WO2023198833A1 - Composé pour l'inhibition de l'inflammasome nlrp3 - Google Patents

Composé pour l'inhibition de l'inflammasome nlrp3 Download PDF

Info

Publication number
WO2023198833A1
WO2023198833A1 PCT/EP2023/059690 EP2023059690W WO2023198833A1 WO 2023198833 A1 WO2023198833 A1 WO 2023198833A1 EP 2023059690 W EP2023059690 W EP 2023059690W WO 2023198833 A1 WO2023198833 A1 WO 2023198833A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
heteroaryl
aryl
alkyl
amino
Prior art date
Application number
PCT/EP2023/059690
Other languages
English (en)
Inventor
Mathilde METNA
Bart WUURMAN
Original Assignee
Apaxen Sa
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Apaxen Sa filed Critical Apaxen Sa
Publication of WO2023198833A1 publication Critical patent/WO2023198833A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/423Oxazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/428Thiazoles condensed with carbocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • 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.
  • 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.
  • inflammasome 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.
  • NLRP3 inflammasome Involvement of the NLRP3 inflammasome in different kinds of diseases necessitates new avenues to design drugs targeting the NLRP3 inflammasome.
  • 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.
  • IL-ip antibodies or recombinant IL-ip receptor antagonist such as canakinumab and anakinra, respectively.
  • 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.
  • BHB P-hydroxybutyrate
  • DMSO dimethyl sulfoxide
  • type I interferon type I interferon
  • 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.
  • 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.
  • the present invention aims to resolve at least some of the problems and disadvantages mentioned above.
  • 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,
  • 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.
  • 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
  • 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.
  • a compartment refers to one or more than one compartment.
  • the value to which the modifier "about” refers is itself also specifically disclosed.
  • 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.
  • 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, t
  • 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.
  • haloalkyl groups are CF3, CHF2 and CH2F.
  • Hydroalkyl 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-
  • 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.
  • solvent 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.
  • 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 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.
  • a therapeutically effective amount 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.
  • 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.
  • 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.
  • 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.
  • administration 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.
  • pharmaceutically acceptable is meant that the ingredients of a pharmaceutical composition are compatible with each other and not deleterious to the subject thereof.
  • pharmaceutically acceptable carrier 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.
  • preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologies standards.
  • MIF 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.
  • 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.
  • the inhibitor of MIF of the invention inhibits MIF binding to CD74 and CXCRs (including CXCR2, CXCR4 and/or CXCR7).
  • 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.
  • 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.
  • 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.
  • 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.
  • ALR AIM2-like receptor
  • pyrin protein or a nucleotide-binding domain, leucine-rich-repeat-containing protein (NOD-like receptor, NLR), such as NLRP1, NLRC4, or NLRP3.
  • NLR leucine-rich-repeat-containing protein
  • 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.
  • ASC caspas
  • Activation of the NLRP3 inflammasome is a two-step process.
  • 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.
  • ATP extracellular adenosine triphosphate
  • pore-forming toxins such as nigericin and particulates
  • uric acid crystals silica, and alum
  • alum as well as bacterial, protozoan, and viral pathogens.
  • NLRP3 inflammasome assembly 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.
  • NIMA-related kinase-7 NIMA-related kinase-7
  • 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.
  • 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.
  • NLRP1 nod-like receptor protein 1
  • NLRP3 nod-like receptor protein 3
  • AIM-2 abent in melanoma 2
  • NLRC4 nod-like receptor family CARD domain containing 4
  • GSDMD gasdermin D
  • caspase-1 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.
  • 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.
  • NLRP3 inflammasome 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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 also known as glycosylation-inhibiting factor, L-dopachrome isomerase, or phenylpyruvate tautomerase
  • MIF Macrophage migration inhibitory factor
  • DDT or MIF-2 D-dopachrome tautomerase
  • 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.
  • 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.
  • the invention relates to a compound or a pharmaceutical composition
  • 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
  • said treatment comprises administering said compound to a subject in need of treatment, wherein said compound inhibits NLRP3 inflammasome activity
  • 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,
  • 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.
  • 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.
  • 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.
  • 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.
  • 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).
  • 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.
  • 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.
  • a disease, disorder or condition responsive to the inhibition of the NLRP3 inflammasome 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • the amount of NLR.P3 activation/signaling is determined by measuring the expression of one or more inflammatory chemokines and/or cytokines.
  • said chemokines and/or cytokines are chosen from: IL-ip, IL-18, IL-lo, IL-37, IL-38.
  • 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.
  • the compound for use according to the current invention 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.
  • 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).
  • 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, phosphoronate, sodium bicarbonate, sodium
  • 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.
  • 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.
  • Ar is a phenyl group, optionally substituted.
  • Ar 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.
  • 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.
  • Ar is a phenol group or a prodrug thereof.
  • the prodrug of the phenol group is selected from aminoacid, carbamate, carbonate, ester, thioester, phosphonate, phosphonate methyloxy, phosphonate methylamino, alkyloxy and alkylthio groups.
  • R 1 and R 4 represent hydrogen atoms.
  • R 1 , R 4 and at least one of R 2 or R 3 represent hydrogen atoms.
  • R 1 , R 3 and R 4 represent hydrogen atoms and R 2 preferably represents an alkyl group, more preferably methyl.
  • 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.
  • 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,
  • 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, alkyny
  • 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, alkyny
  • 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, alkyny
  • 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,
  • 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.
  • R 3 represents H.
  • 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.
  • R 2 represents an alkyl group, preferably methyl, and R 3 represents a hydrogen atom.
  • R 2 represents a haloalkyl group, preferably trifluoromethyl, and R 3 represents a hydrogen atom.
  • R 2 and R 3 represents hydrogen atoms.
  • Z 2 represents a hydroxyl group.
  • Z 1 , Z 3 , Z 4 and Z 5 preferably represent hydrogen atoms.
  • Z 2 represents a hydroxyl group and Z 3 represents a halogen, preferably a fluorine atom.
  • Z 1 , Z 4 and Z 5 preferably represent hydrogen atoms.
  • said compound is a compound according to Formula Ic or a pharmaceutically acceptable enantiomer, salt or solvate thereof, wherein said Formula Ic is
  • 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.
  • R 5 represents H.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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, glyco
  • 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.
  • 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
  • a free acid by reacting a free acid with a suitable organic or inorganic base.
  • a suitable organic or inorganic base 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, ste
  • 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.
  • pharmaceutically acceptable salts include hydrochloride/chloride, hydrobromide/bromide, bisulphate/sulphate, nitrate, citrate, and acetate.
  • the compounds of Formula I and subformulae thereof may also form internal salts, and such compounds are within the scope of the invention.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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 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.
  • inhibition of the NLPR.3 inflammasome is achieved with nanomolar potency.
  • 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).
  • said patient is a mammalian patient, preferably a human patient.
  • 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.
  • 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, silicos
  • 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.
  • MSU monosodium urate crystals
  • said arthritic disease is gout.
  • 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.
  • 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.
  • said auto-immune disease is selected from multiple sclerosis (MS), systemic lupus erythematosus (SLE), rheumatoid arthritis (RA) and/or inflammatory bowel disease (IBD).
  • MS multiple sclerosis
  • SLE systemic lupus erythematosus
  • RA rheumatoid arthritis
  • IBD inflammatory bowel disease
  • metabolic disorders include but are not limited to: diabetes, obesity, steroid- resistance, glucose intolerance, metabolic syndrome.
  • 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.
  • 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.
  • 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
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • the patient is a warm-blooded animal, more preferably a human.
  • 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.
  • the patient or subject is affected with, preferably is diagnosed with a disorder, a disease or a condition related to the NLR.P3 inflammasome.
  • the patient or subject is at risk of developing a disorder, a disease or a condition related to the NLR.P3 inflammasome.
  • the patient or subject presents a non-genetic predisposition to a disorder, a disease or a condition related to the NLRP3 inflammasome.
  • 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.
  • 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.
  • the compounds of Formula I 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.
  • 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.
  • 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.
  • compositions, the pharmaceutical composition or the medicament for use according to the present invention is administered or is to be administered with other active agents.
  • the composition, the pharmaceutical composition or the medicament and the other active agent may be administered separately or in conjunction.
  • 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.
  • 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.
  • 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.
  • parenteral administration such as by intravenous, intramuscular or subcutaneous injection or intravenous infusion
  • topical administration including ocular
  • 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.
  • 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,
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • 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.
  • MIF inhibitors 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 fruct
  • “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.
  • 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).
  • 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
  • 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.
  • 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.
  • 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.
  • 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.
  • 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,
  • 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.
  • 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.
  • 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.
  • 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).
  • iBMM murine immortalised bone-marrow-derived macrophages
  • BMM primary bone marrow-derived macrophages
  • 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.
  • BMM 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.
  • M-CSF macrophage-colony stimulating factor
  • 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
  • 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.
  • LPS lipopolysaccharide
  • 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.
  • 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.
  • LDH Lactate dehydrogenase
  • 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.
  • 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.
  • MSU monosodium urate
  • 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.sciencedirect.com/scie nce/article/pii/S2589936821000244).
  • mice were 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).
  • 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.
  • 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.
  • 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).
  • 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.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne un composé ou une composition pharmaceutique comprenant une quantité thérapeutiquement efficace dudit composé pour une utilisation dans le traitement d'une maladie, d'un trouble ou d'un état sensible à l'inhibition du domaine pyrine de la famille NLR contenant l'inflammasome 3 (NLRP3), ledit traitement comprenant l'administration dudit composé à un sujet ayant besoin d'un traitement, ledit composé inhibant l'activité de l'inflammasome NLRP3, ledit composé étant un composé selon la formule I ou un énantiomère, sel ou solvate pharmaceutiquement acceptable de celui-ci. Dans un dernier aspect, la présente invention concerne l'utilisation d'un composé de formule I tel que divulgué ici, ou d'un énantiomère, sel ou solvate pharmaceutiquement acceptable de celui-ci, pour l'inhibition in vitro de l'inflammasome NLRP3.<i />
PCT/EP2023/059690 2022-04-13 2023-04-13 Composé pour l'inhibition de l'inflammasome nlrp3 WO2023198833A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP22168229 2022-04-13
EP22168229.7 2022-04-13

Publications (1)

Publication Number Publication Date
WO2023198833A1 true WO2023198833A1 (fr) 2023-10-19

Family

ID=81308491

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2023/059690 WO2023198833A1 (fr) 2022-04-13 2023-04-13 Composé pour l'inhibition de l'inflammasome nlrp3

Country Status (1)

Country Link
WO (1) WO2023198833A1 (fr)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015155358A1 (fr) 2014-04-10 2015-10-15 Mifcare Inhibiteurs de mif
WO2015173433A1 (fr) 2014-05-16 2015-11-19 Mifcare Inhibiteurs du mif pour le traitement d'urgence ou chronique de l'hypertension pulmonaire

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015155358A1 (fr) 2014-04-10 2015-10-15 Mifcare Inhibiteurs de mif
WO2015173433A1 (fr) 2014-05-16 2015-11-19 Mifcare Inhibiteurs du mif pour le traitement d'urgence ou chronique de l'hypertension pulmonaire

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
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 *
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
LANG ET AL., NAT COMMUN, 2018, pages 9
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 <URL:http://www.nature.com/articles/s41467-018-04581-2> DOI: 10.1038/s41467-018-04581-2 *
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 <URL:https://www.nature.com/articles/s41420-022-00859-z> 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 *
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 *
SHIN ET AL., ARTHRITIS RHEUMATOL., 2019
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 <URL:https://onlinelibrary.wiley.com/doi/full-xml/10.1002/art.40672> DOI: 10.1002/art.40672 *
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 *
TULSI PATILARUN SONISANJEEV ACHARYA: "A brief review on in vivo models for Gouty Arthritis", METABOLISM OPEN, vol. 11, 2021, Retrieved from the Internet <URL:https://doi.org/10.1016/j.metop.2021.100100.https://www.sciencedirect.com/science/article/pii/S2589936821000244>

Similar Documents

Publication Publication Date Title
US10500209B2 (en) Quinazolinones as bromodomain inhibitors
JP7044769B2 (ja) Usp30の阻害剤としての活性を有するシアノ置換複素環
JP6461118B2 (ja) ブロモドメイン阻害剤としての新規の置換された二環式化合物
KR102531689B1 (ko) 6,7―디히드로피라졸로[1,5―α]피라진―4(5H)―온 화합물 및 MGLUR2 수용체의 음성 알로스테릭 조절제로서의 그 용도
JP2018527309A (ja) ウイルス感染の治療及び予防のための新規な置換アミノチアゾロピリミジンジオン
JP2019533659A (ja) Usp30の阻害剤としての活性を有するシアノ置換ヘテロ環
CA2948589A1 (fr) Composes 5-chloro -2-difluoromethoxyphenyl pyrazolopyrimidine utilises en tant qu&#39;inhibiteurs de jak
US8895581B2 (en) 1H-imidazo[4,5-c]quinolines
KR20190097242A (ko) Magl 억제제로서의 헤테로시클릭 스피로 화합물
WO2014080290A2 (fr) Amines cycliques servant d&#39;inhibiteurs de bromodomaines
JP6609306B2 (ja) 6,7−ジヒドロピラゾロ[1,5−a]ピラジン−4(5H)−オン化合物およびMGLUR2受容体の負のアロステリック調節因子としてのその使用
KR20060123228A (ko) 글루코키나아제 조절물질로서의 피리딘 카르복실산 유도체
CA2865714A1 (fr) Sulfonamides de pyrazol-1-yl benzene convenant comme antagonistes de ccr9
RU2722383C2 (ru) Производные хромена в качестве ингибиторов фосфоинозитид-3-киназ
KR102531693B1 (ko) 6,7―디히드로피라졸로[1,5―a]피라진―4(5H)―온 화합물 및 MGLUR2 수용체의 음성 알로스테릭 조절제로서의 그 용도
JP2020536884A (ja) Fshdの治療のためにdux4および下流遺伝子発現を低減するp38キナーゼ阻害剤
KR20170040299A (ko) 이미다조피리다진 화합물
JP2019517996A (ja) PDE1阻害剤としての1,5−ジヒドロ−4H−ピラゾロ[3,4−d]ピリミジン−4−オン及び1,5−ジヒドロ−4H−ピラゾロ[4,3−c]ピリジン−4−オン
JP2014530807A (ja) H3受容体阻害剤としてのピペリジン及びピペラジン環を含むカルバメート/尿素誘導体
KR20180101568A (ko) 6,7-디히드로-5H-피라졸로[5,1-b][1,3]옥사진-2-카르복스아미드 화합물
CA3158731A1 (fr) Composes antagonistes du recepteur de l&#39;adenosine
EP3847170B1 (fr) Nouveaux composés amidine cyclique pour le traitement d&#39;une maladie auto-immune
JP2022535672A (ja) MRGX2の阻害薬である3-アミノ-4H-ベンゾ[e][1,2,4]チアジアジン1,1-ジオキシド誘導体
WO2023198833A1 (fr) Composé pour l&#39;inhibition de l&#39;inflammasome nlrp3
CN111187251A (zh) 吡啶酰基哌啶衍生物及其用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23719722

Country of ref document: EP

Kind code of ref document: A1