[go: up one dir, main page]

WO2024160691A1 - Pyrrolo[1,2-d][1,2,4]triazines et pyrazolo[1,5-d][1,2,4]triazines utiles en tant qu'inhibiteurs de nlrp3 - Google Patents

Pyrrolo[1,2-d][1,2,4]triazines et pyrazolo[1,5-d][1,2,4]triazines utiles en tant qu'inhibiteurs de nlrp3 Download PDF

Info

Publication number
WO2024160691A1
WO2024160691A1 PCT/EP2024/051979 EP2024051979W WO2024160691A1 WO 2024160691 A1 WO2024160691 A1 WO 2024160691A1 EP 2024051979 W EP2024051979 W EP 2024051979W WO 2024160691 A1 WO2024160691 A1 WO 2024160691A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
triazin
trifluoromethyl
mixture
compounds
Prior art date
Legal status (The legal status 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 status listed.)
Ceased
Application number
PCT/EP2024/051979
Other languages
English (en)
Inventor
Oscar MAMMOLITI
Soufyan JERHAOUI
Laura PEREZ BENITO
Edgar Jacoby
Santiago CAÑELLAS ROMAN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Pharmaceutica NV
Original Assignee
Janssen Pharmaceutica NV
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 Janssen Pharmaceutica NV filed Critical Janssen Pharmaceutica NV
Priority to AU2024214354A priority Critical patent/AU2024214354A1/en
Priority to EP24702714.7A priority patent/EP4658654A1/fr
Priority to KR1020257028762A priority patent/KR20250134147A/ko
Priority to CN202480022485.6A priority patent/CN120957995A/zh
Publication of WO2024160691A1 publication Critical patent/WO2024160691A1/fr
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • pyrrolo[l,2-d][l,2,4]triazines and pyrazolo[l,5- d][l,2,4]triazines that are useful as inhibitors of the NOD-like receptor protein 3 (NLRP3) inflammasome pathway.
  • processes for the preparation of said compounds, pharmaceutical compositions comprising said compounds, methods of using said compounds in the treatment of various diseases and disorders mediated by the NLRP3 inflammasome pathway are also described herein.
  • Inflammasomes considered as central signalling hubs of the innate immune system, are multi-protein complexes that are assembled upon activation of a specific set of intracellular pattern recognition receptors (PRRs) by a wide variety of pathogen- or danger- associated molecular patterns (PAMPs or DAMPs).
  • PRRs pattern recognition receptors
  • PAMPs or DAMPs pathogen- or danger- associated molecular patterns
  • the NLRP3 inflammasome is assembled upon detection of environmental crystals, pollutants, host-derived DAMPs and protein aggregates (Tartey S and Kanneganti TD. Immunology, 2019 Apr;156(4):329-338).
  • Clinically relevant DAMPs that engage NLRP3 include uric acid and cholesterol crystals that cause gout and atherosclerosis, amyloid-P fibrils that are neurotoxic in Alzheimer’s disease and asbestos particles that cause mesothelioma (Kelley et al., IntJMol Sci, 2019 Jul 6;20(13)).
  • NLRP3 is activated by infectious agents such as Vibrio cholerae, fungal pathogens such as Aspergillus fumigatus and Candida albicans,' adenoviruses, influenza A virus and SARS-CoV-2 (Tartey and Kanneganti, 2019; Fung et al. Emerg Microbes Infect, 2020 Mar 14;9(l):558-570).
  • infectious agents such as Vibrio cholerae, fungal pathogens such as Aspergillus fumigatus and Candida albicans,' adenoviruses, influenza A virus and SARS-CoV-2 (Tartey and Kanneganti, 2019; Fung et al. Emerg Microbes Infect, 2020 Mar 14;9(l):558-570).
  • NLRP3 activation mechanism Although the precise NLRP3 activation mechanism remains unclear, for human monocytes, it has been suggested that a one-step activation is sufficient while in mice a two-step mechanism is in place. Given the multitude in triggers, the NLRP3 inflammasome requires add-on regulation at both transcriptional and post- transcriptional level (Yang Y et al., Cell Death Dis, 2019 Feb 12; 10(2): 128).
  • the NLRP3 protein consists of an N-terminal pyrin domain, followed by a nucleotide-binding site domain (NBD) and a leucine-rich repeat (LRR) motif on C- terminal end (Sharif et al., Nature, 2019 Jun; 570(7761):338-343).
  • NBD nucleotide-binding site domain
  • LRR leucine-rich repeat
  • NLRP3 aggregates with the adaptor protein, apoptosis-associated speck-like protein (ASC), and with the protease caspase- 1 to form a functional inflammasome.
  • ASC apoptosis-associated speck-like protein
  • procaspase-1 Upon activation, procaspase-1 undergoes autoproteolysis and consequently cleaves gasdermin D (Gsdmd) to produce the N-terminal Gsdmd molecule that will ultimately lead to pore-formation in the plasma membrane and a lytic form of cell death called pyroptosis.
  • Gsdmd gasdermin D
  • caspase-1 cleaves the pro- inflammatory cytokines pro-IL-ip and pro-IL-18 to allow release of its biological active form by pyroptosis (Kelley et al., 2019).
  • Dysregulation of the NLRP3 inflammasome or its downstream mediators are associated with numerous pathologies ranging from immune/inflammatory diseases, auto-immune/auto-inflammatory diseases (Cryopyrin-associated Periodic Syndrome (Miyamae T. Paediatr Drugs, 2012 Apr 1; 14(2): 109-17); sickle cell disease; systemic lupus erythematosus (SLE)) to hepatic disorders (e.g. non-alcoholic steatohepatitis (NASH), chronic liver disease, viral hepatitis, alcoholic steatohepatitis, non-alcoholic fatty acid liver disease, and alcoholic liver disease) (Szabo G and Petrasek J.
  • NASH non-alcoholic steatohepatitis
  • NASH non-alcoholic steatohepatitis
  • chronic liver disease viral hepatitis
  • viral hepatitis alcoholic steatohepatitis
  • non-alcoholic fatty acid liver disease non-alcoholic fatty acid liver disease
  • kidney related diseases hypertensive nephropathy (Krishnan et al., Br J Pharmacol, 2016 Feb;173(4):752-65), hemodialysis related inflammation and diabetic nephropathy which is a kidney -related complication of diabetes (Type 1, Type 2 and mellitus diabetes), also called diabetic kidney disease (Shahzad et al., Kidney Int, 2015 Jan;87(l):74-84) are associated to NLRP3 inflammasome activation.
  • cardiovascular or metabolic disorders e.g. cardiovascular risk reduction (CvRR), atherosclerosis, type I and type II diabetes and related complications (e.g. nephropathy, retinopathy), peripheral artery disease (PAD), acute heart failure and hypertension.
  • myeloproliferative neoplasms myeloproliferative neoplasms, leukemias, myelodysplastic syndromes (MOS), myelofibrosis, lung cancer, colon cancer (Ridker et al., Lancet, 2017 Oct 21;390(10105): 1833-1842; Derangere et al., Cell Death Differ.
  • Described herein are compounds which inhibit the NLRP3 inflammasome pathway.
  • compositions comprising a therapeutically effective amount of a compound provided herein.
  • compositions comprising such compounds for use in the treatment of a disease or disorder mediated by the NLRP3 inflammasome pathway, for example neurodegenerative disorders such as Alzheimer’s Disease.
  • the instant compounds in the manufacture of a medicament for the treatment of a disease or disorder mediated by the NLRP3 inflammasome pathway, for example neurodegenerative disorders such as Alzheimer’s Disease.
  • a method of treating a disease or disorder mediated by the NLRP3 inflammasome pathway for example neurodegenerative disorders such as Alzheimer’s Disease.
  • R 7 and R 8 each independently are H, CH 3 or F.
  • R 7 and R 8 each independently are H, CH 3 or F, with the proviso that the compound is not
  • compounds of Formula (I) having Formula wherein with the proviso that a) R6 is selected from CH 2 CH 3 , CH(CH 3 ) 2 , CH 2 CH 2 OH, CH 2 CH 2 F, CD 3 , when R7 and R8 each independently are H, CH 3 or F; and b) R6 is CH 3 , CH 2 CH 3 , CH(CH 3 ) 2 , CH 2 CH 2 OH, CH 2 CH 2 F, CD 3 , when R7
  • R 7 and R 8 each independently are H, CH3 or F; with the proviso that a) R 6 is selected from CH2CH3, CH(CH 3 ) 2 , CH2CH2OH, CH2CH2F, CD 3 , when
  • R 7 and R 8 each independently are H, CH3 or F; and b) R 6 is CH 3 , CH2CH3, CH(CH 3 ) 2 , CH2CH2OH, CH2CH2F, CD 3 , when
  • R 7 is CH3 or F
  • R 8 is H, CH3 or F; with the proviso that the compound is not is H, CH3 or F, and R 8 is H, or both R 7 and R 8 are F.
  • R 6 is CH3 or CH2CH3; R 7 is CH3 or F, and R 8 is H, or both R 7 and R 8 are F.
  • R 6 is CH2CH3; R 7 is H, CH3 or F, and R 8 is H, or both R 7 and R 8 are F.
  • R 1 is wherein a) R 6 is CH3 or CH2CH3; R 7 is H, CH3 or F, and R 8 is H, or both R 7 and R 8 are F; or b) R 6 is CH3 or CH2CH3; R 7 is CH3 or F, and R 8 is H, or both R 7 and R 8 are F; or c) R 6 is CH2CH3; R 7 is H, CH3 or F, and R 8 is H, or both R 7 and R 8 are F; or d) R 6 is CH3 or CH2CH3; and both R 7 and R 8 are F; or
  • R 2 , R 4 and R 5 are hydrogen.
  • R 2 is hydrogen and R 3 is CF3.
  • R 3 is CF3.
  • R 6 is CH 3 , CH2CH3, CH(CH 3 ) 2 , CH2CH2OH, CH2CH2F or CD 3 .
  • R 6 is CH2CH3, CH(CH 3 ) 2 , CH2CH2OH, CH2CH2F or CD 3 .
  • salts include acid addition salts and base addition salts.
  • Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound as provided herein with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound provided herein in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids.
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table.
  • the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like.
  • Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine, and tromethamine.
  • the instant compounds may contain double bonds and may thus exist as E (entgegeri) and Z (ziisammeri) geometric isomers about each individual double bond.
  • Compounds as provided herein may contain one or more asymmetric carbon atoms and may therefore exhibit enantiomerism or diastereoisomerism.
  • Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation.
  • the various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques.
  • the desired isomers may be made by reaction of an appropriate enantiomeric starting material under conditions which will not cause racemisation or epimerisation, or by reaction of an appropriate starting material with a ‘chiral auxiliary’ which can subsequently be removed at a suitable stage, by resolution, including dynamic resolution, for example salt formation with a homochiral acid followed by separation of the diastereomeric salts by conventional means such as crystallization, or by reaction with an appropriate chiral reagent or chiral catalyst.
  • resolution including dynamic resolution, for example salt formation with a homochiral acid followed by separation of the diastereomeric salts by conventional means such as crystallization, or by reaction with an appropriate chiral reagent or chiral catalyst.
  • stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated. Where stereochemistry is specified by a solid or dashed wedge representing a particular configuration, then that stereoisomer is so specified and defined.
  • Absolute configurations are specified according to the Cahn-Ingold-Prelog system.
  • the configuration at an asymmetric atom is specified by either R or S.
  • Resolved compounds whose absolute configuration is not known can be designated by (+) or (-) depending on the direction in which they rotate polarized light.
  • a specific stereoisomer is identified, this means that said stereoisomer is substantially free, i.e. associated with less than 50%, preferably less than 20%, more preferably less than 10%, even more preferably less than 5%, in particular less than 2% and most preferably less than 1%, of the other isomers.
  • a compound of formula (I) is for instance specified as (R)
  • the compounds may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like.
  • isotopically-labelled compounds wherein one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature (or the most abundant one found in nature).
  • Exemplary isotopes include isotopes of hydrogen, carbon, nitrogen, oxygen, and fluorine, such as 2H, 3H, 11C, 13C, 14C , 13N, 15O, 17O, 18O, and 18F.
  • Tritiated (3H) and carbon-l4 (14C) isotopes are useful for their ease of preparation and detectability.
  • Isotopes such as 15O, 13N, 11C and 18F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy.
  • PET positron emission tomography
  • Isotopically labelled compounds can generally be prepared by following procedures analogous to those disclosed in the Examples hereinafter.. Unless otherwise specified, C1-q alkyl groups (where q is the upper limit of the range) defined herein may be straight-chain or be branched-chain.
  • C3-q cycloalkyl refers to an alkyl group that is cyclic, for instance cycloalkyl groups may be monocyclic or, if there are sufficient atoms, bicyclic. In an embodiment, such cycloalkyl groups are monocyclic. Substituents may be attached at any point on the cycloalkyl group..
  • halo when used herein, preferably includes fluoro, chloro, bromo and iodo.
  • C1-q alkoxy groups refers to the radical of formula -ORa, where Ra is a C1-q alkyl group as defined herein.
  • HaloC1-q alkyl (where q is the upper limit of the range) groups refer to C1-q alkyl groups, as defined herein, where such group is substituted by one or more halo.
  • HydroxyC1-q alkyl (where q is the upper limit of the range) refers to C1-q alkyl groups, as defined herein, where such group is substituted by one or more (e.g. one) hydroxy (-OH) groups (or one or more, e.g. one, of the hydrogen atoms is replaced with -OH).
  • haloCi- q alkoxy and hydroxyCi- q alkoxy represent corresponding -OCi- q alkyl groups that are substituted by one or more halo, or, substituted by one or more (e.g. one) hydroxy, respectively.
  • the instant compounds can generally be prepared by a succession of steps, each of which is known to the skilled person.
  • the compounds can be prepared according to the following synthesis methods.
  • the compounds of Formula (I) may be synthesized in the form of racemic mixtures of enantiomers which can be separated from one another following art-known resolution procedures.
  • the racemic compounds of Formula (I) may be converted into the corresponding diastereomeric salt forms by reaction with a suitable chiral acid. Said diastereomeric salt forms are subsequently separated, for example, by selective or fractional crystallization and the enantiomers are liberated therefrom by alkalination.
  • An alternative manner of separating the enantiomeric forms of the compounds of Formula (I) involves liquid chromatography using a chiral stationary phase or a chiral supercritical fluid chromatography (SFC).
  • Said pure stereochemically isomeric forms may also be derived from the corresponding pure stereochemically isomeric forms of the appropriate starting materials, provided that the reaction occurs stereospecifically.
  • the absolute configuration of the compounds reported herein was determined by analysis of the racemic mixture by supercritical fluid chromatography (SFC) followed by SFC comparison of the separate enantiomer(s) which were obtained by asymmetric synthesis, followed by vibrational circular dichroism (VCD) analysis of the particular enantiomer(s).
  • Intermediate compounds of formula (Ila) can be synthesized from Intermediates of formulae (la) or (lb) by nucleophilic substitution with hydrazine under thermal conditions in an appropriate solvent, such as methanol, by heating the mixture at an appropriate temperature, such as at the boiling point temperature of the solvent.
  • an appropriate solvent such as methanol
  • Intermediate compounds of formula (Illa) can be synthesized from (Ila) by a condensation reaction in presence of triethyl orthoformate under thermal conditions (e.g. at 165 °C) in an appropriate solvent, such as dimethyl acetamide.
  • Intermediate compounds of formula (IVa) can be obtained by reacting (Illa) with an appropriate brominating agent, such as benzyl trimethyl ammonium tribromide, in an appropriate solvent, such as DMF.
  • a palladium-based pre-catalyst such as the G3 t- BuXPhos-Pd (Accounts of Chemical Research, 2008, 41, 1461-1473)
  • a base such as Z-BuONa
  • solvent such as 2-methyl-2 -butanol.
  • (Va) can be obtained by a nucleophilic substitution of (IVa) under thermal conditions, using DIPEA as a base, CsF and tetrabutylammonium chloride as additives in a solvent such as acetonitrile.
  • Intermediates compounds of formula (Via) can be obtained from (Va) by reaction with Tf2O in a solvent such as dichloromethane in presence of a base, such as pyridine.
  • Intermediates compounds of formula (VIb) can be obtained from (Va) by reaction with a chlorinating agent, such as POCI3 in an appropriate solvent, such as acetonitrile.
  • final compounds may be further purified by supercritical fluid chromatography or chiral reverse phase HPLC to separate their diastereomerically pure or enantiomerically pure components.
  • General Scheme 1 final compounds described by formula (II) can be prepared according to, but not limited to, the General Scheme 2.
  • - Final compounds of formula (II) can be obtained from from (VIa) or (VIb) by a Suzuki cross-coupling reaction.
  • Typical, non-limiting conditions may involve the use of Pd(dppf)Cl2.dichloromethane or Pd(PPh3)4 as catalyst, Na2CO3 or K3PO4 as base in 1,4-dioxane or 1,4-dioxane/water as a solvent at an appropriate temperature (e.g., at 100 °C).
  • the final compounds may be further purified by supercritical fluid chromatography or chiral reverse phase HPLC to separate their diastereomerically pure or enantiomerically pure components.
  • General Scheme 2 Final compounds described by formula (III) can be prepared according to, but not limited to, the General Scheme 3.
  • Intermediate compounds of formula (IXa) can be synthesized from (Villa) by a reaction with a primary amine derivative by a Buchwald-Hartwig cross-coupling reaction using a palladium-based pre-catalyst, such as the G3 t-BuXPhos-Pd (Accounts of Chemical Research, 2008, 41, 1461-1473), a base, such as t- BuONa, and a solvent, such as 2-methyl-2 -butanol.
  • a palladium-based pre-catalyst such as the G3 t-BuXPhos-Pd (Accounts of Chemical Research, 2008, 41, 1461-1473)
  • a base such as t- BuONa
  • solvent such as 2-methyl-2 -butanol.
  • Intermediate compounds of formula (Xia) can be obtained from (Xa) by a Suzuki cross-coupling reaction with appropriate boronic acid or ester derivatives using Pd(dppf)C12. dichloromethane as a source of palladium catalyst, a base such as K2CO3 or K3PO4 in an appropriate solvent, such as 1,4-dioxane/water, at an appropriate temperature (e.g., at 100 °C).
  • Intermediate compounds of formula (Xlla) can be obtained from (Xia) by removal of the Boc protecting group under acidic conditions, such as HC1 in 1,4- dioxane or TFA.
  • Final compounds of general formula (III) can be obtained from (Xlla) by reductive alkylation (e.g., with formaldehyde in presence of sodium triacetoxyborohydride or related agents, such as sodium cyanoborohydride) or nucleophilic substitution on alkyl halides or epoxides.
  • the final compounds may be further purified by supercritical fluid chromatography or chiral reverse phase HPLC to separate their diastereomerically pure or enantiomerically pure components.
  • Intermediate compounds of formula (XlVa) can be obtained from (Xllla) by reductive alkylation (e.g., with formaldehyde in presence of sodium triacetoxyborohydride or another reducing agent, such as NaBFFCN).
  • reductive alkylation e.g., with formaldehyde in presence of sodium triacetoxyborohydride or another reducing agent, such as NaBFFCN.
  • Intermediate compounds of formula (XVa) can be obtained from (XlVa) by reaction with a chlorinating agent, such as POCI3 in an appropriate solvent such as acetonitrile.
  • a chlorinating agent such as POCI3
  • an appropriate solvent such as acetonitrile.
  • Intermediate compounds of formula (XVIa) can be obtained from (Xa) by a Suzuki cross-coupling reaction with appropriate boronic acid or ester derivatives using Pd(dppf)C12. dichloromethane as a source of palladium catalyst, a base such as K2CO3, CS2CO3 or K3PO4 in an appropriate solvent, such as 1,4-dioxane/water, at an appropriate temperature (e.g., at 100 °C).
  • Intermediate compounds of formula (Xia) can be obtained from intermediates (XVIa) by removing the benzyl protecting group by catalytic hydrogenolysis, using palladium on charcoal as catalyst, H2 at a suitable pressure (such as atmospheric pressure or 10 bars) in an appropriate solvent (e.g., methanol, ethanol, or ethyl acetate).
  • a suitable pressure such as atmospheric pressure or 10 bars
  • an appropriate solvent e.g., methanol, ethanol, or ethyl acetate.
  • the benzylic group in (XVIa) can be removed using BBr3.
  • Intermediate compounds of formula (XVIIa) can be obtained from (Xa) by a Suzuki cross-coupling reaction with appropriate boronic acid or ester derivatives using Pd(dppf)C12. dichloromethane as a source of palladium catalyst, a base such as K2CO3, CS2CO3 or K3PO4 in an appropriate solvent, such as 1,4-dioxane/water, at an appropriate temperature (e.g., at 100 °C).
  • Intermediate compounds of formula (XVIIIa) can be synthesized from (Villa) by a reaction with a primary amine derivative by a Buchwald-Hartwig cross- coupling reaction using a palladium-based pre-catalyst, such as the G3 t- BuXPhos-Pd (Accounts of Chemical Research, 2008, 41, 1461-1473), a base, such as Z-BuONa, and a solvent, such as 2-methyl-2-butanol.
  • a palladium-based pre-catalyst such as the G3 t- BuXPhos-Pd (Accounts of Chemical Research, 2008, 41, 1461-1473)
  • a base such as Z-BuONa
  • a solvent such as 2-methyl-2-butanol.
  • Intermediate compounds of formula (XXa) and (XXb) can be obtained from (XIXa) by a Suzuki cross-coupling reaction with appropriate boronic acid or ester derivatives using a palladium-based pre-catalyst such as the G4 cataCxium (Chemistry-A European Journal, 2008, 14, 4267 ⁇ 1279), a base such as CS2CO3 or K3PO4 in an appropriate solvent, such as 1,4-dioxane/water at an appropriate temperature (e.g., at 100 °C).
  • the reaction may be performed using Pd(dppf)C12. dichloromethane as a source of palladium catalyst, a base such as K2CO3 in an appropriate solvent, such as 1,4-dioxane/water at an appropriate temperature (e.g., at 100 °C).
  • Intermediate compounds of formulae (XXIa) and (XXIb) can be obtained from intermediates (XXa) and (XXb) by removing the benzyl protecting group by catalytic hydrogenolysis, using palladium on charcoal as catalyst, H2 at a suitable pressure (such as atmospheric pressure or 10 bars) in an appropriate solvent (e.g., methanol, ethanol, or ethyl acetate).
  • a suitable pressure such as atmospheric pressure or 10 bars
  • an appropriate solvent e.g., methanol, ethanol, or ethyl acetate
  • Intermediate compounds of formulae (XXIIa) and (XXIIb) can be obtained from (XXIa) and (XXIb) by reductive alkylation (e.g., with formaldehyde in presence of sodium triacetoxyborohydride or related agents, such a sodium cyanoborohydride) or nucleophilic substitution on alkyl halides or epoxides.
  • reductive alkylation e.g., with formaldehyde in presence of sodium triacetoxyborohydride or related agents, such a sodium cyanoborohydride
  • Final compounds of formula (IV) can be obtained from (XXIIa) or (XXIIb) by removal of the methoxymethyl (XXIIa) or ethoxymethyl (XXIIb) protecting groups in an acidic medium, such as HC1 in 1,4-di oxane.
  • the final compounds may be further purified by supercritical fluid chromatography or chiral reverse phase HPLC to separate their diastereomerically pure or enantiomerically pure components.
  • Intermediate compounds of formulae (XXIIIa) and (XXIIIb) can be obtained from (XXa) and (XXb) by a one-pot procedure involving the use of palladium on charcoal as catalyst, H2 at a suitable pressure (such as atmospheric pressure or 10 bars) in an appropriate solvent (e.g., methanol, ethanol, or ethyl acetate) in presence of polyoxymethylene - homopolymer at a suitable temperature (e.g., room temperature).
  • a suitable pressure such as atmospheric pressure or 10 bars
  • an appropriate solvent e.g., methanol, ethanol, or ethyl acetate
  • Final compounds of formula (V) can be obtained from (XXIIIa) or (XXIIIb) by removal of the methoxymethyl (XXIIIa) or ethoxymethyl (XXIIIb) protecting groups in an acidic medium, such as HC1 in 1,4-di oxane.
  • the final compounds may be further purified by supercritical fluid chromatography or chiral reverse phase HPLC to separate their diastereomerically pure or enantiomerically pure components.
  • Intermediate compounds of formula (XXIVa) can be obtained by (XVIIa) by reaction with a methylating agent, such as methyl iodide, after treatment with an appropriate base, such as NaH, in an appropriate solvent, such as DMF at an appropriate temperature, such as room temperature.
  • a methylating agent such as methyl iodide
  • an appropriate base such as NaH
  • an appropriate solvent such as DMF
  • - Intermediate compounds of formula (XXVIIa) can be obtained from (XXVIa) by removal of the Boc protecting group under acidic conditions, such as HCl in 1,4- dioxane or TFA.
  • - Intermediate compounds of formula (XXVIIIa) can be obtained from (XXVIIa) by reductive alkylation (e.g., with formaldehyde in presence of sodium triacetoxyborohydride or another reducing agent, such as NaBH3CN).
  • - Intermediates compounds of formula (XXIXa) can be obtained from (XXVIIIa) by reaction with a chlorinating agent, such as POCl3 in an appropriate solvent such as acetonitrile.
  • Intermediate compound of formula (XXXIa) can be synthesized from Intermediates of formulae (XXXa) or (XXXb) by nucleophilic substitution with hydrazine under thermal conditions in solvents such as ethanol by heating the mixture at an appropriate temperature, such as 90 °C.
  • Intermediate compounds of formula (XXXIIa) can be synthesized from Intermediates of formula (XXXIa) by condensation with an electrophilic carbonyl source, such as methyl chloroformate.
  • an electrophilic carbonyl source such as methyl chloroformate
  • the electrophilic carbonyl source such as methyl chloroformate
  • an appropriate solvent such as dichloromethane
  • the mixture is stirred for an appropriate reaction time, such as 16 hours and then it is concentrated.
  • the condensation can be finalized, for example, by taking up the residue in an appropriate solvent, such as ethanol, and stirring the mixture in presence of an appropriate base, such as KOH, at an appropriate temperature, such as 100 °C.
  • Intermediate compounds of formula (XXXIIIa) can be obtained from (XXXIIa) by reaction with a chlorinating agent, such as POCI3 in an appropriate solvent, such as toluene by heating at an appropriate temperature (for example 135 °C).
  • a chlorinating agent such as POCI3
  • an appropriate solvent such as toluene
  • Intermediate compounds of formula (XXXIVa) can be synthesized from (XXXIIIa) by a Suzuki cross-coupling reaction with appropriate boronic acid or ester derivatives using a palladium -based pre-catalyst such as the XPhos Pd G3, an appropriate base such as K3PO4, in an appropriate solvent, such as 1,4- dioxane/water under appropriate non-limiting conditions, such as at 150 °C under microwave irradiation for an appropriate time (e.g., 1 hour).
  • a palladium -based pre-catalyst such as the XPhos Pd G3
  • an appropriate base such as K3PO4
  • an appropriate solvent such as 1,4- dioxane/water under appropriate non-limiting conditions, such as at 150 °C under microwave irradiation for an appropriate time (e.g., 1 hour).
  • Intermediate compounds of formula (XXXVa) can be obtained from (XXXIVa) in a non-limiting manner by treatment with an appropriate thionation agent, such as phosphorous pentasulfide, in an appropriate solvent, such as pyridine under thermal conditions, (e.g., at 150 °C).
  • an appropriate thionation agent such as phosphorous pentasulfide
  • an appropriate solvent such as pyridine
  • Intermediate compounds of formula (XXXVIa) can be obtained from (XXXVa) in a non-limiting manner by treatment with an alkylating agent, such as bromoethane, in presence of an appropriate base, such as K2CO3, in an appropriate solvent, such as THF/water.
  • an alkylating agent such as bromoethane
  • an appropriate base such as K2CO3
  • an appropriate solvent such as THF/water.
  • Intermediate compounds of formula (XXXVIIa) can be obtained from (XXXVIa) in a non-limiting manner by a nucleophilic substitution of (XXXVIa) with an appropriate amine under thermal conditions (e.g., at a temperature of 130 °C) with the appropriate amine used in excess, for an appropriate reaction time (e.g., 48 hours).
  • Intermediate compounds of formula (XXXVIIIa) can be obtained from (XXXVIa) in a non-limiting manner by a nucleophilic substitution of (XXXVIa) with an appropriate amine under thermal conditions (e.g., at a temperature of 130 °C) with the appropriate amine used in excess, for an appropriate reaction time (e.g., 48 hours).
  • Intermediate compounds of formula (XXXIXa) can be obtained from intermediates (XXXVIIIa) by removing the benzyl protecting group by catalytic hydrogenolysis, using palladium on charcoal as catalyst, H2 at a suitable pressure (such as atmospheric pressure or 10 bars) in an appropriate solvent (e.g., methanol, ethanol, or ethyl acetate).
  • a suitable pressure such as atmospheric pressure or 10 bars
  • an appropriate solvent e.g., methanol, ethanol, or ethyl acetate
  • Intermediates compounds of formula (XLIIa) can be obtained from (XLIa) by reductive alkylation (e.g., with formaldehyde in presence of sodium triacetoxyborohydride or related agents, such as sodium cyanoborohydride) or nucleophilic substitution on alkyl halides or epoxides.
  • reductive alkylation e.g., with formaldehyde in presence of sodium triacetoxyborohydride or related agents, such as sodium cyanoborohydride
  • nucleophilic substitution on alkyl halides or epoxides e.g., sodium triacetoxyborohydride or related agents, such as sodium cyanoborohydride
  • Intermediates compounds of formula (XLIIIa) can be obtained from (XLIa) by nucleophilic substitution of an appropriate alkyl halide in presence of an appropriate base, such as DIPEA, in an appropriate solvent, such as acetonitrile, at an appropriate temperature, such as 85 °C.
  • an appropriate base such as DIPEA
  • an appropriate solvent such as acetonitrile
  • Intermediates compounds of formula (XLIVa) can be obtained from (XXXVIa) in a non-limiting manner by a nucleophilic substitution of (XXXVIa) with an appropriate amine under thermal conditions (e.g., at a temperature of 120 °C) with the appropriate amine used in excess, for an appropriate reaction time (e.g., 96 hours), in an appropriate solvent, such as DMSO, in presence of an appropriate base, such as DIPEA.
  • Intermediate compounds of formula (XLVa) can be obtained from intermediates (XLIVa) by removing the benzyl protecting group by catalytic hydrogenolysis, using palladium on charcoal as catalyst, H2 at a suitable pressure (such as atmospheric pressure or 10 bars) in an appropriate solvent (e.g., methanol, ethanol, or ethyl acetate).
  • a suitable pressure such as atmospheric pressure or 10 bars
  • an appropriate solvent e.g., methanol, ethanol, or ethyl acetate.
  • the instant compounds are potent, brain-penetrant, and may be useful in central nervous system diseases such as Parkinson's disease, Alzheimer's disease, dementia, motor neuron disease, Huntington's disease, traumatic brain injury, multiple sclerosis, and amyotrophic lateral sclerosis.
  • central nervous system diseases such as Parkinson's disease, Alzheimer's disease, dementia, motor neuron disease, Huntington's disease, traumatic brain injury, multiple sclerosis, and amyotrophic lateral sclerosis.
  • compositions comprising a pharmaceutically acceptable carrier and, as active ingredient, a therapeutically effective amount of a compound as provided herein.
  • the compounds may be formulated into various pharmaceutical forms for administration purposes.
  • compositions there may be cited all compositions usually employed for systemically administering drugs.
  • an effective amount of the particular compound, optionally in salt form, as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • a pharmaceutically acceptable carrier which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • These pharmaceutical compositions are desirable in unitary dosage form suitable, in particular, for administration orally or by parenteral injection.
  • any of the usual pharmaceutical media may be employed such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs, emulsions and solutions; or solid carriers such as starches, sugars, kaolin, diluents, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules and tablets. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit forms in which case solid pharmaceutical carriers are obviously employed.
  • the carrier will usually comprise sterile water, at least in large part, though other ingredients, for example, to aid solubility, may be included.
  • injectable solutions for example, may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution.
  • injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed.
  • solid form preparations which are intended to be converted, shortly before use, to liquid form preparations.
  • the pharmaceutical composition may additionally contain various other ingredients known in the art, for example, a lubricant, stabilising agent, buffering agent, emulsifying agent, viscosity-regulating agent, surfactant, preservative, flavouring or colorant.
  • a lubricant for example, a lubricant, stabilising agent, buffering agent, emulsifying agent, viscosity-regulating agent, surfactant, preservative, flavouring or colorant.
  • Unit dosage form refers to physically discrete units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • unit dosage forms are tablets (including scored or coated tablets), capsules, pills, powder packets, wafers, suppositories, injectable solutions or suspensions and the like, and segregated multiples thereof.
  • the daily dosage of the compound will, of course, vary with the compound employed, the mode of administration, the treatment desired and the mycobacterial disease indicated. However, in general, satisfactory results will be obtained when the compound is administered at a daily dosage not exceeding 1 gram, e.g. in the range from 10 to 50 mg/kg body weight.
  • pharmaceutical composition refers to a compound as provided herein or a pharmaceutically acceptable salt thereof, together with at least one pharmaceutically acceptable carrier, in a form suitable for oral or parenteral administration.
  • the term "pharmaceutically acceptable carrier” refers to a substance useful in the preparation or use of a pharmaceutical composition and includes, for example, suitable diluents, solvents, dispersion media, surfactants, antioxidants, preservatives, isotonic agents, buffering agents, emulsifiers, absorption delaying agents, salts, drug stabilizers, binders, excipients, disintegration agents, lubricants, wetting agents, sweetening agents, flavoring agents, dyes, and combinations thereof, as would be known to those skilled in the art (see, for example, Remington The Science and Practice of Pharmacy, 22nd Ed. Pharmaceutical Press, 2013, pp. 1049-1070) .
  • subject refers to an animal, preferably a mammal, most preferably a human, for example who is or has been the object of treatment, observation or experiment.
  • terapéuticaally effective amount means that amount of compound that elicits a biological or medicinal response in 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 that, when administered to a subject, is effective to (1) at least partially alleviate, inhibit, prevent and/or ameliorate a condition, or a disorder or a disease (i) mediated by NLRP3, or (ii) associated with NLRP3 activity, or (iii) characterised by activity (normal or abnormal) of NLRP3; or (2) reduce or inhibit the activity ofNLRP3; or (3) reduce or inhibit the expression of NLRP3.
  • a therapeutically effective amount refers to the amount of the compound 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.
  • inhibiting NLRP3 or inhibiting NLRP3 inflammasome pathway comprises reducing the ability ofNLRP3 or NLRP3 inflammasome pathway to induce the production of IL-1 and/or IL-18. This can be achieved by mechanisms including, but not limited to, inactivating, destabilizing, and/or altering distribution of NLRP3.
  • 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.
  • treat refers to alleviating or ameliorating the disease or disorder (i.e., slowing or arresting the development of the disease or at least one of the clinical symptoms thereof); or alleviating or ameliorating at least one physical parameter or biomarker associated with the disease or disorder, including those which may not be discernible to the patient.
  • the term “prevent”, “preventing” or “prevention” of any disease or disorder refers to the prophylactic treatment of the disease or disorder; or delaying the onset or progression of the disease or disorder.
  • a compound for use as a medicament.
  • a compound for use in the treatment of a disease or disorder associated with NLRP3 activity (including inflammasome activity); in the treatment of a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder; in inhibiting NLRP3 inflammasome activity (including in a subject in need thereof); and/or as an NLRP3 inhibitor.
  • NLRP3 activity including inflammasome activity
  • NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder
  • inhibiting NLRP3 inflammasome activity including in a subject in need thereof.
  • a method of treating a disease or disorder in which the NLRP3 signalling contributes to the pathology, and/or symptoms, and/or progression, of said disease/disorder comprising administering a therapeutically effective amount of a compound as provided herein, according to any one of the embodiments described herein (and/or pharmaceutical compositions comprising such compound, according to any one of the embodiment described herein), for instance to a subject (in need thereof).
  • a method of inhibiting the NLRP3 inflammasome activity in a subject comprising administering to the subject in need thereof a therapeutically effective amount of a compound as provided herein, according to any one of the embodiments described herein (and/or pharmaceutical compositions comprising such compound, according to any one of the embodiment described herein).
  • the instant compounds may have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (e.g. higher oral bioavailability and/or lower clearance) than, and/or have other useful pharmacological, physical, or chemical properties over, compounds known in the prior art, whether for use in the above-stated indications or otherwise.
  • a better pharmacokinetic profile e.g. higher oral bioavailability and/or lower clearance
  • the instant compounds may have the advantage that they have a good or an improved thermodynamic solubility (e.g. compared to compounds known in the prior art; and for instance as determined by a known method and/or a method described herein).
  • the instant compounds may have the advantage that they will block pyroptosis, as well as the release of pro-inflammatory cytokines (e.g. IL-10) from the cell.
  • the instant compounds may also have the advantage that they avoid side-effects, for instance as compared to compounds of the prior art, which may be due to selectivity of NLRP3 inhibition.
  • Compounds as provided herein may also have the advantage that they have good or improved in vivo pharmacokinetics and oral bioavailability. They may also have the advantage that they have good or improved in vivo efficacy.
  • the instant compounds may also have advantages over prior art compounds when compared in the tests outlined hereinafter.
  • the compounds according to the invention can generally be prepared by a succession of steps, each of which may be known to the skilled person or described herein.
  • reaction products may be isolated from the reaction medium and, if necessary, further purified according to methodologies generally known in the art, such as extraction, crystallization, and chromatography. It is further evident that reaction products that exist in more than one enantiomeric form, may be isolated from their mixture by known techniques, in particular preparative chromatography, such as preparative HPLC, chiral chromatography. Individual diastereomers or individual enantiomers can also be obtained by Supercritical Fluid Chromatography (SFC).
  • SFC Supercritical Fluid Chromatography
  • the starting materials and the intermediates are compounds that are either commercially available or may be prepared according to conventional reaction procedures generally known in the art.
  • HPLC High-Performance Liquid Chromatography
  • MS Mass Spectrometer
  • tune parameters e.g. scanning range, dwell time. . .
  • ions nominal monoisotopic molecular weight (MW).
  • Data acquisition was performed with appropriate software.
  • Compounds are described by their experimental retention times (Rt) and ions. If not specified differently in the table of data, the reported molecular ion corresponds to the [M+H] + (protonated molecule) and/or [M-H]' (deprotonated molecule).
  • SQL Single Quadrupole Detector
  • MSD Mass Selective Detector
  • RT room temperature
  • BEH bridged ethylsiloxane/silica hybrid
  • DAD Diode Array Detector
  • HSS High Strength silica.
  • Chemical shifts (5) are reported in parts per million (ppm) relative to tetramethylsilane (TMS), which was used as internal standard.
  • Values are either peak values or melt ranges and are obtained with experimental uncertainties that are commonly associated with this analytical method.
  • Method A For a number of compounds, melting points were determined with a DSC823e (Mettler Toledo) apparatus. Melting points were measured with a temperature gradient of 10 °C/minute. Standard maximum temperature was 300 °C.
  • Method B For a number of compounds, melting points were determined in open capillary tubes on a Mettler Toledo MP50. Melting points were measured with a temperature gradient of 10 °C/minute. Maximum temperature was 300 °C. The melting point data was read from a digital display and checked from a video recording system
  • RS Whenever the notation “RS” is indicated herein, it denotes that the compound is a racemic mixture at the indicated center, unless otherwise indicated.
  • the stereochemical configuration for centers in some compounds has been designated “(R)” or “(5)” when the mixture(s) was separated or originated from enantiomerically pure starting materials; for some compounds, the stereochemical configuration at the indicated centers has been designated as “*(R/’ or when the absolute stereochemistry is undetermined although the compound itself has been isolated as a single stereoisomer and is enantiomerically/diastereomerically pure.
  • the enantiomeric excess of compounds reported herein was determined by analysis of the racemic mixture by supercritical fluid chromatography (SFC) followed by SFC comparison of the separated enantiomer(s).
  • the absolute configuration of chiral centers (indicated as R and/or S) can be rationalized.
  • the synthesis of all final compounds started from intermediates of known absolute configuration in agreement with literature precedent or obtained from appropriate synthetic procedures.
  • the assignment of the absolute configuration of additional stereocenters could then be assigned by standard NMR methods.
  • N,N-Diisopropylethylamine [7087-68-5] (21.5 mL, 102 mmol) was added to a coold, 0 °C, solution of 2-iodo-5-(trifluoromethyl)phenol 1-5 (34 g, 102 mmol) in DCM (1.2 L), followed by dropwise addition of chloromethyl methyl ether [107-30-2] (9.4 mL, 121.8 mmol).
  • the reaction mixture was allowed to gradually warm to rt and react for 18 h.
  • the mixture was concentrated, and the residue was purified by flash column chromatography (silica 330 g; EtOAc in heptane 0/100 to 10/90). The desired fractions were collected and concentrated in vacuo to yield l-iodo-2-(methoxymethoxy)-4- (trifluoromethyl)benzene 1-6 (25.5 g, yield: 70%) as a colorless oil.
  • the reaction mixture was stirred for 2 h at 100 °C. The mixture was allowed to cool down to rt. The reaction mixture was diluted with ice/water (2 L). The resulting mixture was extracted with EtOAc (2 x 2 L). The combined organic layers were washed with brine (2 x 2 L), dried over anhydrous Na2SO4. After filtration, the filtrate was concentrated under reduced pressure. The residue was applied onto a silica gel column with petroleum ether. The residue was purified by trituration with n-hexane (300 mL) at -30 °C. The precipitated solids were collected by filtration.
  • Methyl (25,4A)-4-azido-l-tritylpyrrolidine-2-carboxylate 1-14 (10.3 g, 25.0 mmol, 1 equiv) in anhydrous THF (67 mL) was added dropwise to a stirred solution of lithium aluminum hydride [16853-85-3] (3.7 g, 96.9 mmol, 3.9 equiv) in anhydrous THF (90 mL) at 0 °C under nitrogen atmosphere.
  • the reaction mixture was stirred at 0 °C for 30 min. Afterwards, the reaction mixture was stirred at rt for 3h.
  • the reaction mixture was cooled at 0 °C and water (3.7 mL) was added dropwise.
  • Formaldehyde solution [50-00-0] (1.4 mL, 37% in water, 1.09 g/mL, 18.78 mmol, 2 equiv) followed by formic acid [64-18-6] (700 ⁇ L, 1.22 g/mL, 18.55 mmol, 2 equiv) were added to a solution of tert-butyl ((3A,5A)-5-fluoropiperidin-3-yl)carbamate 1-18 (2 g, 9.16 mmol, 1 equiv) in dry 2-methyl-THF (45.0 mL). The resulting colourless solution was stirred at room temperature for 1.5 h and then, heated at 80 °C for 2.5 h.
  • Phosphorus oxychloride [10025-87-3] (5.6 mL, 59.55 mmol) was added dropwise over 15 min to a stirred suspension of pyrazolo[l,5-d][l,2,4]triazin-4(5H)-one 1-28 (1.8 g, 11.91 mmol), DIPEA [7087-68-5] (4.2 mL, 23.82 mmol) and distilled water [7732-18- 5] (279 ⁇ L, 15.48 mmol) in toluene (41 mL) at rt. The suspension was stirred at 135 °C for 8 h in a sealed glass reactor.
  • reaction mixture was stirred at 105 °C overnight.
  • the mixture was concentrated and the residue was purified by flash column chromatography (silica 80 g; dry load in celite; EtOAc in heptane 0/100 to 30/70).
  • the desired fractions were collected and concentrated in vacuo.
  • the product was triturated with heptane to yield to yield l-(2-(benzyloxy)-4- (trifluoromethyl)phenyl)pyrrolo[l,2-d][l,2,4]triazin-4(3H)-one 1-30 (3.9 g, yield: 65%) as a white foamy solid.
  • Phosphorous pentasulfide (8.7 g, 38.73 mmol, 2.5 eq.) was added to a stirred solution of 1 -(2-(benzyloxy)-4-(trifluoromethyl)phenyl)pyrrolo[ 1 ,2-d] [ 1 ,2,4]triazin-4(3H)-one 1-30 (5.97 g, 15.49 mmol) in pyridine (77 mL). The mixture was stirred at 150 °C for 8 h. The reaction was recharged with phosphorous pentasulfide (4.35 g, 19.37 mmol, 1.25 eq.). The mixture was stirred at 150 °C for 8 h.
  • Potassium carbonate [584-08-7] (3.2 g, 22.87 mmol, 6 eq.) diluted in water (19.3 mL) was added dropwise to a stirred solution of l-(2-(benzyloxy)-4- (trifluoromethyl)phenyl)pyrrolo[l,2-d][l,2,4]triazine-4(3H)-thione 1-31 (1.53 g, 3.81 mmol) in THF (38.5 mL) at rt. Then bromoethane [74-96-4] (0.6 mL, 7.62 mmol, 2 eq.) was added and the mixture was stirred at rt for 16 h.
  • Trifluoroacetic acid [76-05-1] (5.6 mL, 74.32 mmol) was added dropwise to a stirred solution of tert-butyl (R)-3-((l-(2-hydroxy-4-(trifluoromethyl)phenyl)pyrrolo[l,2- d][l, 2, 4]triazin-4-yl)amino)piperi dine- 1 -carboxylate 1-41 (478 mg, 1.00 mmol) in DCM anhydrous (5.6 mL) at 0 °C under nitrogen atmosphere. The reaction mixture was stirred at rt for 1 h.
  • Trifluoroacetic acid [76-05-1] (1.43 mL, 19 mmol) was added dropwise to a stirred solution of tert-butyl (R)-3-((l-(2-(benzyloxy)-4-(trifluoromethyl)phenyl)pyrrolo[l,2- d][l, 2, 4]triazin-4-yl)amino)piperi dine- 1 -carboxylate 1-33 (150 mg, 0.26 mmol) in DCM anhydrous (1.5 mL) at 0 °C under nitrogen atmosphere. The reaction mixture was stirred at rt for 1 h.
  • the reaction was recharged with 2-(2- Bromoethoxy)tetrahydro-2h-pyran [17739-45-6] (1.2 eq, 50 ⁇ L, 0.31 mmol) and DIPEA [7087-68-5] (6 eq, 0.28 mL, 1.57 mmol)
  • the reaction mixture was cooled down to rt, diuted with sat. NaHCCf aqueous solution and extracted with EtOAc (x3).
  • the combined organic layers were dried (MgSCU), filtered and solvents evaporated in vacuo.
  • the crude was purified by flash column chromatography (silica 20 g, DCM/MeOH (9: 1) in DCM 0/100 to 35/65).
  • the yellow oil was subjected to silica gel chromatography (12 g irregular 40-60 um; 0-3% MeOH/DCM) to give (R)- l-(2-(benzyloxy)-4-(trifluoromethyl)phenyl)-N-(l-cy cl opropylpiperi din-3 - yl)pyrrolo[l,2-d][l,2,4]triazin-4-amine 1-50 (203 mg, 84%) as a brown foam solid.
  • reaction mixture was stirred at 60 °C for 2h.
  • the reaction mixture was concentrated in vacuo, taken up in sat. aqueous NaHCO3 solution, extracted with EtOAC (x3).
  • the combined organic phases were washed with brine, dried on MgSCU, filtered, and evaporated under reduced pressure.
  • HC1 (4M in dioxane) (2.36 mL, 4 M, 9.42 mmol) was added to a solution of tert-butyl 6-((4-oxo-4,5-dihydropyrazolo[l,5-d][l,2,4]triazin-7-yl)amino)-l,4-oxazepane-4- carboxylate 1-57 (330 mg, 0.942 mmol) in methanol (4 mL). The reaction mixture was stirred at rt until complete conversion. The reaction mixture was concentrated under reduced pressure to give crude product (HC1 salt). The free amine was obtained by solid phase extraction using Si-Propylsulfonic acid SCX-2 resin (SiliCycle).
  • Method 3 A reaction flask was charged with (R)-N-(l-benzylpiperi din-3 -yl)-4- chloropyrazolo[l,5-d][l,2,4]triazin-7-amine 1-75 (2.0 g, 5.83 mmol), 2-(2- (methoxymethoxy)-4-(trifluoromethyl)phenyl)-4,4,5,5-tetramethyl-l,3,2-dioxaborolane 1-7 (2.13 g, 6.42 mmol), potassium phosphate tribasic [7778-53-2] (3.72 g, 17.50 mmol), Pd(dppf)C12 DCM [95464-05-4] (357 mg, 0.44 mmol), 1,4-dioxane (40 mL) and water (4 mL).
  • the reaction mixture was bubbled with nitrogen and stirred at 90°C for 4 hours.
  • the mixture was poured into water, and the resulting mixture was extracted three times with EtOAc.
  • the combined organic layers were washed with brine, dried on MgSO4, filtered, and concentrated.
  • the residue was purified on a column with silica gel, eluent EtOAc in Heptane, from 0 to 100 %.
  • iodomethane [74-88-4] 100 ⁇ L, 2.28 g/mL, 1.6 mmol, 3.6 equiv was added and then the reaction mixture was allowed to stir at rt for 2.5 h. The reaction mixture was quenched with MeOH and concentrated in vacuo.
  • the crude was purified by silicagel column chromatography (gradient elution: 0 to 2% MeOH in DCM) to deliver tert- butyl (A > )-3-((4-(2-(methoxymethoxy)-4-(trifluoromethyl)phenyl)pyrazolo[l,5- d][l,2,4]triazin-7-yl)(methyl)amino)piperidine-l-carboxylate 1-103 (64 mg, yield 24%).
  • Trifluoroacetic acid [76-05-1] (11.5 mL, 153.5 mmol) was added dropwise to a stirred solution of tert-butyl (A)-3-((4-(2-hydroxy-4-(trifluoromethyl)phenyl)pyrazolo[l,5- d][l,2,4]triazin-7-yl)amino)piperidine-l-carboxylate 1-100 (380 mg, 0.80 mmol) in DCM anhydrous (11.5 mL) at 0 °C under nitrogen atmosphere. The reaction mixture was stirred at rt for 1.5 h at rt.
  • HC1 (4M in 1,4-dioxane) [7647-01-0] (1.12 mL, 4M, 4.47 mmol) was added to a solution of tert-butyl (A)-3-((4-(2-hydroxy-4-methylphenyl)pyrazolo[l,5- d][l,2,4]triazin-7-yl)amino)piperidine-l-carboxylate 1-91 (100 mg, 0.24 mmol) in 1,4- dioxane (1.13 mL) and the mixture was stirred at rt for 2 h. The mixture was poured out in sat. NaHCCh solution and extracted three times with EtOAc.
  • Hydrazine hydrate [7803-57-8] (12.1 mL, 162.2 mmol) was added via syringe to a 750 mL sealed stainless steel reactor containing a solution, consisting of ethyl 4-methyl-lH- pyrazole-3 -carboxylate [6076-12-6] (5 g, 32.4 mmol) in ethanol (80 mL) at room temperature under nitrogen atmosphere to give colourless solution. After sealing the reactor, the mixture was stirred at 90 °C for 18 h and then, allowed to rt.
  • Triethyl orthoformate [122-51-0] (6.6 mL, 39.5 mmol) was added via syringe to a 100 mL stainless steel reactor containing a stirred solution, consisting of 4-methyl-lH- pyrazole-5-carbohydrazide 1-120 (4.94 g, 35.25 mmol) and DMF (35 mL) at room temperature to give a colourless solution.
  • a stirred solution consisting of 4-methyl-lH- pyrazole-5-carbohydrazide 1-120 (4.94 g, 35.25 mmol) and DMF (35 mL) at room temperature to give a colourless solution.
  • the mixture was stirred at 165 °C for 16 h.
  • the mixture was allowed to cool to give a brown solution.
  • the mixture was co-distilled wit toluene, 5 times, to give a pale brown slurry.
  • Formaldehyde solution [50-00-0] (4 mL, 53.73 mmol) followed by formic acid [64-18- 6] (2 mL, 53.03 mmol) were added to a solution of tert-butyl (R)-piperi din-3 - ylcarbamate [309956-78-3] (5 g, 24.97 mmol) in 2-methyltetrahydrofuran (50 mL). The resulting solution was stirred 1 h at rt, then Ih at 80 °C.
  • the yellow sticky solid was subjected to silica gel chromatography (80 g of yellow sticky solid, 0-8% MeOH/DCM) to afford (7?)-3-methyl-7-((l-methylpiperidin-3-yl)amino)pyrazolo[l,5- d][l,2,4]triazin-4(5H)-one 1-125 (420 mg, yield: 24 %) as a yellow oil.
  • Phosphorus oxychloride [10025-87-3] (1.2 mL, 12.84 mmol) were added to a solution consisting of (R)-3-methyl-7-((l-methylpiperidin-3-yl)amino)pyrazolo[l,5- d][l,2,4]triazin-4(5H)-one 1-125 (420 mg, 1.6 mmol)and acetonitrile anhydrous (12 mL) at rt in a 35 mL screw-cap vial under N2 atmosphere. The mixture was sparged with N2 for 10 min and then stirred at 90 °C for 18 h.
  • reaction vessel was charged with (2 -hydroxy -4-(trifluoromethyl)phenyl)boronic acid [1072951-50-8] (297.4 mg, 1.44 mmol, 1.5 equiv), Pd(dppf)C12- dichloromethane [95464-05-4] (94.4 mg, 0.12 mmol, 12 mol%) and K2CO3 [584-08-7] (399 mg, 2.89 mmol, 3 equiv) and flushed with nitrogen (3 vacuum/nitrogen cycles).
  • the resulting mixture was purged with nitrogen and stirred at 90 °C for 3 hours.
  • the mixture was purified by solid phase extraction using Si-Propylsulfonic acid SCX-2 resin (SiliCycle).
  • the crude reaction mixture was transfered to a column loaded with Si-Propylsulfonic acid SCX-2 resin (SiliCycle).
  • the column was first eluted with MeOH after which the desired product was released by elution with ammoniated methanol (7 N). Tubes containing the product were concentrated under reduced pressure.
  • the column was first eluted with methanol.
  • the desired product was released by elution with 7 N NH3/methanol. Tubes containing the desired product were concentrated under reduced pressure.
  • the residue was purified using by silica gel column chromatography (gradient elution: 0 to 3% methanol in dichloromethane) to give (R)-2-(7-((l-(2-fluoroethyl)piperidin-3- yl)amino)pyrazolo[l,5-d][l,2,4]triazin-4-yl)-5-(trifluoromethyl)phenol FC-21 (85 mg, yield 56%).
  • FC-32 (100 mg, 0.25 mmol) was purified by SFC to isolate both chiral products. SFC purification was made using a i-amylose-1 column with ISOC 30% Ethanol + 0.1% diethylamine to collect and concentrate to yield FC-33 (25.9 mg, yield: 25%) as a yellow foam solid and FC-34 (21.4 mg, yield: 21%) as a yellow foam solid. The absolute configuration was not determined.
  • FC-11 (92 mg, 0.22 mmol) was purified by SFC to isolate both chiral products. SFC purification was made using a i-amylose-1 column with ISOC 25% 2-propanol + 0.1% diethylamine to yield FC-36 (first eluting enantiomer) (27.4 mg, yield: 28%) as a yellow solid and FC-37 (second eluting enantiomer) (29.6 mg, yield: 31%) as a yellow solid. The absolute configuration was not determined.
  • FC-39 (38 mg, 0.09 mmol) was purified by SFC to isolate both chiral products.
  • Stationary phase Amylose-1 250 x 30 mm 5 pm.
  • Mobile phase 70% CO2 - 30% EtOH + 0.1 % diethylamine. The fractions were collected and concentrated in vacuo to yield FC-41, first eluting enantiomer, (11 mg, 0.03 mmol) as a dark yellow solid and FC-42, second eluting enantiomer, (12 mg, 0.03 mmol) as a dark yellow solid.
  • the absolute configuration was not determined.
  • FC-38 (141 mg, 0.33 mmol) was purified by SFC to isolate both chiral components. Stationary phase: Amylose-1 250 x 30 mm 5 pm, Mobile phase: 60% CO2 - 40% MeOH + 0.1 % diethylamine. The fractions were collected, concentrated in vacuo, and triturated with diisopropyl ether to yield FC-43, first eluting enantiomer, (57 mg, 0.13 mmol) as a yellow solid and FC-44, second eluting enantiomer, (57 mg, 0.13 mmol) as a yellow solid. The absolute configuration was not determined.
  • the mixture was stirred at rt for 16 h.
  • the reaction was recharged with triethylamine [121-44-8] (3 eq, 114 ⁇ L, 0.81 mmol), Formaldehyde (37% aqueous solution) [50-00-0] (2 eq, 34 ⁇ L, 0.46 mmol) and sodium triacetoxyborohydride [56553-60-7] (1.5 eq, 76 mg, 0.35 mmol) and it was stirred at rt for 32 h more.
  • the mixture was diluted with NaHCCL (sat. aq) and extracted with DCM.
  • FC-16 (150 mg, 0.37 mmol) was purified by SFC to isolate both chiral products. SFC conditions: Stationary phase: Chiralpak Diacel AD 20 x 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2. FC-47, first eluting enantiomer, (58 mg, yield: 77%) and FC-48, second eluting enantiomer, (57 mg, yield: 76%) were obtained.
  • Step 1 A solution of 4-chloro-N-(octahydroindolizin-l-yl)pyrazolo[l,5-d][l,2,4]triazin-7- amine, (200 mg, 0.68 mmol) 1-76 and (2-hydroxy-4-(trifluoromethyl)phenyl)boronic acid [1072951-50-8] (85.95 mg, 0.42 mmol) in 1,4-dioxane (10 mL) containing water, (1 mL), was purged with nitrogen for 5 minutes.
  • Step 2 The residue was purified by preparative HPLC (Stationary phase: RP XBridge Prep C18 OBD-lOpm, 50x150mm, Mobile phase: 0.25% NH4HCO3 solution in water, CH3CN) to yield the two diastereomers.
  • the first eluting diastereomer was further purified by preparative chiral SFC (Stationary phase: Chiralcel Diacel OD 20 x 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2) to yield the optically pure TRANS enantiomers 2-(7-(((lS,8aR)-octahydroindolizin-l-yl)amino)pyrazolo[l,5- d][l,2,4]triazin-4-yl)-5-(trifluoromethyl)phenol and 2-(7-(((lR,8aS)- octahydroindolizin- 1 -yl)amino)pyrazolo[ 1 , 5-d] [ 1 ,2,4]triazin-4-yl)-5- (trifluoromethyl)phenol (FC-49 and FC-50).
  • the compound was purified by preparative chiral SFC (Stationary phase: Chiralcel Diacel OD 20 x 250 mm, Mobile phase: CO2, EtOH + 0.4 iPrNH2) to yield the optically pure enantiomers 2-(7-(((8S,8aR)- octahydroindolizin-8-yl)amino)pyrazolo[ 1 , 5-d] [ 1 ,2,4]triazin-4-yl)-5- (trifluoromethyl)phenol and 2-(7-(((8R,8aS)-octahydroindolizin-8- yl)amino)pyrazolo[l,5-d][l,2,4]triazin-4-yl)-5-(trifluoromethyl)phenol (FC-53 and FC- 54).
  • the absolute configuration was not determined. 51 mg (80% yield, for the separation) were obtained for the first eluting enantiomer and 51 mg (80% yield, for the separation
  • the crude reaction mixture was transferred to a column loaded with Si-Propyl sulfonic acid SCX-2 resin (SiliCycle) and eluted with MeOH.
  • the product compound was released by elution with 7 N NH3/MeOH.
  • the tubes containing the product compound were combined and concentrated under reduced pressure.
  • a compound of the invention for instance, a compound of the examples
  • a pharmaceutically acceptable carrier for instance, a compound of the examples
  • a therapeutically effective amount of a compound of the invention is intimately mixed with a pharmaceutically acceptable carrier, in a process for preparing a pharmaceutical composition.
  • a compound according to the present invention exhibits valuable pharmacological properties, e.g. properties susceptible to inhibit NLRP3 activity, for instance as indicated the following test, and are therefore indicated for therapy related to NLRP3 inflammasome activity.
  • PBMCs peripheral blood mononuclear cells
  • Ficoll-Histopaque Sigma- Aldrich, A0561 density gradient centrifugation. After isolation, PBMCs were stored in liquid nitrogen for later use. Upon thawing, PBMC cell viability was determined in growth medium (RPMI media supplemented with 10% fetal bovine serum, 1% Pen- Strep and 1% L-glutamine). Compounds were spotted in a 1 :3 serial dilution in DMSO and diluted to the final concentration in 30 pl medium in 96 well plates (Falcon, 353072).
  • PBMCs peripheral blood mononuclear cells
  • LPS stimulation was performed by addition of 100 ng/ml LPS (final concentration, Invivogen, tlrl-smlps) for 6 hrs followed by collection of cellular supernatant and the analysis of IL-ip (pM), IL6 and TNFa cytokines levels (pM) via MSD technology according to manufacturers’ guidelines (MSD, K151A0H).
  • IC50 values for IL-ip
  • EC 50 values IL6 and TNFa
  • the objective of this assay is to measure the permeability and efflux of test compounds, using MDCK cells transfected with the P-glycoprotein (MDR1). Two control compounds are screened alongside the test compounds, propranolol (highly permeable) and prazosin (a substrate for P-glycoprotein).
  • MDCK cells are an epithelial cell line of canine kidney origin. These cells can be stably transfected to express active P-glycoprotein (MDR1- MDCK) and are ideal for studying drug efflux due to P-gp.
  • Test compound is added to either the apical or basolateral side of a confluent monolayer of MDR1-MDCK cells and permeability in the apical to basolateral (A-B) and basolateral to apical (B-A) direction is measured by monitoring the appearance of the test compound on the opposite side of the membrane using LCMS/MS.
  • Efflux ratios (B-A permeability over A-B permeability) are calculated to determine if the test compound is subject to P-gp efflux.
  • Papp apparent permeability
  • One or more compound(s) may be tested in a number of other assays to evaluate, amongst other properties, permeability, stability (including metabolic stability and blood stability) and solubility. hERG inhibition
  • the whole cell patch clamp technique on transfected cells allows the study of ion channels with no - or limited interference from other ion-channels.
  • the effect of compounds on the hERG current are studied with an automated planar patch clamp system, SyncroPatch 384PE (as described in Obergrussberger, A., Briiggemann, A., Goetze, T.A., Rapedius, M., Haarmann, C., Rinke, I., Becker, N., Oka, T., Ohtsuki, A., Stengel, T., Vogel, M., Steindl, J.,
  • the SyncroPatch 384PE is an automated patch clamp system which allows to conduct parallel recordings from 384 wells.
  • the module is incorporated in a liquid handling pipetting robot system, Biomek FXP, for application of cells and compounds.
  • voltage protocols are constructed, and data acquired using PatchControl384 and analyzed using DataControl384 (both Nanion Technologies).
  • hERG current is determined as the maximal tail current at -30 mV and percent inhibition upon compound addition as well as pICso are reported below.
  • liver microsomes 0.5 mg/ml protein
  • preclinical species incubated up to 60 minutes at 37°C with 1 pM test compound.
  • Fine incubation volume
  • liver hepatocytes (1 milj cells) from human and preclinical species incubated up to 120 minutes at 37°C with 1 pM test compound.
  • the in vitro intrinsic clearance (Clint) (pl/min/million cells) is calculated using the following formula:
  • # cellS/n C number of cells (xlO 6 ) in the incubation
  • Test compound is prepared in species specific plasma (diluted to 25% plasma in buffer). The plasma solution is added to one side of the membrane in an equilibrium dialysis system while buffer (pH 7.4) is added to the other side. The system is allowed to reach equilibrium at 37 °C. Compound on both sides of the membrane is measured by LC-MS/MS and the fraction of unbound compound is calculated. We deliver the fraction unbound in plasma (fu) for each test compound, along with percentage recovery.
  • Plasma from the following strains and species combinations will be used:
  • test compound (1 pM test compound concentration; 0.5 % final DMSO concentration) are prepared in species specific plasma diluted to 25% plasma with buffer. The experiment is performed using equilibrium dialysis with the two compartments separated by a semi-permeable membrane. 500 pL of buffer (pH 7.4) is added to one side of the membrane and 300 pL of the plasma solution containing the test compound is added to the other side. After equilibration for 6 hr at 37°C in an incubator with 5% CO2 and agitation at 250 rpm on an orbital shaker, samples are taken from both sides of the membrane.
  • buffer pH 7.4
  • Samples are matrix matched by addition of either buffer or diluted plasma to relevant samples (i.e. 45 pL of buffer is added to 45 pL of the plasma samples and 45 pL of diluted plasma (25%) is added to 45 pL of the buffer samples). Protein is then precipitated from the matrix-matched samples by addition of 180 pL of methanol containing internal standard followed by centrifugation at 4 °C at 2500 rpm for 30 min. Supernatant (20 pL per compound x 4 compounds) is then diluted with water (100 pL) prior to analysis. Test compound incubations are performed in triplicate. Two control compounds, as specified in the guidance to vendor document, are included in each experiment.
  • BufferF Final Buffer compartment concentration (after dialysis)
  • PlasmaF Final Plasma compartment concentration (after dialysis)
  • the fraction unbound in plasma (fu) and percent recovery is returned in the form of an Excel spreadsheet.
  • the sheet will contain an indication whether the data should be further scrutinized by an internal Janssen Reviewer (based on pre-defined rules supplied in the Janssen guidance document) along with any relevant comments.
  • the objective of this study is to determine brain tissue bindings of test compound(s) in rat and mouse brain tissue using Equilibrium Dialysis Method.
  • the peak area ratios of test compound(s) in brain tissue homogenate and buffer are evaluated by LC-MS/MS.
  • Control compounds verapamil and fluoxetine are purchased from Sigma Chemical Co.
  • Control compound venlafaxine is purchased from MedChemExpress LLC.
  • Na2HPO4, NaH2PO4 and NaCl are purchased from local supplier.
  • Acetonitrile and methanol are purchased from Merck (Darmstadt, Germany).
  • Other reagents are purchased from local supplier.
  • Brain tissue homogenate is prepared by diluting one volume of the whole brain tissue with nine volumes of buffer (PBS, pH 7.4), and the mixture is homogenized using a tissue homogenate machine. Brain tissue homogenate is frozen at -80 °C prior to use. Usually, the brain tissues from three or more individual animals are pooled.
  • Assemble the 48-well RED device apparatus Add 500 pL of PBS to the buffer side of the designated wells. Add 300 uL of spiked brain homogenate immediately to the opposite sides of the designated wells. The assay was performed triplicate. Seal the RED device and place the device in an incubator at 37°C with 5% CO2 at 150 RPM for 6 hours. At the end of incubation, remove the seal and pipette 50 pL of samples from both buffer and brain tissue homogenate chambers into separate wells of a new 96-well plate.
  • Fu ⁇ FUapp brain D+Fu app -(D*Fu app )
  • Rat Sprague Dawley Wistar Supplier: Charles River Germany
  • Sampling site At each time point, animals are sacrificed by decapitation and blood is collected by exsanguination into capillaries in case of micro sampling and otherwise in BD vacutainers. Blood samples are placed immediately on melting ice and plasma is obtained following centrifugation at 4° C for 10 minutes at approximately 1900 x g.
  • Amount Micro sampling: 32 pl on EDTA
  • rat serial blood sampling is performed through the tail vein.
  • Selection of sampling method depends of the amount of plasma needed for bioanalysis.
  • Collection tubes Super Polyethylene vial 20 ml Perkin Elmer (REF.
  • Homogenization tissue Tissue samples were homogenised in demineralised water (1/9 w/v or + 3 ml if tissue weight ⁇ 0.33 g). Homogenisation is carried out under dimmed light conditions.
  • Body weight loss > 20 % body temperature is checked when animals are in sub optimal condition, mobility, changed behavior, pain expression.
  • body temperature ⁇ 33 °C animals will be euthanized and excluded from the experiment.
  • the veterinarian physician will be consulted and he/she decide of the fate of these animals. Deviations will be registered in the amendments of the study file.
  • Kpuu,brain (AUC, last, brain*BTB,r)/(AUC, last, plasma*PPB,m)
  • AUC,last is defined as the area under the concentration-time curve from dosing (time 0) to the time of the last measured concentration respectively in the brain and in the plasma
  • BTB,r is the brain tissue binding, as defined above, in rat
  • PPB,m is the plasma protein binding, as defined above, in mouse Human whole blood assay
  • 125 pl of undiluted blood was added to each well of a 96-well plate, followed by the administration of 25 pl of lipopolysaccharide (LPS E. Coli, L4130, Sigma-Aldrich) at a concentration of 30 ng/ml.
  • LPS E. Coli lipopolysaccharide
  • Blood was primed with LPS for one hour at 37°C before compound dilutions (dose-response, 25 pl/well) were added for 30 minutes at 37°C.
  • the NLRP3 pathway was activated by adding 25 pl of BzATP (A-385, Alomone Labs) to each well at a concentration of ImM.
  • mice blood of several mice was pooled (approximately 300 pl) before adding 75 pl of undiluted mouse blood to each well of a 96-well plate.
  • the NLRP3 pathway was primed by adding 25 pl of LPS (1 pg/ml) to each well for a duration of three hours at 37°C. Compounds were added at different concentrations (dose- response) and incubated for 30 minutes at 37°C before activation of the pathway with BzATP (5 mM, 25 pl/well) for 1 hour.
  • Solubility Assay An aliquot of a DMSO solution containing the test compound is dispensed in a 96-well plate, the DMSO is evaporated, and the pellet is re-dissolved by adding the buffer. The compound solubility in pH 2.0 or 7.0 buffer is measured after three days of agitation at 25 °C. The samples are centrifuged, and the super-natant is filtered. The filtrates are pooled, and the concentration is measured by liquid chromatography/tandem mass spectroscopy (LC-MS/MS). An assessment of the solid-state character of the residues is conducted by polarized light microscopy (PLM).
  • PLM polarized light microscopy
  • the phospholipidogenic potential of the compounds was assessed according to a reported procedure (Mesens, N.; Steemans, M.; Hansen, E.; Peters, A.; Verheyen, G.; Vanparys, P. A 96-well flow cytometric screening assay for detecting in vitro phospholipidosisinduction in the drug discovery phase, Toxicology in Vitro 23, (2009), 217-226. Data are reported as concentration showing a 2-fold increase in fluorescence.
  • CHI LogD Chromatography Hydrophobicity Index (CHI) CHI LogD, also referred as ChromLogD in the literature, values were determined for the compounds of the invention.
  • ChromLogD Chromatography Hydrophobicity Index

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Epidemiology (AREA)
  • Psychology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des composés destinés à être utilisés en tant qu'inhibiteurs de la voie de l'inflammasome NLRP3, de tels composés étant tels que définis par les composés de formule (I) et les radicaux R1, R2, R3, R4 et R5 étant définis dans la description, et ces composés pouvant être utiles en tant que médicaments, par exemple pour une utilisation dans le traitement d'une maladie ou d'un trouble qui est associé à l'activité de l'inflammasome NLRP3.
PCT/EP2024/051979 2023-01-31 2024-01-26 Pyrrolo[1,2-d][1,2,4]triazines et pyrazolo[1,5-d][1,2,4]triazines utiles en tant qu'inhibiteurs de nlrp3 Ceased WO2024160691A1 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU2024214354A AU2024214354A1 (en) 2023-01-31 2024-01-26 PYRROLO[1,2-d][1,2,4]TRIAZINES AND PYRAZOLO[1,5-d] [1,2,4]TRIAZINES AS NLRP3 INHIBITORS
EP24702714.7A EP4658654A1 (fr) 2023-01-31 2024-01-26 Pyrrolo[1,2-d][1,2,4]triazines et pyrazolo[1,5-d][1,2,4]triazines utiles en tant qu'inhibiteurs de nlrp3
KR1020257028762A KR20250134147A (ko) 2023-01-31 2024-01-26 NLRP3 억제제로서의 피롤로[1,2-d][1,2,4]트리아진 및 피라졸로[1,5-d] [1,2,4]트리아진
CN202480022485.6A CN120957995A (zh) 2023-01-31 2024-01-26 作为NLRP3抑制剂的吡咯并[1,2-d][1,2,4]三嗪和吡唑并[1,5-d][1,2,4]三嗪

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP23154277 2023-01-31
EP23154277.0 2023-01-31

Publications (1)

Publication Number Publication Date
WO2024160691A1 true WO2024160691A1 (fr) 2024-08-08

Family

ID=85150807

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2024/051979 Ceased WO2024160691A1 (fr) 2023-01-31 2024-01-26 Pyrrolo[1,2-d][1,2,4]triazines et pyrazolo[1,5-d][1,2,4]triazines utiles en tant qu'inhibiteurs de nlrp3

Country Status (5)

Country Link
EP (1) EP4658654A1 (fr)
KR (1) KR20250134147A (fr)
CN (1) CN120957995A (fr)
AU (1) AU2024214354A1 (fr)
WO (1) WO2024160691A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US12195460B2 (en) 2022-03-25 2025-01-14 Ventus Therapeutics U.S., Inc. Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors
US12281112B2 (en) 2021-04-07 2025-04-22 Ventus Therapeutics U.S., Inc. Compounds for inhibiting NLRP3 and uses thereof
US12312351B2 (en) 2022-10-31 2025-05-27 Ventus Therapeutics U.S., Inc. Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors
WO2025153532A1 (fr) 2024-01-16 2025-07-24 NodThera Limited Polythérapies faisant intervenir des inhibiteurs de nlrp3 et des agonistes de glp-1

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020234715A1 (fr) 2019-05-17 2020-11-26 Novartis Ag Inhibiteurs d'inflammasome nlrp3
WO2021193897A1 (fr) 2020-03-27 2021-09-30 アステラス製薬株式会社 Composé de pyridazine substitué
US11319319B1 (en) 2021-04-07 2022-05-03 Ventus Therapeutics U.S., Inc. Compounds for inhibiting NLRP3 and uses thereof
WO2022135567A1 (fr) 2020-12-25 2022-06-30 上海拓界生物医药科技有限公司 Composé contenant de la pyridazine et son utilisation médicinale
WO2023278438A1 (fr) 2021-06-29 2023-01-05 Zomagen Biosciences Ltd Modulateurs de nlrp3
WO2023028534A1 (fr) * 2021-08-25 2023-03-02 Ptc Therapeutics, Inc. Inhibiteurs de nlrp3

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020234715A1 (fr) 2019-05-17 2020-11-26 Novartis Ag Inhibiteurs d'inflammasome nlrp3
WO2021193897A1 (fr) 2020-03-27 2021-09-30 アステラス製薬株式会社 Composé de pyridazine substitué
WO2022135567A1 (fr) 2020-12-25 2022-06-30 上海拓界生物医药科技有限公司 Composé contenant de la pyridazine et son utilisation médicinale
US11319319B1 (en) 2021-04-07 2022-05-03 Ventus Therapeutics U.S., Inc. Compounds for inhibiting NLRP3 and uses thereof
WO2023278438A1 (fr) 2021-06-29 2023-01-05 Zomagen Biosciences Ltd Modulateurs de nlrp3
WO2023028534A1 (fr) * 2021-08-25 2023-03-02 Ptc Therapeutics, Inc. Inhibiteurs de nlrp3

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
"Remington The Science and Practice of Pharmacy", 2013, PHARMACEUTICAL PRESS, pages: 1049 - 1070
ACCOUNTS OF CHEMICAL RESEARCH, vol. 41, 2008, pages 1461 - 1473
BASIORKA ET AL., LANCET HAEMATOL, vol. 5, no. 9, September 2018 (2018-09-01), pages e393 - e402
CHEMISTRY-A EUROPEAN JOURNAL, vol. 14, 2008, pages 4267 - 4279
DERANGERE ET AL., CELL DEATH DIFFER., vol. 21, no. 12, December 2014 (2014-12-01), pages 1914 - 24
DOYLE ET AL., NAT MED, vol. 18, no. 5, May 2012 (2012-05-01), pages 791 - 8
FUNG ET AL., EMERG MICROBES INFECT, vol. 9, no. 1, 14 March 2020 (2020-03-14), pages 558 - 570
KELLEY ET AL., INTJMOL SCI, vol. 20, no. 13, 6 July 2019 (2019-07-06)
KELLY ET AL., BR J DERMATOL, vol. 173, no. 6, December 2015 (2015-12-01)
KNAUF ET AL., KIDNEY INT, vol. 84, no. 5, November 2013 (2013-11-01), pages 895 - 901
KRISHNAN ET AL., BR J PHARMACOL, vol. 173, no. 4, February 2016 (2016-02-01), pages 752 - 65
MESENS, N.STEEMANS, M.HANSEN, E.PETERS, A.VERHEYEN, G.VANPARYS, P., A 96-WELLFLOW CYTOMETRIC SCREENING ASSAY FOR DETECTING IN VITRO PHOSPHOLIPIDOSIS-INDUCTION IN THE DRUG DISCOVERY PHASE, TOXICOLOGY IN VITRO, vol. 23, 2009, pages 217 - 226
MIYAMAE T., PAEDIATR DRUGS, vol. 14, no. 2, 1 April 2012 (2012-04-01), pages 109 - 17
NIETO-TORRES ET AL., VIROLOGY, vol. 485, November 2015 (2015-11-01), pages 330 - 9
OBERGRUSSBERGER, A.BRUGGEMANN, A.GOETZE, T.A.RAPEDIUS, M.HAARMANN, C.RINKE, I.BECKER, N.OKA, T.OHTSUKI, A.STENGEL, T.: "Automated Patch Clamp Meets High-Throughput Screening: 384 Cells Recorded in Parallel on a Planar Patch Clamp Module", JOURNAL OF LABORATORY AUTOMATION, vol. 21, no. 6, 2016, pages 779 - 793
RIDKER ET AL., LANCET,, vol. 390, no. 10105, 21 October 2017 (2017-10-21), pages 1833 - 1842
RIDKER ET AL.: "CANTOS Trial Group.", N ENGL J MED, vol. 377, no. 12, 21 September 2017 (2017-09-21), pages 1119 - 1131
ROMBOUTS ET AL., J. MED. CHEM., vol. 64, no. 19, 2021, pages 14175 - 14191
SARKAR ET AL., NPJ PARKINSONS DIS, vol. 3, no. 30, 17 October 2017 (2017-10-17)
SHAHZAD ET AL., KIDNEY INT, vol. 87, no. 1, January 2015 (2015-01-01), pages 74 - 84
SHARIF ET AL., NATURE, vol. 570, no. 7761, June 2019 (2019-06-01), pages 338 - 343
SZABO GPETRASEK J., NAT REV GASTROENTEROL HEPATOL, vol. 12, no. 7, July 2015 (2015-07-01), pages 387 - 400
TARTEY SKANNEGANTI TD., IMMUNOLOGY, vol. 156, no. 4, April 2019 (2019-04-01), pages 329 - 338
TOLDO SABBATE A, NAT REV CARDIOL, vol. 15, no. 4, April 2018 (2018-04-01), pages 203 - 214
VAN OPDENBOSCH NLAMKANFI M., IMMUNITY, vol. 50, no. 6, 18 June 2019 (2019-06-18), pages 1352 - 1364
WALLE L ET AL., NATURE, vol. 512, no. 7512, 7 August 2014 (2014-08-07), pages 69 - 73
YANG Y ET AL., CELL DEATH DIS, vol. 10, no. 2, 12 February 2019 (2019-02-12), pages 128
ZHANG ET AL., HUM IMMUNOL, vol. 79, no. 1, January 2018 (2018-01-01), pages 57 - 62
ZHEN YZHANG H., FRONT IMMUNOL, vol. 10, 28 February 2019 (2019-02-28), pages 276

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US12281112B2 (en) 2021-04-07 2025-04-22 Ventus Therapeutics U.S., Inc. Compounds for inhibiting NLRP3 and uses thereof
US12312350B2 (en) 2021-04-07 2025-05-27 Ventus Therapeutics U.S., Inc. Compounds for inhibiting NLRP3 and uses thereof
US12410167B2 (en) 2021-04-07 2025-09-09 Ventus Therapeutics U.S., Inc. Pyridazine compounds for inhibiting NLRP3
US12441728B2 (en) 2021-04-07 2025-10-14 Ventus Therapeutics U.S., Inc. Pyridazine compounds for inhibiting NLRP3
US12195460B2 (en) 2022-03-25 2025-01-14 Ventus Therapeutics U.S., Inc. Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors
US12312351B2 (en) 2022-10-31 2025-05-27 Ventus Therapeutics U.S., Inc. Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors
US12331048B2 (en) 2022-10-31 2025-06-17 Ventus Therapeutics U.S., Inc. Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors
US12398136B2 (en) 2022-10-31 2025-08-26 Ventus Therapeutics U.S., Inc. Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 inhibitors
WO2025153532A1 (fr) 2024-01-16 2025-07-24 NodThera Limited Polythérapies faisant intervenir des inhibiteurs de nlrp3 et des agonistes de glp-1

Also Published As

Publication number Publication date
CN120957995A (zh) 2025-11-14
AU2024214354A1 (en) 2025-09-11
EP4658654A1 (fr) 2025-12-10
KR20250134147A (ko) 2025-09-09

Similar Documents

Publication Publication Date Title
WO2024160691A1 (fr) Pyrrolo[1,2-d][1,2,4]triazines et pyrazolo[1,5-d][1,2,4]triazines utiles en tant qu'inhibiteurs de nlrp3
EP3442947B1 (fr) Dérivés d'aryl ou heteroaryl substitués par une amine comme inhibiteurs de ehmt1 et ehmt2
CA2947338C (fr) Composes polyfluores agissant en tant qu'inhibiteurs de la tyrosine kinase de bruton
EP3342773B9 (fr) Inhibiteurs de syk
EP3416966B1 (fr) Procédé de préparation d'acides [7-[[4-phényl-5-alkoxycarbonyl-2-thiazol-2-yl-1,4-dihydropyrimidin-6-yl]methyl]-3-oxo-5,6,8,8a-tetrahydro-1h-imidazo[1,5-a]pyrazin-2-yl]alkyle carboxyliques
KR20250151413A (ko) NLRP3 억제제로서의 이미다조[1,2-d][1,2,4]트라이아진
HK1197067A1 (en) Cdk inhibitors
US8778893B2 (en) (R)-1-(4-(4-fluoro-2-methyl-1H-indol-5-yloxy)-5-methylpyrrolo[2,1-f][1,2,4]triazin-6-yloxy)propan-2-ol metabolites
EP2280955B1 (fr) Forme crystalline de 6-[(4s)-2-methyl-4-(2-naphthyl)-1,2, 3, 4-tetrahydroisoquinolin-7-yl]pyridazin-3-amine
KR102230383B1 (ko) 히스타민 h4 수용체의 벤조이미다졸-2-일 피리미딘 조절제
EP4658644A1 (fr) 2-(pyridazin-3-yl)-5-(trifluorométhyl)phénols utiles en tant qu'inhibiteurs de nlrp3
WO2024160694A1 (fr) [1,2,4]triazolo[4,3-a]pyridines et [1,2,4]triazolo[4,3-a]pyrazines utiles en tant qu'inhibiteurs de nlrp3
KR20170138998A (ko) 융합-고리 화합물, 약학 조성물 및 이들의 용도
EP4658640A1 (fr) Pyridin-2(1h)-ones et pyrimidin-4(3h)-ones utiles en tant qu'inhibiteurs de nlrp3
EP3362453B1 (fr) Dérivés d'éther spirocycliques de pyrazolo [1,5-a] pyrimidine-3-carboxamide
WO2023222762A1 (fr) Protac de n6-adénosine-méthyltransférase et procédés pour les utiliser
KR101771219B1 (ko) 야누스 키나제 1 선택적 억제제 및 그 의약 용도
EP3247713B1 (fr) Dérivé de pyrazolo [3,4-d]pyrimidine et son utilisation pour le traitement de la leishmaniose
WO2025203062A1 (fr) Antiviraux à petites molécules à base d'indole contre le sars-cov -2
TW202521543A (zh) 取代咪唑并芳烴及其使用方法
HK1256999B (en) Syk inhibitors
HK1195067B (en) Cdk inhibitors
HK1195067A (en) Cdk inhibitors
HK40004853A (en) Amine-substituted aryl or heteroaryl compounds as ehmt1 and ehmt2 inhibitors
HK40004853B (en) Amine-substituted aryl or heteroaryl compounds as ehmt1 and ehmt2 inhibitors

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: 24702714

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2025544779

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 202517079006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: AU2024214354

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2025123590

Country of ref document: RU

NENP Non-entry into the national phase

Ref country code: DE

WWP Wipo information: published in national office

Ref document number: 202517079006

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2024214354

Country of ref document: AU

Date of ref document: 20240126

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: CN2024800224856

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2024702714

Country of ref document: EP