m, 2H), 7.66 - 7.55 (m, 2H), 7.49 - 7.35 (m, 4H), 7.31 - 7.18 (m, 3H), 5.17 (d, 1H), 4.30 - 4.24 (m, 1H), 3.71 (s, 3H), 1.21 (d, 3H).

[0099] Isomer-3_D2E1 : Isolated an Off-white solid (0.016g, 42% yield)

[00100] ESI-MS m/z: Cak’d 464.2 found 465.1 [M+H] + ; >98% ee

[00101] 1 H NMR (400 MHz, DMSO-t/ 6 ): 8 8 11.26 (s, 1H), 9.96 (s, 1H), 8.13 (d, 1H), 7.85 (dd, 1H), 7.80 (ddd, 3H), 7.47 (td, 3H), 7.43 - 7.34 (m, 2H), 7.22 (dt, 3H), 7.14 - 7.00 (m, 1H), 5.16 (d, 1H), 4.39 - 4.35 (m, 1H), 3.59 (s, 3H), 1.21 (d, 3H)

[00102] Isomer-4_D2E2: Isolated an Off-White solid (0.015g, 40% Yield)

[00103] ESI-MS m/z: Cak’d 464.2 found 465.1 [M+H] + ; >98% ee

[00104] 'H NMR (400 MHz, DMSO-t/ 6 ): 8 11.26 (s, 1H), 9.96 (s, 1H), 8.13 (d, 1H), 7.85 (dd, 1H), 7.84 - 7.75 (m, 3H), 7.47 (td, 3H), 7.39 (d, 2H), 7.27 - 7.18 ( m, 3H), 7.09 - 7.06 (m, 1H), 5.16 (d, 1H), 4.39 - 4.35 (m, 1H), 3.59 (s, 3H), 1.21 (d, 3H).

[00105] Example 2

[00106] 2-(l -(2-cyanophenyl)- 1 -phenylpropan-2-yl)-N-(3 -fluorophenyl)-5 -hydroxy- 1 - methyl-6-oxo-l,6-dihydropyrimidine-4-carboxamide

[00107] Prepared according to general procedure C.

[00108] Isomer-I DIEI : Isolated product as a white solid (0.050g, 52% yield)

[00109] ESI-MS m/z: Cak’d 482.2 found 483.2 [M+H] + ; >98% ee [00110] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.32 (s, 1H), 10.15 (d, J = 6.9 Hz, 1H), 7.97 - 7.82 (m, 1H), 7.75-7.78 (m, 1H), 7.72-6.96 (m, 3H), 7.65 - 7.52 (m, 2H), 7.52 - 7.43 (m, 1H), 7.41-7.38 (m, 2H), 7.31 - 7.15 (m, 2H), 7.09-7.06 (m, 1H), 5.21 - 5.19 (m, 1H), 4.28 - 4.25 (m, 1H), 3.71 (dd, 3H), 1.38 - 1.04 (m, 3H).

[00111] Isomer-2_D1E2: Isolated product as a white solid (0.040g, 42% yield)

[00112] ESI-MS m/z: Cak’d 482.2 found 483.2 [M+H] + ; >98% ee

[00113] X H NMR (400 MHz, DMSO- e): 6 11.33 (s, 1H), 10.14 (s, 1H), 7.91 (d, 1H), 7.83 - 7.66 (m, 4H), 7.64 - 7.54 (m, 2H), 7.52 - 7.35 (m, 3H), 7.29 - 7.21 (m, 2H), 7.09 - 7.05 (m, 1H), 5.20 (d, 1H), 4.28 - 4.26 (m, 1H), 3.71 (s, 3H), 1.22 (d, 3H).

[00114] Isomer-3_D2E1 : Isolated an Off-white solid (0.025g, 64% yield)

[00115] ESI-MS m/z: Cak’d 482.2 found 483.2 [M+H] + ; >98% ee

[00116] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.03 (s, 1H), 10.11 (s, 1H), 8.14 (d, 1H), 7.85 (dd, 1H), 7.82 - 7.71 (m, 2H), 7.62 (dd, 1H), 7.54 - 7.43 (m, 2H), 7.41 - 7.33 (m, 2H), 7.19 (t, 2H), 7.07-7.09 (m, 2H), 5.15 (d, 1H), 4.38 - 4.34 (m, 1H), 3.57 (s, 3H), 1.22 (d, 3H).

[00117] Isomer-4_D2E2: Isolated an Off-White solid (0.020g, 51% Yield)

[00118] ESI-MS m/z: Cak’d found [M+H] + ; >98% ee

[00119] 1 H NMR (400 MHz, DMSO-t/ 6 ): 6 11.03 (s, 1H), 10.11 (s, 1H), 8.14 (d, J = 8.4 Hz, 1H), 7.85 (dd, 1H), 7.82 - 7.72 (m, 2H), 7.62 (dt, 1H), 7.54 - 7.43 (m, 2H), 7.40 - 7.33 (m, 2H), 7.19 (t, 2H), 7.10-7.06 (m, 2H), 5.15 (d, 1H), 4.38 - 4.34 (m, 1H), 3.57 (s, 3H), 1.22 (d, 3H).

[00120] Example 3

[00121] 2-(l-(2-cyanophenyl)-l-phenylpropan-2-yl)-N-(2,3-difluorophe nyl)-5-hydroxy-l- methyl-6-oxo-l,6-dihydropyrimidine-4-carboxamide

[00122] Prepared according to general procedure C.

[00123] Isomer-I DIEI : Isolated product as a white solid

[00124] ESI-MS m/z: Cak’d 500.1 found 501.1 [M+H] + ; >98% ee

[00125] X H NMR (400 MHz, DMSO-t/e): 6 11.07 (s, 1H), 10.16 (s, 1H), 7.92 (d, 1H), 7.72 - 7.57 (m, 5H), 7.42 - 7.40 (m, 3H), 7.37 - 7.26 (m, 3H), 5.06 (d, 1H), 4.30 - 4.26 (m, 1H), 3.69 (s, 3H) 1.22 (d, 3H). [00126] Isomer-2_D1E2: Isolated product as a white solid

[00127] ESI-MS m/z: Cak’d 500.1 found 501.1 [M+H] + ; >98% ee

[00128] X H NMR (400 MHz, DMSO- e): 6 11.07 (s, 1H), 10.16 (s, 1H), 7.93 (d, 1H), 7.72

- 7.58 (m, 5H), 7.42 - 7.33 (m, 3H), 7.30 - 7.24 (m, 3H), 5.05 (d, 1H), 4.32 - 4.27 (m, 1H),

3.69 (s, 3H), 1.22 (d, 3H).

[00129] Isomer-3_D2E1 : Isolated an Off-white solid

[00130] ESI-MS m/z: Cak’d 500.1 found 501.1 [M+H] + ; >98% ee

[00131] 1 H NMR (400 MHz, DMSO-t/ 6 ): 6 11.04 (s, 1H), 10.12 (s, 1H), 8.13 (d, 1H), 7.89

- 7.78 (m, 3H), 7.49 - 7.47 (m, 1H), 7.36 - 7.33 (m, 4H), 7.22 - 7.17 (m, 2H), 7.11 - 7.08 (m, 1H), 5.00 (d, 1H), 4.35 - 4.25 (m, 1H), 3.57 (s, 3H),1.22 (d, 3H).

[00132] Isomer-4_D2E2: Isolated an Off-White solid

[00133] ESI-MS m/z: Cak’d 500.1 found 501.1 [M+H] + ; >98% ee

[00134] X H NMR (400 MHz, DMSO- e): 611.04 (s, 1H), 10.12 (s, 1H), 8.13 (d, 1H), 7.89

- 7.78 (m, 3H), 7.49 - 7.47 (m, 1H), 7.36 - 7.33 (m, 4H), 7.22 - 7.17 (m, 2H), 7.11 - 7.08 (m, 1H), 5.00 (d, 1H), 4.33 - 4.29 (m, 1H) , 5.53 (s, 3H), 1.22 (d, 3H).

[00135] Example 4

[00136] 2-(l-(2-cyanophenyl)-l-phenylpropan-2-yl)-N-(2-fluorophenyl) -5-hydroxy-l- methyl-6-oxo-l,6-dihydropyrimidine-4-carboxamide

[00137] Prepared according to general procedure C.

[00138] Isomer-I DIEI : Isolated product as a white solid (0.063g, 54% yield)

[00139] ESI-MS m/z: Cak’d 482.2 found 483.2 [M+H] + ; >98% ee

[00140] 'H NMR (400 MHz, DMSO-t/ 6 ): 6 11.23 (s, 1H), 10.01 (s, 1H), 7.92-7.90 (d, 2H), 7.65-7.58 (m, 4H), 7.41-7.37 (m, 3H), 7.30 - 7.24 (m, 4H), 5.03 (d, 1H), 4.29-4.26 (m, 1H),

3.68 (s, 3H), 1.19 (d, J = 6.4 Hz, 3H).

[00141] Isomer-2_D1E2: Isolated product as a white solid (0.061g, 54% yield)

[00142] ESI-MS m/z: Cak’d 482.2 found 483.1 [M+H] + ; >98% ee

[00143] 'H NMR (400 MHz, DMSO-t/ 6 ): 6 11.23 (s, 1H), 10.01 (s, 1H), 7.93-7.90 (d, 2H), 7.65-7.58 (m, 4H), 7.41-7.37 (m, 3H), 7.30 - 7.24 (m, 4H), 5.02 (d, 1H), 4.30-4.26 (m, 1H),

3.68 (s, 3H), 1.19 (d, 3H). [00144] Isomer-3_D2E1 : Isolated an Off-white solid (0.017g, 54% yield)

[00145] ESI-MS m/z: Calc’d 482.2 found 483.1 [M+H] + ; >98% ee

[00146] 1 H NMR (400 MHz, DMSO-t/ 6 ): 6 11.15 (s, 1H), 9.97 (s, 1H), 8.13 - 8.09 (m, 2H), 7.84-7.82 (m, 1H), 7.79-7.75 (m, 1H), 7.47 - 7.44 (m, 2H), 7.37-7.35 (m, 2H), 7.28-7.20 (m, 2H), 7.18-7.09 (m, 2H), 7.07-7.05 (m, 1H), 5.00 (d, 1H), 4.31 (s, 1H), 3.58 (s, 3H), 1.19 (d, 3H).

[00147] Isomer-4_D2E2: Isolated an Off-White solid (0.021g, 63% Yield)

[00148] ESI-MS m/z: Calc’d 482.2 found 483.2 [M+H] + ; >98% ee

[00149] 1 H NMR (400 MHz, DMSO-t/ 6 ): 6 11.15 (s, 1H), 9.98 (s, 1H), 8.13 - 8.09 (m, 2H), 7.84-7.82 (m, 1H), 7.79-7.75 (m, 1H), 7.47 - 7.44 (m, 2H), 7.37-7.35 (m, 2H), 7.28-7.20 (m, 2H), 7.18-7.09 (m, 2H), 7.07-7.05 (m, 1H), 5.00 (d, 1H), 4.33 (s, 1H), 3.58 (s, 3H), 1.19 (d, 3H).

[00150] General Procedure D:

[00151] Example 5

Step 6 Step 7

[00152] Step 1: tert-butyl 4-[(2-cvanophenyl)methyl]pyrazole-l-carboxylate

[00153] To a stirred solution of 2-(bromomethyl)benzonitrile (10.00 g, 51 mmol), K2CO3 (14.10 g, 102 mmol) and tert-butyl 4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyrazole- 1-carboxylate (22.51 g, 76 mmol) in 1,2-Dimethoxy ethane (100 mL) and water (20 mL) was added Tetrakis(triphenylphosphine)palladium (1.77 g, 1.53 mmol) portion wise. The resulting mixture was heated to 90 °C and stirred for 2.5 h, the mixture was then cooled to rt and filtered. The filtrate was concentrated in vacuo and the remaining aq layer was extracted with EtOAc (3 x 200 mL). The combined organic layers were washed with brine (1 x 500 mL), dried over Na2SO4 then filtered and concentrated in vacuo. The resulting crude material was purified by silica gel column chromatography (EtOAc/Pet. Ether 1 :5) affording the product as a light yellow solid (6.1 g, 42% yield). ESI-MS m/z: Calc’d 283.1 found 589.2 [2M+Na] + T H NMR (400 MHz, DMSO-t/ 6 ) 8 8.08 (d, J= 0.9 Hz, 1H), 7.82 (dd, J= 7.8, 1.4 Hz, 1H), 7.72 - 7.61 (m, 2H), 7.54 - 7.39(m, 2H), 1.56 (s, 9H).

[00154] Step 2: ethyl 2-[l-[l-(tert-butoxycarbonyl)pyrazol-4-yl]-l-(2- cvanophenyl)propan-2-yl]-5-methoxy-l-methyl-6-oxopyrimidine- 4-carboxylate

[00155] To a -60 °C stirred solution of (tert-butyl 4-[(2-cyanophenyl)methyl]pyrazole-l- carboxylate) (4.44 g, 15.7 mmol) and ethyl 2-(l-bromoethyl)-5-methoxy-l-methyl-6- oxopyrimidine-4-carboxylate (2.0 g, 6.3 mmol) in 1,2-Dimethoxy ethane (30 mL) and DMF (15 mL) was added KHMDS (12.53 mL, 12.5 mmol) dropwise over 10 min. The resulting mixture was stirred for 30 min warming to -30 °C at which point desired product was observed by LCMS. The reaction was quenched with NH4CI (sat.) at -30 °C, after warming to rt the resulting mixture was extracted with EtOAc (3 x 100 mL). The organic layers were combined and washed with brine (300 mL), dried over TsfeSCU and concentrated in vacuo. The crude product was purified by silica gel column chromatography (EtOAc/Pet. Ether 1 :2) affording the product as a yellow solid (1.30 g, 39% yield).

[00156] d r .=6: 1 as determined by LCMS

[00157] ESI-MS m/z: Cak’d 521.2 found 522.4 [M+H] +

[00158] Separation of diastereomers could be done at this step using reverse phase HPLC; Column: X-Select CSH C18 column, 19*150, 5um; 34% to 66% water (0.1% FA)/ Acetonitrile over 8 min; Flow rate: 25 mL/min.

[00159] Peak 1: RT 6.96 assigned as DI

[00160] 'H NMR (400 MHz, DMSO-t/ 6 ) 8 8.37 (s, 1H), 7.98 (s, 1H), 7.83 (d, 1H), 7.8 (d, 1H), 7.58 (t, 1H), 7.28 (t, 1H), 4.82 (d, 1H), 4.31-4.24 (m, 3H), 3.71 (s, 3H), 3.66 (s, 3H), 1.57 (s, 9H), 1.32 (t, 3H), 1.22 (d, 3H).

[00161] Peak 2: RT 7.31 assigned as D2

[00162] 'H NMR (400 MHz, DMSO-t/ 6 ) 6 8.09 - 8.04 (m, 2H), 7.84 - 7.78 (m, 2H), 7.66 (s, 1H), 7.48 (t, 1H), 4.92-4.89 (d, 1H), 4.36-4.32 (m, 2H), 4.16 - 4.02 (m, 1H), 3.79 (s, 3H), 3.53 (s, 3H), 1.51 (s, 9H), 1.34 (t, 3H), 1.05 (d, 3H).

[00163] Step 3: ethyl 2-[l-(2-cyanophenyl)-l-(lH-pyrazol-4-yl)propan-2-yl]-5- methoxy-l-methyl-6-oxopyrimidine-4-carboxylate

[00164] To a 0 °C stirred solution of (ethyl 2-[l-[l-(tert-butoxycarbonyl)pyrazol-4-yl]-l- (2-cyanophenyl)propan-2-yl]-5-methoxy-l -methyl-6-oxopyrimidine-4-carboxylate) (7.00 g, 13 mmol, 1.0 equiv) in CH2Q2 (100 mL) was added tri fluoroacetic acid (30 mL). The resulting mixture was warmed to rt and stirred for 1 h at which point complete deprotection was observed. The reaction was then concentrated in vacuo then diluted with EtOAc (100 mL) and washed with NaHCOs (Sat). The aqueous later was extracted with additional EtOAc (3 x 100 mL), the combined organic layers were dried over Na2SO4 then concentrated in vacuo. The crude product was purified by silica gel column chromatography eluting with EtOAc to afford the desired product as a yellow solid (5.10 g, 90% yield) ESI-MS m/z: Cak’d 421.1 found 422.2 [M+H] + 'H NMR (400 MHz, DMSO-t/ 6 ) 6 12.75 (s, 1H), 7.83 - 7.76 (m, 1H), 7.74 (d, J= 8.2 Hz, 1H), 7.62 - 7.49 (m, 3H), 7.22 (t, J= 7.5, 1.1 Hz, 1H), 4.76 (d, J= 11.0 Hz, 1H), 4.31 - 4.19 (m, 2H), 4.14 - 4.05 (m, 1H), 3.68 (s, 3H), 3.63 (s, 3H), 1.31 (t, J= 7.1 Hz, 3H), 1.18 (d, J= 6.6 Hz, 3H).

[00165] Step 4: ethyl 2-[l-(2-cvanophenyl)-l-(l-methylpyrazol-4-yl)propan-2-yl]-5- methoxy-l-methyl-6-oxopyrimidine-4-carboxylate

[00166] To a 0 °C stirred solution of (ethyl 2-[l-(2-cyanophenyl)-l-(lH-pyrazol-4- yl)propan-2-yl]-5-methoxy-l-methyl-6-oxopyrimidine-4-carboxy late) (0.6 g, 1.4 mmol) in DMF (5 mL) was added lodomethane (0.404 g, 2.8 mmol) and K2CO3 (0.394 g, 2.8 mmol). The resulting mixture was heated to 50 °C and stirred for 6 h at which point conversion to the desired product was observed. The reaction was then cooled to 0 °C and quenched with Water the resulting mixture was extracted with EtOAc (3 x 50 mL). The combined organic layers were dried over Na2SO4 and concentrated in vacuo. The residue was purified by silica gel column chromatography (EtOAc/Pet. Ether 1 : 1) affording the product as a yellow solid (0.430 g, 69% yield) ESI-MS m/z: Cak’d 435.1 found 436.2 [M+H] +

[00167] Step 5: lithium 2-(l-(2-cvanophenyl)-l-(l-methyl-lH-pyrazol-4-yl)propan-2- yl)-5-methoxy-l-methyl-6-oxo-l.,6-dihvdropyrimidine-4-carbox ylate

[00168] To a solution of ethyl 2-[l -(cyanophenyl)- 1-(1 -methylpyrazole-4-yl)propan-2-yl]- 5-methoxy-l-6-oxopyrimidine-4-carboxylate (0.410 g, 0.94 mmol) dissolved in MeOH (5 mL) and water (1 mL) was added LiOH-H2O (0.079 g, 1.88 mmol) portion wise. The resulting mixture was stirred at rt for 2 h at which point it was concentrated in vacuo giving the desired product as a yellow solid (0.353 g) which was used in subsequent steps with no further purification. ESI-MS m/z: Cak’d 407.1 found 408.2 [M+H] +

[00169] Step 6: 2-[l-(2-cvanophenyl)-l-(l-methylpyrazol-4-yl)propan-2-yl]-N- (2.,3- difluorophenyl)-5-methoxy-l-methyl-6-oxopyrimidine-4-carboxa mide

[00170] To a stirred solution of lithium 2-(l-(2-cyanophenyl)-l-(l-methyl-lH-pyrazol-4- yl)propan-2-yl)-5-methoxy-l-methyl-6-oxo-l,6-dihydropyrimidi ne-4-carboxylate (0.600 g, 1.45 mmol) and 2, 3 -difluoroaniline (281.1 mg, 2.18 mmol) in DMF (30 mL) was added DIPEA (0.750 mg, 5.81 mmol) and HATU (1.1 g, 2.90 mmol). The resulting mixture was stirred for 1 h at room temperature at which point it was diluted with water and the product was extracted with EtOAc (3 x 50 mL). The organic layers were collected and combined then washed with brine, dried over Na2SO4, and concentrated in vacuo. The resulting crude material was purified by reverse phase Cl 8 chromatography; Column: CSH

C18 column; 10% to 60% water (0.1% FA)/Acetonitrile over 10 min. Fractions containing product were collected and the solvent was removed in vacuo giving the product as an off white solid. (0.570g) [00171] ESI-MS m/z: Cak’d 518.2 found 519.1 [M+H] +

[00172] Separation of diastereomers was done at this step using reverse phase Cl 8 chromatography: Xselect CSH F-Phenyl OBD column, 19*250 mm; 22% to 50% Acetonitrile/water (0.05% FA) in 30 min; Flow rate: 25 mL/min.

[00173] Peak 1_D1 contained 105 mg of a white solid.

[00174] Peak 2_D2 Contained 95 mg of a white solid.

[00175] Enantiomers of this material were separated by Prep-chiral -HPLC:

[00176] DI: Column: NB-Lux 5um i-Cellulose-5, 2.12*25cm, 5pm; Mobile Phase A: Hex:MtBE=l : 1(0.5% 2M NH 3 -ME0H), Mobile Phase B:EtOH— HPLC; Flow rate: 20 mL/min; Gradient: 30% B to 30% B in 12.5 min

[00177] Peak 1 (Isomer-I DIEI): RT 6.48 min; afforded a white solid (27 mg)

[00178] Peak 2 (Isomer-2_D1E2): RT 7.79 min; afforded a white solid (26 mg)

[00179] D2: Column: CHIRALPAK ID, 2*25cm, 5um; Hex:MTBE=l : 1(0.5% 2M NH 3 -

MEOH), Mobile Phase B:EtOH— HPLC; Flow rate:20 mL/min; Gradient: 30% B to 30% B in 12.5 min).

[00180] Peak 1 (Isomer-3_D2E1): RT 7.80 min; afforded a white solid (17 mg)

[00181] Peak 2 (Isomer-4_D2E2): RT 9.76 min; afforded a white solid (25 mg)

[00182] Step 7: 2-[l-(2-cyanoDhenyl)-l-(l-methylDyrazol-4-yl)DroDan-2-yl]-N- (2.,3- difluoroDhenyl)-5-hydroxy-l-methyl-6-oxoDyrimidine-4-carboxa mide

[00183] To a stirred mixture of 2-[l-(2-cyanophenyl)-l-(l-methylpyrazol-4-yl)propan-2- yl]-N-(2,3-difluorophenyl)-5-methoxy-l-methyl-6-oxopyrimidin e-4-carboxamide (27.0 mg, 0.05 mmol) and DMF (1.4 mL) was added LiBr (90.4 mg, 1.04 mmol). This resulting mixture was then heated to 95 °C and stirred for 2h at which point complete conversion to the product was observed by LCMS. The reaction was then cooled to rt and concentrated in vacuo. The resulting crude material was purified by reverse phase Chromatography.

[00184] Isomer-I DIEI : Isolated product as a white solid (0.017g, 63% yield)

[00185] ESI-MS m/z: Cak’d 504.2 found 505.3 [M+H] + ; >98% ee

[00186] 1 H NMR (400 MHz, DMSO-t/ 6 ): 8 11.08 (s, 1H), 10.15 (s, 1H), 7.82-7.80 (m, 2H), 7.77 (s, 1H), 7.71 - 7.56 (m, 3H), 7.39 - 7.22 (m, 3H), 4.87-4.84 (d, 1H), 4.09-4.05 (m, 1H), 3.79 (s, 3H), 3.59 (s, 3H), 1.31-1.29 (d, 3H).

[00187] Isomer-2_D1E2: Isolated product as a white solid (0.015 g, 61% yield)

[00188] ESI-MS m/z: Cak’d 504.2 found 505.3 [M+H] + ; >98% ee [00189] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.08 (s, 1H), 10.15 (s, 1H), 7.82-7.80 (m, 2H), 7.77 (s, 1H), 7.72 - 7.56 (m, 3H), 7.37 - 7.22 (m, 3H), 4.87-4.84 (d, 1H), 4.11-4.03 (m, 1H), 3.79 (s, 3H), 3.59 (s, 3H), 1.31-1.29 (d, 3H).

[00190] Isomer-3_D2E1 : Isolated an Off-white solid (0.010 g, 62% yield)

[00191] ESI-MS m/z: Cak’d 504.2 found 505.3 [M+H] + ; >98% ee

[00192] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.13 (s, 1H), 10.05 (s, 1H), 8.13-7.89 (m, 2H), 7.81-7.74 (m, 2H), 7.48 - 7.43 (m, 2H), 7.31 - 7.22 (m, 3H), 4.88-4.86 (d, 1H), 4.01 (s, 1H), 3.66 (s, 3H), 3.56 (s, 3H), 1.09-1.08 (d, 3H).

[00193] Isomer-4_D2E2: Isolated an Off-White solid (0.009 g, 37% Yield)

[00194] ESI-MS m/z: Cak’d 504.2 found 505.3 [M+H] + ; >98% ee

[00195] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.13 (s, 1H), 10.05 (s, 1H), 8.13-7.89 (m, 2H), 7.81-7.74 (m, 2H), 7.48 - 7.43 (m, 2H), 7.31 - 7.22 (m, 3H), 4.88-4.86 (d, 1H), 4.01 (s, 1H), 3.66 (s, 3H), 3.56 (s, 3H), 1.09-1.08 (d, 3H).

[00196] Example 6

[00197] 2-(l-(2-cyanophenyl)-l-(l-methyl-lH-pyrazol-4-yl)propan-2-yl )-N-(2- fluorophenyl)-5-hydroxy-l -methyl -6-oxo- l,6-dihydropyrimidine-4-carboxamide

[00198] Prepared according to general procedure D.

[00199] Isomer-I DIEI : Isolated product as a white solid (0.062 g, 61% yield)

[00200] ESI-MS m/z: Cak’d 486.2 found 487.1 [M+H] + ; >98% ee

[00201] 'H NMR (400 MHz, DMSO-t/ 6 ): 6 11.24 (s, 1H), 9.98 (s, 1H), 7.92-7.88 (m, 1H), 7.82-7.77 (m, 2H), 7.61 - 7.56 (m, 3H), 7.40 - 7.23 (m, 4H), 4.86-4.83 (d, 1H), 4.09-4.08 (m, 1H), 3.79 (s, 3H), 3.60 (s, 3H), 1.31-1.29 (d, 3H).

[00202] Isomer-2_D1E2: Isolated product as a white solid (0.060 g, 63% yield)

[00203] ESI-MS m/z: Cak’d 486.2 found 487.1 [M+H] + ; >98% ee

[00204] 'H NMR (400 MHz, DMSO-t/ 6 ): 6 11.24 (s, 1H), 9.98 (s, 1H), 7.92-7.88 (m, 1H), 7.82-7.77 (m, 2H), 7.61 - 7.56 (m, 3H), 7.40 - 7.23 (m, 4H), 4.86-4.83 (d, 1H), 4.09-4.08 (m, 1H), 3.79 (s, 3H), 3.60 (s, 3H), 1.31-1.29 (d, 3H).

[00205] Isomer-3_D2E1 : Isolated an Off-white solid (0.011 g, 55% yield) [00206] ESI-MS m/z: Cak’d 486.2 found 487.1 [M+H] + ; >98% ee

[00207] 'H NMR (400 MHz, DMSO-t/ 6 ): 6 11.19 (s, 1H), 9.93 (s, 1H), 8.16-8.11 (m, 1H), 7.91-7.89 (m, 1H), 7.81 - 7.74 (m, 2H), 7.51 (s, 1H), 7.45 - 7.38 (m, 2H), 7.31 - 7.27 (m, 2H), 7.24 (s, 1H), 4.90-4.89 (d, 1H), 4.09-3.98 (m, 1H), 3.67 (s, 3H), 3.59 (s, 3H), 1.10-1.08 (d, 3H).

[00208] Isomer-4_D2E2: Isolated an Off-White solid (0.010 g, 43% Yield)

[00209] ESI-MS m/z: Cak’d 486.2 found 487.1 [M+H] + ; >98% ee

[00210] 'H NMR (400 MHz, DMSO-t/ 6 ): 6 11.19 (s, 1H), 9.93 (s, 1H), 8.16-8.11 (m, 1H), 7.91-7.89 (m, 1H), 7.81 - 7.74 (m, 2H), 7.51 (s, 1H), 7.45 - 7.38 (m, 2H), 7.31 - 7.27 (m, 2H), 7.24 (s, 1H), 4.90-4.89 (d, 1H), 4.09-3.98 (m, 1H), 3.67 (s, 3H), 3.59 (s, 3H), 1.10-1.08 (d, 3H).

[00211] Example 7

[00212] 2-(l-(2-cyano-5-(dimethykarbamoyl)phenyl)-l-(l-methyl-lH-pyr azol-4- yl)propan-2-yl)-N-(2,3-difluorophenyl)-5-hydroxy-l-methyl-6- oxo-l,6-dihydropyrimidine-4- carboxamide

[00213] Prepared according to general procedure D.

[00214] Isomer-I DIEI : Isolated product as a white solid

[00215] ESI-MS m/z: Cak’d 575.2 found 576.2 [M+H] + ; >98% ee

[00216] 'H NMR (400 MHz, DMSO-t/ 6 ): 6 11.09 (s, 1H), 10.09 (s, 1H), 7.84 (d, 1H), 7.80 (s, 1H), 7.74 (s, 1H), 7.66 (d, 1H), 7.62 - 7.56 (m, 2H), 7.29 (s, 2H), 4.87 (d, 1H), 4.09 - 4.06 (m, 1H), 3.80 (s, 3H), 3.59 (s, 3H), 2.91 (s, 3H), 2.74 (s, 3H), 1.30 (d, 3H).

[00217] Isomer-2_D1E2: Isolated product as a white solid (0.050g, 34% yield)

[00218] ESI-MS m/z: Cak’d 575.2 found 576.2 [M+H] + ; >98% ee

[00219] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.10 (s, 1H), 10.09 (s, 1H), 7.85 (d, 1H), 7.80 (s, 1H), 7.73 (s, 1H), 7.66 (d, 1H), 7.62 - 7.55 (m, 2H), 7.34 (s, 1H), 7.32 - 7.25 (m, 1H), 4.87 (d, 1H), 4.09 - 4.06 (m, 1H), 3.80 (s, 3H), 3.59 (s, 3H), 2.91 (s, 3H), 2.74 (s, 3H), 1.31 (d, 3H). [00220] Isomer-3_D2E1 : Isolated an Off-white solid (0.016g, 42% yield) [00221] ESI-MS m/z: Cak’d 575.2 found 576.2 [M+H] + ; >98% ee

[00222] 'H NMR (400 MHz, DMSO-t/ 6 ): 6 11.09 (s, 1H), 10.05 (s, 1H), 7.96 (d, 1H), 7.91 (s, 1H), 7.87 (d, Hz, 1H), 7.78 (m, 1H), 7.53 (s, 1H), 7.35 - 7.24 (m, 2H), 4.91 (d, 1H), 4.06- 4.02 (m, 1H), 3.68 (s, 3H), 3.58 (s, 3H), 2.98 (s, 3H), 2.88 (s, 3H), 1.12 (d, 3H).

[00223] Isomer-4_D2E2: Isolated an Off-White solid (0.015g, 40% Yield)

[00224] ESI-MS m/z: Cak’d 575.2 found 576.2 [M+H] + ; >98% ee

[00225] 'H NMR (400 MHz, DMSO-t/ 6 ): 6 11.09 (s, 1H), 10.05 (s, 1H), 7.96 (d, 1H), 7.92 (s, 1H), 7.87 (d, 1H), 7.78 (m, 1H), 7.53 (s, 1H), 7.31 (d, 2H), 7.26 (s, 1H), 4.91 (d, 1H), 4.06- 4.02 (m, 1H), 3.68 (s, 3H), 3.58 (s, 3H), 2.98 (s, 3H), 2.88 (s, 3H), 1.12 (d, 3H).

[00226] Example 8

[00227] 2-(l-(2-cyanophenyl)-l-(l -methyl- lH-pyrazol-4-yl)propan-2-yl)-5-hydroxy-N-

(isothiazol-4-yl)- 1 -methyl-6-oxo- 1 ,6-dihydropyrimidine-4-carboxamide

[00228] Prepared according to general procedure D.

[00229] Isomer-I DIEI : Isolated product as a white solid

[00230] ESI-MS m/z: Cak’d 475.1 found 476.1 [M+H] + ; >98% ee

[00231] 1 H NMR (400 MHz, DMSO-t/ 6 ): 6 11.30 (s, 1H), 10.62 (s, 1H), 9.16 (s, 1H), 8.92 (s, 1H), 7.81 - 7.78 (m, 2H), 7.60 - 7.54 (m, 3H), 7.23 - 7.19 (m, 1H), 5.02 (d, 1H), 4.11 - 4.06 (m, 1H), 3.81 (s, 3H), 3.63 (s, 3H), 1.34 (d, 3H).

[00232] Isomer-2_D1E2: Isolated product as a white solid (0.050g, 34% yield)

[00233] ESI-MS m/z: Cak’d 475.1 found 476.1 [M+H] + ; >98% ee

[00234] 1 H NMR (400 MHz, DMSO-t/ 6 ): 6 11.32 (s, 1H), 10.62 (s, 1H), 9.16 (d, 1H),

8.93(d, 1H), 7.80 (d, 2H),

[00235] 7.61 - 7.54 (m, 3H), 7.21 (t, 1H), 5.04 (d, 1H), 4.12 -4.08 (m, 1H), 3.81 (s, 3H),

3.64 (s, 3H), 1.35 (d, 3H).

[00236] Isomer-3_D2E1 : Isolated an Off-white solid (0.016g, 42% yield)

[00237] ESI-MS m/z: Cak’d 475.1 found 476.1 [M+H] + ; >98% ee [00238] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.14 (s, 1H), 10.52 (s, 1H), 9.16 (s, 1H), 8.85 (s, 1H), 7.93 (d, 1H), 7.85 - 7.76 (m, 2H), 7.49 - 7.45 (m, 2H), 7.21 (s, 1H), 5.00 (d, 1H), 4.07 - 4.03 (m, 1H), 3.65 (s, 3H), 3.55 (s, 3H), 1.12 (d, 3H).

[00239] Isomer-4_D2E2: Isolated an Off-White solid (0.015g, 40% Yield)

[00240] ESI-MS m/z: Cak’d 475.1 found 476.1 [M+H] + ; >98% ee

[00241] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.14 (s, 1H), 10.52 (s, 1H), 9.16 (s, 1H), 8.85 (s, 1H), 7.93 (d, 1H), 7.85 - 7.83 (m, 1H), 7.80 - 7.76 (m, 1H), 7.49 - 7.45 (m, 2H), 7.21 (s, 1H), 5.00 (d, 1H), 4.07 - 4.03 (m, 1H), 3.65 (s, 3H), 3.55 (s, 3H), 1.12 (d, 3H).

[00242] Example 9

[00243] 2-(l -(2-cyano-5-fluorophenyl)- 1 -( 1 -(2-methoxy ethyl)- lH-pyrazol-4-yl)propan-2- yl)-5-hydroxy-N-(isothiazol-4-yl)-l-methyl-6-oxo-l,6-dihydro pyrimidine-4-carboxamide

[00244] Prepared according to general procedure D.

[00245] Isomer-I DIEI : Isolated product as a white solid

[00246] ESI-MS m/z: Cak’d 537.2 found 538.3 [M+H] + ; >98% ee

[00247] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.31 (s, 1H), 10.63 (s, 1H), 9.16 (s, 1H), 8.91 (s, 1H), 7.84 (s, 1H), 7.82 - 7.78 (m, 1H), 7.74 - 7.70 (m, 1H), 7.66 (s, 1H), 7.12 - 7.07 (m, 1H), 5.08 (d, 1H), 4.23 (t, 2H), 4.11- 4.07 (m, 1H), 3.66 (d, 5H), 3.20 (s, 3H), 1.30 (d, 3H).

[00248] Isomer-2_D1E2: Isolated product as a white solid (0.050g, 34% yield)

[00249] ESI-MS m/z: Cak’d 537.2 found 538.3 [M+H] + ; >98% ee

[00250] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.31 (s, 1H), 10.64 (s, 1H), 9.16 (s, 1H), 8.91 (s, 1H), 7.84 - 7.78 (m, 2H), 7.74 - 7.66 (m, 2H), 7.12 - 7.07 (m, 1H), 5.08 (d, 1H), 4.23 (t, 2H), 4.11- 4.07 (m, 1H), 3.67 (d, 5H), 3.32 (s, 3H), 1.30 (d, 3H). [00251] Example 10

[00252] 2-(l-(2-chloro-5-fluorophenyl)-l-(l-(2-methoxyethyl)-lH-pyra zol-4-yl)propan-2- yl)-5-hydroxy-N-(isothiazol-4-yl)-l-methyl-6-oxo-l,6-dihydro pyrimidine-4-carboxamide

[00253] Prepared according to general procedure D.

[00254] Isomer-I DIEI : Isolated product as a white solid

[00255] ESI-MS m/z: Cak’d 546.1 found 547.1 [M+H] + ; >98% ee

[00256] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.21 (s, 1H), 10.59 (s, 1H), 9.16 (s, 1H), 8.91 (s, 1H), 7.79 (s, 1H), 7.63-7.60 (m, 2H), 7.28-7.24 (m, 1H), 6.91-6.86 (m,lH), 5.19 (d, 1H), 4.22-4.20 (m, 2H), 4.08-4.04 (m, 1H), 3.66-3.64 (m, 5H), 3.19 (s, 3H), 1.27 (d, 3H).

[00257] Isomer-2_D1E2: Isolated product as a white solid (0.050g, 34% yield)

[00258] ESI-MS m/z: Cak’d 546.1 found 547.1 [M+H] + ; >98% ee

[00259] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.21 (s, 1H), 10.59 (s, 1H), 9.16 (s, 1H), 8.91 (s, 1H), 7.79 (s, 1H), 7.63-7.60 (m, 2H), 7.28-7.24 (m, 1H), 6.89 (d,lH), 5.19 (d, 1H), 4.22- 4.20 (m, 2H), 4.08-4.04 (m, 1H), 3.66-3.64 (m, 5H), 3.19 (s, 3H), 1.27 (d, 3H).

[00260] Example 11

[00261] 2-(l-(2-chloro-5-fluorophenyl)-l-(l -(2 -methoxyethyl)- lH-pyrazol-4-yl)propan-2- yl)-5-hydroxy-N-(isothiazol-4-yl)-l-methyl-6-oxo-l,6-dihydro pyrimidine-4-carboxamide

[00262] Prepared according to general procedure D.

[00263] Isomer-I DIEI : Isolated product as a white solid

[00264] ESI-MS m/z: Cak’d 487.1 found 488.1 [M+H] + ; >98% ee

[00265] X H NMR (400 MHz, DMSO-t/ 6 ): 8 11.34 (s, 1H), 10.76 (s, 1H), 9.19 (s, 1H), 8.96 (s, 1H), 8.80 (d, 1H), 8.61 (d, 1H), 7.88 (d, 1H), 7.68 - 7.54 (m, 2H), 7.26 (t, 1H), 5.44 (d, 1H), 4.39- 4.35 (m, 1H), 3.64 (s, 3H), 2.50 (s, 3H), 1.26 (d, 3H).

[00266] Isomer-2_D1E2: Isolated product as a white solid (0.050g, 34% yield) [00267] ESI-MS m/z: Cak’d 487.1 found 488.1 [M+H] + ; >98% ee

[00268] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.34 (s, 1H), 10.75 (s, 1H), 9.18 (s, 1H), 8.96 (s, 1H), 8.80 (d, 1H), 8.61 (d, 1H), 7.88 (d, 1H), 7.66 - 7.53 (m, 2H), 7.26 (t, 1H), 5.44 (d, 1H), 4.39- 4.35 (m, 1H), 3.64 (s, 3H), 2.50 (s, 3H), 1.26 (d, 3H).

[00269] Isomer-3_D2E1 : Isolated an Off-white solid (0.016g, 42% yield)

[00270] ESI-MS m/z: Cak’d 487.1 found 488.1 [M+H] + ; >98% ee

[00271] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.08 (s, 1H), 10.61 (s, 1H), 9.14 (s, 1H), 8.85 (s, 1H), 8.44 (d, 1H), 8.38 (d, 1H), 7.93 - 7.86 (m, 2H), 7.77 (t, 1H), 7.51 (t, 1H), 5.46 (d, 1H), 4.42- 4.34 (m, 1H), 3.70 (s, 3H), 2.34 (s, 3H), 1.19 (d, 3H).

[00272] Isomer-4_D2E2: Isolated an Off-White solid

[00273] ESI-MS m/z: Cak’d 487.1 found 488.1 [M+H] + ; >98% ee

[00274] X H NMR (400 MHz, DMSO-t/e) 8 11.08 (s, 1H), 10.61 (s, 1H), 9.14 (s, 1H), 8.86 (s, 1H), 8.44 (d, 1H), 8.38 (d, 1H), 7.94 - 7.83 (m, 2H), 7.77 (t, 1H), 7.57 - 7.46 (m, 1H), 5.46 (d, 1H), 4.42- 4.34 (m, 1H), 3.70 (s, 3H), 2.35 (s, 3H), 1.19 (d, 3H).

[00275] Example 12

[00276] 2-( 1 -(2-chl oro-5 -fluorophenyl)- 1 -( 1 -(2 -methoxy ethyl)- 1 H-pyrazol-4-yl)propan-2- yl)-5-hydroxy-N-(isothiazol-4-yl)-l-methyl-6-oxo-l,6-dihydro pyrimidine-4-carboxamide

[00277] Prepared according to general procedure D.

[00278] Isomer-I DIEI : Isolated product as a white solid

[00279] ESI-MS m/z: Cak’d 493.1 found 494.2 [M+H] + ; >98% ee

[00280] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.38 (s, 1H), 10.63 (s, 1H), 9.15 (s, 1H), 8.90 (s, 1H), 7.89 (d, 1H), 7.72 (d, 1H), 7.67 - 7.47 (m, 2H), 7.25 (t, 1H), 4.98 (d, 1H), 4.08 - 4.04 (m, 1H), 3.72 (s, 3H), 3.60 (s, 3H), 1.41 (d, 3H).

[00281] Isomer-2_D1E2: Isolated product as a white solid

[00282] ESI-MS m/z: Cak’d 493.1 found 494.2 [M+H] + ; >98% ee

[00283] X H NMR (400 MHz, DMSO-t/ 6 ): 6 11.38 (s, 1H), 10.63 (s, 1H), 9.15 (s, 1H), 8.90 (s, 1H), 7.88 (d, 1H), 7.72 (d, 1H), 7.68 - 7.49 (m, 2H), 7.25 (t, 1H), 4.98 (d, 1H), 4.08 - 4.03 (m, 1H), 3.72 (d, 3H), 3.60 (d, 3H), 1.41 (d, 3H). TREX1 Biochemical Assay

[00284] Compound potency was assessed through a fluorescence assay measuring degradation of a custom dsDNA substrate possessing a fluorophore-quencher pair on opposing strands. Degradation of the dsDNA liberates free fluorophore to produce a fluorescent signal. Specifically, 7.5 pL of N- terminally His-Tev tagged full length human TREX1 (expressed in E. coli and purified in house) in reaction buffer (50 mM Tris (pH 7.4), 150 mM NaCl, 2 mM DTT, 0.1 mg/mL BSA, 0.01% (v/v) Tween-20 and lOOmM MgCh) was added to a 384-well Black ProxiPlate Plus (Perkin Elmer) which already contained compound (150 nL) at varying concentrations as a 10 point dose-response in DMSO. To this was added 7.5 pL of dsDNA substrate (Strand A: 5’ TEX615/GCT AGG CAG 3’; Strand B: 5’ CTG CCT AGC/IAbRQSp (Integrated DNA Technologies)) in reaction buffer. Final concentrations were 150 pM TREX1, 60 nM dsDNA substrate in reaction buffer with 1.0% DMSO (v/v). After 25 minutes at room temperature, reactions were quenched by the addition of 5 pL of stop buffer (same as reaction buffer plus 200 mM EDTA). Final concentrations in the quenched reaction were 112.5 pM TREX1, 45 nM DNA and 50 mM EDTA in a volume of 20 pL. After a 5 -minute incubation at room temperature, plates were read in a laser sourced Envision (Perkin-Elmer), measuring fluorescence at 615 nm following excitation w/ 570 nm light. IC50 values were calculated by comparing the measured fluorescence at 615 nm ratio relative to control wells pre-quenched w/ stop buffer (100% inhibition) and no inhibitor (0% inhibition) controls as using non-linear least square four parameter fits and either Genedata or GraphPad Prism (GraphPad Software, Inc.).

TREX2 Biochemical Assay

[00285] Compound potency was assessed through a fluorescence assay measuring degradation of a custom dsDNA substrate possessing a fluorophore-quencher pair on opposing strands. Degradation of the dsDNA liberates free fluorophore to produce a fluorescent signal. Specifically, 7.5 pL of N- terminally His-Tev tagged human TREX2 (residues M44-A279, expressed in /■/ coli and purified in house) in reaction buffer (50 mM Tris (pH 7.4), 150 mM NaCl, 2 mM DTT, 0.1 mg/mL BSA, 0.01% (v/v) Tween-20 and lOOmM MgCh) was added to a 384-well Black ProxiPlate Plus (Perkin Elmer) which already contained compound (150 nL) at varying concentrations as a 10 point dose-response in DMSO. To this was added 7.5 pL of dsDNA substrate (Strand A: 5’ TEX615/GCT AGG CAG 3’; Strand B: 5’ CTG CCT AGC/IAbRQSp (IDT)) in reaction buffer. Final concentrations were 2.5 nM TREX2, 60 nM dsDNA substrate in reaction buffer with 1.0% DMSO (v/v). After 25 minutes at room temperature, reactions were quenched by the addition of 5 pL of stop buffer (same as reaction buffer plus 200 mM EDTA). Final concentrations in the quenched reaction mixture were 1.875 pM TREX2, 45 nM DNA and 50 mM EDTA in a volume of 20 pL. After a 5-minute incubation at room temperature, plates were read in a laser sourced Envision (Perkin-Elmer), measuring fluorescence at 615 nm following excitation w/ 570 nm light. IC50 values were calculated by comparing the measured fluorescence at 615 nm ratio relative to control wells pre-quenched w/ stop buffer (100% inhibition) and no inhibitor (0% inhibition) controls as using non-linear least square four parameter fits and either Genedata or GraphPad Prism (GraphPad Software, Inc.).

[00286] Results are shown in Table 1. TREX1 IC50 : A = <0.001 pM; B = 0.001 to 0.1 pM; C = 0.1 to 10 pM; D = >10 pM. TREX2 IC50 : A = <1 pM, B = 1 to 10 pM, C = 10 to 100 pM, D = >100 pM.

Table 1

[00287] While we have described a number of embodiments, it is apparent that our basic examples may be altered to provide other embodiments that utilize the compounds and methods of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example.

[00288] The contents of all references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated herein in their entireties by reference. Unless otherwise defined, all technical and scientific terms used herein are accorded the meaning commonly known to one with ordinary skill in the art.