Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CYCLIC VINYL SULFONE COMPOUNDS AS WRN INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2023/062575
Kind Code:
A1
Abstract:
Disclosed herein are certain succinamido derivatives of Formula (I): (I) that inhibit Wemer Syndrome ATP dependent helicase enzyme (WRN) activity, in particular inhibit WRN helicase domain activity and are thereofore useful in treating cancers treatable by inhibition of WRN, including cancers characterized by microsatellite instability (MSI) and/or defective DNA mismatch repair system (dMMR). Also, disclosed are pharmaceutical compositions comprising such compounds, methods of using such compounds, and methods making the same.

Inventors:
ADAMS ASHLEY (US)
BURGESS JOELLE LORRAINE (US)
BURY MICHAEL J (US)
DEMARTINO MICHAEL P (US)
DOWDELL SARAH E (US)
GLOGOWSKI MICHAL PAWEL (US)
GODFREY NICOLE (US)
GRAYBILL TODD L (US)
MANNS SHARADA (US)
MURPHY DENNIS (US)
NEVINS NEYSA (US)
RIDGERS LANCE HOWARD (US)
SHEARER BARRY GEORGE (US)
SHENJE RAYNOLD (US)
TALLANT MATTHEW (US)
YING MABEN (US)
JESO VALER (US)
PHELAN JAMES P (US)
JONES BRIAN T (US)
TAYGERLY JOSHUA P G (US)
Application Number:
PCT/IB2022/059817
Publication Date:
April 20, 2023
Filing Date:
October 13, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
IDEAYA BIOSCIENCES INC (US)
GLAXOSMITHKLINE INTELLECTUAL PROPERTY NO 4 LTD (GB)
International Classes:
C07D409/12; A61K31/4015; A61P35/00; C07D409/14; C07D413/14; C07D417/14
Domestic Patent References:
WO2019241802A22019-12-19
WO2019236448A12019-12-12
Foreign References:
EP3848365A12021-07-14
Other References:
STN REGISTRY ET AL: "N-(2,3-dihydro-1,1-dioxido-3-thienyl)-1,2-dihydro-6-(4-methoxyphenyl)-2-oxo-3-pyridinecarboxamide", 12 March 2014 (2014-03-12), pages 1 - 1, XP093012912, Retrieved from the Internet [retrieved on 20230111]
JOSHUA A. SOMMERS ET AL: "A high-throughput screen to identify novel small molecule inhibitors of the Werner Syndrome Helicase-Nuclease (WRN)", PLOS ONE, vol. 14, no. 1, 9 January 2019 (2019-01-09), pages e0210525, XP055724955, DOI: 10.1371/journal.pone.0210525
AGGARWAL M. ET AL: "Inhibition of helicase activity by a small molecule impairs Werner syndrome helicase (WRN) function in the cellular response to DNA damage or replication stress", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 108, no. 4, 10 January 2011 (2011-01-10), pages 1525 - 1530, XP055795378, ISSN: 0027-8424, Retrieved from the Internet DOI: 10.1073/pnas.1006423108
DOBZHANSKY, T., GENETICS, vol. 31, 1946, pages 269 - 290
HUANG ET AL., NATURE REVIEWS DRUG DISCOVERY, vol. 19, 2020, pages 23 - 38
BAUDRIN ET AL., FRONT. ONCOL., 2018
VAN WIETMARSCHEN N. ET AL., NATURE, vol. 586, 2020, pages 292 - 298
GOEBEL L. ET AL., RSC MED. CHEM., vol. 11, 2020, pages 760
BERGE, S.M. ET AL.: "Pharmaceutical Salts", JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 1 - 19, XP002675560, DOI: 10.1002/jps.2600660104
T.W. GREENP.G.M. WUTS: "Protecting Groups in Organic Synthesis", 1991, JOHN WILEY & SONS
DUDLEY, JONATHAN C. ET AL., CLINICAL CANCER RESEARCH, vol. 22, no. 4, 2016, pages 813 - 820
WU ET AL., AM J HUM GENETICS, vol. 65, 1999, pages 1291 - 1298
JOURNAL OF THE AMERICAN CHEMICAL SOCIETY
JOURNAL OF BIOLOGICAL CHEMISTRY
KORENAGA, T.KOSAKI T.FUKUMURA, R.EMA T.SAKAI, T., ORG. LETT., vol. 7, 2005, pages 4915
Download PDF:
Claims:
What is Claimed:

1. A compound according to Formula (I), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, heteroaryl, or (C3-Cn)cycloalkyl; each R1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-Ce)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, SF5, heterocycloalkyl, aryl, aryloxy, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-O-, aryl-(Ci-C4)alkyl-O-, heterocycloalkyl- (Ci-C4)alkyl-O-, heteroaryl-(Ci-C4)alkyl-O-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-NH-, aryl- (Ci-C4)alkyl-NH-, heterocycloalkyl-(Ci-C4)alkyl-NH-, heteroaryl-(Ci-C4)alkyl-NH-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-, aryl-(Ci-C4)alkyl-, heterocycloalkyl-(Ci-C4)alkyl-, and heteroaryl-(Ci-C4)alkyl-, wherein each of aryl, cycloalkyl, heterocycloalkyl, and heteroaryl moieties in R1 is substituted with 0-3 Rla; [] or two adjacent R1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 Rla; each R2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R3 is hydrogen or (Ci-Ce)alkyl; each R4 is independently halogen, (Ci-Ce)alkyl, or halo(Ci-Ce)alkyl; each Rla is independently halogen, CN, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; p is 1 or 2; and q is 0, 1, or 2; provided that the compound is not N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-6-(4- methoxyphenyl)-2 -oxo-1, 2-dihydropyridine-3-carboxamide.

2. A compound according to Formula (I), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, heteroaryl, or (C3-Cn)cycloalkyl; each R1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-Ce)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, SF5, heterocycloalkyl, aryl, aryloxy, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-O-, aryl-(Ci-C4)alkyl-O-, heterocycloalkyl- (Ci-C4)alkyl-O-, heteroaryl-(Ci-C4)alkyl-O-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-NH-, aryl- (Ci-C4)alkyl-NH-, heterocycloalkyl-(Ci-C4)alkyl-NH-, heteroaryl-(Ci-C4)alkyl-NH-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-, aryl-(Ci-C4)alkyl-, heterocycloalkyl-(Ci-C4)alkyl-, and heteroaryl-(Ci-C4)alkyl-, wherein each of aryl, cycloalkyl, heterocycloalkyl, and heteroaryl moieties in R1 is substituted with 0-3 Rla; or two adjacent R1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 Rla; each R2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R3 is hydrogen or (Ci-Ce)alkyl; each R4 is independently halogen, (Ci-Ce)alkyl, or halo(Ci-Ce)alkyl; each Rla is independently halogen, CN, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; p is 1 or 2; and q is 0, 1, or 2; provided that when ring A is phenyl and m is 1, R1 is not methoxy.

3. A compound according to Formula (I), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, heteroaryl, or (C3-Cn)cycloalkyl; each R1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-Ce)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, SF5, heterocycloalkyl, aryl, aryloxy, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-O-, aryl-(Ci-C4)alkyl-O-, heterocycloalkyl- (Ci-C4)alkyl-O-, heteroaryl-(Ci-C4)alkyl-O-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-NH-, aryl- (Ci-C4)alkyl-NH-, heterocycloalkyl-(Ci-C4)alkyl-NH-, heteroaryl-(Ci-C4)alkyl-NH-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-, aryl-(Ci-C4)alkyl-, heterocycloalkyl-(Ci-C4)alkyl-, and heteroaryl-(Ci-C4)alkyl-, wherein each of aryl, cycloalkyl, heterocycloalkyl, and heteroaryl moieties in R1 is substituted with 0-3 Rla; or two adjacent R1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 Rla; each R2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R3 is hydrogen or (Ci-Ce)alkyl; each R4 is independently halogen, (Ci-Ce)alkyl, or halo(Ci-Ce)alkyl; each Rla is independently halogen, CN, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; p is 1 or 2; and q is 0, 1, or 2.

4. The compound of any one of claims 1 to 3 or a pharmaceutically acceptable salt thereof, wherein the compound has the structure of Formula (la):

5. The compound of any one of claims 1 to 3 or a pharmaceutically acceptable salt thereof, wherein the compound has the structure of Formula (lb):

6. The compound of any one of claims 1 to 5 or a pharmaceutically acceptable salt thereof, wherein ring A is selected from the group consisting of phenyl, naphthalenyl, thiophenyl, pyridyl, pyrazinyl, benzothiophenyl, benzofuranyl, furanyl, 1,3 -thiazolyl, isoxazolyl, cyclohexyl, cyclopentyl, and cyclohexenyl .

7. The compound of any one of claims 1 to 6 or a pharmaceutically acceptable salt thereof, wherein ring A is phenyl.

8. The compound of any one of the claims 1 to 7 or a pharmaceutically acceptable salt thereof, wherein each R1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, (Ci-Ce)alkylsulfanyl, or (C3-Ce)cycloalkyl.

9. The compound of any one of the claims 1 to 7 or a pharmaceutically acceptable salt thereof, wherein each R1 is independently (Ci-Ce)alkyl.

10. The compound of any one of the claims 1 to 7 or a pharmaceutically acceptable salt thereof, wherein each R1 is methyl.

11. The compound of any one of the claims 1 to 10 or a pharmaceutically acceptable salt thereof, wherein m is 0, 1, or 2.

12. The compound of any one of the claims 1 to 10 or a pharmaceutically acceptable salt thereof, wherein m is 0.

13. The compound of any one of the claims 1 to 10 or a pharmaceutically acceptable salt thereof, wherein m is 1.

14. The compound of any one of the claims 1 to 10 or a pharmaceutically acceptable salt thereof, wherein m is 2.

15. The compound of any one of the claims 1 to 7 or a pharmaceutically acceptable salt thereof, wherein two adjacent R1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring.

16. The compound of any one of the claims 1 to 15 or a pharmaceutically acceptable salt thereof, wherein n is 0.

17. The compound of any one of the claims 1 to 15 or a pharmaceutically acceptable salt thereof, wherein n is 1 and R2 is hydroxyl.

18. The compound of any one of the claims 1 to 17 or a pharmaceutically acceptable salt thereof, wherein R3 is hydrogen or methyl.

19. The compound of any one of the claims 1 to 17 or a pharmaceutically acceptable salt thereof, wherein R3 is hydrogen.

20. The compound of any one of the claims 1 to 19 or a pharmaceutically acceptable salt thereof, wherein p is 1.

21. The compound of any one of the claims 1 to 19 or a pharmaceutically acceptable salt thereof, wherein p is 2.

22. The compound of any one of the claims 1 to 21 or a pharmaceutically acceptable salt thereof, wherein q is 0.

23. The compound of claim 1 or a pharmaceutically acceptable salt thereof, wherein the compound is selected from the compounds in Table 1.

24. A pharmaceutical composition comprising a compound of any one of claims 1 to 23 or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipients.

25. A method for treating a cancer treatable by inhibition of WRN in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a compound of any one of claims 1 to 23 or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 24.

26. A method for treating a cancer characterized by MSI-H and/or dMMR in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a compound of any one of claims 1 to 23, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 24.

27. Use of a compound of any one of claims 1 to 23, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 24, in the manufacture of a medicament for treating cancer.

28. A compound of any one of claims 1 to 23, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of claim 24, for use in treating cancer.

Description:
CYCLIC VINYL SULFONE COMPOUNDS AS WRN INHIBITORS

FIELD

Disclosed herein are certain cyclic vinyl sulfone compounds as WRN inhibitors that inhibit Wemer Syndrome helicase enzyme (WRN) activity and in particular, inhibit the ATP dependent helicase domain activity and are therefore useful in treating cancers treatable by inhibition of WRN, including cancers characterized by microsatellite instability (MSI) and/or defective DNA mismatch repair system (dMMR). Also, disclosed are pharmaceutical compositions comprising such compounds and methods making the same.

REFERENCE TO A “SEQUENCE LISTING”

The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled 70050W001_Seq_List.xml created August 23, 2022. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.

BACKGROUND

Cancer is a leading cause of death throughout the world. A limitation of prevailing therapeutic approaches, e.g. chemotherapy is that their cytotoxic effects are not restricted to cancer cells and adverse side effects can occur within normal tissues. Consequently, novel strategies are needed to better target cancer cells.

Synthetic lethality (SL) arises when a combination of genetic deficiencies (e.g. gene mutations, silencing or global genomic lesions) and/or molecular perturbations (e.g. gene expression knockout/knockdown, pharmacological inhibition/activation) corresponding to two or more genes impaired cell wellbeing, whereas presence of single deficiency/perturbation does not (Dobzhansky, T., Genetics 1946; 31, 269-290, Huang et al., Nature Reviews Drug Discovery 2020; volume 19, pages 23-38)

Microsatellite instability is a genomic lesion caused by defects in mismatch repair machinery (dMMR). MSI status is present in colorectal cancer, endometrial cancer, gastric cancer and other cancer types. Mutation or silencing of MMR genes, including MLH1, MSH2, MSH6 and PMS2, abrogates cell’s ability to repair DNA mismatch mutations (Baudrin et al., Front. Oncol. 2018). As a consequence, tumor with MSI-H status carries higher mutation burden, disrupted microsatellite repeat sequences and extended TA dinucleotide repeat sequences across the genome (van Wietmarschen N. et al., Nature 2020; 586, pages 292-298). MSI status can be assessed by molecular testing of certain microsatellites, next-generation sequencing of patient genome or by immunohistochemical evaluation of expression of certain MMR proteins. Tumors can be categorized into MSI high (MSI-H), MSI low (MSI-L) and MSS depending on the number of tested microsatellite showing instability. Based on a consensus NCI-Reference Panel (Bethesda, 1998), MSI can be assessed by molecular testing of five microsatellites - including two mononucleotides (BAT25 and BAT26) and three dinucleotides (D2S123, D5S346, D17S250). Tumors are denoted as MSI-high (MSI-H) if two or more of the microsatellite markers show instability, MSI-low (MSI-L) if only one microsatellite marker shows instability, and MS-stable (MSS) if none of the five microsatellite markers show instability. In some instances, for example where molecular testing or immunohistochemical evaluation is not able to distinguish between MSI-L and general chromosomal instability, tumors can be classified as a MSS neoplasms.

WRN (WRN RecQ helicase) has been identified as a synthetic lethality vulnerability to cancer cells with high microsatellite instability status (MSI-H). WRN contains an exonuclease domain and an ATP -dependent helicase domain. It is localized to the nucleus and unwinds double strand DNA, particularly secondary structures (fork DNA, holliday junction, G4-quadruaplex, DNA hairpin and cruciform etc.) during DNA replication, damage and repair processes. Its helicase activity has been shown to be indispensable to the survival of MSI cell lines as helicase -deficient WRN mutant is insufficient to rescue impaired cell viability from WRN knockout or knockdown. The absence of either the WRN protein or inhibition of its helicase activity prevents normal DNA damage and repair processes, leading to increased DNA double-strand breaks (DSB) and subsequent growth arrest and cell death.

Covalent inhibitors represent a class of small molecules which form covalent bonds with their biological targets to inhibit activities of these targets in physiological or pathological conditions. In general, covalent inhibitors engage with nucleophilic residues (e.g. Cysteine, Serine, Threonine, Histidine, Arginine, Tyrosine) lining specific binding pockets on target proteins, in a nucleophilic addition or substitution reaction, with their reactive electrophilic warhead. To date, a variety of reactive warheads have been identified, including epoxide, aziridine, ester, ketone, a, -unsaturated carbonyl, nitrile, etc. Covalent inhibitors have been discovered as medicines for more than a century, starting with Aspirin being manufactured and marketed as painkillers and anti-inflammatory drug, although its mechanism of action was not revealed until 1970s to be an irreversible inhibitor of cyclooxygenase- 1 (COX- 1).

Other notable covalent inhibitors used as medicine include antibiotics Penicillin, proton pump inhibitor Omeprazole and Lansoprazole, anticoagulant Clopidogrel.

SUMMARY

Disclosed herein are certain cyclic vinyl sulfone compounds that inhibit WRN activity and are therefore useful in treating cancer treatable by inhibition of WRN, including cancers characterized by high microsatellite instability (MSI-H) and/or defective DNA mismatch repair system (dMMR). The cyclic vinyl sulfone compounds disclosed herein can inhibit, in particular, the ATP dependent helicase domain activity of WRN protein. Also, disclosed are pharmaceutical compositions comprising such compounds, methods of using such compounds, and methods for making the same.

In a first aspect, provided is a compound of Formula (I), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, heteroaryl, or (C3-Cn)cycloalkyl; each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-C6)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, SF5, heterocycloalkyl, aryl, aryloxy, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-O-, aryl-(Ci-C4)alkyl-O-, heterocycloalkyl- (Ci-C 4 )alkyl-O-, heteroaryl-(Ci-C4)alkyl-O-, (C3-C6)cycloalkyl-(Ci-C4)alkyl-NH-, aryl- (Ci-C4)alkyl-NH-, heterocycloalkyl-(Ci-C4)alkyl-NH-, heteroaryl-(Ci-C4)alkyl-NH-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-, aryl-(Ci-C4)alkyl-, heterocycloalkyl-(Ci-C4)alkyl-, and heteroaryl-(Ci-C4)alkyl-, wherein each of aryl, cycloalkyl, heterocycloalkyl, and heteroaryl moieties in R 1 is substituted with 0-3 R la ; or two adjacent R 1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la ; each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R 3 is hydrogen or (Ci-Ce)alkyl; each R 4 is independently halogen, (Ci-Ce)alkyl, or halo(Ci-Ce)alkyl; each R la is independently halogen, CN, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; p is 1 or 2; and q is 0, 1, or 2; provided that the compound is not N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-6-(4- methoxyphenyl)-2 -oxo-1, 2-dihydropyridine-3-carboxamide.

In a second aspect, provided is a method of treating a cancer treatable by inhibition of WRN in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.

In a third aspect, provided is a method of treating a cancer characterized by MSI-H and/or dMMR in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.

In a fourth aspect, provided is a pharmaceutical composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.

In a fifth aspect, provided a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of cancer treatable by inhibition of WRN.

In a sixth aspect, provided a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of cancer characterized by high MSI and/or dMMR in a patient.

In a seventh aspect, provided is a method of treating a cancer in a patient, comprising:

(i) determining if the cancer comprises high MSI and/or dMMR; and (ii) if the the cancer comprises high MSI and/or dMMR, then administering to the patient a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.

All the compounds and pharmaceutical compositions provided herein can be used in all the methods provided herein. For example, the compounds and pharmaceutical compositions provided herein can be used in all the methods for treatment and/or prevention of all diseases or disorders provided herein. Thus, the compounds and pharmaceutical compositions provided herein are for use as a medicament.

DETAILED DESCRIPTION

There is evidence in literature suggesting that WRN helicase inhibitor may present a novel therapy to tumor with mismatch repair deficiency (dMMR) and in particular those with high microsatellite instability (MSI-H). And in recent years, covalent inhibitors are being developed more frequently for oncology indications, for example, EGFR inhibitor Afatinib, BTK inhibitor Ibrutinib and Acalabrutinib, and Kras G12C inhibitors sotorasib and adagrasib (Goebel L. et al., RSC Med. Chem. 2020, 11, 760). Notably, these aforementioned irreversible kinase inhibitors and Kras G12C inhibitors are all Cysteine-reactive compounds, suggesting that targeting cysteine residue may be an effective strategy to develop covalent inhibitors for cancer targets.

The present application provides certain cyclic vinyl sulfone compounds as WRN covalent inhibitors that inhibit Werner Syndrome helicase enzyme (WRN) activity and in particular, inhibit the ATP dependent helicase domain activity and are therefore useful in treating cancers treatable by inhibition of WRN.

Before the present disclosure is further described, it is to be understood that the invention is not limited to the particular embodiments set forth herein, and it is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting.

The singular forms “a,” “an,” and “the” as used herein and in the appended claims include plural referents unless the context clearly dictates otherwise. It is further noted that the claims may be drafted to exclude one or more optional elements. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology such as “solely,” “only” and the like in connection with the recitation of claim elements, or use of a “negative limitation.

Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.

When needed, any definition herein may be used in combination with any other definition to describe a composite structural group. By convention, the trailing element of any such definition is that which attaches to the parent moiety. For example, the composite group alkoxyalkyl means that an alkoxy group is attached to the parent molecule through an alkyl group.

The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Further, the dates of publication provided may be different from the actual publication dates, which may need to be independently confirmed.

Definitions:

Unless otherwise stated, the following terms used in the specification and claims are defined for the purposes of this Application and have the following meaning.

As used herein, the term "alkyl" represents a saturated, straight, or branched hydrocarbon moiety. The term "(Ci-Cejalkyl" refers to an alkyl moiety containing from 1 to 6 carbon atoms. Exemplary alkyls include, but are not limited to methyl, ethyl, w-propyl. isopropyl, n- butyl, isobutyl, s-butyl. /-butyl, pentyl, and hexyl.

When the term "alkyl" is used in combination with other substituent groups, such as "halo(Ci-C6)alkyl" or “hydroxy(Ci-C6)alkyl”, the term “alkyl” is intended to encompass a divalent straight or branched-chain hydrocarbon radical, wherein the point of attachment is through the alkyl moiety. The term “halo(Ci-C6)alkyl” is intended to mean a radical having one or more halogen atoms, which may be the same or different, at one or more carbon atoms of an alkyl moiety containing from 1 to 6 carbon atoms, which is a straight or branched-chain carbon radical. Examples of "halo(Ci-C6)alkyl" groups useful in the present disclosure include, but are not limited to, -CHF2 (difluoromethyl), -CF3 (trifluoromethyl), -CCh (trichloromethyl), 1,1 -difluoroethyl, 2,2,2-trifluoroethyl, and hexafluoroisopropyl. Examples of “hydroxy(Ci-C6)alkyl” groups useful in the present disclosure include, but are not limited to, hydroxymethyl, hydroxyethyl, and hydroxyisopropyl.

“Alkoxy” refers to a group containing an alkyl radical, defined hereinabove, attached through an oxygen linking atom. The term “(Ci-C4)alkoxy” refers to a straight- or branched-chain hydrocarbon radical having at least 1 and up to 4 carbon atoms attached through an oxygen linking atom. Exemplary “(Ci-C4)alkoxy” groups useful in the present disclosure include, but are not limited to, methoxy, ethoxy, w-propoxy. isopropoxy, w-butoxy. s-butoxy. isobutoxy, and /-butoxy.

When the term "alkoxy" is used in combination with other substituent groups, such as "halo(Ci-C6)alkoxy", the term “alkoxy” is intended to encompass a divalent straight or branched-chain hydrocarbon radical, wherein the point of attachment is to the alkyl moiety through an oxygen linking atom. The term “halo(Ci-Ce)alkoxy” refers to a straight- or branched-chain hydrocarbon radical, having at least 1 and up to 6 carbon atoms with one or more halogen atoms, which may be the same or different, attached to one or more carbon atoms, which radical is attached through an oxygen linking atom. Exemplary “halo(Ci-Ce)alkoxy” groups useful in the present disclosure include, but are not limited to, - OCHF2 (difluoromethoxy), -OCF3 (trifluoromethoxy), and -OCH(CF3)2 (hexafluoroisopropoxy) .

“Alkylsulfanyl” means a -SR radical where R is alkyl as defined above, e.g., methylsulfanyl, ethylsulfanyl, and the like. The term “(Ci-C6)alkylsulfanyl” refers to a straight- or branched-chain hydrocarbon radical, having at least 1 and up to 6 carbon atoms, which radical is attached through S linking atom.

“Amino” means a -NH2.

“Alkylamino” means a -NHR radical where R is alkyl as defined above, e.g., methylamino, ethylamino, propylamino, or 2-propylamino, and the like. The term “((Ci-C6)alkyl)amino-” refers to a straight- or branched-chain hydrocarbon radical, having at least 1 and up to 6 carbon atoms, which radical is attached through an NH linking group. “Aminoalkyl” means an alkyl radical, defined hereinabove, substituted with -NH2, e.g., aminomethyl, aminoethyl, and the like. The term “amino(Ci-C6)alkyl” refers to a straight- or branched-chain hydrocarbon radical, having at least 1 and up to 6 carbon atoms, which radical is substituted with -NH2.

“Aryl” means a monovalent monocyclic or bicyclic aromatic hydrocarbon radical of 6 to 10 ring atoms, e.g., phenyl or naphthyl.

As used herein, the term “cycloalkyl” refers to a non-aromatic cyclic hydrocarbon ring containing the specified number of carbon atoms which may be saturated or contains one double bond. The term “(C3-C6)cycloalkyl” refers to a non-aromatic cyclic hydrocarbon ring having from three to six ring carbon atoms. Exemplary “(C3-C6)cycloalkyl” groups useful in the present disclosure include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, and cyclohexenyl.

As used herein, the term “cycloalkyloxy” or “cycloalkoxy” refers to a group containing a cycloalkyl radical, defined hereinabove, attached through an oxygen linking atom. Exemplary “(C3-C6)cycloalkyloxy” groups useful in the present disclosure include cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, and cyclohexyloxy.

“Halogen”or “halo” means fluoro, chloro, bromo, or iodo, preferably fluoro or chloro.

As used herein, the term “hydroxy” or “hydroxyl” means a -OH.

“Hydroxyalkyl” means an alkyl radical as defined above, substituted with one or two hydroxy groups, provided that if two hydroxy groups are present, they are not both on the same carbon atom. Representative examples include, but are not limited to, hydroxymethyl, 2-hydroxy- ethyl, 2-hydroxypropyl, and 3 -hydroxypropyl.

“Heteroaryl” means a monovalent monocyclic or bicyclic aromatic radical of 5 to 10 ring atoms, unless otherwise stated, where one or more ring atoms are heteroatoms selected from N, O, and S, the remaining ring atoms being carbon. Monocyclic heteroaryl moieties can have 5 or 6 ring atoms where one or more, (in one embodiment, one, two, or three), ring atoms are heteroatoms selected from N, O, and S, the remaining ring atoms being carbon. Bicyclic heteroaryl moieties can have 9 or 10 ring atoms where one or more, (in one embodiment, one, two, three, or four), ring atoms are heteroatoms selected from N, O, and S, the remaining ring atoms being carbon. Non-limiting examples of heteroaryl groups include pyridyl, pyridazinyl, pyrazinyl, pyrimindinyl, triazinyl, quinolinyl, quinoxalinyl, quinazolinyl, cinnolinyl, phthalazinyl, benzotriazinyl, purinyl, benzimidazolyl, benzopyrazolyl, benzotriazolyl, benzisoxazolyl, isobenzofuryl, isoindolyl, indolizinyl, benzotriazinyl, thienopyridinyl, thienopyrimidinyl, pyrazolopyrimidinyl, imidazopyridines, benzothiaxolyl, benzofuranyl, benzothienyl, indolyl, quinolyl, isoquinolyl, isothiazolyl, pyrazolyl, indazolyl, pteridinyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiadiazolyl, pyrrolyl, thiazolyl, furyl, thienyl, and the like.

“Heterocycloalkyl” means a saturated or unsaturated monovalent monocyclic group of 4 to 8 ring atoms in which one or two ring atoms are heteroatom selected from N, O, and S(O)n, where n is an integer from 0 to 2, the remaining ring atoms being carbon. Additionally, one or two ring carbon atoms in the heterocycloalkyl ring can optionally be replaced by a -CO- group. The term “heterocycloalkyl” includes, but is not limited to, azetidinyl, oxetanyl, pyrrolidine, piperidine, homopiperidino, 2-oxopyrrolidinyl, 2-oxopiperidinyl, morpholino, piperazine, tetrahydro-pyranyl, thiomorpholino, and the like. When the heterocycloalkyl ring is unsaturated it can contain one or two ring double bonds provided that the ring is not aromatic.

"Pharmaceutically acceptable salts" as used herein is meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein. When compounds disclosed herein contain relatively acidic functionalities, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent. Examples of salts derived from pharmaceutically acceptable inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, potassium, sodium, zinc and the like. Salts derived from pharmaceutically acceptable organic bases include salts of primary, secondary and tertiary amines, including substituted amines, cyclic amines, naturally- occurring amines and the like, such as arginine, betaine, caffeine, choline, N,N’- dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine and the like. When compounds of the present disclosure contain relatively basic functionalities, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogen carbonic, phosphoric, monohydrogen phosphoric, dihydrogen phosphoric, sulfuric, monohydrogen sulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, malonic, benzoic, succinic, suberic, fumaric, mandelic, phthalic, benzene sulfonic, p-tolylsulfonic, citric, tartaric, methane sulfonic, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge, S.M., et al, “Pharmaceutical Salts”, Journal of Pharmaceutical Science, 1977, 66, 1-19).

The neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner. The parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present disclosure.

The compounds of Formula (I) or salts thereof may exist in stereoisomeric forms (e.g., it contains one or more asymmetric carbon atoms). The individual stereoisomers (enantiomers and diastereomers) and mixtures of these are included within the scope of the present disclosure. The scope of the present disclosure includes mixtures of stereoisomers as well as purified enantiomers or enantiomerically/diastereomerically enriched mixtures. Unless otherwise indicated, when a stereochemical depiction is shown, it is meant that the isomer with the depicted stereochemistry is present and substantially free of the other isomer(s). “Substantially free of’ another isomer indicates at least an 80/20 ratio of the two isomers, more preferably 90/10, or 95/5 or more.

Likewise, it is understood that a compound of Formula (I) or salts thereof may exist in tautomeric forms other than that shown in the formula and these are also included within the scope of the present disclosure. For example, while the compounds of Formula (I) are depicted as containing a pyridin-2-one moiety, the corresponding 2-hydroxypyridine tautomer is also included within the scope of the present disclosure. It is to be understood that the present disclosure includes all combinations and subsets of the particular groups defined hereinabove.

The compounds of Formula (I) may also contain unnatural amounts of isotopes at one or more of the atoms that constitute such compounds. Unnatural amounts of an isotope may be defined as ranging from the amount found in nature to an amount 100% of the atom in question that differ only in the presence of one or more isotopically enriched atoms. Exemplary isotopes that can be incorporated into the compounds of the present disclosure, such as a compound of Formula (I) include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine, and iodine, such as 2 H, 3 H, n C, 13 C, 14 C, 13 N, 15 N, 15 O, 17 O, 18 O, 32 P, 33 P, 35 S, 18 F, 36 C1, 123 I, and 125 I, respectively. Isotopically labeled compounds (e.g., those labeled with 3 H and 14 C) can be useful in compound or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes can be useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements). In some embodiments, in the compounds disclosed herein, including in Table 1 below one or more hydrogen atoms are replaced by 2 H or 3 H, or one or more carbon atoms are replaced by 13 C- or 14 C-enriched carbon. Positron emitting isotopes such as 15 O, 13 N, n C, and 15 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. Isotopically labeled compounds can generally be prepared by following procedures analogous to those disclosed in the Schemes or in the Examples herein, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.

“Pharmaceutically acceptable carrier or excipient” means a carrier or an excipient that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes a carrier or an excipient that is acceptable for veterinary use as well as human pharmaceutical use. “A pharmaceutically acceptable carrier/excipient” as used in the specification and claims includes both one and more than one such excipient.

“About” as used herein, is intended to qualify the numerical values which it modifies, denoting such a value as variable within a margin of error. When no particular margin of error, such as a standard deviation to a mean value given in a chart or table of data, is recited, the term “about” should be understood to mean that range which would encompass ± 10%, preferably ± 5%, the recited value and the range is included.

“Disease” as used herein is intended to be generally synonymous, and is used interchangeably with, the terms “disorder,” “syndrome,” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms, and causes the human or animal to have a reduced duration or quality of life.

“Patient” is generally synonymous with the term “subject” and as used herein includes all mammals including humans. Examples of patients include humans, livestock such as cows, goats, sheep, pigs, and rabbits, and companion animals such as dogs, cats, rabbits, and horses. Preferably, the patient is a human.

“In need of treatment” or “in need thereof’ as used herein refers to a judgment made by a physician or other caregiver that a subject requires or will benefit from treatment. This judgment is made based on a variety of factors that are in the realm of the physician’s or caregiver's expertise.

“Administration”, “administer” and the like, as they apply to, for example, a patient, cell, tissue, organ, or biological fluid, refer to contact of, for example, a compound of Formula (I), or a pharmaceutical composition comprising same, to the subject, cell, tissue, organ, or biological fluid. In the context of a cell, administration includes contact (e.g., in vitro or ex vivo) of a reagent to the cell, as well as contact of a reagent to a fluid, where the fluid is in contact with the cell.

“Therapeutically effective amount” as used herein means the amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof that, when administered to a patient for treating a disease either alone or as part of a pharmaceutical composition and either in a single dose or as part of a series of doses, is sufficient to affect such treatment for the disease. The “therapeutically effective amount” will vary depending on the compound, the disease and its severity and the age, weight, etc., of the patient to be treated. The therapeutically effective amount can be ascertained by measuring relevant physiological effects, and it can be adjusted in connection with the dosing regimen and diagnostic analysis of the subject’s condition, and the like. By way of example, measurement of the serum level of a compound of Formula (I) (or, e.g., a metabolite thereof) at a particular time post-administration may be indicative of whether a therapeutically effective amount has been used.

As used herein, the term “treatment” or “treating” refers to alleviating the specified condition, eliminating or reducing one or more symptoms of the condition, and slowing or eliminating the progression of the condition in a previously afflicted or diagnosed patient or subject.

"Inhibiting", "reducing," or any variation of these terms in relation ofWRN, includes any measurable decrease or complete inhibition to achieve a desired result. For example, there may be a decrease of about, at most about, or at least about 5%, 10%, 15%, 20%, 25%, 30%,

35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or more, or any range derivable therein, reduction of WRN helicase domain activity compared to its normal activity.

In a first aspect, provided is a compound of Formula (I), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, heteroaryl, or (C3-Cn)cycloalkyl; each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-Ce)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, SF5, heterocycloalkyl, aryl, aryloxy, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-O-, aryl-(Ci-C4)alkyl-O-, heterocycloalkyl- (Ci-C4)alkyl-O-, heteroaryl-(Ci-C4)alkyl-O-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-NH-, aryl- (Ci-C4)alkyl-NH-, heterocycloalkyl-(Ci-C4)alkyl-NH-, heteroaryl-(Ci-C4)alkyl-NH-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-, aryl-(Ci-C4)alkyl-, heterocycloalkyl-(Ci-C4)alkyl-, and heteroaryl-(Ci-C4)alkyl-, wherein each of aryl, cycloalkyl, heterocycloalkyl, and heteroaryl moieties in R 1 is substituted with 0-3 R la ; or two adjacent R 1 groups taken together with the ring atoms to which they are attached form a 5 - or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la ; each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R 3 is hydrogen or (Ci-Ce)alkyl; each R 4 is independently halogen, (Ci-Ce)alkyl, or halo(Ci-Ce)alkyl; each R la is independently halogen, CN, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; p is 1 or 2; and q is 0, 1, or 2; provided that the compound is not N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-6-(4- methoxyphenyl)-2 -oxo- l,2-dihydropyridine-3 -carboxamide (structure shown below).

In a second aspect, provided is a compound of Formula (I’), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, 5- to 10-membered heteroaryl, or (C3-Ce)cycloalkyl; each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-Ce)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, or 3- to 6-membered heterocycloalkyl; or two adjacent R 1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la ; each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R 3 is hydrogen or (Ci-Ce)alkyl; each R la is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; and p is 1 or 2; provided that the compound is not N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-6-(4- methoxyphenyl)-2 -oxo-1, 2-dihydropyridine-3-carboxamide.

In a third aspect, provided is a compound according to Formula (I), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, heteroaryl, or (C3-Cn)cycloalkyl; each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-Ce)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, SF5, heterocycloalkyl, aryl, aryloxy, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-O-, aryl-(Ci-C4)alkyl-0-, heterocycloalkyl- (Ci-C4)alkyl-0-, heteroaryl-(Ci-C4)alkyl-O-, (C3-C6)cycloalkyl-(Ci-C4)alkyl-NH-, aryl- (Ci-C4)alkyl-NH-, heterocycloalkyl-(Ci-C4)alkyl-NH-, heteroaryl-(Ci-C4)alkyl-NH-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-, aryl-(Ci-C4)alkyl-, heterocycloalkyl-(Ci-C4)alkyl-, and heteroaryl-(Ci-C4)alkyl-, wherein each of aryl, cycloalkyl, heterocycloalkyl, and heteroaryl moieties in R 1 is substituted with 0-3 R la ; or two adjacent R 1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la ; each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R 3 is hydrogen or (Ci-Ce)alkyl; each R 4 is independently halogen, (Ci-Ce)alkyl, or halo(Ci-Ce)alkyl; each R la is independently halogen, CN, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; p is 1 or 2; and q is 0, 1, or 2; provided that when ring A is phenyl and m is 1, R 1 is not methoxy.

In a fourth aspect, provided is a compound according to Formula (F), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, 5- to 10-membered heteroaryl, or (C3-Ce)cycloalkyl; each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-C6)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, or 3- to 6-membered heterocycloalkyl; or two adjacent R 1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la ; each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R 3 is hydrogen or (Ci-Ce)alkyl; each R la is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; and p is 1 or 2; provided that when ring A is phenyl and m is 1, R 1 is not methoxy.

In a fifth aspect, provided is a compound according to Formula (I), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, heteroaryl, or (C3-Cn)cycloalkyl; each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-C6)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, SF5, heterocycloalkyl, aryl, aryloxy, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-O-, aryl-(Ci-C4)alkyl-O-, heterocycloalkyl- (Ci-C4)alkyl-O-, heteroaryl-(Ci-C4)alkyl-O-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-NH-, aryl- (Ci-C4)alkyl-NH-, heterocycloalkyl-(Ci-C4)alkyl-NH-, heteroaryl-(Ci-C4)alkyl-NH-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-, aryl-(Ci-C4)alkyl-, heterocycloalkyl-(Ci-C4)alkyl-, and heteroaryl-(Ci-C4)alkyl-, wherein each of aryl, cycloalkyl, heterocycloalkyl, and heteroaryl moieties in R 1 is substituted with 0-3 R la ; or two adjacent R 1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la ; each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R 3 is hydrogen or (Ci-Ce)alkyl; each R 4 is independently halogen, (Ci-Ce)alkyl, or halo(Ci-Ce)alkyl; each R la is independently halogen, CN, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; p is 1 or 2; and q is 0, 1, or 2.

In a sixth aspect, provided is a compound according to Formula (F), or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, 5- to 10-membered heteroaryl, or (C3-Ce)cycloalkyl; each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-C6)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, or 3- to 6-membered heterocycloalkyl; or two adjacent R 1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la ; each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R 3 is hydrogen or (Ci-Ce)alkyl; each R la is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; and p is 1 or 2.

In one embodiment, the present application provides a compound according to Formula (la), or a pharmaceutically acceptable salt thereof, wherein ring A, R 1 , R 2 , R 3 , m, n, and p are as defined herein.

In one embodiment, the present application provides a compound according to Formula (lb), or a pharmaceutically acceptable salt thereof, wherein ring A, R 1 , R 2 , R 3 , m, n, and p are as defined herein.

In one embodiment, the present application provides a compound according to Formula (I) as described herein, wherein ring A is aryl. In one embodiment, ring A is phenyl or naphthalenyl. In one embodiment, ring A is phenyl.

In another embodiment, the present application provides a compound according to Formula (I) as described herein, wherein ring A is heteroaryl. In one embodiment, ring A is 5- to 10- membered heteroaryl. In one embodiment, ring A is 5-membered heteroaryl. In one embodiment, ring A is thiophenyl, thiazolyl, furanyl, or isoxazolyl. In one embodiment, ring A is thiophenyl. In one embodiment, ring A is thiazolyl. In one embodiment, ring A is furanyl. In another embodiment, ring A is 6-membered heteroaryl. In one embodiment, ring A is pyridyl or pyrazinyl. In one embodiment, ring A is pyridyl.

In one embodiment, ring A is 9-membered heteroaryl. In one embodiment, ring A is benzothiophenyl or benzofuranyl. In another embodiment, ring A is 10-membered heteroaryl.

In one embodiment, the present application provides a compound according to Formula (I) as described herein, wherein ring A is (C3-Cn)cycloalkyl. In one embodiment, ring A is (C3-Ce)cycloalkyl. In one embodiment, ring A is (C5-Ce)cycloalkyl. In one embodiment, ring A is cyclohexyl. In one embodiment, ring A is cyclopentyl. In one embodiment, ring A is cyclohexenyl.

In one embodiment, ring A is selected from the group consisting of phenyl, naphthalenyl, thiophenyl, pyridyl, pyrazinyl, benzothiophenyl, benzofuranyl, furanyl, 1,3 -thiazolyl, isoxazolyl, cyclohexyl, cyclopentyl, and cyclohexenyl.

In some embodiments, ring A is not substituted, i.e., m is 0. In one embodiment, m is 1. In one embodiment, m is 2. In one embodiment, m is 3.

In some embodiments, ring A is substituted, and each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-Ce)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, SF5, heterocycloalkyl, aryl, aryloxy, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-O-, aryl-(Ci-C4)alkyl-O-, heterocycloalkyl-(Ci-C4)alkyl-O-, heteroaryl-(Ci-C4)alkyl-O-, (C3-Ce)cycloalkyl- (Ci-C4)alkyl-NH-, aryl-(Ci-C4)alkyl-NH-, heterocycloalkyl-(Ci-C4)alkyl-NH-, heteroaryl- (Ci-C4)alkyl-NH-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-, aryl-(Ci-C4)alkyl-, heterocycloalkyl- (Ci-C4)alkyl-, and heteroaryl-(Ci-C4)alkyl-, wherein each of aryl, cycloalkyl, heterocycloalkyl, and heteroaryl moieties in R 1 is substituted with 0-3 R la .

In one embodiment, each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-Ce)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, or 3- to 6-membered heterocycloalkyl. In one embodiment, each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, (Ci-Ce)alkylsulfanyl, or (C3-C6)cycloalkyl. In one embodiment, each R 1 is independently halogen. In one embodiment, each R 1 is independently chloro or fluoro. In one embodiment, each R 1 is independently (Ci-Ce)alkyl or halo(Ci-Ce)alkyl. In one embodiment, each R 1 is independently (Ci-Ce)alkyl. In one embodiment, each R 1 is independently halo(Ci-Ce)alkyl. In one embodiment, each R 1 is independently methyl or ethyl. In one embodiment, each R 1 is methyl. In one embodiment, each R 1 is ethyl. In one embodiment, each R 1 is independently trifluoromethyl or perfluoroethyl. In one embodiment, each R 1 is trifluoromethyl.

In some embodiments, when ring A is substituted, two adjacent R 1 groups taken together with the two ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered non-aromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la and each R la is independently halogen, CN, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy. In one embodiment, each R la is independently halogen. In one embodiment, each R la is independently (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, or (Ci-Ce)alkoxy. In one embodiment, ring A is aryl fused with a 5- or 6-membered non-aromatic ring. In one embodiment, ring A is phenyl fused with a 5- or 6-membered non-aromatic ring. In one embodiment, ring A is

In one embodiment, n is 0. In one embodiment, n is 1. In one embodiment, n is 2. In some embodiments, each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, or CN. In one embodiment, each R 2 is independently chloro or fluoro. In one embodiment, each R 2 is independently (Ci-Ce)alkyl or halo(Ci-Ce)alkyl. In one embodiment, R 2 is trifluoromethyl. In one embodiment, R 2 is CN. In one embodiment, R 2 is hydroxyl.

In one embodiment, R 3 is hydrogen. In one embodiment, R 3 is (Ci-Ce)alkyl. In one embodiment, R 3 is (Ci-C2)alkyl. In one embodiment, R 3 is methyl.

In one embodiment, each R 4 is independently halogen, (Ci-Ce)alkyl, or halo(Ci-Ce)alkyl. In one embodiment, each R 4 is halogen. In one embodiment, each R 4 is (Ci-Ce)alkyl. In one embodiment, R 4 is F. In one embodiment, R 4 is methyl.

In one embodiment, p is 1. In one embodiment, p is 2.

In one embodiment, q is 0. In one embodiment, q is 1. In one embodiment, q is 2.

In one embodiment, the present application provides a compound according to Formula (Ic), or a pharmaceutically acceptable salt thereof, wherein ring A, R 1 , R 2 , m, and n are as defined herein.

In one embodiment, the present application provides a compound according to Formula (Id), or a pharmaceutically acceptable salt thereof, wherein ring A, R 1 , R 2 , m, and n are as defined herein. In one embodiment, the present application provides a compound according to Formula (le), or a pharmaceutically acceptable salt thereof, wherein ring A, R 1 , R 2 , m, and n are as defined herein.

In one embodiment, the present application provides a compound according to Formula (If), or a pharmaceutically acceptable salt thereof, wherein ring A, R 1 , R 2 , m, and n are as defined herein. In one embodiment, the present application provides a compound according to Formula (Ig), or a pharmaceutically acceptable salt thereof, wherein ring A, R 1 , and m are as defined herein. In one embodiment, ring A is phenyl. In one embodiment, ring A is thiophenyl. In one embodiment, ring A is (C3-Ce)cycloalkyl.

In one embodiment, the present application provides a compound according to Formula (Ih), or a pharmaceutically acceptable salt thereof, wherein ring A, R 1 , and m are as defined herein. In one embodiment, ring A is phenyl. In one embodiment, ring A is thiophenyl. In one embodiment, ring A is (C3-Ce)cycloalkyl.

In one embodiment, the present application provides a compound according to Formula (li), or a pharmaceutically acceptable salt thereof, wherein ring A, R 1 , and m are as defined herein. In one embodiment, ring A is phenyl. In one embodiment, ring A is thiophenyl. In

5 one embodiment, ring A is (C3-Ce)cycloalkyl.

Representative compounds of Formula (I) are listed in Table 1 below.

In some embodiments, the present disclosure provides a compound selected from the group of Examples 1-70. In some embodiments, the present disclosure provides a compound selected from the group of Examples 1-173. It is understood that the embodiments set forth above include all combination of embodiments and subembodiments listed therein.

General Synthetic Schemes

The compounds of this disclosure may be made by a variety of methods, including well-known standard synthetic methods. Illustrative general synthetic methods are set out below and then specific compounds of the invention are prepared in the working examples. A skilled artisan will appreciate that if a substituent described herein is not compatible with the synthetic methods described herein, the substituent may be protected with a suitable protecting group that is stable to the reaction conditions. The protecting group may be removed at a suitable point in the reaction sequence to provide a desired intermediate or target compound. In all of the schemes described below, protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles of synthetic chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T.W. Green and P.G.M. Wuts, (1991) Protecting Groups in Organic Synthesis, John Wiley & Sons, incorporated by reference with regard to protecting groups). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The selection of processes as well as the reaction conditions and order of their execution shall be consistent with the preparation of compounds of the present disclosure. Starting materials are commercially available or are made from commercially available starting materials using methods known to those skilled in the art.

Certain compounds of Formula (I) can be prepared according to Schemes 1-4 or analogous methods.

Scheme 1

Pharmaceutical Composition

The compounds of Formula (I), or a pharmaceutically acceptable salt thereof, provided herein may be in the form of compositions suitable for administration to a subject. In general, such compositions are pharmaceutical compositions comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable or physiologically acceptable excipients. In certain embodiments, the compound of Formula (I), or a pharmaceutically acceptable salt thereof is present in a therapeutically effective amount. The pharmaceutical compositions may be used in the methods disclosed herein; thus, for example, the pharmaceutical compositions can be administered ex vivo or in vivo to a subject in order to practice the therapeutic methods and uses described herein.

The pharmaceutical compositions can be formulated to be compatible with the intended method or route of administration; exemplary routes of administration are set forth herein. Furthermore, the pharmaceutical compositions may be used in combination with other therapeutically active agents or compounds as described herein in order to treat the diseases, disorders and conditions contemplated by the present disclosure.

The pharmaceutical compositions containing the active ingredient (e.g., a compound of Formula (I), or a pharmaceutically acceptable salt thereof) may be in a form suitable for oral use, for example, as tablets, capsules, troches, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups, solutions, microbeads or elixirs. Pharmaceutical compositions intended for oral use may be prepared according to any method known in the art for the manufacture of pharmaceutical compositions, and such compositions may contain one or more agents such as, for example, sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets, capsules and the like contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets, capsules, and the like. These excipients may be, for example, diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, com starch, or alginic acid; binding agents, for example starch, gelatin or acacia, and lubricating agents, for example magnesium stearate, stearic acid or talc.

The tablets, capsules and the like suitable for oral administration may be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action. For example, a time-delay material such as glyceryl monostearate or glyceryl di-stearate may be employed. The tablets may also be coated by techniques known in the art to form osmotic therapeutic tablets for controlled release. Additional agents include biodegradable or biocompatible particles or a polymeric substance such as polyesters, polyamine acids, hydrogel, polyvinyl pyrrolidone, polyanhydrides, polyglycolic acid, ethylene -vinyl acetate, methylcellulose, carboxymethylcellulose, protamine sulfate, or lactide and glycolide copolymers, polylactide and glycolide copolymers, or ethylene vinyl acetate copolymers in order to control delivery of an administered composition. For example, the oral agent can be entrapped in microcapsules prepared by coacervation techniques or by interfacial polymerization, by the use of hydroxymethyl cellulose or gelatin-microcapsules or poly (methyl methacrylate) microcapsules, respectively, or in a colloid drug delivery system. Colloidal dispersion systems include macromolecule complexes, nanocapsules, microspheres, microbeads, and lipid-based systems, including oil- in-water emulsions, micelles, mixed micelles, and liposomes. Methods for the preparation of the above-mentioned formulations are known in the art.

Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate, kaolin or microcrystalline cellulose, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, or olive oil.

Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture thereof. Such excipients can be suspending agents, for example sodium carboxymethylcellulose, methylcellulose, (hydroxypropyl)methyl cellulose, sodium alginate, polyvinyl-pyrrolidone, gum tragacanth and gum acacia; dispersing or wetting agents, for example a naturally-occurring phosphatide (e.g., lecithin), or condensation products of an alkylene oxide with fatty acids (e.g., poly-oxyethylene stearate), or condensation products of ethylene with long chain aliphatic alcohols (e.g., for heptdecaethyleneoxycetanol), or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol (e.g., polyoxyethylene sorbitol monooleate), or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides (e.g., polyethylene sorbitan monooleate). The aqueous suspensions may also contain one or more preservatives.

Oily suspensions may be formulated by suspending the active ingredient in a vegetable oil, for example, arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation.

Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified herein.

The pharmaceutical compositions may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, for example olive oil or arachis oil, or a mineral oil, for example, liquid paraffin, or mixtures of these. Suitable emulsifying agents may be naturally occurring gums, for example, gum acacia or gum tragacanth; naturally occurring phosphatides, for example, soy bean, lecithin, and esters or partial esters derived from fatty acids; hexitol anhydrides, for example, sorbitan monooleate; and condensation products of partial esters with ethylene oxide, for example, polyoxyethylene sorbitan monooleate.

The pharmaceutical compositions typically comprise a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable excipient. Suitable pharmaceutically acceptable excipients include, but are not limited to, antioxidants (e.g., ascorbic acid and sodium bisulfate), preservatives (e.g., benzyl alcohol, methyl parabens, ethyl or n-propyl, p-hydroxybenzoate), emulsifying agents, suspending agents, dispersing agents, solvents, fdlers, bulking agents, detergents, buffers, vehicles, diluents, and/or adjuvants. For example, a suitable vehicle may be physiological saline solution or citrate buffered saline, possibly supplemented with other materials common in pharmaceutical compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles. Those skilled in the art will readily recognize a variety of buffers that can be used in the pharmaceutical compositions and dosage forms contemplated herein. Typical buffers include, but are not limited to, pharmaceutically acceptable weak acids, weak bases, or mixtures thereof. As an example, the buffer components can be water soluble materials such as phosphoric acid, tartaric acids, lactic acid, succinic acid, citric acid, acetic acid, ascorbic acid, aspartic acid, glutamic acid, and salts thereof. Acceptable buffering agents include, for example, a Tris buffer, N-(2-Hydroxyethyl)piperazine-N'-(2 -ethane sulfonic acid) (HEPES), 2-(N-Morpholino)ethanesulfonic acid (MES), 2-(N-Morpholino)ethanesulfonic acid sodium salt (MES), 3 -(N-Morpholino)propane sulfonic acid (MOPS), and N- tris[Hydroxymethyl]methyl-3 -aminopropanesulfonic acid (TAPS).

After a pharmaceutical composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or dehydrated or lyophilized powder. Such formulations may be stored either in a ready-to-use form, a lyophilized form requiring reconstitution prior to use, a liquid form requiring dilution prior to use, or other acceptable form. In some embodiments, the pharmaceutical composition is provided in a single-use container (e.g., a single-use vial, ampoule, syringe, or autoinjector (similar to, e.g., an EpiPen®)), whereas a multi-use container (e.g., a multi-use vial) is provided in other embodiments.

The pharmaceutical compositions may be in the form of a sterile injectable aqueous or oleagenous suspension. The suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents mentioned herein. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butane diol. Acceptable diluents, solvents and dispersion media that may be employed include water, Ringer's solution, isotonic sodium chloride solution, Cremophor EL™ (BASF, Parsippany, NJ) or phosphate buffered saline (PBS), ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), and suitable mixtures thereof. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed, including synthetic mono- or diglycerides. Moreover, fatty acids such as oleic acid, find use in the preparation of injectables. Prolonged absorption of particular injectable formulations can be achieved by including an agent that delays absorption (e.g., aluminum monostearate or gelatin).

A compound of Formula (I), or a pharmaceutically acceptable salt thereof may also be administered in the form of suppositories for rectal administration or sprays for nasal or inhalation use. The suppositories can be prepared by mixing the drug with a suitable nonirritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include, but are not limited to, cocoa butter and polyethylene glycols.

Routes of Administration

Compounds of Formula (I), or a pharmaceutically acceptable salt thereof and compositions containing the same may be administered in any appropriate manner. Suitable routes of administration include oral, parenteral (e.g., intramuscular, intravenous, subcutaneous (e.g., injection or implant), intraperitoneal, intracistemal, intraarticular, intraperitoneal, intracerebral (intraparenchymal) and intracerebro ventricular), nasal, vaginal, sublingual, intraocular, rectal, topical (e.g., transdermal), buccal and inhalation. Depot injections, which are generally administered subcutaneously or intramuscularly, may also be utilized to administer the compounds of Formula (I), or a pharmaceutically acceptable salt thereof over a defined period of time. Particular embodiments of the present disclosure contemplate oral administration.

Treatment of Patients Having Tumors Characterized by High Microsatellite Instability

In one aspect, provided herein is a method for decreasing proliferation in a proliferative cell having a microsatellite instability (MSI), comprising decreasing the helicase activity of Werner syndrome ATP-dependent helicase (WRN) in the proliferative cell. In some embodiments, decreasing the helicase activity of Wemer syndrome ATP-dependent helicase (WRN) in the proliferative cell is achieved by administering a compound of Formula of (I) (or any embodiment thereof disclosed herein) or a pharmaceutically acceptable salt thereof. In some embodiments, the proliferative cell is characterized as having MSI low (MSI-L). In some embodiments, the proliferative cell is characterized as having high MSI (MSI-H), used interchangeably with MSI-high. Cells can be characterized as MSI, including MSI-L or MSI- H, or as MSS (MS-stable), according to the method known in the art (see, for example, Dudley, Jonathan C., et al., Clinical Cancer Research, 22(4): 813-820, 2016.). MSI-H is used to classify tumors as having a high frequency of MSI. A tumor can be classified as MSI, including MSI-low or MSI-high, using polymerase chain reaction (PCR) and/or immunohistochemistry (IHC) assays. As stated in Dudley et al. (supra), a tumor is classified as MSI-H by PCR if (i) there is a shift (usually downward) in the size of at least two microsatellite loci from a reference panel of five microsatellite loci in tumor relative to normal, where the reference panel can be the “Bethesda Panel,” also referred to herein as the “NCI-Reference Panel (Bethesda, 1998)”, which includes two mononucleotide loci (BAT-25 and BAT-26) and three dinucleotide loci (D2S123, D5S346, and D17S250), or alternatively, the reference panel can be Promega Corporation’s MSI Analysis System, which includes five mononucleotide loci (BAT-25, BAT-26, NR-21, NR-24, and MONO-27); or (ii) there is a shift in the size of 30% or more microsatellite loci from a reference panel of more than five microsatellite loci in tumor relative to normal. The MSI-H phenotype is associated with germline defects in the mismatch repair genes MLH1, MSH2, MSH6, and PMS2, and is the primary phenotype observed in tumors from patients with HNPCC/Lynch syndrome. A tumor is classified as MSI-H in IHC test if it shows a loss of protein expression for at least 1 of the above 4 mismatch repair genes. Cells can be similarly classified as MSI-H using the tests described herein for tumors.

In some embodiments, a tumor or cell is classified as MSI-H using PCR to amplify the five microsatellite loci of the “Bethesda Panel” (BAT-25, BAT-26, D2S123, D5S346, and D17S250) from both tumor tissue or cells and normal tissue or cells, wherein the tumor or cell is classified as MSI-H if there is a shift in the size of at least two of the microsatellite loci from the tumor tissue or cells relative to the normal tissue or cells. In some embodiments, the shift in size of the microsatellite loci is a downward shift.

In some embodiments, a tumor or cell is classified as MSI-H using PCR to amplify the five microsatellite loci of Promega Corporation’s MSI Analysis System (BAT-25, BAT-26, NR- 21, NR-24, and MONO-27) from both tumor tissue or cells and normal tissue or cells, wherein the tumor or cell is classified as MSI-H if there is a shift in the size of at least two of the microsatellite loci from the tumor tissue or cells relative to the normal tissue or cells. In some embodiments, the shift in size of the microsatellite loci is a downward shift.

In some embodiments, a tumor is classified as MSI-H using IHC to determine the expression level of the MMR proteins MLH1, MSH2, MSH6, and/or PMS2 in both tumor tissue and normal tissue, wherein the tumor is classified as MSI-H if there is a loss of protein expression for at least one of the MMR proteins in the tumor tissue relative to the normal tissue. In some embodiments, the loss of protein expression is a decrease of at least 20% (such as a decrease of 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more).

In contrast, a tumor is classified as MSI-L by PCR if (i) there is a shift in the size of one microsatellite locus from a reference panel of five microsatellite loci in tumor relative to normal, where the reference panel can be the “Bethesda Panel” or Promega Corporation’s MSI Analysis System; or (ii) there is a shift in the size of less than 30% microsatellite loci from a reference panel of more than five microsatellite loci in tumor relative to normal. MSI- L tumors are thought to represent a distinct mutator phenotype with potentially different molecular etiology than MSI-H tumors (Thibodeau, 1998; Wu et al., 1999, Am J Hum Genetics 65: 1291-1298). Cells can be similarly classified as MSI-L using the tests described herein for tumors.

Cancers classified as MSI-H include, but not limited to, uterine corpus endometrial carcinoma, colon adenocarcinoma, stomach adenocarcinoma, rectal adenocarcinoma, adenoid cystic carcinoma, uterine carcinosarcoma, cervical squamous cell carcinoma, and endocervical adenocarcinoma.

In one embodiment, the present disclosure provides a method for treating cancer in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, heteroaryl, or (C3-Cn)cycloalkyl; each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-C6)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, SF5, heterocycloalkyl, aryloxy, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-O-, aryl-(Ci-C4)alkyl-O-, heterocycloalkyl-(Ci-C4)alkyl-O-, heteroaryl-(Ci-C4)alkyl-O-, (C3-Ce)cycloalkyl-(Ci-C4)alkyl-NH-, aryl-(Ci-C4)alkyl-NH-, heterocycloalkyl-(Ci-C4)alkyl-NH-, heteroaryl-(Ci-C4)alkyl-NH-, (C3-Ce)cycloalkyl- (Ci-C4)alkyl-, aryl-(Ci-C4)alkyl-, heterocycloalkyl-(Ci-C4)alkyl-, and heteroaryl- (Ci-C4)alkyl-, wherein each of aryl, cycloalkyl, heterocycloalkyl, and heteroaryl moieties in R 1 is substituted with 0-3 R la ; or two adjacent R 1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la ; each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R 3 is hydrogen or (Ci-Ce)alkyl; each R 4 is independently halogen, (Ci-Ce)alkyl, or halo(Ci-Ce)alkyl; each R la is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; p is 1 or 2; and q is 0, 1, or 2.

In one embodiment, the present disclosure provides a method for treating cancer in a patient in need thereof comprising administering to the patient a therapeutically effective amount of a compound of Formula (F) or a pharmaceutically acceptable salt thereof: wherein: ring A is aryl, 5- to 10-membered heteroaryl, or (C3-Ce)cycloalkyl; each R 1 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxy(Ci-Ce)alkyl, amino(Ci-C6)alkyl, (C3-Ce)cycloalkyl, hydroxyl, (Ci-Ce)alkoxy, halo(Ci-Ce)alkoxy, (C3-Ce)cycloalkoxy, (Ci-C6)alkylsulfanyl, amino, ((Ci-C6)alkyl)amino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, CN, COOH, CONH2, or 3- to 6-membered heterocycloalkyl; or two adjacent R 1 groups taken together with the ring atoms to which they are attached form a 5- or 6-membered non-aromatic ring, wherein the 5- or 6-membered nonaromatic ring includes 0-2 heteroatoms selected from N and O and is substituted with 0-3 R la ; each R 2 is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, (Ci-Ce)alkoxy, amino, ((Ci-Cejalkyljamino-, ((Ci-C6)alkyl)((Ci-C6)alkyl)amino-, or CN;

R 3 is hydrogen or (Ci-Ce)alkyl; each R la is independently halogen, (Ci-Ce)alkyl, halo(Ci-Ce)alkyl, hydroxyl, or (Ci-Ce)alkoxy; m is 0, 1, 2, or 3; n is 0, 1, or 2; and p is 1 or 2.

In one embodiment, the present disclosure provides use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of comprising the compound, in the manufacture of a medicament for treating cancer. In one embodiment, wherein the cancer is treatable by inhibition of WRN. In one embodiment, the cancer is characterized by MSI-H and/or dMMR.

In another embodiment, the present disclosure provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of comprising the compound, for use a method of treatment. In another embodiment, the present disclosure provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition of comprising the compound, for use_in treating cancer. In one embodiment, wherein the cancer is treatable by inhibition of WRN. In one embodiment, the cancer is characterized by MSI-H and/or dMMR.

Combination Therapy

The present disclosure contemplates the use of compounds of Formula (I), or a pharmaceutically acceptable salt thereof in combination with one or more active therapeutic agents (e.g., chemotherapeutic agents) or other prophylactic or therapeutic modalities (e.g., radiation). In such combination therapy, the various active agents frequently have different, complementary mechanisms of action. Such combination therapy may be especially advantageous by allowing a dose reduction of one or more of the agents, thereby reducing or eliminating the adverse effects associated with one or more of the agents. Furthermore, such combination therapy may have a synergistic therapeutic or prophylactic effect on the underlying disease, disorder, or condition.

As used herein, “combination” is meant to include therapies that can be administered separately, for example, formulated separately for separate administration (e.g., as may be provided in a kit), and therapies that can be administered together in a single formulation (i.e., a “co-formulation”).

In certain embodiments, the compounds of Formula (I), or a pharmaceutically acceptable salt thereof are administered or applied sequentially, e.g., where one agent is administered prior to one or more other agents. In other embodiments, the compounds of Formula (I), or a pharmaceutically acceptable salt thereof are administered simultaneously, e.g., where two or more agents are administered at or about the same time; the two or more agents may be present in two or more separate formulations or combined into a single formulation (i.e., a coformulation). Regardless of whether the two or more agents are administered sequentially or simultaneously, they are considered to be administered in combination for purposes of the present disclosure.

The compounds of Formula (I), or a pharmaceutically acceptable salt thereof may be used in combination with at least one other (active) agent in any manner appropriate under the circumstances. In one embodiment, treatment with the at least one active agent and at least one compound of Formula (I), or a pharmaceutically acceptable salt thereof is maintained over a period of time. In another embodiment, treatment with the at least one active agent is reduced or discontinued (e.g., when the subject is stable), while treatment with the compound of Formula (I), or a pharmaceutically acceptable salt thereof is maintained at a constant dosing regimen. In a further embodiment, treatment with the at least one active agent is reduced or discontinued (e.g., when the subject is stable), while treatment with a compound of Formula (I), or a pharmaceutically acceptable salt thereof is reduced (e.g., lower dose, less frequent dosing or shorter treatment regimen). In yet another embodiment, treatment with the at least one active agent is reduced or discontinued (e.g., when the subject is stable), and treatment with the compound of Formula (I), or a pharmaceutically acceptable salt thereof is increased (e.g., higher dose, more frequent dosing or longer treatment regimen). In yet another embodiment, treatment with the at least one active agent is maintained and treatment with the compound of Formula (I), or a pharmaceutically acceptable salt thereof is reduced or discontinued (e.g., lower dose, less frequent dosing or shorter treatment regimen). In yet another embodiment, treatment with the at least one active agent and treatment with the compound of Formula (I), or a pharmaceutically acceptable salt thereof are reduced or discontinued (e.g., lower dose, less frequent dosing or shorter treatment regimen).

The present disclosure provides methods for treating cancer with a compound of Formula (I), or a pharmaceutically acceptable salt thereof and at least one additional therapeutic or diagnostic agent.

Dosing

The compounds of Formula (I), or a pharmaceutically acceptable salt thereof provided herein may be administered to a subject in an amount that is dependent upon, for example, the goal of administration (e.g., the degree of resolution desired); the age, weight, sex, and health and physical condition of the subject to which the formulation is being administered; the route of administration; and the nature of the disease, disorder, condition or symptom thereof. The dosing regimen may also take into consideration the existence, nature, and extent of any adverse effects associated with the agent(s) being administered. Effective dosage amounts and dosage regimens can readily be determined from, for example, safety and dose-escalation trials, in vivo studies (e.g., animal models), and other methods known to the skilled artisan.

In general, dosing parameters dictate that the dosage amount be less than an amount that could be irreversibly toxic to the subject (the maximum tolerated dose (MTD)) and not less than an amount required to produce a measurable effect on the subject. Such amounts are determined by, for example, the pharmacokinetic and pharmacodynamic parameters associated with ADME, taking into consideration the route of administration and other factors.

An effective dose (ED) is the dose or amount of an agent that produces a therapeutic response or desired effect in some fraction of the subjects taking it. The “median effective dose” or EDso of an agent is the dose or amount of an agent that produces a therapeutic response or desired effect in 50% of the population to which it is administered. Although the ED50 is commonly used as a measure of reasonable expectance of an agent’s effect, it is not necessarily the dose that a clinician might deem appropriate taking into consideration all relevant factors. Thus, in some situations the effective amount is more than the calculated EDso, in other situations the effective amount is less than the calculated ED50, and in still other situations the effective amount is the same as the calculated ED50.

In addition, an effective dose of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as provided herein, may be an amount that, when administered in one or more doses to a subject, produces a desired result relative to a healthy subject. For example, for a subject experiencing a particular disorder, an effective dose may be one that improves a diagnostic parameter, measure, marker and the like of that disorder by at least about 5%, at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more than 90%, where 100% is defined as the diagnostic parameter, measure, marker and the like exhibited by a normal subject.

In certain embodiments, the compounds of Formula (I), or a pharmaceutically acceptable salt thereof disclosed herein may be administered (e.g., orally) at dosage levels of about 0.01 mg/kg to about 50 mg/kg, or about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.

For administration of an oral agent, the compositions can be provided in the form of tablets, capsules and the like containing from 1.0 to 1000 milligrams of the active ingredient, particularly 1.0, 3.0, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 75.0, 100.0, 150.0, 200.0, 250.0, 300.0, 400.0, 500.0, 600.0, 750.0, 800.0, 900.0, and 1000.0 milligrams of the active ingredient.

In certain embodiments, the dosage of the compound of Formula (I), or a pharmaceutically acceptable salt thereof is contained in a “unit dosage form”. The phrase “unit dosage form” refers to physically discrete units, each unit containing a predetermined amount of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, either alone or in combination with one or more additional agents, sufficient to produce the desired effect. It will be appreciated that the parameters of a unit dosage form will depend on the particular agent and the effect to be achieved.

EXAMPLES

The following examples illustrate in the invention. These examples are not intended to limit the scope of the present disclosure, but rather to provide guidance to the skilled artisan to prepare and use the compounds, compositions, and methods of the present disclosure. While particular embodiments of the present disclosure are described, the skilled artisan will appreciate that various changes and modifications can be made without departing from the spirit and scope of the invention. Unless otherwise noted, reagents are commercially available or are prepared according to procedures in the literature. The symbols and conventions used in the descriptions of processes, schemes, and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry.

Preparative HPLC / MS was performed using mass directed auto purification (MDAP) chromatography. HPLC column commonly used was Acquity UPLC CSH C18 column (30 mm x 2. 1 mm i.d. 1.7 pm packing diameter) at 45 °C. HPLC solvents used were: acidic - 0.1% v/v solution of formic acid in MeCN / 0.1% v/v solution of formic acid in H2O (0%-100% gradient); basic - MeCN / 10 mM ammonium bicarbonate in H2O adjusted to pH = 10 with NH3 (0%-100% gradient).

Chemical shifts are expressed in parts per million (ppm) units. Coupling constants (J) are in units of hertz (Hz). Splitting patterns describe apparent multiplicities and are designated as s (single), d (double), t (triplet), dd (double doublet), dt (double triplet), dq (double quartet), m (multiplet), br (broad).

Flash column chromatography was performed on silica gel.

The naming programs used are ACDLABs 11.0 Namebatch, ACD IUPAC, or ChemDraw.

Intermediates

Intermediate 1: 6-(3,4-dimethylphenyl)-2-oxo-l,2-dihydropyridine-3-carboxyli c acid

Step 1: methyl 6-(3,4-dimethylphenyl)-2 -methoxynicotinate methyl 6-bromo-2 -methoxynicotinate (250 mg, 1.0 mmol), (3,4-dimethylphenyl)boronic acid (150 mg, 1.0 mmol), K2CO3 (280 mg, 2.0 mmol) and APhos Pd G3 (65 mg, 0.10 mmol) were placed into a flask vial equipped with a magnetic stir bar and dissolved in THF (4 mL) and H2O (1 mL). The reaction was heated to 80°C and stirred for 17 h then cooled and poured into brine and extracted twice with EtOAc. The combined organic layer was dried over Na2SC>4, filtered, and concentrated. The crude material was purified by normal phase chromatography (EtOAc / heptane; 0% for 1 min, 0-25% over 17 min, 25% for 2 min). The fractions containing desired product were combined and concentrated to afford methyl 6-(3,4-dimethylphenyl)-2- methoxynicotinate (250 mg, 0.93 mmol, 92% yield) as a white solid. J H NMR (400 MHz, DMSO-de) 5 ppm 8.19 (d, J = 7.8 Hz, 1H), 7.95 (br s, 1H), 7.90 (dd, J = 7.8, 1.5 Hz, 1H), 7.65 (d, J = 7.8 Hz, 1H), 7.28 (d, J = 8.3 Hz, 1H), 4.04 (s, 3H), 3.81 (s, 3H), 2.32 (s, 3H), 2.29 (s, 3H). ES-LCMS m/z 272.1 [M+H] + .

Step 2: 6-(3,4-dimethylphenyl)-2-oxo-l,2-dihydropyridine-3-carboxyli c acid

A solution of methyl 6-(3,4-dimethylphenyl)-2-methoxynicotinate (250 mg, 0.920 mmol) in cone. HC1 (10 mL, 120 mmol) was heated to 100°C. After 3 h, the reaction mixture was cooled to r.t. and filtered to obtain 6-(3,4-dimethylphenyl)-2-oxo-l,2-dihydropyridine-3-carboxyli c acid (210 mg, 95%) as a white solid. 'H NMR (400 MHz, DMSO-de) 5 ppm 14.71 (br s, 1H), 13.36 (br s, 1H), 8.39 (d, J = 7.8 Hz, 1H), 7.70 - 7.64 (m, 1H), 7.61 - 7.54 (m, 1H), 7.32 (d, J = 7.8 Hz, 1H), 7.01 (d, J = 7.8 Hz, 1H), 2.33 - 2.25 (m, 6H). ES-LCMS m/z 244.1 [M+H] + . The following compounds were synthesized in an analogous manner to the preparation described above. The palladium catalyst (used for the Suzuki cross-coupling) and HC1 concentration (used for the hydrolysis) are specified.

Intermediate 33: 2-oxo-6-phenyl-4-(trifluoromethyl)-l,2-dihydropyridine-3 -carboxylic acid

Step 1 : 2-chloro-6-phenyl-4-(trifluoromethyl)nicotinic acid

2,6-dichloro-4-(trifluoromethyl)nicotinic acid (1.0 g, 3.9 mmol), phenylboronic acid (0.56 g, 4.6 mmol), PdC^dppf^QfcChadduct (0.16 g, 0.19 mmol), 1 M K2CO3 (11 mb, 12 mmol) in 1,4-dioxane (12 mb) were added to a flask. The mixture was heated to 100°C for 1 h. The reaction was cooled to r.t., acidified to pH ~2 using 6 N HC1, diluted with EtOAc (20 mb) and washed with H2O (20 mb). The aqueous layer was extracted with EtOAc (3 x 20 mL). The combined organic layer was dried over Na2SO4, concentrated, and purified by reverse phase purification (40 - 80% gradient, MeCN (0.1% formic acid modifier) / H2O (0.1% formic acid modifier), 30 min run). 2-chloro-6-phenyl-4-(trifluoromethyl)nicotinic acid (730 mg, 2.4 mmol, 61 % yield) obtained as a white solid. 1H NMR (400 MHz, DMSO-de) 5 14.62 (br s, 1H), 8.41 (s, 1H), 8.24 - 8.14 (m, 2H), 7.61 - 7.51 (m, 3H). ES-LCMS m/z 302.0 [M+H] + .

Step 2: 2-oxo-6-phenyl-4-(trifluoromethyl)-l,2-dihydropyridine-3 -carboxylic acid

2-chloro-6-phenyl-4-(trifluoromethyl)nicotinic acid (350 mg, 1.2 mmol) and NaOH (460 mg,

12 mmol) were dissolved in DMSO (5 mL) to form a suspension. This mixture was stirred at 110°C. After 2 h, the reaction mixture was cooled to r.t, diluted with H2O (50 mL) and acidified to pH = 2 using 6N HC1. The resulting mixture was extracted with EtOAc (2 x 50 mL). The combined organic washed four times with H2O, dried over Na2SC>4 and concentrated to afford 2-oxo-6-phenyl-4-(trifluoromethyl)-l,2-dihydropyridine-3-car boxylic acid (290 mg, 0.97 mmol, 84 % yield) as a yellow-brown solid. 'H NMR (400 MHz, DMSO-de) 5 13.79 - 12.51 (m, 2H), 7.90 (br s, 2H), 7.57 - 7.48 (m, 3H), 7.16 - 6.83 (m, 1H). ES-LCMS m/z 284.1 [M+H] + .

Intermediate 34: 6-cyclohexyl-2-oxo-l,2-dihydropyridine-3-carboxylic acid

Step 1: methyl 6-(cyclohex-l-en-l-yl)-2 -methoxynicotinate

A round bottom flask was charged with a magnetic stirrer and cyclohex- 1-en-l-ylboronic acid (11.4 g, 90.0 mmol), methyl 6-chloro-2-methoxynicotinate (14.0 g, 69.5 mmol), and APhos Pd G3 (0.706 g, 1.11 mmol) under N2. 1,4-dioxane (261 ml), H2O (87 ml), and K2CO3 (25.0 g, 181 mmol) were added, and the resulting mixture was heated to 100 °C. After 10 min, the mixture was cooled to room temperature and concentrated, and an EtOAc / H2O extraction performed. The combined organic layers were dried over MgSO4, filtered, and concentrated. The crude mixture was then purified via normal phase chromatography (7% EtOAc / heptane) to afford methyl 6-(cyclohex-l-en-l-yl)-2 -methoxynicotinate (18 g, 67 mmol, 97 % yield) as a colorless, oil. 'H NMR (400 MHz, DMSO-de) 5 8.09 (d, J = 8.3 Hz, 1H), 7.16 (d, J = 7.8 Hz, 1H), 7.01 - 6.94 (m, 1H), 3.93 (s, 3H), 3.78 (s, 3H), 2.48 - 2.40 (m, 2H), 2.30 - 2.21 (m, 2H), 1.78 - 1.56 (m, 4H). ES-LCMS m/z 248.2 [M+H] + . Step 2: methyl 6-cyclohexyl-2 -methoxynicotinate

To a solution of methyl 6-(cyclohex-l-en-l-yl)-2 -methoxynicotinate (15.7 g, 63.6 mmol) in MeOH (636 ml) was added Pd/C (10wt%) (6.76 g, 6.36 mmol) and the mixture was stirred at r.t. an atmosphere of H2 gas (balloon) over 16 h. The mixture was filtered through a pre-packed celite filter and concentrated to afford methyl 6-cyclohexyl -2 -methoxynicotinate (14 g, 56 mmol, 88 % yield) as a light-yellow oil. 'H NMR (400 MHz, DMSO-de) 5 8.08 - 8.00 (m, 1H), 6.94 (d, J = 7.8 Hz, 1H), 3.91 (s, 3H), 3.78 (s, 3H), 2.63 (tt, J = 11.6, 3.6 Hz, 1H), 1.94 - 1.64 (m, 5H), 1.57 - 1.43 (m, 2H), 1.36 (qt, J = 12.6, 3.2 Hz, 2H), 1.29 - 1.15 (m, 1H). ES-LCMS m/z 250.2 [M+H] + .

Step 3: 6-cyclohexyl-2-oxo-l,2-dihydropyridine-3 -carboxylic acid

To a 500 mL round bottom flask containing a magnetic stirrer and methyl 6-cyclohexyl -2- methoxynicotinate (14.0 g, 56.0 mmol) was added 12 M HC1 (233 ml, 1.40 mol) and the resulting mixture was heated to 100 °C. After 3 h, the mixture was allowed to cool to room temperature then 0 °C. The resulting suspension was filtered and then air-dried to afford 6- cyclohexyl -2 -oxo- l,2-dihydropyridine-3 -carboxylic acid (11 g, 49 mmol, 88 % yield) as a beige solid. 'H NMR (400 MHz, DMSO-de) 5 14.68 (br s, 1H), 13.16 (br s, 1H), 8.31 (d, J = 7.8 Hz, 1H), 6.56 (d, J = 7.3 Hz, 1H), 2.62 (tt, J = 12.0, 3.2 Hz, 1H), 1.91 - 1.74 (m, 4H), 1.73 - 1.62 (m, 1H), 1.54 - 1.37 (m, 2H), 1.36 - 1.13 (m, 3H). ES-LCMS m/z 222.1 [M+H] + .

The following compounds were synthesized in an analogous manner to the preparation described above. The palladium catalyst (used for the cross-coupling) and HC1 concentration (used for the hydrolysis) are specified.

Intermediate 39: l-methyl-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid

Step 1: methyl 2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylate

2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid (180 mg, 0.81 mmol) was dissolved in DMF (4.5 mL) and NaH (60 wt% in mineral oil) (65 mg, 1.6 mmol) was added. The reaction was stirred at r.t for 15 min and then iodomethane (0.051 mL, 0.81 mmol) was added dropwise. After 1 h, additional iodomethane (0.051 mL, 0.81 mmol) was added and continued stirring at r.t for another 1.5 h. The reaction was diluted with H2O and extracted with EtOAc. The combined organic layers were concentrated and purified by normal phase chromatography (EtOAc / heptane; 0%, 0 - 100% gradient) to afford methyl l-methyl-2-oxo-6-phenyl-l,2- dihydropyridine-3 -carboxylate (85 mg, 0.28 mmol, 34 % yield) as a white solid. J H NMR (400 MHz, DMSO-de) 5 8.05 (d, J = 7.3 Hz, 1H), 7.60 - 7.45 (m, 5H), 6.25 (d, J = 7.3 Hz, 1H), 3.78 (s, 3H), 3.29 (s, 3H). ES-LCMS m/z 244.1 [M+H] + .

Step 2: l-methyl-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid methyl l-methyl-2-oxo-6-phenyl-l,2-dihydropyridine-3 -carboxylate (85 mg, 0.35 mmol) and LiOH (34 mg, 1.4 mmol) were combined in methanol (2 mb) and stirred at r.t. for 2 h. The reaction was then concentrated and diluted with 6N HC1 and stirred and the resulting solid filtered, rinsed with H2O, and dried to afford l-methyl-2-oxo-6-phenyl-l,2-dihydropyridine-3- carboxylic acid (59 mg, 0.23 mmol, 66 % yield) as a white solid. 1 H NMR (400 MHz, DMSO- de) 5 14.72 (s, 1H), 8.41 (d, J = 7.3 Hz, 1H), 7.59 (s, 5H), 6.68 (d, J = 7.8 Hz, 1H), 3.46 (s, 3H). ES-LCMS m/z 230.1 [M+H] + .

Intermediate 40: perfluorophenyl 6-(3 ,5 -difluorophenyl)-2-oxo- 1 ,2-dihydropyridine-3 -

5 carboxylate

6-(3,5-difluorophenyl)-2-hydroxynicotinic acid (100 mg, 0.40 mmol) and pyridine (0.80 ml) were combined in a flask and heated to 40°C. Perfluorophenyl 2,2,2-trifluoroacetate (0. 14 ml, 0.80 mmol) was added dropwise and the reaction was heated for 2.5 h. The reaction was then 0 cooled to r.t. and diluted with H2O. The resulting precipitate was fdtered and dried to afford perfluorophenyl 6-(3,5-difluorophenyl)-2-oxo-l,2-dihydropyridine-3-carboxyla te (140 mg, 0.26 mmol, 66 % yield) as a white solid. ES-LCMS m/z 418.1 [M+H] + .

The following compounds were synthesized in an analogous manner to the preparation described above.

Intermediate 51 : (R)-N-( 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)-6-iodo-2-oxo- 1 ,2- dihydropyridine-3-carboxamide Step 1 : 6-bromo-2 -methoxynicotinic acid

In a round bottom flask equipped with a magnetic stirrer was prepared a solution of methyl 6- bromo-2-methoxynicotinate (19 g, 77 mmol) in THF (130 mb), MeOH (130 mb), and H2O (130 mb). To the resulting mixture was added Li OH (7.4 g, 310 mmol) and the reaction was stirred at r.t. for 15 min. The reaction mixture was subsequently cooled in an ice bath and 1 M HC1 (310 ml, 310 mmol) was added slowly. The resulting suspension was filtered and the collected solid was washed with cold H2O (3 x 100 mb), and then hexane (l x 200 mb). The resulting residue was dried to afford 6-bromo-2 -methoxynicotinic acid (16 g, 69 mmol, 89 % yield) as a white solid. NMR (400 MHz, DMSO-de) 5 13.14 (br s, 1H), 8.03 (d, J = 7.8 Hz, 1H), 7.32 (d, J = 7.8 Hz, 1H), 3.92 (s, 3H). ES-LCMS m/z 231.6 [M+H] + .

Step 2: perfluorophenyl 6-bromo-2 -methoxynicotinate

To a solution of 6-bromo-2-methoxynicotinic acid (17.7 g, 76.3 mmol) in anhydrous pyridine (162 mb) was added perfluorophenyl 2,2,2-trifluoroacetate (26.2 mb, 153 mmol). The reaction mixture was stirred at r.t. for 5 min. The solution was then poured into 600 mb of rapidly stirring H2O and the resulting suspension stirred over 15 min then filtered. The residue was washed with H2O (3 x 100 mb) followed by heptane (2 x 100 mb) and air-dried on the filter to afford perfluorophenyl 6-bromo-2 -methoxynicotinate (24 g, 59 mmol, 78 % yield) as a lightyellow solid. 'H NMR (400 MHz, DMSO-de) 5 8.38 (d, J = 7.8 Hz, 1H), 7.50 (d, J = 7.8 Hz, 1H), 4.01 (s, 3H). ES-LCMS m/z 397.8 [M+H] + .

Step 3: (R)-6-bromo-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2 -methoxynicotinamide

To a stirring solution of (R)-3-amino-2,3-dihydrothiophene 1,1-dioxide, hydrochloride (10.6 g, 62.4 mmol) and DIPEA (34.3 mL, 196 mmol) in anhydrous DMF (39.6 mL) was added a solution of perfluorophenyl 6-bromo-2 -methoxynicotinate (23.7 g, 59.4 mmol) in anhydrous DMF (158 mL) and the reaction mixture stirred at r.t. for 5 min. The resulting solution was poured into rapidly stirring water (600 mL) and the suspension fdtered, washed with H2O (3 x 100 mL) and Et2O (2 x 70 mL), and then air-dried to afford (R)-6-bromo-N-(l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-methoxynicotinamide (15 g, 43 mmol, 72 % yield) as an off-white solid. 'H NMR (400 MHz, DMSO-de) 5 8.78 (d, J = 7.8 Hz, 1H), 7.99 (d, J = 7.8 Hz, 1H), 7.37 (d, J = 7.8 Hz, 1H), 7.23 (dd, J = 6.8, 2.4 Hz, 1H), 6.94 - 6.84 (m, 1H), 5.34 (tdt, J = 7.8, 5.3, 2.5 Hz, 1H), 3.95 (s, 3H), 3.76 (dd, J= 13.5, 8.1 Hz, 1H), 3.28 (dd,J= 13.5, 5.6 Hz, 1H). ES-LCMS m/z 346.9 [M+H] + .

Step 4: (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-hydroxy-6-iod onicotinamide

To a mixture of (R)-6-bromo-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2- methoxynicotinamide (7.53 g, 21.7 mmol) in anhydrous MeCN (217 ml) was added 1 M TMS- I in DCM (65.1 mL, 65.1 mmol). The resulting mixture was stirred at r.t for 16 h. At 16 h, additional 1 M TMS-I in DCM (21.7 ml, 21.7 mmol) was added, and the mixture stirred at r.t. for another 24 h. The reaction mixture was then quenched with MeOH (100 mL), and the resulting suspension concentrated. A mixture of 1: 1: 1 mixture of DCM:Heptane:Et2O (150 mL) was added, and the resulting suspension filtered. The residue was dried to afford (R)-N- (l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-hydroxy-6-iodonicot inamide (9.6 g, 22 mmol, 100 % yield) as a light-yellow solid. 'H NMR (400 MHz, DMSO-de) 5 13.37 (br s, 1H), 9.87 - 9.71 (m, 1H), 7.94 (d, J = 7.8 Hz, 1H), 7.24 (dd, J = 6.8, 2.0 Hz, 1H), 6.99 (br d, J = 7.3 Hz, 1H), 6.95 (dd, J = 6.8, 2.9 Hz, 1H), 5.38 - 5.27 (m, 1H), 3.71 (dd, J = 13.7, 7.8 Hz, 1H), 3.24 (dd, J = 13.9, 4.2 Hz, 1H). ES-LCMS m/z 380.7 [M+H] + . Intermediate 52: 5 -bromo-2-oxo-6-phenyl-l,2-dihydropyridine-3 -carboxylic acid

A solution of NaOBr was prepared by adding Bn (0.27 mb, 5.2 mmol) to a stirred, cooled (0°C) solution of 2M NaOH (7.0 mL, 14 mmol). The mixture was warmed to r.t. and then added to a stirred solution of 2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid (1.0 g, 4.7 mmol) in 2 M NaOH (7.0 mL, 14 mmol). The mixture was stirred for 5 min, placed in an ice bath and acidified to pH = 3 using 6N HC1 (4 mL). The resulting solid was filtered and dried to give 5 -bromo-2-oxo-6-phenyl-l,2-dihydropyridine-3 -carboxylic acid (1.3 g, 4.4 mmol, 95% yield) as a beige solid. 'H NMR (400 MHz, Mcthanol-di) 5 ppm 8.62 (s, 1H), 7.64 - 7.57 (m, 5H), 7.54 (s, 1H). ES-LCMS m/z 295.9 [M+H] + .

Intermediate 53: (R)-5-cyano-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-metho xy-6- phenylnicotinamide

Step 1: methyl 5-bromo-2-methoxy-6-phenylnicotinate

To a stirred solution of methyl 2-methoxy-6-phenylnicotinate (850 mg, 3.5 mmol) in MeCN (15 mL) was added NBS (650 mg, 3.7 mmol). The mixture was heated to 70°C and stirred for 21 h after which it was cooled to r.t. and concentrated. The crude material was purified using normal phase chromatography (EtOAc / hexane, 0%-25%). The appropriate fractions were combined and concentrated afford methyl 5-bromo-2-methoxy-6-phenylnicotinate (1.0 g, 3.1 mmol, 90% yield) as a white solid. 'H NMR (400 MHz, DMSO-de) 5 ppm 8.39 (s, 1H), 7.79 - 7.73 (m, 2H), 7.55 - 7.48 (m, 3H), 3.95 (s, 3H), 3.84 (s, 3H). 1H presumed under water peak. ES-LCMS m/z 323.8 [M+H] + .

Step 2: 5-cyano-2-methoxy-6-phenylnicotinate

A mixture of methyl 5-bromo-2-methoxy-6-phenylnicotinate (500 mg, 1.6 mmol), and zinc cyanide (280 mg, 2.4 mmol) in DMF (10 mL) and H2O (1 mL) was degassed using N2 and bis(tri-t-butylphosphine)palladium(0) (50 mg, 0.098 mmol) was added. The reaction mixture was heated to 100°C and stirred for 6 h then cooled to r.t., poured into H2O and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over Na2SC>4, filtered and concentrated. The crude material was purified using normal phase chromatography (EtOAc / hexane, 0%-20%). The appropriate fractions were combined and concentrated to afford methyl 5-cyano-2-methoxy-6-phenylnicotinate (300 mg, 1.1 mmol, 71% yield) as a white solid. 'H NMR (400 MHz, DMSO-de) 5 ppm 8.64 (s, 1H), 8.02 - 7.96 (m, 2H), 7.63 - 7.57 (m, 3H), 4.08 (s, 3H), 3.86 (s, 3H). ES-LCMS m/z 269.1 [M+H] + .

Step 3: 5-cyano-2-methoxy-6-phenylnicotinic acid

To a stirred suspension of methyl 5-cyano-2-methoxy-6-phenylnicotinate (100 mg, 0.37 mmol) in THF (2 mL) and MeOH (2 mL) was added IM aqueous LiOH (2 mL, 2.0 mmol) and the mixture stirred at r.t for 2 h. The reaction mixture was then concentrated and the remaining solid dissolved in H2O, acidified to pH = 2 using IN aqueous HC1, and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over Na2SC>4, filtered, and concentrated. The crude material was dried to afford 5-cyano-2-methoxy-6-phenylnicotinic acid (90 mg, 0.35 mmol, 95% yield) as a white solid. 'H NMR (400 MHz, DMSO-de) 5 ppm 13.46 (br s, 1H), 8.58 (s, 1H), 8.02 - 7.95 (m, 2H), 7.63 - 7.58 (m, 3H), 4.07 (s, 3H). ES-LCMS m/z 255.1 [M+H] + .

Step 4: (R)-5-cyano-N-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6- phenylnicotinamide

To a stirred solution of 5-cyano-2-methoxy-6-phenylnicotinic acid (88 mg, 0.35 mmol) and (R)-3-amino-2,3-dihydrothiophene 1,1-dioxide, hydrochloride (65 mg, 0.38 mmol) in DMF (2 mL) was added DIPEA (0.20 mL, 1.1 mmol) followed by a 50% solution of n- propylphosphonic acid anhydride, cyclic trimer in EtOAc (0.30 mL, 0.51 mmol). The mixture was stirred for 3 h and then filtered and dried to afford (R)-5-cyano-N-(l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-methoxy-6-phenylnicotinamide (93 mg, 0.25 mmol, 73% yield) as a beige solid. 'H NMR (400 MHz, DMSO-de) 5 ppm 5 9.00 (d, J = 7.8 Hz, 1H), 8.52 (s, 1H), 7.99 - 7.95 (m, 2H), 7.62 - 7.58 (m, 3H), 7.27 (dd, J = 6.4, 2.0 Hz, 1H), 6.92 (dd, J = 6.4, 2.4 Hz, 1H), 5.37 (tdt, J = 7.8, 5.3, 2.5 Hz, 1H), 4.08 (s, 3H), 3.80 (dd, J = 13.5, 8.1 Hz, 1H). ES- LCMS m/z 370.0 [M+H] + .

Intermediate 54: 2-oxo-6-phenyl-5 -(trifluoromethyl)- l,2-dihydropyridine-3 -carboxylic acid Step 1: methyl 5-iodo-2-methoxy-6-phenylnicotinate

To a stirred solution of methyl 2-methoxy-6-phenylnicotinate (2.0 g, 8.2 mmol) in TFA (10 mL) was added N-iodosuccinimide (1.9 g, 8.2 mmol) at r.t. and the mixture was stirred for 16 h. The mixture was then quenched with H2O (30 mL) and extracted with EtOAc (2 x 30 mL). The combined organic phase was washed with saturated brine solution (20 mL), dried over Na2SC>4, and evaporated in vacuo to give the crude as a brown solid. The crude mixture was purified by normal phase chromatography (0-100% of EtOAc in pet ether over 30 min) and fractions containing product concentrated to obtain methyl 5-iodo-2-methoxy-6- phenylnicotinate (2.8 g, 5.7 mmol, 69 % yield) as an off-white solid. NMR (400 MHz, DMSO-de) 5 8.55 (s, 1H), 7.70 - 7.63 (m, 2H), 7.55 - 7.44 (m, 3H), 3.91 (s, 3H), 3.83 (s, 3H). ES-LCMS m/z 370.0 [M+H] + .

Step 2: methyl 2-methoxy-6-phenyl-5-(trifluoromethyl)nicotinate

To a stirred solution of methyl 5-iodo-2-methoxy-6-phenylnicotinate (500 mg, 1.4 mmol) in N,N-dimethylformamide (DMF) (8 mL) were added methyl 2,2-difluoro-2- (fluorosulfonyl)acetate (0.35 mL, 2.7 mmol) and copper(I) iodide (26 mg, 0.14 mmol) at r.t. and the reaction mixture was heated at 120 °C and stirred for 34 h. The mixture was quenched with H2O (10 mL) and extracted with EtOAc (2 x 15 mL). The combined organic phase was washed with saturated brine solution (8 mL), dried over Na2SO4, and evaporated in vacuo to give a crude mixture. The crude mixture was purified by normal phase chromatography (0- 100% of EtOAc in pet ether over 30 min) and fractions containing product concentrated to obtain methyl 2-methoxy-6-phenyl-5-(trifluoromethyl)nicotinate (250 mg, 0.63 mmol, 46 % yield) was isolated as a pale-yellow gum. J H NMR (400 MHz, DMSO-de) 5 8.49 (s, 1H), 7.58 - 7.48 (m, 5H), 4.00 (s, 3H), 3.87 (s, 3H). ES-LCMS m/z 312.0 [M+H] + .

Step 3: 2-oxo-6-phenyl-5 -(trifluoromethyl)- l,2-dihydropyridine-3 -carboxylic acid

2-oxo-6-phenyl-5-(trifluoromethyl)-l,2-dihydropyridine-3- carboxylic acid was obtained from hydrolysis of methyl 2-methoxy-6-phenyl-5-(trifluoromethyl)nicotinate using HC1 under similar conditions to those already described above. 'H NMR (400 MHz, DMSO-de) 5 8.45 (s, 1H), 7.63 - 7.51 (m, 5H). ES-LCMS m/z 284.0 [M+H] + .

Intermediate 55: 5-methyl-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid

Step 1: methyl 5-bromo-2-methoxy-6-phenylnicotinate

To a stirred solution of methyl 2-methoxy-6-phenylnicotinate (2.0 g, 8.2 mmol) in chloroform (20 mb) was added a solution of bromine (0.64 mb, 12 mmol) in Chloroform (10 mb), drop wise. The mixture was stirred at room temperature for 32 h. . The mixture was quenched with H2O and extracted with EtOAc (2 x 25 mb). The combined organic phase was washed with saturated brine solution (15 mb), dried over Na2SC>4, and evaporated in vacuo to give an off- white crude solid. This solid was purified by normal phase chromatography (0-40% of EtOAc in pet ether over 30 min) and fractions containing product concentrated to obtain methyl 5- bromo-2-methoxy-6-phenylnicotinate (1.5 g, 4.6 mmol, 56 % yield) as an off-white solid 1 H NMR (400 MHz, Chloroform-d) 5 8.42 (s, 1H), 7.84 - 7.76 (m, 2H), 7.51 - 7.44 (m, 3H), 4.06 (s, 3H), 3.93 (s, 3H). ES-LCMS m/z 322.0, 324.0 [M+H] + .

Step 2: methyl 2-methoxy-5-methyl-6-phenylnicotinate

To a solution of methyl 5-bromo-2-methoxy-6-phenylnicotinate (400 mg, 1.2 mmol), cesium carbonate (890 mg, 2.7 mmol) in 1,4-Dioxane (3 mb) and Water (0.60 mL) stirred under nitrogen a was added a solution of 2,4,6-trimethyl-l,3,5,2,4,6-trioxatriborinane (50% in THF) (0.42 mL, 1.5 mmol) . The reaction mixture was degassed for 10 min and PdCh(dppf)- CFLChadduct (51 mg, 0.062 mmol) was added. The reaction vessel was sealed and heated to 100 °C for 1.75 h. The reaction was then cooled to r.t., fdtered, and washed with MeOH (6 mL) and then concentrated. The resulting mixture was extracted with DCM (3 x 6 mL), washed with brine (5 mL). The combined organic layer was evaporated into vacuo to yield crude product. The crude mixture was purified by normal phase chromatography (0-5% of EtOAc in pet ether over 60 min) and fractions containing product concentrated to obtain methyl 2-methoxy-5- methyl-6-phenylnicotinate (230 mg, 0.81 mmol, 66 % yield) as a white solid. J H NMR (400 MHz, DMSO-de) 5 8.08 (s, 1H), 7.68 - 7.62 (m, 2H), 7.54 - 7.43 (m, 3H), 3.92 (s, 3H), 3.83 (s, 3H), 2.31 (s, 3H). ES-LCMS m/z 258.2 [M+H] + .

Step 3: 5-methyl-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid

5 -methyl -2 -oxo-6-phenyl-l,2-dihydropyridine-3 -carboxylic acid was obtained from hydrolysis of methyl 2-methoxy-5-methyl-6-phenylnicotinate using HC1 under similar conditions to those already described above. 'H NMR (400 MHz, DMSO-de) 5 8.37 (s, 1H), 7.58 - 7.50 (m, 5H), 2.08 (s, 3H). ES-LCMS m/z 230.0 [M+H] + .

Intermediate 56: 5-fluoro-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid

Step 1: methyl 2-chloro-5-fluoro-6-phenylnicotinate methyl 2-chloro-5-fluoro-6-phenylnicotinate was obtained from Suzuki coupling conditions similar to those already above using methyl 2,6-dichloro-5-fluoronicotinate and phenylboronic acid. 'H NMR (400 MHz, DMSO-de) 5 8.36 (d, J = 10.5 Hz, 1H), 7.98 - 7.93 (m, 2H), 7.60 - 7.55 (m, 3H), 3.92 (s, 3H). ES-LCMS m/z 266.0 [M+H] + .

Step 2: methyl 5-fluoro-2-methoxy-6-phenylnicotinate

To a solution of methyl 2-chloro-5-fluoro-6-phenylnicotinate (500 mg, 1.9 mmol) in DCM (10 mL) under nitrogen at -15°C was added sodium methoxide (450 mg, 2.1 mmol). The reaction mixture was stirred at r.t. for 16 h. Reaction mixture was quenched with ice cold H2O (40 mL) and diluted with DCM (30 mL). DCM extractions(2 x 30 mL) were performed, and the combined organic phase dried over Na2SC>4 and evaporated in vacuo to give a crude off-white solid. This solid was purified by normal phase chromatography (0-5% of EtOAc in pet ether over 45 min) and fractions containing product concentrated to obtain methyl 5-fluoro-2- methoxy-6-phenylnicotinate (380 mg, 1.4 mmol, 76 % yield) as off white solid. J H NMR (400 MHz, DMSO-de) 5 8.18 - 8.13 (m, 1H), 8.06 - 8.01 (m, 2H), 7.59 - 7.50 (m, 3H), 4.01 (s, 3H), 3.84 (s, 3H). ES-LCMS m/z 262.2 [M+H] + . Step 3: 5-fluoro-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid

5-fluoro-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid was obtained from hydrolysis of 5-fluoro-2-methoxy-6-phenylnicotinate using HC1 under similar conditions to those already described above. 'H NMR (400 MHz, DMSO-de) 5 8.34 (br d, J = 9.5 Hz, 1H), 7.82 - 7.73 (m, 2H), 7.63 - 7.54 (m, 3H). ES-LCMS m/z iM [M+H] + .

Intermediate 57: 5 -methoxy-2-oxo-6-phenyl-l,2-dihydropyridine-3 -carboxylic acid Step 1: methyl 5-bromo-2-methoxy-6-phenylnicotinate

Procedure and characterization information already described in two separate instances above.

Step 2: methyl 2-methoxy-6-phenyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan -2-yl)nicotinate

To a solution of methyl 5-bromo-2-methoxy-6-phenylnicotinate (1.0 g, 3.1 mmol), in 1,4 Dioxane (5 mL) stirred under nitrogen was added solid potassium acetate (0.91 g, 9.3 mmol), bis(pinacolato)diboron (1.2 g, 4.7 mmol) . The reaction mixture was degassed under nitrogen for 5 min and PdC12(dppf)-CH2C12adduct (0.13 g, 0.16 mmol). The reaction mixture was stirred at 100 °C for 5 h. The reaction was then cooled to r.t., fdtered over celite and concentrated. The reside was diluted with brine (6 mL) and extracted with DCM (3 x 8 mL). The organic layer was concentrated to obtain methyl 2-methoxy-6-phenyl-5 -(4, 4, 5, 5 -tetramethyl- 1,3,2- dioxaborolan-2-yl)nicotinate (1.2 g, 1.8 mmol, 60 % yield) as a black oil. Crude used for next step without further purification. ES-LCMS m/z 370.0 [M+H] + .

Step 3: methyl 5-hydroxy-2-methoxy-6-phenylnicotinate

To a solution of methyl 2-methoxy-6-phenyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan -2- yl)nicotinate (1.8 g, 4.9 mmol), in THF (8 mL) and Water (0.53 mL) stirred under nitrogen at r.t. was added solid sodium perborate tetrahydrate (1.1 g, 7.3 mmol). The reaction mixture was stirred at r.t for 16 h. The mixture was then diluted with water (12 mL), extracted with EtOAc (3 x 10 mL), and washed with brine (10 mL). The combined organic layers were concentrated to a crude solid which was purified by normal phase chromatography (0-5% of EtOAc in pet ether over 60 min). Fractions containing product concentrated to obtain methyl 5-hydroxy-2- methoxy-6-phenylnicotinate (310 mg, 1.1 mmol, 23 % yield) was obtained as an off-white solid. Tf NMR (400 MHz, DMSO-de) 5 10.05 (s, 1H), 8.20 (d, J= 7.5 Hz, 2H), 7.81 (s, 1H), 7.51 - 7.36 (m, 3H), 3.93 (s, 3H), 3.81 (s, 3H). ES-LCMS m/z 274.2 [M+H] + .

Step 4: 2,5-dimethoxy-6-phenylnicotinate

To a solution of methyl 5-hydroxy-2-methoxy-6-phenylnicotinate (150 mg, 0.58 mmol), potassium carbonate (120 mg, 0.87 mmol) in Acetonitrile (5 mL) was added methyl iodide (0.109 mL, 1.7 mmol) . The reaction mixture was stirred at r.t overnight. The solvent was then evaporated, the residue diluted with water, extracted with DCM (3 x 4 mL). The organic layer was dried over sodium sulphate and concentrated to obtain methyl 2,5-dimethoxy-6- phenylnicotinate (130 mg, 0.46 mmol, 79 % yield) as an off-white solid (crude). ES-LCMS m/z TIM [M+H] + .

Step 5: 5-methoxy-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid

5-methoxy-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid was obtained from hydrolysis of 2,5-dimethoxy-6-phenylnicotinate using HC1 under similar conditions to those already described above. 'H NMR (400 MHz, DMSO-de) 5 8.34 - 8.25 (m, 1H), 7.71 - 7.63 (m, 2H), 7.54 - 7.46 (m, 3H), 3.78 (s, 3H). ES-LCMS m/z 246.2 [M+H] + .

Intermediate 58: (R)-5-chloro-N-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6- phenylnicotinamide

Step 1: methyl 5-chloro-2-methoxy-6-phenylnicotinate methyl 5-chloro-2-methoxy-6-phenylnicotinate was obtained from methyl 2-methoxy-6- phenylnicotinate using NCS under similar conditions to those already described above. J H NMR (400 MHz, DMSO-de) 5 8.28 (s, 1H), 7.84 - 7.78 (m, 2H), 7.57 - 7.49 (m, 3H), 3.97 (s, 3H), 3.84 (s, 3H). ES-LCMS m/z 278.0 [M+H] + .

Step 2: 5-chloro-2-methoxy-6-phenylnicotinic acid

5-chloro-2-methoxy-6-phenylnicotinic acid was obtained from hydrolysis of methyl 5-chloro- 2-methoxy-6-phenylnicotinate using Li OH under conditions similar to those already described above. 'H NMR (400 MHz, DMSO-de) 5 8.23 (s, 1H), 7.84 - 7.76 (m, 2H), 7.57 - 7.47 (m, 3H), 3.95 (s, 3H). ES-LCMS m/z 264.0 [M+H] + .

Step 3: (R)-5-chloro-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-meth oxy-6- phenylnicotinamide

(R)-5-chloro-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-m ethoxy-6-phenylnicotinamide was obtained from 5-chloro-2-methoxy-6-phenylnicotinic acid using general amidation method B conditions. 'H NMR (400 MHz, DMSO-de) 58.92 (d, J = 8.0 Hz, 1H), 8.21 (s, 1H), 7.82 - 7.77 (m, 2H), 7.56 - 7.49 (m, 3H), 7.28 - 7.23 (m, 1H), 6.93 (dd, J = 6.8, 2.8 Hz, 1H), 5.41 - 5.33 (m, 1H), 3.98 (s, 3H), 3.79 (dd, J = 13.5, 8.0 Hz, 1H), 3.32 - 3.29 (m, 1H). ES- LCMS m/z 379.0 [M+H] + . Intermediate 59: perfluorophenyl 5-chloro-6-cyclohexyl-2-oxo-l,2-dihydropyridine-3- carboxylate Step 1: 5-chloro-6-cyclohexyl-2-oxo-l,2-dihydropyridine-3-carboxylic acid

5-chloro-6-cyclohexyl-2-oxo-l,2-dihydropyridine-3-carboxy lic acid was obtained from 6- cyclohexyl -2 -oxo- l,2-dihydropyridine-3 -carboxylic acid using NCS under similar conditions to those already described above. J H NMR (400 MHz, DMSO-de) 5 8.24 (s, 1H), 3.12 - 3.01 (m, 1H), 1.88 - 1.62 (m, 7H), 1.42 - 1.16 (m, 3H). ES-LCMS m/z 256.0 [M+H] + .

Step 2: perfluorophenyl 5 -chloro-6-cyclohexyl -2 -oxo- l,2-dihydropyridine-3 -carboxylate perfluorophenyl 5-chloro-6-cyclohexyl-2-oxo- l,2-dihydropyridine-3-carboxylate was obtained from 5 -chloro-6-cyclohexyl -2 -oxo- l,2-dihydropyridine-3 -carboxylic acid and perfluorophenyl 2,2,2-trifluoroacetate in accordance with a procedure already described above. 'H NMR (400 MHz, DMSO-de) 5 12.44 (br s, 1H), 8.37 (s, 1H), 3.10 - 2.98 (m, 1H), 1.88 - 1.62 (m, 7H), 1.42 - 1.16 (m, 3H). ES-LCMS m/z 422.0 [M+H] + . Intermediate 60: tert-butyl (R)-(5-((l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamoyl)-6- methoxy-2-phenylpyridin-3-yl)carbamate

Step 1: methyl 5-((tert-butoxycarbonyl)amino)-2-methoxy-6-phenylnicotinate

To a stirred solution of methyl 5-iodo-2-methoxy-6-phenylnicotinate (1.0 g, 2.0 mmol) in 1,4 dioxane (10 mL) was added tert-butyl carbamate (0.36 g, 3.1 mmol), (9,9-dimethyl-9H- xanthene-4,5-diyl)bis(diphenylphosphane) (0.12 g, 0.20 mmol) and cesium carbonate (1.3 g, 4.1 mmol) at r.t. The reaction mixture was purged with nitrogen for 15 min, followed by the addition of diacetoxypalladium (0.023 g, 0.10 mmol) at r.t. The reaction mixture was then stirred at 90°C for 12 h. The mixture was concentrated in vacuo and taken up in 50 mL water and 50 mL ethyl acetate. Layers were separated and the aqueous layer was extracted with ethyl acetate (2 x 40 mL). The combined organic layer was washed with saturated sodium chloride (50 mL) and water (50 mL), dried over sodium sulphate, and concentrated in vacuo to give crude mixture. This crude mixture was purified by normal phase chromatography (0-50% of EtOAc in pet ether over 50 min) and fractions containing product concentrated to obtain methyl 5-((tert-butoxycarbonyl)amino)-2-methoxy-6-phenylnicotinate (560 mg, 1.5 mmol, 74 % yield) as pale yellow solid; NMR (400 MHz, DMSO-de) 5 8.76 (br s, 1H), 8.01 (br s, 1H), 7.79 - 7.70 (m, 2H), 7.52 - 7.39 (m, 3H), 3.96 (s, 3H), 3.83 (s, 3H), 1.38 - 1.27 (m, 9H). ES- LCMS m/z 359.0 [M+H] + .

Step 2: 5-((tert-butoxycarbonyl)amino)-2-methoxy-6-phenylnicotinic acid

5-((tert-butoxycarbonyl)amino)-2-methoxy-6-phenylnicotini c acid was obtained from methyl 5-((tert-butoxycarbonyl)amino)-2-methoxy-6-phenylnicotinate using Li OH under similar conditions as already described above. T H NMR (400 MHz, DMSO-de) 5 13.04 (br s, 1H), 8.73 (br s, 1H), 7.97 (br s, 1H), 7.75 (br d, J = 7.0 Hz, 2H), 7.51 - 7.40 (m, 3H), 3.95 (s, 3H), 1.32 (br s, 9H). ES-LCMS m/z 345.0 [M+H] + .

Step 3: perfluorophenyl 5-((tert-butoxycarbonyl)amino)-2-methoxy-6-phenylnicotinate perfluorophenyl 5-((tert-butoxycarbonyl)amino)-2-methoxy-6-phenylnicotinate was obtained from 5-((tert-butoxycarbonyl)amino)-2-methoxy-6-phenylnicotinic acid and perfluorophenyl 2,2,2-trifluoroacetate in accordance with a procedure already described above. ES-LCMS m/z 511.0 [M+H] + .

Step 4: tert-butyl (R)-(5-((l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamoyl)-6-m ethoxy-2- phenylpyridin-3 -yl)carbamate tert-butyl (R)-(5-((l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamoyl)-6-m ethoxy-2- phenylpyridin-3-yl)carbamate was obtained from perfluorophenyl 5 -((tert- butoxycarbonyl)amino)-2-methoxy-6-phenylmcotinate using general amidation method B conditions. 'H NMR (400 MHz, DMSO-de) 5 8.84 - 8.71 (m, 2H), 7.97 (br s, 1H), 7.74 (br d, J = 7.0 Hz, 2H), 7.52 - 7.39 (m, 3H), 7.27 - 7.21 (m, 1H), 6.94 (dd, J = 7.0, 2.5 Hz, 1H), 5.41 - 5.33 (m, 1H), 3.99 (s, 3H), 3.77 (dd, J = 13.3, 7.8 Hz, 1H), 1.31 (br s, 9H). ES-LCMS m/z 460.0 [M+H] + .

Intermediate 61: (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-5-formyl-2-oxo- 6-phenyl-l,2- dihydropyridine-3-carboxamide

Step 1: methyl 2-methoxy-6-phenyl-5-vinylnicotinate

A solution of methyl 5-bromo-2-methoxy-6-phenylnicotinate (2.0 g, 6.2 mmol), trifluoro(vinyl)-14-borane, potassium salt (1.2 g, 9.3 mmol), K2CO3 (2.6 g, 19 mmol) in 1,4- Dioxane (20 mL) and Water (2 mL) was degassed for 15 min with nitrogen followed by the addition of PdCh(dppf) (0.454 g, 0.62 mmol). The reaction mixture was heated to 80° C and stirred for 16 h. The reaction mixture was cooled to r.t., filtered, and washed with ethyl acetate (50 mL). The filtrate was diluted with water (90 mL) and extracted with ethyl acetate (3 x 50 mL). The combined organic layer was dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure. This crude mixture was purified by normal phase chromatography (0-8% of EtOAc in pet ether) and fractions containing product concentrated to obtain methyl 2-methoxy-6-phenyl-5-vinylnicotinate (820 mg, 2.6 mmol, 41 % yield) as an off-white solid. 1 H NMR (400 MHz, DMSO-de) 5 8.38 (s, 1H), 7.64 - 7.58 (m, 2H), 7.55 - 7.49 (m, 3H), 6.68 (dd, J = 17.5, 11.0 Hz, 1H), 5.82 (d, J = 17.5 Hz, 1H), 5.31 (d, J = 11.5 Hz, 1H), 3.96 (s, 3H), 3.85 (s, 3H). ES-LCMS z 270.0 [M+H] + .

Step 2: methyl 5-formyl-2-methoxy-6-phenylnicotinate

To a mixture of methyl 2-methoxy-6-phenyl-5-vinylnicotinate (0.82 g, 3.0 mmol) in THF (40 mL) and Water (8.0 mL) was added sodium periodate (2.0 g, 9.1 mmol) followed by the addition of 2.5% osmium tetroxide in tert-butanol (1.2 mL, 0.091 mmol) at r.t. for 1 h. The reaction mixture was diluted with water (80 mL) and extracted with ethyl acetate (2 x 30 mL). The combined organic layer was dried over sodium sulfate, fdtered, and concentrated under reduced pressure. This crude mixture was purified by normal phase chromatography (0-10% of EtOAc in pet ether over 50 min) and fractions containing product concentrated to obtain methyl 5-formyl-2-methoxy-6-phenylnicotinate (490 mg, 1.8 mmol, 59% yield) as an off-white solid. 'H NMR (400 MHz, DMSO-de) 5 9.89 (s, 1H), 8.58 (s, 1H), 7.76 - 7.72 (m, 2H), 7.62 - 7.56 (m, 3H), 4.09 (s, 3H), 3.87 (s, 3H). ES-LCMS m/z 1112 [M+H] + .

Step 3: 5-formyl-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid

5 -formyl -2 -oxo-6-phenyl-l,2-dihydropyridine-3 -carboxylic acid was obtained from hydrolysis of methyl 5-formyl-2-methoxy-6-phenylnicotinate using HC1 under similar conditions to those already described above. 'H NMR (400 MHz, DMSO-de) 5 13.66 (br s, 1H), 9.48 (s, 1H), 8.63 (s, 1H), 7.73 - 7.55 (m, 6H). ES-LCMS m/z 244.0 [M+H] + .

Step 4: (R)-N-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-5-formyl-2-oxo-6-phenyl- l,2- dihydropyridine-3-carboxamide (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-5-formyl-2-oxo- 6-phenyl-l,2- dihydropyridine-3 -carboxamide was obtained from 5-formyl-2-oxo-6-phenyl-l,2- dihydropyridine -3 -carboxylic acid using general amidation method C conditions. J H NMR (400 MHz, DMSO-de) 5 13.19 (br s, 1H), 10.13 (br s, 1H), 9.47 (s, 1H), 8.67 (s, 1H), 7.68 - 7.51 (m, 5H), 7.27 (dd, J = 6.5, 2.0 Hz, 1H), 6.98 (dd, J = 6.5, 3.0 Hz, 1H), 5.43 - 5.34 (m, 1H), 3.79 - 3.70 (m, 1H), 3.27 (dd, J = 13.8, 4.3 Hz, 1H). ES-LCMS m/z 359.2 [M+H] + .

Intermediate 62: 4-hydroxy-2-oxo-6-phenyl-l,2-dihydropyridine-3 -carboxylic acid

Step 1 : methyl 4-chloro-6-phenylnicotinate methyl 4-chloro-6-phenylnicotinate was obtained from Suzuki coupling conditions similar to those already above using methyl 4,6-dichloronicotinate and phenylboronic acid. 'H NMR (400 MHz, DMSO-de) 5 9.05 (s, 1H), 8.27 (s, 1H), 8.24 - 8.17 (m, 2H), 7.58 - 7.45 (m, 3H), 3.91 (s, 3H). ES-LCMS m/z 248.0 [M+H] + .

Step 2: 4-chloro-5-(methoxycarbonyl)-2-phenylpyridine 1-oxide

To a solution of methyl 4-chloro-6-phenylnicotinate (4.5 g, 18 mmol) in DCM (300 mb) stirred in air at 20 °C was added solid m-CPBA (13 g, 73 mmol). The reaction mixture was stirred at 40 °C for 16 h. Reaction was then diluted with 9: 1 DCM:MeOH (100 mb). The organic phase was washed with water (50 mL), saturated bnne (25 mL), dried over sodium sulphate, and evaporated in vacuo to give the crude product. This crude mixture was purified by normal phase chromatography (0-60% of EtOAc in pet ether) and fractions containing product concentrated to obtain 4-chloro-5-(methoxycarbonyl)-2 -phenylpyridine 1-oxide (3.2 g, 11 mmol, 58 % yield) as a yellow solid. ES-LCMS m/z 264.0 [M+H] + .

Step 3: methyl 4-acetoxy-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylate

A solution of 4-chloro-5 -(methoxy carbonyl)-2-phenylpyridine 1-oxide (2.2 g, 8.3 mmol) in acetic anhydride (20 mL) was stirred at 140 °C for 16 h. The reaction was cooled down to r.t. and diluted with 9: 1 DCM:MeOH (25 mL total).The organic phase was washed with water (10 mL), saturated brine (10 mL), dried over sodium sulphate, and evaporated in vacuo to give a crude product. This crude mixture was purified by normal phase chromatography (0-20% of EtOAc in pet ether) and fractions containing product concentrated to obtain methyl 4-acetoxy- 2 -oxo-6-phenyl-l,2-dihydropyridine-3 -carboxylate (750 mg, 2.4 mmol, 29% yield) as a yellow gum. ES-LCMS m/z 288.0 [M+H] + .

Step 4: 4-hydroxy-2-oxo-6-phenyl-l,2-dihydropyridine-3 -carboxylic acid

OH O cA '"

4-hydroxy-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylic acid was obtained from methyl 4-acetoxy-2-oxo-6-phenyl-l,2-dihydropyridine-3-carboxylate using Li OH under similar conditions as already described above. 1 H NMR (400 MHz, DMSO-de) 5 8.51 (s, 1H), 8.27 (br s, 1H), 7.86 - 7.73 (m, 2H), 7.50 - 7.32 (m, 3H). ES-LCMS m/z 232.0 [M+H] + . Intermediate 63: tert-butyl (R)-(5-((l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamoyl)-6-o xo-

2-phenyl- 1 ,6-dihydropyridin-3 -yl)(methyl)carbamate tert-butyl (R)-(5-(( 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)carbamoyl)-6-oxo-2-phenyl- 1,6- dihydropyridin-3-yl)(methyl)carbamate was obtained from an amination then amidation protocol similar to a close intermediate analog already described above. J H NMR (400 MHz, DMSO-de) 5 8.83 (br d, J = 7.5 Hz, 1H), 8.06 (s, 1H), 7.67 - 7.60 (m, 2H), 7.54 - 7.41 (m, 3H), 7.24 (dd, J= 6.8, 2.3 Hz, 1H), 6.94 (dd, J = 6.5, 2.5 Hz, 1H), 5.42 - 5.34 (m, 1H), 4.01 (s, 3H), 3.78 (dd, J= 13.5, 8.0 Hz, 1H), 3.37 - 3.33 (m, 1H), 0.95 (s, 7H) (Boc group integrated low). ES-LCMS m/z 474.1 [M+H] + .

Intermediate 64: (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2,4-dimethoxy-6 - phenylnicotinamide

Step 1 : methyl 2,4-dichloro-6-phenylnicotinate

To a stirred solution of 4-chloro-5 -(methoxy carbonyl)-2 -phenylpyridine 1-oxide (1.0 g, 3.8 mmol) in DCM (10 mL) were added EtsN (1.9 mL, 13 mmol) and oxalyl chloride (1.2 mb, 13 mmol) at 0 °C. The mixture was allowed to warm up to r.t. and stirred for 16 h. The reaction mixture was then diluted with DCM (10 mL) and the organic phase washed with saturated sodium bicarbonate solution (2 x 10 mL), brine (2 x 10 mL), brine again (10 mL), dried over sodium sulphate, and concentrated in vacuo to give the crude product. This crude mixture was

-I l l- purified by normal phase chromatography (0-15% of EtOAc in pet ether, 40 min) and fractions containing product concentrated to obtain methyl 2,4-dichloro-6-phenylnicotinate (2.1 g, 6.9 mmol, 180 % yield) formed as a brown solid (yield >100% because crude was combined with another batch before purification). 'H NMR (400 MHz, DMSO-de) 5 8.40 - 8.37 (m, 1H), 8.17 - 8.11 (m, 2H), 7.59 - 7.51 (m, 3H), 4.00 - 3.95 (m, 3H). ES-LCMS m/z 282.0 [M+H] + .

Step 2: methyl 2-chloro-4-methoxy-6-phenylnicotinate

To a stirred solution of methyl 2,4-dichloro-6-phenylnicotinate (1.1 g, 3.9 mmol) in DCM (3 mL) was added 5% sodium methoxide in MeOH (0.94 mL, 4.7 mmol) at 0 °C and the mixture was stirred for 16 h at r.t. The mixture was quenched with water (10 mL) and extracted with DCM (2 x 10 mL). The organic phase was washed with water (2 x 10 mL), brine (10 mL), dried over sodium sulphate and concentrated in vacuo to give a crude mixture. This crude mixture was purified by normal phase chromatography (0-20% of EtOAc in pet ether, 40 min) and fractions containing product concentrated to obtain methyl 2-chloro-4-methoxy-6- phenylnicotinate (380 mg, 1.3 mmol, 32 % yield) as a colorless solid. 'H NMR (400 MHz, DMSO-de) 5 8.21 - 8.14 (m, 2H), 7.88 - 7.85 (m, 1H), 7.57 - 7.48 (m, 3H), 4.03 (s, 3H), 3.89 (s, 3H). ES-LCMS m/z 278.0 [M+H] + .

Step 3 : 2,4-dimethoxy-6-phenylnicotinic acid

A solution of methyl 2-chloro-4-methoxy-6-phenylnicotinate (320 mg, 1.2 mmol) and sodium methoxide in MeOH (1.5 mL, 7.5 mmol) was stirred at 60 °C for 16 h. The mixture was quenched with water (10 mL) and washed with EtOAc (2 xlO mL). The organic phase was washed with brine (10 mL), dried over sodium sulphate and evaporated in vacuo to obtain 2,4- dimethoxy-6-phenylnicotinic acid (240 mg, 0.89 mmol, 77 % yield) as an off-white solid. T H NMR (400 MHz, DMSO-de) 5 13.00 (br s, 1H), 8.18 - 8.12 (m, 2H), 7.54 - 7.42 (m, 3H), 7.35 (s, 1H), 3.99 - 3.94 (m, 6H). ES-LCMS m/z 260.2 [M+H] + .

Step 4: (R)-N-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-2,4-dimethoxy-6-phenylni cotinamide M 11r .C« °°

To a stirred solution of 2,4-dimethoxy N -6-phe 'nylnicotinic acid (250 mg, 0.96 mmol) in acetonitrile (5 mL) was added (R)-3-amino-2,3-dihydrothiophene 1,1-dioxide, hydrochloride (250 mg, 1.4 mmol), N-(chloro(dimethylamino)methylene)-N-methylmethanaminium hexafluorophosphate(V) (540 mg, 1.9 mmol) and 1-methyl-lH-imidazole (0.31 mL, 3.9 mmol). The resulting solution was stirred at 25 °C for 16 h. The reaction was then partially concentrated, fdtered, and the residue washed with water (3 mL) to obtain a crude white solid, (R)-N-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-2,4-dimethoxy-6-phenylni cotinamide (350 mg, 0.93 mmol, 140 % yield) (impurities present but used in the next step as is). ES-LCMS m/z 375.2 [M+H] + .

Intermediate 65: 6-(l-methyl-lH-pyrazol-5-yl)-2-oxo-l,2-dihydropyridine-3-car boxylic acid

Step 1: 2-methoxy-6-(l-methyl-lH-pyrazol-5-yl)nicotinic acid, formic acid salt

2-methoxy-6-(l-methyl-lH-pyrazol-5-yl)nicotinic acid, formic acid salt was obtained via a Suzuki cross-coupling procedure similar to ones described previously using methyl 6-bromo- 2 -methoxynicotinate and 1 -methyl-5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)- 1H- pyrazole. 'H NMR (400 MHz, DMSO-de) 5 8.36 (s, 1H), 7.91 (d, J = 7.8 Hz, 1H), 7.47 (d, J = 2.0 Hz, 1H), 7.37 (d, J = 7.3 Hz, 1H), 6.82 (d, J = 2.0 Hz, 1H), 4.21 (s, 3H), 3.93 (s, 3H). ES- LCMS z 234.1 [M+H] + .

Step 2: 6-(l-methyl-lH-pyrazol-5-yl)-2-oxo-l,2-dihydropyridine-3-car boxylic acid

6-(l-methyl-lH-pyrazol-5-yl)-2-oxo-l,2-dihydropyridine-3- carboxylic acid was obtained from 2-methoxy-6-(l-methyl-lH-pyrazol-5-yl)nicotinic acid using TMS-I using a procedure similar to ones described previously. 'H NMR (400 MHz, DMSO-de) 5 8.14 (d, J = 7.3 Hz, 1H), 7.49 (d, J = 2.0 Hz, 1H), 7.30 - 7.03 (m, 2H), 7.00 (d, J = 7.3 Hz, 1H), 6.73 (d, J = 2.0 Hz, 1H), 4.09 (s, 3H). ES-LCMS m/z 220.0 [M+H] + .

Intermediate 66: N-((R)- 1, l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-(4- methylcyclohex- 1 -en- 1 -yl)nicotinamide

Step 1: methyl 2-methoxy-6-(4-methylcyclohex-l-en-l-yl)nicotinate methyl 2-methoxy-6-(4-methylcyclohex-l-en-l-yl)nicotinate was obtained via a Suzuki crosscoupling procedure similar to ones described previously using methyl 6-chloro-2- methoxynicotinate and 4,4,5 ,5 -tetramethyl -2-(4-methylcyclohex- 1 -en- 1 -yl)- 1 ,3,2- dioxaborolane. 'H NMR (400 MHz, DMSO-de) 58.09 (d, J= 8.0 Hz, 1H), 7.17 (d, J= 8.0 Hz, 1H), 6.98 - 6.92 (m, 1H), 3.93 (s, 3H), 3.78 (s, 3H), 2.64 - 2.54 (m, 1H), 2.45 - 2.31 (m, 2H), 1.93 - 1.79 (m, 2H), 1.77 - 1.62 (m, 1H), 1.37 - 1.22 (m, 1H), 0.99 (d, J= 6.5 Hz, 3H). ES- LCMS m/z 262.2 [M+H] + .

Step 2: 2-methoxy-6-(4-methylcyclohex-l-en-l-yl)nicotinic acid

2-methoxy-6-(4-methylcyclohex-l-en-l-yl)nicotinic acid was obtained via a Li OH hydrolysis procedure similar to ones described previously using methyl 2-methoxy-6-(4-methylcyclohex- l-en-l-yl)nicotinate. 'H NMR (400 MHz, DMSO-de) 5 8.06 (d, J= 8.0 Hz, 1H), 7.13 (d, J = 8.0 Hz, 1H), 6.95 - 6.89 (m, 1H), 3.91 (s, 3H), 2.44 - 2.30 (m, 2H), 1.92 - 1.78 (m, 2H), 1.76 - 1.61 (m, 1H), 1.37 - 1.11 (m, 2H), 0.99 (d, J= 6.5 Hz, 3H). ES-LCMS m/z 248.2 [M+H] + . Step 3 : N-((R)- 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-(4-methylcycl ohex- 1 -en- 1 -yl)nicotinamide

N-((R)- 1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)-2-methoxy-6-(4-methylcyclohex- 1 -en- 1 - yl)nicotinamide was obtained via aHATU amide coupling procedure similar to ones described previously using 2-methoxy-6-(4-methylcyclohex-l-en-l-yl)nicotinic acid. 'H NMR (400 MHz, DMSO-de) 58.66 (d, J= 8.0 Hz, 1H), 8.11 (d, J= 8.0 Hz, 1H), 7.23 - 7.19 (m, 2H), 6.95 - 6.90 (m, 2H), 5.40 - 5.32 (m, 1H), 3.98 (s, 3H), 3.74 (dd, J= 13.3, 7.8 Hz, 1H), 3.37 - 3.33 (m, 1H), 2.64 - 2.53 (m, 1H), 2.41 - 2.30 (m, 2H), 1.91 - 1.79 (m, 2H), 1.76 - 1.62 (m, 1H), 1.37 - 1.22 (m, 1H), 0.99 (d, J= 6.5 Hz, 3H). ES-LCMS m/z 363.2 [M+H] + .

Intermediate 67: (R)-4-amino-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-metho xy-6- phenylnicotinamide

Step 1: 4-azido-5-(methoxycarbonyl)-2 -phenylpyridine 1 -oxide

To a solution of 4-chloro-5 -(methoxy carbonyl)-2 -phenylpyridine 1 -oxide (4.0 g, 14 mmol) in DMSO (20 mL) stirred under nitrogen at r.t. was added sodium azide (2.7 g, 41 mmol). The reaction mixture was heated to 55 °C for 4 h. The mixture was diluted with water (100 mL) and ethyl acetate (80 mL). The layers were separated, and the aqueous layer washed with ethyl acetate (2 x 40 mL). The combine organic layer was washed with brine (50mL) and water (50mL), dried over sodium sulphate and concentrated in vacuo to give a crude product. This crude mixture was purified by normal phase chromatography (0-100% of EtOAc in pet ether, 65 min) and fractions containing product concentrated to obtain 4-azido-5 -(methoxy carbonyl)- 2 -phenylpyridine 1-oxide (1.6 g, 5.7 mmol, 42 % yield) as a pale yellow solid. J H NMR (400 MHz, DMSO-de) 5 8.58 (s, 1H), 7.96 - 7.89 (m, 2H), 7.63 (s, 1H), 7.56 - 7.51 (m, 3H), 3.88 (s, 3H). ES-LCMS m/z 271.0 [M+H] + .

Step 2: methyl 4-azido-2-chloro-6-phenylnicotinate methyl 4-azido-2-chloro-6-phenylnicotinate was obtained via an oxalyl chloride procedure similar to ones described previously using 4-azido-5-(methoxycarbonyl)-2-phenylpyridine 1- oxide. Tf NMR (400 MHz, DMSO-de) 5 8.17 - 8.11 (m, 2H), 7.99 (s, 1H), 7.57 - 7.52 (m, 3H), 3.91 (s, 3H). ES-LCMS m/z 289.0 [M+H] + . Step 3 : methyl 4-azido-2-methoxy-6-phenylmcotinate methyl 4-azido-2-methoxy-6-phenylnicotinate was obtained via NaOMe alkoxylation procedure similar to ones described previously using methyl 4-azido-2-chloro-6- phenylnicotinate. The desire product was contaminated with unreacted methyl 4-azido-2- chloro-6-phenylnicotinate and used in the next step as is. NMR was a complex mixture of desired product and impurities. 'H NMR (400 MHz, DMSO-de) 58.21 — 8.10 (m, 6H), 7.99 (s, 2H), 7.59 - 7.48 (m, 10H), 4.01 - 3.98 (m, 2H), 3.93 - 3.88 (m, 6H), 3.84 - 3.81 (m, 2H). Intractable LCMS.

Step 4: 4-azido-2-methoxy-6-phenylnicotinic acid

4-azido-2-methoxy-6-phenylnicotinic acid was obtained via a LiOH hydrolysis procedure similar to ones described previously using methyl 4-azido-2-methoxy-6-phenylnicotinate. 'H NMR (400 MHz, DMSO-de) 5 13.39 (br s, 1H), 8.17 (dd, J= 8.0, 1.5 Hz, 2H), 7.58 - 7.44 (m, 4H), 4.00 (s, 3H). ES-LCMS m/z 271.0 [M+H] + .

Step 5 : (R)-4-azido-N-( 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6- phenylnicotinamide

(R)-4-azido-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-me thoxy-6-phenylnicotinamide was obtained via a HATU amide coupling procedure similar to ones described previously using 4- azido-2-methoxy-6-phenylnicotinic acid. J H NMR (400 MHz, DMSO-de) 5 9.09 (d, J = 7.5 Hz, 1H), 8.20 - 8.14 (m, 2H), 7.55 (s, 4H), 7.26 (dd, J= 6.5, 2.0 Hz, 1H), 6.86 (dd, J= 6.5, 2.5 Hz, 1H), 5.32 - 5.24 (m, 1H), 3.98 (s, 3H), 3.85 (dd, J= 13.8, 8.3 Hz, 1H), 3.05 (dd, J= 14.0, 4.5 Hz, 1H). ES-LCMS m/z 360.2 [M-2N+2H+H] + .

Step 6: (R)-4-amino-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-metho xy-6- phenylnicotinamide

To a stirred solution of (R)-4-azido-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-metho xy-6- phenylnicotinamide (220 mg, 0.18 mmol) in THF (10 mL) and water (0.1 mL) was added triphenylphosphine (48 mg, 0.18 mmol) at r.t. and the reaction mixture was stirred for 12 h. The mixture material was extracted using water (30 mL) and ethyl acetate (3 x 30 mL The combined organic phase was washed with brine (20 mL), water (20 mL), and then dried over sodium sulphate and concentrated in vacuo to obtain (R)-4-amino-N-(l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-methoxy-6-phenylnicotinamide (210 mg, 0.070 mmol, 38 % yield) as a pale yellow solid. No purification performed; crude used for next step. ES-LCMS m/z 360.0 [M+H] + .

Intermediate 69: (R)-6-(4,5-dimethylthiophen-2-yl)-N-(l,l-dioxido-2,3-dihydro thiophen-3- yl) -2-methoxynicotinamide

(R)-6-(4,5-dimethylthiophen-2-yl)-N-(l,l-dioxido-2,3-dihy drothiophen-3-yl)-2- methoxynicotinamide was obtained from 6-(4,5-dimethylthiophen-2-yl)-2-methoxynicotinic acid using protocols similar to those already described herein. 6-(4,5-dimethylthiophen-2-yl)- 2-methoxynicotinic acid characterization data: J H NMR (400 MHz, DMSO-de) 5 12.82 (br s, 1H), 8.09 (d, J= 7.8 Hz, 1H), 7.59 (s, 1H), 7.41 (d, J= 7.8 Hz, 1H), 3.94 (s, 3H), 2.35 (s, 3H), 2.13 (s, 3H). ES-LCMS m/z 264.0 [M+H] + . (R)-6-(4,5-dimethylthiophen-2-yl)-N-(l,l- dioxido-2,3-dihydrothiophen-3-yl)-2-methoxynicotinamide characterization data: 'H NMR (400 MHz, DMSO-de) 5 8.69 - 8.62 (m, 1H), 8.13 (d, J= 7.8 Hz, 1H), 7.60 (s, 1H), 7.48 (d, J = 7.8 Hz, 1H), 7.22 (dd, J = 6.6, 2.2 Hz, 1H), 6.95 - 6.91 (m, 1H), 5.36 (tdt, J= 7.8, 5.3, 2.6 Hz, 1H), 4.00 (s, 3H), 3.75 (dd, J= 13.2, 7.8 Hz, 1H), 3.36 - 3.32 (m, 1H), 2.35 (s, 3H), 2.13 (s, 3H). ES-LCMS m/z 379.0 [M+H] + .

Intermediate 68: (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-(th ieno[3,2- b]thiophen-2-yl)nicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6- (thieno[3,2-b]thiophen-2- yl)nicotinamide was obtained from corresponding carboxylic acid using protocols similar to those already described herein. *HNMR (400 MHz, DMSO-de) 58.71 (d, J= 7.8 Hz, 1H), 8.28 (s, IH), 8.19 (d,J= 7.8 Hz, 1H), 7.79 (d, J = 5.4 Hz, 1H), 7.68 (d, J = 7.8 Hz, 1H), 7.52 - 7.47 (m, 1H), 7.23 (dd, J= 6.8, 2.4 Hz, 1H), 6.94 (dd, J= 6.6, 2.7 Hz, 1H), 5.43 - 5.32 (m, 1H), 4.05 (s, 3H), 3.77 (dd, J= 13.7, 7.8 Hz, 1H), 3.37 - 3.32 (m, 1H). ES-LCMS m/z 407.0 [M+H] + .

Intermediate 69: (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-5-hydroxy-2-met hoxy-6-(4-

(trifluoromethyl)phenyl)nicotinamide Step 1: methyl 6-chloro-2-methoxy-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan -2-yl)mcotinate

Combined methyl 6-chloro-2 -methoxynicotinate (13 g, 65 mmol) and bispin (25 g, 98 mmol) in 1,4-Dioxane (150 mL) and degassed under nitrogen for 10 minutes. Added 4,4'-di- tert-butyl-2,2'-bipyridine (3.5 g, 13 mmol) and di-mu-methoxobis(l,5- cyclooctadiene)diiridium(I) (2.0 g, 3.0 mmol) and heated reaction mixture to 100 °C under nitrogen overnight. After 19 h, cooled reaction mixture to r.t. and diluted with water and EtOAc (150 mL each) and stirred for 10 min before separating layers. The aqueous layer was extracted with EtOAc (100 mL), and the combined organic layers were passed through a hydrophobic frit and concentrated down. To the resulting solid was added DCM and a suspension formed. This mixture was filtered, and the residue stirred in hexanes (300 mL) for 20 min and filtered again, finally, the remaining residue was dried under vacuum to afford methyl 6-chloro-2- methoxy-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)nicot inate (7.3 g, 21 mmol, 33 % yield) as a brown solid. NMR (400 MHz, DMSO-de) 5 8.36 (s, 1H), 3.96 (s, 3H), 3.82 (s, 3H), 1.31

Step 2: methyl 6-chloro-5 -hydroxy-2 -methoxynicotinate

Combined methyl 6-chloro-2-methoxy-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan -2- yl)nicotinate (7.3 g, 22 mmol) in tetrahydrofiiran (20 mL) and water (20 mL). Added sodium perborate tetrahydrate (5.2 g, 34 mmol) to the stirred mixture and continued stirring overnight at r.t. After 18 h, diluted the reaction mixture with water and EtOAc (100 mL each), stirred for 15 min, and separated layers. The aqueous layer was extracted with EtOAc (100 mL). The combined organics were passed through a hydrophobic frit, concentrated down and purified by normal phase chromatography (EtOAc / Heptane, 0-100 % gradient, 40 min run) . The fractions containing desired product were combined and concentrated to afford methyl 6-chloro-5- hydroxy-2-methoxynicotinate (7.0 g, 22 mmol, 100 % yield) as a very pale-yellow solid containing about 30-50 % by weight of heptane. 'H NMR (400 MHz, DMSO-de) 5 10.41 (s, 1H), 7.76 (s, 1H), 3.84 (s, 3H), 3.79 (s, 3H). ES-LCMS m/z 218.0 [M+H] + .

Step 3: methyl 5 -hydroxy-2 -methoxy-6-(4-(trifluoromethyl)phenyl)nicotinate

Combined methyl 6-chloro-5 -hydroxy-2 -methoxynicotinate (1.5 g, 6.8 mmol) and (4- (trifluoromethyl)phenyl)boronic acid (1.9 g, 10 mmol) in tetrahydrofuran (17 mL) and water (8 mL) with potassium phosphate tribasic (2.9 g, 14 mmol) and purged under nitrogen for 10 min. Added P(tBu3)Pd G4 (0.20 g, 0.34 mmol) and heated to 60 °C overnight under nitrogen atmosphere. After 19 h, cooled reaction mixture to r.t. and diluted with EtOAc (50 mL) and brine (25 mL) and stirred for 10 min before separating layers. The organic layer was passed through a hydrophobic frit and concentrated down. The resulting residue was stirred in dichloromethane, filtered, and dried under vacuum for 3 days to afford methyl 5 -hydroxy-2 - methoxy-6-(4-(trifluoromethyl)phenyl)nicotinate (1.9 g, 5.5 mmol, 81 % yield) as a light greenish tan solid. 'H NMR (400 MHz, DMSO-de) 5 10.33 (s, 1H), 8.41 (d, J = 8.3 Hz, 2H), 7.87 - 7.79 (m, 3H), 3.94 (s, 3H), 3.82 (s, 3H). ES-LCMS m/z 328.0 [M+H] + .

Step 4: 5 -hydroxy-2 -methoxy-6-(4-(trifluoromethyl)phenyl)nicotinic acid

5-hydroxy-2-methoxy-6-(4-(trifluoromethyl)phenyl)nicotini c acid was obtained via a Li OH hydrolysis procedure similar to ones described previously using methyl 5 -hydroxy-2 -methoxy- 6-(4-(trifluoromethyl)phenyl)nicotinate. 'H NMR (400 MHz, DMSO-de) 5 13.17 - 12.77 (m, 1H), 10.25 (br s, 1H), 8.40 (d, J= 8.3 Hz, 2H), 7.86 - 7.78 (m, 3H), 3.93 (s, 3H). ES-LCMS m/z 314.0 [M+H] + .

Step 5: (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-5-hydroxy-2-met hoxy-6-(4-

(trifluoromethyl)phenyl)nicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-5-hydroxy-2- methoxy-6-(4-

(trifluoromethyl)phenyl)nicotinamide was obtained for a HATU coupling protocol similar to ones already described herein using 5 -hydroxy-2 -methoxy-6-(4-

(trifluoromethyl)phenyl)nicotinic acid. J H NMR (400 MHz, DMSO-de) 5 10.34 (s, 1H), 8.77 (d, J= 7.8 Hz, 1H), 8.40 (d, J= 8.3 Hz, 2H), 7.90 (s, 1H), 7.83 (d, J= 8.3 Hz, 2H), 7.27 - 7.20 (m, 1H), 6.95 (dd, J = 6.6, 2.7 Hz, 1H), 5.38 (tdt, J= 7.8, 5.3, 2.5 Hz, 1H), 4.00 (s, 3H), 3.76 (dd, J= 13.7, 7.8 Hz, 1H), 3.38 - 3.33 (m, 1H).

Intermediate 70: ((R)-6-(5,6-dihydro-4H-cyclopenta[b]thiophen-2-yl)-N-(l,l-di oxido-2,3- dihydrothiophen-3-yl)-2 -methoxynicotinamide) and intermediate 71 ((R)-6-(5,6-dihydro-4H- cyclopenta[b]thiophen-3-yl)-N-(l,l-dioxido-2,3-dihydrothioph en-3-yl)-2- methoxynicotinamide)

Step 1: methyl 6-(5,6-dihydro-4H-cyclopenta[b]thiophen-2-yl)-2 -methoxynicotinate and methyl 6-(5,6-dihydro-4H-cyclopenta[b]thiophen-3-yl)-2 -methoxynicotinate

To a solution of methyl 6-bromo-2 -methoxynicotinate (750 mg, 3.1 mmol), 5,6-dihydro-4H- cyclopenta[b]thiophene-2 -carboxylic acid (770 mg, 4.6 mmol) and CS2CO3 (990 mg, 3.1 mmol) in DMF (8 mL) stirred under nitrogen at r.t. was added bis(tri-tert- butylphoshpine)palladium(O) (62 mg, 0.12 mmol). The reaction vessel was sealed and heated in Biotage Initiator to 170 °C for 10 min. The reaction mixture was cooled and fdtered through a celite bed. The fdtrate was concentrated and purified by normal phase chromatography (EtOAc / Hexanes, 0-100 % gradient, 30 min run). The fractions containing desired product were combined and concentrated to afford a mixture of methyl 6-(5,6-dihydro-4H- cyclopenta[b]thiophen-2-yl)-2 -methoxynicotinate (major product) and methyl 6-(5,6-dihydro- 4H-cyclopenta[b]thiophen-3-yl)-2-methoxynicotinate (minor product) (100 mg, 0.26 mmol, 8.5 % yield) as a pale yellow solid. From this obtained mixture of products, it was inferred that the starting acid commercial material had also likely been a mixture of isomers. The two isomers were close-eluting and carried through the rest of the synthesis in a combined fashion and their separation performed in the last synthesis step. Complex NMR spectrum, overlapping peaks for isomeric mixture. ES-LCMS m/z 290.2 [M+H] + (two isomer peaks, ~3.9: 1 mixture).

Step 2: 6-(5,6-dihydro-4H-cyclopenta[b]thiophen-2-yl)-2-methoxynicot inic acid and 6-(5,6- dihydro-4H-cyclopenta[b]thiophen-3-yl)-2 -methoxynicotinic acid

6-(5,6-dihydro-4H-cyclopenta[b]thiophen-2-yl)-2-methoxyni cotinic acid and 6-(5,6-dihydro-

4H-cyclopenta[b]thiophen-3-yl)-2 -methoxynicotinic acid were obtained via a LiOH hydrolysis procedure similar to ones described previous using the mixture of isomers methyl 6-(5,6- dihydro-4H-cyclopenta[b]thiophen-2-yl)-2-methoxynicotinate and methyl 6-(5,6-dihydro-4H- cyclopenta[b]thiophen-3-yl)-2 -methoxynicotinate. Complex NMR spectrum, overlapping peaks for isomeric mixture. ES-LCMS m/z 276.0 [M+H] + (two isomer peaks, —5.5:1 mixture).

Step 3: (R)-6-(5,6-dihydro-4H-cyclopenta[b]thiophen-2-yl)-N-(l,l-dio xido-2,3- dihydrothiophen-3-yl)-2 -methoxynicotinamide and (R)-6-(5,6-dihydro-4H- cyclopenta[b]thiophen-3-yl)-N-(l,l-dioxido-2,3-dihydrothioph en-3-yl)-2- methoxynicotinamide

(R)-6-(5,6-dihydro-4H-cyclopenta[b]thiophen-2-yl)-N-(l,l- dioxido-2,3-dihydrothiophen-3- yl)-2-methoxynicotinamide and (R)-6-(5,6-dihydro-4H-cyclopenta[b]thiophen-3-yl)-N-(l, 1- dioxido-2,3-dihydrothiophen-3-yl)-2-methoxynicotinamide were obtained for a HATU coupling protocol similar to ones already described herein using 6-(5,6-dihydro-4H- cyclopenta[b]thiophen-2-yl)-2-methoxynicotinic acid and 6-(5,6-dihydro-4H- cyclopenta[b]thiophen-3-yl)-2 -methoxynicotinic acid. Complex NMR spectrum, overlapping peaks for isomeric mixture. ES-LCMS m/z 391.0 [M+H] + (two isomer peaks, —5.3:1 mixture). These isomers were then taken into a deprotection final step (see final compounds synthesis table) after which they were separated and individually characterized.

Intermediate 72: (R)-N-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-6-(l-hydroxycyclohexyl)- 2- methoxynicotinamide Step 1: methyl 6-(l-hydroxycyclohexyl)-2 -methoxynicotinate

A solution of methyl 6-(cyclohex-l-en-l-yl)-2 -methoxynicotinate (0.33 g, 1.3 mmol) and Mn(dpm)s (0.040 g, 0.066 mmol) in DCM (0.82 mL) at 0 °C was evacuated and backfdled with oxygen gas. Next, cold Isopropanol (5.8 mL) was added followed by phenylsilane (0.32 mL, 2.6 mmol) and the resulting mixture was stirred over 4 h at 0 °C under an atmosphere of oxygen gas (balloon). At 4 h, the mixture was quenched with saturated sodium thiosulfate solution (10 mL) and was stirred at room temperature over 16 h. Next, the mixture was combined with additional water and DCM and filtered. The layers were separated, the aqueous layer was extracted with DCM (3 x 10 mL), the combined organic phase passed through a hydrophobic frit and concentrated to afford the crude product mixture. The crude product mixture was purified via normal phase chromatography (15% EtOAc / heptane, isocratic) to afford methyl 6-(l -hydroxy cyclohexyl)-2 -methoxynicotinate (260 mg, 0.85 mmol, 65 % yield) as a thick, transparent, oil. (400 MHz, DMSO-de) 5 8.12 (d, J= 7.8 Hz, 1H), 7.32 (d, J= 7.8 Hz, 1H), 4.99 (s, 1H), 3.92 (s, 3H), 3.79 (s, 3H), 2.00 - 1.85 (m, 2H), 1.79 - 1.60 (m, 3H), 1.58 - 1.48 (m, 4H), 1.32 - 1.21 (m, 1H). ES-LCMS m/z 266.1 [M+H] + .

Step 2: 6-(l -hydroxy cyclohexyl)-2-methoxynicotinic acid

6-(l-hydroxycyclohexyl)-2-methoxynicotinic acid was obtained via a Li OH hydrolysis procedure similar to ones described previously using methyl 6-(l -hydroxy cyclohexyl)-2- methoxynicotinate. 'H NMR (400 MHz, DMSO-de) 5 12.72 (br s, 1H), 8.09 (d, J = 7.8 Hz, 1H), 7.30 (d, J = 7.8 Hz, 1H), 4.96 (s, 1H), 3.91 (s, 3H), 1.98 - 1.87 (m, 2H), 1.78 - 1.60 (m, 3H), 1.57 - 1.46 (m, 4H), 1.31 - 1.21 (m, 1H). ES-LCMS m/z 252.1 [M+H] + . Step 3: (R)-N-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-6-(l-hydroxycyclohexyl)- 2- methoxynicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-6-(l-hydroxy cyclohexyl)-2- methoxynicotinamide was obtained from a perfluorophenyl 2,2,2-trifluoroacetate coupling protocol similar to ones already described herein using 6-(l-hydroxycyclohexyl)-2- methoxynicotinic acid. 'H NMR (400 MHz, DMSO-de) 58.68 (d, J = 7.8 Hz, 1H), 8.10 (d, J = 7.8 Hz, 1H), 7.35 (d, J = 7.8 Hz, 1H), 7.26 - 7.16 (m, 1H), 6.94 - 6.86 (m, 1H), 5.35 (tdt, J = 7.8, 5.3, 2.6 Hz, 1H), 4.97 (s, 1H), 3.96 (s, 3H), 3.75 (dd, J = 13.2, 7.8 Hz, 1H), 3.30 (dd, J = 13.2, 5.4 Hz, 1H), 1.98 - 1.86 (m, 2H), 1.79 - 1.59 (m, 3H), 1.52 (br d, J = 10.8 Hz, 4H), 1.33 - 1.20 (m, 1H). ES-LCMS m/z 367.2 [M+H] + .

Intermediate 73 : (R)-6-(cyclohexyl-di i)-N-( 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)-2- methoxynicotinamide

In an 8 mb glass vial with a magnetic stirrer was placed (R)-6-bromo-N-(l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-methoxynicotinamide (0.20 g, 0.58 mmol), Ni(dtbbpy)(H2O)4Ch (0.013 g, 0.029 mmol), aminosupersilane (0.34 g, 0.86 mmol), [Ir(dF(Me)ppy)2(dtbbpy)]PFe (5.8 mg, 5.8 pmol), and Na2COs (0.12 g, 1.2 mmol) and the solids were purged with nitrogen gas (3x). Next, degassed, anhydrous 1,4-dioxane (5.8 ml) was added followed by 1- bromocyclohexane-l,2,2,3,3,4,4,5,5,6,6-dn (0.14 ml, 1.2 mmol). The resulting mixture was irradiated under blue LED light with stirring and cooling over 16 h (450 nm, Penn PHD reactor). At 16 h, the mixture was diluted with EtOAc (10 mb) and combined with water (10 mL). The phases were separated, and the aqueous phase was extracted with EtOAc (3 x 10 mL). The combined organic portions were dried over MgSC>4, filtered, and concentrated to afford the crude product mixture as a dark yellow oil. The crude product mixture was purified via normal phase chromatography (25-45% EtOAc / heptane) to afford (R)-6-(cyclohexyl- dll)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxynicot inamide (61 mg, 0.17 mmol, 29 % yield) as a white solid. 'H NMR (400 MHz, DMSO-de) 5 8.65 (d, J= 7.8 Hz, 1H), 8.05 (d, J= 7.8 Hz, 1H), 7.21 (dd, J = 6.4, 2.0 Hz, 1H), 6.97 (d, J= 7.8 Hz, 1H), 6.91 (dd, J= 6.6, 2.7 Hz, 1H), 5.35 (tdt, J= 7.8, 5.3, 2.6 Hz, 1H), 3.95 (s, 3H), 3.74 (dd, J= 13.7, 7.8 Hz, 1H), 3.30 (dd, J= 13.2, 5.4 Hz, 1H). ES-LCMS m/z 362.1 [M+H] + .

Intermediate 74: (R)-6-bromo-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-metho xy-5-

(methoxymethoxy)nicotinamide

Step 1: 6-bromo-5 -hydroxy-2 -methoxynicotinate

6-bromo-5-hydroxy-2-methoxynicotinate was obtained from an NBS bromination via a procedure similar to ones described previously using methyl 5-hydroxy-2-methoxynicotinate. 'H NMR (400 MHz, DMSO-de) 5 10.45 (s, 1H), 7.70 (s, 1H), 3.84 (s, 3H), 3.79 (s, 3H). ES- LCMS m/z 262.1 [M+H] + .

Step 2: methyl 6-bromo-2-methoxy-5-(methoxymethoxy)nicotinate

To a solution of methyl 6-bromo-5-hydroxy-2-methoxynicotinate (2.0 g, 7.2 mmol) in anhydrous N,N-Dimethylformamide (DMF) (29 ml) was added K2CO3 (4.0 g, 29 mmol) and the resulting mixture was stirred over 15 min at r.t. Next, MOM-CI (0.82 ml, 11 mmol) was added dropwise, and the resulting mixture was stirred over 45 min at r.t. The reaction mixture was quenched with water (50 mL). Additional water was added (100 mL) followed by diethyl ether (50 mL) and the mixture was transferred to a separatory funnel. The resulting biphasic system was extracted with diethyl ether (3 x 50 mL) and the combined organic portions were combined with hexane (100 mL). The resulting mixture was washed with water (3 x 50 mL), washed once with brine, dried over MgSO-i. fdtered, and concentrated to afford methyl 6- bromo-2-methoxy-5-(methoxymethoxy)nicotinate (2.2 g, 7.1 mmol, 98 % yield) as an off- white solid. 'H NMR (400 MHz, DMSO-de) 5 7.96 (s, 1H), 5.29 (s, 2H), 3.89 (s, 3H), 3.82 (s, 3H), 3.42 (s, 3H). ES-LCMS m/z 306.0 [M+H] + .

Step 3: 6-bromo-2-methoxy-5 -(methoxymethoxy )nicotinic acid

6-bromo-2-methoxy-5-(methoxymethoxy)nicotinic acid was obtained via a Li OH hydrolysis procedure similar to ones described previously using methyl 6-bromo-2-methoxy-5- (methoxymethoxy)nicotinate. 'H NMR (400 MHz, DMSO-de) 5 13.24 (br s, 1H), 7.94 (s, 1H), 5.27 (s, 2H), 3.87 (s, 3H), 3.42 (s, 3H). ES-LCMS m/z 292.0 [M+H] + .

Step 4: (R)-6-bromo-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-metho xy-5-

(methoxymethoxy)nicotinamide

(R)-6-bromo-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-me thoxy-5-

(methoxymethoxy)nicotinamide was obtained from a perfluorophenyl 2,2,2-trifluoroacetate coupling protocol similar to ones already described herein using 6-bromo-2-methoxy-5- (methoxymethoxy)nicotinic acid. 'H NMR (400 MHz, DMSO-de) 5 8.79 (d, J = 7.8 Hz, 1H), 7.96 (s, 1H), 7.23 (dd, J= 6.6, 2.2 Hz, 1H), 6.95 - 6.88 (m, 1H), 5.38 - 5.29 (m, 1H), 5.28 (s, 2H), 3.92 (s, 3H), 3.76 (dd, J= 13.5, 8.1 Hz, 1H), 3.42 (s, 3H), 3.31 (dd, J= 13.7, 5.4 Hz, 1H).

ES-LCMS m/z 407.1 [M+H] + .

Intermediate 75: (R)-6-cycloheptyl-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2 - methoxynicotinamide

(R)-6-cycloheptyl-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl )-2 -methoxynicotinamide was obtained via an Iridium photoredox procedure similar to ones described herein using (R)-6- bromo-N-( 1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)-2 -methoxynicotinamide and bromocycloheptane. 'H NMR (400 MHz, DMSO-de) 5 8.65 (d, J= 7.8 Hz, 1H), 8.04 (d, J = 7.3 Hz, 1H), 7.21 (dd, J= 6.8, 2.4 Hz, 1H), 6.96 (d, J= 7.8 Hz, 1H), 6.91 (dd, J= 6.6, 2.7 Hz, 1H), 5.41 - 5.29 (m, 1H), 3.95 (s, 3H), 3.74 (dd, J= 13.2, 7.8 Hz, 1H), 3.31 (dd, J= 13.7, 5.4 Hz, 1H), 2.90 - 2.77 (m, 1H), 1.93 - 1.44 (m, 12H). ES-LCMS m/z 365.1 [M+H] + .

Intermediate 76: (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-6-(l-fluorocycl ohexyl)-2- methoxynicotinamide

Step 1: methyl 6-(l-fluorocyclohexyl)-2 -methoxynicotinate To a solution of methyl 6-(cyclohex-l-en-l-yl)-2 -methoxynicotinate (1.0 g, 4.1 mmol) in trifluorotoluene (6.4 mL) was added l,l,2,2-tetramethyl-l,2-ethanediamino-N,N'-bis(3,5-di- tert-butylsalicylidene)-cobalt(II) (0.074 g, 0.12 mmol) and l-fluoro-2,4,6-trimethylpyridin-l- ium tetrafluoroborate (1.8 g, 8.1 mmol) followed by another portion of trifluorotoluene (17 mL). The resulting mixture was degassed for 10 min and then cooled to 0 °C at which point 1,1,3,3-tetramethyldisiloxane (2.9 mL, 16 mmol) was added dropwise. The resulting mixture was allowed to stir as it slowly warmed up to r.t. over 16 h. Next, the reaction mixture was diluted with DCM and concentrated to afford the crude product mixture. The crude product mixture was purified via normal phase chromatography on silica gel (7% EtOAc / heptane) to afford a mixture methyl 6-(l-fluorocyclohexyl)-2-methoxynicotinate (520 mg, 1.5 mmol, 38.2 % yield, ~80% wt) containing ~20% by mol of the cyclohexyl byproduct. J H NMR (400 MHz, DMSO-de) 58.18 (d, J= 7.8 Hz, 1H), 7.21 (dd, J= 7.8, 2.0 Hz, 1H), 3.93 (s, 3H), 3.80 (s, 3H), 2.17 - 1.12 (m, 10H). ES-LCMS m/z 268.1 [M+H] + .

Step 2: 6-(l-fluorocyclohexyl)-2 -methoxynicotinic acid

6-(l-fluorocyclohexyl)-2 -methoxynicotinic acid was obtained via a Li OH hydrolysis procedure similar to ones described previously using methyl 6-(l-fluorocyclohexyl)-2- methoxynicotinate. The desired compound contained -20% (by weight) of the cyclohexyl byproduct (carried over from previous step). 'H NMR (400 MHz, DMSO-de) 5 12.87 (br s, 1H), 8.14 (d, J= 7.8 Hz, 1H), 7.17 (dd, J= 7.8, 2.0 Hz, 1H), 3.91 (s, 3H), 2.13 - 1.29 (m, 10H). ES- LCMS m/z 254.1 [M+H] + .

Step 3 : (R)-N-( 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)-6-( 1 -fluorocyclohexyl)-2- methoxynicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-6-(l-fluoroc yclohexyl)-2- methoxynicotinamide was obtained from a perfluorophenyl 2,2,2-trifluoroacetate coupling protocol similar to ones already described herein using 6-(l-fluorocyclohexyl)-2- methoxynicotinic acid. The desired compound contained -20% (by weight) of the cyclohexyl by-product (carried over from previous step). J H NMR (400 MHz, DMSO-de) 5 8.75 (d, J = 7.3 Hz, 1H), 8.16 - 8.10 (m, 1H), 7.24 - 7.21 (m, 2H), 6.91 (dd, J= 6.6, 2.7 Hz, 1H), 5.40 - 5.28 (m, 1H), 3.96 (s, 3H), 3.76 (dd, J = 13.2, 7.8 Hz, 1H), 3.28 (dd, J = 13.5, 5.6 Hz, 1H), 2.14 - 1.29 (m, 10H). ES-LCMS m/z 369.1 [M+H] + .

Intermediate 77: (R)-6-(4,4-difhrorocyclohexyl)-N-(l,l-dioxido-2,3-dihydrothi ophen-3-yl)-2- methoxynicotinamide

In an 8 mL glass vial with a magnetic stirrer was placed (R)-6-bromo-N-(l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-methoxynicotinamide (0.20 g, 0.58 mmol), Ni(dtbbpy)(H2O)4Ch (0.013 g, 0.029 mmol), aminosupersilane (0.34 g, 0.86 mmol), [Ir(dF(Me)ppy)2(dtbbpy)]PFe (5.8 mg, 5.8 pmol), and Na2COs (0.12 g, 1.2 mmol) and the solids were purged with nitrogen gas (3x). Next, degassed, anhydrous 1,4-dioxane (5.8 mL) was added followed by 4-bromo- 1,1 -difluorocyclohexane (0.23 g, 1.2 mmol). The resulting mixture was irradiated under blue LED light with stirring and cooling over 16 h (Penn PHD reactor, 450 nm wavelength, 100% intensity, 800 rpm stirring, cooled to 23 °C). The mixture was then diluted with water and EtOAc and the phases separated. The aqueous portion was extracted with EtOAc (3x) and combined organic portions were dried over MgSCh. fdtered, and concentrated to afford the crude product mixture. The crude product mixture was purified via normal phase chromatography (25-65% EtOAc / heptane, 17 column volumes) to afford (R)-6-(4,4- difluorocyclohexyl)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl) -2 -methoxynicotinamide (92 mg, 0.23 mmol, 41 % yield) as a white solid. J H NMR (400 MHz, DMSO-de) 5 8.68 (d, J = 7.3 Hz, 1H), 8.06 (d, J= 7.3 Hz, 1H), 7.22 (dd, J= 6.8, 2.4 Hz, 1H), 7.03 (d, J= 7.3 Hz, 1H), 6.91 (dd, J= 6.6, 2.7 Hz, 1H), 5.35 (tdt, J= 7.8, 5.3, 2.6 Hz, 1H), 3.95 (s, 3H), 3.75 (dd, J = 13.2, 7.8 Hz, 1H), 3.30 (dd, J = 13.2, 5.4 Hz, 1H), 2.92 - 2.79 (m, 1H), 2.18 - 1.88 (m, 6H), 1.85 - 1.70 (m, 2H). ES-LCMS m/z 387.1 [M+H] + .

Intermediate 78: 6-cyclohexyl-5-hydroxy-2-oxo-l,2-dihydropyridine-3-carboxyli c acid

To a cooled (water ice bath, 5 °C) solution of 6-cyclohexyl-2-oxo-l,2-dihydropyridine-3- carboxylic acid (13 g, 57 mmol) in 10% aq. KOH (180 mL) was added potassium persulfate (27 g, 99 mmol). The resulting mixture was allowed to warm to r.t slowly and stirred for 72 h. The mixture was fdtered, and the filtrate treated with 20% aq. sulfuric acid until pH level of ~6 was obtained. The resulting mixture was cooled on a water/ice bath for 60 min leading to precipitation. More 20% aq. H2SO4 (61 mL) was added, and the mixture stirred vigorously at 100 °C over 16 h. The suspension was cooled to r.t. and then to 5 °C. The suspension was filtered, and the residue washed with water (3x) and then re-suspended in diethyl ether (200 mL) and re-filtered. The final residue was dried to afford 6-cyclohexyl-5 -hydroxy-2 -oxo- 1,2- dihydropyridine-3 -carboxylic acid (8.0 g, 31 mmol, 54 % yield) as a tan solid (containing ~9% starting material by weight by NMR). T H NMR (400 MHz, DMSO-de) 5 15.41 (br s, 1H), 12.83 (br s, 1H), 9.51 (s, 1H), 8.00 (s, 1H), 2.99 (tt, J= 12.2, 3.4 Hz, 1H), 1.92 - 1.12 (m, 10H). ES- LCMS m/z 238.2 [M+H] + .

Intermediate 79 : (R)-N-( 1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)-2-methoxy-6-(2-((tetrahydro- 2H-pyran-4-yl)methoxy)phenyl)nicotinamide

Step 1: methyl 6-(2-hydroxyphenyl)-2 -methoxynicotinate methyl 6-(2-hydroxyphenyl)-2-methoxynicotinate was obtained via a Suzuki coupling protocol similar to ones already described herein using 6-chloro-2-methoxynicotinate and (2- hydroxyphenyl)boronic acid. 'H NMR (400 MHz, DMSO-de) 5 12.00 (s, 1H), 8.27 (d, J= 7.8 Hz, 1H), 8.04 (dd, J = 7.8, 2.0 Hz, 1H), 7.87 (d, J= 8.3 Hz, 1H), 7.35 (ddd, J= 8.4, 7.0, 1.7 Hz, 1H), 7.00 - 6.92 (m, 2H), 4.02 (s, 3H), 3.83 (s, 3H). ES-LCMS m/z 260.1 [M+H] + .

Step 2: methyl 2-methoxy-6-(2-((tetrahydro-2H-pyran-4-yl)methoxy)phenyl)nic otinate

To a solution of methyl 6-(2-hydroxyphenyl)-2-methoxynicotinate (0.16 g, 0.62 mmol), (tetrahydro-2H-pyran-4-yl)methanol (0.072 mb, 0.68 mmol), and 1 M trimethylphosphine solution in THF (0.68 mb, 0.68 mmol) in THF (2.1 mb) cooled to 0 °C was added diisopropyl (E)-diazene-l,2-dicarboxylate (0.13 mL, 0.68 mmol) dropwise. The resulting mixture was stirred on the ice bath for 10 min and then allowed to warm up to r.t. and stirred for 40 h. The mixture was then heated to 60 °C for another 40 h. The mixture was then diluted with diethyl ether and water. The phases were separated, and the aqueous phase was extracted three times with diethyl ether. The combined organic portions were dried over MgSC>4, fdtered, and concentrated to afford the crude product mixture. The crude product mixture was purified via normal phase chromatography (20-30% EtOAc / heptane) to afford methyl 2-methoxy-6-(2- ((tetrahydro-2H-pyran-4-yl)methoxy)phenyl)nicotinate (110 mg, 0.28 mmol, 45 % yield) as a semi-solid. Tf NMR (400 MHz, DMSO-de) 5 8.18 (d, J = 8.3 Hz, 1H), 7.94 (dd, J= 7.8, 1.5 Hz, 1H), 7.66 (d, J= 8.3 Hz, 1H), 7.46 - 7.39 (m, 1H), 7.21 - 7.15 (m, 1H), 7.09 (td, J= 7.6, 1.0 Hz, 1H), 3.99 (s, 3H), 3.95 (d, J= 6.4 Hz, 2H), 3.90 - 3.84 (m, 2H), 3.82 (s, 3H), 3.37 - 3.28 (m, 2H), 2.10 - 1.97 (m, 1H), 1.65 (br dd, J= 12.7, 2.0 Hz, 2H), 1.41 - 1.27 (m, 2H). ES- LCMS m/z 358.2 [M+H] + .

Step 3 : 2-methoxy-6-(2-((tetrahydro-2H-pyran-4-yl)methoxy)phenyl)nic otinic acid methoxy-6-(2-((tetrahydro-2H-pyran-4-yl)methoxy)phenyl)nicot inic acid was obtained via a LiOH hydrolysis protocol similar to ones already described herein using methyl 2-methoxy-6- (2-((tetrahydro-2H-pyran-4-yl)methoxy)phenyl)nicotinate. 'H NMR (400 MHz, DMSO-de) 5 12.83 (s, 1H), 8.15 (d, J= 7.8 Hz, 1H), 7.93 (dd, J= 7.8, 2.0 Hz, 1H), 7.63 (d, J = 7.8 Hz, 1H), 7.42 (ddd, J = 8.3, 7.3, 2.0 Hz, 1H), 7.17 (dd, J= 8.3, 1.0 Hz, 1H), 7.13 - 7.04 (m, 1H), 3.98 (s, 3H), 3.94 (d, J= 6.4 Hz, 2H), 3.86 (dd, J= 11.5, 2.7 Hz, 2H), 3.38 - 3.27 (m, 2H), 2.10 - 1.98 (m, 1H), 1.65 (br dd, J = 12.7, 2.0 Hz, 2H), 1.41 - 1.27 (m, 2H). ES-LCMS m/z 344.2 [M+H] + . Step 4: (R)-N-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-(2-((tetrahy dro-2H- pyran-4-yl)methoxy)phenyl)nicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6- (2-((tetrahydro-2H-pyran-4- yl)methoxy)phenyl)nicotinamide was obtained from a perfluorophenyl 2,2,2-trifluoroacetate coupling protocol similar to ones already described herein using 2-methoxy-6-(2-((tetrahydro- 2H-pyran-4-yl)methoxy)phenyl)nicotinic acid. 'H NMR (400 MHz, DMSO-de) 5 8.76 (d, J = 7.8 Hz, 1H), 8.16 (d, J= 7.8 Hz, 1H), 7.91 (dd, J= 7.6, 1.7 Hz, 1H), 7.68 (d, J= 7.8 Hz, 1H), 7.42 (td, J= 7.8, 1.5 Hz, 1H), 7.23 (dd, J= 6.6, 2.2 Hz, 1H), 7.17 (dd, J= 8.3, 1.0 Hz, 1H), 7.11 - 7.05 (m, 1H), 6.95 (dd, J = 6.6, 2.7 Hz, 1H), 5.38 (tdt, J= 7.8, 5.3, 2.6 Hz, 1H), 4.07 -

3.99 (m, 3H), 3.95 (d, J= 6.4 Hz, 2H), 3.87 (dd, J= 11.2, 2.9 Hz, 2H), 3.77 (dd, J= 13.7, 7.8 Hz, 1H), 3.38 - 3.25 (m, 3H), 2.10 - 1.96 (m, 1H), 1.65 (br dd, J= 13.0, 2.2 Hz, 2H), 1.41 - 1.27 (m, 2H). ES-LCMS m/z 459.1 [M+H] + .

Intermediate 80: (R)-6-(2-(cyclohexylmethoxy)phenyl)-N-( 1, l-dioxido-2,3-dihydrothiophen-

3 -yl) -2 -methoxynicotinamide Step 1 : methyl 6-(2-(cyclohexylmethoxy)phenyl)-2 -methoxynicotinate methyl 6-(2-(cyclohexylmethoxy)phenyl)-2-methoxynicotinate was obtained via a Mitsunobu protocol similar to ones already described herein using methyl 6-(2-hydroxyphenyl)-2- methoxynicotinate. 'H NMR (400 MHz, DMSO-de) 5 8.18 (d, J= 7.8 Hz, 1H), 7.94 (dd, J = 7.8, 2.0 Hz, 1H), 7.68 (d, J = 7.8 Hz, 1H), 7.42 (ddd, J= 8.3, 7.3, 2.0 Hz, 1H), 7.15 (dd, J = 8.3, 1.0 Hz, 1H), 7.07 (td, J= 7.3, 1.0 Hz, 1H), 3.99 (s, 3H), 3.90 (d, J= 5.9 Hz, 2H), 3.82 (s, 3H), 1.84 - 1.57 (m, 6H), 1.36 - 0.97 (m, 5H). ES-LCMS m/z 356.1 [M+H] + .

Step 2: 6-(2-(cyclohexylmethoxy)phenyl)-2 -methoxynicotinic acid

6-(2-(cyclohexylmethoxy)phenyl)-2-methoxynicotinic acid was obtained via a LiOH hydrolysis procedure similar to ones described previously using methyl 6-(2- (cyclohexylmethoxy)phenyl)-2-methoxynicotinate. 'H NMR (400 MHz, DMSO-de) 5 12.83 (br s, 1H), 8.15 (d, J = 7.8 Hz, 1H), 7.93 (dd, J = 7.8, 2.0 Hz, 1H), 7.65 (d, J= 7.8 Hz, 1H), 7.47 - 7.35 (m, 1H), 7.15 (d, J= 8.3 Hz, 1H), 7.07 (td, J= 7.3, 1.0 Hz, 1H), 3.98 (s, 3H), 3.89 (d, J= 5.9 Hz, 2H), 1.87 - 1.58 (m, 6H), 1.35 - 0.97 (m, 5H). ES-LCMS m/z 342.0 [M+H] + . Step 3: (R)-6-(2-(cyclohexylmethoxy)phenyl)-N-(l,l-dioxido-2,3-dihyd rothiophen-3-yl)-2- methoxynicotinamide

(R)-6-(2-(cyclohexylmethoxy)phenyl)-N-(l,l-dioxido-2,3-di hydrothiophen-3-yl)-2- methoxynicotinamide was obtained from a perfluorophenyl 2,2,2-trifluoroacetate coupling protocol similar to ones already described herein using 6-(2-(cyclohexylmethoxy)phenyl)-2- methoxynicotinic acid. 'H NMR (400 MHz, DMSO-de) 5 8.76 (d, J= 7.8 Hz, 1H), 8.16 (d, J = 7.8 Hz, 1H), 7.92 (dd, J= 7.6, 1.7 Hz, 1H), 7.71 (d, J= 7.8 Hz, 1H), 7.47 - 7.37 (m, 1H), 7.23 (dd, J = 6.8, 2.4 Hz, 1H), 7.15 (d, J= 7.8 Hz, 1H), 7.07 (td, J= 7.6, 1.0 Hz, 1H), 6.95 (dd, J= 6.4, 2.4 Hz, 1H), 5.38 (tdt, J= 7.8, 5.3, 2.6 Hz, 1H), 4.03 (s, 3H), 3.89 (d, J= 5.9 Hz, 2H), 3.77 (dd, J= 13.2, 7.8 Hz, 1H), 3.35 (dd, J= 13.2, 5.4 Hz, 1H), 1.85 - 1.60 (m, 6H), 1.34 - 0.99 (m, 5H). ES-LCMS m/z 457.2 [M+H] + .

Intermediate 81 : (R)-N-( 1, l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-( 1- methylcyclohexyl)nicotinamide

Step 1: 2-chloro-6-(l-methylcyclohexyl)nicotinonitrile

To a solution of 1 -methylcyclohexane- 1 -carboxylic acid (2.1 g, 14 mmol), 2- chloronicotinonitrile (1.0 g, 7.2 mmol) and potassium phosphate, dibasic (3.8 g, 22 mmol) in DMSO (3 mL) stirred under nitrogen at r.t. was added (Ir[dF(CF3)ppy]2(dtbpy))PFe (0.16 g, 0.14 mmol) in DMSO (3 mL). The reaction mixture was stirred and irradiated with a blue LED lamp (Penn PhD s/n 00598, photo reactor m2450 nm) forl6 h. The rection mixture was diluted by ice cold water (10 mL) and extracted with ethyl acetate (3 x 15 mL). The combined organic phase was washed with water (10 mL), saturated brine (5 mL) dried over sodium sulphate and evaporated in vacuo to give the crude product of 4 g as a brown oil. The crude mixture was purified via reverse phase chromatography (MeCN / H2O, 0.1% formic acid modifier, 0%- 100% gradient, 60 min run) to afford 2-chloro-6-(l-methylcyclohexyl)nicotinonitrile (380 mg, 1.5 mmol, 20 % yield) as light brown solid. J H NMR (400 MHz, DMSO-de) 5 8.41 (d, J= 8.0 Hz, 1H), 7.70 (d, J= 8.0 Hz, 1H), 2.16 - 2.05 (m, 2H), 1.58 - 1.48 (m, 4H), 1.44 - 1.35 (m, 2H), 1.33 - 1.22 (m, 2H), 1.18 (s, 3H). ES-LCMS m/z 235.0 [M+H] + .

Step 2: 2-chloro-6-(l-methylcyclohexyl)nicotinic acid

To a solution of 2-chloro-6-(l-methylcyclohexyl)nicotinonitrile (360 mg, 1.5 mmol) in ethanol (5 mL) stirred under nitrogen at room temp was added a solution of sodium hydroxide (310 mg, 7.7 mmol) in water (2.5 mL). The reaction mixture was stirred at 80 °C for 3 h. The reaction mixture was concentrated under reduced pressure and then diluted with water (2 mL). The aqueous layer was neutralized with 1.5 M HC1 to a pH = 6. Aqueous layer was injected directly to be purified via reverse phase chromatography (MeCN / H2O, 0.1% formic acid modifier, 0%-100% gradient, 60 min run) to afford 2-chloro-6-(l-methylcyclohexyl)nicotinic acid (200 mg, 0.78 mmol, 51 % yield) as a light brown solid. 'H NMR (400 MHz, DMSO-de) 5 13.58 (br s, 1H), 8.17 (d, J= 8.0 Hz, 1H), 7.55 (d, J= 8.0 Hz, 1H), 2.18 - 2.05 (m, 2H), 1.59 - 1.46 (m, 4H), 1.43 - 1.23 (m, 4H), 1.17 (s, 3H). ES-LCMS m/z 254.0 [M+H] + .

Step 3: 2-methoxy-6-(l-methylcyclohexyl)nicotinic acid

To a solution of 2-chloro-6-(l-methylcyclohexyl)nicotinic acid (200 mg, 0.79 mmol) in methanol (5 mL) stirred under nitrogen at room temp was added sodium methoxide (25% in methanol) (0.89 mL, 3.9 mmol). The reaction mixture was stirred at 80 °C for 16 h. The reaction mixture was concentrated under reduced pressure to give the required crude product as light brown solid. The crude product mixture was dissolved in DCM (2 mL) preabsorbed on silica and purified using normal phase chromatography (0-100% EtOAc / petroleum ether and then 0-20% MeOH/petroleum ether) to afford 2-methoxy-6-(l-methylcyclohexyl)nicotinic acid (80 mg, 0.24 mmol, 30 % yield) as a brown solid. 'H NMR (400 MHz, DMSO-de) 5 13.13 - 12.24 (m, 1H), 8.05 (d, J= 8.0 Hz, 1H), 7.07 (d, J= 7.5 Hz, 1H), 3.91 (s, 3H), 2.22 - 2.07 (m, 2H), 1.57 - 1.26 (m, 8H), 1.17 (s, 3H). ES-LCMS m/z 250.2 [M+H] + .

Step 4: (R)-N-( 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-( 1 - methylcyclohexyl)nicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6- (l- methylcyclohexyl)nicotinamide was obtained from a HATU coupling protocol similar to ones already described herein using 2-methoxy-6-(l-methylcyclohexyl)nicotinic acid. 'H NMR (400 MHz, DMSO-de) 5 8.69 (d, J= 7.5 Hz, 1H), 8.09 (d, J= 7.5 Hz, 1H), 7.23 (dd, J = 6.5, 2.0 Hz, 1H), 7.14 (d, J= 8.0 Hz, 1H), 6.92 (dd, J= 6.8, 2.8 Hz, 1H), 5.36 (tdt, J= 7.7, 5.3, 2.6 Hz, 1H), 3.96 (s, 3H), 3.76 (dd, J = 13.5, 8.0 Hz, 1H), 3.33 - 3.29 (m, 1H), 2.22 - 2.11 (m, 2H), 1.60 - 1.23 (m, 8H), 1.17 (s, 3H). ES-LCMS m/z 365.0 [M+H] + . Intermediate 82: tert-butyl (R)-(l-(5-((l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamoyl)- 6- ethoxypyridin-2-yl)cyclohexyl)carbamate

Step 1: tert-butyl (l-(6-chloro-5-cyanopyridin-2-yl)cyclohexyl)carbamate tert-butyl (l-(6-chloro-5-cyanopyridin-2-yl)cyclohexyl)carbamate was obtained via a photo- Minisci reaction via procedure similar to one already described herein using 2- chloronicotinonitrile and l-((tert-butoxycarbonyl)amino)cyclohexane-l -carboxylic acid. 'H NMR (400 MHz, DMSO-de) 5 8.41 (d, J= 8.0 Hz, 1H), 7.56 (br d, J= 8.0 Hz, 1H), 7.28 (br s, 1H), 2.17 - 2.01 (m, 2H), 1.80 - 1.46 (m, 8H), 1.41 - 1.26 (m, 9H). ES-LCMS m/z 336.0 [M+H] + .

Step 2: 6-(l-((tert-butoxycarbonyl)amino)cyclohexyl)-2-ethoxynicotin ic acid

6-(l-((tert-butoxycarbonyl)amino)cyclohexyl)-2-ethoxynico tinic acid was obtained via a hydrolysis and SNAr using a procedure similar to one already described herein using tert-butyl (l-(6-chloro-5-cyanopyridin-2-yl)cyclohexyl)carbamate and NaOH in EtOH. J H NMR (400 MHz, DMSO-de) 5 12.69 (br s, 1H), 8.01 (br d, J= 7.5 Hz, 1H), 7.03 (br s, 1H), 6.98 (br d, J = 7.5 Hz, 1H), 4.41 (q, J= 7.0 Hz, 2H), 2.19 - 2.03 (m, 2H), 1.84 - 1.72 (m, 2H), 1.67 - 1.46 (m, 6H), 1.35 (br s, 9H), 1.32 (t, J= 7.0 Hz, 3H). ES-LCMS m/z 365.0 [M+H] + .

Step 3: tert-butyl (R)-(l-(5-((l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamoyl)- 6- ethoxypyridin-2-yl)cyclohexyl)carbamate tert-butyl (R) - ( 1 -(5 -(( 1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)carbamoyl)-6-ethoxypyridin-2- yl)cyclohexyl)carbamate was obtained from a HATU coupling protocol similar to ones already described herein using 6-(l-((tert-butoxycarbonyl)amino)cyclohexyl)-2 -ethoxynicotinic acid. 'H NMR (400 MHz, DMSO-de) 5 8.62 (d, J= 8.0 Hz, 1H), 8.07 (br d, J= 7.5 Hz, 1H), 7.25 (dd, J= 6.8, 2.3 Hz, 1H), 7.06 (br d, J= 7.5 Hz, 2H), 6.93 (dd, J= 6.5, 2.5 Hz, 1H), 5.38 (tdt, J= 7.7, 5.1, 2.5 Hz, 1H), 4.46 (q, J= 7.0 Hz, 2H), 3.76 (dd, J= 13.5, 8.0 Hz, 1H), 3.27 (dd, J = 13.5, 5.0 Hz, 1H), 2.20 - 2.03 (m, 2H), 1.87 - 1.72 (m, 2H), 1.68 - 1.46 (m, 6H), 1.38 (t, J = 7.0 Hz, 3H), 1.43 - 1.30 (m, 12H). ES-LCMS m/z 480.0 [M+H] + .

Intermediate 83: N-((R)-l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-((l r,4R)-4-

(trifluoromethyl)cyclohexyl)nicotinamide and Intermediate 84: N-((R)-l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-methoxy-6-((ls,4S)-4-(trifluoromethy l)cyclohexyl)nicotinamide Step 1: l,3-dioxoisoindolin-2-yl (ls,4s)-4-(trifluoromethyl)cyclohexane-l -carboxylate

Stock solution of N-hydroxyphthalimide: In a 40 mL vial, N-hydroxyphthalimide (4.5 g, 28 mmol) was suspended in DCM to produce a total volume of 40 mL (a suspension resulted). Stock solution of DCC: In a 40 mL vial, DCC (5.7 g, 28 mmol) was dissolved in DCM to produce total of 40 mL. Stock solution of DMAP: In a 40 mL vial, DMAP (310 mg, 2.5 mmol) was dissolved in 40 mL of DCM. In a 20 mL vial containing a magnetic stirrer and the desired carboxylic acid (2.50 mmol) were added sequentially suspension of N-hydroxyphthalimide (4 mL of stock solution, slurry dosing), DCC (4 mL of stock solution), and DMAP (4 mL of stock solution). The reactions were stirred at r.t. for 16 h. The resulting mixtures were fdtered through a disposable filter funnel and purified directly via normal phase chromatography. Specific example: (ls,4s)-4-(trifluoromethyl)cyclohexane-l -carboxylic acid (490 mg, 2.5 mmol) was used. The crude reaction mixture was purified via normal phase chromatography (20% EtOAc / heptane) to afford l,3-dioxoisoindolin-2-yl (ls,4s)-4-(trifluoromethyl)cyclohexane-I- carboxylate (790 mg, 2.3 mmol, 92% yield) as a white solid. NMR (400 MHz, DMSO-de) 5 8.03 - 7.92 (m, 4H), 3.32 (s, 1H), 2.48 - 2.31 (m, 1H), 2.25 - 2.10 (m, 2H), 1.90 - 1.70 (m, 4H), 1.57 - 1.41 (m, 2H). ES-LCMS desired m/z not detected.

Step 2: (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-(4-

(trifluoromethyl)cyclohexyl)nicotinamide

To a 20 mL vial were added sodium bicarbonate (220 mg, 2.7 mmol), l,3-dioxoisoindolin-2- yl (Is,4s)-4-(trifluoromethyl)cyclohexane-I -carboxylate (340 mg, 990 pmol), (R)-6-bromo-N- (I,I-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxynicotinamide (0.23 g, 660 pmol), 3,5- pyridinedicarboxylic acid, I,4-dihydro-2,6-dimethyl-, diethyl ester (340 mg, 1.3 mmol), and [Ni(dtbbpy)(H2O)4]C12 (62 mg, 130 pmol). The vial was purged with dry nitrogen gas (3x) and then degassed DMA (6.6 mL) was added. The reaction mixture was irradiated over 16 h using the Penn PHD reactor (395 nm wavelength, 100% intensity, 800 rpm stirring). The mixture was combined with EtOAc and water and transferred to a separatory funnel. The phases were separated, and the aqueous phase was extracted with EtOAc (3x). The combined organic portions were diluted with hexane (30 mL) and the new organic phase was washed with water (3x), once with brine, and then concentrated to afford the crude product mixture. The crude product mixture was purified via normal phase chromatography (35-65% EtOAc / heptane) to afford (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-(4- (trifluoromethyl)cyclohexyl)nicotinamide (94 mg, 0.22 mmol, 33 % yield) as a light yellow solid (~3:2 ratio of isomers by NMR). Complex NMR spectrum. ES-LCMS m/z 419.2 [M+H] + .

Step 3:

N-((R)-l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6- ((lr,4R)-4- (trifluoromethyl)cyclohexyl)nicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6- (4- (trifluoromethyl)cyclohexyl)nicotinamide (94 mg, 0.22 mmol) was purified via chiral chromatography (85: 15% CO2:isopropanol, Chiralpak OJ-H column) to afford N-((R)-1,1- dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-((lr,4R)-4-

(trifluoromethyl)cyclohexyl)nicotinamide (38 mg, 90 pmol, 100% ee). 'H NMR (501 MHz, DMSO-de) 5 8.69 (d, J= 7.6 Hz, 1H), 8.07 (d, J= 7.7 Hz, 1H), 7.23 (dd, J= 6.6, 2.3 Hz, 1H), 7.01 (d, J= 7.6 Hz, 1H), 6.92 (dd, J= 6.6, 2.6 Hz, 1H), 5.35 (tdt, J= 7.7, 5.4, 2.6 Hz, 1H), 3.96 (s, 3H), 3.76 (dd, J= 13.5, 8.0 Hz, 1H), 3.31 (dd, J= 13.5, 5.5 Hz, 1H), 2.72 - 2.65 (m, 1H), 2.36 - 2.28 (m, 1H), 2.05 - 1.94 (m, 4H), 1.60 (qd, J = 12.9, 3.2 Hz, 2H), 1.43 (qd, J= 13.0, 3.4 Hz, 2H). ES-LCMS m/z 419.2 [M+H] + . N-((R)-l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-((l s,4S)-4-

(trifluoromethyl)cyclohexyl)nicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6- (4- (trifluoromethyl)cyclohexyl)nicotinamide (94 mg, 0.22 mmol) was purified via chiral chromatography (85: 15% CChasopropanol. Chiralpak OJ-H column) to afford N-((R)-1,1- dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-6-((ls,4S)-4-

(trifhioromethyl)cyclohexyl)nicotinamide (61 mg, 150 pmol, 100% ee). 1 H NMR (501 MHz, DMSO-de) 5 8.70 (d, J= 8.0 Hz, 1H), 8.08 (d, J= 7.7 Hz, 1H), 7.23 (dd, J= 6.7, 2.1 Hz, 1H), 7.09 (d, J= 8.0 Hz, 1H), 6.92 (dd, J= 6.7, 2.5 Hz, 1H), 5.36 (tdt, J= 7.8, 5.3, 2.6 Hz, 1H), 3.95 (s, 3H), 3.76 (dd,J= 13.3, 7.8 Hz, 1H), 3.31 (dd,J= 13.5, 5.5 Hz, 1H), 3.03 (quin, J = 4.7 Hz, 1H), 2.47 - 2.38 (m, 1H), 2.19 - 2.08 (m, 2H), 1.87 - 1.64 (m, 6H). ES-LCMS m/z 419.2 [M+H] + .

Intermediate 85: N-((R)-l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5-

(methoxymethoxy)-6-((lr,4R)-4-(trifluoromethyl)cyclohexyl )nicotinamide and Intermediate 86: N-((R)-l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5-

(methoxymethoxy)-6-((ls,4S)-4-(trifluoromethyl)cyclohexyl )nicotinamide Step 1: (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5-(me thoxymethoxy)-6-(4- (trifluoromethyl)cyclohexyl)nicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5- (methoxymethoxy)-6-(4- (trifluoromethyl)cyclohexyl)nicotinamide was obtained via photoredox catalysis according to a procedure similar to one already described using (R)-6-bromo-N-(l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-methoxy-5-(methoxymethoxy)nicotinami de and 1,3- dioxoisoindolin-2-yl (ls,4s)-4-(trifluoromethyl)cyclohexane-l-carboxylate. ~1:1 Ratio of cis and trans isomers by NMR. Complex NMR spectrum. ES-LCMS m/z 479.1 [M+H] + .

Step 2:

N-((R)-l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5- (methoxymethoxy)-6-((lr,4R)-4-

(trifluoromethyl)cyclohexyl)nicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5- (methoxymethoxy)-6-(4- (trifluoromethyl)cyclohexyl)nicotinamide (220 mg, 0.37 mmol) was purified via chiral chromatography (85: 15% CChusopropanol, Chiralpak OJ-H column) to afford N-((R)-1,1- dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5-(methoxymethox y)-6-((lr,4R)-4-

(trifluoromethyl)cyclohexyl)nicotinamide (80 mg, 170 pmol, 100% ee) as a white solid. J H NMR (501 MHz, DMSO-de) 5 8.70 (d, J= 7.6 Hz, 1H), 7.89 (s, 1H), 7.23 (dd, J= 6.4, 2.1 Hz, 1H), 6.92 (dd, J= 6.7, 2.4 Hz, 1H), 5.34 (tdt, J= 7.8, 5.3, 2.4 Hz, 1H), 5.21 (s, 2H), 3.94 (s, 3H), 3.75 (dd, J= 13.5, 7.6 Hz, 1H), 3.40 (s, 3H), 3.33 (dd, J= 13.2, 5.5 Hz, 1H), 3.07 (tt, J= 11.7, 3.3 Hz, 1H), 2.37 - 2.30 (m, 1H), 1.99 (br d, J= 10.4 Hz, 2H), 1.86 (br d, J= 11.3 Hz, 2H), 1.68 (qd, J = 12.8, 2.9 Hz, 2H), 1.43 (qd, J = 12.8, 3.2 Hz, 2H). ES-LCMS m/z 479.2 [M+H] + .

N-((R)-l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5- (methoxymethoxy)-6-((ls,4S)-4- (trifluoromethyl)cyclohexyl)nicotinamide

(R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5- (methoxymethoxy)-6-(4- (trifluoromethyl)cyclohexyl)nicotinamide (220 mg, 0.37 mmol) was purified via chiral chromatography (85: 15% CChasopropanol. Chiralpak OJ-H column) to afford N-((R)-1,1- dioxido-2,3-dihydrothiophen-3-yl)-2-methoxy-5-(methoxymethox y)-6-((ls,4S)-4-

(trifluoromethyl)cyclohexyl)nicotinamide (55 mg, 120 pmol, 100% ee) as a white solid. 'H NMR (501 MHz, DMSO-de) 5 8.71 (d, J= 8.0 Hz, 1H), 7.90 (s, 1H), 7.23 (dd, J= 6.7, 2.4 Hz, 1H), 6.92 (dd, J= 6.7, 2.8 Hz, 1H), 5.35 (tdt, J= 7.7, 5.3, 2.6 Hz, 1H), 5.22 (s, 2H), 3.92 (s, 3H), 3.75 (dd, J= 13.5, 8.0 Hz, 1H), 3.39 (s, 3H), 3.38 - 3.37 (m, 1H), 3.33 (dd, J= 13.5, 5.5 Hz, 1H), 2.45 - 2.38 (m, 1H), 2.01 - 1.89 (m, 4H), 1.82 - 1.66 (m, 4H). ES-LCMS m/z 479.2 [M+H] + .

Intermediate 87: (R)-6-(3,3-dimethylcyclobutyl)-N-(l,l-dioxido-2,3-dihydrothi ophen-3-yl)-2- methoxynicotinamide

(R)-6-(3,3-dimethylcyclobutyl)-N-(l,l-dioxido-2,3-dihydro thiophen-3-yl)-2- methoxynicotinamide was obtained via photoredox catalysis according to a procedure similar to one already described using l,3-dioxoisoindolin-2-yl 3,3-dimethylcyclobutane-l- carboxylate and (R)-6-bromo-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2- methoxynicotinamide. 'H NMR (400 MHz, DMSO-de) 5 8.76 - 8.59 (m, 1H), 8.04 (d, J= 7.8 Hz, 1H), 7.23 (dd, J= 6.6, 2.2 Hz, 1H), 7.04 - 6.95 (m, 1H), 6.95 - 6.87 (m, 1H), 5.48 - 5.29 (m, 1H), 4.01 (s, 3H), 3.76 (dd, J= 13.2, 7.8 Hz, 1H), 3.58 (t, J= 8.8 Hz, 1H), 3.30 (d, J= 5.4 Hz, 1H), 2.19 - 2.01 (m, 4H), 1.25 (s, 3H), 1.13 (s, 3H). ES-LC-MS m/z 351.0 [M + H] + .

Intermediate 88: 6-(2,2-difluorocyclohexyl)-N-((R)-l, l-dioxido-2,3-dihydrothiophen-3-yl)-2- methoxynicotinamide

6-(2,2-difluorocyclohexyl)-N-((R)-l,l-dioxido-2,3-dihydro thiophen-3-yl)-2- methoxynicotinamide was obtained via photoredox catalysis according to a procedure similar to one already described using l,3-dioxoisoindolin-2-yl 2,2-difluorocyclohexane-l- carboxylate and (R)-6-bromo-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2- methoxynicotinamide. 'HNMR (400 MHz, DMSO-de) 5 8.77 - 8.71 (m, 1H), 8.07 (d, J= 7.8 Hz, 1H), 7.36 - 7.18 (m, 1H), 7.11 (d, J= 7.8 Hz, 1H), 6.92 (dd, J = 6.6, 2.7 Hz, 1H), 5.47 - 5.28 (m, 1H), 3.98 - 3.92 (m, 4H), 3.85 - 3.65 (m, 1H), 3.30 - 3.25 (m, 1H), 2.23 - 2.06 (m, 1H), 2.07 - 1.73 (m, 5H), 1.69 - 1.29 (m, 2H). ES-LC-MS m/z 387.2 [M + H] + .

Intermediate 89: 6-((3R,5S)-3,5-dimethylcyclohexyl)-N-((R)-l, l-dioxido-2,3- dihydrothiophen-3-yl)-2 -methoxynicotinamide

6-(2,2-difluorocyclohexyl)-N-((R)-l,l-dioxido-2,3-dihydro thiophen-3-yl)-2- methoxynicotinamide was obtained via photoredox catalysis according to a procedure similar to one already described using l,3-dioxoisoindolin-2-yl (ls,3R,5S)-3,5-dimethylcyclohexane- 1 -carboxylate and (R)-6-bromo-N-( 1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)-2- methoxynicotinamide. 'H NMR (400 MHz, DMSO-de) 5 8.67 (d, J= 7.8 Hz, 1H), 8.17 - 8.00 (m, 1H), 7.31 - 7.18 (m, 1H), 7.16 - 6.95 (m, 1H), 6.95 - 6.84 (m, 1H), 5.36 (ddt, J= 7.8, 5.3, 2.5 Hz, 1H), 4.04 - 3.90 (m, 3H), 3.82 - 3.60 (m, 1H), 3.30 (br dd, J= 5.6, 1.7 Hz, 1H), 2.88 - 2.69 (m, 1H), 2.25 (br d, J= 11.7 Hz, 1H), 1.91 - 1.65 (m, 2H), 1.65 - 1.48 (m, 2H), 1.32 - 1.18 (m, 1H), 1.17 - 1.01 (m, 1H), 0.98 - 0.77 (m, 6H), 0.72 - 0.49 (m, 1H). ES-LC-MS m/z 3192 [M + H] + .

Intermediate 90: methyl 2-methoxy-6-(2-phenethylphenyl)nicotinate

Step 1 : (2-bromobenzyl)triphenylphosphonium bromide

A stirring solution of l-bromo-2-(bromomethyl)benzene (5.0 g, 20 mmol) and triphenylphosphine (5.6 g, 21 mmol) in anhydrous toluene (100 mL) was heated at 115 °C for 18 h. The resulting suspension was cooled to room temperature then the solids were collected by filtration, rinsed well with toluene (100 mL) and diethyl ether (200 mL), and dried under suction to afford (2- bromobenzyl)triphenylphosphonium bromide (10 g, 18 mmol, 91 % yield) as a white solid. 'H NMR (400 MHz, DMSO-d6) 7.96 - 7.89 (m, 3H), 7.79 - 7.69 (m, 6H), 7.67 - 7.54 (m, 7H), 7.33 - 7.23 (m, 2H), 7.22 - 7.11 (m, 1H), 5.17 (d, J= 14.7 Hz, 2H). ES- LCMS m/z 431.1, 433.1 [M+H] + , bromine pattern.

Step 2: (E)-l-bromo-2-styrylbenzene and (Z)-l-bromo-2-styrylbenzene

A stirring solution of (2-bromobenzyl)triphenylphosphonium bromide (10 g, 20 mmol) in anhydrous DCM (100 mb) was cooled to 0 °C then slowly treated with portions of 60% sodium hydride in mineral oil (2.9 g, 72 mmol). The resulting suspension stirred at 0 °C for 1 h then benzaldehyde (1.8 mb, 18 mmol) was added. Stirring continued at r.t. for 18 h, then the reaction was slowly quenched with saturated aqueous solution of ammonium chloride (100 mb). The phases were separated and the aqueous was extracted with DCM (50 mb). The combined extracts were washed with saturated aqueous ammonium chloride (50 mL) and brine (50 mb), dried (MgSO4), and evaporated to a crude oil. Purification by normal phased chromatography (0 - 40 % ethyl acetate in heptane) afforded (E)-l-bromo-2-styrylbenzene and (Z)-l-bromo-2- styrylbenzene as amixture (4.6 g, 16 mmol, 88 % yield) as a colorless oil. ES-LCMS m/z 258.8, 261.0 [M+H] + , bromine pattern.

Step 3 : methyl 2-methoxy-6-(2-phenethylphenyl)nicotinate

A degassed suspension of E/Z l-bromo-2-styrylbenzene (4.5 g, 18 mmol) and 10% palladium on carbon and 50% water (0.19 g, 0.88 mmol) in absolute ethanol (35 mL) was hydrogenated (balloon) at r.t for 18 h. The reaction was filtered then concentrated under reduced pressure to a crude oil. Purification by normal phase chromatography (EtOAc / heptane) afforded crude 1- bromo-2-phenethylbenzene as a mixture with some de-brominated side-product.

A degassed suspension of methyl 2-methoxy-6-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)nicotinate (1.0 g, 3.4 mmol), crude l-bromo-2 -phenethylbenzene (1.4 g, 4.1 mmol), APhos Pd G3 (0.11 g, 0.17 mmol), and potassium carbonate (1.1 g, 8.2 mmol) in a mixture of THF (14 mL) and water (3.4 mL) was heated at 80 °C for 18 h. The reaction was diluted with saturated aqueous ammonium chloride and extracted with EtOAc. The extracts were washed with saturated aqueous ammonium chloride and brine, dried over MgSC>4, then evaporated to a crude residue. Purification by normal phase chromatography (0-40% ethyl acetate in heptane) afforded methyl 2-methoxy-6-(2-phenethylphenyl)nicotinate (930 mg, 2.6 mmol, 75 % yield) as a colorless oil. 'H NMR (400 MHz, DMSO-de) 5 8.22 (d, J= 7.8 Hz, 1H), 7.42 - 7.30 (m, 4H), 7.26 - 7.10 (m, 4H), 7.06 - 6.95 (m, 2H), 3.89 (s, 3H), 3.87 - 3.80 (m, 3H), 3.12 - 3.03 (m, 2H), 2.77 - 2.68 (m, 2H). ES-LCMS m/z 348.1 [M+H] + .

Intermediate 91: (R)-3-amino-5-fhroro-2,3-dihydrothiophene 1,1-dioxide hydrobromide

Step 1: tert-butyl (R)-4-(2,2-difluorovinyl)-2,2-dimethyloxazolidine-3-carboxyl ate

A flask charged with THF (100 mL) was placed into a -78 °C dry ice bath, then dibromodifluoromethane (4.0 mL, 44 mmol) was added followed by dropwise addition of N,N,N',N',N",N"-hexamethylphosphanetriamine (16 mL, 87 mmol) and the resulting mixture was stirred at this temperature for 15 min, then taken out of the cooling bath and allowed to warm to rt over the course of 15 min. After this period, a solution of tert-butyl (S)-4-formyl- 2,2-dimethyloxazolidine-3-carboxylate (5.0 g, 22 mmol) in THF (25 mL)was added dropwise and the mixture was stirred at rt for 2 h. The reaction was quenched by ice-cold water (200 mL) and extracted with ether (2 x 250 mL). The combined organic extracts were dried over MgSO4, fdtered and concentrated in vacuo. The evaporation residue was dissolved in hexanes and filtered through a plug of silica eluting with 5% Et2O/hexanes (150 mL) to afford tert-butyl (R)-4-(2,2-difluorovinyl)-2,2-dimethyloxazolidine-3-carboxyl ate (4.0 g, 15 mmol, 70 % yield) as a colorless oil. Due to the relative volatility and the absence of UV absorbance, only an NMR was used to characterize the product. 1 H NMR (400 MHz, Chloroform-d) 5 4.70 - 4.47 (m, 1H), 4.44 - 4.30 (m, 1H), 4.12 - 4.01 (m, 1H), 3.75 (dd, J= 9.0, 1.7 Hz, 1H), 1.61 (br s, 3H), 1.54 - 1.39 (m, 12H).

Step 2: (R)-2-amino-4,4-difluorobut-3-en-l-ol, hydrochloride

To a flask charged with tert-butyl (R)-4-(2,2-difluorovinyl)-2,2-dimethyloxazolidine-3- carboxylate (820 mg, 3.1 mmol) was added 3N HC1 in MeOH (5200 pL, 16 mmol) and the mixture was stirred for 18 h before it was concentrated in vacuo to afford crude (R)-2-amino- 4,4-difluorobut-3-en-l-ol, hydrochloride (550 mg, 3.5 mmol, 110 % yield) (contained residual methanol) as an opaque oily solid. Due to the low molecular weight and the absence of UV absorbance, the final product was only characterized by NMR. J H NMR (501 MHz, DMSO- d 6 ) 5 8.43 (br s, 2H), 5.14 (br s, 1H), 4.75 - 4.68 (m, 1H), 3.85 - 3.81 (m, J= 9.9, 5.0 Hz, 1H), 3.60 - 3.55 (m, 2H).

Step 3: benzyl (R)-(4,4-difluoro-l-hydroxybut-3-en-2-yl)carbamate

To a flask charged with (R)-2-amino-4,4-difluorobut-3-en-l-ol, hydrochloride (2.0 g, 13 mmol) was added potassium carbonate (3.5 g, 25 mmol), water (30 mL), and THF (30 mL). The mixture was stirred at rt for 1 min, then placed into a 0 °C cooling bath, stirred for 5 min, then Cbz-Cl (2.0 mL, 14 mmol) was added dropwise and the mixture was vigorously stirred at 0 °C for 1 h, then gradually allowed to warm to rt in a thawing ice bath over the course of 5 h. The mixture was diluted with DCM (200 mL), the organic layer was separated, dried over MgSO4, filtered, concentrated in vacuo, and subjected to normal phase chromatography (0- 100 % EtOAc / heptane) to afford benzyl (R)-(4,4-difluoro-l-hydroxybut-3-en-2-yl)carbamate (2.6 g, 10 mmol, 81 % yield) as a white solid. 'H NMR (400 MHz, Chloroform-d) 5 7.46 - 7.31 (m, 5H), 5.14 (s, 3H), 4.54 - 4.36 (m, 2H), 3.84 - 3.65 (m, 2H). ES-LCMS m/z 258.1 [M+H] + .

Step 4: (R)-S-(2-(((benzyloxy)carbonyl)amino)-4,4-difluorobut-3-en-l -yl) benzothioate

To a vial charged with triphenylphosphine (3.3 g, 13 mmol) was added THF (12 mL) under nitrogen, the vial was placed into a 0 °C cooling bath, and 40 % DEAD in PhMe (5.8 mL, 13 mmol) was added dropwise at this temperature. After 10 min, a solution of benzyl (R)-(4,4- difluoro-l-hydroxybut-3-en-2-yl)carbamate (2.6 g, 10 mmol) in THF (24 mL)was added dropwise at 0 °C slowly over the course of 4 min, the mixture stirred for 10 min, then thiobenzoic acid (1.7 mL, 13 mmol) was added and the stirring continued for 2.5 h as the cooling bath was gradually warming to rt. After this period, the reaction was quenched with sat. aq. NaHCOs (50 mL) and partitioned between DCM (150 mL) and sat. aq. NaHCOs (100 mL). The aqueous layer was re-extracted with DCM (150mL), the combined organic extracts were dried over MgSCL, filtered, concentrated in vacuo, and subjected to normal phase chromatography (0-100% EtOAc/heptane) to afford (R)-S-(2-(((benzyloxy)carbonyl)amino)- 4,4-difluorobut-3-en-l-yl) benzothioate (3.0 g, 8.0 mmol, 79 % yield) as awhite solid. 'HNMR (400 MHz, Chloroform-d) 5 8.02 - 7.95 (m, J= 1.5 Hz, 2H), 7.66 - 7.60 (m, 1H), 7.49 (s, 2H), 7.34 (s, 5H), 5.21 (br s, 1H), 5.12 (s, 2H), 4.71 - 4.53 (m, J= 6.8 Hz, 1H), 4.51 - 4.35 (m, 1H), 3.50 - 3.26 (m, 2H). ES-LCMS m/z 378.1 [M+H] + .

Step 5: benzyl (R)-(5-fluoro-2,3-dihydrothiophen-3-yl)carbamate

To a 500 mL flask charged with (R)-S-(2-(((benzyloxy)carbonyl)amino)-4,4-difluorobut-3-en- 1-yl) benzothioate (3.0 g, 8.0 mmol) was added THF (220 mL), the flask was sealed and degassed by vigorous bubbling of N2 through it while the solution was stirring (1700 rpm) for 10 min. After this period, the flask was opened, potassium methanolate (0.61 g, 8.7 mmol) was added, the flask was immediately sealed, the degassing needle reinserted, and the mixture was stirred vigorously at rt while being degassed for another 5 min. The flask was positioned into a heating block set to 65 °C (internal temperature of the reaction mixture likely lower), the heating was turned on, and the reaction mixture was stirred at 1700 rpm for 3 h. After this period, the mixture was allowed to cool down to rt, THF was removed in vacuo, the evaporation residue suspended in 50 mL DCM, filtered, the filtrate concentrated in vacuo, and subjected to normal phase chromatography (0-100 % EtOAc / heptane) to yield benzyl (R)-(5-fluoro-2,3- dihydrothiophen-3-yl)carbamate (1.3 g, 5.1 mmol, 65 % yield) as a white solid. 1 H NMR (400 MHz, Chloroform-d) 5 7.46 - 7.30 (m, 5H), 5.21 - 4.98 (m, 5H), 3.82 (br dd, J= 12.2, 7.3 Hz, 1H), 3.24 (br d, J= 13.2 Hz, 1H). ES-LCMS m/z 254.1 [M+H] + .

Step 6: benzyl (R)-(5-fluoro-l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamate

To a solution of benzyl (R)-(5-fluoro-2,3-dihydrothiophen-3-yl)carbamate (1.3 g, 5.1 mmol) in DCM (60 mb) at rt was added 77 % mCPBA (2.9 g, 13 mmol) and the resulting mixture was stirred at rt for 18 h. After this period, the mixture was diluted with DCM (200 mL), transferred into 1 L Erlenmeyer flask, then sat. aq. NaHCOs (100 mL) was added, the mixture stirred for 1 min. Sat. aq. Na2S20s (100 mL) was added, the mixture stirred for 1 min, then brine (200 mL) was added, and the stirring continued for 5 min. The organic layer was separated, the aqueous layer re-extracted with DCM (2 x 200 mL), the combined organic extracts dried over Na2SC>4, filtered, concentrated in vacuo, and subjected to normal phase chromatography (ISCO, 0-100% EtOAc / heptane) to afford benzyl (R)-(5 -fluoro- 1,1 -dioxido-2, 3 -dihydrothiophen-3- yl)carbamate (1.1 g, 3.8 mmol, 75 % yield) as a white solid. 1 H NMR (400 MHz, Chloroform- d) 5 7.44 - 7.29 (m, 5H), 6.06 (t, J= 3.2 Hz, 1H), 5.22 - 5.04 (m, 4H), 3.74 (dd, J= 13.9, 7.6 Hz, 1H), 3.25 (br d, J= 14.2 Hz, 1H). ES-LCMS m/z 284.1 [M-H]’.

Step 7: (R)-3-amino-5-fhioro-2,3-dihydrothiophene 1,1 -dioxide hydrobromide

To a flask charged with benzyl (R)-(5-fluoro-l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamate (1.1 g, 3.8 mmol) was added 33% hydrogen bromide in acetic acid (17 mL, 97 mmol) and the resulting solution was stirred at rt for 25 min. After this period, diethyl ether (200 mL) was added slowly with stirring and the resulting suspension was stirred vigorously for 20 min. The suspension was fdtered, the precipitate washed with additional diethyl ether (100 mL), and airdried (suction fdtration) to afford (R)-3-amino-5-fluoro-2, 3 -dihydrothiophene 1,1-dioxide, hydrobromide (850 mg, 3.6 mmol, 97 % yield) as a free-flowing white non-hygroscopic solid. 'H NMR (501 MHz, DMSO-de) 5 8.52 (s, 2H), 6.68 (t, J= 3.5 Hz, 1H), 4.76 - 4.72 (m, 1H), 4.07 (dd, J= 14.5, 8.4 Hz, 1H), 3.55 (dd, J= 14.7, 4.0 Hz, 1H), 3.36 (br s, 1H). ES-LCMS m/z 152.0 [M+H] + .

Intermediate 92: (R)-3-amino-5-methyl-2,3-dihydrothiophene 1,1-dioxide, hydrobromide

Step 1: tert-butyl (R)-4-ethynyl -2, 2-dimethyloxazolidine-3 -carboxylate

To a solution of tert-butyl (S)-4-formyl-2,2-dimethyloxazolidine-3-carboxylate (20 g, 87 mmol) in methanol (500 mL) was added dimethyl (l-diazo-2-oxopropyl)phosphonate (25 g, 130 mmol) and the resulting mixture was placed into a 0 °C ice bath. After 10 min of stirring (1000 rpm), potassium carbonate (24 g, 170 mmol) was added slowly, and the stirring continued while the ice bath was gradually warming to rt. After 5.5 h, a nitrogen line was attached, and the septum was vented to gradually strip away a portion of the methanol over the course of 24 h. When only about a third of the original volume remained, the mixture was diluted with water (200 mL) and extracted with ether (2 x 350 mL). The combined organic extracts were dried over MgSO-i. filtered, and concentrated in vacuo. The evaporation residue was suspended in 50 mL of 5% ether/hexane, loaded onto a silica gel plug, and washed with additional 350 mL of 5% ether/hexane. The filtrate was concentrated in vacuo to afford tertbutyl (R)-4-ethynyl-2,2-dimethyloxazolidine-3-carboxylate (18 g, 80 mmol, 92 % yield) as a colorless oil. NMR (400 MHz, Chloroform-d) 5 4.09 - 3.97 (m, 2H), 2.27 (br s, 1H), 1.63 (br s, 3H), 1.49 (s, 12H).

Step 2: tert-butyl (R)-2,2-dimethyl-4-(prop-l-yn-l-yl)oxazolidine-3-carboxylate

To a solution of tert-butyl (R)-4-ethynyl-2,2-dimethyloxazolidine-3-carboxylate (3.0 g, 13 mmol) in THF (100 mL) was added 1.35 M nBuLi in hexanes (11 mL, 15 mmol) dropwise at -78 °C and the resulting mixture was stirred for 10 min, then iodomethane (2.5 mL, 40 mmol) was added dropwise and the mixture was stirred for 30 min. Then the cooling bath was removed and the mixture was allowed to reach rt and stirred for 18 h. After this period, the mixture was diluted with water (50 mL), THF removed in vacuo, the evaporation residue (aqueous emulsion) extracted with DCM (150 mL), the organic layer was separated, dried over MgSCL. filtered, and concentrated in vacuo to afford crude tert-butyl (R)-2,2-dimethyl-4-(prop-l-yn-l- yl)oxazolidine-3-carboxylate (3.1 g, 13 mmol, 97 % yield) as a pale yellow oil. 'H NMR (400 MHz, Chloroform-d) 5 4.05 - 3.89 (m, 2H), 1.83 - 1.78 (m, 3H), 1.62 (br s, 3H), 1.52 - 1.46 (m, 12H).

Step 3: benzyl (R)-(l-hydroxypent-3-yn-2-yl)carbamate, hydrochloride

To a flask charged with tert-butyl (R)-2,2-dimethyl-4-(prop-l-yn-l-yl)oxazolidine-3- carboxylate (3.1 g, 13 mmol) was added 3N HC1 in methanol (19 mb, 58 mmol), and stirred at rt for 24 h. After this period, the mixture was concentrated in vacuo. To the evaporation residue was added THF (30 mL), water (30 mb), and potassium carbonate (3.6 g, 26 mmol) at rt and the mixture was stirred at rt for 1 min before it was placed into an ice bath (0 °C), stirred for 5 min, and then Cbz-Cl (2.0 mL, 14 mmol) was added dropwise, and the resulting mixture was stirred vigorously over the course of 5 h while the cooling bath was allowed to gradually reach rt. The mixture was the diluted with DCM (250 mL), the organic layer was separated, dried over MgSC>4, filtered, and concentrated in vacuo. The evaporation residue was subjected to normal phase chromatography (0-100 % EtOAc / heptane) to afford benzyl (R)-(l- hydroxypent-3-yn-2-yl)carbamate, hydrochloride (2.0 g, 7.5 mmol, 58 % yield) as a white solid. 'H NMR (400 MHz, DMSO-de) 5 7.50 (br d, J= 8.3 Hz, 1H), 7.40 - 7.29 (m, 5H), 5.02 (s, 2H), 4.93 (t, J= 6.1 Hz, 1H), 4.28 - 4.18 (m, 1H), 3.49 - 3.32 (m, 2H), 1.76 (d, J= 2.4 Hz, 3H). ES-LCMS m/z 234.2 [M+H] + .

Step 4: (R)-S-(2-(((benzyloxy)carbonyl)amino)pent-3-yn-l-yl) benzothioate

To a vial charged with triphenylphosphine (5.0 g, 19 mmol) was added THF (20 mL) under nitrogen, the vial was placed into a 0 °C cooling bath, and 40 % DEAD in PhMe (8.6 mL, 19 mmol) was added ropwise at this temperature. After 10 min, the intermediate betaine crashed out and (solidified). Then a solution of benzyl (R)-(l-hydroxypent-3-yn-2-yl)carbamate (2.0 g, 8.6 mmol) in THF (20 mL) was added dropwise at 0 °C, the mixture vigorously stirred for 10 min, then thiobenzoic acid (2.5 mL, 19 mmol) was added and the stirring continued for 1 h. After this period, the reaction was quenched with sat. aq. NaHCOs (20 mL) and partitioned between DCM 100 mL and sat. aq. NaHCOs (50 mL). The aqueous layer was re-extracted with DCM (100 mL), the combined organic extracts were dried over MgSO-i. filtered, concentrated in vacuo, and subjected to normal phase chromatography (0-100% EtOAc / heptane) to afford (R)-S-(2-(((benzyloxy)carbonyl)amino)pent-3-yn-l-yl) benzothioate (2.6 g, 7.4 mmol, 87 % yield) as a white solid. 'H NMR (400 MHz, Chloroform-d) 5 8.00 - 7.94 (m, 2H), 7.62 - 7.56 (m, 1H), 7.46 (t, J= 7.5 Hz, 2H), 7.33 - 7.28 (m, 5H), 5.10 (s, 3H), 4.73 (br s, 1H), 3.43 (br d, J= 5.9 Hz, 2H), 1.81 (d, J= 2.4 Hz, 3H). ES-LCMS m/z 354.06 [M+H] + .

Step 5: benzyl (R)-(5-methyl-2,3-dihydrothiophen-3-yl)carbamate

To a solution of (R)-S-(2-(((benzyloxy)carbonyl)amino)pent-3-yn-l-yl) benzothioate (2.6 g, 7.4 mmol) in methanol (250 mL) was added potassium methanolate (0.52 g, 7.4 mmol) and the resulting mixture was stirred vigorously at rt (open to air) for 1 h. After this period, water (50 mL) was added and the resulting mixture was stirred for 15 min, then 1.0 M NaOH (20 mL) was added. The mixture was stirred for 2.5 h before methanol was removed in vacuo. The resulting aqueous suspension was filtered, the precipitate washed with water (2 x 50 mL), and air-dried to afford benzyl (R)-(5-methyl-2,3-dihydrothiophen-3-yl)carbamate (1.8 g, 7.1 mmol, 95 % yield) as an off-white crystalline solid. J H NMR (400 MHz, DMSO-de) 5 7.62 (br d, J = 7.8 Hz, 1H), 7.40 - 7.28 (m, 5H), 5.38 - 5.19 (m, 1H), 5.01 (s, 2H), 4.93 - 4.83 (m, 1H), 3.50 (dd, J= 11.7, 8.3 Hz, 1H), 3.00 (dd, J= 11.7, 5.4 Hz, 1H), 1.90 (t, J= 1.5 Hz, 3H). ES-LCMS z 250.12 [M+H] + .

Step 6: benzyl (R)-(5-methyl-l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamate

To a solution of benzyl (R)-(5-methyl-2,3-dihydrothiophen-3-yl)carbamate (1.8 g, 7.1 mmol) in DCM (100 mL) at rt was added 77 % mCPBA (4.0 g, 18 mmol) and the resulting mixture was stirred at rt for 1 h. After this period, sat. aq. NaHCOs (50 mL) was added, the mixture stirred for 1 min, then 10 % aq. Na2S20s (50 mL) was added, the mixture stirred for 5 min, then brine (100 mL) was added, the mixture stirred for 5 min, the organic layer was separated, the aq. layer re-extracted with DCM (100 mL), the combined organic layers were dried over Na2SC>4, fdtered, and concentrated in vacuo. The evaporation residue was subjected to normal phase chromatography (0- 100 % EtOAc / heptane) to afford benzyl (R)-(5 -methyl- 1 , 1 -dioxido- 2,3-dihydrothiophen-3-yl)carbamate (1.9 g, 6.8 mmol, 97 % yield) as a white solid. 'H NMR (400 MHz, Chloroform-d) 5 7.40 - 7.31 (m, 5H), 6.20 (br s, 1H), 5.17 - 5.07 (m, 3H), 5.03 (br s, 1H), 3.61 (dd,J= 13.9, 7.6 Hz, 1H), 3.12 (dd, J= 14.2, 2.9 Hz, 1H), 2.07 (t, J= 1.7 Hz, 3H). ES-LCMS m/z 282.08 [M+H] + .

Step 7: (R)-3-amino-5-methyl-2,3-dihydrothiophene 1,1-dioxide, hydrobromide

To a flask charged with benzyl (R)-(5 -methyl- 1,1 -dioxido-2, 3 -dihydrothiophen-3- yl)carbamate (1.9 g, 6.8 mmol) was added 33% hydrogen bromide in acetic acid (30 mL, 170 mmol) and the resulting solution was stirred at rt for 25 min. After this period, diethyl ether (400 mL) was added slowly with stirring and the resulting suspension was stirred vigorously for 20 min. The suspension was fdtered, the precipitate washed with additional diethyl ether (100 mb), and air-dried (suction filtration) to afford (R)-3-amino-5-methyl-2,3- dihydrothiophene 1,1-dioxide, hydrobromide (1.5 g, 6.7 mmol, 98% yield) as a pale pink non- hygroscopic microcrystalline solid. 'H NMR (501 MHz, DMSO-de) 5 8.44 (br s, 3H), 6.53 - 6.51 (m, 1H), 4.67 - 4.63 (m, 1H), 3.83 (dd, J= 14.1, 8.3 Hz, 1H), 3.30 (dd, J= 14.4, 4.9 Hz, 1H), 2.02 (t, J= 1.8 Hz, 3H). ES-LCMS m/z 148.06 [M+H] + .

Intermediate 93: l,l-dioxido-2,3-dihydrothiophen-3-yl)carbamoyl)-6-methoxypyr idin-2- yl)benzoate

1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)carbamoyl)-6-methoxypyridin-2-yl)benzoate was obtained from a Suzuki coupling protocol similar to ones already described herein using (/?)- 6-bromo-/V-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-2 -methoxynicotinamide and (2- (methoxycarbonyl)phenyl)boronic acid. 'H NMR (400 MHz, Chloroform-d) 5 ppm 8.57 (d, J = 7.8 Hz, 1H), 8.32 (br d, J= 7.8 Hz, 1H), 7.78 (dd, J= 7.1, 1.2 Hz, 1H), 7.64 - 7.49 (m, 3H), 7.32 (d, J= 7.8 Hz, 1H), 6.88 - 6.77 (m, 2H), 5.65 (ddtd, J= 9.6, 6.3, 3.3, 1.5 Hz, 1H), 4.09 (s, 3H), 3.75 (dd, J= 13.9, 8.1 Hz, 1H), 3.71 (s, 3H), 3.23 (dd, J= 13.7, 3.4 Hz, 1H). ES-LCMS m/z 403.1 [M+H] + .

Intermediate 94: N-((R)-1, l-dioxido-2,3-dihydrothiophen-3-yl)-6-(2-hydroxycyclohexyl)- 2- methoxynicotinamide

Step 1: methyl 6-(2-hydroxycyclohexyl)-2 -methoxynicotinate

To a solution of methyl 6-(cyclohex-l-en-l-yl)-2 -methoxynicotinate (1.5 g, 6.1 mmol) in THF (20 mL) at 0 °C was added borane tetrahydrofuran complex (12 mb, 12 mmol) under nitrogen gas and stirred for 90 min. After 90 min 3 M NaOH solution (2.0 mL, 6.1 mmol) and 30% H2O2 solution (0.56 mL, 9.1 mmol) were added to reaction mixture at 0 °C. After addition completed, reaction mixture was allowed warm to r.t. and stirred for 16 h. The reaction mixture was quenched with water (20 mL) and EtOAc (30 mL) was added. The mixture was then separated two and the organic layer was washed with sat. sodium thiosulfate (2 x 30 mL) and sat. NaCl (8 mL), dried over with Na2SC>4, and concentrated under vacuo to afford the crude product. The crude product was purified via normal phase chromatography (0-100% EtOAc / petroleum ether over 30 min) to afford methyl 6-(2 -hydroxy cyclohexyl)-2 -methoxynicotinate (140 mg, 0.50 mmol, 8.2 % yield) as a yellow gum. 'H NMR (400 MHz, DMSO-de) 5 8.02 (d, J= 7.5 Hz, 1H), 6.94 (d, J= 8.0 Hz, 1H), 4.44 (d, J= 5.5 Hz, 1H), 3.92 (s, 3H), 3.79 (s, 3H), 3.77 - 3.67 (m, 1H), 2.56 - 2.52 (m, 1H), 1.97 - 1.89 (m, 1H), 1.80 - 1.71 (m, 2H), 1.67 (br d, J= 12.5 Hz, 1H), 1.60 - 1.50 (m, 1H), 1.36 - 1.21 (m, 3H). ES-LCMS m/z 266.2 [M+H] + .

Step 2: 6-(2-hydroxycyclohexyl)-2-methoxynicotinic acid

6-(2-hydroxycyclohexyl)-2-methoxynicotinic acid was obtained via a LiOH hydrolysis procedure similar to ones described previously using methyl 6-(2-hydroxycyclohexyl)-2- methoxynicotinate. NMR (400 MHz, DMSO-de) 5 12.66 (br s, 1H), 7.99 (d, J = 7.5 Hz, 1H), 6.91 (d, J= 8.0 Hz, 1H), 4.58 - 4.24 (m, 1H), 3.91 (s, 3H), 3.72 (td, J= 9.9, 3.8 Hz, 1H), 1.99 - 1.89 (m, 1H), 1.82 - 1.15 (m, 8H). ES-LCMS m/z 252.0 [M+H] + . Step 3: N-((R)-1, l-dioxido-2,3-dihydrothiophen-3-yl)-6-(2-hydroxycyclohexyl)- 2- methoxynicotinamide

N-((R)-l,l-dioxido-2,3-dihydrothiophen-3-yl)-6-(2-hydroxy cyclohexyl)-2- methoxynicotinamide was obtained from a HATU coupling protocol similar to ones already described herein using 6-(2-hydroxycyclohexyl)-2 -methoxynicotinic acid. 'H NMR (400 MHz, DMSO-de) 5 8.67 (d, J= 8.0 Hz, 1H), 8.03 (d, J= 7.5 Hz, 1H), 7.22 (dd, J= 6.8, 2.3 Hz, 1H), 6.97 (d, J= 8.0 Hz, 1H), 6.92 (dd, J= 6.5, 2.5 Hz, 1H), 5.36 (tdt, J= 7.7, 5.3, 2.6 Hz, 1H), 4.42 (d, J = 6.5 Hz, 1H), 3.97 (s, 3H), 3.81 - 3.66 (m, 2H), 3.35 - 3.33 (m, 1H), 3.31 - 3.27 (m, 1H), 1.98 - 1.90 (m, 1H), 1.80 - 1.63 (m, 3H), 1.61 - 1.47 (m, 1H), 1.38 - 1.21 (m, 3H). ES- LCMS m/z 367.0 [M+H] + .

Final Compounds

Amidation method A syntheses (tandem acid activation then amidation)

Example 1

6-(3 ,4-dimethylphenyl)-N-( 1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)-2-oxo- 1,2- dihydropyridine-3-carboxamide

A solution of 6-(3,4-dimethylphenyl)-2-oxo-l,2-dihydropyridine-3-carboxyli c acid (210 mg, 0.87 mmol) and perfluorophenyl 2,2,2-trifluoroacetate (0.20 m , 1.2 mmol) in pyridine (5 m ) was heated to 50°C. After 4 h, the reaction was cooled to r.t. and 3-amino-2,3-dihydrothiophene 1,1-dioxide, hydrochloride (300 mg, 1.7 mmol) and DIPEA (0.30 mb, 1.7 mmol) were added. After 65 h, the reaction mixture was concentrated then purified by reverse phase chromatography (MeCN / H2O, 0.1% NH4OH modifier, 10%-80% gradient, 30 min run). The fractions containing desired product were combined and concentrated to afford 6-(3,4- dimethylphenyl)-N-( 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)-2-oxo- 1 ,2-dihydropyridine-3- carboxamide (28 mg, 0.078 mmol, 9.0% yield) as a white solid. J H NMR (400 MHz, DMSO- de) 5 ppm 12.62 (br s, 1H), 10.21 - 10.14 (m, 1H), 8.35 (d, J = 7.8 Hz, 1H), 7.63 (br s, 1H), 7.57 - 7.52 (m, 1H), 7.31 - 7.24 (m, 2H), 7.00 - 6.97 (m, 1H), 6.81 (br d, J = 7.8 Hz, 1H), 5.41 - 5.33 (m, 1H), 3.74 (dd, J = 13.7, 7.8 Hz, 1H), 3.29 - 3.22 (m, 1H), 2.32 - 2.26 (m, 6H). ES-LCMS m/z 359.0 [M+H] + .

The following compounds were synthesized in an analogous manner to the preparation described above (amidation method A) using the relevant carboxylic acid precursors from the intermediate synthesis section. Compounds were purified using one of two options: (1) normal phase chromatography (EtOAc / heptane); (2) reverse phase chromatography (MeCN / H2O, basic or acidic modifiers).

Amidation method B syntheses (using isolated perfluorophenyl esters)

Example 25 (R)-6-(3,5-difluorophenyl)-N-(l,l-dioxido-2,3-dihydrothiophe n-3-yl)-2-oxo-l,2- dihydropyridine-3 -carboxamide

Perfluorophenyl 6-(3,5-difluorophenyl)-2-oxo-l,2-dihydropyridine-3-carboxyla te (80 mg, 0.19 mmol) was suspended in DMSO (320 pL) in a vial at r.t. A solution of (R)-3-amino-2,3- dihydrothiophene 1,1-dioxide (26 mg, 0.19 mmol) in DMSO (320 pL) was added and the 5 reaction was stirred for 1 h. Material was then purified by reverse phase purification (MeCN / H2O, 10 mM NH4HCO3 / 0.075% NH4OH modifier, 15%-55% gradient, 17 min run) to afford (R)-6-(3 ,5 -difluorophenyl)-N-( 1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)-2-oxo- 1 ,2- dihydropyridine-3-carboxamide (40 mg, 0.11 mmol, 55 % yield) as a white solid. J H NMR (400 MHz, DMSO-d 6 ) 5 ppm 12.81 (br s, 1H), 10.11 (br s, 1H), 8.38 (d, J = 7.3 Hz, 1H), 7.68 0 - 7.59 (m, 2H), 7.48 (tt, J = 9.2, 2.3 Hz, 1H), 7.28 (dd, J = 6.6, 2.2 Hz, 1H), 7.02 - 6.94 (m,

2H), 5.42 - 5.34 (m, 1H), 3.75 (dd, J = 13.7, 7.8 Hz, 1H), 3.29 (dd, J = 13.9, 4.2 Hz, 1H). ES- LCMS m/z 367.0 [M+H] + .

The following compounds were synthesized in an analogous manner to the preparation 5 described above (amidation method B) using the relevant carboxylic acid precursors from the intermediate synthesis section. In case where HC1 salt of the reactant amine was used, 1.0 eq of DIPEA was added to the reaction mixture. Compounds were purified using one of two options: (1) normal phase chromatography (EtOAc / heptane); (2) reverse phase chromatography (MeCN / H2O, basic or acidic modifiers).

Amidation method C syntheses

Example 42

(R)-6-(4-cyclopropylphenyl)-N-( 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)-2-oxo- 1 ,2- dihydropyridine-3-carboxamide

To a stirred mixture of 6-(4-cyclopropylphenyl)-2-oxo-l,2-dihydropyridine-3-carboxyl ic acid (50 mg, 0.20 mmol) in DMF (1.5 mL) was added a 50% solution of 2,4,6-tripropyl-l,3,5,2,4,6- trioxatriphosphinane 2,4,6-trioxide in DMF (0.17 mL, 0.30 mmol). To this mixture was added DIPEA (0.10 mL, 0.59 mmol) and (R)-3-amino-2,3-dihydrothiophene 1,1-dioxide (31 mg, 0.24 mmol) and the mixture was stirred at r.t. for 18 h. Upon completion, the reaction mixture was diluted with MeOH and purified by reverse phase chromatography (MeCN / H2O, 0.1% formic acid modifier, 30%-99% gradient, 17 min run). The fractions containing desired product 5 were combined and concentrated to afford (R)-6-(4-cyclopropylphenyl)-N-(l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-oxo-l,2-dihydropyridine-3 -carboxamide (25 mg, 0.067 mmol, 33% yield) as a solid. 1 HNMR (400 MHz, DMSO-de) 5 12.66 (s, 1H), 10.16 (br d, J = 7.8 Hz, 1H), 8.35 (d, J = 7.8 Hz, 1H), 7.70 (d, J = 8.3 Hz, 2H), 7.26 (dd, J = 6.8, 2.0 Hz, 1H), 7.24 - 7.20 (m, 2H), 7.00 - 6.96 (m, 1H), 6.81 (d, J = 7.8 Hz, 1H), 5.41 - 5.32 (m, 1H), 3.74 (dd, J = 13.7, 0 7.8 Hz, 1H), 3.26 (dd, J = 13.7, 4.4 Hz, 1H), 2.04 - 1.95 (m, 1H), 1.06 - 1.00 (m, 2H), 0.79 -

0.73 (m, 2H). ES-LCMS m/z 359.0 [M+H] + .

The following compounds were synthesized in an analogous manner to the preparation described above (amidation method C) using the relevant carboxylic acid precursors from the 5 intermediate synthesis section. Compounds were purified using one of two options: (1) normal phase chromatography (EtOAc / heptane); (2) reverse phase chromatography (MeCN / H2O, basic or acidic modifiers).

Amidation method D syntheses

Example 59

N-((R)-l,l-dioxido-2,3-dihydrothiophen-3-yl)-6-(cis-4-met hylcyclohexyl)-2-oxo-l,2- dihydropyridine-3-carboxamide

To a solution of 6-(cis-4-methylcyclohexyl)-2-oxo-l,2-dihydropyridine-3-carbo xylic acid (30 mg, 0.13 mmol) in DCM (1 mL) was added (R)-3-amino-2,3-dihydrothiophene 1,1-dioxide 5 (26 mg, 0.19 mmol), DIEA (0.067 mL, 0.38 mmol) and HATU (53 mg, 0.14 mmol). The reaction mixture was stirred at r.t. for 2 h then quenched with H2O, concentrated, and purified via reverse phase purification (MeCN / H2O, 0.1% formic acid modifier, 30%-99% gradient, 17 min run). Appropriate fractions were combined and concentrated to afford N-((R)-1,1- dioxido-2,3-dihydrothiophen-3-yl)-6-(cis-4-methylcyclohexyl) -2-oxo-l,2-dihydropyridine-3- 0 carboxamide (27 mg, 0.078 mmol, 61 % yield). J H NMR (400 MHz, Methanol -di) 5 8.39 (d,

J = 7.3 Hz, 1H), 7.02 - 6.98 (m, 1H), 6.92 - 6.87 (m, 1H), 6.48 (d, J = 7.8 Hz, 1H), 5.48 - 5.40 (m, 1H), 3.79 - 3.71 (m, 1H), 3.24 (dd, J = 13.9, 4.2 Hz, 1H), 2.65 - 2.55 (m, 1H), 2.00 - 1.89 (m, 1H), 1.85 - 1.63 (m, 6H), 1.61 - 1.51 (m, 2H), 1.05 (d, J = 7.3 Hz, 3H). ES-LCMS m/z 351.1 [M+H] + . 5

The following compounds were synthesized in an analogous manner to the preparation described above (amidation method D) using the relevant carboxylic acid precursors from the intermediate synthesis section. Compounds were purified using one of two options: (1) normal phase chromatography (EtOAc / heptane); (2) reverse phase chromatography (MeCN / H2O, 0 basic or acidic modifiers).

Amidation method E syntheses

Example 95 (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-oxo-6-(4-(tri fluoromethyl)thiophen-2-yl)-

1 ,2-dihydropyridine-3 -carboxamide

Combined 2-oxo-6-(4-(trifluoromethyl)thiophen-2-yl)-,2-dihydropyridin e-3 -carboxylic acid (49 mg, 0.17 mmol) and CDI (41 mg, 0.26 mmol) in N,N-Dimethylformamide (1.0 mb) and let stir at r.t. After 3 hours, added (R)-3-amino-2, 3 -dihydrothiophene 1,1-dioxide, hydrochloride (35 mg, 0.21 mmol) and continued stirring at r.t. After an additional 1 h, diluted reaction mixture with water (10 mL) and EtOAc and the layers were separated. The aqueous layer was extracted again with EtOAc. The combined organic layers were washed with 5 % aq. LiCl, passed through a hydrophobic frit, and concentrated. The residue was dissolved in 1.0 mL DMSO and purified by reverse phase chromatography (Xselect CSH Prep C18 5pM OBD, 5 MeCN / H2O, 0.1 % FA modifier, 30 % to 85 % gradient, 17 minute run). The fractions containing desired product were lyophilized to afford (R)-N-(l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-oxo-6-(4-(trifluoromethyl)thiophen-2 -yl)-l,2-dihydropyridine-3- carboxamide (N80468-59-B1) (12 mg, 0.027 mmol, 16 % yield) as a pale yellow solid. J H NMR (400 MHz, DMSO-de) 5 12.99 (br s, 1H), 9.94 (br s, 1H), 8.33 (d, J= 7.8 Hz, 1H), 8.01 0 (dd, J= 3.9, 1.0 Hz, 1H), 7.88 - 7.81 (m, 1H), 7.26 (dd, J= 6.6, 2.2 Hz, 1H), 7.18 - 7.00 (m,

1H), 6.98 (dd, J= 6.8, 2.9 Hz, 1H), 5.43 - 5.29 (m, 1H), 3.74 (dd, J= 13.7, 8.3 Hz, 1H), 3.30 - 3.25 (m, 1H). ES-LCMS m/z 405.0 [M+H] + .

The following compounds were synthesized in an analogous manner to the preparation described above (amidation method E) using the relevant carboxylic acid precursors from the 5 intermediate synthesis section. Compounds were purified using one of two options: (1) normal phase chromatography (EtOAc / heptane); (2) reverse phase chromatography (MeCN / H2O, basic or acidic modifiers).

Syntheses via Suzuki Coupling Late-Stage Functionalization 0

Typical procedure A

Example 61 (R)-N-( 1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)-2-oxo-6-phenyl- 1 ,2-dihydropyridine-3 - carboxamide

Phenylboronic acid (0.036 g, 0.30 mmol), (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-6- iodo-2 -oxo- l,2-dihydropyridine-3 -carboxamide (0.075 g, 0.20 mmol), and P(t-Bn)? Pd G4 (0.020 g, 0.020 mmol) were dissolved in 1,4-dioxane (1.6 mb) under N2. Next, 2 M aqueous CS2CO3 (0.40 mb, 0.79 mmol) was added, and the reaction mixture heated to 45 °C for 16 h. The reaction mixture was cooled to r.t., diluted with DCM, filtered, and concentrated. The crude mixture was purified by normal phase chromatography (EtOAc / heptane, 50%-100%) to afford (R)-N-(l, l-dioxido-2,3-dihydrothiophen-3-yl)-2-oxo-6-phenyl-l,2-dihyd ropyridine- 3-carboxamide (0.045 g, 0. 14 mmol, 69% yield) as a white solid. J H NMR (400 MHz, DMSO- d 6 ) 5 12.76 (br s, 1H), 10.17 (br d, J = 7.3 Hz, 1H), 8.38 (d, J = 7.8 Hz, 1H), 7.85 - 7.75 (m, 2H), 7.60 - 7.49 (m, 3H), 7.26 (dd, J = 6.4, 2.0 Hz, 1H), 7.02 - 6.95 (m, 1H), 6.84 (br d, J = 7.8 Hz, 1H), 5.44 - 5.30 (m, 1H), 3.74 (dd, J = 13.9, 8.1 Hz, 1H), 3.27 (dd, J = 13.7, 4.4 Hz, 1H). ES-LCMS m/z 331.0 [M+H] + .

Typical procedure B

Example 117

(R)-N-( 1 , 1 -dioxido-2,3-dihydrothiophen-3-yl)-5-hydroxy-6-(4-methoxyphe nyl)-2-oxo- 1 ,2- dihydropyridine-3-carboxamide

A solution containing of APhos Pd G3 (0.0039 g, 6.1 pmol) and (R)-6-bromo-N-(l,l-dioxido- 2,3-dihydrothiophen-3-yl)-2-methoxy-5- (methoxymethoxy)nicotinamide (0.050 g, 120 pmol) was prepared in 0.75 mL of 1,4-dioxane under dry nitrogen gas. This solution was added to a glass vial containing a magnetic stirrer and 4-methoxyphenyl boronic acid (0.028 g, 180 pmol). Next, CS2CO3 (0.10 g, 153 pL, 2 M, 310 pmol) was added and the vial was capped and stirred over 16 h at 50 °C. At 16 h, the mixtures were diluted with an equal volume of DCM and passed through a phase separator. The resulting mixtures were concentrated, and the resulting residue re-dissolved in acetonitrile (490 pL). TMS-I (0.17 g, 860 pmol) was added. The resulting mixture was heated at 50 °C for 5 min and then quenched with 50 uL of water followed by dissolution in 100 uL of DMSO. Next, purification was performed by reverse phase chromatography (5-100% MeCN/H2O, formic acid modifier). Fractions containing desired product were combined and concentrated to afford (R)-N-(l,l-dioxido-2,3- dihydrothiophen-3 -yl)-5 -hydroxy-6-(4-methoxyphenyl)-2-oxo- 1 ,2-dihydropyridine-3 - carboxamide (0.015 g, 39 pmol, 32% yield) as a white solid. J H NMR (700 MHz, DMSO-de) 5 12.33 - 12.19 (m, 1H), 10.55 - 10.43 (m, 1H), 9.42 (br s, 1H), 8.17 (br s, 1H), 7.69 (br s, 2H), 7.26 (dd, J = 6.5, 1.7 Hz, 1H), 7.04 (d, J= 9.0 Hz, 2H), 6.98 (dd, J = 6.5, 3.0 Hz, 1H), 5.41 - 5.32 (m, 1H), 3.82 (s, 3H), 3.74 (dd, J= 13.8, 7.7 Hz, 1H), 3.24 (dd, J= 14.0, 4.1 Hz, 1H). ES-LCMS m/z 377.1 [M+H] + .

The following compounds were synthesized in an analogous manner to the preparation described above (Suzuki Coupling Late-Stage Functionalization) using the relevant boronic acid or boronate ester intermediates. Compounds were purified using one of two options: (1) normal phase chromatography (EtOAc / heptane); (2) reverse phase chromatography (MeCN / H2O, basic or acidic modifiers).

Final compound synthesis from final step deprotection of methoxy-pyridine precursors

Note: TMS-I was typically used as the deprotecting agent as described below, but other agents are also applicable e.g. BBrs, BCE, HC1, and in situ preparation of TMS-I from TMS-C1 and Nal. An example procedure is shown below.

Example 70

(R)-5-cyano-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-ox o-6-phenyl-l,2-dihydropyridine- 3 carboxamide

To a stirred solution of (R)-5-cyano-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-metho xy-6- phenylnicotinamide (91 mg, 0.25 mmol) in MeCN (2 m ) was added a freshly prepared solution of 1 M TMS-I (0.75 m , 0.75 mmol) in DCM and the reaction stirred at r.t. for 17 h. The mixture was then diluted with MeOH (0.4 mb) and stirred for 10 min. Finally, Et2O was added, and the resulting suspension stirred for 5 min, then filtered and dried to afford (R)-5- cyano-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-2-oxo-6-pheny l-l,2-dihydropyridine-3 carboxamide (70 mg, 0.20 mmol, 80% yield) as a beige solid. J H NMR (400 MHz, DMSO-de) 5 ppm 13.53 (br s, 1H), 9.73 (d, J = 7.8 Hz, 1H), 8.47 (s, 1H), 7.76 - 7.70 (m, 2H), 7.68 - 7.57 (m, 3H), 7.27 (dd, J = 6.4, 2.0 Hz, 1H), 6.97 (dd, J = 6.8, 2.9 Hz, 1H), 5.43 - 5.33 (m, 1H), 3.74 (dd, J = 13.7, 7.8 Hz, 1H), 1H under H2O peak. ES-LCMS m/z 356.0 [M+H] + .

The following compounds were synthesized in an analogous manner to the preparation described above (final step deprotections) using the relevant methoxy-pyridine precursors from the intermediate synthesis section. In some cases, compounds were purified using one of two options: (1) normal phase chromatography (EtOAc / heptane); (2) reverse phase chromatography (MeCN / H2O, basic or acidic modifiers).

Other final compound syntheses

Example 107 : (R)-N-( 1 , 1 -dioxido-2,3 -dihydrothiophen-3 -yl)-5 -hydroxy-2 -oxo-6-phenyl- 1,2- dihydropyridine-3-carboxamide

In a glass vial equipped with a magnetic stirrer was weighed out (R)-6-bromo-N-(l, 1-dioxido- 2,3-dihydrothiophen-3-yl)-2-methoxy-5-(methoxymethoxy)nicoti namide (0.10 g, 250 pmol), 4,4,5,5-tetramethyl-2-phenyl-l,3,2-dioxaborolane (75 mg, 370 pmol), and APhos Pd G3 (7.8 mg, 12 pmol). The contents of the vial were purged with dry nitrogen gas and then 1,4- dioxane (1.3 m ) was added under nitrogen gas. Next, 2 M aqueous cesium carbonate solution (200 mg, 310 pL, 610 pmol) was added and the vial was capped and stirred at 50 °C over 16 h. At 16 h, the mixture was diluted with DCM (equal volume, 1.3 mL) and passed through a disposable phase separator. The vial was rinsed again with DCM (1.3 mL) and this mixture was also passed through the same phase separator. The resulting organic mixture was concentrated and the resulting residue was dissolved in acetonitrile (1.25 mL) and then TMS-I (24 mg, 167 pL, 5 Eq, 1.23 mmol) was added in one portion. The resulting mixture was heated to 50 °C for 2 h. Additional TMS-I (49 mg, 250 pmol) was added and the mixture stirred for an additional 5 min at 50 °C. At this point, LCMS indicated completion of the reaction. The mixture was quenched with the addition of water (100 uL) and combined with DMSO (700 uL). This solution was purified via reverse phase chromatography (MeCN / H2O, 0.1% formic acid modifier, 30%-100% gradient, 30 min run). The desired fractions were combined and lyophilized to afford (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-5-hydroxy-2-oxo -6- phenyl-l,2-dihydropyridine-3-carboxamide (75 mg, 0.21 mmol, 86 % yield) as a light yellow solid. 1 HNMR (400 MHz, DMSO-de) 5 12.35 (br s, 1H), 10.47 (br s, 1H), 9.44 (br s, 1H), 8.20 (s, 1H), 7.75 - 7.63 (m, 2H), 7.52 - 7.42 (m, 3H), 7.26 (dd, J= 6.4, 2.0 Hz, 1H), 6.98 (dd, J = 6.8, 2.9 Hz, 1H), 5.43 - 5.30 (m, 1H), 3.74 (dd, J= 13.7, 7.8 Hz, 1H), 3.24 (dd, J= 13.7, 3.9 Hz, 1H). ES-LCMS m/z 347.2 [M+H] + . Example 78: (R)-5-(difluoromethyl)-N-(l,l-dioxido-2,3-dihydrothiophen-3- yl)-2-oxo-6- phenyl- 1 ,2-dihydropyridine-3 -carboxamide

To a solution of (R)-N-(l,l-dioxido-2,3-dihydrothiophen-3-yl)-5-formyl-2-oxo- 6-phenyl-l,2- dihydropyridine -3 -carboxamide (130 mg, 0.35 mmol) in DCM (6 m ) under nitrogen at 0°C was added N,N-diethyl-l,l,l-trifluoro-14-sulfanamine (0.28 m , 2.1 mmol), dropwise. The reaction mixture was stirred at r.t. for 32 h. The reaction mixture was diluted with water (30 mb) and extracted with DCM (4 x 15 mb). The combined organic phase was dried over sodium sulphate and concentrate in vacuo to get the crude product. The crude mixture was purified using reverse phase purification (20% MeCN in H2O, modified by TP A) over 17 min. Fractions containing product concentrated to obtain (R)-5-(difluoromethyl)-N-(l,l-dioxido-2,3- dihydrothiophen-3-yl)-2-oxo-6-phenyl-l,2-dihydropyridine-3-c arboxamide (33 mg, 0.085 mmol, 24% yield) was obtained as an off-white solid. J H NMR (400 MHz, DMSO-de) 5 13.07 (br s, 1H), 9.97 (br d, J= 7.5 Hz, 1H), 8.51 (s, 1H), 7.65 - 7.55 (m, 3H), 7.54 - 7.49 (m, 2H), 7.28 (dd, J= 6.5, 2.0 Hz, 1H), 6.99 (dd, J= 7.0, 3.0 Hz, 1H), 6.73 - 6.42 (m, 1H), 5.43 - 5.35 (m, 1H), 3.75 (dd, J= 13.8, 7.8 Hz, 1H), 3.30 - 3.26 (m, 1H). ES-ECMS m/z 381.0 [M+H] + .

Biological Examples

Example A

Functional WRN unwinding activity can be measured using a Anorogenic plate based 384 well assay configured to measure the separation of labeled double stranded DNA substrate. Compounds were dosed out in neat DMSO with a 1:3 serial dilution scheme. 100 nF of compound was stamped into Greiner low volume black assay plates (Greiner Cat#784076) using the Echo Acoustic Dispenser to generate assay ready plates. All solutions were prepared in assay buffer (25 mM TRIS (pH8.0), 5 mM NaCl, 2 mM MgCh, 1 mM DTT, 0.05% BSA) for this 10 pb low volume reaction. To prepare the solutions, a 2X WRN Enzyme cocktail was made containing 200 pM of recombinant full-length WRN protein (1- 1432). A 2X Substrate cocktail was made to consist of both 200 pM ATP (any ultrapure ATP sample) and 12 nM of the Auorescent quenched labeled double stranded DNA oligomer (IDT Custom synthesis; 5'-5IABkFQ (SEQ ID NO. 1)/GCA CTG GCC GTC GTT TTA CGG TCG TGA CT-3' (SEQ ID NO. 2): 5'-TTT TTT ACT TAA CGA CGG CCA GTG C (SEQ ID NO. 3)/36-TAMTSP/-3' (SEQ ID NO. 4)). To start the reaction, 5 uL of assay buffer was added to a single column to serve as the low control. Following this, 5pL of 2X WRN Enzyme was added in all wells except the buffer low control wells. The reaction plate was covered and incubated at ambient temperature for 4 hours to allow for time dependent inhibition if it existed. After 4 hours, the addition of 5 pL of 2X-ATP/DNA substrate cocktail was added across all wells of the assay plate. This initiated the reaction, as the plate was incubated at ambient temperature for 60 minutes for the unwinding reaction to occur. A 10 mM EDTA solution was prepared and added at 5 pL across the entire plate after 60 minutes to quench the samples for an endpoint measurement. Fluorescent intensity was measured using excitation and emission wavelengths of 525 nm and 598 nm, respectively. High florescent intensity (DMSO with buffer) represents full inhibition of unwinding activity and low florescent intensity (DMSO with enzyme) represents no inhibition of unwinding activity. The potency of the compounds was determined using a four-parameter inhibition model to generate pICso, Hill Slope, maximum inhibition.

The pICso data of the compounds in Table 1 above are disclosed in Table 2 below. (4.0 < A < 5.0; 5.0 < B < 6.0; 6.0 < C < 7.0; 7.0 < D)

Table 2 DNA Unwinding Assay

Example B

Reversibility of inhibition by certain compounds in the present disclosure was determined utilizing reagents and equipment from the functional WRN unwinding, Anorogenic assay described in Example A, specifically using the assay buffer, recombinant full-length WRN protein, ATP, and Auorescent quenched labeled double stranded DNA oligomer (DNA). Reversibility of inhibition was determined by 128-fold dilution enzyme and inhibitor from a pre-incubation reaction into assay buffer containing the DNA and ATP substrates. Reversible inhibition is inferred by a regain of enzymatic activity over a monitored reaction time course; irreversible inhibition is inferred by a failure to regain enzymatic activity. The compounds were serially diluted 1:2 in neat DMSO and 100 nL dispensed into Greiner low volume black assay plates for the pre-incubation reaction. Ten nanoliters of WRN enzyme was added to the compounds and allowed to react at RT for 30 min. Assay buffer was added to a single row to serve as the no-enzyme control. After incubation 0.85 pL of enzyme/inhibitor solution was transferred with mixing to 108 pL of substrate cocktail in a NUNC black 384, 120 pL volume plate (Cat#262260) containing 100 pM ATP and 60 nM DNA. Fluorescent intensity was measured using excitation and emission wavelengths of 525 nm and 598 nm, respectively over 500 minutes. The compounds showed dose-dependent inhibition relative to the no-inhibitor control reactions and several concentrations showed complete inhibition. No regain of activity was seen in the 500 minute time courses for any of the compounds, and it was therefore concluded that these compounds are irreversible inhibitors of the WRN enzyme.

Certain compounds in the present disclosure were also tested in cell viability assays to investigate the effect of their WRN inhibition on cell health. Colorectal (SW) and endometrial (HEC) cancer cell lines were employed for these studies. In either cell type, the compounds were tested in MSS and MSI-H cell lines. The differential growth inhibition in MSI-H, which is more potent compared to that in MSS cell lines, demonstrates the specificity of WRN inhibition from these compounds. All patent applications, patents, and printed publications cited herein are incorporated herein by reference in the entireties, except for any definitions, subject matter disclaimers or disavowals, and except to the extent that the incorporated material is inconsistent with the express disclosure herein, in which case the language in this disclosure controls.