Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOUND FOR THE INHIBITION OF THE NLRP3 INFLAMMASOME
Document Type and Number:
WIPO Patent Application WO/2023/198833
Kind Code:
A1
Abstract:
The current invention relates to a compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use in the treatment of a disease, disorder or condition responsive to the inhibition of the NLR family pyrin domain containing 3 (NLRP3) inflammasome, wherein said treatment comprises administering said compound to a subject in need of treatment, wherein said compound inhibits NLRP3 inflammasome activity, wherein said compound is a compound according to Formula I or a pharmaceutically acceptable enantiomer, salt or solvate thereof. In a last aspect the current invention relates to the use of a compound of Formula I as disclosed herein, or a pharmaceutically acceptable enantiomer, salt or solvate thereof, for the in vitro inhibition of the NLRP3 inflammasome.

Inventors:
METNA MATHILDE (BE)
WUURMAN BART (BE)
Application Number:
PCT/EP2023/059690
Publication Date:
October 19, 2023
Filing Date:
April 13, 2023
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
APAXEN SA (BE)
International Classes:
A61K31/423; A61K31/4184; A61K31/428; A61P29/00
Domestic Patent References:
WO2015155358A12015-10-15
WO2015173433A12015-11-19
WO2015155358A12015-10-15
WO2015173433A12015-11-19
Other References:
LANG TALI ET AL: "Macrophage migration inhibitory factor is required for NLRP3 inflammasome activation", vol. 9, no. 1, 8 June 2018 (2018-06-08), XP055955782, Retrieved from the Internet DOI: 10.1038/s41467-018-04581-2
SHIN MIN SUN ET AL: "Macrophage Migration Inhibitory Factor Regulates U1 Small Nuclear RNP Immune Complex-Mediated Activation of the NLRP3 Inflammasome", vol. 71, no. 1, 4 December 2018 (2018-12-04), US, pages 109 - 120, XP055955805, ISSN: 2326-5191, Retrieved from the Internet DOI: 10.1002/art.40672
PATEL NIKITA M. ET AL: "Inhibition of Macrophage Migration Inhibitory Factor Activity Attenuates Haemorrhagic Shock-Induced Multiple Organ Dysfunction in Rats", FRONTIERS IN IMMUNOLOGY, vol. 13, 6 April 2022 (2022-04-06), XP055955743, DOI: 10.3389/fimmu.2022.886421
LI TIANLONG ET AL: "Downregulation of macrophage migration inhibitory factor attenuates NLRP3 inflammasome mediated pyroptosis in sepsis-induced AKI", vol. 8, no. 1, 14 February 2022 (2022-02-14), XP055955745, Retrieved from the Internet DOI: 10.1038/s41420-022-00859-z
LIU YANYAN ET AL: "MIF inhibitor ISO-1 alleviates severe acute pancreatitis-associated acute kidney injury by suppressing the NLRP3 inflammasome signaling pathway", INTERNATIONAL IMMUNOPHARMACOLOGY, vol. 96, 1 July 2021 (2021-07-01), NL, pages 107555, XP055955749, ISSN: 1567-5769, DOI: 10.1016/j.intimp.2021.107555
FUSCO ROBERTA ET AL: "Focus on the Role of NLRP3 Inflammasome in Diseases", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 21, no. 12, 13 June 2020 (2020-06-13), pages 4223, XP093062327, DOI: 10.3390/ijms21124223
TARA ELIZABETH SCOTT ET AL: "Inflammasomes: a novel therapeutic target in pulmonary hypertension?", BRITISH JOURNAL OF PHARMACOLOGY, WILEY-BLACKWELL, UK, vol. 176, no. 12, 27 June 2018 (2018-06-27), pages 1880 - 1896, XP071129667, ISSN: 0007-1188, DOI: 10.1111/BPH.14375
LANG ET AL., NAT COMMUN, 2018, pages 9
SHIN ET AL., ARTHRITIS RHEUMATOL., 2019
TULSI PATILARUN SONISANJEEV ACHARYA: "A brief review on in vivo models for Gouty Arthritis", METABOLISM OPEN, vol. 11, 2021, Retrieved from the Internet
GALVAOIZABELA ET AL.: "Macrophage migration inhibitory factor drives neutrophil accumulation by facilitating IL-1(3 production in a murine model of acute gout", JOURNAL OF LEUKOCYTE BIOLOGY, vol. 99, no. 6, 2016, pages 1035 - 43
Attorney, Agent or Firm:
BRANTSANDPATENTS BV (BE)
Download PDF:
Claims:
CLAIMS A compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use in the treatment of a disease, disorder or condition responsive to the inhibition of the NLR family pyrin domain containing 3 (NLR.P3) inflammasome, wherein said treatment comprises administering said compound to a subject in need of treatment, wherein said compound inhibits NLR.P3 inflammasome activity, wherein said compound is a compound according to Formula I or a pharmaceutically acceptable enantiomer, salt or solvate thereof, Formula I is wherein,

X represents O, S or N-R5, wherein R5 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, alkylaryl, alkylheteroaryl, -COR6 wherein R6 is a group selected from alkyl, alkenyl, alkynyl, alkoxy, aryl and heteroaryl; preferably X represents O;

Ar represents an aryl or heteroaryl group, preferably selected from phenyl, pyridine, indole, indazole, 7-azaindole, quinoline, quinolinone, dihydroquinolinone, dihydroquinaolinone, imidazole, pyrrole, or pyrazol, benzimidazolone, benzoxazolone, benzimidazole-thione, benzotriazole, benimidazole, benzoxazinone, indolinedione, hydroxypyridinone, benzothiazolamine; optionally substituted by one or more substituents selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate, ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut-3-ene-l, 2-dione, 3- hydroxythiophen-2-yl-metanone; preferably Ar represents an optionally substituted phenyl group;

R1 -R4 are the same or different and represent a hydrogen atom or a group selected from hydroxyl, amino, halo, nitro, cyano, carboxylic acid, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, hydroxyalkyl, alkoxy, C1-C8 acyl, haloalkyl, preferably R1-R4 represent hydrogen, alkyl, cycloalkyl or haloalkyl, more preferably hydrogen, methyl or CF3. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to claim 1, said compound is a compound according to Formula la or a pharmaceutically acceptable enantiomer, salt or solvate thereof, wherein said Formula la is wherein:

X, R2 and R3 are as defined in claim 1;

Z1 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut- 3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z2 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z1 represents H, F, Cl, Br, NO2, NH2, NMe3, OH, OMe, CH3 or CF3;

Z2 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut- 3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z1 or Z3 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z2 represents H, F, Cl, Br, NO2, NH2, NMe3, OH, OMe, CH3 or CF3;

Z3 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut- 3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z2 or Z4 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z3 represents H, F, Cl, Br, NO2, NH2, NMe3, OH, OMe, CH3 or CF3;

Z4 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut- 3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z3 or Z5 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z4 represents H, F, Cl, Br, NO2, NH2, NMe3, OH, Ome, CH3 or CF3; Z5 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut- 3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z4 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z5 represents H, F, Cl, Br, NO2, NH2, NMe2, OH, Ome, CH3 or CF3. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to any of the previous claims, said compound is a compound according to Formula lb or a pharmaceutically acceptable enantiomer, salt or solvate thereof, wherein said Formula lb is wherein R2, R3, Z1, Z2, Z3, Z4 and Z5 are as defined in claim 1 and 2. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to any of the previous claims, said compound is a compound according to Formula Ic or a pharmaceutically acceptable enantiomer, salt or solvate thereof, wherein said Formula Ic is

5. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to any of the previous claims, wherein said subject in need of treatment is a mammalian subject, preferably a human subject.

6. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to any of the previous claims, wherein said disorder, disease or condition responsive to the inhibition of the NLR.P3 inflammasome is an inflammatory disease, an infectious disease, an autoimmune disease, a metabolic disorder, a cancer, a cardiovascular and/or a cerebrovascular disease.

7. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to claim 6, wherein said inflammatory disease is selected from an arthritic disease, acute pancreatitis, allergic diseases, ophtalmological diseases, renal diseases, hepatic diseases, neurodegenerative diseases, endometriosis, uveitis, fibrotic diseases, osteoarthritis, arteriosclerosis, cryopyrin-associated periodic fever syndromes (CAPS), acute respiratory distress syndrome and/or nonalcoholic steatohepatitis.

8. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to claim 7, wherein said arthritic disease is gout.

9. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to claim 6, wherein said auto-immune disease is selected from multiple sclerosis (MS), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and/or inflammatory bowel disease (IBD). ompound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to any of the previous claims, wherein said compound or pharmaceutical composition is administered to a subject in need of treatment and elicits a decreased NLR.P3 inflammasome activation in cells of said subject compared to NLR.P3 inflammasome activation in said cells of said subject prior to administration of said compound or said pharmaceutical composition to said subject. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to claim 10, wherein said decreased NLR.P3 inflammasome activation results in a decreased expression of one or more inflammatory chemokines and/or cytokines, said chemokines and/or cytokines being chosen from: IL-ip, IL-18, IL-lo, IL-37, IL-38, compared to the expression of one or more of said inflammatory chemokines and/or cytokines prior to administration of said compound or said pharmaceutical composition to said subject. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to any of the previous claims, wherein said compound is administered to a subject by one or more of the following routes of administration: intravenously, orally, rectally, vaginally, transmucosally, topically, transdermally, sublingually, subdurally, nasally, inhalation, intratracheally, intramuscularly, intra-articularly, subcutaneously, intramedullary injection, intratheca lly, intraventricularly, intraperitoneally, intranasally, intracerebroventricularly (ICV), opthalmically and intraocularly. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to any of the previous claims, wherein an effective amount of said compound ranges from about 0.001 mg to about 1000 mg. Compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use according to any of the previous claims, wherein, prior to administration of said compound or pharmaceutical composition to said subject, NLR.P3 inflammasome activation is measured in said subject. Use of a compound of Formula I, or a pharmaceutically acceptable enantiomer, salt or solvate thereof, for the in vitro inhibition of the NLR.P3 inflammasome, wherein said formula I is wherein,

X represents O, S or N-R5, wherein R5 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, alkylaryl, alkylheteroaryl, -COR6 wherein R6 is a group selected from alkyl, alkenyl, alkynyl, alkoxy, aryl and heteroaryl; preferably X represents O;

Ar represents an aryl or heteroaryl group, preferably selected from phenyl, pyridine, indole, indazole, 7-azaindole, quinoline, quinolinone, dihydroquinolinone, dihydroquinaolinone, imidazole, pyrrole, or pyrazol, benzimidazolone, benzoxazolone, benzimidazole-thione, benzotriazole, benimidazole, benzoxazinone, indolinedione, hydroxypyridinone, benzothiazolamine; optionally substituted by one or more substituents selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate, ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut-3-ene-l, 2-dione, 3- hydroxythiophen-2-yl-metanone; preferably Ar represents an optionally substituted phenyl group; R1 -R4 are the same or different and represent a hydrogen atom or a group selected from hydroxyl, amino, halo, nitro, cyano, carboxylic acid, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, hydroxyalkyl, alkoxy, C1-C8 acyl, haloalkyl, preferably R1-R4 represent hydrogen, alkyl, cycloalkyl or haloalkyl, more preferably hydrogen, methyl or

CF3.

Description:
COMPOUND FOR THE INHIBITION OF THE NLRP3 INFLAMMASOME

FIELD OF THE INVENTION

The present invention relates to a compound for the inhibition of the NLRP3 inflammasome. More specifically, the present invention relates to a compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use in the treatment of a disease, disorder or condition responsive to the inhibition of the NLR family pyrin domain containing 3 (NLRP3) inflammasome. In a last aspect the current invention relates to the use of a compound for the in vitro inhibition of the NLRP3 inflammasome.

BACKGROUND

Inflammation is a protective reaction activated in response to detrimental stimuli, such as dead cells, irritants or pathogens, by the evolutionarily conserved immune system and is regulated by the host. The inflammasomes are recognized as innate immune system sensors and receptors that manage the activation of caspase-1 and stimulate inflammation response. They have been associated with several inflammatory disorders. The NLRP3 inflammasome is the most well characterized. It is so called because NLRP3 belongs to the family of nucleotide-binding and oligomerization domain-like receptors (NLRs). Recent evidence has greatly improved our understanding of the mechanisms by which the NLRP3 inflammasome is activated. Additionally, increasing data in animal models, supported by human studies, strongly implicate the involvement of the inflammasome in the initiation or progression of disorders with a high impact on public health, such as metabolic pathologies (obesity, type 2 diabetes, atherosclerosis), cardiovascular diseases (ischemic and non-ischemic heart disease), inflammatory issues (liver diseases, inflammatory bowel diseases, gut microbiome, rheumatoid arthritis) and neurologic disorders (Parkinson's disease, Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis and other neurological disorders). Overactivation of the NLRP3 inflammasome has been described as the common denominator of these diseases, suggesting that intervention dampening its activity could be beneficial therapies for these pathologies.

Involvement of the NLRP3 inflammasome in different kinds of diseases necessitates new avenues to design drugs targeting the NLRP3 inflammasome. To date, clinical treatment of NLRP3-related diseases targets IL-ip with IL-ip antibodies or recombinant IL-ip receptor antagonist, such as canakinumab and anakinra, respectively. In addition, a few small-molecule compounds have shown antiinflammatory effects on NLRP3 inflammasome activation in vitro, including MCC950, P-hydroxybutyrate (BHB), Bay 11-7082, dimethyl sulfoxide (DMSO), and type I interferon. However, most of these inhibitors are relatively nonspecific and have low efficacy. For inhibitors targeting IL-ip, it should be noted that IL-ip secretion is not the only product of NLRP3 inflammasome activation; instead, other proinflammatory cytokines, including HMGB1 and IL-18 may participate in the pathogenesis of these diseases. Moreover, IL- 1(3 can be produced by inflammasome-independent pathways or other inflammasomes. Therefore, inhibitors targeting IL-ip may lead to unintended immunosuppressive effects besides preventing NLRP3 inflammasome activation itself. Pharmacological inhibitors specific to NLRP3 inflammasome may be the best choice for treatment of NLRP3-related diseases.

There is a need for pharmacological inhibitors specific to the NLRP3 inflammasome which are efficacious and have a good pharmacokinetic profile in terms of safety, stability, and bioavailability.

The present invention aims to resolve at least some of the problems and disadvantages mentioned above.

SUMMARY OF THE INVENTION

The present invention and embodiments thereof serve to provide a solution to one or more of above-mentioned disadvantages. To this end, the present invention relates to a compound for use according to claim 1. More specifically, the present invention relates to a compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use in the treatment of a disease, disorder or condition responsive to the inhibition of the NLR family pyrin domain containing 3 (NLRP3) inflammasome, wherein said treatment comprises administering said compound to a subject in need of treatment, wherein said compound inhibits NLRP3 inflammasome activity, wherein said compound is a compound according to Formula I or a pharmaceutically acceptable enantiomer, salt or solvate thereof, Formula I is wherein,

X represents O, S or N-R 5 , wherein R 5 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, alkylaryl, alkylheteroaryl, -COR 6 wherein R 6 is a group selected from alkyl, alkenyl, alkynyl, alkoxy, aryl and heteroaryl; preferably X represents O;

Ar represents an aryl or heteroaryl group, preferably selected from phenyl, pyridine, indole, indazole, 7-azaindole, quinoline, quinolinone, dihydroquinolinone, dihydroquinaolinone, imidazole, pyrrole, or pyrazol, benzimidazolone, benzoxazolone, benzimidazole-thione, benzotriazole, benimidazole, benzoxazinone, indolinedione, hydroxypyridinone, benzothiazolamine; optionally substituted by one or more substituents selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate, ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut-3-ene-l,2- dione, 3-hydroxythiophen-2-yl-metanone; preferably Ar represents an optionally substituted phenyl group;

R 1 -R 4 are the same or different and represent a hydrogen atom or a group selected from hydroxyl, amino, halo, nitro, cyano, carboxylic acid, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, hydroxyalkyl, alkoxy, C1-C8 acyl, haloalkyl, preferably R 1 - R 4 represent hydrogen, alkyl, cycloalkyl or haloalkyl, more preferably hydrogen, methyl or CF3.

In a second aspect, the present invention relates to the use of a compound according to claim 15. More specifically, the present invention relates to use of a compound of Formula I as described above, or a pharmaceutically acceptable enantiomer, salt or solvate thereof, for the in vitro inhibition of the NLR.P3 inflammasome. DESCRIPTION OF FIGURES

The following description of the figures of specific embodiments of the invention is merely exemplary in nature and is not intended to limit the present teachings, their application or uses.

Figure 1 shows the effect of a compound for use according to an embodiment of the current invention on NLRP3-dependent IL-ip release in primary Bone Marrow Macrophages.

Figure 2 shows the effect of 3 compounds for use according to an embodiment of the current invention on NLRP3-dependent IL-ip release and pyroptosis in human THP-1 macrophages.

Figure 3 shows the effect of a compound for use according to an embodiment of the current invention on NLRP3-dependent immune cell infiltration and IL- 1(3 release in vivo in a rodent model of gout.

DETAILED DESCRIPTION OF THE INVENTION

DEFINITIONS

Unless otherwise defined, all terms used in disclosing the invention, including technical and scientific terms, have the meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. By means of further guidance, term definitions are included to better appreciate the teaching of the present invention.

As used herein, the following terms have the following meanings:

"A", "an", and "the" as used herein refers to both singular and plural referents unless the context clearly dictates otherwise. By way of example, "a compartment" refers to one or more than one compartment.

"About" as used herein referring to a measurable value such as a parameter, an amount, a temporal duration, and the like, is meant to encompass variations of +/- 20% or less, preferably +/-10% or less, more preferably +/-5% or less, even more preferably +/-1% or less, and still more preferably +/-0.1% or less of and from the specified value, in so far such variations are appropriate to perform in the disclosed invention. However, it is to be understood that the value to which the modifier "about" refers is itself also specifically disclosed.

"Comprise", "comprising", and "comprises" and "comprised of" as used herein are synonymous with "include", "including", "includes" or "contain", "containing", "contains" and are inclusive or open-ended terms that specifies the presence of what follows e.g. component and do not exclude or preclude the presence of additional, non-recited components, features, element, members, steps, known in the art or disclosed therein.

"Alkyl" by itself or as part of another substituent refers to a hydrocarbyl radical of Formula CnHzn+i, wherein n is a number greater than or equal to 1. Generally, alkyl groups of this invention comprise from 1 to 6 carbon atoms. Alkyl groups may be linear or branched and may be substituted as indicated herein. Examples of alkyl groups are methyl, ethyl, n-propyl, i-propyl, butyl and its isomers (e.g. n-butyl, i- butyl and t-butyl); pentyl and its isomers, hexyl and its isomers.

"Alkenyl" refers to an unsaturated hydrocarbyl group, which may be linear or branched, comprising one or more carbon-carbon double bonds. Suitable alkenyl groups comprise between 2 and 6 carbon atoms, preferably between 2 and 4 carbon atoms, still more preferably between 2 and 3 carbon atoms. Examples of alkenyl groups are ethenyl, 2-propenyl, 2-butenyl, 3-butenyl, 2-pentenyl and its isomers, 2- hexenyl and its isomers, 2,4-pentadienyl and the like.

"Alkynyl" refers to a class of monovalent unsaturated hydrocarbyl groups, wherein the unsaturation arises from the presence of one or more carbon-carbon triple bonds. Alkynyl groups typically, and preferably, have the same number of carbon atoms as described above in relation to alkenyl groups. Non limiting examples of alkynyl groups are ethynyl, 2-propynyl, 2-butynyl, 3-butynyl, 2-pentynyl and its isomers, 2- hexynyl and its isomers-and the like.

"Alkoxy" refers to any O-alkyl group.

Alkylthio" refers to any S-alkyl group. "Aryl" refers to a polyunsaturated, aromatic hydrocarbyl group having a single ring (i.e. phenyl) or multiple aromatic rings fused together (e.g. naphtyl) or linked covalently, typically containing 5 to 12 atoms; preferably 6 to 10, wherein at least one ring is aromatic. The aromatic ring may optionally include one to two additional rings (either cycloalkyl, heterocyclyl or heteroaryl) fused thereto. Aryl is also intended to include the partially hydrogenated derivatives of the carbocyclic systems enumerated herein. Non-limiting examples of aryl comprise phenyl, biphenylyl, biphenylenyl, 5- or 6- tetralinyl, naphthalen-1- or -2-yl, 4-, 5-, 6 or 7-indenyl, 1- 2- , 3-, 4- or 5- acenaphtylenyl, 3-, 4- or 5-acenaphtenyl, 1- or 2-pentalenyl, 4- or 5- indanyl, 5-, 6-, 7- or 8-tetrahydronaphthyl, 1,2,3,4-tetrahydronaphthyl, 1,4- dihydronaphthyl, 1-, 2-, 3-, 4- or 5-pyrenyl.

"Heteroaryl" as used herein by itself or as part of another group refers but is not limited to 5 to 12 carbon-atom aromatic rings or ring systems containing 1 to 2 rings which are fused together or linked covalently, typically containing 5 to 6 atoms; at least one of which is aromatic in which one or more carbon atoms in one or more of these rings can be replaced by oxygen, nitrogen or sulfur atoms where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. Such rings may be fused to an aryl, cycloalkyl, heteroaryl or heterocyclyl ring. Non-limiting examples of such heteroaryl, include: pyrrolyl, furanyl, thiophenyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl, tetrazolyl, oxatriazolyl, thiatriazolyl, pyridinyl, pyrimidyl, pyrazinyl, pyridazinyl, oxazinyl, dioxinyl, thiazinyl, triazinyl, imidazo[2,l-b][l,3]thiazolyl, thieno [3,2-b] furanyl, thieno[3,2-b]thiophenyl, thieno[2,3-d][l,3]thiazolyl, thieno[2,3-d]imidazolyl, tetrazolo[l,5-a]pyridinyl, indolyl, indolizinyl, isoindolyl, benzofuranyl, isobenzofuranyl, benzothiophenyl, isobenzothiophenyl, indazolyl, benzimidazolyl, 1,3-benzoxazolyl, 1,2-benzisoxazolyl, 2,1-benzisoxazolyl, 1,3-benzothiazolyl, 1,2-benzoisothiazolyl, 2,1-benzoisothiazolyl, benzotriazolyl, 1,2,3-benzoxadiazolyl, 2,1,3-benzoxadiazolyl, 1,2,3- benzothiadiazolyl, 2,1,3-benzothiadiazolyl, thienopyridinyl, purinyl, imidazo[l,2- a]pyridinyl, 6-oxo-pyridazin-l(6H)-yl, 2-oxopyridin-l(2H)-yl, 6-oxo-pyrudazin- l(6H)-yl, 2-oxopyridin-l(2H)-yl, 1,3-benzodioxolyl, quinolinyl, isoquinolinyl, cinnolinyl, quinazolinyl, quinoxalinyl.

"Alkylaryl" refers to any group alkyl-aryl-.

Alkylheteroaryl" refers to any group alkyl-heteroaryl-. Halo" refers to fluoro, chloro, bromo, iodo.

"Haloalkyl" refers to any alkyl group substituted by one or more halo groups.

Examples of preferred haloalkyl groups are CF3, CHF2 and CH2F.

"Hydroxyalkyl" refers to any alkyl group substituted by at least one hydroxyl group.

"Cycloalkyl" as used herein is a cyclic alkyl group, that is to say, a monovalent, saturated, or unsaturated hydrocarbyl group having 1 or 2 cyclic structures. Cycloalkyl includes monocyclic or bicyclic hydrocarbyl groups. Cycloalkyl groups may comprise 3 or more carbon atoms in the ring and generally, according to this invention comprise from 3 to 10, more preferably from 3 to 8 carbon atoms, still more preferably from 3 to 6 carbon atoms. Examples of cycloalkyl groups include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl.

"Heterocyclyl" or "heterocycle" as used herein by itself or as part of another group refer to non-aromatic, fully saturated or partially unsaturated cyclic groups (for example, 3 to 7 member monocyclic, 7 to 11 member bicyclic, or containing a total of 3 to 10 ring atoms) which have at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen, oxygen and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. Any of the carbon atoms of the heterocyclic group may be substituted by oxo (for example piperidone, pyrrolidinone). The heterocyclic group may be attached at any heteroatom or carbon atom of the ring or ring system, where valence allows. The rings of multi-ring heterocycles may be fused, bridged and/or joined through one or more spiro atoms. Non limiting exemplary heterocyclic groups include oxetanyl, piperidinyl, azetidinyl, 2-imidazolinyl, pyrazolidinyl imidazolidinyl, isoxazolinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, piperidinyl, 3H-indolyl, indolinyl, isoindolinyl, 2- oxopiperazinyl, piperazinyl, homopiperazinyl, 2-pyrazolinyl, 3-pyrazolinyl, tetrahydro-2H-pyranyl, 2H-pyranyl, 4H-pyranyl, 3,4-dihydro-2H-pyranyl, 3- dioxolanyl, 1,4-dioxanyl, 2,5-dioximidazolidinyl, 2-oxopiperidinyl, 2- oxopyrrolodinyl, indolinyl, tetra hydropyranyl, tetra hydrofuranyl, tetrahydroquinolinyl, tetra hydroisoquinolin-l-yl, tetrahydroisoquinolin-2-yl, tetrahydroisoquinolin-3-yl, tetrahydroisoquinolin-4-yl, thiomorpholin-4-yl, thiomorpholin-4-ylsulf oxide, thiomorpholin-4-ylsulfone, 1,3-dioxolanyl, 1,4- oxathianyl, IH-pyrrolizinyl, tetrahydro-1, 1-dioxothiophenyl, N-formylpiperazinyl, and morpholin-4-yl.

"Amino" refers to any compound derived from ammoniac NH3 by substitution of one or more hydrogen atoms with an organic radical. Amino preferably refers to -NH2, - NHR and -NRR' wherein R and R' are preferably alkyl groups. Therefore "amino" includes monoalkylamino and dialkylamino groups.

"Amide" refers to a group -CO-NH-R or -NH-CO-R wherein R represents preferably an alkyl group, as defined above.

"Aminoacid" refers to a group -O-CO-CHR-NH2 or -NH-CHR-CO-OH wherein R represent the lateral chain of the aminoacid, preferably the lateral chain of a natural aminoacid.

"Carbamate" refers to a group -O-CO-NRR' or -NR-CO-OR' wherein R and R' represent preferably each independently alkyl groups.

"Carbamide" refers to a group -NR-CO-NR'R" wherein R, R' and R" represent preferably each independently alkyl groups.

"Carbonate" refers to a group -O-CO-OR wherein R represents preferably an alkyl group.

"Ester" refers to a group -O-CO-R or -CO-OR wherein R represents preferably an alkyl group.

"Thioester" refers to a group -S-CO-R or -CO-SR wherein R represents preferably an alkyl group.

"Phosphonate" refers to a group -0-PO(OR)2 wherein R represents H, alkyl, Na or Ca.

"Phosphonate methyloxy" refers to a group -0-CH2-O-PO(OR)2 wherein R represents H, alkyl, Na or Ca.

"Phosphonate methylamino" refers to a group -NH-CH2-O-PO(OR)2 wherein R represents H, alkyl, Na or Ca. "Sulfonamide" refers to a group -SO2-NRR' or -NR-SO2-R' wherein R and R' represent preferably each independently alkyl groups.

"Solvate" is used herein to describe a molecular complex comprising the compound of the invention and one or more pharmaceutically acceptable solvent molecules, for example, ethanol.

"Subject" refers to a warm-blooded animal, more preferably a human. Preferably, the subject is a patient, i.e. the subject is awaiting the receipt of, or is receiving medical care or is/will be the object of a medical procedure.

"Human" refers to a subject of both genders and at any stage of development (i.e. neonate, infant, juvenile, adolescent, adult).

"Treatment", "treat" and "treating" refers to therapeutic treatment, prophylactic or preventative measures and deferment of the disease onset; wherein the object is to delay, prevent or slow down (lessen) the targeted pathologic condition or disorder. Those in need of treatment include those already affected with a disease or a condition, as well as those prone to develop a disease or a condition, or those in whom a disease or a condition is to be prevented or delayed. A subject is successfully "treated" for a disease or a condition if, after receiving a therapeutic amount of a composition according to the invention, the subject shows observable and/or measurable inflammation decrease, and/or arterial pressure decrease and/or cell proliferation decrease and/or improvement in quality of life issues. The above parameters for assessing successful treatment and improvement in the disease are readily measurable by routine procedures familiar to the skilled artisan.

The term "a therapeutically effective amount" (or more simply an "effective amount") as used herein means the amount of active agent or active ingredient that is sufficient to achieve the desired therapeutic or prophylactic effect in the subject to which/whom it is administered. The term "a therapeutically effective amount" of a compound for use of the present invention refers to an amount of the compound of the present invention that will elicit the biological or medical 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) characterized 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.

The term "administration", or a variant thereof (e.g. "administering"), means providing the active agent or active ingredient, alone or as part of a pharmaceutically acceptable composition, to the subject in whom/which the condition, symptom, or disease is to be treated or prevented.

By "pharmaceutically acceptable" is meant that the ingredients of a pharmaceutical composition are compatible with each other and not deleterious to the subject thereof.

By "pharmaceutically acceptable carrier" is meant that a carrier that does not produce an adverse, allergic or other untoward reaction when administered to an animal, preferably a human. It includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. For human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologies standards.

The term "MIF" refers to macrophage migration inhibitory factor or an active fragment thereof. An active fragment of MIF may comprise a fragment of a portion of the MIF protein harboring the tautomerase enzymatic activity, or a fragment that is capable of binding to one of its receptors.

"Inhibitor of MIF" refers to any agent that attenuates, inhibits, opposes, counteracts, or decreases the biological activity of MIF. An inhibitor of MIF may be an agent that inhibits or neutralizes MIF activity; an agent that prevents the binding of MIF to CD74; an agent that prevents the interaction between CD74 and CD44; or an agent that prevents the interaction between CD74 and CD44. In one embodiment, the inhibitor of MIF is an inhibitor of MIF CD74 axis, preferably an inhibitor of MIF CD74 pathway, wherein the term MIF CD74 pathway refers to a multi-step biochemical pathway. Each step in this pathway, as in many biochemical pathways, not only passes information downstream but also receives feedback from messengers produced later in the pathway to either enhance or suppress earlier steps in the pathway. According to a specific embodiment, the inhibitor of MIF of the invention inhibits MIF binding to CD74 and CXCRs (including CXCR2, CXCR4 and/or CXCR7).

"Inhibitor of the NLRP3 inflammasome", "a compound which inhibits NLRP3 inflammasome activity", "NLRP3 signaling inhibitors" or "a compound for the inhibition of the NLRP3 inflammasome" refers to any agent that attenuates, inhibits, opposes, counteracts, or decreases the biological activity of the NLRP3 inflammasome. A NLRP3 inflammasome antagonist or inhibitor may be an agent that inhibits or neutralizes NLRP3 inflammasome activity either directly or indirectly.

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 of NLRP3 or NLRP3 inflammasome pathway to induce the production of IL-1 P 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.

The recitation of numerical ranges by endpoints includes all numbers and fractions subsumed within that range, as well as the recited endpoints.

Whereas the terms "one or more" or "at least one", such as one or more or at least one member(s) of a group of members, is clear per se, by means of further exemplification, the term encompasses inter alia a reference to any one of said members, or to any two or more of said members, such as, e.g., any >3, >4, >5, >6 or >7 etc. of said members, and up to all said members. Unless otherwise defined, all terms used in disclosing the invention, including technical and scientific terms, have the meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. By means of further guidance, definitions for the terms used in the description are included to better appreciate the teaching of the present invention. The terms or definitions used herein are provided solely to aid in the understanding of the invention.

Reference throughout this specification to "one embodiment" or "an embodiment" means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment of the present invention. Thus, appearances of the phrases "in one embodiment" or "in an embodiment" in various places throughout this specification are not necessarily all referring to the same embodiment, but may. Furthermore, the particular features, structures or characteristics may be combined in any suitable manner, as would be apparent to a person skilled in the art from this disclosure, in one or more embodiments. Furthermore, while some embodiments described herein include some but not other features included in other embodiments, combinations of features of different embodiments are meant to be within the scope of the invention, and form different embodiments, as would be understood by those in the art. For example, in the following claims, any of the claimed embodiments can be used in any combination.

DETAILED DESCRIPTION

Inflammasomes are multimeric scaffolding complexes that activate caspase-1. Several inflammasome complexes have been identified, with most incorporating at least one adaptor protein such as an AIM2-like receptor (ALR), pyrin protein, or a nucleotide-binding domain, leucine-rich-repeat-containing protein (NOD-like receptor, NLR), such as NLRP1, NLRC4, or NLRP3. This ALR or NLR engages apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC), which, in turn, recruits and activates caspase-1. Caspase-1 cleaves pro-IL-113 into bioactive IL-113.

Activation of the NLRP3 inflammasome is a two-step process. First, a priming signal, such as a TLR ligand, promotes transcription of pro-IL-ip and NLRP3 via nuclear factor-KB (NF-KB)-mediated signaling. A second signal is then required for the formation of the NLRP3 inflammasome complex. Numerous stimuli induce NLRP3 inflammasome formation, including extracellular adenosine triphosphate (ATP), pore-forming toxins such as nigericin and particulates such as uric acid crystals, silica, and alum, as well as bacterial, protozoan, and viral pathogens.

The molecular mechanisms involved in NLRP3 inflammasome assembly are incompletely understood, but studies have demonstrated roles for NIMA-related kinase-7 (NEK7) and the type III intermediate filament protein vimentin. Both proteins have been shown to interact with NLRP3, suggesting direct roles in NLRP3 inflammasome assembly and/or signaling.

As discussed above, the NLRP3 inflammasome activates caspase-1. Caspase-1 cleaves pro-IL-113 into bioactive IL-113. Furthermore, upon activation, the inflammasome also promotes an inflammatory form of cell death named pyroptosis. Varieties of inflammasome formation like NLRP1 (nod-like receptor protein 1), NLRP3 (nod-like receptor protein 3), AIM-2 (absent in melanoma 2), NLRC4 (nod-like receptor family CARD domain containing 4) cause cell pyroptosis in inflammatory diseases and cancers. Pyroptosis activators like LPS, ATP (Adenosine Triphosphate) oxidative stress, and potassium outflow lead to disassembly of TGN (trans-Golgi network), followed by PtdIns4P with negative charge recruiting NLRP3, ASC (apoptosis-associated speck like protein containing a CARD) and caspasel (cysteineaspartic acid protease 1) to dTGN (dispersed TGN) and finishing the assembly of inflammasome. Cells exposed to LPS, ATP or nigericin appear partial swelling and integrality of the membrane is broken up. In this process, GSDMD (gasdermin D) is cleaved by caspase-1 into N and C- terminal fragments, the former of which punches holes on the membrane. This process matures IL-113 (interleukin-113) and IL-18 (interleukin-18) and release them extracellular.

Although IL-1 family cytokines have important functions in protective immunity to various pathogens, dysregulation of these cytokines is also associated with pathology in a number of diseases.

As such, the involvement of these cytokines and the NLRP3 inflammasome (which is important in the production of these cytokines) in different kinds of diseases necessitates new avenues to design drugs targeting the NLRP3 inflammasome. To date, clinical treatment of NLRP3-related diseases targets I L- 1 [3 with I L- 1 [3 antibodies or recombinant IL-ip receptor antagonist, such as canakinumab and anakinra, respectively. In addition, a few small-molecule compounds have shown antiinflammatory effects on NLRP3 inflammasome activation in vitro, including MCC950, [3-hydroxybutyrate (BHB), Bay 11-7082, dimethyl sulfoxide (DMSO), and type I interferon. However, most of these inhibitors are relatively nonspecific and have low efficacy. For inhibitors targeting IL-ip, it should be noted that IL-ip secretion is not the only product of NLRP3 inflammasome activation; instead, other proinflammatory cytokines, including HMGB1 and IL-18 may participate in the pathogenesis of these diseases. Moreover, IL- 1(3 can be produced by inflammasome-independent pathways or other inflammasomes. Therefore, inhibitors targeting IL-ip may lead to unintended immunosuppressive effects besides preventing NLRP3 inflammasome activation itself.

Pharmacological inhibitors specific to the NLR family pyrin domain containing 3 (NLRP3) inflammasome may be the best choice for treatment of NLRP3-related diseases. "NLRP3-related diseases" or "a disease, disorder or condition responsive to the inhibition of the NLRP3 inflammasome" as used herein, refer to a clearly defined pathology which is (i) mediated by NLRP3, or (ii) associated with NLRP3 activity, or (iii) characterized by activity (normal or abnormal) of NLRP3. In an embodiment, a disease, disorder or condition responsive to the inhibition of the NLRP3 inflammasome has aberrant NLRP3 signaling which contributes to the pathology, and/or symptoms, and/or progression, of said disease, disorder or condition. In the present invention, such aberrant NLRP3 signaling can for instance be caused by a dysregulated activation or assembly of the NLRP3 inflammasome and contributes to the pathology, and/or symptoms, and/or progression, of said disease, disorder or condition. Inhibitors specific to the NLRP3 inflammasome may target the NLRP3 inflammasome in various ways, for instance by directly binding to NLRP3 and inhibiting it's ATPase activity, by binding to the NACHT domain of NLRP3 to inhibit NLRP3-NLRP3 interaction and subsequent ASC oligomerization, by directly inhibiting NEK7-NLRP3 interaction or by interfering with the upstream signaling pathways of the NLRP3 inflammasome (for instance NLRP3 and pro-IL-ip expression, K + efflux, mitochondrial damage, ROS production, and chloride efflux).

Macrophage migration inhibitory factor (MIF), also known as glycosylation-inhibiting factor, L-dopachrome isomerase, or phenylpyruvate tautomerase, is a highly conserved protein with pleiotropic actions. Discovered in the mid-1960s as a T cell cytokine that inhibited macrophage migration, its biochemical natures and its biological functions remained enigmatic for a long time. It is now well known that MIF (which exhibits tautomerase and oxidoreductase enzymatic activities) plays roles in cell growth, proliferation, and survival, as well as in leukocytic integrin activation, and induction of pro-inflammatory gene expression. In addition to MIF, a very recent study has identified a functional homologue of MIF with a similar genomic structure and expression patterns: the D-dopachrome tautomerase (DDT or MIF-2).

Circulating (extracellular) MIF and/or DDT bind to and activate CD74 and chemokine receptors CXCR2 and CXCR4. MIF binding to CD74 induces inflammatory cytokine release and cell proliferation, whereas MIF binding to CXCR2/4 induces cell adhesion protein expression and immune cell recruitment to inflamed tissue. Regulation of MIF-CD74 interactions occurs at several levels. MIF is constitutively expressed with increased MIF secretion occurring early in the inflammatory response. Secreted MIF then interacts with CD74 to carry out some if its functions.

In the past few years, significant efforts have been made to develop small molecules to inhibit MIF. These are for instance discussed in Lang et al. (Nat Commun 9, 2018), Shin et al. (Arthritis Rheumatol., 2019) and in WO2015155358 and WO2015173433.

WO2015155358 and WO2015173433 in particular describe thiocarbonyl MIF inhibitors. It has now been surprisingly found that these MIF inhibitors can be used for the inhibition of the NLRP3 inflammasome.

Hence, in a first aspect, the invention relates to a compound or a pharmaceutical composition comprising a therapeutically effective amount of said compound for use in the treatment of a disease, disorder or condition responsive to the inhibition of the NLR family pyrin domain containing 3 (NLRP3) inflammasome, wherein said treatment comprises administering said compound to a subject in need of treatment, wherein said compound inhibits NLRP3 inflammasome activity, wherein said compound is a compound according to Formula I or a pharmaceutically acceptable enantiomer, salt or solvate thereof, Formula I is wherein, X represents O, S or N-R 5 , wherein R 5 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, alkylaryl, alkylheteroaryl, -COR 6 wherein R 6 is a group selected from alkyl, alkenyl, alkynyl, alkoxy, aryl and heteroaryl; preferably X represents O;

Ar represents an aryl or heteroaryl group, preferably selected from phenyl, pyridine, indole, indazole, 7-azaindole, quinoline, quinolinone, dihydroquinolinone, dihydroquinaolinone, imidazole, pyrrole, or pyrazol, benzimidazolone, benzoxazolone, benzimidazole-thione, benzotriazole, benimidazole, benzoxazinone, indolinedione, hydroxypyridinone, benzothiazolamine; optionally substituted by one or more substituents selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate, ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut-3-ene-l,2- dione, 3-hydroxythiophen-2-yl-metanone; preferably Ar represents an optionally substituted phenyl group;

R 1 -R 4 are the same or different and represent a hydrogen atom or a group selected from hydroxyl, amino, halo, nitro, cyano, carboxylic acid, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, hydroxyalkyl, alkoxy, C1-C8 acyl, haloalkyl, preferably R 1 - R 4 represent hydrogen, alkyl, cycloalkyl or haloalkyl, more preferably hydrogen, methyl or CF3.

Hence, in an aspect of the invention, the invention relates to a compound, wherein said compound is a compound according to Formula I, or subformulae thereof, as disclosed herein, or a pharmaceutically acceptable enantiomer, salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of said compound, for the inhibition of the NLR.P3 inflammasome. In an embodiment, the invention relates to a compound, wherein said compound is a compound according to Formula I, or subformulae thereof, as disclosed herein, or a pharmaceutically acceptable enantiomer, salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of said compound, for the inhibition of the NLR.P3 inflammasome, wherein said compound is an inhibitor of MIF.

In an aspect of the invention, the invention relates to a compound, wherein said compound is a compound according to Formula I, or subformulae thereof, as disclosed herein, or a pharmaceutically acceptable enantiomer, salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of said compound, for use as a medicament, in particular for inhibiting NLRP3 activity.

In an aspect of the invention, the invention relates to a compound, wherein said compound is a compound according to Formula I, or subformulae thereof, as disclosed herein, or a pharmaceutically acceptable enantiomer, salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of said compound, for use in the treatment of a disease or disorder in which the NLRP3 signaling contributes to the pathology, and/or symptoms, and/or progression, of said disease or disorder (said compound being an NLRP3 signaling inhibitor).

In another aspect, the invention provides a method of treating a disease or disorder in which the NLRP3 signaling contributes to the pathology, and/or symptoms, and/or progression, of said disease or disorder, comprising administering a therapeutically effective amount of a compound of Formula I, or subformulae thereof, as disclosed herein, or a pharmaceutically acceptable salt thereof.

In another aspect, the invention provides a method of inhibiting the NLRP3 inflammasome activity in a subject in need thereof, the method comprises administering to the subject in need thereof a therapeutically effective amount of a compound of Formula I, or subformulae thereof, as disclosed herein, or a pharmaceutically acceptable salt thereof.

As described above, "a disease, disorder or condition responsive to the inhibition of the NLRP3 inflammasome" as used herein, refers to a clearly defined pathology which is (i) mediated by NLRP3, or (ii) associated with NLRP3 activity, or (iii) characterized by activity (normal or abnormal) of NLRP3. In an embodiment, a disease, disorder or condition responsive to the inhibition of the NLRP3 inflammasome has aberrant NLRP3 signaling which contributes to the pathology, and/or symptoms, and/or progression, of said disease, disorder or condition. In the present invention, such aberrant NLRP3 signaling can for instance be caused by a dysregulated activation or assembly of the NLRP3 inflammasome and contributes to the pathology, and/or symptoms, and/or progression, of said disease, disorder or condition. In a preferred embodiment, said dysregulated activation or aberrant signaling of the NLRP3 inflammasome is confirmed in a subject prior to administration of the compound for use according to the current invention to said subject. As such, in a preferred embodiment, NLRP3 activation/signaling is measured in a subject and based upon the amount of NLR.P3 activation/signaling it is determined whether the compound for use according to the current invention is administered to the subject or not. The amount of NLR.P3 activation/signaling can be measured by any technique known in the art. In an embodiment, said amount of NLR.P3 activation/signaling is measured on body tissues or fluids (for instance blood, saliva, sputum, etc.) after they have been removed from the subject. In an embodiment, the amount of NLR.P3 activation/signaling is determined by measuring the expression of one or more inflammatory chemokines and/or cytokines. In an embodiment, said chemokines and/or cytokines are chosen from: IL-ip, IL-18, IL-lo, IL-37, IL-38. In an embodiment, the amount of NLR.P3 activation/signaling is determined by measuring the transformation of procaspase-1 to caspase-1. In an embodiment, the amount of NLR.P3 activation/signaling is determined by measuring the assembly of NLR.P3, ASC, and procaspase-1 into an inflammasome complex. In an embodiment, the amount of NLR.P3 activation/signaling is determined by measuring the degree of pyroptosis. In an embodiment, the measured amount of NLR.P3 activation/signaling is compared to a certain threshold value, said threshold value determining whether the compound for use according to the current invention is administered to the subject or not. In an embodiment, when the NLR.P3 activation/signaling is below a certain threshold value, the compound for use according to the current invention is not administered to the subject, whereas the compound for use according to the current invention is administered to the subject when the NLR.P3 activation/signaling is above a certain threshold value.

In an embodiment, the invention relates to a compound wherein said compound is a compound according to Formula I or a pharmaceutically acceptable enantiomer, salt or solvate thereof, or a pharmaceutical composition comprising a therapeutically effective amount of said compound, for the treatment of an NLR.P3 inflammasome- related disorder, disease or condition in a patient (or subject in need of treatment).

In an embodiment of said aforementioned uses, compounds of Formula I as described above, are of Formula I' and pharmaceutically acceptable enantiomers, salts or solvates thereof, wherein : Ar represents aryl or heteroaryl group, preferably selected from phenyl, pyridine, indole, indazole, 7-azaindole, quinoline, quinolinone, dihydroquinolinone, dihydroquinaolinone, imidazole, pyrrole, or pyrazol, benzimidazolone, benzoxazolone, benzimidazole-thione, benzotriazole, benimidazole, benzoxazinone, indolinedione, hydroxypyridinone, benzothiazolamine; optionally substituted by or more substituents selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate, ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut-3-ene- 1,2- dione, 3-hydroxythiophen-2-yl-metanone; preferably Ar represents an optionally substituted phenyl group;

R 1 -R 4 are the same or different and represent a hydrogen atom or a group selected from hydroxyl, amino, halo, nitro, cyano, carboxylic acid, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, hydroxyalkyl, alkoxy, C1-C8 acyl, haloalkyl, preferably R 1 -R 4 represent hydrogen, alkyl, cycloalkyl or haloalkyl, more preferably hydrogen, methyl or CF3.

According to a specific embodiment, Ar is optionally substituted and is selected from phenyl, pyridine, indole, indazole, 7-azaindole, quinoline, quinolinone, dihydroquinolinone, dihydroquinaolinone, imidazole, pyrrole, or pyrazol, benzimidazolone, benzoxazolone, benzimidazole-thione, benzotriazole, benimidazole, benzoxazinone, indolinedione, hydroxypyridinone, benzothiazolamine. In a preferred embodiment, Ar is a phenyl group, optionally substituted. In one embodiment, when Ar is substituted, it is preferably substituted by one or more group selected from F, CI, Br, NO2, NH2, NMe2, OH, OMe, CH3 or CF3. In a specific embodiment, when Ar is substituted, it is preferably substituted by one or more groups selected from OH, hydroxyalkyl, aminoacid, carbamate, carbonate, ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, alkoxy and alkylthio.

According to a specific embodiment, Ar is a phenol group or a bio-isostere thereof, wherein preferred phenol bio-isosteres are selected from: wherein R and R' are preferably selected from hydrogen, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, alkylaryl, alkyl heteroaryl.

According to a specific embodiment, Ar is a phenol group or a prodrug thereof. Preferably, the prodrug of the phenol group is selected from aminoacid, carbamate, carbonate, ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, alkyloxy and alkylthio groups. According to a preferred embodiment, R 1 and R 4 represent hydrogen atoms. According to another preferred embodiment, R 1 , R 4 and at least one of R 2 or R 3 represent hydrogen atoms. According to a specific embodiment, R 1 , R 3 and R 4 represent hydrogen atoms and R 2 preferably represents an alkyl group, more preferably methyl. According to a specific embodiment, R 1 , R 3 and R 4 represent hydrogen atoms and R 2 preferably represents a haloalkyl group, more preferably trifluoromethyl. According to a specific embodiment, R 1 , R 3 and R 4 represent hydrogen atoms and R 2 preferably represents an alkyl group, more preferably methyl. According to another specific embodiment, R 1 , R 2 and R 4 represent hydrogen atoms and R 3 preferably represents an alkyl group, more preferably methyl. According to another specific embodiment, R 1 , R 2 and R 4 represent hydrogen atoms and R 3 preferably represents a haloalkyl group, more preferably trifluoromethyl.

In an embodiment of said aforementioned uses, said compound is a compound according to Formula la or a pharmaceutically acceptable enantiomer, salt or solvate thereof, wherein said Formula la is wherein:

X, R 2 and R 3 are as defined in Formula I;

Z 1 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4- amino-cyclobut-3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z 2 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z 1 represents H, F, Cl, Br, NO 2 , NH2, NMe 2 , OH, OMe, CH 3 or CF 3 ;

Z 2 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4- amino-cyclobut-3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z 1 or Z 3 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z 2 represents H, F, Cl, Br, NO 2 , NH 2 , NMe 2 , OH, OMe, CH 3 or CF 3 ;

Z 3 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4- amino-cyclobut-3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z 2 or Z 4 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z 3 represents H, F, Cl, Br, NO 2 , NH 2 , NMe 2 , OH, OMe, CH 3 or CF 3 ;

Z 4 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4- amino-cyclobut-3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z 3 or Z 5 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z 4 represents H, F, Cl, Br, NO 2 , NH 2 , NMe 2 , OH, Ome, CH 3 , or CF 3 ;

Z 5 represents a hydrogen atom or a group selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4- amino-cyclobut-3-ene-l, 2-dione, 3-hydroxythiophen-2-yl-metanone, or form with Z 4 an aryl ring, a heteroaryl ring, a cycloalkyl ring or a heterocyclyl, optionally substituted by one or more groups selected from oxo, halo, hydroxyl, nitro, amino, alkoxy, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl; preferably Z 5 represents H, F, Cl, Br, NO 2 , NH2, NMe 2 , OH, Ome, CH 3 or CF 3 .

In an embodiment of the aforementioned uses, said compound is a compound according to Formula lb or a pharmaceutically acceptable enantiomer, salt or solvate thereof, wherein said Formula lb is wherein R 2 , R 3 , Z 1 , Z 2 , Z 3 , Z 4 and Z 5 are as defined above.

According to a specific embodiment, in compounds of Formula lb, R 3 represents H.

According to a specific embodiment, in compounds of Formula lb, R 3 represents a hydrogen atom or a group selected from hydroxyl, amino, halo, nitro, cyano, carboxylic acid, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, hydroxyalkyl, alkoxy, C1-C8 acyl, haloalkyl.

According to a specific embodiment, in compounds of Formula lb, R 2 represents an alkyl group, preferably methyl, and R 3 represents a hydrogen atom. According to a specific embodiment, in compounds of Formula lb, R 2 represents a haloalkyl group, preferably trifluoromethyl, and R 3 represents a hydrogen atom. According to a specific embodiment, in compounds of Formula lb, R 2 and R 3 represents hydrogen atoms.

According to a specific embodiment, in compounds of Formula lb, Z 2 represents a hydroxyl group. In this embodiment, Z 1 , Z 3 , Z 4 and Z 5 preferably represent hydrogen atoms. According to another specific embodiment, in compounds of Formula lb, Z 2 represents a hydroxyl group and Z 3 represents a halogen, preferably a fluorine atom.

In this embodiment, Z 1 , Z 4 and Z 5 preferably represent hydrogen atoms.

In an embodiment of the aforementioned uses, said compound is a compound according to Formula Ic or a pharmaceutically acceptable enantiomer, salt or solvate thereof, wherein said Formula Ic is

According to an embodiment, compounds of Formula la of the invention as described above, including the proviso, are of Formula Id: and pharmaceutically acceptable enantiomers, salts or solvates thereof, wherein R 2 , R 3 , R 5 , Z 1 , Z 2 , Z 3 , Z 4 and Z 5 are as defined in Formula la.

According to a specific embodiment, in compounds of Formula Id, R 5 represents H.

According to an embodiment, compounds of Formula Id of the invention as described above, including the proviso, are of Formula Id-1 or Id-2: and pharmaceutically acceptable enantiomers, salts or solvates thereof, wherein R 5 is as defined in Formula la.

According to an embodiment, compounds of Formula la of the invention as described above, including the proviso, are of Formula le: and pharmaceutically acceptable enantiomers, salts or solvates thereof, wherein R 2 , R 3 , Z 1 , Z 2 , Z 3 , Z 4 and Z 5 are as defined in Formula la.

According to an embodiment, compounds of Formula Id of the invention as described above, including the proviso, are of Formula Ie-1 or Ie-2: and pharmaceutically acceptable enantiomers, salts or solvates thereof.

The compounds of Formula I and subformulae thereof may contain an asymmetric center and thus may exist as different stereoisomeric forms. Accordingly, the present invention includes all possible stereoisomers and includes not only racemic compounds but the individual enantiomers and their non-racemic mixtures as well. When a compound is desired as a single enantiomer, such may be obtained by stereospecific synthesis, by resolution of the final product or any convenient intermediate, or by chiral chromatographic methods as each are known in the art. Resolution of the final product, an intermediate, or a starting material may be performed by any suitable method known in the art.

The compounds of Formula I and subformulae thereof may be administered in the form of pharmaceutically acceptable salts.

The term "pharmaceutically acceptable salt" is intended to include all acceptable salts such as acetate, lactobionate, benzenesulfonate, laurate, benzoate, malate, bicarbonate, maleate, bisulfate, mandelate, bitartrate, mesylate, borate, methylbromide, bromide, methylnitrate, calcium edetate, methylsulfate, camsylate, mucate, carbonate, napsylate, chloride, nitrate, clavulanate, N-methylglucamine, citrate, ammonium salt, dihydrochloride, oleate, edetate, oxalate, edisylate, pamoate (embonate), estolate, palmitate, esylate, pantothenate, fumarate, phosphate/diphosphate, gluceptate, polygalacturonate, gluconate, salicylate, glutamate, stearate, glycollylarsanilate, sulfate, hexylresorcinate, subacetate, hydrabamine, succinate, hydrobromide, tannate, hydrochloride, tartrate, hydroxynaphthoate, teoclate, iodide, tosylate, isothionate, triethiodide, lactate, panoate, valerate, and the like which can be used as a dosage form for modifying the solubility or hydrolysis characteristics or can be used in sustained release or prodrug formulations.

Depending on the particular functionality of the compounds of Formula I and subformulae thereof, pharmaceutically acceptable salts of the compounds include those formed from cations such as sodium, potassium, aluminum, calcium, lithium, magnesium, zinc, and from bases such as ammonia, ethylenediamine, N-methyl- glutamine, lysine, arginine, ornithine, choline, N,N'-dibenzylethylene-diamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethyl-amine, diethylamine, piperazine, tris(hydroxymethyl)aminomethane, and tetramethylammonium hydroxide. These salts may be prepared by standard procedures, e.g. by reacting a free acid with a suitable organic or inorganic base. Where a basic group is present, such as amino, an acidic salt, i.e. hydrochloride, hydrobromide, acetate, palmoate, and the like, can be used as the dosage form.

Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, adipate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, cyclamate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, pyroglutamate, saccharate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate and xinofoate salts. Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine, 2- (diethylamino)ethanol, ethanolamine, morpholine, 4-(2-hydroxyethyl)morpholine and zinc salts. Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts. Preferred, pharmaceutically acceptable salts include hydrochloride/chloride, hydrobromide/bromide, bisulphate/sulphate, nitrate, citrate, and acetate.

When the compounds of Formula I and subformulae thereof contain an acidic group as well as a basic group the compounds may also form internal salts, and such compounds are within the scope of the invention. When the compounds of the invention contain a hydrogen-donating heteroatom (e.g. NH), the invention also covers salts and/or isomers formed by transfer of said hydrogen atom to a basic group or atom within the molecule.

Pharmaceutically acceptable salts of compounds of Formula I (and of subformulae thereof) may be prepared by one or more of these methods:

(i) by reacting the compound of Formula I with the desired acid;

(ii) by reacting the compound of Formula I with the desired base;

(iii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of Formula I or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid; or

(iv) by converting one salt of the compound of Formula I to another by reaction with an appropriate acid or by means of a suitable ion exchange column.

All these reactions are typically carried out in solution. The salt, may precipitate from solution and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionization in the salt may vary from completely ionized to almost non-ionized. In addition, although generally, with respect to the salts of the compounds of the invention, pharmaceutically acceptable salts are preferred, it should be noted that the invention in its broadest sense also included non- pharmaceutically acceptable salts, which may for example be used in the isolation and/or purification of the compounds of the invention. For example, salts formed with optically active acids or bases may be used to form diastereoisomeric salts that can facilitate the separation of optically active isomers of the compounds of Formula I (or of subformulae thereof) above.

Also, in the case of an alcohol group being present, pharmaceutically acceptable esters can be employed, e.g. acetate, maleate, pivaloyloxymethyl, and the like, and those esters known in the art for modifying solubility or hydrolysis characteristics for use as sustained release or prodrug formulations.

All references to compounds of Formula I and subformulae thereof include references to enantiomers, salts, solvates, polymorphs, multi-component complexes and liquid crystals thereof.

The compounds for use include compounds of Formula I as hereinbefore defined, including all polymorphs and crystal habits thereof, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) and isotopically-labeled compounds of Formula I.

The invention also generally covers all pharmaceutically acceptable predrugs and prodrugs of the compounds of Formula I and of the subformulae thereof.

The compounds of Formula I and subformulae thereof can be prepared by different ways with reactions known to a person skilled in the art. The compounds of Formula I and subformulae thereof can for instance be prepared by a process of manufacturing as previously described in WO2015155358.

The inventors surprisingly found that the compounds of Formula I and subformulae thereof are able to be used as inhibitors of the NLR.P3 inflammasome.

The proposed mechanism of action of the compounds of Formula I and subformulae thereof can for instance be through inhibition of intracellular assembly of the NLPR.3 inflammasome. In an embodiment, inhibition of the NLPR.3 inflammasome is achieved with nanomolar potency.

As described above, the invention relates to the use of compounds of Formula I (and of the subformulae thereof) or pharmaceutically acceptable enantiomers, salts and solvates thereof for the treatment of an NLR.P3 inflammasome-related disorder, disease or condition in a patient (or subject in need of treatment). The present invention also relates to a medicament for use in the treatment of an NLR.P3 inflammasome-related disorder, disease or condition in a patient (or subject in need of treatment).

In an embodiment, said patient is a mammalian patient, preferably a human patient.

In an embodiment, said disorder, disease or condition is an inflammatory disease, an infectious disease, an autoimmune disease, a metabolic disorder, a cancer, a cardiovascular and/or a cerebrovascular disease characterized by an excessive activity of the NR.LP3 inflammasome.

Examples of NRLP3-associated inflammatory diseases include but are not limited to: acute inflammation, chronic inflammation, granulomatous inflammation, fibrinous inflammation, purulent inflammation, serous inflammation, ulcerative inflammation, systemic inflammation, sepsis, acne vulgaris, asthma and other allergic diseases, chronic prostatitis, glomerulonephritis and other renal diseases, pelvic inflammatory disease, reperfusion injury, sarcoidosis, transplant rejection, neuropathic pain, fibromyalgia, colitis, inflammatory bowel disease, Crohn's disease, ulcerative colitis, Castleman's disease, ankylosing spondylitis, hepatitis and other hepatic diseases, otitis, experimental allergic neuritis, Parkinson's disease, Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, traumatic brain injury and other neurodegenerative or neurological disorders, organ transplant rejection, shock, spondylitis, diabetes mellitus type 2, sarcoidosis, meningitis, silicosis, acute respiratory distress syndrome, wet and dry age-related macular degeneration and other ophtalmological diseases, fibrotic diseases, restenosis, interstitial cystitis, cerebral malaria, meningitis, interstitial cystitis, gout, traumatic arthritis, rubella arthritis, acute synovitis, silicosis, muscle degeneration, diabetic retinopathy, macular degeneration, rhinovirus infection, peroral disease, such as gingivitis and periodontitis, eczema, contact dermatitis, psoriasis, and conjunctivitis.

In an embodiment, said inflammatory disease is selected from an arthritic disease, acute pancreatitis, allergic diseases, ophthalmological diseases, renal diseases, hepatic diseases, neurodegenerative diseases, endometriosis, uveitis, fibrotic diseases, arteriosclerosis, cryopyrin-associated periodic fever syndromes (CAPS), acute respiratory distress syndrome and/or nonalcoholic steatohepatitis. Arthritis is defined as "painful inflammation and stiffness of the joints." Arthritis can be broadly classified into two categories, inflammatory arthritis and noninflammatory arthritis.

Inflammatory arthritis is usually associated with the classic symptoms of inflammation - dolor (pain), rubor (erythema), calor (warmth), tumor (swelling), and functio laesa (loss of function), although all the features may not always be present. Inflammatory arthritis can be due to several etiologies, including infectious and non- infectious, and may or may not be associated with systemic features of the underlying condition causing inflammatory arthritis. If left untreated, inflammatory arthritis invariably leads to joint damage and deformities.

Gout is such an inflammatory arthritis characterized by abrupt self-limiting attacks of inflammation caused by precipitation of monosodium urate crystals (MSU) in the joint. Recent studies suggest that orchestration of the MSU-induced inflammatory response is dependent on the proinflammatory cytokine IL-ip. This IL-l-dependent innate inflammatory phenotype, is now understood to rely on the formation of the macromolecular NLRP3 inflammasome complex in response to the MSU 'danger signal'.

As such, in an embodiment of aforementioned uses, said arthritic disease is gout.

Examples of infectious diseases include but are not limited to: bacterial infections, viral infections (including respiratory viral infections, such as COVID-19), fungal infections and parasitic infections.

Examples of autoimmune diseases include but are not limited to: celiac disease, rheumatoid arthritis, juvenile rheumatoid arthritis, vasculitis, psoriasis, psoriatic arthritis, multiple sclerosis, autoimmune uveitis, ankylosing spondylitis, Pemphigus, Myasthenia gravis, Guillain-Barre syndrome, hepatitis, autoimmune glomerulonephritis, systemic lupus erythematosus, lupus nephritis, diabetes mellitus type 1, Reiter's syndrome, polymyositis, graft versus host disease.

In an embodiment, said auto-immune disease is selected from multiple sclerosis (MS), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and/or inflammatory bowel disease (IBD). Examples of metabolic disorders include but are not limited to: diabetes, obesity, steroid- resistance, glucose intolerance, metabolic syndrome.

Examples of cancers include but are not limited to: angiogenesis, multiple myeloma, leukemia, lymphoma, Hodgkin's disease, cancer of the bone, mouth/pharynx, oesophagus, larynx, stomach, intestine, colon, rectum, liver, pancreas, nerve, brain, head and neck, throat, ovary, uterus, prostate, testis, bladder, kidney, breast, nonmelanoma, skin cancer, teratoma, rhabdomyosarcoma, glioma, metastatic bone disease and other forms of metastasis.

Examples of cardiovascular and/or cerebrovascular disease includes but are not limited to: atherosclerosis, arteriosclerosis, restenosis of an atherosclerotic coronary artery, acute coronary syndrome, myocardial infarction, cardiac-allograft vasculopathy, stroke, ischemic and hemorrhagic stroke, neuro trauma/closed head injury, cardiac reperfusion injury.

Examples of the NLR.P3 inflammasome-related diseases, disorders or conditions include but are not limited to: acute inflammation, chronic inflammation, granulomatous inflammation, fibrinous inflammation, purulent inflammation, serous inflammation, ulcerative inflammation, systemic inflammation, Cryopyrin-associated periodic fever syndromes (CAPS), sepsis, acne vulgaris, asthma, celiac disease, chronic prostatitis, glomerulonephritis, pelvic inflammatory disease, reperfusion injury, rheumatoid arthritis, gout, sarcoidosis, transplant rejection, vasculitis, interstitial cystitis, inflammatory myopathies, systemic sclerosis, and include dermatomyositis, polymyositis, inclusion body myositis, psoriatic arthritis, Reiter's syndrome, gout, traumatic arthritis, rubella arthritis and acute synovitis, rheumatoid spondylitis, osteoarthritis, gouty arthritis and other arthritic conditions, septic shock, endotoxic shock, gram negative sepsis, toxic shock syndrome, Alzheimer's disease, stroke, ischemic and hemorrhagic stroke, neurotrauma/closed head injury, atherosclerosis, asthma, acute respiratory distress syndrome, meningitis, silicosis, restenosis of an atherosclerotic coronary artery, acute coronary syndrome, myocardial infarction, cardiac-allograft vasculopathy, restenosis, cardiac reperfusion injury, brain and renal reperfusion injury, chronic renal failure, thrombosis, diabetic retinopathy, macular degeneration, inflammatory bowel disease, Crohn's disease, ulcerative colitis, multiple sclerosis, muscle degeneration, diabetic retinopathy, macular degeneration, rhinovirus infection, peroral disease, such as gingivitis and periodontitis, eczema, contact dermatitis, psoriasis, and conjunctivitis; angiogenesis, multiple myeloma, leukemia, lymphoma, Hodgkin's disease, cancer of the bone, mouth/pharynx, oesophagus, larynx, stomach, intestine, colon, rectum, liver, pancreas, nerve, brain, head and neck, throat, ovary, uterus, prostate, testis, bladder, kidney, breast, non- melanoma, skin cancer, teratoma, rhabdomyosarcoma, glioma, metastatic bone disease and other forms of metastasis.

In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is colorectal cancer, prostate cancer, sepsis, endometriosis, colitis, breast carcinoma, hepatocellular carcinoma, lung adenocarcinoma, melanoma, colon cancer, nasopharyngeal carcinoma, esophageal cancer, systemic inflammation (including polymicrobial sepsis, arthritis and autoimmune diabetes), asthma, viral infection, rheumatoid arthritis, inflammatory bowel disease or atherosclerosis.

In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is rheumatoid arthritis, multiple sclerosis, psoriasis, Crohn's disease, ulcerative colitis, systemic lupus erythematosus, endometriosis, sepsis, prostate cancer.

In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is rheumatoid arthritis. In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is multiple sclerosis. In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is psoriasis. In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is Crohn's disease. In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is ulcerative colitis. In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is systemic lupus erythematosus. In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is endometriosis. In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is sepsis. In an embodiment, the disorder, disease or condition related to the NLR.P3 inflammasome is prostate cancer.

According to a further feature of the present invention there is provided a method for the inhibition of the NLR.P3 inflammasome, in a patient (or subject in need of treatment), preferably a warm blooded animal, and even more preferably a human, in need of such treatment, which comprises administering to said patient an effective amount of the compound of Formula I (or subformulae thereof), or a pharmaceutically acceptable enantiomer, salt and solvate thereof. The invention further relates to a method for treatment of disorders, diseases or conditions related to the NLR.P3 inflammasome, which comprises administering to a patient or subject in need thereof a therapeutically effective amount of the compound of Formula I (or subformulae thereof) or pharmaceutically acceptable enantiomers, salts or solvates thereof.

The invention also provides for a method for delaying in a subject the onset of disorders, diseases or conditions related to the NLR.P3 inflammasome, comprising the administration of a pharmaceutically effective amount of a compound of Formula I (or subformulae thereof) or a pharmaceutically acceptable enantiomer, salt and solvate thereof to a patient in need thereof. Preferably, the patientis a warm-blooded animal, more preferably a human.

In another embodiment, the present invention is directed to the treatment of a disease associated with the NLR.P3 inflammasome, comprising administering an effective amount of a pharmaceutical composition comprising one or more compounds of Formula I (or subformulae thereof), or a pharmaceutically acceptable enantiomer, salt and solvate thereof, to a patient or subject in need thereof.

The compounds of Formula I (or subformulae thereof) or pharmaceutically acceptable enantiomers, salts and solvates thereof are therefore useful as medicaments, in particular in the treatment of disorders, diseases or conditions related to the NLR.P3 inflammasome. The invention further provides the use of a compound of Formula I or a pharmaceutically acceptable enantiomer, salt and solvate thereof for the manufacture of a medicament for treating and/or preventing disorders, diseases or conditions related to the NLR.P3 inflammasome.

The present invention also relates to a method for inhibiting inflammation, comprising administering a compound of Formula I (or subformulae thereof) or pharmaceutically acceptable enantiomers, salts and solvates thereof, thereby treating diseases related to the NLR.P3 inflammasome in a patient or subject in need thereof.

The present invention also relates to a method for inhibiting cell proliferation and/or tumor growth and/or angiogenesis, comprising administering a compound of Formula I (or subformulae thereof) or pharmaceutically acceptable enantiomers, salts and solvates thereof, thereby treating diseases related to the NLRP3 inflammasome in a patient or subject in need thereof. The present invention also relates to a method for inhibiting vasoconstriction, comprising administering a compound of Formula I (or subformulae thereof) or pharmaceutically acceptable enantiomers, salts and solvates thereof, thereby treating diseases related to the NLR.P3 inflammasome in a patient or subject in need thereof.

According to a specific embodiment, compounds for use according to the invention or pharmaceutically acceptable enantiomers, salts and solvates thereof are useful in veterinary field. In an embodiment, the patient or subject is affected with, preferably is diagnosed with a disorder, a disease or a condition related to the NLR.P3 inflammasome. In another embodiment, the patient or subject is at risk of developing a disorder, a disease or a condition related to the NLR.P3 inflammasome. In an embodiment of the invention, the patient or subject presents a non-genetic predisposition to a disorder, a disease or a condition related to the NLRP3 inflammasome. In an embodiment of the invention, the patient or subject has a genetic or familial predisposition to a disorder, a disease or a condition related to the NLR.P3 inflammasome.

The invention also provides pharmaceutical compositions comprising or consisting of a compound of Formula I (or subformulae thereof) or a pharmaceutically acceptable enantiomer, salt and solvate thereof and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant for use as described above. The invention also covers pharmaceutical compositions which contain, in addition to a compound of Formula I (or subformulae thereof), a pharmaceutically acceptable enantiomer, salt and solvate thereof as active ingredient, additional therapeutic agents and/or active ingredients for use as described above. According to an embodiment, the composition for use according to the invention may further comprise, in addition to the compound of Formula I (or subformulae thereof), at least one additional compound, including another MIF inhibitor. Another object of this invention is a medicament comprising or consisting of at least one compound of Formula I (or subformulae thereof), or a pharmaceutically acceptable enantiomer, salt and solvate thereof, as active ingredient for use as described above.

Generally, for pharmaceutical use, the compounds of Formula I (or subformulae thereof) may be formulated as a pharmaceutical preparation comprising at least one compound of Formula I (or subformulae thereof) or a pharmaceutically acceptable enantiomer, salt and solvate thereof and at least one pharmaceutically acceptable carrier, diluent, excipient and/or adjuvant, and optionally one or more further pharmaceutically active compounds.

In one embodiment, the pharmaceutical composition or the medicament for use according to the invention comprises at least one compound of Formula I (or subformulae thereof) or a pharmaceutically acceptable enantiomer, salt and solvate thereof as unique pharmaceutically active compound.

In one embodiment, a therapeutically effective amount of the composition, the pharmaceutical composition or the medicament for use according to the invention is administered or is to be administered alone, i.e. is not administered in combination with another therapeutic agent for treating a disease, or disorder or a condition.

In another embodiment, the composition, the pharmaceutical composition or the medicament for use according to the present invention is administered or is to be administered with other active agents. In an embodiment, the composition, the pharmaceutical composition or the medicament and the other active agent may be administered separately or in conjunction.

In an embodiment, the composition, the pharmaceutical composition or the medicament for use according to the invention is for curing disorders, diseases or conditions related to the NLR.P3 inflammasome.

In another embodiment, the composition, the pharmaceutical composition or the medicament for use according to the invention slows down or stops the progression, aggravation, or deterioration of one or more symptoms of disorders, diseases or conditions related to the NLR.P3 inflammasome; bringing about ameliorations of the symptoms of disorders, diseases or conditions related to the NLR.P3 inflammasome ; reducing the severity or incidence of disorders, diseases or conditions related to the NLR.P3 inflammasome.

By means of non-limiting examples, such a formulation may be in a form suitable for oral administration, for parenteral administration (such as by intravenous, intramuscular or subcutaneous injection or intravenous infusion), for topical administration (including ocular), for administration by inhalation, by a skin patch, by an implant, by a suppository, etc. Such suitable administration forms - which may be solid, semi-solid or liquid, depending on the manner of administration - as well as methods and carriers, diluents and excipients for use in the preparation thereof, will be clear to the skilled person; reference is made to the latest edition of Remington's Pharmaceutical Sciences. Some preferred, but non-limiting examples of such preparations include tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, liposomes, nanoparticles, syrups, aerosols, ointments, cremes, lotions, soft and hard gelatin capsules, suppositories, drops, sterile injectable solutions and sterile packaged powders (which are usually reconstituted prior to use) for administration as a bolus and/or for continuous administration, which may be formulated with carriers, excipients, and diluents that are suitable per se for such formulations, such as lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, polyethylene glycol, cellulose, (sterile) water, methylcellulose, methyl- and propylhydroxybenzoates, talc, magnesium stearate, edible oils, vegetable oils and mineral oils or suitable mixtures thereof. The formulations can optionally contain other substances that are commonly used in pharmaceutical formulations, such as lubricating agents, wetting agents, emulsifying and suspending agents, dispersing agents, desinteg rants, bulking agents, fillers, preserving agents, sweetening agents, flavoring agents, flow regulators, release agents, etc.. The compositions may also be formulated so as to provide rapid, sustained or delayed release of the active compound(s) contained therein. The pharmaceutical preparations of the invention are preferably in a unit dosage form, and may be suitably packaged, for example in a box, blister, vial, bottle, sachet, ampoule or in any other suitable single-dose or multi-dose holder or container (which may be properly labeled); optionally with one or more leaflets containing product information and/or instructions for use. Depending on the condition to be prevented or treated and the route of administration, the active compound of the invention may be administered as a single daily dose, divided over one or more daily doses, or essentially continuously, e.g. using a drip infusion.

In an embodiment, a therapeutically effective amount of the compound for use according to the invention ranges from about 0.001 mg to about 1000 mg, preferably from about 0.01 mg to about 1000 mg, more preferably from about 0.1 mg to about 500 mg, more preferably from about 1 mg to about 500 mg, more preferably from about 10 mg to about 500 mg, and even more preferably from about 100 mg to about 500 mg. In one embodiment, the composition, pharmaceutical composition or medicament for use according to the invention comprises an amount of a compound of Formula I (or subformulae thereof) ranging from about 1 pM to about 1 mM, preferably from about 10 pM to about 50 pM, more preferably from about 0.1 nM to about 1 pM, even more preferably from about 0.5 nM to about 0.1 pM, and still even more preferably is of about 10 nM. In one embodiment, the therapeutically effective amount is administered once a month, once a week, twice a week, at least once a day, twice, or three times a day, most preferably once a day.

In another embodiment, the therapeutically effective amount is administered once a day on consecutive days for at least a week, at least a month, at least a year, or on as needed basis for the rest of the patient's life. In another embodiment, the therapeutically effective amount is administered once a week on consecutive weeks for at least two weeks, one month, at least a year, or on as needed basis for the rest of the patient's life. In another embodiment, the therapeutically effective amount is administered once a month on consecutive months for at least two months, a year, or on as needed basis for the rest of the patient's life.

In another embodiment of the invention, the administration dose of the composition, the pharmaceutical composition or the medicament is determined by the skilled artisan and personally adapted to each patient and/or the severity of the disease.

In an embodiment, said compound is administered to a subject by one or more of the following routes of administration: intravenously, orally, rectally, vaginally, transmucosally, topically, transdermally, sublingually, subdurally, nasally, inhalation, intratracheally, intramuscularly, intra-articularly, subcutaneously, intramedullary injection, intratheca lly, intraventricularly, intraperitoneally, intranasally, intracerebroventricularly (ICV), opthalmically and intraocularly.

The compounds of Formula I and subformulae thereof are known as MIF inhibitors. Technics to measure MIF biological activities are well known to the person skilled in the art. Examples of such assays include but are not limited to: 4- hydroxyphenylpyruvate Tautomerase Assays, Dopachrome Tautomerase Assays, MIF enzymatic activity, MIF immunoregulatory activities, MIF glucocorticoid regulating activity, MIF binding to target cells, inhibition of MIF release or synthesis, inhibition of MIF immunoreactivity with MIF-specific antibodies, alterations of MIF conformation or structural integrity as assessed by circular dichroism spectroscopy, liquid NMR- spectroscopy, X-ray crystallography, thermal stability measurement, inhibition of the pro -pro I iterative effects of MIF on quiescent, non-quiescent cells and inhibition of the associated prolonged ERK activation therein, inhibition of MIF- induced arachadonic acid release from cells, inhibition of MIF-induced fructose 2,6 bisphosphate formation in L6 myocytes, inhibition of MIF toxicity in the MIF, TNF, or LPS-challenged test animals, inhibition of the glucocorticoid counter-regulatory activity of MIF in vitro or in vivo, inhibition of the MIF-induced functional inactivation of the p53 tumor suppressor protein, inhibition of MIF-induced release of prostaglandin E2, and inhibition of morbidity or mortality in any of a number of animal models of human diseases that are characterized by the release, production and/or appearance of MIF.

"Inhibition of the NLR.P3 inflammasome" as described herein can be the result of and refer to, but is not limited to, a decreased expression of one or more of the components of the NLR.P3 inflammasome, a decreased assembly of the components of the NLR.P3 inflammasome and/or a decreased activation of the NLR.P3 inflammasome.

In an embodiment, said decreased expression of one or more of the components of the NLR.P3 inflammasome is mediated due to a decrease upstream signaling (for instance a decreased nuclear factor-KB (NF-KB)-mediated signaling; it is known that MIF may contribute to the activation of NF-KB, which also up-regulates NLRP3).

In an embodiment, said decreased assembly is mediated due to a disturbed interaction between NLR.P3 and vimentin.

Said inhibition of the NLR.P3 inflammasome can be measured by any means known in the art, such as measurement of pyroptosis, ASC speck formation, caspase-1 activation/cleavage and pro-IL-ip cleavage.

In an embodiment, said compound or pharmaceutical composition is administered to a patient and elicits a decreased NLR.P3 inflammasome activation in cells of said patient compared to NLR.P3 inflammasome activation in said cells of said patient prior to administration of said compound or said pharmaceutical composition to said patient.

In a further embodiment, said decreased NLR.P3 inflammasome activation results in a decreased expression of one or more inflammatory chemokines and/or cytokines, said chemokines and/or cytokines being chosen from: IL-ip, IL-18, IL-lo, IL-37, IL- 38, compared to the expression of one or more of said inflammatory chemokines and/or cytokines prior to administration of said compound or said pharmaceutical composition to said patient. In an embodiment, said decreased expression comprises a decrease in protein expression of said one or more inflammatory chemokines and/or cytokines of at least 5%, preferably at least 8%, more preferably at least 10%, more preferably at least 15%, more preferably at least 20%, more preferably at least 25%, more preferably at least 30%, more preferably at least 35%, more preferably at least 40%, more preferably at least 40%, more preferably at least 45%, more preferably at least 50%, more preferably at least 55%, more preferably at least 60%, more preferably at least 65%, more preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99%, such as 100% compared to the expression of one or more of said inflammatory chemokines and/or cytokines prior to administration of said compound or said pharmaceutical composition to said patient.

In a last aspect, the invention relates to the use of a compound of Formula I, or a pharmaceutically acceptable enantiomer, salt or solvate thereof, for the in vitro inhibition of the NLR.P3 inflammasome, wherein said compound is an inhibitor of MIF and wherein said formula I is wherein,

X represents O, S or N-R 5 , wherein R 5 represents a hydrogen atom or a group selected from alkyl, alkenyl, alkynyl, alkylaryl, alkylheteroaryl, -COR 6 wherein R 6 is a group selected from alkyl, alkenyl, alkynyl, alkoxy, aryl and heteroaryl; preferably X represents O;

Ar represents an aryl or heteroaryl group, preferably selected from phenyl, pyridine, indole, indazole, 7-azaindole, quinoline, quinolinone, dihydroquinolinone, dihydroquinaolinone, imidazole, pyrrole, or pyrazol, benzimidazolone, benzoxazolone, benzimidazole-thione, benzotriazole, benimidazole, benzoxazinone, indolinedione, hydroxypyridinone, benzothiazolamine; optionally substituted by one or more substituents selected from halo, hydroxyl, hydroxyalkyl, nitro, amino, amido, aminoacid, carbamate, carbamide, carbonate, ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, sulfonamide, alkoxy, alkylthio, alkyl, haloalkyl, alkenyl, alkynyl, aryl, heteroaryl, 4-amino-cyclobut-3-ene-l,2- dione, 3-hydroxythiophen-2-yl-metanone; preferably Ar represents an optionally substituted phenyl group;

R 1 -R 4 are the same or different and represent a hydrogen atom or a group selected from hydroxyl, amino, halo, nitro, cyano, carboxylic acid, alkyl, cycloalkyl, alkenyl, alkynyl, aryl, heteroaryl, hydroxyalkyl, alkoxy, C1-C8 acyl, haloalkyl, preferably R 1 - R 4 represent hydrogen, alkyl, cycloalkyl or haloalkyl, more preferably hydrogen, methyl or CF3.

As described above, said inhibition of the NLR.P3 inflammasome can be measured by any means known in the art, such as measurement of pyroptosis, ASC speck formation, caspase-1 activation/cleavage and pro-IL-ip cleavage.

In an embodiment, the amount of the compound for use according to the invention is in vitro administered at a dose ranging from about 1 nM to about 0.1 pM, more preferably from about 5 nM to about 50 nM, and still even more preferably from about 5 nM to about 20 nM, such as 10 nM.

Said inhibition of the NLR.P3 inflammasome can also be determined by a decreased expression of one or more inflammatory chemokines and/or cytokines in vitro, said chemokines and/or cytokines being chosen from: IL-ip, IL-18, IL-lo, IL-37, IL-38, compared to the in vitro expression of one or more of said inflammatory chemokines and/or cytokines when said compound or said pharmaceutical composition has not been administered.

In an embodiment, said decreased expression comprises a decrease in protein expression of said one or more inflammatory chemokines and/or cytokines of at least 5%, preferably at least 8%, more preferably at least 10%, more preferably at least 15%, more preferably at least 20%, more preferably at least 25%, more preferably at least 30%, more preferably at least 35%, more preferably at least 40%, more preferably at least 40%, more preferably at least 45%, more preferably at least 50%, more preferably at least 55%, more preferably at least 60%, more preferably at least 65%, more preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 95%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99%, such as 100% compared to the in vitro expression of one or more of said inflammatory chemokines and/or cytokines when said compound or said pharmaceutical composition has not been administered.

Said in vitro inhibition of the NLRP3 inflammasome can be measured in any suited cell culture type, such as for instance murine immortalised bone-marrow-derived macrophages (iBMM), human THP-1 monocytic cells, primary bone marrow-derived macrophages (BMM).

The invention is further described by the following non-limiting examples which further illustrate the invention, and are not intended to, nor should they be interpreted to, limit the scope of the invention.

EXAMPLES AND/OR DESCRIPTION OF FIGURES

The present invention will now be further exemplified with reference to the following examples. The present invention is in no way limited to the given examples or to the embodiments presented in the figures.

Example 1. Investigating the effects of a compound for use according to an embodiment of the current invention on release of IL-1B by mouse myeloid cells treated with NLRP3-stimulating treatments.

Aim

To determine whether the compounds for use according to an embodiment of the current invention affect NLRP3-dependent IL-1 p release in vitro.

Methods

Cells

Primary bone marrow-derived macrophages (BMM) from C57BL/6 mice were cultured in RPMI-1640 medium supplemented with 10% foetal calf serum, 2 mM L- glutamine, 50 U/ml penicillin and 50 mg/ml streptomycin and 15% L-cell conditioned medium, which contains macrophage-colony stimulating factor (M-CSF), for 7-12 days in low-adherence T175 tissue culture flasks. Differentiated macrophages were removed from flask with trypsin, counted, plated on 96 well tissue culture plates at a concentration of 5 x 10 5 cells/ml and cultured overnight prior to treatment.

Stimulations

Cells were first primed with lipopolysaccharide (LPS, 100 ng/ml) for 4 hours. This induces production of pro-IL-ip and pro-IL-18, as well as NLRP3 and pro-caspase- 1. After this, media was replenished, and inhibitor (or vehicle control) added for 30 minutes. In the following examples 1 and 2, a compound according to Formula Ic (denominated "Compound A") for use according to an embodiment of the current invention was tested.

Compound A:

Cells were then treated with 5-10 pM nigericin for 45 minutes to induce NLRP3 inflammasome activation and release of IL-ip. Plates were centrifuged to pellet cells and the culture supernatant (200 pl) collected and stored at -20°C for analysis of IL- ip by ELISA.

ELISA

Culture supernatants were tested for IL- 1(3 using commercial ELISA kits (Biolegend and R&D), according to the manufacturer's protocols.

Results

We tested the effects of Compound A in primary BMM. Using cells from 3 individual animals, we observed significant inhibition of NLRP3-dependent IL-ip release with Compound A (Figure 1). Maximal inhibition with Compound A was in the nanomolar range (Figure 1). For this, cells were primed with LPS (100 ng/ml) for 4 h, treated with Compound A for 30 min and then with nigericin (5 pM) for 45 min. Supernatants were collected and IL-ip measured by ELISA. Data are means +/- SEM of 3 separate mice. * p < 0.05, **p < 0.01, *** p < 0.005. Data analysed by one way ANOVA + Dunnett's multiple comparison test. Summary of Outcomes

Taken together, our data highlight significant activity of Compound A for use according to an embodiment of the current invention against NLRP3-dependent IL- 10 release. Significantly, early IC50 studies suggest activity in the low nanomolar range, which suggests very impressive potency.

Example 2: Investigating the effects of compounds for use according to an embodiment of the current invention on release of IL-1B and pyroptosis in human THP-1 cells treated with NLRP3-stimulating treatments

Aim

To determine whether the compounds for use according to an embodiment of the current invention affect NLRP3-dependent cytokine release and cell death in human THP-1 cells in vitro.

Cells

Human THP-1 cells

Human THP-1 monocytic cells were differentiated into macrophages with phorbol 12- myristate 13-acetate (PMA, 5 ng/ml) for 72 hours prior to stimulations. The cells are cultured in RPMI-1640 medium supplemented with 10% foetal calf serum, 2 mM L- glutamine, 50 U/ml penicillin and 50 mg/ml streptomycin. Experiments with THP-1 cells will be repeated a minimum of 3 times.

Stimulations

Cells were first primed with lipopolysaccharide (LPS, 100 ng/ml) for 3 or 4 hours. This induces production of pro-IL-10 and pro-IL-18, as well as NLRP3 and pro- caspase-1. After this, media was replenished, and Compound A, Compound B, Compound C or Compound D (or vehicle control) was added for 30 to 60 minutes. Compounds A-D are compounds for use according to an embodiment of the current invention (see Example 1 for the formula of Compound A and see below for the formulae of Compounds B-D). A dose response curve was generated by treating cells with Compound A at doubling dilutions. Compounds B and D were exemplified at 1 nM, Compound C at 10 pM. Cells were then treated with 5-10 pM nigericin for 45 minutes to 3 hours to induce NLRP3 inflammasome activation and release of IL-10 and IL-18. Plates were centrifuged to pellet cells at the bottom of wells and half of the culture supernatant (100 pl) was collected and stored at -20°C for analysis of cytokines by ELISA or AlphaLISA. The other half was a used immediately for measurement of LDH release, as a proxy for cell death/pyroptosis.

ELISA and AlphaLISA

Culture supernatants were tested for IL- 1(3 using commercial ELISA kits (Biolegend and R&D) or AlphaLISA kits (PerkinElmer), according to the manufacturer's protocols.

LDH Release assay

NLRP3-dependent cells death was measured by release of Lactate dehydrogenase (LDH) in the culture supernatant, using a Lactate dehydrogenase activity assay kit (Sigma), according to the manufacturer's protocol.

Statistical analyses

All experiments were analysed using appropriate statistical tests (e.g., t-test or oneway ANOVA followed by Tukey's post-hoc if data is parametric, Dunn's test for multiple comparisons if data is non-parametric) on means ± SEM. Statistical significance was displayed as * or # when p<0.05, ** or ## when p<0.01 and *** when p<0.001. The results were graphed using GraphPad Prism software. If appropriate, IC50 curves were be plotted.

Results

In human THP-1 cells, combination of LPS with nigericin induced strong increases in IL-ip levels as compared to LPS alone (Figure 2), reflecting the secretion of this cytokine associated with NLRP3-inflammasome activation. Application of Compound A at concentrations ranging from 0.04 nM to 10 pM dose-dependently inhibited nigericin-induced IL-ip expression (Figure 2A), indicating the inhibitory activity of this compound on NLRP3-inflammasome activation in human macrophages.

Compounds B, C and D showed partial inhibition of NLRP3-dependent release of IL- ip and cell death (pyroptosis, Figure 2B and C) at the respective concentrations tested. Example 3: Effects of a compound for use according to an embodiment of the current invention on NLRP3-associated gout flare in a mouse model of gouty arthritis.

Aim

To determine whether the MIF inhibitors for use according to an embodiment of the current invention affect NLRP3-dependent inflammation in a mouse model of monosodium urate (MSU) crystals induced gout. This model has been selected because it is dependent on NLRP3 inflammasome activation (Tulsi Patil, Arun Soni, Sanjeev Acharya, A brief review on in vivo models for Gouty Arthritis, Metabolism Open, Volume 11, 2021, 100100, ISSN 2589-9368, https://doi.Org/10.1016/j.metop.2021.100100.https://www.scie ncedirect.com/scie nce/article/pii/S2589936821000244).

Animals and treatment

Ten-week-old male C57BL/6J mice were distributed into 3 experimental groups (n=8 per group) and injected with MSU crystals (Sigma-Aldrich, St Louis, MO) or with vehicle only (baseline control group) into the tibiofemoral joint cavity (100 pg/10 pl/cavity) as described into Galvao et al., 2016 (Galvao, Izabela et al. "Macrophage migration inhibitory factor drives neutrophil accumulation by facilitating IL-ip production in a murine model of acute gout." Journal of leukocyte biology vol. 99,6 (2016): 1035-43. doi: 10.1189/jlb.3MA0915-418R) and under intraperitoneal anesthesia (80 mg/kg ketamine : 15 mg/kg xylazine). One group of mice was orally administered with Compound A (see Example 1; 80 mg/kg in 4 ml/kg of Miglyol 812N) and 2 groups of mice with Miglyol 812N only 2 hours before and 3 hours after MSU injection. The mice were euthanized 15 hours post-MSU injection, articular cavity wash (with PBS containing 3% BSA) and periarticular tissue were then collected (Figure 3A).

Quantification of inflammatory markers

Leukocyte numbers in the articular cavity wash were determined in a Neubauer chamber after staining with Turk's solution. Differential counts were performed in Shandon CytoSpin III (Thermo Shandon, Frankfurt, Germany) preparations by evaluating the percentage of neutrophils on a slide stained with May Grunwald Giemsa. IL-ip cytokine was quantified in periarticular tissues collected and homogenized in PBS containing antiproteases. The samples were centrifuged and the supernatant was used for IL-ip ELISA determination in accordance with the manufacturer's instructions.

Statistical analyses

The results were expressed as means ± SEM of the 8 animals endpoints. Student t- test were used to compare the means from the vehicle control group and from the MSU control group on one side, and the means from the MSU control group and from the MSU + Compound A group on the other side. Statistical significance was displayed as ## when p<0.01 and *** when p<0.001. The results were graphed using GraphPad Prism software.

Results

MSU crystal injection induced substantial infiltrations of immune cells into the joints, as exemplified by the increase in total leukocytes and neutrophils counts in articular cavity wash (Figure 3B and C). In joint tissue, MSU also induced drastic elevation of IL- ip (Figure 3D), pointing the involvement of NLRP3 inflammasome in the setting. The systemic administration of Compound A inhibited the response to MSU by 40%, showing that this compound can dampen an inflammatory response associated with NLRP3 inflammasome in vivo.

The present invention is in no way limited to the embodiments described in the examples and/or shown in the figures. On the contrary, methods according to the present invention may be realized in many different ways without departing from the scope of the invention.