Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NEW COMPOUNDS
Document Type and Number:
WIPO Patent Application WO/2022/063876
Kind Code:
A1
Abstract:
The invention relates to novel compounds for use as inhibitors of NLRP3 inflammasone production, wherein such compounds are as defined by compounds of formula (I) and wherein the integers R1, R2 and R3 are defined in the description, and where the compounds may be useful as medicaments, for instance for use in the treatment of a disease or disorder that is associated with NLRP3 inflammasome activity.

Inventors:
OEHLRICH DANIEL (BE)
VAN GOOL MICHIEL (ES)
LEENAERTS JOSEPH (BE)
MURATORE MICHAEL (BE)
TRESADERN GARY (BE)
LAMKANFI MOHAMED (BE)
VAN OPDENBOSCH NINA (BE)
HENDRICKS ROBERT (US)
Application Number:
PCT/EP2021/076164
Publication Date:
March 31, 2022
Filing Date:
September 23, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
JANSSEN PHARMACEUTICA NV (BE)
International Classes:
C07D237/28; A61K31/502; A61P1/16; A61P3/10; A61P9/04; A61P13/12; A61P19/02; A61P25/00; A61P29/00; A61P35/00; C07D401/12; C07D403/12; C07D471/04
Domestic Patent References:
WO2020021447A12020-01-30
WO2019007696A12019-01-10
WO2020018975A12020-01-23
WO2020037116A12020-02-20
WO2020021447A12020-01-30
WO2020010143A12020-01-09
WO2019079119A12019-04-25
WO2019166621A12019-09-06
WO2019121691A12019-06-27
WO2019007696A12019-01-10
Other References:
AMES D. E. ET AL: "1128. Cinnolines. Part VII. Methylation of 4-hydroxy-6,7-dimethoxycinnoline-3-acetic acid", JOURNAL OF THE CHEMICAL SOCIETY, 1 January 1965 (1965-01-01), pages 6036, XP055849674, ISSN: 0368-1769, DOI: 10.1039/jr9650006036
VAN OPDENBOSCH NLAMKANFI M., IMMUNITY, vol. 50, no. 6, 18 June 2019 (2019-06-18), pages 1352 - 1364
TARTEY SKANNEGANTI TD, IMMUNOLOGY, vol. 156, no. 4, April 2019 (2019-04-01), pages 329 - 338
KELLEY ET AL., INTJMOL SCI, vol. 20, 6 July 2019 (2019-07-06), pages 13
FUNG ET AL., EMERGMICROBES INFECT, vol. 9, no. 1, 14 March 2020 (2020-03-14), pages 558 - 570
YANG Y ET AL., CELL DEATH DIS, vol. 10, no. 2, 12 February 2019 (2019-02-12), pages 128
SHARIF ET AL., NATURE, vol. 570, no. 7761, June 2019 (2019-06-01), pages 338 - 343
MIYAMAE T: "Cryopyrin-associated Periodic Syndrome", PAEDIATR DRUGS, vol. 14, no. 2, 1 April 2012 (2012-04-01), pages 109 - 17
SZABO GPETRASEK J., NAT REV GASTROENTEROL HEPATOL, vol. 12, no. 7, July 2015 (2015-07-01), pages 387 - 400
ZHEN YZHANG H, FRONT IMMUNOL, vol. 10, 28 February 2019 (2019-02-28), pages 276
WALLE L ET AL., NATURE, vol. 512, no. 7512, 7 August 2014 (2014-08-07), pages 69 - 73
KNAUF ET AL., KIDNEY INT, vol. 84, no. 5, November 2013 (2013-11-01), pages 895 - 901
KRISHNAN ET AL., BR J PHARMACOL, vol. 173, no. 4, February 2016 (2016-02-01), pages 752 - 65
SHAHZAD ET AL., KIDNEY INT, vol. 87, no. 1, January 2015 (2015-01-01), pages 74 - 84
SARKAR ET AL., NPJ PARKINSONS DIS, vol. 3, 17 October 2017 (2017-10-17), pages 30
RIDKER ET AL., NENGL JMED, vol. 377, no. 12, 21 September 2017 (2017-09-21), pages 1119 - 1131
TOLDO SABBATE A, NAT REV CARDIOL, vol. 15, no. 4, April 2018 (2018-04-01), pages 203 - 214
KELLY ET AL., BR J DERMATOL, vol. 173, December 2015 (2015-12-01), pages 6
NIETO-TORRES ET AL., VIROLOGY, vol. 485, November 2015 (2015-11-01), pages 330 - 9
DOYLE ET AL., NATMED, vol. 18, no. 5, May 2012 (2012-05-01), pages 791 - 8
RIDKER ET AL., LANCET, vol. 390, no. 10105, 21 October 2017 (2017-10-21), pages 1833 - 1842
DERANGERE ET AL., CELL DEATH DIFFER, vol. 21, no. 12, December 2014 (2014-12-01), pages 1914 - 24
BASIORKA ET AL., LANCET HAEMATOL, vol. 5, no. 9, September 2018 (2018-09-01), pages e393 - e402
ZHANG ET AL., HUM IMMUNOL, vol. 79, no. 1, January 2018 (2018-01-01), pages 57 - 62
BUNDEGAARD, H.: "Design of Prodrugs", 1985, ELESEVIER, pages: 1 - 92
T.W. GREENEP.G.M. WUTZ: "Protective Groups in Organic Synthesis", 1999, WILEY-INTERSCIENCE
MENU ET AL., CLINICAL AND EXPERIMENTAL IMMUNOLOGY, vol. 166, 2011, pages 1 - 15
HENAO-MEIJA ET AL., NATURE, vol. 482, 2012, pages 179 - 185
OZAKI ET AL., J. INFLAMMATION RESEARCH, vol. 8, 2015, pages 15 - 27
SCHRODER ET AL., CELL, vol. 140, 2010, pages 821 - 832
COOK ET AL., EUR. J. LMMUNOL., vol. 40, 2010, pages 595 - 653
BRADDOCK ET AL., NAT. REV. DRUG DISC., vol. 3, 2004, pages 1 - 10
INOUE ET AL., IMMUNOLOGY, vol. 139, 2013, pages 11 - 18
COLL ET AL., NAT. MED., vol. 21, no. 3, 2015, pages 248 - 55
SCOTT ET AL., CLIN. EXP. RHEUMATOL., vol. 34, no. 1, 2016, pages 88 - 93
LU ET AL., J. IMMUNOL., vol. 198, no. 3, 2017, pages 1119 - 29
ARTLETT ET AL., ARTHRITIS RHEUM., vol. 63, no. 11, 2011, pages 3563 - 74
DE NARDO ET AL., AM. J. PATHOL., vol. 184, 2014, pages 42 - 54
KIM ET AL., AM. J. RESPIR. CRIT. CARE MED, vol. 196, no. 3, 2017, pages 283 - 97
WALSH ET AL., NATURE REVIEWS, vol. 15, 2014, pages 84 - 97
DUEWELL, BEHAV. LMMUN., vol. 61, 2017, pages 306 - 16
ZHANG ET AL., J. STROKE AND CEREBROVASCULAR DIS., vol. 24, no. 5, 2015, pages 972 - 9
ISMAEL ET AL., J. NEUROTRAUMA., vol. 35, no. 11, 2018, pages 1294 - 1303
WEN ET AL., NATURE IMMUNOLOGY, vol. 13, 2012, pages 352 - 357
DUEWELL ET AL., NATURE, vol. 464, 2010, pages 1357 - 1361
STROWIG ET AL., NATURE, vol. 481, 2014, pages 278 - 286
MRIDHA ET AL., J. HEPATOL., vol. 66, no. 5, 2017, pages 1037 - 46
VAN HOUT ET AL., EUR. HEART J., vol. 38, no. 11, 2017, pages 828 - 36
SANO ET AL., J. AM. COLL. CARDIOL., vol. 71, no. 8, 2018, pages 875 - 66
WU ET AL., ARTERIOSC/ER. THROMB. VASE. BIOL., vol. 37, no. 4, 2017, pages 694 - 706
RIDKER ET AL., N. ENGL. J. MED., vol. 377, no. 12, 2017, pages 1119 - 31
DOYLE ET AL., NATURE MEDICINE, vol. 18, 2012, pages 791 - 798
TARALLO ET AL., CELL, vol. 149, no. 4, 2012, pages 847 - 59
LOUKOVAARA ET AL., ACTA OPHTHALMOL., vol. 95, no. 8, 2017, pages 803 - 8
PUYANG ET AL., SCI. REP., vol. 6, 2016, pages 20998
PRIMIANO ET AL., J. LMMUNOL., vol. 197, no. 6, 2016, pages 2421 - 33
FANG ET AL., J DERMATOL SCI., vol. 83, no. 2, 2016, pages 116 - 23
NIEBUHR ET AL., ALLERGY, vol. 69, no. 8, 2014, pages 1058 - 67
ALIKHAN ET AL., J. AM. ACAD. DERMATOL., vol. 60, no. 4, 2009, pages 539 - 61
JAGER ET AL., AM. J. RESPIR. CRIT. CARE MED., vol. 191, 2015, pages A5816
BRADDOCK ET AL., NAT. REV. DRUG DISC, vol. 3, 2004, pages 1 - 10
GUGLIANDOLO ET AL., INT. J. MOL. SCI., vol. 19, no. 7, 2018, pages E1992
LANNITTI ET AL., NAT. COMMUN., vol. 7, 2016, pages 10791
GRANATA ET AL., PLOS ONE, vol. 10, no. 3, 2015, pages eoi22272
NEUDECKER, J. EXP. MED., vol. 214, no. 6, 2017, pages 1737 - 52
LAZARIDIS ET AL., DIG. DIS. SCI., vol. 62, no. 9, 2017, pages 2348 - 56
DOLUNAY ET AL., INFLAMMATION, vol. 40, 2017, pages 366 - 86
TATE ET AL., SCI REP., vol. 10, no. 6, 2016, pages 27912 - 20
NOVIAS ET AL., PLOS PATHOGENS, vol. 13, no. 2, 2017, pages el006196
RIDKER ET AL., LANCET, vol. 390, no. 10105, 2017, pages 1833 - 42
WANG ET AL., ONCO/ REP., vol. 35, no. 4, 2016, pages 2053 - 64
BASIORKA ET AL., BLOOD, vol. 128, no. 25, 2016, pages 2960 - 75
LI ET AL., AM. J. CANCER RES., vol. 5, no. 1, 2015, pages 442 - 9
ALLEN ET AL., J. EXP. MED., vol. 207, no. 5, 2010, pages 1045 - 56
HU ET AL., PNAS, vol. 107, no. 50, 2010, pages 21635 - 40
LI ET AL., HEMATOLOGY, vol. 21, no. 3, 2016, pages 144 - 51
HUANG ET AL., J. EXP. CLIN. CANCER RES., vol. 36, no. 1, 2017, pages 116
JIA ET AL., MOL. PAIN., vol. 13, 2017, pages 1 - 11
YAN-GANG ET AL., CELL DEATH AND DISEASE, vol. 8, no. 2, 2017, pages 2579
ALEXANDER ET AL., HEPATOLOGY, vol. 59, no. 3, 2014, pages 898 - 910
BALDWIN ET AL., J. MED. CHEM., vol. 59, no. 5, 2016, pages 1691 - 1710
ZHEN ET AL., NEUROIMMUNOLOGY NEUROINFLAMMATION, vol. 1, no. 2, 2014, pages 60 - 65
MATTIA ET AL., J. MED. CHEM., vol. 57, no. 24, 2014, pages 10366 - 82
SATOH ET AL., CELL DEATH AND DISEASE, vol. 4, 2013, pages 644
"Remington The Science and Practice of Pharmacy", 2013, PHARMACEUTICAL PRESS, pages: 1049 - 1070
Attorney, Agent or Firm:
PUREWAL, Savroop (BE)
Download PDF:
Claims:
Claims 1. A compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein: R1 represents: (i) C3-6 cycloalkyl optionally substituted with one or more substituents independently selected from -OH, -C1-3 alkyl and hydroxyC1-3alkyl; (ii) aryl or heteroaryl, each of which is optionally substituted with 1 to 3 substituents independently selected from halo, -CN, =O, -OH, -O-C1-3 alkyl, -C1-3 alkyl, C3-6 cycloalkyl, haloC1-3alkyl, hydroxyC1-3 alkyl, C1-3 alkoxy, haloC1-3alkoxy and -S(O)2C1-4 alkyl; or (iii) heterocyclyl, optionally substituted with 1 to 3 substituents independently selected from =O, C1-3 alkyl and C3-6 cycloalkyl; R2 represents: (i) C1-3 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC1-3 alkyl; (ii) C3-6 cycloalkyl; or (iii) C2-4 alkenyl optionally substituted with -OC1-3 alkyl; R3 represents: (i) hydrogen; (ii) halo; (iii) C1-4 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC1-3 alkyl; (iv) C2-4 alkenyl optionally substituted with -OC1-3 alkyl; (v) C3-6 cycloalkyl; or (vi) -OC1-3 alkyl.

2. The compound of claim 1, wherein R1 represents C3-6 cycloalkyl optionally substituted by one or two substituents selected from C1-3 alkyl, -OH and hydroxyCi-salkyl.

3. The compound of claim 2, wherein R1 represents: where each Rla represents one or two optional substituents selected from -OH, C1-3 alkyl and hydroxyCi-salkyl.

4. The compound of claim 1, wherein R1 represents: (i) phenyl; (ii) a 6-membered monocyclic heteroaryl group; or (iii) a 9- or 10-membered bicyclic heteroaryl group, all of which are optionally substituted with one or two substituent(s) selected from halo, =0, - OH, C1-3 alkyl, -OC1-3 alkyl and -haloCi-salkyl.

5. The compound of claim 4, wherein R1 represents phenyl or a mono-cyclic 6-membered heteroaryl group: wherein Rlb represents one or two optional substituents selected from halo (e.g. fluoro, iodo), =0, -OH, C1-3 alkyl (e.g. methyl), haloCi-salkyl (e.g. -CF3), and, either one or two of Rb, Rc, Rd, Re and Rf represent(s) a nitrogen heteroatom (and the others represent a CH).

6. The compound of claim 4, wherein R1 represents a 9- or 10-membered bicyclic heteroaryl group, for instance: - 165 - wherein Rlb represents one or two optional substituents selected from halo, =0, C1-3 alkyl (e.g. methyl) and haloCi-salkyl (e.g. -CF3), at least one of the rings of the bicyclic system is aromatic (as depicted), Rk represents a N or C atom, Rg represents a N or C atom and any one or two of Rh, Ri and Rj represents N and the other(s) represent(s) C.

7. The compound of any one of claims 1 to 6, wherein R2 represents: (i) C1-3 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC1-2 alkyl; (ii) C3-6 cycloalkyl; or (iii) C2-4 alkenyl optionally substituted with - OC1-2 alkyl.

8. The compound of claim 7, wherein R2 represents unsubstituted C1-3 alkyl.

9. The compound of any one of claims 1 to 8, wherein R3 represents (i) halo; (ii) C1-4 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC1-2 alkyl; or (iii) C3-6 cycloalkyl.

10. The compound of any one of claims 1 to 9, wherein R3 represents: halo (e.g. bromo); C1-3 alkyl optionally (and preferably) substituted by one or more fluoro atoms (so forming, e.g. -CF3); or C3-6 (e.g. C3-4) cycloalkyl (e.g. cylopropyl).

11. A pharmaceutical composition comprising a therapeutically effective amount of a compound as defined in any one of claims 1 to 10 and a pharmaceutically acceptable carrier. - 166 -

12. A process for preparing a pharmaceutical composition as defined in claim 11, characterized in that a pharmaceutically acceptable carrier is intimately mixed with a therapeutically effective amount of a compound as defined in any one of claims

1 to 10.

13. A compound as claimed in any one of claims 1 to 10, for use as a pharmaceutical or medicament.

14. A combination comprising: (a) a compound according to any one of claims 1 to 10; and (b) one or more other therapeutic agents.

15. The compound according to any one of claims 1 to 10, composition according to claim 11 or combination according to claim 14, for use in the treatment of a disease or disorder that is associated with inhibition of NLRP3 inflammasome activity.

16. A method of treating a disease or disorder associated with inhibition of NLRP3 inflammasome activity in a subject in need thereof, the method comprising administering to said subject a therapeutically effective amount of a compound according to any one of claims 1 to 10, a composition according to claim 11 or a combination according to claim 14.

17. The compound, composition or combination for use according to claim 15, or the method of treating according to claim 16 wherein the disease or disorder associated with inhibition of NLRP3 inflammasome activity is selected from inflammasome related diseases and disorders, immune diseases, inflammatory diseases, auto-immune diseases, auto-inflammatory fever syndromes, cryopyrin-associated periodic syndrome, chronic liver disease, viral hepatitis, non-alcoholic steatohepatitis, alcoholic steatohepatitis, alcoholic liver disease, inflammatory arthritis related disorders, gout, chondrocalcinosis, osteoarthritis, rheumatoid arthritis, chronic arthropathy, acute arthropathy, kidney related disease, hyperoxaluria, lupus nephritis, Type I and Type II diabetes, nephropathy, retinopathy, hypertensive nephropathy, hemodialysis related inflammation, neuroinflammation-related diseases, multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, Alzheimer’s disease, cardiovascular diseases, metabolic diseases, cardiovascular risk reduction, hypertension, atherosclerosis, peripheral artery disease, acute heart failure, inflammatory skin diseases, acne, wound healing and scar formation, asthma, sarcoidosis, age-related macular degeneration, colon cancer, lung cancer, myeloproliferative neoplasms, leukemias, myelodysplastic syndromes and myelofibrosis. - 167 -

18. A process for the preparation of a compound of formula (I) as claimed in any of claims 1 to 10, which comprises:

(i) reaction of a compound of formula (II), or a derivative thereof, wherein R2 and R3 are as defined in claim 1, with a compound of formula (III),

H2N-R (III) or a derivative thereof, wherein R1 is as defined in claim 1, under amide-forming reaction conditions;

(ii) reaction of a compound of formula (IV), wherein R1 and R3 are as defined in claim 1, with a compound of formula (V),

R2-LGa (V) wherein LGa represents a suitable leaving group and R2 is as defined in claim 1;

(iii) by transformation of a certain compound of formula (I) into another.

19. A compound of formula (II), as depicted in Claim 18:

Description:
NEW COMPOUNDS

FIELD OF THE INVENTION

The present invention relates to novel compounds that are useful as inhibitors of NOD-like receptor protein 3 (NLRP3) inflammasome pathway. The present invention also relates to processes for the preparation of said compounds, pharmaceutical compositions comprising said compounds, methods of using said compounds in the treatment of various diseases and disorders, and medicaments containing them, and their use in diseases and disorders mediated by NLRP3.

BACKGROUND OF THE INVENTION

Inflammasomes, considered as central signalling hubs of the innate immune system, are multi-protein complexes that are assembled upon activation of a specific set of intracellular pattern recognition receptors (PRRs) by a wide variety of pathogen- or danger- associated molecular patterns (PAMPs or DAMPs). To date, it was shown that inflammasomes can be formed by nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) and Pyrin- and HIN200-domain-containing proteins (Van Opdenbosch N and Lamkanfi M. Immunity, 2019 Jun 18;50(6): 1352-1364). The NLRP3 inflammasome is assembled upon detection of environmental crystals, pollutants, host-derived DAMPs and protein aggregates (Tartey S and Kanneganti TD. Immunology, 2019 Apr;156(4):329-338). Clinically relevant DAMPs that engage NLRP3 include uric acid and cholesterol crystals that cause gout and atherosclerosis, amyloid-P fibrils that are neurotoxic in Alzheimer’s disease and asbestos particles that cause mesothelioma (Kelley et al., IntJMol Sci, 2019 Jul 6;20(13)). Additionally, NLRP3 is activated by infectious agents such as Vibrio cholerae; fungal pathogens such as Aspergillus fumigatus and Candida albicans,' adenoviruses, influenza A virus and SARS-CoV-2 (Tartey and Kanneganti, 2019 (see above); Fung et al. Emerg Microbes Infect, 2020 Mar 14;9(l):558-570).

Although the precise NLRP3 activation mechanism remains unclear, for human monocytes, it has been suggested that a one-step activation is sufficient while in mice a two- step mechanism is in place. Given the multitude in triggers, the NLRP3 inflammasome requires add-on regulation at both transcriptional and post-transcriptional level (Yang Y et al., Cell Death Dis, 2019 Feb 12;10(2):128).

The NLRP3 protein consists of an N-terminal pyrin domain, followed by a nucleotide-binding site domain (NBD) and a leucine-rich repeat (LRR) motif on C-terminal end (Sharif et al., Nature, 2019 Jun; 570(7761):338-343). Upon recognition of PAMP or DAMP, NLRP3 aggregates with the adaptor protein, apoptosis-associated speck -like protein (ASC), and with the protease caspase- 1 to form a functional inflammasome. Upon activation, procaspase-1 undergoes autoproteolysis and consequently cleaves gasdermin D (Gsdmd) to produce the N-terminal Gsdmd molecule that will ultimately lead to pore-formation in the plasma membrane and a lytic form of cell death called pyroptosis. Alternatively, caspase-1 cleaves the pro-inflammatory cytokines pro-IL-1 P and pro-IL-18 to allow release of its biological active form by pyroptosis (Kelley et al., 2019 - see above).

Dysregulation of the NLRP3 inflammasome or its downstream mediators are associated with numerous pathologies ranging from immune/inflammatory diseases, auto- immune/auto-inflammatory diseases (Cryopyrin-associated Periodic Syndrome (Miyamae T. Paediatr Drugs, 2012 Apr 1;14(2): 109-17); sickle cell disease; systemic lupus erythematosus (SLE)) to hepatic disorders (eg. non-alcoholic steatohepatitis (NASH), chronic liver disease, viral hepatitis, alcoholic steatohepatitis, and alcoholic liver disease) (Szabo G and Petrasek J. Nat Rev Gastroenterol Hepatol, 2015 Jul;12(7):387-400) and inflammatory bowel diseases (eg. Crohn’s disease, ulcerative colitis) (Zhen Y and Zhang H. Front Immunol, 2019 Feb 28;10:276). Also, inflammatory joint disorders (eg. gout, pseudogout (chondrocalcinosis), arthropathy, osteoarthritis, and rheumatoid arthritis (Vande Walle L et al., Nature, 2014 Aug 7;512(7512): 69-73) were linked to NLRP3. Additionally, kidney related diseases (hyperoxaluria (Knauf et al., Kidney Int, 2013 Nov;84(5):895-901), lupus nephritis, hypertensive nephropathy (Krishnan et al., Br J Pharmacol, 2016 Feb;173(4):752-65), hemodialysis related inflammation and diabetic nephropathy which is a kidney -related complication of diabetes (Type 1, Type 2 and mellitus diabetes), also called diabetic kidney disease (Shahzad et al., Kidney Int, 2015 Jan;87(l):74-84) are associated to NLRP3 inflammasome activation. Reports link onset and progression of neuroinflammation-related disorders (eg. brain infection, acute injury, multiple sclerosis, Alzheimer's disease) and neurodegenerative diseases (Parkinsons disease) to NLRP3 inflammasome activation (Sarkar et A.. NPJ Parkinsons Dis. 2017 Oct 17;3:30). In addition, cardiovascular or metabolic disorders (eg. cardiovascular risk reduction (CvRR), atherosclerosis, type I and type II diabetes and related complications (e.g. nephropathy, retinopathy), peripheral artery disease (PAD), acute heart failure and hypertension (Ridker et al., CANTOS Trial Group. N Engl JMed, 2017 Sep 21;377(12): 1119-1131 ; andToldo S and Abbate A. Nat Rev Cardiol, 2018 Apr;15(4):203-214) have recently been associated to NLRP3. Also, skin associated diseases were described (eg. wound healing and scar formation; inflammatory skin diseases, eg. acne, hi dradenitis suppurativa (Kelly et al., Br J Dermatol, 2015 Dec;173(6)). In addition, respiratory conditions have been associated with NLRP3 inflammasome activity (eg. asthma, sarcoidosis, Severe Acute Respiratory Syndrome (SARS) (Nieto-Torres et al., Virology, 2015 Nov;485:330-9)) but also age-related macular degeneration (Doyle et al., Nat Med, 2012 May;18(5):791-8). Several cancer related diseases/disorders were described linked to NLRP3 (eg. myeloproliferative neoplasms, leukemias, myelodysplastic syndromes (MOS), myelofibrosis, lung cancer, colon cancer (Ridker et al., Lancet, 2017 Oct 21;390(10105):1833-1842; Derangere et al., Cell Death Differ. 2014 Dec;21(12):1914-24; Basiorka et al., Lancet Haematol, 2018 Sep;5(9): e393- e402, Zhang et al., Hum Immunol, 2018 Jan;79(1):57-62). Several patent applications describe NLRP3 inhibitors, with recent ones including for instance international patent application WO 2020/018975, WO 2020/037116, WO 2020/021447, WO 2020/010143, WO 2019/079119, WO 2019/0166621 and WO 2019/121691, which disclose a range of specific compounds. International patent application WO 2019/007696 discloses various compounds for the treatment of fibrosis There is a need for inhibitors of the NLRP3 inflammasome pathway to provide new and/or alternative treatments for the diseases/disorders mentioned herein. SUMMARY OF THE INVENTION The invention provides compounds which inhibit the NLRP3 inflammasome pathway. Thus, in an aspect of the invention, there is now provided a compound of formula (I), or a R1 represents: (i) C 3-6 cycloalkyl optionally substituted with one or more substituents independently selected from -OH, -C 1-3 alkyl and hydroxyC1-3alkyl; (ii) aryl or heteroaryl, each of which is optionally substituted with 1 to 3 substituents independently selected from halo, =O, -OH, -O-C 1-3 alkyl, -C 1-3 alkyl, haloC1-3alkyl, hydroxyC 1-3 alkyl, C 1-3 alkoxy, haloC1-3alkoxy; or (iii) heterocyclyl, optionally substituted with 1 to 3 substituents independently selected from =O, C 1-3 alkyl and C 3-6 cycloalkyl; R2 represents: (i) C 1-3 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC 1-3 alkyl; (ii) C 3-6 cycloalkyl; or

(iii) C2-4 alkenyl optionally substituted with -OC 1-3 alkyl;

R 3 represents:

(i) halo;

(ii) C 1-4 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC 1-3 alkyl;

(iii) C 2-4 alkenyl optionally substituted with -OC 1-3 alkyl;

(iv) C 3-6 cycloalkyl; or

(v) -OC 1-3 alkyl, which compounds may be referred to herein as “compounds of the invention”.

In an embodiment, compounds of the invention that may be mentioned include a compound of formula (I) as defined herein, or a pharmaceutically acceptable salt thereof, but in which: R 1 represents:

(i) C 3-6 cycloalkyl optionally substituted with one or more substituents independently selected from -OH, -C 1-3 alkyl and hydroxyC 1-3 alkyl;

(ii) aryl or heteroaryl, each of which is optionally substituted with 1 to 3 substituents independently selected from halo, -CN, =O, -OH, -O-C 1-3 alkyl, -C 1-3 alkyl, C 3-6 cycloalkyl, haloCmalkyl, hydroxyC 1-3 alkyl, C 1-3 alkoxy, haloC 1-3 alkoxy and -S(O) 2 C 1-4 alkyl; or

(iii) heterocyclyl, optionally substituted with 1 to 3 substituents independently selected from =O, C 1-3 alkyl and C 3-6 cycloalkyl.

In another aspect, there is provided compounds of the inventon for use as a medicament. In another aspect, there is provided a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention.

In a further aspect, there is provided compounds of the invention (and/or pharmaceutical compositions comprising such compounds) for use: in the treatment of a disease or disorder associated with NLRP3 activity (including inflammasome activity); in the treatment of a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder; in inhibiting NLRP3 inflammasome activity (including in a subject in need thereol); and/or as an NLRP3 inhibitor. Specific diseases or disorders may be mentioned herein, and may for instance be selected from inflammasome-related diseases or disorders, immune diseases, inflammatory diseases, auto-immune diseases, or auto-inflmmatory diseases. In another aspect, there is provided a use of compounds of the invention (and/or pharmaceutical compositions comprising such compounds): in the treatment of a disease or disorder associated with NLRP3 activity (including inflammasome activity); in the treatment of a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder; in inhibiting NLRP3 inflammasome activity (including in a subject in need thereof); and/or as an NLRP3 inhibitor.

In another aspect, there is provided use of compounds of the invention (and/or pharmaceutical compositions comprising such compounds) in the manufacture of a medicament for: the treatment of a disease or disorder associated with NLRP3 activity (including inflammasome activity); the treatment of a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder; and/or inhibiting NLRP3 inflammasome activity (including in a subject in need thereof).

In another aspect, there is provided a method of treating a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder, comprising administering a therapeutically effective amount of a compound of the invention, for instance to a subject (in need thereof). In a further aspect there is provided a method of inhibiting the NLRP3 inflammasome activity in a subject (in need thereof), the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound of the invention.

In further aspect, there is a provided a compound of the invention in combination (including a pharmaceutical combination) with one or more therapeutic agents (for instance as described herein). Such combination may also be provided for use as described herein in respect of compounds of the invention, or, a use of such combination as described herein in respect of compounds of the invention. There may also be provided methods as described herein in repsect of compounds of the invention, but wherein the method comprises administering a therapeutically effective amount of such combination.

DETAILED DESCRIPTION OF THE INVENTION

The invention provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, wherein: R1 represents: (i) C 3-6 cycloalkyl optionally substituted with one or more substituents independently selected from -OH, -C 1-3 alkyl and hydroxyC 1-3 alkyl; (ii) aryl or heteroaryl, each of which is optionally substituted with 1 to 3 substituents independently selected from halo, =O, -OH, -O-C 1-3 alkyl, -C 1-3 alkyl, haloC 1-3 alkyl, hydroxyC 1-3 alkyl, C 1-3 alkoxy, haloC 1-3 alkoxy; or (iii) heterocyclyl, optionally substituted with 1 to 3 substituents independently selected from =O, C 1-3 alkyl and C 3-6 cycloalkyl; R2 represents: (i) C 1-3 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC 1-3 alkyl; (ii) C 3-6 cycloalkyl; or (iii) C 2-4 alkenyl optionally substituted with -OC1-3 alkyl; R3 represents: (i) halo; (ii) C 1-4 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC 1-3 alkyl; (iii) C 2-4 alkenyl optionally substituted with -OC 1-3 alkyl; (iv) C 3-6 cycloalkyl; or (v) -OC 1-3 alkyl. As indicated above, such compounds may be referred to herein as “compounds of the invention”. Pharmaceutically-acceptable salts include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound of the invention with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the invention in the form of a salt with another counter- ion, for example using a suitable ion exchange resin. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.

Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.

Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.

Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.

Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, di ethylamine, lysine, meglumine, piperazine, and tromethamine

For the purposes of this invention solvates, prodrugs, N-oxides and stereoisomers of compounds of the invention are also included within the scope of the invention.

The term “prodrug” of a relevant compound of the invention includes any compound that, following oral or parenteral administration, is metabolised in vivo to form that compound in an experimentally-detectable amount, and within a predetermined time (e.g. within a dosing interval of between 6 and 24 hours (i.e. once to four times daily)). For the avoidance of doubt, the term “parenteral” administration includes all forms of administration other than oral administration.

Prodrugs of compounds of the invention may be prepared by modifying functional groups present on the compound in such a way that the modifications are cleaved, in vivo when such prodrug is administered to a mammalian subject. The modifications typically are achieved by synthesising the parent compound with a prodrug substituent. Prodrugs include compounds of the invention wherein a hydroxyl, amino, sulfhydryl, carboxy or carbonyl group in a compound of the invention is bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl, amino, sulfhydryl, carboxy or carbonyl group, respectively.

Examples of prodrugs include, but are not limited to, esters and carbamates of hydroxy functional groups, esters groups of carboxyl functional groups, N-acyl derivatives and N-Mannich bases. General information on prodrugs may be found e.g. in Bundegaard, H. “Design of Prodrugs” p. 1-92, Elesevier, New York-Oxford (1985).

Compounds of the invention may contain double bonds and may thus exist as E (entgegeri) and Z (zusammeri) geometric isomers about each individual double bond. Positional isomers may also be embraced by the compounds of the invention. All such isomers (e.g. if a compound of the invention incorporates a double bond or a fused ring, the cis- and trans- forms, are embraced) and mixtures thereof are included within the scope of the invention (e.g. single positional isomers and mixtures of positional isomers may be included within the scope of the invention).

Compounds of the invention may also exhibit tautomerism. All tautomeric forms (or tautomers) and mixtures thereof are included within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomerisations. Valence tautomers include interconversions by reorganisation of some of the bonding electrons.

Compounds of the invention may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism. Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation. The various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques. Alternatively the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation (i.e. a ‘chiral pool’ method), by reaction of the appropriate starting material with a ‘chiral auxiliary’ which can subsequently be removed at a suitable stage, by derivatisation (i.e. a resolution, including a dynamic resolution), for example with a homochiral acid followed by separation of the diastereomeric derivatives by conventional means such as chromatography, or by reaction with an appropriate chiral reagent or chiral catalyst all under conditions known to the skilled person.

All stereoisomers (including but not limited to diastereoisomers, enantiomers and atropisomers) and mixtures thereof (e.g. racemic mixtures) are included within the scope of the invention.

In the structures shown herein, where the stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined. When an absolute configuration is specified, it is according to the Cahn-Ingold- Prelog system. The configuration at an asymmetric atom is specified by either R or S. Resolved compounds whose absolute configuration is not known can be designated by (+) or (-) depending on the direction in which they rotate plane polarized light.

When a specific stereoisomer is identified, this means that said stereoisomer is substantially free, i.e. associated with less than 50%, preferably less than 20%, more preferably less than 10%, even more preferably less than 5%, in particular less than 2% and most preferably less than 1%, of the other isomers. Thus, when a compound of formula (I) is for instance specified as (R), this means that the compound is substantially free of the (S) isomer.

The compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms.

The present invention also embraces isotopically-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature (or the most abundant one found in nature). All isotopes of any particular atom or element as specified herein are contemplated within the scope of the compounds of the invention. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and iodine, such as 2 H, 3 H, 1 'C. 13 C, 14 C , 13 N, 15 0, 17 0, 18 O, 32 P, 33 P, 35 S, 18 F, 36 C1, 123 I, and 125 I. Certain isotopically- labeled compounds of the present invention (e.g., those labeled with 3 H and 14 C) are useful in compound and for substrate tissue distribution assays. Tritiated ( 3 H) and carbon-14 ( 14 C) isotopes are 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) and hence may be preferred in some circumstances. Positron emitting isotopes such as 15 O, 13 N, 1 'C and 18 F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the description/Examples hereinbelow, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.

Unless otherwise specified, Ci-q alkyl groups (where q is the upper limit of the range) defined herein may be straight-chain or, when there is a sufficient number (i.e. a minimum of two or three, as appropriate) of carbon atoms, be branched-chain. Such a group is attached to the rest of the molecule by a single bond. C 2-q alkenyl when used herein (again where q is the upper limit of the range) refers to an alkyl group that contains unsaturation, i.e. at least one double bond. C 3-q cycloalkyl (where q is the upper limit of the range) refers to an alkyl group that is cyclic, for instance cycloalkyl groups may be monocyclic or, if there are sufficient atoms, bicyclic. In an embodiment, such cycloalkyl groups are monocyclic. Such cycloalkyl groups are unsaturated. Substituents may be attached at any point on the cycloalkyl group. The term “halo”, when used herein, preferably includes fluoro, chloro, bromo and iodo. C 1-q alkoxy groups (where q is the upper limit of the range) refers to the radical of formula -ORa, where Ra is a C 1-q alkyl group as defined herein. HaloC 1-q alkyl (where q is the upper limit of the range) goups refer to C 1-q alkyl groups, as defined herein, where such group is substituted by one or more halo. HydroxyC 1-q alkyl (where q is the upper limit of the range) refers to C 1-q alkyl groups, as defined herein, where such group is substituted by one or more (e.g. one) hydroxy (- OH) groups (or one or more, e.g. one, of the hydrogen atoms is replaced with -OH). Similarly, haloC 1-q alkoxy and hydroxyC 1-q alkoxy represent corresponding -OC 1-q alkyl groups that are substituted by one or more halo, or, substituted by one or more (e.g. one) hydroxy, respectively. Heterocyclyl groups that may be mentioned include non-aromatic monocyclic and bicyclic heterocyclyl groups in which at least one (e.g. one to four) of the atoms in the ring system is other than carbon (i.e. a heteroatom), and in which the total number of atoms in the ring system is between 3 and 20 (e.g. between three and ten, e.g between 3 and 8, such as 5- to 8-). Such heterocyclyl groups may also be bridged. Such heterocyclyl groups are saturated. C 2-q heterocyclyl groups that may be mentioned include 7- azabicyclo[2.2.1]heptanyl, 6-azabicyclo[3.1.1]heptanyl, 6-azabicyclo[3.2.1]-octanyl, 8- azabicyclo-[3.2.1]octanyl, aziridinyl, azetidinyl, dihydropyranyl, dihydropyridyl, dihydropyrrolyl (including 2,5-dihydropyrrolyl), dioxolanyl (including 1,3-dioxolanyl), dioxanyl (including 1,3-dioxanyl and 1,4-dioxanyl), dithianyl (including 1,4-dithianyl), dithiolanyl (including 1,3-dithiolanyl), imidazolidinyl, imidazolinyl, morpholinyl, 7- oxabicyclo[2.2.1]heptanyl, 6-oxabicyclo-[3.2.1]octanyl, oxetanyl, oxiranyl, piperazinyl, piperidinyl, non-aromatic pyranyl, pyrazolidinyl, pyrrolidinonyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, sulfolanyl, 3-sulfolenyl, tetrahydropyranyl, tetrahydrofuranyl, tetrahydropyridyl (such as 1,2,3,4-tetrahydropyridyl and 1,2,3,6-tetrahydropyridyl), thietanyl, thiiranyl, thiolanyl, thiomorpholinyl, trithianyl (including 1,3,5-trithianyl), tropanyl and the like. Substituents on heterocyclyl groups may, where appropriate, be located on any atom in the ring system including a heteroatom. The point of attachment of heterocyclyl groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system. Heterocyclyl groups may also be in the N- or 5- oxidised form. In an embodiment, heterocyclyl groups mentioned herein are monocyclic.

Aryl groups that may be mentioned include Ce-20, such as Ce-12 (e.g. Ce-io) aryl groups. Such groups may be monocyclic, bicyclic or tricyclic and have between 6 and 12 (e.g. 6 and 10) ring carbon atoms, in which at least one ring is aromatic. Ce-io aryl groups include phenyl, naphthyl and the like, such as 1,2,3,4-tetrahydronaphthyl. The point of attachment of aryl groups may be via any atom of the ring system. For example, when the aryl group is polycyclic the point of attachment may be via atom including an atom of a non-aromatic ring. However, when aryl groups are polycyclic (e.g. bicyclic or tricyclic), they are preferably linked to the rest of the molecule via an aromatic ring. When aryl groups are polycyclic, in an embodiment, each ring is aromatic. In an embodiment, aryl groups mentioned herein are monocyclic or bicyclic. In a further embodiment, aryl groups mentioned herein are monocyclic.

“Heteroaryl” when used herein refers to an aromatic group containing one or more heteroatom(s) (e.g. one to four heteroatoms) preferably selected firomN, O and S. Heteroaryl groups include those which have between 5 and 20 members (e.g. between 5 and 10) and may be monocyclic, bicyclic or tricyclic, provided that at least one of the rings is aromatic (so forming, for example, a mono-, bi-, or tricyclic heteroaromatic group). When the heteroaryl group is polycyclic the point of attachment may be via any atom including an atom of a non-aromatic ring. However, when heteroaryl groups are polycyclic (e.g. bicyclic or tricyclic), they are preferably linked to the rest of the molecule via an aromatic ring. In an embodiment, when heteroaryl groups are polycyclic, then each ring is aromatic. Heteroaryl groups that may be mentioned include 3.4-dihydro- 17/- isoquinolinyl, 1,3-dihydroisoindolyl, 1,3-dihydroisoindolyl (e.g. 3,4-dihydro-177- isoquinolin-2-yl, l,3-dihydroisoindol-2-yl, l,3-dihydroisoindol-2-yl; i.e. heteroaryl groups that are linked via a non-aromatic ring), or, preferably, acridinyl, benzimidazolyl, benzodi oxanyl, benzodi oxepinyl, benzodi oxolyl (including 1,3-benzodioxolyl), benzofuranyl, benzofurazanyl, benzothiadiazolyl (including 2,1,3-benzothiadiazolyl), benzothiazolyl, benzoxadiazolyl (including 2,1,3-benzoxadiazolyl), benzoxazinyl (including 3.4-dihydro-27/-l .4-benzoxazinyl). benzoxazolyl, benzomorpholinyl, benzoselenadiazolyl (including 2,1,3-benzoselenadiazolyl), benzothienyl, carbazolyl, chromanyl, cinnolinyl, furanyl, imidazolyl, imidazo[l,2-a]pyridyl, indazolyl, indolinyl, indolyl, isobenzofuranyl, isochromanyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiaziolyl, isothiochromanyl, isoxazolyl, naphthyridinyl (including 1,6-naphthyridinyl or, preferably, 1,5-naphthyridinyl and 1,8-naphthyridinyl), oxadiazolyl (including 1,2,3- oxadiazolyl, 1,2,4-oxadiazolyl and 1,3,4-oxadiazolyl), oxazolyl, phenazinyl, phenothiazinyl, phthalazinyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinolizinyl, quinoxalinyl, tetrahydroisoquinolinyl (including 1,2,3,4-tetrahydroisoquinolinyl and 5,6,7,8-tetrahydroisoquinolinyl), tetrahydroquinolinyl (including 1, 2,3,4- tetrahydroquinolinyl and 5,6,7,8-tetrahydroquinolinyl), tetrazolyl, thiadiazolyl (including 1,2,3-thiadiazolyl, 1 ,2,4-thiadiazolyl and 1,3,4-thiadiazolyl), thiazolyl, thiochromanyl, thiophenetyl, thienyl, triazolyl (including 1,2,3-triazolyl, 1 ,2,4-triazolyl and 1,3,4-triazolyl) and the like. Substituents on heteroaryl groups may, where appropriate, be located on any atom in the ring system including a heteroatom. The point of attachment of heteroaryl groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system. Heteroaryl groups may also be in the N- or 5- oxidised form. When heteroaryl groups are polycyclic in which there is a non-aromatic ring present, then that non-aromatic ring may be substituted by one or more =0 group. In an embodiment, heteroaryl groups mentioned herein may be monocyclic or bicyclic. In a further embodiment, heteroaryl groups mentioned herein are monocyclic.

Heteroatoms that may be mentioned include phosphorus, silicon, boron and, preferably, oxygen, nitrogen and sulfur.

For the avoidance of doubt, where it is stated herein that a group may be substituted by one or more substituents (e.g. selected from Ci-6 alkyl), then those substituents (e.g. alkyl groups) are independent of one another. That is, such groups may be substituted with the same substituent (e.g. same alkyl substituent) or different (e.g. alkyl) substituents.

All individual features (e.g. preferred features) mentioned herein may be taken in isolation or in combination with any other feature (including preferred feature) mentioned herein (hence, preferred features may be taken in conjunction with other preferred features, or independently of them).

The skilled person will appreciate that compounds of the invention that are the subject of this invention include those that are stable. That is, compounds of the invention include those that are sufficiently robust to survive isolation from e.g. a reaction mixture to a useful degree of purity.

Various embodiments of the invention will now be described, including embodiments of the compounds of the invention.

In an embodiment, compounds of the invention include those in which R 1 represents: (i) C 3-6 cycloalkyl; (ii) aryl or heteroaryl; or (iii) or heterocyclyl, all of which are optionally substituted as herein defined. In a particular embodiment, R 1 represents: (i) C 3-6 cycloalkyl; or (ii) aryl or heteroaryl, all of which are optionally substituted as herein defined.

In an embodiment when R 1 represents optionally substituted C 3-6 cycloalkyl, then it represents C 3-6 cycloalkyl (or, in an embodiment, C3-4 cycloalkyl) optionally substituted by one or two substituents selected from C 1-3 alkyl (e.g. methyl), -OH and hydroxyC 1-3 alkyl (e.g. -C(CH 3 ) 2 OH). In a further embodiment, R1 represents cyclopropyl (e.g. unsubstituted) or cyclobutyl. In a further embodiment, R1 represents cyclohexyl. In yet a further embodiment, R1 represents unsubstituted cyclopropyl or cyclobutyl substituted by - OH and methyl (e.g. at the same carbon atom). In yet a further embodiment, R1 represents cyclohexyl, for instance substituted by -OH (e.g. by one -OH group). In an embodiment therefore, R1 represents: (e.g. methyl) and hydroxyC 1-3 alkyl (e.g.2-propyl substituted by -OH, so forming e.g. a 2- hydroxy-2-propyl group). In a particular embodiment of this aspect, R1 represents C 3-6 cyclolkyl, such as optionally substituted cyclohexyl, optionally substituted cyclobutyl or unsubstituted (or optionally substituted) cyclopropyl, for instance: cted from those defined by R1a, and in an embodiment, represents one optional substituent selected from -OH; R1a, and in an embodiment R1aa represents two substituents, methyl and -OH and in another embodiment R1aa represents one substituent -C(CH 3 ) 2 (OH); or articular embodiment, it is not present. In an embodiment where R1 represents aryl or heteroaryl, optionally substituted as defined herein, then it may represent: (i) phenyl; (ii) a 5- or 6-membered mono-cyclic heteroaryl group; or (iii) a 9- or 10-membered bicyclic heteroaryl group, all of which are optionally substituted by one to three substituents as defined herein. In an embodiment, the aforementioned aryl and heteroaryl groups are optionally substituted with one or two (e.g. one) substituent(s) selected from halo (e.g. fluoro, iodo), =O, -OH, C1-3 alkyl (e.g. methyl), -OC1-3 alkyl and -haloC1-3alkyl (e.g. -CF3). In one embodiment, R1 represents phenyl or a mono-cyclic 6-membered heteroaryl group and in another embodiment it may represent a 9- or 10-membered (e.g.9-membered) bicyclic heteroaryl group. Hence, in an embodiment, R1 may represent: from halo (e.g. fluoro, iodo), =O, -OH, C1-3 alkyl (e.g. methyl), haloC1-3alkyl (e.g. -CF3), and at least one of Rb, Rc, Rd, Re and Rf represents a nitrogen heteroatom (and the others represent CH). In an embodiment, either one or two of Rb, Rc, Rd, Re and Rf represent(s) a nitrogen heteroatom, for instance, Rd represents nitrogen and, optionally, Rb represents nitogen, or, Rc represents nitogen. In an aspect: (i) Rb and Rd represent nitrogen; (ii) Rd represents nitrogen; (iii) Rc represents nitrogen; or (iv) Rb and Rc represent nitrogen. Hence, R1 may represent pyridyl (e.g.3-pyridyl or 4-pyridyl), pyrimidinyl (e.g.4-pyrimidinyl) or pyridazinyl (e.g.3- pyridazinyl or 6-pyridazinyl), all of which are optionally substituted as herein defined; hence, in an embodiment such groups may be substituted by halo (e.g. fluoro, iodo), =O, - OH, C 1-3 alkyl (e.g. methyl), haloC 1-3 alkyl (e.g. -CF 3 ) or such group may be unsubstituted. In another embodiment, R1 may represent: O O wherein R1b is as defined above (i.e. represents one or two optional substituents as defined above, for example selected from halo, =O, C 1-3 alkyl (e.g. methyl) and haloC 1-3 alkyl (e.g. -CF 3 )), at least one of the rings of the bicyclic system is aromatic (as depicted), Rk represents a N or C atom, Rg represents a N or C atom and any one or two of R h , R i and Rj (for instance, one or two of Ri and Rj) represents N and the other(s) represent(s) C (provided that, as the skilled person would understand, the rules of valency are adhered to; for instance when one of the atoms of the (hetero)aromatic ring represents C, then it is understood that it may bear a H atom). Hence, for instance, R1 may represent: in which: R b and R d represent a nitrogen atom; R c represents a nitogen atom; or R b and R c represent a nitrogen atom (and the other R b -R f moieties, e.g. R e and R f , represent a carbon atom), and R 1b represents one or more optional substituents as defined herein. Given that R1b may represent a =O substituent, then the following groups are also included:

In another embodiment, R 1 represents: in which one of Ri and Rj represents N and the other represents C, or, both Ri and Rj represent N, and, in an embodiment, there is one or two independent R lb substituents present or, in another embodiment, no R lb substituent present. Given that R lb may represent =0, R 1 may also represent:

In an embodiment of the invention, R 1 may represents phenyl or a 6-membered heteroaryl group (containing between one and three heteroatoms) and which is optionally substituted as defined herein. In an embodiment, R 1 may represent a 6,5-fused bicyclic ring containing one to five heteroatoms (wherein at least two are nitrogen) and which group is optionally substituted as herein defined, for instance by one or two substituents as defined above (and which may be selected from halo, C1-3 alkyl, C3-4 cycloalkyl, haloCi-salkyl, -CN, =0).

In a particular embodiment, R 1 represents: in which R lb is preferably not present, i.e. the bicycle is unsubstituted.

In an embodiment where R 1 represents heterocyclyl, optionally substituted as defined herein, such goup is in a further aspect a 5- or 6-membered heterocyclyl group, for instance containing at least one nitrogen or oxygen heteroatom; for instance, in a particular embodiment, in this instance R 1 may represent a 6-membered nitrogen-containing heterocyclyl group optionally substituted by one or two substituents as defined herein, e.g. by C 1-3 alkyl. In an aspect of this embodiment, the 6-membered heterocyclyl group may be piperidinyl (e.g. 3-piperidinyl) optionally substituted as defined herein.

In an embodiment R 2 represents: (i) C 1-3 alkyl optionally substituted with one or more substituents independently selected from halo (e.g. fluoro), -OH and -OC 1-2 alkyl; (ii) C 3-6 cycloalkyl; or (iii) C 2-4 alkenyl optionally substituted by -OC 1-2 alkyl. In a further embodiment, R 2 represents C 1-3 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC 1-2 alkyl. In yet a further embodiment, R 2 represents unsubstituted C 1-3 alkyl.

In a particular embodiment R 2 represents unsubstituted isopropyl or unsubstituted ethyl.

In another embodiment of the invention, R 2 represents -CH2CH3, isopropyl, -C(H)(CH 3 )CH 2 CH 3 or cyclopropyl. In a particular embodiment R 2 represents isopropyl.

In an embodiment, R 3 represents (i) halo (e.g. bromo); (ii) C 1-4 alkyl optionally substituted with one or more substituents independently selected from halo, -OH and -OC 1-2 alkyl; or (iii) C 3-6 cycloalkyl (e.g. cyclopropyl). In a further embodiment, R 3 represents: halo (e.g. bromo); C 1-3 alkyl optionally (and preferably) substituted by one or more fluoro atoms (so forming, e.g. -CF 3 ); or C 3-6 (e.g. C 3-4 ) cycloalkyl (e.g. cylopropyl).

In an embodiment when R 3 represents optionally substituted C 1-4 alkyl, then it represents C 1-3 alkyl optionally substituted by one or more fluoro atoms. In an embodiment when R 3 represents C 3-6 cycloalkyl, then it represents cyclopropyl. In an embodiment when R 3 represents -OC 1-3 alkyl, then it represents -OC 1-2 alkyl (e.g. -OCH 3 ).

In a particular embodiment, R 3 represents halo (e.g. bromo), methyl, ethyl, isopropyl -CF 3 , -CHF 2 or cyclopropyl. For instance, R 3 represents bromo, -CF 3 or cyclopropyl.

In another embodiment of the invention, R 3 represents halo (e.g. bromo or chloro), C 1-4 alkyl (e.g. C 1-3 alkyl) optionally substituted by one or more fluoro atoms (so forming, e.g. -CF 3 , isopropyl, -CF 2 CH 3 , -CHF 2 , C(CH 3 )2F) or C 3-6 cycloalkyl (e.g. cyclopropyl or cyclobutyl). In a particular embodiment, R 3 represents -CF 3 or CF 2 CH 3 .

The names of the compounds of the present invention were generated according to the nomenclature rules agreed upon by the Chemical Abstracts Service (CAS) using Advanced Chemical Development, Inc., software (ACD/Name product version 10.01; Build 15494, 1 Dec 2006) or according to the nomenclature rules agreed upon by the International Union of Pure and Applied Chemistry (IUPAC) using Advanced Chemical Development, Inc., software (ACD/Name product version 10.01.0.14105, October 2006). In case of tautomeric forms, the name of the depicted tautomeric form of the structure was generated. The other non-depicted tautomeric form is also included within the scope of the present invention. Preparation of the compounds In an aspect of the invention, there is provided a process for the preparation of compounds of the invention, where reference here is made to compounds of formula (I) as defined herein. Compounds of formula (I) may be prepared by: (i) reaction of a compound of formula (II), C1-3 alkyl ester), wherein R 2 and R 3 are as hereinbefore defined, with a compound of formula (III), H 2 N-R 1 (III) or a derivative thereof, wherein R1 is as hereinbefore defined, under amide-forming reaction conditions (also referred to as amidation), for example in the presence of a suitable coupling reagent (e.g. propylphosphonic anhydride, 1- [bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridi nium 3-oxide hexafluorophosphate (O-(7-azabenzotriazol-1-yl)-N,N,N’,N’-tetramethyluronium hexafluorophosphate), 1,1’-carbonyldiimidazole, N,N’-dicyclohexylcarbodiimide, 1-(3- dimethylaminopropyl)-3-ethylcarbodiimide (or hydrochloride thereof), N,N’- disuccinimidyl carbonate, benzotriazol-1-yloxytris(dimethylamino)phosphonium hexafluoro-phosphate, 2-(1H-benzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexa- fluorophosphate (i.e. O-(1H-benzotriazol-1-yl)-N,N,N´,N´-tetramethyluronium hexafluorophosphate), benzotriazol-1-yloxytris-pyrrolidinophosphonium hexa- fluorophosphate, bromo-tris-pyrrolidinophosponium hexafluorophosphate, 2-(1H- benzotriazol-1-yl)-1,1,3,3-tetramethyluronium tetra-fluorocarbonate, 1- cyclohexylcarbodiimide-3-propyloxymethyl polystyrene, O-benzotriazol-1-yl-N,N,N’,N’- tetramethyluronium tetrafluoroborate), optionally in the presence of a suitable base (e.g. sodium hydride, sodium bicarbonate, potassium carbonate, pyridine, triethylamine, dimethylaminopyridine, diisopropylamine, sodium hydroxide, potassium /c/7-butoxide and/or lithium diisopropylamide (or variants thereof) and an appropriate solvent (e.g. tetrahydrofuran, pyridine, toluene, dichloromethane, chloroform, acetonitrile, dimethylformamide, trifluoromethylbenzene, dioxane or triethylamine). Such reactions may be performed in the presence of a further additive such as 1 -hydroxybenzotriazole hydrate. Alternatively, a carboxylic acid group may be converted under standard conditions to the corresponding acyl chloride (e.g. in the presence of SOCh or oxalyl chloride), which acyl chloride is then reacted with a compound of formula (II), for example under similar conditions to those mentioned above;

(ii) reaction of a compound of formula (IV), or a derivative thereof (e.g. a salt), wherein R 1 and R 3 are as hereinbefore defined, with a compound of formula (V),

R 2 -LG a (V) wherein LG a represents a suitable leaving group (e.g. halo, such as iodo, chloro) and R 2 is as defined herein, under suitable reaction conditions, e.g. in the presence of an appropriate base, e.g. CS 2 CO 3 , K 2 CO 3 or LiHMDS, or the like, or alternative alkylation reaction conditions (although typically such a reaction is performed on derivatives of compounds of formula (IV) or derivatives of compounds of formula (II) (or other intermediates described herein) where the carboxlic acid group may be protected);

(iii) by transformation (such transformation steps may also take place on intermediates) of a certain compound of formula (I) into another, for example: for compounds of formula (I) containing an alkene, reduction to a corresponding compound of formula (I) containing an alkane, under reduction conditions, e.g. with hydrogen in the presence of a suitable catalyst such as, for example, palladium on carbon, in a suitable reaction-inert solvent, such as, for example, ethanol or methanol; - coupling to convert a halo or triflate group to e.g. an alkyl, alkenyl or cycloalkyl group, for example in the presence of a suitable coupling reagent, e.g. where the reagent comprises the appropriate alkyl, alkenyl or aryl/heteroaryl group attached to a suitable group such as -B(OH) 2 , - B(ORwx) 2 , zincates (e.g. including -Zn(Rwx) 2 , -ZnBrRwx) or -Sn(Rwx) 3 , in which each Rwx independently represents a C alkyl group, or, in the case o wx wx 1-6 f -B(OR )2, the respective R groups may be linked together to form a 4- to 6-membered cyclic group (such as a 4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl group), thereby forming e.g. a pinacolato boronate ester group. The reaction may be performed in the presence of a suitable catalyst system, e.g. a metal (or a salt or complex thereof) such as Pd, CuI, Pd/C, PdCl 2 , Pd(OAc) 2 , Pd(Ph 3 P) 2 Cl 2 , Pd(Ph 3 P) 4 (i.e. palladium tetrakistriphenylphosphine), Pd 2 (dba) 3 and/or NiCl 2 (preferred cataysts include RuPhos Pd G3, XPhos Pd and bis(tri-tert-butylphosphine)palladium(0)) and optionally a ligand such as PdCl 2 (dppf).DCM, t-Bu 3 P, (C 6 H 11 ) 3 P, Ph 3 P, AsPh 3 , P(o- Tol) 3 , 1,2-bis(diphenylphosphino)ethane, 2,2'-bis(di-tert-butyl- phosphino)-1,1'-biphenyl, 2,2'-bis(diphenylphosphino)-1,1'-bi-naphthyl, 1,1’-bis(diphenyl-phosphino-ferrocene), 1,3- bis(diphenylphosphino)propane, xantphos, or a mixture thereof, together with a suitable base, such as Na2CO3, K3PO4, Cs2CO3, NaOH, KOH, K2CO3, CsF, Et3N, (i-Pr)2NEt, t-BuONa or t-BuOK (or mixtures thereof; preferred bases include Na2CO3 and K2CO3) in a suitable solvent such as dioxane, toluene, ethanol, dimethylformamide, dimethoxyethane, ethylene glycol dimethyl ether, water, dimethylsulfoxide, acetonitrile, dimethylacetamide, N- methylpyrrolidinone, tetrahydrofuran or mixtures thereof (preferred solvents include dimethylformamide and dimethoxyethane) – as an example compounds in which R3 represent halo may be converted into corresponding compounds in which R3 represents alkyl, alkenyl or cycloalkyl as hereinbefore defined; - reduction of a ketone to an alcohol, in the presence of suitable reducing conditions, e.g. NaBH4 or the like; - conversion of a -C(O)alkyl moiety to a -C(OH)(alkyl)(alkyl) moiety by reaction of an appropriate Grignard reagent, e.g. alkylMgBr; - transformation of a alkene =CH2 moiety to a carbonyl =O moiety, for instance, in the presence of AD-mix-Alpha and methane-sulfonamide, for instance a -CH=CH2 moiety may be converted to a -C(O)H moiety (e.g. by reaction with osmium tetraoxide), which in turn may be converted to a -CHF2 group by reaction with DAST; - transformation of a ketone to an alcohol -OH moiety; - alkylation of a -OH moiety (to -O-alkyl), under appropriate reaction conditions. The compound of formula (II) may be prepared by oxidation of a corresponding compound of formula (VI), I) nbefore defined, under appropriate oxidation reaction conditions such as herein described, e.g. by use of Jones reagent. Alternatively, compounds of formula (II) may be prepared by oxidation of a corresponding compound of formula (VII), II) nbefore defined, under appropriate oxidation reaction conditions such as herein described, e.g. by reaction in a mixture of monosodium phosphate, 2-methyl-2-butene and sodium chlorite in an appropriate solvent system. Compounds of formula (VII) may be prepared by oxidation of compounds of formula (VI) under oxidation conditions such as reaction with Dess-Martin periodinane. Compounds of formula (VI) may need to be protected (and subsequently deprotected at appropriate points in the synthesis). For instance, compounds of formula (VI) that are protected with a silyl protecting group (e.g. tri-isopropyl-silyl) may be deprotected, for instance by reaction with TBAF or the like.

Compounds of formula (VI) that are protected (for instance by a silyl protecting group, such as a tri-isopropyl-silyl group) may be prepared by reaction of a corresponding compound of formula (VIII) or a derivative thereof, wherein PG represents a suitable protecting group (if needed, for instance a silyl protecting group such as tri-isopropyl-silyl) with a compound of formula (IX),

R 2 -LG (IX) wherein R 2 is as hereinbefore defined and LG represents a suitable leaving group, such as halo (e.g. iodo, chloro), for instance under reaction conditions and using reagents such as those described herein, for instance in the presence of an appropriate base such as K2CO3, NaH or the like.

Compounds of formula (VIII) may be prepared by proection of compounds of formula (X), wherein R 3 is as hereinbefore defined, for instance under standard protection conditions, e.g. in the case ofa silyl protecting group, reaction in the presence of the appropriate sily chloride (e.g. triisopropylsilyl chloride).

Compounds of formula (X) may be preprared by cyclisation of a corresponding compound of formula (XI), I) or a er va ve ereo , w ere n s as ere n e ore defined, for instance in the presence of acid (e.g. H 2 SO 4 ) in deionized water and then addition of sodium nitrite (in deionized water). Compound of formula (XI) may be prepared by reaction of a corresponding compound of formula (XII), II) or a er va ve ereo , w ere n s as ere n e ore defined, and LG1 represents a suitable leaving group such as halo (e.g. iodo), with a compound of formula (XIII), H-C ≡C-CH 2 CH 2 -OH (XIII) wherein said reaction may be performed in the presence of a suitable coupling reagent and reaction conditions such as those described in respect of preparation of compounds of formula (I) (process step (iii) where conversion of a halo or triflate group to an alkyl, alkenyl or cycloalkyl group) for instance in the presence of e.g. a metal (or a salt or complex thereof) such as Pd, CuI, Pd/C, PdCl 2 , Pd(OAc) 2 , Pd(Ph 3 P) 2 Cl 2 , Pd(Ph 3 P) 4 (i.e. palladium tetrakistriphenylphosphine), Pd 2 (dba) 3 and/or NiCl 2 (preferred cataysts include RuPhos Pd G3, XPhos Pd and bis(tri-tert-butylphosphine)palladium(0)) and optionally in the presence of a suitable base (e.g. Et 3 N). Certain intermediate compounds may be commercially available, may be known in the literature, or may be obtained either by analogy with the processes described herein, or by conventional synthetic procedures, in accordance with standard techniques, from available starting materials using appropriate reagents and reaction conditions. Certain substituents on/in final compounds of the invention or relevant intermediates may be modified one or more times, after or during the processes described above by way of methods that are well known to those skilled in the art. Examples of such methods include substitutions, reductions, oxidations, alkylations, acylations, hydrolyses, esterifications, etherifications, halogenations, nitrations or couplings.

Compounds of the invention may be isolated from their reaction mixtures using conventional techniques (e.g. recrystallisations, where possible under standard conditions).

It will be appreciated by those skilled in the art that, in the processes described above and hereinafter, the functional groups of intermediate compounds may need to be protected by protecting groups.

The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods (and the need can be readily determined by one skilled in the art). Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxy carbonyl (BOC), benzyloxycarbonyl (CBz), 9-fluorenylmethylene- oxy carbonyl (Fmoc) and 2,4,4-trimethylpentan-2-yl (which may be deprotected by reaction in the presence of an acid, e.g. HC1 in water/alcohol (e.g. MeOH)) or the like. The need for such protection is readily determined by one skilled in the art. For example the a -C(O)O- /c/7-butyl ester moiety may serve as a protecting group for a -C(O)OH moiety, and hence the former may be converted to the latter for instance by reaction in the presence of a mild acid (e.g. TFA, or the like).

The protection and deprotection of functional groups may take place before or after a reaction in the above-mentioned schemes.

Protecting groups may be removed in accordance with techniques that are well known to those skilled in the art and as described hereinafter. For example, protected compounds/intermediates described herein may be converted chemically to unprotected compounds using standard deprotection techniques.

The type of chemistry involved will dictate the need, and type, of protecting groups as well as the sequence for accomplishing the synthesis.

The use of protecting groups is fully described in “Protective Groups in Organic Synthesis”, 3 rd edition, T.W. Greene & P.G.M. Wutz, Wiley-Interscience (1999).

The compounds of the invention as prepared in the hereinabove described processes may be synthesized in the form of racemic mixtures of enantiomers which can be separated from one another following art-known resolution procedures. Those compounds of the invention that are obtained in racemic form may be converted into the corresponding diastereomeric salt forms by reaction with a suitable chiral acid. Said diastereomeric salt forms are subsequently separated, for example, by selective or fractional crystallization and the enantiomers are liberated therefrom by alkali. An alternative manner of separating the enantiomeric forms of the compounds of the invention involves liquid chromatography using a chiral stationary phase. Said pure stereochemically isomeric forms may also be derived from the corresponding pure stereochemically isomeric forms of the appropriate starting materials, provided that the reaction occurs stereospecifically. Preferably if a specific stereoisomer is desired, said compound will be synthesized by stereospecific methods of preparation. These methods will advantageously employ enantiomerically pure starting materials.

PHARMACOLOGY

There is evidence for a role of NLRP3-induced IL-1 and IL-18 in the inflammatory responses occurring in connection with, or as a result of, a multitude of different disorders (Menu etal., Clinical and Experimental Immunology, 2011, 166, 1-15; Strowig et al., Nature, 2012, 481, 278-286). NLRP3 mutations have been found to be responsible for a set of rare autoinflammatory diseases known as CAPS (Ozaki et al., J. Inflammation Research, 2015, 8, 15-27; Schroder et al., Cell, 2010, 140: 821-832; Menu et al., Clinical and Experimental Immunology, 2011, 166, 1-15). CAPS are heritable diseases characterized by recurrent fever and inflammation and are comprised of three autoinflammatory disorders that form a clinical continuum. These diseases, in order of increasing severity, are familial cold autoinflammatory syndrome (FCAS), Muckle- Wells syndrome (MWS), and chronic infantile cutaneous neurological articular syndrome (CINCA; also called neonatal- onset multisystem inflammatory disease, NOMID), and all have been shown to result from gain-of- function mutations in the NLRP3 gene, which leads to increased secretion of IL-1 beta. NLRP3 has also been implicated in a number of autoinflammatory diseases, including pyogenic arthritis, pyoderma gangrenosum and acne (PAPA), Sweet's syndrome, chronic nonbacterial osteomyelitis (CNO), and acne vulgaris (Cook et al., Eur. J. Immunol., 2010, 40, 595- 653).

A number of autoimmune diseases have been shown to involve NLRP3 including, in particular, multiple sclerosis, type-1 diabetes (T1D), psoriasis, rheumatoid arthritis (RA), Behcet's disease, Schnitzler syndrome, macrophage activation syndrome (Braddock et al., Nat. Rev. Drug Disc. 2004, 3, 1-10; Inoue et a/., Immunology, 2013, 139, 11-18; Coll et a/., Nat. Med. 2015, 21(3), 248-55; Scott etal., Clin. Exp. Rheumatol. 2016, 34(1), 88-93), systemic lupus erythematosus and its complications such as lupus nephritis (Lu et al., J. Immunol. , 2017, 198(3), 1119-29), and systemic sclerosis (Artlett et al., Arthritis Rheum. 2011, 63(11), 3563-74). NLRP3 has also been shown to play a role in a number of lung diseases including chronic obstructive pulmonary disorder (COPD), asthma (including steroid-resistant asthma), asbestosis, and silicosis (De Nardo et al., Am. J. Pathol., 2014, 184: 42-54; Kim et al. , Am. J. Respir. Crit. Care Med, 2017, 196(3), 283-97). NLRP3 has also been suggested to have a role in a number of central nervous system conditions, including Multiple Sclerosis (MS), Parkinson's disease (PD), Alzheimer's disease (AD), dementia, Huntington's disease, cerebral malaria, brain injury from pneumococcal meningitis (Walsh et al., Nature Reviews, 2014, 15, 84-97; and Dempsey et al., Brain. Behav. lmmun. 2017, 61, 306-16), intracranial aneurysms (Zhang et al., J. Stroke and Cerebrovascular Dis., 2015, 24, 5, 972-9), and traumatic brain injury (Ismael et al., J. Neurotrauma., 2018, 35(11), 1294-1303). NLRP3 activity has also been shown to be involved in various metabolic diseases including type 2 diabetes (T2D) and its organ- specific complications, atherosclerosis, obesity, gout, pseudo-gout, metabolic syndrome (Wen et al., Nature Immunology, 2012, 13, 352-357; Duewell et al., Nature, 2010, 464, 1357-1361; Strowig et al., Nature, 2014, 481, 278- 286), and non-alcoholic steatohepatitis (Mridha et al., J. Hepatol. 2017, 66(5), 1037-46). A role for NLRP3 via IL-1 beta has also been suggested in atherosclerosis, myocardial infarction (van Hout et al., Eur. Heart J. 2017, 38(11), 828-36), heart failure (Sano et al., J. Am. Coll. Cardiol. 2018, 71(8), 875- 66), aortic aneurysm and dissection (Wu et al., Arteriosc/er. Thromb. Vase. Biol., 2017,37(4), 694-706), and other cardiovascular events (Ridker et al., N. Engl. J. Med., 2017, 377(12), 1119-31). Other diseases in which NLRP3 has been shown to be involved include: ocular diseases such as both wet and dry age-related macular degeneration (Doyle et al., Nature Medicine, 2012, 18, 791-798; Tarallo et al., Cell 2012, 149(4), 847-59), diabetic retinopathy (Loukovaara et al., Acta Ophthalmol., 2017, 95(8), 803-8), non-infectious uveitis and optic nerve damage (Puyang et al., Sci. Rep.2016, 6, 20998); liver diseases including non-alcoholic steatohepatitis (NASH) and acute alcoholic hepatitis (Henao- Meija et al., Nature, 2012, 482, 179-185); inflammatory reactions in the lung and skin (Primiano et al., J. lmmunol. 2016, 197(6), 2421-33) including contact hypersensitivity (such as bullous pemphigoid (Fang et al., J Dermatol Sci. 2016, 83(2), 116-23)), atopic dermatitis (Niebuhr et al., Allergy, 2014, 69(8), 1058-67), Hidradenitis suppurativa (Alikhan et al., J. Am. Acad. Dermatol. , 2009 ,60(4), 539-61), and sarcoidosis (Jager et al., Am. J. Respir. Crit. Care Med., 2015, 191, A5816); inflammatory reactions in the joints (Braddock et al., Nat. Rev. Drug Disc, 2004, 3, 1-10); amyotrophic lateral sclerosis (Gugliandolo et al., Int. J. Mo/. Sci., 2018, 19(7), E1992); cystic fibrosis (lannitti et al., Nat. Commun., 2016, 7, 10791); stroke (Walsh et al., Nature Reviews, 2014, 15, 84-97); chronic kidney disease (Granata et al., PLoS One 2015, 10(3), eoi22272); and inflammatory bowel diseases including ulcerative colitis and Crohn's disease (Braddock et al., Nat. Rev. Drug Disc, 2004, 3, 1-10; Neudecker et a/., J. Exp. Med.2017, 214(6), 1737-52; Lazaridis et al., Dig. Dis. Sci.2017, 62(9), 2348-56). The NLRP3 inflammasome has been found to be activated in response to oxidative stress. NLRP3 has also been shown to be involved in inflammatory hyperalgesia (Dolunay et al., Inflammation, 2017, 40, 366-86). Activation of the NLRP3 inflammasome has been shown to potentiate some pathogenic infections such as influenza and Leishmaniasis (Tate et al., Sci Rep., 2016, 10(6), 27912-20; Novias et al., PLOS Pathogens 2017, 13(2), e1006196). NLRP3 has also been implicated in the pathogenesis of many cancers (Menu et al., Clinical and Experimental Immunology, 2011, 166, 1-15). For example, several previous studies have suggested a role for IL-1 beta in cancer invasiveness, growth and metastasis, and inhibition of IL-1 beta with canakinumab has been shown to reduce the incidence of lung cancer and total cancer mortality in a randomised, double-blind, placebo-controlled trial (Ridker et al., Lancet. , 2017, 390(10105), 1833-42). Inhibition of the NLRP3 inflammasome or IL-1 beta has also been shown to inhibit the proliferation and migration of lung cancer cells in vitro (Wang et al., Onco/ Rep., 2016, 35(4), 2053-64). A role for the NLRP3 inflammasome has been suggested in myelodysplastic syndromes, myelofibrosis and other myeloproliferative neoplasms, and acute myeloid leukemia (AML) (Basiorka et al., Blood, 2016, 128(25), 2960-75.) and also in the carcinogenesis of various other cancers including glioma (Li et al., Am. J. Cancer Res. 2015, 5(1), 442-9), inflammation- induced tumors (Allen et al., J. Exp. Med. 2010, 207(5), 1045-56; Hu et al., PNAS., 2010, 107(50), 21635-40), multiple myeloma (Li et al., Hematology, 2016 21(3), 144-51), and squamous cell carcinoma of the head and neck (Huang et al., J. Exp. Clin. Cancer Res., 2017, 36(1), 116). Activation of the NLRP3 inflammasome has also been shown to mediate chemoresistance of tumor cells to 5-Fluorouracil (Feng et al., J. Exp. Clin. Cancer Res., 2017, 36(1), 81), and activation of NLRP3 inflammasome in peripheral nerve contributes to chemotherapy-induced neuropathic pain (Jia et al., Mol. Pain., 2017, 13, 1-11). NLRP3 has also been shown to be required for the efficient control of viruses, bacteria, and fungi. The activation of NLRP3 leads to cell pyroptosis and this feature plays an important part in the manifestation of clinical disease (Yan-gang et al., Cell Death and Disease, 2017, 8(2), 2579; Alexander et al., Hepatology, 2014, 59(3), 898-910; Baldwin et al., J. Med. Chem., 2016, 59(5), 1691- 1710; Ozaki et a/., J. Inflammation Research, 2015, 8, 15-27; Zhen et a/., Neuroimmunology Neuroinflammation, 2014, 1(2), 60-65; Mattia et a/., J. Med. Chem., 2014, 57(24), 10366-82; Satoh et al., Cell Death and Disease, 2013, 4, 644). Therefore, it is anticipated that inhibitors of NLRP3 will block pyroptosis, as well as the release of pro-inflammatory cytokines (e.g. IL-1 beta) from the cell. Hence, the compounds of the invention, as described herein (e.g. in any of the embodiments described herein, including by the examples, and/or in any of the forms described herein, e.g. in a salt form or free form, etc) exhibit valuable pharmacological properties, e.g. NLRP3 inhibiting properties on the NLRP3 inflammasome pathway e.g. as indicated in vitro tests as provided herein, and are therefore indicated for therapy or for use as research chemicals, e.g. as tool compounds. Compounds of the invention may be useful in the treatment of an indication selected from: inflammasome-related diseases/disorders, immune diseases, inflammatory diseases, auto-immune diseases, or auto-inflammatory diseases, for example, of diseases, disorders or conditions in which NLRP3 signaling contributes to the pathology, and/or symptoms, and/or progression, and which may be responsive to NLRP3 inhibition and which may be treated or prevented, according to any of the methods/uses described herein, e.g. by use or administration of a compound of the invention, and, hence, in an embodiment, such indications may include:

I. Inflammation, including inflammation occurring as a result of an inflammatory disorder, e.g. an autoinflammatory disease, inflammation occurring as a symptom of a non- inflammatory disorder, inflammation occurring as a result of infection, or inflammation secondary to trauma, injury or autoimmunity. Examples of inflammation that may be treated or prevented include inflammatory responses occurring in connection with, or as a result of: a. a skin condition such as contact hypersensitivity, bullous pemphigoid, sunbum, psoriasis, atopical dermatitis, contact dermatitis, allergic contact dermatitis, seborrhoetic dermatitis, lichen planus, scleroderma, pemphigus, epidermolysis bullosa, urticaria, erythemas, or alopecia; b. a joint condition such as osteoarthritis, systemic juvenile idiopathic arthritis, adult-onset Still's disease, relapsing polychondritis, rheumatoid arthritis, juvenile chronic arthritis, crystal induced arthropathy (e.g. pseudo-gout, gout), or a seronegative spondyloarthropathy (e.g. ankylosing spondylitis, psoriatic arthritis or Reiter's disease); c. a muscular condition such as polymyositis or myasthenia gravis; d. a gastrointestinal tract condition such as inflammatory bowel disease (including Crohn's disease and ulcerative colitis), gastric ulcer, coeliac disease, proctitis, pancreatitis, eosinopilic gastro- enteritis, mastocytosis, antiphospholipid syndrome, or a food-related allergy which may have effects remote from the gut (e.g., migraine, rhinitis or eczema); e. a respiratory system condition such as chronic obstructive pulmonary disease (COPD), asthma (including bronchial, allergic, intrinsic, extrinsic or dust asthma, and particularly chronic or inveterate asthma, such as late asthma and airways hyper- responsiveness), bronchitis, rhinitis (including acute rhinitis, allergic rhinitis, atrophic rhinitis, chronic rhinitis, rhinitis caseosa, hypertrophic rhinitis, rhinitis pumlenta, rhinitis sicca, rhinitis medicamentosa, membranous rhinitis, seasonal rhinitis e.g. hay fever, and vasomotor rhinitis), sinusitis, idiopathic pulmonary fibrosis (IPF), sarcoidosis, farmer's lung, silicosis, asbestosis, adult respiratory distress syndrome, hypersensitivity pneumonitis, or idiopathic interstitial pneumonia; f. a vascular condition such as atherosclerosis, Behcet's disease, vasculitides, or Wegener's granulomatosis; g. an immune condition, e.g. autoimmune condition, such as systemic lupus erythematosus (SLE), Sjogren's syndrome, systemic sclerosis, Hashimoto's thyroiditis, type I diabetes, idiopathic thrombocytopenia purpura, or Graves disease; h. an ocular condition such as uveitis, allergic conjunctivitis, or vernal conjunctivitis; i. a nervous condition such as multiple sclerosis or encephalomyelitis; j. an infection or infection-related condition, such as Acquired Immunodeficiency Syndrome (AIDS), acute or chronic bacterial infection, acute or chronic parasitic infection, acute or chronic viral infection, acute or chronic fungal infection, meningitis, hepatitis (A, B or C, or other viral hepatitis), peritonitis, pneumonia, epiglottitis, malaria, dengue hemorrhagic fever, leishmaniasis, streptococcal myositis, mycobacterium tuberculosis, mycobacterium avium intracellulare, Pneumocystis carinii pneumonia, orchitis/epidydimitis, legionella, Lyme disease, influenza A, epstein-barr virus, viral encephalitis/aseptic meningitis, or pelvic inflammatory disease; k. a renal condition such as mesangial proliferative glomerulonephritis, nephrotic syndrome, nephritis, glomerular nephritis, acute renal failure, uremia, or nephritic syndrome; l. a lymphatic condition such as Castleman's disease; m. a condition of, or involving, the immune system, such as hyper IgE syndrome, lepromatous leprosy, familial hemophagocytic lymphohistiocytosis, or graft versus host disease; n. a hepatic condition such as chronic active hepatitis, non-alcoholic steatohepatitis (NASH), alcohol-induced hepatitis, non-alcoholic fatty liver disease (NAFLD), alcoholic fatty liver disease (AFLD), alcoholic steatohepatitis (ASH) or primary biliary cirrhosis; o. a cancer, including those cancers listed herein below; p. a bum, wound, trauma, haemorrhage or stroke; q. radiation exposure; r. obesity; and/or s. pain such as inflammatory hyperalgesia;

II. Inflammatory disease, including inflammation occurring as a result of an inflammatory disorder, e.g. an autoinflammatory disease, such as cryopyrin- associated periodic syndromes (CAPS), Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome (FC AS), familial Mediterranean fever (FMF), neonatal onset multisystem inflammatory disease (NOMID), Majeed syndrome, pyogenic arthritis, pyoderma gangrenosum and acne syndrome (PAPA), adult-onset Still's disease (AOSD), haploinsufficiency of A20 (HA20), pediatric granulomatous arthritis (PGA), PLCG2-associated antibody deficiency and immune dysregulation (PLAID), PLCG2- associated autoinflammatory, antibody deficiency and immune dysregulation (APLAID), or sideroblastic anaemia with B-cell immunodeficiency, periodic fevers and developmental delay (SIFD);

III. Immune diseases, e.g. auto-immune diseases, such as acute disseminated encephalitis, Addison's disease, ankylosing spondylitis, antiphospholipid antibody syndrome (APS), anti-synthetase syndrome, aplastic anemia, autoimmune adrenalitis, autoimmune hepatitis, autoimmune oophoritis, autoimmune polyglandular failure, autoimmune thyroiditis, Coeliac disease, Crohn's disease, type 1 diabetes (T1D), Goodpasture's syndrome, Graves' disease, Guillain-Barre syndrome (GBS), Hashimoto's disease, idiopathic thrombocytopenic purpura, Kawasaki's disease, lupus erythematosus including systemic lupus erythematosus (SLE), multiple sclerosis (MS) including primary progressive multiple sclerosis (PPMS), secondary progressive multiple sclerosis (SPMS) and relapsing remitting multiple sclerosis (RRMS), myasthenia gravis, opsoclonus myoclonus syndrome (OMS), optic neuritis, Ord's thyroiditis, pemphigus, pernicious anaemia, polyarthritis, primary biliary cirrhosis, rheumatoid arthritis (RA), psoriatic arthritis, juvenile idiopathic arthritis or Still's disease, refractory gouty arthritis, Reiter's syndrome, Sjogren's syndrome, systemic sclerosis a systemic connective tissue disorder, Takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, alopecia universalis, Beliefs disease, Chagas' disease, dysautonomia, endometriosis, hidradenitis suppurativa (HS), interstitial cystitis, neuromyotonia, psoriasis, sarcoidosis, scleroderma, ulcerative colitis, Schnitzler syndrome, macrophage activation syndrome, Blau syndrome, giant cell arteritis, vitiligo or vulvodynia;

IV. Cancer including lung cancer, renal cell carcinoma, non-small cell lung carcinoma (NSCLC), Langerhans cell histiocytosis (LCH), myeloproliferative neoplams (MPN), pancreatic cancer, gastric cancer, myelodysplastic syndrome (MOS), leukaemia including acute lymphocytic leukaemia (ALL) and acute myeloid leukaemia (AML), promyelocytic leukemia (APML, or APL), adrenal cancer, anal cancer, basal and squamous cell skin cancer, bile duct cancer, bladder cancer, bone cancer, brain and spinal cord tumours, breast cancer, cervical cancer, chronic lymphocytic leukaemia (CLL), chronic myeloid leukaemia (CML), chronic myelomonocytic leukaemia (CMML), colorectal cancer, endometrial cancer, oesophagus cancer, Ewing family of tumours, eye cancer, gallbladder cancer, gastrointestinal carcinoid tumours, gastrointestinal stromal tumour (GIST), gestational trophoblastic disease, glioma, Hodgkin lymphoma, Kaposi sarcoma, kidney cancer, laryngeal and hypopharyngeal cancer, liver cancer, lung carcinoid tumour, lymphoma including cutaneous T cell lymphoma, malignant mesothelioma, melanoma skin cancer, Merkel cell skin cancer, multiple myeloma, nasal cavity and paranasal sinuses cancer, nasopharyngeal cancer, neuroblastoma, non-Hodgkin lymphoma, non-small cell lung cancer, oral cavity and oropharyngeal cancer, osteosarcoma, ovarian cancer, penile cancer, pituitary tumours, prostate cancer, retinoblastoma, rhabdomyosarcoma, salivary gland cancer, skin cancer, small cell lung cancer, small intestine cancer, soft tissue sarcoma, stomach cancer, testicular cancer, thymus cancer, thyroid cancer including anaplastic thyroid cancer, uterine sarcoma, vaginal cancer, vulvar cancer, Waldenstrom macroglobulinemia, and Wilms tumour;

V. Infections including viral infections (e.g. from influenza virus, human immunodeficiency virus (HIV), alphavirus (such as Chikungunya and Ross River virus), flaviviruses (such as Dengue virus and Zika virus), herpes viruses (such as Epstein Barr Virus, cytomegalovirus, Varicella-zoster virus, and KSHV), poxviruses (such as vaccinia virus (Modified vaccinia virus Ankara) and Myxoma virus), adenoviruses (such as Adenovirus 5), papillomavirus, or SARS-CoV-2) bacterial infections (e.g. from Staphylococcus aureus, Helicobacter pylori, Bacillus anthracis, Bordatella pertussis, Burkholderia pseudomallei, Corynebacterium diptheriae, Clostridium tetani, Clostridium botulinum, Streptococcus pneumoniae, Streptococcus pyogenes, Listeria monocytogenes, Hemophilus influenzae, Pasteurella multicida, Shigella dysenteriae, Mycobacterium tuberculosis, Mycobacterium leprae, Mycoplasma pneumoniae, Mycoplasma hominis, Neisseria meningitidis, Neisseria gonorrhoeae, Rickettsia rickettsii, Legionella pneumophila, Klebsiella pneumoniae, Pseudomonas aeruginosa, Propionibacterium acnes, Treponema pallidum, Chlamydia trachomatis, Vibrio cholerae, Salmonella typhimurium, Salmonella typhi, Borrelia burgdorferi or Yersinia pestis), fungal infections (e.g. from Candida or Aspergillus species), protozoan infections (e.g. from Plasmodium, Babesia, Giardia, Entamoeba, Leishmania or Trypanosomes), helminth infections (e.g. from schistosoma, roundworms, tapeworms or flukes), and prion infections;

VI. Central nervous system diseases such as Parkinson's disease, Alzheimer's disease, dementia, motor neuron disease, Huntington's disease, cerebral malaria, brain injury from pneumococcal meningitis, intracranial aneurysms, traumatic brain injury, multiple sclerosis, and amyotrophic lateral sclerosis;

VII. Metabolic diseases such as type 2 diabetes (T2D), atherosclerosis, obesity, gout, and pseudo-gout;

VIII. Cardiovascular diseases such as hypertension, ischaemia, reperfusion injury including post-Ml ischemic reperfusion injury, stroke including ischemic stroke, transient ischemic attack, myocardial infarction including recurrent myocardial infarction, heart failure including congestive heart failure and heart failure with preserved ejection fraction, embolism, aneurysms including abdominal aortic aneurysm, cardiovascular risk reduction (CvRR), and pericarditis including Dressier's syndrome;

IX. Respiratory diseases including chronic obstructive pulmonary disorder (COPD), asthma such as allergic asthma and steroid-resistant asthma, asbestosis, silicosis, nanoparticle induced inflammation, cystic fibrosis, and idiopathic pulmonary fibrosis;

X. Liver diseases including non-alcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) including advanced fibrosis stages F3 and F4, alcoholic fatty liver disease (AFLD), and alcoholic steatohepatitis (ASH);

XL Renal diseases including acute kidney disease, hyperoxaluria, chronic kidney disease, oxalate nephropathy, nephrocalcinosis, glomerulonephritis, and diabetic nephropathy; XII. Ocular diseases including those of the ocular epithelium, age-related macular degeneration (AMO) (dry and wet), uveitis, comeal infection, diabetic retinopathy, optic nerve damage, dry eye, and glaucoma;

XIII. Skin diseases including dermatitis such as contact dermatitis and atopic dermatitis, contact hypersensitivity, sunburn, skin lesions, hidradenitis suppurativa (HS), other cyst-causing skin diseases, and acne conglobata;

XIV. Lymphatic conditions such as lymphangitis, and Castleman's disease;

XV. Psychological disorders such as depression, and psychological stress;

XVI. Graft versus host disease;

XVII. Bone diseases including osteoporosis, osteopetrosis;

XVIII. Blood disease including sickle cell disease;

XIX. Allodynia including mechanical allodynia; and

XX. Any disease where an individual has been determined to carry a germline or somatic non-silent mutation in NLRP3.

More specifically the compounds of the invention may be useful in the treatment of an indication selected from: inflammasome-related diseases/ disorders, immune diseases, inflammatory diseases, auto-immune diseases, or auto- inflammatory diseases, for example, autoinflammatory fever syndromes (e.g., ciyopyrin-associated periodic syndrome), sickle cell disease, systemic lupus erythematosus (SLE), liver related diseases/disorders (e.g. chronic liver disease, viral hepatitis, non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis, and alcoholic liver disease), inflammatory arthritis related disorders (e.g. gout, pseudogout (chondrocalcinosis), osteoarthritis, rheumatoid arthritis, arthropathy e.g acute, chronic), kidney related diseases (e.g. hyperoxaluria, lupus nephritis, Type I/Type II diabetes and related complications (e.g. nephropathy, retinopathy), hypertensive nephropathy, hemodialysis related inflammation), neuroinflammation- related diseases (e.g. multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, Alzheimer's disease), cardiovascular/metabolic diseases/disorders (e.g. cardiovascular risk reduction (CvRR), hypertension, atherosclerosis, Type I and Type II diabetes and related complications, peripheral artery disease (PAD), acute heart failure), inflammatory skin diseases (e.g. hidradenitis suppurativa, acne), wound healing and scar formation, asthma, sarcoidosis, age-related macular degeneration, and cancer related diseases/disorders (e.g. colon cancer, lung cancer, myeloproliferative neoplasms, leukemias, myelodysplastic syndromes (MOS), myelofibrosis). In particular, autoinflammatory fever syndromes (e.g. CAPS), sickle cell disease, Type I/Type II diabetes and related complications (e.g. nephropathy, retinopathy), hyperoxaluria, gout, pseudogout (chondrocalcinosis), chronic liver disease, NASH, neuroinflammation- related disorders (e.g. multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, Alzheimer's disease), atherosclerosis and cardiovascular risk (e.g. cardiovascular risk reduction (CvRR), hypertension), hidradenitis suppurativa, wound healing and scar formation, and cancer (e.g. colon cancer, lung cancer, myeloproliferative neoplasms, leukemias, myelodysplastic syndromes (MOS), myelofibrosis).

In particular, compounds of the invention, may be useful in the treatment of a disease or disorder selected from autoinflammatory fever syndromes (e.g. CAPS), sickle cell disease, Type 1/ Type II diabetes and related complications (e.g. nephropathy, retinopathy), hyperoxaluria, gout, pseudogout (chondrocalcinosis), chronic liver disease, NASH, neuroinflammation-related disorders (e.g. multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, Alzheimer's disease), atherosclerosis and cardiovascular risk (e.g. cardiovascular risk reduction (CvRR), hypertension), hidradenitis suppurativa, wound healing and scar formation, and cancer (e.g. colon cancer, lung cancer, myeloproliferative neoplasms, leukemias, myelodysplastic syndromes (MOS), myelofibrosis). Thus, as a further aspect, the present invention provides the use of a compound of the invention (hence, including a compound as defined by any of the embodiments/forms/examples herein) in therapy. In a further embodiment, the therapy is selected from a disease, which may be treated by inhibition of NLRP3 inflammasome. In another embodiment, the disease is as defined in any of the lists herein. Hence, there is provided any one of the compounds of the invention described herein (including any of the embodiments/forms/examples) for use in the treatment of any of the diseases or disorders described herein (e.g. as described in the aforementioned lists).

PHARMACEUTICAL COMPOSITIONS AND COMBINATIONS

In an embodiment, the invention also relates to a composition comprising a pharmaceutically acceptable carrier and, as active ingredient, a therapeutically effective amount of a compound of the invention. The compounds of the invention may be formulated into various pharmaceutical forms for administration purposes. As appropriate compositions there may be cited all compositions usually employed for systemically administering drugs. To prepare the pharmaceutical compositions of this invention, an effective amount of the particular compound, optionally in salt form, as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration. These pharmaceutical compositions are desirable in unitary dosage form suitable, in particular, for administration orally or by parenteral injection. For example, in preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs, emulsions and solutions; or solid carriers such as starches, sugars, kaolin, diluents, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules and tablets. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit forms in which case solid pharmaceutical carriers are obviously employed. For parenteral compositions, the carrier will usually comprise sterile water, at least in large part, though other ingredients, for example, to aid solubility, may be included. Injectable solutions, for example, may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution. Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed. Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations.

In an embodiment, and depending on the mode of administration, the pharmaceutical composition will preferably comprise from 0.05 to 99 % by weight, more preferably from 0.1 to 70 % by weight, even more preferably from 0.1 to 50 % by weight of the active ingredient(s), and, from 1 to 99.95 % by weight, more preferably from 30 to 99.9 % by weight, even more preferably from 50 to 99.9 % by weight of a pharmaceutically acceptable carrier, all percentages being based on the total weight of the composition.

The pharmaceutical composition may additionally contain various other ingredients known in the art, for example, a lubricant, stabilising agent, buffering agent, emulsifying agent, viscosity-regulating agent, surfactant, preservative, flavouring or colorant.

It is especially advantageous to formulate the aforementioned pharmaceutical compositions in unit dosage form for ease of administration and uniformity of dosage. Unit dosage form as used herein refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. Examples of such unit dosage forms are tablets (including scored or coated tablets), capsules, pills, powder packets, wafers, suppositories, injectable solutions or suspensions and the like, and segregated multiples thereof.

The daily dosage of the compound according to the invention will, of course, vary with the compound employed, the mode of administration, the treatment desired and the mycobacterial disease indicated. However, in general, satisfactory results will be obtained when the compound according to the invention is administered at a daily dosage not exceeding 1 gram, e.g. in the range from 10 to 50 mg/kg body weight.

In an embodiment, there is provided a combination comprising a therapeutically effective amount of a compound of the invention, according to any one of the embodiments described herein, and another therapeutic agent (including one or more therapeutic agents). In a further embodiment, there is provided such a combination wherein the other therapeutic agent is selected from (and where there is more than one therapeutic agent, each is independently selected from): famesoid X receptor (FXR) agonists; anti-steatotics; anti- fibrotics; JAK inhibitors; checkpoint inhibitors including anti-PDl inhibitors, anti-LAG- 3 inhibitors, anti-TIM-3 inhibitors, or anti-POL 1 inhibitors; chemotherapy, radiation therapy and surgical procedures; urate-lowering therapies; anabolics and cartilage regenerative therapy; blockade of IL-17; complement inhibitors; Bruton's tyrosine Kinase inhibitors (BTK inhibitors); Toll Like receptor inhibitors (TLR7/8 inhibitors); CAR-T therapy; anti-hypertensive agents; cholesterol lowering agents; leukotriene A4 hydrolase (LTAH4) inhibitors; SGLT2 inhibitors; 132-agonists; anti-inflammatory agents; nonsteroidal anti-inflammatory drugs ("NSAIDs"); acetylsalicylic acid drugs (ASA) including aspirin; paracetamol; regenerative therapy treatments; cystic fibrosis treatments; or atherosclerotic treatment. In a further embodiment, there is also provided such (a) combination(s) for use as described herein in respect of compounds of the invention, e.g. for use in the treatment of a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder, or, a disease or disorder associated with NLRP3 activity (including NLRP3 inflammasome activity), including inhibiting NLRP3 inflammasome activity, and in this respect the specific disease/disorder mentioned herein apply equally here. There may also be provided methods as described herein in repsect of compounds of the invention, but wherein the method comprises administering a therapeutically effective amount of such combination (and, in an embodiment, such method may be to treat a disease or disorder mentioned herein in the context of inhibiting NLRP3 inflammasome activity). The combinations mentioned herein may be in a single preparation or they may be formulated in separate preparations so that they can be administered simultaneously, separately or sequentially. Thus, in an embodiment, the present invention also relates to a combination product containing (a) a compound according to the invention, according to any one of the embodiments described herein, and (b) one or more other therapeutic agents (where such therapeutic agents are as described herein), as a combined preparation for simultaneous, separate or sequential use in the treatment of a disease or disorder associated with inhibiting NLRP3 inflammasome activity (and where the disease or disorder may be any one of those described herein), for instance, in an embodiment, the combination may be a kit of parts. Such combinations may be referred to as “pharmaceutical combinations”. The route of administration for a compound of the invention as a component of a combination may be the same or different to the one or more other therapeutic agent(s) with which it is combined. The other therapeutic agent is, for example, a chemical compound, peptide, antibody, antibody fragment or nucleic acid, which is therapeutically active or enhances the therapeutic activity when administered to a patient in combination with a compound of the invention.

The weight ratio of (a) the compound according to the invention and (b) the other therapeutic agent(s) when given as a combination may be determined by the person skilled in the art. Said ratio and the exact dosage and frequency of administration depends on the particular compound according to the invention and the other antibacterial agent(s) used, the particular condition being treated, the severity of the condition being treated, the age, weight, gender, diet, time of administration and general physical condition of the particular patient, the mode of administration as well as other medication the individual may be taking, as is well known to those skilled in the art. Furthermore, it is evident that the effective daily amount may be lowered or increased depending on the response of the treated subject and/or depending on the evaluation of the physician prescribing the compounds of the instant invention. A particular weight ratio for the present compound of the invention and another antibacterial agent may range from 1/10 to 10/1, more in particular from 1/5 to 5/1, even more in particular from 1/3 to 3/1.

The pharmaceutical composition or combination of the present invention can be in unit dosage of about 1-1000 mg of active ingredient(s) for a subject of about 50 - 70 kg, or about 1 - 500 mg, or about 1 - 250 mg, or about 1 - 150 mg, or about 1 - 100 mg, or about 1 - 50 mg of active ingredients. The therapeutically effective dosage of a compound, the pharmaceutical composition, or the combinations thereof, is dependent on the species of the subject, the body weight, age and individual condition, the disorder or disease or the severity thereof being treated. A physician, clinician or veterinarian of ordinary skill can readily determine the effective amount of each of the active ingredients necessary to prevent, treat or inhibit the progress of the disorder or disease.

The above-cited dosage properties are demonstrable in vitro and in vivo tests using advantageously mammals, e.g., mice, rats, dogs, monkeys or isolated organs, tissues and preparations thereof. The compounds of the present invention can be applied in vitro in the form of solutions, e.g, aqueous solutions, and in vivo either enterally, parenterally, advantageously intravenously, e.g, as a suspension or in aqueous solution. The dosage in vitro may range between about 10-3 mo l ar and 10-9 m olar concentrations. A therapeutically effective amount in vivo may range depending on the route of administration, between about 0.1 - 500 mg/kg, or between about 1 - 100 mg/kg.

As used herein, term "pharmaceutical composition" refers to a compound of the invention, or a pharmaceutically acceptable salt thereof, together with at least one pharmaceutically acceptable carrier, in a form suitable for oral or parenteral administration.

As used herein, the term "pharmaceutically acceptable carrier" refers to a substance useful in the preparation or use of a pharmaceutical composition and includes, for example, suitable diluents, solvents, dispersion media, surfactants, antioxidants, preservatives, isotonic agents, buffering agents, emulsifiers, absorption delaying agents, salts, drug stabilizers, binders, excipients, disintegration agents, lubricants, weting agents, sweetening agents, flavoring agents, dyes, and combinations thereof, as would be known to those skilled in the art (see, for example, Remington The Science and Practice of Pharmacy, 22nd Ed. Pharmaceutical Press, 2013, pp. 1049- 1070).

The term "subject" as used herein, refers to an animal, preferably a mammal, most preferably a human, for example who is or has been the object of treatment, observation or experiment.

The term "therapeutically effective amount" as used herein, means that amount of compound of the invention (including, where applicable, form, composition, combination comprising such compound of the invention) elicits the biological or medicinal response of a subject, for example, reduction or inhibition of an enzyme or a protein activity, or ameliorate symptoms, alleviate conditions, slow or delay disease progression, or prevent a disease, etc. In one non-limiting embodiment, the term "a therapeutically effective amount" refers to the amount of the compound of the present invention that, when administered to a subject, is effective to (1) at least partially alleviate, inhibit, prevent and/ or ameliorate a condition, or a disorder or a disease (i) mediated by NLRP3, or (ii) associated with NLRP3 activity, or (iii) characterised by activity (normal or abnormal) of NLRP3; or (2) reduce or inhibit the activity of NLRP3; or (3) reduce or inhibit the expression of NLRP3. In another non-limiting embodiment, the term "a therapeutically effective amount" refers to the amount of the compound of the present invention that, when administered to a cell, or a tissue, or a non-cellular biological material, or a medium, is effective to at least partially reduce or inhibit the activity of NLRP3; or at least partially reduce or inhibit the expression of NLRP3.

As used herein, the term "inhibit", "inhibition" or "inhibiting" refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process. Specifically, inhibiting NLRP3 or inhibiting NLRP3 inflammasome pathway comprises reducing the ability ofNLRP3 orNLRP3 inflammasome pathway to induce the production of IL-1 and/or IL-18. This can be achieved by mechanisms including, but not limited to, inactivating, destabilizing, and/or altering distribution of NLRP3. As used herein, the term "NLRP3" is meant to include, without limitation, nucleic acids, polynucleotides, oligonucleotides, sense and anti-sense polynucleotide strands, complementary sequences, peptides, polypeptides, proteins, homologous and/or orthologous NLRP molecules, isoforms, precursors, mutants, variants, derivatives, splice variants, alleles, different species, and active fragments thereof.

As used herein, the term "treat", "treating" or "treatment" of any disease or disorder refers to alleviating or ameliorating the disease or disorder (i.e., slowing or arresting the development of the disease or at least one of the clinical symptoms thereol); or alleviating or ameliorating at least one physical parameter or biomarker associated with the disease or disorder, including those which may not be discernible to the patient.

As used herein, the term "prevent", "preventing" or "prevention" of any disease or disorder refers to the prophylactic treatment of the disease or disorder; or delaying the onset or progression of the disease or disorder.

As used herein, a subject is "in need of’ a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.

"Combination" refers to either a fixed combination in one dosage unit form, or a combined administration where a compound of the present invention and a combination partner (e.g. another drug as explained below, also referred to as "therapeutic agent" or "coagent") may be administered independently at the same time or separately within time intervals. The single components may be packaged in a kit or separately. One or both of the components (e.g. powders or liquids) may be reconstituted or diluted to a desired dose prior to administration. The terms "co- administration" or "combined administration" or the like as utilized herein are meant to encompass administration of the selected combination partner to a single subject in need thereof (e.g. a patient), and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.

The term "pharmaceutical combination" as used herein means a product that results from the mixing or combining of more than one therapeutic agent and includes both fixed and non-fixed combinations of the therapeutic agents. The term "pharmaceutical combination" as used herein refers to either a fixed combination in one dosage unit form, or non-fixed combination or a kit of parts for the combined administration where two or more therapeutic agents may be administered independently at the same time or separately within time intervals. The term "fixed combination" means that the therapeutic agents, e.g. a compound of the present invention and a combination partner, are both administered to a patient simultaneously in the form of a single entity or dosage. The term "non-fixed combination" means that the therapeutic agents, e.g. a compound of the present invention and a combination partner, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more therapeutic agents.

The term "combination therapy" refers to the administration of two or more therapeutic agents to treat a therapeutic condition or disorder described in the present disclosure. Such administration encompasses co-administration of these therapeutic agents in a substantially simultaneous manner, such as in a single capsule having a fixed ratio of active ingredients. Alternatively, such administration encompasses co-administration in multiple, or in separate containers (e.g. tablets, capsules, powders, and liquids) for each active ingredient. Powders and/or liquids may be reconstituted or diluted to a desired dose prior to administration. In addition, such administration also encompasses use of each type of therapeutic agent in a sequential manner, either at approximately the same time or at different times. In either case, the treatment regimen will provide beneficial effects of the drug combination in treating the conditions or disorders described herein.

Summary of pharmacology, uses, compositions and combinations

In an embodiment, there is provided a pharmaceutical composition comprising a therapeutically effective amount of a compound of the invention, according to any one of the embodiments described herein, and a pharmaceutically acceptable carrier (including one or more pharmaceutically acceptale carriers).

In an embodiment, there is provided a compound of the invention, according to any one of the embodiments described herein, for use as a medicament.

In an embodiment, there is provided a compound of the invention, according to any one of the embodiments described herein (and/or pharmaceutical compositions comprising such compound of the invention, according to any one of the embodiment described herein) for use: in the treatment of a disease or disorder associated with NLRP3 activity (including inflammasome activity); in the treatment of a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder; in inhibiting NLRP3 inflammasome activity (including in a subject in need thereol); and/or as an NLRP3 inhibitor.

In an embodiment, there is provided a use of compounds of the invention, according to any one of the embodiments described herein (and/or pharmaceutical compositions comprising such compound of the invention, according to any one of the embodiment described herein): in the treatment of a disease or disorder associated with NLRP3 activity (including inflammasome activity); in the treatment of a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder; in inhibiting NLRP3 inflammasome activity (including in a subject in need thereof); and/or as an NLRP3 inhibitor.

In an embodiment, there is provided use of compounds of the invention, according to any one of the embodiments described herein (and/or pharmaceutical compositions comprising such compound of the invention, according to any one of the embodiment described herein), in the manufacture of a medicament for: the treatment of a disease or disorder associated with NLRP3 activity (including inflammasome activity); the treatment of a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder; and/or inhibiting NLRP3 inflammasome activity (including in a subject in need thereol).

In an embodiment, there is provided a method of treating a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder, comprising administering a therapeutically effective amount of a compound of the invention, according to any one of the embodiments described herein (and/or pharmaceutical compositions comprising such compound of the invention, according to any one of the embodiment described herein), for instance to a subject (in need thereol). In a further embodiment, there is provided a method of inhibiting the NLRP3 inflammasome activity in a subject (in need thereof), the method comprising administering to the subject in need thereof a therapeutically effective amount of a compound of the invention, according to any one of the embodiments described herein (and/or pharmaceutical compositions comprising such compound of the invention, according to any one of the embodiment described herein).

In all relevant embodiment of the invention, where a disease or disorder is mentioned (e.g. hereinabove), for instance a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder, or, a disease or disorder associated with NLRP3 activity (including NLRP3 inflammasome activity), including inhibiting NLRP3 inflammasome activity, then such disease may include inflammasome-related diseases or disorders, immune diseases, inflammatory diseases, auto-immune diseases, or auto-inflammatory diseases. In a further embodiment, such disease or disorder may include autoinfl ammatory fever syndromes (e.g cryopyrin-associated periodic syndrome), liver related diseases/disorders (e.g. chronic liver disease, viral hepatitis, non-alcoholic steatohepatitis (NASH), alcoholic steatohepatitis, and alcoholic liver disease), inflammatory arthritis related disorders (e.g. gout, pseudogout (chondrocalcinosis), osteoarthritis, rheumatoid arthritis, arthropathy e.g acute, chronic), kidney related diseases (e.g. hyperoxaluria, lupus nephritis, Type I/Type II diabetes and related complications (e.g. nephropathy, retinopathy), hypertensive nephropathy, hemodialysis related inflammation), neuroinflammation-related diseases (e.g. multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, Alzheimer's disease), cardiovascular/ metabolic diseases/ disorders (e.g. cardiovascular risk reduction (CvRR), hypertension, atherosclerosis, Type I and Type II diabetes and related complications, peripheral artery disease (PAD), acute heart failure), inflammatory skin diseases (e.g. hi dradenitis suppurativa, acne), wound healing and scar formation, asthma, sarcoidosis, age-related macular degeneration, and cancer related diseases/ disorders (e.g. colon cancer, lung cancer, myeloproliferative neoplasms, leukaemia, myelodysplastic syndromes (MOS), myelofibrosis). In a particular aspect, such disease or disorder is selected from autoinflammatory fever syndromes (e.g. CAPS), sickle cell disease, Type I/Type II diabetes and related complications (e.g. nephropathy, retinopathy), hyperoxaluria, gout, pseudogout (chondrocalcinosis), chronic liver disease, NASH, neuroinflammation-related disorders (e.g. multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, Alzheimer's disease), atherosclerosis and cardiovascular risk (e.g. cardiovascular risk reduction (CvRR), hypertension), hidradenitis suppurativa, wound healing and scar formation, and cancer (e.g. colon cancer, lung cancer, myeloproliferative neoplasms, leukemias, myelodysplastic syndromes (MOS), myelofibrosis). In a particular embodiment, the disease or disorder associated with inhibition of NLRP3 inflammasome activity is selected from inflammasome related diseases and disorders, immune diseases, inflammatory diseases, auto-immune diseases, auto-inflammatory fever syndromes, cryopyrin-associated periodic syndrome, chronic liver disease, viral hepatitis, nonalcoholic steatohepatitis, alcoholic steatohepatitis, alcoholic liver disease, inflammatory arthritis related disorders, gout, chondrocalcinosis, osteoarthritis, rheumatoid arthritis, chronic arthropathy, acute arthropathy, kidney related disease, hyperoxaluria, lupus nephritis, Type I and Type II diabetes, nephropathy, retinopathy, hypertensive nephropathy, hemodialysis related inflammation, neuroinflammation-related diseases, multiple sclerosis, brain infection, acute injury, neurodegenerative diseases, Alzheimer’s disease, cardiovascular diseases, metabolic diseases, cardiovascular risk reduction, hypertension, atherosclerosis, peripheral artery disease, acute heart failure, inflammatory skin diseases, acne, wound healing and scar formation, asthma, sarcoidosis, age-related macular degeneration, colon cancer, lung cancer, myeloproliferative neoplasms, leukemias, myelodysplastic syndromes and myelofibrosis.

In an embodiment, there is provided a combination comprising a therapeutically effective amount of a compound of the invention, according to any one of the embodiments described herein, and another therapeutic agent (including one or more therapeutic agents). In a further embodiment, there is provided such a combination wherein the other therapeutic agent is selected from (and where there is more than one therapeutic agent, each is independently selected from): famesoid X receptor (FXR) agonists; anti-steatotics; anti- fibrotics; JAK inhibitors; checkpoint inhibitors including anti-PDl inhibitors, anti -LAG- 3 inhibitors, anti-TIM-3 inhibitors, or anti-POL 1 inhibitors; chemotherapy, radiation therapy and surgical procedures; urate-lowering therapies; anabolics and cartilage regenerative therapy; blockade of IL-17; complement inhibitors; Bruton's tyrosine Kinase inhibitors (BTK inhibitors); Toll Like receptor inhibitors (TLR7/8 inhibitors); CAR-T therapy; anti-hypertensive agents; cholesterol lowering agents; leukotriene A4 hydrolase (LTAH4) inhibitors; SGLT2 inhibitors; 132-agonists; anti-inflammatory agents; nonsteroidal anti-inflammatory drugs ("NSAIDs"); acetylsalicylic acid drugs (ASA) including aspirin; paracetamol; regenerative therapy treatments; cystic fibrosis treatments; or atherosclerotic treatment. In a further embodiment, there is also provided such (a) combination(s) for use as described herein in respect of compounds of the invention, e.g. for use in the treatment of a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder, or, a disease or disorder associated with NLRP3 activity (including NLRP3 inflammasome activity), including inhibiting NLRP3 inflammasome activity, and in this respect the specific disease/disorder mentioned herein apply equally here. There may also be provided methods as described herein in repsect of compounds of the invention, but wherein the method comprises administering a therapeutically effective amount of such combination (and, in an embodiment, such method may be to treat a disease or disorder mentioned herein in the context of inhibiting NLRP3 inflammasome activity). The combinations mentioned herein may be in a single preparation or they may be formulated in separate preparations so that they can be administered simultaneously, separately or sequentially. Thus, in an embodiment, the present invention also relates to a combination product containing (a) a compound according to the invention, according to any one of the embodiments described herein, and (b) one or more other therapeutic agents (where such therapeutic agents are as described herein), as a combined preparation for simultaneous, separate or sequential use in the treatment of a disease or disorder associated with inhibiting NLRP3 inflammasome activity (and where the disease or disorder may be any one of those described herein).

Compounds of the invention (including forms and compositions/combinations comprising compounds of the invention) may have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (e.g. higher oral bioavailability and/or lower clearance) than, and/or have other useful pharmacological, physical, or chemical properties over, compounds known in the prior art, whether for use in the above-stated indications or otherwise.

For instance, compounds of the invention may have the advantage that they have a good or an improved thermodynamic solubility (e.g. compared to compounds known in the prior art; and for instance as determined by a known method and/or a method described herein). Compounds of the invention may have the advantage that they will block pyroptosis, as well as the release of pro-inflammatory cytokines (e.g. IL-1 ^) from the cell. Compounds of the invention may also have the advantage that they avoid side-effects, for instance as compared to compounds of the prior art, which may be due to selectivity of NLRP3 inhibition. Compounds of the invention may also have the advantage that they have good or improved in vivo pharmacokinetics and oral bioavailabilty. They may also have the advantage that they have good or improved in vivo efficacy. Specifically, compounds of the invention may also have advantages over prior art compounds when compared in the tests outlined hereinafter (e.g. in Examples C and D). GENERAL PREPARATION AND ANALYTICAL PROCESSES The compounds according to the invention can generally be prepared by a succession of steps, each of which may be known to the skilled person or described herein. It is evident that in the foregoing and in the following reactions, the reaction products may be isolated from the reaction medium and, if necessary, further purified according to methodologies generally known in the art, such as extraction, crystallization and chromatography. It is further evident that reaction products that exist in more than one enantiomeric form, may be isolated from their mixture by known techniques, in particular preparative chromatography, such as preparative HPLC, chiral chromatography. Individual diastereoisomers or individual enantiomers can also be obtained by Supercritical Fluid Chromatography (SFC). The starting materials and the intermediates are compounds that are either commercially available or may be prepared according to conventional reaction procedures generally known in the art. Analytical Part LC-MS (LIQUID CHROMATOGRAPHY/MASS SPECTROMETRY) General procedure The High Performance Liquid Chromatography (HPLC) measurement was performed using a LC pump, a diode-array (DAD) or a UV detector and a column as specified in the respective methods. If necessary, additional detectors were included (see table of methods below). Flow from the column was brought to the Mass Spectrometer (MS) which was configured with an atmospheric pressure ion source. It is within the knowledge of the skilled person to set the tune parameters (e.g. scanning range, dwell time…) in order to obtain ions allowing the identification of the compound’s nominal monoisotopic molecular weight (MW). Data acquisition was performed with appropriate software. Compounds are described by their experimental retention times (R t ) and ions. If not specified differently in the table of data, the reported molecular ion corresponds to the [M+H]+ (protonated molecule) and/or [M-H]- (deprotonated molecule). In case the compound was not directly ionizable the type of adduct is specified (i.e. [M+NH + 4] , [M+HCOO]-, etc…). For molecules with multiple isotopic patterns (Br, Cl..), the reported value is the one obtained for the lowest isotope mass. All results were obtained with experimental uncertainties that are commonly associated with the method used. Hereinafter, “SQD” means Single Quadrupole Detector, “MSD” Mass Selective Detector, “RT” room temperature, “BEH” bridged ethylsiloxane/silica hybrid, “DAD” Diode Array Detector, ”HSS” High Strength silica. Table: LCMS Method codes (Flow expressed in mL/min; column temperature (T) in °C; Run time in minutes). Method Instrume Flow l mn m bil h r di nt ------ Run e Method Instrume Flow l mn m bil h r di nt ------ Run e For a number of compounds, 1H NMR spectra were recorded on a Bruker Avance III spectrometer operating at 300 or 400 MHz, on a Bruker Avance III-HD operating at 400 MHz, on a Bruker Avance NEO spectrometer operating at 400 MHz, on a Bruker Avance Neo spectrometer operating at 500 MHz, or on a Bruker Avance 600 spectrometer operating at 600 MHz, using CHLOROFORM-d (deuterated chloroform, CDCl 3 ), DMSO-d 6 (deuterated DMSO, dimethyl-d6 sulfoxide), METHANOL-d4 (deuterated methanol), BENZENE-d6 (deuterated benzene, C6D6) or ACETONE-d6 (deuterated acetone, (CD3)2CO) as solvents. Chemical shifts ( ^) are reported in parts per million (ppm) relative to tetramethylsilane (TMS), which was used as internal standard. Melting Points Values are either peak values or melt ranges, and are obtained with experimental uncertainties that are commonly associated with this analytical method. Method A: For a number of compounds, melting points were determined with a DSC823e (Mettler Toledo) apparatus. Melting points were measured with a temperature gradient of 10 ºC/minute. Standard maximum temperature was 300 ºC. Method B: For a number of compounds, melting points were determined in open capillary tubes on a Mettler Toledo MP50. Melting points were measured with a temperature gradient of 10 ºC/minute. Maximum temperature was 300 ºC. The melting point data was read from a digital display and checked from a video recording system EXPERIMENTAL PART Hereinafter, the term “m.p.” means melting point, “aq.” means aqueous, “r.m.” means reaction mixture, “rt” means room temperature, ‘DIPEA’ means N,N-diiso- propylethylamine, “DIPE” means diisopropylether, ‘THF’ means tetrahydrofuran, ‘DMF’ means dimethylformamide, ‘DCM’ means dichloromethane, “EtOH” means ethanol ‘EtOAc’ means ethyl acetate, “AcOH” means acetic acid, “iPrOH” means isopropanol, “iPrNH2” means isopropylamine, “MeCN” or “ACN” means acetonitrile, “MeOH” means methanol, “Pd(OAc)2” means palladium(II)diacetate, “rac” means racemic, ‘sat.’ means saturated, ‘SFC’ means supercritical fluid chromatography, ‘SFC-MS’ means supercritical fluid chromatography/mass spectrometry, “LC-MS” means liquid chromatography/mass spectrometry, “GCMS” means gas chromatography/mass spectrometry, “HPLC” means high-performance liquid chromatography, “RP” means reversed phase, “UPLC” means ultra-performance liquid chromatography, “R t ” (or “RT”) means retention time (in minutes), “[M+H]+” means the protonated mass of the free base of the compound, “DAST” means diethylaminosulfur trifluoride, “DMTMM” means 4-(4,6-dimethoxy-1,3,5-triazin-2- yl)-4-methylmorpholinium chloride, “HATU” means O-(7-azabenzotriazol-1-yl)- N,N,N’,N’-tetramethyluronium hexafluorophosphate (1-[bis(dimethylamino)methylene]- 1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate), “Xantphos” means (9,9- dimethyl-9H-xanthene-4,5-diyl)bis[diphenylphosphine], “TBAT” means tetrabutyl ammonium triphenyldifluorosilicate, “TFA” means trifuoroacetic acid, “Et2O” means diethylether, “DMSO” means dimethylsulfoxide, “SiO2” means silica, “XPhos Pd G3” means (2-dicyclohexylphosphino-2′,4′,6′-triisopropyl-1,1′- biphenyl)[2-(2′-amino-1,1′- biphenyl)]palladium(II) ethanesulfonate, “CDCl3” means deuterated chloroform, “MW” means microwave or molecular weight, “min” means minutes, “h” means hours, “rt” means room temperature, “quant” means quantitative, “n.t.” means not tested, “Cpd” means compound, “POCl 3 ” means phosphorus(V) oxychloride. For key intermediates, as well as some final compounds, the absolute configuration of chiral centers (indicated as R and/or S) were established via comparison with samples of known configuration, or the use of analytical techniques suitable for the determination of absolute configuration, such as VCD (vibrational cicular dichroism) or X-ray crystallography. When the absolute configuration at a chiral center is unknown, it is arbitrarily designated R*. Examples – Example A PREPARATION OF INTERMEDIATES Synthesis of 4-(2-amino-4-bromophenyl)but-3-yn-1-ol (I-1) To a s e (147.0 mg, 0.77 mmol) and tetrakis(triphenylphosphine)palladium(0) (446.1 mg, 0.39 mmol) in degassed DI water (187 mL) and under nitrogen, were added 3-butyn-1-ol (3.07 mL, 40.5 mmol) and triethylamine (8.05 mL, 57.9 mmol) simultaneously via syringe. The mixture was stirred vigorously at r.t. for 18 hours. The reaction mixture was extracted with EtOAc (4 x 80 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated in vacuo. The obtained brown residue was purified by column chromatography eluting with gradient elution of Hept/EtOAc (4:1 to 0:1) to give 4-(2-amino-4- bromophenyl)but-3-yn-1-ol I-1 as a thick brown oil (9.35 g, quantitative). 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 1.99 (t, J=5.7 Hz, 1 H), 2.72 (t, J=6.2 Hz, 2 H), 3.82 (q, J=6.0 Hz, 2 H), 4.27 (br s, 2 H), 6.78 (dd, J=8.2, 1.9 Hz, 1 H), 6.84 (d, J=1.9 Hz, 1 H), 7.09 (d, J=8.3 Hz, 1 H). LCMS (Rt = 1.63 min, 96% (UV), m/z (ES+) = 240.1; m/z (ES-) = N.D. Method 1. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Starting material Product Synthesis of 7-bromo-3-(2-hydroxyethyl)cinnolin-4(1H)-one (I-4) To 4-(2- ed a solution of H2SO4 (8.3 mL, 155.8 mmol) in DI water (75 mL). The mixture was cooled to 0 °C for 10 minutes, then a solution of sodium nitrite (4.03 g, 58.4 mmol) in DI water (112 mL) was added dropwise over 45 min at 0 °C. After addition, the mixture was allowed to warm slowly to r.t. (removing the ice from the ice-bath, but leaving the water at 0 °C to warm slowly to r.t.) and stirred overall for 6h. The pale brownish suspension was filtered off, washed with DI water (2 x 10 mL) and the wet paste dried under vacuum at 50 °C for 16 h to afford 7- bromo-3-(2-hydroxyethyl)cinnolin-4(1H)-one I-4 as a pale brown solid (9.35 g, yield 89%). 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 2.85 (t, J=6.9 Hz, 2 H), 3.71 (t, J=6.9 Hz, 2 H), 7.50 (dd, J=8.6, 1.8 Hz, 1 H), 7.72 (d, J=1.8 Hz, 1 H), 7.95 (d, J=8.6 Hz, 1 H), 13.23 (s, 1 H). LCMS Rt = 1.22 min, 100% (UV), m/z (ES+) = 269.0 / 271.0; m/z (ES-) = 267.0 / 269.0, Method 1. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of 7-bromo-3-(2-((triisopropylsilyl)oxy)ethyl)cinnolin-4(1H)-on e (I-7) T mmol) and imidazole (2.84 g, 41.7 mmol) in dry DMF (80.7 mL), under nitrogen and at 0 °C was added triisopropylsilyl chloride (7.8 mL, 36.5 mmol) dropwise. After addition, the ice-bath was removed and the mixture allowed to warm to r.t. and stirred for 16h. The mixture was concentrated to about 10-15 mL of solution and poured onto brine (100 mL) and extracted with EtOAc (5 x 100 mL). The combined organic extracts were concentrated in vacuo and the obtained residue redissolved in DCM and filtered over a pad of Na2SO4. The solution was concentrated to obtain 7-bromo-3-(2-((triisopropylsilyl)oxy)ethyl)cinnolin-4(1H)-on e I-7 as a brown solid (15.9 g, 92% wt purity, yield 99%) that was used without further purification. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 0.96 - 1.08 (m, 21 H), 3.13 (t, J=6.9 Hz, 2 H), 4.10 (t, J=6.9 Hz, 2 H), 7.42 (dd, J=8.8, 1.7 Hz, 1 H), 7.77 (d, J=1.6 Hz, 1 H), 8.13 (d, J=8.8 Hz, 1 H). LCMS Rt = 2.59 min, 98% (UV), m/z (ES+) = 425.2 / 427.2; m/z (ES-) = 423.3 / 425.3, Method 1. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of 7-cyclopropyl-3-(2-((triisopropylsilyl)oxy)ethyl)cinnolin-4( 1H)-one (I-10) l), cesium carbonate [534-17-8] (12.9 g, 39.592 mmol) and Pd(dppf)Cl2·CH2Cl2 [95464-05-4] (1.46 g, 1.788 mmol) were charged in a 250 mL round-bottom flask that was left under nitrogen (3 vacuum/nitrogen cycles).1,4-Dioxane (60 mL) and DI water (20 mL) were added to the solids and the resulting reaction mixture was stirred overnight at 90 °C under nitrogen. It was then partitioned between EtOAc (~100 mL) and brine (~300 mL). The organic layer was collected and the aqueous layer further extracted with EtOAc (3 x ~100 mL). The combined organic layers were dried over MgSO4, filtered and concentrated under reduced pressure at 40 °C. The crude material was purified by flash column chromatography (Hept/EtOAc 1:0 to 3:2) to afford I-10 (2.885 g, yield 61%) as a white solid. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.76 - 0.84 (m, 2 H), 0.89 - 1.04 (m, 22 H), 1.04 - 1.11 (m, 2 H), 2.04 - 2.14 (m, 1 H), 2.91 (t, J=7.04 Hz, 2 H), 3.98 (t, J=6.93 Hz, 2 H), 7.04 (dd, J=8.58, 1.54 Hz, 1 H), 7.19 (d, J=1.54 Hz, 1 H), 7.90 (d, J=8.58 Hz, 1 H). LCMS Rt = 2.57 min, 96% (UV), m/z (ES+) = 387; m/z (ES-) = 385, Method 1. Synthesis of 1-isopropyl-7-(trifluoromethyl)-3-(2-((triisopropylsilyl)oxy )ethyl)-cinnolin- 4(1H)-one (I-11) )-one I-8 (15.8 g, 37.73 mmol), 2-iodopropane [75-30-9] (4.52 mL, 45.28 mmol) and K 2 CO 3 [584-08- 7] (10.43 g, 75.47 mmol) in dry acetonitrile [75-05-8] (98 mL) was stirred and heated at 90 °C in a pressure tube for 18 h. The solvent was evaporated and the residue was taken in water and extracted with EtOAc. The organic layer is separated, washed with brine and dried on MgSO4 and filtered. The solvent was evaporated and the residue was purified by column chromatography eluting with gradient elution of Hept/EtOAc (1:0 to 85:15) to afford 1- isopropyl-7-(trifluoromethyl)-3-(2-((triisopropylsilyl)-oxy) ethyl)cinnolin-4(1H)-one I-11 as a yellow oil (10.99 g, yield 63%). 1H NMR (300 MHz, DMSO-d6) δ ppm 0.91 – 1.00 (m, 21 H), 1.43 (d, J=6.4 Hz, 6 H), 2.98 (t, J=6.6 Hz, 2 H), 4.05 (t, J=6.6 Hz, 2 H), 5.30 - 5.46 (m, 1 H), 7.66 - 7.73 (m, 1 H), 8.30 - 8.37 (m, 2 H). LCMS Rt = 1.62 min, 99% (UV), m/z (ES+) = 457; m/z (ES-) = -, Method 6. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Reagent Substrate Product Reagent Substrate Product Synthesis of 1-cyclopropyl-7-(trifluoromethyl)-3-(2-((triisopropylsilyl)o xy)ethyl)cinnolin- 4(1H)-one (I-18) 2 g, 12.11 mmol) and copper(II) acetate [142-71-2] (6.67 g, 36.722 mmol) in 1,4-dioxane (40 mL) in a 100 mL round-flask and the mixture was stirred 15 min at 110 °C. The mixture was then allowed to cool to rt and cyclopropylboronic acid [411235-57-9] (2.75 g, 32.015 mmol) was added. The reaction mixture was then stirred overnight at 110 °C. Further portions of cyclopropylboronic acid [411235-57-9] (3.02 g, 35.158 mmol) and copper(II) acetate [142- 71-2] (1.1 g, 6.056 mmol) were added and the reaction was stirred 4 h at 110 °C. Cyclopropylboronic acid [411235-57-9] (908 mg, 10.571 mmol) and copper(II) acetate [142-71-2] (3.08 g, 16.957 mmol) were added and the reaction was stirred 1 h at 120 °C and then overnight at rt. The reaction mixture was allowed to cool to rt and flushed with oxygen during 5 min. and final portions of cyclopropylboronic acid [411235-57-9] (1.26 g, 14.668 mmol) and copper(II) acetate [142-71-2] (2.5 g, 13.764 mmol) were added and the reaction was stirred 3 h at 110 °C. The reaction mixture was quenched with a mixture of water (~150 mL) and EtOAc (~50 mL) and the product was extracted with EtOAc (3 x 50 mL). The combined organic extracts were dried over MgSO4, filtered and concentrated under reduced pressure at 40 °C. The crude material was purified by flash column chromatography (Hept/EtOAc 1:0 to 7:3) to afford I-17 (4.2 g, yield 76%). 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 0.86 - 1.06 (m, 21 H), 1.08 - 1.16 (m, 2 H), 1.20 - 1.29 (m, 2 H), 2.93 (t, J=6.7 Hz, 2 H), 3.92 (tt, J=7.1, 3.6 Hz, 1 H), 4.00 (t, J=6.6 Hz, 2 H), 7.74 (dd, J=8.5, 1.4 Hz, 1 H), 8.30 (d, J=8.5 Hz, 1 H), 8.32 (s, 1 H). LCMS Rt = 2.89 min, 100% (UV), m/z (ES+) = 455.4; m/z (ES-) = -, Method 1. Synthesis of 7-bromo-1-cyclopropyl-3-(2-((triisopropylsilyl)oxy)ethyl)cin nolin-4(1H)-one (I-19) d [411235- 57-9] (1.8 g, 21.51 mmol) were added to a solution of 7-bromo-3-(2- ((triisopropylsilyl)oxy)ethyl)cinnolin-4(1H)-one I-7 (3.7 g, 8.60 mmol) and pyridine (2.1 mL) in 1,4-dioxane (36.7 mL) in an open-air system. A condenser and an oxygen balloon were fitted on the reaction set-up and the system was evacuated and backfilled with oxygen three times. The greenish reaction mixture was heated at reflux (110 °C) for 10.5 h and stirred at rt over the weekend. The reaction mixture was then quenched with water and a 25% aqueous ammonia solution (1 mL) and extracted with DCM three times. The combined organic layers were dried over MgSO 4 , filtered and concentrated in vacuo. The brown crude product was purified by flash column chromatography (DCM) to afford 7-bromo-1- cyclopropyl-3-(2-((triisopropylsilyl)oxy)ethyl)cinnolin-4(1H )-one I-19 (3.5 g, yield 87%) as an orange solid. 1H NMR (400 MHz, CHLOROFORM-d) δ ppm 0.96 - 1.11 (m, 22 H), 1.24 (d, J=5.3 Hz, 5 H), 3.05 (t, J=6.7 Hz, 2 H), 3.48 - 3.54 (m, 1 H), 4.06 (t, J=6.7 Hz, 2 H), 7.47 (dd, J=8.7, 1.7 Hz, 1 H), 8.04 (d, J=1.5 Hz, 1 H), 8.16 (d, J=8.6 Hz, 1 H). LCMS Rt = 2.98 min, 100% (UV), m/z (ES+) = 465.3 / 467.3; m/z (ES-) = -, Method 2. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. b P d Synthesis of 7-cyclopropyl-1-ethyl-3-(2-((triisopropylsilyl)oxy)ethyl)cin nolin-4(1H)-one (I-21) trogen were added Pd(tBu3P)2 [53199-31-8] (225.4 mg, 0.44 mmol) followed by a commercial solution of cPr-ZnBr in Me-THF [126403-68-7] (22.05 mL, 0.5 M, 11.03 mmol). The reaction mixture was stirred under inert atmosphere for 3 h at rt. It was quenched by addition of DI water (20 mL) then an aqueous saturated solution of NH4Cl (200 mL), poured onto brine (100 mL) and extracted with EtOAc (3 x 80 mL). The combined organic extracts were dried over Na2SO4, filtered, concentrated and the residue purified by FCC (Hept/EtOAc 7:3 to 0:1) to give I-21 as a white solid that was used as such in the next step.LCMS Rt = 1.61 min, 100% (UV), m/z (ES+) = 415.4; m/z (ES-) = -, Method 5.Synthesis of 7-cyclopropyl-1- isopropyl-3-(2-((triisopropylsilyl)oxy)ethyl)cinnolin-4(1H)- one (I-22) er (80 mL). The solution was sparged with N2 for 5 min in a sealed tube. Then, cyclopropylboronic acid [411235-57-9] (1.76 g, 20.53 mmol), cesium carbonate [534-17-8] (12.27 g, 37.65 mmol) and Pd(dppf)Cl2·CH2Cl2 [95464-05-4] (2.096 g, 2.57 mmol) were sequentially added. After sparging the resulting mixture with N 2 for 5 min, it was stirred at 90 ºC for 16h. The mixture was then diluted with DI water and extracted with EtOAc. The combined organic layers were dried (MgSO 4 ), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (Hept/EtOAc 1:0 to 4:1) to yield I-22 (5.25 g, yield 72%) as a pale yellow oil. 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 0.90 (dd, J=4.8, 2.1 Hz, 2 H), 0.91 – 1.02 (m, 21 H), 1.06 - 1.12 (m, 2 H), 1.41 (d, J=6.4 Hz, 6 H), 2.02 - 2.27 (m, 1 H), 2.92 (t, J=6.7 Hz, 2 H), 4.01 (t, J=6.7 Hz, 2 H), 5.23 (dt, J=12.8, 6.4 Hz, 1 H), 7.05 (dd, J=8.5, 0.9 Hz, 1 H), 7.59 (s, 1 H), 8.00 (d, J=8.5 Hz, 1 H). LCMS Rt = 1.79 min, 99% (UV), m/z (ES+) = 429.3; m/z (ES-) = -, Method 8. Synthesis of 7-(1-ethoxyvinyl)-1-ethyl-3-(2-((triisopropylsilyl)oxy)ethyl )cinnolin-4(1H)- one (I-23) (II) [13965-03-2] (1.1 g, 1.57 mmol) in 1,4-dioxane (125 mL) was sparged with N2 for 10 min. Then tributyl(1-ethoxyvinyl)tin [97674-02-7] (7.93 mL, 23.48 mmol) was added and the mixture was heated at 100 ºC for 4 h. The crude mixture was allowed to cool to rt and concentrated under vacuum. The resulting material was treated with a saturated aqueous solution of NaHCO 3 and extracted with EtOAc (2 x 200 ml). The combine organic layers were evaporated in vacuo and purified by flash column chromatography (Hept/EtOAc 1:0 to 4:1) to yield I-23 (6.01 g, yield 86%) as a yellow oil. LCMS Rt = 1.68 min, 99% (UV), m/z (ES+) = 445; m/z (ES-) = -, Method 4. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of 1-ethyl-7-(prop-1-en-2-yl)-3-(2-((triisopropylsilyl)oxy)ethy l)cinnolin-4(1H)- one (I-26) I water (24 mL). The solution was sparged with N 2 for 5 min in a sealed tube. Then, potassium isopropenyltrifluoroborate [395083-14-4] (1.57 g, 10.58 mmol), cesium carbonate [534-17- 8] (6.32 g, 19.4 mmol) and Pd(dppf)Cl2·CH2Cl2 [95464-05-4] (1.08 g, 1.32 mmol) were added equentially. After sparging for further 5 min with N2, the mixture was stirred at 90 ºC for 16h. It was then diluted with DI water and extracted with EtOAc. The combined organic layers were washed with brine, dried (MgSO4), filtered and the solvents evaporated in vacuo. The crude product was purified by flash column chromatography (Hept/EtOAc 1:0 to 4:1) to yield I-26 (2.8 g, yield 77%) as a colorless oil. LCMS Rt = 1.64 min, 95% (UV), m/z (ES+) = 415; m/z (ES-) = -, Method 5. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of 1-ethyl-7-isopropyl-3-(2-((triisopropylsilyl)oxy)ethyl)cinno lin-4(1H)-one (I- 29) in EtOH (40 mL) in a 250 mL hydrogenation flask. The reaction vessel was purged with hydrogen (3 hydrogen/vacuum cycles) and placed under hydrogen atmosphere. The reaction was stirred at rt until completion was observed by LCMS. The solides were then filtered on a pad of Decalite and the filtrate was concentrated and the obtained residue was used without purification. Note: partial deprotection of the TIPS is observed. LCMS Rt = 1.66 min, 37% (UV), m/z (ES+) = 417.4; m/z (ES-) = - & Rt = 0.82 min, 53% (UV), m/z (ES+) = 261.2; m/z (ES-) = - , Method 5. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of 1-ethyl-7-(2-hydroxypropan-2-yl)-3-(2-((triisopropylsilyl)ox y)ethyl)cinnolin- 4(1H)-one (I-32) ol), NaHCO 3 [144-55-8] (770 mg; 9.2 mmol) and Fe(acac) 3 [14024-18-1] (40 mg; 0.1 mmol) were dissolved/suspended in dry MeOH (35 mL) in a sealed tube. The mixture was sparged with N2 for 5 min and cooled to 0 ºC. Phenylsilane [694-53-1] (1.7 mL; 13.7 mmol) was then added at 0 ºC and the mixture allowed to slowly warm to rt and stirred at rt for 16h. The solvent was removed under vacuum to obtain a yellow solid which was purified by flash column chromatography (Hept/EtOAc 1:0 to 0:1) to yield I-32 (373 mg; ca. 80% purity, yield 15%) as a yellow oil. 1H NMR (300 MHz, DMSO-d6) δ ppm 0.95 - 1.00 (m, 21 H), 1.39 (t, J=7.1 Hz, 3 H), 1.50 (s, 6 H), 2.92 (t, J=6.6 Hz, 2 H), 3.96 - 4.07 (m, 2 H), 4.47 (q, J=7.1 Hz, 2 H), 5.37 (s, 1 H), 7.53 (dd, J=8.6, 1.3 Hz, 1 H), 7.73 (s, 1 H), 8.06 (d, J=8.5 Hz, 1 H). LCMS Rt = 1.27 min, 80% (UV), m/z (ES+) = 433.2; m/z (ES-) = -, Method 6. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of 7-bromo-3-(2-hydroxyethyl)-1-isopropylcinnolin-4(1H)-one (I-34) To a 1H)-one I-13 (5.1 g, 10.91 mmol) in dry THF (16 mL), under nitrogen and at 0 °C, was added dropwise over 5 min TBAF (1M in THF) (16 mL, 16 mmol). The reaction mixture was warmed to R.T. and stirred for 2 h. The liquids were removed and the crude was purified by column chromatography eluting with gradient elution of Hept/EtOAc (1:0 to 1:1) to afford 7-bromo-3-(2-hydroxyethyl)-1-isopropylcinnolin-4(1H)-one I-34 (2.78 g, yield 82%) as a white powder. 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 1.41 (d, J=6.4 Hz, 6 H), 2.88 (t, J=7.0 Hz, 2 H), 3.67 - 3.79 (m, 2 H), 4.56 (t, J=5.6 Hz, 1 H), 5.14 - 5.29 (m, 1 H), 7.56 (dd, J=8.7, 1.7 Hz, 1 H), 8.04 (d, J=8.6 Hz, 1 H), 8.22 (d, J=1.5 Hz, 1 H). LCMS Rt = 1.67 min, 100% (UV), m/z (ES+) = 311; m/z (ES-) = -, Method 1. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Intermediate Product Synthesis of 7-acetyl-1-ethyl-3-(2-hydroxyethyl)cinnolin-4(1H)-one (I-51) 3 (6.01 g, 13.52 mmol) in 1,4-dioxane (67 mL). The reaction mixture was stirred at rt for 16h. It was then treated with a saturated aqueous solution of NaHCO3 and extracted with EtOAc. The combined organic layers were washed with brine, dried over MgSO 4 , filtered and evaporated in vacuo. The obtained residue was stirred in 150 ml DIPE and the solids were filtered and dried under vacuum to yield I-51 (3.3 g, yield 94%) as a pale yellow solid. LCMS Rt = 0.57 min, 92% (UV), m/z (ES+) = 261; m/z (ES-) = -, Method 4. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Intermediate Product Synthesis of 7-bromo-3-(2-hydroxyethyl)-1-isopropylcinnolin-4(1H)-one (I-54) A co s added dropwise to a solution of I-35 (8.32 g, 28 mmol) in acetone [67-64-1] (514.6 mL) at 0 °C. After the addition was complete, the mixture was allowed to reach rt and stirred for 30 min. It was then poured onto DI water (1.5 L) and the resulting solution/suspension stirred for 30- 60 min. The solids were filtered out, washed with water (2x) and heptane (2x) and dried at 50 C under vacuum to afford I-54 (9.17 g, 95% purity, yield 82%) as a white solid. 1H NMR (300 MHz, DMSO-d6) δ ppm 1.36 (t, J=7.1 Hz, 3 H), 3.61 (s, 2 H), 4.49 (q, J=7.1 Hz, 2 H), 7.62 (dd, J=8.7, 1.6 Hz, 1 H), 8.03 (d, J=8.7 Hz, 1 H), 8.15 (d, J=1.5 Hz, 1 H), 12.45 (s, 1 H). LCMS Rt = 0.84 min, 95% (UV), m/z (ES+) = 311.1; m/z (ES-) = -, Method 6.Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of ethyl 2-(7-bromo-l-isopropyl-4-oxo-l,4-dihydrocinnolin-3-yl)acetat e (1-74)

1-Propanephosphonic anhydride [68957-94-8] (50%wt in EtOAc, 1.2 mL, 2.016 mmol,) was added to a solution of 1-55 (300 mg, 0.793 mmol) andEtsN [121-44-8] (0.4 mL, 2.878 mmol) in EtOH (4.5 mL). The reaction was stirred overnight at rt. The crude mixture was concentrated under reduced pressure at 40 °C and the product was purified by flash column chromatography (Hept/EtOAc 1:0 to 0:1) to afford 1-74 (229 mg, yield 82%) as a colorless oil.

LCMS Rt = 2.03 min, 100% (UV), m/z (ES+) = 353; m/z (ES-) = -, Method 1. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of methyl 2-(7-bromo-1-isopropyl-4-oxo-1,4-dihydrocinnolin-3-yl)acetat e (I-87) Iod 4 (7.56 g, 24.3 mmol) and cesium carbonate [534-17-8] (10.29 g, 31.59 mmol) in DMF at rt. The mixture was stirred at rt for 1 h. The mixture was diluted with sat. aqueous NaHCO 3 and extracted with EtOAc. The combined organic layers were dried over MgSO 4 , filtered and concentrated in vacuo. The crude material was purified by flash column chromatography (Hept/EtOAc 1:0 to 4:1) to yield I-87 (6.32 g, yield 79%) as a yellow solid. 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 1.36 (t, J=7.1 Hz, 3 H), 3.61 (s, 3 H), 3.71 (s, 3 H), 4.49 (q, J=7.1 Hz, 2 H), 7.63 (dd, J=8.7, 1.6 Hz, 1 H), 8.03 (d, J=8.7 Hz, 1 H),8.16 (d, J=1.6 Hz, 1 H). LCMS Rt = 2.96 min, 99% (UV), m/z (ES+) = 311.1; m/z (ES-) = -, Method 7.Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate.

Synthesis of ethyl 2-(7-(l,l-difluoroethyl)-l-ethyl-4-oxo-l,4-dihydrocinnolin-3 -yl)acetate (1-95)

DAST [38078-09-0] (5.9 mL, 44.65 mmol) and (HF) 3 Et 3 N [73602-61-6] (4.37 mL, 26.79 mmol) were added to a solution of 1-84 (2.7 g, 8.93 mmol) in dry DCM (20 ml) at rt. After addition., the mixture was stirred at 47 °C for 24 hours. It was then cooled with an ice-bath and quenched with a saturated aqueous solution of NaHCO 3 (dropwise). The organic layer was separated, dried over Na 3 SO4 and concentrated in vacuo. The crude material was purified by flash column chromatography (Hept/EtOAc 1 :0 to 3:2) to yield 1-95 (2.7 g, yield 93%) as an off white solid.

LCMS Rt = 0.96 min, 100% (UV), m/z (ES+) = 325; m/z (ES-) = -, Method 5. Additional analogs were synthesized according to the above procedure substituting the Intermediate Product Synthesis of methyl 2-(1-ethyl-7-(2-fluoropropan-2-yl)-4-oxo-1,4-dihydrocinnolin -3- yl)acetate (I-98) mol) were added to a mixture of I-93 (127 mg, 0.4 mmol) in dry DCM (4 mL) at rt in a sealed plastic tube. The mixture was stirred at 50 ºC for 16h. The crude reaction mixtue was cooled at 0 ºC and quenched with a saturated aqueous solution of NaHCO3. It was then extracted with DCM, the combined organic layers dried over MgSO 4 , filtered and concentrated in vacuo. The crude product was purified by flash column chromatography (Hept/EtOAc 1:0 to 1:1) to yield I-98 (109 mg, yield 84%) as a yellow solid. 1H NMR (300 MHz, DMSO-d6) δ ppm 1.39 (t, J=7.1 Hz, 3 H), 1.72 (s, 3 H), 1.79 (s, 3 H), 3.61 (s, 3 H), 3.72 (s, 2 H), 4.54 (q, J=7.2 Hz, 2 H), 7.55 (dd, J=8.6, 1.3 Hz, 1 H), 7.71 (s, 1 H), 8.14 (d, J=8.5 Hz, 1 H). LCMS Rt = 0.81 min, 99% (UV), m/z (ES+) = 307.2; m/z (ES-) = -, Method 6. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of methyl 2-(7-cyclobutyl-1-ethyl-4-oxo-1,4-dihydrocinnolin-3-yl)aceta te (I-100) O O OMe OMe lution of I- 87 (520 mg, 1.6 mmol), potassium cyclobutyltrifluoroborate [1065010-88-9] (285 mg, 1.76 mmol), cesium carbonate [534-17-8] (1.56 g, 4.8 mmol) and Pd(OAc) 2 [3375-31-3] (35.9 mg, 0.16 mmol) in a mixture of toluene (6.4 mL) and DI water (0.6 mL) while sparging with N2. The resulting mixture was heated at 100 ºC for 16h. The reaction mixture was diluted with DI water and the biphasic mixture extracted with DCM. The combined organic extracts were dried over MgSO 4 , filtered and concentrated in vacuo. The crude product was purified by flash column chromatography (Hept/EtOAc 1:0 to 7:3) to yield I-100 (209 mg, 95% purity, yield 41%) as a white solid. LCMS Rt = 1.21 min, 95% (UV), m/z (ES+) = 301.1; m/z (ES-) = -, Method 6. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of 2-(7-cyclobutyl-1-ethyl-4-oxo-1,4-dihydrocinnolin-3-yl)aceti c acid (I-102) (202 mg, 0.67 mmol) in a mixture of THF (6.84 mL) and DI water (1.69 mL) at rt. The reaction mixture was stirred at 30 ºC for 16h. A 1M aqueous solution of HCl was added until pH=4. The mixture was diluted with DI water and extracted with EtOAc. The combined organic extracts were dried over MgSO4, filtered and concentrated in vacuo to yield I-102 (195 mg, 98% purity, yield 99%) as a yellow solid. The crude product was used in the next step without further purification. 1H NMR (300 MHz, DMSO-d 6 ) δ ppm 1.38 (t, J=7.1 Hz, 3 H), 1.86 (dd, J=10.8, 9.3 Hz, 1 H), 1.99 - 2.13 (m, 1 H), 2.15 - 2.30 (m, 2 H), 2.32 - 2.45 (m, 2 H), 3.60 (s, 2 H), 3.64 - 3.85 (m, 1 H), 4.51 (q, J=7.1 Hz, 2 H), 7.40 (dd, J=8.4, 1.0 Hz, 1 H), 7.54 (s, 1 H), 8.06 (d, J=8.4 Hz, 1 H), 12.17 (s, 1 H). LCMS Rt = 0.97 min, 98% (UV), m/z (ES+) = 287.2; m/z (ES-) = -, Method 6. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of 2-(7-(1,1-difluoroethyl)-1-ethyl-4-oxo-1,4-dihydrocinnolin-3 -yl)acetic acid (I- 106) dded to a stirred solution of I-95 (1.8 g, 5.55 mmol) in 1:1 THF/DI water (11.1 mL:11.1 mL). The resulting mixture was stirred for 2h at rt. It was then neutralized with a 1M aqueous solution of HCL (11.1 ml) and the volatiles were concentrated. The residue was diluted with Di water (20 mL) and extracted with EtOAc (2x). The combined organic extracts were washed with brine, dried over MgSO 4 , filtered and concentrated under vacuum to yield I-106 (1.6 g, yield 97%) as an off-white solid. LCMS Rt = 0.52 min, 100% (UV), m/z (ES+) = 297; m/z (ES-) = -, Method 5. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of ethyl 6-iodoimidazo[1,5-a]pyridine-1-carboxylate (I-109) h NaH (60% dispersion in mineral oil) [7646-69-7] (2,24 g, 56.06 mmol) under N 2 . The mixture was cooled to 0 °C. Ethyl isocyanoacetate [2999-46-4] (6.13 mL, 56.06 mmol) was added dropwise and after 30 min, 2-fluoro-5-iodo-pyridine [171197-80-1] (10.0 g, 44.85 mmol) was added in three portions. The reaction was allowed to warm to rt and then heated at 60 °C for 16 hours. The reaction was allowed to cooled to rt and diluted with EtOAc (500 mL) and DI water (300 mL). The organic layer was separated and washed with brine (2 x 100 mL). The combined aqueous layers were extracted with EtOAc (200 mL). The combined organic layers were dried over MgSO 4 , filtered and concentrated under vacuum. The crude product was purified by FCC (Hept/EtOAc 1:0 to 3:7) to obtain I-109 (2.94 g, yield 21%) as an off-white solid. LCMS Rt = 0.84 min, 97% (UV), m/z (ES+) = 317.0; m/z (ES-) = -, Method 4. Synthesis of 6-iodoimidazo[1,5-a]pyridine-1-carboxylic acid (I-110) ol) was added to a solution of I-109 (3.75 g, 11.86 mmol) in THF (35 mL). The reaction was stirred at 60 °C for 2 hours. It was then concentrated under reduced pressure to and then treated with 1M aqueous HCl until pH slightly acidic. The solid precipitate was filtered off, washed with water and then dried at 50 °C under vacuum to yield I-110 (3.25 g, yield 95%) as an off-white solid. Synthesis of 6-iodoimidazo[1,5-a]pyridine-1-carbonyl chloride (I-111) e to a suspension of I-110 (3.25 g, 11.28 mmol) in dry MeCN (30 mL). The reaction was stirred at 60 °C for 1 hour. Volatiles were removed under reduced pressure and the crude product I-111 (3.45 g, quantitative) was used without further treatment in the following step. Synthesis of 6-iodoimidazo[1,5-a]pyridine-1-carboxylic acid (I-112) 0 mL) was cooled to 0 °C. A 28%wt aqueous solution of NH3 [7664-41-7] (50 mL, 740 mmol) was added portionwise and the mixture was allowed to warm to rt and stirred for 1 hour. The reaction mixture was filtered and the solid cake was washed with water and then dried under vacuum at 50 °C for 16 hours to obtain I-112(2.29g, yield 71%) as a brown solid. LCMS Rt = 0.60 min, 100% (UV), m/z (ES+) = 288.0; m/z (ES-) = -, Method 4. Synthesis of 6-iodoimidazo[1,5-a]pyridine-1-carbonitrile (I-113) tion of I-112 (2.29 g, 7.98 mmol) in anhydrous DMF (23 mL) stirring at 0 °C. The reaction was allowed to warm to rt and stirred for 30 min. The reaction mixture was quenched with ice (~50 mL) and diluted with EtOAc (400 mL) and DI water (150 mL). The organic layer was separated and the aqueous layer extracted again with EtOAc (100 mL). The combined organic layers were dried over MgSO 4 , filtered and concentrated under reduced pressure to yield desired I-113 (2.09 g, yield 97%) as a brown solid. LCMS Rt = 0.76 min, 99% (UV), m/z (ES+) = 270.1; m/z (ES-) = -, Method 4. Synthesis of 6-((diphenylmethylene)amino)imidazo[1,5-a]pyridine-1-carboni trile (I-114) 8-3] (0.65 mL, 3.875 mmol), BINAP [98327-87-8] (322 mg, 0.517 mmol) and sodium tert-butoxide [865-48-5] (397 mg, 4.133 mmol) in anhydrous 1,4-dioxane (28 mL) was sparged with N2 for a few minutes. Pd 2 (dba) 3 [51364-51-3] (237 mg, 0.258 mmol) was added and the reaction was heated at 60 °C for 2 hours. The reaction mixture was allowed to cool to rt and filtered through Celite® (washing wiht EtOAc). The filtrate was concentrated under reduced pressure to give a brown paste. The crude product was purified by FCC (Hept/EtOAc 1:0 to 3:2) to obtain I-114 (360 mg, yield 43%) as a yellow solid. LCMS Rt = 1.08 min, 99% (UV), m/z (ES+) = 323.2; m/z (ES-) = -, Method 4. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of 6-aminoimidazo[1,5-a]pyridine-1-carbonitrile (I-115) d to a solution of I-114 (360 mg, 1.12 mmol) in THF (5 mL). The reaction was stirred at rt for 1 hour. The reaction was diluted with DCM (10 ml), the organic layer was collected and the aqueous layer was treated with solid K 2 CO 3 until saturation and extracted with DCM (5 x 20 mL). The combined organic layers were dried over MgSO 4 , filtered and concentrated to yield I-115 (130 mg, yield 74%) as a light brown solid. LCMS Rt = 0.96 min, 100% (UV), m/z (ES+) = 159.1; m/z (ES-) = 157.1, Method 2 Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of N-((5-bromopyridin-2-yl)methylene)-4-methylbenzenesulfonohyd razide (I- 116) mmol) was added to a solution of 5-bromopyridine-2-carbaldehyde [31181-90-5] (1.0 g, 5.38 mmol) in DCM (10 mL) and MeOH (10 mL). The reaction was stirred at rt for 1 hour. Volatiles were removed under reduced pressure and the obtained solid I-116 was used without purification in the following step. LCMS Rt = 0.90 min, 100% (UV), m/z (ES+) = 354.0 / 356.0; m/z (ES-) = -, Method 4. Synthesis of 6-bromo-[1,2,3]triazolo[1,5-a]pyridine (I-117) antitative, 5.71 g, 16.12 mmol) and morpholine [110-91-8] (25 mL, 289.8 mmol) were stirred at 90 °C for 1 hour. The reaction was allowed to cool to rt and concentrated under reduced pressure. The crude product was purified by FCC (Hept/EtOAc 1:0 to 2:3) to obtain I-117 (3.08 g, yield 96%) as a white solid. LCMS Rt = 0.61 min, 100% (UV), m/z (ES+) = 198.0 / 200.0; m/z (ES-) = -, Method 4. Synthesis of 3-chloro-1-(difluoromethyl)-5-nitropyridin-2(1H)-one (I-120) , 12.5 mmol) was added to a solution of 3-chloro-2-hydroxy-5-nitropyridine [22353-38-4] (2 g, 11.46 mmol) in DMSO (20 mL) under nitrogen at rt. The mixture was allowed to react for 15 minutes at rt and sodium chlorodifluoroacetate [1895-39-2] (2 g, 13.12 mmol) was added. The resulting solution was then heated at 60° C for 16 hours. It was allowed to cool to rt and quenched by addition of DI water. The resulting solution was extracted with EtOAc (3x) and the combined organic layers dried over MgSO 4 , filtered and concentrated under vacuum. The crude product was purified by FCC (Hept/EtOAc 1:0 to 3:2) to obtain I-120 (390 mg, yield 15%) as a white solid. LCMS Rt = 1.50 min, 100% (UV), m/z (ES+) = -; m/z (ES-) = 223.0, Method 2. Synthesis of 5-amino-3-chloro-1-(difluoromethyl)pyridin-2(1H)-one (I-121) mg, 3.83 mmol) and a 7.2M aqueous solution of NH 4 Cl [12125-02-9] (1.21 mL, 8.73 mmol) in EtOH (5 mL) was heated at 80°C for 16 hours in a sealed MW vial under nitrogen atmosphere. The reaction mixture was allowed to cool rt and filtered over Dicalite®, washing throughly with EtOH. The filtrate was evaporated under reduced pressure, suspended in DCM and filtered again. The filtrate was concentrated and purifed by FCC (DCM/MeOH 1:0 to 95:5) to obtain I-121 (110 mg, yield 44%) as a green solid. LCMS Rt = 1.02 min, 100% (UV), m/z (ES+) = 194.9; m/z (ES-) = -, Method 2. Synthesis of 9-methyl-9H-purin-2-amine (I-122) mmol) and Pd/C (10%wt, 289.8 mg, 0.27 mmol) in MeOH (30 mL) and THF (50 mL) was placed under hydrogen atmosphere (1 atm) and stirred at rt for 16 hours. The catalyst was filtered off on Decalite® and the filtrate was concentrated to obtain I-122 (780 mg, yield 85%) as a red solid. LCMS Rt = 0.75 min, 79% (UV), m/z (ES+) = 150.0; m/z (ES-) = -, Method 2. Synthesis of 5-chloro-3-methyl-3H-imidazo[4,5-b]pyridine (I-123) and 5-chloro-1-methyl- 1H-imidazo[4,5-b]pyridine (I-124) portionwise to a stirred mixture of 5-chloro-3H-imidazo[4,5-b]pyridine [52090-89-8] (5 g, 32.56 mmol) in anhydrous DMF (70 mL) at 0 °C. The resulting mixture was allowed to warm to rt and stirred 30 min before dropwise addition of MeI [74-88-4] (2.3 mL, 36.95 mmol). After 2 hours, the mixture was quenched with DI water and extracted with EtOAc. The combined organic extracts were washed with brine (x5), dried over MgSO4, filtered and concentrated under reduced pressure. The crude product was purified by FCC (DCM/MeOH 1:0 to 97:3) to afford I-123(3.7 g, yield 68%) as a pale orange solid and I-124 (880 mg, yield 16%) as a white solid. I-123: LCMS Rt = 0.68 min, 100% (UV), m/z (ES+) = 168.1; m/z (ES-) = -, Method 3.1H NMR (400 MHz, CHLOROFORM-d) δ ppm 3.93 (s, 3 H), 7.29 (d, J=0.9 Hz, 1 H), 8.00 - 8.06 (m, 2 H). I-124: LCMS Rt = 0.58 min, 82% (UV), m/z (ES+) = 168.1; m/z (ES-) = -, Method 3.1H NMR (400 MHz, CHLOROFORM-d) δ ppm 3.91 (s, 3 H), 7.29 (d, J=0.9 Hz, 1 H), 7.71 (d, J=8.4 Hz, 1 H), 8.08 (s, 1 H). Synthesis of N-(3-methyl-3H-imidazo[4,5-b]pyridin-5-yl)-1,1-diphenylmetha nimine (I- 125) 00 mg, 2.98 mmol), Pd(OAc) 2 [3375-31-3] (33.5 mg, 0.15 mmol), XantPhos [161265-03-8] (172.6 mg, 0.3 mmol), benzophenone imine [1013-88-3] (648.8 mg, 3.58 mmol) and cesium carbonate [534-17-8] (1.94 g, 5.97 mmol) in a pressure tube under nitrogen and the mixture was heated at 100 °C for 3 hours. The reaction mixture was allowed to cool to rt, the solids were filtered and the filtercace was washed with EtOAc. The filtrate was concentrated in vacuo and the crude product purified by FCC (Hept/EtOAc 1:0 to 0:1) to yield I-125 (730 mg, yield 78%) as a grey solid. LCMS Rt = 0.94 min, 100% (UV), m/z (ES+) = 313.2; m/z (ES-) = -, Method 5. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Intermediate Product Synthesis of tert-butyl (RS)-cis-3-(((benzyloxy)carbonyl)amino)-4-methylpiperidine-1 - carboxylate (I-129) ed dropwise to a stirred suspension of tert-butyl 3-amino-4-methylpiperidine-1-carboxylate hydrochloride [1312810-20-0] (1.3 g, 4.92 mmol) and K 2 CO 3 [584-08-7] (2.72 g, 20 mmol) in a mixture of EtOAc (2.5 mL) and DI water (2.5 mL). The mixture was stirred at rt for 2 hours. It was diluted with DI water and extracted with EtOAc. The combined organic extracts were dried over MgSO 4 , filtered and concentrated in vacuo. The crude product was purified by FCC (Hept/EtOAc 1:0 to 4:1) to yield I-129 (1.71 g, yield 93%) as a colourless oil. LCMS Rt = 1.30 min, 93% (UV), m/z (ES+) = 249.2 (M – Boc); m/z (ES-) = -, Method 6.1H NMR (300 MHz, CHKOROFORM-d) δ ppm 0.94 (d, J=6.8 Hz, 3 H), 1.26 (t, J=10.7 Hz, 2 H), 1.43 (d, J=12.4 Hz, 9 H), 1.79 (s, 1 H), 2.69 (d, J=12.8 Hz, 1 H), 2.87 (dd, J=13.4, 2.1 Hz, 1 H), 3.81 (s, 1 H), 4.11 (d, J=13.2 Hz, 2 H), 4.85 (d, J=9.2 Hz, 1 H), 5.11 (s, 2 H), 7.30 - 7.40 (m, 5 H). Synthesis of benzyl (RS)-cis-(4-methylpiperidin-3-yl)carbamate (I-130) 29 (1.71 g, 4.91 mmol) in DCM (41.5 mL). The reaction mixture was stirred at rt for 1 hour and the volatiles were concentrated in vacuo to yield I-130 (1.89 g, yield 93%) as a colourless oil. The crude product was used in the next step without further purification. LCMS Rt = 0.72 min, 93% (UV), m/z (ES+) = 249.2; m/z (ES-) = -, Method 6. Synthesis of benzyl (RS)-cis-(1-ethyl-4-methylpiperidin-3-yl)carbamate (I-131) DIPEA [7087-68-5] (10.9 mL, 62.6 mmol) were added to a solution of I-130 (1.89 g, 5.22 mmol) in MeCN (13.6 mL). The mixture was stirred and heated at 85 ºC for 16 hours. The reaction mixture was diluted with DI water and extracted with EtOAc. The combined organic extracts were dried over MgSO 4 , filtered and concentrated under reduced pressure. The crude product was purified by FCC (Hept/EtOAc 10 to 4:1) to yield I-131 (1.19 g, yield 78%) as a brown oil. LCMS Rt = 1.02 min, No UV, m/z (ES+) = 277.2; m/z (ES-) = -, Method 6. Synthesis of (RS)-cis-1-ethyl-4-methylpiperidin-3-amine (I-132) stirred solution of I-131 (1.19 g, 4.31 mmol) in MeOH (20 mL) and the reaction mixture was placed under atmospheric pressure of hydrogen and stirred at rt for 16 hours. The mixture was filtered through a pad of Celite® (washing with MeOH) and the filtrate was concentrated under vacuum to yield I-132 (550 mg, yield 87%) as a yellow solid. LCMS Rt = 0.27 min, No UV, m/z (ES+) = 143.2; m/z (ES-) = -, Method 7.1H NMR (400 MHz, DMSO-d 6 ) δ ppm 0.88 (t, J=9.8 Hz, 3 H), 0.98 (q, J=7.1 Hz, 3 H), 1.38 - 1.53 (m, 2 H), 1.61 - 1.77 (m, 1 H), 1.90 - 2.03 (m, 1 H), 2.11 (d, J=10.3 Hz, 1 H), 2.25 - 2.43 (m, 2 H), 2.67 - 2.84 (m, 2 H), 3.08 (d, J=2.6 Hz, 1 H), 6.49 (s, 2 H). Synthesis of 7-bromocinnolin-4(1H)-one (I-133) water (6 mL) at 0 ºC was added to a solution of 1-(2-amino-4-bromophenyl)ethanone [123858-51-5] (3 g, 14 mmol) in a 37%wt aqueous solution of HCl (7.6 mL) at 0 ºC. The reaction mixture was stirred at 0 ºC for 2 hours and rt for 16 hours. The volatiles were concentrated in vacuo and a saturated aqueous solution of AcOK [127-08-2] (6 mL) was added. The formed precipitate was collected by filtration and washed with DI water to yield I-133(2.73 g, yield 80%) as a brown solid. LCMS Rt = 0.51 min, 92% (UV), m/z (ES+) = 225.0; m/z (ES-) = -, Method 6.1H NMR (300 MHz, DMSO-d 6 ) δ ppm 7.48 (dd, J=8.7, 1.7 Hz, 1 H), 7.69 (d, J=2.5 Hz, 2 H), 7.87 (d, J=8.7 Hz, 1 H). NH was not observed. Synthesis of 7-bromo-3-iodocinnolin-4(1H)-one (I-134) (1.08 mL, 13.34 mmol) and PIDA [3240-34-4] (1.95 g, 6.07 mmol) were added to a stirred solution of I-133 (2.73 g, 12.13 mmol) in DCM (70 mL) at rt. The mixture was stirred at rt for 16 hours. The formed precipitate was collected by filtration and washed with DI water to yield I-134 (3.3 g, yield 74%) as a pale orange solid. LCMS Rt = 0.71 min, 95% (UV), m/z (ES+) = 350.8; m/z (ES-) = -, Method 6.1H NMR (300 MHz, DMSO-d 6 ) δ ppm 7.62 (dd, J=8.7, 1.7 Hz, 1 H), 7.77 (d, J=1.6 Hz, 1 H), 7.96 (d, J=8.7 Hz, 1 H). NH was not observed. Synthesis of 7-bromo-3-iodo-1-isopropylcinnolin-4(1H)-one (I-135) mmol) was added to a stirred suspension of I-134 (3.3 g, 9.4 mmol) in anhydrous DMF (44.8 mL) at 0 ºC.2-Iodopropane [75-30-9] (1.73 mL, 15.05 mmol) was then added and the reaction mixture was stirred and heated at 75ºC for 6 hours. It was diluted with a saturated aqueous solution of NaHCO3 and extracted with EtOAc. The combined organic extracts were dried over MgSO4, filtered and the solvents concentrated in vacuo. The crude product was purified by FCC (Hept/EtOAc 1:0 to 4:1) to yield I-135(1.5 g, yield 39%) as a yellow solid. LCMS Rt = 1.01 min, 95% (UV), m/z (ES+) = 392.9; m/z (ES-) = -, Method 6.1H NMR (300 MHz, DMSO-d6) δ ppm 1.41 (d, J=6.3 Hz, 6 H), 5.25 (spt, J=6.3 Hz, 1 H), 7.69 (dd, J=8.7, 1.5 Hz, 1 H), 8.04 (d, J=8.7 Hz, 1 H), 8.29 (d, J=1.3 Hz, 1 H). Synthesis of tert-butyl (7-bromo-1-isopropyl-4-oxo-1,4-dihydrocinnolin-3-yl)carbamat e (I- 136) 64-51-3] (99.6 mg, 0.11 mmol) were added to a stirred solution/suspension of I-135 (1.5 g, 3.63 mmol), tert-butyl carbamate [4248-19-5] (467.2 mg, 3.99 mmol) and cesium carbonate [534-17-8] (1.77 g, 5.44 mmol) in anhydrous 1,4-dioxane (50 mL) at rt. The mixture was sparged with nitrogen for 5 min and then stirred and heated at 60 ºC for 9 hours. It was diluted with a saturated aqueous solution of NaHCO 3 and extracted with EtOAc. The combined organic extracts were dried over MgSO4, filtered and the solvents evaporated in vacuo. The crude product was purified by FCC (Hept/EtOAc 1:0 to 4:1) to yield I-136 (768 mg, yield 42%) as a white solid. LCMS Rt = 1.07 min, 75% (UV), m/z (ES+) = 326.0 (M - tBu); m/z (ES-) = -, Method 6. Synthesis of 3-amino-7-bromo-1-isopropylcinnolin-4(1H)-one (I-137) 136 (768 mg, 2 mmol) and the mixture was stirred at rt for 3 hours. It was diluted with a saturated aqueous solution of NaHCO 3 and extracted with EtOAc. The combined organic layers were dried over MgSO4, filtered and the solvents concentrated under reduced pressure. The crude product was purified by FCC (Hept/EtOAc 1:0 to 4:1) to yield I-137 (325 mg, yield 57%) as a yellow solid. LCMS Rt = 0.82 min, 99% (UV), m/z (ES+) = 282.0; m/z (ES-) = -, Method 6.1H NMR (300 MHz, DMSO-d6) δ ppm 1.37 (d, J=6.4 Hz, 6 H), 5.11 (spt, J=6.3 Hz, 1 H), 6.01 (s, 2 H), 7.28 (dd, J=8.8, 1.4 Hz, 1 H), 7.98 (d, J=8.8 Hz, 1 H), 8.07 (s, 1 H). Synthesis of 7-bromo-3-isocyanato-1-isopropylcinnolin-4(1H)-one (I-138) sgene [32315-10-9] (53 mg, 0.18 mmol) in anhydrous THF (1 mL) were added to a solution of I-137 (100 mg, 0.35 mmol) in anhydrous THF (2 mL). The mixture was stirred at rt for 3 hours. The reaction was filtered and the filtrate was concentrated to give crude I-138 (105 mg, yield 77%) that was used without further purification in the next reaction. LCMS Rt = 1.03 min, 80% (UV), m/z (ES+) = 308.0; m/z (ES-) = -, Method 6. Synthesis of 3-methyl-[1,2,4]triazolo[4,3-b]pyridazin-6-amine (I-139) ne [1150888-24-6] (1.25 g, 7.37 mmol) in anhydrous THF (60 mL) was stirred at rt and degassed by sparging nitrogen for 5 min. Bis(tri-tert-butylphosphine)palladium(0) [53199-31-8] (565.1 mg, 1.11 mmol) was added and the reaction mixture was degassed again for 5 min. Then, a commercial 2M solution of MeZnCl in THF [5158-46-3] (7.37 mL, 14.74 mmol) was added dropwise and the reaction mixture was stirred under nitrogen at 90 °C for 8 hours. It was then allowed to cool to rt, quenched by addition of with a saturated aqueous solution of NH4Cl (10 mL), stirred for 10 min and the pH finally adjusted to pH > 8 with a saturated aqueous solution of NaHCO3. The volatiles were removed under vacuum and the resulting mixture stirred in MeOH (50 mL) overnight. The solids were filtered and the filtrate was purified by preparative RP-HPLC (Stationary phase: RP XBridge Prep C18 OBD-10µm,50x250mm, Mobile phase: 0.25% NH 4 HCO 3 solution in water, CH 3 CN) to yield I-139 (402 mg, yield 37%) as a white solid. LCMS Rt = 0.31 min, 100% (UV), m/z (ES+) = 150.1; m/z (ES-) = 148.2, Method 4. PREPARATION OF FINAL COMPOUNDS Synthesis of 2-(7-bromo-1-isopropyl-4-oxo-1,4-dihydrocinnolin-3-yl)-N-(1- methyl-2-oxo- 2,3-dihydro-1H-benzo[d]imidazol-5-yl)acetamide, F-1 3P in EtOAc [68957-94-8] (0.18 mL, 1.07 g/mL, 0.31 mmol) were added to a suspension of 2-(7- bromo-1-isopropyl-4-oxo-1,4-dihydrocinnolin-3-yl)acetic acid I-55 (40 mg, 0.12 mmol) and 5-amino-1-methyl-2,3-dihydro-1H-1,3-benzodiazol-2-one [54732-89-7] (27.1 mg, 0.17 mmol) in DCM (1.3 mL). The resulting brownish solution was stirred at rt for 2h. The reaction mixture was concentrated under a stream of nitrogen until a solid was obtained, this solid was dissolved in the minimum amount of hot acetonitrile (ca. 17 mL) and the hot solution was allowed to cool to rt with vigorous stirring. It was then cooled to 5 °C for 1h, the solid filtered off and dried at 55 °C under vacuum for 20h to afford 2-(7-bromo-1- isopropyl-4-oxo-1,4-dihydrocinnolin-3-yl)-N-(1-methyl-2-oxo- 2,3-dihydro-1H- benzo[d]imidazol-5-yl)acetamide F-1 (33 mg, yield 57%) as a light brown/greenish solid. LCMS Rt = 1.65 min, 100% (UV), m/z (ES+) = 470.2 / 472.2; m/z (ES-) = 468.2 / 470.2. Method 1. 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 1.41 (d, J=6.4 Hz, 6 H), 3.24 (s, 3 H), 3.76 (s, 2 H), 5.26 (spt, J=6.4 Hz, 1 H), 6.99 (d, J=8.4 Hz, 1 H), 7.10 (dd, J=8.5, 1.9 Hz, 1 H), 7.48 (d, J=1.8 Hz, 1 H), 7.61 (dd, J=8.6, 1.5 Hz, 1 H), 8.05 (d, J=8.6 Hz, 1 H), 8.27 (d, J=1.5 Hz, 1 H), 10.03 (s, 1 H), 10.75 (s, 1 H). Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. DCM and DMF may be used interchangeably as solvent. Reagent Intermediate Final compound/product N

In the table above, compounds may be separated or isolated using usual separation techniques. More specific techniques may also be used. For example, F-12 and F-13, which are obtained from F-11, the following method may be used: a purification/separation (of F- 11) was performed via preparative SFC (Stationary phase: Chiralcel Diacel IH 20 x 250 mm, Mobile phase: CO 2 , EtOH + 0.4 iPrNH 2 ), yielding F-12 and F-13 as white solids. The skilled person can also identify other methods/techniques. Synthesis of 2-(7-cyclopropyl-1-isopropyl-4-oxo-1,4-dihydrocinnolin-3-yl) -N-(pyrimidin- 4-yl)acetamide F-79 ly, over 2 min, to a suspension of 2-(7-bromo-1-isopropyl-4-oxo-1,4-dihydrocinnolin-3-yl)-N- (pyrimidin-4-yl)acetamide F-2 (43 mg, 0.107 mmol) and bis(tri-tert- butylphosphine)palladium(0) (9.8 mg, 0.0192 mmol) in THF (1 mL) and under nitrogen. The reaction was stirred at rt for 3 h. The solvent was removed in vacuo and the crude product was purified by flash column chromatography (Hept/EtOAc 1:0 to 0:1) to afford 2-(7- cyclopropyl-1-isopropyl-4-oxo-1,4-dihydrocinnolin-3-yl)-N-(p yrimidin-4-yl)acetamide F- 79 (26 mg, yield 67%) as a white solid. 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 0.88 - 0.96 (m, 2 H), 1.07 - 1.17 (m, 2 H), 1.38 - 1.42 (m, 6 H), 2.13 - 2.23 (m, 1 H), 3.87 (s, 2 H), 5.28 (dt, J=12.6, 6.2 Hz, 1 H), 7.12 (d, J=8.1 Hz, 1 H), 7.65 (s, 1 H), 7.96 - 8.05 (m, 2 H), 8.63 (d, J=5.7 Hz, 1 H), 8.88 (s, 1 H), 11.09 (s, 1 H). LCMS Rt = 1.76 min, 100% (UV), m/z (ES+) = 364.2; m/z (ES-) = 362.2. Method 1. Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Substrate Final compound N Substrate Final compound N Synthesis of 2-(1-isopropyl-4-oxo-7-(trifluoromethyl)-1,4-dihydrocinnolin -3-yl)-N-(6- (trifluoromethyl)pyridazine-3-yl)acetamide, F-84 To a stirred suspension of 2-(1-isopropyl-4-oxo-7-(trifluoromethyl)-1,4-dihydrocinnolin -3- yl)acetic acid I-58 (70 mg, 0.22 mmol) and 6-(trifluoromethyl)pyridazine-3-amine [935777- 24-5] (61.8 mg, 0.38 mmol) in MeCN (1.74 mL) was added 1-methylimidazole (89 µL, 1.03 g/mL, 1.11 mmol), followed by addition of solid TCFH [207915-99-9] (125 mg, 0.45 mmol). The mixture was stirred vigorously at rt for 5 hours. After 5h stirring at rt, the reaction mixture was stored in the freezer for 16h and then purified twice by flash column chromatography (Hept/EtOAc 93:7 to 3:2) to afford 2-(l-isopropyl-4-oxo-7- (trifluoromethyl)-l,4-dihydrocinnolin-3-yl)-A-(6-(trifluorom ethyl)pyridazine-3- yl)acetamide F-84 as a dark pink solid (14.8 mg, ca. 94% purity, yield 14%).

LCMS Rt = 2.04-2.06 min, 100% (UV), m/z (ES+) = 460.2; m/z (ES-) = 458.2. Method 1. 'H NMR (400 MHz, CHLOROFORM- ) 6 ppm 1.62 (d, J=6.5 Hz, 6 H), 4.17 (s, 2 H), 5.08 (spt, J=6.5 Hz, 1 H), 7.66 (dd, J=8.5, 1.1 Hz, 1 H), 7.76 (d, J=9.2 Hz, 1 H), 7.90 (s, 1 H), 8.58 (d, J=8.5 Hz, 1 H), 8.64 (d, J=9.2 Hz, 1 H), 10.71 (br s, 1 H).

Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Reagent Intermediate Final compound Synthesis of N-(6-bromoimidazo[1,2-a]pyrazin-2-yl)-2-(1-isopropyl-4-oxo-7 - (trifluoromethyl)-1,4-dihydrocinnolin-3-yl)acetamide, F-116 A (0.23 mmol) was added to a stock solution of I-58 in DMF (0.15 mmol) at rt. The solution was stirred at rt and a stock solution of TCFH [207915-99-9] (0.3mmol) in DMF and 1- methylimidazole [616-47-7] (60 µL, 0.75 mmol) were added sequentially (final overall volume of mixture ~ 1.5 mL). The resulting mixture was stirred at rt for 24 hours. It was diluted with DMSO (0.75 mL) and concentrated in vacuo to removed all the DMF. The resulting solution was diluted with a 1:1 mixture of MeCN/MeOH (2 mL) and filtered (rinsing the filter with 0.5 mL of a 1:1 mixture of MeCN/MeOH) and the filtrate was purified by preparative RP-HPLC (Stationary phase: RP XBridge Prep C18 OBD-10µm, 50x250mm, Mobile phase: 0.25% NH4HCO3 solution in water, CH3CN or MeOH) to yield F-116 (14 mg, yield 18%) as an off-white solid. LCMS Rt = 1.03 min, 100% (UV), m/z (ES+) = 509.3 / 511.3; m/z (ES-) = 507.3 / 509.3.

Method 4..

Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Reagent Final compound Synthesis of N-(6-fluoro-2-oxo-1,2-dihydropyridin-4-yl)-2-(1-isopropyl-4- oxo-7- (trifluoromethyl)-1,4-dihydrocinnolin-3-yl)acetamide, F-130 as added dropwise to a suspension of ethyl 2-(1-isopropyl-4-oxo-7-(trifluoromethyl)-1,4- dihydrocinnolin-3-yl)acetate I-77 (120 mg, 0.35 mmol) and 4-amino-6-fluoro-1H-pyridin- 2-one [105252-99-1] (53.9 mg, 0.42 mmol) in dry toluene (2.7 mL) under nitrogen. The resulting mixture was stirred at rt for 2h. It was then quenched by addition of DI water (2 mL) and a saturated aqueous solution of NH4Cl (3 mL) and extracted with DCM (2 x 3 mL). The combined organic layers were dried on MgSO 4 , filtered and concentrated in vacuo. The crude product was purified by preparative RP-chromatography (Stationary phase: RP Xbridge Prep C18 OBD-10µm, 50x250mm, Mobile phase: 0.25% NH 4 HCO 3 solution in water, CH3CN) to give F-130 (55 mg, yield 37%) as a white solid. m.p.242.5 °C (Method A). LCMS Rt = 1.55 min, 100% (UV), m/z (ES+) = 425.2; m/z (ES-) = 423.1. Method 1. 1H NMR (400 MHz, DMSO-d 6 ) δ ppm 1.43 (d, J=6.2 Hz, 6 H), 3.84 (s, 2 H), 5.42 (spt, J=6.4 Hz, 1 H), 6.75 (s, 1 H), 6.76 (s, 1 H), 7.75 (dd, J=8.6, 1.1 Hz, 1 H), 8.34 (d, J=8.4 Hz, 1 H), 8.36 (s, 1 H), 10.68 (s, 1 H), 11.13 (br s, 1 H). Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Reagent Intermediate Final compound Synthesis of N-([1,2,4]triazolo[4,3-b]pyridazine-6-yl)-2-(7-cyclopropyl-1 -isopropyl-4-oxo- 1,4-dihydrocinnolin-3-yl)acetamide, F-133 ed dropwise to a solution of [1,2,4]triazolo[4,3-b]pyridazine-6-amine [19195-46-1] (161.5 mg, 1.14 mmol) in anhydrous DMF (3.2 mL) at 0 °C under nitrogen. The resulting solution stirred at 0 °C for 2 minutes, whereupon a fine suspension had formed. Then, a solution of I-81 (255 mg, 0.81 mmol) in anhydrous THF (2.6 mL) was added dropwise at 0 °C over 2 minutes. The resulting mixture was allowed to warm to r.t. and stirred at r.t. for 7 hours. The crude mixture was quenched by addition of DI water (10 mL) then 1M aqueous HCl (2.1 mL overall, until pH mildly acidic), then a saturated solution of NaHCO3 (10 mL) was added and the resulting pale brown mixture was extracted with DCM (5 x 10 mL) and DCM/DMF 1:1 (2 x 10 mL). The combined organic extracts were dried over Na2SO4, filtered and concentrated. The obtained residue was purified by preparative RP-HPLC (Stationary phase: RP Xbridge Prep C18 OBD-10µm, 50x250mm, Mobile phase: 0.25% NH 4 HCO 3 solution in water, CH 3 CN) to give F-133 (129 mg, yield 39%) as an off-white fluffy solid. LCMS Rt = 1.61 min, 94% (UV), m/z (ES+) = 404.3; m/z (ES-) = 402.2. Method 1. 1H NMR (400 MHz, DMSO-d6) δ ppm 0.85 - 0.97 (m, 2 H), 1.05 - 1.17 (m, 2 H), 1.42 (d, J=6.4 Hz, 6 H), 2.13 - 2.26 (m, 1 H), 3.89 (s, 2 H), 5.29 (spt, J=6.3 Hz, 1 H), 7.12 (dd, J=8.6, 1.1 Hz, 1 H), 7.66 (s, 1 H), 7.98 (d, J=9.9 Hz, 1 H), 8.02 (d, J=8.6 Hz, 1 H), 8.33 (dd, J=10.1, 0.7 Hz, 1 H), 9.50 (s, 1 H), 11.28 (br s, 1 H). Additional analogs were synthesized according to the above procedure substituting the reagents as appropriate. Amine reagent Intermediate Final compound Amine reagent Intermediate Final compound , , , , -4- oxo-1,4-dihydrocinnolin-3-yl)acetamide, F-149 a mixture of I-107 (55 mg, 0.177 mmol) in dry pyridine (5 ml) under N2. The mixture was sonicated for 10 min and then stirred for 40 min at rt and a 1M solution of titanium(IV) chloride in DCM [7550-45-0] (0.71 mL, 0.709 mmol) was added dropwise at rt. The mixture was stirred for at rt fo 1 hour and then heated at 80 ºC for 24 hours. The volatiles were concentrated in vacuo and the crude material was treated with a 2M aqueous HCl solution until pH < 7. It was extracted with AcOEt (3 x 5 ml) and the combined organic extracts were concentrated in vacuo. The crude product was purified by FCC (DCM/MeOH 100/0 to 95:5) to afford F-149 (30 mg, yield 36%) as an off-white solid. LCMS Rt = 1.53 min, 100% (UV), m/z (ES+) = 427.2; m/z (ES-) = -. Method 9.. 'H NMR (400 MHz, DMSO-tL,) 1.44 (d, J=6.5 Hz, 6 H), 2.09 (t, J=19.2 Hz, 3 H), 3.86 (s, 2 H), 5.38 (spt, J=6.4 Hz, 1 H), 7.31 (dd, J=9.8, 2.0 Hz, 1 H), 7.63 (dd, J=8.6, 1.4 Hz, 1 H), 7.78 (d, J=9.7 Hz, 1 H), 8.07 (s, 1 H), 8.25 (d, J=8.6 Hz, 1 H), 9.17 - 9.32 (m, 2 H), 10.46 (s, 1 H).

Synthesis of l-([l,2,4]triazolo[4,3-a]pyridin-6-yl)-3-(l-isopropyl-4-oxo- 7-

(trifluoromethyl)-l ,4-dihydrocinnolin-3-yl)urea, F-150

A commercial IM solution of LiHMDS in THF [4039-32-1] (0.3 mL, 0.3 mmol) was added to a solution of [l,2,4]triazolo[4,3-a]pyridin-6-amine [1082448-58-5] (40.2 mg, 0.3 mmol) in anhydrous DMF (3 mL) under nitrogen at 0 °C. The mixture was stirred at rt for 1 hour. 1-138 (105 mg, 0.27 mmol) was then added and the reaction mixture was stirred at rt for 16 hours. It was diluted with a saturated aqueous solution of NH4CI and extracted with EtOAc. The combined organic extracts were dried over MgSCL, filtered and concentrated under reduced pressure. The crude product was purified by FCC (DCM to DCM/MeOH 9:1) to yield F-150 ( (4.4 mg, yield 3%) as a green gum.

LCMS Rt = 2.78 min, 04% (UV), m/z (ES+) = 442.1; m/z (ES-) = -. Method 7..

'H NMR (400 MHz, DMSO- e) 5 ppm 1.46 (d, J=6.3 Hz, 6 H), 5.19 - 5.39 (m, 1 H), 7.15 (dd, J=9.7, 1.7 Hz, 1 H), 7.53 (dd, J=8.7, 1.5 Hz, 1 H), 7.77 (d, J=9.7 Hz, 1 H), 8.11 (d, J=8.7 Hz, 1 H), 8.29 (s, 1 H), 9.10 (s, 1 H), 9.18 (s, 1 H), 9.29 (s, 1 H), 9.72 (d, J=4.1 Hz, 1 H).

Additional Characterising Data - LC-MS and melting point

LCMS: [M+H] + means the protonated mass of the free base of the compound, Rt means retention time (in minutes), method refers to the method used for LCMS.

NMR data of final compounds Example B – Pharmaceutical Compositions A compound of the invention (for instance, a compound of the examples) is brought into association with a pharmaceutically acceptable carrier, thereby providing a pharmaceutical composition comprising such active compound. A therapeutically effective amount of a compound of the invention (e.g. a compound of the examples) is intimately mixed with a pharmaceutically acceptable carrier, in a process for preparing a pharmaceutical composition. Example C - Biological Examples The activity of a compound according to the present invention can be assessed by in vitro methods. A compound the invention exhibits valuable pharmacological properties, e.g. properties susceptible to inhibit NLRP3 activity, for instance as indicated the following test, and are therefore indicated for therapy related to NLRP3 inflammasome activity. PBMC assay Peripheral venous blood was collected from healthy individuals and human peripheral blood mononuclear cells (PBMCs) were isolated from blood by Ficoll- Histopaque (Sigma-Aldrich, A0561) density gradient centrifugation. After isolation, PBMCs were stored in liquid nitrogen for later use. Upon thawing, PBMC cell viability was determined in growth medium (RPMI media supplemented with 10% fetal bovine serum, 1% Pen-Strep and 1% L-glutamine). Compounds were spotted in a 1:3 serial dilution in DMSO and diluted to the final concentration in 30 µl medium in 96 well plates (Falcon, 353072). PBMCs were added at a density of 7.5 × 104 cells per well and incubated for 30 min in a 5% CO2 incubator at 37 °C. LPS stimulation was performed by addition of 100 ng/ml LPS (final concentration, Invivogen, tlrl-smlps) for 6 hrs followed by collection of cellular supernatant and the analysis of IL-1β (µM) and TNF cytokines levels (µM) via MSD technology according to manufacturers’ guidelines (MSD, K151A0H). The IC50 values (for IL-1β) and EC50 values (TNF) were obtained on compounds of the invention/examples, and are depicted in the following table:

Example D - Further Testing

One or more compound(s) of the invention (including compounds of the final examples) is/are tested in a number of other methods to evaluate, amongst other properties, permeability, stability (including metabolic stability and blood stability) and solubility. Permeability test The in vitro passive permeability and the ability to be a transported substrate of P- glycoprotein (P-gp) is tested using MDCKcells stably transduced with MDR1 (this may be performed at a commercial organisaiton offering ADME, PK services, e.g. Cyprotex). Permeability experiments are conducted in duplicate at a single concentration (5 µM) in a transwell system with an incubation of 120 min. The apical to basolateral (AtoB) transport in the presence and absence of the P-gp inhibitor GF120918 and the basolateral to apical (BtoA) transport in the absence of the P-gp inhibitor is measured and permeation rates (Apparent Permeability) of the test compounds (P app x10-6 cm/sec) are calculated. Metabolic stability test in liver microsomes The metabolic stability of a test compound is tested (this may be performed at a commercial organisaiton offering ADME, PK services, e.g. Cyprotex) by using liver microsomes (0.5 mg/ml protein) from human and preclinical species incubated up to 60 minutes at 37oC with 1 µM test compound. The in vitro metabolic half-life (t1/2 ) is calculated using the slope of the log-linear regression from the percentage parent compound remaining versus time relationship ( ^), t1/2 = - ln(2)/ ^. The in vitro intrinsic clearance (Clint) (ml/min/mg microsomal protein) is calculated using the following formula: Where: V inc = incubation Wmic prot,inc = weight Metabolic stability test in liver hepatocytes The metabolic stability of a test compound is tested using liver hepatocytes (1 milj cells) from human and preclinical species incubated up to 120 minutes at 37oC with 1 µM test compound. The in vitro metabolic half-life (t1/2) is calculated using the slope of the log-linear regression from the percentage parent compound remaining versus time relationship ( ^), t1/2 = - ln(2)/ ^. The in vitro intrinsic clearance (Clint) (µl/min/million cells) is calculated using the following formula: Where: Vinc = incub ation volume, # cellsinc = number of cells (x106) in the incubation Solubility test The test/assay is run in triplicate and is semi-automated using the Tecan Fluent for all liquid handling with the following general steps: - 20µl of 10mM stock solution is dispensed in a 500µl 96 well plate - DMSO is evaporated (Genevac) - a stir bar and 400µl of buffer/biorelevant media is added - the solution is stirred for 72h (pH2 and pH7) or 24h (FaSSIF and FeSSIF) - the solution is filtered - the filtrate is quantified by UPLC/UV using a three-points calibration curve The LC conditions are: - Waters Acquity UPLC - Mobile phase A: 0.1% formic acid in H2O, B: 0.1% formic acid in CH3CN - Column: Waters HSS T31.8µm 2.1x50mm - Column temp.: 55°C - Inj.vol.: 2µl - Flow: 0.6ml/min - Wavelength UV: 250_350nm - Gradient : 0min: 0%B, 0.3min: 5%B, 1.8min: 95%B, 2.6min: 95%B Blood Stability assay The compound of the invention/examples is spiked at a certain concentration in plasma or blood from the agreed preclinical species; then after incubating to predetermined times and conditions (37°C, 0°C (ice) or room temperature) the concentration of the test compound in the blood or plasma matrix with LCMS/MS can then be determined.